WO2011140325A1 - Indazoles - Google Patents

Indazoles Download PDF

Info

Publication number
WO2011140325A1
WO2011140325A1 PCT/US2011/035340 US2011035340W WO2011140325A1 WO 2011140325 A1 WO2011140325 A1 WO 2011140325A1 US 2011035340 W US2011035340 W US 2011035340W WO 2011140325 A1 WO2011140325 A1 WO 2011140325A1
Authority
WO
WIPO (PCT)
Prior art keywords
methyl
alkyl
indazole
oxo
carboxamide
Prior art date
Application number
PCT/US2011/035340
Other languages
French (fr)
Inventor
Celine Duquenne
Neil Johnson
Steven D. Knight
Louis Lafrance
William H. Miller
Kenneth Newlander
Stuart Romeril
Meagan B. Rouse
Xinrong Tian
Sharad Kumar Verma
Original Assignee
Glaxosmithkline Llc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Glaxosmithkline Llc filed Critical Glaxosmithkline Llc
Priority to ES11778335.7T priority Critical patent/ES2534804T3/en
Priority to US13/696,436 priority patent/US8846935B2/en
Priority to JP2013509260A priority patent/JP5864546B2/en
Priority to EP11778335.7A priority patent/EP2566328B1/en
Publication of WO2011140325A1 publication Critical patent/WO2011140325A1/en
Priority to US14/458,305 priority patent/US9018382B2/en

Links

Classifications

    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/10Drugs for disorders of the urinary system of the bladder
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • 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
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • 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
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems

Definitions

  • This invention relates to substituted indazoles which inhibit EZH2 and thus are useful for inhibiting the proliferation of and/or inducing apoptosis in cancer cells.
  • Epigenetic modifications play an important role in the regulation of many cellular processes including cell proliferation, differentiation, and cell survival.
  • Global epigenetic modifications are common in cancer, and include global changes in DNA and/or histone methylation, dysregulation of non-coding RNAs and nucleosome remodeling leading to aberrant activation or inactivation of oncogenes, tumor suppressors and signaling pathways.
  • these epigenetic changes can be reversed through selective inhibition of the enzymes involved.
  • Several methylases involved in histone or DNA methylation are known to be dysregulated in cancer. Thus, selective inhibitors of particular methylases will be useful in the treatment of proliferative diseases such as cancer.
  • EZH2 (enhancer of zeste homolog 2; human EZH2 gene: Cardoso, C, et al; European J of Human Genetics, Vol. 8, No. 3 Pages 174-180, 2000) is the catalytic subunit of the Polycomb Repressor Complex 2 (PRC2) which functions to silence target genes by tri-methylating lysine 27 of histone H3 (H3K27me3).
  • Histone H3 is one of the five main histone proteins involved in the structure of chromatin in eukaryotic cells. Featuring a main globular domain and a long N-terminal tail, Histones are involved with the structure of the nucleosomes, a 'beads on a string' structure.
  • Histone proteins are highly post-translationally modified however Histone H3 is the most extensively modified of the five histones.
  • Histone H3 alone is purposely ambiguous in that it does not distinguish between sequence variants or modification state.
  • Histone H3 is an important protein in the emerging field of epigenetics. where its sequence variants and variable modification states are thought to play a role in the dynamic and long term regulation of genes.
  • Increased EZH2 expression has been observed in numerous solid tumors including those of the prostate, breast, skin, bladder, liver, pancreas, head and neck and correlates with cancer aggressiveness, metastasis and poor outcome (Varambally et al., 2002; Kleer et al., 2003; Breuer et al., 2004; Bachmann et al., 2005; Weikert et al., 2005; Sudo et al., 2005; Bachmann et al., 2006).
  • this invention relates to compounds of formula (I)
  • X and Z are selected independently from the group consisting of hydrogen, (Ci-C 8 )alkyl, (C 2 -C 8 )alkenyl, (C 2 -C 8 )alkynyl, unsubstituted or substituted (C 3 -C 8 )cycloalkyl, unsubstituted or substituted (C 3 -C 8 )cycloalkyl-(Ci-C 8 )alkyl or -(C 2 -C 8 )alkenyl, unsubstituted or substituted (C 5 - C 8 )cycloalkenyl, unsubstituted or substituted (C 5 -C 8 )cycloalkenyl-(Ci-C 8 )alkyl or -(C 2 -C 8 )alkenyl, (C 6 -Cio)bicycloalkyl, unsubstituted or substituted heterocycloalkyl, unsubstituted or substituted heterocyclo
  • Y is H or halo
  • R 1 is (C r C 8 )alkyl, (C 2 -C 8 )alkenyl, (C 2 -C 8 )alkynyl, unsubstituted or substituted (C 3 - C 8 )cycloalkyl, unsubstituted or substituted (C 3 -C 8 )cycloalkyl-(C 1 -C 8 )alkyl or -(C 2 -C 8 )alkenyl, unsubstituted or substituted (C 5 -C 8 )cycloalkenyl, unsubstituted or substituted (C 5 -C 8 )cycloalkenyl- (Ci-C 8 )alkyl or -(C 2 -C 8 )alkenyl, unsubstituted or substituted (C 6 -Cio)bicycloalkyl, unsubstituted or substituted heterocycloalkyl or -(C 2 -C 8 )alkeny
  • R 3 is hydrogen, (Ci-C 8 )alkyl, cyano, trifluoromethyl, -NR a R b , or halo;
  • R 6 is selected from the group consisting of hydrogen, halo, (Ci-Cg)alkyl, (C 2 -C 8 )alkenyl, - B(OH) 2 , substituted or unsubstituted (C 2 -C 8 )alkynyl, unsubstituted or substituted (C 3 -C 8 )cycloalkyl, unsubstituted or substituted (C 3 -C8)cycloalkyl-(Ci-Cg)alkyl, unsubstituted or substituted (C 5 - C 8 )cycloalkenyl, unsubstituted or substituted (C5-C8)cycloalkenyl-(Ci-C 8 )alkyl, (C6-Cio)bicycloalkyl, unsubstituted or substituted heterocycloalkyl, unsubstituted or substituted heterocycloalkyl-(d- C 8 )alkyl, unsubstituted
  • any (Ci-Cg)alkyl, (C 2 -C 8 )alkenyl, (C 2 -C 8 )alkynyl, cycloalkyl, cycloalkenyl, bicycloalkyl, heterocycloalkyl, aryl, or heteroaryl group is optionally substituted by 1, 2 or 3 groups independently selected from the group consisting of-0(Ci-C6)alkyl(R c )i_ 2 , -S(d- C 6 )alkyl(R c ) 1 . 2 , -(C 1 -C 6 )alkyl(R c ) 1 .
  • (Ci-C 6 )haloalkyl cyano, -COR a , -C0 2 R a ,-CONR a R b , -SR a , -SOR a , -S0 2 R a , -S0 2 NR a R b , nitro, -NR a R b , -NR a C(0)R b , -NR a C(0)NR a R b , -NR a C(0)OR a , -NR a S0 2 R b , -NR a S0 2 NR a R b , -OR a , -OC(0)R a , -OC(0)NR a R b , heterocycloalkyl, aryl, heteroaryl, aryl(Ci-C 4 )alkyl, and heteroaryl(Ci-C 4 )alkyl;
  • any aryl or heteroaryl moiety of said aryl, heteroaryl, aryl(Ci-C4)alkyl, or heteroaryl(Ci-C 4 )alkyl is optionally substituted by 1, 2 or 3 groups independently selected from the group consisting of halo, (Ci-C 6 )alkyl, (C 3 -C 8 )cycloalkyl, (C 5 -C 8 )cycloalkenyl, (C C 6 )haloalkyl, cyano, -COR a , -C0 2 R a , -CONR R b , -SR a , -SOR a , -S0 2 R a , -S0 2 NR a R b , nitro, -NR a R b , -NR a C(0)R b ,
  • R a and R b are each independently hydrogen, (Ci-Cg)alkyl, (C 2 -C 8 )alkenyl, (C 2 -C 8 )alkynyl, (C 3 -C 8 )cycloalkyl, (C 5 -C 8 )cycloalkenyl, (C 6 -Ci 0 )bicycloalkyl, heterocycloalkyl, aryl, heteroaryl, wherein said (Ci-C 8 )alkyl, (C 2 -C 8 )alkenyl, (C 2 -C 8 )alkynyl, cycloalkyl, cycloalkenyl, bicycloalkyl, heterocycloalkyl ,aryl or heteroaryl group is optionally substituted by 1, 2 or 3 groups independently selected from halo, hydroxyl, (Ci-C 4 )alkoxy, amino, (Ci-C 4 )alkylamino,
  • R a and R b taken together with the nitrogen to which they are attached represent a 5-8 membered saturated or unsaturated ring, optionally containing an additional heteroatom selected from oxygen, nitrogen, and sulfur, wherein said ring is optionally substituted by 1, 2 or 3 groups independently selected from (d-d)alkyl, (d-d)haloalkyl, amino, (d-d)alkylamino,
  • each R c is independently (d-C ⁇ alkylamino, -NR a S0 2 R b , -SOR a , -S0 2 R a , -NR a C(0)OR a , - NR a R h , or -C0 2 R a ;
  • Another aspect of the invention relates to the exemplified compounds.
  • this invention relates to a method of treating cancer.
  • compositions comprising compounds of formula (I) and pharmaceutically acceptable excipients.
  • substituted means substituted by one or more defined groups.
  • groups may be selected from a number of alternative groups the selected groups may be the same or different.
  • an “effective amount” means that amount of a drug or pharmaceutical agent that will elicit the biological or medical response of a tissue, system, animal or human that is being sought, for instance, by a researcher or clinician.
  • therapeutically effective amount means any amount which, as compared to a corresponding subject who has not received such amount, results in improved treatment, healing, prevention, or amelioration of a disease, disorder, or side effect, or a decrease in the rate of advancement of a disease or disorder.
  • the term also includes within its scope amounts effective to enhance normal physiological function.
  • alkyl refers to a straight- or branched-chain hydrocarbon radical having the specified number of carbon atoms, so for example, as used herein, the terms “Ci_C 8 alkyl” refers to an alkyl group having at least 1 and up to 8 carbon atoms respectively.
  • alkoxy as used herein means -0(Ci_C 8 alkyl) including -OCH3, -OCH 2 CH 3 and -
  • alkylthio as used herein is meant -S(Ci_C 8 alkyl) including -SCH3, -SCH 2 CH 3 and the like per the definition of alkyl above.
  • acyloxy means -OC(0)Ci.Cgalkyl and the like per the definition of alkyl above.
  • Acylamino means-N(H)C(0)Ci_C 8 alkyl and the like per the definition of alkyl above.
  • Aryloxy means -O(aryl), -0(substituted aryl), -O(heteroaryl) or -0(substituted heteroaryl).
  • Arylamino means -NH(aryl), -NH(substituted aryl), -NH(heteroaryl) or -NH(substituted heteroaryl), and the like.
  • alkenyl refers to straight or branched hydrocarbon chains containing the specified number of carbon atoms and at least 1 and up to 5 carbon-carbon double bonds. Examples include ethenyl (or ethenylene) and propenyl (or propenylene).
  • alkynyl refers to straight or branched hydrocarbon chains containing the specified number of carbon atoms and at least 1 and up to 5 carbon-carbon triple bonds. Examples include ethynyl (or ethynylene) and propynyl (or
  • Haloalkyl refers to an alkyl group group that is substituted with one or more halo substituents, suitably from 1 to 6 substituents. Haloalkyl includes trifluoromethyl.
  • cycloalkyl refers to a non-aromatic, saturated, cyclic hydrocarbon ring containing the specified number of carbon atoms. So, for example, the term “C3_Qcycloalkyl” refers to a non-aromatic cyclic hydrocarbon ring having from three to eight carbon atoms. Exemplary "C3-
  • Cgcycloalkyl groups useful in the present invention include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and cyclooctyl.
  • Cs-Cgcycloalkenyl refers to a non-aromatic monocyclic carboxycyclic ring having the specified number of carbon atoms and up to 3 carbon-carbon double bonds.
  • Cycloalkenyl includes by way of example cyclopentenyl and cyclohexenyl.
  • Cs-Cgheterocycloalkyl means a non-aromatic heterocyclic ring containing the specified number of ring atoms being, saturated or having one or more degrees of unsaturation and containing one or more heteroatom substitutions independently selected from O, S and N. Such a ring may be optionally fused to one or more other "heterocyclic" ring(s) or cycloalkyl ring(s). Examples are given herein below.
  • Aryl refers to optionally substituted monocyclic or polycarbocyclic unfused or fused groups having 6 to 14 carbon atoms and having at least one aromatic ring that complies with Huckel's Rule.
  • aryl groups are phenyl, biphenyl, naphthyl, anthracenyl, phenanthrenyl, and the like, as further illustrated below.
  • Heteroaryl means an optionally substituted aromatic monocyclic ring or polycarbocyclic fused ring system wherein at least one ring complies with Hiickel's Rule, has the specified number of ring atoms, and that ring contains at least one heteratom independently selected from N, O and S. Examples of “heteroaryl” groups are given herein below.
  • event(s) may or may not occur, and includes both event(s), which occur, and events that do not occur.
  • pharmaceutically-acceptable salts refers to salts that retain the desired biological activity of the subject compound and exhibit minimal undesired toxicological effects. These pharmaceutically-acceptable salts may be prepared in situ during the final isolation and purification of the compound, or by separately reacting the purified compound in its free acid or free base form with a suitable base or acid, respectively.
  • X and Z are selected from the group consisting of (Ci-C 8 )alkyl, (C 3 -C 8 )cycloalkyl, heterocycloalkyl, aryl, heteroaryl, -NR a R b , and -OR a ;
  • Y is H or F
  • R 1 is selected from the group consisting of (C 1 -C 8 )alkyl, (C 3 -C 8 )cycloalkyl, heterocycloalkyl, aryl, and heteroaryl;
  • R 3 is selected from the group consisting of hydrogen, (Ci-Cg)alkyl, cyano, trifluoromethyl, -
  • R 6 is selected from the group consisting of hydrogen, halo, cyano, trifluoromethyl, amino, (Ci-C 8 )alkyl, (C 3 -C 8 )cycloalkyl, aryl, heteroaryl, acylamino; (C 2 -C 8 )alkynyl, arylalkynyl, heteroarylalkynyl, -S0 2 R a , -S0 2 NR a R b , and -NR a S0 2 R b ;
  • any (Ci-C 8 )alkyl, (C 3 -C 8 )cycloalkyl, (C 2 -C 8 )alkynyl, arylalkynyl, heteroarylalkynyl group is optionally substituted by 1, 2 or 3 groups independently selected from -0(Ci- C 6 ) ky ⁇ ( c ) 2 , -S(Ci-C 6 )alkyl(R c ) 1 réelle 2 , -(C 1 -C 6 )alkyl(R c ) 1 contend 2 , (Q-C ⁇ alkyl-heterocycloalkyl, (C 3 -C 8 )cycloalkyl-heterocycloalkyl, halo, (Ci-C 6 )alkyl, (C 3 -C 8 )cycloalkyl,
  • each R c is independently (Ci-C 4 )alkylamino, -NR a S0 2 R b , -SOR a , -S0 2 R a , -NR a C(0)OR a ,
  • R a and R b are each independently hydrogen, (Ci-C 8 )alkyl, (C 2 -C 8 )alkenyl, (C 2 -C 8 )alkynyl,
  • R a and R b taken together with the nitrogen to which they are attached represent a 5-8 membered saturated or unsaturated ring, optionally containing an additional heteroatom selected from oxygen, nitrogen, and sulfur, wherein said ring is optionally substituted by 1, 2 or 3 groups independently selected from (Ci-C )alkyl, (Ci-C )haloalkyl, amino, (Ci-C )alkylamino,
  • An aryl or heteroaryl group in this particular subgroup A is selected independently from the group consisting of furan, thiophene, pyrrole, oxazole, thiazole, imidazole, pyrazole, oxadiazole, thiadiazole, triazole, tetrazole, benzofuran, benzothiophene, benzoxazole, benzothiazole, phenyl, pyridine, pyridazine, pyrimidine, pyrazine, triazine, tetrazine, quinoline, e, quinoxaline, and naphthyridine or another aryl or heteroaryl group as follows
  • A is O NH, or S; B is CH or N, and C is hydrogen or Ci-C 8 alkyl; or
  • D is N or C optionally substituted by hydrogen or Ci-C 8 alkyl
  • E is NH or CH 2 ;
  • F is O or CO; and
  • G is NH or CH 2 ; or
  • J is O S or CO;
  • Q is CH or ;
  • M is CH or N
  • L/(5) is hydrogen, halo, amino, cyano, (C C 8 )alkyl, (C 3 -C 8 )cycloalkyl, -COR a , -C0 2 R a , - CONR a R b , -CONR a NR a R b , -S0 2 R a , -S0 2 NR a R b , -NR a R b , -NR a C(0)R b ,-NR a S0 2 R b , - NR a S0 2 NR b , -NR a S0 2 NR a R b , -NR a NR a R b , -NR a NR a C(0)R b , -NR a NR a C(0)NR b , -NR a NR a C(0)NR a R b , -OR a ,
  • any (Ci-C 8 )alkyl, (C 3 -C 8 )cycloalkyl, group is optionally substituted by 1, 2 or 3 groups independently selected from (Ci-C 6 )alkyl, (C3-C 8 )cycloalkyl, (C 5 -C 8 )cycloalkenyl, (C 1 -C 6 )haloalkyl, cyano, -COR a , -C0 2 R a , -CONR a R b , -SR a , -SOR a , -S0 2 R a , -S0 2 NR a R b , nitro, -NR a R b , -NR a C(0)R b , -NR a C(0)NR a R b , -NR a C(0)OR a , -NR a S0 2 R b , -NR a S0 2 NR a R b ,
  • L/(6) is NH or CH 2 ;
  • M/(7) is hydrogen, halo, amino, cyano, (Ci-C 8 )alkyl, (C 3 -C 8 )cycloalkyl, heterocycloalkyl, -COR a , -C0 2 R a , -CONR a R b , -CONR a NR a R b , -S0 2 R a , -S0 2 NR a R b , - NR a R b , -NR a C(0)R b ,-NR a S0 2 R b , -NR a S0 2 NR b , -NR a S0 2 NR a R b , -NR a S0 2 NR a R b , -NR a S0 2 NR a R b , -NR a NR a R b , -NR a NR a C(0)R b , -
  • any (Ci-Cg)alkyl, (C 3 -C 8 )cycloalkyl, heterocycloalkyl group is optionally substituted by 1, 2 or 3 groups independently selected from (Ci-C6)alkyl, (C3-C8)cycloalkyl, (C 5 -C 8 )cycloalkenyl, (Ci-C 6 )haloalkyl, cyano, -COR a , -C0 2 R a , -CONR a R b , -SR a , -SOR a , - S0 2 R a , -S0 2 NR a R b , nitro, -NR a R b , -NR a C(0)R b , -NR a C(0)NR a R b , -NR a C(0)OR a , -NR a C(0)OR a , -NR a C(0)OR a , -NR a C
  • P is CH 2 , NH, O, or S;
  • Q/(8) is CH or N; and
  • n is 0-2; or
  • S/(9) and T(9) is C, or S/(9) is C and T(9) is N, or S/(9) is N and T/(9) is C;
  • R is hydrogen, amino, methyl, trifluoromethyl, halo
  • U is hydrogen, halo, amino, cyano, nitro, trifluoromethyl, (Ci-C 8 )alkyl, (C 3 -C 8 )cycloalkyl, - COR a , -C0 2 R a , -CONR a R b , -S0 2 R a , -S0 2 NR a R b , -NR a R b , -NR a C(0)R b ,-NR a S0 2 R b , - NR a S0 2 NR b , -NR a S0 2 NR a R b , -NR a NR a R b , -NR a NR a C(0)R b , , -OR a , 4-(lH-pyrazol-4-yl),
  • any (Ci-C 8 )alkyl, (C 3 -C 8 )cycloalkyl, group is optionally substituted by 1, 2 or
  • X and Z are selected independently from the group consisting of (C C 8 )alkyl, (C 3 - C 8 )cycloalkyl, heterocycloalkyl, aryl, heteroaryl, -NR R b , and -OR a ;
  • Y is H
  • R 1 is (Ci-Cg)alkyl, (C 3 -C 8 )cycloalkyl, or heterocycloalkyl;
  • R 3 is hydrogen, (Ci-Cg)alkyl or halo
  • R 6 is hydrogen, halo, cyano, trifluoromethyl, amino, (Ci-Cg)alkyl, (d-d)cycloalkyl;, aryl, heteroaryl, acylamino; (C 2 -C 8 )alkynyl, arylalkynyl, heteroarylalkynyl; -S0 2 R a ; -S0 2 NR a R b , or - NR a S0 2 R b ;
  • any (Ci-C 8 )alkyl, (C3-C 8 )cycloalkyl, (C 2 -Cg)alkynyl, arylalkynyl, heteroarylalkynyl group is optionally substituted by 1 , 2 or 3 groups independently selected from halo, (Ci-C 6 )alkyl, (C3-C 8 )cycloalkyl, (C 5 -C 8 )cycloalkenyl, (Ci-C6)haloalkyl, cyano, - COR a , -C0 2 R a , -CONR a R b , -SR a , -SOR a , -S0 2 R a , -S0 2 NR a R b , nitro, -NR a R b , -
  • NR a C(0)R b -NR a C(0)NR a R b , -NR a C(0)OR a , -NR a S0 2 R b , -NR a S0 2 NR a R b , -OR a , - OC(0)R a , -OC(0)NR a R b , heterocycloalkyl, aryl, heteroaryl, aryl(Ci-C 4 )alkyl, and heteroaryl(Ci-C 4 )alkyl;
  • R a and R b are each independently hydrogen, (Ci-C 8 )alkyl, (C 2 -C 8 )alkenyl, (C 2 -C 8 )alkynyl, (C 3 -C 8 )cycloalkyl, (C 5 -C 8 )cycloalkenyl, (d-do)bicycloalkyl, heterocycloalkyl, aryl, heteroaryl, wherein said (Ci-C 8 )alkyl, (C 2 -C 8 )alkenyl, (C 2 -C 8 )alkynyl, cycloalkyl, cycloalkenyl, bicycloalkyl, heterocycloalkyl ,aryl or heteroaryl group is optionally substituted by 1, 2 or 3 groups independently selected from halo, hydroxyl, (Ci-C 4 )alkoxy, amino, (C 1 -C 4 )alkylamino,
  • R a and R b taken together with the nitrogen to which they are attached represent a 5-8 membered saturated or unsaturated ring, optionally containing an additional heteroatom selected from oxygen, nitrogen, and sulfur, wherein said ring is optionally substituted by 1, 2 or 3 groups independently selected from (Ci-C )alkyl, (Ci-C )haloalkyl, amino, (Ci-C )alkylamino,
  • Aryl and heteroaryl in this definition are selected from the group consisting of furan, thiophene, pyrrole, oxazole, thiazole, imidazole, pyrazole, oxadiazole, thiadiazole, triazole, tetrazole, benzofuran, benzothiophene, benzoxazole, benzothiazole, phenyl, pyridine, pyridazine, pyrimidine, pyrazine, triazine, tetrazine, quinoline, cinnoline, quinazoline, quinoxaline, and
  • A is 0 NH, or S; B is CH or N, and C is hydrogen or Ci-Cg alkyl; or
  • D is N or C o tionally substituted by hydrogen or Ci-Cg alkyl
  • E is NH or CH 2 ;
  • F is O or CO; and
  • G is NH or CH 2 ; or
  • J is O S or CO;
  • Q is CH or N
  • M is CH or N
  • L/(5) is hydrogen, halo, amino, cyano, (Ci-Cg)alkyl, (C 3 -C 8 )cycloalkyl, -COR a , -C0 2 R a ,
  • any (Ci-Cg)alkyl, (C3-Cg)cycloalkyl, group is optionally substituted by 1 , 2 or 3 groups independently selected from (Ci-C6)alkyl, (C3-C8)cycloalkyl, (C5-Cg)cycloalkenyl, (C 1 -C 6 )haloalkyl, cyano, -COR a , -C0 2 R a , -CONR a R b , -SR a , -SOR a , -S0 2 R a , -S0 2 NR a R b , nitro, -NR a R b , -NR a C(0)R b , -NR a C(0)NR a R b , -NR a C(0)OR a , -NR a S0 2 R b , -NR a S0 2 NR a R b , -OR a ,
  • R a and R b are defined as above;
  • L/(6) is NH or CH 2 ;
  • M/(7) is hydrogen, halo, amino, cyano, (Ci-C 8 )alkyl, (C 3 -C 8 )cycloalkyl, heterocycloalkyl, -COR a , -C0 2 R a , -CONR a R b , -CONR a NR a R b , -S0 2 R a , -S0 2 NR a R b , - NR a R b , -NR a C(0)R b ,-NR a S0 2 R b , -NR a S0 2 NR b , -NR a S0 2 NR a R b , -NR a S0 2 NR a R b , -NR a S0 2 NR a R b , -NR a NR a R b , -NR a NR a C(0)R b , -
  • any (Ci-Cg)alkyl, (C3-Cg)cycloalkyl, heterocycloalkyl group is optionally substituted by 1, 2 or 3 groups independently selected from (Ci-C6)alkyl, (C3-C8)cycloalkyl, (C 5 -C 8 )cycloalkenyl, (Ci-C 6 )haloalkyl, cyano, -COR a , -C0 2 R a , -CONR a R b , -SR a , -SOR a , - S0 2 R a , -S0 2 NR a R b , nitro, -NR a R b , -NR a C(0)R b , -NR a C(0)NR a R b , -NR a C(0)OR a , -NR a C(0)OR a , -NR a C(0)OR a , -NR a C(0)OR
  • P is CH 2 , NH, O, or S;
  • Q/(8) is CH or N; and
  • n is 0-2; or
  • S/(9) and T(9) is C, or S/(9) is C and T(9) is N, or S/(9) is N and T/(9) is C;
  • R is hydrogen, amino, methyl, trifluoromethyl, halo
  • U is hydrogen, halo, amino, cyano, nitro, trifluoromethyl, (Ci-C 8 )alkyl, (C 3 -C 8 )cycloalkyl, - COR a , -C0 2 R a , -CONR a R b , -S0 2 R a , -S0 2 NR a R b , -NR a R b , -NR a C(0)R b ,-NR a S0 2 R b , - NR a S0 2 NR b , -NR a S0 2 NR a R b , -NR a NR a R b , -NR a NR a C(0)R b , , -OR a , 4-(lH-pyrazol-4-yl),
  • any (Ci-C 8 )alkyl, (C 3 -C 8 )cycloalkyl, group is optionally substituted by 1, 2 or 3 groups independently selected from (Ci-C 6 )alkyl, (C 3 -C 8 )cycloalkyl, (C 5 -C 8 )cycloalkenyl, (C 1 -C 6 )haloalkyl, cyano, -COR a , -C0 2 R a ,-CONR a R b , -SR a , -SOR a , -S0 2 R a , -S0 2 NR a R b , nitro, -NR a R b , -NR a C(0)R b , -NR a C(0)NR a R b , -NR a C(0)OR a , -NR a S0 2 R b , -NR a S0 2 NR a R b ,
  • X is methyl, ethyl, n-propyl, isopropyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, phenyl, trifluoromethyl, tetrahydropyran, hydroxymethyl, methoxymethyl, or benzyl;
  • Y is H
  • Z is methyl, ethyl, n-propyl, isopropyl, trifluoromethyl, or benzyl;
  • R 1 is isopropyl, tert-butyl, cyclobutyl, cyclopentyl, cyclohexyl, (l-methylethyl)cyclopropyl, l,l-dioxo-tetrahydrothiophene-3-yl, 1 -Me-piperidin-4-yl, tetrahydrofuran-3-yl, tetrahydropyran-4-yl, jVN-dimethyl- 1 -propanaminyl, benzyl, or 4-pyridyl;
  • R 3 is H, methyl, or Br
  • R 6 is methyl, bis(l, l-dimethylethyl), bis(l-methylethyl), cyclopropyl, propyl, dimethylamino, ethylamino, (2-hydroxyethyl)amino, 2-propen-l-ylamino, 1 -piperazinyl, 1-piperidinyl, 4-morpholinyl, 4-piperidinylamino, tetrahydro-2H-pyran-4-ylamino, phenylamino, (phenylmethyl)amino, (4- pyridinylmethyl)amino, [2-(2-pyridinylamino)ethyl]amino, 2-(dimethylamino)ethyl] amino, 4- pyridinylamino , 4-(aminocarbonyl)phenyl]amino, 3 -hydroxy-3 -methyl- 1-butyn-l-yl, 4- pyridinylethyny
  • this invention also relates to the exemplified compounds.
  • co-administering and derivatives thereof as used herein is meant either simultaneous administration or any manner of separate sequential administration of one or more additional pharmaceutically active compounds, whether for treating cancer, the side effects of cancer or cancer therapy, or some other disease.
  • the compounds are administered in a close time proximity to each other.
  • the compounds are administered in the same dosage form, e.g. one compound may be administered topically and another compound may be administered orally.
  • compounds according to Formula I may contain an acidic functional group, one acidic enough to form salts.
  • Representative salts include pharmaceutically-acceptable metal salts such as sodium, potassium, lithium, calcium, magnesium, aluminum, and zinc salts;
  • carbonates and bicarbonates of a pharmaceutically-acceptable metal cation such as sodium, potassium, lithium, calcium, magnesium, aluminum, and zinc
  • pharmaceutically-acceptable organic primary, secondary, and tertiary amines including aliphatic amines, aromatic amines, aliphatic diamines, and hydroxy alkylamines such as methylamine, ethylamine, 2-hydroxyethylamine, diethylamine, triethylamine, ethylenediamine, ethanolamine, diethanolamine, and cyclohexylamine.
  • compounds according to Formula (I) may contain a basic functional group and are therefore capable of forming pharmaceutically-acceptable acid addition salts by treatment with a suitable acid.
  • Suitable acids include pharmaceutically-acceptable inorganic acids and pharmaceutically-acceptable organic acids.
  • Representative pharmaceutically-acceptable acid addition salts include hydrochloride, hydrobromide, nitrate, methylnitrate, sulfate, bisulfate, sulfamate, phosphate acetate, hydroxyacetate, phenylacetate, propionate, butyrate, isobutyrate, valerate, maleate, hydroxymaleate, acrylate, fumarate, malate, tartrate, citrate, salicylate, />-aminosalicyclate, glycollate, lactate, heptanoate, phthalate, oxalate, succinate, benzoate, o-acetoxybenzoate, chlorobenzoate, methylbenzoate, dinitrobenzoate, hydroxybenzoate, methoxybenzoate, mandelate, tannate, formate, stearate, ascorbate, palmitate, oleate, pyruvate, pamoate, malonate, laurate, glutarate, glutamate
  • the compounds of formula (I) may be prepared in crystalline or non-crystalline form, and, if crystalline, may optionally be solvated, e.g. as the hydrate.
  • This invention includes within its scope stoichiometric solvates (e.g. hydrates) as well as compounds containing variable amounts of solvent (e.g. water).
  • Certain of the compounds described herein may contain one or more chiral atoms, or may otherwise be capable of existing as two enantiomers.
  • the compounds claimed below include mixtures of enantiomers as well as purified enantiomers or enantiomerically enriched mixtures. Also included within the scope of the invention are the individual isomers of the compounds represented by formula (I), or claimed below, as well as any wholly or partially equilibrated mixtures thereof.
  • the present invention also covers the individual isomers of the claimed compounds as mixtures with isomers thereof in which one or more chiral centers are inverted.
  • compositions which includes a compound of formula (I) and salts, solvates and the like, and one or more pharmaceutically acceptable carriers, diluents, or excipients.
  • the compounds of formula (I) and salts, solvates, etc, are as described above.
  • the carrier(s), diluent(s) or excipient(s) must be acceptable in the sense of being compatible with the other ingredients of the formulation and not deleterious to the recipient thereof.
  • a process for the preparation of a pharmaceutical formulation including admixing a compound of the formula (I), or salts, solvates etc, with one or more pharmaceutically acceptable carriers, diluents or excipients.
  • prodrugs for compounds of the invention include: esters, carbonate esters, hemi- esters, phosphate esters, nitro esters, sulfate esters, sulfoxides, amides, carbamates, azo-compounds, phosphamides, glycosides, ethers, acetals and ketals.
  • the compounds and compositions of the invention are used to treat cellular proliferation diseases.
  • Disease states which can be treated by the methods and compositions provided herein include, but are not limited to, cancer (further discussed below), autoimmune disease, fungal disorders, arthritis, graft rejection, inflammatory bowel disease, proliferation induced after medical procedures, including, but not limited to, surgery, angioplasty, and the like. It is appreciated that in some cases the cells may not be in a hyper or hypo proliferation state (abnormal state) and still requires treatment. For example, during wound healing, the cells may be proliferating "normally", but proliferation enhancement may be desired.
  • the invention herein includes application to cells or individuals afflicted or impending affliction with any one of these disorders or states.
  • compositions and methods provided herein are particularly deemed useful for the treatment of cancer including tumors such as prostate, breast, brain, skin, cervical carcinomas, testicular carcinomas, etc. They are particularly useful in treating metastatic or malignant tumors. More particularly, cancers that may be treated by the compositions and methods of the invention include, but are not limited to tumor types such as astrocytic, breast, cervical, colorectal, endometrial, esophageal, gastric, head and neck, hepatocellular, laryngeal, lung, oral, ovarian, prostate and thyroid carcinomas and sarcomas.
  • tumor types such as astrocytic, breast, cervical, colorectal, endometrial, esophageal, gastric, head and neck, hepatocellular, laryngeal, lung, oral, ovarian, prostate and thyroid carcinomas and sarcomas.
  • these compounds can be used to treat: Cardiac: sarcoma (angiosarcoma, fibrosarcoma, rhabdomyosarcoma, liposarcoma), myxoma, rhabdomyoma, fibroma, lipoma and teratoma; Lung: bronchogenic carcinoma (squamous cell, undifferentiated small cell, undifferentiated large cell, adenocarcinoma), alveolar (bronchiolar) carcinoma, bronchial adenoma, sarcoma, lymphoma, chondromatous hamartoma, mesothelioma; Gastrointestinal: esophagus (squamous cell carcinoma, adenocarcinoma, leiomyosarcoma, lymphoma), stomach (carcinoma, lymphoma, leiomyosarcoma), pancreas (ductal adenocarcinoma, insulinom
  • kidney adenocarcinoma, Wilm's tumor (nephroblastoma), lymphoma, leukemia), bladder and urethra (squamous cell carcinoma, transitional cell carcinoma, adenocarcinoma), prostate (adenocarcinoma, sarcoma), testis (seminoma, teratoma, embryonal carcinoma, teratocarcinoma, choriocarcinoma, sarcoma, interstitial cell carcinoma, fibroma, fibroadenoma, adenomatoid tumors, lipoma); Liver: hepatoma (hepatocellular carcinoma), cholangiocarcinoma, hepatoblastoma, angiosarcoma, hepatocellular adenoma, heman
  • Hematologic blood (myeloid leukemia (acute and chronic), acute lymphoblastic leukemia, chronic lymphocytic leukemia, myeloproliferative diseases, multiple myeloma, myelodysplastic syndrome), Hodgkin's disease, non-Hodgkin's lymphoma (malignant lymphoma); Skin: malignant melanoma, basal cell carcinoma, squamous cell carcinoma, Karposi's sarcoma, moles dysplastic nevi, lipoma, angioma, dermatofibroma, keloids, psoriasis; and Adrenal glands: neuroblastoma.
  • the term "cancerous cell” as provided herein, includes a cell afflicted by any one or related of the above identified conditions.
  • Combination therapies according to the invention comprise the administration of at least one compound of the invention and the use of at least one other treatment method, fn one embodiment, combination therapies according to the invention comprise the administration of at least one compound of the invention and surgical therapy. In one embodiment, combination therapies according to the invention comprise the administration of at least one compound of the invention and radiotherapy. In one embodiment, combination therapies according to the invention comprise the administration of at least one compound of the invention and at least one supportive care agent (e.g., at least one anti-emetic agent).
  • at least one supportive care agent e.g., at least one anti-emetic agent
  • combination therapies according to the present invention comprise the administration of at least one compound of the invention and at least one other chemotherapeutic agent.
  • the invention comprises the administration of at least one compound of the invention and at least one anti-neoplastic agent.
  • the invention comprises a therapeutic regimen where the EZH2 inhibitors of this disclosure are not in and of themselves active or significantly active, but when combined with another therapy, which may or may not be active as a standalone therapy, the combination provides a useful therapeutic outcome.
  • co-administering and derivatives thereof as used herein is meant either simultaneous administration or any manner of separate sequential administration of an EZH2 inhibiting compound, as described herein, and a further active ingredient or ingredients, known to be useful in the treatment of cancer, including chemotherapy and radiation treatment.
  • further active ingredient or ingredients includes any compound or therapeutic agent known to or that demonstrates advantageous properties when administered to a patient in need of treatment for cancer.
  • the compounds are administered in a close time proximity to each other.
  • the compounds are administered in the same dosage form, e.g. one compound may be administered topically and another compound may be administered orally.
  • any anti-neoplastic agent that has activity versus a susceptible tumor being treated may be co-administered in the treatment of specified cancers in the present invention.
  • examples of such agents can be found in Cancer Principles and Practice of Oncology by V.T. Devita and S.
  • Typical anti- neoplastic agents useful in the present invention include, but are not limited to, anti-microtubule agents such as diterpenoids and vinca alkaloids; platinum coordination complexes; alkylating agents such as nitrogen mustards, oxazaphosphorines, alkylsulfonates, nitrosoureas, and triazenes; antibiotic agents such as anthracyclins, actinomycins and bleomycins; topoisomerase II inhibitors such as epipodophyllotoxins; antimetabolites such as purine and pyrimidine analogues and anti-folate compounds; topoisomerase I inhibitors such as camptothecins; hormones and hormonal analogues; DNA methyltransferase inhibitor
  • proapoptotic agents include proapoptotic agents; and cell cycle signaling inhibitors.
  • any chemotherapeutic agent that has activity against a susceptible neoplasm being treated may be utilized in combination with the compounds the invention, provided that the particular agent is clinically compatible with therapy employing a compound of the invention.
  • Typical anti- neoplastic agents useful in the present invention include, but are not limited to: alkylating agents, antimetabolites, antitumor antibiotics, antimitotic agents, nucleoside analogues, topoisomerase I and II inhibitors, hormones and hormonal analogues; retinoids, histone deacetylase inhibitors; signal transduction pathway inhibitors including inhibitors of cell growth or growth factor function, angiogenesis inhibitors, and serine/threonine or other kinase inhibitors; cyclin dependent kinase inhibitors; antisense therapies and immunotherapeutic agents, including monoclonals, vaccines or other biological agents.
  • Nucleoside analogues are those compounds which are converted to deoxynucleotide triphosphates and incorporated into replicating DNA in place of cytosine. DNA methyltransferases become covalently bound to the modified bases resulting in an inactive enzyme and reduced DNA methylation.
  • Examples of nucleoside analogues include azacitidine and decitabine which are used for the treatment of myelodysplastic disorder.
  • Histone deacetylase (HDAC) inhibitors include vorinostat, for the treatment of cutaneous T-cell lymphoma. HDACs modify chromatin through the deactylation of histones. In addition, they have a variety of substrates including numerous transcription factors and signaling molecules. Other HDAC inhibitors are in development.
  • Signal transduction pathway inhibitors are those inhibitors which block or inhibit a chemical process which evokes an intracellular change. As used herein this change is cell proliferation or differentiation or survival.
  • Signal transduction pathway inhibitors useful in the present invention include, but are not limited to, inhibitors of receptor tyrosine kinases, non-receptor tyrosine kinases, SH2/SH3 domain blockers, serine/threonine kinases, phosphatidyl inositol-3-OH kinases, myoinositol signaling, and Ras oncogenes. Signal transduction pathway inhibitors may be employed in combination with the compounds of the invention in the compositions and methods described above.
  • Receptor kinase angiogenesis inhibitors may also find use in the present invention.
  • Inhibitors of angiogenesis related to VEGFR and TIE-2 are discussed above in regard to signal transduction inhibitors (both are receptor tyrosine kinases).
  • Other inhibitors may be used in combination with the compounds of the invention.
  • anti-VEGF antibodies which do not recognize VEGFR (the receptor tyrosine kinase), but bind to the ligand; small molecule inhibitors of integrin (alpha v beta 3 ) that inhibit angiogenesis; endostatin and angiostatin (non-RTK) may also prove useful in combination with the compounds of the invention.
  • VEGFR antibody is bevacizumab (AVASTIN ® ).
  • Trastuzumab (Herceptin ® ) is an example of an anti-erbB2 antibody inhibitor of growth factor function.
  • An anti- erbB 1 antibody inhibitor of growth factor function is cetuximab (ErbituxTM, C225).
  • Bevacizumab (Avastin ) is an example of a monoclonal antibody directed against VEGFR.
  • small molecule inhibitors of epidermal growth factor receptors include but are not limited to lapatinib (TykerbTM) and erlotinib (TARCEVA ® ).
  • Imatinib mesylate is one example of a PDGFR inhibitor.
  • VEGFR inhibitors include pazopanib, ZD6474, AZD2171, PT 787, sunitinib and sorafenib.
  • Anti-microtubule or anti-mitotic agents are phase specific agents active against the microtubules of tumor cells during M or the mitosis phase of the cell cycle.
  • anti-microtubule agents include, but are not limited to, diterpenoids and vinca alkaloids.
  • Diterpenoids which are derived from natural sources, are phase specific anti -cancer agents that operate at the G2/M phases of the cell cycle. It is believed that the diterpenoids stabilize the ⁇ - tubulin subunit of the microtubules, by binding with this protein. Disassembly of the protein appears then to be inhibited with mitosis being arrested and cell death following. Examples of diterpenoids include, but are not limited to, paclitaxel and its analog docetaxel.
  • Paclitaxel, 5P,20-epoxy-l,2a,4,7P,10p,13a-hexa-hydroxytax-l l-en-9-one 4,10-diacetate 2- benzoate 13-ester with (2R,3S)-N-benzoyl-3-phenylisoserine; is a natural diterpene product isolated from the Pacific yew tree Taxus brevifolia and is commercially available as an injectable solution TAXOL ® . It is a member of the taxane family of terpenes. It was first isolated in 1971 by Wani et al. J. Am. Chem, Soc, 93:2325.
  • Paclitaxel has been approved for clinical use in the treatment of refractory ovarian cancer in the United States (Markman et al., Yale Journal of Biology and Medicine, 64:583, 1991 ; McGuire et al., Ann. Intern, Med., I l l :273,1989) and for the treatment of breast cancer (Holmes et al., J. Nat. Cancer Inst., 83 : 1797, 1991.) It is a potential candidate for treatment of neoplasms in the skin (Einzig et. al., Proc. Am. Soc. Clin. Oncol., 20:46) and head and neck carcinomas (Forastire et. al., Sem. Oncol., 20:56, 1990).
  • the compound also shows potential for the treatment of polycystic kidney disease (Woo et. al., Nature, 368:750. 1994), lung cancer and malaria.
  • Treatment of patients with paclitaxel results in bone marrow suppression (multiple cell lineages, Ignoff, R.J. et. al, Cancer Chemotherapy Pocket Guide,, 1998) related to the duration of dosing above a threshold concentration (50nM) (Kearns, CM. et. al., Seminars in Oncology, 3(6) p.16-23, 1995).
  • Docetaxel (2R,3S)- N-carboxy-3-phenylisoserine,N-ter?-buryl ester, 13-ester with 5 ⁇ -20- epoxy- l,2a,4,7P, 10P,13a-hexahydroxytax-l l-en-9-one 4-acetate 2-benzoate, trihydrate; is commercially available as an injectable solution as TAXOTERE ® .
  • Docetaxel is indicated for the treatment of breast cancer.
  • Docetaxel is a semisynthetic derivative of paclitaxel q.v., prepared using a natural precursor, 10-deacetyl-baccatin III, extracted from the needle of the European Yew tree. The dose limiting toxicity of docetaxel is neutropenia.
  • Vinca alkaloids are phase specific anti-neoplastic agents derived from the periwinkle plant. Vinca alkaloids act at the M phase (mitosis) of the cell cycle by binding specifically to tubulin. Consequently, the bound tubulin molecule is unable to polymerize into microtubules. Mitosis is believed to be arrested in metaphase with cell death following. Examples of vinca alkaloids include, but are not limited to, vinblastine, vincristine, and vinorelbine.
  • Vinblastine vincaleukoblastine sulfate
  • VELBAN ® an injectable solution
  • Myelosuppression is the dose limiting side effect of vinblastine.
  • Vincristine vincaleukoblastine, 22-oxo-, sulfate
  • ONCOVIN ® an injectable solution.
  • Vincristine is indicated for the treatment of acute leukemias and has also found use in treatment regimens for Hodgkin's and non-Hodgkin's malignant lymphomas. Alopecia and neurologic effects are the most common side effect of vincristine and to a lesser extent
  • Vinorelbine 3',4'-didehydro -4'-deoxy-C'-norvincaleukoblastine [R-(R*,R*)-2,3- dihydroxybutanedioate (1 :2)(salt)], commercially available as an injectable solution of vinorelbine tartrate (NAVELBINE ® ), is a semisynthetic vinca alkaloid.
  • Vinorelbine is indicated as a single agent or in combination with other chemotherapeutic agents, such as cisplatin, in the treatment of various solid tumors, particularly non-small cell lung, advanced breast, and hormone refractory prostate cancers. Myelosuppression is the most common dose limiting side effect of vinorelbine.
  • Platinum coordination complexes are non-phase specific anti-cancer agents, which are interactive with DNA.
  • the platinum complexes enter tumor cells, undergo, aquation and form intra- and interstrand crosslinks with DNA causing adverse biological effects to the tumor.
  • Examples of platinum coordination complexes include, but are not limited to, cisplatin and carboplatin.
  • Cisplatin cis-diamminedichloroplatinum
  • PLATINOL ® an injectable solution.
  • Cisplatin is primarily indicated in the treatment of metastatic testicular and ovarian cancer and advanced bladder cancer.
  • the primary dose limiting side effects of cisplatin are nephrotoxicity, which may be controlled by hydration and diuresis, and ototoxicity.
  • Carboplatin, platinum, diammine [l,l-cyclobutane-dicarboxylate(2-)-0,0'] is commercially available as PARAPLATIN ® as an injectable solution.
  • Carboplatin is primarily indicated in the first and second line treatment of advanced ovarian carcinoma. Bone marrow suppression is the dose limiting toxicity of carboplatin.
  • Alkylating agents are non-phase anti-cancer specific agents and strong electrophiles.
  • alkylating agents form covalent linkages, by alkylation, to DNA through nucleophilic moieties of the DNA molecule such as phosphate, amino, sulfhydryl, hydroxyl, carboxyl, and imidazole groups. Such alkylation disrupts nucleic acid function leading to cell death.
  • alkylating agents include, but are not limited to, nitrogen mustards such as cyclophosphamide, melphalan, and chlorambucil; alkyl sulfonates such as busulfan; nitrosoureas such as carmustine; and triazenes such as dacarbazine.
  • Cyclophosphamide 2-[bis(2-chloroethyl)amino]tetrahydro-2H-l,3,2-oxazaphosphorine 2- oxide monohydrate, is commercially available as an injectable solution or tablets as CYTOXAN ® . Cyclophosphamide is indicated as a single agent or in combination with other chemotherapeutic agents, in the treatment of malignant lymphomas, multiple myeloma, and leukemias. Alopecia, nausea, vomiting and leukopenia are the most common dose limiting side effects of
  • Melphalan 4-[bis(2-chloroethyl)amino]-L-phenylalanine, is commercially available as an injectable solution or tablets as ALKERAN ® .
  • Melphalan is indicated for the palliative treatment of multiple myeloma and non-resectable epithelial carcinoma of the ovary. Bone marrow suppression is the most common dose limiting side effect of melphalan.
  • Chlorambucil 4-[bis(2-chloroethyl)amino]benzenebutanoic acid, is commercially available as LEUKERAN ® tablets. Chlorambucil is indicated for the palliative treatment of chronic lymphatic leukemia, and malignant lymphomas such as lymphosarcoma, giant follicular lymphoma, and Hodgkin's disease. Bone marrow suppression is the most common dose limiting side effect of chlorambucil.
  • Busulfan 1,4-butanediol dimethanesulfonate, is commercially available as MYLERAN ® TABLETS. Busulfan is indicated for the palliative treatment of chronic myelogenous leukemia. Bone marrow suppression is the most common dose limiting side effects of busulfan.
  • Carmustine, l,3-[bis(2-chloroethyl)-l -nitrosourea, is commercially available as single vials of lyophilized material as BiCNU ® .
  • Carmustine is indicated for the palliative treatment as a single agent or in combination with other agents for brain tumors, multiple myeloma, Hodgkin's disease, and non- Hodgkin's lymphomas. Delayed myelosuppression is the most common dose limiting side effects of carmustine.
  • dacarbazine, 5-(3,3-dimethyl-l-triazeno)-imidazole-4-carboxamide, is commercially available as single vials of material as DTIC-Dome ® .
  • dacarbazine is indicated for the treatment of metastatic malignant melanoma and in combination with other agents for the second line treatment of Hodgkin's Disease. Nausea, vomiting, and anorexia are the most common dose limiting side effects of dacarbazine.
  • Antibiotic anti-neoplastics are non-phase specific agents, which bind or intercalate with DNA. Typically, such action results in stable DNA complexes or strand breakage, which disrupts ordinary function of the nucleic acids leading to cell death.
  • antibiotic anti-neoplastic agents include, but are not limited to, actinomycins such as dactinomycin, anthrocyclins such as
  • daunorubicin and doxorubicin are excreted by doxorubicin and bleomycins.
  • Dactinomycin also know as Actinomycin D, is commercially available in injectable form as COSMEGEN ® . Dactinomycin is indicated for the treatment of Wilm's tumor and
  • Daunorubicin (8S-cis-)-8-acetyl- 10-[(3-amino-2,3,6-trideoxy-a-L-lyxo-hexopyranosyl)oxy]-
  • Daunorubicin is indicated for remission induction in the treatment of acute nonlymphocytic leukemia and advanced HIV associated Kaposi's sarcoma. Myelosuppression is the most common dose limiting side effect of daunorubicin.
  • Doxorubicin (8S, 10S)-10-[(3-amino-2,3,6-trideoxy-a-L-lyxo-hexopyranosyl)oxy]-8- glycoloyl, 7,8,9, 10-tetrahydro-6,8,l l-trihydroxy- l-methoxy-5, 12 naphthacenedione hydrochloride, is commercially available as an injectable form as RUBEX ® or ADRIAMYCIN RDF ® .
  • Doxorubicin is primarily indicated for the treatment of acute lymphoblastic leukemia and acute myeloblastic leukemia, but is also a useful component in the treatment of some solid tumors and lymphomas. Myelosuppression is the most common dose limiting side effect of doxorubicin.
  • Bleomycin a mixture of cytotoxic glycopeptide antibiotics isolated from a strain of
  • Streptomyces verticillus is commercially available as BLENOXANE ® .
  • Bleomycin is indicated as a palliative treatment, as a single agent or in combination with other agents, of squamous cell carcinoma, lymphomas, and testicular carcinomas. Pulmonary and cutaneous toxicities are the most common dose limiting side effects of bleomycin.
  • Topoisomerase II inhibitors include, but are not limited to, epipodophyllotoxins.
  • Epipodophyllotoxins are phase specific anti-neoplastic agents derived from the mandrake plant. Epipodophyllotoxins typically affect cells in the S and G 2 phases of the cell cycle by forming a ternary complex with topoisomerase II and DNA causing DNA strand breaks. The strand breaks accumulate and cell death follows. Examples of epipodophyllotoxins include, but are not limited to, etoposide and teniposide.
  • Etoposide 4'-demethyl-epipodophyllotoxin 9[4,6-0-(R )-ethylidene-P-D-glucopyranoside] is commercially available as an injectable solution or capsules as VePESID ® and is commonly known as VP- 16.
  • Etoposide is indicated as a single agent or in combination with other chemotherapy agents in the treatment of testicular and non-small cell lung cancers. Myelosuppression is the most common side effect of etoposide. The incidence of leucopenia tends to be more severe than thrombocytopenia.
  • Teniposide 4'-demethyl-epipodophyllotoxin 9[4,6-0-(R )-thenylidene- -D-glucopyranoside], is commercially available as an injectable solution as VUMON ® and is commonly known as VM-26.
  • Teniposide is indicated as a single agent or in combination with other chemotherapy agents in the treatment of acute leukemia in children. Myelosuppression is the most common dose limiting side effect of teniposide. Teniposide can induce both leucopenia and thrombocytopenia.
  • Antimetabolite neoplastic agents are phase specific anti-neoplastic agents that act at S phase (DNA synthesis) of the cell cycle by inhibiting DNA synthesis or by inhibiting purine or pyrimidine base synthesis and thereby limiting DNA synthesis. Consequently, S phase does not proceed and cell death follows.
  • Examples of antimetabolite anti-neoplastic agents include, but are not limited to, fluorouracil, methotrexate, cytarabine, mecaptopurine, thioguanine, and gemcitabine.
  • 5-fluorouracil 5-fluoro-2,4- (1H,3H) pyrimidinedione
  • fluorouracil is commercially available as fluorouracil.
  • Administration of 5-fluorouracil leads to inhibition of thymidylate synthesis and is also incorporated into both RNA and DNA. The result typically is cell death.
  • 5-fluorouracil is indicated as a single agent or in combination with other chemotherapy agents in the treatment of carcinomas of the breast, colon, rectum, stomach and pancreas. Myelosuppression and mucositis are dose limiting side effects of 5-fluorouracil.
  • Other fluoropyrimidine analogs include 5-fluoro deoxyuridine (floxuridine) and 5-fluorodeoxyuridine monophosphate.
  • Cytarabine 4-amino- l-P-D-arabinofuranosyl-2 (lH)-pyrimidinone, is commercially available as CYTOSAR-U ® and is commonly known as Ara-C. It is believed that cytarabine exhibits cell phase specificity at S-phase by inhibiting DNA chain elongation by terminal incorporation of cytarabine into the growing DNA chain. Cytarabine is indicated as a single agent or in combination with other chemotherapy agents in the treatment of acute leukemia. Other cytidine analogs include 5-azacytidine and 2',2'-difluorodeoxycytidine (gemcitabine). Cytarabine induces leucopenia, thrombocytopenia, and mucositis.
  • Mercaptopurine 1 ,7-dihydro-6H-purine-6-thione monohydrate, is commercially available as PURTNETHOL ® .
  • Mercaptopurine exhibits cell phase specificity at S-phase by inhibiting DNA synthesis by an as of yet unspecified mechanism.
  • Mercaptopurine is indicated as a single agent or in combination with other chemotherapy agents in the treatment of acute leukemia. Myelosuppression and gastrointestinal mucositis are expected side effects of mercaptopurine at high doses.
  • a useful mercaptopurine analog is azathioprine.
  • Thioguanine 2-amino-l,7-dihydro-6H-purine-6-thione
  • TABLOID ® Thioguanine exhibits cell phase specificity at S-phase by inhibiting DNA synthesis by an as of yet unspecified mechanism.
  • Thioguanine is indicated as a single agent or in combination with other chemotherapy agents in the treatment of acute leukemia.
  • Myelosuppression including leucopenia, thrombocytopenia, and anemia, is the most common dose limiting side effect of thioguanine administration.
  • Other purine analogs include pentostatin, erythrohydroxynonyladenine, fludarabine phosphate, and cladribine.
  • Gemcitabine 2'-deoxy-2', 2'-difluorocytidine monohydrochloride ( ⁇ -isomer), is commercially available as GEMZAR ® .
  • GEMZAR ® 2'-deoxy-2', 2'-difluorocytidine monohydrochloride
  • Gemcitabine exhibits cell phase specificity at S-phase and by blocking progression of cells through the Gl/S boundary.
  • Gemcitabine is indicated in combination with cisp latin in the treatment of locally advanced non-small cell lung cancer and alone in the treatment of locally advanced pancreatic cancer.
  • Myelosuppression including leucopenia, thrombocytopenia, and anemia, is the most common dose limiting side effect of gemcitabine administration.
  • Methotrexate N-[4[[(2,4-diamino-6-pteridinyl) methyl]methylamino] benzoyl]-L-glutamic acid, is commercially available as methotrexate sodium. Methotrexate exhibits cell phase effects specifically at S-phase by inhibiting DNA synthesis, repair and/or replication through the inhibition of dyhydrofolic acid reductase which is required for synthesis of purine nucleotides and thymidylate.
