WO2002079192A1 - Novel tyrosine kinase inhibitors - Google Patents

Novel tyrosine kinase inhibitors Download PDF

Info

Publication number
WO2002079192A1
WO2002079192A1 PCT/US2002/009402 US0209402W WO02079192A1 WO 2002079192 A1 WO2002079192 A1 WO 2002079192A1 US 0209402 W US0209402 W US 0209402W WO 02079192 A1 WO02079192 A1 WO 02079192A1
Authority
WO
WIPO (PCT)
Prior art keywords
methyl
hydroxy
ethylamino
pyridin
phenyl
Prior art date
Application number
PCT/US2002/009402
Other languages
French (fr)
Inventor
Mark D. Wittman
Neelakantan Balasubramanian
Upender Velaparthi
Kurt Zimmermann
Mark G. Saulnier
Peiying Liu
Xiaopeng Sang
David B. Frennesson
Karen M. Stoffan
James G. Tarrant
Original Assignee
Bristol-Myers Squibb Company
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to EEP200300475A priority Critical patent/EE200300475A/en
Priority to HU0400323A priority patent/HUP0400323A2/en
Priority to EP02723631A priority patent/EP1381598A4/en
Priority to IL15804102A priority patent/IL158041A0/en
Priority to CA002442428A priority patent/CA2442428A1/en
Priority to BR0208373-6A priority patent/BR0208373A/en
Priority to JP2002577817A priority patent/JP2004534010A/en
Priority to MXPA03008690A priority patent/MXPA03008690A/en
Application filed by Bristol-Myers Squibb Company filed Critical Bristol-Myers Squibb Company
Priority to KR10-2003-7012594A priority patent/KR20030083016A/en
Priority to SK12002003A priority patent/SK12002003A3/en
Publication of WO2002079192A1 publication Critical patent/WO2002079192A1/en
Priority to NO20034308A priority patent/NO20034308L/en
Priority to IS6968A priority patent/IS6968A/en
Priority to HR20030844A priority patent/HRP20030844A2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic 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
    • C07D401/04Heterocyclic 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 directly linked by a ring-member-to-ring-member bond
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41841,3-Diazoles condensed with carbocyclic rings, e.g. benzimidazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D231/00Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings
    • C07D231/02Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings
    • C07D231/10Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D231/12Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D233/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings
    • C07D233/54Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members
    • C07D233/56Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members with only hydrogen atoms or radicals containing only hydrogen and carbon atoms, attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D249/00Heterocyclic compounds containing five-membered rings having three nitrogen atoms as the only ring hetero atoms
    • C07D249/02Heterocyclic compounds containing five-membered rings having three nitrogen atoms as the only ring hetero atoms not condensed with other rings
    • C07D249/081,2,4-Triazoles; Hydrogenated 1,2,4-triazoles
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/14Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings

Definitions

  • the present invention relates generally to the field of tyrosine kinase enzyme inhibition using novel small molecules.
  • Tyrosine Kinases are a class of enzymes, which catalyze the transfer of the terminal phosphate of adenosine triphosphate to the phenolic hydroxyl group of a tyrosine residue present in the target protein.
  • Tyrosine kinases play a critical role in signal transduction for several cellular functions including cell proliferation, carcinogenesis, apoptosis, and cell differentiation (Plowman, G. D.; Ullrich, A.; Shawver, L. K.: Receptor Tyrosine Kinases As Targets For Drug Intervention. DN&P (1994) 7: 334-339). Therefore inhibitors of these enzymes would be useful for the treatment or prevention of proliferative diseases which are dependent on these enzymes.
  • Tyrosine kinases that have been implicated in these processes include Abl, CDK's, EGF, EMT, FGF, FAK, Flk-1/KDR, HER-2, IGF-1R, IR, LCK, MET, PDGF, Src, and NEGF (Traxler, P.M. Protein Tyrosine Kinase Inhibitors in Cancer Treatment. Exp. Opin. Ther. Patents (1997) 7: 571-588; incorporated herein by reference). Hence, there is an ongoing need to investigate novel compounds that can be used to regulate or inhibit tyrosine kinase enzymes.
  • the present invention relates to compounds which inhibit tyrosine kinase enzymes, compositions which contain tyrosine kinase inhibiting compounds and methods of using inhibitors of tyrosine kinase enzymes to treat diseases which are characterized by an overexpression or upregulation of tyrosine kinase activity such as cancer, diabetes, restenosis, arteriosclerosis, psoriasis, angiogenic diseases and immunologic disorders (Powis, G.; Workman, P. Signaling targets For The Development of Cancer Drugs. Anti-Cancer Drug Design (1994), 9: 263-277;
  • tyrosine kinase inhibitors can enhance the activity of cytotoxic or cytostatic treatments when used in combination with standard therapies known in the art.
  • the present invention is directed to compounds having formula I
  • X is selected from the group consisting of N, C, C 1 -C 3 alkyl, C 1 -C 3 alkyl substituted with one or more R , and a direct bond;
  • Y is selected from the group consisting of O and S ;
  • W is selected from the group consisting of N, C, O, and S, provided that when W is O or S, R 9 is absent;
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 are each independently selected from the group consisting of H, C ⁇ _ 6 alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, halo, amino, OR 60 , NO 2 , OH, SR 60 , NR 60 R 61 , CN, CO 2 R 60 , CONR 60 R 61 , CO 2 NR 60 R 61 , NR 62 CONR 60 R 61 , NR 60 SO 2 R 61 , SO 2 NR 60 R 61 , C(NR 62 )NR 60 R 61 , aryl, heteroaryl, (CH 2 ) n OR 60 , (CH 2 ) n NR 60 R 61 , (CH 2
  • Z is selected from the group consisting of Ci - C 4 alkyl, alkenyl, and alkynyl chain; Z having one or more hydroxy, thiol, alkoxy, thioalkoxy, amino, halo, NR 60 SO 2 R 61 groups; Z optionally incorporating one or more groups selected from the group consisting of CO, CNOH, CNOR 60 , CNNR 60 , CNNCOR 60 and CNNSO 2 R 60 ; and
  • R 25 is hydrogen, alkenyl, hydroxy, thiol, alkoxy, thioalkoxy, amino, alkylamino, dialkylamino, aryl, heteroaryl, cyano, halo, sulfoxy, sulfonyl, - NR 30 COOR 31 , -NR 30 C(O)R 31 , -C(O)NR 30 R 31 , heteroaryl or heterocycloalkyl; and
  • R 30 and R 31 are, independently, hydrogen, alkyl, cycloalkyl, or alkyl-R 25 .
  • R 1 , R 7 , R 8 and R 9 are H;
  • R 2 and R 4 are H or F
  • Y is O; X is selected from the group consisting of N and CH;
  • W is N
  • R 5 is selected from the group consisting of H, methyl, ethyl, isopropyl, secondary butyl, cyclopropyl, F, and CF 3 ;
  • R 6 is selected from the group consisting of H, 2-aminomethylpyridine, NHCH(CH 2 OH)CH 2 Ph, NHCH 2 CH(OH)aryl, and NHCH(CH 2 OH)CH 2 aryl; and R 3 is selected from the group consisting of OR 60 , C(NH)NHR 60 , C(O)NHR 60 imidazole, imidazoline, tetrahydropyrimidine, piperazine, morpholine, homomorpholine, piperidine, pyrrolidine, homopiperazine and amino; wherein
  • R 60 is selected from the group consisting of H, alkyl, cycloalkyl, heterocycloalkyl, and alkyl-R wherein R is hydrogen, alkenyl, hydroxy, thiol, thioalkoxy, alkoxy, thioalkoxy, halo, cyano, sulfoxy, sulfonyl, -NR COOR , - NR 30 C(O)R 31 , -NR 30 SO 2 R 31 , -C(O)NR 30 R 31 , or a heteroaryl or heterocycloalkyl; and
  • R 3 o and R 31 are, independently, hydrogen, alkyl, cycloalkyl, or alkyl-R .
  • the invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of formula I, as defined above, and a pharmaceutically acceptable carrier.
  • the invention further provides a pharmaceutical composition comprising a compound of formula I, as defined above, in combination with pharmaceutically acceptable carrier and at least one other anti-cancer agent optionally formulated as a fixed dose. Additionally provided is a method of treating a condition associated with at least one tyrosine kinase enzyme comprising administering to a mammalian species in need of such treatment an effective amount of a compound of formula I, as defined above. Furthermore, the invention provides a method of treating a condition associated with at least one tyrosine kinase enzyme comprising administering to a mammalian species at least one other anti-cancer agent in combination with a compound of formula I, as defined above.
  • the present invention provides for compounds of formula I, as defined above, pharmaceutical compositions employing such compounds and methods of using such compounds.
  • alkyl herein alone or as part of another group refers to a monovalent alkane (hydrocarbon) derived radical containing from 1 to 12 carbon atoms unless otherwise defined.
  • An alkyl group is an optionally substituted straight, branched or cyclic saturated hydrocarbon group. When substituted, alkyl groups may be substituted with up to four substituent groups, R as defined, at any available point of attachment. When the alkyl group is said to be substituted with an alkyl group, this is used interchangeably with "branched alkyl group”.
  • Exemplary unsubstituted such groups include methyl, ethyl, propyl, isopropyl, n-butyl, t-butyl, isobutyl, pentyl, hexyl, isohexyl, heptyl, 4,4-dimethylpentyl, octyl, 2,2,4-trimethylpentyl, nonyl, decyl, undecyl, dodecyl, and the like.
  • substituents may include but are not limited to one or more of the following groups: hydroxy, halo (such as F, CI, Br, I), haloalkyl (such as CCI3 or CF3), alkoxy, alkylthio, cyano, carboxy (-COOH), alkylcarbonyl (-C(O)R), alkoxycarbonyl (-OCOR), amino, carbamoyl (-NHCOOR or -OCONHR), urea (-NHCONHR), thiol, (-SH), sulfoxy, sulfonyl, aryl, heteroaryl, and heterocycloalkyl.
  • Alkyl groups as defined may also comprise one or more carbon to carbon double bonds or one or more carbon to carbon triple bonds.
  • Alkyl groups may also be represented by the formula alkyl-R 25 .
  • the alkyl group is a methyl, ethyl, propyl or butyl group and include substituted methyl, ethyl, propyl or butyl groups.
  • alkenyl herein alone or as part of another group refers to a hydrocarbon radical straight, branched or cyclic containing from 2 to 12 carbon atoms and at least one carbon to carbon double bond. An alkenyl group may be optionally substituted in the same manner as described for an alkyl group.
  • alkynyl herein alone or as part of another group refers to a hydrocarbon radical straight, branched or cyclic containing from 2 to 12 carbon atoms and at least one carbon to carbon triple bond.
  • An alkynyl group may be optionally substituted in the same manner as described for an alkyl group.
  • alkoxy refers to a straight or branched chain alkyl group covalently bonded to the parent molecule through an oxygen atom linkage containing from one to ten carbon atoms and the terms "C].
  • 6 alkoxy and “lower alkoxy” refer to such groups containing from one to six carbon atoms, examples include, but are not limited to, methoxy, ethoxy, propoxy, isopropoxy, butoxy, t-butoxy and the like.
  • alkoxy substituent when used in connection with an alkoxy substituent refers to the replacement of up to two hydrogens, preferably on different carbon atoms with a radical selected form the group of lower alkyl, phenyl, cyano, halo, trifluoromethyl, nitro, hydroxy, alkanoyl, amino, monoalkyl amino and dialkylamino.
  • Alkoxy groups may be substituted in the same manner that alkyl groups can be substituted as described above.
  • sulfoxy herein alone or as part of a group refers to -SO and may be substituted with, for example, alkyl or aryl groups.
  • sulfonyl herein alone or as part of a group refers to -SO 2 and may be substituted with alkyl or aryl groups.
  • amino herein alone or as part of another group refers to -NH .
  • An “amino” may optionally be substituted with one or two substituents, which may be the same or different, such as alkyl, aryl, arylalkyl, alkenyl, alkynyl, heteroaryl, heteroarylalkyl, cycloheteroalkyl, cycloheteroalkylalkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, hydroxyalkyl, alkoxyalkyl or thioalkyl.
  • substituents include alkylamino and dialkylamino, such as methylamino, ethylamino, dimethylamino, and diethylamino. These substituents may be further substituted with a carboxylic acid or any of the alkyl or aryl substituents set out herein.
  • amino substituents may be taken together with the nitrogen atom to which they are attached to form 1- pyrrolidinyl, 1-piperidinyl, 1-azepinyl, 4-morpholinyl, 4-thiamorpholinyl, 4- sulfoxymorpholine, 4-sulfonylmorpholine, 1-piperazinyl, 4-alkyl-l-piperazinyl, 4- arylalkyl-1-piperazinyl, 4-diarylalkyl- 1-piperazinyl , 1-homopiperazinyl, 4-alkyl-l- homopiperazinyl, 4-arylalkyl- 1-homopiperazinyl, 4-diarylalkyl- 1-homopiperazinyl; 1- pyrrolidinyl, 1-piperidinyl, or 1-azepinyl, optionally substituted with alkyl, alkoxy, alkylthio, halo, trifluoromethyl or hydroxy .
  • aryl herein alone or as part of another group refers to monocyclic or bicyclic aromatic rings, e.g. phenyl, substituted phenyl and the like, as well as groups which are fused, e.g., napthyl, phenanthrenyl and the like.
  • An aryl group thus contains at least one ring having at least 6 atoms, with up to five such rings being present, containing up to 22 atoms therein, with alternating (resonating) double bonds between adjacent carbon atoms or suitable heteroatoms.
  • Aryl groups may also be substituted with heterocycloalkyl and heterocycloaryl groups to form fused rings, such as dihydrobenzfuranyl, oxindolyl, indolyl, indolinyl, oxindolyl, benzoxazolidinonyl, benzoxazolinyl and benzoxazolidinone.
  • cycloalkyl herein alone or as part of another group refers to fully saturated and partially unsaturated hydrocarbon rings of 3 to 9, preferably 3 to 7 carbon atoms. Further, a cycloalkyl may be substituted.
  • R' and R" is independently selected from hydrogen, alkyl, substituted alkyl, and cycloalkyl, or R' and R" together form a heterocyclo or heteroaryl ring.
  • Cycloalkyl groups may also be substituted with hetero atoms such as O, N, and S to form heterocycloalkyl groups.
  • Preferred heterocycloalkyl groups include optionally substituted morpholine, homomorpholine (7 membered ring), hiomorpholine, piperazine, homopiperazine (7 membered ring), and piperidine.
  • heteroaryl herein alone or as part of another group refers to substituted and unsubstituted aromatic 5 or 6 membered monocyclic groups, 9 or 10 membered bicyclic groups, and 11 to 14 membered tricyclic groups which have at least one heteroatom (O, S or N) in at least one of the rings.
  • Each ring of the heteroaryl group containing a heteroatom can contain one or two oxygen or sulfur atoms and/or from one to four nitrogen atoms provided that the total number of heteroatoms in each ring is four or less and each ring has at least one carbon atom.
  • the fused rings completing the bicyclic and tricyclic groups may contain only carbon atoms and may be saturated, partially saturated, or unsaturated.
  • the nitrogen and sulfur atoms may optionally be oxidized and the nitrogen atoms may optionally be quaternized.
  • Heteroaryl groups which are bicyclic or tricyclic must include at least one fully aromatic ring but the other fused ring or rings may be aromatic or non- aromatic.
  • the heteroaryl group may be attached at any available nitrogen or carbon atom of any ring.
  • Exemplary monocyclic heteroaryl groups include pyrrolyl, pyrrolidinyl, imidazolinyl, pyrazolyl, pyrazolinyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl, thiadiazolyl, isothiazolyl, furanyl, thienyl, oxadiazolyl, pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, triazinyl, and the like.
  • Exemplary bicyclic heteroaryl groups include indolyl, indolinyl, oxindolyl, benzoxazolidinone, benzothiazolyl, benzodioxolyl, benzoxazolyl, benzothienyl, quinolinyl, tetrahydroisoquinolinyl, isoquinolinyl, benzimidazolyl, benzopyranyl, indolizinyl, benzofuranyl, chromonyl, coumarinyl, benzopyranyl, cinnolinyl, quinoxalinyl, indazolyl, pyrrolopyridyl, furopyridinyl, dihydroisoindolyl, tetrahydroquinolinyl and the like.
  • Exemplary tricyclic heteroaryl groups include carbazolyl, benzindolyl, phenanthrollinyl, acridinyl, phenanthridinyl, xanthenyl and the like.
  • halogen or halo
  • hydroxy herein alone or as part of another group refers to -OH.
  • thioalkoxy herein alone or as part of another group refers to an alkyl group as defined herein attached to the parent molecular group through a sulfur atom.
  • thioalkoxy include, but are not limited to, thiomethoxy, thioethoxy, and the like.
  • an "anti-cancer agent” as used herein includes known anti-cancer treatments such as radiation therapy or with cytostatic or cytotoxic agents, such as for example, but not limited to, DNA interactive agents, such as cisplatin or doxorubicin; topoisomerase II inhibitors, such as etoposide; topoisomerase I inhibitors such as CPT-11 or topotecan; tubulin interacting agents, such as paclitaxel, docetaxel or the epothilones; hormonal agents, such as tamoxifen; thymidilate synthase inhibitors, such as 5-fluorouracil; anti-metabolites, such as methotrexate; tyrosine kinase inhibitors such as Iressa and OSI-774; angiogenesis inhibitors; EGF inhibitors; VEGF inhibitors; CDK inhibitors; Her 1/2 inhibitors and monoclonal antibodies directed against growth factor receptors such as erbitux (EGF) and herceptin (Her2).
  • protecting groups for the compounds of the present invention will be recognized from the present application taking into account the level of skill in the art, and with reference to standard textbooks, such as Greene, T. W. et al., Protective Groups in Organic Synthesis, Wiley, N.Y. (1991).
  • alkenyl, alkynyl, cycloalkyl are substituted, they are preferably substituted with one or more hydroxy, cyano, carbamoyl, hydroxy, alkoxy, thiol, alkenyl, thioalkoxy, amino, alkylamino, amido, sulfonyl, sulfoxy, sulfonamido, halo, heterocycloalkyl, aryl or heteroaryl.
  • aryl or heteroaryl When aryl or heteroaryl are substituted, they are preferably substituted with one or more alkyl, alkenyl, alkynyl, cyano, carbamoyl, hydroxy, alkoxy, thioalkoxy, amino, amido, sulfonamido, halo or with R', R" wherein R', R" form a ring that is fused to the aryl group.
  • CH aryl or heteroaryl When CH aryl or heteroaryl are substituted, they are preferably substituted with one or more alkyl, alkyenyl, alkynyl, cyano, carbamoyl, hydroxy, alkoxy, thioalkoxy, amino, amido, sulfonamido, or halogen.
  • NH-Z-aryl or NH-Z-heteroaryl groups are substituted, they are preferably substituted with one or more alkyl, alkenyl, alkynyl, hydroxy, alkoxy, thioalkoxy, amino, halogen, nitro, nitrile, carboxylate, alkoxycarbonyl, carbamoyl, ester, amide, aryl, or heteroaryl
  • C The numbers in the subscript after the symbol "C” define the number of carbon atoms a particular group can contain.
  • “Ci .5 alkyl” means a straight or branched saturated carbon chain having from one to six carbon atoms; examples include methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl, t- butyl, n-pentyl, sec-pentyl, isopentyl, and n-hexyl.
  • Ci . ⁇ alkyl can also refer to Ci .5 alkylene which bridges two groups; examples include propane- 1, 3 -diyl, butane- 1,4-diyl, 2-methyl-butane-l,4-diyl, etc.
  • C2-6 alkenyl means a straight or branched carbon chain having at least one carbon-carbon double bond, and having from two to six carbon atoms; examples include ethenyl, propenyl, isopropenyl, butenyl, isobutenyl, pentenyl, and hexenyl.
  • C2-6 alkenyl can also refer to C2-6 alkenediyl which bridges two groups; examples include ethylene- 1,2-diyl (vinylene), 2-methyl-2-butene- 1,4-diyl, 2-hexene-l,6-diyl, etc.
  • C2-6 alkynyl means a straight or branched carbon chain having at least one carbon-carbon triple bond, and from two to six carbon atoms; examples include ethynyl, propynyl, butynyl, and hexynyl.
  • alkyl-R 25 includes optionally substituted alkyl groups such as methyl, ethyl, propyl, and butyl, attached to an R 25 group.
  • R 5 generally includes hydrogen, alkenyl, hydroxy, thiol, alkoxy, thioalkoxy, amino, alkylamino, dialkylamino, aryl, heteroaryl, cyano, halo, sulfoxy, sulfonyl, -NHCOOH, -NHC(O)-, -NHSO 2 -, -C(O)NH , heteroaryl or heterocycloalkyl groups such as morpholinyl or group having the formula: O O O
  • imidazole and “imidazoline” herein alone or as part of another group includes substituted imidazoles and substituted imidazolines.
  • tetrahydropyrimidine includes substituted tetrahydropyrimidines.
  • piperazine and “piperidine” “morpholines”, “homopiperazines”,
  • homomorpholines and “pyrrolidine” include substituted piperazines, substituted piperidines, substituted morpholines, substituted homomorpholines and substituted pyrrolidines, respectively.
  • X is selected from the group consisting of N, C, C1-C 3 alkyl, C 1 -C 3 alkyl substituted with one or more R , and a direct bond;
  • Y is selected from the group consisting of O and S ;
  • W is selected from the group consisting of N, C, O, and S, provided that when W is O or S, R 9 is absent;
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 are each independently selected from the group consisting of H, C 1 - 6 alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, halo, amino, OR 60 , NO 2 , OH, SR 60 , NR 60 R 61 , CN, CO 2 R 60 , CONR 60 R 61 , CO 2 NR o0 R 61 , NR 62 CONR 60 R 61 , NR 60 SO 2 R 61 , SO 2 NR o0 R 61 , C(NR 62 )NR 60 R 6 ', aryl, heteroaryl, (CH 2 ) favorOR 60 , (CH 2 ) n NR 60 R Dl , (CH 2 ) n SR 60 , (CH 2 ) contradict aryl, (CH 2 ) n heteroaryl,
  • Z is selected from the group consisting of - C 4 alkyl, alkenyl, and alkynyl chain; Z having one or more hydroxy, thiol, alkoxy, thioalkoxy, amino, halo, NR 60 SO R 61 groups; Z optionally inco ⁇ orating one or more groups selected from the group consisting of CO, CNOH, CNOR 60 , CNNR 60 , CNNCOR 60 and CNNSO 2 R 60 • and
  • R , and R ' are independently selected from the group consisting of H, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, hydroxy, alkoxy, aryl, heteroaryl, heteroarylalkyl, and alkyl-R wherein
  • R is hydrogen, alkenyl, hydroxy, thiol, alkoxy, thioalkoxy, amino, alkylamino, dialkylamino, aryl, heteroaryl, cyano, halo, sulfoxy, sulfonyl, - NR 30 COOR 31 , -NR 30 C(O)R 31 , -NR 30 SO 2 R 31 , -C(O)NR 30 R 31 , heteroaryl or heterocycloalkyl; and
  • R 30 and R 31 are, independently, hydrogen, alkyl, cycloalkyl, or alkyl-R 25 .
  • R 3 is -OR °.
  • R 60 is alkyl, or - alkyl-R 25 , wherein R 25 is hydrogen, alkenyl, hydroxy, thiol, alkoxy, thioalkoxy, amino, halo, cyano, alkylsulfoxy, alkylsulfonyl, -R 30 COOR 31 , -NR 30 C(O)R 31 , - NR 30 SO 2 R 31 , -C(O)NR 30 R 31 , heteroaryl or heterocycloalkyl; and R 30 and R 31 are, S independently, hydrogen, alkyl, cycloalkyl, or alkyl-R .
  • R 60 is methyl, -(CH 2 ) n CH 2 OH, or -(CH 2 ) n CH 2 N(CH 2 CH 2 ) 2 O, and n is 0, 1, or 2.
  • R is piperazine, homopiperazine, 3-methylpiperazine, or 3,5-dimethylpiperazine being optionally substituted at the 4-N position with a compound selected from the group consisting of alkyl, aryl, cycloalkyl, cycloalkylalkyl, heterocycloalkyl, alkyl-R 25 , -C(O)-R 15 , or -CO 2 R 15 wherein R 15 is
  • 9S S hydrogen, alkyl, aryl, alkyl-R , amino or aryl; and R is hydrogen, alkenyl, hydroxy, thiol, alkoxy, thioalkoxy, amino, alkylamino, dialkylamino, cyano, halo, sulfoxy, sulfonyl, arylsulfonyl, -NR 30 COOR 31 , -NR 30 SO 2 R 31 , -C(O)NR 30 R 31 , heteroaryl or heterocycloalkyl and R 30 and R 31 are, independently, hydrogen, alkyl, cycloalkyl, or alkyl-R 25 .
  • piperazine is substituted with Me, CH 2 cyclopropyl, CH 2 CH 2 NMe 2 , CH 2 CH 2 NEt 2 , CH 2 CH 2 NH 2 , CH 2 CH 2 NHMe, CH 2 CH 2 NHEt, N-CH 2 CH 2 N(CH 2 CH 2 ) 2 ⁇ , (CH 2 ) n CH 2 -R 25 wherein R 25 is OH, OMe, F, CN, CF 3 , SOCH 3 or SO 2 CH 3 , wherein n is 0, 1, or 2.
  • R " is an amino group.
  • Preferred amino groups include NHCH 2 CH 2 OH, NMeCH 2 CH 2 OH, NEtCH 2 CH 2 OH, NHCH 2 CH 2 NH 2 , NMeCH 2 CH 2 NH 2 , NEtCH 2 CH 2 NH 2 , NHCH 2 CH 2 NMe 2 , NMeCH 2 CH 2 NMe 2 , NEtCH 2 CH 2 NMe 2 , NHCH 2 CH 2 NEt 2 , NMeCH 2 CH 2 NEt 2 , NEtCH 2 CH 2 NEt 2 , NHCH 2 CH 2 N(CH 2 CH 2 ) 2 ⁇ , NMeCH 2 CH 2 N(CH 2 CH 2 ) 2 O, NEtCH 2 CH 2 N(CH 2 CH 2 ) 2 ⁇ .
  • R is an optionally substitued piperidine.
  • Preferred substituents are selected from the group consisting of hydroxy, thiol, amino, alkylamino, dialkylamino, alkoxy, thioalkoxy, 1,3 dioxolane (-OCHR 15 ) 2 , 1,3 dioxane (-OCHR 15 CHR 15 CHR 15 O-) -NHC(O)R 15 , -NHCO 2 R 15 , wherein R 15 is hydrogen, alkyl 9S or alkyl-R wherein R is hydrogen, alkenyl, hydroxy, thiol, alkoxy, thioalkoxy, amino, halo, cyano, alkylsulfoxy, alkylsulfonyl, -NR 30 COOR 31 , -NR 30 C(O)R 31 , - NR 30 SO 2 R 31 , -C(O)NR 30 R 31 , heteroaryl or heterocycloalkyl; and R 30
  • R 3 is an optionally substituted mo ⁇ holine, homomorpholine, thiomo ⁇ holine, sulfoxymo ⁇ holine, or sulfonylmo ⁇ holine.
  • Preferred substituents include hydroxy, thiol, amino, alkylamino, dialkylamino, alkoxy, thioalkoxy, alkyl-R 25 , -NHC(O)R 15 , -NHCO 2 R 15 , wherein R 15 is hydrogen,
  • R 3 is a pyrrolidine.
  • Preferred pyrrolidines include, 3- hydroxyl pyrrolidine, 3- alkoxy pyrrolidine, and 3-alkylamino pyrrolidine.
  • R 3 is an optionally substituted N-tetrahydropyrimidine or N-imidazoline wherein the substituents are, preferably, alkyl, hydroxyalkyl, alkoxyalkyl, haloalkyl, cyanoalkyl, carboxyl, or carboxamide.
  • R 6 is is selected from the group consisting of H, 2- aminomethylpyridine, NHCH 2 CH(OH)aryl, and NHCH(CH 2 OH)CH 2 aryl, wherein the aryl group is optionally substituted. In preferred embodiments, the aryl group is substituted with Br, CI, F, or methoxy. In some embodiments, R 6 has one of the following formulae:
  • R ,40 is hydrogen or alkyl, preferably methyl, and R 17 is hydrogen or halogen, such as Br, CI or F.
  • Preferred compounds of the present invention have one of the following formulae:
  • R 19 I ⁇ 9* ⁇ R and R are, independently, hydrogen, alkyl, or alkyl-R ;
  • R 15 is hydrogen, alkyl, or alkyl-R 25 ;
  • R 1 is independently, hydrogen or methyl;
  • R , R and R are, independently, hydrogen, halogen, or alkoxy,or R and R 19 together form a heterocycloalkyl or heteroaryl group;
  • R 25 is hydrogen, hydroxy, thiol, alkenyl, alkoxy, thioalkoxy, amino, halo, cyano, sulfoxy, sulfonyl, -NR 30 COOR 31 , -NR 30 C(O)R 31 , -NR 30 SO 2 R 31 , - C(O)NR 30 R 31 , heteroaryl or heterocycloalkyl; and
  • R and R are, independently, hydrogen, alkyl, cycloalkyl or alkyl-R .
  • R 12 is hydrogen, methyl, hydroxymethyl, methoxymethyl, CH 2 F, CH 2 CN, CO 2 H, or -CONR 30 R 31 wherein R 30 and R 31 are, independently, hydrogen, or alkyl-R 25 ;
  • R l is H; R 17 is Br, CI or F;
  • R 18 is halo or methoxy
  • R 19 is H.
  • Suitable examples of salts of the compounds according to the invention include inorganic or organic acids. These include, but are not limited to, hydrochloride, hydrobromide, sulfate, methanesulfonate, maleate, fumarate, phosphate and other pharmaceutically acceptable salts. Salts which are unsuitable for pharmaceutical uses but which can be employed, for example, for the isolation or purification of free compounds of formula I or their pharmaceutically acceptable salts, are also included. All stereoisomers of the compounds of the instant invention are contemplated, either in admixture or in pure or substantially pure form. The definition of the compounds according to the invention embraces all possible stereoisomers and their mixtures. It very particularly embraces the racemic forms and the isolated optical isomers having the specified activity.
  • racemic forms can be resolved by physical methods, such as, for example, fractional crystallization, separation or crystallization of diastereomeric derivatives or separation by chiral column chromatography.
  • the individual optical isomers can be obtained from the racemates by conventional methods, such as, for example, salt formation with an optically active acid followed by crystallization.
  • prodrug forms of the compounds of formula I Various forms of prodrugs are well known in the art. For examples of such prodrug derivatives, see:
  • the invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of formula I, as defined above, and a pharmaceutically acceptable carrier and at least one other anti-cancer agent formulated as a fixed dose.
  • Preferred anticancer agents are selected from the group consisting of: tamoxifen, toremifen, raloxifene, droloxifene, iodoxyfene, megestrol acetate, anastrozole, letrazole, borazole, exemestane, flutamide, nilutamide, bicalutamide, cyproterone acetate, goserelin acetate, luprolide, finasteride, herceptin, methotrexate, 5-fluorouracil, cytosine arabinoside, doxorubicin, daunomycin, epirubicin, idarubicin, mitomycin-C, dactinomycin, mithramycin, cisplatin, carboplatin, melphalan, chlorambucil, busulphan, cyclophosphamide, ifosfamide, nitrosoureas, thiotephan
  • the invention provides a method of treating a condition via modulation of at least one tyrosine kinase enzyme comprising administering to a mammalian species in need of such treatment an effective amount of a compound of formula I, as defined above, in combination (simultaneously or sequentially) with at least one other anti-cancer agent.
  • a preferred condition, treated by said methods of the instant invention, is cancer.
  • the tyrosine kinase enzyme may include (but is not limited to): Abl, CDK's, EGF, EMT, FGF, FAK, Flk-1/KDR, HER-2, IGF-1R, IR, LCK, MET, PDGF, Src, and VEGF.
  • the invention also provides a method for treating cancer, comprising administering to a mammalian species in need of such treatment, a therapeutically effective amount of at least one of the pharmaceutical compositions defined above.
  • the invention further provides a method for treating proliferative diseases, comprising administering to a mammalian species in need of such treatment a therapeutically effective amount of at least one of the pharmaceutical compositions defined above.
  • Certain compounds of formula I may generally be prepared according to the following schemes and the knowledge of one skilled in the art. Solvates (e.g., hydrates) of the compounds of formula I are also within the scope of the present invention. Methods of solvation are generally known in the art. Accordingly, the compounds of the instant invention may be in the free or hydrate form, and may be obtained by methods exemplified by the following schemes below. More specifically, Schemes I-V ⁇ illustrate the preparation of compounds claimed in this invention. The examples, which follow, illustrate the compounds that can be synthesized by these schemes. The schemes are not limited by the examples listed or by any substituents employed for illustrative pu ⁇ oses.
  • Scheme I describes the preparation of the benzimidazoles.
  • the starting diamines 1 are readily available using literature methods or are obtained commercially.
  • the diamine is then condensed with an aldehyde 2 to provide the benzimidazole 3. Further modification of the functional groups on the aryl group of the benzimidazole or heterocycle of 3 are then envisioned.
  • the benzimidazole could be formed in a step-wise manner (see Scheme II) by amide formation using the acid chloride of 5 or any of the commonly used peptide coupling reagents such as DCC (dicyclohexylcarbodiimide), EDCI (l-(3- dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride), etc.
  • DCC dicyclohexylcarbodiimide
  • EDCI l-(3- dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride
  • the amide 6 was formed the nitro group could be reduced using catalytic hydrogenation, transfer hydrogenation or chemical reduction such as SnCl 2 or iron powder or other methods known in the art for reduction of aryl nitro groups. Treatment of the aniline with acid would then form the benzimidazole.
  • Scheme III illustrates the use of 4-iodo-2-methoxy-pyridine-3- carbaldehyde 7 to provide the functionalized benzimidazole 8.
  • Hydrolysis of the methoxy group using protic acid conditions, TMSI (trimethylsilyl iodide), BBr 3 , or other conditions known in the art for cleaving a methyl ether would provide the halopyridone 9.
  • Addition of heteroatom nucleophiles using amines, alcohols or thiols would then provide the substituted pyridones 10.
  • Other functionality could be inco ⁇ orated into the aldehyde and the above example is included for illustrative pu ⁇ oses only.
  • aryl ring of the benzimidazole prepared using Schemes I or II can be modified.
  • introduction of a cyano group for R 3 on the benzimidazole allows for the formation of heterocycles such as imidazole, imidazolines, oxazolines, thiazolines, amides, or amidines.
  • Scheme IV illustrates such transformations. Starting from the cyano-substituted benzimidazole 11 the heterocycle can be modified as illustrated in Scheme IV to provide 12. Imidate formation preferably using ethanol and acid provides intermediate 13.
  • Imidate 13 can be transformed using diamines to form imidazolines, amino alcohols to form oxazolines, amino acetals to form imidazoles, and amino thiols to form thiazolines 14.
  • the imidate can be hydrolyzed to the acid and coupled with amines using any of the standard amide formation reagents (DCC, EDCI, etc.) to form amides 15.
  • Imidate 13 is also a useful intermediate for the preparation of amidines 16 by reacting with amines.
  • Scheme V illustrates further transformation of benzimidazoles that bear a halogen atom using palladium catalysis using conditions developed by Suzuki [Yang et al. Acta Chem. Scand. (1993) 221; Suzuki et al. Synth. Commun. (1981) 11: 513] or Buchwald/Hartwig [Buchwald et al. J. Am. Chem. Soc. (1994) 116: 7901; Hartwig et al. J. Am. Chem. Soc. (1994) 116: 5969; Hartwig. Angew. Chem., Int. Ed. Engl (1998) 37: 2046] and variations of these methods.
  • bromide substituted benzimidazole 17 could be envisioned to provide a substrate for Suzuki coupling with aryl, vinyl, and heterocyclic boronic acids to provide benzimidazoles 18.
  • amines and heterocycles such as piperazine or mo ⁇ holine derivatives 19 can be prepared from the same bromide using amines under conditions described by Buchwald and Hartwig or variations thereof.
  • amine and heterocyclic derivatives such as 19 can be prepared using intermediate 6 described in Scheme II.
  • the halogen can be displaced with amines, alcohols, heterocyclic amines and other nitrogen containing heterocycles such as piperazine, piperidine, 4-amino piperidine, mo ⁇ holine, imidazole, etc (Scheme VI).
  • the terminal nitrogen of piperazine or 4-amino piperidine can then be alkylated using standard alkylation conditions or reacted with aldehydes in a reductive amination reaction to provide alkylated derivatives.
  • terminal nitrogen atom of piperazine or 4- amino piperidine can be acylated or carbamoylated using any number of conditions that are routine for someone skilled in the art of organic synthesis. Following the example illustrated in Scheme II compounds such as 19 could be prepared.
  • amines, heterocycles, and alcohols can be introduced at R 3 using a nucleophilic aromatic substitution reaction started from an intermediate 20 were R 3 is halogen, preferably F, the halogen can be displaced with amines, alcohols, heterocyclic amines and other nitrogen containing heterocycles such as piperazine,
  • the compounds according to the invention have pharmacological properties; in particular, the compounds of formula I are tyrosine kinase enzyme inhibitors.
  • the novel compounds of formula I are thus useful in the therapy of a variety of proliferative diseases (including but not limited to diseases associated with tyrosine kinase enzymes) such as cancer, autoimmune diseases, viral diseases, fungal diseases, neurodegenerative disorders and cardiovascular disease.
  • the compounds of formula I are useful in the treatment of a variety of cancers, including, but not limited to, the following: a) carcinoma, including that of the bladder, breast, colon, kidney, liver, lung, including small cell lung cancer, esophagus, gall bladder, ovary, pancreas, stomach, cervix, thyroid, prostate, and skin, including squamous cell carcinoma; b) hematopoietic tumors of lymphoid lineage, including leukemia, acute lymphocytic leukemia, acute lymphoblastic leukemia, B-cell lymphoma, T-cell lymphoma, Hodgkins lymphoma, non-Hodgkins lymphoma, hairy cell lymphoma and Burkett's lymphoma; c) hematopoietic tumors of myeloid lineage, including acute and chronic myelogenous leukemias, myelodysplastic syndrome and promyelocytic leukemia; d) tumors of me
  • inhibitors could act as reversible cytostatic agents which may be useful in the treatment of any disease process which features abnormal cellular proliferation, e.g., benign prostatic hype ⁇ lasia, familial adenomatosis polyposis, neuro-fibromatosis, atherosclerosis, pulmonary fibrosis, arthritis, psoriasis, glomerulonephritis, restenosis following angioplasty or vascular surgery, hypertrophic scar formation, inflammatory bowel disease, transplantation rejection, endotoxic shock, and fungal infections.
  • benign prostatic hype ⁇ lasia familial adenomatosis polyposis
  • neuro-fibromatosis e.g., atherosclerosis, pulmonary fibrosis, arthritis, psoriasis, glomerulonephritis, restenosis following angioplasty or vascular surgery
  • hypertrophic scar formation e.g., benign prostatic hype ⁇ lasia, familial adenomatosis polyposis, neuro-fibromatosis,
  • Compounds of formula I may induce apoptosis.
  • the apoptotic response is aberrant in a variety of human diseases.
  • Compounds of formula I, as modulators of apoptosis, will be useful in the treatment of cancer (including but not limited to those types mentioned herein above), viral infections (including but not limited to he ⁇ esvirus, poxvirus, Epstein-Barr virus, Sindbis virus and adenovirus), prevention of AIDS development in HIV-infected individuals, autoimmune diseases (including but not limited to systemic lupus, erythematosus, autoimmune mediated glomerulonephritis, rheumatoid arthritis, psoriasis, inflammatory bowel disease, and autoimmune diabetes mellitus), neurodegenerative disorders (including but not limited to Alzheimer's disease, AIDS-related dementia, Parkinson's disease, amyotrophic lateral sclerosis, retinitis pigmentosa, spinal muscular atrophy and cerebellar degeneration), my
  • Compounds of formula I may modulate the level of cellular RNA and DNA synthesis. These agents would therefore be useful in the treatment of viral infections (including but not limited to HIV, human papilloma virus, he ⁇ esvirus, poxvirus, Epstein-Barr virus, Sindbis virus and adenovirus).
  • viral infections including but not limited to HIV, human papilloma virus, he ⁇ esvirus, poxvirus, Epstein-Barr virus, Sindbis virus and adenovirus.
  • Compounds of formula I may also be useful in the chemoprevention of cancer. Chemoprevention is defined as inhibiting the development of invasive cancer by either blocking the initiating mutagenic event or by blocking the progression of pre- malignant cells that have already suffered an insult or inhibiting tumor relapse. Compounds of formula I may also be useful in inhibiting tumor angiogenesis and metastasis.
  • the compounds of this invention may also be useful in combination (administered together or sequentially) with known anti-cancer treatments such as radiation therapy or with cytostatic or cytotoxic agents, such as for example, but not limited to, DNA interactive agents, such as cisplatin or doxorubicin; topoisomerase II inhibitors, such as etoposide; topoisomerase I inhibitors such as CPT-11 or topotecan; tubulin interacting agents, such as paclitaxel, docetaxel or the epothilones; hormonal agents, such as tamoxifen; thymidilate synthase inhibitors, such as 5-fluorouracil; and anti-metabolites, such as methotrexate; tyrosine kinase inhibitors such as Iressa and OSI-774; angiogenesis inhibitors; EGF inhibitors; VEGF inhibitors; CDK inhibitors; Herl/2 inhibitors and monoclonal antibodies directed against growth factor receptors such as erbitux (
  • Such combination products employ the compounds of this invention within the dosage range described below and the other pharmaceutically active agent or treatment within its approved dosage range.
  • Compounds of formula I may also be administered sequentially with known anticancer or cytotoxic agents when a combination formulation is inappropriate. The invention is not limited in the sequence of administration; compounds of formula I may be administered either prior to or after administration of the known anticancer or cytotoxic agent(s).
  • compositions for use as described above, including controlling cancer, inflammation and arthritis, which contain at least one compound of the formula I as defined above or at least one of its pharmacologically acceptable acid addition salts, and the use of a compound of the formula I as defined above for the preparation of a pharmaceutical having activity against proliferative diseases as described previously including against cancer, inflammation and/or arthritis.
  • the pharmacological properties of the compounds of this invention may be confirmed by a number of pharmacological assays.
  • the exemplified pharmacological assays which follow have been carried out with the compounds according to the invention and their salts.
  • CDK2/cyclin E complex 0.5 ⁇ g GST-RB fusion protein (amino acids 776-928 of retinoblastoma protein), 0.2 ⁇ Ci 33 P ⁇ -ATP, 25 ⁇ M ATP in 50 ⁇ l kinase buffer (50 mM Hepes, pH 8.0, 10 mM MgCl 2 , 1 mM EGTA, 2 mM DTT). Reactions were incubated for 45 minutes at 30° C and stopped by the addition of cold trichloroacetic acid (TCA) to a final concentration of 15% .
  • TCA cold trichloroacetic acid
  • TCA precipitates were collected onto GF/C unifilter plates (Packard Instrument Co., Meriden, CT) using a Filtermate universal harvester (Packard Instrument Co., Meriden, CT) and the filters were quantitated using a TopCount 96-well liquid scintillation counter (Packard Instrument Co., Meriden, CT).
  • Dose response curves were generated to determine the concentration required inhibiting 50% of kinase activity (IC 50 ).
  • Compounds were dissolved at 10 mM in dimethylsulfoxide (DMSO) and evaluated at six concentrations, each in triplicate. The final concentration of DMSO in the assay equaled 2%.
  • a filter-based kinase assay measuring the phosphorylation of Gst-SLP76 by Gst-Emtk, was employed to determine the compound inhibitory activity against Emt.
  • the kinase reaction was performed in a 96-well plate at room temperature for 15 min before being terminated by adding 100 ⁇ l of 20% trichloroacetic acid (TCA) containing 0.1 M sodium pyrophosphate.
  • TCA trichloroacetic acid
  • the kinase reaction mixture (60 ⁇ l ) contained 25 mM HEPES, pH 7.0, 0.1 mg/ml BSA, 5 mM MgCl 2 , 5 mM MnCl 2 , 8 ng of enzyme (Gst-Emtk), 5 ⁇ g of the substrate protein (Gst-SLP76), 1 ⁇ M ATP, 0.4 ⁇ Ci of [ ⁇ -P ] ATP and the tested compound (at various concentrations). After termination, the proteins were allowed to precipitate in the presence of TCA for 1 fir at 4 °C.
  • the precipitated proteins were then harvested on a filter plate (UniFilter-96, GF/C, Packard Instrument) and washed to remove excess [ ⁇ -P ] ATP.
  • the radioactivity was determined using a TopCount NXT (Packard Instrument) after adding 35 ⁇ l of Microscint 20 (Packard Instrument).
  • the Focal Adhesion kinase was assayed using the synthetic polymer poly(Glu/Tyr) (Sigma Chemicals) as a phosphoacceptor substrate.
  • Each reaction mixture consisted of a total volume of 50 ul and contained 100 ng of baculovirus- expressed enzyme, 2 ⁇ g of poly(Glu/Tyr), l ⁇ M of ATP, and 0.2 ⁇ Ci of [ ⁇ - 33 P]ATP.
  • the mixtures also contained 40 mM Tris.HCl, pH 7.4, 1 mM MnCl 2 , 0.5 mM DTT, and 0.1 mg/ml bovine serum albumin.
  • kinase activity was determined by quantitation of the amount of radioactive phosphate transferred to the poly(Glu/Tyr) substrate.
  • Inco ⁇ oration was measured by the addition of cold trichloroacetic acid (TCA) precipitation of the proteins which were collected onto GF/C unifilter plates (Packard Instrument Co., Meriden, CT) using a Filtermate universal harvester and the filters were quantitated using a TopCount 96-well liquid scintillation counter (Packard Instrument Co., Meriden, CT).
  • TCA cold trichloroacetic acid
  • kinase reactions consisted of 10 ng of baculovirus expressed GST- HER1, 100 ng of HER2, 100 ng/ml poly(Glu/Tyr) (Sigma), 0.2 ⁇ Ci 33P ⁇ -ATP, 1 ⁇ M ATP in 50 ⁇ l kinase buffer (50 mM Tris, pH 7.5, 10 mM MnC12, 0.5 mM DTT). Reactions were incubated for lh at 27 C and stopped by the addition of cold trichloroacetic acid (TCA) to a final concentration 15%.
  • TCA cold trichloroacetic acid
  • TCA precipitates were collected onto GF/C unifilter plates (Packard Instrument Co., Meriden, CT) using a Filtermate universal harvester (Packard Instrument Co., Meriden, CT) and the filters were quantitated using a TopCount 96-well liquid scintillation counter (Packard Instrument Co., Meriden, CT).
  • Dose response curves were generated to determine the concentration required to inhibit 50% of kinase activity (IC 50 ).
  • Compounds were dissolved at 10 mM in dimethylsulfoxide (DMSO) and evaluated at six concentrations, each in triplicate. The final concentration of DMSO in the assay equaled 1%.
  • the IGF-1 receptor tyrosine kinase was assayed using the synthetic polymer poly(Glu/Tyr) (Sigma Chemicals) as a phosphoacceptor substrate.
  • Each reaction mixture consisted of a total volume of 50 ul and contained 125 ng of baculovirus expressed enzyme, 2.5 ⁇ g of poly(Glu/Tyr), 25 ⁇ M of ATP, and 0.1 ⁇ Ci of [ ⁇ - 33 P]ATP.
  • the mixtures also contained 20 mM MOPS, pH 7.0, 5 mM MnCl 2 , 0.5 mM DDT, and 0.1 mg/ml bovine serum albumin.
  • reaction mixtures were incubated at 30 ° C for 45 minutes and kinase activity was determined by quantitation of the amount of radioactive phosphate transferred to the poly(Glu/Tyr) substrate.
  • Inco ⁇ oration was measured by the addition of cold trichloroacetic acid (TCA) precipitation of the proteins which were collected onto GF/C unifilter plates (Packard Instrument Co., Meriden, CT) using a Filtermate universal harvester and the filters were quantitated using a TopCount 96-well liquid scintillation counter (Packard Instrument Co.,
  • the Insulin Receptor Tryrosine kinase was assayed using the synthetic polymer poly(GlutTyr) (Sigma Chemicals) as a phosphoacceptor substrate.
  • Each reaction mixture consisted of a total volume of 50 ul and contained 90 ng of baculovirus-expressed enzyme, 2.5 ⁇ g of poly(Glu/Tyr), 25 ⁇ M of ATP, and 0.1 ⁇ Ci of [ ⁇ - 33 P]ATP.
  • the mixtures contained also 20 mM Tris.HCl, pH 7.4, 5 mM MnCl 2 , 0.5 mM DTT, and 0.1 mg/ml bovine serum.
  • kinase activity was determined by quantitation of the amount of radioactive phosphate transferred to the poly(Glu/Tyr) substrate.
  • Inco ⁇ oration was measured by the addition of cold trichloroacetic acid (TCA) precipitation of the proteins which were collected onto GF/C unifilter plates (Packard Instrument Co., Meriden, CT) using a Filtermate universal harvester and the filters were quantitated using a TopCount 96-well liquid scintillation counter (Packard Instrument Co.,
  • kinase reactions consisted of 10 ng of baculovirus expressed lOng GST-Lck, 100 ng/ml poly(Glu/Tyr) (Sigma), 0.2 ⁇ Ci 33P ⁇ -ATP, 1 ⁇ M ATP in 50 ⁇ l kinase buffer (50 mM Tris, pH 7.5, 10 mM MnC12, 0.5 mM DTT). Reactions were incubated for lh at 27 C and stopped by the addition of cold trichloroacetic acid (TCA) to a final concentration 15%.
  • TCA cold trichloroacetic acid
  • TCA precipitates were collected onto GF/C unifilter plates (Packard Instrument Co., Meriden, CT) using a Filtermate universal harvester (Packard Instrument Co., Meriden, CT) and the filters were quantitated using a TopCount 96-well liquid scintillation counter (Packard Instrument Co.,
  • IC 50 concentration required to inhibit 50% of kinase activity
  • Compounds were dissolved at 10 mM in dimethylsulfoxide (DMSO) and evaluated at six concentrations, each in triplicate. The final concentration of DMSO in the assay equaled 1%.
  • MET Kinase Assay Kinase reactions consisted of lOng of baculovirus expressed GST-Met, 2.5ug poly(Glu/Tyr) (Sigma), 0.2 ⁇ Ci 33P ⁇ -ATP, 10 ⁇ M ATP in 50 ⁇ l kinase buffer (40mM Tris, pH 7.5, ImM MnC12, 0.50 mM DTT). Reactions were incubated for lh at 27 C and stopped by the addition of cold trichloroacetic acid (TCA) to a final concentration 3.5%.
  • TCA cold trichloroacetic acid
  • TCA precipitates were collected onto GF/C unifilter plates (Packard Instrument Co., Meriden, CT) using a Filtermate universal harvester (Packard Instrument Co., Meriden, CT) and the filters were quantitated using a TopCount 96-well liquid scintillation counter (Packard Instrument Co., Meriden, CT).
  • Dose response curves were generated to determine the concentration required to inhibit 50% of kinase activity (IC 50 ).
  • Compounds were dissolved at 10 mM in dimethylsulfoxide (DMSO) and evaluated at seven concentrations, each in triplicate. The final concentration of DMSO in the assay equaled 1%.
  • Kinase reactions consisted of 70ng of baculovirus expressed GST-PDGFbR, 0.3ug biotinylated poly(Glu/Tyr) (Sigma), in 50 ⁇ l kinase buffer (20 mM Hepes, pH 7.5, 0.7uM ATP, lOmM MnC12, 0.5mM DTT, 0.15mM NaCl, O.lmg/ml BSA). Reactions were incubated for 30 minutes at room temperature with shaking and stopped by the addition of lOul of 0.2M EDTA, pH8.0. 150ul of HTRF detection buffer was added and incubated for 1 hour and 30 minutes at room temperature. Counts were quantitated on Discovery HTRF Packard Instrument.
  • kinase reactions consisted of 7.5ng of baculovirus expressed GST-KDR, 1.5ug ⁇ oly(Glu/Tyr) (Sigma), 0.04 ⁇ Ci 33P ⁇ -ATP, 2.5 ⁇ M ATP in 50 ⁇ l kinase buffer (25 mM Tris, pH 7.5, 1.8 mM MnC12, 0.0.625 mM DTT). Reactions were incubated for lh at 27 C and stopped by the addition of cold trichloroacetic acid (TCA) to a final concentration 15%.
  • TCA cold trichloroacetic acid
  • TCA precipitates were collected onto GF/C unifilter plates (Packard Instrument Co., Meriden, CT) using a Filtermate universal harvester (Packard Instrument Co., Meriden, CT) and the filters were quantitated using a TopCount 96-well liquid scintillation counter (Packard Instrument Co.,
  • IC 50 concentration required to inhibit 50% of kinase activity
  • Compounds were dissolved at 10 mM in dimethylsulfoxide (DMSO) and evaluated at six concentrations, each in triplicate. The final concentration of DMSO in the assay equaled 1%.
  • Cytotoxicity assay (HT-29-colon; Colo205, MCF-7-breast) Tumor cell lines are maintained in McCoy's 5A medium (GIBCO) and 10% heat inactivated fetal bovine serum (GIBCO). The in vitro cytotoxicity is assessed in tumor cells by a tetrazolium-based colorimetric assay which takes advantage of the metabolic conversion of MTS (3-(4,5-dimefhylthiazol-2-yl)-5-(3- carboxymethoxyphenyl)-2-(4-sulphenyl)-2H-tetrazolium, inner salt) (Promega) to a reduced form that absorbs light at 492 nm (1).
  • MTS 3-(4,5-dimefhylthiazol-2-yl)-5-(3- carboxymethoxyphenyl)-2-(4-sulphenyl)-2H-tetrazolium, inner salt
  • HT-29 and Colo205 are human colon tumor cell lines, and MCF-7 is a human breast tumor cell line.
  • 5-Chloroacetyl-7-chlorooxindole To a suspension of AICI 3 (13.4g, 0.10 mol) in methylene chloride (40mL) is added a solution of 7-Chlorooxindole (0.10 mol) and chloroacetyl chloride (8 mL, 0.10 mol) at 0°C. The solution is warmed to ambient temperature for two hours and poured onto ice and extracted with methylene chloride, washing with saturated bicarbonate solution, brine, and drying over MgSO 4 would provide the desired chloroketone.
  • Methyl-CBS-oxazaborolidine (IM in toluene, 0.745 mL, 0.745 mmol) and BH 3 -THF (8mL, 8 mmol) is added at the same time a solution of BH 3 -THF (19mL, 19 mmol) and a solution of the 5-Chloroacetyl-7-chlorooxindole (37.98 mmol) in 19 mL of THF. Both solutions are added dropwise over 30 minutes. The solution is stirred for 1 hour and quenched with the slow addition of methanol (50mL). The solution is concentrated and the residue chromatographed over a short silica gel column (1:1 hexane/ethyl acetate).
  • 6-ChloroacetyI-4-chloro-2-benzooxazolinone To a suspension of AICI 3 (13.4g, 0.10 mol) in methylene chloride (40mL) is added a solution of 4-chloro-2- benzooxazolinone (0.10 mol) and chloroacetyl chloride (8 mL, 0.10 mol) at 0°C. The solution is warmed to ambient temperature for two hours and poured onto ice and extracted with methylene chloride, washing with saturated bicarbonate solution, brine, and drying over MgSO 4 would provide the desired chloroketone.
  • N-MethyI-7-chloroindoline To a solution of the 7-Chloroindoline (0.10 mol) in 500 mL of acetone is added K 2 CO 3 (0.15mol) and Mel (0.15mol) and refluxed until the starting material is consumed. The reaction is filtered and washed with water and saturated bicarbonate solution, drying over MgSO 4 would provide the N-Me -7- chloroindoline
  • N-Methyl-5-chloroacetyl-7-chloro-indoline To a suspension of AICI 3 (13.4g, 0.10 mol) in methylene chloride (40mL) is added a solution of N-Me-7-Chloroindoline (0.10 mol) and chloroacetyl chloride (8 mL, 0.10 mol) at 0°C. The solution is warmed to ambient temperature for two hours and poured onto ice and extracted with methylene chloride, washing with saturated bicarbonate solution, brine, and drying over MgSO 4 provides the desired chloroketone.
  • 6-Chloroacetyl-4-chloro-2-methyl-benzooxazole To a suspension of AICI 3 (13.4g, 0.10 mol) in methylene chloride (40mL) is added a solution of 2-Methyl-4-chIoro- benzooxazole (0.10 mol) and chloroacetyl chloride (8 mL, 0.10 mol) at 0°C. The solution is warmed to ambient temperature for two hours and poured onto ice and extracted with methylene chloride, washing with saturated bicarbonate solution, brine, and drying over MgSO would provide the desired chloroketone.
  • N-(4-Imidazol-l-yl-2-methyl-6-nitro-phenyI)-acetamide To a solution of 4- imidazol-l-yl-2-methyl-phenylamine (1 g, 5.78 mmol) in CH2CI 2 (20 mL) was added AC 2 O (0.7 mL, 7.28 mmol) at 0°C. The reaction mixture was stir at room temperature for 14 h and diluted with water. The aqueous layer was extracted with CH2CI2 and the combined organic layers were washed with saturated ⁇ aHC ⁇ 3 and brine, dried over Na 2 SO 4 , and concentrated in vacuo to give a white solid. The white solid was suspended in H 2 SO 4 (cone.) (15 mL).
  • HNO 3 (cone.) (0.312 mL) was added to the suspension at 0°C.
  • the reaction mixture was slowly warmed to room temperature and stirred at room temperature for 4 h. After cooling to -10°C, the reaction mixture was neutralized with ammonium hydroxide and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over Na 2 SO 4 , and concentrated. The residue was purified by flash chromatography (1:9:5 MeOH / THF / hexane) to yield the title compound (0.61 g, 41%).
  • N-formyl-N, N', N'- trimethylethylenediamine (176 mL, 1.37 mol) dropwise at -78°C.
  • the reaction mixture was stirred for ca. 30 min at -78°C before warming to -23°C over ca. 30 min.
  • ethylene glycol dimethyl ether (1 L) followed by rt-butyllithium (2.5 M solution in hexane) (800 mL, 2.0 mol). The resulting mixture was stirred for ca. 2 h during which time the reaction mixture turned deep green.
  • a 12-L 4-necked round flask was charged with iodine (571 g, 2.25 mol) and ethylene glycol dimethyl ether (2 L) and the resultant solution was cooled to -78°C.
  • the contents of the 5-L flask were transferred via canula to the mixture of iodine and ethylene glycol dimethyl ether in the 12-L flask at -78°C.
  • the reaction mixture was stirred for an additional 1 h at -78°C.
  • the cooling bath was removed and the mixture was allowed to warm to about 0°C and treated with 2 L of water and 2 L of 1 ⁇ hydrochloric acid. Methyl t-butyl ether (2L) was added and the layers were separated.
  • N-(4-Cyano-2-methyl-6-nitro-phenyl)-2,2,2-trifluoro-acetamide To the ice-cold trifluoroacetic anhydride (60 mL) was added 4-amino-3-methyl-benzonitrile (14.33 g, 0.108 mol) in portion. The resulting white slurry was stirred at 0°C for 30 min. Then ammonium nitrate (17.28 g, 0.216 mol) was added. The reaction mixture was allowed to stir at 0°C for 1 h and at room temperature for 14 h. After removal of most solvent, the reaction mixture was cooled with ice and quenched with ice.
  • 6-Bromo-2-(4-iodo-2-methoxy-pyridin-3-yl)-4-methyl-lH-benzimidazole To a solution of 5-bromo-3-methyl-l,2-phenylenediamine (4 g, 19.9 mmol) in methanol (80 mL) was added 4-iodo-2-methoxy-pyridine-3-carbaldehyde (5.23 g, 19.9 mmol) in methanol (20 mL) dropwise at 0°C. The resulting slurry was stirred for 30 min at room temperature. Then iodine (2.53 g, 9.95 mmol) in methanol (20 mL) was added via a dropping funnel.
  • the reaction mixture was heated to reflux for 14 h under nitrogen and then cooled to room temperature. After removal of the solvent, the residue was diluted with ethyl acetate and washed with water. The aqueous fraction was extracted with ethyl acetate and the combined organic layers were washed with water and brine, dried over Na 2 SO 4 . After concentration in vacuo, the residue was purified by flash column chromatography (30% EtOAc / hexane) to yield the title compound (1 g, 73%) as a white solid.
  • 2-(3-Amino-5-fluoro-2-nitro-phenyl)-malonic acid di-tert-butyl ester To the crude 2-(3,5-Difluoro-2-nitro-phenyl)-malonic acid di-tert-butyl ester (62g, 0.42 mol) was added 700 mL of 2M ammonia in methanol in a pressure bottle. The vessel was sealed and heated to 85°C for 18 hours. The reaction mixture was cooled and the vessel opened carefully and the methanol solution concentrated to provide 140 g of crude material.
  • 3-Amino-5-fluoro-2-nitro phenyl acetic acid To the 2-(3-Amino-5-fluoro-2-nitro- phenyl)-malonic acid di-tert-butyl ester (140g) in 500 mL of 4N HCl in dioxane was added 50 mL of water and heated to 40°C for 2 days. The solution was extracted with ethyl acetate (3X's) and the ethyl acetate washed with water (3X's) and brine.
  • 2-Amino-4-fluoro-6-methyI nitrobenzene To the crude 3-Amino-5-fluoro-2-nitro phenyl acetic acid (3.6g, 16.8 mmol) was added Cu 2 O (lO.lg, 70.6 mmol) in 120 mL of acetonitrile along with 50uL of methanol and the suspension was refluxed for 12 hours. The reaction mixture was filtered through Celite and the Celite pad washed with water and ethyl acetate. The filtrate was extracted with ethyl acetate, washed with water and brine, dried over Na 2 SO 4 and concentrated to give 2.95 g of material which by ⁇ NMR was 80% pure.
  • 2-(4-Iodo-2-methoxy-pyridin-3-yl)-4-methyl-6-morpholin-4-yl-lH- benzoimidazole 2-Methyl-4-mo ⁇ holin-4-yl-6-nitro-phenylamine (15.2 g, 64 mmol) was suspended in methanol (200 ml) in a PARR flask. Palladium on carbon (1.0 g, 10% Pd) was added and the suspension shaken under 60 psi of hydrogen overnight.
  • the mixture was filtered through a pad of celite (under argon) into a 3-neck flask, the celite rinsed with methanol and the filtrate diluted with methanol to a total volume of 500 ml and cooled to 0°C.
  • a solution of 4-Iodo-2-methoxy-pyridine-3 -carbaldehyde (14.6 g, 55.5 mmol) in methanol (500 ml) was added slowly (during 3 hours). After addition of ⁇ l A of the solution the system was opened to air and stirred over the weekend, thereby reaching room temperature.
  • the resulting solution was then stirred at 0 °C for an additional 30 min,. the cooling bath was removed, and the reaction mixture was allowed to stir at room temperature in the presence of air for 72 h.
  • the resulting solution was concentrated in vacuo and the residue was dissolved in dichloromethane (1500 mL) and the solvent removed in vacuo (repeated 3x). The resulting dark foamy solid was dried under high vacuum.
  • Examples 29-35 were prepared from commercially or readily available diamines which were prepared and condensed with 4-iodo-2-methoxy-pyridine-3-carbaldehyde as described in Scheme UI.
  • LCMS conditions a) YMC C18 S5 4.6 x 50 mm; 0 - 100% gradient over 4 min*; 4 mL/min flow rate b) YMC ODS-A C18 S7 3.0 x 50 mm; 0 - 100% gradient over 2 min*; 5 mL/min flow rate c) YMC C18 S5 4.5 x 50 mm; 0 - 100% gradient over 8 min*; 2.5 mLmin flow rate d) YMC C18 S7 3.0 x 50 mm; 0 - 100% gradient over 3 min*; 5 mL/min flow rate e) YMC ODSA S3 6.0 xl50 mm; 0 - 100% gradient over 5 min*; 1.5 mL/min flow rate f) PHS-PRIMESPHERE C 18 4.6 x 30 mm; 0 - 100% gradient over 2 min* ; 5 mL/min flow rate g) YMC C18 S7 3.0 x 50 mm;
  • (+)-2- ⁇ 4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-2-oxo-l,2-dihydro-pyridin- 3-yl ⁇ -7,N,N,-trimethyl-3H-benzimidazole-5-carboxamidine: 1H NMR (300 MHz, CD 3 OD) ⁇ 7.63 (IH, s), 7.54 (IH, s), 7.18 - 7.41 (5H, m), 6.26 (IH, d, 7 7.
  • (+)-2- ⁇ 4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-2-oxo-l,2-dihydro-pyridin- 3-yl ⁇ -7-methyl-3H-benzimidazole-5-carboxylic acid The ethyl ester (419 mg, 0.90 mmol) obtained above was diluted with methanol (15 mL) and water (5 mL) followed by addition of sodium hydroxide (180 mg, 4.5 mmol). The mixture was stirred at room temperature for 14 h. After removal of methanol, the residue was neutralized with 2 N HCl solution. The resulting slurry was filtered and washed with ice-cold water.
  • Example 520 (General procedure for examples 520-522)
  • the trifluoroaceticacid salt of the pure title compound was dissolved in methanol and applied to a Varian Mega Bond-Elute SCX cartridge. Elution with methanol followed by 2.0 M NH3 / MeOH gave the free base. This material was suspended in MeOH and 1.00 N aqueous HCl (2 equiv.) was added.
  • the reaction was heated at 80 °C for 12 h.
  • the reaction mixture was then purified on reverse phase preparative HPLC using a methanol / water / 0.1% trifluoroacetic acid gradient. The fractions were evaporated to give the title compound as a trifluoroacetic acid salt, which was dissolved in methanol and applied to a Varian Mega Bond-Elute SCX cartridge. Elution with methanol followed by 2.0 M NH3 / MeOH gave the free base (39.3 mg). This material was suspended in MeOH and 1.00 N aqueous HCl (2 equiv.) was added.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The present invention provides compounds of formula I[ ] and pharmaceutically acceptable salts thereof. The formula I compounds inhibit tyrosine kinase enzymes thereby making them useful as anti-cancer agents. The formula I compounds are also useful for the treatment of other diseases which can be treated by inhibiting tyrosine kinase enzymes.

