WO2001027103A1 - Fab i inhibitors - Google Patents

Fab i inhibitors Download PDF

Info

Publication number
WO2001027103A1
WO2001027103A1 PCT/US2000/027844 US0027844W WO0127103A1 WO 2001027103 A1 WO2001027103 A1 WO 2001027103A1 US 0027844 W US0027844 W US 0027844W WO 0127103 A1 WO0127103 A1 WO 0127103A1
Authority
WO
WIPO (PCT)
Prior art keywords
methyl
indol
ylmethyl
mmole
aminopyridin
Prior art date
Application number
PCT/US2000/027844
Other languages
French (fr)
Inventor
William H. Miller
Kenneth A. Newlander
Mark A. Seefeld
Irene N. Uzinskas
Walter E. Dewolf, Jr.
Dalia R. Jakas
Original Assignee
Smithkline Beecham Corporation
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 JP2001530321A priority Critical patent/JP4803935B2/en
Application filed by Smithkline Beecham Corporation filed Critical Smithkline Beecham Corporation
Priority to AU78747/00A priority patent/AU773218B2/en
Priority to AT00968895T priority patent/ATE285821T1/en
Priority to CA2387016A priority patent/CA2387016C/en
Priority to NZ517706A priority patent/NZ517706A/en
Priority to HU0203122A priority patent/HU230030B1/en
Priority to BRPI0014470A priority patent/BRPI0014470B1/en
Priority to IL14882000A priority patent/IL148820A0/en
Priority to US10/089,755 priority patent/US6846819B1/en
Priority to EP00968895A priority patent/EP1226138B1/en
Priority to DE60017180T priority patent/DE60017180T2/en
Publication of WO2001027103A1 publication Critical patent/WO2001027103A1/en
Priority to IL148820A priority patent/IL148820A/en
Priority to NO20021638A priority patent/NO322708B1/en
Priority to HK03100341.3A priority patent/HK1049656A1/en
Priority to US11/007,927 priority patent/US7557125B2/en
Priority to US11/779,578 priority patent/US7524843B2/en
Priority to US12/403,078 priority patent/US7790716B2/en
Priority to US12/830,729 priority patent/US8173646B2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/72Nitrogen atoms
    • C07D213/75Amino or imino radicals, acylated by carboxylic or carbonic acids, or by sulfur or nitrogen analogues thereof, e.g. carbamates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/38Heterocyclic compounds having sulfur as a ring hetero atom
    • A61K31/381Heterocyclic compounds having sulfur as a ring hetero atom having five-membered rings
    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4436Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a heterocyclic ring having sulfur as a ring hetero atom
    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • 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
    • 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
    • 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
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/5365Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines ortho- or peri-condensed with heterocyclic ring systems
    • 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
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/02Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
    • C07D209/04Indoles; Hydrogenated indoles
    • C07D209/10Indoles; Hydrogenated indoles with substituted hydrocarbon radicals attached to carbon atoms of the hetero ring
    • C07D209/14Radicals substituted by nitrogen atoms, not forming part of a nitro radical
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/02Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
    • C07D209/04Indoles; Hydrogenated indoles
    • C07D209/10Indoles; Hydrogenated indoles with substituted hydrocarbon radicals attached to carbon atoms of the hetero ring
    • C07D209/18Radicals substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D209/24Radicals substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals with an alkyl or cycloalkyl radical attached to the ring nitrogen atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/02Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
    • C07D209/52Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring condensed with a ring other than six-membered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/32One oxygen, sulfur or nitrogen atom
    • C07D239/42One nitrogen atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D333/00Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom
    • C07D333/50Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom condensed with carbocyclic rings or ring systems
    • C07D333/52Benzo[b]thiophenes; Hydrogenated benzo[b]thiophenes
    • 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
    • C07D401/02Heterocyclic 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 two hetero rings
    • C07D401/12Heterocyclic 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 two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/12Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • 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
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/02Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D495/04Ortho-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
    • C07D498/02Heterocyclic 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
    • C07D498/04Ortho-condensed systems
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • This invention relates to pharmaceutically active compounds which inhibit Fab I and are useful for the treatment of bacterial infections.
  • FAS fatty acid synthase
  • ACP acyl carrier protein
  • each of the reactions is catalyzed by a distinct, mono-functional enzyme and the ACP is a discrete protein. Therefore, there is considerable potential for the selective inhibition of the bacterial system by antibacterial agents.
  • Fab I functions as an enoyl-ACP reductase (Bergler, et al, (1994), J.Biol.Chem. 269, 5493-5496) in the final step of the four reactions involved in each cycle of bacterial fatty acid biosynthesis.
  • the first step is catalyzed by ⁇ -ketoacyl-ACP synthase, which condenses malonyl-ACP with acetyl-CoA (FabH, synthase III).
  • FabH acetyl-CoA
  • malonyl-ACP is condensed with the growing-chain acyl-ACP (FabB and FabF, synthases I and II, respectively).
  • the second step in the elongation cycle is ketoester reduction by NADPH-dependent ⁇ -ketoacyl-ACP reductase (FabG).
  • FabG NADPH-dependent ⁇ -ketoacyl-ACP reductase
  • ⁇ -hydroxyacyl-ACP dehydrase either FabA or FabZ
  • trans-2-enoyl-ACP which in turn is converted to acyl-ACP by NADH-dependent enoyl-ACP reductase (Fab I).
  • Fab I is a major biosynthetic enzyme and is a key regulatory point in the overall synthetic pathwayof bacterial fatty acid biosynthesis. Therefore, Fab I is an ideal target for antibacterial intervention.
  • diazaborine antibiotics inhibit fatty acid, phospholipid and lipopolysaccharide (LPS) biosynthesis and that the antibacterial target of these compounds is Fab I.
  • derivative 2b 18 from Grassberger, et al, (1984) J. Med Chem 27, 947-953 has been reported to be a non-competitive inhibitor of Fab I (Bergler, et al, ( 1994 ) J.Biol.Chem. 269, 5493-5496).
  • plasmids containing the Fab I gene from diazaborine resistant S.
  • Fab I is also the target for the broad spectrum antibacterial agent triclosan (McMurry, et al, (1998) Nature 394, 531-532).
  • a crystal structure of the E. Coli Fab I complexed with NAD and triclosan shows that triclosan acts as a site-directed, very potent inhibitor of Fab I by mimicking its natural substrate (Levy, et al, (1999) Nature 398, 383-384). Ward, et al ((1999) Biochem.
  • Fab I inhibiting compounds have been found to be inhibitors of Streptococcus Fab K.
  • Fab I is not present in Streptococcus, and is not essential in Pseudomonas.
  • Fab K another enoyl reductase
  • Pseudomonas and Enterococcus contain both Fab I and Fab K
  • Streptococcus contains only Fab K. Consequently, pure Fab I inhibitors are not expected to have antibacterial activity in these organisms.
  • compounds that inhibit both Fab I and Fab K have the potential to be broad-spectrum antibacterial agents.
  • This invention comprises compounds of the formula (I), as described hereinafter, which inhibit Fab I and are useful in the treatment of bacterial infections.
  • This invention is also a pharmaceutical composition
  • a pharmaceutical composition comprising a compound according to formula (I) and a pharmaceutically acceptable carrier.
  • This invention is a method of treating bacterial infections by inhibiting Fab I and, for certain compounds, also inhibiting Fab K.
  • the compounds of this invention are useful as antibacterial agents.
  • This invention also comprises the preparation and purification of crotonoyl-ACP and the use of this purified enzyme in a Fab I enzyme inhibition assay.
  • This invention comprises compounds of formula (I):
  • R 1 isHorC ⁇ _4alkyl
  • R 2 is H, C1.4a.kyl or C3.6cycloa.kyl
  • R 4 is H or C 1 . 4 alkyl; indicates that one of the two designated bonds is a double bond and the other is a single bond; R ⁇ is CH2 when the bond to which it is attached is a double bond; or R ⁇ is H or
  • R 6 is H or C ⁇ _4alkyl
  • R 7 is H, Ci-6alkyl or -C ⁇ -6alkyl-Ar
  • W is S or O
  • E is O or NR'; each R' independently is H, Cj- ⁇ alkyl or -C ⁇ -6 a lkyl-Ar; and r is 0, 1 or 2; or a pharmaceutically acceptable salt thereof. Also included in this invention are pharmaceutically acceptable addition salts and complexes of the compounds of this invention. In cases wherein the compounds of this invention may have one or more chiral centers, unless specified, this invention includes each unique racemic compound, as well as each unique nonracemic compound.
  • O compounds may exist in tautomeric forms, such as keto-enol tautomers, such as --' ⁇ -- OR' and ⁇ sr , each tautomeric form is contemplated as being included within this invention, whether existing in equilibrium or locked in one form by appropriate substitution with R ⁇
  • keto-enol tautomers such as --' ⁇ -- OR' and ⁇ sr
  • each tautomeric form is contemplated as being included within this invention, whether existing in equilibrium or locked in one form by appropriate substitution with R ⁇
  • the meaning of any substituent at any one occurrence is independent of its meaning, or any other substituent's meaning, at any other occurrence.
  • prodrugs of the compounds of this invention are considered to be any covalently bonded carriers which release the active parent drug according to formula (I) in vivo.
  • the compounds of formula (I) inhibit Fab I. Inhibition of this enzyme is useful in the treatment of bacterial infections. Also, the compounds of this invention may be useful as antifungal agents. Additionally, the compounds may be useful in combination with known antibiotics.
  • this invention preferably includes compounds of formula (la):
  • this invention preferably includes compounds of formula (II):
  • R , R2, R3 and X are as defined for formula (I) compounds.
  • this invention preferably includes compounds of formula (Ila):
  • R , R2, R and X are as defined for formula (I) compounds.
  • this invention preferably includes compounds of formula (lib):
  • R 3 is as defined for formula (I) compounds.
  • R 3 is:
  • the compounds of this invention are Fab I inhibitors useful in the treatment of bacterial infections.
  • Two compounds of this invention namely (E)-N- methyl-N-(l-methyl-lH-indol-3-ylmethyl)-3-(7-oxo-5,6,7,8-tetrahydro-l,8-naphthyridin-3- yl)acrylamide and (E)-N-methyl-N-(2-methyl-lH-indol-3-ylmethyl)-3-(7-oxo-5,6,7,8- tetrahydro-l,8-naphthyridin-3-yl)acrylamide, are dual Fab I/Fab K inhibitors. These compounds have the potential to be broad spectrum antibiotics.
  • C ⁇ alkyl as applied herein means an optionally substituted alkyl group of 1 to 4 carbon atoms, and includes methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl and t-butyl.
  • Cj.galkyl additionally includes pentyl, n-pentyl, isopentyl, neopentyl and hexyl and the simple aliphatic isomers thereof.
  • C () -4alkyl and Cg-galkyl additionally indicates that no alkyl group need be present (e.g., that a covalent bond is present).
  • Any Cj ⁇ alkyl or Ci .g alkyl may be optionally substituted with the group R x , which may be on any carbon atom that results in a stable structure and is available by conventional synthetic techniques.
  • Suitable groups for R x are C1.4a.kyl, OR , SR , CN, N(R )2, CH 2 N(R') 2 , -N0 2 , -CF 3 , -C0 2 R' -CON(R ' )2, -COR ' , -NR C(0)R ' , F, Cl, Br, I, or -S(0) r CF3, wherein R' and r are as defined for formula (I) compounds.
  • Halogen or halo means F, Cl, Br, and I.
  • Ar, or aryl as applied herein, means phenyl or naphthyl, or phenyl or naphthyl substituted by one to three substituents, such as those defined above for alkyl, or substituted by methylenedioxy.
  • Het, or heterocycle indicates an optionally substituted five or six membered monocyclic ring, or a nine or ten-membered bicyclic ring containing one to three heteroatoms chosen from the group of nitrogen, oxygen and sulfur, which are stable and available by conventional chemical synthesis.
  • heterocycles are benzofuryl, benzimidazolyl, benzopyranyl, benzothienyl, furyl, imidazolyl, indolinyl, morpholinyl, piperidinyl, piperazinyl, pyrrolyl, pyrrolidinyl, tetrahydropyridinyl, pyridinyl, thiazolyl, thienyl, quinolinyl, isoquinolinyl, and tetra- and perhydro- quinolinyl and isoquinolinyl.
  • t-Bu refers to the tertiary butyl radical
  • Boc refers to the t-butyloxycarbonyl radical
  • Fmoc refers to the fluorenylmethoxycarbonyl radical
  • Ph refers to the phenyl radical
  • Cbz refers to the benzyloxycarbonyl radical
  • Bn refers to the benzyl radical
  • Me refers to methyl
  • Et refers to ethyl
  • Ac refers to acetyl
  • Alk refers to C1.4a.kyl
  • Nph refers to 1- or 2-naphthyl
  • cHex refers to cyclohexyl.
  • Tet refers to 5-tetrazolyl.
  • DCC refers to dicyclohexylcarbodiimide
  • DMAP refers to dimethylaminopyridine
  • EDC refers to l-(3-dimethylaminopropyl)-3- ethylcarbodiimide
  • hydrochloride HOBt refers to 1-hydroxybenzotriazole
  • THF refers to tetrahydrofuran
  • DIEA refers to diisopropylethylamine
  • DEAD refers to diethyl azodicarboxylate
  • PPh3 refers to triphenylphosphine
  • DIAD refers to diisopropyl azodicarboxylate
  • DME refers to dimethoxyethane
  • DMF refers to dimethylformamide
  • NBS refers to N-bromosuccinimide
  • Pd/C refers to a palladium on carbon catalyst
  • PPA refers to polyphosphoric acid
  • DPPA diphenylphosphoryl
  • R 2 , R3, R4 t R5 anc j x are as defined in formula (I), with any reactive functional groups protected, in the presence of EDC and HOBT;
  • R R3 and X are as defined in formula (I) and Halo is Br, Cl, F or I, with any reactive functional groups protected, in the presence of a palladium (II) salt, a phosphine ligand and base; and thereafter removing any protecting groups, and optionally forming a pharmaceutically acceptable salt.
  • the reaction is mediated by a palladium(O) species, and generally is conducted in an inert solvent, such as CH3CN, propionitrile, or toluene, in the presence of an appropriate acid scavenger, such as triethylamine (Et3N) or diisopropylethylamine ((i-Pr) 2 NEt).
  • an inert solvent such as CH3CN, propionitrile, or toluene
  • an appropriate acid scavenger such as triethylamine (Et3N) or diisopropylethylamine ((i-Pr) 2 NEt).
  • Typical sources of the palladium(O) species include palladium (II) acetate (Pd(OAc) 2 ) and palladium(H) chloride (PdCl 2 ), and oftentimes phosphine ligands, for instance triphenylphosphine (PPh3) or tri-ortho-tolylphosphine (P(tol)3), are included.
  • aqueous base for example, LiOH in aqueous THF or NaOH in aqueous methanol or ethanol
  • the intermediate carboxylate salt is acidified with a suitable acid, for instance TFA or HCl, to afford the carboxylic acid 1-3.
  • the carboxylic acid of 1-3 is converted to an activated form using, for example, EDC and HOBt, or SOCl 2 , and the activated form is subsequently reacted with an appropriate amine, for instance 1- methyl-2-(methylaminomethyl)indole, in a suitable solvent such as DMF, CH 2 C1 2 , or CH3CN, to afford 1-4.
  • an added base such as triethylamine (Et3N), diisopropylethylamine ((i-Pr) 2 NEt), or pyridine, may be used.
  • the amine is coupled via its free amino group to an appropriate carboxylic acid substrate using a suitable carbodiimide coupling agent, such as N,N' dicyclohexyl carbodiimide (DCC), optionally in the presence of catalysts such as 1- hydroxybenzotriazole (HOBt) and dimethylamino pyridine (DMAP).
  • a suitable carbodiimide coupling agent such as N,N' dicyclohexyl carbodiimide (DCC)
  • catalysts such as 1- hydroxybenzotriazole (HOBt) and dimethylamino pyridine (DMAP).
  • HABt 1- hydroxybenzotriazole
  • DMAP dimethylamino pyridine
  • Other methods such as the formation of activated esters, anhydrides or acid halides, of the free carboxyl of a suitably protected acid substrate, and subsequent reaction with the free amine, optionally in the presence of a base, are also suitable.
  • a benzoic acid is treated in an anhydrous solvent, such as methylene chloride or tetrahydrofuran (THF), in the presence of a base, such as N-methylmorpholine, DMAP or a trialkylamine, with isobutyl chloroformate to form the "activated anhydride", which is subsequently reacted with the free amine.
  • anhydrous solvent such as methylene chloride or tetrahydrofuran (THF)
  • a base such as N-methylmorpholine, DMAP or a trialkylamine
  • amine coupling partners used in the present invention were prepared by established methods well-known to those of skill in the art.
  • amine II-4 is prepared by the straightforward procedure outlined in Scheme II.
  • a suitable base generally sodium hydride (NaH)
  • the intermediate sodium salt is reacted with an appropriate alkylating agent, for instance methyl iodide, to afford II-2.
  • Polar solvents such as DMF , THF, or mixtures thereof are generally preferred for this reaction.
  • Compound II-2 can be conveniently converted to II-3 by reaction with an excess of an amine, such as methylamine, in a polar solvent, generally H 0 or a mixture of H 0 and methanol.
  • an amine such as methylamine
  • a polar solvent generally H 0 or a mixture of H 0 and methanol.
  • the ester of II-2 can be saponified under standard conditions, typically with an alkali metal hydroxide such as LiOH, NaOH, or KOH, in an aqueous solvent, such as THF, ethanol, or methanol, and the resulting carboxylic acid can be converted to the desired amide.
  • an alkali metal hydroxide such as LiOH, NaOH, or KOH
  • an aqueous solvent such as THF, ethanol, or methanol
  • Reduction of the amide II-3 to the amine II-4 is typically accomplished with lithium aluminum hydride (L.AIH4) in refluxing THF, although many other methods can be used to reduce amides to amines. Such methods are well-known to those of skill in the art, and can be found in standard reference volumes, such as "Compendium of Organic Synthetic Methods” (published by Wiley-Interscience).
  • the amine coupling partners used in the present invention can also be prepared by the reductive amination of an appropriate aldehyde (Scheme III).
  • This method involves the initial conversion of an aldehyde to an intermediate imine, which is subsequently reduced, oftentimes in situ, to afford the amine.
  • the commercially-available aldehyde III-l reacts with an appropriate amine, for instance methylamine, to afford an intermediate imine (not shown), which is reduced in situ to amine III-2 by reaction with a suitable reducing agent, usually sodium cyanoborohydride or sodium (triacetoxy)borohydride.
  • a suitable reducing agent usually sodium cyanoborohydride or sodium (triacetoxy)borohydride.
  • the reaction is conducted in the presence of an acid, such as acetic acid, in a polar solvent such as methanol or DMF.
  • the amine of compound IV-1 reacts with a variety of acylating agents to produce amides, sulfonamides, ureas, and carbamates.
  • IV-1 reacts with acetic anhydride (Ac 2 0) in a neutral solvent, typically THF, in the presence of a suitable base, such as sodium bicarbonate (NaHC03), to afford IV-2.
  • a neutral solvent typically THF
  • a suitable base such as sodium bicarbonate (NaHC03)
  • Other acylating agents including sulfonyl halides, isocyanates, and chlorocarbonates, also participate in this reaction to afford sulfonamides, ureas, and carbamates, respectively.
  • 1,8-Naphthyridine can be selectively reduced to l,2,3,4-tetrahydro-l,8- naphthyridine (V-2) by reaction with hydrogen gas in the presence of a suitable catalyst, preferably palladium metal on activated carbon (Pd/C), in an inert solvent, generally MeOH, EtOH, EtOAc, or mixtures thereof.
  • a suitable catalyst preferably palladium metal on activated carbon (Pd/C)
  • an inert solvent generally MeOH, EtOH, EtOAc, or mixtures thereof.
  • V-2 is converted to a suitably protected derivative, for instance the N-Boc protected derivative V-3, by reaction with di-fert-butyl dicarbonate in the presence of an appropriate base, preferably lithium hexamethyldisilazide (LiHMDS).
  • LiHMDS lithium hexamethyldisilazide
  • V-3 is selectively brominated at the 6-position by reaction with a suitable brominating agent, such as bromine (Br 2 ) or N-bromosuccinimide (NBS).
  • a suitable brominating agent such as bromine (Br 2 ) or N-bromosuccinimide (NBS).
  • Typical solvents for a bromination reaction include CH 2 C1 2 , CCI4, MeOH, AcOH, or mixtures thereof.
  • the resulting 6-bromo- 1 ,2,3,4-tetrahydro- 1 ,8-naphthyridine V-4 participates in a Heck reaction as described in Scheme I to afford V-5. Removal of the Boc protecting group is accomplished under standard acidic conditions well-known to those of skill in the art (see Greene above), and the benzyl ester is saponified as described in Scheme I to afford V-6.
  • VI- 1 2-aminonicotinic acid
  • VI-2 is reduced to alcohol VI-2 under standard conditions (LiAlH4, THF), and the aromatic ring of VI-2 is brominated using, for example, bromine or N-bromosuccinimide (NBS), in a neutral solvent such as CH 2 C1 2 , to afford VI-3.
  • NBS N-bromosuccinimide
  • VI-3 is converted to bromide VI-4, which reacts with a diester of malonic acid, for instance dimethyl malonate, in the presence of a suitable base, typically sodium methoxide, in an alcoholic solvent such as methanol, to afford the naphthyridone derivative VI-S.
  • a suitable base typically sodium methoxide
  • alcoholic solvent such as methanol
  • Benzylic bromide VII-1 reacts with an amine, for example aqueous methylamine, to afford benzylic amine VII-2.
  • Polar solvents such as THF, DMF, DMSO, or mixture thereof, are generally preferred for this reaction.
  • VII-2 reacts with a dialkyl carbonate, preferably dimethyl carbonate, in the presence of a suitable base, typically sodium methoxide, in an alcoholic solvent, generally methanol, to afford the cyclic urea derivative VII-3.
  • a suitable base typically sodium methoxide
  • an alcoholic solvent generally methanol
  • IX-1 2,2 -dipyridylamine
  • a suitable brominating agent such as bromine (Br 2 ) or N- bromosuccinimide (NBS).
  • Typical solvents for a bromination reaction include CH 2 C1 2 , CCI4, MeOH, AcOH, or mixtures thereof.
  • the resulting mono-bromo derivative IX-2 reacts with N-methyl-N-(l-methyl-lH-indol-2-ylmethyl)acrylamide in a ⁇ eck-type reaction as described in Scheme I to afford IX-3.
  • X-l 2H-pyrido[3,2-b]-l,4-oxazin-3(4H)-one
  • X-l is selectively brominated at the 5-position by reaction with a suitable brominating agent, such as bromine (Br 2 ) or N-bromosuccinimide (NBS).
  • Typical solvents for a bromination reaction include C ⁇ 2 C1 2 , CCI4, MeOH, AcOH, or mixtures thereof.
  • the resulting mono-bromo derivative X-2 reacts with N-methyl-N-(l -methyl- lH-indol-2-ylmethyl)acrylamide in a ⁇ eck-type reaction as described in Scheme I to afford X-3.
  • Acid addition salts of the compounds are prepared in a standard manner in a suitable solvent from the parent compound and an excess of an acid, such as hydrochloric, hydrobromic, hydrofluoric, sulfuric, phosphoric, acetic, trifluoroacetic, maleic, succinic or methanesulfonic. Certain of the compounds form inner salts or zwitterions which may be acceptable.
  • Cationic salts are prepared by treating the parent compound with an excess of an alkaline reagent, such as a hydroxide, carbonate or alkoxide, containing the appropriate cation; or with an appropriate organic amine.
  • Cations such as Li + , Na + , K + , Ca ++ , Mg ++ and NH4 + are specific examples of cations present in pharmaceutically acceptable salts.
  • This invention also provides a pharmaceutical composition which comprises a compound according to formula (I) and a pharmaceutically acceptable carrier. Accordingly, the compounds of formula (I) may be used in the manufacture of a medicament.
  • Pharmaceutical compositions of the compounds of formula (I) prepared as hereinbefore described may be formulated as solutions or lyophilized powders for parenteral administration. Powders may be reconstituted by addition of a suitable diluent or other pharmaceutically acceptable carrier prior to use.
  • the liquid formulation may be a buffered, isotonic, aqueous solution.
  • Suitable diluents are normal isotonic saline solution, standard 5% dextrose in water or buffered sodium or ammonium acetate solution.
  • Such formulation is especially suitable for parenteral administration, but may also be used for oral administration or contained in a metered dose inhaler or nebulizer for insufflation. It may be desirable to add excipients such as polyvinylpyrrolidone, gelatin, hydroxy cellulose, acacia, polyethylene glycol, mannitol, sodium chloride or sodium citrate.
  • these compounds may be encapsulated, tableted or prepared in a emulsion or syrup for oral administration.
  • Pharmaceutically acceptable solid or liquid carriers may be added to enhance or stabilize the composition, or to facilitate preparation of the composition.
  • Solid carriers include starch, lactose, calcium sulfate dihydrate, terra alba, magnesium stearate or stearic acid, talc, pectin, acacia, agar or gelatin.
  • Liquid carriers include syrup, peanut oil, olive oil, saline and water.
  • the carrier may also include a sustained release material such as glyceryl monostearate or glyceryl distearate, alone or with a wax.
  • the amount of solid carrier varies but, preferably, will be between about 20 mg to about 1 g per dosage unit.
  • the pharmaceutical preparations are made following the conventional techniques of pharmacy involving milling, mixing, granulating, and compressing, when necessary, for tablet forms; or milling, mixing and filling for hard gelatin capsule forms.
  • a liquid carrier When a liquid carrier is used, the preparation will be in the form of a syrup, elixir, emulsion or an aqueous or non-aqueous suspension.
  • Such a liquid formulation may be administered directly p.o. or filled into a soft gelatin capsule.
  • the compounds of this invention may also be combined with excipients, such as cocoa butter, glycerin, gelatin or polyethylene glycols, and molded into a suppository.
  • excipients such as cocoa butter, glycerin, gelatin or polyethylene glycols
  • the compounds of this invention may be combined with diluents to take the form of ointments, gels, pastes, creams, powders or sprays.
  • the compositions which are ointments, gels, pastes or creams contain diluents, for example, animal and vegetable fats, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures of these substances.
  • the compositions which are powders or sprays contain diluents, for example, lactose, talc, silicic acid, aluminum hydroxide, calcium silicate and polyamide powder, or mixtures of these substances.
  • the typical carriers are water, mixtures of water and water miscible solvents, such as lower alkanols or vegetable oils, and water-soluble non-toxic polymers, for example cellulose derivatives, such as methyl cellulose.
  • the compounds described herein are inhibitors of Fab I, and are useful for treating bacterial infections.
  • these compounds are useful for the treatment of bacterial infections, such as, for example, infections of upper respiratory tract (e.g. otitis media, bacterial tracheitis, acute epiglottitis, thyroiditis), lower respiratory (e.g. empyema, lung abscess), cardiac (e.g. infective endocarditis), gastrointestinal (e.g. secretory diarrhoea, splenic abscess, retroperitoneal abscess), CNS (e.g. cerebral abscess), eye (e.g.
  • the compounds of this invention may be useful as antifungal agents. Additionally, the compounds may be useful in combination with known antibiotics.
  • the compounds of this invention are administered to the patient, in a manner such that the concentration of drug is sufficient to treat bacterial infections.
  • the pharmaceutical composition containing the compound is administered at an oral dose of between about 10 mg to about 1000 mg, taken once or several times daily, in a manner consistent with the condition of the patient.
  • the oral dose would be about 50 mg to about 500 mg, although the dose may be varied depending upon the age, body weight and symptoms of the patient.
  • parenteral administration is preferred.
  • An intravenous infusion of the compound of formula (I) in 5% dextrose in water or normal saline, or a similar formulation with suitable excipients, is most effective, although an intramuscular bolus injection is also useful.
  • the precise level and method by which the compounds are administered is readily determined by one skilled in the art.
  • the compounds may be tested in one of several biological assays to determine the concentration of compound which is required to have a given pharmacological effect.
  • the fabl gene was cloned from the chromosomal DNA of 5. aureus strain WCUH29 using the polymerase chain reaction. Amplification was performed using Taq DNA polymerase (BRL) and the following primers: 5 - CGCCTCGAGATGTTAAATCTTGAAAACAAAACATATGTC-3' and 5 - CGCGGATCCAATCAAGTCAGGTTGAAATATCCA-3' (Xhol and BamWl sites underlined). The resulting fragment was then digested with Xhol and BamHl and ligated into Xhol- and B ⁇ HI-digested expression vector pET-16b (Novagen), producing pET- His ⁇ Q -fabI.
  • BBL Taq DNA polymerase
  • the gene sequence of fabl was confirmed by automated cycle sequencing using an Applied Biosystems model 377 machine.
  • the untagged version of pET-fabl was constructed by digesting pET-His ⁇ )-/ ⁇ -?/ with Ncol and Ndel to remove a 97 bp fragment encoding the His 10 tag, the factor Xa cleavage site and the first 8 amino acids of Fabl, and replacing it with a linker encoding the first 8 amino acids of Fabl plus a glycine residue between the initiator methionine and the lysine at position 2.
  • This plasmid was called pET- fabl.
  • the linker was made by annealing the following two oligonucleotides: 5 - CATGGGCTTAAATCTTGAAAACAAAACA-3' and 5 - TATGTTTTGTTTTCAAGATTTAAGCC-3'.
  • the linker sequence in pET-fabl was confirmed by dideoxy sequencing. Only native Fabl was used for compound evaluation.
  • plasmid pET-fabl was transformed into BL21(DE3) (Novagen) cells, to form strain BL21(DE3):pET-/--£>/.
  • aureus Fabl was expressed as soluble protein to 10% of total cell protein, 400g cells being recovered from 15L fermentation in tryptone phosphate medium. The cells were lysed and the sample centrifuged. The resulting supernatant was filtered and purified using three consecutive chromatography columns: ion-exchange (Sourse 15Q), dye-affinity (Blue sepharose), and size exclusion chromatography columns (Superose 12). After each column the Fabl containing fractions were pooled, concentrated, and checked for purity and biological activity.
  • a PCR fragment of correct size for E. coli Fabl was PCR amplified from E. coli chromosomal DNA, subcloned into the TOPO TA cloning vector, and verified by colony PCR + restriction endonuclease analysis.
  • the presumptive E. coli Fabl PCR fragment was subcloned into the expression vector pBluePet.
  • the Fabl clone was transformed into E. coli strain BL21(DE3).
  • Small Scale expression studies show an over-expressed protein band of correct molecular weight (-28 Kda) for E. coli Fabl clearly visible following Coomassie staining of SDS PAGE gels. DNA sequencing of the E. coli Fabl expression constructs illustrated that no errors were apparent. N' terminal amino acid sequencing has confirmed the over-expressed protein band to be E. coli Fabl.
  • E. coli Fabl was expressed as soluble protein to 15% of total cell protein, 120g cells being recovered from 3L fermentation in shake flasks in modified terrific broth. The cells were lysed and the sample centrifuged. The resulting supernatant was filtered and purified using three consecutive chromatography columns: ion-exchange (Sourse 15Q), dye-affinity (blue sepharose), and size exclusion (superose 12). After each column the Fabl containing fractions were pooled, concentrated and checked for purity and biological activity.
  • NADH S aureus Fabl Enzyme Inhibition Assay
  • Reactions contained 5 mg/mL E. coli apo-ACP, 0.8 mM crotonoyl-CoA (Fluka), 10 mM MgCl 2 , and 30 uM S. pneumoniae ACP synthase in 50 mM NaHEPES, pH 7.5.
  • the mixture was gently mixed on a magnetic stirrer at 23 °C for 2 hr, and the reaction was terminated by the addition of 15 mM EDTA .
  • the reaction mixture was filtered through a 0.2 micron filter (Millipore) and applied to a MonoQ column (Pharmacia) equilibrated with 20 mM Tris-Cl, pH 7.5. The column was washed with buffer until all non-adherent material was removed (as observed by UV detection), and the crotonoyl-ACP was eluted with a linear gradient of 0 to 400 mM NaCl.
  • IC50's are estimated from a fit of the initial velocities to a standard, 4-parameter model (Equation 1) and are typically reported as the mean ⁇ S.D. of duplicate determinations.
  • Compounds of this invention in this assay have IC5o's from about 100.0 micromolar to about 0.04 micromolar.
  • the apparent Ki is calculated from Equation 2 assuming the inhibition is competitve with crotonoyl-ACP.
  • Ki(app) IC50/( 1+[S]/Ks) FabK Enzyme Inhibition Assay
  • FabK catalyses the reduction of enoyl-ACPs with the concomitant oxidation of NADH.
  • the reduction of crotonoyl-ACP to butyryl- ACP can be monitored by following the change in absorbance at 340 nm as NADH is oxidized.
  • Assays were carried out in Costar 3696 half-area plates in a final assay volume of 150 uL on a Spectramax platereader.
  • the substrates (NADH and crotonoyl-ACP) were incubated with FabK enzyme in 100 mM N-[2-acetamido]-2 iminodiacetic acid (ADA), pH 6.5, 100 mM NH4CI, 4% glycerol at 30 °C and the reaction was monitored at 340 nm.
  • IC50s were determined for compounds by carrying out the assay at 8 different concentrations of compound (100 uM-0.75 uM) in duplicate. The IC50 was calculated using Grafit software (v 4.09). The two Fab K inhibitors of this invention have IC50's of about 5 micromolar.
  • Test organisms were selected from the following laboratory strains: Staphylococcus aureus Oxford, Staphylococcus aureus WCUH29, Streptococcus pneumoniae ERY2, Streptococcus pneumoniae 1629, Streptococcus pneumoniae N 1387, Enterococcus faecalis I, Enterococcus faecalis 7, Haemophilus influenzae Ql, Haemophilus influenzae NEMC1, Moraxella Catarrhalis 1502, Escherichia coli 7623 AcrABEFD+, Escherichia coli 120 AcrAB-, Escherichia coli MG1655, Escherichia coli MG1658.
  • the minimum inhibitory concentration (MIC) was determined as the lowest concentration of compound that inhibited visible growth. A mirror reader was used to assist in determining the MIC endpoint.
  • the compounds used in the antimicrobial assays of the present invention have a MIC value of less than 128 ⁇ g/mL. Most preferably, said compounds have a MIC value of less than 64 ⁇ g/mL.
  • the preferred Fab I and Fab K enzyme inhibition assays use crotonoyl-ACP, rather than crotonoyl CoA, as a substrate.
  • this invention comprises the preparation and purification of crotonoyl-ACP and the use of this purified enzyme in Fab I and Fab K enzyme inhibition assays.
  • Crotonoyl-ACP was synthesised using 5. pneumoniae ACP synthase to catalyse the addition of a crotonoyl group from crotonoyl CoA to E.coli apo-acyl carrier protein (ACP).
  • an apo-acyl carrier protein from any bacterial species such as from Escherichia coli, Staphylococcus and Streptococcus, can be used in the preparation of crotonoyl-ACP.
  • This synthesis was carried out in the presence of magnesium chloride in NaHEPES, pH 7.5. The reaction was complete in 2 hours at a reaction temperature of about 20-30°C, preferably at 23°C.
  • the purified crotonoyl-ACP prepared above is then used in the Fab I and Fab K assays to determine the inhibitors of the instant invention.
  • Assays may be carried out, for example, in Costar 3696 half-area plates, preferably at a final assay volume of 150 ul on a Spectramax platereader.
  • Preferred substrates used in the methods of the invention are NADH, NADPH, an NADH analogue and crotonoyl-ACP.
  • Further provided comprising the step of incubating substrates with Fab I or Fab K in 100 mM N-[2- acetamido]-2 iminodiacetic acid (ADA), pH 6.5. This reaction may be monitored at 340 nm, among other wavelengths.
  • Mass spectra were obtained using electrospray (ES) ionization techniques. Elemental analyses were performed by Quantitative Technologies Inc., Whitehouse, NJ. Melting points were obtained on a Thomas-Hoover melting point apparatus and are uncon n ected. All temperatures are reported in degrees Celsius.
  • Analtech Silica Gel GF and E. Merck Silica Gel 60 F-254 thin layer plates were used for thin layer chromatography. Flash chromatography was carried out on E. Merck Kieselgel 60 (230-400 mesh) silica gel.
  • ODS refers to an octadecylsilyl derivatized silica gel chromatographic support.
  • YMC ODS-AQ® is an ODS chromatographic support and is a registered trademark of YMC Co. Ltd., Kyoto, Japan.
  • PRP-1® is a polymeric (styrene-divinylbenzene) chromatographic support, and is a registered trademark of Hamilton Co., Reno, Nevada.
  • Celite® is a filter aid composed of acid-washed diatomaceous silica, and is a registered trademark of Manville Corp., Denver, Colorado.
  • 1,8-Naphthyridine (1.0 g, 7.68 mmole) was hydrogenated (50 psi) with 10% Pd/C (100 mg) in absolute ethanol (40 mL) for 18 hr. The mixture was filtered through a pad of Celite® and the filtrate was concentrated to give the title compound (1.04 g) which was sufficiently pure for use in the next step: MS (ES) m/e 135 (M + H) + .
  • EDC (1.53 g, 0.01 mole) was added to a solution of 3-methyl-2-inden-2-carboxylic acid (1.91 g, 0.01 mole), methylamine hydrochloride (0.675 g, 0.01 mole), HOBt • H 2 0 (1.53 g, 0.01 mole) and triethylamine (4.0 mL, 0.028 mole) in anhydrous DMF (80 mL) at RT. The reaction was stirred overnight, then was concentrated in vacuo.
  • the reaction was purged with argon and heated at reflux for 6 hr, then was cooled to RT and concentrated to dryness under vacuum.
  • the residue was taken up in 80% ethyl acetate/hexanes (100 mL), and the solution was filtered through a pad of silica gel, eluting with 80% ethyl acetate/hexanes (400 mL) until all the product was eluted off.
  • the yellowish filtrate was concentrated under vacuum, and the residue was taken up in a small volume of 1 : 1 Et 2 0/petroleum ether.
  • N-(Benzyloxycarbonyloxy)succinimide (8.9 g, 35.7 mmole) was added to a solution of 3-(methylaminomethyl)-l /- indole (5.2 g, 32.5 mmole) and triethylamine (5.0 mL, 65.7 mmole) in DMF (100 mL) at RT. The reaction was stirred overnight then was concentrated in vacuo. The residue was diluted with water and the mixture was extracted with ethyl acetate. The combined extracts were dried over Na 2 S04 and concentrated. Flash chromatography on silica gel (33% ethyl acetate/hexanes) gave the title compound (7.0 g, 74%) as an off-white solid: MS (ES) m/e 295 (M + ⁇ ) + .
  • EDC (0.70 g, 3.7 mmole) was added to a solution of (E)-3-(6-aminopyridin-3- yl)acrylic acid (0.61 g, 3.7 mmole), l-methyl-2-(methylaminomethyl)-lH-indole (0.65 g, 3.7 mmole), ⁇ OBt • ⁇ 2 0 (0.50 g, 3.7 mmole), and triethylamine (0.52 mL, 3.7 mmole) in DMF (30 mL) at RT. The reaction was stirred overnight, then was concentrated in vacuo. The residue was diluted with 5% NaHC03 and extracted with CH 2 C1 2 .
  • Example 19 According to the procedure of Example 1, except substituting (E)-3-(6-amino-5- methylpyridin-3-yl)acrylic acid (0.40 g, 2.24 mmole) for (E)-3-(6-aminopyridin-3- yPacrylic acid, and substituting 2-(methylaminomethyl)naphthalene (0.42 g, 2.47 mmole) for l-methyl-2-(methylaminomethyl)indole, the title compound (0.65 g, 87%) was prepared as an off-white solid: MS (ES) m/e 332 (M + H)+.
  • Example 19 Example 19
  • Example 2 According to the procedure of Example 1, except substituting 1 -methy 1-2- (propylaminomethyl)-lH-indole (0.2 g,l mmole) for l-methyl-2-(methylaminomethyl)- lH- indole, the title compound (0.14 g, 40%) was prepared as a white solid: MS (ES) m/e 349 (M + ⁇ )+.
  • EDC (0.30 g, 1.58 mmole) was added to a solution of 3-(6-aminopyridin-3- y Pacrylic acid (0.26 g, 1.58 mmole), 2-(methy laminomethyl)- lH-indole (0.23 g, 1.43 mmole), ⁇ OBt • ⁇ 2 0 (0.21 g, 1.58 mmole) and diisopropylethylamine (0.51 mL, 2.86 mmole) in DMF (20 mL) at RT. The reaction was stirred overnight, then was concentrated in vacuo. The residue was diluted with water and extracted with ethyl acetate.
  • EDC (0.35 g, 1.89 mmole) was added to a solution of 3-(6-aminopyridin-3- yl)acrylic acid (0.31 g, 1.89 mmole), l-methyl-3-(methylaminomethyl)-lH-indole (0.30 g, 1.72 mmole), ⁇ OBt • ⁇ 2 0 (0.24 g, 1.89 mmole) and diisopropylethylamine (0.60 mL, 3.44 mmole) in DMF (20 mL) at RT. The reaction was stirred overnight, then was concentrated in vacuo. The residue was diluted with water and extracted with ethyl acetate.
  • Crotonic anhydride (0.29 mL, 1.96 mmole) was added to a solution of (E)-3-(6- aminopyridin-3-yl)-N-methyl-N-(l -methyl- lH-indol-2-ylmethyl)acry lamide (0.16 g, 0.49 mmole) and sodium bicarbonate (0.20 g, 2.45 mmole) in T ⁇ F (30 mL) at RT, and the reaction was heated at reflux under nitrogen. After 48 hr, the reaction was concentrated in vacuo and the residue was diluted with water and extracted with ethyl acetate. The combined extracts were dried over Na 2 S04 and concentrated in vacuo to afford the title compound (0.10 g, 53%) as a tan solid: MS (ES) m/e 389 (M + H) + .
  • Example 31 According to the procedure of Example 31, except substituting 1 -methy 1-3- (methylaminomethyl)indole (1.96 g, 8.6 mmole) for 3-methyl-2- (methylaminomethyPindene hydrochloride, and substituting 2-amino-5-bromopyrimidine (1.0 g, 5.75 mmole) for 6-bromo-3,4-dihydro-lH-l,8-naphthyridin-2-one, the title compound (1.44 g, 78%) was prepared as a yellow solid: MS (ES) m/e 322 (M + ⁇ ) + .
  • Example 68 According to the procedure of Example 31, except substituting 1 -methy 1-2- (methylaminomethyl)indole (1.17 g, 6.75 mmole) for 3-methyl-2- (methylaminomethyl)indene hydrochloride, and substituting 5-bromo-2H-pyrido[3,2- ⁇ >]- l,4-oxazin-3(4 )-one (1.03 g, 4.50 mmole) for 6-bromo-3,4-dihydro-l /-l,8-naphthyridin- 2-one, the title compound (0.90 g, 53%) was prepared as a light yellow solid: MS (ES) m/e 377 (M + ⁇ ) + .
  • Example 68 Example 68
  • Example 31 According to the procedure of Example 31, except substituting l-methyl-3- (methylaminomethyl)indole (0.38 g, 2.20 mmole) for 3-methyl-2- (methylaminomethyl)indene hydrochloride, and substituting 5-bromo- 2H-pyrido[3,2-Z>]- l,4-oxazin-3(4//)-one (0.32 g, 1.40 mmole) for 6-bromo-3,4-dihydro-lH-l,8-naphthyridin- 2-one, the title compound (0.26 g, 50%) was prepared as a light yellow solid: MS (ES) m/e 311 (M + ⁇ )+.
  • EDC (0.42 g, 2.20 mmole) was added to a solution of 3-(6-aminopyridin-3- yl)acrylic acid (0.36 g, 2.20 mmole), l-benzyl-3-(methy laminomethy l)-lH-indole (0.50 g, 2.00 mmole), ⁇ OBt • ⁇ 2 0 (0.30 g, 2.20 mmole) and diisopropylethylamine (0.70 mL, 4.00 mmole) in DMF (30 mL) at RT. The reaction was stirred overnight, then was concentrated in vacuo. The residue was diluted with water and extracted with ethyl acetate.
  • N-Methyl-N-(1 -methyl- lH-indol-2-ylmethyl)acry lamide (from Example 76 (a)) was taken up in propionitrile (50 mL). To this solution was added with stirring 2- phenylamino-5-bromopyridine (1.3 g, 5.2 mmole), DIEA (1.8 mL, 10 mmole), Pd(OAc) (1 12 mg, 0.5 mmole) and P(o-tol)3 (304 mg, 1.0 mmole). The reaction was purged with argon then stirred at reflux for 16 h. After cooling to room temperature the reaction was concentrated to dryness under vacuum.
  • Example 81 According to the procedure of Example 76 (b), except substituting 6-bromo-3,4- dihydro-lH-l,8-naphthyridin-2-one (1.3 g, 5.7 mmole) for the 2-phenylamino-5- bromopyridine, the title compound (0.57 g, 26%) was prepared as a white solid: MS (ES) m/e 389.19 (M + ⁇ )+.
  • Example 81 Example 81
  • Example 76 According to the procedure of Example 76 (a), except substituting 2- (methylarninomethyl)-3-methylbenzo[b]thiophene (1.53 g, 8 mmole) for the l-methyl-2- (methylaminomethyl)-lH-indole, the title compound was prepared and used without further purification.
  • Example 76 According to the procedure of Example 76 (b), except substituting 6-bromo-3,4- dihydro-lH-l,8-naphthyridin-2-one (1.3 g, 5.7 mmole) for the 2-phenylamino-5- bromopyridine, the title compound (0.85 g, 33%) was prepared as an off-white solid: MS (ES) m/e 392.2 (M + ⁇ ) + .
  • Example 2 According to the procedure of Example 1, except substituting 1 -methy 1-3- (methylaminomethyl)-l ⁇ -pyrrolo[2,3-o]pyridine (0.2 g, 1.14 mmole) for the l-methyl-2- (methylaminomethyP-lH-indole, the title compound (0.19 g, 52%) was prepared as a white solid: MS (ES) m/e 322 (M + ⁇ ) + .
  • Example 87 According to the procedure of Example 1 , except substituting 2,3-dihydro-8- (methylaminomethyl)-lH-3a-azacyclopenta[ ⁇ ]indene (0.100 g, 0.5 mmole) for the 1- methy l-2-(methylaminomethyl)- 1 //-indole, the title compoud (0.063 g, 36%) was prepared as a white solid: MS (ES) m/e 347 (M + ⁇ ) + .
  • Example 87 Example 87
  • a preparation which contains 20 mg of the compound of Example 1 as a sterile dry powder is prepared as follows: 20 mg of the compound is dissolved in 15 mL of distilled water. The solution is filtered under sterile conditions into a 25 mL multi-dose ampoule and lyophilized. The powder is reconstituted by addition of 20 mL of 5% dextrose in water (D5W) for intravenous or intramuscular injection. The dosage is thereby determined by the injection volume. Subsequent dilution may be made by addition of a metered volume of this dosage unit to another volume of D5W for injection, or a metered dose may be added to another mechanism for dispensing the drug, as in a bottle or bag for IV drip infusion or other injection-infusion system.
  • D5W dextrose in water
  • a capsule for oral administration is prepared by mixing and milling 50 mg of the compound of Example 1 with 75 mg of lactose and 5 mg of magnesium stearate. The resulting powder is screened and filled into a hard gelatin capsule.
  • a tablet for oral administration is prepared by mixing and granulating 20 mg of sucrose, 150 mg of calcium sulfate dihydrate and 50 mg of the compound of Example 1 with a 10% gelatin solution.
  • the wet granules are screened, dried, mixed with 10 mg starch, 5 mg talc and 3 mg stearic acid; and compressed into a tablet.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Heterocyclic Carbon Compounds Containing A Hetero Ring Having Oxygen Or Sulfur (AREA)
  • Indole Compounds (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
  • Pyridine Compounds (AREA)
  • Nitrogen And Oxygen Or Sulfur-Condensed Heterocyclic Ring Systems (AREA)

Abstract

Compounds of formula (I) are disclosed which are Fab I inhibitors and are useful in the treatment of bacterial infections. In said formula, (A) is (a), (b), (c), (d), (e), (f), (g), (h), (i), (j), (k), (l), (m), (n), (o) or (p); (I) wherein R1 is H or C¿1-4?alkyl; R?2¿ is H, C¿1-4?alkyl or C3-6cycloalkyl; R?3¿ is (q), (r), (s), (t), (u), (v) or (w); R4 is H or C¿1-4?alkyl; (x) indicates that one of the two designated bonds is a double bond and the other is a single bond; R?5¿ is CH¿2? when the bond to which it is attached is a double bond; or R?5¿ is H or C¿1-4?alkyl when the bond to which it is attached is a single bond; R?6¿ is H or C¿1-4?alkyl; R?7¿ is H, C¿1-6?alkyl or -C0-6alkyl-Ar; Y is H, C1-4alkyl, N(R')2, NHC(O)R', NHCH2C(O)R' or NHC(O)CH=CHR'; each X independently is H, C1-4alkyl, CH2OH, OR', SR', CN, N(R')2, CH2N(R')2, NO2, CF3, CO2R', CON(R')2, COR', NR'C(O)R', F, Cl, Br, I or -S(O)rCF3; W is S or O, Q is H or C1-4alkyl; M is CH2 or O; L is CH2 or C(O); E is O or NR'; each R' independently is H, C1-6alkyl or -C0-6alkyl-Ar; and r is 0, 1 or 2; or a pharmaceutically acceptable salt thereof.