  • Methotrexate is indicated as a single agent or in combination with other chemotherapy agents in the treatment of choriocarcinoma, meningeal leukemia, non-Hodgkin's lymphoma, and carcinomas of the breast, head, neck, ovary and bladder.
  • Myelosuppression (leucopenia, thrombocytopenia, and anemia) and mucositis are expected side effect of methotrexate administration.
  • Camptothecins including, camptothecin and camptothecin derivatives are available or under development as Topoisomerase I inhibitors. Camptothecins cytotoxic activity is believed to be related to its Topoisomerase I inhibitory activity. Examples of camptothecins include, but are not limited to irinotecan, topotecan, and the various optical forms of 7-(4-methylpiperazino-methylene)- 10, 1 1 - ethylenedioxy-20-camptothecin described below.
  • Irinotecan is a derivative of camptothecin which binds, along with its active metabolite SN- 38, to the topoisomerase I - DNA complex. It is believed that cytotoxicity occurs as a result of irreparable double strand breaks caused by interaction of the topoisomerase I : DNA : irintecan or SN- 38 ternary complex with replication enzymes. Irinotecan is indicated for treatment of metastatic cancer of the colon or rectum.
  • the dose limiting side effects of irinotecan HC1 are myelosuppression, including neutropenia, and GI effects, including diarrhea.
  • Topotecan HC1 (S)- 10- [(dimethylamino)methyl]-4-ethyl-4,9-dihydroxy- 1 H- pyrano[3',4',6,7]indolizino[l,2-b]quinoline-3, 14-(4H, 12H)-dione monohydrochloride, is commercially available as the injectable solution HYCAMTIN ® .
  • Topotecan is a derivative of camptothecin which binds to the topoisomerase I - DNA complex and prevents religation of singles strand breaks caused by Topoisomerase I in response to torsional strain of the DNA molecule.
  • Topotecan is indicated for second line treatment of metastatic carcinoma of the ovary and small cell lung cancer.
  • the dose limiting side effect of topotecan HC1 is myelosuppression, primarily neutropenia.
  • compositions may be presented in unit dose forms containing a predetermined amount of active ingredient per unit dose.
  • a unit may contain, for example, 0.5 mg to 1 g, preferably 1 mg to 700 mg, more preferably 5 mg to 100 mg of a compound of the formula (I), depending on the condition being treated, the route of administration and the age, weight and condition of the patient, or pharmaceutical compositions may be presented in unit dose forms containing a predetermined amount of active ingredient per unit dose.
  • Preferred unit dosage compositions are those containing a daily dose or sub-dose, as herein above recited, or an appropriate fraction thereof, of an active ingredient.
  • such pharmaceutical compositions may be prepared by any of the methods well known in the pharmacy art.
  • compositions may be adapted for administration by any appropriate route, for example by the oral (including buccal or sublingual), rectal, nasal, topical (including buccal, sublingual or transdermal), vaginal or parenteral (including subcutaneous, intramuscular, intravenous or intradermal) route.
  • Such compositions may be prepared by any method known in the art of pharmacy, for example by bringing into association a compound of formal (I) with the carrier(s) or excipient(s).
  • compositions adapted for oral administration may be presented as discrete units such as capsules or tablets; powders or granules; solutions or suspensions in aqueous or nonaqueous liquids; edible foams or whips; or oil-in-water liquid emulsions or water-in-oil liquid emulsions.
  • Capsules are made by preparing a powder mixture, as described above, and filling formed gelatin sheaths.
  • Glidants and lubricants such as colloidal silica, talc, magnesium stearate, calcium stearate or solid polyethylene glycol can be added to the powder mixture before the filling operation.
  • a disintegrating or solubilizing agent such as agar-agar, calcium carbonate or sodium carbonate can also be added to improve the availability of the medicament when the capsule is ingested.
  • suitable binders, lubricants, disintegrating agents and coloring agents can also be incorporated into the mixture.
  • Suitable binders include starch, gelatin, natural sugars such as glucose or beta-lactose, corn sweeteners, natural and synthetic gums such as acacia, tragacanth or sodium alginate, carboxymethylcellulose, polyethylene glycol, waxes and the like.
  • Lubricants used in these dosage forms include sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride and the like.
  • Disintegrators include, without limitation, starch, methyl cellulose, agar, bentonite, xanthan gum and the like. Tablets are formulated, for example, by preparing a powder mixture, granulating or slugging, adding a lubricant and disintegrant and pressing into tablets.
  • a powder mixture is prepared by mixing the compound, suitably comminuted, with a diluent or base as described above, and optionally, with a binder such as carboxymethylcellulose, an aliginate, gelatin, or polyvinyl pyrrolidone, a solution retardant such as paraffin, a resorption accelerator such as a quaternary salt and/or an absorption agent such as bentonite, kaolin or dicalcium phosphate.
  • the powder mixture can be granulated by tablet forming dies by means of the addition of stearic acid, a stearate salt, talc or mineral oil. The lubricated mixture is then compressed into tablets.
  • the compounds of the present invention can also be combined with a free flowing inert carrier and compressed into tablets directly without going through the granulating or slugging steps.
  • a clear or opaque protective coating consisting of a sealing coat of shellac, a coating of sugar or polymeric material and a polish coating of wax can be provided.
  • Dyestuffs can be added to these coatings to distinguish different unit dosages.
  • Oral fluids such as solution, syrups and elixirs can be prepared in dosage unit form so that a given quantity contains a predetermined amount of a compound of formula (I).
  • Syrups can be prepared by dissolving the compound in a suitably flavored aqueous solution, while elixirs are prepared through the use of a non-toxic alcoholic vehicle.
  • Suspensions can be formulated by dispersing the compound in a non-toxic vehicle.
  • Solubilizers and emulsifiers such as ethoxylated isostearyl alcohols and polyoxy ethylene sorbitol ethers, preservatives, flavor additive such as peppermint oil or natural sweeteners or saccharin or other artificial sweeteners, and the like can also be added.
  • dosage unit pharmaceutical compositions for oral administration can be microencapsulated.
  • the formulation can also be prepared to prolong or sustain the release as for example by coating or embedding particulate material in polymers, wax or the like.
  • compositions adapted for rectal administration may be presented as suppositories or as enemas.
  • compositions adapted for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams or spray formulations.
  • compositions adapted for parenteral administration include aqueous and nonaqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the composition isotonic with the blood of the intended recipient; and aqueous and nonaqueous sterile suspensions which may include suspending agents and thickening agents.
  • the pharmaceutical compositions may be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use.
  • Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets.
  • compositions may include other agents conventional in the art having regard to the type of formulation in question, for example those suitable for oral administration may include flavouring agents.
  • a therapeutically effective amount of a compound of the present invention will depend upon a number of factors including, for example, the age and weight of the intended recipient, the precise condition requiring treatment and its severity, the nature of the formulation, and the route of administration, and will ultimately be at the discretion of the attendant prescribing the medication.
  • an effective amount of a compound of formula (I) for the treatment of anemia will generally be in the range of O.001 to 100 mg/kg body weight of recipient per day, suitably in the range of .01 to 10 mg/kg body weight per day.
  • the actual amount per day would suitably be from 7 to 700 mg and this amount may be given in a single dose per day or in a number (such as two, three, four, five or six) of sub-doses per day such that the total daily dose is the same.
  • An effective amount of a salt or solvate, etc. may be determined as a proportion of the effective amount of the compound of formula (I) per se. It is envisaged that similar dosages would be appropriate for treatment of the other conditions referred to above. Chemical Background
  • the compounds of this invention may be made by a variety of methods, including standard chemistry. Any previously defined variable will continue to have the previously defined meaning unless otherwise indicated. Illustrative general synthetic methods are set out below and then specific compounds of the invention as prepared are given in the examples.
  • the present invention includes both possible stereoisomers and includes not only racemic compounds but the individual enantiomers as well.
  • a compound When a compound is desired as a single enantiomer, it may be obtained by stereospecific synthesis or by resolution of the final product or any convenient intermediate.
  • Resolution of the final product, an intermediate, or a starting material may be effected by any suitable method known in the art. See, for example, Stereochemistry of Organic Compounds by E. L. Eliel, S. H. Wilen, and L. N. Mander (Wiley- Interscience, 1994).
  • Scheme 1 illustrates a route to prepare compounds of formula (VI).
  • Compounds of formula (I) are commercially available and were alkylated using sodium hydride, DMF, and an appropriate alkyl halide to afford a mixture of N1 N2 alkylated regioisomers which were separated by chromatography and fully characterized.
  • Compounds of formula (II) were converted to compounds of formula (III) by standard base-catalyzed hydrolysis.
  • Treatment of compounds of formula (III) with substituted aminomethyl pyridones of formula (IV) using EDC, HOAT, N-methylmorpholine, and DMSO at room temperature for a period of 3-48 h stirring afforded compounds of formula (V).
  • Compounds of formula (V) were substituted at the 6-position using standard methods known to those skilled in the art (i.e. palladium, copper, mediated cross couplings), to afford compounds of formula (VI).
  • Scheme 3 illustrates a route to prepare compounds of formula (XV).
  • a compound of formula (IX) which is commercially available, was converted to a compound of formula (X) using sodium nitrite, 6N HC1, water, and stirring at room temperature for 48 h.
  • Compounds of formula (XI) were converted to compounds of formula (XII) typically by Wolff-Kischner mediated reduction conditions using sodium cyanoborohydride as the reducing agent.
  • Compounds of formula (XII) were converted to compounds of formula (XIII) by standard base-catalyzed hydrolysis.
  • Scheme 5 illustrates the method to synthesize a compound of formula (IV). Heating of compounds of formula (XVIII) with cyanoacetamide in ethanol at reflux containing catalytic piperidine for typically 30 min, affords compounds of formula (XX). Alternatively, treatment of compounds of formula (XIX) with cyanoacetamide in DMSO at room temperature with an excess of potassium tert-butoxide under an atmosphere of oxygen for ca. 90 min. also affords compounds of formula (XX).
  • Celite ® registered trademark of Celite Corp. brand of diatomaceous earth
  • Preparative normal phase silica gel chromatography was carried out using either a Teledyne ISCO CombiFlash Companion instrument with RediSep or ISCO Gold silica gel cartridges (4 g-330 g), or an Analogix IF280 instrument with SF25 silica gel cartridges (4 g - 3-00g), or a Biotage SP1 instrument with HP silica gel cartridges (lOg - 100 g).
  • Purification by reverse phase HPLC was conducted using a YMC-pack column (ODS-A 75x30mm) as solid phase, unless otherwise noted.
  • a mobile phase of 25mL/min A (acetonitrile-0.1%TFA) : B (water-0.1% TFA), 10-80% gradient A (10 min) was utilized with UV detection at 214 nM, unless otherwise noted.
  • a PE Sciex API 150 single quadrupole mass spectrometer (PE Sciex, Thornhill, Ontario, Canada) was operated using electrospray ionization in the positive ion detection mode.
  • the nebulizing gas was generated from a zero air generator (Balston Inc., Haverhill, MA, USA) and delivered at 65 psi and the curtain gas was high purity nitrogen delivered from a Dewar liquid nitrogen vessel at 50 psi.
  • the voltage applied to the electrospray needle was 4.8 kV.
  • the orifice was set at 25 V and mass spectrometer was scanned at a rate of 0.5 scan/sec using a step mass of 0.2 amu and collecting profile data.
  • Method A LCMS Method A LCMS. Samples were introduced into the mass spectrometer using a CTC PAL autosampler (LEAP Technologies, Carrboro, NC) equipped with a hamilton 10 uL syringe which performed the injection into a Valco 10-port injection valve.
  • the HPLC pump was a Shimadzu LC- lOADvp (Shimadzu Scientific Instruments, Columbia, MD) operated at 0.3 mL/min and a linear gradient 4.5% A to 90% B in 3.2 min. with a 0.4 min. hold.
  • the mobile phase was composed of 100% (H 2 0 0.02% TFA) in vessel A and 100% (CH 3 CN 0.018% TFA) in vessel B.
  • the stationary phase is Aquasil (C I 8) and the column dimensions were 1 mm x 40 mm. Detection was by UV at 214 nm, evaporative light- scattering (ELSD) and MS.
  • Method B LCMS.
  • an Agilent 1 100 analytical HPLC system with an LC/MS was used and operated at 1 mL/min and a linear gradient 5% A to 100% B in 2.2 min with a 0.4 min hold.
  • the mobile phase was composed of 100% (H 2 0 0.02% TFA) in vessel A and 100% (CH 3 CN 0.018% TFA) in vessel B.
  • the stationary phase was Zobax (C8) with a 3.5 um partical size and the column dimensions were 2.1 mm x 50 mm. Detection was by UV at 214 nm, evaporative light- scattering (ELSD) and MS.
  • Method C LCMS.
  • an MDSSCIEX API 2000 equipped with a capillary column of (50 x 4.6 mm, 5 ⁇ ) was used.
  • HPLC was done on Agilent- 1200 series UPLC system equipped with column Zorbax SB-C 18 (50 x 4.6 mm, 1.8 /mi) eluting with CH 3 CN: ammonium acetate buffer. The reactions were performed in the microwave (CEM, Discover).
  • Analytical HPLC Products were analyzed by Agilent 1 100 Analytical Chromatography system, with 4.5 x 75 mm Zorbax XDB-C18 column (3.5 um) at 2 mL/min with a 4 min gradient from 5% CH 3 CN (0.1% formic acid) to 95% CH 3 CN (0.1% formic acid) in H 2 0 (0.1% formic acid) and a 1 min hold. 1.
  • Agilent 1 100 Analytical Chromatography system with 4.5 x 75 mm Zorbax XDB-C18 column (3.5 um) at 2 mL/min with a 4 min gradient from 5% CH 3 CN (0.1% formic acid) to 95% CH 3 CN (0.1% formic acid) in H 2 0 (0.1% formic acid) and a 1 min hold. 1.
  • Agilent 1 100 Analytical Chromatography system with 4.5 x 75 mm Zorbax XDB-C18 column (3.5 um) at 2 mL/min with
  • Methyl 6-chloro-lH-indazole-4-carboxylate (0.4 lOg, 1.947 mmol) was dissolved in DMF (10 mL) and placed into an ice bath and stirred for 15 min.
  • Sodium hydride (0.101 g, 2.53 mmol) was added, the contents stirred for 15 min., and then 2-iodopropane (0.389 mL, 3.89 mmol) was added.
  • the contents were stirred with warming to RT. After stirring at RT for 2h, a scoop of sodium carbonate and then 0.4 mL of methyl iodide were added.
  • Methyl 6-chloro-l-(l-methylethyl)-lH-indazole-4-carboxylate (0.22g, 0.871 mmol) was dissolved in methanol (6 mL) and THF (1 mL), followed by addition of NaOH (1.451 mL, 4.35 mmol) via syringe. The contents were stirred at RT for 3h. The volatiles were removed in vacuo, the contents diluted with water, and then slowly acidifed to pH 4-5 by addition of 1M HC1 (solid precipitation occured). The contents were then extracted with EtOAc (3x). The combined organic layers were washed with brine, dried over MgSC , filtered, and concentrated in vacuo.
  • the contents were stirred at RT overnight Additional HOAT and EDC were added, and the contents were stirred for another 6h.
  • the contents were slowly poured onto 45 mL of ice-water wherein solid precipiatation occurred.
  • the contents were stirred for 10 min., and then allowed to stand for another 10 min.
  • the contents were filtered, the collected solid washed with ca. 5 mL of water.
  • the solid was air dried for 15 min. then dried in vacuum oven at 45 °C for 24h.
  • the title compound was collected as a white solid (0.160g, 93 % yield).
  • 6-fluoro- lH-indazole-4-carboxylate (.500g, 2.58 mmol), sodium hydride (0.129 g, 3.22 mmol), and 2- iodopropane (0.386 mL, 3.86 mmol).
  • step c The title compound was prepared in the same manner as described for example 1 (step c) from 6- fluoro- l -(l-methylethyl)- lH-indazole-4-carboxylic acid (.085g, 0.383 mmol),3-(aminomethyl)-4,6- dimethyl-2(lH)-pyridinone (0.108 g, 0.574 mmol), 1 -hydroxy-7-azabenzotriazole (0.078 g, 0.574 mmol), EDC (0.1 10 g, 0.574 mmol), and N-methylmorpho line (0.210 mL, 1.913 mmol). The product was collected as a white solid (117 mg, 82 %) !
  • N-methylmorpholine (0.64 ml, 5.79 mmol) and EDC (433 mg, 2.26 mmol) were added, the vessel was sealed, and the mixture was stirred at room temperature for 2 days. The mixture was poured onto 10 mL of ice-water and solids crashed out. 10% K 2 C0 3 was added to adjust the pH ⁇ 8-9 and the contents were stirred for 10 min and then allowed to stand for another 10 min. Solids were filtered and air-dried. DMF along with some water were added and solids that precipitated were filtered. DCM was added and the contents purified by Si0 2 chromatography (eluent: gradient 0 to 80:20:2 DCM/MeOH/NEL j OH).
  • the title compound was prepared in the same manner as described for the example 4 from 6-bromo- 1 - (l-methylethyl)-lH-indazole-4-carboxylic acid (100 mg, 0.353 mmol) and 3-(aminomethyl)-6- methyl-4-(phenylmethyl)-2(lH)-pyridinone (129 mg, 0.565 mmol), wherein the reaction stir time was 12h.
  • the product was collected as a white solid (130 mg, 71%).
  • reaction contents were stirred at RT for 12 hr, after which time an addtional 20 mg each of amine, EDC, and HOAt were added. After stirring for an additional 2h, the reaction mixture was poured onto ice water (lOmL), stirred for 20 min, allowed to stand for 10 min, and filtered. The collected solid was rinsed with water (10 mL), and then purified by reverse phase HPLC (10-90% acetonitrile/water + 0.1% TFA). The product fractions were treated with NaHCOg (sat aq), extracted with EtOAc, and evaporated from water to aford the final product as a white solid (69 mg, 43%).
  • the mixture was stirred under atmosphere of hydrogen (balloon) for 6h.
  • the mixture was purged with nitrogen and then diluted with DCM (10 mL) and a small amount of celite, After stirring for 10 min, the contents were then filtered through analytical grade celite and washed with 10% MeOH/DCM, EtOH, and then DCM.
  • the filtrate was concentrated in vacuo and dried under hi vacuum overnight.
  • the crude product was purified by silica gel chromatography (eluent: 5- 50% gradient of 10% methanol in dichloromethane and dichloromethane). The final product was concentrated from MTBE and dried in vacuum oven at 45 °C for 18h.
  • the title compound was prepared in a similar manner as described for example 8 from 6-bromo-l-(l- methylethyl)-N-[(6-methyl-2-oxo-4-methyl- 1 ,2-dihydro-3-pyridinyl)methyl]- lH-indazole-4- carboxamide (90 mg, 0.216 mmol) and l-[5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-2- pyridinyl]piperazine (94 mg, 0.324 mmol).
  • the product obtained from HPLC purification was treated with a saturated solution of NaHC0 3 , and solids that crashed out were filtered .
  • the title compound was prepared in a similar manner as described for example 8 from 6-bromo-l-(l- methylethyl)-N- ⁇ [6-methyl-4-(l-methylethyl)-2-oxo-l,2-dihydro-3-pyridinyl]methyl ⁇ -lH-indazole-4- carboxamide (90 mg, 0.202 mmol) and dimethyl ⁇ [4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)phenyl]methyl ⁇ mine (90 mg, 0.303 mmol). The product was collected as a white solid (54 mg, 49%).
  • the collected product was then further purified by reverse- phase HPLC (15% to 80% CH 3 CN in water with 0.1% TFA), which afforded the product as a TFA salt.
  • CH3CN was evaporated and a saturated solution of sodium bicarbonate was added to the water layer followed by DCM/isopropanol (70:30).
  • the organic layer was separated, and the aqueous layer was further extracted with DCM/isopropanol (70:30).
  • the combined organic layers were washed with brine, dried over MgS0 4 , filtered, and concentrated. DMF was added along with some water, and after sitting overnight, the solids that precipitated were filtered.
  • PdCl 2 (dppf)-CH 2 Ci 2 adduct (10.8 mg, 0.013 mmol) was added and the resulting mixture was degassed with nitrogen for 10 min.
  • Sodium bicarbonate (66.4 mg, 0.79 mmol) was added, the vessel was sealed, and the insoluble mixture was heated in a microwave at 100 °C for 20 min. The mixture was evaporated, DCM/MeOH (1 : 1) was added, and the contents pre-absorbed on silica gel and purified by Si0 2 chromatography (eluent: gradient 0 to 80:20:2 DCM/MeOH/NH 4 OH).
  • the title compound was prepared in a similar manner as described for example 8 from 6-bromo-l-(l- methylethyl)-N-[(6-methyl-2-oxo-4-propyl- 1 ,2-dihydro-3-pyridinyl)methyl]- 1 H-indazole-4- carboxamide (90 mg, 0.202 mmol) and l-[5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-2- pyridinyl]piperazine (88 mg, 0.303 mmol). The final product was collected as a white solid (91 mg, 83%).
  • the collected product was further purified by reversed-phase HPLC (15% to 80% CH3CN in water with 0.1% TFA) which afforded the TFA salt.
  • CH3CN was evaporated, and a saturated solution of sodium bicarbonate was added to the water layer along with EtOAc.
  • the organic layer was separated, and the aqueous layer was further extracted with EtOAc, DCM and DCM/isopropanol (8:2).
  • EtOAc was added along with some hexanes, and the contents were sonicated and then allowed sit overnight at room temperature. Solids that precipitated were filtered and dried to afford the title compound (84 mg, 76%) as a white solid.
  • PdCl 2 (dppf)-CH 2 Cl 2 adduct (9.2 mg, 0.01 1 mmol) was added and the resulting mixture was degassed with nitrogen for 10 min.
  • Sodium bicarbonate (56.6 mg, 0.67 mmol) was added, the vessel was sealed, and the insoluble mixture was heated in a microwave at 1 10 °C for 20 min.
  • the mixture was evaporated, DCM/MeOH (1 : 1) was added, and the contents pre-absorbed on silica gel and purified by Si0 2 chromatography (eluent: gradient 0 to 80:20:2 DCM/MeOH/NH 4 OH).
  • the title compound was prepared in a similar manner as described for example 13 from 6-bromo- 1- (l-methylethyl)-N-[(6-methyl-2-oxo-4-propyl-l,2-dihydro-3-pyridinyl)methyl]- lH-indazole-4- carboxamide (100 mg, 0.225 mmol) and [6-(dimethylamino)-3-pyridinyl]boronic acid (49 mg, 0.292 mmol). The final product was collected as a white solid (49 mg, 44%).
  • the title compound was prepared in a similar manner as described for example 8 from 6-bromo- 1 -(1 - methylethyl)-N-[(6-methyl-2-oxo-4-propyl- 1 ,2-dihydro-3-pyridinyl)methyl]- 1 H-indazole-4- carboxamide (100 mg, 0.225 mmol) and [6-(4-morpholinyl)-3-pyridinyl]boronic acid (70 mg, 0.337 mmol). The final product was collected as a white solid (98 mg, 81%). H NMR (400 MHz, DMSO- d 6 ) ⁇ ppm 1 1.46 (br.
  • the title compound was prepared in the same manner as described for example 75 from 6-bromo- 1 - (1 -methylethyl)-N- [(6-methyl-2-oxo-4-propyl- 1 ,2-dihydro-3 -pyridinyl)methyl] - 1 H-indazole-4- carboxamide (100 mg, 0.225 mmol) and 5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-2- pyrimidinamine (75 mg, 0.337 mmol). The final product was collected as a white solid (87 mg, 82%).
  • the title compound was prepared in the same manner as described for example 75 from 6-bromo- 1 - (l-methylethyl)-N-[(6-methyl-2-oxo-4-propyl-l,2-dihydro-3-pyridinyl)methyl]-lH-indazole-4- carboxamide (100 mg, 0.225 mmol) and 5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-2- pyridinamine (75 mg, 0.337 mmol). The final product was collected as a white solid (60 mg, 57%).
  • the title compound was prepared in the same manner as described for example 75 from 6-bromo- 1 - (1 -methylethyl)-N- [(6-methyl-2-oxo-4-propyl- 1 ,2-dihydro-3 -pyridinyl)methyl] - 1 H-indazole-4- carboxamide (100 mg, 0.225 mmol) and 5-pyrimidinylboronic acid (42 mg, 0.337 mmol). The final product was collected as a white solid (77 mg, 75%).
  • the title compound was prepared in a similar manner as described for example 8 from 6-bromo- 1-(1- methylethyl)-N-[(6-methyl-2-oxo-4-propyl- 1 ,2-dihydro-3-pyridinyl)methyl]- 1 H-indazole-4- carboxamide (90 mg, 0.202 mmol) and 1-methyl [5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-2- pyridinyljpiperazine (92 mg, 0.303 mmol). The final product was collected as a light brown solid (54 mg, 49%).
  • the title compound was prepared in a similar manner as described for example 8 from 6-bromo-N- [(4,6-dimethyl-2-oxo- 1 ,2-dihydro-3-pyridinyl)methyl]- 1 -(1 -methylethyl)- lH-indazole-4-carboxamide (1 10 mg, 0.264 mmol) and [6-(dimethylamino)-3-pyridinyl]boronic acid (66 mg, 0.395 mmol). The product was collected as a white solid (33 mg, 27%).
  • the title compound was prepared in a similar manner as described for example 8 from 6-bromo-N- [(4,6-dimethyl-2-oxo- 1 ,2-dihydro-3-pyridinyl)methyl]- 1 -(1 -methylethyl)- lH-indazole-4-carboxamide (1 10 mg, 0.264 mmol) and [6-(trifluoromethyl)-3-pyridinyl]boronic acid (75 mg, 0.395 mmol). The product was collected as a solid (75 mg, 57%). !
  • the title compound was prepared in a similar manner as described for example 12 from 6-bromo- l- (1 -methylethyl)-N- [(6-methyl-2-oxo-4-methyl- 1 ,2-dihydro-3-pyridinyl)methyl]- lH-indazole-4- carboxamide (100 mg, 0.240 mmol) and dimethyl ⁇ [3-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)phenyl]methyl ⁇ amine (93 mg, 0.312 mmol).
  • the product obtained from HPLC purification was treated with a saturated solution of NaHC0 3 , solids that crashed out were filtered, and air-dried for 15 min.
  • the title compound was prepared in a similar manner as described for example 12 from 6-bromo- l- (1 -methylethyl)-N- [(6-methyl-2-oxo-4-methyl- 1 ,2-dihydro-3-pyridinyl)methyl]- lH-indazole-4- carboxamide (100 mg, 0.240 mmol) and dimethyl ⁇ [3-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-3- yl)phenyl]methyl ⁇ amine (93 mg, 0.312 mmol).
  • the product obtained from HPLC purification was treated with a saturated solution of NaHC0 3 , solids that crashed out were filtered, and air-dried for 15 min.
  • the title compound was prepared from 6-bromo-N-((l,2-dihydro-6-methyl-2-oxo-4-propylpyridin-3- yl)methyl)-l-isopropyl-lH-indazole-4-carboxamide (0.2 g, 0.44 mmol) and 2-(furan-3-yl)-4,4,5,5- tetramethyl- l,3,2-dioxaborolane (0.1 g, 0.51 mmol) in the same manner as described for example 31.
  • the product was collected as an off-white solid (55 mg, 25.7 %).
  • 6-bromo-l-(l-methylethyl)-N-[(6-methyl-2-oxo-4-propyl-l,2- dihydro-3-pyridinyl)methyl]-lH-indazole-4-carboxamide 200 mg, 0.449 mmol
  • 4-(N,N- dimethylaminomethyl)phenylboronic acid pinacol ester hydrochloride 170 mg, 0.571 mmol
  • potassium phosphate 300 mg, 1.413 mmol
  • the solid was taken up in CH 2 C1 2 and treated with Silicycle Si-Thiol (2 g, 1.46 mMol/g). After stirring for 30 minutes on a rotary evaporator (no vacuum) the mixture was filtered through a pad of Celite, washed with CH 2 C1 2 , and evaporated to dryness. The light yellow colored solid was taken up in a small volume of MeOH and treated with 6 N HC1 (200 uL) and re-evaporated to dryness. The residue was dissolved in a small volume of MeOH, ppt.
  • the title compound was prepared in the same manner as described for example 33 using from 6- bromo- 1 -( 1 -methylethyl)-N- [(6-methyl-2-oxo-4-propyl- 1 ,2-dihydro-3 -pyridinyl)methyl] - 1 H- indazole-4-carboxamide (200 mg, 0.449 mmol), 3-(N,N dimethylaminomethyl)phenylboronic acid pinacol ester hydrochloride (170 mg, 0.571 mmol), potassium phosphate (300 mg, 1.413 mmol), dioxane (12 mL), water (3 mL), and PdCl 2 (dppf)-CH 2 Cl 2 adduct (50 mg, 0.061 mmol).
  • step a from methyl 6-bromo-l-isopropyl-3 -methyl- lH-indazole-4-carboxy late (1 g, 3.36 mmol) and 5-(4,4,5,5- tetramethyl- l,3,2-dioxaborolan-2-yl)-lH-pyrrolo[2,3-b]pyridine (0.98 g, 4.04 mmol) wherein the reaction mixture was stirred at 100 °C for 5 h. The product was collected as an off white solid (600 mg, 53%). LCMS (ES-) m/z: 333.08. b) l-isopropyl-6-(lH-pyrrolo[2,3-b]pyridin-5-yl)-lH-indazole-4-carboxylic acid
  • step b The title compound was prepared in the same manner as described for example 35 (step b) from methyl l-isopropyl-6-(lH-pyrrolo[2,3-b]pyridin-5-yl)-lH-indazole-4-carboxylate 1 (0.6 g, 1.79 mmol) and LiOH H 2 0 (0.22 g, 5.37 mmol) wherein the mixture was heated at 80 °C for 5 h. The product was collected as an off-white solid (0.5 g, 87.7%).
  • step c The title compound was prepared in the same manner as described for example 35 (step c) from 1- isopropyl-3-methyl-6-(pyridin-3-yl)-lH-indazole-4-carboxylic acid and 3-(aminomethyl)-6-methyl-4- propylpyridin-2(lH)-one (0.28 g, 1.56 mmol).
  • the crude product was purified by silica gel chromatography (eluent: 5% MeOHYEtOAc) to afford the product as an off-white solid (200 mg, 26 %).
  • step c The title compound was prepared using the procedure described for example 37 (step c) from 6-(4- ((dimethylamino)methyl)piperidin- 1 -yl)- 1 -isopropyl- 1 H-indazole-4-carboxylic acid and 3 -(aminomethyl)-6- methyl-4-propyl-2(lH)-pyridinone.
  • 6-bromo-l-(l-methylethyl)-lH-indazole-4-carboxylic acid (237 mg, 0.84 mmol), 3-(aminomethyl)-4- cyclohexyl-6-methyl-2(lH)-pyridinone ' TFA (378 mg, 1.13 mmol) and 1 -hydroxy-7-azabenzotriazole (171 mg, 1.26 mmol) were stirred in 10 mL of DMSO for 10 min under nitrogen. N- methylmorpholine (0.37 ml, 3.35 mmol) was added along with EDC (241 mg, 1.26 mmol) and the mixture was stirred at rt overnight.
  • the crude product was purified by silica gel chromatography (eluent: 0 to 30% EtOAc in petroleum ether) to afford the title compound 6-bromo-l-ethyl-lH-indazole-4-carboxylic acid methyl ester (700 mg, 25%) and the undesired isomer 6-bromo-2-ethyl-2H-indazole-4-carboxylic acid methyl ester (500 mg, 18%) as white solids.
  • Step 1 To a stirred solution of 6-bromo-l -ethyl- lH-indazole-4-carboxylic acid methyl ester (700 mg,
  • Step 2 To a stirred suspension of 6-bromo-l-ethyl-lH-indazole-4-carboxylic acid, 4 (450 mg, 1.67 mmol) in DCM (50 mL) were added EDC.HC1 (384 mg, 2.00 mmol), HOBt.H 2 0 (306 mg, 2.00 mmol) and then stirred for 15 min at RT. To the resulting mixture, DIPEA (1.2 mL, 5.59 mmol) followed by 3-aminomethyl-6-methyl-4-propyl-lH-pyridin-2-one (301 mg, 1.67 mmol) were added and the contents stirred for 18 h at RT.
  • EDC.HC1 384 mg, 2.00 mmol
  • HOBt.H 2 0 306 mg, 2.00 mmol
  • the reaction mixture was diluted with DCM (50 mL) and washed with water (2x50 mL), 10% aq citric acid solution (2x50 mL), saturated aq NaHCCh solution (2x30 mL) and brine (2x50 mL).
  • the organic layer was dried over Na SO/i, filtered, and concentrated under reduced pressure.
  • the obtained solid was washed with diethyl ether (2 x 50 mL) to afford the title compound as a white solid (230 mg, 32 %).
  • step a The title compounds were prepared in the same manner as described for example 42 (step a) from 6- bromo-lH-indazole-4-carboxylic acid methyl ester (3.0 g, 11.66 mmol) and 1-propyl bromide (1.59 g, 12.94 mmol) wherein the contents were heated at at 40 °C for 30 min.
  • step b, part 1 The title compound was prepared in the same manner as described for example 42 (step b, part 1) from 6-bromo-l -propyl- lH-indazole-4-carboxylic acid, 7 (700 mg, 2.35 mmol ) and LiOH.H 2 0 (290 mg, 7.07 mmol). The product was collected as an off-white solid (600 mg, 90 %).
  • step b, part 2 The title compound was prepared in the same manner as described for example 42 (step b, part 2) from 6-bromo-l -propyl- lH-indazole-4-carboxy lie acid (300 mg, 1.06 mmol) and 3-aminomethyl-6- methyl-4-propyl-lH-pyridin-2-one (190 mg, 1.06 mmol).
  • the crude product was triturated with diethyl ether (10 mL) and n-pentane (10 mL) to afford the title compound as a white solid ( 200 mg,
  • Step 2 6-bromo-l-cyclopentyl-lH-indazole-4-carboxylic acid (1.44 g, 4.61 mmol), 3-(aminomethyl)- 4,6-dimethyl-2(lH)-pyridinone hydrochloride (1.138 g, 6.03 mmol), N-[3-(dimethylamino)propyl]- N'-ethylcarbodiimide hydrochloride (1.335 g, 6.96 mmol), and 3H-[l,2,3]triazolo[4,5-b]pyridin-3-ol hydrate (1.073 g, 6.96 mmol) were suspended in DMSO (8.00 mL), followed by N-methylmorpholine (2.82 g, 27.8 mmol).
  • 6-bromo-l-cyclopentyl-lH-indazole-4- carboxylic acid 370 mg, 1.2 mmol
  • 3-(aminomethyl)-4-ethyl-6-methyl-2(lH)-pyridinone HCl (340 mg, 1.68 mmol) in DMSO (3 mL).
  • 1 -hydroxy-7-azabenzotriazole (277 mg, 2.04 mmol) was added and the resulting mixture was degassed with nitrogen for 10 minutes.
  • N-methylmorpholine (0.55 ml, 5 mmol) and EDC (390 mg, 2 mmol) were added, the vessel was sealed, and the mixture was stirred at room temperature overnight.
  • the contents were irradiated in a microwave reactor at 150 °C for 30 min.
  • the reaction solution was filtered through a Whatman 0.45 ⁇ 1 Teflon syringless filter device and diluted with DMSO (6 mL).
  • the DMSO solution was purified by HPLC reverse phase chromatography (phenomenex Gemini-NX, 30x100 5 ⁇ column, 5-30% acetonitrile/water 0.01% formic acid, 8 min gradient).
  • the fractions containing the desired product were concentrated to dryness using a Genovac HT-4 instrument at 40 °C.
  • the title compound was obtained as a pale cream colored foam (28 mg, 14.83 % yield).
  • Examples 47-52 were prepared in a similar manner as described above using 6-bromo- 1 -cyclopentyl- N-[(4,6-dimethyl-2-oxo-l,2-dihydro-3-pyridinyl)methyl]-lH-indazole-4-carboxamide and the appropriate boronic acid reagent.
  • the title compound was prepared in a similar manner as described for example 8 from 6-bromo-l- cyclopentyl-N-[(4,6-dimethyl-2-oxo-l,2-dihydro-3-pyridinyl)methyl]-lH-indazole-4-carboxamide (100 mg, 0.226 mmol) and l-methyl-4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-lH-pyrazole (70 mg, 0.338 mmol). The product was collected as a white solid (82 mg, 81%). l U NMR (400 MHz, DMSO-6? 6 ) ⁇ ppm 1 1.53 (br.
  • the title compound was prepared in a similar manner as described for example 8 from 6-bromo-l- cyclopentyl-N-[(4,6-dimethyl-2-oxo-l,2-dihydro-3-pyridinyl)methyl]-lH-indazole-4-carboxamide (200 mg, 0.451 mmol) and l-[5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-2-pyridinyl]piperazine (196 mg, 0.677 mmol). The product was collected as a white solid (92 mg, 37%). ! H NMR (400 MHz, DMSO-J 6 ) ⁇ ppm 11.54 (br.
  • 6-bromo-l-cyclopentyl-N-[(4,6-dimethyl-2-oxo-l,2-dihydro-3- pyridinyl)methyl]-lH-indazole-4-carboxamide (.10g, 0.226 mmol)
  • N,N-dimethyl-l-[4-(4,4,5,5- tetramethyl-l,3,2-dioxaborolan-2-yl)phenyl]methanamine 0.081 g, 0.271 mmol
  • potassium phosphate tribasic
  • the suspension was stirred with degassing under N 2 for 10 min, followed by addition of PdCl 2 (dppf)- CH 2 C1 2 adduct (0.018 g, 0.023 mmol).
  • the reaction vessel was sealed and stirred with heating at 100 °C (heat block) for 2h. The contents were allowed to stir with cooling to RT overnight.
  • the mixture was diluted with EtOAc followed by addition of silica gel.
  • the mixture was concentrated in vacuo and the obtained solid purified by silica gel chromatography (dry loaded, eluent: 8-95% gradient of chloroform (containing 10% 2M Ammonia in Methanol) and DCM.
  • the collected product was concentrated from DCM/MTBE and then dried in vacuum oven for 16h.
  • step 2 The title compound was prepared in the same manner as described for example 1 (step 2) from methyl 6-chloro- l-cyclopentyl- lH-indazole-4-carboxylate (1.1 lg, 3.98 mmol), wherein the reaction stir time was 12h. The product was collected as 0.99g (85%).
  • step c The title compound was prepared in the same manner as described for example 3 (step c) from 6- chloro- l -cyclopentyl- lH-indazole-4-carboxylic acid (.12g, 0.453 mmol) and 3-(aminomethyl)-4,6- dimethyl-2(lH)-pyridinone (0.128 g, 0.680 mmol), wherein the stir time was 12h.
  • the crude product was purified by silica gel chromatography (eluent: 5-85% gradient chloroform (containing 10% 2M Ammonia in methanol) and dichloromethane). The isolated product was concentrated from MTBE to afford an off-white solid that was dried in hi-vac oven for 6h.
  • step c The title compound was prepared in the same manner as described for example 3 (step c) from 6- chloro-l-cyclopentyl-lH-indazole-4-carboxylic acid (.12g, 0.453 mmol) and 3-(aminomethyl)-6- methyl-4-propyl-2(lH)-pyridinone (0.128 g, 0.680 mmol).
  • the contents were purified by silica gel chromatograrphy (dry loaded; eluent: 2-65% gradient of 10% methanol in dichloromethane and dichloromethane).
  • the product was concentrated from MTBE and dried in hi-vac oven for 6 h at 45
  • 6-cyano-l-cyclopentyl-N-[(4,6-dimethyl-2-oxo-l,2-dihydro-3-pyridinyl)methyl]-lH-indazole-4- carboxamide (.10g, 0.257 mmol) was suspended in acetic acid (8 mL). The contents were placed heated at 40 °C under an atmosphere of hydrogen (50 psi; H-cube reactor) at a flow rate of lmL/min with recirculation 2 h. Then contents were diluted with MeOH, concentrated in vacuo, and dried under hi vacuum for 18 h.
  • the crude product was dissolved in DCM and purified by silica gel chromatography (eluent: 10- 100% gradient of chloroform (containing 10% 2M ammonia in methanol) and dichloromethane.
  • the product was concentrated from MTBE and dried in a vacuum oven at 45
  • the residue was dried under hi-vacuum, and then dissolved in MeOH/DCM followed by addition of silica gel.
  • the contents were concentrated to dryness and dried under hi-vacuum for lh.
  • the contents were purified by silica gel chromatography (dry loaded, eluent: 3-95% gradient chloroform (containing 10% 2M ammonia in methanol) and DCM).
  • the collected solid was triturated from hot acetontrile, and then placed in freezer for 15 min. The contents were filtered cold and washed with additional acetonitrile.
  • 6-amino- 1 -cyclopentyl-N- [(4,6-dimethyl-2-oxo- 1 ,2-dihydro-3-pyridinyl)methyl]- lH-indazole-4- carboxamide (30 mg, 0.079 mmol) was suspended in DCM (3 mL) followed by pyridine (0.032 mL, 0.395 mmol). The contents were vigorously stirred and then benzenesulfonyl chloride (0.01 1 mL, 0.087 mmol) was added and the contents stirred at RT for 1 h. The volatiles were removed in vacuo and dried on hi-vac for lh.
  • the crude product was purified by reverse phase HPLC (Gradient B: 15- 70%. A: Water + .1% TFA. B: CH3CN + .1% TFA). The product was neutralized with water and sat. NaHC03, and extracted with 10% THF/EtOAC (hot) in duplicate. The combined organic layers were dried over MgSC>4, filtered, and concentrated in vacuo. The solid was concentrated from DCM and dried in vacuum oven for 4 h at 45 °C.
  • the crude product was purified by silica gel chromatography (eluent: 5-80% gradient chloroform (containing 10% 2M ammonia in methanol) in DCM).
  • the collected product was purified by reverse phase HPLC ( Gradient B: 15-75 %; A: Water + .1% TFA. B: CH3CN + .1% TFA).
  • the product was suspended in 10% MeOH/CHCl3. Next added 0.5g Silicycle carbonate resin and stirred for 15 min. After standing for 10 min, the contents were filtered through celite and concentrated in vacuo. The product was further concentrated from DCM and MTBE and then dried in hi-vac oven at 45 °C overnight.
  • Methyl 6-bromo-lH-indazole-4-carboxylate (1.0 g, 3.92 mmol) was dissolved in 1 ,2-Dichloroethane (DCE) (14 mL) and stirred for 15 min. Next added cyclopropylboronic acid (0.674 g, 7.84 mmol) and sodium carbonate (0.831 g, 7.84 mmol). The reaction was stirred at RT (suspension). Copper (II) acetate (0.712 g, 3.92 mmol) and 2,2'-bipyridine (0.612 g, 3.92 mmol) were suspended in DCE (24 mL) with heating and the hot suspension was added to the reaction mixture.
  • DCE 1,2-Dichloroethane
  • Methyl 6-bromo- l-cyclopropyl-lH-indazole-4-carboxylate (0.54g, 1.830 mmol) was dissolved in methanol (16 mL) and THF (4 mL) with stirring at RT. A solution of 3N NaOH (1.830 mL, 5.49 mmol) was added the contents were stirred at RT for 2 days. The volatiles were removed in vacuo. The residue was diluted with water and slowly acidifed to pH 3-4 with 1M HCl wherein solids were observed to precipitate. The contents were extracted with EtOAc (2x).
  • step c The title compound was prepared in the same manner as described for example 3 (step c) from 6- bromo-l-cyclopropyl- lH-indazole-4-carboxylic acid (.22 g, 0.783 mmol) and 3-(aminomethyl)-4,6- dimethyl-2(lH)-pyridinone (0.221 g, 1.174 mmol), wherein the reaction stir time was 12h.
  • the title compound was collected as a white solid (0.27g, 81%); !
  • step c The title compound was prepared in the same manner as described for example 3 (step c) from 6- bromo-l-cyclopropyl-lH-indazole-4-carboxylic acid (.22 g, 0.783 mmol) and 3-(aminomethyl)-6- methyl-4-propyl-2(lH)-pyridinone (0.236 g, 1.089 mmol), wherein the reaction stir time was 12 h.
  • the title compound was collected as a white solid (0.36g, 91%); !
  • the title compound was prepared in the same manner as described for example 67 from 6-bromo- 1 - cyclopropyl-N-[(6-methyl-2-oxo-4-propyl-l,2-dihydro-3-pyridinyl)methyl]-lH-indazole-4- carboxamide (.075g, 0.169 mmol) and N,N-dimethyl-l -[4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)phenyl]methanamine (0.060 g, 0.203 mmol).
  • the title compound was collected as a white solid (59 mg, 69 %); !
  • the reaction mixture was then filtered through a celite pad.
  • the filtrate was poured onto ice water (1 L) and extracted with diethyl ether (3 X 700 ml).
  • the combined organic layers were washed with sat NaHC0 3 , brine, and dried over anhydrous Na 2 S0 4 , filtered, and evaporated under vaccum.
  • the crude product was purified by silica gel chromatography (eluent: 10 % ethyl acetate in pet ether) and afforded the title compound as a solid (80g, 59%). !
  • step c The title compound was prepared in the same manner as described for example 3 (step c) from 6- bromo-3-methyl-l-(l-methylethyl)-lH-indazole-4-carboxylic acid (0.24g, 0.808 mmol) and 3- (aminomethyl)-6-methyl-4-propyl-2(lH)-pyridinone (0.219 g, 1.010 mmol).
  • the title compound was collected as a white solid (0.35g, 92%); !
  • PdCl 2 (dppf)-CH 2 Cl 2 adduct (7.6 mg, 0.0093 mmol) was added and the resulting mixture was degassed with nitrogen for 10 min.
  • Sodium bicarbonate (46.6 mg, 0.55 mmol) was added, the vessel was sealed, and the insoluble mixture was heated in a microwave at 110 °C for 20 min. The mixture was evaporated, DCM was added and the contents purified by Si0 2 chromatography (eluent: gradient 0 to 80:20:2 DCM/MeOH/NH 4 OH). The collected product was suspended in EtOAc along with some hexanes. The contents were sonicated and solids that precipitated were filtered.
  • the title compound was prepared in a similar manner as described for example 8 from 6-bromo-3- methyl- 1 -( 1 -methylethyl)-N- [(6-methyl-2-oxo-4-propyl- 1 ,2-dihydro-3 -pyridinyl)methyl] - 1 H- indazole-4-carboxamide (65 mg, 0.141 mmol) and l-[5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)- 2-pyridinyl]piperazine (61 mg, 0.212 mmol). The product was collected as a white solid (54 mg, 49%).
  • the title compound was prepared from 6-bromo-N-((l,2-dihydro-4,6-dimethyl-2-oxopyridin-3- yl)methyl)-l-isopropyl-lH-indazole-4-carboxamide (500 mg, 1.14 mmol) and N,N-dimethyl(3- (4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)phenyl)methanamine (300 mg, 1.14 mmol) in the same manner as described for example 73 wherein the contents were heated at 120 °C for 2 h. The product was collected as a pale red solid (120 mg, 21%).
  • the title compound was prepared from 6-bromo-N-((l,2-dihydro-4,6-dimethyl-2-oxopyridin-3- yl)methyl)-l-isopropyl-lH-indazole-4-carboxamide (300 mg, 0.696 mmol) and dimethyl-[4-(4,4,5,5- tetramethyl-[l,3,2]dioxaborolan-2-yl)-benzyl]-amine (199 mg, 0.765 mmol) in the same manner as described for example 73. The final product was collected as a grey colored solid (80 mg, 26%).
  • the reaction mixture was stirred for 5 min, then sodium bicarbonate (0.4 g, 4.80 mmol) dissolved in water (8 mL) was added and the reaction mixture was stirred at 100 °C for 6 h.
  • the reaction mixture was diluted with water and extracted with ethyl acetate (3x40 mL). The combined organic layers were washed with cold water (2x25 mL), brine solution (25 mL), dried over anhydrous Na 2 S0 4, filtered, and concentrated to afford the crude product.
  • the crude compound was purified by silica gel chromatography (eluent: 100 % ethyl acetate) to afford the title compound as an off-white solid (470 mg, 94%).
  • reaction mixture was filtered and the precipitate was washed with water followed by ether and dried.
  • the solid compound was further purified by silica gel chromatography (eluent: 10 % MeOH in ethyl acetate) to afford the title compound as an off-white solid (225 mg, 31%).
  • the title compound was prepared from methyl 6-bromo- 1 -isopropy 1-3 -methyl- lH-indazole-4- carboxylate, (0.7 g, 2.25 mmol) and 5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-lH-pyrrolo[2,3- b]pyridine (0.65 g, 2.70 mmol) in the same manner as described for example 80 (step a). The product was collected as an off-white solid (600 mg, 82.5%). !
  • step c The title compound was prepared in the same manner as described for example 80 (step c) from 1- isopropyl-3-methyl-6-(lH-pyrrolo[2,3-b]pyridin-5-yl)-lH-indazole-4-carboxylic acid (50 mg, 0.149 mmol) and 3-(aminomethyl)-6-methyl-4-propylpyridin-2(lH)-one (26 mg, 0.149 mmol) wherein the contents were stirred at RT for 5h. The crude product was washed with DCM, filtered, and dried to afford the title compound as an off-white solid. (48 mg, 64 %). !
  • the title compound was prepared in the same manner as described for example 80 (step a) from methyl 6-bromo-l-isopropyl-3-methyl-lH-indazole-4-carboxylate (0.5 g, 1.60 mmol) and 4-(4,4,5,5- tetramethyl-l,3,2-dioxaborolan-2-yl)-lH-pyrazole (0.37 g, 1.92 mmol) wherein the reaction mixture was heated at 115 °C for 3 h.
  • the crude compound was purified silica gel chromatography (eluent: 50 % ethyl acetate ⁇ pet ether) to afford the title compound as an off-white solid. !
  • the title compound was prepared from methyl l-isopropyl-3-methyl-6-(lH-pyrazol-4-yl)-lH- indazole-4-carboxylate (0.2 g, 0.67 mmol) and L1OH.H 2 O (0.084 g, 2.01 mmol) in the same manner as described for example 80 (step b).
  • the product was collected as a white solid (0.2 g).
  • the title compound was prepared in the same manner as described for example 80 (step c) from 1 - isopropyl-3-methyl-6-(lH-pyrazol-4-yl)- lH-indazole-4-carboxylic acid (0.15 g, 0.52 mmol) and 3- (aminomethyl)-6-methyl-4-propylpyridin-2(lH)-one (0.09 g, 0.52 mmol) wherein the contents were stirred at RT for 5h.
  • the crude compound was purified by silica gel chromatography (eluent: 2 % MeOHYDCM) to afford the title compound as an off-white solid (60 mg, 25%).
  • the title compound was prepared in the same manner as described for example 76 from 6-bromo-N- ((l ,2-dihydro-4,6-dimethyl-2-oxopyridin-3-yl)methyl)- l -isopropyl-3-methyl-lH-indazole-4- carboxamide (300 mg, 0.696 mmol) and 2-memyl-5-(4,4,5,5-tetramethyl- l,3,2-dioxaborolan-2- yl)pyridine (183 mg, 0.835 mmol). The title compound was collected as 75 mg (24%); !
  • the title compound was prepared in the same manner as described for example 67 from 6-bromo-3-methyl- l - (l-methylethyl)-N-[(6-methyl-2-oxo-4-propyl-l,2-dihydro-3-pyridinyl)methyl]- lH-indazole-4-carboxamide (.079 g, 0.172 mmol) and [6-(dimethylamino)-3-pyridinyl]boronic acid (0.034 g, 0.206 mmol).
  • the produd was triturated from EtOAc spiked with DCM, and dried in a hi-vacuum oven for 4h.
  • the title compound was prepared in the same manner as described for example 67 from 6-bromo-3-methyl- l - (l-methylethyl)-N-[(6-methyl-2-oxo-4-propyl-l,2-dihydro-3-pyridinyl)methyl]- lH-indazole-4-carboxamide (.090 g, 0.196 mmol).
  • the crude product was purified by reverse phase HPLC (Gradient B: 10-60%. 8mir A: Water + .1% TFA. B: CH3CN + .1% TFA).
  • the isolated product was then treated with 1.0 g of Silicyck Carbonate resin, and filtered through celite washing with 10% MeOH/DCM.
  • the title compound was prepared in the same manner as described for example 67 from 6-bromo-3-methyl- l - (l-methylethyl)-N-[(6-methyl-2-oxo-4-propyl-l,2-dihydro-3-pyridinyl)methyl]- lH-indazole-4-carboxamide (.090 g, 0.196 mmol) and N,N-dimethyl-l-[4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)phenyl]methanamine (0.070 g, 0.235 mmol).
  • the title compound was prepared in the same manner as described for example 67 from 6-bromo-3-methyl-l- (l-methylethyl)-N-[(6-methyl-2-oxo-4-propyl-l,2-dihydro-3-pyridinyl)methyl]- lH-indazole-4-carboxamide (.10g, 0.218 mmol).
  • the title compound was prepared in the same manner as described for example 67 from 6-bromo-3- methyl- 1 -( 1 -methylethyl)-N- [(6-methyl-2-oxo-4-propyl- 1 ,2-dihydro-3 -pyridinyl)methyl] - 1 H- indazole-4-carboxamide (0.10g, 0.218 mmol) and l-methyl-4-[5-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)-2-pyridinyl]piperazine (0.086 g, 0.283 mmol).
  • the vessel was sealed, and the insoluble mixture was heated to 120 °C for 6 hours. Upon cooling down, the mixture was quenched with water, and dark grey solids that crashed out and were filtered.
  • DCM/MeOH (1 : 1) was added, the contents pre-absorbed on silica gel and purified by Si0 2 chromatography (eluent: gradient 0 to 80:20:2 DCM/MeOH/NH 4 OH).
  • the collected product was futher purified by reverse-phase HPLC (20% to 80% CH3CN in water with 0.1% TFA) which afforded the TFA salt. CH3CN was evaporated, and a saturated solution of sodium bicarbonate was added to the water layer.
  • the title compound was prepared from 6-bromo-N-[(4-ethyl-6-methyl-2-oxo-l,2-dihydro-3- pyridinyl)methyl]-l-(l-methylethyl)-lH-indazole-4-carboxamide (70 mg, 0.162 mmol) and the sodium salt of cyclopropanesulfmic acid (41.9 mg, 0.325 mmol) in the same manner as described for example 91. The product was collected an off-white solid (52 mg).
  • the title compound was prepared from 6-bromo-N-[(4-ethyl-6-methyl-2-oxo-l,2-dihydro-3- pyridinyl)methyl]-l-(l-methylethyl)-lH-indazole-4-carboxamide (70 mg, 0.162 mmol) and the sodium salt of methanesulfinic acid (33.5 mg, 0.325 mmol), in the same manner as described for example 91.
  • reaction vial was sealed anc the mixture was stirred with heating at 90 °C (heat block) for 18h. The contents were then irradiatec (microwave) at 130 °C for 2hr. After cooling to RT, the reaction mixture was poured onto 50 mL of water anc stirred. The contents were extracted with hot 30%THF/EtOAc (2x) (need to heat to break up emulsion). The combined organic layers were washed with brine, dried over MgSOzi, filtered, and concentrated in vacuo. ⁇ crude product was purified by silica gel chromatography (eluent: 5-75% chloroform (containing 10% 2M ammonia in methanol) and dichloromethane.
  • Methyl l-(l-methylethyl)-6-nitro-lH-indazole-4-carboxylate (1.39 g, 5.28 mmol) was dissolved in ethanol (70 mL) and hydrogenated using an H-Cube instrument (full H 2 mode and 10% Pd/C). The solvent was removed in vacuo and the residue was purified via silica gel chromatography (eluent: 0% to 40% gradient; EtOAc:Hex). The product was obtained as 1.05 g (85%).
  • Methyl 6-amino-l-(l-methylethyl)-lH-indazole-4-carboxylate (500 mg, 2.143 mmol) was dissolved in cone, hydrochloric acid (5 mL) and cooled in an ice water bath. A solution of sodium nitrite (155 mg, 2.251 mmol) in 2 mL of water was then added dropwise, and the contents were stirred for 90 min. The contents were added portion- wise to a solution of ca. 5 mL of S0 2 , copper(II) chloride (303 mg, 2.251 mmol) and acetic acid (20 mL). The contents were stirred at room temperature for 15 h, and then concentrated in vacuo.