Description

NOVEL TYROSINE KINASE INHIBITORS
RELATED APPLICATIONS
This application claims the benefit under 35 U.S.C. Section 119(e) of U.S. Provisional Patent Application No. 60/279,327 filed March 28, 2001.
FIELD OF INVENTION The present invention relates generally to the field of tyrosine kinase enzyme inhibition using novel small molecules.
BACKGROUND OF THE INVENTION
Tyrosine Kinases are a class of enzymes, which catalyze the transfer of the terminal phosphate of adenosine triphosphate to the phenolic hydroxyl group of a tyrosine residue present in the target protein. Tyrosine kinases play a critical role in signal transduction for several cellular functions including cell proliferation, carcinogenesis, apoptosis, and cell differentiation (Plowman, G. D.; Ullrich, A.; Shawver, L. K.: Receptor Tyrosine Kinases As Targets For Drug Intervention. DN&P (1994) 7: 334-339). Therefore inhibitors of these enzymes would be useful for the treatment or prevention of proliferative diseases which are dependent on these enzymes. Strong epidemiologic evidence suggests that the overexpression or activation of receptor protein tyrosine kinases leading to constitutive mitogenic signaling is an important factor in a growing number of human malignancies. Tyrosine kinases that have been implicated in these processes include Abl, CDK's, EGF, EMT, FGF, FAK, Flk-1/KDR, HER-2, IGF-1R, IR, LCK, MET, PDGF, Src, and NEGF (Traxler, P.M. Protein Tyrosine Kinase Inhibitors in Cancer Treatment. Exp. Opin. Ther. Patents (1997) 7: 571-588; incorporated herein by reference). Hence, there is an ongoing need to investigate novel compounds that can be used to regulate or inhibit tyrosine kinase enzymes.
SUMMARY OF THE INVENTION
The present invention relates to compounds which inhibit tyrosine kinase enzymes, compositions which contain tyrosine kinase inhibiting compounds and methods of using inhibitors of tyrosine kinase enzymes to treat diseases which are characterized by an overexpression or upregulation of tyrosine kinase activity such as cancer, diabetes, restenosis, arteriosclerosis, psoriasis, angiogenic diseases and immunologic disorders (Powis, G.; Workman, P. Signaling targets For The Development of Cancer Drugs. Anti-Cancer Drug Design (1994), 9: 263-277;
Merenmies, J.; Parada, L. F.; Henkemeyer, M. Receptor Tyrosine Kinase Signaling in Vascular Development. Cell Growth Differ (1997) 8: 3-10; Shawver, L. K.; Lipsosn, K. E.; Fong, T. A. T.; McMahon, G.; Plowman, G. D.; Strawn, L. M. Receptor Tyrosine Kinases As Targets For Inhibition of Angiogenesis. Drug Discovery Today (1997) 2: 50-63; all herein incorporated by reference).
In addition to being used as single agents, it is contemplated that tyrosine kinase inhibitors can enhance the activity of cytotoxic or cytostatic treatments when used in combination with standard therapies known in the art.
The present invention is directed to compounds having formula I
Figure imgf000003_0001
its enantiomers, diastereomers, pharmaceutically acceptable salts, hydrates, prodrugs and solvates thereof;
wherein
X is selected from the group consisting of N, C, C1-C3 alkyl, C1-C3 alkyl substituted with one or more R , and a direct bond;
Y is selected from the group consisting of O and S ;
W is selected from the group consisting of N, C, O, and S, provided that when W is O or S, R9 is absent; R1, R2, R3, R4, R5, R6, R7, R8, R9 are each independently selected from the group consisting of H, Cι_6 alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, halo, amino, OR60, NO2, OH, SR60, NR60R61 , CN, CO2R60, CONR60R61, CO2NR60R61 , NR62CONR60R61, NR60SO2R61 , SO2NR60R61, C(NR62)NR60R61, aryl, heteroaryl, (CH2)nOR60, (CH2)nNR60R61 , (CH2)nSR60, (CH2)n aryl, (CH2)n heteroaryl, , (CH2)n heterocycloalkyl, NH-Z-aryl, and NH-Z-heteroaryl; wherein n is 1 to 3; and
Z is selected from the group consisting of Ci - C4 alkyl, alkenyl, and alkynyl chain; Z having one or more hydroxy, thiol, alkoxy, thioalkoxy, amino, halo, NR60SO2R61 groups; Z optionally incorporating one or more groups selected from the group consisting of CO, CNOH, CNOR60, CNNR60, CNNCOR60 and CNNSO2R60 ; and
R60, ^6i an(j ^62 ^& independently selected from the group consisting of H, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, hydroxy, alkoxy, aryl, heteroaryl, heteroarylalkyl, and alkyl-R25 wherein
R25 is hydrogen, alkenyl, hydroxy, thiol, alkoxy, thioalkoxy, amino, alkylamino, dialkylamino, aryl, heteroaryl, cyano, halo, sulfoxy, sulfonyl, - NR30COOR31, -NR30C(O)R31,
Figure imgf000004_0001
-C(O)NR30R31, heteroaryl or heterocycloalkyl; and
R30 and R31 are, independently, hydrogen, alkyl, cycloalkyl, or alkyl-R25.
In preferred embodiments, R1, R7, R8and R9 are H;
R2 and R4 are H or F;
Y is O; X is selected from the group consisting of N and CH;
W is N;
R5 is selected from the group consisting of H, methyl, ethyl, isopropyl, secondary butyl, cyclopropyl, F, and CF3;
R6 is selected from the group consisting of H, 2-aminomethylpyridine, NHCH(CH2OH)CH2Ph, NHCH2CH(OH)aryl, and NHCH(CH2OH)CH2aryl; and R3 is selected from the group consisting of OR60, C(NH)NHR60, C(O)NHR60 imidazole, imidazoline, tetrahydropyrimidine, piperazine, morpholine, homomorpholine, piperidine, pyrrolidine, homopiperazine and amino; wherein
R60 is selected from the group consisting of H, alkyl, cycloalkyl, heterocycloalkyl, and alkyl-R wherein R is hydrogen, alkenyl, hydroxy, thiol, thioalkoxy, alkoxy, thioalkoxy, halo, cyano, sulfoxy, sulfonyl, -NR COOR , - NR30C(O)R31, -NR30SO2R31, -C(O)NR30R31, or a heteroaryl or heterocycloalkyl; and
R3o and R31 are, independently, hydrogen, alkyl, cycloalkyl, or alkyl-R .
The invention also provides a pharmaceutical composition comprising a compound of formula I, as defined above, and a pharmaceutically acceptable carrier.
The invention further provides a pharmaceutical composition comprising a compound of formula I, as defined above, in combination with pharmaceutically acceptable carrier and at least one other anti-cancer agent optionally formulated as a fixed dose. Additionally provided is a method of treating a condition associated with at least one tyrosine kinase enzyme comprising administering to a mammalian species in need of such treatment an effective amount of a compound of formula I, as defined above. Furthermore, the invention provides a method of treating a condition associated with at least one tyrosine kinase enzyme comprising administering to a mammalian species at least one other anti-cancer agent in combination with a compound of formula I, as defined above.
DESCRIPTION
The present invention provides for compounds of formula I, as defined above, pharmaceutical compositions employing such compounds and methods of using such compounds.
Listed below are definitions of various terms used to describe the compounds of the instant invention. These definitions apply to the terms as they are used throughout the specification (unless they are otherwise limited in specific instances) either individually or as part of a larger group. The term "alkyl" herein alone or as part of another group refers to a monovalent alkane (hydrocarbon) derived radical containing from 1 to 12 carbon atoms unless otherwise defined. An alkyl group is an optionally substituted straight, branched or cyclic saturated hydrocarbon group. When substituted, alkyl groups may be substituted with up to four substituent groups, R as defined, at any available point of attachment. When the alkyl group is said to be substituted with an alkyl group, this is used interchangeably with "branched alkyl group". Exemplary unsubstituted such groups include methyl, ethyl, propyl, isopropyl, n-butyl, t-butyl, isobutyl, pentyl, hexyl, isohexyl, heptyl, 4,4-dimethylpentyl, octyl, 2,2,4-trimethylpentyl, nonyl, decyl, undecyl, dodecyl, and the like. Exemplary substituents may include but are not limited to one or more of the following groups: hydroxy, halo (such as F, CI, Br, I), haloalkyl (such as CCI3 or CF3), alkoxy, alkylthio, cyano, carboxy (-COOH), alkylcarbonyl (-C(O)R), alkoxycarbonyl (-OCOR), amino, carbamoyl (-NHCOOR or -OCONHR), urea (-NHCONHR), thiol, (-SH), sulfoxy, sulfonyl, aryl, heteroaryl, and heterocycloalkyl. Alkyl groups as defined may also comprise one or more carbon to carbon double bonds or one or more carbon to carbon triple bonds. Alkyl groups may also be represented by the formula alkyl-R25. In preferred embodiments, the alkyl group is a methyl, ethyl, propyl or butyl group and include substituted methyl, ethyl, propyl or butyl groups. The term "alkenyl" herein alone or as part of another group refers to a hydrocarbon radical straight, branched or cyclic containing from 2 to 12 carbon atoms and at least one carbon to carbon double bond. An alkenyl group may be optionally substituted in the same manner as described for an alkyl group.
The term "alkynyl" herein alone or as part of another group refers to a hydrocarbon radical straight, branched or cyclic containing from 2 to 12 carbon atoms and at least one carbon to carbon triple bond. An alkynyl group may be optionally substituted in the same manner as described for an alkyl group.
The term "alkoxy" as used alone or in combination herein refers to a straight or branched chain alkyl group covalently bonded to the parent molecule through an oxygen atom linkage containing from one to ten carbon atoms and the terms "C].6 alkoxy" and "lower alkoxy" refer to such groups containing from one to six carbon atoms, examples include, but are not limited to, methoxy, ethoxy, propoxy, isopropoxy, butoxy, t-butoxy and the like. The term "optionally substituted" when used in connection with an alkoxy substituent refers to the replacement of up to two hydrogens, preferably on different carbon atoms with a radical selected form the group of lower alkyl, phenyl, cyano, halo, trifluoromethyl, nitro, hydroxy, alkanoyl, amino, monoalkyl amino and dialkylamino. Alkoxy groups may be substituted in the same manner that alkyl groups can be substituted as described above.
The term "sulfoxy" herein alone or as part of a group refers to -SO and may be substituted with, for example, alkyl or aryl groups.
The term "sulfonyl" herein alone or as part of a group refers to -SO2 and may be substituted with alkyl or aryl groups.
The term "amino" herein alone or as part of another group refers to -NH . An "amino" may optionally be substituted with one or two substituents, which may be the same or different, such as alkyl, aryl, arylalkyl, alkenyl, alkynyl, heteroaryl, heteroarylalkyl, cycloheteroalkyl, cycloheteroalkylalkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, hydroxyalkyl, alkoxyalkyl or thioalkyl. Preferred substituents include alkylamino and dialkylamino, such as methylamino, ethylamino, dimethylamino, and diethylamino. These substituents may be further substituted with a carboxylic acid or any of the alkyl or aryl substituents set out herein. In addition, the amino substituents may be taken together with the nitrogen atom to which they are attached to form 1- pyrrolidinyl, 1-piperidinyl, 1-azepinyl, 4-morpholinyl, 4-thiamorpholinyl, 4- sulfoxymorpholine, 4-sulfonylmorpholine, 1-piperazinyl, 4-alkyl-l-piperazinyl, 4- arylalkyl-1-piperazinyl, 4-diarylalkyl- 1-piperazinyl , 1-homopiperazinyl, 4-alkyl-l- homopiperazinyl, 4-arylalkyl- 1-homopiperazinyl, 4-diarylalkyl- 1-homopiperazinyl; 1- pyrrolidinyl, 1-piperidinyl, or 1-azepinyl, optionally substituted with alkyl, alkoxy, alkylthio, halo, trifluoromethyl or hydroxy .
The term "aryl" herein alone or as part of another group refers to monocyclic or bicyclic aromatic rings, e.g. phenyl, substituted phenyl and the like, as well as groups which are fused, e.g., napthyl, phenanthrenyl and the like. An aryl group thus contains at least one ring having at least 6 atoms, with up to five such rings being present, containing up to 22 atoms therein, with alternating (resonating) double bonds between adjacent carbon atoms or suitable heteroatoms. Aryl groups may optionally be substituted with one or more groups including, but not limited to halogen, alkyl, alkenyl, alkynyl, alkoxy, hydroxy, carboxy, carbamoyl, alkyloxycarbonyl, alkylaminocarbonyl, nitro, trifluoromethyl, amino, cycloalkyl, cyano, alkyl S(O)m (m=O, 1, 2), or thiol. Aryl groups may also be substituted with heterocycloalkyl and heterocycloaryl groups to form fused rings, such as dihydrobenzfuranyl, oxindolyl, indolyl, indolinyl, oxindolyl, benzoxazolidinonyl, benzoxazolinyl and benzoxazolidinone.
The term "cycloalkyl" herein alone or as part of another group refers to fully saturated and partially unsaturated hydrocarbon rings of 3 to 9, preferably 3 to 7 carbon atoms. Further, a cycloalkyl may be substituted. A substituted cycloalkyl refers to such rings having one, two, or three substituents, preferably one, selected from the group consisting of halo, alkyl, substituted alkyl, alkenyl, alkynyl, nitro, cyano, oxo (=O), hydroxy, alkoxy, thioalkyl, -CO2H, -OC(=O)H, CO2-alkyl, - OC(=O)alkyl, =N-OH, =N-O-alkyl, aryl, heteroaryl, heterocyclo, a five or six membered ketal (i.e. 1,3-dioxolane or 1,3-dioxane), -NR'R", -C(=O)NR'R", - OC(=O)NR'R", -NR'CO2'R", -NR'C(=O)R", -SO2NR'R", and -NR'SO2'R", wherein each of R' and R" is independently selected from hydrogen, alkyl, substituted alkyl, and cycloalkyl, or R' and R" together form a heterocyclo or heteroaryl ring. Cycloalkyl groups may also be substituted with hetero atoms such as O, N, and S to form heterocycloalkyl groups. Preferred heterocycloalkyl groups include optionally substituted morpholine, homomorpholine (7 membered ring), hiomorpholine, piperazine, homopiperazine (7 membered ring), and piperidine.
The term "heteroaryl" herein alone or as part of another group refers to substituted and unsubstituted aromatic 5 or 6 membered monocyclic groups, 9 or 10 membered bicyclic groups, and 11 to 14 membered tricyclic groups which have at least one heteroatom (O, S or N) in at least one of the rings. Each ring of the heteroaryl group containing a heteroatom can contain one or two oxygen or sulfur atoms and/or from one to four nitrogen atoms provided that the total number of heteroatoms in each ring is four or less and each ring has at least one carbon atom. The fused rings completing the bicyclic and tricyclic groups may contain only carbon atoms and may be saturated, partially saturated, or unsaturated. The nitrogen and sulfur atoms may optionally be oxidized and the nitrogen atoms may optionally be quaternized. Heteroaryl groups which are bicyclic or tricyclic must include at least one fully aromatic ring but the other fused ring or rings may be aromatic or non- aromatic. The heteroaryl group may be attached at any available nitrogen or carbon atom of any ring. The heteroaryl ring system may contain zero, one, two or three substituents selected from the group consisting of halo, alkyl, substituted alkyl, alkenyl, alkynyl, nitro, cyano, hydroxy, alkoxy, thioalkyl, -CO2H, -OC(=O)H, -CO2- alkyl, -OC(=O)alkyl, phenyl, benzyl, phenylethyl, phenyloxy, phenylthio, cycloalkyl, substituted cycloalkyl, heterocyclo, heteroaryl, -NR'R", -C(=O)NR'R", - OC(=O)NR'R", -NR'CO2'R", -NR'C(=O)R", -SO2NR'R", and -NR'SO2R", wherein each of R' and R" is independently selected from hydrogen, alkyl, substituted alkyl, and cycloalkyl, or R' and R" together form a heterocyclo or heteroaryl ring. Exemplary monocyclic heteroaryl groups include pyrrolyl, pyrrolidinyl, imidazolinyl, pyrazolyl, pyrazolinyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl, thiadiazolyl, isothiazolyl, furanyl, thienyl, oxadiazolyl, pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, triazinyl, and the like. Exemplary bicyclic heteroaryl groups include indolyl, indolinyl, oxindolyl, benzoxazolidinone, benzothiazolyl, benzodioxolyl, benzoxazolyl, benzothienyl, quinolinyl, tetrahydroisoquinolinyl, isoquinolinyl, benzimidazolyl, benzopyranyl, indolizinyl, benzofuranyl, chromonyl, coumarinyl, benzopyranyl, cinnolinyl, quinoxalinyl, indazolyl, pyrrolopyridyl, furopyridinyl, dihydroisoindolyl, tetrahydroquinolinyl and the like.
Exemplary tricyclic heteroaryl groups include carbazolyl, benzindolyl, phenanthrollinyl, acridinyl, phenanthridinyl, xanthenyl and the like.
The term "halogen" or "halo" herein alone or as part of another group refers to chlorine, bromine, fluorine or iodine selected on an independent basis. The term "hydroxy" herein alone or as part of another group refers to -OH.
The term "thioalkoxy" herein alone or as part of another group refers to an alkyl group as defined herein attached to the parent molecular group through a sulfur atom. Examples of thioalkoxy include, but are not limited to, thiomethoxy, thioethoxy, and the like. Abbreviations: "Ph" represents phenyl; "Me" represents methyl; and "Et" represents ethyl. An "anti-cancer agent" as used herein includes known anti-cancer treatments such as radiation therapy or with cytostatic or cytotoxic agents, such as for example, but not limited to, DNA interactive agents, such as cisplatin or doxorubicin; topoisomerase II inhibitors, such as etoposide; topoisomerase I inhibitors such as CPT-11 or topotecan; tubulin interacting agents, such as paclitaxel, docetaxel or the epothilones; hormonal agents, such as tamoxifen; thymidilate synthase inhibitors, such as 5-fluorouracil; anti-metabolites, such as methotrexate; tyrosine kinase inhibitors such as Iressa and OSI-774; angiogenesis inhibitors; EGF inhibitors; VEGF inhibitors; CDK inhibitors; Her 1/2 inhibitors and monoclonal antibodies directed against growth factor receptors such as erbitux (EGF) and herceptin (Her2).
When a functional group is termed "protected", this means that the group is in modified form to preclude undesired side reactions at the protected site. Suitable protecting groups for the compounds of the present invention will be recognized from the present application taking into account the level of skill in the art, and with reference to standard textbooks, such as Greene, T. W. et al., Protective Groups in Organic Synthesis, Wiley, N.Y. (1991).
When C 1-6 alkyl, alkenyl, alkynyl, cycloalkyl are substituted, they are preferably substituted with one or more hydroxy, cyano, carbamoyl, hydroxy, alkoxy, thiol, alkenyl, thioalkoxy, amino, alkylamino, amido, sulfonyl, sulfoxy, sulfonamido, halo, heterocycloalkyl, aryl or heteroaryl.
When aryl or heteroaryl are substituted, they are preferably substituted with one or more alkyl, alkenyl, alkynyl, cyano, carbamoyl, hydroxy, alkoxy, thioalkoxy, amino, amido, sulfonamido, halo or with R', R" wherein R', R" form a ring that is fused to the aryl group. When CH aryl or heteroaryl are substituted, they are preferably substituted with one or more alkyl, alkyenyl, alkynyl, cyano, carbamoyl, hydroxy, alkoxy, thioalkoxy, amino, amido, sulfonamido, or halogen.
When NH-Z-aryl or NH-Z-heteroaryl groups are substituted, they are preferably substituted with one or more alkyl, alkenyl, alkynyl, hydroxy, alkoxy, thioalkoxy, amino, halogen, nitro, nitrile, carboxylate, alkoxycarbonyl, carbamoyl, ester, amide, aryl, or heteroaryl
The numbers in the subscript after the symbol "C" define the number of carbon atoms a particular group can contain. For example "Ci .5 alkyl" means a straight or branched saturated carbon chain having from one to six carbon atoms; examples include methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl, t- butyl, n-pentyl, sec-pentyl, isopentyl, and n-hexyl. Depending on the context, "Ci .β alkyl" can also refer to Ci .5 alkylene which bridges two groups; examples include propane- 1, 3 -diyl, butane- 1,4-diyl, 2-methyl-butane-l,4-diyl, etc. "C2-6 alkenyl" means a straight or branched carbon chain having at least one carbon-carbon double bond, and having from two to six carbon atoms; examples include ethenyl, propenyl, isopropenyl, butenyl, isobutenyl, pentenyl, and hexenyl. Depending on the context, "C2-6 alkenyl" can also refer to C2-6 alkenediyl which bridges two groups; examples include ethylene- 1,2-diyl (vinylene), 2-methyl-2-butene- 1,4-diyl, 2-hexene-l,6-diyl, etc. "C2-6 alkynyl" means a straight or branched carbon chain having at least one carbon-carbon triple bond, and from two to six carbon atoms; examples include ethynyl, propynyl, butynyl, and hexynyl.
The term "alkyl-R25" includes optionally substituted alkyl groups such as methyl, ethyl, propyl, and butyl, attached to an R25group. R 5generally includes hydrogen, alkenyl, hydroxy, thiol, alkoxy, thioalkoxy, amino, alkylamino, dialkylamino, aryl, heteroaryl, cyano, halo, sulfoxy, sulfonyl, -NHCOOH, -NHC(O)-, -NHSO2-, -C(O)NH , heteroaryl or heterocycloalkyl groups such as morpholinyl or group having the formula: O O
-O -O " 1 "N-N →^ ^"
Figure imgf000011_0001
The terms "imidazole" and "imidazoline" herein alone or as part of another group includes substituted imidazoles and substituted imidazolines. Similarly, the term "tetrahydropyrimidine" includes substituted tetrahydropyrimidines. Likewise, the terms "piperazine", "piperidine" "morpholines", "homopiperazines",
"homomorpholines" and "pyrrolidine" include substituted piperazines, substituted piperidines, substituted morpholines, substituted homomorpholines and substituted pyrrolidines, respectively.
Compounds of the present invention have the general formula I:
Figure imgf000012_0001
I its enantiomers, diastereomers, pharmaceutically acceptable salts, hydrates, prodrugs and solvates thereof;
wherein
X is selected from the group consisting of N, C, C1-C3 alkyl, C1-C3 alkyl substituted with one or more R , and a direct bond;
Y is selected from the group consisting of O and S ;
W is selected from the group consisting of N, C, O, and S, provided that when W is O or S, R9 is absent;
R1, R2, R3, R4, R5, R6, R7, R8, R9 are each independently selected from the group consisting of H, C 1-6 alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, halo, amino, OR60, NO2, OH, SR60, NR60R61 , CN, CO2R60, CONR60R61, CO2NRo0R61 , NR62CONR60R61, NR60SO2R61 , SO2NRo0R61, C(NR62)NR60R6', aryl, heteroaryl, (CH2)„OR60, (CH2)nNR60RDl , (CH2)nSR60, (CH2)„ aryl, (CH2)n heteroaryl, , (CH2)n heterocycloalkyl, NH-Z-aryl, and NH-Z-heteroaryl; wherein n is 1 to 3; and
Z is selected from the group consisting of - C4 alkyl, alkenyl, and alkynyl chain; Z having one or more hydroxy, thiol, alkoxy, thioalkoxy, amino, halo, NR60SO R61 groups; Z optionally incoφorating one or more groups selected from the group consisting of CO, CNOH, CNOR60, CNNR60 , CNNCOR60 and CNNSO2R60 • and
R , and R ' are independently selected from the group consisting of H, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, hydroxy, alkoxy, aryl, heteroaryl, heteroarylalkyl, and alkyl-R wherein
R is hydrogen, alkenyl, hydroxy, thiol, alkoxy, thioalkoxy, amino, alkylamino, dialkylamino, aryl, heteroaryl, cyano, halo, sulfoxy, sulfonyl, - NR30COOR31, -NR30C(O)R31, -NR30SO2R31, -C(O)NR30R31, heteroaryl or heterocycloalkyl; and
R30 and R31 are, independently, hydrogen, alkyl, cycloalkyl, or alkyl-R25.
In some embodiments of the present invention, R3 is -OR °. R60 is alkyl, or - alkyl-R25, wherein R25 is hydrogen, alkenyl, hydroxy, thiol, alkoxy, thioalkoxy, amino, halo, cyano, alkylsulfoxy, alkylsulfonyl, -R30COOR31, -NR30C(O)R31, - NR30SO2R31, -C(O)NR30R31, heteroaryl or heterocycloalkyl; and R30 and R31 are, S independently, hydrogen, alkyl, cycloalkyl, or alkyl-R . In preferred embodiments, R60 is methyl, -(CH2)nCH2OH, or -(CH2)nCH2N(CH2CH2)2O, and n is 0, 1, or 2.
In some embodiments, R is piperazine, homopiperazine, 3-methylpiperazine, or 3,5-dimethylpiperazine being optionally substituted at the 4-N position with a compound selected from the group consisting of alkyl, aryl, cycloalkyl, cycloalkylalkyl, heterocycloalkyl, alkyl-R25, -C(O)-R15, or -CO2R15 wherein R15 is
9S S hydrogen, alkyl, aryl, alkyl-R , amino or aryl; and R is hydrogen, alkenyl, hydroxy, thiol, alkoxy, thioalkoxy, amino, alkylamino, dialkylamino, cyano, halo, sulfoxy, sulfonyl, arylsulfonyl, -NR30COOR31,
Figure imgf000013_0001
-NR30SO2R31, -C(O)NR30R31, heteroaryl or heterocycloalkyl and R30 and R31 are, independently, hydrogen, alkyl, cycloalkyl, or alkyl-R25. In preferred embodiments, piperazine is substituted with Me, CH2cyclopropyl, CH2CH2NMe2, CH2CH2NEt2, CH2CH2NH2, CH2CH2NHMe, CH2CH2NHEt, N-CH2CH2N(CH2CH2)2θ, (CH2)nCH2-R25 wherein R25 is OH, OMe, F, CN, CF3, SOCH3 or SO2CH3, wherein n is 0, 1, or 2.
In some embodiments, R" is an amino group. Preferred amino groups include NHCH2CH2OH, NMeCH2CH2OH, NEtCH2CH2OH, NHCH2CH2NH2, NMeCH2CH2NH2, NEtCH2CH2NH2, NHCH2CH2NMe2, NMeCH2CH2NMe2, NEtCH2CH2NMe2, NHCH2CH2NEt2, NMeCH2CH2NEt2, NEtCH2CH2NEt2, NHCH2CH2N(CH2CH2)2θ, NMeCH2CH2N(CH2CH2)2O, NEtCH2CH2N(CH2CH2)2θ. In some embodiments, R is an optionally substitued piperidine. Preferred substituents are selected from the group consisting of hydroxy, thiol, amino, alkylamino, dialkylamino, alkoxy, thioalkoxy, 1,3 dioxolane (-OCHR15)2, 1,3 dioxane (-OCHR15CHR15CHR15O-) -NHC(O)R15, -NHCO2R15, wherein R15 is hydrogen, alkyl 9S or alkyl-R wherein R is hydrogen, alkenyl, hydroxy, thiol, alkoxy, thioalkoxy, amino, halo, cyano, alkylsulfoxy, alkylsulfonyl, -NR30COOR31, -NR30C(O)R31, - NR30SO2R31, -C(O)NR30R31, heteroaryl or heterocycloalkyl; and R30 and R31 are, independently, hydrogen, alkyl, cycloalkyl, or alkyl-R25.
In some embodiments R3 is an optionally substituted moφholine, homomorpholine, thiomoφholine, sulfoxymoφholine, or sulfonylmoφholine. Preferred substituents include hydroxy, thiol, amino, alkylamino, dialkylamino, alkoxy, thioalkoxy, alkyl-R25, -NHC(O)R15, -NHCO2R15, wherein R15 is hydrogen,
9S 9S alkyl or alkyl-R wherein R is hydrogen, alkenyl, hydroxy, thiol, alkoxy, thioalkoxy, amino, halo, cyano, alkylsulfoxy, alkylsulfonyl, -NR30COOR31, - NR30C(O)R31, -NR30SO2R31, -C(O)NR30R31, heteroaryl or heterocycloalkyl; and R30 and R" are, independently, hydrogen, alkyl, cycloalkyl, or alkyl-R . In some embodiments, R3 is a pyrrolidine. Preferred pyrrolidines include, 3- hydroxyl pyrrolidine, 3- alkoxy pyrrolidine, and 3-alkylamino pyrrolidine.
According to one embodiment of the present invention, R3 is an optionally substituted N-tetrahydropyrimidine or N-imidazoline wherein the substituents are, preferably, alkyl, hydroxyalkyl, alkoxyalkyl, haloalkyl, cyanoalkyl, carboxyl, or carboxamide.
In some embodiments, R6 is is selected from the group consisting of H, 2- aminomethylpyridine, NHCH2CH(OH)aryl, and NHCH(CH2OH)CH2aryl, wherein the aryl group is optionally substituted. In preferred embodiments, the aryl group is substituted with Br, CI, F, or methoxy. In some embodiments, R6 has one of the following formulae:
Figure imgf000015_0001
wherein R ,40 is hydrogen or alkyl, preferably methyl, and R 17 is hydrogen or halogen, such as Br, CI or F.
Preferred compounds of the present invention have one of the following formulae:
Figure imgf000016_0001
wherein
19 I ^ 9*ϊ R and R are, independently, hydrogen, alkyl, or alkyl-R ;
R15is hydrogen, alkyl, or alkyl-R25; R1 is independently, hydrogen or methyl;
17 18 I Q 18
R , R and R are, independently, hydrogen, halogen, or alkoxy,or R and R19 together form a heterocycloalkyl or heteroaryl group; R25 is hydrogen, hydroxy, thiol, alkenyl, alkoxy, thioalkoxy, amino, halo, cyano, sulfoxy, sulfonyl, -NR30COOR31, -NR30C(O)R31, -NR30SO2R31, - C(O)NR30R31, heteroaryl or heterocycloalkyl; and
R and R are, independently, hydrogen, alkyl, cycloalkyl or alkyl-R . In preferred embodiments, R12 is hydrogen, methyl, hydroxymethyl, methoxymethyl, CH2F, CH2CN, CO2H, or -CONR30R31 wherein R30 and R31 are, independently, hydrogen, or alkyl-R25;
Rl is H; R17 is Br, CI or F;
R18 is halo or methoxy; and
R19 is H.
Suitable examples of salts of the compounds according to the invention include inorganic or organic acids. These include, but are not limited to, hydrochloride, hydrobromide, sulfate, methanesulfonate, maleate, fumarate, phosphate and other pharmaceutically acceptable salts. Salts which are unsuitable for pharmaceutical uses but which can be employed, for example, for the isolation or purification of free compounds of formula I or their pharmaceutically acceptable salts, are also included. All stereoisomers of the compounds of the instant invention are contemplated, either in admixture or in pure or substantially pure form. The definition of the compounds according to the invention embraces all possible stereoisomers and their mixtures. It very particularly embraces the racemic forms and the isolated optical isomers having the specified activity. The racemic forms can be resolved by physical methods, such as, for example, fractional crystallization, separation or crystallization of diastereomeric derivatives or separation by chiral column chromatography. The individual optical isomers can be obtained from the racemates by conventional methods, such as, for example, salt formation with an optically active acid followed by crystallization. It should be understood that the present invention includes prodrug forms of the compounds of formula I. Various forms of prodrugs are well known in the art. For examples of such prodrug derivatives, see:
(a) Design of Prodrugs, edited by H. Bundgaard (Elsevier, 1985); and Methods in Enzymology, Vol. 42, pp. 309-396, edited by K. Widder et al., (Academic Press, 1985);
(b) A Textbook of Drug Design and Development, edited by Krosgaard-Larsen and H. Bundgaard, Chapter 5, "Design and Application of Prodrugs," by H. Bundgaard, pp. 113-191 (1991);
(c) H. Bundgaard, Advanced Drug Deliver Reviews, 8, pp. 1-38 (1992);
(d) H. Bundgaard et al., Journal of Pharmaceutical Sciences, 77, 285 (1988); and
(e) N. Kayeka et al., Chem. Phar. Bull, 32, 692 (1984). The invention also provides a pharmaceutical composition comprising a compound of formula I, as defined above, and a pharmaceutically acceptable carrier and at least one other anti-cancer agent formulated as a fixed dose. Preferred anticancer agents are selected from the group consisting of: tamoxifen, toremifen, raloxifene, droloxifene, iodoxyfene, megestrol acetate, anastrozole, letrazole, borazole, exemestane, flutamide, nilutamide, bicalutamide, cyproterone acetate, goserelin acetate, luprolide, finasteride, herceptin, methotrexate, 5-fluorouracil, cytosine arabinoside, doxorubicin, daunomycin, epirubicin, idarubicin, mitomycin-C, dactinomycin, mithramycin, cisplatin, carboplatin, melphalan, chlorambucil, busulphan, cyclophosphamide, ifosfamide, nitrosoureas, thiotephan, vincristine, taxol, taxotere, etoposide, teniposide, amsacrine, irinotecan, topotecan, an epothilone; a tyrosine kinase inhibitor such as Iressa or OSI-774; an angiogenesis inhibitor; an EGF inhibitor; a VEGF inhibitor; a CDK inhibitor; a Herl/2 inhibitor and monoclonal antibodies directed against growth factor receptors such as erbitux (EGF) and herceptin (Her2). The invention further provides a method of treating a condition via modulation of at least one tyrosine kinase enzyme comprising administering to a mammalian species in need of such treatment an effective amount of a compound of formula I, as defined above.
Additionally, the invention provides a method of treating a condition via modulation of at least one tyrosine kinase enzyme comprising administering to a mammalian species in need of such treatment an effective amount of a compound of formula I, as defined above, in combination (simultaneously or sequentially) with at least one other anti-cancer agent.
A preferred condition, treated by said methods of the instant invention, is cancer. Additionally, the tyrosine kinase enzyme may include (but is not limited to): Abl, CDK's, EGF, EMT, FGF, FAK, Flk-1/KDR, HER-2, IGF-1R, IR, LCK, MET, PDGF, Src, and VEGF. The invention also provides a method for treating cancer, comprising administering to a mammalian species in need of such treatment, a therapeutically effective amount of at least one of the pharmaceutical compositions defined above. The invention further provides a method for treating proliferative diseases, comprising administering to a mammalian species in need of such treatment a therapeutically effective amount of at least one of the pharmaceutical compositions defined above.
Certain compounds of formula I may generally be prepared according to the following schemes and the knowledge of one skilled in the art. Solvates (e.g., hydrates) of the compounds of formula I are also within the scope of the present invention. Methods of solvation are generally known in the art. Accordingly, the compounds of the instant invention may be in the free or hydrate form, and may be obtained by methods exemplified by the following schemes below. More specifically, Schemes I-Vπ illustrate the preparation of compounds claimed in this invention. The examples, which follow, illustrate the compounds that can be synthesized by these schemes. The schemes are not limited by the examples listed or by any substituents employed for illustrative puφoses.
Scheme I describes the preparation of the benzimidazoles. The starting diamines 1 are readily available using literature methods or are obtained commercially. The diamine is then condensed with an aldehyde 2 to provide the benzimidazole 3. Further modification of the functional groups on the aryl group of the benzimidazole or heterocycle of 3 are then envisioned.
Scheme I.
Figure imgf000019_0001
1 Alternatively, the benzimidazole could be formed in a step-wise manner (see Scheme II) by amide formation using the acid chloride of 5 or any of the commonly used peptide coupling reagents such as DCC (dicyclohexylcarbodiimide), EDCI (l-(3- dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride), etc. Once the amide 6 was formed the nitro group could be reduced using catalytic hydrogenation, transfer hydrogenation or chemical reduction such as SnCl2 or iron powder or other methods known in the art for reduction of aryl nitro groups. Treatment of the aniline with acid would then form the benzimidazole.
Scheme II.
Figure imgf000020_0001
For example, Scheme III illustrates the use of 4-iodo-2-methoxy-pyridine-3- carbaldehyde 7 to provide the functionalized benzimidazole 8. Hydrolysis of the methoxy group using protic acid conditions, TMSI (trimethylsilyl iodide), BBr3, or other conditions known in the art for cleaving a methyl ether would provide the halopyridone 9. Addition of heteroatom nucleophiles using amines, alcohols or thiols would then provide the substituted pyridones 10. Other functionality could be incoφorated into the aldehyde and the above example is included for illustrative puφoses only. Scheme III
Figure imgf000021_0001
9 10
Likewise the aryl ring of the benzimidazole prepared using Schemes I or II can be modified. For example introduction of a cyano group for R3 on the benzimidazole allows for the formation of heterocycles such as imidazole, imidazolines, oxazolines, thiazolines, amides, or amidines. Scheme IV illustrates such transformations. Starting from the cyano-substituted benzimidazole 11 the heterocycle can be modified as illustrated in Scheme IV to provide 12. Imidate formation preferably using ethanol and acid provides intermediate 13. Imidate 13 can be transformed using diamines to form imidazolines, amino alcohols to form oxazolines, amino acetals to form imidazoles, and amino thiols to form thiazolines 14. Alternatively the imidate can be hydrolyzed to the acid and coupled with amines using any of the standard amide formation reagents (DCC, EDCI, etc.) to form amides 15. Imidate 13 is also a useful intermediate for the preparation of amidines 16 by reacting with amines.
Scheme IV
Figure imgf000022_0001
15
X= NH, 0, S
Scheme V illustrates further transformation of benzimidazoles that bear a halogen atom using palladium catalysis using conditions developed by Suzuki [Yang et al. Acta Chem. Scand. (1993) 221; Suzuki et al. Synth. Commun. (1981) 11: 513] or Buchwald/Hartwig [Buchwald et al. J. Am. Chem. Soc. (1994) 116: 7901; Hartwig et al. J. Am. Chem. Soc. (1994) 116: 5969; Hartwig. Angew. Chem., Int. Ed. Engl (1998) 37: 2046] and variations of these methods. Preparation of a bromide substituted benzimidazole 17 could be envisioned to provide a substrate for Suzuki coupling with aryl, vinyl, and heterocyclic boronic acids to provide benzimidazoles 18. Likewise, amines and heterocycles such as piperazine or moφholine derivatives 19 can be prepared from the same bromide using amines under conditions described by Buchwald and Hartwig or variations thereof. Scheme V
Figure imgf000023_0001
18 19
X = NH, O, S
Alternatively amine and heterocyclic derivatives such as 19 can be prepared using intermediate 6 described in Scheme II. When the R3 of 6 is a halogen, preferably F, the halogen can be displaced with amines, alcohols, heterocyclic amines and other nitrogen containing heterocycles such as piperazine, piperidine, 4-amino piperidine, moφholine, imidazole, etc (Scheme VI). The terminal nitrogen of piperazine or 4-amino piperidine can then be alkylated using standard alkylation conditions or reacted with aldehydes in a reductive amination reaction to provide alkylated derivatives. Alternatively the terminal nitrogen atom of piperazine or 4- amino piperidine can be acylated or carbamoylated using any number of conditions that are routine for someone skilled in the art of organic synthesis. Following the example illustrated in Scheme II compounds such as 19 could be prepared.
Scheme VI.
Figure imgf000023_0002
19 Alternatively amines, heterocycles, and alcohols can be introduced at R3 using a nucleophilic aromatic substitution reaction started from an intermediate 20 were R3 is halogen, preferably F, the halogen can be displaced with amines, alcohols, heterocyclic amines and other nitrogen containing heterocycles such as piperazine,
5 piperidine, 4-amino piperidine, moφholine, imidazole, etc (Scheme VII). The terminal nitrogen of piperazine or 4-amino piperidine can then be alkylated using standard alkylation conditions or reacted with aldehydes in a reductive animation reaction to provide alkylated derivatives. Alternatively the terminal nitrogen atom of piperazine or 4-amino piperidine can be acylated or carbamoylated using any number
) of conditions that are routine for someone skilled in the art of organic synthesis. The resulting nitro aniline could be reduced to the diamine 21 and processed as illustrated in Scheme III.
Scheme VII.
Figure imgf000024_0001
21
20
Utility
The compounds according to the invention have pharmacological properties; in particular, the compounds of formula I are tyrosine kinase enzyme inhibitors. The novel compounds of formula I are thus useful in the therapy of a variety of proliferative diseases (including but not limited to diseases associated with tyrosine kinase enzymes) such as cancer, autoimmune diseases, viral diseases, fungal diseases, neurodegenerative disorders and cardiovascular disease.
More specifically, the compounds of formula I are useful in the treatment of a variety of cancers, including, but not limited to, the following: a) carcinoma, including that of the bladder, breast, colon, kidney, liver, lung, including small cell lung cancer, esophagus, gall bladder, ovary, pancreas, stomach, cervix, thyroid, prostate, and skin, including squamous cell carcinoma; b) hematopoietic tumors of lymphoid lineage, including leukemia, acute lymphocytic leukemia, acute lymphoblastic leukemia, B-cell lymphoma, T-cell lymphoma, Hodgkins lymphoma, non-Hodgkins lymphoma, hairy cell lymphoma and Burkett's lymphoma; c) hematopoietic tumors of myeloid lineage, including acute and chronic myelogenous leukemias, myelodysplastic syndrome and promyelocytic leukemia; d) tumors of mesenchymal origin, including fibrosarcoma and rhabdomyosarcoma; e) tumors of the central and peripheral nervous system, including astrocytoma, neuroblastoma, glioma and schwannomas; and f) other tumors, including sacroma, melanoma, seminoma, teratocarcinoma, osteosarcoma, xenoderoma pigmentosum, keratoctanthoma, thyroid follicular cancer and Kaposi's sarcoma.
Due to the key role of tyrosine kinases in the regulation of cellular proliferation in general, inhibitors could act as reversible cytostatic agents which may be useful in the treatment of any disease process which features abnormal cellular proliferation, e.g., benign prostatic hypeφlasia, familial adenomatosis polyposis, neuro-fibromatosis, atherosclerosis, pulmonary fibrosis, arthritis, psoriasis, glomerulonephritis, restenosis following angioplasty or vascular surgery, hypertrophic scar formation, inflammatory bowel disease, transplantation rejection, endotoxic shock, and fungal infections.
Compounds of formula I may induce apoptosis. The apoptotic response is aberrant in a variety of human diseases. Compounds of formula I, as modulators of apoptosis, will be useful in the treatment of cancer (including but not limited to those types mentioned herein above), viral infections (including but not limited to heφesvirus, poxvirus, Epstein-Barr virus, Sindbis virus and adenovirus), prevention of AIDS development in HIV-infected individuals, autoimmune diseases (including but not limited to systemic lupus, erythematosus, autoimmune mediated glomerulonephritis, rheumatoid arthritis, psoriasis, inflammatory bowel disease, and autoimmune diabetes mellitus), neurodegenerative disorders (including but not limited to Alzheimer's disease, AIDS-related dementia, Parkinson's disease, amyotrophic lateral sclerosis, retinitis pigmentosa, spinal muscular atrophy and cerebellar degeneration), myelodysplastic syndromes, aplastic anemia, ischemic injury associated with myocardial infarctions, stroke and reperfusion injury, arrhythmia, atherosclerosis, toxin-induced or alcohol related liver diseases, hematological diseases (including but not limited to chronic anemia and aplastic anemia), degenerative diseases of the musculoskeletal system (including but not limited to osteoporosis and arthritis) aspirin-sensitive rhinosinusitis, cystic fibrosis, multiple sclerosis, kidney diseases and cancer pain.
Compounds of formula I may modulate the level of cellular RNA and DNA synthesis. These agents would therefore be useful in the treatment of viral infections (including but not limited to HIV, human papilloma virus, heφesvirus, poxvirus, Epstein-Barr virus, Sindbis virus and adenovirus).
Compounds of formula I may also be useful in the chemoprevention of cancer. Chemoprevention is defined as inhibiting the development of invasive cancer by either blocking the initiating mutagenic event or by blocking the progression of pre- malignant cells that have already suffered an insult or inhibiting tumor relapse. Compounds of formula I may also be useful in inhibiting tumor angiogenesis and metastasis.
The compounds of this invention may also be useful in combination (administered together or sequentially) with known anti-cancer treatments such as radiation therapy or with cytostatic or cytotoxic agents, such as for example, but not limited to, DNA interactive agents, such as cisplatin or doxorubicin; topoisomerase II inhibitors, such as etoposide; topoisomerase I inhibitors such as CPT-11 or topotecan; tubulin interacting agents, such as paclitaxel, docetaxel or the epothilones; hormonal agents, such as tamoxifen; thymidilate synthase inhibitors, such as 5-fluorouracil; and anti-metabolites, such as methotrexate; tyrosine kinase inhibitors such as Iressa and OSI-774; angiogenesis inhibitors; EGF inhibitors; VEGF inhibitors; CDK inhibitors; Herl/2 inhibitors and monoclonal antibodies directed against growth factor receptors such as erbitux (EGF) and herceptin (Her2). If formulated as a fixed dose, such combination products employ the compounds of this invention within the dosage range described below and the other pharmaceutically active agent or treatment within its approved dosage range. Compounds of formula I may also be administered sequentially with known anticancer or cytotoxic agents when a combination formulation is inappropriate. The invention is not limited in the sequence of administration; compounds of formula I may be administered either prior to or after administration of the known anticancer or cytotoxic agent(s).
