WO2013067302A1 - Method of treatment - Google Patents

Method of treatment Download PDF

Info

Publication number
WO2013067302A1
WO2013067302A1 PCT/US2012/063243 US2012063243W WO2013067302A1 WO 2013067302 A1 WO2013067302 A1 WO 2013067302A1 US 2012063243 W US2012063243 W US 2012063243W WO 2013067302 A1 WO2013067302 A1 WO 2013067302A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
methyl
pyridinyl
mmol
cycloalkyl
Prior art date
Application number
PCT/US2012/063243
Other languages
French (fr)
Inventor
Anna K. BASSIL
Soren BEINKE
Rabinder Kumar Prinjha
Original Assignee
Glaxosmithkline Intellectual Property (No. 2) Limited
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 Intellectual Property (No. 2) Limited filed Critical Glaxosmithkline Intellectual Property (No. 2) Limited
Publication of WO2013067302A1 publication Critical patent/WO2013067302A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/444Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring heteroatom, e.g. amrinone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/4545Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring hetero atom, e.g. pipamperone, anabasine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/513Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim having oxo groups directly attached to the heterocyclic ring, e.g. cytosine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/1003Transferases (2.) transferring one-carbon groups (2.1)
    • C12N9/1007Methyltransferases (general) (2.1.1.)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y201/00Transferases transferring one-carbon groups (2.1)
    • C12Y201/01Methyltransferases (2.1.1)
    • C12Y201/01043Histone-lysine N-methyltransferase (2.1.1.43)
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • This invention relates to compounds which inhibit EZH2/EZH1 and their uses for treating T cell mediated inflammatory immune diseases.
  • Posttranslational modifications of proteins play a critical role in the regulation of signal transduction from receptors, chromatin remodelling and gene transcription. These modifications include acetylation, methylation, phosphorylation, ubiquitinylation, SUMOylation.
  • EZH (enhancer of zeste homolog) 1 and 2 are the catalytic subunits of the Polycomb Repressor Complex 2 (PRC2) and exhibit methyltransferase activity that can catalyse the methylation of lysine amino acids (Margueron R, Reinberg D:The Polycomb complex PRC2 and its mark in life. Nature. 201 1 Jan 20;469 (7330):343-9)
  • Histone H3 is one of the five main histone proteins involved in the structure of chromatin in eukaryotic cells. Chromatin is the complex combination of DNA and protein that makes up chromosomes. It is found inside the nuclei of eukaryotic cells and is divided between heterochromatin (condensed) and euchromatin (extended).
  • the basic building blocks of chromatin are nucleosomes, each of which is composed of 146 base pairs of DNA wrapped around a histone octamer that consists of 2 copies of each H2A, H2B, H3 and H4.
  • the functions of chromatin are to package DNA into a smaller volume to fit in the cell, to strengthen the DNA to allow mitosis and meiosis, and to serve as a mechanism to control gene expression and DNA replication.
  • the chromatin structure is controlled by a series of post translational modifications to histone proteins, notably histones H3 and H4, and most commonly within the "histone tails" which extend beyond the core nucleosome structure.
  • H3K27me3 Binding of enzymes and adaptor proteins to posttranslational modification in histone tails regulates chromatin dynamics and gene expression.
  • H3K27me3 is thought to silence gene expression by recruiting histone deacetylases to the modified nucleosomes and stall transcriptional elongation by polymerase II.
  • inhibition of the enzymatic activity of EZH1 and EZH2 may result in a loss of H3K27me3 and up-regulation of target genes.
  • EZH2 has been implicated in the regulation of signal transduction that leads to actin polymerization in the cytoplasm of cells (Su IH, Dobenecker MW, Dickinson E, Oser M, Basavaraj A, Marqueron R, Viale A, Reinberg D, WQIfing C, Tarakhovsky A: Polycomb group protein ezh2 controls actin polymerization and cell signaling. Cell. 2005 May 6; 121 (3):425-36).
  • the reorganization of the actin cytoskeleton critically contributes to T cell responses by facilitating the interaction of T cells with antigen presenting cells or target cells.
  • actin remodelling plays an important role in T cell migration and motility during their recruitment to the sites of inflammation.
  • a fraction of EZH2 protein was found to localize to the cytoplasm of T cells and to interact with the small GTPase VAV1 , which is involved in actin remodelling.
  • Genetic elimination of EZH2 resulted in impaired polymerization of actin in TCR stimulated T cells or at the T cell - antigen presenting cell interphase.
  • actin polymerization induced by EZH2 over-expression was dependent on the methytransferase activity of EZH2. Proliferation of T cells in response to TCR was also impaired in the absence of EZH2.
  • inhibition of EZH1 and / or EZH2 may suppress the activation of T cells.
  • Mature T cell respond to foreign peptide antigens in the presence of appropriate co-stimulation by antigen presenting cells. They have the capability to discriminate between self and non self as a consequence of the selection of a TCR repertoire specific for foreign antigens in the thymus, tolerance induction of self reactive T cell clones in the periphery, and control of T cell activation by self antigen by regulatory T cells.
  • T cells provide protection against different classes of pathogens by mediating distinct types of adaptive immune responses as a consequence of the expression of distinct sets of cytokines and other soluble and cell-bound products. In addition, they act as principle amplifiers and inducers of the appropriate inflammatory and effector responses in cells of the innate immune system and nonimmune cells.
  • CD8 T cells can lyse cells bearing intracellular pathogens but may also contribute to tissue damage and secrete proinflammatory cytokines, e.g. TNF and IFNg.
  • CD4 T cells can have diverse functions in inflammation depending on their specific cytokine expression profiles.
  • CD4 + Th1 cells are important for the clearance of intracellular pathogens but also play a critical role in inflammation through the expression of TNF and IFNg.
  • IL-17 expressing CD4 + Th17 cells which mediate neutrophilia and tissue remodelling and repair, have also been shown to be involved in many inflammatory conditions.
  • CD4 + Th2 cells are involved in allergic responses by expressing IL-13, IL-5 and IL-4 which mediate airway hyper reactivity, eosinophil recruitment and IgE production.
  • T cell activation is considered central to many inflammatory immune diseases.
  • compounds that inhibit EZH1 and / or EZH2 activity and suppress T cell activation would be useful for the treatment of T cell mediated inflammatory immune diseases.
  • Inhibitors of EZH1/EZH2 that are useful in treating cancer have been reported in PCT applications PCT/US201 1/035336, PCT/US201 1/035340, and PCT/US201 1/035344.
  • the present invention relates to a method of treating T cell mediated inflammatory immune diseases or T cell mediated hypersensitivity diseases, which comprises administering to a human in need thereof an effective amount of a compound which inhibits EZH2 and/or EZH1 , or a pharmaceutically acceptable salt thereof.
  • the invention relates to a compound or a pharmaceutically acceptable salt thereof which inhibits EZH2 and/or EZH1 for use in treating T cell mediated inflammatory immune diseases or T cell mediated hypersensitivity diseases.
  • the invention relates to the use of a compound or a pharmaceutically acceptable salt thereof which inhibits EZH2 and/or EZH1 in the manufacture of a medicament for treating T cell mediated inflammatory immune diseases or T cell mediated hypersensitivity diseases.
  • Figure 1 inhibitors impair T cell receptor-induced CD4+ T cell proliferation.
  • Figure 3 inhibitors impair T cell receptor-induced IL-2 production in CD4+ T cells.
  • Table 1 EZH-
  • /EZH 2 inhibitors impair T cell receptor-induced effector cytokine production in CD4+ T cells. Cytokine production was measured 72 h post stimulation with 10 g/ml_ CD3 + 2 g/ml_ CD28. Data are presented as plC 50 ⁇ sem; n 4
  • the present invention relates to a method of treating T cell mediated inflammatory immune diseases as described above.
  • the present invention relates to a method of treating T cell mediated hypersensitivity diseases as described above.
  • the present invention also relates to a method of treating T cell mediated inflammatory immune diseases or T cell mediated hypersensitivity diseases, which comprises administering to a human in need thereof an effective amount of a compound, which is represented by Formula (I)
  • X and Z are selected independently from the group consisting of hydrogen, (d- 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-(CrC 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 -Ci 0 )bicycloalkyl, unsubstituted or substituted
  • heterocycloalkyl unsubstituted or substituted heterocycloalkyl-(Ci-C 8 )alkyl or - (C 2 -C 8 )alkenyl, unsubstituted or substituted aryl, unsubstituted or substituted aryl-(Ci- C 8 )alkyl or -(C 2 -C 8 )alkenyl, unsubstituted or substituted heteroaryl, unsubstituted or substituted heteroaryl-(C C 8 )alkyl or -(C 2 -C 8 )alkenyl, halo, cyano, -COR a , -C0 2 R a , - CONR a R b , -CONR a NR a R b , -SR a , -SOR a , -S0 2 R a , -S0 2 NR a R b , nitro, -NR
  • R 1 is (C 1 -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 heterocycloalkyi or -(C 2 -C 8 )alken
  • 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-C 8 )alkyl,
  • heterocycloalkyi unsubstituted or substituted heterocycloalkyl-(Ci-C 8 )alkyl, unsubstituted or substituted aryl, unsubstituted or substituted aryl-(C 1 -C 8 )alkyl, unsubstituted or substituted heteroaryl, unsubstituted or substituted heteroaryl-(C 1 -C 8 )alkyl, cyano, -COR a , -C0 2 R a , -CONR a R b , -CONR a NR 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
  • any (Ci-C 8 )alkyl, (C 2 -C 8 )alkenyl, (C 2 -C 8 )alkynyl, cycloalkyl, cycloalkenyl, bicycloalkyl, heterocycloalkyi, aryl, or heteroaryl group is optionally substituted by
  • 1 , 2 or 3 groups independently selected from the group consisting of -0(Ci- C 6 )alkyl(R c ) 1-2 , -S(Ci-C 6 )alkyl(R c ) 1-2 , -(Ci-C 6 )alkyl(R c ) 1-2 , (Ci-C 8 )alkyl- heterocycloalkyl, (C 3 -C 8 )cycloalkyl-heterocycloalkyl, halo, (CrC 6 )alkyl,
  • any aryl or heteroaryl moiety of said aryl, heteroaryl, aryl(Ci-C 4 )alkyl, or heteroaryl(Ci-C 4 )alkyl is optionally substituted by 1 , 2 or
  • 3 groups independently selected from the group consisting of halo, (C 1 -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 ,
  • R a and R b are each independently hydrogen, (d-C 8 )alkyl, (C 2 -C 8 )alkenyl,
  • heterocycloalkyi aryl, heteroaryl, wherein said (Ci-C 8 )alkyl, (C 2 -C 8 )alkenyl, (C 2 -C 8 )alkynyl, cycloalkyi, cycloalkenyl, bicycloalkyi, heterocycloalkyi ,aryl or heteroaryl group is optionally substituted by 1 , 2 or 3 groups independently selected from halo, hydroxyl, (CrC 4 )alkoxy, amino, (C C 4 )alkylamino, ((Ci-C 4 )alkyl)((Ci-C 4 )alkyl)amino, -C0 2 H, -C0 2 (C C 4 )alkyl, - CONH 2 ,-CONH(Ci-C 4 )alkyl, -CON((C C 4 )alkyl)((Ci-C 4 )alkyl), -S0 2 (C C 4
  • 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 (CrC 4 )alkyl, (Ci-C 4 )haloalkyl, amino, (C 1 -C 4 )alkylamino, ((C 1 -C 4 )alkyl)((C 1 -C 4 )alkyl)amino, hydroxyl, oxo, (C 1 -C 4 )alkoxy, and (C 1 -C 4 )alkoxy(C 1 -C 4 )alkyl, wherein said ring is optionally fused to a (C 3 -C 8 )cycloalkyl, heterocycloalkyi, aryl, or heteroaryl ring;
  • R a and R b taken together with the nitrogen to which they are attached represent a 6- to 10-membered bridged bicyclic ring system optionally fused to a (C 3 -C 8 )cycloalkyl, heterocycloalkyi, aryl, or heteroaryl ring;
  • 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 , -NR a R b , or -C0 2 R a ;
  • the present invention also relates to the above method, wherein the compound of Formula (III) is further defined in one of the subgroups below:
  • X and Z are selected from the group consisting of (Ci-C 8 )alkyl, (C 3 -C 8 )cycloalkyl, heterocycloalkyi, aryl, heteroaryl, -NR a R b , and -OR a ;
  • Y is H or F
  • R 1 is selected from the group consisting of (Ci-C 8 )alkyl, (C 3 -C 8 )cycloalkyl, heterocycloalkyi, aryl, and heteroaryl;
  • R 3 is selected from the group consisting of hydrogen, (Ci-C 8 )alkyl, cyano, trifluoromethyl, -NR a R b , and halo;
  • R 6 is selected from the group consisting of hydrogen, halo, cyano, trifluoromethyl, amino, (C 1 -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 (d-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 )alkyl(R c ) 1-2 , -S(Ci-C 6 )alkyl(R c ) 1-2 , -(CrC 6 )alkyl(R c ) 1-2 , (C C 8 )alkyl-heterocycloalkyl, (C 3 -C 8 )cycloalkyl-heterocycloalkyl, halo, (CrC 6 )alkyl, (C 3 -C 8 )cycloalkyl, (C 5 -C 8 )cycloalkenyl, (C C 6 )haloalkyl, cyano, -
  • 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 , -NR a R b , or -C0 2 R a ;
  • R a and R b are each independently hydrogen, (Ci-C 8 )alkyl, (C 2 -C 8 )alkenyl,
  • heterocycloalkyi aryl, heteroaryl, wherein said (Ci-C 8 )alkyl, (C 2 -C 8 )alkenyl, (C 2 -C 8 )alkynyl, cycloalkyi, cycloalkenyl, bicycloalkyi, heterocycloalkyi ,aryl or heteroaryl group is optionally substituted by 1 , 2 or 3 groups independently selected from halo, hydroxyl, (C 1 -C 4 )alkoxy, amino, (C C 4 )alkylamino, ((C 1 -C 4 )alkyl)((C 1 -C 4 )alkyl)amino, -C0 2 H, -C0 2 (C C 4 )alkyl, - CONH 2 , -CONH(d-C 4 )alkyl, -CON((d-d)alkyl)((d-d)alkyl), -S0 2 (d-d
  • 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, ((d-C 4 )alkyl)((d-C 4 )alkyl)amino, hydroxyl, oxo, (d-d)alkoxy, and (d-C 4 )alkoxy(d-C 4 )alkyl, wherein said ring is optionally fused to a (C 3 -C 8 )cycloalkyl, heterocycloalkyi, aryl, or heteroaryl ring;
  • R a and R b taken together with the nitrogen to which they are attached represent a 6- to 10-membered bridged bicyclic ring system optionally fused to a (C 3 -C 8 )cycloalkyl, heterocycloalkyi, 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:
  • A is O, NH, or S; B is CH or N, and C is h drogen or CrC 8 alkyl; or
  • D is N or C optionally substituted b 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 N
  • M is CH or N
  • L/(5) is hydrogen, halo, amino, cyano, (CrC 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 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 , -N R a N R a C(0)N R a R b , -OR a , wherein any (Ci-C 8 )alkyl, (C 3
  • L/(6) is NH or CH 2 ;
  • M/(7) is hydrogen, halo, amino, cyano, (CrC 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 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 , -NR a NR a C(0)NR b , -NR a NR a C
  • any (d-C 8 )alkyl, (C 3 -C 8 )cycloalkyl, heterocycloalkyl group is optionally substituted by 1 , 2 or 3 groups independently selected from (CrC 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 R
  • P is CH 2 , NH, O, or S; 0/(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, (C 1 -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 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 (d-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, (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 NR a R b , -NR a S0 2 NR a
  • X and Z are selected independently from the group consisting of (Ci-C 8 )alkyl, (C 3 - C 8 )cycloalkyl, heterocycloalkyi, aryl, heteroaryl, -NR a R b , and -OR a ;
  • Y is H
  • R 1 is (Ci-C 8 )alkyl, (C 3 -C 8 )cycloalkyl, or heterocycloalkyi;
  • R 3 is hydrogen, (Ci-C 8 )alkyl or halo
  • R 6 is 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 , or -NR a S0 2 R b ;
  • any (C 1 -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 halo, (Ci-C 6 )alkyl, (C 3 -C 8 )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 S0 2 R b , -NR a S0 2 NR b , -OR a , -OC(0)R a , -OC(0)NR a R b , heterocycloalkyi, 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,
  • heterocycloalkyi aryl, heteroaryl, wherein said (Ci-C 8 )alkyl, (C 2 -C 8 )alkenyl, (C 2 -C 8 )alkynyl, cycloalkyi, cycloalkenyl, bicycloalkyi, heterocycloalkyi ,aryl or heteroaryl group is optionally substituted by 1 , 2 or 3 groups independently selected from halo, hydroxyl, (CrC 4 )alkoxy, amino, (C C 4 )alkylamino, ((Ci-C 4 )alkyl)((Ci-C 4 )alkyl)amino, -C0 2 H, -C0 2 (C C 4 )alkyl, - CONH 2 ,-CONH(Ci-C 4 )alkyl, -CON((C C 4 )alkyl)((Ci-C 4 )alkyl), -S0 2 (C C 4
  • 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 (C 1 -C 4 )alkyl, (C 1 -C 4 )haloalkyl, amino, (C 1 -C 4 )alkylamino, ((C 1 -C 4 )alkyl)((C 1 -C 4 )alkyl)amino, hydroxyl, oxo, (C 1 -C 4 )alkoxy, and (Ci-C 4 )alkoxy(Ci-C 4 )alkyl, wherein said ring is optionally fused to a (C 3 -C 8 )cycloalkyl, heterocycloalkyi, aryl, or heteroaryl ring;
  • R a and R b taken together with the nitrogen to which they are attached represent a 6- to 10-membered bridged bicyclic ring system optionally fused to a (C 3 -C 8 )cycloalkyl, heterocycloalkyi, 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:
  • A is O, NH, or S; B is CH or N, and C is h drogen or CrC 8 alkyl; or
  • D is N or C optionally substituted b hydrogen or C-
  • 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, (CrC 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 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 , -NR a NR a C(0)NR a R b , -OR a , wherein any (d-C 8 )alkyl, (C 3 -C
  • 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 , -
  • any (Ci-C 8 )alkyl, (C 3 -C 8 )cycloalkyl, heterocycloalkyl group is optionally substituted by 1 , 2 or 3 groups independently selected from (CrC 6 )alkyl, (C 3 -C 8 )cycloalkyl, (C 5 -C 8 )cycloalkenyl, (Ci-C 6 )haloalkyl, cyano, -COR a , -C0 2 R a , -
  • P is CH 2 , NH, O, or S; 0/(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, (d-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 S0 2 NR a R b , -NR a NR a R b , -NR a NR a C(0)R b , , -OR a , 4-(1 H- 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 (C 1 -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)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, (1 - methylethyl)cyclopropyl, 1 , 1-dioxo-tetrahydrothiophene-3-yl, 1-Me-piperidin-4-yl, tetrahydrofuran-3-yl, tetrahydropyran-4-yl, /V,/V-dimethyl-1-propanaminyl, benzyl, or 4- pyridyl;
  • R 3 is H, methyl, or Br; and R 6 is methyl, bis(1 , 1-dimethylethyl), bis(l -methylethyl), cyclopropyl, propyl, dimethylamino, ethylamino, (2-hydroxyethyl)amino, 2-propen-1 -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-1 -yl, 4-pyridinylethynyl, phenylethynyl, 2-furanyl, 3-thienyl; 1 H-pyrazol-4-yl, 1 H-indazol-5-yl, 1 H-indazol-6-yl, 3- methyl-1 H-indazol-5-yl, 1 H-1 ,2,3-benzotriazol-5-yl, 2-oxo-2,3-dihydro-1 H-benzimidazol-5- yl, 2-0X0-2, 3-dihydro-1 H-ind
  • the present invention also relates to a method of treating T cell mediated inflammatory immune diseases or T cell mediated hypersensitivity diseases, which comprises administering to a human in need thereof an effective amount of a compound, which is selected from the group comprising:
  • the phrase "optionally substituted” or variations thereof denote an optional substitution, including multiple degrees of substitution, with one or more substitutent group. The phrase should not be interpreted as duplicative of the substitutions herein described and depicted.
  • Exemplary optional substituent groups include acyl, CrC 6 alkyl, d-C 3 alkylsulfonyl, CrC 3 alkoxy, C
  • 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.
  • 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.
  • alkoxy as used herein means -0(Ci.C 8 alkyl) including -OCH3, -
  • alkylthio as used herein is meant -S(C-
  • acyloxy means -OC(0)Ci.C 8 alkyl 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 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.
  • cycloalkyl refers to a non-aromatic, saturated, cyclic hydrocarbon ring containing the specified number of carbon atoms.
  • C 3- CecycloalkyI refers to a non-aromatic cyclic hydrocarbon ring having from three to eight carbon atoms.
  • Exemplary "C 3 -C 8 cycloalkyl” groups useful in the present invention include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and cyclooctyl.
  • C 5 -C 8 cycloalkenyl 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.
  • C 3 -C 8 heterocycloalkyl 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 aromatic, hydrocarbon, ring system.
  • the ring system may be monocyclic or fused polycyclic (e.g., bicyclic, tricyclic, etc.), substituted or unsubstituted.
  • the monocyclic aryl ring is C5-C10, or C5-C7, or C5-C6, where these carbon numbers refer to the number of carbon atoms that form the ring system.
  • a C6 ring system i.e. a phenyl ring, is a suitable aryl group.
  • the polycyclic ring is a bicyclic aryl group, where suitable bicyclic aryl groups are C8-C12, or C9-C10.
  • a naphthyl ring, which has 10 carbon atoms, is a suitable polycyclic aryl group.
  • Suitable substituents for aryl, unless otherwise defined, are described below in the definition of "optionally substituted".
  • heteroaryl an aromatic ring system containing carbon(s) and at least one heteroatom.
  • Heteroaryl may be monocyclic or polycyclic, substituted or unsubstituted.
  • a monocyclic heteroaryl group may have 1 to 4 heteroatoms in the ring, while a polycyclic heteroaryl may contain 1 to 10 hetero atoms.
  • a polycyclic heteroaryl ring may contain fused, spiro or bridged ring junctions, for example, bicyclic heteroaryl is a polycyclic heteroaryl.
  • Bicyclic heteroaryl rings may contain from 8 to 12 member atoms.
  • Monocyclic heteroaryl rings may contain from 5 to 8 member atoms (carbons and heteroatoms).
  • heteroaryl groups include benzofuran, benzothiophene, furan, imidazole, indole, isothiazole, oxazole, pyrazine, pyrazole, pyridazine, pyridine, pyrimidine, pyrrole, quinoline, quinazoline, quinoxaline, thiazole, and thiophene.
  • Suitable substituents for heteroaryl, unless otherwise defined are described below in the definition of "optionally substituted"
  • a method may comprise administering to a human, e.g. a human in need thereof, a therapeutically effective amount of an agent described herein.
  • a method of treatment of T cell mediated inflammatory immune diseases or T cell mediated hypersensitivity diseases in a human comprising administering a therapeutically effective amount of an EZH1 and / EZH2 inhibitor.
  • Inflammation represents a group of vascular, cellular and neurological responses to trauma. Inflammation can be characterised as the movement of inflammatory cells such as monocytes, neutrophils and granulocytes into the tissues. This is usually associated with reduced endothelial barrier function and oedema into the tissues. Inflammation can be classified as either acute or chronic. Acute inflammation is the initial response of the body to harmful stimuli and is achieved by the increased movement of plasma and leukocytes from the blood into the injured tissues. A cascade of biochemical event propagates and matures the inflammatory response, involving the local vascular system, the immune system, and various cells within the injured tissue. Prolonged inflammation, known as chronic inflammation, leads to a progressive shift in the type of cells which are present at the site of inflammation and is characterised by simultaneous destruction and healing of the tissue from the inflammatory process.
  • Acute inflammation is the initial response of the body to harmful stimuli and is achieved by the increased movement of plasma and leukocytes from the blood into the injured tissues.
  • a cascade of biochemical event propag
  • inflammation When occurring as part of an immune response to infection or as an acute response to trauma, inflammation can be beneficial and is normally self-limiting. However, inflammation can be detrimental under various conditions. This includes the production of excessive inflammation in response to infectious agents, which can lead to significant organ damage and death (for example, in the setting of sepsis). Moreover, chronic inflammation is generally deleterious and is at the root of numerous chronic diseases, causing severe and irreversible damage to tissues. In such settings, the immune response is often directed against self-tissues (autoimmunity), although chronic responses to foreign entities can also lead to bystander damage to self tissues.
  • autoimmunity autoimmunity
  • the aim of anti-inflammatory therapy is therefore to reduce this inflammation, to inhibit autoimmunity when present and to allow for the physiological process or healing and tissue repair to progress.
  • the agents may be used to treat inflammation of any tissue and organs of the body, including musculoskeletal inflammation, vascular inflammation, neural inflammation, digestive system inflammation, ocular inflammation, inflammation of the reproductive system, and other inflammation, as exemplified below.
  • Musculoskeletal inflammation refers to any inflammatory condition of the musculoskeletal system, particularly those conditions affecting skeletal joints, including joints of the hand, wrist, elbow, shoulder, jaw, spine, neck, hip, knew, ankle, and foot, and conditions affecting tissues connecting muscles to bones such as tendons.
  • musculoskeletal inflammation examples include arthritis (including, for example, osteoarthritis, psoriatic arthritis, ankylosing spondylitis, acute and chronic infectious arthritis, arthritis associated with gout and pseudogout, and juvenile idiopathic arthritis), tendonitis, synovitis, tenosynovitis, bursitis, fibrositis (fibromyalgia), epicondylitis, myositis, and osteitis (including, for example, Paget's disease, osteitis pubis, and osteitis fibrosa cystic).
  • arthritis including, for example, osteoarthritis, psoriatic arthritis, ankylosing spondylitis, acute and chronic infectious arthritis, arthritis associated with gout and pseudogout, and juvenile idiopathic arthritis
  • tendonitis synovitis
  • tenosynovitis bursitis
  • fibrositis fibromyalgia
  • epicondylitis myos
  • Ocular inflammation refers to inflammation of any structure of the eye, including the eye lids.
  • ocular inflammation which may be treated in this invention include blepharitis, blepharochalasis, conjunctivitis, dacryoadenitis, keratitis, keratoconjunctivitis sicca (dry eye), scleritis, trichiasis, and uveitis.
  • Examples of inflammation of the nervous system which may be treated in this invention include encephalitis, Guillain-Barre syndrome, meningitis, neuromyotonia, narcolepsy, multiple sclerosis, myelitis and schizophrenia.
  • Examples of inflammation of the vasculature or lymphatic system which may be treated in this invention include arthrosclerosis, arthritis, phlebitis, vasculitis, and lymphangitis.
  • Examples of inflammatory conditions of the digestive system which may be treated in this invention include cholangitis, cholecystitis, enteritis, enterocolitis, gastritis, gastroenteritis, ileitis, and proctitis.
  • Examples of inflammatory conditions of the reproductive system which may be treated in this invention include cervicitis, chorioamnionitis, endometritis, epididymitis, omphalitis, oophoritis, orchitis, salpingitis, tubo-ovarian abscess, urethritis, vaginitis, vulvitis, and vulvodynia.
  • the agents may be used to treat autoimmune conditions having an inflammatory component.
  • Such conditions include acute disseminated alopecia universalise, Behcet's disease, Chagas' disease, chronic fatigue syndrome, dysautonomia, encephalomyelitis, ankylosing spondylitis, aplastic anemia, hidradenitis suppurativa, autoimmune hepatitis, autoimmune oophoritis, celiac disease, Crohn's disease, diabetes mellitus type 1 , giant cell arteritis, goodpasture's syndrome, Grave's disease, Guillain-Barre syndrome, Hashimoto's disease, Henoch-Schonlein purpura, Kawasaki's disease, lupus erythematosus, microscopic colitis, microscopic polyarteritis, mixed connective tissue disease, multiple sclerosis, myasthenia gravis, opsoclonus myoclonus syndrome, optic neuritis, ord's thyroid
  • the agents may be used to treat T-cell mediated hypersensitivity diseases having an inflammatory component.
  • T-cell mediated hypersensitivity diseases having an inflammatory component.
  • Such conditions include contact hypersensitivity, contact dermatitis (including that due to poison ivy), uticaria, skin allergies, respiratory allergies (hayfever, allergic rhinitis) and gluten-sensitive enteropathy (Celliac disease).
  • inflammatory conditions which may be treated in this invention include, for example, appendicitis, dermatitis, dermatomyositis, endocarditis, fibrositis, gingivitis, glossitis, hepatitis, hidradenitis suppurativa, ulceris, laryngitis, mastitis, myocarditis, nephritis, otitis, pancreatitis, parotitis, percarditis, peritonoitis, pharyngitis, pleuritis, pneumonitis, prostatistis, pyelonephritis, and stomatisi, transplant rejection (involving organs such as kidney, liver, heart, lung, pancreas (e.g., islet cells), bone marrow, cornea, small bowel, skin allografts, skin homografts, and heart valve xengrafts, sewrum sickness, and graft vs host disease), acute pan
  • Preferred treatments include any one of treatment of transplant rejection, psoriatic arthritis, multiple sclerosis, Type 1 diabetes, asthma, systemic lupus erythematosis, chronic pulmonary disease, and inflammation accompanying infectious conditions (e.g., sepsis).
  • salts of the compounds for use in the inevntion are pharmaceutically acceptable salts. Salts encompassed within the term
  • salts of the compounds of the present invention may comprise acid addition salts.
  • the salts are formed from pharmaceutically acceptable inorganic and organic acids. More specific examples of suitable acid salts include maleic, hydrochloric, hydrobromic, sulphuric, phosphoric, nitric, perchloric, fumic, acetic, propionic, succinic, glycolic, formic, lactic, aleic, tartaric, citric, palmoic, malonic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicylic, fumaric, toluenesulfonic, methansulfonic (mesylate), naphthalene-2-sulfonic, benzenesulfonic, hydroxynaphthoic, hydroiodic, malic, teroic, tannic, and the like.
  • salts include acetate, benzenesulfonate, benzoate, bicarbonate, bisulfate, bitartrate, borate, calcium edetate, camsylate, carbonate, clavulanate, citrate, dihydrochloride, edisylate, estolate, esylate, fumarate, gluceptate, gluconate, glutamate, glycollylarsanilate, hexylresorcinate, hydrobromide, hydrochloride, hydroxynaphthoate, iodide, isethionate, lactate, lactobionate, laurate, malate, maleate, mandelate, mesylate, methylsulfate, monopotassium maleate, mucate, napsylate, nitrate, oxalate, pamoate (embonate), palmitate, pantothenate, phosphate/diphosphate, polygalacturonate, salicylate, a
  • salts which are not pharmaceutically acceptable, may be useful in the preparation of compounds for use in this invention.
  • These salts such as oxalic or trifluoroacetate, while not in themselves pharmaceutically acceptable, may be useful in the preparation of salts useful as intermediates in obtaining the compounds of the invention and their pharmaceutically acceptable salts.
  • 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 non-aqueous 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 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.
  • 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
  • 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 for use in this 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 non-aqueous 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 non-aqueous 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 for use in this 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 0.001 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.
  • 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-lnterscience, 1994).
  • Irradiation levels utilized i.e. high, normal, low
  • 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 (1 Og - 100 g).
  • 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.,
  • 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-10ADvp (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 (C18) 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 * 4.6 mm, 5 /jm) was used.
  • HPLC was done on Agilent-1200 series UPLC system equipped with column Zorbax SB-C18 (50 * 4.6 mm, 1.8 /jm) 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.
  • Scheme 1 illustrates two methods to synthesize a compound of formula (VII).
  • Substituted aminopyrazoles of formula (I) are heated with diethyl oxobutanedione in benzene or toluene at 62 °C overnight.
  • Treatment of the putative intermediate with acetic acid and typically heating at reflux furnishes azaindazole compounds of formula (II).
  • Compounds of formula (II) are converted to compounds of formula (III) by base-catalyzed hydrolysis of the ethyl ester and then chlorination of the putative carboxylic acid intermediate with POCI 3 under standard conditions to afford compounds of formula (IV).
  • compounds of formula (II) are converted to the corresponding triflate (I la) using standard methods.
  • Compounds of formula (I la) are then substituted at the 6-pos. using standard palladium mediated cross- coupling conditions, followed by base-catalyzed hydrolysis of the ethyl ester group to afford compounds of formula (IVa).
  • Treatment of compounds of formula (IVa) with substituted aminomethyl pyridones of formula (V) using EDC, HOAT, N-methylmorpholine, and DMSO at room temperature for a period of no less than 12 h stirring at room temperature affords compounds of formula (VII).
  • Scheme 4 illustrates the method to synthesize a compound of formula (V).
  • Step 1
  • Step 1
  • step 2 from 4-cyclopropyl-6-methyl-2-oxo-1,2-dihydro-3- pyridinecarbonitrile (5 g, 28.7 mmol).
  • the product was collected as a TFA salt, 0.50 g.
  • LCMS E-S (M+H) 179.1.
  • 1 H NMR 400 MHz, DMSO-d 6 ) ⁇ ppm 11.76 - 11.78 (br s, 1H), 7.82-7.92 (brs, 3H), 5.61 (s, 1H), 3.94-3.99 (m, 2H), 2.11 (s, 3H), 1.98-2.05 (m, 1H), 0.95 - 1.01 (m, 2H), 0.74 - 0.79 (m, 2H).
  • 6-Cyclopropyl-4-methyl-2-oxo-1 ,2-dihydro-3-pyridinecarbonitrile (0.35 g, 2.01 mmol) was added to methanol (20 mL) and the stirring contents cooled to -10 °C.
  • di-tert-butyloxycarbonyl (0.933 mL, 4.02 mmol) and the suspension was stirred for 15 min.
  • NiCI 2 -6H 2 0 0.055 g, 0.201 mmol
  • NaBH 4 0.532 g, 14.06 mmol
  • the crude product was purified by silica gel chromatography (eluent: 10% Methanol in Dichloromethane). The collected product was dried under hi-vacuum for 1 h, and then treated with ether and filtered. After drying in vacuum oven at 45 °C for 2 h, the product was collected as 0.28 g (50%).
  • 6-Chloro-1-(1-methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid (0.12g, 0.501 mmol), 1 -hydroxy-7-azabenzotriazole (0.102 g, 0.751 mmol), EDC (0.144 g, 0.751 mmol), and 3-(aminomethyl)-4,6-dimethyl-2(1 H)-pyridinone (0.123 g, 0.651 mmol) were dissolved in dimethyl sulfoxide (3.0 mL) and stirred at room temperature. Added next to the stirring contents was N-methylmorpholine (0.220 mL, 2.003 mmol) via syringe at once.
  • 6-Cyclopropyl-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid (167 mg, 0.680 mmol), 3-(aminomethyl)-4-methyl-6-propyl-2(1 /-/)-pyridinone trifluoroacetate (200 mg, 0.680 mmol), HOAT (139 mg, 1 .019 mmol), EDC (195 mg, 1 .019 mmol), and N- methylmorpholine (0.299 mL, 2.72 mmol) were dissolved in DMF(6 mL) and stirred at 40 °C for 24 h.
  • the title compound was prepared in the same manner as described in example 7 from 6-cyclopropyl-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid (250 mg, 1 .019 mmol), 3-(aminomethyl)-6-methyl-4-(1 -methylethyl)-2(1 H)-pyridinone « TFA (300 mg, 1 .019 mmol), HOAT (208 mg, 1.529 mmol), EDC (293 mg, 1 .529 mmol), N- methylmorpholine (0.448 mL, 4.08 mmol), and DMF(6 mL), wherein the reaction time was 48 h.
  • the title compound was prepared in the same manner as described in example 9 from 1 -(1 -methylethyl)-6-(4-pyridinyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid (403 mg, 1 .427 mmol), 3-(aminomethyl)-6-ethyl-4-methyl-2(1 H)-pyridinone « TFA (400 mg, 1 .427 mmol), HOAT (291 mg, 2.141 mmol), EDC (1094 mg, 5.71 mmol), N-methylmorpholine (0.628 mL, 5.71 mmol), and DMF(6 mL). The final product was collected as a white solid, 232 mg (38%).
  • the title compound was prepared in the same manner as described in example 15 from 6-cyclopropyl-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid (147 mg, 0.60 mmol), 3-(aminomethyl)-4-cyclohexyl-6-methyl-2(1 H)-pyridinone trifluoroacetate (221 mg, 0.660 mmol), EDC (150 mg, 0.780 mmol), HOAT (106 mg, 0.780 mmol), N- methylmorpholine (0.264 mL, 2.400 mmol) and DMF(3 mL), wherein the stir time was 3 d and the final product was not treated with EtOH.
  • the title compound was prepared in the same manner as described in example 19 from 6-cyclopropyl-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid (0.12 g, 0.489 mmol), 1 -hydroxy-7-azabenzotriazole (0.100 g, 0.734 mmol), 3-(aminomethyl)-6- methyl-4-(trifluoromethyl)-2(1 H)-pyridinone (0.154 g, 0.636 mmol, DMSO (3.0 ml_), N- methylmorpholine (0.215 ml_, 1 .957 mmol), and EDC (0.141 g, 0.734 mmol)
  • the crude solid was purified by silica gel chromatography (eluent: gradient 5-100% of 10% 2M NH 3 (in MeOH/DCM) and DCM) and the collected product dried in vacuum oven for 5h. The final product was collected as 0.1 12
  • the suspension was stirred and degassed with nitrogen for 5 min, forming an emulsion.
  • 2-methyl-3-butyn-2-ol 0.194 mL, 2.006 mmol
  • Pd(Ph 3 P) 4 0.046 g, 0.040 mmol.
  • the stirring contents were heated at 90 °C for 3 hr, and then allowed to cool to room temperature.
  • the reaction mixture was poured into a solution of water and 20% THF/EtOAc, and stirred. The layers were separated, and the organic layer washed with brine. The organic layer was dried over MgS0 4 , and then filtered through Celite, washing the filter pad with additional EtOAc.
  • the stirring suspension was degassed with nitrogen for 5 min., wherein an emulsion had formed.
  • Added next were phenylacetylene (0.1 10 mL, 1 .003 mmol) and Pd(Ph 3 P) 4 (0.039 g, 0.033 mmol).
  • the sealed reaction mixture was placed onto a heat block, stirred at 90 °C for 3 hr, and then allowed to cool to room temperature overnight.
  • the contents were poured onto water and 20% THF/EtOAc, stirred, and the layers separted.
  • the organic layer was washed with brine, dried over MgS0 4 , filtered, and concentrated in vacuo.
  • the filter pad was washed with additional EtOAc.
  • the combined filtrates were concentrated in vacuo to a yellow/orange residue that was dried on hi-vac pump.
  • the crude solid was then pre-adsorbed onto silica gel and purified by silica gel chromatography (dry loaded, eluent: 5-80 % gradient of DCM and chloroform containing 10% 2M Ammonia (in methanol)).
  • the isolated product was obtained as a yellow solid which was then further purified by reverse phase HPLC (mobile phase: 20-90% ACN in H 2 0, 0.1 % TFA, Gradient time: 8min).
  • the isolated solid was dissolved in 10% MeOH/ CH 2 CI 2 and treated with 0.6 g of Silicycle carbonate resin for 30 min.
  • the title compound was prepared in the same manner as described in example 31 from 6-chloro-N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-1 H- pyrazolo[3,4-b]pyridine-4-carboxamide (.15 g, 0.401 mmol), sodium iodide (0.012 g, 0.080 mmol), zinc (5.25 mg, 0.080 mmol), DMSO (4.0 ml_), triethylamine (0.168 ml_, 1 .204 mmol), DBU (0.121 ml_, 0.802), 4-ethynylpyridine (0.1 12 g, 0.802 mmol), and Pd(Ph 3 P) 4 (0.046 g, 0.040 mmol).
  • the contents were transferred to a heat block and heated at 135 °C for 16 hr., and then at 145 °C for an additional 12 h. After cooling to room temperature, the contents were diluted with CH 2 CI 2 and pre-absorbed onto silica gel.
  • the crude product was purified by silica gel chromatography (dry loaded, eluent; gradient of 5-80% DCM and chloroform containing 10% 2M Ammonia (in methanol)). The isolated solid was triturated with MTBE, filtered, and washed with additional MTBE. The collected solid was dried in vacuum oven at 45 °C overnight to afford the final product as 0.067 g (55%).
  • LCMS E-S (M+H) 445.3.
  • PdCI 2 (dppf)-CH 2 CI 2 adduct (8.74 mg, 0.01 1 mmol) was added and the resulting mixture was degassed with nitrogen for 10 min.
  • Sodium bicarbonate (53.9 mg, 0.64 mmol) was added, the vessel was sealed, and reaction mixture was heated in a microwave at 150 °C for 30 min. After cooling, 2 mL of water was added to the black mixture and solids that precipitated were filtered.
  • the reaction mixture was degassed with nitrogen for 5 min.
  • Sodium bicarbonate (67.4 mg, 0.802 mmol) was added and the contents sealed and irradiated (microwave) at 140 °C.
  • the reaction mixture was cooled to room temperature and poured on a silica column (through Na 2 S0 4 ) and purified by silica gel chromatography (eluent : 5% MeOH/CH 2 CI 2 ) which provided the desired product as an off-white solid after preciptation from EtOAc/MeOH.
  • the final product was collected as
  • the title compound was prepared in the same manner as described in example 74 from 6-chloro-N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-1 H- pyrazolo[3,4-b]pyridine-4-carboxamide (70 mg, 0.187 mmol), 6-(4,4,5,5-tetramethyl-1 ,3,2- dioxaborolan-2-yl)-1 H-indazole (59.4 mg, 0.243 mmol), DMSO(1 .5 mL), sodium carbonate (0.281 mL, 0.562 mmol), and bis(triphenylphosphine)palladium(l l) chloride (10.51 mg,
  • the title compound was prepared in the same manner as described in example 74 using 6-chloro-N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)- 1 H-pyrazolo[3,4-b]pyridine-4-carboxamide (70 mg, 0.187 mmol), 1 H-1 ,2,3-benzotriazol-5- ylboronic acid (39.7 mg, 0.243 mmol), DMSO(2 mL), sodium carbonate (0.281 mL, 0.562 mmol) and bis(triphenylphosphine)palladium(ll) chloride (10.51 mg, 0.015 mmol).
  • the title compound was prepared in the same manner as described in example 74 using 6-chloro-N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)- 1 H-pyrazolo[3,4-b]pyridine-4-carboxamide (70 mg, 0.187 mmol), 6-(4,4,5,5-tetramethyl- 1 ,3,2-dioxaborolan-2-yl)-1 ,3-dihydro-2H-indol-2-one (63.1 mg, 0.243 mmol), DMSO(1 .5 mL), sodium carbonate (0.281 mL, 0.562 mmol), and bis(triphenylphosphine)palladium(l l) chloride (10.51 mg, 0.015 mmol).
  • the title compound was prepared in the same manner as described in example 74 using 6-chloro-N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)- 1 H-pyrazolo[3,4-b]pyridine-4-carboxamide (70 mg, 0.187 mmol), 5-(4,4,5,5-tetramethyl- 1 ,3,2-dioxaborolan-2-yl)-1 ,3-dihydro-2H-indol-2-one (63.1 mg, 0.243 mmol), DMSO(1 .5 mL), sodium carbonate (0.281 mL, 0.562 mmol), and bis(triphenylphosphine)palladium(ll) chloride (10.51 mg, 0.015 mmol).

Abstract

The present invention relates to a method of treating T cell mediated inflammatory immune diseases or T cell mediated hypersensitivity diseases, which comprises administering to a human in need thereof an effective amount of a compound which inhibits EZH2 and/or EZH1, or a pharmaceutically acceptable salt thereof.