Abstract

Herein are disclosed indazoles of formula (I) where the various groups are defined herein, and which are useful for treating cancer.

Description

INDAZOLES
FIELD OF THE INVENTION
This invention relates to substituted indazoles which inhibit EZH2 and thus are useful for inhibiting the proliferation of and/or inducing apoptosis in cancer cells.
BACKGROUND OF THE INVENTION
Epigenetic modifications play an important role in the regulation of many cellular processes including cell proliferation, differentiation, and cell survival. Global epigenetic modifications are common in cancer, and include global changes in DNA and/or histone methylation, dysregulation of non-coding RNAs and nucleosome remodeling leading to aberrant activation or inactivation of oncogenes, tumor suppressors and signaling pathways. However, unlike genetic mutations which arise in cancer, these epigenetic changes can be reversed through selective inhibition of the enzymes involved. Several methylases involved in histone or DNA methylation are known to be dysregulated in cancer. Thus, selective inhibitors of particular methylases will be useful in the treatment of proliferative diseases such as cancer.
EZH2 (enhancer of zeste homolog 2; human EZH2 gene: Cardoso, C, et al; European J of Human Genetics, Vol. 8, No. 3 Pages 174-180, 2000) is the catalytic subunit of the Polycomb Repressor Complex 2 (PRC2) which functions to silence target genes by tri-methylating lysine 27 of histone H3 (H3K27me3). Histone H3 is one of the five main histone proteins involved in the structure of chromatin in eukaryotic cells. Featuring a main globular domain and a long N-terminal tail, Histones are involved with the structure of the nucleosomes, a 'beads on a string' structure.
Histone proteins are highly post-translationally modified however Histone H3 is the most extensively modified of the five histones. The term "Histone H3" alone is purposely ambiguous in that it does not distinguish between sequence variants or modification state. Histone H3 is an important protein in the emerging field of epigenetics. where its sequence variants and variable modification states are thought to play a role in the dynamic and long term regulation of genes.
Increased EZH2 expression has been observed in numerous solid tumors including those of the prostate, breast, skin, bladder, liver, pancreas, head and neck and correlates with cancer aggressiveness, metastasis and poor outcome (Varambally et al., 2002; Kleer et al., 2003; Breuer et al., 2004; Bachmann et al., 2005; Weikert et al., 2005; Sudo et al., 2005; Bachmann et al., 2006). For instance, there is a greater risk of recurrence after prostatectomy in tumors expressing high levels of EZH2, increased metastasis, shorter disease-free survival and increased death in breast cancer patients with high EZH2 levels (Varambally et al., 2002; Kleer et al., 2003). More recently, inactivating mutations in UTX (ubiquitously transcribed tetratricopeptixe repeas X), a H3K27 demethylase which functions in opposition to EZH2, have been identified in multiple solid and hematological tumor types (including renal, glioblastoma, esophageal, breast, colon, non-small cell lung, small cell lung, bladder, multiple myeloma, and chronic myeloid leukemia tumors), and low UTX levels correlate with poor survival in breast cancer suggesting that loss of UTX function leads to increased H3K27me3 and repression of target genes (Wang et al., 2010). Together, these data suggest that increased H3K27me3 levels contribute to cancer aggressiveness in many tumor types and that inhibition of EZH2 activity may provide therapeutic benefit.
Numerous studies have reported that direct knockdown of EZH2 via siRNA or shRNA or indirect loss of EZH2 via treatment with the SAH hydrolase inhibitor 3-deazaneplanocin A (DZNep) decreases cancer cell line proliferation and invasion in vitro and tumor growth in vivo (Gonzalez et al., 2008, GBM 2009). While the precise mechanism by which aberrant EZH2 activity leads to cancer progression is not known, many EZH2 target genes are tumor suppressors suggesting that loss of tumor suppressor function is a key mechanism (refs). In addition, EZH2 overexpression in immortalized or primary epithelial cells promotes anchorage independent growth and invasion and requires EZH2 catalytic activity. (Kleer et al., 2003; Cao et al., 2008).
Thus, there is strong evidence to suggest that inhibition of EZH2 activity decreases cellular proliferation and invasion. Accordingly, compounds that inhibit EZH2 activity would be useful for the treatment of cancer. The indazoles of this invention provide such treatment.
SUMMARY OF THE INVENTION
In a first instance, this invention relates to compounds of formula (I)
Figure imgf000005_0001
wherein
X and Z are selected independently from the group consisting of hydrogen, (Ci-C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, unsubstituted or substituted (C3-C8)cycloalkyl, unsubstituted or substituted (C3-C8)cycloalkyl-(Ci-C8)alkyl or -(C2-C8)alkenyl, unsubstituted or substituted (C5- C8)cycloalkenyl, unsubstituted or substituted (C5-C8)cycloalkenyl-(Ci-C8)alkyl or -(C2-C8)alkenyl, (C6-Cio)bicycloalkyl, unsubstituted or substituted heterocycloalkyl, unsubstituted or substituted heterocycloalkyl-(Ci-C8)alkyl or -(C2-C8)alkenyl, unsubstituted or substituted aryl, unsubstituted or substituted aryl-(Ci-C8)alkyl or -(C2-C8)alkenyl, unsubstituted or substituted heteroaryl, unsubstituted or substituted heteroaryl-(C C8)alkyl or -(C2-C8)alkenyl, halo, cyano, -CORa, -C02Ra, -CONRaRb, - CONRaNRaRb, -SRa, -SORa, -S02Ra, -S02NRaRb, nitro, -NRaRb, -NRaC(0)Rb, -NRaC(0)NRaRb, - NRaC(0)ORa, -NRaS02Rb, -NRaS02NRaRb, -NRaNRaRb, -NRaNRaC(0)Rb, -NRaNRaC(0)NRaRb, - NRaNRaC(0)ORa, -ORa, -OC(0)Ra, and -OC(0)NRaRb;
Y is H or halo;
R1 is (CrC8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, unsubstituted or substituted (C3- C8)cycloalkyl, unsubstituted or substituted (C3-C8)cycloalkyl-(C1-C8)alkyl or -(C2-C8)alkenyl, unsubstituted or substituted (C5-C8)cycloalkenyl, unsubstituted or substituted (C5-C8)cycloalkenyl- (Ci-C8)alkyl or -(C2-C8)alkenyl, unsubstituted or substituted (C6-Cio)bicycloalkyl, unsubstituted or substituted heterocycloalkyl or -(C2-C8)alkenyl, unsubstituted or substituted heterocycloalkyl-(Ci- C8)alkyl, unsubstituted or substituted aryl, unsubstituted or substituted aryl-(Ci-C8)alkyl or - (C2-C8)alkenyl, unsubstituted or substituted heteroaryl, unsubstituted or substituted heteroaryl-(Ci- C8)alkyl or -(C2-C8)alkenyl, -CORa, -C02Ra, -CONRaRb, -CONRaNRaRb;
R3 is hydrogen, (Ci-C8)alkyl, cyano, trifluoromethyl, -NRaRb, or halo;
R6 is selected from the group consisting of hydrogen, halo, (Ci-Cg)alkyl, (C2-C8)alkenyl, - B(OH)2, substituted or unsubstituted (C2-C8)alkynyl, unsubstituted or substituted (C3-C8)cycloalkyl, unsubstituted or substituted (C3-C8)cycloalkyl-(Ci-Cg)alkyl, unsubstituted or substituted (C5- C8)cycloalkenyl, unsubstituted or substituted (C5-C8)cycloalkenyl-(Ci-C8)alkyl, (C6-Cio)bicycloalkyl, unsubstituted or substituted heterocycloalkyl, unsubstituted or substituted heterocycloalkyl-(d- C8)alkyl, unsubstituted or substituted aryl, unsubstituted or substituted aryl-(Ci-C8)alkyl, unsubstituted or substituted heteroaryl, unsubstituted or substituted heteroaryl-(Ci-C8)alkyl, cyano, - CORa, -C02Ra, -CONRaRb, -CONRaNRaRb, -SRa, -SORa, -S02Ra, -S02NRaRh, nitro, -NRaRb, - NRaC(0)Rb, -NRaC(0)NRaRb, -NRaC(0)ORa, -NRaS02Rb, -NRaS02NRaRb, -NRaNRaR , - NRaNRaC(0)Rb, -NRaNRaC(0)NRaRb, -NRaNRaC(0)ORa, -ORa, -OC(0)Ra, -OC(0)NRaRb;
wherein any (Ci-Cg)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, cycloalkyl, cycloalkenyl, bicycloalkyl, heterocycloalkyl, aryl, or heteroaryl group is optionally substituted by 1, 2 or 3 groups independently selected from the group consisting of-0(Ci-C6)alkyl(Rc)i_2, -S(d- C6)alkyl(Rc)1.2, -(C1-C6)alkyl(Rc)1.2, (Ci-C8)alkyl-heterocycloalkyl, (C3-C8)cycloalkyl- heterocycloalkyl, halo, (d-d)alkyl, (C3-C8)cycloalkyl, (C5-C8)cycloalkenyl,
(Ci-C6)haloalkyl, cyano, -CORa, -C02Ra,-CONRaRb, -SRa, -SORa, -S02Ra, -S02NRaRb, nitro, -NRaRb, -NRaC(0)Rb, -NRaC(0)NRaRb, -NRaC(0)ORa, -NRaS02Rb, -NRaS02NRaRb, -ORa, -OC(0)Ra, -OC(0)NRaRb, heterocycloalkyl, aryl, heteroaryl, aryl(Ci-C4)alkyl, and heteroaryl(Ci-C4)alkyl;
wherein any aryl or heteroaryl moiety of said aryl, heteroaryl, aryl(Ci-C4)alkyl, or heteroaryl(Ci-C4)alkyl is optionally substituted by 1, 2 or 3 groups independently selected from the group consisting of halo, (Ci-C6)alkyl, (C3-C8)cycloalkyl, (C5-C8)cycloalkenyl, (C C6)haloalkyl, cyano, -CORa, -C02Ra, -CONR Rb, -SRa, -SORa, -S02Ra, -S02NRaRb, nitro, -NRaRb, -NRaC(0)Rb,
-NRaC(0)NR Rb, -NRaC(0)ORa, -NRaS02Rb, -NRaS02NRaRb, -ORa, -OC(0)Ra, and -OC(0)NRaRb;
Ra and Rb are each independently hydrogen, (Ci-Cg)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, (C3-C8)cycloalkyl, (C5-C8)cycloalkenyl, (C6-Ci0)bicycloalkyl, heterocycloalkyl, aryl, heteroaryl, wherein said (Ci-C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, cycloalkyl, cycloalkenyl, bicycloalkyl, heterocycloalkyl ,aryl or heteroaryl group is optionally substituted by 1, 2 or 3 groups independently selected from halo, hydroxyl, (Ci-C4)alkoxy, amino, (Ci-C4)alkylamino,
((d-d)alkyl)((d-d)alkyl)amino, -C02H, -C02(d-C4)alkyl, -CONH2 -CONH(d-C4)alkyl, - CON((d-C4)alkyl)((d-C4)alkyl), -S02(d-d)alkyl, -S02NH2 ,-S02NH(d-d)alkyl, or - S02N((d-C4)alkyl)((d-C4)alkyl);
or Ra and Rb taken together with the nitrogen to which they are attached represent a 5-8 membered saturated or unsaturated ring, optionally containing an additional heteroatom selected from oxygen, nitrogen, and sulfur, wherein said ring is optionally substituted by 1, 2 or 3 groups independently selected from (d-d)alkyl, (d-d)haloalkyl, amino, (d-d)alkylamino,
((d-d)alkyl)((d-C4)alkyl)amino, hydroxyl, oxo, (d-d)alkoxy, and (C1-C4)alkoxy(C1-C4)alkyl, wherein said ring is optionally fused to a (d-d)cycloalkyl, heterocycloalkyl, aryl, or heteroaryl ring; or a and Rb taken together with the nitrogen to which they are attached represent a 6- to 10- membered bridged bicyclic ring system optionally fused to a (C3-C8)cycloalkyl, heterocycloalkyl, aryl, or heteroaryl ring;
each Rc is independently (d-C^alkylamino, -NRaS02Rb, -SORa, -S02Ra, -NRaC(0)ORa, - NRaRh, or -C02Ra;
or a salt thereof.
Another aspect of the invention relates to the exemplified compounds.
In a further iteration of this invention it relates to a method of treating cancer.
Another aspect of the invention are pharmaceutical preparations comprising compounds of formula (I) and pharmaceutically acceptable excipients.
In a fourth aspect, there is provided the use of a compound of formula (I) or a
pharmaceutically acceptable salt or solvate thereof, in the preparation of a medicament for use in the treatment of a disorder mediated by inhibiting EZH2, such as inducing apoptosis in cancer cells.
In a fifth aspect there is provided methods of co-administering the presently invented compounds of formula (I) with another active ingredients.
DETAILED DESCRIPTION OF THE INVENTION
For the avoidance of doubt, unless otherwise indicated, the term "substituted" means substituted by one or more defined groups. In the case where groups may be selected from a number of alternative groups the selected groups may be the same or different.
The term "independently" means that where more than one substituent is selected from a number of possible substituents, those substituents may be the same or different.
An "effective amount" means that amount of a drug or pharmaceutical agent that will elicit the biological or medical response of a tissue, system, animal or human that is being sought, for instance, by a researcher or clinician. Furthermore, the term "therapeutically effective amount" means any amount which, as compared to a corresponding subject who has not received such amount, results in improved treatment, healing, prevention, or amelioration of a disease, disorder, or side effect, or a decrease in the rate of advancement of a disease or disorder. The term also includes within its scope amounts effective to enhance normal physiological function.
As used herein the term "alkyl" refers to a straight- or branched-chain hydrocarbon radical having the specified number of carbon atoms, so for example, as used herein, the terms "Ci_C8alkyl" refers to an alkyl group having at least 1 and up to 8 carbon atoms respectively. Examples of such branched or straight-chained alkyl groups useful in the present invention include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, isobutyl, n-butyl, t-butyl, n-pentyl, isopentyl, n-hexyl, n-heptyl, and n-octyl and branched analogs of the latter 5 normal alkanes. The term "alkoxy" as used herein means -0(Ci_C8alkyl) including -OCH3, -OCH2CH3 and -
OC(CH3)3 and the like per the definition of alkyl above.
The term "alkylthio" as used herein is meant -S(Ci_C8alkyl) including -SCH3, -SCH2CH3 and the like per the definition of alkyl above.
The term "acyloxy" means -OC(0)Ci.Cgalkyl and the like per the definition of alkyl above.
"Acylamino" means-N(H)C(0)Ci_C8alkyl and the like per the definition of alkyl above.
"Aryloxy" means -O(aryl), -0(substituted aryl), -O(heteroaryl) or -0(substituted heteroaryl).
"Arylamino" means -NH(aryl), -NH(substituted aryl), -NH(heteroaryl) or -NH(substituted heteroaryl), and the like.
When the term "alkenyl" (or "alkenylene") is used it refers to straight or branched hydrocarbon chains containing the specified number of carbon atoms and at least 1 and up to 5 carbon-carbon double bonds. Examples include ethenyl (or ethenylene) and propenyl (or propenylene).
When the term "alkynyl" (or "alkynylene") is used it refers to straight or branched hydrocarbon chains containing the specified number of carbon atoms and at least 1 and up to 5 carbon-carbon triple bonds. Examples include ethynyl (or ethynylene) and propynyl (or
propynylene).
"Haloalkyl" refers to an alkyl group group that is substituted with one or more halo substituents, suitably from 1 to 6 substituents. Haloalkyl includes trifluoromethyl.
When "cycloalkyl" is used it refers to a non-aromatic, saturated, cyclic hydrocarbon ring containing the specified number of carbon atoms. So, for example, the term "C3_Qcycloalkyl" refers to a non-aromatic cyclic hydrocarbon ring having from three to eight carbon atoms. Exemplary "C3-
Cgcycloalkyl" groups useful in the present invention include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and cyclooctyl.
The term "Cs-Cgcycloalkenyl" refers to a non-aromatic monocyclic carboxycyclic ring having the specified number of carbon atoms and up to 3 carbon-carbon double bonds. "Cycloalkenyl" includes by way of example cyclopentenyl and cyclohexenyl.
Where "Cs-Cgheterocycloalkyl" is used, it means a non-aromatic heterocyclic ring containing the specified number of ring atoms being, saturated or having one or more degrees of unsaturation and containing one or more heteroatom substitutions independently selected from O, S and N. Such a ring may be optionally fused to one or more other "heterocyclic" ring(s) or cycloalkyl ring(s). Examples are given herein below.
"Aryl" refers to optionally substituted monocyclic or polycarbocyclic unfused or fused groups having 6 to 14 carbon atoms and having at least one aromatic ring that complies with Huckel's Rule. Examples of aryl groups are phenyl, biphenyl, naphthyl, anthracenyl, phenanthrenyl, and the like, as further illustrated below.
"Heteroaryl" means an optionally substituted aromatic monocyclic ring or polycarbocyclic fused ring system wherein at least one ring complies with Hiickel's Rule, has the specified number of ring atoms, and that ring contains at least one heteratom independently selected from N, O and S. Examples of "heteroaryl" groups are given herein below.
The term "optionally" means that the subsequently described event(s) may or may not occur, and includes both event(s), which occur, and events that do not occur.
Herein, the term "pharmaceutically-acceptable salts" refers to salts that retain the desired biological activity of the subject compound and exhibit minimal undesired toxicological effects. These pharmaceutically-acceptable salts may be prepared in situ during the final isolation and purification of the compound, or by separately reacting the purified compound in its free acid or free base form with a suitable base or acid, respectively.
While the compounds encompassed by the general structure of formula (I) as defined herein are believed to be useful for inducing apoptosis in cancer cells, some of these compounds are more active that others. In that vein, the following subgroups delineate certain compounds believed to have greater potency or other properties which suggest they may be a better choice for use in therapy, versus other. Those subgroups are represented as follows:
Subgroup A
X and Z are selected from the group consisting of (Ci-C8)alkyl, (C3-C8)cycloalkyl, heterocycloalkyl, aryl, heteroaryl, -NRaRb, and -ORa;
Y is H or F;
R1 is selected from the group consisting of (C1-C8)alkyl, (C3-C8)cycloalkyl, heterocycloalkyl, aryl, and heteroaryl;
R3 is selected from the group consisting of hydrogen, (Ci-Cg)alkyl, cyano, trifluoromethyl, -
NRaRb, and halo;
R6 is selected from the group consisting of hydrogen, halo, cyano, trifluoromethyl, amino, (Ci-C8)alkyl, (C3-C8)cycloalkyl, aryl, heteroaryl, acylamino; (C2-C8)alkynyl, arylalkynyl, heteroarylalkynyl, -S02Ra, -S02NRaRb , and -NRaS02Rb ;
wherein any (Ci-C8)alkyl, (C3-C8)cycloalkyl, (C2-C8)alkynyl, arylalkynyl, heteroarylalkynyl group is optionally substituted by 1, 2 or 3 groups independently selected from -0(Ci- C6) ky\( c) 2, -S(Ci-C6)alkyl(Rc)12, -(C1-C6)alkyl(Rc)12, (Q-C^alkyl-heterocycloalkyl, (C3-C8)cycloalkyl-heterocycloalkyl, halo, (Ci-C6)alkyl, (C3-C8)cycloalkyl,
(C5-C8)cycloalkenyl, (C1-C6)haloalkyl, cyano, -CORa, -C02Ra, -CONRaRb, -SRa, -SORa, - S02Ra, -S02NRaRb, nitro, -NRaRb, -NRaC(0)Rb, -NRaC(0)NRaRb, -NRaC(0)ORa, - NRaS02Rb, -NRaS02NRaRb, -ORa, -OC(0)Ra, -OC(0)NRaRb, heterocycloalkyl, aryl, heteroaryl, aryl(Ci-C4)alkyl, and heteroaryl(Ci-C4)alkyl;
each Rc is independently (Ci-C4)alkylamino, -NRaS02Rb, -SORa, -S02Ra, -NRaC(0)ORa,
, or -C02Ra;
Ra and Rb are each independently hydrogen, (Ci-C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl,
(C3-C8)cycloalkyl, (C5-C8)cycloalkenyl, (C6-Ci0)bicycloalkyl, heterocycloalkyl, aryl, heteroaryl, wherein said (Ci-Cg)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, cycloalkyl, cycloalkenyl, bicycloalkyl, heterocycloalkyl ,aryl or heteroaryl group is optionally substituted by 1, 2 or 3 groups independently selected from halo, hydroxyl, (Ci-C4)alkoxy, amino, (Ci-C4)alkylamino,
((Ci-C4)alkyl)((Ci-C4)alkyl)amiiio, -C02H, -C02(d-C4)alkyl, -CONH2, -CONH(Ci-C4)alkyl, - CON((Ci-C4)alkyl)((Ci-C4)alkyl), -S02(C!-C4)alkyl, -SOzNHz -SOzNHCd-C^alkyl, and - S02N((Ci-C )allQrl)((Ci-C )alkyl);
or Ra and Rb taken together with the nitrogen to which they are attached represent a 5-8 membered saturated or unsaturated ring, optionally containing an additional heteroatom selected from oxygen, nitrogen, and sulfur, wherein said ring is optionally substituted by 1, 2 or 3 groups independently selected from (Ci-C )alkyl, (Ci-C )haloalkyl, amino, (Ci-C )alkylamino,
((Ci-C )alkyl)((Ci-C )alkyl)amino, hydroxyl, oxo, (Ci-C )alkoxy, and (Ci-C )alkoxy(Ci-C )alkyl, wherein said ring is optionally fused to a (C3-C8)cycloalkyl, heterocycloalkyl, aryl, or heteroaryl ring; or Ra and Rb taken together with the nitrogen to which they are attached represent a 6- to 10 membered bridged bicyclic ring system optionally fused to a (C3-C8)cycloalkyl, heterocycloalkyl, aryl, or heteroaryl ring. An aryl or heteroaryl group in this particular subgroup A is selected independently from the group consisting of furan, thiophene, pyrrole, oxazole, thiazole, imidazole, pyrazole, oxadiazole, thiadiazole, triazole, tetrazole, benzofuran, benzothiophene, benzoxazole, benzothiazole, phenyl, pyridine, pyridazine, pyrimidine, pyrazine, triazine, tetrazine, quinoline, e, quinoxaline, and naphthyridine or another aryl or heteroaryl group as follows
Figure imgf000010_0001
wherein in (1),
A is O NH, or S; B is CH or N, and C is hydrogen or Ci-C8 alkyl; or
Figure imgf000010_0002
wherein in (2),
D is N or C optionally substituted by hydrogen or Ci-C8 alkyl; or
Figure imgf000011_0001
wherein in (3),
E is NH or CH2; F is O or CO; and G is NH or CH2; or
Figure imgf000011_0002
wherein in (4),
J is O S or CO; or
Figure imgf000011_0003
wherein in (5),
Q is CH or ;
M is CH or N; and
L/(5) is hydrogen, halo, amino, cyano, (C C8)alkyl, (C3-C8)cycloalkyl, -CORa, -C02Ra, - CONRaRb, -CONRaNRaRb, -S02Ra, -S02NRaRb, -NRaRb, -NRaC(0)Rb,-NRaS02Rb, - NRaS02NRaRb, -NRaNRaRb, -NRaNRaC(0)Rb, -NRaNRaC(0)NRaRb, -ORa,
wherein any (Ci-C8)alkyl, (C3-C8)cycloalkyl, group is optionally substituted by 1, 2 or 3 groups independently selected from (Ci-C6)alkyl, (C3-C8)cycloalkyl, (C5-C8)cycloalkenyl, (C1-C6)haloalkyl, cyano, -CORa, -C02Ra, -CONRaRb, -SRa, -SORa, -S02Ra, -S02NRaRb, nitro, -NRaRb, -NRaC(0)Rb, -NRaC(0)NRaRb, -NRaC(0)ORa, -NRaS02Rb, -NRaS02NRaRb, -0Ra, -OC(0)Ra, -OC(0)NRaRb; wherein Ra and Rb are defined as above; or
Figure imgf000011_0004
wherein in 6,
L/(6) is NH or CH2; or
Figure imgf000012_0001
wherein in 7,
M/(7) is hydrogen, halo, amino, cyano, (Ci-C8)alkyl, (C3-C8)cycloalkyl, heterocycloalkyl, -CORa, -C02Ra, -CONRaRb, -CONRaNRaRb, -S02Ra, -S02NRaRb, - NRaRb, -NRaC(0)Rb,-NRaS02Rb, -NRaS02NRaRb, -NRaNRaRb, -NRaNRaC(0)Rb, -
NRa RaC(0)NRaRb, -ORa,
wherein any (Ci-Cg)alkyl, (C3-C8)cycloalkyl, heterocycloalkyl group is optionally substituted by 1, 2 or 3 groups independently selected from (Ci-C6)alkyl, (C3-C8)cycloalkyl, (C5-C8)cycloalkenyl, (Ci-C6)haloalkyl, cyano, -CORa, -C02Ra, -CONRaRb, -SRa, -SORa, - S02Ra, -S02NRaRb, nitro, -NRaRb, -NRaC(0)Rb, -NRaC(0)NRaRb, -NRaC(0)ORa, -
NRaS02Rb, -NRaS02NRaRb, -ORa, -OC(0)Ra, -OC(0)NRaRb; wherein Ra andRb are defined as above; or
Figure imgf000012_0002
wherein in (8),
P is CH2, NH, O, or S; Q/(8) is CH or N; and n is 0-2; or
Figure imgf000012_0003
wherein in (9),
S/(9) and T(9) is C, or S/(9) is C and T(9) is N, or S/(9) is N and T/(9) is C;
R is hydrogen, amino, methyl, trifluoromethyl, halo;
U is hydrogen, halo, amino, cyano, nitro, trifluoromethyl, (Ci-C8)alkyl, (C3-C8)cycloalkyl, - CORa, -C02Ra, -CONRaRb, -S02Ra, -S02NRaRb, -NRaRb, -NRaC(0)Rb,-NRaS02Rb, - NRaS02NRaRb, -NRaNRaRb, -NRaNRaC(0)Rb, , -ORa, 4-(lH-pyrazol-4-yl),
wherein any (Ci-C8)alkyl, (C3-C8)cycloalkyl, group is optionally substituted by 1, 2 or
3 groups independently selected from (Ci-C6)alkyl, (C3-C8)cycloalkyl, (C5-C8)cycloalkenyl, (C C6)haloalkyl, cyano, -CORa, -C02Ra,-CONRaRb, -SRa, -SORa, -S02Ra, -S02NRaRb, nitro, -NRaRb, -NRaC(0)Rb, -NRaC(0)NRaRb, -NRaC(0)ORa, -NRaS02Rb, -NRaS02NRaRb, -ORa, -OC(0)Ra, -OC(0)NRaRb; wherein Ra and Rb are defined as above.
Subgroup B
X and Z are selected independently from the group consisting of (C C8)alkyl, (C3- C8)cycloalkyl, heterocycloalkyl, aryl, heteroaryl, -NR Rb, and -ORa;
Y is H;
R1 is (Ci-Cg)alkyl, (C3-C8)cycloalkyl, or heterocycloalkyl;
R3 is hydrogen, (Ci-Cg)alkyl or halo;
R6 is hydrogen, halo, cyano, trifluoromethyl, amino, (Ci-Cg)alkyl, (d-d)cycloalkyl;, aryl, heteroaryl, acylamino; (C2-C8)alkynyl, arylalkynyl, heteroarylalkynyl; -S02Ra; -S02NRaRb, or - NRaS02Rb;
wherein any (Ci-C8)alkyl, (C3-C8)cycloalkyl, (C2-Cg)alkynyl, arylalkynyl, heteroarylalkynyl group is optionally substituted by 1 , 2 or 3 groups independently selected from halo, (Ci-C6)alkyl, (C3-C8)cycloalkyl, (C5-C8)cycloalkenyl, (Ci-C6)haloalkyl, cyano, - CORa, -C02Ra, -CONRaRb, -SRa, -SORa, -S02Ra, -S02NRaRb, nitro, -NRaRb, -
NRaC(0)Rb, -NRaC(0)NRaRb, -NRaC(0)ORa, -NRaS02Rb, -NRaS02NRaRb, -ORa, - OC(0)Ra, -OC(0)NRaRb, heterocycloalkyl, aryl, heteroaryl, aryl(Ci-C4)alkyl, and heteroaryl(Ci-C4)alkyl;
Ra and Rb are each independently hydrogen, (Ci-C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, (C3-C8)cycloalkyl, (C5-C8)cycloalkenyl, (d-do)bicycloalkyl, heterocycloalkyl, aryl, heteroaryl, wherein said (Ci-C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, cycloalkyl, cycloalkenyl, bicycloalkyl, heterocycloalkyl ,aryl or heteroaryl group is optionally substituted by 1, 2 or 3 groups independently selected from halo, hydroxyl, (Ci-C4)alkoxy, amino, (C1-C4)alkylamino,
((d-d)alkyl)((d-d)alkyl)amino, -C02H, -C02(C C4)alkyl, -CONH2 -CONH(C1-C4)alkyl, - CON((d-d)alkyl)((d-d)alkyl), -S02(C C4)alkyl, -S02NH2, -S02NH(C C4)alkyl, and - S02N((d-d)alkyl)((d-d)alkyl);
or Ra and Rb taken together with the nitrogen to which they are attached represent a 5-8 membered saturated or unsaturated ring, optionally containing an additional heteroatom selected from oxygen, nitrogen, and sulfur, wherein said ring is optionally substituted by 1, 2 or 3 groups independently selected from (Ci-C )alkyl, (Ci-C )haloalkyl, amino, (Ci-C )alkylamino,
((C1-C4)alkyl)((C1-C4)alkyl)amino, hydroxyl, oxo, (d-d)alkoxy, and (d-C4)alkoxy(d-C4)alkyl, wherein said ring is optionally fused to a (C3-C8)cycloalkyl, heterocycloalkyl, aryl, or heteroaryl ring; or Ra and Rb taken together with the nitrogen to which they are attached represent a 6- to 10- membered bridged bicyclic ring system optionally fused to a (C3-C8)cycloalkyl, heterocycloalkyl, aryl, or heteroaryl ring. Aryl and heteroaryl in this definition are selected from the group consisting of furan, thiophene, pyrrole, oxazole, thiazole, imidazole, pyrazole, oxadiazole, thiadiazole, triazole, tetrazole, benzofuran, benzothiophene, benzoxazole, benzothiazole, phenyl, pyridine, pyridazine, pyrimidine, pyrazine, triazine, tetrazine, quinoline, cinnoline, quinazoline, quinoxaline, and
a compound of or another aryl or heteroaryl group as follows:
Figure imgf000014_0001
wherein in (1),
A is 0 NH, or S; B is CH or N, and C is hydrogen or Ci-Cg alkyl; or
Figure imgf000014_0002
wherein in (2),
D is N or C o tionally substituted by hydrogen or Ci-Cg alkyl; or
Figure imgf000014_0003
wherein in (3),
E is NH or CH2; F is O or CO; and G is NH or CH2; or
Figure imgf000014_0004
wherein in (4),
J is O S or CO; or
Figure imgf000014_0005
wherein in (5),
Q is CH or N;
M is CH or N; and
L/(5) is hydrogen, halo, amino, cyano, (Ci-Cg)alkyl, (C3-C8)cycloalkyl, -CORa, -C02Ra,
CONRaRb, -CONRaNRaRb, -S02Ra, -S02NRaRb, -NRaRb, -NRaC(0)Rb,-NRaS02Rb,
NRaS02NRaRb, -NRaNRaRb, -NRaNRaC(0)Rb, -NRaNRaC(0)NRaRb, -ORa,
wherein any (Ci-Cg)alkyl, (C3-Cg)cycloalkyl, group is optionally substituted by 1 , 2 or 3 groups independently selected from (Ci-C6)alkyl, (C3-C8)cycloalkyl, (C5-Cg)cycloalkenyl, (C1-C6)haloalkyl, cyano, -CORa, -C02Ra, -CONRaRb, -SRa, -SORa, -S02Ra, -S02NRaRb, nitro, -NRaRb, -NRaC(0)Rb, -NRaC(0)NRaRb, -NRaC(0)ORa, -NRaS02Rb, -NRaS02NRaRb, -ORa, -OC(0)Ra, -OC(0)NRaRb,
wherein Ra and Rb are defined as above; or
Figure imgf000015_0001
wherein in 6,
L/(6) is NH or CH2; or
Figure imgf000015_0002
wherein in 7,
M/(7) is hydrogen, halo, amino, cyano, (Ci-C8)alkyl, (C3-C8)cycloalkyl, heterocycloalkyl, -CORa, -C02Ra, -CONRaRb, -CONRaNRaRb, -S02Ra, -S02NRaRb, - NRaRb, -NRaC(0)Rb,-NRaS02Rb, -NRaS02NRaRb, -NRaNRaRb, -NRaNRaC(0)Rb, -
NRa RaC(0)NRaRb, -ORa,
wherein any (Ci-Cg)alkyl, (C3-Cg)cycloalkyl, heterocycloalkyl group is optionally substituted by 1, 2 or 3 groups independently selected from (Ci-C6)alkyl, (C3-C8)cycloalkyl, (C5-C8)cycloalkenyl, (Ci-C6)haloalkyl, cyano, -CORa, -C02Ra, -CONRaRb, -SRa, -SORa, - S02Ra, -S02NRaRb, nitro, -NRaRb, -NRaC(0)Rb, -NRaC(0)NRaRb, -NRaC(0)ORa, -
NRaS02Rb, -NRaS02NRaRb, -ORa, -OC(0)Ra, -OC(0)NRaRb; wherein Ra andRb are defined as above; or
Figure imgf000015_0003
wherein in (8),
P is CH2, NH, O, or S; Q/(8) is CH or N; and n is 0-2; or
Figure imgf000016_0001
wherein in (9),
S/(9) and T(9) is C, or S/(9) is C and T(9) is N, or S/(9) is N and T/(9) is C;
R is hydrogen, amino, methyl, trifluoromethyl, halo;
U is hydrogen, halo, amino, cyano, nitro, trifluoromethyl, (Ci-C8)alkyl, (C3-C8)cycloalkyl, - CORa, -C02Ra, -CONRaRb, -S02Ra, -S02NRaRb, -NRaRb, -NRaC(0)Rb ,-NRaS02Rb, - NRaS02NRaRb, -NRaNRaRb, -NRaNRaC(0)Rb, , -ORa, 4-(lH-pyrazol-4-yl),
wherein any (Ci-C8)alkyl, (C3-C8)cycloalkyl, group is optionally substituted by 1, 2 or 3 groups independently selected from (Ci-C6)alkyl, (C3-C8)cycloalkyl, (C5-C8)cycloalkenyl, (C1-C6)haloalkyl, cyano, -CORa, -C02Ra,-CONRaRb, -SRa, -SORa, -S02Ra, -S02NRaRb, nitro, -NRaRb, -NRaC(0)Rb, -NRaC(0)NRaRb, -NRaC(0)ORa, -NRaS02Rb, -NRaS02NRaRb, -ORa, -OC(0)Ra, -OC(0)NRaRb, wherein Ra andRb are defined as above.
Subgroup C
X is methyl, ethyl, n-propyl, isopropyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, phenyl, trifluoromethyl, tetrahydropyran, hydroxymethyl, methoxymethyl, or benzyl;
Y is H;
Z is methyl, ethyl, n-propyl, isopropyl, trifluoromethyl, or benzyl;
R1 is isopropyl, tert-butyl, cyclobutyl, cyclopentyl, cyclohexyl, (l-methylethyl)cyclopropyl, l,l-dioxo-tetrahydrothiophene-3-yl, 1 -Me-piperidin-4-yl, tetrahydrofuran-3-yl, tetrahydropyran-4-yl, jVN-dimethyl- 1 -propanaminyl, benzyl, or 4-pyridyl;
R3 is H, methyl, or Br; and
R6 is methyl, bis(l, l-dimethylethyl), bis(l-methylethyl), cyclopropyl, propyl, dimethylamino, ethylamino, (2-hydroxyethyl)amino, 2-propen-l-ylamino, 1 -piperazinyl, 1-piperidinyl, 4-morpholinyl, 4-piperidinylamino, tetrahydro-2H-pyran-4-ylamino, phenylamino, (phenylmethyl)amino, (4- pyridinylmethyl)amino, [2-(2-pyridinylamino)ethyl]amino, 2-(dimethylamino)ethyl] amino, 4- pyridinylamino , 4-(aminocarbonyl)phenyl]amino, 3 -hydroxy-3 -methyl- 1-butyn-l-yl, 4- pyridinylethynyl, phenylethynyl, 2-furanyl, 3-thienyl; lH-pyrazol-4-yl, lH-indazol-5-yl, lH-indazol- 6-yl, 3-methyl-lH-indazol-5-yl, lH- l,2,3-benzotriazol-5-yl, 2-oxo-2,3-dihydro-lH-benzimidazol-5- yl, 2-oxo-2,3-dihydro-lH-indol-5-yl, 2-oxo-2,3-dihydro-lH-indol-6-yl, 2,l,3-benzoxadiazol-5-yl, 2- amino-6-quinazolinyl, 2,4-dioxo-l,2,3,4-tetrahydro-5-pyrimidinyl, 2-amino-5-pyrimidinyl, 7-oxo- l,5,6,7-tetrahydro-l,8-naphthyridin-3-yl, phenyl, 2-methylphenyl, 2-nitrophenyl, 2-phenylethyl, 3- aminophenyl, 4-aminophenyl, 4-chlorophenyl, 4- fluorophenyl, 4-(methyloxy)phenyl, 3- (acetylamino)phenyl, 4-(acetylamino)phenyl, 4-(aminocarbonyl)phenyl, 4-(lH-pyrazol-4-yl)phenyl,
4- (aminosulfonyl)phenyl, 4-(methylsulfonyl)phenyl, 4-[(dimethylamino)sulfonyl]phenyl, 4- [(methylamino)carbonyl]phenyl, 4- [(methylamino)sulfonyl]phenyl, 4- [(methylsulfonyl)amino]phenyl,
3- pyridinyl, 4-pyridinyl, 2-(4-morpholinyl)-4-pyridinyl, 2-amino-4-pyridinyl, 5-(methyloxy)-3- pyridinyl, 5-(methylsulfonyl)-3-pyridinyl, 5-[(cyclopropylsulfonyl)amino]-6-(methyloxy)-3-pyridinyl,
5- [(phenylsulfonyl)amino]-3-pyridinyl, 6-(4-methyl- 1 -piperazinyl)-3 -pyridinyl, 6-(4-morpholinyl)-3- pyridinyl, 6-(acetylamino)-3-pyridinyl, 6-(dimethylamino)-3-pyridinyl, 6-(methyloxy)-3-pyridinyl, 6- [(methylamino)carbonyl]-3-pyridinyl, 6-[(methylamino)sulfonyl]-3-pyridinyl, 6-methyl-3 -pyridinyl,
4- pyridinyloxy.
In one aspect, this invention also relates to the exemplified compounds.
Individual compounds can be found in the Examples set out below.
By the term "co-administering" and derivatives thereof as used herein is meant either simultaneous administration or any manner of separate sequential administration of one or more additional pharmaceutically active compounds, whether for treating cancer, the side effects of cancer or cancer therapy, or some other disease. Preferably, if the administration is not simultaneous, the compounds are administered in a close time proximity to each other. Furthermore, it does not matter if the compounds are administered in the same dosage form, e.g. one compound may be administered topically and another compound may be administered orally.
In certain embodiments, compounds according to Formula I may contain an acidic functional group, one acidic enough to form salts. Representative salts include pharmaceutically-acceptable metal salts such as sodium, potassium, lithium, calcium, magnesium, aluminum, and zinc salts;
carbonates and bicarbonates of a pharmaceutically-acceptable metal cation such as sodium, potassium, lithium, calcium, magnesium, aluminum, and zinc; pharmaceutically-acceptable organic primary, secondary, and tertiary amines including aliphatic amines, aromatic amines, aliphatic diamines, and hydroxy alkylamines such as methylamine, ethylamine, 2-hydroxyethylamine, diethylamine, triethylamine, ethylenediamine, ethanolamine, diethanolamine, and cyclohexylamine.
In certain embodiments, compounds according to Formula (I) may contain a basic functional group and are therefore capable of forming pharmaceutically-acceptable acid addition salts by treatment with a suitable acid. Suitable acids include pharmaceutically-acceptable inorganic acids and pharmaceutically-acceptable organic acids. Representative pharmaceutically-acceptable acid addition salts include hydrochloride, hydrobromide, nitrate, methylnitrate, sulfate, bisulfate, sulfamate, phosphate acetate, hydroxyacetate, phenylacetate, propionate, butyrate, isobutyrate, valerate, maleate, hydroxymaleate, acrylate, fumarate, malate, tartrate, citrate, salicylate, />-aminosalicyclate, glycollate, lactate, heptanoate, phthalate, oxalate, succinate, benzoate, o-acetoxybenzoate, chlorobenzoate, methylbenzoate, dinitrobenzoate, hydroxybenzoate, methoxybenzoate, mandelate, tannate, formate, stearate, ascorbate, palmitate, oleate, pyruvate, pamoate, malonate, laurate, glutarate, glutamate, estolate, methanesulfonate (mesylate), ethanesulfonate (esylate), 2-hydroxyethanesulfonate, benzenesulfonate (besylate), >-aminobenzenesulfonate, / toluenesulfonate (tosylate) and napthalene- 2-sulfonate.
All tautomeric forms of the compounds described herein, including mixtures thereof, are intended to be encompassed within the scope of the invention. Generally, the compounds exemplified herein have been assigned names based on the structure of the tautomer of formaula (IA). It should be understood that any reference to named compounds of this invention is intended to encompass all tautomers of the named compounds and any mixtures of tautomers of the named compounds.
The compounds of formula (I) may be prepared in crystalline or non-crystalline form, and, if crystalline, may optionally be solvated, e.g. as the hydrate. This invention includes within its scope stoichiometric solvates (e.g. hydrates) as well as compounds containing variable amounts of solvent (e.g. water).
Certain of the compounds described herein may contain one or more chiral atoms, or may otherwise be capable of existing as two enantiomers. The compounds claimed below include mixtures of enantiomers as well as purified enantiomers or enantiomerically enriched mixtures. Also included within the scope of the invention are the individual isomers of the compounds represented by formula (I), or claimed below, as well as any wholly or partially equilibrated mixtures thereof. The present invention also covers the individual isomers of the claimed compounds as mixtures with isomers thereof in which one or more chiral centers are inverted.
Where there are different isomeric forms they may be separated or resolved one from the other by conventional methods, or any given isomer may be obtained by conventional synthetic methods or by stereospecific or asymmetric syntheses.
While it is possible that, for use in therapy, a compound of formula (I), as well as salts, solvates and the like, may be administered as a neat preparation, i.e. no additional carrier, the more usual practice is to present the active ingredient confected with a carrier or diluent. Accordingly, the invention further provides pharmaceutical compositions, which includes a compound of formula (I) and salts, solvates and the like, and one or more pharmaceutically acceptable carriers, diluents, or excipients. The compounds of formula (I) and salts, solvates, etc, are as described above. The carrier(s), diluent(s) or excipient(s) must be acceptable in the sense of being compatible with the other ingredients of the formulation and not deleterious to the recipient thereof. In accordance with another aspect of the invention there is also provided a process for the preparation of a pharmaceutical formulation including admixing a compound of the formula (I), or salts, solvates etc, with one or more pharmaceutically acceptable carriers, diluents or excipients.
It will be appreciated by those skilled in the art that certain protected derivatives of compounds of formula (I), which may be made prior to a final deprotection stage, may not possess pharmacological activity as such, but may, in certain instances, be administered orally or parenterally and thereafter metabolised in the body to form compounds of the invention which are pharmacologically active. Such derivatives may therefore be described as "prodrugs". Further, certain compounds of the invention may act as prodrugs of other compounds of the invention. All protected derivatives and prodrugs of compounds of the invention are included within the scope of the invention. It will further be appreciated by those skilled in the art, that certain moieties, known to those skilled in the art as "pro-moieties" may be placed on appropriate functionalities when such functionalities are present within compounds of the invention. Preferred prodrugs for compounds of the invention include: esters, carbonate esters, hemi- esters, phosphate esters, nitro esters, sulfate esters, sulfoxides, amides, carbamates, azo-compounds, phosphamides, glycosides, ethers, acetals and ketals.
Treatments
The compounds and compositions of the invention are used to treat cellular proliferation diseases. Disease states which can be treated by the methods and compositions provided herein include, but are not limited to, cancer (further discussed below), autoimmune disease, fungal disorders, arthritis, graft rejection, inflammatory bowel disease, proliferation induced after medical procedures, including, but not limited to, surgery, angioplasty, and the like. It is appreciated that in some cases the cells may not be in a hyper or hypo proliferation state (abnormal state) and still requires treatment. For example, during wound healing, the cells may be proliferating "normally", but proliferation enhancement may be desired. Thus, in one embodiment, the invention herein includes application to cells or individuals afflicted or impending affliction with any one of these disorders or states.
The compositions and methods provided herein are particularly deemed useful for the treatment of cancer including tumors such as prostate, breast, brain, skin, cervical carcinomas, testicular carcinomas, etc. They are particularly useful in treating metastatic or malignant tumors. More particularly, cancers that may be treated by the compositions and methods of the invention include, but are not limited to tumor types such as astrocytic, breast, cervical, colorectal, endometrial, esophageal, gastric, head and neck, hepatocellular, laryngeal, lung, oral, ovarian, prostate and thyroid carcinomas and sarcomas. More specifically, these compounds can be used to treat: Cardiac: sarcoma (angiosarcoma, fibrosarcoma, rhabdomyosarcoma, liposarcoma), myxoma, rhabdomyoma, fibroma, lipoma and teratoma; Lung: bronchogenic carcinoma (squamous cell, undifferentiated small cell, undifferentiated large cell, adenocarcinoma), alveolar (bronchiolar) carcinoma, bronchial adenoma, sarcoma, lymphoma, chondromatous hamartoma, mesothelioma; Gastrointestinal: esophagus (squamous cell carcinoma, adenocarcinoma, leiomyosarcoma, lymphoma), stomach (carcinoma, lymphoma, leiomyosarcoma), pancreas (ductal adenocarcinoma, insulinoma, glucagonoma, gastrinoma, carcinoid tumors, vipoma), small bowel (adenocarcinoma, lymphoma, carcinoid tumors, Karposi's sarcoma, leiomyoma, hemangioma, lipoma, neurofibroma, fibroma), large bowel
(adenocarcinoma, tubular adenoma, villous adenoma, hamartoma, leiomyoma); Genitourinary tract: kidney (adenocarcinoma, Wilm's tumor (nephroblastoma), lymphoma, leukemia), bladder and urethra (squamous cell carcinoma, transitional cell carcinoma, adenocarcinoma), prostate (adenocarcinoma, sarcoma), testis (seminoma, teratoma, embryonal carcinoma, teratocarcinoma, choriocarcinoma, sarcoma, interstitial cell carcinoma, fibroma, fibroadenoma, adenomatoid tumors, lipoma); Liver: hepatoma (hepatocellular carcinoma), cholangiocarcinoma, hepatoblastoma, angiosarcoma, hepatocellular adenoma, hemangioma; Biliary tract: gall bladder carcinoma, ampullary carcinoma, cholangiocarcinoma; Bone: osteogenic sarcoma (osteosarcoma), fibrosarcoma, malignant fibrous histiocytoma, chondrosarcoma, Ewing's sarcoma, malignant lymphoma (reticulum cell sarcoma), multiple myeloma, malignant giant cell tumor chordoma, osteochronfroma (osteocartilaginous exostoses), benign chondroma, chondroblastoma, chondromyxofibroma, osteoid osteoma and giant cell tumors; Nervous system: skull (osteoma, hemangioma, granuloma, xanthoma, osteitis deformans), meninges (meningioma, meningiosarcoma, gliomatosis), brain (astrocytoma, medulloblastoma, glioma, ependymoma, germinoma (pinealoma), glioblastoma multiform, oligodendroglioma, schwannoma, retinoblastoma, congenital tumors), spinal cord neurofibroma, meningioma, glioma, sarcoma); Gynecological: uterus (endometrial carcinoma), cervix (cervical carcinoma, pre-tumor cervical dysplasia), ovaries (ovarian carcinoma (serous cystadenocarcinoma, mucinous cystadenocarcinoma, unclassified carcinoma), granulosa-thecal cell tumors, Sertoli-Leydig cell tumors, dysgerminoma, malignant teratoma), vulva (squamous cell carcinoma, intraepithelial carcinoma, adenocarcinoma, fibrosarcoma, melanoma), vagina (clear cell carcinoma, squamous cell carcinoma, botryoid sarcoma (embryonal rhabdomyosarcoma), fallopian tubes (carcinoma);
Hematologic: blood (myeloid leukemia (acute and chronic), acute lymphoblastic leukemia, chronic lymphocytic leukemia, myeloproliferative diseases, multiple myeloma, myelodysplastic syndrome), Hodgkin's disease, non-Hodgkin's lymphoma (malignant lymphoma); Skin: malignant melanoma, basal cell carcinoma, squamous cell carcinoma, Karposi's sarcoma, moles dysplastic nevi, lipoma, angioma, dermatofibroma, keloids, psoriasis; and Adrenal glands: neuroblastoma. Thus, the term "cancerous cell" as provided herein, includes a cell afflicted by any one or related of the above identified conditions.
The instant compounds can be combined with or co-administered with other therapeutic agents, particularly agents that may enhance the activity or time of disposition of the compounds. Combination therapies according to the invention comprise the administration of at least one compound of the invention and the use of at least one other treatment method, fn one embodiment, combination therapies according to the invention comprise the administration of at least one compound of the invention and surgical therapy. In one embodiment, combination therapies according to the invention comprise the administration of at least one compound of the invention and radiotherapy. In one embodiment, combination therapies according to the invention comprise the administration of at least one compound of the invention and at least one supportive care agent (e.g., at least one anti-emetic agent). In one embodiment, combination therapies according to the present invention comprise the administration of at least one compound of the invention and at least one other chemotherapeutic agent. In one particular embodiment, the invention comprises the administration of at least one compound of the invention and at least one anti-neoplastic agent. In yet another embodiment, the invention comprises a therapeutic regimen where the EZH2 inhibitors of this disclosure are not in and of themselves active or significantly active, but when combined with another therapy, which may or may not be active as a standalone therapy, the combination provides a useful therapeutic outcome.
By the term "co-administering" and derivatives thereof as used herein is meant either simultaneous administration or any manner of separate sequential administration of an EZH2 inhibiting compound, as described herein, and a further active ingredient or ingredients, known to be useful in the treatment of cancer, including chemotherapy and radiation treatment. The term further active ingredient or ingredients, as used herein, includes any compound or therapeutic agent known to or that demonstrates advantageous properties when administered to a patient in need of treatment for cancer. Preferably, if the administration is not simultaneous, the compounds are administered in a close time proximity to each other. Furthermore, it does not matter if the compounds are administered in the same dosage form, e.g. one compound may be administered topically and another compound may be administered orally.
Typically, any anti-neoplastic agent that has activity versus a susceptible tumor being treated may be co-administered in the treatment of specified cancers in the present invention. Examples of such agents can be found in Cancer Principles and Practice of Oncology by V.T. Devita and S.
Hellman (editors), 6th edition (February 15, 2001), Lippincott Williams & Wilkins Publishers. A person of ordinary skill in the art would be able to discern which combinations of agents would be useful based on the particular characteristics of the drugs and the cancer involved. Typical anti- neoplastic agents useful in the present invention include, but are not limited to, anti-microtubule agents such as diterpenoids and vinca alkaloids; platinum coordination complexes; alkylating agents such as nitrogen mustards, oxazaphosphorines, alkylsulfonates, nitrosoureas, and triazenes; antibiotic agents such as anthracyclins, actinomycins and bleomycins; topoisomerase II inhibitors such as epipodophyllotoxins; antimetabolites such as purine and pyrimidine analogues and anti-folate compounds; topoisomerase I inhibitors such as camptothecins; hormones and hormonal analogues; DNA methyltransferase inhibitors such as azacitidine and decitabine; signal transduction pathway inhibitors; non-receptor tyrosine kinase angiogenesis inhibitors; immunotherapeutic agents;
proapoptotic agents; and cell cycle signaling inhibitors.
Typically, any chemotherapeutic agent that has activity against a susceptible neoplasm being treated may be utilized in combination with the compounds the invention, provided that the particular agent is clinically compatible with therapy employing a compound of the invention. Typical anti- neoplastic agents useful in the present invention include, but are not limited to: alkylating agents, antimetabolites, antitumor antibiotics, antimitotic agents, nucleoside analogues, topoisomerase I and II inhibitors, hormones and hormonal analogues; retinoids, histone deacetylase inhibitors; signal transduction pathway inhibitors including inhibitors of cell growth or growth factor function, angiogenesis inhibitors, and serine/threonine or other kinase inhibitors; cyclin dependent kinase inhibitors; antisense therapies and immunotherapeutic agents, including monoclonals, vaccines or other biological agents.
Nucleoside analogues are those compounds which are converted to deoxynucleotide triphosphates and incorporated into replicating DNA in place of cytosine. DNA methyltransferases become covalently bound to the modified bases resulting in an inactive enzyme and reduced DNA methylation. Examples of nucleoside analogues include azacitidine and decitabine which are used for the treatment of myelodysplastic disorder.Histone deacetylase (HDAC) inhibitors include vorinostat, for the treatment of cutaneous T-cell lymphoma. HDACs modify chromatin through the deactylation of histones. In addition, they have a variety of substrates including numerous transcription factors and signaling molecules. Other HDAC inhibitors are in development.
Signal transduction pathway inhibitors are those inhibitors which block or inhibit a chemical process which evokes an intracellular change. As used herein this change is cell proliferation or differentiation or survival. Signal transduction pathway inhibitors useful in the present invention include, but are not limited to, inhibitors of receptor tyrosine kinases, non-receptor tyrosine kinases, SH2/SH3 domain blockers, serine/threonine kinases, phosphatidyl inositol-3-OH kinases, myoinositol signaling, and Ras oncogenes. Signal transduction pathway inhibitors may be employed in combination with the compounds of the invention in the compositions and methods described above.
Receptor kinase angiogenesis inhibitors may also find use in the present invention. Inhibitors of angiogenesis related to VEGFR and TIE-2 are discussed above in regard to signal transduction inhibitors (both are receptor tyrosine kinases). Other inhibitors may be used in combination with the compounds of the invention. For example, anti-VEGF antibodies, which do not recognize VEGFR (the receptor tyrosine kinase), but bind to the ligand; small molecule inhibitors of integrin (alphav beta3) that inhibit angiogenesis; endostatin and angiostatin (non-RTK) may also prove useful in combination with the compounds of the invention. One example of a VEGFR antibody is bevacizumab (AVASTIN®).
Several inhibitors of growth factor receptors are under development and include ligand antagonists, antibodies, tyrosine kinase inhibitors, anti-sense oligonucleotides and aptamers. Any of these growth factor receptor inhibitors may be employed in combination with the compounds of the invention in any of the compositions and methods/uses described herein. Trastuzumab (Herceptin®) is an example of an anti-erbB2 antibody inhibitor of growth factor function. One example of an anti- erbB 1 antibody inhibitor of growth factor function is cetuximab (Erbitux™, C225). Bevacizumab (Avastin ) is an example of a monoclonal antibody directed against VEGFR. Examples of small molecule inhibitors of epidermal growth factor receptors include but are not limited to lapatinib (Tykerb™) and erlotinib (TARCEVA®). Imatinib mesylate (GLEEVEC®) is one example of a PDGFR inhibitor. Examples of VEGFR inhibitors include pazopanib, ZD6474, AZD2171, PT 787, sunitinib and sorafenib.
Anti-microtubule or anti-mitotic agents are phase specific agents active against the microtubules of tumor cells during M or the mitosis phase of the cell cycle. Examples of anti- microtubule agents include, but are not limited to, diterpenoids and vinca alkaloids.
Diterpenoids, which are derived from natural sources, are phase specific anti -cancer agents that operate at the G2/M phases of the cell cycle. It is believed that the diterpenoids stabilize the β- tubulin subunit of the microtubules, by binding with this protein. Disassembly of the protein appears then to be inhibited with mitosis being arrested and cell death following. Examples of diterpenoids include, but are not limited to, paclitaxel and its analog docetaxel.
Paclitaxel, 5P,20-epoxy-l,2a,4,7P,10p,13a-hexa-hydroxytax-l l-en-9-one 4,10-diacetate 2- benzoate 13-ester with (2R,3S)-N-benzoyl-3-phenylisoserine; is a natural diterpene product isolated from the Pacific yew tree Taxus brevifolia and is commercially available as an injectable solution TAXOL®. It is a member of the taxane family of terpenes. It was first isolated in 1971 by Wani et al. J. Am. Chem, Soc, 93:2325. 1971), who characterized its structure by chemical and X-ray crystallographic methods. One mechanism for its activity relates to paclitaxel's capacity to bind tubulin, thereby inhibiting cancer cell growth. Schiff et al., Proc. Natl, Acad, Sci. USA, 77: 1561- 1565 (1980); Schiff et al., Nature, 277:665-667 (1979); Kumar, J. Biol, Chem, 256: 10435-10441 (1981). For a review of synthesis and anticancer activity of some paclitaxel derivatives see: D. G. I. Kingston et al., Studies in Organic Chemistry vol. 26, entitled "New trends in Natural Products Chemistry 1986", Attaur- Rahman, P.W. Le Quesne, Eds. (Elsevier, Amsterdam, 1986) pp 219-235.
Paclitaxel has been approved for clinical use in the treatment of refractory ovarian cancer in the United States (Markman et al., Yale Journal of Biology and Medicine, 64:583, 1991 ; McGuire et al., Ann. Intern, Med., I l l :273,1989) and for the treatment of breast cancer (Holmes et al., J. Nat. Cancer Inst., 83 : 1797, 1991.) It is a potential candidate for treatment of neoplasms in the skin (Einzig et. al., Proc. Am. Soc. Clin. Oncol., 20:46) and head and neck carcinomas (Forastire et. al., Sem. Oncol., 20:56, 1990). The compound also shows potential for the treatment of polycystic kidney disease (Woo et. al., Nature, 368:750. 1994), lung cancer and malaria. Treatment of patients with paclitaxel results in bone marrow suppression (multiple cell lineages, Ignoff, R.J. et. al, Cancer Chemotherapy Pocket Guide,, 1998) related to the duration of dosing above a threshold concentration (50nM) (Kearns, CM. et. al., Seminars in Oncology, 3(6) p.16-23, 1995). Docetaxel, (2R,3S)- N-carboxy-3-phenylisoserine,N-ter?-buryl ester, 13-ester with 5β-20- epoxy- l,2a,4,7P, 10P,13a-hexahydroxytax-l l-en-9-one 4-acetate 2-benzoate, trihydrate; is commercially available as an injectable solution as TAXOTERE®. Docetaxel is indicated for the treatment of breast cancer. Docetaxel is a semisynthetic derivative of paclitaxel q.v., prepared using a natural precursor, 10-deacetyl-baccatin III, extracted from the needle of the European Yew tree. The dose limiting toxicity of docetaxel is neutropenia.
Vinca alkaloids are phase specific anti-neoplastic agents derived from the periwinkle plant. Vinca alkaloids act at the M phase (mitosis) of the cell cycle by binding specifically to tubulin. Consequently, the bound tubulin molecule is unable to polymerize into microtubules. Mitosis is believed to be arrested in metaphase with cell death following. Examples of vinca alkaloids include, but are not limited to, vinblastine, vincristine, and vinorelbine.
Vinblastine, vincaleukoblastine sulfate, is commercially available as VELBAN® as an injectable solution. Although, it has possible indication as a second line therapy of various solid tumors, it is primarily indicated in the treatment of testicular cancer and various lymphomas including Hodgkin's Disease; and lymphocytic and histiocytic lymphomas. Myelosuppression is the dose limiting side effect of vinblastine.
Vincristine, vincaleukoblastine, 22-oxo-, sulfate, is commercially available as ONCOVIN® as an injectable solution. Vincristine is indicated for the treatment of acute leukemias and has also found use in treatment regimens for Hodgkin's and non-Hodgkin's malignant lymphomas. Alopecia and neurologic effects are the most common side effect of vincristine and to a lesser extent
myelosupression and gastrointestinal mucositis effects occur.
Vinorelbine, 3',4'-didehydro -4'-deoxy-C'-norvincaleukoblastine [R-(R*,R*)-2,3- dihydroxybutanedioate (1 :2)(salt)], commercially available as an injectable solution of vinorelbine tartrate (NAVELBINE®), is a semisynthetic vinca alkaloid. Vinorelbine is indicated as a single agent or in combination with other chemotherapeutic agents, such as cisplatin, in the treatment of various solid tumors, particularly non-small cell lung, advanced breast, and hormone refractory prostate cancers. Myelosuppression is the most common dose limiting side effect of vinorelbine.
Platinum coordination complexes are non-phase specific anti-cancer agents, which are interactive with DNA. The platinum complexes enter tumor cells, undergo, aquation and form intra- and interstrand crosslinks with DNA causing adverse biological effects to the tumor. Examples of platinum coordination complexes include, but are not limited to, cisplatin and carboplatin.
Cisplatin, cis-diamminedichloroplatinum, is commercially available as PLATINOL® as an injectable solution. Cisplatin is primarily indicated in the treatment of metastatic testicular and ovarian cancer and advanced bladder cancer. The primary dose limiting side effects of cisplatin are nephrotoxicity, which may be controlled by hydration and diuresis, and ototoxicity. Carboplatin, platinum, diammine [l,l-cyclobutane-dicarboxylate(2-)-0,0'], is commercially available as PARAPLATIN® as an injectable solution. Carboplatin is primarily indicated in the first and second line treatment of advanced ovarian carcinoma. Bone marrow suppression is the dose limiting toxicity of carboplatin.
Alkylating agents are non-phase anti-cancer specific agents and strong electrophiles.
Typically, alkylating agents form covalent linkages, by alkylation, to DNA through nucleophilic moieties of the DNA molecule such as phosphate, amino, sulfhydryl, hydroxyl, carboxyl, and imidazole groups. Such alkylation disrupts nucleic acid function leading to cell death. Examples of alkylating agents include, but are not limited to, nitrogen mustards such as cyclophosphamide, melphalan, and chlorambucil; alkyl sulfonates such as busulfan; nitrosoureas such as carmustine; and triazenes such as dacarbazine.
Cyclophosphamide, 2-[bis(2-chloroethyl)amino]tetrahydro-2H-l,3,2-oxazaphosphorine 2- oxide monohydrate, is commercially available as an injectable solution or tablets as CYTOXAN®. Cyclophosphamide is indicated as a single agent or in combination with other chemotherapeutic agents, in the treatment of malignant lymphomas, multiple myeloma, and leukemias. Alopecia, nausea, vomiting and leukopenia are the most common dose limiting side effects of
cyclophosphamide.
Melphalan, 4-[bis(2-chloroethyl)amino]-L-phenylalanine, is commercially available as an injectable solution or tablets as ALKERAN®. Melphalan is indicated for the palliative treatment of multiple myeloma and non-resectable epithelial carcinoma of the ovary. Bone marrow suppression is the most common dose limiting side effect of melphalan.
Chlorambucil, 4-[bis(2-chloroethyl)amino]benzenebutanoic acid, is commercially available as LEUKERAN® tablets. Chlorambucil is indicated for the palliative treatment of chronic lymphatic leukemia, and malignant lymphomas such as lymphosarcoma, giant follicular lymphoma, and Hodgkin's disease. Bone marrow suppression is the most common dose limiting side effect of chlorambucil.
Busulfan, 1,4-butanediol dimethanesulfonate, is commercially available as MYLERAN® TABLETS. Busulfan is indicated for the palliative treatment of chronic myelogenous leukemia. Bone marrow suppression is the most common dose limiting side effects of busulfan.
Carmustine, l,3-[bis(2-chloroethyl)-l -nitrosourea, is commercially available as single vials of lyophilized material as BiCNU®. Carmustine is indicated for the palliative treatment as a single agent or in combination with other agents for brain tumors, multiple myeloma, Hodgkin's disease, and non- Hodgkin's lymphomas. Delayed myelosuppression is the most common dose limiting side effects of carmustine. Dacarbazine, 5-(3,3-dimethyl-l-triazeno)-imidazole-4-carboxamide, is commercially available as single vials of material as DTIC-Dome®. Dacarbazine is indicated for the treatment of metastatic malignant melanoma and in combination with other agents for the second line treatment of Hodgkin's Disease. Nausea, vomiting, and anorexia are the most common dose limiting side effects of dacarbazine.
Antibiotic anti-neoplastics are non-phase specific agents, which bind or intercalate with DNA. Typically, such action results in stable DNA complexes or strand breakage, which disrupts ordinary function of the nucleic acids leading to cell death. Examples of antibiotic anti-neoplastic agents include, but are not limited to, actinomycins such as dactinomycin, anthrocyclins such as
daunorubicin and doxorubicin; and bleomycins.
Dactinomycin, also know as Actinomycin D, is commercially available in injectable form as COSMEGEN®. Dactinomycin is indicated for the treatment of Wilm's tumor and
rhabdomyosarcoma. Nausea, vomiting, and anorexia are the most common dose limiting side effects of dactinomycin.
Daunorubicin, (8S-cis-)-8-acetyl- 10-[(3-amino-2,3,6-trideoxy-a-L-lyxo-hexopyranosyl)oxy]-
7,8,9, 10-tetrahydro-6,8, 1 l-trihydroxy-l-methoxy-5, 12 naphthacenedione hydrochloride, is commercially available as a liposomal injectable form as DAUNOXOME® or as an injectable as CERUBIDINE®. Daunorubicin is indicated for remission induction in the treatment of acute nonlymphocytic leukemia and advanced HIV associated Kaposi's sarcoma. Myelosuppression is the most common dose limiting side effect of daunorubicin.