Description

j TITLE
Fab I Inhibitors
FIELD OF THE INVENTION This invention relates to pharmaceutically active compounds which inhibit Fab I and are useful for the treatment of bacterial infections.
BACKGROUND OF THE INVENTION
While the overall pathway of saturated fatty acid biosynthesis is similar in all organisms, the fatty acid synthase (FAS) systems vary considerably with respect to their structural organization. Vertebrates and yeast possess a FAS in which all the enzymatic activities are encoded on one or two polypeptide chains, respectively, and the acyl carrier protein (ACP) is an integral part of the complex. In contrast, in bacterial FAS, each of the reactions is catalyzed by a distinct, mono-functional enzyme and the ACP is a discrete protein. Therefore, there is considerable potential for the selective inhibition of the bacterial system by antibacterial agents.
Fab I (previously designated EnvM) functions as an enoyl-ACP reductase (Bergler, et al, (1994), J.Biol.Chem. 269, 5493-5496) in the final step of the four reactions involved in each cycle of bacterial fatty acid biosynthesis. In this pathway, the first step is catalyzed by β-ketoacyl-ACP synthase, which condenses malonyl-ACP with acetyl-CoA (FabH, synthase III). In subsequent rounds, malonyl-ACP is condensed with the growing-chain acyl-ACP (FabB and FabF, synthases I and II, respectively). The second step in the elongation cycle is ketoester reduction by NADPH-dependent β-ketoacyl-ACP reductase (FabG). Subsequent dehydration by β-hydroxyacyl-ACP dehydrase (either FabA or FabZ) leads to trans-2-enoyl-ACP, which in turn is converted to acyl-ACP by NADH-dependent enoyl-ACP reductase (Fab I). Further rounds of this cycle, adding two carbon atoms per cycle, eventually lead to palmitoyl-ACP (16C), where upon the cycle is stopped largely due to feedback inhibition of Fab I by palmitoyl-ACP (Heath, et al, (1996), J.Biol.Chem. 271, 1833-1836). Thus, Fab I is a major biosynthetic enzyme and is a key regulatory point in the overall synthetic pathwayof bacterial fatty acid biosynthesis. Therefore, Fab I is an ideal target for antibacterial intervention.
Studies have shown that diazaborine antibiotics inhibit fatty acid, phospholipid and lipopolysaccharide (LPS) biosynthesis and that the antibacterial target of these compounds is Fab I. For example, derivative 2b 18 from Grassberger, et al, (1984) J. Med Chem 27, 947-953 has been reported to be a non-competitive inhibitor of Fab I (Bergler, et al, ( 1994 ) J.Biol.Chem. 269, 5493-5496). Also, plasmids containing the Fab I gene from diazaborine resistant S. typhimurium conferred diazaborine resistance in E.coli (Turnowsky, et al, (1989) J.Bacteriol., 171, 6555-6565). Furthermore, inhibition of Fab I either by diazaborine or by raising the temperature in a Fab I temperature sensitive mutant is lethal. These results demonstrate that Fab I is essential to the survival of the organism (Bergler, et al, (1994) J.Biol.Chem. 269, 5493-5496).
Recent studies have shown that Fab I is also the target for the broad spectrum antibacterial agent triclosan (McMurry, et al, (1998) Nature 394, 531-532). A crystal structure of the E. Coli Fab I complexed with NAD and triclosan shows that triclosan acts as a site-directed, very potent inhibitor of Fab I by mimicking its natural substrate (Levy, et al, (1999) Nature 398, 383-384). Ward, et al ((1999) Biochem. 38, 12514-12525) have shown that there is no evidence for the formation of a covalent complex between Fab I and triclosan, which would be analogous to the diazaborines; triclosan differs from these compounds in that it is a reversible inhibitor of Fab I. The structural data for the complex of Fab I with NAD and triclosan provides important information about Fab I as a therapeutic target.
Importantly, it has now been discovered that certain compounds are Fab I inhibitors and have antibacterial activity, and, therefore, may be useful for the treatment of bacterial infections in mammals, particularly in man.
Additionally, two of the instant Fab I inhibiting compounds have been found to be inhibitors of Streptococcus Fab K. Fab I is not present in Streptococcus, and is not essential in Pseudomonas. There is also reason to believe that Fab I may not be essential in Enterococcus. In all of these organisms, another enoyl reductase, termed Fab K, is present (Heath, R. J.; Rock, CO., Nature (2000), 406, 145-146). Pseudomonas and Enterococcus contain both Fab I and Fab K, and Streptococcus contains only Fab K. Consequently, pure Fab I inhibitors are not expected to have antibacterial activity in these organisms. Thus, compounds that inhibit both Fab I and Fab K have the potential to be broad-spectrum antibacterial agents.
SUMMARY OF THE INVENTION This invention comprises compounds of the formula (I), as described hereinafter, which inhibit Fab I and are useful in the treatment of bacterial infections.
This invention is also a pharmaceutical composition comprising a compound according to formula (I) and a pharmaceutically acceptable carrier.
This invention is a method of treating bacterial infections by inhibiting Fab I and, for certain compounds, also inhibiting Fab K. In a particular aspect, the compounds of this invention are useful as antibacterial agents. This invention also comprises the preparation and purification of crotonoyl-ACP and the use of this purified enzyme in a Fab I enzyme inhibition assay.
DETAILED DESCRIPTION
This invention comprises compounds of formula (I):
Figure imgf000005_0001
Figure imgf000006_0001
R1 isHorCι_4alkyl;
R2 is H, C1.4a.kyl or C3.6cycloa.kyl;
R is
Figure imgf000006_0002
Figure imgf000006_0003
Figure imgf000007_0001
R4 is H or C1.4alkyl;
Figure imgf000007_0002
indicates that one of the two designated bonds is a double bond and the other is a single bond; R^ is CH2 when the bond to which it is attached is a double bond; or R^ is H or
Cj.4a.kyl when the bond to which it is attached is a single bond;
R6 is H or Cι_4alkyl;
R7 is H, Ci-6alkyl or -Cθ-6alkyl-Ar;
Y is H, Cι_4alkyl, N(R^2. NHC(O)R\ NHCH2C(0)R' or NHC(0)CH=CHR'; each X independently is H, C 1.4alkyl, CH2OH, OR', SR', CN, N(R')2, CH2N(R')2,
N02, CF3, C02R', CON(R')2, COR', NR C(0)R', F, Cl, Br, I or -S(0)rCF3;
W is S or O;
Figure imgf000007_0003
M is CH or O; L is CH2 or C(O);
E is O or NR'; each R' independently is H, Cj-όalkyl or -Cθ-6alkyl-Ar; and r is 0, 1 or 2; or a pharmaceutically acceptable salt thereof. Also included in this invention are pharmaceutically acceptable addition salts and complexes of the compounds of this invention. In cases wherein the compounds of this invention may have one or more chiral centers, unless specified, this invention includes each unique racemic compound, as well as each unique nonracemic compound.
In cases in which compounds have unsaturated carbon-carbon double bonds, both the cis (Z) and trans (E) isomers are within the scope of this invention. In cases wherein
O compounds may exist in tautomeric forms, such as keto-enol tautomers, such as --'^-- OR' and ^^^sr , each tautomeric form is contemplated as being included within this invention, whether existing in equilibrium or locked in one form by appropriate substitution with R\ The meaning of any substituent at any one occurrence is independent of its meaning, or any other substituent's meaning, at any other occurrence.
Also included in this invention are prodrugs of the compounds of this invention. Prodrugs are considered to be any covalently bonded carriers which release the active parent drug according to formula (I) in vivo.
The compounds of formula (I) inhibit Fab I. Inhibition of this enzyme is useful in the treatment of bacterial infections. Also, the compounds of this invention may be useful as antifungal agents. Additionally, the compounds may be useful in combination with known antibiotics.
With respect to formula (I), this invention preferably includes compounds of formula (la):
Figure imgf000008_0001
in which R2, R3, R4, 5 an(j x Αre as defined for formula (I) compounds.
With respect to formula (I), this invention preferably includes compounds of formula (II):
Figure imgf000008_0002
in which R , R2, R3 and X are as defined for formula (I) compounds.
With respect to formula (II), this invention preferably includes compounds of formula (Ila):
Figure imgf000008_0003
in which R , R2, R and X are as defined for formula (I) compounds.
In particular, with respect to formula (II), this invention preferably includes compounds of formula (lib):
Figure imgf000009_0001
(lib) in which R3 is as defined for formula (I) compounds. Suitably, with respect to formula (I), R3 is:
Figure imgf000009_0002
Figure imgf000009_0003
in which X, Y, M, L and E are as defined for formula (I) compounds. Representative of the novel compounds of this invention are the compounds of examples 1-86 hereinafter. The compounds of this invention are Fab I inhibitors useful in the treatment of bacterial infections. Two compounds of this invention, namely (E)-N- methyl-N-(l-methyl-lH-indol-3-ylmethyl)-3-(7-oxo-5,6,7,8-tetrahydro-l,8-naphthyridin-3- yl)acrylamide and (E)-N-methyl-N-(2-methyl-lH-indol-3-ylmethyl)-3-(7-oxo-5,6,7,8- tetrahydro-l,8-naphthyridin-3-yl)acrylamide, are dual Fab I/Fab K inhibitors. These compounds have the potential to be broad spectrum antibiotics.
Abbreviations and symbols commonly used in the peptide and chemical arts are used herein to describe the compounds of this invention. In general, the amino acid abbreviations follow the IUPAC-IUB Joint Commission on Biochemical Nomenclature as described in Eur. J. Biochem., 158, 9 (1984).
C^alkyl as applied herein means an optionally substituted alkyl group of 1 to 4 carbon atoms, and includes methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl and t-butyl. Cj.galkyl additionally includes pentyl, n-pentyl, isopentyl, neopentyl and hexyl and the simple aliphatic isomers thereof. C()-4alkyl and Cg-galkyl additionally indicates that no alkyl group need be present (e.g., that a covalent bond is present). Any Cj^alkyl or Ci .g alkyl may be optionally substituted with the group Rx, which may be on any carbon atom that results in a stable structure and is available by conventional synthetic techniques. Suitable groups for Rx are C1.4a.kyl, OR , SR , CN, N(R )2, CH2N(R')2, -N02, -CF3, -C02R' -CON(R')2, -COR', -NR C(0)R', F, Cl, Br, I, or -S(0)rCF3, wherein R' and r are as defined for formula (I) compounds. Halogen or halo means F, Cl, Br, and I.
Ar, or aryl, as applied herein, means phenyl or naphthyl, or phenyl or naphthyl substituted by one to three substituents, such as those defined above for alkyl, or substituted by methylenedioxy. Het, or heterocycle, indicates an optionally substituted five or six membered monocyclic ring, or a nine or ten-membered bicyclic ring containing one to three heteroatoms chosen from the group of nitrogen, oxygen and sulfur, which are stable and available by conventional chemical synthesis. Illustrative heterocycles are benzofuryl, benzimidazolyl, benzopyranyl, benzothienyl, furyl, imidazolyl, indolinyl, morpholinyl, piperidinyl, piperazinyl, pyrrolyl, pyrrolidinyl, tetrahydropyridinyl, pyridinyl, thiazolyl, thienyl, quinolinyl, isoquinolinyl, and tetra- and perhydro- quinolinyl and isoquinolinyl. Any accessible combination of up to three substituents on the Het ring, such as those defined above for alkyl, that are available by chemical synthesis and are stable are within the scope of this invention. Certain radical groups are abbreviated herein. t-Bu refers to the tertiary butyl radical, Boc refers to the t-butyloxycarbonyl radical, Fmoc refers to the fluorenylmethoxycarbonyl radical, Ph refers to the phenyl radical, Cbz refers to the benzyloxycarbonyl radical, Bn refers to the benzyl radical, Me refers to methyl, Et refers to ethyl, Ac refers to acetyl, Alk refers to C1.4a.kyl, Nph refers to 1- or 2-naphthyl and cHex refers to cyclohexyl. Tet refers to 5-tetrazolyl.
Certain reagents are abbreviated herein. DCC refers to dicyclohexylcarbodiimide, DMAP refers to dimethylaminopyridine, EDC refers to l-(3-dimethylaminopropyl)-3- ethylcarbodiimide, hydrochloride, HOBt refers to 1-hydroxybenzotriazole, THF refers to tetrahydrofuran, DIEA refers to diisopropylethylamine, DEAD refers to diethyl azodicarboxylate, PPh3 refers to triphenylphosphine, DIAD refers to diisopropyl azodicarboxylate, DME refers to dimethoxyethane, DMF refers to dimethylformamide, NBS refers to N-bromosuccinimide, Pd/C refers to a palladium on carbon catalyst, PPA refers to polyphosphoric acid, DPPA refers to diphenylphosphoryl azide, BOP refers to benzotriazol-l-yloxy-tris(dimethyl-amino)phosphonium hexafluorophosphate, HF refers to hydrofluoric acid, TEA refers to triethylamine, TFA refers to trifluoroacetic acid, PCC refers to pyridinium chlorochromate. Generally, compounds of this invention are prepared by:
(i) reacting a compound of formula (III) with a compound of formula (IV):
Figure imgf000011_0001
wherein R2, R3, R4t R5 ancj x are as defined in formula (I), with any reactive functional groups protected, in the presence of EDC and HOBT;
(ii) reacting a compound of formula (V) with a compound of formula (VI):
Figure imgf000011_0002
wherein R R3 and X are as defined in formula (I) and Halo is Br, Cl, F or I, with any reactive functional groups protected, in the presence of a palladium (II) salt, a phosphine ligand and base; and thereafter removing any protecting groups, and optionally forming a pharmaceutically acceptable salt.
In particular, compounds of the formula (I) are prepared by the general methods described in the Schemes hereinafter.
Scheme I
Figure imgf000011_0003
(a) benzyl acrylate, Pd(OAc)2, P(o-tol)3, (i-Pr)2NEt, propionitrile; (b) 1.0 N NaOH, MeOH; (c) l-methyl-2-(methylaminomethyl)indole, EDC, HOBt • H20, Et3N, DMF. A suitable haloaromatic derivative, for instance for instance 2-amino-5- bromopyridine (1-1), reacts with an appropriate α,β-unsaturated ester, for example benzyl acrylate, in a Heck-type reaction (see Heck, Org. Reactions 1982, 27, 345) to afford 1-2. The reaction is mediated by a palladium(O) species, and generally is conducted in an inert solvent, such as CH3CN, propionitrile, or toluene, in the presence of an appropriate acid scavenger, such as triethylamine (Et3N) or diisopropylethylamine ((i-Pr)2NEt). Typical sources of the palladium(O) species include palladium (II) acetate (Pd(OAc)2) and palladium(H) chloride (PdCl2), and oftentimes phosphine ligands, for instance triphenylphosphine (PPh3) or tri-ortho-tolylphosphine (P(tol)3), are included. The ethyl ester of 1-2 is hydrolyzed using aqueous base, for example, LiOH in aqueous THF or NaOH in aqueous methanol or ethanol, and the intermediate carboxylate salt is acidified with a suitable acid, for instance TFA or HCl, to afford the carboxylic acid 1-3. The carboxylic acid of 1-3 is converted to an activated form using, for example, EDC and HOBt, or SOCl2, and the activated form is subsequently reacted with an appropriate amine, for instance 1- methyl-2-(methylaminomethyl)indole, in a suitable solvent such as DMF, CH2C12, or CH3CN, to afford 1-4. Depending on whether acid neutralization is required, an added base, such as triethylamine (Et3N), diisopropylethylamine ((i-Pr)2NEt), or pyridine, may be used. Many additional methods for converting a carboxylic acid to an amide are known, and can be found in standard reference books, such as "Compendium of Organic Synthetic Methods", Vol. I - VI (published by Wiley-Interscience), or Bodansky, "The Practice of Peptide Synthesis" (published by Springer- Verlag), which are incorporated herein by reference. Amide coupling reagents as used herein denote reagents which may be used to form peptide bonds. Typical coupling methods employ carbodiimides, activated anhydrides and esters and acyl halides. Reagents such as EDC, DCC, DPPA, PPA, BOP reagent, HOBt, N- hydroxysuccinimide and oxalyl chloride are typical.
Typically, the amine is coupled via its free amino group to an appropriate carboxylic acid substrate using a suitable carbodiimide coupling agent, such as N,N' dicyclohexyl carbodiimide (DCC), optionally in the presence of catalysts such as 1- hydroxybenzotriazole (HOBt) and dimethylamino pyridine (DMAP). Other methods, such as the formation of activated esters, anhydrides or acid halides, of the free carboxyl of a suitably protected acid substrate, and subsequent reaction with the free amine, optionally in the presence of a base, are also suitable. For example, a benzoic acid is treated in an anhydrous solvent, such as methylene chloride or tetrahydrofuran (THF), in the presence of a base, such as N-methylmorpholine, DMAP or a trialkylamine, with isobutyl chloroformate to form the "activated anhydride", which is subsequently reacted with the free amine.
Scheme II
Figure imgf000013_0001
(a) NaH, Mel, DMF; (b) CH3NH2, H20, MeOH; (c) LiAlH4, THF.
The amine coupling partners used in the present invention were prepared by established methods well-known to those of skill in the art. For example, amine II-4 is prepared by the straightforward procedure outlined in Scheme II. Commercially available ethyl indole-2- carboxylate (II-l) is deprotonated with a suitable base, generally sodium hydride (NaH), . and the intermediate sodium salt is reacted with an appropriate alkylating agent, for instance methyl iodide, to afford II-2. Polar solvents such as DMF , THF, or mixtures thereof are generally preferred for this reaction. Compound II-2 can be conveniently converted to II-3 by reaction with an excess of an amine, such as methylamine, in a polar solvent, generally H 0 or a mixture of H 0 and methanol. Alternatively, the ester of II-2 can be saponified under standard conditions, typically with an alkali metal hydroxide such as LiOH, NaOH, or KOH, in an aqueous solvent, such as THF, ethanol, or methanol, and the resulting carboxylic acid can be converted to the desired amide. Typical methods for forming amides are described in Scheme I. Reduction of the amide II-3 to the amine II-4 is typically accomplished with lithium aluminum hydride (L.AIH4) in refluxing THF, although many other methods can be used to reduce amides to amines. Such methods are well-known to those of skill in the art, and can be found in standard reference volumes, such as "Compendium of Organic Synthetic Methods" (published by Wiley-Interscience). Scheme III
Figure imgf000014_0001
(a) CH3NH2, NaCNBH3, MeOH.
The amine coupling partners used in the present invention can also be prepared by the reductive amination of an appropriate aldehyde (Scheme III). This method, which is well- known to those of skill in the art, involves the initial conversion of an aldehyde to an intermediate imine, which is subsequently reduced, oftentimes in situ, to afford the amine. For example, the commercially-available aldehyde III-l reacts with an appropriate amine, for instance methylamine, to afford an intermediate imine (not shown), which is reduced in situ to amine III-2 by reaction with a suitable reducing agent, usually sodium cyanoborohydride or sodium (triacetoxy)borohydride. Frequently, the reaction is conducted in the presence of an acid, such as acetic acid, in a polar solvent such as methanol or DMF.
Scheme IV
Figure imgf000014_0002
(a) Ac20, NaHC03, THF.
The amine of compound IV-1 (prepared as described in Scheme I) reacts with a variety of acylating agents to produce amides, sulfonamides, ureas, and carbamates. For example, IV-1 reacts with acetic anhydride (Ac20) in a neutral solvent, typically THF, in the presence of a suitable base, such as sodium bicarbonate (NaHC03), to afford IV-2. Other acylating agents, including sulfonyl halides, isocyanates, and chlorocarbonates, also participate in this reaction to afford sulfonamides, ureas, and carbamates, respectively. Scheme V
Figure imgf000015_0001
(a) H2, Pd/C, EtOH; (b) (Boc)20, LiHMDS, THF; (c) NBS, AcOH, CH2C12; (d) benzyl acrylate, Pd(OAc)2, P(ø-tol)3, (i-Pr)2NEt, propionitrile; (e) 4 N HCl/dioxane; (f) LiOH, H20, MeOH.
1,8-Naphthyridine (V-l) can be selectively reduced to l,2,3,4-tetrahydro-l,8- naphthyridine (V-2) by reaction with hydrogen gas in the presence of a suitable catalyst, preferably palladium metal on activated carbon (Pd/C), in an inert solvent, generally MeOH, EtOH, EtOAc, or mixtures thereof. V-2 is converted to a suitably protected derivative, for instance the N-Boc protected derivative V-3, by reaction with di-fert-butyl dicarbonate in the presence of an appropriate base, preferably lithium hexamethyldisilazide (LiHMDS). The protecting group for the amine must be compatible with subsequent chemistry, and must be readily removable when desired. Methods for the protection of amines are well-known to those of skill in the art, and are described in standard reference volumes, such as Greene "Protective Groups in Organic Synthesis" (published by Wiley- Interscience). V-3 is selectively brominated at the 6-position by reaction with a suitable brominating agent, such as bromine (Br2) or N-bromosuccinimide (NBS). Typical solvents for a bromination reaction include CH2C12, CCI4, MeOH, AcOH, or mixtures thereof. The resulting 6-bromo- 1 ,2,3,4-tetrahydro- 1 ,8-naphthyridine V-4 participates in a Heck reaction as described in Scheme I to afford V-5. Removal of the Boc protecting group is accomplished under standard acidic conditions well-known to those of skill in the art (see Greene above), and the benzyl ester is saponified as described in Scheme I to afford V-6. Scheme VI
Figure imgf000016_0001
Figure imgf000016_0002
(a) LiAlH4, THF; (b) NBS, CH2C12; (c) 48% HBr; (d) (Me02C)2CH2, NaOMe, MeOH; (e) NaOH, H20, MeOH, (f) HCl, H20, MeOH; (g) acryloyl chloride, Et3N, CH2C12; (h) Pd(OAc)2, P(o-tol)3, (i-Pr)2NEt, propionitrile.
Commercially available 2-aminonicotinic acid (VI- 1) is reduced to alcohol VI-2 under standard conditions (LiAlH4, THF), and the aromatic ring of VI-2 is brominated using, for example, bromine or N-bromosuccinimide (NBS), in a neutral solvent such as CH2C12, to afford VI-3. On reaction with 48% aqueous HBr, VI-3 is converted to bromide VI-4, which reacts with a diester of malonic acid, for instance dimethyl malonate, in the presence of a suitable base, typically sodium methoxide, in an alcoholic solvent such as methanol, to afford the naphthyridone derivative VI-S. Saponification and neutralization under standard conditions affords an intermediate carboxylic acid (not shown), which is typically not isolated, but is subject to decarboxylation on gentle warming to afford the naphthyridone VI-6. This compound reacts with acrylamide VI-8 in a Heck-type reaction as described in Scheme I to afford VI-9. Alternatively, VI-6 might be converted to VI-9 according to the general procedure described in Scheme I for the conversion of 1-1 to 1-4. The acrylamide VI-8 is conveniently prepared by reaction of amine VI-7 (see Scheme II) with an activated form of acrylic acid in an amide bond-forming reaction. Typical conditions for the formation of amides are described in Scheme I, and are well-known to those of skill in the art.
Scheme VII
Figure imgf000017_0001
(a) CH3NH2, H20, THF; (b) (MeO)2C=0, NaOMe, MeOH; (c) compound VI-8, Pd(OAc)2, P(o-tol)3, (i-Pr)2NEt, propionitrile.
Benzylic bromide VII-1, prepared as described in Scheme VI, reacts with an amine, for example aqueous methylamine, to afford benzylic amine VII-2. Polar solvents such as THF, DMF, DMSO, or mixture thereof, are generally preferred for this reaction. VII-2 reacts with a dialkyl carbonate, preferably dimethyl carbonate, in the presence of a suitable base, typically sodium methoxide, in an alcoholic solvent, generally methanol, to afford the cyclic urea derivative VII-3. This compound is converted to VII-4 by reaction with compound VI-8 as described in Scheme VI.
Scheme VIII
Figure imgf000018_0001
(a) SnCl2 • H20, EtOH; (b) 96% HC02H; (c) TrCl, Et3N, CH2C12; (d) benzyl acrylate, Pd(OAc)2, P(o-tol)3, (i-Pr)2NEt, propionitrile; (e) 4 N HCl/dioxane; (f) NaOH, H20, MeOH.
The nitro group of commercially available 2-amino-5-bromo-3-nitropyridine (VIII- 1) is reduced under standard conditions using, for example, tin (II) chloride in EtOH. The resulting diamine, VIII-2, reacts with formic acid, or an appropriate equivalent, to afford the imidazopyridine derivative VIII-3. This compound is converted to a suitably protected derivative, for instance the N-trityl protected derivative VIII-4, by reaction with trityl chloride in the presence of an appropriate base, typically triethylamine or diisopropylethylamine. Typical solvents for this reaction include CH2C12, DMF, or mixtures thereof. As discussed in Scheme V, the protecting group for the amine must be compatible with the subsequent chemistry, and must be readily removable when desired. VIII-4 is converted to VIII-6 according to the general procedure described in Scheme V.
Scheme IX
Figure imgf000019_0001
Figure imgf000019_0002
(a) Br2, AcOH; (b) N-methyl-N-(l -methyl- lH-indol-2-ylmethyl)acrylamide, Pd(OAc)2, P(o-tol)3, (i-Pr)2NEt, propionitrile.
Commercially-available 2,2 -dipyridylamine (IX-1) is mono-brominated at the 5-position by reaction with a suitable brominating agent, such as bromine (Br2) or N- bromosuccinimide (NBS). Typical solvents for a bromination reaction include CH2C12, CCI4, MeOH, AcOH, or mixtures thereof. The resulting mono-bromo derivative IX-2 reacts with N-methyl-N-(l-methyl-lH-indol-2-ylmethyl)acrylamide in a Ηeck-type reaction as described in Scheme I to afford IX-3.
Scheme X
Figure imgf000019_0003
(a) Br2, AcOΗ; (b) N-methyl-N-(l-methyl-lH-indol-2-ylmethyl)acrylamide, Pd(OAc)2, P(o-tol)3, (i-Pr)2NEt, propionitrile.
Commercially-available 2H-pyrido[3,2-b]-l,4-oxazin-3(4H)-one (X-l) is selectively brominated at the 5-position by reaction with a suitable brominating agent, such as bromine (Br2) or N-bromosuccinimide (NBS). Typical solvents for a bromination reaction include CΗ2C12, CCI4, MeOH, AcOH, or mixtures thereof. The resulting mono-bromo derivative X-2 reacts with N-methyl-N-(l -methyl- lH-indol-2-ylmethyl)acrylamide in a Ηeck-type reaction as described in Scheme I to afford X-3.
Acid addition salts of the compounds are prepared in a standard manner in a suitable solvent from the parent compound and an excess of an acid, such as hydrochloric, hydrobromic, hydrofluoric, sulfuric, phosphoric, acetic, trifluoroacetic, maleic, succinic or methanesulfonic. Certain of the compounds form inner salts or zwitterions which may be acceptable. Cationic salts are prepared by treating the parent compound with an excess of an alkaline reagent, such as a hydroxide, carbonate or alkoxide, containing the appropriate cation; or with an appropriate organic amine. Cations such as Li+, Na+, K+, Ca++, Mg++ and NH4+ are specific examples of cations present in pharmaceutically acceptable salts. This invention also provides a pharmaceutical composition which comprises a compound according to formula (I) and a pharmaceutically acceptable carrier. Accordingly, the compounds of formula (I) may be used in the manufacture of a medicament. Pharmaceutical compositions of the compounds of formula (I) prepared as hereinbefore described may be formulated as solutions or lyophilized powders for parenteral administration. Powders may be reconstituted by addition of a suitable diluent or other pharmaceutically acceptable carrier prior to use. The liquid formulation may be a buffered, isotonic, aqueous solution. Examples of suitable diluents are normal isotonic saline solution, standard 5% dextrose in water or buffered sodium or ammonium acetate solution. Such formulation is especially suitable for parenteral administration, but may also be used for oral administration or contained in a metered dose inhaler or nebulizer for insufflation. It may be desirable to add excipients such as polyvinylpyrrolidone, gelatin, hydroxy cellulose, acacia, polyethylene glycol, mannitol, sodium chloride or sodium citrate.
Alternately, these compounds may be encapsulated, tableted or prepared in a emulsion or syrup for oral administration. Pharmaceutically acceptable solid or liquid carriers may be added to enhance or stabilize the composition, or to facilitate preparation of the composition. Solid carriers include starch, lactose, calcium sulfate dihydrate, terra alba, magnesium stearate or stearic acid, talc, pectin, acacia, agar or gelatin. Liquid carriers include syrup, peanut oil, olive oil, saline and water. The carrier may also include a sustained release material such as glyceryl monostearate or glyceryl distearate, alone or with a wax. The amount of solid carrier varies but, preferably, will be between about 20 mg to about 1 g per dosage unit. The pharmaceutical preparations are made following the conventional techniques of pharmacy involving milling, mixing, granulating, and compressing, when necessary, for tablet forms; or milling, mixing and filling for hard gelatin capsule forms. When a liquid carrier is used, the preparation will be in the form of a syrup, elixir, emulsion or an aqueous or non-aqueous suspension. Such a liquid formulation may be administered directly p.o. or filled into a soft gelatin capsule.
For rectal administration, the compounds of this invention may also be combined with excipients, such as cocoa butter, glycerin, gelatin or polyethylene glycols, and molded into a suppository.
For topical administration, the compounds of this invention may be combined with diluents to take the form of ointments, gels, pastes, creams, powders or sprays. The compositions which are ointments, gels, pastes or creams contain diluents, for example, animal and vegetable fats, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures of these substances. The compositions which are powders or sprays contain diluents, for example, lactose, talc, silicic acid, aluminum hydroxide, calcium silicate and polyamide powder, or mixtures of these substances. Additionally, for topical ophthalmologic administration, the typical carriers are water, mixtures of water and water miscible solvents, such as lower alkanols or vegetable oils, and water-soluble non-toxic polymers, for example cellulose derivatives, such as methyl cellulose.
The compounds described herein are inhibitors of Fab I, and are useful for treating bacterial infections. For instance, these compounds are useful for the treatment of bacterial infections, such as, for example, infections of upper respiratory tract (e.g. otitis media, bacterial tracheitis, acute epiglottitis, thyroiditis), lower respiratory (e.g. empyema, lung abscess), cardiac (e.g. infective endocarditis), gastrointestinal (e.g. secretory diarrhoea, splenic abscess, retroperitoneal abscess), CNS (e.g. cerebral abscess), eye (e.g. blepharitis, conjunctivitis, keratitis, endophthal itis, preseptal and orbital cellulitis, darcryocystitis), kidney and urinary tract (e.g. epididymitis, intrarenal and perinephric abscess, toxic shock syndrome), skin (e.g. impetigo, folliculitis, cutaneous abscesses, cellulitis, wound infection, bacterial myositis), and bone and joint (e.g. septic arthritis, osteomyelitis). Also, the compounds of this invention may be useful as antifungal agents. Additionally, the compounds may be useful in combination with known antibiotics.
The compounds of this invention are administered to the patient, in a manner such that the concentration of drug is sufficient to treat bacterial infections. The pharmaceutical composition containing the compound is administered at an oral dose of between about 10 mg to about 1000 mg, taken once or several times daily, in a manner consistent with the condition of the patient. Preferably, the oral dose would be about 50 mg to about 500 mg, although the dose may be varied depending upon the age, body weight and symptoms of the patient. For acute therapy, parenteral administration is preferred. An intravenous infusion of the compound of formula (I) in 5% dextrose in water or normal saline, or a similar formulation with suitable excipients, is most effective, although an intramuscular bolus injection is also useful. The precise level and method by which the compounds are administered is readily determined by one skilled in the art.
The compounds may be tested in one of several biological assays to determine the concentration of compound which is required to have a given pharmacological effect.
Cloning of S. aureus Fabl:
The fabl gene was cloned from the chromosomal DNA of 5. aureus strain WCUH29 using the polymerase chain reaction. Amplification was performed using Taq DNA polymerase (BRL) and the following primers: 5 - CGCCTCGAGATGTTAAATCTTGAAAACAAAACATATGTC-3' and 5 - CGCGGATCCAATCAAGTCAGGTTGAAATATCCA-3' (Xhol and BamWl sites underlined). The resulting fragment was then digested with Xhol and BamHl and ligated into Xhol- and Bα HI-digested expression vector pET-16b (Novagen), producing pET- His\Q-fabI. The gene sequence of fabl was confirmed by automated cycle sequencing using an Applied Biosystems model 377 machine. The untagged version of pET-fabl was constructed by digesting pET-Hisκ)-/α-?/ with Ncol and Ndel to remove a 97 bp fragment encoding the His 10 tag, the factor Xa cleavage site and the first 8 amino acids of Fabl, and replacing it with a linker encoding the first 8 amino acids of Fabl plus a glycine residue between the initiator methionine and the lysine at position 2. This plasmid was called pET- fabl. The linker was made by annealing the following two oligonucleotides: 5 - CATGGGCTTAAATCTTGAAAACAAAACA-3' and 5 - TATGTTTTGTTTTCAAGATTTAAGCC-3'. The linker sequence in pET-fabl was confirmed by dideoxy sequencing. Only native Fabl was used for compound evaluation. For overproduction of native Fabl, plasmid pET-fabl was transformed into BL21(DE3) (Novagen) cells, to form strain BL21(DE3):pET-/--£>/.
Purification of 5. aureus Fabl
5. aureus Fabl was expressed as soluble protein to 10% of total cell protein, 400g cells being recovered from 15L fermentation in tryptone phosphate medium. The cells were lysed and the sample centrifuged. The resulting supernatant was filtered and purified using three consecutive chromatography columns: ion-exchange (Sourse 15Q), dye-affinity (Blue sepharose), and size exclusion chromatography columns (Superose 12). After each column the Fabl containing fractions were pooled, concentrated, and checked for purity and biological activity.
Cloning of E. coli Fabl:
A PCR fragment of correct size for E. coli Fabl was PCR amplified from E. coli chromosomal DNA, subcloned into the TOPO TA cloning vector, and verified by colony PCR + restriction endonuclease analysis. The presumptive E. coli Fabl PCR fragment was subcloned into the expression vector pBluePet. The Fabl clone was transformed into E. coli strain BL21(DE3). Small Scale expression studies show an over-expressed protein band of correct molecular weight (-28 Kda) for E. coli Fabl clearly visible following Coomassie staining of SDS PAGE gels. DNA sequencing of the E. coli Fabl expression constructs illustrated that no errors were apparent. N' terminal amino acid sequencing has confirmed the over-expressed protein band to be E. coli Fabl.
Purification of E. coli Fabl
E. coli Fabl was expressed as soluble protein to 15% of total cell protein, 120g cells being recovered from 3L fermentation in shake flasks in modified terrific broth. The cells were lysed and the sample centrifuged. The resulting supernatant was filtered and purified using three consecutive chromatography columns: ion-exchange (Sourse 15Q), dye-affinity (blue sepharose), and size exclusion (superose 12). After each column the Fabl containing fractions were pooled, concentrated and checked for purity and biological activity.
S aureus Fabl Enzyme Inhibition Assay (NADH):
Assays were carried out in half-area, 96-well microtitre plates. Compounds were evaluated in 50-uL assay mixtures containing 100 mM NaADA, pH 6.5 (ADA = N-[2- acetamido]-2-iminodiacetic acid), 4 % glycerol, 0.25 mM crotonoyl CoA, 1 mM NADH, and an appropriate dilution of S. aureus Fabl. Inhibitors were typically varied over the range of 0.01-10 uM. The consumption of NADH was monitored for 20 minutes at 30 °C by following the change in absorbance at 340 nm. Initial velocities were estimated from an exponential fit of the non-linear progress curves represented by the slope of the tangent at t = 0 min. K-50's were estimated from a fit of the initial velocities to a standard, 4-parameter model and are typically reported as the mean ± S.D. of duplicate determinations. Triclosan, a commercial antibacterial agent and inhibitor of Fabl, is currently included in all assays as a positive control. Compounds of this invention have ^Q'S from about 5.0 micromolar to about 0.05 micromolar.
S aureus Fabl Enzyme Inhibition Assay (NADPH):
Assays were carried out in half-area, 96-well microtitre plates. Compounds were evaluated in 150-uL assay mixtures containing 100 mM NaADA, pH 6.5 (ADA = N-[2- acetamido]-2-iminodiacetic acid), 4 % glycerol, 0.25 mM crotonoyl CoA, 50 uM NADPH, and an appropriate dilution of S. aureus Fabl. Inhibitors were typically varied over the range of 0.01-10 uM. The consumption of NADPH was monitored for 20 minutes at 30 °C by following the change in absorbance at 340 nm. Initial velocities were estimated from an exponential fit of the non-linear progress curves represented by the slope of the tangent at t = 0 min. ICSQ'S were estimated from a fit of the initial velocities to a standard, 4-parameter model and are typically reported as the mean ± S.D. of duplicate determinations. Triclosan, a commercial antibacterial agent and inhibitor of Fabl, is currently included in all assays as a positive control.
E. coli Fabl Enzyme Inhibition Assay:
Assays were carried out in half-area, 96-well microtitre plates. Compounds were evaluated in 150-uL assay mixtures containing 100 mM NaADA, pH 6.5 (ADA = N-[2- acetamido]-2-iminodiacetic acid), 4 % glycerol, 0.25 mM crotonoyl CoA, 50 uM NADH, and an appropriate dilution of E. coli Fabl. Inhibitors were typically varied over the range of 0.01-10 uM. The consumption of NADH was monitored for 20 minutes at 30 °C by following the change in absorbance at 340 nm. Initial velocities were estimated from an exponential fit of the non-linear progress curves represented by the slope of the tangent at t = 0 min. IC5o's were estimated from a fit of the initial velocities to a standard, 4-parameter model and are typically reported as the mean ± S.D. of duplicate determinations. Triclosan, a commercial antibacterial agent and inhibitor of Fabl, is currently included in all assays as a positive control. Compounds of this invention have IC50 S from about 100.0 micromolar to about 0.05 micromolar.
Preparation and purification of crotonoyl-ACP:
Reactions contained 5 mg/mL E. coli apo-ACP, 0.8 mM crotonoyl-CoA (Fluka), 10 mM MgCl2, and 30 uM S. pneumoniae ACP synthase in 50 mM NaHEPES, pH 7.5. The mixture was gently mixed on a magnetic stirrer at 23 °C for 2 hr, and the reaction was terminated by the addition of 15 mM EDTA . The reaction mixture was filtered through a 0.2 micron filter (Millipore) and applied to a MonoQ column (Pharmacia) equilibrated with 20 mM Tris-Cl, pH 7.5. The column was washed with buffer until all non-adherent material was removed (as observed by UV detection), and the crotonoyl-ACP was eluted with a linear gradient of 0 to 400 mM NaCl.
S. aureus Fabl Enzyme Inhibition Assay using crotonoyl-ACP:
Assays are carried out in half-area, 96-well microtitre plates. Compounds are evaluated in 150 uL assay mixtures containing 100 mM NaADA, pH 6.5 (ADA = N-(2-acetamido)-2- iminodiacetic acid), 4 % glycerol, 25 uM crotonoyl-ACP, 50 uM NADPH, and an appropriate dilution of 5. aureus Fab I (approximately 20 nM). Inhibitors are typically varied over the range of 0.01-10 uM. The consumption of NADPH is monitored for 20 minutes at 30 °C by following the change in absorbance at 340 nm. Initial velocities are estimated from a linear fit of the progress curves. IC50's are estimated from a fit of the initial velocities to a standard, 4-parameter model (Equation 1) and are typically reported as the mean ± S.D. of duplicate determinations. Compounds of this invention in this assay have IC5o's from about 100.0 micromolar to about 0.04 micromolar. The apparent Ki is calculated from Equation 2 assuming the inhibition is competitve with crotonoyl-ACP.
Equation 1: v = Range/(l+[I]/IC50) s + Background
Equation 2: Ki(app) = IC50/( 1+[S]/Ks) FabK Enzyme Inhibition Assay
FabK catalyses the reduction of enoyl-ACPs with the concomitant oxidation of NADH. The reduction of crotonoyl-ACP to butyryl- ACP can be monitored by following the change in absorbance at 340 nm as NADH is oxidized.
Assays were carried out in Costar 3696 half-area plates in a final assay volume of 150 uL on a Spectramax platereader. The substrates (NADH and crotonoyl-ACP) were incubated with FabK enzyme in 100 mM N-[2-acetamido]-2 iminodiacetic acid (ADA), pH 6.5, 100 mM NH4CI, 4% glycerol at 30 °C and the reaction was monitored at 340 nm.
Using the above assay, compounds were tested for inhibition of FabK. 30 uL of inhibitor was added to a well of the plate. 30 uL of a 250 uM stock of NADH and 60 uL of a 67.5 uM stock of crotonoyl ACP were then added to the well. The plate was incubated at 30 °C for 5 min. The reaction was initiated by adding 30 uL of a 6.25 nM stock of enzyme to the well (also pre-incubated at 30 °C). The reaction was then monitored at A340 nm for 30 min at 30 °C. Positive controls were reactions without compound. Negative controls were reactions without enzyme and without compound. Final concentrations in the assay mixture were 25 uM crotonoyl-ACP, 50 uM NADH, and 1.25 nM enzyme.
IC50s were determined for compounds by carrying out the assay at 8 different concentrations of compound (100 uM-0.75 uM) in duplicate. The IC50 was calculated using Grafit software (v 4.09). The two Fab K inhibitors of this invention have IC50's of about 5 micromolar.
Antimicrobial Activity Assay:
Whole-cell antimicrobial activity was determined by broth microdilution using the National Committee for Clinical Laboratory Standards (NCCLS) recommended procedure, Document M7-A4, "Methods for Dilution Susceptibility Tests for Bacteria that Grow Aerobically". The compound was tested in serial two-fold dilutions ranging from 0.06 to 64 mcg/mL. Test organisms were selected from the following laboratory strains: Staphylococcus aureus Oxford, Staphylococcus aureus WCUH29, Streptococcus pneumoniae ERY2, Streptococcus pneumoniae 1629, Streptococcus pneumoniae N 1387, Enterococcus faecalis I, Enterococcus faecalis 7, Haemophilus influenzae Ql, Haemophilus influenzae NEMC1, Moraxella Catarrhalis 1502, Escherichia coli 7623 AcrABEFD+, Escherichia coli 120 AcrAB-, Escherichia coli MG1655, Escherichia coli MG1658. The minimum inhibitory concentration (MIC) was determined as the lowest concentration of compound that inhibited visible growth. A mirror reader was used to assist in determining the MIC endpoint.
One skilled in the art would consider any compound with a MIC of less than 256 μg/mL to be a potential lead compound. Preferably, the compounds used in the antimicrobial assays of the present invention have a MIC value of less than 128 μg/mL. Most preferably, said compounds have a MIC value of less than 64 μg/mL.
According to the instant invention, the preferred Fab I and Fab K enzyme inhibition assays use crotonoyl-ACP, rather than crotonoyl CoA, as a substrate. Thus, this invention comprises the preparation and purification of crotonoyl-ACP and the use of this purified enzyme in Fab I and Fab K enzyme inhibition assays. Crotonoyl-ACP was synthesised using 5. pneumoniae ACP synthase to catalyse the addition of a crotonoyl group from crotonoyl CoA to E.coli apo-acyl carrier protein (ACP). In a further aspect of this invention, it is contemplated that an apo-acyl carrier protein from any bacterial species, such as from Escherichia coli, Staphylococcus and Streptococcus, can be used in the preparation of crotonoyl-ACP. This synthesis was carried out in the presence of magnesium chloride in NaHEPES, pH 7.5. The reaction was complete in 2 hours at a reaction temperature of about 20-30°C, preferably at 23°C.
The purified crotonoyl-ACP prepared above is then used in the Fab I and Fab K assays to determine the inhibitors of the instant invention. Assays may be carried out, for example, in Costar 3696 half-area plates, preferably at a final assay volume of 150 ul on a Spectramax platereader. Preferred substrates used in the methods of the invention are NADH, NADPH, an NADH analogue and crotonoyl-ACP. Further provided are preferred methods comprising the step of incubating substrates with Fab I or Fab K in 100 mM N-[2- acetamido]-2 iminodiacetic acid (ADA), pH 6.5. This reaction may be monitored at 340 nm, among other wavelengths.
The examples which follow are intended in no way to limit the scope of this invention, but are provided to illustrate how to make and use the compounds of this invention. Many other embodiments will be readily apparent to those skilled in the art.
EXAMPLES
General
Proton nuclear magnetic resonance (*H NMR) spectra were recorded at either 300 or 360 MHz, and chemical shifts are reported in parts per million (δ)downfield from the internal standard tetramethylsilane (TMS). Abbreviations for NMR data are as follows: s = singlet, d = doublet, t = triplet, q = quartet, m = multiplet, dd = doublet of doublets, dt = doublet of triplets, app = apparent, br = broad. J indicates the NMR coupling constant measured in Hertz. CDCI3 is deuteriochloroform, DMSO-d6 is hexadeuteriodimethylsulfoxide, and CD3OD is tetradeuteriomethanol. Mass spectra were obtained using electrospray (ES) ionization techniques. Elemental analyses were performed by Quantitative Technologies Inc., Whitehouse, NJ. Melting points were obtained on a Thomas-Hoover melting point apparatus and are unconnected. All temperatures are reported in degrees Celsius. Analtech Silica Gel GF and E. Merck Silica Gel 60 F-254 thin layer plates were used for thin layer chromatography. Flash chromatography was carried out on E. Merck Kieselgel 60 (230-400 mesh) silica gel.
Analytical HPLC was performed on Beckman chromatography systems. Preparative HPLC was performed using Gilson chromatography systems. ODS refers to an octadecylsilyl derivatized silica gel chromatographic support. YMC ODS-AQ® is an ODS chromatographic support and is a registered trademark of YMC Co. Ltd., Kyoto, Japan. PRP-1® is a polymeric (styrene-divinylbenzene) chromatographic support, and is a registered trademark of Hamilton Co., Reno, Nevada. Celite® is a filter aid composed of acid-washed diatomaceous silica, and is a registered trademark of Manville Corp., Denver, Colorado.
Preparation 1
Preparation of l-methyl-2-(methylaminomethyl1-lH-indole
a) Ethyl l-methyl-l//-indole-2-carboxylate
NaΗ (60% dispersion in mineral oil, 8.02 g, 200.49 mmole) was washed with hexanes, then was suspended in dry DMF (530 mL). Solid ethyl indole-2-carboxylate (25.29 g, 133.66 mmole) was added portionwise over 5 - 10 min, allowing gas evolution to subside between additions. When the addition was complete, the yellow mixture was stirred for 15 min, then methyl iodide (42 mL, 668.3 mmole) was added all at once. The reaction was exothermic, and the internal temperature rose to 40 - 45°C. After 1 hr, the reaction was quenched with 10% NΗ4CI (100 mL) and concentrated on the rotavap (high vacuum). The residue was partitioned between Et2O(500 mL) and H20 (100 mL), and the layers were separated. The Et20 layer was washed with H20 (100 mL), dried (MgSθ4), and concentrated to leave the title compound (27.10 g, quantitative) as a light yellow solid. This was used without further purification: TLC (10% EtOAc/hexanes) Rf = 0.39.
b) N, 1 -Dimethyl- lH-indole-2-carboxamide
A suspension of ethyl l-methyl-lH-indole-2-carboxylate (27.10 g, 133.34 mmole) in 40% aqueous CΗ3NΗ2 (300 mL) and MeOH (30 mL) was stirred at RT. A solid tended to gradually creep up the walls of the flask, and was washed down periodically with MeOH. The flask was tightly stoppered to keep the material inside the flask. As the reaction proceeded, the solid dissolved, but eventually the product began to precipitate. The reaction was stirred at RT for 5 days, then was concentrated to remove approximately 200 mL of the solvent. The remaining residue was diluted with H20 (300 mL), and the solid was collected by suction filtration and washed with H20. Drying at 50 - 60°C in high vacuum left the title compound (23.45 g, 93%) as a faintly yellow solid: ΪH NMR (300 MHz, CDCI3) 8 7.63 (d, J = 8.0 Hz, 1 H), 7.27 - 7.43 (m, 2 H), 7.10 - 7.20 (m, 1 H), 6.80 (s, 1 H), 6.10 - 6.30 (m, 1 H), 4.06 (s, 3 H), 3.01 (d, J = 4.9 Hz, 3 H).
c) 1 -Methyl-2-(methylaminomethy 1)- 1 H-indole
A 3-liter 3-necked roundbottom flask equipped with overhead stirring was charged with N,l -dimethyl- l -'-indole-2-carboxamide (23.45 g, 124.58 mmole) and anhydrous TΗF (170 mL). The solution was stirred while a solution of LiAlΗ4 in THF (1.0 M, 250 mL, 250 mmole) was added via syringe. Gas was evolved during the addition of the first 50 L of L.AIH4 solution. When the addition was complete, the resulting light yellow solution was heated at gentle reflux. After 23 hr, the reaction was cooled in ice and quenched by the sequential dropwise addition of H20 (9.5 mL), 15% NaOH (9.5 mL), and H20 (28.5 mL). The mixture was stirred for 15 min, then was filtered through celite®, and the filter pad was washed thoroughly with THF. The filtrate was concentrated and the residue was flash chromatographed on silica gel (10% MeOH/CHCl3 containing 0.5% cone. NH4OH). The title compound (20.17 g, 93%) was obtained as a light yellow oil: 1H NMR (300 MHz, CDCI3) δ 7.56 (d, J = 7.8 Hz, 1 H), 7.02 - 7.35 (m, 3 H), 6.38 (s, 1 H), 3.88 (s, 2 H), 3.75 (s, 3 H), 2.49 (s, 3 H).
Preparation 2
Preparation of (E)-3-(6-aminopyridin-3-yl)acrylic acid (Method A)
a) Benzyl (E)-3-(6-aminopyridin-3-yl)acrylate A solution of 2-amino-5-bromopyridine (2.25 g, 13.0 mmole), benzyl acrylate (3.2 g, 19.7 mmole), Pd(OAc)2 (0.31 g, 1.4 mmole), tri-ort/iσ-tolylphosphine (0.73 g, 2.4 mmole), and diisopropylethylamine (3.5 mL, 20.0 mmole) in propionitrile (50 mL) was heated at reflux overnight. The dark mixture was filtered through celite®, and the filtrate was concentrated. Flash chromatography on silica gel (3% MeOH/CH2Cl2) gave the title compound (1.3 g, 39%): MS (ES) m/e 255 (M + H)+.
b) (E)-3-(6-Aminopyridin-3-yl)acrylic acid
A solution of benzyl (E)-3-(6-aminopyridin-3-y.)acrylate (1.3 g, 5.1 mmole) and 1.0 N NaOH (10 mL, 10 mmole) in MeOH was heated at reflux overnight. The solution was concentrated in vacuo, and the residue was dissolved in H20. The pH was adjusted to 6 with dilute HCl, and the solid precipitate was collected by suction filtration and dried to give the title compound (0.6 g, 72%) as a white solid: MS (ES) m/e 165 (M + H)+.
Preparation 3
Preparation of (E)-3-(6-aminopyridin-3-yl)acrylic acid (Method B)
a) (E)-3-(6-Aminopyridin-3-yl)acrylic acid
Acrylic acid (23 mL, 0.33 mole) was added carefully to a solution of 2-amino-5- bromopyridine (25.92 g, 0.15 mole) and Na2C03 (55.64 g, 0.53 mole) in H20 (600 mL). PdCl2 (0.53 g, 0.003 mole) was then added, and the mixture was heated at reflux. After 24 hr, the reaction was cooled to RT and filtered, and the filtrate was adjusted to pH 6 with aqueous HCl. Additional H20 (0.5 L) was added to improve mixing, and the mixture was stirred for 1 hr. The pH was readjusted to 6, then the solid was collected by suction filtration. The filter pad was washed sequentially with H 0 (2 x 0.5 L), cold absolute EtOH (100 mL), and Et 0 (2 x 250 mL). Drying in high vacuum at elevated temperature gave the title compound (15.38 g, 62%) as a tan solid: IH NMR (300 MHz, DMSO-dg) δ 8.1 1 (d, J = 2.0 Hz, 1 H), 7.75 (dd, J = 8.7, 2.0 Hz, 1 H), 7.43 (d, J = 15.8 Hz, 1 H), 6.53 (s,- 2 H), 6.45 (d, J = 8.7 Hz, 1 H), 6.22 (d, J = 15.8 Hz, 1 H); MS (ES) m/e 165 (M + H)+.
Preparation 4
Preparation of l-rnethyl-3-(methylarninomethyl)-lH-indazole
a) Methyl (l-methyl-lH-indazole)carboxylate Indazole-3-carboxylic acid (5.0 g, 30 mmole), K2C03 (12.4 g, 90 mmole), and Mel
(9.3 mL, 150 mmole) were combined in dry DMF (100 mL) and heated to 50 °C. After 18 hr the mixture was cooled to RT and concentrated in vacuo. The residue was taken up in EtOAc and filtered, and the filtrate was concentrated under reduced pressure. The residue was chromatographed on silica gel (25% EtOAc/hexanes) to give the title compound (3.88 g, 68%) as a yellow solid: 1H NMR (300 MHz, CDC13) δ 8.24 (m, 1 H), 7.47 (m, 2 H), 7.34 (m, 1 H), 4.19 (s, 3 H), 4.05 (s, 3 H).
b) N,l -Dimethyl- lH-indazole-3-carboxamide
A suspension of methyl (l-methyl-lH-indazole)carboxylate (3.88 g, 20.4 mmole) in 40% aqueous CΗ3NΗ2 (100 mL) and MeOH (5 mL) was stirred at RT for 4 hr. During that time the suspension became a solution. The mixture was concentrated to approximately 1/3 by volume at which time the product precipitated as a pale yellow solid. The solid was collected by filtration, washed with H20, and dried in vacuo to give the title compound (3.42 g, 89%) which was sufficiently pure for use in the next step: 1H NMR (300 MHz, CDCI3) δ 8.24 (m, 1 H), 7.47 (m, 2 H), 7.34 (m, 1 H), 6.95 (bs, 1 H), 4.19 (s, 3 H), 3.05 (d, J = 12.0 Hz, 3 H).
c) l-Methyl-3-(methylaminomethyl)- lH-indazole
To a solution of N,l-dimethyl-lH-indazole-3-carboxamide (3.42 g, 18 mmole) in dry TΗF (90 mL) was added a solution of LiAlΗ4 in THF (1.0 M, 36 mL, 36 mmole) slowly at RT. After 2 hr the mixture was heated to a gentle reflux. After 4 hr the mixture was cooled to RT and quenched by dropwise addition of 2.0 M NaOH until a white solid had formed. The mixture was dried (MgSθ4), filtered, and concentrated under reduced pressure to give the title compound (3.28 g, 100%) as an oil which was sufficiently pure for use in the next step: MS (ES) m/e 176 (M + H)+.
Preparation 5
Preparation of (E)-3-(3,4-dihydro-2//-pyrido[3,2-b]-l,4-oxazin-7-yl)acrylic acid
a) 3,4-Dihydro-2H-pyrido[3,2-fc]-l,4-oxazine
To a suspension of 2H-pyrido[3,2-έ]-l,4-oxazin-3(4H)-one (2.0 g, 13.3 mmole) in dry TΗF (40 mL) was added a solution of LiAlΗ4 in THF (1.0 M, 26.6 mL, 26.6 mmole) slowly at 0°C. After 1 hr the mixture was quenched with 2.0 M NaOH until a solid formed.
The mixture was dried (MgSθ4), filtered, and concentrated under reduced pressure to give the title compound (1.44 g, 79%) as a white solid which was sufficiently pure for use in the next step: MS (ES) /e 137 (M + H)+.
b) 4-(terr-Butoxycarbonyl)-3,4-dihydro-2H-pyrido[3,2-/?]- 1 ,4-oxazine
To a solution of 3,4-dihydro-2H-pyrido[3,2-fo]-l,4-oxazine (1.44 g, 10.6 mmole) and di-tert-butyl dicarbonate (2.78 g, 12.7 mmole) in dry TΗF (50 mL) was added a solution of LiHMDS in THF (1.0 M, 12.7 mL, 12.7 mmole) dropwise at 0°C. After 30 min the mixture was quenched with saturated NH4CI and extracted with EtOAc (3x). The combined organic layers were dried (MgS04), filtered, and concentrated. Flash chromatography on silica gel (40% EtOAc/hexanes) gave the title compound (2.0 g, 80%) as a clear oil: MS (ES) m/e 237 (M + H)+.
c) 4-(t -Butoxycarbonyl)-7-bromo-3,4-dihydro-2H-pyrido[3,2-έ>]-l,4-oxazine
To a solution of 4-(tert-butoxycarbonyl)-3,4-dihydro-2H-pyrido[3,2-b]-l,4-oxazine (2.0 g, 8.46 mmole) in MeOΗ (40 mL) was added Br2 (0.53 mL, 10.2 mmole) dropwise at 0°C. After 1 hr the mixture was concentrated. The residue was taken up in 1: 1
Et20/hexanes and filtered. The filtrate was concentrated under reduced pressure to give the title compound (1.27 g, 48%) as an oil which solidified under vacuum: ^Η NMR (400 MHz, CDCI3) δ 8.10 (s, 1 H), 7.33 (s, 1 H), 4.25 (m, 2 H), 3.92 (m, 2 H), 1.54 (s, 9 H). d) (E)-3-[4-(tert-Butoxycarbonyl)-3,4-dihydro-2H-pyrido[3,2-/?]-l,4-oxazin-7-yl]acrylic acid
A solution of 4-(rert-butoxycarbonyl)-7-bromo-3,4-dihydro-2H-pyrido[3,2-_?]- 1 ,4- oxazine (1.27 g, 4.03 mmole), benzyl acrylate (785 mg, 4.84 mmole), Pd(OAc)2 (45 mg, 0.20 mmole), P(o-tolyl)3 (122 mg, 0.4 mmole), and (i-Pr)2NEt (1.76 mL, 10.1 mmole) in propionitrile (20 mL) was degassed (3 x N2/vacuum) then heated to reflux. After 18 hr the mixture was cooled to RT and concentrated. Flash chromatography on silica gel (25% EtOAc/hexanes) gave the title compound (1.17 g, 73%) as a yellow oil: MS (ES) m/e 397 (M + Η)+.
e) (E)-3-(3,4-dihydro-2H-pyrido[3,2--?][l,4]oxazin-7-yl)acrylic acid
(E)-3-[4-(tert-Butoxycarbonyl)-3,4-dihydro-2H-pyrido[3,2--j]-l,4-oxazin-7- yljacrylic acid (1.17 g, 2.95 mmole) was dissolved in 4 N ΗC1 in dioxane (15 mL). After 72 hr the mixture was concentrated. The residue was taken up in 1:1 MeOΗ/Η20 (20 mL). 1.0 N LiOH (15 mL, 15 mmole) was added and the mixture was heated to reflux. After 18 hr the mixture was cooled to RT and concentrated to approximately 1/3 volume. The mixture was adjusted to pH 6 using 10% HCl. The solid was collected by filtration, washed with H20 and dried in vacuo to give the title compound (315 mg, 52% over 2 steps): MS (ES) /e 207 (M + H)+.
Preparation 6
Preparation of (E)-3-(5,6,7,8-tetrahydro-l,8-naphthyridin-3-yl)acrylic acid
a) l,2,3,4-Tetrahydro-l,8-naphthyridine
1,8-Naphthyridine (1.0 g, 7.68 mmole) was hydrogenated (50 psi) with 10% Pd/C (100 mg) in absolute ethanol (40 mL) for 18 hr. The mixture was filtered through a pad of Celite® and the filtrate was concentrated to give the title compound (1.04 g) which was sufficiently pure for use in the next step: MS (ES) m/e 135 (M + H)+.
b) l-(tert-Butoxycarbonyl)- 1 ,2,3,4-tetrahydro- 1 ,8-naphthyridine
To a solution of l,2,3,4-tetrahydro-l,8-naphthyridine (1.04 g, 7.68 mmole) and di- tert-butyl dicarbonate (2.01 g, 9.22 mmole) in dry THF (40 mL) was added a solution of LiHMDS in TΗF (1.0 M, 9.22 mL, 9.22 mmole) dropwise at 0°C. After 30 min the mixture was quenched with saturated NH4CI and extracted with EtOAc (3x). The combined organic layers were dried (MgS04), filtered, and concentrated. Flash chromatography on silica gel (40% EtOAc/hexanes) gave the title compound (1.37 g, 76% over 2 steps) as an orange oil which solidified under vacuum: lH NMR (400 MHz, CDC1 ) δ 8.33 (m, 1 H), 7.37 (m, 1 H), 6.94 (m, 1 H), 3.77 (m, 2 H), 2.75 (t, J = 6.5 Hz, 2 H), 1.93 (m, 2 H), 1.54 (s, 9 H).
c) 1 -(r -Butoxycarbonyl)-6-bromo- 1 ,2,3,4-tetrahydro- 1 ,8-naphthyridine
To a solution of l-( rt-butoxycarbonyl)-l,2,3,4-tetrahydro-l,8-naphthyridine (1.37 g, 5.85 mmole) in CH2C12 (30 mL) was added glacial HOAc (3.4 mL, 58.5 mmole) and NBS (1.09 g, 6.14 mmole). After 72 hr the mixture was washed with 2 .0 M NaOH, H20, and brine. The mixture was dried (MgS04), filtered, and concentrated under reduced pressure to give the title compound (1.79 g, 98%) which was sufficiently pure for use in the next step: *H NMR (400 MHz, CDCI3) δ 8.35 (s, 1 H), 7.51 (s, 1 H), 3.77 (m, 2 H), 2.75 (t, J = 6.5 Hz, 2 H), 1.93 (m, 2 H), 1.54 (s, 9 H).
d) Benzyl (E)-3-[8-(rert-butoxycarbonyl)-5,6,7,8-tetrahydro-l,8-naphthyridin-3-yl]acrylate A solution of l-(tert-butoxycarbonyl)-6-bromo-l,2,3,4-tetrahydro-l,8- naphthyridine (1.79 g, 5.70 mmole), benzyl acrylate (1.11 g, 6.84 mmole), Pd(OAc)2 (65 mg, 0.29 mmole), P(o-tolyl)3 (173 mg, 0.57 mmole), and (i-Pr)2NEt (2.5 mL, 14.25 mmole) in propionitrile (30 mL) was degassed (3 x N2/vacuum) then heated to reflux. After 18 hr the mixture was cooled to RT and concentrated. Flash chromatography on silica gel (25% EtOAc/hexanes) gave the title compound (1.21 g, 54%) as a yellow solid: 1H NMR (400 MHz, CDCI3) δ 8.44 (s, 1 H), 7.65 (d, J = 16.0 Hz, 1 H), 7.53 (s, 1 H), 7.40 (m, 5 H), 6.43 (d, J = 16.0 Hz, 1 H), 5.25 (s, 2 H). 3.77 (m, 2 H), 2.75 (t, J = 6.5 Hz, 2 H), 1.93 (m, 2 H), 1.54 (s, 9 H)
e) (E)-3-(5,6,7,8-Tetrahydro-l,8-naphthyridin-3-yl)acrylic acid
Benzyl (E)-3-[8-(t-;rt-butoxycarbony l)-5,6,7,8-tetrahydro- 1 ,8-naphthyridin-3- yljacrylate (1.21 g, 3.07 mmole) was dissolved in 4 N HCl in dioxane (15 mL). After 18 hr the mixture was concentrated. The residue was taken up in 1:1 MeOH/H20 (15 mL). 1.0 N LiOH (15 mL, 15 mmole) was added and the mixture heated to reflux. After 18 hr the mixture was cooled to RT and concentrated to approximately 1/3 volume. The mixture was adjusted to pH 6 using 10% HCl. The solid was collected by filtration, washed with H20, and dried in vacuo to give the title compound (180 mg, 29% over 2 steps): MS (ES) m/e 205 (M + H)+. Preparation 7
Preparation of 2-(methylaminomethyl)thieno[2,3-i>]thiophene
a) 3-(l,3-Dioxolan-2-yl)thiophene
To a solution of thiophene-3-carboxaldehyde (5.0 g, 44.58 mmole) in benzene (200 mL) was added ethylene glycol (25 mL, 445.8 mmole) and p-toluenesulfonic acid hydrate (848 mg, 4.458 mmole). The mixture was heated to reflux under a Dean-Stark trap. After 18 hr the mixture was cooled to RT, washed with saturated NaHCθ3 then with H20, dried (MgS04), and concentrated under reduced pressure to give the title compound (6.32 g,
91%) as a light amber oil: *H NMR (400 MHz, CDC13) δ 7.42 (s, 1 H), 7.32 (m, 1 H), 7.16 (m, 1 H), 5.91 (s, 1 H), 4.12-3.99 (m, 4 H).
b) 2-(Carboethoxy methy lthio)-3-( 1 ,3-dioxolan-2-y l)thiophene To a solution of 3-(l,3-dioxolan-2-yl)thiophene (6.32 g, 40.46 mmole) in dry THF
(200 mL) was added asolution of n-BuLi in hexanes (1.7 M,- 28.8 mL, 49 mmole) slowly at -78°C. After 30 min sulfur (1.57 g, 49 mmole) was added all at once. After 30 min ethyl bromoacetate (7.4 mL, 66.87 mmole) was added slowly, and after another 30 min the mixture was warmed to RT. After 2 hr at RT the mixture was concentrated under reduced pressure. The residue was taken up in Et 0, washed with H20 (3x), dried (MgSθ4), and concentrated to give the title compound as an oil which was sufficiently pure for use in the next step.
c) 2-(Carboethoxymethylthio)-3-formylthiophene To a solution of 2-(carboethoxymethy.thio)-3-(l,3-dioxolan-2-yl)thiophene (from step b) in acetone (200 mL) was added p-toluenesulfonic acid (761 mg, 4.0 mmole) at RT. After 18 hr the mixture was concentrated. The residue was taken up in Et20, washed with saturated NaHCθ3, H20 (2x), dried (MgSθ4), and concentrated under reduced pressure to give the title compound as an oil which was sufficiently pure for use in the next step.
d) Ethyl thieno[2,3-£>]thiophene-2-carboxylate
To a solution of 2-(carboethoxymethylthio)-3-formylthiophene (from step c) in MeOH (200 mL) was added DBU (0.6 mL, 4.0 mmole) at 0°C. After 1 hr the mixture was warmed to RT and concentrated. The residue was taken up in EtOAc, washed with 10% HCl, H20 (3x), dried (MgS04), and concentrated. Flash chromatography on silica gel
(50% toluene/hexanes) gave the title compound (3.84 g, 45% over 4 steps) as an off-white solid: H NMR (400 MHz, CDCI3) δ 7.95 (s, 1 H), 7.40 (d, J = 5.2 Hz, 1 H), 7.26 (d, J = 5.2 Hz, 1 H), methyl ester 3.92 (s, 3 H), ethyl ester 4.38 (q, J = 7.1 Hz, 2 H)) and 1.41 (t, J = 2.4 Hz, 3 H).
e) N-Methyl-2-(thieno[2,3-b]thiophene)carboxamide
A suspension of ethyl thieno[2,3-fc]thiophene-2-carboxylate (3.84 g, 18.1 mmole) in 40% aqueous CH3NH2 (100 mL) and MeOH (lOmL) was stirred at RT for 18 hr. During that time the suspension became a solution. The mixture was concentrated to approximately 1/3 volume at which time the product precipitated. The solid was collected by filtration, washed with H20, and dried in vacuo to give the title compound (3.01 g,
85%): !H NMR (400 MHz, d6-DMSO) δ 8.60 (bs, 1 H), 7.92 (s, 1 H), 7.67 (d, J = 5.2 Hz, 1 H), 7.38 (d, J = 5.2 Hz, 1 H), 2.78 (d, J = 4.6 Hz, 3 H).
f) 2-(Methylaminomethyl)thieno[2,3-Z?]thiophene To a solution of N-methyl-2-(thieno[2,3-έ»]thiophene)carboxamide (3.01 g, 15.26 mmole) in dry THF (75 mL) was added a solution of L1AIH4 in THF (1.0 M, 30 mL, 30 mmole) slowly at RT. After gas evolution had ceased the mixture was heated to a gentle reflux. After 18 hr the mixture was cooled to RT and quenched by dropwise addition of 2.0 M NaOH until a white solid had formed. The mixture was dried over MgSθ4, filtered, and concentrated under reduced pressure to give the title compound (2.18 g, 78%) as a brown oil: 1H NMR (400 MHz, CDCI3) δ 7.30 (d, J = 5.2 Hz, 1 H), 7.15 (d, J = 5.2 Hz, 1 H), 7.04 (s, 1 H), 4.00 (s, 2 H), 2.49 (s, 3 H).
Preparation 8
Preparation of 2-(methylaminomethyl)thieno[3,2-£>]thiophene
a) N-Methyl-2-(thieno[3,2-b]thiophene)carboxamide
EDC (624 mg, 3.26 mmole) was added to a solution thieno[3,2-6]thiophene-2- carboxylic acid (500 mg, 2.71 mmole), CH3NH2 (2.0 M in THF, 2.7 mL, 5.42 mmole),
HOBt • H20 (440 mg, 3.26 mmole), and Et3N (0.95 mL, 6.78 mmole) in dry DMF (14 L) at RT. After 18 hr the mixture was diluted with H20 and extracted with EtOAc (3x). The combined organic layers were dried (MgSθ4) and concentrated to give the title compound (415 mg, 78%) which was sufficiently pure for use in the next step: ^H NMR (400 MHz, CDCI3) δ 7.70 (s, 1 H), 7.52 (d, J = 5.3 Hz, 1 H), 7.27 (d, J = 5.3 Hz, 1 H), 3.02 (d, J = 4.9 Hz, 3 H). b) 2-(Methylarninomethyl)thieno[3,2-b]thiophene
To a solution of N-methyl-2-(thieno[3,2-i>]thiophene)carboxamide (415 mg, 2.1 mmole) in dry THF (10 mL) was added a solution of L-AIH4 in THF (1.0 M, 4.2 mL, 4.2 mmole) slowly at RT. After gas evolution had ceased the mixture was heated to a gentle reflux. After 18 hr the mixture was cooled to RT and quenched by dropwise addition of 2.0 M NaOH until a white solid had formed. The mixture was dried (MgS04), filtered, and concentrated to give the title compound (361 mg, 94%) as a brown oil: !H NMR (400 MHz, CDCI3) δ 7.31 (d, J = 5.2 Hz, 1 H), 7.21 (d, J = 5.2 Hz, 1 H), 7.11 (s, 1 H), 4.01 (s, 2 H), 2.50 (s, 3 H).
Preparation 9
Preparation of (E)-3-(3/ -imidazo.4.5-£>lpyridin-6-yl)acrylic acid
a) 5-Bromo-2,3-diaminopyridine
To a suspension of 2-amino-5-bromo-3-nitropyridine (2.0 g, 9.17 mmole) in absolute EtOH (50 mL) was added SnCl2 hydrate (9.3 g, 41.3 mmole), then the mixture was heated to reflux. After 3 hr the mixture was cooled to RT and concentrated. The residue was taken up in 2.0 M NaOH and extracted with EtOAc (3x). The combined organic layers were dried (MgSθ4), filtered, and concentrated to give the title compound
(1.69 g, 98%) which was sufficiently pure for use in the next step: MS (ES) m/e 188/190
(M + H)+.
b) 6-Bromo-3H-imidazo[4,5-b]pyridine
5-Bromo-2,3-diaminopyridine ( 1.69 g, 8.99 mmole) was taken up in 96% formic acid (50 mL) and heated to reflux. After 18 hr the mixture was cooled to RT and concentrated. The residue was taken up in Η 0 and the pH was adjusted to 7 with 2.0 M NaOH. The title compound (1.54 g, 87%) was collected as a solid by filtration, washed with H20, and dried in vacuo: MS (ES) m/e 198/200 (M + H)+.
c) 6-Bromo-4-trityl-3 -imidazo[4,5-i>]pyridine
To a suspension of 6-bromo-3H-imidazo[4,5-b]pyridine (1.2 g, 6.06 mmole) in CΗ2C12 (30 mL) was added Et3N (1.3 mL, 9.09 mmole) then trityl chloride (2.03 g, 7.27 mmole) at RT. After 72 hr the mixture was washed with H20 (2x) and brine, then was dried (MgS04), filtered, and concentrated under reduced pressure to afford the title compound. This was used directly in the next step.
d) Benzyl (E)-3-(4-trityl-3H-imidazo[4,5-έ>]pyridin-6-yl)acrylate A solution of 6-bromo-4-trityl-3H-imidazo[4,5-£>]pyridine (from step a) (6.06 mmole), benzyl acrylate (1.18 g, 7.27 mmole), Pd(OAc)2 (67 mg, 0.30 mmole), P(o-tolyl)3 (183 mg, 0.6 mmole), and (i-Pr)2NEt (2.64 mL, 15.15 mmole) in propionitrile (30 mL) was degassed (3 x N2/vacuum) then heated to reflux. After 4 hr the mixture was cooled to RT and concentrated. Flash chromatography on silica gel (30% EtOAc/hexanes) gave the title compound (1.75 g, 55% over 2 steps) as an off-white foam: lK NMR (400 MHz, CDC13) δ 8.24 (d, J = 2.0 Hz, 1 H), 8.19 (d, J = 2.0 Hz, 1 H), 8.06 (s, 1 H), 7.77 (d, J = 16.0 Hz, 1 H), 7.42-7.11 (m, 20 H), 6.48 (d, J = 16.0 Hz, 1 H), 5.25 (s, 2 H).
d) (E)-3-(3H-Imidazo[4,5-b]pyridin-6-yl)acrylic acid Benzyl (E)-3-(4-trityl-3H-imidazo[4,5-Z?]pyridin-6-y-)acrylate (1.75 g, 3.35 mmole) was dissolved in 4 N ΗC1 in dioxane (20 mL). After 1 hr the mixture was concentrated. The residue was taken up in 1: 1 MeOΗ/Η20 (15 mL). 2.0 N NaOH (15 mL, 15 mmole) was added and the mixture was heated to reflux. After 18 hr the mixture was cooled to RT and concentrated to approximately 1/3 volume. The mixture was adjusted to pH 4 using 10% HCl. The solid was collected by filtration, washed with H20, and dried in vacuo to give the title compound (329 mg, 52% over 2 steps) as a white solid: 1H NMR (400 MHz, d6-DMSO) δ 9.10 (s, 1 H), 8.94 (s, 1 H), 8.84 (s, 1 H), 8.20 (d, J = 16.0 Hz, 1 H), 7.10 (d, J = 16.0 Hz, I H).
Preparation 10
Preparation of 6-methyl-5-(methylaminomethyl)-6H-thieno[2,3-fo]pyrrole
a) Ethyl (Z)-2-azido-3-(thiophen-3-yl)acrylate To a solution of thiophene-3-carboxaldehyde (500 mg, 4.46 mmole) and ethyl 2- azido acetate (863 mg, 6.69 mmole) in absolute EtOΗ (20 mL) was added NaOEt (21 %, 2.2 mL, 6.69 mmole) at 0°C. After 1 hr the mixture was quenched with saturated NΗ4CI and extracted with Et20 (3x). The combined organic layers were dried (MgS04), filtered, and concentrated. Flash chromatography on silica gel (50% CHC^/hexanes) gave the title compound (208 mg, 21%) as a pale yellow oil: !H NMR (400 MHz, CDCI3) δ 7.87 (m. I H), 7.49 (m, 1 H), 7.31 (m, 1 H), 6.96 (s, 1 H), 4.36 (q, J = 7.1 Hz, 2 H), 1.39 (t, J = 7.1 Hz, 3 H).
b) Ethyl 6H-thieno[2,3-£>]pyrrole-5-carboxylate
A solution of ethyl (Z)-2-azido-3-(thiophen-3-yl)acrylate (208 mg, 0.93 mmole) in xylenes (5 mL) was heated to reflux. After 30 min the mixture was cooled to RT and concentrated to give the title compound (175 mg, 96%) which was sufficiently pure for use in the next step: 1H NMR (400 MHz, CDC13) δ 9.26 (bs, 1 H), 7.10 (m, 1 H), 7.00 (m, 1 H), 6.91 (m, 1 H), 4.36 (q, J = 7.1 Hz, 2 H), 1.39 (t, J = 7.1 Hz, 3 H).
c) N,6-Dimethyl-6H-thieno[2,3--7]pyrrole-5-carboxamide
To a solution of ethyl 6/ -thieno[2,3-fc]pyrrole-5-carboxylate (175 mg, 0.9 mmole, see J. Het. Chem. 1984, 21, 215-217) and Mel (0.08 mL, 1.35 mmole) in dry DMF (5 mL) was added NaΗ (60% dispersion in mineral oil, 43 mg, 1.08 mmole) at 0°C. After 2 hr the mixture was quenched with saturated NΗ4CI and extracted with EtOAc (3x). The combined organic layers were dried (MgSθ4), filtered, and concentrated to an oil.
A solution of the above oil in 40% aqueous CH3NH2 (20 mL) and MeOH (1 mL) was stirred at RT for 18 hr. The mixture was concentrated to approximately 1/3 by volume at which time the product precipitated. The solid was collected by filtration, washed with H20, and dried in vacuo to give the title compound (134 mg, 74% over 2 steps): MS (ES) /e 195 (M + H)+.
d) 6-Methyl-5-(methylaminomethyl)-6/t -thieno[2,3-i»]pyrrole
To a solution of N,6-dimethyl-6-¥-thieno[2,3-b]pyrtole-5-carboxamide (134 mg, 0.69 mmole) in dry THF (5 mL) was added a solution of L.AIH4 in THF (1.0 M, 1.38 mL, 1.38 mmole) slowly at RT. After gas evolution had ceased the mixture was heated to a gentle reflux. After 2 hr the mixture was cooled to RT and quenched by dropwise addition of 2M NaOH until a white solid had formed. The mixture was dried (MgS04), filtered, and concentrated to give the title compound as a brown oil (142 mg, 100%) which was sufficiently pure for use in the next step: *H NMR (400 MHz, CDCI3) δ
6.95 (d, J = 5.2 Hz, 1 H), 6.78 (d, J = 5.2 Hz, 1 H), 6.27 (s, 1 H), 3.78 (s, 2 H), 3.72 (s, 3 H), 2.47 (s, 3 H). Preparation 11
Preparation of (E)-3-(2-aminopyrimidin-5-yl)acrylic acid
a) Benzyl (E)-3-(2-aminopyrimidin-5-yl)acrylate
According to the procedure of Preparation 2 (a), except substituting 5-bromo-2- aminopyrimidine ( 1.95 g, 11.2 mmole) for 2-amino-5-bromopyridine, the title compound (2.25 g, 79%) was prepared as a light orange solid: MS (ES) m/e 256 (M + H)+.
b) (E)-3-(2-Aminopyrimidin-5-yl)acrylic acid
According to the procedure of Preparation 2 (b), except substituting benzyl (E)-3- (2-aminopyrimidin-5-yl)acrylate (2.93 g, 11.5 mmole) for benzyl (E)-3-(6-aminopyridin-3- yl)acrylate, the title compound (1.71 g, 90%) was prepared as an off-white solid: MS (ES) m/e 166 (M + H)+.
Preparation 12
Preparation of (E)-3-(6-aminopyridin-3-yl)-2-methylacrylic acid
a) Methyl (E)-3-(6-aminopyridin-3-yl)-2-methylacrylate
According to the procedure of Preparation 2 (a), except substituting methyl crotonate (4.33 g, 43.3 mmole) for benzyl acrylate, the title compound (1.0 g, 18%) was prepared as an off-white solid: MS (ES) m/e 193 (M + H)+.
b) (E)-3-(6-Aminopyridin-3-yl)-2-methylacrylic acid
According to the procedure of Preparation 2 (b), except substituting methyl (E)-3- (6-aminopyridin-3-yl)-2-methylacrylate (1.0 g, 5.2 mmole) for benzyl (E)-3-(6- aminopyridin-3-yl)acrylate, the title compound (0.83 g, 90%) was prepared as an off-white solid: MS (ES) e 179 (M + H)+.
Preparation 13
Preparation of (EV3-(6-amino-2-methylpyridin-3-yl')acrylic acid
a) Benzyl (E)-3-(6-amino-2-methylpyridin-3-yl)acrylate
According to the procedure of Preparation 2 (a), except substituting 2-amino-5- bromo-6-methylpyridine (5.00 g, 26.7 mmole) for 2-amino-5-bromopyridine, the title compound (5.58 g, 78%) was prepared as an off-white solid: MS (ES) m/e 269 (M + H)+.
b) (E)-3-(6-Amino-2-methylpyridin-3-yl)acrylic acid
According to the procedure of Preparation 2 (b), except substituting benzyl (E)-3- (6-amino-2-methylpyridin-3-yl)acrylate (2.20 g, 8.2 mmole) for benzyl (E)-3-(6- aminopyridin-3-yl)acrylate, the title compound (1.31 g, 90%) was prepared as an off-white solid: MS (ES) /e 179 (M + H)+.
Preparation 14
Preparation of (E)-3-(6-amino-5-methylpyridin-3-yl)acrylic acid
a) Benzyl (E)-3-(6-amino-5-methylpyridin-3-yl)acrylate
According to the procedure of Preparation 2 (a), except substituting 2-amino-5- bromo-3-methylpyridine (5.00 g, 26.7 mmole) for 2-amino-5-bromopyridine, the title compound (6.37 g, 89%) was prepared as an off-white solid: MS (ES) m/e 269 (M + H)+.
b) (E)-3-(6-Amino-5-methylpyridin-3-yl)acrylic acid
According to the procedure of Preparation 2 (b), except substituting benzyl (E)-3- (6-amino-5-methylpyridin-3-yl)acrylate (5.00 g, 18.6 mmole) for benzyl (E)-3-(6- aminopyridin-3-yl)acrylate, the title compound (2.98 g, 90%) was prepared as an off-white solid: MS (ES) /e 179 (M + H)+.
Preparation 15
Preparation of (EV3-r6-amino-5-(hydroxymethyl')pyridin-3-yllacrylic acid
a) 2-Amino-3-(hydroxymethyl)pyridine
To a solution of 2-aminonicotinic acid (20.5 g, 148.1 mmole) in THF was added lithium aluminum hydride (300 mL, 1.0 M in THF) over 30 minutes. The reaction solution was heated to reflux for 18 hrs and then was cooled to room temperature. The reaction was quenched by the sequential dropwise addition of H20 (11.5 mL), 15% NaOH (11.5 mL), and H20 (34.5 mL). The mixture was stirred for 15 min, then was filtered through celite®, and the filter pad was washed thoroughly with THF followed by 5% CH3OH/CHCI3. The filtrate was concentrated to give the title compound (15.24 g, 83%) as a waxy light yellow solid: MS (ES) m/e 125 (M + H)+.
b) 2-Amino-5-bromo-3-(hydroxymethyl)pyridine
To a solution of 2-amino-3-(hydroxymethyl)pyridine (13.0 g, 116.0 mmole) in
CH2C12 (300 mL) at RT was added NBS (22.71 g, 127.6 mmole). After stirring at RT for
45 min the reaction solution was concentrated and the residue was dissolved in CHCI3.
The resulting suspension was filtered and the filtrate was concentrated to a dark oil. Purification on silica gel (EtOAc) afforded the title compound (78%, 18.36 g) as a tan solid: MS (ES) m/e 204 (M + H)+.
c) Benzyl (E)-3-[6-amino-5-(hydroxymethyl)pyridin-3-yl]acrylate
According to the procedure of Preparation 2 (a), except substituting 2-amino-3- (hydroxymethyl)-5-bromopyridine (1.10 g, 5.42 mmole) for 2-amino-5-bromopyridine, the title compound (1.25 g, 81%) was prepared as an off-white solid: MS (ES) m/e 285 (M + H)+.
d) (E)-3-[6-Amino-5-(hydroxymethyl)pyridin-3-yl]acrylic acid According to the procedure of Preparation 2 (b) except substituting benzyl-(E)-3-
[6-amino-5-(hydroxymethyl)pyridin-3-yl]acrylate (1.10 g, 5.42 mmole) for benzyl (E)-3- (6-aminopyridin-3-yl)acrylate, the title compound (0.68 g, 65%) was prepared as an off- white solid: MS (ES) m/e 194 (M + H)+. Preparation 16
Preparation of 6-bromo-3.4-dihydro- IH- 1.8-naphthyridin-2-one
a) 2-Amino-5-bromo-3-(bromomethyl)pyridine hydrobromide
A solution of 2-amino-5-bromo-3-hydroxymethylpyridine (5.00 g, 24.6 mmole), from Preparation 14 (b), in 48% aqueous ΗBr (50 mL), was heated at reflux for 12 hrs. The reaction was concentrated and toluene was used to azeotrope the residual Η20. The resulting light brown solid was placed under high vacuum overnight and used directly.
