WO2018011681A1 - Novel pyrimidine carboxamides as inhibitors of vanin-1 enzyme - Google Patents

Novel pyrimidine carboxamides as inhibitors of vanin-1 enzyme Download PDF

Info

Publication number
WO2018011681A1
WO2018011681A1 PCT/IB2017/054104 IB2017054104W WO2018011681A1 WO 2018011681 A1 WO2018011681 A1 WO 2018011681A1 IB 2017054104 W IB2017054104 W IB 2017054104W WO 2018011681 A1 WO2018011681 A1 WO 2018011681A1
Authority
WO
WIPO (PCT)
Prior art keywords
pyrimidin
amino
methanone
azaspiro
oxa
Prior art date
Application number
PCT/IB2017/054104
Other languages
French (fr)
Inventor
Agustin Casimiro-Garcia
Joseph Walter Strohbach
David Hepworth
Frank Eldridge Lovering
Chulho Choi
Christophe Philippe ALLAIS
Stephen Wayne Wright
Original Assignee
Pfizer Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to CN201780043514.7A priority Critical patent/CN109476645A/en
Application filed by Pfizer Inc. filed Critical Pfizer Inc.
Priority to MX2019000536A priority patent/MX2019000536A/en
Priority to CA3030381A priority patent/CA3030381A1/en
Priority to JP2019500621A priority patent/JP2019524716A/en
Priority to BR112019000589-7A priority patent/BR112019000589A2/en
Priority to EP17740116.3A priority patent/EP3484876A1/en
Priority to KR1020197004221A priority patent/KR20190026902A/en
Priority to AU2017296338A priority patent/AU2017296338A1/en
Priority to SG11201811161YA priority patent/SG11201811161YA/en
Priority to RU2019100559A priority patent/RU2019100559A/en
Priority to US16/316,688 priority patent/US10906888B2/en
Publication of WO2018011681A1 publication Critical patent/WO2018011681A1/en
Priority to IL263662A priority patent/IL263662A/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/10Spiro-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/02Stomatological preparations, e.g. drugs for caries, aphtae, periodontitis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • 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
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • 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
    • C07D471/08Bridged systems
    • 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
    • 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
    • C07D487/10Spiro-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/10Spiro-condensed systems
    • C07D491/107Spiro-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D498/10Spiro-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/12Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains three hetero rings
    • C07D498/18Bridged systems