Doxorubicin, (8S, 10S)-10-[(3-amino-2,3,6-trideoxy-a-L-lyxo-hexopyranosyl)oxy]-8- glycoloyl, 7,8,9, 10-tetrahydro-6,8,l l-trihydroxy- l-methoxy-5, 12 naphthacenedione hydrochloride, is commercially available as an injectable form as RUBEX® or ADRIAMYCIN RDF®. Doxorubicin is primarily indicated for the treatment of acute lymphoblastic leukemia and acute myeloblastic leukemia, but is also a useful component in the treatment of some solid tumors and lymphomas. Myelosuppression is the most common dose limiting side effect of doxorubicin.
Bleomycin, a mixture of cytotoxic glycopeptide antibiotics isolated from a strain of
Streptomyces verticillus, is commercially available as BLENOXANE®. Bleomycin is indicated as a palliative treatment, as a single agent or in combination with other agents, of squamous cell carcinoma, lymphomas, and testicular carcinomas. Pulmonary and cutaneous toxicities are the most common dose limiting side effects of bleomycin.
Topoisomerase II inhibitors include, but are not limited to, epipodophyllotoxins.
Epipodophyllotoxins are phase specific anti-neoplastic agents derived from the mandrake plant. Epipodophyllotoxins typically affect cells in the S and G2 phases of the cell cycle by forming a ternary complex with topoisomerase II and DNA causing DNA strand breaks. The strand breaks accumulate and cell death follows. Examples of epipodophyllotoxins include, but are not limited to, etoposide and teniposide.
Etoposide, 4'-demethyl-epipodophyllotoxin 9[4,6-0-(R )-ethylidene-P-D-glucopyranoside], is commercially available as an injectable solution or capsules as VePESID® and is commonly known as VP- 16. Etoposide is indicated as a single agent or in combination with other chemotherapy agents in the treatment of testicular and non-small cell lung cancers. Myelosuppression is the most common side effect of etoposide. The incidence of leucopenia tends to be more severe than thrombocytopenia.
Teniposide, 4'-demethyl-epipodophyllotoxin 9[4,6-0-(R )-thenylidene- -D-glucopyranoside], is commercially available as an injectable solution as VUMON® and is commonly known as VM-26. Teniposide is indicated as a single agent or in combination with other chemotherapy agents in the treatment of acute leukemia in children. Myelosuppression is the most common dose limiting side effect of teniposide. Teniposide can induce both leucopenia and thrombocytopenia.
Antimetabolite neoplastic agents are phase specific anti-neoplastic agents that act at S phase (DNA synthesis) of the cell cycle by inhibiting DNA synthesis or by inhibiting purine or pyrimidine base synthesis and thereby limiting DNA synthesis. Consequently, S phase does not proceed and cell death follows. Examples of antimetabolite anti-neoplastic agents include, but are not limited to, fluorouracil, methotrexate, cytarabine, mecaptopurine, thioguanine, and gemcitabine.
5-fluorouracil, 5-fluoro-2,4- (1H,3H) pyrimidinedione, is commercially available as fluorouracil. Administration of 5-fluorouracil leads to inhibition of thymidylate synthesis and is also incorporated into both RNA and DNA. The result typically is cell death. 5-fluorouracil is indicated as a single agent or in combination with other chemotherapy agents in the treatment of carcinomas of the breast, colon, rectum, stomach and pancreas. Myelosuppression and mucositis are dose limiting side effects of 5-fluorouracil. Other fluoropyrimidine analogs include 5-fluoro deoxyuridine (floxuridine) and 5-fluorodeoxyuridine monophosphate.
Cytarabine, 4-amino- l-P-D-arabinofuranosyl-2 (lH)-pyrimidinone, is commercially available as CYTOSAR-U® and is commonly known as Ara-C. It is believed that cytarabine exhibits cell phase specificity at S-phase by inhibiting DNA chain elongation by terminal incorporation of cytarabine into the growing DNA chain. Cytarabine is indicated as a single agent or in combination with other chemotherapy agents in the treatment of acute leukemia. Other cytidine analogs include 5-azacytidine and 2',2'-difluorodeoxycytidine (gemcitabine). Cytarabine induces leucopenia, thrombocytopenia, and mucositis.
Mercaptopurine, 1 ,7-dihydro-6H-purine-6-thione monohydrate, is commercially available as PURTNETHOL®. Mercaptopurine exhibits cell phase specificity at S-phase by inhibiting DNA synthesis by an as of yet unspecified mechanism. Mercaptopurine is indicated as a single agent or in combination with other chemotherapy agents in the treatment of acute leukemia. Myelosuppression and gastrointestinal mucositis are expected side effects of mercaptopurine at high doses. A useful mercaptopurine analog is azathioprine.
Thioguanine, 2-amino-l,7-dihydro-6H-purine-6-thione, is commercially available as TABLOID®. Thio guanine exhibits cell phase specificity at S-phase by inhibiting DNA synthesis by an as of yet unspecified mechanism. Thioguanine is indicated as a single agent or in combination with other chemotherapy agents in the treatment of acute leukemia. Myelosuppression, including leucopenia, thrombocytopenia, and anemia, is the most common dose limiting side effect of thioguanine administration. However, gastrointestinal side effects occur and can be dose limiting. Other purine analogs include pentostatin, erythrohydroxynonyladenine, fludarabine phosphate, and cladribine.
Gemcitabine, 2'-deoxy-2', 2'-difluorocytidine monohydrochloride (β-isomer), is commercially available as GEMZAR®. Gemcitabine exhibits cell phase specificity at S-phase and by blocking progression of cells through the Gl/S boundary. Gemcitabine is indicated in combination with cisp latin in the treatment of locally advanced non-small cell lung cancer and alone in the treatment of locally advanced pancreatic cancer. Myelosuppression, including leucopenia, thrombocytopenia, and anemia, is the most common dose limiting side effect of gemcitabine administration.
Methotrexate, N-[4[[(2,4-diamino-6-pteridinyl) methyl]methylamino] benzoyl]-L-glutamic acid, is commercially available as methotrexate sodium. Methotrexate exhibits cell phase effects specifically at S-phase by inhibiting DNA synthesis, repair and/or replication through the inhibition of dyhydrofolic acid reductase which is required for synthesis of purine nucleotides and thymidylate. Methotrexate is indicated as a single agent or in combination with other chemotherapy agents in the treatment of choriocarcinoma, meningeal leukemia, non-Hodgkin's lymphoma, and carcinomas of the breast, head, neck, ovary and bladder. Myelosuppression (leucopenia, thrombocytopenia, and anemia) and mucositis are expected side effect of methotrexate administration.
Camptothecins, including, camptothecin and camptothecin derivatives are available or under development as Topoisomerase I inhibitors. Camptothecins cytotoxic activity is believed to be related to its Topoisomerase I inhibitory activity. Examples of camptothecins include, but are not limited to irinotecan, topotecan, and the various optical forms of 7-(4-methylpiperazino-methylene)- 10, 1 1 - ethylenedioxy-20-camptothecin described below.
Irinotecan HC1, (4S)-4,1 l-diethyl-4-hydroxy-9-[(4-piperidinopiperidino) carbonyloxy]-lH- pyrano[3',4',6,7]indolizino[l,2-b]quinoline-3,14(4H,12H)-dione hydrochloride, is commercially available as the injectable solution CAMPTOSAR®.
Irinotecan is a derivative of camptothecin which binds, along with its active metabolite SN- 38, to the topoisomerase I - DNA complex. It is believed that cytotoxicity occurs as a result of irreparable double strand breaks caused by interaction of the topoisomerase I : DNA : irintecan or SN- 38 ternary complex with replication enzymes. Irinotecan is indicated for treatment of metastatic cancer of the colon or rectum. The dose limiting side effects of irinotecan HC1 are myelosuppression, including neutropenia, and GI effects, including diarrhea.
Topotecan HC1, (S)- 10- [(dimethylamino)methyl]-4-ethyl-4,9-dihydroxy- 1 H- pyrano[3',4',6,7]indolizino[l,2-b]quinoline-3, 14-(4H, 12H)-dione monohydrochloride, is commercially available as the injectable solution HYCAMTIN®. Topotecan is a derivative of camptothecin which binds to the topoisomerase I - DNA complex and prevents religation of singles strand breaks caused by Topoisomerase I in response to torsional strain of the DNA molecule.
Topotecan is indicated for second line treatment of metastatic carcinoma of the ovary and small cell lung cancer. The dose limiting side effect of topotecan HC1 is myelosuppression, primarily neutropenia.
Pharmaceutical compositions may be presented in unit dose forms containing a predetermined amount of active ingredient per unit dose. Such a unit may contain, for example, 0.5 mg to 1 g, preferably 1 mg to 700 mg, more preferably 5 mg to 100 mg of a compound of the formula (I), depending on the condition being treated, the route of administration and the age, weight and condition of the patient, or pharmaceutical compositions may be presented in unit dose forms containing a predetermined amount of active ingredient per unit dose. Preferred unit dosage compositions are those containing a daily dose or sub-dose, as herein above recited, or an appropriate fraction thereof, of an active ingredient. Furthermore, such pharmaceutical compositions may be prepared by any of the methods well known in the pharmacy art.
Pharmaceutical compositions may be adapted for administration by any appropriate route, for example by the oral (including buccal or sublingual), rectal, nasal, topical (including buccal, sublingual or transdermal), vaginal or parenteral (including subcutaneous, intramuscular, intravenous or intradermal) route. Such compositions may be prepared by any method known in the art of pharmacy, for example by bringing into association a compound of formal (I) with the carrier(s) or excipient(s).
Pharmaceutical compositions adapted for oral administration may be presented as discrete units such as capsules or tablets; powders or granules; solutions or suspensions in aqueous or nonaqueous liquids; edible foams or whips; or oil-in-water liquid emulsions or water-in-oil liquid emulsions.
Capsules are made by preparing a powder mixture, as described above, and filling formed gelatin sheaths. Glidants and lubricants such as colloidal silica, talc, magnesium stearate, calcium stearate or solid polyethylene glycol can be added to the powder mixture before the filling operation. A disintegrating or solubilizing agent such as agar-agar, calcium carbonate or sodium carbonate can also be added to improve the availability of the medicament when the capsule is ingested. Moreover, when desired or necessary, suitable binders, lubricants, disintegrating agents and coloring agents can also be incorporated into the mixture. Suitable binders include starch, gelatin, natural sugars such as glucose or beta-lactose, corn sweeteners, natural and synthetic gums such as acacia, tragacanth or sodium alginate, carboxymethylcellulose, polyethylene glycol, waxes and the like. Lubricants used in these dosage forms include sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride and the like. Disintegrators include, without limitation, starch, methyl cellulose, agar, bentonite, xanthan gum and the like. Tablets are formulated, for example, by preparing a powder mixture, granulating or slugging, adding a lubricant and disintegrant and pressing into tablets. A powder mixture is prepared by mixing the compound, suitably comminuted, with a diluent or base as described above, and optionally, with a binder such as carboxymethylcellulose, an aliginate, gelatin, or polyvinyl pyrrolidone, a solution retardant such as paraffin, a resorption accelerator such as a quaternary salt and/or an absorption agent such as bentonite, kaolin or dicalcium phosphate. The powder mixture can be granulated by tablet forming dies by means of the addition of stearic acid, a stearate salt, talc or mineral oil. The lubricated mixture is then compressed into tablets. The compounds of the present invention can also be combined with a free flowing inert carrier and compressed into tablets directly without going through the granulating or slugging steps. A clear or opaque protective coating consisting of a sealing coat of shellac, a coating of sugar or polymeric material and a polish coating of wax can be provided. Dyestuffs can be added to these coatings to distinguish different unit dosages.
Oral fluids such as solution, syrups and elixirs can be prepared in dosage unit form so that a given quantity contains a predetermined amount of a compound of formula (I). Syrups can be prepared by dissolving the compound in a suitably flavored aqueous solution, while elixirs are prepared through the use of a non-toxic alcoholic vehicle. Suspensions can be formulated by dispersing the compound in a non-toxic vehicle. Solubilizers and emulsifiers such as ethoxylated isostearyl alcohols and polyoxy ethylene sorbitol ethers, preservatives, flavor additive such as peppermint oil or natural sweeteners or saccharin or other artificial sweeteners, and the like can also be added.
Where appropriate, dosage unit pharmaceutical compositions for oral administration can be microencapsulated. The formulation can also be prepared to prolong or sustain the release as for example by coating or embedding particulate material in polymers, wax or the like.
Pharmaceutical compositions adapted for rectal administration may be presented as suppositories or as enemas.
Pharmaceutical compositions adapted for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams or spray formulations.
Pharmaceutical formulations adapted for parenteral administration include aqueous and nonaqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the composition isotonic with the blood of the intended recipient; and aqueous and nonaqueous sterile suspensions which may include suspending agents and thickening agents. The pharmaceutical compositions may be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets.
It should be understood that in addition to the ingredients particularly mentioned above, the pharmaceutical compositions may include other agents conventional in the art having regard to the type of formulation in question, for example those suitable for oral administration may include flavouring agents.
A therapeutically effective amount of a compound of the present invention will depend upon a number of factors including, for example, the age and weight of the intended recipient, the precise condition requiring treatment and its severity, the nature of the formulation, and the route of administration, and will ultimately be at the discretion of the attendant prescribing the medication.
However, an effective amount of a compound of formula (I) for the treatment of anemia will generally be in the range of O.001 to 100 mg/kg body weight of recipient per day, suitably in the range of .01 to 10 mg/kg body weight per day. For a 70kg adult mammal, the actual amount per day would suitably be from 7 to 700 mg and this amount may be given in a single dose per day or in a number (such as two, three, four, five or six) of sub-doses per day such that the total daily dose is the same. An effective amount of a salt or solvate, etc., may be determined as a proportion of the effective amount of the compound of formula (I) per se. It is envisaged that similar dosages would be appropriate for treatment of the other conditions referred to above. Chemical Background
The compounds of this invention may be made by a variety of methods, including standard chemistry. Any previously defined variable will continue to have the previously defined meaning unless otherwise indicated. Illustrative general synthetic methods are set out below and then specific compounds of the invention as prepared are given in the examples.
Compounds of general formula (I) may be prepared by methods known in the art of organic synthesis as set forth in part by the following synthesis schemes. In all of the schemes described below, it is well understood that protecting groups for sensitive or reactive groups are employed where necessary in accordance with general principles of chemistry. Protecting groups are manipulated according to standard methods of organic synthesis (T. W. Green and P. G. M. Wuts (19911 Protecting Groups in Organic Synthesis, John Wiley & Sons). These groups are removed at a convenient stage of the compound synthesis using methods that are readily apparent to those skilled in the art. The selection of processes as well as the reaction conditions and order of their execution shall be consistent with the preparation of compounds of formula (I). Those skilled in the art will recognize if a stereocenter exists in compounds of formula (I). Accordingly, the present invention includes both possible stereoisomers and includes not only racemic compounds but the individual enantiomers as well. When a compound is desired as a single enantiomer, it may be obtained by stereospecific synthesis or by resolution of the final product or any convenient intermediate.
Resolution of the final product, an intermediate, or a starting material may be effected by any suitable method known in the art. See, for example, Stereochemistry of Organic Compounds by E. L. Eliel, S. H. Wilen, and L. N. Mander (Wiley- Interscience, 1994).
EXAMPLES
The compounds of the present invention were prepared as depicted in schemes 1-5. The groups and substituents shown in the schemes 1-5, such as X, Y, Z and the various R groups have the same definition in what follows as they have herein above. The solvents and conditions referred to are illustrative and are not intended to be limiting.
Scheme 1
Figure imgf000033_0001
Scheme 1 illustrates a route to prepare compounds of formula (VI). Compounds of formula (I) are commercially available and were alkylated using sodium hydride, DMF, and an appropriate alkyl halide to afford a mixture of N1 N2 alkylated regioisomers which were separated by chromatography and fully characterized. Compounds of formula (II) were converted to compounds of formula (III) by standard base-catalyzed hydrolysis. Treatment of compounds of formula (III) with substituted aminomethyl pyridones of formula (IV) using EDC, HOAT, N-methylmorpholine, and DMSO at room temperature for a period of 3-48 h stirring afforded compounds of formula (V). Compounds of formula (V) were substituted at the 6-position using standard methods known to those skilled in the art (i.e. palladium, copper, mediated cross couplings), to afford compounds of formula (VI).
Sc
Figure imgf000033_0002
Compounds of formula (VI) were also prepared as depicted in scheme 2. In this embodiment, compounds of formula (II) were substituted at the 6-pos. using standard methods known to those skilled in the art (i.e. palladium, copper, mediated cross couplings, nucleophillic substitution) to afford compounds of formula (VII), which were then converted to compounds of formula (VIII) by standard base-catalyzed hydrolysis. Treatment of compounds of formula (VIII) with substituted aminomethyl pyridones of formula (IV) using EDC, HOAT, N-methylmorpholine, and DMSO at room temperature for a period of 3-48 h stirring, afforded compounds of formula (VI). Compounds of formula (VI) wherein 6 = sulfonamide were prepared from compounds of formula (II) in which the 6-nitro substituted conjeger was used as starting material. Conversion of the nrtro group by standard reduction conditions afforded a compound of formula (VII) in which R6 = NH2. Treatment of the putative amine intermediate with a sulfonyl chloride under standard conditions afforded compound of formula (VII), wherein R6 = sulfonamide.
Figure imgf000034_0001
Scheme 3 illustrates a route to prepare compounds of formula (XV). A compound of formula (IX), which is commercially available, was converted to a compound of formula (X) using sodium nitrite, 6N HC1, water, and stirring at room temperature for 48 h. Treatment of (X) with an alkyl halide under basic conditions afforded a compound of formula (XI). Compounds of formula (XI) were converted to compounds of formula (XII) typically by Wolff-Kischner mediated reduction conditions using sodium cyanoborohydride as the reducing agent. Compounds of formula (XII) were converted to compounds of formula (XIII) by standard base-catalyzed hydrolysis. Treatment of compounds of formula (XIII) with substituted aminomethyl pyridones of formula (IV) using EDC, HOAT, N- methylmorpholine, and DMSO at room temperature for a period of 3-48 h stirring afforded compounds of formula (XIV). Compounds of formula (XIV) were substituted at the 6-position using standard methods known to those skilled in the art (i.e. palladium, copper, mediated cross couplings), to afford compounds of formula (XV). Scheme 4
Figure imgf000035_0001
Compounds of formula (XV) were also prepared as depicted in scheme 4. In this embodiment, compounds of formula (XII) were substituted at the 6-pos. using standard methods known to those skilled in the art (i.e. palladium, copper, mediated cross couplings, nucleophillic substitution) to afford compounds of formula (XVI), which were then converted to compounds of formula (XVII) by standard base-catalyzed hydrolysis. Treatment of compounds of formula (XVII) with substituted aminomethyl pyridones of formula (IV) using EDC, HOAT, N-methylmorpholine, and DMSO at room temperature for a period of 3-48 h stirring afforded compounds of formula (XV).
Scheme 5
Figure imgf000035_0002
Scheme 5 illustrates the method to synthesize a compound of formula (IV). Heating of compounds of formula (XVIII) with cyanoacetamide in ethanol at reflux containing catalytic piperidine for typically 30 min, affords compounds of formula (XX). Alternatively, treatment of compounds of formula (XIX) with cyanoacetamide in DMSO at room temperature with an excess of potassium tert-butoxide under an atmosphere of oxygen for ca. 90 min. also affords compounds of formula (XX).
Regioisomeric mixtures, are separable and the individual compounds regiochemical assignments can be confirmed by 2D HNMR techniques. Compounds of formula (XVIII) and (XIX) which were not commercially available were prepared using standard methods known to those skilled in the art, and are described herein. Compounds of formula (XX) were converted to compounds of formula (V) either by hydrogenation using sodium acetate, palladium on carbon, and platinum oxide, or reduction conditions using NaBH4 with either iodine or N1CI2-6H2O. General Experimental Methods
The following abbreviations are used throughout the experimental and have the following meaning: aq aqueous
ΒΓΝΑΡ 2,2' -bis(diphenylphosphino) -l ,l '-binapthyl
ca. circa
CDCl3-(i chloroform-t/
CD3OD-£/4 methanol-^
CS2CO3 cesium carbonate
CHCI3 chloroform
ACN acetonitrile
CH3CN acetonitrile
Celite® registered trademark of Celite Corp. brand of diatomaceous earth
DBU l,8-diazabicyclo[5.4.0]undeca-7-ene
DCE dichloroethane
DCM methylene chloride
DME 1,2 dimethoxyethane
DMF N,N-dimethylformamide
DIEA diisopropyl ethylamine
DMSO-<f6 dimethylsulfoxide-i/6
EtOAc ethyl acetate
EDC l-(3-dimethylaminopropyl)-3-ethylcarbodimmide hydrochloride
h hour(s)
l~H NMR proton nuclear magnetic resonance
HC1 hydrochloric acid
HOAT 1 -hydro xy-7-azabenzotriazole
HPLC high performance liquid chromatography
IP A 2-propanol
K2C03 potassium carbonate
KOH potassium hydroxide
LC/MS liquid chromatography/mass spectroscopy
MgS04 magnesium sulfate
MeOH methanol
min minute(s)
MTBE methyl tert-butyl ether
MS mass spectrometry
NaOH sodium hydroxide Na2S04 sodium sulfate
NH4OH ammonium hydroxide
NMM 4-methylmorpholine
NMP N-Methyl-2-pyrrolidone
Pd/C Palladium ( 10% by wt) on carbon
PdCl2(d f)-CH2Cl2 1 , 1 '-Bis(diphenylphosphino)fenOcene-palladium(II)dichloride
dichloromethane complex
Pd(Ph3P)4 tetrakis(triphenylphosphine)palladium(0)
RT room temperature
SOCI2 thionyl chloride
SPhos 2-Dicyclohexylphosphino-2',6'-dimethoxybiphenyl
TFA trifluoroacetic acd
THF tetrahydrofuran
TLC thin layer chromatography
The following guidelines apply to all experimental procedures described herein. All reactions were conducted under a positive pressure of nitrogen using oven-dried glassware, unless otherwise indicated. Temperatures designated are external (i.e. bath temperatures), and are approximate. Air and moisture-sensitive liquids were transferred via syringe. Reagents were used as received. Solvents utilized were those listed as "anhydrous" by vendors. Molarities listed for reagents in solutions are approximate, and were used without prior titration against a corresponding standard. All reactions were agitated by stir bar, unless otherwise indicated. Heating was conducted using heating baths containing silicon oil, unless otherwise indicated. Reactions conducted by microwave irradiation (0 - 400 W at 2.45 GHz) were done so using a Biotage Initiator™ 2.0 instrument with Biotage microwave EXP vials (0.2 - 20 mL) and septa and caps. Irradiation levels utilized (i.e. high, normal, low) based on solvent and ionic charge were based on vendor specifications. Cooling to temperatures below -70 °C was conducted using dry ice/acetone or dry ice/2 -propanol. Magnesium sulfate and sodium sulfate used as drying agents were of anhydrous grade, and were used interchangeably. Solvents described as being removed "in vacuo " or "under reduced pressure" were done so by rotary evaporation.
Preparative normal phase silica gel chromatography was carried out using either a Teledyne ISCO CombiFlash Companion instrument with RediSep or ISCO Gold silica gel cartridges (4 g-330 g), or an Analogix IF280 instrument with SF25 silica gel cartridges (4 g - 3-00g), or a Biotage SP1 instrument with HP silica gel cartridges (lOg - 100 g). Purification by reverse phase HPLC was conducted using a YMC-pack column (ODS-A 75x30mm) as solid phase, unless otherwise noted. A mobile phase of 25mL/min A (acetonitrile-0.1%TFA) : B (water-0.1% TFA), 10-80% gradient A (10 min) was utilized with UV detection at 214 nM, unless otherwise noted.
A PE Sciex API 150 single quadrupole mass spectrometer (PE Sciex, Thornhill, Ontario, Canada) was operated using electrospray ionization in the positive ion detection mode. The nebulizing gas was generated from a zero air generator (Balston Inc., Haverhill, MA, USA) and delivered at 65 psi and the curtain gas was high purity nitrogen delivered from a Dewar liquid nitrogen vessel at 50 psi. The voltage applied to the electrospray needle was 4.8 kV. The orifice was set at 25 V and mass spectrometer was scanned at a rate of 0.5 scan/sec using a step mass of 0.2 amu and collecting profile data.
Method A LCMS. Samples were introduced into the mass spectrometer using a CTC PAL autosampler (LEAP Technologies, Carrboro, NC) equipped with a hamilton 10 uL syringe which performed the injection into a Valco 10-port injection valve. The HPLC pump was a Shimadzu LC- lOADvp (Shimadzu Scientific Instruments, Columbia, MD) operated at 0.3 mL/min and a linear gradient 4.5% A to 90% B in 3.2 min. with a 0.4 min. hold. The mobile phase was composed of 100% (H20 0.02% TFA) in vessel A and 100% (CH3CN 0.018% TFA) in vessel B. The stationary phase is Aquasil (C I 8) and the column dimensions were 1 mm x 40 mm. Detection was by UV at 214 nm, evaporative light- scattering (ELSD) and MS.
Method B, LCMS. Alternatively, an Agilent 1 100 analytical HPLC system with an LC/MS was used and operated at 1 mL/min and a linear gradient 5% A to 100% B in 2.2 min with a 0.4 min hold. The mobile phase was composed of 100% (H20 0.02% TFA) in vessel A and 100% (CH3CN 0.018% TFA) in vessel B. The stationary phase was Zobax (C8) with a 3.5 um partical size and the column dimensions were 2.1 mm x 50 mm. Detection was by UV at 214 nm, evaporative light- scattering (ELSD) and MS.
Method C, LCMS. Alternatively, an MDSSCIEX API 2000 equipped with a capillary column of (50 x 4.6 mm, 5 μτα) was used. HPLC was done on Agilent- 1200 series UPLC system equipped with column Zorbax SB-C 18 (50 x 4.6 mm, 1.8 /mi) eluting with CH3CN: ammonium acetate buffer. The reactions were performed in the microwave (CEM, Discover).
'H-NMR spectra were taken using deuterated DMSO (unless otherwise noted) and were recorded at 400 MHz using a Bruker AVANCE 400 MHz instrument, with ACD Spect manager v. 10 used for reprocessing. Multiplicities indicated are: s=singlet, d=doublet, t=triplet, q=quartet, quint= quintet, sxt= sextet, m=multiplet, dd = doublet of doublets, dt=doublet of triplets etc. and br indicates a broad signal.
Analytical HPLC: Products were analyzed by Agilent 1 100 Analytical Chromatography system, with 4.5 x 75 mm Zorbax XDB-C18 column (3.5 um) at 2 mL/min with a 4 min gradient from 5% CH3CN (0.1% formic acid) to 95% CH3CN (0.1% formic acid) in H20 (0.1% formic acid) and a 1 min hold. 1. Experimentals Example 1
6-chloro-l-(l-methylethy])-Ar-[(6-methyl-2-oxo-4-propyl-l,2-dihydro-3^yridinyl)methyl]-l /- indazole-4-carboxamide
Figure imgf000039_0001
a) Methyl 6-chloro-l-(l-methylethyl)-l/f-indazole-4-carboxylate
Figure imgf000039_0002
Methyl 6-chloro-lH-indazole-4-carboxylate (0.4 lOg, 1.947 mmol) was dissolved in DMF (10 mL) and placed into an ice bath and stirred for 15 min. Sodium hydride (0.101 g, 2.53 mmol) was added, the contents stirred for 15 min., and then 2-iodopropane (0.389 mL, 3.89 mmol) was added. The contents were stirred with warming to RT. After stirring at RT for 2h, a scoop of sodium carbonate and then 0.4 mL of methyl iodide were added. After stirring at RT for an additional lh, the reaction mixture was diluted with saturated NH4CI and then extracted with EtOAc (2x). The combined organic layers were washed with brine, dried over MgS04, filtered, and concentrated in vacuo. The crude residue was dissolved in a minimal amount of DCM and purified by silica gel chromatography (eluent: 3-20% ethyl acetate in hexanes). The title compound was the less polar of the two products separated from chromatography, and was collected as 0.22g (45% yield). ¾ NMR (400 MHz, DMSO-i/6) δ ppm 1.45 - 1.52 (m, 6 H), 3.93 - 3.99 (m, 3 H), 5.11 (quin, J=6.57 Hz, 1 H), 7.73 (d, J=1.77 Hz, 1 H), 8.30 (s, 1 H), 8.42 (s, 1 H). b) 6-chloro-l-(l-methylethyl)-l//-indazole-4-carboxylic acid
Figure imgf000040_0001
Methyl 6-chloro-l-(l-methylethyl)-lH-indazole-4-carboxylate (0.22g, 0.871 mmol) was dissolved in methanol (6 mL) and THF (1 mL), followed by addition of NaOH (1.451 mL, 4.35 mmol) via syringe. The contents were stirred at RT for 3h. The volatiles were removed in vacuo, the contents diluted with water, and then slowly acidifed to pH 4-5 by addition of 1M HC1 (solid precipitation occured). The contents were then extracted with EtOAc (3x). The combined organic layers were washed with brine, dried over MgSC , filtered, and concentrated in vacuo. The collected solid was dried under hi- vac for 2h, and collected as 0.19g (90%). :H NMR (400 MHz, DMSO-c 6) δ ppm 1.47 (d, .7=6.57 Hz, 6 H) 5.09 (quin, J=6.57 Hz, 1 H) 7.70 (d, J=1.52 Hz, 1 H) 8.25 (s, 1 H) 8.40 (s, 1 H), 13.54 (br. s., 1 H). c) 6-chloro-l-(l-methylethyl)-lH-indazole-4-carboxylic acid (.10 g, 0.419 mmol), 3-(aminomethyl)- 6-methyl-4-propyl-2(lH)-pyridinone (0.109 g, 0.503 mmol), 1 -hydroxy-7-azabenzotriazole (0.080 g, 0.587 mmol), and then EDC (0.112 g, 0.587 mmol) were added to DMSO (4.0 mL). N- methylmorpholine (0.184 mL, 1.676 mmol) was quickly added via syringe. The contents were stirred at RT overnight Additional HOAT and EDC were added, and the contents were stirred for another 6h. The contents were slowly poured onto 45 mL of ice-water wherein solid precipiatation occurred. The contents were stirred for 10 min., and then allowed to stand for another 10 min. The contents were filtered, the collected solid washed with ca. 5 mL of water. The solid was air dried for 15 min. then dried in vacuum oven at 45 °C for 24h. The title compound was collected as a white solid (0.160g, 93 % yield). :H NMR (400 MHz, DMSO- 6) δ ppm 0.89 (t, J=7.33 Hz, 3 H) 1.42 - 1.58 (m, 8 H) 2.14 (s, 3 H) 2.52 - 2.58 (m, 1 H) 4.36 (d, J=5.05 Hz, 2 H) 5.04 (quin, J=6.57 Hz, 1 H) 5.91 (s, 1 H) 7.59 (d, J=1.52 Hz, 1 H) 8.06 (s, 1 H) 8.38 (s, 1 H) 8.61 (t, J=4.80 Hz, 1 H) 11.54 (s, 1 H); MS(ES) [M+H]+ 400.9.
Example 2
V-[(4,6-dimethyl-2-oxo-l,2-dihydro-3-pyridinyl)methyl]-6-fluoro-l-(l-methylethyl)-l/?- indazole-4-carboxamide
Figure imgf000041_0001
a) Methyl 6-fluoro-l-(l-methylethyl)-l/ -indazole-4-carboxylate
Figure imgf000041_0002
The title compound was prepared in the same manner as described for example 1 (step a) from methyl
6-fluoro- lH-indazole-4-carboxylate (.500g, 2.58 mmol), sodium hydride (0.129 g, 3.22 mmol), and 2- iodopropane (0.386 mL, 3.86 mmol). The product was collected as a yellow solid (0.32g, 52 %) ; 1H NMR (400 MHz, DMSO-c 6) δ ppm 1.48 (d, J=6.57 Hz, 6 H) 3.96 (s, 3 H) 5.04 (quin, .7=6.57 Hz, 1 H) 7.61 (dd, J=9.60, 2.27 Hz, 1 H) 8.03 (dt, J=8.46, 1.20 Hz, 1 H) 8.41 (s, 1 H). b) 6-fluoro-l-(l-methylethyl)-l/ -indazole-4-carboxylic acid
Figure imgf000041_0003
The title compound was prepared in the same manner as described for example 1 (step b) from methyl 6-fluoro- l -(l-methylethyl)- lH-indazole-4-carboxylate (0.31g, 1.312 mmol) and NaOH (1.750 mL, 5.25 mmol) The product was collected as a yellow solid (0.27g, 89%); :H NMR (400 MHz, DMSO- d6) 5ppm 1.48 (d, .7=6.57 Hz, 6 H), 5.03 (quin, J=6.63 Hz, 1 H) , 7.57 (dd, J=9.60, 2.27 Hz, 1 H), 7.97 (dd, J=9.35, 1.26 Hz, 1 H), 8.40 (s, 1 H),13.51 (br. s., 1 H)
The title compound was prepared in the same manner as described for example 1 (step c) from 6- fluoro- l -(l-methylethyl)- lH-indazole-4-carboxylic acid (.085g, 0.383 mmol),3-(aminomethyl)-4,6- dimethyl-2(lH)-pyridinone (0.108 g, 0.574 mmol), 1 -hydroxy-7-azabenzotriazole (0.078 g, 0.574 mmol), EDC (0.1 10 g, 0.574 mmol), and N-methylmorpho line (0.210 mL, 1.913 mmol). The product was collected as a white solid (117 mg, 82 %) !H NMR (400 MHz, DMSO-J6) δ ppm 1.42 - 1.50 (m, 6 H) 2.09 - 2.17 (m, 3 H) 2.21 (s, 3 H) 4.34 (d, J=5.05 Hz, 2 H) 4.98 (quin, J=6.57 Hz, 1 H), 5.88 (s, 1 H) 7.46 (dd, .7=9.98, 2.15 Hz, 1 H) 7.78 (dd, J=9.60, 1.26 Hz, 1 H) 8.36 (s, 1 H) 8.56 (t, J=4.93 Hz, 1 H) 11.53 (br. s., 1 H); MS(ES) [M+H]+ 357.2
Example 3
6-Bromo-Ar-[(4,6-dimethyl-2-oxo-l,2-dihydro-3-pyridinyl)methyl]-l-(l-methylethyl)-l /- indazole-4-carboxamide
Figure imgf000042_0001
a) Methyl 6-bromo-l-(l-methylethyl)-l//-indazole-4-carboxylate
Figure imgf000042_0002
To a cooled (0 °C) solution of methyl 6-bromo- lH-indazole-4-carboxylate (1.25g, 4.90 mmol) in N,N-dimethylformamide (25 mL) was added sodium hydride (0.216 g, 5.39 mmol). The reaction mixture was stirred for 15 min, then 2-bromopropane (0.920 mL, 9.80 mmol) was added and the reaction allowed to warm to RT. The reaction was maintained at RT overnight. The contents were concentrated to about 1/2 volume, then poured into saturated NH4C1 (200 mL) with stirring. The contents were extracted with ether (2x) and the combined organics washed with brine, dried (MgS04), filtered, and concentrated to give an orange residue (1.55 g crude). Purification by silica gel chromatography (eluent : 5-25% ethyl acetate in hexanes) gave methyl 6-bromo-l-(l-methylethyl)- lH-indazole-4-carboxylate (0.60 g, 40 % yield) and methyl 6-bromo-2-(l-methylethyl)-2H-indazole- 4-carboxylate (0.65 g, 43 % yield). Both products were isolated and methyl 6-bromo-l-(l- methylethyl)-lH-indazole-4-carboxylate was taken on to the next step. Data for 1 -alkylated isomer : !H NMR (400 MHz, CHLOROFORM- d) δ ppm 8.48 (s, 1 H) 8.02 (d, J=1.52 Hz, 1 H) 7.85 (s, 1 H) 4.83 (dt, J=13.33, 6.60 Hz, 1 H) 4.04 (s, 3 H) 1.63 (s, 3 H) 1.61 (s, 3 H); LC-MS(ES) [M+H]+
297.5/299.5. Data for 2-alkylated isomer : !H NMR (400 MHz, DMSO-J6) δ ppm 8.72 (s, 1 H), 8.25 (s, 1 H), 7.80 (d, J=1.5 Hz, 1 H), 4.96 (m, 1 H), 3.95 (s, 3 H), 1.57 (d, J=6.6 Hz, 6 H); LC-MS(ES) [M+H]+ 297.5/299.5. b) 6-bromo-l-(l-methylethyl)-l//-indazole-4-carboxylic acid
Figure imgf000043_0001
To a solution of 6-bromo-l-(l-methylethyl)-lH-indazole-4-carboxylate (0.58 g, 1.952 mmol in methanol (12 mL) and tetrahydrofuran (3 mL) was added 3 N NaOH (3.25 mL, 9.76 mmol) via syringe and stirred for 3 h at RT. The volatiles were removed in vacuo then diluted with water and slowly acidifed with 1 N HC1 to pH 4-5. The contents were extracted with 20% THF/EtOAc (3x). The combined organics were washed with brine, dried over MgS04, filtered, and concentrated to give 6-bromo-l-(l-methylethyl)-lH-indazole-4-carboxylic acid (0.52 g, 94 % yield). H NMR (400 MHz, DMSO-i/6) d ppm 13.54 (s, 1 H) 8.36 - 8.43 (m, 2 H) 7.82 (d, J=1.52 Hz, 1 H) 5.11 (quin, J=6.57 Hz, 1 H) 1.48 (s, 3 H) 1.47 (s, 3 H); LC-MS(ES) [M+H]+ 283.4/285.2
c) 6-Bromo-N-[(4,6-dimethyl-2-oxo-l,2-dihydro-3-pyridinyl)methyl]-l-(l-methylethyl)-l /- indazole-4-carboxamide
Figure imgf000043_0002
To a mixture of 6-bromo-l-(l-methylethyl)-lH-indazole-4-carboxylic acid (0.52g, 1.837 mmol), 3- (aminomethyl)-4,6-dimethyl-2(lH)-pyridinone (0.485 g, 2.57 mmol), 1 -hydroxy-7-azabenzotriazole (0.375 g, 2.76 mmol) and EDC (0.528 g, 2.76 mmol) in dimethyl sulfoxide (15 mL) was added N- methylmorpholine (0.606 mL, 5.51 mmol) via syringe. The reaction was stirred at RT for 48h. The contents were slowly diluted into 200 mL of water, stirred for 10 min, then allowed to sit for 20 min. The suspension was filtered. The collected solid was washed with about 50 mL of water, filtered, air dried for 15 min then dried in vac oven to afford the title compound (0.67g, 85 %), which was used without further purification. XH NMR (400 MHz, DMSO-c/6) δ ppm 1.45 (d, J=6.57 Hz, 6 H) 2.12 (s, 3 H) 2.20 (s, 3 H) 4.33 (d, J=5.05 Hz, 2 H) 5.05 (quin, J=6.57 Hz, 1 H) 5.89 (s, 1 H) 7.71 (d, J=1.26 Hz, 1 H) 8.20 (s, 1 H) 8.37 (s, 1 H) 8.62 (t, J=4.67 Hz, 1 H) 1 1.54 (s, 1 H). MS(ES) [M+H]+ 417.1. Example 4
6-bromo-N-((4-ethyl-6-methyl-2-oxo-l,2-dihydropyridin-3-yl)methyl)-l-isopropyl-lH-indazole- 4-carboxamide
Figure imgf000044_0001
In a 25 mL sealable tube under nitrogen were combined 6-bromo-l-(l-methylethyl)-lH-indazole-4- carboxylic acid (400 mg, 1.41 mmol) and 3-(aminomethyl)-4-ethyl-6-methyl-2(lH)-pyridinone.HCl (401 mg, 1.98 mmol) in DMSO (15 mL). 1 -hydroxy-7-azabenzotriazole (308 mg, 2.26 mmol) was added and the resulting mixture was degassed with nitrogen for 10 minutes. N-methylmorpholine (0.64 ml, 5.79 mmol) and EDC (433 mg, 2.26 mmol) were added, the vessel was sealed, and the mixture was stirred at room temperature for 2 days. The mixture was poured onto 10 mL of ice-water and solids crashed out. 10% K2C03 was added to adjust the pH~8-9 and the contents were stirred for 10 min and then allowed to stand for another 10 min. Solids were filtered and air-dried. DMF along with some water were added and solids that precipitated were filtered. DCM was added and the contents purified by Si02 chromatography (eluent: gradient 0 to 80:20:2 DCM/MeOH/NELjOH). The collected product was suspended in EtOAc along with some hexanes. Solids that precipitated were filtered and dried to afford the title compound as a white solid (504 mg, 82%). H NM (400 MHz, DMSO-6?6) δ ppm 1 1.54 (s, 1 H) 8.63 (t, .7=4.80 Hz, 1 H) 8.37 (s, 1 H) 8.20 (s, 1 H) 7.70 (d, .7=1.26
Hz, 1 H) 5.93 (s, 1 H) 5.06 (quin, J=6.57 Hz, 1 H) 4.37 (s, 1 H) 4.35 (s, 1 H) 2.52 - 2.58 (m, 2 H) 2.14 (s, 3 H) 1.47 (s, 3 H) 1.45 (s, 3 H) 1.10 (t, .7=7.58 Hz, 3 H); LC-MS (ES) m/z = 431.0/433.0
Example 5
6-bromo-l-(l-methylethyl)-/V-{[6-methyl-2-oxo-4-(phenylmethyl)-l,2-dihydro-3- pyridinyl]methyl}-l/i-indazole-4-carboxamide
Figure imgf000044_0002
The title compound was prepared in the same manner as described for the example 4 from 6-bromo- 1 - (l-methylethyl)-lH-indazole-4-carboxylic acid (100 mg, 0.353 mmol) and 3-(aminomethyl)-6- methyl-4-(phenylmethyl)-2(lH)-pyridinone (129 mg, 0.565 mmol), wherein the reaction stir time was 12h. The product was collected as a white solid (130 mg, 71%). ¾ NMR (400 MHz, DMSO-t/6) δ ppm 1 1.61 (s, 1H), 8.69 (s, 1H), 8.35 (s, 1H), 8.20 (s, 1H), 7.66 (s, 1H), 7.17-7.26 (m, 6H), 5.81 (s, 1H), 5.04 (m, 2H), 4.42 (s, 2H), 3.96 (s, 2H), 2.10 (s, 3H), 1.45 (d, 6H) ; MS(ES) [M+H]+ 495.0.
Example 6
6-bromo-l-(l-methylethyl)-A'-[(6-methyl-2-oxo-l,2-dihydro-4,4'-bipyridin-3-yl)methyl]-l/ - indazole-4-carboxamide
Figure imgf000045_0001
To a reaction vial were added 6-bromo- 1-(1 -methyl ethyl)- lH-indazole-4-carboxylic acid (90 mg, 0.318 mmol), 3-(aminomethyl)-6-methyl-4,4'-bipyridin-2(lH)-one (103 mg, 0.477 mmol), 1- hydroxy-7-azabenzotriazole (64.9 mg, 0.477 mmol), EDC (91 mg, 0.477 mmol) and DMSO (10 mL) followed by N-methylmorpholine (0.140 mL, 1.272 mmol) in one portion. The reaction contents were stirred at RT for 12 hr, after which time an addtional 20 mg each of amine, EDC, and HOAt were added. After stirring for an additional 2h, the reaction mixture was poured onto ice water (lOmL), stirred for 20 min, allowed to stand for 10 min, and filtered. The collected solid was rinsed with water (10 mL), and then purified by reverse phase HPLC (10-90% acetonitrile/water + 0.1% TFA). The product fractions were treated with NaHCOg (sat aq), extracted with EtOAc, and evaporated from water to aford the final product as a white solid (69 mg, 43%). XH NMR (400 MHz, DMSO-J6) 8 ppm 11.95 (s, 1H), 8.63-8.65 (d, 2H), 8.60 (s, 1H), 8.32 (s, 1H), 8.21 (s, 1H), 7.61 (s, 1H), 7.42 (d, 2H), 6.00 (s, 1H), 5.02 (m, 2H), 4.15 (s, 2H), 2.23 (s, 3H), 1.45 (d, 6H) ; MS(ES) [M+H]+ 481.8. Example 7
l-(l-methylethyl)-A-{[6-methyl-2-oxo-4-^henylmethyl)-l,2-dihydro-3^yridinyl]methyl}-l /- indazole-4-carboxamide
Figure imgf000046_0001
To a dried 50 mL round bottom flask was placed 10% Pd/C (0.020 g, 9.63 μηιοΐ), followed by degassing with N2. Added about 5 mL EtOH and then added 6-bromo-l-(l-methylethyl)-N- {[6- methyl-2-oxo-4-(phenylmethyl)-l,2-dihydro-3-pyridinyl]methyl}-lH-indazole-4-carboxamide (.095g, 0.193 mmol) and ethanol (5 mL) and THF (1 mL). Next added in triethylamine (0.081 mL, 0.578 mmol) via syringe. The mixture was stirred under atmosphere of hydrogen (balloon) for 6h. The mixture was purged with nitrogen and then diluted with DCM (10 mL) and a small amount of celite, After stirring for 10 min, the contents were then filtered through analytical grade celite and washed with 10% MeOH/DCM, EtOH, and then DCM. The filtrate was concentrated in vacuo and dried under hi vacuum overnight. The crude product was purified by silica gel chromatography (eluent: 5- 50% gradient of 10% methanol in dichloromethane and dichloromethane). The final product was concentrated from MTBE and dried in vacuum oven at 45 °C for 18h. The title compound was collected as a white solid (0.75g, 92 % yield); ¾ NMR (400 MHz, DMSO-c 6) δ ppm 1.48 (d, J=6.57 Hz, 6 H) 2.10 (s, 3 H) 3.98 (s, 2 H) 4.43 (d, J=5.05 Hz, 2 H) 5.03 (quin, J=6.63 Hz, 1 H) 5.81 (s, 1 H) 7.15 - 7.31 (m, 5 H) 7.39 (t, J=7.71 Hz, 1 H) 7.52 (d, J=6.82 Hz, 1 H) 7.85 (d, J=8.34 Hz, 1 H) 8.34 (s, 1 H) 8.50 (t, J=5.05 Hz, 1 H) 11.61 (s, 1 H) ; MS(ES) [M+H]+ 415.0.
Example 8
N-((4,6-dimethyl-2-oxo-l,2-dihydropyridin-3-yl)methyl)-6-(4-(2-
(dimethylamino)ethoxy)phenyl)-l-isopropyl-lH-indazole-4-carboxamide
Figure imgf000046_0002
6-bromo-N-(4,6-dimethyl-2-oxo- 1 ,2-dihydropyridin-3-yl)methyl)- 1 -isopropyl- 1 H-indazole-4- carboxamide (80 mg, 0.19 mmol), N,N-dimethyl-2-(4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)phenoxy)ethanamine (84 mg, 0.29 mmol) and PdCl2(dppf)-CH2Cl2 adduct (7.8 mg, 0.009 mmol) in dioxane/water (3 ml: l ml) were stirred for 10 min under nitrogen. Sodium bicarbonate (48.3 mg, 0.58 mmol) was added and the insoluble mixture was irradiated in a microwave at 100 °C for 20 min. The reaction mixture was evaporated, dissolved in DCM/MeOH (1 : 1), and preabsorbed on silica gel and purified using silica gel chromatography (eluent: DCM/MeOH/NE^OH; gradient 0 to 80:20:2 in
DCM). The isolated product was dissolved in hot DMSO/MeOH and purified using reversed-phase HPLC (25-80% gradient of MeCN in water with 0.1% TFA). Most of the solvent from the combined product fractions were evaporated and sat. sol. NaHC03 was added, solids that crashed out were filtered, air-dried for 15 min, and dried in vaccum-oven overnight. The product was collected as a white solid (56 mg, 56%). :H NMR (400 MHz, DMSO-c/6) δ ppm 11.54 (br. s., 1 H) 8.64 (t, J=4.80 Hz, 1 H) 8.35 (s, 1 H) 8.05 (s, 1 H) 7.81 - 7.84 (m, 2 H). LC-MS (ES) m/z = 528.1 [M+H]+ Example 9
N-[(4,6-dimethyl-2-oxo-l,2-dihydro-3-pyridinyl)methyl]-l-(l-methylethyl)-6-[4-(l- piperazinyl)phenyl]-lH-indazole-4-carboxamide
Figure imgf000047_0001
The title compound was prepared in a similar manner as described for example 8 from 6-bromo-l-(l- methylethyl)-N-[(6-methyl-2-oxo-4-propyl- 1 ,2-dihydro-3-pyridinyl)methyl]- 1 H-indazole-4- carboxamide (80 mg, 0.192 mmol) and [4-(l-piperazinyl)phenyl]boronic acid (59 mg, 0.288 mmol). The product was collected as a white solid (34 mg, 35%). :H NMR (400 MHz, DMSO-J6) δ ppm 11.53 (br. s., 1 H) 8.66 (br. s., 1 H) 8.33 (s, 1 H) 8.00 (s, 1 H) 7.82 (s, 1 H) 7.73 - 7.76 (m, 1 H) 7.72 (s, 1 H) 7.01 - 7.09 (m, 2 H) 5.89 (s, 1 H) 5.14 (dt, .7=13.33, 6.60 Hz, 1 H) 4.39 (br. s., 1 H) 4.38 (br. s., 1 H) 3.38 - 3.52 (m, 4 H) 3.09 - 3.14 (m, 1 H) 3.00 - 3.09 (m, 3 H) 2.81 - 2.90 (m, 1 H) 2.21 (s, 3 H) 2.13 (s, 3 H) 1.50 (s, 3 H) 1.48 (s, 3 H). LC-MS (ES) m/z = 499.4 [M+H]+
Example 10
/V-[(4,6-dimethyl-2-oxo-l,2-dihydro-3-pyridinyl)methyl]-l-(l-methylethyl)-6-[6-(l-piperazinyl)- 3-pyridinyl]-l//-indazole-4-carboxamide
Figure imgf000048_0001
The title compound was prepared in a similar manner as described for example 8 from 6-bromo-l-(l- methylethyl)-N-[(6-methyl-2-oxo-4-methyl- 1 ,2-dihydro-3-pyridinyl)methyl]- lH-indazole-4- carboxamide (90 mg, 0.216 mmol) and l-[5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-2- pyridinyl]piperazine (94 mg, 0.324 mmol). The product obtained from HPLC purification was treated with a saturated solution of NaHC03, and solids that crashed out were filtered . The product was evaporated from MeOH, triturated from ether and then CH3CN. After drying in vacuum oven overnight, the final product was collected as a white solid (87 mg, 78%). ¾ NMR (400 MHz, DMSO- ) δ ppm 1 1.53 (br. s., 1 H) 8.59 - 8.67 (m, 2 H) 8.54 (s, 1 H) 8.35 (s, 1 H) 8.02 - 8.09 (m, 2 H) 7.84 (s, 1 H) 6.94 (d, J=9.09 Hz, 1 H) 5.89 (s, 1 H) 5.14 (dt, .7=13.07, 6.47 Hz, 1 H) 4.39 (br. s., 1 H) 4.38 (br. s., 1 H) 3.41 - 3.47 (m, 4 H) 3.17 (s, 1 H) 2.22 (s, 3 H) 2.13 (s, 3 H) 1.50 (s, 3 H) 1.49 (s, 3 H). LC-MS (ES) m/z = 500.4 [M+H]+ Note: some piperinzyl proton atoms hidden under water peak.
Example 11
6-{4-[(dimethylamino)methyl]phenyl}-l-(l-methylethyl)-N-{[6-methyl-4-(l-methylethyl)-2-oxo- l,2-dihydro-3-pyridinyl]methyl}-lH-indazole-4-carboxamide
Figure imgf000048_0002
The title compound was prepared in a similar manner as described for example 8 from 6-bromo-l-(l- methylethyl)-N-{[6-methyl-4-(l-methylethyl)-2-oxo-l,2-dihydro-3-pyridinyl]methyl}-lH-indazole-4- carboxamide (90 mg, 0.202 mmol) and dimethyl{[4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)phenyl]methyl} mine (90 mg, 0.303 mmol). The product was collected as a white solid (54 mg, 49%). H NMR (400 MHz, DMSO-<i6) δ ppm 11.48 (br. s., 1 H) 8.69 (br. s., 1 H) 8.37 (s, 1 H) 8.10 (s, 1 H) 7.86 (s, 1 H) 7.82 (s, 1 H) 7.80 (s, 1 H) 7.42 (s, 1 H) 7.40 (s, 1 H) 6.02 (s, 1 H) 5.17 (dt, .7=13.14, 6.57 Hz, 1 H) 4.47 (d, .7=4.80 Hz, 2 H) 3.44 (s, 2 H) 3.24 (dt, .7=13.71, 6.92 Hz, 1 H) 2.17 (s, 6 H) 2.15 (s, 3 H) 1.51 (s, 3 H) 1.49 (s, 3 H) 1.11 (s, 3 H) 1.09 (s, 3 H). LC-MS (ES) m/z = 500.1 [M+H]+
Example 12
l-isopropyl-N-((4-isopropyl-6-methyl-2-oxo-l,2-dihydropyridin-3-yl)methyl)-6-(6-(piperazin-l- yl)pyridin-3-yl)-lH-indazole-4-carboxamide
Figure imgf000049_0001
In a 25 niL sealable tube under nitrogen were combined 6-bromo-l-(l-methylethyl)-N- {[6-methyl-4- (l-methylethyl)-2-oxo- l,2-dihydro-3-pyridinyl]methyl}-lH-indazole-4-carboxamide (90 mg, 0.2 mmol), { l-[5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-2-pyridinyl]piperazine (88 mg, 0.3 mmol) in dioxane/water (3 mL:lmL). PdCl2(dppi)-CH2Cl2 adduct (4.95 mg, 0.006 mmol) was added and the resulting mixture was degassed with nitrogen for 10 min. Sodium bicarbonate (50.9 mg, 0.61 mmol) was added, the vessel was sealed, and the insoluble mixture was heated in a microwave at 100 °C for 20 min. Upon cooling down, any solids that crashed out were filtered. DCM/MeOH (1 : 1) was added, the contents pre-absorbed on silica gel, and purified by Si02 chromatography (eluent: gradient 0 to 80:20:2 DCM/MeOH/NH4OH). The collected product was then further purified by reverse- phase HPLC (15% to 80% CH3CN in water with 0.1% TFA), which afforded the product as a TFA salt. CH3CN was evaporated and a saturated solution of sodium bicarbonate was added to the water layer followed by DCM/isopropanol (70:30). The organic layer was separated, and the aqueous layer was further extracted with DCM/isopropanol (70:30). The combined organic layers were washed with brine, dried over MgS04, filtered, and concentrated. DMF was added along with some water, and after sitting overnight, the solids that precipitated were filtered. EtOAc was added along with some hexanes, the contents sonicated, and the solids that precipitated were filtered and dried to afford the title compound as an off-white solid (29.5 mg, 26%). ¾ NMR (400 MHz, DMSO- 6) δ ppm 11.54 (br. s., 1 H) 8.59 - 8.68 (m, 2 H) 8.35 (s, 1 H) 8.00 - 8.12 (m, 2 H) 7.83 (s, 1 H) 6.92 (d, .7=8.84 Hz, 1 H) 6.03 (s, 1 H) 5.13 (quin, .7=6.63 Hz, 1 H) 4.47 (br. s., 1 H) 4.46 (br. s., 1 H) 3.42 - 3.56 (m, 4 H) 3.20 - 3.27 (m, 1 H) 2.76 - 2.85 (m, 4 H) 2.16 (s, 3 H) 1.50 (s, 3 H) 1.49 (s, 3 H) 1.11 (s, 3 H) 1.09 (s, 3 H). LC-MS (ES) m/z = 528.1 [M+H]+
Example 13
N-((4,6-dimethyl-2-oxo-l,2-dihydropyridin-3-yl)methyl)-l-isopropyl-6-(l-methyl-lH-pyrazol-4- yl)-lH-indazole-4-carboxamide
Figure imgf000050_0001
In a 25 mL sealable tube under nitrogen were combined 6-bromo-N-[(4,6-dimethyl-2-oxo-l,2- dihydro-3-pyridinyl)methyl]-l-(l-methylethyl)-lH-indazole-4-carboxamide (1 10 mg, 0.26 mmol), 1- methyl-4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-lH-pyrazole (82 mg, 0.4 mmol) in dioxane/water (3 mL: lmL). PdCl2(dppf)-CH2Ci2 adduct (10.8 mg, 0.013 mmol) was added and the resulting mixture was degassed with nitrogen for 10 min. Sodium bicarbonate (66.4 mg, 0.79 mmol) was added, the vessel was sealed, and the insoluble mixture was heated in a microwave at 100 °C for 20 min. The mixture was evaporated, DCM/MeOH (1 : 1) was added, and the contents pre-absorbed on silica gel and purified by Si02 chromatography (eluent: gradient 0 to 80:20:2 DCM/MeOH/NH4OH).
The collected product was suspended in EtOAc along with some hexanes, it was sonicated, and the solids that precipitated were filtered. Acetonitrile was added, solids that precipitated were filtered, and further washed with acetonitrile/ether (2:8) and dried to afford the title compound as an off-white solid (84 mg, 76 %). H NMR (400 MHz, DMSO- 6) δ ppm 1 1.54 (s, 1 H) 8.48 (t, J=4.93 Hz, 1 H) 8.31 (s, 1 H) 8.26 (s, 1 H) 8.04 (s, 1 H) 8.03 (s, 1 H) 7.81 (d, J=1.01 Hz, 1 H) 5.90 (s, 1 H) 5.06 (quin, J=6.57 Hz, 1 H) 4.39 (s, 1 H) 4.38 (s, 1 H) 3.90 (s, 3 H) 2.23 (s, 3 H) 2.13 (s, 3 H) 1.50 (s, 3 H) 1.48 (s, 3 H); LC-MS (ES) m/z = 419.0
Example 14
6-(4-((dimethylamino)methyl)phenyl)-N-((4-ethyl-6-methyl-2-oxo-l,2-dihydropyridin-3- yl)methyl)-l-isopropyl-lH-indazole-4-carboxamide
Figure imgf000051_0001
In a 25 mL sealable tube under nitrogen were combined 6-bromo-N-[(4-ethyl-6-methyl-2-oxo-l,2- dihydro-3-pyridinyl)methyl]-l-(l-methylethyl)-lH-indazole-4-carboxamide (90 mg, 0.21 mmol), dimethyl{[4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)phenyl]methyl}amine.HCl (93 mg, 0.31 mmol) in dioxane/water (3 mL: lmL). PdCl2(dppf)-CH2Cl2 adduct (8.52 mg, 0.01 mmol) was added and the resulting mixture was degassed with nitrogen for 10 min. Sodium bicarbonate (52.6 mg, 0.63 mmol) was added, the vessel was sealed, and the insoluble mixture was heated in a microwave at 110 °C for 20 min. The mixture was evaporated, DCM/MeOH (1 : 1) added, and the contents pre-absorbed on silica gel and purified by Si02 chromatography (eluent: gradient 0 to 80:20:2 DCM/MeOH/NH4OH). The collected product was further purified by reversed-phase HPLC (15% to
80% CH3CN in water with 0.1% TFA) which afforded the TFA salt. CH3CN was evaporated, and a saturated solution of sodium bicarbonate was added to the water layer along with EtOAc. The organic layer was separated, and the aqueous layer was further extracted with EtOAc, DCM and DCM/isopropanol (8:2). The combined organic layers were washed with brine, dried over MgS04, filtered and concentrated. The solid was triturated in ether and dried to afford the title compound as an-off-white solid (40 mg, 38%). ¾ NMR (400 MHz, DMSO-c¾) δ ppm 1 1.54 (s, 1 H) 8.67 (t, J=4.93 Hz, 1 H) 8.38 (s, 1 H) 8.11 (s, 1 H) 7.86 (s, 1 H) 7.82 (s, 1 H) 7.80 (s, 1 H) 7.42 (s, 1 H) 7.40 (s, 1 H) 5.93 (s, 1 H) 5.17 (quin, J=6.57 Hz, 1 H) 4.43 (s, 1 H) 4.41 (s, 1 H) 3.44 (s, 2 H) 2.57 (q, J=7.58 Hz, 2 H) 2.17 (s, 6 H) 2.14 (s, 3H), 1.51 (s, 3 H) 1.49 (s, 3 H) 1.10 (t, J=7.58 Hz, 3 H); LC-MS (ES) m/z = 486.3 [M+H]+
Example 15
l-(l-methylethyl)-N-[(6-methyl-2-oxo-4-propyl-l,2-dihydro-3-pyridinyl)methy]]-6-[6-(l- piperazinyl)-3-pyridinyl]-lH-indazole-4-carboxamide
Figure imgf000052_0001
The title compound was prepared in a similar manner as described for example 8 from 6-bromo-l-(l- methylethyl)-N-[(6-methyl-2-oxo-4-propyl- 1 ,2-dihydro-3-pyridinyl)methyl]- 1 H-indazole-4- carboxamide (90 mg, 0.202 mmol) and l-[5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-2- pyridinyl]piperazine (88 mg, 0.303 mmol). The final product was collected as a white solid (91 mg, 83%). H NMR (400 MHz, DMSO-i¾) δ ppm 11.54 (br. s., 1 H) 8.65 (d, .7=2.53 Hz, 1 H) 8.61 (t, .7=4.80 Hz, 1 H) 8.36 (s, 1 H) 8.03 - 8.07 (m, 2 H) 7.83 (s, 1 H) 6.92 (d, .7=9.09 Hz, 1 H) 5.92 (s, 1 H) 5.14 (quin, .7=6.63 Hz, 1 H) 4.42 (br. s., 1 H) 4.41 (br. s., 1 H) 3.45 - 3.51 (m, 4 H) 2.75 - 2.84 (m, 4 H) 2.54 (m, 2 H) 2.14 (s, 3 H) 1.51 - 1.58 (m, 2 H) 1.50 (s, 3 H) 1.49 (s, 3 H) 0.88 (t, .7=7.33 Hz, 3 H). LC-MS (ES) m/z = 528.0 [M+H]+
Example 16
N-((4-ethyl-6-methyl-2-oxo-l,2-dihydropyridin-3-y])methyl)-l-isopropyl-6-(6-(4- methylpiperazin-l-yl)pyridin-3-yl)-lH-indazole-4-carboxamide
Figure imgf000052_0002
In a 25 mL sealable tube under nitrogen were combined 6-bromo-N-[(4-ethyl-6-methyl-2-oxo-l,2- dihydro-3-pyridinyl)methyl]-l-(l-methylethyl)-lH-indazole-4-carboxamide (90 mg, 0.21 mmol), 1- methyl-4-[5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-2-pyridinyl]piperazine (95 mg, 0.31 mmol) in dioxane/water (3 mL: lmL). PdCl2(dppf)-CH2Cl2 adduct (8.52 mg, 0.01 mmol) was added and the resulting mixture was degassed with nitrogen for 10 min. Sodium bicarbonate (52.6 mg, 0.63 mmol) was added, the vessel was sealed, and the insoluble mixture was heated in a microwave at 1 10 °C for 20 min. The mixture was evaporated, DCM/MeOH (1 : 1) was added, and the contents pre-absorbed on silica gel and purified by Si02 chromatography (eluent: gradient 0 to 80:20:2 DCM/MeOH/NE^OH).
The collected product was further purified by reversed-phase HPLC (15% to 80% CH3CN in water with 0.1% TFA) which afforded the TFA salt. CH3CN was evaporated, and a saturated solution of sodium bicarbonate was added to the water layer along with EtOAc. The organic layer was separated, and the aqueous layer was further extracted with EtOAc, DCM and DCM/isopropanol (8:2). The combined organic layers were washed with brine, dried over MgS04, filtered and concentrated. EtOAc was added along with some hexanes, and the contents were sonicated and then allowed sit overnight at room temperature. Solids that precipitated were filtered and dried to afford the title compound (84 mg, 76%) as a white solid. ¾ NMR (400 MHz, DMSO- 6) δ ppm 1 1.54 (br. s., 1 H) 8.65 (d, J=2.27 Hz, 1 H) 8.62 (t, J=4.80 Hz, 1 H) 8.36 (s, 1 H) 8.04 - 8.08 (m, 2 H) 7.82 - 7.85 (m, 1 H) 6.97 (d, .7=8.84 Hz, 1 H) 5.94 (s, 1 H) 5.14 (quin, J=6.57 Hz, 1 H) 4.42 (s, 1 H) 4.41 (s, 1 H) 3.52 - 3.59 (m, 4 H) 2.56 (q, .7=7.58 Hz, 2 H) 2.39 - 2.44 (m, 4 H) 2.23 (s, 3 H) 2.14 (s, 3 H) 1.50 (s, 3 H) 1.49 (s, 3 H) 1.10 (t, J=7.58 Hz, 3 H); LC-MS (ES) m/z = 486.3 Example 17
l-isopropy]-N-((6-methyl-2-oxo-4-propyl-l,2-dihydropyridin-3-yl)methyl)-6-(2- methylpyrimidin-5-yl)-lH-indazole-4-carboxamide
Figure imgf000053_0001
In a 25 mL sealable tube under nitrogen were combined 6-bromo-l-(l-methylethyl)-N-[(6-methyl-2- oxo-4-propyl-l,2-dihydro-3-pyridinyl)methyl]-lH-indazole-4-carboxamide (100 mg, 0.23 mmol), 2- methyl-5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrimidine (64.2 mg, 0.29 mmol) in dioxane/water (3 mL: lmL). PdCl2(dppf)-CH2Cl2 adduct (9.2 mg, 0.01 1 mmol) was added and the resulting mixture was degassed with nitrogen for 10 min. Sodium bicarbonate (56.6 mg, 0.67 mmol) was added, the vessel was sealed, and the insoluble mixture was heated in a microwave at 1 10 °C for 20 min. The mixture was evaporated, DCM/MeOH (1 : 1) was added, and the contents pre-absorbed on silica gel and purified by Si02 chromatography (eluent: gradient 0 to 80:20:2 DCM/MeOH/NH4OH).
The collected product was suspended in acetonitrile along with a few drops of EtOH and some hexanes. The contents were sonicated, and solids that precipitated were filtered and dried to afford the title compound as an off-white solid (44 mg, 42%). H NMR (400 MHz, DMSO-<76) δ ppm 11.55 (s, 1 H) 9.21 (s, 2 H) 8.63 (t, .7=4.80 Hz, 1 H) 8.43 (s, 1 H) 8.32 (s, 1 H) 7.94 - 7.96 (m, 1 H) 5.92 (s, 1 H) 5.17 (quin, .7=6.63 Hz, 1 H) 4.43 (br. s., 1 H) 4.42 (br. s., 1 H) 2.69 (s, 3 H) 2.52 - 2.56 (m, 2 H) 2.14 (s, 3 H) 1.49 - 1.55 (m, 8 H) 0.88 (t, .7=7.33 Hz, 3 H); LC-MS (ES) mix = 459.2
Example 18
6-[6-(dimethylamino)-3-pyridinyl]-l-(l-methylethyl)-N- [(6-methyl-2-oxo-4-propyl-l,2-dihydro- 3-pyridinyl)methyl]-lH-indazole-4-carboxamide
Figure imgf000054_0001
The title compound was prepared in a similar manner as described for example 13 from 6-bromo- 1- (l-methylethyl)-N-[(6-methyl-2-oxo-4-propyl-l,2-dihydro-3-pyridinyl)methyl]- lH-indazole-4- carboxamide (100 mg, 0.225 mmol) and [6-(dimethylamino)-3-pyridinyl]boronic acid (49 mg, 0.292 mmol). The final product was collected as a white solid (49 mg, 44%). H NMR (400 MHz, DMSO- d6) δ ppm 1 1.55 (s, 1 H) 8.57 - 8.65 (m, 2 H) 8.35 (s, 1 H) 8.01 - 8.06 (m, 2 H) 7.83 (s, 1 H) 6.76 (d, 7=8.84 Hz, 1 H) 5.92 (s, 1 H) 5.13 (dt, J=13.14, 6.57 Hz, 1 H) 4.42 (br. s., 1 H) 4.41 (br. s., 1 H) 3.09 (s, 6 H) 2.54 (m, 2 H) 2.14 (s, 3 H) 1.51 - 1.58 (m, 2 H) 1.50 (s, 3 H) 1.49 (s, 3 H) 0.88 (t, ,7=7.33 Hz, 3 H). LC-MS (ES) m/z = 487.4 [M+H]+
Example 19
l-(l-methylethyl)-N-[(6-methyl-2-oxo-4-propyl-l,2-dihydro-3-pyridinyl)methyl]-6-[6-(4- morpholinyl)-3-pyridinyl]-lH-indazole-4-carboxamide
Figure imgf000054_0002
The title compound was prepared in a similar manner as described for example 8 from 6-bromo- 1 -(1 - methylethyl)-N-[(6-methyl-2-oxo-4-propyl- 1 ,2-dihydro-3-pyridinyl)methyl]- 1 H-indazole-4- carboxamide (100 mg, 0.225 mmol) and [6-(4-morpholinyl)-3-pyridinyl]boronic acid (70 mg, 0.337 mmol). The final product was collected as a white solid (98 mg, 81%). H NMR (400 MHz, DMSO- d6) δ ppm 1 1.46 (br. s., 1 H) 8.68 (d, .7=2.53 Hz, 2 H) 8.37 (s, 1 H) 8.07 - 8.12 (m, 2 H) 7.84 (d, .7=1.01 Hz, 1 H) 6.98 (d, .7=8.84 Hz, 1 H) 5.91 (s, 1 H) 5.14 (quin, .7=6.57 Hz, 1 H) 4.42 (d, .7=4.80 Hz, 2 H) 3.71 - 3.76 (m, 4 H) 3.50 - 3.56 (m, 4 H) 2.52 - 2.56 (m, 2 H) 2.13 (s, 3 H) 1.51 - 1.58 (m, 2 H) 1.51 (s, 3 H) 1.49 (s, 3 H) 0.88 (t, .7=7.33 Hz, 3 H). LC-MS (ES) m/z = 529.0 [M+H]+
Example 20
6-(2-amino-5-pyrimidinyl)-l-(l-methylethyl)-N-[(6-methyl-2-oxo-4-propy]-l,2-dihydro-3- pyridinyl)methyl]-lH-indazole-4-carboxamide
Figure imgf000055_0001
The title compound was prepared in the same manner as described for example 75 from 6-bromo- 1 - (1 -methylethyl)-N- [(6-methyl-2-oxo-4-propyl- 1 ,2-dihydro-3 -pyridinyl)methyl] - 1 H-indazole-4- carboxamide (100 mg, 0.225 mmol) and 5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-2- pyrimidinamine (75 mg, 0.337 mmol). The final product was collected as a white solid (87 mg, 82%). H NMR (400 MHz, DMSO-i/6) δ ppm 11.54 (s, 1 H) 8.78 (s, 2 H) 8.59 (t, .7=4.80 Hz, 1 H) 8.37 (s, 1 H) 8.11 (s, 1 H) 7.82 (s, 1 H) 6.87 (s, 2 H) 5.91 (s, 1 H) 5.14 (quin, .7=6.57 Hz, 1 H) 4.42 (s, 1 H) 4.41 (s, 1 H) 2.52 - 2.57 (m, 2 H) 2.13 (s, 3 H) 1.51 - 1.58 (m, 2 H) 1.50 (s, 3 H) 1.49 (s, 3 H) 0.88 (t, .7=7.33 Hz, 3 H). LC-MS (ES) m/z = 460.7
Example 21
6-(6-amino-3-pyridinyl)-l-(l-methylethy])-N-[(6-methyl-2-oxo-4-propyl-l,2-dihydro-3- pyridinyl)methyl]-lH-indazole-4-carboxamide
Figure imgf000055_0002