Further subject matter of the invention also includes pharmaceuticals for use, as described above, including controlling cancer, inflammation and arthritis, which contain at least one compound of the formula I as defined above or at least one of its pharmacologically acceptable acid addition salts, and the use of a compound of the formula I as defined above for the preparation of a pharmaceutical having activity against proliferative diseases as described previously including against cancer, inflammation and/or arthritis.
The pharmacological properties of the compounds of this invention may be confirmed by a number of pharmacological assays. The exemplified pharmacological assays which follow have been carried out with the compounds according to the invention and their salts.
Biological Assays
A. CDK 2/cyclin E Kinase Assay Kinase reactions consisted of 5 ng of baculovirus expressed GST-
CDK2/cyclin E complex, 0.5 μg GST-RB fusion protein (amino acids 776-928 of retinoblastoma protein), 0.2 μCi 33P γ-ATP, 25 μM ATP in 50 μl kinase buffer (50 mM Hepes, pH 8.0, 10 mM MgCl2, 1 mM EGTA, 2 mM DTT). Reactions were incubated for 45 minutes at 30° C and stopped by the addition of cold trichloroacetic acid (TCA) to a final concentration of 15% . TCA precipitates were collected onto GF/C unifilter plates (Packard Instrument Co., Meriden, CT) using a Filtermate universal harvester (Packard Instrument Co., Meriden, CT) and the filters were quantitated using a TopCount 96-well liquid scintillation counter (Packard Instrument Co., Meriden, CT). Dose response curves were generated to determine the concentration required inhibiting 50% of kinase activity (IC50). Compounds were dissolved at 10 mM in dimethylsulfoxide (DMSO) and evaluated at six concentrations, each in triplicate. The final concentration of DMSO in the assay equaled 2%. IC50 values were derived by non-linear regression analysis and have a coefficient of variance (SD/mean, n=6) = 14%.
B. EMT Kinase Assay
A filter-based kinase assay, measuring the phosphorylation of Gst-SLP76 by Gst-Emtk, was employed to determine the compound inhibitory activity against Emt. The kinase reaction was performed in a 96-well plate at room temperature for 15 min before being terminated by adding 100 μl of 20% trichloroacetic acid (TCA) containing 0.1 M sodium pyrophosphate. The kinase reaction mixture (60 μl ) contained 25 mM HEPES, pH 7.0, 0.1 mg/ml BSA, 5 mM MgCl2, 5 mM MnCl2, 8 ng of enzyme (Gst-Emtk), 5 μg of the substrate protein (Gst-SLP76), 1 μM ATP, 0.4 μCi of [γ-P ] ATP and the tested compound (at various concentrations). After termination, the proteins were allowed to precipitate in the presence of TCA for 1 fir at 4 °C. The precipitated proteins were then harvested on a filter plate (UniFilter-96, GF/C, Packard Instrument) and washed to remove excess [γ-P ] ATP. The radioactivity was determined using a TopCount NXT (Packard Instrument) after adding 35 μl of Microscint 20 (Packard Instrument).
C. FAK Tyrosine Kinase Assay The Focal Adhesion kinase was assayed using the synthetic polymer poly(Glu/Tyr) (Sigma Chemicals) as a phosphoacceptor substrate. Each reaction mixture consisted of a total volume of 50 ul and contained 100 ng of baculovirus- expressed enzyme, 2μg of poly(Glu/Tyr), lμM of ATP, and 0.2 μCi of [γ-33P]ATP. The mixtures also contained 40 mM Tris.HCl, pH 7.4, 1 mM MnCl2, 0.5 mM DTT, and 0.1 mg/ml bovine serum albumin. The reaction mixtures were incubated at 26°C for 1 hour and kinase activity was determined by quantitation of the amount of radioactive phosphate transferred to the poly(Glu/Tyr) substrate. Incoφoration was measured by the addition of cold trichloroacetic acid (TCA) precipitation of the proteins which were collected onto GF/C unifilter plates (Packard Instrument Co., Meriden, CT) using a Filtermate universal harvester and the filters were quantitated using a TopCount 96-well liquid scintillation counter (Packard Instrument Co., Meriden, CT). Compounds were dissolved in dimethyl sulfoxide to a concentration of 10 mM and were evaluated at six concentrations, each in triplicate. The final concentration of DMSO added to the kinase assays was 0.5%, which has been shown to have no effect on kinase activity. IC50 values were derived non-linear regression analysis and have a coefficient of variance (SD/mean, n=6) = 16%.
D. HER-l/HER-2 Kinase Assay
Kinase reactions consisted of 10 ng of baculovirus expressed GST- HER1, 100 ng of HER2, 100 ng/ml poly(Glu/Tyr) (Sigma), 0.2 μCi 33P γ-ATP, 1 μM ATP in 50 μl kinase buffer (50 mM Tris, pH 7.5, 10 mM MnC12, 0.5 mM DTT). Reactions were incubated for lh at 27 C and stopped by the addition of cold trichloroacetic acid (TCA) to a final concentration 15%. TCA precipitates were collected onto GF/C unifilter plates (Packard Instrument Co., Meriden, CT) using a Filtermate universal harvester (Packard Instrument Co., Meriden, CT) and the filters were quantitated using a TopCount 96-well liquid scintillation counter (Packard Instrument Co., Meriden, CT). Dose response curves were generated to determine the concentration required to inhibit 50% of kinase activity (IC50). Compounds were dissolved at 10 mM in dimethylsulfoxide (DMSO) and evaluated at six concentrations, each in triplicate. The final concentration of DMSO in the assay equaled 1%. IC50 values were derived by non-linear regression analysis and have a coefficient of variance (SD/mean, n=6) = 16%.
E. IGF- Receptor Tyrosine Kinase Assay
The IGF-1 receptor tyrosine kinase was assayed using the synthetic polymer poly(Glu/Tyr) (Sigma Chemicals) as a phosphoacceptor substrate. Each reaction mixture consisted of a total volume of 50 ul and contained 125 ng of baculovirus expressed enzyme, 2.5μg of poly(Glu/Tyr), 25μM of ATP, and 0.1 μCi of [γ- 33P]ATP. The mixtures also contained 20 mM MOPS, pH 7.0, 5 mM MnCl2, 0.5 mM DDT, and 0.1 mg/ml bovine serum albumin. The reaction mixtures were incubated at 30°C for 45 minutes and kinase activity was determined by quantitation of the amount of radioactive phosphate transferred to the poly(Glu/Tyr) substrate. Incoφoration was measured by the addition of cold trichloroacetic acid (TCA) precipitation of the proteins which were collected onto GF/C unifilter plates (Packard Instrument Co., Meriden, CT) using a Filtermate universal harvester and the filters were quantitated using a TopCount 96-well liquid scintillation counter (Packard Instrument Co.,
Meriden, CT). Compounds were dissolved in dimethyl sulfoxide to a concentration of 10 mM and were evaluated at six concentrations, each in triplicate. The final concentration of DMSO added to the kinase assays was 0.5%, which has been shown to have no effect on kinase activity. IC50 values were derived by non-linear regression analysis and have a coefficient of variance (SD/mean, n=6) = 16%.
F. Insulin Receptor Tyrosine Kinase Assay
The Insulin Receptor Tryrosine kinase was assayed using the synthetic polymer poly(GlutTyr) (Sigma Chemicals) as a phosphoacceptor substrate. Each reaction mixture consisted of a total volume of 50 ul and contained 90 ng of baculovirus-expressed enzyme, 2.5μg of poly(Glu/Tyr), 25μM of ATP, and 0.1 μCi of [γ-33P]ATP. The mixtures contained also 20 mM Tris.HCl, pH 7.4, 5 mM MnCl2, 0.5 mM DTT, and 0.1 mg/ml bovine serum. The reaction mixtures were incubated at 26°C for 1 hour and kinase activity was determined by quantitation of the amount of radioactive phosphate transferred to the poly(Glu/Tyr) substrate. Incoφoration was measured by the addition of cold trichloroacetic acid (TCA) precipitation of the proteins which were collected onto GF/C unifilter plates (Packard Instrument Co., Meriden, CT) using a Filtermate universal harvester and the filters were quantitated using a TopCount 96-well liquid scintillation counter (Packard Instrument Co.,
Meriden, CT). Compounds were dissolved in dimethyl sulfoxide to a concentration of 10 mM and were evaluated at six concentrations, each in triplicate. The final concentration of DMSO added to the kinase assays was 0.5%, which has been shown to have no effect on kinase activity. IC50 values were derived non-linear regression analysis and have a coefficient of variance (SD/mean, n=6) = 16%. G. LCK Kinase Assay
Kinase reactions consisted of 10 ng of baculovirus expressed lOng GST-Lck, 100 ng/ml poly(Glu/Tyr) (Sigma), 0.2 μCi 33P γ-ATP, 1 μM ATP in 50 μl kinase buffer (50 mM Tris, pH 7.5, 10 mM MnC12, 0.5 mM DTT). Reactions were incubated for lh at 27 C and stopped by the addition of cold trichloroacetic acid (TCA) to a final concentration 15%. TCA precipitates were collected onto GF/C unifilter plates (Packard Instrument Co., Meriden, CT) using a Filtermate universal harvester (Packard Instrument Co., Meriden, CT) and the filters were quantitated using a TopCount 96-well liquid scintillation counter (Packard Instrument Co.,
Meriden, CT). Dose response curves were generated to determine the concentration required to inhibit 50% of kinase activity (IC50). Compounds were dissolved at 10 mM in dimethylsulfoxide (DMSO) and evaluated at six concentrations, each in triplicate. The final concentration of DMSO in the assay equaled 1%. IC50 values were derived by non-linear regression analysis and have a coefficient of variance (SD/mean, n=6) = 16%.
H. MET Kinase Assay Kinase reactions consisted of lOng of baculovirus expressed GST-Met, 2.5ug poly(Glu/Tyr) (Sigma), 0.2μCi 33P γ-ATP, 10 μM ATP in 50 μl kinase buffer (40mM Tris, pH 7.5, ImM MnC12, 0.50 mM DTT). Reactions were incubated for lh at 27 C and stopped by the addition of cold trichloroacetic acid (TCA) to a final concentration 3.5%. TCA precipitates were collected onto GF/C unifilter plates (Packard Instrument Co., Meriden, CT) using a Filtermate universal harvester (Packard Instrument Co., Meriden, CT) and the filters were quantitated using a TopCount 96-well liquid scintillation counter (Packard Instrument Co., Meriden, CT). Dose response curves were generated to determine the concentration required to inhibit 50% of kinase activity (IC50). Compounds were dissolved at 10 mM in dimethylsulfoxide (DMSO) and evaluated at seven concentrations, each in triplicate. The final concentration of DMSO in the assay equaled 1%. IC50 values were derived by non-linear regression analysis and have a coefficient of variance (SD/mean, n=6) = 16%. I. PDGF Receptor Kinase Assay
Kinase reactions consisted of 70ng of baculovirus expressed GST-PDGFbR, 0.3ug biotinylated poly(Glu/Tyr) (Sigma), in 50 μl kinase buffer (20 mM Hepes, pH 7.5, 0.7uM ATP, lOmM MnC12, 0.5mM DTT, 0.15mM NaCl, O.lmg/ml BSA). Reactions were incubated for 30 minutes at room temperature with shaking and stopped by the addition of lOul of 0.2M EDTA, pH8.0. 150ul of HTRF detection buffer was added and incubated for 1 hour and 30 minutes at room temperature. Counts were quantitated on Discovery HTRF Packard Instrument.
J. VEGFR-2 (KDR) Kinase Assay
Kinase reactions consisted of 7.5ng of baculovirus expressed GST-KDR, 1.5ug ρoly(Glu/Tyr) (Sigma), 0.04μCi 33P γ-ATP, 2.5 μM ATP in 50 μl kinase buffer (25 mM Tris, pH 7.5, 1.8 mM MnC12, 0.0.625 mM DTT). Reactions were incubated for lh at 27 C and stopped by the addition of cold trichloroacetic acid (TCA) to a final concentration 15%. TCA precipitates were collected onto GF/C unifilter plates (Packard Instrument Co., Meriden, CT) using a Filtermate universal harvester (Packard Instrument Co., Meriden, CT) and the filters were quantitated using a TopCount 96-well liquid scintillation counter (Packard Instrument Co.,
Meriden, CT). Dose response curves were generated to determine the concentration required to inhibit 50% of kinase activity (IC50). Compounds were dissolved at 10 mM in dimethylsulfoxide (DMSO) and evaluated at six concentrations, each in triplicate. The final concentration of DMSO in the assay equaled 1%. IC50 values were derived by non-linear regression analysis and have a coefficient of variance (SD/mean, n=6) = 16%.
K. Cytotoxicity assay (HT-29-colon; Colo205, MCF-7-breast) Tumor cell lines are maintained in McCoy's 5A medium (GIBCO) and 10% heat inactivated fetal bovine serum (GIBCO). The in vitro cytotoxicity is assessed in tumor cells by a tetrazolium-based colorimetric assay which takes advantage of the metabolic conversion of MTS (3-(4,5-dimefhylthiazol-2-yl)-5-(3- carboxymethoxyphenyl)-2-(4-sulphenyl)-2H-tetrazolium, inner salt) (Promega) to a reduced form that absorbs light at 492 nm (1). Cells are seeded 24 hr prior to drug addition. Following a 72 hour incubation at 37°C with serially diluted test compound, MTS (Riss, T.L, et al., Comparison of MTT, XTT, and a novel tetrazolium compound MTS for in vitro proliferation and chemosensitivity assays.," Mol. Biol. Cell 3 (Suppl.):184a, 1992), in combination with the electron coupling agent phenazine methosulfate, is added to the cells. The incubation is continued for 3 hours, then the absorbency of the medium at 492 nm is measured with a spectrophotometer to obtain the number of surviving cells relative to control populations. The results are expressed as median cytotoxic concentrations (IC50 values).
Table 1 - Biological Activity (uM); all compounds in the table had kinase activity of
<25uM against one or more of the following kinases CDK, EMT, FAK, Herl, Her2, IGF, IR, LCK, MET, PDGF, VEGF. HT-29 and Colo205 are human colon tumor cell lines, and MCF-7 is a human breast tumor cell line.
Figure imgf000033_0001
Figure imgf000034_0003
General Procedure for the Preparation of 2-Hydroxy-2-(substituted-phenyl)- ethylamines:
Figure imgf000034_0001
l-(3-Chloro-phenyI)-2-nitro-ethanol: To a solution of 3-chloro-benzaldehyde (20 g, 0.142 mol) in nitromethane (100 mL) were added magnesium sulfate (37.6 g, 0.312 mol) and phosphazene base P1-t-bu-tris(tetramethylene) (4.43 g, 0.014 mol). The reaction mixture was stirred at room temperature for 2 h. After concentration in vacuo, the residue was purified by flash chromatography (25% EtOAc / hexane) to yield the title compound (26.4 g, 100%) as a green-yellow oil. 1H NMR (300 MHz, DMSO-d6) δ 7.53 (IH, s), 7.35 - 7.42 (3H, m), 6.23 (IH, broad s), 5.32 - 5.33 (IH, m), 4.90 (lH, dd, J = 3.2, 12.4 Hz), 4.60 (IH, dd, J = 1.2, 12.4 Hz).
Figure imgf000034_0002
[l-(3-Chloro-phenyl)-2-nitro-ethoxy]-triethyl-siIane: To a solution of l-(3-chloro- phenyl)-2-nitro-ethanol (26 g, 0.14 mol) in DMF (50 mL) were added imidazole (28.6 g, 0.42 mol) and chlorotriethylsilane (25.3 g, 0.17 mol). The reaction mixture was stirred at room temperature for 2 h. After quenching with water, the mixture was extracted with ethyl acetate. The combined organic layers were washed with water and brine, dried over Na2SO4, and filtered. After removal of solvent, the crude product was purified by flash chromatography (2% EtOAc / hexane) to yield the title compound (37 g, 91%) as a colorless oil. 1H NMR (300 MHz, CDC13) δ 7.40 (IH, s), 7.27 - 7.32 (3H, m), 5.40 (IH, dd, J = 3.2, 9.5 Hz), 4.51 (IH, dd, 7 = 9.5, 12.1 Hz), 4.36 (IH, dd, 7 = 3.3, 12.1 Hz), 0.85 (9H, t, J = 7.5 Hz), 0.54 (6H, q, J = 7.5 Hz).
Figure imgf000035_0001
2-(3-Chloro-phenyI)-2-triethylsilanyloxy-ethylamine: Raney nickel (1 g) was washed with distilled water five times and methanol three times. [l-(3-Chloro- phenyl)-2-nitro-ethoxy]-triethyl-silane (10 g, 0.032 mol) and Raney nickel in methanol (100 mL) was hydrogenated (35 psi) at room temperature for 14 h. The reaction mixture was filtered through a pad of celite and rinsed with methanol. Concentration of the filtrate to dryness gave the title compound (5.6 g, 62%) as a colorless oil which was used for the next step without purification. 1H NMR (300 MHz, CDC13) δ 7.32 (IH, s), 7.18-7.26 (3H, m), 4.70 (IH, t, J = 5.8 Hz), 2.86 (2H, m), 0.89 (9H, t, / = 7.9 Hz), 0.56 (6H, q, / = 7.8 Hz). LRMS (M+H)+ m z 286. General Procedure for the Preparation of 2-Hydroxy-2-(substituted-phenyl)- ethylamines;
Figure imgf000035_0002
4-methoxy-3-bromophenyl chloroacetophenone: To a suspension of A1C13 (13.4g, 0.10 mol) in methylene chloride (40mL) was added a solution of 2-bromoanisole (12.5 mL, 0.10 mol) and chloroacetyl chloride (8 mL, 0.10 mol) at 0°C. The solution was warmed to ambient temperature for two hours and poured onto ice and extracted with methylene chloride, washed with saturated sodium bicarbonate solution, brine, and dried over MgSO4. The solution was filtered, concentrated and crystalized from EtOH to give 15.37g of white solid. LRMS [M-H]- 260.8; IR (KBr) 1697, 1048, 1255 cm"1; 1H NMR (300MHz, CDC13) δ 8.18 (s, IH), 7.94 (dd, J= 8.67 Hz, IH), 6.96 (d, J=8.67 Hz, IH), 4.62 (s, 2H), 3.98 (s, 3H); 13C NMR (CDC13, 75.5 Hz) δ 188.8, 160.3, 134.1, 129.9, 128.2, 112.4, 111.3, 56.6, 45.3. General Procedure for chiral reduction of chloroketones and ammonolysis :
Figure imgf000036_0001
(S)-l-[4-methoxy-3-bromophenyl] -2-chloro ethanol : To a solution of (S)-Methyl- CBS-oxazaborolidine (IM in toluene, 0.745 mL, 0.745 mmol) and BH3-THF (8mL, 8 mmol) was added at the same time a solution of BH3-THF (19mL, 19 mmol) and a solution of the chloroketone (10.03g, 37.98 mmol) in 19 mL of THF. Both solutions were added dropwise over 30 minutes. The solution was stirred for 1 hour and quenched with the slow addition of methanol (50mL). The solution was concentrated and the residue chromatographed over a short silica gel column (1:1 hexane/ethyl acetate) to give a quantitative yield (lO.Og) of chlorohydrin as a clear oil. IR (KBr)
1053, 1258, 3406 cm'1; 'H NMR (300MHz, CDC13) δ 7.59 (s, IH), 7.30 (dd, J=2.16 Hz, IH), 6.90 (d, J=8.46 Hz, IH), 4.83 (dd, J= 3.57 Hz, IH), 3.90 (s, 3H), 3.64 (ddd, J= 3.6, 11.1, 8.7, 2H), 2.04 (b s, IH). "c NMR (CDC , 75.5 MHz) δ 155.9, 133.5,
131.1, 126.3, 111.9, 73.1, 60.4, 56.3, 50.7.
OH
Br ,NH2-HCI
^Y
MeO' ^
(S) 2-Amino-l-[3-chloro-4-methoxyphenyl]ethanol Hydrochloride : To a solution of the chlorohydrin (lO.Og, 37.9 mmol) in 120 mL of methanol at -40°C was added 100 grams of ammonia. The solution was sealed in a pressure bottle and warmed to ambient temperature and stirred for 48 hours. The solution was cooled and opened. The ammonia was allowed to evaporate and solution concentrated. The residue was crystalized from ethanol/ethyl acetate to give 3.83g of white solid (35%). The material was reacted with B0C2O in ethyl acetate and saturated sodium bicarbonate and analyzed by chiral HPLC using a chiralcel OJ column using 95% hexane/ethanol as elutant and determined to by 98%ee. Additional crops were collected - 2.96g and 1.41g for a total of 75% yield. LRMS [M+H]+ 246; IR (cm"1, KBr) 1055, 1261,
3001, 2948, 3356; *H NMR (500MHz, DMSO) δ 8.09 (b s, 2 H), 7.58 (s, IH), 7.36 (dd, J=2.05, 6.45 Hz, IH), 7.11 (d, J=8.5 Hz, IH) 6.10 (s, IH), 4.80 (m, IH), 3.84 (s, 3H), 3.00 (ddd, J= 2.7, 12.6, 9.5 Hz, 2H); '3C NMR (DMSO, 75.5 MHz) δ 154.8, 135.4, 130.4, 126.6, 112.4, 110.4, 67.9, 56.2, 45.4.
OH
CI . ,NH2-HCI
(S) 2-Amino-l-[3-chlorophenyl]ethanol Hydrochloride : was prepared according to the general procedure outlined above. LRMS [M+H]+ 172; IR (KBr, cm-1) 3048,
3351, 2952; Η NMR (300MHz, MeOD) δ 7.48 (s, IH), 7.35 (m, 3H), 3.31 (ddd, J= 1.5, 3.12, 9.15 Hz 2H).
OH
Br„ ,NH2-HCI
(S)-2-Amino-l-[3-bromophenyl]ethanol Hydrochloride : was prepared according to the general procedure outlined above. LRMS [MH]+ 217.9; IR (KBr, cm-1) 3025, 3443, 2891; H NMR (500MHz, DMSO) δ 7.93 (b s, 2H), 7.60 (s, IH), 7.52 (d, IH),
7.41 (s, IH), 7.35 (d, J=7.7 Hz, IH) 6.17 (s, IH), 4.82 (m, IH), 3.08 (ddd, J= 2.6,
13
1 122..77,, 99..66 HHz, 2H); C NMR (DMSO, 75.5 MHz) δ 144.4, 130.5, 128.7, 125.0, 121.6,
68.3, 45.1.
OH
MeS"^^
(S)-2-Amino-l-[3-chloro-4-methyIthiophenyl]ethanol Hydrochloride : was prepared according to the general procedure outlined above. LRMS [M+H]+ 217.9;
IR (KBr, cm-1) 3007, 3358; H NMR (500MHz, DMSO) δ 8.12 (b s, 2H), 7.46 (s, IH), 7.37 (s, IH), 7.35 (d, IH) 6.19 (d, IH), 4.83 (m, IH), 3.01 (ddd, J= 3.2, 12.8, 9.3 Hz, 2H); '3C NMR (DMSO, 75.5 MHz) δ 139.6, 136.5, 129.8, 126.6, 125.4, 68.0, 45.2, 14.2.
Figure imgf000038_0001
(S)-2-Amino-l-[3-chloro-4-fluoro-phenyl]ethanol Hydrochloride : was prepared according to the general procedure outlined above. LRMS [M+H]+ 189.9; IR (KBr, cm-1) 1509, 3008, 3359; Η NMR (500MHz, DMSO) δ 8.21 (b s, 2H), 7.61 (d,
J=7.85 Hz, IH), 7.42 (m, 2H), 6.29 (s, IH), 4.88 (m, IH), 3.03 (ddd, J= 3.4, 12.8, 9.2 Hz, 2H); 13C NMR (DMSO, 75.5 MHz) δ 157.5, 155.5, 139.7, 128.1, 126.7, 119.3, 116.7, 109.0, 67.8, 45.2.
OH C'^- "^NH2-HCI xJ
(S)-2-Amino-l-[3-chloro-4-methoxyphenyl]ethanol Hydrochloride : was prepared according to the general procedure outlined above. LRMS [M+H]+ 202; IR (KBr, cm-1) 3354, 3003, 2949, 1288, 1064; *H NMR (500MHz, DMSO) δ 8.18 (brs, 3H), 7.43 (d, J=2.0Hz, IH), 7.31 (dd, J=8.5, 2.0Hz, IH), 7.14 (d, J=5.1Hz, IH), 6.11 (s, IH), 4.81 (m, IH), 3.84 (s, 3H), 2.99 (dd, J=13, 3.5Hz, IH), 2.83 (dd, J=12.5, 9Hz, IH); 13C NMR (DMSO, 125MHz) δ 153.9, 135.0, 127.3, 125.8, 120.8, 112.6, 68.0, 56.1, 45.5; Elemental Analysis Calcd for C9H,2ClNO2-HCl: C, 45.39; H, 5.50; N, 5.88. Found: C, 45.38; H, 5.43; N, 5.70.
Figure imgf000038_0002
(S)-2-Amino-l-(7-bromo-2,3-dihydrobenzfuran-5-yl)-2-aminoethanol Hydrochloride : was prepared according to the general procedure outlined above. LRMS [M+H]+ 258; IR (KBr, cm-1) 3349, 3006, 2928, 1485, 1045, 983; Η NMR (500MHz, DMSO) δ 8.13 (brs, 3H), 7.29 (s, IH), 7.23 (s, IH), 6.08 (d, J=4 Hz, IH), 4.76 (m, IH), 4.61 (t, J=9Hz, 2H), 3.29 (t, J=9Hz, 2H), 2.96 (dd, J=13, 3.5Hz, IH), 2.82 (dd, J=13,9.5Hz, IH); '3C NMR (DMSO, 125MHz) δ 156.3, 135.9, 129.1, 128.1, 122.1, 100.9, 71.5, 68.2, 45.6, 29.9; Elemental Analysis Calcd for Cι02BrNO2-HCl: C, 40.77; H, 4.44; N, 4.75. Found: C, 40.77; H, 4.63; N, 4.63.
General Procedure for the Preparation of 2-Amino-3-(substituted-phenyl)- propanol:
Figure imgf000039_0001
(S)-[2-(3-Bromo-phenyl)-l-hydroxymethyl-ethyl]-carbamic acid tert-butyl ester:
To a solution of (S)-3-(3-bromo-phenyl)-2-tert-butoxycarbonylamino-propinic acid (500 mg, 1.45 mmol) in THF (30 mL) was added borane-tetrahydrofuran complex (1.0 M solution) (4.35 mL, 4.35 mmol). The reaction mixture was stirred at room temperature for 14 h and quenched with acetic acid (1 mL). After removal of most solvent, the residue was extracted with EtOAc, washed with brine, dried over Na2SO4. After concentration, the crude product (400 mg, 83%) was used for the next step without purification. LCMS (M+H)+ m z 330 (t = 1.61 min
Figure imgf000039_0002
(S)-2-Amino-3-(3-bromo-phenyl)-propan-l-ol: To a solution of (S)-[2-(3-bromo- phenyl)-l-hydroxymethyl-ethyl]-carbamic acid tert-butyl ester (400 mg, 1.21 mmol) in methanol (30 mL) was added 4 M HCl in dioxane (2 mL, excess). The reaction mixture was stirred at room temperature for 14 h. After concentration in vacuo, the residue was used for the next step without purification. LCMS (M+H)+ m z 230 (t = 0.78 min.)
Figure imgf000039_0003
5-Chloroacetyl-7-chlorooxindole: To a suspension of AICI3 (13.4g, 0.10 mol) in methylene chloride (40mL) is added a solution of 7-Chlorooxindole (0.10 mol) and chloroacetyl chloride (8 mL, 0.10 mol) at 0°C. The solution is warmed to ambient temperature for two hours and poured onto ice and extracted with methylene chloride, washing with saturated bicarbonate solution, brine, and drying over MgSO4 would provide the desired chloroketone.
Figure imgf000040_0001
(S)-7-Chloro-5-(2-chloro-l-hydroxy-ethyl)-2-oxoindole : To a solution of (S)-
Methyl-CBS-oxazaborolidine (IM in toluene, 0.745 mL, 0.745 mmol) and BH3-THF (8mL, 8 mmol) is added at the same time a solution of BH3-THF (19mL, 19 mmol) and a solution of the 5-Chloroacetyl-7-chlorooxindole (37.98 mmol) in 19 mL of THF. Both solutions are added dropwise over 30 minutes. The solution is stirred for 1 hour and quenched with the slow addition of methanol (50mL). The solution is concentrated and the residue chromatographed over a short silica gel column (1:1 hexane/ethyl acetate).
Figure imgf000040_0002
(S)-2-Amino-l-(7-chlorooxindole-5-yl)-ethanol Hydrochloride : To a solution of the chlorohydrin (37.9 mmol) in 120 mL of methanol at -40°C is added 100 grams of ammonia. The solution is sealed in a pressure bottle and warmed to ambient temperature and stirred for 48 hours. The solution is cooled and opened. The ammonia is allowed to evaporate and solution concentrated to provide the hydrochloride salt, which can be crystallized from ethanol/ethyl acetate.
Figure imgf000041_0001
6-ChloroacetyI-4-chloro-2-benzooxazolinone: To a suspension of AICI3 (13.4g, 0.10 mol) in methylene chloride (40mL) is added a solution of 4-chloro-2- benzooxazolinone (0.10 mol) and chloroacetyl chloride (8 mL, 0.10 mol) at 0°C. The solution is warmed to ambient temperature for two hours and poured onto ice and extracted with methylene chloride, washing with saturated bicarbonate solution, brine, and drying over MgSO4 would provide the desired chloroketone.
OH
Figure imgf000041_0002
(S)-6-(2-Chloro-l-hydroxy-ethyl)-4-chloro-2-benzooxazolinone: To a solution of (S)-Methyl-CBS-oxazaborolidine (IM in toluene, 0.745 mL, 0.745 mmol) and BH3- THF (8mL, 8 mmol) is added at the same time a solution of BH3-THF (19mL, 19 mmol) and a solution of the 6-Chloroacetyl-4-chloro-2-benzooxazolinone (37.98 mmol) in 19 mL of THF. Both solutions are added dropwise over 30 minutes. The solution is stirred for 1 hour and quenched with the slow addition of methanol (50mL). The solution is concentrated and the residue chromatographed over a short silica gel column (1:1 hexane/ethyl acetate).
Figure imgf000041_0003
(S)-2-Amino-l-(4-chIoro-2-benzooxazoIinone-6-yl)-ethanol Hydrochloride : To a solution of the chlorohydrin (37.9 mmol) in 120 mL of methanol at -40°C is added 100 grams of ammonia. The solution is sealed in a pressure bottle and warmed to ambient temperature and stirred for 48 hours. The solution is cooled and opened. The ammonia is allowed to evaporate and solution concentrated to provide the hydrochloride salt, which can be crystallized from ethanol/ethyl acetate.
Figure imgf000042_0001
N-MethyI-7-chloroindoline: To a solution of the 7-Chloroindoline (0.10 mol) in 500 mL of acetone is added K2CO3 (0.15mol) and Mel (0.15mol) and refluxed until the starting material is consumed. The reaction is filtered and washed with water and saturated bicarbonate solution, drying over MgSO4 would provide the N-Me -7- chloroindoline
Figure imgf000042_0002
N-Methyl-5-chloroacetyl-7-chloro-indoline: To a suspension of AICI3 (13.4g, 0.10 mol) in methylene chloride (40mL) is added a solution of N-Me-7-Chloroindoline (0.10 mol) and chloroacetyl chloride (8 mL, 0.10 mol) at 0°C. The solution is warmed to ambient temperature for two hours and poured onto ice and extracted with methylene chloride, washing with saturated bicarbonate solution, brine, and drying over MgSO4 provides the desired chloroketone.
Figure imgf000042_0003
(S)-N-Methyl-5-(2-chloro-l-hydroxylcthyl)-7-chloro-indoline: To a solution of (S)- Methyl-CBS-oxazaborolidine (IM in toluene, 0.745 mL, 0.745 mmol) and BH3-THF (8mL, 8 mmol) is added at the same time a solution of BH3-THF (19mL, 19 mmol) and a solution of N-Methyl-5-chloroacetyl-7-chloro-indoline (37.98 mmol) in 19 mL of THF. Both solutions are added dropwise over 30 minutes. The solution is stirred for 1 hour and quenched with the slow addition of methanol (50mL). The solution is concentrated and the residue chromatographed over a short silica gel column (1:1 hexane/ethyl acetate).
Figure imgf000043_0001
(S)-2-Amino-l-(7-chloro-N-methyl-indoline-5-yl)-ethanol Hydrochloride : To a solution of the chlorohydrin (37.9 mmol) in 120 mL of methanol at -40°C is added 100 grams of ammonia. The solution is sealed in a pressure bottle and warmed to ambient temperature and stirred for 48 hours. The solution is cooled and opened. The ammonia is allowed to evaporate and solution concentrated to provide the hydrochloride salt, which can be crystallized from ethanol/ethyl acetate.
Figure imgf000043_0002
(S)-2-Chloro-l-(7-chloro-N-methyI-indol-5-yl)-ethanol: A solution (S)-N-Methyl- 5-(2-chloro-l-hydroxylethyl)-7-chloro-indoline (0.10 mmol) in lOOmL of t-butyl methyl ether is treated with o-chloroanil (0. lOmmol) at ambient temperature. The solution is concentrated and the residue chromatographed over silica gel (1: 1 hexane/ethyl acetate) to provide the corresponding indole.
Figure imgf000043_0003
(S)-2-Amino-l-(7-chloro-N-methyl-indoI-5-yl)-ethanol Hydrochloride : To a solution of the chlorohydrin (37.9 mmol) in 120 mL of methanol at -40°C is added 100 grams of ammonia. The solution is sealed in a pressure bottle and warmed to ambient temperature and stirred for 48 hours. The solution is cooled and opened. The ammonia is allowed to evaporate and solution concentrated to provide the hydrochloride salt, which can be crystalized from ethanol/ethyl acetate.
Figure imgf000044_0001
4-Chloro-2-methyl-benzooxazole : To a solution of the 4-chloro-2- benzooxazolinone (0.10 mol) in 200 mL ethanol is added LiOH (0.20 mol) in 100 mL of water. The solution is refluxed for 8 hr and cooled. The solution is neutralized with IN HCl and extracted with ethyl acetate followed by drying over MgSO4. The solution is concentrated and taken up in 200 mL of toluene and 0.10 mol of acetic acid. The solution is refluxed in a Dean Stark trap for 12 hours, concentrated and purified by flash chromatography.
Figure imgf000044_0002
6-Chloroacetyl-4-chloro-2-methyl-benzooxazole: To a suspension of AICI3 (13.4g, 0.10 mol) in methylene chloride (40mL) is added a solution of 2-Methyl-4-chIoro- benzooxazole (0.10 mol) and chloroacetyl chloride (8 mL, 0.10 mol) at 0°C. The solution is warmed to ambient temperature for two hours and poured onto ice and extracted with methylene chloride, washing with saturated bicarbonate solution, brine, and drying over MgSO would provide the desired chloroketone.
Figure imgf000044_0003
(S)-6-(2-chloro-l-hydroxy-ethyI)-4-chloro-2-methyl-benzooxazole: To a solution of (S)-Methyl-CBS-oxazaborolidine (IM in toluene, 0.745 mL, 0.745 mmol) and BH3-THF (8mL, 8 mmol) is added at the same time a solution of BH3-THF (19mL, 19 mmol) and a solution of 6-ChloroacetyI-4-chloro-2-methyl-benzooxazole (37.98 mmol) in 19 mL of THF. Both solutions are added dropwise over 30 minutes. The solution is stirred for 1 hour and quenched with the slow addition of methanol (50mL). The solution is concentrated and the residue chromatographed over a short silica gel column (1: 1 hexane/ethyl acetate).
Figure imgf000045_0001
(S)-2-Amino-l-(4-chloro-2-methyl-benzooxazol-6-yl)-ethanol Hydrochloride : To a solution of the chlorohydrin (37.9 mmol) in 120 mL of methanol at -40°C is added 100 grams of ammonia. The solution is sealed in a pressure bottle and warmed to ambient temperature and stirred for 48 hours. The solution is cooled and opened. The ammonia is allowed to evaporate and solution concentrated to provide the hydrochloride salt, which can be crystallized from ethanol/ethyl acetate.
Preparation of 3-(6-Imidazol-l-yl-4-methyl-lH-benzimidazol-2-yl)-4-iodo-lH- pyridin-2-one;
Figure imgf000045_0002
l-(3-Methyl-4-nitro-phenyl)-lH-imidazole: To a solution of 4-fluoro-2-methyl-l- nitro-benzene (300 mg, 1.84 mmol) in DMSO (2 mL) were added KOH (20 mg, 3.87 mmol) and imidazole (263 mg, 3.88 mmol). The reaction mixture was heated to 100°C for 3.5 h, cooled to room temperature, and diluted with ice-cold water. The resulting precipitate was filtered, washed with ice-cold water, and dried under vacuum to give the title compound (310 mg, 80%) as a yellow powder. 1H NMR (300 MHz, DMSO-d6) δ 8.46 (IH, s), 8.16 (IH, d, J= 8.9 Hz), 7.90 - 7.92 (2H, m), 7.78 (IH, dd, 1 = 2.5, 8.9 Hz), 7.17 (IH, s), 2.61 (3H, s). LRMS (M+H)+ m/z 204.
Figure imgf000046_0001
4-Imidazol-l-yl-2-methyl-phenylamine: To 1 -(3 -methyl -4-nitro-phenyl)-lH- imidazole (200 mg, 0.98 mmol) and 10% Palladium on carbon (35 mg) was added degassed methanol (3 mL). The suspension was flushed and evacuated with hydrogen / vacuum line. The suspension was allowed to stir at room temperature for 14 h under hydrogen atmosphere (hydrogen balloon). The dark reaction mixture was filtered through a pad of celite and rinsed with methanol. Concentration of the filtrate gave the title compound (166 mg, 98%) which was used for the next step without purification. 1H NMR (300 MHz, DMSO-d6) δ 7.95 (IH, s), 7.48 (IH, s), 7.16 (IH, narrow d, J = 2.5 Hz), 7.09 (IH, dd, J = 2.5, 8.4 Hz), 7.01 (IH, s), 6.67 (IH, ά, J = 8.4 Hz), 5.03 (2H, broad s), 2,10 (3H, s).
Figure imgf000046_0002
N-(4-Imidazol-l-yl-2-methyl-6-nitro-phenyI)-acetamide: To a solution of 4- imidazol-l-yl-2-methyl-phenylamine (1 g, 5.78 mmol) in CH2CI2 (20 mL) was added AC2O (0.7 mL, 7.28 mmol) at 0°C. The reaction mixture was stir at room temperature for 14 h and diluted with water. The aqueous layer was extracted with CH2CI2 and the combined organic layers were washed with saturated ΝaHCθ3 and brine, dried over Na2SO4, and concentrated in vacuo to give a white solid. The white solid was suspended in H2SO4 (cone.) (15 mL). Then HNO3 (cone.) (0.312 mL) was added to the suspension at 0°C. The reaction mixture was slowly warmed to room temperature and stirred at room temperature for 4 h. After cooling to -10°C, the reaction mixture was neutralized with ammonium hydroxide and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over Na2SO4, and concentrated. The residue was purified by flash chromatography (1:9:5 MeOH / THF / hexane) to yield the title compound (0.61 g, 41%). 1H NMR (300 MHz, CD3OD) δ 8.11 (IH, s), 7.45 - 7.56 (2H, m), 7.38 (IH, dd, J= 2.4, 8.4 Hz), 7.14 (IH, s), 2.33 (3H, s), 2.18 (3H, s).
Figure imgf000047_0001
4-Imidazol-l-yI-2-methyI-6-nitro-phenylamine: To a suspension of N-(4-imidazol- l-yl-2-methyl-6-nitro-phenyl)-acetamide (279 mg, 1.07 mmol) in ethanol (3 mL) was added 2 Ν HCl (2 mL). The reaction mixture was heated to reflux for 14 h, cooled to room temperature, and neutralized with saturated ΝaHCU3. The resulting bright orange solid was filtered and dried under vacuum. The title compound (179 mg, 76%) was obtained as an orange solid. Η NMR (300 MHz, CD3OD) δ 8.78 (IH, s), 8.24 (IH, s), 7.78 (IH, s), 7.64 (IH, s), 7.46 (IH, s), 2.36 (3H, s).
Figure imgf000047_0002
5-Imidazol-l-yl-3-methyl-benzene-l,2-diamine: To 4-imidazol- 1 -yl-2-methyl-6- nitro-phenylamine (350 mg, 1.61 mmol) and 10% Palladium on carbon (40 mg) were added degassed methanol (5 mL) and TFA (5 drops). The reaction mixture was flushed and evacuated with hydrogen / vacuum line, stirred at room temperature for 14 h under hydrogen atmosphere (hydrogen balloon). The dark reaction mixture was filtered through a pad of celite and rinsed with methanol. Concentration of the filtrate gave the residue, which was diluted with water, extracted with ethyl acetate. The combined organic layers were washed with saturated NaHCO3, brine, dried over Na2SO4. Concentration to dryness gave the title compound (275 mg, 91%) as a solid. 1H NMR (300 MHz, CD3OD) δ 7.87 (IH, s), 7.34 (IH, s), 7.05 (IH, s), 6.72 (IH, d, J = 2.4 Hz), 6.65 (IH, d, J = 2.4 Hz) 2.21 (3H, s). LCMS (M+H)+ m z 189 (t = 0.23 min.).
Figure imgf000048_0001
4-Iodo-2-methoxy-pyridine-3-carbaldehyde (WO 95/29917): A 5-liter three-necked round bottom flask was equipped with an overhead mechanical stirrer under nitrogen, the flask was charged with THF (1 L) and cooled to -78°C. To this stirred solution was added tert-butyllithium (1.7 M solution in pentane) (800 mL, 1.36 mol) via canula followed by 2-methoxypyridine (132.2 g, 1.21 mol) at -78°C. The mixture was stirred for 1 h at -78°C. To the mixture was added N-formyl-N, N', N'- trimethylethylenediamine (176 mL, 1.37 mol) dropwise at -78°C. The reaction mixture was stirred for ca. 30 min at -78°C before warming to -23°C over ca. 30 min. To the mixture at -23°C was added ethylene glycol dimethyl ether (1 L) followed by rt-butyllithium (2.5 M solution in hexane) (800 mL, 2.0 mol). The resulting mixture was stirred for ca. 2 h during which time the reaction mixture turned deep green. A 12-L 4-necked round flask was charged with iodine (571 g, 2.25 mol) and ethylene glycol dimethyl ether (2 L) and the resultant solution was cooled to -78°C. The contents of the 5-L flask were transferred via canula to the mixture of iodine and ethylene glycol dimethyl ether in the 12-L flask at -78°C. After the addition was complete, the reaction mixture was stirred for an additional 1 h at -78°C. The cooling bath was removed and the mixture was allowed to warm to about 0°C and treated with 2 L of water and 2 L of 1 Ν hydrochloric acid. Methyl t-butyl ether (2L) was added and the layers were separated. The aqueous layer was extracted with 2 x 1 L of methyl t-butyl ether. The combined organic layers were washed with saturated Νa2S2θ3 (1.2 L), brine (1.2 L), dried over Na SO4. After concentration in vacuo, the thick slurry was diluted with hexane (1 L). The mixture was cooled with an ice / water bath for ca. 30 min. The precipitate was filtered and dried in vacuum to yield the title compound as a light yellow solid. Η NMR (300 MHz, CDC13) δ 10.22 (s, IH), 7.86 (IH, d, J = 5.3 Hz), 7.54 (IH, d, 7= 5.3 Hz), 4.06 (3H, s). LCMS (M+H)+ m/z 364 (t = 2.26 min.).
Figure imgf000049_0001
6-Imidazol-l-yl-2-(4-iodo-2-methoxy-pyridin-3-yl)-4-methyl-lH-benzimidazole:
To a solution of 5-imidazol-l-yl-3-methyl-benzene-l,2-diamine (175 mg, 0.93 mmol) in methanol (8 mL) was added a solution of 4-iodo-2-mefhoxy-pyridine-3- carbaldehyde (245 mg, 0.93 mmol) in methanol (5 mL) at 0°C. The reaction mixture was stirred at 0°C for 1.5 h and then at room temperature for 2 h. After concentration, the residue was purified by flash column chromatography (10% MeOH / CH2C12) to give the title compound (291 mg, 60%). 1H NMR (300 MHz, CD3OD) δ 8.13 (IH, s), 7.98 (IH, d, 7 = 5.4 Hz), 7.62 (IH, d, 7 = 5.4 Hz), 7.59 (2H, s), 7.33 (IH, s), 7.16 (IH, s), 3.90 (3H, s), 2.67 (3H, s). LCMS (M+H)+ m/z 432 (t = 0.99 min.).
Figure imgf000049_0002
3-(6-Imidazol-l-yl-4-methyl-lH-benzimidazol-2-yI)-4-iodo-lH-pyridin-2-one:
The suspension of 6-imidazol-l-yl-2-(4-iodo-2-methoxy-pyridin-3-yl)-4-methyl-lH- benzimidazole in 1 N HCl (6 mL) was heated to 70°C for 3 days, cooled to room temperature, and diluted with ethyl acetate. After extraction, the combined organic layers were washed with brine, dried over Na2SO4 and concentrated. The residue was purified by flash chromatography (1% NH4OH / 10% MeOH / CH2C12) to yield the title compound (78 mg, 81%) as a solid. 1H NMR (300 MHz, CD3OD) δ 8.12 (IH, s), 7.58 (2H, s), 7.29 - 7.31 (2H, m), 7.16 (IH, s), 7.01 (IH, 7 = 6.8 Hz), 2.66 (3H, s). LCMS (M+H)+ m/z 418 (t = 0.75 min.)
Preparation of 2-(4-Chloro-2-oxo-l,2-dihvdro-pyridin-3-yl)-7-methyl-3H- benzimidazole-5 -carbonitrile :
Figure imgf000050_0001
4-Amino-3-methyl-benzonitrile: To a solution of 3-methyl-4-nitro-benzonitrile (20 g, 0.123 mol) in HOAc (200 mL) was added iron powder (17.55 g, 0.309 mol). After 10 min, the reaction was exothermic and turned to dark color. The reaction mixture was allowed to stir at room temperature for 14 h and then diluted with EtOAc (200 mL). The brown precipitate was filtered through a pad of celite and the filtercake was rinsed with EtOAc. The filtrate was concentrated in vacuo and the residue was purified by flash chromatography (40% EtOAc / hexane) to yield the title compound (15.3 g, 92%). 1H NMR (300 MHz, CDC13) δ 7.30 - 7.34 (2H, m), 6.64 (IH, d, 7 = 8.7 Hz), 2.16 (3H, s). LCMS (M+H)+ m/z 133 (t = 0.93 min).
NH 0'^CF3
N-(4-Cyano-2-methyl-6-nitro-phenyl)-2,2,2-trifluoro-acetamide: To the ice-cold trifluoroacetic anhydride (60 mL) was added 4-amino-3-methyl-benzonitrile (14.33 g, 0.108 mol) in portion. The resulting white slurry was stirred at 0°C for 30 min. Then ammonium nitrate (17.28 g, 0.216 mol) was added. The reaction mixture was allowed to stir at 0°C for 1 h and at room temperature for 14 h. After removal of most solvent, the reaction mixture was cooled with ice and quenched with ice. The yellow precipitate was filtered, washed with cold water, and dried under vacuum. The crude product (15.5 g, 52% yield, and ca. 80% pure) was used for the next step without purification. 1H ΝMR (300 MHz, CD3OD) δ 8.05 (IH, s), 7.74 (IH, s), 2.30 (3H, s). LRMS (neg. ESI, (M-H)") m z 272.
Figure imgf000050_0002
4-Amino-3-methyl-5-nitro-benzonitrile: A mixture of N-(4-cyano-2-methyl-6-nitro- phenyl)-2,2,2-trifluoro-acetamide (5 g, 18.3 mmol) and 2 M ammonia in methanol
(80 mL) was heated to reflux for 14 h and then cooled to room temperature. After concentration in vacuo, the residue was purified by flash chromatography (20% EtOAc / hexane) to yield the title compound (3.24 g, 100%, ca 80% pure). 1H ΝMR
(300 MHz, CDC13) δ 8.40 (IH, s), 7.47 (IH, s), 6.6 - 6.8 (2H, broad s), 2.89 (3H, s).
Figure imgf000051_0001
3,4-Diamino-5-methyl-benzonitrile: To a solution of 4-amino-3-methyl-5-nitro- benzonitrile (3.24 g, 18.3 mmol) in ethanol (80 mL) was added tin chloride dihydrate (8.67 g, 49.75 mmol). The reaction mixture was heated to reflux for 14 h, then cooled to room temperature, and concentrated in vacuum. The residue was diluted with ethyl acetate (100 mL) and treated with triethylamine (20 mL). The resulting slurry was filtered through a pad of celite and the filtercake was rinsed with three-portion ethyl acetate (50 mL). The filtrate was washed with saturated NaHCO3, water, and brine, then dried over Na2SO4 and filtered. After removal of solvent, the residue was purified by flash chromatography on silica gel (30% - 50% EtOAc / hexane) to yield the title compound (2.17 g, 81%) as a light yellow solid. 1H NMR (300 MHz, CDC13) δ 6.94 (IH, s), 6.85 (IH, s), 2.16 (3H, s). LCMS (M+H)+ m z 148 (t = 0.67 min.).
Figure imgf000051_0002
2-(4-Iodo-2-methoxy-pyridin-3-yl)-7-methyl-3H-benzimidazole-5-carbonitrile: To a solution of 3,4-diamino-5-methyl-benzonitrile (2.00 g, 13.6 mmol) in MeOH (40 mL) was added 4-iodo-2-methoxy-pyridine-3-carbaldehyde (3.6 g, 13.6 mmol) in MeOH (20 mL) at 0°C. The resulting slurry was stirred at 0°C for 1 h. Iodine (1.73 g, 8.8 mmol) in MeOH (10 mL) was added dropwise via a dropping funnel to the reaction mixture at 0°C. The reaction mixture was stirred at room temperature for 14 h. After removal of MeOH, the residue was diluted with saturated Na2S2θ3 and extracted with EtOAc. The combined organic layers were washed with water and brine, dried over Na2SO4. The crude product was purified by flash column chromatography (3% MeOH / CH2C12) to yield the title compound (1.81 g, 46%). Η NMR (300 MHz, CDC13) δ 7.90 (IH, s), 7.49 (IH, d, 7 = 5.4 Hz), 7.46 (IH, s), 7.41 (IH, d, 7 = 5.3 Hz), 3.78 (3H, s), 2.68 (3H, s). LCMS (M+H)+ m/z 391 (t = 1.27 min.).
Figure imgf000052_0001
2-(4-Chloro-2-oxo-l,2-dihydro-pyridin-3-yl)-7-methyl-3H-benzimidazole-5- carbonitrile: A suspension of 2-(4-iodo-2-methoxy-pyridin-3-yl)-7-methyl-3H- benzimidazole-5-carbonitrile (1.8 g, 4.63 mmol) in 4 M HCl dioxane (40 mL) was heated to 80°C for 6 h and cooled to room temperature. The precipitate was filtered and dried. The crude product (1.08 g, 82%) was used for the next step without purification. LRMS (neg. ESI, (M-H)") m z 283.
Preparation of (S)-4-(l-Benzyl-2-trityloxy-ethylamino)-3-(6-bromo-4-methyl-lH- benzimidazol-2-yl)-lH-pyridin-2-one:
Figure imgf000052_0002
5-Bromo-3-methyl-benzene-l,2-diamine: To a suspension of 4-bromo-2-methyl-6- nitro-phenyl amine (20 g, 0.086 mol) in ethanol (200 mL) was added tin chloride dihydrate (49.2 g, 0.258 mol). The reaction mixture was heated to reflux for 14 h, cooled to room temperature, and concentrated in vacuo. The residue was diluted with ethyl acetate (150 mL) and treated with triethylamine (40 mL). The resulting slurry was filtered through a pad of celite, and the filtercake was rinsed with three portions ethyl acetate (50 mL). The filtrate was washed with saturated NaHCO3, water, and brine, then dried over Na2SO4 and filtered. After removal of the solvent, the residue was purified by flash chromatography on silica gel (30% EtOAc / hexane, then 5% MeOH / CH2C12) to yield the title compound (10.26 g, 59%) as a yellow oil. 1H NMR (300 MHz, CDCI3) δ 6.77 (IH, d, 7 = 2.0 Hz), 6.74 (IH, d, 7 = 2.0 Hz), 2.16 (3H, s). LCMS (M+H)+ m/z 201. (t = 0.83 min.).