Definitions

  • the present invention relates to novel heterocyclic compounds, or pharmaceutically acceptable salts thereof, and pharmaceutical compositions comprising the same.
  • the present invention also relates to methods of treating a subject by administering a therapeutically effective amount of these compounds, or salts thereof, to a subject. In general, these compounds act as inhibitors of vanin-1 enzyme.
  • Vanin-1 is a cell surface associated, giycosyiphosphatidyS inositol (GPi) ⁇ anchored protein which is expressed at high levels in kidney, liver and the small intestine. Vanin-1 expression can be up-regulated in multiple cell types under various inflammatory and oxidative stress conditions. Soluble Vanin-1 is found in serum of mice and humans indicating that Vanin-1 can be shed off the cell surface (Rommelaere S, et al. PPARalpha regulates the production of serum Vanin-1 by liver. FEBS Lett. 2013 Nov 15;587(22):3742-8).
  • Vanin-1 Three Vanin family members have been described in humans (Vanin-1 , Vanin-2 and Vanin-3) and these are classified as members of the biotinidase branch of the nitrilase superfamily (Kaskow BJ, et al. Diverse biological activities of the vascular non-inflammatory molecules - the Vanin pantetheinases. Biochem Biophys Res Commun. 2012 Jan 13;417(2):653-8).
  • Vanin-1 acts as the predominant pantetheinase in vivo catalyzing its hydrolysis to produce pantothenic acid (vitamin B5) and cysteamine (Pitari G, et al. Pantetheinase activity of membrane-bound vanin-1 ; lack of free cysteamine in tissues of vanin-1 deficient mice. FEBS Lett. 2000;483: 149-154). These products impact diverse biological processes. Panthothenic acid is a necessary factor in the synthesis of Coenzyme A (CoA), a cofactor involved in many metabolic processes such as fatty acid synthesis and oxidation of pyruvate.
  • CoA Coenzyme A
  • Vanin-1 -dependent phenotypes are revealed in situations of metabolic challenge and/or oxidative stress and tissue damage.
  • Vanin-1 -deficient mice exhibit resistance to oxidative tissue injury caused by y-irradiation or by the administration of paraquat which is correlated with significantly increased glutathione levels (Berruyer C, et al. Vanin-1 -/- mice exhibit a glutathione mediated tissue resistance to oxidative stress. Mol Cell Biol. 2004;24:7214-7224).
  • Vanin-1 deficient animals are also protected against multiple mouse models of IBD including DSS (dextran sulfate) and TNBS (trinitrobenzene sulfonate) colitis as evidenced by preserved mucosal barrier and reduced inflammatory infiltrate (Berruyer C, et al. Vanin-1 licenses inflammatory mediator production by gut epithelial cells and controls colitis by antagonizing peroxisome proiiferator-activated receptor ⁇ activity. J Exp Med. 2008;203:2817-2827 and et a!. Vanin-1 -/- mice show decreased NSAID- and Schistosoma-induced intestinal inflammation associated with higher glutathione stores. J Clin Invest. 2004; 1 13:591-597).
  • Vanin-1 expression is significantly increased in the colonic mucosa from IBD patients and functional polymorphisms in the regulatory regions of the Vanin-1 gene are associated with susceptibility to inflammatory bowel diseases (GensoNen T, et.al. Functional polymorphisms in the regulatory regions of the VNN1 gene are associated with susceptibility to inflammatory bowel diseases. Inflamm Bowel Dis. 2013 Oct; 19(1 1 ):2315-25).
  • patients with ulcerative colitis have an increased risk of developing colorectal cancer and Vanin- 1 knock-out mice exhibit drastically reduced incidence of tumors in colitis associated cancer model (Pouyet L, et al. Epithelial vanin-1 controls inflammation-driven carcinogenesis in the colitis-associated colon cancer model. Inflamm Bowel Dis. 2010 Jan; 16(1 ):96-104).
  • Vanin-1 is a key activator for hepatic gluconeogenesis (Chen S, et al. Vanin-1 is a key activator for hepatic gluconeogenesis. Diabetes. 2014 Jun;63(6):2073-85. doi: 10.2337/db13-0788. Epub 2014 Feb 18). Vanin-1 regulates the activation of smooth muscle cells in vitro and development of neointimal hyperplasia in response to carotid artery ligation in vivo. Polymorphysims in VNN1 gene are associated with blood pressure and HDL levels further supporting Vanin-1 's role in cardiovascular diseases.
  • Vanin-1 deficiency prevents mice from the development of adrenocortical neoplasia in Sf-1 transgenic mice suggesting a role for Vanin-1 in certain cancers.
  • Vanin-1 deficiency reduces granuloma formation and tissue damage against Coxieila burnetii, a bacterium that causes Q fever.
  • Vanin-1 is highly up-regulated in psoriatic skin lesions compared with normal individuals.
  • Vnn-1 gene expression is also up-regulated in whole blood of patients with pediatric immune thrombocytopenia (ITP) where overexpression of VNN1 , is associated with progression to chronic ITP.
  • ITP pediatric immune thrombocytopenia
  • Vanin-1 has been detected in the urine of patients with multiple renal disorders including systemic lupus erythematosus, nephrotoxicant-induced renal injury and type 2-diabetes (Rommelaere S, et al. PPARalpha regulates the production of serum Vanin-1 by liver. FEBS Lett. 2013 Nov 15;587(22):3742-8).
  • This invention relates to a com ound of Formula I,
  • G is a 6-membered heteroaryl, with one, two or three N, wherein the
  • heteroaryl is optionally substituted with one, two or three substituents selected from halogen, OH, cyano, Ci-C 4 alkyl, -NR 8a C(O)R 8b , -NR 8a SO 2 R 8b - (CR 6a R 6b )tC(O)N(R 8a ) 2 , -C(O)OH, -N(R 8a ) 2 , -(CR 6a R 6b ) t SO 2 R 8b , - (CR 6a R 6b ) t SO 2 N(R 8a ) 2 , Ci-C alkoxy, -S(Ci-C 3 alkyl) or C 3 -C 5 cycloalkyl, wherein the alkyl, cycloalkyl and alkoxy are optionally substituted with one, two or three halogen, OH, OCH 3 , or C 3 -C 5 cycloalkyl;
  • L is NH or O
  • Z is a bond; -(CR 5a R 5b ) q -; -CH 2 (CR 5a R 5b ) m -; or -(CR 5a R 5b ) m -W-(CR 5a R 5b ) n -, wherein W is S, O or NR 7 ;
  • the invention also provides for pharmaceutical compositions comprising the compounds, methods of using the compounds, combination therapies utilizing the compounds and other therapeutic agents and methods of preparing the compounds.
  • the invention also provides for intermediates useful in the preparation of the compounds of the invention.
  • the compounds of the invention may inhibit the vanin-1 enzyme.
  • Such compounds may therefore be useful for treating diseases or disorders that are mediated by, or otherwise associated with, inhibition of the vanin-1 enzyme, the method comprising administering to a subject in need thereof, an effective amount of a compound of the invention.
  • the present invention further provides a method of inhibiting vanin-1 enzyme in a cell, comprising contacting the cell with a therapeutically effective amount of a compound of Formula I, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising a therapeutically effective amount of a compound of Formula I, or a pharmaceutically acceptable salt thereof.
  • Inhibitors of vanin-1 enzyme may be used in the treatment of a variety of diseases or disorders related to systemic or tissue inflammation, inflammatory responses to infection or hypoxia, cellular activation and proliferation, lipid metabolism, fibrosis and in the treatment of viral infections. Therefore, inhibition of Vanin-1 would have the potential for multiple therapeutic indications over a wide range of unmet needs.
  • Figure 1 is a PXRD pattern of Example 142.
  • Figure 2 is an X-ray crystal structure (ORTEP drawing) of Example 145a, 8-oxa- 2-azaspiro[4.5]dec-2-yl(2- ⁇ [(1 S)-1 -(pyrazin-2-yl)ethyl]amino ⁇ pyrimidin-5-yl)methanone methanesulfonate.
  • the present invention relates to novel heterocyclic compounds of the invention which, in general, inhibit vanin-1 enzyme.
  • the present invention may be understood more readily by reference to the following detailed description of exemplary embodiments of the invention and the examples included therein. It is to be understood that this invention is not limited to specific methods of synthesis, which may of course vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting.
  • alkyl refers to a linear or branched saturated hydrocarbon moiety, consisting solely of carbon and hydrogen atoms. In one embodiment from one to six carbon atoms; and in another embodiment from one to four carbon atoms; and in another embodiment one to three carbon atoms.
  • substituents include methyl, ethyl, propyl (including n-propyl and isopropyl), butyl (including n-butyl, isobutyl, sec-butyl and fe/f-butyl), pentyl, isoamyl, hexyl and the like.
  • an alkyl may be optionally substituted at each carbon as defined in the claims.
  • Typical substitution includes, but is not limited to, fluoro, chloro, OH, cyano, alkyl (optionally substituted), alkoxy, cycloalkyl and the like.
  • the number of carbon atoms in a hydrocarbon substituent is indicated by the prefix “Cx-C y -” or “C x-y ", wherein x is the minimum and y is the maximum number of carbon atoms in the substituent.
  • Ci -C6-alkyl or “C-i -6 alkyl” refers to an alkyl substituent containing from 1 to 6 carbon atoms.
  • C3-C6-cycloalkyl or C3-6-cycloalkyl refers to saturated cycloalkyl containing from 3 to 6 carbon ring atoms.
  • cycloalkyl refers to a nonaromatic ring containing 3 to 12 carbons that is fully hydrogenated consisting of mono-, bi- or tricyclic rings. Accordingly, a cycloalkyl may be a single ring, which typically contains from 3 to 7 ring atoms. Examples include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl. Alternatively, 2 or 3 rings may be fused together, such as bicyclodecanyl and decalinyl.
  • cycloalkyl also includes bridged bicycloalkyl systems such as, but not limited to, bicyclo[2.2.1 ]heptane and bicyclo[1 .1 .1 ]pentane.
  • the cycloalkyl group may be optionally substituted as described herein, as appropriate, by 1 to 5 suitable substituents as defined herein, including but not limited to, for example, Ci-C 4 alkyl, oxo, OH, CH 2 OH, halogen, Ci-C 4 alkoxy, cyano or C(O)NH 2 , wherein said alkyl and alkoxy are optionally substituted with OH, halogen, C(O)NH 2 , C(O)NHCH 3 , C(O)N(CH 3 ) 2 , - S(Ci-C alkyl) or C 3 -C 5 cycloalkyl.
  • heterocycloalkyl means a monovalent saturated moiety, consisting of one to three rings, incorporating one, two, three or four heteroatoms (selected from N, O or S) and three to 12 carbon atoms.
  • the heterocycloalkyl may be optionally substituted as defined herein.
  • heterocycloalkyl moieties include, but are not limited to, optionally substituted piperidinyl, piperazinyl, homopiperazinyl, azepinyl, pyrrolidinyl, imidazolidinyl, morpholinyl, quinuclidinyl, thiadiazolylidinyl, benzothiazolidinyl, benzoazolylidinyl, dihydrofuryl, tetrahydrofuryl, dihydropyranyl, tetrahydropyranyl, thiamorpholinyl, thiamorpholinylsulfoxide, thiamorphilinylsulfone, dihydroquinolinyl, tetrahydroquinolinyl, tetrahydrisoquinolinyl, and the like.
  • Heterocycloalkyls may be optionally substituted, as appropriate, by 1 to 5 suitable substituents as defined herein such as including but not limited to, for example, C-i - C 4 alkyl, oxo, OH, CH 2 OH, halogen, Ci-C 4 alkoxy, cyano or C(O)NH 2 , wherein said alkyl and alkoxy are optionally substituted with OH, halogen, C(O)NH 2 , C(O)NHCH 3 , C(O)N(CH 3 ) 2 , -S(Ci-C alkyl) or C 3 -C 5 cycloalkyl.
  • suitable substituents as defined herein such as including but not limited to, for example, C-i - C 4 alkyl, oxo, OH, CH 2 OH, halogen, Ci-C 4 alkoxy, cyano or C(O)NH 2 , wherein said alkyl and alkoxy are optionally substituted with
  • heterocycloalkyl also includes fused ring systems with, for example, a cycloalkyl, aryl or heteroaryl.
  • heteroalkyl by itself or in combination with another term, means, unless otherwise stated, a saturated, straight or branched chain hydrocarbon radical consisting of the stated number of carbon atoms and from one to three heteroatoms selected from the group consisting of 0, N and S, and wherein the nitrogen and sulfur atoms may optionally be oxidized and the nitrogen heteroatom may optionally be quaternized.
  • the heteroatom(s) 0, N and S may be placed at any interior position of the heteroalkyl group.
  • the heteroatom S may be placed at any position of the heteroalkyl group, including the position at which the alkyl group is attached to the remainder of the molecule. Up to two heteroatoms may be consecutive.
  • alkoxy and alkyloxy which may be used interchangeably, refers to a moiety of the formula -OR, wherein R is a straight chain saturated alkyl or branched chain saturated alkyl moiety, as defined herein, bonded through an oxygen atom.
  • the alkoxy group may be optionally substituted as defined herein.
  • Non-limiting examples of such alkoxy groups are methoxy, ethoxy, propoxy, isopropoxy, butoxy, isobutoxy, tertiary butoxy, pentoxy and the like.
  • aryl means a carbocyclic aromatic system containing one or two rings wherein such rings may be fused. If the rings are fused, one of the rings must be fully unsaturated and the fused ring(s) may be fully saturated, partially unsaturated or fully unsaturated.
  • fused means that a second ring is present (i.e., attached or formed) by having two adjacent atoms in common (i.e., shared) with the first ring.
  • fused is equivalent to the term “condensed”.
  • the aryl group may be optionally substituted as defined herein.
  • aryl embraces aromatic radicals such as phenyl, naphthyl, tetrahydronaphthyl, indanyl, biphenyl, benzo[b][1 ,4]oxazin-3(4H)-onyl, 2,3-dihydro-1 /-/ indenyl and 1 ,2,3,4-tetrahydronaphthalenyl.
  • Aryls may be optionally substituted, as appropriate, by 1 to 5 suitable substituents as defined herein such as Ci-C 4 alkyl, oxo, OH, CH 2 OH, halogen, Ci-C 4 alkoxy, cyano or C(O)NH 2 , wherein said alkyl and alkoxy are optionally substituted with OH, halogen, C(O)NH 2 , C(O)NHCH 3 , C(O)N(CH 3 ) 2 , -S(Ci-C alkyl) or C 3 -C 5 cycloalkyland the like.
  • suitable substituents as defined herein such as Ci-C 4 alkyl, oxo, OH, CH 2 OH, halogen, Ci-C 4 alkoxy, cyano or C(O)NH 2 , wherein said alkyl and alkoxy are optionally substituted with OH, halogen, C(O)NH 2 , C(O)NHCH 3 , C(O)
  • heteroalkyl by itself or in combination with another term, means, unless otherwise stated, a saturated, straight or branched chain hydrocarbon radical consisting of the stated number of carbon atoms and from one to three heteroatoms selected from the group consisting of O, N and S, and wherein the nitrogen and sulfur atoms may optionally be oxidized and the nitrogen heteroatom may optionally be quaternized.
  • the heteroatom(s) O, N and S may be placed at any interior position of the heteroalkyl group.
  • the heteroatom S may be placed at any position of the heteroalkyl group, including the position at which the alkyl group is attached to the remainder of the molecule. Up to two heteroatoms may be consecutive.
  • heteroaryl refers to an aromatic ring structure containing from 5 to 6 ring atoms in which at least one of the ring atoms is a heteroatom (i.e., oxygen, nitrogen, or sulfur), with the remaining ring atoms being independently selected from the group consisting of carbon, oxygen, nitrogen, and sulfur.
  • heteroaryl substituents include 6-membered ring substituents such as pyridyl, pyrazyl, pyrimidinyl, and pyridazinyl; and 5-membered ring substituents such as triazolyl, imidazolyl, furanyl, thiophenyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl, 1 ,2,3-, 1 ,2,4-, 1 ,2,5-, or 1 ,3,4-oxadiazolyl and isothiazolyl.
  • 6-membered ring substituents such as pyridyl, pyrazyl, pyrimidinyl, and pyridazinyl
  • 5-membered ring substituents such as triazolyl, imidazolyl, furanyl, thiophenyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl, 1
  • heteroaryl In a group that has a heteroaryl substituent, the ring atom of the heteroaryl substituent that is bound to the group may be one of the heteroatoms, or it may be a ring carbon atom. Similarly, if the heteroaryl substituent is in turn substituted with a group or substituent, the group or substituent may be bound to one of the heteroatoms, or it may be bound to a ring carbon atom.
  • heteroaryl also includes pyridyl /V-oxides and groups containing a pyridine /V-oxide ring.
  • Further examples include furyl, thienyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, thiadiazolyl, pyridinyl, pyridazinyl, pyrimidinyl, pyrazinyl, pyridin-2(1 /-/)-onyl, pyridazin-2(1 /-/)-onyl, pyrimidin- 2(1 /-/)-onyl, pyrazin-2(1 /-/)-onyl, imidazo[1 ,2-a]pyridinyl, pyrazolo[1 ,5-a]pyridinyl, 5,6,7,8- tetrahydroisoquinolinyl, 5,6,7,8-tetrahydroquinolinyl, 6,7
  • the heteroaryl can be optionally substituted, as appropriate, by 1 to 5 suitable substituents as defined herein such as Ci-C 4 alkyl, oxo, OH, CH 2 OH, halogen, Ci-C 4 alkoxy, cyano or C(O)NH 2 , wherein said alkyl and alkoxy are optionally substituted with OH, halogen, C(O)NH 2 , C(O)NHCH 3 , C(O)N(CH 3 ) 2 , -S(Ci-C alkyl) or C 3 -C 5 cycloalkyl and the like.
  • suitable substituents as defined herein such as Ci-C 4 alkyl, oxo, OH, CH 2 OH, halogen, Ci-C 4 alkoxy, cyano or C(O)NH 2 , wherein said alkyl and alkoxy are optionally substituted with OH, halogen, C(O)NH 2 , C(O)NHCH 3 , C(
  • heteroaryls and heterocycloalkyls examples include furanyl, dihydrofuranyl, tetrahydrofuranyl, thiophenyl, dihydrothiophenyl, tetrahydrothiophenyl, pyrrolyl, isopyrrolyl, pyrrolinyl, pyrrolidinyl, imidazolyl, isoimidazolyl, imidazolinyl, imidazolidinyl, pyrazolyl, pyrazolinyl, pyrazolidinyl, triazolyl, tetrazolyl, dithiolyl, oxathiolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, thiazolinyl, isothiazolinyl, thiazolidinyl, isothiazolidinyl, thiaoxadiazolyl, oxathiazolyl,
  • heteroaryl also includes fused ring systems having one or two rings wherein such rings may be fused, wherein fused is as defined above. It is to be understood that if a carbocyclic or heterocyclic moiety may be bonded or otherwise attached to a designated substrate through differing ring atoms without denoting a specific point of attachment, then all possible points are intended, whether through a carbon atom or, for example, a trivalent nitrogen atom.
  • pyridyl means 2-, 3- or 4-pyridyl
  • thienyl means 2- or 3-thienyl, and so forth.
  • the number of atoms in a cyclic substituent containing one or more heteroatoms is indicated by the prefix "x- to y- membered", wherein x is the minimum and y is the maximum number of atoms forming the cyclic moiety of the substituent.
  • x is the minimum and y is the maximum number of atoms forming the cyclic moiety of the substituent.
  • “5- to 6-membered heteroaryl” refers to a heteroaryl containing from 5 to 6 atoms, including one or more heteroatoms, in the cyclic moiety of the heteroaryl.
  • the heteroatoms for this invention are selected from nitrogen, oxygen and sulfur. Specific embodiments of ring systems include, for example: 8-oxa-2-
  • ring systems include the following, that also incorporate exemplary ring substituents: 8-oxa-2-azaspiro[4.5]dec-2-yl, 7-oxa-2-azaspiro[3.5]non-2-yl, (3aR,4R,7aS)-rel- 4-hydroxyoctahydro-2H-isoindol-2-yl, (3aR,4R,7aS)-rel-4-hydroxyoctahydro-2H- isoindol-2-yl, (8-anti)-8-methoxy-3-azabicyclo[3.2.1 ]oct-3-yl, (1 R,5S,6R)-rel-6- (hydroxymethyl)-3-azabicyclo[3.1 .0]hex-3-yl, 1 ,3-dihydro-2H-isoindol-2-yl, 3- azabicyclo[3.2.2]non-3-yl, [(3-endo)-3-hydroxy-8-
  • haloalkyl and haloalkoxy are intended to include both branched and straight-chain saturated aliphatic “alkyl” or “alkoxy” groups respectively, wherein “alkyl” and “alkoxy” are as defined herein, having the specified number of carbon atoms and in which at least one hydrogen is replaced with a halogen atom.
  • halogen atom refers to F, CI, Br and I.
  • Haloalkyl groups include perhaloalkyl groups, wherein all hydrogens of an alkyl group have been replaced with halogens (e.g., -CF 3 , -CF 2 CF 3 ).
  • halogen atoms can be the same (e.g., CHF 2 , -CF 3 ) or different (e.g., CF 2 CI).
  • haloalkyl or haloalkoxy groups can optionally be substituted with one or more substituents in addition to halogen.
  • haloalkyl groups include, but are not limited to, fluoromethyl, dichloroethyl, trifluoromethyl, trichloromethyl, pentafluoroethyl, and pentachloroethyl groups.
  • halogen or halogen atom refers to the group consisting of fluorine (which may be depicted as -F), chlorine (which may be depicted as -CI), bromine (which may be depicted as -Br), or iodine (which may be depicted as -I).
  • hydroxy and hydroxyl are used interchangeably and as used herein mean an -OH group.
  • hydroxyalkyl and hydroxyalkoxy are intended to include both branched and straight-chain saturated aliphatic “alkyl” or “alkoxy” groups respectively, wherein “alkyl” and “alkoxy” are as defined herein, having the specified number of carbon atoms and in which at least one hydrogen is replaced with a -OH group. Where so indicated, hydroxyalkyl and hydroxyalkoxy groups can optionally be substituted with one or more substituents in addition to -OH.
  • hydroxyalkyl groups include, but are not limited to, CH 2 OH, CH 2 CH 2 OH or CH 2 (OH)CH 2 OH.
  • oxo or “carbonyl” refers to
  • sulfonyl refers to -SO 2 -.
  • substituted is used throughout the specification.
  • the term “substituted” is defined herein as a moiety, whether acyclic or cyclic, which has one or more (e.g. 1 -10) hydrogen atoms replaced by a substituent as defined herein below.
  • Substituents include those that are capable of replacing one or two hydrogen atoms of a single moiety at a time, and also those that can replace two hydrogen atoms on two adjacent carbons to form said substituent.
  • substituents that replace single hydrogen atoms include, but are not limited to, halogen, hydroxy, and the like.
  • a two hydrogen atom replacement includes, but is not limited to, carbonyl, oximino, and the like.
  • Substituents that replace two hydrogen atoms from adjacent carbon atoms include, but are not limited to, epoxy, and the like.
  • any number of its hydrogen atoms can be replaced, as described above.
  • difluoromethyl is a substituted Ci alkyl
  • trifluoromethyl is a substituted Ci alkyl
  • 4-hydroxyphenyl is a substituted aryl ring
  • (N,N-dimethyl-5- amino)octanyl is a substituted Cs alkyl
  • 3-guanidinopropyl is a substituted C3 alkyl
  • 2-carboxy-3-fluoropyridinyl is a substituted heteroaryl.
  • a multi-moiety substituent is bound through the atom indicated by To illustrate this the term "-OC-i-Cshydroxyalkyl” is an OCi-C3alkyl group substituted by a hydroxy group. Further, any carbon number pre-fix attached to a multi-moiety substituent only applies to the moiety it immediately precedes. To illustrate, the term “cycloalkyl(Ci-C4)alkyl” contains two moieties: alkyl and cycloalkyl.
  • the (C1-C4) pre-fix on the cycloalkyl(Ci-C4)alkyl means that the alkyl moiety of the alkylcycloalkyl contains from 1 to 4 carbon atoms, the (C1-C4) pre-fix does not describe the cycloalkyl moiety.
  • substituents are collectively described as being optionally substituted by one or more of a list of substituents, the group may include (1 ) unsubstitutable substituents, (2) substitutable substituents that are not substituted by the optional substituents, and/or (3) substitutable substituents that are substituted by one or more of the optional substituents.
  • a substituent such that it "may be substituted” or as being “optionally substituted” with up to a particular number of non-hydrogen substituents, that substituent may be either (1 ) not substituted; or (2) substituted by up to that particular number of non-hydrogen substituents or by up to the maximum number of substitutable positions on the substituents, whichever is less.
  • a substituent is described as a heteroaryl optionally substituted with one, two or three substituents, then any heteroaryl with less than three substitutable positions would be optionally substituted by up to only as many non-hydrogen substituents as the heteroaryl has substitutable positions.
  • tetrazolyl which has only one substitutable position
  • substituents of compounds are disclosed in groups or in ranges. It is specifically intended that the description include each and every individual sub-combination of the members of such groups and ranges.
  • C-i -6 alkyl is specifically intended to individually disclose C-i , C2, C3, C4, C5, C6, C1-C6, C1-C5, C1-C4, C1-C3, C1-C2, C2-C6, C2-C5, C2-C4, C2-C3, C3-C6, C3- C5, C3-C4, C4-C6, C4-C5, and C5-C6 alkyl.
  • C1-3 alkyl is specifically intended to individually disclose C-i , C2, C3, C1-C3, C1-C2, and C2-C3 alkyl.
  • Compounds of the present invention may contain basic nitrogen atoms (e.g. alkyl amines or heterocycles such as pyridine etc.) which may be converted to N-oxides by treatment with an oxidizing agent (e.g. mCPBA and/or hydrogen peroxides) to afford other compounds of this invention.
  • an oxidizing agent e.g. mCPBA and/or hydrogen peroxides
  • Forma la-lg may be hereinafter referred to as a "compound(s) of the invention,” “the present invention,” and collectively the “compound of Formula I.” Accordingly, the term “compound of Formula I” or “compound of formula (I), and the like, includes the compounds of Formula I, la, lb, Ic, Id, le, If and Ig, as well as the compounds of Formula II, Ma, Mb, lie, lid, Me, llf and llg, whether capitalized, bolded or not. Such terms are also defined to include all forms of the compound of Formula I, including hydrates, solvates, isomers, crystalline and non-crystalline forms, isomorphs, polymorphs, tautomers and metabolites thereof.
  • the compounds of the invention may exist in unsolvated and solvated forms.
  • the complex When the solvent or water is tightly bound, the complex will have a well-defined stoichiometry independent of humidity.
  • the solvent or water When, however, the solvent or water is weakly bound, as in channel solvates and hygroscopic compounds, the water/solvent content will be dependent on humidity and drying conditions. In such cases, non-stoichiometry will be the norm.
  • the compounds of the invention have asymmetric carbon atoms.
  • the carbon- carbon bonds of the compounds of the invention may be depicted herein using a solid line ( ), a solid wedge ( ⁇ ), or a dotted wedge ( 11 ).
  • a solid line to depict bonds to asymmetric carbon atoms is meant to indicate that all possible stereoisomers (e.g., specific enantiomers, racemic mixtures, etc.) at that carbon atom are included.
  • the use of either a solid or dotted wedge to depict bonds to asymmetric carbon atoms is meant to indicate that only the stereoisomer shown is meant to be included or that the stereoisomer predominates the other stereoisomer.
  • compounds of Formula I may contain more than one asymmetric carbon atom. In those compounds, the use of a solid line to depict bonds to asymmetric carbon atoms is meant to indicate that all possible stereoisomers are meant to be included.
  • the compounds of Formula I can exist as enantiomers and diastereomers or as racemates and mixtures thereof.
  • the use of a solid line to depict bonds to one or more asymmetric carbon atoms in a compound of Formula I and the use of a solid or dotted wedge to depict bonds to other asymmetric carbon atoms in the same compound is meant to indicate that a mixture of diastereomers is present.
  • Stereoisomers of Formula I include cis and trans isomers, optical isomers such as R and S enantiomers, diastereomers, geometric isomers, rotational isomers, conformational isomers, and tautomers of the compounds of the invention, including compounds exhibiting more than one type of isomerism; and mixtures thereof (such as racemates and diastereomeric pairs). Also included are acid addition or base addition salts wherein the counterion is optically active, for example, D-lactate or L-lysine, or racemic, for example, DL-tartrate or DL-arginine.
  • Figure 2 depicts an X-ray crystal structure (ORTEP drawing) of Example 145a, 8-oxa-2-azaspiro[4.5]dec-2-yl(2- ⁇ [(1 S)-1 -(pyrazin-2- yl)ethyl]amino ⁇ pyrimidin-5-yl)methanone methanesulfonate.
  • the single crystal X-Ray structure of example 145a is consistent with Example 145 having a "S" absolute configuration.
  • Example 146 was assigned the "R" enantiomer of this pair and dis layed a ⁇ 100-fold loss of potency against vanin in the assay.
  • Example 145 asymmetric carbon at the same position as Example 145, as detailed in the Examples herein.
  • the most potent enantiomer was assigned the "S" absolute configuration, based upon the configuration of Example 145, while the least potent enantiomer was assigned the "R” configuration. Accordingly, some of the Examples have a designation of "absolute stereochemistry inferred", based upon the above assumptions.
  • Optical isomers can be obtained in pure form by standard procedures known to those skilled in the art, which include, but are not limited to for example, chiral chromatography, diastereomeric salt formation, kinetic resolution, and asymmetric synthesis. It is also understood that the present teachings encompass all possible regioisomers, and mixtures thereof, which can be obtained in pure form by standard separation procedures known to those skilled in the art, and include, but are not limited to, column chromatography, thin-layer chromatography, and high-performance liquid chromatography.
  • the first type is the racemic compound (true racemate) referred to above wherein one homogeneous form of crystal is produced containing both enantiomers in equimolar amounts.
  • the second type is the racemic mixture or conglomerate wherein two forms of crystal are produced in equimolar amounts each comprising a single enantiomer.
  • the compounds of the invention not only include compounds as hereinbefore defined, but also all forms of the compounds of the invention, including isomers (including optical, geometric and tautomeric isomers), hydrates, solvates, complexes, salts (including solvates and complexes thereof) crystalline and non-crystalline forms, isomorphs, polymorphs, isotopically-labeled derivatives, metabolites and prodrugs (including tautomeric forms of such prodrugs) thereof.
  • a “metabolite” of a compound disclosed herein is a derivative of that compound that is formed when the compound is metabolized.
  • active metabolite refers to a biologically active derivative of a compound that is formed when the compound is metabolized.
  • metabolism refers to the sum of the processes (including, but not limited to, hydrolysis reactions and reactions catalyzed by enzymes, such as, oxidation reactions) by which a particular substance is changed by an organism. Thus, enzymes may produce specific structural alterations to a compound.
  • cytochrome P450 catalyzes a variety of oxidative and reductive reactions while uridine diphosphate glucuronyl transferases catalyze the transfer of an activated glucuronic-acid molecule to aromatic alcohols, aliphatic alcohols, carboxylic acids, amines and free sulfhydryl groups.
  • uridine diphosphate glucuronyl transferases catalyze the transfer of an activated glucuronic-acid molecule to aromatic alcohols, aliphatic alcohols, carboxylic acids, amines and free sulfhydryl groups.
  • Metabolites of the compounds disclosed herein can be identified either by administration of compounds to a host and analysis of tissue samples from the host, or by incubation of compounds with hepatic cells in vitro and analysis of the resulting compounds. Both methods are well known in the art.
  • metabolites of a compound are formed by oxidative processes and correspond to the corresponding hydroxy-containing compound.
  • a compound is metabolized to pharmacologically active metabolites.
  • the compounds of the invention may exist in both unsolvated and solvated forms.
  • solvate as used herein means a physical association of a compound with one or more solvent molecules, whether organic or inorganic, including water ('hydrate').
  • the compounds of the invention, or pharmaceutically acceptable salts thereof may exist in unsolvated and solvated forms.
  • the solvent or water is tightly bound, the complex will have a well-defined stoichiometry independent of humidity.
  • the solvent or water is weakly bound, as in channel solvates and hygroscopic compounds, the water/solvent content will be dependent on humidity and drying conditions. In such cases, non-stoichiometry will be the norm.
  • the compounds of this invention may be used in the form of salts derived from inorganic or organic acids.
  • a salt of the compound may be advantageous due to one or more of the salt's physical properties, such as enhanced pharmaceutical stability in differing temperatures and humidities, or a desirable solubility in water or oil.
  • a salt of a compound also may be used as an aid in the isolation, purification, and/or resolution of the compound.
  • a salt is intended to be administered to a patient (as opposed to, for example, being used in an in vitro context)
  • the salt preferably is pharmaceutically acceptable.
  • pharmaceutically acceptable salt refers to a salt prepared by combining a compound of the invention (e.g. a compound of Formula (I)) with an acid whose anion, or a base whose cation, is generally considered suitable for human consumption.
  • Pharmaceutically acceptable salts are particularly useful as products of the methods of the present invention because of their greater aqueous solubility relative to the parent compound.
  • salts of the compounds of this invention are non-toxic “pharmaceutically acceptable salts.”
  • Salts encompassed within the term “pharmaceutically acceptable salts” refer to non-toxic salts of the compounds of this invention which are generally prepared by reacting the free base with a suitable organic or inorganic acid.
  • Suitable pharmaceutically acceptable acid addition salts of the compounds of the present invention when possible include those derived from inorganic acids, such as hydrochloric, hydrobromic, hydrofluoric, boric, fluoroboric, phosphoric, metaphosphoric, nitric, carbonic, sulfonic, and sulfuric acids, and organic acids such as acetic, benzenesulfonic, benzoic, citric, ethanesulfonic, fumaric, gluconic, glycolic, isothionic, lactic, lactobionic, maleic, malic, methanesulfonic, trifluoromethanesulfonic, succinic, toluenesulfonic, tartaric, and trifluoroacetic acids.
  • Suitable organic acids generally include but are not limited to aliphatic, cycloaliphatic, aromatic, araliphatic, heterocyclic, carboxylic, and sulfonic classes of organic acids.
  • suitable organic acids include but are not limited to acetate, trifluoroacetate, formate, propionate, succinate, glycolate, gluconate, digluconate, lactate, malate, tartaric acid, citrate, ascorbate, glucuronate, maleate, fumarate, pyruvate, aspartate, glutamate, benzoate, anthranilic acid, stearate, salicylate, p- hydroxybenzoate, phenylacetate, mandelate, embonate (pamoate), methanesulfonate, ethanesulfonate, benzenesulfonate, pantothenate, toluenesulfonate, 2- hydroxyethanesulfonate, sufanilate, cyclohexylaminosulfonate, algenic acid, .beta.- hydroxybutyric acid, galactarate, galacturonate, adipate, alginate,
  • suitable pharmaceutically acceptable salts thereof may include alkali metal salts, i.e., sodium or potassium salts; alkaline earth metal salts, e.g., calcium or magnesium salts; and salts formed with suitable organic ligands, e.g., quaternary ammonium salts.
  • base salts are formed from bases which form non-toxic salts, including aluminum, arginine, benzathine, choline, diethylamine, diolamine, glycine, lysine, meglumine, olamine, tromethamine and zinc salts.
  • Organic salts may be made from secondary, tertiary or quaternary amine salts, such as tromethamine, diethylamine, ⁇ , ⁇ '-benzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, meglumine (N-methylglucamine), and procaine.
  • secondary, tertiary or quaternary amine salts such as tromethamine, diethylamine, ⁇ , ⁇ '-benzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, meglumine (N-methylglucamine), and procaine.
  • Basic nitrogen-containing groups may be quaternized with agents such as lower alkyl (C.sub.1 -C.sub.6) halides (e.g., methyl, ethyl, propyl, and butyl chlorides, bromides, and iodides), dialkyl sulfates (i.e., dimethyl, diethyl, dibutyl, and diamyl sulfates), long chain halides (i.e., decyl, lauryl, myristyl, and stearyl chlorides, bromides, and iodides), arylalkyl halides (i.e., benzyl and phenethyl bromides), and others.
  • lower alkyl (C.sub.1 -C.sub.6) halides e.g., methyl, ethyl, propyl, and butyl chlorides, bromides, and iodides
  • hemisalts of acids and bases may also be formed, for example, hemisulphate and hemicalcium salts.
  • complexes such as clathrates, drug-host inclusion complexes wherein, in contrast to the aforementioned solvates, the drug and host are present in stoichiometric or non-stoichiometric amounts.
  • complexes of the drug containing two or more organic and/or inorganic components which may be in stoichiometric or non-stoichiometric amounts.
  • the resulting complexes may be ionised, partially ionised, or non-ionised.
  • the present invention includes all pharmaceutically acceptable isotopically- labelled compounds of the invention wherein one or more atoms are replaced by atoms having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number usually found in nature.
  • isotopes suitable for inclusion in the compounds of the invention include isotopes of hydrogen, such as 2 H and 3 H, carbon, such as 11 C, 13 C and 1 C, chlorine, such as 36 CI, fluorine, such as 18 F, iodine, such as 123 l and 125 l, nitrogen, such as 13 N and 15 N, oxygen, such as 15 0, 17 0 and 18 0, phosphorus, such as 32 P, and sulphur, such as 35 S.
  • isotopically- labelled compounds of formula (I), for example, those incorporating a radioactive isotope, are useful in drug and/or substrate tissue distribution studies.
  • the radioactive isotopes tritium, i.e. 3 H, and carbon-14, i.e. 1 C, and 125 l are particularly useful for this purpose in view of their ease of incorporation and ready means of detection.
  • Substitution with heavier isotopes such as deuterium, i.e. 2 H may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements, and hence may be preferred in some circumstances.
  • Isotopically-labeled compounds of formula (I) can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described in the accompanying Examples and Preparations using an appropriate isotopically-labeled reagents in place of the non- labeled reagent previously employed.
  • prodrugs refers to an agent that is converted (e.g., either spontaneous or enzymatic) within the target physiological system into the parent drug in vivo.
  • Prodrugs are designed to overcome problems associated with stability, toxicity, lack of specificity, or limited bioavailability. In some situations, they may be easier to administer than the parent drug. They may, for instance, be bioavailable by oral administration whereas the parent is not.
  • the prodrug may also have improved solubility in pharmaceutical compositions over the parent drug.
  • prodrug a compound described herein, which is administered as an ester (the "prodrug") to facilitate transmittal across a cell membrane where water solubility is detrimental to mobility but which then is metabolically hydrolyzed to the carboxylic acid, the active entity, once inside the cell where water-solubility is beneficial.
  • prodrug a short peptide (polyaminoacid) bonded to an acid group where the peptide is metabolized to reveal the active moiety.
  • a prodrug upon in vivo administration, a prodrug is chemically converted to the biologically, pharmaceutically or therapeutically active form of the compound.
  • a prodrug is enzymatically metabolized by one or more steps or processes to the biologically, pharmaceutically or therapeutically active form of the compound.
  • a pharmaceutically active compound is modified such that the active compound will be regenerated upon in vivo administration.
  • the prodrug can be designed to alter the metabolic stability or the transport characteristics of a drug, to mask side effects or toxicity, to improve the flavor of a drug or to alter other characteristics or properties of a drug.
  • Prodrugs may be designed as reversible drug derivatives, for use as modifiers to enhance drug transport to site-specific tissues. See, e.g., Fedorak et al., Am. J.
  • Some preferred prodrugs are variations or derivatives of compounds that have groups cleavable under metabolic conditions.
  • Common prodrugs include acid derivatives such as esters, such as carboxylic esters (eg ethyl esters) and phosphate esters prepared by reaction of parent acids with a suitable alcohol (e.g., a lower alkanol), or of parent alcohols with a suitable acid (e.g. phosphate esters of hydroxyl groups); amides prepared by reaction of the parent acid compound with an amine, or basic groups reacted to form an acylated base derivative (e.g., a lower alkylamide).
  • esters such as carboxylic esters (eg ethyl esters) and phosphate esters prepared by reaction of parent acids with a suitable alcohol (e.g., a lower alkanol), or of parent alcohols with a suitable acid (e.g. phosphate esters of hydroxyl groups); amides prepared by reaction of the parent acid compound with an amine,
  • the invention is directed to the compound of Formula I, including the racemates, and/or diastereomer mixtures, as well as specific enantiomers and/or diastereomers thereof,
  • G is a 6-membered heteroaryl, with one, two or three N, wherein the
  • heteroaryl is optionally substituted with one, two or three substituents selected from halogen, OH, cyano, Ci-C 4 alkyl, -NR 8a C(O)R 8b , -NR 8a SO 2 R 8b - (CR 6a R 6b )tC(O)N(R 8a ) 2 , -C(O)OH, -N(R 8a ) 2 , -(CR 6a R 6b ) t SO 2 R 8b , -
  • L is NH or O
  • Z is a bond; -(CR 5a R 5b ) q -; -CH 2 (CR 5a R 5b ) m -; or -(CR 5a R 5b ) m -W-(CR 5a R 5b ) n -, wherein W is S, O or NR 7 ;
  • R a , R b , R 1 a and R 1 b are each independently hydrogen, Ci-C 4 alkyl, wherein the alkyl is optionally substituted with one, two or three halogen, OH, cyano, -S(Ci- Csalkyl) or Ci-C 4 alkoxy, optionally substituted with one, two or three fluoro;
  • R 1 a and R 1 b together with the carbon to which they are bonded, form an oxo, C3-C5cycloalkyl, -(4- to 5-membered heterocycloalkyl) wherein said cycloalkyl or heterocycloalkyl are optionally substituted with one, two, three or four halogen, OH, Ci-C 4 alkyl, -S(Ci-C3alkyl) Ci-C 4 alkoxy or cyano; and the heteroatom is
  • R 2a and R 2b are each independently hydrogen, OH, halogen, - (CR 6a R 6b )tSO 2 R 8b -(CR 6a R 6b )tC(O)N(R 8a ) 2 , -NR 8a C(O)R 8b , -NR 8a C(O)N(R 8a ) 2 , - SO 2 N(R 8a ) 2 , Ci-C 4 alkyl, Ci-C alkoxy, S(Ci-C 3 alkyl), cyano, -(CR 6a R 6b ) t -(C 3 - Cecycloalkyl), -(CR 6a R 6b ) n -(5- to 6- membered heterocycloalkyi) or -(CR 6a R 6b ) n -(5- to 6-membered heteroaryl), wherein said heteroatoms of said heteroalkyl and heteroaryl are selected from one
  • R 2a and R 2b together with the carbon to which they are bonded form a C 3 - Cgcycloalkyl or a -(4- to 1 1 -membered heterocycloalkyi), having one to three heteroatoms selected from N, O or S; wherein the cycloalkyi and heterocycloalkyi are optionally substituted with one, two or three Ci-C 4 alkyl, S(Ci-C3alkyl), OH, halogen, oxo, -C(O)NH 2 , -C(O)NHCH 3 , -C(O)N(CH 3 ) 2 , C 3 -C 5 cycloalkyl or Ci- C 4 alkoxy; or if substitution is at a N atom, then such N atom is substituted with R 7 ; or R 2a or R 2b , and one of R 5a or R 5b , together with the respective carbons to which they are bonded, form a C 3 -Ci 2
  • R 2a or R 2b , and R 7 together with the respective atoms to which they are bonded form a -(4- to 12- membered heterocycloalkyi) or a -(5- to 6-membered heteroaryl), wherein said heterocycloalkyi or heteroaryl have one, two to three heteroatoms selected from N, O or S, wherein said heterocycloalkyi and heteroaryl are optionally substituted with one, two, three or four R 9 ; or if substitution is at a N atom, then such N atom is substituted with R 7 ;
  • R 3 is hydrogen, -(CR 6a R 6b ) t C(O)NH 2 ,or Ci-C alkyl, wherein said alkyl is optionally substituted with one, two, three or four R 9 ; or
  • R 3 and R b together with the carbon to which they are attached, form an oxo;
  • R 4 is hydrogen, -(CR 6a R 6b ) t C(O)NH 2 , or Ci-C alkyl, wherein said alkyl is optionally substituted with one, two, three or four R 9 ; or
  • R 3 and R 4 taken together with the respective carbons to which they are bonded form a -(4- to 1 1 -membered heterocycloalkyi), having one to two
  • heteroatoms selected from N, O or S wherein the heterocycloalkyi are optionally substituted with one, two, three or four R 9 ; or if substitution is at a N atom, then such N atom is substituted with R 7 ; or R 4 and R a , together with the carbon to which they are attached, form an oxo;
  • R 5a and R 5b are each independently hydrogen, halogen, OH, - (CR 6a R 6b )tC(O)R 8b , -(CR 6a R 6b )tC(O)NH 2 , d-C 4 alkyl, S(Ci-C 3 alkyl), Ci-C alkoxy, cyano, -(CR 6a R 6b ) t -(C 3 -C 6 cycloalkyl) or -(CR 6a R 6b ) t -(C 3 -C 6 heterocycloalkyl), wherein said alkyl, cycloalkyi and heterocycloalkyl are optionally substituted with one, two, three or four R 9 ; and the heteroatom is selected from one or two N, O, or S; or
  • R 5a and R 5b taken together with the carbon to which they are bonded form a Cs-Cgcycloalkyl or a 4- to 1 1 -membered heterocycloalkyl, wherein the heteroatom is selected from one or two N, S or O, wherein said cycloalkyi or heterocycloalkyl is optionally substituted with one, two, three or four R 9 ; or if substitution is at a N atom, then such N atom is substituted with R 7 ; or
  • R 6a and R 6b are each independently hydrogen, Ci-C alkyl, S(Ci-C 3 alkyl), OH, Ci-C 4 alkoxy, cyano or halogen;
  • R 7 is hydrogen; -(4- to 6-membered heterocycloalkyl), having 1 to 2 heteroatoms wherein said heteroatom is selected from O, N and S; d-Csalkyl; S(Ci- C 3 alkyl); C(O)R 8b ; SO 2 R 8b ; SO 2 N(R 8a ) 2 ; C(O)N(R 8a ) 2 or -(C 3 -C 7 cycloalkyl), wherein said alkyl, heterocycloalkyl and cycloalkyi are optionally substituted with R a ;
  • R 8a is hydrogen, Ci-C 4 alkyl or -(C 3 -C 7 cycloalkyl);
  • R 8b is Ci-C alkyl, -(C 3 -C 7 cycloalkyl), -(CR 6a R 6b ) t SO 2 N(R 8a ) 2, -
  • R 9 is hydrogen, Ci-C alkyl, S(Ci-C 3 alkyl), OH, CH 2 OH, halogen, Ci-C alkoxy, cyano or -C(O)NH 2 , wherein said alkyl and alkoxy are optionally substituted with OH, halogen, -C(O)NH 2 , -C(O)NHCH 3 , -C(O)N(CH 3 ) 2 , -S(Ci-C alkyl) or C 3 - Cscycloalkyl; or R 9 is oxo, provided that it is attached to a non-aromatic group;
  • R 10 is hydrogen or Ci-C 3 alkyl
  • n and t are each independently 0, 1 or 2; q is 1 , 2 or 3; and x is 1 or 2; or pharmaceutically acceptable salts thereof.
  • the invention is directed to compounds having the Formula la, lb, lc, Id, le, If and Ig.
  • G is a 6-membered heteroaryl, with one, two or three N, wherein the heteroaryl is optionally substituted with one, two or three substituents selected from halogen, OH, cyano, Ci-C 4 alkyl, -NR 8a C(O)R 8b , -NR 8a SO 2 R 8b -
  • L is NH or O
  • Z is a bond; -(CR 5a R 5b ) q -; -CH 2 (CR 5a R 5b ) m -; or -(CR 5a R 5b ) m -W-(CR 5a R 5b ) n -, wherein W is S, O or NR 7 ; and
  • R a , R b , R 1 a , R 1 b , R 2a , R 2b , R 3 , R 4 , R 5a , R 5b , R 6 R 7 , R 8 , R 9 and R 10 are as previously described herein; or a pharmaceutically acceptable salt thereof.
  • G is a triazinyl, pyridazinyl, pyridonyl, pyridinyl, pyrazinyl or pyrimidinyl, optionally substituted with one, two or three substituents selected from halogen, OH, cyano, Ci-C alkyl, -NR 8a C(O)R 8b , -(CR 6a R 6b ) t C(O)NH 2 , -C(0)OH, -N(R ) 2 , Ci-C 4 alkoxy, wherein the alkyl and alkoxy are optionally substituted with one, two or three halogen, OH, OCH 3 , Cs-Cscycloalkyl or -S(Ci- Csalkyl);. or a pharmaceutically acceptable salt thereof.
  • G is a pyrazinyl or a pyrimidinyl.
  • L is NH
  • FIG. 1 A block diagram illustrating an exemplary computing environment in accordance with the present disclosure.
  • FIG. 1 A block diagram illustrating an exemplary computing environment in accordance with the present disclosure.
  • FIG. 1 A block diagram illustrating an exemplary computing environment in accordance with the present disclosure.
  • FIG. 1 A block diagram illustrating an exemplary computing environment in accordance with the present disclosure.
  • FIG. 1 A block diagram illustrating an exemplary computing environment in accordance with the present disclosure.
  • alkylene refers to a saturated, branched or straight chain or cyclic hydrocarbon diradical of the stated number of carbon atoms, typically 1 -6 carbon atoms, and having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent alkane.
  • Typical alkylene radicals include, but are not limited to methylene (-CH 2 -), 1 ,2-ethylene (-CH 2 CH 2 -), 2,2-dimethylene, 1 ,3-propylene (-CH 2 CH 2 CH 2 -), 2-methylpropylene, 1 ,4- butylene (-CH 2 CH 2 CH 2 CH 2 -), and the like; optionally substituted as defined herein.
  • heteroalkylene means a divalent group derived from heteroalkyl (as defined above).
  • heteroatoms can also occupy either or both of the chain termini.
  • a new ring formation between R 2a and R 2b can create a c cloalkyl, optionally substituted, as exemplified in the following structure,
  • R or R comprises an ethylene (-CH 2 CH 2 -) and one a bond.
  • R 2a or R 2b comprise a methylene (-CH 2 -).
  • R 2a and R 2b could form the following new ring formation, optionally substituted as appropriate, (R 2a and R 2b labels left in for further clarification), , wherein one of R 2a or R 2b independently is a bond, methylene (-CH 2 -), 1 ,2-ethylene (-CH 2 CH 2 -), 2,2-dimethylene, 1 ,3-propylene (-CH 2 CH 2 CH 2 -), 2- methylpropylene, 1 ,4-butylene (-CH 2 CH 2 CH 2 CH 2 -), and the like; and x is 0-5.
  • R 2a and R 2b Another embodiment of a new ring formation between R 2a and R 2b is wherein the two substituents form a -(4- to 1 1 -membered heterocycloalkyl), optionally substituted as a ropriate, such as described in the following exemplary formula.
  • the two substituents form a -(4- to 1 1 -membered heterocycloalkyl), optionally substituted as a ropriate, such as described in the following exemplary formula.
  • heterocycloalkyl is a 4-membered ring, optionall substituted as appropriate.
  • R a heterocycloalkyl
  • R or R independently is a bond, methylene (-CH 2 -), 1 ,2-ethylene (-CH 2 CH 2 -), 2,2-dimethylene, 1 ,3-propylene (-CH 2 CH 2 CH 2 -), 2-methylpropylene, 1 ,4-butylene (-CH 2 CH 2 CH 2 CH 2 -), and the like; and the other of R 2a or R 2b is a heteroalkylene.
  • a 9-carbon cycloalkyi new ring formation and an 1 1 - membered heterocycloalkyl between R 2a and R 2b are de icted in the following
  • R 2a and R 2b may form a new ring system as described above, may be extrapolated to the other substituents (R 1 a and R 1 b , R 3 and R 4 , R 2a or R 2b and either R 5a or R 5b , R 3 and either R 5a or R 5b , and R 2a or R 2b and R 7 ) that are described as forming new rings and such description is applicable, as appropriate.
  • substituents R 1 a and R 1 b , R 3 and R 4 , R 2a or R 2b and either R 5a or R 5b , R 3 and either R 5a or R 5b , and R 2a or R 2b and R 7 .
  • a new ring formation can occur between
  • R 5a or R 5b comprise a methylene (- CH 2 -).
  • R 5a or R 5b comprise an ethylene (-CH 2 CH 2 -). and the other a bond.
  • such substituents could form a 4- to 7-membered
  • heterocycloalkyi optionally substituted, , wherein the smallest heterocycloalkyi is a 4-membered ring (wherein X is N, 0 or S) and wherein R 5a or R 5b are independently a bond, an alkylene and, at least one of R 5a or R 5b comprises a heteroalkylene (e.g. -0-CH 2 -).
  • R 5a and R 5b are independently a bond, a methylene (-CH 2 -), 1 ,2-ethylene (-CH 2 CH 2 -), 2,2-dimethylene, 1 ,3-propylene (-CH 2 CH 2 CH 2 -), 2-methylpropylene, or 1 ,4-butylene
  • R and R j 3 5 D b being an alkylene or heteroalk lene group
  • a new ring formation can occur between substituents R 2a or R 2b and one Qf R 5a or R 5b tQ provjc
  • W is defined herein as N, 0, or S
  • the smallest heterocycloalkyl formed is a 4-membered heterocycloalkyl, wherein one of R 2a or R 5a comprises a methylene (-CH 2 -) and one a bond, said heterocycloalkyl optionally substituted as appropriate.
  • R 2a and R 5 could form the following specific exemplary new ring formations, optionally substituted as defined herein, (R 2a and R 5a labels remaining for
  • Ci-C 4 alkylene independently Ci-C 4 alkylene; Z is -(CR 5a R 5b ) q -; and q is 1 ); or R a , (R 2a and R 5a are independently Ci-C alkylene; Z is -(CR 5a R 5b ) q -; and P is 3 ); an ar y'.
  • V R a heteroaryl, v R ; and a heterocycloalkyl
  • R 2a and R 5a are independently -0-Ci-C 4 alkylene or Ci-C 4 alkylene; Z is -(CR 5a R 5b ) q -; and q is 3); all of which are particular embodiments of this invention, as well as other possible ring formations.
  • a new ring formation can occur between substituents
  • R 2a or R 2b and R 7 to form a -(4- to 12- membered heterocycloalkyl) or a -(5- to 6- membered heteroaryl), having two to three heteroatoms selected from N, 0 or S, and wherein W is N as exemplified by the following structures:
  • the smallest heterocycloalky formed is a 4-membered ring, wherein R 2a and R 7 is, for example, -CH 2 - , optionally substituted as defined herein.
  • R 2a and R 7 is, for example, -CH 2 - , optionally substituted as defined herein.
  • substituent groups for R 2a and R 7 could arrive at the 4-membered ring, as well as other ring sizes.
  • R is Ci-C 4 alkylene (and in this example is 1 ,4-butylene (-CH2CH2CH2CH2-)); R 7 is d-Csalkylene (and in this example, 1 ,5-pentylene (-CH2CH2CH2CH2-)); Z is -(CR 5a R 5b ) m -W-( CR 5a R 5b ) n -; and m and n are 2, to form a 12-membered heterocycloalkyl ring.
  • R 2a and R 7 could form the following specific new ring formations, optionally substituted as described herein, (R 2a and R 7 labels
  • a new ring formation can occur between substituents R 3 and R 4 to provide a -(4- to 12-membered heterocycloalkyl), optionally substituted as defined herein, having one to two heteroatoms selected from N, O or S, as exemplified in the following structure, wherein the smallest heterocycloalkyl formed is a 4-membered ring, (assuming one of R 3 or R 4 comprises a methylene (- CH -) and the other a bond), such as depicted in the following:
  • the N of the amide is part of the new ring being formed and is, therefore, included in numbering the new ring system.
  • R 3 and R 4 could form the following specific new rin formations (R 3 and R 4 labels remaining for clarification): the heterocycloalkyl,
  • R 3 and R 4 independently are a bond or Ci-C2alkylene (e.g. methylene (-CH 2 -), 1 ,2-ethylene (-CH2CH2-)).
  • Ci-C2alkylene e.g. methylene (-CH 2 -), 1 ,2-ethylene (-CH2CH2-)
  • a larger heterocycloalkyl may be formed,
  • R 3 and R 4 in this example, could each be 1 ,4-butylene
  • a new ring formation may occur between R 3 and R 5a or R 5b to provide for a C3-C7cycloalkyl or -(4- to 12-membered heterocycloalkyl), as exemplified in the following structure, , wherein the smallest heterocycloalkyi formed is a 4-membered ring (assuming R 3 or R 5a is a bond).
  • R 3 and R 5 could form the following specific new ring formations (R 3 and R 5 labels remaining for clarification): a heterocycloalkyi
  • a new ring formation may occur between R 3 and R 5a or R 5b to provide for a C -Crcycloalkyl, wherein W is O or NR 7 , as exemplified in the
  • R 3 and R 5a form an optionally substituted
  • R 3 and R 5a are each 1 ,4-butylene (-CH2CH2CH2CH2-)); Z is -(CR 5a R 5b ) m -W-( CR 5a R 5 V; and m and n are 2.
  • R 3 and R 5a form an optionally substituted 12-
  • R 3 and R 5a in this example, are each 1 ,4-butylene (-CH2CH2CH2CH2-)).
  • R 1 a or R 1 b are each independently hydrogen, or C-i- Csalkyl, wherein the alkyl is optionally substituted with one, two or three fluoro, OH, cyano or Ci-C 4 alkoxy, optionally substituted with one, two or three fluoro; or R 1 a and R 1 b , (as described above) together with the carbon to which they are bonded, form a C 3 -C 4 cycloalkyl or a 4-membered heterocycloalkyi, wherein said cycloalkyl or heterocycloalkyi are optionally substituted with one, two, three or four halogen, OH, C-i- C 4 alkyl, S(Ci-C 3 alkyl), Ci-C 4 alkoxy or cyano; or a pharmaceutically acceptable salt thereof.
  • R 1 a and R 1 b are each independently hydrogen or methyl; or R 1 a and R 1 b , together with the carbon to which they are bonded, form an optionally substituted cyclopropyl, cyclobutyl or an oxetane; or a pharmaceutically acceptable salt thereof.
  • L is NH; R a and R b are H; and x is 1 ; or a pharmaceutically acceptable salt thereof.
  • R 3 and R 4 are each independently hydrogen or Ci-C 4 alkyl, wherein said alkyl is optionally substituted with one, two, three or four R 9 ; or R 3 and R 4 taken together with the respective carbons to which they are bonded (as described above) form a -(4- to 12- membered heterocycloalkyi), having one to two heteroatoms selected from N, O or S, wherein the heterocycloalkyi is optionally substituted with one, two, three or four R 9 ; R 9 is OH, CH 2 OH, halogen, Ci-C 4 alkyl, Ci-C 4 alkoxy or cyano; or a pharmaceutically acceptable salt thereof.
  • G is selected from the following exemplary moieties: pyrazinyl, pyrimidinyl, pyridinyl or pyridazinyl, (each of which is optionally substituted with methyl, CH 2 F, CHF 2 or CF 3 ); and wherein such moieties are either carbon-linked or nitrogen-linked; or a pharmaceutically acceptable salt thereof.
  • R 2a and R 2b together with the carbon to which they are bonded form a oxetane, tetrahydrofuran, tetrahydropyran, cyclobutyl, cyclopentyl, or cyclohexyl, each of which is optionally substituted with one or two Ci-C 4 alkyl or OH; or a pharmaceutically acceptable salt thereof.
  • R 2a and R 2b are each independently hydrogen; fluoro; OH; Ci-C 4 alkyl; Ci-C 4 alkoxy; C3-C6cycloalkyl; 5-membered heteroaryl, having one or two N; cyano; -SO 2 CH 3 ; -C(O)NHR 8a ; -NHC(O)NHR 8a ; wherein said alkyl, alkoxy, cycloalkyi and heteroaryl are optionally substituted by one, two, three or four R 9 ; wherein R 9 is OH, fluoro, methyl, ethyl, methoxy or ethoxy; or a pharmaceutically acceptable salt thereof.
  • one of R 2a or R 2b taken together with the carbon to which they are bonded, and one of R 5a or R 5b , form a cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, tetrahydrofuran or phenyl, wherein each is optionally substituted with one, two, three or four R 9 , wherein R 9 is OH, CH 2 F, CHF 2 , CF 3 , CH 2 OH; or a pharmaceutically acceptable salt thereof.
  • the invention comprises a compound of Formula la
  • Z is -(CR 5a R 5b ) q -; and q is 1 ;
  • R 2a and R 2b are each independently hydrogen, methyl, ethyl, propyl,
  • R 2a and R 2b together with the carbon to which they are bonded, form a
  • R 5a or R 5b together with the respective carbons to which they are bonded, form a cyclopentane or cyclohexane, optionally substituted with one, two or three R 9 ; or a pharmaceutically acceptable salt of said compound or a tautomer of said compound or said salt.
  • the invention is directed to a compound of Formula
  • R 10 is hydrogen; and L is NH; G is pyrimidinyl or pyrazinyl; or a
  • the invention is directed to a compound having the following absolute stereochemistry,
  • R 1 a is hydrogen; and R 1 b is methyl, ethyl, propyl, wherein each is optionally substituted with one, two or three fluoro; or a pharmaceutically acceptable salt thereof.
  • R a , R b , R 3 , R 4 , R 5a and R 5b are hydrogen; and x is 1 ; R 1 b is methyl or ethyl, optionally substituted with one, two or three fluoro, or a pharmaceutically acceptable salt thereof.
  • the invention is directed to a compound of Formula lb
  • R 2a and R 2b are each independently hydrogen, methyl, ethyl, propyl, isopropyl, methoxy or ethoxy, optionally substituted with R 9 wherein R 9 is OH; or R 2a and R 2b together with the carbon to which they are bonded form a tetrahydrofuran, cyclobutane, cyclopentane, cyclohexane, oxetane, tetrahydropyran, pyrrolidine, azetidine, each of which is optionally substituted with one, two, three or four R 9 ; or a pharmaceutically acceptable salt thereof.
  • the invention is directed to the compound of Formula lib,
  • R 10 is hydrogen; and L is NH; or a pharmaceutically acceptable salt thereof.
  • the compound of Formula Mb has the absolute
  • R 1 a is hydrogen; and R 1 b is methyl, ethyl or propyl, each of which is optionally substituted by one, two or three fluoro; or a pharmaceutically acceptable salt thereof.
  • R a , R b , R 3 and R 4 are hydrogen; x is 1 ; R 1 b is methyl or ethyl, optionally substituted with one, two or three fluoro; and G is pyrimidinyl or pyrazinyl; or a pharmaceutically acceptable salt thereof.
  • the invention is directed to a compound of Formula le
  • Z is-(CR 5a R 5b ) m -W-(CR 5a R 5b ) n -, W is NR 7 , m is 1 , and n is 0; or a
  • L is NH; and R 2a or R' and R 7 , together with the respective atoms to which they are bonded form a -(4- to 12- membered heterocycloalkyi), having one or two heteroatoms selected from N or 0 , wherein said heterocycloalkyi is optionally substituted with one, two, three or four R 9 ; or a pharmaceutically acceptable salt thereof.
  • R a , R b , R 3 , R 4 , R 5a and R 5b are hydrogen and the heterocycloalkyi formed is a pyrrole; or a pharmaceutically acceptable salt thereof.
  • Yet another aspect of the invention is the compound of Formula lie,
  • R 1a is hydrogen; and R 1b is methyl, ethyl, propyl, each of which is optionally substituted by one, two or three fluoro; or a pharmaceutically acceptable salt thereof.
  • G is pyrimidinyl or pyrazinyl, or a pharmaceutically acceptable salt thereof.
  • the invention is directed to a compound of Formula Ic
  • Z is-(CR 5a R 5b ) q -; and q is 2;
  • R 5a and R 5b are each independently hydrogen, OH, fluoro, cyano, Ci-C 4 alkyl,
  • R 5a and R 5b taken together with the carbon to which they are bonded form a oxetane, tetrahydrofuran, tetrahydropyran, oxazolidinone, cyclopentane, cyclohexane, cyclobutane, cyclopropane, wherein said cycloalkyi or heterocycloalkyi are optionally substituted with one, two, three or four R 9 ;
  • R 9 is fluoro, OH or Ci-C 4 alkoxy, and t is 0 or 1 ;
  • R a , R b , R 3 , R 4 , R 5a and R 5b are hydrogen and the heterocycloalkyl formed is a pyrrole; and L is NH; or a pharmaceutically acceptable salt thereof.
  • the compound of Formula lie is
  • G is pyrimidinyl or pyrazinyl; or a pharmaceutically acceptable salt thereof.
  • the invention is directed to a compound having the absolute
  • R 1 a is hydrogen; and R 1 b is methyl, ethyl, propyl, each of which is optionally substituted with one, two or three fluoro; or a pharmaceutically acceptable salt thereof
  • the compounds of the invention are also useful in treating and/or preventing a disease or condition mediated by or otherwise associated with a Vanin-1 enzyme.
  • the use of compounds of the invention may be useful in diseases where there is evidence of oxidative stress and/or Vanin-1 enzyme upregulation; the method comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the disease may be, but not limited to, one of the following classes: autoimmune diseases, inflammatory diseases, allergic diseases, metabolic diseases, infection-based diseases, trauma or tissue-injury based diseases, fibrotic diseases, cardiovascular diseases, respiratory diseases, renal diseases, dermatological diseases, liver diseases, gastrointestinal diseases, oral diseases, pain and sensory diseases, and hematopoietic diseases.
  • autoimmune diseases include, but are not limited to: rheumatoid arthritis, osteoarthritis, psoriasis, allergic dermatitis, systemic lupus erythematosus (and resulting complications), Sjogren's syndrome, multiple sclerosis, asthma, glomerular nephritis, inflammatory bowel disease, Crohn's disease, ankylosing spondylitis, Behget's disease, lupus nephritis, scleroderma, systemic scleroderma, alopecia universalis, acute disseminated encephalomyelitis, antiphospholipid antibody syndrome, atrophic gastritis of pernicious anemia, autoimmune alopecia, autoimmune hepatitis, autoimmune encephalomyelitis, autoimmune thrombocytopenia, chronic hepatitis, Cogan's syndrome, endometriosis, Goodpasture's syndrome, Graves' disease, Guillain
  • Specific inflammatory diseases include, but are not limited to: chronic obstructive pulmonary diseases, airway hyper-responsiveness, cystic fibrosis, acute respiratory distress syndrome, sinusitis, rhinitis, gingivitis, atherosclerosis, chronic prostatitis, glomerular nephritis, ulcerative colitis, uveitis, periodontal disease, or an indication listed in a separate category herein.
  • Specific pain conditions include, but are not limited to: inflammatory pain, pain due to burns, interstitial cystitis, post-traumatic injury, pain associated with irritable bowel syndrome, gout, pain associated with any of the other indications listed within this specification, or an indication listed in a separate category herein.
  • Specific respiratory, airway and pulmonary conditions include, but are not limited to: asthma (which may encompass chronic, late, bronchial, allergic, intrinsic, extrinsic or dust), chronic obstructive pulmonary disease, idiopathic pulmonary fibrosis, pulmonary arterial hypertension, cystic fibrosis, interstitial lung disease, acute lung injury, sarcoidosis, allergic rhinitis, chronic cough, bronchitis, recurrent airway obstruction, emphysema, or bronchospasm, or an indication listed in a separate disease category herein.
  • asthma which may encompass chronic, late, bronchial, allergic, intrinsic, extrinsic or dust
  • chronic obstructive pulmonary disease idiopathic pulmonary fibrosis
  • pulmonary arterial hypertension cystic fibrosis
  • cystic fibrosis interstitial lung disease
  • acute lung injury sarcoidosis
  • allergic rhinitis allergic rhinitis
  • chronic cough bronchi
  • Gl disorders include, but are not limited to: Irritable Bowel Syndrome (IBS), Inflammatory Bowel Disease (IBD), biliary colic and other biliary disorders, renal colic, diarrhea-dominant IBS, pain associated with Gl distension, ulcerative colitis, Crohn's Disease, irritable bowel syndrome, Celiac disease, proctitis, eosinophilic gastroenteritis, mastocytosis, or an indication listed in a separate disease category herein.
  • IBS Irritable Bowel Syndrome
  • IBD Inflammatory Bowel Disease
  • biliary colic and other biliary disorders renal colic
  • diarrhea-dominant IBS pain associated with Gl distension
  • ulcerative colitis Crohn's Disease
  • irritable bowel syndrome Celiac disease
  • proctitis proctitis
  • eosinophilic gastroenteritis eosinophilic gastroenteritis
  • mastocytosis or an indication listed in a separate
  • Specific allergic diseases include, but are not limited to: anaphylaxis, allergic rhinitis, allergic dermatitis, allergic urticaria, angioedema, allergic asthma, allergic reactions to: food, drugs, insect bites, pollen; or an indication listed in a separate disease category herein.
  • Specific infection-based diseases include, but are not limited to: sepsis, septic shock, viral diseases, malaria, Lyme disease, ocular infections, conjunctivitis, Whipple Disease, or an indication listed in a separate disease category herein.
  • Specific trauma and tissue injury-based conditions include, but are not limited to: Renal glomerular damage, reperfusion injury (for example to heart, kidney, lung), spinal cord injury, tissue scarring, tissue adhesion, or an indication listed in a separate disease category herein.
  • Specific fibrotic diseases include, but are not limited to: Idiopathic pulmonary fibrosis, liver fibrosis, renal fibrosis, or an indication listed in a separate disease category herein.
  • Specific skin/ dermatological diseases include, but are not limited to: psoriasis, atopic dermatitis, cutaneous lupus, acne, eczema, pruritus, scleroderma, Sweet Syndrome/neutrophilic dermatosis, neutrophilic panniculitis, acrodermatitis (form of pustular psoriasis), or an indication listed in a separate disease category herein.
  • Specific renal diseases include, but are not limited to: acute kidney injury (AKI)
  • AKI acute kidney injury
  • glomerulonephritis IgA nephropathy, crescentic GN, lupus nephritis, HIV associated nephropathy, membraneous nephropathy, C3 glomerulopathy, ANCA vasculitis, diabetic nephropathy, nephrotic syndrome, hypertensive nephrosclerosis, focal segmental glomerulosclerosis, Alport syndrome, Fanconi, syndrome, crystal nephropathy, nephrotic syndrome, amyloidosis, glomerulonephritis in SJIA, or an indication listed in a separate disease category herein.