Description

METHOD OF TREATMENT
Field of the Invention
This invention relates to compounds which inhibit EZH2/EZH1 and their uses for treating T cell mediated inflammatory immune diseases.
Background of the Invention
Posttranslational modifications of proteins play a critical role in the regulation of signal transduction from receptors, chromatin remodelling and gene transcription. These modifications include acetylation, methylation, phosphorylation, ubiquitinylation, SUMOylation. EZH (enhancer of zeste homolog) 1 and 2 are the catalytic subunits of the Polycomb Repressor Complex 2 (PRC2) and exhibit methyltransferase activity that can catalyse the methylation of lysine amino acids (Margueron R, Reinberg D:The Polycomb complex PRC2 and its mark in life. Nature. 201 1 Jan 20;469 (7330):343-9)
EZH1 and EZH2 play a critical role in the epigenetic long term silencing of gene expression by di- or tri-methylating lysine 27 of histone H3 (H3K27me2/3). Histone H3 is one of the five main histone proteins involved in the structure of chromatin in eukaryotic cells. Chromatin is the complex combination of DNA and protein that makes up chromosomes. It is found inside the nuclei of eukaryotic cells and is divided between heterochromatin (condensed) and euchromatin (extended). The basic building blocks of chromatin are nucleosomes, each of which is composed of 146 base pairs of DNA wrapped around a histone octamer that consists of 2 copies of each H2A, H2B, H3 and H4. The functions of chromatin are to package DNA into a smaller volume to fit in the cell, to strengthen the DNA to allow mitosis and meiosis, and to serve as a mechanism to control gene expression and DNA replication. The chromatin structure is controlled by a series of post translational modifications to histone proteins, notably histones H3 and H4, and most commonly within the "histone tails" which extend beyond the core nucleosome structure. Binding of enzymes and adaptor proteins to posttranslational modification in histone tails regulates chromatin dynamics and gene expression. H3K27me3 is thought to silence gene expression by recruiting histone deacetylases to the modified nucleosomes and stall transcriptional elongation by polymerase II. Thus, inhibition of the enzymatic activity of EZH1 and EZH2 may result in a loss of H3K27me3 and up-regulation of target genes.
In addition to its nuclear function in histone H3 modification EZH2 has been implicated in the regulation of signal transduction that leads to actin polymerization in the cytoplasm of cells (Su IH, Dobenecker MW, Dickinson E, Oser M, Basavaraj A, Marqueron R, Viale A, Reinberg D, WQIfing C, Tarakhovsky A: Polycomb group protein ezh2 controls actin polymerization and cell signaling. Cell. 2005 May 6; 121 (3):425-36). The reorganization of the actin cytoskeleton critically contributes to T cell responses by facilitating the interaction of T cells with antigen presenting cells or target cells. In addition, actin remodelling plays an important role in T cell migration and motility during their recruitment to the sites of inflammation. A fraction of EZH2 protein was found to localize to the cytoplasm of T cells and to interact with the small GTPase VAV1 , which is involved in actin remodelling. Genetic elimination of EZH2 resulted in impaired polymerization of actin in TCR stimulated T cells or at the T cell - antigen presenting cell interphase. Furthermore, actin polymerization induced by EZH2 over-expression was dependent on the methytransferase activity of EZH2. Proliferation of T cells in response to TCR was also impaired in the absence of EZH2. Thus, inhibition of EZH1 and / or EZH2 may suppress the activation of T cells.
Mature T cell respond to foreign peptide antigens in the presence of appropriate co-stimulation by antigen presenting cells. They have the capability to discriminate between self and non self as a consequence of the selection of a TCR repertoire specific for foreign antigens in the thymus, tolerance induction of self reactive T cell clones in the periphery, and control of T cell activation by self antigen by regulatory T cells. T cells provide protection against different classes of pathogens by mediating distinct types of adaptive immune responses as a consequence of the expression of distinct sets of cytokines and other soluble and cell-bound products. In addition, they act as principle amplifiers and inducers of the appropriate inflammatory and effector responses in cells of the innate immune system and nonimmune cells. While such concerted immune responses can provide powerful protection against pathogens it can also result in inflammation associated with unwanted immune responses against self and environmental antigens and commensal microorganisms as well as collateral damage to the host as a side effect of immune responses against pathogens. CD8 T cells can lyse cells bearing intracellular pathogens but may also contribute to tissue damage and secrete proinflammatory cytokines, e.g. TNF and IFNg. CD4 T cells can have diverse functions in inflammation depending on their specific cytokine expression profiles. CD4+ Th1 cells are important for the clearance of intracellular pathogens but also play a critical role in inflammation through the expression of TNF and IFNg. IL-17 expressing CD4+ Th17 cells, which mediate neutrophilia and tissue remodelling and repair, have also been shown to be involved in many inflammatory conditions. CD4+ Th2 cells are involved in allergic responses by expressing IL-13, IL-5 and IL-4 which mediate airway hyper reactivity, eosinophil recruitment and IgE production. Thus, T cell activation is considered central to many inflammatory immune diseases. Accordingly, compounds that inhibit EZH1 and / or EZH2 activity and suppress T cell activation would be useful for the treatment of T cell mediated inflammatory immune diseases. Inhibitors of EZH1/EZH2 that are useful in treating cancer have been reported in PCT applications PCT/US201 1/035336, PCT/US201 1/035340, and PCT/US201 1/035344.
Summary of the Invention
The present invention relates to a method of treating T cell mediated inflammatory immune diseases or T cell mediated hypersensitivity diseases, which comprises administering to a human in need thereof an effective amount of a compound which inhibits EZH2 and/or EZH1 , or a pharmaceutically acceptable salt thereof.
In a further aspect the invention relates to a compound or a pharmaceutically acceptable salt thereof which inhibits EZH2 and/or EZH1 for use in treating T cell mediated inflammatory immune diseases or T cell mediated hypersensitivity diseases.
In a further aspect the invention relates to the use of a compound or a pharmaceutically acceptable salt thereof which inhibits EZH2 and/or EZH1 in the manufacture of a medicament for treating T cell mediated inflammatory immune diseases or T cell mediated hypersensitivity diseases.
Brief Description of Drawings
Figure 1 : inhibitors impair T cell receptor-induced CD4+ T cell proliferation.
Division index was calculated by CFSE fluorescence 6 days post stimulation with 10 g/mL CD3 + 2 g/mL CD28. Data are presented as mean ± standard error of the mean, n=4.
Figure 2: inhibitors impair T cell receptor-induced effector cytokine production in CD4+ T cells. Cytokine production was measured 72 h post stimulation with 10 g/mL CD3 + 2 g/mL CD28. Data are presented as means ± standard error of the mean, n=4.
Figure 3: inhibitors impair T cell receptor-induced IL-2 production in CD4+ T cells. IL-2 production was measured 18 h post stimulation with 10 g/mL CD3 + 2 g/mL CD28. Data are presented as means ± standard error of the mean, n=4. Table 1 : EZH-|/EZH2 inhibitors impair T cell receptor-induced effector cytokine production in CD4+ T cells. Cytokine production was measured 72 h post stimulation with 10 g/ml_ CD3 + 2 g/ml_ CD28. Data are presented as plC50 ± sem; n=4
Detailed Description of the Invention
The present invention relates to a method of treating T cell mediated inflammatory immune diseases as described above.
The present invention relates to a method of treating T cell mediated hypersensitivity diseases as described above.
The present invention also relates to a method of treating T cell mediated inflammatory immune diseases or T cell mediated hypersensitivity diseases, which comprises administering to a human in need thereof an effective amount of a compound, which is represented by Formula (I)
Figure imgf000005_0001
wherein
X and Z are selected independently from the group consisting of hydrogen, (d- C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, unsubstituted or substituted (C3-C8)cycloalkyl, unsubstituted or substituted (C3-C8)cycloalkyl-(CrC8)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-Ci0)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 (C1-C8)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 heterocycloalkyi 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-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-(d-C8)alkyl, unsubstituted or substituted (C5-C8)cycloalkenyl, unsubstituted or substituted (C5- C8)cycloalkenyl-(Ci-C8)alkyl, (C6-Ci0)bicycloalkyl, unsubstituted or substituted
heterocycloalkyi, unsubstituted or substituted heterocycloalkyl-(Ci-C8)alkyl, unsubstituted or substituted aryl, unsubstituted or substituted aryl-(C1-C8)alkyl, unsubstituted or substituted heteroaryl, unsubstituted or substituted heteroaryl-(C1-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, heterocycloalkyi, 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)1-2, -S(Ci-C6)alkyl(Rc)1-2, -(Ci-C6)alkyl(Rc)1-2, (Ci-C8)alkyl- heterocycloalkyl, (C3-C8)cycloalkyl-heterocycloalkyl, halo, (CrC6)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, heterocycloalkyi, 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, (C1-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, and -OC(0)NRaRb;
Ra and Rb are each independently hydrogen, (d-C8)alkyl, (C2-C8)alkenyl,
(C2-C8)alkynyl, (C3-C8)cycloalkyl, (C5-C8)cycloalkenyl, (C6-Ci0)bicycloalkyl,
heterocycloalkyi, aryl, heteroaryl, wherein said (Ci-C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, cycloalkyi, cycloalkenyl, bicycloalkyi, heterocycloalkyi ,aryl or heteroaryl group is optionally substituted by 1 , 2 or 3 groups independently selected from halo, hydroxyl, (CrC4)alkoxy, amino, (C C4)alkylamino, ((Ci-C4)alkyl)((Ci-C4)alkyl)amino, -C02H, -C02(C C4)alkyl, - CONH2,-CONH(Ci-C4)alkyl, -CON((C C4)alkyl)((Ci-C4)alkyl), -S02(C C4)alkyl, - S02NH2,-S02NH(Ci-C4)alkyl, or -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 (CrC4)alkyl, (Ci-C4)haloalkyl, amino, (C1-C4)alkylamino, ((C1-C4)alkyl)((C1-C4)alkyl)amino, hydroxyl, oxo, (C1-C4)alkoxy, and (C1-C4)alkoxy(C1-C4)alkyl, wherein said ring is optionally fused to a (C3-C8)cycloalkyl, heterocycloalkyi, 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, heterocycloalkyi, aryl, or heteroaryl ring;
each Rc is independently (Ci-C4)alkylamino, -NRaS02Rb, -SORa, -S02Ra, - NRaC(0)ORa, -NRaRb, or -C02Ra;
or a salt thereof.
The present invention also relates to the above method, wherein the compound of Formula (III) is further defined in one of the subgroups below:
Subgroup (l)(A)
X and Z are selected from the group consisting of (Ci-C8)alkyl, (C3-C8)cycloalkyl, heterocycloalkyi, aryl, heteroaryl, -NRaRb, and -ORa;
Y is H or F;
R1 is selected from the group consisting of (Ci-C8)alkyl, (C3-C8)cycloalkyl, heterocycloalkyi, 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, (C1-C8)alkyl, (C3-C8)cycloalkyl;, aryl, heteroaryl, acylamino; (C2-C8)alkynyl, arylalkynyl, heteroarylalkynyl; -S02Ra: -S02NRaRb , and -NRaS02Rb ;
wherein any (d-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)alkyl(Rc)1-2, -S(Ci-C6)alkyl(Rc)1-2, -(CrC6)alkyl(Rc)1-2, (C C8)alkyl-heterocycloalkyl, (C3-C8)cycloalkyl-heterocycloalkyl, halo, (CrC6)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, heterocycloalkyi, aryl, heteroaryl, aryl(Ci-C4)alkyl, and
heteroaryl(Ci-C4)alkyl;
each Rc is independently (Ci-C4)alkylamino, -NRaS02Rb, -SORa, -S02Ra, - NRaC(0)ORa, -NRaRb, 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,
heterocycloalkyi, aryl, heteroaryl, wherein said (Ci-C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, cycloalkyi, cycloalkenyl, bicycloalkyi, heterocycloalkyi ,aryl or heteroaryl group is optionally substituted by 1 , 2 or 3 groups independently selected from halo, hydroxyl, (C1-C4)alkoxy, amino, (C C4)alkylamino, ((C1-C4)alkyl)((C1-C4)alkyl)amino, -C02H, -C02(C C4)alkyl, - CONH2, -CONH(d-C4)alkyl, -CON((d-d)alkyl)((d-d)alkyl), -S02(d-d)alkyl, - S02NH2,-S02NH(d-C4)alkyl, and -S02N((Ci-d)alkyl)((C 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 (d-d)alkyl, (d-d)haloalkyl, amino, (d-d)alkylamino, ((d-C4)alkyl)((d-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, heterocycloalkyi, 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, heterocycloalkyi, 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 imgf000009_0001
(1 )
wherein in (1 ),
A is O, NH, or S; B is CH or N, and C is h drogen or CrC8 alkyl; or
Figure imgf000009_0002
wherein in (2),
D is N or C optionally substituted b hydrogen or Ci-C8 alkyl; or
Figure imgf000009_0003
wherein in (3),
E is NH or CH2; F is O or CO; and G is NH or CH2; or
Figure imgf000009_0004
wherein in (4),
J is O, S or CO; or
Figure imgf000009_0005
wherein in (5),
Q is CH or N;
M is CH or N; and
L/(5) is hydrogen, halo, amino, cyano, (CrC8)alkyl, (C3-C8)cycloalkyl, -CORa, - C02Ra, -CONRaRb, -CONRaNRaRb, -S02Ra, -S02NRaRb, -NRaRb, -NRaC(0)Rb - NRaS02Rb, -NRaS02NRaRb, -NRaNRaRb, -NRaNRaC(0)Rb, -N RaN RaC(0)N RaRb, -ORa, wherein any (Ci-C8)alkyl, (C3-C8)cycloalkyl, group is optionally substituted by 1 , 2 or 3 groups independently selected from (d-C6)alkyl, (C3-C8)cycloalkyl, (C5-Ce)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 imgf000010_0001
wherein in 6,
L/(6) is NH or CH2; or
Figure imgf000010_0002
wherein in 7,
M/(7) is hydrogen, halo, amino, cyano, (CrC8)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 (d-C8)alkyl, (C3-C8)cycloalkyl, heterocycloalkyl group is optionally substituted by 1 , 2 or 3 groups independently selected from (CrC6)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 and Rb are defined as above; or
Figure imgf000010_0003
wherein in (8),
P is CH2, NH, O, or S; 0/(8) is CH or N; and n is 0-2; or
Figure imgf000010_0004
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, (C1-C8)alkyl, (C3- C8)cycloalkyl, -CORa, -C02Ra, -CONRaRb, -S02Ra, -S02NRaRb, -NRaRb, - NRaC(0)Rb,-NRaS02Rb, -NRaS02NRaRb, -NRaNRaRb, -NRaNRaC(0)Rb, -ORa, 4-(1 H- pyrazol-4-yl),
wherein any (d-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, (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.
Subgroup (l)(B)
X and Z are selected independently from the group consisting of (Ci-C8)alkyl, (C3- C8)cycloalkyl, heterocycloalkyi, aryl, heteroaryl, -NRaRb, and -ORa;
Y is H;
R1 is (Ci-C8)alkyl, (C3-C8)cycloalkyl, or heterocycloalkyi;
R3 is hydrogen, (Ci-C8)alkyl or halo;
R6 is hydrogen, halo, cyano, trifluoromethyl, amino, (Ci-C8)alkyl, (C3- C8)cycloalkyl;, aryl, heteroaryl, acylamino; (C2-C8)alkynyl, arylalkynyl, heteroarylalkynyl; - S02Ra; -S02NRaRb, or -NRaS02Rb;
wherein any (C1-C8)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, heterocycloalkyi, 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, (C6-Ci0)bicycloalkyl,
heterocycloalkyi, aryl, heteroaryl, wherein said (Ci-C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, cycloalkyi, cycloalkenyl, bicycloalkyi, heterocycloalkyi ,aryl or heteroaryl group is optionally substituted by 1 , 2 or 3 groups independently selected from halo, hydroxyl, (CrC4)alkoxy, amino, (C C4)alkylamino, ((Ci-C4)alkyl)((Ci-C4)alkyl)amino, -C02H, -C02(C C4)alkyl, - CONH2,-CONH(Ci-C4)alkyl, -CON((C C4)alkyl)((Ci-C4)alkyl), -S02(C C4)alkyl, - S02NH2, -S02NH(CrC4)alkyl, and -S02N((Ci-C4)alkyl)((C 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 (C1-C4)alkyl, (C1-C4)haloalkyl, amino, (C1-C4)alkylamino, ((C1-C4)alkyl)((C1-C4)alkyl)amino, hydroxyl, oxo, (C1-C4)alkoxy, and (Ci-C4)alkoxy(Ci-C4)alkyl, wherein said ring is optionally fused to a (C3-C8)cycloalkyl, heterocycloalkyi, 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, heterocycloalkyi, 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 imgf000012_0001
wherein in (1 ),
A is O, NH, or S; B is CH or N, and C is h drogen or CrC8 alkyl; or
Figure imgf000012_0002
wherein in (2),
D is N or C optionally substituted b hydrogen or C-|-C8 alkyl; or
Figure imgf000012_0003
wherein in (3),
E is NH or CH2; F is O or CO; and G is NH or CH2; or
Figure imgf000012_0004
wherein in (4),
J is O, S or CO; or
Figure imgf000013_0001
wherein in (5),
Q is CH or N;
M is CH or N; and
L/(5) is hydrogen, halo, amino, cyano, (CrC8)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 (d-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-Ce)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 imgf000013_0002
wherein in 6,
L/(6) is NH or CH2; or
Figure imgf000013_0003
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 (CrC6)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 imgf000014_0001
wherein in (8),
P is CH2, NH, O, or S; 0/(8) is CH or N; and n is 0-2; or
Figure imgf000014_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, (d-C8)alkyl, (C3- C8)cycloalkyl, -CORa, -C02Ra, -CONRaRb, -S02Ra, -S02NRaRb, -NRaRb, - NRaC(0)Rb,-NRaS02Rb, -NRaS02NRaRb, -NRaNRaRb, -NRaNRaC(0)Rb, , -ORa, 4-(1 H- pyrazol-4-yl),
wherein any (Ci-C8)alkyl, (C3-C8)cycloalkyl, group is optionally substituted by 1 , 2 or 3 groups independently selected from (C1-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 and Rb are defined as above.
Subgroup (l)(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, (1 - methylethyl)cyclopropyl, 1 , 1-dioxo-tetrahydrothiophene-3-yl, 1-Me-piperidin-4-yl, tetrahydrofuran-3-yl, tetrahydropyran-4-yl, /V,/V-dimethyl-1-propanaminyl, benzyl, or 4- pyridyl;
R3 is H, methyl, or Br; and R6 is methyl, bis(1 , 1-dimethylethyl), bis(l -methylethyl), cyclopropyl, propyl, dimethylamino, ethylamino, (2-hydroxyethyl)amino, 2-propen-1 -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-1 -yl, 4-pyridinylethynyl, phenylethynyl, 2-furanyl, 3-thienyl; 1 H-pyrazol-4-yl, 1 H-indazol-5-yl, 1 H-indazol-6-yl, 3- methyl-1 H-indazol-5-yl, 1 H-1 ,2,3-benzotriazol-5-yl, 2-oxo-2,3-dihydro-1 H-benzimidazol-5- yl, 2-0X0-2, 3-dihydro-1 H-indol-5-yl, 2-oxo-2,3-dihydro-1 H-indol-6-yl, 2,1 ,3-benzoxadiazol- 5-yl, 2-amino-6-quinazolinyl, 2,4-dioxo-1 ,2,3,4-tetrahydro-5-pyrimidinyl, 2-amino-5- pyrimidinyl, 7-oxo-1 ,5,6,7-tetrahydro-1 ,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-(1 H-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. The present invention also relates to a method of treating T cell mediated inflammatory immune diseases or T cell mediated hypersensitivity diseases, which comprises administering to a human in need thereof an effective amount of a compound, which is selected from the group comprising:
6-chloro-N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)- 1 H-pyrazolo[3,4-b]pyridine-4-carboxamide;
6-chloro-/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-3-methyl-1 -(1 - methylethyl)-1 H-pyrazolo[3,4-i)]pyridine-4-carboxamide;
6-chloro-1-(1 , 1-dimethylethyl)-/\/-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3- pyridinyl)methyl]-3-methyl-1 H-pyrazolo[3,4-i)]pyridine-4-carboxamide;
A/-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-6-(4- pyridinyloxy)-1 H-pyrazolo[3,4-ib]pyridine-4-carboxamide; /V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-6-(2- propen-1-ylamino)-1 /-/-pyrazolo[3,4-6]pyridine-4-carboxamide;
6-amino-/\/-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)- 1 /-/-pyrazolo[3,4-6]pyridine-4-carboxamide;
6-cyclopropyl-1 -(1 -methylethyl)-/\/-[(4-methyl-2-oxo-6-propyl-1 ,2-dihydro-3- pyridinyl)methyl]-1 /-/-pyrazolo[3,4-i)]pyridine-4-carboxamide;
6-cyclopropyl-1 -(1 -methylethyl)-/\/-{[6-methyl-4-(1-methylethyl)-2-oxo-1 ,2-dihydro- 3-pyridinyl]methyl}-1 /-/-pyrazolo[3,4-i)]pyridine-4-carboxamide;
1-(1-methylethyl)-/V-[(4-methyl-2-oxo-6-propyl-1 ,2-dihydro-3-pyridinyl)methyl]-6-(4- pyridinyl)-1 /-/-pyrazolo[3,4-6]pyridine-4-carboxamide;
/V-[(6-ethyl-4-methyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1 -methylethyl)-6-(4- pyridinyl)-1 /-/-pyrazolo[3,4-6]pyridine-4-carboxamide;
1-(1-methylethyl)-/V-[(6-methyl-2-oxo-4-propyl-1 ,2-dihydro-3-pyridinyl)methyl]-6-(4- pyridinyl)-1 /-/-pyrazolo[3,4-6]pyridine-4-carboxamide;
1-(1-methylethyl)-/V-{[6-methyl-4-(1 -methylethyl)-2-oxo-1 ,2-dihydro-3- pyridinyl]methyl}-6-(4-pyridinyl)-1 /-/-pyrazolo[3,4-i)]pyridine-4-carboxamide;
6-cyclopropyl-1 -(1 -methylethyl)-/\/-[(6-methyl-2-oxo-4-propyl-1 ,2-dihydro-3- pyridinyl)methyl]-1 /-/-pyrazolo[3,4-i)]pyridine-4-carboxamide;
1-(1-methylethyl)-/V-[(6-methyl-2-oxo-4-phenyl-1 ,2-dihydro-3-pyridinyl)methyl]-6-(4- pyridinyl)-1 /-/-pyrazolo[3,4-6]pyridine-4-carboxamide;
A/-[(4-cyclohexyl-6-methyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)- 6-(4-pyridinyl)-1 /-/-pyrazolo[3,4-6]pyridine-4-carboxamide;
A/-[(4-cyclohexyl-6-methyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-6-cyclopropyl-1 -(1 - methylethyl)-1 /-/-pyrazolo[3,4-i)]pyridine-4-carboxamide;
6-cyclopropyl-1 -(1 -methylethyl)-/\/-[(6-methyl-2-oxo-4-phenyl-1 ,2-dihydro-3- pyridinyl)methyl]-1 /-/-pyrazolo[3,4-i)]pyridine-4-carboxamide;
A/-[(4-cyclopropyl-6-methyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)- 6-(4-pyridinyl)-1 /-/-pyrazolo[3,4-6]pyridine-4-carboxamide;
6-cyclopropyl-/\/-[(6-cyclopropyl-4-methyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 - (1 -methylethyl)-1 H-pyrazolo[3,4-i)]pyridine-4-carboxamide;
/V-[(5-fluoro-4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-6- (4-pyridinyl)-1 /-/-pyrazolo[3,4-6]pyridine-4-carboxamide;
/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-6-(ethylamino)-1-(1- methylethyl)-1 /-/-pyrazolo[3,4-i)]pyridine-4-carboxamide;
N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-1 H- pyrazolo[3,4-b]pyridine-4-carboxamide; 6-cyclopropyl-1-(1-methylethyl)-N-{[6-methyl-2-oxo-4-(trifluoromethyl)-1 ,2-dihydro- 3-pyridinyl]methyl}-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide;
6-(dimethylamino)-N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1- methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide;
N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-6-(1- piperidinyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide;
N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-6-(4- morpholinyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide;
/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-3-methyl-1-(1-methylethyl)- 6-[6-(4-methyl-1 -piperazinyl)-3-pyridinyl]-1 H-pyrazolo[3,4-i)]pyridine-4-carboxamide;
N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-6-(1- piperazinyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide;
N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-6-(3-hydroxy-3-methyl-1- butyn-1 -yl)-1-(1-methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide;
N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-6-(3- methyl-1 H-indazol-5-yl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide;
N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-6- (phenylethynyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide;
/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-6-(2- phenylethyl)-1 H-pyrazolo[3,4-i)]pyridine-4-carboxamide;
N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-6-(4- pyridinylethynyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide;
N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-6- (phenylamino)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide;
N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-6- [(phenylmethyl)amino]-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide;
/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-6-methyl-1-(1-methylethyl)- 1 /-/-pyrazolo[3,4-6]pyridine-4-carboxamide;
A/-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 ,6-bis(1 -methylethyl)-1 H- pyrazolo[3,4-6]pyridine-4-carboxamide;
/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-6-phenyl- 1 /-/-pyrazolo[3,4-6]pyridine-4-carboxamide;
A/-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-6-(4-fluorophenyl)-1 -(1 - methylethyl)-1 /-/-pyrazolo[3,4-i)]pyridine-4-carboxamide;
6-{4-[(dimethylamino)sulfonyl]phenyl}-/\/-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3- pyridinyl)methyl]-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-i)]pyridine-4-carboxamide; 6-[6-(dimethylamino)-3-pyridinyl]-/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3- pyridinyl)methyl]-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-i)]pyridine-4-carboxamide;
/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-6-{4- [(methylamino)sulfonyl]phenyl}-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-i)]pyridine-4- carboxamide;
6-(4-aminophenyl)-/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1- methylethyl)-1 /-/-pyrazolo[3,4-i)]pyridine-4-carboxamide;
6-[4-(acetylamino)phenyl]-/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 - (1 -methylethyl)-1 H-pyrazolo[3,4-i)]pyridine-4-carboxamide;
6-(3-aminophenyl)-/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1- methylethyl)-1 /-/-pyrazolo[3,4-i)]pyridine-4-carboxamide;
/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-6-{4- [(methylamino)carbonyl]phenyl}-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-i)]pyridine-4- carboxamide;
6-[3-(acetylamino)phenyl]-/\/-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -
(1 -methylethyl)-1 H-pyrazolo[3,4-i)]pyridine-4-carboxamide;
A/-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-6-(2,4-dioxo-1 ,2,3,4- tetrahydro-5-pyrimidinyl)-1-(1-methylethyl)-1 H-pyrazolo[3,4-6]pyridine-4-carboxamide;
6-(2-amino-4-pyridinyl)-/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1 - methylethyl)-1 H-pyrazolo[3,4-i)]pyridine-4-carboxamide;
6-[4-(aminosulfonyl)phenyl]-/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]- 1-(1-methylethyl)-1 /-/-pyrazolo[3,4-i)]pyridine-4-carboxamide;
/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-6-{4- [(methylsulfonyl)amino]phenyl}-1 /-/-pyrazolo[3,4-i)]pyridine-4-carboxamide;
6-{4-[(dimethylamino)sulfonyl]phenyl}-/\/-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3- pyridinyl)methyl]-3-methyl-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-i)]pyridine-4-carboxamide;
/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-6-(4- piperidinylamino)-1 /-/-pyrazolo[3,4-i)]pyridine-4-carboxamide;
/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-6- (tetrahydro-2H-pyran-4-ylamino)-1 /-/-pyrazolo[3,4-i)]pyridine-4-carboxamide;
/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-6-[(4- pyridinylmethyl)amino]-1 /-/-pyrazolo[3,4-i)]pyridine-4-carboxamide;
6-{[2-(dimethylamino)ethyl]amino}-/\/-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3- pyridinyl)methyl]-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-i)]pyridine-4-carboxamide;
A/-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-6-[(2-hydroxyethyl)amino]-1 -
(1 -methylethyl)-1 H-pyrazolo[3,4-i)]pyridine-4-carboxamide; N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-6-[5- (methyloxy)-3-pyridinyl]-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide;
/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-6-[2- (methyloxy)-4-pyridinyl]-1 /-/-pyrazolo[3,4-i)]pyridine-4-carboxamide;
6-(6-amino-3-pyridinyl)-/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1 - methylethyl)-1 /-/-pyrazolo[3,4-i)]pyridine-4-carboxamide;
/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-6-[5- (methylsulfonyl)-3-pyridinyl]-1 /-/-pyrazolo[3,4-i)]pyridine-4-carboxamide;
A/-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-6-(2-furanyl)-1 -(1 - methylethyl)-1 H-pyrazolo[3,4-i)]pyridine-4-carboxamide;
/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-6-{6- [(methylamino)carbonyl]-3-pyridinyl}-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-i)]pyridine-4- carboxamide;
6-[5-[(cyclopropylsulfonyl)amino]-6-(methyloxy)-3-pyridinyl]-N-[(4,6-dimethyl-2-oxo- 1 ,2-dihydro-3-pyridinyl)methyl]-1-(1 -methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4- carboxamide;
N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-6-{5- [(phenylsulfonyl)amino]-3-pyridinyl}-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide;
N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-6-[2-(4- morpholinyl)-4-pyridinyl]-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide;
N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-6-[6-(4- morpholinyl)-3-pyridinyl]-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide;
N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-6-[6- (methyloxy)-3-pyridinyl]-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide;
6-[6-(acetylamino)-3-pyridinyl]-N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3- pyridinyl)methyl]-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide;
/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-6-(2- methylphenyl)-1 /-/-pyrazolo[3,4-i)]pyridine-4-carboxamide;
/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-6-(1 H- pyrazol-4-yl)-1 /-/-pyrazolo[3,4-6]pyridine-4-carboxamide;
6-(2-amino-5-pyrimidinyl)-/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1^ methylethyl)-1 /-/-pyrazolo[3,4-i)]pyridine-4-carboxamide;
/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-6-(3-pyridinyl)-1 H^ pyrazolo[3,4-6]pyridine-4-carboxamide;
A/-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-6-(1 H-indazol-5-yl)-1 -(1 - methylethyl)-1 /-/-pyrazolo[3,4-i)]pyridine-4-carboxamide; A/-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-6-(1 H-indazol-6-yl)-1 -(1 - methylethyl)-1 /-/-pyrazolo[3,4-i)]pyridine-4-carboxamide;
6-(1 H-1 ,2,3-benzotriazol-5-yl)-/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 - methylethyl)-1 /-/-pyrazolo[3,4-i)]pyridine-4-carboxamide;
/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-3-methyl-1-(1-methylethyl)-6-(2- oxo-2,3-dihydro-1 H-benzimidazol-5-yl)-1 /-/-pyrazolo[3,4-i)]pyridine-4-carboxamide;
/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-6-(2-oxo- 2,3-dihydro-1 /-/-indol-6-yl)-1 /-/-pyrazolo[3,4-6]pyridine-4-carboxamide;
/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-6-(2-oxo- 2,3-dihydro-1 H-indol-5-yl)-1 /-/-pyrazolo[3,4-6]pyridine-4-carboxamide;
/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-6-(2-oxo-2,3- dihydro-1 /-/-benzimidazol-5-yl)-1 /-/-pyrazolo[3,4-i)]pyridine-4-carboxamide;
/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-6-[6-(4-methyl-1 - piperazinyl)-3-pyridinyl]-1 /-/-pyrazolo[3,4-6]pyridine-4-carboxamide;
/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-6-[6-(4- methyl-1 -piperazinyl)-3-pyridinyl]-1 /-/-pyrazolo[3,4-i)]pyridine-4-carboxamide;
/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-3-methyl-1-(1-methylethyl)- 6-[6-(4-morpholinyl)-3-pyridinyl]-1 /-/-pyrazolo[3,4-i)]pyridine-4-carboxamide;
/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-6-(4- pyridinylamino)-1 /-/-pyrazolo[3,4-6]pyridine-4-carboxamide;
/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-6-[(2-oxo-2,3- dihydro-1 H-benzimidazol-5-yl)amino]-1 /-/-pyrazolo[3,4-i)]pyridine-4-carboxamide;
6-{[4-(aminocarbonyl)phenyl]amino}-/\/-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3- pyridinyl)methyl]-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-i)]pyridine-4-carboxamide;
1-(1 -dimethylethyl)-/V-[(4,6-dime ^
methyl-6-[6-(4-methyl-1-piperazinyl)-3-pyridinyl]-1 /-/-pyrazolo[3,4-i)]pyridine-4- carboxamide;
1-(1 -dimethylethyl)-/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-3-methyl-6-(2- oxo-2,3-dihydro-1 H-benzimidazol-5-yl)-1 /-/-pyrazolo[3,4-i)]pyridine-4-carboxamide;
1-(1 -dimethylethyl)-/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-3-methyl-6-[6-
(4-morpholinyl)-3-pyridinyl]-1 /-/-pyrazolo[3,4-i)]pyridine-4-carboxamide;
6-(2, 1 ,3-benzoxadiazol-5-yl)-/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-3- methyl-1 -(1-methylethyl)-1 /-/-pyrazolo[3,4-i)]pyridine-4-carboxamide;
6-(2-amino-6-quinazolinyl)-/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-3- methyl-1 -(1-methylethyl)-1 /-/-pyrazolo[3,4-i)]pyridine-4-carboxamide; /V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-3-methyl-6-{4- [(methylamino)sulfonyl]phenyl}-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-i)]pyridine-4- carboxamide;
6-[4-(acetylamino)phenyl]-/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-3- methyl-1 -(1-methylethyl)-1 /-/-pyrazolo[3,4-i)]pyridine-4-carboxamide;
6-[4-(aminocarbonyl)phenyl]-/\/-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3- pyridinyl)methyl]-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-i)]pyridine-4-carboxamide;
/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-6-{[2-(2- pyridinylamino)ethyl]amino}-1 /-/-pyrazolo[3,4-i)]pyridine-4-carboxamide;
/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-6-(7-oxo- 1 ,5,6,7-tetrahydro-1 ,8-naphthyridin-3-yl)-1 /-/-pyrazolo[3,4-6]pyridine-4-carboxamide;
/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-6-{6- [(methylamino)sulfonyl]-3-pyridinyl}-1-(1 -methylethyl)-1 /-/-pyrazolo[3,4-i)]pyridine-4- carboxamide;
1-(2-aminoethyl)-6-cyclopropyl-/\/-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3- pyridinyl)methyl]-1 /-/-pyrazolo[3,4-i)]pyridine-4-carboxamide;
6-cyclopropyl-/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-{2- [(methylsulfonyl)amino]ethyl}-1 /-/-pyrazolo[3,4-i)]pyridine-4-carboxamide;
/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-6-methyl-1-(phenylmethyl)- 1 H-pyrazolo[3,4-ib]pyridine-4-carboxamide;
6-cyclopropyl-/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-ethyl-1 /-/- pyrazolo[3,4-6]pyridine-4-carboxamide;
6-cyclopropyl-/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-propyl-1 /-/- pyrazolo[3,4-6]pyridine-4-carboxamide;
6-cyclopropyl-/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-3-methyl-1-(1 - methylethyl)-1 /-/-pyrazolo[3,4-i)]pyridine-4-carboxamide;
1-amino-6-cyclopropyl-/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 /-/- pyrazolo[3,4-6]pyridine-4-carboxamide;
1-cyclobutyl-6-cyclopropyl-/\/-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]- 3-methyl-1 /-/-pyrazolo[3,4-i)]pyridine-4-carboxamide;
1-cyclopentyl-6-cyclopropyl-/\/-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]- 3-methyl-1 /-/-pyrazolo[3,4-i)]pyridine-4-carboxamide;
6-cyclopropyl-1 -(1 ,1 -dimethylethyl)-/\/-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3- pyridinyl)methyl]-3-methyl-1 /-/-pyrazolo[3,4-i)]pyridine-4-carboxamide;
6-cyclopropyl-/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(4- pyridinyl)-1 /-/-pyrazolo[3,4-6]pyridine-4-carboxamide; 6-cyclopropyl-1 -(1 -cyclopropylethyl)-/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3- pyridinyl)methyl]-3-methyl-1 /-/-pyrazolo[3,4-i)]pyridine-4-carboxamide;
1-cyclohexyl-6-cyclopropyl-/\/-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-
3- methyl-1 /-/-pyrazolo[3,4-i)]pyridine-4-carboxamide;
6-cyclopropyl-/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-3-methyl-1-(1- methyl-4-piperidinyl)-1 /-/-pyrazolo[3,4-i)]pyridine-4-carboxamide;
6-cyclopropyl-/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-3-methyl-1- (tetrahydro-2/-/-pyran-4-yl)-1 H-pyrazolo[3,4-i)]pyridine-4-carboxamide;
/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-6-[4- (methyloxy)phenyl]-1 /-/-pyrazolo[3,4-i)]pyridine-4-carboxamide;
/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-6-(4-pyridinyl)^ pyrazolo[3,4-6]pyridine-4-carboxamide;
6-(4-chlorophenyl)-/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1- methylethyl)-1 /-/-pyrazolo[3,4-i)]pyridine-4-carboxamide;
/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-6-(3- thienyl)-1 /-/-pyrazolo[3,4-i)]pyridine-4-carboxamide;
/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-3-methyl-1-(1-methylethyl)-6-(4- pyridinyl)-1 /-/-pyrazolo[3,4-6]pyridine-4-carboxamide;
1 ,6-bis(1 -dimethylethyl)-/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)m
methyl-1 H-pyrazolo[3,4-i)]pyridine-4-carboxamide;
/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-6-[4- (methylsulfonyl)phenyl]-1 /-/-pyrazolo[3,4-i)]pyridine-4-carboxamide;
/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-6-[4-(1 /-/-pyrazol-
4- yl)phenyl]-1 /-/-pyrazolo[3,4-6]pyridine-4-carboxamide;
1-(1 -dimethylethyl)-/V-[(4,6-dime ^
methyl-6-[4-(methyloxy)phenyl]-1 /-/-pyrazolo[3,4-i)]pyridine-4-carboxamide;
1-(1 -dimethylethyl)-/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-3-methyl-6-(3- pyridinyl)-1 /-/-pyrazolo[3,4-6]pyridine-4-carboxamide;
3-bromo-/\/-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)- 6-phenyl-1 /-/-pyrazolo[3,4-6]pyridine-4-carboxamide;
/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-6-(2- nitrophenyl)-1 H-pyrazolo[3,4-i)]pyridine-4-carboxamide;
/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-6-propyl-1 /-/- pyrazolo[3,4-6]pyridine-4-carboxamide;
N-((4,6-dimethyl-2-oxo-1 ,2-dihydropyridin-3-yl)methyl)-1-isopropyl-3-methyl-6- (pyridin-3-yl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide; N-((4,6-dimethyl-2-oxo-1 ,2-dihydropyridin-3-yl)methyl)-1-isopropyl-3-methyl-6- phenyl-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide; N-((4,6-dimethyl-2-oxo-1 ,2-dihydropyridin-3-yl)methyl)-6-(4-
((dimethylamino)methyl)phenyl)-1 -isopropyl-3-methyl-1 H-pyrazolo[3,4-b]pyridine-4- carboxamide;
N-((4,6-dimethyl-2-oxo-1 ,2-dihydropyridin-3-yl)methyl)-6-(4-fluorophenyl)-1 - isopropyl-3-methyl-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide;
1-isopropyl-N-((6-methyl-2-oxo-4-propyl-1 ,2-dihydropyridin-3-yl)methyl)-6-phenyl- 1 H-pyrazolo[3,4-b]pyridine-4-carboxamide;
1-cyclopropyl-/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-3-methyl-6-(2- methyl-5-pyrimidinyl)-1 H-pyrazolo[3,4-i)]pyridine-4-carboxamide;
1-cyclopropyl-/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-3-methyl-6-[4- (methyloxy)phenyl]-1 /-/-pyrazolo[3,4-i)]pyridine-4-carboxamide; 1-cyclopropyl-/V-[(41 -cyclopropyl-/\/-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3- pyridinyl)methyl]-3-methyl-6-[6-(4-methyl-1-piperazinyl)-3-pyridinyl]-1 /-/-pyrazolo[3,4- 6]pyridine-4-carboxamide;
1-cyclopropyl-/V-[(41 -cyclopropyl-/\/-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3- pyridinyl)methyl]-3-methyl-6-[6-(4-morpholiny
carboxamide;
6-cyclopropyl-1 -(1 ,1 -dimethylethyl)-3-methyl-/\/-[(6-methyl-2-oxo-4-propyl-1 ,2- dihydro-3-pyridinyl)methyl]-1 /-/-pyrazolo[3,4-i)]pyridine-4-carboxamide;
1 ,6-dicyclopropyl-3-methyl-/\/-[(6-methyl-2-oxo-4-propyl-1 ,2-dihydro-3- pyridinyl)methyl]-1 /-/-pyrazolo[3,4-i)]pyridine-4-carboxamide;
1 ,6-dicyclopropyl-/V-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-3-methyl- 1 H-pyrazolo[3,4-ib]pyridine-4-carboxamide; 6-(cyclopropylamino)-N-((4,6-dimethyL
isopropyl-3-methyl-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide; N-((4-benzyl-6-methyl-2-oxo-1 ,2-dihydropyridin-3-yl)methyl)-1 -isopropyl-1 H- pyrazolo[3,4-b]pyridine-4-carboxamide;
N-((4-benzyl-6-methyl-2-oxo-1 ,2-dihydropyridin-3-yl)methyl)-1 -isopropyl-3-methyl- 1 H-pyrazolo[3,4-b]pyridine-4-carboxamide;
6-cyclopropyl-/V-[(4-cyclopropyl-6-methyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-3- methyl-1 -(1-methylethyl)-1 /-/-pyrazolo[3,4-i)]pyridine-4-carboxamide;
6-cyclopropyl-3-methyl-1-(1-methylethyl)-/V-{[6-meth^
dihydro-3-pyridinyl]methyl}-1 H-pyrazolo[3,4-i)]pyridine-4-carboxamide ;
6-cyclopropyl-3-methyl-1-(1-methylethyl)-/V-[(6-methyl-2-oxo-4-propyl-1 ,2-dihydro- 3-pyridinyl)methyl]-1 H-pyrazolo[3,4-ib]pyridine-4-carboxamide ; 6-cyclopropyl-3-methyl-1-(1-methylethyl)-/V-{[6-meth^
dihydro-3-pyridinyl]methyl}-1 /-/-pyrazolo[3,4-i)]pyridine-4-carboxamide; or a pharmaceutically acceptable salt thereof. As used herein, the term "optionally" means that the subsequently described event(s) may or may not occur, and includes both event(s) that occur and event(s) that do not occur.
As used herein, unless otherwise defined, the phrase "optionally substituted" or variations thereof denote an optional substitution, including multiple degrees of substitution, with one or more substitutent group. The phrase should not be interpreted as duplicative of the substitutions herein described and depicted. Exemplary optional substituent groups include acyl, CrC6alkyl, d-C3alkylsulfonyl, CrC3alkoxy, C
C3alkoxycarbonyl, cyano, halogen, haloalkyl, hydroxyl, oxo, and nitro.
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(C-|.C8alkyl) including -SCH3, -
SCH2CH3 and the like per the definition of alkyl above.