Figure imgf000053_0001
6-Bromo-2-(4-iodo-2-methoxy-pyridin-3-yl)-4-methyl-lH-benzimidazole: To a solution of 5-bromo-3-methyl-l,2-phenylenediamine (4 g, 19.9 mmol) in methanol (80 mL) was added 4-iodo-2-methoxy-pyridine-3-carbaldehyde (5.23 g, 19.9 mmol) in methanol (20 mL) dropwise at 0°C. The resulting slurry was stirred for 30 min at room temperature. Then iodine (2.53 g, 9.95 mmol) in methanol (20 mL) was added via a dropping funnel. After 14 h, the reaction mixture was concentrated in vacuo, diluted with 5% Na2S2O3, and extracted with ethyl acetate. The combined organic layers were washed with water, brine, and dried over Na SO4. After removal of solvent, the residue was purified by careful flash chromatography (20% EtOAc / hexane) to yield the title compound (4.05 g, 46%) as a yellow foam. ]H NMR (300 MHz, CDCI3) δ 7.86 (IH, d, 7 = 5.31 Hz), 7.53 (IH, d, 7 = 5.3 Hz), 7.26 (2H, broad s), 3.91 (3H, s), 2.63 (3H, s). LCMS (M+H)+ m/z 444 (t = 1.39 min.).
Figure imgf000053_0002
3-(6-Bromo-4-methyl-lH-benzimidazol-2-yl)-4-chloro-lH-pyridin-2-one: The suspension of 6-bromo-2-(4-iodo-2-methoxy-pyridin-3-yl)-4-methyl- 1H- benzimidazole (4 g, 9.03 mmol) and 60 mL of 4 M HCl in dioxane was heated to
80°C for 6 h and cooled to room temperature. The precipitate was filtered and dried to yield the title compound (3.0 g, 100%) as a brown powder. The crude product was used for the next step without purification. 1H NMR (300 MHz, CD3OD) δ 7.55 (IH, s), 7.42 (IH, d, 7 = 6.0 Hz), 7.17 (IH, s), 6.91 (IH, d, 7 = 6.0 Hz), 2.55 (3H, s). LC MS (M+H)+ m/z 338 (t = 1.33 min.).
Figure imgf000054_0001
(5)-4-(l-Benzyl-2-hydroxy-ethylamino)-3-(6-bromo-4-methyI-lH-benzimidazol- 2-yl)-lH-pyridin-2-one: To a solution of 3-(6-bromo-4-methyl-lH-benzimidazol-2- yl)-4-chloro-lH-ρyridin-2-one (1.42 g, 3.78 mmol) in DMF (15 mL) were added (S)- (-)-2-amino-3-phenyl-l-propanol (1.43 g, 9.45 mmol) and N-methyl moφholine (1.5 mL). The reaction mixture was heated to 80°C for 6 h and cooled to room temperature. The solvent was removed with high vacuum and the residue was purified by flash chromatography (5% MeOH / CH2CI2) to yield the title compound (1.26 g, 74%) as a yellow foam. Η ΝMR (300 MHz, CDCI3) δ 6.9 - 7.2 (8H, m), 5.86 (IH, d, 7 = 7.1 Hz), 3.7-3.9 (3H, m), 2.9-3.1(2H, m), 2.57 (3H, s). LCMS (M+H)+ m z 453 (t = 2.03 min.).
Figure imgf000054_0002
(S)-4-(l-Benzyl-2-trityloxy-ethylamino)-3-(6-bromo-4-methyl-lH-benzimidazol- 2-yl)-lH-pyridin-2-one: To a solution of (S)-4-(l-benzyl-2-hydroxy-ethylamino)-3- (6-bromo-4-methyl-lH-benzimidazol-2-yl)-lH-pyridin-2-one (0.9 g, 1.98 mmol) in THF (30 mL), was added Cs2CO3 (1.29 g, 3.96 mmol) followed by triphenylmethyl chloride (1.10 g, 3.96 mmol). The reaction mixture was heated to reflux for 14 h under nitrogen and then cooled to room temperature. After removal of the solvent, the residue was diluted with ethyl acetate and washed with water. The aqueous fraction was extracted with ethyl acetate and the combined organic layers were washed with water and brine, dried over Na2SO4. After concentration in vacuo, the residue was purified by flash column chromatography (30% EtOAc / hexane) to yield the title compound (1 g, 73%) as a white solid. Η NMR (300 MHz, DMSO-d6) 6 11.77 (IH, broad s), 11.73 (IH, d, 7 = 5.2 Hz), 11.46 (IH, broad s), 7.13 - 7.54 (23H, m), 5.87 (IH, d, 7 = 4.5 Hz), 4.09 - 4.14 (IH, m), 3.07 - 3.42 (4H, m), 2.54 (3H, s). LCMS (M+H)+ m/z 695 (t = 2.79 min.).
Preparation of 3-r6-(4-Acetyl-piperazin-l-yl)-4-methyl-lH-benzimidazol-2-yll-4- chloro-lH-pyridin-2-one:
Figure imgf000055_0001
l-[4-(4-Amino-3-methyl-5-nitro-phenyl)-piperazin-l-yl]-ethanone: A mixture of 4- bromo-2-methyl-6-nitro-phenylamine (5g, 21.64 mmol), 1-acetylpiperazine (4.2g, 32.46 mmol), palladium acetate (244 mg, 1.08 mmol), tri-tert-butylphosphine (440 mg, 2.16 mmol) and sodium tert-butoxide (4.2 g, 43.29 mmol) in toluene (70 mL) was heated to 100°C for 14 h under nitrogen. The reaction mixture was cooled to room temperature and diluted with EtOAc. After extraction, the combined organic layers were washed with water, brine, dried over Na2SO4. Concentration gave a brownish residue which was purified by flash column chromatography (10% MeOH / CH2CI2) to yield the title compound (4.21 g, 70%). 1H NMR (400 MHz, CD3OD) δ 7.42 (IH, d, 7 = 2.8 Hz), 7.23 (IH, d, 7 = 2.8 Hz), 3.71 (2H, t, 7 = 5.1 Hz), 3.67 (2H, t, 7 = 5.1 Hz), 3.04 (2H, t, 7 = 5.2 Hz), 2.98 (2H, t, 7= 5.2 Hz), 2.24 (3H, s), 2.13 (3H, s). LCMS (M+H)+ m/z 279 (t = 1.46 min.).
Figure imgf000055_0002
l-[4-(3,4-Diamino-5-methyl-phenyl)-piperazin-l-yl]-ethanone: To l-[4-(4-amino-3- methyl-5-nitro-phenyl)-piperazin-l-yl]-ethanone (4.5 g, 16.2 mmol) and 10% palladium on carbon (400 mg) were added methanol (50 mL) and acetic acid (5 mL) under nitrogen. The reaction mixture was stirred under hydrogen atmosphere (hydrogen balloon) for 14 h. The dark solution was filtered through a pad of celite and the filtercake was washed with methanol. Concentration of the filtrate gave the title compound (4.00 g, 100%) which was used for the next step without purification. LCMS (M+H)+ m/z 207 (t = 0.41 min.).
Figure imgf000056_0001
l-{4-[2-(4-Iodo-2-methoxy-pyridin-3-yl)-7-methyl-3H-benzimidazol-5-yl]-piperazin- 1-ylj-ethanone: To a solution of l-[4-(3,4-diamino-5-methyl-phenyl)-piperazin-l-yl]- ethanone (4.00 g, 16.18 mmol) in methanol (100 mL) was added 4-iodo-2-methoxy- pyridine-3-carbaldehyde (4.25 g, 16.18 mmol). The reaction mixture was stirred at room temperature for 14 h. After concentration, the residue was purified by flash column chromatography (10% MeOH / CH2C12) to yield the title compound (5.25 g, 66%). 1H NMR (400 MHz, CD3OD) δ 7.81 (IH, d, 7 = 5.4 Hz), 7.48 (IH, d, 7 = 5.4 Hz), 7.26 (IH, s), 6.85 (IH, s), 3.85 (3H, s), 3.78 (2H, t, 7 = 5.0 Hz), 3.64 (2H, t, 7 = 5.0 Hz), 3.16 (2H, t, 7 = 5.2 Hz), 3.11 (2H, t, 7 = 5.2 Hz), 2.62 (3H, s), 2.13 (3H, s). LCMS (M+H)+ m z 492 (t = 1.71 min.).
Figure imgf000056_0002
3-[6-(4-Acetyl-piperazin-l-yl)-4-methyl-lH-benzimidazol-2-yl]-4-chloro-lH- pyridin-2-one: To a solution of l-{4-[2-(4-Iodo-2-methoxy-pyridin-3-yl)-7-methyl- 3H-benzimidazol-5-yl]-piperazin-l-yl}-ethanone (5.2 g, 10.6 mmol) in 4 M HCl in dioxane (60 mL) was added water (5 mL). The reaction mixture was stirred at room temperature for 14 h. Concentration of the reaction mixture gave the title compound (4.02 g, 100%) which was used for the next step without purification. LCMS (M+H)+ m/z 486 (t = 1.55 min.).
Preparation of (S)-4-(l-Hvdroxymethyl-2-phenyl-ethylamino)-3-(4-methyl-6- piperazin-l-yl-lH-benzimidazol-2-yl)-lH-pyridin-2-one:
Figure imgf000057_0001
(S)-3-[6-(4-Acetyl-piperazin-l-yl)-4-methyl-lH-benzimidazol-2-yI]-4-(l- hydroxymethyl-2-phenyl-ethylamino)-lH-pyridin-2-one: To a solution of 3-[6-(4- acetyl-piperazin- 1 -yl)-4-methyl- 1 H-benzimidazol-2-yl] -4-chloro- 1 H-pyridin-2-one ( 1 g, 2.6 mmol) in DMF (10 mL) was added (S)-(-)-2-amino-3-phenyl-propanol (0.78 mg, 5.2 mmol) and N-methyl moφholine (2 mL). The reaction mixture was heated to 80°C for 12 h, cooled to room temperature and concentrated with high vacuum. The residue was purified by flash column chromatography (10% MeOH / CH2CI2) to yield the title compound (0.90 g, 69%). 1H ΝMR (400 MHz, CD3OD) δ 7.36 (IH, s), 7.02 - 7.23 (6H, m), 6.80 (IH, s), 5.98 (IH, d, 7 = 7.5 Hz), 4.10 - 4.12 (3H, m), 3.67 - 3.78 (6H, m), 3.06 - 3.11 (3H, m), 2.90 (IH, dd, 7 = 7.8, 13.6 Hz), 2.54 (3H, s), 2.12 (3H, s). LCMS (M+H)+ m/z 501 (t = 1.30 min.).
Figure imgf000057_0002
(S)-4-(l-Hydroxymethyl-2-phenyl-ethylamino)-3-(4-methyl-6-piperazin-l-yl-lH- benzimidazol-2-yl)-lH-pyridin-2-one: To a solution of (S)-3-[6-(4-acetyl-piperazin- 1 -yl)-4-methyl- 1 H-benzimidazol-2-yl] -4-( 1 -hydroxymethyl-2-phenyl-ethylamino)- lH-pyridin-2-one (900 mg, 18 mmol) in 4 M HCl in dioxane (10 mL) was added water (1 mL). The reaction mixture was heated to 80°C for 14 h and cooled to room temperature. Concentration with high vacuum gave the title compound (0.83 g, 100%) which was used for the next step without purification. LCMS (M+H)+ m/z 459 (t = 1.13 min.).
Preparation of 2-(4,6-Dichloro-pyrimidin-5-yl)-6-imidazol-l-yl-4-methyl-lH- benzimidazole:
Figure imgf000058_0001
4,6-Dichloro-pyrimidine-5-carbaldehyde: DMF (7 mL, 0.09 mol) was added to POCI3 (21 mL, 0.23 mol) at 0°C. The reaction mixture was stirred at room temperature for 0.5 h. 4,6-Dihydroxy-pyrimidine-5-carbaldehyde (5 g, 0.045 mol) was added in small portions. The reaction mixture was heated to 90°C for 6 h and cooled to room temperature. A large excess of crushed ice was added to the reaction mixture very slowly under ice-bath. The mixture was extracted with CH2CI2. The combined organic layers were washed with water, brine, and dried over Na2SO4. After concentration, the residue was purified by column chromatography (20% EtOAc / hexane) to yield the title compound (4 g, 50%). Η NMR (400 MHz, CDCI3) δ 8.91 (IH, s), 7.87 (IH, s). LRMS (M+H)+ m/z 177.
Figure imgf000058_0002
2-(4,6-Dichloro-pyrimidin-5-yl)-6-imidazol-l-yl-4-methyl-lH-benzimidazole: To a solution of 5-imidazol- 1 -yl-3-methyl-benzene- 1 ,2-diamine ( 180 mg, 0.96 mmol) in methanol (4 mL) was added a solution of 4,6-dichloro-pyrimidine-5-carbaldehyde ( 183 mg, 0.96 mmol) in methanol (1 mL). The reaction mixture was stirred at room temperature for 14 h. After concentration, the residue was purified by flash column chromatography (5% methanol / CH2Cl2) to yield the title compound (180 mg, 55%). LCMS (M+H)+ m z 344 (t = 1.31 min.).
Procedure for the Preparation of 2-Amino-4-Fluoro-6-methyl nitrobenzene;
Figure imgf000059_0001
2-(3,5-Difluoro-2-nitro-phenyl)-malonic acid di-tert-butyl ester: To a suspension of NaH (54.6g, 60%, 1.365 mol) in 600mL of DMF was added di-t-Butyl malonate (118g, 0.546 mol) at 0°C and stirred for 30 min. 2,4,6 trifluoronitrobenzene was added as a solution in 400 mL of DMF (75 g, 0.42mol) over 3 hours and the solution stirred at ambient temperature for 12 hours. The reaction mixture was extracted with ethyl acetate (3X's). The ethyl acetate was washed with water (3X's) and with brine and dried over MgSO4 and concentrated to give 62 g of crude product. LCMS [M+Na] - 396; Η NMR (500MHz, DMSO) δ 7.81 (m, IH), 7.27 (m, IH), 5.00 (s, IH), 1.41 (m, 18H).
Figure imgf000059_0002
2-(3-Amino-5-fluoro-2-nitro-phenyl)-malonic acid di-tert-butyl ester: To the crude 2-(3,5-Difluoro-2-nitro-phenyl)-malonic acid di-tert-butyl ester (62g, 0.42 mol) was added 700 mL of 2M ammonia in methanol in a pressure bottle. The vessel was sealed and heated to 85°C for 18 hours. The reaction mixture was cooled and the vessel opened carefully and the methanol solution concentrated to provide 140 g of crude material. LCMS [M+Na] - 393; 1H NMR (500MHz, DMSO) δ 6.76 (dd, J=10.8 2.8Hz, IH), 6.29 (dd, J=10.8, 2.8Hz, IH), 4.99 (brs, 2H), 4.80 (s, IH), 1.40 (m, 18H).
Figure imgf000060_0001
3-Amino-5-fluoro-2-nitro phenyl acetic acid: To the 2-(3-Amino-5-fluoro-2-nitro- phenyl)-malonic acid di-tert-butyl ester (140g) in 500 mL of 4N HCl in dioxane was added 50 mL of water and heated to 40°C for 2 days. The solution was extracted with ethyl acetate (3X's) and the ethyl acetate washed with water (3X's) and brine. The organic fraction was dried over MgSO4 and was concentrated to give 78g of crude (66% pure by LC/MS); 1H NMR (500MHz, DMSO) δ 12.40 (brs, IH), 7.04 (s, 2H), 6.68 (dd, J=10.9 2.8Hz, IH), 6.47 (dd, J=10.9, 2.8Hz, IH), 3.80 (s, 2H).
Figure imgf000060_0002
2-Amino-4-fluoro-6-methyI nitrobenzene: To the crude 3-Amino-5-fluoro-2-nitro phenyl acetic acid (3.6g, 16.8 mmol) was added Cu2O (lO.lg, 70.6 mmol) in 120 mL of acetonitrile along with 50uL of methanol and the suspension was refluxed for 12 hours. The reaction mixture was filtered through Celite and the Celite pad washed with water and ethyl acetate. The filtrate was extracted with ethyl acetate, washed with water and brine, dried over Na2SO4 and concentrated to give 2.95 g of material which by Η NMR was 80% pure. ESEVIS [M+Na] - 193; 1H NMR (500MHz, DMSO) δ 6.67 (s, 2H), 6.56 (dd, J=l l, 2.8Hz, IH), 6.39 (dd, J=l l, 2.8Hz, IH), 2.50 (s, 3H).
Procedure for the preparation of 4-Chloro-3-(4-methyl-6-morpholin-4-yl-lH- benzoimidazol-2-yl)-lH-pyridin-2-one and 4-Iodo-3-(4-methyl-6-morpholin-4-yl- lH-benzoimidazol-2-yl)-lH-pyridin-2-one
Figure imgf000061_0001
2-Methyl-4-morphoIin-4-yl-6-nitro-phenylamine: To a 800 ml pressure flask was added Tris(dibenzylideneacetone)dipalladium (2.64 g, 2.88 mmol), 2-(Di-t- butylphosphino)biphenyl (1.42 g, 4.75 mmol) and sodium tert-butoxide (17.5 g, 182 mmol). Then dry THF (500 mL), 4-bromo-2-methyl-6-nitroaniline (30.0 g, 130 mmol) and moφholine (34 ml, 390 mmol) were added. Argon was bubbled through the solution for 1 minute and the flask was sealed. The reaction mixture was stirred at 85 °C for 3 days. THF was evaporated in vacuo and the crude product was preabsorbed on silica and this then transferred on top of a silica gel column. Elution with hexane-ethyl acetate (6:4 to 4:6 to 0: 1 gradient) gave, after evaporation of solvents, the title compound (15.2 g red-brown solid, 49.3%). LCMS (M+H)+ m/z 238 (t = 0.64 min.) 1H NMR (500 MHz, DMSO-d6) δ 7.32 (IH, s), 7.22 (IH, s), 6.96 (2H, s), 3.72 (4H, broad s), 2.96 (4H, broad s), 2.21 (3H, s).
Figure imgf000061_0002
2-(4-Iodo-2-methoxy-pyridin-3-yl)-4-methyl-6-morpholin-4-yl-lH- benzoimidazole : 2-Methyl-4-moφholin-4-yl-6-nitro-phenylamine (15.2 g, 64 mmol) was suspended in methanol (200 ml) in a PARR flask. Palladium on carbon (1.0 g, 10% Pd) was added and the suspension shaken under 60 psi of hydrogen overnight. The mixture was filtered through a pad of celite (under argon) into a 3-neck flask, the celite rinsed with methanol and the filtrate diluted with methanol to a total volume of 500 ml and cooled to 0°C. A solution of 4-Iodo-2-methoxy-pyridine-3 -carbaldehyde (14.6 g, 55.5 mmol) in methanol (500 ml) was added slowly (during 3 hours). After addition of ~ lA of the solution the system was opened to air and stirred over the weekend, thereby reaching room temperature. The reaction mixture was concentrated in vacuo, filtered through a pad of silica (eluent: methylenechloride - ethyl acetate - methanol 55-40-5) then crystallized from ethyl acetate. The title compound was isolated as brown solid (12.68 g, 51%). Flash column chromatography of the mother liquor (gave additional 2.90 g (12%). [pack column with methylene chloride, elute compound with methylene chloride - ethyl acetate 6-4, then methylenechloride - ethyl acetate - methanol 58-40- 2]. LCMS (M+H)+ m z 451 (t = 1.03 min.). Η NMR (500 MHz, CDC13) δ 7.76 (IH, d, 7 = 5.3 Hz),), 7.42 (IH, d, 7 = 5.3 Hz), 6.85 (IH, broad s), 6.82 (IH, s), 3.86 (4H, t, 7 = 4.5 Hz), 3.79 (3H, s), 3.12 (4H, t, 7 = 4.5 Hz), 2.60 (3H, s), 2.21 (3H, s).
Figure imgf000062_0001
4-Chloro-3-(4-methyl-6-morpholin-4-yl-lH-benzoimidazol-2-yl)-lH-pyridin-2- one and 4-Iodo-3-(4-methyl-6-morpholin-4-yl-lH-benzoimidazol-2-yl)-lH- pyridin-2-one: 2-(4-Iodo-2-methoxy-pyridin-3-yl)-4-methyl-6-moφholin-4-yl- IH-benzoimidazole (15.58 g, 34.6 mmol) was suspended in 1,4-dioxane (300 ml) and cone, aqueous HCl (50 ml) was added. The mixture was stirred at ambient temperature overnight, then 3 hours at 50°C. The mixture was cooled to room temperature and poured into an ice cold solution of NaHCO3 (67 g, 0.8 mol) and filtered. The aqueous phase was extracted with ethyl acetate. The solid material was dissolved in CH2CI2 with some methanol, then extracted between water and CH2C12 . The combined organic layers were dried over Na2SO4, and concentrated in vacuo to give an inseparable mixture of the 4-Chloro- and 4-Iodo- title compounds. The product was used without further purification. LCMS (M+H)+ m/z 437 and m/z 345 (both t = 0.92 min.).
Procedure for the preparation of 4-r2-(4-Chloro-2-oxo-1.2-dihvdro-pyridin-3-yl)- 7-methyl-3H-benzoimidazol-5-yll-piperazine-l-carboxylic acid tert-butyl ester and 4-r2-(4-Iodo-2-oxo-l,2-dihydro-pyridin-3-yl)-7-methyl-3H-benzoimidazol-5- yl] -piperazine- 1 -carboxylic acid tert-butyl ester
Figure imgf000063_0001
4-(3-Amino-5-methyl-4-nitro-phenyl)-piperazine-l-carboxylic acid tert-butyl ester: To a stirred solution of 3-fluoro-5-amino-6-nitrotoluene (10 g, 58.79 mmol) in anhydrous NMP (160 mL) under nitrogen was addded BOC-piperazine (39 g, 209.4 mmol) and 4-methylmoφholine (25.9 mL). The resulting dark solution was heated to reflux for 72 h, cooled to room temperature and diluted with ethyl acetate (4000 mL). The organic layer was washed with water (8 x 1500 mL), brine (1 x 1500 mL), dried over sodium sulfate and evaporated in vacuo. The resulting dark oil was dissolved in boiling absolute ethanol (800 mL) and concentrated to a total volume of 400 mL and left to stand overnight at room temperature. The solution was further cooled to -20 °C for 5 h and the resulting solid was filtered off and dried in vacuo to give 16.3 g (83%) of a light yellow solid. 1H NMR (500 MHz, CDC13) δ 6.16 (brs, IH), 6.04 (brs, IH), 3.70 - 3.60 (m,4H), 3.38 - 3.25 (m, 4H), 2.53 (s, 3H), 1.48 (s, 9H); LCMS (M+H)+ m/z 337.
Figure imgf000063_0002
4-(3,4-Diamino-5-methyl-phenyl)-piperazine-l-carboxylic acid tert-butyl ester: To a stirred solution of 4-(3-Amino-5-methyl-4-nitro-phenyl)-piperazine-l -carboxylic acid tert-butyl ester (15 g, 44.6 mmol) in methanol (2200 mL) was added 20% Pd(OH)2/C (1.6 g) and the suspension flushed well with nitrogen, followed by hydrogen. The resulting suspension was stirred overnight at room temperature under an atmosphere of hydrogen (ca. 1 atm). The resulting suspension was filtered under nitrogen through a pad of Celite and washed with methanol (400 - 500 mL). The resulting material was used immediately. LCMS (M+H)+ m/z 307.
Figure imgf000064_0001
4-[2-(4-Iodo-2-methoxy-pyridin-3-yl)-7-methyl-3H-benzoimidazol-5-yl]- piperazine-1-carboxylic acid tert-butyl ester: To a stirred solution of 4-(3,4- Diamino-5-methyl-phenyl)-piperazine-l -carboxylic acid tert-butyl ester (44.6 mmol - assuming 100% conversion in previous step) in methanol (ca 2700 mL) at 0 oC under a nitrogen atmosphere was slowly added (2 h) via addition funnel, a solution of 4- iodo-2-methoxy-pyridine-3-carbaldehyde (15.0g, 57.1 mmol) in anhydrous methanol (225 mL). The resulting solution was then stirred at 0 °C for an additional 30 min,. the cooling bath was removed, and the reaction mixture was allowed to stir at room temperature in the presence of air for 72 h. The resulting solution was concentrated in vacuo and the residue was dissolved in dichloromethane (1500 mL) and the solvent removed in vacuo (repeated 3x). The resulting dark foamy solid was dried under high vacuum. 1H NMR (500 MHz, CDC13) δ 7.82 (d, IH, J = 5.4 Hz), 7.50 (d, IH, J = 5.4 Hz), 6.98 (brs, IH), 6.90 (brs, IH), 4.05 (s, 3H), 3.67 - 3.58 (m, 4H), 3.18 - 3.09 (m, 4H), 2.63 (s, 3H), 1.49 (s, 9H);. LCMS (M+H)+ m z 550.
Figure imgf000064_0002
HCl
4-Iodo-3-(4-methyl-6-piperazin-l-yl-lH-benzoimidazol-2-yl)-lH-pyridin-2-one:
To a stirred solution of 4-[2-(4-Iodo-2-methoxy-pyridin-3-yl)-7-methyl-3H- benzoimidazol-5-yl]-piperazine-l -carboxylic acid tert-butyl ester (24g, 43.7 mmol) was added 1,4-dioxane (750 mL) and 6 N aqueous HCl (30 mL) and the mixture was heated to 75 °C overnight. The solution was cooled to room temperature, the supernatent was poured off, and the resulting dark gummy solid was washed with anhydrous diethyl ether (3 x 500 mL) and dried in vacuo to give 17.7 g (93%) of the title compound as a dark solid that was used as described to prepare 4-[2-(4-Chloro-2- oxo-1, 2-dihydro-pyridin-3-yl)-7-methyl-3H-benzoimidazol-5-yl]-piperazine-l- carboxylic acid tert-butyl ester and 4-[2-(4-Iodo-2-oxo-l,2-dihydro-pyridin-3-yl)-7- methyl-3H-benzoimidazol-5-yl] -piperazine- 1 -carboxylic acid tert-butyl ester; LCMS (M+H)+ m z 436. (Note: A small peak (4-chloro-pyridin-2-one) in the LC/MS shows m/z 344, 346).
Figure imgf000065_0001
4-[2-(4-Chloro-2-oxo-l,2-dihydro-pyridin-3-yl)-7-methyl-3H-benzoimidazol-5- yl]-piperazine-l-carboxylic acid tert-butyl ester and 4-[2-(4-Iodo-2-oxo-l,2- dihydro-pyridin-3-yl)-7-methyl-3H-benzoimidazol-5-yl]-piperazine-l-carboxylic acid tert-butyl ester: To a stirred suspension of 4-Iodo-3-(4-methyl-6-piperazin-l- yl-lH-benzoimidazol-2-yl)-lH-pyridin-2-one (17.7 g, 40.7 mmol) in dichloromethane (750 mL) was added di-tert-butyl dicarbonate (9.8 g, 44.8 mmol) and triethylamine (67.4 mL, 483.6 mmol). The mixture was stirred at room temperature for 30 min. and purified via flash chromatography on silica gel. After elution with dichloromethane followed by 2.5% methanol/ethyl acetate, homogeneous fractions were combined and partially evaporated in vacuo to give the product as a yellow solid, following filtration (8.9 g, ca 41%, 2 crops). Η NMR (500 MHz, CD3OD) δ 7.25 (d, IH, J = 6.9 Hz), 6.97 (d, IH, J = 6.9 Hz), 6.97 (brs, IH), 6.89 (brs, IH), 3.65 - 3.56 (m, 4H), 3.16 - 3.07 (m, 4H), 2.55 (s, 3H), 1.49 (s, 9H); LCMS (M+H)+ m/z 536. (Note: A small peak (4-chloro-pyridin-2-one) in the LC/MS shows m z 444, 446).
Procedure for the preparation of 3-r6-(4-Amino-piperidin-l-yl)-4-methyl-lH- benzoimidazol-2-yll-4-iodo-lH-pyridin-2-one and 3-r6-(4-Amino-piperidin-l-yl)- 4-methyl-lH-benzoimidazol-2-yl1-4-chloro-lH-pyridin-2-one
Figure imgf000065_0002
[l-(3-Amino-5-methyl-4-nitro-phenyl)-piperidin-4-yl]-carbamic acid tert-butyl ester: 5-Fluoro-3-methyl-2-nitro-phenylamine (0.97 g, 5.7 mmol), 4-N-BOC- aminopiperidine (1.60g, 8.0 mmol), diisopropylethylamine (2.5 ml, 14 mmol) and DMSO (10 ml) are combined and stirred at 85°C for 3 hours. The reaction mixture was poured on saturated aqueous NaHCO3 solution and extracted with ethyl acetate. The organic layers were washed with water (3x) and brine, dried over Na2SO4 and concentrated. Flash column chromatography on silica (eluent hexanes-ethyl acetate - triethylamine 50-50-1, then 33-66-1) gave the title compound as a yellow solid. (1.57 g, 79%). LCMS (M+H)+ m z 351 (t = 1.55 min.). 1H NMR (500 MHz, CD3OD) δ 6.70 (IH, broad s), 6.22 (IH, d, 7 = 2.5 Hz)), 6.13 (IH, d, 7 = 2.5 Hz), 3.88 (2H, d, 7 = 13.3 Hz), 3.58 (IH, broad s), 2.98 (2H, t, 7 = 11.8 Hz), 2.48 (3H, s), 1.92 (2H, d, 7 = 11.3 Hz), 1.48 (2H, m), 1.45 (9H, s).
Figure imgf000066_0001
{l-[2-(4-Iodo-2-methoxy-pyridin-3-yl)-7-methyI-3H-benzoimidazol-5-yl]- piperidin-4-yl}-carbamic acid tert-butyl ester: [l-(3-Amino-5-methyl-4-nitro- phenyl)-piperidin-4-yl]-carbamic acid tert-butyl ester (1.54 g, 4.4 mmol) was dissolved in methanol (100 ml). Palladium on carbon (0.3 g, 10% Pd) was added and the suspension stirred vigorously under a balloon pressure of hydrogen overnight. The mixture was filtered through a pad of celite (under argon) into a 3-neck flask, the celite rinsed with methanol and the filtrate cooled to 0°C. A solution of 4-Iodo-2- methoxy-pyridine-3-carbaldehyde (1.21 g, 4.6 mmol) in methanol (50 ml) was added slowly (during 2 hours). The mixture was stirred overnight under air at ambient temperature, then concentrated in vacuo. Flash column chromatography on silica
(eluent hexanes-ethyl acetate -methanol 5-4-1 gave the title compound. (0.79 g, 32%). LCMS (M+H)+ m z 564 (t = 1.31 min.).
Figure imgf000067_0001
3-[6-(4-Amino-piperidin-l-yl)-4-methyl-lH-benzoimidazol-2-yl]-4-iodo-lH- pyridin-2-one and 3-[6-(4-Amino-piperidin-l-yl)-4-methyl-lH-benzoimidazol-2- yl]-4-chloro-lH-pyridin-2-one : { l-[2-(4-Iodo-2-methoxy-pyridin-3-yl)-7-methyl- 3H-benzoimidazol-5-yl]-piperidin-4-yl}-carbamic acid tert-butyl ester (330 mg, 0.59 mmol) was suspended in 4M HCl in 1 ,4-dioxane (20 ml) and water (3 ml) was added, (exothermic reaction). The mixture was stirred at ambient temperature overnight, then concentrated in vacuo to give an inseparable mixture of the 4-Chloro- and 4-Iodo- title compounds. The product was used without further purification. LCMS (M+H)+ m/z 450 and m/z 358 (both t = 0.69 min.).
Figure imgf000067_0002
3-Methyl-5-(2-morpholin-4-ethoxy)-2-nitro-phenylamine: To a solution of 2- moφholin-4-yl-ethanol (5 g, excess) in THF (30 mL) was added NaH (0.21 g, 8.82 mmol) in portion under ice bath. The reaction mixture was stirred at room temperature for 30 min. Then 5-fluoro-3-methyl-2-nitro-phenylamine was added. The reaction mixture was heated to reflux for 6 h, cooled to room temperature, and concentrated. The residue was diluted with water and extracted with EtOAc. The combined organic layers were washed with water, brine, and dried over Na2SO4. After concentration, the residue was purified by column chromatography (20% EtOAc / hexane) to yield the title compound (0.70 g, 85%). 1H NMR (400 MHz, CD3OD) δ 6.10 (IH, s), 6.09 (IH, s), 4.38 - 4.42 (2H, m), 3.92 - 4.08 (4H, m), 3.72 (IH, d, 7 = 12 Hz), 3.53 - 3.56 (2H, m), 3.05 - 3.10 (2H, m), 2.48 (3H, s). LCMS (M+H)+ m/z 282 (t = 0.73 min.).
Figure imgf000068_0001
(5)-7-Bromo-2-{4-[2-(3-chloro-phenyl)-2-hydroxy-ethylamino]-2-oxo-l,2- dihydro-pyridin-3-yl}-3H-benzimidazole-5-carbaldehyde: To a solution of (S)-7- Bromo-2-{4-[2-(3-chloro-phenyl)-2-hydroxy-ethylamino]-2-oxo-l,2-dihydro-pyridin- 3-yl}-3H-benzimidazole-5-carbonitrile (150 mg, 0.31 mmol) in THF (50 mL) was added DDBAL-H (1 M toluene solution, 1.55 mL, 1.55 mmol) at -78°C. The reaction mixture was stirred at -40°C for 10 h and cooled to -78°C. EtOAc (0.5 mL) was added. The reaction mixture was stirred for 30 min at-78°C before water (1 mL) was added. The reaction mixture was warmed to room temperature and concentrated. The residue was passed through a small pad of celite. The filtrate was concentrated and purified by prep. HPLC to give the titled compound (67 mg, 43%). Η NMR (400 MHz, CD3OD) δ 9.45 (IH, s), 7.62 (IH, s), 7.56 (IH, narrow d, 7 = 1.6 Hz), 7.44 (IH, narrow d, 7 = 1.0 Hz), 7.32 - 7.42 (2H, m), 7.24 - 7.30 (3H, m), 6.24 (IH, d, 7 = 7.6 Hz), 5.01 (IH, m), 3.65 - 3.76 (2H, m). LCMS (M+H)+ m z 487 (t = 1.76 min.).
Figure imgf000068_0002
5-(l,4,5,6-Tetrahydropyrimidin-l-yl)-3-methyl-2-nitro aniline: To a stirred solution of 2.0 g (11.76 mmol) of the 5-Flouro-3-methyl-2-nitro aniline in 10 mL of DMSO was added 1.2 g (14.11 mmol) of 1,4,5,6-Tetrahydropyrimidine, and 2.43 g (17.64 mmol) of potassium carbonate, and the mixture was heated at 100°C for 10 hrs, cooled, diluted with water, and extracted with Ethylacetate containing 5% methanol. The combined organic extract was washed with water, brine and dried (Na2SO4). Evaporation of the solvent furnished the residue, which was chromatographed (20% of 2M ammonia in methanol and dichloromethane) to produce 1.85 g (67%) of the product as red solid. Η NMR (400 MHz, CD3OD) δ 7.89 (lH,s), 6.53 (lH,d, 7 = 2.57 Hz), 6.44 ((lH,d, 7 = 2.1 Hz), 7.04 (IH, d, 7 = 2.1Hz), 3.70 (2H, t, 7 = 6.0 Hz), 3.41 (2H, t, J = 5.65 Hz, 2.43 (3H, s), 2.05 (2H, m) LCMS (M+H)+ m/z 235 (t = 0.78 min).
Example 1 (General procedure for examples 1-21)
Figure imgf000069_0001
(S)-4-(2-Hydroxy-l-phenyl-ethylamino)-3-(6-imidazoI-l-yl-4-methyl-lH- benzimidazol-2-yl)-lH-pyridin-2-one: To a solution of 3-(6-imidazol-l-yl-4-methyl- lH-benzimidazol-2-yl)-4-iodo-lH-ρyridin-2-one (30 mg, 0.072 mmol) in DMF (1 mL) were added (S)-(-)-2-phenylglycinol (26 mg, 0.18 mmol) and N- methylmoφholine (0.1 mL). The reaction mixture was heated to 80°C for 6 h and cooled to room temperature. The solvent was removed under vacuum and the residue was purified by prep. HPLC to yield the title compound (16 mg, 52%). 1H ΝMR (300 MHz, CD3OD) δ 8.07 (IH, narrow t, 7 = 1.7 Hz), 7.76 (2H, s), 7.27 - 7.48 (7H, m), 7.21 (IH, d, 7 = 7.5 Hz), 6.11 (IH, d, 7 = 7.5 Hz), 4.92 (IH, m), 4.03 (IH, dd, 7 = 4.5, 11.2 Hz), 3.95 (IH, dd, 7 = 6.2, 11.2 Hz), 2.75 (3H, s). LCMS (M+H)+ m/z 427 (t = 1.44 min.)
Example 2
Figure imgf000069_0002
(±)-4-[2-Hydroxy-2-(3-iodo-phenyI)-ethylamino]-3-(6-imidazol-l-yl-4-methyl-lH- benzimidazol-2-yl)-lH-pyridin-2-one: Η NMR (300 MHz, CD3OD) δ 9.41 (IH, s), 8.06 (IH, s), 7.88 (IH, s), 7.76 (IH, s), 7.71 (IH, s), 7.59 (IH, d, 7 = 7.8 Hz), 7.47 (IH, d, 7 = 7.8 Hz) 7.32 (IH, s) 7.29 (IH, d, 7 = 7.6 Hz) 7.09 (IH, t, 7 = 7.8 Hz), (6.24 (IH, d, 7 = 7.6 Hz), 4.97 (IH, dd, 7 = 5.0, 6.0 Hz), 3.75 (IH, dd, 7= 5.0, 13.5 Hz), 3.67 (IH, dd, 7 = 6.0, 13.5 Hz), 2.68 (3H, s). LCMS (M+H)+ m/z 553 (t = 1.43 min.).
Example 3
Figure imgf000070_0001
(±)-4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-(6-imidazol-l-yl-4-methyl- lH-benzimidazol-2-yl)-lH-pyridin-2-one: 1H NMR (300 MHz, CD3OD) δ 9.39 (IH, narrow t, 7 = 1.4 Hz), 8.04 (IH, narrow t, 7 = 1.7 Hz), 7.76 (IH, narrow t, 7 = 1.7 Hz), 7.69 (IH, narrow d, 7 = 1.9 Hz), 7.55 (IH, s), 7.23 - 7.42 (5H, m), 6.25 (IH, d, 7 = 7.6 Hz), 5.01 (IH, dd, 7 = 4.8, 6.4 Hz), 3.76 (IH, dd, 7 = 4.8, 13.4 Hz), 3.66 (IH, dd, 7 = 6.4, 13.4 Hz), 2.67 (3H, s). LCMS (M+H)+ m/z 461 (t = 1.46 min.).
Example 4
Figure imgf000070_0002
(±)-4-[2-(3-Bromo-phenyl)-2-hydroxy-ethylamino]-3-(6-imidazol-l-yl-4-methyl- lH-benzimidazol-2-yl)-lH-pyridin-2-one: Η NMR (300 MHz, CD3OD) δ 9.38 (IH, s), 8.02 (IH, s), 7.74 (IH, s), 7.69 (IH, s), 7.66 (IH, narrow d, 7 = 1.4 Hz), 7.20 - 7.48 (5H, m), 6.21 (IH, d, 7 = 7.6 Hz), 4.99 (IH, dd, 7 = 4.8, 6.3 Hz), 3.73 (IH, dd, 7 = 4.8, 13.5 Hz), 3.64 (IH, dd, 7 = 6.3, 13.5 Hz), 2.65 (3H, s). LCMS (M+H)+ m z 505 (t = 1.47 min.).
Example 5
Figure imgf000071_0001
(+)-N-(2-Chloro-4-{l-hydroxy-2-[3-(6-imidazol-l-yl-4-methyI-lH-benzimidazol- 2-yl)-2-oxo-l,2-dihydro-pyridin-4-ylamino]-ethyl}-phenyl)-methanesulfonamide:
1H ΝMR (300 MHz, CD3OD) δ 9.39 (IH, s), 8.05 (IH, s), 7.76 (IH, s), 7.68 (IH, s), 7.62 (IH, narrow d, 7 = 1.5 Hz), 7.52 (IH, s), 7.42 - 7.49 (2H, m), 7.31 (IH, s), 7.30 (IH, d, 7 = 7.6 Hz), 6.26 (IH, d, 7 = 7.6 Hz), 5.01 (IH, dd, 7 = 5.0, 5.6 Hz), 3.76 (IH, d, 7 = 5.0, 13.4 Hz), 3.74 (IH, dd, 7 = 5.6, 13.4 Hz), 2.98 (3H, s), 2.68 (3H, s). LCMS (M+H)+ m/z 554 (t = 1.11 min.).
Example 6
Figure imgf000071_0002
3-Bromo-benzoic-acid N'-[3-(6-imidazol-l-yl-4-methyl-lH-benzimidazol-2-yl)-2- oxo-l,2-dihydro-pyridin-4-yl]-hydrazide: Η ΝMR (300 MHz, CD3OD) δ 9.40 (IH, narrow t, 7 = 1.4 Hz), 8.14 (IH, narrow t, 7 = 1.7 Hz), 8.05 (IH, narrow t, 7 = 1.7 Hz), 7.95 (IH, d, 7 = 7.9 Hz), 7.81 (IH, d, 7 = 7.9 Hz), 7.74 - 7.77 (2H, m), 7.49 (IH, t, 7 = 7.9 Hz), 7.41 (IH, d, 7 = 7.4 Hz), 7.34 (IH, s), 6.38 (IH, d, 7 = 7.4 Hz), 2.70 (3H, s). LCMS (M+H)+ m/z 504 (t = 1.44 min.).
Example 7
Figure imgf000072_0001
4-Amino-benzoic-acid N'-[3-(6-imidazol-l-yl-4-methyl-lH-benzimidazol-2-yl)-2- oxo-l,2-dihydro-pyridin-4-yl]-hydrazide: 1H ΝMR (300 MHz, CD3OD) δ 9.41 (IH, s), 8.06 (IH, s), 7.76 - 7.79 (4H, m), 7.39 (IH, d, 7 = 7.4 Hz), 7.36 (IH, s), 6.81 (2H, d, 7 = 8.6 Hz), 6.36 (IH, d, 7 = 7.4 Hz), 2.70 (3H, s). LCMS (M+H)+ m/z 441 (t = 0.96 min.).
Example 8
Figure imgf000072_0002
(S)-4-[2-(2-Chloro-phenyl)-l-hydroxymethyl-ethylamino]-3-(6-imidazol-l-yl-4- methyl-lH-benzimidazol-2-yl)-lH-pyridin-2-one: Η ΝMR (300 MHz, CD3OD) δ 9.41 (IH, s), 8.05 (IH, narrow t, 7 = 1.7 Hz), 7.76 (IH, narrow t, 7 = 1.7 Hz), 7.72 (IH, narrow d, 7 = 1.9 Hz), 7.08 - 7.36 (6H, m), 6.10 (IH, d, 7 = 7.7 Hz), 3.98 - 4.24 (IH, m), 3.84 (IH, dd, 7 = 4.4, 11.2 Hz), 3.79 (IH, dd, 7 = 4.8, 11.2 Hz), 3.35 (IH, dd, 7 = 5.4, 13.6 Hz), 3.09 (IH, dd, 7 = 7.8, 13.6 Hz), 2.72 (3H, s). LCMS (M+H)+ m/z 475 (t = 1.56 min.).
Example 9
Figure imgf000073_0001
(5)-4-[2-(3-Chloro-phenyl)-l-hydroxymethyl-ethylamino]-3-(6-imidazol-l-yl-4- methyl-lH-benzimidazol-2-yl)-lH-pyridin-2-one: 1H NMR (300 MHz, CD3OD) δ 9.42 (IH, s), 8.07 (IH, narrow t, 7 = 1.7 Hz), 7.75 - 7.78 (2H, m), 7.14 - 7.37 (6H, m), 6.18 (IH, d, 7 = 7.7 Hz), 4.07 - 4.11 (IH, m), 3.76 - 3.77 (2H, m), 3.17 (IH, dd, 7 = 5.1, 13.7 Hz), 2.98 (IH, dd, 7 = 8.2, 13.7 Hz), 2.71 (3H, s). LCMS (M+H)+ m/z 475 (t = 1.57 min.).
Example 10
Figure imgf000073_0002
(S)-4-[2-(4-Chloro-phenyl)-l-hydroxymethyl-ethylamino]-3-(6-imidazoI-l-yl-4- methyl-lH-benzimidazol-2-yl)-lH-pyridin-2-one: Η NMR (300 MHz, CD3OD) δ 9.42 (IH, s), 8.07 (IH, narrow t, 7 = 1.6 Hz), 7.77 (IH, narrow t, 7 = 1.6 Hz), 7.73 (IH, narrow d, 7 = 1.9 Hz), 7.16 - 7.37 (6H, m), 6.19 (IH, d, 7 = 7.7 Hz), 4.06 - 4.10 (IH, m), 3.72 - 3.77 (2H, m), 3.14 (IH, dd, 7 = 5.3, 13.8 Hz), 2.98 (IH, dd, 7 = 7.8, 13.8 Hz), 2.69 (3H, s). LCMS (M+H)+ m/z 475 (t = 1.61 min.). Example 11
Figure imgf000074_0001
(S)-4-[2-(2-Bromo-phenyI)-l-hydroxymethyl-ethylamino]-3-(6-imidazol-l-yl-4- methyl-lH-benzimidazoI-2-yl)-lH-pyridin-2-one: 1H NMR (300 MHz, CD3OD) δ 9.41 (IH, s), 8.06 (IH, s), 7.75 - 7.77 (2H, m), 7.52 (IH, dd, 7 = 1.5, 7.5 Hz), 7.36 (IH, d, 7 = 1.9 Hz), 7.34 (IH, s), 7.03 - 7.16 (3H, m), 6.09 (IH, d, 7 = 7.7 Hz), 4.15 - 4.27 (IH, m), 3.82 (2H, m), 3.35 (IH, dd, 7 = 5.0, 13.6 Hz), 3.10 (IH, dd, 7 = 9.0, 13.6 Hz), 2.74 (3H, s). LCMS (M+H)+ m z 519 (t = 1.56 min.).
Example 12
Figure imgf000074_0002
(5)-4-[2-(3-Bromo-phenyl)-l-hydroxymethyl-ethylamino]-3-(6-imidazol-l-yl-4- methyl-lH-benzimidazol-2-yl)-lH-pyridin-2-one: 1H NMR (300 MHz, CD3OD) δ
9.41 (IH, s), 8.06 (IH, s), 7.74 - 7.77 (2H, m), 7.47 (IH, s), 7.24 - 7.31 (4H, m), 7.11 (IH, d, 7 = 7.7 Hz), 6.16 (IH, d, 7 = 7.7 Hz), 4.05 - 4.11 (IH, m), 3.76 (2H, m), 3.15 (IH, dd, 7 = 5.0, 13.6 Hz), 2.96 (IH, dd, 7 = 8.3, 13.6 Hz), 2.70 (3H, s). LCMS (M+H)+ m/z 519 (t = 1.54 min.). Example 13
Figure imgf000075_0001
(+)-4-(l-Hydroxymethyl-2-pentafluorophenyl-ethyIamino)-3-(6-imidazol-l-yl-4- methyl-lH-benzimidazol-2-yl)-lH-pyridin-2-one: 1H NMR (300 MHz, CD3OD) δ 9.40 (IH, s), 8.06 (IH, narrow t, 7 = 1.8 Hz), 7.77 (IH, narrow t, 7 = 1.7 Hz), 7.74 (IH, narrow d, 7 = 1.8 Hz), 7.35 (IH, s), 7.29 (IH, d, 7 = 7.6 Hz), 6.22 (IH, d, 7 = 7.6 Hz), 4.24 (IH, m), 3.82 (2H, dd, 7 = 2.6, 4.5 Hz), 3.23 (2H, t, 7 = 6.5 Hz), 2.70 (3H, s). LCMS (M+H)+ m z 531 (t = 1.61 min.).
Example 14
Figure imgf000075_0002
(S)-4-(l-Hydroxymethyl-2-pyridin-4-yl-ethylamino)-3-(6-imidazol-l-yl-4-methyl- lH-benzimidazol-2-yl)-lH-pyridin-2-one: 1H NMR (400 MHz, CD3OD) δ 9.42 (IH, s), 8.67 (2H, d, 7 = 6.6 Hz), 8.07 (2H, d, 7 = 6.6 Hz), 8.06 (IH, s), 7.77 (2H, s), 7.36 (IH, s), 7.28 (IH, d, 7 = 7. 6 Hz), 6.24 (IH, d, 7 = 7.6 Hz), 4.35 (IH, m), 3.82 (2H, d, 7 = 4.4 Hz), 3.50 (IH, dd, 7 = 4.4, 13.6 Hz), 3.40 (IH, dd, 7 = 8.7, 13.6 Hz), 2.71 (3H, s). LCMS (M+H)+ m z 442 (t = 0.96 min.). Example 15
Figure imgf000076_0001
(S)-4-(l-Hydroxymethyl-2-naphthalen-2-yl-ethylamino)-3-(6-imidazol-l-yl-4- methyl-lH-benzimidazol-2-yl)-lH-pyridin-2-one: 1H NMR (300 MHz, CD3OD) δ 7.99 (IH, narrow t, 7 = 1.7 Hz), 7.26 - 7.75 (1 IH, m), 7.15 (IH, d, 7 = 7.6 Hz), 6.19 (IH, d, 7 = 7.6 Hz), 4.16 - 4.20 (IH, m), 3.75 - 3.86 (2H, m), 3.30 (IH, dd, 7 = 5.4, 13.6 Hz), 3.15 (IH, dd, 7 = 7.6, 13.6 Hz), 2.60 (3H, s). LCMS (M+H)+ m/z 491 (t = 1.71 min.).
Example 16
Figure imgf000076_0002
(5)-4-(2-Cyclohexyl-l-hydroxymethyl-ethylamino)-3-(6-imidazol-l-yl-4-methyl- lH-benzimidazol-2-yl)-lH-pyridin-2-one: 1H NMR (300 MHz, CD3OD) δ 9.40 (IH, s), 8.06 (IH, narrow t, 7 = 1.7 Hz), 7.76 (IH, narrow t, 7 = 1.7 Hz), 7.73 (IH, narrow d, 7 = 1.7 Hz), 7.34 (IH, d, 7 = 7.6 Hz), 7.33 (IH, s), 6.34 (IH, d, 7= 7.6 Hz), 3.89 - 3.94 (IH, m), 3.68 (2H, d, 7 = 4.9 Hz), 2.68 (3H, s), 1.62 - 1.83 (7H, m), 0.95 - 1.26 (6H, m). LCMS (M+H)+ m/z 447 (t = 1.71 min.). Example 17
Figure imgf000077_0001
(3S,4R)-4-(3-Hydroxy-2,2-dimethyl-chroman-4-ylamino)-3-(6-imidazol-l-yl-4- methyl-lH-benzimidazol-2-yl)-lH-pyridin-2-one: 1H NMR (300 MHz, CD3OD) δ 9.36 (IH, s), 8.02 (IH, narrow t, 7 = 1.7 Hz), 7.73 (IH, narrow t, 7 = 1.7 Hz), 7.69 (IH, narrow d, 7 = 2.0 Hz), 7.37 (IH, d, 7 = 7.6 Hz), 7.18 - 7.30 (3H, m), 6.89 (IH, t, 7 = 7.6 Hz), 6.85 (IH, d, 7 = 8.4 Hz), 6.61 (IH, d, 7 = 7.6 Hz), 4.93 (IH, d, 7 = 8.2 Hz), 3.80 (IH, d, 7 = 8.2 Hz), 2.46 (3H, s), 1.48 (3H, s), 1.35 (3H, s). LCMS (M+H)+ m/z 483 (t = 1.70 min.).
Example 18
Figure imgf000077_0002
3-(6-Imidazol-l-yI-4-methyl-lH-benzimidazol-2-yl)-4-(2-thiophen-2-yl- ethylamino)-lH-pyridin-2-one: 1H NMR (500 MHz, DMSO-de) δ 13.17 (IH, broad s), 11.32 (IH, broad s), 11.03 (IH, broad s), 9.59 (IH, broad s), 8.22 (IH, broad s), 7.91 (IH, s), 7.82 (IH, d, 7 = 2.0 Hz), 7.41 (IH, t, 7 = 6.9 Hz), 7.36 (IH, dd, 7 = 1.0, 5.1 Hz), 7.30 - 7.40 (IH, m), 7.05 (IH, broad s), 7.01 (IH, t, 7 = 4.2 Hz), 6.23 (IH, d, 7 = 7.5 Hz), 3.74 (2H, t, 7 = 6.5 Hz), 3.25 (2H, t, 7 = 6.5 Hz), 2.58 (3H, s). Example 19
Figure imgf000078_0001
3-(6-Imidazol-l-yl-4-methyI-lH-benzimidazol-2-yl)-4-[2-(lH-indol-3-yl)- ethylamino]-lH-pyridin-2-one: Η NMR (500 MHz, DMSO-d6) δ 13.17 ( IH, broad s), 11.27 (broad s, IH), 11.00 (IH, broad s), 10.89 (IH, s), 9.58 (IH, broad s), 8.30 (IH, broad s), 7.91 (IH, s), 7.80 (IH, s), 7.63 (IH, d, 7 = 7.9 Hz), 7.40 (IH, t, 7 = 6.9 Hz), 7.34 (IH, d, 7 = 8.0 Hz), 7.30 - 7.40 (2H, m), 7.07 (IH, t, 7 = 7.5 Hz), 6.99 (IH, t, 7 = 7.6 Hz), 6.23 (IH, dd, 7 = 0.8, 7.2 Hz), 3.76 (2H, broad s), 3.17 (2H, t, 7 = 6.7 Hz), 2.50 (3H, s).
Example 20
Figure imgf000078_0002
3-(6-Imidazol-l-yl-4-methyl-lH-benzimidazol-2-yl)-4-(pyridin-2-yImethoxy)-lH- pyridin-2-one: To a solution of 3-(6-imidazol-l-yl-4-methyl-lH-benzimidazol-2-yl)- 4-iodo-lH-pyridin-2-one (25 mg, 0.06 mmol) in DMF (2 mL) were added pyridine carbinol (26 mg, 0.24 mmol) and cesium fluoride (36 mg, 0.24 mmol). The reaction mixture was heated to 130°C for 14 h and cooled to room temperature. After concentration in vacuo, the residue was purified by prep. HPLC to yield the title compound (8.2 mg, 34%). Η NMR (300 MHz, CD3OD) δ 9.58 (IH, narrow d, 7 = 1.2 Hz), 8.92 (IH, d, 7 = 4.9 Hz), 8.19 (IH, narrow t, 7 = 1.2 Hz), 8.12 (IH, s), 7.97 - 8.02 (2H, m), 7.85 (IH, narrow t, 7 = 1.8 Hz), 7.81 (IH, narrow t, 7 = 1.0 Hz), 7.63 (IH, d, 7 = 7.9 Hz), 7.54 (IH, t, 7 = 6.2 Hz), 6.81 (IH, 7 = 7.5 Hz), 5.86 (2H, s), 2.86 (3H, s). LCMS (M+H)+ m z 399 (t = 1.07 min.).
Example 21
Figure imgf000079_0001
(±)-4-[2-(3-Bromo-phenyI)-2-fluoro-ethylamino]-3-(6-imidazol-l-yl-4-methyl-lH- benzimidazol-2-yl)-lH-pyridin-2-one: 1H NMR (300 MHz, CD3OD) δ 9.40 (IH, t, 7 = 1.4 Hz), 8.06 (IH, t, 7 = 1.9 Hz), 7.76 (IH, t, 7 = 1.7 Hz), 7.72 (IH, d, 7 = 1.9 Hz), 7.66 (IH, s), 7.27 - 7.50 (5H, m), 6.29 (IH, d, 7 = 7.6 Hz), 5.75 - 5.94 (IH, m), 3.86 - 4.06 (2H, m), 2.64 (3H, s). LCMS (M+H)+ m/z 507 (t = 1.70 min.).
Example 22 (General procedure for examples 22-28)
Figure imgf000079_0002
(5)-2-[4-(l-Hydroxymethyl-2-phenyl-ethylamino)-2-oxo-l,2-dihydro-pyridin-3- yl]-7-methyl-3H-benzimidazole-5-carbonitrile: To a solution of 2-(4-chloro-2-oxo- l,2-dihydro-pyridin-3-yl)-7-methyl-3H-benzimidazole-5-carbonitrile (0.7 g, 2.19 mmol) in DMF (15 mL) were added N-methylmoφholine (0.66 g, 6.57 mmol) and (S)-(-)-2-amino-3-phenyl-l-propanol (0.40 g, 2.63 mmol). The reaction mixture was heated to 80°C for 6 h and then cooled to room temperature. After concentration in vacuo, the residue was purified by flash chromatography (3% MeOH / CH2CI2) to yield the title compound (0.59 g, 68%) as a yellow foam.Η ΝMR (400 MHz, CD3OD) δ 7.74 (IH, s), 7.63 (IH, s), 7.12 - 7.27 (6H, m), 6.07 (IH, d, 7 = 7.5 Hz), 3.97 (IH, m), 3.74 (2H, t, 7 = 5 Hz), 3.14 (IH, dd, 7 = 5.5, 14.0 Hz), 2.94 (IH, dd, 7 : 7.9, 14.0 Hz), 2.60 (3H, s). LCMS (M+H)+ m/z 400 (t = 1.71 min.).
Example 23
Figure imgf000080_0001
(±)-2-{4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-2-oxo-l,2-dihydro-pyridin- 3-yl}-7-methyl-3H-benzimidazole-5-carbonitriIe: 1H NMR (300 MHz, DMSO-d6) δ 7.92 (IH, s), 7.59 (IH, s), 7.28 - 7.47 (5H, m), 6.19 (IH, d, 7 = 7.3 Hz), 4.92 - 4.96 (IH, m), 3.53 - 3.73 (2H, m), 2.58 (3H, s). LCMS (M+H)+ m/z 420 (t = 1.99 min.).
Example 24
Figure imgf000080_0002
(5)-2-{4-[2-(3-ChIoro-phenyl)-l-hydroxymethyl-ethyIamino]-2-oxo-l,2-dihydro- pyridin-3-yl}-7-methyl-3H-benzimidazole-5-carbonitrile: 1H NMR (400 MHz, CD3OD) δ 7.86 (IH, s), 7.10 - 7.33 (6H, m), 6.12 (IH, d, 7 = 7.6 Hz), 4.01 - 4.05 (IH, m), 3.75 (2H, d, 7 = 4.9 Hz), 3.15 (IH, dd, 7 = 4.9, 13.5 Hz), 2.86 - 3.00 (IH, m), 2.63 (3H, s). LCMS (M+H)+ m/z 434 (t = 1.81 min.). Example 25
Figure imgf000081_0001
(±)-2-{4-[2-(3-Bromo-4-methoxy-phenyl)-2-hydroxy-ethylamino]-2-oxo-l,2- dihydro-pyridin-3-yl}-7-methyl-3H-benzimidazole-5-carbonitrile: 1H NMR (300 MHz, CD OD) δ 7.86 (IH, s), 7.75 (IH, s), 7.67 (IH, s), 7.28 - 7.40 (2H, m), 6.95 (IH, d, 7 = 8.5 Hz), 6.24 (IH, d, 7 = 7.4 Hz), 4.93 (IH, m), 3.65 - 3.97 (2H, m), 3.82 (3H, s), 2.57 (3H, s). LCMS (M+H)+ m/z 494 (t = 2.10 min.).
Example 26
Figure imgf000081_0002
(±)-2-{4-[2-(3-Fluoro-phenyl)-2-hydroxy-ethylamino]-2-oxo-l,2-dihydro-pyridin- 3-yI}-7-methyl-3H-benzimidazole-5-carbonitrile: 1H NMR (400 MHz, CD3OD) δ 7.70 (IH, s), 7.49 (IH, s), 7.22 - 7.32 (4H, m), 6.92 (IH, d, 7 = 8.9 Hz), 6.17 (IH, d, 7 = 7.2 Hz), 4.92 (IH, t, 7 = 6.3 Hz), 3.66 (2H, d, 7 = 5.9 Hz), 2.56 (3H, s). LCMS (M+H)+ m z 404 (t = 1.65 min.).
Example 27
Figure imgf000081_0003
(±)-2-{4-[2-(3-Bromo-phenyl)-2-hydroxy-ethylamino]-2-oxo-l,2-dihydro-pyridin- 3-yl}-7-methyl-3H-benzimidazoIe-5-carbonitrile: Η NMR (300 MHz, CD3OD) δ 7.92 (IH, s), 7.72 (IH, s), 7.26 - 7.50 (5H, m), 6.19 (IH, d, 7 = 7.1 Hz), 4.93 (IH, t, 7 = 4.3 Hz), 3.47 - 3.73 (2H, m), 2.58 (3H, s). LCMS (M+H)+ m/z 464 (t = 2.00 min.).
Example 28
Figure imgf000082_0001
(S)-2-[4-(2-Hydroxy-2-phenyl-ethylamino)-2-oxo-l,2-dihydro-pyridin-3-yl]-7- methyl-3H-benzimidazole-5-carbonitrile: 1H NMR (400 MHz, CD3OD) δ 8.04 (IH, s), 7.74 (IH, s), 7.24 - 7.51 (6H, m), 6.22 (IH, d, 7 = 7.5 Hz), 5.00 (IH, m), 3.64 - 3.74 (2H, m), 2.60 (3H, s). LCMS (M+H)+ m/z 386 (t = 1.65 min.).
Examples 29-35 were prepared from commercially or readily available diamines which were prepared and condensed with 4-iodo-2-methoxy-pyridine-3-carbaldehyde as described in Scheme UI.
Example 29
Figure imgf000082_0002
(+)-4-[2-(3-Bromo-4-methoxy-phenyl)-2-hydroxy-ethyIamino]-3-(4,5,6-trifluoro- lH-benzimidazol-2-yl)-lH-pyridin-2-one: Η NMR (500 MHz, CD3COCD3) δ
13.26 (IH, broad s), 10.93 (IH, broad s), 10.27 (IH, broad s), 7.76 (IH, s), 7.54 - 7.56 (2H, m), 7.41 - 7.42 (IH, m), 7.06 (IH, d, 7 = 8.2 Hz), 6.29 (IH, d, 7= 7.5 Hz), 5.05 - 5.07 (IH, m), 3.87 (3H, s), 3.74 - 3.79 (IH, m), 3.65 - 3.69 (IH, m).
Example 30
Figure imgf000083_0001
(+)-4-[2-(3-Bromo-4-methoxy-phenyl)-2-hydroxy-ethylamino]-3-(4,6-dibromo- lH-benzimidazol-2-yl)-lH-pyridin-2-one: 1H NMR (500 MHz, CD3COCD3) δ
13.18 (IH, broad s), 11.18 (IH, broad s), 10.20 (IH, broad s), 7.92 (IH, s), 7.78 (IH, s), 7.52 - 7.55 (3H, m), 7.03 (IH, d, 7 = 8.5 Hz), 6.28 (IH, d, 7 = 7.4 Hz), 5.06 - 5.08 (IH, m), 3.86 (3H, s), 3.73 - 3.77 (IH, m), 3.66 -3.70 (IH, m).
Example 31
Figure imgf000083_0002
(±)-4-[2-(3-Bromo-4-methoxy-phenyl)-2-hydroxy-ethylamino]-3-(5,6-dichloro- lH-benzimidazol-2-yl)-lH-pyridin-2-one: Η NMR (500 MHz, CD3COCD3) δ 7.79
(3H, m), 7.53 (IH, dd, 7 = 2.0, 8.4 Hz), 7.41 - 7.44 (IH, m), 7.08 (IH, d, 7 = 8.4 Hz),
6.29 (IH, d, 7 = 7.5 Hz), 5.05 - 5.08 (IH, m), 3.89 (3H, s), 3.74 - 3.78 (IH, m), 3.70 - 3.72 (lH, m). Example 32
Figure imgf000084_0001
(±)-3-(lH-Benzimidazol-2-yl)-4-[2-(3-bromo-phenyl)-2-hydroxy-ethylamino]-lH- pyridin-2-one: 1H NMR (500 MHz, CD3COCD3) δ 7.80 (IH, s), 7.64 - 7.69 (2H, m), 7.60 (IH, d, 7 = 7.7 Hz), 7.46 - 7.48 (2H, m), 7.33 (IH, t, 7 = 7.8 Hz), 7.19 - 7.22 (2H, m), 6.59 (IH, d, 7 = 4.5 Hz), 5.12 - 5.14 (IH, m), 5.13 (IH, dd, 7 = 4.5, 7.2 Hz), 3.82 (IH, dd, 7 = 4.5, 13.6 Hz), 3.71 (IH, dd, 7 = 7.2, 13.6 Hz).
Example 33
.
Figure imgf000084_0002
3-(lH-Benzimidazol-2-yl)-4-[(pyridin-2-yImethyl)-amino]-lH-pyridin-2-one: 1H NMR (500 MHz, CD3COCD3) δ 8.96 - 8.98 (IH, m), 8.46 - 8.49 (IH, m), 8.03 (IH, d, 7 = 8.0 Hz), 7.93 (IH, t, 7 = 6.4 Hz), 7.69 - 7.73 (2H, m), 7.53 (IH, d, 7 = 7.5 Hz), 7.28 - 7.32 (2H, s), 6.34 (IH, d, 7 = 7.5 Hz), 5.29 (2H, m).
Example 34
Figure imgf000084_0003
(S)-3-(lH-Benzimidazol-2-yl)-4-(l-hydroxymethyl-2-phenyl-ethyIamino)-lH- pyridin-2-one: Η NMR (500 MHz, CD3COCD3) δ 7.67 - 7.69 (2H, m), 7.15 - 7.44 (8H, m), 6.31 (IH, d, 7 = 7.5 Hz), 4.10 - 4.13 (IH, m), 3.76 - 3.82 (2H, m), 3.23 (IH, dd, 7 = 5.6, 13.7 Hz), 3.04 (IH, dd, 7 = 8.1, 13.7 Hz).
Example 35
Figure imgf000085_0001
(±)-3-(lH-Benzimidazol-2-yl)-4-[2-(3-bromo-4-methoxy-phenyl)-2-hydroxy- ethylamino]-lH-pyridin-2-one: ]H NMR (500 MHz, CD3COCD3) δ 7.78 (IH, s), 7.68 (2H, s), 7.50 - 7.55 (2H, m), 7.21 - 7.24 (2H, m), 7.06 (IH, d, 7 = 8.4 Hz), 6.44 (IH, d, 7 = 7.5 Hz), 5.08 (IH, dd, 7 = 4.6, 7.2 Hz), 3.87 (3H, s), 3.79 (IH, dd, 7 = 4.6, 13.4 Hz), 3.71 (IH, dd, 7 = 7.2, 13.4 Hz).
Examples 36 - 43 were prepared according to Scheme V.
Example 36 (General procedure for examples 36-43)