  • liver diseases include, but are not limited to: liver fibrosis, liver cirrhosis, nonalcoholic steatohepatitis (NASH), or an indication listed in a separate disease category herein.
  • liver fibrosis liver cirrhosis
  • NASH nonalcoholic steatohepatitis
  • Specific oral diseases include, but are not limited to: gingivitis, periodontal disease or an indication listed in a separate disease category herein.
  • Specific metabolic diseases include, but are not limited to: Type 2 diabetes (and resulting complications), hyperlipidemia, non-alcholic fatty liver disease, metabolic syndrome, insulin resistance, obesity, or an indication listed in a separate disease category herein.
  • a proliferative disease selected from a benign or malignant tumor, solid tumor, carcinoma of the brain, kidney, liver, adrenal gland, bladder, breast, stomach, gastric tumors, ovaries, colon, rectum, prostate, pancreas, lung, vagina, cervix, testis, genitourinary tract, esophagus, larynx, skin, bone or thyroid, sarcoma, glioblastomas, neuroblastomas, gastrointestinal cancer, especially colon carcinoma or colorectal adenoma, an epidermal hyperproliferation, psoriasis, prostate hyperplasia, a neoplasia, a neoplasia of epithelial character, adenoma, adenocarcinoma, keratoacanthoma, epidermoid carcinoma, large cell carcinoma, nonsmall-cell lung carcinoma, a mammary carcinoma, follicular carcinoma, undifferent
  • Cardiovascular conditions include, but are not limited to coronary heart disease, acute coronary syndrome, ischaemic heart disease, post-myocardial infarction cardiac remodeling atrial fibrillation, myocardial and vascular fibrosis, vascular wall hypertrophy, endothelial thickening, adverse remodeling, stroke, and the like, or an indication listed in a separate disease category herein.
  • Cardiovascular complications of type 2 diabetes are associated with inflammation, accordingly, the compounds of the present invention may be used to treat diabetes and diabetic complications such as macrovascular disease, hyperglycemia, metabolic syndrome, impaired glucose tolerance, fatty liver disease, cataracts, diabetic neuropathy, diabetic nephropathy, diabetic retinopathy, obesity, dyslipidemia, hypertension, hyperinsulinemia, and insulin resistance syndrome, or an indication listed in a separate disease category herein.
  • diabetes and diabetic complications such as macrovascular disease, hyperglycemia, metabolic syndrome, impaired glucose tolerance, fatty liver disease, cataracts, diabetic neuropathy, diabetic nephropathy, diabetic retinopathy, obesity, dyslipidemia, hypertension, hyperinsulinemia, and insulin resistance syndrome, or an indication listed in a separate disease category herein.
  • the compounds of the present invention are particularly indicated for use in the treatment of neuroinflammatory and neurodegenerative conditions (i.e., disorders or diseases) in mammals including humans such as multiple sclerosis, Alzheimer's disease; Parkinson's disease; brain injury; stroke; cerebrovascular diseases; dementia, acute stress disorder, generalized anxiety disorder, social anxiety disorder, panic disorder, post-traumatic stress disorder and obsessive-compulsive disorder; depression, or an indication listed in a separate disease category herein.
  • neuroinflammatory and neurodegenerative conditions i.e., disorders or diseases
  • mammals including humans such as multiple sclerosis, Alzheimer's disease; Parkinson's disease; brain injury; stroke; cerebrovascular diseases; dementia, acute stress disorder, generalized anxiety disorder, social anxiety disorder, panic disorder, post-traumatic stress disorder and obsessive-compulsive disorder; depression, or an indication listed in a separate disease category herein.
  • a compound of the invention is administered in an amount effective to treat a condition as described herein.
  • the compounds of the invention are administered by any suitable route in the form of a pharmaceutical composition adapted to such a route, and in a dose effective for the treatment intended.
  • Therapeutically effective doses of the compounds required to treat the progress of the medical condition are readily ascertained by one of ordinary skill in the art using preclinical and clinical approaches familiar to the medicinal arts.
  • pharmaceutically acceptable means the substance or composition must be compatible, chemically and/or toxicologically, with the other ingredients comprising a formulation, and/or the mammal being treated therewith.
  • terapéuticaally effective amount means an amount of a compound of the present invention that (i) treats or prevents the particular disease, condition, or disorder, (ii) attenuates, ameliorates, or eliminates one or more symptoms of the particular disease, condition, or disorder, or (iii) prevents or delays the onset of one or more symptoms of the particular disease, condition, or disorder described herein.
  • effective amounts of the compounds of the invention generally include any amount sufficient to detectably modulate vanin-1 activity, and in one embodiment inhibit vanin-1 enzyme, or to alleviate symptoms of diseases associated with vanin-1 activity, and in one embodiment those associated with inhibition of vanin-1 enzyme, or susceptible to vanin-1 activity modulation, in one embodiment inhibition of vanin-1 enzyme.
  • a therapeutically effective amount preferably refers to the amount of a therapeutic agent that increases peak air flow by at least 5%, preferably at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or at least 100%.%.
  • a therapeutically effective amount refers to that amount which has the effect of (1 ) reducing the size of the tumor, (2) inhibiting (that is, slowing to some extent, preferably stopping) tumor metastasis, (3) inhibiting to some extent (that is, slowing to some extent, preferably stopping) tumor growth or tumor invasiveness, and/or (4) relieving to some extent (or, preferably, eliminating) one or more signs or symptoms associated with the cancer.
  • abnormal cell growth refers to cell growth that is independent of normal regulatory mechanisms (e.g., loss of contact inhibition). Abnormal cell growth may be benign (not cancerous), or malignant (cancerous).
  • cancer refers to any malignant and/or invasive growth or tumor caused by abnormal cell growth.
  • cancer refers to solid tumors named for the type of cells that form them, or cancers of blood, bone marrow, or the lymphatic system. Examples of solid tumors include but not limited to sarcomas and carcinomas. Examples of cancers of the blood include but not limited to leukemias, lymphomas and myeloma.
  • cancer includes but is not limited to a primary cancer that originates at a specific site in the body, a metastatic cancer that has spread from the place in which it started to other parts of the body, a recurrence from the original primary cancer after remission, and a second primary cancer that is a new primary cancer in a person with a history of previous cancer of different type from latter one.
  • treating means reversing, alleviating, inhibiting the progress of, delaying the progression of, delaying the onset of, or preventing the disorder or condition to which such term applies, or one or more symptoms of such disorder or condition.
  • treatment refers to the act of treating as “treating” is defined immediately above.
  • treating also includes adjuvant and neo-adjuvant treatment of a subject.
  • reference herein to “treatment” includes reference to curative, palliative and prophylactic treatment, and to the administration of a medicament for use in such treatment.
  • the compounds of the invention may be administered orally.
  • Oral administration may involve swallowing, so that the compound enters the gastrointestinal tract, or buccal or sublingual administration may be employed, by which the compound enters the blood stream directly from the mouth.
  • the compounds of the invention may also be administered directly into the blood stream, into muscle, or into an internal organ.
  • Suitable means for parenteral administration include intravenous, intraarterial, intraperitoneal, intrathecal, intraventricular, intraurethral, intrasternal, intracranial, intramuscular and subcutaneous.
  • Suitable devices for parenteral administration include needle (including microneedle) injectors, needle-free injectors and infusion techniques.
  • the compounds of the invention may also be administered topically to the skin or mucosa, that is, dermally or transdermally.
  • the compounds of the invention can also be administered intranasally or by inhalation.
  • the compounds of the invention may be administered rectally or vaginally.
  • the compounds of the invention may also be administered directly to the eye or ear.
  • the dosage regimen for the compounds and/or compositions containing the compounds is based on a variety of factors, including the type, age, weight, sex and medical condition of the patient; the severity of the condition; the route of administration; and the activity of the particular compound employed. Thus the dosage regimen may vary widely. Dosage levels of the order from about 0.01 mg to about 100 mg per kilogram of body weight per day are useful in the treatment of the above- indicated conditions. In one embodiment, the total daily dose of a compound of the invention (administered in single or divided doses) is typically from about 0.01 to about 100 mg/kg.
  • the total daily dose of the compound of the invention is from about 0.1 to about 50 mg/kg, and in another embodiment, from about 0.5 to about 30 mg/kg (i.e., mg compound of the invention per kg body weight). In one embodiment, dosing is from 0.01 to 10 mg/kg/day. In another embodiment, dosing is from 0.1 to 1 .0 mg/kg/day. Dosage unit compositions may contain such amounts or submultiples thereof to make up the daily dose. In many instances, the administration of the compound will be repeated a plurality of times in a day (typically no greater than 4 times). Multiple doses per day typically may be used to increase the total daily dose, if desired.
  • compositions may be provided in the form of tablets containing from about 0.01 mg to about 500 mg of the active ingredient, or in another embodiment, from about 1 mg to about 100 mg of active ingredient.
  • doses may range from about 0.1 to about 10 mg/kg/minute during a constant rate infusion.
  • “Patient” or “subject” refers to mammals and include, but are not limited to, canine, feline, bovine, caprine, equine, ovine, porcine, rodents, lagomorphs, primates, and the like, and encompass mammals in utero. In one embodiment, humans are suitable subjects. Human subjects may be of either gender and at any stage of development.
  • the term "subject” or “patient” as used herein means any mammalian patient or subject to which the compounds of the invention can be administered.
  • accepted screening methods are employed to determine risk factors associated with a targeted or suspected disease or condition or to determine the status of an existing disease or condition in a subject. These screening methods include, but are not limited to for example, conventional work-ups to determine risk factors that may be associated with the targeted or suspected disease or condition. These and other routine methods allow the clinician to select patients in need of therapy using the methods and compounds of the present invention.
  • inhibitor(s) of vanin-1 enzyme refers to a compound that binds to the vanin-1 enzyme and decreases the resulting enzymatic activity.
  • modulate refers to encompasses either a decrease or an increase in activity or expression depending on the target molecule.
  • other therapeutic agents refers to any therapeutic agent that has been used, is currently used or is known to be useful for treating a disease or a disorder encompassed by the present invention.
  • IC50 refers to an amount, concentration or dosage of a particular test compound that achieves a 50% inhibition of a maximal response in an assay that measures such response. The value depends on the assay used.
  • the invention comprises the use of one or more compounds of the invention for the preparation of a medicament for the treatment of the conditions recited herein.
  • the compound of the invention can be administered as compound per se.
  • pharmaceutically acceptable salts are suitable for medical applications because of their greater aqueous solubility relative to the parent compound.
  • the present invention comprises pharmaceutical compositions.
  • Such pharmaceutical compositions comprise a compound of the invention presented with a pharmaceutically acceptable carrier.
  • the carrier can be a solid, a liquid, or both, and may be formulated with the compound as a unit-dose composition, for example, a tablet, which can contain from 0.05% to 95% by weight of the active compounds.
  • a compound of the invention may be coupled with suitable polymers as targetable drug carriers. Other pharmacologically active substances can also be present.
  • 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 active compounds and compositions for example, may be administered orally, rectally, parenterally, or topically.
  • Oral administration of a solid dose form may be, for example, presented in discrete units, such as hard or soft capsules, pills, cachets, lozenges, or tablets, each containing a predetermined amount of at least one compound of the present invention.
  • the oral administration may be in a powder or granule form.
  • the oral dose form is sub-lingual, such as, for example, a lozenge.
  • the compounds of Formula I are ordinarily combined with one or more adjuvants.
  • Such capsules or tablets may contain a controlled-release formulation.
  • the dosage forms also may comprise buffering agents or may be prepared with enteric coatings.
  • oral administration may be in a liquid dose form.
  • Liquid dosage forms for oral administration include, for example, pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and elixirs containing inert diluents commonly used in the art (e.g., water). Such compositions also may comprise adjuvants, such as wetting, emulsifying, suspending, flavoring (e.g., sweetening), and/or perfuming agents.
  • adjuvants such as wetting, emulsifying, suspending, flavoring (e.g., sweetening), and/or perfuming agents.
  • the present invention comprises a parenteral dose form.
  • Parenteral administration includes, for example, subcutaneous injections, intravenous injections, intraperitoneal injections, intramuscular injections, intrasternal injections, and infusion.
  • injectable preparations e.g., sterile injectable aqueous or oleaginous suspensions
  • suitable dispersing, wetting agents, and/or suspending agents may be formulated according to the known art using suitable dispersing, wetting agents, and/or suspending agents.
  • Topical administration includes, for example, transdermal administration, such as via transdermal patches or iontophoresis devices, intraocular administration, or intranasal or inhalation administration.
  • Compositions for topical administration also include, for example, topical gels, sprays, ointments, and creams.
  • a topical formulation may include a compound that enhances absorption or penetration of the active ingredient through the skin or other affected areas.
  • Typical formulations for this purpose include gels, hydrogels, lotions, solutions, creams, ointments, dusting powders, dressings, foams, films, skin patches, wafers, implants, sponges, fibers, bandages and microemulsions. Liposomes may also be used.
  • Typical carriers include alcohol, water, mineral oil, liquid petrolatum, white petrolatum, glycerin, polyethylene glycol and propylene glycol.
  • Penetration enhancers may be incorporated; see, for example, J. Pharm. Sci., 88(10), 955-958, by Finnin and Morgan (October 1999).
  • Formulations suitable for topical administration to the eye include, for example, eye drops wherein the compound of this invention is dissolved or suspended in a suitable carrier.
  • a typical formulation suitable for ocular or aural administration may be in the form of drops of a micronized suspension or solution in isotonic, pH-adjusted, sterile saline.
  • Other formulations suitable for ocular and aural administration include ointments, biodegradable (e.g., absorbable gel sponges, collagen) and nonbiodegradable (e.g., silicone) implants, wafers, lenses and particulate or vesicular systems, such as niosomes or liposomes.
  • a polymer such as cross-linked polyacrylic acid, polyvinyl alcohol, hyaluronic acid, a cellulosic polymer, for example, hydroxypropyl methyl cellulose, hydroxyethyl cellulose, or methyl cellulose, or a heteropolysaccharide polymer, for example, gelan gum, may be incorporated together with a preservative, such as benzalkonium chloride.
  • a preservative such as benzalkonium chloride.
  • Such formulations may also be delivered by iontophoresis.
  • the active compounds of the invention are conveniently delivered in the form of a solution or suspension from a pump spray container that is squeezed or pumped by the patient or as an aerosol spray presentation from a pressurized container or a nebulizer, with the use of a suitable propellant.
  • Formulations suitable for intranasal administration are typically administered in the form of a dry powder (either alone, as a mixture, for example, in a dry blend with lactose, or as a mixed component particle, for example, mixed with phospholipids, such as phosphatidylcholine) from a dry powder inhaler or as an aerosol spray from a pressurized container, pump, spray, atomizer (preferably an atomizer using electrohydrodynamics to produce a fine mist), or nebulizer, with or without the use of a suitable propellant, such as 1 , 1 , 1 ,2-tetrafluoroethane or 1 , 1 ,1 ,2,3,3, 3-heptafluoropropane.
  • the powder may comprise a bioadhesive agent, for example, chitosan or cyclodextrin.
  • the present invention comprises a rectal dose form.
  • rectal dose form may be in the form of, for example, a suppository. Cocoa butter is a traditional suppository base, but various alternatives may be used as appropriate.
  • Other carrier materials and modes of administration known in the pharmaceutical art may also be used.
  • Pharmaceutical compositions of the invention may be prepared by any of the well-known techniques of pharmacy, such as effective formulation and administration procedures. The above considerations in regard to effective formulations and administration procedures are well known in the art and are described in standard textbooks.
  • pharmaceutically acceptable carrier means a pharmaceutically-acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting the subject agent from one organ, or portion of the body, to another organ, or portion of the body.
  • a pharmaceutically-acceptable material such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting the subject agent from one organ, or portion of the body, to another organ, or portion of the body.
  • Each carrier must be “acceptable” in the sense of being compatible with the other ingredients of the formulation and not injurious to the patient.
  • materials which can serve as pharmaceutically-acceptable carriers include: (1 ) sugars, such as lactose, glucose and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; (10) glycols, such as propylene glycol; (1 1 ) polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; (12) esters, such as ethyl oleate and ethyl laurate; (13) agar; (14) buffering agents, such as magnesium hydroxide, such
  • excipient refers to an inert substance added to a pharmacological composition to further facilitate administration of a compound.
  • excipients include but are not limited to calcium carbonate, calcium phosphate, various sugars and types of starch, cellulose derivatives, gelatin, vegetable oils and polyethylene glycols.
  • the compounds of the present invention can be used, alone or in combination with other therapeutic agents, in the treatment of various conditions or disease states.
  • the compound(s) of the present invention and other therapeutic agent(s) may be may be administered simultaneously (either in the same dosage form or in separate dosage forms) or sequentially. Simultaneous administration may be carried out by mixing the compounds prior to administration or by administering the compounds at the same point in time but at different anatomic sites or using different routes of administration.
  • the present invention includes the use of a combination of an Vanin-1 inhibitor compound as provided in the compound of Formula I and one or more additional pharmaceutically active agent(s). Accordingly, the present invention also includes pharmaceutical compositions comprising an amount of: (a) a first agent comprising a compound of Formula I or a pharmaceutically acceptable salt of the compound; (b) a second pharmaceutically active agent; and (c) a pharmaceutically acceptable carrier, vehicle or diluent.
  • the compounds of the present invention can be administered alone or in combination with one or more additional therapeutic agents.
  • administered in combination or “combination therapy” it is meant that a compound of the present invention and one or more additional therapeutic agents are administered concurrently to the mammal being treated.
  • each component When administered in combination each component may be administered at the same time or sequentially in any order at different points in time. Thus, each component may be administered separately but sufficiently closely in time so as to provide the desired therapeutic effect.
  • the methods of prevention and treatment described herein include use of combination agents.
  • the combination agents are administered to a mammal, including a human, in a therapeutically effective amount.
  • therapeutically effective amount it is meant an amount of a compound of the present invention that, when administered alone or in combination with an additional therapeutic agent to a mammal, is effective to treat the desired disease/condition e.g., inflammatory condition such as systemic lupus erythematosus. See also, T. Koutsokeras and T. Healy, Systemic lupus erythematosus and lupus nephritis, Nat Rev Drug Discov, 2014, 13(3), 173-174, for therapeutic agents useful treating lupus.
  • Non-steroidal anti-inflammatory drugs including but not limited to, nonselective COX1/2 inhibitors such as piroxicam, naproxen, flubiprofen, fenoprofen, ketoprofen, ibuprofen, etodolac (Lodine), mefanamic acid, sulindac, apazone, pyrazolones (such as phenylbutazone), salicylates (such as aspirin); selective COX2 inhibitors such as: celecoxib, rofecoxib, etoricoxib, valdecoxib, meloxicam; Immunomodulatory and/ or anti-inflammatory agents, including but not limited to, methotrexate, leflunomide, ciclesonide chloroquine, hydroxychloroquine, d- penicillamine, auranofin, sulfasalazine,
  • NSAIDs Non-steroidal anti-inflammatory drugs
  • COX1/2 inhibitors such as piroxicam
  • Antimalarials including but not limited to, hydroxychloroquine (Plaquenil) and chloroquine (Aralen), cyclophosphamide (Cytoxan), methotrexate (Rheumatrex), azathioprine (Imuran), mesalamine (Asacol) and sulfasalazine (Azulfidine):
  • Antibiotics including but not limited to, Flagyl or ciprofloxacin;
  • Anti-TNFa agents including but not limited to, infliximab, adalimumab, certolizumab pegol, golimumab and etanercept;
  • Anti-CD20 agents including but not limited to, rituximab, ocrelizumab, ofatumumab and PF-05280586;
  • Antidiarrheals such as diphenoxylate (Lomotil) and loperamide (Imodium);
  • Bile acid binding agents such as cholestyramine, alosetron (Lotronex) and ubiprostone (Amitiza);
  • Laxatives such as Milk of Magnesia, polyethylene glycol (MiraLax), Dulcolax,
  • T lymphocyte activation inhibitors including but not limited to, abatacept
  • Glucocorticoid receptor modulators that may be dosed orally, by inhalation, by injection, topically, rectally, by ocular delivery, including but not limited to, betamethasone, prednisone, hydrocortisone, prednisolone, flunisolide, triamcinoline acetonide, beclomethasone, dipropionate, budesonide, fluticasone propionate, ciclesonide, mometasone furoate, fluocinonide, desoximetasone, methylprednisolone or PF-04171327;
  • Aminosalicyic acid derivatives including but not limited to, sulfasalazine and mesalazine;
  • Anti integrin agents including but not limited to, natalizumab, vedolizumab, PF- 00547659, etrolizumab;
  • a1 - or a2-adrenergic agonist agents including but not limited to: propylhexidrine, phenylephrine, phenylpropanolamine, pseudoephedrine or naphazoline hydrochloride, oxymethazoline hydrochloride, tetrahydrozoline hydrochloride, xylometazoline hydrochloride or ethylnorepinephrine hydrochloride;
  • ⁇ -adrenergic agonists including but not limited to, metaproterenol, isoprotenerol, isoprenaline, albuterol, salbutamol, formoterol, salmeterol, terbutaline, orciprenaline, botolterol mesylate, pirbuterol;
  • Anticholinergic agents including but not limited to, ipratropium bromide, tiotropium bromide, oxitropium bromide, aclindinium bromide, glycopyrrolate, pirenzipine or telenzepine;
  • Leukotriene pathway modulators including but not limited to, 5-LO Inhibitors (such as zileuton), FLAP antagonists (such as veliflapon, fiboflapon), LTD4 antagonists (such as montelukast, zafirlukast or pranlukast;
  • 5-LO Inhibitors such as zileuton
  • FLAP antagonists such as veliflapon, fiboflapon
  • LTD4 antagonists such as montelukast, zafirlukast or pranlukast
  • H1 receptor antagonists including but not limited to, cetirizine, loratidine, desloratidine, fexofenadine, astemizole, azelastine or chlorpheniramine;
  • PDE4 inhibitors including but not limited to, apremilast, roflumilast or AN2728;
  • Vitamin D receptor modulators including but not limited to, paricalcitol
  • Nrf2 pathway activators including but not limited to, fumarates, sulfurophane and bardoxolone methyl
  • RAR-related orphan receptor (ROR) family in particular
  • Modulator and/ or antagonists of the chemokine receptors including but not limited to, CCR2 antagonists (such as CCX140, BMS-741672, PF-4634817, CCX-872, NOX-E36), CCR2/5 antagonists (such as PF-4634817), CCR9 (such as vercirnon, CCX507), CCR1 modulators, CCR4 modulators, CCR5 modulators, CCR6 modulators, CXCR6 modulators, CXCR7 modulators) and CXCR2 modulators (such as danirixin, AZD5069);
  • CCR2 antagonists such as CCX140, BMS-741672, PF-4634817, CCX-872, NOX-E36
  • CCR2/5 antagonists such as PF-4634817
  • CCR9 such as vercirnon, CCX507
  • CCR1 modulators such as CCX140, BMS-741672, PF-4634817, CC
  • Prostaglandins including but not limited to, prostacyclin;
  • PDE5 inhibitors including but not limited to, sildenafil, PF-489791 , vardenafil and tadalafil;
  • Endothelin receptor antagonists including but not limited to, bosentan, ambrisentan, sparsentan, atrasentan, zibotentan and macitentan;
  • Soluble guanylate cyclase activators including but not limited to, riociguat;
  • Interferons including but not limited to, interferon beta-1 a interferon beta-1 b; Sphingosine 1 -phosphate receptor modulators, including but not limited to, fingolimod, ponesimod;
  • Inhibitors of the complement pathway including but not limited to, C5aR antagonists (such as CCX168, PMX-53, NN8210), C5 inhibitors (such as eculizumab), inhibitors of complement factors B and D, inhibitors of MASP2 (such as OMS-721 ) and ARC-1905;
  • Inhibitors of Janus kinases include but not limited to, decernotinib, cerdulatinib, JTE-052, ruxolitinib, tofacitnib, Baricitinib, Peficitinib, GLPG-0634, INCB-47986, INCB-0391 10, PF-04965842, XL-019, ABT-494, R-348, GSK-2586184, AC-410, BMS-91 1543, PF-06651600, and PF-06263276;
  • Inhibitors of other anti-inflammatory or immunomodulatory kinases including but not limited to, spleen tyrosine kinase (SYK) inhibitors, p38 MAP kinase inhibitors (such as PF-3715455, PH-797804, AZD-7624, AKP-001 , UR-13870, FX-005, semapimod, pexmetinib, ARRY-797, RV-568, dilmapimod, ralimetinib), PI3K inhibitors (such as GSK-2126458, pilaralisib, GSK-2269557), PI3Kg and/ or PI3Kd inhibitors (such as CAL- 101/GS-1 101 , duvelisib), JNK inhibitors, ERK1 and/ or 2 inhibitors, IKKb inhibitors, BTK inhibitors, ITK inhibitors, ASK1 inhibitors (such as GS-4997), PKC inhibitors (such as sotrastaurin
  • Antioxidants including but not limited to, myeloperoxidase inhibitors (such as myeloperoxidase inhibitors).
  • NOX4 and other NOX enzymes (such as GKT-137831 ) and N-acetyl cysteine;
  • Inhibitors of IL5 including but not limited to, mepolizumab, reslizumab and benralizumab;
  • Inhibitors of IL4 including but not limited to, pascolizumab, altrakincept and pitrakinra;
  • Inhibitors of IL13 including but not limited to, tralokinumab, anrukinzumab and lebrikizumab;
  • Anti-IL6 agents including but not limited to, tocilizumab, olokizumab, siltuximab, PF-4236921 and sirukumab;
  • Inhibitors/Antagonists of IL17/IL17R including but not limited to, secukinumab, RG-7624, brodalumab and ixekizumab;
  • Antagonists of IL12 and/or IL23 including but not limited to, tildrakizumab, guselkumab, MEDI2070 and AMG 139;
  • Inhibitors of IL33 including but not limited to, AMG 282;
  • Inhibitors of IL9 including but not limited to, MEDI-528;
  • Inhibitors of GM-CSF including but not limited to, MT203;
  • Anti CD4 agents including but not limited to, tregalizumab and rigerimod;
  • CRTH2 antagonists including but not limited to, AZD-1981 ; Inhibitors of B lymphocyte stimulator (BLYS; also known as BAFF), a protein that is often increased in patients with SLE, including but not limited to, belimumab, tabalumab, blisibimod, and atacicept;
  • BLYS B lymphocyte stimulator
  • BAFF B lymphocyte stimulator
  • CD22-specific monoclonal antibodies including but not limited to, epratuzumab; Inhibitors of interferon-a, including but not limited to, sifalimumab and rontalizumab;
  • Inhibitor of type I interferon receptors including but not limited to, MEDI-546; FcyRIIB agonists, including but not limited to, SM-101 ;
  • Hsp10 also known as Chaperonin 10 or EPF
  • INV-103 Heat Shock Protein 10
  • TWEAK receptor TNF superfamily receptor 12A
  • BIIB-023, enavatuzumab, and RG-7212 TNF superfamily receptor 12A
  • Inhibitors of xanthine oxidase including but not limited to, allopurinol, benzbromarone, febuxostat, topiroxostat, tisopurine and inositols;
  • Inhibitors of URAT1 include but not limited to, lesinurad, RDEA 3170, UR1 102 and levotofispam;
  • TLRs toll-like receptors
  • TLR7, TLR8, TLR9 such as IMO-8400, IMO-3100, DV-1 179
  • TLR2 and/ or TLR 4 such as VB-201 , OPN-305
  • TLR7 such as GSK2245035
  • AZD8848 AZD8848
  • TLR9 such as AZD1419
  • SIRT1 Activators SIRT1 , including but not limited to, SRT2104;
  • A3 receptor agonists including but not limited to, CF101 ;
  • agents of use of the treatment of psoriasis including but not limited to, IDP-1 18, LAS41004, LEO 80185, LEO 90100, PH-10, WBI-1001 , C T01959, BT-061 , cimzia, ustekinumab, MK-3222/SCH 900222, ACT-128800, AEB071 , aliireiinoin, ASP015K, Apo805K1 , BMS-582949, FP187, hectorai (doxerca!ciferol), LEO 2281 1 , Ly3009104 (INCB28050), calcipotriene foam (STF 1 15489), iofaciiinib (CP-690,550), 518101 and Cyc!oPsorbTM;
  • Antifibrotic agents including but not limited to: pirfenidone, inhibitors of LOXL2
  • FT-01 1 modulators of epiregulin and/ or TGFp (such as LY- 3016859), modulators of TGFp (such as LY-2382770, fresolimumab);
  • Prolyl hydroxylase inhibitors including but not limited to, GSK1278863, FG-2216, ASP-1517/FG-4592, AKB-6548, JTZ-951 , BAY-85-3934 and DS-1093;
  • Inhibitors of granulocyte macrophage colony-stimulating factor including but not limited to, GSK3196165 (MOR103), PD-0360324 and methosimumab;
  • Inhibitors of MAdCAM and/ or other cell adhesion molecules including but not limited to, PF-00547659;
  • CTGF connective tissue growth factor
  • Inhibitors of cathepsin C including but not limited to, GSK2793660;
  • Inhibitors of soluble epoxide hydrolase including but not limited to, GSK2269557;
  • Inhibitors of the TNFR1 associated death domain protein including but not limited to, GSK2862277;
  • Anti-CD19 agents including but not limited to, MEDI-551 and AMG 729;
  • Anti-B7RP1 agents/ inhibitors of ICOS ligand including but not limited to, MEDI5872 and AMG-557;
  • Inhibitors of thymic stromal lymphoprotein including but not limited to, AMG157; Inhibitors of IL2, including but not limited to, daclizumab;
  • Checkpoint inhibitors including but not limited to those which target CTLA4, PD- 1 , PD-L1 , including but not limited to Ipilimumab, tremelimumab, nivolumab, pembrolizumab, avelumab,
  • Inhibitors of Leucine rich repeat neuronal protein 6A including but not limited to, Anti-Lingo (Biogen);
  • Inhibitors of integrins including but not limited to, alpha-V/beta-6 (STX-100) and alpha-V/beta-3 (VPI-2690B);
  • Anti-CD40L agents including but not limited to, CDP-7657;
  • Modulators of the dopamine D3 receptor including but not limited to, ABT-614; Inhibitors and/ or modulators of galectin-3, including but not limited to, GCS-100 and GR-MD-02;
  • Agents for treating diabetic nephropathy including but not limited to, DA-9801 and ASP-8232;
  • Agents for treating acute kidney injury including but not limited to, THR-184, TRC-160334, NX-001 , EA-230, ABT-719, CMX-2043, BB-3 and MTP-131 ;
  • Modulators of inflammasomes including but not limited to, inhibitors of NLRP3; Modulators of bromodomains, including but not limited to, BRD4;
  • Modulators of short-chain fatty acid receptors including but not limited to, GPR43, GPR109; and Inhibitors of TRP channels, including but not limited to, TRPA1 , TRPC3, TRPC5, TRPC6 and TRPC6.
  • Additional therapeutic agents include anti-coagulant or coagulation inhibitory agents, anti-platelet or platelet inhibitory agents, thrombin inhibitors, thrombolytic or fibrinolytic agents, anti-arrhythmic agents, anti-hypertensive agents, calcium channel blockers (L-type and T-type), cardiac glycosides, diuretics, mineralocorticoid receptor antagonists, NO donating agents such as organonitrates, NO promoting agents such as phosphodiesterase inhibitors, cholesterol/lipid lowering agents and lipid profile therapies, anti-diabetic agents, anti-depressants, anti-inflammatory agents (steroidal and non-steroidal), anti-osteoporosis agents, hormone replacement therapies, oral contraceptives, anti-obesity agents, anti-anxiety agents, anti-proliferative agents, antitumor agents, anti-ulcer and gastroesophageal reflux disease agents, growth hormone and/or growth hormone secretagogues, thyroid mimetics (including thyroid hormone receptor antagonist), anti-infective agents,
  • Agents used in an ICU setting are included, for example, dobutamine, dopamine, epinephrine, nitroglycerin, nitroprusside, etc.
  • Combination agents useful for treating vasculitis are included, for example, azathioprine, cyclophosphamide, mycophenolate, mofetil, rituximab, etc.
  • the present invention provides a combination wherein the second agent is at least one agent selected from a factor Xa inhibitor, an anti-coagulant agent, an anti-platelet agent, a thrombin inhibiting agent, a thrombolytic agent, and a fibrinolytic agent.
  • a factor Xa inhibitor include apixaban and rivaroxaban.
  • suitable anti-coagulants for use in combination with the compounds of the present invention include heparins (e.g., unfractioned and low molecular weight heparins such as enoxaparin and dalteparin).
  • the second agent is at least one agent selected from warfarin, unfractionated heparin, low molecular weight heparin, synthetic pentasaccharide, hirudin, argatrobanas, aspirin, ibuprofen, naproxen, sulindac, indomethacin, mefenamate, droxicam, diclofenac, sulfinpyrazone, piroxicam, ticlopidine, clopidogrel, tirofiban, eptifibatide, abciximab, melagatran, disulfatohirudin, tissue plasminogen activator, modified tissue plasminogen activator, anistreplase, urokinase, and streptokinase.
  • warfarin unfractionated heparin, low molecular weight heparin, synthetic pentasaccharide, hirudin, argatrobanas, aspirin, ibuprofen, naprox
  • the agent is at least one anti-platelet agent.
  • anti-platelet agents are aspirin and clopidogrel.
  • anti-platelet agents denotes agents that inhibit platelet function, for example by inhibiting the aggregation, adhesion or granular secretion of platelets.
  • Agents include, but are not limited to, the various known non-steroidal antiinflammatory drugs (NSAIDS) such as aspirin, ibuprofen, naproxen, sulindac, indomethacin, mefenamate, droxicam, diclofenac, sulfinpyrazone, piroxicam, and pharmaceutically acceptable salts or prodrugs thereof.
  • NSAIDS non-steroidal antiinflammatory drugs
  • NSAIDS aspirin (acetylsalicyclic acid or ASA) and COX-2 inhibitors such as celecoxib or piroxicam are preferred.
  • Suitable platelet inhibitory agents include llb/llla antagonists (e.g., tirofiban, eptifibatide, and abciximab), thromboxane-A2-receptor antagonists (e.g., ifetroban), thromboxane-A2-synthetase inhibitors, PDE3 inhibitors (e.g., Pletal, dipyridamole), and pharmaceutically acceptable salts or prodrugs thereof.
  • llb/llla antagonists e.g., tirofiban, eptifibatide, and abciximab
  • thromboxane-A2-receptor antagonists e.g., ifetroban
  • thromboxane-A2-synthetase inhibitors e.g., Pletal, dip
  • anti-platelet agents or platelet inhibitory agents
  • ADP adenosine diphosphate
  • P2Y12 receptor antagonists include ticagrelor, prasugrel, ticlopidine and clopidogrel, including pharmaceutically acceptable salts or prodrugs thereof.
  • Clopidogrel is an even more preferred agent. Ticlopidine and clopidogrel are also preferred compounds since they are known to be gentle on the gastro-intestinal tract in use.
  • thrombin inhibitors denotes inhibitors of the serine protease thrombin.
  • various thrombin-mediated processes such as thrombin-mediated platelet activation (that is, for example, the aggregation of platelets, and/or the granular secretion of plasminogen activator inhibitor-1 and/or serotonin) and/or fibrin formation are disrupted.
  • thrombin inhibitors are known to one of skill in the art and these inhibitors are contemplated to be used in combination with the present compounds.
  • Such inhibitors include, but are not limited to, boroarginine derivatives, boropeptides, heparins, hirudin, argatroban, and melagatran, including pharmaceutically acceptable salts and prodrugs thereof.
  • Boroarginine derivatives and boropeptides include N-acetyl and peptide derivatives of boronic acid, such as C-terminal alpha-aminoboronic acid derivatives of lysine, ornithine, arginine, homoarginine and corresponding isothiouronium analogs thereof.
  • hirudin includes suitable derivatives or analogs of hirudin, referred to herein as hirulogs, such as disulfatohirudin.
  • thrombolytics or fibrinolytic agents denote agents that lyse blood clots (thrombi).
  • agents include tissue plasminogen activator (natural or recombinant) and modified forms thereof, anistreplase, urokinase, streptokinase, tenecteplase (TNK), lanoteplase (nPA), factor Vila inhibitors, PAI-1 inhibitors (i.e., inactivators of tissue plasminogen activator inhibitors), alpha2- antiplasmin inhibitors, and anisoylated plasminogen streptokinase activator complex, including pharmaceutically acceptable salts or prodrugs thereof.
  • anistreplase refers to anisoylated plasminogen streptokinase activator complex, as described, for example, in EP 028,489, the disclosure of which is hereby incorporated herein by reference herein.
  • urokinase as used herein, is intended to denote both dual and single chain urokinase, the latter also being referred to herein as prourokinase.
  • Suitable anti-arrythmic agents include: Class I agents (such as propafenone); Class II agents (such as metoprolol, atenolol, carvadiol and propranolol); Class III agents (such as sotalol, dofetilide, amiodarone, azimilide and ibutilide); Class IV agents (such as ditiazem and verapamil); K + channel openers such as lAch inhibitors, and lj ⁇ ur inhibitors (e.g., compounds such as those disclosed in WO01/40231 ).
  • Class I agents such as propafenone
  • Class II agents such as metoprolol, atenolol, carvadiol and propranolol
  • Class III agents such as sotalol, dofetilide, amiodarone, azimilide and ibutilide
  • Class IV agents such as ditiazem and verapamil
  • K + channel openers such as l
  • antihypertensive agents include: alpha adrenergic blockers; beta adrenergic blockers; calcium channel blockers (e.g., diltiazem, verapamil, nifedipine and amlodipine); vasodilators (e.g., hydralazine), diruetics (e.g., chlorothiazide, hydrochlorothiazide, flumethiazide, hydroflumethiazide, bendroflumethiazide, methylchlorothiazide, trichloromethiazide, polythiazide, benzthiazide, ethacrynic acid tricrynafen, chlorthalidone, torsemide, fu
  • An exemplary antianginal agent is ivabradine.
  • suitable calcium channel blockers include diltiazem, verapamil, nifedipine and amlodipine and mibefradil.
  • suitable cardiac glycosides include digitalis and ouabain.
  • a compound of the invention may be co-administered with one or more diuretics.
  • suitable diuretics include (a) loop diuretics such as furosemide (such as LASIXTM), torsemide (such as DEMADEXTM), bemetanide (such as BUMEXTM), and ethacrynic acid (such as EDECRINTM); (b) thiazide-type diuretics such as chlorothiazide (such as DIURILTM, ESIDRIXTM or HYDRODIURILTM), hydrochlorothiazide (such as MICROZIDETM or ORETICTM), benzthiazide, hydroflumethiazide (such as SALURONTM), bendroflumethiazide, methychlorthiazide, polythiazide, trichlormethiazide, and indapamide (such as LOZOLTM); (c) phthalimidine- type diuretics such as chlorothia
  • a compound of the invention may be co-administered with a loop diuretic.
  • the loop diuretic is selected from furosemide and torsemide.
  • one or more compounds of the invention may be co-administered with furosemide.
  • one or more compounds of the invention may be co- administered with torsemide which may optionally be a controlled or modified release form of torsemide.
  • a compound of the invention may be co-administered with a thiazide-type diuretic.
  • the thiazide-type diuretic is selected from the group consisting of chlorothiazide and hydrochlorothiazide.
  • one or more compounds of the invention may be co-administered with chlorothiazide.
  • one or more compounds of the invention may be co-administered with hydrochlorothiazide.
  • one or more compounds of the invention may be co-administered with a phthalimidine- type diuretic.
  • the phthalimidine-type diuretic is chlorthalidone.
  • suitable combination mineralocorticoid receptor antagonists include spironolactone and eplerenone.
  • suitable combination phosphodiesterase inhibitors include: PDE3 inhibitors (such as cilostazol); and PDE5 inhibitors (such as sildenafil).
  • the compounds of the present invention may be used in combination with cholesterol modulating agents (including cholesterol lowering agents) such as a lipase inhibitor, an HMG-CoA reductase inhibitor, an HMG-CoA synthase inhibitor, an HMG- CoA reductase gene expression inhibitor, an HMG-CoA synthase gene expression inhibitor, an MTP/Apo B secretion inhibitor, a CETP inhibitor, a bile acid absorption inhibitor, a cholesterol absorption inhibitor, a cholesterol synthesis inhibitor, a squalene synthetase inhibitor, a squalene epoxidase inhibitor, a squalene cyclase inhibitor, a combined squalene epoxidase/squalene cyclase inhibitor, a fibrate, niacin, an ion- exchange resin, an antioxidant, an ACAT inhibitor or a bile acid sequestrant or an agent such as mipomersen.
  • cholesterol modulating agents
  • Suitable cholesterol/lipid lowering agents and lipid profile therapies include: HMG-CoA reductase inhibitors (e.g., pravastatin, lovastatin, atorvastatin, simvastatin, fluvastatin, NK-104 (a.k.a. itavastatin, or nisvastatin or nisbastatin) and ZD-4522 (a.k.a.
  • HMG-CoA reductase inhibitors e.g., pravastatin, lovastatin, atorvastatin, simvastatin, fluvastatin, NK-104 (a.k.a. itavastatin, or nisvastatin or nisbastatin)
  • ZD-4522 a.k.a.
  • rosuvastatin or atavastatin or visastatin
  • squalene synthetase inhibitors squalene synthetase inhibitors
  • fibrates such as questran
  • bile acid sequestrants such as questran
  • ACAT inhibitors MTP inhibitors
  • lipooxygenase inhibitors cholesterol absorption inhibitors
  • cholesteryl ester transfer protein inhibitors cholesteryl ester transfer protein inhibitors
  • Anti-inflammatory agents also include sPLA2 and lpPLA2 inhibitors (such as darapiadib), 5 LO inhibitors (such as atrelueton) and IL-1 and IL-1 r antagonists (such as canakinumab).
  • sPLA2 and lpPLA2 inhibitors such as darapiadib
  • 5 LO inhibitors such as atrelueton
  • IL-1 and IL-1 r antagonists such as canakinumab
  • Atherosclerotic agents include agents that modulate the action of PCSK9, for example, called bococizumab.
  • the compounds of the present invention may be used in combination with anti-diabetic agents, particularly type 2 anti-diabetic agents.
  • anti-diabetic agents include (e.g.
  • Suitable anti-diabetic agents include an acetyl-CoA carboxylase- (ACC) inhibitor such as those described in WO2009144554, WO2003072197, WO2009144555 and WO2008065508, a diacylglycerol O-acyltransferase 1 (DGAT-1 ) inhibitor, such as those described in WO09016462 or WO2010086820, AZD7687 or LCQ908, diacylglycerol O- acyltransferase 2 (DGAT-2) inhibitor, monoacylglycerol O-acyltransferase inhibitors, a PDE10 inhibitor, an AMPK activator, a sulfonylurea (e.g., acetohexamide, chlorpropamide, diabinese, glibenclamide
  • ACC acetyl-CoA carboxylase-
  • DGAT-1 diacylglycerol O-acyltransferase 1
  • GSK1362885 a VPAC2 receptor agonist
  • SGLT2 inhibitors such as those described in E.C. Chao et al. Nature Reviews Drug Discovery 9, 551 -559 (July 2010) including dapagliflozin, canagliflozin, empagliflozin, tofogliflozin (CSG452), ertugliflozin, ASP-1941 , THR1474, TS-071 , ISIS388626 and LX421 1 as well as those in WO2010023594, a glucagon receptor modulator such as those described in Demong, D.E. et al.
  • GPR1 19 modulators particularly agonists, such as those described in WO2010140092, WO2010128425, WO2010128414, WO2010106457, Jones, R.M. et al. in Medicinal Chemistry 2009, 44, 149-170 (e.g. MBX-2982, GSK1292263, APD597 and PSN821 ), FGF21 derivatives or analogs such as those described in Kharitonenkov, A. et al.
  • TGR5 also termed GPBAR1
  • GPBAR1 TGR5 receptor modulators
  • agonists such as those described in Zhong, M., Current Topics in Medicinal Chemistry, 2010, 10(4), 386-396 and INT777
  • GPR40 agonists such as those described in Medina, J.C., Annual Reports in Medicinal Chemistry, 2008, 43, 75-85, including but not limited to TAK-875
  • GPR120 modulators particularly agonists, high affinity nicotinic acid receptor (HM74A) activators, and SGLT1 inhibitors, such as GSK1614235.
  • HM74A high affinity nicotinic acid receptor
  • anti-diabetic agents that can be combined with the compounds of the present invention can be found, for example, at page 28, line 35 through page 30, line 19 of WO201 100561 1 .
  • Preferred anti-diabetic agents are metformin and DPP-IV inhibitors (e.g., sitagliptin, vildagliptin, alogliptin, dutogliptin, linagliptin and saxagliptin).
  • antidiabetic agents could include inhibitors or modulators of carnitine palmitoyl transferase enzymes, inhibitors of fructose 1 ,6-diphosphatase, inhibitors of aldose reductase, mineralocorticoid receptor inhibitors, inhibitors of TORC2, inhibitors of CCR2 and/or CCR5, inhibitors of PKC isoforms (e.g.
  • PKCa ⁇ , PKCy
  • inhibitors of fatty acid synthetase inhibitors of serine palmitoyl transferase
  • modulators of GPR81 , GPR39, GPR43, GPR41 , GPR105, Kv1.3 retinol binding protein 4, glucocorticoid receptor, somatostain receptors (e.g. SSTR1 , SSTR2, SSTR3 and SSTR5), inhibitors or modulators of PDHK2 or PDHK4, inhibitors of MAP4K4, modulators of IL1 family including ILI beta, modulators of RXRalpha.
  • suitable anti-diabetic agents include mechanisms listed by Carpino, P. A., Goodwin, B. Expert Opin. Ther. Pat, 2010, 20(12), 1627-51 .
  • the compounds of this invention may also be used in conjunction with other cardiovascular or cerebrovascular treatments including PCI, stenting, drug eluting stents, stem cell therapy and medical devices such as implanted pacemakers, defibrillators, or cardiac resynchronization therapy.
  • the compounds of the present invention may be used in combination with neuroinflammatory and neurodegenerative agents in mammals. Examples of additional neuroinflammatory and neurodegenerative agents include antidepressants, antipsychotics, anti-pain agents, anti-Alzheimer's agents, and anti-anxiety agents.
  • Examples of particular classes of antidepressants that can be used in combination with the compounds of the invention include norepinephrine reuptake inhibitors, selective serotonin reuptake inhibitors (SSRIs), NK-1 receptor antagonists, monoamine oxidase inhibitors (MAOIs), reversible inhibitors of monoamine oxidase (RIMAs), serotonin and noradrenaline reuptake inhibitors (SNRIs), corticotropin releasing factor (CRF) antagonists, and atypical antidepressants.
  • Suitable norepinephrine reuptake inhibitors include tertiary amine tricyclics and secondary amine tricyclics.
  • Suitable tertiary amine tricyclics and secondary amine tricyclics include amitriptyline, clomipramine, doxepin, imipramine, trimipramine, dothiepin, butriptyline, nortriptyline, protriptyline, amoxapine, desipramine and maprotiline.
  • suitable SSRIs include fluoxetine, fluvoxamine, paroxetine, and sertraline.
  • monoamine oxidase inhibitors include isocarboxazid, phenelzine, and tranylcyclopramine.
  • suitable reversible inhibitors of monoamine oxidase include moclobemide.
  • SNRIs of use in the present invention include venlafaxine.
  • suitable atypical anti-depressants include bupropion, lithium, trazodone and viloxazine.
  • anti-Alzheimer's agents include NMDA receptor antagonists such as memantine; and cholinesterase inhibitors such as donepezil and galantamine.
  • suitable classes of anti-anxiety agents that can be used in combination with the compounds of the invention include benzodiazepines and serotonin 1A receptor (5-HT1A) agonists, and CRF antagonists.
  • Suitable benzodiazepines include alprazolam, chlordiazepoxide, clonazepam, chlorazepate, diazepam, lorazepam, oxazepam, and prazepam.
  • Suitable 5-HT1A receptor agonists include buspirone and ipsapirone.
  • Suitable CRF antagonists include verucerfont.
  • Suitable atypical antipsychotics include paliperidone, ziprasidone, risperidone, aripiprazole, olanzapine, and quetiapine.
  • Suitable nicotine acetylcholine agonists include CP-601927 and varenicline.
  • Anti-pain agents include pregabalin, gabapentin, clonidine, neostigmine, baclofen, midazolam, ketamine and ziconotide.
  • the pharmaceutical combination comprises a therapeutically effective amount of a composition comprising:
  • first compound being a compound of Formula I or a pharmaceutically acceptable salt thereof
  • a second compound being selected from an approved drug or a clinical candidate useful for the treatment of infectious or inflammatory diseases
  • the invention provides for a pharmaceutical combination
  • a first compound the first compound being a compound of Formula I or a pharmaceutically acceptable salt thereof; and a second compound, the second compound being selected from the group consisting of antibodies or small molecules which include but are not limited to those that block the action of specific cytokines such as TNFa, IL12 and/or IL23, or inhibitors of leukocyte recruitment such as modulators of S1 P receptors or integrin antagonists, or selective or non-selective inhibitors of the JAK kinases JAK1 , JAK2, JAK3 and/ or TYK2, inhibitors of leukocyte function such as PDE4 or SMAD7.
  • the invention is directed to a pharmaceutical composition of a compound of Formula I wherein the second compound is selected from
  • an anti-TNFa agent selected from infliximab, adalimumab, golimumab, and certolizumab pegol;
  • an integrin antagonist selected from vedolizumab, etrolizumab, and natalizumab;
  • a SMAD7 antisense oligonucleotides selected from mongersen (g) a SMAD7 antisense oligonucleotides selected from mongersen.
  • kits include at least one compound of the present invention and a package insert or other labeling including directions.
  • spectroscopic means such as nuclear magnetic resonance spectroscopy (e.g., 1 H or 13 C), infrared spectroscopy, spectrophotometry (e.g., UV-visible), mass spectrometry, or by chromatography such as high-performance liquid chromatograpy (HPLC), gas chromatography (GC), gel-permeation chromatography (GPC), or thin layer chromatography (TLC).
  • HPLC high-performance liquid chromatograpy
  • GC gas chromatography
  • GPC gel-permeation chromatography
  • TLC thin layer chromatography
  • Suitable solvents typically are substantially nonreactive with the reactants, intermediates, and/or products at the temperatures at which the reactions are carried out, i.e., temperatures that can range from the solvent's freezing temperature to the solvent's boiling temperature.
  • a given reaction can be carried out in one solvent or a mixture of more than one solvent.
  • suitable solvents for a particular reaction step can be selected.
  • ratios of solvents are given, the ratios are by volume.
  • the derivatives of formula (I), exemplified herein, can be prepared by the procedures described in the general methods presented below or by routine modifications thereof.
  • the present invention also encompasses any one or more of these processes for preparing the derivatives of formula (I), in addition to any novel intermediates used therein.
  • the person skilled in the art will appreciate that the following reactions may be heated thermally or under microwave irradiation.
  • compounds of formula (I), wherein L is N may be prepared from compounds of formulae (IV) and (V), as illustrated by Scheme 1.
  • Hal halogen, typically, CI or F
  • Compounds of formula (IV) may be prepared from an acyl chloride of formula (a) and the amine of formula (b) according to process step (i), an amide bond formation step.
  • Preferred conditions include the reaction in the presence of an organic base, such as triethylamine in THF or EtOAc, at elevated temperatures (60 °C).
  • Compounds of formula (I) may be prepared from compounds of formula (IV) and the amine of formula (V) according to process step (ii).
  • Preferred conditions include the reaction of the amine of formula (V) with the halo compound of formula (IV) in the presence of a suitable organic base, such as DIPEA in a suitable aprotic solvent such DMF or NMP at elevated temperature e.g. 140 °C.
  • Hal halogen, typically CI or F
  • Compounds of formula (VI) are commercially available or may be synthesized by those skilled in the art according to the literature or preparations described herein.
  • Compounds of formula (IV) may be prepared from compounds of formula (VI) according to process step (iii), an amide bond formation step with amines of formula (b), wherein Z defined herein elsewhere, mediated by a suitable combination of amide bond coupling agent and organic base.
  • Typical conditions comprise HATU or HBTU with triethylamine or DIPEA in DCM, DMF or DMA at room or elevated temperatures (e.g.
  • compounds of formula (I) may be prepared in an alternative sequence from compounds of formulae (b) and (VIII) as illustrated by Scheme 3.
  • halogen typically chloro or fluoro.
  • Compounds of formula (VIII) may be prepared from compounds of formula (VII), wherein R x is C1 -C4, typically ethyl or methyl, and compounds of formula (V), wherein the amine of (V) is previously described as "L", according to process step (ii), as described in scheme 1 .
  • Preferred conditions include the reaction of the amine and halide in the presence of an organic base such as triethylamine or DIPEA in a solvent such as NMP under microwave irradiation at elevated temperatures eg 140 °C for up to 1 hr.
  • reaction of the amine and halide is conducted in the presence of an organic base, preferably DIPEA, in a suitable solvent, such as 2-propanol, dioxane, or THF, (optionally with DMSO as a co-solvent), at elevated temperatures, typically between 60 and 80 °C.
  • organic base preferably DIPEA
  • suitable solvent such as 2-propanol, dioxane, or THF, (optionally with DMSO as a co-solvent)
  • Compounds of formula (I) may be prepared from compounds of formula (VIII) by treatment with an amine of formula (b) according to process step (iv).
  • Preferred conditions are reaction in the presence of suitable coupling agent, typically TBD in a suitable aprotic solvent, such as DMF or NMP, at elevated temperature e.g. 50 °C.
  • compounds of formula (I) may be prepared in an alternative sequence from compounds of formulae (IX) and (b) as illustrated by Scheme 4.
  • Hal halogen, typically CI or F
  • Compounds of formula (IX), wherein L is previously described herein, may be prepared from the ester of formula (VIII), wherein R x is C1 -C4 alkyl, typically ethyl or methyl, according to process step (v), a hydrolysis step mediated by an inorganic base.
  • Preferred conditions include aqueous lithium or sodium hydroxide in methanol or ethanol optionally with THF as a co-solvent between room temperature and 60 °C.
  • Compounds of formula (I) may be prepared from compounds of formula (IX) according to process step (iii), an amide bond formation step with an amine of formula (b), wherein Z is previously described herein, mediated by a suitable combination of amide bond coupling agent and organic base, as described in Scheme 2.
  • Preferred conditions comprise HATU with triethylamine or DIPEA at elevated temperatures (e.g. about 60 °C), or using propylphosphonic anhydride in THF with DIPEA and triethylamine at an elevated temperature (e.g.about 60 °C).
  • compounds of formula (I) wherein L is O may be prepared from compounds of formula (IV), wherein Z is previously described herein, and an alcohol of formula (X) as illustrated by scheme 5.
  • Compounds of formula (I) may be prepared from the compounds of formulae (IV) and (X) according to process step (vi).
  • Typical conditions include treatment with a suitable non-nucleophilic base, such as LiHMDS in a suitable solvent such as DMF at temperatures below room tem erature (e.g. 0 °C).
  • Hal halogen, typically CI or F
  • Compounds of formula (XI) may be prepared from compounds of formula (IV), wherein Z is previously described herein, according to process step (vii), a substitution nucleophilic aromatic with an ammonia source as described in Scheme 6.
  • a preferred source of ammonia is an aqueous solution of an ammonium salt such as ammonium hydroxide. This step could also be performed in a different temperature range, typically a temperature above 80 °C and, preferentially, under microwave irradiations above 120 °C.
  • Compounds of formula (I) may be prepared from compounds of formula (XI) according to process step (iii), an amide bond formation step with an acid of formula (XII), mediated by a suitable combination of amide bond coupling agent and organic base, as described in Scheme 2.
  • Formula I can include intermediates of compounds of Formula I.
  • AgF is silver fluoride
  • AIBN is azobisisobutyronitrile
  • Ar is argon
  • Bn is benzyl
  • Boc is tert-butoxycarbonyl
  • Boc 2 O is di-fe/f-butyl dicarbonate
  • tBu is tert-butyl
  • tBuOH is fe/f-butanol
  • CBz-CI is benzyl chloroform ate
  • CS2CO3 is cesium carbonate
  • CsF cesium fluoride
  • DCM is dichloromethane; methylene chloride;
  • DBU is 1 ,8-Diazabicyclo[5.4.0]undec-7-ene
  • DIPEA is N-ethyldiisopropylamine, N,N-diisopropylethylamine;
  • DMA is N,N-dimethylacetamide
  • DMF is N,N-dimethylformamide
  • DMSO dimethyl sulphoxide
  • DPPA is diphenyl phosphoryl azide
  • EDC N-(3-Dimethylaminopropyl)-N'-ethylcarbodiimide hydrochloride and HOBt
  • Et 2 O is diethyl ether
  • EtOAc is ethyl acetate
  • Et 3 N is triethylamine
  • g is gram
  • HATU is 1 -[Bis(dimethylamino)methylene]- H-1 ,2,3-triazolo[4,5-b]pyridinium 3- oxid hexafluorophosphate;
  • HBTU is N,N,N',N'-tetramethyl-O-(1 H-benzotriazol-1 -yl)uronium
  • HCI is hydrochloric acid
  • HCO2H is formic acid
  • HPLC high pressure liquid chromatography
  • H 2 O is water
  • H2O2 is hydrogen peroxide
  • Hr is hour, hrs are hours
  • K2CO3 is potassium carbonate
  • KHSO4 is potassium hydrogen sulphate
  • KOAc is potassium acetate; L is litre;
  • LCMS is liquid chromatography mass spectrometry
  • LiALH 4 is lithium aluminium hydride
  • LiHMDS is lithium bis(trimethylsilyl)amide
  • UOH. H2O is lithium hydroxide monohydrate
  • Li-Selectride® is lithium tri-sec-butylborohydride; m is multiplet;
  • M is molar
  • MeCN is acetonitrile
  • MeMgBr is methyl magnesium bromide
  • MeOH is methanol
  • 2-MeTHF is 2-methyl tetrahydrofuran
  • MgS0 4 is magnesium sulphate
  • MHz is mega Hertz
  • MS m/z is mass spectrum peak
  • MsCI mesyl chloride
  • NaCN sodium cyanide
  • NaBH 4 is sodium borohydride
  • Na 2 C03 is sodium carbonate
  • NaH sodium hydride
  • NaHS0 4 is sodium hydrogen sulfate
  • NaOH sodium hydroxide
  • Na 2 S0 4 is sodium sulphate
  • NBS is N-bromosuccinimide
  • NH 3 is ammonia
  • NH 4 CI is ammonium chloride
  • NH 4 HC0 3 is ammonium hydrogen carbonate
  • NH 2 NH 2 .H 2 0 is hydrazine hydrate
  • NH2OH.HCI is hydroxylamine hydrochloride
  • NH 4 OH is ammonium hydroxide
  • NH OAc is ammonium acetate
  • NMP is 1 -methyl-2-pyrrolidinone
  • NMR nuclear magnetic resonance
  • Pd/C is palladium on carbon
  • Pd(dppf)CI 2 is [1 , 1 '-bis(diphenylphosphino)ferrocene]dichloropalladium(ll);
  • Pd(OH) 2 is palladium hydroxide
  • Pd(OAc) 2 is palladium acetate
  • Pet. Ether is petroleum ether
  • pH is power of hydrogen
  • ppm is parts per million
  • Pt0 2 is platinum (IV) oxide
  • RT retention time
  • SCX is strong cation exchange
  • t is triplet
  • T3P is propylphosphonic anhydride
  • TBAF is tert-butyl ammonium fluoride
  • TBD is 1 ,5,7-triazabicyclo[4.4.0]dec-5-ene
  • TBME is tert-butyl dimethyl ether
  • TFA is trifluoroacetic acid
  • THF is tetrahydrofuran
  • Ti(OiPr) 4 is titanium (IV) propoxide
  • TPTU is 2-(2-pyridon-1 -yl)-1 , 1 ,3,3-tetramethyluronium tetrafluoroborate, an amide coupling agent.
  • ⁇ _ is microlitre
  • tautomers may be recorded within the NMR data; and some exchangeable protons may not be visible.
  • Method CA-A column: Lux Cellulose-2 150x4.6mm I.D., 3pm; mobile phase: 40% EtOH (0.05% DEA) in CO 2 ; flow rate of 2.5 mL/min at 40 °C.
  • Method CA-B column: Chiralcel OD-3 150x4.6mm I.D., 3pm; mobile phase:
  • Method CA-F column: Chiralpak AD-3 150x4.6mm I.D., 3pm; mobile phase: iso-propanol (0.05% DEA) in CO 2 (from 5% to 40% in 5.5 min); flow rate: 2.5 mL/min at 35 °C
  • Method CP-A C2 250mm * 30mm, 10 ⁇ column, eluting with 55% EtOH (0.1 %NH 3 H 2 0) in C0 2 at a flow rate of 80 mL/min.
  • Method CP-B OD 250mm * 30mm, 10pm column, eluting with 40% EtOH (0.1 % ⁇ 3 ⁇ 2 ⁇ ) in CO 2 at a flow rate of 80 mL/min.
  • Method CP-C AD 250mm * 30mm, 10pm column, eluting with 50% of EtOH (0.05% ⁇ 3 ⁇ 2 ⁇ ) in CO 2 at a flow rate of 80 mL/min.
  • Method CP-D AD 250mm * 30mm, 5 m column, eluting with 40% of iso-propanol (0.05% DEA) in CO 2 at a flow rate of 50 mL/min.
  • Method CP-F OJ 250mm * 30mm, 5pm column, eluting with 25% MeOH (0.05% DEA) in CO 2 at a flow rate of 60 mL/min.
  • Method CP-G AY 250mm * 30mm, 10 m column, eluting with 45% iso-propanol (0.1 % ⁇ 3 ⁇ 2 ⁇ ) in CO 2 at flow rate of 80 mL/min.
  • reaction conditions length of reaction and temperature
  • purifications may vary between experiments: in general, sorbents, solvents and the solvent ratios used for eluants/gradients were chosen to provide appropriate R f s or retention times.
  • HPLC purifications may be effected in a variety of ways, including the use of normal stationary phases, reverse stationary phases, chiral stationary phases, and supercritical eluants. The appropriate choices of conditions for chromatographic and HPLC purifications will be discerned by one skilled in the art.
  • the content of the erlenmeyer was transferred into a 10-L tank equipped with a jacket, a short-path distillation set-up connected to a 1 L flask and a mechanical stirrer.
  • the erlenmeyer and the flask were rinsed with water until all the solids have been transferred.
  • the resulting suspension was stirred at 50 °C under vacuum for 5 hrs and the mixture then allowed to cool to rt.
  • Water (2 L) was added, the mixture stirred for 2 hrs then the solid was filtered off, washing through with water (500 ml_).
  • the solid was dried in vacuo.
  • the filtrate was concentrated in vacuo and the resulting solid filtered off and dried to provide additional product.
  • 6-Methyl-3-pyridinemethanamine (1 .0 g, 8.19 mmol) was added to a solution of ethyl 2-chloropyrimidine-5-carboxylate (1 .53 g, 8.19 mmol) and DIPEA (1 .59 g, 12.3 mmol) in 2-propanol (8 mL) and the resulting mixture was heated under reflux for 8 hrs. The cooled mixture was concentrated under reduced pressure and the residue purified by column chromatography on silica gel eluting with EtOAc: Pet. Ether (0: 100 to 30:70) to afford the title compound as a yellow solid, 1 .5 g, 67%.
  • Preparation 16 was further purified by SFC separation using the following: AD 250mm * 50mm, 10pm column; 60% EtOH (0.1 %NH 3. H 2 O) in C0 2 at 200ml/min, 38 ° C; to provide ethyl 2- ⁇ [(1 S)-1 -(6-methylpyridin-3-yl)ethyl]amino ⁇ pyrimidine-5-carboxylate as a yellow oil (RT: 6.31 1 min, 5.73 g, 44%).
  • DPPA (2330 mg, 8.45 mmol) was added to a solution of Et 3 N (dried over KOH) (855 mg, 8.45 mmol) and 2-methyl-2-(pyrimidin-5-yl)propanoic acid (Preparation 36, 1 170 mg, 7.04 mmol) in distilled f-BuOH (20 mL) and the mixture heated to 1 10 °C for 16 hrs. The cooled mixture was poured into aq. NH 4 CI solution and extracted with EtOAc (100 mL x 3). The combined organic extracts were dried (MgS0 4 ), filtered and evaporated under reduced pressure. The residue was purified by column chromatography on silica gel eluting with 0-40% EtOAc in Pet.
  • the reaction mixture was again cooled to -70 °C and an additional portion of LiHMDS (1 M in THF, 56.2 mL, 56.2 mmol) was added drop wise. After addition, the reaction was allowed to warm to 20 °C slowly and stirred for an additional hour. The reaction was poured into saturated NH 4 CI solution (60 mL) the mixture extracted with EtOAc (300 mL x 3). The combined organic extracts were washed with brine, dried (Na 2 S0 4 ) filtered and concentrated in vacuo. The crude product was purified by column chromatography on silica gel eluting with pet.
  • n-BuLi (25.4 ml_, 63.5 mmol, 2.5 M in hexane) was added drop wise to a -78 °C solution of 2-bromopyrazine (10.1 g, 63.5 mmol) in toluene (150 ml_) under N 2 .
  • a solution of N-cyclobutylidene-2-methylpropane-2-sulfinamide (Preparation 50, 10.0 g, 57.71 mmol) in toluene (50 ml_) was added slowly.
  • the resulting dark red solution was stirred for 1 hr at -78 °C.
  • the reaction was quenched by the addition of sat.
  • Dimethyl zinc (21 .3 mL, 21 .3 mmol, 1 .0 M in toluene) was added in an oven- dried and N 2 -purged flask, followed by MeMgBr (6.25 ml_,18.7 mmol, 3.0 M in ether) over 1 min with stirring at 15°C and the solution allowed to stir for 20 mins.
  • Me 2 Zn (5.63 mmol, 1 M in toluene, 5.63 ml_) was added to an oven-dried and N 2 - purged round-bottomed flask and MeMgBr (0.497 mmol, 3 M in ether) was added over 1 min. The solution was stirred at 15 °C for 30 min.
  • Boc-anhydride (10.79 ml_, 49.45 mmol) was added drop wise to an ice-cooled stirred solution of the gum in 50% EtOAc/water (100 ml) and Na 2 C0 3 (10.48 g, 98.89 mmol) and the resulting reaction was stirred at rt for 15 hrs.
  • the mixture was extracted with EtOAc, the combined organic extracts washed with brine, dried (Na 2 SO 4 ) and concentrated in vacuo.
  • the crude product was purified by column chromatography on silica gel eluting with EtOAc: Hexane (20:80) to afford the title compound as a yellow liquid, 3.5 g, 49%.