The term "acyloxy" means -OC(0)Ci.C8alkyl 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- CecycloalkyI" refers to a non-aromatic cyclic hydrocarbon ring having from three to eight carbon atoms. Exemplary "C3-C8cycloalkyl" groups useful in the present invention include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and cyclooctyl.
The term "C5-C8cycloalkenyl" 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 "C3-C8heterocycloalkyl" 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.
As used herein, the term "aryl", unless otherwise defined, is meant aromatic, hydrocarbon, ring system. The ring system may be monocyclic or fused polycyclic (e.g., bicyclic, tricyclic, etc.), substituted or unsubstituted. In various embodiments, the monocyclic aryl ring is C5-C10, or C5-C7, or C5-C6, where these carbon numbers refer to the number of carbon atoms that form the ring system. A C6 ring system, i.e. a phenyl ring, is a suitable aryl group. In various embodiments, the polycyclic ring is a bicyclic aryl group, where suitable bicyclic aryl groups are C8-C12, or C9-C10. A naphthyl ring, which has 10 carbon atoms, is a suitable polycyclic aryl group. Suitable substituents for aryl, unless otherwise defined, are described below in the definition of "optionally substituted".
As used herein, the term "heteroaryl", unless otherwise defined, is meant an aromatic ring system containing carbon(s) and at least one heteroatom. Heteroaryl may be monocyclic or polycyclic, substituted or unsubstituted. A monocyclic heteroaryl group may have 1 to 4 heteroatoms in the ring, while a polycyclic heteroaryl may contain 1 to 10 hetero atoms. A polycyclic heteroaryl ring may contain fused, spiro or bridged ring junctions, for example, bicyclic heteroaryl is a polycyclic heteroaryl. Bicyclic heteroaryl rings may contain from 8 to 12 member atoms. Monocyclic heteroaryl rings may contain from 5 to 8 member atoms (carbons and heteroatoms). Exemplary heteroaryl groups include benzofuran, benzothiophene, furan, imidazole, indole, isothiazole, oxazole, pyrazine, pyrazole, pyridazine, pyridine, pyrimidine, pyrrole, quinoline, quinazoline, quinoxaline, thiazole, and thiophene. Suitable substituents for heteroaryl, unless otherwise defined are described below in the definition of "optionally substituted"
Provided herein are methods of treatment or prevention of autoimmune and inflammatory conditions and diseases that can be improved by inhibiting EZH1 and / or EZH2 and thereby, e.g., modulate the level of expression of methylation activated and methylation repressed target genes, or modulate the activity of signalling proteins. A method may comprise administering to a human, e.g. a human in need thereof, a therapeutically effective amount of an agent described herein.
Thus in one aspect there is provided the use of use of a compound or a pharmaceutically acceptable salt thereof which inhibits EZH2 and/or EZH1 (i.e. an EZH2 and/or EZH1 inhibitor) in the manufacture of a medicament for treating T cell mediated inflammatory immune diseases or T cell mediated hypersensitivity diseases.
In a further aspect there is provided a method of treatment of T cell mediated inflammatory immune diseases or T cell mediated hypersensitivity diseases in a human comprising administering a therapeutically effective amount of an EZH1 and / EZH2 inhibitor.
Inflammation represents a group of vascular, cellular and neurological responses to trauma. Inflammation can be characterised as the movement of inflammatory cells such as monocytes, neutrophils and granulocytes into the tissues. This is usually associated with reduced endothelial barrier function and oedema into the tissues. Inflammation can be classified as either acute or chronic. Acute inflammation is the initial response of the body to harmful stimuli and is achieved by the increased movement of plasma and leukocytes from the blood into the injured tissues. A cascade of biochemical event propagates and matures the inflammatory response, involving the local vascular system, the immune system, and various cells within the injured tissue. Prolonged inflammation, known as chronic inflammation, leads to a progressive shift in the type of cells which are present at the site of inflammation and is characterised by simultaneous destruction and healing of the tissue from the inflammatory process.
When occurring as part of an immune response to infection or as an acute response to trauma, inflammation can be beneficial and is normally self-limiting. However, inflammation can be detrimental under various conditions. This includes the production of excessive inflammation in response to infectious agents, which can lead to significant organ damage and death (for example, in the setting of sepsis). Moreover, chronic inflammation is generally deleterious and is at the root of numerous chronic diseases, causing severe and irreversible damage to tissues. In such settings, the immune response is often directed against self-tissues (autoimmunity), although chronic responses to foreign entities can also lead to bystander damage to self tissues.
The aim of anti-inflammatory therapy is therefore to reduce this inflammation, to inhibit autoimmunity when present and to allow for the physiological process or healing and tissue repair to progress.
The agents may be used to treat inflammation of any tissue and organs of the body, including musculoskeletal inflammation, vascular inflammation, neural inflammation, digestive system inflammation, ocular inflammation, inflammation of the reproductive system, and other inflammation, as exemplified below.
Musculoskeletal inflammation refers to any inflammatory condition of the musculoskeletal system, particularly those conditions affecting skeletal joints, including joints of the hand, wrist, elbow, shoulder, jaw, spine, neck, hip, knew, ankle, and foot, and conditions affecting tissues connecting muscles to bones such as tendons. Examples of musculoskeletal inflammation which may be treated with compounds of the invention include arthritis (including, for example, osteoarthritis, psoriatic arthritis, ankylosing spondylitis, acute and chronic infectious arthritis, arthritis associated with gout and pseudogout, and juvenile idiopathic arthritis), tendonitis, synovitis, tenosynovitis, bursitis, fibrositis (fibromyalgia), epicondylitis, myositis, and osteitis (including, for example, Paget's disease, osteitis pubis, and osteitis fibrosa cystic).
Ocular inflammation refers to inflammation of any structure of the eye, including the eye lids. Examples of ocular inflammation which may be treated in this invention include blepharitis, blepharochalasis, conjunctivitis, dacryoadenitis, keratitis, keratoconjunctivitis sicca (dry eye), scleritis, trichiasis, and uveitis.
Examples of inflammation of the nervous system which may be treated in this invention include encephalitis, Guillain-Barre syndrome, meningitis, neuromyotonia, narcolepsy, multiple sclerosis, myelitis and schizophrenia. Examples of inflammation of the vasculature or lymphatic system which may be treated in this invention include arthrosclerosis, arthritis, phlebitis, vasculitis, and lymphangitis. Examples of inflammatory conditions of the digestive system which may be treated in this invention include cholangitis, cholecystitis, enteritis, enterocolitis, gastritis, gastroenteritis, ileitis, and proctitis.
Examples of inflammatory conditions of the reproductive system which may be treated in this invention include cervicitis, chorioamnionitis, endometritis, epididymitis, omphalitis, oophoritis, orchitis, salpingitis, tubo-ovarian abscess, urethritis, vaginitis, vulvitis, and vulvodynia.
The agents may be used to treat autoimmune conditions having an inflammatory component. Such conditions include acute disseminated alopecia universalise, Behcet's disease, Chagas' disease, chronic fatigue syndrome, dysautonomia, encephalomyelitis, ankylosing spondylitis, aplastic anemia, hidradenitis suppurativa, autoimmune hepatitis, autoimmune oophoritis, celiac disease, Crohn's disease, diabetes mellitus type 1 , giant cell arteritis, goodpasture's syndrome, Grave's disease, Guillain-Barre syndrome, Hashimoto's disease, Henoch-Schonlein purpura, Kawasaki's disease, lupus erythematosus, microscopic colitis, microscopic polyarteritis, mixed connective tissue disease, multiple sclerosis, myasthenia gravis, opsoclonus myoclonus syndrome, optic neuritis, ord's thyroiditis, pemphigus, polyarteritis nodosa, polymyalgia, Reiter's syndrome, Sjogren's syndrome, temporal arteritis, Wegener's granulomatosis, warm autoimmune haemolytic anemia, interstitial cystitis, lyme disease, morphea, sarcoidosis, scleroderma, ulcerative colitis, and vitiligo.
The agents may be used to treat T-cell mediated hypersensitivity diseases having an inflammatory component. Such conditions include contact hypersensitivity, contact dermatitis (including that due to poison ivy), uticaria, skin allergies, respiratory allergies (hayfever, allergic rhinitis) and gluten-sensitive enteropathy (Celliac disease).
Other inflammatory conditions which may be treated in this invention include, for example, appendicitis, dermatitis, dermatomyositis, endocarditis, fibrositis, gingivitis, glossitis, hepatitis, hidradenitis suppurativa, iritis, laryngitis, mastitis, myocarditis, nephritis, otitis, pancreatitis, parotitis, percarditis, peritonoitis, pharyngitis, pleuritis, pneumonitis, prostatistis, pyelonephritis, and stomatisi, transplant rejection (involving organs such as kidney, liver, heart, lung, pancreas (e.g., islet cells), bone marrow, cornea, small bowel, skin allografts, skin homografts, and heart valve xengrafts, sewrum sickness, and graft vs host disease), acute pancreatitis, chronic pancreatitis, acute respiratory distress syndrome, Sexary's syndrome, congenital adrenal hyperplasis, nonsuppurative thyroiditis, hypercalcemia associated with cancer, pemphigus, bullous dermatitis herpetiformis, severe erythema multiforme, exfoliative dermatitis, seborrheic dermatitis, seasonal or perennial allergic rhinitis, bronchial asthma, contact dermatitis, astopic dermatitis, drug hypersensistivity reactions, allergic conjunctivitis, keratitis, herpes zoster ophthalmicus, iritis and oiridocyclitis, chorioretinitis, optic neuritis, symptomatic sarcoidosis, fulminating or disseminated pulmonary tuberculosis chemotherapy, idiopathic thrombocytopenic purpura in adults, secondary thrombocytopenia in adults, acquired (autoimmune) haemolytic anemia, leukaemia and lymphomas in adults, acute leukaemia of childhood, regional enteritis, autoimmune vasculitis, multiple sclerosis, chronic obstructive pulmonary disease, solid organ transplant rejection, sepsis.
Preferred treatments include any one of treatment of transplant rejection, psoriatic arthritis, multiple sclerosis, Type 1 diabetes, asthma, systemic lupus erythematosis, chronic pulmonary disease, and inflammation accompanying infectious conditions (e.g., sepsis).
Typically, but not absolutely, the salts of the compounds for use in the inevntion are pharmaceutically acceptable salts. Salts encompassed within the term
"pharmaceutically acceptable salts" refer to non-toxic salts of the compounds of this invention. Salts of the compounds of the present invention may comprise acid addition salts. In general, the salts are formed from pharmaceutically acceptable inorganic and organic acids. More specific examples of suitable acid salts include maleic, hydrochloric, hydrobromic, sulphuric, phosphoric, nitric, perchloric, fumic, acetic, propionic, succinic, glycolic, formic, lactic, aleic, tartaric, citric, palmoic, malonic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicylic, fumaric, toluenesulfonic, methansulfonic (mesylate), naphthalene-2-sulfonic, benzenesulfonic, hydroxynaphthoic, hydroiodic, malic, teroic, tannic, and the like.
Other representative salts include acetate, benzenesulfonate, benzoate, bicarbonate, bisulfate, bitartrate, borate, calcium edetate, camsylate, carbonate, clavulanate, citrate, dihydrochloride, edisylate, estolate, esylate, fumarate, gluceptate, gluconate, glutamate, glycollylarsanilate, hexylresorcinate, hydrobromide, hydrochloride, hydroxynaphthoate, iodide, isethionate, lactate, lactobionate, laurate, malate, maleate, mandelate, mesylate, methylsulfate, monopotassium maleate, mucate, napsylate, nitrate, oxalate, pamoate (embonate), palmitate, pantothenate, phosphate/diphosphate, polygalacturonate, salicylate, stearate, subacetate, succinate, sulfate, tannate, tartrate, teoclate, tosylate, triethiodide, and valerate salts.
Other salts, which are not pharmaceutically acceptable, may be useful in the preparation of compounds for use in this invention. These salts, such as oxalic or trifluoroacetate, while not in themselves pharmaceutically acceptable, may be useful in the preparation of salts useful as intermediates in obtaining the compounds of the invention and their pharmaceutically acceptable salts.
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 non-aqueous 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 for use in this 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 non-aqueous 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 non-aqueous 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 for use in this 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 0.001 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.
Experimentals:
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 (1991 ) 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-lnterscience, 1994).
EXAMPLES
General Experimental Methods
The following abbreviations are used throughout the experimental and have the following meaning:
aq aqueous
BINAP 2,2'-bis(diphenylphosphino) -1 ,1 '-binapthyl
ca. circa
CDCIs-d chloroform-d
CD3OD-C/4 methanol-c/4
Cs2C03 cesium carbonate
CHCI3 chloroform
ACN acetonitrile
CH3CN acetonitrile
Celite® registered trademark of Celite Corp. brand of diatomaceous earth
DBU 1 ,8-diazabicyclo[5.4.0]undeca-7-ene
DCE dichloroethane
DCM methylene chloride
DME 1 ,2 dimethoxyethane
DMF Ν,Ν-dimethyl formamide
DIEA diisopropyl ethylamine
DMSO-d6 dimethylsulfoxide-c/6
EtOAc ethyl acetate
EDC 1-(3-dimethylaminopropyl)-3-ethylcarbodimmide hydrochloride
h hour(s) 1H NMR proton nuclear magnetic resonance
HCI hydrochloric acid
HOAT 1 -hydroxy-7-azabenzotriazole
HPLC high performance liquid chromatography
I PA 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
PdCI2(dppf)-CH2CI2 1 , 1 '-bis(diphenylphosphino)ferrocene-palladium(ll)dichloride dichloromethane complex
Pd(Ph3P)4 tetrakis(triphenylphosphine)palladium(0)
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 (1 Og - 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-10ADvp (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 (C18) 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 * 4.6 mm, 5 /jm) was used. HPLC was done on Agilent-1200 series UPLC system equipped with column Zorbax SB-C18 (50 * 4.6 mm, 1.8 /jm) eluting with CH3CN: ammonium acetate buffer. The reactions were performed in the microwave (CEM, Discover).
1H-NMR spectra were taken in deuterated DMSO (unless otherwise noted) and 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.
The compounds of the present invention were prepared according to the following schemes 1 -4 described in detail below. The groups and substituents shown in the schemes 1 -4, 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.
Figure imgf000038_0001
Scheme 1 illustrates two methods to synthesize a compound of formula (VII). Substituted aminopyrazoles of formula (I) are heated with diethyl oxobutanedione in benzene or toluene at 62 °C overnight. Treatment of the putative intermediate with acetic acid and typically heating at reflux furnishes azaindazole compounds of formula (II). Compounds of formula (II) are converted to compounds of formula (III) by base-catalyzed hydrolysis of the ethyl ester and then chlorination of the putative carboxylic acid intermediate with POCI3 under standard conditions to afford compounds of formula (IV). Treatment of compounds of formula (IV) with substituted aminomethyl pyridones of formula (V) using EDC, HOAT, N-methylmorpholine, and DMSO for a period of no less than 12 h stirring typically at room temperature (in some instances, heating at 40 °C may be required), affords compounds of formula (VI). Compounds of formula (VI) are substituted at the 6-position using standard methods known to those skilled in the art (i.e. nucleophillic substitution, palladium mediated cross couplings), to afford compounds of formula (VII). Alternatively, compounds of formula (VII) can be obtained from a compound of formula (II) via compounds (I la) and (IVa). In this route, compounds of formula (II) are converted to the corresponding triflate (I la) using standard methods. Compounds of formula (I la) are then substituted at the 6-pos. using standard palladium mediated cross- coupling conditions, followed by base-catalyzed hydrolysis of the ethyl ester group to afford compounds of formula (IVa). Treatment of compounds of formula (IVa) with substituted aminomethyl pyridones of formula (V) using EDC, HOAT, N-methylmorpholine, and DMSO at room temperature for a period of no less than 12 h stirring at room temperature affords compounds of formula (VII).
Scheme 2
Figure imgf000039_0001
Compounds of formula (VII) are also prepared as depicted in scheme 2. In this embodiment, substituted oxobutanediones of formula (VIII) (prepared by Claisen condensation between an appropriately substitutued ketone and diethyloxalate) are heated with substituted aminopyrazoles of formula (I) (as described for scheme 1 ), to afford compounds of formula (IX). Compounds of formula (IX) are converted to
compounds of formula (X) by base-catalyzed hydrolysis. Substitutions respectively at either the R3-position, or on the R6 substituted group of compounds of formulas (IX) and (X) are done so using methods known to those skilled in the art (e.g. bromination, nitration). Treatment of compounds of formula (X) with substituted aminomethyl pyridones of formula (V) using EDC, HOAT, N-methylmorpholine, and DMSO at room temperature for a period no less than 12 h stirring at room temperature, affords compounds of formula (VII).
Scheme 3
Figure imgf000040_0001
Compounds of formula (VII) are also prepared as depicted in scheme 3. In this embodiment, substituted aminopyrazoles of formula (I) are heated with a keto-ester of formula (XI) in benzene containing catalytic acetic acid at 62 °C overnight. Exposure of the putative intermediate to refluxing Dowtherm A overnight affords a compound of formula (XII). Compounds of formula (XI) that are not commercially available are prepared using standard methods known to those skilled in the art, and are described herein. Compounds of formula (XII) are converted to compounds of formula (XIII) by bromination with refluxing POBr3 in toluene/DMF for 1 h. Heating of compounds of formula (XIII) with dicyano zinc, tris(dibenzylideneacetone)dipalladium(0), and SPhos in DMFand water at 120° Cfor 2 hours furnishes compounds of formula (XIV). The compounds of formula (XIV) are hydrolyzed to the compounds of formula (X) using standard base- catalyzed hydrolysis conditions.
Compounds of formula (X) are treated with substituted aminomethyl pyridones of formula (V) using EDC, HOAT, N-methylmorpholine, and DMSO at room temperature for a period no less than 12 h stirring at room temperature, to afford compounds of formula (VII). Scheme 4
Figure imgf000041_0001
Scheme 4 illustrates the method to synthesize a compound of formula (V).
Heating of compounds of formula (XV) with cyanoacetamide in ethanol at reflux containing catalytic piperidine for typically 30 min, affords compounds of formula (XVII). Alternatively, treatment of compounds of formula (XVI) 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 (XVII). Regioisomeric mixtures, are separable and the individual compounds regiochemical assignments were confirmed by 2D HNMR techniques. Compounds of formula (XV) and (XVI) which are not commercially available are prepared using standard methods known to those skilled in the art, and are described herein. Compounds of formula (XVII) can be 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 NiCI2-6H20.
Examples
Intermediate 1
6-Hydroxy-1 -(1 -methylethyl)-1H-pyrazolo[3,4-b]pyridine-4-carboxylic acid
Figure imgf000041_0002
Step 1 :
1 -(1 -methylethyl)-1 H-pyrazol-5-amine
Figure imgf000042_0001
To a solution of ethyl (2Z)-2-cyano-3-(ethyloxy)-2-propenoate (1 14.2 g, 0.67 mol) in ethanol (250 ml.) was slowly added isopropylhydrazine (55 g, 0.74 mol) in a dropwise manner. The mixture was heated at reflux for 4 h, and then cooled to room temperature. The mixture was concentrated in vacuo. Approximately half of the crude 5-amino-1- isopropyl-1 H-pyrazole-4-carboxylic acid ethyl ester (50 g) was suspended in an aqueous solution of sodium hydroxide (4M, 130 ml_), and stirred with heating at reflux for 2 h. The reaction mixture was then cooled to room temperature and adjusted to pH=3.5 with concentrated HCI, wherein precipitate formation ensued. The solid was collected by filtration and dried in vacuum oven overnight to afford crude 5-amino-1-isopropyl-1 H- pyrazole-4-carboxylic acid (30 g). The solid was suspended in diphenyl ether (120 ml.) and stirred with heating at 160-165 °C for 2 h. The solution was then cooled to room temperature and the solvent removed in vacuo. The crude product was purified by silica gel chromatography (eluent: petroleum ether/EtOAc=1 :1 ) to afford the product as 12 g. 1H NMR (400 MHz, DMSO-d6) δ ppm 1.24 (d, 6H, J = 6.4 Hz), 4.32 (m, 1 H), 5.06 (s, 2H), 5.21 (s, 1 H), 7.00 (s, 1 H).
Step 2:
To a solution of diethyl 2-oxobutanedione (96 g) in toluene (500 ml.) was added 1-
(1 -methylethyl)-1 H-pyrazol-5-amine (30 g, 0.24 mol) and the mixture was stirred at 60 °C overnight. The mixture was concentrated in vacuo, the crude residue dissolved into acetic acid (500 ml_), and then heated at reflux for 2 h. The mixture was then cooled to room temperature and concentrated in vacuo to give a residue, which was recrystallized from DCM to afford the product as a yellow colored solid, collected as 40 g. This solid was suspended in in ethanol (700 ml.) and THF (100 ml_), followed by addition of 3 M NaOH (150 ml_). The reaction mixture was stirred at 40 °C for 40 min. The mixture was concentrated in vacuo to remove the volatiles, and the aqueous layer then acidified using 1 M HCI. The resulting precipitate was collected by filtration and dried under high vacuum to give the title compound, 6-hydroxy-1-(1-methylethyl)-1 H-pyrazolo[3,4-i)]pyridine-4- carboxylic acid, as 27 g. LCMS E-S (M+H) = 222.0. 1H NMR (400 MHz, DMSO-d6) δ ppm 1.48 (d, J=6.82 Hz, 6 H), 4.89 - 4.96 (m, 1 H), 6.81 (s, 1 H), 8.13 (s, 1 H). Intermediate 2
3-Methyl-1 -(1 -methylethyl)-6-oxo-6,7-dihydro-1 H-pyrazolo[3,4-b]pyridine-4- carboxylic acid
Figure imgf000043_0001
Step 1 :
3-Methyl-1 -(1 -methylethyl)-1 H-pyrazol-5-amine
Figure imgf000043_0002
3-Amino-2-butenenitrile (33.3 g, 0.41 mol) and ethanol (170 mL) were combined and stirred at room temperature for 30 min., after which time isopropylhydrazine (50 g, 0.67 mol) was added at once. After stirring at room temperature for 5 min., the contents were then heated at reflux for 10 h. After cooling to room temperature, the mixture was concentrated in vacuo to give the desired product (85 g) which was used in the next step directly. 1H NMR (400 MHz, DMSO-d6) δ ppm 1 .21 (d, 6 H, J =6.4 Hz), 1.94 (s, 3H), 4.21 (m, 1 H), 4.93 (s, 2H), 5.02 (s, 1 H).
Step 2:
To a solution of diethyl 2-oxobutanedione (176 g, 0.94 mol) in toluene (2 L) was added 3-methyl-1-(1-methylethyl)-1 H-pyrazol-5-amine (82.5 g, 0.59 mol), and the mixture stirred at 62 °C, overnight. After cooling to room temperature, the mixture was
concentrated in vacuo and the crude residue dissolved into acetic acid (1.5 L). The mixture was heated at reflux for 2 h. After cooling to room temperature, the mixture was concentrated in vacuo to afford a solid residue, which was recrystallized from DCM to afford the desired product as a yellow colored solid. The collected solid was suspended in ethanol (1510 mL) and THF (216 mL) followed by addition of 3N NaOH (334 mL) and the reaction mixture was stirred at 40 °C for 40 min. The mixture was concentrated in vacuo to remove the volatiles and the aqueous phase acidified using 1 N HCI. The resulting precipitate was collected by filtration and dried under high vacuum to give the title compound, 3-methyl-1 -(1 -methylethyl)-6-oxo-6,7-dihydro-1 H-pyrazolo[3,4-ib]pyridine-4- carboxylic acid, as 51.38 g. LCMS E-S (M+H) = 236.1. 1H NMR (400 MHz, DMSO-d6) δ ppm 1.35 (d, J=6.8 Hz, 6 H), 2.41 (s, 3H), 4.84-4.91 (m, 1 H), 6.64 (s, 1 H). Carboxylic acid proton not observed.
Intermediate 3
Ethyl 1 -(1 -dimethylethyl)-6-hydroxy-3-methyl-1H-pyrazolo[3,4-b]pyridine-4- carbox late
Figure imgf000044_0001
1-(1 , 1 -Dimethylethyl)-3-methyl-1 H-pyrazol-5-amine (5 g, 32.6 mmol), diethyl 2- oxobutanedione (6.14 g, 32.6 mmol) and toluene (100 ml.) were heated at 70 °C for 16 hours. The solvent was removed in vacuo, the crude residue dissolved in acetic acid (100 ml_), and heated at reflux for 4 hours. The solvent was removed in vacuo, and the crude product purified via silica gel chromatography (eluent: gradient of 0 to 10%
EtOAc/Hexanes). The product was collected as a solid, 6.32 g (70%). LCMS E-S (M+H) = 278.4. 1H NMR (400 MHz, DMSO-d6) δ ppm 1 1.39 (s, 1 H), 6.82 (s, 1 H), 4.36 (q, 2H, J = 7.2 Hz), 2.45 (s, 3H), 1 .69 (s, 9H), 1 .32 (t, 3H, J = 7.2 Hz).
Intermediate 4
1 -(1 ,1 -Dimethylethyl)-6-hydroxy-3-methyl-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid
Figure imgf000044_0002
Sodium hydroxide (52.6 ml_, 52.6 mmol) was added to an EtOH solution (100 ml.) of ethyl 1 -(1 , 1-dimethylethyl)-3-methyl-6-oxo-6,7-dihydro-1 H-pyrazolo[3,4-b]pyridine-4- carboxylate (7.3 g, 26.3 mmol) and stirred at room temperature for 16 h. The solvent was removed in vacuo. The crude residue was suspended in water, and extracted with EtOAc. The combined organic layers were washed with water, dried over MgS04, filtered, and concentrated in vacuo. The solid product obtained was set aside. The aqueous phase was concentrated in vacuo and the crude product purified by reverse phase HPLC (mobile phase : 20 -50% ACN/H20, 0.1 %TFA) to afford additional product. The combined products were collected as a solid, 5.76 g (88%). LCMS E-S (M+H) = 250.3. 1H NMR (400 MHz, DMSO-d6) δ ppm 13.5 - 13.9 (br s, 1 H), 1 1 .2 - 1 1.5 (br s, 1 H), 6.78 (s, 1 H), 2.46 (s, 3H), 1.70 (s, 9H).
Intermediate 5
6-Chloro-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid
Figure imgf000045_0001
To a 75 ml. pressure vessel was added 1 -(1 -methylethyl)-6-oxo-6,7-dihydro-1 H- pyrazolo[3,4-b]pyridine-4-carboxylic acid (4.12 g, 18.62 mmol), followed by phosphorous oxychloride (26.0 ml, 279 mmol). The flask was sealed and the stirring mixture heated at ca. 105 °C for ca. 18 h. After cooling to room temperature, the contents were
concentrated in vacuo to remove most of volatiles. The residual contents were poured into a mixture of ice and 3M NaOH (60 ml_), followed by additional 3M NaOH to ensure the pH stayed basic. The mixture was stirred for 30 min., and then cooled in an ice bath. The heterogenous mixture was slowly acidified to pH = 3-4 with 6M HCI. The resulting suspension was extracted with EtOAc (3x). The combined organic layers were dried over MgS04, filtered, and concentrated in vacuo to afford a yellow solid which was dried in a hi- vac oven overnight. The title compound was collected as 4.1 1 g (90%), and used without further purification. LCMS E-S (M+H) = 240.2/242.2. 1H NMR (400 MHz, DMSO-d6) δ ppm 1.51 (d, J=6.57 Hz, 6 H) 5.17 (quin, J=6.63 Hz, 1 H) 7.65 (s, 1 H) 8.41 (s, 1 H) 14.25 (br. s., 1 H).
Intermediate 6
6-Chloro-3-methyl-1 -(1 -methylethyl)-1H-pyrazolo[3,4-b]pyridine-4-carboxylic acid
Figure imgf000045_0002
3-Methyl-1-(1 -methylethyl)-6-oxo-6,7-dihydro-1 H-pyrazolo[3,4-b]pyridine-4- carboxylic acid (1.5 g, 6.38 mmol) was added to a solution of THF (15 mL) followed by addition of phosphorous oxychloride (8.9 mL, 96 mmol), and the contents heated at 105
°C overnight. After cooling to room temperature, the contents were concentrated in vacuo to remove most of the volatiles. The residual contents were slowly poured into a solution of iced water and 1 N NaOH (10 mL), and the contents were stirred at room temperature for 24 h, during which time solid precipitation ensued. Additional 1 N NaOH was added, upon which the solids went into solution. After stirring at room temperature for an
additional 30 min., the contents were cooled in an ice bath and the mixture slowly acidified to pH = 3-4 by slow addition of 6N HCI, to afford a heterogenous mixture. The mixture was filtered and a white solid set aside. The aq. layer was further extracted with EtOAc and DCM. The combined organic layers dried were dried over MgSC^, filtered and
concentrated in vacuo. The resultant light brown solid was triturated in EtOAc/EtOH (1 :1 ) and filtered to afford a first crop of white solid product, which was set aside. The filtrate was again concentrated in vacuo. The residue was diluted with EtOAc, sonicated, treated with hexanes, and filtered. The process was repeated. The isolated solid product crops were dried under vacuum (3 h) and collected as 1 .33 g (80%). LCMS E-S (M+H) : 254.3
1H NMR (400 MHz, DMSO-d6) δ ppm 1 .46 (d, 6 H), 2.60 (s, 3 H), 5.10 (quin, J=6.63 Hz, 1 H), 7.50 (s, 1 H), 14.17 (br. s., 1 H).
Intermediate 7
6-Chloro-1 -(1 -dimethylethyl)-3-methyl-1H-pyrazolo[3,4-b]pyridine-4-carboxylic
acid
Figure imgf000046_0001
1-(1 , 1 -Dimethylethyl)-3-methyl-6-oxo-6,7-dihydro-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid (5.07 g, 20.34 mmol) and phosphorus oxychloride (28.4 ml, 305 mmol) were heated at 100 °C for 16 hours. The contents were concentrated in vacuo. The residue was added to ice water followed by 1 N NaOH until basic (pH > 10). After stirring for 15 minutes, the mixture was adjusted to pH 3-4 by addition of 1 N HCI. The contents were extracted with EtOAc, then washed with water, brine and concentrated in vacuo. The crude product was purified by reverse phase HPLC (40 - 70% ACN/H20, 0.1 %TFA). The product was collected as a solid, 0.50 g (9%). LCMS E-S (M+H) =268.3 1H NMR (400 MHz, DMSO-de) δ ppm 1 .73 (s, 9 H), 2.56 (s, 3 H), 7.48 (s, 1 H), 14.17 (br. s., 1 H). Intermediate 8
nomethyl)-4,6-dim pyridinone hydrochloride
Figure imgf000047_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. HCI, and the formed solids were filtered. The yellow filtrate was concentrated . To the crude compound was added 30 mL of cone. HCI 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.. 1H NMR (400 MHz, DMSO-d6) δ ppm 1 1.85 (br s, 1 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 9
3-(Aminomethyl)-6-methyl-4-(trifluoromethyl)-2(1 H)-pyridinone
Figure imgf000047_0002
To a dried 500 mL Parr bottle equipped with nitrogen inlet were placed sodium acetate (1 .502 g, 18.30 mmol), 10% palladium on carbon (1.579 g, 0.742 mmol), platinum(IV) oxide (0.01 1 g, 0.049 mmol) and a small amount of acetic acid to wet the catalysts under nitrogen stream. Next added 2-hydroxy-6-methyl-4-(trifluoromethyl)-3- pyridinecarbonitrile (2.0g, 9.89 mmol) followed by acetic acid (175 mL) while under nitrogen atmosphere. The contents were sealed, placed on a Parr shaker, and reacted at 40 psi of H2 for ca. 6 hr., keeping the H2 psi between 20 and 40 psi (vessel was refilled twice). The vessel was purged with nitrogen and the reaction mixture filtered through Celite, and the filter pad was further washed with a small amount of acetic acid. The volatiles were removed in vacuo to afford a residue, which was dried under hi-vacuum for 45 min. The solid was suspended in cone. HCI (12 ml_), stirred, and filtered (removed NaCI). The clear filtrate was concentrated in vacuo and the residue dried under hi- vacuum. The collected solid was suspended in cone. HCI (2 ml.) and diluted with EtOH (13 ml_). The contents were agitated (i.e. spatula) and stored at ca. 0 °C (i.e. freezer) for 30 min to give a white solid. The solid was filtered and washed with cold ethanol (5 ml_). The solid was filtered and dried in vacuum oven for 1 h. The final product was collected as 0.95 g (40%). LCMS E-S (M+H) = 206.9. 1H NMR (400 MHz, DMSO-d6) δ ppm 2.31 (s, 3 H), 3.87 (d, J=5.05 Hz, 2 H), 6.41 (s, 1 H), 8.12 - 8.37 (m, 3 H).
Intermediate 10
3-(Aminomethyl)-4-cyclohexyl-6-methyl-2(1 H)-pyridinone
Figure imgf000048_0001
Step 1
To a stirred suspension of CrCI2 (58 g, 472.8 mmol in THF (1500 ml.) was added a THF solution (500 ml.) of 1 , 1-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 HCI. 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-BuOK (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 HCI, which were added slowly. The reaction mixture was filtered, washed with water and dried to give 4-cyclohexyl-6-methyl- 2-OXO-1 ,2-dihydro-3-pyridinecarbonitrile as 4.5 g (32%). 1 H NMR (400 MHz, DMSO-c/6) δ ppm 6.25 (s, 1 H), 2.61 -2.65 (m, 1 H), 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 .3mmol), and \2 (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 3N HCI (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 (TFA salt), 0.5 g (25%). LCMS E-S (M+H) =221.1. 1H NMR (400 MHz, DMSO-d6) δ ppm 11.8 - 11.9 (brs, 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 11
3-(Aminomethyl)-4-cyclopropyl-6-methyl-2(1H)-pyridinone hydrochloride
Figure imgf000049_0001
The title compound was prepared in the same manner as described for
intermediate 10 (step 2) from 4-cyclopropyl-6-methyl-2-oxo-1,2-dihydro-3- pyridinecarbonitrile (5 g, 28.7 mmol). The product was collected as a TFA salt, 0.50 g. LCMS E-S (M+H) = 179.1. 1H NMR (400 MHz, DMSO-d6) δ ppm 11.76 - 11.78 (br s, 1H), 7.82-7.92 (brs, 3H), 5.61 (s, 1H), 3.94-3.99 (m, 2H), 2.11 (s, 3H), 1.98-2.05 (m, 1H), 0.95 - 1.01 (m, 2H), 0.74 - 0.79 (m, 2H).
Intermediate 12
3-(Aminomethyl)-6-methyl-4-propyl-2(1H)-pyridinone
Figure imgf000049_0002
Step 1
To a solution of DMSO (300 ml.) containing t-BuOK (20 g, 178 mmol) and cyanoacetamide (16.5 g, 196 mmol) was added (3£)-3-hepten-2-one (20 g, 178 mmol), and contents stirred at room temperature for 30 min. Additional t-BuOK (60 g, 534 mmol) was added and the reaction mixture was under an atmosphere of oxygen for an additional 1 h. The reaction mixture was purged with argon, diluted with 4 volumes of H20, and then 5 volumes of 4 N HCI, which were added slowly. The reaction mixture was filtered, washed with water, and dried to give the product as 10 g (32%). 1H NMR (400 MHz, DMSO-d6) δ ppm 12.25 - 12.40 (br s, 1 H), 6.18 (s, 1 H), 2.53 (t, 2H), 2.22 (s, 3H), 1.57 - 1.64 (m, 2H), 0.84 (t, 3H).
Step 2
The title compound was prepared in the same manner as described for intermediate 10 (step 2) from the product of step 1 (2 g, 1 1.2 mmol). The product was collected as 1 .2 g (60%). LCMS E-S (M+H) = 181.1. 1H NMR (400 MHz, DMSO-d6) δ ppm 7.85 - 7.95 (br s, 3H), 5.99 (s, 1 H), 3.80 - 3.85 (m, 2H), 2.42 (t, 2H), 2.14 (s, 3H), 1.43 - 1 .49 (m, 2H), 0.86 (t, 3H).
Intermediate 13
nomethyl)-6-methyl-4-phenyl-2(1H)-pyridinone
Figure imgf000050_0001
The title compound was prepared in the same manner as described for intermediate 12 (steps 1 and 2) from (3£)-4-phenyl-3-buten-2-one (20 g, 137 mmol). The crude nitrile intermediate was obtained as 10 g (35%), of which 4 g of this putative intermediate was converted to the title compound 1 .2 g as a TFA salt. LCMS E-S (M+H) = 215.0. 1H NMR (400 MHz, DMSO-d6) δ ppm 12.2 - 12.3 (br s, 1 H), 7.88 - 8.00 (br s, 3H), 7.43 - 7.51 (m, 3H), 7.29 - 7.38 (m, 2H), 6.08 (s, 1 H), 3.67 - 3.70 (m, 2H), 2.23 (s, 3H). Intermediate 14
nomethyl)-6-methyl-4-(1 -methylethyl)-2(1 H)-pyridinone
Figure imgf000051_0001
The title compound was prepared in the same manner as described for intermediate 12 (steps 1 and 2) from (3£)-5-methyl-3-hexen-2-one (20 g, 137 mmol). The crude nitrile intermediate was obtained as 7 g (22 %), of which 3 g of this putative intermediate was converted to the title compound 1 .3 g as a TFA salt. LCMS E-S (M+H) = 181.1 . 1H N MR (400 MHz, DMSO-d6) δ ppm 1 1 .8 - 1 1 .9 (br s, 1 H), 7.86 - 7.96 (br s, 3H), 6,.10 (s, 1 H), 3.82 - 3.86 (m, 2H), 3.02 - 3.09 (m, 1 H), 2.17 (s, 3H), 1 .08 (d, 6H).
Intermediate 15
3-(Aminomethyl)-4-methyl-6-propyl-2(1H)-pyridinone
Figure imgf000051_0002
Step 1
To a solution of NaNH2 (32.5 g ,862 mmol) in anhydrous ether (500 ml.) at 30 °C was added dropwise a mixture of butyric acid ethyl ester (50 g ,431 mmol) and acetone (37.5 g 646.5 mol). After addition, the reaction mixture was stirred for 4 h. The reaction mixture was poured onto ice water with stirring. Additional ether was added, and the layers were separated. The aqueous layer was acidified to pH 5.0 with 2 N HCI and then to pH 7.5 with Na2C03. The aq. layer was then extracted with ether. The combined organic layers were dried over Na2S04, filtered, and concentrated in vacuo. The crude product (20 g, 156 mmol) and 2-cyanoacetamide (13.12 g , 156 mmol) were suspended in EtOH (160 ml.) at 75 °C, followed by addition of piperidine (13.2 g, 156 mmol ). The contents were stirred and heated at reflux for 1 h. The mixture was cooled to room temperature, and filtered. The collected solid was suspended in water and stirred for 1 h. The mixture was filtered and dried to give 4-methyl-2-oxo-6-propyl-1 ,2-dihydro-3- pyridinecarbonitrile (1 1 g, 40%). LCMS E-S (M+H) = 181.1. 1H NMR (400 MHz, DMSO-d6) 5 ppm 12.3 - 12.4 (br s, 1 H), 6.25 (s, 1 H), 3.64 (s, 3H), 2.50 (t, 2H), 1.63 (m, 2H), 0.94 (t, 3H). Step 2
Sodium acetate (3.5 g, 42.6 mmol), palladium on carbon (0.81 g) and platinum oxide (0.1 g) were placed in a dried Parr bottle flushed with nitrogen, followed by addition of a small amount of acetic acid (to wet the catalysts). A solution of 4-methyl-2-oxo-6- propyl-1 ,2-dihydro-pyridine-3-carbonitrile (5 g, 28 mmol) in acetic acid was added to the Parr bottle followed by additional acetic acid (200 mL). The vessel was capped, placed on Parr apparatus and hydrogenated at 45 psi for 12 h. The reaction mixture was filtered and the filtrate concentrated in vacuo. The crude product was purified by preparative HPLC to afford the title compound (TFA salt) as 4.1 g. LCMS E-S (M+H)) = 181.1. 1H N MR (400 MHz, DMSO-d6) δ ppm 1 1.8 - 1 1.9 (br s, 1 H), 7.83 - 7.88 (br s, 3H), 5.99 (s, 1 H), 3.77 - 3.81 (m, 2H), 2.37 (t, 2H), 1.53 (m, 2H), 0.83 (t, 3H).
Intermediate 16
-6-cyclopropyl-4-methyl-2(1 H)-pyridinone
Figure imgf000052_0001
Step 1
1 -Cyclopropyl-1 ,3-butanedione
To a a stirring solution of THF (100 mL) was suspended potassium tert-butoxide
(5.60 g, 49.5 mmol), followed by a mixture of cyclopropyl methyl ketone (3.27mL, 33 mmol) and ethyl acetate (9.69 mL, 99 mmol) in 30 mL THF at 35 °C, via addition funnel over a 25 min period. The contents were heated and stirred at 60 °C. After 3 h, the contents were removed from heating, and allowed to stir with cooling to room temperature. The reaction mixture was carefully diluted with 30 mL 2N HCI and stirred for 10 min. The mixture was extracted with diethyl ether (3 x 50 mL), and the combined organic layers washed with brine (1 x 50 mL). The organic layer was dried over MgS04, filtered, and concentrated in vacuo. The crude oil was chromatographed on silica gel (eluent: 0 to 15% EtOAc in hexanes) with good separation to afford the desired product as a light yellow colored oil, 3.9 g in -75% purity (residual solvent), for an overall yield of 70%. 1 H NMR (400 MHz, CHLOROFORM-d) δ ppm 0.89 - 0.96 (m, 2 H), 1 .09-1 .15 (m, 2 H), 1 .59-1 .69 (m, 1 H), 2.04 (s, 3H), 5.63 (s, 1 H), 15.5 - 16.0 (br s, 1 H). Step 2
6-Cyclopropyl-4-methyl-2-o -1 ,2-dihydro-3-pyridinecarbonitrile
Figure imgf000053_0001
To a stirring solution of ethanol (5 mL) were suspended 1 -cyclopropyl-1 ,3- butanedione (505 mg, 3.00 mmol) and cyanoacetamide (252 mg, 3.00 mmol), and the heterogenous contents heated until homogenous (ca. 75 °C). Next added piperidine (0.395 mL, 4.00 mmol), and the mixture was stirred with warming at reflux for 30 min. The reaction mixture was allowed to cool to room temperature, wherein precipitation ensued. The solid precipitate was filtered and set aside. The filtrate was concentrated in vacuo, and the oily residue treated with minimal EtOAc and then 10 mL hexanes to afford a 2nd crop of solid. The solid product crops were combined, suspended in water (7 mL), vigorously stirred, and vacuum filtered to afford a nearly white solid as 380 mg (73%). LCMS E-S (M+H) = 175.1 . 1H N MR (400 MHz, CHLOROFORM-d) δ ppm 1.01 - 1.09 (m, 2 H), 1 .28 (dd, J=8.59, 2.27 Hz, 2 H), 1 .95-2.01 (m, 1 H), 2.43 (s, 3H), 5.82 (s, 1 H).