The title compound was prepared in the same manner as described for example 75 from 6-bromo- 1 - (l-methylethyl)-N-[(6-methyl-2-oxo-4-propyl-l,2-dihydro-3-pyridinyl)methyl]-lH-indazole-4- carboxamide (100 mg, 0.225 mmol) and 5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-2- pyridinamine (75 mg, 0.337 mmol). The final product was collected as a white solid (60 mg, 57%). H NMR (400 MHz, DMSO-<76) δ ppm 1 1.55 (s, 1 H) 8.61 (t, .7=4.93 Hz, 1 H) 8.45 (d, .7=2.27 Hz, 1 H) 8.34 (s, 1 H) 8.00 (s, 1 H) 7.90 (dd, J=8.59, 2.53 Hz, 1 H) 7.78 (s, 1 H) 6.55 (d, .7=8.59 Hz, 1 H) 6.10 - 6.17 (m, 2 H) 5.91 (s, 1 H) 5.13 (quin, .7=6.57 Hz, 1 H) 4.42 (br. s., 1 H) 4.41 (br. s., 1 H) 2.52 - 2.56 (m, 2 H) 2.13 (s, 3 H) 1.51 - 1.57 (m, 2 H) 1.50 (s, 3 H) 1.48 (s, 3 H) 0.88 (t, .7=7.33 Hz, 3 H). LC-MS (ES) m/z = 459.2
Example 22
l-(l-methylethyl)-N-[(6-methyl-2-oxo-4-propyl-l,2-dihydro-3-pyridinyl)methyl]-6-(5- pyrimidinyl)-lH-indazole-4-carboxamide
Figure imgf000056_0001
The title compound was prepared in the same manner as described for example 75 from 6-bromo- 1 - (1 -methylethyl)-N- [(6-methyl-2-oxo-4-propyl- 1 ,2-dihydro-3 -pyridinyl)methyl] - 1 H-indazole-4- carboxamide (100 mg, 0.225 mmol) and 5-pyrimidinylboronic acid (42 mg, 0.337 mmol). The final product was collected as a white solid (77 mg, 75%). lB NMR (400 MHz, DMSO-i¾) δ ppm 1 1.55 (s, 1 H) 9.35 (s, 2 H) 9.21 - 9.26 (m, 1 H) 8.63 (t, J=4.93 Hz, 1 H) 8.45 (s, 1 H) 8.38 (s, 1 H) 7.99 (d, 7=1.26 Hz, 1 H) 5.92 (s, 1 H) 5.18 (quin, J=6.57 Hz, 1 H) 4.44 (s, 1 H) 4.42 (s, 1 H) 2.52 - 2.56 (m, 2 H) 2.13 (s, 3 H) 1.52 - 1.56 (m, 4 H) 1.48 - 1.52 (m, 4 H) 0.88 (t, J=7.33 Hz, 3 H). LC-MS (ES) m/z = 445.2 Example 23
l-(l-methylethyl)-N- [(6-methyl-2-oxo-4-propyl-l ,2-dihydro-3-pyridinyl)methyl] -6- [6- (methyloxy)-3-pyridinyl]-lH-indazole-4-carboxamide
Figure imgf000057_0001
The title compound was prepared in the same manner as described for example 75 from 6-bromo- 1 - (l-methylethyl)-N-[(6-methyl-2-oxo-4-propyl-l,2-dihydro-3-pyridinyl)methyl]-lH-indazole-4- carboxamide (100 mg, 0.225 mmol) and [6-(methyloxy)-3-pyridinyl]boronic acid (52 mg, 0.337 mmol). The final product was collected as a white solid (72 mg, 67%). H NMR (400 MHz, DMSO- d6) 8 ppm 11.55 (br. s., 1 H) 8.68 (br. s., 1 H) 8.62 (br. s., 1 H) 8.39 (s, 1 H) 8.21 (br. s., 1 H) 8.14 (s, 1 H) 7.86 (s, 1 H) 6.97 (d, J=8.59 Hz, 1 H) 5.92 (s, 1 H) 5.09 - 5.21 (m, 1 H) 4.37 - 4.48 (m, 2 H) 3.92 (s, 3 H) 2.54 (br. s., 2 H) 2.14 (s, 3 H) 1.55 (m, 2H) 1.51 (br. s., 3 H) 1.50 (br. s., 3 H) 0.88 (t, J=7.20 Hz, 3 H). LC-MS (ES) m/z = 474.0
Example 24
l-(l-methylethyl)-A'-[(6-methyl-2-oxo-4-propyl-l,2-dihydro-3-pyridinyl)methyl]-6-[6-(4-methyl- l-piperazinyl)-3-pyridinyl]-l//-indazole-4-carboxamide
Figure imgf000057_0002
The title compound was prepared in a similar manner as described for example 8 from 6-bromo- 1-(1- methylethyl)-N-[(6-methyl-2-oxo-4-propyl- 1 ,2-dihydro-3-pyridinyl)methyl]- 1 H-indazole-4- carboxamide (90 mg, 0.202 mmol) and 1-methyl [5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-2- pyridinyljpiperazine (92 mg, 0.303 mmol). The final product was collected as a light brown solid (54 mg, 49%). H NMR (400 MHz, DMSO-<i6) δ ppm 1H NMR (400 MHz, DMSO- 6) δ ppm 11.54 (br. s., 1 H) 8.65 (d, J=2.27 Hz, 1 H) 8.61 (t, J=4.80 Hz, 1 H) 8.36 (s, 1 H) 8.04 - 8.08 (m, 2 H) 7.83 (s, 1 H) 6.96 (d, J=9.09 Hz, 1 H) 5.92 (s, 1 H) 5.14 (quin, J=6.57 Hz, 1H) 4.42 (d, J=4.80 Hz, 2 H) 3.53 - 3.59 (m, 4 H) 2.53 - 2.61 (m, 2 H) 2.40 - 2.45 (m, 4 H) 2.23 (s, 3 H) 2.14 (s, 3 H) 1.51 - 1.58 (m, 2 H) 1.50 (s, 3 H) 1.49 (s, 3 H) 0.88 (t, J=7.33 Hz, 3 H). LC-MS (ES) m/z = 542.2 [M+H]+ Example 25
l-(l-methylethyl)-N-[(6-methyl-2-oxo-4-propyl-l,2-dihydro-3-pyridinyl)methy]]-6-(2-methyl-3- pyridinyl)-lH-indazole-4-carboxamide
Figure imgf000058_0001
The title compound was prepared in a similar manner as described for example 8 from 6-bromo-l-(l- methylethyl)-N-[(6-methyl-2-oxo-4-propyl- 1 ,2-dihydro-3-pyridinyl)methyl]- 1 H-indazole-4- carboxamide (100 mg, 0.225mmol) and (2-methyl-3-pyridinyl)boronic acid (46 mg, 0.337 mmol). The product was collected as a white solid (68 mg, 66%). :H NMR (400 MHz, DMSO-i¾) δ ppm 11.44 (br. s., 1 H) 8.57 (br. s., 1H) 8.50 (dd, J=4.93, 1.64 Hz, 1 H) 8.43 (s, 1 H) 7.90 (s, 1 H) 7.73 (dd, J=7.71, 1.64 Hz, 1 H) 7.59 (d, J=1.26 Hz, 1 H) 7.34 (dd, J=7.33, 4.80 Hz, 1 H) 5.90 (s, 1 H) 5.10 (quin, J=6.57 Hz, 1 H) 4.40 (s, 1 H) 4.38 (s, 1 H) 2.46 (s, 3 H) 2.12 (s, 3 H) 1.50 - 1.58 (m, 2 H) 1.50 (s, 3 H) 1.48 (s, 3 H) 0.88 (t, J=7.33 Hz, 3 H). LC-MS (ES) m/z = 458.3 [M+H]+
Example 26
l-(l-methylethyl)-N-[(6-methyl-2-oxo-4-propyl-l,2-dihydro-3-pyridinyl)methy]]-6-(6-methyl-3- pyridinyl)-lH-indazole-4-carboxamide
Figure imgf000058_0002
The title compound was prepared in a similar manner as described for example 8 from 6-bromo-l-(l- methylethyl)-N-[(6-methyl-2-oxo-4-propyl- 1 ,2-dihydro-3-pyridinyl)methyl]- 1 H-indazole-4- carboxamide (100 mg, 0.225mmol) and (6-methyl-3-pyridinyl)boronic acid (46 mg, 0.337 mmol). The product was collected as a white solid (99 mg, 94%). :H NMR (400 MHz, DMSO-J6) δ ppm 11.47 (br. s., 1 H) 8.96 (d, J=2.02 Hz, 1 H) 8.67 (br. s., 1 H) 8.40 (s, 1 H) 8.20 (s, 1 H) 8.17 (dd, .7=8.08, 2.53 Hz, 1 H) 7.89 (s, 1 H) 7.39 (d, J=8.08 Hz, 1 H) 5.91 (s, 1 H) 5.18 (quin, J=6.57 Hz, 1 H) 4.43 (br. s., 1 H) 4.42 (br. s., 1 H) 2.52 - 2.58 (m, 5 H) 2.13 (s, 3 H) 1.52 - 1.58 (m, 2 H) 1.52 (s, 3 H) 1.50 (s, 3 H) 0.88 (t, J=7.33 Hz, 3 H). LC-MS (ES) m/z = 458.3 [M+H]+
Example 27
6-[6-(dimethylamino)-3^yridinyl]-N-[(4,6-dimethyl-2-oxo
methylethyl)-lH-indazole-4-carboxamide
Figure imgf000059_0001
The title compound was prepared in a similar manner as described for example 8 from 6-bromo-N- [(4,6-dimethyl-2-oxo- 1 ,2-dihydro-3-pyridinyl)methyl]- 1 -(1 -methylethyl)- lH-indazole-4-carboxamide (1 10 mg, 0.264 mmol) and [6-(dimethylamino)-3-pyridinyl]boronic acid (66 mg, 0.395 mmol). The product was collected as a white solid (33 mg, 27%). lB NMR (400 MHz, DMSO- d6) δ ppm 1 1.54 (s, 1 H) 8.64 (d, J=2.27 Hz, 1 H) 8.60 (t, J=4.80 Hz, 1 H) 8.35 (s, 1 H) 8.02 - 8.06 (m, 2 H) 7.84 (s, 1 H) 6.77 (d, J=8.84 Hz, 1 H) 5.89 (s, 1 H) 5.13 (dt, J=13.14, 6.57 Hz, 1 H) 4.39 (br. s., 1 H) 4.38 (br. s., 1 H) 3.09 (s, 6 H) 2.22 (s, 3 H) 2.13 (s, 3 H) 1.50 (s, 3 H) 1.49 (s, 3 H) LC-MS (ES) m/z = 459.2 [M+H]+
Example 28
N-[(4,6-dimethyl-2-oxo-l,2-dihydro-3-pyridinyl)methyl]-l-(l-methylethyl)-6-[6- (trifluoromethyl)-3-pyridinyl]-lH-indazole-4-carboxamide
Figure imgf000059_0002
The title compound was prepared in a similar manner as described for example 8 from 6-bromo-N- [(4,6-dimethyl-2-oxo- 1 ,2-dihydro-3-pyridinyl)methyl]- 1 -(1 -methylethyl)- lH-indazole-4-carboxamide (1 10 mg, 0.264 mmol) and [6-(trifluoromethyl)-3-pyridinyl]boronic acid (75 mg, 0.395 mmol). The product was collected as a solid (75 mg, 57%). !H NMR (400 MHz, DMSO-J6) δ ppm 1 1.54 (s, 1 H) 9.30 (d, J=2.02 Hz, 1 H) 8.66 (t, J=4.93 Hz, 1 H) 8.58 (dd, J=8.08, 2.02 Hz, 1 H) 8.45 (s, 1 H) 8.38 (s, 1 H) 8.07 (d, J=8.34 Hz, 1 H) 7.98 - 8.01 (m, 1 H) 5.90 (s, 1 H) 5.20 (dt, J=13.14, 6.57 Hz, 1 H) 4.41 (br. s., 1 H) 4.40 (br. s., 1 H) 2.22 (s, 3 H) 2.13 (s, 3 H) 1.53 (s, 3 H) 1.51 (s, 3 H). LC-MS (ES) m/z = 484.1 [M+H]+
Example 29
6-{3-[(dimethylamino)methyl]phenyl}-Ar-[(4,6-dimethyl-2-oxo-l,2-dihydro-3-pyridinyl)met l-(l-methylethyl)-l/f-indazole-4-carboxamide
Figure imgf000060_0001
The title compound was prepared in a similar manner as described for example 12 from 6-bromo- l- (1 -methylethyl)-N- [(6-methyl-2-oxo-4-methyl- 1 ,2-dihydro-3-pyridinyl)methyl]- lH-indazole-4- carboxamide (100 mg, 0.240 mmol) and dimethyl{[3-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)phenyl]methyl} amine (93 mg, 0.312 mmol). The product obtained from HPLC purification was treated with a saturated solution of NaHC03, solids that crashed out were filtered, and air-dried for 15 min. The product was then triturated first from EtOAc (containing EtOH) and then hexanes. The solid was then concentrated from MeOH, and triturated with ether, and then dried in vacuum oven overnight. The product was collected as a white solid (56 mg, 56%). !H NMR (400 MHz, DMSO-J6) δ ppm 1 1.53 (s, 1 H) 8.66 (t, J=4.93 Hz, 1 H) 8.37 (s, 1 H) 8.09 (s, 1 H) 7.83 (s, 1 H) 7.71 - 7.76 (m, 2 H) 7.46 (t, J=7.96 Hz, 1 H) 7.33 (d, J=7.58 Hz, 1 H) 5.89 (s, 1 H) 5.19 (quin, J=6.63 Hz, 1 H) 4.40 (s, 1 H) 4.39 (s, 1 H) 3.49 (s, 2 H) 2.22 (s, 3 H) 2.19 (s, 6 H) 2.12 (s, 3 H) 1.51 (s, 3 H) 1.49 (s, 3 H); LC- MS (ES) m/z = 472.2 [M+H]+
Example 30
6-{4-[(dimethylamino)methyl]phenyl}-N-[(4,6-dimethyl-2-oxo-l,2-dihydro-3-pyridinyl)methyl]- l-(l-methylethyl)-lH-indazole-4-carboxamide
Figure imgf000061_0001
The title compound was prepared in a similar manner as described for example 12 from 6-bromo- l- (1 -methylethyl)-N- [(6-methyl-2-oxo-4-methyl- 1 ,2-dihydro-3-pyridinyl)methyl]- lH-indazole-4- carboxamide (100 mg, 0.240 mmol) and dimethyl{[3-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-3- yl)phenyl]methyl} amine (93 mg, 0.312 mmol). The product obtained from HPLC purification was treated with a saturated solution of NaHC03, solids that crashed out were filtered, and air-dried for 15 min. The product was then triturated first from EtOAc and then hexanes. The solid was washed with acetonitrile, filtered, and then dried in vacuum oven overnight. The product was collected as a white solid (80 mg, 69%). H NMR (400 MHz, DMSO-<i6) δ ppm 11.53 (br. s., 1 H) 8.65 (t, .7=4.93 Hz, 1 H) 8.38 (s, 1 H) 8.11 (s, 1 H) 7.87 (d, J=1.01 Hz, 1 H) 7.83 (s, 1 H) 7.81 (s, 1 H) 7.42 (s, 1 H) 7.40 (s, 1 H) 5.89 (s, 1 H) 5.12 - 5.21 (m, 1 H) 4.40 (s, 1 H) 4.39 (s, 1 H) 3.44 (s, 2 H) 2.22 (s, 3 H) 2.17 (s, 6 H) 2.12 (s, 3 H) 1.51 (s, 3 H) 1.49 (s, 3 H). LC-MS (ES) m/z = 472.2 [M+H]+ 472.2 Example 31
N-((l,2-dihydro-6-methyl-2-oxo-4-propylpyridin-3-yl)methyl)-l-isopropyl-6-(thiophen-3-yl)-lH- indazole-4-carboxamide
Figure imgf000061_0002
To a stirred solution of 6-bromo-N-((l,2-dihydro-6-methyl-2-oxo-4-propylpyridin-3-yl)methyl)-l- isopropyl-lH-indazole-4-carboxamide (0.2 g, 0.44 mmol) in DMF (10 mL) was added 4,4,5,5- tetramethyl-2-(thiophen-3-yl)-l,3,2-dioxaborolane (0.113 g, 0.53 mmol) followed by PdCl2(PPh3)2 (0.06 g, 0.08 mmol) and the reaction mixture was stirred for 5 min. Sodium carbonate (0.119 g, 0.940 mmol) dissolved in water (2 mL) was added and the contents were stirred at 120 °C for 3 h. The reaction mixture was diluted with NaHCC>3 solution and extracted with ethyl acetate (3x20 mL). The combined organic layers were washed with brine solution (20 mL), dried over anhydrous Na2S04 , filtered, and concentrated in vacuo to afford the crude product. The crude product was purified by silica gel chromatography (eluent: 4% MeOHYEtOAc) to afford the title compound as an off white solid (75 mg, 38 %) . !H NM (DMSO-d6; 400 MHz): δ 0.883 (t, J = 7.2 Hz, 3H), 1.491-1.550 (m, 8H), 2.13 (s, 3H), 2.501 (s, 2H), 4.419 (d, J = 3.6 Hz, 2H), 5.130 (t, J = 6.4 Ηζ,ΙΗ), 5.912 (s, 1H), 7.691 (s, 1H), 7.766 (d, J = 4.4 Hz, 1H), 7.930 (s, 1H), 8.041 (s, 1H), 8.171 (s, 1H), 8.353 (s, 1H), 8.563 (s, 1H), 1 1.530 (s, 1H). LCMS (ES+) m/z: 449.09.
Example 32
l-isopropyl-N-((6-methyl-2-oxo-4-propyl-l,2-dihydropyridin-3-yl)methyl)-6-(thiophen-3-yl)-lH- indazole-4-carboxamide
Figure imgf000062_0001
The title compound was prepared from 6-bromo-N-((l,2-dihydro-6-methyl-2-oxo-4-propylpyridin-3- yl)methyl)-l-isopropyl-lH-indazole-4-carboxamide (0.2 g, 0.44 mmol) and 2-(furan-3-yl)-4,4,5,5- tetramethyl- l,3,2-dioxaborolane (0.1 g, 0.51 mmol) in the same manner as described for example 31. The product was collected as an off-white solid (55 mg, 25.7 %). ¾ NMR (DMSO-d6; 400 MHz): δ 0.896 (t, J = 7.2 Hz, 3H), 1.483-1.576 (m, 8H), 2.138 (s, 3H), 2.533 (s, 2H), 4.408 (d, J = 4.4 Hz, 2H), 5.076-5.140 (m, 1H), 5.915 (s, 1H), 6.656 (s, 1H), 7.105 (d, J = 2.8 Hz, 1H), 7.814 (s, 1H), 7.909 (s, 1H), 8.092 (s, 1H), 8.340 (s, 1H), 8.573 (s, 1H), 11.540 (s, 1H). LCMS (ES+) m/z: 433.1
Example 33
6-{4-[(dimethylamino)methyl]phenyl}-l-(l-methylethyl)-N-[(6-methyl-2-oxo-4-propyl-l,2- dihydro-3-pyridinyl)methyl]-lH-indazole-4-carboxamide hydrochloride salt
Figure imgf000062_0002
In a glass pressure tube was added 6-bromo-l-(l-methylethyl)-N-[(6-methyl-2-oxo-4-propyl-l,2- dihydro-3-pyridinyl)methyl]-lH-indazole-4-carboxamide (200 mg, 0.449 mmol), 4-(N,N- dimethylaminomethyl)phenylboronic acid pinacol ester hydrochloride (170 mg, 0.571 mmol), potassium phosphate (300 mg, 1.413 mmol), dioxane (12 mL) and water (3 mL). The suspension was stirred and purged with N2 then PdCl2(dppf)-CH2Cl2 adduct (50 mg, 0.061 mmol) was added. The reaction mixture was capped and stirred at 110 °C for 4 hr. The dark black reaction mixture was evaporated to dryness. The crude product was purified by silica gel chromatography (eluent: 0 to 70%, 20% (5% NH4OH in MeOH) / CH2C12) in CH2C12). The pure fractions were combined and evaporated to dryness to give the product as a very dark brown-black solid. The solid was taken up in CH2C12 and treated with Silicycle Si-Thiol (2 g, 1.46 mMol/g). After stirring for 30 minutes on a rotary evaporator (no vacuum) the mixture was filtered through a pad of Celite, washed with CH2C12, and evaporated to dryness. The light yellow colored solid was taken up in a small volume of MeOH and treated with 6 N HC1 (200 uL) and re-evaporated to dryness. The residue was dissolved in a small volume of MeOH, ppt. with Et20, triturated, filtered and dried under vacuum to give the product 6- {4- [(dimethylamino)methyl]phenyl} - 1 -(1 -methylethyl)-N-[(6-methyl-2-oxo-4-propyl- 1 ,2-dihydro-3- pyridinyl)methyl]-lH-indazole-4-carboxamide hydrochloride salt as a light tan solid (205 mg, 85 % yield). :H NMR (400MHz ,DMSO-d6) δ = 11.60 (br. s., 1 H), 10.81 (br. s., 1 H), 8.69 (t, J = 4.9 Hz,
1 H), 8.40 (s, 1 H), 8.19 (s, 1 H), 7.97 (d, J = 8.3 Hz, 2 H), 7.90 (d, J= 1.0 Hz, 1 H), 7.71 (d, J= 8.3 Hz, 2 H), 5.94 (s, 1 H), 5.18 (dt, J= 6.7, 13.2 Hz, 1 H), 4.43 (d, J= 4.8 Hz, 2 H), 4.34 (d, J = 5.3 Hz,
2 H), 2.72 (d, J = 5.1 Hz, 6 H), 2.58 - 2.52 (m, 2 H), 2.14 (s, 3 H), 1.59 - 1.52 (m, 2 H), 1.51 (d, J =
6.6 Hz, 6 H), 0.89 (t, J= 7.3 Hz, 3 H); LC-MS(ES)+ m/e 500.2 [M+H]+.
Example 34
6-{3-[(dimethylamino)methyl]phenyl}-l-(l-methylethyl)-N-[(6-methyl-2-oxo-4-propyl-l,2- dihydro-3-pyridinyl)methyl]-lH-indazole-4-carboxamide hydrochloride salt
Figure imgf000063_0001
The title compound was prepared in the same manner as described for example 33 using from 6- bromo- 1 -( 1 -methylethyl)-N- [(6-methyl-2-oxo-4-propyl- 1 ,2-dihydro-3 -pyridinyl)methyl] - 1 H- indazole-4-carboxamide (200 mg, 0.449 mmol), 3-(N,N dimethylaminomethyl)phenylboronic acid pinacol ester hydrochloride (170 mg, 0.571 mmol), potassium phosphate (300 mg, 1.413 mmol), dioxane (12 mL), water (3 mL), and PdCl2(dppf)-CH2Cl2 adduct (50 mg, 0.061 mmol). The product was obtained as a light tan solid (204 mg, 85 % yield). ¾ NMR (400MHz ,DMSO-d6) δ = 11.67 (br. s., 1 H), 10.97 (br. s., 1 H), 8.68 (t, J = 5.1 Hz, 1 H), 8.38 (s, 1 H), 8.30 (s, 1 H), 8.16 (s, 1 H), 8.00 - 7.94 (m, 1 H), 7.92 (d, J= 1.0 Hz, 1 H), 7.65 - 7.53 (m, 2 H), 5.96 (s, 1 H), 5.18 (dt, J= 6.6, 13.3 Hz, 1 H), 4.43 (d, J = 5.1 Hz, 2 H), 4.38 (d, J = 5.6 Hz, 2 H), 2.74 (d, .7 = 4.8 Hz, 6 H), 2.61 - 2.52 (m, 2 H), 2.15 (s, 3 H), 1.59 - 1.52 (m, 2 H), 1.51 (d, J = 6.6 Hz, 6 H), 0.90 (t, J = 7.3 Hz, 3 H); LC-
MS(ES)+ m/e 500.2 [M+H]+
Example 35
N-((l,2-dihydro-6-methyl-2-oxo-4-propylpyridin-3-yl)methyl)-l-isopropyl-6-(lH-pyrazol-4-yl)- lH-indazole-4-carboxamide
Figure imgf000064_0001
a) Methyl l-isopropyl-6-(lH-pyrazol-4 arboxylate
Figure imgf000064_0002
To a stirred solution of methyl 6-bromo- l-isopropyl-lH-indazole-4-carboxylate, 1 (0.8 g, 2.7 mmol) in 1,4-dioxane (20 mL) were added 4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-lH-pyrazole (0.576 g, 2.97 mmol) and NaHC03 (0.567 g, 6.75 mmol) dissolved in water (8 mL). The reaction mixture was degassed with argon gas for 30 min. To the resulting mixture was added PdCl2(dppf)- CH2CI2 adduct (0.110 g, 0.135mmol) and the mixture heated at reflux for 3 h. The reaction mixture was diluted with water (30 mL) and extracted with ethyl acetate (100 mL). The organic layer was dried over anhydrous Na2S04, filtered, and concentrated under reduced pressure to afford 800 mg of crude product. The crude compound was purified by silica gel chromatography (eluent: 5% MeOH/DCM) to the title compound as a pale liquid (375 mg, 47% ). LCMS (ES-) m/z = 283.11. b) l-isopropyl-6-(lH-pyrazol-4-yl)-lH-indazole-4-carboxylic acid
Figure imgf000065_0001
To a stirred solution of methyl l-isopropyl-6-(lH-pyrazol-4-yl)- lH-indazole-4-carboxylate (750 mg, 2.64 mmol) in THF:H20 (30 mL) was added LiOH H20 (330 mg, 7.85 mmol) and the resulting mixture was refluxed at 80 °C for 8 h. THF was distilled off under reduced pressure and the aqueous layer was acidified with 10% HC1 (to pH ~5 ) at 0 °C. The precipitated solid was collected by filtration and dried to afford the title compound l-isopropyl-6-(lH-pyrazol-4-yl)-lH-indazole-4- carboxylic acid as an off-white solid (540 mg, 76%). LCMS (ES+) m/z = 271.09. c) N-((l,2-dihydro-6-methyl-2-oxo-4-propylpyridin-3-yl)methyl)-l-isopropyl-6-(lH-pyrazol-4- yl)-lH-indazole-4-carboxamide
To a stirred solution of N-((l,2-dihydro-6-methyl-2-oxo-4-propylpyridin-3-yl)methyl)-l-isopropyl-6- (lH-pyrazol-4-yl)-lH-indazole-4-carboxamide (550 mg, 2.037mmol) in DCM (20 mL) were added EDC HCl (466 mg, 2.44 mmol), HOBT (374 mg, 2.44 mmol) and DIPEA (1.8 mL, 10.18 mmol) and the contents stirred at room temperature for 15 min. To the resulting mixture 3-(amino methyl)-4,6- dimethylpyridin-2(lH)-one (338 mg, 2.037 mmol) was added and stirred at RT for 3 h. The reaction mixture was diluted with DCM (50 mL) and washed with water (20 mL). The DCM layer was dried over anhydrous sodium sulphate, filtered, and concentrated to afford 200 mg of crude product. The crude product was diluted with DCM (5 mL), filtered, and dried to afford N-((l,2-dihydro-6-methyl- 2-oxo-4-propylpyridin-3 -yl)methyl)- 1 -isopropyl-6-(l H-pyrazol-4-yl)- 1 H-indazole-4-carboxamide as an off-white solid (80 mg, 10%).^ NMR(DMSO-d6, 400 MHz): δ 0.907-0.87 l(t, 3H, J = 14.4 Hz), 1.55-1.48 (m, 8H), 2.134 (s, 3H), 2.555 (s, 2H), 4.417-4.406 (d, 1H, J = 4.4 Hz), 5.059 (m, 1H), 5.913 (s, 1H), 7.830 (s, 1H), 8.047 (s, 1H), 8.080 (s, 1H), 8.306 (s, 2H), 8.462 (s, 1H), 1 1.53 (s, 1H), 13 (bs, 1H). LCMS (ES+) m/z = 433.17.
Example 36
N-((l,2-dihydro-6-methyl-2-oxo-4-propy]pyridin-3-yl)methyl)-l-isopropyl-6-(lH-pyrrolo[2,3- b] py ridin-5-yl)- 1 H-indazole-4-carboxamide a) Methyl l-isopropyl-6-(lH-p ndazole-4-carboxylate
Figure imgf000066_0001
The title compound was prepared in the same manner as described for example 35 (step a) from methyl 6-bromo-l-isopropyl-3 -methyl- lH-indazole-4-carboxy late (1 g, 3.36 mmol) and 5-(4,4,5,5- tetramethyl- l,3,2-dioxaborolan-2-yl)-lH-pyrrolo[2,3-b]pyridine (0.98 g, 4.04 mmol) wherein the reaction mixture was stirred at 100 °C for 5 h. The product was collected as an off white solid (600 mg, 53%). LCMS (ES-) m/z: 333.08. b) l-isopropyl-6-(lH-pyrrolo[2,3-b]pyridin-5-yl)-lH-indazole-4-carboxylic acid
Figure imgf000066_0002
The title compound was prepared in the same manner as described for example 35 (step b) from methyl l-isopropyl-6-(lH-pyrrolo[2,3-b]pyridin-5-yl)-lH-indazole-4-carboxylate 1 (0.6 g, 1.79 mmol) and LiOH H20 (0.22 g, 5.37 mmol) wherein the mixture was heated at 80 °C for 5 h. The product was collected as an off-white solid (0.5 g, 87.7%). ¾ NMR (DMSO-d6, 400 MHz) : 81.530 (d, J = 6.4 Hz, 6H), 5.199-5.265 (m, 1H), 6.552 (d, J = 1.6 Hz, 1H), 7.550 (t, J = 2.4 Hz, 1H), 8.11 (s,
1H), 8.328 (s, 1H), 8.383 (d, J = 1.6 Hz, 1H ), 8.401 (s, 1H), 8.671 (d, J
1H). LCMS (ES+) m/z: 321.24. c) N-((l,2-dihydro-6-methyl-2-oxo-4-propylpyridin-3-y])methyl)-l-isopropyl-6-(lH-pyrrolo[2,3- b] py ridin-5-yl)- 1 H-indazole-4-carboxamide
The title compound was prepared in the same manner as described for example 35 (step c) from 1- isopropyl-3-methyl-6-(pyridin-3-yl)-lH-indazole-4-carboxylic acid and 3-(aminomethyl)-6-methyl-4- propylpyridin-2(lH)-one (0.28 g, 1.56 mmol). The crude product was purified by silica gel chromatography (eluent: 5% MeOHYEtOAc) to afford the product as an off-white solid (200 mg, 26 %). H NM (DMSO-d6, 400 MHz) : 50.886 (t, J = 7.2 Hz, 3H), 1.513 (d, J = 6.4 Hz, 8H), 2.131 (s, 3H), 2.552 (s, 2H), 4.431 (d, J = 4.4 Hz, 1H), 5.153-5.216 (m, 1H), 5.91 1 (s, 1H), 6.535 (d, J = 2Hz, 2H), 7.535 (s, 1H), 7.930 (s, 1H), 8.174 (s, 1H), 8.40 (d, J = 9.6 Hz, 2H), 8.656 (s, 1H), 8.730 (s, 1H), 11.526 (brs, 1H), 1 1.756 (brs, 1H); LC-MS (ES+) m/z: 483.19.
Example 37
6-(4-(dimethylamino)piperidin-l-yl)-l-isopropyl-N-((6-methyl-2-oxo-4-propyl-l,2-dihydropyridin-3- yl)methyl)-lH-indazole-4-carboxamide
Figure imgf000067_0001
a) Methyl 6-(4-(dimethylamino)piperidin-l-yl)-l-isopropy]-lH-indazole-4-carboxylate
Figure imgf000067_0002
To a 10-mL microwave tube were added methyl 6-bromo-l-(l-methylethyl)-lH-indazole-4- carboxylate (200 mg, 0.673 mmol), N,N-dimethyl-4-piperidinamine (95 mg, 0.740 mmol), toluene (3 mL), and cesium carbonate (329 mg, 1.010 mmol), and the mixture was degassed for 5 min by bubbling N2. BINAP (62.9 mg, 0.101 mmol) and Pd2(dba)3 (30.8 mg, 0.034 mmol) were added. The tube was sealed and the mixture was heated at 115 °C with stirring for 15h. The mixture was diluted with methanol (5 mL) and filtered. The filtrate was concentrated and the residue was purified using reverse-phase HPLC to give 58 mg of product as an off-white solid. ¾ NMR (400 MHz, DMSO-J6) δ ppm 1.45 (d, J = 8.0 Hz, 6 H), 1.54 - 1.61 (m, 2 H), 1.87 - 1.96 (m, 2 H), 2.31 (m, 6H), 2.80-2.83 (m, 2H), 3.83-3.86 (m, 2H), 3.93 (s, 3H), 4.98-5.04 (m, 1H), 7.29 (s, 1H), 7.57 (s, 1H), 8.17 (s, 1H).
LCMS: (M+H)+=345.2 b) 6-(4-(dimethylamino)piperidin-l-yl)-l-isopropyl-lH-indazole-4-carboxylic acid
Figure imgf000068_0001
To a solution of methyl 6-bromo-3-methyl-l-(l-methylethyl)-lH-indole-4-carboxylate (520 mg, 1.676 mmof in methanol (8 mL) and tetrahydrofuran (3.00 mL) was added sodium hydroxide (2.79 mL, 8.38 mmol), anc the mixture was stirred overnight. The mixture was concentrated to remove organic solvents and diluted witl water (5 mL). The aqueous solution was acidified to ~pH 3 using IN HC1 and the precipitate was collected bj filtration and dried under high vacuum to give 480 mg of product as a white solid. ¾ NMR (400 MHz. DMSO-6?6) δ ppm 1.45- 1.46 (d, J= 8.0 Hz, 6H), 1.75 - 1.78 (m, 2 H), 2.10-2.12 (m, 2H), 2.80-2.89 (m, 8H): 3.35-3.40 (m, 1H), 3.97-4.00 (m, 2H), 4.97-5.04 (m, 1H), 7.34 (s, 1H), 7.58 (s, 1H), 8.18 (s, 1H); LCMS: (M+H)+=296.3 c) 6-(4-(dimethylamino)piperidin-l-yl)-l-isopropyl-N-((6-methyl-2-oxo-4-propyl-l,2-dihydropyridin-3- yl)methyl)-lH-indazole-4-carboxamide
To a solution of 6-[4-(dimethylamino)-l-piperidinyl]-l-(l-methylethyl)-lH-indazole-4-carboxylic acid (34 mg, 0.103 mmol) in DMSO (1 mL) were added 3-(aminomethyl)-6-methyl-4-propyl-2(lH)- pyridinone (29.0 mg, 0.134 mmol), N-methylmorpholine (0.045 mL, 0.412 mmol), l-hydroxy-7- azabenzotriazole (28.0 mg, 0.206 mmol) and EDC (39.5 mg, 0.206 mmol), and the mixture was stirred for 48h. The mixture was purified using reverse-phase HPLC to afford the title compound as a pale yellow solid (34 mg). !H NMR (400 MHz, DMSO-i¾) δ ppm 0.77 - 0.96 (m, 3 H), 1.43 (d, J = 6.57 Hz, 6 H), 1.46 - 1.66 (m, 4 H), 1.89 (m, 2 H), 2.13 (s, 3 H), 2.31 (s, 6H), 2.33( m, 2 H), 2.55 (s, 3 H), 2.65 - 2.84 (m, 2 H), 3.86 (m. 2 H), 4.38 (m, 2 H), 4.85 - 5.04 (m, 1 H), 5.91 (s, 1 H), 7.07 (s, 1 H), 7.32 (m, 1 H), 8.15 (m, 1 H), 8.25 (br. s., 1 H), 8.44 (t, J= 4.93 Hz, 1 H); LCMS: (M+H)+=493.2 Example 38
6-(4-((dimethylamino)methyl)piperidin-l-yl)-l-isopropyl-N-((6-methyl-2-oxo-4-propyl-l,2- dihydropyridin-3-yl)methyl)-lH-indazole-4-carboxamide
Figure imgf000069_0001
a) Methyl 6-(4-((dimethylamino)methyl)piperidin-l-yl)-l-isopropyl-lH-indazole-4-carboxylate
Figure imgf000069_0002
To a 10-mL microwave tube were added methyl 6-bromo-l-(l-methylethyl)-lH-indazole-4-carboxylate (25C mg, 0.841 mmol), N,N-dimethyl-l-(4-piperidinyl)methanamine (132 mg, 0.925 mmol), Toluene (3 mL), anc cesium carbonate (411 mg, 1.262 mmol), and the mixture was degassed for 5 min by bubbling N2 through ΒΓΝΑΡ (79 mg, 0.126 mmol) and Pd2(dba)3 (38.5 mg, 0.042 mmol) were added. The tube was sealed and the mixture was heated at 1 15 °C with stirring for 15h. The mixture was diluted with methanol (5 mL) anc filtered. The fitrate was concentrated and the residue was purified using reverse-phase HPLC to give 35 mg ol product as a off-white solid. LCMS MH+ = 359.3 b) 6-(4-((dimethylamino)methyl)piperidin-l-yl)-l-isopropyl-lH-indazole-4-carboxylic acid
Figure imgf000069_0003
The title compound was prepared using the procedure described for example 37 (step b) ¾ NMR (400 MHz, DMSO-< ) δ ppm 1.26- 1.29 (m, 2 H), 1.43-1.45 (m, 6 H), 2.27-2.30 (m, 8 H), 2.76-2.78 (m, 2 H), 3.77-3.81 (m, 2 H), 3.97-4.00 (m, 2 H), 4.97-5.03 (m, 1 H), 7.20 (s, 1 H), 7.53 (s, 1 H), 8.16 (s, 1 H); LCMS:
(M+H)+=345.2 c) 6-(4-((dimethylaraino)raethyl)piperidin-l-yl)-l-isopropyl-N-((6-raethyl-2-oxo-4-propyl-l,2- dihydropyridin-3-yl)methyl)-lH-indazole-4-carboxamide
The title compound was prepared using the procedure described for example 37 (step c) from 6-(4- ((dimethylamino)methyl)piperidin- 1 -yl)- 1 -isopropyl- 1 H-indazole-4-carboxylic acid and 3 -(aminomethyl)-6- methyl-4-propyl-2(lH)-pyridinone. H NMR (400 MHz, DMSO-<f6) δ ppm 0.89 (t, J = 7.33 Hz, 3 H), 1.14■ 1.32 (m, 2 H), 1.42 (m, 6 H), 1.45 - 1.55 (m, 2 H), 1.57 - 1.68 (m, 1 H), 1.72 - 1.89 (m, 3 H), 2.03 - 2.21 (m 12 H), 2.63 - 2.79 (m, 2 H), 3.54 - 3.66 (m, 1 H), 3.81 (m, 2 H), 4.37 (d, J = 4.80 Hz, 2 H), 4.94 (quin, J = 6.63 Hz, 1 H), 5.91 (s, 1 H), 7.05 (s, 1 H), 7.32 (d, J=1.77 Hz, 1 H), 8.14 (s, 1 H), 8.44 (t, J= 5.05 Hz, 2 H) 11.24 - 11.80 (m, 1 H); LCMS: (M+H)+=507.1
Example 39
N-((4-ethyl-6-methyl-2-oxo-l,2-dihydropyridin-3-yl)methyl)-l-isopropyl-6-morpholino-lH- indazole-4-carboxamide
Figure imgf000070_0001
a) l-isopropyl-6-morpholino-lH-indazole-4-carboxylic acid
Figure imgf000070_0002
In a 25 mL sealable tube under nitrogen were combined methyl 6-bromo-l-(l-methylethyl)-lH- indazole-4-carboxylate (250 mg, 0.84 mmol), sodium tert-butoxide (178 mg, 1.85 mmol), Pd-XPhos precatalyst (chloro(2-dicyclohexylphosphino-2',4',6'-tri-i-propyl-l,r-biphenyl)[2-(2- aminoethyl)phenyl]Pd(II); Me-t-butylether adduct) (34.8 mg, 0.042 mmol) and morpholine (81 mg, 0.93 mmol) in Dioxane (4 mL). The resulting mixture was degassed with nitrogen for 10 minutes. The vessel was sealed and the mixture was heated at 98 °C for 20 hours. The contents were diluted with water and IN HCl was added to adjust the pH~3-4. The contents were extracted with EtOAc and DCM/isopropanol (8:2), and the combined organic layers were washed with brine, dried over MgSO/i, filtered and concentrated. The residue was dissolved in DCM and purified by S1O2 chromatography (eluent: Hexanes/EtOAc gradient 0 to 100% EtOAc). Fractions were evaporated and solids were dried to afford the title compound as a light yellow solid (115 mg, 46%). LC-MS (ES) m/z = 290.2. lB NMR (400 MHz, DMSO-< 6) δ ppm 13.13 (br. s., 1 H) 8.18 (s, 1 H) 7.56 (d, J=2.02 Hz, 1 H) 7.26 - 7.29 (m, 1 H) 5.00 (quin, .7=6.57 Hz, 1 H) 3.77 - 3.81 (m, 4 H) 3.22 - 3.26 (m, 4 H) 1.46 (s, 3 H) 1.44 (s, 3 H) b) In a 25 mL sealable tube under nitrogen were combined l-(l-methylethyl)-6-(4-moφholinyl)- lH-indazole-4-carboxylic acid (55 mg, 0.19 mmol) and 3-(aminomethyl)-4-ethyl-6-methyl- 2(lH)-pyridinone.HCl (57.8 mg, 0.29 mmol) in DMSO (2 mL). l-hydroxy-7- azabenzotriazole (44 mg, 0.32 mmol) was added and the resulting mixture was degassed with nitrogen for 10 minutes. N-methylmorpholine (0.088 ml, 0.8 mmol) and EDC (62 mg, 0.32 mmol) were added, the vessel was sealed, and the mixture was stirred at room temperature overnight. The crude product was purified by reversed-phase HPLC (15% to 80% CH3CN in water with 0.1% TFA) which afforded the TFA salt. CH3CN was evaporated, a saturated solution of sodium bicarbonate was added, and solids that precipitated were filtered. EtOAc was added along with some hexanes and the contents were sonicated. Solids that precipitated were filtered and dried to afford the title compound as an off-white solid (47.5 mg, 56%). lU NMR (400 MHz, DMSO-4) δ ppm 11.46 (br. s., 1 H) 8.48 (br. s., 1 H) 8.18 (s, 1 H) 7.32 - 7.37 (m, 1 H) 7.10 (s, 1 H) 5.93 (s, 1 H) 4.95 (dt, J=13.14, 6.57 Hz, 1 H) 4.38 (br. s., 1 H) 4.37 (br. s., 1 H) 3.74 - 3.81 (m, 4 H) 3.20 - 3.25 (m, 4 H) 2.53 - 2.60 (m, 2 H) 2.14 (s, 3 H) 1.45 (s, 3 H) 1.43 (s, 3 H) 1.09 (t, J=7.45 Hz, 3 H); LC-MS (ES) m/z = 437.9
Example 40
N-((4,6-dimethyl-2-oxo-l,2-dihydropyridin-3-yl)methyl)-l-isopropyl-6-morpholino-lH-indazole- 4-carboxamide
Figure imgf000072_0001
The title compound was prepared in the same manner as described for example 39 from 1-(1- methylethyl)-6-(4-mo holinyl)-lH-indazole-4-carboxylic acid (55 mg, 0.19 mmol) and 3- (aminomethyl)-4,6-dimethyl-2(lH)-pyridinone.HCl (53.8 mg, 0.29 mmol) The product was collected as an off-white solid (48 mg, 57%). H NMR (400 MHz, DMSO-J6) δ ppm 1 1.44 (br. s., 1 H) 8.49 (br. s., 1 H) 8.18 (s, 1 H) 7.35 (d, J=1.77 Hz, 1 H) 7.10 (s, 1 H) 5.88 (s, 1 H) 4.95 (quin, J=6.57 Hz, 1 H) 4.36 (br. s., 1 H) 4.34 (br. s., 1 H) 3.75 - 3.80 (m, 4 H) 3.21 - 3.25 (m, 4 H) 2.20 (s, 3 H) 2.12 (s, 3 H) 1.44 (s, 3 H) 1.43 (s, 3 H); LC-MS (ES) m/z = 423.9
Example 41
N-((4-cyclohexy]-6-methyl-2-oxo-l,2-dihydropyridin-3-yl)methyl)-6-cyclopropyl-l-isopropyl- lH-indazole-4-carboxamide
Figure imgf000072_0002
a) 6-bromo-N-((4-cyclohexyl-6-methyl-2-oxo-l,2-dihydropyridin-3-yl)methyl)-l-isopropyl-lH- indazole-4-carboxamide
Figure imgf000072_0003
6-bromo-l-(l-methylethyl)-lH-indazole-4-carboxylic acid (237 mg, 0.84 mmol), 3-(aminomethyl)-4- cyclohexyl-6-methyl-2(lH)-pyridinone'TFA (378 mg, 1.13 mmol) and 1 -hydroxy-7-azabenzotriazole (171 mg, 1.26 mmol) were stirred in 10 mL of DMSO for 10 min under nitrogen. N- methylmorpholine (0.37 ml, 3.35 mmol) was added along with EDC (241 mg, 1.26 mmol) and the mixture was stirred at rt overnight. An additional 0.2 eq of pyrdinone, HOAt, NMM and EDC were added and the contents were stirred at rt overnight. The reaction mixture was then poured onto ice- water . A solution of 10% K2C03 in water was added to afford a pH ~ 8-9 solution. The reaction mixture was stirred at rt for 30 min and then allowed to stand at rt for 30 min. Precipitated solids were filtered and air-dried. The collected solid was treated with EtOAc (not soluble) and hexanes, and then concentrated in vacuo. To the isolated solid was then added DMF followed by heating and sonication. Water was added and beige solids crashed out. The precipitated solids were filtered, washed with water, air-dried, and dried in vaccum oven for 2 hours. The title compound was collected as a solid (308 mg, 74%). !H NMR (400 MHz, DMSO-< 6) δ ppm 1 1.53 (s, 1 H) 8.63 (t, .7=4.80 Hz, 1 H) 8.37 (s, 1 H) 8.20 (s, 1 H) 7.68 (d, .7=1.26 Hz, 1 H) 6.01 (s, 1 H) 5.05 (dt, .7=13.14, 6.57 Hz, 1 H) 4.43 (d, J=4.80 Hz, 2 H) 2.84 (t, J=\ 1.24 Hz, 1 H) 2.15 (s, 3 H) 1.70 (d, .7=13.39 Hz, 2 H) 1.60 (d, J=12.13 Hz, 3 H) 1.47 (s, 3 H) 1.45 (s, 3 H) 1.42 (br. s., 1 H) 1.17 - 1.39 (m, 4 H); LCMS:485.2/487.2 (Br pattern).
b) N-((4-cyclohexyl-6-methyl-2-oxo-l,2-dihydropyridin-3-yl)methyl)-6-cyclopropyl-l-isopropyl- lH-indazole-4-carboxamide
In a 25 mL sealable tube under nitrogen were combined 6-bromo-N-[(4-cyclohexyl-6-methyl-2-oxo- l,2-dihydro-3-pyridinyl)methyl]- l -(l -methylethyl)- lH-indazole-4-carboxamide (98 mg, 0.2 mmol), 2-cyclopropyl-4,4,5,5-tetramethyl-l,3,2-dioxaborolane (50.9 mg, 0.3 mmol) in dioxane/water (3 mL: lmL). PdCl2(dppf)-CH2Cl2 adduct (8.24 mg, 0.01 1 mmol) was added and the resulting mixture was degassed with nitrogen for 10 min. Sodium bicarbonate (50.9 mg, 0.61 mmol) was added, the vessel was sealed, and the insoluble mixture was heated in a microwave at 1 10 °C for 20 min, then at 120 °C for 15 min and then at 130 °C for 15 min. Upon cooling down, water was added, and solids that crashed out were filtered. DCM MeOH (1 : 1) was added, the contents pre-absorbed on silica gel, and purified by Si02 chromatography (eluent: gradient 0 to 80:20:2 DCM/MeOH/NH4OH). The collected product was further purified by reverse-phase HPLC (15% to 80% CH^CN in water with
0.1% TFA) which afforded the TFA salt. CH^CN was evaporated, a saturated solution of sodium bicarbonate was added to the water layer and it was further concentrated. Upon sitting for 2 weeks, solids that precipitated were filtered and dried to afford the title compound as a light orange solid (17 mg, 18%). ¾ NMR (400 MHz, OMSO-d6) δ ppm 1 1.48 (br. s., 1 H) 8.46 (br. s., 1 H) 8.25 (s, 1 H) 7.57 (s, 1 H) 7.16 (s, 1 H) 6.01 (s, 1 H) 4.93 - 5.04 (m, 1 H) 4.45 (br. s., 1 H) 4.43 (br. s., 1 H) 2.84 (t, .7=1 1.49 Hz, 1 H) 2.15 (s, 3 H) 2.03 - 2.10 (m, 1 H) 1.60 (br. s., 5 H) 1.47 (s, 3 H) 1.45 (s, 3 H) 1.16 - 1.42 (m, 5 H) 0.96 - 1.02 (m, 2 H) 0.80 - 0.85 (m, 2 H). LC-MS (ES) m/z = 447
Example 42
6-bromo-l-ethyl-lH-indazole-4-carboxylic acid (6-methyl-2-oxo-4-propyl-l, 2-dihydro-pyridin- 3-ylmethyl)-amide
Figure imgf000074_0001
a) 6-bromo-l-ethyl-lH-indazole-4-carboxylic acid methyl ester and methyl 6-bromo-2-ethyl-2 - indazole-4-carboxylate
Figure imgf000074_0002
To a stirred suspension of 6-bromo-lH-indazole-4-carboxylic acid methyl ester (2.5 g, 9.8 mmol) and K2C03 (2 g,14.7 mmol) in DMF (50 mL) was added 1 -bromo ethane (1.28 g,l 1.76 mmol) at RT. The contents were stirred for 2 h at 50 °C. The reaction mixture was then diluted with water (100 mL) and extracted with ethyl acetate (3x50 mL). The combined organic layers were washed with water (3x30 mL), brine solution (2x30 mL), dried over anhydrous sodium sulphate, and filtered. The crude product was purified by silica gel chromatography (eluent: 0 to 30% EtOAc in petroleum ether) to afford the title compound 6-bromo-l-ethyl-lH-indazole-4-carboxylic acid methyl ester (700 mg, 25%) and the undesired isomer 6-bromo-2-ethyl-2H-indazole-4-carboxylic acid methyl ester (500 mg, 18%) as white solids. ¾ NMR (400 MHz, DMSO-d6) (1 -ethyl isomer) : δ 1.41-1.37 (t, 3H), 3.95 (s, 3H), 4.51-4.49 (m, 2H), 7.85-7.84 (d, J = 1.2 Hz, 1H), 8.41-8.39 (d, J = 10 Hz, 1H). LCMS (ES+): m/z=283 [M+l], 285.02 [M+2]. b) 6-bromo-l-ethyl-lH-indazole-4-carboxylic acid (6-methyl-2-oxo-4-propyl-l, 2-dihydro- pyridin-3-ylmethyl)-amide
Step 1: To a stirred solution of 6-bromo-l -ethyl- lH-indazole-4-carboxylic acid methyl ester (700 mg,
2.47 mmol) in THF (35 mL) was added a solution of LiOH H20 (312 mg, 7.42 mmol) in water (15 mL) and the mixture was stirred at RT for 4 h. The reaction mixture was concentrated under reduced pressure, diluted with water (30 mL) and washed with EtOAc (2x25 mL). The aqueous layer was acidified (pH ~ 5) with IN HC1. The precipitated solid was collected by filtration and dried to furnish 6-bromo- l -ethyl- lH-indazole-4-carboxylic acid as a white solid (450 mg, 68 %). Step 2: To a stirred suspension of 6-bromo-l-ethyl-lH-indazole-4-carboxylic acid, 4 (450 mg, 1.67 mmol) in DCM (50 mL) were added EDC.HC1 (384 mg, 2.00 mmol), HOBt.H20 (306 mg, 2.00 mmol) and then stirred for 15 min at RT. To the resulting mixture, DIPEA (1.2 mL, 5.59 mmol) followed by 3-aminomethyl-6-methyl-4-propyl-lH-pyridin-2-one (301 mg, 1.67 mmol) were added and the contents stirred for 18 h at RT. The reaction mixture was diluted with DCM (50 mL) and washed with water (2x50 mL), 10% aq citric acid solution (2x50 mL), saturated aq NaHCCh solution (2x30 mL) and brine (2x50 mL). The organic layer was dried over Na SO/i, filtered, and concentrated under reduced pressure. The obtained solid was washed with diethyl ether (2 x 50 mL) to afford the title compound as a white solid (230 mg, 32 %). XH NMR (400 MHz, DMSO-d6): δ 0.83-0.79 δ (t, 3H), 1.38 (t, 3H), 1.55 (m, 2H), 2.15 (s, 3H), 2.49 (s, 2H), 4.38 (d, 2H), 4.43 (m, 2H), 5.90 (s, 1H), 7.64 (d, 1H), 8.19 (s, 1H), 8.38 (s, 1H), 8.61 (m, 1H), 1 1.51 (bs, 1H). LCMS (ES+): m/z = 431.15.
Example 43
6-bromo-N-((6-methyl-2-oxo-4-propyl-l,2-dihydropyridin-3-yl)methyl)-l-propyl-lH-indazole-4- carboxamide
Figure imgf000075_0001
a) 6-Bromo-l-propyl-lH-indazole-4-carboxylic acid methyl ester
Figure imgf000075_0002
The title compounds were prepared in the same manner as described for example 42 (step a) from 6- bromo-lH-indazole-4-carboxylic acid methyl ester (3.0 g, 11.66 mmol) and 1-propyl bromide (1.59 g, 12.94 mmol) wherein the contents were heated at at 40 °C for 30 min. The obtained crude products were purified by silica gel chromatography (eluent: 10 % ethyl acetate in petroleum ether) to afford the title compounds 6-bromo-l -propyl- lH-indazole-4-carboxylic acid methyl ester (700 mg, 20 %) and undesired isomer 6-bromo-2-propyl-2H-indazole-4-carboxylic acid methyl ester (700 mg, 20 %) as white solids. !H NMR (400 MHz, DMSO-d6): ( l-propyl isomer): δ 0.83-0.80 δ (t, J=7.6, 3H), 1.84-1.81 (t, J=14.4 Hz, 2H), 3.95-3.94 (d, J=5.2 Hz 3H), 4.45-4.43 (t, 2H), 7.84 (d, J=1.2 Hz, 1H), 8.43 (d, J= 14 Hz, 2H). LCMS (ES+): m/z= 297.06. b) 6-bromo-l-propyl-lH-indazole-4-carboxylic acid
Figure imgf000076_0001
The title compound was prepared in the same manner as described for example 42 (step b, part 1) from 6-bromo-l -propyl- lH-indazole-4-carboxylic acid, 7 (700 mg, 2.35 mmol ) and LiOH.H20 (290 mg, 7.07 mmol). The product was collected as an off-white solid (600 mg, 90 %). l NMR (400 MHz, DMSO-dg): δ 0.83-0.80 δ (t, 3H), 1.86- 1.81 (m, 2H), 4.44-4.40 (t, 2H), 7.81 (d, J=1.2 Hz, 1H), 8.38 (d, J= 4.4 Hz, 2H). LCMS (ES-): m/z= 281.14 [M-2H]. c) 6-bromo-N-((6-methyl-2-oxo-4-propyl-l,2-dihydropyridin-3-yl)methyl)-l-propyl-lH- indazole-4-carboxamide
The title compound was prepared in the same manner as described for example 42 (step b, part 2) from 6-bromo-l -propyl- lH-indazole-4-carboxy lie acid (300 mg, 1.06 mmol) and 3-aminomethyl-6- methyl-4-propyl-lH-pyridin-2-one (190 mg, 1.06 mmol). The crude product was triturated with diethyl ether (10 mL) and n-pentane (10 mL) to afford the title compound as a white solid ( 200 mg,
42.5 %). H NMR (400 MHz, DMSO-d6): δ 0.88 (t, 3H), 0.90 (t, 3H), 1.48-1.46 (t, 2H), 1.86-1.80 (m, 2H), 2.13 (s, 3H), 2.50-2.32 (t, 2H), 4.40-4.35 (m, 4H), 5.90 (s, 1H), 7.69 (s, 1H), 8.20 (s, 1H), 8.35 (s, 1H), 8.62 (s, 1H), 11.52 (s, 1H). LCMS (ES+): m/z= 445.17.
Example 44
6-bromo-l-cyclopentyl-N-((4,6-dimethyl-2-oxo-l,2-dihydropyridin-3-yl)methyl)-lH-indazole-4- carboxamide
Figure imgf000077_0001
a) methyl 6-bromo-l-cyclopentyl-lH-indazole-4-carboxylate
Figure imgf000077_0002
Ice-cooled methyl 6-bromo-lH-indazole-4-carboxylate (2 g, 7.84 mmol) in 30 mL of DMF was treated with NaH (60%, 345 mg, 8.63 mmol) and the mixture was stirred for 1 hr at 0 °C. Iodocyclopentane (2.31 g, 1 1.8 mmol) was then added and the mixture was stirred at 100 °C overnight. After cooling to RT, the reaction mixture was partitioned between water and ethyl acetate. The organic phase was washed with water and brine, dried over MgSO^, filtered and evaporated. Hexanes was added to the brown oil and it was purified using silica gel chromatography (eluent: Hex/EtOAc , gradient 0 to 25%). The less polar product was evaporated to give an orange oil, and was dried on hivac overnight. The product was confirmed to be the alkylated 1 -isomer as suggested by 2D HNMR, and was collected as 807 mg (32%). !H NMR (400 MHz, DMSO-d6): δ 8.40 (s, 1 H) 8.37 (s, 1 H) 7.81 (d, J=1.52 Hz, 1 H) 5.26 (quin, J=7.07 Hz, 1 H) 3.95 (s, 3 H) 2.08 - 2.17 (m, 2 H) 1.93 - 2.01 (m, 2 H) 1.82 - 1.92 (m, 2 H) 1.64 - 1.73 (m, 2 H); LCMS (ES+): m/z= 323.3/325.3 b) 6-bromo-l-cyclopentyl-N-((4,6-dimethyl-2-oxo-l,2-dihydropyridin-3-yl)methyl)-lH-indazole- 4-carboxamide Step 1 : Methyl 6-bromo-l-cyclopentyl-lH-indazole-4-carboxylate (1.5 g, 4.64 mmol) was suspended in Methanol (8 mL) and Tetrahydrofuran (THF) (16 mL) followed by addition of 3N Sodium Hydroxide (3.09 mL, 9.28 mmol). The solution was heated to 55 °C with stirring overnight (16h). The organic solvents were removed in vacuo and the residue was diluted with water (20 mL) and stirred in an ice bath. To the chilled aqueous solution was added IN HC1, dropwise, until precipitation stopped. The suspension was stirred in the ice bath for 20 min and then filtered. The solid cake was washed with water, dried, and used directly in step 2. Step 2: 6-bromo-l-cyclopentyl-lH-indazole-4-carboxylic acid (1.44 g, 4.61 mmol), 3-(aminomethyl)- 4,6-dimethyl-2(lH)-pyridinone hydrochloride (1.138 g, 6.03 mmol), N-[3-(dimethylamino)propyl]- N'-ethylcarbodiimide hydrochloride (1.335 g, 6.96 mmol), and 3H-[l,2,3]triazolo[4,5-b]pyridin-3-ol hydrate (1.073 g, 6.96 mmol) were suspended in DMSO (8.00 mL), followed by N-methylmorpholine (2.82 g, 27.8 mmol). The reaction mixture was stirred at room temperature overnight to afford a slurry. The slurry was diluted with DMSO (10 mL) and added dropwise to a ice-chilled aqueous solution of IN Na2CC>3 (50 ml) and water (200 mL), and stirred rapidly for 30 min. The precipitate was collected by vacuum filtration and washed with water. The cake was dried in the vacuum oven at 60 °C overnight (16h). The title compound was obtained as a pale yellow-white solid (2.05 g, 98 % yield). lB NMR (400 MHz, DMSO-56) δ 11.50 (br. s., 1H), 8.64 (br. s., 1H), 8.37 (s, 1H), 8.20 (s,
1H), 7.70 (d, J= 1.52 Hz, 1H), 5.89 (s, 1H), 5.22 (quin, J = 7.01 Hz, 1H), 4.34 (d, J = 4.55 Hz, 2H), 2.20 (s, 3H), 2.13 (s, 3H), 2.06 - 2.11 (m, 2H), 1.91 - 2.03 (m, 2H), 1.80 - 1.91 (m, 2H), 1.61 - 1.75 (m, 2H). LC-MS(ES) [M+H]+ 443.0.
Example 45
6-bromo-l-cyclopenryl-N-((4-ethyl-6-methy]-2-oxo-l,2-dihytlropyridin-3-y])methyl)
indazole-4-carboxamide
Figure imgf000078_0001
In a 25 mL sealable tube under nitrogen were combined 6-bromo-l-cyclopentyl-lH-indazole-4- carboxylic acid (370 mg, 1.2 mmol) and 3-(aminomethyl)-4-ethyl-6-methyl-2(lH)-pyridinone.HCl (340 mg, 1.68 mmol) in DMSO (3 mL). 1 -hydroxy-7-azabenzotriazole (277 mg, 2.04 mmol) was added and the resulting mixture was degassed with nitrogen for 10 minutes. N-methylmorpholine (0.55 ml, 5 mmol) and EDC (390 mg, 2 mmol) were added, the vessel was sealed, and the mixture was stirred at room temperature overnight. The mixture was poured onto 10 mL of ice-water and solids crashed out. 10% K2C03 was added to adjust the pH~8-9. The mixture was stirred for 4 hours and then allowed to stand for another 2 hours. Solids were filtered and air-dried. DMF along with some water was then added. Solids that precipitated were filtered and dried to afford the title compound as a light orange solid (515 mg, 91%). H NMR (400 MHz, DMSO-<¾) δ ppm 1 1.54 (s, 1 H) 8.63 (t, J=4.80 Hz, 1 H) 8.36 (s, 1 H) 8.20 (s, 1 H) 7.70 (d, J=1.52 Hz, 1 H) 5.93 (s, 1 H) 5.23 (t, .7=7.07 Hz, 1 H) 4.36 (s, 1 H) 4.35 (s, 1 H) 2.53 - 2.59 (m, 2 H) 2.14 (s, 3 H) 2.07 - 2.13 (m, 2 H) 1.93 - 2.01 (m, 2 H) 1.83 - 1.91 (m, 2 H) 1.65 - 1.73 (m, 2 H) 1.10 (t, J=7.58 Hz, 3 H); LC-MS (ES) m/z = 456.9/459.1
Example 46
l-cyclopent l-N-((4,6-dimethyl-2-oxo-l,2-dihydropyridin-3-yl)methyl)-6-(2-(4-methy]piperazin- l-yl)pyrimidin-5-yl)-l
Figure imgf000079_0001
To a 2 mL microwave vial were added 6-bromo- l-cyclopentyl-N-[(4,6-dimethyl-2-oxo-l,2-dihydro- 3-pyridinyl)methyl]-lH-indazole-4-carboxamide (150 mg, 0.338 mmol), 2-(4-methyl- 1 -piperazinyl)- 5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrimidine (134 mg, 0.440 mmol), Na2C03 (108 mg, 1.015 mmol), PdCl2(dppf)-CH2Cl2 adduct (27.6 mg, 0.034 mmol), 1 ,2-Dimethoxy ethane (1269 μΐ) and water (423 μΐ). The contents were irradiated in a microwave reactor at 150 °C for 30 min. The reaction solution was filtered through a Whatman 0.45μ1 Teflon syringless filter device and diluted with DMSO (6 mL). The DMSO solution was purified by HPLC reverse phase chromatography (phenomenex Gemini-NX, 30x100 5μ column, 5-30% acetonitrile/water 0.01% formic acid, 8 min gradient). The fractions containing the desired product were concentrated to dryness using a Genovac HT-4 instrument at 40 °C. The title compound was obtained as a pale cream colored foam (28 mg, 14.83 % yield). ¾ NMR (400 MHz, CHLOROFORM-δ) δ 8.62 (s, 2H), 8.39 (s, 1H), 7.99 (t, J = 5.56 Hz, 1H), 7.71 (s, 1H), 7.57 (s, 1H), 5.98 (s, 1H), 5.03 (quin, J = 7.14 Hz, 1H), 4.66 (d, J = 5.81 Hz, 2H), 3.97 (br. s., 4H), 2.64 (br. s., 4H), 2.44 (s, 6H), 2.12 - 2.25 (m, 7H), 1.89 - 2.07 (m, 2H), 1.67 - 1.85 (m, 2H). LCMS(ES) [M+H]+ 541.5. Examples 47-52 were prepared in a similar manner as described above using 6-bromo- 1 -cyclopentyl- N-[(4,6-dimethyl-2-oxo-l,2-dihydro-3-pyridinyl)methyl]-lH-indazole-4-carboxamide and the appropriate boronic acid reagent.
Example 47
l-cyclopentyl-N-((4,6-dimethyl-2-oxo-l,2-dihydropyridin-3-yl)methyl)-6-(3-(pyrrolidin-l- yl)phenyl)-lH-indazole-4-carboxamide
Figure imgf000080_0001
H NMR (400 MHz, MeOD) δ 8.35 (s, 1H), 7.93 (s, 1H), 7.81 (d, J = 1.26 Hz, 1H), 7.29 (t, J = 7.96 Hz, 1H), 6.99 (d, J = 8.08 Hz, 1H), 6.91 (t, J = 2.02 Hz, 1H), 6.63 (dd, J = 1.77, 8.08 Hz, 1H), 6.14 (s, 1H), 5.27 (quin, J = 7.26 Hz, 1H), 4.60 (s, 2H), 3.36 - 3.44 (m, 4H), 2.45 (s, 3H), 2.27 (s, 8H), 2.07 (dt, J = 3.38, 6.63 Hz, 3H), 1.90 - 2.05 (m, 2H), 1.74 - 1.88 (m, 2H). LCMS(ES) [M+H]+ 510.1.
Example 48
l-cyclopentyl-N-((4,6-dimethyl-2-oxo-l,2-dihydropyridin-3-yl)methyl)-6-(4-methylthiophen-2- yl)-lH-indazole-4-carboxamide
Figure imgf000080_0002
H NMR (400 MHz, DMSO-86) δ 1 1.55 (s, 1H), 8.64 (t, J = 4.93 Hz, 1H), 8.32 (s, 1H), 8.05 (s, 1H), 7.80 (d, J = 1.26 Hz, 1H), 7.55 (d, J = 1.26 Hz, 1H), 7.19 (t, J = 1.26 Hz, 1H), 5.90 (s, 1H), 5.30 (s, 1H), 4.38 (d, J = 4.80 Hz, 2H), 2.28 (s, 3H), 2.23 (s, 3H), 2.13 (s, 5H), 1.95 - 2.06 (m, J = 6.32 Hz, 2H), 1.82 - 1.94 (m, 2H), 1.64 - 1.77 (m, J = 4.55 Hz, 2H). LCMS(ES) [M+H]+ 461.1. Example 49
l-cyclopentyl-N-((4,6-dimethyl-2-oxo-l,2-dihydropyridin-3-yl)methyl)-6-(2-(piperazin-l- yl)pyrimidin-5-yl)-lH-indazole-4-carboxamide
Figure imgf000081_0001
H NMR (400 MHz, MeOD) δ 8.97 (s, 2H), 8.43 (s, 1H), 8.15 (s, 1H), 8.02 (s, 1H), 6.99 (br. s., 1H), 5.31 (qd, J = 6.69, 6.86 Hz, 1H), 4.72 (br. s., 2H), 4.21 (br. s., 4H), 3.38 (br. s., 4H), 2.69 (s, 3H), 2.53 (s, 3H), 2.20 - 2.32 (m, 2H), 2.07 - 2.20 (m, 2H), 2.03 (d, J = 18.19 Hz, 2H), 1.73 - 1.90 (m, 2H). LCMS(ES) [M+H]+ 527.2.
Example 50
l-cyclopentyl-N-((4,6-dimethyl-2-oxo-l,2-dihydropyridin-3-yl)methyl)-6-(3-(3- hydroxypropyl)pheny
Figure imgf000081_0002
H NMR (400 MHz, MeOD) δ 8.37 (s, 1H), 7.97 (s, 1H), 7.83 (d, J = 1.26 Hz, 1H), 7.64 (s, 1H), 7.60 (d, J = 7.83 Hz, 1H), 7.41 (t, J = 7.58 Hz, 1H), 7.27 (d, J = 7.58 Hz, 1H), 6.15 (s, 1H), 5.28 (quin, J = 7.26 Hz, 1H), 4.57 - 4.62 (m, 2H), 3.63 (t, J = 6.44 Hz, 2H), 2.77 - 2.85 (m, 2H), 2.45 (s, 3H), 2.27 (s, 3H), 2.19 - 2.26 (m, 2H), 2.09 - 2.20 (m, 2H), 1.87 - 2.07 (m, 4H), 1.74 - 1.87 (m, 2H). LCMS(ES) [M+H]+ 499.5.
Example 51
l-cyclopentyl-N-((4,6-dimethyl-2-oxo-l,2-dihydropyridin-3-yl)methyl)-6-(3- (trifluoromethoxy)phenyl)-lH-indazole-4-carboxamide
Figure imgf000082_0001
H NMR (400 MHz, MeOD) δ 8.39 (s, 1H), 8.04 (s, 1H), 7.79 - 7.85 (m, 2H), 7.73 (s, 1H), 7.61 (t, J = 7.96 Hz, 1H), 7.34 (dt, J = 1.17, 8.27 Hz, 1H), 6.15 (s, 1H), 5.26 - 5.35 (m, 1H), 4.60 (s, 2H), 2.45 (s, 3H), 2.27 (s, 3H), 2.24 (br. s., 2H), 2.09 - 2.20 (m, 2H), 1.94 - 2.07 (m, 2H), 1.81 (d, J = 4.55 Hz, 2H). LCMS(ES) [M+H]+ 525.2.
Example 52
l-cyclopentyl-N-((4,6-dimethyl-2-oxo-l,2-dihydropyridin-3-y])methyl)-6-(6-methoxypyridin-3- yl)-lH-indazole-4-carboxamide
Figure imgf000082_0002
!H NMR (400 MHz, DMSO-d6) δ 1 1.54 (s, 1H), 8.69 (d, J = 1.77 Hz, 1H), 8.61 (t, J = 4.93 Hz, 1H), 8.38 (s, 1H), 8.22 (dd, J = 2.65, 8.72 Hz, 1H), 8.16 (s, 1H), 7.87 (d, J = 1.26 Hz, 1H), 6.97 (d, J = 8.59 Hz, 1H), 5.89 (s, 1H), 5.32 (quin, J = 7.14 Hz, 1H), 4.39 (d, J = 4.80 Hz, 2H), 3.92 (s, 3H), 2.22 (s, 3H), 2.13 (s, 5H), 1.99 (s, 2H), 1.84 - 1.95 (m, 2H), 1.65 - 1.78 (m, 2H). LCMS(ES) [M+H]+ 472.4.
Example 53
l-cyclopentyl-N-((4,6-dimethyl-2-oxo-l,2-dihydropyridin-3-yl)methyl)-6-(4-fluorophenyl)-lH- indazole-4-carboxamide
Figure imgf000083_0001
In a 25 mL sealable tube under nitrogen were combined 6-bromo- l-cyclopentyl-N-[(4,6-dimethyl-2- oxo-l,2-dihydro-3-pyridinyl)methyl]-lH-indazole-4-carboxamide (100 mg, 0.23 mmol) and (4- fluorophenyl)boronic acid (47.3 mg, 0.34 mmol) in dioxane/water (3 mL: lmL). PdCl2(dppf)-CH2Cl2 adduct (9.2 mg, 0.011 mmol) was added and the resulting mixture was degassed with nitrogen for 10 min. Sodium bicarbonate (56.8 mg, 0.68 mmol) was added, the vessel was sealed, and the insoluble mixture was heated in a microwave at 100 °C for 20 min. Upon cooling down, water was added, and solids that crashed out were filtered. DCM/MeOH (1 :1) was added, the contents pre-absorbed on silica gel and purified by Si02 chromatography (eluent: gradient 0 to 80:20:2 DCM/MeOH/NEL^OH).
The collected product was suspended in EtOAc along with some hexanes. The mixture was sonicated, and the solids that precipitated were filtered and dried to afford the title compound as a light grey solid (78 mg, 75%). !H NMR (400 MHz, DMSO-c 6) δ ppm 11.54 (s, 1 H) 8.64 (t, J=4.80 Hz, 1 H) 8.37 (s, 1 H) 8.13 (s, 1 H) 7.89 - 7.94 (m, 2 H) 7.85 (s, 1 H) 7.35 (t, .7=8.97 Hz, 2 H) 5.89 (s, 1 H) 5.34 (quin, .7=7.14 Hz, 1 H) 4.40 (br. s., 1 H) 4.38 (br. s., 1 H) 2.22 (s, 3 H) 2.1 1 - 2.18 (m, 5 H) 1.98 - 2.06 (m, 2 H) 1.86 - 1.94 (m, 2 H) 1.66 - 1.75 (m, 2 H). LC-MS (ES) m/z = 459.1
Example 54
l-cyclopentyl-N-[(4,6-dimethyl-2-oxo-l,2-dihydro-3-pyridinyl)methyl]-6-(l-methyl-lH-pyrazol- 4-yl)-lH-indazole-4-carboxamide
Figure imgf000083_0002
The title compound was prepared in a similar manner as described for example 8 from 6-bromo-l- cyclopentyl-N-[(4,6-dimethyl-2-oxo-l,2-dihydro-3-pyridinyl)methyl]-lH-indazole-4-carboxamide (100 mg, 0.226 mmol) and l-methyl-4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-lH-pyrazole (70 mg, 0.338 mmol). The product was collected as a white solid (82 mg, 81%). lU NMR (400 MHz, DMSO-6?6) δ ppm 1 1.53 (br. s., 1 H) 8.48 (t, J=4.93 Hz, 1 H) 8.30 (s, 1 H) 8.26 (s, 1 H) 8.04 (s, 2 H) 7.81 (s, 1 H) 5.90 (s, 1 H) 5.22 (quin, J=7.14 Hz, 1 H) 4.39 (s, 1 H) 4.38 (s, 1 H) 3.87 - 3.92 (m, 3 H) 2.23 (s, 3 H) 2.14 - 2.19 (m, 1 H) 2.13 (s, 4 H) 1.96 - 2.05 (m, 2 H) 1.84 - 1.94 (m, 2 H) 1.66 - 1.76 (m, 2 H). LC-MS (ES) m z = 445.2 [M+H]+
Example 55
l-cyclopent l-N-((6-methyl-2-oxo-4-propyl-l,2-dihydropyridin-3-yl)methyl)-6-(6-(4- methylpiperazin-l-yl)pyridin-3-yl)-lH-indazole-4-carboxamide
Figure imgf000084_0001
In a 25 mL sealable tube under nitrogen were combined 6-bromo-l-cyclopentyl-N-[(6-methyl-2-oxo- 4-propyl-l,2-dihydro-3-pyridinyl)methyl]-lH-indazole-4-carboxamide (100 mg, 0.21 mmol), 1- methyl-4-[5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-2-pyridinyl]piperazine (96 mg, 0.32 mmol) in DME/water (3 mL: lmL). PdCl2(dppf)-CH2Cl2 adduct (8.7 mg, 0.01 mmol) was added and the resulting mixture was degassed with nitrogen for 10 min. Sodium bicarbonate (53.5 mg, 0.64 mmol) was added, the vessel was sealed, and the insoluble mixture was heated in a microwave at 1 10 °C for 20 min. The mixture was evaporated, DCM/MeOH (1 : 1) was added, and the contents pre-absorbed on silica gel and purified by Si02 chromatography (eluent: gradient 0 to 80:20:2 DCM/MeOH/NH4OH).
The collected product was further purified by reversed-phase HPLC (15% to 80% CH^CN in water with 0.1% TFA) which afforded the TFA salt. CH^CN was evaporated and a saturated solution of sodium bicarbonate was added. Solids that precipitated were filtered and dried to afford the title compound as a white solid (87 mg, 71%). *H NMR (400 MHz, DMSO-c/6) δ ppm 1 1.45 (br. s., 1 H) 8.68 (br. s., 1 H) 8.39 (s, 1 H) 8.24 (s, 1 H) 8.21 (d, J=5.30 Hz, 1 H) 7.84 - 7.88 (m, 1 H) 7.20 (s, 1 H) 7.13 (dd, J=5.31, 1.26 Hz, 1 H) 5.91 (s, 1 H) 5.38 (quin, J=7.07 Hz, 1 H) 4.43 (br. s., 1 H) 4.42 (br. s., 1 H) 3.55 - 3.63 (m, 4 H) 2.53 - 2.56 (m, 2 H) 2.41 - 2.46 (m, 4 H) 2.24 (s, 3 H) 2.12 - 2.20 (m, 5 H) 1.97 - 2.06 (m, 2 H) 1.86 - 1.95 (m, 2 H) 1.67 - 1.76 (m, 2 H) 1.48 - 1.57 (m, 2 H) 0.89 (t, J=7.33 Hz, 3 H); LC-MS (ES) m/z = 568.3 Example 56
l-cyclopentyl-N-[(4,6-dimethyl-2-oxo-l,2-dihydro-3-pyridinyl)methyl]-6-[6-(l-piperazinyl)-3 pyridinyl]-lH-indazole-4-carboxamide
Figure imgf000085_0001
The title compound was prepared in a similar manner as described for example 8 from 6-bromo-l- cyclopentyl-N-[(4,6-dimethyl-2-oxo-l,2-dihydro-3-pyridinyl)methyl]-lH-indazole-4-carboxamide (200 mg, 0.451 mmol) and l-[5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-2-pyridinyl]piperazine (196 mg, 0.677 mmol). The product was collected as a white solid (92 mg, 37%). !H NMR (400 MHz, DMSO-J6) δ ppm 11.54 (br. s., 1 H) 8.64 - 8.67 (m, 1 H) 8.60 (t, J=4.80 Hz, 1 H) 8.35 (s, 1 H) 8.03 - 8.09 (m, 2 H) 7.84 (d, J=1.26 Hz, 1 H) 6.89 - 6.99 (m, 1 H) 5.89 (s, 1 H) 5.27 - 5.35 (m, 1 H) 4.39 (br. s., 1 H) 4.38 (br. s., 1 H) 3.41 - 3.56 (m, 4 H) 2.69 - 2.91 (m, 4 H) 2.22 (s, 3 H) 2.07 - 2.20 (m, 6 H) 1.97 - 2.06 (m, 2 H) 1.85 - 1.93 (m, 2 H) 1.65 - 1.74 (m, 2 H). LC-MS (ES) m/z = 526.3 [M+H]+
Example 57
l-cyclopentyl-N-((4,6-dimethyl-2-oxo-l,2-dihydropyridin-3-yl)methyl)-6-(4- ((dimethylamino)methyl)phenyl)-lH-indazole-4-carboxamide
Figure imgf000085_0002
To a sealed tube were added 6-bromo-l-cyclopentyl-N-[(4,6-dimethyl-2-oxo-l,2-dihydro-3- pyridinyl)methyl]-lH-indazole-4-carboxamide (.10g, 0.226 mmol), N,N-dimethyl-l-[4-(4,4,5,5- tetramethyl-l,3,2-dioxaborolan-2-yl)phenyl]methanamine (0.081 g, 0.271 mmol), and potassium phosphate (tribasic) (0.192 g, 0.902 mmol) followed by 1,4-Dioxane (2.0 mL) and water (.5 niL). The suspension was stirred with degassing under N2 for 10 min, followed by addition of PdCl2(dppf)- CH2C12 adduct (0.018 g, 0.023 mmol). The reaction vessel was sealed and stirred with heating at 100 °C (heat block) for 2h. The contents were allowed to stir with cooling to RT overnight. The mixture was diluted with EtOAc followed by addition of silica gel. The mixture was concentrated in vacuo and the obtained solid purified by silica gel chromatography (dry loaded, eluent: 8-95% gradient of chloroform (containing 10% 2M Ammonia in Methanol) and DCM. The collected product was concentrated from DCM/MTBE and then dried in vacuum oven for 16h. The title compound was collected as an off-white solid (45 mg, 39%). lH NMR (400 MHz, DMSO-t 6) δ ppm 1.65 - 1.76 (m, 2 H), 1.83 - 1.94 (m, 2 H), 1.97 - 2.07 (m, 2 H), 2.09 - 2.19 (m, 1 1 H), 2.21 (s, 3 H), 3.44 (s, 2H), 4.39 (d, J=4.80 Hz, 2 H), 5.34 (quin, J=7.14 Hz, 1 H), 5.88 (s, 1 H), 7.41 (m, J=8.34 Hz, 2 H), 7.81 (m, J=8.08 Hz, 2 H), 7.87 (d, J=1.01 Hz, 1 H), 8.12 (s, 1 H), 8.36 (s, 1 H), 8.65 (t, J=4.93 Hz, 1 H), 1 1.53 (s, 1 H); LC-MS (ES) m/z = 498.3 [M+H]+
Example 58
6-chloro-l-cyclopentyl-iV-[(4,6-dimethyl-2-oxo-l,2-dihydro-3-pyridinyl)methyl]-l -indazole-4- carboxamide
Figure imgf000086_0001
a) Methyl 6-chloro-l-cyclopentyl-l /-indazole-4-carboxylate
Figure imgf000086_0002
Methyl 6-chloro-l-cyclopentyl- lH-indazole-4-carboxylate (2.1g, 9.97 mmol) was suspended in DMF
(40 mL), placed into an ice bath, and stirred for 15 min. Next added sodium hydride (0.997 g, 24.93 mmol) slowly over 5 min (gas evolution) and stirred for 15 min. Bromocyclopentane (3.21 mL, 29.9 mmol) was added at once via syringe, and the mixture allowed to stir with warming to RT. After 15 min stirring at RT, the contents were stirred with heating at 45 °C for 16h. The contents were cooled to RT, and then 0.5g sodium carbonate and 1 mL of iodomethane were added. The contents were stirred at RT for 3h, after which time the mixture was poured onto 400 mL of ice/water with stirring. After 5 min stirring, the contents were extracted with ether (2xl00mL). The combined organic layers were concentrated in vacuo. The crude product was purified by silica gel chromatography (eluent : gradient 3-25% EtOAc in hexanes) The first product off the column was determined to be the desired Nl -substituted isomer, and was collected after drying (vacuum pump, lh) as an orange solid (1.1 1 g, 39%). H NMR (400 MHz, DMSO-c 6) δ ppm 1.64 - 1.74 (m, 2 H), 1.82 - 1.93 (m, 2 H), 1.94 - 2.04 (m, 2 H), 2.09 - 2.19 (m, 2 H), 3.96 (s, 3 H), 5.27 (t, J=7.07 Hz, 1 H), 7.72 (d, J=1.77 Hz, 1H), 8.28 (s, 1 H), 8.40 (s, 1 H); LC-MS (ES) m/z = 278.7 [M+H]+ b) 6-chloro-l-cyclopentyl-l//-indazole-4-carboxylic acid
Figure imgf000087_0001
The title compound was prepared in the same manner as described for example 1 (step 2) from methyl 6-chloro- l-cyclopentyl- lH-indazole-4-carboxylate (1.1 lg, 3.98 mmol), wherein the reaction stir time was 12h. The product was collected as 0.99g (85%). lH NMR (400 MHz, DMSO-d6) δ ppm 1.63 - 1.75 (m, 2 H) 1.82 - 1.93 (m, 2 H) 1.93 - 2.05 (m, 2 H) 2.08 - 2.19 (m, 2 H) 5.25 (quin, J=7.07 Hz, 1H) 7.69 (d, J=1.77 Hz, 1 H) 8.20 (s, 1 H) 8.40 (s, 1 H) 1 1.88 - 15.25 (m, 1 H); LC-MS (ES) m/z = 264.9 [M+H]+ c) 6-chloro-l-cyclopentyl-N-[(4,6-dimethyl-2-oxo-l,2-dihydro-3-pyridinyl)methyl]-l//-indazole- 4-carboxaniide
The title compound was prepared in the same manner as described for example 3 (step c) from 6- chloro- l -cyclopentyl- lH-indazole-4-carboxylic acid (.12g, 0.453 mmol) and 3-(aminomethyl)-4,6- dimethyl-2(lH)-pyridinone (0.128 g, 0.680 mmol), wherein the stir time was 12h. The crude product was purified by silica gel chromatography (eluent: 5-85% gradient chloroform (containing 10% 2M Ammonia in methanol) and dichloromethane). The isolated product was concentrated from MTBE to afford an off-white solid that was dried in hi-vac oven for 6h. The final product was collected as 0.165g (89%); JH NMR (400 MHz, DMSO-d6) δ ppm 1.62 - 1.74 (m, 2 H) 1.81 - 1.91 (m, 2 H) 1.91 - 2.02 (m, 2 H) 2.06 - 2.17 (m, 5 H) 2.20 (s, 3 H) 4.34 (d, J=4.80 Hz, 2 H) 5.16 - 5.28 (m, 1 H) 5.88 (s, 1 H) 7.60 (d, J=1.52 Hz, 1 H) 8.05 (s, 1 H) 8.37 (s, 8.61 (t, J=4.93 Hz, 1 H) 1 1.54 (s, 1 H); LC-MS (ES) m/z = 398.8 [M+H]+
Example 59
6-chloro-l-cyclopentyl-7V- [(6-methyl-2-oxo-4-propyl-l ,2-dihydro-3-pyridinyl)methyl] -\H- indazole-4-carboxamide
Figure imgf000088_0001
The title compound was prepared in the same manner as described for example 3 (step c) from 6- chloro-l-cyclopentyl-lH-indazole-4-carboxylic acid (.12g, 0.453 mmol) and 3-(aminomethyl)-6- methyl-4-propyl-2(lH)-pyridinone (0.128 g, 0.680 mmol). The final product was collected as 0.170g (86%); H NMR (400 MHz, DMSO-d6) δ ppm 0.89 (t, J=7.33 Hz, 3 H), 1.45 - 1.57 (m, 2 H), 1.61 - 1.74 (m, 2 H), 1.80 - 1.92 (m, 2 H), 1.92 - 2.03 (m, 2 H), 2.05 - 2.19 (m, 5 H), 4.36 (d, J=5.05 Hz, 2 H), 5.21 (t, J=7.07 Hz, 1 H), 5.91 (s, 1 H), 7.59 (d, J=1.52 Hz, 1 H), 8.06 (s, 1 H), 8.37 (s, 1 H), 8.62 (t, J=4.80 Hz, 1 H), 1 1.55 (s, 1 H); LC-MS (ES) m/z = 426.7 [M+H]+
Example 60
6-cyano-l-cyclopentyl-N-[(4,6-dimethyl-2-oxo-l,2-dihydro-3-pyridinyl)methyl]-l/ -indazole-4- carboxamide
Figure imgf000088_0002
To 20 mL microwave vial containing a mixture of 6-bromo-l-cyclopentyl-N-[(4,6-dimethyl-2-oxo- l,2-dihydro-3-pyridinyl)methyl]-lH-indazole-4-carboxamide (0.25g, 0.564 mmol), zinc cyanide (0.066 g, 0.564 mmol), zinc (7.37 mg, 0.113 mmol), and dppf (0.063 g, 0.113 mmol) was added Ν,Ν-Dimethylacetamide (DMA) (5.0 mL). The suspension was stirred with degassing under nitrogen for 5 min. Next added PdC^dppfj-CTLC . adduct (0.046 g, 0.056 mmol). The reaction contents were sealed and stirred with heating at 135 °C (heat block) for 16 h. The reaction mixture was allowed to cool to RT, diluted with 80 mL of water, and adjusted to pH~8 with sat NaHC03. The contents were stirred for 30 min, filtered, and the filter cake was washed with water. The collected solid was dried under hi vacuum for 4 h. The crude product was dissolved in DCM/MeOH followed by addition of silica gel, and concentrated in vacuo to dryness. The contents were purified by silica gel chromatograrphy (dry loaded; eluent: 2-65% gradient of 10% methanol in dichloromethane and dichloromethane). The product was concentrated from MTBE and dried in hi-vac oven for 6 h at 45
°C to afford the title compound as 0.190 g (85 %) . Ή NMR (400 MHz, DMSO-d6) δ ppm 1.64 - 1.75 (m, 2 H) 1.83 - 1.93 (m, 2 H) 1.94 - 2.06 (m, 2 H) 2.10 - 2.19 (m, 5 H) 2.21 (s, 3 H) 4.35 (d, J=4.80 Hz, 2 H) 5.31 (t, J=7.07 Hz, 1 H) 5.89 (s, 1 H) 7.91 (d, J=1.26 Hz, 1 H) 8.51 (s, 1 H) 8.60 (s, 1 H) 8.66 (t, J=4.93 Hz, 1 H) 1 1.54 (s, 1 H) ; LC-MS (ES) m/z = 390.2 [M+H]+
Example 61
6-(aminomethyl)-l-cyclopentyl-/V-[(4,6-dimethyl-2-oxo-l,2-dihydro-3-pyridinyl)methyl]-l/i- indazole-4-carboxamide
Figure imgf000089_0001
6-cyano-l-cyclopentyl-N-[(4,6-dimethyl-2-oxo-l,2-dihydro-3-pyridinyl)methyl]-lH-indazole-4- carboxamide (.10g, 0.257 mmol) was suspended in acetic acid (8 mL). The contents were placed heated at 40 °C under an atmosphere of hydrogen (50 psi; H-cube reactor) at a flow rate of lmL/min with recirculation 2 h. Then contents were diluted with MeOH, concentrated in vacuo, and dried under hi vacuum for 18 h. The crude product was dissolved in DCM and purified by silica gel chromatography (eluent: 10- 100% gradient of chloroform (containing 10% 2M ammonia in methanol) and dichloromethane. The product was concentrated from MTBE and dried in a vacuum oven at 45