Figure imgf000085_0002
(S)-4-{2-[4-(l-Hydroxymethyl-2-phenyl-ethylamino)-2-oxo-l,2-dihydro-pyridin- 3-yl]-7-methyl-3H-benzimidazol-5-yI}-piperazine-l-carboxylic acid isøpropylamide: To a solution of (S)-4-(l-hydroxymethyl-2-phenyl-ethylamino)-3- (4-methyl-6-piperazin-l-yl-lH-benzimidazol-2-yl)-lH-pyridin-2-one (30 mg, 0.063 mmol) in methanol (2 mL) was added isopropyl isocyanate (2 drops). The reaction mixture was stirred at room temperature for 5 min. Concentration gave the residue, which was purified by prep. HPLC to yield the title compound (23.8 mg, 69%). 1H NMR (400 MHz, CD3OD) δ 7.55 (IH, s), 7.14 - 7.28 (7H, m), 6.12 (IH, d, 7 = 7.8 Hz), 4.01 - 4.03 (IH, m), 3.92 (quintet, 7 = 6.6 Hz), 3.80 (4H, m), 3.76 (IH, dd, 7 = 4.8, 11.2 Hz), 3.70 (IH, dd, 7 = 5.2, 11.2 Hz), 3.61 - 3.64 (4H, m), 3.09 (IH, dd, 7 = 5.6, 13.7 Hz), 2.91 (IH, dd, 7 = 8.0, 13.7 Hz), 2.64 (3H, s), 1.19 (6H, d, 7 = 6.8 Hz). LCMS (M+H)+ m z 545 (t = 1.99 min.).
Example 37
Figure imgf000086_0001
(S)-4-{2-[4-(l-Hydroxymethyl-2-phenyl-ethylamino)-2-oxo-l,2-dihydro-pyridin-3-yl]- 7-methyl-3H-benzimidazol-5-yl}-piperazine-l-carboxylic acid ethylamide: 1H NMR (400 MHz, CD3OD) δ 7.55 (IH, s), 7.12 - 7.28 (7H, m), 6.12 (IH, d, 7 = 7.8 Hz), 4.01 - 4.05 (IH, m), 3.62 - 3.81 (10H, m), 3.24 (2H, q, 7= 7.2 Hz), 3.08 (IH, dd, 7 = 5.6, 13.7 Hz), 2.91 (IH, dd, 7 = 8.0, 13.7 Hz), 2.65 (3H, s), 1.14 (3H, t, 7 = 7.2 Hz). LCMS (M+H)+ m/z 531 (t = 1.93 min.).
Example 38
Figure imgf000086_0002
(S)-4-(l-Hydroxymethyl-2-phenyl-ethylamino)-3-{4-methyl-6-[4-(l-phenyl- methanoyl)-piperazin-l-yl]-lH-benzimidazoI-2-yl}-lH-pyridin-2-one: To a solution of (S)-4-( 1 -hydroxymethyl-2-phenyl-ethylamino)-3-(4-methyl-6-piperazin- 1 -yl- 1 H- benzimidazol-2-yl)-lH-pyridin-2-one (30 mg, 0.063 mmol) in methanol (2 mL) was added benzoyl chloride (1 drop). The reaction mixture was stirred for 5 min. and concentrated. The residue was purified by prep. HPLC to yield the title compound (11 mg, 33%). 1H NMR (400 MHz, CD3OD) δ 7.48 - 7.54 (5H, m), 7.37 (IH, s), 7.13 - 7.26 (7H, m), 6.08 (IH, d, 7 = 7.7 Hz), 3.79 - 4.02 (5H, m), 3.76 (IH, dd, 7 = 4.7, 11.2 Hz), 3.67 (IH, dd, 7 = 5.7, 11.2 Hz), 3.54 (4H, m), 3.03 (IH, dd, 7 = 5.7, 13.7 Hz), 2.89 (IH, dd, 7 = 8.0, 13.7 Hz), 2.62 (3H, s). LCMS (M+H)+ m z 563 (t = 2.19 min.).
Example 39
Figure imgf000087_0001
(S)-4-(l-Hydroxymethyl-2-phenyl-ethylamino)-3-[6-(4-wopropyI-piperazin-l-yl)-4- methyl-lH-benzimidazol-2-yl]-lH-pyridin-2-one: To a solution of (S)-4-(l- hydroxymethyl-2-phenyl-ethylamino)-3 -(4-methyl-6-piperazin- 1 -yl- 1 H-benzimidazol-2- yl)-lH-pyridin-2-one (25 mg, 0.054 mmol) in methanol (0.5 mL) was added acetone (0.25 mL) and 1 M THF solution of NaCNBH3 (0.2 mL). The reaction mixture was stirred at room temperature for 1 h and concentrated in vacuo. The residue was purified by prep. HPLC to yield the title compound (12 mg, 44%). 1H NMR (400 MHz, CD3OD) δ 7.10 - 7.24 (6H, m), 7.08 (IH, s), 7.06 (IH, s), 6.06 (IH, d, 7 = 7.9 Hz), 3.58 - 4.01 (10H, m), 3.34 (IH, m), 3.14 (IH, m), 2.98 (IH, dd, 7 = 5.9, 13.7 Hz), 2.83 (IH, dd, 7 = 7.9, 13.7 Hz), 2.60 (3H, s), 1.44 (6H, d, 7 = 6.7 Hz). LCMS (M+H)+ m/z 501 (t = 1.80 min.). Example 40
Figure imgf000088_0001
(5)-3-[6-(4-Benzyl-piperazine-l-yl)-4-methyl-lH-benzimidazol-2-yl]-4-(l- hydroxymethyl-2-phenyl-ethylamino)-lH-pyridin-2-one: 1H NMR (400 MHz, CD3OD) δ 7.50 - 7.57 (6H, m), 7.04 - 7.27 (7H, m), 6.07 (IH, d, 7 = 7.8 Hz), 4.22 (2H, s), 3.97 - 4.00 (IH, m), 3.34 - 3.77 (2H, m), 2.82 - 3.04 (10H, m), 2.58 (3H, s). LCMS (M+H)+ m z 549 (t = 1.93 min.).
Example 41
Figure imgf000088_0002
(±)-3-[6-(4-Acetyl-piperazine-l-yl)-4-methyl-lH-benzimidazol-2-yl]-4-[2-(3-chloro- phenyl)-2-hydroxy-ethylamino]-lH-pyridin-2-one: 1H NMR (400 MHz, CD3OD) δ 7.50 (IH, s), 7.25 - 7.49 (5H, m), 7.20 (IH, s), 6.25 (IH, d, 7 = 8.0 Hz), 4.94 (IH, dd, 7 = 4.6, 7.1 Hz), 3.88 - 3.92 (4H, m), 3.50 - 3.65 (6H, m), 2.62 (3H, s), 2.20 (3H, s). LCMS (M+H)+ m/z 521 (t = 2.13 min.).
Example 42
Figure imgf000089_0001
(±)-4-[2-(3-ChIoro-phenyl)-2-hydroxy-ethylamino]-3-(4-methyl-6-piperazin-l-yl-lH- benzimidazol-2-yI) -lH-pyridin-2-one: 1H NMR (400 MHz, CD3OD) δ 7.47 (IH, s), 7.25 - 7.38 (4H, m), 7.07 (2H, s), 6.25 (IH, d, 7 = 7.6 Hz), 4.90 (IH, m), 3.42 - 3.66 (10 H, m), 2.59 (3H, s). LCMS (M+H)+ m/z 479 (t = 1.90 min.).
Example 43
Figure imgf000089_0002
(±)-4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-[6-(4-isøpropyl-piperazine- l-yl)-4-methyl-lH-benzimidazol-2-yl]-lH-pyridin-2-one: 1H NMR (400 MHz, CD3OD) δ 7.47 (IH, s), 7.25 - 7.38 (4H, m), 7.06 (2H, s), 6.26 (IH, d, 7= 7.6 Hz), 4.91 - 4.93 (IH, m), 3.89 - 3.92 (2H, m), 3.51 - 3.64 (5H, m), 3.31 - 3.37 (2H, m), 3.09 - 3.30 (2H, m), 2.59 (3H, s), 1.44 (6H, d, 7 = 6.6 Hz). LCMS (M+H)+ m z 521 (t = 1.95 min.).
Example 44
Figure imgf000089_0003
(S)-6-(l-Hydroxymethyl-2-phenyl-ethylamino)-5-(6-imidazoI-l-yl-4-methyl-lH- benzimidazol-2-yl)-3H-pyrimidin-4-one:
(S)-2-[6-Chloro-5-(6-imidazol-l-yl-4-methyl-lH-benzimidazol-2-yI)-pyrimidin-4- ylamino]-3-phenyl-propan-l-ol: To a solution of 2-(4,6-dichloro-pyrimidin-5-yl)-6- imidazol-l-yl-4-methyl-lH-benzimidazole (40 mg, 0.16 mmol) in tsøpropanol (5 mL) was added (S)-(-)-2-amino-3-phenyl-propanol (35 mg, 0.23 mmol) and triethylamine (0.5 mL). The reaction mixture was heated to 80°C for 4 h, cooled to room temperature and concentrated with high vacuum. The crude product was used for the next step without purification. LCMS (M+H)+ m/z 460 (t = 2.13 min.).
(S)-6-(l-HydroxymethyI-2-phenyl-ethylamino)-5-(6-imidazol-l-yl-4-methyl-lH- benzimidazoI-2-yl)-3H-pyrimidin-4-one: To a solution of (S)-2-[6-chloro-5-(6- imidazol-l-yl-4-methyl-lH-benzimidazol-2-yl)-pyrimidin-4-ylamino]-3-phenyl- propan-1-ol in 4 N HCl (0.5 mL) and acetic acid (0.5 mL) was added two drops of water. The reaction mixture was heated to 100°C for 8 h, cooled to room temperature, and neutralized with ammonia in methanol. After concentration, the residue was purified by prep. HPLC to yield the title compound (26 mg, 37% for two steps). 1H NMR (400 MHz, CD3OD) δ 9.41 (IH, narrow t, 7 = 1.5 Hz), 8.06 (IH, narrow dd, 7 = 1.6, 1.9 Hz), 7.93 91H, s), 7.77 (IH, narrow dd, 7= 1.6, 1.8 Hz), 7.73 (IH, narrow d, 7 = 1.9 Hz), 7.11 - 7.21 (6H, m), 4.70 - 4.74 (IH, m), 3.74 (2H, d, 7 = 4.7 Hz), 3.11 (IH, dd, 7 = 6.1, 13.5 Hz), 3.00 (IH, dd, 7 = 7.8, 13.5 Hz), 2.70 (3H, s). LCMS (M+H)+ m/z 442 (t = 2.17 min.).
Examples 45 - 383 were prepared according to the general methods described above (Scheme III)
LCMS conditions: a) YMC C18 S5 4.6 x 50 mm; 0 - 100% gradient over 4 min*; 4 mL/min flow rate b) YMC ODS-A C18 S7 3.0 x 50 mm; 0 - 100% gradient over 2 min*; 5 mL/min flow rate c) YMC C18 S5 4.5 x 50 mm; 0 - 100% gradient over 8 min*; 2.5 mLmin flow rate d) YMC C18 S7 3.0 x 50 mm; 0 - 100% gradient over 3 min*; 5 mL/min flow rate e) YMC ODSA S3 6.0 xl50 mm; 0 - 100% gradient over 5 min*; 1.5 mL/min flow rate f) PHS-PRIMESPHERE C 18 4.6 x 30 mm; 0 - 100% gradient over 2 min* ; 5 mL/min flow rate g) YMC C18 S7 3.0 x 50 mm; 0 - 100% gradient over 4 min*; 5 mL/min flow rate h) YMC ODS-A C18 S7 3.0 x 50 mm; 0 - 100% gradient over 2 min*; 5 mL/min flow rate i) YMC ODS-A C18 S7 3.0 x 50 mm; 0 - 100% gradient over 1.5 min*; 5 mL/min flow rate j) YMC Xterra C18 S7 3.0 x 50 mm; 0 - 100% gradient over 2 min*; 5 mL/min flow rate k) YMC Pro-ODS C18 S5 4.6x33 mm; 0 - 100% gradient over 3 min*; 4 mL/min flow rate 1) YMC ODS-A C18 S7 3.0 x 50 mm; 0 - 100% gradient over 4 min*; 4 mlJmin flow rate * Gradient begin with 10% methanol/90% water (0.1% TFA) and end with 90% methanol/10% water (0.1% TFA)
Figure imgf000091_0001
Figure imgf000092_0001
Figure imgf000093_0001
Figure imgf000094_0001
Figure imgf000095_0001
Figure imgf000096_0001
Figure imgf000097_0001
Figure imgf000098_0001
Figure imgf000099_0001
Figure imgf000100_0001
Figure imgf000101_0001
Figure imgf000102_0001
Figure imgf000103_0001
Figure imgf000104_0001
Figure imgf000105_0001
Figure imgf000106_0001
Figure imgf000107_0001
Figure imgf000108_0001
Figure imgf000109_0001
Figure imgf000110_0001
Figure imgf000111_0001
Figure imgf000112_0001
Figure imgf000113_0001
Figure imgf000114_0001
Figure imgf000115_0001
Figure imgf000116_0001
Figure imgf000117_0001
Figure imgf000118_0001
Figure imgf000119_0001
Figure imgf000120_0001
11!
Figure imgf000121_0001
Figure imgf000122_0001
Figure imgf000123_0001
Figure imgf000124_0001
Figure imgf000125_0001
Figure imgf000126_0001
Figure imgf000127_0001
Figure imgf000128_0001
Figure imgf000129_0001
Figure imgf000130_0001
Figure imgf000131_0001
Figure imgf000132_0001
Figure imgf000133_0001
Figure imgf000134_0001
Figure imgf000135_0001
Figure imgf000136_0001
Figure imgf000137_0001
Figure imgf000138_0001
Figure imgf000139_0001
Intermediate Imidate Formation (Scheme IV, 13)
Figure imgf000140_0001
(S)-2-[4-(l-Hydroxymethyl-2-phenyl-ethylamino)-2-oxo-l,2-dihydro-pyridin-3- yl]-7-methyl-3H-benzimidazole-5-carboximidic ethyl ester: To a suspension of (S)- 2-[4-(l-hydroxymethyl-2-phenyl-ethylamino)-2-oxo-l,2-dihydro-pyridin-3-yl]-7- methyl-3H-benzimidazole-5-carbonitrile (0.8 g, 2.0 mmol) in ethanol (anhydrous, 80 mL) was bubbled HCl (anhydrous) at 0°C until saturation. The mixture became a clear solution after a few minutes of bubbling and the solution was stirred at room temperature for 14 h. After concentration in vacuo, the crude product (0.89 g, 100%) was directly used for the next step without purification. LCMS (M+H)+ m/z 446 (t = 1.55 min).
Figure imgf000140_0002
(+)-2-{4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-2-oxo-l,2-dihydro-pyridin- 3-yl}-7-methyl-3H-benzimidazole-5-carboximidic ethyl ester: The title compound was prepared using the General Procedure for Imidate Formation. LCMS (M+H)+ m/z 466 (t = 1.43 min.).
General Procedure for Examples 384 - 397 - Imidazoline Formation (Scheme IV, 14) Example 384
Figure imgf000141_0001
(S)-3-[6-(4,5-Dihydro-lH-imidazol-2-yl)-4-methyl-lH-benzimidazol-2-yl]-4-(l- hydroxymethyl-2-phenyl-ethylamino)-lH-pyridin-2-one: The crude imidate ester (60 mg, 0.135 mmol) was diluted with methanol followed by addition of ethylenediamine (24 mg, 0.40 mmol). The reaction mixture was heated to reflux for 6 h. After concentration in vacuo, the residue was purified by prep. HPLC to yield the title compound (37 mg, 62%). 1H NMR (300 MHz, CD3OD) δ 7.94 (IH, s), 7.50 (IH, s), 7.12 - 7.30 (6H, m), 6.16 (IH, d, J = 7.7 Hz), 4.04 - 4.10 (5H, m), 3.75 - 3.77
(2H, m), 3.15 (IH, dd, J = 5.2, 13. 6 Hz), 2.96 (IH, dd, J = 8.1, 13.6 Hz), 2.67 (3H, s). LCMS (M+H)+ m/z 443 (t = 1.50 min.).
Example 385
Figure imgf000141_0002
(±)-4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-[6-(4,5-dihydro-lH- imidazol-2-yl)-4-methyI-lH-benzimidazol-2yl]-lH-pyridin-2-one: Η NMR (300 MHz, CD3OD) 5 7.93 (IH, s), 7.54 (IH, s), 7.23 - 7.45 (5H, m), 6.26 (IH, d, J = 7.6 Hz), 5.01 (IH, t, 7 = 6.3 Hz), 4.01 (4H, s), 3.78 (IH, dd, J = 4.7, 13.5 Hz), 3.67 (IH, dd, J = 6.6, 13.5 Hz), 2.66 (3H, s). LCMS (M+H)+ m/z 463 (t = 1.54 min Example 386
4-[2-(3-Chloro-phenyl)-2R-hydroxy-ethylamino]-3-[4-methyI-6-(4S-methyl-4,5- dihydro-lH-imidazol-2-yl)-lH-benzoimidazoI-2-yl]-lH-pyridin-2-one and 4-[2- (3-Chloro-phenyl)-25-hydroxy-ethylamino]-3-[4-methyl-6-(4S-methyl-4,5- dihydro-lH-imidazol-2-yl)-lH-benzoimidazol-2-yl]-lH-pyridin-2-one: 1H NMR (300 MHz, CD3OD) δ 7.90 (2H, s), 7.53 (2H, s), 7.22 - 7.47 (10H, m), 6.23 (2H, d, 7 = 7.5 Hz), 5.00 (2H, t, 7 = 6.4 Hz), 4.49 - 4.57 (2H, m), 4.21 (2H, t, 7 = 11.0 Hz), 3.61 - 3.78 (6H, m), 2.63 (6H, s), 1.48 (6H, d, 7 = 6.3 Hz). LCMS (M+H)+ m/z 477 (t = 1.71 min.).
Example 387
Figure imgf000142_0002
(±)-4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-[4-methyl-6-(l-methyI-4,5- dihydro-lH-imidazol-2-yl)-lH-benzimidazol-2yl]-lH-pyridin-2-one: 'H NMR (300 MHz, CD3OD) δ 7.94 (IH, s), 7.57 (IH, s), 7.23 - 7.52 (5H, m), 6.22 (IH, d, 7 = 7.5 Hz), 4.99 (IH, m), 4.00 - 4.15 (2H, m), 3.57 - 3.75 (4H, m), 2.77 (3H, s), 2.61 (3H, s). LCMS (M+H)+ m/z 477 (t = 1.60 min.).
Example 388
Figure imgf000143_0001
(±)-4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-[6-(4,4-dimethyl-4,5- dihydro-lH-imidazol-2-yl)-4-methyl-lH-benzimidazol-2yl] -lH-pyridin-2-one: ' H NMR (300 MHz, CD3OD) δ 7.89 (IH, s), 7.52 (IH, s), 7.22 - 7.45 (5H, m), 6.19 (IH, d, 7 = 7.4 Hz), 4.99 (IH, t, 7 = 6.2 Hz), 3.84 (2H, s), 3.78 (IH, dd, 7 = 4.4, 13.4 Hz), 3.60 (IH, dd, 7 = 6.7, 13.4 Hz), 2.61 (3H, s), 1.55 (6H, s). LCMS (M+H)+ m/z 491 (t = 1.73 min.).
Example 389
Figure imgf000143_0002
2-(2-{4-[2-(3-Chloro-phenyl)-2R-hydroxy-ethylamino]-2-oxo-l,2-dihydro- pyridin-3-yl}-7-methyl-3H-benzimidazol-5-yl)-4,5-dihydro-lH-imidazole-4S- carboxylic acid and 2-(2-{4-[2-(3-Chloro-phenyl)-2S-hydroxy-ethylamino]-2-oxo- l,2-dihydro-pyridin-3-yI}-7-methyl-3H-benzimidazol-5-yl)-4,5-dihydro-lH- imidazole-4S-carboxylic acid: Η NMR (300 MHz, CD3OD) 6 7.88 (2H, s), 7.52 (2H, s), 7.22 - 7.44 (10H, m), 6.17 (2H, d, 7 = 7.4 Hz), 5.05 (2H, m), 4.23 - 4.38 (4H, m), 3.56 - 3.72 (6H, m), 2.58 (6H, s). LCMS (M+H)+ m/z 507 (t = 1.64 min.). Example 390
Figure imgf000144_0001
(±)-4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-[4-methyI-6-(l,4,5,6- tetrahydro-pyrimidin-2-yl)-lH-benzimidazol-2-yl]-lH-pyridin-2-one: 1H NMR
(300 MHz, CD3OD) δ 7.90 (IH, s), 7.53 (IH, s), 7.22 - 7.47 (5H, m), 6.23 (IH, d, 7 = 7.6 Hz), 5.00 (IH, t, 7 = 6.3 Hz), 4.49 - 4.57 (IH, m), 3.60 - 3.77 (5H, m), 3.24 - 3.34 (2H, m), 2.63 (3H, s). LCMS (M+H)+ m/z 477 (t = 1.59 min.).
Example 391
Figure imgf000144_0002
(±)-4-[2-(3-Chloro-4-methoxy-phenyl)-2-methoxy-ethylamino]-3-[6-(4,5-dihydro- lH-imidazol-2-yl)-4-methyl-lH-benzimidazol-2-yl]-lH-pyridin-2-one: 1H NMR (300 MHz, CD3OD) δ 7.88 (IH, s), 7.46 (IH, s), 7.44 (IH, s), 7.31 (IH, d, 7 = 6.4 Hz), 7.28 (IH, d, 7 = 5.6 Hz), 7.01 (IH, d, 7 = 6.4 Hz), 6.19 (IH, d, 7 = 5.6 Hz), 4.49 (IH, dd, 7 = 3.4, 5.0 Hz), 4.09 (4H, s), 3.82 (3H, s), 3.36 (3H, s), 3.63 - 3.67 (2H, m), 2.62 (3H, s). LCMS (M+H)+ m/z 507 (t = 1.72 min.). Example 392
Figure imgf000145_0001
(±)-4-[2-(3-Bromo-4-methoxy-phenyl)-2-hydroxy-ethylamino]-3-[6-(4,5-dihydro- lH-imidazol-2-yl)-4-methyl-lH-benzimidazol-2-yl]-lH-pyridin-2-one: The title compound was prepared following hydrolysis of the benzylic chloride to the hydroxyl. Η NMR (400 MHz, CD3OD) δ 7.90 (IH, s), 7.67 (IH, s), 7.48 (IH, s), 7.38 (IH, dd, 7 = 2.0, 8.5 Hz), 7.31 (IH, d, 7 = 7. 6 Hz), 6.95 (IH, d, 7 = 8.5 Hz), 6.26 (IH, d, 7 = 7.6 Hz), 4.94 (IH, m), 4.10 (4H, s), 3.80 (3H, s), 3.62 - 3.74 (2H, m), 2.62 (3H, s). LCMS (M+H)+ m/z 537 (t = 1.49 min.).
Example 393
Figure imgf000145_0002
(5)-3-[6-(4,4-Dimethyl-4,5-dihydro-lH-imidazol-2-yI)-4-methyl-lH- benzimidazol-2-yl]-4-(l-hydroxymethyl-2-phenyl-ethylamino)-lH-pyridin-2-one:
Η NMR (300 MHz, CD3OD) δ 7.94 (IH, s), 7.50 (IH, s), 7.12 - 7.30 (6H, m), 6.15 (IH, dd, 7 = 7.7 Hz), 4.02 - 4.10 (IH, m), 3.85 (2H, s), 3.75 - 3.77 (2H, m), 2.96 - 3.17 (2H, m), 2.67 (3H, s), 1.56 (6H, s). LCMS (M+H)+ m/z 471 (t = 1.62 min.). Example 394
Figure imgf000146_0001
(S)-2-{2-[4-(lS-HydroxymethyI-2-phenyl-ethylamino)-2-oxo-l,2-dihydro-pyridin- 3-yl]-7-methyl-3H-benzimidazol-5-yl}-4,5-dihydro-lH-imidazoIe-4S-carboxylic acid. Η NMR (300 MHz, CD3OD) δ 8.03 (IH, s), 7.60 (IH, s), 7.11 - 7.29 (6H, m), 6.15 (IH, d, 7 = 7.3 Hz), 5.07 (IH, dd, 7 = 6.9, 11.8 Hz), 4.25 - 4.41 (2H, m), 4.05 - 4.07 (IH, m), 3.70 - 3.82 (2H, m), 3.12 (IH, dd, 7 = 5.5, 13.6 Hz), 2.95 (IH, dd, 7 = 8.0, 13.6 Hz), 2.69 (3H, s). LCMS (M+H)+ m/z 487 (t = 1.35 min.).
Example 395
Figure imgf000146_0002
(S)-4-(l-Hydroxymethyl-2-phenyl-ethylamino)-3-[4-methyI-6(l,4,5,6-tetrahydro- pyrimidin-2-yl)-lH-benzimidazol-2-yl]-lH-pyridin-2-one: 1H NMR (300 MHz, CD3OD) δ 7.76 (IH, s), 7.32 (IH, s), 7.09 - 7.29 (6H, m), 6.15 (IH, d, 7 = 7.6 Hz), 4.02 - 4.07 (IH, m), 3.71 - 3.79 (2H, m), 3.61 (4H, t, 7 = 5.6 Hz), 3.14 (IH, dd, 7 = 5.3, 13.6 Hz), 2.95 (IH, dd, 7 = 8.1, 13.6 Hz), 2.65 (3H, s), 2.09 - 2.16 (2H, m). LCMS (M+H)+ m/z 457 (t = 1.49 min.). Example 396
Figure imgf000147_0001
(S)-4-(l-Hydroxymethyl-2-phenyl-ethyIamino)-3-[4-methyl-6-(l-methyl-4,5- dihydro-lH-imidazol-2-yl)-lH-benzoimidazo-2-yl]-lH-pyridin-2-one: 1H NMR (300 MHz, CD3OD) δ 7.77 (IH, s), 7.10 - 7.30 (7H, m), 6.16 (IH, d, 7 = 7.6 Hz), 3.97 - 4.20 (5H, m), 3.73 - 3.77 (2H, m), 3.26 (3H, s), 3.15 (IH, dd, 7= 5.3, 13.6 Hz), 2.96 (IH, dd, 7 = 8.0, 13.6 Hz), 2.68 (3H, s). LCMS (M+H)+ m/z 457 (t = 1.53 min.)
Example 397
Figure imgf000147_0002
(S)-3-[6-(4,5-Dihydro-lH-imidazol-2-yl)-4-methyl-lH-benzimidazol-2-yl]-4-(2- hydroxy-2-phenyI-ethylamino)-lH-pyridin-2-one: 1H NMR (400 MHz, CD3OD) δ 7.91 (IH, s), 7.50 (IH, s), 7.48 (2H, s), 7.24 - 7.35 (4H, m), 6.25 (IH, d, 7 = 7.6 Hz), 5.01 (IH, dd, 7 = 4.6, 7.0 Hz), 4.10 (4H, s), 3.65 - 3.76 (2H, m), 2.64 (3H, s). LCMS (M+H)+ m/z 429 (t = 1.48 min.). Example 398
Figure imgf000148_0001
(S)-4-(l-Hydroxymethyl-2-phenyl-ethylamino)-3-[6-(lH-imidazol-2-yl)-4-methyl- lH-benzimidazol-2-yl]-lH-pyridin-2-one: To a solution of the imidate ester (150 mg, 0.31 mmol) in methanol (10 mL) was added aminoacetaldehyde diethyl acetal (97 mg, 0.93 mmol). The mixture was stirred at room temperature for 14 h. The solvent was removed in vacuo and the residue was treated with 60% HClO4 (5 mL) at room temperature for 14 h. The reaction mixture was then neutralized with ammonium hydroxide (cone). The inorganic salt was filtered. The filtrate was concentrated and purified by prep. HPLC to yield the title compound (37 mg, 27%) as a white solid. 1H NMR (300 MHz, CD3OD) δ 7.94 (IH, narrow d, 7 = 1.3 Hz), 7.58 (2H, s), 7.53 (IH, narrow d, 7 = 0.6 Hz), 7.12 - 7.31 (6H, m), 6.16 (IH, d, 7 = 7.7 Hz), 4.04 - 4.08 (IH, m), 3.76 - 3.79 (2H, m), 3.16 (IH, dd, 7 = 5.4, 13.6 Hz), 3.97 (IH, dd, 7 = 8.1, 13.6 Hz), 2.69 (3H, s). LCMS (M+H)+ m/z 441 (t = 1.60 min.).
General Procedure for Amidine Formation, Examples 399 - 412 (Scheme IV, 16) Example 399
Figure imgf000148_0002
(S)-2-[4-(l-Hydroxymethyl-2-phenyI-ethylamino)-2-oxo-l,2-dihydro-pyridin-3- yl]-7,N-dimethyl-3H-benzimidazole-5-carboxamidine: The crude imidate ester (60 mg, 0.135 mmol) was diluted with methanol (10 mL), then methylamine (2.0 M methanol solution, 0.5 mL, excess) was added to the solution. After stirring for 2 h, the reaction mixture was concentrated and purified by prep. HPLC to yield the title compound (32 mg, 55%) as a white solid. 1H NMR (300 MHz, CD3OD) δ 7.82 (IH, s), 7.38 (IH, s), 7.10 - 7.30 (6H, m), 6.15 (IH, 7 = 7.7 Hz), 4.03 - 4.07 (IH, m), 4.72 - 4.76 (2H, m), 3.06 - 3.18 (IH, m), 3.12 (3H, s), 2.96 (IH, dd, 7 = 8.0, 13.5 Hz), 2.67 (3H, s). LCMS (M+H)+ m z 431 (t = 1.34 min.).
Example 400
Figure imgf000149_0001
(±)-2-{4-[2-(3-Chloro-phenyl)-2-hydroxy-ethyIamino]-2-oxo-l,2-dihydro-pyridin- 3-yl}-7,N-dimethyl-3H-benzimidazole-5-carboxamidine: 1H ΝMR (300 MHz, CD3OD) δ 7.81 (IH, s), 7.54 (IH, s), 7.23 - 7.54 (5H, m), 6.26 (IH, d, 7 = 7.6 Hz), 5.01 (IH, t, 7 = 4.8 Hz), 3.77 (IH, dd, 7 = 4.5, 13.4 Hz), 3.67 (IH, dd, 7= 6.6, 13.4 Hz), 3.12 (3H, s), 2.66 (3H, s). LCMS (M+H)+ m/z 451 (t = 1.32 min.).
Example 401
Figure imgf000149_0002
(±)-2-{4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-2-oxo-l,2-dihydro-pyridin- 3-yl}-7-methyI-3H-benzimidazole-5-carboxamidine: 1H NMR (300 MHz, CD3OD) δ 7.90 (IH, s), 7.26 -7.57 (6H, m), 6.25 (IH, d, 7 = 7.6 Hz), 5.01 (IH, t, 7 = 6.4 Hz), 3.75 (IH, dd, 7 = 4.8, 13.4 Hz), 3.66 (IH, dd, 7 = 6.7, 13.4 Hz), 2.62 (3H, s). LCMS (M+H)+ m/z 437 (t = 1.59 min.). Example 402
Figure imgf000150_0001
(+)-2-{4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-2-oxo-l,2-dihydro-pyridin- 3-yl}-7,N,N,-trimethyl-3H-benzimidazole-5-carboxamidine: 1H NMR (300 MHz, CD3OD) δ 7.63 (IH, s), 7.54 (IH, s), 7.18 - 7.41 (5H, m), 6.26 (IH, d, 7 = 7. 6 Hz), 5.01 (IH, t, 7 = 6.3 Hz), 3.77 (IH, dd, 7 = 4.7, 13.5 Hz), 3.67 (IH, dd, 7 = 6.6, 13.5 Hz), 3.34 (6H, s), 2.66 (3H, s). LCMS (M+H)+ m/z 465 (t = 1.57 min.).
Example 403
Figure imgf000150_0002
(+)-2-{4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-2-oxo-l,2-dihydro-pyridin- 3-yl}-N-cyclopropyl-7-methyl-3H-benzimidazole-5-carboxamidine: Η ΝMR (300 MHz, CD3OD) δ 7.78 (IH, s), 7.54 (IH, s), 7.22 - 7.41 (5H, m), 6.25 (IH, d, 7 = 7.6 Hz), 5.01 (IH, t, 7 = 6.4 Hz), 3.76 (IH, dd, 7 = 4.7, 13.5 Hz), 3.67 (IH, dd, 7 = 6.6, 13.5 Hz), 2.77 - 2.83 (IH, m), 2.65 (3H, s), 1.03 - 1.10 (2H, m), 0.86 - 0.92 (2H, m). LCMS (M+H)+ m/z 477 (t = 1.43 min.). Example 404
Figure imgf000151_0001
(±)-2-{4-[2-(3-Chloro-4-methoxy-phenyl)-2-methoxy-ethylamino]-2-oxo-l,2- dihydro-pyridin-3-yl}-7,N-dimethyl-3H-benzimidazole-5-carboxamidine: 1H NMR (300 MHz, CD3OD) δ 7.79 (IH, narrow d, 7 = 1.0 Hz), 7.45 (IH, narrow d, 7 = 1.5 Hz), 7.38 (IH, s), 7.28 - 7.31 (2H, m), 7.02 (IH, d, 7 = 6.4 Hz), 6.22 (IH, d, 7 = 5.7 Hz), 4.52 (IH, t, 7 = 5.9 Hz), 3.83 (3H, s), 3.60 - 3.71 (2H, m), 3.36 (3H, s), 3.13 (3H, s), 2.65 (3H, s). LCMS (M+H)+ m/z 495 (t = 1.64 min.).
Example 405
Figure imgf000151_0002
(±)-2-{4-[2-(3-Chloro-4-methoxy-phenyl)-2-hydroxy-ethylamino]-2-oxo-l,2- dihydro-pyridin-3-yl}-7,N-dimethyl-3H-benzimidazole-5-carboxamidine: To a solution of 2-{4-[2-(3-chloro-4-methoxy-phenyl)-2-methoxy-ethylamino]-2-oxo-l,2- dihydro-pyridin-3-yl}-7,N-dimethyl-3H-benzimidazole-5-carboxamidine (40 mg, 0.08 mmol) in methanol (5 mL) was added 2 Ν ΝaOH (0.5 mL) and the reaction mixture was stirred at room temperature for 14 h. After concentration in vacuo, the residue was purified by prep. HPLC to yield the title compound (7.2 mg, 19%) as oil. 1H
ΝMR (300 MHz, CD3OD) δ 7.78 (IH, narrow d, 7 = 1.6 Hz), 7.52 (IH, narrow d, 7 = 2.1 Hz), 7.30 - 7.36 (3H, m), 6.99 (IH, d, 7 = 8.5 Hz), 6.27 (IH, d, 7 = 7.6 Hz), 4.94 (IH, t, 7= 5.9 Hz), 3.82 (3H, s), 3.68 - 3.72 (2H, m), 3.13 (3H, s), 2.63 (3H, s). LCMS (M+H)+ m/z 481 (t = 1.44 min.). Example 406
Figure imgf000152_0001
(±)-2-{4-[2-(3-Bromo-4-methoxy-phenyl)-2-hydroxy-ethylamino]-2-oxo-l,2- dihydro-pyridin-3-yl}-7^V-dimethyl-3H-benzimidazole-5-carboxamidine: Η
NMR (400 MHz, CD3OD) δ 7.77 (IH, s), 7.67 (IH, s), 7.29 - 7.39 (3H, m), 6.95 (IH, d, 7 = 8.4 Hz), 6.23 (IH, d, 7 = 4.4 Hz), 4.92 (IH, m), 3.80 (3H, s), 3.69 (2H, m), 3.13 (3H, s), 2.61 (3H, s). LCMS (M+H)+ m/z 525 (t = 1.44 min.).
Example 407
Figure imgf000152_0002
(S)-N-Cyclopropyl-2-[4-(l-hydroxymethyl-2-phenyI-ethylamino)-2-oxo-l,2- dihydro-pyridin-3-yl]-7-methyl-3H-benzimidazole-5-carboxamidine: 1H ΝMR (300 MHz, CD3OD) δ 7.81 (IH, s), 7.37 (IH, s), 7.10 - 7.30 (6H, m), 6.16 (IH, d, 7 = 7.6 Hz), 4.03 - 4.08 (IH, m), 3.74 - 3.76 (2H, m), 3.15 (IH, dd, 7 = 5.3, 13.6 Hz), 2.96 (IH, dd, 7= 8.1, 13.6 Hz), 2.78 - 2.84 (IH, m), 2.67 (3H, s), 1.03 - 1.10 (2H, m), 0.87 - 0.92 (2H, m). LCMS (M+H)+ m/z 457 (t = 1.51 min.). Example 408
Figure imgf000153_0001
(5)-N-Ethyl-2-[4-(l-hydroxymethyl-2-phenyl-ethyIamino)-2-oxo-l,2-dihydro- pyridin-3-yl]-7-methyI-3H-benzimidazole-5-carboxamidine: 1H NMR (400 MHz, CD3OD) δ 7.82 (IH, s), 7.38 (IH, s), 7.12 - 7.30 (6H, m), 6.15 (IH, d, 7= 7.8 Hz), 4.04 - 4.07 (IH, m), 3.75 - 3.77 (2H, m), 3.51 (2H, q, 7 = 7.3 Hz), 3.15 (IH, dd, 7 = 5.4, 13.7 Hz), 2.96 (IH, dd, 7 = 8.1, 13.7 Hz), 2.67 (3H,s), 1.40 (3H, t, 7= 7.3 Hz). LCMS (M+H)+ m/z 445 (t = 1.44 min.).
Example 409
Figure imgf000153_0002
(5)-2-{4-[2-(3-Chloro-phenyl)-l-hydroxymethyl-ethylamino]-2-oxo-l,2-dihydro- pyridin-3-yl}-7,N-dimethyl-3H-benzimidazole-5-carboxamidine: Η ΝMR (400 MHz, CD3OD) δ 7.83 (IH, s), 7.38 (IH, s), 7.31 (IH, s), 7.10 - 7.24 (4H, m), 6.16 (IH, d, 7 = 7.6 Hz), 4.06 - 4.09 (IH, m), 3.72 - 3.77 (2H, m), 3.16 (3H, s), 3.15 (IH, m), 2.96 (IH, dd, 7 = 8.2, 13.7 Hz), 2.67 (3H, s). LCMS (M+H)+ m/z 465 (t = 1.49 min.). Example 410
Figure imgf000154_0001
(S)-2-{4-[2-(2-ChIoro-phenyl)-l-hydroxymethyl-ethylamino]-2-oxo-l,2-dihydro- pyridin-3-yl}-7,N-dimethyl-3H-benzimidazole-5-carboxamidine: Η ΝMR (400 MHz, CD3OD) δ 7.84 (IH, s), 7.41 (IH, s), 7.32 - 7.35 (2H, m), 7.06 - 7.18 (3H, m), 6.10 (IH, d, 7 = 7.5 Hz), 4.20 - 4.24 (IH, m), 3.76 - 3.85 (2H, m), 3.30 - 3.34 (IH, m), 3.06 - 3.14 (IH, m), 3.13 (3H, s), 2.64 (3H, s). LCMS (M+H)+ m/z 465 (t = 1.48 min.).
Example 411
Figure imgf000154_0002
(±)-2-{4-[2-(3-Bromo-phenyl)-2-hydroxy-ethylamino]-2-oxo-l,2-dihydro-pyridin- 3-yl}-7,N-dimethyl-3H-benzimidazole-5-carboxamidine: 1H ΝMR (300 MHz, CD3OD) δ 7.80 (IH, s), 7.69 (IH, s), 7.20 - 7.45 (5H, m), 6.25 (IH, d, 7 = 7.3 Hz), 5.00 (IH, t, 7 = 5.9 Hz), 3.64 - 3.80 (2H, m), 3.12 (3H, s), 2.66 (3H, s). LCMS (M+H)+ m/z 495 (t = 1.49 min.).
Example 412
Figure imgf000154_0003
(S)-2-[4-(2-Hydroxy-2-phenyl-ethylamino)-2-oxo-l,2-dihydro-pyridin-3-yl]-7^V- dimethyl-3H-benzimidazole-5-carboxamidine: 1H NMR (400 MHz, CD3OD) δ 7.78 (IH, s), 7.24 - 7.49 (7H, m), 6.21 (IH, d, 7 = 7.0 Hz), 4.00 - 5.05 (IH, m), 3.61 - 3.74 (2H, m), 3.12 (3H, s), 2.63 (3H, s). LCMS (M+H)+ m/z 417 (t = 1.28 min.).
Figure imgf000155_0001
Figure imgf000156_0001
Figure imgf000157_0001
Figure imgf000158_0001
Figure imgf000159_0001
Figure imgf000160_0001
Figure imgf000161_0001
Figure imgf000162_0001
Figure imgf000163_0001
Amide Formation Intermediate Synthesis
Figure imgf000163_0002
(±)-2-{4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-2-oxo-l,2-dihydro-pyridin- 3-yl}-7-methyI-3H-benzimidazole-5-carboxylic acid ethyl ester: A solution of (±)- 2-{4-[2-(3-chloro-phenyl)-2-hydroxy-ethylamino-]-2-oxo-l,2-dihydro-pyridin-3-yl}- 7-methyl-3H-benzimidazole-5-carboximidic acid ethyl ester (400 mg, 0.90 mmol) was diluted with 2 N HCl (20 mL) solution and the mixture was allowed to stir at room temperature for 14 h. After concentration to dryness, the crude product (419 mg, 100%) was used for the next step without purification. Small amount was purified by prep. HPLC to yield the title compound. Η NMR (300 MHz, CD3OD) δ 8.11 ( IH, s), 7.74 (IH, s), 7.25 - 7.54 (5H, m), 6.25 (IH, d, 7 = 7.6 Hz), 4.99 (IH, t, 7 = 7.2 Hz), 4.38 (2H, q, 7 = 7.1 Hz), 3.61 - 3.76 (2H, m), 2.61 (3H, s), 1.42 (3H, q, 7 = 7.1 Hz). LCMS (M+H)+ m/z 467 (t = 2.23 min.).
Figure imgf000164_0001
(+)-2-{4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-2-oxo-l,2-dihydro-pyridin- 3-yl}-7-methyl-3H-benzimidazole-5-carboxylic acid: The ethyl ester (419 mg, 0.90 mmol) obtained above was diluted with methanol (15 mL) and water (5 mL) followed by addition of sodium hydroxide (180 mg, 4.5 mmol). The mixture was stirred at room temperature for 14 h. After removal of methanol, the residue was neutralized with 2 N HCl solution. The resulting slurry was filtered and washed with ice-cold water. The solid was collected after drying over high vacuum. The crude product (395 mg, 100% yield, 80% pure) was used for the next step without further purification. 'NMR (300 MHz, CD3OD) δ 8.20 (IH, s), 7.92 (IH, s), 7.27 - 7.47 (5H, m), 6.27
(IH, d, 7 = 7.6 Hz), 4.85 (IH, m), 3.63 (2H, m), 2.67 (3H, s). LCMS (M+H)+ m/z 439 (t = 1.88 min.).
Example 475
Figure imgf000164_0002
(S)-2-[4-(l-HydroxymethyI-2-phenyl-ethylamino)-2-oxo-l,2-dihydro-pyridin-3- yl]-7-methyl-3H-benzimidazole-5-carboxylic acid ethyl ester: The imidate ester (30 mg, 0.067 mmol) was dilute with methanol (10 mL) and two drops of water was added. The reaction mixture was stirred at room temperature for 20 h. After concentration in vacuo, the residue was purified by prep. HPLC to yield the title compound (18.2 mg, 61%) as a white solid. 1H NMR (300 MHz, CD3OD) δ 7.74 (IH, s), 7.11 - 7.30 (7H, m), 6.10 (IH, d, 7 = 6.9 Hz), 4.37 (2H, q, 7 = 6.6 Hz), 4.02 (IH, broad s), 3.69 - 3.81 (2H, m), 3.10 (IH, dd, 7 = 4.8, 13.5 Hz), 2.93 (IH, dd, 7 = 7.8, 13.5 Hz), 2.62 (3H, s), 1.41 (3H, 17 = 6.6 Hz). LCMS (M+H)+ m/z 447 (t = 1.95 min.).
Preparation of Amides Starting from (+)-2-{4-[2-(3-Chloro-phenyl)-2-hydroxy- ethylamino]-2-oxo-l,2-dihydro-pyridin-3-yl}-7-methyl-3H-benzimidazole-5- carboxylic acid, Examples 476 - 504, Scheme IV, 15
Example 476
Figure imgf000165_0001
(±)-2-{4-[2-(3-ChIoro-phenyl)-2-hydroxy-ethylamino]-2-oxo-l,2-dihydro-pyridin- 3-yl}-7-methyl-3H-benzimidazole-5-carboxylic acid methylamide: To a solution of the acid (50 mg, 0.11 mmol) in DMF (5 mL) was added diphenylphosphoryl azide (38 mg, 0.14 mmol). The mixture was allowed to stir for 5 min. Then methylamine (2.0 M solution in THF)(0.11 mL, 0.22 mmol) was added. The mixture was stirred at room temperature for 14 h. After removal of DMF with high vacuum, the residue was purified by prep. HPLC to yield the title compound (18 mg, 36%) as a white solid. 1H NMR (300 MHz, CD3OD) δ 7.89 (IH, s), 7.18 - 7.53 (6H, m), 6.23 (IH, d, 7 = 7.6 Hz), 4.98 (IH, m), 3.70 (IH, dd, 7 = 4.7, 13.5 Hz), 3.61 (IH, dd, 7 = 7.0, 13.5 Hz), 2.94 (3H, s), 2.61 (3H, s). LCMS (M+H)+ m/z 452 (t = 1.57 min.).
Example 477
Figure imgf000166_0001
(±)-2-{4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylanιino]-2-oxo-l,2-dihydro-pyridin- 3-yl}-7-methyl-3H-benzimidazole-5-carboxylic acid dimethylamide: Η NMR (300 MHz, CD3OD) δ 7.52 (IH, s), 7.24 - 7.40 (5H, m), 7.15 (IH, s), 6.25 (IH, d, 7 = 7.6 Hz), 4.98 (IH, dd, 7 = 5.0, 6.9 Hz), 3.70 (IH, dd, 7 = 4.8, 13.5 Hz), 3.62 (IH, dd, 7 = 7.0, 13.5 Hz), 3.3 (3H, s), 3.08 (3H, s), 2.62 (3H, s). LCMS (M+H)+ m/z 466 (t = 1.84 min.).
Example 478
Figure imgf000166_0002
(±)-2-{4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-2-oxo-l,2-dihydro-pyridin- 3-yl}-7-methyl-3H-benzimidazole-5-carboxylic acid tort-butylamide: 1H NMR (300 MHz, CD3OD) δ 7.82 (IH, s), 7.53 (IH, s), 7.48 (IH, s), 7.24 - 7.40 (4H, m), 6.25 (IH, d, 7 = 7.6 Hz), 5.00 (IH, m), 3.64 - 3.70 (2H, m), 2.62 (3H, s), 1.49 (9H, s). LCMS (M+H)+ m/z 494 (t = 1.79 min.). Example 479
Figure imgf000167_0001
(±)-2-{4-[2-(3-Chloro-phenyI)-2-hydroxy-ethylamino]-2-oxo-l,2-dihydro-pyridin- 3-yl}-7-methyl-3H-benzimidazole-5-carboxylic acid butyl-methyl-amide: 1H NMR (300 MHz, CD3OD) δ 7.51 (IH, s), 7.48 (IH, s), 7.24 - 7.39 (4H, m), 7.13 (IH, s), 6.22 (IH, d, 7 = 7.6 Hz), 4.95 (IH, m), 3.55 - 3.70 (3H, m), 3.34 (IH, m), 3.05 (3H, s), 2.62 (3H, s), 0.79 - 1.69 (7H, m). LCMS (M+H)+ m/z 508 (t = 1.88 min.).
Example 480
Figure imgf000167_0002
(±)-2-{4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-2-oxo-l,2-dihydro-pyridin- 3-yI}-7-methyl-3H-benzimidazole-5-carboxylic acid (2-morpholine-4-yl-ethyl)- amide: 1H NMR (300 MHz, CD3OD) δ 7.58 (IH, s), 7.52 (IH, s), 7.05 - 7.42 (5H, m), 6.22 (IH, d, 7 = 7.6 Hz), 4.97 (IH, dd, 7 = 4.9, 6.7 Hz), 2.60 - 4.07 (14H, m), 2.61 (3H, s). LCMS (M+H)+ m/z 551 (t = 1.49 min.).
Example 481
Figure imgf000167_0003
(±)-4-[l-(2-{4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-2-oxo-l,2-dihydro- pyridin-3-yl}-7-methyl-3H-benzimidazol-5-yl)-methanoyl]-piperazine-l- carboxylic acid tert-butyl ester: 1H NMR (300 MHz, CD3OD) δ 7.52 (IH, s), 7.50 (IH, s), 7.24 - 7.38 (4H, m), 7.18 (IH, m), 6.22 (IH, d, 7= 7.6 Hz), 4.97 (IH, m), 3.49 - 3.69 (10H, m), 2.62 (3H, s), 1.47 (9H, s). LCMS (M+H)+ m/z 607 (t = 1.90 min.).
Example 482
Figure imgf000168_0001
(+)-4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-[4-methyl-6-(l-piperazin-l- yl-methanoyl)-lH-benzimidazol-2-yl]-lH-pyridin-2-one: To a solution of (±)-4-[l- (2-{4-[2-(3-chloro-phenyl)-2-hydroxy-ethylamino]-2-oxo-l,2-dihydro-pyridin-3-yl}- 7-methyl-3H-benzimidazol-5-yl)-methanoyl]-piperazine-l-carboxylic acid tert-butyl ester (40 mg, 0.06 mmol) in methanol (5 mL) was added 4.0 M HCl dioxane solution (0.1 mL, excess). The reaction mixture was stirred at room temperature for 14 h. After concentration, the residue was purified by prep. HPLC to give the title compound (16 mg, 63%) as a white foam. 1H NMR (300 MHz, CD3OD) δ 7.56 (IH, s), 7.53 (IH, s), 7.23 - 7.41 (4H, m), 7.17 (IH, m), 6.26 (IH, d, 7 = 7.6 Hz), 4.99 (IH, dd, 7= 4.7, 6.4 Hz), 3.91 (4H, broad s), 3.74 (IH, dd, 7 = 4.7, 13.5 Hz), 3.65 (IH, dd, 7 = 6.4, 13.5 Hz), 3.31 (4H, broad s), 2.63 (3H, s). LCMS (M+H)+ m/z 507 (t = 1.38 min.).
Example 483
Figure imgf000168_0002
(±)-2-{4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-2-oxo-l,2-dihydro-pyridin- 3-yl}-7-methyl-3H-benzimidazole-5-carboxyIic acid cyclopropylamide: 1H NMR (300 MHz, CD3OD) δ 7.80 (IH, s), 7.21 - 7.53 (6H, m), 6.22 (IH, d, 7 = 7.6 Hz), 4.99 (IH, t, 7 = 6.4 Hz), 3.59 - 3.76 (2H, m), 2.83 - 2.90 (IH, m), 2.60 (3H, s), 0.81 - 0.89 (2H, m), 0.64 - 0.73 (2H, m). LCMS (M+H)+ m/z 478 (t = 1.60 min.).
Example 484
Figure imgf000169_0001
(±)-2-{4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-2-oxo-l,2-dihydro-pyridin- 3-yl}-7-methyl-3H-benzimidazole-5-carboxyIic acid cyclopentylamide: 1H NMR
(300 MHz, CD3OD) δ 7.97 (IH, s), 7.16 - 7.60 (6H, m), 6.22 (IH, d, 7 = 7.6 Hz), 4.35 (IH, m), 3.58 - 3.68 (2H, m), 2.62 (3H, s), 1.29 - 2.07 (9H, m). LCMS (M+H)+ m/z 506 (t = 1.86 min.).
Example 485
Figure imgf000169_0002
(±)-4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-[4-methyl-6-(l-piperidin-l- yl-methanoyl)-lH-benzoimidazol-2-yl]-lH-pyridin-2-one: Η NMR (300 MHz, CD3OD) δ 7.53 (IH, s), 7.46 (IH, s), 7.18 - 7.41 (4H, m), 7.07 (IH, s), 6.23 (IH, d, 7 = 7.6 Hz), 4.98 (IH, dd, 7 = 4.8, 6.6 Hz), 3.60 - 3.74 (5H, m), 3.18 - 3.24 (IH, m), 2.60 (3H, m), 1.43 - 1.7 (6H, m). LCMS (M+H)+ m/z 506 (t = 1.78 min.). Example 486
Figure imgf000170_0001
(±)-2-{4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-2-oxo-l,2-dihydro-pyridin- 3-yl}-7-methyl-3H-benzimidazole-5-carboxylic acid cyclohexylamide: 1H NMR (300 MHz, CD3OD) δ 7.88 (IH, s), 7.54 (IH, s), 7.52 (IH, s), 7.16 - 7.40 (4H, m), 6.22 (IH, 7 = 7.6 Hz), 4.97 (IH, dd, 7 = 4.7, 6.9 Hz), 3.89 (H, m), 3.69 (IH, dd, 7 = 4.7, 13.5 Hz), 3.61 (IH, dd, 7= 6.9, 13.5 Hz), 2.61 (3H, s), 1.19 - 2.00 (10H, m). LCMS (M+H)+ m/z 520 (t = 1.93 min.).
Example 487
Figure imgf000170_0002
(±)-4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-{4-methyl-6-[l-(4-methyI- piperidin-l-yl)-methanoyl]-lH-benzimidazoI-2-yl}-lH-pyridin-2-one: Η NMR (300 MHz, CD3OD) δ 7.51 (IH, s), 7.49 (IH, s), 7.21 - 7.39 (4H, m), 7.14 (IH, s), 6.24 (IH, d, 7 = 7.6 Hz), 4.96 (IH, dd, 7 = 4.7, 7.0 Hz), 3.68 (IH, dd, 7 = 4.7, 13. 6 Hz), 3.60 (IH, dd, 7 = 7.0, 13.6 Hz), 2.62 (3H, s), 1.93 - 1.72 (9H, m), 1.00 (3H, d, 7 = 6.3 Hz). LCMS (M+H)+ m/z 520 (t = 1.97 min.). Example 488
Figure imgf000171_0001
(±)-4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-[6-(4,5-dihydro-thiazol-2- yl)-4-methyl-lH-benzimidazol-2-yl]-lH-pyridin-2-one: To a solution of (±)-2-{4- [2-(3-chloro-phenyl)-2-hydroxy-ethylamino]-2-oxo- 1 ,2-dihydro-pyridin-3-yl } -7- methyl-3H-benzimidazole-5-carbonitrile (100 mg, 0.24 mmol) in methanol (20 mL) was added 2-aminoethanethiol hydrochloride (41 mg, 0.36 mmol) and triethylamine (0.1 mL, excess). The reaction mixture was heated to reflux for 14 h and cooled to room temperature. After concentration, the residue was purified by prep. HPLC to yield the title compound (76 mg, 66%) as a yellow solid. 1H NMR (300 MHz, CD3OD) δ 8.10 (IH, s), 7.71 (IH, s), 7.66 (IH, s), 7.25 - 7.53 (4H, m), 6.33 (IH, d, 7 = 7.6 Hz), 5.01 (IH, dd, 7 = 4.2, 7.0 Hz), 4.58 (2H, t, 7 = 8.6 Hz), 3.91 (2H, t, 7 = 8.6 Hz), 3.73 (IH, dd, 7 = 4.2, 13.7 Hz), 3.63 (IH, dd, 7 = 7.0, 13.7 Hz), 2.64 (3H, s). LCMS (M+H)+ m/z 480 (t = 1.70 min.). Example 489
Figure imgf000171_0002
(±)-2-{4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-2-oxo-l,2-dihydro-pyridin- 3-yl}-7-methyl-3H-benzimidazoIe-5-carbaldehyde: To a suspension of (+)-2-{4-[2- (3-chloro-phenyl)-2-hydroxy-ethylamino]-2-oxo-l,2-dihydro-pyridin-3-yl}-7-methyl- 3H-benzimidazole-5-carbonitrile (76 mg, 0.18 mmol) in toluene (anhydrous, 20 mL) was added diisobutylaluminium (1.4 M toluene solution) (0.65 mL, 0.97 mmol) at - 78°C under nitrogen. The mixture was stirred at -78°C for 6 h. Ethyl acetate (1 mL) was then added followed by water (0.5 mL). The mixture was stirred at room temperature for 20 min. The mixture was then passed through a pad of celite and the filtrate was concentrated. The crude product was purified by prep. HPLC to yield the title compound (4 mg, 2.5%) as a brown solid. Η NMR (300 MHz, CD3OD) δ 7.65 (IH, s), 7.57 (IH, s), 7.24 - 7.50 (5H, m), 6.26 (IH, d, 7 = 7.6 Hz), 5.49 (IH, s), 4.96 (IH, m), 3.53 - 3.79 (2H, m), 2.62 (3H, s). LCMS (M+H)+ m/z 423 (t = 1.79 min.).
Example 490
Figure imgf000172_0001
(±)-4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-[6-(l-hydroxy-l-methyl- ethyl)-4-methyI-lH-benzimidazol-2-yl]-lH-pyridin-2-one: To a solution of (±)-2- {4-[2-(3-chloro-phenyl)-2-hydroxy-ethylamino]-2-oxo- 1 ,2-dihydro-pyridin-3-yl } -7- methyl-3H-benzimidazole-5-carboxylic acid ethyl ester (40 mg, 0.08 mmol) in THF (5 mL) was added methyllithium (1.4 M THF solution, 0.57 mL, 0.8 mmol) at -78°C under nitrogen. The reaction mixture was gradually warmed to room temperature overnight. After quenching with water, the mixture was partitioned between ethyl acetate and water. The organic layers were washed with brine, dried over Na SO4, and concentrated in vacuo. The residue was purified by prep HPLC to yield the title compound (13 mg, 36%) as a colorless oil. 1H NMR (300 MHz, CD3OD) δ 7.66 (IH, s), 7.47 (IH, s), 7.26 - 7.41 (5H, m), 6.26 (IH, d, 7 = 7.6 Hz), 4.92 (IH, m), 3.54 - 3.61 (2H, m), 2.62 (3H, s), 1.60 (6H, s). LCMS (M+H)+ m/z 453 (t = 1.46 min.).
Example 491
Figure imgf000172_0002
(±)-3-(6-Aminomethyl-4-methyl-lH-benzimidazol-2-yl)-4-[2-(3-chloro-phenyl)-2- hydroxy-ethylamino]-lH-pyridin-2-one: To a solution of (±)-2-{4-[2-(3-Chloro- phenyl)-2-hydroxy-ethylamino]-2-oxo-l,2-dihydro-pyridin-3-yl}-7-methyl-3H- benzimidazole-5-carboxylic acid amide (20 mg, 0.046 mmol) in THF (1 mL) was added borane-tetrahydrofuran complex (1 M solution) (0.45 mL, 0.45 mmol). The reaction mixture was stirred at room temperature for 10 h and quenched with acetic acid (2 drops). After removal of most solvent, the residue was extracted with EtOAc, washed with brine, dried over Na2SO4. After concentration, the crude product was purified by prep. HPLC to yield the title compound (11.5 mg, 60%). 1H NMR (400 MHz, CD3OD) δ 7.53 (IH, s), 7.50 (IH, s), 7.39 (IH, d, 7 = 7.5 Hz), 7.24 - 7.32 (3H, m), 7.13 (IH, s), 6.26 (IH, d, 7 = 7.5 Hz), 4.99 (IH, dd, 7 = 4.8, 6.8 Hz), 4.19 (2H, s), 3.72 (IH, dd, 7= 4.8, 13.6 Hz), 3.64 (IH, dd, 7 = 6.8, 13.6 Hz), 2.62 (3H, s). LCMS (M+H)+ m/z 424 (t = 2.10 min.).
Example 492
Figure imgf000173_0001
(±)-4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-(6-hydroxymethyl-4-methyl-lH- benzimidazol-2-yl)-lH-pyridin-2-one: To a solution of (±)-2-{4-[2-(3-Chloro-phenyl)-2- hydroxy-ethylamino] -2-oxo- 1 ,2-dihydro-pyridin-3-yl }- -3H-benzimidazole-5-carboxylic acid ethyl ester (25 mg, 0,054 mmol) in methanol (2 mL) and acetic acid (1 mL) was added NaBH4 (10 mg, 0.27 mmol) at -20°C. The reaction mixture was stirred at -20°C for 30 min. and quenched with wopropanol (5 drops). After removal of most solvent, the residue was extracted with EtOAc, washed with water and brine, dried over Na SO4. After concentration, the crude product was purified by prep. HPLC to yield the title compound (18 mg, 62%). Η NMR (300 MHz, CD3OD) δ 7.50 (IH, s), 7.48 (IH, s), 7.25 - 7.38 (4H, m), 7.22 (IH, s), 6.25 (IH, d, 7 = 7.6 Hz), 4.89 - 4.94 (IH, m), 4.76 (2H, s), 3.51 - 3.62 (2H, m), 2.62 (3H, s). LCMS (M+H)+ m/z 425 (t = 1.64 min.).
Figure imgf000174_0001
Figure imgf000175_0002
Example 505
Figure imgf000175_0001
(S)-4-(l-BenzyI-2-hydroxy-ethylamino)-3-(4-methyl-6-phenyl-lH-benzimidazol- 2-yl)-lH-pyridin-2-one: To a solution of (S)-4-(l-benzyl-2-trityloxy-ethylamino)-3- [6-bromo-4-methyl-lH-benzimidazole-2-yl]-lH-pyridin-2-one (50 mg, 0.072 mmol), phenylboronic acid (13 mg, 0.11 mmol), and 2 M K2CO3 (0.108 mL, 0.22 mmol) in THF (5 mL) was added Pd(PPh3)4 (8.3 mg, 0.007 mmol). The mixture was heated to reflux 14 h. Upon cooling, the reaction mixture was diluted with CH C1 , washed with saturated NaHCO3, dried over Na SO4, and concentrated in vacuo. The residue was used for the next step without purification. LCMS (M+H)+ m/z 693 (t = 2.82 min.). The crude product was treated with 4 N HCl dioxane solution (5 mL) at room temperature for 6 h. After concentration in vacuo, the residue was purified by prep. HPLC to yield the title compound (17 mg, 34%) as a white solid. 1H NMR (300 MHz, CD3OD) δ 7.13 - 7. 67 (13H, m), 6.15 (IH, d, 7 = 7.4 Hz), 3.99 - 4.11 (IH, m), 3.74 - 3.77 (2H, m), 3.16 (IH, dd, 7 = 5.4, 13.6 Hz), 2.97 (IH, dd, 7 = 7.8, 13.6 Hz), 2.69 (3H, s). LCMS (M+H)+ m/z 451 (t = 2.04 min.).
Example 506
Figure imgf000176_0001
(S)-4-(l-Benzyl-2-hydroxy-ethylamino)-3-(4-methyl-lH-benzimidazol-2-yI)-lH- pyridin-2-one: 1H NMR (300 MHz, CD3OD) δ 6.98 - 7.46 (9H, m), 6.15 (IH, d, 7 = 7.5 Hz), 3.99 - 4.04 (IH, m), 3.75 (2H, d, 7 = 5.1 Hz), 3.16 (IH, dd, 7 = 5.1, 13.6 Hz), 2.95 (IH, dd, 7 = 8.1, 13.6 Hz), 2.61 (3H, s). LCMS (M+H)+ m/z 375 (t = 1.72 min.).
Example 507
Figure imgf000176_0002
(S)-4-(l-Benzyl-2-hydroxy-ethylamino)-3-[6-(2-methoxy-phenyl)-4-methyl-lH- benzimidazol-2-yl]-lH-pyridin-2-one: Η NMR (300 MHz, CD3OD) δ 6.99 - 7.59 (12H, m), 6.16 (IH, d, 7 =7.5 Hz), 4.04 (IH, m), 3.81 (3H, s), 3.76 (2H, d, 7 = 4.1 Hz), 2.96 - 3.20 (2H, m), 2.66 (3H, s). LCMS (M+H)+ m/z 481 (t = 2.00 min.). Example 508
Figure imgf000177_0001
(S)-4-(l-Benzyl-2-hydroxy-ethylamino)-3-[6-(4-fluoro-phenyl)-4-methyl-lH- benzimidazol-2-yl]-lH-pyridin-2-one: 1H NMR (300 MHz, CD3OD) δ 7.10 - 7.66 (12H, m), 6.13 (IH, d, 7 = 7.6 Hz), 3.99 - 4.04 (IH, m), 3.76 (2H, d, 7= 5.0 Hz), 3.16 (IH, dd, 7 = 5.0, 13.6 Hz), 2.96 (IH, dd, 7 = 8.1, 13.6 Hz), 2.65 (3H, s). LCMS (M+H)+ m/z 469 (t = 2.07 min.).
Example 509
Figure imgf000177_0002
(S)-4-(l-Benzyl-2-hydroxy-ethylamino)-3-[6-(4-methoxy-phenyl)-4-methyl-lH- benzimidazol-2-yl]-lH-pyridin-2-one: 1H NMR (300 MHz, CD3OD) δ 6.97 - 7.65 (12H, m), 6.14 (IH, d, 7 = 7.6 Hz), 4.00 - 4.04 (IH, m), 3.83 (3H, s), 3.76 (2H, d, 7 = 5.0 Hz), 2.95 - 3.19 (2H, m), 2.67 (3H, s). LCMS (M+H)+ m/z 481 (t = 2.01 min.).
General Procedure for Buchwald couplings, Examples 510 - 516 Scheme V, 19 Example 510
Figure imgf000177_0003
(S)-4-(l-Benzyl-2-hydroxy-ethylamino)-3-(4-methyl-6-morphoIin-4-yl-lH- benzimidazol-2-yl)-lH-pyridin-2-one: A mixture of (S)-4-(l-benzyl-2-trityloxy- ethylamino)-3-[6-bromo-4-methyl-lH-benzimidazol-2-yl]-lH-pyridin-2-one (150 mg, 0.216 mmol), morpholine (28.2 mg, 0.324 mmol), palladium acetate (2.4 mg, 0.01 mmol), tri-tert-buylphosphine (4.4 mg. 0.02 mmol), and sodium tert-butoxide (104 mg, 1.08 mmol) in toluene (5 mL) was heated to 100°C for 14 h under nitrogen. The reaction mixture was cooled to room temperature and diluted with EtOAc. After extraction, the combined organic layers were washed with water, brine, dried over Na2SO4. Concentration gave a brownish residue, which was treated with 4 N HCl dioxane solution (3 mL) at room temperature for 6 h. After removal of the solvent, the residue was purified by prep. HPLC to yield the title compound (18 mg, 18%). 1H NMR (400 MHz, CD3OD) δ 7.59 (IH, s), 7.12 - 7.28 (7H, m), 6.12 (IH, d, 7 = 7.6 Hz), 4.01 - 4.08 (5H, m), 3.76 (IH, dd, 7 = 4.8, 11.1 Hz), 3.71 (IH, dd, 7 = 5.2, 11.1 Hz), 3.32 - 3.10 (4H, m), 3.09 (IH, dd, 7 = 5.6, 13.7 Hz), 2.92 (IH, dd, 7 = 8.0, 13.7 Hz), 2.65 (3H, s). LCMS (M+H)+ m/z 460 (t = 1.30 min.).
Example 511
Figure imgf000178_0001
(±)-4-[2-(3-ChIoro-phenyl)-2-hydroxy-ethylamino]-3-(4-methyl-lH- benzimidazol-2-yl)-lH-pyridin-2-one: Η NMR (300 MHz, CD3OD) δ 7.17 - 7.64 (8H, m), 6.23 (IH, d, 7 = 10.2 Hz), 4.94 (IH, m), 3.61 (IH, dd, 7= 4.8, 13.8 Hz), 3.54 (IH, dd, 7 = 7.4, 13.8 Hz), 2.61 (3H, s). LCMS (M+H)+ m/z 395 (t = 1.65 min.). Example 512
Figure imgf000179_0001
(+)-4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-(4-methyl-6-pyridin-3-yl- lH-benzimidazol-2-yl)-lH-pyridin-2-one: Η NMR (300 MHz, CD3OD) δ 9.14 (IH, s), 8.88 (IH, d, 7 = 8.2 Hz), 8.72 (IH, d, 7 = 5.4 Hz), 8.08 (IH, dd, 7 = 5.8, 8.2 Hz), 7.82 (IH, s), 7.56 (IH, s), 7.24 - 7.45 (5H, m), 6.26 (IH, d, 7 = 7.6 Hz), 5.02 (IH, dd, 7 = 5.0, 6.5 Hz), 3.77 (IH, dd, 7 = 5.0, 13.5 Hz), 3.68 (IH, dd, 7 = 6.5, 13.5 Hz), 2.68 (3H, s). LCMS (M+H)+ m/z 472 (t = 1.66 min.).
Figure imgf000179_0002
The following examples 517-519 were prepared according to Schemes VII and III.
Example 517
Figure imgf000180_0001
(S)-4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-(4-methyl-6-piperazin-l-yI- lH-benzoimidazol-2-yl)-lH-pyridin-2-one: 1H NMR (500 MHz, CD3OD) δ 7.54 (brs, IH), 7.40 - 7.20 (m, 4H), 7.03 (brs, IH), 6.84 (brs, IH), 6.25 (d, IH, J = 7.60 Hz), 5.01 - 4.91 (m, IH), 3.73 (dd, IH), 3.65 (dd, IH), 3.45 - 3.25 (m, 8H), 2.56 (s, 3H); LCMS (M+H)+ m/z 479,481.
Example 518
Figure imgf000180_0002