b) Methyl (±)-6-bromo-2-oxo-l,2,3,4-tetrahydro-lH-l,8-naphthyridine-3-carboxylate
To a solution of sodium methoxide (20.57 mL, 25% wt in CΗ3OΗ) in CH3OH (75 mL) was added dimethyl malonate (11.87 g, 89.9 mmole). After 30 min the 2-amino-5- bromo-3-(bromomethyl)pyridine hydrobromide salt prepared above was added to the methoxide solution and the reaction was stirred at RT overnight. The reaction slurry was concentrated to dryness under vacuum and then suspended in 1 : 1 H20/Et20. The remaining solids were filtered and washed with H20 then with hexanes to afford the title compound (4.08 g, 58 %) as a white solid after drying: MS (ES) m/e 286 (M + H)+.
c) 6-Bromo-3,4-dihydro-lH-l,8-naphthyridin-2-one
To a solution of methyl (±)-6-bromo-2-oxo-l,2,3,4-tetrahydro-lH-l,8- naphthyridine-3-carboxylate (2.00 g, 7.0 mmole) in CΗ3OΗ (75 mL) was added 1.0 M NaOH (30 mL). The reaction was heated to reflux for 4 hrs and then cooled to RT. The reaction was neutralized with 1.0 M HCl (30 mL) then was heated at reflux overnight. The reaction slurry was concentrated to dryness and the residues was suspended in 95:5
CHCI3/CH3OH. The solids were removed by filtration and the filtrate was concentrated to afford the title compound (1.40 g, 88%) as an off-white solid: MS (ES) m/e 228 (M + H)+.
Preparation 17
Preparation of (E)-3-r6-amino-5-r(2-hydroxyethylamino)carbonyllpyridin-3-yllacrylic acid
a) 2-Amino-5-bromo-N-(2-hydroxyethyl)nicotinamide
EDC (2.91 g, 15.2 mmole) was added to a solution 2-amino-5-bromonicotinic acid (3.00 g, 13.8 mmole), ethanola ine (0.93 g, 15.2 mmole), HOBt • H20 (2.05 g, 15.2 mmole), and diisopropylethylamine (2.64 mL, 15.2 mmole) in DMF (50 mL) at RT and the reaction solution was stirred overnight. The reaction contents were poured into H 0 (200 mL) and the resulting mixture was extracted with EtOAc (2 x 200 mL). The combined organic extracts were washed with H20 and brine and then dried over Na2Sθ4. Concentration of the organic extracts afforded the title compound as a yellow solid which was used without further purification: MS (ES) m/e 261 (M + H)+.
b) Benzyl (E)-3-[6-amino-5-[(2-hydroxyethylamino)carbonyl]pyridin-3-yl]acrylate
According to the procedure of Preparation 2 (a), except substituting 2-amino-5- bromo-N-(2-hydroxyethyl)nicotinamide (2.70 g, 10.4 mmole) for 2-amino-5- bromopyridine, the title compound (2.67 g, 75%) was prepared as an off-white solid: MS (ES) m/e 342 (M + H)+.
c) (E)-3-[6-Amino-5-[(2-hydroxyethylamino)carbonyl]pyridin-3-yl]acrylic acid
According to the procedure of Preparation 2 (b), except substituting benzyl (E)-3- [6-amino-5-[(2-hydroxyethy.amino)carbonyl]pyridin-3-yl]acrylate (2.67 g, 7.8 mmole) for benzyl (E)-3-(6-aminopyridin-3-yl)acrylate the title compound (1.37 g, 70%) was prepared as an off-white solid: MS (ES) m/e 252 (M + H)+.
Preparation 18
Preparation of 6-bromo-3-methyl-3.4-dihydro-l -pyrido[2.3--Jlpyrimidin-2-one
a) 2-Amino-5-bromo-3-(methylaminornethyl)pyridine
A solution of 2-amino-5-bromo-3-(hydroxymethyl)pyridine (5.00 g, 24.6 mmole), from Preparation 14 (b), in 48% aqueous HBr (50 mL) was heated at reflux for 12 hrs. The reaction was concentrated and toluene was used to azeotrope the residual H 0. The resulting light brown solid was placed under high vacuum overnight and used directly. A solution of the 2-amino-3-(bromomethyl)-5-bromopyridine hydrobromide salt
(prepared above) in 40% aqueous methylamine (50 mL) and THF (50 mL) was stirred at RT overnight in a pressure bottle. The reaction solution was concentrated and extracted with EtOAc (2 x 100 mL). The combined organic phases were washed with H20, dried over Na Sθ4 and concentrated. Purification on silica gel afforded the title compound (4.25 g, 80 %) as a yellow oil: MS (ES) m/e 217 (M + H)+. b) 6-Bromo-3-methyl-3,4-dihydro-lH-pyrido[2,3-- |pyrimidin-2-one
To a solution of dimethyl carbonate (2.14 g, 23.7 mmole) and sodium methoxide (1.0 mL, 4.5 mmole, 25% wt in CΗ3OΗ) in CH3OH (25 mL) was added 2-amino-5-bromo- 3-(methylaminomethyl)pyridine (1.0 g, 4.62 mmole). The reaction was heated at 50 °C overnight, diluted with H20 (1 mL) and concentrated. Toluene was added to the reaction residue and the contents were heated to reflux for 12 hr under a Dean-Stark apparatus. The reaction was cooled to RT, diluted with EtOAc, and washed with H20. Purification on silica gel (9: 1 CHCI3/CH3OH containing 5% NH4OH) gave the title compound (0.75 g, 67 %) as an off-white solid: MS (ES) m/e 243 (M + H)+.
Preparation 19
Preparation of 4-methyl-5-(methylaminomethyl)-4H-thienol3.2-Z?lpyrrole
a) Ethyl 4-methyl-4H-theino[3,2-έ»]pyrrole-5-carboxylate
According to the procedure of Preparation 1 (a), except substituting ethyl 4H- theino[3,2-/?]pyrrole-5-carboxylate (1.30 g, 6.7 mmole, see J. Het. Chem. 1984, 21, 215- 217) for ethyl indole-2-carboxylate, the title compound (1.35 g, 97%) was prepared as a yellow solid: MS (ES) m/e 210 (M + Η)+.
b) N,4-Dimethyl-4H-theino[3,2--?]pyrrole-5-carboxamide
According to the procedure of Preparation 1 (b), except substituting ethyl 4-methyl- 4H-theino[3,2-&]pyrrole-5-carboxylate (1.35 g, 6.5 mmole) for ethyl- 1-methy lindole-2- carboxylate, the title compound (1.19 g, 95%) was prepared as a yellow solid: MS (ES) m/e 195 (M + Η)+.
c) 4-Methyl-5-(methylaminomethyl)-4H-thieno[3,2--?]pyrrole
According to the procedure of Preparation 1 (c), except substituting N,4-dimethyl- 4H-theino[3,2--?]pyrrole-5-carboxamide (0.70 g, 3.6 mmole) for N,l-dimethylindole-2- carboxamide, the title compound (0.60 g, 92%) was prepared as a yellow oil: MS (ES) m/e 181 (M + Η)+. Preparation 20
Preparation of 3-methyl-2-(methylaminomethyl')indene hydrochloride
a) N,3-Dimethylinden-2-carboxamide
EDC (1.53 g, 0.01 mole) was added to a solution of 3-methyl-2-inden-2-carboxylic acid (1.91 g, 0.01 mole), methylamine hydrochloride (0.675 g, 0.01 mole), HOBt • H20 (1.53 g, 0.01 mole) and triethylamine (4.0 mL, 0.028 mole) in anhydrous DMF (80 mL) at RT. The reaction was stirred overnight, then was concentrated in vacuo. The residue was diluted with 5% NaHCθ3 and the resulting white precipitate was collected, washed with water and dried at 50°C in a vacuum oven to afford the title compound (1.6 g, 86%) as a white solid: MS (ES) m/e 188.2 (M + H)+.
b) 3-Methyl-2-(methylaminomethyl)indene hydrochloride A flame-dried flask was charged with anhydrous THF (15 mL) followed by solid lithium aluminum hydride (760 mg, 0.02 mole) at 0 °C. The mixture was stirred for 15 min, then a solution of N,3-dimethylindene-2-carboxamide (1.5 g, 0.008 mole) in anhydrous THF (20 mL) was added dropwise. When the addition was complete, the reaction was heated at gentle reflux for 30 hr, then was cooled in ice and quenched with H20 (1.4 mL) and NaF (2.5 g, 0.06 mole). The reaction mixture was stirred for 40 min then was filtered through celite®, and the filter pad was washed with THF. The filtrate was dried over K2Cθ3, filtered and concentrated to an oil, which was dissolved in anhydrous ethyl ether and treated with 4 M HCl in diethyl ether. The precipitated light tan solid was collected by suction filtration and washed with diethyl ether. Drying at 50 °C in a vacuum oven gave the title compound (1.05 g, 80.7%) as a light tan solid: MS (ES) /e 174.2 (M + H)+.
Preparation 21
Preparation of 2-(methylaminomethyl)indene hydrochloride
a) N-Methylindene-2-carboxamide
According to the procedure of Preparation 20 (a), except substituting 2-inden- carboxylic acid for 3-methyl-2-inden-2-carboxylic acid, the title compound was obtained as a white crystalline solid (1.45 g, 83.3%): MS (ES) m/e 174.2 (M + H)+. b) 2-(Methylaminomethyl)indene hydrochloride
According to the procedure of Preparation 20 (b), except substituting N- methylindene-2-carboxamide for N,3-dimethylindene-2-carboxamide, the title compound was obtained as an off-white solid (0.685 g, 87.6%): MS (ES) m/e 160.0 (M + H)+.
Preparation 22
Preparation of 4-methoxy-l-methyl-2-(methylarninomethyl)- lH-indole hydrochloride
a) Methyl 4-methoxy-l-methyl-lH-indol-2-carboxylate
NaΗ (60% dispersion in mineral oil, 0.3 g, 7.3 mmole) was washed with hexane then suspended in anhydrous DMF (16 mL). The mixture was cooled to 0 °C and methyl 4- methoxy-lH-indol-2-carboxylate (1.0 g, 4.87 mmole) was added. The mixture was stirred under argon for 10 min, then Mel (1.3 mL, 20 mmole) was added, and the thick slurry was stirred at RT for 2.5 hr. The reaction was quenched with 10% NΗ4CI (2 mL) and concentrated. The residue was partitioned between H20 and Et20, and the organic layer was dried over MgSU4 and concentrated to yield the title compound (1.03 g, 96%) as a white solid: MS (ES) m/e 220.2 (M + H)+.
b) N,l-Dimethyl-4-methoxy-lH-indol-2-carboxamide
A solution of methyl 4-methoxy-l -methyl- lH-indol-2-carboxylate (1.03 g, 4.7 mmole) in 2.0 M methylamine in methanol (40 mL) was sealed in a pressure bottle and heated at 55-60 °C for 60 hr. Concentration in vacuo yielded the title compound (1.05 g, quantitative) as a white solid: MS (ES) m/e 219.2 (M + Η)+.
c) 4-Methoxy- 1 -methyl-2-(methylaminomethy 1)- 1 W-indole hydrochloride
According to the procedure of Preparation 20 (b), except substituting N,l-dimethyl- 4-methoxy-lH-indol-2-carboxamide for N,3-dimethylindene-2-carboxamide, the title compound was obtained as an off white solid (0.72 g, 75%): MS (ES) m/e 205.2 (M + Η)+.
Preparation 23
Preparation of 1.4-dimethyl-2-(methylaminomethylHH-indole hydrochloride
a) l,4-Dimethyl- lH-indol-2-carboxylic acid
A solution of 1,4-dimethyl-lH-indole (0.9 g, 6.2 mmole) in anhydrous Et20 (20 mL) was treated with 2.5 M n-BuLi in hexanes (5.0 mL, 12 mmole) and the reaction was heated at reflux for 15 hr. The dark reaction mixture was poured into a slurry of excess crushed dry ice in Et20, and the mixture was allowed to stand for 1 hr. Water (10 mL) was added, the layers separated, and the aqueous layer was filtered through celite®. The clear filtrate was acidified with 2.0 N ΗC1 to pΗ 2, and the precipitate was collected and dried to afford the title compound (0.29 g, 26.4%) as an off-white solid: MS (ES) m/e 190.2 (M + Η)+.
b) N,l,4-Trimethyl-lH-indol-2-carboxamide
According to the procedure in Preparation 20 (a), except substituting 1,4-dimethyl- lH-indole-2-carboxylic acid for 3-methyl-2-indene-2-carboxylic, the title compound was obtained (0.184 g, 91%): MS (ES) m/e 203.2 (M + Η)+.
c) l,4-Dimethyl-2-(methylaminomethyl)-lH-indole hydrochloride
According to the procedure in Preparation 20 (b), except substituting N,l,4- trimethyl-lH-indole-2-carboxamide for N,3-dimethylindene-2-carboxamide, the title compound was obtained (0.13 g, 65%): MS (ES) m/e 189.2 (M + Η)+.
Preparation 24
Preparation of 2-(cyclopropylamino)-l -methyl- lH-indole
a) 2-(Cyclopropy lamino)- 1 -methyl- 1 H-indole To a solution of l-methylindole-2-carboxaldehyde (1.5 g,10 mmole), cyclopropylamine (1.14 g, 20 mmole), and glacial acetic acid (0.6 mL, 10 mmole)in MeOΗ (30 mL) was added NaBΗ3CN (0.69 g, 11 mmole). The reaction was stirred at RT overnight, then was concentrated in vacuo. The residue was diluted with 10% NaOH and extracted with CH2C12. The combined organic extracts were washed with brine, dried over MgSθ4, and concentrated. Flash chromatography on silica gel (3% MeOH CH2Cl ) gave the title compound (1.3 g.65%) as a semi-solid: MS (ES) m e-201 (M + H)+. Preparation 25
Preparation of 5-fluoro-2-(methylarninomethyl)- lH-indole
a) Ethyl 5-fluoro-l -methyl- lH-indole-2-carboxylate
According to the procedure of Preparation 1 (a), except substituting ethyl 5-fluoro- indole-2-carboxylate for the ethyl-indole-2-carboxylate, the title compound (3.3 g, 100%) was prepared as a white solid: MS (ES) m/e 222 (M + Η)+.
b) N, 1 -Dimethyl-5-fluoro- 1 H-indole-2-carboxamide
According to the procedure of Preparation 1 (b), except substituting ethyl 5-fluoro- 1 -methyl- lH-indole-2-carboxylate for the ethyl l-methyl-lH-indole-2-carboxylate, the title compound (2.1 g, 68%) was prepared as a white solid: MS (ES) m/e 207 (M + Η)+.
c) 5-Fluoro-2-(methy laminomethyl)- 1 H-indole
According to the procedure of Preparation 1 (c), except substituting N,l-dimethyl- 5-fluoro-lH-indole-2-carboxam.de for the N,l-dimethyl-lH-indole-2-carboxamide, the title compound (1.5 g, 78%) was prepared as a white solid: MS (ES) m/e 193 (M + Η)+.
Preparation 26
Preparation of 3-(methylaminomethyl)quinoline
a) 3-(Methylaminomethyl)quinoline
A solution of 3-quinolinecarboxaldehyde (1.5 g,10 mmole), 2.0 M CH3NH2/MeOH (10 mL, 20 mmole), glacial AcOH (0.6 mL, 10 mmole), and NaBH CN (0.35 g, 11 mmole) in MeOH (20 mL) was stirred at RT overnight, then was concentrated in vacuo. The residue was diluted with 5% NaOH and extracted with CH2C1 . The combined organic extracts were washed with brine, dried over MgS04, and concentrated. Flash chromatography on silica gel (10% MeOH/CH2Cl2) gave the title compound (0.83 g, 24%) as a slightly yellow viscous oil: MS (ES) m/e 173 (M + H)+. Preparation 27
Preparation of 2-(methylaminomethyl)benzofuran
a) N-Methylbenzofuran-2-carboxamide
To a solution of 2-benzofurancarboxylic acid (1.62 g,10 mmole), methylamine hydrochloride (0.79 g, l 1 mmole), triethylamine (3.1 mL, 22 mmole), and HOBt ■ H 0 (1.5 g,l 1 mmole) in DMF (30 mL) was added EDC (2.1 g,l 1 mmole). The reaction was stirred overnight then was concentrated in vacuo. The residue was diluted with 5% NaHCθ3 and extracted with CH2C12. The combined organic extracts were washed with brine, dried over MgSθ4, and concentrated. Flash chromatography on silica gel (3% MeOH/CH2Cl2) gave the title compound (1.75 g,100%) as white solid: MS (ES) m/e 176 (M + H)+.
b) 2-(Methylaminomethyl)benzofuran To a solution of 1.0 M BH3 THF (30 mL, 30 mmole) at 0 °C was added N- methylbenzofuran-2-carboxamide (1.75 g, 10 mmole). The reaction mixture was allowed to warm to RT, then was heated at reflux overnight. The reaction was cooled to 0 °C and excess methanol was added. The resulting solution was concentrated in vacuo and the residue was purified by flash chromatography on silica gel (3% MeOH/CH2Cl ). The tile compound (0.2 g ,12% ) was obtained as a white solid: MS (ES) m/e 162 (M + H)+.
Preparation 28
Preparation of l-methyl-2-(propylaminomethyl)-lH-indole
a) l-Methyl-N-cyclopropylindole-2-carboxamide
According to the procedure of Preparation 27 (a), except substituting 1 -methyl- 1H- indole-2-carboxylic acid (3.5 g, 20 mmole) for 2-benzofurancarboxylic acid, and substituting cyclopropylamine for methylamine hydrochloride, the title compound (2.1 g, 49%) was prepared as white solid: MS (ES) m/e 215 (M + Η)+.
b) l-Methyl-2-(propylaminomethyl)-lH-indole
To a solution of l-methyl-N-cyclopropylindole-2-carboxamide (2.1 g, 9.8 mmole) in dry TΗF (40 mL) was added dropwise a solution of 1.0 M L1AIΗ4 in THF (2.2 mL, 22 mmole). The reaction mixture was heated at reflux overnight, then was cooled and quenched with 10% NaOH. The mixture was filtered and the filtrate was concentrated in vacuo. Flash chromatography on silica gel (5% MeOH CH Cl2) gave the title compound (0.65 g, 33%) as a viscous oil: MS (ES) /e 203 (M + H)+.
Preparation 29
Preparation of 5-bromo-2-(methylamino)pyridine and 5-bromo-2-(dimethylamino)pyridine
a) 5-Bromo-2-(methylamino)pyridine and 5-bromo-2-(dimethylamino)pyridine
To a suspension of NaH (60% dispersion in mineral oil, 0.44 g, 11 mmole) in dry DMF (40 mL) was added solid 2-amino-5-bromopyridine (1.73 g, 10 mmole) in portions over 5-10 min. Gas evolution was allowed to subside between additions. The resulting amber mixture was stirred for 15 min, then methyl iodide (0.61 mL, 10 mmole) was added all at once. The reaction mixture was stirred at RT overnight, then was concentrated in vacuo. The residue was diluted with 5% NH4CI (30 mL) and the mixture was extracted with CH2C12. The combined organic extracts were washed with brine, dried (MgS04), and concentrated. Flash chromatography on silica gel (3% MeOH CH2Cl2) separated the products. 5-Bromo-2-(methylamino)pyridine (0.60 g, 32 %) was obtained as a semisolid: TLC (3% MeOH/CH2Cl2) Rf 0.35; MS (ES) m/e 187 (M + H)+. 5-Bromo-2- (dimethylamino)pyridine (0.70 g, 34%) was obtained as a semisolid: TLC (3% MeOH/CH2Cl2) Rf 0.77; MS (ES) m/e 201 (M + H)+.
Preparation 30
Preparation of (E)-3-16-(methylamino)pyridin-3-yllacrylic acid
a) Benzyl (E)-3-[6-methylamino)pyridin-3-yl)acrylate
According to the procedure of Preparation 2 (a), except substituting 5-bromo-2- (methylamino)pyridine for 2-amino-5-bromopyridine, the title compound (0.52 g, 60%) was prepared as a white solid: MS (ES) m/e 269 (M + H)+.
b) (E)-3-[6-(Methylamino)pyridin-3-yl]acrylic acid
According to the procedure of Preparation 2 (b), except substituting benzyl (E)-3- [6-(methylamino)pyridin-3-yl]acrylate for benzyl (E)-3-(6-aminopyridin-3-yl)acrylate, the title compound (0.15 g, 43%) was prepared as a white solid: MS (ES) m/e 179 (M + H)+. Preparation 31
Preparation of (EV3-r6-(dimethylamino)pyridin-3-yllacrylic acid
a) Benzyl (E)-3-[6-(dimethylamino)pyridin-3-yl]acrylate
According to the procedure of Preparation 2 (a), except substituting 5-bromo-2- (dimethylamino)pyridine for 2-amino-5-bromopyridine, the title compound (0.82 g, 84%) was prepared as a white solid: MS (ES) m/e 283 (M + H)+.
b) (E)-3-[6-(Dimethylamino)pyridin-3-yl]acrylic acid
According to the procedure of Preparation 2 (b), except substituting benzyl (E)-3- [6-(dimethylamino)pyridin-3-yl]acrylate for benzyl (E)-3-(6-aminopyridin-3-yl)acrylate, the title compound (0.20 g, 36%) was prepared as a white solid: MS (ES) m/e 193 (M + H)+.
Preparation 32
Preparation of (E -3-(6-methylpyridin-3-yl)acrylic acid
a) Benzyl (E)-3-(6-methylpyridin-3-yl)acrylic acid
According to the procedure of Preparation 2 (a), except substituting 5-bromo-2- methylpyridine for 2-amino-5-bromopyridine, the title compound (0.85 g, 34%) was prepared as a white solid: MS (ES) m/e 253 (M + H)+.
b) (E)-3-(6-Methylpyridin-3-yl)acrylic acid
According to the procedure of Preparation 2 (b), except substituting benzyl (E)-3- (6-methylpyridin-3-yl)acrylic acid for benzyl (E)-3-(6-aminopyridin-3-yl)acrylate, the title compound (0.18 g, 33%) was prepared as a white solid: MS (ES) m/e 164 (M + H)+.
Preparation 33
Preparation of 2-(methylaminomethyD-l/-'-indole
a) N-Methyl-lH-indol-2-carboxamide A suspension of ethyl indole-2-carboxylate (25.30 g, 133.7 mmole) in 40% aqueous
CΗ3NΗ (400 mL) was stirred at RT. The flask was tightly stoppered to keep the material inside the flask. As the reaction proceeded the product began to precipitate. The reaction was stirred at RT for 3 days, then was concentrated to remove approximately 200 mL of the solvent. The remaining residue was diluted with H 0 (500 mL), and the solid was collected by suction filtration and washed with H20. Drying under high vacuum left the title compound (21.50 g, 92%) as a light yellow solid: MS (ES) m/e 175 (M + H)+.
b) 2-(Methylaminomethyl)- lH-indole
A solution of LiAlΗ4 in THF (1.0 M, 250 mL, 250 mmole) was slowly added via syringe to a solution of N-methyl- lH-indol-2-carboxamide (21.50 g, 12.34 mmole) in anhydrous TΗF (100 mL). Gas was evolved during the addition of the first 50 mL of
L.AIΗ4 solution. When the addition was complete, the resulting light yellow solution was heated at gentle reflux. After 23 hr, the reaction was cooled in ice and quenched by the sequential dropwise addition of H20 (9.5 mL), 1.0 N NaOH (20 mL), and H20 (28.5 mL). The mixture was stirred for 15 min, then was filtered through celite®, and the filter pad was washed thoroughly with THF. The filtrate was concentrated and the residue was flash chromatographed on silica gel (10% MeOH/CHCl3 containing 0.5% cone. NH4OH). The title compound (10.10 g, 51%) was obtained as a light yellow oil: MS (ES) m/e 161 (M + H)+
Preparation 34
Preparation of l-ethyl-2-(methylaminomethyl)-lH-indole
a) 2-[N-(Benzyloxycarbonyl)-N-methylaminomethyl]- lH-indole N-(Benzyloxycarbonyloxy)succinimide (17.10 g, 68.6 mmole) was added to a solution of 2-(methylaminomethyl)-lH-indole (10.00 g, 62.4 mmole), from Preparation 33, and triethylamine (9.60 mL, 68.6 mmole) in DMF (100 mL) at RT. The reaction was stirred overnight then was concentrated in vacuo. The residue was diluted with water and the mixture was extracted with ethyl acetate. The combined extracts were dried over K2Cθ3 and concentrated. Flash chromatography on silica gel (20% ethyl acetate/hexanes) gave the title compound (14.80 g, 80%) as an off-white solid: MS (ES) m/e 295 (M + Η)+.
b) 2-[N-(Benzyloxycarbonyl)-N-methylaminomethyl]-l-ethyl-lH-indole
NaΗ (60% dispersion in mineral oil, 0.25 g, 7.1 mmole) was added portionwise. allowing for gas evolution, to a solution of 2-[N-(benzyloxycarbonyl)-N- methylaminomethylΗH-indole (1.40 g, 4.75 mmole) in DMF (35 mL) at 0 °C. When the NaH addition was complete, ethyl iodide (0.42 mL, 5.2 mmole) was added at 0 °C. The reaction was stirred at 0 °C for 15 minutes then at RT overnight. The reaction was diluted with water and extracted with ethyl acetate. The combined extracts were dried over K C03 and concentrated to afford the title compound (1.30 g, 87%) as an orange solid: MS (ES) m/e 323 (M + H)+.
e) 1 -Ethy l-2-(methy laminomethy 1)- 1 H-indole
2-[N-(Benzyloxycarbonyl)-N-methylaminomethyl]-l-ethyl-lH-indole (1.30 g, 4.0 mmole) was added to a suspension of Pearlman's catalyst (about 0.30 g) in MeOΗ at RT in a Parr flask. The reaction was placed under 50 p.s.i. of Η2 and shaken for 8 hr. The mixture was filtered through celite® and the filter pad was washed with MeOH. The filtrate was concentrated to afford the title compound (0.75 g, 100%) as a light yellow solid: MS (ES) m/e 189 (M + H)+.
Preparation 35
Preparation of l-methyl-3-(methylaminomethyl)-lH-indole (Method A)
a) Methyl l-methyl-lH-indole-3-carboxylate NaΗ (60% dispersion in mineral oil, 8.56 g, 214.0 mmole) was added portionwise, allowing for gas evolution, to a solution of methyl lH-indole-3-carboxylate (25.00 g, 142.7 mmole) in DMF (350 mL) at 0 °C. When the NaΗ addition was complete, methyl iodide (44.4 mL, 713.5 mmole) was added at 0 °C. The reaction was stirred at 0 °C for 15 minutes then at RT overnight. The reaction was diluted with water and extracted with ethyl acetate. The combined extracts were dried over K2C03 and concentrated to afford the title compound (26.00 g, 96%) as an orange solid: MS (ES) m/e 190 (M + Η)+.
b) N, 1 -Dimethyl- 1 H-indole-3-carboxamide
A suspension of methyl l-methyl-lH-indole-3-carboxylate (4.30 g, 22.74 mmole) in 40% aqueous CΗ3NΗ2 (400 mL) was stirred at RT. The flask was tightly stoppered to keep the material inside the flask. As the reaction proceeded the product began to precipitate. The reaction was stirred at RT for 3 days, then was concentrated to remove approximately 200 mL of the solvent. The remaining residue was diluted with H20 (500 mL), and the solid was collected by suction filtration and washed with H 0. Flash chromatography on silica gel (ethyl acetate) gave the title compound (2.4 g, 56%) as a white solid: MS (ES) m/e 189 (M + H)+. c) l-Methyl-3-(methylaminomethyl)- lH-indole
A solution of LiAlΗ4 in THF (1.0 M, 5.20 mL, 5.2 mmole) was slowly added via syringe to a solution of N,l -dimethyl- lH-indole-3-carboxamide (0.50 g, 2.6 mmole) in anhydrous TΗF (15 mL). Gas was evolved during the addition of the first 2 mL of LiAlΗ4 solution. When the addition was complete, the resulting light yellow solution was heated at gentle reflux. After 23 hr, the reaction was cooled in ice and quenched by the sequential dropwise addition of H20 (0.5 mL), 1.0 N NaOH (0.5 mL), and H20 (0.5 mL). The mixture was stirred for 15 min, then was filtered through celite®, and the filter pad was washed thoroughly with THF. The filtrate was concentrated and the residue was flash chromatographed on silica gel (10% MeOH/CHCl3 containing 0.5% cone. NH4OH) to afford the title compound (0.30 g, 67%) as a light yellow oil: MS (ES) m/e 175 (M + H)+.
Preparation 36
Preparation of l-methyl-3-(methylaminomethyl)-lH-indole (Method B)
To a solution of l-methylindole-3-carboxaldehyde (10.0 g, 62.8 mmole) in MeOΗ (100 mL) was added a solution of 2.0 M CΗ3NΗ2 in MeOH (126 mL, 252.0 mmole). The reaction was stirred at RT for 2 hrs, then was concentrated to a light yellow oil. This oil was dissolved in EtOH (300 mL), and NaBH4 (2.38 g, 62.8 mmole) was added. After 2 hrs the reaction was concentrated to a slurry and dissolved in 1.0 N NaOH (75 mL). The aqueous solution was extracted with Et 0 (2 x 200 mL) and the combined organic fractions were dried over Na2Sθ4 and concentrated. Flash chromatography on silica gel (9: 1 CHC^/MeOH containing 5% NH4OH) and drying in high vacuum left the title compound (10.1 g, 92%) as a faintly yellow oil: MS (ES) m/e 175 (M + H)+.
Preparation 37
Preparation of (E)-3-(6-aminopyridin-3-yl)-2-methylacrylic acid HCl salt and 2-(6- aminopyridin-3-ylmethyl)acrylic acid HCl salt
a) Ethyl (E)-3-(6-aminopyridin-3-yl)-2-methylacrylate and ethyl 2-(6-aminopyridin-3- ylmethyl)acrylate To a stirred solution of 2-amino-5-bromopyridine (25 g, 140 mmole) in propionitrile (150 mL) was added ethyl methacrylate (50 mL, 400 mmole), DIEA (50 mL, 287 mmole), palladium(II) acetate (1.57 g, 7 mmole), and tri-ø-tolylphosphine (4.3 g, 14 mmole). The reaction was purged with argon and heated at reflux for 6 hr, then was cooled to RT and concentrated to dryness under vacuum. The residue was taken up in 80% ethyl acetate/hexanes (100 mL), and the solution was filtered through a pad of silica gel, eluting with 80% ethyl acetate/hexanes (400 mL) until all the product was eluted off. The yellowish filtrate was concentrated under vacuum, and the residue was taken up in a small volume of 1 : 1 Et20/petroleum ether. The precipitate which formed was collected and dried under vacuum to give ethyl (E)-3-(6-aminopyridin-3-yl)-2-methylacrylate (10.77 g, 37%) as a pale yellow solid: LCMS (ES) m/e 207.0 (M + H)+; !H NMR (300 MHz, CDC13) δ 8.05 (d, J = 1.7 Hz, 1 H), 7.63 (dd, 1 H), 7.48 (s, 1 H), 6.75 (d, J = 8.8 Hz, 1 H), 5.79 (br s, 2 H), 4.26 (q, 2 H), 2.10 (s, 3 H), 1.34 (t, 3 H). The filtrate was concentrated to dryness and purified by flash chromatography on silica gel (4: 1 ethyl acetate/hexanes) to give additional ethyl (E)-3-(6-aminopyridin-3-yl)-2-methylacrylate (0.87 g, 3% ) and ethyl 2-(6- aminopyridin-3-ylmethyl)acrylate (5.77 g, 20%) as a yellow oil: LCMS (ES) /e 207.0 (M + H)+; 1H NMR (300 MHz, CDCI3) δ 7.86 (d, J = 2.1 Hz, 1 H), 7.32 (dd, 1 H), 6.53 (d, J = 8.5 Hz, 1 H), 6.21 (d, J = 1.8 Hz, 1 H), 5.48 (d, J = 1.4 Hz, 1 H), 4.17 (q, 2 H), 3.47 (s, 2 H), 1.27 (t, 3 H).
b) (E)-3-(6-Aminopyridin-3-yl)-2-methylacrylic acid HCl salt To ethyl (E)-3-(6-aminopyridin-3-yl)-2-methylacrylate (5.0 g, 24.2 mmole) was added HOAc (25 mL) and cone. HCl (25 mL). The reaction was stirred and heated at 100 °C for 6 hr, cooled to RT and concentrated to dryness. The remaining residue was triturated with Et 0, filtered and dried under vacuum to give the title compound (5.5 g, quantitative) as a white solid: LCMS (ES) m/e 179.0 (M + H)+; lH NMR (300 MHz, DMSO-d6) δ 8.47 (br s, 2 H), 8.16 (d, J = 1.7 Hz, 1 H), 8.08 (dd, 1 H), 7.42 (s, 1 H), 7.08 (d, J = 9.3 Hz, 1 H), 2.01 (s, 3 H).
c) 2-(6-Aminopyridin-3-ylmethyl)acrylic acid HCl salt
According to the procedure of Preparation 37 (b), except substituting ethyl 2-(6- aminopyridin-3-ylmethyl)acrylate (3.1 g, 15 mmole) for ethyl (E)-3-(6-aminopyridin-3-yl)- 2-methylacrylate gave the title compound (3.0 g, 93%) as a white solid: LCMS (ES) m/e 179.0 (M + H)+; 1H NMR (300 MHz, DMSO-d6) δ 8.10 (br s, 2 H), 7.79 (dd, 1 H), 7.78 (s, 1 H), 7.00 (d, J = 9.7 Hz, 1 H), 6.15 (d, J = 1.2 Hz, 1 H), 5.67 (d, J = 1.2 Hz, 1 H), 3.45 (s, 2 H). Preparation 38
Preparation of 2-(methylaminomethyl)naphthalene
To a stirred solution of 40 wt% methylamine in H20 (50 mL, 581 mmole) in THF
(50 mL) at 0 °C was added 2-(bromomethyl)naphthalene (10 g, 43 mmole) in one portion. The reaction was allowed to warm to RT and stirred for 16 hr, then was then concentrated under vacuum. The residue was taken up in Et20 and washed with 1.0 N NaOH then with brine, dried (Na2Sθ4), and concentrated to dryness. Purification by flash chromatography on silica gel (98:2 to 9: 1 CHC^/methanol containing 5% NH4OH) gave the title compound (3.95 g, 54%) as a clear oil: lH NMR (400 MHz, CDCI3) δ 7.85 (m, 3 H), 7.79 (s, 1 H), 7.49 (m, 3 H), 3.94 (s, 2 H), 2.53 (s, 3 H).
Preparation 39
Preparation of (E)-3-(6-amino-4-methylpyridin-3-yl)acrylic acid HCl salt
a) 2-Amino-5-bromo-4-methylpyridine
To a stirred solution of 2-amino-4-methylpyridine (22 g, 203 mmole) in 48% HBr (200 mL) at 70 °C was added dropwise a solution of 15% H 02 in H20 (60 mL) over 60 minutes. The reaction became slightly exothermic and the oil bath was removed after 15 minutes. The reaction stirred for an additional 1 hr, then was poured into ice (approximately 500 mL). The clear solution was adjusted to pH 4-5 with solid Na2C03 (80 g, 755 mmole), and the resulting thick white suspension was filtered. The filter pad was washed with a small volume of H20 and pressed dry. Drying under high vacuum gave a 2:3 mixture of 2-amino-5-bromo-4-methylpyridine and 2-amino-3,5-dibromo-4- methylpyridine (27.08 g). Flash chromatography on silica gel (50% ethyl acetate/hexanes then ethyl acetate) gave the title compound ( 12.1 1 g, 32%) as a white solid: LCMS (ES) m/e 187.2 (M + H)+; 1H NMR (400 MHz, DMSO-d6) δ 7.92 (s, 1 H), 6.41 (s, 1 H), 6.03 (br s, 2 H), 2.17 (s, 3 H).
b) Ethyl (E)-3-(6-amino-4-methylpyridin-3-yl)acrylate
To a stirred solution of 2-amino-5-bromo-4-methylpyridine (10 g, 54 mmole) in propionitrile (50 mL) was added ethyl acrylate (17 mL, 157 mmole), DIEA (19 mL, 106 mmole), palladium(II) acetate (0.61 g, 2.7 mmole) and tri-ø-tolylphosphine (1.64 g, 5.4 mmole). The reaction was purged with argon and heated at reflux for 6 hr, then was cooled to RT and concentrated to dryness under vacuum. The resulting residue was taken up in ethyl acetate and filtered through a pad of silica gel. The filtrate was concentrated and the remaining residue was triturated with 1 : 1 Et 0/petroleum ether (50 mL), filtered, and dried under vacuum to give the title compound (6.50 g, 59%) as a pale yellow solid: ^H NMR (400 MHz, DMSO-d6) δ 8.31 (s, 1 H), 7.66 (d, J = 16.0 Hz, 1 H), 6.40 (br s, 2 H), 6.32 (d, J = 16.0 Hz, 1 H), 6.28 (s, 1 H), 4.15 (q, 2 H), 2.24 (s, 3 H), 1.24 (t, 3 H).
c) (E)-3-(6-Amino-4-methylpyridin-3-yl)acrylic acid HCl salt
To ethyl (E)-3-(6-amino-4-methylpyridin-3-yl)acrylate (1.50 g, 7.3 mmole) was added HO Ac (15 mL) and cone. HCl (15 mL). The solution was stirred at 100 °C for 10 hr, cooled to RT, and concentrated to dryness. Trituration with Et20, filtration and drying under vacuum gave the title compound (1.65 g, quantitative) as a white solid: LCMS (ES) m/e 179.2 (M + H)+; 1H NMR (400 MHz, DMSO-d6) δ 8.37 (s, 1 H), 8.28 (br s, 3 H), 7.51 (d, J = 16.0 Hz, 1 H), 6.86 (s, 1 H), 6.46 (d, J = 16.0 Hz, 1 H), 2.41 (s, 3 H).
Preparation 40
Preparation of 1.3-dimethyl-2-(methylaminomethyl)-lH-indole
a) 1,3-Dimethyl-lH-indole
To a stirred solution of 3-methylindole (15.0 g, 114 mmole) in dry DMF (200 mL) was added NaΗ (60% dispersion in oil, 5.0 g, 125 mmole) in portions. Gas evolution was observed. The mixture was stirred for 30 min, then iodomethane (8 mL, 129 mmole) was added in one portion. The reaction became exothermic and was cooled in an ice bath. After 16 hr at RT, the reaction was concentrated under vacuum and the residue was taken up in ethyl acetate. The solution was washed with Η20 then with brine, dried (MgSθ4), and concentrated to dryness. Purification by short path distillation under vacuum (bp 88- 92°C, 0.5 mmHg) gave the title compound (16.10 g, 97%) as a pale yellow oil: lH NMR (400 MHz, CDC13) δ 7.47 (d, J = 7.9 Hz, 1 H), 7.35 (d, J = 8.2 Hz, 1 H), 7.13 (t, 1 H), 7.06 (s, 1 H), 7.00 (t, 1 H), 3.71 (s, 3 H), 2.24 (s, 3 H).
b) 1 ,3-Dimethyl- 1 H-indole-2-carboxaldehyde
To a stirred solution of phosphorus oxychloride (7.0 mL, 75 mmole) in DMF (25 mL) was added dropwise a solution of 1 ,3-dimethylindole (12.0 g, 83 mmole) in dry DMF (6.0 mL). The reaction was stirred at RT for 2 hr then was poured onto ice. The mixture was basified with a solution of NaOΗ (13.2 g, 330 mmole) in Η20 (44 mL), then was extracted with Et20 (2x 50 mL). The combined organic layers were washed with brine, dried (MgSθ4), and concentrated under vacuum. Flash chromatography on silica gel (10% ethyl acetate/hexanes) gave the title compound (13.03 g, 91%) as an off-white solid: LCMS (ES) /e 174.2 (M + H)+; lH NMR (400 MHz, CDC13) δ 10.16 (s, 1 H), 7.68 (d, J = 8.1 Hz, 1 H), 7.42 (t, 1 H), 7.32 (d, J = 8.5 Hz, 1 H), 7.15 (t, 1 H), 4.04 (s, 3 H), 2.63 (s, 3 H).
c) 1 ,3-Dimethyl-2-(methylarninomethyl)- lH-indole
To l,3-dimethyl-lH-indole-2-carboxaldehyde (13.0 g, 75 mmole) was added a solution of 2.0 M methylamine in methanol (150 mL, 300 mmole) and ΗOAc (4.3 mL, 75 mmole). The solution was stirred at RT for 4 hr, then was cooled to 0 °C, and sodium cyanoborohydride (5.0 g, 80 mmole) was added portionwise over 5 min. The reaction was then allowed to warm to RT. After 16 hr, the reaction was concentrated under vacuum and the residue was taken up in Et20. The solution was washed with 1.0 N NaOΗ then with brine, dried (Na2Sθ4), and concentrated to dryness. Flash chromatography on silica gel (95:5 CΗCl3/methanol containing 5% NH4OH) gave the title compound (7.34 g, 52%) as a yellow oil: lH NMR (400 MHz, CDCI3) δ 7.53 (d, J = 7.8 Hz, 1 H), 7.26 (d, J = 7.8 Hz, 1 H), 7.20 (t, 1 H), 7.09 (t, 1 H), 3.88 (s, 2 H), 3.76 (s, 3 H), 2.46 (s, 3 H), 2.32 (s, 3 H), 1.36 (br s, 1 H).
Preparation 41
Preparation of 6-bromo-2-oxo-1.4-dihvdro-2H-pyridor2.3-rfl-1.3-oxazine
a) 2-Amino-3-(hydroxymethyl)pyridine
To a stirred solution of 2-aminonicotinic acid (20 g, 145 mmole) in dry TΗF (200 mL) under argon was added 1.0 M LiAlΗ4 in THF (300 mL, 300 mmole) carefully, portionwise, through a reflux condenser, over 4 hr. The reaction became exothermic and refluxed without external heating. After the addition was complete, the reaction was heated at reflux for an additional 16 hr, then was cooled to 0 °C and carefully quenched by sequential addition of H20 (12 mL), 15% NaOH in H 0 (12 mL), and H20 (35 mL). The resulting thick suspension was stirred for 1 hr, then was filtered through a pad of celite®. The filter pad was rinsed with THF (300 mL), and the filtrate was concentrated to dryness to give the title compound (17.04 g, 95%) as a pale yellow waxy solid: LCMS (ES) m/e 125.1 (M + H)+; 1H NMR (400 MHz, DMSO-d6) δ 7.84 (dd, 1 H), 7.37 (m, 1 H), 6.53 (dd, 1 H), 5.65 (br s, 2 H), 5.16 (t, 1 H), 4.34 (d, J = 4.6 Hz, 2 H). b) 2-Amino-5-bromo-3-(hydroxymethyl)pyridine
To a stirred solution of 2-amino-3-(hydroxymethyl)pyridine (15.0 g, 121 mmole) in HO Ac (300 mL) at RT was added bromine (6.2 mL, 121 mmole) dropwise over 1 hr. A suspension formed after approximately 15 min. After the addition, the reaction was stirred for an additional 1 hr, then was concentrated under vacuum. The residue was taken up in 1.0 M Na2Cθ3 (500 mL), and the solution was extracted with ethyl acetate (2 x 250 mL). The combined organic layers were washed with brine, dried (Na2Sθ4), and concentrated to dryness. The resulting residue was triturated with a small volume of petroleum ether, filtered and dried under vacuum to give the title compound (18.45 g, 75%) as a beige solid: LCMS (ES) /e 203.2 (M + H)+; lH NMR (400 MHz, DMSO-d6) δ 7.89 (d, J = 2.3 Hz, 1 H), 7.52 (s, 1 H), 5.92 (br s, 2 H), 5.29 (br s, 1 H), 4.30 (s, 2 H).
c) 6-Bromo-2-oxo- 1 ,4-dihydro-2H-pyrido[2,3--i]- 1 ,3-oxazine To a stirred solution of 2-amino-5-bromo-3-(hydroxymethyl)pyridine (3.0 g, 15 mmole) in methanol (30 mL) was added dimethyl carbonate (5 mL, 60 mmole) and sodium methoxide (25 wt% solution in methanol, 4 mL, 17.4 mmole). The reaction was heated at reflux for 18 hr, cooled to RT, and concentrated to dryness. The remaining residue was triturated with saturated aqueous NΗ4CI (50 mL), filtered, washed with cold H20 (50 mL), and dried under vacuum to give the title compound (1.75 g, 51%) as a beige solid: LCMS (ES) m/e 229.0 (M + H)+; lH NMR (400 MHz, DMSO-d6) δ 10.90 (s, 1 H), 8.31 (s, 1 H), 7.90 (s, I H), 5.31 (s, 2 H).
Preparation 42
Preparation of 3-methyl-2-(methylaminomethyl)benzorplthiophene
To a stirred solution of 3-rnethylbenzo[σ]thiophene-2-carboxaldehyde (0.5 g, 2.8 mmole) in methanol (15 mL) was added 2.0 M methylamine in methanol (6 mL, 12 mmole) and HOAc (0.32 mL, 5.7 mmole). The reaction was stirred at RT for hr, then sodium cyanoborohydride (0.2 g, 3 mmole) was added in one portion. After stirring for an additional 16 hr the reaction was concentrated to dryness. The residue was taken up in Et20 and washed with 1.0 N NaOH then with brine, dried (Na2Sθ4), and concentrated under vacuum. Purification by flash chromatography on silica gel (95:5 CHC^/methanol containing 5% NH4OH) gave the title compound (0.30 g, 56%) as a yellow oil: lU NMR (400 MHz, CDCI3) δ 7.77 (d, J = 7.8 Hz, 1 H), 7.62 (d, J = 7.9 Hz, 1 H), 7.34 (t, 1 H), 7.28 (t, 1 H), 3.99 (s, 2 H), 2.49 (s, 3 H), 2.34 (s, 3 H), 1.77 (br s, 1 H).
Preparation 43
Preparation of 2-(methylaminomethyl)benzothiophene
a) N-methyl benzothiophene-2-carboxamide
To a stirred solution of 2.0 M methylamine in THF (60 mL) and THF (60 mL) was added dropwise at 0 °C a solution of benzothiophene-2-carbonyl chloride (10.8 g, 55 mmole) in THF (50 mL) over 15 minutes. After the addition the reaction was allowed to warm to RT then was concentrated under vacuum. Trituration with a cold solution of 4: 1 H20/methanol (50 L), filtration, and drying under vacuum gave the title compound (10.35 g, 98%) as a white solid: MS (ES) m/e 191.9 (M + H)+.
b) 2-(Methylaminomethyl)benzothiophene