Step 3
1 ,1 -Dimethylethyl [(6-cyclopropyl-4-methyl-2-oxo-1 ,2-dihydro-3- pyridinyl)methyl]carbamate
Figure imgf000053_0002
6-Cyclopropyl-4-methyl-2-oxo-1 ,2-dihydro-3-pyridinecarbonitrile (0.35 g, 2.01 mmol) was added to methanol (20 mL) and the stirring contents cooled to -10 °C. Next added di-tert-butyloxycarbonyl (0.933 mL, 4.02 mmol) and the suspension was stirred for 15 min. Next added in NiCI2-6H20 (0.055 g, 0.201 mmol) as a solid and stirred for 5 min. Next added NaBH4 (0.532 g, 14.06 mmol) in 6 portions with 5 min. increments between each portion. After completed addition (ca. 30min), the ice bath was removed and the contents were stirred with warming to room temperature overnight. The reaction mixture was returned to - 10 °C, followed by addition of 3 more portions of NaBH4 (0.532 g, 14.06 mmol). The ice bath was removed and the mixture stirred at room temperature for 1 h. The contents were quenched by addition of diethylethylene amine (0.218 mL, 2.01 mmol) and stirred for 45 min. at room temperature. The volatiles were removed in vacuo and the residue suspended in EtOAc and sat. NaHC03. The organic layer was washed with additional NaHC03. The layers were separated, and the organic layer dried over MgS04, filtered, and concentrated in vacuo. The crude product was purified by silica gel chromatography (eluent: 10% Methanol in Dichloromethane). The collected product was dried under hi-vacuum for 1 h, and then treated with ether and filtered. After drying in vacuum oven at 45 °C for 2 h, the product was collected as 0.28 g (50%). 1 H NMR (400 MHz, DMSO-d6) δ ppm 0.73 - 0.80 (m, 2 H), 0.88 - 0.96 (m, 2 H), 1.36 (s, 9 H), 1 .70 - 1 .82 (m, 1 H), 2.1 1 (s, 3 H), 3.95 (d, J=5.31 Hz, 2 H), 5.66 (s, 1 H), 6.51 (t, J=4.80 Hz, 1 H) , 1 1 .50 (br. s., 1 H).
Step 4
3-(Aminomethyl)-6-cyclopropyl-4-methyl-2(1 H)-pyridinone hydrochloride
1 , 1 -Dimethylethyl [(6-cyclopropyl-4-methyl-2-oxo-1 ,2-dihydro-3- pyridinyl)methyl]carbamate (0.28g, 1.006 mmol) was added to EtOAc (9 mL) and methanol (1.0ml_). The suspension was stirred at room temperature for 5 min., followed by addition of 4M HCI in dioxane (5.03 mL, 20.12 mmol), and the contents were stirred at room temperature overnight. The volatiles were then removed in vacuo to afford a solid. The solid was triturated with ether, filtered, and dried in a vacuum oven at 45°C for 4 h. The title compound was collected as 0.22 g (100% yield). 1H NMR (400 MHz, DMSO-c/6) δ ppm 0.78 - 0.86 (m, 2 H), 0.95 - 1.03 (m, 2 H), 1.83 (tt, J=8.46, 5.05 Hz, 1 H), 2.16 -
2.22 (m, 3 H), 3.75 (q, J=5.47 Hz, 2 H), 5.79 (s, 1 H), 8.02 (br. s., 3 H), 1 1 .92 (br. s., 1 H).
Example 1
6-Chloro-N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)- 1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000054_0001
6-Chloro-1-(1-methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid (0.12g, 0.501 mmol), 1 -hydroxy-7-azabenzotriazole (0.102 g, 0.751 mmol), EDC (0.144 g, 0.751 mmol), and 3-(aminomethyl)-4,6-dimethyl-2(1 H)-pyridinone (0.123 g, 0.651 mmol) were dissolved in dimethyl sulfoxide (3.0 mL) and stirred at room temperature. Added next to the stirring contents was N-methylmorpholine (0.220 mL, 2.003 mmol) via syringe at once. After stirring at room temperature overnight, the reaction mixture was slowly added to water (75 mL) and stirred for 10 min. After sitting for 10 min. at room temperature, the contents were filtered to afford a tan solid which was washed with water and then cold 50% aq EtOH. The contents were filtered, air-dried for 10 min., and then dried under vacuum for 1 hr. The collected solid was then further dried in a vacuum oven at 45 °C for 4 hr. The title compound was collected as 0.105 g (55%). LCMS E-S (M+H) = 373.9. 1H NMR (400 MHz, DMSO-d6) δ ppm 1.49 (d, J=6.82 Hz, 6 H), 2.13 (s, 3 H), 2.20 (s, 3 H), 4.35 (d, J=4.80 Hz, 2 H), 5.07 - 5.20 (m, 1 H), 5.89 (s, 1 H), 7.66 (s, 1 H), 8.39 (s, 1 H), 8.91 (t, J=4.80 Hz, 1 H), 1 1.55 (s, 1 H).
Example 2
6-Chloro-W-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-3-methyl-1 -(1 methylethyl)-1 H-pyraz -b]pyridine-4-carboxamide
Figure imgf000055_0001
6-chloro-3-methyl-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid (1324 mg, 5.22 mmol), 3-(aminomethyl)-4,6-dimethyl-2(1 H)-pyridinone. HCI (1329 mg, 7.05 mmol) and 1 -hydroxy-7-azabenzotriazole (1066 mg, 7.83 mmol) were stirred in 10 mL of DMSO for 10 min under nitrogen. N-methylmorpholine (2.3 mL, 20.88 mmol) was added along with EDC (1501 mg, 7.83 mmol) and the mixture became dark yellow. After a few hours solids precipitated and the contents became very thick, so 10 mL DMSO was added to facilitate stirring, The contents were stirred at RT overnight. Next added ice-water and then 10% K2C03 (pH ~ 8-9). The contents were stirred at RT for 30 min and then allowed to stand at RT for another 30 min. The contents were filtered, washed with water, and dried in vacuo. The title compound was collected as 1.67g (81 %) and used without further purification. 1H NMR (400 MHz, DMSO-d6) δ ppm 1.44 (d, 6 H), 2.12 (s, 3 H), 2.22 (s, 3 H), 2.40 (s, 3 H), 4.34 (d, J=5.05 Hz, 2 H), 5.05 (quin, J=6.63 Hz, 1 H), 5.88 (s, 1 H), 7.16 (s, 1 H), 8.78 (t, J=4.93 Hz, 1 H), 1 1.53 (br. s., 1 H). LCMS E-S (M+H) = 388.1 . Example 3
6-Chloro-1 -(1 ,1 -dimethylethyl)-W-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3- pyridinyl)methyl]-3-methyl -b]pyridine-4-carboxamide
Figure imgf000056_0001
To a solution of 6-chloro-1 -(1 , 1 -dimethylethyl)-3-methyl-1 H-pyrazolo[3,4- b]pyridine-4-carboxylic acid (320 mg, 1 .195 mmol) in DMSO(7 mL) were added 3- (aminomethyl)-4,6-dimethyl-2(1 H)-pyridinone (338 mg, 1 .793 mmol), N-methylmorpholine (0.526 mL, 4.78 mmol), 1 -hydroxy-7-azabenzotriazole (325 mg, 2.391 mmol) and EDC (458 mg, 2.391 mmol), and the reaction mixture was stirred overnight. The reaction mixture was quenched with water (20 mL) and stirred for 10 min. The precipitate was collected by filtration and further dried under high vacuum to give the product as a solid, 450 mg (94%). LCMS E-S (M+H) = 402.1 . 1 H NMR (400 MHz, DMSO-d6) δ ppm 1 .71 (s, 9 H), 2.12 (s, 3 H), 2.22 (s, 3 H), 2.37 (s, 3 H), 4.33 (d, J = 4.80 Hz, 2 H), 5.88 (s, 1 H), 7.09 - 7.21 (m, 1 H), 8.77 (t, J = 4.93 Hz, 1 H), 1 1 .53 (s, 1 H).
Example 4
W-[(4,6-Dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-6-(4- pyridinyloxy)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000056_0002
6-Chloro-N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)- 1 H-pyrazolo[3,4-b]pyridine-4-carboxamide (0.15 g, 0.401 mmol), 4-pyridinol (0.057 g, 0.602 mmol), cesium carbonate (0.261 g, 0.802 mmol), and 1 -(2-pyridinyl)-2-propanone (10.85 mg, 0.080 mmol) were sequentially dissolved in DMSO (4.0 mL). Next added copper(l) bromide (5.76 mg, 0.040 mmol) and the suspension was stirred under nitrogen (degassed) for 1 min. The sealed reaction mixture was stirred with heating at 1 10 °C (heating block) for 20 h, and then allowed to cool to room temperature overnight. The reaction mixture was diluted with EtOAc (25 mL) and water. The contents were vigorously stirred and then fitlered through Celite, washing the filter pad with 20%THF/EtOAc. The layers were separated and the aq. layer extracted with EtOAc. The combined organic layers were dried over MgS04, filtered, and concentrated in vacuo to a dark residue that was dried under hi-vacuum overnight. The crude product was purified by silica gel chromatography (eluent: gradient of 5-95 % Dichloromethane/ Chloroform containing 2M Ammonia (in methanol). The collected product was washed with MTBE, filtered, and dried in a vacuum oven at 45 °C for 5 hr. The final product was collected as 0.075 g (42%). LCMS E-S (M+H) = 433.2. 1H NMR (400 MHz, DMSO-d6) δ ppm 1H NMR (400 MHz, DMSO-d6) δ ppm 1.52 (d, J=6.57 Hz, 6 H), 2.13 (s, 3 H), 2.22 (s, 3 H), 4.41 (d, J=5.05 Hz, 2 H), 5.18 - 5.30 (m, 1 H), 5.91 (s, 1 H), 6.30 - 6.40 (m, 2 H), 7.91 (s, 1 H), 8.41 (s, 1 H), 8.57 - 8.65 (m, 2 H), 8.92 (t, J=4.93 Hz, 1 H), 1 1.58 (s, 1 H).
Example 5
W-[(4,6-Dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-6-(2- propen-1 -ylamino)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000057_0001
6-Chloro-N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)- 1 H-pyrazolo[3,4-b]pyridine-4-carboxamide (0.25 g, 0.669 mmol) was suspended in ethanol (4 mL) followed by addition of allylamine (0.753 mL, 10.03 mmol). The sealed contents were heated (heat block) at 140 °C for ca. 60 h, and then allowed to cool to room temperature. The reaction mixture was diluted with water (100 mL) and then filtered. The collected solid was washed with additional water and then dried in vacuum oven at 45 °C for 18h. The final product was isolated as an off-white solid, 0.225 g (83%). LCMS E- S (M+H) = 394.2. 1H NMR (400 MHz, DMSO-d6) δ ppm 1.42 (d, J=6.82 Hz, 6 H), 2.12 (s, 3 H), 2.21 (s, 3 H), 4.00 (t, J=5.43 Hz, 2 H), 4.32 (d, J=5.05 Hz, 2 H), 4.94 (quin, J=6.69 Hz, 1 H), 5.09 (dd, J=10.36, 1.77 Hz, 1 H), 5.18 - 5.29 (m, 1 H), 5.85 - 6.01 (m, 2 H), 6.68 (s, 1 H), 7.37 (t, J=5.56 Hz, 1 H), 7.84 (s, 1 H), 8.45 (t, J=5.05 Hz, 1 H), 1 1.55 (br. s., 1 H). Example 6
6-Amino-A -[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)- 1H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000058_0001
A mixture of 10% Pd/C (0.200 g, 0.188 mmol) and N-[(4,6-dimethyl-2-oxo-1 ,2- dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-6-(2-propen-1 -ylamino)-1 H-pyrazolo[3,4- b]pyridine-4-carboxamide (0.200g, 0.507 mmol) was suspended in ethanol (10 ml.) and stirred. Added next was methanesulfonic acid (0.033 ml_, 0.507 mmol), and the stirring contents were heated at reflux for 2 h. After cooling to room temperature, the reaction mixture was diluted with DCM and filtered through Celite. The filter pad was washed with 10% MeOH/DCM. The filtrate was pre-adsorped onto silica gel, and the crude product purified by silica gel chromatography (dry loaded, eluent: gradient of 5-80%
Dichloromethane/Chloroform containing 10% 2M Ammonia (in methanol)). The collected product was dried in vacuum oven at 45 °C overnight. The final product was collected as 0.065 g (36%). LCMS E-S (M+H) = 355.3. 1H NMR (400 MHz, DMSO-d6) δ ppm 1.40 (d, J=6.82 Hz, 6 H), 2.12 (s, 3 H), 2.21 (s, 3 H), 4.31 (d, J=5.05 Hz, 2 H), 4.85 - 5.00 (m, 1 H), 5.89 (s, 1 H), 6.49 - 6.68 (m, 3 H), 7.85 (s, 1 H), 8.46 (t, J=5.05 Hz, 1 H), 1 1.54 (br. s., 1 H).
Example 7
6-Cyclopropyl-1 -(1 -methylethyl)-W-[(4-methyl-2-oxo-6-propyl-1 ,2-dihydro-3- pyridinyl)methy yridine-4-carboxamide
Figure imgf000058_0002
6-Cyclopropyl-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid (167 mg, 0.680 mmol), 3-(aminomethyl)-4-methyl-6-propyl-2(1 /-/)-pyridinone trifluoroacetate (200 mg, 0.680 mmol), HOAT (139 mg, 1 .019 mmol), EDC (195 mg, 1 .019 mmol), and N- methylmorpholine (0.299 mL, 2.72 mmol) were dissolved in DMF(6 mL) and stirred at 40 °C for 24 h. The reaction mixture was poured into water (20 mL) and extracted with ethyl acetate (3 x 50 mL). The combined organic layers were dried over sodium sulfate and concentrated to an orange oil. The residue was dissolved in DMSO, and purified by reverse phase HPLC (mobile phase: 40-60% ACN in H20, 0.1 % TFA). The isolated product was dried in a vacuum oven overnight and furnished the TFA salt of the title compound as a white solid, 0.1 13 g (32%). LCMS E-S (M+H) = 408.1. 1H NMR (400 MHz, DMSO-d6) δ ppm 0.80 - 0.98 (m, 3 H), 1.06 (d, J = 7.07 Hz, 4 H), 1.46 (d, J = 6.82 Hz, 6 H), 1.52 - 1 .67 (m, 2 H), 2.17 - 2.31 (m, 4 H), 2.37 (t, J = 7.58 Hz, 2 H), 4.36 (d, J = 4.80 Hz, 2 H), 5.02 - 5.27 (m, 1 H), 5.91 (s, 1 H), 7.43 (s, 1 H), 8.21 (s, 1 H), 8.62 - 8.87 (m, 1 H), 1 1 .54 (br. s., 1 H).
Example 8
6-Cyclopropyl-1 -(1 -methylethyl)-W-{[6-methyl-4-(1 -methylethyl)-2-oxo-1 ,2-dihydro-3- pyridinyl]methyl}-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000059_0001
The title compound was prepared in the same manner as described in example 7 from 6-cyclopropyl-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid (250 mg, 1 .019 mmol), 3-(aminomethyl)-6-methyl-4-(1 -methylethyl)-2(1 H)-pyridinone«TFA (300 mg, 1 .019 mmol), HOAT (208 mg, 1.529 mmol), EDC (293 mg, 1 .529 mmol), N- methylmorpholine (0.448 mL, 4.08 mmol), and DMF(6 mL), wherein the reaction time was 48 h. The final product was collected following a basic extraction (to remove residual starting material) as a white solid, 30 mg (7%). LCMS E-S (M+H) = 408.3. 1H NMR (400 MHz, DMSO-de) δ ppm 0.95 - 1.16 (m, 10 H), 1 .38 - 1 .54 (m, 6 H), 2.16 (s 3 H), 2.19 - 2.30 (m, 1 H), 3.12 - 3.29 (m, 1 H), 4.29 - 4.48 (m, 2 H), 5.03 - 5.21 (m, 1 H), 6.03 (s, 1 H), 7.39 (s, 1 H), 8.21 (s, 1 H), 8.63 - 8.81 (m, 1 H), 1 1.56 (s, 1 H). Example 9
1 -(1 -Methylethyl)-A -[(4-methyl-2-oxo-6-propyl-1 ,2-dihydro-3-pyridinyl)methyl]-6-(4- pyridinyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000060_0001
1 -(1 -Methylethyl)-6-(4-pyridinyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid (192 mg, 0.680 mmol), 3-(aminomethyl)-4-methyl-6-propyl-2(1 H)-pyridinone«TFA (200 mg, 0.680 mmol), HOAT (139 mg, 1 .019 mmol), EDC (195 mg, 1 .019 mmol), and N- methylmorpholine (0.299 mL, 2.72 mmol) were sequentially added to DMF(6 mL), and the mixture stirred at 40 °C overnight. The reaction mixture was then filtered. The collected solid was washed with ethanol, and dried, affording the final product as a white solid,
0.160 g (53%). LCMS E-S (M+H) = 445.3. 1 H NMR (400 MHz, DMSO-d6) δ ppm 0.88 (t, 3 H), 1 .46 - 1 .65 (m, 8 H), 2.24 (s, 3 H), 2.38 (t, J = 7.58 Hz, 2 H), 4.43 (d, J = 4.80 Hz, 2 H), 5.37 (s, 1 H), 5.93 (s, 1 H), 8.23 (d, J = 5.81 Hz, 2 H), 8.30 (s, 1 H), 8.45 (s, 1 H), 8.78 (d, J = 5.31 Hz, 2 H), 9.00 (br. s., 1 H), 1 1 .56 (s, 1 H).
Example 10
W-[(6-Ethyl-4-methyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-6-(4- pyridinyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000060_0002
The title compound was prepared in the same manner as described in example 9 from 1 -(1 -methylethyl)-6-(4-pyridinyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid (403 mg, 1 .427 mmol), 3-(aminomethyl)-6-ethyl-4-methyl-2(1 H)-pyridinone«TFA (400 mg, 1 .427 mmol), HOAT (291 mg, 2.141 mmol), EDC (1094 mg, 5.71 mmol), N-methylmorpholine (0.628 mL, 5.71 mmol), and DMF(6 mL). The final product was collected as a white solid, 232 mg (38%). LCMS E-S (M+H) = 432.4. 1 H NMR (400 MHz, DMSO-d6) δ ppm 1 .14 (t, 3 H), 1 .56 (d, J = 6.57 Hz, 6 H), 2.25 (s, 3 H), 2.42 (q, J = 7.41 Hz, 2 H), 4.43 (d, J = 4.55 Hz, 2 H), 5.29 - 5.45 (m, 1 H), 5.93 (s, 1 H), 8.22 (d, J = 6.06 Hz, 2 H), 8.30 (s, 1 H), 8.45 (s, 1 H), 8.78 (d, J = 5.81 Hz, 2 H), 9.01 (t, J = 4.55 Hz, 1 H), 1 1.58 (s, 1 H). Example 11
1 -(1 -Methylethyl)-A -[(6-methyl-2-oxo-4-propyl-1 ,2-dihydro-3-pyridinyl)methyl]-6-(4- pyridinyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000061_0001
1-(1-Methylethyl)-6-(4-pyridinyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid (150 mg, 0.531 mmol), 3-(aminomethyl)-6-methyl-4-propyl-2(1 /-/)-pyridinone (1 15 mg, 0.531 mmol), EDC (122 mg, 0.638 mmol), HOAt (72.3 mg, 0.531 mmol), and N- methylmorpholine (0.233 mL, 2.12 mmol) were suspended in DMF (5 mL) and stirred at room temperature overnight, water was added to the reaction mixture, and the contents were filtered. The filter cake was washed with addional water (2x). The crude solid was purified by reverse phase HPLC (mobile phase: 10-30% ACN in H20, 0.1 % TFA). The isolated solid was then neutralized with saturated NaHC03, and washed with EtOAc (5 x 15 mL). The combined organic layers were dried over Na2S04, filtered, and concentrated in vacuo to afford the title compound as an off-white solid, 0.060 g (24%). LCMS E-S (M+H) = 445.3. 1H NMR (400 MHz, DMSO-d6) δ ppm 0.84 - 0.92 (m, 3 H), 1 .47 - 1 .54 (m, 2 H), 1 .57 (d, J=6.82 Hz, 6 H), 2.14 (s, 3 H), 2.48 - 2.52 (m, 2 H), 4.42 - 4.49 (m, 2 H),
5.33 - 5.43 (m, 1 H), 5.90 - 5.96 (m, 1 H), 8.28 - 8.37 (m, 3 H), 8.44 - 8.48 (m, 1 H), 8.82 - 8.87 (m, 2 H), 8.99 - 9.04 (m, 1 H), 1 1 .58 (s, 1 H).
Example 12
1 -(1 -Methylethyl)-W-{[6-methyl-4-(1 -methylethyl)-2-oxo-1 ,2-dihydro-3- pyridinyl]methyl}-6-(4^yridinyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000062_0001
A DMF solution (6 mL) of 1-(1-methylethyl)-6-(4-pyridinyl)-1 H-pyrazolo[3,4- b]pyridine-4-carboxylic acid (288 mg, 1 .019 mmol), 3-(aminomethyl)-6-methyl-4-(1- methylethyl)-2(1 H)-pyridinone«TFA (300 mg, 1.019 mmol), HOAT (208 mg, 1 .529 mmol), EDC (782 mg, 4.08 mmol), and N-methylmorpholine (0.448 mL, 4.08 mmol) was stirred overnight at 40 °C. After cooling to room temperature, the reaction mixture was poured into H20 (50 mL) and EtOAc (50 mL). The aqueous layer was extracted with EtOAC (2 x 50 mL). The organic layers were dried over sodium sulfate, filtered, and concentrated in vacuo. The residue was dissolved in DMSO and purified by reverse phase (15-40% ACN in H20, 0.1 % TFA). The product fractions were poured into sodium bicarbonate and extracted with ethyl acetate. The organic layer was dried over sodium sulfate, filtered, and concentrated in vacuo to afford the title compound as a white solid (123 mg, 27% yield). LCMS E-S (M+H) = 445.4. 1H N MR (400 MHz, DMSO-d6) δ ppm 1.10 (m, 6 H), 1 .56 (m, 6 H), 2.17 (s, 3 H), 3.17 - 3.28 (m, 1 H), 4.45 - 4.56 (m, 2 H), 5.29 - 5.43 (m, 1 H), 5.99 - 6.12 (m, 1 H), 8.16 - 8.23 (m, 2 H), 8.24 - 8.29 (m, 1 H), 8.40 - 8.46 (m, 1 H), 8.71 - 8.82 (m, 2 H), 8.96 - 9.04 (m, 1 H), 1 1.53 - 1 1.63 (m, 1 H).
Example 13
6-Cyclopropyl-1 -(1 -methylethyl)-A -[(6-methyl-2-oxo-4-propyl-1 ,2-dihydro-3- pyridinyl)methy -b]pyridine-4-carboxamide
Figure imgf000062_0002
The title compound was prepared in the same manner as described in example 1 1 from 1 -(1 -methylethyl)-6-(cyclopropyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid (150 mg, 0.612 mmol). The product was collected as a white solid, 0.043 g (16%). LCMS E-S (M+H) = 408.3. 1H NMR (400 MHz, DMSO-d6) δ ppm 0.89 (t, J=7.20 Hz, 3 H), 1 .06 (d, J=2.78 Hz, 3 H), 1.08 (br. s., 1 H), 1.47 (d, J=6.57 Hz, 8 H), 1.49 - 1.57 (m, 2 H), 2.14 (s, 3 H), 2.19 - 2.30 (m, 1 H), 4.38 (d, J=4.80 Hz, 2 H), 5.03 - 5.18 (m, 1 H), 5.92 (s, 1 H), 7.40 (s, 1 H), 8.21 (s, 1 H), 8.69 - 8.76 (m, 1 H), 1 1.55 (s, 1 H).
Example 14
1 -(1 -Methylethyl)-A -[(6-methyl-2-oxo-4-phenyl-1 ,2-dihydro-3-pyridinyl)methyl]-6-(4- pyridinyl)-1 -pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000063_0001
The title compound was prepared in the same manner as described in example 1 1 from 1 -(1 -methylethyl)-6-(4-pyridinyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid (150 mg, 0.531 mmol) and 3-(aminomethyl)-6-methyl-4-phenyl-2(1 /-/)-pyridinone (133 mg, 0.531 mmol). The product was collected as a white solid, 0.041 g (15%). LCMS E-S (M+H) = 479.3. 1H NMR (400 MHz, DMSO-d6) δ ppm 1.57 (d, J=6.82 Hz, 6 H) 2.23 (s, 3 H) 4.23 - 4.27 (m, 2 H) 5.33 - 5.42 (m, 1 H) 6.02 - 6.04 (m, 1 H) 7.35 - 7.42 (m, 1 H) 7.43 (s, 4 H) 8.23 - 8.26 (m, 1 H) 8.34 - 8.39 (m, 2 H) 8.41 - 8.42 (m, 1 H) 8.84 - 8.88 (m, 2 H) 8.91 - 8.96 (m, 1 H) 1 1.91 (s, 1 H).
Example 15
W-[(4-Cyclohexyl-6-methyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-6- (4-pyridinyl)- -pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000064_0001
A mixture of 1 -(1 -methylethyl)-6-(4-pyridinyl)-1 H-pyrazolo[3,4-b]pyridine-4- carboxylic acid (169 mg, 0.6 mmol), 3-(aminomethyl)-4-cyclohexyl-6-methyl-2(1 H)- pyridinone trifluoroacetate (221 mg, 0.660 mmol), EDC (150 mg, 0.780 mmol), HOAT (106 mg, 0.780 mmol), and N-methylmorpholine (0.264 ml_, 2.400 mmol) in DMF(3 mL) were stirred at room temperature for 6 days, water (15 mL) was added to the slurry, and it was stirred for an hour. The precipitate was then collected by vacuum filtration and rinsed with EtOH (4 mL), and the solid was dried in the vacuum oven overnight to give the title compounds as an off-white solid (137 mg, 45%). LCMS E-S (M+H) = 485.2. 1 H NMR (400 MHz, DMSO-d6) δ ppm 1 .09 - 1 .44 (m, 5 H), 1 .56 (d, J = 6.57 Hz, 6 H), 1 .55 - 1 .72 (m, 5 H), 2.15 (s, 3 H), 2.80 - 2.92 (m, 1 H), 4.53 (d, J = 4.55 Hz, 2 H), 5.36 (spt, J = 6.53 Hz, 1 H), 6.03 (s, 1 H), 8.18 - 8.23 (m, 2 H), 8.26 (s, 1 H), 8.45 (s, 1 H), 8.75 - 8.82 (m, 2 H), 9.01 (t, J = 4.55 Hz, 1 H), 1 1 .59 (s, 1 H).
Example 16
A -[(4-Cyclohexyl-6-methyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-6-cyclopropyl-1 -(1 - methylethyl)- -pyrazolo[3,4-fo]pyridine-4-carboxamide
Figure imgf000064_0002
The title compound was prepared in the same manner as described in example 15 from 6-cyclopropyl-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid (147 mg, 0.60 mmol), 3-(aminomethyl)-4-cyclohexyl-6-methyl-2(1 H)-pyridinone trifluoroacetate (221 mg, 0.660 mmol), EDC (150 mg, 0.780 mmol), HOAT (106 mg, 0.780 mmol), N- methylmorpholine (0.264 mL, 2.400 mmol) and DMF(3 mL), wherein the stir time was 3 d and the final product was not treated with EtOH. The final product was collected as 193 mg (68%). LCMS E-S (M+H) = 448.4. 1H NMR (400 MHz, DMSO-d6) δ ppm 1 .01 - 1 .1 1 (m, 4 H), 1.15 - 1 .43 (m, 5 H), 1.47 (d, J = 6.57 Hz, 6 H), 1.55 - 1.76 (m, 5 H), 2.15 (s, 3 H), 2.19 - 2.28 (m, 1 H), 2.83 (t, J = 1 1.12 Hz, 1 H), 4.45 (d, J = 5.05 Hz, 2 H), 5.12 (spt, J = 6.69 Hz, 1 H), 6.02 (s, 1 H), 7.37 (s, 1 H), 8.21 (s, 1 H), 8.74 (t, J = 4.80 Hz, 1 H), 1 1.56 (s, 1 H).
Example 17
6-Cyclopropyl-1 -(1 -methylethyl)-A -[(6-methyl-2-oxo-4-phenyl-1 ,2-dihydro-3- pyridinyl)methy -1H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000065_0001
The title compound was prepared in the same manner as described in example 1 1 from 1 -(1 -methylethyl)-6-(cyclopropyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid (150 mg, 0.612 mmol) and 3-(aminomethyl)-6-methyl-4-phenyl-2(1 /-/)-pyridinone (153 mg, 0.612 mmol). The product was collected as a white solid, 0.067 g (24%). LCMS E-S (M+H) = 442.3. 1H NMR (400 MHz, DMSO-d6) δ ppm 1 .04 - 1 .1 1 (m, 3 H), 1 .47 (d, J=6.57 Hz, 5 H), 2.22 (s, 3 H), 2.24 - 2.29 (m, 1 H), 3.17 (d, J=5.31 Hz, 2 H), 4.07 - 4.15 (m, 1 H), 4.19 (d, J=4.29 Hz, 2 H), 5.06 - 5.18 (m, 1 H), 6.01 (s, 1 H), 7.36 (s, 1 H), 7.38 - 7.49 (m, 5 H), 8.18 (s, 1 H), 8.70 (s, 1 H), 1 1.88 (s, 1 H). Example 18
A -[(4-Cyclopropyl-6-methyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)- 6-(4-pyridinyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000066_0001
1-(1-Methylethyl)-6-(4-pyridinyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid (100 mg, 0.354 mmol), 3-(aminomethyl)-4-cyclopropyl-6-methyl-2(1 /-/)-pyridinone (104 mg, 0.354 mmol), EDC (81 mg, 0.425 mmol), HOAt (48 mg, 0.354 mmol), and N- methylmorpholine (0.156 ml_, 1 .42 mmol) were suspended in DMF(5 mL) and stirred at room temperature overnight. The contents were filtered, washed with ethanol, and then concentrated in vacuo. The crude solid was purified by reverse phase HPLC (mobile phase : 20-40%, ACN in H20, 0.1 % TFA). The isolated solid was then neutralized with saturated NaHC03, and washed with EtOAc (3x). The combined organic layers were dried over Na2S04, filtered, and concentrated in vacuo to afford the title compound as an off- white solid, 0.042 g (26%). LCMS E-S (M+H) = 443.3. 1H NMR (400 MHz, DMSO-d6) δ ppm 0.71 - 0.78 (m, 2 H), 0.94 (dd, J=8.34, 2.27 Hz, 2 H), 1.57 (d, J=6.57 Hz, 6 H), 2.12 (s, 3 H), 2.13 - 2.18 (m, 1 H), 4.62 (d, J=4.80 Hz, 2 H), 5.38 (s, 1 H), 5.54 (s, 1 H), 8.37 (s, 1 H), 8.44 - 8.54 (m, 3 H), 8.91 (d, J=6.06 Hz, 2 H), 9.04 (s, 1 H), 1 1 .51 (br. s., 1 H).
Example 19
6-Cyclopropyl-A -[(6-cyclopropyl-4-methyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 - (1 -methylethyl)-1 H-pyrazolo 3,4-fo]pyridine-4-carboxamide
Figure imgf000066_0002
To a 4 mL solution of DMSO were sequentially added 6-cyclopropyl-1 -(1- methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid (0.1 1 g, 0.448 mmol), 3- (aminomethyl)-6-cyclopropyl-4-methyl-2(1 H)-pyridinone hydrochloride (0.106 g, 0.493 mmol), 1-hydroxy-7-azabenzotriazole (0.073 g, 0.538 mmol), EDC (0.103 g, 0.538 mmol), and then N-methylmorpholine (0.197 mL, 1 .79 mmol) via syringe. After stirring overnight at room temperature, the suspension was diluted with 50 mL of water and stirred for 15 min. After standing at room temperature for 15 min., the reaction mixture was filtered and the collected solid washed with additional water. The solid was then dried in a vacuum oven at 45 °C for 4 h. The product was obtained as 0.165 g (89%). LCMS E-S (M+H) = 406.5. 1H NMR (400 MHz, DMSO-d6) δ ppm 0.76 - 0.82 (m, 2 H), 0.91 - 0.98 (m, 2 H), 1.03 - 1.12 (m, 4 H), 1.46 (d, J=6.82 Hz, 6 H), 1 .74 - 1 .84 (m, 1 H), 2.19 (s, 3 H), 2.21 - 2.31 (m, 1 H), 4.34 (d, J=5.05 Hz, 2 H), 5.05 - 5.18 (m, 1 H), 5.73 (s, 1 H), 7.42 (s, 1 H), 8.21 (s, 1 H), 8.72 (t, J=4.93 Hz, 1 H), 1 1.61 (br. s., 1 H).
Example 20
W-[(5-Fluoro-4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-6- (4-pyridinyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000067_0001
A mixture of 1 -(1 -methylethyl)-6-(4-pyridinyl)-1 H-pyrazolo[3,4-b]pyridine-4- carboxylic acid (169 mg, 0.60 mmol), 3-(aminomethyl)-5-fluoro-4,6-dimethyl-2(1 H)- pyridinone hydrochloride (136 mg, 0.660 mmol), EDC (150 mg, 0.780 mmol), HOAT (106 mg, 0.780 mmol), and N-methylmorpholine (0.264 mL, 2.400 mmol) in DMF(3 mL) was stirred at room temperature for 3 days, water (15 mL) was added to the slurry, and it was stirred for an hour. The precipitate was then collected by vacuum filtration and rinsed with EtOH (4 mL), and the solid was dried in the vacuum oven overnight to give the title compound as 220 mg (76%). LCMS E-S (M+H) = 435.0. 1 H NMR (400 MHz, DMSO-d6) δ ppm 1.56 (d, 7 H), 2.18 (d, J = 2.78 Hz, 3 H), 2.25 (d, J = 2.02 Hz, 3 H), 4.45 (d, J = 4.80 Hz, 2 H), 5.37 (spt, J = 6.65 Hz, 1 H), 8.19 - 8.25 (m, 2 H), 8.29 (s, 1 H), 8.44 (s, 1 H), 8.74 - 8.81 (m, 2 H), 9.07 (t, J = 4.80 Hz, 1 H), 1 1 .66 (br. s., 1 H). Example 21
W-[(4,6-Di methyl -2 -oxo-1 ,2-dihydro-3-pyridinyl)methyl]-6-(ethylamino)-1 -(1 methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000068_0001
To a 10 mL microwave vial equipped with stir bar, septum cap and nitrogen inlet were added 6-chloro-N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 - methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide (23 mg, 0.062 mmol) and ethanol (1 mL). Added next to the stirring suspension was ethylamine (0.100 mL, 1 .230 mmol) via syringe at once. The sealed reaction mixture was irradiated (microwave) at 1 10 °C for 40 min, followed by addition of 0.1 mL ethylamine and further irradiation for 7 hr at 130 °C. After cooling to room temperature, about 50% of the volatiles were removed by a stream of nitrogen. The reaction mixture was diluted with water (20 mL) and stirred for 10 min. The mixture was then filtered and the collected solid washed with water. The solid was dried in vacuum oven at 50 °C for 18h, to afford the title compound as 0.17 g (71 %). LCMS E-S (M+H) = 383.3. 1H NMR (400 MHz, DMSO-d6) δ ppm 1.17 (t, J=7.07 Hz, 3 H), 1.43 (d, J=6.57 Hz, 6 H), 2.12 (s, 3 H), 2.21 (s, 3 H), 3.35 - 3.39 (m, 2 H), 4.31 (d,J=5.05 Hz, 2 H), 4.89 - 5.01 (m, 1 H), 5.89 (s, 1 H), 6.61 (s, 1 H), 7.18 (t, J=5.31 Hz, 1 H), 7.83 (s, 1 H), 8.44 (t, J=5.05 Hz, 1 H), 1 1.55 (br. s., 1 H).
Example 22
N-[(4,6-Dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-1 H- pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000068_0002
To a 50 mL round bottom flask fitted with a 3-way inlet was placed 10% Pd/C (0.071 g, 0.033 mmol) followed by degassing with N2, and then addition of 2 mL ethanol. Added next to the stirring slurry was 6-chloro-N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3- pyridinyl)methyl]-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide (.25 g, 0.669 mmol). Additional ethanol (8ml_) and THF (10ml_) were added with slight warming to facilitate dissolution. The stirring contents were allowed to cool (15 min.) then fitted with a balloon of H2 and allowed to stir at room temperature overnight. The contents were then purged with N2. The mixture was then diluted with 10% MeOH/DCM (20ml_), stirred for 10 min., and filtered through Celite. The filtrate was concentrated in vacuo and the crude solid was triturated with ethanol. After filtration and washing the collected solid with additional ethanol (cold), the solid was air-dried for 15 min, then in vacuum oven at 40 °C overnight. The final product was collected as 185 mg (80%). LCMS E-S (M+H) = 340.2. 1H NMR (400 MHz, DMSO-d6) δ ppm 1 .50 (d, J=6.82 Hz, 6 H), 2.13 (s, 3 H), 2.22 (s, 3 H), 4.37 (d, J=4.80 Hz, 2 H), 5.18 - 5.31 (m, 1 H), 5.89 (s, 1 H), 7.54 (d, J=4.80 Hz, 1 H), 8.36 (s, 1 H), 8.63 (d, J=4.55 Hz, 1 H), 8.82 (t, J=4.93 Hz, 1 H), 1 1.56 (s, 1 H).
Example 23
6-Cyclopropyl-1 -(1 -methylethyl)-N-{[6-methyl-2-oxo-4-(trifluoromethyl)-1 ,2-dihydro- 3-pyridinyl]methyl}-1 H- razolo[3,4-b]pyridine-4-carboxamide
Figure imgf000069_0001
The title compound was prepared in the same manner as described in example 19 from 6-cyclopropyl-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid (0.12 g, 0.489 mmol), 1 -hydroxy-7-azabenzotriazole (0.100 g, 0.734 mmol), 3-(aminomethyl)-6- methyl-4-(trifluoromethyl)-2(1 H)-pyridinone (0.154 g, 0.636 mmol, DMSO (3.0 ml_), N- methylmorpholine (0.215 ml_, 1 .957 mmol), and EDC (0.141 g, 0.734 mmol) The crude solid was purified by silica gel chromatography (eluent: gradient 5-100% of 10% 2M NH3 (in MeOH/DCM) and DCM) and the collected product dried in vacuum oven for 5h. The final product was collected as 0.1 12g (50%). LCMS E-S (M+H) = 434.3. 1H NMR (400
MHz, DMSO-d6) 5 ppm 1 .03 - 1.12 (m, 4 H), 1.47 (d, J=6.82 Hz, 6 H), 2.20 - 2.33 (m, 4 H), 4.34 - 4.54 (m, 2 H), 5.12 (quin, J=6.63Hz, 1 H), 6.33 (s, 1 H), 7.39 (s, 1 H), 8.18 (s, 1 H), 8.70 (t, J=4.04 Hz, 1 H), 12.43 (s, 1 H). Example 24
6-(Dimethylamino)-N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000070_0001
The title compound was prepared in the same manner as described in example 21 from 6-chloro-N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-1 H- pyrazolo[3,4-b]pyridine-4-carboxamide (23 mg, 0.062 mmol), ethanol (0.7 mL), and dimethylamine (0.461 mL, 0.923 mmol). The product was dried in vacuum oven at 50 °C for 5 hr and collected as 0.016 g (67%). LCMS E-S (M+H) = 383.3. 1H NMR (400 MHz, DMSO-de) δ ppm 1.44 (d, J=6.57 Hz, 6 H), 2.05 - 2.25 (m, 6 H), 3.14 (s, 6 H), 4.34 (d,
J=4.80 Hz, 2 H), 4.99 (dt, J=13.39, 6.69 Hz, 1 H), 5.89 (s, 1 H), 6.94 (s, 1 H), 7.96 (s, 1 H), 8.64 (t, J=4.93 Hz, 1 H), 1 1.56 (br. s., 1 H).
Example 25
N-[(4,6-Dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-6-(1 - piperidinyl)-1 H-p razolo[3,4-b]pyridine-4-carboxamide
Figure imgf000070_0002
To a 10 mL reaction vial containing stir bar were added 6-chloro-N-[(4,6-dimethyl- 2-OXO-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4- carboxamide (.060g, 0.160 mmol), ethanol (1 .5 mL), and then piperidine (0.318 mL, 3.21 mmol) via syringe at once. The contents were capped, placed into a heat block, and heated at 120 °C for 18 h. After cooling to room temperature, the reaction mixture was diluted with water (40 mL), adjusted to pH 6-7, and stirred for 15 min. The contents were filtered and washed with water. The product was dried in vacuum oven at 50 °C for 5 hr. The product was obtained as 57 mg (82%). LCMS E-S (M+H) = 422.8. 1H NMR (400 MHz, DMSO-d6) δ ppm 1 .43 (d, J=6.82 Hz, 6 H), 1 .53 - 1 .67 (m, 6 H), 2.12 (s, 3 H), 2.20 (s, 3 H), 3.61 - 3.73 (m, 4 H), 4.34 (d, J=5.05Hz, 2 H), 4.91 - 5.04 (m, 1 H), 5.89 (s, 1 H), 7.10 (s, 1 H), 7.97 (s, 1 H), 8.65 (t, J=4.93 Hz, 1 H), 1 1 .51 (br. s., 1 H).
Example 26
methyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-6-(4- morpholinyl)-1 H-p razolo[3,4-b]pyridine-4-carboxamide
Figure imgf000071_0001
To a 10 mL reaction vial containing stir bar were added 6-chloro-N-[(4,6-dimethyl- 2-OXO-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4- carboxamide (.060 g, 0.160 mmol), ethanol (1.5 mL), and then morpholine (0.280 mL, 3.21 mmol) via syringe at once. The contents were capped, placed into a heat block, and heated at 120 °C for 18 h and then at 135 °C for 2 hr. After cooling to room temperature, the reaction mixture was diluted with water (40 mL), adjusted to pH 6-7, and stirred for 15 min. The contents were filtered and washed with water. The product was dried in vacuum oven at 50 °C for 5 hr. The product was obtained as 57mg (79%). LCMS E-S (M+H) = 425.0. 1H NMR (400 MHz, DMSO-d6) δ ppm 1.44 (d, J=6.57 Hz, 6 H), 2.12 (s, 3 H), 2.19 (s, 3 H), 3.56 - 3.66 (m, 4 H), 3.69 - 3.78 (m, 4 H), 4.35 (d, J=4.80 Hz, 2 H), 5.00 (quin, J=6.63 Hz, 1 H), 5.89 (s, 1 H), 7.13 (s, 1 H), 8.03 (s, 1 H), 8.65 (t, J=4.93 Hz, 1 H), 1 1 .56 (br. s., 1 H).