(±)-4-[2-(3-Chloro-4-methylsulfanyl-phenyl)-2-hydroxy-ethylamino]-3-(4-methyI- 6-piperazin-l-yl-lH-benzoimidazol-2-yl)-lH-pyridin-2-one: Η NMR (500 MHz, CD3OD) δ 7.47 (brs, IH), 7.38 - 7.29 (m, 2H), 7.19 (d, IH, J = 8.3 Hz), 7.03 (brs, IH), 6.98 (brs, IH), 6.26 (d, IH, J = 7.7 Hz), 4.90 - 4.81 (m, IH), 3.65 - 3.35 (m, 10H), 2.56 (s, 3H), 2.42 (s, 3H); LCMS (M+H)+ m/z 525, 527. Example 519
Figure imgf000181_0001
(S)-4-[2-(3-Chloro-4-fluoro-phenyl)-2-hydroxy-ethylamino]-3-(4-methyl-6- piperazin-l-yl-lH-benzoimidazol-2-yl)-lH-pyridin-2-one: 1H NMR (500 MHz, CD3OD) δ 7.61 (dd, IH, J = 2.1, 7.2 Hz), 7.40 (ddd, IH), 7.28 (d, IH, J = 7.5 Hz), 7.17 (dd, IH, J = 8.9, 8.8 Hz), 7.02 (brs, IH), 6.87 (brs, IH), 6.25 (d, IH, J = 7.6 Hz), 4.99 - 4.90 (m, IH), 3.73 - 3.60 (m, 2H), 3.45 - 3.30 (m, 8H), 2.54 (s, 3H); LCMS (M+H)+ m/z 497, 499.
The following examples (520-522) were prepared according to Scheme VII and III and illustrate the alkylation of a piperazine derivative
Example 520 (General procedure for examples 520-522)
Figure imgf000181_0002
(S)-3-[4-(2-{4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-2-oxo-l,2-dihydro- pyridin-3-yl}-7-methyl-3H-benzoimidazol-5-yI)-piperazin-l-yl]-propionitrile: To a stirred solution of 4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-(4-methyl-6- piperazin-l-yl-lH-benzoimidazol-2-yl)-lH-pyridin-2-one (17 mg, 0.021 mmol) in anhydrous methanol (1 mL) was added Hunigs base (36 μL). The resulting solution was cooled to 0 °C and acrylonitrile (5 μL) was added in portions until the reaction was complete as judged by LCMS. The reaction was warmed to room temperature and the solvent evaporated in vacuo. The resulting residue was purified on reverse phase preparative HPLC using a methanol / water / 0.1% trifluoroacetic acid gradient. The fractions were evaporated to give the title compound as a trifluoroacetic acid salt (12.1 mg): 1H NMR (500 MHz, CD3OD) δ 7.49 (brs, IH), 7.39 - 7.23 (m, 4H), 7.15 (brs, IH), 7.06 (brs, IH), 6.25 (d, IH, J = 7.7 Hz), 4.97 - 4.88 (m, IH), 3.70 - 3.40 (m, 12 H), 3.06 (t, 2H, J = 7.0 Hz), 2.59 (s, 3H); LCMS (M+H)+ m/z 532,534. The trifluoroaceticacid salt of the pure title compound was dissolved in methanol and applied to a Varian Mega Bond-Elute SCX cartridge. Elution with methanol followed by 2.0 M NH3 / MeOH gave the free base. This material was suspended in MeOH and 1.00 N aqueous HCl (2 equiv.) was added. The resulting solution was filtered through a 45 μm filter and evaporated to give the bis HCl salt of the title compound: 1H NMR (300 MHz, CD3OD) δ 7.50 - 7.25 (m, 5H), 7.16 (brs, IH), 7.13 (brs, IH), 6.27 (d, IH, J = 7.7 Hz), 4.95 - 4.87 (m, IH), 3.70 - 3.40 (m, 12H), 3.13 (t, 2H, J = 7.0 Hz), 2.62 (s, 3H); LCMS (M+H)+ m/z 532,534. Example 521
Figure imgf000182_0001
(S)-4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-{6-[4-(2-methanesulfonyl- ethyl)-piperazin-l-yl]-4-methyl-lH-benzoimidazol-2-yl}-lH-pyridin-2-one: Η NMR (500 MHz, CD3OD) δ 7.53 (brs, IH), 7.41 - 7.20 (m, 4H), 7.13 (brs, IH), 6.92 (brs, IH), 6.24 (d, IH, J = 7.6 Hz), 5.00 - 4.92 (m, IH), 3.80 - 3.25 (m, 14H), 3.13 (s, 3H), 2.57 (s, 3H); LCMS (M+H)+ m/z 585, 587.
J 81 Example 522
Figure imgf000183_0001
(S)-3-[4-(2-{4-[2-(3-Bromo-4-methoxy-phenyI)-2-hydroxy-ethylamino]-2-oxo-l,2- dihydro-pyridin-3-yl}-7-methyI-3H-benzoimidazol-5-yl)-piperazin-l-yl]- propionitrile: Η NMR (500 MHz, CD3OD) δ 7.59 (brs, IH), 7.41 (d, IH, J = 7.5 Hz), 7.34 (dd, IH, J = 2.0, 8.5 Hz), 7.17 (brs, IH), 7.12 (brs, IH), 7.00 (d, IH, J = 8.5 Hz), 6.25 (d, IH, J = 7.5 Hz), 4.85 - 4.76 (m, IH), 3.85 (s, 3H), 3.80 - 3.30 (m, 12H), 3.16 (t, 2H, J = 7.0 Hz), 2.55 (s, 3H); LCMS (M+H)+ m/z 606, 608.
The following examples (523-528) were prepared according to Scheme VII and III and illustrate the carbamoylation of a piperazine derivative
Example 523 (General procedure for examples 523-528)
Figure imgf000183_0002
(S)-4-(2-{4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-2-oxo-l,2-dihydro- pyridin-3-yl}-7-methyl-3H-benzoimidazol-5-yl)-piperazine-l-carboxylic acid 2- fluoro-ethyl ester: To a stirred solution of 4-[2-(3-Chloro-phenyl)-2-hydroxy- ethylamino]-3-(4-methyl-6-piperazin-l-yl-lH-benzoimidazol-2-yl)-lH-pyridin-2-one (~ 2 TFA salt, 80 mg, ~0.1 mmol) in methanol (2 mL) at 0 °C was added N,N- diisopropylethylamine (170 μL) and 2-fluoroethyl chloroformate (37 mg). The cooling bath was removed and the solution was stirred at room temperature for 30 minutes, after which LC/MS analysis showed the reaction to be complete. The reaction mixture was then purified on reverse phase preparative HPLC using a methanol / water / 0.1% trifluoroacetic acid gradient. The fractions were evaporated to give the title compound as a trifluoroacetic acid salt, which was dissolved in methanol and applied to a Varian Mega Bond-Elute SCX cartridge. Elution with methanol followed by 2.0 M NH3 / MeOH gave the free base (46.2 mg). This material was suspended in MeOH and 1.00 N aqueous HCl (1 equiv.) was added. The resulting solution was filtered through a 45 μm filter and evaporated to give the mono HCl salt of the title compound (46 mg): 1H NMR (500 MHz, CD3OD) δ 7.50 (brs, IH), 7.45 - 7.20 (m, 6H), 6.26 (d, IH, J = 7.7 Hz), 4.98 - 4.91 (m, IH), 4.64 (dm, 2H, J = 47.9 Hz), 4.39 (dm, 2H, J = 29 Hz), 3.95 - 3.50 (m, 10H), 2.63 (s, 3H); . LCMS (M+H)+ m/z 569,571.
Example 524
Figure imgf000184_0001
(S)-4-(2-{4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-2-oxo-l,2-dihydro- pyridin-3-yl}-7-methyl-3H-benzoimidazol-5-yl)-piperazine-l-carboxylic acid 2- methoxy-ethyl ester: 1H NMR (500 MHz, CD3OD) δ 7.53 - 7.22 (m, 7H), 6.26 (d, IH, J = 7.6 Hz), 4.96 (dd, IH, J = 7.0, 4.6 Hz), 4.31 - 4.27 (m, 2H), 4.05 - 3.55 (m, 12H), 3.39 (s, 3H), 2.64 (s, 3H); LCMS (M+H)+ m/z 581, 583. Example 525
Figure imgf000185_0001
(S)-4-(2-{4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-2-oxo-l,2-dihydro- pyridin-3-yl}-7-methyl-3H-benzoimidazol-5-yl)-piperazine-l-carboxylic acid tert- butyl ester: Η NMR (500 MHz, CD3OD) δ 7.51 (brs, IH), 7.40 - 7.22 (m, 6H), 6.26 (d, IH, J = 7.7 Hz), 4.96 - 4.90 (m, IH), 3.90 - 3.30 (m, 10H), 2.64 (s, 3H), 1.51 (s, 9H); LCMS (M+H)+ m/z 579, 581.
Example 526
Figure imgf000185_0002
(S)-4-(2-{4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-2-oxo-l,2-dihydro- pyridin-3-yl}-7-methyI-3H-benzoimidazol-5-yl)-piperazine-l-carboxylic acid prop-2-ynyl ester: 1H NMR (500 MHz, CD3OD) δ 7.50 (brs, IH), 7.43 - 7.20 (m, 5H), 7.19 (brs, IH), 6.25 (d, IH, J = 7.6 Hz), 4.98 - 4.90 (m, IH), 4.78 (d, 2H, J = 2.5 Hz), 3.95 - 3.30 (m, 10H), 2.97 (t, IH, J = 2.5 Hz), 2.62 (s, 3H); LCMS (M+H)+ m/z 561, 563.
Example 527
Figure imgf000186_0001
(S)-4-(2-{4-[2-(3-Bromo-4-methoxy-phenyl)-2-hydroxy-ethylamino]-2-oxo-l,2- dihydro-pyridin-3-yl}-7-methyl-3H-benzoimidazol-5-yl)-piperazine-l-carboxylic acid tert-butyl ester: 1H NMR (300 MHz, CD3OD) δ 7.65 (brs, IH), 7.48 - 7.28 (m, 3H), 7.25 (brs, IH), 6.97 (d, IH, J = 8.5 Hz), 6.25 (d, IH, J = 7.7 Hz), 4.94 - 4.86 (m, IH), 3.90 - 3.45 (m, 10H), 3.82 (s, 3H), 2.61 (s, 3H), 1.52 (s, 9H); LCMS (M+H)+ m/z 653, 655.
Example 528
Figure imgf000186_0002
(S)-4-(2-{4-[2-(3-Bromo-4-methoxy-phenyl)-2-hydroxy-ethylamino]-2-oxo-l,2- dihydro-pyridin-3-yl}-7-methyI-3H-benzimidazol-5-yl)-piperazine-l-carboxylic acid ethyl ester: 1H NMR (400 MHz, CD3OD) δ 7.80 (IH, s), 7.63 (IH, narrow d, 7 = 1.8 Hz), 7.52 (IH, s), 7.42 (IH, d, 7 = 7.5 Hz), 7.37 (IH, dd, 7 = 1.8, 8.4 Hz), 7.00 (IH, d, 7 = 8.4 Hz), 6.31 (IH, d, 7 = 7.5 Hz), 4.89 (IH, m), 4.22 (2H, q, 7 = 7.1 Hz), 3.98 (4H, br s), 3.84 (3H, s), 3.70 - 3.72 (4H, m), 3.59 - 3.60 (2H, m), 2.67 (3H, s), 1.31 (3H, t, 7 = 7.1 Hz). LCMS (M+H)+ m z 625 (t = 1.45 min.). The following examples (529-540) were prepared according to Scheme VII and III and illustrate an alternative method of alkylation of a piperazine derivative
Example 529 (General procedure for examples 529-540)
Figure imgf000187_0001
(S)-4-[2-(3-ChIoro-4-methoxy-phenyI)-2-hydroxy-ethylamino]-3-{6-[4-(3-fluoro- propyl)-piperazin-l-yl]-4-methyl-lH-benzoimidazol-2-yl}-lH-pyridin-2-one: To a stirred solution of 4-[2-(3-Chloro-4-methoxy-phenyl)-2-hydroxy-ethylamino]-3-(4- methyl-6-piperazin-l-yl-lH-benzoimidazol-2-yl)-lH-pyridin-2-one (100 mg, 0.162 mmol) in 1,4-dioxane (4.0 mL), ethanol (0.8 mL), methanol (0.8 mL) was added N,N- diisopropylethylamine (0.30 mL) and l-bromo-3-fluoropropane (85 μl). The reaction was heated at 80 °C for 12 h. The reaction mixture was then purified on reverse phase preparative HPLC using a methanol / water / 0.1% trifluoroacetic acid gradient. The fractions were evaporated to give the title compound as a trifluoroacetic acid salt, which was dissolved in methanol and applied to a Varian Mega Bond-Elute SCX cartridge. Elution with methanol followed by 2.0 M NH3 / MeOH gave the free base (39.3 mg). This material was suspended in MeOH and 1.00 N aqueous HCl (2 equiv.) was added. The resulting solution was filtered through a 45 μm filter and evaporated to give the bis HCl salt of the title compound (43.5 mg): 1H NMR (400 MHz, CD3OD) δ 7.43 - 7.37 (m, 2H), 7.28 (dd, IH, J = 2.0, 8.6 Hz), 7.16 (brs, IH), 7.08 (d, IH, J = 1.7 Hz), 7.01 (d, IH, J = 8.5 Hz), 6.24 (d, IH, J = 7.7 Hz), 4.84 - 4.78 (m, IH), 4.60 (dt, 2H, J = 5.7, 47.1 Hz), 3.95 - 3.88 (m, 2H), 3.83 (s, 3H), 3.78 - 3.71 (m, 2H), 3.53 - 3.14 (m, 8H), 2.59 (s, 3H), 2.31 - 2.16 (m, 2H); LCMS (M+H)+ m/z 569, 571. Example 530
Figure imgf000188_0001
(S)-4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-{6-[4-(2-fluoro-ethyl)- piperazin-l-yl]-4-methyl-lH-benzoimidazol-2-yl}-lH-pyridin-2-one: 1H NMR (400 MHz, CD3OD) δ 7.45 - 7.20 (m, 5H), 7.16 (brs, IH), 7.09 (brs, IH), 6.25 (d, IH, J = 7.6 Hz), 5.00 - 4.92 (m, IH), 4.92 - 4.78 (m, 2H), 3.98 - 3.15 (m, 12H), 2.60 (s, 3H); LCMS (M+H)+ m/z 525, 527.
Example 531
Figure imgf000188_0002
(S)-4-[2-(3-Chloro-4-fluoro-phenyl)-2-hydroxy-ethylamino]-3-{6-[4-(3-fluoro- propyl)-piperazin-l-yI]-4-methyl-lH-benzoimidazol-2-yl}-lH-pyridin-2-one: 1H NMR (400 MHz, CD3OD) δ 7.52 (dd, IH, J = 2.1, 7.2 Hz), 7.40 (d, IH, J = 7.6 Hz), 7.35 - 7.12 (m, 3H), 7.08 (d, IH, J = 1.7Hz), 6.25 (d, IH, J = 7.7Hz), 4.90 - 4.82 (m, IH), 4.60 (dt, 2H, J = 5.4, 47.1 Hz), 3.96 - 3.10 (m, 12H), 2.60 (s, 3H), 2.28 - 2.13 (m, 2H); LCMS (M+H)+ m/z 557, 559. Example 532
Figure imgf000189_0001
(S)-4-[2-(3-Bromo-4-methoxy-phenyl)-2-hydroxy-ethylamino]-3-{6-[4-(3-fluoro- propyl)-piperazin-l-yl]-4-methyl-lH-benzoimidazol-2-yl}-lH-pyridin-2-one: 1H
NMR (400 MHz, CD3OD) δ 7.58 (d, IH, J = 2.0 Hz), 7.43 (d, IH, J = 7.6 Hz), 7.34 (dd, IH, J = 2.0, 8.5 Hz), 7.19 (brs, IH), 7.11 (brs, IH), 7.00 (d, IH, J = 8.5 Hz), 6.26 (d, IH, J = 7.7 Hz), 5.90 - 4.82 (m, IH), 4.61 (dt, 2H, J = 5.4, 47.1 Hz), 3.95 - 3.12 (m, 12H), 3.85 (s, 3H), 2.62 (s, 3H), 2.30 - 2.14 (m, 2H); LCMS (M+H)+ m/z 613, 615.
Example 533
Figure imgf000189_0002
(S)-4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-{4-methyI-6-[4-(3,3,3- trifluoro-propyI)-piperazin-l-yI]-lH-benzoimidazol-2-yl}-lH-pyridin-2-one: Η
NMR (300 MHz, CD3OD) δ 7.50 - 7.10 (m, 7H), 6.26 (d, IH, J = 7.5 Hz), 4.96 - 4.88 (m, IH), 4.00 - 3.15 (m, 12H), 3.00 - 2.82 (m, 2H), 2.61 (s, 3H); LCMS (M+H)+ m/z 575, 577. Example 534
Figure imgf000190_0001
(5)-4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-{6-[4-(3-fluoro-propyl)- piperazin-l-yl]-4-methyl-lH-benzoimidazol-2-yI}-lH-pyridin-2-one: 1H NMR (300 MHz, CD3OD) δ 7.46 - 7.25 (m, 5H), 7.19 (brs, IH), 7.11 (brs, IH), 6.27 (d, IH, J = 7.7 Hz), 4.95 - 4.86 (m, IH), 4.62 (dt, 2H, J = 5.4, 47.1 Hz), 3.98 - 3.15 (m, 12H), 2.62 (s, 3H), 2.35 - 2.12 (m, 2H); LCMS (M+H)+ m/z 539, 541.
Example 535
Figure imgf000190_0002
(S)-4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-{4-methyl-6-[4-(3,4,4- trifluoro-but-3-enyl)-piperazin-l-yl]-lH-benzoimidazol-2-yl}-lH-pyridin-2-one:
LCMS (M+H)+ m/z 587, 589.
Example 536
Figure imgf000190_0003
4-[2-(3-Chloro-phenyl)-2(S)-hydroxy-ethylamino]-3-{6-[4-(3-fluoro-2-hydroxy- propyI)-piperazin-l-yl]-4-methyl-lH-benzoimidazol-2-yl}-lH-pyridin-2-one:
LCMS (M+H)+ m z 555, 557.
Example 537
Figure imgf000191_0001
(S)-4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-{6-[4-(2-hydroxy-2-methyl- propyl)-piperazin-l-yl]-4-methyl-lH-benzoimidazol-2-yI}-lH-pyridin-2-one: 1H
NMR (400 MHz, CD3OD) δ 7.44 - 7.22 (m, 5H), 7.15 (brs, IH), 7.08 (brs, IH), 6.25 (d, IH, J = 7.6 Hz), 4.88 - 4.80 (m, IH), 3.86 - 3.81 (m, 4H), 3.55 - 3.32 (m, 6H), 3.28 (s, 2H), 2.60 (s, 3H), 1.38 (s, 6H); LCMS (M+H)+ m/z 551, 553.
Example 538
Figure imgf000191_0002
(S)-4-[2-(3-Chloro-phenyI)-2-hydroxy-ethylamino]-3-{6-[4-(2-hydroxy-ethyl)- piperazin-l-yl]-4-methyl-lH-benzimidazoI-2-yl}-lH-pyridin-2-one: ΗNMR (400 MHz, CD3OD) δ 7.50 (IH, s), 7.25 - 7.35 (4H, m), 7.04 (IH, s), 6.97 (IH, s), 6.22 (IH, d, 7 = 6.8 Hz), 4.93 - 4.95 (IH, m), 3.21 - 3.96 (14H, m), 2.57 (3H, s). LCMS (M+H)+ m/z 523 (t = 1.11 min.). Example 539
Figure imgf000192_0001
(S)-4-[2-(3-Bromo-4-methoxy-phenyl)-2-hydroxy-ethyIamino]-3-{6-[4-(2- hydroxy-ethyl)-piperazin-l-yl]-4-methyl-lH-benzimidazol-2-yl}-lH-pyridin-2- one: 1H NMR (400 MHz, CD3OD) δ 7.60 (IH, narrow d, 7 = 2.0 Hz), 7.40 (IH, d, 7 = 7.6 Hz), 7.35 (IH, dd, 7 = 2.0, 8.4 Hz), 7.13 (IH, s), 7.09 (IH, s), 6.99 (IH, d, 7 = 8.4 Hz), 6.25 (IH, d, 7 = 7.6 Hz), 4.82 - 4.87 (IH, m), 3.76 - 3.97 (6H, m), 3.84 (3H, s), 3.24 - 3.52 (8H, m), 2.60 (3H, s). LCMS (M+H)+ m/z 597 (t = 1.09 min.).
Example 540
Figure imgf000192_0002
(S)-[4-(2-{4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-2-oxo-l,2-dihydro- pyridin-3-yl}-7-methyl-3H-benzoimidazol-5-yl)-piperazin-l-yl]-acetonitrile: 1H NMR (400 MHz, CD3OD) δ 7.50 - 7.25 (m, 7H), 6.28 (d, IH, J = 8.0 Hz), 4.95 - 4.88 (m, IH), 4.32 (s, 2H), 3.75 - 3.62 (m, 4H), 3.60 - 3.35 (m, 6H), 2.63 (s, 3H); LCMS (M+H)+ m/z 518, 520.
The following examples (541-553) were prepared according to Scheme VII and III and illustrate the acylation of a piperazine derivative Example 541 (General procedure for examples 541-553)
Figure imgf000193_0001
(S)-4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-{6-[4-(4-fluoro-butyryl)- piperazin-l-yl]-4-methyl-lH-benzoimidazol-2-yI}-lH-pyridin-2-one: To a stirred solution of 4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-(4-methyl-6-piperazin- l-yl-lH-benzoimidazol-2-yl)-lH-pyridin-2-one (70 mg, 0.136 mmol) in anhydrous N,N-dimethylformamide (750 μl) was added l-(3-dimethylaminopropyl)-3- ethylcarbodiimide hydrochloride (112 mg, 0.584 mmol), l-hydroxybenzotriazole hydrate (59 mg, 0.438 mmol), N-methylmorpholine (0.048 mL, 0.438 mmol) and 4- fluorobutyric acid (31 mg, 0.291 mmol; see O'Hagan,D., J. Fluorine Chem., 43, (1989), 371-377) and the mixture heated to 80 °C for 3 h. The reaction mixture was then purified on reverse phase preparative HPLC using a methanol / water 1 .1% trifluoroacetic acid gradient. The fractions were evaporated to give the title compound as a trifluoroacetic acid salt, which was dissolved in methanol and applied to a Varian Mega Bond-Elute SCX cartridge. Elution with methanol followed by 2.0 M NH3 / MeOH gave the free base (35.9 mg). This material was suspended in MeOH and 1.00 N aqueous HCl (2 equiv.) was added. The resulting solution was filtered through a 45 μm filter and evaporated to give the bis HCl salt of the title compound (37.6 mg): 1H NMR (400 MHz, CD3OD) δ. 7.61 (brs, IH), 7.47 (brs, IH), 7.44 - 7.20 (m, 5H), 6.27 (d, IH, J = 7.6 Hz), 4.92 (dd, IH, J = 4.4, 7.3 Hz), 4.50 (dt, 2H, J = 5,9, 47.3 Hz), 4.05 - 3.40 (m, 10H), 2.64 (s, 3H), 2.63 (t, 2H, J = 7.6 Hz), 2.09 - 1.95 (m, 2H); LCMS (M+H)+ m/z 567,569. Example 542
Figure imgf000194_0001
(5)-4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-{6-[4-(2,2-difluoro-acetyl)- piperazin-l-yl]-4-methyl-lH-benzoimidazol-2-yl}-lH-pyridin-2-one: LCMS
(M+H)+ m/z 557, 559.
Example 543
Figure imgf000194_0002
(5)-4-[2-(3-ChIoro-phenyl)-2-hydroxy-ethylamino]-3-{6-[4-(2-methanesulfonyl- acetyl)-piperazin-l-yl]-4-methyl-lH-benzoimidazol-2-yl}-lH-pyridin-2-one:
LCMS (M+H)+ m/z 599, 601.
Example 544
Figure imgf000194_0003
(S)-3-[6-(4-AcetyI-piperazin-l-yl)-4-methyl-lH-benzoimidazol-2-yl]-4-[2-(3- chloro-phenyl)-2-hydroxy-ethylamino]-lH-pyridin-2-one: Η NMR (500 MHz, CD3OD) δ 7.57 (brs, IH), 7.49 (brs, IH), 7.40 - 7.20 (m, 5H), 6.29 (d, IH, J = 7.6 Hz), 4.98 - 4.90 (m, IH), 4.02 - 3.91 (m, 4H), 3.70 - 3.50 (m, 6H), 2.66 (s, 3H), 2.21 (s, 3H); LCMS (M+H)+ m/z 521, 523.
Example 545
Figure imgf000195_0001
(S)-4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-(4-methyl-6-{4-[2-(l-oxo- 114-thiomorpholin-4-yl)-acetyl]-piperazin-l-yl}-lH-benzoimidazol-2-yl)-lH- pyridin-2-one: LCMS (M+H)+ m/z 638, 640.
Example 546
Figure imgf000195_0002
(5)-4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-(6-{4-[2-(l,l-dioxo-1 6- thiomorpholin-4-yI)-acetyl]-piperazin-l-yl}-4-methyl-lH-benzoimidazol-2-yl)- lH-pyridin-2-one: LCMS (M+H)+ m/z 654, 656.
Example 547
Figure imgf000196_0001
(S)-4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-{4-methyl-6-[4-(2- thiomorpholin-4-yl-acetyl)-piperazin-l-yl]-lH-benzoimidazol-2-yl}-lH-pyridin- 2-one: LCMS (M+H)+ m/z 622, 624.
Example 548
Figure imgf000196_0002
4-[2-(3-Chloro-phenyl)-2(5)-hydroxy-ethylamino]-3-{6-[4-(2-methanesulfinyl- acetyl)-piperazin-l-yl]-4-methyl-lH-benzoimidazol-2-yI}-lH-pyridin-2-one:
LCMS (M+H)+ m/z 583, 585.
Example 549
Figure imgf000196_0003
(S)-4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-{6-[4-(2-methoxy-acetyl)- piperazin-l-yl]-4-methyl-lH-benzoimidazol-2-yl}-lH-pyridin-2-one: 1H MR (500 MHz, CD3OD) δ 7.49 (brs, IH), 7.40 - 7.18 (m, 6H), 6.26 (d, IH, J = 7.7 Hz), 4.98 - 4.90 (m, IH), 4.25 (s, 2H), 3.95 - 3.46 (m, 10H), 3.44 (s, 3H), 2.63 (s, 3H); LCMS (M+H)+ m/z 551, 553.
Example 550
Figure imgf000197_0001
(S)-4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-{4-methyl-6-[4-(2- methylsulfanyl-acetyl)-piperazin-l-yl]-lH-benzoimidazol-2-yl}-lH-pyridin-2- one: ]H NMR (500 MHz, CD3OD) δ 7.49 (brs, IH), 7.45 - 7.20 (m, 6H), 6.26 (d, IH, J = 7.60 Hz), 4.98 - 4.90 (m, IH), 3.97 - 3.40 (m, 12H), 2.63 (s, 3H), 2.20 (s, 3H); LCMS (M+H)+ m/z 567, 569.
Example 551
Figure imgf000197_0002
(S)-3-{6-[4-(2-Chloro-acetyl)-piperazin-l-yl]-4-methyl-lH-benzoimidazol-2-yl}-4- [2-(3-chloro-phenyl)-2-hydroxy-ethyIamino]-lH-pyridin-2-one: Η NMR (500 MHz, CD3OD) δ 7.49 (brs, IH), 7.38 - 7.20 (m, 5H), 7.15 (brs, IH), 6.25 (d, IH, J = 7.6 Hz), 4.98 - 4.90 (m, IH), 4.35 (s, 2H), 3.90 - 3.80 (m, 4H), 3.66 - 3.30 (m, 6H), 2.61 (s, 3H); LCMS (M+H)+ m/z 555, 557. Example 552
Figure imgf000198_0001
(S)-4-(2-{4-[2-(3-Bromo-4-methoxy-phenyl)-2-hydroxy-ethylamino]-2-oxo-l,2- dihydro-pyridin-3-yl}-7-methyl-3H-benzimidazol-5-yl)-piperazine-l- carbaldehyde: 1H NMR (400 MHz, CD3OD) δ 8.18 (IH, s), 7.65 (IH, s), 7.61 (IH, narrow d, 7 = 2.0 Hz), 7.46 (IH, s), 7.43 (IH, d, 7 = 7.6 Hz), 7.36 (IH, dd, 7 = 2.0, 8.5 Hz), 7.00 (IH, d, 7 = 8.5 Hz), 6.32 (IH, d, 7 = 7.6 Hz), 4.86 - 4.89 (IH, m), 3.91 - 3.96 (4H, m), 3.84 (3H, s), 3.57 - 3.67 (7H, m), 2.66 (3H, s). LCMS (M+H)+ m/z 581 (t = 1.24 min.).
Example 553
Figure imgf000198_0002
(S)-4-(2-{4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-2-oxo-l,2-dihydro- pyridin-3-yl}-7-methyl-3H-benzimidazol-5-yl)-piperazine-l-carbaldehyde: 1H
NMR (400 MHz, CD3OD) δ 8.12 (IH, s), 7.50 (IH, s), 7.24 - 7.38 (6H, m), 6.23 (IH, d, 7 = 7.6 Hz), 4.93 - 4.96 (IH, m), 3.54 - 3.79 (10H, m), 2.58 (3H, s). LCMS (M+H)+ m/z 507 (t = 1.29 min.). The following examples (Examples 554-575) were prepared according to Scheme VII and III
Example 554
Figure imgf000199_0001
(S)-4-[2-(3-ChIoro-phenyl)-2-hydroxy-ethylamino]-3-(4-methyl-6-morpholin-4- yl-lH-benzoimidazol-2-yl)-l H-pyridin-2-one: To a suspension of 4-Chloro-3-(4- methyl-6-morpholin-4-yl- 1 H-benzoimidazol-2-yl)- 1 H-pyridin-2-one and the corresponding iodo compound 4-Iodo-3-(4-methyl-6-morpholin-4-yl-lH- benzoimidazol-2-yl)-lH-pyridin-2-one (4.43 g, - 11 mmol) in acetonitrile (100 ml) triethylamine (7.0 ml, 50 mmol) and 2-(S)-2-(3-Chloro-phenyl)-2-hydroxy-ethylamine hydrochloride (2.55 g, 12.2 mmol) were added. The mixture was stirred at 85°C overnight. LCMS showed some starting material pyridone left. 2-(S)-2-(3-Chloro- phenyl)-2-hydroxy-ethylamine hydrochloride (0.22 g, 1.06 mmol) was added and the mixture stinted at 85°C another 24 hours. After evaporation of volatiles an aqueous solution of Cs2CO3 (200 ml, 10%) was added, the suspension sonicated for 5 minutes and stirred overnight. The product was filtered, washed with water and recrystallized from methanol - chloroform. The title compound was isolated as yellow crystals. (3.951 g, 75%). LCMS (M+H)+ m/z 480 (t = 1.31 min.). HPLC t = 4.93 min, YMC- Pack ODS-A 3.0 x 50 mm; 0 - 100% gradient over 8 min; 2.5 mL min flow rate. Η NMR of mono-HCl salt (500 MHz, DMSO-d6) δ 13.3 (IH, broad s), 11.22 (IH, s), 10.9 (IH, broad s), 7.65 (IH, broad s), 7.60 (IH, s), 7.45 (d, 7 = 7.6 Hz), 7.38-7.30 (4H, m) 6.19 (IH, d, 7 = 7.5 Hz), 4.92 (IH, t, 7 = 5.3 Hz), 4.00 (6H, broad), 3.67 (IH, m), 3.52 (5H, m), 2.58 (3H, s).
General procedure for preparation of mono- and bis- HCl salts:
A solution or suspension of free base in methanol is treated with 1.00 (or 2.00 resp.) equivalent of 1.00 N aqueous HCl. If significant amounts of compound remain insoluble an equal volume of dichlorethane is added to improve solubility. The mixture is filtered and concentrated in vacuo. .Small amounts were evaporate to dryness, large-scale preparations for in vivo studies were concentrated until most of the compound crystallized, then filtered.
Example 555
Figure imgf000200_0001
(S)-4-[2-(3-Bromo-4-methoxy-phenyl)-2-hydroxy-ethylamino]-3-(4-methyl-6- morpholin-4-yl-lH-benzoimidazol-2-yl)-lH-pyridin-2-one: 1H NMR (300 MHz, CD3OD) δ 7.68 - 7.60 (m, 2H), 7.43 - 7.28 (m, 3H), 6.98 (d, IH, J = 8.50 Hz), 6.26 (d, IH, J = 7.7 Hz), 4.97 - 4.89 (m, IH), 4.18 - 4.04 (m, 4H), 3.82 (s, 3H), 3.73 - 3.55 (m, 6H), 2.63 (s, 3H); LCMS (M+H)+ m/z 554, 556.
Example 556
Figure imgf000200_0002
(S)-4-[2-(3-Chloro-4-fluoro-phenyl)-2-hydroxy-ethylamino]-3-(4-methyl-6- morpholin-4-yl-lH-benzoimidazol-2-yl)-lH-pyridin-2-one: Η NMR (300 MHz, CD3OD) δ 7.65 - 7.50 (m, 2H), 7.43 - 7.20 (m, 3H), 7.18 (dd, IH, J = 8.9, 8.8 Hz), 6.27 (d, IH, J = 7.6 Hz), 5.00 - 4.91 (m, IH), 4.15 - 4.02 (m, 4H), 3.75 - 3.60 (m, 6H), 2.64 (s, 3H); LCMS (M+H)+ m z 498, 500. Example 557
Figure imgf000201_0001
(S)-4-[2-(3-Chloro-4-methoxy-phenyl)-2-hydroxy-ethylamino]-3-(4-methyl-6- morpholin-4-yl-lH-benzoimidazol-2-yl)-lH-pyridin-2-one: 1H NMR (400 MHz, CD3OD) δ 7.67 (brs, IH), 7.45 (brs, IH), 7.40 - 7.28 (m, 3H), 7.00 (d, IH, J = 8.4 Hz), 6.27 (d, IH, J = 7.6 Hz), 4.95 - 4.84 (m, IH), 4.15 - 4.05 (m, 4H), 3.82 (s, 3H), 3.75 - 3.55 (m, 6H), 2.64 (s, 3H); LCMS (M+H)+ m/z 510, 512.
Example 558
Figure imgf000201_0002
(S)-4-[2-(7-Bromo-2,3-dihydro-benzofuran-5-yl)-2-hydroxy-ethylamino]-3-(4- methyl-6-morphoIin-4-yI-lH-benzoimidazol-2-yl)-lH-pyridin-2-one: 'H NMR (400 MHz, CD3OD) δ 7.54 (brs, IH), 7.35 - 7.30 (m, 2H), 7.27 (brs, IH), 7.22 (brs, IH), 6.27 (d IH, J = 7.6 Hz), 4.95 - 4.87 (m, IH), 4.60 - 4.50 (m, 2H), 4.10 - 4.00 (m, 4H), 3.75 - 3.60 (m, 6H), 3.25 - 3.13 (m, 2H), 2.62 (s, 3H); LCMS (M+H)+ m/z 566, 568. Example 559
Figure imgf000202_0001
4-[2-(3-Chloro-phenyl)-2(S)-hydroxy-ethyIamino]-3-[4-methyl-6-[2(S),6(R)- dimethyI-morpholine-4-yl]-lH-benzoimidazol-2-yl]-lH-pyridine-2-one. 1H NMR (400 MHz, CD3OD) δ 7.23-7.57 (7H, - m), 6.22 (IH, d, 7 = 7.6 Hz), 4.97 (IH, m), 4.06 (2H, m), 3.62-3.68 (4H, m), 3.20-3.34 (2H, m), 2.63 (3H, s), 1.30 (6H, d, 7 = 6.28 Hz), LCMS (M+H)+ m/z 508 (t = 2.12 min.) Example 560
Figure imgf000202_0002
4-[2-(3-Bromo-4-methoxy-phenyl)-2(5)-hydroxy-ethylamino]-3-[4-methyl-6- [2(S),6(R)-dimethyl-morpholine-4-yl]-lH-benzoimidazol-2-yl]-lH-pyridine-2- one.1H NMR (400 MHz, CD3OD) δ 7.73 (IH- s), 7.63 (IH, s), 7.42 (IH, s), 7.34 (2H, m), 6.96 (IH, d, 7 = 8.48 Hz), 6.21 (IH, d, 7 = 7.48 Hz), 4.87 (IH, m), 4.23 (2H, m), 3.57-3.67(4H, m), 3.34 (3H, s) 3.30-3.32(2H, m), 2.60 (IH, s), 1.30 (6H, d, 7 = 6.2 Hz) LCMS (M+H)+ m/z 582 (t = 2.03 min.) Example 561
Figure imgf000203_0001
4-[2-(3-Chloro-phenyl)-(S)-2-hydroxy-ethylamino]-3-{6-[(R 2-fluoromethyl- morpholin-4-yl]-4-methyl-lH-benzimidazol-2-yl}-lH-pyridin-2-one and 4-[2-(3- chloro-phenyl)-(S)-2-hydroxy-ethylamino]-3-{6-[(S)-2-fluoromethyl-morpholin- 4-yl]-4-methyl-lH-benzimidazol-2-yl}-lH-pyridin-2-one : 1H NMR (400 MHz, CD3OD) δ 7.47 (IH, s), 7.23 - 7.43 (6H, m), 6.22 (IH, d, 7 = 7.2 Hz), 4.87 - 4.94 (IH, m), 4.60 (IH, d, 7= 3.4 Hz), 4.48 (IH, d, 7 = 3.4 Hz), 4.04 - 4.15 (3H, m), 3.28 - 3.62 (6H, m), 2.56 (3H, s). LCMS (M+H)+ m/z 512 (t = 1.35 min.).
Example 562
Figure imgf000203_0002
4-[2-(3-Bromo-4-methoxy-phenyl)-(S)-2-hydroxy-ethylamino]-3-{6-[(Rj-2- fluoromethyl-morpholin-4-yl]-4-methyl-lH-benzimidazol-2-yl}-lH-pyridin-2-one and 4-[2-(3-bromo-4-methoxy-phenyl)-(S)-2-hydroxy-ethylamino]-3-{6-[(Sj-2- fluoromethyI-morpholin-4-yI]-4-methyl-lH-benzimidazol-2-yl}-lH-pyridin-2- one: Η NMR (400 MHz, CD3OD) δ 7.60 (IH, narrow d, J = 2.0 Hz), 7.56 (IH, s), 7.39 (IH, br s), 7.34 (IH, dd, 7 = 2.0, 8.4 Hz), 6.97 (IH, d, J 8.4 Hz), 6.27 (IH, d, 7 = 6.4 Hz), 4.86 (IH, m), 4.61 (IH, m), 4.50 (IH, m), 4.77 - 4.19 (3H, m), 3.82 (3H, s), 3.38 - 3.74 (8H, m), 2.63 (3H, s). LCMS (M+H)+ m/z 586 (t = 1.31 min.).
Example 563
Figure imgf000204_0001
4-[2-(3-Chloro-4-methoxy-phenyl)-(S)-2-hydroxy-ethylamino]-3-{6-[(R)-2- fluoromethyl-morpholin-4-yl]-4-methyl-lH-benzimidazol-2-yl}-lH-pyridin-2-one and 4-[2-(3-chloro-4-methoxy-phenyl)-(S)-2-hydroxy-ethylamino]-3-{6-[(S 2- fluoromethyl-morpholin-4-yl]-4-methyl-lH-benzimidazol-2-yl}-lH-pyridin-2- one: Η NMR (400 MHz, CD3OD) δ 7.45 (IH, s), 7.28 - 7.30 (3H, m), 7.10 (IH, s), 6.97 (IH, d, 7 = 8.0 Hz), 6.20 (IH, d, 7 = 6.4 Hz), 4.85 (IH, m), 4.58 (IH, br s), 4.46 (IH, br s), 3.93 - 4.10 (3H, m), 3.80 (3H, s), 3.59 (4H, m), 3.07 - 3.29 (2H, m), 2.55 (3H, s). LCMS (M+H)+ m/z 542 (t = 1.28 min.).
Example 564
Figure imgf000204_0002
4-[2-(7-Bromo-2,3-dihydro-benzofuran-5-yl)-(S)-2-hydroxy-ethylamino]-3-{6- [(R)-2-fluoromethyl-morpholin-4-yl]-4-methyl-lH-benzimidazol-2-yl}-lH- pyridin-2-one and 4-[2-(7-bromo-2,3-dihydro-benzofuran-5-yl)-(S)-2-hydroxy- ethylamino]-3-{6-[(SJ-2-fluoromethyl-morpholin-4-yI]-4-methyl-lH- benzimidazol-2-yl}-lH-pyridin-2-one: 1H NMR (400 MHz, CD3OD) δ 7.26 -7.40 (4H, m), 7.19 (IH, s), 6.24 (IH, d, 7 = 7.6 Hz), 4.81 - 4.82 (IH, m), 4.49 - 4.61 (4H, m), 3.18 - 4.18 (11H, m), 2.59 (3H, s). LCMS (M+H)+ m/z 598 (t = 1.32 min.).
Example 565
Figure imgf000205_0001
4-[2-(3-Chloro-phenyl)-(S)-2-hydroxy-ethylamino]-3-{6-[(R)-2-hydroxymethyl- morpholin-4-yl]-4-methyl-lH-benzimidazol-2-yI}-lH-pyridin-2-one and 4-[2-(3- chIoro -phenyl)-(S)-2-hydroxy-ethylamino]-3-{6-[(5y)-2-hydroxy-methyl- morpholin-4-yl]-4-methyl-lH-benzimidazol-2-yl}-lH-pyridin-2-one: 1H NMR (400 MHz, CD3OD) δ 7.49 (IH, s), 7.23 - 7.37 (6H, m), 7.10 (IH, s), 6.24 (IH, d, 7 = 7.6 Hz), 4.95 - 4.96 (IH, m), 4.19 (IH, m), 3.94 - 4.80 (2H, m), 3.59 - 3.71 (8H, m), 2.63 (3H, s). LCMS (M+H)+ m/z 510 (t = 1.21 min.).
Example 566
Figure imgf000205_0002
4-[2-(3-Bromo-4-methoxy-phenyl)-(S)-2-hydroxy-ethylamino]-3-{6-[(Rj-2- hydroxymethyl-morpholin-4-yl]-4-methyI-lH-benzimidazoI-2-yl}-lH-pyridin-2- one and 4-[2-(3-bromo-4-methoxy-phenyl)-(S)-2-hydroxy-ethylamino]-3-{6-[ Sj- 2-hydroxy-methyl-morpholin-4-yl]-4-methyl-lH-benzimidazol-2-yl}-lH-pyridin- 2-one: 1H NMR (400 MHz, CD3OD) δ 7.62 (IH, narrow d, 7 = 2.0 Hz), 7.54 (IH, s), 7.29 - 7.36 (3H, m), 6.95 (IH, d, 7 = 8.4 Hz), 6.23 (IH, d, 7 = 7.6 Hz), 4.88 - 4.89 (IH, m), 3.56 - 4.19 (1 IH, m), 3.80 (3H, s), 2.60 (3H, s). LCMS (M+H)+ m/z 584 (t = 1.16 min.).
Example 567
Figure imgf000206_0001
4-[2-(3-Chloro-4-methoxy-phenyl)-(S)-2-hydroxy-ethylamino]-3-{6-[(RJ-2- hydroxymethyI-morpholin-4-yl]-4-methyl-lH-benzimidazol-2-yI}-lH-pyridin-2- one and 4-[2-(3-chloro-4-methoxy-phenyl)-(S)-2-hydroxy-ethylamino]-3-{6-[ S 2-hydroxy-methyl-morpholin-4-yl]-4-methyl-lH-benzimidazol-2-yl}-lH-pyridin- 2-one: 1H NMR (400 MHz, CD3OD) δ 7.68 (IH, s), 7.54 (IH, s), 7.45 (IH, s), 7.39 (IH, s), 7.30 - 7.32 (2H, m), 6.98 (IH, d, 7 = 8.4 Hz), 6.22 (IH, d, 7 = 7.6 Hz), 4.89 (IH, m), 4.11 - 4.19 (3H, m), 3.80 (3H, s), 3.49 - 3.72 (8H, m), 2.60 (3H, s). LCMS (M+H)+ m/z 540 (t = 1.09 min.).
Example 568
Figure imgf000207_0001
4-[2-(3-Chloro-phenyI)-(5)-2-hydroxy-ethylamino]-3-{6-[(R>2-methyl- morpholin-4-yl]-4-methyl-lH-benzimidazol-2-yl}-lH-pyridin-2-one and 4-[2-(3- chloro-phenyl)-(S)-2-hydroxy-ethylamino]-3-{6-[(S -2-methyl-morpholin-4-yl]-4- methyl-lH-benzimidazol-2-yl}-lH-pyridin-2-one: 1H NMR (400 MHz, CD3OD) δ
7.75 (IH, s), 7.48 (IH, s), 7.42 (IH, s), 7.23 - 7.37 (4H, m), 6.24 (IH, d, 7 = 7.2 Hz), 4.94 - 4.97 (IH, m), 4.11 - 4.16 (3H, m), 3.61 - 3.68 (5H, m), 3.38 (IH, m), 2.64 (3H, s), 1.29 (3H, d, 7 = 6.4 Hz). LCMS (M+H)+ m/z 494 (t = 1.32 min.).
Example 569
Figure imgf000207_0002
4-[2-(3-Bromo-4-methoxy-phenyl)-(5)-2-hydroxy-ethyIamino]-3-{6-[(R)-2- methyl-morpholin-4-yl]-4-methyl-lH-benzimidazol-2-yl}-lH-pyridin-2-one and 4-[2-(3-bromo-4-methoxy-phenyl)-(5)-2-hydroxy-ethylamino]-3-{6-[(S)-2-methyl- morpholin-4-yl]-4-methyl-lH-benzimidazol-2-yl}-lH-pyridin-2-one: 1H NMR (400 MHz, CD3OD) δ 7.63 (IH, s), 7.62 (IH, s), 7.30 - 7.61 (3H, m), 6.95 (IH, d, 7 = 8.4 Hz), 6.22 (IH, d, 7 = 7.6 Hz), 4.88 - 4.90 (IH, m), 4.08 - 4.18 (3H, m), 3.80 (3H, s), 3.61 - 3.67 (5H, m), 3.32 - 3.34 (IH, m), 2.60 (3H, s), 1.28 (3H, d, 7 = 6.0 Hz). LCMS (M+H)+ m/z 568 (t = 1.31 min.).
Example 570
Figure imgf000208_0001
4-[2-(3-Chloro-4-methoxy-phenyl)-(S)-2-hydroxy-ethylamino]-3-{6-[(R 2- methyl-morpholin-4-yl]-4-methyl-lH-benzimidazoI-2-yl}-lH-pyridin-2-one and
4-[2-(3-chloro-4-methoxy-phenyl)-(S)-2-hydroxy-ethylamino]-3-{6-[(5)-2-methyl- morpholin-4-yl]-4-methyl-lH-benzimidazol-2-yI}-lH-pyridin-2-one: 1H NMR (400 MHz, CD3OD) δ 7.48 (IH, narrow d, 7 = 2.0 Hz), 7.40 (IH, br s), 7.31 (IH, narrow d, 7 = 2.0 Hz), 7.29 (IH, narrow d, 7 = 2.0 Hz), 6.97 (IH, d, 7 = 8.4 Hz), 6.22 (IH, d, 7 = 7.6 Hz), 4.87 - 4.90 (IH, m), 4.11 (IH, m), 3.95 - 4.01 (2H, m), 3.80 (3H, s), 3.47 (4H, m), 3.32 (IH, m), 3.25 (IH, m), 2.57 (3H, s), 1.27 (3H, d, 7 = 6.4 Hz). LCMS (M+H)+ m/z 525 (t = 1.27 min.).
Example 571
Figure imgf000208_0002
4-[2-(3-Chloro-phenyl)-(S)-2-hydroxy-ethylamino]-3-{6-[(R 2-methoxymethyl- morpholin-4-yl]-4-methyl-lH-benzimidazol-2-yl}-lH-pyridin-2-one and 4-[2-(3- chloro -phenyl)-(S)-2-hydroxy-ethylamino]-3-{6-[(S)-2-methoxy-methyl- morpholin-4-yl]-4-methyl-lH-benzimidazol-2-yl}-lH-pyridin-2-one: 1H NMR (400 MHz, CD3OD) δ 7.49 (IH, narrow d, 7 = 2.0 Hz), 7.23 - 7.36 (6H, m), 6.22 (IH, d, 7 = 7.2 Hz), 4.93 - 4.96 (IH, m), 4.08 - 4.21 (1 IH, m), 3.38 (3H, s), 2.60 (3H, s), 1.27 (3H, d, 7 = 6.4 Hz). LCMS (M+H)+ m/z 524 (t = 1.35 min.).
Example 572
Figure imgf000209_0001
4-[2-(3-Bromo-4-methoxy-phenyI)-(S)-2-hydroxy-ethylamino]-3-{6-[(R 2- methoxymethyl-morpholin-4-yl]-4-methyl-lH-benzimidazol-2-yl}-lH-pyridin-2- one and 4-[2-(3-bromo-4-methoxy-phenyl)-(5)-2-hydroxy-ethyIamino]-3-{6-[ 5)- 2-methoxymethyl-morpholin-4-yl]-4-methyl-lH-benzimidazol-2-yl}-lH-pyridin- 2-one: 1H NMR (400 MHz, CD3OD) δ 7.63 (IH, narrow d, 7 = 2.0 Hz), 7.40 (IH, br s), 7.30 -7.36 (3H, m), 7.19 (IH, br s), 6.95 (IH, d, 7 = 8.4 Hz), 6.23 (IH, d, 7 = 7.2 Hz), 4.87 - 4.89 (IH, m), 4.18 (IH, m), 3.97 - 4.03 (3H, m), 3.80 (3H, s), 3.54 - 3.66 (7H, m), 3.39 (3H, s), 2.59 (3H, s). LCMS (M+H)+ m/z 598 (t = 1.31 min.).
Example 573
Figure imgf000209_0002
4-[2-(3-Chloro-4-methoxy-phenyl)-(S)-2-hydroxy-ethylamino]-3-{6-[(R 2- methoxymethyl-morpholin-4-yl]-4-methyl-lH-benzimidazol-2-yl}-lH-pyridin-2- one and 4-[2-(3-chIoro-4-methoxy-phenyl)-(S)-2-hydroxy-ethylamino]-3-{6-[(Sj- 2-methoxymethyl-morpholin-4-yl]-4-methyl-lH-benzimidazol-2-yI}-lH-pyridin- 2-one: 1H NMR (400 MHz, CD3OD) δ 7.52 (IH, s), 7.46 (IH, narrow d, 7 = 1.6 Hz), 7.29 -7.34 (3H, m), 6.99 (IH, d, 7 = 8.4 Hz), 6.24 (IH, d, 7 = 7.2 Hz), 4.89 (IH, m), 4.06 - 4.17 (3H, m), 3.81 (3H, s), 3.54 - 3.68 (8H, m), 3.38 (3H, s), 2.60 (3H, s). LCMS (M+H)+ m/z 554 (t = 1.28 min.).
Example 574
Figure imgf000210_0001
4-[2-(3-Chloro-phenyl)-2(S)-hydroxy-ethylamino]-3-[4-methyl-6-(4-methyl- piperazin-l-yl)-lH-benzoimidazoI-2-yl]-lH-pyridine-2-one. 'H NMR (400 MHZ, CD3OD) δ 7.46 (IH, s), 7.24-7.36 (m, 4H), 7.04 (2H, s), 6.22 (IH, d, 7 = 7.64 Hz), 4.89 (IH, m), 3.30-3.82 (10H, m) 2.97 (3H, s), 2.57 (3H, -s). LCMS (M+H)+ m/z 493 (t = 1.56 min.)
Example 575
Figure imgf000210_0002
4-[2-(3-Bromo-4-methoxy-phenyl)-2(S)-hydroxy-ethyIamino]-3-[4-methyl-6-(4- methyl-piperazin-l-yl)-lH-benzoimidazol-2-yl]-lH-pyridine-2-one. 1H NMR (400 MHz, CD3OD) δ 7.61 (IH, d, 7 = 2.0 Hz), 7.32-7.37 (2H, m), 7.05 (2H, s), 6.97 (IH, d, 7 = 8.52 Hz), 6.24 (IH, d, 7 = 7.64 Hz), 4.82 (lH,m), 3.82 (3H, s) 3.30-3.64 (10H, m), 2.98 (3H, s), 2.56 (3H, s) LCMS (M+H)+ m/z 567 (t = 1.53 min.)
The following examples (576-581) were prepared according to Scheme VII and III and illustrate the acylation of a 4-amino-piperidine_derivative
Example 576 (General procedure for Examples 576-581)
Figure imgf000211_0001
4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-{6-[4-(acetamido)-piperidin-l- yl]-4-methyl-lH-benzoimidazol-2-yl}-lH-pyridin-2-one: A solution of the 4- amino-piperidine compound (~ 50-100 umol) in 5 ml MeOH is cooled to 0°C. Then -10 equivalent of Huenigs base and ~ 3 equivalent of acyl chloride are added. The vial is shaken once and allowed to stand at ambient temperature for 1 hour. Evaporation of volatiles and purification by prep. HPLC gives the 4-acyl- aminopiperidine compounds. Η NMR (500 MHz, CD3OD) δ 8.06 (IH, s), 7.68 (IH, s), 7.49 (IH, s), 7.44 (IH, d, J = 7.6), 7.27 - 7.38 (4H, m), 6.33 (IH, d, 7= 7.6 Hz), 4.96 (IH, dd, 7 = 4.1, 7.6 Hz), 4.17 (IH, m), 3.87 (4H, m), 3.65 (IH, dd, 7 = 4.2, 14 Hz), 3.58 (IH, dd, 7 = 7.8, 13.8 Hz), 2.72 (3H, s), 2.31 (2H, m), 2.19 (2H, m), 2.02 (3H, s). LCMS (M+H)+ m/z 535 (t = 1.03 min, YMC Xterra C18 S7 3.0 x 50 mm; 0 - 100% gradient over 1.5 min; 5 mL/min flow rate). Example 577
Figure imgf000212_0001
4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-{6-[4-(2-hydroxyacetamido)- piperidin-l-yl]-4-methyl-lH-benzoimidazol-2-yl}-lH-pyridin-2-one:
LCMS (M+H)+ m/z 551 (t = 1.27 min.). HPLC t = 5.01 min, Waters Xterra C18 S5 4.6 x 30 mm; 0 - 100% gradient over 12 min; 5 mL/min flow rate.
Example 578
Figure imgf000212_0002
4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-{6-[4-(2-fluoroacetamido)- piperidin-l-yl]-4-methyl-lH-benzoimidazol-2-yl}-lH-pyridin-2-one: LCMS (M+H)+ m/z 597 (t = 1.31 min.). HPLC t = 6.90 min, YMC-Pack ODS-A 3.0 x 50 mm, 0 - 100% gradient over 12 min; 2.5 mL/min flow rate. Example 579
Figure imgf000212_0003
21 4-[2-(3-Bromo-4-methoxy-phenyl)-2-hydroxy-ethylamino]-3-{6-[4-(acetamido)- piperidin-l-yl]-4-methyl-lH-benzoimidazol-2-yl}-lH-pyridin-2-one:
LCMS (M+H)+ m/z 609 (t = 1.27 min.). HPLC t = 5.00 min, Waters Xterra C18 S5 4.6 x 30 mm; 0 - 100% gradient over 12 min; 5 mL/min flow rate.
Example 580
Figure imgf000213_0001
4-[2-(3-Bromo-phenyl)-2-hydroxy-ethylamino]-3-{6-[4-(2-hydroxyacetamido)- piperidin-l-yl]-4-methyl-lH-benzoimidazol-2-yl}-lH-pyridin-2-one:
LCMS (M+H)+ m/z 595 (t = 1.30 min.). HPLC t = 5.09 min, Waters Xterra C18 S5 4.6 x 30 mm; 0 - 100% gradient over 12 min; 5 mL/min flow rate.
Example 581
Figure imgf000213_0002
4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-{6-[4-(2-fluoroacetamido)- piperidin-l-yl]-4-methyl-lH-benzoimidazol-2-yl}-lH-pyridin-2-one:
LCMS (M+H)+ m/z 553 (t = 1.35 min.). HPLC t = 5.90 min, YMC-Pack ODS-A 3.0 x 50 mm; 0 - 100% gradient over 10 min; 2.5 mL/min flow rate.
The following examples (582-584) were prepared according to Scheme VII and III and illustrate the carbamoylation of a 4-amino-piperidine derivative
General Procedure for Examples 582-584 A solution of the 4-amino-piperidine compound (~ 50-100 umol) in 5 ml MeOH is cooled to 0°C. Then -10 equivalent of Huenigs base and - 3 equivalent of carbamoyl chloride are added. The vial is shaken once and allowed to stand at ambient temperature overnight. Evaporation of volatiles and purification by prep. HPLC gives the 4-carbamoyl-aminopiperidine compounds.
Example 582
Figure imgf000214_0001
4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-{6-[4-(2- methoxyethoxy carbamoyl)- piperidin-l-yl]-4-methyl-lH-benzoimidazol-2-yl}- lH-pyridin-2-one: 1H NMR (500 MHz, CD3OD) δ 7.90 (IH, s), 7.26 -7.52 (6H, m), 6.29 (IH, d, 7 = 7.5 Hz), 4.97 (IH, dd, 7 = 4.1, 7.0 Hz), 4.20 (2H, m), 3.93 (IH, m), 3.60 - 3.84 (8H, m), 3.37 (3H, s), 2.69 (3H, s), 2.33 (2H, m), 2.13 (2H, m). LCMS (M+H)+ m/z 595 (t = 1.09 min, YMC Xterra C18 S7 3.0 x 50 mm; 0 - 100% gradient over 1.5 min; 5 mL/min flow rate).
Example 583
Figure imgf000214_0002
4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-{6-[4-(methoxycarbamoyl)- piperidin-l-yl]-4-methyl-lH-benzoimidazol-2-yl}-lH-pyridin-2-one: 1H NMR (500 MHz, CD3OD) δ 8.00 (IH, s), 7.63 (IH, s), 7.26 -7.49 (5H, m), 6.32 (IH, d, 7 = 7.6 Hz), 4.96 (IH, dd, 7 = 4.2, 7.6 Hz), 3.93 (IH, m), 3.79 - 3.88 (4H, m), 3.67 (3H, s), 3.56 -3.65 (2H, m), 2.71 (3H, s), 2.32 (2H, m), 2.17 (2H, m). LCMS (M+H)+ m/z 551 (t = 1.07 min, YMC Xterra C18 S7 3.0 x 50 mm; 0 - 100% gradient over 1.5 min; 5 mL/min flow rate).
Example 584
Figure imgf000215_0001
4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-{6-[4-(2-fluoroethoxy carbamoyl)- piperidin-l-yl]-4-methyl-lH-benzoimidazol-2-yl}-lH-pyridin-2-one:
1H NMR (500 MHz, CD3OD) δ 7.92 (IH, s), 7.26 -7.54 (6H, m), 6.3 (IH, d, 7= 7.5 Hz), 4.97 (IH, dd, 7 = 4.5, 7.5 Hz), 4.63 (IH, broad s), 4.53 (IH, d, J = 2.5 Hz), 4.32 (IH, broad s), 4.27 (IH, broad s), 3.93 (IH, m), 3.79 - 3.85 (4H, m), 3.58 - 3.72 (2H, m), 2.70 (3H, s), 2.33 (2H, m), 2.15 (2H, m). LCMS (M+H)+ m/z 583 (t = 1.29 min, YMC Xterra C18 S7 3.0 x 50 mm; 0 - 100% gradient over 2 min; 5 mL/min flow rate).
The following examples (585-590) were prepared according to Scheme VII and III and illustrate the use of an alcohol as the nucleophile in Scheme VII Example 585 (General procedure for Examples 585-590)
Figure imgf000215_0002
(S)-4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-[4-methyl-6-(2-morpholin- 4-yl-ethoxy)-lH-benzimidazol-2-yl]-lH-pyridin-2-one: Η NMR (400 MHz, CD3OD) δ 7.53 (IH, s), 7.37 -7.39 (IH, m), 7.23 - 7.30 (3H, m), 7.01 (IH, s), 6.75 (IH, s), 6.21 (IH, d, 7 = 7.2 Hz), 4.98 (IH, t, 7 = 5.6 Hz), 4.40 (2H, br s), 3.97 (4H, br s), 3.45 - 3.73 (8H, m), 2.54 (3H, s). LCMS (M+H)+ m/z 524 (t = 1.24 min.).
Example 586
Figure imgf000216_0001
(5)-4-[2-(3-Bromo-4-methoxy-phenyl)-2-hydroxy-ethylamino]-3-[4-methyl-6-(2- morpholin-4-yl-ethoxy)-lH-benzimidazol-2-yl]-lH-pyridin-2-one : 1H NMR (400 MHz, CD3OD) δ 7.67 (IH, narrow d, 7 = 1.6 Hz), 7.36 (IH, dd, 7 = 1.6, 8.4 Hz), 7.25 (IH, d, 7 = 7.2 Hz), 6.98 (IH, s), 6.92 (IH, d, 7 = 8.4 Hz), 6.75 (IH, s), 6.22 (IH, d, 7 = 7.2 Hz), 4.89 - 4.92 (IH, m), 4.41 (2H, br s), 3.97 (4H, br s), 3.79 (3H, s), 3.34 - 3.66 (8H, m), 2.51 (3H, s). LCMS (M+H)+ m/z 598 (t = 1.22 min.).
Example 587
Figure imgf000216_0002
(5)-4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-[4-methyl-6-(2-methoxy- ethoxy)-lH-benzimidazol-2-yl]-lH-pyridin-2-one : 1H NMR (400 MHz, CD3OD) δ 7.48 (IH, s), 7.25 -7.36 (4H, m), 6.96 (IH, s), 6.81 (IH, s), 6.20 (IH, d, 7 = 7.4 Hz), 4.90 - 4.93 (IH, m), 4.13 - 4.14 (2H, m), 3.76 - 3.77 (2H, m), 3.50 - 3.61 (2H, m), 3.43 (3H, s), 2.53 (3H, s). LCMS (M+H)+ m/z 469 (t = 1.52 min.). Example 588
Figure imgf000217_0001
(S)-4-[2-(3-Chloro-phenyI)-2-hydroxy-ethylamino]-3-[4-methyl-6-(2-hydroxy- ethoxy)-lH-benzimidazol-2-yl]-lH-pyridin-2-one : 1H NMR (400 MHz, CD3OD) δ 7.43 - 7.45 (2H, m), 7.27 -7.34 (3H, m), 7.08 (IH, s), 7.04 (IH, narrow d, 7 = 1.0 Hz), 6.28 (IH, d, 7 = 7.6 Hz), 4.87 (IH, m), 4.13 (2H, t, 7 = 4.6 Hz), 3.92 (2H, t, 7 = 4.6 Hz), 3.45 - 3.54 (2H, m), 2.60 (3H, s). LCMS (M+H)+ m/z 455 (t = 1.35 min.).
Example 589
Figure imgf000217_0002
(S)-4-[2-(3-Bromo-4-methoxy-phenyl)-2-hydroxy-ethylamino]-3-[4-methyl-6-(3- morpholin-4-yl-propoxy)-lH-benzimidazol-2-yl]-lH-pyridin-2-one: 1H NMR (400 MHz, CD3OD) δ 7.66 (IH, s), 7.35 (IH, dd, 7 = 1.6, 7.9 Hz), 7.25 (IH, br s), 6.93 (IH, s), 6.91 (IH, s), 6.68 (IH, s), 6.19 (IH, br s), 4.86 (IH, m), 4.05 - 4.10 (4H, br s), 3.79 (3H, s), 3.17 - 3.73 (12H, m), 2.50 (3H, s). LCMS (M+H)+ m/z 612 (t = 1.16 min.). Example 590
Figure imgf000218_0001
(S)-4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-[4-methyl-6-(3-morpholin- 4-yl-propoxy)-lH-benzimidazol-2-yl]-lH-pyridin-2-one: 1H NMR (400 MHz, CD3OD) δ 7.52 (IH, s), 7.36 - 7.38 (IH, m), 7.23 - 7.30 (3H, m), 6.92 (IH, s), 6.67 (IH, s), 6.18 (IH, d, 7 = 6.9 Hz), 4.96 (IH, t, 7 = 5.9 Hz), 4.04 - 4.08 (4H, m), 3.82 (2H, m), 3.56 - 3.65 (8H, m), 3.15 - 3.18 (2H, m), 2.52 (3H, s). LCMS (M+H)+ m/z 538 (t = 1.19 min.).
The following examples (591-593) were prepared according to Scheme VII and III wherein a cyano group (Scheme IV) is converted to an aldehyde which undergo reductive animation with an amine
Example 591 (General Procedure for Examples 591-593)
Figure imgf000218_0002
(S)-3-(4-Bromo-6-morpholin-4-ylmethyl-lH-benzimidazol-2-yl)-4-[2-(3-chloro- phenyl)-2-hydroxy-ethylamino]-lH-pyridin-2-one: To a solution of (S)-7-Bromo- 2- { 4-[2-(3-chloro-phenyl)-2-hydroxy-ethylamino]-2-oxo- 1 ,2-dihydro-pyridin-3-yl } - 3H-benzimidazole-5-carbaldehyde (130 mg, 0.27 mmol) in methanol (60 mL) was added morpholine (0.2 mL, excess). The reaction mixture was stirred 1 h at room temperature. Then NaCNBH3 (IM THF solution, 1.35 mL, 1.35 mmol) was added. The reaction mixture was stirred at room temperature overnight and concentrated in vacuo. The residue was purified by prep. HPLC to yield the title compound (68 mg, 45%).'H NMR (400 MHz, CD3OD) δ 7.83 (IH, s), 7.69 (IH, s), 7.68 (IH, s), 7.24 - 7.54 (4H, m), 6.30 (IH, d, 7 = 7.6 Hz), 4.89 - 5.02 (IH, m), 4.50 (2H, s), 3.66 - 3.71 (4H, m), 3.17 - 3.43 (6H, m). LCMS (M+H)+ m/z 558 (t = 1.41 min.).
Example 592
Figure imgf000219_0001
(5)-3-[4-Bromo-6-(4-methyI-piperazin-l-ylmethyl)-lH-benzimidazol-2-yl]-4-[2- (3-chloro-phenyl)-2-hydroxy-ethylamino]-lH-pyridin-2-one: 1H NMR (400 MHz, CD3OD) δ 7.70 (IH, s), 7.55 (2H, s), 7.42 (IH, d, 7 = 7.6 Hz), 7.24 - 7.32 (3H, m), 6.22 (IH, d, 7 = 7.6 Hz), 5.01 (IH, t, 7 = 6.2 Hz), 4.35 (2H, br s), 3.48 - 3.71 (10H, m), 2.98 (3H, s). LCMS (M+H)+ m/z 571 (t = 1.37 min.).
Example 593
Figure imgf000219_0002
(5)-4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-[4-methyl-6-(4-methyl- piperazin-l-ylmethyl)-lH-benzimidazol-2-yl]-lH-pyridin-2-one: Through a mixture of (5)-3-[4-Bromo-6-(4-methyl-piperazin-l-ylmethyl)-lH-benzimidazol-2- yl]-4-[2-(3-chloro-phenyl)-2-hydroxy-ethylamino]-lH-pyridin-2-one (80 mg, 0.14 mmol), tetramethyl tin (2.5 mL, excess), PdC12 (PPh3)4 (10 mg, 0.014 mmol), and KF (40 mg, 0.7 mmol) in DMF (2 mL) in a vial was bubbled nitrogen, sealed, and heated to 100°C for two days. The reaction mixture was passed through a small pad of celite. After concentration, the residue was purified by prep. HPLC to give the titled; compound (34 mg, 48%). 1H NMR (400 MHz, CD3OD) δ 7.53 (IH, s), 7.45 (IH, s), 7.24 - 7.40 (4H, m), 7.09 (IH, s), 6.22 (IH, d, 7 = 7.6 Hz), 4.99 (IH, t, 7 = 6.4 Hz), 4.04 (2H, br s), 3.61 - 3.74 (2H, m), 2.98 - 3.34 (8H, m), 2.80 (3H, s), 2.58 (3H, s). LCMS (M+H)+ m/z 507 (t = 1.29 min.).
The following examples (594-595) were prepared according to Scheme VII and III and illustrate the use of tetrahydropyrimidine as the nucleophile in Scheme VII
Example 594
Figure imgf000220_0001
4-[2-(3-Chloro-phenyl)-2(S)-hydroxy-ethylamino]-3-[4-methyl-6-(l,4,5,6- tetrahydropyrimidine-l-yl)-lH-benzoimidazol-2-yl]-lH-pyridine-2-one: Η NMR (400 MHz, CD3OD) δ 8.38 (IH, s), 7.52 (lH,s), 7.21-7.41 (5H, m), 7.04 (IH, d, 7 = 1.2Hz), 6.21(1H, d, 7 = 7.6Hz), 4.97 (IH, t, 7 = 4.9 Hz) 3.97(2H, m) 3.60-3.73 (2H, m),3.52(2H, t, J = 5.74 Hz) 2.60 (3H, s), 2.25 (2H, m) LCMS (M+H)+ m/z 477 (t = 1.79 min.)
Example 595
Figure imgf000220_0002
4-[2-(4-Methoxy-3-chloro-phenyl)-2(S)-hydroxy-ethylamino]-3-[4-methyl-6- (l,4,5,6-tetrahydropyrimidine-l-yl)-lH-benzoimidazol-2-yl]-lH-pyridine-2-one: Η NMR (400 MHz, CD3OD) δ 8.37 (IH, s), 7.52 (lH,s), 7.04-7.52(4H, m), 7.04 (IH, d, 7 = 1.2Hz), 6.21(1H, d, 7 = 7.6Hz), 4.98 (IH, t, 7 = 4.92 Hz) 3.97(5H, m) 3.60-3.73 (2H, m),3.52(2H, t, J = 5.74 Hz), 2.60 (3H, s), 2.26 (2H, m). LCMS (M+H)+ m/z 507 (t = 1.67 min.)
It is understood that the examples described above in no way serve to limit the true scope of this invention, but rather are presented for illustrative purposes. All references cited herein are incorporated by reference in their entirety.