Abstract

Compounds, pharmaceutically acceptable salts thereof, are disclosed wherein the compounds have the structure of (I) as defined in the specification. Corresponding pharmaceutical compositions, methods of treatment, methods of synthesis, and intermediates are also disclosed.

Description

NOVEL PYRIMIDINE CARBOXAMIDES
AS INHIBITORS OF VANIN-1 ENZYME
The present invention relates to novel heterocyclic compounds, or pharmaceutically acceptable salts thereof, and pharmaceutical compositions comprising the same. The present invention also relates to methods of treating a subject by administering a therapeutically effective amount of these compounds, or salts thereof, to a subject. In general, these compounds act as inhibitors of vanin-1 enzyme.
BACKGROUND
Vanin-1 is a cell surface associated, giycosyiphosphatidyS inositol (GPi) ~ anchored protein which is expressed at high levels in kidney, liver and the small intestine. Vanin-1 expression can be up-regulated in multiple cell types under various inflammatory and oxidative stress conditions. Soluble Vanin-1 is found in serum of mice and humans indicating that Vanin-1 can be shed off the cell surface (Rommelaere S, et al. PPARalpha regulates the production of serum Vanin-1 by liver. FEBS Lett. 2013 Nov 15;587(22):3742-8). Three Vanin family members have been described in humans (Vanin-1 , Vanin-2 and Vanin-3) and these are classified as members of the biotinidase branch of the nitrilase superfamily (Kaskow BJ, et al. Diverse biological activities of the vascular non-inflammatory molecules - the Vanin pantetheinases. Biochem Biophys Res Commun. 2012 Jan 13;417(2):653-8).
To date the only known substrate for Vanin-1 is pantetheine and it is believed that Vanin-1 acts as the predominant pantetheinase in vivo catalyzing its hydrolysis to produce pantothenic acid (vitamin B5) and cysteamine (Pitari G, et al. Pantetheinase activity of membrane-bound vanin-1 ; lack of free cysteamine in tissues of vanin-1 deficient mice. FEBS Lett. 2000;483: 149-154). These products impact diverse biological processes. Panthothenic acid is a necessary factor in the synthesis of Coenzyme A (CoA), a cofactor involved in many metabolic processes such as fatty acid synthesis and oxidation of pyruvate. The amino-thiol cysteamine, the second product of Vanin-1 enzymatic reaction, impacts the cellular redox status (Kaskow BJ, et al. Diverse biological activities of the vascular non-inflammatory molecules - the Vanin pantetheinases. Biochem Biophys Res Commun. 2012 Jan 13;417(2):653-8 and Nitto T, Onodera K. The Linkage between coenzyme A metabolism and inflammation: roles of Pantetheinase. Journal of pharmacological sciences 2013: 123: 1-8). Vanin-1 -deficient mice show no developmental defects nor do they show obvious spontaneous phenotype. However, diverse Vanin-1 -dependent phenotypes are revealed in situations of metabolic challenge and/or oxidative stress and tissue damage. Vanin-1 -deficient mice exhibit resistance to oxidative tissue injury caused by y-irradiation or by the administration of paraquat which is correlated with significantly increased glutathione levels (Berruyer C, et al. Vanin-1 -/- mice exhibit a glutathione mediated tissue resistance to oxidative stress. Mol Cell Biol. 2004;24:7214-7224). Vanin-1 deficient animals are also protected against multiple mouse models of IBD including DSS (dextran sulfate) and TNBS (trinitrobenzene sulfonate) colitis as evidenced by preserved mucosal barrier and reduced inflammatory infiltrate (Berruyer C, et al. Vanin-1 licenses inflammatory mediator production by gut epithelial cells and controls colitis by antagonizing peroxisome proiiferator-activated receptor γ activity. J Exp Med. 2008;203:2817-2827 and et a!. Vanin-1 -/- mice show decreased NSAID- and Schistosoma-induced intestinal inflammation associated with higher glutathione stores. J Clin Invest. 2004; 1 13:591-597). In humans, Vanin-1 expression is significantly increased in the colonic mucosa from IBD patients and functional polymorphisms in the regulatory regions of the Vanin-1 gene are associated with susceptibility to inflammatory bowel diseases (GensoNen T, et.al. Functional polymorphisms in the regulatory regions of the VNN1 gene are associated with susceptibility to inflammatory bowel diseases. Inflamm Bowel Dis. 2013 Oct; 19(1 1 ):2315-25). In addition, patients with ulcerative colitis have an increased risk of developing colorectal cancer and Vanin- 1 knock-out mice exhibit drastically reduced incidence of tumors in colitis associated cancer model (Pouyet L, et al. Epithelial vanin-1 controls inflammation-driven carcinogenesis in the colitis-associated colon cancer model. Inflamm Bowel Dis. 2010 Jan; 16(1 ):96-104).
Vanin-1 is a key activator for hepatic gluconeogenesis (Chen S, et al. Vanin-1 is a key activator for hepatic gluconeogenesis. Diabetes. 2014 Jun;63(6):2073-85. doi: 10.2337/db13-0788. Epub 2014 Feb 18). Vanin-1 regulates the activation of smooth muscle cells in vitro and development of neointimal hyperplasia in response to carotid artery ligation in vivo. Polymorphysims in VNN1 gene are associated with blood pressure and HDL levels further supporting Vanin-1 's role in cardiovascular diseases. Vanin-1 deficiency prevents mice from the development of adrenocortical neoplasia in Sf-1 transgenic mice suggesting a role for Vanin-1 in certain cancers. In the context of infection, Vanin-1 deficiency reduces granuloma formation and tissue damage against Coxieila burnetii, a bacterium that causes Q fever. Vanin-1 is highly up-regulated in psoriatic skin lesions compared with normal individuals. Vnn-1 gene expression is also up-regulated in whole blood of patients with pediatric immune thrombocytopenia (ITP) where overexpression of VNN1 , is associated with progression to chronic ITP. In addition, elevated Vanin-1 has been detected in the urine of patients with multiple renal disorders including systemic lupus erythematosus, nephrotoxicant-induced renal injury and type 2-diabetes (Rommelaere S, et al. PPARalpha regulates the production of serum Vanin-1 by liver. FEBS Lett. 2013 Nov 15;587(22):3742-8).
There is a need for novel and potent small molecule compounds which act as inhibitors of vanin-1 enzyme. Compounds reported as having vanin activity include, for example, those disclosed in WO 2014/048547. Co-pending U.S. Provisional Application 62/167962, filed by Pfizer Inc on May 29, 2015, and co-pending U.S. Provisional Application 62/195005, filed by Pfizer Inc on July 21 , 2015, both of which are incorporated herein by reference in its entirety.
SUMMARY
This invention relates to a com ound of Formula I,
Figure imgf000004_0001
I wherein
G is a 6-membered heteroaryl, with one, two or three N, wherein the
heteroaryl is optionally substituted with one, two or three substituents selected from halogen, OH, cyano, Ci-C4alkyl, -NR8aC(O)R8b, -NR8aSO2R8b - (CR6aR6b)tC(O)N(R8a)2, -C(O)OH, -N(R8a)2, -(CR6aR6b)tSO2R8b, - (CR6aR6b)tSO2N(R8a)2, Ci-C alkoxy, -S(Ci-C3alkyl) or C3-C5cycloalkyl, wherein the alkyl, cycloalkyl and alkoxy are optionally substituted with one, two or three halogen, OH, OCH3, or C3-C5cycloalkyl;
L is NH or O;
Z is a bond; -(CR5aR5b)q-; -CH2(CR5aR5b)m-; or -(CR5aR5b)m-W-(CR5aR5b)n-, wherein W is S, O or NR7;
or a pharmaceutically acceptable salt of said compound or a tautomer of said compound or said salt. The invention also provides for pharmaceutical compositions comprising the compounds, methods of using the compounds, combination therapies utilizing the compounds and other therapeutic agents and methods of preparing the compounds. The invention also provides for intermediates useful in the preparation of the compounds of the invention.
In particular, the compounds of the invention, or pharmaceutically acceptable salts thereof, may inhibit the vanin-1 enzyme. Such compounds may therefore be useful for treating diseases or disorders that are mediated by, or otherwise associated with, inhibition of the vanin-1 enzyme, the method comprising administering to a subject in need thereof, an effective amount of a compound of the invention.
In another embodiment, the present invention further provides a method of inhibiting vanin-1 enzyme in a cell, comprising contacting the cell with a therapeutically effective amount of a compound of Formula I, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising a therapeutically effective amount of a compound of Formula I, or a pharmaceutically acceptable salt thereof.
Inhibitors of vanin-1 enzyme may be used in the treatment of a variety of diseases or disorders related to systemic or tissue inflammation, inflammatory responses to infection or hypoxia, cellular activation and proliferation, lipid metabolism, fibrosis and in the treatment of viral infections. Therefore, inhibition of Vanin-1 would have the potential for multiple therapeutic indications over a wide range of unmet needs.
BRIEF DESCRIPTION OF FIGURES
Figure 1 is a PXRD pattern of Example 142.
Figure 2 is an X-ray crystal structure (ORTEP drawing) of Example 145a, 8-oxa- 2-azaspiro[4.5]dec-2-yl(2-{[(1 S)-1 -(pyrazin-2-yl)ethyl]amino}pyrimidin-5-yl)methanone methanesulfonate.
DETAILED DESCRIPTION
The present invention relates to novel heterocyclic compounds of the invention which, in general, inhibit vanin-1 enzyme. The present invention may be understood more readily by reference to the following detailed description of exemplary embodiments of the invention and the examples included therein. It is to be understood that this invention is not limited to specific methods of synthesis, which may of course vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting.
All patents, patent applications and references referred to herein are hereby incorporated by reference in their entirety.
Other features and advantages of this invention will be apparent from this specification and the appendent claims which describe the invention. There are many features of this invention that are not necessarily fully captured by the claims. It is understood, however, that all such novel subject matter is part of the invention.
Definitions
Unless otherwise defined herein, scientific and technical terms used in connection with the present invention have the meaning commonly understood by those of ordinary skill in the art. As used in the specification and the appended claims, the singular forms "a", "an", and "the" include plural referents unless the context clearly dictates otherwise.
The term "about" refers to a relative term denoting an approximation of plus or minus 10% of the nominal value it refers, in one embodiment, to plus or minus 5%, in another embodiment, to plus or minus 2%. For the field of this disclosure, this level of approximation is appropriate unless the value is specifically stated require a tighter range.
The term "alkyl" refers to a linear or branched saturated hydrocarbon moiety, consisting solely of carbon and hydrogen atoms. In one embodiment from one to six carbon atoms; and in another embodiment from one to four carbon atoms; and in another embodiment one to three carbon atoms. Non-limiting examples of such substituents include methyl, ethyl, propyl (including n-propyl and isopropyl), butyl (including n-butyl, isobutyl, sec-butyl and fe/f-butyl), pentyl, isoamyl, hexyl and the like. As appropriate, an alkyl may be optionally substituted at each carbon as defined in the claims. Typical substitution includes, but is not limited to, fluoro, chloro, OH, cyano, alkyl (optionally substituted), alkoxy, cycloalkyl and the like. In some instances, the number of carbon atoms in a hydrocarbon substituent (i.e., alkyl, cycloalkyl, etc.) is indicated by the prefix "Cx-Cy-" or "Cx-y", wherein x is the minimum and y is the maximum number of carbon atoms in the substituent. Thus, for example, "Ci -C6-alkyl" or "C-i-6 alkyl" refers to an alkyl substituent containing from 1 to 6 carbon atoms. Illustrating further, C3-C6-cycloalkyl or C3-6-cycloalkyl refers to saturated cycloalkyl containing from 3 to 6 carbon ring atoms.
The term "cycloalkyl" refers to a nonaromatic ring containing 3 to 12 carbons that is fully hydrogenated consisting of mono-, bi- or tricyclic rings. Accordingly, a cycloalkyl may be a single ring, which typically contains from 3 to 7 ring atoms. Examples include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl. Alternatively, 2 or 3 rings may be fused together, such as bicyclodecanyl and decalinyl. The term "cycloalkyl" also includes bridged bicycloalkyl systems such as, but not limited to, bicyclo[2.2.1 ]heptane and bicyclo[1 .1 .1 ]pentane. The cycloalkyl group may be optionally substituted as described herein, as appropriate, by 1 to 5 suitable substituents as defined herein, including but not limited to, for example, Ci-C4alkyl, oxo, OH, CH2OH, halogen, Ci-C4alkoxy, cyano or C(O)NH2, wherein said alkyl and alkoxy are optionally substituted with OH, halogen, C(O)NH2, C(O)NHCH3, C(O)N(CH3)2, - S(Ci-C alkyl) or C3-C5cycloalkyl.
The term "heterocycloalkyl" means a monovalent saturated moiety, consisting of one to three rings, incorporating one, two, three or four heteroatoms (selected from N, O or S) and three to 12 carbon atoms. The heterocycloalkyl may be optionally substituted as defined herein. Examples of heterocycloalkyl moieties include, but are not limited to, optionally substituted piperidinyl, piperazinyl, homopiperazinyl, azepinyl, pyrrolidinyl, imidazolidinyl, morpholinyl, quinuclidinyl, thiadiazolylidinyl, benzothiazolidinyl, benzoazolylidinyl, dihydrofuryl, tetrahydrofuryl, dihydropyranyl, tetrahydropyranyl, thiamorpholinyl, thiamorpholinylsulfoxide, thiamorphilinylsulfone, dihydroquinolinyl, tetrahydroquinolinyl, tetrahydrisoquinolinyl, and the like. Heterocycloalkyls may be optionally substituted, as appropriate, by 1 to 5 suitable substituents as defined herein such as including but not limited to, for example, C-i - C4alkyl, oxo, OH, CH2OH, halogen, Ci-C4alkoxy, cyano or C(O)NH2, wherein said alkyl and alkoxy are optionally substituted with OH, halogen, C(O)NH2, C(O)NHCH3, C(O)N(CH3)2, -S(Ci-C alkyl) or C3-C5cycloalkyl. The term "heterocycloalkyl" also includes fused ring systems with, for example, a cycloalkyl, aryl or heteroaryl. Unless otherwise indicated, the term "heteroalkyl," by itself or in combination with another term, means, unless otherwise stated, a saturated, straight or branched chain hydrocarbon radical consisting of the stated number of carbon atoms and from one to three heteroatoms selected from the group consisting of 0, N and S, and wherein the nitrogen and sulfur atoms may optionally be oxidized and the nitrogen heteroatom may optionally be quaternized. The heteroatom(s) 0, N and S may be placed at any interior position of the heteroalkyl group. The heteroatom S may be placed at any position of the heteroalkyl group, including the position at which the alkyl group is attached to the remainder of the molecule. Up to two heteroatoms may be consecutive. The term "alkoxy" and "alkyloxy", which may be used interchangeably, refers to a moiety of the formula -OR, wherein R is a straight chain saturated alkyl or branched chain saturated alkyl moiety, as defined herein, bonded through an oxygen atom. The alkoxy group may be optionally substituted as defined herein. Non-limiting examples of such alkoxy groups are methoxy, ethoxy, propoxy, isopropoxy, butoxy, isobutoxy, tertiary butoxy, pentoxy and the like.
The term "aryl" means a carbocyclic aromatic system containing one or two rings wherein such rings may be fused. If the rings are fused, one of the rings must be fully unsaturated and the fused ring(s) may be fully saturated, partially unsaturated or fully unsaturated. The term "fused" means that a second ring is present (i.e., attached or formed) by having two adjacent atoms in common (i.e., shared) with the first ring. The term "fused" is equivalent to the term "condensed". The aryl group may be optionally substituted as defined herein. The term "aryl" embraces aromatic radicals such as phenyl, naphthyl, tetrahydronaphthyl, indanyl, biphenyl, benzo[b][1 ,4]oxazin-3(4H)-onyl, 2,3-dihydro-1 /-/ indenyl and 1 ,2,3,4-tetrahydronaphthalenyl. Aryls may be optionally substituted, as appropriate, by 1 to 5 suitable substituents as defined herein such as Ci-C4alkyl, oxo, OH, CH2OH, halogen, Ci-C4alkoxy, cyano or C(O)NH2, wherein said alkyl and alkoxy are optionally substituted with OH, halogen, C(O)NH2, C(O)NHCH3, C(O)N(CH3)2, -S(Ci-C alkyl) or C3-C5cycloalkyland the like.
Unless otherwise indicated, the term "heteroalkyl," by itself or in combination with another term, means, unless otherwise stated, a saturated, straight or branched chain hydrocarbon radical consisting of the stated number of carbon atoms and from one to three heteroatoms selected from the group consisting of O, N and S, and wherein the nitrogen and sulfur atoms may optionally be oxidized and the nitrogen heteroatom may optionally be quaternized. The heteroatom(s) O, N and S may be placed at any interior position of the heteroalkyl group. The heteroatom S may be placed at any position of the heteroalkyl group, including the position at which the alkyl group is attached to the remainder of the molecule. Up to two heteroatoms may be consecutive.
The term "heteroaryl" refers to an aromatic ring structure containing from 5 to 6 ring atoms in which at least one of the ring atoms is a heteroatom (i.e., oxygen, nitrogen, or sulfur), with the remaining ring atoms being independently selected from the group consisting of carbon, oxygen, nitrogen, and sulfur. Examples of heteroaryl substituents include 6-membered ring substituents such as pyridyl, pyrazyl, pyrimidinyl, and pyridazinyl; and 5-membered ring substituents such as triazolyl, imidazolyl, furanyl, thiophenyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl, 1 ,2,3-, 1 ,2,4-, 1 ,2,5-, or 1 ,3,4-oxadiazolyl and isothiazolyl. In a group that has a heteroaryl substituent, the ring atom of the heteroaryl substituent that is bound to the group may be one of the heteroatoms, or it may be a ring carbon atom. Similarly, if the heteroaryl substituent is in turn substituted with a group or substituent, the group or substituent may be bound to one of the heteroatoms, or it may be bound to a ring carbon atom. The term "heteroaryl" also includes pyridyl /V-oxides and groups containing a pyridine /V-oxide ring.
Further examples include furyl, thienyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, thiadiazolyl, pyridinyl, pyridazinyl, pyrimidinyl, pyrazinyl, pyridin-2(1 /-/)-onyl, pyridazin-2(1 /-/)-onyl, pyrimidin- 2(1 /-/)-onyl, pyrazin-2(1 /-/)-onyl, imidazo[1 ,2-a]pyridinyl, pyrazolo[1 ,5-a]pyridinyl, 5,6,7,8- tetrahydroisoquinolinyl, 5,6,7,8-tetrahydroquinolinyl, 6,7-dihydro-5/-/- cyclopenta[jb]pyridinyl, 6,7-dihydro-5/-/-cyclopenta[c]pyridinyl, 1 ,4,5,6- tetrahydrocyclopenta[c]pyrazolyl, 2,4,5,6-tetrahydrocyclopenta[c]pyrazolyl, 5,6-dihydro- 4H-pyrrolo[1 ,2-Jb]pyrazolyl, 6,7-dihydro-5H-pyrrolo[1 ,2-b][1 ,2,4]triazolyl, 5,6,7,8- tetrahydro-[1 ,2,4]triazolo[1 ,5-a]pyridinyl, 4,5,6,7-tetrahydropyrazolo[1 ,5-a]pyridinyl, 4,5,6,7-tetrahydro-1 /-/-indazolyl and 4,5,6,7-tetrahydro-2/-/-indazolyl. The heteroaryl can be optionally substituted, as appropriate, by 1 to 5 suitable substituents as defined herein such as Ci-C4alkyl, oxo, OH, CH2OH, halogen, Ci-C4alkoxy, cyano or C(O)NH2, wherein said alkyl and alkoxy are optionally substituted with OH, halogen, C(O)NH2, C(O)NHCH3, C(O)N(CH3)2, -S(Ci-C alkyl) or C3-C5cycloalkyl and the like.
Examples of single-ring heteroaryls and heterocycloalkyls include furanyl, dihydrofuranyl, tetrahydrofuranyl, thiophenyl, dihydrothiophenyl, tetrahydrothiophenyl, pyrrolyl, isopyrrolyl, pyrrolinyl, pyrrolidinyl, imidazolyl, isoimidazolyl, imidazolinyl, imidazolidinyl, pyrazolyl, pyrazolinyl, pyrazolidinyl, triazolyl, tetrazolyl, dithiolyl, oxathiolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, thiazolinyl, isothiazolinyl, thiazolidinyl, isothiazolidinyl, thiaoxadiazolyl, oxathiazolyl, oxadiazolyl (including oxadiazolyl, 1 ,2,4-oxadiazolyl, 1 ,2,5-oxadiazolyl, or 1 ,3,4-oxadiazolyl), pyranyl (including 1 ,2-pyranyl or 1 ,4-pyranyl), dihydropyranyl, pyridinyl, piperidinyl, diazinyl (including pyridazinyl, pyrimidinyl, piperazinyl, triazinyl (including s-triazinyl, as-triazinyl and v-triazinyl), oxazinyl (including 2/-/-1 ,2-oxazinyl, 6/-/-1 ,3-oxazinyl, or 2H- 1 ,4-oxazinyl), isoxazinyl (including o-isoxazinyl or p-isoxazinyl), oxazolidinyl, isoxazolidinyl, oxathiazinyl (including 1 ,2,5-oxathiazinyl or 1 ,2,6-oxathiazinyl), oxadiazinyl (including 2/-/-1 ,2,4-oxadiazinyl or 2/-/-1 ,2,5-oxadiazinyl), and morpholinyl.
The term "heteroaryl" also includes fused ring systems having one or two rings wherein such rings may be fused, wherein fused is as defined above. It is to be understood that if a carbocyclic or heterocyclic moiety may be bonded or otherwise attached to a designated substrate through differing ring atoms without denoting a specific point of attachment, then all possible points are intended, whether through a carbon atom or, for example, a trivalent nitrogen atom. For example, the term "pyridyl" means 2-, 3- or 4-pyridyl, the term "thienyl" means 2- or 3-thienyl, and so forth.
In some instances, the number of atoms in a cyclic substituent containing one or more heteroatoms (i.e., heteroaryl or heterocycloalkyl) is indicated by the prefix "x- to y- membered", wherein x is the minimum and y is the maximum number of atoms forming the cyclic moiety of the substituent. Thus, for example, "5- to 6-membered heteroaryl" refers to a heteroaryl containing from 5 to 6 atoms, including one or more heteroatoms, in the cyclic moiety of the heteroaryl. The heteroatoms for this invention are selected from nitrogen, oxygen and sulfur. Specific embodiments of ring systems include, for example: 8-oxa-2-
azaspiro[4.5]dec-2-yl,
Figure imgf000010_0001
It would be apparent to one skilled in the art, based upon the examples described herein, that other ring systems are contemplated as part of this invention.
Further embodiments of ring systems include the following, that also incorporate exemplary ring substituents: 8-oxa-2-azaspiro[4.5]dec-2-yl, 7-oxa-2-azaspiro[3.5]non-2-yl, (3aR,4R,7aS)-rel- 4-hydroxyoctahydro-2H-isoindol-2-yl, (3aR,4R,7aS)-rel-4-hydroxyoctahydro-2H- isoindol-2-yl, (8-anti)-8-methoxy-3-azabicyclo[3.2.1 ]oct-3-yl, (1 R,5S,6R)-rel-6- (hydroxymethyl)-3-azabicyclo[3.1 .0]hex-3-yl, 1 ,3-dihydro-2H-isoindol-2-yl, 3- azabicyclo[3.2.2]non-3-yl, [(3-endo)-3-hydroxy-8-azabicyclo[3.2.1 ]oct-8-yl, 2-oxa-6- azaspiro[3.5]non-6-yl, 8-oxa-3-azabicyclo[3.2.1 ]oct-3-yl, 1 -oxa-7-azaspiro[3.5]non-7-yl, (7S,8aS)-7-fluorohexahydropyrrolo[1 ,2-a]pyrazin-2(1 H)-yl, octahydropyrazino[1 ,2- a]azepin-2(1 H)-yl, 8-azaspiro[4.5]dec-8-yl, 1 -oxa-9-azaspiro[5.5]undec-9-yl, 6-oxa-9- azaspiro[4.5]dec-9-yl, 7,9-dimethyl-8-oxa-2-azaspiro[4.5]dec-2-yl, 1 -oxa-8- azaspiro[4.5]dec-8-yl, 3-methyl-1 -oxa-3,8-diazaspiro[4.5]decan-2-on-yl, 2-oxa-7- azaspiro[3.5]non-7-yl, (3aR,7aR)-rel-3a-(hydroxymethyl)octahydro-2H-isoindol-2-yl, 6- (trifluoromethyl)-3-azabicyclo[3.1.0]hex-3-yl, hexahydrocyclopenta[c]pyrrol-2(1 H)-yl, 3- methyl-1 ,7-dioxa-3, 10-diazaspiro[4.6]undecan-2-on-yl, (6S,7S)-rel-7-hydroxy-2- azaspiro[5.5]undec-2-yl, 4-methoxy-1 -oxa-9-azaspiro[5.5]undec-9-yl, 3-oxa-8- azabicyclo[3.2.1 ]oct-8-yl, (6S,7R)-rel-7-hydroxy-2-azaspiro[5.5]undec-2-yl, 6-hydroxy-2- azaspiro[3.3]hept-2-yl, 2-oxa-5-azabicyclo[2.2.2]oct-5-yl, (7R,8aS)-7- hydroxyhexahydropyrrolo[1 ,2-a]pyrazin-2(1 H)-yl, (1 S,4S)-5-methyl-2,5- diazabicyclo[2.2.1 ]hept-2-yl, (8aS)-7,7-difluorohexahydropyrrolo[1 ,2-a]pyrazin-2(1 H)-yl, 7-ethyl-2,7-diazaspiro[4.4]non-2-yl, 2-methyl-2,6-diazaspiro[3.4]oct-6-yl, (3aR,6aS)-5- methylhexahydropyrrolo[3,4-c]pyrrol-2(1 H)-yl, (8aS)-hexahydropyrrolo[1 ,2-a]pyrazin- 2(1 H)-yl, and (8aR)-hexahydropyrrolo[1 ,2-a]pyrazin-2(1 H)-yl.
As used herein, unless otherwise noted, the terms "haloalkyl" and "haloalkoxy" are intended to include both branched and straight-chain saturated aliphatic "alkyl" or "alkoxy" groups respectively, wherein "alkyl" and "alkoxy" are as defined herein, having the specified number of carbon atoms and in which at least one hydrogen is replaced with a halogen atom. As used herein, the term "halogen atom" refers to F, CI, Br and I. Haloalkyl groups include perhaloalkyl groups, wherein all hydrogens of an alkyl group have been replaced with halogens (e.g., -CF3, -CF2CF3). In certain embodiments in which two or more hydrogen atoms are replaced by halogen atoms, the halogen atoms can be the same (e.g., CHF2, -CF3) or different (e.g., CF2CI). Where so indicated, haloalkyl or haloalkoxy groups can optionally be substituted with one or more substituents in addition to halogen. Examples of haloalkyl groups include, but are not limited to, fluoromethyl, dichloroethyl, trifluoromethyl, trichloromethyl, pentafluoroethyl, and pentachloroethyl groups. As used herein, unless otherwise stated, the term "amido" refers to -C(=0)NH2.
As used herein, unless otherwise stated, the term "halogen" or "halogen atom" refers to the group consisting of fluorine (which may be depicted as -F), chlorine (which may be depicted as -CI), bromine (which may be depicted as -Br), or iodine (which may be depicted as -I).
As used herein, unless otherwise stated, the terms "hydroxy" and "hydroxyl" are used interchangeably and as used herein mean an -OH group. As used herein, unless otherwise noted, the terms "hydroxyalkyl" and "hydroxyalkoxy" are intended to include both branched and straight-chain saturated aliphatic "alkyl" or "alkoxy" groups respectively, wherein "alkyl" and "alkoxy" are as defined herein, having the specified number of carbon atoms and in which at least one hydrogen is replaced with a -OH group. Where so indicated, hydroxyalkyl and hydroxyalkoxy groups can optionally be substituted with one or more substituents in addition to -OH. Examples of hydroxyalkyl groups include, but are not limited to, CH2OH, CH2CH2OH or CH2(OH)CH2OH. As used herein, unless otherwise stated, the term "oxo" or "carbonyl" refers to
=0.
As used herein, unless otherwise stated, the term "carboxy" refers to -CO2H.
As used herein, unless otherwise stated, the term sulfonyl refers to -SO2-.
As used herein, the term "substituted" is used throughout the specification. The term "substituted" is defined herein as a moiety, whether acyclic or cyclic, which has one or more (e.g. 1 -10) hydrogen atoms replaced by a substituent as defined herein below. Substituents include those that are capable of replacing one or two hydrogen atoms of a single moiety at a time, and also those that can replace two hydrogen atoms on two adjacent carbons to form said substituent. For example, substituents that replace single hydrogen atoms include, but are not limited to, halogen, hydroxy, and the like. A two hydrogen atom replacement includes, but is not limited to, carbonyl, oximino, and the like. Substituents that replace two hydrogen atoms from adjacent carbon atoms include, but are not limited to, epoxy, and the like. When a moiety is described as "substituted" any number of its hydrogen atoms can be replaced, as described above. For example, difluoromethyl is a substituted Ci alkyl; trifluoromethyl is a substituted Ci alkyl; 4-hydroxyphenyl is a substituted aryl ring; (N,N-dimethyl-5- amino)octanyl is a substituted Cs alkyl; 3-guanidinopropyl is a substituted C3 alkyl; and 2-carboxy-3-fluoropyridinyl is a substituted heteroaryl. A multi-moiety substituent is bound through the atom indicated by To illustrate this the term "-OC-i-Cshydroxyalkyl" is an OCi-C3alkyl group substituted by a hydroxy group. Further, any carbon number pre-fix attached to a multi-moiety substituent only applies to the moiety it immediately precedes. To illustrate, the term "cycloalkyl(Ci-C4)alkyl" contains two moieties: alkyl and cycloalkyl. The (C1-C4) pre-fix on the cycloalkyl(Ci-C4)alkyl means that the alkyl moiety of the alkylcycloalkyl contains from 1 to 4 carbon atoms, the (C1-C4) pre-fix does not describe the cycloalkyl moiety.
If a group of substituents are collectively described as being optionally substituted by one or more of a list of substituents, the group may include (1 ) unsubstitutable substituents, (2) substitutable substituents that are not substituted by the optional substituents, and/or (3) substitutable substituents that are substituted by one or more of the optional substituents.
If a substituent is described such that it "may be substituted" or as being "optionally substituted" with up to a particular number of non-hydrogen substituents, that substituent may be either (1 ) not substituted; or (2) substituted by up to that particular number of non-hydrogen substituents or by up to the maximum number of substitutable positions on the substituents, whichever is less. Thus, for example, if a substituent is described as a heteroaryl optionally substituted with one, two or three substituents, then any heteroaryl with less than three substitutable positions would be optionally substituted by up to only as many non-hydrogen substituents as the heteroaryl has substitutable positions. To illustrate, tetrazolyl (which has only one substitutable position) would be optionally substituted with up to one non-hydrogen substituent.
At various places in the present specification, substituents of compounds are disclosed in groups or in ranges. It is specifically intended that the description include each and every individual sub-combination of the members of such groups and ranges. For example, the term "C-i-6 alkyl" is specifically intended to individually disclose C-i , C2, C3, C4, C5, C6, C1-C6, C1-C5, C1-C4, C1-C3, C1-C2, C2-C6, C2-C5, C2-C4, C2-C3, C3-C6, C3- C5, C3-C4, C4-C6, C4-C5, and C5-C6 alkyl. For example, the term "C1-3 alkyl" is specifically intended to individually disclose C-i , C2, C3, C1-C3, C1-C2, and C2-C3 alkyl. Compounds of the present invention may contain basic nitrogen atoms (e.g. alkyl amines or heterocycles such as pyridine etc.) which may be converted to N-oxides by treatment with an oxidizing agent (e.g. mCPBA and/or hydrogen peroxides) to afford other compounds of this invention. Thus, all nitrogen-containing compounds that may be converted to N-oxide (N - ) or -N+-0") derivatives are part of the invention. If substituents are described as "independently" having more than one variable, each instance of a substituent is selected independent of the other from the list of variables available. Each substituent therefore may be identical to or different from the other substituent(s). As used herein, the terms "Formula I", "Formula II", "Formula lla-llg: and/or
"Formula la-lg", may be hereinafter referred to as a "compound(s) of the invention," "the present invention," and collectively the "compound of Formula I." Accordingly, the term "compound of Formula I" or "compound of formula (I), and the like, includes the compounds of Formula I, la, lb, Ic, Id, le, If and Ig, as well as the compounds of Formula II, Ma, Mb, lie, lid, Me, llf and llg, whether capitalized, bolded or not. Such terms are also defined to include all forms of the compound of Formula I, including hydrates, solvates, isomers, crystalline and non-crystalline forms, isomorphs, polymorphs, tautomers and metabolites thereof. For example, the compounds of the invention, or pharmaceutically acceptable salts thereof, may exist in unsolvated and solvated forms. When the solvent or water is tightly bound, the complex will have a well-defined stoichiometry independent of humidity. When, however, the solvent or water is weakly bound, as in channel solvates and hygroscopic compounds, the water/solvent content will be dependent on humidity and drying conditions. In such cases, non-stoichiometry will be the norm. The compounds of the invention have asymmetric carbon atoms. The carbon- carbon bonds of the compounds of the invention may be depicted herein using a solid line ( ), a solid wedge ( ), or a dotted wedge ( 11 ). The use of a solid line to depict bonds to asymmetric carbon atoms is meant to indicate that all possible stereoisomers (e.g., specific enantiomers, racemic mixtures, etc.) at that carbon atom are included. The use of either a solid or dotted wedge to depict bonds to asymmetric carbon atoms is meant to indicate that only the stereoisomer shown is meant to be included or that the stereoisomer predominates the other stereoisomer. It is possible that compounds of Formula I may contain more than one asymmetric carbon atom. In those compounds, the use of a solid line to depict bonds to asymmetric carbon atoms is meant to indicate that all possible stereoisomers are meant to be included. For example, unless stated otherwise, it is intended that the compounds of Formula I can exist as enantiomers and diastereomers or as racemates and mixtures thereof. The use of a solid line to depict bonds to one or more asymmetric carbon atoms in a compound of Formula I and the use of a solid or dotted wedge to depict bonds to other asymmetric carbon atoms in the same compound is meant to indicate that a mixture of diastereomers is present.
Stereoisomers of Formula I include cis and trans isomers, optical isomers such as R and S enantiomers, diastereomers, geometric isomers, rotational isomers, conformational isomers, and tautomers of the compounds of the invention, including compounds exhibiting more than one type of isomerism; and mixtures thereof (such as racemates and diastereomeric pairs). Also included are acid addition or base addition salts wherein the counterion is optically active, for example, D-lactate or L-lysine, or racemic, for example, DL-tartrate or DL-arginine.
For example, Figure 2 depicts an X-ray crystal structure (ORTEP drawing) of Example 145a, 8-oxa-2-azaspiro[4.5]dec-2-yl(2-{[(1 S)-1 -(pyrazin-2- yl)ethyl]amino}pyrimidin-5-yl)methanone methanesulfonate. The single crystal X-Ray structure of example 145a is consistent with Example 145 having a "S" absolute configuration. By deduction, Example 146 was assigned the "R" enantiomer of this pair and dis layed a ~100-fold loss of potency against vanin in the assay.
Figure imgf000015_0001
Example 145 Example 146
The above determinations were then used to extrapolate absolute configurations of other enantiomeric pairs of the compounds of the invention, possessing an
asymmetric carbon at the same position as Example 145, as detailed in the Examples herein. In particular, the most potent enantiomer was assigned the "S" absolute configuration, based upon the configuration of Example 145, while the least potent enantiomer was assigned the "R" configuration. Accordingly, some of the Examples have a designation of "absolute stereochemistry inferred", based upon the above assumptions.
Optical isomers can be obtained in pure form by standard procedures known to those skilled in the art, which include, but are not limited to for example, chiral chromatography, diastereomeric salt formation, kinetic resolution, and asymmetric synthesis. It is also understood that the present teachings encompass all possible regioisomers, and mixtures thereof, which can be obtained in pure form by standard separation procedures known to those skilled in the art, and include, but are not limited to, column chromatography, thin-layer chromatography, and high-performance liquid chromatography.
When any racemate crystallizes, crystals of two different types are possible. The first type is the racemic compound (true racemate) referred to above wherein one homogeneous form of crystal is produced containing both enantiomers in equimolar amounts. The second type is the racemic mixture or conglomerate wherein two forms of crystal are produced in equimolar amounts each comprising a single enantiomer.
The compounds of the invention not only include compounds as hereinbefore defined, but also all forms of the compounds of the invention, including isomers (including optical, geometric and tautomeric isomers), hydrates, solvates, complexes, salts (including solvates and complexes thereof) crystalline and non-crystalline forms, isomorphs, polymorphs, isotopically-labeled derivatives, metabolites and prodrugs (including tautomeric forms of such prodrugs) thereof.
A "metabolite" of a compound disclosed herein is a derivative of that compound that is formed when the compound is metabolized. The term "active metabolite" refers to a biologically active derivative of a compound that is formed when the compound is metabolized. The term "metabolized," as used herein, refers to the sum of the processes (including, but not limited to, hydrolysis reactions and reactions catalyzed by enzymes, such as, oxidation reactions) by which a particular substance is changed by an organism. Thus, enzymes may produce specific structural alterations to a compound. For example, cytochrome P450 catalyzes a variety of oxidative and reductive reactions while uridine diphosphate glucuronyl transferases catalyze the transfer of an activated glucuronic-acid molecule to aromatic alcohols, aliphatic alcohols, carboxylic acids, amines and free sulfhydryl groups. Further information on metabolism may be obtained from The Pharmacological Basis of Therapeutics, 9th Edition, McGraw-Hill (1996), incorporated herein by reference. Metabolites of the compounds disclosed herein can be identified either by administration of compounds to a host and analysis of tissue samples from the host, or by incubation of compounds with hepatic cells in vitro and analysis of the resulting compounds. Both methods are well known in the art. In some embodiments, metabolites of a compound are formed by oxidative processes and correspond to the corresponding hydroxy-containing compound. In some embodiments, a compound is metabolized to pharmacologically active metabolites. The compounds of the invention may exist in both unsolvated and solvated forms. The term "solvate" as used herein means a physical association of a compound with one or more solvent molecules, whether organic or inorganic, including water ('hydrate'). As noted above, the compounds of the invention, or pharmaceutically acceptable salts thereof, may exist in unsolvated and solvated forms. When the solvent or water is tightly bound, the complex will have a well-defined stoichiometry independent of humidity. When, however, the solvent or water is weakly bound, as in channel solvates and hygroscopic compounds, the water/solvent content will be dependent on humidity and drying conditions. In such cases, non-stoichiometry will be the norm.
The compounds of this invention may be used in the form of salts derived from inorganic or organic acids. Depending on the particular compound, a salt of the compound may be advantageous due to one or more of the salt's physical properties, such as enhanced pharmaceutical stability in differing temperatures and humidities, or a desirable solubility in water or oil. In some instances, a salt of a compound also may be used as an aid in the isolation, purification, and/or resolution of the compound.
Where a salt is intended to be administered to a patient (as opposed to, for example, being used in an in vitro context), the salt preferably is pharmaceutically acceptable. The term "pharmaceutically acceptable salt" refers to a salt prepared by combining a compound of the invention (e.g. a compound of Formula (I)) with an acid whose anion, or a base whose cation, is generally considered suitable for human consumption. Pharmaceutically acceptable salts are particularly useful as products of the methods of the present invention because of their greater aqueous solubility relative to the parent compound. For use in medicine, the salts of the compounds of this invention are non-toxic "pharmaceutically acceptable salts." Salts encompassed within the term "pharmaceutically acceptable salts" refer to non-toxic salts of the compounds of this invention which are generally prepared by reacting the free base with a suitable organic or inorganic acid.
Suitable pharmaceutically acceptable acid addition salts of the compounds of the present invention when possible include those derived from inorganic acids, such as hydrochloric, hydrobromic, hydrofluoric, boric, fluoroboric, phosphoric, metaphosphoric, nitric, carbonic, sulfonic, and sulfuric acids, and organic acids such as acetic, benzenesulfonic, benzoic, citric, ethanesulfonic, fumaric, gluconic, glycolic, isothionic, lactic, lactobionic, maleic, malic, methanesulfonic, trifluoromethanesulfonic, succinic, toluenesulfonic, tartaric, and trifluoroacetic acids. Suitable organic acids generally include but are not limited to aliphatic, cycloaliphatic, aromatic, araliphatic, heterocyclic, carboxylic, and sulfonic classes of organic acids.
Specific examples of suitable organic acids include but are not limited to acetate, trifluoroacetate, formate, propionate, succinate, glycolate, gluconate, digluconate, lactate, malate, tartaric acid, citrate, ascorbate, glucuronate, maleate, fumarate, pyruvate, aspartate, glutamate, benzoate, anthranilic acid, stearate, salicylate, p- hydroxybenzoate, phenylacetate, mandelate, embonate (pamoate), methanesulfonate, ethanesulfonate, benzenesulfonate, pantothenate, toluenesulfonate, 2- hydroxyethanesulfonate, sufanilate, cyclohexylaminosulfonate, algenic acid, .beta.- hydroxybutyric acid, galactarate, galacturonate, adipate, alginate, butyrate, camphorate, camphorsulfonate, cyclopentanepropionate, dodecylsulfate, glycoheptanoate, glycerophosphate, heptanoate, hexanoate, nicotinate, 2-naphthalesulfonate, oxalate, palmoate, pectinate, 3-phenylpropionate, picrate, pivalate, thiocyanate, and undecanoate.
Furthermore, where the compounds of the invention carry an acidic moiety, suitable pharmaceutically acceptable salts thereof may include alkali metal salts, i.e., sodium or potassium salts; alkaline earth metal salts, e.g., calcium or magnesium salts; and salts formed with suitable organic ligands, e.g., quaternary ammonium salts. In another embodiment, base salts are formed from bases which form non-toxic salts, including aluminum, arginine, benzathine, choline, diethylamine, diolamine, glycine, lysine, meglumine, olamine, tromethamine and zinc salts.
Organic salts may be made from secondary, tertiary or quaternary amine salts, such as tromethamine, diethylamine, Ν,Ν'-benzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, meglumine (N-methylglucamine), and procaine. Basic nitrogen-containing groups may be quaternized with agents such as lower alkyl (C.sub.1 -C.sub.6) halides (e.g., methyl, ethyl, propyl, and butyl chlorides, bromides, and iodides), dialkyl sulfates (i.e., dimethyl, diethyl, dibutyl, and diamyl sulfates), long chain halides (i.e., decyl, lauryl, myristyl, and stearyl chlorides, bromides, and iodides), arylalkyl halides (i.e., benzyl and phenethyl bromides), and others.
In one embodiment, hemisalts of acids and bases may also be formed, for example, hemisulphate and hemicalcium salts. Included within the scope of the invention are complexes such as clathrates, drug-host inclusion complexes wherein, in contrast to the aforementioned solvates, the drug and host are present in stoichiometric or non-stoichiometric amounts. Also included are complexes of the drug containing two or more organic and/or inorganic components which may be in stoichiometric or non-stoichiometric amounts. The resulting complexes may be ionised, partially ionised, or non-ionised. For a review of such complexes, see J Pharm Sci, 64 (8), 1269-1288 by Haleblian (August 1975).
The present invention includes all pharmaceutically acceptable isotopically- labelled compounds of the invention wherein one or more atoms are replaced by atoms having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Examples of isotopes suitable for inclusion in the compounds of the invention include isotopes of hydrogen, such as 2H and 3H, carbon, such as 11C, 13C and 1 C, chlorine, such as 36CI, fluorine, such as 18F, iodine, such as 123l and 125l, nitrogen, such as 13N and 15N, oxygen, such as 150, 170 and 180, phosphorus, such as 32P, and sulphur, such as 35S. Certain isotopically- labelled compounds of formula (I), for example, those incorporating a radioactive isotope, are useful in drug and/or substrate tissue distribution studies. The radioactive isotopes tritium, i.e. 3H, and carbon-14, i.e. 1 C, and 125l are particularly useful for this purpose in view of their ease of incorporation and ready means of detection. Substitution with heavier isotopes such as deuterium, i.e. 2H, may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements, and hence may be preferred in some circumstances. Substitution with positron emitting isotopes, such as 11C, 18F, 150 and 13N, can be useful in Positron Emission Topography (PET) studies for examining substrate receptor occupancy. Isotopically-labeled compounds of formula (I) can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described in the accompanying Examples and Preparations using an appropriate isotopically-labeled reagents in place of the non- labeled reagent previously employed.
In some embodiments, compounds described herein could be prepared as prodrugs. A "prodrug" refers to an agent that is converted (e.g., either spontaneous or enzymatic) within the target physiological system into the parent drug in vivo. Prodrugs are designed to overcome problems associated with stability, toxicity, lack of specificity, or limited bioavailability. In some situations, they may be easier to administer than the parent drug. They may, for instance, be bioavailable by oral administration whereas the parent is not. The prodrug may also have improved solubility in pharmaceutical compositions over the parent drug. An example, without limitation, of a prodrug would be a compound described herein, which is administered as an ester (the "prodrug") to facilitate transmittal across a cell membrane where water solubility is detrimental to mobility but which then is metabolically hydrolyzed to the carboxylic acid, the active entity, once inside the cell where water-solubility is beneficial. A further example of a prodrug might be a short peptide (polyaminoacid) bonded to an acid group where the peptide is metabolized to reveal the active moiety. In certain embodiments, upon in vivo administration, a prodrug is chemically converted to the biologically, pharmaceutically or therapeutically active form of the compound. In certain embodiments, a prodrug is enzymatically metabolized by one or more steps or processes to the biologically, pharmaceutically or therapeutically active form of the compound. To produce a prodrug, a pharmaceutically active compound is modified such that the active compound will be regenerated upon in vivo administration. The prodrug can be designed to alter the metabolic stability or the transport characteristics of a drug, to mask side effects or toxicity, to improve the flavor of a drug or to alter other characteristics or properties of a drug. By virtue of knowledge of pharmacodynamic processes and drug metabolism in vivo, those of skill in this art, once a pharmaceutically active compound is known, can design prodrugs of the compound. (see, for example, Nogrady (1985) Medicinal Chemistry A Biochemical Approach, Oxford University Press, New York, pages 388-392; Silverman (1992), The Organic Chemistry of Drug Design and Drug Action, Academic Press, Inc., San Diego, pages 352-401 , Saulnier et al., (1994), Bioorganic and Medicinal Chemistry Letters, Vol. 4, p. 1985). Prodrugs may be designed as reversible drug derivatives, for use as modifiers to enhance drug transport to site-specific tissues. See, e.g., Fedorak et al., Am. J. Physiol., 269:G210-218 (1995); McLoed et al., Gastroenterol, 106:405-413 (1994); Hochhaus et al., Biomed. Chrom., 6:283-286 (1992); J. Larsen and H. Bundgaard, Int. J. Pharmaceutics, 37, 87 (1987); J. Larsen et al., Int. J. Pharmaceutics, 47, 103 (1988); Sinkula et al., J. Pharm. Sci., 64: 181 -210 (1975); T. Higuchi and V. Stella, Pro-drugs as Novel Delivery Systems, Vol. 14 of the A.C.S. Symposium Series; and Edward B. Roche, Bioreversible Carriers in Drug Design, American Pharmaceutical Association and Pergamon Press, 1987, all incorporated herein in their entirety.
Some preferred prodrugs are variations or derivatives of compounds that have groups cleavable under metabolic conditions. Common prodrugs include acid derivatives such as esters, such as carboxylic esters (eg ethyl esters) and phosphate esters prepared by reaction of parent acids with a suitable alcohol (e.g., a lower alkanol), or of parent alcohols with a suitable acid (e.g. phosphate esters of hydroxyl groups); amides prepared by reaction of the parent acid compound with an amine, or basic groups reacted to form an acylated base derivative (e.g., a lower alkylamide).
Compounds of the Invention
The invention is directed to the compound of Formula I, including the racemates, and/or diastereomer mixtures, as well as specific enantiomers and/or diastereomers thereof,
Figure imgf000021_0001
wherein
G is a 6-membered heteroaryl, with one, two or three N, wherein the
heteroaryl is optionally substituted with one, two or three substituents selected from halogen, OH, cyano, Ci-C4alkyl, -NR8aC(O)R8b, -NR8aSO2R8b - (CR6aR6b)tC(O)N(R8a)2, -C(O)OH, -N(R8a)2, -(CR6aR6b)tSO2R8b, -
(CR6aR6b)tSO2N(R8a)2, Ci-C alkoxy, -S(Ci-C3alkyl) or C3-C5cycloalkyl, wherein the alkyl, cycloalkyl and alkoxy are optionally substituted with one, two or three halogen, OH, OCH3, or C3-C5cycloalkyl;
L is NH or O;
Z is a bond; -(CR5aR5b)q-; -CH2(CR5aR5b)m-; or -(CR5aR5b)m-W-(CR5aR5b)n-, wherein W is S, O or NR7;
Ra, Rb, R1 a and R1 b are each independently hydrogen, Ci-C4alkyl, wherein the alkyl is optionally substituted with one, two or three halogen, OH, cyano, -S(Ci- Csalkyl) or Ci-C4alkoxy, optionally substituted with one, two or three fluoro;
or R1 a and R1 b, together with the carbon to which they are bonded, form an oxo, C3-C5cycloalkyl, -(4- to 5-membered heterocycloalkyl) wherein said cycloalkyl or heterocycloalkyl are optionally substituted with one, two, three or four halogen, OH, Ci-C4alkyl, -S(Ci-C3alkyl) Ci-C4alkoxy or cyano; and the heteroatom is
selected from one or two N, S or O; R2a and R2b are each independently hydrogen, OH, halogen, - (CR6aR6b)tSO2R8b -(CR6aR6b)tC(O)N(R8a)2, -NR8aC(O)R8b, -NR8aC(O)N(R8a)2, - SO2N(R8a)2, Ci-C4alkyl, Ci-C alkoxy, S(Ci-C3alkyl), cyano, -(CR6aR6b)t-(C3- Cecycloalkyl), -(CR6aR6b)n-(5- to 6- membered heterocycloalkyi) or -(CR6aR6b)n-(5- to 6-membered heteroaryl), wherein said heteroatoms of said heteroalkyl and heteroaryl are selected from one, two or three N, O or S; wherein said alkyl, cycloalkyi, heterocycloakyl and heteroaryl are optionally substituted with one, two, three or four R9; or
R2a and R2b together with the carbon to which they are bonded form a C3- Cgcycloalkyl or a -(4- to 1 1 -membered heterocycloalkyi), having one to three heteroatoms selected from N, O or S; wherein the cycloalkyi and heterocycloalkyi are optionally substituted with one, two or three Ci-C4alkyl, S(Ci-C3alkyl), OH, halogen, oxo, -C(O)NH2, -C(O)NHCH3, -C(O)N(CH3)2, C3-C5cycloalkyl or Ci- C4alkoxy; or if substitution is at a N atom, then such N atom is substituted with R7; or R2a or R2b, and one of R5a or R5b, together with the respective carbons to which they are bonded, form a C3-Ci2cycloalkyl, C6-Cioaryl, -(5- to 6-membered heteroaryl) or a -(4- to 12-membered heterocycloalkyi), wherein said heteroaryl or heterocycloalkyi is optionally substituted with one, two, three or four R9; or if substitution is at a N atom, then such N atom is substituted with R7; or
R2a or R2b, and R7, together with the respective atoms to which they are bonded form a -(4- to 12- membered heterocycloalkyi) or a -(5- to 6-membered heteroaryl), wherein said heterocycloalkyi or heteroaryl have one, two to three heteroatoms selected from N, O or S, wherein said heterocycloalkyi and heteroaryl are optionally substituted with one, two, three or four R9; or if substitution is at a N atom, then such N atom is substituted with R7;
R3 is hydrogen, -(CR6aR6b)tC(O)NH2,or Ci-C alkyl, wherein said alkyl is optionally substituted with one, two, three or four R9; or
R3 and Rb, together with the carbon to which they are attached, form an oxo; R4 is hydrogen, -(CR6aR6b)tC(O)NH2, or Ci-C alkyl, wherein said alkyl is optionally substituted with one, two, three or four R9; or
R3 and R4 taken together with the respective carbons to which they are bonded form a -(4- to 1 1 -membered heterocycloalkyi), having one to two
heteroatoms selected from N, O or S, wherein the heterocycloalkyi are optionally substituted with one, two, three or four R9; or if substitution is at a N atom, then such N atom is substituted with R7; or R4 and Ra, together with the carbon to which they are attached, form an oxo;
R5a and R5b are each independently hydrogen, halogen, OH, - (CR6aR6b)tC(O)R8b, -(CR6aR6b)tC(O)NH2, d-C4alkyl, S(Ci-C3alkyl), Ci-C alkoxy, cyano, -(CR6aR6b)t-(C3-C6cycloalkyl) or -(CR6aR6b)t-(C3-C6heterocycloalkyl), wherein said alkyl, cycloalkyi and heterocycloalkyl are optionally substituted with one, two, three or four R9; and the heteroatom is selected from one or two N, O, or S; or
R5a and R5b taken together with the carbon to which they are bonded form a Cs-Cgcycloalkyl or a 4- to 1 1 -membered heterocycloalkyl, wherein the heteroatom is selected from one or two N, S or O, wherein said cycloalkyi or heterocycloalkyl is optionally substituted with one, two, three or four R9; or if substitution is at a N atom, then such N atom is substituted with R7; or
R3 and either R5a or R5b taken together with the respective carbons to which they are bonded form a C3-Ciocycloalkyl or -(4- to 12-membered heterocycloalkyl), wherein the heteroatom is selected from one or two N or O, wherein said cycloalkyi and heterocycloalkyl are optionally substituted with one, two, three or four R9 or oxo; or if substitution is at a N atom, then such N atom is substituted with R7;
R6a and R6b are each independently hydrogen, Ci-C alkyl, S(Ci-C3alkyl), OH, Ci-C4alkoxy, cyano or halogen;
R7 is hydrogen; -(4- to 6-membered heterocycloalkyl), having 1 to 2 heteroatoms wherein said heteroatom is selected from O, N and S; d-Csalkyl; S(Ci- C3alkyl); C(O)R8b; SO2R8b; SO2N(R8a)2; C(O)N(R8a)2 or -(C3-C7cycloalkyl), wherein said alkyl, heterocycloalkyl and cycloalkyi are optionally substituted with Ra;
R8a is hydrogen, Ci-C4alkyl or -(C3-C7cycloalkyl);
R8b is Ci-C alkyl, -(C3-C7cycloalkyl), -(CR6aR6b)tSO2N(R8a)2, -
(CR6aR6b)tSO2R8a or -(CR6aR6b)tNHC(O)N(R8a)2;
R9 is hydrogen, Ci-C alkyl, S(Ci-C3alkyl), OH, CH2OH, halogen, Ci-C alkoxy, cyano or -C(O)NH2, wherein said alkyl and alkoxy are optionally substituted with OH, halogen, -C(O)NH2, -C(O)NHCH3, -C(O)N(CH3)2, -S(Ci-C alkyl) or C3- Cscycloalkyl; or R9 is oxo, provided that it is attached to a non-aromatic group;
R10 is hydrogen or Ci-C3alkyl;
m, n and t are each independently 0, 1 or 2; q is 1 , 2 or 3; and x is 1 or 2; or pharmaceutically acceptable salts thereof.
In another embodiment, the invention is directed to compounds having the Formula la, lb, lc, Id, le, If and Ig.
Figure imgf000024_0001
Figure imgf000024_0002
Figure imgf000024_0003
or ig
G is a 6-membered heteroaryl, with one, two or three N, wherein the heteroaryl is optionally substituted with one, two or three substituents selected from halogen, OH, cyano, Ci-C4alkyl, -NR8aC(O)R8b, -NR8aSO2R8b -
(CR6aR6b)tC(O)N(R8a)2, -C(O)OH, -N(R8a)2, -(CR6aR6b)tSO2R8b, -
(CR6aR6b)tSO2N(R8a)2, Ci-C alkoxy, -S(Ci-C3alkyl) or C3-C5cycloalkyl, wherein the alkyl, cycloalkyi and alkoxy are optionally substituted with one, two or three halogen,
OH, OCH3, or Cs-Cscycloalkyl;
L is NH or O;
Z is a bond; -(CR5aR5b)q-; -CH2(CR5aR5b)m-; or -(CR5aR5b)m-W-(CR5aR5b)n-, wherein W is S, O or NR7; and
Ra, Rb, R1 a, R1 b, R2a, R2b, R3, R4, R5a, R5b, R6 R7, R8, R9 and R10 are as previously described herein; or a pharmaceutically acceptable salt thereof.
In another embodiment, G is a triazinyl, pyridazinyl, pyridonyl, pyridinyl, pyrazinyl or pyrimidinyl, optionally substituted with one, two or three substituents selected from halogen, OH, cyano, Ci-C alkyl, -NR8aC(O)R8b, -(CR6aR6b)tC(O)NH2, -C(0)OH, -N(R )2 , Ci-C4alkoxy, wherein the alkyl and alkoxy are optionally substituted with one, two or three halogen, OH, OCH3, Cs-Cscycloalkyl or -S(Ci- Csalkyl);. or a pharmaceutically acceptable salt thereof.
In yet another embodiment, G is a pyrazinyl or a pyrimidinyl.
In another embodiment, L is NH.
Further embodiments of the invention include, but are not limited to compounds of Formula I, Formula la-lg, and Formula lla-llg with new ring formations between, for example, the following substituents: R1 a and R1 b, R2a and R2b, R3 and R4, R2a or R2b and either R5a or R5b, R3 and either R5a or R5b, and R2a or R2b and R7 In such embodiments, one skilled in the art would appreciate that upon such selected substituents, taken together with the atoms to which they are bonded, such substituents may take the form of a bond, if appropriate, or an "alkylene" or a "heteroalkylene" and would, respectively, therefore, ultimately form a cycloalkyl or a heterocycloalkyl. The term "alkylene", as used herein, refers to a saturated, branched or straight chain or cyclic hydrocarbon diradical of the stated number of carbon atoms, typically 1 -6 carbon atoms, and having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent alkane. Typical alkylene radicals include, but are not limited to methylene (-CH2-), 1 ,2-ethylene (-CH2CH2-), 2,2-dimethylene, 1 ,3-propylene (-CH2CH2CH2-), 2-methylpropylene, 1 ,4- butylene (-CH2CH2CH2CH2-), and the like; optionally substituted as defined herein. Likewise, the term "heteroalkylene" means a divalent group derived from heteroalkyl (as defined above). For heteroalkylene groups, heteroatoms can also occupy either or both of the chain termini. For example, in one embodiment, a new ring formation between R2a and R2b can create a c cloalkyl, optionally substituted, as exemplified in the following structure,
Figure imgf000025_0001
, wherein the smallest cycloalkyl formed is cyclopropyl, and in that case, one of R or R comprises an ethylene (-CH2CH2-) and one a bond. Alternatively, both R2a or R2b comprise a methylene (-CH2-). In a more specific embodiment, one skilled in the art would appreciate from the description herein and the examples that R2a and R2b could form the following new ring formation, optionally substituted as appropriate, (R2a and R2b labels left in for further clarification),
Figure imgf000026_0001
, wherein one of R2a or R2b independently is a bond, methylene (-CH2-), 1 ,2-ethylene (-CH2CH2-), 2,2-dimethylene, 1 ,3-propylene (-CH2CH2CH2-), 2- methylpropylene, 1 ,4-butylene (-CH2CH2CH2CH2-), and the like; and x is 0-5.
Another embodiment of a new ring formation between R2a and R2b is wherein the two substituents form a -(4- to 1 1 -membered heterocycloalkyl), optionally substituted as a ropriate, such as described in the following exemplary formula. In this case,
Figure imgf000026_0002
and the smallest heterocycloalkyl is a 4-membered ring, optionall substituted as appropriate. In particular, a specific embodiment
Figure imgf000026_0003
includes, K , a heterocycloalkyl, wherein, for example, one of R or R independently is a bond, methylene (-CH2-), 1 ,2-ethylene (-CH2CH2-), 2,2-dimethylene, 1 ,3-propylene (-CH2CH2CH2-), 2-methylpropylene, 1 ,4-butylene (-CH2CH2CH2CH2-), and the like; and the other of R2a or R2b is a heteroalkylene.
In a further embodiment, a 9-carbon cycloalkyi new ring formation and an 1 1 - membered heterocycloalkyl between R2a and R2b are de icted in the following
structures:
Figure imgf000026_0004
and (R2a and R2b labels left in for further clarification). Different variations of these cycloalkyi and heterocycloalkys are considered part of the present invention and are further described herein.
One skilled in the art would appreciate that the description of how R2a and R2b may form a new ring system as described above, may be extrapolated to the other substituents (R1 a and R1 b, R3 and R4, R2a or R2b and either R5a or R5b, R3 and either R5a or R5b, and R2a or R2b and R7) that are described as forming new rings and such description is applicable, as appropriate. For example, in another embodiment, a new ring formation can occur between
substituents R5a and R5b to provide an optionally substituted cycloalkyl,
Figure imgf000027_0001
with the smallest being a cyclopropyl; wherein both R5a or R5b comprise a methylene (- CH2-). Alternatively, one of R5a or R5b comprise an ethylene (-CH2CH2-). and the other a bond.
In another embodiment, such substituents could form a 4- to 7-membered
heterocycloalkyi, optionally substituted,
Figure imgf000027_0002
, wherein the smallest heterocycloalkyi is a 4-membered ring (wherein X is N, 0 or S) and wherein R5a or R5b are independently a bond, an alkylene and, at least one of R5a or R5b comprises a heteroalkylene (e.g. -0-CH2-).
In a more specific embodiment, one skilled in the art would appreciate from the description herein and the examples that R5a and R5b could form the followin new ring
formations (R5a and R5b labels left in for further clarification):
Figure imgf000027_0003
, wherein
R5a and R5b are independently a bond, a methylene (-CH2-), 1 ,2-ethylene (-CH2CH2-), 2,2-dimethylene, 1 ,3-propylene (-CH2CH2CH2-), 2-methylpropylene, or 1 ,4-butylene
(-CH2CH2CH2CH2-), and the like; or
Figure imgf000027_0004
, forming a cycloalkyl, wherein R and R5b are each 1 ,4-butylene (-CH2CH2CH2CH2-) in the later example. In another embodiment, R and R j35Db, being an alkylene or heteroalk lene group,
form an optionally substituted heterocycloalkyl, such
Figure imgf000028_0001
or
Figure imgf000028_0002
as described herein. Other specific embodiments of ring systems
include, for example: 8-oxa-2-azaspiro[4.5]dec-2-yl,
Figure imgf000028_0003
and 7-oxa-2-
azaspiro[3.5]nonan-2-yl,
Figure imgf000028_0004
. it would be apparent to one skilled in the art, based upon the examples described herein, that other possible ring systems are contemplated as part of this invention.
In another embodiment, a new ring formation can occur between substituents R2a or R2b and one Qf R5a or R5b tQ provjc|e a c3-C12cycloalkyl, C6-C10aryl, -(5- to 12- membered heteroar l) or a -(4- to 12-membered heterocycloalkyl)as exemplified in the
following depictions,
Figure imgf000028_0005
, wherein the smallest cycloalkyl is a cyclopropyl and one of R or R comprises a methylene (-CH2-) and one a bond, said cycloalkyl optionally substituted as appropriate. The following structures demonstrate the formation of the new cyclopropyl ring, wherein one of R2a or R5a is a bond:
Figure imgf000028_0006
ln another embodiment, or
Figure imgf000029_0001
, wherein W is defined herein as N, 0, or S, and the smallest heterocycloalkyl formed is a 4-membered heterocycloalkyl, wherein one of R2a or R5a comprises a methylene (-CH2-) and one a bond, said heterocycloalkyl optionally substituted as appropriate.
In particular, one skilled in the art would appreciate from the description herein and the examples that R2a and R5 could form the following specific exemplary new ring formations, optionally substituted as defined herein, (R2a and R5a labels remaining for
clarification): a cycloalkyi,
Figure imgf000029_0002
independently Ci-C4alkylene; Z is -(CR5aR5b)q-; and q is 1 ); or R a , (R2a and R5a are independently Ci-C alkylene; Z is -(CR5aR5b)q-; and P is 3); an ary'.
Figure imgf000029_0003
V R a heteroaryl, v R ; and a heterocycloalkyl,
Figure imgf000029_0004
, (R2a and R5a are independently -0-Ci-C4alkylene or Ci-C4alkylene; Z is -(CR5aR5b)q-; and q is 3); all of which are particular embodiments of this invention, as well as other possible ring formations. In another embodiment, a new ring formation can occur between substituents
R2a or R2b and R7 to form a -(4- to 12- membered heterocycloalkyl) or a -(5- to 6- membered heteroaryl), having two to three heteroatoms selected from N, 0 or S, and wherein W is N as exemplified by the following structures:
Figure imgf000030_0001
wherein the smallest heterocycloalky formed is a 4-membered ring, wherein R2a and R7 is, for example, -CH2- , optionally substituted as defined herein. One of ordinary skill in the art would appreciate that different combinations of substituent groups for R2a and R7 could arrive at the 4-membered ring, as well as other ring sizes.
For example, in another embodiment of an heterocycloalkyl, optionally
Figure imgf000030_0002
substituted as defined herein, R is Ci-C4alkylene (and in this example is 1 ,4-butylene (-CH2CH2CH2CH2-)); R7 is d-Csalkylene (and in this example, 1 ,5-pentylene (-CH2CH2CH2CH2-)); Z is -(CR5aR5b)m-W-( CR5aR5b)n-; and m and n are 2, to form a 12-membered heterocycloalkyl ring.
In a particular embodiment, one skilled in the art would appreciate from the description herein and the examples that R2a and R7 could form the following specific new ring formations, optionally substituted as described herein, (R2a and R7 labels
remainin for clarification): a heterocycloalkyl,
Figure imgf000030_0003
and a heteroaryl,
Figure imgf000030_0004
. Both of these embodiments, and variations thereof, are contemplated as part of this invention, as well as other possible ring formations described herein.
In another embodiment, a new ring formation can occur between substituents R3 and R4 to provide a -(4- to 12-membered heterocycloalkyl), optionally substituted as defined herein, having one to two heteroatoms selected from N, O or S, as exemplified in the following structure,
Figure imgf000031_0001
wherein the smallest heterocycloalkyl formed is a 4-membered ring, (assuming one of R3 or R4 comprises a methylene (- CH -) and the other a bond), such as depicted in the following:
Figure imgf000031_0002
One of ordinary skill in the art would understand that, in this example, as well as others exemplified herein, the N of the amide is part of the new ring being formed and is, therefore, included in numbering the new ring system.
In a specific embodiment, one skilled in the art would appreciate from the description herein and the examples that R3 and R4 could form the following specific new rin formations (R3 and R4 labels remaining for clarification): the heterocycloalkyl,
Figure imgf000031_0003
, wherein different variations of R3 or R4 substituents could arrive at the 5-membered heterocycloalkyl described above. For example, R3 and R4 independently are a bond or Ci-C2alkylene (e.g. methylene (-CH2-), 1 ,2-ethylene (-CH2CH2-)). One of ordinary skill in the art would appreciate that many other new ring size formations are possible and are part of the contemplated invention described herein. For example, in another embodiment, a larger heterocycloalkyl may be formed,
Figure imgf000031_0004
r4 , wherein R3 and R4 , in this example, could each be 1 ,4-butylene
(-CH2CH2CH2CH2-)) to form a 1 1 -membered heterocycloalkyl ring.
In another embodiment, a new ring formation may occur between R3 and R5a or R5b to provide for a C3-C7cycloalkyl or -(4- to 12-membered heterocycloalkyl), as exemplified in the following structure,
Figure imgf000032_0001
, wherein the smallest heterocycloalkyi formed is a 4-membered ring (assuming R3 or R5a is a bond).
In a particular embodiment, one skilled in the art would appreciate from the description herein and the examples that R3 and R5 could form the following specific new ring formations (R3 and R5 labels remaining for clarification): a heterocycloalkyi,
Figure imgf000032_0002
. Other specific ring formations are contemplated as part of this invention and are described herein.
In another embodiment, a new ring formation may occur between R3 and R5a or R5b to provide for a C -Crcycloalkyl, wherein W is O or NR7, as exemplified in the
following structure,
Figure imgf000032_0003
, wherein the smallest cycloalkyl formed is cyclobutyl (assuming one of R3 and R5a is a bond or d-C^alkylene (e.g. methylene (-
CH2-),), as depicted in the following:
Figure imgf000032_0004
. In a
specific embodiment,
Figure imgf000032_0005
, R3 and R5a form an optionally substituted
Cucycloalkyl, wherein R3 and R5a , in this example, are each 1 ,4-butylene (-CH2CH2CH2CH2-)); Z is -(CR5aR5b)m-W-( CR5aR5V; and m and n are 2. One skilled in the art would appreciate that different variations of R3 and R5a are described herein, each of which would form different new ring sizes. In an alternative embodiment, R3 and R5a form an optionally substituted 12-
membered heterocycloalkyi,
Figure imgf000033_0001
, wherein W is N, 0 or S; and R3 and R5a , in this example, are each 1 ,4-butylene (-CH2CH2CH2CH2-)).