Example 27
W-[(4,6-Dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-3-methyl-1 -(1 -methylethyl)-6- [6-(4-methyl-1 ^iperazin l)-3-pyridinyl]-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000072_0001
To a 10-mL microwave vial were added 6-chloro-N-[(4,6-dimethyl-2-oxo-1 ,2- dihydro-3-pyridinyl)methyl]-3-methyl-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4- carboxamide (70 mg, 0.180 mmol),1 -methyl-4-[5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan- 2-yl)-2-pyridinyl]piperazine (71.1 mg, 0.235 mmol) , DMSO (2.0 mL) and sodium carbonate (0.271 mL, 0.541 mmol) and the mixture was degassed for 10 min under nitrogen. Bis(triphenylphosphine)palladium(ll) chloride (10.13 mg, 0.014 mmol) was added and the contents were sealed. The mixture was irradiated (microwave) at 140 °C for 8 h. The reaction mixture was then quenched with water (5 mL) and filtered. The crude product was washed with water, dried, and then purified via silica gel chromatography (eluent: gradient of 0 to 15% (9 :1 MeOH/NH4OH)/DCM). The resulting product was treated with MeOH and 1 N HCI. The mixture was concentrated in vacuo, dried under high vacuum and collected as the HCI salt, 42 mg (36%). LCMS E-S (M+H) = 529.0 1 H NMR (400 MHz, DMSO-d6) δ ppm 1 .50 (d, 6 H), 2.13 (s, 3 H), 2.25 (s, 3 H), 2.42 (s, 3 H), 2.81 (d, J = 4.55 Hz, 3 H), 3.03 - 3.21 (m, 2 H), 3.41 (br. s., 2 H), 3.52 (d, J = 1 1.37 Hz, 2 H), 4.39 (d, J = 5.05 Hz, 2 H), 4.56 (d, J = 14.40 Hz, 2 H), 5.23 (quin, J = 6.63 Hz, 1 H), 5.91 (s, 1 H), 7.19 (d, J = 9.09 Hz, 1 H), 7.68 (s, 1 H), 8.50 (dd, J = 9.09, 2.27 Hz, 1 H) , 8.73 (t, J = 4.93 Hz, 3 H), 8.98 (d, J =2.02 Hz, 3 H), 10.97 - 1 1 .21 (m, 2 H)
Example 28
N-[(4,6-Dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-6-(1 piperazinyl)-1 H-py ine-4-carboxamide
Figure imgf000073_0001
To a 10 mL microwave vial equipped with stir bar were added 6-chloro-N-[(4,6- dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-1 H-pyrazolo[3,4- b]pyridine-4-carboxamide (.060g, 0.160 mmol), ethanol (1.5 mL), and 1 , 1-dimethylethyl 1- piperazinecarboxylate (0.299 g, 1 .605 mmol). The stirring suspension was placed onto heat block and heated at 120 °C for 18 h, and then irradiated (microwave) at 160 °C for 2 hr. The contents were cooled to room temperature, diluted into water (40 mL), and then adjusted to pH 6-7 and stirred for 15 min. The contents were filtered, washed with water, and then dried in hi-vac oven at 50 °C for 5 hr. The collected solid (52 mg) was dissolved in CH2CI2/TFA (3 mL, 2:1 ) and stirred at room temperature for 2 hr. The volatiles were removed in vacuo to afford a residue which was then dissolved in CH2CI2 and 10% 2M Ammonia (in methanol) in Chloroform. The crude product was purified by silica gel chromatography (eluent : gradient of 10% 2M Ammonia (in Methanol/Chloroform) and Dichloromethane). The isolated solid was dried in vacuum oven at 45 °C for 18 hr. to afford the final product as 38 mg (55 % yield). LCMS E-S (M+H) = 423.9. 1 H NMR (400 MHz, DMSO-d6) δ ppm 1 .43 (d, J=6.82 Hz, 6 H), 2.08 - 2.24 (m, 6 H), 2.75 - 2.89 (m, 4 H), 3.53 - 3.64 (m, 4 H), 4.34 (d, J=4.80 Hz, 2H), 4.98 (quin, J=6.69 Hz, 1 H), 5.89 (s, 1 H), 7.10 (s, 1 H), 8.00 (s, 1 H), 8.65 (t, J=4.93 Hz, 1 H), 1 1 .55 (br. s., 1 H).
Example 29
N-[(4,6-Dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-6-(3-hydroxy-3-methyl-1 - butyn-1 -yl)-1 -(1 -methylethyl -1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000073_0002
To a 10 mL microwave vial were sequentially added 6-chloro-N-[(4,6-dimethyl-2- oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4- carboxamide (.15 g, 0.401 mmol), sodium iodide (0.012 g, 0.080 mmol), zinc (5.25 mg, 0.080 mmol), DMSO(2.5 mL), triethylamine (0.1 12 mL, 0.802 mmol), and DBU (0.121 mL, 0.802 mmol). The suspension was stirred and degassed with nitrogen for 5 min, forming an emulsion. Next added 2-methyl-3-butyn-2-ol (0.194 mL, 2.006 mmol) and Pd(Ph3P)4 (0.046 g, 0.040 mmol). The stirring contents were heated at 90 °C for 3 hr, and then allowed to cool to room temperature. The reaction mixture was poured into a solution of water and 20% THF/EtOAc, and stirred. The layers were separated, and the organic layer washed with brine. The organic layer was dried over MgS04, and then filtered through Celite, washing the filter pad with additional EtOAc. The filtrate was concentrated in vacuo and the crude residue dried overnight on a hi-vac pump. The crude product was purified by silica gel chromatography (eluent: gradient: 5-80% of DCM and Chloroform containing 10% 2M Ammonia (in methanol)). The collected product was dried in vacuum oven at 45 °C overnight, to afford the final product as 0.1 16 g (67% yield). LCMS E-S
(M+H) = 421.9. 1H NMR (400 MHz, DMSO-d6) δ ppm 1.44 - 1.55 (m, 12 H), 2.13 (s, 3 H), 2.20 (s, 3 H), 4.35 (d, J=4.80 Hz, 2 H), 5.15 - 5.26 (m, 1 H), 5.70 (s, 1 H), 5.89 (s, 1 H), 7.65 (s, 1 H), 8.38 (s, 1 H), 8.93 (t, J=4.80 Hz, 1 H), 1 1 .55 (s, 1 H).
Example 30
N-[(4,6-Dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-6-(3- methyl-1 H-indazol-5-yl)- -pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000074_0001
To a 10 mL microwave vial were sequentially added 6-chloro-N-[(4,6-dimethyl-2- oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4- carboxamide (0.12 g, 0.321 mmol), 1 ,1 -dimethylethyl 3-methyl-5-(4,4,5,5-tetramethyl- 1 ,3,2-dioxaborolan-2-yl)-1 H-indazole-1-carboxylate (0.138 g, 0.385 mmol), potassium phosphate (tribasic) (0.204 g, 0.963 mmol), 1 ,4-dioxane (3 mL), and water (0.75 mL). The stirring suspension was degassed with nitrogen for 10 min., wherein an emulsion had formed. Next added PdCI2(dppf)-CH2CI2 adduct (0.039 g, 0.048 mmol) and the contents were placed onto a heat block and stirred at 105 °C overnight. After cooling to room temperature, the reaction mixture was diluted with EtOAc and filtered through Celite. The filter pad was washed with 50% THF/EtOAc. Silica gel was added to the combined filtrates and the mixture concentrated in vacuo to a solid. The contents were purified by silica gel chromatography (dry loaded, eluent: 5 - 80% gradient of DCM and chloroform containing 10% 2M Ammonia (in methanol)). The isolated solid was dried in vacuum oven at 45 °C overnight to afford the final product as 0.126 g (81 %). LCMS E-S (M+H) = 470.3. 1H NMR (400 MHz, DMSO-d6) δ ppm 1.56 (d, J=6.57 Hz, 6 H), 2.13 (s, 3 H), 2.25 (s, 3 H), 2.60 (s, 3 H), 4.43 (d, J=5.05 Hz, 2 H), 5.31 - 5.43 (m, 1 H), 5.91 (s, 1 H), 7.60 (d, J=8.84 Hz, 1 H), 8.22 (s, 1 H), 8.29 - 8.37 (m, 2 H), 8.60 (s, 1 H), 8.97 (t, J=5.05 Hz, 1 H), 1 1.58 (br. s., 1 H), 12.84 (s, 1 H).
Example 31
N-[(4,6-Dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-6- (phenylethynyl)-1 H- razolo[3,4-b]pyridine-4-carboxamide
Figure imgf000075_0001
To a 10 mL microwave vial were sequentially added 6-chloro-N-[(4,6-dimethyl-2- oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4- carboxamide (0.125 g, 0.334 mmol), sodium iodide (10.02 mg, 0.067 mmol) and zinc (4.37 mg, 0.067 mmol), DMSO(2.5 mL), triethylamine (0.093 mL, 0.669 mmol) and DBU (0.101 mL, 0.669 mmol). The stirring suspension was degassed with nitrogen for 5 min., wherein an emulsion had formed. Added next were phenylacetylene (0.1 10 mL, 1 .003 mmol) and Pd(Ph3P)4 (0.039 g, 0.033 mmol). The sealed reaction mixture was placed onto a heat block, stirred at 90 °C for 3 hr, and then allowed to cool to room temperature overnight. The contents were poured onto water and 20% THF/EtOAc, stirred, and the layers separted. The organic layer was washed with brine, dried over MgS04, filtered, and concentrated in vacuo. The filter pad was washed with additional EtOAc. The combined filtrates were concentrated in vacuo to a yellow/orange residue that was dried on hi-vac pump. The crude solid was then pre-adsorbed onto silica gel and purified by silica gel chromatography (dry loaded, eluent: 5-80 % gradient of DCM and chloroform containing 10% 2M Ammonia (in methanol)). The isolated product was obtained as a yellow solid which was then further purified by reverse phase HPLC (mobile phase: 20-90% ACN in H20, 0.1 % TFA, Gradient time: 8min). The isolated solid was dissolved in 10% MeOH/ CH2CI2 and treated with 0.6 g of Silicycle carbonate resin for 30 min. The contents were filtered through Celite and the filter pad washed with additional 10% MeOH/CH2CI2. The combined filtrates were concentrated in vacuo to afford a solid which was dried in vacuum oven for 18 hr. The final product was collected as 0.045 g (30%). LCMS E-S (M+H) = 440.2. 1H NMR (400 MHz, DMSO-d6) δ ppm 1.51 (d, J=6.82 Hz, 6 H), 2.13 (s, 3 H), 2.21 (s, 3 H), 4.37 (d, J=4.80 Hz, 2 H), 5.18 - 5.33 (m, 1 H), 5.90 (s, 1 H), 7.45 - 7.56 (m, 3 H), 7.64 - 7.73 (m, 2 H), 7.86 (s, 1 H), 8.42 (s, 1 H), 8.92 (t, J=4.80 Hz, 1 H), 1 1 .55 (br. s., 1 H).
Example 32
W-[(4,6-Dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-6-(2- phenylethyl)- -pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000076_0001
To a suspension of palladium on carbon (0.063 g, 0.059 mmol) in ethanol (1 ml.) under nitrogen was added N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 - methylethyl)-6-(4-pyridinylethynyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide (0.13 g, 0.295 mmol) and then ethanol (5 ml.) and tetrahydrofuran (THF) (1.5 ml_). The suspension was stirred under an atmosphere of hydrogen (ca. 1 atm, balloon) overnight. The reaction mixture was then evacuated with nitrogen, and diluted with 10%
MeOH/DCM. Celite was added and the contents stirred for 15 min., and then filtered through Celite (analytical grade) and washed with 10%MeOH/DCM. The filtrate was concentrated in vacuo and purified by silica gel chromatography (eluent: gradient of 5-95 %. Dichloromethane/Chloroform containing 10% 2M ammonia (in methanol). The collected solid was dried in vacuum oven at 45 °C for 18 h. The final product was collected as 0.1 12 g (84%). LCMS E-S (M+H) = 445.3. 1H NMR (400 MHz, DMSO-d6) δ ppm 1.46 (d, J=6.57 Hz, 6 H), 2.13 (s, 3 H), 2.21 (s, 3 H), 3.08 - 3.18 (m, 2 H), 3.22 - 3.29 (m, 2 H), 4.36 (d, J=5.05Hz, 2 H), 5.18 (quin, J=6.69 Hz, 1 H), 5.90 (s, 1 H), 7.28 (d, J=6.06 Hz, 2 H), 7.53 (s, 1 H), 8.26 (s, 1 H), 8.38 - 8.47 (m, 2 H), 8.72 (t, J=5.05 Hz, 1 H), 1 1.56 (s, 1 H). Example 33
N-[(4,6-Dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-6-(4- pyridinylethynyl)-1 H- razolo[3,4-b]pyridine-4-carboxamide
Figure imgf000077_0001
The title compound was prepared in the same manner as described in example 31 from 6-chloro-N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-1 H- pyrazolo[3,4-b]pyridine-4-carboxamide (.15 g, 0.401 mmol), sodium iodide (0.012 g, 0.080 mmol), zinc (5.25 mg, 0.080 mmol), DMSO (4.0 ml_), triethylamine (0.168 ml_, 1 .204 mmol), DBU (0.121 ml_, 0.802), 4-ethynylpyridine (0.1 12 g, 0.802 mmol), and Pd(Ph3P)4 (0.046 g, 0.040 mmol). The crude product was purified by silica gel chromatography (eluent: Gradient of 5-100% of DCM and chloroform containing 10% 2M Ammonia (in methanol)). The isolated product was dried in vacuum oven for 18 hr. to afford the final product as 0.032 g (18%). LCMS E-S (M+H) = 441.1. 1H NMR (400 MHz, DMSO-d6) δ ppm 1.51 (d, J=6.57 Hz, 6 H), 2.13 (s, 3 H), 2.21 (s, 3 H), 4.37 (d, J=4.80 Hz, 2 H), 5.20 - 5.32 (m, 1 H), 5.90 (s, 1 H), 7.62 - 7.69 (m, 2 H), 7.93 (s, 1 H), 8.45 (s, 1 H), 8.67 - 8.75 (m, 2 H), 8.93 (t, J=4.80 Hz, 1 H), 1 1.55 (s, 1 H).
Example 34
N-[(4,6-Dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-6- (phenylamino)-l H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000077_0002
To a 10 mL microwave vial were sequentially added 6-chloro-N-[(4,6-dimethyl-2- oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4- carboxamide (0.100 g, 0.267 mmol), cesium carbonate (0.305 g, 0.936 mmol), 1 ,4- Dioxane (2.5 mL) and aniline (0.049ml_, 0.535 mmol). The stirring suspension was degassed with nitrogen for 10 min. Added next were BINAP (0.033 g, 0.053 mmol) and palladium(ll) acetate (6.01 mg, 0.027 mmol). The sealed mixture was stirred at 105 °C overnight. After cooling to room temperature, the contents were poured onto EtOAc and filtered through Celite. The filter pad was washed with additional 50% THF/EtOAc. The combined filtrates were treated with silica gel and concentrated in vacuo. The crude product was purified by silica gel chromatography (dry loaded, eluent: Gradient 5-80% of DCM and chloroform containing 10% 2M Ammonia (in methanol))). The isolated solid was dried in a vacuum oven overnight and the final product collected as 0.061 g (52%). LCMS E-S (M+H) = 431.3. 1H NMR (400 MHz, DMSO-d6) δ ppm 1.50 (d, J=6.57 Hz, 6 H), 2.13 (s, 3 H), 2.23 (s, 3 H), 4.35 (d, J=5.05 Hz, 2 H), 4.98 - 5.1 1 (m, 1 H), 5.90 (s, 1 H), 6.93 - 7.00 (m, 2 H), 7.34 (t, J=7.96 Hz, 2 H), 7.86 (d, J=7.58 Hz, 2 H), 7.97 (s, 1 H), 8.59 (t, J=5.05 Hz, 1 H), 9.62 (s, 1 H), 1 1 .58 (br. s., 1 H).
Example 35
N-[(4,6-Dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-6- [(phenylmethyl)amino]-1 H- razolo[3,4-b]pyridine-4-carboxamide
Figure imgf000078_0001
To a 10 mL microwave vial were sequentially added 6-chloro-N-[(4,6-dimethyl-2- oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4- carboxamide (0.100 g, 0.267 mmol), ethanol (2.0ml_) and then benzylamine (0.350 mL, 3.21 mmol) via syringe at once. The sealed contents were irradiated at 140 °C for 3 hr. The contents were transferred to a heat block and heated at 135 °C for 16 hr., and then at 145 °C for an additional 12 h. After cooling to room temperature, the contents were diluted with CH2CI2 and pre-absorbed onto silica gel. The crude product was purified by silica gel chromatography (dry loaded, eluent; gradient of 5-80% DCM and chloroform containing 10% 2M Ammonia (in methanol)). The isolated solid was triturated with MTBE, filtered, and washed with additional MTBE. The collected solid was dried in vacuum oven at 45 °C overnight to afford the final product as 0.067 g (55%). LCMS E-S (M+H) = 445.3. 1H NMR (400 MHz, DMSO-d6) δ ppm 1.38 (d, J=6.57 Hz, 6 H), 2.12 (s, 3 H), 2.20 (s, 3 H), 4.31 (d, J=5.05 Hz, 2 H), 4.56 (d, J=6.06 Hz, 2 H), 4.92 (quin, J=6.69 Hz, 1 H), 5.88 (s, 1 H), 6.69 (s, 1 H), 7.18 - 7.25 (m, 1 H), 7.26 - 7.35 (m, 2 H), 7.35 - 7.43 (m, 2 H), 7.77 (t, J=5.94 Hz, 1 H), 7.83 (s, 1 H), 8.47 (t, J=5.05 Hz, 1 H), 1 1.55 (br. s., 1 H). Example 36
W-[(4,6-Dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-6-methyl-1 -(1 -methylethyl)- 1H-pyrazolo[ -b]pyridine-4-carboxamide
Figure imgf000079_0001
In a 25 ml. sealable tube under nitrogen were combined 6-methyl-1-(1 - methylethyl)-1 H-pyrazolo [3, 4-b] pyridine-4-carboxylic acid (60 mg, 0.27 mmol) and 3- (aminomethyl)-4,6-dimethyl-2(1 H)-pyridinone.HCI (62 mg, 0.33 mmol) in DMSO (3 ml_). 1 - hydroxy-7-azabenzotriazole (56 mg, 0.41 mmol) was added and the resulting mixture was degassed with nitrogen for 10 minutes. N-methylmorpholine (0.1 1 ml, 0.96 mmol) and EDC (79 mg, 0.41 mmol) were added, the vessel was sealed, and the light brown mixture was stirred at room temperature for 2 days. Next added 2 ml. of water and the mixture was stirred for 10 min. Solids that precipitated were sonicated, and allowed to stand at room temperature for 10 min. The contents were filtered, washed with water, and dried to afford the title compound (68 mg, 68%) as a light pink solid. LCMS E-S (M+H) = 354.3. 1 H NMR (400 MHz, DMSO-d6) δ ppm 1 1 .54 (s, 1 H), 8.69 (t, J=4.80 Hz, 1 H), 8.26 (s, 1 H), 7.46 (s, 1 H), 5.89 (s, 1 H), 5.20 (quin, J=6.69 Hz, 1 H), 4.36 (d, J=5.05 Hz, 2 H), 2.63 (s, 3 H), 2.21 (s, 3 H), 2.13 (s, 3 H), 1 .48 (d, J=6.57 Hz, 6 H).
Example 37
W-[(4,6-Di methyl -2-OXO-1 ,2-dihydro-3-pyridinyl)methyl]-1 ,6-bis(1 -methylethyl)-1 H- pyrazolo[ -b]pyridine-4-carboxamide
Figure imgf000079_0002
The title compound was prepared in the same manner as described in example 52 using 1 ,6-bis(1 -methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid (70 mg, 0.28 mmol) to afford an off-white solid (95 mg, 86%). LCMS E-S (M+H) = 382.3. 1 H NMR (400 MHz, DMSO-de) δ ppm 1 1.56 (br. s., 1 H, 8.76 (t, J=4.93 Hz, 1 H), 8.26 (s, 1 H), 7.50 (s, 1 H), 5.90 (s, 1 H), 5.15 - 5.25 (m, 1 H), 4.37 (d, J=5.05 Hz, 2 H), 3.13 - 3.23 (m, 1 H), 2.22 (s, 3 H), 2.13 (s, 3 H), 1 .50 (d, J=6.57 Hz, 6 H), 1.32 (d, J=6.82 Hz, 6 H).
Example 38
W-[(4,6-Dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-6-phenyl-
1H-pyrazol -b]pyridine-4-carboxamide
Figure imgf000080_0001
In a 25 ml. sealable tube under nitrogen were combined 1-(1-methylethyl)-6- phenyl-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid (70 mg, 0.25 mmol) and 3- (aminomethyl)-4,6-dimethyl-2(1 H)-pyridinone.HCI (56.3 mg, 0.3 mmol) in DMSO (3 ml_). 1-hydroxy-7-azabenzotriazole (51 mg, 0.37 mmol) was added and the resulting mixture was degassed with nitrogen for 10 min. N-methylmorpholine (0.1 ml, 0.87 mmol) and EDC (72 mg, 0.37 mmol) were added, the vessel was sealed, and the bright yellow mixture was stirred at room temperature for 2 days. Next added 2 ml. of water, and the contents were stirred for 10 min. Solids that precipitated were sonicated, and allowed to stand at room temperature for 10 min. The reaction contents were filtered and washed with water. The solid was treated with 2 ml. of EtOH, sonicated and heated, and then allowed to cool to room temperature. The contents were filtered, washed with water and dried to afford the title compound (74 mg, 70%) as a white solid. LCMS E-S (M+H) = 416.3. 1 H NMR (400 MHz, DMSO-d6) δ ppm 1 1.58 (s, 1 H), 8.98 (t, J=4.80 Hz, 1 H), 8.38 (s, 1 H), 8.25 - 8.30 (m, 2 H), 8.17 (s, 1 H), 7.49 - 7.59 (m, 3 H), 5.91 (s, 1 H), 5.30 - 5.38 (m, 1 H), 4.42 (d, J=4.80 Hz, 2 H), 2.23 (s, 3 H), 2.13 (s, 3 H), 1 .55 (d, J=6.57 Hz, 6 H). Example 39
methyl -2-OXO-1 ,2-dihydro-3-pyridinyl)methyl]-6-(4-fluorophenyl)-1 -(1 - methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000081_0001
In a 25 ml. sealable tube under nitrogen were combined 6-chloro-N-[(4,6-dimethyl-
2-OXO-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4- carboxamide (90 mg, 0.24 mmol), (4-fluorophenyl)boronic acid (33.7 mg, 0.24 mmol) in 1 ,4-dioxane (3 ml_). PdCI2(dppf)-CH2CI2 adduct (19.7 mg, 0.024 mmol) was added and the resulting mixture was degassed with nitrogen for 10 min. Sodium carbonate (77 mg, 0.72 mmol) was added, the vessel was sealed, and the mixture was heated at 85 °C for 2 hrs and then at 100 °C overnight. Added an additional 2 eq of boronic acid and 0.2 eq of PdCI2(dppf)-CH2CI2 adduct. The contents were heated at 120 °C for 4 hr. and then irradiated (microwave) first at 160 °C for 90 min, and then at 190 °C for 2 hrs. After cooling to room temperature, the solids were filtered, washed with DMSO and the filtrate was evaporated. The crude product was purified first by reverse-phase HPLC (C18, 5% to 80% CH3CN in water with 0.1 % TFA, 18 minute gradient) and then silica gel chromatography (eluent: gradient 0 to 90:10:1 DCM/MeOH/NH4OH). The collected solid was dried to afford the title compound (13 mg, 12%) as a white solid. LCMS E-S (M+H) = 434.2. 1 H NMR (400 MHz, DMSO-d6) δ ppm 1 1.58 (br. s., 1 H), 8.96 (t, J=4.67 Hz, 1 H), 8.38 (s, 1 H), 8.33 (dd, J=8.84, 5.56 Hz, 2 H), 8.16 (s, 1 H), 7.40 (t, J=8.84 Hz, 2 H), 5.91 (s, 1 H), 5.33 (dt, J=13.26, 6.76 Hz, 1 H), 4.41 (d, J=4.55 Hz, 2 H), 2.23 (s, 3 H), 2.13 (s, 3 H), 1.55 (d, J=6.57 Hz, 6 H).
Example 40
6-{4-[(Dimethylamino)sulfonyl]phenyl}-A -[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3- pyridinyl)methyl]-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-i ]pyridine-4-carboxamide
Figure imgf000082_0001
In a 25 ml. sealable tube under nitrogen were combined 6-chloro-N-[(4,6-dimethyl-
2-OXO-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4- carboxamide (85 mg, 0.23 mmol) and {4-[(dimethylamino)sulfonyl]phenyl}boronic acid (104 mg, 0.46 mmol) in DME/water (3 ml:1 ml). PdCI2(dppf)-CH2CI2 adduct (9.3 mg, 0.01 1 mmol) was added and the resulting mixture was degassed with nitrogen for 10 min. Sodium bicarbonate (57.3 mg, 0.68 mmol) was added and the insoluble light brown mixture was heated in an oil bath at 1 10 °C for 3 hrs. After cooling, 2 ml. of water was added to the black mixture and solids that precipitated were filtered. DMF was added along with a few drops of water and solids were filtered. DCM/MeOH (1 :1 ) was added to the grey solids and they were filtered and dried to afford the title compound (69 mg, 57%) as a grayish solid. LCMS E-S (M+H) = 523.2 . 1H NMR (400 MHz, DMSO-d6) δ ppm
1 1 .59 (br. s., 1 H), 9.01 (br. s., 1 H), 8.51 (m, J=8.34 Hz, 2 H), 8.44 (s, 1 H), 8.27 (s, 1 H), 7.93 (m, J=8.34 Hz, 2 H), 5.91 (s, 1 H), 5.36 (dt, J=13.14, 6.57 Hz, 1 H), 4.43 (d, J=4.55 Hz, 2 H), 2.66 (s, 6 H), 2.23 (s, 3 H), 2.13 (s, 3 H), 1 .56 (d, J=6.57 Hz, 6 H). Example 41
6-[6-(Dimethylamino)-3-pyridinyl]-A -[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3- pyridinyl)methyl]-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-fo]pyridine-4-carboxamide
Figure imgf000082_0002
In a 25 mL sealable tube under nitrogen were combined 6-chloro-N-[(4,6-dimethyl- 2-OXO-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4- carboxamide (80 mg, 0.21 mmol) and [6-(dimethylamino)-3-pyridinyl]boronic acid (53.3 mg, 0.32 mmol) in DME/water (3 mL : 1 mL). PdCI2 (dppf)-CH2CI2 adduct (8.7 mg, 0.01 1 mmol) was added and the resulting mixture was degassed with nitrogen for 10 min.
Sodium bicarbonate (53.9 mg, 0.64 mmol) was added, the vessel was sealed, and the insoluble green mixture was irradiated (microwave) at 150 °C for 20 min. After cooling, 2 mL of water was added to the dark green mixture and solids that precipitated were filtered. EtOAc was added, the mixture was heated and some hexanes were added. Solids were filtered, dissolved in DCM/MeOH (1 :1 ), and filtered through a pad of silica gel and the filtrate was evaporated. The residue was dissolved in DCM and purified by Si02 chromatography (eluent: gradient of 0 to 90:10 DCM/MeOH) to afford a residual oil that was triturated with EtOH/EtOAc (1 :1 ). The resultant solid was filtered and dried to afford the title compound (46 mg, 45%) as a yellow solid. LCMS E-S (M+H) = 460.2. 1H NMR (400 MHz, DMSO-de) δ ppm 1 1 .58 (s, 1 H), 9.02 (d, J=2.27 Hz, 1 H), 8.87 (t, J=4.67 Hz, 1 H), 8.36 (dd, J=8.84, 2.53 Hz, 1 H), 8.31 (s, 1 H), 8.08 (s, 1 H), 6.79 (d, J=8.84 Hz, 1 H), 5.91 (s, 1 H), 5.25 - 5.34 (m, 1 H), 4.40 (d, J=4.80 Hz, 2 H), 3.12 (s, 6 H), 2.23 (s, 3 H), 2.13 (s, 3 H), 1.53 (d, J=6.57 Hz, 6 H). Example 42
A -[(4,6-Dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-6-{4- [(methylamino)sulfonyl]phenyl}-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4- carboxamide
Figure imgf000083_0001
In a 25 mL sealable tube under nitrogen were combined 6-chloro-N-[(4,6-dimethyl- 2-OXO-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4- carboxamide (75 mg, 0.2 mmol), {4-[(methylamino)sulfonyl]phenyl}boronic acid (69 mg, 0.32 mmol) in DME/water (3 ml:1 ml). PdCI2(dppf)-CH2CI2 adduct (8.2 mg, 0.01 mmol) was added and the resulting mixture was degassed with nitrogen for 10 min. Sodium bicarbonate (50.6 mg, 0.6 mmol) was added , the vessel was sealed, and the insoluble mixture was heated in a microwave at 150 °C for 30 min. After cooling, 2 mL of water was added to the mixture and solids that precipitated were filtered. DCM/MeOH (1 :1 ) was added and the solution was filtered through a pad of silica gel and the filtrate was evaporated. The residue was purified by silica gel chromatography (eluent: gradient of 0 to 90:10 DCM/MeOH) and the isolated solid triturated in EtOAc. The solids were filtered and dried to afford the title compound (29 mg, 28%) as an off-white solid. LCMS E-S (M+H) = 509.0. 1H NMR (400 MHz, DMSO-d6) δ ppm 1 1 .58 (br. s., 1 H), 8.99 (t, J=4.93 Hz, 1 H), 8.47 (m, J=8.59 Hz, 2 H), 8.43 (s, 1 H), 8.25 (s, 1 H), 7.95 (m, J=8.59 Hz, 2 H), 7.60 (br. s., 1 H), 5.91 (s, 1 H), 5.36 (quin, J=6.69 Hz, 1 H), 4.42 (d, J=4.80 Hz, 2 H), 2.46 (s, 3 H), 2.23 (s, 3 H), 2.13 (s, 3 H), 1 .56 (d, J=6.82 Hz, 6 H).
Example 43
6-(4-Aminophenyl)-A -[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 - methylethyl)-1 -pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000084_0001
In a 25 mL sealable tube under nitrogen were combined 6-chloro-N-[(4,6-dimethyl- 2-OXO-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4- carboxamide (80 mg, 0.21 mmol) and (4-aminophenyl)boronic acid (44 mg, 0.32 mmol) in DME/water (3 ml:1 ml). PdCI2(dppf)-CH2CI2 adduct (8.74 mg, 0.01 1 mmol) was added and the resulting mixture was degassed with nitrogen for 10 min. Sodium bicarbonate (53.9 mg, 0.64 mmol) was added, the vessel was sealed, and the insoluble light pink mixture was irradiated (microwave) at 150 °C for 30 min. After cooling, DCM/MeOH (1 :1 ) was added, it was pre-absorbed on silica gel and purified by silica gel chromatography (eluent: gradient 0 to 90:10:1 DCM/MeOH/NH4OH). The isolated solid was treated with
MeOH. The solids that precipitated were filtered and dried to afford the title compound (63 mg, 66%) as a yellow solid. LCMS E-S (M+H) = 431.3. 1 H NMR (400 MHz, DMSO-d6) δ ppm 1 1 .56 (s, 1 H), 8.88 (t, J=4.93 Hz, 1 H), 8.27 (s, 1 H), 8.01 (s, 1 H), 7.99 (d, J=2.02 Hz, 2 H), 6.69 (s, 1 H), 6.67 (s, 1 H), 5.90 (s, 1 H), 5.61 (s, 2 H), 5.24 - 5.32 (m, 1 H), 4.40 (d, J=4.80 Hz, 2 H), 2.22 (s, 3 H), 2.13 (s, 3 H), 1 .53 (s, 3 H), 1.51 (s, 3 H).
Example 44
6-[4-(acetylamino)phenyl]-A -[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 - (1 -methylethyl)- -pyrazolo[3,4-fo]pyridine-4-carboxamide
Figure imgf000085_0001
In a 25 ml. sealable tube under nitrogen were combined 6-chloro-N-[(4,6-dimethyl- 2-OXO-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4- carboxamide (80 mg, 0.21 mmol) and [4-(acetylamino)phenyl]boronic acid (57.5 mg, 0.32 mmol) in DME/water (3 ml:1 ml). PdCI2(dppf)-CH2CI2 adduct (8.7 mg, 0.010 mmol) was added and the resulting mixture was degassed with nitrogen for 10 min. Sodium bicarbonate (53.9 mg, 0.64 mmol) was added, the vessel was sealed, and the insoluble light brown mixture was heated at 1 10 °C for 2.5 h. After cooling, 2 ml. of water was added to the dark grey mixture and solids that precipitated were filtered. EtOAc was added, the mixture was heated and some hexanes were added. The solids were filtered, dissolved in DCM/MeOH (1 :1 ) and 1 ml. of DMF. The contents were pre-absorbed onto silica gel and purified by silica gel chromatography (eluent: gradient 0 to 90:10:1
DCM/MeOH/NH40H). The isolated product was treated with MeOH, and the solids that precipitated were filtered, washed with hexanes and dried to afford the title compound (55 mg, 52%) as a white solid. LCMS E-S (M+H) = 473.1. 1H NMR (400 MHz, DMSO-d6) δ ppm 1 1 .57 (s, 1 H), 10.18 (s, 1 H), 8.93 (t, J=4.93 Hz, 1 H), 8.35 (s, 1 H), 8.22 (m, J=8.84 Hz, 2 H), 8.13 (s, 1 H), 7.76 (m, J=8.84 Hz, 2 H), 5.91 (s, 1 H), 5.28 - 5.37 (m, 1 H), 4.41 (d, J=4.80 Hz, 2 H), 2.23 (s, 3 H), 2.13 (s, 3 H), 2.09 (s, 3 H), 1 .54 (d, J=6.82 Hz, 6 H). Example 45
6-(3-aminophenyl)-A -[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000086_0001
In a 25 mL sealable tube under nitrogen were combined 6-chloro-N-[(4,6-dimethyl-
2-OXO-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4- carboxamide (80 mg, 0.21 mmol) and (3-aminophenyl)boronic acid (46.9 mg, 0.34 mmol) in DME/water (3 ml:1 ml). PdCI2(dppf)-CH2CI2 adduct (8.74 mg, 0.01 mmol) was added and the resulting mixture was degassed with nitrogen for 10 min. Sodium bicarbonate (53.9 mg, 0.64 mmol) was added, the vessel was sealed, and the reaction mixture was irradiated (microwave) at 150 °C for 25 min. After cooling, 2 mL of water was added to the black mixture and solids that precipitated were filtered. DCM/MeOH (1 :1 ) was added, the mixture was pre-absorbed on silica gel and purified by silica gel chromatography (eluent: gradient 0 to 90:10:1 DCM/MeOH/NH4OH). The isolated product was treated with EtOH/EtOAc/hexanes (1 :1 :1 ). The solids that precipitated were filtered and dried to afford the title compound (40 mg, 42%) as a light grey solid. LCMS E-S (M+H) = 431 .3. 1H NMR (400 MHz, DMSO-de) δ ppm 1 1.56 (br. s., 1 H), 8.96 (t, J=4.93 Hz, 1 H), 8.34 (s, 1 H), 8.02 (s, 1 H), 7.46 (t, J=1.89 Hz, 1 H), 7.36 (d, J=7.58 Hz, 1 H), 7.18 (t, J=7.71 Hz, 1 H), 6.69 (dd, J=7.83, 1.52 Hz, 1 H), 5.90 (s, 1 H), 5.25 - 5.37 (m, 3 H), 4.40 (d, J=4.80 Hz, 2 H), 2.22 (s, 3 H), 2.13 (s, 3 H), 1 .55 (s, 3 H), 1.54 (s, 3 H).
Example 46
A -[(4,6-Dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-6-{4- amino)carbonyl]phenyl}-1 -(1 -methylethyl)-1H-pyrazolo[3,4-b]pyridine-' carboxamide
Figure imgf000087_0001
The title compound was prepared in the same manner as described in example 61 using {4-[(methylamino) carbonyl] phenyl} boronic acid (57.5 mg, 0.32 mmol) to afford a light grey solid (70 mg, 68%). LCMS E-S (M+H) = 473.1 . 1H NMR (400 MHz, DMSO-d6) δ ppm 1 1 .58 (s, 1 H), 9.00 (t, J=4.80 Hz, 1 H), 8.59 (q, J=4.38 Hz, 1 H), 8.41 (s, 1 H), 8.35 (m, J=8.59 Hz, 2 H), 8.24 (s, 1 H) 8.01 (m, J=8.59 Hz, 2 H), 5.91 (s, 1 H), 5.33 - 5.41 (m, 1 H), 4.42 (d, J=4.80 Hz, 2 H), 2.83 (d, J=4.55 Hz, 3 H), 2.23 (s, 3 H), 2.13 (s, 3 H), 1.56 (s, 3 H), 1 .55 (s, 3 H).
Example 47
6-[3-(Acetylamino)phenyl]-W-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 - (1 -methylethyl)-1 H-pyrazolo[3,4-fo]pyridine-4-carboxamide
Figure imgf000087_0002
In a 25 ml. sealable tube under nitrogen were combined 6-chloro-N-[(4,6-dimethyl- 2-OXO-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4- carboxamide (80 mg, 0.21 mmol) and [3-(acetylamino)phenyl]boronic acid (57.5 mg, 0.32 mmol) in DME/water (3 ml:1 ml). PdCI2(dppf)-CH2CI2 adduct (8.7 mg, 0.010 mmol) was added and the resulting mixture was degassed with nitrogen for 10 min. Sodium bicarbonate (53.9 mg, 0.64 mmol) was added, the vessel was sealed and the reaction mixture was irradiated (microwave) at 150 °C for 30 min. After cooling, 2 mL of water was added and solids that precipitated were filtered. DCM/MeOH (1 :1 ) was added, the mixture was pre-absorbed on silica gel and purified by silica gel chromatography (eluent: gradient 0 to 90:10:1 DCM/MeOH/NH4OH). The isolated product was treated with EtOH and DCM, filtered, and dried to afford the title compound (65 mg, 63%) as a white solid. LCMS E-S (M+H) = 473.1 . 1H NMR (400 MHz, DMSO-d6) δ ppm 1 1 .58 (s, 1 H), 9.02 (t, J=4.80 Hz, 1 H), 8.61 - 8.68 (m, 2 H), 8.37 - 8.43 (m, 2 H), 8.21 (s, 1 H), 7.94 (d, J=7.83 Hz, 1 H), 7.65 (t, J=7.83 Hz, 1 H), 5.91 (s, 1 H), 5.37 (quin, J=6.63 Hz, 1 H), 4.42 (d, J=4.80 Hz, 2 H), 2.84 (d, J=4.29 Hz, 3 H), 2.24 (s, 3 H), 2.13 (s, 3 H), 1 .57 (s, 3 H), 1.55 (s, 3 H).
Example 48
A -[(4,6-Dimethyl-2-oxo-1 ,2-dihydro-3^yridinyl)methyl]-6-(2,4-dioxo-1 ,2,3,4- tetrahydro-5-pyrimidinyl)-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4- carboxamide
Figure imgf000088_0001
In a 25 mL sealable tube under nitrogen were combined 6-chloro-N-[(4,6-dimethyl- 2-OXO-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4- carboxamide (80 mg, 0.21 mmol) and (2,4-dioxo-1 ,2,3,4-tetrahydro-5-pyrimidinyl)boronic acid (50 mg, 0.32 mmol) in DME/water (3 ml:1 ml). PdCI2(dppf)-CH2CI2 adduct (8.74 mg, 0.01 1 mmol) was added and the resulting mixture was degassed with nitrogen for 10 min. Sodium bicarbonate (53.9 mg, 0.64 mmol) was added, the vessel was sealed, and reaction mixture was heated in a microwave at 150 °C for 30 min. After cooling, 2 mL of water was added to the black mixture and solids that precipitated were filtered.
DCM/MeOH (1 :1 ) was added, the mixture was pre-absorbed on silica gel and purified by silica gel chromatography (eluent: gradient 0 to 90:10:1 DCM/MeOH/NH4OH). The isolated product was treated with EtOH/EtOAc (1 :1 ) and sonicated. The solids that precipitated were filtered, washed with EtOH and DCM, and dried to afford the title compound (41 mg, 42%) as a light grey solid. LCMS E-S (M+H) = 449.9. 1 H NMR (400 MHz, DMSO-de) δ ppm 1 1.58 (s, 1 H), 1 1.52 (br. s., 1 H), 1 1.47 (s, 1 H), 8.75 (t, J=5.05 Hz, 1 H), 8.36 (s, 1 H), 8.31 (s, 1 H), 8.23 (s, 1 H), 5.90 (s, 1 H), 5.27 (quin, J=6.69 Hz, 1 H), 4.37 (d, J=5.05 Hz, 2 H), 2.23 (s, 3 H), 2.13 (s, 3 H), 1 .52 (s, 3 H), 1.50 (s, 3 H).
Example 49
6-(2-Amino-4-pyridinyl)-A -[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 - methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000089_0001
The title compound was prepared in the same manner as example 48 using 4- (4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)-2-pyridinamine (65.9 mg, 0.3 mmol) to afford an off-white solid (60 mg, 73%). LCMS E-S (M+H) = 432.3. 1H NMR (400 MHz, DMSO- d6) δ ppm 1 1 .57 (s, 1 H), 8.99 (t, J=4.80 Hz, 1 H), 8.41 (s, 1 H), 8.05 - 8.13 (m, 2 H), 7.24 - 7.32 (m, 2 H), 6.14 (s, 2 H), 5.91 (s, 1 H), 5.33 (quin, J=6.69 Hz, 1 H), 4.41 (d, J=4.80 Hz, 2 H), 2.22 (s, 3 H), 2.13 (s, 3 H), 1 .55 (d, J=6.57 Hz, 6 H). Example 50
6-[4-(Aminosulfonyl)phenyl]-A -[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]- 1 -(1 -methylethyl)- -pyrazolo[3,4-fo]pyridine-4-carboxamide
Figure imgf000089_0002
The title compound was prepared in the same manner as example 48 using [4- (aminosulfonyl) phenyl] boronic acid (64.5 mg, 0.32 mmol) to afford a light grey solid (76 mg, 74 %). LCMS E-S (M+H) = 495.0. 1 H NMR (400 MHz, DMSO-d6) δ ppm 1 1 .58 (br. s., 1 H), 8.99 (t, J=4.80 Hz, 1 H), 8.41 - 8.48 (m, 3 H), 8.25 (s, 1 H), 8.00 (d, J=8.59 Hz, 2 H), .50 (s, 2 H), 5.91 (s, 1 H), 5.37 (quin, J=6.69 Hz, 1 H), 4.43 (d, J=4.80 Hz, 2 H), 2.23 (s,H), 2.14 (s, 3 H), 1.56 (d, J=6.57 Hz, 6 H).