Claims

What is claimed is:
1. A compound according to formula I
Figure imgf000222_0001
its enantiomers, diastereomers, pharmaceutically acceptable salts, hydrates, prodrugs and solvates thereof;
wherein
X is selected from the group consisting of N, C, C1-C3 alkyl, -C3 alkyl substituted with one or more R7, and a direct bond; Y is selected from the group consisting of O and S ;
W is selected from the group consisting of N, C, O, and S; provided that if W is O or S, R9 is absent;
R1, R2, R3, R4, R5, R6, R7, R8, R9 are each independently selected from the group consisting of H, Cι-6 alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, halo, amino, OR60, NO2, OH, SR60, NR60R61 , CN, CO2R60, CONR60R61, OCONR60R61,
NR62CONR60R61, NR60SO2R61 , SO2NR60R61, C(NR62)NR60R61, aryl, heteroaryl,
(CH2)nOR60, (CH2)nNR60Ro1, (CH2)nSR60, (CH2)naryl, (CH2)n heteroaryl, ,
(CH )n heterocycloalkyl, NH-Z-aryl, and NH-Z-heteroaryl; wherein n is 1 to 3; and Z is selected from the group consisting of C\ - C4 alkyl, alkenyl, and alkynyl chain; Z having one or more hydroxy, thiol, alkoxy, thioalkoxy, amino, halo,
NR60SO2R61 groups; Z optionally incorporating one or more groups selected from the group consisting of CO, CNOH, CNOR60, CNNR60 , CNNCOR60 and CNNSO2R60 ; and R60, and R61 are independently selected from the group consisting of H, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, hydroxy, alkoxy, aryl, heteroaryl, heteroarylalkyl, and alkyl-R " wherein
R25 is hydrogen, alkenyl, hydroxy, thiol, alkoxy, thioalkoxy, amino, alkylamino, dialkylamino, aryl, heteroaryl, cyano, halo, sulfoxy, sulfonyl, - ; NR30COOR31, -NR30C(O)R31, -NR30SO2R31, -C(O)NR30R31, heteroaryl or heterocycloalkyl; and
R and R are, independently, hydrogen, alkyl, cycloalkyl, or alkyl-R .
2. The compound according to claim 1 wherein R1, R7, R8and R9 are H;
R2 and R4 are H or F;
Y is O;
X is selected from the group consisting of N and CH;
W is N; R5 is selected from the group consisting of H, methyl, ethyl, isopropyl, secondary butyl, cyclopropyl, F, and CF3;
R6 is selected from the group consisting of H, 2-aminomethylpyridine, NHCH2CH(OH)Ph, NHCH2CH(OH)(3-Cl-Ph), NHCH2CH(OH)(3-Br-Ph), NHCH2CH(OH)(3-Br-4-OMe-Ph), NHCH(CH2OH)CH2Ph, NHCH2CH(OH)aryl, and NHCH(CH2OH)CH2aryl; and
R3 is selected from the group consisting of OR60, C(NH)NHR60, C(O)NHR60 imidazole, imidazoline, tetrahydropyrimidine, piperazine, morpholine, homomorpholine, piperidine, pyrrolidine, homopiperazine and amino; wherein
R60 is selected from the group consisting of H, alkyl, cycloalkyl, heterocycloalkyl, and alkyl-R25 wherein R25 is hydrogen, alkenyl, hydroxy, thiol, thioalkoxy, alkoxy, thioalkoxy, halo, cyano, sulfoxy, sulfonyl, -NR30COOR31, - NR30C(O)R31, -NR30SO2R31, -C(O)NR30R31, or a heteroaryl or heterocycloalkyl; and
R30 and R31 are, independently, hydrogen, alkyl, cycloalkyl, or alkyl-R .
3. The compound according to claim 2 wherein R is -OR60 and R is alkyl, or alkyl-R25, wherein R25 is hydrogen, alkenyl, hydroxy, thiol, alkoxy, thioalkoxy, amino, halo, cyano, sulfoxy, sulfonyl, -NR30COOR31, -NR30C(O)R31, -NR30SO2R31, -
C(O)NR30R31, heteroaryl or heterocycloalkyl; and
R30 and R3i are, independently, hydrogen, alkyl, cycloalkyl, or alkyl-R 25
4. The compound according to claim 3 wherein R60 is methyl, -(CH )nCH2OH, or -(CH2)nCH2N(CH2CH2)2O, and n is 0, 1, or 2.
The compound according to claim 3 wherein R is morpholine,
Figure imgf000224_0001
wherein R >33 is hydrogen, alkyl, or substituted alkyl.
6. The compound according to claim 2 wherein R3 is piperazine, homopiperazine, 3-methylpiperazine, or 3,5-dimethylpiperazine being optionally substituted at the 4-N position with a compound selected from the group consisting of alkyl, cycloalkyl, cycloalkylalkyl, alkyl-R25, -C(O)-R15, or -CO2R15 wherein R15 is hydrogen, alkyl, aryl, alkyl-R25, amino or aryl; and
R25 is hydrogen, alkenyl, hydroxy, thiol, alkoxy, thioalkoxy, amino, alkylamino, dialkylamino, cyano, halo, sulfoxy, sulfonyl, arylsulfonyl, -
NR30COOR31, -NR30C(O)R31, -NR30SO2R31, -C(O)NR30R31, heteroaryl or heterocycloalkyl; and
R and R are, independently, hydrogen, alkyl, cycloalkyl, or alkyl-R .
7. The compound according to claim 6 wherein said piperazine is substituted with methyl, ethyl, CH2-cyclopropyl, hydroxyethyl, 2-dimethylaminoethyl, 2- diethylaminoethyl, 2-aminoethyl, 2-methylaminoethyl, 2-ethylaminoethyl, methoxyethyl, ethoxyethyl, morpholine, and morpholinylethyl.
8. The compound according to claim 2 wherein R is an amino group selected from the group consisting of hydroxyalkylamino, aminoalkylamino, dialkylaminoalkylamino, and heterocycloalkylalkylamino.
9. The compound according to claim 8 wherein said amino is selected from the group consisting of NHCH2CH2OH, NMeCH2CH2OH, NEtCH2CH2OH, NHCH2CH2NH2, NMeCH2CH2NH2, NEtCH2CH2NH2, NHCH2CH2NMe2, NMeCH2CH2NMe2, NEtCH2CH2NMe2, NHCH2CH2NEt2, NMeCH2CH2NEt2, NEtCH2CH2NEt2, NHCH2CH2N(CH2CH2)2O, NMeCH2CH2N(CH2CH2)2θ, and NEtCH2CH2N(CH2CH2)2O.
10. The compound according to claim 2 wherein R is piperidine, optionally substituted with hydroxy, thiol, amino, alkylamino, dialkylamino, alkoxy, thioalkoxy, 1,3 dioxolane, 1,3 dioxane, -NHC(O)R15, -NHCO2R15, wherein R15 is hydrogen, alkyl, aryl or alkyl-R25 wherein R25 is hydrogen, alkenyl, hydroxy, thiol, alkoxy, thioalkoxy, amino, halo, cyano, sulfoxy, sulfonyl, -NR30COOR31, -NR30C(O)R31, -NR30SO2R31, -
C(O)NR30R31, heteroaryl or heterocycloalkyl; and
R30 and R31 are, independently, hydrogen, alkyl, cycloalkyl, or alkyl-R 25
11. The compound according to claim 2 wherein R3 is morpholine, thiomorpholine, sulfoxymorpholine, sulfonylmorpholine, or homomorpholine, or a substituted morpholine, thiomorpholine, sulfoxymorpholine, sulfonylmorpholine, or homomorpholine.
12. The compound according to claim 11 wherein said morpholine, thiomorpholine, sulfoxymorpholine, sulfonyl morpholine, or homomorpholine is substituted with hydroxy, thiol, amino, alkylamino, dialkylamino, alkoxy, thioalkoxy, alkyl-R25 -NHC(O)R15, -NHCO2R15, wherein R15 is hydrogen, alkyl, aryl or alkyl-R25 wherein R25 is hydrogen, alkenyl, hydroxy, thiol, alkoxy, thioalkoxy, amino, halo, cyano, sulfoxy, sulfonyl, -NR30COOR31, -NR30C(O)R31, -NR30SO2R31, -C(O)NR30R31, heteroaryl or heterocycloalkyl; and
R30 and R31 are, independently, hydrogen, alkyl, cycloalkyl, or alkyl-R25.
13. The compound according to claim 2 wherein R is (CH )n-morpholine or (CH2)„- piperazine, wherein n is 1 to 3.
14. The compound according to claim 2 wherein R is an optionally substituted N- tetrahydropyrimidine or N-imidazoline.
15. The compound according to claim 14 wherein at least one of said substituents is alkyl, hydroxyalkyl, alkoxyalkyl, haloalkyl, cyanoalkyl, carboxyl, or carboxamide.
16. The compound according to claim 2 wherein R3 is pyrrolidine.
17. The compound according to claim 2 wherein said pyrrolidine is selected from the group consisting of 3-hydroxyl pyrrolidine, 3- alkoxy pyrrolidine, and 3- alkylamino pyrrolidine, 3-dialkylamino pyrrolidine.
18. The compound according to claim 2 wherein R6 is selected from the group consisting of H, 2-aminomethylpyridine, NHCH2CH(OH)aryl, and NHCH(CH2OH)CH2aryl.
19. The compound according to claim 18 wherein said aryl is an optionally substituted phenyl.
20. The compound according to claim 19 wherein said phenyl is substituted with a Br, Cl, F, alkoxy or-NHSO2CH .
21. The compound according to claim 19 wherein said substituent is 3-Br, 3-C1 or 3-F.
22. The compound according to claim 19 wherein said substituent is 4-F or 4- methoxy.
23. The compound according to claim 19 wherein R is
Figure imgf000227_0001
wherein R17 is H, Br, Cl, or F and R40 is H or alkyl.
24. A compound having the formula:
Figure imgf000227_0002
wherein R12 and R13 are, independently, hydrogen, alkyl, or alkyl-R25 wherein
R25 is hydrogen, hydroxy, thiol, alkenyl, amino, alkoxy, thioalkoxy, halo, cyano, sulfoxy, sulfonyl, -CO2H, -C(O)NR30R31, -NR30SO2R31, -NR30C(O)R31, - NR30C(O)OR31, heteroaryl or heterocycloalkyl;
R . 17 , R and R , 19 are, independently, hydrogen, halogen, or alkoxy, or R and
R , 19 together form a heterocycloalkyl or heteroaryl group; and
^0 ^1 S
R and R are, independently, hydrogen, alkyl, or alkyl-R .
25. The compound according to claim 24 wherein R 25 is morpholine,
Figure imgf000228_0001
wherein R > 33 i;s hydrogen, alkyl, or substituted alkyl.
1
26. The compound according to claim 24 wherein R is hydrogen, methyl, hhyyddrrooxxyymmeetthhyyll,, mmeetthhooxxyymmeetthhyyll,, CCHH22FF,, CCHH22CCNN,, C CO2H, or -CONR30R31 wherein R30 and R are, independently, hydrogen, or alkyl-R .
27. The compound according to claim 24 incorporating the side chain
Figure imgf000228_0002
wherein R40 is hydrogen or alkyl, and R70 hydrogen or halogen.
28. The compound according to claim 24 wherein R12 and R13 are H; R17 is Br, F, or Cl; R18 is methoxy or fluoro; and R19 is H; or R18 and R19 together form 4-O, 5 dihydrofuranyl.
29. The compound according to claim 24 wherein R and R13 are methyl; R17 is Br, F or Cl, R18 is hydrogen or methoxy; and R19 is H.
30. A compound having the formula
Figure imgf000229_0001
wherein R15 is hydrogen, alkyl, aryl, or -alkyl-R25, wherein
R25 is is-hydrogen, hydroxy, thiol, alkenyl, amino, alkoxy, thioalkoxy, halo, cyano, sulfoxy, sulfonyl,
-NR30COOR31, -NR30C(O)R31, -NR30SO2R31, -C(O)NR30R31, heteroaryl or heterocycloalkyl;
R16 is each independently hydrogen or methyl;
R17, R18 and R19 are, independently, hydrogen, halogen, or alkoxy; or R18 and R19 together form a heterocycloalkyl or heteroaryl group; and
R30 and R31 are, independently, hydrogen, alkyl or alkyl-R 25
31. The compound according to claim 30 wherein R is morpholine, thiomorpholine,
Figure imgf000229_0002
32. The compound according to claim 30 incorporating the sidechain
Figure imgf000230_0001
Figure imgf000230_0002
Figure imgf000230_0003
wherein R40 is hydrogen or alkyl and R17 hydrogen or halogen.
33. The compound according to claim 30 wherein R15 is hydrogen, methyl, ethyl, or
(CH2)nCH2-R25 wherein R25 is OH, OMe, F, CN, CF3, SOCH3 or SO2CH3, wherein n is O or 1.
34. The compound according to claim 30 wherein R 15 is cyanoethyl, hydroxyethyl, CH2CH2SOCH3, CH2CH2CH2F, CH2CH2CH2CN, or CH2CH2CF3; R 16 and R19 are H; R17 is Br, or Cl; and R18 is hydrogen or methoxy.
35. A compound having the formula:
Figure imgf000230_0004
wherein
R15 is hydrogen, alkyl, aryl or alkyl-R25 R25 is hydrogen, hydroxy, thiol, alkenyl, alkoxy, thioalkoxy, amino, halo, cyano, sulfoxy, sulfonyl, -NR30COOR31, -NR30C(O)R31, -NR30SO2R31, - C(O)NR30R31, heteroaryl or heterocycloalkyl;
R16 is each independently hydrogen or methyl;
17 1 R 1 Q 18
R , R and R are, independently, hydrogen, halogen, or alkoxy, or R and R1 together form a heterocycloalkylor heteroaryl group; and r* i i >y
R and R are, independently, hydrogen, alkyl, cycloalkyl or alkyl-R .
36. The compound according to claim 35 wherein R >25 i •s morpholine, thiomorpholine,
Figure imgf000231_0001
wherein R , 33 is hydrogen, alkyl, or substituted alkyl.
37. The compound according to claim 35 incoφorating the sidechain
Figure imgf000231_0002
wherein R40 is hydrogen or alkyl and R17 hydrogen or halo.
38. The compound according to claim 35 wherein R is hydrogen or methyl; R is bromo, chloro or fluoro; R18 is hydrogen or methoxy; and R19 is hydrogen .
39. A compound having the formula
Figure imgf000232_0001
wherein
R15 is hydrogen, alkyl, or, -alkyl-R25;
R25 is hydrogen, hydroxy, thiol, alkenyl, alkoxy, thioalkoxy, amino, halo, cyano, sulfoxy, sulfonyl, -NR30COOR31, -NR30C(O)R31, -NR30SO2R31, -C(O)NR30R31, heteroaryl or heterocycloalkyl ;
R16 is each independently hydrogen or methyl;
17 1 8 1 Q 18
R , R and R are, independently, hydrogen, halogen, or alkoxy; or R and R19 together form a heterocyclo or heteroaryl group; and R30 and R31 are, independently, hydrogen, alkyl, cycloalkyl or alkyl-R25.
40. The compound according to claim 39 incoφorating the sidechain
Figure imgf000233_0001
wherein R40 is hydrogen or alkyl and R17 hydrogen or halo.
41. The compound according to claim 39 wherein R15 is hydrogen, methyl, ethyl, or -(CH2)nCH2-R25 wherein n is 0, 1, or 2; and R25 is OH, OMe, F, CN, CF3, SOCH3 or SO2CH3, -NR30COR31, -NR30COOR31, -NR30SO2R31, -C(O)NR30R31, or has the formula:
Figure imgf000233_0002
wherein R >33 ' i •s hydrogen, alkyl, or substituted alkyl.
42. The compound according to claim 39 wherein R15 is ethyl, methoxyethyl, CH2CH2F, or CH2CH2CN; R17 is bromo or chloro; R18 is methoxy or hydrogen; and R19 is hydrogen.
43. A compound having the formula:
Figure imgf000234_0001
wherein
R15 is hydrogen, alkyl, aryl, or -alkyl-R25;
R25 is hydrogen, hydroxy, thiol, alkenyl, alkoxy, thioalkoxy, amino, halo, cyano, sulfoxy, sulfonyl, -NR30COOR31, -NR30C(O)R31, -NR30SO2R31, - C(O)NR30R31, heteroaryl or heterocycloalkyl;
R , R and R are, independently, hydrogen, halogen, or alkoxy; or R and R19 together form a heterocyclo or heteroaryl group; and
R and R are, independently, hydrogen, alkyl, cycloalkyl or alkyl-R .
44. The compound according to claim 43 wherein R15 is -(CH2)nCH2R25 n is 0, 1, 2 or 3 and R25 is OH, OMe, F, CN, CF3, SOCH3 or SO2CH3 , -NR30COOR31, - NR30C(O)R31, -NR30SO2R31, -C(O)NR31R32 or has the formula:
Figure imgf000234_0002
wherein R , 33 is hydrogen, alkyl, or substituted alkyl.
45. The compound according to claim 43 wherein R15 is methyl, ethyl, CH2F, methoxyethyl, CH2CH2F, or CH2CH2CH2SOCH3; R 1"7 is bromo; R , 1'8° is hydrogen, methoxy, or fluoro; and R 19 i:,s H
46. The compound according to claim 45 incoφorating the sidechain
Figure imgf000235_0001
Figure imgf000235_0002
wherein R ,40 is hydrogen or alkyl, and R 17 is hydrogen or halo.
47. A compound having the formula:
Figure imgf000235_0003
wherein each R , 15 is independently hydrogen, alkyl, aryl, or -alkyl-R 25. ; R25 is hydrogen, hydroxy, thiol, alkenyl, alkoxy, thioalkoxy, amino, halo, cyano, sulfoxy, sulfonyl, -NR30COOR31, -NR30C(O)R31, -NR30SO2R31, - C(O)NR30R31, heteroaryl or heterocycloalkyl;
R17, R18 and R19 are, independently, hydrogen, halogen, or alkoxy; or R18 and R19 together form a heterocyclo or heteroaryl group; and
R30 and R31 are, independently, hydrogen, alkyl, cycloalkyl or alkyl-R25.
48. The compound according to claim 1 selected from the group consisting of :
A compound according to claim 1 having the formula: (S)-4-(2-Hydroxy- 1 -phenyl-ethylamino)-3-(6-imidazol- 1 -yl-4-methyl- 1 H- benzimidazol-2-yl)-lH-pyridin-2-one;
(±)-4-[2-Hydroxy-2-(3-iodo-phenyl)-ethylamino]-3-(6-imidazol-l-yl-4-methyl-lH- benzimidazol-2-yl)-lH-pyridin-2-one;
(±)-4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-(6-imidazol-l-yl -4-methyl- 1H- benzimidazol-2-yl)- lH-pyridin-2-one;
(±)-4-[2-(3-Bromo-phenyl)-2-hydroxy-ethylamino]-3-(6-imidazol-l-yl-4-methyl-lH- benzimidazol-2-yl)-lH-pyridin-2-one;
(S)-4- [2-(2-Chloro-phenyl)- 1 -hydroxymethyl-ethylamino] -3-(6-imidazol- 1 -yl-4- methyl- 1 H-benzimidazol-2-yl)- 1 H-pyridin-2-one; (S)-4-[2-(3-Chloro-phenyl)-l-hydroxymethyl-ethylamino]-3-(6-imidazol-l-yl-4- methyl- 1 H-benzimidazol-2-yl)- 1 H-pyridin-2-one;
(S)-4-[2-(4-Chloro-phenyl)-l-hydroxymethyl-ethylamino]-3-(6-imidazol-l-yl-4- methyl-lH-benzimidazol-2-yl)-lH-pyridin-2-one;
(S)-4-[2-(2-Bromo-phenyl)-l-hydroxymethyl-ethylamino]-3-(6-imidazol-l-yl-4- methyl-lH-benzimidazol-2-yl)-lH-pyridin-2-one;
(S)-4-[2-(3-Bromo-phenyl)-l-hydroxymethyl-ethylamino]-3-(6-imidazol-l-yl-4- methyl-lH-benzimidazol-2-yl)-lH-pyridin-2-one;
(+)-4-( 1 -Hydroxymethyl-2-pentafluorophenyl-ethylamino)-3-(6-imidazol- 1 -yl-4- methyl- lH-benzimidazol-2-yl)- 1 H-pyridin-2-one; (S)-4-(l-Hydroxymethyl-2-pyridin-4-yl-ethylamino)-3 -(6-imidazol- 1-yl -4-methyl- 1H- benzimidazol-2-yl)- 1 H-pyridin-2-one; (S)-4-[l-Hydroxymethyl-2-(2-naphthalenyl)-ethylamino]-3-(6-imidazol-l-yl-4- methyl-lH-benzimidazol-2-yl)-lH-pyridin-2-one;
3-(6-Imidazol-l-yl -4-methyl- lH-benzimidazol-2-yl)-4-(pyridin-2-ylmethoxy)-lH- pyridin-2-one; (±)-4-[2-(3-Bromo-phenyl)-2-fluoro-ethylamino]-3-(6-imidazol-l-yl-4-methyl-lH- benzimidazol-2-yl)-lH-pyridin-2-one;
(S)-2- [4-( 1 -Hydroxymethyl-2-phenyl-ethylamino)-2-oxo- 1 ,2-dihydro-pyridin-3-yl] -7- methyl-3H-benzimidazole-5-carbonitrile;
(±)-2- {4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-2-oxo- 1 ,2-dihydro-pyridin-3- yl }-7-methyl-3H-benzimidazole-5-carbonitrile;
(S)-2- { 4-[2-(3-Chloro-phenyl)- 1 -hydroxymethyl-ethylamino]-2-oxo- 1 ,2-dihydro- pyridin-3-yl } -7-methyl-3H-benzimidazole-5-carbonitrile;
(±)-2-{4-[2-(3-Bromo-4-methoxy-phenyl)-2-hydroxy-ethylamino]-2-oxo-l,2-dihydro- pyridin-3-yl}-7-methyl-3H-benzimidazole-5-carbonitrile; (±)-2- { 4-[2-(3-Fluoro-phenyl)-2-hydroxy-ethylamino]-2-oxo- 1 ,2-dihydro-pyridin-3- yl } -7-methyl-3H-benzimidazole-5-carbonitrile;
(±)-2-{4-[2-(3-Bromo-phenyl)-2-hydroxy-ethylamino] -2-oxo- l,2-dihydro-pyridin-3- yl } -7-methyl-3H-benzimidazole-5-carbonitrile;
(S)-2-[4-(2-Hydroxy-2-phenyl-ethylamino)-2-oxo-l,2-dihydro-pyridin-3-yl]-7- methyl-3H-benzimidazole-5-carbonitrile;
(+)-3-(lH-Benzimidazol-2-yl)-4-[2-(3-bromo-phenyl)-2-hydroxy-ethylamino]-lH- pyridin-2-one;
(S)-3-(lH-Benzimidazol-2-yl)-4-(l-hydroxymethyl-2-phenyl-ethylamino)-lH- pyridin-2-one; (±)-3-(lH-Benzimidazol-2-yl)-4-[2-(3-bromo-4-methoxy-phenyl)-2-hydroxy- ethylamino] - 1 H-pyridin-2-one;
(S)-4- { 2- [4-( 1 -hydroxymethyl-2-phenyl-ethylamino)-2-oxo- 1 ,2-dihydro-pyridin-3- yl]-7-methyl-3H-benzimidazol-5-yl } -piperazine- 1 -carboxylic acid /.søpropylamide;
(S)-4- { 2-[4-( 1 -hydroxymethyl-2-phenyl-ethylamino)-2-oxo- 1 ,2-dihydro-pyridin-3-yl] -7- methyl-3H-benzimidazol-5-yl } -piperazine- 1 -carboxylic acid ethylamide;
(S)-4-( 1 -Hydroxymethyl-2-phenyl-ethylamino)-3- { 4-methyl-6- [4-( 1 -phenyl-methanoyl)- piperazin- 1 -yl] - 1 H-benzimidazol-2-yl } - 1 H-pyridin-2-one; (S)-4-(l-Hydroxymethyl-2-phenyl-ethylamino)-3-[6-(4- 5opropyl-piperazin-l-yl)-4- methyl-lH-benzimidazol-2-yl]-lH-pyridin-2-one;
(S)-3- [6-(4-Benzyl-piperazine- 1 -yl)-4-methyl- 1 H-benzimidazol-2-yl] -4-( 1 - hydroxymethyl-2-phenyl-ethylamino)- 1 H-pyridin-2-one; (±)-3-[6-(4- Acetyl-piperazine- 1 -yl)-4-methyl- 1 H-benzimidazol-2-yl] -4-[2-(3-chloro- phenyl)-2-hydroxy-ethylamino]-lH-pyridin-2-one;
(±)-4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-(4-methyl-6-piperazin-l-yl-lH- benzimidazol-2-yl) - lH-pyridin-2-one;
(+)-4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-[6-(4- 5opropyl-piperazine-l-yl)-4- methyl- 1 H-benzimidazol-2-yl] - 1 H-pyridin-2-one;
(S)-6-( 1 -Hydroxymethyl-2-phenyl-ethylamino)-5-(6-imidazol- 1 -yl-4-methyl- 1H- benzimidazol-2-yl)-3H-pyrimidin-4-one;
(S)-2-[6-Chloro-5-(6-imidazol- 1 -yl-4-methyl- lH-benzimidazol-2-yl)-pyrimidin-4- ylamino]-3-phenyl-propan-l-ol; (S)-4-(2-Hydroxy-2-phenyl-ethylamino)-3-(6- imidazol-l-yl-4-methyl-lH- benzimidazol-2-yl)- IH- pyridin-2-one;
(R)-4-(2-Hydroxy-2-phenyl-ethylamino)-3-(6-imidazol-l-yl-4-methyl-lH- benzimidazol-2-yl)-lH-pyridin-2-one;
( 1 S,2R)-4-( 1 -Hydroxy-indan-2-ylamino)-3-(6-imidazol- 1 -yl-4-methyl- 1 H- benzimidazol-2-yl)- 1 H-pyridin-2-one;
(±)-4-[2-Hydroxy-2-(3-hydroxy-phenyl)-ethylamino]-3-(6-imidazol-l-yl-4-methyl- lH-benzimidazol-2-yl)- 1 H-pyridin-2-one;
(S)-4-(2-Hydroxy-2-pyridin-2-yl-ethylamino)-3-(6-imidazol-l-yl-4-methyl-lH- benzimidazol-2-yl)- 1 H-pyridin-2-one; (+)-N-(3-{ l-Hydroxy-2-[3-(6-imidazol-l-yl-4-methyl-lH-benzimidazol-2-yl)-2-oxo-
1 ,2-dihydro-pyridin-4-ylamino] -ethyl } -phenyl)-methanesulfonamide;
(±)-4-[2-(3-Fluoro-phenyl)-2-hydroxy-ethylamino]-3-(6-imidazol-l-yl-4-methyl-lH- benzimidazol-2-yl)-lH-pyridin-2-one;
(±)-4-[2-(3-Chloro-4-fluoro-phenyl)-2-hydroxy-ethylamino]-3-(6-imidazol-l-yl-4- methyl-lH-benzimidazol-2-yl)-lH-pyridin-2-one;
(S)-4-[2-(3-Fluoro-phenyl)-l -hydroxymethyl -ethylamino]-3-(6-imidazol-l-yl -4- methyl- 1 H-benzimidazol-2-yl)- 1 H-pyri din-2 one; (±)-4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-(6-imidazol-l-yl-lH- benzimidazol-2-yl)- 1 H-pyridin-2-one;
(±)-4-[2-(3-Bromo-4-methoxy-phenyl)-2-hydroxy-ethylamino]-3-(6-imidazol-l-yl-4- methyl- 1 H-benzimidazol-2-yl)- 1 H-pyridin-2-one; (S)-4-[2-(3-Bromo-phenyl)-2-hydroxy-ethylamino]-3-(6-imidazol-l-yl-4-methyl-lH- benzimidazol-2-yl)- 1 H-pyridin-2-one;
(S)-4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-(6-imidazol-l-yl-4-methyl-lH- benzimidazol-2-yl)- 1 H-pyridin-2-one;
(R)-4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-(6-imidazol-l-yl-4-methyl-lH- benzimidazol-2-yl)- 1 H-pyridin-2-one;
(+)-4-[2-(3-Chloro-4-methoxy-phenyl)-2-hydroxy-ethylamino]-3-(6-imidazol-l-yl-4- methyl- 1 H-benzimidazol-2-yl)- 1 H-pyridin-2-one;
(±)-(2-Chloro-4- { 1 -hydroxy-2-[3-(6-imidazol- 1 -yl-4-methyl- 1 H-benzimidazol-2-yl)-
2-oxo-l,2-dihydro-pyridin-4-ylamino] -ethyl }-phenyl)-carbamic acid methyl ester; (S)-4-( 1 -Hydroxymethyl-2-phenyl-ethylamino)-3-[4-methyl-6-(4-methyl -piperazin- 1 - yl)- 1 H-benzimidazol-2-yl] - 1 H-pyridin-2-one;
(S)-4-( 1 -Hydroxymethyl-2-phenyl-ethylamino)-3-[4-methyl-6-(4-n-butyl-piperazin- 1 -yl)- lH-benzimidazol-2-yl]-lH-pyridin-2-one;
(S)-3- { 6-[4-(2-Hydroxy-ethyl)-piperazin- 1 -yl] -4-methyl- 1 H-benzimidazol-2-yl } -4-( 1 - hydroxymethyl-2-phenyl-ethylamino)- 1 H-pyridin-2-one;
(S)-4- { 2-[4-( 1 -Hydroxymethyl-2-phenyl-ethylamino)-2-oxo- 1 ,2-dihydro-pyridin-3- yl]-7-methyl-3H-benzimidazol-5-yl}-piperazine-l-carboxylic acid amide;
(±)-4- [2-(3-Chloro-phenyl)-2-hydroxy-ethylamino] -3-(4-methyl-6-piperazin- 1 -yl- 1 H- benzimidazol-2-yl)-lH-pyridin-2-one; (+)-4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-[6-(4-ethyl-piperazin-l-yl)-4- methyl- 1 H-benzimidazol-2-yl] - 1 H-pyri din-2-one;
(±)-4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-{6-[4-(2-hydroxy- ethyl)piperazin- 1 -yl] -4-methyl- 1 H-benzimidazol-2-yl } - 1 Hpyridin-2-one;
(±)-4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-(4-methyl-6-moφholin-4-yl- lH-benzimidazol-2-yl)-lH-pyridin-2-one;
(+)-4-[2-(3-Bromo-phenyl)-2-hydroxy-ethylamino]-3-(4-methyl-6-moφholin-
4-yl- 1 H-benzimidazol-2-yl)- 1H-; (±)-4-[2-(3-Bromo-4-methoxy-phenyl)-2-hydroxy-ethylamino]-3-(4-methyl-6- moφholin-4-yl- 1 H-benzimidazol-2-yl)- lH-pyridin-2-one;
(±)-4-[2-(3-Bromo-phenyl)-2-hydroxy-ethylamino]-3-{6-[4-(2-hydroxy-ethyl)- piperazin- l-yl]-4-methyl- lH-benzimidazol-2-yl } - lH-pyridin-2-one; (+)-4-[2-(3-Bromo-phenyl)-2-hydroxy-ethylamino]-3-(4-methyl-6-piperazin-
1 -yl- 1 H-benzimidazol-2-yl } - 1 H-pyridin-2-one;
(±)-4-[2-(3-Bromo-phenyl)-2-hydroxy-ethylamino]-3-(4-methyl-6-piperazin-
1 -yl- 1 H-benzimidazol-2-yl } - 1 H-pyridin-2-one;
(±)-3-[6-(4-Acetyl-piperazin-l-yl)-4-methyl-lH-benzimidazol-2-yl]-4-[2-(3-bromo- phenyl)-2-hydrox y-ethylamino]- lH-pyridin-2-one;
(S)-4-(l-hydroxymethyl-2-phenyl-ethylamino)-3-[4-methyl-6-(2-moφholin-4-yl- ethylamino)- 1 H-benzimidazol-2-yl] - 1 H-pyridin-2-one;
(±)-6-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-5-(6-imidazol-l-yl-4-methyl-lH- benzimidazol-2-yl)-3H-pyrimidin-4-one; (±)-4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-[6-(l-hydroxy-l-methyl-ethyl)-
4-methyl- 1 H-benzimidazol-2-yl] - 1 H-pyridin-2-one;
(±)-3-(6-Aminomethyl-4-methyl-lH-benzimidazol-2-yl)-4-[2-(3-chloro-phenyl)-2- hydroxy-ethylamino]-lH-pyridin-2-one;
(±)-4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-(6-hydroxymethyl-4-methyl- 1 H-benzimidazol-2-yl)- 1 H-pyri din-2-one;
(S)-4-(l-Benzyl-2-hydroxy-ethylamino)-3-(4-methyl-6-moφholin-4-yl-lH- benzimidazol-2-yl)- 1 H-pyridin-2-one; and
(S)-4-(l-Benzyl-2-hydroxy-ethylamino)-3-(4-methyl-6-piperidin-l-yl-lH- benzimidazol-2-yl)- 1 H-pyri din-2-one;
49. The compound according to claim 1 selected from the group consisting of:
(S)-4-( 1 -Benzyl-2-hydroxy-ethylamino)-3-(4-methyl-6-piperidin- 1 -yl- 1H- benzimidazol-2-yl)- 1 H-pyridin-2-one;
4-[2-(3-Chloro-4-methylsulfanyl-phenyl)-2-hydroxy-ethylamino]-3-(4-methyl-6- piperazin- 1 -yl- 1 H-benzoimidazol-2-yl)- 1 H-pyridin-2-one;
4-[2-(3-Chloro-4-fluoro-phenyl)-2-hydroxy-ethylamino]-3-(4-methyl-6-piperazin-l- yl- 1 H-benzoimidazol-2-yl)- 1 H-pyridin-2-one; 3-[4-(2-{4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-2-oxo-l,2-dihydro-pyridin-
3-yl}-7-methyl-3H-benzoimidazol-5-yl)-piperazin-l-yl]-propionitrile;
4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-{6-[4-(2-methanesulfonyl-ethyl)- piperazin- 1 -yl] -4-methyl- 1 H-benzoimidazol-2-yl } - 1 H-pyridin-2-one; 3-[4-(2-{4-[2-(3-Bromo-4-methoxy-phenyl)-2-hydroxy-ethylamino]-2-oxo-l,2- dihydro-pyridin-3-yl }-3H-benzoimidazol-5-yl)-7-methyl -piperazin- 1-yl]- propionitrile;
4-(2-{4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-2-oxo-l,2-dihydro-pyridin-3- yl } -7-methyl-3H-benzoimidazol-5-yl)-piperazine- 1 -carboxylic acid 2-fluoro-ethyl ester;
4-(2-{4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-2-oxo-l,2-dihydro-pyridin-3- yl}-7-methyl-3H-benzoimidazol-5-yl)-piperazine-l-carboxylic acid 2-methoxy-ethyl ester;
4-(2-{4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-2-oxo-l,2-dihydro-pyridin-3- yl}-7-methyl-3H-benzoimidazol-5-yl)-piperazine-l -carboxylic acid tert-butyl ester;
4-(2-{4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-2-oxo-l,2-dihydro-pyridin-3- yl}-7-methyl-3H-benzoimidazol-5-yl)-piperazine-l -carboxylic acid prop-2-ynyl ester;
4-(2-{4-[2-(3-Bromo-4-methoxy-phenyl)-2-hydroxy-ethylamino]-2-oxo-l,2-dihydro- pyridin-3-yl } -7-methyl-3H-benzoimidazol-5-yl)-piperazine- 1 -carboxylic acid tert- butyl ester;
(S)-4-(2- { 4-[2-(3-Bromo-4-methoxy-phenyl)-2-hydroxy-ethylamino]-2-oxo- 1 ,2- dihydro-pyridin-3-yl } -7-methyl-3H-benzimidazol-5-yl)-piperazine- 1 -carboxylic acid ethyl ester;
4-[2-(3-Chloro-4-methoxy-phenyl)-2-hydroxy-ethylamino]-3-{6-[4-(3-fluoro-propyl)- piperazin- 1 -yl] -4-methyl- 1 H-benzoimidazol-2-yl } - 1 H-pyridin-2-one;
4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-{6-[4-(2-fluoro-ethyl)-piperazin-l- yl] -4-methyl- 1 H-benzoimidazol-2-yl } - 1 H-pyridin-2-one;
4-[2-(3-Chloro-4-fluoro-phenyl)-2-hydroxy-ethylamino]-3-{6-[4-(3-fluoro-propyl)- piperazin- 1 -yl] -4-methyl- 1 H-benzoimidazol-2-yl } - 1 H-pyridin-2-one; 4-[2-(3-Bromo-4-methoxy-phenyl)-2-hydroxy-ethylamino]-3- { 6-[4-(3-fluoro-propyl)- piperazin- 1 -yl]-4-methyl- 1 H-benzoimidazol-2-yl } - 1 H-pyridin-2-one; 4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-{4-methyl-6-[4-(3,3,3-trifluoro- propyl)-piperazin- 1 -yl] - 1 H-benzoimidazol-2-yl } - 1 H-pyridin-2-one;
4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-{6-[4-(3-fluoro-propyl)-piperazin-
1 -yl] -4-methyl- 1 H-benzoimidazol-2-yl } - 1 H-pyridin-2-one; 4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-{4-methyl-6-[4-(3,4,4-trifluoro-but-
3-enyl)-piperazin-l-yl]-lH-benzoimidazol-2-yl}-lH-pyridin-2-one;
4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-{6-[4-(3-fluoro-2-hydroxy-propyl)- piperazin- 1 -yl] -4-methyl- 1 H-benzoimidazol-2-yl } - 1 H-pyridin-2-one;
4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-{6-[4-(2-hydroxy-2-methyl- propyl)-piperazin- 1 -yl]-4-methyl- 1 H-benzoimidazol-2-yl } - 1 H-pyridin-2-one;
(S)-4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-{6-[4-(2-hydroxy-ethyl)- piperazin- 1 -yl] -4-methyl- 1 H-benzimidazol-2-yl } - 1 H-pyridin-2-one;
(S)-4-[2-(3-Bromo-4-methoxy-phenyl)-2-hydroxy-ethylamino]-3-{6-[4-(2-hydroxy- ethyl)-piperazin- 1 -yl] -4-methyl- 1 H-benzimidazol-2-yl } - 1 H-pyri din-2-one; [4-(2- [4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-2-oxo- 1 ,2-dihydro-pyridin-3- yl } -7-methyl-3H-benzoimidazol-5-yl)-piperazin- 1 -yl] -acetonitrile;
4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-{6-[4-(4-fluoro-butyryl)-piperazin-
1 -yl] -4-methyl- 1 H-benzoimidazol-2-yl } - 1 H-pyridin-2-one;
4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-{6-[4-(2,2-difluoro-acetyl)- piperazin- 1 -yl] -4-methyl- 1 H-benzoimidazol-2-yl } - 1 H-pyridin-2-one;
4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-{6-[4-(2-methanesulfonyl-acetyl)- piperazin- 1 -yl] -4-methyl- 1 H-benzoimidazol-2-yl } - 1 H-pyridin-2-one;
3- [6-(4- Acetyl -piperazin- 1 -yl)-4-methyl- 1 H-benzoimidazol-2-yl] -4- [2-(3-chloro- phenyl)-2-hydroxy-ethylamino]-lH-pyridin-2-one; 4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-(4-methyl-6-{4-[2-(l-oxo-114- thiomoφholin-4-yl)-acetyl] -piperazin- 1-yl } - lH-benzoimidazol-2-yl)- lH-pyridin-2- one;
4- [2-(3-Chloro-phenyl)-2-hydroxy-ethylamino] -3-(6- { 4- [2-( 1 , 1 -dioxo- 116- thiomoφholin-4-yl)-acetyl] -piperazin- 1 -yl } -4-methyl- 1 H-benzoimidazol-2-yl)- 1 H- pyridin-2-one;
4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-{4-methyl-6-[4-(2-thiomoφholin- 4-yl-acetyl)-piperazin- 1 -yl]- 1 H-benzoimidazol-2-yl } - 1 H-pyri din-2-one; 4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-{6-[4-(2-methanesulfinyl-acetyl)- piperazin- 1 -yl] -4-methyl- 1 H-benzoimidazol-2-yl } - lH-pyridin-2-one;
4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-{6-[4-(2-methoxy-acetyl)- piperazin- 1 -yl]-4-methyl- 1 H-benzoimidazol-2-yl } - 1 H-pyridin-2-one; 4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-{4-methyl-6-[4-(2-methylsulfanyl- acetyl)-piperazin- 1 -yl]- 1 H-benzoimidazol-2-yl } - 1 H-pyridin-2-one;
3- { 6- [4-(2-Chloro-acetyl)-piperazin-l-yl] -4-methyl- lH-benzoimidazol-2-yl } -4-[2-(3- chloro-phenyl)-2-hydroxy-ethylamino]-lH-pyridin-2-one;
(S)-4-(2- { 4- [2-(3-Bromo-4-methoxy-phenyl)-2-hydroxy-ethylamino] -2-oxo- 1 ,2- dihydro-pyridin-3-yl } -7-methyl-3H-benzimidazol-5-yl)-piperazine- 1 -carbaldehyde;
(S)-4-(2-{4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-2-oxo-l,2-dihydro-pyridin-
3-yl}-7-methyl-3H-benzimidazol-5-yl)-piperazine-l-carbaldehyde;
(S)-4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-(4-methyl-6-moφholin-4-yl- lH-benzoimidazol-2-yl)- 1 H-pyridin-2-one; 4-[2-(3-Bromo-4-methoxy-phenyl)-2-hydroxy-ethylamino]-3-(4-methyl-6-moφholin-
4-yl-lH-benzoimidazol-2-yl)-lH-pyridin-2-one;
4-[2-(3-Chloro-4-fluoro-phenyl)-2-hydroxy-ethylamino]-3-(4-methyl-6-moφholin-4- yl- lH-benzoimidazol-2-yl)- 1 H-pyridin-2-one;
4-[2-(3-Chloro-4-methoxy-phenyl)-2-hydroxy-ethylamino]-3-(4-methyl-6-moφholin- 4-yl-lH-benzoimidazol-2-yl)-lH-pyridin-2-one;
4-[2-(7-Bromo-2,3-dihydro-benzofuran-5-yl)-2-hydroxy-ethylamino]-3-(4-methyl-6- moφholin-4-yl- 1 H-benzoimidazol-2-yl)- 1 H-pyridin-2-one;
4-[2-(3-Chloro-phenyl)-2(S)-hydroxy-ethylamino]-3-[4-methyl-6-[2(S),6(R)- dimethyl-moφholine-4-yl] - 1 H-benzoimidazol-2-yl] - 1 H-pyridine-2-one; 4-[2-(3-Bromo-4-methoxy-phenyl)-2(S)-hydroxy-ethylamino]-3-[4-methyl-6-
[2(S),6(R)-dimethyl-morpholine-4-yl]-lH-benzoimidazol-2-yl]-lH-pyridine-2-one;
4-[2-(3-Chloro-phenyl)-(S)-2-hydroxy-ethylamino]-3-{6-[(R)-2-fluoromethyl- moφholin-4-yl]-4-methyl-lH-benzimidazol-2-yl}-lH-pyridin-2-one and 4-[2-(3- chloro-phenyl)-(S)-2-hydroxy-ethylamino]-3-{6-[(S/)-2-fluoromethyl-moφholin-4-yl]- 4-methyl- 1 H-benzimidazol-2-yl } - 1 H-pyridin-2-one;
4-[2-(3-Bromo-4-methoxy-phenyl)-(S)-2-hydroxy-ethylamino]-3-{6-[(R)-2- fluoromethyl-moφholin-4-yl] -4-methyl- 1 H-benzimidazol-2-yl } - 1 H-pyridin-2-one and 4-[2-(3-bromo-4-methoxy-phenyl)-(S)-2-hydroxy-ethylamino]-3-{6-[(S;-2- fluoromethyl-moφholin-4-yl] -4-methyl - lH-benzimidazol-2-yl } - lH-pyridin-2-one; 4-[2-(3-Chloro-4-methoxy-phenyl)-(S)-2-hydroxy-ethylamino]-3-{6-[(R)-2- fluoromethyl-moφholin-4-yl]-4-methyl- lH-benzimidazol-2-yl } - lH-pyridin-2-one and 4-[2-(3-chloro-4-methoxy-phenyl)-(S)-2-hydroxy-ethylamino]-3-{6-[(S)-2- fluoromethyl-moφholin-4-yl] -4-methyl- 1 H-benzimidazol-2-yl } - 1 H-pyridin-2-one; 4-[2-(7-Bromo-2,3-dihydro-benzofuran-4-yl)-(S)-2-hydroxy-ethylamino]-3-{6-[(R/)-2- fluoromethyl-moφholin-4-yl] -4-methyl- lH-benzimidazol-2-yl } - lH-pyridin-2-one and 4-[2-(7-bromo-2,3-dihydro-benzofuran-4-yl)-(S)-2-hydroxy-ethylamino]-3-{6- [(S)-2-fluoromethyl-moφholin-4-yl] -4-methyl- lH-benzimidazol-2-yl}-lH-pyridin-2- one;
4-[2-(3-Chloro-ρhenyl)-(S)-2-hydroxy-ethylamino]-3-{6-[ R)-2-hydroxymethyl- moφholin-4-yl] -4-methyl- lH-benzimidazol-2-yl }-lH-pyridin-2-one and 4-[2-(3- chloro -phenyl)-(S)-2-hydroxy-ethylamino]-3- { 6-[(SJ-2-hydroxy-methyl-moφholin-4- yl] -4-methyl- 1 H-benzimidazol-2-yl } - 1 H-pyridin-2-one;
4-[2-(3-Bromo-4-methoxy-phenyl)-(S)-2-hydroxy-ethylamino]-3-{6-[(R)-2- hydroxymethyl-moφholin-4-yl] -4-methyl- 1 H-benzimidazol-2-yl } - 1 H-pyridin-2-one and 4-[2-(3-bromo-4-methoxy-phenyl)-(S)-2-hydroxy-ethylamino]-3- { 6-[(S)-2- hydroxy-methyl-moφholin-4-yl] -4-methyl- 1 H-benzimidazol-2-yl } - 1 H-pyridin-2-one; 4-[2-(3-Chloro-4-methoxy-phenyl)-(S)-2-hydroxy-ethylamino]-3- { 6-[(R)-2- hydroxymethyl-moφholin-4-yl]-4-methyl- 1 H-benzimidazol-2-yl } - 1 H-pyridin-2-one and 4-[2-(3-chloro-4-methoxy-phenyl)-(S)-2-hydroxy-ethylamino]-3- { 6-[(S)-2- hydroxy-methyl-moφholin-4-yl] -4-methyl- 1 H-benzimidazol-2-yl } - 1 H-pyridin-2-one; 4-[2-(3-Chloro-phenyl)-(S)-2-hydroxy-ethylamino]-3-{6-[(R 2-methyl-moφholin-4- yl] -4-methyl- lH-benzimidazol-2-yl } - lH-pyridin-2-one and 4-[2-(3-chloro-phenyl)- (S)-2-hydroxy-ethylamino]-3-{6-[(Sj-2-methyl-moφholin-4-yl]-4-methyl-lH- benzimidazol-2-yl } - 1 H-pyridin-2-one;
4-[2-(3-Bromo-4-methoxy-phenyl)-(S)-2-hydroxy-ethylamino]-3-{6-[(R)-2-methyl- moφholin-4-yl] -4-methyl- 1 H-benzimidazol-2-yl } - 1 H-pyridin-2-one and 4-[2-(3- bromo-4-methoxy-phenyl)-(S)-2-hydroxy-ethylamino]-3- { 6-[(S)-2-methyl-moφholin- 4-yl]-4-methyl- lH-benzimidazol-2-yl } - lH-pyridin-2-one; 4-[2-(3-Chloro-4-methoxy-phenyl)-(S)-2-hydroxy-ethylamino]-3-{6-[(R)-2-methyl- moφholin-4-yl] -4-methyl- lH-benzimidazol-2-yl }-lH-pyridin-2-one and 4-[2-(3- chloro-4-methoxy-phenyl)-(S)-2-hydroxy-ethylamino]-3-{6-[(S)-2-methyl-moφholin-
4- yl] -4-methyl- 1 H-benzimidazol-2-yl } - 1 H-pyridin-2-one; 4-[2-(3-Chloro-phenyl)-(S)-2-hydroxy-ethylamino]-3-{6-[ Rj-2-methoxymethyl- moφholin-4-yl]-4-methyl-lH-benzimidazol-2-yl}-lH-pyridin-2-one and 4-[2-(3- chloro -phenyl)-(S)-2-hydroxy-ethylamino]-3-{6-[(S)-2-methoxy-methyl-moφholin-
4-yl] -4-methyl- lH-benzimidazol-2-yl } - lH-pyridin-2-one;
4-[2-(3-Bromo-4-methoxy-phenyl)-(S)-2-hydroxy-ethylamino]-3-{6-[(R)-2- methoxymethyl-moφholin-4-yl] -4-methyl- 1 H-benzimidazol-2-yl } - 1 H-pyridin-2-one and 4-[2-(3-bromo-4-methoxy-phenyl)-(S)-2-hydroxy-ethylamino]-3- { 6-[(S)-2- methoxymethyl-moφholin-4-yl] -4-methyl- 1 H-benzimidazol-2-yl } - 1 H-pyridin-2-one;
4-[2-(3-Chloro-4-methoxy-ρhenyl)-(S)-2-hydroxy-ethylamino]-3-{6-[(R)-2- methoxymethyl-moφholin-4-yl] -4-methyl- 1 H-benzimidazol-2-yl } - 1 H-pyridin-2-one and 4-[2-(3-chloro-4-methoxy-phenyl)-(S)-2-hydroxy-ethylamino]-3-{6-[(S -2- methoxymethyl-moφholin-4-yl] -4-methyl- 1 H-benzimidazol-2-yl } - 1 H-pyridin-2-one;
4-[2-(3-Chloro-phenyl)-2(S)-hydroxy-ethylamino]-3-[4-methyl-6-(4-methyl- piperazin- 1 -yl)- 1 H-benzoimidazol-2-yl]- 1 H-pyridine-2-one;
4-[2-(3-Bromo-4-methoxy-phenyl)-2(S)-hydroxy-ethylamino]-3-[4-methyl-6-(4- methyl-piperazin- 1 -yl)- 1 H-benzoimidazol-2-yl] - 1 H-pyridine-2-one;
4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-{6-[4-(acetamido)- piperidin-1-yl]-
4-methyl- 1 H-benzoimidazol-2-yl } - 1 H-pyridin-2-one;
4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-{6-[4-(2-hydroxyacetamido)- piperidin- 1 -yl] -4-methyl- 1 H-benzoimidazol-2-yl } - 1 H-pyridin-2-one; 4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-{6-[4-(2-fluoroacetamido)- piperidin- 1 -yl] -4-methyl- 1 H-benzoimidazol-2-yl } - 1 H-pyridin-2-one;
4-[2-(3-Bromo-4-methoxy-phenyl)-2-hydroxy-ethylamino]-3-{6-[4-(acetamido)- piperidin- 1 -yl]-4-methyl- 1 H-benzoimidazol-2-yl } - 1 H-pyridin-2 -one;
4-[2-(3-Bromo -phenyl)-2-hydroxy-ethylamino]-3-{6-[4-(2-hydroxyacetamido)- piperidin- 1 -yl]-4-methyl- 1 H-benzoimidazol-2-yl } - 1 H-pyridin-2-one;
4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-{6-[4-(2-iluoroacetamido)- piper idi n- 1 -yl] -4-methyl- 1 H-benzoimidazol-2-yl } - 1 H-pyridin-2-one; 4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-{6-[4-(2- methoxyethoxycarbamoyl)- piperidin - 1 -yl]-4-methyl- lH-benzoimidazol-2-yl } - 1 H- pyridin-2-one;
4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-{6-[4-(methoxycarbamoyl)- piperidin -l-yl]-4-methyl-lH-benzoimidazol-2-yl}-lH-pyridin-2-one;
4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-{6-[4-(2-fluoroethoxy carbamoyl)- piperidin -l-yl]-4-methyl-lH-benzoimidazol-2-yl}-lH-pyridin-2-one;
(S)-4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-[4-methyl-6-(2-moφholin-4-yl- ethoxy)- 1 H-benzimidazol-2-yl] - 1 H-pyridin-2-one; (S)-4-[2-(3-Bromo-4-methoxy-phenyl)-2-hydroxy-ethylamino]-3-[4-methyl-6-(2- moφholin-4-yl-ethoxy)- lH-benzimidazol-2-yl]- lH-pyridin-2-one;
(S)-4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-[4-methyl-6-(2-methoxy- ethoxy)- 1 H-benzimidazol-2-yl] - 1 H-pyridin-2-one;
(S)-4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-[4-methyl-6-(2-hydroxy- ethoxy)-lH-benzimidazol-2-yl]-lH-pyridin-2-one;
(S)-4-[2-(3-Bromo-4-methoxy-phenyl)-2-hydroxy-ethylamino]-3-[4-methyl-6-(2- moφholin-4-yl-propoxy)- 1 H-benzimidazol-2-yl] - 1 H-pyridin-2-one;
(S)-4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-[4-methyl-6-(2-moφholin-4-yl- propoxy)- lH-benzimidazol-2-yl]- lH-pyridin-2-one; (S)-3-(4-Bromo-6-moφholin-4-ylmethyl-lH-benzimidazol-2-yl)-4-[2-(3-chloro- phenyl)-2-hydroxy-ethylamino]-lH-pyridin-2-one;
(S)-3-[4-Bromo-6-(4-methyl-piperazin-l-ylmethyl-lH-benzimidazol-2-yl)-4-[2-(3- chloro-phenyl)-2-hydroxy-ethylamino]-lH-pyridin-2-one;
(S)-4-[2-(3-Chloro-phenyl)-2-hydroxy-ethylamino]-3-[4-methyl-6-(4-methyl- piperazin- 1 -ylmethyl)- 1 H-benzimidazol-2-yl] - 1 H-pyridin-2-one;
4-[2-(3-Chloro-phenyl)-2(S)-hydroxy-ethylamino]-3-[4-methyl-6-(l,4,5,6- tetrahydropyrimidine- 1 -yl)- lH-benzoimidazol-2-yl] - 1 H-pyridine-2-one; and
4-[2-(4-Methoxy-3-Chloro-phenyl)-2(S)-hydroxy-ethylamino]-3-[4-methyl-6-
( 1 ,4,5 ,6-tetrahydropyrimidine- 1 -yl)- 1 H-benzoimidazol-2-yl] - 1 H-pyridine-2-one.
50. A pharmaceutical composition comprising a compound according to claim 1 and a pharmaceutically acceptable carrier.
51. The pharmaceutical composition according to claim 50 further comprising at least one other anti-cancer agent formulated as a fixed dose.
52. The pharmaceutical composition according to claim 51, wherein said anticancer agent is selected from the group consisting of: tamoxifen, toremifen, raloxifene, droloxifene, iodoxyfene, megestrol acetate, anastrozole, letrazole, borazole, exemestane, flutamide, nilutamide, bicalutamide, cyproterone acetate, goserelin acetate, luprolide, finasteride, herceptin, methotrexate, 5-fluorouracil, cytosine arabinoside, doxorubicin, daunomycin, epirubicin, idarubicin, mitomycin-C, dactinomycin, mithramycin, cisplatin, carboplatin, melphalan, chlorambucil, busulphan, cyclophosphamide, ifosfamide, nitrosoureas, thiotephan, vincristine, taxol, taxotere, etoposide, teniposide, amsacrine, irinotecan, topotecan, an epothilone, Iressa, OSI-774, angiogenesis inhibitors, EGF inhibitors, NEGF inhibitors, CDK inhibitors, Herl and Her2 inhibitors and monoclonal antibodies.
53. A method of treating a condition associated with at least one tyrosine kinase enzyme comprising administering to a mammalian species in need of such treatment an effective amount of a compound according to claim 1.
54. The method according to claim 53 wherein said tyrosine kinase enzyme is Abl, CDK's, EGF, EMT, FGF, FAK, Flk-1/KDR, HER-2, IGF-1R, IR, LCK, MET, PDGF, Src, or NEGF.
55. The method according to claim 53 further comprising administering to said mammalian species at least one other anti-cancer agent in combination with said compound.
56. The method according to claim 53 wherein the condition is cancer.
57. A method for treating cancer, comprising administering to a mammalian species in need of such treatment, a therapeutically effective amount of the composition of claim 50 or 51.
58. A method for treating proliferative diseases, comprising administering to a mammalian species in need of such treatment a therapeutically effective amount of the composition of claim 50 or 51.
PCT/US2002/009402 2001-03-28 2002-03-26 Novel tyrosine kinase inhibitors WO2002079192A1 (en)