In another embodiment, R1 a or R1 b are each independently hydrogen, or C-i- Csalkyl, wherein the alkyl is optionally substituted with one, two or three fluoro, OH, cyano or Ci-C4alkoxy, optionally substituted with one, two or three fluoro; or R1 a and R1 b, (as described above) together with the carbon to which they are bonded, form a C3-C4cycloalkyl or a 4-membered heterocycloalkyi, wherein said cycloalkyl or heterocycloalkyi are optionally substituted with one, two, three or four halogen, OH, C-i- C4alkyl, S(Ci-C3alkyl), Ci-C4alkoxy or cyano; or a pharmaceutically acceptable salt thereof.
In a specific embodiment, R1 a and R1 b are each independently hydrogen or methyl; or R1 a and R1 b, together with the carbon to which they are bonded, form an optionally substituted cyclopropyl, cyclobutyl or an oxetane; or a pharmaceutically acceptable salt thereof.
In another embodiment, L is NH; Ra and Rb are H; and x is 1 ; or a pharmaceutically acceptable salt thereof. In another specific embodiment, R3 and R4 are each independently hydrogen or Ci-C4alkyl, wherein said alkyl is optionally substituted with one, two, three or four R9; or R3 and R4 taken together with the respective carbons to which they are bonded (as described above) form a -(4- to 12- membered heterocycloalkyi), having one to two heteroatoms selected from N, O or S, wherein the heterocycloalkyi is optionally substituted with one, two, three or four R9; R9 is OH, CH2OH, halogen, Ci-C4alkyl, Ci-C4alkoxy or cyano; or a pharmaceutically acceptable salt thereof. In another embodiment, G is selected from the following exemplary moieties: pyrazinyl, pyrimidinyl, pyridinyl or pyridazinyl, (each of which is optionally substituted with methyl, CH2F, CHF2 or CF3); and wherein such moieties are either carbon-linked or nitrogen-linked; or a pharmaceutically acceptable salt thereof. In a specific embodiment, R2a and R2b together with the carbon to which they are bonded form a oxetane, tetrahydrofuran, tetrahydropyran, cyclobutyl, cyclopentyl, or cyclohexyl, each of which is optionally substituted with one or two Ci-C4alkyl or OH; or a pharmaceutically acceptable salt thereof. In an another particular embodiment, R2a and R2b are each independently hydrogen; fluoro; OH; Ci-C4alkyl; Ci-C4alkoxy; C3-C6cycloalkyl; 5-membered heteroaryl, having one or two N; cyano; -SO2CH3; -C(O)NHR8a; -NHC(O)NHR8a; wherein said alkyl, alkoxy, cycloalkyi and heteroaryl are optionally substituted by one, two, three or four R9; wherein R9 is OH, fluoro, methyl, ethyl, methoxy or ethoxy; or a pharmaceutically acceptable salt thereof.
In another emobodiment, one of R2a or R2b, taken together with the carbon to which they are bonded, and one of R5a or R5b, form a cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, tetrahydrofuran or phenyl, wherein each is optionally substituted with one, two, three or four R9, wherein R9 is OH, CH2F, CHF2, CF3, CH2OH; or a pharmaceutically acceptable salt thereof.
In a particular, embodiment, the invention comprises a compound of Formula la
Figure imgf000034_0001
la
wherein Z is -(CR5aR5b)q-; and q is 1 ;
R2a and R2b are each independently hydrogen, methyl, ethyl, propyl,
isopropyl, methoxy or ethoxy, optionally substituted with R9 wherein R9 is OH; or
R2a and R2b, together with the carbon to which they are bonded, form a
tetrahydrofuran, cyclobutane, cyclopentane, cyclohexane, oxetane, tetrahydropyran, pyrrolidine, azetidine, each of which is optionally substituted with one, two, three or four R9; or R2a or R2b, and
one of R5a or R5b, together with the respective carbons to which they are bonded, form a cyclopentane or cyclohexane, optionally substituted with one, two or three R9; or a pharmaceutically acceptable salt of said compound or a tautomer of said compound or said salt. In specific embodiments, the invention is directed to a compound of Formula
Ila,
Figure imgf000035_0001
wherein R10 is hydrogen; and L is NH; G is pyrimidinyl or pyrazinyl; or a
pharmaceutically acceptable salt thereof.
In a more specific embodiment, the invention is directed to a compound having the following absolute stereochemistry,
Figure imgf000035_0002
wherein R1 a is hydrogen; and R1 b is methyl, ethyl, propyl, wherein each is optionally substituted with one, two or three fluoro; or a pharmaceutically acceptable salt thereof. And, in another embodiment, Ra, Rb, R3, R4, R5a and R5b are hydrogen; and x is 1 ; R1 b is methyl or ethyl, optionally substituted with one, two or three fluoro, or a pharmaceutically acceptable salt thereof.
In another embodiment, the invention is directed to a compound of Formula lb
Figure imgf000035_0003
lb
wherein Z is a bond; or a pharmaceutically acceptable salt thereof. In particular, the invention is directed to compounds wherein R2a and R2b are each independently hydrogen, methyl, ethyl, propyl, isopropyl, methoxy or ethoxy, optionally substituted with R9 wherein R9 is OH; or R2a and R2b together with the carbon to which they are bonded form a tetrahydrofuran, cyclobutane, cyclopentane, cyclohexane, oxetane, tetrahydropyran, pyrrolidine, azetidine, each of which is optionally substituted with one, two, three or four R9; or a pharmaceutically acceptable salt thereof.
In another aspect, the invention is directed to the compound of Formula lib,
Figure imgf000036_0001
lib
wherein R10 is hydrogen; and L is NH; or a pharmaceutically acceptable salt thereof. In a specific embodiment, the compound of Formula Mb has the absolute
stereochemistry as depicted herein:
Figure imgf000036_0002
wherein R1 a is hydrogen; and R1 b is methyl, ethyl or propyl, each of which is optionally substituted by one, two or three fluoro; or a pharmaceutically acceptable salt thereof. In another aspect, Ra, Rb, R3 and R4 are hydrogen; x is 1 ; R1 b is methyl or ethyl, optionally substituted with one, two or three fluoro; and G is pyrimidinyl or pyrazinyl; or a pharmaceutically acceptable salt thereof.
In another embodiment, the invention is directed to a compound of Formula le
Figure imgf000036_0003
le
wherein Z is-(CR5aR5b)m-W-(CR5aR5b)n-, W is NR7, m is 1 , and n is 0; or a
pharmaceutically acceptable salt thereof. In another aspect, L is NH; and R2a or R' and R7, together with the respective atoms to which they are bonded form a -(4- to 12- membered heterocycloalkyi), having one or two heteroatoms selected from N or 0 , wherein said heterocycloalkyi is optionally substituted with one, two, three or four R9; or a pharmaceutically acceptable salt thereof. In another aspect, Ra, Rb, R3, R4, R5a and R5b are hydrogen and the heterocycloalkyi formed is a pyrrole; or a pharmaceutically acceptable salt thereof. Yet another aspect of the invention is the compound of Formula lie,
Figure imgf000037_0001
wherein R1a is hydrogen; and R1b is methyl, ethyl, propyl, each of which is optionally substituted by one, two or three fluoro; or a pharmaceutically acceptable salt thereof. In a particular aspect, G is pyrimidinyl or pyrazinyl, or a pharmaceutically acceptable salt thereof.
In another embodiment, the invention is directed to a compound of Formula Ic
Figure imgf000037_0002
Ic
wherein Z is-(CR5aR5b)q-; and q is 2;
R5a and R5b are each independently hydrogen, OH, fluoro, cyano, Ci-C4alkyl,
Ci-C4alkoxy, cyclopropyl, cyclobutyl, cyclopentyl, oxazolidinone, - (CR6aR6b)tC(O)NH2, optionally substituted with one, two, three or four R9; or
R5a and R5b taken together with the carbon to which they are bonded form a oxetane, tetrahydrofuran, tetrahydropyran, oxazolidinone, cyclopentane, cyclohexane, cyclobutane, cyclopropane, wherein said cycloalkyi or heterocycloalkyi are optionally substituted with one, two, three or four R9;
R9 is fluoro, OH or Ci-C4alkoxy, and t is 0 or 1 ;
or a pharmaceutically acceptable salt thereof. In a particular aspect of the invention, Ra, Rb, R3, R4, R5a and R5b are hydrogen and the heterocycloalkyl formed is a pyrrole; and L is NH; or a pharmaceutically acceptable salt thereof. In a more specific aspect, the compound of Formula lie is
Figure imgf000038_0001
lie
wherein G is pyrimidinyl or pyrazinyl; or a pharmaceutically acceptable salt thereof. In particular, the invention is directed to a compound having the absolute
stereochemistry of,
Figure imgf000038_0002
wherein R1 a is hydrogen; and R1 b is methyl, ethyl, propyl, each of which is optionally substituted with one, two or three fluoro; or a pharmaceutically acceptable salt thereof
Specific embodiments of the invention include the examples 1 -205, as described herein, the specific enantiomers thereof, as well as the racemate mixtures. Vanin-1 Indications
On the basis of what is known the in the state of the art literature and the pattern of Vanin-1 expression in human health and disease systems, the compounds of the invention are also useful in treating and/or preventing a disease or condition mediated by or otherwise associated with a Vanin-1 enzyme. The use of compounds of the invention may be useful in diseases where there is evidence of oxidative stress and/or Vanin-1 enzyme upregulation; the method comprising administering to a subject in need thereof an effective amount of a compound of the invention.
The disease may be, but not limited to, one of the following classes: autoimmune diseases, inflammatory diseases, allergic diseases, metabolic diseases, infection-based diseases, trauma or tissue-injury based diseases, fibrotic diseases, cardiovascular diseases, respiratory diseases, renal diseases, dermatological diseases, liver diseases, gastrointestinal diseases, oral diseases, pain and sensory diseases, and hematopoietic diseases. Specific autoimmune diseases include, but are not limited to: rheumatoid arthritis, osteoarthritis, psoriasis, allergic dermatitis, systemic lupus erythematosus (and resulting complications), Sjogren's syndrome, multiple sclerosis, asthma, glomerular nephritis, inflammatory bowel disease, Crohn's disease, ankylosing spondylitis, Behget's disease, lupus nephritis, scleroderma, systemic scleroderma, alopecia universalis, acute disseminated encephalomyelitis, antiphospholipid antibody syndrome, atrophic gastritis of pernicious anemia, autoimmune alopecia, autoimmune hepatitis, autoimmune encephalomyelitis, autoimmune thrombocytopenia, chronic hepatitis, Cogan's syndrome, endometriosis, Goodpasture's syndrome, Graves' disease, Guillain-Barre syndrome, Hashimoto's disease (or Hashimoto's thyroiditis), hidradentitis suppurativa, idiopathic thrombocytopenia purpura, interstitial cystitis, membranous glomerulopathy, morphea, polyarteritis nodosa, polymyositis, primary biliary cirrhosis, systemic sclerosis, temporal arteritis, thyroiditis, vasculitis, vitiglio, Wegner's granulomatosis, palmoplantar keratoderma, systemic-onset Juvenile Idiopathic Arthritis (SJIA), or an indication listed in a separate category herein. Specific inflammatory diseases include, but are not limited to: chronic obstructive pulmonary diseases, airway hyper-responsiveness, cystic fibrosis, acute respiratory distress syndrome, sinusitis, rhinitis, gingivitis, atherosclerosis, chronic prostatitis, glomerular nephritis, ulcerative colitis, uveitis, periodontal disease, or an indication listed in a separate category herein. Specific pain conditions include, but are not limited to: inflammatory pain, pain due to burns, interstitial cystitis, post-traumatic injury, pain associated with irritable bowel syndrome, gout, pain associated with any of the other indications listed within this specification, or an indication listed in a separate category herein.
Specific respiratory, airway and pulmonary conditions include, but are not limited to: asthma (which may encompass chronic, late, bronchial, allergic, intrinsic, extrinsic or dust), chronic obstructive pulmonary disease, idiopathic pulmonary fibrosis, pulmonary arterial hypertension, cystic fibrosis, interstitial lung disease, acute lung injury, sarcoidosis, allergic rhinitis, chronic cough, bronchitis, recurrent airway obstruction, emphysema, or bronchospasm, or an indication listed in a separate disease category herein.
Specific gastrointestinal (Gl) disorders include, but are not limited to: Irritable Bowel Syndrome (IBS), Inflammatory Bowel Disease (IBD), biliary colic and other biliary disorders, renal colic, diarrhea-dominant IBS, pain associated with Gl distension, ulcerative colitis, Crohn's Disease, irritable bowel syndrome, Celiac disease, proctitis, eosinophilic gastroenteritis, mastocytosis, or an indication listed in a separate disease category herein.
Specific allergic diseases include, but are not limited to: anaphylaxis, allergic rhinitis, allergic dermatitis, allergic urticaria, angioedema, allergic asthma, allergic reactions to: food, drugs, insect bites, pollen; or an indication listed in a separate disease category herein.
Specific infection-based diseases include, but are not limited to: sepsis, septic shock, viral diseases, malaria, Lyme disease, ocular infections, conjunctivitis, Whipple Disease, or an indication listed in a separate disease category herein.
Specific trauma and tissue injury-based conditions include, but are not limited to: Renal glomerular damage, reperfusion injury (for example to heart, kidney, lung), spinal cord injury, tissue scarring, tissue adhesion, or an indication listed in a separate disease category herein. Specific fibrotic diseases include, but are not limited to: Idiopathic pulmonary fibrosis, liver fibrosis, renal fibrosis, or an indication listed in a separate disease category herein.
Specific skin/ dermatological diseases include, but are not limited to: psoriasis, atopic dermatitis, cutaneous lupus, acne, eczema, pruritus, scleroderma, Sweet Syndrome/neutrophilic dermatosis, neutrophilic panniculitis, acrodermatitis (form of pustular psoriasis), or an indication listed in a separate disease category herein.
Specific renal diseases include, but are not limited to: acute kidney injury (AKI)
(sepsis-AKI, coronary artery bypass graft-AKI, cardiac surgery-AKI, non-cardiac surgery-AKI, transplant surgery-AKI cisplatin-AKI, contrast/imaging agent induced-AKI), glomerulonephritis, IgA nephropathy, crescentic GN, lupus nephritis, HIV associated nephropathy, membraneous nephropathy, C3 glomerulopathy, ANCA vasculitis, diabetic nephropathy, nephrotic syndrome, hypertensive nephrosclerosis, focal segmental glomerulosclerosis, Alport syndrome, Fanconi, syndrome, crystal nephropathy, nephrotic syndrome, amyloidosis, glomerulonephritis in SJIA, or an indication listed in a separate disease category herein.
Specific liver diseases include, but are not limited to: liver fibrosis, liver cirrhosis, nonalcoholic steatohepatitis (NASH), or an indication listed in a separate disease category herein.
Specific oral diseases include, but are not limited to: gingivitis, periodontal disease or an indication listed in a separate disease category herein.
Specific metabolic diseases include, but are not limited to: Type 2 diabetes (and resulting complications), hyperlipidemia, non-alcholic fatty liver disease, metabolic syndrome, insulin resistance, obesity, or an indication listed in a separate disease category herein.
Compounds of the current invention are also useful in the treatment of a proliferative disease selected from a benign or malignant tumor, solid tumor, carcinoma of the brain, kidney, liver, adrenal gland, bladder, breast, stomach, gastric tumors, ovaries, colon, rectum, prostate, pancreas, lung, vagina, cervix, testis, genitourinary tract, esophagus, larynx, skin, bone or thyroid, sarcoma, glioblastomas, neuroblastomas, gastrointestinal cancer, especially colon carcinoma or colorectal adenoma, an epidermal hyperproliferation, psoriasis, prostate hyperplasia, a neoplasia, a neoplasia of epithelial character, adenoma, adenocarcinoma, keratoacanthoma, epidermoid carcinoma, large cell carcinoma, nonsmall-cell lung carcinoma, a mammary carcinoma, follicular carcinoma, undifferentiated carcinoma, or an indication listed in a separate disease category herein.
Cardiovascular conditions include, but are not limited to coronary heart disease, acute coronary syndrome, ischaemic heart disease, post-myocardial infarction cardiac remodeling atrial fibrillation, myocardial and vascular fibrosis, vascular wall hypertrophy, endothelial thickening, adverse remodeling, stroke, and the like, or an indication listed in a separate disease category herein.
Cardiovascular complications of type 2 diabetes are associated with inflammation, accordingly, the compounds of the present invention may be used to treat diabetes and diabetic complications such as macrovascular disease, hyperglycemia, metabolic syndrome, impaired glucose tolerance, fatty liver disease, cataracts, diabetic neuropathy, diabetic nephropathy, diabetic retinopathy, obesity, dyslipidemia, hypertension, hyperinsulinemia, and insulin resistance syndrome, or an indication listed in a separate disease category herein.
Linkage of oxidative stress and inflammation to disease has been demonstrated in neuroinflammatory and neurodegenerative conditions. Therefore, the compounds of the present invention are particularly indicated for use in the treatment of neuroinflammatory and neurodegenerative conditions (i.e., disorders or diseases) in mammals including humans such as multiple sclerosis, Alzheimer's disease; Parkinson's disease; brain injury; stroke; cerebrovascular diseases; dementia, acute stress disorder, generalized anxiety disorder, social anxiety disorder, panic disorder, post-traumatic stress disorder and obsessive-compulsive disorder; depression, or an indication listed in a separate disease category herein.
Typically, a compound of the invention is administered in an amount effective to treat a condition as described herein. The compounds of the invention are administered by any suitable route in the form of a pharmaceutical composition adapted to such a route, and in a dose effective for the treatment intended. Therapeutically effective doses of the compounds required to treat the progress of the medical condition are readily ascertained by one of ordinary skill in the art using preclinical and clinical approaches familiar to the medicinal arts.
The term "pharmaceutically acceptable" means the substance or composition must be compatible, chemically and/or toxicologically, with the other ingredients comprising a formulation, and/or the mammal being treated therewith.
The term "therapeutically effective amount" means an amount of a compound of the present invention that (i) treats or prevents the particular disease, condition, or disorder, (ii) attenuates, ameliorates, or eliminates one or more symptoms of the particular disease, condition, or disorder, or (iii) prevents or delays the onset of one or more symptoms of the particular disease, condition, or disorder described herein. In particular, effective amounts of the compounds of the invention generally include any amount sufficient to detectably modulate vanin-1 activity, and in one embodiment inhibit vanin-1 enzyme, or to alleviate symptoms of diseases associated with vanin-1 activity, and in one embodiment those associated with inhibition of vanin-1 enzyme, or susceptible to vanin-1 activity modulation, in one embodiment inhibition of vanin-1 enzyme. For example, with respect to the treatment of asthma, a therapeutically effective amount preferably refers to the amount of a therapeutic agent that increases peak air flow by at least 5%, preferably at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or at least 100%.%. In reference to the treatment of cancer, a therapeutically effective amount refers to that amount which has the effect of (1 ) reducing the size of the tumor, (2) inhibiting (that is, slowing to some extent, preferably stopping) tumor metastasis, (3) inhibiting to some extent (that is, slowing to some extent, preferably stopping) tumor growth or tumor invasiveness, and/or (4) relieving to some extent (or, preferably, eliminating) one or more signs or symptoms associated with the cancer.
The term "abnormal cell growth" as used herein, unless otherwise indicated, refers to cell growth that is independent of normal regulatory mechanisms (e.g., loss of contact inhibition). Abnormal cell growth may be benign (not cancerous), or malignant (cancerous).
As used herein "cancer" refers to any malignant and/or invasive growth or tumor caused by abnormal cell growth. As used herein "cancer" refers to solid tumors named for the type of cells that form them, or cancers of blood, bone marrow, or the lymphatic system. Examples of solid tumors include but not limited to sarcomas and carcinomas. Examples of cancers of the blood include but not limited to leukemias, lymphomas and myeloma. The term "cancer" includes but is not limited to a primary cancer that originates at a specific site in the body, a metastatic cancer that has spread from the place in which it started to other parts of the body, a recurrence from the original primary cancer after remission, and a second primary cancer that is a new primary cancer in a person with a history of previous cancer of different type from latter one.
The term "treating", as used herein, unless otherwise indicated, means reversing, alleviating, inhibiting the progress of, delaying the progression of, delaying the onset of, or preventing the disorder or condition to which such term applies, or one or more symptoms of such disorder or condition. The term "treatment", as used herein, unless otherwise indicated, refers to the act of treating as "treating" is defined immediately above. The term "treating" also includes adjuvant and neo-adjuvant treatment of a subject. For the avoidance of doubt, reference herein to "treatment" includes reference to curative, palliative and prophylactic treatment, and to the administration of a medicament for use in such treatment.
The compounds of the invention may be administered orally. Oral administration may involve swallowing, so that the compound enters the gastrointestinal tract, or buccal or sublingual administration may be employed, by which the compound enters the blood stream directly from the mouth.
In another embodiment, the compounds of the invention may also be administered directly into the blood stream, into muscle, or into an internal organ. Suitable means for parenteral administration include intravenous, intraarterial, intraperitoneal, intrathecal, intraventricular, intraurethral, intrasternal, intracranial, intramuscular and subcutaneous. Suitable devices for parenteral administration include needle (including microneedle) injectors, needle-free injectors and infusion techniques.
In another embodiment, the compounds of the invention may also be administered topically to the skin or mucosa, that is, dermally or transdermally. In another embodiment, the compounds of the invention can also be administered intranasally or by inhalation. In another embodiment, the compounds of the invention may be administered rectally or vaginally. In another embodiment, the compounds of the invention may also be administered directly to the eye or ear.
The dosage regimen for the compounds and/or compositions containing the compounds is based on a variety of factors, including the type, age, weight, sex and medical condition of the patient; the severity of the condition; the route of administration; and the activity of the particular compound employed. Thus the dosage regimen may vary widely. Dosage levels of the order from about 0.01 mg to about 100 mg per kilogram of body weight per day are useful in the treatment of the above- indicated conditions. In one embodiment, the total daily dose of a compound of the invention (administered in single or divided doses) is typically from about 0.01 to about 100 mg/kg. In another embodiment, the total daily dose of the compound of the invention is from about 0.1 to about 50 mg/kg, and in another embodiment, from about 0.5 to about 30 mg/kg (i.e., mg compound of the invention per kg body weight). In one embodiment, dosing is from 0.01 to 10 mg/kg/day. In another embodiment, dosing is from 0.1 to 1 .0 mg/kg/day. Dosage unit compositions may contain such amounts or submultiples thereof to make up the daily dose. In many instances, the administration of the compound will be repeated a plurality of times in a day (typically no greater than 4 times). Multiple doses per day typically may be used to increase the total daily dose, if desired.
For oral administration, the compositions may be provided in the form of tablets containing from about 0.01 mg to about 500 mg of the active ingredient, or in another embodiment, from about 1 mg to about 100 mg of active ingredient. Intravenously, doses may range from about 0.1 to about 10 mg/kg/minute during a constant rate infusion.
"Patient" or "subject" refers to mammals and include, but are not limited to, canine, feline, bovine, caprine, equine, ovine, porcine, rodents, lagomorphs, primates, and the like, and encompass mammals in utero. In one embodiment, humans are suitable subjects. Human subjects may be of either gender and at any stage of development. The term "subject" or "patient" as used herein means any mammalian patient or subject to which the compounds of the invention can be administered. In an exemplary embodiment of the present invention, to identify subject patients for treatment according to the methods of the invention, accepted screening methods are employed to determine risk factors associated with a targeted or suspected disease or condition or to determine the status of an existing disease or condition in a subject. These screening methods include, but are not limited to for example, conventional work-ups to determine risk factors that may be associated with the targeted or suspected disease or condition. These and other routine methods allow the clinician to select patients in need of therapy using the methods and compounds of the present invention.
As used herein, the term "inhibitor(s) of vanin-1 enzyme" refers to a compound that binds to the vanin-1 enzyme and decreases the resulting enzymatic activity. As used herein, the term "modulate" as used herein, refers to encompasses either a decrease or an increase in activity or expression depending on the target molecule.
As used herein, the term "other therapeutic agents" as used herein, refers to any therapeutic agent that has been used, is currently used or is known to be useful for treating a disease or a disorder encompassed by the present invention.
As used herein, the term "IC50" refers to an amount, concentration or dosage of a particular test compound that achieves a 50% inhibition of a maximal response in an assay that measures such response. The value depends on the assay used. In another embodiment, the invention comprises the use of one or more compounds of the invention for the preparation of a medicament for the treatment of the conditions recited herein.
For the treatment of the conditions referred to above, the compound of the invention can be administered as compound per se. Alternatively, pharmaceutically acceptable salts are suitable for medical applications because of their greater aqueous solubility relative to the parent compound.
In another embodiment, the present invention comprises pharmaceutical compositions. Such pharmaceutical compositions comprise a compound of the invention presented with a pharmaceutically acceptable carrier. The carrier can be a solid, a liquid, or both, and may be formulated with the compound as a unit-dose composition, for example, a tablet, which can contain from 0.05% to 95% by weight of the active compounds. A compound of the invention may be coupled with suitable polymers as targetable drug carriers. Other pharmacologically active substances can also be present.
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 active compounds and compositions, for example, may be administered orally, rectally, parenterally, or topically.
Oral administration of a solid dose form may be, for example, presented in discrete units, such as hard or soft capsules, pills, cachets, lozenges, or tablets, each containing a predetermined amount of at least one compound of the present invention. In another embodiment, the oral administration may be in a powder or granule form. In another embodiment, the oral dose form is sub-lingual, such as, for example, a lozenge. In such solid dosage forms, the compounds of Formula I are ordinarily combined with one or more adjuvants. Such capsules or tablets may contain a controlled-release formulation. In the case of capsules, tablets, and pills, the dosage forms also may comprise buffering agents or may be prepared with enteric coatings. In another embodiment, oral administration may be in a liquid dose form. Liquid dosage forms for oral administration include, for example, pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and elixirs containing inert diluents commonly used in the art (e.g., water). Such compositions also may comprise adjuvants, such as wetting, emulsifying, suspending, flavoring (e.g., sweetening), and/or perfuming agents.
In another embodiment, the present invention comprises a parenteral dose form. "Parenteral administration" includes, for example, subcutaneous injections, intravenous injections, intraperitoneal injections, intramuscular injections, intrasternal injections, and infusion. Injectable preparations (e.g., sterile injectable aqueous or oleaginous suspensions) may be formulated according to the known art using suitable dispersing, wetting agents, and/or suspending agents.
In another embodiment, the present invention comprises a topical dose form. "Topical administration" includes, for example, transdermal administration, such as via transdermal patches or iontophoresis devices, intraocular administration, or intranasal or inhalation administration. Compositions for topical administration also include, for example, topical gels, sprays, ointments, and creams. A topical formulation may include a compound that enhances absorption or penetration of the active ingredient through the skin or other affected areas. When the compounds of this invention are administered by a transdermal device, administration will be accomplished using a patch either of the reservoir and porous membrane type or of a solid matrix variety. Typical formulations for this purpose include gels, hydrogels, lotions, solutions, creams, ointments, dusting powders, dressings, foams, films, skin patches, wafers, implants, sponges, fibers, bandages and microemulsions. Liposomes may also be used. Typical carriers include alcohol, water, mineral oil, liquid petrolatum, white petrolatum, glycerin, polyethylene glycol and propylene glycol. Penetration enhancers may be incorporated; see, for example, J. Pharm. Sci., 88(10), 955-958, by Finnin and Morgan (October 1999). Formulations suitable for topical administration to the eye include, for example, eye drops wherein the compound of this invention is dissolved or suspended in a suitable carrier. A typical formulation suitable for ocular or aural administration may be in the form of drops of a micronized suspension or solution in isotonic, pH-adjusted, sterile saline. Other formulations suitable for ocular and aural administration include ointments, biodegradable (e.g., absorbable gel sponges, collagen) and nonbiodegradable (e.g., silicone) implants, wafers, lenses and particulate or vesicular systems, such as niosomes or liposomes. A polymer such as cross-linked polyacrylic acid, polyvinyl alcohol, hyaluronic acid, a cellulosic polymer, for example, hydroxypropyl methyl cellulose, hydroxyethyl cellulose, or methyl cellulose, or a heteropolysaccharide polymer, for example, gelan gum, may be incorporated together with a preservative, such as benzalkonium chloride. Such formulations may also be delivered by iontophoresis.
For intranasal administration or administration by inhalation, the active compounds of the invention are conveniently delivered in the form of a solution or suspension from a pump spray container that is squeezed or pumped by the patient or as an aerosol spray presentation from a pressurized container or a nebulizer, with the use of a suitable propellant. Formulations suitable for intranasal administration are typically administered in the form of a dry powder (either alone, as a mixture, for example, in a dry blend with lactose, or as a mixed component particle, for example, mixed with phospholipids, such as phosphatidylcholine) from a dry powder inhaler or as an aerosol spray from a pressurized container, pump, spray, atomizer (preferably an atomizer using electrohydrodynamics to produce a fine mist), or nebulizer, with or without the use of a suitable propellant, such as 1 , 1 , 1 ,2-tetrafluoroethane or 1 , 1 ,1 ,2,3,3, 3-heptafluoropropane. For intranasal use, the powder may comprise a bioadhesive agent, for example, chitosan or cyclodextrin.
In another embodiment, the present invention comprises a rectal dose form. Such rectal dose form may be in the form of, for example, a suppository. Cocoa butter is a traditional suppository base, but various alternatives may be used as appropriate. Other carrier materials and modes of administration known in the pharmaceutical art may also be used. Pharmaceutical compositions of the invention may be prepared by any of the well-known techniques of pharmacy, such as effective formulation and administration procedures. The above considerations in regard to effective formulations and administration procedures are well known in the art and are described in standard textbooks. Formulation of drugs is discussed in, for example, Remington: The Science and Practice of Pharmacy, Mack Publishing Company, Easton, Pa., 21 st Edition (2005); Liberman et al., Eds., Pharmaceutical Dosage Forms, Marcel Decker, New York, N.Y. , 1980; and Kibbe et al., Eds., Handbook of Pharmaceutical Excipients (3rd Ed.), American Pharmaceutical Association, Washington, 1999. These articles are incorporated herein by reference.
The phrase "pharmaceutically acceptable carrier" as used herein means a pharmaceutically-acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting the subject agent from one organ, or portion of the body, to another organ, or portion of the body. Each carrier must be "acceptable" in the sense of being compatible with the other ingredients of the formulation and not injurious to the patient. Some examples of materials which can serve as pharmaceutically-acceptable carriers include: (1 ) sugars, such as lactose, glucose and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; (10) glycols, such as propylene glycol; (1 1 ) polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; (12) esters, such as ethyl oleate and ethyl laurate; (13) agar; (14) buffering agents, such as magnesium hydroxide and aluminum hydroxide; (15) alginic acid; (16) pyrogen-free water; (17) isotonic saline; (18) Ringer's solution; (19) ethyl alcohol; (20) phosphate buffer solutions; and (21 ) other non-toxic compatible substances employed in p harmaceutical formulations. A physiologically acceptable carrier should not cause significant irritation to an organism and does not abrogate the biological activity and properties of the administered compound.
An "excipient" refers to an inert substance added to a pharmacological composition to further facilitate administration of a compound. Examples of excipients include but are not limited to calcium carbonate, calcium phosphate, various sugars and types of starch, cellulose derivatives, gelatin, vegetable oils and polyethylene glycols.
The compounds of the present invention can be used, alone or in combination with other therapeutic agents, in the treatment of various conditions or disease states. The compound(s) of the present invention and other therapeutic agent(s) may be may be administered simultaneously (either in the same dosage form or in separate dosage forms) or sequentially. Simultaneous administration may be carried out by mixing the compounds prior to administration or by administering the compounds at the same point in time but at different anatomic sites or using different routes of administration.
The phrases "concurrent administration," "co-administration," "simultaneous administration," and "administered simultaneously" mean that the compounds are administered in combination.
The present invention includes the use of a combination of an Vanin-1 inhibitor compound as provided in the compound of Formula I and one or more additional pharmaceutically active agent(s). Accordingly, the present invention also includes pharmaceutical compositions comprising an amount of: (a) a first agent comprising a compound of Formula I or a pharmaceutically acceptable salt of the compound; (b) a second pharmaceutically active agent; and (c) a pharmaceutically acceptable carrier, vehicle or diluent. The compounds of the present invention can be administered alone or in combination with one or more additional therapeutic agents. By "administered in combination" or "combination therapy" it is meant that a compound of the present invention and one or more additional therapeutic agents are administered concurrently to the mammal being treated. When administered in combination each component may be administered at the same time or sequentially in any order at different points in time. Thus, each component may be administered separately but sufficiently closely in time so as to provide the desired therapeutic effect. Thus, the methods of prevention and treatment described herein include use of combination agents.
The combination agents are administered to a mammal, including a human, in a therapeutically effective amount. By "therapeutically effective amount" it is meant an amount of a compound of the present invention that, when administered alone or in combination with an additional therapeutic agent to a mammal, is effective to treat the desired disease/condition e.g., inflammatory condition such as systemic lupus erythematosus. See also, T. Koutsokeras and T. Healy, Systemic lupus erythematosus and lupus nephritis, Nat Rev Drug Discov, 2014, 13(3), 173-174, for therapeutic agents useful treating lupus.
Combination Therapies
In particular, it is contemplated that the compounds of the invention may be administered with the following therapeutic agents: Non-steroidal anti-inflammatory drugs (NSAIDs), including but not limited to, nonselective COX1/2 inhibitors such as piroxicam, naproxen, flubiprofen, fenoprofen, ketoprofen, ibuprofen, etodolac (Lodine), mefanamic acid, sulindac, apazone, pyrazolones (such as phenylbutazone), salicylates (such as aspirin); selective COX2 inhibitors such as: celecoxib, rofecoxib, etoricoxib, valdecoxib, meloxicam; Immunomodulatory and/ or anti-inflammatory agents, including but not limited to, methotrexate, leflunomide, ciclesonide chloroquine, hydroxychloroquine, d- penicillamine, auranofin, sulfasalazine, sodium aurothiomalate, cyclosporine, azathioprine, cromolyn, hydroxycarbamide, retinoids, fumarates (such as monomethyl and dimethyl fumarate), glatiramer acetate, mitoxantrone, teriflunomide, suplatast tosilate, mycophenolate mofetil and cyclophosphamide, laquinimod, voclosporin, PUR- 1 18, AMG 357, AMG 81 1 , BCT197;
Antimalarials, including but not limited to, hydroxychloroquine (Plaquenil) and chloroquine (Aralen), cyclophosphamide (Cytoxan), methotrexate (Rheumatrex), azathioprine (Imuran), mesalamine (Asacol) and sulfasalazine (Azulfidine):
Antibiotics, including but not limited to, Flagyl or ciprofloxacin;
Anti-TNFa agents, including but not limited to, infliximab, adalimumab, certolizumab pegol, golimumab and etanercept;
Anti-CD20 agents, including but not limited to, rituximab, ocrelizumab, ofatumumab and PF-05280586;
Antidiarrheals, such as diphenoxylate (Lomotil) and loperamide (Imodium);
Bile acid binding agents, such as cholestyramine, alosetron (Lotronex) and ubiprostone (Amitiza);
Laxatives, such as Milk of Magnesia, polyethylene glycol (MiraLax), Dulcolax,
Correctol and Senokot, and anticholinergics or antispasmodics such as dicyclomine (Bentyl);
T lymphocyte activation inhibitors, including but not limited to, abatacept;
Glucocorticoid receptor modulators that may be dosed orally, by inhalation, by injection, topically, rectally, by ocular delivery, including but not limited to, betamethasone, prednisone, hydrocortisone, prednisolone, flunisolide, triamcinoline acetonide, beclomethasone, dipropionate, budesonide, fluticasone propionate, ciclesonide, mometasone furoate, fluocinonide, desoximetasone, methylprednisolone or PF-04171327;
Aminosalicyic acid derivatives, including but not limited to, sulfasalazine and mesalazine;
Anti integrin agents, including but not limited to, natalizumab, vedolizumab, PF- 00547659, etrolizumab;
a1 - or a2-adrenergic agonist agents including but not limited to: propylhexidrine, phenylephrine, phenylpropanolamine, pseudoephedrine or naphazoline hydrochloride, oxymethazoline hydrochloride, tetrahydrozoline hydrochloride, xylometazoline hydrochloride or ethylnorepinephrine hydrochloride;
β-adrenergic agonists, including but not limited to, metaproterenol, isoprotenerol, isoprenaline, albuterol, salbutamol, formoterol, salmeterol, terbutaline, orciprenaline, botolterol mesylate, pirbuterol; Anticholinergic agents, including but not limited to, ipratropium bromide, tiotropium bromide, oxitropium bromide, aclindinium bromide, glycopyrrolate, pirenzipine or telenzepine;
Inhaled long acting beta-agonists, long acting muscarinic antagonists and long acting corticosteroids, including but not limited, to those included in the following reference: Y. Mushtaq, The COPD pipeline, Nat Rev Drug Discov, 2014, 13(4), 253- 254. http://dx.doi. org/10.1038/nrd425;
Leukotriene pathway modulators, including but not limited to, 5-LO Inhibitors (such as zileuton), FLAP antagonists (such as veliflapon, fiboflapon), LTD4 antagonists (such as montelukast, zafirlukast or pranlukast;
H1 receptor antagonists, including but not limited to, cetirizine, loratidine, desloratidine, fexofenadine, astemizole, azelastine or chlorpheniramine;
PDE4 inhibitors, including but not limited to, apremilast, roflumilast or AN2728;
Vitamin D receptor modulators, including but not limited to, paricalcitol;
Nrf2 pathway activators, including but not limited to, fumarates, sulfurophane and bardoxolone methyl;
Modulators of the RAR-related orphan receptor (ROR) family, in particular
RORg;
Modulator and/ or antagonists of the chemokine receptors, including but not limited to, CCR2 antagonists (such as CCX140, BMS-741672, PF-4634817, CCX-872, NOX-E36), CCR2/5 antagonists (such as PF-4634817), CCR9 (such as vercirnon, CCX507), CCR1 modulators, CCR4 modulators, CCR5 modulators, CCR6 modulators, CXCR6 modulators, CXCR7 modulators) and CXCR2 modulators (such as danirixin, AZD5069);
Prostaglandins, including but not limited to, prostacyclin;
PDE5 inhibitors, including but not limited to, sildenafil, PF-489791 , vardenafil and tadalafil;
Endothelin receptor antagonists, including but not limited to, bosentan, ambrisentan, sparsentan, atrasentan, zibotentan and macitentan;
Soluble guanylate cyclase activators, including but not limited to, riociguat;
Interferons, including but not limited to, interferon beta-1 a interferon beta-1 b; Sphingosine 1 -phosphate receptor modulators, including but not limited to, fingolimod, ponesimod;
Inhibitors of the complement pathway, including but not limited to, C5aR antagonists (such as CCX168, PMX-53, NN8210), C5 inhibitors (such as eculizumab), inhibitors of complement factors B and D, inhibitors of MASP2 (such as OMS-721 ) and ARC-1905;
Inhibitors of Janus kinases (one of more of JAK1 , JAK2, JAK3, TYK2), including but not limited to, decernotinib, cerdulatinib, JTE-052, ruxolitinib, tofacitnib, Baricitinib, Peficitinib, GLPG-0634, INCB-47986, INCB-0391 10, PF-04965842, XL-019, ABT-494, R-348, GSK-2586184, AC-410, BMS-91 1543, PF-06651600, and PF-06263276;
Inhibitors of other anti-inflammatory or immunomodulatory kinases, including but not limited to, spleen tyrosine kinase (SYK) inhibitors, p38 MAP kinase inhibitors (such as PF-3715455, PH-797804, AZD-7624, AKP-001 , UR-13870, FX-005, semapimod, pexmetinib, ARRY-797, RV-568, dilmapimod, ralimetinib), PI3K inhibitors (such as GSK-2126458, pilaralisib, GSK-2269557), PI3Kg and/ or PI3Kd inhibitors (such as CAL- 101/GS-1 101 , duvelisib), JNK inhibitors, ERK1 and/ or 2 inhibitors, IKKb inhibitors, BTK inhibitors, ITK inhibitors, ASK1 inhibitors (such as GS-4997), PKC inhibitors (such as sotrastaurin), TrkA antagonists (such as CT-327), MEK1 inhibitors (such as E6201 );
Antioxidants, including but not limited to, myeloperoxidase inhibitors (such as
AZD-3241 ), NOX4 and other NOX enzymes (such as GKT-137831 ) and N-acetyl cysteine;
Inhibitors of IL5, including but not limited to, mepolizumab, reslizumab and benralizumab;
Inhibitors of IL4, including but not limited to, pascolizumab, altrakincept and pitrakinra;
Inhibitors of IL13, including but not limited to, tralokinumab, anrukinzumab and lebrikizumab;
Anti-IL6 agents, including but not limited to, tocilizumab, olokizumab, siltuximab, PF-4236921 and sirukumab;
Inhibitors/Antagonists of IL17/IL17R, including but not limited to, secukinumab, RG-7624, brodalumab and ixekizumab;
Antagonists of IL12 and/or IL23, including but not limited to, tildrakizumab, guselkumab, MEDI2070 and AMG 139;
Inhibitors of IL33, including but not limited to, AMG 282;
Inhibitors of IL9, including but not limited to, MEDI-528;
Inhibitors of GM-CSF, including but not limited to, MT203;
Anti CD4 agents, including but not limited to, tregalizumab and rigerimod;
CRTH2 antagonists, including but not limited to, AZD-1981 ; Inhibitors of B lymphocyte stimulator (BLYS; also known as BAFF), a protein that is often increased in patients with SLE, including but not limited to, belimumab, tabalumab, blisibimod, and atacicept;
CD22-specific monoclonal antibodies, including but not limited to, epratuzumab; Inhibitors of interferon-a, including but not limited to, sifalimumab and rontalizumab;
Inhibitor of type I interferon receptors, including but not limited to, MEDI-546; FcyRIIB agonists, including but not limited to, SM-101 ;
Modified and/or recombinant versions of Heat Shock Protein 10 (Hsp10, also known as Chaperonin 10 or EPF), including but not limited to, INV-103;
Inhibitors of the TNF superfamily receptor 12A (TWEAK receptor), including but not limited to, BIIB-023, enavatuzumab, and RG-7212;
Inhibitors of xanthine oxidase, including but not limited to, allopurinol, benzbromarone, febuxostat, topiroxostat, tisopurine and inositols;
Inhibitors of URAT1 (also known as SLC22A12), including but not limited to, lesinurad, RDEA 3170, UR1 102 and levotofispam;
Inhibitors of toll-like receptors (TLRs), including but not limited to, one or more of TLR7, TLR8, TLR9 (such as IMO-8400, IMO-3100, DV-1 179), TLR2 and/ or TLR 4 (such as VB-201 , OPN-305);
Agonists of TLRs, including but not limited to, TLR7 (such as GSK2245035,
AZD8848), TLR9 (such as AZD1419);
Activators SIRT1 , including but not limited to, SRT2104;
A3 receptor agonists, including but not limited to, CF101 ;
Other agents of use of the treatment of psoriasis, including but not limited to, IDP-1 18, LAS41004, LEO 80185, LEO 90100, PH-10, WBI-1001 , C T01959, BT-061 , cimzia, ustekinumab, MK-3222/SCH 900222, ACT-128800, AEB071 , aliireiinoin, ASP015K, Apo805K1 , BMS-582949, FP187, hectorai (doxerca!ciferol), LEO 2281 1 , Ly3009104 (INCB28050), calcipotriene foam (STF 1 15489), iofaciiinib (CP-690,550), 518101 and Cyc!oPsorb™;
Antifibrotic agents, including but not limited to: pirfenidone, inhibitors of LOXL2
(such as Simtuzumab), FT-01 1 , modulators of epiregulin and/ or TGFp (such as LY- 3016859), modulators of TGFp (such as LY-2382770, fresolimumab);
Prolyl hydroxylase inhibitors, including but not limited to, GSK1278863, FG-2216, ASP-1517/FG-4592, AKB-6548, JTZ-951 , BAY-85-3934 and DS-1093; Inhibitors of granulocyte macrophage colony-stimulating factor, including but not limited to, GSK3196165 (MOR103), PD-0360324 and mavrilimumab;
Inhibitors of MAdCAM and/ or other cell adhesion molecules, including but not limited to, PF-00547659;
Inhibitors of connective tissue growth factor (CTGF), including but not limited to,
PF-06473871 ; Inhibitors of cathepsin C, including but not limited to, GSK2793660;
Inhibitors of soluble epoxide hydrolase, including but not limited to, GSK2269557;
Inhibitors of the TNFR1 associated death domain protein, including but not limited to, GSK2862277;
Anti-CD19 agents, including but not limited to, MEDI-551 and AMG 729;
Anti-B7RP1 agents/ inhibitors of ICOS ligand, including but not limited to, MEDI5872 and AMG-557;
Inhibitors of thymic stromal lymphoprotein, including but not limited to, AMG157; Inhibitors of IL2, including but not limited to, daclizumab;
Checkpoint inhibitors, including but not limited to those which target CTLA4, PD- 1 , PD-L1 , including but not limited to Ipilimumab, tremelimumab, nivolumab, pembrolizumab, avelumab,
Inhibitors of Leucine rich repeat neuronal protein 6A, including but not limited to, Anti-Lingo (Biogen);
Inhibitors of integrins, including but not limited to, alpha-V/beta-6 (STX-100) and alpha-V/beta-3 (VPI-2690B);
Anti-CD40L agents, including but not limited to, CDP-7657;
Modulators of the dopamine D3 receptor, including but not limited to, ABT-614; Inhibitors and/ or modulators of galectin-3, including but not limited to, GCS-100 and GR-MD-02;
Agents for treating diabetic nephropathy, including but not limited to, DA-9801 and ASP-8232;
Agents for treating acute kidney injury, including but not limited to, THR-184, TRC-160334, NX-001 , EA-230, ABT-719, CMX-2043, BB-3 and MTP-131 ;
Modulators of inflammasomes, including but not limited to, inhibitors of NLRP3; Modulators of bromodomains, including but not limited to, BRD4;
Modulators of short-chain fatty acid receptors, including but not limited to, GPR43, GPR109; and Inhibitors of TRP channels, including but not limited to, TRPA1 , TRPC3, TRPC5, TRPC6 and TRPC6.
Additional therapeutic agents include anti-coagulant or coagulation inhibitory agents, anti-platelet or platelet inhibitory agents, thrombin inhibitors, thrombolytic or fibrinolytic agents, anti-arrhythmic agents, anti-hypertensive agents, calcium channel blockers (L-type and T-type), cardiac glycosides, diuretics, mineralocorticoid receptor antagonists, NO donating agents such as organonitrates, NO promoting agents such as phosphodiesterase inhibitors, cholesterol/lipid lowering agents and lipid profile therapies, anti-diabetic agents, anti-depressants, anti-inflammatory agents (steroidal and non-steroidal), anti-osteoporosis agents, hormone replacement therapies, oral contraceptives, anti-obesity agents, anti-anxiety agents, anti-proliferative agents, antitumor agents, anti-ulcer and gastroesophageal reflux disease agents, growth hormone and/or growth hormone secretagogues, thyroid mimetics (including thyroid hormone receptor antagonist), anti-infective agents, anti-viral agents, anti-bacterial agents, and anti-fungal agents.
Agents used in an ICU setting are included, for example, dobutamine, dopamine, epinephrine, nitroglycerin, nitroprusside, etc.
Combination agents useful for treating vasculitis are included, for example, azathioprine, cyclophosphamide, mycophenolate, mofetil, rituximab, etc.
In another embodiment, the present invention provides a combination wherein the second agent is at least one agent selected from a factor Xa inhibitor, an anti-coagulant agent, an anti-platelet agent, a thrombin inhibiting agent, a thrombolytic agent, and a fibrinolytic agent. Exemplary factor Xa inhibitors include apixaban and rivaroxaban. Examples of suitable anti-coagulants for use in combination with the compounds of the present invention include heparins (e.g., unfractioned and low molecular weight heparins such as enoxaparin and dalteparin).
In another embodiment the second agent is at least one agent selected from warfarin, unfractionated heparin, low molecular weight heparin, synthetic pentasaccharide, hirudin, argatrobanas, aspirin, ibuprofen, naproxen, sulindac, indomethacin, mefenamate, droxicam, diclofenac, sulfinpyrazone, piroxicam, ticlopidine, clopidogrel, tirofiban, eptifibatide, abciximab, melagatran, disulfatohirudin, tissue plasminogen activator, modified tissue plasminogen activator, anistreplase, urokinase, and streptokinase.
In another embodiment, the agent is at least one anti-platelet agent. Especially preferred anti-platelet agents are aspirin and clopidogrel. The term anti-platelet agents (or platelet inhibitory agents), as used herein, denotes agents that inhibit platelet function, for example by inhibiting the aggregation, adhesion or granular secretion of platelets. Agents include, but are not limited to, the various known non-steroidal antiinflammatory drugs (NSAIDS) such as aspirin, ibuprofen, naproxen, sulindac, indomethacin, mefenamate, droxicam, diclofenac, sulfinpyrazone, piroxicam, and pharmaceutically acceptable salts or prodrugs thereof. Of the NSAIDS, aspirin (acetylsalicyclic acid or ASA) and COX-2 inhibitors such as celecoxib or piroxicam are preferred. Other suitable platelet inhibitory agents include llb/llla antagonists (e.g., tirofiban, eptifibatide, and abciximab), thromboxane-A2-receptor antagonists (e.g., ifetroban), thromboxane-A2-synthetase inhibitors, PDE3 inhibitors (e.g., Pletal, dipyridamole), and pharmaceutically acceptable salts or prodrugs thereof.
The term anti-platelet agents (or platelet inhibitory agents), as used herein, is also intended to include ADP (adenosine diphosphate) receptor antagonists, preferably antagonists of the purinergic receptors P2Y1 and P2Y12. w'tn ^2^12 being even more preferred. Preferred P2Y12 receptor antagonists include ticagrelor, prasugrel, ticlopidine and clopidogrel, including pharmaceutically acceptable salts or prodrugs thereof. Clopidogrel is an even more preferred agent. Ticlopidine and clopidogrel are also preferred compounds since they are known to be gentle on the gastro-intestinal tract in use.
The term thrombin inhibitors (or anti-thrombin agents), as used herein, denotes inhibitors of the serine protease thrombin. By inhibiting thrombin, various thrombin-mediated processes, such as thrombin-mediated platelet activation (that is, for example, the aggregation of platelets, and/or the granular secretion of plasminogen activator inhibitor-1 and/or serotonin) and/or fibrin formation are disrupted. A number of thrombin inhibitors are known to one of skill in the art and these inhibitors are contemplated to be used in combination with the present compounds. Such inhibitors include, but are not limited to, boroarginine derivatives, boropeptides, heparins, hirudin, argatroban, and melagatran, including pharmaceutically acceptable salts and prodrugs thereof. Boroarginine derivatives and boropeptides include N-acetyl and peptide derivatives of boronic acid, such as C-terminal alpha-aminoboronic acid derivatives of lysine, ornithine, arginine, homoarginine and corresponding isothiouronium analogs thereof. The term hirudin, as used herein, includes suitable derivatives or analogs of hirudin, referred to herein as hirulogs, such as disulfatohirudin. The term thrombolytics or fibrinolytic agents (or thrombolytics or fibrinolytics), as used herein, denote agents that lyse blood clots (thrombi). Such agents include tissue plasminogen activator (natural or recombinant) and modified forms thereof, anistreplase, urokinase, streptokinase, tenecteplase (TNK), lanoteplase (nPA), factor Vila inhibitors, PAI-1 inhibitors (i.e., inactivators of tissue plasminogen activator inhibitors), alpha2- antiplasmin inhibitors, and anisoylated plasminogen streptokinase activator complex, including pharmaceutically acceptable salts or prodrugs thereof. The term anistreplase, as used herein, refers to anisoylated plasminogen streptokinase activator complex, as described, for example, in EP 028,489, the disclosure of which is hereby incorporated herein by reference herein. The term urokinase, as used herein, is intended to denote both dual and single chain urokinase, the latter also being referred to herein as prourokinase. Examples of suitable anti-arrythmic agents include: Class I agents (such as propafenone); Class II agents (such as metoprolol, atenolol, carvadiol and propranolol); Class III agents (such as sotalol, dofetilide, amiodarone, azimilide and ibutilide); Class IV agents (such as ditiazem and verapamil); K+ channel openers such as lAch inhibitors, and lj<ur inhibitors (e.g., compounds such as those disclosed in WO01/40231 ).
The compounds of the present invention may be used in combination with antihypertensive agents and such antihypertensive activity is readily determined by those skilled in the art according to standard assays (e.g., blood pressure measurements). Examples of suitable anti-hypertensive agents include: alpha adrenergic blockers; beta adrenergic blockers; calcium channel blockers (e.g., diltiazem, verapamil, nifedipine and amlodipine); vasodilators (e.g., hydralazine), diruetics (e.g., chlorothiazide, hydrochlorothiazide, flumethiazide, hydroflumethiazide, bendroflumethiazide, methylchlorothiazide, trichloromethiazide, polythiazide, benzthiazide, ethacrynic acid tricrynafen, chlorthalidone, torsemide, furosemide, musolimine, bumetanide, triamtrenene, amiloride, spironolactone); renin inhibitors; ACE inhibitors (e.g., captopril, zofenopril, fosinopril, enalapril, ceranopril, cilazopril, delapril, pentopril, quinapril, ramipril, lisinopril); AT-1 receptor antagonists (e.g., losartan, irbesartan, valsartan); ET receptor antagonists (e.g., sitaxsentan, atrasentan and compounds disclosed in U.S. Patent Nos. 5,612,359 and 6,043,265); Dual ET/AII antagonist (e.g., compounds disclosed in WO 00/01389); neutral endopeptidase (NEP) inhibitors; vasopepsidase inhibitors (dual NEP-ACE inhibitors) (e.g., gemopatrilat and nitrates). An exemplary antianginal agent is ivabradine. Examples of suitable calcium channel blockers (L-type or T-type) include diltiazem, verapamil, nifedipine and amlodipine and mibefradil. Examples of suitable cardiac glycosides include digitalis and ouabain.
In one embodiment, a compound of the invention may be co-administered with one or more diuretics. Examples of suitable diuretics include (a) loop diuretics such as furosemide (such as LASIX™), torsemide (such as DEMADEX™), bemetanide (such as BUMEX™), and ethacrynic acid (such as EDECRIN™); (b) thiazide-type diuretics such as chlorothiazide (such as DIURIL™, ESIDRIX™ or HYDRODIURIL™), hydrochlorothiazide (such as MICROZIDE™ or ORETIC™), benzthiazide, hydroflumethiazide (such as SALURON™), bendroflumethiazide, methychlorthiazide, polythiazide, trichlormethiazide, and indapamide (such as LOZOL™); (c) phthalimidine- type diuretics such as chlorthalidone (such as HYGROTON™), and metolazone (such as ZAROXOLYN™); (d) quinazoline-type diuretics such as quinethazone; and (e) potassium-sparing diuretics such as triamterene (such as DYRENIUM™), and amiloride (such as MIDAMOR™ or MODURETIC™). In another embodiment, a compound of the invention may be co-administered with a loop diuretic. In still another embodiment, the loop diuretic is selected from furosemide and torsemide. In still another embodiment, one or more compounds of the invention may be co-administered with furosemide. In still another embodiment, one or more compounds of the invention may be co- administered with torsemide which may optionally be a controlled or modified release form of torsemide.
In another embodiment, a compound of the invention may be co-administered with a thiazide-type diuretic. In still another embodiment, the thiazide-type diuretic is selected from the group consisting of chlorothiazide and hydrochlorothiazide. In still another embodiment, one or more compounds of the invention may be co-administered with chlorothiazide. In still another embodiment, one or more compounds of the invention may be co-administered with hydrochlorothiazide. In another embodiment, one or more compounds of the invention may be co-administered with a phthalimidine- type diuretic. In still another embodiment, the phthalimidine-type diuretic is chlorthalidone.
Examples of suitable combination mineralocorticoid receptor antagonists include spironolactone and eplerenone. Examples of suitable combination phosphodiesterase inhibitors include: PDE3 inhibitors (such as cilostazol); and PDE5 inhibitors (such as sildenafil). The compounds of the present invention may be used in combination with cholesterol modulating agents (including cholesterol lowering agents) such as a lipase inhibitor, an HMG-CoA reductase inhibitor, an HMG-CoA synthase inhibitor, an HMG- CoA reductase gene expression inhibitor, an HMG-CoA synthase gene expression inhibitor, an MTP/Apo B secretion inhibitor, a CETP inhibitor, a bile acid absorption inhibitor, a cholesterol absorption inhibitor, a cholesterol synthesis inhibitor, a squalene synthetase inhibitor, a squalene epoxidase inhibitor, a squalene cyclase inhibitor, a combined squalene epoxidase/squalene cyclase inhibitor, a fibrate, niacin, an ion- exchange resin, an antioxidant, an ACAT inhibitor or a bile acid sequestrant or an agent such as mipomersen.
Examples of suitable cholesterol/lipid lowering agents and lipid profile therapies include: HMG-CoA reductase inhibitors (e.g., pravastatin, lovastatin, atorvastatin, simvastatin, fluvastatin, NK-104 (a.k.a. itavastatin, or nisvastatin or nisbastatin) and ZD-4522 (a.k.a. rosuvastatin, or atavastatin or visastatin)); squalene synthetase inhibitors; fibrates; bile acid sequestrants (such as questran); ACAT inhibitors; MTP inhibitors; lipooxygenase inhibitors; cholesterol absorption inhibitors; and cholesteryl ester transfer protein inhibitors.
Anti-inflammatory agents also include sPLA2 and lpPLA2 inhibitors (such as darapiadib), 5 LO inhibitors (such as atrelueton) and IL-1 and IL-1 r antagonists (such as canakinumab).
Other atherosclerotic agents include agents that modulate the action of PCSK9, for example, called bococizumab.
Cardiovascular complications of type 2 diabetes are associated with deleterious levels of MPO, accordingly, the compounds of the present invention may be used in combination with anti-diabetic agents, particularly type 2 anti-diabetic agents. Examples of suitable anti-diabetic agents include (e.g. insulins, metfomin, DPPIV inhibitors, GLP-1 agonists, analogues and mimetics, SGLT1 and SGLT2 inhibitors) Suitable anti-diabetic agents include an acetyl-CoA carboxylase- (ACC) inhibitor such as those described in WO2009144554, WO2003072197, WO2009144555 and WO2008065508, a diacylglycerol O-acyltransferase 1 (DGAT-1 ) inhibitor, such as those described in WO09016462 or WO2010086820, AZD7687 or LCQ908, diacylglycerol O- acyltransferase 2 (DGAT-2) inhibitor, monoacylglycerol O-acyltransferase inhibitors, a PDE10 inhibitor, an AMPK activator, a sulfonylurea (e.g., acetohexamide, chlorpropamide, diabinese, glibenclamide, glipizide, glyburide, glimepiride, gliclazide, glipentide, gliquidone, glisolamide, tolazamide, and tolbutamide), a meglitinide, an a- amylase inhibitor (e.g., tendamistat, trestatin and AL-3688), an a-glucoside hydrolase inhibitor (e.g., acarbose), an a-glucosidase inhibitor (e.g., adiposine, camiglibose, emiglitate, miglitol, voglibose, pradimicin-Q, and salbostatin), a PPARy agonist (e.g., balaglitazone, ciglitazone, darglitazone, englitazone, isaglitazone, pioglitazone and rosiglitazone), a PPAR α/γ agonist (e.g., CLX-0940, GW-1536, GW-1929, GW-2433, KRP-297, L-796449, LR-90, MK-0767 and SB-219994), a biguanide (e.g., metformin), a glucagon-like peptide 1 (GLP-1 ) modulator such as an agonist (e.g., exendin-3 and exendin-4), liraglutide, albiglutide, exenatide (Byetta®), albiglutide, lixisenatide, dulaglutide, semaglutide, NN-9924,TTP-054, a protein tyrosine phosphatase-1 B (PTP- 1 B) inhibitor (e.g., trodusquemine, hyrtiosal extract, and compounds disclosed by Zhang, S., et al., Drug Discovery Today, 12(9/10), 373-381 (2007)), SIRT-1 inhibitor (e.g., resveratrol, GSK2245840 or GSK184072), a dipeptidyl peptidease IV (DPP-IV) inhibitor (e.g., those in WO20051 16014, sitagliptin, vildagliptin, alogliptin, dutogliptin, linagliptin and saxagliptin), an insulin secreatagogue, a fatty acid oxidation inhibitor, an A2 antagonist, a c-jun amino-terminal kinase (JNK) inhibitor, glucokinase activators (GKa) such as those described in WO2010103437, WO2010103438, WO2010013161 , WO2007122482, TTP-399, TTP-355, TTP-547, AZD1656, ARRY403, MK-0599, TAK- 329, AZD5658 or GKM-001 , insulin, an insulin mimetic, a glycogen phosphorylase inhibitor (e.g. GSK1362885), a VPAC2 receptor agonist, SGLT2 inhibitors, such as those described in E.C. Chao et al. Nature Reviews Drug Discovery 9, 551 -559 (July 2010) including dapagliflozin, canagliflozin, empagliflozin, tofogliflozin (CSG452), ertugliflozin, ASP-1941 , THR1474, TS-071 , ISIS388626 and LX421 1 as well as those in WO2010023594, a glucagon receptor modulator such as those described in Demong, D.E. et al. Annual Reports in Medicinal Chemistry 2008, 43, 1 19-137, GPR1 19 modulators, particularly agonists, such as those described in WO2010140092, WO2010128425, WO2010128414, WO2010106457, Jones, R.M. et al. in Medicinal Chemistry 2009, 44, 149-170 (e.g. MBX-2982, GSK1292263, APD597 and PSN821 ), FGF21 derivatives or analogs such as those described in Kharitonenkov, A. et al. et al., Current Opinion in Investigational Drugs 2009, 10(4)359-364, TGR5 (also termed GPBAR1 ) receptor modulators, particularly agonists, such as those described in Zhong, M., Current Topics in Medicinal Chemistry, 2010, 10(4), 386-396 and INT777, GPR40 agonists, such as those described in Medina, J.C., Annual Reports in Medicinal Chemistry, 2008, 43, 75-85, including but not limited to TAK-875, GPR120 modulators, particularly agonists, high affinity nicotinic acid receptor (HM74A) activators, and SGLT1 inhibitors, such as GSK1614235. A further representative listing of anti-diabetic agents that can be combined with the compounds of the present invention can be found, for example, at page 28, line 35 through page 30, line 19 of WO201 100561 1 . Preferred anti-diabetic agents are metformin and DPP-IV inhibitors (e.g., sitagliptin, vildagliptin, alogliptin, dutogliptin, linagliptin and saxagliptin). Other antidiabetic agents could include inhibitors or modulators of carnitine palmitoyl transferase enzymes, inhibitors of fructose 1 ,6-diphosphatase, inhibitors of aldose reductase, mineralocorticoid receptor inhibitors, inhibitors of TORC2, inhibitors of CCR2 and/or CCR5, inhibitors of PKC isoforms (e.g. PKCa, ΡΚΟβ, PKCy), inhibitors of fatty acid synthetase, inhibitors of serine palmitoyl transferase, modulators of GPR81 , GPR39, GPR43, GPR41 , GPR105, Kv1.3, retinol binding protein 4, glucocorticoid receptor, somatostain receptors (e.g. SSTR1 , SSTR2, SSTR3 and SSTR5), inhibitors or modulators of PDHK2 or PDHK4, inhibitors of MAP4K4, modulators of IL1 family including ILI beta, modulators of RXRalpha. In addition suitable anti-diabetic agents include mechanisms listed by Carpino, P. A., Goodwin, B. Expert Opin. Ther. Pat, 2010, 20(12), 1627-51 .