Example 51
W-[(4,6-Di methyl -2 -oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-6-{4- [(methylsulfonyl)amino]phenyl}-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000090_0001
In a 25 mL sealable tube under nitrogen were combined 6-chloro-N-[(4,6- dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-1 H-pyrazolo[3,4- b]pyridine-4-carboxamide (75 mg, 0.2 mmol) and {4-
[(methylsulfonyl)amino]phenyl}boronic acid (43.1 mg, 0.2 mmol) in DME/water (3 ml:1 ml). PdCI2(dppf)-CH2Cl2 adduct (8.2 mg, 0.01 mmol) was added and the resulting mixture was degassed with nitrogen for 10 min. Sodium bicarbonate (50.6 mg, 0.6 mmol) was added, the vessel was sealed, and the reaction mixture was irradiated (microwave) at 150 °C for 25 min. After cooling, 2 mL of water was added to the black mixture and solids that precipitated were filtered. DCM/MeOH (1 :1 ) was added, the mixture was pre-absorbed on silica gel and purified by silica gel chromatography (eluent: gradient 0 to 90:10:1 DCM/MeOH/NH4OH). The isolated product was suspended in EtOAc heated, and sonicated. Some hexanes was added and the contents allowed to cool to room temperature. Solids that precipitated were filtered. The solid was then suspended in EtOH, heated, and sonicated. After cooling to room temperature, the solids were filtered and dried to afford the title compound (38 mg, 36%) as a beige solid. LCMS E-S (M+H) = 509.0. 1H NMR (400 MHz, DMSO-d6) δ ppm 1 1 .57 (s, 1 H), 10.05 (br. s., 1 H), 8.93 (t, J=4.93 Hz, 1 H), 8.35 (s, 1 H), 8.24 (m, J=8.59 Hz, 2 H), 8.1 1 (s, 1 H), 7.37 (m, J=8.84 Hz, 2 H), 5.91 (s, 1 H), 5.32 (quin, J=6.69 Hz, 1 H), 4.41 (d, J=4.80 Hz, 2 H), 3.08 (s, 3 H), 2.23 (s, 3 H), 2.13 (s, 3 H),
1.55 (s, 3 H), 1.54 (s, 3 H). Example 52
6-{4-[(Dimethylamino)sulfonyl]phenyl}-A -[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3- pyridinyl)methyl]-3-methyl-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4- carboxamide
Figure imgf000091_0001
In a 25 ml. sealable tube under nitrogen were combined 6-chloro-N-[(4,6-dimethyl- 2-OXO-1 ,2-dihydro-3-pyridinyl)methyl]-3-methyl-1 -(1 -methylethyl)-1 H-pyrazolo[3,4- b]pyridine-4-carboxamide (75 mg, 0.19 mmol) and N,N-dimethyl-4-(4,4,5,5-tetramethyl- 1 ,3,2-dioxaborolan-2-yl)benzenesulfonamide (90 mg, 0.29 mmol) in DME/water (3 ml:1 ml). PdCI2 (dppf)-CH2CI2 adduct (7.9 mg, 0.009 mmol) was added and the resulting mixture was degassed with nitrogen for 10 min. Sodium bicarbonate (48.7 mg, 0.58 mmol) was added, the vessel was sealed, and the reaction mixture was irradiated (microwave) at 150 °C for 30 min. After cooling, 2 ml. of water was added and solids that precipitated were filtered. DCM/MeOH (1 :1 ) was added, the mixture was pre-absorbed on silica gel and purified by silica gel chromatography (eluent: gradient 0 to 90:10:1
DCM/MeOH/NH4OH). The isolated product was treated with EtOH/EtOAc (1 :1 ), the solids filtered and washed with EtOAc and DCM, and then dried to afford the title compound (83 mg, 78%) as a white solid. LCMS E-S (M+H) = 537.1. 1H NMR (400 MHz, DMSO-d6) δ ppm 1 1 .54 (s, 1 H) 8.79 (t, J=4.93 Hz, 1 H) 8.47 (m, J=8.59 Hz, 2 H) 7.90 (m, J=8.59 Hz, 2 H) 7.80 (s, 1 H) 5.89 (s, 1 H) 5.28 (quin, J=6.69 Hz, 1 H) 4.40 (d, J=5.05 Hz, 2 H) 2.66 (s, 6 H) 2.47 (s, 3 H) 2.25 (s, 3 H) 2.12 (s, 3 H) 1.53 (s, 3 H) 1.51 (s, 3 H). Example 53
W-[(4,6-Dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-6-(4- piperidinylamino)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000092_0001
In a 25 ml. sealable tube under nitrogen were combined 6-chloro-N-[(4,6-dimethyl- 2-OXO-1 , 2-dihydro-3-pyridinyl)methyl]-1 -(1-methylethyl)-1 H-pyrazolo [3,4-b]pyridine-4- carboxamide ( 60 mg, 0.16 mmol) and 4-piperidinamine (2 ml.) in EtOH (3 ml_). The vessel was sealed and the reaction mixture was irradiated (microwave) at 125 °C for 5 hr and then at 160 °C for 90 min. The mixture was evaporated under vacuum and the resulting residue was partitioned between EtOAc and water. Organics were washed with water (2x) and brine, dried over MgSC^, filtered and evaporated. The residue was purified by silica gel chromatography (eluent: gradient 0 to 90:10:1 DCM/MeOH/NH4OH). The isolated product was treated with EtOAc/ether (1 :1 ) and allowed to stand at room temperature overnight. The solids that had precipitated were triturated, filtered, and dried to afford the title compound as a pale yellow solid 25 mg (35%). LCMS E-S (M+H) = 438.0. 1H NMR (400 MHz, DMSO-d6) δ ppm 8.66 (t, J=4.67 Hz, 1 H) 7.98 (s, 1 H) 7.12 (s,
1 H) 5.89 (s, 1 H) 4.98 (dt, J=13.33, 6.60 Hz, 1 H) 4.30 - 4.41 (m, 4 H) 3.01 (t, J=1 1 .62 Hz,
2 H) 2.85 (br. s., 1 H) 2.20 (s, 3 H) 2.12 (s, 3 H) 1 .80 (d, J=10.36 Hz, 2 H) 1.44 (s, 3 H) 1.43 (s, 3 H) 1.18 - 1.30 (m, 3 H) 0.79 - 0.91 (m, 1 H)
Example 54
W-[(4,6-Dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-6- (tetrahydro-2H-pyran-4-ylamino)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000093_0001
In a 25 mL sealable tube under nitrogen were combined 6-chloro-N-[(4,6-dimethyl-
2-OXO-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4- carboxamide (60 mg, 0.16 mmol) and tetrahydro-2H-pyran-4-amine (2 ml, 19 mmol) in EtOH (3 mL). The vessel was sealed and the reaction mixture was irradiated (microwave) at 125 °C for 2 hr. and then at 170 °C for 1 hr. The mixture was concentrated to ca. 20% volume and NMP (2 mL) added to the reaction mixture. The contents were again irradiated (microwave) first at 180 °C for 2.5 h and then at 190 °C for 75 min. The mixture was evaporated under vacuum, and the resulting residue was purified by silica gel chromatography (eluent: gradient 0 to 90:10:1 DCM/MeOH/Nh^OH). The isolated product was treated with EtOAc/hexanes (1 :1 ), sonicated, and heated. After cooling, solids that precipitated were filtered and dried to afford the title compound (46 mg, 64%) as an off- white solid. LCMS E-S (M+H) = 439.1. 1H NMR (400 MHz, DMSO-d6) δ ppm 1 1 .55 (s, 1 H) 8.44 (t, J=5.05 Hz, 1 H) 7.83 (s, 1 H) 7.20 (d, J=7.33 Hz, 1 H) 6.63 (s, 1 H) 5.89 (s, 1 H) 4.93 (quin, J=6.69 Hz, 1 H) 4.32 (d, J=5.05 Hz, 2 H) 3.99 - 4.06 (m, 1 H) 3.88 (dt, J=1 1.49, 3.47 Hz, 2 H) 3.45 (td, J=1 1.37, 2.02 Hz, 2 H) 2.21 (s, 3 H) 2.13 (s, 3 H) 1 .95 (d, J=10.61 Hz, 2 H) 1.41 - 1.51 (m, 8 H).
Example 55
A -[(4,6-Di methyl -2-OXO-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-6-[(4- pyridinylmethyl)ami -1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000094_0001
In a 25 mL sealable tube under nitrogen were combined 6-chloro-N-[(4,6-dimethyl-
2-OXO-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4- carboxamide (60 mg, 0.16 mmol) and (4-pyridinylmethyl)amine (0.065 ml, 0.64 mmol) in EtOH (2 mL). The vessel was sealed and the reaction mixture was heated at 90 °C overnight. An additional 1 mL of (4-pyridinylmethyl) amine was added and the mixture was heated in a microwave at 160 °C for 90 min. The EtOH solvent was removed under reduced pressure, 2 mL of NMP was added, and the mixture was then irradiated
(microwave) at 180 °C for 90 min. The mixture was evaporated under vacuum, and the resulting residue was partitioned between EtOAc and water. The crude residue was extracted with EtOAc, DCM and DCM/isopropanol (70:30). The combined organics were washed with water (2x) and brine, dried over MgS0 , filtered and evaporated. The residue was purified by silica gel chromatography (eluent: gradient 0 to 90:10:1
DCM/MeOH/NH40H). The isolated yellow oil was treated with EtOAc and sonicated. The filtered solid was washed with hexanes and dried to afford the title compound (26 mg, 36%) as a white solid. LCMS E-S (M+H) = 446.1. 1H NMR (400 MHz, DMSO-d6) δ ppm 1 1 .56 (s, 1 H) 8.50 (t, J=5.18 Hz, 1 H) 8.43 - 8.48 (m, 2 H) 7.89 (t, J=5.94 Hz, 1 H) 7.84 (s, 1 H) 7.34 (d, J=6.06 Hz, 2 H) 6.73 (s, 1 H) 5.89 (s, 1 H) 4.83 (quin, J=6.69 Hz, 1 H) 4.58 (d, J=5.81 Hz, 2 H) 4.32 (d, J=5.05 Hz, 2 H) 2.21 (s, 3 H) 2.13 (s, 3 H) 1 .32 (d, J=6.82 Hz, 6 H). Example 56
6-{[2-(Dimethylamino)ethyl]amino}-A -[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3- pyridinyl)methyl]-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-i ]pyridine-4 -carboxamide
Figure imgf000095_0001
In a 25 ml. sealable tube under nitrogen were combined 6-chloro-N-[(4,6-dimethyl-
2-OXO-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4- carboxamide (80 mg, 0.21 mmol) and (2-aminoethyl)dimethylamine (37.7 mg, 0.43 mmol) in NMP (2 ml_). The vessel was sealed and the reaction mixture was irradiated
(microwave) at 150 °C for 1 h. The mixture was evaporated under vacuum, and the resulting residue was partitioned between EtOAc and water. The contents were extracted with EtOAc, DCM and DCM/isopropanol (70:30). The combined organics were washed with water (2x) and brine, dried over MgSC^, filtered and evaporated. The residue was purified by silica gel chromatography (eluent: gradient 0 to 90:10:1 DCM/MeOH/Nh^OH).
The isolated yellow oil was treated with EtOAc and sonicated. Solids that precipitated were filtered, washed with hexanes, and dried to afford the title compound (60 mg, 65%) as a white solid. LCMS E-S (M+H) = 425.9. 1 H NMR (400 MHz, DMSO-d6) δ ppm 1 1 .55 (br. s., 1 H) 8.43 (t, J=5.05 Hz, 1 H) 7.84 (s, 1 H) 7.08 (t, J=5.05 Hz, 1 H) 6.67 (s, 1 H) 5.89 (s, 1 H) 4.94 (quin, J=6.63 Hz, 1 H) 4.32 (d, J=5.05 Hz, 2 H) 3.44 (q, J=6.48 Hz, 2 H) 2.44 (t, J=6.69 Hz, 2 H) 2.17 - 2.22 (m, 9 H) 2.12 (s, 3 H) 1.43 (d, J=6.57 Hz, 6 H).
Example 57
W-[(4, 6-Di methy I -2-OXO-1 ,2 -d i hyd ro-3 ri d i ^
(1 -methylethyl)-1 H-pyrazolo[3,4-fo]pyridine-4-carboxamide
Figure imgf000096_0001
In a 25 mL sealable tube under nitrogen were combined 6-chloro-N-[(4,6-dimethyl- 2-OXO-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4- carboxamide (80 mg, 0.21 mmol) and 2-aminoethanol (1 ml) in NMP (2 mL). The vessel was sealed and the reaction mixture was irradiated (microwave) at 130 °C for 1 h and then at 140 °C for 90 min. The mixture was evaporated under vacuum and the residue was purified by silica gel chromatography (eluent: gradient 0 to 90:10:1 DCM/MeOH/Nh^OH).
The isolated yellow oil was treated with EtOAc, sonicated, and treated with hexanes. The solids were filtered, dissolved in DMF (1 mL), and a few drops of water were added. The solution was allowed to stand at room temperature overnight, and then sonicated. The filtered solid was washed with Hexanes/EtOAc (1 :1 ), and then dried to afford the title compound (55 mg, 63%) as a light yellow solid. LCMS E-S (M+H) = 399.0. 1H NMR (400 MHz, DMSO-de) δ ppm 1 1.56 (br. s., 1 H) 8.44 (br. s., 1 H) 7.84 (s, 1 H) 7.20 (br. s., 1 H) 6.68 (s, 1 H) 5.89 (s, 1 H) 4.95 (dt, J=13.01 , 6.38 Hz, 1 H) 4.72 (t, J=4.93 Hz, 1 H) 4.32 (d, J=4.80 Hz, 2 H) 3.58 (d, J=5.56 Hz, 2 H) 3.38 - 3.46 (m, 2 H) 2.21 (s, 3 H) 2.12 (s, 3 H) 1.43 (d, J=6.57 Hz, 6 H)
Example 58
N-[(4,6-Dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-6-[5- (methyloxy)-3-pyridinyl]-1 b]pyridine-4-carboxamide
Figure imgf000096_0002
To a 20 mL microwave vial were sequentially added 6-chloro-N-[(4,6-dimethyl-2- oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4- carboxamide (100 mg, 0.267 mmol, 3-(methyloxy)-5-(4,4,5,5-tetramethyl-1 ,3,2- dioxaborolan-2-yl)pyridine (69.2 mg, 0.294 mmol), PdCI2(dppf)-CH2CI2 adduct (21 .84 mg, 0.027 mmol), (DME) (5 mL), and water (2 mL). The reaction mixture was degassed with nitrogen for 5 min. Sodium bicarbonate (67.4 mg, 0.802 mmol) was added and the contents sealed and irradiated (microwave) at 140 °C. The reaction mixture was cooled to room temperature and poured on a silica column (through Na2S04) and purified by silica gel chromatography (eluent : 5% MeOH/CH2CI2) which provided the desired product as an off-white solid after preciptation from EtOAc/MeOH. The final product was collected as
0.090 g (76%). LCMS E-S (M+H) 447.2. 1H NMR (400 MHz, DMSO-d6) δ ppm 1 1 .57 (s., 1 H), 9.06 (d, J=1.52 Hz, 1 H), 8.92 - 9.00 (m, 1 H), 8.39 - 8.48 (m, 2 H), 8.24 (s, 1 H), 8.14 (d, J=1.77 Hz, 1 H), 5.91 (s, 1 H), 5.30 - 5.45 (m, 1 H), 4.42 (d, J=4.80 Hz, 2 H), 3.97 (s, 3 H), 2.24 (s, 3 H), 2.13 (s, 3 H), 1 .55 (d, J=6.82 Hz, 6 H).
Example 59
W-[(4,6-Dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-6-[2- (methyloxy)-4-pyridinyl]-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000097_0001
The title compound was prepared in the same manner as described in example 58 using 2-methoxypyridine-4-boronic acid pinacol ester (69.2 mg, 0.294 mmol) to give the desired product as a grey solid after evaporation and preciptation from EtOAc/MeOH. The final product was collected as 0.051 g (41 %). LCMS E-S (M+H) 447.2. 1H NMR (400 MHz, DMSO-de) δ ppm 1 1.57 (br. s., 1 H) 9.00 (t, J=4.80 Hz, 1 H) 8.44 (s, 1 H) 8.36 (s, 1 H) 8.26 (s, 1 H) 7.84 (dd, J=5.31 , 1 .52 Hz, 1 H) 7.67 (s, 1 H) 5.91 (s, 1 H) 5.32 - 5.40 (m, 1 H) 4.42 (d, J=4.80 Hz, 2 H) 3.94 (s, 3 H) 2.23 (s, 3 H) 2.13 (s, 3 H) 1.55 (d, 6 H). Example 60
6-(6-Amino-3-pyridinyl)-A -[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000098_0001
The title compound was prepared in the same manner as described in example 58 using 6-aminopyridine-3-boronic acid pinacol ester (64.8 mg, 0.294 mmol) to give the desired product as a grey solid after evaporation and preciptation from EtOAc/MeOH. The final product was collected as 0.060 g (49%). LCMS E-S (M+H) = 432.2. 1H NMR (400 MHz, DMSO-de) δ ppm 1 1.57 (br. s., 1 H) 8.85 - 8.92 (m, 2 H) 8.31 (s, 1 H) 8.26 (dd,
J=8.59, 2.53 Hz, 1 H) 8.04 (s, 1 H) 6.57 (d, J=8.59 Hz, 1 H) 6.46 (s, 2 H) 5.90 (s, 1 H) 5.25 - 5.33 (m, 1 H) 4.40 (d, J=4.80 Hz, 2 H) 2.22 (s, 3 H) 2.13 (s, 3 H) 1.53 (d, 6 H).
Example 61
W-[(4,6-Dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-6-[5-
(methylsulfonyl)-3-pyridinyl]-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000098_0002
The title compound was prepared in the same manner as described in example 58 using [5-(methylsulfonyl)-3-pyridinyl]boronic acid (59.1 mg, 0.294 mmol) to give the desired product as an off-white solid after evaporation and preciptation from
EtOAc/MeOH. The final product was collected as 0.079 g (59%). LCMS E-S (M+H) = 495.0. 1H NMR (400 MHz, DMSO-d6) δ ppm 1 1 .59 (br. s., 1 H), 9.76 (d, J=2.27 Hz, 1 H), 9.21 (d, J=2.02 Hz, 1 H), 9.00 - 9.06 (m, 2 H), 8.46 (s, 1 H), 8.37 (s, 1 H), 5.91 (s, 1 H), 5.36 - 5.44 (m, 1 H), 4.43 (d, J=4.80 Hz, 2 H), 3.45 (s, 3 H), 2.24 (s, 3 H), 2.13 (s, 3 H), 1.56 (d, 6 H).
Example 62
W-[(4,6-Dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-6-(2-furanyl)-1 -(1
methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000099_0001
The title compound was prepared in the same manner as described in example 74 using 2-(2-furanyl)-4,4,5,5-tetramethyl-1 ,3,2-dioxaborolane (57.1 mg, 0.294 mmol) to give the desired product as an white solid after evaporation and preciptation from
EtOAc/MeOH. The final product was collected as 0.049 g (44%). LCMS E-S (M+H) 406.2. 1H NMR (400 MHz, DMSO-d6) δ ppm 1 1.57 (s, 1 H) 8.93 (t, J=4.93 Hz, 1 H) 8.33 (s, 1 H) 7.97 (s, 1 H) 7.94 (d, J=1.01 Hz, 1 H) 7.34 (d, J=2.78 Hz, 1 H) 6.73 (dd, J=3.54, 1.77 Hz, 1 H) 5.90 (s, 1 H) 5.19 - 5.32 (m, 1 H) 4.39 (d, J=4.80 Hz, 2 H) 2.23 (s, 3 H) 2.13 (s, 3 H) 1.52 (d, 6 H).
Example 63
A -[(4,6-Dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-6-{6- [(methylamino)carbonyl]-3-pyridinyl}-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4- carboxamide
Figure imgf000099_0002
The title compound was prepared in the same manner as described in example 58 using 2-(N-methylamidocarboxy)-5-pyridineboronic acid pinacol ester (70.1 mg, 0.267 mmol) to give the desired product as an off-white solid after evaporation and preciptation from EtOAc/MeOH. The final product was collected as 0.080 g (60%). LCMS E-S (M+H) = 474.1. 1H NMR (400 MHz, DMSO-d6) δ ppm 1 1.59 (br. s., 1 H), 9.46 (d, J=1.52 Hz, 1 H), 8.98 (t, J=4.55 Hz, 1 H), 8.85 (d, J=5.05 Hz, 1 H), 8.78 (dd, J=8.21 , 2.15 Hz, 1 H), 8.44 (s, 1 H), 8.31 (s, 1 H), 8.20 (d, J=8.08 Hz, 1 H), 5.91 (s, 1 H), 5.30 - 5.42 (m, 1 H), 4.42 (d, J=4.55 Hz, 2 H), 2.87 (d, J=4.80 Hz, 3 H), 2.23 (s, 3 H), 2.13 (s, 3 H), 1.56 (d, 6 H).
Example 64
6-[5-[(Cyclopropylsulfonyl)amino]-6-(methyloxy)-3-pyridinyl]-N-[(4,6-dimethyl-2-oxo- 1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4- carboxamide
Figure imgf000100_0001
The title compound was prepared in the same manner as described in example 58 using N-[2-(methyloxy)-5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)-3- pyridinyl]cyclopropanesulfonamide (95 mg, 0.267 mmol) to give the desired product as an off-white solid after evaporation and preciptation from EtOAc/MeOH. The final product was collected as 0.100 g (66%). LCMS E-S (M+H) 566.4. 1H NMR (400 MHz, DMSO-d6) δ ppm 1 1.57 (s, 1 H) 9.47 (s, 1 H) 8.94 (t, J=4.93 Hz, 1 H) 8.89 (d, J=2.27 Hz, 1 H) 8.49 (d, J=2.27 Hz, 1 H) 8.37 (s, 1 H) 8.15 (s, 1 H) 5.91 (s, 1 H) 5.29 (m, 1 H) 4.41 (d, J=4.80 Hz, 2 H) 4.01 (s, 3 H) 2.70 - 2.78 (m, 1 H) 2.23 (s, 3 H) 2.13 (s, 3 H) 1.56 (d, J=6.57 Hz, 6 H) 0.91 - 1.00 (m, 4 H).
Example 65
N-[(4,6-Dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-6-{5- [(phenylsulfonyl)amino]-3-pyridinyl}-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000100_0002
The title compound was prepared in the same manner as described in example 58 using N-[5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)-3-pyridinyl]benzenesulfonamide (96 mg, 0.267 mmol) to give the desired product as an off-whitesolid after evaporation and preciptation from EtOAc/MeOH. The final product was collected as 0.050 g (33%). LCMS E-S (M+H) = 572.2. 1H NMR (400 MHz, DMSO-d6) δ ppm 1 1 .57 (s, 1 H), 10.83 (s, 1 H), 9.10 (d, J=1.77 Hz, 1 H), 8.95 (t, J=5.05 Hz, 1 H), 8.37 - 8.45 (m, 2 H), 8.32 (t, J=2.15 Hz, 1 H), 8.17 (s, 1 H), 7.82 - 7.91 (m, 2 H), 7.55 - 7.69 (m, 3 H), 5.90 (s, 1 H), 5.18 - 5.30 (m, 1 H), 4.41 (d, J=5.05 Hz, 2 H), 2.22 (s, 3 H), 2.13 (s, 3 H), 1.58 (d, J=6.82 Hz, 6 H). Example 66
N-[(4,6-Dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-6-[2-(4- morpholinyl)-4-pyridinyl] ]pyridine-4-carboxamide
Figure imgf000101_0001
The title compound was prepared in the same manner as described in example 58 using 4-[4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)-2-pyridinyl]morpholine (78 mg, 0.267 mmol) to give the desired product as an off-white solid after evaporation and preciptation from EtOAc/MeOH. The final product was collected as 0.1 10 g (58%). LCMS E-S (M+H) 502.1. 1H NMR (400 MHz, DMSO-d6) δ ppm 1 1.57 (s, 1 H), 8.95 (t, J=5.05 Hz, 1 H), 8.40 (s, 1 H), 8.31 (d, J=5.30 Hz, 1 H), 8.18 (s, 1 H), 7.58 (s, 1 H), 7.51 (dd, J=5.31 , 1.26 Hz, 1 H), 5.90 (s, 1 H), 5.39-5.30 (m, 1 H), 4.42 (d, J=4.80 Hz, 2 H), 3.72 - 3.80 (m, 4 H), 3.54 - 3.62 (m, 4 H), 2.24 (s, 3 H), 2.13 (s, 3 H), 1.55 (d, J=6.57 Hz, 6 H).
Example 67
N-[(4,6-Dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-6-[6-(4- morpholinyl)-3-pyridiny pyridine-4-carboxamide
Figure imgf000102_0001
The title compound was prepared in the same manner as described in example 58 using 6-morpholinopyridine-3-boronic acid (55.6 mg 0.267 mmol) to give the desired product as an off-white solid after evaporation and preciptation from EtOAc/MeOH. The final product was collected as 0.1 10 g (82%). LCMS E-S (M+H) 502.1 . 1 H NMR (400 MHz, DMSO-de) δ ppm 1 1.57 (s, 1 H), 9.06 (d, J=2.27 Hz, 1 H), 8.88 (t, J=4.93 Hz, 1 H), 8.41 (dd, J=8.97, 2.40 Hz, 1 H), 8.33 (s, 1 H), 8.1 1 (s, 1 H), 7.00 (d, J=9.09 Hz, 1 H), 5.91 (s, 1 H), 5.34-5.28 (m, 1 H), 4.40 (d, J=4.80 Hz, 2 H), 3.71 - 3.78 (m, 4 H), 3.55 - 3.62 (m, 4 H), 2.23 (s, 3 H), 2.13 (s, 3 H), 1 .54 (d, J=6.82 Hz, 6 H).
Example 68
N-[(4,6-Dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-6-[6-
(methyloxy)-3-pyridinyl]-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000102_0002
The title compound was prepared in the same manner as described in example 58 using [6-(methyloxy)-3-pyridinyl]boronic acid ( 40.9 mg 0.267 mmol) to give the desired product as an off-white solid after evaporation and preciptation from EtOAc/MeOH. The final product was collected as 0.060 g (50%). LCMS E-S (M+H) 447.2. 1H NMR (400 MHz, DMSO-de) δ ppm 1 1.58 (s, 1 H), 9.07 (d, J=2.02 Hz, 1 H), 8.90 (t, J=4.93 Hz, 1 H), 8.55 (dd, J=8.59, 2.53 Hz, 1 H), 8.37 (s, 1 H), 8.17 (s, 1 H), 7.01 (d, J=8.84 Hz, 1 H), 5.91 (s, 1 H), 5.36-5.28 (m, 1 H), 4.41 (d, J=5.05 Hz, 2 H), 3.95 (s, 3 H), 2.23 (s, 3 H), 2.13 (s, 3 H), 1 .55 (d, J=6.57 Hz, 6 H). Example 69
6-[6-(Acetylamino)-3^yridinyl]-N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3- pyridinyl)methyl]-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000103_0001
The title compound was prepared in the same manner as described in example 58 using 2-acetamidopyridine-5-boronic acid pinacol ester (77 mg, 0.294 mmol) to give the desired product as an off-white solid after evaporation and preciptation from
EtOAc/MeOH. The final product was collected as 0.065 g (51 %). LCMS E-S (M+H) = 474.0. 1 H NMR (400 MHz, DMSO-d6) δ ppm 1 1 .57 (s, 1 H), 10.76 (s, 1 H), 9.19 (d, J=1.77 Hz, 1 H), 8.93 (t, J=4.80 Hz, 1 H), 8.61 (dd, J=8.84, 2.27 Hz, 1 H), 8.38 (s, 1 H), 8.25 (d, J=8.84 Hz, 1 H), 8.20 (s, 1 H), 5.91 (s, 1 H), 5.30 - 5.40 (m, 1 H), 4.42 (d, J=5.05 Hz, 2 H), 2.23 (s, 3 H), 2.14 (s, 3 H), 2.13 (s, 3 H), 1.54 (d, J=6.82 Hz, 6 H).
Example 70
W-[(4,6-Dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-6-(2- methylphenyl)-1 H-py arboxamide
Figure imgf000103_0002
The title compound was prepared in the same manner as described in example 58 using 2-methylphenyl boronic acid (40.0 mg, 0.294 mmol) to give the desired product as an off-white solid after evaporation and preciptation from MeOH. The final product was collected as 0.065 g (51 %). LCMS E-S (M+H) = 430.2. 1 H NMR (400 MHz, DMSO-d6) δ ppm 1 1 .54 (s, 1 H) 8.89 (t, J=4.93 Hz, 1 H) 8.39 (s, 1 H) 7.74 (s, 1 H) 7.56 (d, J=7.07 Hz, 1 H) 7.31 - 7.41 (m, 3 H) 5.89 (s, 1 H) 5.20 - 5.27 (m, 1 H) 4.38 (d, J=4.80 Hz, 2 H) 2.40 (s, 3 H) 2.21 (s, 3 H) 2.12 (s, 3 H) 1.53 (d, 6 H). Example 71
methyl -2-OXO-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-6-(1 H- pyrazol-4-yl)-1 H-pyrazolo 3,4-b]pyridine-4-carboxamide
Figure imgf000104_0001
To a 5-mL microwave vial were added 6-chloro-N-[(4,6-dimethyl-2-oxo-1 ,2- dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxami
(70 mg, 0.187 mmol), 4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)-1 H-pyrazole (47.2 mg, 0.243 mmol), DMSO(1 .5 mL) and sodium carbonate (0.281 mL, 0.562 mmol), and the mixture was degassed with nitrogen for 5 min. Next added bis(triphenylphosphine)
palladium(l l) chloride (10.51 mg, 0.015 mmol) and the vial was sealed. The mixture was irradiated (microwave) at 140 °C for 12h. The reaction mixture was filtered and the
residue was washed with DMSO. The DMSO solution of crude product was purified using reverse-phase HPLC. The TFA salt of the product obtained was neutralized with
saturated NaHC03, washed with water, and dried under high vacuum to give 12 mg (16%) of product. LCMS: (M+H)+=406.2 1 H NMR (400 MHz, DMSO-d6) δ ppm 1 .45 - 1 .60 (m, J = 6.4 Hz, 6 H), 2.13 (s, 3 H), 2.23 (s, 3 H), 4.39 (d, J = 4.6 Hz, 2 H), 5.27 (quin, J = 6.7 Hz, 1 H), 5.90 (s, 1 H), 7.91 (s, 1 H), 8.22 - 8.31 (m, 1 H), 8.33 (m, 2 H), 8.77 (t, J = 4.7 Hz, 1 H).
Example 72
6-(2-Amino-5-pyrimidinyl)-A -[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 - methyleth l)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000104_0002
To a 5-mL microwave vial was added DMSO (2 mL) and it was degassed with
nitrogen for 5 min. Next added 6-chloro-N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3- pyridinyl)methyl]-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide (70 mg,
0.187 mmol), 5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)-2-pyrimidinamine (49.7 mg,
0.225 mmol), sodium carbonate (59.5 mg, 0.562 mmol) and
bis(triphenylphosphine)palladium(l l) chloride (9.20 mg, 0.013 mmol). The mixture was degassed for additional 5 min, sealed, and irradiated (microwave) at 135 °C for 15h. The mixture was filtered and the residue was washed with DMSO. The DMSO solution of the crude product was purified using reverse-phase HPLC. The TFA salt of the product was neutralized with saturated NaHC03 solution, filtered, washed with water, and dried under high vacuum to give 14 mg (17%) of product. LCMS E-S (M+H) = 433.2 1 H NMR (400
MHz, DMSO-de) δ ppm 1 .52 (d, J = 6.4 Hz, 6 H), 2.13 (s, 3 H), 2.22 (s, 3 H), 4.40 (d, J =
4.6 Hz, 2 H) 5.22 - 5.41 (m, 1 H), 5.90 (s, 1 H), 7.18 (m, 2 H), 8.08 (s, 1 H), 8.34 (s, 1 H), 8.86 (m, 1 H), 9.1 1 (s, 2 H), 1 1 .58 (br. s., 1 H).
Example 73
W-[(4,6-Dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-6-(3-pyridinyl)-1 H- p razolo[3,4-b]pyridine-4-carboxamide
Figure imgf000105_0001
To a 5-mL microwave vial were added 6-chloro-N-[(4,6-dimethyl-2-oxo-1 ,2- dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
(70 mg, 0.187 mmol), 3-pyridinylboronic acid (29.9 mg, 0.243 mmol), DMSO (1 .5 mL) and sodium carbonate (0.281 mL, 0.562 mmol), and the mixture was degassed with nitrogen for 10 min. Next was added bis(triphenylphosphine)palladium(l l) chloride (10.51 mg,
0.015 mmol) and the vial was sealed. The reaction mixture was irradiated (microwave) at 140 °C overnight. The reaction mixture was filtered and the residue was washed with
DMSO. The crude product in DMSO was purified using reverse-phase HPLC. The TFA salt of the product was neutralized using saturated NaHC03, filtered, washed with water and dried under high vacuum to give 33 mg (42%) of product. LCMS E-S (M+H) = 417.2
1 H NMR (400 MHz, DMSO-d6) δ ppm 1 .56 (d, J = 6.4 Hz , 6 H), 2.13 (s, 3 H), 2.23 (s, 3 H), 4.42 (d, J = 4.29 Hz, 2 H), 5.26 - 5.43 (m, 1 H), 5.91 (s, 1 H), 7.62 (dd, J = 7.3, 5.0 Hz, 1 H), 8.26 (s, 1 H), 8.43 (s, 1 H), 8.64 (d, J = 7.8 Hz, 1 H), 8.70 - 8.79 (m, 1 H), 8.96 (br.
s., 1 H), 9.46 (br. s., 1 H), 1 1 .59 (br. s., 1 H). Example 74
W-[(4,6-Di methyl -2 -oxo-1 ,2-dihydro-3-pyridinyl)methyl]-6-(1 H-indazol-5-yl)-1 -(1 - methyleth l)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000106_0001
To a 5-mL microwave vial were added 6-chloro-N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3- pyridinyl)methyl]-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide (70 mg, 0.187 mmol), 5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)-1 H-indazole (59.4 mg, 0.243 mmol), DMSO(1.5 ml.) and sodium carbonate (0.281 ml_, 0.562 mmol), and the mixture was degassed with nitrogen for 10 min. Next was added bis(triphenylphosphine)palladium(ll) chloride (10.51 mg, 0.015 mmol). The contents were sealed and irradiated (microwave) at 140 °C overnight. The mixture was filtered and the residue was washed with DMSO. The crude product in DMSO was purified using reverse-phase HPLC. The TFA salt of the product was neutralized using saturated NaHC03, filtered, washed with water and dried under high vacuum to give 41 mg (48%) of product. LCMS E-S (M+H) = 456.0 1 H NMR (400 MHz, DMSO-d6) δ ppm 1 .56 (d, J = 6.8 Hz , 6 H), 2.14 (s, 3 H), 2.23 (s, 3 H), 4.38 - 4.53 (m, 2 H), 5.27 - 5.49 (m, 1 H), 5.91 (s, 1 H), 7.59 - 7.75 (m, 1 H), 8.24 (s, 2 H), 8.37 (s, 2 H), 8.69 (s, 1 H), 8.95 - 9.09 (m, 1 H), 1 1 .49 - 1 1.67 (m, 1 H), 13.21 - 13.35 (m, 1 H).
Example 75
W-[(4,6-Di methyl -2 -oxo-1 ,2-dihydro-3-pyridinyl)methyl]-6-(1 H-indazol-6-yl)-1 -(1 - methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000107_0001
The title compound was prepared in the same manner as described in example 74 from 6-chloro-N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-1 H- pyrazolo[3,4-b]pyridine-4-carboxamide (70 mg, 0.187 mmol), 6-(4,4,5,5-tetramethyl-1 ,3,2- dioxaborolan-2-yl)-1 H-indazole (59.4 mg, 0.243 mmol), DMSO(1 .5 mL), sodium carbonate (0.281 mL, 0.562 mmol), and bis(triphenylphosphine)palladium(l l) chloride (10.51 mg,
0.015 mmol). The final product was collected as 55 mg (65%). LCMS E-S (M+H) = 456.0 1 H NMR (400 MHz, DMSO-d6) δ ppm 1 .49 - 1 .65 (m, J = 6.4 Hz, 6 H), 2.14 (s, 3 H), 2.23 (s, 3 H), 4.43 (d, J = 4.6 Hz, 2 H), 5.36 (quin, J = 6.7 Hz, 1 H), 5.87 - 5.97 (m, 1 H), 7.52 - 7.72 (m, 2 H) 7.92 (d, J = 8.6 Hz, 1 H), 8.08 (d, J = 8.6 Hz, 1 H), 8.16 (s, 1 H), 8.26 (s, 1 H) , 8.37 - 8.49 (m, 2 H), 8.96 - 9.14 (m, 1 H), 1 1 .59 (br. s., 1 H).
Example 76
6-(1 H-1 ,2,3-Benzotriazol-5-yl)-W-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 - methyleth l)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000107_0002
The title compound was prepared in the same manner as described in example 74 using 6-chloro-N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)- 1 H-pyrazolo[3,4-b]pyridine-4-carboxamide (70 mg, 0.187 mmol), 1 H-1 ,2,3-benzotriazol-5- ylboronic acid (39.7 mg, 0.243 mmol), DMSO(2 mL), sodium carbonate (0.281 mL, 0.562 mmol) and bis(triphenylphosphine)palladium(ll) chloride (10.51 mg, 0.015 mmol). The final product was collected as 22 mg (26%). LCMS E-S (M+H) = 457.1 1H NMR (400 MHz, DMSO-de) δ ppm 1.57 (d, 6 J = 6.4 Hz, H), 2.14 (s, 3 H), 2.24 (s, 3 H), 4.43 (d, J = 4.3 Hz, 2 H), 5.31 - 5.46 (m, 1 H), 5.91 (s, 1 H), 8.07 (br. s., 1 H), 8.34 (s, 1 H), 8.41 (m, 2 H), 8.85 (br. s., 1 H), 9.02 (br. s., 1 H), 1 1.59 (br. s., 1 H).
Example 77
W-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-3-methyl-1 -(1 -methylethyl)-6-(2- 2,3-dihydro-1H-benzimidazol-5- l)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000108_0001
The title compound was prepared in the same manner as described in example 74 using 6-chloro-N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)- 1 H-pyrazolo[3,4-b]pyridine-4-carboxamide (70 mg, 0.187 mmol), 5-(4,4,5,5-tetramethyl- 1 ,3,2-dioxaborolan-2-yl)-1 ,3-dihydro-2H-benzimidazol-2-one (61 .0 mg, 0.235 mmol), DMSO(1.5 mL) sodium carbonate (0.281 mL, 0.562 mmol), and
bis(triphenylphosphine)palladium(ll) chloride (10.13 mg, 0.014 mmol). The final product was collected as 19 mg (21 %). LCMS E-S (M+H) = 486.3 1H NMR (400 MHz, DMSO-d6) δ ppm 1 .50 (d, J = 6.8 Hz, 6 H), 2.12 (s, 3 H), 2.24 (s, 3 H), 2.43 (s, 3 H), 4.39 (d, J = 4.6 Hz, 2 H), 5.15 - 5.35 (m, 1 H), 5.89 (s, 1 H), 7.05 (d, J = 8.1 Hz, 1 H), 7.61 (s, 1 H), 7.77 - 7.93 (m, 2 H), 8.70 - 8.85 (m, 1 H), 10.79 (br. s., 1 H), 10.87 (br. s., 1 H), 1 1 .54 (br. s., 1 H).
Example 78
W-[(4,6-Dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-6-(2- 2,3-dihydro-1 H-indol-6- l)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000109_0001
The title compound was prepared in the same manner as described in example 74 using 6-chloro-N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)- 1 H-pyrazolo[3,4-b]pyridine-4-carboxamide (70 mg, 0.187 mmol), 6-(4,4,5,5-tetramethyl- 1 ,3,2-dioxaborolan-2-yl)-1 ,3-dihydro-2H-indol-2-one (63.1 mg, 0.243 mmol), DMSO(1 .5 mL), sodium carbonate (0.281 mL, 0.562 mmol), and bis(triphenylphosphine)palladium(l l) chloride (10.51 mg, 0.015 mmol). The final product was collected as 10 mg (1 1 %). LCMS E-S (M+H) = 471 .1 1 H NMR (400 MHz, DMSO-d6) δ ppm 1 .55 (d, J = 6.8 Hz, 6 H), 2.13 (s, 3 H), 2.22 (s, 3 H), 3.58 (s, 2 H), 4.41 (d, J = 4.8 Hz, 2 H), 5.31 (quin, J = 6.6 Hz, 1 H), 5.91 (s, 1 H), 7.38 (d, J = 7.6 Hz, 1 H), 7.73 (d, J = 1 .0 Hz, 1 H), 7.87 (dd, J=7.8, 1 .5 Hz, 1 H), 8.12 (s, 1 H), 8.37 (s, 1 H), 9.00 (t, J = 4.8 Hz, 1 H), 10.55 (s, 1 H), 1 1 .58 (s, 1 H).
Example 79
W-[(4,6-Dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-6-(2- 2,3-dihydro-1 H-indol-5- l)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000109_0002
The title compound was prepared in the same manner as described in example 74 using 6-chloro-N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)- 1 H-pyrazolo[3,4-b]pyridine-4-carboxamide (70 mg, 0.187 mmol), 5-(4,4,5,5-tetramethyl- 1 ,3,2-dioxaborolan-2-yl)-1 ,3-dihydro-2H-indol-2-one (63.1 mg, 0.243 mmol), DMSO(1 .5 mL), sodium carbonate (0.281 mL, 0.562 mmol), and bis(triphenylphosphine)palladium(ll) chloride (10.51 mg, 0.015 mmol). The final product was collected as 30 mg (34%). LCMS E-S (M+H) = 471.3 1 H NMR (400 MHz, DMSO-d6) δ ppm 1.54 (d, J = 6.4 Hz, 6 H), 2.13 (s, 3 H), 2.22 (s, 3 H), 3.40 (br. s., 2 H), 4.41 (d, J = 4.6 Hz, 2 H), 5.25 - 5.41 (m, 1 H), 5.91 (s, 1 H), 6.97 (d, J = 8.1 Hz, 1 H), 8.08 - 8.21 (m, 3 H), 8.34 (s, 1 H), 8.96 (br. s., 1 H), 10.64 (s, 1 H), 1 1 .58 (br. s., 1 H).
Example 80
W-[(4,6-Dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-6-(2-oxo-2,3- dihydro-1 H-benzimidazol-5- l)-1H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000110_0001
The title compound was prepared in the same manner as described in example 74 using 6-chloro-N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)- 1 H-pyrazolo[3,4-b]pyridine-4-carboxamide (70 mg, 0.187 mmol), 5-(4,4,5,5-tetramethyl- 1 ,3,2-dioxaborolan-2-yl)-1 ,3-dihydro-2H-benzimidazol-2-one (63.3 mg, 0.243 mmol),
DMSO(2 mL), sodium carbonate (0.281 mL, 0.562 mmol), and
bis(triphenylphosphine)palladium(ll) chloride (10.51 mg, 0.015 mmol). The final product was collected as 35 mg (40%). LCMS E-S (M+H) = 472.4 1H NMR (400 MHz, DMSO-d6) δ ppm 1 .55 (d, J = 6.4 Hz, 6 H), 2.13 (s, 3 H), 2.22 (s, 3 H), 4.41 (d, J = 4.8 Hz, 2 H), 5.30 (quin, J = 6.6 Hz, 1 H), 5.91 (s, 1 H), 7.07 (d, J = 8.3 Hz, 1 H), 7.86 (s, 1 H), 7.92 (dd, J = 8.3, 1 .8 Hz, 1 H), 8.10 (s, 1 H), 8.30 - 8.41 (m, 1 H), 8.98 (t, J = 4.8 Hz, 1 H), 10.82 (s, 1 H), 10.89 (s, 1 H). 1 1 .58 (s, 1 H).
Example 81
W-[(4,6-Dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-6-[6-(4-methyl-1 - piperazinyl)-3-p ridinyl]-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000111_0001
The title compound was prepared in the same manner as described in example 74 using 6-chloro-N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)- 1 H-pyrazolo[3,4-b]pyridine-4-carboxamide (70 mg, 0.187 mmol), 1 -methyl-4-[5-(4,4,5,5- tetramethyl-1 ,3,2-dioxaborolan-2-yl)-2-pyridinyl]piperazine (73.8 mg, 0.243 mmol),
DMSO(2 ml_), sodium carbonate (0.281 mL, 0.562 mmol), and
bis(triphenylphosphine)palladium(l l) chloride (10.51 mg, 0.015 mmol). The final product was collected as 12 mg (13%). LCMS E-S (M+H) = 515.1 1 H NMR (400 MHz, DMSO-d6) δ ppm 1 .53 (d, J = 6.4 Hz , 6 H), 2.13 (s, 3 H), 2.23 (m, 6 H), 2.37 - 2.46 (m, 4 H), 3.56 - 3.69 (m, 4 H), 4.40 (d, J = 4.6 Hz, 2 H), 5.30 (quin, J = 6.7 Hz, 1 H), 5.91 (s, 1 H), 6.99 (d,
J = 9.1 Hz, 1 H), 8.10 (s, 1 H), 8.32 (s, 1 H), 8.38 (dd, J = 9.1 , 2.5 Hz, 1 H), 8.91 (br. s., 1 H), 9.03 (d, J=2.3 Hz, 1 H).