Priority Applications (13)

Application Number Priority Date Filing Date Title
JP2002577817A JP2004534010A (en) 2001-03-28 2002-03-26 Novel tyrosine kinase inhibitors
EP02723631A EP1381598A4 (en) 2001-03-28 2002-03-26 Novel tyrosine kinase inhibitors
IL15804102A IL158041A0 (en) 2001-03-28 2002-03-26 Benzimidazole derivatives and pharmaceutical compositions containing the same
CA002442428A CA2442428A1 (en) 2001-03-28 2002-03-26 Novel tyrosine kinase inhibitors
BR0208373-6A BR0208373A (en) 2001-03-28 2002-03-26 Tyrosine kinase inhibitors
EEP200300475A EE200300475A (en) 2001-03-28 2002-03-26 Tyrosine kinase inhibitors, pharmaceutical compositions containing them, and said compounds for use in the treatment of diseases
MXPA03008690A MXPA03008690A (en) 2001-03-28 2002-03-26 Novel tyrosine kinase inhibitors.
HU0400323A HUP0400323A2 (en) 2001-03-28 2002-03-26 Tyrosine kinase inhibitors and pharmaceutical compositions containing them
KR10-2003-7012594A KR20030083016A (en) 2001-03-28 2002-03-26 Novel Tyrosine Kinase Inhibitors
SK12002003A SK12002003A3 (en) 2001-03-28 2002-03-26 Novel tyrosine kinase inhibitors
NO20034308A NO20034308L (en) 2001-03-28 2003-09-26 New tyrokinase inhibitors
IS6968A IS6968A (en) 2001-03-28 2003-09-26 New tyrosine kinase inhibitors
HR20030844A HRP20030844A2 (en) 2001-03-28 2003-10-17 Novel tyrosine kinase inhibitors

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US27932701P 2001-03-28 2001-03-28
US60/279,327 2001-03-28

Publications (1)

Publication Number Publication Date
WO2002079192A1 true WO2002079192A1 (en) 2002-10-10

Family

ID=23068491

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2002/009402 WO2002079192A1 (en) 2001-03-28 2002-03-26 Novel tyrosine kinase inhibitors

Country Status (25)

Country Link
EP (1) EP1381598A4 (en)
JP (1) JP2004534010A (en)
KR (1) KR20030083016A (en)
CN (1) CN1514833A (en)
AR (1) AR035804A1 (en)
BG (1) BG108206A (en)
BR (1) BR0208373A (en)
CA (1) CA2442428A1 (en)
CZ (1) CZ20032615A3 (en)
EE (1) EE200300475A (en)
GE (1) GEP20053660B (en)
HR (1) HRP20030844A2 (en)
HU (1) HUP0400323A2 (en)
IL (1) IL158041A0 (en)
IS (1) IS6968A (en)
MX (1) MXPA03008690A (en)
NO (1) NO20034308L (en)
PE (1) PE20021015A1 (en)
PL (1) PL373300A1 (en)
RU (1) RU2003131693A (en)
SK (1) SK12002003A3 (en)
UY (1) UY27234A1 (en)
WO (1) WO2002079192A1 (en)
YU (1) YU84603A (en)
ZA (1) ZA200307466B (en)

Cited By (42)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003106452A3 (en) * 2002-06-12 2004-04-08 Millennium Pharm Inc Antagonists of melanin concentrating hormone receptor
WO2005034868A3 (en) * 2003-10-06 2005-06-02 Bristol Myers Squibb Co Methods for treating igf1r-inhibitor induced hyperglycemia
EP1545543A2 (en) * 2002-10-02 2005-06-29 Bristol-Myers Squibb Company Novel tyrosine kinases inhibitors
WO2005085231A1 (en) * 2004-03-02 2005-09-15 Aventis Pharma S.A. Novel 4-benzimidazol-2-ylpyridazin-3-one derivatives
EP1575580A2 (en) * 2002-12-02 2005-09-21 Arqule, Inc. Method of treating cancers
EP1581539A2 (en) * 2003-01-03 2005-10-05 Bristol-Myers Squibb Company Novel tyrosine kinase inhibitors
JP2006516626A (en) * 2003-01-28 2006-07-06 スミスクライン ビーチャム コーポレーション Chemical compound
JP2007504160A (en) * 2003-08-29 2007-03-01 エグゼリクシス, インコーポレイテッド c-Kit Regulator and Method of Use
WO2007027528A2 (en) 2005-08-29 2007-03-08 Vertex Pharmaceuticals Incorporated 3,5-disubstituted pyrid-2-ones useful as inhibitors of tec family of non-.receptor tyrosine kinases
WO2007027729A1 (en) 2005-08-29 2007-03-08 Vertex Pharmaceuticals Incorporated 3, 5-disubstituted pyrid-2-ones useful as inhibitors of tec family of non-receptor tyrosine kinases
WO2007027594A1 (en) 2005-08-29 2007-03-08 Vertex Pharmaceuticals Incorporated 3,5-disubstituted pyrid-2-ones useful as inhibitors of tec family of non-receptor tyrosine kinases
US7265260B2 (en) 2002-03-01 2007-09-04 Bristol-Myers Squibb Company Transgenic non-human mammals expressing constitutively activated tyrosine kinase receptors
US7312215B2 (en) 2003-07-29 2007-12-25 Bristol-Myers Squibb Company Benzimidazole C-2 heterocycles as kinase inhibitors
WO2008022747A1 (en) * 2006-08-21 2008-02-28 F. Hoffmann-La Roche Ag Tricyclic lactam derivatives, their manufacture and use as pharmaceutical agents
WO2008025526A1 (en) * 2006-08-31 2008-03-06 F. Hoffmann-La Roche Ag Indole derivatives, their manufacture and use as pharmaceutical agents
WO2008021369A3 (en) * 2006-08-14 2008-05-29 Chembridge Res Lab Inc Tricyclic compounds and its use as tyrosine kinase modulators
US7393667B2 (en) 2005-05-31 2008-07-01 Bristol-Myers Squibb Company Stereoselective reduction process for the preparation of pyrrolotriazine compounds
WO2008144345A2 (en) 2007-05-17 2008-11-27 Bristol-Myers Squibb Company Biomarkers and methods for determining sensitivity to insulin growth factor-1 receptor modulators
WO2009024332A1 (en) * 2007-08-22 2009-02-26 F. Hoffmann-La Roche Ag Pyridoneamide derivatives as focal adhesion kinase (fak) inhibitors and their use for the treatment of cancer
WO2009093012A1 (en) * 2008-01-22 2009-07-30 Vernalis (R & D) Ltd Indolyl- pyridone derivatives having checkpoint kinase 1 inhibitory activity
JP2010531316A (en) * 2007-06-25 2010-09-24 エフ.ホフマン−ラ ロシュ アーゲー Benzimidazolamide derivatives as kinase inhibitors
US7820662B2 (en) 2004-04-02 2010-10-26 Osi Pharmaceuticals, Inc. 6,6-bicyclic ring substituted heterobicyclic protein kinase inhibitors
US7855289B2 (en) 2005-08-04 2010-12-21 Sirtris Pharmaceuticals, Inc. Sirtuin modulating compounds
US7893086B2 (en) 2007-06-20 2011-02-22 Sirtris Pharmaceuticals, Inc. Sirtuin modulating compounds
WO2011053779A2 (en) 2009-10-30 2011-05-05 Bristol-Myers Squibb Company Methods for treating cancer in patients having igf-1r inhibitor resistance
US8088928B2 (en) 2005-08-04 2012-01-03 Sirtris Pharmaceuticals, Inc. Sirtuin modulating compounds
US8093401B2 (en) 2005-08-04 2012-01-10 Sirtris Pharmaceuticals, Inc. Sirtuin modulating compounds
US8163908B2 (en) 2005-08-04 2012-04-24 Sirtris Pharmaceuticals, Inc. Sirtuin modulating compounds
US8343997B2 (en) 2008-12-19 2013-01-01 Sirtris Pharmaceuticals, Inc. Thiazolopyridine sirtuin modulating compounds
WO2013056069A1 (en) 2011-10-13 2013-04-18 Bristol-Myers Squibb Company Methods for selecting and treating cancer in patients with igf-1r/ir inhibitors
US8481733B2 (en) 2008-05-19 2013-07-09 OSI Pharmaceuticals, LLC Substituted imidazopyr- and imidazotri-azines
US8513415B2 (en) 2009-04-20 2013-08-20 OSI Pharmaceuticals, LLC Preparation of C-pyrazine-methylamines
US8575164B2 (en) 2005-12-19 2013-11-05 OSI Pharmaceuticals, LLC Combination cancer therapy
WO2016016894A1 (en) 2014-07-30 2016-02-04 Yeda Research And Development Co. Ltd. Media for culturing pluripotent stem cells
US10166215B2 (en) 2013-06-21 2019-01-01 Zenith Epigenetics Ltd. Substituted bicyclic compounds as bromodomain inhibitors
US10231953B2 (en) 2014-12-17 2019-03-19 Zenith Epigenetics Ltd. Inhibitors of bromodomains
US10292968B2 (en) 2014-12-11 2019-05-21 Zenith Epigenetics Ltd. Substituted heterocycles as bromodomain inhibitors
US10363257B2 (en) 2013-06-21 2019-07-30 Zenith Epigenetics Ltd. Bicyclic bromodomain inhibitors
US10500209B2 (en) 2013-07-31 2019-12-10 Zenith Epigenetics Ltd. Quinazolinones as bromodomain inhibitors
US10710992B2 (en) 2014-12-01 2020-07-14 Zenith Epigenetics Ltd. Substituted pyridinones as bromodomain inhibitors
WO2020152686A1 (en) 2019-01-23 2020-07-30 Yeda Research And Development Co. Ltd. Culture media for pluripotent stem cells
CN117069696A (en) * 2023-08-17 2023-11-17 中国药科大学 Double-target small molecule inhibitor and preparation method and application thereof

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2591413A1 (en) * 2004-12-16 2006-06-22 Vertex Pharmaceuticals Incorporated Pyrid-2-ones useful as inhibitors of tec family protein kinases for the treatment of inflammatory, proliferative and immunologically-mediated diseases
WO2007026720A1 (en) * 2005-08-31 2007-03-08 Taisho Pharmaceutical Co., Ltd. Ring-fused pyrazole derivative
WO2007145203A1 (en) * 2006-06-13 2007-12-21 Daiichi Fine Chemical Co., Ltd. Optically active 2-amino-1-(4-fluorophenyl)ethanol
US7816540B2 (en) * 2007-12-21 2010-10-19 Hoffmann-La Roche Inc. Carboxyl- or hydroxyl-substituted benzimidazole derivatives
DE102010001064A1 (en) * 2009-03-18 2010-09-23 Bayer Schering Pharma Aktiengesellschaft Substituted 2-acetamido-5-aryl-1,2,4-triazolones and their use
US8536180B2 (en) * 2009-05-27 2013-09-17 Abbvie Inc. Pyrimidine inhibitors of kinase activity
EA022623B1 (en) * 2010-10-06 2016-02-29 ГЛАКСОСМИТКЛАЙН ЭлЭлСи Benzimidazole derivatives as pi3 kinase inhibitors
WO2012061169A1 (en) * 2010-11-01 2012-05-10 Boehringer Ingelheim International Gmbh Benzimidazole inhibitors of leukotriene production
CN103936719A (en) * 2014-05-14 2014-07-23 中国药科大学 Preparation method and application of benzimidazoles derivatives
CA2982664A1 (en) * 2015-04-16 2016-10-20 Merck Patent Gmbh 3-(1h-benzimidazol-2-yl)-1h-pyridin-2-one derivatives

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6162804A (en) * 1997-09-26 2000-12-19 Merck & Co., Inc. Tyrosine kinase inhibitors
PL206826B1 (en) * 1999-06-23 2010-09-30 Sanofi Aventis Deutschland Substituted benzimidazoles
US7081454B2 (en) * 2001-03-28 2006-07-25 Bristol-Myers Squibb Co. Tyrosine kinase inhibitors

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
CHEMICAL ABSTRACTS, vol. 120, no. 13, 28 March 1994, Columbus, Ohio, US; abstract no. 164052R, NAWWAR ET AL.: "Some reaction of 2-(cyanomethyl)benzothiazole versus the corresponding benzimidazole derivative" page 1172; column 1; XP002954730 *
CHEMICAL ABSTRACTS, vol. 96, no. 22, 31 May 1982, Columbus, Ohio, US; abstract no. 182731B, PEDNEKAR ET AL.: "Synthesis of benzimidazole-substituted pyridone azo disperse dyes" page 87-88; XP002954731 *
CHIMIRRI ET AL.: "Synthesis and structural features of new cyclofunctionalized benzimidazoles", HETEROCYCLES, vol. 53, no. 3, 2000, pages 613 - 620, XP002954719 *
INDIAN J. CHEM., vol. 20B, no. 12, 1981, pages 1047 - 1049 *
PHOSPHORUS, SULFUR AND SILICON AND THE RELATED ELEMENTS, vol. 79, no. 1-4, 1993, pages 195 - 205 *
See also references of EP1381598A4 *

Cited By (83)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7265260B2 (en) 2002-03-01 2007-09-04 Bristol-Myers Squibb Company Transgenic non-human mammals expressing constitutively activated tyrosine kinase receptors
WO2003106452A3 (en) * 2002-06-12 2004-04-08 Millennium Pharm Inc Antagonists of melanin concentrating hormone receptor
EP1545543A4 (en) * 2002-10-02 2010-03-10 Bristol Myers Squibb Co Novel tyrosine kinases inhibitors
EP1545543A2 (en) * 2002-10-02 2005-06-29 Bristol-Myers Squibb Company Novel tyrosine kinases inhibitors
EP1575580A2 (en) * 2002-12-02 2005-09-21 Arqule, Inc. Method of treating cancers
EP1575580A4 (en) * 2002-12-02 2009-06-10 Arqule Inc Method of treating cancers
EP1581539A2 (en) * 2003-01-03 2005-10-05 Bristol-Myers Squibb Company Novel tyrosine kinase inhibitors
US7470696B2 (en) 2003-01-03 2008-12-30 Bristol-Myers Squibb Company Tyrosine kinase inhibitors
US7189716B2 (en) 2003-01-03 2007-03-13 Bristol-Myers Squibb Company Tyrosine kinase inhibitors
EP1581539A4 (en) * 2003-01-03 2007-09-19 Bristol Myers Squibb Co Novel tyrosine kinase inhibitors
JP2006516626A (en) * 2003-01-28 2006-07-06 スミスクライン ビーチャム コーポレーション Chemical compound
US7312215B2 (en) 2003-07-29 2007-12-25 Bristol-Myers Squibb Company Benzimidazole C-2 heterocycles as kinase inhibitors
US8362017B2 (en) 2003-08-29 2013-01-29 Exelixis, Inc. C-kit modulators and methods of use
JP2007504160A (en) * 2003-08-29 2007-03-01 エグゼリクシス, インコーポレイテッド c-Kit Regulator and Method of Use
EP1670416A4 (en) * 2003-10-06 2009-06-10 Bristol Myers Squibb Co Methods for treating igf1r-inhibitor induced hyperglycemia
WO2005034868A3 (en) * 2003-10-06 2005-06-02 Bristol Myers Squibb Co Methods for treating igf1r-inhibitor induced hyperglycemia
EP1670416A2 (en) * 2003-10-06 2006-06-21 Bristol-Myers Squibb Company Methods for treating igf1r-inhibitor induced hyperglycemia
JP4792455B2 (en) * 2004-03-02 2011-10-12 アバンテイス・フアルマ・エス・アー Novel 4-benzimidazol-2-ylpyridazin-3-one derivatives
WO2005085231A1 (en) * 2004-03-02 2005-09-15 Aventis Pharma S.A. Novel 4-benzimidazol-2-ylpyridazin-3-one derivatives
US7470689B2 (en) 2004-03-02 2008-12-30 Aventis Pharma S.A. 4-benzimidazol-2-ylpyridazin-3-one derivatives
US7754713B2 (en) 2004-03-02 2010-07-13 Aventis Pharma S.A. 4-benzimidazol-2-ylpyridazin-3-one derivatives
US8735405B2 (en) 2004-04-02 2014-05-27 OSI Pharmaceuticals, LLC 6,6-bicyclic ring substituted heterobicyclic protein kinase inhibitors
US8367826B2 (en) 2004-04-02 2013-02-05 OSI Pharmaceuticals, LLC 6,6-bicyclic ring substituted heterobicyclic protein kinase inhibitors
US8101613B2 (en) 2004-04-02 2012-01-24 OSI Pharmaceuticals, LLC 6,6-bicyclic ring substituted heterobicyclic protein kinase inhibitors
US8653268B2 (en) 2004-04-02 2014-02-18 OSI Pharmaceuticals, LLC 6,6-bicyclic ring substituted heterobicyclic protein kinase inhibitors
US7820662B2 (en) 2004-04-02 2010-10-26 Osi Pharmaceuticals, Inc. 6,6-bicyclic ring substituted heterobicyclic protein kinase inhibitors
US7393667B2 (en) 2005-05-31 2008-07-01 Bristol-Myers Squibb Company Stereoselective reduction process for the preparation of pyrrolotriazine compounds
US8093401B2 (en) 2005-08-04 2012-01-10 Sirtris Pharmaceuticals, Inc. Sirtuin modulating compounds
US7855289B2 (en) 2005-08-04 2010-12-21 Sirtris Pharmaceuticals, Inc. Sirtuin modulating compounds
US8088928B2 (en) 2005-08-04 2012-01-03 Sirtris Pharmaceuticals, Inc. Sirtuin modulating compounds
US8163908B2 (en) 2005-08-04 2012-04-24 Sirtris Pharmaceuticals, Inc. Sirtuin modulating compounds
US8178536B2 (en) 2005-08-04 2012-05-15 Sirtris Pharmaceuticals, Inc. Sirtuin modulating compounds
US7786130B2 (en) 2005-08-29 2010-08-31 Vertex Pharmaceuticals Incorporated Pyridones useful as inhibitors of kinases
WO2007027528A2 (en) 2005-08-29 2007-03-08 Vertex Pharmaceuticals Incorporated 3,5-disubstituted pyrid-2-ones useful as inhibitors of tec family of non-.receptor tyrosine kinases
WO2007027528A3 (en) * 2005-08-29 2007-05-03 Vertex Pharma 3,5-disubstituted pyrid-2-ones useful as inhibitors of tec family of non-.receptor tyrosine kinases
WO2007027594A1 (en) 2005-08-29 2007-03-08 Vertex Pharmaceuticals Incorporated 3,5-disubstituted pyrid-2-ones useful as inhibitors of tec family of non-receptor tyrosine kinases
US7786120B2 (en) 2005-08-29 2010-08-31 Vertex Pharmaceuticals Incorporated Pyridones useful as inhibitors of kinases
US7691885B2 (en) 2005-08-29 2010-04-06 Vertex Pharmaceuticals Incorporated Pyridones useful as inhibitors of kinases
WO2007027729A1 (en) 2005-08-29 2007-03-08 Vertex Pharmaceuticals Incorporated 3, 5-disubstituted pyrid-2-ones useful as inhibitors of tec family of non-receptor tyrosine kinases
US8575164B2 (en) 2005-12-19 2013-11-05 OSI Pharmaceuticals, LLC Combination cancer therapy
WO2008021369A3 (en) * 2006-08-14 2008-05-29 Chembridge Res Lab Inc Tricyclic compounds and its use as tyrosine kinase modulators
AU2007284542B2 (en) * 2006-08-14 2013-01-31 Chembridge Corporation Tricyclic compounds and its use as tyrosine kinase modulators
US8686005B2 (en) 2006-08-14 2014-04-01 Chembridge Corporation Tricyclic compound derivatives useful in the treatment of neoplastic diseases, inflammatory disorders and immunomodulatory disorders
US8063225B2 (en) 2006-08-14 2011-11-22 Chembridge Corporation Tricyclic compound derivatives useful in the treatment of neoplastic diseases, inflammatory disorders and immunomodulatory disorders
WO2008022747A1 (en) * 2006-08-21 2008-02-28 F. Hoffmann-La Roche Ag Tricyclic lactam derivatives, their manufacture and use as pharmaceutical agents
WO2008025526A1 (en) * 2006-08-31 2008-03-06 F. Hoffmann-La Roche Ag Indole derivatives, their manufacture and use as pharmaceutical agents
WO2008144345A2 (en) 2007-05-17 2008-11-27 Bristol-Myers Squibb Company Biomarkers and methods for determining sensitivity to insulin growth factor-1 receptor modulators
US8492328B2 (en) 2007-05-17 2013-07-23 Bristol-Myers Squibb Company Biomarkers and methods for determining sensitivity to insulin growth factor-1 receptor modulators
EP2559771A2 (en) 2007-05-17 2013-02-20 Bristol-Myers Squibb Company Biomarkers and methods for determining sensitivity to insulin growth factor-1 receptor modulators
US7893086B2 (en) 2007-06-20 2011-02-22 Sirtris Pharmaceuticals, Inc. Sirtuin modulating compounds
US8268862B2 (en) 2007-06-20 2012-09-18 Sirtris Pharmaceuticals, Inc. Sirtuin modulating compounds
US7956190B2 (en) 2007-06-25 2011-06-07 Hoffmann-La Roche Inc. Benzimidazole amido derivatives as kinase inhibitors
JP2010531316A (en) * 2007-06-25 2010-09-24 エフ.ホフマン−ラ ロシュ アーゲー Benzimidazolamide derivatives as kinase inhibitors
WO2009024332A1 (en) * 2007-08-22 2009-02-26 F. Hoffmann-La Roche Ag Pyridoneamide derivatives as focal adhesion kinase (fak) inhibitors and their use for the treatment of cancer
EP2065380A1 (en) * 2007-08-22 2009-06-03 F.Hoffmann-La Roche Ag Pyridoneamide derivatives as focal adhesion kinase (FAK) inhibitors and their use for the treatment of cancer
JP2011510055A (en) * 2008-01-22 2011-03-31 ヴァーナリス アールアンドディー リミテッド Indolyl-pyridone derivatives
US9604975B2 (en) 2008-01-22 2017-03-28 Vernalis (R&D) Ltd Indolyl-pyridone derivatives having checkpoint kinase 1 inhibitory activity
US10696652B2 (en) 2008-01-22 2020-06-30 Vernalis (R&D) Ltd. Indolyl-pyridone derivatives having checkpoint kinase 1 inhibitory activity
CN101970424B (en) * 2008-01-22 2013-06-12 弗奈利斯(R&D)有限公司 Indolylpyridone derivatives with checkpoint kinase 1 inhibitory activity
CN101970424A (en) * 2008-01-22 2011-02-09 弗奈利斯(R&D)有限公司 Indolylpyridone derivatives with checkpoint kinase 1 inhibitory activity
WO2009093012A1 (en) * 2008-01-22 2009-07-30 Vernalis (R & D) Ltd Indolyl- pyridone derivatives having checkpoint kinase 1 inhibitory activity
US8916591B2 (en) 2008-01-22 2014-12-23 Vernalis (R&D) Ltd Indolyl-pyridone derivatives having checkpoint kinase 1 inhibitory activity
EA021464B1 (en) * 2008-01-22 2015-06-30 Вернэлис (Р&Д) Лтд. Indolyl-pyridone derivatives having kinase 1 inhibitory activity, pharmaceutical composition containing same and use thereof
US8481733B2 (en) 2008-05-19 2013-07-09 OSI Pharmaceuticals, LLC Substituted imidazopyr- and imidazotri-azines
US8492401B2 (en) 2008-12-19 2013-07-23 Glaxosmithkline Llc Thiazolopyridine sirtuin modulating compounds
US8343997B2 (en) 2008-12-19 2013-01-01 Sirtris Pharmaceuticals, Inc. Thiazolopyridine sirtuin modulating compounds
US8513415B2 (en) 2009-04-20 2013-08-20 OSI Pharmaceuticals, LLC Preparation of C-pyrazine-methylamines
WO2011053779A2 (en) 2009-10-30 2011-05-05 Bristol-Myers Squibb Company Methods for treating cancer in patients having igf-1r inhibitor resistance
WO2013056069A1 (en) 2011-10-13 2013-04-18 Bristol-Myers Squibb Company Methods for selecting and treating cancer in patients with igf-1r/ir inhibitors
US11026926B2 (en) 2013-06-21 2021-06-08 Zenith Epigenetics Ltd. Substituted bicyclic compounds as bromodomain inhibitors
US10166215B2 (en) 2013-06-21 2019-01-01 Zenith Epigenetics Ltd. Substituted bicyclic compounds as bromodomain inhibitors
US10226451B2 (en) 2013-06-21 2019-03-12 Zenith Epigenetics Ltd. Substituted bicyclic compounds as bromodomain inhibitors
US10363257B2 (en) 2013-06-21 2019-07-30 Zenith Epigenetics Ltd. Bicyclic bromodomain inhibitors
US11446306B2 (en) 2013-06-21 2022-09-20 Zenith Epigenetics Ltd. Bicyclic bromodomain inhibitors
US10772892B2 (en) 2013-06-21 2020-09-15 Zenith Epigenetics Ltd. Bicyclic bromodomain inhibitors
US10500209B2 (en) 2013-07-31 2019-12-10 Zenith Epigenetics Ltd. Quinazolinones as bromodomain inhibitors
WO2016016894A1 (en) 2014-07-30 2016-02-04 Yeda Research And Development Co. Ltd. Media for culturing pluripotent stem cells
US10710992B2 (en) 2014-12-01 2020-07-14 Zenith Epigenetics Ltd. Substituted pyridinones as bromodomain inhibitors
US10292968B2 (en) 2014-12-11 2019-05-21 Zenith Epigenetics Ltd. Substituted heterocycles as bromodomain inhibitors
US10231953B2 (en) 2014-12-17 2019-03-19 Zenith Epigenetics Ltd. Inhibitors of bromodomains
WO2020152686A1 (en) 2019-01-23 2020-07-30 Yeda Research And Development Co. Ltd. Culture media for pluripotent stem cells
CN117069696A (en) * 2023-08-17 2023-11-17 中国药科大学 Double-target small molecule inhibitor and preparation method and application thereof
CN117069696B (en) * 2023-08-17 2024-04-26 中国药科大学 Double-target small molecule inhibitor and preparation method and application thereof

Also Published As

Publication number Publication date
AR035804A1 (en) 2004-07-14
PE20021015A1 (en) 2002-11-10
CA2442428A1 (en) 2002-10-10
EP1381598A1 (en) 2004-01-21
SK12002003A3 (en) 2004-10-05
IS6968A (en) 2003-09-26
EE200300475A (en) 2004-02-16
KR20030083016A (en) 2003-10-23
HRP20030844A2 (en) 2005-08-31
NO20034308D0 (en) 2003-09-26
EP1381598A4 (en) 2008-03-19
GEP20053660B (en) 2005-11-10
NO20034308L (en) 2003-11-26
JP2004534010A (en) 2004-11-11
HUP0400323A2 (en) 2005-11-28
BG108206A (en) 2004-11-30
IL158041A0 (en) 2004-03-28
RU2003131693A (en) 2005-05-10
MXPA03008690A (en) 2003-12-12
CN1514833A (en) 2004-07-21
CZ20032615A3 (en) 2004-03-17
ZA200307466B (en) 2005-01-13
YU84603A (en) 2006-03-03
UY27234A1 (en) 2002-10-31
PL373300A1 (en) 2005-08-22
BR0208373A (en) 2005-02-22

Similar Documents

Publication Publication Date Title
WO2002079192A1 (en) Novel tyrosine kinase inhibitors
EP1545543A2 (en) Novel tyrosine kinases inhibitors
US11390584B2 (en) 1-cyano-pyrrolidine compounds as USP30 inhibitors
US7189716B2 (en) Tyrosine kinase inhibitors
US7232826B2 (en) Tyrosine kinase inhibitors
US7244735B2 (en) Heterocyclic protein kinase inhibitors and uses thereof
KR20130028732A (en) 4-aminopyrimidine derivatives and their as as adenosine a2a receptor antagonists
MX2007009843A (en) Chemical compounds.
KR20060129040A (en) Imidazolo-5-yl-2-anilinopyrimidines as agents for the inhibition of cell proliferation
EP1853602A1 (en) Chemical compounds
AU2004212421A1 (en) Heteroaryl substituted pyrolls useful as inhibitors of protein kinases
MXPA04008807A (en) Derivatives of 4- (imidazol-5-yl)-2-(4-sulfoanilino) pyrimidine with cdk inhibitory activity.
CA3075477A1 (en) Novel heterocyclic compounds as cdk8/19 inhibitors
AU2002254399A1 (en) Novel tyrosine kinase inhibitors

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: P-846/03

Country of ref document: YU

AK Designated states

Kind code of ref document: A1

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

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 ZM 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 TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
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: 158041

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: PA/a/2003/008690

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 2003/07466

Country of ref document: ZA

Ref document number: 2442428

Country of ref document: CA

Ref document number: 528476

Country of ref document: NZ

Ref document number: 200307466

Country of ref document: ZA

ENP Entry into the national phase

Ref document number: 10820602

Country of ref document: BG

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 373300

Country of ref document: PL

Ref document number: 12002003

Country of ref document: SK

Ref document number: PV2003-2615

Country of ref document: CZ

Ref document number: 01548/DELNP/2003

Country of ref document: IN

Ref document number: 1-2003-500936

Country of ref document: PH

Ref document number: 1020037012594

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 2002254399

Country of ref document: AU

Ref document number: 2002577817

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: P20030844A

Country of ref document: HR

WWE Wipo information: entry into national phase

Ref document number: 5363

Country of ref document: GE

Ref document number: 7163

Country of ref document: GE

Ref document number: 1200300952

Country of ref document: VN

WWE Wipo information: entry into national phase

Ref document number: 2002723631

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 028105168

Country of ref document: CN

WWP Wipo information: published in national office

Ref document number: 2002723631

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWP Wipo information: published in national office

Ref document number: PV2003-2615

Country of ref document: CZ