Those skilled in the art will recognize that the compounds of this invention may also be used in conjunction with other cardiovascular or cerebrovascular treatments including PCI, stenting, drug eluting stents, stem cell therapy and medical devices such as implanted pacemakers, defibrillators, or cardiac resynchronization therapy. The compounds of the present invention may be used in combination with neuroinflammatory and neurodegenerative agents in mammals. Examples of additional neuroinflammatory and neurodegenerative agents include antidepressants, antipsychotics, anti-pain agents, anti-Alzheimer's agents, and anti-anxiety agents. Examples of particular classes of antidepressants that can be used in combination with the compounds of the invention include norepinephrine reuptake inhibitors, selective serotonin reuptake inhibitors (SSRIs), NK-1 receptor antagonists, monoamine oxidase inhibitors (MAOIs), reversible inhibitors of monoamine oxidase (RIMAs), serotonin and noradrenaline reuptake inhibitors (SNRIs), corticotropin releasing factor (CRF) antagonists, and atypical antidepressants. Suitable norepinephrine reuptake inhibitors include tertiary amine tricyclics and secondary amine tricyclics. Examples of suitable tertiary amine tricyclics and secondary amine tricyclics include amitriptyline, clomipramine, doxepin, imipramine, trimipramine, dothiepin, butriptyline, nortriptyline, protriptyline, amoxapine, desipramine and maprotiline. Examples of suitable SSRIs include fluoxetine, fluvoxamine, paroxetine, and sertraline. Examples of monoamine oxidase inhibitors include isocarboxazid, phenelzine, and tranylcyclopramine. Examples of suitable reversible inhibitors of monoamine oxidase include moclobemide. Examples of suitable SNRIs of use in the present invention include venlafaxine. Examples of suitable atypical anti-depressants include bupropion, lithium, trazodone and viloxazine. Examples of anti-Alzheimer's agents include NMDA receptor antagonists such as memantine; and cholinesterase inhibitors such as donepezil and galantamine. Examples of suitable classes of anti-anxiety agents that can be used in combination with the compounds of the invention include benzodiazepines and serotonin 1A receptor (5-HT1A) agonists, and CRF antagonists. Suitable benzodiazepines include alprazolam, chlordiazepoxide, clonazepam, chlorazepate, diazepam, lorazepam, oxazepam, and prazepam. Suitable 5-HT1A receptor agonists include buspirone and ipsapirone. Suitable CRF antagonists include verucerfont. Suitable atypical antipsychotics include paliperidone, ziprasidone, risperidone, aripiprazole, olanzapine, and quetiapine. Suitable nicotine acetylcholine agonists include CP-601927 and varenicline. Anti-pain agents include pregabalin, gabapentin, clonidine, neostigmine, baclofen, midazolam, ketamine and ziconotide.
Accordingly, in one embodiment, the pharmaceutical combination comprises a therapeutically effective amount of a composition comprising:
a first compound, the first compound being a compound of Formula I or a pharmaceutically acceptable salt thereof;
a second compound being selected from an approved drug or a clinical candidate useful for the treatment of infectious or inflammatory diseases; and
an optional pharmaceutically acceptable carrier, vehicle or diluent.
In particular, the invention provides for a pharmaceutical combination
comprising a therapeutically effective amount of a composition comprising:
a first compound, the first compound being a compound of Formula I or a pharmaceutically acceptable salt thereof; and a second compound, the second compound being selected from the group consisting of antibodies or small molecules which include but are not limited to those that block the action of specific cytokines such as TNFa, IL12 and/or IL23, or inhibitors of leukocyte recruitment such as modulators of S1 P receptors or integrin antagonists, or selective or non-selective inhibitors of the JAK kinases JAK1 , JAK2, JAK3 and/ or TYK2, inhibitors of leukocyte function such as PDE4 or SMAD7. In a specific embodiment, the invention is directed to a pharmaceutical composition of a compound of Formula I wherein the second compound is selected from
(a) an anti-TNFa agent selected from infliximab, adalimumab, golimumab, and certolizumab pegol;
(b) an anti-IL-12 and/or IL-23 agent selected from ustekinumab;
(c) a modulator of S1 P receptors selected from ozanimod;
(d) an integrin antagonist selected from vedolizumab, etrolizumab, and natalizumab;
(e) an inhibitor of JAK kinases selected from tofacitinib, filgotinib, PF-
04965842, PF-06651600, and PF-06263276;
(f) a PDE4 inhibitor selected from apremilast; or
(g) a SMAD7 antisense oligonucleotides selected from mongersen.
Inasmuch as it may be desirable to administer a combination of active compounds, for example, for the purpose of treating a particular disease or condition, it is within the scope of the present invention that two or more pharmaceutical compositions, at least one of which comprises a compound of the invention, may conveniently be combined in the form of a kit suitable for co-administration of the compositions. Representative kits include at least one compound of the present invention and a package insert or other labeling including directions.
General Synthetic Schemes
Compounds of the present invention can be prepared in accordance with the procedures outlined herein, from commercially available starting materials, compounds known in the literature, or readily prepared intermediates, by employing standard synthetic methods and procedures known to those skilled in the art. Standard synthetic methods and procedures for the preparation of organic molecules and functional group transformations and manipulations can be readily obtained from the relevant scientific literature or from standard textbooks in the field. It will be appreciated that where typical or preferred process conditions (i.e. , reaction temperatures, times, mole ratios of reactants, solvents, pressures, etc.) are given; other process conditions can also be used unless otherwise stated. Optimum reaction conditions can vary with the particular reactants or solvent used. Those skilled in the art will recognize that the nature and order of the synthetic steps presented can be varied for the purpose of optimizing the formation of the compounds described herein. The processes described herein can be monitored according to any suitable method known in the art. For example, product formation can be monitored by spectroscopic means, such as nuclear magnetic resonance spectroscopy (e.g., 1H or 13C), infrared spectroscopy, spectrophotometry (e.g., UV-visible), mass spectrometry, or by chromatography such as high-performance liquid chromatograpy (HPLC), gas chromatography (GC), gel-permeation chromatography (GPC), or thin layer chromatography (TLC).
The reactions or the processes described herein can be carried out in suitable solvents, which can be readily selected by one skilled in the art. Suitable solvents typically are substantially nonreactive with the reactants, intermediates, and/or products at the temperatures at which the reactions are carried out, i.e., temperatures that can range from the solvent's freezing temperature to the solvent's boiling temperature. A given reaction can be carried out in one solvent or a mixture of more than one solvent. Depending on the particular reaction step, suitable solvents for a particular reaction step can be selected.
The routes below, including those mentioned in the Examples and Preparations, illustrate methods of synthesizing compounds of formula I. The skilled person will appreciate that the compounds of the invention, and intermediates thereof, may be made by methods other than those specifically described herein. One skilled in the art could, therefore, adapt the methods described herein, by synthetic methods known in the art. In particular, suitable guides to synthesis, functional group interconversions, use of protecting groups, and the like, include, for example: "Comprehensive Organic Transformations" by RC Larock, VCH Publishers Inc. (1989); Advanced Organic Chemistry" by J. March, Wiley Interscience (1985); "Designing Organic Synthesis" by S Warren, Wiley Interscience (1978); "Organic Synthesis - The Disconnection Approach" by S Warren, Wiley Interscience (1982); "Guidebook to Organic Synthesis" by RK Mackie and DM Smith, Longman (1982); "Protective Groups in Organic Synthesis" by TW Greene and PGM Wuts, John Wiley and Sons, Inc. (1999); and "Protecting Groups" by PJ, Kocienski, Georg Thieme Verlag (1994); and any updated versions of said standard works.
In addition, the skilled person will appreciate that it may be necessary or desirable at any stage in the synthesis of compounds of the invention to protect one or more sensitive groups, so as to prevent undesirable side reactions. In particular, it may be necessary or desirable to protect amino or carboxylic acid groups. The protecting groups used in the preparation of the compounds of the invention may be used in conventional manner. See, for example, those described in 'Greene's Protective Groups in Organic Synthesis' by Theodora W Greene and Peter G M Wuts, third edition, (John Wiley and Sons, 1999), in particular chapters 7 ("Protection for the Amino Group") and 5 ("Protection for the Carboxyl Group"), incorporated herein by reference, which also describes methods for the removal of such groups. In the general synthetic methods below, unless otherwise specified, the substituents are as defined above with reference to the compounds of formula I above.
Where ratios of solvents are given, the ratios are by volume.
The skilled person will appreciate that the experimental conditions set forth in the schemes that follow are illustrative of suitable conditions for effecting the transformations shown, and that it may be necessary or desirable to vary the precise conditions employed for the preparation of compounds of formula (I).
The derivatives of formula (I), exemplified herein, can be prepared by the procedures described in the general methods presented below or by routine modifications thereof. The present invention also encompasses any one or more of these processes for preparing the derivatives of formula (I), in addition to any novel intermediates used therein. The person skilled in the art will appreciate that the following reactions may be heated thermally or under microwave irradiation.
It will be further appreciated that it may be necessary or desirable to carry out the transformations in a different order from that described in the schemes, or to modify one or more of the transformations, to provide the desired compound of the invention.
According to a first process, compounds of formula (I), wherein L is N, may be prepared from compounds of formulae (IV) and (V), as illustrated by Scheme 1.
Figure imgf000067_0001
Figure imgf000067_0002
Hal = halogen, typically, CI or F
Scheme 1
Compounds of formulae (a), (b) and (V) are commercially available or may be synthesized by those skilled in the art according to the literature or preparations described herein.
Compounds of formula (IV) may be prepared from an acyl chloride of formula (a) and the amine of formula (b) according to process step (i), an amide bond formation step. Preferred conditions include the reaction in the presence of an organic base, such as triethylamine in THF or EtOAc, at elevated temperatures (60 °C).
Compounds of formula (I) may be prepared from compounds of formula (IV) and the amine of formula (V) according to process step (ii). Preferred conditions include the reaction of the amine of formula (V) with the halo compound of formula (IV) in the presence of a suitable organic base, such as DIPEA in a suitable aprotic solvent such DMF or NMP at elevated temperature e.g. 140 °C.
Compounds of formula (IV) may alternatively be prepared from the acid of formula (VI) and the amine of formula (b) as illustrated in Scheme 2.
Figure imgf000068_0001
Hal = halogen, typically CI or F
Scheme 2
Compounds of formula (VI) are commercially available or may be synthesized by those skilled in the art according to the literature or preparations described herein. Compounds of formula (IV) may be prepared from compounds of formula (VI) according to process step (iii), an amide bond formation step with amines of formula (b), wherein Z defined herein elsewhere, mediated by a suitable combination of amide bond coupling agent and organic base. Typical conditions comprise HATU or HBTU with triethylamine or DIPEA in DCM, DMF or DMA at room or elevated temperatures (e.g. about 80 °C), or using propylphosphonic anhydride (50% in EtOAc) in 2-MeTHF, THF or toluene with pyridine or DIPEA at elevated temperature e.g. about 60 °C. Alternative conditions comprise treatment with phosphoryl trichloride in pyridine or with Ghosez's reagent in acetonitrile or dichloromethane at between -10 °C and reflux.
According to a second process, compounds of formula (I) may be prepared in an alternative sequence from compounds of formulae (b) and (VIII) as illustrated by Scheme 3.
Figure imgf000069_0001
(V)
Figure imgf000069_0002
(VIII)
halogen, typically chloro or fluoro.
Compounds of formula (VIII) may be prepared from compounds of formula (VII), wherein Rx is C1 -C4, typically ethyl or methyl, and compounds of formula (V), wherein the amine of (V) is previously described as "L", according to process step (ii), as described in scheme 1 . Preferred conditions include the reaction of the amine and halide in the presence of an organic base such as triethylamine or DIPEA in a solvent such as NMP under microwave irradiation at elevated temperatures eg 140 °C for up to 1 hr. Alternatively, the reaction of the amine and halide is conducted in the presence of an organic base, preferably DIPEA, in a suitable solvent, such as 2-propanol, dioxane, or THF, (optionally with DMSO as a co-solvent), at elevated temperatures, typically between 60 and 80 °C.
Compounds of formula (I) may be prepared from compounds of formula (VIII) by treatment with an amine of formula (b) according to process step (iv). Preferred conditions are reaction in the presence of suitable coupling agent, typically TBD in a suitable aprotic solvent, such as DMF or NMP, at elevated temperature e.g. 50 °C.
According to a third process, compounds of formula (I) may be prepared in an alternative sequence from compounds of formulae (IX) and (b) as illustrated by Scheme 4.
Figure imgf000070_0001
(VIII) (IX)
Figure imgf000070_0002
(iii) G V|_ N R4 Z
(I)
Hal = halogen, typically CI or F
Scheme 4
Compounds of formula (IX), wherein L is previously described herein, may be prepared from the ester of formula (VIII), wherein Rx is C1 -C4 alkyl, typically ethyl or methyl, according to process step (v), a hydrolysis step mediated by an inorganic base. Preferred conditions include aqueous lithium or sodium hydroxide in methanol or ethanol optionally with THF as a co-solvent between room temperature and 60 °C.
Compounds of formula (I) may be prepared from compounds of formula (IX) according to process step (iii), an amide bond formation step with an amine of formula (b), wherein Z is previously described herein, mediated by a suitable combination of amide bond coupling agent and organic base, as described in Scheme 2.
Preferred conditions comprise HATU with triethylamine or DIPEA at elevated temperatures (e.g. about 60 °C), or using propylphosphonic anhydride in THF with DIPEA and triethylamine at an elevated temperature (e.g.about 60 °C).
According to a fourth process, compounds of formula (I) wherein L is O may be prepared from compounds of formula (IV), wherein Z is previously described herein, and an alcohol of formula (X) as illustrated by scheme 5.
Figure imgf000071_0001
Compounds of formula (I) may be prepared from the compounds of formulae (IV) and (X) according to process step (vi). Typical conditions include treatment with a suitable non-nucleophilic base, such as LiHMDS in a suitable solvent such as DMF at temperatures below room tem erature (e.g. 0 °C).
Figure imgf000071_0002
Hal = halogen, typically CI or F
Scheme 6
Compounds of formula (XI) may be prepared from compounds of formula (IV), wherein Z is previously described herein, according to process step (vii), a substitution nucleophilic aromatic with an ammonia source as described in Scheme 6. A preferred source of ammonia is an aqueous solution of an ammonium salt such as ammonium hydroxide. This step could also be performed in a different temperature range, typically a temperature above 80 °C and, preferentially, under microwave irradiations above 120 °C. Compounds of formula (I) may be prepared from compounds of formula (XI) according to process step (iii), an amide bond formation step with an acid of formula (XII), mediated by a suitable combination of amide bond coupling agent and organic base, as described in Scheme 2.
It will be apparent to those skilled in the art that many of the intermediates are commercially avaialbe and that where intermediates are not commercially available, numerous synthetic methods are available from the synthetic literature from which one skilled in the art would be able prepare such intermediates. EXPERIMENTAL PROCEDURES AND WORKING EXAMPLES
The following illustrate the synthesis of various compounds of the present invention. Additional compounds within the scope of this invention may be prepared using the methods illustrated in these Examples, either alone or in combination with techniques generally known in the art.
It will be understood that the intermediate compounds of the invention depicted above are not limited to the particular enantiomer shown, but also include all stereoisomers and mixtures thereof. It will also be understood that compounds of
Formula I can include intermediates of compounds of Formula I.
Abbreviations
In the Examples and Preparations that are set out below, and in the aforementioned Schemes, the following abbreviations, definitions and analytical procedures may be referred to. Other abbreviations common in the art are also used. Standard lUPAC nomenclature has been used.
AcOH is acetic acid;
AgF is silver fluoride;
AIBN is azobisisobutyronitrile;
Ar is argon;
aq is aqueous;
Bn is benzyl;
Boc is tert-butoxycarbonyl;
Boc2O is di-fe/f-butyl dicarbonate;
br is broad;
tBu is tert-butyl;
tBuOH is fe/f-butanol;
n-BuLi is n-butyl lithium; °C is degrees celcius;
CBz-CI is benzyl chloroform ate;
CDC is deutero-chloroform;
CS2CO3 is cesium carbonate;
CsF is cesium fluoride;
δ is chemical shift;
d is doublet;
DCM is dichloromethane; methylene chloride;
DBU is 1 ,8-Diazabicyclo[5.4.0]undec-7-ene
DIPEA is N-ethyldiisopropylamine, N,N-diisopropylethylamine;
DMA is N,N-dimethylacetamide;
DMF is N,N-dimethylformamide;
DMSO is dimethyl sulphoxide;
DPPA is diphenyl phosphoryl azide;
EDC = N-(3-Dimethylaminopropyl)-N'-ethylcarbodiimide hydrochloride and HOBt
= 1 -Hydroxybenzotriazole hydrate are amide coupling reagents
Et2O is diethyl ether;
EtOAc is ethyl acetate;
EtOH is ethanol;
Et3N is triethylamine;
g is gram;
HATU is 1 -[Bis(dimethylamino)methylene]- H-1 ,2,3-triazolo[4,5-b]pyridinium 3- oxid hexafluorophosphate;
HBTU is N,N,N',N'-tetramethyl-O-(1 H-benzotriazol-1 -yl)uronium
hexafluorophosphate;
HCI is hydrochloric acid;
HCO2H is formic acid;
HPLC is high pressure liquid chromatography;
H2O is water;
H2O2 is hydrogen peroxide;
Hr is hour, hrs are hours;
IMS is industrial methylated spirit;
K2CO3 is potassium carbonate;
KHSO4 is potassium hydrogen sulphate;
KOAc is potassium acetate; L is litre;
LCMS is liquid chromatography mass spectrometry; LiALH4 is lithium aluminium hydride;
LiHMDS is lithium bis(trimethylsilyl)amide;
UOH. H2O is lithium hydroxide monohydrate;
Li-Selectride® is lithium tri-sec-butylborohydride; m is multiplet;
M is molar;
MeCN is acetonitrile;
MeMgBr is methyl magnesium bromide;
MeOH is methanol;
2-MeTHF is 2-methyl tetrahydrofuran
mg is milligram;
MgS04 is magnesium sulphate;
MHz is mega Hertz;
min is minutes;
mL is millilitre;
mmol is millimole;
mol is mole;
MS m/z is mass spectrum peak;
MsCI is mesyl chloride
NaCN is sodium cyanide;
NaBH4 is sodium borohydride;
Na2C03 is sodium carbonate;
NaH is sodium hydride;
NaHS04 is sodium hydrogen sulfate;
NaOH is sodium hydroxide;
Na2S04 is sodium sulphate;
NBS is N-bromosuccinimide;
NH3 is ammonia;
NH4CI is ammonium chloride;
NH4HC03 is ammonium hydrogen carbonate;
NH2NH2.H20 is hydrazine hydrate;
NH2OH.HCI is hydroxylamine hydrochloride;
NH4OH is ammonium hydroxide; NH OAc is ammonium acetate;
NMP is 1 -methyl-2-pyrrolidinone;
NMR is nuclear magnetic resonance;
Pd/C is palladium on carbon;
Pd(dppf)CI2 is [1 , 1 '-bis(diphenylphosphino)ferrocene]dichloropalladium(ll);
Pd(OH)2 is palladium hydroxide;
Pd(OAc)2 is palladium acetate;
Pet. Ether is petroleum ether;
pH is power of hydrogen;
ppm is parts per million;
Pt02 is platinum (IV) oxide;
q is quartet;
rt is room temperature;
RT is retention time;
s is singlet;
sat. is saturated
SCX is strong cation exchange;
SFC is supercritical fluid chromatography
t is triplet;
T3P is propylphosphonic anhydride
TBAF is tert-butyl ammonium fluoride;
TBD is 1 ,5,7-triazabicyclo[4.4.0]dec-5-ene;
TBME is tert-butyl dimethyl ether;
TFA is trifluoroacetic acid;
THF is tetrahydrofuran;
Ti(OiPr)4 is titanium (IV) propoxide;
TPTU is 2-(2-pyridon-1 -yl)-1 , 1 ,3,3-tetramethyluronium tetrafluoroborate, an amide coupling agent.
μΙ_ is microlitre
μιηοΙ is micromole
1H and 19F Nuclear magnetic resonance (NMR) spectra were in all cases consistent with the proposed structures. Characteristic chemical shifts (δ) are given in parts-per-million downfield from tetramethylsilane (for H-NMR) and upfield from trichloro-fluoro-methane (for 19F NMR) using conventional abbreviations for designation of major peaks: e.g. s, singlet; d, doublet; t, triplet; q, quartet; m, multiplet; br, broad. The following abbreviations have been used for common solvents: CDCI3, deuterochloroform; DMSO-d6, deuterodimethylsulphoxide; and MeOH-d4,
deuteromethanol. Where appropriate, tautomers may be recorded within the NMR data; and some exchangeable protons may not be visible.
Mass spectra, MS (m/z), were recorded using either electrospray ionisation (ESI) or atmospheric pressure chemical ionisation (APCI).
Where relevant, and unless otherwise stated, the m/z data provided are for isotopes 19F, 35CI, 79Br and 127l.
Wherein preparative TLC or silica gel chromatography have been used, one skilled in the art may choose any combination of solvents to purify the desired compound.
In general, reactions were followed by thin layer chromatography and / or liquid chromatography-mass spectrometry, and subjected to work-up when appropriate. It will be recognized by one skilled in the art that purifications may vary between experiments: in general, sorbents, solvents and the solvent ratios used for eluants/gradients were chosen to provide appropriate Rfs or retention times. It will also be recognized by one skilled in the art that HPLC purifications may be effected in a variety of ways, including the use of normal stationary phases, reverse stationary phases, chiral stationary phases, and supercritical eluants. The appropriate choices of conditions for
chromatographic and HPLC purifications will be discerned by one skilled in the art.
Achiral analytical HPLC conditions
The following methods were used to characterize examples 1 to 129 from library protocols.
Figure imgf000076_0001
Time 4.20 mins 100% B Time 4.30 mins 1 % B
Time 4.21 mins 5% B Time 4.70 mins 1 % B
Time 4.70 mins 5% B Flow rate 0.8 ml/min
Flow rate 0.8 ml/min Injection volume 2 μΙ
Injection volume 2 μΙ
Agilent 1200 HPLC/1956 MSD/SEDEX 75
Agilent 1200 HPLC/1956 MSD/SEDEX 75 ELSD
ELSD Ionization Mode API-ES
Ionization Mode API-ES Polarity Positive
Polarity Positive
Method AB00
Column Xbridge C18 2.1 x50mm 5 m
Temperature 40 °C
Mobile Phase A 0.0375% TFA in water
Mobile Phase B 0.01875% TFA in MeCN
Gradient - Initial 0% B
Time 0.00 mins 0% B
Time 1.00 mins 5% B
Time 4.00 mins 70% B
Time 4.10 mins 0% B
Time 4.70 mins 0% B
Flow rate 0.8 ml/min
Injection volume 2 μΙ
Agilent 1200 HPLC/1956 MSD/SEDEX 75
ELSD
Ionization Mode API-ES
Polarity Positive
Achiral preparative HPLC conditions
The following methods were used to purify examples 1 to 129 from library protocols. Anyone skilled in the art will apply an appropriate gradient of solvent to afford the title compounds in adequate purity. AD01 AD02
Agela Durashell C18 250x21 .2mm*5um Agela Durashell C18 250x21 .2mm*5um column column
MeCN-Water(0.225% TFA) MeCN-NH4OH (pH10)
AD03 AD04
Agela Durashell C18 150x25mm*5um Agela Durashell C18 150x25mm*5um column column
MeCN-Water(0.225% TFA) MeCN-NH4OH (pH10)
PG01 PG02
Phenomenex Gemini C18 Phenomenex Gemini C18 250x21 .2mm*10um 250x21 .2mm*10um
MeCN-Water(0.225% TFA) MeCN-NH4OH (pH10)
WX01
Waters Xbridge Prep OBD C18
150x30mm*5um
MeCN-NH4OH (pH10)
Chiral analytical SFC Conditions
Method CA-A: column: Lux Cellulose-2 150x4.6mm I.D., 3pm; mobile phase: 40% EtOH (0.05% DEA) in CO2; flow rate of 2.5 mL/min at 40 °C.
Method CA-B: column: Chiralcel OD-3 150x4.6mm I.D., 3pm; mobile phase:
EtOH (0.05%DEA) in CO2 (from 5% to 40% in 5 min); flow rate: 2.5 mL/min at 35 °C.
Method CA-C: column: Chiralpak AD-3 50x4.6mm I.D., 3pm; mobile phase: 40% of EtOH (0.05% DEA) in CO2; flow rate: 4 mL/min at 40 °C
Method CA-D: column: Chiralpak AD-3 150x4.6mm I.D., 3pm; mobile phase: 40% of iso-propanol (0.05% DEA) in CO2; flow rate of 2.5 mL/min
Method CA-E: column: Chiralpak AS-3 100x4.6mm I.D., 3pm; mobile phase: EtOH (0.05% DEA) in CO2 (from 5% to 40% in 4.5 min); flow rate: 2.8 mL/min
Method CA-F: column: Chiralpak AD-3 150x4.6mm I.D., 3pm; mobile phase: iso-propanol (0.05% DEA) in CO2 (from 5% to 40% in 5.5 min); flow rate: 2.5 mL/min at 35 °C
Method CA-G: column: Chiralpak AD-3 150x4.6mm I.D., 3pm; mobile phase: EtOH (0.05% DEA) in CO2 (from 5% to 40% in 5.0 min); flow rate: 2.5 mL/min at 35 °C Chiral preparative SFC conditions
Method CP-A: C2 250mm*30mm, 10μηι column, eluting with 55% EtOH (0.1 %NH3 H20) in C02 at a flow rate of 80 mL/min.
Method CP-B: OD 250mm*30mm, 10pm column, eluting with 40% EtOH (0.1 % ΝΗ3Ή2Ο) in CO2 at a flow rate of 80 mL/min.
Method CP-C: AD 250mm*30mm, 10pm column, eluting with 50% of EtOH (0.05% ΝΗ3Ή2Ο) in CO2 at a flow rate of 80 mL/min.
Method CP-D: AD 250mm*30mm, 5 m column, eluting with 40% of iso-propanol (0.05% DEA) in CO2 at a flow rate of 50 mL/min.
Method CP-E: AS 250mm*30mm, 10pm column, eluting with 20% EtOH (0.05%
DEA) in CO2 at a flow rate of 60 mL/min.
Method CP-F: OJ 250mm*30mm, 5pm column, eluting with 25% MeOH (0.05% DEA) in CO2 at a flow rate of 60 mL/min.
Method CP-G: AY 250mm*30mm, 10 m column, eluting with 45% iso-propanol (0.1 % ΝΗ3Ή2Ο) in CO2 at flow rate of 80 mL/min.
Experimental Procedures
Experiments were generally carried out under inert atmosphere (nitrogen or argon), particularly in cases where oxygen- or moisture-sensitive reagents or intermediates were employed. Commercial solvents and reagents were generally used without further purification, including anhydrous solvents where appropriate (generally Sure-Seal™ products from the Aldrich Chemical Company, Milwaukee, Wisconsin). Products were generally dried under vacuum before being carried on to further reactions or submitted for biological testing. Mass spectrometry data is reported from either liquid chromatography-mass spectrometry (LCMS), atmospheric pressure chemical ionization (APCI) or gas chromatography-mass spectrometry (GCMS) instrumentation. Chemical shifts for nuclear magnetic resonance (NMR) data are expressed in parts per million (ppm, δ) referenced to residual peaks from the deuterated solvents employed.
For syntheses referencing procedures in other Examples or Methods, reaction conditions (length of reaction and temperature) may vary. In general, reactions were followed by thin layer chromatography and / or liquid chromatography-mass spectrometry, and subjected to work-up when appropriate. It will be recognized by one skilled in the art that purifications may vary between experiments: in general, sorbents, solvents and the solvent ratios used for eluants/gradients were chosen to provide appropriate Rfs or retention times. It will also be recognized by one skilled in the art that HPLC purifications may be effected in a variety of ways, including the use of normal stationary phases, reverse stationary phases, chiral stationary phases, and supercritical eluants. The appropriate choices of conditions for chromatographic and HPLC purifications will be discerned by one skilled in the art.
The following Preparations describe the preparation of certain intermediates used in the Methods and Examples that follow. The following Preparations, Methods and Examples are intended to illustrate particular embodiments of the invention and preparations thereto and are not intended to limit the specification, including the claims, in any manner. Unless noted otherwise, all reactants were obtained commercially. Furthermore, some preparations, such as 59, 60 and 73, describe a synthetic route to a free base, while the salt form is the one actually characterized. One skilled in the art will appreciate that isolation of a salt could be performed by mixing the free base with corresponding acid in an appropriate solvent, according to standard procedure that can be found in the literature.
Vanin-1 Preparations
Preparation 1
2-[(Pyridin-3-ylmethyl)aminolpyrimidine-5-carboxylic acid
Figure imgf000080_0001
LiOH H2O (1 .22 g, 29.0 mmol) was added to a solution of ethyl 2-[(pyridin-3- ylmethyl)amino]pyrimidine-5-carboxylate (Preparation 7, 2.50 g, 9.68 mmol) in MeOH/H2O (1 : 1 , 20 ml_) and the reaction stirred at 20 °C for 16 hrs. The mixture was concentrated in vacuo and the residue acidified to pH 4 by the dropwise addition of 1 N HCI. The resulting solid was filtered off and dried under vacuum to afford the title compound as a brown solid, 2.00 g, 89%. 1H NMR (400 MHz, DMSO-d6): δ 4.59 (d, 2H), 7.34 (dd, 1 H), 7.71 (d, 1 H), 8.42-8.60 (m, 3H), 8.73 (s, 2H). LCMS m/z = 231 [M+H]+
Preparation 2
2-[(Pyrimidin-5-ylmethyl)aminolpyrimidine-5-carboxylic acid
Figure imgf000081_0001
UOH.H2O (971 mg, 23.1 mmol) was added to a suspension of ethyl 2- [(pynmidin-5-ylmethyl)amino]pynmidine-5-carboxylate (Preparation 8, 1 .2 g, 4.63 mmol) in EtOH/THF/H20 (70 ml_, 4:2: 1 ) and the reaction stirred at 60 °C for 2 hrs. The cooled reaction mixture was evaporated under reduced pressure. The residue was adjusted to pH 3 using 1 N HCI solution and the resulting suspension stirred at 20 °C for 10 min. The solid was collected by filtration and washed with water (5 ml_ x2). The solid was co-evaporated with toluene three times to afford the title compound as a pale solid, 850 mg, 79%. 1H NMR (400 MHz, DMSO-d6): δ 4.59 (d, 2H), 8.57 (dd, 1 H), 8.71 -8.78 (m, 3H), 9.07 (s, 1 H), 12.85 (br s, 1 H). LCMS m/z = 232 [M+H]+
Preparation 3
2-[(Pyrazin-5-ylmethyl)aminolpyrimidine-5-carboxylic acid
Figure imgf000081_0002
A solution of ethyl 2-[(pyrazin-5-ylmethyl)amino]pyrimidine-5-carboxylate (Preparation 9, 1710 mg, 6.60 mmol) in THF (13.2 ml_) and MeOH (6.6 ml_) was treated with a solution of LiOH.H20 (830 mg, 19.8 mmol) in water (13.2 ml_) and the resulting solution stirred at rt for 2 hrs. 1 N HCI (35 ml_) was added followed by sat. NH4CI solution and the mixture was concentrated under reduced pressure. The resulting aqueous layer was filtered and the resulting brown solid dried under reduced pressure to afford the title compound, 1 .12 g, 73%. 1H NMR (400 MHz, DMSO-d6): δ 4.72 (d, 2H), 8.50-8.61 (m, 4H), 8.72 (d, 1 H), 12.79 (br s, 1 H). LCMS m/z = 232 [M+H]+
Preparation 4
2-{[(1 S)-(1 -(Pyrazin-2-yl)ethyl1amino)pyrimidine-5-carboxylic acid
Figure imgf000081_0003
A solution of NaOH (838 mg, 21 .0 mmol) in water (10 mL) was added drop wise over 30 mins. to a mixture of ethyl 2-{[(1 S)-1 -(pyrazin-2-yl)ethyl]amino}pyrimidine-5-carboxylate (Preparation 10, 2.29g, 8.38 mmol) in THF (10 mL), MeOH (10 mL) and water (5 mL) and the reaction was then stirred for 20 mins. The mixture was concentrated in vacuo, and 6M HCI (3.5 mL) was added to lower the pH to ~2. The resulting solid was filtered off, washed with water and dried to afford the title compound as a pale yellow solid, 1 .7 g, 82%. 1H NMR (400 MHz, DMSO-d6): δ 1 .55 (d, 3H), 5.24-5.32 (m, 1 H), 8.50-8.59 (m, 3H), 8.63-8.76 (m, 3H), 12.75 (s, 1 H). LCMS m/z = 246 [M+H]+
Preparation 5 2-{[1 -(Pyrimidin-5-yl)cvclopropyl1amino)pyrimidine-5-carboxylic acid
Figure imgf000082_0001
NaOH (1 .68 g, 42.1 mmol) was added to a yellow solution of ethyl 2-{[1 -(pyrimidin-5- yl)cyclopropyl]amino}pyrimidine-5-carboxylate (Preparation 11 , 2.00 g, 7.01 mmol) in MeOH/H2O/THF (80.0 mL/ 15.0 mL/ 15.0 mL) and the resulting suspension stirred at 30 °C for 18 hrs. The reaction mixture was acidified to pH 2 using aq.HCI (2M). The solvent was removed under reduced pressure to give a yellow solid which was triturated with MeOH/THF (80 mL/40 mL), the solid was filtered off and the filtrate was concentrated in vacuo to provide the title compound as a grey solid, in quantitative yield. 1H NMR (400 MHz, DMSO-de) : δ 1 .30-1 .35 (m, 2H), 1 .46-1 .50 (m, 2H), 8.61 (s, 2H), 8.70-8.76 (m, 2H), 8.93 (s, 1 H), 9.00 (s, 1 H). LCMS m/z = 258 [M+H]+
Preparation 6
2-{[1 -(Pyrimidin-5-yl)ethyl1amino)pyrimidine-5-carboxylic acid
Figure imgf000082_0002
LiOH.H2O (691 mg, 16.5 mmol) was added to a yellow suspension of ethyl 2-{[1 - (pyrimidin-5-yl)ethyl]amino}pyrimidine-5-carboxylate (Preparation 12, 900 mg, 3.29 mmol) in EtOH/THF/H2O (70 mL, 4:2: 1 ) and the reaction was stirred at 60 °C for 2 hrs. The cooled reaction was concentrated in vacuo, and the residue was adjusted to pH 3 with 1 N HCI solution. The resulting suspension was stirred at 20 °C for 10 min, the solid collected by filtration and washed with water (5 ml_ x2). The solid was co-evaporated with toluene (3x) to give the title compound as a yellow solid, 170 mg, 21 %. The filtrate was evaporated under reduced pressure and the resulting yellow oil purified by preparative HPLC using a Phenomenex Gemini C18 250*50 10μ column eluting with 0.225% aq. TFA in MeCN at a flow rate of 30 mL/min to afford additional product as a pale yellow solid, 195 mg, 24% yield. 1HNMR (400 MHz, CDCI3): δ 1 .58 (d, 3H), 5.21 - 5.27 (m, 1 H), 8.64-8.72 (m, 3H), 8.76 (s, 2H), 8.84 (s, 1 H), 9.07 (s, 1 H). LCMS m/z = 246 [M+H]+ Preparation 7
Ethyl 2-[(pyridin-3-ylmethyl)aminolpyrimidine-5-carboxylate
Figure imgf000083_0001
DIPEA (7.27 g, 56.3 mmol) was added drop wise to a stirred solution of ethyl 2- chloropyrimidine-5-carboxylate (3.50 g, 218.8 mmol) and 3-pyridinemethanamine (2.03 g, 18.8 mmol) in dry THF (30 ml_) and the reaction stirred at 60 °C for 16 hrs. The solvent was removed under reduced pressure and water (10 ml_) was added. The mixture was extracted with EtOAc (40 ml_ x3) and the combined organic extracts dried (Na2S04), filtered and concentrated in vacuo. The crude product was purified by column chromatography on silica gel eluting with Pet. EthenEtOAc (100:0 to 7:93) to afford the title compound as a brown solid, 3.50 g, 72%. 1H NMR (400 MHz, CDCI3): δ 1 .38 (t, 3H), 4.36 (q, 2H), 4.74 (d, 2H), 6.03 (br s, 1 H), 7.29 (d, 1 H), 7.68 (d, 1 H), 8.55 (d, 1 H), 8.63 (d, 1 H), 8.85 (br s, 2H). LCMS m/z = 259 [M+H]+
Preparation 8
Ethyl 2-[(Pyrimidin-5-ylmethyl)aminolpyrimidine-5-carboxylate
Figure imgf000083_0002
A yellow solution of ethyl 2-chloropyrimidine-5-carboxylate (700 mg, 3.75 mmol), 5- pyrimidinemethanamine (450 mg, 4.13 mmol) and DIPEA (2420 mg, 18.8 mmol) in THF (20 ml_) was stirred at 60 °C for 16 hrs. The cooled reaction was concentrated in vacuo to give a yellow oil. The crude product was purified by column chromatography on silica gel eluting with DCM: MeOH (100:0 to 88: 12) to afford the title compound, as a yellow solid, 933 mg, 96%. 1H NMR (400 MHz, DMSO-d6): δ 1 .29 (t, 3H), 4.26 (q, 2H), 4.61 (d, 2H), 8.66 (br t, 1 H), 8.77 (s, 4H), 9.08 (s, 1 H). LCMS m/z = 260 [M+H]+
Preparation 9
Ethyl 2-[(pyrazin-5-ylmethyl)aminolpyrimidine-5-carboxylate
Figure imgf000084_0001
2-Aminomethylpyrazine (3.51 g, 32.2 mmol) was added to a solution of ethyl 2- chloropyrimidine-5-carboxylate (6 g, 32.16 mmol) and DIPEA (5.4 g, 41 .8 mmol) in 2- propanol (20 ml_) and the reaction mixture was heated under reflux for 16 hrs. The cooled reaction mixture was concentrated in vacuo and the residue purified by column chromatography on silica gel eluting with EtOAc: petroleum ether (0: 100 to 60:40) to afford the title compound as a yellow solid, 7.2 g, 86%. 1H NMR (400 MHz, CDCIs) : δ 1 .37 (t, 3H), 4.35 (q, 2H), 4.86 (s, 2H), 6.71 (br s, 1 H), 8.50 (s, 1 H), 8.55 (s, 1 H), 8.58 (s, 1 H), 8.86 (br s, 2H). LCMS m/z = 260 [M+H]+
Preparation 10
Ethyl 2-{[(1 S)-1 -(pyrazin-2-yl)ethyl1amino)pyrimidine-5-carboxylate
Figure imgf000084_0002
A solution of 1 -(pyrazin-2-yl)ethanamine (469.0 g, 3810 mmol) in 2-propanol (1 .9
L) was placed in a 5-L reactor equipped with a mechanical stirrer (glass rod, teflon paddle) and internal thermometer under N2. Ethyl 2-chloropyrimidine-5-carboxylate (71 1 g, 3810 mmol) was added as a solid with stirring followed by DIPEA (640 g, 4950 mmol). The resulting solution was gradually warmed to 88 °C, stirred for 7 hrs then allowed to cool. The content of the tank was transferred into a 6-L erlen meyer flask and the tank rinsed with 2-propanol. The solution was concentrated in vacuo to remove approximately half of the volume. The content of the erlenmeyer was transferred into a 10-L tank equipped with a jacket, a short-path distillation set-up connected to a 1 L flask and a mechanical stirrer. The erlenmeyer and the flask were rinsed with water until all the solids have been transferred. The resulting suspension was stirred at 50 °C under vacuum for 5 hrs and the mixture then allowed to cool to rt. Water (2 L) was added, the mixture stirred for 2 hrs then the solid was filtered off, washing through with water (500 ml_). The solid was dried in vacuo. The filtrate was concentrated in vacuo and the resulting solid filtered off and dried to provide additional product. This was purified by chiral SFC separation, using a Chiralcel IC-H 50 x 250 column, eluting with 65:35 CO2: MeCN at a flow rate of 250 mL/min, wavelength 215 nm to afford the title compound as a pale orange oil that crystallized on standing, 441 g. RT = 1 .38 min; 1H NMR (400 MHz, CDCI3): δ 1.37 (t, 3H), 1.63 (d, 3H), 4.35 (q, 2H), 5.42 (quint., 1 H), 6.58 (d, 1 H), 8.49 (d, 1 H), 8.54 (dd, 1 H), 8.65 (d, 1 H), 8.85 (br s, 2H). LCMS m/z = 274 [M+H]+
Further elution provided the second enantiomer as an orange solid, 487 g.
Preparation 11 Ethyl 2-{[1 -(pyrimidin-5-yl)cvclopropyl1amino)pyrimidine-5-carboxylate
Figure imgf000085_0001
DIPEA (17.3 ml_, 99.5 mmol) was added in one portion to a yellow suspension of 1 -(pyrimidin-5-yl)cyclopropanamine hydrochloride (Preparation 26, 4.87 g, 19.9 mmol) in THF (120 ml_) and the solution was stirred at 45 °C for 15 min. Ethyl 2- chloropyrimidine-5-carboxylate (3.71 g, 19.9 mmol) was added in one portion and the resulting yellow suspension was heated at 60 °C for 18 hrs. The mixture was cooled to 18 °C, the resulting solid filtered off and the filtrate concentrated in vacuo to give a yellow oil. The crude product was purified by column chromatography on silica gel eluting with EtOAc: Pet. Ether (50:50 to 90: 10) to afford the title compound as a yellow solid, 3.31 g, 58%. 1H NMR (400 MHz, MeOD-d4): δ 1 .35 (t, 3H), 1 .40-1 .45 (m, 2H), 1 .47-1 .52 (m, 2H), 4.32 (q, 2H), 8.69 (s, 2H), 8.76-8.84 (m, 2H), 8.96 (s, 1 H). LCMS m/z = 286 [M+H]+
Preparation 12
Ethyl 2-{[1 -(pyrimidin-5-yl)ethyl1amino)pyrimidine-5-carboxylate
Figure imgf000086_0001
A yellow suspension of ethyl 2-chloropyrimidine-5-carboxylate (600 mg, 3.22 mmol), 1 -(5-pyrimidin-yl)ethanamine (436 mg, 3.54 mmol) and DIPEA (2.08g, 16.1 mmol) in THF (20 mL) and DMSO (3 mL) was stirred at 60 °C for 16 hrs. The cooled reaction was concentrated in vacuo to give a yellow oil. The residue was partitioned between DCM (20 mL) and water (10 mL), the layers were separated and the aqueous was extracted with DCM (35 mL x2). The combined organic layers were washed with water (10 mL x2), dried (Na2S04), filtered and concentrated under reduced pressure to give a yellow oil. The crude product was purified by column chromatography on silica gel eluting with DCM: MeOH (100:0 to 88: 12) to afford the title compound as a yellow oil, 88% yield. 1H NMR (400 MHz, CDCI3) : δ 1 .36 (t, 3H), 1 .67 (d, 3H), 4.34 (q, 2H), 5.21 -5.29 (m, 1 H), 6.06 (br d, 1 H), 8.77 (s, 2H), 8.82 (s, 2H), 9.13 (s, 1 H). LCMS m/z = 274 [M+H]+
Preparation 13 Ethyl 2-{[(6-methylpyridin-3-yl)methyl1amino)pyrimidine-5-carboxylate
Figure imgf000086_0002
6-Methyl-3-pyridinemethanamine (1 .0 g, 8.19 mmol) was added to a solution of ethyl 2-chloropyrimidine-5-carboxylate (1 .53 g, 8.19 mmol) and DIPEA (1 .59 g, 12.3 mmol) in 2-propanol (8 mL) and the resulting mixture was heated under reflux for 8 hrs. The cooled mixture was concentrated under reduced pressure and the residue purified by column chromatography on silica gel eluting with EtOAc: Pet. Ether (0: 100 to 30:70) to afford the title compound as a yellow solid, 1 .5 g, 67%. 1H NMR (400 MHz, CDCI3) : δ 1 .37 (t, 3H), 2.55 (s, 3H), 4.35 (q, 2H), 4.67 (d, 2H), 7.12 (d, 1 H), 7.56 (dd, 1 H), 8.45 (d, 1 H). 8.85 (br s, 2H). LCMS m/z =273 [M+H]+ Preparation 14
Ethyl 2-{[1 -(pyrazin-2-yl)cvclobutyl1amino)pyrimidine-5-carboxylate
Figure imgf000087_0001
Ethyl 2-fluoropyrimidine-5-carboxylate (Preparation 21 , 1 .7 g, 9.99 mmol) was added to a solution of 1 -(pyrazin-2-yl)cyclobutanamine (Preparation 42, 1 .7 g, 1 1 .39 mmol) and DIPEA (2.58 g, 20.0 mmol) in dioxane (60 mL) under N2 and the reaction stirred at 80 °C for 16 hrs. The mixture was concentrated in vacuo and the residue purified by column chromatography on silica gel eluting with pet. ether: EtOAc (100:0 to 35:65) to provide the title compound as a yellow oil, 2.1 g, 70%. 1H NMR (400 MHz, CDCIs) : δ 1.33 (t, 3H), 2.06-2.18 (m, 1 H), 2.19-2.25 (m, 1 H), 2.48-2.52 (m, 2H), 2.86- 2.91 (m, 2H), 4.31 (q, 2H), 6.53 (br s, 1 H), 8.41 (s, 1 H), 8.55 (s, 1 H), 8.73-8.87 (m, 3H). LCMS m/z = 300 [M+H]+
Preparation 15
Ethyl 2-{[2-(pyrazin-2-yl)propan-2-yl1amino)pyrimidine-5-carboxylate
Figure imgf000087_0002
Ethyl 2-fluoropyrimidine-5-carboxylate (Preparation 21 , 1 .4 g, 8.23 mmol) was added to a solution of 2-pyrazin-2-ylpropan-2-amine (Commercial, 1 .69 g, 12.3 mmol) and DIPEA (2.13 g, 16.5 mmol) in dioxane (25 mL) under N2 and the reaction stirred at 80 °C for 16 hrs. The mixture was concentrated in vacuo and the residue purified by column chromatography on silica gel eluting with pet. ether: EtOAc (100:0 to 60:40) to provide the title compound, 1 .2 g, 51 %. 1HNMR (400MHz, CDCI3): δ 1 .33 (t, 3H), 1 .85 (s, 6H), 4.31 (q, 2H), 6.57 (s, 1 H), 8.42 (s, 1 H), 8.49 (s, 1 H), 8.66-8.80 (m, 3H). LCMS m/z = 288 [M+H]+
Preparation 16
Ethyl 2-{[1 -(6-methylpyridin-3-yl)ethyl1amino)pyrimidine-5-carboxylate
Figure imgf000087_0003
1 -(6-Methylpyridin-3-yl)ethanamine (Preparation 30 a, 10.9 g, 80.4 mmol) was added to a solution of ethyl 2-chloropyrimidine-5-carboxylate (15 g, 80.39 mmol) and DIPEA (13.5 g, 105 mmol) in 2-propanol (48 ml_) and the resulting mixture heated under reflux for 16 hrs. The cooled mixture was concentrated in vacuo and the residue was purified by column chromatography on silica gel eluting with EtOAc: pet. Ether (0: 100 to 30:70) to provide the title compound as a yellow solid, 16 g (69%). 1H NMR (400 MHz, CDCI3): δ 1 .36 (t, 3H), 1 .61 (d, 3H), 2.55 (s, 3H), 4.33 (q, 2H), 5.22-5.30 (m, 1 H), 5.92 (br d, 1 H), 7.13 (d, 1 H), 7.58 (dd, 1 H), 8.54 (d, 1 H), 8.82 (s, 2H). LCMS m/z = 287 [M+H]+
Preparation 17 Ethyl 2-{[(1 S)-1 -(6-methylpyridin-3-yl)ethyl1amino)pyrimidine-5-carboxylate and
Preparation 18
Ethyl 2-{[(1 R)-1 -(6-methylpyridin-3-yl)ethyl1amino)pyrimidine-5-carboxylate
Figure imgf000088_0001
Ethyl 2-{[1 -(6-methylpyridin-3-yl)ethyl]amino}pyrimidine-5-carboxylate
(Preparation 16) was further purified by SFC separation using the following: AD 250mm*50mm, 10pm column; 60% EtOH (0.1 %NH3.H2O) in C02 at 200ml/min, 38 °C; to provide ethyl 2-{[(1 S)-1 -(6-methylpyridin-3-yl)ethyl]amino}pyrimidine-5-carboxylate as a yellow oil (RT: 6.31 1 min, 5.73 g, 44%). 1H NMR (400 MHz, CDCI3): δ 1 .36 (t, 3H), 1 .60 (d, 3H), 2.54 (s, 3H), 4.33 (q, 2H), 5.22-5.29 (m, 1 H), 5.94 (br d, 1 H), 7.12 (d, 1 H), 7.57 (dd, 1 H), 8.53 (d, 1 H), 8.81 (s, 2H). LCMS m/z = 287 [M+H]+. a[D] 26 = 134.9° (c = 1 .012, CHCIs).
Further elution provided ethyl 2-{[(1 R)-1 -(6-methylpyridin-3- yl)ethyl]amino}pyrimidine-5-carboxylate as a yellow oil (RT: 7.481 min, 5.77 g, 44%). 1H NMR (400 MHz, CDCI3): δ 1 .36 (t, 3H), 1 .60 (d, 3H), 2.53 (s, 3H), 4.33 (q, 2H), 5.22- 5.29 (m, 1 H), 5.98 (br d, 1 H), 7.1 1 (d, 1 H), 7.56 (dd, 1 H), 8.53 (d, 1 H), 8.81 (s, 2H). LCMS m/z = 287 [M+H]+. a[D] 25 1 = -129.4° (c = 1 .151 , CHCI3)
Preparation 19
Ethyl 4-methyl-2-{[1 -(pyrimidin-5-yl)cvclopropyl1amino)pyrimidine-5-carboxylate
Figure imgf000089_0001
1 -(Pyrimidin-5-yl)cyclopropanamine hydrochloride (Preparation 26, 573 mg, 2.39 mmol) was added to a solution of ethyl 2-chloro-4-methylpyrimidine-5-carboxylate (400 mg, 1 .99 mmol) and DIPEA (515 mg, 3.99 mmol) in NMP (4 mL) and the resulting mixture stirred under microwave irradiation at 140°C for 40 min. The cooled mixture was diluted with EtOAc (200 mL), washed with brine (100 mL), H20 (100 mL), dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel eluting with Pet. Ether: EtOAc (80:20 to 40:60) to afford the title compound as a yellow gum, 200 mg, 33%. 1H NMR (400 MHz, CDCI3): δ 1 .35 (t, 3H), 1 .42 (br s, 4H), 2.64 (s, 3H), 4.31 (q, 2H), 6.22 (br s, 1 H), 8.67 (br s, 2H), 8.82 (br s, 1 H), 9.04 (s, 1 H). LCMS m/z = 300 [M+H]+
Preparation 20
Ethyl 4-methyl-2-[(pyrazine-2-ylmethyl)aminolpyrimidine-5-carboxylate
Figure imgf000089_0002
The title compound was obtained as an off white solid, in 59% yield from ethyl 2- chloro-4-methylpyrimidine-5-carboxylate and 2-(aminomethyl)pyrazine, following an analogous method to that described in Preparation 19, except DCM:MeOH was used as the column eluent. 1H NMR (400 MHz, CDCI3): δ 1 .36 (t, 3H), 2.66 (s, 3H), 4.32 (q, 2H), 4.85 (d, 2H), 6.63 (br s, 1 H), 8.48 (s, 1 H), 8.53 (dd, 1 H), 8.66 (s, 1 H), 8.63 (br s, 1 H). LCMS m/z = 274 [M+H]+
Preparation 21
Ethyl 2-fluoropyrimidine-5-carboxylate
Figure imgf000089_0003
AgF (6.8 g, 53.6 mmol) was added in one portion to a colorless solution of ethyl 2-chloropyrimidine-5-carboxylate (5.0 g, 26.80 mmol) in MeCN (100 mL) and the resulting suspension stirred at 70 °C for 16 hrs. The cooled mixture was filtered and the filtrate was concentrated in vacuo to a volume of approx. 10 mL. The solution was purified by column chromatography on silica gel eluting with pet. ether: EtOAc (80:20) to provide the title compound as a yellow solid 2.9 g, 64%. 1H NMR (400 MHz, CDCI3) : δ 1 .43 (t, 3H), 4.45 (q, 2H), 9.21 (s, 2H). LCMS m/z = 171 [M+H]+
Preparation 22
(2-Chloropyrimidin-5-yl)(8-oxa-2-azaspiro[4.51dec-2-yl)methanone
Figure imgf000090_0001
To a -10 °C slurry of 2-chloropyrimidine-5-carboxylic acid (30 g, 189.2 mmol) and 8-oxa-2-azaspiro[4.5]decane hydrochloride (36.6 g, 206.0 mmol) in acetonitrile (210 mL), was slowly added propylphosphonic anhydride solution (273 mL, 458.9 mmol, 50% in EtOAc). Then a solution of Et3N (96 mL, 688.3 mmol) in acetonitrile (90 mL) was added over a period of 3h, keeping the temperature below -5 °C. The mixture was stirred at this temperature for 10 minutes, then water (300 mL) was added. The resulting slurry was evaporated under reduced pressure (35 °C, 90 mmHg) until no more distillation was observed, then cooled down to 5 °C. The slurry was filtered and the solid washed with water (90 mL). The resulting white solid was dried in a vacuum oven at 40 °C, providing the title compound, 49.75 g, 96%. 1H NMR (400 MHz, CDCI3): δ 1 .55-1 .61 (m, 2H), 1 .65-1 .69 (m, 2H), 1 .89-1 .98 (m, 2H), 3.37 (s, 1 H), 3.57-3.82 (m, 7H), 8.82 (s, 2H). LCMS m/z = 282 [M+H]+
Preparation 23
(2-Chloropyrimidin-5-yl)(7-oxa-2-azaspiro[3.51non-2-yl)methanone
Figure imgf000090_0002
A yellow suspension of 2-chloropyrimidine-5-carboxylic acid (3.0 g, 18.92 mmol), 7-oxa-2-azaspiro[3.5]nonane (2.89 g, 22.72 mmol), propylphosphonic anhydride solution (12.0 g, 37.8 mmol, 50% in EtOAc) and Et3N (9.57 g, 94.6 mmol) in THF (50 mL) was stirred at 60 °C for 16 hrs. The cooled mixture was diluted wth EtOAc (300 mL) washed with sat. aqueous NH4CI (150 mL), brine (150 mL), dried (Na2SO4) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel eluting with Pet. EtherEtOAc (100:0 to 50:50) to afford the title compound as a pale yellow solid, 3.07 g, 61 %. 1H NMR (400 MHz, DMSO-d6): δ 1 .64-1 .79 (m, 4H), 3.43-3.48 (m, 2H), 3.52-3.56 (m, 2H), 3.81 (s, 2H), 4.16 (s, 2H), 9.00 (s, 2H). LCMS m/z = 268 [M+H]+
Preparation 24 2-(Pyrimidin-5-yl)propan-2-amine hydrochloride
Figure imgf000091_0001
A solution of fe/f-butyl [2-(pyrimidin-5-yl)propan-2-yl]carbamate (Preparation 33, 5.7 g, 24.02 mmol) in 1 M HCI in MeOH (20 mL) was stirred at 20 °C for 2 hrs. The reaction mixture was evaporated under reduced pressure, the residue washed with EtOAc, filtered and dried to afford the title compound as a white solid, 4.0 g, 96%. 1 H NMR (400 MHz, DMSO-d6): δ 1 .71 (s, 6H), 8.20 (br s, 1 H), 9.09 (s, 2H), 9.15-9.18 (m, 3H). LCMS m/z = 138 [M+H]+
Preparation 25
1 -(Pyrimidin-5-yl)cvclobutanamine hydrochloride
Figure imgf000091_0002
The title compound was obtained in quantitative yield from fe/f-butyl [1 -(pyrimidin-5- yl)cyclobutyl]carbamate (Preparation 34), following the procedure described in Preparation 24. 1H NMR (400 MHz, DMSO-d6): δ 1 .81 -1 .86 (m, 1 H), 2.17-2.24 (m, 1 H), 2.63-2.72 (m, 4H), 9.03 (s, 2H), 9.17-9.28 (m, 4H). LCMS m/z =150 [M+H]+
Preparation 26
1 -(Pyrimidin-5-yl)cyclopropanamine hydrochloride
Figure imgf000092_0001
A solution of fe/f-butyl [1 -(pyrimidin-5-yl)cyclopropyl]carbamate (Preparation 35) (19.0 g, 80.8 mmol) in 1 M HCI in MeOH (200 mL) was stirred at 25 °C for 2 hrs. The solvent was removed under reduced pressure to afford the title compound as a yellow solid, 13.6 g (98%). 1H NMR (400 MHz, DMSO-d6): δ 1 .30-1 .35 (m, 2H), 1 .46-1 .51 (m, 2H), 8.92 (s, 2H), 9.15 (s, 1 H), 9.33 (br s, 3H). LCMS m/z = 136 [M+H]+
Preparation 27
(1 S)-1 -(Pyrazin-2-vQethanamine hydrochloride N^|^NH2 HCI A mixture of 2-methyl-N-[(1 S)-1 -(pyrazin-2-yl)ethyl]propane-2-sulfinamide
(Preparation 46, 320 mg, 1 .41 mmol) in 1 M HCI in MeOH (8 mL) was stirred at 0 °C for 1 hr. The resulting suspension was evaporated under reduced pressure to afford the title compound as an off- white solid in quantitative yield. 1H NMR (400 MHz, MeOD-d4): δ 1 .69 (d, 3H), 4.75-4.82 (m, 1 H), 8.69 (d, 1 H), 8.78 (dd, 1 H), 8.82 (d, 1 H). LCMS m/z = 124 [M+H]+
Preparations 28 to 30
H R1 b
V
G NH2 HCI
The compounds were prepared from the appropriate sulfinamide starting material in approximately quantitative yield, following the procedure described in Preparation 27.
H R1 b
V
Preparation G NH2 HCI Starting Material; Name; Data
28 Preparation 47: 2-methyl-N-[(1 R)-1 -(pyrazin-2- yl)ethyl]propane-2-sulfinamide
Figure imgf000093_0001
(1 R)-1 -(Pyrazin-2-yl)ethanamine hydrochloride
1H NMR (400 MHz, MeOD-d4) : δ 1 .68 (d, 3H), 4.77-4.84 (m, 1 H), 8.70 (d, 1 H), 8.81 (dd, 1 H),
8.84 (d, 1 H). LCMS m/z = 124 [M+H]+
29 Preparation 48, 2-methyl-N-[1 -(pyrimidin-5- yl)ethyl]propane-2-sulfinamide ■ HCI 1 -(Pyrimidin-5-yl)ethanamine hydrochloride
Figure imgf000093_0002
1H NMR (400 MHz, DMSO-d6) : δ 1 .61 (d, 3H),
4.46-4.58 (m, 1 H), 8.94 (br s, 2H), 9.04 (s, 2H), 9.20 (s, 1 H), 10.33 (br s, 1 H). LCMS m/z = 124
[M+H]+
30 Preparation 49, 2-methyl-N-[1 -(6-methylpyridin-3- yl)ethyl]propane-2-sulfinamide ■ HCI 1 -(6-Methylpyridin-3-yl)ethanamine hydrochloride
Figure imgf000093_0003
1H NMR (400 MHz, MeOD-d4) : δ 1 .76 (d, 3H),
2.87 (s, 3H), 4.77-4.85 (m, 1 H), 8.07 (d, 1 H), 8.67 (dd, 1 H), 8.92 (d, 1 H). LCMS m/z = 137 [M+H]+
Preparation 30
1 -(6-Methylpyridin-3-yl)ethanam ine
Figure imgf000093_0004
Zn powder (20.9 g, 320 mmol) was added in portions to a refluxing solution of
(1 E)-N-hydroxy-1 -(6-methylpyridin-3-yl)ethanamine (Preparation 55, 16.0 g, 106.54 mmol) and NH4OAc (10.7 g, 139 mmol) in EtOH (137 mL) and NH4OH (228 mL) and the resulting mixture was stirred at 95 °C for 16 hrs. The cooled mixture was evaporated under reduced pressure and the residue basified with aq. NaOH to pH>12. The resulting suspension was filtered, the solids washed with DCM, the organic filtrate was washed with brine, dried (Na2S04), filtered off and the filtrate evaporated under reduced pressure to afford the title compound as a yellow oil, 14.3g (98%). 1H NMR (400 MHz, DMSO-de): δ 1 .23 (d, 3H), 2.42 (s, 3H), 3.94-4.01 (m, 1 H), 7.16 (d, 1 H), 7.64 (dd, 1 H), 8.40 (d, 1 H). LCMS m/z = 137 [M+H]+
Preparation 31 3-(Pyrazin-2-yl)oxetan-3-am ine
Figure imgf000094_0001
A solution of 2-methyl-/V-[3-(pyrazin-2-yl)oxetan-3-yl]propane-2-sulfinamide (Preparation 44, 2.2 g, 8.62 mmol) in EtOAc (10 mL) and HCI/EtOAc (1 M, 5 mL) was stirred at 0 °C for 5 min. The resulting precipitate was filtered off. The filter cake was dissolved in MeOH (50 mL), NaHCO3 added (4 g) and the mixture stirred at 25 °C for 15 min. The mixture was diluted with DCM (80 mL) filtered and the filtrate concentrated in vacuo. The crude material was purified by column chromatography on silica gel eluting with MeOH: EtOAc (0: 100 to 5:95) to afford the compound as a yellow oil, 400 mg, 10%. 1H NMR (400 MHz, CDCI3): δ 4.77-4.82 (m, 2H), 4.89-4.93 (m, 2H), 9.03 (s, 2H), 9.18 (s, 1 H). LCMS m/z = 152 [M+H]+
Preparation 32
3-(Pyrimidin-5-yl)oxetan-3-amine
Figure imgf000094_0002
The title compound was obtained as a yellow oil in 93% yield from 2-methyl-/V-[3- (pyrazin-2-yl)oxetan-3-yl]propane-2-sulfinamide (Preparation 44), following an analogous method to that described in Preparation 31 . 1H NMR (400 MHz, CDCI3): δ 4.78 (m, 2H), 4.90 (m, 2H), 9.03 (s, 2H), 9.18 (s, 1 H). LCMS m/z = 152.1 [M+H]+
Preparation 33 Te/f-butyl [2-(pyrimidin-5-yl)propan-2-yl1carbamate
Figure imgf000095_0001
DPPA (2330 mg, 8.45 mmol) was added to a solution of Et3N (dried over KOH) (855 mg, 8.45 mmol) and 2-methyl-2-(pyrimidin-5-yl)propanoic acid (Preparation 36, 1 170 mg, 7.04 mmol) in distilled f-BuOH (20 mL) and the mixture heated to 1 10 °C for 16 hrs. The cooled mixture was poured into aq. NH4CI solution and extracted with EtOAc (100 mL x 3). The combined organic extracts were dried (MgS04), filtered and evaporated under reduced pressure. The residue was purified by column chromatography on silica gel eluting with 0-40% EtOAc in Pet. Ether to provide the title compound as a colorless oil, 1.03 g, 62%. 1H NMR (400 MHz, CDCI3): δ 1 .38 (br s, 9H), 1 .65 (s, 6H), 5.12 (br s, 1 H), 8.76 (s, 2H), 9.08 (s, 1 H). LCMS m/z = 238 [M+H]+
Preparation 34
Te/f-butyl [1 -(pyrimidin-5-yl)cvclobutyl1carbamate
Figure imgf000095_0002
The title compound was obtained in 34% yield from 1 -(pyrimidin-5- yl)cyclobutane-1 -carboxylic acid (Preparation 37), following the method described in Preparation 33. 1H NMR (400 MHz, CDCI3): δ 1 .36 (br s, 9H), 1 .80-1 .98 (m, 1 H), 2.10- 2.21 (m, 1 H), 2.42-2.48 (m, 2H), 2.53-2.57 (m, 2H), 8.79 (s, 2H), 9.09 (s, 1 H). LCMS m/z = 250 [M+H]+
Preparation 35
Te/f-butyl [1 -(pyrimidin-5-yl)cvclopropyl1carbamate
Figure imgf000095_0003
DPPA (13.50 g, 49.0 mmol) and Et3N (6.19 g, 61.20 mmol) were added to a suspension of 1 -(pyrimidin-5-yl)cyclopropane-1 -carboxylic acid (Preparation 38, 6.7 g, 40.81 mmol) in f-BuOH (120 mL) at 30 °C. The resulting mixture was degassed with N2 and stirred at 100 °C for 16 hrs. The cooled mixture was concentrated in vacuo and the residue partitioned between aq. NH4CI (50 mL) and pet. ether (50 mL). The mixture was stirred at 0 °C for 10 min and the resulting solid collected by filtration. The solid was washed consecutively with aq.NaHCOs (20 mL), water (20 mL) and pet. ether (20 mL) then dried in vacuo to afford the title product as a white solid, 8.1 g, 84% yield. 1H NMR (400 MHz, CDCI3): δ 1 .24-1 .32 (m, 2H), 1 .38-1 .48 (m, 1 1 H), 5.27-5.40 (m, 1 H), 8.61 (s, 2H), 9.06 (s, 1 H). LCMS m/z = 236 [M+H]+
Preparation 36
2-Methyl-2-(pyrimidin-5-yl)propanoic acid
Figure imgf000096_0001
NaOH (635 mg, 15.9 mmol) was added to a solution of methyl 2-methyl-2-
(pyrimidin-5-yl)propanoate (Preparation 39, 1.43 g, 7.94 mmol) in THF (5 mL) and H20 (5 mL) and the reaction stirred at 15 °C for 2 hrs. The mixture was concentrated in vacuo, the aqueous solution extracted with EtOAc (5 mL) then acidified with HCI (1 N) to pH=3. This aqueous solution was extracted with further EtOAc, and the organic extract evaporated under reduced pressure. The crude product was suspended in a solution of (DCM:MeOH=5: 1 )(20 mL). The mixture was filtered and the filtrate evaporated under reduced pressure to provide the title compound as a white solid, 1 .3 g (99% yield). 1H NMR (400 MHz, DMSO-d6): δ 1 .47 (s, 6H), 8.76 (s, 2H), 8.99 (s, 1 H). LCMS m/z = 165 [M-H]- Preparation 37
1 -(Pyrimidin-5-yl)cvclobutane-1 -carboxylic acid
Figure imgf000096_0002
NaOH (1 .08 g, 27.1 mmol) was added to a solution of methyl 1 -(pyrimidin-5- yl)cyclobutane-1 -carboxylate (Preparation 40, 2.6 g, 13.53 mmol) in THF (5 mL) and H20 (5 mL) and the resulting mixture stirred at 18 °C for 2 hrs. The reaction was concentrated in vacuo and the residue acidified with 2N HCI to pH=6 (white solid formed). The solid was collected by filtration and washed with water (5 mL x 2). The solid was co-evaporated with toluene three times to afford the title compound as a white solid, 1 .10 g, 43% yield. 1H N MR (400 MHz, DMSO-d6): δ 1 .81 -1 .92 (m, 1 H), 1 .98-2.10 (m, 1 H), 2.49-2.58 (m, 2H), 2.70-2.79 (m, 2H), 8.74 (s, 2H), 9.09 (s, 1 H). LCMS m/z = 179 [M+H]+
Preparation 38
1 -(Pyrimidin-5-yl)cvclopropane-1 -carboxylic acid
Figure imgf000097_0001
The title compound was obtained in 77% yield from methyl 1 -(pyrimidin-5- yl)cyclopropane-1 -carboxylate (Preparation 41 ), following a similar method to that described in Preparation 37, except the reaction was conducted using THF/MeOH/H20 (1 : 1 : 1 ) as solvent. 1H NMR (400 MHz, DMSO-d6): δ 1 .28-1 .33 (m, 2H), 1 .49-1 .54 (m, 2H), 8.78 (s, 2H), 9.07 (s, 1 H), 12.69 (br s, 1 H). LCMS m/z = 163 [M-H]"
Preparation 39
Methyl 2-methyl-2-(pyrimidin-5-yl)propanoate
Figure imgf000097_0002
LiHMDS (39.4 ml_, 39.4 mmol, 1 M solution in THF) was added drop wise to a -
78 °C solution of methyl 2-(pyrimidin-5-yl)acetate (1 .5 g, 9.86 mmol) in THF (20 ml_) under N2. The resulting mixture was stirred at -78 °C for 1 hr. A solution of iodomethane (15.1 g, 106.38 mmol) in THF (10 ml_) was added drop wise to the reaction mixture at -78 °C. After complete addition, the resulting brown solution was allowed to warm to 18 °C and stirred for 16 hrs. The reaction was poured into saturated NH4CI solution (200 ml_) and the mixture extracted with EtOAc (200 ml_ x3). The combined organic extracts were washed with brine, dried (Na2S04), filtered and concentrated in vacuo to afford a brown liquid. The crude product was purified by column chromatography on silica gel eluting with Pet. Ether: EtOAc (100:0 to 70:30) to provide the title compound as a yellow oil, 1 .3 g, 73%. 1H NMR (400 MHz, CDCI3): δ 1 .63 (s, 6H), 3.67 (s, 3H), 8.73 (s, 2H), 9.10 (s, 1 H). LCMS m/z = 181 [M+H]+
Preparation 40
Methyl 1 -(pyrimidin-5-yl)cvclobutan-1 -carboxylate
Figure imgf000098_0001
LiHMDS (56.2 mL, 56 mmol, 1 M solution in THF) was added drop wise to a stirred yellow solution of methyl 2-(pyrimidin-5-yl)acetate (8.00 g, 47 mmol) in THF (150 mL) at -70 °C under N2 and the reaction was stirred at -70 °C for 1 hr. A solution of 1 ,3-diiodopropane (13.8 g, 46.8 mmol) in THF (20 mL) was added drop wise to the reaction at -70 °C and the mixture allowed to warm to 20 °C slowly and stirred for 1 hr. The reaction mixture was again cooled to -70 °C and an additional portion of LiHMDS (1 M in THF, 56.2 mL, 56.2 mmol) was added drop wise. After addition, the reaction was allowed to warm to 20 °C slowly and stirred for an additional hour. The reaction was poured into saturated NH4CI solution (60 mL) the mixture extracted with EtOAc (300 mL x 3). The combined organic extracts were washed with brine, dried (Na2S04) filtered and concentrated in vacuo. The crude product was purified by column chromatography on silica gel eluting with pet. ether: EtOAc (100:0 to 65:35) to afford the title compound as a yellow oil, 1 .95 g, 22% yield. 1H NMR (400 MHz, CDCI3): 1 .90-2.04 (m, 1 H), 2.12- 2.25 (m, 1 H), 2.48-2.59 (m, 2H), 2.87-2.96 (m, 2H), 3.67 (s, 3H), 8.66 (s, 2H), 9.09 (s, 1 H). LCMS m/z = 193 [M+H]+
Preparation 41
Methyl 1 -(pyrimidin-5-yl)cvclopropan-1 -carboxylate
Figure imgf000098_0002
LiHMDS (1 M solution in THF, 21 1 mL, 21 1 mmol) was added drop wise at -70
°C to a stirred solution of methyl 2-(pyrimidin-5-yl)acetate (26.73 g, 175.7 mmol) in THF (200 mL) and the resulting mixture was stirred at this temperature for 1 hr. A solution of 1 ,3,2-dioxathiolane 2,2-dioxide (26.2 g, 21 1 mmol) in THF (200 mL) was added drop wise so as to maintain the temperature at -70 °C and on complete addition, the reaction was brought slowly to -20 °C and stirred for 1 .5 hr. The reaction mixture was again cooled to -72 °C and an additional portion of LiHMDS (1 M in THF, 21 1 mL, 21 1 mmol) added. The reaction mixture was then allowed to warm to room temperature and stirred for 15 hrs. The reaction was quenched with saturated NH4CI solution (300 mL) and the mixture extracted with EtOAc (3 x 500 mL). The combined organic extracts were washed with brine, dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography eluting with pet. Ether: EtOAc (100:0 to 65:35) to afford the title compound as a yellow oil, 25.0 g, 80 %. 1H NMR (400MHz, CDCI3): δ 1 .20-1 .25 (m, 2H), 1 .67-1.74 (m, 2H), 3.63 (s, 3H), 8.69 (s, 2H), 9.03 (s, 1 H). LCMS m/z = 179 [M+H]+
Preparation 42
1 -(Pyrazin-2-yl)cvclobutanamine
Figure imgf000099_0001
A solution of acetyl chloride (18.8 g, 240 mmol) in MeOH (60 ml_) was added to a solution of 2-methyl-N-[1 -(pyrazin-2-yl)cyclobutyl]propane-2-sulfinamide (Preparation 43, 3.4 g, 13.42 mmol) in MeOH (5 ml_) maintaining the reaction temperature below 10 °C. On complete addition, the reaction was stirred at 20 °C for 1 hr. The reaction mixture was concentrated in vacuo and the residue dissolved in MeOH (60 ml_). NaHCO3 solid was added until no gas was released. The suspension was filtered and washed with MeOH. The filtrate was concentrated in vacuo and the residue dissolved in DCM (60 ml_). The suspension was filtered again, washing through with DCM. The filtrate was evaporated under reduced pressure to afford the title compound as a brown solid, 1 .7 g, 85%. 1H NMR (400 MHz, DMSO-d6): 1 .90-2.02 (m, 1 H), 2.1 1 -2.25 (m, 1 H), 2.53-2.70 (m, 4H), 8.68 (d, 1 H), 8.71 (dd, 1 H), 8.38-8.92 (br s, 2H), 9.09 (s, 1 H). LCMS m/z = 150 [M+H]+
Preparation 43
2-Methyl-N-[1 -(pyrazin-2-yl)cvclobutyllpropane-2-sulfinamide
Figure imgf000099_0002
n-BuLi (25.4 ml_, 63.5 mmol, 2.5 M in hexane) was added drop wise to a -78 °C solution of 2-bromopyrazine (10.1 g, 63.5 mmol) in toluene (150 ml_) under N2. After 10 min at -78 °C, a solution of N-cyclobutylidene-2-methylpropane-2-sulfinamide (Preparation 50, 10.0 g, 57.71 mmol) in toluene (50 ml_) was added slowly. The resulting dark red solution was stirred for 1 hr at -78 °C. The reaction was quenched by the addition of sat. NH4CI solution (10 ml_) and the reaction mixture dried (MgS04), filtered and concentrated in vacuo to give a brown oil. The crude product was purified by column chromatography on silica gel eluting with pet. Ether: EtOAc (100:0 to 0: 100) to MeOH: EtOAc (9:91 ) to afford the title compound as a yellow oil, 4.0 g, 27%. 1H NMR (400 MHz, CDCI3): δ 1 .21 (s, 9H), 1.86-1 .96 (m, 1 H), 2.08-2.19 (m, 1 H), 2.58-2.78 (m, 4H), 3.62-3.75 (br s, 1 H), 8.49 (d, 1 H), 8.56 (dd, 1 H), 8.80 (d, 1 H). LCMS m/z = 254 [M+H]+
Preparation 44
2-Methyl-/V-[3-(pyrazin-2-yl)oxetan-3-yllpropane-2-sulfinamide
Figure imgf000100_0001
The title compound was prepared as a yellow oil in 25% yield from 2-methyl-N- (oxetan-3-ylidene)propane-2-sulfinamide (Preparation 51 ) and 2-bromopyrazine following the method described in Preparation 43. 1H NMR (400 MHz, CDCI3): δ 1 .29 (s, 9H), 4.94 (d, 1 H), 5.07-5.15 (m, 2H), 5.35 (d, 1 H), 8.56-8.58 (m, 1 H), 8.59-8.60 (m, 1 H), 9.08 (d, 1 H). LCMS m/z = 256 [M+H]+
Preparation 45
2-Methyl-/V-[3-(pyrimidin-5-yl)oxetan-3-yllpropane-2-sulfinamide
Figure imgf000100_0002
The title compound was prepared as a yellow oil in 14% yield from 2-methyl-N- (oxetan-3-ylidene)propane-2-sulfinamide (Preparation 51 ) and 5-bromopyrimidine following an analogous method to that described in Preparation 43, except THF was used as the reaction solvent. 1H NMR (400 MHz, CDCI3): δ 1 .22 (s, 9H), 4.86 (d, 1 H), 5.06 (d, 1 H), 5.14 (s, 2H), 8.86 (s, 2H), 9.20 (s, 1 H). LCMS m/z = 139 [M+H]+
Preparation 46 (S)-2-methyl-N-((S)-1 -(pyrazin-2-yl)ethyl)propane-2-sulfinamide and Preparation 47
(S)-2-methyl-N-((R)-1 -(pyrazin-2-yl)ethyl)propane-2-sulfinamide
Figure imgf000101_0001
Dimethyl zinc (21 .3 mL, 21 .3 mmol, 1 .0 M in toluene) was added in an oven- dried and N2-purged flask, followed by MeMgBr (6.25 ml_,18.7 mmol, 3.0 M in ether) over 1 min with stirring at 15°C and the solution allowed to stir for 20 mins. This solution was added drop wise over 30 min into a cooled (-68 °C) suspension of 2-methyl-N-[(E)- pyrazin-2-ylmethylidene]propane-2-sulfinamide (Preparation 53, 1 .8 g, 8.52 mmol) in anhydrous THF (25.8 mL) and the reaction then allowed to stir at -68 °C for 1 hr. The reaction was quenched by the drop wise addition of saturated NH4CI soln. (10 mL) maintaining the temperature below -60 °C. The mixture was then allowed to warm to rt, the resulting solid filtered off, washed with EtOAc (200 mL) and MeOH (20 mL) and the filtrate concentrated in vacuo. The crude product was purified by automated column chromatography on silica gel, eluting with EtOAc: DCM (20:80 to 95:5) to afford a yellow gum, 2.4 g. This was further purified by preparative HPLC using an Agela ASB 150*25mm*5um column, eluting with 16-46% (0.225% TFA in water): MeCN at a flow rate of 25 mL/min to afford 2-methyl-N-[(1 S)-1 -(pyrazin-2-yl)ethyl]propane-2-sulfinamide as a yellow oil, 740 mg. 1H NMR (400 MHz, CDCI3): δ 1 .20 (s, 9H), 1 .64 (d, 3H), 3.95 (br d, 1 H), 4.67-4.75 (m, 1 H), 8.48 (d, 1 H), 8.53 (dd, 1 H), 8.61 (d, 1 H). SFC RT [Method CA-G] = 4.632 min
Further elution provided 2-methyl-N-[(1 R)-1 -(pyrazin-2-yl)ethyl]propane-2- sulfinamide as a yellow oil, 700 mg. 1H NMR (400 MHz, CDCI3): δ 1 .25 (s, 9H), 1 .55 (d, 3H), 4.26 (br s, 1 H), 4.65-4.70 (m, 1 H), 8.49 (d, 1 H), 8.52 (dd, 1 H), 8.62 (d, 1 H). SFC RT [Method CA-G] = 3.593 min
Preparation 48
2-Methyl-/V-[1 -(pyrimidin-5-yl)ethyllpropane-2-sulfinamide
Figure imgf000101_0002
NaBH4 (1610 mg, 42.6 mmol) was added portion wise to an ice-cooled solution of 2-methyl-N-[(1 E)-1 -(pyrimidin-5-yl)ethylidene]propane-2-sulfinamide (Preparation 52, 3.2 g, 14.20 mmol) in THF (25 ml_) and MeOH (25 ml_) and the resulting mixture stirred at 20 °C for 1 hr. The mixture was cooled to 0 °C and quenched with saturated NH4CI soln. (10 ml_). The resulting solid was filtered off and the solid washed with DCM (100 ml_) and MeOH (20 ml_). The combined organic filtrates were dried over MgSO4 and concentrated in vacuo. The residue was purified by column chromatography on silica gel eluting with pet. Ether: EtOAc (100:0 to 0: 100), then MeOH: DCM, (0: 100 to 30:70) to afford the title compound as a white solid, 1 .6 g, 50 % (title compound was isolated as a 1 : 1 mixture of diastereomers and used as is in the next step). 1H NMR (400 MHz, CDCIs): δ 1.20 (s, 4.5H), 1 .23 (s, 4.5H), 1 .59 (d, 1 .5H), 1 .60 (d, 1 .5H), 3.47 (br d, 0.5H), 3.59 (br d, 0.5H), 4.55-4.66 (m, 1 H), 8.71 (s, 1 H), 8.74 (s, 1 H), 9.14 (s, 0.5H), 9.15 (s, 0.5H). LCMS m/z = 228 [M+H]+
Preparation 49 2-Methyl-N-[1 -(6-methylpyridin-3-yl)ethyllpropane-2-sulfinamide
Figure imgf000102_0001
Me2Zn (5.63 mmol, 1 M in toluene, 5.63 ml_) was added to an oven-dried and N2- purged round-bottomed flask and MeMgBr (0.497 mmol, 3 M in ether) was added over 1 min. The solution was stirred at 15 °C for 30 min. This solution was added drop wise over 30 mins to a -78 °C solution of 2-methyl-N-[(E)-(6-methylpyridin-3- yl)methylidene]propane-2-sulfinamide (Preparation 54, 0.7 g, 3.31 mmol) in anhydrous THF (10 ml_) so as to maintain the internal temperature below -70 °C. Upon complete addition the reaction was stirred at -78 °C for 1 hr and then allowed to warm to 15 °C. The reaction was quenched with sat. aqueous NH4CI solution, the mixture was filtered, and the residue concentrated under reduced pressure. The residue was purified by column chromatography on silica gel eluting with DCM:MeOH (100 :0 to 60 :40) to afford the title compound as a colorless oil, 650 mg, 76% (title compound was isolated in ~3: 1 ratio with 1 -(6-methylpyridin-3-yl)ethanol and was used as is in the next step). 1H NMR (400 MHz, CDCI3): δ 1 .23 (s, 9H), 1 .54 (d, 3H), 2.56 (s, 3H), 4.53-4.60 (m, 1 H), 7.13-7.19 (m, 1 H), 7.60 (dd, 1 H), 8.50 (br s, 1 H). LCMS m/z = 241 [M+H]+ Preparation 50
/V-Cvclobutylidene-2-methylpropane-2-sulfinamide
Figure imgf000103_0001
2-Methyl-2-propanesulfinamide (19 g, 153 mmol) and Ti(OiPr)4 (81 .1 g, 285 mmol) were added to a solution of cyclobutanone (10.0 g, 142 mmol) in anhydrous THF (180 mL) and the resulting yellow solution was heated at 50 °C for 6 hrs. The reaction was cooled with an ice-water bath, diluted with anhydrous MeOH (100 mL) and EtOAc (100 mL) then sat. aq. NaHCO3 (20 mL) was added. The resulting suspension was stirred for 1 hr. The mixture was filtered, washed through with EtOAc and the filtrate was dried (Na2SO4) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel eluting with pet. Ether: EtOAc (100:0 to 80:20) to provide the title compound as a pale yellow oil, 17.0 g, 69%. 1H NMR (400 MHz, CDCIs): δ 1 .23 (s, 9H), 2.02-2.17 (m, 2H), 3.03-3.19 (m, 2H), 3.22-3.32 (m, 1 H), 3.44- 3.56 (m, 1 H). LCMS m/z =174 [M+H]+ Preparation 51