To a stirred suspension of N-methyl benzothiophene-2-carboxamide (10.0 g, 52 mmole) in dry THF (75 mL) under argon was added a solution of 1.0 M LiAlH4 in THF (135 mL, 135 mmole) over 15 minutes. The reaction quickly became clear and was heated at reflux for 2 days. After cooling to 0 °C the reaction was carefully quenched with the sequential addition of H20 (5.1 mL), 15% NaOH in H20 (5.1 mL), and H20 (15.3 mL). The mixture was filtered through a pad of celite® and the filter pad was rinsed with Et20 (50 mL). The filtrate was concentrated to afford the title compound (9.11 g, 99%) as a pale yellow oil which solidified in the freezer: 1H NMR (400 MHz, CDCI3) δ 7.83 (d, J = 7.3 Hz, 1 H), 7.72 (d, J = 7.3 Hz, 1 H), 7.33 (m, 2 H), 7.17 (s, 1 H), 4.06 (s, 2 H), 2.53 (s, 3 H), 1.56 (br s, I H).
Preparation 44
Preparation of 2-methyl-3-(methylaminomethyl)indole
To a solution of 2-methylindole-3-carboxaldehyde (10.00 g, 62.84 mmole) in MeOH (100 mL) was added 2 M CH3NH2 in MeOH (200 mL). After stirring for 3 hours at RT, the reaction solution was concentrated to a yellow oil which solidified under vacuum. This solid was dissolved in ethanol (350 mL) and NaBH4 (2.38 g, 62.8 mmole) was added. The reaction was stirred at RT for 6 hours, then was concentrated under vacuum. The remaining residue was diluted with saturated aqueous Na C03 (50 mL) and extracted with EtOAc (2 x 200 mL). The organic phase was separated, washed with brine, and dried over Na2Sθ4. Flash chromatography on silica gel (9:1 CHC^ MeOH containing 5% NH4OH) and drying under high vacuum gave the title compound (6.88 g, 63%) as a faintly yellow viscous solid: MS (ES) m/e 175 (M + H)+.
Preparation 45
Preparation of 5-bromo-2H-pyridol3.2-ol-1.4-oxazin-3(4H)-one
To a solution of 2#-pyrido[3,2-ø]-l,4-oxazin-3(4H)-one (5.00 g, 33.3 mmole) in ΗOAc (100 mL) was added Br2 (2.6 mL, 50.0 mmole). After stirring for 48 hours at RT, the reaction solution was concentrated to an orange solid, which was suspended in 1 N NaOΗ (50 mL) and extracted with EtOAc (2 x 100 mL). The combined organic layers were washed with brine and dried over Na S04- Flash chromatography on silica gel (9: 1 CΗC^/MeOΗ containing 5% NH4OH) and drying under high vacuum gave the title compound (5.49 g, 72%) as a yellow solid: MS (ES) m/e 230 (M + H)+.
Preparation 46
Preparation of 5-bromo-2-acetylaminopyrimidine
To a solution of 5-bromor2-aminopyrimidine (2.0 g, 11.5 mmole) in CH2C12 (75 mL) at RT was added 2,6-lutidine (2.7 mL, 23.0 mmole) followed by acetyl chloride (0.99 g, 12.6 mmole). After stirring for 8 hours, the reaction solution was concentrated under vacuum. The remaining residue was dissolved EtOAc (200 mL), washed with H 0 (100 mL) and brine, and dried over Na Sθ4- Flash chromatography on silica gel (95:5 CHC^/MeOH) and drying under high vacuum gave the title compound (1.74 g, 70%) as a yellow solid: MS (ES) m/e 217 (M + H)+. Preparation 47
Preparation of l-methyl-2-(methylamino methyll-6-methoxy-lH-indole
a) Methyl- l-methyl-6-methoxy-lH-indole-2-carboxy late
According to the procedure of Preparation 1 (a), except substituting methyl-6- methoxyindole-2-carboxylate for ethyl indole-2-carboxylate, the title compound (90%) was prepared as a tan solid: MS (ES) m/e 220.2 (M + Η)+.
b) N,l-Dimethyl-6-methoxy-lH-indole-2-carboxamide
According to the procedure of Preparation 1 (b), except substituting methyl- 1-methy 1-6- methoxy-lH-indole-2-carboxylate for ethyl- l-methyl-lH-indole-2-carboxylate, the title compound (95%) was prepared as an off-white solid: MS (ES) m/e 219.2 (M + Η)+ and 437.4 (2M + H)+.
c) l-Methyl-2-(methylamino methyl)-6-methoxy-lH-indole
According to the procedure of Preparation 1 (c), except substituting N,l-dimethyl- 6-methoxy-lH-indole-2-carboxamide for N,l -dimethyl- lH-indole-2-carboxamide, the title compound (76%) was prepared as a light gray solid: MS (ES) m/e 205.2 (M + Η)+, 409.4 (2M + H)+.
Preparation 48
Preparation of 1.7-dimethyl-3-(methylaminomethyl)-lH-indole
a) 1,7-Dimethyl-lH-indole
According to the procedure of Preparation 1 (a), except substituting 7-methylindole for ethyl indole-2-carboxylate, the title compound (89%) was prepared as a tan solid: MS (ES) m/e 146.2 (M + Η)+.
b) 1 ,7-Dimethy 1- 1 H-indole-3-carboxaldehyde
According to the procedure of Preparation 40 (b), except substituting 1,7-dimethyI- lH-indole for 1,3-dimethylindole, the title compound (82%) was prepared as a light tan solid: MS (ES) m/e 174.2 (M + Η)+. c) l,7-Dimethyl-3-(methylaminomethyl)-l//-indole
According to the procedure of Preparation 40 (c), except substituting 1,7-dimethyl- l /-indole-3-carboxaldehyde for l,3-dimethyl-l/ -indole-l-carboxaldehyde, the title compound (98%) was prepared as a white, crystalline solid: MS (ES) m/e 189.2 (M + H)+.
Preparation 49
Preparation of 1.5-dimethyl-3-(methylaminomethyl)-ltf-indole
a) 1,5-Dimethyl-l -indole
According to the method of Preparation 1 (a), except substituting 5-methylindole for ethyl indole-2-carboxylate, the title compound (92%) was prepared as an amber oil: MS (ES) m/e 146.2 (M + H)+.
b) l,5-Dimethyl-l /-indole-3-carboxaldehyde
According to the procedure of Preparation 40 (b), except substituting 1, 5 -dimethyl- \H- indole for 1 ,3-dimethylindole, the title compound (82%) was prepared as a light tan solid: MS (ES) m/e 174.2 (M + H)+.
c) l,5-Dimethyl-3-(methylaminomethyl)-lH-indole
According to the procedure of Preparation 36, except substituting 1,5-dimethyl-lH- indole-3-carboxaldehyde for 1,3-dimethyl-lH-indole-l-carboxaldehyde, the title compound (89%) was prepared as an oil: MS (ES) m/e 189.2 (M + Η)+.
Preparation 50
Preparation of 1.6-dimethyl-3-(methylaminomethyl)-lH-indole
a) 1,6-Dimethy 1-1 -indole According to the procedure of Preparation 1 (a), except substituting 5-methylindole for ethyl indole-2-carboxylate, the title compound (96%) was prepared as an amber oil: MS (ES) m/e 146.2 (M + Η)+. b) 1 ,6-Dimethyl- 1 / -indole-3-carboxaldehyde
According to the procedure of Preparation 40 (b), except substituting 1,5-dimethyl-l//- indole for 1,3-dimethylindole, the title compound (99%) was prepared as a light tan solid: MS (ES) /e 174.2 (M + H)+.
c) l,6-Dimethyl-3-(methylaminomethyl)-l -indole
According to the procedure of Preparation 36, except substituting 1,5-dimethyl-l//- indole-3-carboxaldehyde for l,3-dimethyl-l /-indole-l-carboxaldehyde, the title compound (95%) was prepared as an oil: MS (ES) /e 189.2 (M + H)+.
Preparation 51
Preparation of l-benzyl-3-(methylaminomethyl)-l/ -indole
a) 3-(Methylaminomethyl)-lH-indole
To a solution of indole-3-carboxaldehyde (5.4 g, 34.1 mmole) in MeOΗ (30 mL) was added a solution of 2.0 M CΗ3NΗ in MeOH (51.3 mL, 102.6 mmole). The reaction was stirred at RT overnight, then was concentrated to a light yellow oil. This oil was dissolved in EtOH (40 mL), and N-1BH4 (1.3 g, 34.1 mmole) was added. After 16 hrs the reaction was concentrated to a slurry and dissolved in 10% Na2Cθ3(100 mL). The aqueous solution was extracted with EtOAc (2 x 200 mL) and the combined organic fractions were dried over Na2Sθ4 and concentrated. Drying in high vacuum left the title compound (5.2 g, 94%) as a faintly yellow oil: MS (ES) m e 161 (M + H)+.
b) 3-[N-(Benzy.oxycarbonyl)-N-methylam.nomethyl]-lH-indole
N-(Benzyloxycarbonyloxy)succinimide (8.9 g, 35.7 mmole) was added to a solution of 3-(methylaminomethyl)-l /- indole (5.2 g, 32.5 mmole) and triethylamine (5.0 mL, 65.7 mmole) in DMF (100 mL) at RT. The reaction was stirred overnight then was concentrated in vacuo. The residue was diluted with water and the mixture was extracted with ethyl acetate. The combined extracts were dried over Na2S04 and concentrated. Flash chromatography on silica gel (33% ethyl acetate/hexanes) gave the title compound (7.0 g, 74%) as an off-white solid: MS (ES) m/e 295 (M + Η)+.
c) 3-[N-(Benzyloxycarbonyl)-N-methylaminomethyl}- 1-benzyl- lH-indole NaΗ (60% dispersion in mineral oil, 0.15 g, 3.8 mmole) was added portionwise, allowing for gas evolution, to a solution of 3-[N-(benzyloxycarbonyl)-N- methylaminomethylH -indole (0.7 g, 2.5 mmole) in DMF (25 mL) at 0 °C. When the NaH addition was complete, benzyl bromide (1.2 mL, 10.0 mmole) was added at 0 °C. The reaction was stirred at 0 °C for 15 minutes then at RT overnight. The reaction was diluted with water and extracted with ethyl acetate. The combined extracts were dried over Na2S04 and concentrated. Flash chromatography on silica gel (33% ethyl acetate/hexanes) gave the title compound (0.9 g, 93%) as an off white solid: MS (ES) m/e 385 (M + H)+.
d) l-Benzyl-3-(methylaminomethyl)- lH-indole
3-[N-(Benzyloxycarbonyl)-N-methylaminomethyl]-l-benzyl-lH-indole (0.9 g, 2.3 mmole) was added to a suspension of Pearlman's catalyst (about 0.30 g) in MeOΗ at RT in a Parr flask. The reaction was placed under 50 p.s.i. of Η and shaken for 5 hr. The mixture was filtered through celite® and the filter pad was washed with MeOH. The filtrate was concentrated to afford the title compound (0.5 g, 86%) as a light yellow solid: MS (ES) m/e 251 (M + H)+.
Preparation 52
Preparation of 2-phenylamino-3-bromopyridine
A mixture of 2,5-dibromopyridine (10.2 g, 43 mmole) in aniline (25 mL) was stirred and heated at reflux for 3 h. The reaction was cooled to RT and most of the aniline was distilled off under vacuum. The remaining residue was taken up in ethyl acetate and the solution was washed with 1.0 N Na C03 then with brine, dried (Na2Sθ4), and concentrated under vacuum. Trituration with petroleum ether, filtration and drying under vacuum gave the title compound (7.20 g, 67%) as a tan solid: 1H NMR (400 MHz, CDCI3) δ 8.25 (d, J = 2.4 Hz, 1 H), 7.58 (dd, 1 H), 7.31-7.39 (m, 4 H), 7.11 (m, 1 H), 6.79 (br s, 1 H); MS (ES) m/e 249.0 (M + H)+
Preparation 53
Preparation of 1.2-dimethyl-3-(methylaminomethy.VlH-indole
a) 1 ,2-Dimethylindole-3-carboxaldehyde
A solution of POCI3 (7.0 mL, 75 mmole) in DMF (100 mL) was stirred for 5 minutes at 0 °C, then 1, 2-dimethylindole (10.0 g, 69 mmole) was added in one portion. The reaction was allowed to warm to RT and stirred for 4 h. The thick slurry was poured into ice water (300 mL) and the flask was rinsed with additional water (50 mL). The aqueous mixture was basified with a solution of NaOH (13.2 g, 330 mmole) in H20 (50 mL), and the thick suspension was filtered to collect the solid. This was washed with water and dried under vacuum to give the title compound (11.59 g, 97%) as an off-white solid: ! H NMR (400 MHz, CDC13) δ 10.07 (s, 1 H), 8.09 (d, J = 7.9 Hz, 1 H), 7.54 (d, J = 7.6 Hz, 1 H), 7.21 (dt, 2 H), 3.73 (s, 3 H), 2.70 (s, 3 H).
b) l,2-Dimethyl-3-(methyliminornethyl)-lH-indole
To l,2-dimethylindole-3-carboxaldehyde (11.50 g, 66.4 mmole) was added a solution of 2 M methylamine in methanol (100 mL, 200 mmole). The reaction was stirred for 4 h at RT then was concentrated to dryness to afford the crude title compound: ^Η NMR (400 MHz, CDCI3) δ 8.55 (d, J = 1.4 Hz, 1 H), 8.16 (d, J = 7.5 Hz, 1 H), 7.42 (d, J = 7.8 Hz, 1 H), 7.15 (t, 1 H), 7.07 (t, 1 H), 3.68 (s, 3 H), 3.41 (s, 3 H), 2.55 (s, 3 H).
c) l,2-Dimethyl-3-(methylaminomethyl)-lH-indole l,2-Dimethyl-3-(methyliminomethyl)-lH-indole was taken up in ethanol (200 mL) and NaBΗ4 (2.6 g, 68.7 mmole) was added portionwise with stirring at RT (vigorous gas evolution). After 16 h the reaction was concentrated under vacuum, and the residue was basified with aqueous 1.0 N NaOH (200 mL). The mixture was extracted with Et20 (250 mL), and the combined Et20 extracts were washed with brine, dried (Na2Sθ4) and concentrated. Purification by flash chromatography on silica gel (5-10% (5% NH4θH MeOH)/CHCl3) gave the title compound (8.47 g, 68%) as an oil which solidified in the freezer: *H NMR (400 MHz, CDCI3) δ 7.60 (d, J = 7.7 Hz, 1 H), 7.29 (d, J = 8.0 Hz, 1 H), 7.19 (t, 1 H), 7.12 (t, 1 H), 3:93 (s, 2 H), 3.69 (s, 3 H), 2.49 (s, 3 H), 2.45 (s, 3 H).
Preparation 54
Preparation of 3-(methylaminomethyl)benzo[/?]thiophene
To a stirred solution of 2 M methylamine in methanol (75 mmole, 150 mmole) was added benzo[ø]thiophen-3-carboxaldehyde (5.3 g, 33 mmole) and HOAc (4.3 mL, 75 mmole). The reaction was stirred at RT for 1 h, then NaB^CN (2.1 g, 33 mmole) was added portionwise over 5 minutes. The reaction was stirred for an additional 16 h then was concentrated under vacuum. The residue was taken up in Et20 (300 mL) and washed with 1.0 N NaOH (300 mL) then with brine, dried (Na2S04), and concentrated. Purification by flash chromatography on silica gel (5% (5% NH4θH/MeOH)/CHCl3) gave the title compound (2.81 g, 48%) as a brownish oil: lH NMR (400 MHz, CDCI3) δ»7.87 (2d, 2 H), 7.40 (m, 2 H), 7.32 (s, 1 H), 4.02 (s, 2 H), 2.56 (s, 3 H), 1.5 (br s, 1 H).
Preparation 55
Preparation of 5-bromo-2.2'-dipyridylamine
Bromine (3.0 mL, 58.2 mmole) was added dropwise over 15 minutes to a stirred solution of 2,2'-dipyridylamine (10 g, 58.4 mmole) in HOAc (100 mL). The reaction quickly became a thick suspension.. After 2 h the reaction was concentrated under vacuum and the residue was purified by flash chromatography on silica gel (0.5% (5% NH4θH/MeOH)/CHCl3). The resulting residue was triturated with hexane and dried under vacuum gave the title product (1.77 g, 12%) as an off-white solid: 1H NMR (400 MHz, CDCI3) δ 9.88 (s, 1 H), 8.31 (s, 1 H), 8.23 (d, J = 4.8 Hz, 1 H), 7.83 ( , 2 H), 7.67 (t, 1 H), 7.62 (d, J = 8.4 Hz, 1 H), 6.90 (t, 1 H); MS (ES) m/e 250.0 (M + H)+. 5,5'-dibromo-2,2'- dipyridylamine (4.04 g, 21%) was also isolated as a white solid after trituration with hexane and drying under vacuum: !H NMR (400 MHz, CDCI3) δ 10.08 (s, 1 H), 8.32 (d, J = 2.5 Hz, 2 H), 7.88 (dd, 2 H), 7.68 (d, J = 9.0 Hz, 2 H); MS (ES) m/e 328.0 (M + H)+.
Preparation 56
Preparation of 2-(methylaminomethyl)-3-methylbenzorolthiophene
According to the procedures of Preparation 53 (b) and (c), except substituting 3- methylbenzo[6]thiophene-2-carboxaldehyde (7.40 g, 42 mmole) for l,2-dimethylindole-3- carboxaldehyde, the title compound (6.02 g, 75%) was prepared as a pale yellow oil which solidified in the freezer: 1H NMR (400 MHz, CDCI3) δ 7.77 (d, J = 7.8 Hz, 1 H), 7.62 (d, J = 7.9 Hz, 1 H), 7.34 (t, 1 H), 7.28 (t, 1 H), 3.99 (s, 2 H), 2.49 (s, 3 H), 2.34 (s, 3 H), 1.77 (br s, 1 H).
Preparation 57
Preparation of 2-methyl-3-(methylaminomethyπbenzorfclthiophene
a) 2-Methylbenzo[o]thiophene-3-carboxaldehyde
SnC-4 (20 mL, 67 mmole) was added over 5 min to a stirred solution of 2- methylbenzo[i>]thiophene (5.0 g, 33.7 mmole) in CH2C12 (75 mL) at 0 °C under argon. After 15 minutes, dichloromethyl methyl ether (3.7 mL, 41 mmole) was added. The reaction became a yellowish colored suspension. The reaction was allowed to warm to RT and stirred for 16 h, then was poured onto ice water (200 mL). The aqueous mixture was acidified with 1.0 N HCl (100 mL) and stirred until the suspension dissolved. The organic phase was separated, dried (MgS04), and concentrated under vacuum. Purification by flash chromatography on silica gel (10% ethyl acetate/hexane) gave the title compound (5.83 g, 98%) as a white crystalline solid: 1H NMR (400 MHz, CDCI3) δ 10.38 (s, 1 H), 8.61 (d, J = 8.1 Hz, 1 H), 7.77 (d, J = 8.0 Hz, 1 H), 7.48 (t, 1 H), 7.39 (t, 1 H), 2.93 (s, 3 H).
b) 2-Methyl-3-(methylaminomethyl)benzo[-?]thiophene
According to the procedures of Preparation 53 (b) and (c), except substituting 2- methylbenzo[Z>]thiophene-3-carboxaldehyde (5.0 g, 28.4 mmole) for l,2-dimethylindole-3- carboxaldehyde, the title compound (4.89 g, 90%) was prepared as an oil which solidified in the freezer: 1H NMR (400 MHz, CDCI3) δ 7.78 (d, J = 7.9 Hz, 1 H), 7.75 (d, J = 7.9 Hz, 1 H), 7.37 (t, 1 H), 7.29 (t, 1 H), 3.95 (s, 2 H), 2.60 (s, 3 H), 2.50 (s, 3 H).
Preparation 58
Preparation of 3.4-dimethyl-2-(methylaminomethyl)thienof2.3-blthiophene
According to the procedure of Preparation 24 (a), except substituting 3,4- dimethylthieno[2,3-b]thiophene-2-carboxaldehyde (0.5 g, 2.5 mmole) for the 1- methylindole-2-carboxaldehyde, the title compound (0.28 g, 53%) was prepared as a colorless oil: MS (ES) m/e 212 (M + H)+. Preparation 59
Preparation of l-methyl-2-(methylaminomethyDnaphthalene
a) N, l-Dimethylnaphthalene-2-carboxamide
According to the procedure of Preparation 20 (a), except substituting 1- methylnaphthalene-2-carboxylic acid (J. Org. Chem. 1965, 22, 3869; 0.3 g, 1.6 mmole) for the 3-methyl-2-inden-2-carboxylic acid, the title compound (0.3 g, 94%) was prepared as a white solid: MS (ES) m/e 200 (M + H)+.
b) 1 -Methy l-2-(methylaminomethyl)naphthalene
According to the procedure of Preparation 20 (b), except substituting N,l- dimethylnaphthalene-2-carboxamide (0.3 g, 1.5 mmole) for the N,3-dimethylindene-2- carboxamide, the title compound (0.1 g, 36%) was prepared as a colorless oil: MS (ES) m/e 186 (M + H)+.
Preparation 60
Preparation of l-methyl-3-(methylaminomethyl)- lH-pyrrolof2,3-blpyridine
a) 1 -Methyl- 1 H-pyrrolo[2,3-b]pyridine
According to the procedure of Preparation 40 (a), except substituting 7-azaindole (2.28 g, 1.83 mmole) for the 3-methylindole, the title compound (1.4 g, 58%) was prepared as a yellow oil: MS (ES) m/e 133 (M + H)+.
b) 1 -Methyl- lH-pyrrolo[2,3-b]pyridine-3-carboxa.dehyde
According to the procedure of Preparation 40 (b), except substituting 1 -methyl- 1/ - pyrrolo[2,3-b]pyridine (0.7 g, 5.3 mmole) for the 1 ,3-dimethylindole, the title compound (0.4 g, 47%) was prepared as a white solid: MS (ES) m/e 161 (M + Η)+.
c) 1 -Methy l-3-(methylaminomethyl)- l/ -pyrrolo[2,3-fc]pyridine
According to the procedure of Preparation 40 (c), except substituting 1-methyl-l//- pyrrolo[2,3-b]pyridine-3-carboxaldehyde (0.4 g, 2.5 mmole) for the 1,3-dimethyl-l//- indole-2-carboxaldehyde, the title compound (0.2 g, 45%) was prepared as a yellow oil: MS (ES) m/e 176 (M + H)+. Preparation 61
Preparation of 2.3-dihydro-8-(methylaminomethyl')-lH-3a-azacvclopental lindene
a) 2,3-Dihydro-lH3a-azacyclopenta[ ]indene-8-carboxaldehyde
According to the procedure of Preparation 40 (b), except substituting 2,3-dihydro- lH-3a-azacyclopenta[-.]indene (J. Med. Chem. 1965, 8, 700; 0.24 g, 1.53 mmole) for the 1,3-dimethylindole, the title compound (0.17 g, 60%) was prepared as a yellow solid: MS (ES) m/e 186 (M + Η)+.
b) 2,3-Dihydro-8-(methylaminomethyl)-lH-3a-azacyclopenta[α]indene
According to the procedure of Preparation 40 (c), except substituting 2,3-dihydro- lH-3a-azacyclopenta[α]indene-8-carboxaldehyde (0.17 g, 0.92 mmole) for the 1,3- dimethyl-lH-indole-2-carboxaldehyde, the title compound (0.1 g, 54%) was prepared as a yellow oil: MS (ES) m/e 201 (M + Η)+.
The following examples illustrate methods for preparing the biologically active compounds of this invention from intermediate compounds such as those described in the foregoing Preparations.
Example 1
Preparation of (E)-3-(6-aminopyridin-3-yl)-N-methyl-N-( 1 -methyl- lH-indol-2- ylmethyDacrylamide
EDC (0.70 g, 3.7 mmole) was added to a solution of (E)-3-(6-aminopyridin-3- yl)acrylic acid (0.61 g, 3.7 mmole), l-methyl-2-(methylaminomethyl)-lH-indole (0.65 g, 3.7 mmole), ΗOBt Η20 (0.50 g, 3.7 mmole), and triethylamine (0.52 mL, 3.7 mmole) in DMF (30 mL) at RT. The reaction was stirred overnight, then was concentrated in vacuo. The residue was diluted with 5% NaHC03 and extracted with CH2C12. The combined organic extracts were washed with brine and dried over MgS04. Flash chromatography on silica gel (3% MeOH/CH Cl2) gave a colorless semisolid which was triturated with Et20 and dried. The title compound (1.0 g, 83%) was obtained as a white solid: ^H NMR (300 MHz, CDC13) δ 8.20 (br s, 1 H), 7.45 - 7.70 (m, 3 H), 7.00 - 7.30 (m, 3 H), 6.69 (d, J = 15.4 Hz, 1 H), 6.30 - 6.50 (m, 2 H), 4.89 (s, 2 H), 4.67 (br s, 2 H), 3.68 (s, 3 H), 3.01 (s, 3 H); MS (ES) /e 321 (M + H)+. Anal. Calcd for C19H20N4O 0.40 H20: C, 69.66; H, 6.40; N, 17.10. Found: C, 69.99; H, 6.27; N, 16.84.
Example 2
Preparation of (E)-3-(4-aminophenyl)-N-methy l-N-( 1 -methyl- 1 / -indol-2- ylmethyDacrylamide
EDC (218 mg, 1.14 mmole) was added to a solution of 4-aminocinnamic acid hydrochloride (220 mg, 1.10 mmole), l-methyl-2-(methylaminomethy.)-lH-indole (0.20 g, 1.15 mmole), ΗOBt • Η 0 (154 mg, 1.14 mmole), and triethylamine (0.20 mL, 1.43 mmole) in DMF (20 mL) at RT. The reaction was stirred overnight, then was concentrated in vacuo. The residue was diluted with 5% NaHCθ3 and extracted with CH2C1 . The combined organic extracts were washed with brine (2 x 30 mL) and dried over MgSθ4. Flash chromatography on silica gel (3% MeOH/CH2Cl2) gave the title compound (68 mg, 19%) as a yellow foam: 1H NMR (360 MHz, DMSO-d6, 330K) δ 7.46 (d, J = 7.8 Hz, 1 H), 7.42 (d, J = 15.3 Hz, 1 H), 7.37 (d, J = 8.3 Hz, 1 H), 7.32 (d, J = 8.5 Hz, 2 H), 7.06 - 7.15 (m, 1 H), 6.94 - 7.03 (m, 1 H), 6.81 (d, J = 15.3 Hz, 1 H), 6.58 (d, J = 8.5 Hz, 2 H), 6.33 (s, 1 H), 5.25 (br s, 2 H), 4.85 (s, 2 H), 3.70 (s, 3 H), 3.02 (s, 3 H); MS (ES) m/e 320 (M + H)+. Anal. Calcd for C20H21N3O • 0.20 H20: C, 74.37; H, 6.68; N, 13.01. Found: C, 74.21; H, 6.60; N, 12.80.
Example 3
Preparation of (E)-N-Methyl-N-(l-methyl-l//-indol-2-ylmethyl)-3-(pyridin-3- yPacrylamide
EDC (0.22 g, 1.14 mmole) was added to a solution of trans-3-(3-pyridyl)acrylic acid (0.17 g, 1.14 mmole), l-methyl-2-(methylaminomethyl)-lH-indole (0.20 g, 1.15 mmole), and ΗOBt • Η20 (0.15 g, 1.11 mmole) in DMF (10 mL) at RT. The reaction was stirred overnight, then was concentrated in vacuo. The residue was diluted with 5% NaHCθ3 and extracted with CH2C12. The combined organic extracts were washed with brine and dried over MgSθ4. Flash chromatography on silica gel (3% MeOH/CH2Cl2) followed by preparative TLC (3% MeOH/CH2Cl2) gave the title compound (0.14 g, 40%) as a white solid: ^H NMR (360 MHz, CDCI3) indicated an approximately 8: 1 mixture of amide rotamers; for the major rotamer: δ 8.79 (s, 1 H), 8.59 (d, J = 3.9 Hz, 1 H), 7.84 (d, J = 7.6 Hz, 1 H), 7.76 (d, J = 15.5 Hz, 1 H), 7.59 (d, J = 7.8 Hz, 1 H), 7.38 - 7.48 (m, 2 H), 7.19 - 7.27 (m, 1 H), 7.08 - 7.17 (m, 1 H), 6.98 (d, J = 15.5 Hz, 1 H), 6.51 (s, 1 H), 4.94 (s, 2 H), 3.73 (s, 3 H), 3.09 (s, 3 H); MS (ES) m/e 306 (M + H)+. Anal. Calcd for C19H19N30 • 0.20 H20: C, 73.86; H, 6.33; N, 13.60. Found: C, 73.52; H, 6.32; N, 13.43.
Example 4
Preparation of (E)-3-(6-aminopyridin-3-yl)-N-methyl-N-(l-methyl-lH-indazol-3- ylmethyPacrylamide
a) (E)-3-(6-Aminopyridin-3-yl)-N-methyl-N-( 1 -methyl- 1 H-indazol-3-ylmethy l)acrylamide
EDC (230 mg, 1.2 mmole) was added to a solution (E)-3-(6-aminopyridin-3- yl)acrylic acid (164 mg, 1.0 mmole), l-methyl-3-(methylaminomethyl)-lH-indazole (210 mg, 1.2 mmole), ΗOBt • Η20 (162 mg, 1.2 mmole), and Et3N (0.28 mL, 2.0 mmole) in dry DMF (5 mL) at RT. After 18 hr the mixture was concentrated. Flash chromatography on silica gel (5% EtOH/EtOAc) gave the title compound (238 mg, 74%) as a white foam: ^H NMR (400 MHz, CDC13) δ 8.24 (m, 1 H), 7.90 (m, 1 H), 7.65 (m, 2 H), 7.35 (m, 2 H), 7.09 (m, 1 H), 6.73 (m, 1 H), 6.50 (m, 1 H), 5.04 (s, 2 H), 4.83 (bs, 2 H), 4.04 (s, 3 H), 3.10 (s, 3 H); MS (ES) m/e 322 (M + H)+.
Example 5
Preparation of (E)-3-(3.4-dihvdro-2H-pyridor3.2--7l-1.4-oxazin-7-yl)-N-methyl-N-(l- methyl- lH-indol-2-ylmethyl)acrylamide
a) (E)-3-(3,4-Dihydro-2H-pyrido[3,2-Z>][ 1 ,4]oxazin-7-yl)-N-methyl-N-(l -methyl- 1H- indol-2-ylmethyl)acrylamide
EDC (230 mg, 1.2 mmole) was added to a solution (E)-3-(3,4-dihydro-2H- pyrido[3,2-Z?]-l,4-oxazin-7-yl)acrylic acid (206 mg, 1.0 mmole), l-methyl-2- (methylaminomethyl)-lH-indole (209 mg, 1.2 mmole), ΗOBt • Η20 (162 mg, 1.2 mmole), and E-3N (0.21 mL, 1.5 mmole) in dry DMF (5 mL) at RT. After 18 hr the mixture was concentrated. Flash chromatography on silica gel (5% EtOH/EtOAc) gave the title compound (238 mg, 66%) as a yellow solid: lH NMR (400 MHz, d6-DMSO) δ 7.99-6.95 (m, 8 H), 6.40 (s, 1 H), 4.82 (s, 2 H), 4.11 (bs, 2 H), 3.72 (bs, 3 H), 3.67 (bs, 2 H), 3.08 (s, 3 H); for minor rotomer δ 6.15 (s, 1 H), 5.02 (s, 2 H), 2.96 (s, 3 H); MS (ES) m/e 363 (M + H)+. Example 6
Preparation of (EVN-methyl-N-[(l-methyl-l -indol-2-ylmethvPl-3-(5.6.7.8-tetrahydro- 1.8-naphthyridin-3-vPacrylamide
a) (E)-N-Methyl-N-[(l-methyl-lH-indol-2-ylmethyl)]-3-(5,6,7,8-tetrahydro-l,8- naphthyridin-3-yl)acrylamide
EDC (203 mg, 1.06 mmole) was added to a solution (E)-3-(5,6,7,8-tetrahydro-l,8- naphthyridin-3-yl)acrylic acid (180 mg, 0.88 mmole), l-methyl-2-(methylaminomethyl)- lH-indole (185 mg, 1.06 mmole), ΗOBt • Η 0 (143 mg, 1.06 mmole), and Et3N (0.31 mL, 2.2 mmole) in dry DMF (5 mL) at RT. After 18 hr the mixture was concentrated. Flash chromatography on silica gel (10% EtOH/EtOAc) gave the title compound (222 mg, 70%) as a yellow solid: !H NMR (400 MHz, DMSO-d6) δ 7.99-6.82 (m, 8 H), 6.40 (s, 1 H), 4.82 (s, 2 H), 3.67 (m, 2 H), 3.29 (m, 3 H), 3.07 (m, 3 H), 2.73 ( , 2 H), 1.77 (m, 2 H); for minor rotomer δ 6.16 (s, 1 H), 5.00 (s, 2 H); MS (ES) m/e 361 (M + H)+.
Example 7
Preparation of (E)-3-(6-aminopyridin-3-yP-N-methyl-N-(thieno[2.3-fclthiophen-2- ylmethypacrylamide
a) (E)-3-(6-Aminopyridin-3-yl)-N-methyl-N-(thieno[2,3-fc]thiophen-2- ylmethyl)acrylamide EDC (230 mg, 1.2 mmole) was added to a solution (E)-3-(6-aminopyridin-3- yl)acrylic acid (164 mg, 1.0 mmole), 2-(methylaminomethyl)thieno[2,3-i>]thiophene (220 mg, 1.2 mmole), HOBt • H20 (162 mg, 1.2 mmole), and Et3N (0.35 mL, 2.5 mmole) in dry DMF (5 mL) at RT. After 18 hr the mixture was concentrated. Flash chromatography on silica gel (5% EtOH/EtOAc) gave the title compound (138 mg, 42%) as a tan solid: 1H NMR (400 MHz, d6-DMSO) δ 8.15 (d, J = 2.0 Hz, 1 H), 7.84 (bs, 1 H), 7.57 (d, J = 5.2 Hz, 1 H), 7.43 (d, J = 15.2 Hz, 1 H), 7.27 (m, 2 H), 6.44 (m, 2 H), 4.75 (s, 2 H), 3.13 (s, 3 H); for minor rotomer δ 5.00 (s, 2 H), 2.95 (s, 3 H); MS (ES) m/e 330 (M + H)+. Example 8
Preparation of (EV3-(6-aminopyridin-3-yP-N-methyl-N-(thienor3.2-felthiophen-2- ylmethvDacrylamide
a) (E)-3-(6-Aminopyridin-3-yl)-N-methyl-N-(thieno[3,2-6]thiophen-2- ylmethyl)acrylamide
EDC (230 mg, 1.2 mmole) was added to a solution (E)-3-(6-aminopyridin-3- yl)acrylic acid (164 mg, 1.0 mmole), 2-(methylaminomethyl)thieno[3,2-fc]thiophene (220 mg, 1.2 mmole), HOBt • H20 (162 mg, 1.2 mmole), and Et3N (0.35 mL, 2.5 mmole) in dry
DMF (5 mL) at RT. After 18 hr the mixture was diluted with H20 and extracted with
EtOAc (3x). The combined organic layers were dried (MgSθ4) and concentrated. The solid was taken up in 1: 1 MeOH/H20 and filtered. The filtrate was concentrated to approximately 1/3 volume. The precipitate was collected by filtration, washed with H20, and dried in vacuo to afford the title compound (139 mg, 42%) as a light tan solid: 1H
NMR (400 MHz, d6-DMSO) δ 8.15 (d, J = 2.0 Hz, 1 H), 7.83 (bd, 1 H), 7.61 (d, J = 5.2 Hz,
1 H), 7.40 (m, 3 H), 6.45 (m, 2 H), 4.75 (s, 2 H), 3.13 (s, 3 H); for minor rotomer δ 5.00 (s,
2 H), 2.95 (s, 3 H); MS (ES) m/e 330 (M + H)+.
Example 9
Preparation of (E)-3-(3H-imidazor4,5-frlpyridin-6-vP-N-methyl-N-(l-methyl-lH-indol-2- ylmethyPacrylamide
a) (E)-3-(3H-Imidazo[4,5-*>]pyridin-6-y l)-N-methy l-N-( 1 -methy 1- 1 H-indol-2- ylmethyl)acrylamide
EDC (230 mg, 1.2 mmole) was added to a solution (E)-3-(3H-imidazo[4,5- Z?]pyridin-6-yl)acrylic acid (189 mg, 1.0 mmole), l-methyl-2-(methylaminomethyl)-lH- indole (209 mg, 1.2 mmole), ΗOBt Η20 (162 mg, 1.2 mmole), and E-3N (0.28 mL, 2.0 mmole) in dry DMF (5 mL) at RT. After 18 hr the mixture was diluted with H20. The title compound (193 mg, 56%) was collected as a white solid by filtration, washed with H20, and dried in vacuo: lH NMR (400 MHz, d6-DMSO) δ 8.72 (s, 1 H), 8.50 (s, 2 H), 7.68 (d, J = 15.4 Hz, 1 H), 7.45 (m, 3 H), 7.13 (m, 1 H), 7.01 (m, 1 H), 6.43 (s, 1 H), 4.87 (s, 2 H), 3.70 (s, 3 H), 3.15 (s, 3 H); for minor rotomer δ 8.68 (s, 1 H), 8.47 (s, 2 H), 6.19 (s, 1 H), 5.10 (s, 2 H), 3.74 (s, 3 H), 3.01 (s, 3 H); MS (ES) m/e 346 (M + H)+. Example 10
Preparation of (E)-3-(6-aminoρyridin-3-yP-N-methyl-N-(6-methyl-6//-thienor2.3-fclpyrrol-
5-ylmethvPacrylamide
a) (E)-3-(6-Aminopyridin-3-yl)-N-methyl-N-(6-methyl-6H-thieno[2,3-/j]pyrrol-5- ylmethyPacrylamide
EDC (132 mg, 0.69 mmole) was added to a solution (E)-3-(6-aminopyridin-3- yPacrylic acid (95 mg, 0.58 mmole), 6-methyl-5-(methylaminomethyl)-6H-thieno[2,3- ^pyrrole (142 mg, 0.69 mmole), ΗOBt • Η20 (93 mg, 0.69 mmole), and Et3N (0.16 mL,
1.16 mmole) in dry DMF (3 mL) at RT. After 18 hr the mixture was diluted with H 0 and extracted with EtOAc (3x). The combined organic layers were dried (MgSθ4) and concentrated. The residue was taken up in MeOH and collected by filtration to give the title compound (65 mg, 34%) as a yellow solid: !H NMR (400 MHz, d6-DMSO) δ 8.15 (s, 1 H), 7.81 (d, J = 8.1 Hz, 1 H), 7.43 (d, J = 15.2 Hz, 1 H), 6.96 (m, 2 H), 6.43 (m, 3 H), 4.70
(s, 2 H), 3.61 (s, 3 H), 3.00 (s, 3 H); for minor rotomer δ 4.87 (s, 2 H), 2.90 (s, 3 H); MS
(ES) m/e 321 (M + H)+.
Example 11
Preparation of (E)-3-(2-aminopyrimidin-5- vP-N-methy l-N-( 1 -methyl- 1 H-indol-2- ylmethyPacrylamide
According to the procedure of Example 1, except substituting (E)-3-(2- aminopyrimidin-5-yl)acrylic acid (0.50 g, 3.0 mmole) for (E)-3-(6-aminopyridin-3- yl)acrylic acid, the title compound (0.86 g, 89%) was prepared as an off-white solid: MS (ES) m/e 322 (M + Η)+.
Example 12
Preparation of (E)-3-(6-aminopyridin-3-vP-N-(benzor/?lthiophen-2-ylmethvP-N- methylacrylamide
According to the procedure of Example 1, except substituting 2- (methylaminomethyl)benzo[Z?]thiophene (0.47 g, 2.68 mmole) for l-methyl-2- (methylaminomethyl)indole, the title compound (0.71 g, 91%) was prepared as an off-white solid: MS (ES) m/e 324 (M + H)+.
Example 13
Preparation of (E)-3-(6-aminopyridin-3-yl)-N-methyl-N-(l-methyl-lH-indol-2-ylmethyl)- 2-butenamide
According to the procedure of Example 1, except substituting (E)-3-(6- aminopyridin-3-yl)-2-methylacrylic acid (0.40 g, 2.24 mmole) for (E)-3-(6-aminopyridin-3- yl)acrylic acid, the title compound (0.65 g, 87%) was prepared as an off-white solid: MS (ES) m/e 335 (M + Η)+.
Example 14
Preparation of (E)-3-(6-amino-2- methy lpyridin-3-y P-N- methy l-N-( 1 -methyl- 1 /-indol-2- ylmethyDacrylamide
According to the procedure of Example 1, except substituting (E)-3-(6-amino-2- methylpyridin-3-yl)acrylic acid (0.40 g, 2.24 mmole) for (E)-3-(6-aminopyridin-3- ypacrylic acid, the title compound (0.70 g, 94%) was prepared as an off-white solid: MS (ES) m/e 335 (M + H)+.
Example 15
Preparation of (E)-3-(6-amino-5-methylpyridin-3-yl)-N-methyl-N-(l-methyl-lH-indol-2- ylmethvDacrylamide
According to the procedure of Example 1, except substituting (E)-3-(6-amino-5- methylpyridin-3-yl)acrylic acid (1.00 g, 5.62 mmole) for (E)-3-(6-aminopyridin-3- yPacrylic acid, the title compound (1.78 g, 95%) was prepared as an off-white solid: MS (ES) m/e 335 (M + Η)+. Example 16
Preparation of (E)-3-r6-acetylamino)pyridin-3-yll-N-methyl-N-(l-methyl-l//-indol-2- ylmethyPacrylamide
To a stirred suspension of (E)-3-(6-aminopyridin-3-yl)-N-methyl-N-[(l-rnethyl- l//-indol-2-yl)methyl]acrylamide (0.50 g, 1.56 mmole) and NaHC03 (0.51 g, 6.09 mmole) in THF (75 mL) was added acetic anhydride (0.38 g, 3.74 mmole). The reaction was heated at reflux for 24 hrs and then concentrated. The remaining residue was extracted with EtOAc and purified on silica gel (95:5 CHCI3/CH3OH) to give the title compound (0.54 g, 96 %) as an off-white solid: MS (ES) /e 363 (M + H)+.
Example 17
Preparation of (E)-3-(6-amino-5-methylpyridin-3-vP-N-(benzoffclthiophen-2-ylmethyl)-N- methylacrylamide
According to the procedure of Example 1 , except substituting (E)-3-(6-amino-5- methylpyridin-3-yl)acrylic acid (0.40 g, 2.24 mmole) for (E)-3-(6-aminopyridin-3- yl)acrylic acid, and substituting 2-(methylaminomethyl)benzo[Z?]thiophene (0.44 g, 2.47 mmole) for l-methyl-2-(methylaminomethyl)indole, the title compound (0.69 g, 91%) was prepared as an off-white solid: MS (ES) m/e 338 (M + H)+.
Example 18
Preparation of (E)-3-(6-amino-5-methylpyridin-3-yl)-N-methyl-N-(naphthalen-2- ylmefhyDacrylam.de
According to the procedure of Example 1, except substituting (E)-3-(6-amino-5- methylpyridin-3-yl)acrylic acid (0.40 g, 2.24 mmole) for (E)-3-(6-aminopyridin-3- yPacrylic acid, and substituting 2-(methylaminomethyl)naphthalene (0.42 g, 2.47 mmole) for l-methyl-2-(methylaminomethyl)indole, the title compound (0.65 g, 87%) was prepared as an off-white solid: MS (ES) m/e 332 (M + H)+. Example 19
Preparation of (E)-3-f6-acetylamino-5-methylpyridin-3-vP-N-methyl-N-(l-methyl-l - indol-2-ylmethvPacrylamide
According to the procedure of Example 16, except substituting (E)-3-(6-amino-5- methylpyridin-3-yl)-N-methyl-N-(l-methyl-lH-indol-2-ylmethyl)acrylamide (0.47 g, 1.4 mmole) for (E)-3-(6-aminopyridin-3-yl)-N-methyl-N-[(l-methyl-l//-indol-2- yl)methyl]acrylamide, the title compound (0.49 g, 93%) was prepared as an off-white solid: MS (ES) m/e 377 (M + Η)+.
Example 20
Preparation of (E)-3-16-amino-5-(hydroxymethyPpyridin-3-yll-N-methyl-N-(l-methyl-l//- indol-2-ylmethyl)acrylamide
According to the procedure of Example 1, except substituting (E)-3-[6-amino-5- (hydroxymethyl)pyridin-3-yl]acrylic acid (0.40 g, 2.1 mmole) for (E)-3-(6-am.nopyridin-3- yl)acrylic acid, the title compound (0.56 g, 77%) was prepared as an off-white solid: MS (ES) m/e 351 (M + H)+.
Example 21
Preparation of (EVN-methyl-N-( 1 -methyl- l//-indol-2-ylmethvP-3-(7-oxo-5.6.7.8- tetrahydro- 1 ,8-naphthyridin-3-y Pacrylamide
a) N-Methyl-N-(1 -methyl- l//-indol-2-ylmethyl)acrylam.de
To a solution of l-methyl-2-(methylaminomethyl)indole (0.78 g, 4,5 mmole), from Preparation 1, and triethylamine (1.4 mL, 10.0 mmole) in CH C12 (50 mL) at 5 °C was added acryloyl chloride (0.41 mL, 4.95 mmole). After 45 min, the reaction solution was poured onto H 0 and the layers were separated. The organic phase was dried over Na Sθ4 and concentrated to afford the title compound as a yellow oil. This was used directly without further purification. b) (E)-N-Methyl-N-(l-methyl-lH-indol-2-ylmethyl)-3-(7-oxo-5,6,7,8-tetrahydro-l,8- naphthyridin-3-yl)acrylamide
According to the procedure of Preparation 2 (a), except substituting N-methyl-N- (1 -methyl- lH-indol-2-ylmethyl)acrylamide (0.90 g, 3.96 mmole) for benzyl acrylate, and substituting 6-bromo-3,4-dihydro-lH-l,8-naphthyridin-2-one (0.60 g, 2.64 mmole) for 2- amino-5-bromopyridine, the title compound (0.85 g, 86%) was prepared as an off-white solid: MS (ES) m/e 375 (M + Η)+.
Example 22
Preparation of (E)-3-f6-amino-5-r(2-hydroxyethylamino)carbonyllpyridin-3-yll-N-(l- methyl-l -indol-2-ylmethyP-N-methylacrylamide
According to the procedure of Example 1, except substituting (E)-3-[6-amino-5- [(2-hydroxyethylamino)carbonyl]pyridin-3-yl]acrylic acid (1.35 g, 5.4 mmole) for (E)-3-(6- aminopyridin-3-yl)acrylic acid, the title compound ( 1.95 g, 89%) was prepared as an off- white solid: MS (ES) m/e 408 (M + H)+.
Example 23
Preparation of (E)-N-methyl-N-(l -methyl- lH-indol-2-ylmethyl)-3-(3-methyl-2-oxo- 1.2.3.4-tetrahydropyridof2.3-dlpyrimidin-6-yl)acrylamide
a) N-Methyl-N-(l-methyl-lH-indol-2-ylmethyl)acrylamide To a solution of l-methyl-2-(methylaminomethyl)indole (0.96 g, 5.5 mmole), from
Preparation 1, and triethylamine (1.54 mL, 11.0 mmole) in CΗ2C12 (50 mL) at 5 °C was added acryloyl chloride (0.48 mL, 6.0 mmole). After 45 min, the reaction solution was poured onto H20 and the layers were separated. The organic phase was dried over Na2S04 and concentrated to afford the title compound as a yellow oil. This was used directly without further purification.
b) (E)-N-Methyl-N-(l-methyl-lH-indol-2-ylmethyl)-3-(3-methyl-2-oxo-l,2,3,4- tetrahydropyrido[2,3-d]pyrimidin-6-yl)acrylamide
According to the procedure of Preparation 2 (a), except substituting N-methyl-N- (l-methyl-lH-indol-2-ylmethyl)acrylamide (1.25 g, 5.5 mmole) for benzyl acrylate, and substituting 6-bromo-3-methyl-3,4-dihydro-lH-pyrido[2,3--i]pyrimidin-2-one (0.80 g, 3.3 mmole) for 2-amino-5-bromopyridine, the title compound (0.62 g, 49%) was prepared as an off-white solid: MS (ES) m/e 390 (M + H)+.
Example 24
Preparation of (E)-3-(6-aminopyridin-3-vP-N-methyl-N-(4-methyl-4H-theinol3.2-felpyrrol- 5-ylmethyPacrylamide
According to the procedure of Example of 1 , except substituting 4-methyl-5- (methylaminomethyl)-4H-thieno[3,2-/?]pyrrole (0.60 g, 3.3 mmole) for l-methyl-2-
(methylaminomethyPindole, the title compound (0.90 g, 92%) was prepared as an off-white solid: MS (ES) m/e 327 (M + Η)+.
Example 26
Preparation of (E)-3-r6-aminopyridin-3-yl1-N-methyl-N-(3-methyl-lH-inden-2- ylmethyDacrylamide
EDC (0.383 g, 2.0 mmole) was added to a solution of (E)-3-(6-aminopyridin-3- yPacrylic acid (0.328 g, 2.0 mmole), 3-methyl-2-(methylaminomethyl)indene hydrochloride (0.420 g, 2.0 mmole), ΗOBt • Η20 (0.306 g, 2.0 mmole), and triethylamine (0.57 mL, 4.0 mmole) in anhydrous DMF (18 mL) at RT. The reaction was stirred overnight and concentrated in vacuo. The residue was diluted with 5% NaHCθ3 and extracted with CH2C12. The combined organic extracts were washed with brine and dried over MgSθ4. Flash chromatography on silica gel (3% MeOH/ CH2C12) gave the title compound (0.33 g, 52%) as a colorless solid: MS (ES) m/e 320.2 (M + H)+. Anal. Calcd for C20H21N O • 0.4 H20: C, 73.57; H, 6.72; N, 12.86. Found: C, 73.94; H, 6.92; N, 12.50.
Example 27
Preparation of (E)-3-f6-aminopyridin-3-yll-N-(lH-inden-2-ylmethyl)-N-methylacrylamide
According to the procedure in Example 26, except substituting 2- (methylaminomethyPindene hydrochloride for 3-methyl-2-(methylaminomethyl)indene hydrochloride, the title compound (0.23 g, 38%) was obtained as an off-white solid: MS (ES) m/e 306.2 (M + H)+. Anal. Calcd for C199N30 • 0.125 H20: C, 74.18; H, 6.30; N, 13.64. Found: C.74.21; H, 6.25; N, 13.27.
Example 28
Preparation of (E)-3-(6-aminopyridin-3-vP-N-(4-methoxy-l-methyl- lH-indol-2-ylmethvP- N-methylacrylamide
According to the procedure in Example 26, except substituting 4-methoxy-l- methyl-2-(methylaminomethyl)-lH-indole hydrochloride for 3-methyl-2-
(methylaminomethyl)indene hydrochloride, the title compound (0.115 g, 68%) was obtained as an off white solid: MS (ES) m/e 351.2 (M + Η)+. Anal. Calcd for C20H22N4O2: C, 68.55; H, 6.32; N, 15.98. Found: C, 68.15; H, 6.33; N, 15.73.
Example 29
Preparation of (E)-3-16-(acetylamino)pyridin-3-yll-N-methyl-N-(3-methyl-lH-inden-2- ylmethyPacrylamide
To a solution of (E)-3-[6-aminopyridin-3-yl]-N-methyl-N-(3-methyl-lH-inden-2- ylmethyl)acrylamide (0.159 g, 0.5 mmole), from Example 26, in anhydrous TΗF (20 mL) was added NaΗC03 (0.126 g, 1.5 mmole) followed by acetic anhydride (0.153 g, 0.15 mmole). The mixture was heated at reflux for 40 hr, then was concentrated under vacuum. The residue was partitioned between H20 and EtOAc, and the organic layer was dried over MgSθ4, filtered and concentrated. The residue was triturated with diethyl ether to give the title compound (0.135 g, 74.8%) as an off-white solid: MS (ES) m/e 362.2 (M + H)+. Anal. Calcd for C22H23N302 0.25 H20: C, 72.20; H, 6.47; N, 11.47. Found: C, 72.42; H, 6.45; N, 11.07.