°C for 4 h. The title compound was collected as 68 mg (66 %); H NMR (400 MHz, DMSO-d6) δ ppm 1.64 - 1.75 (m, 2 H), 1.82 - 1.93 (m, 2 H), 1.94 - 2.04 (m, 2 H), 2.06 - 2.17 (m, 6 H), 2.21 (s, 3 H), 3.85 (s, 2 H), 4.35 (d, J=5.05 Hz, 2 H), 5.14 (t, J=7.07 Hz, 1 H), 5.89 (s, 1 H), 7.54 (s, 1 H), 7.76 (s, 1 H), 8.27 (s, 1 H), 8.32 (t, J=5.05 Hz, 1 H),
; LC-MS (ES) m/z = 394.0 [M+H]+ Example 62
6-amino-l-cyclopentyl-A^(4,6-dimethyl-2-oxo-l,2-dihydro-3^yri^
carboxamide
Figure imgf000090_0001
To a 20 mL microwave vial containing a mixture of 6-bromo-l-cyclopentyl-N-[(4,6-dimethyl-2-oxo- l,2-dihydro-3-pyridinyl)methyl]-lH-indazole-4-carboxamide (0.25g, 0.564 mmol), benzophenone imine (0.142 mL, 0.846 mmol) and Cs2C03 (0.735 g, 2.256 mmol) was added 1,4-dioxane (5 mL) and the suspension stirred with degassing under N2 for 5 min. Next added Xantphos (0.098 g, 0.169 mmol), degassed for 5 min, and then added Pd2(dba)3 (0.077 g, 0.085 mmol) and degassed for 1 min. The reaction vial was sealed and the mixture heated at 100 °C (heat block) with stirring for 2h. The contents were cooled to RT, diluted with water (80 mL), and extracted with EtOAc (2x). The combined organic layers were dried over MgS04, filtered through Celite, and concentrated in vacuo. The crude solid was dissolved in THF (5 mL) and treated with HC1 (0.564 mL, 1.692 mmol). The volatiles were removed in vacuo. The residue was dried under hi-vacuum, and then dissolved in MeOH/DCM followed by addition of silica gel. The contents were concentrated to dryness and dried under hi-vacuum for lh. The contents were purified by silica gel chromatography (dry loaded, eluent: 3-95% gradient chloroform (containing 10% 2M ammonia in methanol) and DCM). The collected solid was triturated from hot acetontrile, and then placed in freezer for 15 min. The contents were filtered cold and washed with additional acetonitrile. The title compound was collected as 90 mg (41 %); *H NMR (400 MHz, DMSO-d6) δ ppm 1.60 - 1.72 (m, 2 H), 1.78 - 1.89 (m, 2 H), 1.91 - 2.00 (m, 2 H), 2.01 - 2.10 (m, 2 H), 2.12 (s, 3 H), 2.22 (s, 3 H), 4.32 (d, J=5.05 Hz, 2 H), 4.84 (t, J=7.07 Hz, 1 H), 5.41 (s, 2 H), 5.88 (s, 1 H), 6.65 (s, 1 H), 6.88 (d, J=1.52 Hz, 1 H), 7.95 (s, 1 H), 8.14 (t, J=5.18 Hz, 1 H), 11.53 (s, 1 H) ; LC-MS (ES) m/z = 379.2 [M+H]
Example 63
l-cyclopentyl-/V-[(4,6-dimethyl-2-oxo-l,2-dihydro-3-pyridinyl)methyl]-6- [(phenylsulfonyl)amino]-l//-indazole-4-carboxamide
Figure imgf000091_0001
6-amino- 1 -cyclopentyl-N- [(4,6-dimethyl-2-oxo- 1 ,2-dihydro-3-pyridinyl)methyl]- lH-indazole-4- carboxamide (30 mg, 0.079 mmol) was suspended in DCM (3 mL) followed by pyridine (0.032 mL, 0.395 mmol). The contents were vigorously stirred and then benzenesulfonyl chloride (0.01 1 mL, 0.087 mmol) was added and the contents stirred at RT for 1 h. The volatiles were removed in vacuo and dried on hi-vac for lh. The crude product was purified by reverse phase HPLC (Gradient B: 15- 70%. A: Water + .1% TFA. B: CH3CN + .1% TFA). The product was neutralized with water and sat. NaHC03, and extracted with 10% THF/EtOAC (hot) in duplicate. The combined organic layers were dried over MgSC>4, filtered, and concentrated in vacuo. The solid was concentrated from DCM and dried in vacuum oven for 4 h at 45 °C. The title compound was collected as 30 mg (72%); l NMR (400 MHz, DMSO-d6) δ ppm 1.68 (td, J=5.49, 3.41 Hz, 2 H), 1.75 - 1.94 (m, 4 H), 1.99 - 2.10 (m, 2 H), 2.12 (s, 3 H), 2.20 (s, 3 H), 4.30 (d,J=5.05 Hz, 2 H), 4.89 - 5.04 (m, 1 H), 5.89 (s, 1 H), 7.22 (d, J=1.52 Hz, 1 H), 7.36 (s, 1 H), 7.48 - 7.63 (m, 3 H), 7.77 - 7.84 (m, 2 H), 8.11 (s, 1 H), 8.36 (t, J=5.05 Hz, 1 H), 10.56 (s, 1 H), 11.55 (s, 1 H); LC-MS (ES) m/z = 519.2 [M+H]
Example 64
l-cyclopentyl- r-[(4,6-dimethyl-2-oxo-l,2-dihydro-3-pyridinyl)methyl]-6-(3-hydroxy-3-methyl-l- butyn-l-yi)-l/f-indazole-4-carboxamide
Figure imgf000091_0002
To a 10 mL microwave vial containing a mixture of 6-bromo-l-cyclopentyl-N-[(4,6-dimethyl-2-oxo- l,2-dihydro-3-pyridinyl)methyl]-lH-indazole-4-carboxamide (0.12 g, 0.271 mmol), sodium iodide (8.1 1 mg, 0.054 mmol) and zinc (3.54 mg, 0.054 mmol) were added DMSO (2.5 mL), TEA (0.075 mL, 0.541 mmol), and DBU (0.082 mL, 0.541 mmol). The suspension was stirred and degassed with N2 for 5 min. Next added in 2-methyl-3-butyn-2-ol (0.131 mL, 1.353 mmol) and Pd(Ph3P)4 (0.031 g, 0.027 mmol). The reaction vial was sealed and stirred with heating at 85 °C (heat block) for 3h. The mixture was then diluted into water and 20% THF/ EtOAc. The contents were stirred and the layers were separated. The organic layer was washed with brine, dried over MgS04, filtered through Celite, and concentrated in vacuo. The residue was dried on hi-vacuum pump overnight. The crude product was purified by silica gel chromatography (eluent: 5-80% gradient chloroform (containing 10% 2M ammonia in methanol) in DCM). The collected product was purified by reverse phase HPLC ( Gradient B: 15-75 %; A: Water + .1% TFA. B: CH3CN + .1% TFA). The product was suspended in 10% MeOH/CHCl3. Next added 0.5g Silicycle carbonate resin and stirred for 15 min. After standing for 10 min, the contents were filtered through celite and concentrated in vacuo. The product was further concentrated from DCM and MTBE and then dried in hi-vac oven at 45 °C overnight. The title compound was collected as a white solid (38mg, 30%); lB NMR (400 MHz, DMSO-d6) δ ppm 1.47 - 1.52 (m, 6 H), 1.64 - 1.73 (m, 2 H), 1.80 - 1.91 (m, 2 H), 1.92 - 2.02 (m, 2 H), 2.06 - 2.15 (m, 5 H), 2.18 -2.23 (m, 3 H), 4.28 - 4.38 (m, 2 H), 5.25 (quin, J=7.07 Hz, 1 H), 5.52 (s, 1 H), 5.84 - 5.91 (m, 1 H), 7.54 (d, J=1.01 Hz, 1 H), 7.93 (s, 1 H), 8.35 (s, 1 H), 8.60 (t, J=4.93 Hz, 1 H), 11.51 (br. s., 1 H); LC-MS (ES) m/z = 446.9 [M+H]
Example 65
6-bromo-l-cyclopropyl-N-[(4,6-dimethyl-2-oxo-l,2-dihydro-3-pyridinyl)methyl]-l//-indazole-4- carboxamide
Figure imgf000092_0001
a) Methyl 6-bromo-l-cyclopropyl-l/ -indazole-4-carboxylate
Figure imgf000092_0002
Methyl 6-bromo-lH-indazole-4-carboxylate (1.0 g, 3.92 mmol) was dissolved in 1 ,2-Dichloroethane (DCE) (14 mL) and stirred for 15 min. Next added cyclopropylboronic acid (0.674 g, 7.84 mmol) and sodium carbonate (0.831 g, 7.84 mmol). The reaction was stirred at RT (suspension). Copper (II) acetate (0.712 g, 3.92 mmol) and 2,2'-bipyridine (0.612 g, 3.92 mmol) were suspended in DCE (24 mL) with heating and the hot suspension was added to the reaction mixture. The contents were stirred with heating at 70 °C overnight. After cooling to RT, the reaction mixture was poured onto sat. NH4CI and ice. Next added DCM and stirred for 10 min. The contents were filtered through Celite, washing with water and DCM. The layers were separated and the aq. layer was extracted with DCM (lx). The combined organic layers were washed with brine, dried over MgS04, filtered, and concentrated in vacuo. The crude product was purified by silica gel chromatography (3-25% gradient ethyl acetate in hexanes) wherein the less polar product was observed to be the title compound, and was collected as a yellow solid (0.54g, 46%); H NMR (400 MHz, DMSO-d6) δ ppm 1.10 - 1.20 (m, 4 H), 3.81 - 3.90 (m, 1 H), 3.95 (s, 3 H), 7.86 (d, J=1.52 Hz, 1 H), 8.30 (d, J=1.77 Hz, 1 H), 8.32 (d, J=1.01 Hz, 1 H); LC-MS (ES) m/z = 295.1 [M+H] b) 6-bromo-l-cyclopropyl-l//-indazole-4-carboxylate6-bromo-l-cyclopropyl-l/ -indazole-4- carboxylic acid
Figure imgf000093_0001
Methyl 6-bromo- l-cyclopropyl-lH-indazole-4-carboxylate (0.54g, 1.830 mmol) was dissolved in methanol (16 mL) and THF (4 mL) with stirring at RT. A solution of 3N NaOH (1.830 mL, 5.49 mmol) was added the contents were stirred at RT for 2 days. The volatiles were removed in vacuo. The residue was diluted with water and slowly acidifed to pH 3-4 with 1M HCl wherein solids were observed to precipitate. The contents were extracted with EtOAc (2x). The combined organic layers were washed with brine, dried over MgS04, filtered, and concentrated in vacuo to a solid to which was dried under vacuum for lh. The title compound was collected as a white solid (0.48g, 91%); l NMR (400 MHz, DMSO-d6) δ ppm 1.10 - 1.17 (m, 4 H), 3.80 - 3.89 (m, 1 H), 7.84 (d, J=1.77 Hz, 1 H), 8.26 (s, 1 H), 8.31 (s, 1 H), 13.60 (br. s., 1H); LC-MS (ES) m/z = 281.1 [M+H].
c) 6-bromo-l-cyclopropyl-/V-[(4,6-dimethyl-2-oxo-l,2-dihydro-3-pyridinyl)methyl]-l//-indazole- 4-carboxamide
The title compound was prepared in the same manner as described for example 3 (step c) from 6- bromo-l-cyclopropyl- lH-indazole-4-carboxylic acid (.22 g, 0.783 mmol) and 3-(aminomethyl)-4,6- dimethyl-2(lH)-pyridinone (0.221 g, 1.174 mmol), wherein the reaction stir time was 12h. The title compound was collected as a white solid (0.27g, 81%); !H NMR (400 MHz, DMSO-d6) δ ppm 1.06 - 1.17 (m, 4 H), 2.12 (s, 3 H), 2.20 (s, 3 H), 3.76 - 3.85 (m, 1 H), 4.33 (d, J=5.05 Hz, 2 H), 5.88 (s, 1 H), 7.76 (d, J=1.52 Hz, 1 H), 8.10 (s, 1 H), 8.30 (d, J=1.01 Hz, 1 H), 8.64 (t, J=4.93 Hz, 1 H), 1 1.54 (s, 1 H); LC-MS (ES) m z = 414.8 [M+H].
Example 66
6-bromo-l-cyclopropyl-N-[(6-methyl-2-oxo-4-propyl-l,2-dihydro-3-pyridinyl)methyl]-l//- indazole-4-carboxamide
Figure imgf000094_0001
The title compound was prepared in the same manner as described for example 3 (step c) from 6- bromo-l-cyclopropyl-lH-indazole-4-carboxylic acid (.22 g, 0.783 mmol) and 3-(aminomethyl)-6- methyl-4-propyl-2(lH)-pyridinone (0.236 g, 1.089 mmol), wherein the reaction stir time was 12 h. The title compound was collected as a white solid (0.36g, 91%); !H NMR (400 MHz, DMSO-d6) δ ppm 0.88 (t, J=7.33 Hz, 3 H), 1.06 - 1.16 (m, 4 H), 1.50 (sxt, J=7.53 Hz, 2 H), 2.13 (s, 3 H), 3.76 - 3.86 (m, 1 H), 4.36 (d, J=5.05 Hz, 2 H), 5.90 (s, 1 H), 7.74 (d, J=1.52 Hz, 1 H), 8.09 (s, 1 H), 8.31 (s, 1 H), 8.64 (t, J=4.80 Hz, 1 H), 1 1.54 (br. s., 1 H) ; LC-MS (ES) m/z = 443.0 [M+H]. Example 67
l-cyclopropyl-6-{4-[(dimethy]amino)methyl]pheny]}-/V-[(4,6-dimethyl-2-oxo-l,2-dihydro-3- pyridinyl)methyl]-l/i-indazole-4-carboxamide
Figure imgf000094_0002
To a 20 mL microwave vial containing a mixture of 6-bromo-l-cyclopropyl-N-[(4,6-dimethyl-2-oxo- l,2-dihydro-3-pyridinyl)methyl]-lH-indazole-4-carboxamide (.070g, 0.169 mmol), Ν,Ν-dimethyl- 1 - [4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)phenyl]methanamine (0.060 g, 0.202 mmol), and potassium phosphate (tribasic) (0.107 g, 0.506 mmol) were added 1,4-dioxane (2.0 mL) and water (0.5 mL). The suspension was stirred with degassing under N2 for 10 min. (emulsion), and then PdCl2(dppf)-CH2Cl2 adduct (0.021 g, 0.025 mmol) was added. The contents were sealed and heated at 100 °C (heat block) for 2h. After cooling to RT, the reaction mixture was diluted with EtOAc. Silica gel was added the mixture was concentrated in vacuo to dryness. The contents were purified by silica gel chromatography (dry loaded, eluent : 8-95% gradient chloroform (containing 10% 2M Ammonia in Methanol) and DCM). The collected product was concentrated from MTBE and dried in a vacuum oven for 2h. The title compound was collected as a white solid (67 mg, 81%); H NMR (400 MHz, DMSO-d6) δ ppm 1.12 - 1.20 (m, 4 H), 2.12 (s, 3 H), 2.15 - 2.24 (m, 9 H), 3.44 (s, 2 H), 3.87 (t, J=5.31 Hz, 1 H), 4.38 (d, J=4.80 Hz, 2 H), 5.88 (s, 1 H), 7.42 (m, J=8.34 Hz, 2 H), 7.80 (m, J=8.08 Hz, 2 H), 7.90 (s, 1 H), 8.04 (s, 1 H), 8.31 (s, 1 H), 8.67 (t, J=4.93 Hz, 1 H), 11.53 (s,l H); LC- MS (ES) mix = 470.3 [M+H].
Example 68
l-cyclopropyl-6-{4-[(dimethylamino)methyl]phenyl}-A'-[(6-methyl-2-oxo-4-propyl-l,2-dihydro- 3-
Figure imgf000095_0001
The title compound was prepared in the same manner as described for example 67 from 6-bromo- 1 - cyclopropyl-N-[(6-methyl-2-oxo-4-propyl-l,2-dihydro-3-pyridinyl)methyl]-lH-indazole-4- carboxamide (.075g, 0.169 mmol) and N,N-dimethyl-l -[4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)phenyl]methanamine (0.060 g, 0.203 mmol). The title compound was collected as a white solid (59 mg, 69 %); !H NMR (400 MHz, DMSO-d6) δ ppm 0.88 (t, J=7.33 Hz, 3 H), 1.12 - 1.21 (m, 4 H), 1.45 - 1.58 (m, 2 H), 2.13 (s, 3 H), 2.18 (s, 6 H), 3.45 (s, 2 H), 3.88 (quin, J=5.37 Hz, 1 H), 4.41 (d, J=5.05 Hz, 2 H), 5.91 (s, 1 H), 7.43 (m, J=8.34 Hz, 2 H), 7.80 (m, J=8.08 Hz, 2 H), 7.89 (d, J=1.26 Hz, 1 H), 8.04 (s, 1 H), 8.32 (s, 1 H), 8.68 (t, J=4.93 Hz, 1 H), 11.54 (s, 1 H); LC-MS (ES) m/z = 498.4 [M+H].
Example 69
6-bromo-N-((l,2-dihydro-4,6-dimethyl-2-oxopyridin-3-yl)methyl)-l-isopropyl-3-methyl-lH- indazole-4-carboxamide
Figure imgf000096_0001
a) S-BromG-2-roeth I-3-idtrii-ber!zoic acid
Figure imgf000096_0002
To a stirred solution of 2-methyl-3-nitro benzoic acid (300 g, 1647 mmol) in cone. H2S04 (1.5 L) was added l ,3-dibromo-5,5 dimethyl -2,4-imadazolidinedione (258 g, 906 mmol) and the mixture was stirred at room temperature for 5 h. The reaction mixture was slowly added to ice water (4 L), and solid was precipitated out. The solid was filtered off and washed with water (1.2 L), pet ether (1 1) and dried to afford the title compound as a white solid (41 1 g, 96 %), which was used without further purification. lU NMR (DMSO; 400 MHz) : δ 2.446 (s, 3H), 8.136 (s, 1H), 8.294 (s, 1H). LCMS (ES-) m/z = 257.93 (M-H)- b) Methyl 6-bromo-lH-indole-4-carboxylate
Figure imgf000096_0003
To a stirred solution of 5-Bromo-2-methyi-3~nitro-be«zoic acid (140 g, 538.4 mmol) in DMF (550 ml) was added DMF-DMA (599 mL, 4846 mmol) at room temperature. The reaction mixture was stirred at 1 15 °C for 18 h. The reaction mixture was then concentrated in vacuo. The residual contents (176 g, 536.5 mmol) were dissolved in acetic acid (696 mL) and added to a suspension of Iron (329.2 g, 5902 mmol) in acetic acid (1.4 L) at 50 °C. After completion of addition, the reaction mixture was stirred at 80-90 °C for 4 h. The reaction mixture was then filtered through a celite pad. The filtrate was poured onto ice water (1 L) and extracted with diethyl ether (3 X 700 ml). The combined organic layers were washed with sat NaHC03, brine, and dried over anhydrous Na2S04, filtered, and evaporated under vaccum. The crude product was purified by silica gel chromatography (eluent: 10 % ethyl acetate in pet ether) and afforded the title compound as a solid (80g, 59%). !H NMR (DMSO-d6; 400 MHz) δ: 3.980 (s, 3H), 7.168 (d, J = 3.2 Hz, 1H), 7.334 (d, J = 3.2 Hz, 1H), 7.734 (s, 1H), 8.017 (s, 1H), 8.384 (brs, 1H); LCMS (ES-) m/z = 251.9 (M-H). c) Methyl 6~brome~3~fennyS H ndazoIe~4-earbi>xySate
Figure imgf000097_0001
To a stirred solution of sodium nitrite (68.4 g, 991.3 mmol) in water (1425 niL) was added methyl 6- bromo-l.ff-indole-4-carboxylate (20.64 g, 81.25 mmol) at RT and the mixture stirred for 15 min at RT. To the mixture was added 6N HC1 (159.6 mL) slowly dropwise over a period of 1 h and the reaction mixture was then stirred at room temperature for 48 h. The reaction mixture was extracted with 10% THF in ethyl acetate (5 x 500 mL). The combined organic layers were washed with water, brine, dried over anhydrous Na2S04 , and filtered. The filtrate was concentrated to 1/3 volume and cooled in freezer for 4 h. Precipitated solids were filtered and washed with cold ethyl acetate and dried under high vacuum to afford the title compound as an off white solid (13.7g, 59.6%). !H NMR (DMSO-d6, 400 MHz): δ 3.905 (s, 3H), 7.754 (s, 1H), 8.183 (s, 1H), 10.274 (s, 1H), 14.563 (brs, 1H). LCMS (ES+): 281.06 [M-H] ion present. d) Methyl 6-bromo-3-formyl-l-isopropyl-lH-indazole-4-carboxylate
Figure imgf000097_0002
To a stirred solution of methyl 6-bromo-3-formyl-lH-indazole-4-carboxylate 1 (11.5 g, 40.63 mmol) in DMF (230 mL)) was added K2C03 (14.08 g, 102.02 mmol) at RT under argon, and stirred for 10 min. Then 2-iodopropane (7.24 g, 42.58 mmol) was added at RT and the mixture stirred at 40 °C for 2 h. The reaction mixture was diluted with cold water and extracted with ethyl acetate (4 x 300 mL). The combined organic layers were washed with water, brine, dried over anhydrous Na2S04 , filtered, and concentrated. The crude residue (12.4 g) was purified by silica gel chromatography (eluent: 0- 10% ethyl acetate: pet ether) to afford the title compound as an off white solid (5.55 g, 42 %). lH NMR (CDC13, 400 MHz): δ 1.659 ( d, J = 6.4 Hz, 6H ), 4.017 (s, 3H), 4.862-4.960 (m, 1H), 7.807 (s, 1H), 7.854 (s, 1H), 10.375 (s, 1H). HPLC: 98.66 %. e) Methyl 6-bromo-l-isopropyl-3-methyl-lH-indazole-4-carboxylate
Figure imgf000098_0001
To a stirred solution of methyl 6-bromo-3-formyl-l-isopropyl-lH-indazole-4-carboxylate (4.5 g, 13.84 mmol) in DMF (31.5 mL) was added >-toluenesulfonic acid monohydrate (0.342 g, 1.8 mmol), />-toluenesulfonyl hydrazide (3.35 g, 18.0 mmol) followed by sulfolane (31.5 mL), and the reaction mixture was stirred at 100 °C for 1 h. The reaction mixture was cooled to room temperature and sodium cyanoborohydride (3.489 g, 55.0 mmol) was added portion wise over a period of 25 min. The resulting reaction mixture was stirred at 100 °C for 2 h and then at RT for 6 h. The reaction mixture was diluted with water and extracted with ethyl acetate (4x300 mL). The combined organic layers were washed with water, brine, dried over anhydrous Na2S04 , filtered, and concentrated. The crude residue (4.4 g) was purified by silica gel chromatography (eluent: 0-5% ethyl acetate: pet ether) to afford the title compound as an off white solid (2.95 g, 68 % ). !H NMR (CDC13, 400 MHz): δ 1.559 (d, J = 6.8 Hz, 6H), 2.674 (s, 3H), 3.976 (s, 3H), 4.667-4.767 ( m, 1H), 7.715 (d, J=1.6 Hz, 1H), 7.761 (d, J=1.6 Hz, 1H).-LCMS (ES+): 98.63 %, 313.06 [M+H] ion present. f) 6-bromo-l-isopropyl-3-methyl-lH-indazole-4-carboxylic acid
Figure imgf000098_0002
To a stirred solution of methyl 6-bromo- 1 -isopropyl-3 -methyl- lH-indazole-4-carboxylate (7.5 g, 24.11 mmol) in ethanol (400 mL) was added sodium hydroxide (1.45 g, 36.17 mmol) in water (60 mL) and the reaction mixture was stirred at reflux for 6 h. The reaction mixture was concentrated under reduced pressure and the residue was diluted with water (150 mL), and acidified with 2N HC1 to pH~2. The precipitated acid was collected by filtration, washed with ether (200 mL, and dried to afford 6-bromo-l-isopropyl-3-methyl-lH-indazole-4-carboxylic acid as an off white solid (6.85 g, 95.6%). LCMS (ES-): 294.9 [M-H]. g) 6-bromo-N-((l,2-dihydro-4,6-dimethyl-2-oxopyridin-3-yl)methyl)-l-isopropyl-3-methyl-lH- indazole-4-carboxamide
To a stirred solution of 6-bromo-l-isopropyl-3-methyl-lH-indazole-4-carboxylic acid, 1 (6.5 g, 21.88 mmol) in DCM (250 mL) was added EDC HCl (5.01 g, 26.23 mmol), HOBt (3.545 g, 26.20 mmol) followed by diisopropyl ethyl amine (14.1 1 g, 109.37 mmol) and stirred at room temperature for 15 min. To the resulting mixture, 3-(amino methyl)-4,6-dimethylpyridin-2(lH)-one (3.32 g, 21.84 mmol) followed by DMAP (catalytic amount) was added and the reaction mixture was stirred at room temperature for 5 h. The reaction mixture was diluted with DCM (300 mL) and washed with IN HC1 solution (250 mL), saturated NaHC03 solution (250 mL) and brine solution. The organic layer was dried over anhydrous Na2S04 , filtered, and concentrated under reduced pressure. The residue was triturated with diethyl ether (100 mL), filtered, and dried to afford 6-bromo-N-((l,2-dihydro-4,6- dimethyl-2-oxopyridin-3-yl)methyl)-l-isopropyl-3-methyl-lH-indazole-4-carboxamide as an off white solid (5.5 g, 58 %). :H NMR (DMSO-d6, 400 MHz): δ 1.412 (d, J = 6.4 Hz, 6H), 2.1 12 (s, 3H), 2.215 (s, 3H), 2.383 (s, 3H), 4.319 (d, J = 5.2 Hz, 2H), 4.907 - 4.972 (m, 1H), 5.864 (s, 1H), 7.129 (d, J = 1.2 Hz, 1H), 8.01 1 (d, J = 1.2 Hz, 1H), 8.473 (t, J = 5 Hz, 1H), 11.479 (brs, 1H). LCMS (ES+): 431.02 [M+H].
Example 70
6-bromo-l-isopropyl-3-methyl-N-((6-methyl-2-oxo-4-propyl-l,2-dihydropyridin-3-yl)methyl)- lH-indazole-4-carboxamide
Figure imgf000099_0001
The title compound was prepared in the same manner as described for example 3 (step c) from 6- bromo-3-methyl-l-(l-methylethyl)-lH-indazole-4-carboxylic acid (0.24g, 0.808 mmol) and 3- (aminomethyl)-6-methyl-4-propyl-2(lH)-pyridinone (0.219 g, 1.010 mmol). The title compound was collected as a white solid (0.35g, 92%); !H NMR (400 MHz, DMSO-d6) δ ppm 0.92 (t, J=7.33 Hz, 3 H) 1.41 (d, J=6.57 Hz, 6 H) 1.49 - 1.60 (m, 2 H) 2.12 (s, 3 H) 2.39 (s, 3 H) 2.53 (s, 1
H) 4.33 (d, J=5.05 Hz, 2 H) 4.88 - 5.00 (m, 1 H) 5.89 (s, 1 H) 7.12 (d, J=1.52 Hz, 1 H) 8.02 (d, J=1.77
Hz, 1 H) 8.48 (t, J=4.93 Hz, 1 H) 11.50 (s, 1H); LCMS (ES+): 459.1 [M+H].
Example 71
l-isopropyl-3-methyl-N-((6-methyl-2-oxo-4-propyl-l,2-dihydropyridin-3-yl)methyl)-6-(2-(4- methylpiperazin-l-yl)pyridin-4-yl)-lH-indazole-4-carboxamide
Figure imgf000100_0001
In a 25 mL sealable tube under nitrogen were combined 6-bromo-3-methyl-l-(l-methylethyl)-N-[(6- methyl-2-oxo-4-propyl-l,2-dihydro-3-pyridinyl)methyl]-lH-indazole-4-carboxamide (85 mg, 0.19 mmol), 1 -methyl-4-[4-(4,4,5,5-tetramethyl- 1 ,3,2-dioxaborolan-2-yl)-2-pyridinyl]piperazine (72.9 mg, 0.24 mmol) in dioxane/water (3 mL: lmL). PdCl2(dppf)-CH2Cl2 adduct (7.6 mg, 0.0093 mmol) was added and the resulting mixture was degassed with nitrogen for 10 min. Sodium bicarbonate (46.6 mg, 0.55 mmol) was added, the vessel was sealed, and the insoluble mixture was heated in a microwave at 110 °C for 20 min. The mixture was evaporated, DCM was added and the contents purified by Si02 chromatography (eluent: gradient 0 to 80:20:2 DCM/MeOH/NH4OH). The collected product was suspended in EtOAc along with some hexanes. The contents were sonicated and solids that precipitated were filtered. Acetonitrile was added, the solids were triturated, and again filtered. The collected solid was then suspended in DMF along with some water and then allowed to sit at room temperature overnight. Solids that precipitated were filtered, washed with DCM, and dried to afford the title compound as a light grey solid (34 mg, 32%). !H NMR (400 MHz, DMSO-<i6) δ ppm 11.50 (s, 1 H) 8.45 (t, J=4.93 Hz, 1 H) 8.19 (d, J=5.31 Hz, 1 H) 8.06 - 8.09 (m, 1 H) 7.40 (s, 1 H) 7.17 (s, 1 H) 7.08 (d, J=5.05 Hz, 1 H) 5.91 (s, 1 H) 5.1 1 (quin, J=6.57 Hz, 1 H) 4.39 (br. s., 1 H) 4.38 (br. s., 1 H) 3.55 - 3.62 (m, 4 H) 2.53 - 2.57 (m, 2 H) 2.41 - 2.46 (m, 7 H) 2.24 (s, 3 H) 2.13 (s, 3 H) 1.53 - 1.61 (m, 2 H) 1.47 (s, 3 H) 1.46 (s, 3 H) 0.93 (t, J=7.33 Hz, 3 H); LC-MS (ES) m/z = 459.2 Example 72
3-methyl-l-(l-methylethyl)-N-[(6-methyl-2-oxo-4-propyl-l,2-dihydro-3-pyridinyl)methyl]-6-[6- (l-piperazinyl)-3-pyridinyl]-lH-indazole-4-carboxamide
Figure imgf000101_0001
The title compound was prepared in a similar manner as described for example 8 from 6-bromo-3- methyl- 1 -( 1 -methylethyl)-N- [(6-methyl-2-oxo-4-propyl- 1 ,2-dihydro-3 -pyridinyl)methyl] - 1 H- indazole-4-carboxamide (65 mg, 0.141 mmol) and l-[5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)- 2-pyridinyl]piperazine (61 mg, 0.212 mmol). The product was collected as a white solid (54 mg, 49%). lB NMR (400 MHz, DMSO-i/6) δ ppm 11.54 (br. s., 1 H) 8.49 - 8.63 (m, 2 H) 8.43 (br. s., 1 H) 7.97 (dd, J=8.84, 2.53 Hz, 1 H) 7.89 (s, 1 H) 7.32 (s, 1 H) 6.87 - 6.96 (m, 1 H) 5.91 (s, 1 H) 5.03 (dt, .7=13.07, 6.47 Hz, 1 H) 4.38 (d, .7=4.55 Hz, 2 H) 3.40 - 3.61 (m, 5 H) 2.77 - 2.82 (m, 3 H) 2.53 - 2.57 (m, 2 H) 2.43 (s, 3 H) 2.13 (s, 3 H) 1.53 - 1.60 (m, 2 H) 1.46 (s, 3 H) 1.44 (s, 3 H) 0.93 (t, J=7.33 Hz, 3 H); LC-MS (ES) m/z = 542.2 [M+H]+
Example 73
6-(6-(dimethy]amino)pyridin-3-yl)-N-((l,2-dihydro-4,6-dimethyl-2-oxopyridin-3-yl)
isopropyl-3-methyl- 1 H-indazole-4-carboxamide
Figure imgf000101_0002
To a stirred solution of 6-bromo-N-((l,2-dihydro-4,6-dimethyl-2-oxopyridin-3-yl)methyl)-l- isopropy 1-3 -methyl- lH-indazole-4-carboxamide (300 mg, 0.696 mmol) in DMF (30 mL) was added 6-(dimethylamino)pyridin-3-yl-3-boronic acid, 1 (127 mg, 0.765 mmol) followed by sodium carbonate (184.4 mg,1.74 mmol) dissolved in water (3 mL) and degassed with argon for 1 h. Then PdCl2(PPh3)2 (48.8 mg,0.069 mmol) was added and again degassed with argon for 15 min and stirred at 1 10°C for 4 h. The reaction mixture was filtered through Celite pad and washed with ethyl acetate (50 mL). The filtrate was diluted with water and extracted with ethyl acetate (4x90 mL). The combined organic layers were separated and washed with cold water (2x50 mL), brine solution (50 mL), dried over anhydrous Na2S04, filtered, and concentrated to afford the crude product (450 mg). The crude compound was purified by silica gel chromatography (eluent: 5% MeOH: DCM) to afford 100 mg of desired product which was triturated with hexane and the solid was filtered and dried to afford the title compound as an off white solid (100 mg, 30%). H NMR (DMSO-d6; 400 MHz) : δ 1.559 (d, J = 6.8 Hz, 6H), 2.102 (s, 3H), 2.403 (s, 3H), 2.539 (s, 3H), 3.125 (s, 6H), 4.616 (s, 2H), 4.754 - 4.820 (m, 1H), 5.893 (s, 1H), 6.557 (d, J = 8.8 Hz, 1H),7.313 (s,lH),7.401 (d, J = 5.2 Hz, 1H), 7.448 (s,lH), 7.707 (d, J = 8.8 Hz, 1H), 8.438 (s, 1H), 11.502 (brs, 1H). H NMR (D20 Ex in DMSO, 400 MHz) : δ 1.559 (d, J = 6.8 Hz, 6H), 2.1 15 (s,3H), 2.408 (s, 3H), 2.538 (s, 3H), 3.124 (s, 6H), 4.616 (s, 2H), 4.754 - 4.820 (m, 1H), 5.899 (s, 1H), 6.557 (d, J = 8.8 Hz, 1H), 7.318 (s,lH), 7.450 (s, lH), 7.718 (d, J = 8.8 Hz, 1H), 8.438 (s, 1H). LCMS (ES+): 473.19.
Example 74
6-(3-((diraethylaraino)methyl)phenyl)-N-((l,2-dihydro-4,6-diraethyl-2-oxopyridin-3-yl)methyl)- l-isopropyl-lH-indazole-4-carboxamide
Figure imgf000102_0001
The title compound was prepared from 6-bromo-N-((l,2-dihydro-4,6-dimethyl-2-oxopyridin-3- yl)methyl)-l-isopropyl-lH-indazole-4-carboxamide (500 mg, 1.14 mmol) and N,N-dimethyl(3- (4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)phenyl)methanamine (300 mg, 1.14 mmol) in the same manner as described for example 73 wherein the contents were heated at 120 °C for 2 h. The product was collected as a pale red solid (120 mg, 21%). 'HNMR (Cods, 400 MHz): 51.568 (d, J = 6.8 Hz, 6H), 2.12 (s, 3H), 2.41 (s, 8H), 2.55 (s, 3H), 3.67 (s, 2H), 4.614 (d, J = 5.2 Hz, 2H), 4.899-4.834 (m, 1H), 5.90 (s, 1H), 7.326-7.308 (m, 1Η),7.408-7.368 (m, 3H), 7.558 (d, J = 7.6 Hz, 1H, ), 7.639 (s, 1H), 7.699 (s, 1H), 11.193 (brs,lH). LCMS (ES+) m/z: 486.31.
Example 75
N-((4,6-dimethyl-2-oxo-l,2-dihydropyridin-3-yl)methyl)-6-(4-fluorophenyl)-l-isopropyl-3- methyl-lH-indazole-4-carboxamide
Figure imgf000102_0002
In a 25 mL sealable tube under nitrogen were combined 6-bromo-N-[(4,6-dimethyl-2-oxo-l,2- dihydro-3-pyridinyl)methyl]-3-methyl-l-(l-methylethyl)-lH-indazole-4-carboxamide (90 mg, 0.21 mmol), (4-fluorophenyl)boronic acid (43.8 mg, 0.31 mmol) in dioxane/water (3 mL: lmL). PdCl2(dppf)-CH2Cl2 adduct (5.11 mg, 0.006 mmol) was added and the resulting mixture was degassed with nitrogen for 10 min. Sodium bicarbonate (52.6 mg, 0.63 mmol) was added, the vessel was sealed, and the insoluble mixture was heated in a microwave at 100 °C for 20 min. The mixture was evaporated, DCM/MeOH (1 : 1) was added, the contents pre-absorbed on silica gel and purified by Si02 chromatography (eluent: gradient 0 to 80:20:2 DCM7MeOH/NH4OH). The collected product was suspended in EtOAc along with some hexanes. The mixture was sonicated, and the solids that precipitated were filtered and dried to afford the title compound as a white solid (73 mg, 77%). lB NMR (400 MHz, DMSO-c 6) δ ppm 11.50 (s, 1 H) 8.46 (t, J=5.05 Hz, 1 H) 7.94 (d, J=1.26 Hz, 1 H) 7.83 - 7.88 (m, 2 H) 7.31 - 7.36 (m, 3 H) 5.88 (s, 1 H) 5.06 (quin, J=6.63 Hz, 1 H) 4.38 (s, 1 H) 4.36 (s, 1 H) 2.43 (s, 3 H) 2.24 (s, 3 H) 2.12 (s, 3 H) 1.47 (s, 3 H) 1.45 (s, 3 H). LC-MS (ES) m/z = 446.9
Example 76
N-((l,2-dihydro-4,6-dimethyl-2-oxopyridin-3-yl)methyl)-l-isopropyl-3-methyl-6-(6-(4- methylpiperazin-l-yl)pyridin-3-yl)-lH-indazole-4-carboxamide
Figure imgf000103_0001
To a stirred solution of 6-bromo-N-((l,2-dihydro-4,6-dimethyl-2-oxopyridin-3-yl)methyl)-l- isopropy 1-3 -methyl- lH-indazole-4-carboxamide (0.3 g, 0.69 mmol) in DMF (15 mL) was added 1- methyl-4-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyridin-2-yl)piperazine (0.25 g, 0.82 mmol) followed by PdCl2(PPh3)2 (0.097 g, 0.13 mmol) and the mixture stirred 5 min. Sodium carbonate (0.184 g, 1.73 mmol) dissolved in water (2 mL) was added and the resulting reaction mixture was stirred at 1 10 °C for 4 h. The contents were then diluted with sodium bicarbonate solution and extracted with ethyl acetate (3x20 mL). The combined organic layers were washed with brine solution (20 mL), dried over anhydrous Na2S04, filtered, and concentrated to afford the crude product. The crude compound was purified by silica gel chromatography (eluent: 5% MeOHMDCM). The desired product was isolated as an off white solid with trace impurities. The impure compound was washed several times with cold water and triturated with hexane to afford the title compound as an off-white solid (80 mg, 22%). ¾ NM (DMSO-d6; 400 MHz) : δ 1.447 (d, J = 6.4 Hz, 6H), 2.112 (s, 3H), 2.235 (s, 6H), 2.41 1 (s, 7H), 3.551 (s, 4H), 4.363 (d, J = 4.4 Hz, 2H), 4.991-5.054 (m, 1H), 5.870 (s, 1H), 6.942 (d, J = 9.2 Hz, 1H), 7.321 (s, 1H), 7.880 (s, 1H), 7.99 (d, J = 8.8 Hz, 1H), 8.403 (s, 1H ), 8.569 (s, 1H), 1 1.483 (brs, 1H). LCMS (ES+) m/z: 528.29.
Example 77
6-(4-((dimethylamino)methyl)phenyl)-N-((l,2-dihydro-4,6-dimethyl-2-oxopyridin-3-yl)methyl)- l-isopropy]-3-methyl-lH-indazole-4-carboxamide
Figure imgf000104_0001
The title compound was prepared from 6-bromo-N-((l,2-dihydro-4,6-dimethyl-2-oxopyridin-3- yl)methyl)-l-isopropyl-lH-indazole-4-carboxamide (300 mg, 0.696 mmol) and dimethyl-[4-(4,4,5,5- tetramethyl-[l,3,2]dioxaborolan-2-yl)-benzyl]-amine (199 mg, 0.765 mmol) in the same manner as described for example 73. The final product was collected as a grey colored solid (80 mg, 26%). 1HNMR (DMSO-d6, 400 ΜΗζ):δ 1.458 (d, J = 6.4 Hz, 6H), 2.108 (s, 3H), 2.176 (s, 8H), 2.55 (s, 3H), 3.67 (s, 2H), 4.614 (d, J = 5.2 Hz, 2H), 4.899-4.834 (m, 1H), 5.90 (s, 1H), 7.326-7.308 (m, 1H), 7.408-7.368 (m, 3H), 7.558 (d, J = 7.6 Hz, 1H), 7.639 (s, 1H), 7.699 (s, 1H), 1 1.193 (brs, 1H). LCMS (ES+) m/z: 486.20 (M+H).
Example 78
N-((l,2-dihydro-4,6-dimethyl-2-oxopyridin-3-yl)methyl)-l-isopropyl-3-methyl-6-(6-(piperazin-l- yl)pyridin-3-yl)-lH-indazole-4-carboxamide hydrochloride
Figure imgf000105_0001
.HCI a) tert-butyl 4-(5-(4-((l,2-dihydro-4,6-dimethyl-2-oxopyridin-3-yl)methylcarbamoyl)-l- isopropyl-3-methyl-lH-indazol-6-yl)pyridin-2-yl)piperazine-l-carboxylate
Figure imgf000105_0002
To a stirred solution of 6-brorno-N-((l,2-dihydro-4,6-dimethyl-2-oxopyridin-3-yl)methyl)- l - isopropy 1-3 -methyl- lH-indazole-4-carboxamide (400 mg, 0.928 mmol) in DMF (20 mL) was added tert-butyl 4-(5-(4,4,5,5-tetramethyl- 1 ,3,2-dioxaborolan-2-yl)pyridin-2-yl)piperazine- 1 -carboxylate (397 mg, 1.02 mmol) followed by sodium carbonate (246 mg, 2.32 mmol) dissolved in water (4 mL) and degassed with argon for 30 min. PdCl2(PPh3)2 (65 mg, 0.092 mmol) was added and the mixture again degassed with argon for 10 min. The reaction mixture was stirred at 100 °C for 3 h. The reaction mixture was diluted with water (100 mL) and extracted with ethyl acetate (3x150 mL). The combined organic layers were dried over anhydrous Na2S04 , filtered, and concentrated to afford the crude product (650 mg). The crude compound was purified by silica gel chromatography (eluent: 0- 10% MeOH: DCM). The product was triturated with diethyl ether (150 mL) to afford the title compound as an off white solid (270 mg, 47%). *H NMR (DMSO-d6, 400 MHz): δ 1.498 (s, 9H), 1.560 (d, J = 6.4 Hz, 6H), 2.128 (s, 3H), 2.413 (s, 3H), 2.526 (s, 3H), 3.579 (s, 8H), 4.610 (d, J = 5.6 Hz, 2H), 4.760-4.824 (m, 1H), 5.907 (s, 1H), 6.70 (d, J = 8.8 Hz, 1H ), 7.31 1-7.346 (m, 2H),7.454 (s, 1H), 7.735 (d, J = 8.8 Hz, 1H ), 8.463 (s, 1H), 10.991 (brs, 1H). LCMS (ES+): 614.25. b) N-((l,2-dihydro-4,6-dimethyl-2-oxopyridin-3-yl)methyl)-l-isopropyl-3-methyl-6-(6- (piperazin-l-yl)pyridin-3-yl)-lH-indazole-4-carboxamide hydrochloride To a stirred solution of tert-butyl 4-(5-(4-((l ,2-dihydro-4,6-dimethyl-2-oxopyridin-3- yl)methylcarbamoyl)- 1 -isopropyl-3 -methyl- 1 H-indazol-6-yl)pyridin-2-yl)piperazine- 1 -carboxylate (250 mg, 0.407 mmol) in DCM (50 mL) was added 4M HC1 in 1,4-dioxane (6 mL) at 0 °C and stirred at room temperature for 2 h. The reaction mixture was concentrated and then co-distilled with toluene (50 mL). The residue was dried under reduced pressure to get the crude HC1 salt (195 mg). The residue was triturated with diethyl ether (100 mL), DCM (100 mL) and n-hexane (100 mL) to afford the title compound as a pale brown solid (135 mg, 60%). lU NMR (DMSO-d6, 400 MHz): δ 1.454 (d, J = 6.4 Hz, 6H), 2.1 19 (s, 3H), 2.244 (s, 3H), 2.412 (s, 3H), 3.222 (s, 4H), 3.839 (s, 4H), 4.369 (d, J= 4.4 Hz, 2H), 5.005-5.071 (m, 1H), 5.888 (s, 1H), 7.125 (d, J = 8.8 Hz, 1H ), 7.348 (s, 1H), 7.942 (s, 1H), 8.158 (d, J = 8 Hz, 1H), 8.429 (t, J = 4.6 Hz, 1H ), 8.613 (d, J = 2.0 Hz, 1H), 9.145 (s, 2H), 1 1.519 (brs, 1H).LCMS (ES+): 514.24 [M+H].
Example 79
N-((4,6-dimethyl-2-oxo-l,2-dihydropyridin-3-yl)methyl)-l-isopropyl-3-methyl-6-(6-(4- methylpiperazin-l-yl)pyridin-2-yl)-lH-indazole-4-carboxamide
Figure imgf000106_0001
A stirred solution of 6-bromo-N-((4,6-dimethyl-2-oxo- l ,2-dihydropyridin-3-yl)methyl)-l-isopropyl- 3-methyl-lH-indazole-4-carboxamide, 3 (200 mg, 0.464 mmol), l -methyl-4-(4-(4,4,5,5-tetramethyl- l,3,2-dioxaborolan-2-yl)pyridin-2-yl)piperazine (154 mg, 0.510 mmol) and Na2C03 (123 mg, 1.160 mmol) in a mixture of water (3 mL) and DMF (15 mL) was degassed with argon gas for 30 min. Then PdCl2(PPh3)2 (16.28 mg, 0.023 mmol) was added and heated to 120 °C for 3 h. Then the reaction mixture was filtered through Celite bed and the Celite bed was washed with ice cold water (2x100 mL). The filtrate was extracted with EtOAc (20 mL), dried over anhydrous sodium sulphate, filtered, and concentrated to afford 150 mg of crude product. The crude product was purified by silica gel chromatography over (eluent: 5% MeOH/DCM) to afford the title compound as an off-white solid (85 mg, 32%).1HNMR(DMSO-d6, 400MHz): δ 0.889 (t, J = 14.4 Hz, 3H), 1.554-1.483 (m, 8H), 2.134 (s, 3H), 2.55 (s, 3H), 4.411 (d, J = 1.6 Hz, 2H), 5.059 (m, 1H), 5.913 (s, 1H), 7.830 (s, 1H), 8.047 (s, 1H), 8.08 (s, 1H), 8.306 (s, 1H), 8.462 (s, 1H), 11.53 (s, 1H), 13 (brs, 1H). LCMS (ES+) m/z = 528.2.
Example 80
N-((l,2-dihydro-4,6-dimethyl-2-oxopyridin-3-yl)methyl)-l-isopropyl-3-methyl-6-(pyridin-3-yl)- lH-indazole-4-carboxamide
Figure imgf000107_0001
a) methyl 3-methyl-l-(l-methylethyl)-6-(3-pyridinyl)-l -indazole-4-carboxylate
Figure imgf000107_0002
To a stirred solution of methyl 6-bromo-l-isopropyl-3-methyl-lH-indazole-4-carboxylate (0.5 g, 1.60 mmol) in 1,4-dioxane (25 mL) was added 3-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyridine (0.39 g, 1.92 mmol) followed by PdCl2(dppf)-CH2Cl2 adduct (0.26 g, 0.32 mmol). The reaction mixture was stirred for 5 min, then sodium bicarbonate (0.4 g, 4.80 mmol) dissolved in water (8 mL) was added and the reaction mixture was stirred at 100 °C for 6 h. The reaction mixture was diluted with water and extracted with ethyl acetate (3x40 mL). The combined organic layers were washed with cold water (2x25 mL), brine solution (25 mL), dried over anhydrous Na2S04, filtered, and concentrated to afford the crude product. The crude compound was purified by silica gel chromatography (eluent: 100 % ethyl acetate) to afford the title compound as an off-white solid (470 mg, 94%). ¾ NMR (DMSO-d6, 400 MHz): δ 1.485 (d, J = 6.4 Hz, 6H), 2.601 (s, 3H), 3.945 (s, 3H), 5.121-5.186 (m, 1H), 7.523-7.554 (m,lH), 7.902 (s, 1H) , 8.232 (d, J = 7.6 Hz, 1H), 8.316 (s, 1H), 8.625 (d, J = 4Hz, 1H), 9.045 (s, 1H). b) l-isopropyl-3-methyl-6-(pyridin-3-yl)-lH-indazole-4-carboxylic acid
Figure imgf000108_0001
To a stirred solution of methyl l-isopropyl-3-methyl-6-(pyridin-3-yl)-lH-indazole-4-carboxylate, 1 (0.5 g, 1.618 mmol) in a mixture of THF and H20 (30 mL) was added LiOH¾0 (0.2 g, 4.85 mmol) and the mixture was refluxed at 80 °C for 8 h. THF was distilled off and the aqueous layer was adjusted to pH~5 with 10% HCl at 0 °C and the precipitated solid was collected by filtration and dried to afford l-isopropyl-3-methyl-6-(pyridin-3-yl)-lH-indazole-4-carboxylic acid as an off- white solid (0.51 g). LCMS (ES+) m/z: 296.7. c) N-((l,2-dihydro-4,6-dimethyl-2-oxopyridin-3-yl)methyl)-l-isopropyl-3-methyl-6-(pyridin-3- yl)-lH-indazole-4-carboxamide
To a stirred solution of l-isopropyl-3-methyl-6-(pyridin-3-yl)-lH-indazole-4-carboxylic acid (0.5 g, 1.69 mmol) in DCM (30 mL) was added EDC HCl (0.39 g, 2.03 mmol), HOBT (2.74 g, 2.03 mmol) followed by DIPEA (1.47 mL, 8.45 mmol) and stirred at room temperature for 15 min. To the resulting reaction mixture, 3-(amino methyl)-4,6-dimethylpyridin-2(lH)-one (0.257 g, 1.69 mmol) was added and stirred at room temperature for overnight. The reaction mixture was filtered and the precipitate was washed with water followed by ether and dried. The solid compound was further purified by silica gel chromatography (eluent: 10 % MeOH in ethyl acetate) to afford the title compound as an off-white solid (225 mg, 31%). *H NMR (DMSO-d6, 400 MHz): 81.461 (d, J = 6.4 Hz, 6H), 2.109 (s, 3H), 2.240 (s, 3H), 2.438 (s, 3H), 4.372 (d, J = 4.8 Hz, 2H), 5.042-5.107 (m, 1H), 5.870 (s, 1H), 7.410 (s, 1H), 7.501-7.533 (m, 1H), 8.061 (s, 1H), 8.215 (d, J = 8 Hz, 1H ), 8.461 (t, J = 4.8 Hz, 1H), 8.593 (d, J = 4.8 Hz, 1H), 9.036 (s, 1H), 11.489 (brs, 1H). LCMS (ES+) m/z: 430.12 Example 81
N-((l,2-dihydro-6-methyl-2-oxo-4-propylpyridin-3-yl)methyl)-l-isopropyl-3-methyl-6-(lH- pyrrolo[2,3-b]pyridin-5-yl)-lH-indazole-4-carboxamide
Figure imgf000108_0002
a) Methyl l-isopropyl-3-methyl-6-(lH-pyrrolo[2,3-b]pyridin-5-yl)-lH-indazole-4-carboxylate
Figure imgf000109_0001
The title compound was prepared from methyl 6-bromo- 1 -isopropy 1-3 -methyl- lH-indazole-4- carboxylate, (0.7 g, 2.25 mmol) and 5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-lH-pyrrolo[2,3- b]pyridine (0.65 g, 2.70 mmol) in the same manner as described for example 80 (step a). The product was collected as an off-white solid (600 mg, 82.5%). !H NMR (DMSO-d6, 400 MHz) : δ 1.489 (d, J = 6.4 Hz, 6H), 2.603 (s, 3H), 3.949 (s, 3H), 5.140-5.172 (m, 1H), 6.547 (s, 1H), 7.545 (s, 1H), 7.935 (s, 1H), 8.253 (s, 1H), 8.377 (s, 1H), 8.667 (s, 1H), 11.774 (s, 1H). LCMS (ES+) m/z: 349. b) l-isopropyl-3-methyl-6-(lH-pyrrolo[2,3-b]pyridin-5-yl)-lH-indazole-4-carboxylic acid
Figure imgf000109_0002
The title compound was prepared from methyl l-isopropyl-3-methyl-6-(lH-pyrrolo[2,3-b]pyridin-5- yl)-lH-indazole-4-carboxylate, 1 (50 mg, 0.14 mmol) and LiOH H20 (20 mg, 0.43 mmol) in the same manner as described for example 80 (step b). The product was collected as an off-white solid (60 mg) and used in the next step without any further purification. LCMS (ES+) m/z: 335.15. c) N-((l,2-dihydro-6-methyl-2-oxo-4-propylpyridin-3-yl)methyl)-l-isopropyl-3-methyl-6-(lH- pyrrolo[2,3-b]pyridin-5-yl)-lH-indazole-4-carboxamide
The title compound was prepared in the same manner as described for example 80 (step c) from 1- isopropyl-3-methyl-6-(lH-pyrrolo[2,3-b]pyridin-5-yl)-lH-indazole-4-carboxylic acid (50 mg, 0.149 mmol) and 3-(aminomethyl)-6-methyl-4-propylpyridin-2(lH)-one (26 mg, 0.149 mmol) wherein the contents were stirred at RT for 5h. The crude product was washed with DCM, filtered, and dried to afford the title compound as an off-white solid. (48 mg, 64 %). !H NMR (DMSO-d6i 400 MHz) : δ0.930 (t, J = 7.6 Hz, 3H), 1.468 (d, J = 6.4 Hz, 6H), 1.544-1.601 (m, 2H), 2.123 (s, 3H), 2.452 (s, 3H), 2.501 (s, 2H), 4.394 (d, J = 4 Hz, 2H), 5.060-5.093 (m, 1H), 5.902 (s, 1H), 6.526 (s, 1H), 7.420 (s, 1H), 7.528 (s, 1H), 7.996 (s, 1H), 8.348 (s, 1H), 8.445 (s, 1H), 8.656 (s, 1H), 11.490 (brs, 1H), 11.735 (brs, 1H). LCMS (ES+) m/z: 497.49 Example 82
N-((l,2-dihydro-4,6-dimethyl-2-oxopyridin-3-yl)methyl)-l-isopropyl-3-methyl-6-(lH-pyrazol-4- yl)-lH-indazole-4-carboxamide
Figure imgf000110_0001
a) Methyl l-isopropyl-3-methyl-6-(lH-pyrazol-4-yl)-lH-indazole-4-carboxylate
Figure imgf000110_0002
The title compound was prepared in the same manner as described for example 80 (step a) from methyl 6-bromo-l-isopropyl-3-methyl-lH-indazole-4-carboxylate (0.5 g, 1.60 mmol) and 4-(4,4,5,5- tetramethyl-l,3,2-dioxaborolan-2-yl)-lH-pyrazole (0.37 g, 1.92 mmol) wherein the reaction mixture was heated at 115 °C for 3 h. The crude compound was purified silica gel chromatography (eluent: 50 % ethyl acetate\pet ether) to afford the title compound as an off-white solid. !H NM (DMSO-d6> 400 MHz) : 51.470 (d, J = 6.4 Hz, 6H), 2.543 (s, 3H), 3.928 (s, 3H), 5.000 - 5.063 (m, 1H), 7.822 (s, 1H), 8.102 (d, J = 9.6 Hz, 2H), 8.374 (s, 1H), 13.030 (brs, 1H). LCMS (ES+) m/z: 299.25. b) l-isopropyl-3-methy]-6-(lH-pyrazol-4-yl)-lH-indazole-4-carboxylic acid
Figure imgf000110_0003
The title compound was prepared from methyl l-isopropyl-3-methyl-6-(lH-pyrazol-4-yl)-lH- indazole-4-carboxylate (0.2 g, 0.67 mmol) and L1OH.H2O (0.084 g, 2.01 mmol) in the same manner as described for example 80 (step b). The product was collected as a white solid (0.2 g). l NMR (DMSO-d6, 400 MHz) : δ 1.467 (d, J = 6.4 Hz, 6H), 2.568 (s,3H), 4.984-5.048 (m, 1H), 7.790 (s, 1H), 8.058 (s, 1H), 8.219 (brs, 2H), 13.05 (brs, 2H). LCMS (ES+) m/z: 285.10 [M+H]. c) N-((l,2-dihydro-4,6-dimethyl-2-oxopyridin-3-yl)methyl)-l-isopropyl-3-methyl-6-(lH-pyrazol- 4-yl)-lH-indazole-4-carboxamide The title compound was prepared from l -isopropyl-3-methyl-6-(lH-pyrazol-4-yl)- lH-indazole-4- carboxylic acid and 3-(aminomethyl)-4,6-dimethylpyridin-2(lH)-one (0.059 g, 0.387 mmol) in the same manner as described for example 80 (step c) wherein the contents were stirred at RT for 5h. The product was collected as a pale yellow solid (30 mg, 20%). lU NMR (DMSO-d6, 400 MHz): 51.563 (d, J = 6.8 Hz, 6H), 2.207 (s, 3H), 2.455 (s, 3H), 2.556 (s, 3H), 4.616 (d, J = 6 Hz, 2H), 4.781 - 4.798 (m, 1H), 5.899 (s, 1H), 7.236 (s, 1H), 7.488 (s, 1H) 7.522 (s, 1H), 7.797 (d, J = 9.6 Hz, 2H).LCMS (ES+) m/z: 419.45.
Example 83
N-((l,2-dihydro-4,6-dimethyl-2-oxopyridin-3-yl)methyl)-l-isopropyl-3-methyl-6-(pyridin-3-yl)- lH-indazole-4-carboxamide
Figure imgf000111_0001
The title compound was prepared in the same manner as described for example 80 (step c) from 1 - isopropyl-3-methyl-6-(lH-pyrazol-4-yl)- lH-indazole-4-carboxylic acid (0.15 g, 0.52 mmol) and 3- (aminomethyl)-6-methyl-4-propylpyridin-2(lH)-one (0.09 g, 0.52 mmol) wherein the contents were stirred at RT for 5h. The crude compound was purified by silica gel chromatography (eluent: 2 % MeOHYDCM) to afford the title compound as an off-white solid (60 mg, 25%). lB NMR (DMSO-d6> 400 MHz): 50.938 (t, J = 7.2 Hz, 3H), 1.447 (d, J = 6.8 Hz, 6H), 1.540-1.597 (m, 2H), 2.122 (s, 3H), 2.380 (s, 3H), 2.548 (t, J = 3.6 Hz, 2H), 4.369 (d, J = 5.2 Hz, 2H), 4.920 - 4.987 (m, 1H), 5.899 (s, 1H), 7.330 (s, 1H), 7.869 (s, 1H), 8.021 (s, 1H), 8.293 (t, J = 4.8 Hz, 2H), 1 1.483 (brs, 1H), 12.963 (bra, 1H).-LCMS (ES+) m/z: 447.48.
Example 84
V-[(4,6-dimethyl-2-oxo-l,2-dihydro-3-pyridinyl)methyl]-3-methyl-l-(l-methylethyl)-6-(6- methyl-3-pyridinyl)-l -indazole-4-carboxamide
Figure imgf000112_0001
The title compound was prepared in the same manner as described for example 76 from 6-bromo-N- ((l ,2-dihydro-4,6-dimethyl-2-oxopyridin-3-yl)methyl)- l -isopropyl-3-methyl-lH-indazole-4- carboxamide (300 mg, 0.696 mmol) and 2-memyl-5-(4,4,5,5-tetramethyl- l,3,2-dioxaborolan-2- yl)pyridine (183 mg, 0.835 mmol). The title compound was collected as 75 mg (24%); !H NMR (DMSO-d6, 400 MHz): δ 1,45 (d, J = 6.4 Hz, 3H), 2.1 1 (s, 3H), 2.24 (s, 3H), 2.49 (s, 3H), 2.51 (s, 3H), 4.36-4.38 (m, 2H), 5.03-5.09 (m, 1H), 5.87 (s, 1H), 7.36 (s, 1H), 7.38 (s, 1H), 8.01 (s, 1H), 8.1 1 (d, J = 2.4 Hz, 1H), 8.45 (s, 1H), 8.89 (s, 1H), 1 1.48 (s, 1H); LCMS (ES+) m/z: 444.21.
Example 85
6-[6-(dimethylamino)-3-pyridinyl]-3-methyl-l-(l-methylethyl)-N-[(6-methyl-2-oxo-4-propyl-l,2- dihydro-3-pyridinyl)methyl]-l//-indazole-4-carboxamide
Figure imgf000112_0002
The title compound was prepared in the same manner as described for example 67 from 6-bromo-3-methyl- l - (l-methylethyl)-N-[(6-methyl-2-oxo-4-propyl-l,2-dihydro-3-pyridinyl)methyl]- lH-indazole-4-carboxamide (.079 g, 0.172 mmol) and [6-(dimethylamino)-3-pyridinyl]boronic acid (0.034 g, 0.206 mmol). The produd was triturated from EtOAc spiked with DCM, and dried in a hi-vacuum oven for 4h. The title compound was collected as a white solid (49 mg, 56%); H NMR (400 MHz, DMSO-d6) δ ppm 0.93 (t, J=7.33 Hz, 3 H), 1 Ai (d, J=6.57 Hz, 6 H), 1.51 - 1.64 (m, 2 H), 2.12 (s, 3 H), 2.42 (s, 3 H), 2.52 - 2.59 (m, 2 H), 3.08 (s, 6 H), 4.3 (d, J=5.05 Hz, 2 H), 4.96 - 5.09 (m, 1 H), 5.90 (s, 1 H), 6.75 (d, J=9.09 Hz, 1 H), 7.31 (d, J=1.26 Hz, 1 H) 7.86 (d, J=1.26 Hz, 1 H), 7.96 (dd, J=8.97, 2.65 Hz, 1 H), 8.40 (t, J=4.93 Hz, 1 H), 8.55 (d, J=2.27 Hz, 1 H) 1 1.50 (s, 1 H); LCMS (ES+) m/z: 501.1. Example 86
6-{3-[(dimethylamino)methyl]phenyl}-3-methyl-l-(l-methylethyl)-A'-[(6-methyl-2-oxo-4-propyl-l,2- dihydro-3-pyridinyl)methyl]-l//-indazole-4-carboxamide
Figure imgf000113_0001
The title compound was prepared in the same manner as described for example 67 from 6-bromo-3-methyl- l - (l-methylethyl)-N-[(6-methyl-2-oxo-4-propyl-l,2-dihydro-3-pyridinyl)methyl]- lH-indazole-4-carboxamide (.090 g, 0.196 mmol). The crude product was purified by reverse phase HPLC (Gradient B: 10-60%. 8mir A: Water + .1% TFA. B: CH3CN + .1% TFA). The isolated product was then treated with 1.0 g of Silicyck Carbonate resin, and filtered through celite washing with 10% MeOH/DCM. The product was collected as ί white solid (58 mg, 57 %); ¾ NMR (400 MHz, DMSO-d6) δ ppm 0.93 (t, J=7.33 Hz, 3 H) 1.46 (d, 1=6.51 Hz, 6 H) 1.52 - 1.63 (m, 2 H) 2.10 - 2.21 (m, 9 H) 2.44 (s, 3 H) 2.52 - 2.59 (m, 2 H) 3.47 (s, 2 H) 4.38 (d J=5.05 Hz, 2 H) 5.02 - 5.14 (m, 1 H) 5.90 (s, 1 H) 7.28 - 7.36 (m, 2 H) 7.44 (t, J=7.58 Hz, 1 H) 7.63 - 7.73 (m 2 H) 7.92 (d, J=1.26 Hz, 1 H) 8.47 (t, J= 93 Hz, 1 H) 1 1.50 (s, 1 H); LCMS (ES+) m/z: 514.3.
Example 87
6-{4-[(dimethylamino)methyl]phenyl}-3-methyl-l-(l-methylethyl)-/V-[(6-methyl-2-oxo-4-propyl-l,2- dihydro-3-pyridinyl)methyl]-l//-indazole-4-carboxamide
Figure imgf000113_0002
The title compound was prepared in the same manner as described for example 67 from 6-bromo-3-methyl- l - (l-methylethyl)-N-[(6-methyl-2-oxo-4-propyl-l,2-dihydro-3-pyridinyl)methyl]- lH-indazole-4-carboxamide (.090 g, 0.196 mmol) and N,N-dimethyl-l-[4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)phenyl]methanamine (0.070 g, 0.235 mmol). The product was collected as a white solid (72 mg, 70%); ¾ NMR (400 MHz, DMSO-d6) δ ppm 0.93 (t, J=7.33 Hz, 3 H), 1.45 (d, J=6.57 Hz, 6 H), 1.56 (sxt, J=7.53 Hz, 2 H), 2.07 - 2.23 (m, 9 H), 2.44 (s, 3H), 2.52 - 2.59 (m, 2 H), 3.43 (s, 2 H), 4.38 (d, J=5.05 Hz, 2 H), 5.06 (quin J=6.63 Hz, 1 H), 5.90 (s, 1 H), 7.33 - 7.45 (m, 3 H), 7.75 (d, J=8.08 Hz, 2 H), 7.93 (d, J=1.26 Hz, 1 H), 8.4f (t, J=4.93 Hz, 1 H), 1 1.50 (s, 1 H); LCMS (ES+) m/z: 514.5.
Example 88
3-methyl-l-(l-methylethyl)-N-[(6-methyl-2-oxo-4^ropyl-l,2-dihydro-3-pyridinyl)methyl]
pyridinyl)-liMndazole-4-carboxamide
Figure imgf000114_0001
The title compound was prepared in the same manner as described for example 67 from 6-bromo-3-methyl-l- (l-methylethyl)-N-[(6-methyl-2-oxo-4-propyl-l,2-dihydro-3-pyridinyl)methyl]- lH-indazole-4-carboxamide (.10g, 0.218 mmol). The product was collected as a white solid (.097g, 95%); ¾ NMR (400 MHz, DMSO d6) δ ppm 0.92 (t, J=7.33 Hz, 3 H), 1.46 (d, J=6.57 Hz, 6 H), 1.51 - 1.64 (m, 2 H), 2.12 (s, 3 H), 2.45 (s, 3 H): 2.54 (dd, J=8.72, 6.95 Hz, 2 H), 4.38 (d, J=5.05 Hz, 2 H), 5.08 (quin, J=6.57 Hz, 1 H) ,5.90 (s, 1 H), 7.40 (d J=1.52 Hz, 1 H), 7.52 (dd, J=8.46, 5.18 Hz, 1 H), 8.07 (d, J=1.52 Hz, 1 H), 8.21 (dt, J=8.27, 1.80 Hz, 1 H) 8.46 (t, J=4.93 Hz, 1 H), 8.59 (dd, J=4.67, 1.64 Hz, 1 H), 9.03 (d, J=1.52 Hz, 1 H), 11.51 (s, 1 H); LCMS (ES+) m/z: 457.2. Example 89
3-methyl-l-(l-methylethyl)-A'-[(6-methyl-2-oxo-4-propyl-l,2-dihydro-3-pyridinyl)methyl]-6-[6-(4- methyl-l-piperazinyl)-3-pyridinyl]-l//-indazole-4-carboxamide
Figure imgf000114_0002
The title compound was prepared in the same manner as described for example 67 from 6-bromo-3- methyl- 1 -( 1 -methylethyl)-N- [(6-methyl-2-oxo-4-propyl- 1 ,2-dihydro-3 -pyridinyl)methyl] - 1 H- indazole-4-carboxamide (0.10g, 0.218 mmol) and l-methyl-4-[5-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)-2-pyridinyl]piperazine (0.086 g, 0.283 mmol). The title compound was collected as a white solid (0.105 g, 84%); ¾ NMR (400 MHz, DMSO-d6) δ ppm 0.92 (t, J=7.33 Hz, 3 H), 1.45 (d, J=6.57 Hz, 6 H), 1.50 - 1.63 (m, 2 H), 2.12 (s, 3 H), 2.22 (s, 3 H), 2.38 - 2.45 (m, 7 H), 2.52 - 2.59 (m, 2 H), 3.51 - 3.60 (m, 4 H), 4.37 (d, J=5.05 Hz, 2 H), 4.96 - 5.10 (m, 1 H), 5.90 (s, 1 H), 6.94 (d, J=8.84 Hz, 1 H), 7.32 (d, J=1.26 Hz, 1 H), 7.89 (d, J=1.26 Hz, 1 H), 7.98 (dd, J=8.84, 2.53 Hz, 1 H), 8.40 (t, J=5.05 Hz, 1 H), 8.56 (d, J=2.27 Hz, 1 H), 1 1.50 (s, 1 H); LCMS (ES+) m/z: 556.1.
Example 90
6-cyano-N-((4,6-dimethyl-2-oxo-l,2-dihydropyridin-3-yl)methyl)-l-isopropyl-3-methyl-lH- indazole-4-carboxamide
Figure imgf000115_0001
In a 25 mL sealable tube under nitrogen were combined 6-bromo-N-[(4,6-dimethyl-2-oxo-l,2- dihydro-3-pyridinyl)methyl]-3-methyl-l-(l-methylethyl)-lH-indazole-4-carboxamide (130 mg, 0.3 mmol), dicyanozinc (40.7 mg, 0.35 mmol), Pd2(dba)3 (317 mg, 0.35 mmol), dppf (21.7 mg, 0.04 mmol) and zinc (4.9 mg, 0.08 mmol) in DMF (4 mL) and the resulting mixture was degassed with nitrogen for 10 min. The vessel was sealed, and the insoluble mixture was heated to 120 °C for 6 hours. Upon cooling down, the mixture was quenched with water, and dark grey solids that crashed out and were filtered. DCM/MeOH (1 : 1) was added, the contents pre-absorbed on silica gel and purified by Si02 chromatography (eluent: gradient 0 to 80:20:2 DCM/MeOH/NH4OH). The collected product was futher purified by reverse-phase HPLC (20% to 80% CH3CN in water with 0.1% TFA) which afforded the TFA salt. CH3CN was evaporated, and a saturated solution of sodium bicarbonate was added to the water layer. The solids that precipitated were filtered and dried to afford the title compound as a white solid (70.3 mg, 60%). ¾ NMR (400 MHz, DMSO-J6) δ ppm 11.39 (br. s., 1 H) 8.56 (t, .7=4.80 Hz, 1 H) 8.43 (d, J=1.01 Hz, 1 H) 7.35 (d, .7=1.01 Hz, 1 H) 5.88 (s, 1 H) 5.04 (quin, =6.57 Hz, 1 H) 4.35 (s, 1 H) 4.34 (s, 1 H) 2.43 (s, 3 H) 2.23 (s, 3 H) 2.12 (s, 3 H) 1.46 (s, 3 H) 1.44 (s, 3 H); LC-MS (ES) m/z = 378.3[M+H]+
Example 91
l-cyclopentyl-6-(cyclopropylsulfonyl)-N-((4,6-dimethyl-2-oxo-l,2-dihydropyridin-3-yl)methyl)-lH- indazole-4-carboxamide
Figure imgf000116_0001
To a 10-mL microwave tube were added 6-bromo- l-cyclopentyl-N-[(4,6-dimethyl-2-oxo-l,2-dihydro- 3-pyridinyl)methyl]-lH-indazole-4-carboxamide (70 mg, 0.158 mmol), cyclopropanesulfinic acid (40.8 mg, 0.316 mmol), dimethyl sulfoxide (3 mL), Ν,Ν'-dim ethyl- 1,2-ethanediamine methyl[2- (methylamino) ethyl] amine (55.7 mg, 0.632 mmol), and copper(II) trifluoromethanesulfonate (57.1 mg, 0.158 mmol), and the mixture was degassed for 5 min by bubbling nitrogen. The tube was sealed and the mixture irradiated at 120 °C (microwave) for 3h. The mixture was filtered and the DMSO solution was purified using reverse-phase HPLC to give 45 mg of product. The product was collected as an off-white solid. H NMR (400 MHz, DMSO-<f6) δ ppm 1.00 - 1.12 (m, 2 H), 1.15 - 1.31 (m, 2 H), 1.67 - 1.79 (m, 2 H), 1.84 - 1.94 (m, 2 H), 1.96 - 2.08 (m, 2 H), 2.12 - 2.28 (m, 8 H), 2.93 - 3.05 (m, 1 H), 4.38 (d, J= 5.05 Hz, 2 H), 5.43 (quin, J= 7.01 Hz, 1 H), 5.90 (s, 1 H), 8.01 (d, J=1.26 Hz, 1 H), 8.44 (s, 1 H), 8.52 (s, 1 H), 8.83 (t, J= 4.93 Hz, 1 H) ,1 1.57 (s, 1 H); LCMS: (M+H)+=469.0 Example 92
6-(cyclopropylsulfonyl)-N-((4-ethyl-6-methyl-2-oxo-l,2-dihydropyridin-3-yl)methy])-l-isopropyl-lH- indazole-4-carboxamide
Figure imgf000116_0002
The title compound was prepared from 6-bromo-N-[(4-ethyl-6-methyl-2-oxo-l,2-dihydro-3- pyridinyl)methyl]-l-(l-methylethyl)-lH-indazole-4-carboxamide (70 mg, 0.162 mmol) and the sodium salt of cyclopropanesulfmic acid (41.9 mg, 0.325 mmol) in the same manner as described for example 91. The product was collected an off-white solid (52 mg). ¾ NMR (400 MHz, DMSO-c 6) δ ppm 1.02 - 1.16 (m, 5 H), 1.19 - 1.27 (m, 2 H), 1.50 (d, J = 6.57 Hz, 6 H), 2.15 (s, 3 H), 2.58 (m, 2 H), 2.97 (m, 1 H), 4.41 (d, J= 4.80 Hz, 2 H), 5.26 (m, 1 H), 5.94 (s, 1 H), 8.01 (d, J= 1.26 Hz, 1 H), 8.43 (s, 1 H), 8.53 (s, 1 H), 8.83 (m, 1 H), 1 1.58 (s, 1 H); LCMS: (M+H)+=457.0
Example 93
N-((4-ethyl-6-methyl-2-oxo-l,2-dihydropyridin-3-yl)methyl)-l-isopropyl-6-(methylsulfonyl)-lH- indazole-4-carboxamide
Figure imgf000117_0001
The title compound was prepared from 6-bromo-N-[(4-ethyl-6-methyl-2-oxo-l,2-dihydro-3- pyridinyl)methyl]-l-(l-methylethyl)-lH-indazole-4-carboxamide (70 mg, 0.162 mmol) and the sodium salt of methanesulfinic acid (33.5 mg, 0.325 mmol), in the same manner as described for example 91. The product was collected as a white solid (21 mg) H NMR (400 MHz, DMSO-< ) δ ppm 1.06 - 1.17 (m, 3 H), 1.47 - 1.56 (m, 6 H), 2.15 (s, 3 H) ,2.58 (q, J= 7.58 Hz, 2 H), 3.03 (s, 3 H), 4.33 - 4.47 (m, 2 H), 5.17 - 5.30 (m, 1 H), 5.94 (s, 1 H), 8.01 (d, J = 1.26 Hz, 1 H), 8.45 (m, 1 H), 8.53 (s, 1 H), 8.78 (t, J= 4.93 Hz, 1 H); LCMS: (M+H)+=431.1
Example 94
A^-[(4,6-dimethyl-2-oxo-l,2-dihydro-3-pyridinyl)methyl]-l-(l-methylethyl)-6-(methylsulfonyl)-lfi- indazole-4-carboxamide
Figure imgf000117_0002
To a 10 mL microwave vial containing a stirring mixture of L-valine (8.42 mg, 0.072 mmol), NaOH (0.024 mL, 0.072 mmol), and DMSO (1.6 mL) were added sodium methanesulfinate (0.065 g, 0.539 mmol) copper(I) iodide (6.85 mg, 0.036 mmol) and l-Ethyl-3-methylimidazoliym bromide (0.014 g, 0.072 mmol) After stirring for 15 min, 6-bromo-N-[(4,6-dimethyl-2-oxo-l,2-dihydro-3-pyridinyl)methyl]-l-(l- methylethyl)-lH-indazole-4-carboxamide (0.15 g, 0.359 mmol) was added. The reaction vial was sealed anc the mixture was stirred with heating at 90 °C (heat block) for 18h. The contents were then irradiatec (microwave) at 130 °C for 2hr. After cooling to RT, the reaction mixture was poured onto 50 mL of water anc stirred. The contents were extracted with hot 30%THF/EtOAc (2x) (need to heat to break up emulsion). The combined organic layers were washed with brine, dried over MgSOzi, filtered, and concentrated in vacuo. Τΐκ crude product was purified by silica gel chromatography (eluent: 5-75% chloroform (containing 10% 2M ammonia in methanol) and dichloromethane. The isolated product was concentrated from DCM and MTBE. and dried under hi vacuum. The title compound was collected as 75 mg (49%); !H NMR (400 MHz, DMSO- d6) δ ppm 1.50 (d, J=6.57 Hz, 6 H) 2.13 (s, 3 H) 2.22 (s, 3 H) 3.30 (s, 3 H) 4.38 (d, J=4.80 Hz, 2 H) 5.22 (quin, J=6.57 Hz, 1 H) 5.90 (s, 1 H) 8.02 (d, J=1.01 Hz, 1 H) 8.45 (s, 1 H) 8.52 (s, 1 H) 8.77 (t, J=4.80 Hz, 1 H) 11.56 (s, 1 H); LCMS: (M+H)+= 417.1.
Methyl l-(l-methylethyl)-6-nitro-l//-indazole-4-carboxylate
Figure imgf000118_0001
Sodium hydride (1.142 g, 45.2 mmol) was added to Ν,Ν-Dimethylformamide (DMF) (100 mL) which was cooled in a ice water bath. A DMSO solution (20 mL) of methyl 6-nitro-lH-indazole-4- carboxylate (5 g, 22.61 mmol) was then added dropwise. After 15 minutes was added 2- bromopropane (4.25 mL, 45.2 mmol) dropwise and the contents were stirred at room temperature for 16 hours. Next added iodomethane (2.83 mL, 45.2 mmol) and let stir at room temperature for 3 hours followed by heating at 50 °C for 3 hours. The solvent was removed in vacuo. The residue was purified via silica gel chromatography (eluent: 0% to 25% EtOAc:Hex) to afford the product as 1.39 g (23%). ¾ NMR (400 MHz, CHLOROFORM-d) δ ppm 4.07 (s, 3 H), 8.61 - 8.63 (m, 1 H), 8.64 (s, 1 H), 8.73 (d, J=2.02 Hz, 1 H) ' LCMS(ES) [M+H]+264.0
Methyl 6-amino-l-(l-methylethyl)-l/ -indazole-4-carboxylate
Figure imgf000119_0001
Methyl l-(l-methylethyl)-6-nitro-lH-indazole-4-carboxylate (1.39 g, 5.28 mmol) was dissolved in ethanol (70 mL) and hydrogenated using an H-Cube instrument (full H2 mode and 10% Pd/C). The solvent was removed in vacuo and the residue was purified via silica gel chromatography (eluent: 0% to 40% gradient; EtOAc:Hex). The product was obtained as 1.05 g (85%). *H NMR (400 MHz, CHLOROFORM-d) δ ppm 1.58 (d, J=6.57 Hz, 6 H), 4.00 (s, 5 H), 4.70 (dt, 7=13.39, 6.69 Hz, 1 H), 6.80 (s, 1 H), 7.37 (d, J=2.02 Hz, 1 H), 8.30 (s, 1 H). LCMS(ES) [M+H]+234.0
Methyl 6-(chlorosulfonyl)-l-(l-methylethyl)-l/i-indazole-4-carboxylate
Figure imgf000119_0002
Methyl 6-amino-l-(l-methylethyl)-lH-indazole-4-carboxylate (500 mg, 2.143 mmol) was dissolved in cone, hydrochloric acid (5 mL) and cooled in an ice water bath. A solution of sodium nitrite (155 mg, 2.251 mmol) in 2 mL of water was then added dropwise, and the contents were stirred for 90 min. The contents were added portion- wise to a solution of ca. 5 mL of S02, copper(II) chloride (303 mg, 2.251 mmol) and acetic acid (20 mL). The contents were stirred at room temperature for 15 h, and then concentrated in vacuo. The residue was suspended in 100 mL DCM, washed with water (2 x 50 mL), dried (MgSC ), filtered and concentrated in vacuo. The residue was purified via silica gel chromatography (eluent : 0% to 10 gradient EtOAc:Hex). The product was collected as 510 mg (75%). ¾ NMR (400 MHz, CHLOROFORM-d) δ ppm 1.68 (d, 6 H), 4.09 (s, 3 H), 5.01 (dt, J=13.33, 6.60 Hz, 1 H), 8.40 (s, 1 H), 8.51 (d, .7=1.52 Hz, 1 H), 8.70 (s, 1 H). LCMS(ES) [M+H]+317.0
Methyl l-(l-methylethyl)-6-(4-morpholinylsulfonyl)-l/i-indazole-4-carboxylate
Figure imgf000120_0001
A solution of morpholine (0.030 n L, 0.347 mmol), triethylamine (0.055 n L, 0.395 mmol) and dichloromethane (DCM) (20 mL) were cooled in an ice-water bath. A DCM solution (5 mL) of methyl 6-(chlorosulfonyl)-l-(l -methylethyl)- lH-indazole-4-carboxylate (100 mg, 0.316 mmol) was added dropwise. The contents were stirred at room temperature for 10 min. The contents were then washed with water, brine, dried over MgS04, filtered, and concentrated in vacuo. The product was collected as 90 mg (77%). Ή NMR (400 MHz, CHLOROFORM-d) δ ppm 1.67 (d, 6 H), 2.99 - 3.19 (m, 4 H), 3.75 - 3.86 (m, 4 H), 4.08 (s, 3 H), 4.98 (ddd, J=13.26, 6.82, 6.69 Hz, 1 H), 8.14 (s, 1 H), 8.22 (d, J=1.26 Hz, 1 H), 8.65 (s, 1 H); LCMS(ES) [M+H]+368.0
The following intermediates were prepared using the general procedures outlined for the above compound from methyl 6-(chlorosulfonyl)- 1 -( 1 -methylethyl)- 1 H-indazole-4-carboxylate and the appropriate amine.
Methyl 6-[(cyclopropylamino)sulfonyl]-l-(l-methylethyl)-l /-indazole-4-carboxylate
Figure imgf000120_0002
!H NMR (400 MHz, CHLOROFORM-d) δ ppm 0.57 - 0.76 (m, 4 H), 1.66 (d, .7=6.82 Hz, 6 H), 2.22 2.39 (m, 1 H), 4.07 (s, 3 H), 4.89 - 5.17 (m, 2 H), 8.33 (s, 1 H), 8.36 (d, .7=1.26 Hz, 1 H), 8.63 (s, 1 H); LCMS(ES) [M+H]+324.4
Methyl l-(l-methylethyl)-6-(l-pyrrolidinylsulfonyl)-l -indazole-4-carboxylate
Figure imgf000120_0003
!H NMR (400 MHz, CHLOROFORM-d) δ ppm 1.62 - 1.74 (m, 6 H), 1.85 (ddd, .7=6.57, 3.66, 3.41 Hz, 4 H), 3.37 (t, .7=6.69 Hz, 4 H), 5.01 (spt, .7=6.65 Hz, 1 H), 8.30 (s, 1 H), 8.42 (d, .7=1.26 Hz, 1 H), 8.69 (s, 1 H); LCMS(ES) [M+H]+338.3
Methyl 6-({[3-(dimethylamino)propyl]amino}sulfonyl)-l-(l-methylethyl)-l/ -indazole-4- carboxylate
Figure imgf000121_0001
*H NMR (400 MHz, CHLOROFORM-d) δ ppm 1.60 - 1.67 (m, 6 H), 1.74 (quin, .7=5.81 Hz, 2 H), 2.37 (s, 6 H), 2.55 (t, .7=5.81 Hz, 2 H), 3.07 - 3.16 (m, 2 H), 4.04 (s, 3 H), 4.98 (dt, .7=13.33, 6.60 Hz, 1 H), 8.30 (d, .7=1.77 Hz, 2 H), 8.59 (s, 1 H). LCMS(ES) [M+H]+383.1
Methyl l-(l-methylethyl)-6-[(phenylamino)sulfonyl]-l//-indazole-4-carboxylate
Figure imgf000121_0002
¾ NMR (400 MHz, CHLOROFORM-d) δ ppm 1.53 (d, 6 H), 4.01 (s, 3 H), 4.81 (dt, .7=13.33, 6.60 Hz, 1 H), 6.94 (s, 1 H), 7.02 - 7.16 (m, 3 H), 7.20 - 7.26 (m, 2 H), 8.04 (s, 1 H), 8.32 (d, .7=1.26 Hz, 1 H), 8.55 (s, 1 H). LCMS(ES) [M+H]+ 373.9 l-(l-methylethyl)-6-(4-morpholinylsulfonyl)-l/i-indazole-4-carboxylic acid
Figure imgf000121_0003
Sodium hydroxide (0.980 mL, 0.980 mmol) was added to a solution of methyl l-(l-methylethyl)-6-(4- mo holinylsulfonyl)-lH-indazole-4-carbo ylate (90 mg, 0.245 mmol) in ethanol (30 mL) and heated at reflux for 1 hour. The solvent was removed in vacuo and the residue suspended in water (20 mL). The contents were acidifed by addition of acetic acid, and extracted with DCM (4 x 30 mL). The combined organic layers were washed with water, brine, dried over MgS04, filtered, and concentrated in vacuo. The product was collected as 90 mg. ¾ NMR (400 MHz, CHLOROFORM-d) δ ppm 1.63 - 1.78 (m, 6 H), 3.02 - 3.24 (m, 4 H), 3.67 - 3.88 (m, 4 H), 5.00 (dt, .7=13.20, 6.66 Hz, 1 H), 8.21 (s, 1 H), 8.31 (s, 1 H), 8.71 (s, 1 H), LCMS(ES) [M+H]+354.2