2-Methyl-N-(oxetan-3-ylidene)propane-2-sulfinamide
Figure imgf000103_0002
2-Methyl-2-propanesulfinamide (25.9 g, 214 mmol) followed by Ti(OiPr) (1 10 g, 389 mmol) were added to a solution of oxetan-3-one (14.0 g, 194 mmol) in anhydrous THF (250 mL) and the reaction mixture was heated at 50 °C for 16 hrs. The reaction mixture was cooled in an ice bath, anhydrous MeOH (140 mL) was added followed by sat. aq. NaHCO3 (20 mL) and the resulting suspension stirred for 1 hr. The solids were filtered off washing through with EtOAc. The filtrate was dried (Na2SO4) and concentrated in vacuo.The crude product was purified by column chromatography on silica gel column eluting with pet. Ether: EtOAc (100:0 to 80:20) to give the title product as a pale yellow oil, 15.0 g, 44%. 1H NMR (400 MHz, CDCI3): δ 1 .25 (s, 9H), 5.38-5.50 (m, 2H), 5.61 - 5.67 (m, 1 H), 5.74-5.81 (m, 1 H). LCMS m/z = 176 [M+H]+
Preparation 52
2-Methyl-N-[(1 E)-1 -(pyrimidin-5-yl)ethylidenelpropane-2-sulfinamide
Figure imgf000104_0001
2-Methyl-2-propanesulfinamide (2.73 g, 22.5 mmol) and Ti(OiPr)4 (1 1 .6 g, 40.9 mmol) were added to a solution of 1 -(5-pyrimidinyl)ethanone (2.5 g, 20.47 mmol) in DCM (100 mL) and the mixture was heated under reflux for 16 hrs. The mixture was cooled in an ice bath and diluted with anhydrous MeOH (20 mL) and aq. NaHCO3 (5 mL). The resulting suspension was stirred for 1 hr then filtered, washed through with EtOAC. The filtrate was dried (Na2SO4) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel column eluting with pet. Ether: EtOAc (100:0 to 50:50) to afford the title compound as a yellow oil, 3.2 g, 69%. (title compound was isolated in a ~85:15 mixture with 1 -(5-pyrimidinyl)ethanone, and the mixture was used as is in the next step). 1H NMR (400 MHz, CDCI3): δ 1 .32 (s, 9H), 2.80 (s, 3H), 9.15 (s, 2H), 9.28 (s, 1 H). LCMS m/ z= 226 [M+H]+
Preparation 53
(S)-2-Methyl-/V-[(E)-pyrazin-2-ylmethylidenelpropane-2-sulfinamide
Figure imgf000104_0002
The title compound was obtained as a yellow gum in 37% yield from (S)-(-)-2- methyl-2-propanesulfinamide and pyrazine 2-carbaldehyde, following an analogous procedure to that described in Preparation 52. 1H NMR (400 MHz, CDCI3): δ 1 .31 (s, 9H), 8.67 (d, 1 H), 8.72 (dd, 1 H), 8.75 (s, 1 H), 9.25 (d, 1 H). LCMS m/z = 212 [M+H]+ Preparation 54
2-Methyl-N-[(EH6-methylpyridin-3-yl)methylidenelpropane-2-sulfinamide
Figure imgf000104_0003
2-Methyl-2-propanesulfinamide (1 .5 g, 12.4 mmol) and copper(ll) sulfate (2.64 g, 16.5 mmol) were added to a solution of 6-methylpyridine-3-carboxaldehyde (1 g, 9.25 mmol) in DCM (30 mL) and the reaction heated to 50 °C for 18 hrs. The cooled mixture was filtered and washed with DCM. The filtrate was concentrated in vacuo. The residue was purified by column chromatography on silica gel eluting with EtOAc: Pet. Ether (0: 100 to 80:20) to afford the title compound as a colourless oil, 1 .55 g, 84% (title compound was isolated in a ~3: 1 mixture with 6-methylpyridine-3-carboxaldehyde, and the mixture was used as is in the next step). 1H NMR (400 MHz, CDCI3): δ 1 .26 (s, 9H), 2.63 (s, 3H), 7.28 (d, 1 H), 8.08 (dd, 1 H), 8.62 (s, 1 H), 8.89 (d, 1 H). LCMS m/z = 225 [M+H]+
Preparation 55
(1 E)-N-Hydroxy-1 -(6-methylpyridin-3-yl)ethanamine
Figure imgf000105_0001
NH2OH.HCI (26.2 g, 377 mmol) was added to a solution of 1 -(6-methylpyridin-3- yl)ethan-1 -one (17.0 g, 125.77 mmol) and sodium acetate (41 .3 g, 503 mmol) in EtOH (125 mL) and H20 (25 mL) and the resulting mixture was stirred at 25 °C for 7 hrs. The reaction mixture was diluted with EtOAc (1 L), washed with brine (400 mLx2), dried (Na2SO4), filtered and the filtrate evaporated under reduced pressure. The residue was washed with EtOH (10 mL) to provide the title compound as a white solid, 16.0 g, 85%. 1H NMR (400 MHz, DMSO-d6) : 2.16 (s, 3H), 2.48 (s, 3H), 7.26 (d, 1 H), 7.90 (dd, 1 H), 8.70 (d, 1 H), 1 1 .36 (s, 1 H). LCMS m/z = 151 [M+H]+
Preparation 56
(3aR,4R,7aS)-re/-Octahvdro-1 H-isoindol-4-ol hydrochloride
Figure imgf000105_0002
8.4% aq. HCI (300 mL) was added to a solution of fe/f-butyl (3aR,4R,7aS)-re/-4- hydroxyoctahydro-2H-isoindole-2-carboxylate (Preparation 57, 32.66 g, 0.136 mol) in 2-propanol (200 mL) and the mixture was heated under reflux for 1 hr. The cooled mixture was evaporated under reduced pressure and the residue triturated with ether, the resulting solid filtered off, washed with ether and dried to afford the title compound, 23.92g, 99%. 1H NMR (400 MHz, MeOD-d4): δ 1 .28-1 .80 (m, 8H), 2.41 -2.45 (m, 1 H), 2.54-2.59 (m, 1 H), 3.19-3.39 (m, 4H), 3.96-3.99 (m, 1 H). LCMS m/z = 142 [M+H]+
Preparation 57 fe/f-butyl (3aR,4R,7aS)-re/-4-hvdroxyoctahvdro-2H-isoindole-2-carboxylate
Figure imgf000106_0001
A mixture of (3aR,4R,7aS)-re/-2-benzyloctahydro-1 H-isoindol-4-ol (Preparation 58, 56 g, 0.242 mol), ammonium formate (45.8 g, 0.726 mol) and 10% Pd/C (5 g) in MeOH (500 mL) was stirred at rt until no starting material remained by tic analysis. The mixture was filtered and the filtrate evaporated under reduced pressure. The residue was dissolved in 2-propanol (500 mL) and Boc2O (52.8 g, 0.242 mol) added. The mixture was heated under reflux for 30 min, then concentrated under reduced pressure. DCM (500 mL) was added, the mixture was washed with water (100 mL), 5% NaHSO4 (100 mL) and again with water (100 mL). The organic layer was dried (MgSO4) and co- evaporated with silica gel (100 g). The residue was purified on a silica gel column eluting with hexane:EtOAc (50:50) to afford the title compound, 32.66 g, 56%.
Preparation 58
(3aR.4R.7aS)-re/-2-Benzyloctahvdro-1 H-isoindol-4-ol
Figure imgf000106_0002
1 N Solution of Li-Selectride® in THF (268 mL, 0.268 mol) was added at -78 C to a solution of 2-(phenylmethyl)-3aa,7aa-octahydro-1 H-isoindol-4-one (Procedure R, WO 9422823) (56.0 g, 0.244 mol) in THF (600 mL) and the solution stirred for 2 hrs at - 78°C, then allowed to warm to rt and stirred for a further 18 hrs. The mixture was concentrated in vacuo, the residue diluted with DCM, washed with water, sat. aq Na2CO3 and water then evaporated under reduced pressure to afford the title compound, 56.0 g, 99%.
Preparation 59
3-(Pyrrolidin-3-yl)-1 H-pyrazole hydrochloride bis HCI salt
Figure imgf000106_0003
A mixture of fe/f-butyl (E)-3-(3-(dimethylamino)acryloyl)pyrrolidine-1 -carboxylate (3.0 g, 1 1.18 mmol) and hydrazine (1 .1 1 mL, 12.3 mmol) in EtOH (20 mL) was heated under reflux for 2 hr and the cooled reaction was evaporated under reduced pressure. The residue was dissolved in EtOH and treated with HCI (4M in dioxane) and the mixture was stirred at rt for 1 hr. 2-MeTHF was added and the resulting solid filtered off and dried to afford the title compound as a solid. 1H NMR (400 MHz, DMSO-d6): δ 1 .95-2.05 (m, 1 H), 2.30-2.38 (m, 1 H), 3.14-3.35 (m, 3H), 3.53-3.66 (m, 2H), 6.59 (d, 1 H), 8.01 (d, 1 H), 9.86 (br s, 2H), 13.29 (br s, 2H). LCMS m/z = 138 [M+H]+
Preparation 60
3-Methyl-5-(Pyrrolidin-3-yl)-1 H-pyrazole hydrochloride bis HCI salt
Figure imgf000107_0001
HCI in dioxane (4M solution, 50 ml_) was added to a stirred solution of fe/f-butyl
3-(3-methyl-1 H-pyrazol-5-yl)pyrrolidine-1 -carboxylate (Preparation 61, 10.0 g, 39.84 mmol) in MeOH (30 ml_) at 0 °C and the reaction stirred at rt for 16 hrs. The mixture was concentrated in vacuo and the residue triturated with dry Et2O and dried under vacuum to afford the title compound as light yellow solid, 7.0 g, 94%. 1H NMR (400 MHz, DMSO-de): δ 1 .93-2.05 (m, 1 H), 2.28 (s, 3H), 2.30-2.40 (m, 1 H), 3.1 1 -3.34 (m, 3H), 3.57-3.67 (m, 2H), 6.55 (s, 1 H), 9.88 (br s, 2H), 14.48 (br s, 2H). LCMS m/z = 152 [M+H]+
Preparation 61
fe/f-Butyl 3-(3-methyl-1 H-pyrazol-5-yl)pyrrolidine-1 -carboxylate
Figure imgf000107_0002
ΝΗ2ΝΗ2Ή2Ο (5.32 g, 106.38 mmol) was added to a stirred solution of fe/f-butyl 3-[(2E)-3-(dimethylamino)but-2-enoyl]pyrrolidine-1 -carboxylate (Preparation 62, 20.0 g, 70.92 mmol) in MeOH (250 ml_) and the resulting mixture was heated under reflux for 2 hrs. The cooled mixture was concentrated in vacuo and the crude product purified by column chromatography on silica gel, eluting with MeOH: DCM (2:98 to 3:97) to afford the title compound as colorless oil, 12.0 g, 67%. LCMS m/z = 251 [M+H]+
Preparation 62
fe/f-Butyl 3-[(2E)-3-(dimethylamino)but-2-enoyllpyrrolidine-1 -carboxylate
Bocx
Figure imgf000107_0003
A stirred solution of fe/f-butyl 3-acetyl-1 -pyrrolidinecarboxylate (Commercial, 30 g, 140.66 mmol) and Ν, Ν-dimethylacetamide dimethyl acetal (180 ml_) in a sealed tube was heated to 105 °C for 15 hrs. The cooled reaction mixture was concentrated under reduced pressure to afford the title compound, 20 g, which was used without further purification. LCMS m/z = 283 [M+H]+
Preparation 63
Figure imgf000108_0001
10% Pd/C (300 mg) was added to a solution of benzyl 3-ethyl-3- methoxypyrrolidine-1 -carboxylate (Preparation 64, 1 .4 g, 5.32 mmol) in MeOH (10 ml_) and the reaction stirred at 25 °C under an atmosphere of 15 psi H2 for 2 hrs. The mixture was filtered through Celite® and the filtrate concentrated under reduced pressure to afford the title product as a colorless oil, 400 mg, 81 %. 1H NMR (400 MHz, CDCIs): δ 0.90 (t, 3H), 1 .52-1.68 (m, 3H), 1 .93 (ddd, 1 H), 2.57 (d, 1 H), 2.90-2.97 (m, 1 H), 3.04-3.12 (m, 2H), 3.14 (s, 3H), 3.47 (br s, 1 H). LCMS m/z = 130 [M+H]+
Preparation 64
Benzyl 3-ethy -3-methoxypyrrolidine-1 -carboxylate
Figure imgf000108_0002
NaH (375 mg, 15.6 mmol, 60% in mineral oil) was added to a solution of benzyl 3-ethyl-3-hydroxypyrrolidine-1 -carboxylate (Preparation 65, 2.6 g, 10.43 mmol) in THF (25 ml_) and the mixture stirred at 20 °C for 30 mins. lodomethane (4.44 g, 31 .3 mmol) was added and the reaction stirred at 60 °C for 16 hrs. The reaction was quenched by the addition of ice water, then extracted with EtOAc (200ml_ x2). The combined organic extracts were washed with H20 (200 ml_), brine (200 ml_), dried (Na2S04) and concentrated. The crude product was purified by column chromatography on silica gel eluting with pet. Ether: EtOAc (100:0-40:60) to afford the title compound as a colourless oil, 1.5 g, 55%. 1H NMR (400 MHz, CDCI3): δ 0.90-0.95 (m, 3H), 1 .59-1 .75 (m, 3H), 2.04-2.10 (m, 1 H), 3.14-3.18 (m. 4H), 3.45-3.68 (m, 3H), 5.10-5.18 (m, 2H), 7.31 -7.39 (m, 5H). LCMS m/z = 264 [M+H]+ Preparation 65
Benzyl 3-ethyl-3-hvdroxypyrrolidine-1 -carboxylate
Figure imgf000109_0001
1 -Carbobenzyloxy-3-pyrrolidinone (5.0 g, 22.81 mmol) in THF (10 mL) was added to a solution of EtMgBr (15.2 mL, 45.6 mmol, 3.0 M in Et20) in THF (40 mL) at 0°C and the reaction was stirred for 2 hrs. Saturated aqueous NH4CI solution (100 mL) was added and the mixture was extracted with EtOAc (200 mLx3). The combined organic extracts were washed with H20 (200 mL), dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel eluting with pet. Ether: EtOAc (100:0 to 50:50) to afford the title compound as a colourless oil, 2.6g , 46%. 1H NMR (400 MHz, CDCI3): δ 0.96-1 .04 (m, 3H), 1 .63-1 .71 (m, 2H), 1 .76- 1 .98 (m, 2H), 3.29 (dd, 1 H), 3.45 (dd, 1 H), 3.55-3.64 (m, 2H), 5.08-5.18 (m, 2H), 7.29- 7.39 (m, 5H). LCMS m/z = 250 [M+H]+
Preparation 66
(3-Ethylpyrrolidin-3-yl)methanol
Figure imgf000109_0002
A mixture of (1 -benzyl-3-ethylpyrrolidin-3-yl)methanol (104 g, 0.474 mol), ammonium formate (89.7 g, 1 .42 mol) and 10% Pd/C (10 g) in MeOH (1 L) was heated under reflux for 1 hr. The cooled mixture was filtered and Et3N (20 mL) was added. The mixture was evaporated under reduced pressure, and the residue was distilled twice, collecting at first the wide fraction and then the fraction in a boiling point (75-84°C at 0.3-0.4 mmHg) to afford the title compound, 25.5 g, 42%. 1H NMR (400 MHz, CDCI3): δ 0.80 (t, 3H), 1 .23-1 .51 (m, 4H), 2.32 (d, 1 H), 2.60-2.81 (m, 3H), 3.21 (s, 2H), 3.58 (br s, 2H). LCMS m/z = 130 [M+H]+
Preparation 67
-Benzyl-3-ethylpyrrolidin-3-yl)methanol
Figure imgf000109_0003
Methyl 1 -benzyl-3-ethylpyrrolidine-3-carboxylate (1 10 g, 0.51 mol) was dissolved in THF (600 mL), and water (240 mL) added. NaBH (9.5 g, 0.25 mol) was added in portions under stirring and cooling, maintaining the temperature of the mixture below 30°C. THF was evaporated, and 20% HCI was added to obtain an acid solution. The mixture was extracted with ether (3x150 mL), the pH of the aqueous phase was adjusted to 9 with concentrated alkali. This aqueous solution was extracted with DCM (2x300 mL), and the combined organic extracts were dried (Na2S04) and evaporated to afford the title compound, 104 g, 94%. Preparation 68
Methyl 1 -benzyl-3-ethylpyrrolidine-3-carboxylate
Figure imgf000110_0001
A solution of methyl 2-ethylacrylate (99.5 g, 1 .18 mol) and /V-benzyl-1 -methoxy- /V-[(trimethylsilyl)methyl]methanamine (337 g, 1 .42 mol) in toluene (1 L) was cooled to -3 C, and a 1 N solution of TFA in DCM (1 18 mL, 1 18 mmol) was added drop wise under stirring. The reaction mixture was stirred under cooling for 40 min and then at room temperature for a further 18 hrs. The mixture was washed with saturated NaHC03 solution and brine, dried (MgS04) and evaporated under reduced pressure. The residue was distilled (bp 145°C at 3 mmHg) to afford the title compound, 1 10 g, 43%. Preparation 69
((3S,4S)-4-(Trifluoromethyl)pyrrolidin-3-yl)methanol
Figure imgf000110_0002
A solution of benzyl (3S,4S)-3-(hydroxymethyl)-4-(trifluoromethyl)pyrrolidine-1 - carboxylate (Preparation 70, 5.6 g, 18.47 mmol) in EtOH (75 mL) was degassed using Ar(g) and 20% Pd(OH)2 (1 .81 g, 12.93 mmol) was added. The reaction mixture was hydrogenated at rt for 16 hrs and then filtered through Celite® washing through with 0.1 % aq. NH3 in EtOH (300 mL). The filtrate was evaporated under reduced pressure to afford the title compound as a brown gel, 3.0 g, 96%. 1H NMR (400 MHz, CDCI3): δ 2.37-2.46 (m, 1 H), 2.64-2.71 (m, 2H), 2.90 (dd, 1 H), 3.04 (dd, 1 H), 3.12 (dd, 1 H), 3.17- 3.22 (m, 1 H), 3.61 (dd, 1 H), 3.72 (dd, 1 H). a[D] 25 0 = -44.4° (c = 1 .00, MeOH)
Preparation 70
Figure imgf000110_0003
NaHC03 (29.79 g, 354.72 mmol) was added portion wise to a stirred solution of ((3S,4S)-re/-4-(trifluoromethyl)pyrrolidin-3-yl)methanol {Bioorg. Med. Chem. Lett. 1998, 8, 2833) (12.0 g, 70.94 mmol) in DCM:H20 (396 mL, 3:2) at rt. The mixture was cooled to 0-5 °C and CBz-CI (1 1.91 mL, 70.94 mmol) was added drop wise and the resulting reaction was stirred at rt for 16 hrs. The reaction was quenched with water and extracted with DCM (300 mL). The combined organic extracts were washed with water, dried (Na2S04) and concentrated under reduced pressure. The crude product was purified by column chromatography on silica gel eluting with EtOAC:hexane, (30:70) to afford a racemic mixture, 17.4 g. This was separated by chiral prep. SFC using a CHIRALPAK AD-H (250x21 mm) column, mobile phase: C02: [MeCN:MeOH (1 : 1 )] = 80:20, and a total flow of 45 g/min to afford the title compound, 7.6 g. RT = 3.56 min; 1H NMR (400 MHz, DMSO-d6): δ 2.40-2.53 (m, 1 H), 3.02-3.08 (m, 1 H), 3.28-3.72 (m, 6H), 4.89-5.07 (m, 3H), 7.32-7.40 (m, 5H). LCMS m/z = 321 [M+H]+
Preparation 71
(3R.5S)-rel- 5-Methylpiperidin-3-ol hydrochloride
Figure imgf000111_0001
HCI in dioxane (4M, 40 mL) was added to a stirred solution of fe/f-butyl (3S,5R)- re/-hydroxy-5-methylpiperidine-1 -carboxylate (Preparation 72, 3.5 g, 16.26 mmol) in DCM (40 mL) at 0° C and the reaction was stirred at room temperature for 16 hrs. The mixture was concentrated in vacuo and the resulting solid triturated with diethyl ether and MeOH to afford the title compound as a white solid, 2.2 g, 89%. 1H NMR (400 MHz, DMSO-de): δ 0.85 (d, 3H), 1.01 (q, 1 H), 1 .77-1 .95 (m, 2H), 2.28-2.40 (m, 2H), 3.00-3.12 (m, 1 H), 3.13-3.25 (m, 1 H), 3.70-3.80 (m, 1 H), 5.32 (d, 1 H), 9.09 (br s, 2H). LCMS m/z = 1 16 [M+H]+ Preparation 72
Figure imgf000111_0002
A saturated solution of HCI in EtOAc (80 mL) was added to a solution of 5- methyl-3-pyridinol (10.0 g, 91 .6 mmol) in EtOAc (15 mL) and MeOH (5 mL) and the mixture stirred for 4 hr at 23 °C. The resulting solid was filtered off and washed with EtOAc and dried under vacuum. The solid was dissolved in acetic acid (50 ml_), 10% Pt02 (1 .0 g) was added and the resulting reaction mixture was hydrogenated in a Parr autoclave (pressure 200 psi) for 16 hrs at 50 °C. The cooled mixture was filtered through Celite® and the filtrate was concentrated to afford a brown gum, 5.0 g.
Boc-anhydride (10.79 ml_, 49.45 mmol) was added drop wise to an ice-cooled stirred solution of the gum in 50% EtOAc/water (100 ml) and Na2C03 (10.48 g, 98.89 mmol) and the resulting reaction was stirred at rt for 15 hrs. The mixture was extracted with EtOAc, the combined organic extracts washed with brine, dried (Na2SO4) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel eluting with EtOAc: Hexane (20:80) to afford the title compound as a yellow liquid, 3.5 g, 49%. 1H NMR (400 MHz, DMSO-d6): δ 0.80-0.91 (m, 4H), 1 .38 (s, 9H), 1 .44-1 .47 (m, 1 H), 1 .86-1 .90 (m, 1 H), 2.05-2.32 (m, 2H), 3.28-3.34 (m, 1 H), 3.75-3.86 (m, 1 H), 3.94-4.03 (m, 1 H), 4.91 (d, 1 H).
Preparation 73
Figure imgf000112_0001
Δ
Pd(OH)2/C (1 .0 g) was added to a solution of (3R,5R)-re/-1 -benzyl-3-(benzyloxy)- 5-cyclopropylpiperidine (Preparation 74, 4.0 g, 15.85 mmol) in MeOH (150 ml_), and the mixture stirred under 50 Psi H2 at 60 °C for 18 hrs. The mixture was filtered and the filtrate concentrated in vacuo to afford the title compound, 1 .2 g, 67%. 1H NMR (400 MHz, MeOH-d4): δ 0.12-0.14 (m, 2H), 0.40-0.46 (m, 2H), 0.47-0.58 (m, 1 H), 0.70-0.83 (m, 1 H), 1 .15 (q, 1 H), 2.1 1 -2.18 (m, 1 H), 2.22-2.35 (m, 2H), 3.02 (dd, 1 H), 3.09 (dd, 1 H), 3.48-3.58 (m, 1 H). LCMS m/z = 142 [M+H]+
Preparation 74
Figure imgf000112_0002
Δ
PtO2 (0.90 g, 3.93 mmol) was added to a solution of 1 -benzyl-3-(benzyloxy)-5- cyclopropylpyridinium bromide (Preparation 77, 15.6 g, 39.37 mmol) and Et3N (7.1 ml_, 51 .17 mmol) in MeOH (360 mL) and the mixture was stirred under H2 at 50 Psi for 6 hrs. The mixture was filtered and the filtrate was concentrated in vacuo. The crude was purified by column chromatography on silica gel eluting with pet. Ether: EtOAc (96:4 to 66:34) to afford the title compound as an oil, 4.5 g, 35% and the trans isomer as an oil, 0.9 g, 7%. 1H NMR (400 MHz, MeOH-d4): δ 0.03-0.1 1 (m, 2H), 0.37-0.52 (m, 3H), 0.74- 0.86 (m, 1 H), 1 .07 (q, 1 H), 1 .72-1.83 (m, 2H), 2.19-2.28 (m, 1 H), 2.93 (dd, 1 H), 3.13 (dd, 1 H), 3.42-3.62 (m, 3H), 4.54 (q, 2H), 7.20-7.40 (m, 10H).
Preparation 75
(3 5R)-re/-5-lsopropylpiperidin-3-ol
Figure imgf000113_0001
Pd/C (1 .0g) was added to (3R,5R)-re/-1 -benzyl-5-isopropylpiperidin-3-ol (Preparation 76, 4.0 g, 15.85 mmol) in MeOH (150 mL), and the mixture stirred under 50 Psi H2 at rt for 18 hrs. The mixture was filtered and the filtrate concentrated in vacuo to give the title compound as an oil, 2.3 g, 95%. 1H NMR (400 MHz, MeOH-d ): 0.94 (d, 6H), 0.97 (q, 1 H), 1 .28-1.39 (m, 1 H), 1 .42-1 .53 (m, 1 H), 2.00-2.32 (m, 3H), 2.97 (d, 1 H), 3.09 (dd, 1 H), 3.50-3.61 (m, 1 H). LCMS m/z = 144 [M+H]+
Preparation 76
(3R5R)-re/-1 -Benzyl-5-isopropylpiperidin-3-ol
Figure imgf000113_0002
PtO2 (2.1 g, 9.40 mmol) was added to a solution of 1 -benzyl-3-hydroxy-5-
(propan-2-yl)pyridinium bromide (Preparation 78, 29 g, 94.09 mmol) and Et3N (1 1.76 mL, 122.31 mmol) in MeOH (600 mL) and the mixture stirred under H2 at 50 Psi for 6 hrs. PtO2 was removed by filtration, and the filtrate was concentrated in vacuo. The crude product was purified by column chromatography on silica gel eluting with Pet. ether: EtOAc, (96:4 to 66:34) to afford the title compound as an oil, 7.4 g, 34%. Further elution provided the trans isomer as an oil, 3.7 g, 17%. 1H NMR (400 MHz, CDCI3): δ 0.80-0.93 (m, 7H), 1 .37-1 .50 (m, 1 H), 1 .60-1 .72 (m, 2H), 1 .97-2.09 (m, 1 H), 2.21 (s, 1 H), 2.84 (d, 1 H), 2.98 (dd, 1 H), 3.40-3.63 (m, 2H), 3.65-3.76 (m, 1 H), 7.20-7.40 (m, 5H). LCMS m/z = 234 [M+H]+ Preparation 77
Figure imgf000114_0001
To a solution of 3-(benzyloxy)-5-cyclopropylpyridine (Preparation 79, 7.80 g, 34.62 mmol) in MeCN (250 mL) was added benzyl bromide (5.92 g, 34.62 mmol) and the mixture stirred at 70-80 °C for 12 hrs. The reaction solution was concentrated in vacuo to afford the title compound, 13.70 g, 99%. LCMS m/z = 316 [M+H]+
Preparation 78
1 -benzyl-3-hvdro -5-(propan-2-yl)pyridinium bromide
Figure imgf000114_0002
Benzyl bromide (13.7 g, 80.19 mmol) was added to a solution of 5- isopropylpyridin-3-ol (1 1 .0 g, 80.19 mmol) in MeCN (300 mL) and the mixture heated at 70-80 °C for 6 hrs. The mixture was evaporated under reduced pressure to afford the title compound, 24.7 g, 99%. LCMS m/z = 228 [M+H]+
Preparation 79
3-(Benzyloxy)-5-cvclopropylpyridine
Figure imgf000114_0003
To a solution of 3-(benzyloxy)-5-bromopyridine (12.7 g, 48.08 mmol) in dioxane:H20 (4: 1 , v/v, 300 mL) were added cyclopropylboronic acid (8.26 g, 96.17 mmol), Na2C03 (10.2 g, 96.17 mmol) and Pd(dppf)CI2 (1 .5 g, 2.05 mmol) under N2. The mixture was stirred at 80-100 °C for 3 days. The cooled reaction was extracted with DCM (400mL x3), and the combined organic extracts washed with aq.NaHCOs (150 mL x2) and brine (100 mL), dried (Na2S04), and concentrated in vacuo. The crude product was purified by column chromatography on silica gel eluting with Pet. ether: EtOAc, from (100: 1 to 90: 10) to afford the title compound as a solid, 9.2 g, 84%. LCMS m/z = 226 [M+H]+ Preparation 80
Figure imgf000115_0001
((3aR,7aR)-rel-Benzyloctahydro-3aH-isoindol-3a-yl)methanol (Preparation 81 , 69 g, 0.282 mol) was dissolved in MeOH (500 mL), Pd(OH)2 (14 g) was added and the reaction stirred at 45 °C under an atmosphere of H2 for 18 hrs. The mixture was filtered and the filtrate concentrated in vacuo to afford the title compound as a yellow oil, 41 g, 94%. 1H NMR (300 MHz, CDCI3): δ 1 .20-1 .65 (m, 8H), 1 .87-2.00 (m, 1 H), 2.84-2.96 (m, 2H), 3.13-3.25 (m, 1 H), 3.40-3.60 (m, 2H), 3.70-3.82 (m, 3H). Preparation 81
((3aR,7aR)-re/-Benzyloctahvdro-3aH-isoindol-3a-yl)methanol
Figure imgf000115_0002
Ethyl (3aR,7aR)-rel-benzyloctahydro-3aH-isoindole-3a-carboxylate (Preparation 82, 128.7 g, 0.471 mol) was dissolved in THF (500 mL), the solution cooled in ice and LiAIH4 (18 g, 0.471 mol) added portion wise and the reaction stirred for 2 hrs. NaOH solution was added slowly until no further bubbles formed, then the mixture was filtered and concentrated in vacuo. The crude was purified by column chromatography on silica gel to afford the title compound as a yellow oil, 69 g 60%.
Preparation 82
Ethyl (3aR,7aR)-rel-benzyloctahvdro-3aH-isoindole-3a-carboxylate
Figure imgf000115_0003
Ethyl cyclohex-1 -ene-1 -carboxylate (1 10 g, 0.786 mol) and N-(methoxymethyl)- N-(trimethylsilylmethyl)benzylamine (175.2 g, 0.786 mol) were dissolved in DCM (300 mL) and a solution of acetic acid (8.960 g, 0.079 mol) in DCM (50 mL) was added drop wise over a period for 30 mins with stirring. The reaction was stirred for 3 hrs then quenched with water, the layers separated and the organic layer dried and concentrated in vacuo. The crude was purified by column chromatography on silica gel to afford the title compound as a yellow oil, 128.7 g, 60%. Preparation 83
r(3R,4R)-re/-3,4-Dimethylpyrrolidin-3-yl1methanol
Figure imgf000116_0001
A mixture of ((3R,4R)-re/-1 -benzyl-3,4-dimethylpyrrolidin-3-yl)methanol
(Preparation 84, 68 g, 0.31 mol), ammonium formate (63 g, 0.93 mol) and 10% Pd/C (9 g) in MeOH (1 L) was stirred at rt for 18 hrs. The mixture was filtered, the filtrate evaporated, and the residue was distilled (bp 90-95 °C at 2-5 mmHg) to afford the title compound as an oil, 26 g, 65%. 1H NMR (400 MHz, DMSO-d6): δ 0.77 (s, 3H), 0.79 (d, 3H), 1 .69-1.77 (m, 1 H), 2.28-2.36 (m, 2H), 2.78 (d, 1 H), 2.97 (dd, 1 H), 3.17 (s, 2H), 3.67 (br s, 2H). GCMS: 129 [M]
Preparation 84
((3R.4R)-re/-1 -Benzyl-3.4-dimethylpyrrolidin-3-yl)methanol
Figure imgf000116_0002
A solution of methyl (3R,4R)-re/-1 -benzyl-3,4-dimethylpyrrolidine-3-carboxylate (Preparation 85, 81 g, 0.327 mol) in THF (100 mL) was added at -3 °C to a
suspension of LiAIH4 (24.8 g, 0.655 mol) in THF (1 .2 L). The reaction mixture was heated to room temperature over a period of 30 min and then refluxed for 1 hr. Then the mixture was cooled, quenched by the addition of water (45 mL), 15% NaOH (45 mL) and water (135 mL), filtered, washed with ether (3x200 mL) and evaporated to give the title product, 68 g, 95%.
Preparation 85
Methyl (3R,4R)-re/-1 -benzyl-3,4-dimethylpyrrolidine-3-carboxylate
Figure imgf000116_0003
A solution of methyl 2-methylbut-2-enoate (70 g, 0.61 mol) and /V-benzyl-1 - methoxy-/V-[(trimethylsilyl)methyl]methanamine (175 g, 0.74 mol) in toluene (1 L) was cooled to 0 °C, and 1 N solution of trifluoroacetic acid in DCM (61 mL) was added drop wise under stirring. The reaction mixture was stirred under cooling for 40 min and then at rt for a further 18 hrs. The mixture was washed with saturated NaHC03 solution, then brine, dried (MgS04) and evaporated under reduced pressure. The residue was distilled (bp 123-125 °C at 0.3-0.4 mmHg) to afford the title compound, 81 g, 53%.
Preparation 86
Ethyl 2-{[3-(pyrazin-2-yl)oxetan-3-yl1amino)pyrimidine-5-carboxylate
Figure imgf000117_0001
To a solution of 3-(pyrazin-2-yl)oxetan-3-amine (Preparation 31 , 213 mg, 1 .41 mmol) and DIPEA (304 mg, 2.35 mmol) in dioxane (10 mL) was added ethyl 2- fluoropyrimidine-5-carboxylate (Preparation 21 , 200 mg, 1 .18 mmol) and the reaction was stirred at 100 °C for 4 hrs. The cooled reaction was diluted with EtOAc (50 mL) washed with brine (30 mL x3), dried (Na2S04), filtered and the filtrate evaporated under reduced pressure. The residue was purified by column chromatography on silica gel eluting with pet. ether: EtOAc (50:50 to 100:0) to afford the title compound as a yellow solid, 158 mg, 44%. 1 H NMR (400 MHz, MeOD-d4) : δ 1 .33 (t, 3H), 4.31 (q, 2H), 4.99 (d, 2H), 5.15 (d, 2H), 8.50 (d, 1 H), 8.61 (br s, 1 H), 8.63 (d, 1 H), 8.70 (dd, 1 H), 8.87 (br s, 1 H). LCMS m/z = 302 [M+H]+
Preparation 87 Ethyl 2-{[3-(pyrimidin-5-yl)oxetan-3-yl1amino)pyrimidine-5-carboxylate
Figure imgf000117_0002
To a solution of 3-(pyrimidin-5-yl)oxetan-3-amine (Preparation 32, 437 mg, 2.89 mmol) and DIPEA (622 mg, 4.81 mmol) in dioxane (20 mL) was added ethyl 2-chloro- pyrimidine-5-carboxylate (400 mg, 2.41 mmol) and the reaction stirred at 100 °C for 4 hrs. The cooled mixture was diluted with EtOAc (100 mL), washed with brine (30 mL x3), dried (Na2SO4), filtered and the filtrate evaporated under reduced pressure. The residue was purified by column chromatography eluting with pet. Ether: EtOAc (50:50 to 0: 100) to afford the title compound as a yellow oil, 100 mg, 14%. 1H NMR (400 MHz, CDCIs): δ 1 .36 (t, 3H), 4.35 (q, 2H), 4.97-5.03 (m, 2H), 5.06 (d, 2H), 6.52 (s, 1 H), 8.85 (br s, 2H), 8.95 (s, 2H), 9.18 (s, 1 H). LCMS m/z = 302 [M+H]+
Preparation 88
2-{[2-(pyrazin-2-yl)propan-2-yl1amino)pyrimidine-5-carboxylic acid
Figure imgf000118_0001
Water (2 ml_) and LiOH H20 (161 mg, 6.66 mmol) was added to a mixture of ethyl 2-{[2-(pyrazin-2-yl)propan-2-yl]amino}pyrimidine-5-carboxylate (Preparation 15, 736 mg, 2.56 mmol) in THF (10 ml_) and the reaction was stirred at ambient temperature for 2 hrs. The mixture was concentrated to 1/3 of the original volume and 1 N HCI (6.66 ml_, 6.66 mmol) slowly added. The resulting solids were filtered and washed with water. The solids were transferred to a round bottom flask and MeCN (5 ml_) added and removed under reduced pressure twice. The resulting solids were dried to give afford the title compound as a tan solid, 571 mg, 86%. 1H NMR (400 MHz, DMSO-de): δ 1 .72 (s, 6H), 8.36-8.83 (m, 6H), 12.82 (br s, 1 H). LCMS m/z = 260 [M+H]+
Preparation 89
2-aminopyrimidin-5-yl)(8-oxa-2-azaspiro[4.51dec-2-yl)methanone
Figure imgf000118_0002
A suspension of (2-chloropyrimidin-5-yl)(8-oxa-2-azaspiro[4.5]dec-2- yl)methanone (Preparation 22, 200 mg, 0.710 mmol) in ammonium hydroxide solution (1 .42 ml_, c=0.5 M) was subjected to microwave irradiation at 120 °C for 30 minutes. The mixture was poured into brine and the pH was adjusted to ~ 7 by the addition of 6N HCI. The mixture was extracted with CHCI3/IPA (3: 1 , 10x). The organic extracts were dried over anhydrous sodium sulfate and concentrated under reduced pressure to give the desired material which was used without further purification, 171 mg, 92%. 1H NMR (400 MHz, CDCI3): δ 1 .52-1.72 (m, 4H), 1 .85-1 .94 (m, 2H), 3.40-3.83 (m, 8H), 5.28-5.40 (br s, 2H), 8.57 (s, 2H). LCMS m/z = 263 [M+H]+ EXAMPLES
Examples 1 to 32 were prepared in a library through an amide coupling of 2- [(pyridin-3-ylmethyl)amino]pyrimidine-5-carboxylic acid (Preparation 1 ) and 32 different amines or common amine salts using the reaction protocol described below.
Figure imgf000119_0001
1 . 2-[(Pyridin-3-ylmethyl)amino]pyrimidine-5-carboxylic acid (Preparation 1 , 23.0 mg, 100 μηιοΙ, 1 .0 eq.) was dispensed into 8 ml_ vials.
2. HATU (45.6 mg, 120 pmol, 1 .2 eq.) was dispensed into the above vials.
3. DMF (1 ml_) was dispensed into the above vials.
4. The vials were capped and shaken at 50 °C for 2 hrs.
5. The selected amine (120 pmol, 1 .2 eq.) was dispensed into the above vials.
6. Et3N (50 μΙ, 345 pmol, 3.45 eq.) was added into the above vial.
7. The vials were capped and shaken at 50 °C for 18 hrs.
8. Solvent was evaporated using a Speedvac.
9. The residues were purified by preparative HPLC using a Phenomenex Gemini C18 250 x 21 .2 mm*10Mm column, eluting with MeCN: aq NH4OH at an appropriate gradient of between 0 and 60% over up to 10 minutes to afford the title compounds.
Figure imgf000119_0002
Figure imgf000120_0001
ylmethyl)amino]pyrimidin-5-yl}methanone
Figure imgf000121_0001
Figure imgf000122_0001
[(pyndin-3-ylmethyl)amino]pyrimidin-5-yl}methanone
Figure imgf000123_0001
yl}carbonyl)piperidin-3-yl]acetamide CD05
Figure imgf000124_0001
yl}carbonyl)piperidine-4-carExampleamide
Figure imgf000125_0001
All amine starting materials are commercially available, with the exception of:
a (3aR,4R,7aS)-re/-octahydro-1 H-isoindol hydrochloride (Preparation 56) b (3-ethylpyrrolidin-3-yl)methanol (Preparation 66) Examples 33 to 109 were prepared in a library through an amide coupling of 2-
{[1 -(pyrimidin-5-yl)cyclopropyl]amino}pyrimidine-5-carboxylic acid (Preparation 5) and 77 different amines or common amine salts, using the reaction protocol described below.
Figure imgf000125_0002
1 . 2-{[1 -(Pyrimidin-5-yl)cyclopropyl]amino}pyrimidine-5-carboxylic acid (0.2 M solution in DMF, 120 μηιοΙ, 1.2 eq.) was dispensed into 8 mL vials.
2. The selected amine (R1NHR2) (100 μηιοΙ, 1 .0 eq.) was dispensed into each vial. 3. HATU (120 μηποΙ, 1.2 eq., 0.24 M solution in DMF) was added to each vial.
4. DIPEA (70 μΙ, 400 μηποΙ, 4.0 eq.) was added to each vial.
5. The vials were capped and shaken at 50 °C for 16 hrs.
6. The solvent was evaporated on Speedvac.
7. The residues were purified by preparative HPLC using the purification methods (PM) described in the table below and an appropriate solvent gradient, to provide the title compounds
Figure imgf000126_0001
Figure imgf000126_0002
Figure imgf000127_0001
yl)cyclopropyl]amino}pynmidin-5-yl)methanone
Figure imgf000128_0001
Figure imgf000129_0001
Figure imgf000130_0001
yl)cyclopropyl]amino}pyrimidin-5-yl)methanone
Figure imgf000131_0001
yl)methanone
Figure imgf000132_0001
yl)[4-(trifluoromethyl)pipendin-1 -yl]methanone
Figure imgf000133_0001
yl)cyclopropyl]amino}pyrimidin-5-yl)methanone
Figure imgf000134_0001
yl)[2-(trifluoromethyl)pyrrolidin-1 -yl]methanone
Figure imgf000135_0001
yl)methanone
Figure imgf000136_0001
yl)methanone
Figure imgf000137_0001
5-yl)methanone
Figure imgf000138_0001
yl)cyclopropyl]amino}pyrimidin-5-yl)methanone
Figure imgf000139_0001
yl)cyclopropyl]amino}pynmidin-5-yl)methanone
Figure imgf000140_0001
yl)cyclopropyl]amino}pyrim idin-5-yl)methanone
Figure imgf000141_0001
yl)methanone
Figure imgf000142_0001
a (3R,5R)-re/-5-isopropylpiperidin-3-ol (Preparation 75)
b 3-(pyrrolidin-3-yl)-1 H-pyrazole hydrochloride (Preparation 59)
c [(3S,4S)-4-(trifluoromethyl)pyrrolidin-3-yl]methanol (Preparation 69)
d (3aR,7aR)-re/-octahydro-3aH-isoindol-3a-ylmethanol (Preparation 80)
e [(3R,4R)-re/-3,4-dimethylpyrrolidin-3-yl]methanol (Preparation 83)
f 3-Methyl-5-(pyrrolidin-3-yl)-1 H-pyrazole hydrochloride (Preparation 60)
9 (3R,5S)-re/-5-methylpiperidin-3-ol hydrochloride (Preparation 71)
h (3R,5R)-re/-5-cyclopropylpiperidin-3-ol hydrochloride (Preparation 73)
Examples 1 10 to 129 were prepared in a library through an amide coupling of 2- [(pyrazin-2-ylmethyl)amino]pyrimidine-5-carboxylic acid (Preparation 3) or 2-{[(1 S)-(1 - (pyrazin-2-yl)ethyl]amino}pyrimidine-5-carboxylic acid (Preparation 4) with 10 different amines using the reaction protocol described below.
Figure imgf000142_0002
1 . 2-[(Pyrazin-2-ylmethyl)amino]pyrimidine-5-carboxylic acid (140 μηιοΙ, 1 .0 eq.) was added to 10 separate 8 ml_ reaction vials.
2. 2-{[(1 S)-(1 -(pyrazin-2-yl)ethyl]amino}pyrimidine-5-carboxylic acid (140 μίτιοΙ, 1.0 eq.) was added to 10 separate 8 ml_ reaction vials.
3. The selected amine (R1NHR2) (168 μηιοΙ, 1 .2 eq.) was dispensed into each vial. 4. HATU (53.2 mg, 140 μιτηοΙ, 1 .0 eq.) was added to each vial.
5. DMA (1400 μΙ_) was added to each vial.
6. DIPEA (-73 μΙ_, 420 μηποΙ, 3.0 eq.) was added to each vial.
7. The vials were capped and shaken at 50 °C for 16 hrs.
8. The solvent was evaporated by Speedvac.
9. The residues were purified by preparative HPLC using the columns described in the table below and an appropriate solvent gradient and flow rate, to provide the title compounds
Figure imgf000143_0001
Figure imgf000144_0001
Figure imgf000145_0001
(pyrazin-2-yl)ethyl]amino}pyrimidin-5-yl)methanone 354 [M+H]+
Figure imgf000146_0001
5-yl)methanone
Figure imgf000147_0001
Example 130
8-oxa-2-azaspiro[4.51dec-2-yl(2-{[1 -(pyrazin-2-yl)cvclobutyl1amino)pyrimidin-5- vDmethanone
Figure imgf000147_0002
Ethyl 2-{[(1 -(pyrazin-2-yl)cyclobutyl]amino}pyrimidine-5-carboxylate (Preparation 14, 2.1 g, 7.016 mmol), 8-oxa-2-azaspiro[4.5]decane (1 .49 g, 10.5 mmol) and TBD (1 .95 g, 14.0 mmol) were dissolved in DMF (40 mL) and the resulting mixture was stirred at 50 °C for 18 hrs. The reaction mixture was concentrated under reduced pressure to give a residue which was partially purified by column chromatography on silica gel eluting with petroleum ether: EtOAc (100:0 to 0: 100) to give a yellow oil. The product was isolated using preparative HPLC using a Phenomenex Gemini C18 250*50 10μ column, eluting with water (0.05% ammonium hydroxide):MeCN (10 to 34%) over 21 mins at a flow rate of 120 mL/min, to afford the title compound as a white solid (1 .48 g, 53%).
1H NMR (400 MHz, CDCI3): δ 1 .54-1 .62 (m, 4H), 1 .80-1 .92 (m, 2H), 2.07-2.16 (m, 1 H), 2.18-2.28 (m, 1 H), 2.43-2.54 (m, 2H), 2.85-2.95 (m, 2H), 3.39 (br s, 1 H), 3.53 (br s, 1 H), 3.58-3.80 (m, 6H), 6.28 (br s, 1 H), 8.42 (d, 1 H), 8.47 (br s, 2H), 8.57 (s, 1 H), 8.73 (br s, 1 H). LCMS m/z = 395 [M+H]+
Example 131 7-oxa-2-azaspiro[3.51non-2-yl(2-{[2-(pyrazin-2-yl)propan-2-yl1amino)pyrimidin-5- vQmethanone
Figure imgf000148_0001
Ethyl 2-{[2-(pyrazin-2-yl)propan-2-yl]amino}pyrimidine-5-carboxylate (Preparation 15, 2.15 g, 7.48 mmol), 7-oxa-2-azaspiro[3.5]nonane (1 .43 g, 1 1 .2 mmol) and TBD (2.08 g, 15.0 mmol) were dissolved in DMF (40 mL) and the resulting mixture was stirred at 50 °C for 16 hrs. The reaction mixture was concentrated under reduced pressure to give a residue which was partially purified by column chromatography on silica gel eluting with petroleum ether:EtOAc (100:0 to 0: 100) then EtOAC:MeOH(100:0 to 85: 15) to give a yellow oil. The product was isolated using preparative HPLC using a Phenomenex Gemini C18 250*50 10μ column, eluting with water (0.05% ammonium hydroxide): MeCN (10 to 31 %) over 20 mins at a flow rate of 120 mL/min, to afford the title compound as a white solid (1.74 g, 63%). 1H NMR (400 MHz, CDCI3): δ 1 .79 (dd, 4H), 1 .84 (s, 6H), 3.62 (dd, 4H), 3.85-4.09 (m, 4H), 6.42 (br s, 1 H), 8.43 (d, 1 H), 8.45- 8.59 (m, 3H), 8.73 (s, 1 H). LCMS m/z = 369 [M+H]+
The following Examples 132-142 were prepared according to the general synthetic schemes and methods outlined above for Example 130, using the appropriate ester and amine, in a transamidation reaction.
Example Structure and name Starting ester; Yield; Data
132 ethyl 2-{[3-(pyrazin-2-yl)oxetan-3- yl]am ino}pyrim idine-5-carboxylate;
H (Preparation 86); 62 mg, 32%
1H NMR (400 MHz, MeOD-d4): δ 1 .50-1.72
8-oxa-2-azaspiro[4.5]dec-2- (m, 4H), 1 .86-1 .97 (m, 2H), 3.49 (s, 2H), yl(2-{[3-(pyrazin-2- 3.55-3.80 (m, 6H), 4.97-5.04 (m, 2H), 5.17 yl)oxetan-3- (d, 2H), 8.40-8.80 (m, 5H). LCMS m/z = 397 yl]amino}pyrimidin-5- [M+H+]
yl)methanone
133 ethyl 2-{[3-(pyrimidin-5-yl)oxetan-3- yl]am ino}pyrim idine-5-carboxylate;
V H (Preparation 87); 13 mg, 24%
8-oxa-2-azaspiro[4.5]dec-2-
1H NMR (400 MHz, MeOD-d4): δ 1 .50-1.68 yl(2-{[3-(pyrimidin-5- (m, 4H), 1 .83-1 .96 (m, 2H), 3.48 (s, 2H), yl)oxetan-3- 3.55-3.80 (m, 6H), 4.90-5.00 (m, 4H), 5.04- yl]amino}pyrimidin-5- 5.1 1 (m, 2H), 8.51 (br s, 2H), 9.00 (d, 2H), yl)methanone
9.09 (s, 1 H). LCMS m/z = 397 [M+H+]
134 ethyl 2-{[3-(pyrimidin-5-yl)oxetan-3- yl]am ino}pyrim idine-5-carboxylate;
(Preparation 87); 8 mg, 14%
Figure imgf000149_0001
1H NMR (400MHz, MeOD-d4): δ 1 .77-1.82
7-oxa-2-azaspiro[3.5]non-2- (m, 4H), 3.56-3.68 (m, 4H), 3.87 (s, 2H), yl(2-{[3-(pyrimidin-5- 4.16 (s, 2H), 4.96 (d, 2H), 5.06 (d, 2H), yl)oxetan-3- 8.43-8.79 (br m, 2H), 8.99 (s, 2H), 9.09 (s, yl]amino}pyrimidin-5- 1 H). LCMS m/z = 383 [M+H]+
yl)methanone
Figure imgf000150_0001
138 Ethyl 2-{[1-(pyrazin-2- yl)cyclobutyl]amino}pyrimidine-5-
Figure imgf000151_0001
carboxylate; (Preparation 14); 298 mg,
7-oxa-2-azaspiro[3.5]non-2- 65%.
yl(2-{[1-(pyrazin-2- 1H NMR (400 MHz, DMSO-d6): δ 1.60-1.72 yl)cyclobutyl]am ino}pyrim idi (m, 4H), 1.90-2.10 (m, 2H), 2.40-2.55 (m, n-5-yl)methanone 2H), 2.66-2.77 (m, 2H), 3.30-3.55 (m, 4H),
3.71 (s, 2H), 4.10 (s, 2H), 8.37 (br s, 1H), 8.47 (d, 1H), 8.56 (d, 1H), 8.63 (br s, 2H), 8.80 (s, 1H). LCMS m/z = 381 [M+H]+
139 Ethyl 4-methyl-2-[(pyrazine-2- ylmethyl)amino]pyrimidine-5-carboxylate; (Preparation 20); 17 mg, 14%
Figure imgf000151_0002
1H NMR (400 MHz, CDCI3): δ 1.49-1.58 (m,
{4-methyl-2-[(pyrazin-2- 2H), 1.60-1.69 (m, 2H), 1.81-1.95 (m, 2H), ylmethyl)amino]pyrimidin-5- 2.37 (s, 3H), 3.16 (s, 1H), 3.34-3.40 (m, 1H), yl}(8-oxa-2- 3.55-3.80 (m, 6H), 4.79-4.86 (m, 2H), 6.11- azaspiro[4.5]dec-2- 6.19 (m, 1H), 8.17 (d, 1H), 8.48 (s, 1H), 8.53 yl)methanone
(s, 1H), 8.66 (d, 1H). LCMS m/z = 369 [M+H]+
140 Ethyl 2-{[(1 S)-1 -(6-methylpyridin-3- yl)ethyl]amino}pyrimidine-5-carboxylate; (Preparation 17); 4.35 g, 63%
Figure imgf000151_0003
(2-{[(1 S)-1 -(6-methylpyridin- 1H NMR (400 MHz, CDCI3): δ 1.45-1.65 (m,
3-yl)ethyl]amino}pyrimidin- 7H), 1.81-1.92 (m, 2H), 2.52 (s, 3H), 3.35-
5-yl)(8-oxa-2- 3.80 (m, 8H), 5.18-5.26 (m, 1H), 5.62-5.69 azaspiro[4.5]dec-2- (m, 1H), 7.11 (d, 1H), 7.56 (dd, 1H), 8.43- yl)methanone 8.60 (m, 3H). LCMS m/z = 382 [M+H]+ 141 o Ethyl 2-{[(1 R)-1 -(6-methylpyridin-3- o yl)ethyl]amino}pyrimidine-5-carboxylate;
(Preparation 18); 1 .65 g, 56%
(2-{[(1 R)-1 -(6-methylpyridin- Ή NMR (400 MHz, CDCI3): δ 1 .52-1 .68 (m,
3-yl)ethyl]amino}pyrimidin- 7H), 1 .81 -1 .93 (m, 2H), 2.54 (s, 3H), 3.39-
5-yl)(8-oxa-2- 3.83 (m, 8H), 5.17-5.27 (m, 1 H), 5.61 -5.67 azaspiro[4.5]dec-2- (m, 1 H), 7.13 (d, 1 H), 7.54-7.60 (m, 1 H), yl)methanone 8.52-8.56 (m, 3H). LCMS m/z = 382 [M+H]+
Example 142
8-oxa-2-azaspiro[4.51dec-2-yl{2-[(pyrazin-2-ylmethyl)aminolpyrimidin-5-yl)methanone
Figure imgf000152_0001
A 200-mL flask was charged with (2-chloropyrimidin-5-yl)(8-oxa-2- azaspiro[4.5]dec-2-yl)methanone (Preparation 22,. 4.0 g 13.92 mmol), potassium carbonate (2.33 g, 16.7 mmol, 325 mesh), 1 -(pyrazin-2-yl)methanamine (CAS# 20010- 99-5, 1 .65 g, 14.7 mmol) at 25 °C. An isopropanol/water solution (40 mL, 99: 1 v/v) was added to the mixture which was heated at 80 °C for 2h. The mixture was cooled down to 45 °C, then acetone (80 mL) was added and the stirring was continued for 1 h at 45 °C. the mixture was cooled down to 40 °C and filtered with a Buchner funnel and filter paper. The cake was washed with acetone (12 mL) and the combined filtrate was transferred in a 500-mL round bottom flask. The solvent was evaporated at 65 °C under reduced pressure while feeding the solution with isopropanol (80 mL), until a final volume of ~40 mL was obtained. Water (0.2 mL) was added to the solution which was slowly cooled from 65 °C to 2 °C over at least 10.5 h (~ -0.1 C/min rate) and maintained at 2 °C for an extra 2 h period. The resulting slurry was filtered on a Buchner funnel with filter paper and isopropanol (12 mL) was used to rinse the flask and wash the cake. The off-white solid was dried under vacuum for 1 h then in a vacuum oven (house vacuum, 40 °C) for 2 h, affording the title compound, 4.25 g, 86%. 1H NMR (400 MHz, DMSO- de): δ 1 .38-1 .58 (m, 4H), 1 .74-1 .82 (m, 2H), 3.34 (br s, 1 H), 3.44 (br s, 1 H), 3.47-3.65 (m, 6H), 4.68 (d, 2H), 8.28 (br s, 1 H), 8.48-8.55 (m, 3H), 8.56-8.62 (m, 2H). LCMS: m/z = 355 [M+H]+ Powder X-Ray Diffraction of Example 142:
The divergence slit was set at 0.6 mm while the secondary optics used variable slits. Diffracted radiation was detected by a PSD-Lynx Eye detector. The X-ray tube voltage and amperage were set to 40 kV and 40 mA respectively. Data was collected in the Theta-2Theta goniometer at the Cu wavelength (k-alpha average) from 3.0 to 40.0 degrees 2-Theta using a step size of 0.019 degrees and a step time of 5 second. Samples were prepared by placing them in a silicon low background sample holder (Bruker part number: C79298A3244B261 ) and rotated during collection. Data were collected using Bruker DIFFRAC Plus software and analysis was performed by EVA diffract plus software.
The PXRD data file was not processed prior to peak searching. Using the peak search algorithm in the EVA software, peaks selected with a threshold value of 5 and a width value of 0.3 were used to make preliminary peak assignments. The output of automated assignments was visually checked to ensure validity and adjustments were manually made if necessary. Peaks with relative intensity of > 10% were generally chosen. The peaks which were not resolved or were consistent with noise were not selected. A typical error associated with the peak position from PXRD stated in USP is within +/- 0.2° 2-Theta (USP-941 ). Table 1 details the PXRD peak list associated with Example 142. Asterisked peak positions represent characteristic peaks.
Table 1 : PXRD peak list for EXAMPLE 142.
Figure imgf000153_0001
23.9 12
26.4 25
28.4 15
28.4 15
Table 2 sets forth the comparison peak data for characteristic peaks from replicate preparations. All values listed are in Angles (2-Theta°). Asterisked peak positions represent characteristic peaks. See Figure 1.
Table 2: Comparison of peak positions of the characteristics peaks
Figure imgf000154_0002
*Rounded values for important peaks are within 0.1 of each other.
Example 143
8-oxa-2-azaspiro[4.51dec-2-yl(2-{[2-(pyrimidin-5-yl)propan-2-yl1amino)pyrimidin-5- vDmethanone
Figure imgf000154_0001
To a solution of 2-(pyrimidin-5-yl)propan-2-amine hydrochloride (Preparation 24, 123 mg, 0.495 mmol) and DIPEA (183 mg, 1 .42 mmol) in NMP (0.3 ml_) was added (2- chloropyrimidin-5-yl)(8-oxa-2-azaspiro[4.5]dec-2-yl)methanone (Preparation 22, 100 mg, 0.355 mmol). The resulting mixture was stirred at 140 °C for 0.5 hr. The cooled reaction was purified directly by prep. HPLC using a Luna C18 150*25 5u column, eluting with 18-38% (0.225% TFA in water):MeCN over 1 1 minutes at a flow rate of 35 mL/min to afford the title compound as a white solid, 21 mg, 15%. 1H NMR (400 MHz, DMSO-de): δ 1 .42-1.55 (m, 4H), 1 .70-1 .80 (m, 8H), 3.36 (s, 2H), 3.45-3.65 (s, 2H), 3.45- 3.60 (m, 6H), 7.80 (br s, 1 H), 8.40 (s, 2H), 8.76 (s, 2H), 8.98 (s, 1 H). LCMS: m/z = 383 [M+H]+
Example 145
8-oxa-2-azaspiro[4.51dec-2-yl(2-{[(1 S)-1 -(pyrazin-2-yl)ethyl1amino)pyrimidin-5- vQmethanone
Figure imgf000155_0001
A mixture of (1 S)-1 -(pyrazin-2-yl)ethanamine hydrochloride (Preparation 27, 529 mg, 4.29 mmol), (2-chloropyrimidin-5-yl)(8-oxa-2-azaspiro[4.5]dec-2-yl)methanone (Preparation 22, 1.1 g, 3.90 mmol), cesium carbonate (2.54 g, 7.81 mmol) and cesium fluoride (1 .78 g, 1 1 .70 mmol) in acetonitrile (50 ml_) was stirred at 80 °C for 16 hours. The resulting suspension was filtered, and the filtrate was evaporated to dryness under reduced pressure. The residue was then purified by column chromatography on silica gel (CH2CI2/MeOH, 100:0 to 80:20), to provide the title compound, 545 mg, 38%. 1H NMR (400 MHz, CDCI3): δ 1.53-1.68 (m, 7H), 1 .82-1 .92 (m, 2H), 3.43 (br s, 1 H), 3.53- 3.81 (m, 7H), 5.33-5.43 (m, 1 H), 6.32 (br s, 1 H), 8.49 (d, 1 H), 8.54-8.57 (m, 3H), 8.66 (br s, 1 H). LCMS: m/z = 369 [M+H]+
Determination of absolute stereochemistry for Example 145a: A single crystal structure of the methanesulfonate salt of Example 145 was obtained (Figure 2). The absolute stereochemistry of Example 145a was determined as the S enantiomer from this crystal structure.
Example 145a
8-oxa-2-azaspiro[4.51dec-2-yl(2-{[(1 S)-1 -(pyrazin-2-yl)ethyl1amino)pyrimidin-5- vQmethanone methanesulfonate
Figure imgf000155_0002
A solution of Example 145 (1 .47g. 3.89 mmol) in ethyl acetate (77.8 ml_) stirred at room temperature was treated with a solution of methane sulfonic acid (CAS# 75-75- 2, 376 mg, 3.89 mmol) in ethyl acetate (266 μΙ_). The resulting mixture was heated at 50 °C for 2 hrs, then slowly cooled down to room temperature, and further stirred at this temperature for 3 days. The slurry was then filtered off, rinsed with ethyl acetate and the solids were dried under vacuum, providing the title material as a white crystalline solid (1 .66 g, 92%). 1 H NMR (400 MHz, CDCI3): δ 1 .55-1 .66 (m, 4H), 1 .75 (d, 3H), 1 .92 (t, 2H), 2.94 (s, 3H), 3.40 (br. s. , 1 H), 3.55 (br. s. , 1 H), 3.61 -3.81 (m, 6H), 5.42-5.51 (m, 1 H), 8.29-8.41 (m, 1 H), 8.53 (d, 1 H), 8.57 (s, 1 H), 8.71 (s, 1 H), 8.95 (br. s. , 1 H), 9.98- 10.20 (m, 1 H). LCMS m/z 369 [M+H]+
Data collection was performed on a Bruker APEX diffractometer at room temperature. Data collection consisted of omega and phi scans. The structure was solved by direct methods using SHELX software suite in the space group P212121 . The structure was subsequently refined by the full-matrix least squares method. All non-hydrogen atoms were found and refined using anisotropic displacement parameters.
The hydrogen atoms located on nitrogen were placed in reasonable constrained positions based on bond lengths. The bond lengths and the pkas of the acid/base support this assignment of the proton (salt).
The remaining hydrogen atoms were placed in calculated positions and were allowed to ride on their carrier atoms. The final refinement included isotropic displacement parameters for all hydrogen atoms.
The final R-index was 6%. A final difference Fourier revealed no missing or misplaced electron density. See Figure 2 for an X-ray crystal structure (ORTEP drawing of Example 145a). Table 3 contains relevant structure data on Example 145a.
Table 3. Crystal data and structure refinement for Example 145a. Empirical formula C20 H28 N6 05 S
Formula weight 464.54
Temperature 296(2) K
Wavelength 1 .54178 A
Crystal system Orthorhombic
Space group P2i2i2i
Unit cell dimensions a = 8.2609(17) A a= 90°.
b = 14.686(3) A b= 90°.
c = 18.670(4) A g = = 90°
Volume 2265.1 (8) A3
Z 4 Density (calculated) 1 .362 Mg/m3
Goodness-of-fit on 0.971
Final R indices [l>2sigma(l)] R1 = 0.0597, wR2 0.1 129
R indices (all data) R1 = 0.1 178, wR2 0.1364
Inferred assignment of absolute stereochemistry for other enantiomeric pairs
Analysis of the absolute structure using likelihood methods (Hooft 2008) was performed using PLATON (Spek 2010). Assuming the sample submitted is enantiopure, the results indicate that the absolute stereochemistry was correctly assigned. Therefore, the single X-Ray of Example 145a is consistent with Example 145 having a "S" absolute configuration. By deduction, Example 146 is the "R" enantiomer of this pair, and it displayed a ~100-fold loss of potency against vanin in the assay. This analysis was used to extrapolate absolute configurations of other enantiomeric pairs of this series: in each case, the most potent enantiomer was assigned with the "S" absolute configuration, based upon the above assumptions. The examples in which absolute stereochemistry was inferred are indicated as such in the description thereof.
Examples 144 and 146-181 , Table 4, were prepared using the method of Example 143, from (2-chloropyrimidin-5-yl)(8-oxa-2-azaspiro[4.5]dec-2-yl)methanone (Preparation 22) or (2-chloropyrimidin-5-yl)(7-oxa-2-azaspiro[4.5]non-2-yl)methanone (Preparation 23) and the appropriate amine upon heating either with or without microwave irradiation.
Table 4. Data for Examples 144 and 146-181
Figure imgf000157_0001
Figure imgf000158_0001
Figure imgf000159_0001
Figure imgf000160_0001
Figure imgf000161_0001
160 o 19 mg, 9%; ηΗ NMR (400 MHz,
DMSO-de): δ 1.51 (d, 3H), 1.65- 1.70 (m, 4H), 2.57 (s, 3H), 3.40-
(2-{[1 -(2-methylpyrimidin-5- 3.60 (m, 4H), 3.73 (s, 2H), 4.11 yl)ethyl]amino}pyrimidin-5-yl)(7-oxa- (s, 2H), 5.10-5.15 (m, 1H), 8.39
2-azaspiro[3.5]non-2-yl)methanone (d, 1H), 8.56 (br s, 2H), 8.69 (s,
2H). LCMS: m/z = 369 [M+H]+
161 315 mg, 55%; ηΗ NMR (400 MHz,
MeOD-d4): δ 1.54 (d, 5H), 1.60- 1.68 (m, 2H), 1.90 (td, 2H), 2.32 (s, 3H), 3.49 (d, 2H), 3.54-3.79
(2-{[1 -(5-methylpyridin-2- (m, 6H), 4.83 (s, 3H), 5.21 (q, yl)ethyl]amino}pyrimidin-5-yl)(8-oxa- 1H), 7.33 (d, 1H), 7.59 (d, 1H),
2-azaspiro[4.5]dec-2-yl)methanone
8.32 (s, 1H), 8.50 (s, 2H). LCMS m/z = 382 [M+H]+
162e 15 mg, 11%; 1H NMR (400 MHz,
MeOD-d4): δ 1.50-1.69 (m, 7H), 1.85-1.97 (m, 2H), 2.52 (s, 3H),
(2-{[1 -(6-methylpyridin-3- 3.50 (d, 2H), 3.55-3.80 (m, 6H), yl)ethyl]amino}pyrimidin-5-yl)(8-oxa- 5.17-5.27 (m, 1H), 7.28 (d, 1H),
2-azaspiro[4.5]dec-2-yl)methanone 7.80 (d, 1H), 8.46 (s, 1H), 8.52 (s,
2H). LCMS: m/z = 382 [M+H]+
163e 0 30 mg, 22%; 1H NMR (400 MHz,
MeOD-d4): δ 1.58 (d, 3H), 1.82 (dd, 4H), 2.52 (s, 3H), 3.55-3.70
(2-{[1 -(6-methylpyridin-3- (m, 4H), 3.89 (s, 2H), 4.18 (s, yl)ethyl]amino}pyrimidin-5-yl)(7-oxa- 2H), 5.19-5.28 (m, 1H), 7.29 (d,
2-azaspiro[3.5]non-2-yl)methanone 1H), 7.79 (dd, 1H), 8.45 (d, 1H),
8.59 (s, 2H). LCMS: m/z = 368 [M+H]+ 164 0 48 mg, 39%; ηΗ NMR (400 MHz,
DMSO-de): δ 1 .35-1 .58 (m, 4H),
Λ H 1 .72-1 .84 (m, 2H), 2.46 (s, 3H),
(2-{[(5-methylpyrazin-2- 3.40-3.65 (m, 8H), 4.58-4.67 (m, yl)methyl]amino}pyrimidin-5-yl)(8- 2H), 8.26-8.30 (m, 1 H), 8.40-8.60 oxa-2-azaspiro[4.5]dec-2- (m, 4H). LCMS: m/z = 369 yl)methanone [M+H]+
165 0 25 mg, 30%; ηΗ NMR (400 MHz,
DMSO-de): δ 1 .66-1 .70 (m, 4H), 2.46 (s, 3H), 3.40-3.55 (m, 4H),
(2-{[(5-methylpyrazin-2- 3.74 (s, 2H), 4.12 (s, 2H), 4.64 (d, yl)methyl]amino}pyrimidin-5-yl)(7- 2H), 8.34-8.40 (m, 1 H), 8.44 (d, oxa-2-azaspiro[3.5]non-2- 2H), 8.56 (br d, 2H). LCMS: m/z = yl)methanone 355 [M+H]+
166 33 mg, 33%; 1H NMR (400 MHz,
DMSO-de): δ 1 .40-1 .60 (m, 4H), 1 .72-1 .84 (m, 2H), 3.40-3.65 (m,
Figure imgf000163_0001
8H), 4.73-4.79 (m, 2H), 8.30 (br s,
8-oxa-2-azaspiro[4.5]dec-2-yl[2- 1 H), 8.56 (br s, 2H), 9.06 (d, 1 H), ({[4-(trifluoromethyl)pyrimidin-5- 9.34 (s, 1 H). LCMS: m/z = 423 yl]methyl}amino)pyrimidin-5- [M+H]+
l]methanone
167 25 mg, 24%; 1H NMR (400 MHz,
DMSO-de): δ 1 .65-1 .78 (m, 4H), 3.40-3.60 (m, 4H), 3.75 (s, 2H),
Figure imgf000163_0002
4.13 (s, 2H), 4.71 -4.82 (m, 2H),
7-oxa-2-azaspiro[3.5]non-2-yl[2- 8.36-8.46 (m, 1 H), 8.66 (br s, 2H), ({[4-(trifluoromethyl)pyrimidin-5- 9.05 (s, 1 H), 9.34 (s, 1 H). LCMS: yl]methyl}amino)pyrimidin-5- m/z = 409 [M+H]+
yl]methanone
Figure imgf000164_0001
yl)methanone
Figure imgf000165_0001
yl)methanone (s, 1H). LCMS: m/z = 394 [M+H]+
Figure imgf000166_0001
180 0 223 mg, 27%; ηΗ NMR (400 MHz,
DMSO-de): δ 1 .63-1 .70 (m, 10H), 2.43 (s, 3H), 3.40-3.57 (m, 4H),
(2-{[2-(5-methylpyrazin-2-yl)propan- 3.70 (s, 2H), 4.08 (s, 2H), 8.23 (s, 2-yl]am ino}pyrim idin-5-yl)(7-oxa-2- 1 H), 8.26-8.70 (m, 4H). LCMS: azaspiro[3.5]non-2-yl)methanone m/z = 383 [M+H]+
181 36 mg, 18%; 1 H NMR (400 MHz,
MeOD-d4): δ 1 .53-1 .68 (m, 4H),
H 1 .87-1 .97 (m, 2H), 2.98 (t, 2H),
8-oxa-2-azaspiro[4.5]dec-2-yl(2-{[2- 3.50 (d, 2H), 3.55-3.78 (m, 8H), (pyrimidin-5- 8.50 (s, 2H), 8.69 (s, 2H), 8.98 (s, yl)ethyl]amino}pyrimidin-5- 1 H). LCMS: m/z = 369 [M+H]+ yl)methanone
All amines, or their common salts are commercia ly available with the exception of:
a 2-(Pyrimidin-5-yl)propan-2 -amine hydrochloride (Preparation 24)
b (1 R)-1 -(Pyrazin-2-yl)ethanamine hydrochloride (Preparation 28)
c (1 S)-1 -(Pyrazin-2-yl)ethanamine hydrochloride (Preparation 27)
d 1 -(Pyrimidin-5-yl)ethanamine hydrochloride (Preparation 29)
e 1 -(6-Methylpyridin-3-yl)ethanamine hydrochloride (Preparation 30)
f 1 -(Pyrimidin-5-yl)cyclobutanamine hydrochloride (Preparation 25)
Example 182
8-oxa-2-azaspiro[4.51dec-2-yl(2-{[2-(pyrazin-2-yl)propan-2-yl1amino)pyrimidin-5- vDmethanone
Figure imgf000167_0001
To a mixture of 2-{[2-(pyrazin-2-yl)propan-2-yl]amino}pyrimidine-5-carboxylic acid, (Preparation 88, 571 mg, 2.20 mmol) and 8-oxa-2-azaspiro[4.5]decane (31 1 mg, 2.20 mmol) in THF (10 mL) was added DIPEA (1 .73 g, 13.2 mmol) and T3P (50%w in EtOAc, 2.8 g, 4.40 mmol) and the resulting mixture heated at 60 °C for 18 hrs. The mixture was cooled, partitioned between brine and EtOAc and the layers separated. The aqueous phase extracted with EtOAc and the combined extracts washed with brine, dried (Na2S04) and concentrated under reduced pressure. The residue was purified by column chromatography on silica gel eluting with DCM:MeOH (90:10) to afford a tan foam which was further purified by column chromatography eluting with (EtOAc: MeCN). The residue was treated with Et2O which was removed under reduced pressure to give the title compound as a white foam (319 mg, 38%). 1 .42-1 .62 (m, 4H), 1 .77 (s, 6H), 1 .78-1 .84 (m, 2H), 3.38 (s, 2H), 3.46-3.68 (m, 6H), 6.66 (br s, 1 H), 8.33 (br s, 2H), 8.38 (d, 1 H), 8.46 (dd, 1 H), 8.70 (s, 1 H). LCMS: m/z = 383 [M+H]+
Example 188
8-oxa-2-azaspiro[4.51dec-2-yl(2-{[1 -(pyrimidin-5-yl)cvclopropyl1amino) pyrimidin-5- vQmethanone
Figure imgf000168_0001
A suspension of 2-{[1 -(Pyrimidin-5-yl)cyclopropyl]amino}pyrimidine-5-carboxylic acid (Preparation 5, 79.6 mg, 0.31 mmol), 8-oxa-2-azaspiro[4.5]decane hydrochloride (50.0 mg, 0.28 mmol), 2-chloro-1 -methylpyridinium iodide (Mukaiyama reagent, CAS# 14338-32-0, 108.0 mg, 0.42 mmol) and DIPEA (0.15 ml_, 0.84 mmol) in THF (15 ml_) was stirred at 65 °C for 18 hours. The reaction mixture was diluted with EtOAc/water (5:3, v/v, 80 ml_), and the aqueous layer was extracted with EtOAc (50 ml_*2). Combined organic layers were washed with brine (30 ml_), dried over MgSO4, filtered and concentrated under reduced pressure to give a yellow oil. The crude product was purified by flash column (MeOH:CH2Cl2 from 0% to 10%, 12 g silica gel) to provide the title compound, 40 mg, 38%. 1H NMR (400 MHz, MeOD-d4): δ 1 .40-1 .70 (m, 8H), 1 .87- 1 .98 (m, 2H), 3.52 (d, 2H), 3.57-3.80 (m, 6H), 8.58 (br s, 2H), 8.71 (d, 2H), 8.97 (s, 1 H). LCMS m/z = 381 [M+H]+
The following Examples 183-187 and 189-202 (Table 5) were prepared according to the general procedure outlined above for Example 182, using the appropriate carboxylic acid (Preparations 2, 5 and 6) and the appropriate amine. One skilled-in-the-art could also use any known peptidic coupling reagents and conditions. Table 5: Data for Examples 183-187 and 189-202
Figure imgf000169_0001
Figure imgf000170_0001
Figure imgf000171_0001
o 81 mg, 50%; ηΗ NMR (400 MHz,
MeOD-d4): δ 1.40-1.53 (m, 4H),
W N i h 1.65-1.75 (m, 1H), 1.78-1.93 (m,
1 -[(2-{[1 -(pyrimidin-5- 1H), 1.96-2.11 (m, 2H), 3.02-3.10 yl)cyclopropyl]amino}pyrimidin-5- (m, 1H), 3.36-3.48 (m, 1H), 3.65- yl)carbonyl]piperidine-3- 3.99 (m, 3H), 8.45 (br s, 2H), 8.72 carbonitrile (s, 2H), 8.97 (s, 1H). LCMS: m/z =
350 [M+H]+
First eluting enantiomer: 15 mg,
20% (after SFC on Example 195, Method CP-E); RT = 3.053 mins
Figure imgf000172_0001
(Method CA-E); 1H NMR (400 MHz,
1-[(2-{[1-(Pyrimidin-5- MeOD-d4) : δ 1.42-1.52 (m, 4H), yl)cyclopropyl]amino}pyrimidin-5- 1.65-1.75 (m, 1H), 1.78-1.91 (m, yl)carbonyl]piperidine-3- 1H), 1.98-2.09 (m, 2H), 3.05 (m, carbonitrile
1H), 3.36-3.47 (m, 1H), 3.68-3.97 (m, 3H), 8.45 (br s, 2H), 8.71 (s, 2H), 8.97 (s, 1H). LCMS m/z = 372 [M+Na]+
Second eluting enantiomer: 13 mg, 17% (after SFC on Example 195, Method CP-E); RT = 3.262
Figure imgf000172_0002
mins (Method CA-E); 1H NMR (400
1-[(2-{[1-(Pyrimidin-5- MHz, MeOD-d4) : δ 1.41-1.53 (m, yl)cyclopropyl]amino}pyrimidin-5- 4H), 1.65-1.75 (m, 1H), 1.78-1.92 yl)carbonyl]piperidine-3- (m, 1H), 1.99-2.09 (m, 2H), 3.05 (m, carbonitrile
1H), 3.36-3.47 (m, 1H), 3.68-3.99 (m, 3H), 8.45 (br s, 2H), 8.70 (s, 2H), 8.97 (s, 1H). LCMS m/z = 350 [M+H]+
Figure imgf000173_0001
201 a O HO First eluting enantiomer: 21 mg,
7% (after SFC Method CP-G); RT = 4.818 mins (Method CA-F);1H NMR
(400 MHz, MeOH-d4): δ 0.85-0.98
[3-Ethyl-3-(hydroxymethyl)
(m, 3H), 1 .17 (d, 2H), 1 .35-1 .65 (m, pyrrolidin-1 -yl](2-{[1 -(pyrimidin-5- 5H), 1 .73-1 .99 (m, 2H), 3.41 -3.73 yl)cyclopropyl]amino}pyrimidin-5- (m, 5H), 8.56 (br s, 2H), 8.71 (s, yl)methanone
2H), 8.97 (s, 1 H). LCMS m/z = 369 [M+H]+
202a Second eluting enantiomer: 21 mg, 7% (after SFC Method CP-G); RT = 5.098 mins (Method CA-F); 1H
NMR (400 MHz, MeOH-d4): δ 0.86-
[3-Ethyl-3-(hydroxymethyl)
0.98 (m, 3H), 1 .17 (d, 2H), 1 .37-1 .66 pyrrolidin-1 -yl](2-{[1 -(pyrimidin-5- (m, 5H), 1 .72-1 .97 (m, 2H), 3.41 - yl)cyclopropyl]amino}pyrimidin-5- 3.74 (m, 5H), 8.56 (br s, 2H), 8.71 yl)methanone
(s, 2H), 8.97 (s, 1 H). LCMS m/z = 369 [M+H]+
All amines, or their common salts are commercially available with the exception of: a 3-ethylpyrrolidin-3-yl)methanol (Preparation 66)
Example 203
8-oxa-2-azaspiro[4.51dec-2-yl[2-(pyrimidin-5-ylmethoxy)pyrimidin-5-yl1methanone
Figure imgf000174_0001
To a solution of pyrimidin-5-ylmethanol (70.4 mg, 0.639 mmol) and LiHMDS (1 .33 mL, 1 .33 mmol) in DMF (5 mL) at 0°C was added (2-chloropyrimidin-5-yl)(8-oxa- 2-azaspiro[4.5]dec-2-yl)methanone (Preparation 22, 150 mg, 0.532 mmol) and the resulting mixture stirred at 0°C for 2 hr. The mixture was purified directly by HPLC using a DuraShell 150*25mm*5um column, eluting with 5-35% water (0.05% ammonium hydroxide):MeCN over 10 mins and a flow rate of 30 mL/min to afford the title compound as a white solid, 10 mg, 5%. 1H NMR (400 MHz, DMSO-d6): δ 1 .42-1 .48 (m, 2H), 1 .54-1.59 (m, 2H), 1 .78-1 .85 (m, 2H), 3.40-3.48 (m, 2H), 3.50-3.65 (m, 6H), 5.52 (s, 2H), 8.85 (d, 1 H), 8.97 (d, 1 H), 9.19 (s, 1 H). LCMS m/z = 356 [M+H]+
Example 204
N-[5-(8-oxa-2-azaspiro[4.51dec-2-ylcarbonyl)pyrimidin-2-yllpyrazine-2-carboxamide
Figure imgf000175_0001
To a -15 °C solution of pyrazine-2-carboxylic acid (CAS# 98-97-5, 47.3 mg, 0.381 mmol) and (2-aminopyrimidin-5-yl)(8-oxa-2-azaspiro[4.5]dec-2-yl)methanone (Preparation 89, 100 mg, 0.381 mmol) in pyridine (0.5 ml_), was added phosphoryl trichloride (70.1 mg, 0.457 mmol) dropwise. The cold bath was removed and the resulting yellow mixture was stirred at room temperature for 1 hour. The reaction was quenched with water (5 ml_) and then extracted with EtOAc (5 ml_) twice. The combined organic extarcts were washed with 0.5N HCI (5 ml_), sat.NaHC03 (5 ml_) and brine (5 ml_) in turns, dried over Na2S04 and concentrated to give crude product. Purification by preparative HPLC (Agela Durashell C18 150*25 5pm, using water(0.225%FA)-MeCN, from 10% to 30% over 1 1 min, at a flow rate of 35 mL/min) followed by lyophilisation gave the title compound, 48 mg, 34%. 1H NMR (400 MHz, DMSO-d6): δ 1 .41 -1 .64 (m, 4H), 1 .82 (dt, 2H), 3.41 (s, 1 H), 3.45-3.67 (m, 7H), 8.81 -8.85 (m, 1 H), 8.93 (s, 1 H), 8.95 (s, 1 H), 8.97 (t, 1 H), 9.31 (dd, 1 H), 10.83 (br s, 1 H). LCMS m/z = 369 [M+H]+ Example 205
N-[5-(8-oxa-2-azaspiro[4.51dec-2-ylcarbonyl)pyrimidin-2-yllpyrimidine-5-carboxamide
Figure imgf000175_0002
To a solution of pyrimidine-5-carboxylic acid (CAS# 4595-61 -3, 400 mg, 3.22 mmol) in dichloromethane (30 mL) was added dropwise a solution of Ghosez reagent (861 mg, 6.45 mmol) in dichloromethane (10 mL). The solution was stirred at 20 °C for
30 minutes, then cooled to -5 °C in an ice-salt bath. A solution of (2-aminopyrimidin-5- yl)(8-oxa-2-azaspiro[4.5]dec-2-yl)methanone (Preparation 89, 676 mg, 2.58 mmol) and
Et3N (978 mg, 9.67 mmol) in dichloromethane (10 mL) was added slowly to the reaction mixture. The reaction was stirred at 20 °C for 2.5 hrs then quenched by NaHC03 aqueous solution. The layers were separated and the organic layer was evaporated to dryness. Purification by preparative HPLC (Daiso 150*25 5pm, water(10mM NH4HCO3)-MeCN, from 0% to 30% over 10 min, at a flow rate of 30 mL/min) gave the title product 7 mg, 1 %. 1H NMR (400 MHz, CDCI3): δ 1 .22-1 .71 (m, 4H), 1 .84-2.00 (m, 2H), 3.39-3.82 (m, 8H), 5.25-5.37 (m, 1 H), 8.57 (s, 1 H), 8.89 (br. s., 2H), 9.27 (s, 2H), 9.43 (s, 1 H). LCMS m/z = 369 [M+H]+
SUMMARY OF BIOLOGICAL ASSAYS AND DATA
Human Vanin-1 Enzyme Assay 1 . The in vitro assay measures enzymatic cleavage of the fluorescently-labeled vanin substrate, pantetheine 7-amino-4- trifluoromethylcoumarin, by human vanin-1 .
Figure imgf000176_0001
The vanin-1 protein was prepared in-house from a construct expressing the extracellular domain of human vanin-1 (GenBank ID NM_004666) preceded N- terminally by the honey bee melittin signal peptide, a GSG linker sequence, a His6X tag and a FLAG tag. The secreted, soluble enzyme was purified from the conditioned medium from a CHO cell line stably expressing the resulting protein. Enzyme purification was performed through sequential Ni NTA and size-exclusion chromatography steps.
The test inhibitors were solubilized in DMSO to a stock concentration of 30 mM. On the day of the assay, dose response plates were prepared by diluting the inhibitors in DMSO at compound concentration 200-fold the final in-assay concentration. Intermediate concentrations were prepared by diluting in DMSO in a four-fold series for a total of 1 1 data points.
To prepare a working solution of human vanin-1 , the enzyme was diluted to 33.3 pM in the assay buffer consisting of 50 mM Tris-HCI pH=8.0, 50 mM KCI, 0.005% Brij-
35 and 1 .6 mM cysteamine. To begin the assay 100 nL was transferred from the compound plate to the assay plate. Next, 15 μί of the vanin-1 working solution were transferred to the assay plate. The inhibitor and enzyme were incubated at room temperature for 30 minutes. The enzyme reaction was then initiated by the addition of 5 μΙ_ of 200 μΜ pantetheine 7-amino-4-trifluoromethylcoumarin prepared in assay buffer. The final concentrations in the assay were 25 pM human vanin-1 and 50 uM substrate. The final concentration of DMSO was 0.5%. The assay plates were incubated for 60 minutes and before they were read on a Perkin Elmer EnVision Model 2103 using a 405 nm excitation wavelength and a 510 nm emission wavelength for detection.
Vanin-1 in Human Plasma Assay. The in vitro assay measures enzymatic cleavage of the fluorescently-labeled vanin substrate, pantetheine 7-amino-4- trifluoromethylcoumarin, by human vanin-1 present in human plasma.
Figure imgf000177_0001
Human plasma was prepared from whole blood drawn from healthy donors, collected in tubes containing sodium heparin. The plasma fractions were separated from the whole blood by centrifugation for 10 minutes at 2000xg and pooled. The concentration of vanin-1 in the pooled plasma was determined by measuring the rate of enzymatic hydrolysis of the fluorescently-labeled substrate by the vanin-1 in the plasma sample and comparing the observed hydrolysis rate to a recombinant human vanin-1 standard.
The test inhibitors were solubilized in DMSO to a stock concentration of 30 mM.
On the day of the assay, dose response plates were prepared by diluting the inhibitors in DMSO to a compound concentration 200-fold the final in-assay concentration.
Concentration series were prepared by serially diluting in 100% DMSO in a three-fold series. Intermediate compound plates containing compound in 2% DMSO were then created by diluting the compounds 50-fold in assay buffer consisting of 50 mM Tris-HCI pH=8.0, 50 mM KCI, 0.005% Brij-35. From this intermediate compound plate, 10 μΙ_ were transferred to the assay plate and mixed with 20 uL of the pooled human plasma. The inhibitor and enzyme were co-incubated at room temperature for 30 minutes. The enzyme reaction was initiated by the addition of 10 μΙ_ of 200 uM pantetheine 7-amino- 4-trifluoromethylcoumarin prepared in assay buffer. The final concentrations in the assay were 50% plasma, and 50 uM substrate. The final concentration of DMSO was 0.5%. Datapoints were measured over time using a Tecan Safire 2 platereader at 405 nm excitation wavelength and a 510 nm emission wavelength for detection.
The biological activity of certain compounds of the invention was tested in one or more of the assays described above. The results are shown in Table 6.
Vanin-1 Vanin-1
Human Human Human Human
Example Vanin-1 Plasma Example Vanin-1 Plasma Number ICso (nM) ICso (nM) Number ICso (nM) ICso (nM)
1 1 .312 27 1372.853
2 1 .332 28 1547.699
3 1 .694 29 1916.482
4 8.578 30 2170.505
5 16.049 31 3086.957
6 17.942 32 20000.000
7 17.946 33 108.335
8 37.101 34 47.165
9 48.089 35 5.424
10 75.872 36 1000.997
1 1 81 .672 37 0.504
12 94.108 38 62.610
13 101 .188 39 9.438
14 155.857 40 26.921
15 219.545 41 45.005
16 323.01 1 42 35.137
17 327.776 43 1 .353
18 384.808 44 2269.128
19 406.956 45 1848.644
20 452.621 46 6.285
21 528.418 47 29.564
22 559.349 48 20000.000
23 612.584 49 7.502
24 749.160 50 19.855
25 1039.731 51 97.512
26 1226.543 52 60.278 Example Human Vanin-1 Example Human Vanin-1
Number Vanin-1 Human Number Vanin-1 Human
ICso (nM) Plasma ICso (nM) Plasma
ICso (nM) ICso (nM)
53 12.158 79 21 .320
54 0.283 80 545.210
55 2519.219 81 101 .565
56 24.949 82 146.851
57 610.814 83 55.133
58 1 16.254 84 75.401
59 47.374 49.159 85 167.434
60 2846.082 86 24.159
61 2.507 87 79.792 68.047
62 3.921 88 28.653
63 420.940 89 34.957
64 100.438 90 347.876
65 1 .822 91 315.544
66 230.557 92 50.914
67 4242.905 93 40.492
68 8.135 94 20.448
69 1 .852 95 30.075
70 175.384 205.142 96 10.931 9.855
71 5.41 1 97 344.799
72 147.586 98 5.657
73 829.361 99 590.914
74 3.616 100 135.449
75 1075.367 101 1355.166
76 122.487 102 40.820
77 31 .293 103 590.277
78 1598.104 104 2.446 Example Human Vanin-1 Example Human Vanin-1
Number Vanin-1 Human Number Vanin-1 Human
ICso (nM) Plasma ICso (nM) Plasma
ICso (nM) ICso (nM)
105 35.950 131 <0.094 0.838
106 30.756 132 0.349
107 38.597 133 0.430 0.524
108 387.300 451.801 134 0.504
109 249.066 135 3.384 3.705
110 171.121 136 2330.344
111 3585.665 137 5.236 5.017
112 >18897.44 138 0.043
113 508.231 139 >17588.525
114 20.878 140 0.283 0.950
115 16011.757 141 87.341
116 112.894 142 7.656 10.147
117 5.109 143 0.082 1.219
118 4.642 144 0.743
119 1369.249 145 0.242 1.222
120 84.111 146 31.264 72.617
121 3.948 147 0.650 1.887
122 43.903 148 54.546
123 11387.672 149 0.677 1.297
124 4616.109 150 27.832 35.632
125 3.038 151 0.955
126 604.220 152 91.346
127 586.544 153 4.291
128 383.800 154 336.856
129 65.881 155 4.394 27.468
130 <0.041 0.743 156 838.451 Example Human Vanin-1 Example Human Vanin-1
Number Vanin-1 Human Number Vanin-1 Human
ICso (nM) Plasma ICso (nM) Plasma
ICso (nM) ICso (nM)
157 0.875 12.945 183 0.745
158 1 .1 12 2.031 184 1 .446 2.210
159 1 .291 185 7.707
160 2.837 186 5.482 4.627
161 8.057 187 83.71 1
162 0.629 2.967 188 0.277 1 .167
163 1 .770 189 0.867
164 27.300 190 12.516 12.418
165 26.305 191 1 .571
166 32.596 39.069 192 2.209
167 57.694 193 7.1 17 5.392
168 191 .839 194 20.155
169 157.567 195 10.650
170 6.330 16.459 196 1 176.654 >792.957
171 7.333 197 5.221
172 4.528 17.434 198 20.487
173 0.1 16 1.608 199 1 175.069
174 0.293 0.905 200 10.580 32.612
175 0.240 201 0.582
176 0.91 1 202 14.259
177 0.551 203 1087.464
178 2.184 204 5156.678
179 7.330 14.408 205 8801 .420
180 0.196
181 200.058
182 0.062 1.085 Variations, modifications, and other implementations of what is described herein will occur to those skilled in the art without departing from the spirit and the essential characteristics of the present teachings. Accordingly, the scope of the present teachings is to be defined not by the preceding illustrative description but instead by the following claims, and all changes that come within the meaning and range of equivalency of the claims are intended to be embraced therein.
Each of the printed publications, including but not limited to patents, patent applications, books, technical papers, trade publications and journal articles described or referenced in this specification are herein incorporated by reference in their entirety and for all purposes.