Example 30
Preparation of (EV3-(6-aminopyridin-3-yl)-N-(1.4-dimethyl-lH-indol-2-ylmethvP-N- methylacrylamide
According to the procedure in Example 26, except substituting l,4-dimethyl-2- (methylaminomethyl)-lH-indole hydrochloride for 3-methyl-2-(methylaminomethyl)- indene hydrochloride, the title compound (0.088 g, 52.7%) was obtained as an off white solid: MS (ES) m/e 335.2 (M + Η)+. Anal. Calcd for C20H22N4O 0.125 H20: C, 71.35; H, 6.66; N, 16.64. Found: C, 71.23; H, 6.65; N, 16.67.
Example 31
Preparation of (E)-N-methyl-N-(3-methyl- lH-inden-2-ylmethyl)-3-(7-oxo-5.6.7.8- tetrahydro- 1.8-naphthyridin-3-yPacry lamide
a) N-Methyl-N-(3-methyl- lH-inden-2-ylmethyl)-acrylamide
To a solution of 3-methyl-2-(methylaminomethyl)indene hydrochloride (0.132 g, 0.63 mmole), from Preparation 19, and triethylamine (0.19 g, 1.89 mmole) in CΗ2C12 (6 mL) at 0 °C was added a solution of acryloyl chloride ( 0.06 mL, 0. 7 mmole) in CH2C1 (2 mL). The reaction was stirred at 0 °C for 1 hr, then was poured into water. The layers were separated, and the organic layer was washed with brine, dried over Mg SO4 and concentrated in vacuo to yield the title compound (0.145 g, quantitative) as an oily solid: MS (ES) m/e 228.2 (M + H)+.
b) (E)-N-Methyl-N-(3-methyl-lH-inden-2-ylmethyl)-3-(7-oxo-5,6,7,8-tetrahydro-l,8- naphthyridin-3-yl)acry lamide
A mixture of 6-bromo-3,4-dihydro- lH- l ,8-naphthyridin-2-one (0.096 g, 0.42 mmole), from Preparation 15, and N-methyl-N-(3-methyl-lH-inden-2-ylmethyl)acrylamide (0.141 g, 0.62 mmole) in propionitrile (10 mL) was treated with (j-Pr)2NEt (0.15 mL, 0.08 mmole), palladium acetate (0.014 g, 0.062 mmole), and (o-tolyl^P (0.025 g, 0.08 mmole), and the resulting mixture was heated at gentle reflux. After 18 hr, the reaction was cooled, filtered through celite®, and concentrated. Flash chromatography on silica gel (2% MeOΗ/CΗ Cl2) gave the title compound (0.06 g, 41%) as a glassy solid: MS (ES) m/e 374.2 (M + H)+. Anal. Calcd for C23H23N302 1.25 H20: C, 69.76; H, 6.41; N, 10.61. Found: C, 69.86; H, 6.67; N, 10.51. Example 32
Preparation of (E~)-3-(6-aminopyridin-3-yl)-N-(l -methyl- lH-indol-2-ylmethyl)-N- propylacrylamide
According to the procedure of Example 1, except substituting 1 -methy 1-2- (propylaminomethyl)-lH-indole (0.2 g,l mmole) for l-methyl-2-(methylaminomethyl)- lH- indole, the title compound (0.14 g, 40%) was prepared as a white solid: MS (ES) m/e 349 (M + Η)+.
Example 33
Preparation of (E)-3-(6-aminopyridin-3-vP-N-(5-fluoro-l-methyl-lH-indol-2-ylmethyl)-N- methylacrylamide
According to the procedure of Example 1, except substituting 5-fluoro-2- (methylaminomethyP-lH-indole (0.192 g, 1 mmole) for l-methyl-2-(methylaminomethyl)- lH-indole, the title compound (0.1 g, 30%) was prepared as a white solid : MS (ES) m/e 339 (M + Η)+.
Example 34
Preparation of (E)-3-(6-aminopyridin-3-y P-N-methy l-N-(naphthalen- 1 - ylmethvDacrylam.de
According to the procedure of Example 1, except substituting N-methyl- 1- (methylaminomethyl)naphthalene hydrochloride (0.2 g, 1 mmole) for 1 -methy 1-2- (methylaminomethyl)-lH-indole, the title compound (0.09 g, 28%) was prepared as a white solid: MS (ES) m/e 318 (M + Η)+.
Example 35
Preparation of (E)-3-(6-aminopyridin-3-yl)-N-(benzofuran-2-ylmethvD-N- methylacrylamide
According to the procedure of Example 1, except substituting 2-(methylamino methyPbenzofuran (0.17 g.1.1 mmole) for l-methyl-2-(methylaminomethyl)-lH-indole, the title compound (0.10 g, 30%) was prepared as a white solid: MS (ES) m/e 308 (M + Η)+.
Example 36
Preparation of (E)-N-methyl-3-16-(methylamino)pyridin-3-yll-N-(l -methyl- lH-indol-2- ylmethvDacrylamide
According to the procedure of Example 1, except substituting (E)-3-[6- (methylamino)pyridin-3-yl]acrylic acid (0.15 g, 0.84 mmole) for (E)-3-(6-aminopyridin-3- yl)acrylic acid, the title compound (0.1 g, 37%) was prepared as a white solid: MS (ES) m/e 335 (M + Η)+.
Example 37
Preparation of (E)-3-r6-(dimethylamino)pyridin-3-yll-N-methyl-N-( 1-methyl- lH-indol-2- ylmethyDacrylamide
According to the procedure of Example 1, except substituting (E)-3-[6- (dimethylamino)pyridin-3-yl]acrylic acid (0.20 g, 1.0 mmole) for (E)-3-(6-aminopyridin-3- yPacrylic acid, the title compound (0.22 g, 63%) was prepared as a white solid: MS (ES) m/e 349 (M + Η)+.
Example 38
Preparation of (E>3-(6-aminopyridin-3-vP-N-cvclopropyl-N-(l-methyl-lH-indol-2- ylmethvPacrylamide
According to the procedure of Example 1, except substituting 2- (cyclopropylamino)- 1 -methy 1-lH-indole (0.22 g, 1.1 mmole) for l-methyl-2- (methylaminomethyl)-lH-indole, the title compound (0.154 g, 53%) was prepared as a white solid: MS (ES) m/e 347 (M + Η)+.
Example 39
Preparation of (E)-3-(6-aminopyridin-3-vD-N-methyl-N-(quinolin-3-ylmethyDacrylamide
According to the procedure of Example 1, except substituting 3-
(methylaminomethyPquinoline (0.172 g, 1 mmole) for 1 -methy l-2-(methy laminomethyl)- lH-indole, the title compound (0.100 g, 31%) was prepared as a white solid: MS (ES) m/e 319 (M + Η)+.
Example 40
Preparation of (E)-N-methyl-N-(l-methyl-lH-indol-2-ylmethyl)-3-(6-methylpyridine-3- vPacry lamide
According to the procedure of Example 1, except substituting (E)-3-(6- methylpyridin-3-yl)acrylic acid (0.18 g, 1.1 mmole) for (E)-3-(6-aminopyridin-3-yl)acrylic acid, the title compound (0.11 g, 31%) was prepared as a white solid: MS (ES) m/e 320 (M + Η)+.
Example 41
Preparation of (E)-N-methyl-N-(l-methyl-lH-indol-2-ylmethvP-3-16-(2- oxopropylamino)pyridin-3-yllacrylamide
To a solution of (E)-3-(6-aminopyridin-3-yl)-N-methyl-N-(l -methyl- lH-indol-2- ylmethyl)acrylamide (0.12 g, 0.32 mmol), from Example 1, in DMF (1 mL) was added NaΗ (14 mg, 60% dispersion in oil, 0.35 mmol) and l-bromo-2,2-dimethoxy-propane (0.05 L, 0.37 mmol). After 18 h at RT, the reaction was complete by TLC analysis. The solvent was removed under vacuum and the residue was purified by reverse phase preparative ΗPLC (YMC CombiPrep® ODS- A, 10% to 90% CΗ3CN/Η20 + 0.1% TFA) to give the title compound (11.6 mg) as a pale yellow oil: *H NMR (400 MHz, MeOH-d4, 2: 1 mixture of rotamers, minor rotamer in italics) δ 9.28 and 9.22 (s, 1 H), 8.60 and 8.52 (s, 1 H), 8.25 and 8.15 (d, 1 H), 7.68 (d, J = 16 Hz, 1 H), 7.50 ( , 1 H), 7.35 (m, 3 H), 7.15 ( , 1 H), 7.02 (m, 1 H), 6.55 and 6.25 (s, 1 H), 5.05 and 4.95 (s, 2 H), 3.72 and 3.68 (s, 3 H), 3.50 and 3.48 (s, 3 H), 3.35 (s, 2 H), 3.15 and 3.10 (s, 3 H). MS (ES+) m/e 376.3 ( M + H)+. Unreacted (E)-3-(6-aminopyridin-3-yl)-N-methyl-N-(l-methyl-lH-indol-2- ylmethyl)acrylamide (68 mg) was also recovered.
Example 42
Preparation of (E)-3-(6-aminopyridin-3-yD-N-(lH-indol-2-ylmethyD-N-methylacrylamide
EDC (0.30 g, 1.58 mmole) was added to a solution of 3-(6-aminopyridin-3- y Pacrylic acid (0.26 g, 1.58 mmole), 2-(methy laminomethyl)- lH-indole (0.23 g, 1.43 mmole), ΗOBt • Η20 (0.21 g, 1.58 mmole) and diisopropylethylamine (0.51 mL, 2.86 mmole) in DMF (20 mL) at RT. The reaction was stirred overnight, then was concentrated in vacuo. The residue was diluted with water and extracted with ethyl acetate. The combined organic extracts were washed with brine and dried over Na2Sθ4. Flash chromatography on silica gel (10% MeOH/CHCl^) gave the title compound (0.30 g, 68%) as a light yellow solid: MS (ES) m/e 307 (M + H)+. Example 43
Preparation of (E)-3-(6-aminopyridin-3-y D-N-( 1 -ethyl- lH-indol-2-ylmethv D-N- methylacrylamide
EDC (0.84 g, 4.38 mmole) was added to a solution of 3-(6-aminopyridin-3- yl)acrylic acid (0.72 g, 4.38 mmole), l-ethyl-2-(methylaminomethyl)-lH-indole (0.75 g, 3.98 mmole), ΗOBt Η 0 (0.59 g, 4.38 mmole) and diisopropylethylamine (1.40 mL, 7.96 mmole) in DMF (30 mL) at RT. The reaction was stirred overnight, then was concentrated in vacuo. The residue was diluted with water and extracted with ethyl acetate. The combined organic extracts were washed with brine and dried over Na Sθ4- Flash chromatography on silica gel (5% MeOH/CHCl3) gave the title compound (0.40 g, 30%) as a light tan solid: MS (ES) m/e 335 (M + H)+.
Example 44
Preparation of (E)-3-(6-aminopyridin-3-yl)-N-methyl-N-(l-methyl-lH-indol-3- ylmethyDacrylam.de
EDC (0.35 g, 1.89 mmole) was added to a solution of 3-(6-aminopyridin-3- yl)acrylic acid (0.31 g, 1.89 mmole), l-methyl-3-(methylaminomethyl)-lH-indole (0.30 g, 1.72 mmole), ΗOBt • Η20 (0.24 g, 1.89 mmole) and diisopropylethylamine (0.60 mL, 3.44 mmole) in DMF (20 mL) at RT. The reaction was stirred overnight, then was concentrated in vacuo. The residue was diluted with water and extracted with ethyl acetate. The combined organic extracts were washed with brine and dried over Na2Sθ4. Flash chromatography on silica gel (5% MeOH/CHCl3) gave the title compound (0.30 g, 55%) as a light yellow solid: MS (ES) m/e 321 (M + H)+.
Example 45
Preparation of (E)-3-f6-((E)-but-2-enoylamino)pyridin-3-yll-N-methyl-N-(l-methyl-lH- indol-2-ylmethyl)acrylamide
Crotonic anhydride (0.29 mL, 1.96 mmole) was added to a solution of (E)-3-(6- aminopyridin-3-yl)-N-methyl-N-(l -methyl- lH-indol-2-ylmethyl)acry lamide (0.16 g, 0.49 mmole) and sodium bicarbonate (0.20 g, 2.45 mmole) in TΗF (30 mL) at RT, and the reaction was heated at reflux under nitrogen. After 48 hr, the reaction was concentrated in vacuo and the residue was diluted with water and extracted with ethyl acetate. The combined extracts were dried over Na2S04 and concentrated in vacuo to afford the title compound (0.10 g, 53%) as a tan solid: MS (ES) m/e 389 (M + H)+.
Example 46
Preparation of (E)-3-[6-( 1 ,3-dioxo- 1.3-dihydroisoindol-2-yl)pyridin-3-yll-N-methy l-N-( 1- methy 1- 1 -indol-2-ylmethy Dacry lamide
Phthalic anhydride (0.81 g, 5.48 mmole) was added to a solution of (E)-3-(6- aminopyridin-3-yl)-N-methyl-N-(l-methyl-lH-indol-2-ylmethyl)acrylamide (0.44 g, 1.37 mmole) and sodium bicarbonate (0.58 g, 6.85 mmole) in TΗF (70 mL) at RT, and the reaction was heated at reflux under nitrogen. After 48 hr, the reaction was concentrated in vacuo and the residue was purified by flash chromatography on silica gel (ethyl acetate). The title compound (0.21 g, 33%) was obtained as a white solid: MS (ES) m/e 451 (M + Η)+.
Example 47
Preparation of (EV3-16-1 (2-carboxybenzoyl)amino1pyridin-3-yll-N-methyl-N-( 1-methyl- lH-indol-2-ylmethyl)acry lamide
Phthalic anhydride (0.81 g, 5.48 mmole) was added to a solution of (E)-3-(6- aminopyridin-3-yl)-N-methyl-N-(l-methyl-lH-indol-2-ylmethyl)acrylamide (0.44 g, 1.37 mmole) and sodium bicarbonate (0.58 g, 6.85 mmole) in TΗF (70 mL) at RT, and the reaction was heated at reflux under nitrogen. After 48 hr, the reaction was concentrated in vacuo and the residue was diluted with water and extracted with ethyl acetate. The combined extracts were dried over Na S0 and concentrated. Flash chromatography on silica gel (10% MeOΗ/CΗCl3) gave the title compound (0.10 g, 16%) as a light yellow solid: MS (ES) m/e 469 (M + H)+. Example 48
Preparation of (E)-N-Methyl-N-(l-methyl-lH-indol-2-ylmethyl)-3-16- (propionylamino)pyridin-3-yllacrylamide
Propionic anhydride (0.90 mL, 7.04 mmole) was added to a solution of (E)-3-(6- aminopyridin-3-yl)-N-methyl-N-(l-methyl-lH-indol-2-ylmethyl)acrylamide (0.56 g, 1.76 mmole) and sodium bicarbonate (0.74 g, 8.8 mmole) in TΗF (40 mL) at RT, and the reaction was heated at reflux under nitrogen. After 48 hr, the reaction was concentrated in vacuo and the residue was diluted with water and extracted with ethyl acetate. The combined extracts were dried over Na2S04 and concentrated. Flash chromatography on silica gel (ethyl acetate) gave the title compound (0.35 g, 53%) as a white solid: MS (ES) m/e 377 (M + Η)+.
Example 49
Preparation of (E)-3-16-(3-Ethylureido)pyridin-3-yll-N-methyl-N-(l-methyl-lH-indol-2- ylmethyPacrylamide
Ethyl isocyanate (0.13 mL, 1.68 mmole) was added to a solution of (E)-3-(6- aminopyridin-3-yl)-N-methyl-N-(l -methyl- lH-indol-2-ylmethyl)acry lamide (0.27 g, 0.84 mmole) and triethylamine (0.29 mL, 2.1 mmole) in DMF (30 mL) at RT. The reaction was stirred for 6 days, then was concentrated in vacuo, and the residue was diluted with water and extracted with ethyl acetate. The combined organic extracts were washed with brine, dried over Na2Sθ4, and concentrated. Flash chromatography on silica gel (ethyl acetate) gave the title compound (80 mg, 24%) as a light yellow solid: MS (ES) m/e 392 (M + Η)+.
Example 50
Preparation of (E)-N-methyl-N-(l-methyl-lH-indol-2-ylmethyP-3-r6-(3-methyl- ureido)pyridin-3-yllacrylamide
Methyl isocyanate (0.18 mL, 3.05 mmole) was added to a solution of (E)-3-(6- aminopyridin-3-yl)-N-methyl-N-(l -methyl- lH-indol-2-ylmethyl)acrylamide (0.20 g, 0.61 mmole) and triethylamine (0.17 mL, 1.22 mmole) in DMF (20 mL) at RT. The reaction was stirred for 5 days, then was concentrated in vacuo, and the residue was diluted with water and extracted with ethyl acetate. The combined organic extracts were washed with brine, dried over Na2Sθ4, and concentrated. Flash chromatography on silica gel (ethyl acetate) gave the title compound (0.10 g, 43%) as an off white solid: MS (ES) m/e 378 (M + H)+.
Example 51
Preparation of (E)-3-r6-(acetylamino)pyridin-3-yll-N-methyl-N-(l-methyl-lH-indol-3- ylmethvPacrylamide
Acetic anhydride (0.12 mL, 1.24 mmole) was added to a solution of (E)-3-(6- aminopyridin-3-yl)-N-(l-methyl-lH-indol-3-ylmethyl)-N-methylacrylamide (0.10 g, 0.31 mmole) and sodium bicarbonate (0.13 g, 1.55 mmole) in TΗF (20 mL) at RT, and the reaction was heated at reflux under nitrogen. After 48 hr, the reaction was concentrated in vacuo and the residue was diluted with water and extracted with ethyl acetate. The combined extracts were dried over Na Sθ4 and concentrated. Flash chromatography on silica gel (ethyl acetate) gave the title compound (50 mg, 45%) as a white solid: MS (ES) m/e 363 (M + Η)+.
Example 52
Preparation of (EV3-(6-aminopyridin-3-vD-2-methyl-N-methyl-N-(l-methyl-lH-indol-2- ylmethyDacrylamide
To a stirred solution of (E)-3-(6-aminopyridin-3-yl)-2-methylacrylic acid HCl salt
(0.5 g, 2.3 mmole) in dry 1 :1 DMF/CH2C12 (30 mL) at RT was added l-methyl-2- (methylaminomethyl)indole (0.42 g, 2.4 mmole), HOBt • H20 (0.32 g, 2.4 mmole), Et3N (0.66 mL, 4.7 mmole), and EDC (0.46 g, 2.4 mmole). After stirring for 24 hr the reaction was concentrated to dryness. The residue was taken up in ethyl acetate and the solution was washed with H20, dried (Na2SU4), and concentrated under vacuum. Flash chromatography on silica gel (4% methanol/CHC^) followed by trituration with ethyl acetate/hexane gave the title compound (0.55 g, 75%) as an off-white solid: LCMS (ES) m/e 335.2 (M + H)+; lK NMR (300 MHz, DMSO-d6) δ 7.96 (s, 1 H), 7.52 (d, J = 7.8 Hz, 1 H), 7.48 (dd, 1 H), 7.43 (d, J = 7.8 Hz, 1 H), 7.14 (t, 1 H), 7.03 (t, 1 H), 6.46 (d, J = 8.7 Hz, 1 H), 6.43 (s, 1 H), 6.40 (s, 1 H), 6.17 (br s, 2 H). Example 53
Preparation of 2-(6-aminopyridin-3-ylmethyl)-N-methyl-N-(l-methyl-lH-indol-2- ylmethvDacrylamide
According to the procedure of Example 52, except substituting 2-(6-aminopyridin- 3-ylmethyl)acrylic acid HCl salt (0.50 g, 2.3 mmole) for (E)-3-(6-aminopyridin-3-yl)-2- methylacrylic acid HCl salt, the title compound (0.55 g, 75%) was prepared as an off-white solid following purification by flash chromatography on silica gel (4% methanol/CHC^): LCMS (ES) m/e 335.2 (M + H)+; !H NMR (300 MHz, DMSO-d6) δ 7.75 (d, J = 2.0 Hz, 1 H), 7.50 (d, J = 7.6 Hz, 1 H), 7.38 (d, J = 8.1 Hz, 1 H), 7.22 (dd, 1 H), 7.12 (t, 1 H), 7.01 (t, 1 H), 6.40 (d, J = 8.4 Hz, 1 H), 6.17 (s, 1 H), 5.83 (br s, 2 H), 5.23 (s, 1 H), 5.14 (s, 1 H), 4.73 (s, 2 H), 3.57 (s, 3 H), 3.36 (s, 2 H), 2.82 (s, 3 H).
Example 54
Preparation of (E)-3-(6-aminopyridin-3-yl)-N-rnethyl-N-(naphthalen-2- ylmethypacry lamide
To a stirred solution of (E)-3-(6-aminopyridin-3-yl)acrylic acid (0.30 g, 1.8 mmole) in 1:1 DMF/CH2C12 (25 mL) was added 2-(methylaminomethyl)naphthalene (0.34 g, 2 mmole), HOBt H20 (0.27 g, 2 mmole), Et3N (0.28 mL, 2 mmole), and EDC (0.38 g, 2 mmole). After stirring at RT for 16 hr the reaction was concentrated under vacuum. The residue was taken up in ethyl acetate and the solution was washed with H20, dried (Na2S04) and concentrated to dryness. Purification by flash chromatography on silica gel (4% methanol/CHCl3), trituration with 1 : 1 ethyl acetate/hexane, filtration, and drying under vacuum gave the title compound (0.49 g, 81%) as an off-white solid: LCMS (ES) m/e 318.0 (M + H)+.
Example 55
Preparation of (E)-3-(6-amino-4-methylpyridin-3-yl)-N-methyl-N-(l-methyl-lH-indol-2- ylmethyPacrylamide
To a stirred solution of (E)-3-(6-amino-4-methylpyridin-3-yl)acrylic acid ΗC1 salt
(0.70 g, 3.3 mmole) in 1:1 DMF/CΗ2C12 (30 mL) was added Et3N (0.42 mL, 3 mmole), 1- methyl-2-(methylaminomethyl)indole (0.50 g, 2.9 mmole), HOBt • H 0 (0.41 g, 3 mmole), and DCC (0.70 g, 3 mmole). After stirring at RT for 16 hr the reaction was concentrated under vacuum. The residue was taken up in ethyl acetate and filtered. The filtrate was washed with 1.0 N Na2C03 then with brine, dried (Na Sθ4), and concentrated under vacuum. Purification by flash chromatography on silica gel (4% methanol/CHCl3) gave the title compound (0.74 g, 74%) as a pale yellow solid: LCMS (ES) m/e 335.2 (M + H)+.
Example 56
Preparation of (E)-3-(6-aminopyridin-3-yl)-N-(l,3-dimethyl-lH-indol-2-ylmethyl)-N- methylacrylamide
To a stirred solution of l,3-dimethyl-2-(methylaminomethyl)-l -indole (0.6 g, 3.2 mmole) in 1:1 DMF/CΗ2C12 (25 mL) was added (E)-3-(6-aminopyridin-3-yl)acrylic acid (0.50 g, 3 mmole), HOBt H20 (0.43 g, 3.2 mmole), and DCC (0.66 g, 3.2 mmole). After stirring at RT for 16 hr the reaction was concentrated under vacuum. Purification by flash chromatography on silica gel (3% methanol/CHC^) gave the title compound (0.83 g, 83%) as an off-white solid: LCMS (ES) m/e 335.4 (M + H)+.
Example 57
Preparation of (E)-N-methyl-N-t 1 -methyl- 1 -indol-2-ylmethyl)-3-(2-oxo- 1.4-dihydro-2/ - pyridof 2.3-til- 1.3-oxazin-6-yl)acry lamide
a) N-Methyl-N-(l-methyl-lH-indol-2-ylmethyl)acrylamide
To a stirred solution of l-methyl-2-(methylaminomethyl)-lH-indole (1.0 g, 5.7 mmole), from Preparation 1, and Et3N (0.9 mL, 6.4 mmole) in CΗ2C12 (50 mL) at 0 °C was added dropwise acryloyl chloride (0.51 mL, 6 mmole) over 5 minutes. The reaction was stirred at 0 °C for 1 hr, then was poured into ice water. The organic phase was separated, washed with brine, dried (MgS04), and concentrated to dryness to give the title compound (1.19 g, 91%) as a yellow oil. This was used without further purification: TLC (silica gel, 50% EtOAc/hexanes) Rf = 0.31. b) (E)-N-methyl-N-(l -methy 1-1 /-indol-2-ylmethyl)-3-(2-oxo-l,4-dihydro-2H-pyrido[2,3- d]- 1 ,3-oxazin-6-yl)acry lamide
To a stirred solution of N-methyl-N-(l -methyl- l/ -indol-2-ylmethyl)acrylamide (1.19 g, 5.2 mmole) in propionitrile (50 mL) was added 6-bromo-2-oxo-l,4-dihydro-2//- pyrido[2,3--/]- 1 ,3-oxazine (1.1 g, 4.9 mmole), DIEA ( 1.75 mL, 10 mmole), palladium(II) acetate (112 mg, 0.5 mmole), and tri-o-tolylphosphine (304 mg, 1.0 mmole). The reaction was purged with argon and heated at reflux for 16 hr, then was cooled to RT and concentrated under vacuum. The residue was taken up in CΗCI3 and the solution was filtered through a pad of silica gel (3% methanol/CHCl3). The filtrate was concentrated and the residue was triturated with ethyl acetate, collected by suction filtration, and dried under vacuum gave the title compound (1.02 g, 55%) as an off-white solid: LCMS (ES) m/e 377.4 (M + H)+.
Example 58
Preparation of (E)-N-(1.3-dimethyl-lH-indol-2-ylmethyl)-N-methyl-3-(7-oxo-5,6.7.8- tetrahydro-l,8-naphthyridin-3-yl)acrylamide
a) N-(l,3-Dimethyl-lH-indol-2-ylmethyl)-N-methylacrylamide To a stirred solution of l,3-dimethyl-2-(methylaminomethyl)indole (1.5 g, 8 mmole), from Preparation 40, and Et3N (1.12 mL, 8 mmole) in CΗ C12 (75 mL) at 0 °C was added acryloyl chloride (0.65 mL, 8 mmole) dropwise over 5 minutes. The reaction was stirred at 0 °C for 1 hr then was poured into ice water. The organic phase was separated, washed with brine, dried (MgSθ4), and concentrated to dryness to give the title compound (1.7 g, 90%) as a yellow oil. This was used without further purification: TLC silica gel (50% EtOAc/hexanes) Rf = 0.41.
b) (E)-N-( 1 ,3-Dimethy 1- lH-indol-2-ylmethyl)-N-methyl-3-(7-oxo-5,6,7,8- tetrahydro[ 1 ,8]naphthyridin-3-yl)acrylamide To a stirred solution of N-( 1 ,3-dimethyl- lH-indol-2-ylmethyl)-N-methy lacry lamide
(1.7 g, 7 mmole) in propionitrile (50 mL) was added 6-bromo-3,4-dihydro-lH-l,8- naphthyridin-2-one (1.16 g, 5.1 mmole), DIEA (1.8 mL, 10.3 mmole), palladium(II) acetate (112 mg, 0.5 mmole), and tri-o-tolylphosphine (304 mg, 1.0 mmole). The reaction was purged with argon and heated at reflux for 16 hr, then was cooled to RT and concentrated under vacuum. Purification by flash chromatography on silica gel (5% methanol/CΗCl3), trituration with ethyl acetate, filtration, and drying under vacuum gave the title compound (1.17 g, 59%) as an off-white solid: LCMS (ES) m/e 389.2 (M + H)+.
Example 59
Preparation of (E)-3-(6-aminopyridin-3-yl)-N-methyl-N-(3-methylbenzorfrlthiophen-2- ylmethvPacrylamide
To a stirred solution of 3-methyl-2-(methylaminomethyl)benzo[4»]thiophene (0.30 g, 1.6 mmole) in 1: 1 DMF/CH2C12 was added (E)-3-(6-aminopyridin-3-yl)acrylic acid (0.33 g, 2 mmole), HOBt • H20 (0.27 g, 2 mmole), and DCC (0.41 g, 2 mmole). The reaction was stirred for 16 hr, then was concentrated under vacuum. The residue was taken up in CHCI3, washed with H20, dried (Na Sθ4) and concentrated. Purification by flash chromatography on silica gel (4% methanol/CHC^) gave the title compound (0.39 g, 72%) as a pale yellow solid: LCMS (ES) m/e 338.2 (M + H)+.
Example 60
Preparation of (E)-3-(2-aminopyrimidin-5-yl)-N-(benzor-7lthiophen-2-ylmethvD-N- methylacrylamide
According to the procedure of Example 1 , except substituting (E)-3-(2- aminopyrimidin-5-yl)acrylic acid (1.49 g, 7.1 mmole) for (E)-3-(6-aminopyridin-3- yl)acrylic acid, and substituting 2-(methylaminomethyl)theino[2,3-σ]thiophene (1.38 g, 7.8 mmole) for l-methyl-2-(methylaminomethyl)indole, the title compound (2.04 g, 89%) was prepared as a yellow solid: MS (ES) m/e 325 (M + H)+.
Example 61
Preparation of (E)-3-(2-aminopyrimidin-5-yl)-N-methyl-N-(l-methyl-lH-indol-3- yl methvPacry lamide
According to the procedure of Example 31, except substituting 1 -methy 1-3- (methylaminomethyl)indole (1.96 g, 8.6 mmole) for 3-methyl-2- (methylaminomethyPindene hydrochloride, and substituting 2-amino-5-bromopyrimidine (1.0 g, 5.75 mmole) for 6-bromo-3,4-dihydro-lH-l,8-naphthyridin-2-one, the title compound (1.44 g, 78%) was prepared as a yellow solid: MS (ES) m/e 322 (M + Η)+.
Example 62
Preparation of (E)-N-methyl-N-(l-methyl-lH-indol-3-ylmethyl)-3-(7-oxo-5,6,7,8- tetrahvdro- 1.8-naphthyridin-3-yl)acrylamide
According to the procedure of Example 31, except substituting 1 -methy 1-3- (methylaminomethyPindole (0.75 g, 3.3 mmole) for 3-methyl-2-
(methylaminomethyl)indene hydrochloride, the title compound (0.59 g, 72%) was prepared as a light yellow solid: MS (ES) m/e 375 (M + Η)+.
Example 63
Preparation of (E)-3-r2-aminopyrimidin-5-yll-N-methyl-N-(3-methyl-lH-inden-2- ylmethyPacrylamide
According to the procedure of Example 31, except substituting 2-amino-5- bromopyrimidine (0.32 g, 1.84 mmole) for 6-bromo-3,4-dihydro-lH-l,8-naphthyridin-2- one, the title compound (0.47 g, 80%) was prepared as a light yellow solid: MS (ES) m/e 321 (M + Η)+.
Example 64
Preparation of (E)-3-f2-(acetylamino)pyrimidin-5-yll-N-methyl-N-(l -methyl- lH-indol-2- ylmethyPacrylamide
According to the procedure of Example 31, except substituting l-methyl-2- (methylaminomethyl)indole (1.45 g, 8.33 mmole) for 3-methyl-2-
(methylaminomethyl)indene hydrochloride, and substituting 2-acetylamino-5- bromopyrimidine (1.20 g, 5.55 mmole) for 6-bromo-3,4-dihydro-lH-l,8-naphthyridin-2- one, the title compound (2.38 g, 43%) was prepared as a yellow solid: MS (ES) m/e 364 (M + Η)+. Example 65
Preparation of (E)-3-(6-aminopyridin-3-yl)-N-methyl-N-(2-methyl-l/ -indol-3- ylmethyPacrylamide
According to the procedure of Example 1, except substituting 2-methyl-3- (methylaminomethyl)indole (0.45 g, 2.58 mmole) for l-methyl-2- (methylaminomethyl)indole, the title compound (0.68 g, 90%) was prepared as a yellow solid: MS (ES) m/e 321 (M + H)+.
Example 66
Preparation of (E)-3-(2-aminopyrimidin-5-yP-N-(1.2-dimeth l-l//-indol-3-ylmethyl)-N- methylacrylamide
According to the procedure of Example 31, except substituting l,2-dimethyl-3- (methylaminomethyl)indole (1.62 g, 8.62 mmole) for 3-methyl-2- (methylaminomethyPindene hydrochloride, and substituting 2-amino-5-bromopyrimidine (1.00 g, 5.75 mmole) for 6-bromo-3,4-dihydro-l/ -l,8-naphthyridin-2-one, the title compound (1.33 g, 69%) was prepared as a yellow solid: MS (ES) m/e 336 (M + H -.+^
Example 67
Preparation of (E)-N-methyl-N-( 1 -methyl- l -indol-2-ylmethyP-3-(3-oxo-3.4-dihydro-2H- pyrido[3,2-o1-1.4-oxazin-7-yPacrylamide
According to the procedure of Example 31, except substituting 1 -methy 1-2- (methylaminomethyl)indole (1.17 g, 6.75 mmole) for 3-methyl-2- (methylaminomethyl)indene hydrochloride, and substituting 5-bromo-2H-pyrido[3,2-έ>]- l,4-oxazin-3(4 )-one (1.03 g, 4.50 mmole) for 6-bromo-3,4-dihydro-l /-l,8-naphthyridin- 2-one, the title compound (0.90 g, 53%) was prepared as a light yellow solid: MS (ES) m/e 377 (M + Η)+. Example 68
Preparation of (E)-N-methyl-N-(2-methyl-l /-indol-3-ylmethyl)-3-(7-oχo-5.6,7,8- tetrahvdro- 1.8-naphthyridin-3-yl)acrylamide
According to the procedure of Example 31, except substituting 2-methyl-3- (methylaminomethypindole (1.40 g, 8.00 mmole) for 3-methyl-2-
(methylaminomethypindene hydrochloride, the title compound (1.30 g, 65%) was prepared as a light yellow solid: MS (ES) m/e 376 (M + H)+.
Example 69
Preparation of (E)-N-methyl-N-(l-methyl-l /-indol-3-ylmethvP-3-(3-oxo-3.4-dihydro-2H- pyridor3.2-/?1- 1.4-oxazin-7-yl)acrylamide
According to the procedure of Example 31, except substituting l-methyl-3- (methylaminomethyl)indole (0.38 g, 2.20 mmole) for 3-methyl-2- (methylaminomethyl)indene hydrochloride, and substituting 5-bromo- 2H-pyrido[3,2-Z>]- l,4-oxazin-3(4//)-one (0.32 g, 1.40 mmole) for 6-bromo-3,4-dihydro-lH-l,8-naphthyridin- 2-one, the title compound (0.26 g, 50%) was prepared as a light yellow solid: MS (ES) m/e 311 (M + Η)+.
Example 70
Preparation of (E)-N-methyl-N-(l-methyl- l/ -indol-2-ylmethyl)-3-(7-oxo-5,6.7,8- tetrahydro- 1 ,8-naphthyridin-3-yl)propionamide
To a solution of (E)-N-methyl-N-( l -methyl- lH-indol-2-ylmethyl)-3-(7-oxo- 5,6,7, 8-tetrahydro-l,8-naphthyridin-3-yl)acrylamide (0.15 g, 0.40 mmole) in dioxane at RT was added Pd(OΗ)2. The flask was sealed with a septum through which a balloon containing hydrogen (1 atm) was inserted. The reaction was stirred at RT overnight and then filtered through a pad of celite®, washing with methanol. The filtrate was concentrated to give the title compound (0.14 g, 94 %) as a light yellow solid: MS (ES) m/e 378 (M + H)+. Example 71
Preparation of (EV3-(6-aminopyridin-3-yl)-N-(6-methoxy- l-methyl-lH-indol-2-ylmethvP- N-methylacrylamide
According to the procedure of Example 1, except substituting 1 -methy 1-2- (methylaminomethyl)-6-methoxy-l /-indole for 1 -methy l-2-(methy laminomethy 1)- 1//- indole, the title compound (50%) was prepared as a light yellow solid: MS (ES) m/e 351.4 (M + Η)+ Anal. Calcd for C20H22N4O2 • 1.5 H20: C, 63.64; H, 6.66; N, 14.84. Found: C, 63.51; H, 6.21; N, 14.71.
Example 72
Preparation of (E)-3-(7-aminopyridin-3-yl)-N-(1.7-dimethyl-lH-indol-3-ylmethyl)-N- methylacrylamide
According to the procedure of Example 1, except substituting l,7-dimethyl-3- (methylaminomethyl)-l //-indole for l-methyl-2-(methylaminomethyl)-lH-indole, the title compound (50%) was prepared as a light yellow solid: MS (ES) m/e 335.2 (M + Η)+. Anal. Calcd for C20H22N4O2 • 0.5 H20: C, 69.99; H, 6.76; N, 16.31. Found: C, 70.02; H, 6.59; N, 16.43.
Example 73
Preparation of (E)-3-(6-aminopyridin-3-yl)-N-( 1.5-dimethyl- 1 H-indol-3-ylmethy P-N- methylacrylamide
According to the procedure of Example 1, except substituting l,5-dimethyl-3- (methylaminomethyP-lH-indole for 1 -methy l-2-(methy laminomethy 1)-1Z/- indole, the title compound (33%) was prepared as a light yellow solid: MS (ES) m/e 335.2 (M + Η)+. Anal. Calcd for C20H22N4O2 • H20: C, 68.16; H, 6.86; N, 15.89. Found: C, 68.37; H, 6.70; N, 15.62. Example 74
Preparation of (E)-3-(6-aminopyridin-3-yP-N-(1.6-dimethyl-lH-indol-3-ylmethvP-N- methylacrylamide
According to the procedure of Example 1, except substituting l,6-dimethyl-3- (methylaminomethyl)-lH-indole for l-methyl-2-(methylaminomethyl)-lH-indole, the title compound (33%) was prepared as a light tan solid: MS (ES) m/e 335.2 (M + Η)+. Anal. Calcd for C20H22N4O2 • 0.375 H20: C, 70.41; H, 6.64; N, 16.42. Found: C, 70.40; H, 6.61; N, 16.19.
Example 75
Preparation of (E)-3-(6-aminopyridin-3- yl)-N-( 1 -benzyl- 1 H-indol-3-y lmethvP-N- methylacrylamide
EDC (0.42 g, 2.20 mmole) was added to a solution of 3-(6-aminopyridin-3- yl)acrylic acid (0.36 g, 2.20 mmole), l-benzyl-3-(methy laminomethy l)-lH-indole (0.50 g, 2.00 mmole), ΗOBt • Η20 (0.30 g, 2.20 mmole) and diisopropylethylamine (0.70 mL, 4.00 mmole) in DMF (30 mL) at RT. The reaction was stirred overnight, then was concentrated in vacuo. The residue was diluted with water and extracted with ethyl acetate. The combined organic extracts were washed with brine and dried over Na Sθ4- Flash chromatography on silica gel (10% MeOH/CHCl3) gave the title compound (0.48 g, 60%) as a light yellow solid: MS (ES) m/e 397 (M + H)+.
Example 76
Preparation of (E)-N-methyl-N-(l-methyl-lH-indol-2-ylmethyl)-3-r6- (phenylamino)pyridin-3-vnacrylamide
a) N-Methy l-N-( 1 -methyl- 1 H-indol-2-y lmethy l)acry lamide
To a stirred solution of l-methyl-2-(methylaminomethyl)-lH-indole (1.5 g, 8.6 mmole) and E-3N (1.35 mL, 9.6 mmole) in CΗ2C1 (75 mL) at 0 °C was added dropwise acryloyl chloride (0.77 mL, 9.5 mmole) over 5 minutes. After 2 h the reaction was washed with cold H 0, brine, dried (MgS04) and concentrated under vacuum. The residue was used without further purification. b) (E)-N-Methyl-N-(l-methyl- lr/-indol-2-yImethyI)-3-[6-(phenylamino)pyridin-3- yl]acrylamide
N-Methyl-N-(1 -methyl- lH-indol-2-ylmethyl)acry lamide (from Example 76 (a)) was taken up in propionitrile (50 mL). To this solution was added with stirring 2- phenylamino-5-bromopyridine (1.3 g, 5.2 mmole), DIEA (1.8 mL, 10 mmole), Pd(OAc) (1 12 mg, 0.5 mmole) and P(o-tol)3 (304 mg, 1.0 mmole). The reaction was purged with argon then stirred at reflux for 16 h. After cooling to room temperature the reaction was concentrated to dryness under vacuum. Flash chromatography on silica gel (5% methanol/CΗCl3) followed by a second flash column on silica gel (50-70% EtOAc/CHC^) left a residue that was triturated with EtOAc/petroleum ether. Filtration and drying under vacuum gave the title compound (1.42 g, 69%) as an off-white powder: MS (ES) m/e 396.20 (M + H)+.
Example 77
Preparation of (E)-3-(6-aminopyridin-3-yl)-N-( 1.2-dimethyl- 1 H-indol-3-ylmethy P-N- methylacrylamide
To a stirred solution of l,2-dimethyl-3-(methylaminomethyl)-lH-indole (0.8 g, 4.2 mmole) in 1 : 1 DMF/CΗ C1 (30 mL) at RT was added (E)-3-(6-aminopyridin-3-yl)acrylic acid (0.7 g, 4.3 mmole), Et3N (0.61 mL, 4.3 mmole), HOBt • H20 (0.58 g, 4.3 mmole) and EDC (0.83 g, 4.3 mmole). After 16 h the reaction was concentrated under vacuum and the residue was taken up in EtOAc (100 mL). The solution was washed with 1.0 N Na Cθ3 (100 mL) then with brine, dried (Na2Sθ4), and concentrated. Purification by flash chromatography on silica gel (4% MeOH/CHCl3) followed by trituration with 1 : 1 Et 0/petroleum ether and drying under vacuum gave the title compound (1.36 g, 97%) as an off-white solid: MS (ES) m/e 335.2 (M + H)+.
Example 78
Preparation of (E)-3-(6-aminopyridin-3-yl)-N-(benzorfrlthiophen-3-ylmethvP-N- methylacrylamide
According to the procedure of Example 77, except substituting 3-
( methy laminomethyl)benzo[/>]thiophene (0.75 g, 4.2 mmole) for the l,2-dimethyl-3- ( methy laminomethy 1)-1 //-indole, the title compound (1.05 g, 83%) was prepared as an off- white solid: MS (ES) m/e 324.2 (M + H)+.
Example 79
Preparation of (E)-N-methyl-N-(l-methyl-l -indol-2-ylmethyl)-3-r6-(pyridin-2- ylamino)pyridin-3-yllacrylamide
According to the procedure of Example 76 (a) and (b), except substituting 5- bromo-2,2'-dipyridylamine (1.3 g, 5.2 mmole) for the 2-phenylamino-5-bromopyridine, the title compound (1.54 g, 75%) was prepared as an off-white solid: MS (ES) m/e 398.2 (M + H)+.
Example 80
Preparation of (E)-N-(l,2-dimethyl-l//-indol-3-ylmethyP-N-methyl-3-(7-oxo-5.6,7.8- tetrahydro- 1.8-naphthyridin-3-yl)acrylamide
a) N-Methyl-N-(l,2-dimethyl-l/ -indol-2-ylmethyl)acry lamide According to the procedure of Example 76 (a), except substituting l,2-dimethyl-3-
(methylaminomethyl)-lH-indole (1.5 g, 8 mmole) for the l-methyl-2- (methy laminomethy p-l / -indole, the title compound was prepared and used without further purification.
b) (E)-N-(l,2-Dimethyl-l /-indol-3-ylmethyl)-N-methyl-3-(7-oxo-5,6,7,8-tetrahydro-l,8- naphthyridin-3-yl)acrylamide
According to the procedure of Example 76 (b), except substituting 6-bromo-3,4- dihydro-lH-l,8-naphthyridin-2-one (1.3 g, 5.7 mmole) for the 2-phenylamino-5- bromopyridine, the title compound (0.57 g, 26%) was prepared as a white solid: MS (ES) m/e 389.19 (M + Η)+. Example 81
Preparation of (E)-N-methyl-N-(3-methv.benzorfrlthiophen-2-ylmethvπ-3-(7-oxo-5.6,7,8- tetrahydro- 1.8-naphthyridin-3-yPacrylamide
a) N-Methyl-N-(3-methylbenzo[b]thiophen-2-ylmethyl)acrylamide
According to the procedure of Example 76 (a), except substituting 2- (methylarninomethyl)-3-methylbenzo[b]thiophene (1.53 g, 8 mmole) for the l-methyl-2- (methylaminomethyl)-lH-indole, the title compound was prepared and used without further purification.
b) (E)-N-Methyl-N-(3-methylbenzo[σ]thiophen-2-ylmethyl)-3-(7-oxo-5,6,7,8-tetrahydro- 1 ,8-naphthyridin-3-yl)acrylamide
According to the procedure of Example 76 (b), except substituting 6-bromo-3,4- dihydro-lH-l,8-naphthyridin-2-one (1.3 g, 5.7 mmole) for the 2-phenylamino-5- bromopyridine, the title compound (0.85 g, 33%) was prepared as an off-white solid: MS (ES) m/e 392.2 (M + Η)+.
Example 82
Preparation of (E)-3-(6-aminopyridin-3-yl)-N-methyl-(2-methylbenzorolthiophen-3- ylmethvDacrylamide
According to the procedure of Example 77, except substituting 2-methyl-3- (methylaminomethyl)benzo[fo]thiophene (1.2 g, 6.1 mmole) for the l,2-dimethyl-3-
(methylaminornethyl)-lH-indole, the title compound (1.22 g, 59%) was prepared as a pale yellow solid: MS (ES) m/e 338.2 (M + Η)+.
Example 83
Preparation of (E)-3-(6-aminopyridin-3-vP-N-(3,4-dimethylthieno.2.3-plthiophen-2- ylmethyP-N-methylacrylamide
According to the procedure of Example 1, except substituting 3,4-dimethyl-2- (methylaminomethyl)thieno[2,3-b]thiophene (0.026 g, 0.126 mmole) for the l-methyl-2- (methylaminomethyl)-lH-indole, the title compound (0.013 g,72%) was prepared as a white solid: MS (ES) m/e 358 (M + Η)+.
Example 84
Preparation of (E)-3-(6-aminopyridin-3-yl)-N-methy l-N-( 1 -methy lnaphthalen-2- ylmethvDacrylamide
According to the procedure of Example 1, except substituting l-methyl-2- (methylaminornethyl)naphthalene (0.100 g, 0.54 mmole) for the l-methyl-2-
( methy laminomethyl)- 1 /-indole, the title compound (0.088 g, 49%) was prepared as a white solid: MS (ES) m/e 332 (M + H)+.
Example 85
Preparation of (E)-3-(6-aminopyridin-3-yl)-N-methyl-N-(l -methyl- lH-pyrrolo[2,3- plpyridin-3-ylmethyl)acry lamide
According to the procedure of Example 1, except substituting 1 -methy 1-3- (methylaminomethyl)-lΗ-pyrrolo[2,3-o]pyridine (0.2 g, 1.14 mmole) for the l-methyl-2- (methylaminomethyP-lH-indole, the title compound (0.19 g, 52%) was prepared as a white solid: MS (ES) m/e 322 (M + Η)+.
Example 86
Preparation of (E)-3-(6-aminopyridin-3-vP-N-(2,3-dihvdro-lH-3a-azacyclopentafα1inden- 8-yP-N-methylacrylamide
According to the procedure of Example 1 , except substituting 2,3-dihydro-8- (methylaminomethyl)-lH-3a-azacyclopenta[α]indene (0.100 g, 0.5 mmole) for the 1- methy l-2-(methylaminomethyl)- 1 //-indole, the title compoud (0.063 g, 36%) was prepared as a white solid: MS (ES) m/e 347 (M + Η)+. Example 87
Parenteral Dosage Unit Composition
A preparation which contains 20 mg of the compound of Example 1 as a sterile dry powder is prepared as follows: 20 mg of the compound is dissolved in 15 mL of distilled water. The solution is filtered under sterile conditions into a 25 mL multi-dose ampoule and lyophilized. The powder is reconstituted by addition of 20 mL of 5% dextrose in water (D5W) for intravenous or intramuscular injection. The dosage is thereby determined by the injection volume. Subsequent dilution may be made by addition of a metered volume of this dosage unit to another volume of D5W for injection, or a metered dose may be added to another mechanism for dispensing the drug, as in a bottle or bag for IV drip infusion or other injection-infusion system.
Example 88
Oral Dosage Unit Composition
A capsule for oral administration is prepared by mixing and milling 50 mg of the compound of Example 1 with 75 mg of lactose and 5 mg of magnesium stearate. The resulting powder is screened and filled into a hard gelatin capsule.
Example 89
Oral Dosage Unit Composition
A tablet for oral administration is prepared by mixing and granulating 20 mg of sucrose, 150 mg of calcium sulfate dihydrate and 50 mg of the compound of Example 1 with a 10% gelatin solution. The wet granules are screened, dried, mixed with 10 mg starch, 5 mg talc and 3 mg stearic acid; and compressed into a tablet.
The above description fully discloses how to make and use the present invention. However, the present invention is not limited to the particular embodiments described hereinabove, but includes all modifications thereof within the scope of the following claims. The various references to journals, patents and other publications which are cited herein comprises the state of the art and are incorporated herein by reference as though fully set forth.