Example 82
W-[(4,6-Dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-6-[6-(4- methyl-1 -piperazin l)-3-pyridinyl]-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000111_0002
The title compound was prepared in the same manner as described in example 74 using 6-chloro-N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)- 1 H-pyrazolo[3,4-b]pyridine-4-carboxamide (70 mg, 0.187 mmol), 1 -methyl-4-[4-(4,4,5,5- tetramethyl-1 ,3,2-dioxaborolan-2-yl)phenyl]piperazine (73.6 mg, 0.243 mmol), 1 ,2- Dimethoxyethane (DME) (3 mL), water (1 mL) sodium carbonate (0.281 mL, 0.562 mmol), and bis(triphenylphosphine)palladium(ll) chloride (12.23 mg, 0.015 mmol), wherein the reaction time was 30 min. The crude product was purified by column chromatography (eluent: gradient of 0 to 15% (9 :1 MeOH/NH4OH)/DCM). The product was dried under high vacuum and collected as 58 mg (59%). LCMS E-S (M+H) = 514.3 1 H NMR (400 MHz, DMSO-de) δ ppm 1.51 - 1.58 (m, 6 H), 2.13 (s, 3 H), 2.23 (s, 6 H), 2.24 (s, 3H), 2.47 (m, 4H), 3.24 - 3.30 (m, 4 H), 4.41 (d, J = 5.05 Hz, 2 H), 5.30 (quin, J = 6.63 Hz, 1 H), 5.90 (s, 1 H), 7.07 (d, J = 9.09 Hz, 2 H), 8.06 (s, 1 H), 8.15 (d, J = 8.84 Hz, 2H), 8.30 (s, 1 H), 8.92 (t, J = 4.80 Hz, 1 H), 1 1 .55 (s, 1 H).
Example 83
W-[(4,6-Dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-3-methyl-1 -(1 -methylethyl)-6-
[6-(4-morpholinyl)-3-p ridinyl]-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000112_0001
The title compound was prepared in the same manner as described in example 74 using 6-chloro-N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-3-methyl-1 -(1 - methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide (70 mg, 0.180 mmol), [6-(4- morpholinyl)-3-pyridinyl]boronic acid (48.8 mg, 0.235 mmol), DMSO(2.0 mL), sodium carbonate (0.271 mL, 0.541 mmol), and bis(triphenylphosphine)palladium(ll) chloride (10.13 mg, 0.014 mmol), wherein the reaction time was 8 h. The mixture was filtered and the residue was washed with DMSO. The crude product in DMSO was purified by reverse- phase HPLC (mobile phase: 25 - 60 % CAN in H20, 0.1 %TFA). The TFA product salt obtained was neutralized with saturated NaHC03, filtered, washed with water, and dried under high vacuum to give the product as 24 mg (25%). LCMS E-S (M+H) = 516.2 1H NMR (400 MHz, DMSO-d6) δ ppm 1 .50 (d, J = 6.57 Hz, 6 H), 2.12 (s, 3 H), 2.25 (s, 3 H), 2.42 (s, 3 H), 3.54 - 3.63 (m, 4 H), 3.73 (m, 4 H), 4.39 (d, J= 4.80 Hz, 2 H), 5.14 - 5.29 (m, 1 H), 5.89 (s, 1 H), 6.97 (d, J = 8.84 Hz, 1 H), 7.63 (s, 1 H), 8.37 (dd, J = 9.09, 2.53 Hz, 1 H), 8.68 (t, J = 4.93 Hz, 1 H), 8.98 (d, J = 2.27 Hz, 1 H), 1 1 .51 (s, 1 H).
Example 84
W-[(4,6-Dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-6-(4- pyridinylamino -1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000113_0001
The title compound was prepared in the same manner as described in example 74 using 6-chloro-N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)- 1 H-pyrazolo[3,4-b]pyridine-4-carboxamide (50 mg, 0.134 mmol), 4-pyridinamine (12.59 mg, 0.134 mmol), cesium carbonate (131 mg, 0.401 mmol), 1 ,4-dioxane (2 ml_),
palladium(ll) acetate (1 .501 mg, 6.69 mol) and BINAP (8.33 mg, 0.013 mmol), wherein the reaction time was 2h. The final product was collected as 29 mg (50%). LCMS E-S (M+H) = 432.3 1H NMR (400 MHz, DMSO-d6) δ ppm 1 .45 - 1 .59 (d, J = 6.8 Hz, 6 H), 2.14 (s, 3 H), 2.23 (s, 3 H), 4.36 (d, J = 5.0 Hz, 2 H), 5.16 (m, 1 H), 5.91 (s, 1 H), 7.09 (s, 1 H), 7.99 (d, J = 6.1 Hz, 2 H), 8.12 (s, 1 H) 8.49 (d, J = 6.6 Hz, 2 H), 8.74 (t, J = 5.1 Hz, 1 H), 10.58 (s, 1 H), 1 1 .58 (s, 1 H).
Example 85
W-[(4,6-Dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-6-[(2-oxo-2, dihydro-1 H-benzimidazol-5- l)amino]-1H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000113_0002
The title compound was prepared in the same manner as described in example 74 using 6-chloro-N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)- 1 H-pyrazolo[3,4-b]pyridine-4-carboxamide (70 mg, 0.187 mmol), 5-amino-1 ,3-dihydro-2H- benzimidazol-2-one (33.5 mg, 0.225 mmol), cesium carbonate (92 mg, 0.281 mmol), N,N- dimethylacetamide (DMA) (2 ml_), palladium(ll) acetate (2.52 mg, 0.01 1 mmol) and Xantphos (10.83 mg, 0.019 mmol) wherein the reaction temperature was 150 °C and reaction time was 8h. The final product was collected as 25 mg (27%). LCMS E-S (M+H) = 487.2 1H NMR (400 MHz, DMSO-de) δ ppm 1 .50 - 1.61 (m, 6 H), 2.13 (s, 3 H), 2.23 (s, 3 H), 4.38 (d, J = 5.0 Hz, 2 H), 5.20 (quin, J = 6.6 Hz, 1 H), 5.90 (s, 1 H), 6.79 - 6.91 (m, 2 H), 8.04 (d, J = 8.3 Hz, 1 H), 8.26 - 8.33 (m, 2 H), 8.84 (t, J = 5.0 Hz, 1 H), 1 1.50 (s, 1 H).
Example 86
6-{[4-(Aminocarbonyl)phenyl]amino}-A -[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3- pyridinyl)methyl]-1 -(1 -meth lethyl)-1 H-pyrazolo[3,4-fo]pyridine-4-carboxamide
Figure imgf000114_0001
The title compound was prepared in the same manner as described in example 74 using 6-chloro-N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-3-methyl-1 -(1 - methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide (70 mg, 0.180 mmol), 4- aminobenzamide (29.5 mg, 0.217 mmol), cesium carbonate (88 mg, 0.271 mmol), N,N- dimethylacetamide (DMA) (1.5 mL), palladium(ll) acetate (2.431 mg, 10.83 μηηοΙ) and Xantphos (10.44 mg, 0.018 mmol) wherein the reaction temperature was 150 °C and reaction time was 1 h. The final product was collected as 12 mg (14%). LCMS E-S (M+H) = 488.2 1H NMR (400 MHz, DMSO-d6) δ ppm 1 .48 (d, J = 6.4 Hz, 6 H), 2.13 (s, 3 H),
2.23 (s, 3 H), 2.32 (s, 3 H), 4.35 (d, J = 4.8 Hz, 2 H), 4.99 (quin, J = 6.8 Hz, 1 H), 5.89 (s, 1 H), 6.65 (s, 1 H), 7.15 (br. s., 1 H), 7.75 - 7.82 (m, 1 H), 7.84 - 7.96 (m, 4 H), 8.56 (t, J = 4.9 Hz, 1 H), 9.77 (s, 1 H). Example 87
1 -(1 ,1 -Dimethylethyl)-A -[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-3- methyl-6-[6-(4-methyl-1 -piperazinyl)-3-pyridinyl]-1H-pyrazolo[3,4-b]pyridine-4- carboxamide
Figure imgf000115_0001
The title compound was prepared in the same manner as described in example 74 using 6-chloro-1-(1 , 1 -dimethylethyl)-N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3- pyridinyl)methyl]-3-methyl-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide (70 mg, 0.174 mmol), 1-methyl-4-[5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)-2-pyridinyl]piperazine (68.7 mg, 0.226 mmol), DME (3 mL), water (1 .00 mL), sodium carbonate (0.261 mL, 0.523 mmol) and PdCl2(dppf)-CH2Cl2 adduct (1 1 .38 mg, 0.014 mmol), wherein the reaction time was 40 min. The crude product was purified by column chromatography (eluent: gradient of 0 to 15% (9:1 MeOH/NH4OH)/DCM). The final product was collected as a solid, 41 mg (43%).. LCMS E-S (M+H) = 543.2 1 H NMR (400 MHz, DMSO-d6) δ ppm 1 .78 (s, 9 H), 2.12 (s, 3 H), 2.24 (m, 7 H), 2.38 (s, 3 H), 2.39 - 2.49 (m, 4 H), 3.34 (s, 3 H), 4.38 (m, 2 H), 5.88 (s, 1 H), 6.98 (d, J = 9.09 Hz, 1 H), 7.59 (s, 1 H), 8.93 - 8.97 (m, 1 H), 1 1.49 - 1 1.57 (m, 1 H), 1 1 .53 (s, 1 H), 1 1.53 (s, 1 H).
Example 88
1 -(1 -Dimethylethyl)-W-[(4,6-dimethyl-2-oxo- oxo-2,3-dihydro-1 H-benzimidazol-5- l)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000115_0002
The title compound was prepared in the same manner as described in example 74 using 6-chloro-1-(1 -dimethylethyl)-N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-3-m pyrazolo[3,4-b]pyridine-4-carboxamide (70 mg, 0.174 mmol), 5-(4,4,5,5-tetramethyl-1 ,3,2- dioxaborolan-2-yl)-1 ,3-dihydro-2H-benzimidazol-2-one (58.9 mg, 0.226 mmol), DME (3 ml_), water (1 .00 ml_), sodium carbonate (0.261 ml_, 0.523 mmol) and PdCI2(dppf)-CH2CI2 adduct
(1 1.38 mg, 0.014 mmol) wherein the reaction time was 40 min. The crude product was purified by column chromatography (eluent: gradient of 0 to 15% (9:1 MeOH/NH4OH)/DCM). The final product was collected as a solid, 24 mg (27%). LCMS E-S (M+H) = 500.2 1 H NMR (400 MHz, DMSO-de) δ ppm 1 .77 (m, 9 H), 2.12 (s, 3 H), 2.24 (s, 3 H), 2.40 (s, 3 H), 4.38 (d, J = 4.55 Hz, 2 H), 5.89 (s, 1 H), 7.06 (d, J = 8.08 Hz, 1 H), 7.59 (s, 1 H), 7.72 - 7.86 (m, 2 H), 8.73 (br. s., 2 H), 10.74 - 10.92 (m, 1 H), 1 1 .53 (br. s., 1 H).
Example 89
1 -(1 ,1 -Dimethylethyl)-W-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-3-methyl-6-[6-
(4-morpholin l)-3-pyridinyl]-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000116_0001
The title compound was prepared in the same manner as described in example 74 using 6-chloro-1-(1 , 1-dimethylethyl)-N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-3-methyl-1 H- pyrazolo[3,4-b]pyridine-4-carboxamide (70 mg, 0.174 mmol), [6-(4-morpholinyl)-3- pyridinyl]boronic acid (47.1 mg, 0.226 mmol), DME (3 ml_), water (1 .00 ml_), sodium carbonate (0.261 ml_, 0.523 mmol) and PdCI2(dppf)-CH2CI2 adduct (1 1.38 mg, 0.014 mmol) wherein the reaction time was 40 min. The crude product was purified by column chromatography (eluent: gradient of 0 to 15% (9:1 MeOH/NH4OH)/DCM). The final product was collected as a solid, 61 mg (65%). LCMS E-S (M+H) = 530.0 1H NMR (400 MHz, DMSO-d6) δ ppm 1.78 (s, 9 H), 2.12 (s, 3 H), 2.24 (s, 3 H), 2.38 (s, 3 H), 3.52 - 3.64 (m, 4 H), 3.68 - 3.80 (m, 4 H), 4.38 (d, J = 5.05 Hz, 2 H), 5.88 (s, 1 H), 6.99 (d, J = 9.09 Hz, 1 H), 7.61 (s, 1 H), 8.34 (dd, J=8.84, 2.53 Hz, 1 H), 8.68 (t, J = 5.05 Hz, 1 H), 8.97 (d, J = 2.27 Hz, 1 H), 1 1 .53 (s, 1 H). Example 90
6-(2 ,3-Benzoxadiazol-5-yl)-A -[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3^yridinyl)m
methyl-1 -(1 -meth lethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000117_0001
The title compound was prepared in the same manner as described in example 74 using 6-chloro-N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-3-methyl-1 -(1 - methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide (70 mg, 0.180 mmol), 5-(4,4,5,5- tetramethyl-1 ,3,2-dioxaborolan-2-yl)-2, 1 ,3-benzoxadiazole (57.7 mg, 0.235 mmol),
DMSO(2 mL), sodium carbonate (0.271 ml_, 0.541 mmol), and
bis(triphenylphosphine)palladium(l l) chloride (12.67 mg, 0.018 mmol) wherein the reaction time was 8h. The final product was collected as 17 mg (20%). LCMS LCMS E-S (M+H) = 472.4 1 H NMR (400 MHz, DMSO-d6) δ ppm 1 .53 (d, J = 6.8 Hz ,6 H), 2.12 (s, 3 H), 2.27
(s, 3 H) 4.42 (d, J = 4.8 Hz, 2 H) 5.32 (quin, J = 6.6 Hz, 1 H), 5.90 (s, 1 H), 8.00 (s, 1 H),
8.23 (d, J = 9.4 Hz, 1 H), 8.54 - 8.64 (m, 1 H), 8.79 (t, J = 4.7 Hz, 1 H), 8.91 (s, 1 H),
1 1 .54 (br. s., 1 H).
Example 91
6-(2-Amino-6-quinazolinyl)-A -[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-3-methyl- 1 -(1 -methyleth l)-1 H-pyrazolo[3,4-fo]pyridine-4-carboxamide
Figure imgf000117_0002
The title compound was prepared in the same manner as described in example 74 using 6-chloro-N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-3-methyl-1 -(1 - methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide (70 mg, 0.180 mmol), 5-(4, 4,5,5- tetramethyl-1 ,3,2-dioxaborolan-2-yl)-2,1 ,3-benzoxadiazole (57.7 mg, 0.235 mmol), DMSO(2 ml_), sodium carbonate (0.271 ml_, 0.541 mmol), and
bis(triphenylphosphine)palladium(ll) chloride (12.67 mg, 0.018 mmol), wherein the reaction time was 8h. The final product was collected as 28 mg (31 %). LCMS E-S (M+H) = 497.3 1H NMR (400 MHz, DMSO-d6) δ ppm 1 .52 (d, J = 6.8 Hz , 6 H), 2.12 (s, 3 H), 2.26 (s, 3 H), 4.41 (d, J = 5.0 Hz, 2 H) 5.30 (quin, J = 6.6 Hz, 1 H), 5.89 (s, 1 H), 7.16 (br. s., 2 H), 7.56 (d, J = 9.1 Hz, 1 H), 7.78 (s, 1 H) 8.60 (dd, J = 8.8, 2.0 Hz, 1 H), 8.69 (d, J = 1.8 Hz, 1 H), 8.76 (t, J = 4.9 Hz, 1 H), 9.28 (s, 1 H), 1 1 .53 (s, 1 H).
Example 92
A -[(4,6-Dimethyl-2-oxo-1 ,2-dihydro-3^yridinyl)methyl]-3-methyl-6-{4- [(methylamino)sulfonyl]phenyl}-1 -(1 -methylethyl)-1H-pyrazolo[3,4-b]pyridine-4- carboxamide
Figure imgf000118_0001
6-Chloro-N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-3-methyl-1 -(1 - methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide (80 mg, 0.21 mmol), {4- [(methylamino)sulfonyl]phenyl}boronic acid (66.5 mg, 0.31 mmol) and
bis(triphenylphosphine)palladium(ll) chloride (8.4 mg, 0.01 mmol) were suspended in DME/water (4 ml_, 3:1 ) and stirred for 10 min under nitrogen at room temperature.
Sodium bicarbonate (52 mg, 0.62 mmol) was added and the heterogenous mixture was irradiated (microwave) at 150 °C for 30 min. After cooling to room temperature, water was added to the black mixture, and the contents were vacuum filtered. The crude product was dissolved in DCM/MeOH (1 :1 ) and preabsorbed onto silica gel. The product was purified by silica gel chromatography (eluent: DCM/MeOH/NH^H, gradient of 0 to 90:10:1 ). The light beige solid that was collected was suspended in EtOH, sonicated, and filtered. The solid was then air-dried for 15 min, and then in vacuum oven overnight. The final product was collected as 77mg (70%). LCMS E-S (M+H) = 523.2 1H NMR (400 MHz, DMSO-de) δ ppm 1 1 .54 (br. s., 1 H) 8.78 (t, J=5.05 Hz, 1 H) 8.43 (m, J=8.59 Hz, 2 H) 7.93 (m, J=8.59 Hz, 2 H) 7.78 (s, 1 H) 7.58 (br. s., 1 H) 5.89 (s, 1 H) 5.29 (quin, J=6.63 Hz, 1 H) 4.40 (d, J=4.80 Hz, 2 H) 2.46 (d, J=1.26 Hz, 6 H) 2.25 (s, 3 H) 2.12 (s, 3 H) 1 .52 (s, 3 H) 1 .51 (s, 3 H).
Example 93
6-[4-(Acetylamino)phenyl]-A -[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3^yridinyl)methyl]-3^ methyl-1 -(1 -methyleth l)-1 H-pyrazolo[3,4-fo]pyridine-4-carboxamide
Figure imgf000119_0001
The title compound was prepared in the same manner as described in example 93 using 6-chloro-N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-3-methyl-1 -(1 - methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide (80 mg, 0.21 mmol), [4- (acetylamino)phenyl]boronic acid (55.4 mg, 0.31 mmol),
bis(triphenylphosphine)palladium(ll) chloride (8.4 mg, 0.01 mmol), DME/water (4 mL, 3:1 ) and sodium bicarbonate (52 mg, 0.62 mmol). The crude product was dissolved in
DCM/MeOH (1 :1 ) and preabsorbed onto silica gel. The product was purified by silica gel chromatography (eluent: DCM/MeOH/NH4OH. gradient of 0 to 80:20:2). The collected solid was suspended in EtOH/EtOAc (1 :1 ), sonicated, and filtered. After further washing with hexanes, the solid was then air-dried for 15 min, and then in vacuum oven overnight. The final product was collected as 70 mg (69%). LCMS E-S (M+H) = 487.3 1H NMR (400 MHz, DMSO-de) δ ppm 1 1 .53 (br. s., 1 H) 10.16 (s, 1 H) 8.73 (t, J=5.05 Hz, 1 H) 8.17 (m, J=8.84 Hz, 2 H) 7.74 (m, J=8.84 Hz, 2 H) 7.64 (s, 1 H) 5.89 (s, 1 H) 5.25 (quin, J=6.69 Hz, 1 H) 4.39 (d, J=4.80 Hz, 2 H) 2.43 (s, 3 H) 2.25 (s, 3 H) 2.12 (s, 3 H) 2.09 (s, 3 H) 1.51 (s, 3 H) 1 .49 (s, 3 H). Example 94
6-[4-(Aminocarbonyl)phenyl]-W-[(4,6-dimeth^
1 -(1 -methylethyl)-1 H-p razolo[3,4-fo]pyridine-4-carboxamide
Figure imgf000120_0001
The title compound was prepared in the same manner as described in example 93 using 6-chloro-N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)- 1 H-pyrazolo[3,4-b]pyridine-4-carboxamide (80 mg, 0.21 mmol), [4- (aminocarbonyl)phenyl]boronic acid (52.9 mg, 0.31 mmol),
bis(triphenylphosphine)palladium(ll) chloride (8.7 mg, 0.01 mmol), DME/water (4 mL, 3:1 ) and sodium bicarbonate (54 mg, 0.62 mmol). The crude product was dissolved in
DCM/MeOH (1 :1 ) and preabsorbed onto silica gel. The product was purified by silica gel chromatography (eluent: DCM/MeOH/NH4OH (gradient 0 to 90:10:1 ). The collected solid was suspended in EtOH, sonicated, and filtered. After further washing with EtOH/DCM, the filtered solid was then air-dried for 15 min, and then in vacuum oven overnight. The final product was collected as 70 mg (69%). LCMS E-S (M+H) = 459.3 1 H NMR (400
MHz, DMSO-de) δ ppm 1 1 .58 (s, 1 H) 9.00 (t, J=4.93 Hz, 1 H) 8.41 (s, 1 H) 8.36 (s, 1 H) 8.34 (s, 1 H) 8.24 (s, 1 H) 8.13 (s, 1 H) 8.06 (s, 1 H) 8.04 (s, 1 H) 7.51 (s, 1 H) 5.91 (s, 1 H) 5.37 (quin, J=6.69 Hz, 1 H) 4.42 (d, J=4.80 Hz, 2 H) 2.23 (s, 3 H) 2.13 (s, 3 H) 1.56 (s, 3 H) 1 .55 (s, 3 H).
Example 95
W-[(4,6-Dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-6-{[2-(2- pyridinylamino)ethyl]amino -1 H-pyrazolo[3,4-j ]pyridine-4-carboxamide
Figure imgf000121_0001
6-Chloro-N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)- 1 H-pyrazolo[3,4-b]pyridine-4-carboxamide (80 mg, 0.21 mmol) and (2-aminoethyl)2- pyridinylamine (88 mg, 0.64 mmol) were suspended in 2 ml. of NMP and irradiated (microwave) as follows: 180 °C for 30 min, 200 °C for 30 min, 220°C for 30 min, 230°C for 30 min and then 240 °C for 1 h. After cooling to room temperature, some of the solvent was removed in vacuo and the residue was purified by silica gel chromatography (eluent: DCM/MeOH/NH40H, gradient 0 to 90:10:1 ). The collected solid was partitioned between water and EtOAc/toluene (1 :1 ) and then extracted twice with DCM/isopropanol. The combined organic layers were washed with water, dried over MgSC^, filtered, and concentrated in vacuo to afford a dark brown oil. After drying under vacuum overnight, the residue was again purified by silica gel chromatography (eluent: DCM/MeOH/NI-^OH
(gradient 0 to 90:10:1 ). The collected solid was suspended in EtOH, sonicated, and filtered. The solid was further washed with EtOH/DCM, filtered, and dried in vacuum-oven for 2 days. The final product was collected as 47 mg (45%). LCMS E-S (M+H) = 475.0 1H NMR (400 MHz, DMSO-d6) δ ppm 1 1 .56 (br. s., 1 H) 8.47 (t, J=5.05 Hz, 1 H) 7.97 (dd, J=4.93, 1 .14 Hz, 1 H) 7.85 (s, 1 H) 7.32 - 7.39 (m, 2 H) 6.65 (s, 1 H) 6.61 (t, J=5.43 Hz, 1 H) 6.46 - 6.51 (m, 2 H) 5.89 (s, 1 H) 4.97 (quin, J=6.69 Hz, 1 H) 4.32 (d, J=5.05 Hz, 2 H) 3.49 (dt, J=15.28, 5.62 Hz, 4 H) 2.21 (s, 3 H) 2.13 (s, 3 H) 1.44 (s, 3 H) 1.43 (s, 3 H). Example 96
W-[(4,6-Dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-6-(7-oxo- 1 ,5,6,7-tetrahydro-1 ,8-naphth ridin-3-yl)-1H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000122_0001
6-Bromo-3,4-dihydro-1 ,8-naphthyridin-2(1 H)-one (200 mg, 0.881 mmol) (J. Med.
Chem. 2003; 46; 9; 1627-1635), bis(pinacolato)diboron (268 mg, 1 .057 mmol), Pd(dppf) (35.7 mg, 0.044 mmol) and potassium acetate (259 mg, 2.64 mmol) were suspended in 1 ,4-Dioxane (8 mL), and stirred with heating at 100 °C for 1 h. After cooling to room temperature, 6-chloro-N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1- methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide (329 mg, 0.881 mmol), bis(triphenylphosphine)palladium(ll) chloride (71 .9 mg, 0.088 mmol) and sodium bicarbonate (222 mg, 2.64 mmol) were added, followed by DME (5 mL) and water (3 mL). The reaction mixture was irradiated (microwave) at 120 °C for 2 h. The reaction mxiture was cooled cooled to room temperature and filtered through Na2S0 - The contents were purified directly by silica gel chromatography (eluent : 5% MeOH/CH2Cl2) to furnish the desired product as a grey solid after evaporation and preciptation from warm
EtOAc/MeOH (1 :9). The product was collected as 55 mg (13%). LCMS E-S (M+H) = 486.3 1 H NMR (400 MHz, DMSO-d6) δ ppm 1 1.57 (br. s., 1 H), 10.74 (s, 1 H), 8.99 (d, J=2.27 Hz, 1 H), 8.93 (t, J=4.93 Hz, 1 H), 8.44 (d, J=1.77 Hz, 1 H), 8.37 (s, 1 H), 8.16 (s, 1 H), 5.91 (s, 1 H), 5.29 - 5.40 (m, 1 H), 4.42 (d, J=5.05 Hz, 2 H), 3.04 (t, J=7.58 Hz, 2 H), 2.53 - 2.62 (m, 2 H), 2.23 (s, 3 H), 2.13 (s, 3 H), 1 .54 (d, 6 H).
Example 97
A -[(4,6-Dimethyl-2-oxo-1 ,2-dihydro-3^yridinyl)methyl]-6-{6-[(methylamino)s^
3-pyridinyl}-1 -(1 -meth lethyl)-1 H-pyrazolo[3,4-i ]pyridine-4-carboxamide
Figure imgf000123_0001
5-Bromo-N-methyl-2-pyridinesulfonamide (225 mg, 0.896 mmol),
Bis(pinacolato)diboron (296 mg, 1.165 mmol), Pd(dppf) (35.7 mg, 0.044 mmol) and potassium acetate (264 mg, 2.69 mmol) were suspended in 1 ,4-Dioxane (8 ml_), and stirred with heating at 100 °C for 1 h. After cooling to room temperature, 6-chloro-N-[(4,6- dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1-(1-methylethyl)-1 H-pyrazolo[3,4- b]pyridine-4-carboxamide (368 mg, 0.986 mmol), bis(triphenylphosphine)palladium(ll) chloride (73.2 mg, 0.090 mmol) and sodium bicarbonate (226 mg, 2.69 mmol) were added, followed by DME (5 ml.) and water (3 ml_). The reaction mixture was irradiated (microwave) at 120 °C for 2h. The reaction mxiture was cooled cooled to room
temperature and filtered through Na2S0 - The contents were purified directly by silica gel chromatography (eluent : 15% MeOH/CH2Cl2) to furnish the desired product as a grey solid after evaporation and preciptation from warm EtOAc/MeOH (1 :9). The product was again purified by silica gel chromatography (eluent : 5% MeOH/CH2CI2) to afford the final product as a white solid, 83 mg (18%). LCMS E-S (M+H) = 510.0 1H NMR (400 MHz, DMSO-de) δ ppm 1 1 .59 (br. s., 1 H), 9.56 (d, J=1.52 Hz, 1 H), 8.96 (s, 1 H), 8.86 (dd, J=8.34, 2.27 Hz, 1 H), 8.46 (s, 1 H), 8.34 (s, 1 H), 8.12 (d, J=8.34 Hz, 1 H), 7.85 (br. s., 1 H), 5.91 (s, 1 H), 5.27 - 5.46 (m, 1 H), 4.43 (d, J=4.80 Hz, 2 H), 2.60 (s, 3 H), 2.23 (s, 3 H), 2.13 (s, 3 H), 1.56 (d, 6 H).
Intermediate 17
5-Bromo-N-methyl-2-pyridinesulfonamide
Figure imgf000123_0002
5-Bromo-2-pyridinesulfonyl chloride (500 mg, 1.949 mmol) was added to a 0 °C solution of pyridine (0.315 ml, 3.90 mmol), methylamine (0.975 ml, 1.949 mmol, 2M in THF) and CH2CI2 (2 mL). The reaction mixture was stirred at room temperature for 1 hr, then quenched with brine. The contents were extracted with DCM, dried, filtered, and concentrated in vacuo. The crude product was purified by silica gel chromatography (eluent: 50%, EtOAC/CH2CI2). The product was collected as a clear oil, 225 mg (75%). LCMS E-S (M+H) = 251.1 . 1H NMR (400 MHz, Chloroform-d) δ ppm 8.77 (s, 1 H), 8.07 (dd, J=8.34, 2.27 Hz, 1 H), 7.92 (d, J=8.34 Hz, 1 H), 5.45 (d, J=5.05 Hz, 1 H), 2.76 (d, 3 H).
Intermediate 18
1 -{[4-(Methyloxy)phen l]methyl}-1H-pyrazol-5-amine
Figure imgf000124_0001
Hydrazine hydrate (12.82 g, 400 mmol) was added dropwise to a cooled (< 20 °C) solution of 2-propenenitrile (21 .76 g, 410 mmol) and ethanol (200 mL). After 16 h stirring the reaction mixture was cooled in an ice water bath and 4-(methyloxy)benzaldehyde
(53.8 g, 395 mmol) was added dropwise. The mixture was stirred at room temperature for 16 h. The reaction mixture was concentrated to dryness. The residue was dissolved in n- butanol (200 mL), sodium hydroxide was added (1 g, 25.00 mmol), and the mixture heated at 120 °C for 6 h. The reaction mixture was concentrated to 50% volume under reduced pressure, poured onto 300 mL of water, and then extracted with Et20 (2 x 200 mL). The combined ether phases were extracted with 1 N HCI (3 x 100 mL). The combined HCI extracts were combined and cooled in an ice/water bath. Added next was 6N NaOH until basic (pH > 12). The contents were extracted with Et20 (4 x 100 mL), washed with water, dried over MgS04, filtered, and concentrated under reduced pressure. The residue was purified via silica gel chromatography (eluent : 0 - 50% EtOAc:Hex). The final product was collected as 8.94 g (1 1 %). 1H NMR (400 MHz, CHLOROFORM-d) δ ppm 3.79 (s, 3 H), 5.14 (s, 2 H), 5.55 (d, J=1 .77 Hz, 1 H), 6.81 - 6.94 (m, 2 H), 7.12 (d, J=8.84 Hz, 2 H), 7.31 (d, J=2.02 Hz, 1 H). Intermediate 19
1 -Cyclobut l-3-methyl-1H-pyrazol-5-amine
Figure imgf000125_0001
(2Z)-3-Amino-2-butenenitrile (1.339 g, 16.31 mmol) and cyclobutylhydrazine HCI (2 g, 16.31 mmol) were added to ethanol (20 mL) and heated at 75 °C for 16 h. After cooling to room temperature, the solvent was removed under reduced pressure. The crude residue was suspended in saturated NaHC03 (30 mL) and EtOAc (50 mL), and stirred for 10 min. The phases were separated and the aq. phase extracted with EtOAc (3 x 50 mL). The combined EtOAc extracts were washed with brine, dried over MgS04, filtered, and concentrated in vacuo. The crude residue was purified via silica gel chromatography (eluent: 0 to 5% EtOAc:DCM, then gradient to 100% EtOAc). The final product was collected as 0.4 g (16%). LCMS E-S (M+H) =:152.0. 1 H NMR (400 MHz, CHLOROFORM-d) δ ppm 1.67 - 2.00 (m, 2 H) 2.21 (s, 3 H) 2.32 - 2.43 (m, 2 H) 2.69 (ddd, J=10.55, 9.41 , 2.53 Hz, 2 H) 3.26 - 3.55 (br s, 2 H) 4.48 - 4.59 (m, 1 H) 5.36 (s, 1 H)
Intermediate 20
1 -Cyclopentyl-3-meth l-1H-pyrazol-5-amine
Figure imgf000125_0002
The title compound was prepared in the same manner as described for
intermediate 19 using cyclopentylhydrazine hydrochloride (2 g, 14.64 mmol), (2Z)-3- amino-2-butenenitrile (1.202 g, 14.64 mmol) and ethanol (20 mL). The final product was collected as 0.57 g (24%). LCMS E-S (M+H) =166.0.1H NMR (400 MHz, CHLOROFORM- d) δ ppm 1.60 - 1 .69 (m, 2 H), 1 .86 - 1 .95 (m, 2 H), 1 .99 - 2.09 (m, 4 H), 2.19 (s, 3 H), 3.46 (br. s., 2 H), 4.38 (quin, J=7.89 Hz, 1 H), 5.36 (s, 1 H). Intermediate 21
1 -(Phenylmeth l)-1H-pyrazol-5-amine
Figure imgf000126_0001
A solution of hydrazine hydrate (14.55 mL, 300 mmol) and ethanol (75 mL) was cooled in an ice water bath and then 2-propenenitrile (15.3 g, 288 mmol) was added dropwise. After stirring at room temperature for 2 h, benzaldehyde (31 .8 g, 300 mmol) was added dropwise and the reaction mixture was allowed to stir at room temperature for 2 d. The solvent was removed under reduced pressure. The crude oil was cooled in an ice/water bath followed by dropwise addition of a n-BuONa solution (Na 6.9 g, (300 mmol) in n-Butanol (300 mL)). The reaction mixture was heated at reflux for 1 h, and then cooled to room temperature. The contents were poured onto 300 mL of water and then extracted with Et20 (2 x 200 mL). The ether phase was extracted with 1 N HCI (3 x 100 mL). The combined HCI extracts were combined and cooled in a ice/water bath, followed by addition of 6N NaOH until basic (pH > 12). The mixture was extracted with Et20 (4 x 100 mL), washed with water, dried over MgS04, filtered, and concentrated under reduced pressure. The crude residue was purified via silica gel chromatography (eluent: 0 to 50% EtOAc:Hex). The final product was collected as 6.96 g (14%). LCMS E-S (M+H) = 174.0. 1H NMR (400 MHz, CHLOROFORM-d) δ ppm 3.39 (br. s., 2 H), 5.23 (s, 2 H), 5.59 (d, J=1.77 Hz, 1 H), 7.17 (d, J=6.82 Hz, 2 H), 7.28 - 7.37 (m, 4 H)
Intermediate 22
1 ,1 -Dimethylethyl 2-(tetrah dro-2H-pyran-4-yl)hydrazinecarboxylate
Figure imgf000126_0002
Tetrahydro-4H-pyran-4-one (9.69 g, 97 mmol) was added to a solution of 1 ,1 - dimethylethyl hydrazinecarboxylate (14.07 g, 106 mmol) in methanol (100 mL) and stirred at room temperature for 3 h. The solvent was removed in vacuo, and the crude residue was suspended in acetic acid (140 mL). Next added sodium cyanoborohydride (6.69 g, 106 mmol) in portions over 3 minutes. The contents were stirred at room temperature for 60 h. The solvent was removed in vacuo, and the crude residue was suspended in DCM (100 mL). The reaction mixture was adjusted to pH 7 with 6N NaOH. The layers were separated, and the aq. layer extracted with DCM. The combined organic layers were washed with saturated NaHC03, and brine. The organic layer was dried over MgS04, filtered, and concentrated in vacuo to afford the product as a solid, 18.4 g (88%). 1 H NMR (400 MHz, DMSO-de) δ ppm 1.22 (m, 2 H), 1 .39 (s, 9 H), 1 .43 - 1 .48 (m, 1 H), 1 .59 - 1.69 (m, 2 H), 2.82 - 2.98 (m, 1 H), 3.20 - 3.32 (m, 2 H), 3.80 (d, J=1 1.62 Hz, 2 H), 4.36 (br. s., 1 H), 8.07 - 8.34 (m, 1 H).
Intermediate 23
Tetrahyd r -2H-py ra n -4-y I hyd razi ne
Figure imgf000127_0001
1 , 1 -Dimethylethyl 2-(tetrahydro-2H-pyran-4-yl)hydrazinecarboxylate was added to 1 ,4-Dioxane (10 mL) followed by hydrochloric acid (4M in 1 ,4-Dioxane, 10 mL, 329 mmol). The reaction mixture was stirred at room temperature for 60 h. The reaction mixture was filtered to afford the product as a solid, 1 .02 g. (72%). 1H NMR (400 MHz,
CHLOROFORM-d) δ ppm 1.45 (qd, J=1 1.79, 4.29 Hz, 2 H), 1.81 - 2.01 (m, 2 H), 3.00 - 3.19 (m, 1 H), 3.20 - 3.35 (m, 2 H), 3.78 - 3.97 (m, 2 H), 6.5-9.5(br m, 3 H).
Intermediate 24
3-Methyl-1 -(tetrahydro-2H- ran-4-yl)-1H-pyrazol-5-amine
Figure imgf000127_0002
(2Z)-3-Amino-2-butenenitrile (0.538 g, 6.55 mmol), tetrahydro-2H-pyran-4- ylhydrazine (1 g, 6.55 mmol) and triethylamine (0.913 mL, 6.55 mmol) were added to ethanol (300 mL), and the reaction mixture was stirred at 75 °C for 16 hours. The solvent was removed in vacuo, and the crude material suspended in EtOAc. The organic phase was washed with water and then brine. The organic layer was then dried over MgS04, filtered, and concentrated in vacuo. Half of the crude material was purified by reverse HPLC (mobile phase : 0 - 30% ACN/H20, 0.1 %TFA) to afford 380 mg of the desired product. The other half of the crude material was purified via silica gel chromatography (eluent : 0% to 100% EtOAc:Hex then 0% to 20% MeOH:DCM). An additional 300 mg of the desired product was obtained (overall yield : 57%). LCMS E-S (M+H) = 182.0. 1H NMR (400 MHz, CHLOROFORM-d) δ ppm 1 .78 (dd, 2 H), 1.94 (qd, J=12.13, 4.55 Hz, 2 H), 2.19 (s, 3 H), 3.30 - 3.46 (m, 2 H), 3.99 (dd, J=1 1 .49, 4.17 Hz, 2 H), 4.49 (m, J=1 1 .65, 1 1 .65, 4.1 1 , 3.92 Hz, 1 H), 5.53 (s, 1 H).
Intermediate 25
6-Methyl-1 -(phenylmethyl)-1H-pyrazolo[3,4-b]pyridin
Figure imgf000128_0001
A mixture of 1 -(phenylmethyl)-1 H-pyrazol-4-amine (6.25 g, 36.1 mmol), ethyl 3- oxobutanoate (4.70 g, 36.1 mmol), acetic acid (.2 mL, 3.49 mmol) and benzene (50 mL) were refluxed for 16 h (Dean-Stark trap used). The solvent was removed under reduced pressure and the crude residue purified via silica gel chromatography (eluent; 0 to 60% EtOAc:Hex). The collected product (7.35 g, 25.8 mmol) was then dissolved in 10 mL of Dowtherm A and this solution added dropwise to 10 mL of refluxing Dowtherm A. After refluxing for an additional 20 minutes, the reaction mixture was cooled to room
temperature, and 20 mL of petroleum ether were added. After stirring at room
temperature for 16 hr, the solid product was filtered and dried on hi-vacuum. The final product was collected as 6.23 g (72%). LCMS E-S (M+H) = 240.0. 1H NMR (400 MHz, MeOD) δ ppm 4.89 (s, 3 H), 5.59 (s, 2 H), 6.17 (br. s., 1 H), 7.17 (d, J=6.82 Hz, 2 H), 7.25
- 7.37 (m, 3 H), 8.05 (s, 1 H).
Intermediate 26
4-Bromo-6-methyl-1 -(phenylmethyl)-1H-pyrazolo[3,4-b]pyridine
Figure imgf000128_0002
Phosphorus oxybromide (5.31 g, 18.54 mmol) was added to a suspension of 6- methyl-1 -(phenylmethyl)-1 H-pyrazolo[3,4-b]pyridin-4-ol (3.08 g, 12.87 mmol) and toluene (25 mL). The contents were heated to reflux for 1 h, wherein 10 mL DMF were added during this process. The solvent was removed under reduced pressure. The crude residue was suspended in 30 mL water containing ice. The contents were adjusted to pH = 10 by addition of sat. NaHC03. The solid product was filtered and dried under hi- vacuum. The final product was collected as 3.55 g (91 %). LCMS E-S (M+H) =
302.1/304.0. 1H NMR (400 MHz, DMSO-d6) δ ppm 2.61 (s, 3 H), 5.65 (s, 2 H), 7.16 - 7.35 (m, 5 H), 7.48 (s, 1 H), 8.12 s, 1 H).
Intermediate 27
6-Methyl-1 -(phenyl methyl)-1 -pyrazolo[3,4-b]pyridine-4-carbonitrile
Figure imgf000129_0001
A mixture of 4-bromo-6-methyl-1 -(phenylmethyl)-1 H-pyrazolo[3,4-b]pyridine (3.45 g, 1 1 .42 mmol), dicyanozinc (1 .542 g, 13.13 mmol),
tris(dibenzylideneacetone)dipalladium(0) (0.523 g, 0.571 mmol), SPhos (0.562 g, 1.370 mmol), DMF(49 mL), and water (0.5 mL) was degassed with nitrogen for 10 minutes. The reaction mixture was heated at 120 °C for 2 hours, and then cooled to room temperature. The contents were concentrated to 50% volume and then 50 mL of 1 N NaOH and 50 mL of EtOAc were added. The solids were filtered off. The phases were separated and the aqueous phase extracted with EtOAc (3 x 50 mL). The combined EtOAc extracts were washed with water (3 x 50 mL), brine (50 mL), dried over MgS04, filtered, and
concentrated in vacuo. The crude product containing SPhos reagent (-30-40%) was collected as 3.69 g and used without further purification. LCMS E-S (M+H) = 249.1. 1H NMR (400 MHz, CHLOROFORM-d) δ ppm 2.77 (s, 3 H), 5.73 (s, 2 H), 7.08 - 7.44 (m, 6 H), 8.16 (s, 1 H) Intermediate 28
6-Methyl-1 -(phenylmethyl)-l -pyrazolo[3,4-b]pyridine-4-carboxylic acid
Figure imgf000130_0001
6-Methyl-1-(phenylmethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carbonitrile (1 g), sodium hydroxide (0.805 g, 20.14 mmol), ethanol (25 mL) and water (10 mL) were heated at reflux for 2 hours. After cooling to room temperature, the solvent was removed under reduced pressure. The crude product was suspended in water (50 mL) and acidified by dropwise addition of 6N HCI. The solid product was filtered off and dissolved in 100ml of EtOAc. The solution dried over MgS04, filtered, and concentrated in vacuo. The final product was obtained as 0.730 g. LCMS E-S (M+H) =267.9. 1H NMR (400 MHz, DMSO-d6) δ ppm 2.71 (s, 3 H), 5.70 (s, 2 H), 7.15 - 7.37 (m, 5 H), 7.63 (s, 1 H), 8.33 (s, 1 H), 13.86 (s, 1 H).