The following intermediates were prepared using the general procedure outlined for the above compound.
6-[(cyclopropylamino)sulfonyl]-l-(l-methylethyl)-l/i-indazole-4-carboxylic acid
Figure imgf000122_0001
H NMR (400 MHz, CHLOROFORM-d) δ ppm 0.62 - 0.72 (m, 4 H), 1.67 (d, .7=6.57 Hz, 6 H), 2.27 2.44 (m, 1 H), 5.02 (spt, .7=6.61 Hz, 1 H), 5.37 (s, 1 H), 8.41 (s, 1 H), 8.44 (d, .7=1.52 Hz, 1 H), 8.65 (s, 1 H); LCMS(ES) [M+H]+324.4 l-(l-methylethyl)-6-(l-pyrrolidinylsulfonyl)-l/7-indazole-4-carboxylic acid
Figure imgf000122_0002
l¥L NMR (400 MHz, CHLOROFORM-d) δ ppm 1.62 - 1.74 (m, 6 H), 1.85 (ddd, J=6.57, 3.66, 3.41 Hz, 4 H), 3.37 (t, .7=6.69 Hz, 4 H), 5.01 (spt, .7=6.65 Hz, 1 H), 8.30 (s, 1 H), 8.42 (d, .7=1.26 Hz, 1 H), 8.69 (s, 1 H); LCMS(ES) [M+H]+338.3 6-({[3-(dimethylamino)propyl]amino}sulfonyl)-l-(l-methylethyl)-l^-indazole-4-carboxylic acid
Figure imgf000122_0003
!H NMR (400 MHz, DMSO-d6) δ ppm 1.50 (d, 6 H), 1.62 (qd, .7=7.07, 6.82 Hz, 2 H), 2.30 (s, 6 H), 2.82 (t, .7=6.82 Hz, 2 H), 5.19 (dt, J=13.14, 6.57 Hz, 1 H), 7.98 (d, .7=8.59 Hz, 1 H), 8.13 (d, .7=1.26 Hz, 1 H), 8.32 (s, 1 H), 8.55 (s, 1 H); LCMS(ES) [M+H]+ 368.9 l-(l-methylethyl)-6-[(phenylamino)sulfonyl]-l -indazole-4-carboxylic acid
Figure imgf000123_0001
*H NMR (400 MHz, MeOD) δ ppm 1.52 (d, J=6.82 Hz, 6 H), 4.98 (ddd, J=13.26, 6.82, 6.69 Hz, 1 H), 7.00 - 7.16 (m, 3 H), 7.18 - 7.28 (m, 2 H), 8.15 (s, 1 H), 8.25 (d, J=1.52 Hz, 1 H), 8.52 (s, 1 H);
LCMS(ES) [M+H]+ 360.1
Example 95
N-[(4,6-dimethyl-2-oxo-l,2-dihydro-3-pyridinyl)raethyl]-l-(l-methylethyl)-6-(4- morpholinylsulfonyl)-lH-indazole-4-carboxamide
Figure imgf000123_0002
l-(l-methylethyl)-6-(4-morpholinylsulfonyl)-lH-indazole-4-carboxylic acid (90 mg, 0.255 mmol), 3- (aminomethyl)-4,6-dimethyl-2(lH)-pyridinone (62.5 mg, 0.331 mmol), l-hydroxy-7- azabenzotriazole (69.3 mg, 0.509 mmol), EDC (98 mg, 0.509 mmol) and N-methylmorpholine (0.112 mL, 1.019 mmol) were added to Dimethyl Sulfoxide (DMSO) (10 mL) and stirred at room temperature for 16 hours. Next added 40 mL of water and stirred for 10 minutes. The contents were extracted with DCM (5 x 30 mL), washed with water, dried (MgSO^, filtered, and concentrated in vacuo. The residue was purified via chromatography (eluent : 0% to 100% gradient Hex:EtOAc then 0 to 10% MeOH:EtOAc). The product was collected as 110 mg (88%). ¾ NMR (400 MHz, DMSO- d6) δ ppm 1.50 (d, 6 H), 2.13 (s, 3 H), 2.23 (s, 3 H), 2.89 - 3.05 (m, 4 H), 3.54 - 3.71 (m, 4 H), 4.38 (d, .7=4.80 Hz, 2 H), 5.27 (dt, .7=13.07, 6.47 Hz, 1 H), 5.90 (s, 1 H), 7.80 (d, .7=1.01 Hz, 1 H), 8.29 (s, 1 H), 8.52 (s, 1 H), 8.88 (t, J=5.05 Hz, 1 H), 1 1.56 (br. s., 1 H); LCMS(ES) [M+H]+488.3
Examples 96-99 were prepared using the general procedures outlined for the above compound.
Example 96
6-[(cyclopropylamino)sulfonyl]-N-[(4,6-dimethyl-2-oxo-l,2-dihydro-3-pyridinyl)methyl]-l-(l- methylethyl)-l /-indazole-4-carboxamide
Figure imgf000124_0001
H NM (400 MHz, CHLOROFORM- d) δ ppm 0.42 - 0.65 (m, 4 H), 1.61 (d, .7=6.57 Hz, 6 H), 2.19 (dt, .7=6.63, 3.38 Hz, 1 H), 2.25 (s, 3 H), 2.45 (s, 3 H), 4.63 (d, .7=5.56 Hz, 2 H), 4.95 (quin, .7=6.63 Hz, 1 H), 6.02 (s, 1 H), 6.32 (br. s., 1 H), 8.17 (s, 1 H), 8.20 (s, 1 H), 8.25 (t, .7=5.18 Hz, 1 H), 8.60 (s, 1 H), 1 1.46 (br. s., 1 H); LCMS(ES) [M+H]+ 458.1
Example 97
Ar-[(4,6-dimethyl-2-oxo-l,2-dihydro-3-pyridinyl)methyl]-l-(l-methylethyl)-6-(l- pyrrolidinylsulfonyl)-l//-indazole-4-carboxamide
Figure imgf000124_0002
H NMR (400 MHz, DMSO-d6) δ ppm 1.49 (d, 6 H), 1.58 - 1.71 (m, 4 H), 2.13 (s, 3 H), 2.23 (s, 3 H), 3.18 - 3.29 (m, 4 H), 4.38 (d, J=5.05 Hz, 2 H), 5.28 (quin, J=6.57 Hz, 1 H), 5.90 (s, 1 H), 7.89 (d, J=1.01 Hz, 1 H), 8.33 (s, 1 H), 8.51 (s, 1 H), 8.90 (t, J=4.93 Hz, 1 H), 1 1.56 (s, 1 H); LCMS(ES) [M+H]+ 472.4
Example 98
6-({ [3-(dimethylamino)propyl] amino} sulfonyl)-N- [(4,6-dimethyl-2-oxo-l ,2-dihydro-3- pyridiny])methyl]-l-(l-methylethyl)-l/i-indazole-4-carboxamide
Figure imgf000125_0001
!H NMR (400 MHz, DMSO-d6) δ ppm 1.37 - 1.54 (m, 8 H), 1.97 (s, 6 H), 2.05 - 2.17 (m, 5 H), 2.22 (s, 3 H), 2.78 (t, J=6.95 Hz, 2 H), 4.36 (d, .7=4.55 Hz, 2 H), 5.16 (quin, J=6.57 Hz, 1 H), 5.89 (s, 1 H), 7.87 (s, 1 H), 8.25 (s, 1 H), 8.43 (s, 1 H), 8.53 (s, 0 H), 8.73 (br. s., 1 H); LCMS(ES) [M+H]+ 502.9
Example 99
N-((4,6-dimethyl-2-oxo-l,2-dihydropyridin-3-yl)methyl)-l-isopropyl-6-((4-methylpiperazin-l-
Figure imgf000125_0002
!H NMR (400 MHz, CHLOROFORM-d) δ ppm 1.62 (d, J=6.82 Hz, 6 H), 2.27 (d, J=6.32 Hz, 6 H), 2.43 (s, 3 H), 2.52 (br. s., 4 H), 3.14 (br. s., 4 H), 4.65 (d, J=5.81 Hz, 2 H), 4.93 (dq, J=6.82, 6.65 Hz, 1 H), 5.99 (s, 1 H), 7.86 (s, 1 H), 8.01 (s, 1 H), 8.06 - 8.16 (m, 1 H), 8.56 (s, 1 H), 12.02 (br. s., 1 H); LCMS(ES) [M+H]+ 501.0.
Example 100
6-bromo-N-((4-(sec-butyl)-6-methyl-2-oxo-l,2-dihydropyridin-3-yl)methyl)-l-isopropyl 1H- indazole-4-carboxamide
Figure imgf000126_0001
The title compound was prepared in the same manner as described for example 3 (step c) from 6- bromo-l-(l-methylethyl)-lH-indazole-4-carboxylic acid (0.50 g, 1.77 mmol) and 3-(aminomethyl)-4- (sec-butyl)-6-methylpyridin-2(lH)-one (0.446 g, 2.296 mmol). The product was collected as 0.81 g (98%). ¾ NMR (400 MHz, DMSO-d6) δ ppm 1 1.53 (s, 1 H), 8.62 (t, J=4.7 Hz, 1 H), 8.37 (s, 1 H), 8.20 (s, 1 H), 7.70 (d, J=1.5 Hz, 1 H), 5.96 (s, 1 H), 5.05 (quin, J=6.6 Hz, 1 H), 4.41 (d, J=4.3 Hz, 2 H), 2.95 (q, J=7.1 Hz, 1 H), 2.15 (s, 3 H), 1.48 (m, 8 H), 1.07 (d, J=6.6 Hz, 3 H), 0.74 (t, J=7.3 Hz, 3 H). LC-MS(ES) [M+H]+ 459.2. Example 101
6-Bromo-l-(l-methylethyl)-N-[(6-methyl-2-oxo-4^ropyl-l,2-dihydro-3-pyridinyl)methyl]-lH- indazole-4-carboxamide
Figure imgf000126_0002
6-Bromo-l-(l-methylethyl)-lH-indazole-4-carboxylic acid (80 mg, 0.28 mmol), 3- (aminomethyl)-6-methyl-4-propyl-2(lH)-pyridinone (112 mg, 0.38 mmol) and l-hydroxy-7- azabenzotriazole (57.7 mg, 0.42 mmol) were stirred in 3 mL of DMSO for 10 min under nitrogen. N- Methylmorpholine (0.12 ml, 1.13 mmol) was added along with EDC (81 mg, 0.42 mmol) and the mixture was stirred at room temperature overnight under nitrogen. Ice-water was added and solids crashed out. Then 10% K2C03 was added to adjust the pH to about 8-9. Then the reaction was stirred at room temperature for 10 min and let stand for 10 min. Solids were filtered off, dissolved in DMF and water was added. Solids that precipitated out were filtered off, air-dried for 15 min and dried in vacuum oven for 2 h to give the title compound (94 mg, 73%). ¾ NMR (400 MHz, DMSO-J6) δ ppm 11.54 (s, 1 H) 8.62 (t, J=4.93 Hz, 1 H) 8.37 (s, 1 H) 8.20 (s, 1 H) 7.69 (d, J=1.26 Hz, 1 H) 5.91 (s, 1 H) 5.06 (dt, J=13.14, 6.57 Hz, 1 H) 4.36 (d, J=4.80 Hz, 2 H) 2.14 (s, 3 H) 1.48 - 1.56 (m, 2 H) 1.47 (s, 3 H) 1.45 (s, 3 H) 0.89 (t, J=7.33 Hz, 3 H); MS(ES) [M+H]+ 445.1, 446.9. Example 102
6-Bromo-l-(l-methylethyl)- V-{[6-methyl-4-(l-methylethyl)-2-oxo-l,2-dihydro-3- pyridinyl]methyl}-l/i-i
Figure imgf000127_0001
The title compound was prepared in the same manner as example 101 from 6-Bromo-l-(l- methylethyl)-lH-indazole-4-carboxylic acid and 3-(ammomethyl)-6-methyl-4-(l-methylethyl)2(lH)- pyridinone. :H NMR (400 MHz, DMSO-c/e) δ ppm 11.53 (s, 1 H) 8.63 (t, J=4.80 Hz, 1 H) 8.37 (s, 1 H) 8.20 (s, 1 H) 7.69 (d, J=1.26 Hz, 1 H) 6.02 (s, 1 H) 5.05 (quin, J=6.57 Hz, 1 H) 4.41 (d, J=4.80 Hz, 2 H) 3.21 (quin, J=6.76 Hz, 1 H) 2.16 (s, 3 H) 1.47 (s, 3 H) 1.45 (s, 3 H) 1.11 (s, 3 H) 1.09 (s, 3 H); MS(ES) [M+H]+ 445.1, 446.9.
Example 103
N- [(4,6-Dimethyl-2-oxo- 1 ,2-dihydro-3-pyridinyl)methyl]- 1 -( 1 -methylethyl)-6-phenyl- 1 H-indazole-4- carboxamide
Figure imgf000127_0002
To a mixture of 6-bromo-N-[(4,6-dimethyl-2-oxo-l,2-dihydro-3-pyridinyl)methyl]-l-(l- methylethyl)-lH-indazole-4-carboxamide (0.15g, 0.359 mmol), phenylboronic acid (0.088 g, 0.719 mmol) and potassium phosphate (tribasic) (0.229 g, 1.078 mmol) was added 1,4-dioxane (3 mL) and water (0.75 mL). The suspension was degassed with N2 for 10 min, at which time PdCl2(dppf)-
CH2C12 (0.044 g, 0.054 mmol) was added and the sealed reaction heated at 100 °C overnight. Diluted reaction with EtOAc and filtered through Celite, washing with EtOAc and concentrating in vacuo to a residue that was dissolve in a minimum amount of DCM. Purification by silica gel chromatography (12 gram Isco GOLD silica column; Gradient B: 5-80%; A: dichloromethane, B: 10% (2 M ammonia in methanol) in chloroform) gave N-[(4,6-dimethyl-2-oxo-l,2-dihydro-3-pyridinyl)methyl]- l-(l- methylethyl)-6-phenyl-lH-indazole-4-carboxamide (0.105g, 0.248 mmol, 69.1 % yield). ¾ NMR (400 MHz, DMSO-t 6) δ ppm 1.50 (d, J=6.57 Hz, 6 H) 2.12 (s, 3 H) 2.22 (s, 3 H) 4.39 (d, .7=4.80 Hz, 2 H) 5.10 - 5.24 (m, 1 H) 5.89 (s, 1 H) 7.37 - 7.46 (m, 1 H) 7.48 - 7.55 (m, 2 H) 7.82 - 7.93 (m, 3 H) 8.12 (s, 1 H) 8.38 (s, 1 H) 8.66 (t, J=4.93 Hz, 1 H) 11.54 (s, 1 H). MS(ES) [M+H]+ 415.1.
Example 104
N-[(4,6-Dimethyl-2-oxo- 1 ,2-dihydro-3 -pyridinyl)methyl] - 1 -( 1 -memylethyl)-6-(4-pyridinyl)-l/i- indazole-4-carboxamide
Figure imgf000128_0001
To a mixture of 6-bromo-N-[(4,6-dimethyl-2-oxo-l,2-dihydro-3-pyridinyl)methyl]-l-(l- methylethyl)-lH-indazole-4-carboxamide (0.15g, 0.359 mmol),4-pyridinylboronic acid (0.088 g, 0.647 mmol) and potassium phosphate (tribasic) (0.229 g, 1.078 mmol) was added 1,4-dioxane (3 mL) and water (0.75 mL). The suspension was degassed with N2 for 10 min, at which time
PdCl2(dppf)-CH2Cl2 (0.044 g, 0.054 mmol) was added and the sealed reaction heated at 100 °C overnight (reaction got very dark). Diluted reaction with EtOAc and filtered through Celite, washing with EtOAc and concentrated in vacuo to a residue that was dissolved in a minimum amount of DCM. Purification by silica gel chromatography (12 gram Isco GOLD silica column; Gradient B: 5-90%; A: dichloromethane, B: 10% (2 M ammonia in methanol) in chloroform) gave N-[(4,6-dimethyl-2-oxo- l,2-dihydro-3-pyridinyl)methyl]-l-(l-methylethyl)-6-(4-pyridinyl)-lH-indazole-4-carboxamide (1 12 mg, 0.264 mmol, 73.5 % yield). !H NMR (400 MHz, DMSO-c 6) δ ppm 1.51 (d, J=6.57 Hz, 6 H) 2.12 (s, 3 H) 2.22 (s, 3 H) 4.40 (d, J=4.80 Hz, 2 H) 5.15 - 5.27 (m, 1 H) 5.89 (s, 1 H) 7.90 - 8.00 (m, 3 H) 8.34 (s, 1 H) 8.43 (s, 1 H) 8.65 - 8.75 (m, 3 H) 11.55 (s, 1 H). MS(ES) [M+H]+ 416.1.
Example 105
N-[(4,6-Dimethyl-2-oxo-l,2-dihydro-3-pyridinyl)methyl]-6- {4-[(methylamino)sulfonyl]ph(
methylethyl)-l /-indazole-4-carboxamide
Figure imgf000129_0001
In a 25 mL sealable tube under nitrogen were combined 6-bromo-N-[(4,6-dimethyl-2-oxo-
I, 2-dihydro-3-pyridinyl)methyl]-l-(l-methylethyl)-lH-indazole-4-carboxamide (100 mg, 0.24 mmol), {4-[(methylamino)sulfonyl]phenyl}boronic acid (77 mg, 0.36 mmol) in DME/water (3 mL:lmL). PdCl2(dppf)-CH2Cl2 (9.8 mg, 0.012 mmol) was added and the resulting mixture was degassed with nitrogen for 10 min. Sodium bicarbonate (60.4 mg, 0.72 mmol) was added, the vessel was sealed, and the insoluble mixture was heated in a microwave at 140 °C for 30 min. EtOAc was added and the solution was filtered thru Celite and evaporated. DCM/MeOH (1 : 1) was added, it was pre-absorbed on silica gel and purified by Si02 chromatography (eluent: gradient 100% DCM to 90: 10:1 DCM/MeOH/NH4OH). Fractions were evaporated. EtOAc was added along with some hexanes, it was sonicated and the solids that precipitated were filtered, washed with DCM and dried to afford the title compound (67 mg, 54%) as a beige solid. H NMR (400 MHz, DMSO-< ) δ ppm
I I .54 (s, 1 H) 9.87 (br. s., 1 H) 8.62 (t, J=4.80 Hz, 1 H) 8.37 (s, 1 H) 8.07 (s, 1 H) 7.82 - 7.86 (m, 3 H) 7.35 (s, 1 H) 7.32 (s, 1 H) 5.89 (s, 1 H) 5.15 (quin, J=6.63 Hz, 1 H) 4.39 (d, J=4.80 Hz, 2 H) 3.04 (s, 3 H) 2.22 (s, 3 H) 2.12 (s, 3 H) 1.51 (s, 3 H) 1.49 (s, 3 H). MS(ES) [M+H]+ 507.9.
Example 106
V-[(4,6-Dimethyl-2-oxo-l,2-dihydro-3-pyridinyl)methyl]-l-(l-methylethyl)-6-[6-(4-methyl-l- piperazinyl)-3-pyridinyl]-l/ -indazole-4-carboxamide
Figure imgf000129_0002
In a 25 mL sealable tube under nitrogen were combined 6-bromo-N-[(4,6-dimethyl-2-oxo- l,2-dihydro-3-pyridinyl)methyl]-l-(l-methylethyl)-lH-indazole-4-carboxamide (100 mg, 0.24 mmol), l-methyl-4-[5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-2-pyridinyl]piperazine (109 mg, 0.36 mmol) in DME/water (3 mL: lmL). PdCl2(dppf)-CH2Cl2 (9.8 mg, 0.012 mmol) was added and the resulting mixture was degassed with nitrogen for 10 min. Sodium bicarbonate (60.4 mg, 0.72 mmol) was added, the vessel was sealed, and the insoluble mixture was heated in a microwave at 130 °C for 30 min. Solvent was evaporated, DCM was added and it was purified by Si02 chromatography (eluent: gradient 100% DCM to 80:20:2 DCM/MeOH/NH4OH). Further purification by reversed- phase HPLC (C18, 15% to 80% CH3CN in water with 0.1% TFA, 12 minute gradient) afforded the TFA salt. The CH3CN was evaporated and the mixture was diluted with saturated sodium bicarbonate and EtOAc. The organic layer was separated and the aqueous layer was further extracted with DCM and DCM/isopropanol (80:20). The combined organic layers were washed with water, dried (MgS04), filtered and concentrated. EtOAc was added along with some hexanes, it was sonicated and the solids that precipitated were filtered and dried to afford the title compound (71 mg, 56%) as a white solid. ¾ NMR (400 MHz, DMSO-J6) δ ppm 1 1.55 (br. s., 1 H) 8.66 (d, =2.27 Hz, 1 H) 8.61 (t, J=4.93 Hz, 1 H) 8.36 (s, 1 H) 8.04 - 8.09 (m, 2 H) 7.84 (d, J=1.26 Hz, 1 H) 6.96 (d, J=8.84 Hz, 1 H) 5.89 (s, 1 H) 5.14 (quin, J=6.57 Hz, 1 H) 4.39 (d, J=5.05 Hz, 2 H) 3.52 - 3.60 (m, 4 H) 2.38 - 2.46 (m, 4 H) 2.22 (d, .7=4.55 Hz, 6 H) 2.13 (s, 3 H) 1.50 (s, 3 H) 1.49 (s, 3 H). MS(ES) [M+H]+ 514.5.
Example 107
A^-[(4,6-Dimethyl-2-oxo- l ,2-dihydro-3-pyridinyl)methyl]-l-(l -methylethyl)-6-(2-oxo-2,3-dihydro- l /-be arboxamide
Figure imgf000130_0001
In a 25 mL sealable tube under nitrogen were combined 6-bromo-N-[(4,6-dimethyl-2-oxo- l,2-dihydro-3-pyridinyl)methyl]- l -(l -methylethyl)- lH-indazole-4-carboxamide (100 mg, 0.24 mmol), 5-(4,4,5,5-tetramethyl- 1 ,3,2-dioxaborolan-2-yl)- 1 ,3-dihydro-2H-benzimidazol-2-one (93 mg, 0.36 mmol) in dioxane/water (3 mL: 1 mL). PdC /dppfj-CHiC . (9.8 mg, 0.012 mmol) was added and the resulting mixture was degassed with nitrogen for 10 min. Sodium bicarbonate (60.4 mg, 0.72 mmol) was added, the vessel was sealed, and the insoluble mixture was heated in a microwave at 130 °C for 20 min. DCM/MeOH (1 : 1) was added, it was pre-absorbed on silica gel and purified by S1O2 chromatography (eluent: gradient 100% DCM to 80:20:2 DCM/MeOH/NH4OH). Fractions were evaporated. EtOH was added, it was sonicated and the solids that precipitated were filtered, washed with EtOH and DCM and dried to afford the title compound (43 mg, 37%) as a beige solid. l NMR (400 MHz, DMSO-c/s) 5ppm 1 1.53 (s, 1 H) 10.78 (s, 1 H) 10.74 (s, 1 H) 8.64 (t, J=4.80 Hz, 1 H) 8.35 (s, 1 H) 8.02 (s, 1 H) 7.81 (s, 1 H) 7.44 (dd, J=8.08, 1.77 Hz, 1 H) 7.36 - 7.39 (m, 1 H) 7.03 (d, J=8.08 Hz, 1 H) 5.89 (s, 1 H) 5.16 (quin, J=6.57 Hz, 1 H) 4.39 (d, J=4.80 Hz, 2 H) 2.21 (s, 3 H) 2.13 (s, 3 H) 1.50 (s, 3 H) 1.49 (s, 3 H). MS(ES) [M+H]+ 471.3
Example 108
6-[6-(Acetylamino)-3-pyridinyl]-W-[(4,6-dimethyl-2-oxo- l,2-dihydro-3-pyridinyl)methyl]- 1 -(1 - m de
Figure imgf000131_0001
In a 25 mL sealable tube under nitrogen were combined 6-bromo-N-[(4,6-dimethyl-2-oxo- l,2-dihydro-3-pyridinyl)methyl]-l-(l-methylethyl)-lH-indazole-4-carboxamide (110 mg, 0.24 mmol), N-[5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-2-pyridinyl]acetamide (104 mg, 0.4 mmol) in DME/water (3 mL: lmL). PdCl2(dppf)-CH2Cl2 (10.76 mg, 0.013 mmol) was added and the resulting mixture was degassed with nitrogen for 10 min. Sodium bicarbonate (66.4 mg, 0.79 mmol) was added, the vessel was sealed, and the insoluble mixture was heated in a microwave at 120 °C for 20 min. Water was added and the solids that precipitated were filtered off. DCM was added to the solids and it was purified by Si02 chromatography (eluent: gradient 100% DCM to 80:20:2
DCM/MeOH/NH4OH). Fractions were evaporated. EtOAc was added, it was sonicated and the solids that precipitated were filtered, washed with hexanes and dried to afford the title compound (91 mg, 72%) as a light beige solid. *H NMR (400 MHz, DMSO-c¾) δ ppm 1 1.54 (s, 1 H) 10.65 (s, 1 H) 8.84 (d, J=2.27 Hz, 1 H) 8.64 (t, J=4.93 Hz, 1 H) 8.39 (s, 1 H) 8.25 - 8.30 (m, 1 H) 8.15 - 8.24 (m, 2 H) 7.89 - 7.93 (m, 1 H) 5.89 (s, 1 H) 5.18 (quin, J=6.63 Hz, 1 H) 4.40 (d, J=4.80 Hz, 2 H) 2.22 (s, 3 H) 2.13 (s, 3 H) 2.12 (s, 3 H) 1.51 (s, 3 H) 1.50 (s, 3 H). MS(ES) [M+H]+ 473.1.
Example 109
AL[(4,6-Dimethyl-2-oxo-l,2-dihydro-3-pyridinyl)methyl]-l-(l-methylethyl)-6-(3-pyridinyl)-l/7- indazole-4-carboxamide
Figure imgf000132_0001
In a 25 mL sealable tube under nitrogen were combined 6-bromo-N-[(4,6-dimethyl-2-oxo- l,2-dihydro-3-pyridinyl)methyl]-l-(l-methylethyl)-lH-indazole-4-carboxamide (120 mg, 0.29 mmol) and 3-pyridinylboronic acid (53 mg, 0.43 mmol) in DME/water (3 mL: 1 mL). PdCl2(dppf)-CH2Cl2 (11.7 mg, 0.014 mmol) was added and the resulting mixture was degassed with nitrogen for 10 min. Sodium bicarbonate (72.5 mg, 0.86 mmol) was added and the insoluble mixture was heated in a microwave at 120 °C for 20 min. It was evaporated and the residue was dissolved in DCM and purified by Si02 chromatography (eluent: gradient 100% DCM to 80:20:2 DCM/MeOH/NH4OH). Fractions were evaporated. DMF was added along with some water and the solids that precipitated were filtered, washed with water and hexanes and dried to afford the title compound (74 mg, 61%) as a light grey solid. *H NMR (400 MHz, DMSO-c¾) δ ppm 1 1.54 (s, 1 H) 9.11 (d, J=2.02 Hz, 1 H) 8.66 (t, J=4.80 Hz, 1 H) 8.61 (dd, J=4.80, 1.26 Hz, 1 H) 8.42 (s, 1 H) 8.28 (dt, J=8.02, 1.93 Hz, 1 H) 8.25 (s, 1 H) 7.93 (s, 1 H) 7.54 (dd, J=7.83, 4.80 Hz, 1 H) 5.89 (s, 1 H) 5.19 (dt, J=13.14, 6.57 Hz, 1 H) 4.40 (d, J=4.80 Hz, 2 H) 2.22 (s, 3 H) 2.12 (s, 3 H) 1.52 (s, 3 H) 1.50 (s, 3 H). MS(ES) [M+H]+ 416.1.
Example 110
^-[(4,6-Dimethyl-2-oxo-l,2-dihydro-3-pyridinyl)methyl]-l-(l-methylethyl)-6-[6-(4-morpholinyl)-3- pyridinyl] - l//-indazole-4-carboxamide
Figure imgf000132_0002
In a 25 mL sealable tube under nitrogen were combined 6-bromo-N-[(4,6-dimethyl-2-oxo- l,2-dihydro-3-pyridinyl)methyl]-l-(l-methylethyl)-lH-indazole-4-carboxamide (110 mg, 0.26 mmol) and [6-(4-morpholinyl)-3-pyridinyl]boronic acid (82 mg, 0.39 mmol) in DME/water (3 mL: l mL). PdCl2(dppf)-CH2Cl2 (10.76 mg, 0.013 mmol) was added and the resulting mixture was degassed with nitrogen for 10 min. Sodium bicarbonate (66.4 mg, 0.79 mmol) was added and the insoluble mixture was heated in a microwave at 110 °C for 20 min. It was evaporated, DCM/MeOH (1 : 1) was added, it was pre-absorbed on silica gel and purified by Si02 chromatography (eluent: gradient 100% DCM to 80:20:2 DCM/MeOH/NH4OH). Fractions were evaporated. EtOH was added, it was sonicated and the solids that precipitated were filtered, washed with hexanes and dried to afford the title compound (123 mg, 89%) as a light beige solid. H NMR (400 MHz, DMSO-<i6) δ ppm 1 1.54 (s, 1 H) 8.69 (d, J=2.27 Hz, 1 H) 8.61 (t, J=4.93 Hz, 1 H) 8.36 (s, 1 H) 8.08 - 8.12 (m, 2 H) 7.85 (d, J=1.01 Hz, 1 H) 6.98 (d, ,7=8.84 Hz, 1 H) 5.89 (s, 1 H) 5.14 (quin, .7=6.57 Hz, 1 H) 4.39 (d, .7=5.05 Hz, 2 H) 3.71 - 3.75 (m, 4 H) 3.50 - 3.55 (m, 4 H) 2.22 (s, 3 H) 2.13 (s, 3 H) 1.50 (s, 3 H) 1.49 (s, 3 H). MS(ES) [M+H]+ 501.1.
Example 111
A/-[(4,6-Dimethyl-2-oxo-l,2-dihydro-3-pyridinyl)methyl]-l-(l-methylethyl)-6-[6-(methyloxy)-3- pyridinyl] - 17/-indazole-4-carboxamide
Figure imgf000133_0001
In a 25 mL sealable tube under nitrogen were combined 6-bromo-N-[(4,6-dimethyl-2-oxo- l,2-dihydro-3-pyridinyl)methyl]-l-(l-methylethyl)-lH-indazole-4-carboxamide (120 mg, 0.29 mmol) and [6-(methyloxy)-3-pyridinyl]boronic acid (66 mg, 0.43 mmol) in DME/water (3 mL: l mL). PdCl2(dppf)-CH2Cl2 (11.7 mg, 0.014 mmol) was added and the resulting mixture was degassed with nitrogen for 10 min. Sodium bicarbonate (72.5 mg, 0.86 mmol) was added and the insoluble mixture was heated in a microwave at 110 °C for 20 min. Water was added and the solids that precipitated were filtered. DCM was added and it was purified by Si02 chromatography (eluent: gradient 100% DCM to 80:20:2 DCM/MeOH NH4OH). Fractions were evaporated. EtOAc was added along with some hexanes, it was sonicated and the solids that precipitated were filtered. DMF was added along with some water and the solids that precipitated were filtered and dried to afford the title compound (57 mg, 43%) as an off-white solid. lB NMR (400 MHz, DMSO- 6) δ ppm 11.54 (s, 1 H) 8.69 (d, 7=2.27 Hz, 1 H) 8.62 (t, .7=4.80 Hz, 1 H) 8.39 (s, 1 H) 8.22 (dd, J=8.72, 2.65 Hz, 1 H) 8.15 (s, 1 H) 7.87 (s, 1 H) 6.97 (d, .7=8.59 Hz, 1 H) 5.89 (s, 1 H) 5.15 (dt, J=13.07, 6.47 Hz, 1 H) 4.39 (d, J=4.80 Hz, 2 H) 3.92 (s, 3 H) 2.22 (s, 3 H) 2.12 (s, 3 H) 1.51 (s, 3 H) 1.49 (s, 3 H). MS(ES) [M+H]+ 446.1.
Example 112
N- [(4,6-Dimethyl-2-oxo- 1 ,2-dihydro-3-pyridinyl)methyl] - 1 -( 1 -methylethyl)-6-[2- (trifluoromethyl)phenyl]-l/7-indazole-4-carboxamide
Figure imgf000134_0001
In a 25 niL sealable tube under nitrogen were combined 6-bromo-N-[(4,6-dimethyl-2-oxo- l,2-dihydro-3-pyridinyl)methyl]-l-(l-methylethyl)-lH-indazole-4-carboxamide (100 mg, 0.24 mmol) and [2-(trifluoromethyl)phenyl]boronic acid (68.3 mg, 0.36 mmol) in dioxane/water (3 mL: l mL). PdCl2(dppf)-CH2Cl2 (9.78 mg, 0.012 mmol) was added and the resulting mixture was degassed with nitrogen for 10 min. Sodium bicarbonate (60.4 mg, 0.72 mmol) was added and the insoluble mixture was heated in a microwave at 110 °C for 15 min. Water was added and the solids that precipitated were filtered off. DCM/MeOH (1 : 1) was added, it was pre-absorbed on silica gel and purified by S1O2 chromatography (eluent: gradient 100% DCM to 80:20:2 DCM/MeOH/NH4OH). Fractions were evaporated. EtOAc was added along with some hexanes, it was sonicated and the solids that precipitated were filtered and dried to afford the title compound (87 mg, 74%) as an off-white solid. ¾ NMR (400 MHz, DMSO-c¾) δ ppm 1 1.52 (s, 1 H) 8.53 (t, J=4.80 Hz, 1 H) 8.43 (s, 1 H) 7.88 (d, J=7.83 Hz, 1 H) 7.81 (s, 1 H) 7.77 (t, J=7.45 Hz, 1 H) 7.66 (t, J=7.71 Hz, 1 H) 7.53 - 7.57 (m, 2 H) 5.87 (s, 1 H) 5.05 (dt, J=13.14, 6.57 Hz, 1 H) 4.35 (d, J=4.80 Hz, 2 H) 2.19 (s, 3 H) 2.10 (s, 3 H) 1.48 (s, 3 H) 1.47 (s, 3 H). MS(ES) [M+H]+ 483.0.
Example 113
6- {4-[(Dimethylamino)sulfonyl]phenyl}-N-[(4,6-dimethyl-2-oxo-l,2-dihydro-3-pyridinyl)methyl]-l-
(l- oxamide
Figure imgf000134_0002
In a 25 mL sealable tube under nitrogen were combined 6-bromo-N-[(4,6-dimethyl-2-oxo- l,2-dihydro-3-pyridinyl)methyl]-l-(l-methylethyl)-lH-indazole-4-carboxamide (220 mg, 0.53 mmol) and {4-[(dimethylamino)sulfonyl]phenyl}boronic acid (181 mg, 0.79 mmol) in 3: 1 dioxane/water (4 mL). PdCl2(dppf)-CH2Cl2 (21.5 mg, 0.026 mmol) was added and the resulting mixture was degassed with nitrogen for 10 min. Sodium bicarbonate (133 mg, 1.58 mmol) was added and the insoluble mixture was heated in a microwave at 1 10 °C for 20 min. Water was added and the solids that precipitated were filtered. Purification by reversed-phase HPLC (CI 8, 15% to 80% CH3CN in water with 0.1% TFA, 12 minute gradient) afforded the TFA salt. Acetonitrile was evaporated off and the resulting mixture was diluted with saturated sodium bicarbonate. The solids that precipitated were filtered off. EtOAc /EtOH (5: 1) was added, along with some hexanes, and the mixture was sonicated. The solids that precipitated were filtered off and dried to afford the title compound (132 mg, 47%) as a white solid. !H NMR (400 MHz, DMSO-<i6) δ ppm 11.49 (br. s., 1 H) 8.71 (t, J=4.42 Hz, 1 H) 8.43 (s, 1 H) 8.27 (s, 1 H) 8.15 (s, 1 H) 8.13 (s, 1 H) 7.94 (d, J=1.26 Hz, 1 H) 7.88 (s, 1 H) 7.86 (s, 1 H) 5.89 (s, 1 H) 5.21 (quin, J=6.57 Hz, 1 H) 4.40 (d, J=4.80 Hz, 2 H) 2.66 (s, 6 H) 2.21 (s, 3 H) 2.12 (s, 3 H) 1.52 (s, 3 H) 1.50 (s, 3 H). MS(ES) [M+H]+ 522.
Example 114
jV-[(4,6-Dimethyl-2-oxo-l,2-dihydro-3-pyridm^^
indazole-4-carboxamide
Figure imgf000135_0001
In a 25 mL sealable tube under nitrogen were combined 6-bromo-N-[(4,6-dimethyl-2-oxo- l,2-dihydro-3-pyridinyl)methyl]-l-(l-methylethyl)-lH-indazole-4-carboxamide (120 mg, 0.29 mmol) and [(2-methylphenyl)boronic acid (58.6 mg, 0.43 mmol) in dioxane/water (3 mL: l mL).
PdCl2(dppf)-CH2Cl2 (11.74 mg, 0.014 mmol) was added and the resulting mixture was degassed with nitrogen for 10 min. Sodium bicarbonate (72.5 mg, 0.86 mmol) was added and the insoluble mixture was heated in a microwave at 100 °C for 15 min. DCM/MeOH (1 : 1) was added, it was pre-absorbed on silica gel and purified by S1O2 chromatography (eluent: gradient 100% DCM to 80:20:2
DCM/MeOH/NHziOH). Fractions were evaporated. EtOAc was added along with some hexanes, it was sonicated and the solids that precipitated were filtered and dried to afford the title compound (84 mg, 66%) as an off-white solid. Ή NMR (400 MHz, DMSO-c 6) δ ppm 1 1.51 (s, 1 H) 8.55 (t, J=4.80 Hz, 1 H) 8.41 (s, 1 H) 7.80 (s, 1 H) 7.55 (d, J=1.01 Hz, 1 H) 7.27 - 7.35 (m, 4 H) 5.87 (s, 1 H) 5.09 (quin, .7=6.57 Hz, 1 H) 4.36 (d, J=5.05 Hz, 2 H) 2.26 (s, 3 H) 2.20 (s, 3 H) 2.11 (s, 3 H) 1.49 (s, 3 H) 1.47 (s, 3 H). MS(ES) [M+H]+ 429.1. Example 115
6-Bromo-N- [(4-cyclohexyl-6-methy 1-2-oxo- 1 ,2-dihydro-3 -pyridinyl)methyl] - 1 -(1 -methylethyl)- 1 H- indazole-4-carboxamide
Figure imgf000136_0001
In a 25 mL sealable tube under nitrogen were combined 6-bromo- 1-(1 -methylethyl)- 1H- indazole-4-carboxylic acid (237 mg, 0.84 mmol) and 3-(aminomethyl)-4-cyclohexyl-6-methyl-2(lH)- pyridinone.TFA (434 mg, 1.3 mmol) in DMSO (10 mL). 1 -hydroxy-7-azabenzotriazole (194 mg, 1.42 mmol) was added and the resulting mixture was degassed with nitrogen for 10 minutes. N- methylmorpholine (0.39 mL, 3.52 mmol) and EDC (273 mg, 1.42 mmol) were added, the vessel was sealed, and the mixture was stirred at room temperature for 2 days. The mixture was poured into 10 mL of ice- water and beige solids crashed out. K2C03 (10%) was added to adjust the pH~8-9 and the mixture stirred for 30 min and let stand for another 30 min. Solids were filtered off and air-dried. DMF was added, it was heated and sonicated and some water was added. Solids that precipitated were filtered, washed with water and dried to afford the title compound (307 mg, 73%) as a light beige solid. ¾ NMR (400 MHz, DMSO-<i6) δ ppm 1 1.53 (s, 1 H) 8.63 (t, ,7=4.80 Hz, 1 H) 8.37 (s, 1 H) 8.20 (s, 1 H) 7.68 (d, J=1.26 Hz, 1 H) 6.01 (s, 1 H) 5.05 (dt, J=13.14, 6.57 Hz, 1 H) 4.43 (d, .7=4.80 Hz, 2 H) 2.84 (t, J=11.24 Hz, 1 H) 2.15 (s, 3 H) 1.70 (d, 7=13.39 Hz, 2 H) 1.60 (d, J=12.13 Hz, 3 H) 1.47 (s, 3 H) 1.45 (s, 3 H) 1.42 (br. s., 1 H) 1.17 - 1.39 (m, 4 H). MS(ES) [M+H]+ 485.2, 487.2.
Example 116
l-(l-Methylethyl)-N-[(6-methyl-2-oxo-4-propyl-l,2-dihydro-3-pyridinyl)methyl]-lH-indazole-4- carboxamide
Figure imgf000136_0002
6-Bromo- 1 -(1 -methylethyl)-N-[(6-methyl-2-oxo-4-phenyl- 1 ,2-dihydro-3-pyridinyl)methyl]- lH-indazole-4-carboxamide (70.3 mg, 0.15 mmol) in 3: 1 EtOH/THF (4 mL) was hydrogenated using Pd/C (10% wet Degussa type) and a balloon of hydrogen. The reactin mixture was stirred at room temperature overnight. It was filtered through Celite, washed with EtOAc and EtOH and the solvent was evaporated. DMF was added with some water and white solids that crashed out were filtered off, air-dried for 15 min and dried in a vacuum oven for 3 h. The residue was purified by Gilson reversed- phase HPLC (30 100 Varian Polaris CI 8, 15-80% gradient of MeCN in water with 0.1% TFA over 12 minutes). Most of the solvent was removed and saturated NaHCCh was added. The solids that crashed out were filtered off, air-dried for 15 min and dried in vacuum oven overnight to give the title compound (19 mg, 37%). ¾ NMR (400 MHz, DMSO-i6) δ ppm 11.75 (br. s., 1 H) 8.63 (br. s., 1 H) 8.34 (s, 1 H) 7.85 (d, J=8.34 Hz, 1 H) 7.39 - 7.51 (m, 7 H) 5.99 (s, 1 H) 5.03 (quin, J=6.57 Hz, 1 H) 4.22 (d, J=4.29 Hz, 2 H) 2.21 (s, 3 H) 1.49 (s, 3 H) 1.47 (s, 3 H). MS(ES) [M+H]+ 367.2.
The following compounds were prepared using the general procedures outlined for Exampli 116:
Example 117
6-Bromo- 1 -cyclopentyl-N- [(6-methyl-2-oxo-4-propyl- 1 ,2-dihydro-3 -pyridinyl)methyl] - lif-indazole-
4-carboxamide
Figure imgf000137_0001
H NMR (400 MHz, DMSO-i/6) δ ppm 1 1.54 (s, 1 H) 8.62 (t, J=4.80 Hz, 1 H) 8.36 (s, 1 H) 8.20 (s, 1 H) 7.69 (d, J=1.26 Hz, 1 H) 5.91 (s, 1 H) 5.22 (quin, J=7.01 Hz, 1 H) 4.36 (d, J=4.80 Hz, 2 H) 2.07 - 2.15 (m, 5 H) 1.93 - 2.01 (m, 2 H) 1.83 - 1.91 (m, 2 H) 1.65 - 1.73 (m, 2 H) 1.47 - 1.56 (m, 2 H) 0.89 (t, J=7.33 Hz, 3 H). MS(ES) [M+H]+ 471.2, 473.0.
Example 118
6-Bromo-AT-[(4-cyclopropyl-6-methyl-2-oxo- l,2-dihydro-3-pyridinyl)methyl]-l-(l-methylethyl)-l/i- indazole-4-carboxamide
Figure imgf000138_0001
¾ NMR (400 MHz, DMSO- 6) § ppm 1 1.48 (br. s., 1 H) 8.66 (t, .7=4.67 Hz, 1 H) 8.32 - 8.38 (m, 1 H) 8.20 (s, 1 H) 7.71 (d, .7=1.26 Hz, 1 H) 5.52 (s, 1 H) 5.06 (quin, .7=6.57 Hz, 1 H) 4.53 (d, . =4.80 Hz, 2 H) 2.06 - 2.17 (m, 4 H) 1.47 (s, 3 H) 1.45 (s, 3 H) 0.89 - 0.97 (m, 2 H) 0.68 - 0.78 (m, 2 H).
MS(ES) [M+H]+ 443.0, 445.1.
Example 1 19
l-cyclopentyl-N-[(4,6-dimethyl-2-oxo- l ,2-dihydro-3-pyridinyl)methyl]-6-[2-(4-methyl- l - piperazinyl)-4-pyridinyl]- l/f-indazole-4-carboxamide
Figure imgf000138_0002
!H NMR (400 MHz, OMSO-d6) δ ppm 1 1.54 (s, 1 H) 8.65 (t, .7=4.80 Hz, 1 H) 8.39 (s, 1 H) 8.24 (s, 1 H) 8.21 (d, .7=5.05 Hz, 1 H) 7.87 (s, 1 H) 7.21 (s, 1 H) 7.14 (d, .7=5.31 Hz, 1 H) 5.89 (s, 1 H) 5.38 (quin, ,7=7.14 Hz, 1 H) 4.39 (d, .7=5.05 Hz, 2 H) 3.56 - 3.63 (m, 4 H) 2.44 (t, .7=4.80 Hz, 4 H) 2.24 (s, 3 H) 2.22 (s, 3 H) 2.14 - 2.20 (m, 2 H) 2.12 (s, 3 H) 1.98 - 2.05 (m, 2 H) 1.91 (dd, .7=8.84, 5.56 Hz, 2 H) 1.68 - 1.75 (m, 2 H). MS(ES) [M+H]+ 540.3.
Example 120
l -Cyclopentyl-A^-[(4,6-dimethyl-2-oxo-l,2-dihydro-3-pyridinyl)methyl]- 17^-indazole-4-carboxamide
Figure imgf000138_0003
!H NMR (400 MHz, DMSO- 6) δ ppm 1 1.39 (br. s., 1 H) 8.50 (br. s., 1 H) 8.35 (s, 1 H) 7.85 (d, .7=8.59 Hz, 1 H) 7.54 (d, .7=7.07 Hz, 1 H) 7.40 (dd, J=8.46, 7.20 Hz, 1 H) 5.88 (s, 1 H) 5.21 (quin, .7=7.01 Hz, 1 H) 4.36 (d, 7=4.80 Hz, 2 H) 2.22 (s, 3 H) 2.09 - 2.16 (m, 6 H) 1.96 - 2.04 (m, 2 H) 1.84 - 1.92 (m, 2 H) 1.66 - 1.74 (m, 2 H). MS(ES) [M+H]+ 365.3.
Example 121
6-Bromo-l-(l-methylethyl)-N-[(6-methyl-2^
indazole-4-carboxamide
Figure imgf000139_0001
*H NMR (400 MHz, DMSO-i¾) δ ppm 1 1.86 (br. s., 1 H) 8.60 (t, J=4.04 Hz, 1 H) 8.34 (s, 1 H) 8.21 (s, 1 H) 7.64 (d, J=1.26 Hz, 1 H) 7.39 - 7.47 (m, 5 H) 6.00 (s, 1 H) 5.06 (dt, J=13.14, 6.57 Hz, 1 H) 4.17 (d, J=4.04 Hz, 2 H) 2.22 (s, 3 H) 1.47 (s, 3 H) 1.45 (s, 3 H). MS(ES) [M+H]+ 478.9, 480.8.
Example 122
l-Cyclopentyl-N-[(4,6-dimethyl-2-oxo-l,2-dihydro-3-pyridinyl)methyl]-6-(2-oxo-2,3-dihydro benzimidazol-5-yl)- 17i-indazole-4-carboxamide
Figure imgf000139_0002
!H NMR (400 MHz, DMSO- 6) δ ppm 1 1.53 (s, 1 H) 10.78 (s, 1 H) 10.74 (s, 1 H) 8.64 (t, J=4.80 Hz, 1 H) 8.34 (s, 1 H) 8.03 (s, 1 H) 7.79 - 7.84 (m, 1 H) 7.44 (dd, J=8.21, 1.64 Hz, 1 H) 7.37 (s, 1 H) 7.03 (d, J=8.08 Hz, 1 H) 5.89 (s, 1 H) 5.34 (quin, J=7.14 Hz, 1 H) 4.39 (d, J=4.80 Hz, 2 H) 2.21 (s, 3 H) 2.15 - 2.19 (m, 1 H) 2.13 (s, 4 H) 1.98 - 2.06 (m, 2 H) 1.85 - 1.94 (m, 2 H) 1.67 - 1.76 (m, 2 H). MS(ES) [M+H]+ 497.3. Example 123
1 -(1 -methylethyl)-N- { [6-methyl-4-( 1 -methylethyl)-2-oxo- 1 ,2-dihydro-3 -pyridinyl]methyl} -6- [2-(4- methyl- l-piperazin l)-4-pyridinyl]- lii-indazole-4-carboxamide
Figure imgf000140_0001
!H NMR (400 MHz, DMSO-tfe) δ ppm 1 1.51 (br. s., 1 H) 8.67 (t, J=4.55 Hz, 1 H) 8.39 (s, 1 H) 8.24 (s, 1 H) 8.20 (d, J=5.31 Hz, 1 H) 7.85 (d, J=1.01 Hz, 1 H) 7.21 (s, 1 H) 7.14 (dd, J=5.31 , 1.26 Hz, 1 H) 6.03 (s, 1 H) 5.21 (quin, J=6.63 Hz, 1 H) 4.48 (br. s., 1 H) 4.46 (br. s., 1 H) 3.54 - 3.64 (m, 4 H) 3.25 (dt, J=13.64, 6.82 Hz, 1 H) 2.41 - 2.46 (m, 4 H) 2.24 (s, 3 H) 2.16 (s, 3 H) 1.52 (s, 3 H) 1.50 (s, H) 1.12 (s, 3 H) 1.10 (s, 3 H). MS(ES) [M+H]+ 542.3.
Example 124
l-(l-Methylethyl)-N-[(6-methyl-2-oxo-4-propyl-l,2-dihydro-3-pyridinyl)methyl]-6-(2-oxo-2,3- dihydro- l /-benzimidazol-5- l)- l//-indazole-4-carboxamide
Figure imgf000140_0002
!H NMR (400 MHz, DMSO- 6) δ ppm 1 1.54 (s, 1 H) 10.79 (s, 1 H) 10.74 (s, 1 H) 8.65 (t, J=4.93 Hz, 1 H) 8.35 (s, 1 H) 8.02 (s, 1 H) 7.80 (s, 1 H) 7.43 (dd, J=8.08, 1.77 Hz, 1 H) 7.37 (s, 1 H) 7.03 (d, J=8.08 Hz, 1 H) 5.91 (s, 1 H) 5.16 (quin, J=6.63 Hz, 1 H) 4.42 (d, J=4.80 Hz, 2 H) 2.52 - 2.57 (m, 1 H) 2.14 (s, 3 H) 1.51 - 1.59 (m, 2 H) 1.50 (s, 3 H) 1.49 (s, 3 H) 0.89 (t, J=7.33 Hz, 3 H). MS(ES) [M+H]+ 499.3.
Example 125
l-Cyclopentyl-A¾4,6-dimethyl-2-oxo ,2-d ^
indazole-4-carboxamide
Figure imgf000141_0001
H NMR (400 MHz, DMSO-i/6) δ ppm 1 1.45 (br. s., 1 H) 8.97 (d, .7=2.27 Hz, 1 H) 8.69 (br. s., 1 H) 8.39 (s, 1 H) 8.21 (s, 1 H) 8.17 (dd, .7=8.08, 2.53 Hz, 1 H) 7.90 (s, 1 H) 7.40 (d, J=8.08 Hz, 1 H) 5.89 (s, 1 H) 5.35 (quin, J=7.14 Hz, 1 H) 4.39 (d, =4.80 Hz, 2 H) 2.54 (s, 3 H) 2.22 (s, 3 H) 2.16 (d, .7=7.58 Hz, 2 H) 2.12 (s, 3 H) 1.98 - 2.06 (m, 2 H) 1.86 - 1.94 (m, 2 H) 1.68 - 1.76 (m, 2 H). MS(ES) [M+H]+ 456.0.
Example 126
1 -(1 -methylethyl)- V- [(6-methyl-2-oxo-4-propyl- 1 ,2-dihydro-3 -pyridinyl)methyl] -6- [2-(4-methyl- 1 - piperazinyl)-4- ridinyl] - 177-indazole-4-carboxamide
Figure imgf000141_0002
!H NMR (400 MHz, DMSO-c¾) δ ppm 1 1.54 (s, 1 H) 8.65 (t, J=4.80 Hz, 1 H) 8.40 (s, 1 H) 8.24 (s, 1 H) 8.21 (d, J=5.30 Hz, 1 H) 7.86 (s, 1 H) 7.21 (s, 1 H) 7.14 (d, J=5.31 Hz, 1 H) 5.91 (s, 1 H) 5.21 (dt, 7=13.14, 6.57 Hz, 1 H) 4.42 (d, J=4.80 Hz, 2 H) 3.56 - 3.63 (m, 4 H) 2.53 - 2.56 (m, 2 H) 2.44 (t, J=4.93 Hz, 4 H) 2.24 (s, 3 H) 2.13 (s, 3 H) 1.52 - 1.61 (m, 2 H) 1.52 (s, 3 H) 1.50 (s, 3 H) 0.89 (t, J=7.33 Hz, 3 H). MS(ES) [M+H]+ 542.2.
Example 127
l-Cyclopentyl-N-[(6-methyl-2-oxo-4-propyl-l,2-dihydro-3-pyridinyl)methyl]-6-(6-methyl-3- pyridinyl -177-indazole-4-carboxamide
Figure imgf000141_0003
!H NMR (400 MHz, DMSO- 6) δ ppm 1 1.46 (br. s., 1 H) 8.96 (d, .7=2.02 Hz, 1 H) 8.70 (br. s., 1 H) 8.40 (s, 1 H) 8.21 (s, 1 H) 8.17 (dd, .7=8.08, 2.53 Hz, 1 H) 7.89 (s, 1 H) 7.40 (d, J=8.08 Hz, 1 H) 5.91 (s, 1 H) 5.31 - 5.39 (m, 1 H) 4.42 (d, 7=4.55 Hz, 2 H) 2.54 (s, 5 H) 2.12 - 2.20 (m, 5 H) 2.05 (br. s., 2 H) 1.90 (dd, J=8.97, 5.68 Hz, 2 H) 1.67 - 1.76 (m, 2 H) 1.48 - 1.57 (m, 2 H) 0.88 (t, J=7.33 Hz, 3 H). MS(ES) [M+H]+ 484.2.
Example 128
l-Cyclopentyl- -[(6-methyl-2-oxo-4-propyl^^
indazole-4-carboxamide
Figure imgf000142_0001
H NMR (400 MHz, DMSO- 6) δ ppm 1 1.55 (br. s., 1 H) 9.14 (s, 1 H) 8.60 - 8.70 (m, 2 H) 8.42 (s, 1 H) 8.35 (d, J=8.08 Hz, 1 H) 8.28 (s, 1 H) 7.93 (d, J=1.01 Hz, 1 H) 7.61 (dd, .7=7.83, 4.80 Hz, 1 H) 5.92 (s, 1 H) 5.36 (quin, J=7.14 Hz, 1 H) 4.43 (d, J=4.80 Hz, 2 H) 2.54 (d, .7=6.82 Hz, 2 H) 2.12 - 2.21 (m, 5 H) 1.99 - 2.07 (m, 2 H) 1.86 - 1.95 (m, 2 H) 1.67 - 1.77 (m, 2 H) 1.48 - 1.57 (m, 2 H) 0.88 (t, .7=7.33 Hz, 3 H). MS(ES) [M+H]+ 470.2.
Example 129
l-(l-Methylethyl)-JV-[(6-methyl-2-oxo-4-propyU
indazole-4-carboxamide
Figure imgf000142_0002
!H NMR (400 MHz, DMSO-c¾) δ ppm 1 1.54 (s, 1 H) 9.1 1 (br. s., 1 H) 8.66 (t, J=4.80 Hz, 1 H) 8.62 (d, J=4.04 Hz, 1 H) 8.42 (s, 1 H) 8.28 (dt, J=8.21, 1.83 Hz, 1 H) 8.25 (s, 1 H) 7.92 (d, J=1.01 Hz, 1 H) 7.55 (dd, J=7.96, 4.67 Hz, 1 H) 5.92 (s, 1 H) 5.19 (quin, J=6.63 Hz, 1 H) 4.43 (d, J=4.80 Hz, 2 H) 2.54 (d, J=7.07 Hz, 2 H) 2.13 (s, 3 H) 1.52 - 1.59 (m, 2 H) 1.52 (s, 3 H) 1.50 (s, 3 H) 0.88 (t, J=7.33 Hz, 3 H). MS(ES) [M+H]+ 444.3. Example 130
l-Cyclopentyl-A¾4,6-dimethyl-2-oxo-l,2-dihy^
pyridinyl] - li/-indazole-4-carboxamide
Figure imgf000143_0001
H NMR (400 MHz, DMSO-4) δ ppm 1 1.54 (s, 1H), 8.69 (d, J = 2.27 Hz, 1H), 8.60 (t, J = 5.05 Hz, 1H), 8.35 (s, 1H), 8.10 (ddd, J = 2.78, 2.91, 5.94 Hz, 2H), 7.85 (d, J = 1.01 Hz, 1H), 6.98 (d, J = 8.84 Hz, 1H), 5.89 (s, 1H), 5.31 (t, J = 7.07 Hz, 1H), 4.39 (d, J = 4.80 Hz, 2H), 3.70 - 3.76 (m, 4H), 3.50 - 3.55 (m, 4H), 2.22 (s, 3H), 2.09 - 2.18 (m, 5H), 1.96 - 2.07 (m, 2H), 1.89 (dd, J = 5.68, 9.22 Hz, 2H), 1.66 - 1.77 (m, 2H). MS(ES) [M+H]+ 527.1.
Example 131
1 -Cyclopentyl-N- [(4,6-dimethyl-2-oxo- 1 ,2-dihydro-^
indazole-4-carboxamide
Figure imgf000143_0002
H NMR (400 MHz, DMSO- d6) δ ppm 1 1.54 (s, 1H), 9.35 (s, 2H), 9.23 (s, 1H), 8.63 (t, J = 4.93 Hz, 1H), 8.44 (s, 1H), 8.39 (s, 1H), 8.01 (d, J = 1.26 Hz, 1H), 5.90 (s, 1H), 5.35 (dq, J = 7.07, 7.33 Hz, 1H), 4.40 (d, J = 4.80 Hz, 2H), 2.22 (s, 3H), 2.13 (s, 5H), 1.98 - 2.08 (m, 2H), 1.85 - 1.96 (m, 2H), 1.67 - 1.77 (m, 2H). MS(ES) [M+H]+ 443.0. Intermediates
Intermediate 1
3-(Aminomethyl)-4,6-dimethyl-2(l//)-pyridinone hydrochloride
Figure imgf000144_0001
Palladium on carbon (10%) (3.24 g) was charged into a 2L dry Parr bottle and a small amount of acetic acid was added. Next added 4,6-dimethyl-2-oxo- 1 ,2-dihydro- pyridine-3-carbonitrile (30 g , 202.7 mmol), sodium acetate (30.75 g, 375.0 mmol), platinum oxide (0.218 g), and acetic acid (1 L).. The bottle was capped, placed on Parr apparatus, and shaken under an atmosphere of H2 (100 psi) for 2 days. The reaction mixture was filtered. The solvent was removed to give a residue, which was treated with 150 mL of cone. HC1, and the formed solids were filtered. The yellow filtrate was concentrated . To the crude compound was added 30 mL of cone. HC1 and 150 mL EtOH, the contents cooled to 0 °C, and stirred at 0 °C for 2h. The formed solids were filtered, washed with cold EtOH, ether, and dried. The product was collected as 36 g. This batch was combined with other batches prepared on smaller scales and triturated with ether to give 51 g of pure compound.. lH NM (400 MHz, DMSO-<i6) δ ppm 11.85 (br s,l H) 8.13 (br s, 3 H) 5.93 - 6.01 (m, 1 H) 3.72 - 3.80 (m, 2 H) 2.22 (s, 3 H) 2.16 (s, 3 H). Intermediate 2
3-(aminomethyl)-6-methyl-4-propyl-2(l/ )-pyridinone
Figure imgf000144_0002
Step 1
To a stirred solution of t-BuOK (20 g,178.5 mmol) and cyanoacetamide (16.5 g, 196 mmol) in DMSO (300 mL) was added (3E)-3-hepten-2-one (20 g, 178.3 mmol) under argon atmosphere at room temperature. The reaction mixture was stirred at room temperature for 30 min and then additional t- BuOK (60 g,535.7 mmol) was added to the reaction mixture. The argon was then displaced by oxygen gas and stirred for 48 hrs at room temperature under oxygen. Reaction mixture was cooled to 0 °C and diluted with water (80 mL) followed by 4N HC1 (120 mL). The mixture was stirred for 15 min and the solid was filtered. The solid was washed with water (1 L) and dried to afford l,2-dihydro-6- methyl-2-oxo-4-propylpyridine-3-carbonitrile as an off white solid (12 g, 38 %). ¾ NMR (CDC13 400 MHz) : δ 1.030 (t, 3H, J = 7.2 Hz), 1.728 (m, 2H),2.429 ( s, 3H), 2.701 (t, 2H, J=7.6 Hz), 6.083 (s, lH). LCMS E-S (M-H) = 175.1 1. Step 2
To a suspension of Raney Ni (2 g) in methanol (250 mL) was added I ,2-dihydro-6-methyl-2-oxo-4- propyipyridine-3-carbonitrile (5 g, 28.4 mmol) and methanolic ammonia (750 mL). The reaction mixture was stirred at room temperature under hydrogen pressure (60 psi) for 48 hrs. The reaction mixture was filtered through celite pad and washed with methanol (250 mL). The filtrate was concentrated under reduced pressure to afford the crude product (5.1 g). The crude product was purified by column chromatography by using a short column of silica gel (60-120 mesh) treated with methanolic NH3, (eluant: 0-25% methanol in DCM) and isolated to afford the desired product 3- (amino methyl)-6-methyl-4-propylpyridin-2(lH)-one as an off white solid (2 g , 39 % yield). lB NMR (DMSO, 400 MHz) : δ 0.921(t, 3H, J = 7.2 Hz), 1.518 (m, 2H),2.09 ( s, 3H), 2.418 (t, 2H,J=7.6 Hz), 3.451 (s,2H), 4-5.6 (br s, 2H, exchangeable), 5.822(s, lH). LCMS E-S (M+H) = 181.22.
Intermediate 3
3-(Aminomethyl)-6-methyl-4-(l-methylethyl)-2(l//)-pyridinone
Figure imgf000145_0001
The title compound was prepared in the same manner as described for 3-(aminomethyl)-6-methyl-4- propyl-2(lH)-pyridinone (Intermediate 2, steps 1 and 2) from (3£)-5-methyl-3-hexen-2-one (20 g, 137 mmol). LCMS E-S (M+H) = 181.1. :H NMR (400 MHz, DMSO-iie) δ ppm 11.8 - 11.9 (br s, 1H), 7.86 - 7.96 (br s, 3H), 6..10 (s, 1H), 3.82 - 3.86 (m, 2H), 3.02 - 3.09 (m, 1H), 2.17 (s, 3H), 1.08
(d, 6H).
Intermediate 4
3-(Aminomethyl)-4-cyclohexyl-6-methyl-2(l//)-pyridinone
Figure imgf000145_0002
Step 1 To a stirred suspension of CrCl2 (58 g, 472.8 mmol in THF (1500 niL) was added a THF solution (500 mL) of l, l-dichloro-2-propanone ( 10 g, 78.8 mmol) and cyclohexanecarbaldehyde (8.84 g, 78.8 mmol). The reaction mixture was heated at reflux for 2 h, and then quenched by the addition of 1.0 M HCl. The reaction mixture was filtered through a pad of Celite and concentrated in vacuo. The crude residue ( 10 g) was added to a solution of DMSO (150 mL) containing t-BuO (7.5 g, 65.7 mmol) and cyanoacetamide (6.1 g, 72.3 mmol) and stirred at room temperature for 30 min. Additional t- BuOK (22.5 g, 197.1 mmol) was added and the reaction mixture was stirred under an atmosphere of oxygen for an additional 1 h. The contents were purged with argon, diluted with 4 volumes of H20, and then 5 volumes of 4 N HCl, which were added slowly. The reaction mixture was filtered, washed with water and dried to give 4-cyclohexyl-6-methyl-2-oxo-l,2-dihydro-3-pyridinecarbonitrile (4.5 g, 32%). !H NMR (400 MHz, DMSO-c 6) δ ppm 6.25 (s, 1H), 2.61-2.65 (m, 1H), 2.22 (s, 3H), 1.66-1.79 (m, 4H), 1.24-1.46 (m, 6H).
Step 2
To an ice-bath cooled THF (100 mL) solution of the product from step 1 (2 g, 9.26 mmol) were added NaBH4 (0.81 g, 21.3 mmol), and ¾ (2.3 g, 9.26 mmol), and the mixture stirred for 30 min. The reaction mixture was then heated at reflux for 3h, and then allowed to cool to room temperature. After cooling to 0 °C, the reaction mixture was acidified by slow addition of 3 N HCl (1 mL). The reaction mixture was concentrated in vacuo and the crude product purified by reverse phase HPLC to give the title compound as a solid (0.5 g, 25%). LCMS E-S (M+H) = 221.1. ιΉ NMR (400 MHz, DMSO-<f6) δ ppm 11.8 - 1 1.9 (br s, 1H), 7.80-7.93 (br s, 3H), 6.07 (s, 1H), 3.69 (s, 2H), 2.67-2.75 (m, 1H), 2.17 (s, 3H), 1.58-1.72 (m, 5H), 1.19 - 1.41 (m, 5H).
Intermediate 5
3-(Aminomethy])-4-ethyl-6-methyl-2(l//)-pyridinone hydrochloride
Figure imgf000146_0001
Stepl
Hex-3-en-2-one
Figure imgf000147_0001
To a stirred solution of l-(triphenylphosphoranylidene)-2-propanone (100 g, 307 mmol) in DCM (500 mL) was added propionaldehyde (140 mL, 1929 mmol) at room temperature. The reaction mixture was then stirred at room temperature for 18 hours. The reaction was monitored by TLC. The solvent (DCM) was distilled off using ordinary distillation. The residue was then distilled using fractional distillation under vacuum (-450 mbar) and the desired product was isolated. The title compound, hex-
3-en-2-one (20 g, 66%), was collected at 110 °C. *H NMR (CDC13 400 MHz) δ ppm 1.071-1.121 (t, 3H, J = 7.4 Hz), 2.250-2.299 (m, 5H), 6.054-6.094 (d, 1H, J =16Hz), 6.823-6.895 (m, 1H).
Step2
4~Eth l-l,2-dihydro-6-methyl-2-oxopyridiiie-3-carbonitrile
Figure imgf000147_0002
To a stirred solution of t-BuOK (22.85 g, 204.08 mmol) and cyanoacetamide (18.8 g, 224.1 mmol) in DMSO (300 mL) was added hex-3-en-2-one (20 g, 204.08 mmol) under argon atmosphere at room temperature. The reaction mixture was then stirred at room temperature for 30 min and then added additional t-BuOK (68.5 g, 612.05 mmol) was added. Argon gas was displaced by oxygen gas and the mixture stirred for 48 hrs at room temperature in presence of oxygen. Reaction was monitored by TLC. The reaction mixture was cooled to 0 °C and diluted with water (100 mL) followed by 4 N HC1 (120 mL). The mixture was stirred for 15 min and the resulting solid was filtered. The solid was washed with water (1 L) and dried to afford the title compound, 4-ethyl-l,2-dihydro-6-methyl-2- oxopyridine-3-carbonitrile (10.5 g, 31 %), as an off white solid. !H NMR (CDC13 400 MHz) : δ ppm
1.148-1.185 (t, 3H, J = 7.4 Hz), 2.237 ( s, 3H), 2.557-2.614(m, 2H), 6.211 (s, 1H), 12.330 (broad s, 1H). MS(ES) [M+H]+ 161.06.
Step 3
3-(Amino methyl)-4-ethyl-6-methylpyridin-2(lH)-one
Figure imgf000147_0003
To a suspension of Raney Nickel (6 g) in methanol (200 mL) was added 4-ethyl-l,2-dihydro-6- methyl-2-oxopyridine-3-carbonitrile (10 g, 61.7 mmol) and methanolic ammonia (750 mL). The reaction mixture was stirred at room temperature under hydrogen pressure (80 psi) for 48 hrs. The reaction mixture was filtered through Celite and washed with methanol (250 mL). The filtrate was concentrated under reduced pressure and the residue purified by filter column using silica gel (60-120 mesh), eluted with 10% MeOH in CHC13, to afford 3-(amino methyl)-4-ethyl-6-methylpyridin-2(lH)- one (5.6 g, 54 %) as an off white solid. :H NMR (DMSO-D6; 400 MHz) (free amine): δ ppm 1.063- 1.101 (t, 3H, J = 7.6 Hz), 2.101 (s, 3H), 2.412-2.449 ( m, 2H), 3.448 (s, 2H), 5.835 (s,lH). MS(ES) [M+H]+ 167.06.
Step 4
3-(Aminomethy])-4-ethyl-6-methylpyridin-2(lH)-one hydrochloride
Figure imgf000148_0001
3-(Amino methyl)-4-ethyl-6-methylpyridin-2(lH)-one, (5.6 g, 33 mmol) was suspended in DCM (560 mL) and the insoluble contents/particles were filtered. The filtrate was concentrated and dried. The residue was dissolved in DCM (10 mL) and 4 M HCI inl,4-dioxane (16 mL, 66 mmol) was added at 0 °C and stirred for 10 min, at which time the reaction mixture was concentrated under high- vacuum and dried. The resulting crude solid was triturated with hexane (150 mL) and filtered. The solid was dried under vacuum. Collected 3-(amino methyl)-4-ethyl-6-methylpyridin-2(lH)-one hydrochloride (5.9 g, 86%). ¾ NMR (400 MHz, DMSO-<i6) δ ppm 1.082-1.120 (t, 3H, J = 7.6 Hz), 2.179 (s, 3H), 2.503-2.544 (m, 2H), 3.785-3.798 (d, 2H, J = 5.2 Hz), 6.024 (s,lH),7.985 (broad s,2H), 11.858 (broad s,lH). MS(ES) [M+H]+ 167.2.
Intermediate 6
3-(aniinoniethyl)-6-methyl-4,4'-bipyridin-2(l/7)-one
Figure imgf000148_0002
Step 1
(Z)-3-hydroxy-l-(pyridin-4-y])but-2-en-l-one
Figure imgf000149_0001
To a solution of ethyl 4-pyridinecarboxylate (30 g, 198 mmol) and acetone (34.58 g, 595 mmol) in THF (150 n L) was slowly added NaOMe (12.87 g, 238 mmol) at 35-40 °C. The mixture was stirred at room temperature for 0.5 h, and then heated at reflux for 3 h. The mixture was cooled to room temperature and filtered to give a solid, which was washed with M3uOMe, and dissolved in H20. The solution was acidified with acetic acid and the resulting oily product was extracted with CHC13. The solvent was removed in vacuo, and the crude product was obtained (12 g, 37%) and used without further purification. ¾ NMR (400 MHz, DMSO-d6) δ 8.73 (d, 2H), 7.76 (d, 2H), 6.63 (s, 1H), 2.21 (s, 3H); note: enolic OH does not appear. Step 2
6-methyl-2-oxo-1 ,2-dihydro-[4,4'-bi ridine]-3-carbonitrile
Figure imgf000149_0002
To a solution of (2Z)-3 -hydroxy- l-(4-pyridinyl)-2-buten-l -one (8 g, crude, 49 mmol) and cyanoacetamide (4.12 g, 49 mmol) in anhydrous EtOH (100 mL) was added piperidine (4.17 g, 49 mmol) under N2 at 75 °C. The mixture was heated at reflux for 1 h, and then cooled to room temperature. After filtration, the solid was collected and washed with H20 to give the crude product (4 g) as two isomers. After separation by HPLC, 1.8 g of 6-methyl-2-oxo-l,2-dihydro-4,4'-bipyridine-3- carbonitrile and 1.2 g of 4-methyl-6-oxo-l,6-dihydro-2,4'-bipyridine-5-carbonitrile were obtained. The identity of 6-methyl-2-oxo-l,2-dihydro-4,4'-bipyridine-3-carbonitrile was established by nOE analysis. !H NMR (400 MHz, DMSO-d6) δ 12.79 (br. s., 1H), 8.75 (d, 2H), 7.58 (d, 2 H), 6.37 (s 1H), 2.31 (s, 3H).
Step 3
3-(aminomethyl)-6-methyl-4,4'-bipyridin-2(l//)-one
Figure imgf000150_0001
To an ice bath cooled THF (100 mL) solution of 6-methyl-2-oxo-l,2-dihydro-4,4'-bipyridine-3- carbonitrile (4 g, 18.9 mmol) was added NaBH4 (1.43 g, 37.9 mmol), and I2 (4.81 g, 18.9 mmol), and the mixture was stirred for 0.5 h. The reaction mixture was then heated at reflux for 4 h. After cooling to 0 °C, the reaction mixture was adjusted to pH 5 with 4 N HC1. The mixture was concentrated in vacuo to give the crude compound, which was purified by HPLC to give 3-(aminomethyl)-6-methyl- 4,4'-bipyridin-2(lH)-one (1.9 g, 31 %) as a TFA salt. LCMS MH+ = 216.0 lB NMR (400 MHz, DMSO-d6 in D20) δ 8.87 (d, 2H), 7.87 (d, 2H), 6.13 (s, 1H), 3.65 (br s, 2H), 2.17 (s, 3H).
Intermediate 7
3-(aminomethyl)-6-methyl-4-(phenylmethyl)-2(l /)-pyridinone and 3-(aminomethyl)-4-methyl- 6-(phenylmethyl)-2(l//)-pyridinone
Figure imgf000150_0002
Step 1
1 -phenylpentane-2,4-dion
Figure imgf000150_0003
To a solution of NaNH2 (19.02 g, 480 mmol) in anhydrous ether (400 mL) under N2 at - 5 °C was added dropwise ethyl phenylacetate (19.2 g , 150 mmol) and then acetone (21.23 g, 370 mmol) with vigorous stirring. After addition, the reaction mixture was stirred at room temperature overnight. The mixture was then acidified to pH 4.0 - 5.0 with IN HC1. The organic layer was separated and concentrated in vacuo. The crude product was purified by silica gel chromatography to give 1- phenyl-2,4-pentanedione (18.32 g, 44 %). ¾ NMR (400 MHz, CDC13-d3) δ 15.49 (br s, 1H), 7.33- 7.45 (m, 5H), 5.53 (s, 1H), 3.66 (s, 2H), 2.10 (s, 3H).
Step 2
4-benz l-6-methyl-2-oxo-l,2-dihydropyridine-3-carbonitrile and 6-benzyl-4-methyl-2-oxo-l,2- dihydropyridine-3-carbo
Figure imgf000151_0001
l-phenyl-2,4-pentanedione (18.32 g, 104 mmol) and cyanoacetamide (8.74 g, 104 mmol) were dissolved in EtOH (104 mL) and heated until homogenous (ca. 75 °C). Piperidine (8.86 g, 104 mmol) was added and the reaction mixture heated at reflux for 15 -30 min. followed by cooling to room temperature, during which time precipitation occurred. The heterogenous contents were filtered to give a solid which was suspended in 200 mL water and stirred vigorously for 20 min. The heterogenous mixture was filtered to afford 6-methyl-2-oxo-4-(phenylmethyl)-l,2-dihydro-3- pyridinecarbonitrile and 4-methyl-2-oxo-6-(phenylmethyl)-l,2-dihydro-3-pyridinecarbonitrile (12.06 g, 52%). LCMS MH+ = 225.1 ¾ NMR (400 MHz, DMSO-d6) δ 7.21-7.31 (m, 10H), 6.06 (s, 2H), 3.89 (s, 2H), 3.79 (s, 2H), 2.24 (s, 3H), 2.15 (s, 3H).
Step 3
tert-butyl ((4-benzyl-6-methyl-2-oxo-l,2-dihydropyridin-3-yl)methyl)carbamate compound and tert-butyl ((6-benzyl-4-niethyl-2-oxo-l,2-dihydropyridin-3-yl)methyl)carbamate
Figure imgf000151_0002
Sodium acetate (6.14 g, 74.8 mmol), Pd C (0.65 g, 1 mmol), and platinum (II) oxide (45 mg, 1 mmol) were placed in a dried Parr bottle equipped with nitrogen inlet. A small amount of acetic acid was added to wet the catalysts. A solution of 6-methyl-2-oxo-4-(phenylmethyl)-l,2-dihydro-3- pyridinecarbonitrile and 4-methyl-2-oxo-6-(phenylmethyl)-l,2-dihydro-3-p}Tidinecarbonitrile (6 g, 26.7 mmol) in acetic acid (300 mL) was added to the vessel. The contents were sealed and hydrogenated on Parr shaker at 45 psi for 12 h. The reaction mixture was filtered and washed with acetic acid. The filtrate was removed under reduced pressure. The residue was washed with methanol and filtered to afford a crude mixture of 3-(aminomethyl)-6-methyl-4-(phenylmethyl)-2(lH)- pyridinone and 3-(aminomethyl)-4-methyl-6-(phenylmethyl)-2(lH)-pyridinone. The reaction was run in duplicate to afford a total crude recovery of 14.5 g. To a solution of the above crude product mixture (4.0 g, 17.5 mmol) in THF (10 mL) and DMF (10 mL) was added di-tert-butoxycarbonyl anhydride (5.0 g, 23.4 mmoL) and triethylamine (5.2 g, 52.5 mmol) at 0 °C. The reaction mixture was stirred with warming to room temperature and then stirred for an additional 4 h. The contents were diluted with ice water and then filtered. The collected solid was dried and the products separated by HPLC to furnish 1.2 g of 1, 1 -dimethylethyl {[4-methyl-2-oxo-6-(phenylmethyl)- l ,2-dihydro-3- pyridinyljmethyl} carbamate (¾ NMR (400 MHz, DMSO-d6) δ 1 1.55- 1.60 (br s, 1H), 7.20-7.29 (m, 5H), 5.85 (s, 1H), 3.92 (s, 2H), 3.90 (s, 2H), 2.10 (s, 3H), 1.32 (s, 9H) and 1.0 g of 1, 1 -dimethylethyl {[6-methyl-2-oxo-4-(phenylmethyl)-l,2-dihydro-3-pyridinyl]methyl}carbamate ( H NMR (400 MHz, DMSO-d6) δ 1 1.50- 1 1.55 (br s, 1H), 7.18-7.25 (m, 5H), 5.75 (s, 1H), 4.02 (s, 2H), 3.85 (s, 2H), 2.05 (s, 3H), 1.32 (s, 9H).
Step 4
3-(aminomethyl)-6-benzyl-4-methylpyridin-2(lH)-
Figure imgf000152_0001
A solution of 1 , 1 -dimethylethyl {[4-methyl-2-oxo-6-(phenylmethyl)- l ,2-dihydro-3- pyridinyljmethyl} carbamate (1.2 g, 3.66 mmol) in 4N HC1 (in 15 mL 1 ,4 dioxane) was heated to 60 °C for 1 h. The mixture was cooled to room temperature. The mixture was filtered and dried to give 3- (aminomethyl)-4-methyl-6-(phenylmethyl)-2(lH)-pyridinone as an HC1 salt (0.725 g, 87%). LCMS MH+ = 229.1 H NMR (400 MHz, DMSO-d6) δ 1 1.9- 12.0 (br s, 1H), 7.99 (br s, 3H), 7.20 (s, 5H), 5.97 (s, 1H), 3.72-3.75 (m, 4H), 2.17 (s, 3H).
3-(aminomethyl)-4-benzyl-4-methylpyridin-2(lH)-one
Figure imgf000153_0001
A solution of 1 , 1 -dimethylethyl {[6-methyl-2-oxo-4-(phenylmethyl)- l ,2-dihydro-3- pyridinyljmethyl} carbamate (1.0 g, 3.0 mmol) in 4N HC1 (in 15 mL 1,4 dioxane) was heated to 60 °C for 1 h. The mixture was cooled to room temperature. The mixture was filtered and dried to give 3- (aminomethyl)-6-methyl-4-(phenylmethyl)-2(lH)-pyridinone as an HC1 salt (0.600 g, 86%). LCMS MH+ = 229.1 ¾ NMR (400 MHz, DMSO-d6) δ 1 1.9- 12.0 (br s, 1H), 8.03 (br s, 3H), 7.16-7.30 (m, 5H), 5.84 (s, 1H), 3.91 (s, 2H), 3.81 (s, 2H), 2.10 (s, 3H). Intermediate 8
3-(aminomethyl)-4-(sec-butyl)-6-methylpyridin-2(lH)-one
Figure imgf000153_0002
a) (E)-5-methylhept-3-en-2-one
Figure imgf000153_0003
To a stirred solution of l -(triphenylphosphoranylidene)-2-propanone (73.9 g, 232.55 mmol) in DCM (150 mL) was added 2-methyl butanal (20 g, 232.55 mmol) and the resulting reaction mixture was stirred at room temperature for 16 h. DCM was distilled off from the reaction mixture using short path condenser and after that, the desired product (E)-5-methylhept-3-en-2-one, 2 (12 g, 41%) was collected as colorless liquid by fractional distillation. !H NMR (CDC13 400 MHz): δ 0.909-0.891 (t, 3H, J = 7.2 Hz), 1.085-0.976 (m, 3H), 1.46- 1.103 (m, 2H), 1.632-1.606 (d, 1H), 2.252-2.205 (m, 3H), 6.061-6.018 (dd, 1H), 6.716-6.657 (m, lH). GCMS = 126.2 (M)-Purity 87%. b) 4-sec-buty]-l,2-dihydro-6-methyl-2-oxopyridine-3-carbonitrile
Figure imgf000154_0001
To a stirred solution of t-BuOK (8 g, 71.42 mmol) and cyanoacetamide (6.6 g, 78.57 mmol) in DMSO (80 mL) was added (E)-5-methylhept-3-en-2-one, 2 (9 g, 71.42 mmol) under argon atmosphere at room temperature. The reaction mixture was stirred at room temperature for 1 h and then added additional t-BuOK (24 g, 214.28 mmol) was added. Then argon was displaced by oxygen and stirred at RT for 48 h. The reaction mixture was cooled to 0 °C, diluted with water (36 mL) followed by 4N HC1 (up to pH~4) and the resulting reaction mixture was stirred for 15 min. The separated solid was collected by filtration, washed with water (10 mL), pet ether (100 mL) and dried to afford the title compound as yellow solid (7 g, 53 %). ¾ NMR (DMSO-d6 400 MHz): δ 0.816-0.779 (t, 3H, J = 7.6 Hz), 1.178-1.161 (d, 3H, J=6.8Hz), 1637-1.505 (m, 2H), 2.24 (s, 3H), 2.814-2.760 (m, 1H), 6.239 (s, lH), 12.324 (bs, lH); LCMS (ES-) m/z = 189.20 (M-H) c) 4-sec-buty]-3-(aminomethyl)-6-methylpyridin-2(lH)-one
To a suspension of Raney Ni (10 g) in methanol (50 mL) was added 4-sec -butyl- l,2-dihydro-6- methyl-2-oxopyridine-3-carbonitrile (7 g, 36.8 mmol) followed by methanolic ammonia (200 mL) and the resulting reaction mixture was stirred at room temperature under hydrogen pressure (80 psi) for 18 h. The reaction mixture was filtered through Celite pad and washed with methanol (250 mL). The filtrate was concentrated under reduced pressure to afford the crude product (7 g). The reaction was repeated again under same condition. The crude products were combined and purified by silica gel chromatography (eluent: 8% MeOH in CHC13 , spiked with NH3) and the obtained solid was triturated with diethyl ether (50 mL) and dried under vacuum to afford the title compound as yellow solid (3.5 g, 28%). ¾ NMR (DMSO-d6,400 MHz): δ 0.809-0.774 (t, 3H, J = 6.8 Hz), 1.113-1.097 (d, 3H, J = 6.4 Hz), 1.504-1.468 (t, 2H, J =7.2 Hz), 2.184 (s, 3H), 2.839-2.822 (d, 1H, J = 6.8 Hz), 3.822 (s, 2H), 6.059 (s, 1H), 8.315 (bs, 2H); LCMS (ES+) m/z = 195.22 (M+H)
Assay Protocol
Compounds contained herein were evaluated for their ability to inhibit the methyltransferase activity of EZH2 within the PRC2 complex. Human PRC2 complex was prepared by co-expressing each of the 5 member proteins (FLAG-EZH2, EED, SUZ12, RbAp48, AEBP2) in Sf9 cells followed by co-purification. Enzyme activity was measured in a scintillation proximity assay (SPA) where a tritiated methyl group is transferred from 3H-SAM to a lysine residue on Histone H3 of a mononucleosome, purified from HeLa cells. Mononucleosomes were captured on SPA beads and the resulting signal is read on a ViewLux plate reader.
Part A. Compound Preparation
1. Prepare 10 mM stock of compounds from solid in 100% DMSO.
2. Set up an 11 -point serial dilution (1 :3 dilution, top concentration 10 mM) in 100% DMSO for each test compound in a 384 well plate leaving columns 6 and 18 for DMSO controls.
3. Dispense 100 nL of compound from the dilution plate into reaction plates (Grenier Bio-One, 384-well, Cat# 784075).
Part B. Reagent Preparation
Prepare the following solutions:
1. 50 mM Tris-HCl, pH 8: Per 1 L of base buffer, combine 1 M Tris-HCl, pH 8 (50 mL) and distilled water (950 mL).
2. lx Assay Buffer: Per 10 mL of lx Assay Buffer, combine 50 mM Tris-HCl, pH 8 (9958 uL), 1 M MgCl2 (20 uL), 2 M DTT (20 uL), and 10% Tween-20 (2 uL) to provide a final concentration of 50 mM Tris-HCl, pH 8, 2 mM MgCl2, 4 mM DTT, 0.002% Tween-20.
3. 2x Enzyme Solution: Per 10 mL of 2x Enzyme Solution, combine lx Assay Buffer and PRC2 complex to provide a final enzyme concentration of 10 nM.
4. SPA Bead Suspension: Per 1 mL of SPA Bead Suspension, combine PS-PEI coated
LEAD Seeker beads (40 mg) and ddH20 (1 mL) to provide a final concentration of 40 mg/mL.
5. 2x Substrate Solution: Per 10 mL of 2x Substrate Solution, combine lx Assay Buffer
(9728.55 uL), 800 ug/mL mononucleosomes (125 uL), 1 mM cold SAM (4 uL), and 7.02 uM 3H-SAM (142.45 uL; 0.55 mCi/mL) to provide a final concentration of 5 ug/mL
nucleosomes, 0.2 uM cold SAM, and 0.05 uM 3H-SAM.
6. 2.67x Quench/Bead Mixture: Per 10 mL of 2.67x Quench/Bead Mixture, combine dd¾0 (9358 uL), 10 mM cold SAM (267 uL), 40 mg/mL Bead Suspension (375 uL) to provide a final concentration of 100 uM cold SAM and 0.5 mg/mL SPA beads.
Part C. Assay Reaction in 384-well Grenier Bio-One Plates
Compound Addition
1. Dispense 100 nL/well of lOOx Compound to test wells (as noted above).
2. Dispense 100 nL/well of 100% DMSO to columns 6 & 18 for high and low controls,
respectively. Assay
1. Dispense 5 uL/well of 1 x Assay Buffer to column 18 (low control reactions).
2. Dispense 5 uL/well of 2x Enzyme Solution to columns 1 - 17, 19-24.
3. Spin assay plates for ~1 minute at 500 rpm.
4. Stack the assay plates, covering the top plate.
5. Incubate the compound/DMSO with the enzyme for 30 minutes at room temperature.
6. Dispense 5 uL/well of 2x Substrate Solution to columns 1 -24.
7. Spin assay plates for ~1 minute at 500 rpm.
8. Stack the assay plates, covering the top plate.
9. Incubate the assay plates at room temperature for 1 hour.
Quench/Bead Addition
1. Dispense 5 uL/well of the 3x Quench/Bead Mixture to columns 1 -24.
2. Seal the top of each assay plate with adhesive TopSeal.
3. Spin assay plates for ~1 minute at 500 rpm.
4. Equilibrate the plates for > 20 min.
Read plates
1. Read the assay plates on the Viewlux Plate Reader utilizing the 613 nm emission filter with a 300 s read time.
Reagent addition can be done manually or with automated liquid handler.
*The final DMSO concentration in this assay is 1%.
*The positive control is in column 6; negative control is in column 18.
*Final starting concentration of compounds is 100 μΜ.
Part D. Data analysis
Percent inhibition was calculated relative to the DMSO control for each compound concentration and the resulting values were fit using standard IC5o fitting parameters within the ABASE data fitting software package.
Exemplified compounds of the present invention were generally tested according to the above or an analogous assay and were found to be inhibitors of EZH2. The IC5o values ranged from about 1 nM to about 10 μΜ; The IC5o values of the more active compounds range from about 1 nM to about 500 nM; The most active compounds are under 50 nM. As tested in the foregoing assay or an analogous assay, compounds of the various Examples gave the IC50 data (nM) in the paragraph below. Repeating the assay run(s) may result in a somewhat different. Ex 1, 32; Ex 3, 158; Ex 4, 25; Ex 5, 8; Ex 7, 40; Ex 8, 10; Ex 9, 16; Ex 10, 13; Ex 12, 4 ; Ex, 126; Ex 14, 5; Ex 15, 2; Ex 16, 3; Ex 17, 8; Ex 18, 10; Ex 19, 6; Ex 20, 10; Ex 21, 5; Ex 22, 20; 23, 8; Ex 24, 5; Ex 25,13; Ex 27, 32; Ex 29, 40; Ex 30, 32; Ex 31, 20; Ex 32, 25; Ex 33, 5.