Claims

What is claimed is:
A compound according to formula (I):
Figure imgf000106_0001
wherein:
Figure imgf000106_0002
Figure imgf000106_0003
Figure imgf000107_0001
R1 isH0rC1.4a.kyl;
R2 is H, Cj.4alkyl or C3_6cycloalkyl;
R3is
Figure imgf000107_0002
Figure imgf000108_0001
R4isHorC!.4alkyl;
indicates that one of the two designated bonds is a double bond and the other is a single bond;
R5 is CH2 when the bond to which it is attached is a double bond; or R^ is H . or Cj_4alkyl when the bond to which it is attached is a single bond;
R6isHorCι_4alkyl;
R7 is H, Ci-6alkyl or -Q)-6alkyl-Ar;
Y is H, Cι_4alkyl, N(RT)2, NHC(0)R\ NHCH2C(0)R' or NHC(0)CH=CHR'; each X independently is H, Cι_ alkyl, CH2OH, OR', SR', CN, N(R')2, CH2N(R)2, N02, CF3, C02R', CON(R)2, COR', NR C(0)R', F, Cl, Br, I or -S(0)rCF3;
W is S or O;
QisH0r .4a.kyl;
M is CH or O;
L is CH2 or C(O);
E is O or NR'; each R' independently is H, Ci-6alkyl or -Q)-6alky--Ar; and r is 0, 1 or 2; or a pharmaceutically acceptable salt thereof.
2. A compound according to claim 1 of formula (la):
Figure imgf000108_0002
3. A compound according to claim 2 of formula (II):
Figure imgf000109_0001
(II).
4. A compound according to claim 1 of formula (Ila):
Figure imgf000109_0002
x (Ila).
A compound according to claim 1 of formula (lib):
Figure imgf000109_0003
(lib).
A compound according to claim 1 in which R3 is:
Figure imgf000109_0004
7. A compound according to claim 1 in which R3 is:
Figure imgf000109_0005
8. A compound according to claim 1 in which R3 is:
Figure imgf000110_0001
9. A compound according to claim 1 which is:
(E)-3-(6-Aminopyridin-3-yl)-N-methyl-N-(l-methyl-lH-indol-2- ylmethyl)acrylamide;
(E)-3-(4-Aminophenyl)-N-methyl-N-(l-methyl-lH-indol-2-ylmethyl)acrylamide; (E)-N-Methyl-N-(l -methyl- lH-indol-2-ylmethyl)-3-(pyridin-3-yl)acrylamide;
(E)-3-(2-Aminopyrimidin-5-yl)-N-methy.-N-(l-methyl- lH-indol-2- ylmethyl)acrylamide;
(E)-3-(6-Aminopyridin-3-yl)-N-(benzo[ ?]thiophen-2-yImethyl)-N- methylacrylamide; (E)-3-(6-Aminopyridin-3-yl)-N-methyl-N-(l -methy 1-1 H-indol-2-ylmethyl)-2- butenamide;
(E)-3-(6- Aminopyridin-3-yl)-N-methy l-N-( 1 -methyl- lH-indazol-3- ylmethyPacrylamide;
(E)-3-(6-Amino-2-methylpyridin-3-yl)-N-methyl-N-(l -methyl- lH-indol-2- ylmethyPacrylam.de;
(E)-3-(6-Amino-5-methylpyridin-3-yl)-N-methyl-N-(l -methyl- lH-indol-2- ylmethyPacrylamide;
(E)-3-(6-Aminopyridin-3-yl)-N-(l-methyl-lH-indol-2-ylmethyl)-N- propylacrylamide; (E)-3-(6-Aminopyridin-3-yl)-N-(5-fluoro-l-methyl-lH-indol-2-ylmethyl)-N- methylacrylamide;
(E)-3-(6-Aminopyridin-3-yl)-N-methyl-N-(naphtha.en-l-ylmethyl)acrylamide;
(E)-3-(6-Aminopyridin-3-yl)-2,N-dimethyl-N-(l-methyl-lH-indol-2- ylmethyPacrylamide; (E)-3-(6-Aminopyridin-3-yl)-N-methyl-N-(naphthalen-2-ylmethyl)acrylamide;
2-(6-Aminopyridin-3-ylmethyl)-N-methyl-N-(l-methyl-lH-indol-2- ylmethyl)acrylamide;
(E)-3-(6-Aminopyridin-3-yl)-N-(benzofuran-2-ylmethyl)-N-methylacrylamide; (E)-3-(3,4-Dihydro-2H-pyrido[3,2-/?]-l,4-oxazin-7-yl)-N-methyl-(l-methyl-lH- indol-2-ylmethyl)acrylamide;
(E)-N-Methyl-3-[6-(methylamino)pyridin-3-yl]-N-(l-methyl- lH-indol-2- ylmethyl)acry lamide; (E)-3-[6-(Dimethylamino)pyridin-3-yl]-N-methyl-N-(l-methyl- lH-indol-2- ylmethyl)acrylamide;
(E)-N-Methyl-N-[(l-methyl-lH-indol-2-yl)methyl]-3-(5,6,7,8-tetrahydro- l,8- naphthyridin-3-yl)acrylamide;
(E)-3-[6-(Acetylamino)pyridin-3-yl]-N-methyl-N-(l-methyl-lH-indol-2- ylmethypacrylamide;
(E)-3-(6-Amino-5-methylpyridin-3-yl)-N-(benzo[Z?]thiophen-2-ylmethyl)-N- methylacrylamide;
(E)-3-(6-Amino-5-methylpyridin-3-yl)-N-methyl-N-(naphthalen-2- ylmethyl)acrylamide; (E)-3-(6-Amino-4-methylpyridin-3-y l)-N-methyl-N-( 1 -methyl- 1 H-indol-2- ylmethypacrylamide;
(E)-3-(6-Aminopyridin-3-yl)-N-cyclopropyl-N-( 1 -methyl- 1 H-indol-2- ylmethyPacrylamide;
(E)-3-(6-Aminopyridin-3-yl)-N-methyl-N-(l-methyl-lH-indol-3- ylmethyPacrylamide;
(E)-3-(6-Aminopyridin-3-yl)-N-methyl-N-(quinolin-3-ylmethyl)acry lamide;
(E)-3-(6-Aminopyridin-3-yl)-N-methyl-N-(thieno[2,3-/7]thiophen-2- ylmethyl)acrylamide;
(E)-N-Methyl-N-(l-methyl-lH-indol-2-ylmethyl)-3-(6-methylpyridine-3- yl)acrylamide;
(E)-3-[6-(Acetylamino)-5-methylpyridin-3-yl]-N-methyl-N-(l-methyl-lH-indol-2- ylmethyl)acrylamide;
(E)-3-(6-Aminopyridin-3-yl)-N-(lH-indol-2-ylmethyl)-N-methylacrylamide;
(E)-N-Methyl-N-(l-methyl-lH-indol-2-ylmethyl)-3-[6-(2-oxopropylamino)pyridin- 3-yl]acrylamide;
(E)-3-(6-Aminopyridin-3-yl)-N-methyl-N-(thieno[3,2-/7]thiophen-2- ylmethyPacrylamide;
(E)-3-[6-Amino-5-(hydroxymethyl)pyridin-3-yl]-N-methyl-N-(l-methyl-lH-indol- 2-ylmethyl)acrylamide; (E)-3-(3HImidazo[4,5-i ]pyridin-6-yl)-N-methyl-N-(l -methyl- lH-indol-2- ylmethyPacrylamide; (E)-3-[6-Aminopyridin-3-yl]-N-(l-ethyl- l//-indol-2-ylmethyl)-N- methylacrylamide;
(E)-3-[6-Aminopyridin-3-yl]-N-(l,3-dimethyl-l//-indol-2-ylmethyl)-N- methylacrylamide; (E)-3-[6-((E)-But-2-enoylamino)pyridin-3-yl]-N-methyl-N-(l-methyl- l//-indol-2- ylmethyPacrylamide;
(E)-N-Methyl-N-(l-methyl-l//-indol-2-ylmethyl)-3-(7-oxo-5,6,7,8-tetrahydro-l,8- naphthyridin-3-yl)acrylamide;
(E)-3-[6-Amino-5-[(2-hydroxyethylamino)carbonyl]pyridin-3-yl]-N-(l-methyl-l /- indol-2-ylmethyl)-N-methylacry lamide;
(E)-3-[6-Aminopyridin-3-yl]-N-methyl-N-(3-methyl-l /-inden-2- ylmethypacrylamide;
(E)-3-[6-Aminopyridin-3-yl]-N-(l/ -inden-2-ylmethyl)-N-methylacrylamide;
(E)-3-(6-Aminopyridin-3-yl)-N-methyl-N-(6-methyl-6H-thieno[2,3-fe]pyrrol-5- ylmethyPacrylamide;
(E)-N-Methyl-N-(l-methyl-lH-indol-2-ylmethyl)-3-(2-oxo-l,4-dihydro-2/ - pyrido[2,3-d]- 1 ,3-oxazin-6-yl)acrylamide;
(E)-3-[6-( 1 ,3-dioxo- 1 ,3-dihydroisoindol-2-yl)pyridin-3-y l]-N-methyl-N-( 1 -methy 1- 1 //-indol-2-ylmethy Pacrylamide; (E)-3-[6-[(2-Carboxybenzoyl)amino]pyridin-3-yl]-N-methyl-N-(l-methyl-lH- indol-2-ylmethyl)acrylamide;
(E)-3-[6-(3-Ethylureido)pyridin-3-yl]-N-methyl-N-(l-methyl-lH-indol-2- ylmethyl)acrylamide;
(E)-N-(l,3-Dimethyl-lH-indol-2-ylmethyl)-N-methyl-3-(7-oxo-5,6,7,8-tetrahydro- 1 ,8-naphthyridin-3-yl)acrylamide;
(E)-3-(6-Aminopyridin-3-yl)-N-methyl-N-(3-methylbenzo[o]thiophen-2- ylmethyl)acrylamide;
(E)-3-(6-Aminopyridin-3-yl)-N-(4-methoxy-l-methyl-lH-indol-2-ylmethyl)-N- methylacrylamide; (E)-3-t6-(Acetylamino)pyridin-3-yl]-N-methyl-N-(3-methyl-lH-inden-2- ylmethyl)acrylamide;
(E)-3-[6-(Acetylamino)pyridin-3-yl]-N-methyl-N-(l-methyl-lH-indol-3- ylmethyPacrylamide;
(E)-N-Methyl-N-(l-methyl-lH-indol-2-ylmethyl)-3-(3-methyl-2-oxo-l,2,3,4- tetrahydropyrido[2,3--i]pyrimidin-6-yl)acrylamide;
(E)-N-Methyl-N-(l-methyl-lH-indol-2-ylmethyl)-3-[6-(propionylamino)pyridin-3- yl]acrylamide;
(E)-3-(6- Aminopyridin-3-y l)-N-( 1 ,4-dimethyl- 1 H-indol-2-ylmethy 1)-N- methylacrylamide;
(E)-N-methyl-N-(l-methyl-lH-indol-2-ylmethyl)-3-[6-(3-methylureido)pyridin-3- yl]acrylamide;
(E)-N-Methyl-N-(3-methyl-l/ -inden-2-ylmethyl)-3-(7-oxo-5,6,7,8-tetrahydro-l,8- naphthyridin-3-yl)acrylamide;
(E)-3-(6-Aminopyridin-3-yl)-N-methyl-N-(4-methyl-4//-theino[3,2-o]pyrrol-5- ylmethyPacrylamide; (E)-3-(6-Aminopyridin-3-yl)-N-(3,4-dimethylthieno[2,3-£]thiophen-2-ylmethyl)-
N-methylacrylamide;
(E)-N-Methyl-N-(l-methyl- lH-indol-2-ylmethyl)-3-[6-(phenylamino)pyridin-3- yljacrylamide;
(E)-3-(6-Aminopyridin-3-yl)-N-(6-methoxy-l-methyl-lH-indol-2-ylmethyl)-N- methylacrylamide;
(E)-3-(2-Aminopyrimidin-5-yl)-N-(benzo[b]thiophen-2-ylmethyl)-N- methylacrylamide;
(E)-3-(2-Aminopyrimidin-5-yl)-N-methyl-N-(l-methyl-lH-indol-3- ylmethyPacrylamide; (E)-3-(6- Aminopyridin-3-y l)-N-methy l-N-( 1 -methylnaphthalen-2- ylmethyPacrylamide;
(E)-3-(6-Aminopyridin-3-yl)-N-( 1 ,2-dimethyl- lH-indol-3-ylmethyl)-N- methylacrylamide;
(E)-3-(6-Aminopyridin-3-yl)-N-(benzo[fe]thiophen-3-ylmethyl)-N- methylacrylamide;
(E)-N-Methyl-N-(l-methyl-l//-indol-3-ylmethyl)-3-(7-oxo-5,6,7,8-tetrahydro- l ,8- naphthyridin-3-yl)acry lamide;
(E)-3-[2-Aminopyrimidin-5-yl]-N-methyl-N-(3-methyl-lH-inden-2- ylmethyPacry lamide; (E)-N-Methyl-N-(l-methyl-l/ -indol-2-ylmethyl)-3-[6-(pyridin-2-ylamino)pyridin-
3-yl]acrylamide;
(E)-3-[2-(Acetylamino)pyrimidin-5-yl]-N-methyl-N-(l-methyl-lH-indόl-2- ylmethyPacrylamide;
(E)-3-(6-Aminopyridin-3-yl)-N-methyl-N-(2-methyl-lH-indol-3- ylmethyPacrylamide;
(E)-3-(2-Aminopyrimidin-5-yl)-N-( 1 ,2-dimethyl- lH-indol-3-ylmethyl)-N- methylacrylamide;
(E)-N-(l,2-Dimethyl-l/ -indol-3-ylmethyl)-N-methyl-3-(7-oxo-5,6,7,8-tetrahydro- l,8-naphthyridin-3-yl)acrylamide;
(E)-N-Methyl-N-( l-methyl-lH-indol-2-ylmethyl)-3-(3-oxo-3,4-dihydro-2//- pyrido[3,2-i>]- l,4-oxazin-7-yl)acrylamide;
(E)-N-Methyl-N-(3-methylbenzo[/j]thiophen-2-ylmethyl)-3-(7-oxo-5,6,7,8- tetrahydro- 1 ,8-naphthyridin-3-yl)acrylamide;
(E)-3-(6-Aminopyridin-3-yl)-N-methyl-N-(l-methyl-lH-pyrrolo[2,3- ?]pyridin-3- ylmethyPacrylamide; (E)-3-(6-Aminopyridin-3-yl)-N-(l,7-dimethyl-lH-indol-3-ylmethyl)-N- methylacrylamide;
(E)-3-(6- Aminopyridin-3-yl)-N-( 1 ,5-dimethyl- 1 H-indol-3-ylmethy 1)-N- methylacry lamide ;
E)-N-Methyl-N-(2-methyl- lH-indol-3-ylmethyl)-3-(7-oxo-5,6,7,8-tetrahydro- 1 ,8- naphthyridin-3-yl)acrylamide;
(E)-3-(6-Aminopyridin-3-yl)-N-( 1 ,6-dimethyl- 1 H-indol-3-y lmethyl)-N- methylacrylamide;
(E)-3-(6-Aminopyridin-3-yl)-N-(2,3-dihydro-lH-3a-azacyclopenta[ ]inden-8-yl)- N-methylacrylamide; (E)-3-(6-Aminopyridin-3-yl)-N-methyl-(2-methylbenzo[/»]thiophen-3- ylmethyPacrylamide;
(E)-3-(6-Aminopyridin-3-yl)-N-(l-benzyl-lH-indol-3-ylmethyl)-N- methylacrylamide;
(E)-N-Methyl-N-(l -methyl- lH-indol-3-ylmethyl)-3-(3-oxo-3,4-dihydro-2H- pyrido[3,2-Z?]-l,4-oxazin-7-yl)acrylamide;or
(E)-N-Methyl-N-(l-methyl-lH-indol-2-ylmethyl)-3-(7-oxo-5,6,7,8-tetrahydro- l ,8- naphthyridin-3-yl)propionamide; or a pharmaceutically acceptable salt thereof.
10. A pharmaceutical composition which comprises a compound according to claim 1 and a pharmaceutically acceptable carrier.
11. A method for inhibiting Fab I which comprises administering to a subject in need thereof a compound according to claim 1.
12. A method for inhibiting Fab K which comprises administering to a subject in need thereof:
(E)-N-methyl-N-(l-methyl- l/ -indol-3-ylmethyl)-3-(7-oxo-5,6,7,8-tetrahydro-l,8- naphthyridin-3-yl)acrylamide; or (E)-N-methyl-N-(2-methyl-l//-indol-3-ylmethyl)-3-(7-oxo-5,6,7,8-tetrahydro-l,8- naphthyridin-3-yl)acrylamide; or a pharmaceutically acceptable salt thereof.
13. A method of treating bacterial infections which comprises administering to a subject in need thereof a compound according to claim 1.
14. A compound according to any one of claims 1 to 9 for use as a medicament.
15. The use of a compound of the formula (I) as defined in claim 1 in the manufacture of a medicament for the treatment of bacterial infections.
16. The use of a compound of the formula (I) as defined in claim 1 in the manufacture of a medicament for the treatment of diseases in which inhibition of Fab I is indicated.
17. The use of (E)-N-methyl-N-( 1 -methy 1- 1 H-indol-3-ylmethy l)-3-(7-oxo- 5,6,7, 8-tetrahydro-l,8-naphthyridin-3-yl)acrylamide or (E)-N-methyl-N-(2- methyl- 1H- indol-3-ylmethyl)-3-(7-oxo-5,6,7,8-tetrahydro- 1 ,8-naphthyridin-3-yl)acrylamide in the manufacture of a medicament for the treatment of diseases in which inhibition of Fab K is indicated.
18. A process for preparing compounds of formula (I) as defined in claim 1, which process comprises: (i) reacting a compound of formula (III) with a compound of formula (IV):
Figure imgf000115_0001
wherein R2, R3, R4, R^ and X are as defined in formula (I), with any reactive functional groups protected, in the presence of EDC and HOBT;
(ii) reacting a compound of formula (V) with a compound of formula (VI):
Figure imgf000116_0001
wherein R2, R3 and X are as defined in formula (I) and Halo is Br, Cl, F or I, with any reactive functional groups protected, in the presence of a palladium (II) salt, a phosphine ligand and base; and thereafter removing any protecting groups, and optionally forming a pharmaceutically acceptable salt.
19. A process for preparing crotonoyl-ACP, which process comprises reacting crotonoyl CoA and apo-acyl carrier protein, in the presence of S. pneumoniae ACP synthase.
20. A method of screening for an inhibitor of Fab I or FabK which comprises: (i) providing a reaction mixture comprising Fab I or Fab K, crotonoyl-ACP and NADPH or NADH;
(ii) contacting a candidate compound to said reaction mixture; and (iii) detecting inhibition of Fab I or Fab K.
PCT/US2000/027844 1999-10-08 2000-10-06 Fab i inhibitors WO2001027103A1 (en)

Priority Applications (18)

Application Number Priority Date Filing Date Title
IL14882000A IL148820A0 (en) 1999-10-08 2000-10-06 Fab i inhibitors
AU78747/00A AU773218B2 (en) 1999-10-08 2000-10-06 Fab I inhibitors
AT00968895T ATE285821T1 (en) 1999-10-08 2000-10-06 FAB I INHIBITORS
CA2387016A CA2387016C (en) 1999-10-08 2000-10-06 Acrylamide derivatives as fab i inhibitors
NZ517706A NZ517706A (en) 1999-10-08 2000-10-06 Fab I inhibitors
HU0203122A HU230030B1 (en) 1999-10-08 2000-10-06 Fab i inhibitors
EP00968895A EP1226138B1 (en) 1999-10-08 2000-10-06 Fab i inhibitors
US10/089,755 US6846819B1 (en) 1999-10-08 2000-10-06 Fab I inhibitors
BRPI0014470A BRPI0014470B1 (en) 1999-10-08 2000-10-06 fab i inhibitor compounds, pharmaceutical composition, compound preparation process and use of the compounds for the manufacture of medicaments for treating bacterial infections
JP2001530321A JP4803935B2 (en) 1999-10-08 2000-10-06 FABI inhibitor
DE60017180T DE60017180T2 (en) 1999-10-08 2000-10-06 FAB I INHIBITORS
IL148820A IL148820A (en) 1999-10-08 2002-03-21 Fab i inhibitors, process for their preparation, pharmaceutical compositions containing them and the use thereof in the preparation of medicaments for treating bacterial infections
NO20021638A NO322708B1 (en) 1999-10-08 2002-04-05 FAB I inhibitors, pharmaceutical composition comprising the same, use of the same for the preparation of drug, and method for the preparation of the same.
HK03100341.3A HK1049656A1 (en) 1999-10-08 2003-01-14 Fab i inhibitors
US11/007,927 US7557125B2 (en) 1999-10-08 2004-12-09 Fab I inhibitors
US11/779,578 US7524843B2 (en) 1999-10-08 2007-07-18 Fab I inhibitors
US12/403,078 US7790716B2 (en) 1999-10-08 2009-03-12 Fab I inhibitors
US12/830,729 US8173646B2 (en) 1999-10-08 2010-07-06 FAB I inhibitors

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US15870499P 1999-10-08 1999-10-08
US60/158,704 1999-10-08

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US10089755 A-371-Of-International 2000-10-06
US11/007,927 Continuation US7557125B2 (en) 1999-10-08 2004-12-09 Fab I inhibitors

Publications (1)

Publication Number Publication Date
WO2001027103A1 true WO2001027103A1 (en) 2001-04-19

Family

ID=22569335

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2000/027844 WO2001027103A1 (en) 1999-10-08 2000-10-06 Fab i inhibitors

Country Status (25)

Country Link
US (5) US6846819B1 (en)
EP (1) EP1226138B1 (en)
JP (1) JP4803935B2 (en)
KR (1) KR100823382B1 (en)
CN (1) CN1197860C (en)
AR (1) AR025976A1 (en)
AT (1) ATE285821T1 (en)
AU (1) AU773218B2 (en)
BR (1) BRPI0014470B1 (en)
CA (1) CA2387016C (en)
CZ (1) CZ302015B6 (en)
DE (1) DE60017180T2 (en)
EC (1) ECSP003699A (en)
ES (1) ES2231275T3 (en)
HK (1) HK1049656A1 (en)
HU (1) HU230030B1 (en)
IL (2) IL148820A0 (en)
NO (1) NO322708B1 (en)
NZ (1) NZ517706A (en)
PE (1) PE20010635A1 (en)
PL (1) PL201627B1 (en)
TW (1) TW534909B (en)
UY (1) UY26380A1 (en)
WO (1) WO2001027103A1 (en)
ZA (1) ZA200202631B (en)