Claims

We claim:
1 . A compound of Formula I
Figure imgf000184_0001
wherein
G is a 6-membered heteroaryl, with one, two or three N, wherein the heteroaryl is optionally substituted with one, two or three substituents selected from Ci-C4alkyl, -NR8aC(0)R8b, -NR8aS02R8b -(CR6aR6b)tC(0)N(R8a)2, -C(0)OH, halogen, cyano, -N(R8a)2, -(CR6aR6b)tS02R8b, -(CR6aR6b)tS02N(R8a)2, Ci-C alkoxy, OH, -S(Ci-C3alkyl) or Cs-Cscycloalkyl, wherein the alkyl, cycloalkyl and alkoxy are optionally substituted with one, two or three halogen, OH, OCH3, or Cs-Cscycloalkyl;
L is NH or O;
Z is a bond; -(CR5aR5b)q-; -CH2(CR5aR5b)m-; or -(CR5aR5b)m-W-(CR5aR5b)n-, wherein W is S, O or NR7;
Ra, Rb, R1 a and R1 b are each independently hydrogen, Ci-C4alkyl, wherein the alkyl is optionally substituted with one, two or three halogen, OH, cyano, -S(Ci- Csalkyl) or Ci-C4alkoxy, optionally substituted with one, two or three fluoro;
or R1 a and R1 b, together with the carbon to which they are bonded, form an oxo, C3-C5cycloalkyl, -(4- to 5-membered heterocycloalkyi) wherein said cycloalkyl or heterocycloalkyi are optionally substituted with one, two, three or four halogen, OH, Ci-C4alkyl, -S(Ci-C3alkyl) Ci-C4alkoxy or cyano; and the heteroatom is selected from one or two N, S or O;
R2a and R2b are each independently hydrogen, -(CR6aR6b)tSO2R8b, OH, halogen, -(CR6aR6b)tC(O)N(R8a)2, -NR8aC(O)R8b, -NR8aC(O)N(R8a)2, -SO2N(R8a)2, Ci-C alkyl, Ci-C alkoxy, S(Ci-C3alkyl), -(CR6aR6b)t-(C3-C6cycloalkyl), cyano, -(CR6aR6b)n-(5- to 6- membered heterocycloalkyi) or -(CR6aR6b)n-(5- to 6- membered heteroaryl), wherein said heteroatoms of said heteroalkyl and heteroaryl are selected from one, two or three N, O or S; wherein said alkyl, cycloalkyl, heterocycloakyl and heteroaryl are optionally substituted with one, two, three or four R9; or
R2a and R2b together with the carbon to which they are bonded form a C3- Cgcycloalkyl or a -(4- to 1 1 -membered heterocycloalkyl), having one to three heteroatoms selected from N, 0 or S; wherein the cycloalkyl and heterocycloalkyl are optionally substituted with one, two or three Ci-C4alkyl, S(Ci-C3alkyl), OH, halogen, oxo, -C(O)NH2, -C(O)NHCH3, -C(O)N(CH3)2, C3-C5cycloalkyl or Ci- C4alkoxy; or if substitution is at a N atom, then such N atom is substituted with R7; or
R2a or R2b, and one of R5a or R5b, together with the respective carbons to which they are bonded, form a C3-Ci2cycloalkyl, C6-Cioaryl, -(5- to 6-membered heteroaryl) or a -(4- to 12-membered heterocycloalkyl), wherein said heteroaryl or heterocycloalkyl is optionally substituted with one, two, three or four R9; or if substitution is at a N atom, then such N atom is substituted with R7; or
R2a or R2b, and R7, together with the respective atoms to which they are bonded form a -(4- to 12- membered heterocycloalkyl) or a -(5- to 6-membered heteroaryl), wherein said heterocycloalkyl or heteroaryl have one, two to three heteroatoms selected from N, O or S, wherein said heterocycloalkyl and heteroaryl are optionally substituted with one, two, three or four R9; or if substitution is at a N atom, then such N atom is substituted with R7;
R3 is hydrogen, -(CR6aR6b)tC(O)NH2,or Ci-C alkyl, wherein said alkyl is optionally substituted with one, two, three or four R9; or
R3 and Rb, together with the carbon to which they are attached, form an oxo;
R4 is hydrogen, -(CR6aR6b)tC(O)NH2, or Ci-C alkyl, wherein said alkyl is optionally substituted with one, two, three or four R9; or
R3 and R4 taken together with the respective carbons to which they are bonded form a -(4- to 1 1 -membered heterocycloalkyl), having one to two heteroatoms selected from N, O or S, wherein the heterocycloalkyl are optionally substituted with one, two, three or four R9; or if substitution is at a N atom, then such N atom is substituted with R7; or
R4 and Ra, together with the carbon to which they are attached, form an oxo;
R5a and R5b are each independently hydrogen, halogen, OH, -(CR6aR6b)tC(O)R8b, -(CR6aR6b)tC(O)NH2, Ci-C alkyl, S(Ci-C3alkyl), Ci-C alkoxy, cyano, -(CR6aR6b)t-(C3-C6cycloalkyl) or -(CR6aR6b)t- (C3-C6heterocycloalkyl), wherein said alkyl, cycloalkyl and heterocycloalkyl are optionally substituted with one, two, three or four R9; and the heteroatom is selected from one or two N, 0, or S; or
R5a and R5b taken together with the carbon to which they are bonded form a Cs-Cgcycloalkyl or a 4- to 1 1 -membered heterocycloalkyi, wherein the heteroatom is selected from one or two N, S or 0, wherein said cycloalkyi or heterocycloalkyi is optionally substituted with one, two, three or four R9; or if substitution is at a N atom, then such N atom is substituted with R7; or
R3 and either R5a or R5b taken together with the respective carbons to which they are bonded form a C3-Ciocycloalkyl or -(4- to 12-membered heterocycloalkyi), wherein the heteroatom is selected from one or two N or 0, wherein said cycloalkyi and heterocycloalkyi are optionally substituted with one, two, three or four R9 or oxo; or if substitution is at a N atom, then such N atom is substituted with R7;
R6a and R6b are each independently hydrogen, Ci-C4alkyl, S(Ci-C3alkyl), OH, Ci-C4alkoxy, cyano or halogen;
R7 is hydrogen; -(4- to 6-membered heterocycloalkyi), having 1 to 2 heteroatoms wherein said heteroatom is selected from O, N and S; d-Csalkyl; S(Ci- C3alkyl); C(O)R8b; SO2R8b; SO2N(R8a)2; C(O)N(R8a)2 or -(C3-C7cycloalkyl), wherein said alkyl, heterocycloalkyi and cycloalkyi are optionally substituted with Ra;
R8a is hydrogen, Ci-C4alkyl or -(C3-C7cycloalkyl);
R8b is Ci-C alkyl, -(C3-C7cycloalkyl), (CR6aR6b)tSO2N(R8a)2, -(CR6aR6b)tSO2R8a or -(CR6aR6b)tNHC(O)N(R8a)2;
R9 is hydrogen, Ci-C alkyl, S(Ci-C3alkyl), OH, CH2OH, halogen, Ci-C alkoxy, cyano or -C(O)NH2, wherein said alkyl and alkoxy are optionally substituted with OH, halogen, -C(O)NH2, -C(O)NHCH3, -C(O)N(CH3)2, -S(Ci-C alkyl) or C3- Cscycloalkyl; or R9 is oxo, provided that it is attached to a non-aromatic group;
R10 is hydrogen or Ci-C3alkyl;
m, n and t are each independently 0, 1 or 2;
q is 1 , 2 or 3; and
x is 1 or 2;
or a pharmaceutically acceptable salt thereof.
2. The compound of formula la, lb, lc, Id, le, If or Ig,
Figure imgf000187_0001
Figure imgf000187_0002
Figure imgf000187_0003
wherein
G is a triazinyl, pyridazinyl, pyridonyl, pyridinyl, pyrazinyl or pyrimidinyl, optionally substituted with one, two or three substituents selected from halogen, OH, cyano, Ci-C4alkyl, -NR8aC(O)R8b, -NR8aSO2R8b -(CR6aR6b)tC(O)N(R8a)2, C(O)OH, -N(R8a)2, -(CR6aR6b)tSO2R8b, -(CR6aR6b)tSO2N(R8a)2, Ci-C4alkoxy, S(Ci-C3alkyl) or Cs-Cscycloalkyl, wherein the alkyl, cycloalkyl and alkoxy are optionally substituted with one, two or three halogen, OH, OCH3, or Cs-Cscycloalkyl;
L is NH or O;
Ra, Rb, R1 a and R1 b are each independently hydrogen, Ci-C4alkyl, wherein the alkyl is optionally substituted with one, two or three halogen, OH, cyano or Ci-C4alkoxy (optionally substituted with one, two or three fluoro);
or R1 a and R1 b, together with the carbon to which they are bonded, form an oxo, C3-C5cycloalkyl, -(4- to 5-membered heterocycloalkyl) wherein said cycloalkyl or heterocycloalkyl are optionally substituted with one, two, three or four halogen, OH, Ci-C4alkyl, Ci-C4alkoxy or cyano;
R2a and R2b are each independently hydrogen, halogen, Ci-C4alkyl, -(CR6aR6b)tSO2R8b -(CR6aR6b)tC(O)N(R8a)2, -NR8C(O)R8b, -SO2N(R8a)2, -NR8C(O)N(R8a)2, S(Ci-C3alkyl), cyano, -(CR6aR6b)t(C3-C6cycloalkyl), OH, Ci-C4alkoxy, -(CR6aR6b)n(5- to 6- membered heterocycloalkyl), having one to three heteroatoms selected from N, O or S, -(CR6aR6b)n-(5- to 6-membered heteroaryl), having one to three heteroatoms selected from N, O or S, wherein said alkyl, cycloalkyl, heterocycloakyl and heteroaryl are optionally substituted with one, two, three or four R9; or
R2a and R2b together with the carbon to which they are bonded form a C3-C7cycloalkyl or a -(4- to 12-membered heterocycloalkyl), having one to three heteroatoms selected from N, O or S; wherein the cycloalkyl and heterocycloalkyl are optionally substituted with one, two or three Ci-C4alkyl, S(Ci-C3alkyl), OH, halogen, oxo, C(O)NH2, C(O)NHCH3, C(O)N(CH3)2, C3-C5cycloalkyl or Ci-C alkoxy; or if substitution is at a N atom, then such N atom is substituted with R7; or
R2a or R2b, and one of R5a or R5b, together with the respective carbons to which they are bonded, form a C3-Ci2cycloalkyl, C6-Cioaryl, -(5- to 6-membered heteroaryl) or a -(4- to 12-membered heterocycloalkyl), wherein said heteroaryl or heterocycloalkyl is optionally substituted with one, two, three or four R9; or if substitution is at a N atom, then such N atom is substituted with R7; or
R2a or R2b, and R7, together with the respective atoms to which they are bonded form a -(4- to 12- membered heterocycloalkyl) or a -(5- to 6-membered heteroaryl), wherein said heterocycloalkyl or heteroaryl have one, two to three heteroatoms selected from N, O or S, wherein said heterocycloalkyl and heteroaryl are optionally substituted with one, two, three or four R9;
R3 is hydrogen, -(CR6aR6b)tC(O)NH2 or Ci-C alkyl, wherein said alkyl is optionally substituted with one, two, three or four R9; or
R3 and Rb, together with the carbon to which they are attached, form an oxo; R4 is hydrogen, -(CR6aR6b)tC(O)NH2 or Ci-C alkyl, wherein said alkyl is optionally substituted with one, two, three or four R9; or
R4 and Ra, together with the carbon to which they are attached, form an oxo; or
R3 and R4 taken together with the respective carbons to which they are bonded form a -(4- to 12-membered heterocycloalkyl), having one to two heteroatoms selected from N, 0 or S, wherein the heterocycloalkyi are optionally substituted with one, two, three or four R9;
R5a and R5b are each independently hydrogen, halogen, OH, - (CR6aR6b)tC(O)R8b, -(CR6aR6b)tC(O)NH2, d-C4alkyl, S(Ci-C3alkyl), Ci-C alkoxy, cyano, -(CR6aR6b)t-(C3-C6cycloalkyl) or -(CR6aR6b)t-(C3-C6heterocycloalkyl) (having one, to two heteroatoms selected from N, O or S), wherein said alkyl, cycloalkyi and heterocycloalkyi are optionally substituted with one, two, three or four R9; or
R5a and R5b taken together with the carbon to which they are bonded form a C3-C6cycloalkyl or a 4- to 7-membered heterocycloalkyi, wherein the heteroatom is selected from one or two N, S or O, wherein said cycloalkyi or heterocycloalkyi is optionally substituted with one, two, three or four R9; or
R3 and either R5a or R5b taken together with the respective carbons to which they are bonded form a C3-C7cycloalkyl or -(4- to 12-membered heterocycloalkyi), wherein the heteroatom is selected from one or two N or O, wherein said cycloalkyi and heterocycloalkyi are optionally substituted with one, two, three or four R9 or oxo;
R6a and R6b are each independently hydrogen, Ci-C alkyl, S(Ci-C3alkyl), OH, Ci-C4alkoxy, — S(Ci-C alkyl), cyano or halogen;
R7 is hydrogen; -(4- to 6-membered heterocycloalkyi), having 1 to 2 heteroatoms wherein said heteroatom is selected from O, N and S; d-Csalkyl; S(Ci- C3alkyl); C(O)R8b; SO2R8b; SO2N(R8a)2; C(O)N(R8a)2 or -(C3-C7cycloalkyl), wherein said alkyl, heterocycloalkyi and cycloalkyi are optionally substituted with Ra;
R8a is hydrogen, Ci-C4alkyl or -(C3-C7cycloalkyl);
R8b is Ci-C alkyl, -(C3-C7cycloalkyl), -(CR6aR6b)tSO2N(R8a)2, - (CR6aR6b)tSO2R8a or -(CR6aR6b)tNHC(O)N(R8a)2;
R9 is hydrogen, Ci-C alkyl, S(Ci-C3alkyl), oxo, OH, CH2OH, halogen,
Ci-C4alkoxy, cyano or C(O)NH2, wherein said alkyl and alkoxy are optionally substituted with OH, halogen, C(O)NH2, C(O)NHCH3, C(O)N(CH3)2, -S(Ci-C alkyl) or C3-C5cycloalkyl;
R10 is hydrogen or Ci-C3alkyl;
m, n and t are each independently 0, 1 or 2;
q is 1 , 2 or 3; and
x is 1 or 2;
or a pharmaceutically acceptable salt thereof.
3. The compound of Claims 1 or 2 wherein R1 a or R1 b are each independently hydrogen, or Ci-C3alkyl, wherein the alkyl is optionally substituted with one, two or three fluoro, OH, cyano or Ci-C4alkoxy, optionally substituted with one, two or three fluoro; or R1 a and R1 b, together with the carbon to which they are bonded, form a C3-C4cycloalkyl or a 4-membered heterocycloalkyl, wherein said cycloalkyi or heterocycloalkyl are optionally substituted with one, two, three or four halogen, OH, Ci-C4alkyl, S(Ci-C3alkyl), Ci-C4alkoxy or cyano;
or a pharmaceutically acceptable salt thereof. 4. The compound of Claim 3 wherein R1 a and R1 b are each independently hydrogen or methyl; or R1 a and R1 b, together with the carbon to which they are bonded, form a cyclopropyl, cyclobutyl or an oxetane; or a pharmaceutically acceptable salt thereof. 5. The compound of Claim 4 wherein L is NH; Ra and Rb are H; and x is
1 ; or a pharmaceutically acceptable salt thereof.
6. The compound of Claim 5 wherein R3 and R4 are each independently hydrogen or Ci-C4alkyl, wherein said alkyl is optionally substituted with one, two, three or four R9; or
R3 and R4 taken together with the respective carbons to which they are bonded form a -(4- to 12-membered heterocycloalkyl), having one to two heteroatoms selected from N, O or S, wherein the heterocycloalkyl is optionally substituted with one, two, three or four R9; R9 is OH, CH2OH, halogen, Ci-C4alkyl, Ci-C4alkoxy or cyano; or a pharmaceutically acceptable salt thereof.
7. The compound of Claim 6 wherein G is selected from pyrazinyl, pyrimidinyl, pyridinyl or pyridazinyl, optionally substituted with methyl, CH2F, CHF2 or CF3; or a pharmaceutically acceptable salt thereof.
8. The compound of Claim 7 wherein R2a and R2b together with the carbon to which they are bonded form a oxetane, tetrahydrofuran, tetrahydropyran, cyclobutyl, cyclopentyl, or cyclohexyl, each of which is optionally substituted with one or two Ci-C4alkyl or OH; or a pharmaceutically acceptable salt thereof.
9. The compound of Claim 7 wherein R and R are each independently hydrogen; fluoro; OH; Ci-C4alkyl; Ci-C4alkoxy; C3-C6cycloalkyl; 5-membered heteroaryl, having one or two N; cyano; -SO2CH3; -C(O)NHR8a; -NHC(O)NHR8a; wherein said alkyl, alkoxy, cycloalkyi and heteroaryl are optionally substituted by one, two, three or four R9; wherein R9 is OH, fluoro, methyl, ethyl, methoxy or ethoxy; or a pharmaceutically acceptable salt thereof.
10. The compound of Claim 7 wherein one of R2a or R2b, taken together with the carbon to which they are bonded, and one of R5a or R5b, form a cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, tetrahydrofuran or phenyl, wherein each is optionally substituted with one, two, three or four R9, wherein R9 is OH, CH2F, CHF2, CF3, CH2OH; or a pharmaceutically acceptable salt thereof.
1 1 . The compound of Formula la of Claims 8, 9 or 10
Figure imgf000191_0001
la
wherein Z is -(CR5aR5b)q-; and q is 1 ; or a pharmaceutically acceptable salt thereof.
12. The compound of Claim 1 1 wherein R2a and R2b are each independently hydrogen, methyl, ethyl, propyl, isopropyl, methoxy or ethoxy, optionally substituted with R9 wherein R9 is OH; or R2a and R2b, together with the carbon to which they are bonded, form a tetrahydrofuran, cyclobutane, cyclopentane, cyclohexane, oxetane, tetrahydropyran, pyrrolidine, azetidine, each of which is optionally substituted with one, two, three or four R9; or R2a or R2b, and one of R5a or R5b, together with the respective carbons to which they are bonded, form a cyclopentane or cyclohexane, optionally substituted with one, two or three R9; or a pharmaceutically acceptable salt of said compound or a tautomer of said compound or said salt. The compound of Claim 12, having the formula Ila,
Figure imgf000192_0001
Ila
wherein R10 is hydrogen; and L is NH; G is pyrimidinyl or pyrazinyl; or a pharmaceutically acceptable salt thereof.
14. The compound of Claim 13, having the following absolute stereochemistry,
Figure imgf000192_0002
Ila
wherein R1 a is hydrogen; and R1 b is methyl, ethyl, propyl, wherein each is optionally substituted with one, two or three fluoro; or a pharmaceutically acceptable salt thereof.
15. The compound of Claim 14 wherein Ra, Rb, R3, R4, R5a and R5b are hydrogen; and x is 1 ; R1 b is methyl or ethyl, optionally substituted with one, two or three fluoro, or a pharmaceutically acceptable salt thereof.
The compound of Formula lb of Claims 8 9 or 10
Figure imgf000192_0003
lb
wherein Z is a bond; or a pharmaceutically acceptable salt thereof.
17. The compound of Claim 16 wherein R and R are each independently hydrogen, methyl, ethyl, propyl, isopropyl, methoxy or ethoxy, optionally substituted with R9 wherein R9 is OH; or R2a and R2b together with the carbon to which they are bonded form a tetrahydrofuran, cyclobutane, cyclopentane, cyclohexane, oxetane, tetrahydropyran, pyrrolidine, azetidine, each of which is optionally substituted with one, two, three or four R9; or a pharmaceutically acceptable salt thereof.
The compound of Claim 17, having the formula Mb,
Figure imgf000193_0001
lib
wherein R10 is hydrogen; and L is NH; or a pharmaceutically acceptable salt thereof. 19. The compound of claim 18 having the absolute stereochemistry of
Figure imgf000193_0002
lib
wherein R1 a is hydrogen; and R1 b is methyl, ethyl or propyl, each of which is optionally substituted by one, two or three fluoro; or a pharmaceutically acceptable salt thereof.
20. The Compound of Claim 19 wherein Ra, Rb, R3 and R4 are hydrogen; x is 1 ; R1 b is methyl or ethyl, optionally substituted with one, two or three fluoro; and G is pyrimidinyl or pyrazinyl; or a pharmaceutically acceptable salt thereof.
21 The compound of Formula le of Claims 8, 9 or 10,
Figure imgf000194_0001
le
wherein Z is-(CR5aR5b)m-W-(CR5aR5b)n-, W is NR7, m is 1 , and n is 0; or a
pharmaceutically acceptable salt thereof.
22. The compound of Claim 21 wherein L is NH; and R2a or R2b and R7, together with the respective atoms to which they are bonded form a -(4- to 12-membered heterocycloalkyl), having one or two heteroatoms selected from N or 0 , wherein said heterocycloalkyl is optionally substituted with one, two, three or four R9; or a pharmaceutically acceptable salt thereof.
23. The compound of Claim 22 wherein Ra, Rb, R3, R4, R5a and R5b are hydrogen and the heterocycloalkyl formed is a pyrrole; or a pharmaceutically acceptable salt thereof.
24. The compound of Claim 23 of the formula lie,
Figure imgf000194_0002
lie
or a pharmaceutically acceptable salt thereof.
25. The compound of Claim 24, having the absolute stereochemistry of
Figure imgf000194_0003
lie wherein R1 a is hydrogen; and R1 b is methyl, ethyl, propyl, each of which is optionally substituted by one, two or three fluoro; or a pharmaceutically acceptable salt thereof.
26. The Compound of Claim 25 wherein G is pyrimidinyl or pyrazinyl, or a pharmaceutically acceptable salt thereof.
27. The compound of Formula Ic of Claims 8 9 or 10,
Figure imgf000195_0001
Ic
wherein Z is-(CRsaFrD)q-; and q is 2;
R5a and R5b are each independently hydrogen, OH, fluoro, cyano, Ci-C4alkyl, Ci-C alkoxy, cyclopropyl, cyclobutyl, cyclopentyl, -(CR6aR6b)tC(O)NH2, or oxazolidinone, optionally substituted with one, two, three or four R9; or
R5a and R5b taken together with the carbon to which they are bonded form a oxetane, tetrahydrofuran, tetrahydropyran, oxazolidinone, cyclopentane, cyclohexane, cyclobutane, cyclopropane, wherein said cycloalkyi or heterocycloalkyi are optionally substituted with one, two, three or four R9;
R9 is fluoro, OH or Ci-C4alkoxy, and
t is 0 or 1 ;
or a pharmaceutically acceptable salt thereof.
28. The compound of Claim 26 wherein Ra, Rb, R3, R4, R5a and R5b are hydrogen and the heterocycloalkyi formed is a pyrrole; and L is NH; or a pharmaceutically acceptable salt thereof.
29. The compound of Claim 27 having the formula lie,
Figure imgf000196_0001
lie
wherein G is pyrimidinyl or pyrazinyl; or a pharmaceutically acceptable salt thereof. 30. The compound of Claim 28, having the absolute stereochemistry of,
Figure imgf000196_0002
lie
wherein R1 a is hydrogen; and R1 b is methyl, ethyl, propyl, each of which is optionally substituted with one, two or three fluoro; or a pharmaceutically acceptable salt thereof.
31 . The compound of Claim 1 selected from
7- oxa-2-azaspiro[3.5]non-2-yl(2-{[1 -(pyrazin-2-yl)cyclobutyl]amino}pyrimidin- 5-yl)methanone;
8- oxa-2-azaspiro[4.5]dec-2-yl(2-{[1 -(pyrazin-2-yl)cyclobutyl]amino}pyrimidin- 5-yl)methanone;
8-oxa-2-azaspiro[4.5]dec-2-yl(2-{[2-(pyrazin-2-yl)propan-2-yl]amino}pyrimidin- 5-yl)methanone;
8-oxa-2-azaspiro[4.5]dec-2-yl(2-{[2-(pyrimidin-5-yl)propan-2- yl]amino}pyrimidin-5-yl)methanone;
7- oxa-2-azaspiro[3.5]non-2-yl(2-{[2-(pyrazin-2-yl)propan-2-yl]amino}pyrimidin- 5-yl)methanone;
8- oxa-2-azaspiro[4.5]dec-2-yl(2-{[1 -(pyrimidin-5- yl)cyclobutyl]amino}pyrimidin-5-yl)methanone;
(2-{[2-(5-methylpyrazin-2-yl)propan-2-yl]amino}pyrimidin-5-yl)(7-oxa-2- azaspiro[3.5]non-2-yl)methanone; 8-oxa-2-azaspiro[4.5]dec-2-yl(2-{[(1 S)-1 -(pyrazin-2-yl)ethyl]amino}pyrimidin- 5-yl)methanone;
(2-{[1 -(6-methylpyndin-3-yl)cyclopropyl]amino}pyrimidin-5-yl)(8-oxa-2- azaspiro[4.5]dec-2-yl)methanone;
8-oxa-2-azaspiro[4.5]dec-2-yl(2-{[1 -(pyrimidin-5- yl)cyclopropyl]amino}pyrimidin-5-yl)methanone;
(2-{[(1 S)-1 -(6-methylpyndin-3-yl)ethyl]amino}pyrimidin-5-yl)(8-oxa-2- azaspiro[4.5]dec-2-yl)methanone;
7- oxa-2-azaspiro[3.5]non-2-yl(2-{[1 -(pyrimidin-5- yl)cyclobutyl]amino}pyrimidin-5-yl)methanone;
8- oxa-2-azaspiro[4.5]dec-2-yl(2-{[3-(pyrazin-2-yl)oxetan-3-yl]amino}pyrimidin- 5-yl)methanone;
8-oxa-2-azaspiro[4.5]dec-2-yl(2-{[3-(pyrimidin-5-yl)oxetan-3- yl]amino}pyrimidin-5-yl)methanone;
7-oxa-2-azaspiro[3.5]non-2-yl(2-{[3-(pyrimidin-5-yl)oxetan-3- yl]amino}pyrimidin-5-yl)methanone;
(2-{[1 -(5-methylpyndin-2-yl)ethyl]amino}pyrimidin-5-yl)(8-oxa-2- azaspiro[4.5]dec-2-yl)methanone;
8-oxa-2-azaspiro[4.5]dec-2-yl(2-{[1 -(pyndin-2-yl)cyclopropyl]amino}pyrimidin- 5-yl)methanone;
7-oxa-2-azaspiro[3.5]non-2-yl(2-{[2-(pyrimidin-5-yl)propan-2- yl]amino}pyrimidin-5-yl)methanone;
7- oxa-2-azaspiro[3.5]non-2-yl(2-{[(1 S)-1 -(pyrazin-2-yl)ethyl]amino}pyrimidin- 5-yl)methanone;
8-oxa-2-azaspiro[4.5]dec-2-yl(2-{[(1 S)-1 -(pynmidin-5-yl)ethyl]amino}pyrimidin- 5-yl)methanone;
(2-{[1 -(6-methylpyndin-3-yl)ethyl]amino}pyrimidin-5-yl)(8-oxa-2- azaspiro[4.5]dec-2-yl)methanone;
8- oxa-2-azaspiro[4.5]dec-2-yl(2-{[1 -(pynmidin-5-yl)ethyl]amino}pyrimidin-5- yl)methanone;
(2-{[1 -(6-methylpyndin-3-yl)cyclopropyl]amino}pyrimidin-5-yl)(7-oxa-2- azaspiro[3.5]non-2-yl)methanone;
7-oxa-2-azaspiro[3.5]non-2-yl(2-{[1 -(pyrimidin-5- yl)cyclopropyl]amino}pyrimidin-5-yl)methanone; 8-oxa-2-azaspiro[4.5]dec-2-yl(2-{[1 -(pyrazin-2-yl)cyclopropyl]amino}pyrimidin- 5-yl)methanone;
7- oxa-2-azaspiro[3.5]non-2-yl(2-{[(1 S)-1 -(pyrimidin-5- yl)ethyl]amino}pyrimidin-5-yl)methanone;
7-oxa-2-azaspiro[3.5]non-2-yl(2-{[1 -(pyrazin-2-yl)cyclopropyl]amino}pyrimidin- 5-yl)methanone;
(2-{[1 -(2-methylpynmidin-5-yl)ethyl]amino}pyrimidin-5-yl)(8-oxa-2- azaspiro[4.5]dec-2-yl)methanone;
8- oxa-2-azaspiro[4.5]dec-2-yl{2-[(pyndin-3-ylmethyl)amino]pyrimidin-5- yl}methanone;
7-oxa-2-azaspiro[3.5]non-2-yl{2-[(pyndin-3-ylmethyl)amino]pyrimidin-5- yl}methanone;
7- oxa-2-azaspiro[3.5]non-2-yl(2-{[1 -(pynmidin-5-yl)ethyl]amino}pyrimidin-5- yl)methanone;
(2-{[1 -(6-methylpyridin-3-yl)ethyl]amino}pyrimidin-5-yl)(7-oxa-2- azaspiro[3.5]non-2-yl)methanone;
8- oxa-2-azaspiro[4.5]dec-2-yl(2-{[1 -(pyrimidin-2- yl)cyclopropyl]amino}pyrimidin-5-yl)methanone;
(2-{[1 -(2-methylpynmidin-5-yl)ethyl]amino}pyrimidin-5-yl)(7-oxa-2- azaspiro[3.5]non-2-yl)methanone;
7- oxa-2-azaspiro[3.5]non-2-yl{2-[(pyrazin-2-ylmethyl)amino]pyrimidin-5- yl}methanone;
8- oxa-2-azaspiro[4.5]dec-2-yl(2-{[(1 S)-1 -(pyndazin-4-yl)ethyl]amino}pyrimidin- 5-yl)methanone;
8-oxa-2-azaspiro[4.5]dec-2-yl(2-{[(1 S)-1 -(pynmidin-4-yl)ethyl]amino}pyrimidin- 5-yl)methanone;
(2-{[(6-methylpyridin-3-yl)methyl]amino}pyrimidin-5-yl)(7-oxa-2- azaspiro[3.5]non-2-yl)methanone;
(2-{[(6-methylpyridin-3-yl)methyl]amino}pyrimidin-5-yl)(8-oxa-2- azaspiro[4.5]dec-2-yl)methanone;
7-oxa-2-azaspiro[3.5]non-2-yl{2-[(pynmidin-5-ylmethyl)amino]pyrimidin-5- yl}methanone;
(2-{[(2-methylpynmidin-5-yl)methyl]amino}pyrimidin-5-yl)(8-oxa-2- azaspiro[4.5]dec-2-yl)methanone; 8-oxa-2-azaspiro[4.5]dec-2-yl{2-[(pynmidin-5-ylmethyl)amino]pyrimidin-5- yl}methanone;
8-oxa-2-azaspiro[4.5]dec-2-yl{2-[(pyrazin-2-ylmethyl)amino]pyrimidin-5- yl}methanone;
(7,9-dimethyl-8-oxa-2-azaspiro[4.5]dec-2-yl)(2-{[1 -(pyrimidin-5- yl)cyclopropyl]amino}pyrimidin-5-yl)methanone;
(3,3-diethylpyrrolidin-1 -yl)(2-{[1 -(pyrimidin-5-yl)cyclopropyl]amino}pynmidin yl)methanone;
[(3S)-3-ethyl-3-(hydroxymethyl)pyrrolidin-1 -yl](2-{[1 -(pyrimidin-5- yl)cyclopropyl]amino}pyrimidin-5-yl)methanone;
[3-(1 H-pyrazol-3-yl)pyrrolidin-1 -yl](2-{[1 -(pyrimidin-5- yl)cyclopropyl]amino}pyrimidin-5-yl)methanone;
(3,3-dimethylpyrrolidin-1 -yl)(2-{[1 -(pyrimidin-5-yl)cyclopropyl]amino}pynmi 5-yl)methanone;
[(3aR,4R,7aS)-4-hydroxyoctahydro-2H-isoindol-2-yl]{2-[(pyridin-3- ylmethyl)amino]pyrimidin-5-yl}methanone;
[3-(3-methyl-1 H-pyrazol-5-yl)pyrrolidin-1 -yl](2-{[1 -(pyrimidin-5- yl)cyclopropyl]amino}pyrimidin-5-yl)methanone;
hexahydrocyclopenta[c]pyrrol-2(1 H)-yl(2-{[1 -(pyrimidin-5- yl)cyclopropyl]amino}pyrimidin-5-yl)methanone;
(3-ethyl-3-methoxypyrrolidin-1 -yl)(2-{[1 -(pyrimidin-5- yl)cyclopropyl]amino}pyrimidin-5-yl)methanone;
(3-ethoxypiperidin-1 -yl)(2-{[1 -(pynmidin-5-yl)cyclopropyl]amino}pyrimidin-5- yl)methanone;
[(3aR7aR)-3a-(hydroxymethyl)octahydro-2H-isoindol-2-yl](2-{[1 -(pyrimidin-5- yl)cyclopropyl]amino}pyrimidin-5-yl)methanone;
(2-{[1 -(pynmidin-5-yl)cyclopropyl]amino}pyrimidin-5-yl)[3- (trifluoromethyl)pipendin-1 -yl]methanone;
[(3R,4R)-3-(hydroxymethyl)-3,4-dimethylpyrrolidin-1 -yl](2-{[1 -(pynmidin-5- yl)cyclopropyl]amino}pyrimidin-5-yl)methanone;
(3R)-1 -[(2-{[1 -(pynmidin-5-yl)cyclopropyl]amino}pyrimidin-5- yl)carbonyl]piperidine-3-carbonitrile;
[3-(methoxymethyl)piperidin-1 -yl](2-{[1 -(pyrimidin-5- yl)cyclopropyl]amino}pyrimidin-5-yl)methanone; [(7S,8aS)-7-fluorohexahydropyrrolo[1 ,2-a]pyrazin-2(1 H)-yl](2-{[1 -(pynmidin-5- yl)cyclopropyl]amino}pyrimidin-5-yl)methanone;
(4,4-difluoropiperidin-1 -yl)(2-{[1 -(pyrim^
yl)methanone;
(4-ethyl-4-methylpiperidin-1 -yl)(2-{[1 -(pyrim idin-5- yl)cyclopropyl]amino}pyrimidin-5-yl)methanone;
(2-{[1 -(pyrim idin-5-yl)cyclopropyl]amino}pyrimidin-5-yl)[(3R)-3- (trifluoromethyl)pyrrolidin-1 -yl]methanone;
8-azaspiro[4.5]dec-8-yl(2-{[1 -(pynmidin-5-yl)cyclopropyl]amino}pyrimidin-5- yl)methanone;
(4-fluoropiperidin-1 -yl)(2-{[1 -(pynmidin-5-yl)cyclopropyl]amino}pyrimidi yl)methanone;
[3-ethyl-3-(hydroxymethyl)pyrrolidin-1 -yl]{2-[(pyridin-3- ylmethyl)amino]pynmidin-5-yl}methanone;
(3-methylpiperidin-1 -yl)(2-{[1 -(pynmidin-5-yl)cyclopropyl]amino}pynmi yl)methanone;
[(3R)-3-(methylsulfonyl)piperidin-1 -yl](2-{[1 -(pyrimidin-5- yl)cyclopropyl]amino}pynmidin-5-yl)methanone;
1 -[(2-{[1 -(pyrimidin-5-yl)cyclopropyl]amino}pyrimidin-5-yl)carbonyl]pipend 3-carbonitrile;
(3-methoxypiperidin-1 -yl)(2-{[1 -(pynmidin-5-yl)cyclopropyl]amino}pynmi yl)methanone;
[(3S,4S)-3-(hydroxymethyl)-4-(tnfluoromethyl)pyrrolidin-1 -yl](2-{[1 -(pyrimidin- 5-yl)cyclopropyl]amino}pynmidin-5-yl)methanone;
pipendin-1 -yl(2-{[1 -(pynmidin-5-yl)cyclopropyl]amino}pyrimidin-5- yl)methanone;
[(3R)-3-ethyl-3-(hydroxymethyl)pyrrolidin-1 -yl](2-{[1 -(pyrimidin-5- yl)cyclopropyl]amino}pynmidin-5-yl)methanone;
(3-cyclopropyl-3-hydroxyazetidin-1 -yl){2-[(pyridin-3-ylmethyl)amino]pynmidin- 5-yl}methanone;
[(3R,4R)-3-(hydroxymethyl)-4-methylpyrrolidin-1 -yl]{2-[(pyridin-3- ylmethyl)amino]pynmidin-5-yl}methanone;
{2-[(pyridin-3-ylmethyl)amino]pynmidin-5-yl}[3-(trifluorometh^
yl]methanone; 1 -oxa-9-azaspiro[5.5]undec-9-yl(2-{[1 -(pyrimidin-5- yl)cyclopropyl]amino}pyrimidin-5-yl)methanone;
(2-{[1 -(pynmidin-5-yl)cyclopropyl]amino}pyrimidin-5-yl)[(3S)-3- (trifluoromethyl)pyrrolidin-1 -yl]methanone;
[4-hydroxy-3-(trifluoromethyl)piperidin-1 -yl](2-{[1 -(pyrimidin-5- yl)cyclopropyl]amino}pyrimidin-5-yl)methanone;
[3-(methylsulfonyl)piperidin-1 -yl](2-{[1 -(pyrimidin-5- yl)cyclopropyl]amino}pyrimidin-5-yl)methanone;
[(3R)-3-methoxypyrrolidin-1 -yl](2-{[1 -(pyrimidin-5- yl)cyclopropyl]amino}pyrimidin-5-yl)methanone;
[(3R,5S)-3-hydroxy-5-methylpiperidin-1 -yl](2-{[1 -(pyrimidin-5- yl)cyclopropyl]amino}pyrimidin-5-yl)methanone;
1 -oxa-8-azaspiro[4.5]dec-8-yl(2-{[1 -(pyrimidin-5- yl)cyclopropyl]amino}pyrimidin-5-yl)methanone;
(2-{[(5-methylpyrazin-2-yl)methyl]amino}pyrimidin-5-yl)(7-oxa-2- azaspiro[3.5]non-2-yl)methanone;
[(3R,5R)-3-hydroxy-5-(propan-2-yl)pipendin-1 -yl](2-{[1 -(pyrimidin-5- yl)cyclopropyl]amino}pyrimidin-5-yl)methanone;
(2-{[(5-methylpyrazin-2-yl)methyl]amino}pyrimidin-5-yl)(8-oxa-2- azaspiro[4.5]dec-2-yl)methanone;
8-oxa-2-azaspiro[4.5]dec-2-yl(2-{[(1 R)-1 -(pyrimidin-5- yl)ethyl]amino}pyrimidin-5-yl)methanone;
[(3R,5R)-3-cyclopropyl-5-hydroxypiperidin-1 -yl](2-{[1 -(pynmidin-5- yl)cyclopropyl]amino}pyrimidin-5-yl)methanone;
[(2S)-2-methylpyrrolidin-1 -yl](2-{[1 -(pyrimidin-5- yl)cyclopropyl]amino}pyrimidin-5-yl)methanone;
[4-ethyl-4-(hydroxymethyl)piperidin-1 -yl](2-{[1 -(pyrimidin-5- yl)cyclopropyl]amino}pyrimidin-5-yl)methanone;
[3-(hydroxymethyl)piperidin-1 -yl](2-{[1 -(pyrimidin-5- yl)cyclopropyl]amino}pyrimidin-5-yl)methanone;
8-oxa-2-azaspiro[4.5]dec-2-yl(2-{[(1 R)-1 -(pyrazin-2-yl)ethyl]amino}pyrimidin- 5-yl)methanone;
8-oxa-2-azaspiro[4.5]dec-2-yl[2-({[4-(trifluoromethyl)pyrimidin-5- yl]methyl}amino)pyrimidin-5-yl]methanone; [(3R,5S)-3-hydroxy-5-(trifluoromethyl)pipendin-1 -yl](2-{[1 -(pynmidin-5^ yl)cyclopropyl]amino}pyrimidin-5-yl)methanone;
1 ,4-oxazepan-4-yl(2-{[1 -(pynmidin-5-yl)cyclopropyl]amino}pyrimidin-5- yl)methanone;
2-oxa-5-azabicyclo[2.2.2]oct-5-yl(2-{[1 -(pyrimidin-5- yl)cyclopropyl]amino}pyrimidin-5-yl)methanone;
5,6-dihydroimidazo[1 ,2-a]pyrazin-7(8H)-yl{2-[(pyndin-3- ylmethyl)amino]pyrimidin-5-yl}methanone;
[(2S)-2-methylmorpholin-4-yl](2-{[1 -(pyrimidin-5- yl)cyclopropyl]amino}pyrimidin-5-yl)methanone;
(3-fluoropipendin-1 -yl)(2-{[1 -(pynmidin-5-yl)cyclopropyl]amino}pyrimidin-5- yl)methanone;
(6-hydroxy-2-azaspiro[3.3]hept-2-yl)(2-{[1 -(pyrimidin-5- yl)cyclopropyl]amino}pyrimidin-5-yl)methanone;
octahydropyrazino[1 ,2-a]azepin-2(1 H)-yl(2-{[1 -(pyrimidin-5- yl)cyclopropyl]amino}pyrimidin-5-yl)methanone;
1 -oxa-7-azaspiro[3.5]non-7-yl(2-{[1 -(pyrimidin-5- yl)cyclopropyl]amino}pyrimidin-5-yl)methanone;
2-oxa-7-azaspiro[3.5]non-7-yl(2-{[1 -(pyrimidin-5- yl)cyclopropyl]amino}pyrimidin-5-yl)methanone;
(4-hydroxy-4-methylpiperidin-1 -yl){2-[(pyndin-3-ylmethyl)amino]pynmidin^ yl}methanone;
[4-(ethoxymethyl)-4-fluoropiperidin-1 -yl](2-{[1 -(pynmidin-5- yl)cyclopropyl]amino}pyrimidin-5-yl)methanone;
7-oxa-2-azaspiro[3.5]non-2-yl(2-{[(1 R)-1 -(pyrazin-2-yl)ethyl]amino}pyrimidin-
5-yl)methanone;
[4-(hydroxymethyl)-4-methylpipendin-1 -yl](2-{[1 -(pyrimidin-5- yl)cyclopropyl]amino}pyrimidin-5-yl)methanone;
7-oxa-2-azaspiro[3.5]non-2-yl[2-({[4-(trifluoromethyl)pyrimidin-5- yl]methyl}amino)pyrimidin-5-yl]methanone;
6-oxa-9-azaspiro[4.5]dec-9-yl(2-{[1 -(pyrimidin-5- yl)cyclopropyl]amino}pyrimidin-5-yl)methanone;
(6-methyl-1 ,4-oxazepan-4-yl)(2-{[1 -(pyrimidin-5- yl)cyclopropyl]amino}pyrimidin-5-yl)methanone; [(3R,4R)-3-fluoro-4-methoxypyrrolidin-1 -yl](2-{[1 -(pynmidin-5- yl)cyclopropyl]amino}pynmidin-5-yl)methanone;
(3-ethoxypyrrolidin-1 -yl){2-[(pyridin-3-ylmethyl)amino]pyrimidin-5- yl}methanone;
(2-{[1 -(pynmidin-5-yl)cyclopropyl]amino}pynmidin-5-yl)(pyrrolidin-1 - yl)methanone;
[3-(hydroxymethyl)pyrrolidin-1 -yl]{2-[(pyridin-3-ylmethyl)amino]pynmi yl}methanone;
piperidin-1 -yl{2-[(pynmidin-5-ylmethyl)amino]pynmidin-5-yl}m
(2-{[(1 R)-1 -(6-methylpyndin-3-yl)ethyl]amino}pyrimidin-5-yl)(8-oxa-2- azaspiro[4.5]dec-2-yl)methanone;
7-oxa-2-azaspiro[3.5]non-2-yl(2-{[(1 R)-1 -(pyrimidin-5- yl)ethyl]amino}pyrimidin-5-yl)methanone;
[(8-anti)-8-methoxy-3-azabicyclo[3.2.1 ]oct-3-yl]{2-[(pyridin-3- ylmethyl)amino]pynmidin-5-yl}methanone;
[(3S)-3-methoxypyrrolidin-1 -yl](2-{[1 -(pyrimidin-5- yl)cyclopropyl]amino}pynmidin-5-yl)methanone;
[(7S,8aS)-7-fluorohexahydropyrrolo[1 ,2-a]pyrazin-2(1 H)-yl]{2-[(pyrazin-2- ylmethyl)amino]pynmidin-5-yl}methanone;
[(7R,8aS)-7-hydroxyhexahydropyrrolo[1 ,2-a]pyrazin-2(1 H)-yl]{2-[(pyrazin-2- ylmethyl)amino]pynmidin-5-yl}methanone;
[(1 S,4S)-5-methyl-2,5-diazabicyclo[2.2.1 ]hept-2-yl]{2-[(pyrazin-2- ylmethyl)amino]pynmidin-5-yl}methanone;
[(3S,4S)-3-hydroxy-4-(morpholin-4-yl)pyrrolidin-1 -yl](2-{[(1 S)-1 -(pyrazin-2- yl)ethyl]amino}pynmidin-5-yl)methanone;
(8aR)-hexahydropyrrolo[1 ,2-a]pyrazin-2(1 H)-yl(2-{[(1 S)-1 -(pyrazin-2- yl)ethyl]amino}pynmidin-5-yl)methanone;
[(3S,4S)-3-hydroxy-4-(morpholin-4-yl)pyrrolidin-1 -yl]{2-[(pyrazin-2- ylmethyl)amino]pynmidin-5-yl}methanone;
[(8aS)-7,7-difluorohexahydropyrrolo[1 ,2-a]pyrazin-2(1 H)-yl]{2-[(pyrazin-2- ylmethyl)amino]pynmidin-5-yl}methanone;
(7-ethyl-2,7-diazaspiro[4.4]non-2-yl)(2-{[(1 S)-1 -(pyrazin-2- yl)ethyl]amino}pynmidin-5-yl)methanone;
[(8aS)-7,7-difluorohexahydropyrrolo[1 ,2-a]pyrazin-2(1 H)-yl](2-{[(1 S)-1 - (pyrazin-2 -yl)ethyl]amino}pynmidin-5-yl)methanone; (2-methyl-2,6-diazaspiro[3.4]oct-6-yl){2-[(pyrazin-2-ylmethyl)amino]pynmidin- 5-yl}methanone;
[(3R)-3-(morpholin-4-yl)pyrrolidin-1 -yl](2-{[(1 S)-1 -(pyrazin-2- yl)ethyl]amino}pyrimidin-5-yl)methanone;
[(7S,8aS)-7-fluorohexahydropyrrolo[1 ,2-a]pyrazin-2(1 H)-yl](2-{[(1 S)-1 -
(pyrazin-2-yl)ethyl]amino}pyrimidin-5-yl)methanone;
(2-methyl-2,6-diazaspiro[3.4]oct-6-yl)(2-{[(1 S)-1 -(pyrazin-2- yl)ethyl]amino}pyrimidin-5-yl)methanone;
[(3aR,6aS)-5-methylhexahydropyrrolo[3,4-c]pyrrol-2(1 H)-yl]{2-[(pyrazin-2- ylmethyl)amino]pyrimidin-5-yl}methanone;
[(3R)-3-(morpholin-4-yl)pyrrolidin-1 -yl]{2-[(pyrazin-2-ylmethyl)amino]pyrimi 5-yl}methanone;
(8aS)-hexahydropyrrolo[1 ,2-a]pyrazin-2(1 H)-yl(2-{[(1 S)-1 -(pyrazin-2- yl)ethyl]amino}pyrimidin-5-yl)methanone;
[(1 S,4S)-5-methyl-2,5-diazabicyclo[2.2.1 ]hept-2-yl](2-{[(1 S)-1 -(pyrazin-2- yl)ethyl]amino}pyrimidin-5-yl)methanone;
[(3aR,6aS)-5-methylhexahydropyrrolo[3,4-c]pyrrol-2(1 H)-yl](2-{[(1 S)-1 - (pyrazin-2-yl)ethyl]amino}pyrimidin-5-yl)methanone;
(8aR)-hexahydropyrrolo[1 ,2-a]pyrazin-2(1 H)-yl{2-[(pyrazin-2- ylmethyl)amino]pyrimidin-5-yl}methanone;
(8aS)-hexahydropyrrolo[1 ,2-a]pyrazin-2(1 H)-yl{2-[(pyrazin-2- ylmethyl)amino]pyrimidin-5-yl}methanone;
((6S,7S)-7-hydroxy-2-azaspiro[5.5]undecan-2-yl)(2-((1 -(pyrimidin-5- yl)cyclopropyl)amino)pyrimidin-5-yl)methanone;
or a pharmaceutically acceptable salt thereof.
32. The compound of Claim 20 selected from
7- oxa-2-azaspiro[3.5]non-2-yl(2-{[1 -(pyrazin-2-yl)cyclobutyl]amino}pyrimidin- 5-yl)methanone;
8-oxa-2-azaspiro[4.5]dec-2-yl(2-{[1 -(pyrazin-2-yl)cyclobutyl]amino}pyrimidin- 5-yl)methanone;
8- oxa-2-azaspiro[4.5]dec-2-yl(2-{[2-(pyrazin-2-yl)propan-2-yl]amino}pyrimidin- 5-yl)methanone;
8-oxa-2-azaspiro[4.5]dec-2-yl(2-{[2-(pyrimidin-5-yl)propan-2- yl]amino}pyrimidin-5-yl)methanone; 7- oxa-2-azaspiro[3.5]non-2-yl(2-{[2-(pyrazin-2-yl)propan-2-yl]amino}pyrimidin- 5-yl)methanone;
8- oxa-2-azaspiro[4.5]dec-2-yl(2-{[1 -(pyrimidin-5- yl)cyclobutyl]amino}pyrimidin-5-yl)methanone;
(2-{[2-(5-methylpyrazin-2-yl)propan-2-yl]amino}pyrimidin-5-yl)(7-oxa-2- azaspiro[3.5]non-2-yl)methanone;
8-oxa-2-azaspiro[4.5]dec-2-yl(2-{[(1 S)-1 -(pyrazin-2-yl)ethyl]amino}pyrimidin- 5-yl)methanone;
(2-{[1 -(6-methylpyndin-3-yl)cyclopropyl]amino}pyrimidin-5-yl)(8-oxa-2- azaspiro[4.5]dec-2-yl)methanone;
8-oxa-2-azaspiro[4.5]dec-2-yl(2-{[1 -(pyrimidin-5- yl)cyclopropyl]amino}pyrimidin-5-yl)methanone;
(2-{[(1 S)-1 -(6-methylpyndin-3-yl)ethyl]amino}pyrimidin-5-yl)(8-oxa-2- azaspiro[4.5]dec-2-yl)methanone;
7-oxa-2-azaspiro[3.5]non-2-yl(2-{[1 -(pyrimidin-5- yl)cyclobutyl]amino}pyrimidin-5-yl)methanone;
8-oxa-2-azaspiro[4.5]dec-2-yl(2-{[3-(pyrazin-2-yl)oxetan-3-yl]amino}pyrimidin- 5-yl)methanone;
8-oxa-2-azaspiro[4.5]dec-2-yl(2-{[3-(pyrimidin-5-yl)oxetan-3- yl]amino}pyrimidin-5-yl)methanone;
7-oxa-2-azaspiro[3.5]non-2-yl(2-{[3-(pyrimidin-5-yl)oxetan-3- yl]amino}pyrimidin-5-yl)methanone;
(2-{[1 -(5-methylpyndin-2-yl)ethyl]amino}pyrimidin-5-yl)(8-oxa-2- azaspiro[4.5]dec-2-yl)methanone;
8-oxa-2-azaspiro[4.5]dec-2-yl(2-{[1 -(pyndin-2-yl)cyclopropyl]amino}pyrimidin- 5-yl)methanone;
7-oxa-2-azaspiro[3.5]non-2-yl(2-{[2-(pyrimidin-5-yl)propan-2- yl]amino}pyrimidin-5-yl)methanone;
7- oxa-2-azaspiro[3.5]non-2-yl(2-{[(1 S)-1 -(pyrazin-2-yl)ethyl]amino}pyrimidin- 5-yl)methanone;
8- oxa-2-azaspiro[4.5]dec-2-yl(2-{[(1 S)-1 -(pynmidin-5-yl)ethyl]amino}pyrimidin- 5-yl)methanone;
(2-{[1 -(6-methylpyndin-3-yl)ethyl]amino}pyrimidin-5-yl)(8-oxa-2- azaspiro[4.5]dec-2-yl)methanone; 8-oxa-2-azaspiro[4.5]dec-2-yl(2-{[1 -(pyrimidin-5-yl)ethyl]amino}pyrimidin-5- yl)methanone;
(2-{[1 -(6-methylpyndin-3-yl)cyclopropyl]amino}pyrimidin-5-yl)(7-oxa-2- azaspiro[3.5]non-2-yl)methanone;
7-oxa-2-azaspiro[3.5]non-2-yl(2-{[1 -(pyrimidin-5- yl)cyclopropyl]amino}pyrimidin-5-yl)methanone;
8-oxa-2-azaspiro[4.5]dec-2-yl(2-{[1 -(pyrazin-2-yl)cyclopropyl]amino}pyrimidin- 5-yl)methanone;
7-oxa-2-azaspiro[3.5]non-2-yl(2-{[(1 S)-1 -(pyrimidin-5- yl)ethyl]amino}pyrimidin-5-yl)methanone;
7-oxa-2-azaspiro[3.5]non-2-yl(2-{[1 -(pyrazin-2-yl)cyclopropyl]amino}pyrimidin- 5-yl)methanone;
(2-{[1 -(2-methylpynmidin-5-yl)ethyl]amino}pyrimidin-5-yl)(8-oxa-2- azaspiro[4.5]dec-2-yl)methanone;
8-oxa-2-azaspiro[4.5]dec-2-yl{2-[(pyndin-3-ylmethyl)amino]pyrimidin-5- yl}methanone;
7-oxa-2-azaspiro[3.5]non-2-yl{2-[(pyndin-3-ylmethyl)amino]pyrimidin-5- yl}methanone;
7- oxa-2-azaspiro[3.5]non-2-yl(2-{[1 -(pynmidin-5-yl)ethyl]amino}pyrimidin-5- yl)methanone;
(2-{[1 -(6-methylpyndin-3-yl)ethyl]amino}pyrimidin-5-yl)(7-oxa-2- azaspiro[3.5]non-2-yl)methanone;
8- oxa-2-azaspiro[4.5]dec-2-yl(2-{[1 -(pyrimidin-2- yl)cyclopropyl]amino}pyrimidin-5-yl)methanone;
(2-{[1 -(2-methylpynmidin-5-yl)ethyl]amino}pyrimidin-5-yl)(7-oxa-2- azaspiro[3.5]non-2-yl)methanone;
7- oxa-2-azaspiro[3.5]non-2-yl{2-[(pyrazin-2-ylmethyl)amino]pyrimidin-5- yl}methanone;
8- oxa-2-azaspiro[4.5]dec-2-yl(2-{[(1 S)-1 -(pyndazin-4-yl)ethyl]amino}pyrimidin- 5-yl)methanone;
8-oxa-2-azaspiro[4.5]dec-2-yl(2-{[(1 S)-1 -(pynmidin-4-yl)ethyl]amino}pyrimidin- 5-yl)methanone;
(2-{[(6-methylpyridin-3-yl)methyl]amino}pyrimidin-5-yl)(7-oxa-2- azaspiro[3.5]non-2-yl)methanone; (2-{[(6-methylpyridin-3-yl)methyl]amino}pyrimidin-5-yl)(8-oxa-2- azaspiro[4.5]dec-2-yl)methanone;
7- oxa-2-azaspiro[3.5]non-2-yl{2-[(pyrimidi
yl}methanone;
(2-{[(2-methylpyrimidin-5-yl)methyl]amino}py^
azaspiro[4.5]dec-2-yl)methanone;
8- oxa-2-azaspiro[4.5]dec-2-yl{2-[(pynmidin-5-ylmethyl)amino]pyrimidin-5- yl}methanone;
8-oxa-2-azaspiro[4.5]dec-2-yl{2-[(pyrazin-2-ylmethyl)amino]pyrimidin-5- yl}methanone;
or a pharmaceutically acceptable salt thereof.
33. A pharmaceutical composition comprising a compound of any of the preceding claims, or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable vehicle, diluent, carrier or other excipient.
34. A method of treating a disease or disorder treating mediated by, or otherwise associated with, inhibition of the vanin-1 enzyme, comprising administering to a patient in need thereof a therapeutically effective amount of a compound of any of the preceding claims, or a pharmaceutically acceptable salt thereof.
35. The method of Claim 34 wherein the disease or disorder is selected from: auto-immune diseases, inflammatory diseases, allergic diseases, metabolic diseases, infection-based diseases, fibrotic diseases, cardiovascular diseases, respiratory diseases, renal diseases, dermatological diseases, liver diseases, gastrointestinal diseases, oral diseases, and hematopoietic diseases.
36. The method of Claim 35 wherein the disease or disorder is selected from the group consisting of disease or disorder is selected from the group consisting of inflammatory bowel disease, ulcerative colitis, Crohn's disease, colorectal cancer, and gastritis.
37. A pharmaceutical combination comprising a therapeutically effective amount of a composition comprising: a first compound, the first compound being a compound of any of the preceding claims or a pharmaceutically acceptable salt thereof;
a second compound being selected from an approved drug or a clinical candidate useful for the treatment of infectious or inflammatory diseases; and
an optional pharmaceutically acceptable carrier, vehicle or diluent.
38. A pharmaceutical combination comprising a therapeutically effective amount of a composition comprising:
a first compound, the first compound being a compound of any of the preceding claims or a pharmaceutically acceptable salt thereof; a second compound, the second compound being selected from the group consisting of antibodies or small molecules which include but are not limited to those that block the action of specific cytokines such as TNFa, IL12 and/or IL23, or inhibitors of leukocyte recruitment such as modulators of S1 P receptors or integrin antagonists, or selective or non-selective inhibitors of the JAK kinases JAK1 , JAK2, JAK3 and/ or TYK2, inhibitors of leukocyte function such as PDE4 or SMAD7.
39. The pharmaceutical composition of Claim 38 wherein the second compound is selected from
(a) an anti-TNFa agent selected from infliximab, adalimumab, golimumab, and certolizumab pegol;
(b) an anti-IL-12 and/or IL-23 agent selected from ustekinumab;
(c) a modulator of S1 P receptors selected from ozanimod;
(d) an integrin antagonist selected from vedolizumab, etrolizumab, and natalizumab;
(e) an inhibitor of JAK kinases selected from tofacitinib, filgotinib, PF- 04965842, PF-06651600, and PF-06263276;
(f) a PDE4 inhibitor selected from apremilast; or
(g) a SMAD7 antisense oligonucleotides selected from mongersen.
40. A compound selected from
[(8aS)-7,7-difluorohexahydropyrrolo[1 ,2-a]pyrazin-2(1 H)-yl](2-{[(1 S)-1 - (pyrazin-2-yl)ethyl]amino}pyrimidin-5-yl)methanone; [(7S,8aS)-7-fluorohexahydropyrrolo[1 ,2-a]pyrazin-2(1 H)-yl](2-{[(1 S)-1 - (pyrazin-2-yl)ethyl]amino}pyrimidin-5-yl)methanone;
(8aS)-hexahydropyrrolo[1 ,2-a]pyrazin-2(1 H)-yl(2-{[(1 S)-1 -(pyrazin-2- yl)ethyl]amino}pyrimidin-5-yl)methanone;
8-oxa-2-azaspiro[4.5]dec-2-yl(2-{[1 -(pyrazin-2-yl)cyclobutyl]amino}pyrimidin- 5-yl)methanone;
7- oxa-2-azaspiro[3.5]non-2-yl(2-{[2-(pyrazin-2-yl)propan-2-yl]amino}pyrimidin- 5-yl)methanone;
8- oxa-2-azaspiro[4.5]dec-2-yl(2-{[3-(pyrimidin-5-yl)oxetan-3- yl]amino}pyrimidin-5-yl)methanone;
7-oxa-2-azaspiro[3.5]non-2-yl{2-[(pyrazin-2-ylmethyl)amino]pyrimidin-5- yl}methanone;
(2-{[(6-methylpyridin-3-yl)methyl]amino}pyrimidin-5-yl)(8-oxa-2- azaspiro[4.5]dec-2-yl)methanone;
8-oxa-2-azaspiro[4.5]dec-2-yl{2-[(pyrazin-2-ylmethyl)amino]pyrimidin-5- yl}methanone;
7- oxa-2-azaspiro[3.5]non-2-yl(2-{[2-(pyrimidin-5-yl)propan-2- yl]amino}pyrimidin-5-yl)methanone;
8- oxa-2-azaspiro[4.5]dec-2-yl(2-{[(1 S)-1 -(pyrazin-2-yl)ethyl]amino}pyrimidin- 5-yl)methanone;
7- oxa-2-azaspiro[3.5]non-2-yl(2-{[1 -(pyrazin-2-yl)cyclopropyl]amino}pyrimidin- 5-yl)methanone;
8- oxa-2-azaspiro[4.5]dec-2-yl(2-{[1 -(pyndin-2-yl)cyclopropyl]amino}pyrimidin- 5-yl)methanone;
(2-{[2-(5-methylpyrazin-2-yl)propan-2-yl]amino}pyrimidin-5-yl)(7-oxa-2- azaspiro[3.5]non-2-yl)methanone; and
8-oxa-2-azaspiro[4.5]dec-2-yl(2-{[1 -(pyrimidin-5- yl)cyclopropyl]amino}pyrimidin-5-yl)methanone;
or a pharmaceutically acceptable salt of said compounds.
PCT/IB2017/054104 2016-07-14 2017-07-07 Novel pyrimidine carboxamides as inhibitors of vanin-1 enzyme WO2018011681A1 (en)

Priority Applications (12)

Application Number Priority Date Filing Date Title
EP17740116.3A EP3484876A1 (en) 2016-07-14 2017-07-07 Novel pyrimidine carboxamides as inhibitors of vanin-1 enzyme
MX2019000536A MX2019000536A (en) 2016-07-14 2017-07-07 Novel pyrimidine carboxamides as inhibitors of vanin-1 enzyme.
CA3030381A CA3030381A1 (en) 2016-07-14 2017-07-07 Pyrimidine carboxamides as inhibitors of vanin-1 enzyme
JP2019500621A JP2019524716A (en) 2016-07-14 2017-07-07 Novel pyrimidinecarboxamides as inhibitors of vanin 1 enzyme
BR112019000589-7A BR112019000589A2 (en) 2016-07-14 2017-07-07 pyrimidine carboxamides as inhibitors of vanin-1 enzyme
CN201780043514.7A CN109476645A (en) 2016-07-14 2017-07-07 New pyrimidine carboxamide as VANIN-1 enzyme inhibitor
KR1020197004221A KR20190026902A (en) 2016-07-14 2017-07-07 Novel pyrimidinecarboxamide as an inhibitor of vanin-1 enzyme
RU2019100559A RU2019100559A (en) 2016-07-14 2017-07-07 NEW PYRIMIDINE CARBOXAMIDES AS INHIBITORS OF THE ENZYM VANIN-1
SG11201811161YA SG11201811161YA (en) 2016-07-14 2017-07-07 Novel pyrimidine carboxamides as inhibitors of vanin-1 enzyme
AU2017296338A AU2017296338A1 (en) 2016-07-14 2017-07-07 Novel pyrimidine carboxamides as inhibitors of vanin-1 enzyme
US16/316,688 US10906888B2 (en) 2016-07-14 2017-07-07 Pyrimidine carboxamides as inhibitors of Vanin-1 enzyme
IL263662A IL263662A (en) 2016-07-14 2018-12-12 Novel pyrimidine carboxamides as inhibitors of vanin-1 enzyme

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201662362098P 2016-07-14 2016-07-14
US62/362,098 2016-07-14

Publications (1)

Publication Number Publication Date
WO2018011681A1 true WO2018011681A1 (en) 2018-01-18

Family

ID=59351004

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2017/054104 WO2018011681A1 (en) 2016-07-14 2017-07-07 Novel pyrimidine carboxamides as inhibitors of vanin-1 enzyme

Country Status (14)

Country Link
US (1) US10906888B2 (en)
EP (1) EP3484876A1 (en)
JP (1) JP2019524716A (en)
KR (1) KR20190026902A (en)
CN (1) CN109476645A (en)
AU (1) AU2017296338A1 (en)
BR (1) BR112019000589A2 (en)
CA (1) CA3030381A1 (en)
IL (1) IL263662A (en)
MA (1) MA45656A (en)
MX (1) MX2019000536A (en)
RU (1) RU2019100559A (en)
SG (1) SG11201811161YA (en)
WO (1) WO2018011681A1 (en)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018228934A1 (en) 2017-06-12 2018-12-20 Boehringer Ingelheim International Gmbh Heteroaromatic compounds as vanin inhibitors
WO2019133445A1 (en) * 2017-12-28 2019-07-04 Inception Ibd, Inc. Aminothiazoles as inhibitors of vanin-1
WO2020043658A1 (en) 2018-08-28 2020-03-05 Boehringer Ingelheim International Gmbh Heteroaromatic compounds as vanin inhibitors
WO2020114949A1 (en) 2018-12-03 2020-06-11 Boehringer Ingelheim International Gmbh Heteroaromatic compounds as vanin inhibitors
WO2020114947A1 (en) 2018-12-03 2020-06-11 Boehringer Ingelheim International Gmbh Heteroaromatic compounds as vanin inhibitors
WO2020114943A1 (en) 2018-12-03 2020-06-11 Boehringer Ingelheim International Gmbh Heteroaromatic compounds as vanin inhibitors
CN112703037A (en) * 2018-09-04 2021-04-23 施万生物制药研发Ip有限责任公司 5-to 7-membered heterocyclic amides as JAK inhibitors
US11713325B2 (en) 2017-04-27 2023-08-01 Pharma Mar, S.A. Antitumoral compounds
TWI811901B (en) * 2020-12-17 2023-08-11 大陸商上海美悅生物科技發展有限公司 A kind of pyrimidine formamide compound and application thereof
TWI838430B (en) 2018-12-03 2024-04-11 德商百靈佳殷格翰國際股份有限公司 Heteroaromatic compounds as vanin inhibitors

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111388452B (en) * 2020-04-30 2021-10-26 中国药科大学 Adipose tissue targeting peptide P3-chitosan oligomeric lactic acid-polyethylene glycol delivery system and application thereof in nucleic acid drug delivery
CN114933594A (en) * 2022-07-20 2022-08-23 北京科翔中升医药科技有限公司 Fluotriazines compound, pharmaceutical composition and application

Citations (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994022823A1 (en) 1993-03-26 1994-10-13 Ortho Pharmaceutical Corporation 4-arylisoindole analgesics
US5612359A (en) 1994-08-26 1997-03-18 Bristol-Myers Squibb Company Substituted biphenyl isoxazole sulfonamides
WO2000001389A1 (en) 1998-07-06 2000-01-13 Bristol-Myers Squibb Co. Biphenyl sulfonamides as dual angiotensin endothelin receptor antagonists
US6043265A (en) 1997-01-30 2000-03-28 Bristol-Myers Squibb Co. Isoxazolyl endothelin antagonists
WO2001040231A1 (en) 1999-12-06 2001-06-07 Bristol-Myers Squibb Company Heterocyclic dihydropyrimidines as potassium channel inhibitors
WO2003072197A1 (en) 2002-02-27 2003-09-04 Pfizer Products Inc. Acc inhibitors
WO2005116014A1 (en) 2004-05-12 2005-12-08 Pfizer Products Inc. Proline derivatives and their use as dipeptidyl peptidase iv inhibitors
WO2007122482A1 (en) 2006-04-20 2007-11-01 Pfizer Products Inc. Fused phenyl amido heterocyclic compounds for the prevention and treatment of glucokinase-mediated diseases
WO2008065508A1 (en) 2006-11-29 2008-06-05 Pfizer Products Inc. Spiroketone acetyl-coa carboxylase inhibitors
WO2009016462A2 (en) 2007-08-02 2009-02-05 Pfizer Products Inc. Substituted bicyclolactam compounds
WO2009144554A1 (en) 2008-05-28 2009-12-03 Pfizer, Inc. Pyrazolospiroketone acetyl-c0a carboxylase inhibitors
WO2009144555A1 (en) 2008-05-28 2009-12-03 Pfizer Inc. Pyrazolospiroketone acetyl-coa carboxylase inhibitors
WO2010013161A1 (en) 2008-07-29 2010-02-04 Pfizer Inc. Fluorinated heteroaryls
WO2010023594A1 (en) 2008-08-28 2010-03-04 Pfizer Inc. Dioxa-bicyclo[3.2.1.]octane-2,3,4-triol derivatives
WO2010086820A1 (en) 2009-02-02 2010-08-05 Pfizer Inc. 4-amino-5-oxo-7, 8-dihydropyrimido [5,4-f] [1,4] oxazepin-6 (5h) -yl) phenyl derivatives, pharmaceutical compositions and uses thereof
WO2010103438A1 (en) 2009-03-11 2010-09-16 Pfizer Inc. Substituted indazole amides and their use as glucokinase activators
WO2010103437A1 (en) 2009-03-11 2010-09-16 Pfizer Inc. Benzofuranyl derivatives used as glucokinase inhibitors
WO2010106457A2 (en) 2009-03-20 2010-09-23 Pfizer Inc. 3-oxa-7-azabicyclo[3.3.1]nonanes
WO2010128425A1 (en) 2009-05-08 2010-11-11 Pfizer Inc. Gpr 119 modulators
WO2010128414A1 (en) 2009-05-08 2010-11-11 Pfizer Inc. Gpr 119 modulators
WO2010140092A1 (en) 2009-06-05 2010-12-09 Pfizer Inc. L- ( piperidin-4-yl) -pyrazole derivatives as gpr 119 modulators
WO2011005611A1 (en) 2009-07-09 2011-01-13 Merck Sharp & Dohme Corp. Neuromedin u receptor agonists and uses thereof
US20110053912A1 (en) * 2009-03-30 2011-03-03 Astellas Pharma Inc. Pyrimidine compound
WO2014048547A1 (en) 2012-09-25 2014-04-03 Merck Patent Gmbh Alpha hydroxy amides
WO2016193844A1 (en) * 2015-05-29 2016-12-08 Pfizer Inc. Novel heterocyclic compounds as inhibitors of vanin-1 enzyme

Family Cites Families (42)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5811428A (en) 1995-12-18 1998-09-22 Signal Pharmaceuticals, Inc. Pyrimidine carboxamides and related compounds and methods for treating inflammatory conditions
EP2308833A3 (en) 1999-04-15 2011-09-28 Bristol-Myers Squibb Company Cyclic protein tyrosine kinase inhibitors
RU2267484C2 (en) 1999-04-28 2006-01-10 Авентис Фарма Дойчланд Гмбх Derivatives of diarylic acid and pharmaceutical composition based on thereof
AU5723500A (en) 1999-05-24 2000-12-12 Cor Therapeutics, Inc. Inhibitors of factor xa
WO2001019802A1 (en) 1999-09-16 2001-03-22 Tanabe Seiyaku Co., Ltd. Aromatic nitrogenous six-membered ring compounds
KR100711042B1 (en) 2000-04-28 2007-04-24 다나베 세이야꾸 가부시키가이샤 Cyclic compounds
US20020183316A1 (en) 2000-10-27 2002-12-05 Kevin Pan Amidoalkyl-piperidine and amidoalkyl-piperazine derivatives useful for the treatment of nervous system disorders
US6770647B2 (en) 2001-08-17 2004-08-03 Bristol-Myers Squibb Pharma Company Bicyclic hydroxamates as inhibitors of matrix metalloproteinases and/or TNF-α converting enzyme (TACE)
US20030195192A1 (en) 2002-04-05 2003-10-16 Fortuna Haviv Nicotinamides having antiangiogenic activity
AU2003228701A1 (en) 2002-04-25 2003-11-10 Bristol-Myers Squibb Company Spirobarbituric acid derivatives useful as inhibitors of matrix metalloproteases
AU2004234258B2 (en) 2003-04-28 2009-04-09 Tacurion 4,4-difluoro-1,2,3,4-tetrahydro-5H-1-benzazepine derivative or salt thereof
MY141220A (en) 2003-11-17 2010-03-31 Astrazeneca Ab Pyrazole derivatives as inhibitors of receptor tyrosine kinases
WO2005075482A1 (en) 2004-02-04 2005-08-18 Neurosearch A/S Diazabicyclic aryl derivatives as nicotinic acetylcholine receptor ligands
US7435831B2 (en) 2004-03-03 2008-10-14 Chemocentryx, Inc. Bicyclic and bridged nitrogen heterocycles
US7435830B2 (en) 2004-03-03 2008-10-14 Chemocentryx, Inc. Bicyclic and bridged nitrogen heterocycles
US7423147B2 (en) 2004-03-31 2008-09-09 Janssen Pharmaceutical, N.V. Pyridine compounds as histamine H3 modulators
EP1761542B1 (en) 2004-06-09 2008-01-02 F.Hoffmann-La Roche Ag Octahydropyrrolo[3,4-c]pyrrole derivatives an their use as antiviral agents
WO2006044174A2 (en) 2004-10-13 2006-04-27 Neurogen Corporation Aryl substituted 8-azabicyclo[3.2.1]octane compounds as ligands of the melanin concentrating hormone receptor
JP4765545B2 (en) 2004-10-27 2011-09-07 アステラス製薬株式会社 Pharmaceutical composition comprising a benzazepine derivative as an active ingredient
PE20071025A1 (en) 2006-01-31 2007-10-17 Mitsubishi Tanabe Pharma Corp TRISUSTITUTED AMINE COMPOUND
CN101384596A (en) 2006-02-15 2009-03-11 弗·哈夫曼-拉罗切有限公司 Heterocylic antiviral compounds
KR100909953B1 (en) 2007-06-15 2009-07-31 한국화학연구원 Triazole derivatives having antifungal activity, method for the preparation thereof and pharmaceutical composition containing same
WO2008157552A2 (en) 2007-06-18 2008-12-24 Mayo Foundation For Medical Education And Research Invertebrate acetylcholinesterase inhibitors
AU2008281509A1 (en) 2007-08-02 2009-02-05 Pfizer Inc. Pyrimidine and pyridine derivatives and their pharmaceutical use and compositions
UY32055A (en) 2008-08-19 2010-03-26 Astrazeneca Ab Substituted derivatives of 5-Halo-N2- [substituted] (1-methyl-1H-imidazol-4-yl) pyrimidin-2, -diamine and its pharmaceutically acceptable salts, preparation processes, compositions and applications.
WO2010132999A1 (en) 2009-05-21 2010-11-25 Chlorion Pharma, Inc. Methyl sulfanyl pyrmidmes useful as antiinflammatories, analgesics, and antiepileptics
AR081331A1 (en) 2010-04-23 2012-08-08 Cytokinetics Inc AMINO- PYRIMIDINES COMPOSITIONS OF THE SAME AND METHODS FOR THE USE OF THE SAME
US9133123B2 (en) 2010-04-23 2015-09-15 Cytokinetics, Inc. Certain amino-pyridines and amino-triazines, compositions thereof, and methods for their use
AR081626A1 (en) 2010-04-23 2012-10-10 Cytokinetics Inc AMINO-PYRIDAZINIC COMPOUNDS, PHARMACEUTICAL COMPOSITIONS THAT CONTAIN THEM AND USE OF THE SAME TO TREAT CARDIAC AND SKELETIC MUSCULAR DISORDERS
JP5782430B2 (en) 2010-04-27 2015-09-24 田辺三菱製薬株式会社 Novel amide derivatives and their use as pharmaceuticals
CN103038229B (en) 2010-05-26 2016-05-11 桑诺维恩药品公司 Heteroaryl compound and using method thereof
EP2598483B1 (en) 2010-07-29 2020-07-29 Rigel Pharmaceuticals, Inc. Ampk-activating heterocyclic compounds and methods for using the same
WO2012031090A2 (en) 2010-09-01 2012-03-08 President And Fellows Of Harvard College Small molecule inhibitors of ebola and lassa fever viruses and methods of use
US20130310379A1 (en) 2010-11-19 2013-11-21 Constellation Pharmaceuticals Modulators of methyl modifying enzymes, compositions and uses thereof
US20140018540A1 (en) 2010-12-14 2014-01-16 Electrophoretics Limited Casein kinase 1delta (ck 1delta) inhibitors
RU2014115476A (en) 2011-09-20 2015-10-27 Целльзом Лимитед PYRAZOLO [4, 3-C] PTRIDINE DERIVATIVES AS KINASE INHIBITORS
EP2776393B1 (en) * 2011-11-11 2017-12-20 Abbvie Inc. Nampt inhibitors
CA2868507A1 (en) 2012-04-02 2013-10-10 Cytokinetics, Inc. Methods for improving diaphragm function
SG11201406359TA (en) 2012-04-11 2014-11-27 Cytokinetics Inc Improving resistance to skeletal muscle fatigue
US9624197B2 (en) 2012-11-27 2017-04-18 Merck Sharp & Dohme Corp. 2-pyridylamino-4-nitrile-piperidinyl orexin receptor antagonists
WO2014181287A1 (en) 2013-05-09 2014-11-13 Piramal Enterprises Limited Heterocyclyl compounds and uses thereof
US20150087673A1 (en) 2013-09-26 2015-03-26 Rigel Pharmaceuticals, Inc. Methods for using and biomarkers for ampk-activating compounds

Patent Citations (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994022823A1 (en) 1993-03-26 1994-10-13 Ortho Pharmaceutical Corporation 4-arylisoindole analgesics
US5612359A (en) 1994-08-26 1997-03-18 Bristol-Myers Squibb Company Substituted biphenyl isoxazole sulfonamides
US6043265A (en) 1997-01-30 2000-03-28 Bristol-Myers Squibb Co. Isoxazolyl endothelin antagonists
WO2000001389A1 (en) 1998-07-06 2000-01-13 Bristol-Myers Squibb Co. Biphenyl sulfonamides as dual angiotensin endothelin receptor antagonists
WO2001040231A1 (en) 1999-12-06 2001-06-07 Bristol-Myers Squibb Company Heterocyclic dihydropyrimidines as potassium channel inhibitors
WO2003072197A1 (en) 2002-02-27 2003-09-04 Pfizer Products Inc. Acc inhibitors
WO2005116014A1 (en) 2004-05-12 2005-12-08 Pfizer Products Inc. Proline derivatives and their use as dipeptidyl peptidase iv inhibitors
WO2007122482A1 (en) 2006-04-20 2007-11-01 Pfizer Products Inc. Fused phenyl amido heterocyclic compounds for the prevention and treatment of glucokinase-mediated diseases
WO2008065508A1 (en) 2006-11-29 2008-06-05 Pfizer Products Inc. Spiroketone acetyl-coa carboxylase inhibitors
WO2009016462A2 (en) 2007-08-02 2009-02-05 Pfizer Products Inc. Substituted bicyclolactam compounds
WO2009144554A1 (en) 2008-05-28 2009-12-03 Pfizer, Inc. Pyrazolospiroketone acetyl-c0a carboxylase inhibitors
WO2009144555A1 (en) 2008-05-28 2009-12-03 Pfizer Inc. Pyrazolospiroketone acetyl-coa carboxylase inhibitors
WO2010013161A1 (en) 2008-07-29 2010-02-04 Pfizer Inc. Fluorinated heteroaryls
WO2010023594A1 (en) 2008-08-28 2010-03-04 Pfizer Inc. Dioxa-bicyclo[3.2.1.]octane-2,3,4-triol derivatives
WO2010086820A1 (en) 2009-02-02 2010-08-05 Pfizer Inc. 4-amino-5-oxo-7, 8-dihydropyrimido [5,4-f] [1,4] oxazepin-6 (5h) -yl) phenyl derivatives, pharmaceutical compositions and uses thereof
WO2010103438A1 (en) 2009-03-11 2010-09-16 Pfizer Inc. Substituted indazole amides and their use as glucokinase activators
WO2010103437A1 (en) 2009-03-11 2010-09-16 Pfizer Inc. Benzofuranyl derivatives used as glucokinase inhibitors
WO2010106457A2 (en) 2009-03-20 2010-09-23 Pfizer Inc. 3-oxa-7-azabicyclo[3.3.1]nonanes
US20110053912A1 (en) * 2009-03-30 2011-03-03 Astellas Pharma Inc. Pyrimidine compound
WO2010128425A1 (en) 2009-05-08 2010-11-11 Pfizer Inc. Gpr 119 modulators
WO2010128414A1 (en) 2009-05-08 2010-11-11 Pfizer Inc. Gpr 119 modulators
WO2010140092A1 (en) 2009-06-05 2010-12-09 Pfizer Inc. L- ( piperidin-4-yl) -pyrazole derivatives as gpr 119 modulators
WO2011005611A1 (en) 2009-07-09 2011-01-13 Merck Sharp & Dohme Corp. Neuromedin u receptor agonists and uses thereof
WO2014048547A1 (en) 2012-09-25 2014-04-03 Merck Patent Gmbh Alpha hydroxy amides
WO2016193844A1 (en) * 2015-05-29 2016-12-08 Pfizer Inc. Novel heterocyclic compounds as inhibitors of vanin-1 enzyme

Non-Patent Citations (46)

* Cited by examiner, † Cited by third party
Title
"Remington: The Science and Practice of Pharmacy., 21 st Edition", 2005, MACK PUBLISHING COMPANY
"The Pharmacological Basis of Therapeutics, 9th Edition,", 1996, MCGRAW-HILL
BERRUYER C ET AL.: "Vanin-1 licenses inflammatory mediator production by gut epithelial cells and controls colitis by antagonizing peroxisome proliferator-activated receptor y activity", J EXP MED., vol. 203, 2006, pages 2817 - 2827, XP002687503, DOI: doi:10.1084/JEM.20061640
BERRUYER C ET AL.: "Vanin-1-/- mice exhibit a glutathione mediated tissue resistance to oxidative stress", MOL CELL BIOL., vol. 24, 2004, pages 7214 - 7224
BIOORG. MED. CHEM. LETT., vol. 8, 1998, pages 2833
CARPINO, P.A.; GOODWIN, B., EXPERT OPIN. THER. PAT, vol. 20, no. 12, 2010, pages 1627 - 51
CHEN S ET AL.: "Vanin-1 is a key activator for hepatic gluconeogenesis", DIABETES, vol. 63, no. 6, 18 February 2014 (2014-02-18), pages 2073 - 85
D.E. ET AL., ANNUAL REPORTS IN MEDICINAL CHEMISTRY, vol. 43, 2008, pages 119 - 137
E.C. CHAO ET AL., NATURE REVIEWS DRUG DISCOVERY, vol. 9, July 2010 (2010-07-01), pages 551 - 559
EDWARD; B. ROCHE: "Bioreversible Carriers in Drug Design", 1987, AMERICAN PHARMACEUTICAL ASSOCIATION AND PERGAMON PRESS
FEDORAK ET AL., AM. J. PHYSIOL., vol. 269, 1995, pages G210 - 218
FINNIN; MORGAN, J. PHARM. SCI., vol. 88, no. 10, October 1999 (1999-10-01), pages 955 - 958
HALEBLIAN, J PHARM SCI, vol. 64, no. 8, August 1975 (1975-08-01), pages 1269 - 1288
HOCHHAUS ET AL., BIOMED. CHROM., vol. 6, 1992, pages 283 - 286
INFLAMM BOWEL DIS., vol. 19, no. 11, October 2013 (2013-10-01), pages 2315 - 25
J CLIN INVEST., vol. 113, 2004, pages 591 - 597
J. LARSEN ET AL., INT. J. PHARMACEUTICS, vol. 47, 1988, pages 103
J. LARSEN; H. BUNDGAARD, INT. J. PHARMACEUTICS, vol. 37, 1987, pages 87
J. MARCH: "Advanced Organic Chemistry", 1985, WILEY INTERSCIENCE
JONES, R.M. ET AL., MEDICINAL CHEMISTRY, vol. 44, 2009, pages 149 - 170
KASKOW BJ ET AL.: "Diverse biological activities of the vascular non-inflammatory molecules - the Vanin pantetheinases", BIOCHEM BIOPHYS RES COMMUN., vol. 417, no. 2, 13 January 2012 (2012-01-13), pages 653 - 8, XP028441631, DOI: doi:10.1016/j.bbrc.2011.11.099
KHARITONENKOV, A. ET AL., CURRENT OPINION IN INVESTIGATIONAL DRUGS, vol. 10, no. 4, 2009, pages 359 - 364
KIBBE ET AL.,: "Handbook of Pharmaceutical Excipients (3rd Ed.)", 1999, AMERICAN PHARMACEUTICAL ASSOCIATION
LIBERMAN ET AL.: "Pharmaceutical Dosage Forms", 1980, MARCEL DECKER
MCLOED ET AL., GASTROENTEROL, vol. 106, 1994, pages 405 - 413
MEDICINAL CHEMISTRY, vol. 10, no. 4, 2010, pages 386 - 396
MEDINA, J.C., ANNUAL REPORTS IN MEDICINAL CHEMISTRY, vol. 43, 2008, pages 75 - 85
NITTO T; ONODERA K.: "The Linkage between coenzyme A metabolism and inflammation: roles of Pantetheinase", JOURNAL OF PHARMACOLOGICAL SCIENCES, vol. 123, 2013, pages 1 - 8
NOGRADY: "Medicinal Chemistry A Biochemical Approach", 1985, OXFORD UNIVERSITY PRESS, pages: 388 - 392
PITARI G ET AL.: "Pantetheinase activity of membrane-bound vanin-1: lack of free cysteamine in tissues of vanin-1 deficient mice", FEBS LETT., vol. 483, 2000, pages 149 - 154, XP004337693, DOI: doi:10.1016/S0014-5793(00)02110-4
PJ, KOCIENSKI: "Protecting Groups", 1994, GEORG THIEME VERLAG
POUYET L ET AL.: "Epithelial vanin-1 controls inflammation-driven carcinogenesis in the colitis-associated colon cancer model", INFLAMM BOWEL DIS, vol. 16, no. 1, January 2010 (2010-01-01), pages 96 - 104
RC LAROCK: "Comprehensive Organic Transformations", 1989, VCH PUBLISHERS INC.
RK MACKIE; DM SMITH: "Guidebook to Organic Synthesis", 1982, LONGMAN
ROMMELAERE S ET AL.: "PPARalpha regulates the production of serum Vanin-1 by liver", FEBS LETT., vol. 587, no. 22, 15 November 2013 (2013-11-15), pages 3742 - 8, XP028766011, DOI: doi:10.1016/j.febslet.2013.09.046
S WARREN: "Designing Organic Synthesis", 1978, WILEY INTERSCIENCE
S WARREN: "Organic Synthesis - The Disconnection Approach", 1982, WILEY INTERSCIENCE
SAULNIER ET AL., BIOORGANIC AND MEDICINAL CHEMISTRY LETTERS, vol. 4, 1994
SILVERMAN: "The Organic Chemistry of Drug Design and Drug Action", 1992, ACADEMIC PRESS, INC., pages: 352 - 401
SINKULA ET AL., J. PHARM. SCI., vol. 64, 1975, pages 181 - 210
T. HIGUCHI; V. STELLA, PRO-DRUGS AS NOVEL DELIVERY SYSTEMS, vol. 14
T. KOUTSOKERAS; T. HEALY: "Systemic lupus erythematosus and lupus nephritis", NAT REV DRUG DISCOV, vol. 13, no. 3, 2014, pages 173 - 174
THEODORA W GREENE; PETER G M WUTS: "Greene's Protective Groups in Organic Synthesis", 1999, JOHN WILEY AND SONS
TW GREENE; PGM WUTS: "Protective Groups in Organic Synthesis", 1999, JOHN WILEY AND SONS, INC.
Y. MUSHTAQ: "The COPD pipeline", NAT REV DRUG DISCOV, vol. 13, no. 4, 2014, pages 253 - 254, Retrieved from the Internet <URL:http://dx.doi.org/10.1038/nrd425>
ZHANG, S. ET AL., DRUG DISCOVERY TODAY, vol. 12, no. 9/10, 2007, pages 373 - 381

Cited By (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11713325B2 (en) 2017-04-27 2023-08-01 Pharma Mar, S.A. Antitumoral compounds
EA039223B1 (en) * 2017-06-12 2021-12-20 Бёрингер Ингельхайм Интернациональ Гмбх Heteroaromatic compounds as vanin inhibitors
US10364255B2 (en) 2017-06-12 2019-07-30 Boehringer Ingleheim International Gmbh Heteroaromatic compounds as Vanin inhibitors
CN110770242A (en) * 2017-06-12 2020-02-07 勃林格殷格翰国际有限公司 Heteroaromatic compounds as VANIN inhibitors
KR20200013718A (en) * 2017-06-12 2020-02-07 베링거 인겔하임 인터내셔날 게엠베하 Heteroaromatic Compounds as Banin Inhibitors
KR102607934B1 (en) 2017-06-12 2023-12-01 베링거 인겔하임 인터내셔날 게엠베하 Heteroaromatic compounds as vanin inhibitors
WO2018228934A1 (en) 2017-06-12 2018-12-20 Boehringer Ingelheim International Gmbh Heteroaromatic compounds as vanin inhibitors
WO2019133445A1 (en) * 2017-12-28 2019-07-04 Inception Ibd, Inc. Aminothiazoles as inhibitors of vanin-1
JP7148709B2 (en) 2018-08-28 2022-10-05 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Heteroaromatic compounds as vanin inhibitors
JP2021535154A (en) * 2018-08-28 2021-12-16 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Heteroaromatic compounds as vanin inhibitors
CN112601748A (en) * 2018-08-28 2021-04-02 勃林格殷格翰国际有限公司 Heteroaromatic compounds as VANIN inhibitors
TWI826509B (en) * 2018-08-28 2023-12-21 德商百靈佳殷格翰國際股份有限公司 Heteroaromatic compounds as vanin inhibitors
WO2020043658A1 (en) 2018-08-28 2020-03-05 Boehringer Ingelheim International Gmbh Heteroaromatic compounds as vanin inhibitors
US10864201B2 (en) 2018-08-28 2020-12-15 Boehringer Ingelheim International Gmbh Heteroaromatic compounds as Vanin inhibitors
CN113788825A (en) * 2018-08-28 2021-12-14 勃林格殷格翰国际有限公司 Heteroaromatic compounds as VANIN inhibitors
US11713315B2 (en) 2018-09-04 2023-08-01 Theravance Biopharma R&D Ip, Llc 5 to 7 membered heterocyclic amides as JAK inhibitors
CN112703037B (en) * 2018-09-04 2023-10-20 施万生物制药研发Ip有限责任公司 5-to 7-membered heterocyclic amides as JAK inhibitors
CN112703037A (en) * 2018-09-04 2021-04-23 施万生物制药研发Ip有限责任公司 5-to 7-membered heterocyclic amides as JAK inhibitors
WO2020114947A1 (en) 2018-12-03 2020-06-11 Boehringer Ingelheim International Gmbh Heteroaromatic compounds as vanin inhibitors
JP7195436B2 (en) 2018-12-03 2022-12-23 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Heteroaromatic compounds as vanin inhibitors
US20220048889A1 (en) * 2018-12-03 2022-02-17 Boehringer Ingelheim International Gmbh Heteroaromatic compounds as vanin inhibitors
CN114478485A (en) * 2018-12-03 2022-05-13 勃林格殷格翰国际有限公司 Heteroaromatic compounds as VANIN inhibitors
JP7080405B2 (en) 2018-12-03 2022-06-03 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Heteroaromatic compounds as vanine inhibitors
JP7130873B2 (en) 2018-12-03 2022-09-05 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Heteroaromatic compounds as vanin inhibitors
WO2020114943A1 (en) 2018-12-03 2020-06-11 Boehringer Ingelheim International Gmbh Heteroaromatic compounds as vanin inhibitors
JP2022512086A (en) * 2018-12-03 2022-02-02 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Complex aromatic compounds as vanin inhibitors
WO2020114949A1 (en) 2018-12-03 2020-06-11 Boehringer Ingelheim International Gmbh Heteroaromatic compounds as vanin inhibitors
US11078182B2 (en) 2018-12-03 2021-08-03 Boehringer Ingelheim International Gmbh Heteroaromatic compounds as Vanin inhibitors
TWI838430B (en) 2018-12-03 2024-04-11 德商百靈佳殷格翰國際股份有限公司 Heteroaromatic compounds as vanin inhibitors
JP2022510351A (en) * 2018-12-03 2022-01-26 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Complex aromatic compounds as vanin inhibitors
CN113166104A (en) * 2018-12-03 2021-07-23 勃林格殷格翰国际有限公司 Heteroaromatic compounds as VANIN inhibitors
JP2022509317A (en) * 2018-12-03 2022-01-20 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Heteroaromatic compounds as vanin inhibitors
CN114478485B (en) * 2018-12-03 2024-04-05 勃林格殷格翰国际有限公司 Heteroaromatic compounds as VANIN inhibitors
TWI811901B (en) * 2020-12-17 2023-08-11 大陸商上海美悅生物科技發展有限公司 A kind of pyrimidine formamide compound and application thereof

Also Published As

Publication number Publication date
KR20190026902A (en) 2019-03-13
CA3030381A1 (en) 2018-01-18
IL263662A (en) 2019-01-31
RU2019100559A (en) 2020-07-14
MA45656A (en) 2021-03-24
RU2019100559A3 (en) 2020-07-14
US10906888B2 (en) 2021-02-02
JP2019524716A (en) 2019-09-05
BR112019000589A2 (en) 2019-04-24
AU2017296338A1 (en) 2019-01-03
EP3484876A1 (en) 2019-05-22
CN109476645A (en) 2019-03-15
SG11201811161YA (en) 2019-01-30
US20190315715A1 (en) 2019-10-17
MX2019000536A (en) 2019-04-01

Similar Documents

Publication Publication Date Title
US10906888B2 (en) Pyrimidine carboxamides as inhibitors of Vanin-1 enzyme
US10316018B2 (en) Bicyclic-fused heteroaryl or aryl compounds as IRAK4 modulators
US11702424B2 (en) Bicyclic-fused heteroaryl or aryl compounds
JP2018527337A (en) Bicyclic fused heteroaryl or aryl compounds
WO2016193844A1 (en) Novel heterocyclic compounds as inhibitors of vanin-1 enzyme
US20160368919A1 (en) Novel pyridine pyrazinones as bet-family bromodomain inhibitors
RU2785126C2 (en) New compounds and their pharmaceutical compositions for treatment of inflammatory diseases

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: 17740116

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2017296338

Country of ref document: AU

Date of ref document: 20170707

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2019500621

Country of ref document: JP

Kind code of ref document: A

Ref document number: 3030381

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112019000589

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 20197004221

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2017740116

Country of ref document: EP

Effective date: 20190214

ENP Entry into the national phase

Ref document number: 112019000589

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20190111