Claims

claimed is:
A compound of compounds of formula (I)
Figure imgf000158_0001
wherein
X and Z are selected independently from the group consisting of hydrogen, (Ci-C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, unsubstituted or substituted (C3-C8)cycloalkyl, unsubstituted or substituted (C3-C8)cycloalkyl-(Ci-C8)alkyl or -(C2-C8)alkenyl, unsubstituted or substituted (C5- C8)cycloalkenyl, unsubstituted or substituted (C5-C8)cycioaikenyl-(Ci-C8)alkyl or -(C2-C8)alkenyl, (C6-Cio)bicycloalkyl, unsubstituted or substituted heterocycloalkyl, unsubstituted or substituted heterocycloalkyl-(Ci-C8)alkyl or -(C2-C8)alkenyl, unsubstituted or substituted aryl, unsubstituted or substituted aryl-(Ci-C8)alkyl or -(C2-C8)alkenyl, unsubstituted or substituted heteroaryl, unsubstituted or substituted heteroaryl-(Ci-C8)alkyl or -(C2-C8)alkenyl, halo, cyano, -CORa, - C02Ra, -CONRaRb, -CONRaNRaRb, -SRa, -SORa, -S02Ra, -S02NRaRb, nitro, -NRaRb, - NRaC(0)Rb, -NRaC(0)NRaRb, -NRaC(0)ORa, -NRaS02Rb, -NRaS02NRaRb, -NRaNRaRb, - NRaNRaC(0)Rb, -NRaNRaC(0)NRaRb, -NRaNRaC(0) ORa, -ORa, -OC(0)Ra, and -OC(0)NRaRb;
Y is H or halo;
R1 is (Ci-C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, unsubstituted or substituted (C3- C8)cycloalkyl, unsubstituted or substituted (C3-C8)cycloalkyl-(Ci-C8)alkyl or -(C2-C8)alkenyl, unsubstituted or substituted (C5-C8)cycloalkenyl, unsubstituted or substituted (C5-C8)cycloalkenyl- (Ci-C8)alkyl or -(C2-C8)alkenyl, unsubstituted or substituted (C6-Ci0)bicycloalkyl, unsubstituted or substituted heterocycloalkyl or -(C2-C8)alkenyl, unsubstituted or substituted heterocycloalkyl-(Ci- C8)alkyl, unsubstituted or substituted aryl, unsubstituted or substituted aryl-(Ci-C8)alkyl or - (C2-C8)alkenyl, unsubstituted or substituted heteroaryl, unsubstituted or substituted heteroaryl-(Cr C8)alkyl or -(C2-C8)alkenyl, -CORa, -C02Ra, -CONRaRb, -CONRaNRaRb;
R3 is hydrogen, (C C8)alkyl, cyano, trifluoromethyl, -NRaRb, or halo;
R6 is selected from the group consisting of hydrogen, halo, (Ci-C8)alkyl, (C2-C8)alkenyl, - B(OH)2, substituted or unsubstituted (C2-C8)alkynyl, unsubstituted or substituted (C3-C8)cycloalkyl, unsubstituted or substituted (C3-C8)cycloalkyl-(Ci-C8)alkyl, unsubstituted or substituted (C5- C8)cycloalkenyl, unsubstituted or substituted (Q-C8)cycloalkenyl-(Ci-C8)alkyl, (C6- Cio)bicycloalkyl, unsubstituted or substituted heterocycloalkyl, unsubstituted or substituted heterocycloalkyl-(Ci-C8)alkyl, unsubstituted or substituted aryl, unsubstituted or substituted aryl- (Ci-Cs)alkyl, unsubstituted or substituted heteroaryl, unsubstituted or substituted heteroaryl-(Ci- C8)alkyl, cyano, -CORa, -C02Ra, -CONRaRb, -CONRaNRaRb, -SRa, -SORa, -S02Ra, -S02NRaRb, nitro, -NRaRb, -NRaC(0)Rb, -NRaC(0)NRaRb, -NRaC(0)ORa, -NRaS02Rb, -NRaS02NRaRb, - NRaNRaRb, -NRaNRaC(0)Rb, -NRaNRaC(0)NRaRb, -NRaNRaC(0)ORa, -ORa, -OC(0)Ra, - OC(0)NRaRb;
wherein any (Ci-C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, cycloalkyl, cycloalkenyl, bicycloalkyl, heterocycloalkyl, aryl, or heteroaryl group is optionally substituted by 1 , 2 or 3 groups independently selected from the group consisting of -0(Ci-C6)alkyl(R )1_2, -S(Cr C6)alkyl(Rc)1.2, -(C1-C6)alkyl(Rc)1.2, (CrC8)alkyl-heterocycloalkyl, (C3-C8)cycloalkyl- heterocycloalkyl, halo, (Ci-C6)alkyl, (C3-C8)cycloalkyl, (C5-C8)cycloalkenyl,
(C C6)haloalkyl, cyano, -CORa, -C02Ra,-CONRaRb, -SRa, -SORa, -S02Ra, -S02NRaRb, nitro, -NRaRb, -NRaC(0)Rb, -NRaC(0)NRaRb, -NRaC(0)ORa, -NRaS02Rb, - NRaS02NRaRb, -ORa, -OC(0)Ra, -OC(0)NRaRb, heterocycloalkyl, aryl, heteroaryl, aryl(Ci-C4)alkyl, and heteroaryl(Ci-C4)alkyl;
wherein any aryl or heteroaryl moiety of said aryl, heteroaryl, aryl(Ci-C4)alkyl, or heteroaryl(Ci-C4)alkyl is optionally substituted by 1, 2 or 3 groups independently selected from the group consisting of halo, (Ci-C6)alkyl, (C3-C8)cycloalkyl, (C5-C8)cycloalkenyl, (Ci-C6)haloalkyl, cyano, -CORa, -C02Ra, -CONRaRb, -SRa, -SORa, -S02Ra, -S02NRaRb, nitro, -NRaRb, -NRaC(0)Rb, -NRaC(0)NR¾b, -NRaC(0)ORa, -NRaS02Rb, -NRaS02NRaRb, -ORa,
-OC(0)Ra, and -OC(0)NRaRb;
Ra and Rb are each independently hydrogen, (Ci-C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, (C3-C8)cycloalkyl, (C5-C8)cycloalkenyl, (C6-C10)bicycloalkyl, heterocycloalkyl, aryl, heteroaryl, wherein said (d-d)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, cycloalkyl, cycloalkenyl, bicycloalkyl, heterocycloalkyl ,aryl or heteroaryl group is optionally substituted by 1, 2 or 3 groups
independently selected from halo, hydroxyl, (d-d)alkoxy, amino, (d-d)alkylamino,
((Ci-C4)alkyl)((Ci-C4)alkyl)amino, -C02H, -C02(Ci-C4)alkyl, -CONH2,-CONH(Ci-C4)alkyl, - CON((Ci-C4)alkyl)((Ci-C4)alkyl), -S02(Ci-C4)alkyl, -S02NH2,-S02NH(Ci-C4)alkyl, or - S02N((d-d)alkyl)((d-d)alkyl);
or Ra and Rb taken together with the nitrogen to which they are attached represent a 5-8 membered saturated or unsaturated ring, optionally containing an additional heteroatom selected from oxygen, nitrogen, and sulfur, wherein said ring is optionally substituted by 1 , 2 or 3 groups independently selected from (Ci-C4)alkyl, (Ci-C4)haloalkyl, amino, (CrC4)alkylamino,
((C C4)alkyl)((Ci-C4)alkyl)amino, hydroxyl, oxo, (C C4)alkoxy, and (d-d)alkoxy(d-d)alkyl, wherein said ring is optionally fused to a (C3-Cg)cycloalkyl, heterocycloalkyl, aryl, or heteroaryl ring;
or Ra and Rb taken together with the nitrogen to which they are attached represent a 6- to 10-membered bridged bicyclic ring system optionally fused to a (C3-C8)cycloalkyl,
heterocycloalkyl, aryl, or heteroaryl ring;
each Rc is independently (CrC4)alkylamino, -NRaS02Rb, -SORa, -S02Ra, -NRaC(0)ORa, -NRaRb, or -C02Ra;
or a salt thereof.
2. A compound of formula (I) according to claim 1 wherein:
X and Z are independently selected from the group consisting of (Ci-C8)alkyl, (C3- C8)cycloalkyl, heterocycloalkyl, aryl, heteroaryl, -NRaRb, and -ORa;
Y is H or F;
R1 is selected from the group consisting of (Ci-C8)alkyl, (C3-C8)cycloalkyl,
heterocycloalkyl, aryl, and heteroaryl;
R3 is selected from the group consisting of hydrogen, (Ci-C8)alkyl, cyano, trifluoromethyl, -NRaRb, and halo;
R6 is selected from the group consisting of hydrogen, halo, cyano, trifluoromethyl, amino, (Ci-C8)alkyl, (C3-C8)cycloalkyl, aryl, heteroaryl, acylamino; (C2-C8)alkynyl, arylalkynyl, heteroarylalkynyl, -S02Ra, -S02NRaRb , and -NRaS02Rb ;
wherein any (Ci-C8)alkyl, (C3-C8)cycloalkyl, (C2-C8)alkynyl, arylalkynyl,
heteroarylalkynyl group is optionally substituted by 1 , 2 or 3 groups independently selected from
Figure imgf000160_0001
(C C8)alkyl- heterocycloalkyl, (C3-C8)cycloalkyl-heterocycloalkyl, halo, (Ci-C6)alkyl,
(C3-C8)cycloalkyl, (C5-C8)cycloalkenyl, (C C6)haloalkyl, cyano, -CORa, -C02Ra, - CONRaRb, -SRa, -SORa, -S02Ra, -S02NRaRb, nitro, -NRaRb, -NRaC(0)Rb, - NRaC(0)NRaRb, -NRaC(0)ORa, -NRaS02Rb, -NRaS02NRaRb, -ORa, -OC(0)Ra, - OC(0)NRaRb, heterocycloalkyl, aryl, heteroaryl, aryl(Ci-C4)alkyl, and
heteroaryl(Ci-C4)alkyl;
Ra and Rb are each independently hydrogen, (Ci-C8)alkyl, (C2-C8)alkenyl, (C2-Cg)alkynyl, (C3-C8)cycloalkyl, (C5-C8)cycloalkenyl, (C6-Cio)bicycloalkyl, heterocycloalkyl, aryl, heteroaryl, wherein said (Ci-C8)alkyl, (C2-Cg)alkenyl, (C2-C8)alkynyl, cycloalkyl, cycloalkenyl, bicycloalkyl, heterocycloalkyl ,aryl or heteroaryl group is optionally substituted by 1, 2 or 3 groups independently selected from halo, hydroxyl, (Ci-C4)alkoxy, amino, (Ci-C4)alkylamino,
((Ci-C4)alkyl)((Ci-C4)alkyl)amino, -C02H, -C02(CrC4)alkyl, -CONH2, -CONH(C C4)alkyl, - CON((Ci-C4)alk l)((Ci-C4)alkyl), -S02(Ci-C4)alkyl, -S02NH2 ,-S02NH(d-d)alkyl, and - S02N((d-d)alkyl)((d-d)alkyl);
each Rc is independently (Ci-C4)alkylamino, -NRaS02Rb, -SORa, -S02Ra, -NRaC(0)ORa, -NRaRb, or -C02Ra;
or Ra and Rb taken together with the nitrogen to which they are attached represent a 5-8 membered saturated or unsaturated ring, optionally containing an additional heteroatom selected from oxygen, nitrogen, and sulfur, wherein said ring is optionally substituted by 1 , 2 or 3 groups independently selected from (Ci-C4)alkyl, (Ci-C4)haloalkyl, amino, (Ci-C4)alkylamino,
((C C4)alkyl)((Ci-C4)alkyl)amino, hydroxyl, oxo, (C C4)alkoxy, and (d-d)alkoxy(d-d)alkyl, wherein said ring is optionally fused to a (d-d)cycloalkyl, heterocycloalkyl, aryl, or heteroaryl ring;
or Ra and Rb taken together with the nitrogen to which they are attached represent a 6- to 10-membered bridged bicyclic ring system optionally fused to a (C3-C8)cycloalkyl,
heterocycloalkyl, aryl, or heteroaryl ring. An aryl or heteroaryl group in this particular subgroup A is selected independently from the group consisting of furan, thiophene, pyrrole, oxazole, thiazole, imidazole, pyrazole, oxadiazole, thiadiazole, triazole, tetrazole, benzofuran, benzothiophene, benzoxazole, benzothiazole, phenyl, pyridine, pyridazine, pyrimidine, pyrazine, triazine, tetrazine, quinoline, cinnoline, quinazoline, quinoxaline, and naphthyridine or another aryl or heteroaryl group as follows:
Figure imgf000161_0001
wherein in (1),
A is O NH, or S; B is CH or N, and C is hydrogen or d- alkyl; or
Figure imgf000161_0002
wherein in (2),
D is N or C o tionally substituted by hydrogen or d-d alkyl; or
Figure imgf000161_0003
wherein in (3),
E is NH or CH2; F is O or CO; and G is NH or CH2; or
Figure imgf000162_0001
wherein in (4),
J is 0 S or CO; or
Figure imgf000162_0002
wherein in (5),
Q is CH or N;
M is CH or N; and
L/(5) is hydrogen, halo, amino, cyano, (C C8)alkyl, (C3-C8)cycloalkyl, -CORa, -C02Ra, - CONRaRb, -CONRaNRaRb, -S02Ra, -S02NRaRb, -NRaRb, -NRaC(0)Rb ,-NRaS02Rb, - NRaS02NRaRb, -NRaNRaRb, -NRaNRaC(0)Rb, -NRaNRaC(0)NRaRb, -ORa,
wherein any (Ci-C8)alkyl, (C3-C8)cycloalkyl, group is optionally substituted by 1, 2 or 3 groups independently selected from (Ci-C6)alkyl, (C3-C8)cycloalkyl,
(C5-C8)cycloalkenyl, (C1-C6)haloalkyl, cyano, -CORa, -C02Ra, -CONRaRb, -SRa, -SORa, -S02Ra, -S02NRaRb, nitro, -NRaRb, -NRaC(0)Rb, -NRaC(0)NRaRb, -NRaC(0)ORa, - NRaS02Rb, -NRaS02NRaRb, -ORa, -OC(0)Ra, -OC(0)NRaRb; wherein Ra andRb are defined as above; or
Figure imgf000162_0003
wherein in 6,
L/(6) is NH or CH2; or
Figure imgf000162_0004
wherein in 7,
M/(7) is hydrogen, halo, amino, cyano, (C1-C8)alkyl, (C3-C8)cycloalkyl, heterocycloalkyl, -CORa, -C02Ra, -CONRaRb, -CONRaNRaRb, -S02Ra, -S02NRaRb, - NRaRb, -NRaC(0)Rb ,-NRaS02Rb, -NRaS02NRaRb, -NRaNRaRb, -NRaNRaC(0)Rb, - NRaNRaC(0)NRaRb, -ORa,
wherein any (Ci-C8)alkyl, (C3-C8)cycloalkyl, heterocycloalkyl group is optionally substituted by 1 , 2 or 3 groups independently selected from (Ci-C6)alkyl,
(C3-C8)cycloalkyl, (C5-C8)cycloalkenyl, (Ci-C6)haloalkyl, cyano, -CORa, -C02Ra, - CONRaRb, -SRa, -SORa, -S02Ra, -S02NRaRb, nitro, -NRaRb, -NRaC(0)Rb, - NRaC(0)NRaRb, -NRaC(0)ORa, -NRaS02Rb, -NRaS02NRaRb, -ORa, -OC(0)Ra, - OC(0)NRaRb; wherein Ra and Rb are defined as above; or
Figure imgf000163_0001
wherein in (8),
P is CH2, NH, O, or S; Q/(8) is CH or N; and n is 0-2; or
Figure imgf000163_0002
wherein in (9),
S/(9) and T(9) is C, or S/(9) is C and T(9) is N, or S/(9) is N and T/(9) is C;
R is hydrogen, amino, methyl, trifluoromethyl, halo;
U is hydrogen, halo, amino, cyano, nitro, trifluoromethyl, (Ci-C8)alkyl, (C3-C8)cycloalkyl, -CORa, -C02Ra, -CONRaRb, -S02Ra, -S02NRaRb, -NRaRb, -NRaC(0)Rb ,-NRaS02Rb, - NRaS02NRaRb, -NRaNRaRb, -NRaNRaC(0)Rb, , -ORa, 4-(lH-pyrazol-4-yl),
wherein any (Ci-C8)alkyl, (C3-C8)cycloalkyl, group is optionally substituted by 1, 2 or 3 groups independently selected from (Ci-C6) lkyl, (C3-C8)cycloalkyl,
(C5-C8)cycloalkenyl, (C1-C6)haloalkyl, cyano, -CORa, -C02Ra,-CONRaRb, -SRa, -SORa, - S02Ra, -S02NRaRb, nitro, -NRaRb, -NRaC(0)Rb, -NRaC(0)NRaRb, -NRaC(0)ORa, - NRaS02Rb, -NRaS02NRaRb, -ORa, -OC(0)Ra, -OC(0)NRaRb; wherein Ra andRb are defined as above.
3. A compound of formula (I) according to claim 1 or 2 wherein:
X and Z are selected independently from the group consisting of (Ci-C8)alkyl, (C3-
C8)cycloalkyl, heterocycloalkyl, aryl, heteroaryl, -NRaRb, and -ORa; Y is H;
R1 is (Ci-C8)alkyl, (d-d)cycloalkyl, or heterocycloalkyl;
R3 is hydrogen, (Ci-Cs)alkyl or halo;
R6 is hydrogen, halo, cyano, trifluoromethyl, amino, (Ci-Cg)alkyl, (C3-C8)cycloalkyl;, aryl, heteroaryl, acylamino; (d-d)alkynyl, arylalkynyl, heteroarylalkynyl; -S02Ra; -S02NRaRb, or - NRaS02Rb;
wherein any (d-Cg)alkyl, (C3-C8)cycloalkyl, (C2-C8)alkynyl, arylalkynyl, heteroarylalkynyl group is optionally substituted by 1 , 2 or 3 groups independently selected from halo, (Ci-C6)alkyl, (C3-C8)cycloalkyl, (C5-C8)cycloalkenyl, (Ci-C6)haloalkyl, cyano, -CORa, -C02Ra, -CONRaRb, -SRa, -SORa, -S02Ra, -S02NRaRb, nitro, -NRaRb, - NRaC(0)Rb, -NRaC(0)NRaRb, -NRaC(0)ORa, -NRaS02Rb, -NRaS02NRaRb, -ORa, - OC(0)Ra, -OC(0)NRaRb, heterocycloalkyl, aryl, heteroaryl, aryl(CrC4)alkyl, and heteroaryl(Ci-C4)alkyl;
Ra and Rb are each independently hydrogen, (Ci-C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, (C3-C8)cycloalkyl, (C5-C8)cycloalkenyl, (C6-do)bicycloalkyl, heterocycloalkyl, aryl, heteroaryl, wherein said (Ci-C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, cycloalkyl, cycloalkenyl, bicycloalkyl, heterocycloalkyl ,aryl or heteroaryl group is optionally substituted by 1, 2 or 3 groups
independently selected from halo, hydroxyl, (Ci-C4)alkoxy, amino, (Ci-C4)alkylamino,
((d-d)alkyl)((d-d)alkyl)amino, -C02H, -C02(Ci-C4)alkyl, -CONH2,-CONH(d-d)alkyl, - CON((Ci-C4)alkyl)((Ci-C4)alkyl), -S02(Ci-C4)alkyl, -S02NH2, -S02NH(d-d)alkyl, and - S02N((Ci-C4)alkyl)((Ci-C4)alkyl);
or Ra and Rb taken together with the nitrogen to which they are attached represent a 5-8 membered saturated or unsaturated ring, optionally containing an additional heteroatom selected from oxygen, nitrogen, and sulfur, wherein said ring is optionally substituted by 1 , 2 or 3 groups independently selected from (Ci-C4)alkyl, (d-d)haloalkyl, amino, (Ci-C4)alkylamino,
((d-d)alkyl)((d-d)alkyl)amino, hydroxyl, oxo, (C C4)alkoxy, and (d-d)alkoxy(d-d)alkyl, wherein said ring is optionally fused to a (d-d)cycloalkyl, heterocycloalkyl, aryl, or heteroaryl ring;
or Ra and Rb taken together with the nitrogen to which they are attached represent a 6- to 10-membered bridged bicyclic ring system optionally fused to a (C3-Cg)cycloalkyl,
heterocycloalkyl, aryl, or heteroaryl ring. Aryl and heteroaryl in this definition are selected from the group consisting of furan, thiophene, pyrrole, oxazole, thiazole, imidazole, pyrazole, oxadiazole, thiadiazole, triazole, tetrazole, benzofuran, benzothiophene, benzoxazole,
benzothiazole, phenyl, pyridine, pyridazine, pyrimidine, pyrazine, triazine, tetrazine, quinoline, cinnoline, quinazoline, quinoxaline, and naphthyridine as or a compound of or another aryl or heteroaryl group as follows:
Figure imgf000165_0001
wherein in (1),
A is 0 NH, or S; B is CH or N, and C is hydrogen or Ci-Cg alkyl; or
Figure imgf000165_0002
wherein in (2),
D is N or C o tionally substituted by hydrogen or Ci-Cg alkyl; or
Figure imgf000165_0003
wherein in (3),
E is NH or CH2; F is O or CO; and G is NH or CH2; or
Figure imgf000165_0004
wherein in (4),
J is 0 S or CO; or
Figure imgf000165_0005
wherein in (5),
Q is CH or N;
M is CH or N; and
L/(5) is hydrogen, halo, amino, cyano, (C C8)alkyl, (C3-C8)cycloalkyl, -CORa, -C02Ra, - CONRaRb, -CONRaNRaRb, -S02Ra, -S02NRaRb, -NRaRb, -NRaC(0)Rb -NRaS02Rb, - NRaS02NRaRb, -NRaNRaRb, -NRaNRaC(0)Rb, -NRaNRaC(0)NRaRb, -ORa,
wherein any (Ci-C8)alkyl, (C3-Cs)cycloalkyl, group is optionally substituted by 1, 2 or 3 groups independently selected from (Ci-C6)alkyl, (C3-C8)cycloalkyl,
(C5-C8)cycloalkenyl, (C C6)haloalkyl, cyano, -CORa, -C02Ra, -CONRaRb, -SRa, -SORa, -S02Ra, -S02NRaRb, nitro, -NRaRb, -NRaC(0)Rb, -NRaC(0)NRaRb, -NRaC(0)ORa, - NRaS02Rb, -NRaS02NRaRb, -ORa, -OC(0)Ra, -OC(0)NRaRb,
wherein Raand Rb are defined as above; or
Figure imgf000166_0001
wherein in (6),
L/(6) is NH or CH2; or
Figure imgf000166_0002
wherein in (7),
M/(7) is hydrogen, halo, amino, cyano, (Ci-C8)alkyl, (C3-C8)cycloalkyl, heterocycloalkyl, -CORa, -C02Ra, -CONRaRb, -CONRaNRaRb, -S02Ra, -S02NRaRb, - NRaRb, -NRaC(0)Rb,-NRaS02Rb, -NRaS02NRaRb, -NRaNRaRb, -NRaNRaC(0)Rb, - NRaNRaC(0)NRaRb, -ORa,
wherein any (Ci-C8)alkyl, (C3-C8)cycloalkyl, heterocycloalkyl group is optionally substituted by 1, 2 or 3 groups independently selected from (Ci-C6)alkyl,
(C3-C8)cycloalkyl, (C5-C8)cycloalkenyl, (C C6)haloalkyl, cyano, -CORa, -C02Ra, - CONRaRb, -SRa, -SORa, -S02Ra, -S02NRaRb, nitro, -NRaRb, -NRaC(0)Rb, - NRaC(0)NRaRb, -NRaC(0)ORa, -NRaS02Rb, -NRaS02NRaRb, -ORa, -OC(0)Ra, - OC(0)NRaRb; wherein RaandRb are defined as above; or
Figure imgf000166_0003
wherein in (8),
P is CH2, NH, O, or S; Q/(8) is CH or N; and n is 0-2; or
Figure imgf000167_0001
wherein in (9),
S/(9) and T(9) is C, or S/(9) is C and T(9) is N, or S/(9) is N and T/(9) is C;
R is hydrogen, amino, methyl, trifluoromethyl, halo;
U is hydrogen, halo, amino, cyano, nitro, trifluoromethyl, (Ci-Cg)alkyl, (C3-Cg)cycloalkyl, -CORa, -C02Ra, -CONRaRb, -S02Ra, -S02NRaRb, -NRaRb, -NRaC(0)Rb ,-NRaS02Rb, - NRaS02NRaRb, -NRaNRaRb, -NRaNRaC(0)Rb, , -ORa, 4-(lH-pyrazol-4-yl),
wherein any (Ci-Cg)alkyl, (C3-Cg)cycloalkyl, group is optionally substituted by 1, 2 or 3 groups independently selected from (Ci-C6)alkyl, (C3-Cg)cycloalkyl,
(C5-Cg)cycloalkenyl, (C1-C6)haloalkyl, cyano, -CORa, -C02Ra,-CONRaRb, -SRa, -SORa, - S02Ra, -S02NRaRb, nitro, -NRaRb, -NRaC(0)Rb, -NRaC(0)NRaRb, -NRaC(0)ORa, - NRaS02Rb, -NRaS02NRaRb, -ORa, -OC(0)Ra, -OC(0)NRaRb, wherein Ra and Rb are defined as above;
or a salt thereof.
4. A compound according to any one of claims 1 , 2 or 3 wherein:
X is methyl, ethyl, n-propyl, isopropyl , cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, phenyl, trifluoromethyl, tetrahydropyran, hydroxymethyl, methoxymethyl, or benzyl;
Y is H;
Z is methyl, ethyl, n-propyl, isopropyl, trifluoromethyl, or benzyl;
R1 is isopropyl, tert-butyl, cyclobutyl, cyclopentyl, cyclohexyl, (1 - methylethyl)cyclopropyl, 1 , 1 -dioxo-tetrahydrothiophene-3-yl, 1 -Me-piperidin-4-yl,
tetrahydrofuran-3-yl, tetrahydropyran-4-yl, A^, V-dimethyl- l-propanaminyl, benzyl, or 4-pyridyl;
R3 is H, methyl, or Br; and
R6 is methyl, bis(l , l -dimethylethyl), bis(l -methylethyl), cyclopropyl, propyl, dimethylamino, ethylamino, (2-hydroxyethyl)amino, 2-propen-l-ylamino, 1 -piperazinyl, 1- piperidinyl, 4-morpholinyl, 4-piperidinylamino, tetrahydro-2H-pyran-4-ylamino, phenylamino, (phenylmethyl)amino, (4-pyridinylmethyl)amino, [2-(2-pyridinylamino)ethyl]amino, 2- (dimethylamino)ethyl]amino, 4-pyridinylamino , 4-(aminocarbonyl)phenyl]amino, 3-hydroxy-3- methyl-l-butyn-l -yl, 4-pyridinylethynyl, phenylethynyl, 2-furanyl, 3-thienyl; lH-pyrazol-4-yl, 1H- indazol-5-yl, lH-indazol-6-yl, 3-methyl-lH-indazol-5-yl, lH- l ,2,3-benzotriazol-5-yl, 2-oxo-2,3- dihydro- lH-benzimidazol-5-yl, 2-oxo-2,3-dihydro- lH-indol-5-yl, 2-oxo-2,3-dihydro- lH-indol-6- yl, 2, 1 ,3-benzoxadiazol-5-yl, 2-amino-6-quinazolinyl, 2,4-dioxo- l,2,3,4-tetrahydro-5-pyrimidinyl,
2- amino-5-pyrimidinyl, 7-oxo-l,5,6,7-tetrahydro-l,8-naphthyridin-3-yl, phenyl, 2-methylphenyl, 2- nitrophenyl, 2-phenylethyl, 3-aminophenyl, 4-aminophenyl, 4-chlorophenyl, 4-fluorophenyl, 4- (methyloxy)phenyl, 3-(acetylamino)phenyl, 4-(acetylamino)phenyl, 4-(aminocarbonyl)phenyl, 4- (lH-pyrazol-4-yl)phenyl, 4-(aminosulfonyl)phenyl, 4-(methylsulfonyl)phenyl, 4- [(dimethylamino)sulfonyl]phenyl, 4- [(methylamino)carbonyl]phenyl, 4-
[(methylamino)sulfonyl]phenyl, 4-[(methylsulfonyl)amino]phenyl, 3-pyridinyl, 4-pyridinyl, 2-(4- morpholinyl)-4-pyridinyl, 2-amino-4-pyridinyl, 5-(methyloxy)-3-pyridinyl, 5-(methylsulfonyl)-3- pyridinyl, 5 - [(cyclopropylsulfonyl)amino ] - 6-(methy loxy) -3 -pyridinyl, 5 - [(phenylsulfonyl)amino] -
3- pyridinyl, 6-(4-methyl-l-piperazinyl)-3 -pyridinyl, 6-(4-morpholinyl)-3-pyridinyl, 6- (acetylamino)-3-pyridinyl, 6-(dimethylamino)-3-pyridinyl, 6-(methyloxy)-3-pyridinyl, 6- [(methylamino)carbonyl] -3 -pyridinyl, 6-[(methylamino)sulfonyl]-3-pyridinyl, 6-methyl-3- pyridinyl, 4-pyridinyloxy;
or a salt thereof.
5. A compound of formula (I) according to claim 1 or 2 wherein:
X is (C1-C8)alkyl or (C3-C8)cycloalkyl;
Y is H;
Z is (Ci-C3)alkyl;
R1 is (C3-C8)alkyl, (C3-C8)cycloalkyl, or heterocycloalkyl;
R3 is hydrogen, (Ci-C3)alkyl or halo;
R6 is selected from the group consisting of hydrogen, halo, cyano, trifluoromethyl, amino, (C!-C8)alkyl, (C3-C8)cycloalkyl, aryl, heteroaryl, acylamino, (C2-C8)alkynyl, arylalkynyl, heteroarylalkynyl, -S02Ra; -S02NRaRb , and -NRaS02Rb ;
wherein any (C C8)alkyl, (C3-C8)cycloalkyl, (C2-C8)alkynyl, arylalkynyl,
heteroarylalkynyl group is optionally substituted by 1 , 2 or 3 groups independently selected from
Figure imgf000168_0001
-S(C1-C6)alkyl(Rc)1.2, (C C8)alkyl- heterocycloalkyl, (C3-C8)cycloalkyl-heterocycloalkyl, halo, (Ci-C6)alkyl,
(C3-C8)cycloalkyl, (C5-C8)cycloalkenyl, (C1-C6)haloalkyl, cyano, -CORa, -C02Ra, - CONRaRb, -SRa, -SORa, -S02Ra, -S02NRaRb, nitro, -NRaRb, -NRaC(0)Rb, - NRaC(0)NRaRb, -NRaC(0)ORa, -NRaS02Rb, -NRaS02NRaRb, -ORa, -OC(0)Ra, - OC(0)NRaRb, heterocycloalkyl, aryl, heteroaryl, aryl(Ci-C4)alkyl, and
heteroaryl(Ci-C4)alkyl;
Ra and Rb are each independently hydrogen, (Ci-Cs)alkyl, (C2-C8)alkenyl, (C2-Cg)alkynyl, (C3-C8)cycloalkyl, (C5-C8)cycloalkenyl, (C6-Cio)bicycloalkyl, heterocycloalkyl, aryl, heteroaryl, wherein said (Ci-Cg)alkyl, (C2-C8) lkenyl, (C2-C8)alkynyl, cycloalkyl, cycloalkenyl, bicycloalkyl, heterocycloalkyl ,aryl or heteroaryl group is optionally substituted by 1, 2 or 3 groups independently selected from halo, hydroxyl, (d-d)alkoxy, amino, (d-d)alkylamino,
((Ci-C4)alkyl)((Ci-C4)alkyl)amino, -C02H, -C02(d-d)alkyl, -CONH2, -CONH(d-d)alkyl, - CON((Ci-C4)alkyl)((Ci-C4)alkyl), -S02(Ci-C4)alkyl, -S02NH2 -S02NH(Ci-C4)alkyl, and - S02N((Ci-C4)alkyl)((Ci-C4)alkyl);
each Rc is independently (C C4)alkylamino, -NRaS02Rb, -SORa, -S02Ra, -NRaC(0)ORa, -NRaRb, or -C02Ra;
or a pharmaceutically acceptable salt thereof.
6. A compound according to any preceeding claim wherein R6 is selected from the group consisting of: halo, cyano, trifluoromethyl, amino, (d-Cg)alkyl, (d-d)cycloalkyl, acylamino, (C2- C8)alkynyl, arylalkynyl, heteroarylalkynyl, -S02Ra; -S02NRaRb , and -NRaS02Rb ; or a pharmaceutically acceptable salt thereof.
7. A compound according to any preceeding claim wherein R6 is aryl or heteroaryl, wherein said aryl or heteroaryl is optionally substituted by 1, 2 or 3 groups independently selected from - 0(C1-C6)alkyl(Rc)1_2, -SCd-QDalky RV,, -(C1-C6)alkyl(Rc)1.2, (C1-C8)alkyl-heterocycloalkyl, (C3-C8)cycloalkyl-heterocycloalkyl, halo, (Ci-C6)alkyl, (C3-C8)cycloalkyl, (C5-C8)cycloalkenyl, (C1-C6)haloalkyl, cyano, -CORa, -C02Ra, -CONRaRb, -SRa, -SORa, -S02Ra, -S02NRaRb, nitro, - NRaRb, -NRaC(0)Rb, -NRaC(0)NRaRb, -NRaC(0)ORa, -NRaS02Rb, -NRaS02 RaRb, -ORa, - OC(0)Ra, -OC(0)NRaRb, heterocycloalkyl, aryl, heteroaryl, aryl(C C4)alkyl, and
heteroaryl(Ci-C4)alkyl;
Ra and Rb are each independently hydrogen, (Ci-C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, (C3-C8)cycloalkyl, (C5-C8)cycloalkenyl, (C6-C10)bicycloalkyl, heterocycloalkyl, aryl, heteroaryl, wherein said (d-d)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, cycloalkyl, cycloalkenyl, bicycloalkyl, heterocycloalkyl ,aryl or heteroaryl group is optionally substituted by 1, 2 or 3 groups
independently selected from halo, hydroxyl, (d-d)alkoxy, amino, (d-Q)alkylamino,
((d-d)alkyl)((d-d)alkyl)amino, -C02H, -C02(C C4)alkyl, -CONH2, -CONH(C C4)alkyl, - CON((Ci-C4)alkyl)((Ci-C4)alkyl), -S02(Ci-C4)alkyl, -S02NH2 ,-S02NH(d-d)alkyl, and - S02N((d-d)alkyl)((d-d)alkyl);
each Rc is independently (d-d)alkylamino, -NRaS02Rb, -SORa, -S02Ra, -NRaC(0)ORa, -NRaRb, or -C02Ra;
or a pharmaceutically acceptable salt thereof.
8. A compound according to any preceeding claim which is
6-chloro-l -(1 -methylethyl)-_V-[(6-methyl-2-oxo-4-propyl- 1 ,2-dihydro-3-pyridinyl)methyl]- l f-indazole-4-carboxamide;
N-[(4,6-dimethyl-2-oxo- 1 ,2-dihydro-3-pyridinyl)methyl]-6-fluoro- 1 -(1-methylethyl)- IH- indazole-4-carboxamide; 6-Bromo-N-[(4,6-dimethyl-2-oxo-l ,2-dihydro-3-pyridi^^
indazole-4-carboxamide;
6-bromo-N-((4-ethyl-6-methyl-2-oxo- l,2-dihydropyridin-3-yl)methyl)-l -isopropyl-lH- indazole-4-carboxamide;
6-bromo- 1 -( 1 -methylethyl)-iV- { [6-methyl-2-oxo-4-(phenylmethyl)- 1 ,2-dihydro-3- pyridinyl]methyl} - l/f-indazole-4-carboxamide;
6-bromo- 1 -( 1 -methylethyl)-^- [(6-methyl-2-oxo- 1 ,2-dihydro-4,4'-bipyridin-3 -yl)methyl]- l/i-indazole-4-carboxamide;
1 -(1 -methylethyl)-N- { [6-methyl-2-oxo-4-(phenylmethyl)- 1 ,2-dihydro-3-pyridinyl]methyl} - l/i-indazole-4-carboxamide ;
N-((4,6-dimethyl-2-oxo- l,2-dihydropyridin-3-yl)methyl)-6-(4-(2- (dimethylamino)ethoxy)phenyl)- 1 -isopropyl- 1 H-indazole-4-carboxamide;
N-[(4,6-dimethyl-2-oxo- l,2-dihydro-3-pyridinyl)methyl]- 1 -(l-methylethyl)-6-[4-(l - piperazinyl)phenyl]- lH-indazole-4-carboxamide;
V-[(4,6-dimethyl-2-oxo- 1 ,2-dihydro-3-pyridinyl)methyl]- 1 -(l-methylethyl)-6-[6-(l - piperazinyl)-3-pyridinyl]- l i-indazole-4-carboxamide;
6- {4-[(dimethylamino)methyl]phenyl} - 1 -(1 -methylethyl)-N- {[6-methyl-4-(l -methylethyl)- 2-oxo- 1 ,2-dihydro-3-pyridinyl]methyl} - 1 H-indazole-4-carboxamide;
l -isopropyl-N-((4-isopropyl-6-methyl-2-oxo-l,2-dihydropyridin-3-yl)methyl)-6-(6- (piperazin- 1 -yl)pyridin-3 -yl)- 1 H-indazole-4-carboxamide;
N-((4,6-dimethyl-2-oxo- 1 ,2-dihydropyridin-3-yl)methyl)- 1 -isopropyl-6-(l -methyl- 1 H- pyrazol-4-yl)- lH-indazole-4-carboxamide;
6-(4-((dimethylamino)methyl)phenyl)-N-((4-ethyl-6-methyl-2-oxo- l ,2-dihydropyridin-3- yl)methyl)- 1 -isopropyl- 1 H-indazole-4-carboxamide;
l -(l-methylethyl)-N-[(6-methyl-2-oxo-4-propyl-l,2-dihydro-3-pyridinyl)methyl]-6-[6-(l- piperazinyl)-3-pyridinyl]- lH-indazole-4-carboxamide;
N-((4-ethyl-6-methyl-2-oxo- l ,2-dihydropyridin-3-yl)methyl)- l-isopropyl-6-(6-(4- methylpiperazin- 1 -yl)pyridin-3-yl)- 1 H-indazole-4-carboxamide;
1 -isopropyl-N-((6-methyl-2-oxo-4-propyl- 1 ,2-dihydropyridin-3-yl)methyl)-6-(2- methylpyrimidin-5-yl)- lH-indazole-4-carboxamide;
6-[6-(dimethylamino)-3-pyridinyl]-l-(l -methylethyl)-N-[(6-methyl-2-oxo-4-propyl- l ,2- dihydro-3-pyridinyl)methyl]- lH-indazole-4-carboxamide;
l -(l-methylethyl)-N-[(6-methyl-2-oxo-4-propyl-l,2-dihydro-3-pyridinyl)methyl]-6-[6-(4- morpholinyl)-3-pyridinyl]- lH-indazole-4-carboxamide;
6-(2-amino-5-pyrimidinyl)- 1 -( 1 -methylethyl)-N-[(6-methyl-2-oxo-4-propyl- 1 ,2-dihydro-3- pyridinyl)methyl]- 1 H-indazole-4-carboxamide; 6-(6-amino-3-pyridinyl)- l-(l-methylethyl)-N-[(6-methyl-2-oxo-4-propyl-l,2-dihydro-3- pyridinyl)methyl]- 1 H-indazole-4-carboxamide;
l -(l-methylethyl)-N-[(6-methyl-2-oxo-4-propyl-l,2-dihydro-3-pyridinyl)methyl]-6-(5- pyrimidinyl)-lH-indazole-4-carboxamide;
l -(l-methylethyl)-N-[(6-methyl-2-oxo-4-propyl-l,2-dihydro-3-pyridinyl)methyl]-6-[6- (methyloxy)-3-pyridinyl]- lH-indazole-4-carboxamide;
l -(l-methylethyl)-N-[(6-methyl-2-oxo-4-propyl-l,2-dihydro-3-pyridinyl)methyl]-6-[6-(4- methyl- 1 -piperazinyl)-3 -pyridinyl] - lii-indazole-4-carboxamide;
l -(l-methylethyl)-N-[(6-methyl-2-oxo-4-propyl-l,2-dihydro-3-pyridinyl)methyl]-6-(2- methyl-3-pyridinyl)- 1 H-indazole-4-carboxamide;
l -(l-methylethyl)-N-[(6-methyl-2-oxo-4-propyl-l,2-dihydro-3-pyridinyl)methyl]-6-(6- methyl-3-pyridinyl)- 1 H-indazole-4-carboxamide;
6-[6-(dimethylamino)-3-pyridinyl]-N-[(4,6-dimethyl-2-oxo- l ,2-dihydro-3- pyridinyl)methyl]- 1 -( 1 -methylethyl)- 1 H-indazole-4-carboxamide;
N-[(4,6-dimethyl-2-oxo- l,2-dihydro-3-pyridinyl)methyl]- l -(l-methylethyl)-6-[6- (trifluoromethyl)-3-pyridinyl]-lH-indazole-4-carboxamide;
6- {3-[(dimethylamino)methyl]phenyl} -N-[(4,6-dimethyl-2-oxo-l,2-dihydro-3- pyridinyl)methyl]- l-(l -methylethyl)- lif-indazole-4-carboxamide;
6- {4-[(dimethylamino)methyl]phenyl} -N-[(4,6-dimethyl-2-oxo- 1 ,2-dihydro-3- pyridinyl)methyl]- 1 -( 1 -methylethyl)- 1 H-indazole-4-carboxamide;
N-((l ,2-dihydro-6-methyl-2-oxo-4-propylpyridin-3-yl)methyl)- 1 -isopropyl-6-(thiophen-3- yl)-lH-indazole-4-carboxamide;
1 -isopropyl-N-((6-methyl-2-oxo-4-propyl- 1 ,2-dihydropyridin-3-yl)methyl)-6-(thiophen-3- yl)-lH-indazole-4-carboxamide;
6- {4- [(dimethylamino)methyl]phenyl} - 1 -(1 -methylethyl)-N-[(6-methyl-2-oxo-4-propyl- 1 ,2-dihydro-3-pyridinyl)methyl]- lH-indazole-4-carboxamide hydrochloride salt;
6- {3-[(dimethylamino)methyl]phenyl} - 1 -(1 -methylethyl)-N-[(6-methyl-2-oxo-4-propyl- 1 ,2-dihydro-3-pyridinyl)methyl]- lH-indazole-4-carboxamide hydrochloride salt;
N-((l ,2-dihydro-6-methyl-2-oxo-4-propylpyridin-3-yl)methyl)- l -isopropyl-6-(lH-pyrazol- 4-yl)- lH-indazole-4-carboxamide;
N-((l ,2-dihydro-6-methyl-2-oxo-4-propylpyridin-3-yl)methyl)- l -isopropyl-6-(lH- pyrrolo[2,3-b]pyridin-5-yl)- lH-indazole-4-carboxamide;
6-(4-(dimethylamino)piperidin- 1 -yl)- 1 -isopropyl-N-((6-methyl-2-oxo-4-propyl- 1 ,2-dihydropyridin- 3 -yl)methyl)- 1 H-indazole-4-carboxamide ;
6-(4-((dimethylamino)methyl)piperidin- 1 -yl)- 1 -isopropyl-N-((6-methyl-2-oxo-4-propyl- 1 ,2- dihydropyridin-3-yl)methyl)- lH-indazole-4-carboxamide ; N-((4-ethyl-6-methyl-2-oxo-l,2-dmydropyridin-3-yl)methy
1 H-indazole-4-carboxamide;
N-((4,6-dimethy 1-2-oxo- 1 ,2-dihydropyridin-3-yl)methyl)- 1 -isopropyl-6-morpholino- 1 H- indazole-4-carboxamide;
N-((4-cyclohexyl-6-methyl-2-oxo-l,2-dihydropyridin-3-yl)methyl)-6-cyclopropyl-l- isopropyl-lH-indazole-4-carboxamide;
6-bromo- 1 -ethyl- lH-indazole-4-carboxy lie acid (6-methyl-2-oxo-4-propyl- l, 2-dihydro- pyridin-3 -ylmethyl)-amide;
6-bromo-N-((6-methyl-2-oxo-4-propyl- 1 ,2-dihydropyridin-3-yl)methyl)- 1 -propyl- 1H- indazole-4-carboxamide;
6-bromo- 1 -cyclopentyl-N-((4,6-dimethyl-2-oxo- 1 ,2-dihydropyridin-3-yl)methyl)- 1 H- indazole-4-carboxamide;
6-bromo- 1 -cyclopentyl-N-((4-ethyl-6-methyl-2-oxo- 1 ,2-dihydropyridin-3-yl)methyl)- 1H- indazole-4-carboxamide;
1 -cyclopentyl-N-((4,6-dimethyl-2-oxo- 1 ,2-dihydropyridin-3-yl)methyl)-6-(2-(4- methylpiperazin-l-yl)pyrimidin-5-yl)-lH-indazole-4-carboxamide;
l-cyclopentyl-N-((4,6-dimethyl-2-oxo-l,2-dihydropyridin-3-yl)methyl)-6-(3-(pyrrolidin-l- yl)phenyl)-lH-indazole-4-carboxamide;
1 -cyclopentyl-N-((4,6-dimethyl-2-oxo- 1 ,2-dihydropyridin-3-yl)methyl)-6-(4- methylthiophen-2-yl)-lH-indazole-4-carboxamide;
1 -cyclopentyl-N-((4,6-dimethyl-2-oxo- 1 ,2-dihydropyridin-3-yl)methyl)-6-(2-(piperazin- 1 - yl)pyrimidin-5-yl)- 1 H-indazole-4-carboxamide;
1 -cyclopentyl-N-((4,6-dimethyl-2-oxo- 1 ,2-dihydropyridin-3-yl)methyl)-6-(3-(3- hydroxypropyl)phenyl)- 1 H-indazole-4-carboxamide;
1 -cyclopentyl-N-((4,6-dimethyl-2-oxo- 1 ,2-dihydropyridin-3-yl)methyl)-6-(3- (trifluoromethoxy)phenyl)- 1 H-indazole-4-carboxamide;
1 -cyclopentyl-N-((4,6-dimethyl-2-oxo- 1 ,2-dihydropyridin-3-yl)methyl)-6-(6- methoxypyridin-3-yl)-lH-indazole-4-carboxamide;
1 -cyclopentyl-N-((4,6-dimethyl-2-oxo- 1 ,2-dihydropyridin-3-yl)methyl)-6-(4- fluorophenyl)-lH-indazole-4-carboxamide; l-cyclopentyl-N-[(4,6-dimethyl-2-oxo- l,2-dihydro-3-pyridinyl)methyl]-6-(l -methyl- 1H- pyrazol-4-yl)-lH-indazole-4-carboxamide; l-cyclopentyl-N-((6-methyl-2-oxo-4-propyl- l,2-dihydropyridin-3-yl)methyl)-6-(6-(4- methylpiperazin- 1 -yl)pyridin-3-yl)- 1 H-indazole-4-carboxamide; l -cyclopentyl-N-[(4,6-dimethyl-2-oxo- l,2-dihydro-3^yridinyl)methyl]-6-[6-(l- piperazinyl)-3-pyridinyl]- lH-indazole-4-carboxamide;
1 -cyclopentyl-N-((4,6-dimethyl-2-oxo- 1 ,2-dihydropyridin-3-yl)methyl)-6-(4- ((dimethylamino)methyl)phenyl)- 1 H-indazole-4-carboxamide;
6-chloro- l-cyclopentyl-N-[(4,6-dimethyl-2-oxo- l ,2-dihydro-3-pyridinyl)methyl]-l - indazole-4-carboxamide;
6-chloro- l-cyclopentyl-N-[(6-methyl-2-oxo-4-propyl-l,2-dihydro-3-pyridinyl)methyl]- li/- indazole-4-carboxamide;
6-cyano- 1 -cyclopentyl-Ar-[(4,6-dimethyl-2-oxo- 1 ,2-dihydro-3-pyridinyl)methyl]- \H- indazole-4-carboxamide;
6-(aminomethyl)- 1 -cyclopentyl- V-[(4,6-dimethyl-2-oxo- 1 ,2-dihydro-3-pyridinyl)methyl]- l/f-indazole-4-carboxamide;
6-amino- 1 -cyclopentyl-iV-[(4,6-dimethyl-2-oxo- 1 ,2-dihydro-3-pyridinyl)methyl]-l//- indazole-4-carboxamide;
l -cyclopentyl-jV-[(4,6-dimethyl-2-oxo-l,2-dihydro-3-pyridinyl)methyl]-6- [(phenylsulfonyl)amino] - l/f-indazole-4-carboxamide;
1 -cyclopentyl-jV-[(4,6-dimethyl-2-oxo- 1 ,2-dihydro-3 -pyridinyl)methyl] -6-(3 -hydroxy-3 - methyl- 1 -butyn- 1 -yl)- l/ -indazole-4-carboxamide;
6-bromo- 1 -cyclopropyl-N- [(4,6-dimethyl-2-oxo- 1 ,2-dihydro-3 -pyridinyl)methyl] - 1 H- indazole-4-carboxamide;
6-bromo-l-cyclopropyl-N-[(6-methyl-2-oxo-4-propyl- l ,2-dihydro-3-pyridinyl)methyl]- li7- indazole-4-carboxamide;
1 -cyclopropyl-6- {4-[(dimethylamino)methyl]phenyl} - V-[(4,6-dimethyl-2-oxo- 1 ,2-dihydro- 3-pyridinyl)methyl]- lif-indazole-4-carboxamide;
1 -cyclopropyl-6- {4- [(dimethylamino)methyl]phenyl} -TV- [(6-methyl-2-oxo-4-propyl-l, 2- dihydro-3-p} idinyl)methyl]- l f-indazole-4-carboxamide;
6-bromo-N-((l,2-dihydro-4,6-dimethyl-2-oxopyridin-3-yl)methyl)-l-isopropyl-3-methyl- lH-indazole-4-carboxamide ;
6-bromo- l-isopropyl-3 -methyl-N-((6-methyl-2-oxo-4-propyl-l,2-dihydropyridin-3- yl)methyl)- 1 H-indazole-4-carboxamide;
1 -isopropyl-3-methyl-N-((6-methyl-2-oxo-4-propyl- 1 ,2-dihydropyridin-3-yl)methyl)-6-(2- (4-methylpiperazin- l-yl)pyridin-4-yl)- lH-indazole-4-carboxamide;
3-methyl- l -(l-methylethyl)-N-[(6-methyl-2-oxo-4-propyl- l,2-dihydro-3- pyridinyl)methyl]-6-[6-(l-piperazinyl)-3-pyridinyl]- lH-indazole-4-carboxamide;
6-(6-(dimethylamino)pyridin-3-yl)-N-((l,2-dihydro-4,6-dimethyl-2-oxopyridin-3- yl)methyl)- 1 -isopropy 1-3 -methyl- 1 H-indazole-4-carboxamide; 6-(3-((dimethylamino)methyl)phenyl)-N-((l,2-dihydro-4,6-dimethyl-2-oxopyridin-3- yl)methyl)- 1 -isopropyl- 1 H-indazole-4-carboxamide;
N-((4,6-dimethyl-2-oxo-l,2-dihydropyridin-3-yl)methyl)-6-(4-fluorophenyl)- l -isopropyl- 3 -methyl- 1 H-indazole-4-carboxamide;
N-((l ,2-dihydro-4,6-dimethyl-2-oxopyridin-3-yl)methyl)- 1 -isopropyl-3-methyl-6-(6-(4- methylpiperazin- 1 -yl)pyridin-3 -yl)- 1 H-indazole-4-carboxamide;
6-(4-((dimethylamino)methyl)phenyl)-N-((l,2-dihydro-4,6-dimethyl-2-oxopyridin-3- yl)methyl)- 1 -isopropy 1-3 -methyl- 1 H-indazole-4-carboxamide;
N-((l ,2-dihydro-4,6-dimethyl-2-oxopyridin-3-yl)methyl)- 1 -isopropyl-3-methyl-6-(6- (piperazin-l-yl)pyridin-3-yl)-lH-indazole-4-carboxamide hydrochloride;
N-((4,6-dimethyl-2-oxo-l,2-dihydropyridin-3-yl)methyl)- l -isopropyl-3-methyl-6-(6-(4- methylpiperazin- 1 -yl)pyridin-2-yl)- 1 H-indazole-4-carboxamide;
N-((l ,2-dihydro-4,6-dimethyl-2-oxopyridin-3-yl)methyl)- 1 -isopropyl-3-methyl-6-(pyridin- 3-yl)- lH-indazole-4-carboxamide;
N-((l ,2-dihydro-6-methyl-2-oxo-4-propylpyridin-3-yl)methyl)- 1 -isopropyl-3-methyl-6- (lH-pyrrolo[2,3-b]pyridin-5-yl)- lH-indazole-4-carboxamide;
N-((l ,2-dihydro-4,6-dimethyl-2-oxopyridin-3-yl)methyl)- l -isopropyl-3-methyl-6-(lH- pyrazol-4-yl)- 1 H-indazole-4-carboxamide;
N-((l ,2-dihydro-4,6-dimethyl-2-oxopyridin-3-yl)methyl)- 1 -isopropyl-3-methyl-6-(pyridin- 3-yl)- lH-indazole-4-carboxamide;
N-[(4,6-dimethyl-2-oxo- 1 ,2-dihydro-3-pyridinyl)methyl] -3 -methyl- 1 -(l-methylethyl)-6-(6- methyl-3-pyridinyl)- l f-indazole-4-carboxamide;
6-[6-(dimethylamino)-3-pyridinyl]-3-methyl-l-(l -methylethyl)-Ai-[(6-methyl-2-oxo-4-propyl- l ,2- dihydro-3-pyridmyl)methyl]- lii-indazole-4-carboxamide;
6- {3-[(dimethylamino)methyl]phenyl} -3-methyl- 1 -(1 -methylethyl)-A^-[(6-methyl-2-oxo-4-propyl- 1 ,2-dihydro-3-pyridinyl)methyl]- l/ -indazole-4-carboxamide
6- {4-[(dimethylamino)methyl]phenyl} -3-methyl- 1 -(1 -methylethyl)-A¾6-methyl-2-oxo-4-propyl- 1 ,2-dihydro-3-pyridinyl)methyl]- lif-indazole-4-carboxamide;
3-methyl- 1 -(1 -methylethyl)-N-[(6-methyl-2-oxo-4-propyl-l,2-dihydro-3-pyridinyl)methyl]-6-(3- pyridinyl)- l/i-indazole-4-carboxamide;
3-methyl- l -(l-methylethyl)-N-[(6-methyl-2-oxo-4-propyl-l,2-dihydro-3-pyridinyl)methyl]-6-[6-(4- methyl- 1 -piperazinyl)-3 -pyridinyl] - lii-indazole-4-carboxamide; 6-cyano-N-((4,6-dimethyl-2-oxo-l,2-dihydropyridin-3-yl)methyl)- l -isopropyl-3-methyl- 1 H-indazole-4-carboxamide;
1 -cyclopentyl-6-(cyclopropylsulfonyl)-N-((4,6-dimethyl-2-oxo- 1 ,2-dihydropyridin-3-yl)methyl)- lH-indazole-4-carboxamide ;
6-(cyclopropylsulfonyl)-N-((4-ethyl-6-methyl-2-oxo- l ,2-dihydropyridin-3-yl)methyl)-l-isopropyl- lH-indazole-4-carboxamide ;
N-((4-ethyl-6-methyl-2-oxo- 1 ,2-dihydropyridin-3 -yl)methyl)- 1 -isopropyl-6-(methylsulfonyl)- 1 H- indazole-4-carboxamide;
V-[(4,6-dimethyl-2-oxo- 1 ,2-dihydro-3-pyridinyl)methyl]- 1 -(l-methylethyl)-6-(methylsulfonyl)- \H indazole-4-carboxamide;
N- [(4, 6-dimethy 1-2-oxo- 1 ,2-dihydro-3 -pyridinyl)methyl] - 1 -( 1 -methylethyl)-6-(4- morpholinylsulfonyl)-lH-indazole-4-carboxamide;
6- [(cyclopropylamino)sulfonyl] -N- [(4,6-dimethyl-2-oxo- 1 ,2-dihydro-3 -pyridinyl)methyl] - l -(l -methylethyl)-l f-indazole-4-carboxamide;
N- [(4,6-dimethyl-2-oxo- 1 ,2-dihydro-3-pyridinyl)methyl]- 1 -(1 -methylethyl)-6-(l - pyrrolidinylsulfonyl)- l/i-indazole-4-carboxamide ;
6-({[3-(dimethylamino)propyl]amino} sulfonyl)-A^-[(4,6-dimethyl-2-oxo- l ,2-dihydro-3- pyridinyl)methyl]- 1 -(1 -methylemyl)- l/f-indazole-4-carboxamide ;
N-((4,6-dimethyl-2-oxo- l,2-dihydropyridin-3-yl)methyl)- 1 -isopropyl-6-((4- methylpiperazin- 1 -yl)sulfonyl)- lH-indazole-4-carboxamide ;
6-bromo-N-((4-(sec-butyl)-6-methyl-2-oxo- 1 ,2-dihydropyridin-3-yl)methyl)- 1 -isopropyl 1 H-indazole-4-carboxamide;
6-Bromo- 1 -(1 -methylethyl)-N-[(6-methyl-2-oxo-4-propyl- l,2-dihydro-3- pyridinyl)methyl]- 1 H-indazole-4-carboxamide; or
6-Bromo- 1 -(1 -methylethyl)-N- { [6-methyl-4-( 1 -methylethyl)-2-oxo- 1 ,2-dihydro-3- pyridinyl]methyl} - l/f-indazole-4-carboxamide;
or a pharmaceutically acceptable salt thereof.
9. A compound according to any preceeding claim which is:
6-bromo-yV- [(4,6-dimethyl-2-oxo- 1 ,2-dihydro-3-pyridinyl)methyl]- 1 -(1 -methylethyl)- \H- indazole-4-carboxamide;
N- [(4,6-dimethyl-2-oxo- 1 ,2-dihydro-3-pyridinyl)methyl]- 1 -(1 -methylethyl)-6-phenyl- \H- indazole-4-carboxamide; N-[(4,6-dimethyl-2-oxo- 1 ,2-dihydro-3-pyridinyl)methyl]- 1 -(l-methylethyl)-6-(4- pyridinyl)- l f-indazole-4-carboxamide;
N-[(4,6-dimethyl-2-oxo-l,2-dihydro-3-pyridinyl)methyl]-6- {4- [(methylamino)sulfonyl]phenyl} - 1 -( 1 -methylethyl)- l//-indazole-4-carboxamide;
N- [(4,6-dimethyl-2-oxo- 1 ,2-dihydro-3-pyridinyl)methyl]- 1 -(1 -methylethyl)-6- [6-(4- methyl- 1 -piperazinyl)-3 -pyridinyl] - l /-indazole-4-carboxamide;
N-[(4,6-dimethyl-2-oxo-l,2-dihydro-3-pyridinyl)methyl]-l-(l-methylethyl)-6-(2-oxo-2,3- dihydro- l f-benzimidazol-5-yl)- l /-indazole-4-carboxamide;
6- [6-(acetylamino)-3 -pyridinyl] -N- [(4,6-dimethyl-2-oxo- 1 ,2-dihydro-3 -pyridinyl)methyl] - 1-(1 -methylethyl)- \H- indazole-4-carboxamide;
N- [(4,6-dimethyl-2-oxo- 1 ,2-dihydro-3-pyridinyl)methyl]- 1 -(1 -methylethyl)-6-(3- pyridinyl)- l /-indazole-4-carboxamide;
N-[(4,6-dimethyl-2-oxo-l,2-dihydro-3-pyridinyl)methyl]-l-(l-methylethyl)-6-[6-(4- morpholinyl)-3-pyridinyl]- lii-indazole-4-carboxamide;
N-[(4,6-dimethyl-2-oxo- 1 ,2-dihydro-3-pyridinyl)methyl]- 1 -(l-methylethyl)-6-[6- (methyloxy)-3-pyridinyl]- l/f-indazole-4-carboxamide;
N-[(4,6-dimethyl-2-oxo- 1 ,2-dihydro-3-pyridinyl)methyl]- 1 -(l-methylethyl)-6-[2- (trifluoromethyl)phenyl] - lif-indazole-4-carboxamide;
6- {4-[(dimethylamino)sulfonyl]phenyl}-N-[(4,6-dimethyl-2-oxo- l,2-dihydro-3- pyridinyl)methyl]- 1 -(1 -methylethyl)- l/f-mdazole-4-carboxamide;
TV- [(4,6-dimethyl-2-oxo- 1 ,2-dihydro-3-pyridinyl)methyl]- 1 -(1 -methylethyl^-^- methylpheny^-l/i-indazole^-carboxamide;
6-bromo-Af-[(4-cyclohexyl-6-methyl-2-oxo-l,2-dihydro-3-pyridinyl)methyl]-l-(l- methylethyl)-lf -indazole-4-carboxamide;
6-bromo- l-(l-methy lethyl)-A^-[(6-methyl-2-oxo-4-propyl- l,2-dihydro-3-pyridinyl)methyl]- l f-indazole-4-carboxamide;
1 -(1 -methylethyl)-N- [(6-methyl-2-oxo-4-propyl- 1 ,2-dihydro-3 -pyridinyl)methyl] - 1H- indazole-4-carboxamide;
6-bromo- l-cyclopen1yl-N-[(6-methyl-2-oxo-4-propyl-l,2-dihydro-3-pyridinyl)methyl]-lH- indazole-4-carboxamide;
6-bromo-N- [(4-cyclopropyl-6-methyl-2-oxo- 1 ,2-dihydro-3-pyridinyl)methyl] - 1 -( 1 - methylethyl)- l//-indazole-4-carboxamide;
l-cyclopentyl-N-[(4,6-dimethyl-2-oxo-l,2-dihydro-3-pyridinyl)methyl]-6-[2-(4-methyl-l- piperazinyl)-4-pyridinyl]-li/-indazole-4-carboxamide;
6-bromo-l-cyclopentyl-N-[(4,6-dimethyl-2-oxo-l,2-dihydro-3-pyridinyl)methyl]-li/- indazole-4-carboxamide; 6-bromo- 1 -( 1 -methylethyl)-.V- { [6-methyl-4-(l -methylethyl)-2-oxo- 1 ,2-dihydro-3- pyridinyl]methyl} - l//-indazole-4-carboxamide;
l -cyclopentyl-N-[(4,6-dimethyl-2-oxo- l,2-dihydro-3-pyridinyl)methyl]- l 7-indazole-4- carboxamide;
6-bromo- 1 -( 1 -methylethyl)-iV- [(6-methyl-2-oxo-4-phenyl- 1 ,2-dihydro-3 -pyridinyl)methyl] - l /-indazole-4-carboxamide;
l -cyclopentyl-N-[(4,6-dimethyl-2-oxo- l,2-dihydro-3-pyridinyl)methyl]-6-(2-oxo-2,3- dihydro- l/f-benzimidazol-5-yl)- l//-indazole-4-carboxamide;
1 -(1 -methylethyl)-N- { [6-methyl-4-( 1 -methylethyl)-2-oxo- 1 ,2-dihydro-3- pyridmyl]methyl} 6-[2-(4-methyl-l
Figure imgf000177_0001
l -(l-methylethyl)-N-[(6-methyl-2-oxo-4-propyl-l,2-dihydro-3-pyridinyl)methyl]-6-(2-oxo- 2,3-dihydro-li/-benzimidazol-5-yl)- l /-indazole-4-carboxamide;
l -cyclopentyl-N-[(4,6-dimethyl-2-oxo- l,2-dihydro-3-pyridinyl)methyl]-6-(6-methyl-3- pyridmyl)- lif-indazole-4-carboxamide;
l -(l-methylethyl)-N-[(6-methyl-2-oxo-4-propyl-l,2-dihydro-3-pyridinyl)methyl]-6-[2-(4- methyl- l-piperazinyl)-4-pyridinyl]- lii-indazole-4-carboxarnide;
1 -cyclopentyl-JV-[(6-methyl-2-oxo-4-propyl- 1 ,2-dihydro-3-pyridinyl)methyl]-6-(6-methyl- 3-pyridinyl)- lii-indazole-4-carboxamide;
1 -cyclopentyl-JV-[(6-methyl-2-oxo-4-propyl- 1 ,2-dihydro-3-pyridinyl)methyl]-6-(3- pyridinyl)- l/f-indazole-4-carboxamide;
l -(l-methylethyl)-N-[(6-methyl-2-oxo-4-propyl-l,2-dihydro-3-pyridinyl)methyl]-6-(3- pyridinyl)- l/f-indazole-4-carboxamide;
l -cyclopentyl-^[(4,6-dimethyl-2-oxo-l,2-dihydro-3-pyridinyl)methyl]-6-[6-(4- morpholinyl)-3-pyridinyl]- lii-mdazole-4-carboxamide;
l -cyclopentyl-A -[(4,6-dimethyl-2-oxo- l,2-dihydro-3-pyridm^
l f-indazole-4-carboxamide; or a salt thereof.
10. A method of treating cancer comprising administering to a patient with cancer a therapeutically effective amount of a compound of formula (I) according to claim 1 or a pharmaceutically acceptable salt thereof, alone or admixed with a pharmaceutically acceptable carrier.
1 1. A pharmaceutically acceptable preparations comprising a compound of formula (I) according to claim 1 and pharmaceutically acceptable excipient.
12. A method of claim 9, wherein said cancer is selected from the group consisting of: brain (gliomas), glioblastomas, leukemias, lymphomas, Bannayan-Zonana syndrome, Cowden disease, Lhermitte-Duclos disease, breast, inflammatory breast cancer, Wilm's tumor, Ewing's sarcoma, Rhabdomyosarcoma, ependymoma, meduUoblastoma, colon, gastric, bladder, head and neck, kidney, lung, liver, melanoma, renal, ovarian, pancreatic, prostate, sarcoma, osteosarcoma, giant cell tumor of bone and thyroid.
13. The use of a compound of formula (I) according to claim 1 or a pharmaceutically acceptable salt or solvate thereof, in the preparation of a medicament for use in the treatment of a disorder mediated by inhibiting EZH2.
PCT/US2011/035340 2010-05-07 2011-05-05 Indazoles WO2011140325A1 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
ES11778335.7T ES2534804T3 (en) 2010-05-07 2011-05-05 Indazoles
US13/696,436 US8846935B2 (en) 2010-05-07 2011-05-05 Indazoles
JP2013509260A JP5864546B2 (en) 2010-05-07 2011-05-05 Indazole
EP11778335.7A EP2566328B1 (en) 2010-05-07 2011-05-05 Indazoles
US14/458,305 US9018382B2 (en) 2010-05-07 2014-08-13 Indazoles

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US33231210P 2010-05-07 2010-05-07
US61/332,312 2010-05-07

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US13/696,436 A-371-Of-International US8846935B2 (en) 2010-05-07 2011-05-05 Indazoles
US14/458,305 Division US9018382B2 (en) 2010-05-07 2014-08-13 Indazoles

Publications (1)

Publication Number Publication Date
WO2011140325A1 true WO2011140325A1 (en) 2011-11-10

Family

ID=44904073

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2011/035340 WO2011140325A1 (en) 2010-05-07 2011-05-05 Indazoles

Country Status (5)

Country Link
US (2) US8846935B2 (en)
EP (1) EP2566328B1 (en)
JP (1) JP5864546B2 (en)
ES (1) ES2534804T3 (en)
WO (1) WO2011140325A1 (en)

Cited By (66)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012118812A2 (en) 2011-02-28 2012-09-07 Epizyme, Inc. Substituted 6,5-fused bicyclic heteroaryl compounds
WO2012142504A1 (en) * 2011-04-13 2012-10-18 Epizyme, Inc. Aryl-or heteroaryl-substituted benzene compounds
EP2566327A1 (en) * 2010-05-07 2013-03-13 GlaxoSmithKline LLC Indoles
WO2013037960A1 (en) 2011-09-16 2013-03-21 Laboratorios Del Dr. Esteve, S.A. Ep1 receptor ligands
WO2013049770A2 (en) 2011-09-30 2013-04-04 Glaxosmithkline Llc Methods of treating cancer
WO2013075084A1 (en) 2011-11-18 2013-05-23 Constellation Pharmaceuticals Modulators of methyl modifying enzymes, compositions and uses thereof
WO2013075083A1 (en) 2011-11-18 2013-05-23 Constellation Pharmaceuticals Modulators of methyl modifying enzymes, compositions and uses thereof
WO2013138361A1 (en) 2012-03-12 2013-09-19 Epizyme, Inc. Inhibitors of human ezh2, and methods of use thereof
EP2646020A1 (en) * 2010-12-01 2013-10-09 GlaxoSmithKline LLC Indoles
WO2013155317A1 (en) * 2012-04-13 2013-10-17 Epizyme, Inc. Salt form of a human hi stone methyltransf erase ezh2 inhibitor
WO2013188848A2 (en) * 2012-06-15 2013-12-19 The Regents Of The University Of California Antiviral compounds and methods of use
US8691507B2 (en) 2010-09-10 2014-04-08 Epizyme, Inc. Inhibitors of human EZH2 and methods of use thereof
WO2014062720A2 (en) 2012-10-15 2014-04-24 Epizyme, Inc. Methods of treating cancer
WO2014100080A1 (en) * 2012-12-19 2014-06-26 Glaxosmithkline Llc Combination
WO2014100463A1 (en) * 2012-12-19 2014-06-26 Quanticel Pharmaceuticals, Inc. Histone demethylase inhibitors
WO2014100665A1 (en) * 2012-12-21 2014-06-26 Epizyme, Inc. 1,4-pyridone bicyclic heteroaryl compounds
WO2014153030A2 (en) 2013-03-14 2014-09-25 Genentech, Inc. Methods of treating cancer and preventing cancer drug resistance
JP2015506985A (en) * 2012-02-10 2015-03-05 コンステレーション・ファーマシューティカルズ・インコーポレイテッドConstellation Pharmaceuticals,Inc. Methyl group-modifying enzyme regulator, composition and use thereof
US9006242B2 (en) 2012-10-15 2015-04-14 Epizyme, Inc. Substituted benzene compounds
US9040515B2 (en) 2012-12-21 2015-05-26 Pfizer Inc. Aryl and heteroaryl fused lactams
WO2015077194A1 (en) * 2013-11-22 2015-05-28 Bristol-Myers Squibb Company Inhibitors of lysine methyl transferase
WO2015128837A1 (en) 2014-02-26 2015-09-03 Glaxosmithkline Intellectual Property (No.2) Limited Methods of treating cancer patients responding to ezh2 inhibitor gsk126
JP2015531366A (en) * 2012-09-28 2015-11-02 ファイザー・インク Benzamide and heterobenzamide compounds
US9175331B2 (en) 2010-09-10 2015-11-03 Epizyme, Inc. Inhibitors of human EZH2, and methods of use thereof
CN105017221A (en) * 2014-04-30 2015-11-04 中国医学科学院药物研究所 Benzimidazole derivative and preparation method, drug composition and application thereof
AU2012332297B2 (en) * 2011-11-04 2016-01-07 Glaxosmithkline Intellectual Property (No.2) Limited Method of treatment
CN105308038A (en) * 2013-04-30 2016-02-03 葛兰素史密斯克莱知识产权(第2号)有限公司 Enhancer of zeste homolog 2 inhibitors
WO2016097749A1 (en) * 2014-12-19 2016-06-23 Cancer Research Technology Limited Parg inhibitory compounds
US9376422B2 (en) 2011-04-13 2016-06-28 Epizyme, Inc. Dihidropyridin-2-one benzamine compounds
WO2016101956A2 (en) 2014-12-23 2016-06-30 University Of Copenhagen Treatment of cancer by inhibiting ezh2 activity
AU2014262280B2 (en) * 2011-04-13 2016-08-25 Epizyme, Inc. Aryl-or heteroaryl-substituted benzene compounds
US9481666B2 (en) 2014-06-17 2016-11-01 Pfizer Inc. Substituted dihydroisoquinolinone compounds
US9527837B2 (en) 2014-12-05 2016-12-27 Eli Lilly And Company Inhibitors of EZH2
US9718838B2 (en) 2015-08-27 2017-08-01 Eli Lilly And Company Inhibitors of EZH2
WO2017139455A1 (en) 2016-02-10 2017-08-17 Wake Forest University Health Sciences Model system of liver fibrosis and method of making and using the same
US9745305B2 (en) 2013-03-15 2017-08-29 Constellation Pharmaceuticals, Inc. Modulators of methyl modifying enzymes, compositions and uses thereof
WO2017157825A1 (en) 2016-03-15 2017-09-21 F. Hoffmann-La Roche Ag Combinations of lsd1 inhibitors for use in the treatment of solid tumors
CN107573327A (en) * 2016-07-05 2018-01-12 四川大学 Indazolecarboxamides Pyridione derivatives and its production and use
US9969716B2 (en) 2013-08-15 2018-05-15 Constellation Pharmaceuticals, Inc. Indole derivatives as modulators of methyl modifying enzymes, compositions and uses thereof
WO2018086592A1 (en) * 2016-11-11 2018-05-17 上海海雁医药科技有限公司 4,5,6-tri-substituted indazoles derivatives, preparation thereof, and use thereof in medicines
US10040782B2 (en) 2013-10-16 2018-08-07 Epizyme, Inc. Hydrochloride salt form for EZH2 inhibition
WO2018210302A1 (en) 2017-05-18 2018-11-22 江苏恒瑞医药股份有限公司 Crystal of benzofuran derivative free base and preparation method
US10239843B2 (en) 2014-12-12 2019-03-26 Cancer Research Technology Limited 2,4-dioxo-quinazoline-6-sulfonamide derivatives as inhibitors of PARG
WO2019057946A1 (en) 2017-09-25 2019-03-28 F. Hoffmann-La Roche Ag Multi-cyclic aromatic compounds as factor d inhibitors
US10266542B2 (en) 2017-03-15 2019-04-23 Mirati Therapeutics, Inc. EZH2 inhibitors
WO2019091450A1 (en) 2017-11-10 2019-05-16 江苏恒瑞医药股份有限公司 Method for preparing benzofuran derivative
WO2019097369A1 (en) 2017-11-14 2019-05-23 Pfizer Inc. Ezh2 inhibitor combination therapies
CN110016014A (en) * 2018-01-08 2019-07-16 中国科学院上海药物研究所 Application in EZH2 inhibitor and its preparation and antineoplaston
CN110229157A (en) * 2018-03-06 2019-09-13 上海海和药物研究开发有限公司 Pyrimido 5-membered aromatic heterocycle class compound, preparation method and the usage
US10457640B2 (en) 2016-10-19 2019-10-29 Constellation Pharmaceuticals, Inc. Synthesis of inhibitors of EZH2
WO2020011607A1 (en) 2018-07-09 2020-01-16 Fondation Asile Des Aveugles Inhibition of prc2 subunits to treat eye disorders
US10577350B2 (en) 2015-08-28 2020-03-03 Constellation Pharmaceuticals, Inc. Crystalline forms of (R)-N-((4-methoxy-6-methyl-2-oxo-1,2-dihydropyridin-3-yl)methyl)-2-methyl-1-(1-(1-(2,2,2-trifluoroethyl)piperidin-4-yl)ethyl)-1H-indole-3-carboxamide
CZ308400B6 (en) * 2018-04-11 2020-07-29 Univerzita Karlova Pharmaceutical preparation for treating malignant melanoma
US10759787B2 (en) 2015-11-19 2020-09-01 Jiangsu Hengrui Medicine Co., Ltd. Benzofuran derivative, preparation method thereof and use thereof in medicine
WO2021035194A1 (en) 2019-08-22 2021-02-25 Juno Therapeutics, Inc. Combination therapy of a t cell therapy and an enhancer of zeste homolog 2 (ezh2) inhibitor and related methods
US11059817B2 (en) 2015-09-23 2021-07-13 The Regents Of The University Of California Potent antiviral pyrazolopyridine compounds
US11091495B2 (en) 2018-01-31 2021-08-17 Mirati Therapeutics, Inc. Substituted imidazo[1,2-c]pyrimidines as PRC2 inhibitors
US11236082B2 (en) * 2014-11-06 2022-02-01 Dana-Farber Cancer Institute, Inc. EZH2 inhibitors and uses thereof
EP3831822A4 (en) * 2018-07-27 2022-03-23 Suzhou Sinovent Pharmaceuticals Co., Ltd. Polysubstituted benzene compound and preparation method and use thereof
US11326212B2 (en) 2010-06-23 2022-05-10 British Columbia Cancer Agency Branch Biomarkers for non-hodgkin lymphomas and uses thereof
RU2777624C2 (en) * 2017-11-10 2022-08-08 Цзянсу Хэнжуй Медисин Ко., Лтд. Method for production of benzofuran derivative
WO2023025856A1 (en) 2021-08-25 2023-03-02 INSERM (Institut National de la Santé et de la Recherche Médicale) Use of ezh2 inhibitors for the treatment of aortic valve stenosis
US11642349B2 (en) 2015-08-24 2023-05-09 Epizyme, Inc. Method for treating cancer
WO2023244917A1 (en) 2022-06-13 2023-12-21 Treeline Biosciences, Inc. 1,8-naphthyridin-2-one heterobifunctional bcl6 degraders
WO2023244918A1 (en) 2022-06-13 2023-12-21 Treeline Biosciences, Inc. Quinolone bcl6 bifunctional degraders
US11912994B2 (en) 2015-04-07 2024-02-27 The General Hospital Corporation Methods for reactivating genes on the inactive X chromosome

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013173441A2 (en) 2012-05-16 2013-11-21 Glaxosmithkline Llc Enhancer of zeste homolog 2 inhibitors
RU2016104044A (en) 2013-07-10 2017-08-15 Глэксосмитклайн Интеллекчуал Проперти (Nо.2) Лимитед ZESTE Homolog Amplifier Inhibitors 2
BR112017005510A2 (en) 2014-09-17 2018-08-14 Celgene Quanticel Res Inc histone demethylase inhibitors.
CA3000983A1 (en) * 2015-10-06 2017-04-13 The Regents Of The University Of Colorado, A Body Corporate Method of treating medulloblastoma with an ezh2 inhibitor
US10987353B2 (en) 2016-05-04 2021-04-27 The Wistar Institute Of Anatomy And Biology Methods of treating cancers overexpressing CARM1 with EZH2 inhibitors and platinum-based antineoplastic drugs
KR102351782B1 (en) * 2016-09-07 2022-01-17 상하이 하이헤 파마수티컬 컴퍼니 리미티드 Pyrido 5-membered aromatic ring compound, preparation method and use thereof

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000069826A1 (en) * 1999-05-19 2000-11-23 Pharmacia Corporation Substituted polycyclic aryl and heteroaryl pyridones useful for selective inhibition of the coagulation cascade
WO2004112719A2 (en) 2003-06-19 2004-12-29 Smithkline Beecham Corporation Chemical compounds
US20090012031A1 (en) * 2007-07-03 2009-01-08 The Regents Of The University Of Michigan EZH2 Cancer Markers
WO2009012312A1 (en) * 2007-07-16 2009-01-22 Abbott Laboratories Indazoles, benzisoxazoles and benzisothiazoles as inhibitors of protein kinases
WO2010036213A1 (en) * 2008-09-26 2010-04-01 Agency For Science, Technology And Research 3-deazaneplanocin derivatives
US7709500B2 (en) * 2002-02-18 2010-05-04 Astrazeneca Ab Chemical compounds

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2722923C (en) * 2008-04-29 2016-08-02 Boehringer Ingelheim International Gmbh Indazole compounds as ccr1 receptor antagonists
HUE034787T2 (en) 2010-05-07 2018-02-28 Glaxosmithkline Llc Indoles
US8637509B2 (en) 2010-05-07 2014-01-28 Glaxosmithkline Llc Azaindazoles
WO2012118812A2 (en) 2011-02-28 2012-09-07 Epizyme, Inc. Substituted 6,5-fused bicyclic heteroaryl compounds

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000069826A1 (en) * 1999-05-19 2000-11-23 Pharmacia Corporation Substituted polycyclic aryl and heteroaryl pyridones useful for selective inhibition of the coagulation cascade
US7709500B2 (en) * 2002-02-18 2010-05-04 Astrazeneca Ab Chemical compounds
WO2004112719A2 (en) 2003-06-19 2004-12-29 Smithkline Beecham Corporation Chemical compounds
US20090012031A1 (en) * 2007-07-03 2009-01-08 The Regents Of The University Of Michigan EZH2 Cancer Markers
WO2009012312A1 (en) * 2007-07-16 2009-01-22 Abbott Laboratories Indazoles, benzisoxazoles and benzisothiazoles as inhibitors of protein kinases
WO2010036213A1 (en) * 2008-09-26 2010-04-01 Agency For Science, Technology And Research 3-deazaneplanocin derivatives

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP2566328A4

Cited By (178)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9649307B2 (en) 2010-05-07 2017-05-16 Glaxosmithkline Llc Indoles
EP2566327A1 (en) * 2010-05-07 2013-03-13 GlaxoSmithKline LLC Indoles
US9402836B2 (en) 2010-05-07 2016-08-02 Glaxosmithkline Llc Indoles
US8975291B2 (en) 2010-05-07 2015-03-10 Glaxosmithkline Llc Indoles
EP2566327A4 (en) * 2010-05-07 2013-12-11 Glaxosmithkline Llc Indoles
US9114141B2 (en) 2010-05-07 2015-08-25 Glaxosmithkline Llc Indoles
EP3246027A1 (en) * 2010-05-07 2017-11-22 GlaxoSmithKline LLC Indole derivatives for the treatment of cancer
US11326212B2 (en) 2010-06-23 2022-05-10 British Columbia Cancer Agency Branch Biomarkers for non-hodgkin lymphomas and uses thereof
US8895245B2 (en) 2010-09-10 2014-11-25 Epizyme, Inc. Inhibitors of human EZH2 and methods of use thereof
US9175331B2 (en) 2010-09-10 2015-11-03 Epizyme, Inc. Inhibitors of human EZH2, and methods of use thereof
US9334527B2 (en) 2010-09-10 2016-05-10 Epizyme, Inc. Inhibitors of human EZH2, and methods of use thereof
US8691507B2 (en) 2010-09-10 2014-04-08 Epizyme, Inc. Inhibitors of human EZH2 and methods of use thereof
US9949999B2 (en) 2010-09-10 2018-04-24 Epizyme, Inc. Inhibitors of human EZH2, and methods of use thereof
US9333217B2 (en) 2010-09-10 2016-05-10 Epizyme, Inc. Inhibitors of human EZH2, and methods of use thereof
EP2646020A4 (en) * 2010-12-01 2014-04-23 Glaxosmithkline Llc Indoles
JP2013544840A (en) * 2010-12-01 2013-12-19 グラクソスミスクライン・リミテッド・ライアビリティ・カンパニー Indole
EP2646020A1 (en) * 2010-12-01 2013-10-09 GlaxoSmithKline LLC Indoles
EP2681216A2 (en) * 2011-02-28 2014-01-08 Epizyme, Inc. Substituted 6,5-fused bicyclic heteroaryl compounds
US8598167B1 (en) 2011-02-28 2013-12-03 Epizyme, Inc. Substituted 6,5-fused bicyclic heteroaryl compounds
EP2681216A4 (en) * 2011-02-28 2014-10-01 Epizyme Inc Substituted 6,5-fused bicyclic heteroaryl compounds
US9637472B2 (en) 2011-02-28 2017-05-02 Epizyme, Inc. Substituted 6,5-fused bicyclic heteroaryl compounds
US9206157B2 (en) 2011-02-28 2015-12-08 Epizyme, Inc. Substituted 6,5-fused bicyclic heteroaryl compounds
WO2012118812A2 (en) 2011-02-28 2012-09-07 Epizyme, Inc. Substituted 6,5-fused bicyclic heteroaryl compounds
EP3323820A1 (en) * 2011-02-28 2018-05-23 Epizyme, Inc. Substituted 6,5-fused bicyclic heteroaryl compounds
US8962620B2 (en) 2011-02-28 2015-02-24 Epizyme, Inc. Substituted 6,5-fused bicyclic heteroaryl compounds
US10273223B2 (en) 2011-02-28 2019-04-30 Epizyme, Inc. Substituted 6,5-fused bicyclic heteroaryl compounds
US9855275B2 (en) 2011-04-13 2018-01-02 Epizyme, Inc. Aryl-or heteroaryl-substituted benzene compounds
US9549931B2 (en) 2011-04-13 2017-01-24 Epizyme, Inc. Aryl- or heteroaryl-substituted benzene compounds
CN104080769A (en) * 2011-04-13 2014-10-01 Epizyme股份有限公司 Aryl- or heteroaryl-substituted benzene compounds
EP3150580A1 (en) * 2011-04-13 2017-04-05 Epizyme, Inc. Aryl- or heteroaryl-substituted benzene compounds
JP2014516931A (en) * 2011-04-13 2014-07-17 エピザイム,インコーポレイティド Aryl-substituted or heteroaryl-substituted benzene compounds
AU2021202894B2 (en) * 2011-04-13 2023-07-06 Epizyme, Inc. Aryl-or heteroaryl-substituted benzene compounds
US9522152B2 (en) 2011-04-13 2016-12-20 Epizyme, Inc. Aryl- or heteroaryl-substituted benzene compounds
US8765732B2 (en) 2011-04-13 2014-07-01 Epizyme, Inc. Aryl- or heteroaryl-substituted benzene compounds
WO2012142504A1 (en) * 2011-04-13 2012-10-18 Epizyme, Inc. Aryl-or heteroaryl-substituted benzene compounds
EP3943485A1 (en) * 2011-04-13 2022-01-26 Epizyme, Inc. Aryl- or heteroaryl-substituted benzene compounds
US11052093B2 (en) 2011-04-13 2021-07-06 Epizyme, Inc. Aryl-or heteroaryl-substituted benzene compounds
JP2021042243A (en) * 2011-04-13 2021-03-18 エピザイム,インコーポレイティド Aryl- or heteroaryl-substituted benzene compounds
CN107311921B (en) * 2011-04-13 2021-02-12 Epizyme股份有限公司 Aryl or heteroaryl substituted benzene compounds
CN104080769B (en) * 2011-04-13 2017-06-20 Epizyme股份有限公司 Aryl or heteroaryl-substituted compound
AU2014262280B2 (en) * 2011-04-13 2016-08-25 Epizyme, Inc. Aryl-or heteroaryl-substituted benzene compounds
CN107311921A (en) * 2011-04-13 2017-11-03 Epizyme股份有限公司 Aryl or heteroaryl-substituted compound
US9376422B2 (en) 2011-04-13 2016-06-28 Epizyme, Inc. Dihidropyridin-2-one benzamine compounds
US9090562B2 (en) 2011-04-13 2015-07-28 Epizyme, Inc. Aryl- or heteroaryl-substituted benzene compounds
US10420775B2 (en) 2011-04-13 2019-09-24 Epizyme, Inc. Aryl-or heteroaryl-substituted benzene compounds
AU2018200826B2 (en) * 2011-04-13 2019-09-19 Epizyme, Inc. Aryl-or heteroaryl-substituted benzene compounds
US8410088B2 (en) 2011-04-13 2013-04-02 Epizyme, Inc. Aryl- or heteroaryl-substituted benzene compounds
US10155002B2 (en) 2011-04-13 2018-12-18 Epizyme, Inc. Aryl- or heteroaryl-substituted benzene compounds
EP3486234A1 (en) * 2011-04-13 2019-05-22 Epizyme Inc Aryl- or heteroaryl-substituted benzene compounds
WO2013037960A1 (en) 2011-09-16 2013-03-21 Laboratorios Del Dr. Esteve, S.A. Ep1 receptor ligands
WO2013049770A2 (en) 2011-09-30 2013-04-04 Glaxosmithkline Llc Methods of treating cancer
CN103987842A (en) * 2011-09-30 2014-08-13 葛兰素史密斯克莱有限责任公司 Methods of treating cancer
US9730925B2 (en) 2011-09-30 2017-08-15 Glaxomithkline Llc Methods of treating cancer
AU2012332297B2 (en) * 2011-11-04 2016-01-07 Glaxosmithkline Intellectual Property (No.2) Limited Method of treatment
US9242962B2 (en) 2011-11-04 2016-01-26 Glaxosmithkline Intellectual Property (No. 2) Limited Method of treatment
US9446041B2 (en) 2011-11-04 2016-09-20 Glaxosmithkline Intellectual Property (No. 2) Limited Method of treatment
WO2013075084A1 (en) 2011-11-18 2013-05-23 Constellation Pharmaceuticals Modulators of methyl modifying enzymes, compositions and uses thereof
US9409865B2 (en) 2011-11-18 2016-08-09 Constellation_Pharmaceuticals, Inc. Modulators of methyl modifying enzymes, compositions and uses thereof
US9206128B2 (en) 2011-11-18 2015-12-08 Constellation Pharmaceuticals, Inc. Modulators of methyl modifying enzymes, compositions and uses thereof
US9051269B2 (en) 2011-11-18 2015-06-09 Constellation Pharmaceuticals, Inc. Modulators of methyl modifying enzymes, compositions and uses thereof
EP2780014A4 (en) * 2011-11-18 2015-07-01 Constellation Pharmaceuticals Inc Modulators of methyl modifying enzymes, compositions and uses thereof
WO2013075083A1 (en) 2011-11-18 2013-05-23 Constellation Pharmaceuticals Modulators of methyl modifying enzymes, compositions and uses thereof
EP2780013A4 (en) * 2011-11-18 2015-07-01 Constellation Pharmaceuticals Inc Modulators of methyl modifying enzymes, compositions and uses thereof
USRE47428E1 (en) 2012-02-10 2019-06-11 Constellation Pharmaceuticals, Inc. Modulators of methyl modifying enzymes, compositions and uses thereof
US9371331B2 (en) 2012-02-10 2016-06-21 Constellation Pharmaceuticals, Inc. Modulators of methyl modifying enzymes, compositions and uses thereof
US9980952B2 (en) 2012-02-10 2018-05-29 Constellation Pharmaceuticals, Inc. Modulators of methyl modifying enzymes, compositions and uses thereof
US10016405B2 (en) 2012-02-10 2018-07-10 Constellation Pharmaceuticals, Inc. Modulators of methyl modifying enzymes, compositions and uses thereof
US9469646B2 (en) 2012-02-10 2016-10-18 Constellation Pharmaceuticals, Inc. Modulators of methyl modifying enzymes, compositions and uses thereof
US9085583B2 (en) 2012-02-10 2015-07-21 Constellation—Pharmaceuticals, Inc. Modulators of methyl modifying enzymes, compositions and uses thereof
JP2015506985A (en) * 2012-02-10 2015-03-05 コンステレーション・ファーマシューティカルズ・インコーポレイテッドConstellation Pharmaceuticals,Inc. Methyl group-modifying enzyme regulator, composition and use thereof
WO2013138361A1 (en) 2012-03-12 2013-09-19 Epizyme, Inc. Inhibitors of human ezh2, and methods of use thereof
EP3536314A1 (en) 2012-03-12 2019-09-11 Epizyme, Inc. Inhibitors of human ezh2, and methods of use thereof
RU2658911C2 (en) * 2012-04-13 2018-06-26 Эпизайм, Инк. Salt form of human histone methyltranserase ezh2 inhibitor
CN104603130B (en) * 2012-04-13 2018-04-27 Epizyme股份有限公司 The salt form of mankind's histone methyltransferase EZH2 inhibitor
US9394283B2 (en) 2012-04-13 2016-07-19 Epizyme, Inc. Salt form of a human histone methyltransferase EZH2 inhibitor
AU2018200168B2 (en) * 2012-04-13 2019-08-29 Eisai R&D Management Co.Ltd. Salt form of a human histone methyltransferase ezh2 inhibitor
WO2013155317A1 (en) * 2012-04-13 2013-10-17 Epizyme, Inc. Salt form of a human hi stone methyltransf erase ezh2 inhibitor
US10245269B2 (en) 2012-04-13 2019-04-02 Epizyme, Inc. Salt form of a human histone methyltransferase EZH2 inhibitor
CN108358899A (en) * 2012-04-13 2018-08-03 Epizyme股份有限公司 The salt form of mankind's histone methyltransferase EZH2 inhibitor
US9872862B2 (en) 2012-04-13 2018-01-23 Epizyme, Inc. Salt form of a human histone methyltransferase EZH2 inhibitor
US11491163B2 (en) 2012-04-13 2022-11-08 Epizyme, Inc. Salt form of a human histone methyltransferase EZH2 inhibitor
JP2018002742A (en) * 2012-04-13 2018-01-11 エピザイム,インコーポレイティド Salt form of human histone methyltransferase ezh2 inhibitor
JP2015512942A (en) * 2012-04-13 2015-04-30 エピザイム,インコーポレイティド Salt form of human histone methyltransferase EZH2 inhibitor
US10821113B2 (en) 2012-04-13 2020-11-03 Epizyme, Inc. Salt form of a human histone methyltransferase EZH2 inhibitor
CN108358899B (en) * 2012-04-13 2021-07-27 Epizyme股份有限公司 Salt forms of human histone methyltransferase EZH2 inhibitor
CN104603130A (en) * 2012-04-13 2015-05-06 Epizyme股份有限公司 Salt form of a human hi stone methyltransf erase EZH2 inhibitor
WO2013188848A2 (en) * 2012-06-15 2013-12-19 The Regents Of The University Of California Antiviral compounds and methods of use
WO2013188848A3 (en) * 2012-06-15 2014-02-20 The Regents Of The University Of California Antiviral compounds and methods of use
JP2015531366A (en) * 2012-09-28 2015-11-02 ファイザー・インク Benzamide and heterobenzamide compounds
US10098888B2 (en) 2012-10-15 2018-10-16 Epizyme, Inc. Substituted benzene compounds
US9089575B2 (en) 2012-10-15 2015-07-28 Epizyme, Inc. Substituted benzene compounds
US9532992B2 (en) 2012-10-15 2017-01-03 Epizyme, Inc. Substituted benzene compounds
US11642348B2 (en) 2012-10-15 2023-05-09 Epizyme, Inc. Substituted benzene compounds
US10092572B2 (en) 2012-10-15 2018-10-09 Epizyme, Inc. Substituted benzene compounds
US9006242B2 (en) 2012-10-15 2015-04-14 Epizyme, Inc. Substituted benzene compounds
EP3659606A1 (en) 2012-10-15 2020-06-03 Epizyme, Inc. Methods of treating cancer
WO2014062720A2 (en) 2012-10-15 2014-04-24 Epizyme, Inc. Methods of treating cancer
US9617242B2 (en) 2012-12-19 2017-04-11 Celgene Quanticel Research, Inc. Histone demethylase inhibitors
WO2014100080A1 (en) * 2012-12-19 2014-06-26 Glaxosmithkline Llc Combination
AU2013361255B2 (en) * 2012-12-19 2017-01-05 Celgene Quanticel Research, Inc. Histone demethylase inhibitors
US8969343B2 (en) 2012-12-19 2015-03-03 Quanticel Pharmaceuticals, Inc. Histone demethylase inhibitors
WO2014100463A1 (en) * 2012-12-19 2014-06-26 Quanticel Pharmaceuticals, Inc. Histone demethylase inhibitors
US10336727B2 (en) 2012-12-19 2019-07-02 Celgene Quanticel Research, Inc. Histone demethylase inhibitors
US10040779B2 (en) 2012-12-19 2018-08-07 Celgene Quanticel Research, Inc. Histone demethylase inhibitors
WO2014100665A1 (en) * 2012-12-21 2014-06-26 Epizyme, Inc. 1,4-pyridone bicyclic heteroaryl compounds
US9040515B2 (en) 2012-12-21 2015-05-26 Pfizer Inc. Aryl and heteroaryl fused lactams
US10246433B2 (en) 2012-12-21 2019-04-02 Pfizer Inc. Aryl and heteroaryl fused lactams
US10150759B2 (en) 2012-12-21 2018-12-11 Epizyme, Inc. 1,4-pyridone bicycic heteroaryl compounds
US9701666B2 (en) 2012-12-21 2017-07-11 Epizyme, Inc. 1,4-pyridone bicyclic heteroaryl compounds
WO2014153030A2 (en) 2013-03-14 2014-09-25 Genentech, Inc. Methods of treating cancer and preventing cancer drug resistance
US9745305B2 (en) 2013-03-15 2017-08-29 Constellation Pharmaceuticals, Inc. Modulators of methyl modifying enzymes, compositions and uses thereof
CN105308038B (en) * 2013-04-30 2018-05-29 葛兰素史密斯克莱知识产权(第2 号)有限公司 The inhibitor of Zeste enhancers homologue 2
CN105308038A (en) * 2013-04-30 2016-02-03 葛兰素史密斯克莱知识产权(第2号)有限公司 Enhancer of zeste homolog 2 inhibitors
US9969716B2 (en) 2013-08-15 2018-05-15 Constellation Pharmaceuticals, Inc. Indole derivatives as modulators of methyl modifying enzymes, compositions and uses thereof
US10710987B2 (en) 2013-10-16 2020-07-14 Epizyme, Inc. Hydrochloride salt form for EZH2 inhibition
US10040782B2 (en) 2013-10-16 2018-08-07 Epizyme, Inc. Hydrochloride salt form for EZH2 inhibition
US9822103B2 (en) 2013-11-22 2017-11-21 Bristol-Myers Squibb Company Inhibitors of lysine methyl transferase
WO2015077194A1 (en) * 2013-11-22 2015-05-28 Bristol-Myers Squibb Company Inhibitors of lysine methyl transferase
WO2015128837A1 (en) 2014-02-26 2015-09-03 Glaxosmithkline Intellectual Property (No.2) Limited Methods of treating cancer patients responding to ezh2 inhibitor gsk126
CN105017221B (en) * 2014-04-30 2019-05-28 中国医学科学院药物研究所 Benzimidizole derivatives and its preparation method and pharmaceutical composition and purposes
CN105017221A (en) * 2014-04-30 2015-11-04 中国医学科学院药物研究所 Benzimidazole derivative and preparation method, drug composition and application thereof
US9481666B2 (en) 2014-06-17 2016-11-01 Pfizer Inc. Substituted dihydroisoquinolinone compounds
US10570121B2 (en) 2014-06-17 2020-02-25 Pfizer Inc. Substituted dihydroisoquinolinone compounds
US11236082B2 (en) * 2014-11-06 2022-02-01 Dana-Farber Cancer Institute, Inc. EZH2 inhibitors and uses thereof
US9527837B2 (en) 2014-12-05 2016-12-27 Eli Lilly And Company Inhibitors of EZH2
US10239843B2 (en) 2014-12-12 2019-03-26 Cancer Research Technology Limited 2,4-dioxo-quinazoline-6-sulfonamide derivatives as inhibitors of PARG
CN107295799A (en) * 2014-12-19 2017-10-24 癌症研究科技有限公司 Parg inhibiting compounds
US10995073B2 (en) 2014-12-19 2021-05-04 Cancer Research Technology Limited PARG inhibitory compounds
US10508086B2 (en) 2014-12-19 2019-12-17 Cancer Research Technology Limited PARG inhibitory compounds
CN107295799B (en) * 2014-12-19 2021-03-16 癌症研究科技有限公司 PARG inhibiting compounds
WO2016097749A1 (en) * 2014-12-19 2016-06-23 Cancer Research Technology Limited Parg inhibitory compounds
AU2015365602B2 (en) * 2014-12-19 2020-02-27 Cancer Research Technology Limited PARG inhibitory compounds
EP3907224A1 (en) * 2014-12-19 2021-11-10 Cancer Research Technology Limited Parg inhibitory compounds
WO2016101956A3 (en) * 2014-12-23 2016-09-15 University Of Copenhagen Treatment of cancer by inhibiting ezh2 activity
WO2016101956A2 (en) 2014-12-23 2016-06-30 University Of Copenhagen Treatment of cancer by inhibiting ezh2 activity
US11912994B2 (en) 2015-04-07 2024-02-27 The General Hospital Corporation Methods for reactivating genes on the inactive X chromosome
US11642349B2 (en) 2015-08-24 2023-05-09 Epizyme, Inc. Method for treating cancer
US9718838B2 (en) 2015-08-27 2017-08-01 Eli Lilly And Company Inhibitors of EZH2
US10577350B2 (en) 2015-08-28 2020-03-03 Constellation Pharmaceuticals, Inc. Crystalline forms of (R)-N-((4-methoxy-6-methyl-2-oxo-1,2-dihydropyridin-3-yl)methyl)-2-methyl-1-(1-(1-(2,2,2-trifluoroethyl)piperidin-4-yl)ethyl)-1H-indole-3-carboxamide
US11059817B2 (en) 2015-09-23 2021-07-13 The Regents Of The University Of California Potent antiviral pyrazolopyridine compounds
US11787799B2 (en) 2015-09-23 2023-10-17 The Regents Of The University Of California Potent antiviral pyrazolopyridine compounds
US10759787B2 (en) 2015-11-19 2020-09-01 Jiangsu Hengrui Medicine Co., Ltd. Benzofuran derivative, preparation method thereof and use thereof in medicine
US11059811B2 (en) 2015-11-19 2021-07-13 Jiangsu Hengrui Medicine Co., Ltd. Benzofuran derivative, preparation method thereof and use thereof in medicine
WO2017139455A1 (en) 2016-02-10 2017-08-17 Wake Forest University Health Sciences Model system of liver fibrosis and method of making and using the same
US10265279B2 (en) 2016-03-15 2019-04-23 Oryzon Genomics, S.A. Combinations of LSD1 inhibitors for use in the treatment of solid tumors
WO2017157825A1 (en) 2016-03-15 2017-09-21 F. Hoffmann-La Roche Ag Combinations of lsd1 inhibitors for use in the treatment of solid tumors
CN107573327B (en) * 2016-07-05 2020-03-31 四川大学 Indazole-formamide-pyridone derivative and preparation method and application thereof
CN107573327A (en) * 2016-07-05 2018-01-12 四川大学 Indazolecarboxamides Pyridione derivatives and its production and use
US10457640B2 (en) 2016-10-19 2019-10-29 Constellation Pharmaceuticals, Inc. Synthesis of inhibitors of EZH2
WO2018086592A1 (en) * 2016-11-11 2018-05-17 上海海雁医药科技有限公司 4,5,6-tri-substituted indazoles derivatives, preparation thereof, and use thereof in medicines
CN108699029B (en) * 2016-11-11 2021-07-30 上海海雁医药科技有限公司 4,5, 6-trisubstituted indazole derivatives, preparation method and medical application thereof
US11104664B2 (en) 2016-11-11 2021-08-31 Shanghai Haiyan Pharmaceutical Technology Co., Ltd. 4,5,6-trisubstituted indazole derivatives, and preparation method and pharmaceutical use thereof
CN108699029A (en) * 2016-11-11 2018-10-23 上海海雁医药科技有限公司 Tri- substituted indazole analog derivatives of 4,5,6-, its preparation method and purposes pharmaceutically
US10266542B2 (en) 2017-03-15 2019-04-23 Mirati Therapeutics, Inc. EZH2 inhibitors
WO2018210302A1 (en) 2017-05-18 2018-11-22 江苏恒瑞医药股份有限公司 Crystal of benzofuran derivative free base and preparation method
US11155537B2 (en) 2017-05-18 2021-10-26 Jiangsu Hengrui Medicine Co., Ltd. Crystal of benzofuran derivative free base and preparation method
WO2019057946A1 (en) 2017-09-25 2019-03-28 F. Hoffmann-La Roche Ag Multi-cyclic aromatic compounds as factor d inhibitors
CN111094279B (en) * 2017-11-10 2023-06-16 江苏恒瑞医药股份有限公司 Preparation method of benzofuran derivative
CN111094279A (en) * 2017-11-10 2020-05-01 江苏恒瑞医药股份有限公司 Preparation method of benzofuran derivative
WO2019091450A1 (en) 2017-11-10 2019-05-16 江苏恒瑞医药股份有限公司 Method for preparing benzofuran derivative
US11390614B2 (en) 2017-11-10 2022-07-19 Jiangsu Hengrui Medicine Co., Ltd. Method for preparing benzofuran derivative
RU2777624C2 (en) * 2017-11-10 2022-08-08 Цзянсу Хэнжуй Медисин Ко., Лтд. Method for production of benzofuran derivative
WO2019097369A1 (en) 2017-11-14 2019-05-23 Pfizer Inc. Ezh2 inhibitor combination therapies
US11529344B2 (en) 2017-11-14 2022-12-20 Pfizer Inc. EZH2 inhibitor combination therapies
CN110016014A (en) * 2018-01-08 2019-07-16 中国科学院上海药物研究所 Application in EZH2 inhibitor and its preparation and antineoplaston
CN110016014B (en) * 2018-01-08 2023-02-17 中国科学院上海药物研究所 EZH2 inhibitor, preparation thereof and application thereof in antitumor therapy
US11220509B2 (en) 2018-01-31 2022-01-11 Mirati Therapeutics, Inc. Substituted imidazo[1,2-c]pyrimidines as PRC2 inhibitors
US11485738B2 (en) 2018-01-31 2022-11-01 Mirati Therapeutics, Inc. Substituted imidazo[1,2-c]pyrimidines as PRC2 inhibitors
US11091495B2 (en) 2018-01-31 2021-08-17 Mirati Therapeutics, Inc. Substituted imidazo[1,2-c]pyrimidines as PRC2 inhibitors
CN110229157B (en) * 2018-03-06 2022-06-21 上海海和药物研究开发股份有限公司 Pyrimido five-membered heteroaromatic compound, preparation method and application thereof
CN110229157A (en) * 2018-03-06 2019-09-13 上海海和药物研究开发有限公司 Pyrimido 5-membered aromatic heterocycle class compound, preparation method and the usage
CZ308400B6 (en) * 2018-04-11 2020-07-29 Univerzita Karlova Pharmaceutical preparation for treating malignant melanoma
WO2020011607A1 (en) 2018-07-09 2020-01-16 Fondation Asile Des Aveugles Inhibition of prc2 subunits to treat eye disorders
EP3831822A4 (en) * 2018-07-27 2022-03-23 Suzhou Sinovent Pharmaceuticals Co., Ltd. Polysubstituted benzene compound and preparation method and use thereof
WO2021035194A1 (en) 2019-08-22 2021-02-25 Juno Therapeutics, Inc. Combination therapy of a t cell therapy and an enhancer of zeste homolog 2 (ezh2) inhibitor and related methods
WO2023025856A1 (en) 2021-08-25 2023-03-02 INSERM (Institut National de la Santé et de la Recherche Médicale) Use of ezh2 inhibitors for the treatment of aortic valve stenosis
WO2023244917A1 (en) 2022-06-13 2023-12-21 Treeline Biosciences, Inc. 1,8-naphthyridin-2-one heterobifunctional bcl6 degraders
WO2023244918A1 (en) 2022-06-13 2023-12-21 Treeline Biosciences, Inc. Quinolone bcl6 bifunctional degraders

Also Published As

Publication number Publication date
EP2566328B1 (en) 2015-03-04
US8846935B2 (en) 2014-09-30
JP5864546B2 (en) 2016-02-17
US20130053383A1 (en) 2013-02-28
EP2566328A4 (en) 2013-10-16
US9018382B2 (en) 2015-04-28
ES2534804T3 (en) 2015-04-28
JP2013528591A (en) 2013-07-11
EP2566328A1 (en) 2013-03-13
US20140350013A1 (en) 2014-11-27

Similar Documents

Publication Publication Date Title
EP2566328B1 (en) Indazoles
US9649307B2 (en) Indoles
EP2646020B1 (en) Indoles
EP2755962B1 (en) Azaindazoles
EP2566479B1 (en) Azaindazoles

Legal Events

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

Ref document number: 11778335

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2013509260

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 13696436

Country of ref document: US

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2011778335

Country of ref document: EP