Cited By (45)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002024693A1 (en) * 2000-09-22 2002-03-28 Bayer Aktiengesellschaft A process for producing 3,4-dihydro naphthyridinone analogs
EP1225894A1 (en) * 1999-10-08 2002-07-31 Smithkline Beecham Corporation Fab i inhibitors
EP1330515A1 (en) * 2000-10-06 2003-07-30 SmithKline Beecham Corporation Methods of agonizing and antagonizing fabk
WO2003088897A2 (en) * 2001-04-06 2003-10-30 Affinium Pharmaceuticals, Inc. Fab i inhibitors
US6730684B1 (en) 1999-10-08 2004-05-04 Affinium Pharmaceuticals, Inc. Fab I inhibitors
WO2004052890A1 (en) 2002-12-06 2004-06-24 Affinium Pharmaceuticals, Inc. Heterocyclic compounds, methods of making them and their use in therapy
US6762201B1 (en) 1999-10-08 2004-07-13 Affinium Pharmaceuticals, Inc. Fab I inhibitors
US6765005B2 (en) 1999-04-19 2004-07-20 Affinium Pharmaceuticals, Inc. Fab I inhibitors
WO2004082586A2 (en) * 2003-03-17 2004-09-30 Affinium Pharmaceuticals, Inc. Phamaceutical compositions comprising inhibitors of fab i and further antibiotics
US6846819B1 (en) 1999-10-08 2005-01-25 Affinium Pharmaceuticals, Inc. Fab I inhibitors
US6964970B2 (en) 1999-06-01 2005-11-15 Affinium Pharmaceuticals, Inc. Antibacterial compounds
WO2006021277A1 (en) 2004-08-21 2006-03-02 Merck Patent Gmbh MONOMERS, OLIGOMERS AND POLYMERS OF THIENO[2,3-b]THIOPHENE
US7048926B2 (en) 2000-10-06 2006-05-23 Affinium Pharmaceuticals, Inc. Methods of agonizing and antagonizing FabK
WO2007043835A1 (en) 2005-10-13 2007-04-19 Crystalgenomics, Inc. Fab i inhibitor and process for preparing same
WO2007067416A2 (en) * 2005-12-05 2007-06-14 Affinium Pharmaceuticals, Inc. Heterocyclylacrylamide compounds as fabi inhibitors and antibacterial agents
WO2007053131A3 (en) * 2004-06-04 2007-08-02 Affinium Pharm Inc Acrylamide derivatives as antibiotic agents
WO2008009122A1 (en) 2006-07-20 2008-01-24 Affinium Pharmaceuticals, Inc. Acrylamide derivatives as fab i inhibitors
EP2014287A1 (en) * 2007-06-13 2009-01-14 Bayer Schering Pharma Aktiengesellschaft Use of cinnamic acid derivatives as modulators of an EP2 receptor
US7884124B2 (en) 2006-06-30 2011-02-08 Sepracor Inc. Fluoro-substituted inhibitors of D-amino acid oxidase
US7893098B2 (en) 2003-12-29 2011-02-22 Sepracor Inc. Pyrrole and pyrazole DAAO inhibitors
EP2286808A1 (en) * 2009-08-18 2011-02-23 Rheinische Friedrich-Wilhelms Universität Cytohesin inhibitors
US7902252B2 (en) 2007-01-18 2011-03-08 Sepracor, Inc. Inhibitors of D-amino acid oxidase
WO2011061214A1 (en) 2009-11-18 2011-05-26 Fab Pharma Sas Novel heterocyclic acrylamides and their use as pharmaceuticals
US8053603B2 (en) 2006-01-06 2011-11-08 Sunovion Pharmaceuticals Inc. Tetralone-based monoamine reuptake inhibitors
US8097760B2 (en) 2006-03-31 2012-01-17 Sunovion Pharmacuticals Inc. Preparation of chiral amides and amines
WO2013021051A1 (en) 2011-08-10 2013-02-14 Janssen R&D Ireland Antibacterial homopiperidinyl substituted 3,4 dihydro 1h [1,8]naphthyridinones
WO2013021052A1 (en) 2011-08-10 2013-02-14 Janssen R&D Ireland Antibacterial piperidinyl substituted 3,4-dihydro-1h-[1,8]naphthyridinones
WO2013021054A1 (en) 2011-08-10 2013-02-14 Janssen R&D Ireland Antibacterial cyclopenta[c]pyrrole substituted 3,4-dihydro-1h-[1,8]naphthyridinones
WO2014023815A1 (en) 2012-08-10 2014-02-13 Janssen R&D Ireland New antibacterial compounds
US8669291B2 (en) 2007-05-31 2014-03-11 Sunovion Pharmaceuticals Inc. Phenyl substituted cycloalkylamines as monoamine reuptake inhibitors
WO2014072930A2 (en) * 2012-11-09 2014-05-15 Aurigene Discovery Technologies Limited Fused pyridine derivatives as antibacterial agents
US8877975B2 (en) 2006-01-06 2014-11-04 Sunovion Pharmaceuticals Inc. Cycloalkylamines as monoamine reuptake inhibitors
US8901105B2 (en) 2012-06-19 2014-12-02 Debiopharm International Sa Prodrug derivatives of (E)-N-methyl-N-((3-M ethylbenzofuran-2-yl)methyl)-3-(7-oxo-5,6,7,8-tetrahydro-1,8-naphthyridin-3-yl)acrylamide
EP2758376A4 (en) * 2011-09-19 2015-04-29 Vitas Pharma Res Private Ltd Heterocyclic compounds as inhibitors of fatty acid biosynthesis for bacterial infections
US9315522B2 (en) 2012-08-10 2016-04-19 Janssen Sciences Ireland Uc Antibacterial compounds
US9394287B2 (en) 2010-11-05 2016-07-19 Senomyx, Inc. Compounds useful as modulators of TRPM8
US9732071B2 (en) 2015-10-01 2017-08-15 Senomyx, Inc. Compounds useful as modulators of TRPM8
US9862737B2 (en) 2007-02-16 2018-01-09 Debiopharm International Sa Salts, prodrugs and polymorphs of fab I inhibitors
US10167257B2 (en) 2014-04-04 2019-01-01 Iomet Pharma Ltd. Indole derivatives for use in medicine
WO2020099341A1 (en) 2018-11-12 2020-05-22 Debiopharm International S.A. Antibiotic compounds, methods of manufacturing the same, pharmaceutical compositions containing the same and uses thereof
US10751351B2 (en) 2016-02-26 2020-08-25 Debiopharm International S.A. Medicament for treatment of diabetic foot infections
US10858319B2 (en) 2016-10-03 2020-12-08 Iomet Pharma Ltd. Indole derivatives for use in medicine
US11667651B2 (en) 2017-12-22 2023-06-06 Hibercell, Inc. Aminopyridine derivatives as phosphatidylinositol phosphate kinase inhibitors
US11691967B2 (en) 2018-03-12 2023-07-04 The Board Of Trustees Of The University Of Illinois Antibiotics effective for gram-negative pathogens
US12006332B2 (en) 2019-06-17 2024-06-11 Hibercell, Inc. Aminopyrimidine derivatives as phosphatidylinositol phosphate kinase inhibitors

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007086584A1 (en) * 2006-01-30 2007-08-02 Meiji Seika Kaisha, Ltd. NOVEL INHIBITOR OF FabK AND FabI/K
EP2579863A4 (en) * 2010-06-11 2013-11-27 Affinium Pharm Inc Compounds for treatment of bovine mastitis
AR088729A1 (en) * 2011-03-29 2014-07-02 Actelion Pharmaceuticals Ltd DERIVATIVES OF 3-UREIDOISOQUINOLIN-8-ILO AND A PHARMACEUTICAL COMPOSITION
CN102675311A (en) * 2011-06-14 2012-09-19 苏春华 Fluoro-acrylamide derivative
US9062075B2 (en) 2011-12-02 2015-06-23 Aurigene Discovery Technologies Limited Tetrahydropyridine derivatives as FabI inhibitors
US9062002B2 (en) 2011-12-02 2015-06-23 Aurigene Discovery Technologies Limited Substituted pyridine derivatives as FabI inhibitors
WO2014195844A1 (en) 2013-06-04 2014-12-11 Aurigene Discovery Technologies Limited TETRAHYDROPYRIDINE DERIVATIVES AS FabI INHIBITORS
CN105669519B (en) * 2016-01-04 2018-01-05 北方民族大学 Benzazole compounds, preparation method and its application as drug-resistance bacteria medicine

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0407200A1 (en) * 1989-07-05 1991-01-09 Kanegafuchi Kagaku Kogyo Kabushiki Kaisha Cinnamamide derivatives
WO1999024406A1 (en) * 1997-11-07 1999-05-20 Schering Corporation Phenyl-alkyl-imidazoles as h3 receptor antagonists

Family Cites Families (120)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3828068A (en) 1971-05-10 1974-08-06 Tenneco Chem ((substituted indazolyl)-n1-methyl)carbamates
US4154943A (en) 1977-12-29 1979-05-15 University Of Vermont Preparation of vincadifformine
FR2619111B1 (en) 1987-08-07 1991-01-11 Synthelabo (PIPERIDINYL-4) METHYL-2 TETRAHYDRO-1,2,3,4 9H-PYRIDO (3,4-B) INDOLE DERIVATIVES, THEIR PREPARATION AND THEIR THERAPEUTIC APPLICATION
HU210679B (en) 1991-11-21 1995-06-28 Richter Gedeon Vegyeszet Process for producing new tetrahydro-pyrido/3,4-b/indol derivatives and pharmaceutical compositions containing the same
FR2692575B1 (en) 1992-06-23 1995-06-30 Sanofi Elf NOVEL PYRAZOLE DERIVATIVES, PROCESS FOR THEIR PREPARATION AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM.
US5416193A (en) 1993-04-30 1995-05-16 Pfizer Inc. Coupling reagent and method
MA23420A1 (en) 1994-01-07 1995-10-01 Smithkline Beecham Corp BICYCLIC FIBRINOGEN ANTAGONISTS.
US5614551A (en) 1994-01-24 1997-03-25 The Johns Hopkins University Inhibitors of fatty acid synthesis as antimicrobial agents
BR9508178A (en) 1994-06-29 1997-11-18 Smithkline Beecham Corp Vitronectin receptor antagonists
US6176842B1 (en) 1995-03-08 2001-01-23 Ekos Corporation Ultrasound assembly for use with light activated drugs
US5989832A (en) 1995-04-21 1999-11-23 Microcide Pharmaceuticals, Inc. Method for screening for non-tetracycline efflux pump inhibitors
DE69634581T2 (en) 1995-05-11 2006-02-09 Sandoz Ag Antibacterial cephalosporins
US6057291A (en) 1995-06-02 2000-05-02 University Of British Columbia Antimicrobial cationic peptides
WO1997031639A1 (en) 1996-02-29 1997-09-04 Fujisawa Pharmaceutical Co., Ltd. TABLETS CONTAINING β-LACTAM ANTIBIOTIC AND PROCESS FOR PRODUCING THE SAME
US6239154B1 (en) 1996-03-08 2001-05-29 Adolor Corporation Kappa agonist compounds pharmaceutical formulations and method of prevention and treatment of pruritus therewith
US6367985B1 (en) 1996-03-12 2002-04-09 Intellectual Property Company Optical connector using large diameter alignment features
US6451816B1 (en) 1997-06-20 2002-09-17 Klinge Pharma Gmbh Use of pyridyl alkane, pyridyl alkene and/or pyridyl alkine acid amides in the treatment of tumors or for immunosuppression
DE19624659A1 (en) 1996-06-20 1998-01-08 Klinge Co Chem Pharm Fab New pyridylalkene and pyridylalkanoic acid amides
US6503881B2 (en) 1996-08-21 2003-01-07 Micrologix Biotech Inc. Compositions and methods for treating infections using cationic peptides alone or in combination with antibiotics
US6995254B1 (en) 1996-08-28 2006-02-07 Affinium Pharmaceuticals, Inc. Polynucleotide encoding the enoyl-acyl carrier protein reductase of Staphylococcus aureus, FAB I
ES2198593T3 (en) 1996-09-20 2004-02-01 Meiji Seika Kaisha, Ltd. A CRYSTAL SUBSTANCE OF CEFDITOREN PIVOXIL AND THE PRODUCTION OF THE SAME.
US6521408B1 (en) 1997-09-25 2003-02-18 National Institute Of Agrobiological Sciences Method for assessing a function of a gene
DE19641437A1 (en) 1996-10-08 1998-04-09 Basf Ag 1,3-bis (N-lactamyl) propanes and their pharmaceutical and cosmetic use
DE19652239A1 (en) 1996-12-16 1998-06-18 Bayer Ag Use of 7- (2-oxa-5,8-diazabicyclo [4.3.0] non-8-yl) -quinolone and naphthyridonecarboxylic acid derivatives for the therapy of Helicobacter pylori infections and the associated gastroduodenal diseases
SI9600371B (en) 1996-12-18 2005-04-30 LEK, tovarna farmacevtskih in kemi�nih izdelkov, d.d. Ethylidene derivatives of tricyclic carbapenemes
WO1998043969A1 (en) 1997-03-31 1998-10-08 Dupont Pharmaceuticals Company Indazoles of cyclic ureas useful as hiv protease inhibitors
JP2001524951A (en) 1997-04-01 2001-12-04 トーマス ジュリアス ボロディー Methods and compositions for treating inflammatory bowel disease
US6406880B1 (en) 1997-05-02 2002-06-18 Integrated Research Technology, Llc Betaines as adjuvants to susceptibility testing and antimicrobial therapy
US6184363B1 (en) 1997-06-13 2001-02-06 Northwestern University Inhibitors of β-lactamases and uses therefor
US6207679B1 (en) 1997-06-19 2001-03-27 Sepracor, Inc. Antimicrobial agents uses and compositions related thereto
CA2294453A1 (en) 1997-06-23 1998-12-30 Fumihiro Hirayama Prophylactic or therapeutic agent for diseases attributable to infection with helicobacters
AUPO758297A0 (en) 1997-06-27 1997-07-24 Rowe, James Baber Control of acidic gut syndrome
US6198000B1 (en) 1997-07-07 2001-03-06 Pfizer Inc. Intermediates useful in the synthesis of quinoline antibiotics
EP1000935A4 (en) 1997-07-25 2001-03-21 Tsumura & Co Pyridylacrylamide derivatives and nephritis remedies and tgf-beta inhibitors containing the same
HN1998000106A (en) 1997-08-01 1999-01-08 Pfizer Prod Inc PARENTERAL COMPOSITIONS OF ALATROFLAXACINO
US5932743A (en) 1997-08-21 1999-08-03 American Home Products Corporation Methods for the solid phase synthesis of substituted indole compounds
GB9717804D0 (en) 1997-08-22 1997-10-29 Zeneca Ltd Chemical compounds
CZ300208B6 (en) 1997-10-31 2009-03-18 Syngenta Participations Ag Transgenic plant tolerant to glyphosate herbicide and process for preparing such plant
US6432444B1 (en) 1997-10-31 2002-08-13 New Horizons Diagnostics Corp Use of bacterial phage associated lysing enzymes for treating dermatological infections
SE9704404D0 (en) 1997-11-28 1997-11-28 Astra Ab New compounds
GB9725244D0 (en) 1997-11-29 1998-01-28 Zeneca Ltd Chemical compounds
DE19753298A1 (en) 1997-12-01 1999-06-02 Basf Ag Process for the preparation of solid dosage forms
TR200001903T2 (en) 1998-01-07 2000-11-21 Meiji Seika Kaisha, Ltd. Crystallographically stable amorphous cephalosporin compositions and process for their production.
US6184380B1 (en) 1999-01-25 2001-02-06 Pfizer Inc. Process for preparing naphthyridones and intermediates
PA8466701A1 (en) 1998-01-21 2000-09-29 Pfizer Prod Inc TROVAFLOXACINO MESYLATE TABLET
US6204279B1 (en) 1998-06-03 2001-03-20 Microcide Pharmaceuticals, Inc. Peptidomimetic efflux pump inhibitors
US6503539B2 (en) 1998-02-27 2003-01-07 Biora Bioex Ab Matrix protein compositions for wound healing
US6350738B1 (en) 1998-03-06 2002-02-26 Brigham Young University Steroid derived antibiotics
DE19820801A1 (en) 1998-05-09 1999-11-25 Gruenenthal Gmbh Oral dosage form for gatifloxacin, providing reproducible decomposition time and drug release
DE19821039A1 (en) 1998-05-11 1999-11-18 Bayer Ag Production of (S,S)-8-benzyl-2,8-diazabicyclo(4.3.0)nonane useful as intermediate for antibiotic moxifloxacin
US6399629B1 (en) 1998-06-01 2002-06-04 Microcide Pharmaceuticals, Inc. Efflux pump inhibitors
US6428579B1 (en) 1998-07-01 2002-08-06 Brown University Research Foundation Implantable prosthetic devices coated with bioactive molecules
US6423741B1 (en) 1998-07-10 2002-07-23 Council Of Scientific And Industrial Research Anti-microbial composition and method for producing the same
GB9815567D0 (en) 1998-07-18 1998-09-16 Glaxo Group Ltd Antiviral compound
EP1101497B1 (en) 1998-08-04 2008-02-20 Takeda Schering-Plough Animal Health K.K. Stabilized oily preparations of tobicillin (beta-lactam antibiotic)
ATE332692T1 (en) 1998-08-21 2006-08-15 Kyorin Seiyaku Kk AQUEOUS LIQUID PREPARATIONS
US6461607B1 (en) 1998-08-24 2002-10-08 Ganeden Biotech, Inc. Probiotic, lactic acid-producing bacteria and uses thereof
US6518487B1 (en) 1998-09-23 2003-02-11 Pioneer Hi-Bred International, Inc. Cyclin D polynucleotides, polypeptides and uses thereof
US6395746B1 (en) 1998-09-30 2002-05-28 Alcon Manufacturing, Ltd. Methods of treating ophthalmic, otic and nasal infections and attendant inflammation
US6509327B1 (en) 1998-09-30 2003-01-21 Alcon Manufacturing, Ltd. Compositions and methods for treating otic, ophthalmic and nasal infections
TW526202B (en) 1998-11-27 2003-04-01 Shionogi & Amp Co Broad spectrum cephem having benzo[4,5-b]pyridium methyl group of antibiotic activity
CA2371816C (en) 1999-02-18 2010-04-27 The Regents Of The University Of California Phthalamide-lanthanide complexes for use as luminescent markers
US6248363B1 (en) 1999-11-23 2001-06-19 Lipocine, Inc. Solid carriers for improved delivery of active ingredients in pharmaceutical compositions
US6267985B1 (en) 1999-06-30 2001-07-31 Lipocine Inc. Clear oil-containing pharmaceutical compositions
US6294192B1 (en) 1999-02-26 2001-09-25 Lipocine, Inc. Triglyceride-free compositions and methods for improved delivery of hydrophobic therapeutic agents
US6461829B1 (en) 1999-03-03 2002-10-08 The Trustees Of Princeton University Bacterial transglycosylases: assays for monitoring the activity using Lipid II substrates analogs and methods for discovering new antibiotics
US6495161B1 (en) 1999-03-09 2002-12-17 Vivorx, Inc. Cytoprotective biocompatible containment systems for biologically active materials and methods of making same
US6239113B1 (en) 1999-03-31 2001-05-29 Insite Vision, Incorporated Topical treatment or prevention of ocular infections
AU4230900A (en) 1999-04-08 2000-10-23 General Hospital Corporation, The Purposeful movement of human migratory cells away from an agent source
CO5180550A1 (en) * 1999-04-19 2002-07-30 Smithkline Beecham Corp FAB I INHIBITORS
US6290946B1 (en) 1999-05-13 2001-09-18 Geltex Pharmaceuticals, Inc. Anionic polymers as toxin binders and antibacterial agents
US6514535B2 (en) 1999-05-21 2003-02-04 Noveon Ip Holdings Corp. Bioadhesive hydrogels with functionalized degradable crosslinks
AR024077A1 (en) 1999-05-25 2002-09-04 Smithkline Beecham Corp ANTIBACTERIAL COMPOUNDS
CO5370679A1 (en) * 1999-06-01 2004-02-27 Smithkline Beecham Corp FAB INHIBITORS 1
US6239141B1 (en) 1999-06-04 2001-05-29 Pfizer Inc. Trovafloxacin oral suspensions
CO5180605A1 (en) 1999-06-23 2002-07-30 Smithkline Beecham Corp INDOL COMPOUNDS
CO5190665A1 (en) * 1999-06-23 2002-08-29 Smithkline Beecham Corp INDOL COMPOUNDS
CA2282066C (en) 1999-06-29 2010-09-07 Smithkline Beecham Corporation Methods of use of quinolone compounds against atypical upper respiratory pathogenic bacteria
US6309663B1 (en) 1999-08-17 2001-10-30 Lipocine Inc. Triglyceride-free compositions and methods for enhanced absorption of hydrophilic therapeutic agents
US6500459B1 (en) 1999-07-21 2002-12-31 Harinderpal Chhabra Controlled onset and sustained release dosage forms and the preparation thereof
US6346391B1 (en) 1999-07-22 2002-02-12 Trustees Of Tufts College Methods of reducing microbial resistance to drugs
EP1206464A4 (en) * 1999-08-23 2002-12-18 Smithkline Beecham Corp Fatty acid synthase inhibitors
WO2001013956A2 (en) 1999-08-26 2001-03-01 Ganeden Biotech, Inc. Use of emu oil a carrier for antifungal, antibacterial, and antiviral medications
US6221859B1 (en) 1999-08-27 2001-04-24 Merck & Co., Inc. Carbapenem antibacterial compositions and methods of the treatment
US6174878B1 (en) 1999-08-31 2001-01-16 Alcon Laboratories, Inc. Topical use of kappa opioid agonists to treat otic pain
AU770516B2 (en) 1999-09-11 2004-02-26 Procter & Gamble Company, The Polyoxyalkylene copolymers containing pourable liquid vehicles
DE60025503T2 (en) 1999-09-17 2006-09-07 Daiichi Asubio Pharma Co., Ltd. Pharmaceutical compositions containing faropenem sodium and a diamine acetate compound for improving gastrointestinal absorption
US6500463B1 (en) 1999-10-01 2002-12-31 General Mills, Inc. Encapsulation of sensitive components into a matrix to obtain discrete shelf-stable particles
US6762201B1 (en) * 1999-10-08 2004-07-13 Affinium Pharmaceuticals, Inc. Fab I inhibitors
WO2001026654A1 (en) 1999-10-08 2001-04-19 Smithkline Beecham Corporation Fab i inhibitors
WO2001027103A1 (en) 1999-10-08 2001-04-19 Smithkline Beecham Corporation Fab i inhibitors
US6730684B1 (en) * 1999-10-08 2004-05-04 Affinium Pharmaceuticals, Inc. Fab I inhibitors
CO5251397A1 (en) 1999-10-08 2003-02-28 Smithkline Beecham Corp FAB I INHIBITORS
CA2323008C (en) 1999-10-11 2005-07-12 Pfizer Inc. Pharmaceutically active compounds
ES2225237T3 (en) 1999-10-19 2005-03-16 Sato Pharmaceutical Co. Ltd. ANTIMICROBIAL AGENTS OF TYPE 4-OXOQUINOLIZINE THAT HAVE MAIN CHAINS OF 2-PIRIDONE AS A PARTIAL STRUCTURE.
US6951729B1 (en) 1999-10-27 2005-10-04 Affinium Pharmaceuticals, Inc. High throughput screening method for biological agents affecting fatty acid biosynthesis
ATE461692T1 (en) 1999-10-29 2010-04-15 Novartis Ag DRY POWDER COMPOSITIONS WITH IMPROVED DISPERSITY
US6531291B1 (en) 1999-11-10 2003-03-11 The Trustees Of Columbia University In The City Of New York Antimicrobial activity of gemfibrozil and related compounds and derivatives and metabolites thereof
US6514986B2 (en) 2000-11-22 2003-02-04 Wockhardt Limited Chiral fluoroquinolone arginine salt forms
US6656703B1 (en) 1999-12-29 2003-12-02 Millennium Pharamaceuticals, Inc. High throughput screen for inhibitors of fatty acid biosynthesis in bacteria
US6372752B1 (en) 2000-02-07 2002-04-16 Genzyme Corporation Inha inhibitors and methods of use thereof
WO2001072317A1 (en) 2000-03-28 2001-10-04 Council Of Scientific And Industrial Research Formulation comprising thymol useful in the treatment of drug resistant bacterial infections
EP1280753A1 (en) 2000-04-21 2003-02-05 Rhodia/Chirex, Inc. Process for preparation of r-1-(aryloxy)propan-2-ol
WO2002007682A1 (en) 2000-07-26 2002-01-31 Vyden John K Methods for treating atopic disorders
US6288239B1 (en) 2000-09-19 2001-09-11 Board Of Trustees Operating Michigan State University 5-trityloxymethyl-oxazolidinones and process for the preparation thereof
US7048926B2 (en) * 2000-10-06 2006-05-23 Affinium Pharmaceuticals, Inc. Methods of agonizing and antagonizing FabK
US6821746B2 (en) 2000-10-06 2004-11-23 Affinium Pharmaceuticals, Inc. Methods of screening for FabK antagonists and agonists
EP1332131A2 (en) 2000-11-07 2003-08-06 Bristol-Myers Squibb Company Acid derivatives useful as serine protease inhibitors
AU2002232558A1 (en) 2000-12-12 2002-06-24 Corvas International, Inc. Compounds, compositions and methods for treatment of parasitic infections
US6388070B1 (en) 2001-01-05 2002-05-14 Orchid Chemicals & Pharmaceuticals Ltd. Thioester derivatives of thiazolyl acetic acid and their use in the preparation of cephalosporin compounds
US6495158B1 (en) 2001-01-19 2002-12-17 Lec Tec Corporation Acne patch
EP1560584B1 (en) 2001-04-06 2009-01-14 Affinium Pharmaceuticals, Inc. Fab i inhibitors
US6573941B1 (en) * 2002-04-22 2003-06-03 Thomson Licensing Sa Low bit rate compression format conversion for improved resolution
US6503906B1 (en) 2002-02-21 2003-01-07 Ren-Jin Lee Method for optimizing ciprofloxacin treatment of anthrax-exposed patients according to the patient's characteristics
WO2004052890A1 (en) 2002-12-06 2004-06-24 Affinium Pharmaceuticals, Inc. Heterocyclic compounds, methods of making them and their use in therapy
CA2519429C (en) 2003-03-17 2013-08-06 Affinium Pharmaceuticals, Inc. Pharmaceutical compositions comprising inhibitors of fab i and further antibiotics
PT1828167E (en) 2004-06-04 2014-10-08 Debiopharm Int Sa Acrylamide derivatives as antibiotic agents
CA2632476A1 (en) 2005-12-05 2007-06-14 Affinium Pharmaceuticals, Inc. Heterocyclylacrylamide compounds as fabi inhibitors and antibacterial agents
EP2687533B1 (en) 2006-07-20 2017-07-19 Debiopharm International SA Acrylamide derivatives as FAB I inhibitors
EP2125802A4 (en) 2007-02-16 2014-08-20 Debiopharm Int Sa Salts, prodrugs and polymorphs of fab i inhibitors

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0407200A1 (en) * 1989-07-05 1991-01-09 Kanegafuchi Kagaku Kogyo Kabushiki Kaisha Cinnamamide derivatives
WO1999024406A1 (en) * 1997-11-07 1999-05-20 Schering Corporation Phenyl-alkyl-imidazoles as h3 receptor antagonists

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
DATABASE CAPLUS [online] ASLANIAN ET AL.: "Preparation of phenylalkylimidazoles as H3 receptor antagonists", XP002936688, accession no. STN Database accession no. 1999:325910 *
DATABASE CAPLUS [online] FUSE ET AL.: "Preparation of cinnamamide derivatives as antihyperlipidemics", XP002936687, accession no. STN Database accession no. 1991:428908 *
DATABASE CAPLUS [online] STUETZ ET AL.: "Synthesis and structure-activity relationships of naftifine-related allylamine antimycotics", XP002936686, accession no. STN Database accession no. 1986:68547 *
J. MED. CHEM., vol. 29, no. 1, 1986, pages 112 - 125 *

Cited By (94)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6765005B2 (en) 1999-04-19 2004-07-20 Affinium Pharmaceuticals, Inc. Fab I inhibitors
US6964970B2 (en) 1999-06-01 2005-11-15 Affinium Pharmaceuticals, Inc. Antibacterial compounds
US6730684B1 (en) 1999-10-08 2004-05-04 Affinium Pharmaceuticals, Inc. Fab I inhibitors
EP1225894A4 (en) * 1999-10-08 2003-01-08 Smithkline Beecham Corp Fab i inhibitors
US7557125B2 (en) 1999-10-08 2009-07-07 Affinium Pharmaceuticals, Inc. Fab I inhibitors
US6762201B1 (en) 1999-10-08 2004-07-13 Affinium Pharmaceuticals, Inc. Fab I inhibitors
EP1225894A1 (en) * 1999-10-08 2002-07-31 Smithkline Beecham Corporation Fab i inhibitors
US7524843B2 (en) 1999-10-08 2009-04-28 Affinium Pharmaceuticals, Inc. Fab I inhibitors
US6846819B1 (en) 1999-10-08 2005-01-25 Affinium Pharmaceuticals, Inc. Fab I inhibitors
US6562811B1 (en) 2000-09-22 2003-05-13 Bayer Aktiengesellschaft Pyridine derivatives
US7232909B2 (en) 2000-09-22 2007-06-19 Bayer Aktiengesellschaft Pyridine derivatives
US6984649B1 (en) 2000-09-22 2006-01-10 Bayer Aktiengesellschaft Pyridine derivatives
US7435743B2 (en) 2000-09-22 2008-10-14 Bayer Aktiengesellschaft Pyridine derivatives
WO2002024693A1 (en) * 2000-09-22 2002-03-28 Bayer Aktiengesellschaft A process for producing 3,4-dihydro naphthyridinone analogs
EP1330515A4 (en) * 2000-10-06 2005-12-07 Affinium Pharm Inc Methods of agonizing and antagonizing fabk
EP1330515A1 (en) * 2000-10-06 2003-07-30 SmithKline Beecham Corporation Methods of agonizing and antagonizing fabk
US7048926B2 (en) 2000-10-06 2006-05-23 Affinium Pharmaceuticals, Inc. Methods of agonizing and antagonizing FabK
WO2003088897A3 (en) * 2001-04-06 2005-06-09 Affinium Pharm Inc Fab i inhibitors
WO2003088897A2 (en) * 2001-04-06 2003-10-30 Affinium Pharmaceuticals, Inc. Fab i inhibitors
US7250424B2 (en) 2001-04-06 2007-07-31 Affinium Pharmaceuticals, Inc. Fab I inhibitors
US7049310B2 (en) 2001-04-06 2006-05-23 Affinium Pharmaceuticals, Inc. Fab I inhibitors
JP2006513262A (en) * 2002-12-06 2006-04-20 アフィニウム ファーマシューティカルズ, インク. Heterocyclic compounds, methods for their preparation and their use in therapy
US7790709B2 (en) * 2002-12-06 2010-09-07 Affinium Pharmaceuticals, Inc. Heterocyclic compounds, methods of making them and their use in therapy
JP2012006927A (en) * 2002-12-06 2012-01-12 Affinium Pharmaceuticals Inc Heterocyclic compound, method of making the same and its use in therapy
JP4859460B2 (en) * 2002-12-06 2012-01-25 アフィニウム ファーマシューティカルズ, インク. Heterocyclic compounds, methods for their preparation and their use in therapy
WO2004052890A1 (en) 2002-12-06 2004-06-24 Affinium Pharmaceuticals, Inc. Heterocyclic compounds, methods of making them and their use in therapy
JP2006523207A (en) * 2003-03-17 2006-10-12 アフィナム ファーマシューティカルズ,インコーポレーテッド Composition comprising a plurality of antibiotics and method of using the same
WO2004082586A3 (en) * 2003-03-17 2004-12-23 Affinium Pharm Inc Phamaceutical compositions comprising inhibitors of fab i and further antibiotics
JP4880448B2 (en) * 2003-03-17 2012-02-22 アフィナム ファーマシューティカルズ,インコーポレーテッド Composition comprising a plurality of antibiotics and method of using the same
WO2004082586A2 (en) * 2003-03-17 2004-09-30 Affinium Pharmaceuticals, Inc. Phamaceutical compositions comprising inhibitors of fab i and further antibiotics
US7893098B2 (en) 2003-12-29 2011-02-22 Sepracor Inc. Pyrrole and pyrazole DAAO inhibitors
EP2848614A3 (en) * 2004-06-04 2015-07-29 Debiopharm International SA Acrylamide derivatives as antibiotic agents
WO2007053131A3 (en) * 2004-06-04 2007-08-02 Affinium Pharm Inc Acrylamide derivatives as antibiotic agents
KR101299399B1 (en) * 2004-06-04 2013-08-22 아피늄 파마슈티컬스, 인크. Therapeutic Agents, and Methods of Making and Using the Same
WO2006021277A1 (en) 2004-08-21 2006-03-02 Merck Patent Gmbh MONOMERS, OLIGOMERS AND POLYMERS OF THIENO[2,3-b]THIOPHENE
US8114316B2 (en) 2004-08-21 2012-02-14 Merck Patent Gmbh Monomers, oligomers and polymers of thieno[2,3-b]thiophene
TWI393728B (en) * 2004-08-21 2013-04-21 Merck Patent Gmbh Monomers, oligomers and polymers of thieno[2,3-b]thiophene
WO2007043835A1 (en) 2005-10-13 2007-04-19 Crystalgenomics, Inc. Fab i inhibitor and process for preparing same
WO2007067416A2 (en) * 2005-12-05 2007-06-14 Affinium Pharmaceuticals, Inc. Heterocyclylacrylamide compounds as fabi inhibitors and antibacterial agents
WO2007067416A3 (en) * 2005-12-05 2007-11-15 Affinium Pharm Inc Heterocyclylacrylamide compounds as fabi inhibitors and antibacterial agents
US8877975B2 (en) 2006-01-06 2014-11-04 Sunovion Pharmaceuticals Inc. Cycloalkylamines as monoamine reuptake inhibitors
US9868718B2 (en) 2006-01-06 2018-01-16 Sunovion Pharmaceuticals Inc. Cycloalkylamines as monoamine reuptake inhibitors
US8053603B2 (en) 2006-01-06 2011-11-08 Sunovion Pharmaceuticals Inc. Tetralone-based monoamine reuptake inhibitors
US10562878B2 (en) 2006-01-06 2020-02-18 Sunovion Pharamceuticals Inc. Cycloalkylamines as monoamine reuptake inhibitors
US8097760B2 (en) 2006-03-31 2012-01-17 Sunovion Pharmacuticals Inc. Preparation of chiral amides and amines
US7884124B2 (en) 2006-06-30 2011-02-08 Sepracor Inc. Fluoro-substituted inhibitors of D-amino acid oxidase
EP2054422A1 (en) * 2006-07-20 2009-05-06 Affinium Pharmaceuticals, Inc. Acrylamide derivatives as fab i inhibitors
EP2054422A4 (en) * 2006-07-20 2012-10-03 Affinium Pharm Inc Acrylamide derivatives as fab i inhibitors
US8895545B2 (en) 2006-07-20 2014-11-25 Debiopharm International Sa Acrylamide derivatives as Fab I inhibitors
WO2008009122A1 (en) 2006-07-20 2008-01-24 Affinium Pharmaceuticals, Inc. Acrylamide derivatives as fab i inhibitors
US7902252B2 (en) 2007-01-18 2011-03-08 Sepracor, Inc. Inhibitors of D-amino acid oxidase
US9862737B2 (en) 2007-02-16 2018-01-09 Debiopharm International Sa Salts, prodrugs and polymorphs of fab I inhibitors
US9586888B2 (en) 2007-05-31 2017-03-07 Sunovion Pharmaceuticals Inc. Phenyl substituted cycloalkylamines as monoamine reuptake inhibitors
US8669291B2 (en) 2007-05-31 2014-03-11 Sunovion Pharmaceuticals Inc. Phenyl substituted cycloalkylamines as monoamine reuptake inhibitors
EP2014287A1 (en) * 2007-06-13 2009-01-14 Bayer Schering Pharma Aktiengesellschaft Use of cinnamic acid derivatives as modulators of an EP2 receptor
EP2286808A1 (en) * 2009-08-18 2011-02-23 Rheinische Friedrich-Wilhelms Universität Cytohesin inhibitors
WO2011020849A1 (en) * 2009-08-18 2011-02-24 Rheinische Friedrich-Wilhelms Universität Cytohesin inhibitors
US9051321B2 (en) 2009-11-18 2015-06-09 Fab Pharma S.A.S. Heterocyclic acrylamides and their use as pharmaceuticals
US8846711B2 (en) 2009-11-18 2014-09-30 Fab Pharma S.A.S. Heterocyclic acrylamides and their use as pharmaceuticals
US9321769B2 (en) 2009-11-18 2016-04-26 Fab Pharma S.A.S. Heterocyclic acrylamides and their use as pharmaceuticals
WO2011061214A1 (en) 2009-11-18 2011-05-26 Fab Pharma Sas Novel heterocyclic acrylamides and their use as pharmaceuticals
US10016418B2 (en) 2010-11-05 2018-07-10 Seaomyx, Inc. Compounds useful as modulators of TRPM8
US10953007B2 (en) 2010-11-05 2021-03-23 Firmenich Incorporated Compounds useful as modulators of TRPM8
US9394287B2 (en) 2010-11-05 2016-07-19 Senomyx, Inc. Compounds useful as modulators of TRPM8
US9290493B2 (en) 2011-08-10 2016-03-22 Janssen Sciences Ireland Uc Antibacterial cyclopenta[c]pyrrole substituted 3,4-dihydro-1H-[1,8]naphthyridinones
US9884864B2 (en) 2011-08-10 2018-02-06 Janssen Sciences Ireland Uc Antibacterial cyclopenta[C]pyrrole substituted 3,4-Dihydro-1H-[1,8]naphthyridinones
US9012634B2 (en) 2011-08-10 2015-04-21 Janssen R&D Ireland Antibacterial piperidinyl substituted 3,4-dihydro-1H-[1,8]naphthyridinones
WO2013021051A1 (en) 2011-08-10 2013-02-14 Janssen R&D Ireland Antibacterial homopiperidinyl substituted 3,4 dihydro 1h [1,8]naphthyridinones
WO2013021052A1 (en) 2011-08-10 2013-02-14 Janssen R&D Ireland Antibacterial piperidinyl substituted 3,4-dihydro-1h-[1,8]naphthyridinones
US8906923B2 (en) 2011-08-10 2014-12-09 Janssen R&D Ireland Antibacterial cyclopenta[c]pyrrole substituted 3,4-dihydro-1H-[1,8]naphthyridinones
US9394295B2 (en) 2011-08-10 2016-07-19 Janssen Sciences Ireland Uc Antibacterial homopiperidinyl substituted 3,4-dihydro-1H-[1,8]naphthyridinones
US10526331B2 (en) 2011-08-10 2020-01-07 Janssen Sciences Ireland Unlimited Company Antibacterial cyclopenta[C]pyrrole substituted 3,4-dihydro-1H-[1,8]naphthyridinones
US10501463B2 (en) 2011-08-10 2019-12-10 Janssen Sciences Ireland Unlimited Company Antibacterial cyclopenta[C]pyrrole substituted 3,4-dihydro-1H-[1,8]naphthyridinones
US9598407B2 (en) 2011-08-10 2017-03-21 Janssen Sciences Ireland Uc Antibacterial piperidinyl substituted 3,4-dihydro-1H-[1,8]naphthyridinones
US9617262B2 (en) 2011-08-10 2017-04-11 Janssen Sciences Ireland Uc Antibacterial cyclopental[C]pyrrole substituted 3,4-dihydro-1H-[1,8]naphthyridinones
US10155759B2 (en) 2011-08-10 2018-12-18 Janssen Sciences Ireland Uc Antibacterial cyclopenta[c]pyrrole substituted 3,4-dihydro-1H-[1,8]naphthyridinones
WO2013021054A1 (en) 2011-08-10 2013-02-14 Janssen R&D Ireland Antibacterial cyclopenta[c]pyrrole substituted 3,4-dihydro-1h-[1,8]naphthyridinones
EP3330269A1 (en) * 2011-09-19 2018-06-06 Vitas Pharma Research Private Limited Heterocyclic compounds as inhibitors of fatty acid biosynthesis for bacterial infections
EP2758376A4 (en) * 2011-09-19 2015-04-29 Vitas Pharma Res Private Ltd Heterocyclic compounds as inhibitors of fatty acid biosynthesis for bacterial infections
US10035813B2 (en) 2012-06-19 2018-07-31 Debiopharm International Sa Prodrug derivatives of (E)-N-methyl-N-((3-methylbenzofuran-2-yl)methyl)-3-(7-oxo-5,6,7,8-tetrahydro-1,8-naphthyridin-3-yl)acrylamide
US8901105B2 (en) 2012-06-19 2014-12-02 Debiopharm International Sa Prodrug derivatives of (E)-N-methyl-N-((3-M ethylbenzofuran-2-yl)methyl)-3-(7-oxo-5,6,7,8-tetrahydro-1,8-naphthyridin-3-yl)acrylamide
US9296760B2 (en) 2012-08-10 2016-03-29 Janssen Sciences Ireland Uc Antibacterial compounds
US9315522B2 (en) 2012-08-10 2016-04-19 Janssen Sciences Ireland Uc Antibacterial compounds
WO2014023815A1 (en) 2012-08-10 2014-02-13 Janssen R&D Ireland New antibacterial compounds
WO2014072930A3 (en) * 2012-11-09 2014-07-31 Aurigene Discovery Technologies Limited Fused pyridine derivatives as antibacterial agents
WO2014072930A2 (en) * 2012-11-09 2014-05-15 Aurigene Discovery Technologies Limited Fused pyridine derivatives as antibacterial agents
US10167257B2 (en) 2014-04-04 2019-01-01 Iomet Pharma Ltd. Indole derivatives for use in medicine
US9732071B2 (en) 2015-10-01 2017-08-15 Senomyx, Inc. Compounds useful as modulators of TRPM8
US10751351B2 (en) 2016-02-26 2020-08-25 Debiopharm International S.A. Medicament for treatment of diabetic foot infections
US10858319B2 (en) 2016-10-03 2020-12-08 Iomet Pharma Ltd. Indole derivatives for use in medicine
US11667651B2 (en) 2017-12-22 2023-06-06 Hibercell, Inc. Aminopyridine derivatives as phosphatidylinositol phosphate kinase inhibitors
US11691967B2 (en) 2018-03-12 2023-07-04 The Board Of Trustees Of The University Of Illinois Antibiotics effective for gram-negative pathogens
WO2020099341A1 (en) 2018-11-12 2020-05-22 Debiopharm International S.A. Antibiotic compounds, methods of manufacturing the same, pharmaceutical compositions containing the same and uses thereof
US12006332B2 (en) 2019-06-17 2024-06-11 Hibercell, Inc. Aminopyrimidine derivatives as phosphatidylinositol phosphate kinase inhibitors

Also Published As

Publication number Publication date
US7524843B2 (en) 2009-04-28
US20110190283A1 (en) 2011-08-04
ECSP003699A (en) 2002-04-23
EP1226138A1 (en) 2002-07-31
JP4803935B2 (en) 2011-10-26
UY26380A1 (en) 2001-04-30
KR20020037770A (en) 2002-05-22
PL354892A1 (en) 2004-03-22
DE60017180D1 (en) 2005-02-03
BR0014470A (en) 2002-09-24
CA2387016A1 (en) 2001-04-19
US7790716B2 (en) 2010-09-07
TW534909B (en) 2003-06-01
AU773218B2 (en) 2004-05-20
US6846819B1 (en) 2005-01-25
JP2003511448A (en) 2003-03-25
KR100823382B1 (en) 2008-04-17
CZ302015B6 (en) 2010-09-08
US20050250810A1 (en) 2005-11-10
HUP0203122A2 (en) 2003-01-28
EP1226138B1 (en) 2004-12-29
CN1378542A (en) 2002-11-06
CZ20021167A3 (en) 2002-06-12
CA2387016C (en) 2010-09-28
IL148820A (en) 2007-08-19
DE60017180T2 (en) 2005-12-08
US20080125423A1 (en) 2008-05-29
AU7874700A (en) 2001-04-23
US8173646B2 (en) 2012-05-08
US7557125B2 (en) 2009-07-07
US20090275572A1 (en) 2009-11-05
ES2231275T3 (en) 2005-05-16
IL148820A0 (en) 2002-09-12
AR025976A1 (en) 2002-12-26
NO20021638L (en) 2002-06-05
PL201627B1 (en) 2009-04-30
EP1226138A4 (en) 2003-03-05
ATE285821T1 (en) 2005-01-15
BRPI0014470B1 (en) 2016-08-23
CN1197860C (en) 2005-04-20
NZ517706A (en) 2004-01-30
HU230030B1 (en) 2015-05-28
NO322708B1 (en) 2006-11-27
PE20010635A1 (en) 2001-08-15
NO20021638D0 (en) 2002-04-05
HK1049656A1 (en) 2003-05-23
HUP0203122A3 (en) 2004-01-28
ZA200202631B (en) 2003-05-28

Similar Documents

Publication Publication Date Title
CA2387016C (en) Acrylamide derivatives as fab i inhibitors
US7250424B2 (en) Fab I inhibitors
EP1180030B1 (en) Antibacterial compounds

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AL AU BA BB BG BR BZ CA CN CZ DZ EE GE GH GM HR HU ID IL IN IS JP KP KR LC LK LR LT LV MA MG MK MN MX MZ NO NZ PL RO SG SI SK SL TR TT TZ UA US UZ VN YU ZA

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 517706

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: IN/PCT/2002/00326/MU

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 78747/00

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 148820

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: PV2002-1167

Country of ref document: CZ

Ref document number: 10089755

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 2002/02631

Country of ref document: ZA

Ref document number: 200202631

Country of ref document: ZA

WWE Wipo information: entry into national phase

Ref document number: 1020027004456

Country of ref document: KR

ENP Entry into the national phase

Ref document number: 2001 530321

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: PA/a/2002/003561

Country of ref document: MX

Ref document number: 2387016

Country of ref document: CA

Ref document number: 00813989X

Country of ref document: CN

Ref document number: 2002/00950

Country of ref document: TR

WWE Wipo information: entry into national phase

Ref document number: 2000968895

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1020027004456

Country of ref document: KR

WWP Wipo information: published in national office

Ref document number: PV2002-1167

Country of ref document: CZ

WWP Wipo information: published in national office

Ref document number: 2000968895

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 517706

Country of ref document: NZ

WWG Wipo information: grant in national office

Ref document number: 517706

Country of ref document: NZ

WWG Wipo information: grant in national office

Ref document number: 78747/00

Country of ref document: AU

WWG Wipo information: grant in national office

Ref document number: 2000968895

Country of ref document: EP