Intermediate 29
ethyl 4-cyclopropyl-2,4-dioxobutanoate
Figure imgf000130_0002
Sodium metal (2.41 1 g, 105 mmol) was dissolved in ethanol (50 mL). The solution was heated to reflux followed by addition of a mixture of 1-cyclopropylethanone (8.4 g, 100 mmol) and diethyl oxalate (14.59 g, 100 mmol) dropwise over 30 minutes. The reaction mixture was heated at reflux for an additional 2h, and then allowed to cool to room temperature over a 2 d period. The contents were diluted with water (200 mL) and acidified by dropwise addition of 6N HCI. The contents were extracted with EtOAc (3 x 75 mL), washed with water, brine, dried over MgS04, filtered, and concentrated in vacuo. The final product was collected as 14.3g (74%). LCMS E-S (M+H) =184.8 1 H NMR (400 MHz, CHLOROFORM-d) δ ppm 1.03 (d, J=7.83 Hz, 2 H) 1.13 - 1.19 (m, 2 H) 1 .31 (t, J=7.07 Hz, 3 H) 1 .81 - 1 .90 (m, 1 H) 4.29 (q, J=7.16 Hz, 2 H) 6.43 (s, 1 H). Intermediate 30
Ethyl 6-cyclopropyl-1 -{[4-(methyloxy)phenyl]methyl}-1 H-pyrazolo[3,4-b]pyridine-4- carboxylate
Figure imgf000131_0001
A mixture of 1 -{[4-(methyloxy)phenyl]methyl}-1 H-pyrazol-5-amine (3 g, 14.76 mmol), ethyl 4-cyclopropyl-2,4-dioxobutanoate (2.72 g, 14.76 mmol) and benzene (50 mL) were heated at 63 °C for 16 h. The solvent was removed under reduced pressure. The crude residue was purified via silica gel chromatography (eluent: 0 to 25%
EtOAc:Hex) to afford 2.56 g of the desired cyclized product and 1.71 g of the uncyclized adduct. The uncyclized adduct was dissolved in 25 mL of AcOH and heated to reflux for 16 hours. The solvent was removed under reduced pressure and the residue purified via silica gel chromatography (eluent: 0 to 25% EtOAc:Hex) to afford an additional 1.15 g of the desired cyclized product (combined yield = 71 %). LCMS E-S (M+H) = 352.3. 1H NMR (400 MHz, DMSO-de) δ ppm 1.07 - 1.13 (m, 4 H) 1.38 (t, J=7.07 Hz, 3 H) 2.39 (s, 1 H) 3.67 (s, 3 H) 4.41 (q, J=7.07 Hz, 2 H) 5.51 (s, 2 H) 6.84 (d, J=8.84 Hz, 2 H) 7.21 (d, J=8.59 Hz, 2 H) 7.64 (s, 1 H) 8.23 (s, 1 H).
Intermediate 31
Ethyl 6-cyclopropyl-1H-pyrazolo[3,4-b]pyridine-4-carboxylate
Figure imgf000131_0002
To a 20 mL microwave vial were combined ethyl 6-cyclopropyl-1-{[4- (methyloxy)phenyl]methyl}-1 H-pyrazolo[3,4-b]pyridine-4-carboxylate (2.05 g, 5.83 mmol), trifluoroacetic acid (6.74 ml, 88 mmol) and anisole (1.912 ml, 17.50 mmol). The reaction vessel was sealed and irradiated (microwave) at 100 °C for 5 minutes. After cooling to room temperature, the reaction mixture was concentrated in vacuo. The residue was diluted with water (20 mL) and then saturated NaHC03 until basic. The contents were extracted with DCM (4 x 20 mL). The combined organic layers were washed with water, brine, dried over MgS04, filtered and concentrated in vacuo. The crude product was purified via silica gel chromatography (eluent: 0 to 20% EtOAc:Hex). The final product was obtained as 1.06 g (79%). LCMS E-S (M+H) = 232.2. 1H NMR (400 MHz, DMSO-de) δ ppm 0.95 - 1.21 (m, 4 H) 1 .41 (t, J=7.07 Hz, 3 H) 2.33 - 2.46 (m, 1 H) 4.44 (q, J=7.07 Hz, 2 H) 7.67 (s, 1 H) 8.26 (d, J=1.26 Hz, 1 H) 13.75 (s, 1 H).
Intermediate 32
Ethyl 1 -amino-6-cycloprop l-1 H-pyrazolo[3,4-b]pyridine-4-carboxylate
Figure imgf000132_0001
Ethyl 6-cyclopropyl-1 H-pyrazolo[3,4-b]pyridine-4-carboxylate (200 mg, 0.865 mmol) was dissolved in N-Methyl-2-pyrrolidone (NMP) (8 mL), followed by addition of potassium tert-butoxide (1 16 mg, 1 .038 mmol). After 20 minutes stirring, 0-{[4- (methyloxy)phenyl]carbonyl}hydroxylamine 1 -[(aminooxy)carbonyl]-4-(methyloxy)benzene (289 mg, 1.730 mmol) was added and the mixture stirred at room temperature for 16 h. The contents were diluted with EtOAc, and then washed with brine, and saturated NaHC03. The organic phase was dried over MgS04, filtered, and concentrated in vacuo. The crude product was purified via silica gel chromatography (eluent: 0 to 50% EtOAc). The final product as collected as 106 mg (50%). LCMS E-S (M+H) = 274.4. 1H NMR (400 MHz, DMSO-de) δ ppm 1 .00 - 1.14 (m, 4 H), 1.41 (t, J=7.07 Hz, 3 H), 2.34 - 2.47 (m, 1 H), 4.44 (q, J=7.07 Hz, 2 H), 6.39 (s, 2 H), 7.64 (s, 1 H), 8.09 (s, 1 H).
Intermediate 33
Ethyl 6-cyclopropyl-3-methyl-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4- carboxylate
Figure imgf000133_0001
A solution of ethyl 4-cyclopropyl-2,4-dioxobutanoate (700 mg, 3.76 mmol), 3- methyl-1 -(1 -methylethyl)-1 H-pyrazol-5-amine (523 mg, 3.76 mmol) and benzene (20 mL) were heated at 65 °C for 6 hr, and then allowed to cool to room temperature for 2 d. The solvent was removed under reduced pressure and the residue purified via silica gel chromatography (eluent: 0 to 25% EtOAc:Hex). The final product was collected as 0.77 g (71 %). LCMS E-S (M+H) = 288.0. 1 H NMR (400 MHz, CHLOROFORM-d) ppm 1.04 - 1.1 1 (m, 2 H), 1.13 - 1.20 (m, 4 H), 1.47 (t, J=7.20 Hz, 6 H), 1 .55 (d, J=6.82 Hz, 12 H), 2.16 - 2.25 (m, 2 H), 2.68 (s, 6 H), 4.49 (q, J=7.07 Hz, 4 H), 5.20 (spt, J=6.78 Hz, 2 H), 7.41 (s, 1 H).
Intermediate 34
Ethyl 1 -cyclobutyl-6-cyclopropyl-3-methyl-1H-pyrazolo[3,4-b]pyridine-4-carboxylate
Figure imgf000133_0002
The title compound was prepared in the same manner as described for intermediate 33 using 1 -cyclobutyl-3-methyl-1 H-pyrazol-5-amine (400 mg, 2.65 mmol), ethyl 4-cyclopropyl-2,4-dioxobutanoate (487 mg, 2.65 mmol) and benzene (50 mL), wherein the reaction time was 4 h. The final product was collected as 0.556 g (70%). LCMS E-S (M+H) = 300.6 1 H NMR (400 MHz, CHLOROFORM-d) δ ppm 1.02 - 1 .22 (m, 4 H) 1 .47 (t, J=7.07 Hz, 3 H) 1.80 - 2.01 (m, 2 H) 2.15 - 2.28 (m, 1 H) 2.38 - 2.51 (m, 2 H) 2.70 (s, 3 H) 2.82 (td, J=9.85, 2.53 Hz, 2 H) 4.49 (q, J=7.07 Hz, 2 H) 5.32 - 5.57 (m, 1 H) 7.41 (s, 1 H). Intermediate 35
ethyl 1 -cyclopentyl-6-cyclopropyl-3-methyl-1 H-pyrazolo[3,4-b]pyridine-4- carboxylate
Figure imgf000134_0001
The title compound was prepared in the same manner as described for intermediate 33 using 1 -cyclopentyl-3-methyl-1 H-pyrazol-5-amine (570 mg, 3.45 mmol), ethyl 4-cyclopropyl-2,4-dioxobutanoate (635 mg, 3.45 mmol), and benzene (50 ml.) wherein the reaction time was 4 h. The crude product was purified via silica gel chromatography (eluent: 0 to 10% EtOAc:Hex). The final product was collected as 0.740 g (68%). LCMS E-S (M+H) = 314.3 1H NMR (400 MHz, CHLOROFORM-d) δ ppm 1 .04 - 1.21 (m, 4 H) 1 .47 (t, J=7.20 Hz, 3 H) 1 .73 (br. s., 2 H) 2.00 (d, J=2.78 Hz, 2 H) 2.08 - 2.16 (m, 4 H) 2.21 (s, 1 H) 2.67 (s, 3 H) 4.49 (q, J=7.16 Hz, 2 H) 5.32 (t, J=7.83 Hz, 1 H) 7.40 (s, 1 H). Intermediate 36
ethyl 6-cyclopropyl-1 -(1 ,1 -dimethylethyl)-3-methyl-1 H-pyrazolo[3,4-b]pyridine-4- carboxylate
Figure imgf000134_0002
The title compound was prepared in the same manner as described for intermediate 33 using ethyl 4-cyclopropyl-2,4-dioxobutanoate (481 mg, 2.61 mmol), 1- (1 ,1 -dimethylethyl)-3-methyl-1 H-pyrazol-5-amine (400 mg, 2.61 mmol), and toluene (20 ml_), wherein the reaction time was 3 h. The crude product was purified via silica gel chromatography (eluent: 0 to 10% EtOAc:Hex). The final product was collected as 0.24 g (30%). LCMS E-S (M+H) = 302.5. 1 H NMR (400 MHz, CHLOROFORM-d) δ ppm 1 .02 - 1.23 (m, 4 H) 1 .47 (t, J=7.20 Hz, 3 H) 1.79 (s, 9 H) 2.19 (s, 1 H) 2.66 (s, 3 H) 4.49 (q, J=7.07 Hz, 2 H) 7.44 (s, 1 H).
Intermediate 37
Ethyl 6-cyclopropyl-1 -(1 -cyclopropylethyl)-3-methyl-1 H-pyrazolo[3,4-b]pyridine-4- carbox late
Figure imgf000135_0001
1-(1-Cyclopropylethyl)-3-methyl-1 H-pyrazol-5-amine (500 mg, 3.03 mmol) and ethyl 4-cyclopropyl-2,4-dioxobutanoate (557 mg, 3.03 mmol) were suspended in Toluene (10 ml.) and heated at 70 °C for 16 h. The solvent was removed in vacuo and the crude residue was purified via silica gel chromatography (eluent: gradient of 0 to 10%
EtOAc:Hex). The final product was collected as a solid, 0.722 g (76%). LCMS E-S (M+H) = 314.3 1H NMR (400 MHz, DMSO-de) δ ppm 0.17 - 0.32 (m, 2 H), 0.39 (m, J=9.69, 4.82, 4.82, 4.67 Hz, 1 H), 0.51 - 0.62 (m, 1 H), 0.94 - 1.1 1 (m, 4 H), 1.28 - 1.41 (m, 4 H), 1.58 (d, J=6.82 Hz, 3 H), 2.30 - 2.38 (m, 1 H), 2.56 (s, 3 H), 4.19 (dq, J=9.44, 6.87 Hz, 1 H), 4.43 (q, J=7.07 Hz, 2 H), 7.48 (s, 1 H)
Intermediate 38
Ethyl 1 -cyclohexyl-6-cyclopropyl-3-meth l-1H-pyrazolo[3,4-b]pyridine-4-carboxylate
Figure imgf000135_0002
The title compound was prepared in the same manner as described for
intermediate 37 using 1-(1 -cyclohexyl)-3-methyl-1 H-pyrazol-5-amine (500 mg, 2.79 mmol) and ethyl 4-cyclopropyl-2,4-dioxobutanoate (514 mg, 2.79 mmol). The final product was collected as a solid, 0.827 g (91 %). LCMS E-S (M+H) = 328.3 1 H NMR (400 MHz, DMSO-de) δ ppm 1.01 - 1.10 (m, 3 H), 1 .20 - 1 .30 (m, 1 H), 1 .37 (t, J=7.07 Hz, 3 H), 1.41 - 1.52 (m, 2 H), 1.70 (d, 1 H), 1.79 - 1.98 (m, 6 H), 2.30 - 2.38 (m, 1 H), 2.54 (s, 3 H), 4.42 (q, J=7.07 Hz, 2 H), 4.60 - 4.74 (m, 1 H), 7.44 (s, 1 H).
Intermediate 39
Ethyl 6-cyclopropyl-3-methyl-1 -(1 -methyl-4-piperidinyl)-1H-pyrazolo[3,4-b]pyridine-
4-carbox late
Figure imgf000136_0001
3-Methyl-1-(1-methyl-4-piperidinyl)-1 H-pyrazol-5-amine (500 mg, 2.57 mmol) and ethyl 4-cyclopropyl-2,4-dioxobutanoate (474 mg, 2.57 mmol) were suspended in Toluene (10 ml.) and heated at 70 °C for 5 h. The solvent was removed in vacuo and the crude residue was purified via silica gel chromatography (eluent: gradient of 0 to 10%
MeOH:DCM). The final product was collected as a solid, 0.722 g (76%). LCMS E-S (M+H) = 343.1 1H NMR (400 MHz, DMSO-de) δ ppm 1.03 - 1.1 1 (m, 4 H), 1 .38 (t, J=7.07 Hz, 3 H), 1.83 (d, J=6.06 Hz, 2 H), 2.12 - 2.22 (m, 4 H), 2.27 (s, 3 H), 2.35 (m, J=7.83, 7.83, 5.05, 4.80 Hz, 1 H), 2.54 (s, 3 H), 2.94 (d, J=6.57 Hz, 2 H), 4.42 (q, J=7.16 Hz, 2 H), 4.57 - 4.73 (m, 1 H), 7.46 (s, 1 H).
Intermediate 40
Ethyl 6-cyclopropyl-3-methyl-1 -(tetrahydro-2H-pyran-4-yl)-1 H-pyrazolo[3,4- b] py ri d i ne-4-carbox I ate
Figure imgf000136_0002
A mixture of 3-methyl-1 -(tetrahydro-2H-pyran-4-yl)-1 H-pyrazol-5-amine (380 mg, 2.097 mmol), ethyl (3Z)-4-cyclopropyl-4-hydroxy-2-oxo-3-butenoate (386 mg, 2.097 mmol) and acetic acid (50 ml.) were heated at 1 17 °C for 2 hours. The solvent was removed in vacuo, and the crude residue was purified via silica gel chromatography (eluent: 0 to 25% EtOAc:Hex). The desired product was collected as a solid, 300 mg (43%). LCMS E-S (M+H) = 330.3 1H NMR (400 MHz, DMSO-de) δ ppm 1.00 - 1 .12 (m, 4 H), 1.37 (t, J=7.20 Hz, 3 H), 1 .82 (dd, J=12.51 , 2.40 Hz, 2 H), 2.16 (qd, J=12.21 , 4.55 Hz, 2 H), 2.29 - 2.42 (m, 1 H), 2.54 (s, 3 H), 3.47 - 3.60 (m, 2 H), 3.99 (dd, J=1 1 .37, 3.79 Hz, 2 H), 4.42 (q, J=7.24 Hz, 2 H), 4.92 (tt, J=1 1 .59, 4.20 Hz, 1 H), 7.46 (s, 1 H).
Intermediate 41
Ethyl 6-cyclopropyl-1 -[2-({[(1 ,1 -dimethylethyl)oxy]carbonyl}amino)ethyl]-1 H- pyrazolo[3,4-b]pyridine-4-carboxylate
Figure imgf000137_0001
Ethyl 6-cyclopropyl-1 H-pyrazolo[3,4-b]pyridine-4-carboxylate (125 mg, 0.541 mmol) was dissolved in DMF(10 ml_), followed by addition of potassium tert-butoxide (79 mg, 0.703 mmol). After stirring for 15 minutes, 1 , 1-dimethylethyl (2-bromoethyl)carbamate (121 mg, 0.541 mmol) was added and the mixture was stirred at room temperature for 16 h. The reaction mixture was concentrated to dryness. The crude residue was diluted with water (50 ml.) and acidified with acetic acid. The contents were extracted with DCM (4 x 50 ml_). The combined organic layers were washed with water, brine, dried over MgS04, filtered, and concentrated in vacuo. The crude residue was purified via silica gel chromatography (eluent: 0 to 50% EtOAc:Hex). The final product was obtained as 0.1 14 g (56%). LCMS E-S (M+H) = 375.1. 1H NMR (400 MHz, DMSO-de) δ ppm 1 .04 - 1.14 (m, 4 H), 1.15 - 1 .20 (m, 2 H), 1.27 (s, 7 H), 1.41 (t, J=7.07 Hz, 3 H), 2.36 - 2.45 (m, 1 H), 3.31 - 3.41 (m, 3 H), 4.36 - 4.50 (m, 5 H), 6.84 (t, J=5.81 Hz, 1 H), 7.65 (s, 1 H), 8.24 (s, 3 H). Intermediate 42
Ethyl 6-cyclopropyl-3-methyl-1 -(4-pyridinyl)-1 H-pyrazolo[3,4-b]pyridine-4- carboxylate
Figure imgf000138_0001
The title compound was prepared in the same manner as described for
intermediate 30 using 3-methyl-1 -(4-pyridinyl)-1 H-pyrazol-5-amine (310 mg, 1 .780 mmol), ethyl 4-cyclopropyl-2,4-dioxobutanoate (328 mg, 1 .780 mmol), benzene (50 mL), and acetic acid (25 mL). The final product was collected as 3.71 (71 % overall). LCMS E-S (M+H) = 323.5 1H NMR (400 MHz, DMSO-de) δ ppm 1.15 - 1.22 (m, 4 H), 1 .41 (t, J=7.07 Hz, 3 H), 2.64 - 2.69 (m, 4 H), 4.47 (q, J=7.07 Hz, 2 H), 7.74 (s, 1 H), 8.27 - 8.36 (m, 2 H), 8.66 - 8.71 (m, 2 H).
Intermediate 43
6-Cyclopropyl-1 -ethyl-1H-pyrazolo[3,4-b]pyridine-4-carboxylic acid
Figure imgf000138_0002
To a solution of ethyl 6-cyclopropyl-1 H-pyrazolo[3,4-b]pyridine-4-carboxylate (200 mg, 0.865 mmol) in DMF(10 mL) was carefully added sodium hydride (29.1 mg, 1 .21 1 mmol). After 15 minutes stirring, iodoethane (0.077 mL, 0.951 mmol) was added and the mixture stirred at room temperature for 2 hr. Sodium hydroxide (1 mL, 1 .000 mmol) was added and the mixture allowed to stir at room temperature for 1 h. The contents were concentrated in vacuo. The crude residue was diluted with water (50 mL) and acidified with acetic acid. The contents were then extracted with DCM (4 x 50 mL). The combined organic layers were washed with water, brine, dried over MgS04, filtered and
concentrated in vacuo. The crude product was purified by silica gel chromatography (eluent: hexanes to 100% EtOAc, then DCM to 20% MeOH:DCM). The final product was collected as 90 mg (45%). LCMS E-S (M+H) = 232.1 1H NMR (400 MHz, DMSO-de) δ ppm 1.06 - 1.17 (m, 4 H) 1.41 (t, J=7.33 Hz, 3 H) 2.35 - 2.45 (m, 1 H) 4.34 - 4.57 (m, 2 H) 7.62 (s, 1 H) 8.24 (s, 1 H) 13.83 (br. s., 1 H). Regiochemical assignment supported by 2D HNMR.
Intermediate 44
6-Cyclopropyl-1 -propyl-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid
Figure imgf000139_0001
The title compound was prepared as in the same manner as described for intermediate 43 using ethyl 6-cyclopropyl-1 H-pyrazolo[3,4-b]pyridine-4-carboxylate (200 mg, 0.865 mmol), sodium hydride (29.1 mg, 1 .21 1 mmol), DMF (10 mL), 1-iodopropane (162 mg, 0.951 mmol), and NaOH (1 mL). The crude product was purified by silica gel chromatography (eluent: 0 to 10% MeOH:DCM) to afford the final product as a solid, 90 mg (42%). Regiochemical assignment supported by 2D HNMR. LCMS E-S (M+H) = 246.0 1 H NMR (400 MHz, DMSO-d6) δ ppm 13.83 (br s, 1 H), 8.24 (s, 1 H), 7.62 (s, 1 H), 4.26 (t, 2H, J = 7.2 Hz), 2.37 - 2.40 (m, 2H), 1.83 - 1 .89 (m, 2H), 1 .07 - 1 .1 1 (m, 4H), 0.77 (t, 3H, J = 7.2 Hz).
Intermediate 45
1 -Amino-6-cyclopropyl-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid
Figure imgf000139_0002
To a solution of ethyl 1-amino-6-cyclopropyl-1 H-pyrazolo[3,4-b]pyridine-4- carboxylate (100 mg, 0.406 mmol) in ethanol (10 mL) was added sodium hydroxide (1 ml, 0.406 mmol), and the mixture stirred at room temperature for 2h. The solvent was removed in vacuo, and the residue diluted with EtOAc (30 mL) and water (20 mL). The reaction mixture was acidified to pH 3 with citric acid. The phases were separated and the aq. phase extracted with EtOAc (4 x 20 mL). The combined organic layers were washed with water, dried over MgS04, filtered, and concentrated in vacuo. The final product was collected as 0.075 g (84%). LCMS E-S (M+H) =219.5. 1H NMR (400 MHz, DMSO-de) δ m 1.00 - 1.13 (m, 4 H), 2.27 - 2.45 (m, 1 H), 6.36 (s, 2 H), 7.60 (s, 1 H), 8.06 (s, 1 .81 (br. s., 1 H).
Intermediate 46
6-Cyclopropyl-3-methyl-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid
Figure imgf000140_0001
To a solution of ethyl 6-cyclopropyl-3-methyl-1-(1-methylethyl)-1 H-pyrazolo[3,4- b]pyridine-4-carboxylate (200 mg, 0.696 mmol) in ethanol (5 mL) was added sodium hydroxide (2.088 mL, 2.088 mmol), and the mixture stirred at room temperature for 30 min. The reaction mixture was concentrated in vacuo. The residue was diluted with water (30 mL) and then acidified with acetic acid. The contents were extracted with DCM (3 x 30 mL). The combined organic layers were washed with water, brine, dried over MgS04, filtered, and concentrated in vacuo. The final product was obtained as 0.17 g (94%). LCMS E-S (M+H) = 260.0. 1H NMR (400 MHz, DMSO-de) δ ppm 0.97 - 1.12 (m, 4 H) 1.44 (d, J=6.57 Hz, 6 H) 2.27 - 2.38 (m, 1 H) 2.55 (s, 3 H) 5.07 (quin, J=6.69 Hz, 1 H) 7.43 (s, 1 H) 13.30 - 14.08 (m, 1 H).
Intermediate 47
6-Cyclopropyl-1 -[2-({[(1 ,1 -dimethylethyl)oxy]carbonyl}amino)ethyl]-1 H-pyrazolo[3,4- b]pyridine-4-carboxylic acid
Figure imgf000140_0002
The title compound was prepared in the same manner as described for intermediate 46 using ethyl 6-cyclopropyl-1 -[2-({[(1 ,1 - dimethylethyl)oxy]carbonyl}amino)ethyl]-1 H-pyrazolo[3,4-b]pyridine-4-carboxylate (1 10 mg, 0.294 mmol), sodium hydroxide (1 mL, 0.294 mmol), and ethanol (10 mL) wherein the stir time was 16h. The final product was collected as 0.060 g (59%). LCMS E-S (M+H) = 347.1 1 H NMR (400 MHz, DMSO-d6) δ ppm 1 .06 - 1.13 (m, 4 H) 1 .27 (s, 9 H) 2.33 - 2.43 (m, 1 H) 3.37 (q, J=5.81 Hz, 2 H) 4.44 (t, J=5.94 Hz, 2 H) 6.84 (t, J=5.81 Hz, 1 H) 7.62 (s, 5 H) 8.24 (s, 1 H).
Intermediate 48
1 -Cyclobutyl-6-cyclopropyl-3-methyl-1H-pyrazolo[3,4-b]pyridine-4-carboxylic acid
Figure imgf000141_0001
The title compound was prepared in the same manner as described for intermediate 46 using ethyl 1-cyclobutyl-6-cyclopropyl-3-methyl-1 H-pyrazolo[3,4- b]pyridine-4-carboxylate (550 mg, 1 .837 mmol), sodium hydroxide (3 ml, 3.00 mmol), and ethanol (30 mL) wherein the stir time was 1 h. The final product was collected as 0.490 g (98%). LCMS E-S (M+H) = 272.5 1H NMR (400 MHz, DMSO-de) δ ppm 1.03 - 1 .10 (m, 4 H), 1.80 - 1.91 (m, 2 H), 2.26 - 2.43 (m, 3 H), 2.58 (s, 3 H), 2.59 - 2.71 (m, 2 H), 5.35 (dq, J=8.59, 8.42 Hz, 1 H), 7.44 (s, 1 H), 13.73 (br. s., 1 H).
Intermediate 49
1 -Cyclopentyl-6-cyclopropyl-3-methyl-1H-pyrazolo[3,4-b]pyridine-4-carboxylic acid
Figure imgf000141_0002
The title compound was prepared in the same manner as described for intermediate 46 using ethyl 1 -cyclopentyl-6-cyclopropyl-3-methyl-1 H-pyrazolo[3,4- b]pyridine-4-carboxylate (740 mg, 2.361 mmol), sodium hydroxide (4ml, 4.00 mmol), and ethanol (30 mL), wherein the stir time was 1 h. The final product was collected as 0.530 g (79%). LCMS E-S (M+H) = 286.3 1H NMR (400 MHz, DMSO-de) δ ppm 0.96 - 1 .12 (m, 4 H) 1 .52 - 1.74 (m, 2 H) 1 .82 - 2.13 (m, 6 H) 2.26 - 2.37 (m, 1 H) 2.55 (s, 3 H) 5.17 - 5.30 (m, 1 H) 7.43 (s, 1 H) 13.70 (br. s., 1 H). Intermediate 50
6-Cyclopropyl-1 -(1 ,1 -dimethylethyl)-3-methyl-1 H-pyrazolo[3,4-b]pyridine-4- carboxylic acid
The title compound was prepared in the same manner as described for intermediate 46 using ethyl 6-cyclopropyl-1 -(1 , 1-dimethylethyl)-3-methyl-1 H-pyrazolo[3,4- b]pyridine-4-carboxylate (240 mg, 0.796 mmol), sodium hydroxide (4 mL, 4.00 mmol), and ethanol (30 mL), wherein the stir time was 1 h. The final product was collected as 0.210 g (96%). LCMS E-S (M+H) = 274.4. 1 H NMR (400 MHz, DMSO-de) δ ppm 1 .05 (m, 4 H) 1.70 (s, 9 H) 2.33 (m, 1 H) 2.51 (s, 3 H) 7.46 (s, 1 H) 13.70 (br. s., 1 H).
Intermediate 51
6-Cyclopropyl-3-methyl-1 -(4-pyridinyl)-1H-pyrazolo[3,4-b]pyridine-4-carboxylic acid
Figure imgf000142_0002
The title compound was prepared in the same manner as described for intermediate 46 using ethyl 6-cyclopropyl-1 -(4-pyridinyl)-3-methyl-1 H-pyrazolo[3,4- b]pyridine-4-carboxylate (240 mg, 0.796 mmol), sodium hydroxide (4 mL, 4.00 mmol), and ethanol (30 mL), wherein the stir time was 1 h. The final product was collected as 0.210 g (89%). LCMS E-S (M+H) = 295.3.
Intermediate 52
6-Cyclopropyl-1 -(1 -cyclopropylethyl)-3-methyl-1 H-pyrazolo[3,4-b]pyridine-4- carboxylic acid
Figure imgf000143_0001
EtOH (10 mL) solution of ethyl 6-cyclopropyl-1-(1-cyclopropylethyl)-3- methyl-1 H-pyrazolo[3,4-b]pyridine-4-carboxylate (720 mg, 2.297 mmol) was added sodium hydroxide (6.89 mL, 6.89 mmol) and the mixture heated at 70 °C for 1 hour. The solvent was removed in vacuo and the residue was dissolved in 20 mL of water. The contents were acidifed with acetic acid, and extracted with EtOAc (4 x 30mL). The combined organic extracts were washed with water, brine, dried over MgS04, filtered, and concentrated in vacuo. The final product was collected as a solid, 0.560 g (85%). LCMS E-S (M+H) = 286.3 1 H NMR (400 MHz, DMSO-d6) δ ppm 0.15 - 0.33 (m, 2 H), 0.38 (dq, J=9.57, 4.89 Hz, 1 H), 0.50 - 0.65 (m, 1 H), 0.90 - 1 .10 (m, 4 H), 1 .27 - 1 .42 (m, 1 H), 1 .57 (d, J=6.82 Hz, 3 H), 2.23 - 2.36 (m, 1 H), 2.57 (s, 3 H), 4.18 (dq, J=9.32, 6.83 Hz, 1 H), 7.44 (s, 1 H), 12.79 (br. s., 1 H).
Intermediate 53
1 -cyclohexyl-6-cyclopropyl-3-methyl-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid
Figure imgf000143_0002
The title compound was prepared in the same manner as described for intermediate 52 using ethyl 6-cyclopropyl-1-(1 -cyclohexyl)-3-methyl-1 H-pyrazolo[3,4- b]pyridine-4-carboxylate (820 mg, 2.5 mmol) and sodium hydroxide (7.51 mL, 7.51 mmol). The final product was collected as a solid, 0.660 g (88%). LCMS E-S (M+H) = 300.4 1H NMR (400 MHz, DMSO-de) δ ppm 1.01 - 1.1 1 (m, 4 H), 1 .16 - 1 .31 (m, 1 H), 1 .36 - 1 .55 (m, 2 H), 1.70 (d, J=12.38 Hz, 1 H), 1.80 - 1.98 (m, 6 H), 2.32 (m, J=7.80, 7.80, 5.05, 4.86 Hz, 1 H), 2.55 (s, 3 H), 4.60 - 4.73 (m, 1 H), 7.40 (s, 1 H), 13.71 (br. s., 1 H). Intermediate 54
6-Cyclopropyl-3-methyl-1 -(1 -methyl -4-piperidinyl)-1H-pyrazolo[3,4-b]pyridine-4- carboxylic acid
Figure imgf000144_0001
To an EtOH solution (10 mL) of ethyl 6-cyclopropyl-3-methyl-1-(1-methyl-4- piperidinyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxylate (620 mg, 1.81 1 mmol) was added sodium hydroxide (5.43 mL, 5.43 mmol) and the mixture heated at 70 °C for 1 hour. The solvent was removed in vacuo and the residue was dissolved in 20 mL of water. The contents were acidifed with acetic acid, and extracted with EtOAc (4 x 30mL). The combined organic extracts were washed with water, brine, dried over MgS04, filtered, and concentrated in vacuo to afford a minor amount of product. The aqueous phase was concentrated in vacuo and the crude residue purified by reverse phase HPLC purification (eluent : 0% ACN/H20, 0.1 %TFA to 45% ACN/H20, 0.1 % TFA). The isolated solid product was concentrated from toluene to afford the final product as 510 mg (90%).
LCMS E-S (M+H) = 315.2. 1 H NMR (400 MHz, DMSO-d6) δ ppm 1.08 (d, J=6.57 Hz, 4
H), 2.10 (d, J=13.14 Hz, 2 H), 2.30 - 2.42 (m, 3 H), 2.55 (s, 3 H), 2.77 - 2.92 (m, 3 H), 3.30 (br. s., 2 H), 3.57 (d, J=12.13 Hz, 2 H), 4.98 (m, J=1 1 .78, 1 1.78, 3.85, 3.66 Hz, 1 H), 7.46 (s, 1 H), 9.87 (br. s., 1 H). Intermediate 55
6-Cyclopropyl-3-methyl-1 -(tetrahydro-2H-pyran-4-yl)-1H-pyrazolo[3,4-b]pyridine-4- carboxylic acid
Figure imgf000144_0002
To an EtOH solution (30 mL) of ethyl 6-cyclopropyl-3-methyl-1-(tetrahydro-2H- pyran-4-yl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxylate (300 mg, 0.91 1 mmol) was added sodium hydroxide (1 .82 mL, 1 .82 mmol) and the mixture stirred at room temperature for 16 h. The solvent was removed in vacuo and the residue was dissolved in 20 ml. of water. The contents were acidifed with acetic acid, and extracted with EtOAc (4 x 30ml_). The combined organic extracts were washed with water, brine, dried over MgS04, filtered, and concentrated in vacuo to afford the final product as 265 mg (97%). LCMS E-S (M+H)= 302.4 1H NMR (400 MHz, DMSO-de) δ ppm 1 .02 - 1 .09 (m, 4 H), 1 .81 (dd,
J=12.38, 2.27 Hz, 2 H), 2.15 (qd, J=12.25, 4.42 Hz, 2 H), 2.28 - 2.37 (m, 1 H), 2.54 (s, 3 H), 3.46 - 3.60 (m, 2 H), 3.89 - 4.02 (m, 2 H), 4.91 (m, J=1 1.56, 1 1.56, 4.17, 4.04 Hz, 1 H), 7.42 (s, 1 H), 13.73 (br. s., 1 H). Intermediate 56
(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl 6-cyclopropyl-1 -[2-({[(1 ,1 - dimethylethyl)oxy]carbonyl}amino)ethyl]-1 H-pyrazolo[3,4-b]pyridine-4-carboxylate
Figure imgf000145_0001
To a solution of DMSO (10 mL) were sequentially added 6-cyclopropyl-1-[2-({[(1 , 1- dimethylethyl)oxy]carbonyl}amino)ethyl]-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid (270 mg, 0.779 mmol), 3-(aminomethyl)-4,6-dimethyl-2(1 H)-pyridinone (178 mg, 1 .169 mmol), and 1 -hydroxy-7-azabenzotriazole (212 mg, 1.559 mmol), and the mixture stirred at room temperature for 10 min. Next was added EDC (299 mg, 1 .559 mmol) and N- methylmorpholine (0.343 mL, 3.12 mmol), and reaction mixture was stirred at room temperature for 16 h.. The contents were diluted with water (25 mL) and stirred for 10 min. The solid product was filtered off, dried, and collected as 0.290 g (98%). LCMS E-S (M+H) = 381.2. 1H NMR (400 MHz, DMSO-de) δ ppm 1.00 - 1 .15 (m, 4 H), 1 .30 (s, 9H), 2.13 (s, 3 H), 2.18 - 2.32 (m, 4 H), 4.36 (d, J=4.80 Hz, 2 H), 4.42 (t, J=5.81 Hz, 2 H), 5.90 (s, 1 H), 6.85 (t, J=5.56 Hz, 1 H), 7.44 (s, 1 H), 8.22 (s, 2 H), 8.73 (t, J=4.67 Hz, 1 H), 1 1 .57 (br. s., 1 H). Example 98
1 -(2-Aminoethyl)-6-cyclopropyl-W-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3- pyridinyl)methyl]-1H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000146_0001
To a solution of 1 , 1 -dimethylethyl {2-[6-cyclopropyl-4-({[(4,6-dimethyl-2-oxo-1 ,2- dihydro-3-pyridinyl)methyl]amino}carbonyl)-1 H-pyrazolo[3,4-b]pyridin-1 -yl]ethyl}carbamate (260 mg, 0.541 mmol) in DCM (15 mL) was added trifluoroacetic acid (3 ml, 38.9 mmol) and the mixture stirred at room temperature for 2 h. The contents were concentrated in vacuo and the crude residue purified by reverse phase HPLC. (mobile phase : 10 to 70% ACN in H20, 0.1 % NH4OH). The final product was collected as 0.170 g (89%). LCMS E- S (M+H) = 353.3 1 H NMR (400 MHz, DMSO-de) δ ppm 1 .07 (d, J=6.32 Hz, 4 H), 2.13 (s, 3 H), 2.21 (s, 4 H), 3.00 (t, J=6.19 Hz, 2 H), 4.29 - 4.47 (m, 4 H), 5.90 (s, 1 H,) 7.43 (s, 1 H), 8.22 (s, 1 H), 8.75 (t, 1 H). Example 99
6-Cyclopropyl-A -[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -{2- [(methylsulfonyl)amino]ethyl}-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000146_0002
To a solution of 1-(2-aminoethyl)-6-cyclopropyl-N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro- 3-pyridinyl)methyl]-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide (120 mg, 0.315 mmol) in pyridine (2 mL) was added methanesulfonyl chloride (0.025 mL, 0.315 mmol) and the reaction mixture stirred at room temperature for 16 hours. The reaction mixture was concentrated in vacuo, diluted with water, and extracted with EtOAc. The combined organic extracts were washed with brine, dried over MgS04, filtered, and concentrated in vacuo. The crude product was purified by reverse phase HPLC to afford a solid (25 mg). This solid was suspended in MeOH (1 mL) and treated with 4N HCI (1 mL). The solvent was removed under reduced pressure to afford the final product as a solid which was collected as 15 mg (9%). LCMS E-S (M+H) = 459.2. 1H NMR (400 MHz, DMSO-de) δ ppm 1.02 - 1.15 (m, 4 H), 2.13 (s, 3 H), 2.22 (s, 3 H), 2.23 - 2.31 (m, 1 H), 2.82 (s, 3 H), 3.45 (q,2 H), 4.36 (d, J=5.05 Hz, 2 H), 4.49 (t, J=6.44 Hz, 2 H), 5.90 (s, 1 H), 7.19 (s, 1 H), 7.47 (s, 1 H), 8.25 (s, 1 H), 8.76 (t, J=4.93 Hz, 1 H), 1 1 .56 (s, 1 H).
Example 100
A -[(4,6-Dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-6-methyl-1 -(phenylmethyl)- 1H-pyrazolo -b]pyridine-4-carboxamide
Figure imgf000147_0001
The title compound was prepared in the same manner as described for
intermediate 56 using 6-methyl-1 -(phenylmethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid (200 mg, 0.748 mmol), 3-(aminomethyl)-4,6-dimethyl-2(1 H)-pyridinone (142 mg, 0.935 mmol), 1-hydroxy-7-azabenzotriazole (204 mg, 1.497 mmol) , DMSO (10 mL), EDC (287 mg, 1.497 mmol), and N-methylmorpholine (0.329 mL, 2.99 mmol). The final product was collected as 0.25 g (83%). LCMS E-S (M+H) = 402.2. 1H NMR (400 MHz, DMSO-d6) δ ppm 0.93 - 1.12 (m, 4 H), 1.42 (d, J=6.82 Hz, 6 H), 2.12 (s, 3 H), 2.22 (s, 4 H), 2.36 (s, 3 H), 4.34 (d, J=4.80 Hz, 2 H), 4.90 - 5.1 1 (m, 1 H), 5.87 (s, 1 H), 6.99 (s, 1 H), 8.58 (s, 1 H), 1 1 .51 (s, 1 H. Example 101
6-Cyclopropyl-W-[(4,6-dimethyl-2-oxo-1 ,2-dihyd
pyrazolo[ -b]pyridine-4-carboxamide
Figure imgf000148_0001
The title compound was prepared in the same manner as described for intermediate 56 using 6-cyclopropyl-1-ethyl-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid (91 mg, 0.394 mmol), 3-(aminomethyl)-4,6-dimethyl-2(1 H)-pyridinone (75 mg, 0.493 mmol), 1 -hydroxy-7-azabenzotriazole (107 mg, 0.788 mmol), DMSO(10 ml_), EDC (151 mg, 0.788 mmol), and N-methylmorpholine (0.173 ml_, 1.577 mmol). The final product was collected as 0.090 g (63%). LCMS E-S (M+H) = 366.3 1 H NMR (400 MHz, DMSO- d6) 5 ppm 1.07 (d, J=6.32 Hz, 4 H), 1.39 (t, J=7.20 Hz, 3 H), 2.13 (s, 3 H), 2.19 - 2.32 (m, 4 H), 4.36 (d, J=4.80 Hz, 2 H), 4.42 (q, J=7.07 Hz, 2 H), 5.90 (s, 1 H), 7.43 (s, 1 H), 8.21 (s, 1 H), 8.75 (t, J=4.67 Hz, 1 H), 1 1.57 (br. s., 1 H). Example 102
6-Cyclopropyl-A -[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -propyl-1 H- pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000148_0002
The title compound was prepared in the same manner as described for intermediate 56 using 6-cyclopropyl-1 -propyl-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid (90 mg, 0.368 mmol), 3-(aminomethyl)-4,6-dimethyl-2(1 H)-pyridinone (70 mg, 0.460 mmol), 1 -hydroxy-7-azabenzotriazole (100 mg, 0.736 mmol), DMSO(10 ml_), EDC (141 mg, 0.736 mmol), and N-methylmorpholine (0.162 mL, 1.472 mmol). The final product was collected as 0.1 18 g (84%). LCMS E-S (M+H) = 380.3. 1H NMR (400 MHz, DMSO- d6) δ ppm 0.78 (t, 3 H), 0.99 - 1 .16 (m, 4 H), 1 .85 (d, J=7.33 Hz, 2 H), 2.13 (s, 3 H), 2.22 (s, 4 H), 4.25 - 4.43 (m, 4 H), 5.90 (s, 1 H), 7.43 (s, 1 H), 8.21 (s, 1 H), 8.75 (s, 1 H), 1 1.57 (s, 1 H).
Example 103
6-Cyclopropyl-W-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-3-methyl-1 -(1 methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000149_0001
The title compound was prepared in the same manner as described for intermediate 56 using 6-cyclopropyl-1-(2-methylpropyl)-1 H-pyrazolo[3,4-b]pyridine-4- carboxylic acid (80 mg, 0.309 mmol), 3-(aminomethyl)-4,6-dimethyl-2(1 H)-pyridinone (70.4 mg, 0.463 mmol), 1 -hydroxy-7-azabenzotriazole (84 mg, 0.617 mmol), DMSO(10 mL), EDC (1 18 mg, 0.617 mmol), and N-methylmorpholine (0.136 mL, 1.234 mmol). The final product was collected as 0.123 g (100%). LCMS E-S (M+H) = 394.2 1H NMR (400 MHz, DMSO-de) δ ppm 0.93 - 1 .12 (m, 4 H), 1.42 (d, J=6.82 Hz, 6 H), 2.12 (s, 3 H), 2.22 (s, 4 H), 2.36 (s, 3 H), 4.34 (d, J=4.80 Hz, 2 H), 4.90 - 5.1 1 (m, 1 H), 5.87 (s, 1 H), 6.99 (s, 1 H), 8.58 (s, 1 H), 1 1 .51 (s, 1 H).
Example 104
-6-cyclopropyl-A -[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 H- pyrazolo[ -b]pyridine-4-carboxamide
Figure imgf000150_0001
The title compound was prepared in the same manner as described for
intermediate 56 using 1-amino-6-cyclopropyl-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid (73 mg, 0.335 mmol), 3-(aminomethyl)-4,6-dimethyl-2(1 H)-pyridinone (76 mg, 0.502 mmol), 1 -hydroxy-7-azabenzotriazole (91 mg, 0.669 mmol), DMSO(10 ml_), EDC (128 mg, 0.669 mmol), and N-methylmorpholine (0.147 ml_, 1 .338 mmol). The aq. phase was extracted with EtOAc (5 x 30 ml_). The combined EtOAc extracts were dried over MgS04, filtered, and concentrated in vacuo. The final product was collected as 0.068 g (58%). LCMS E-S (M+H) = 353.3. 1H NMR (400 MHz, DMSO-de) δ ppm 0.99 - 1.14 (m, 4 H), 2.13 (s, 3 H), 2.22 (s, 4 H), 4.35 (d, J=4.80 Hz, 2 H), 5.90 (s, 1 H), 6.31 (s, 2 H), 7.41 (s, 1 H), 8.04 (s, 1 H), 8.74 (s, 1 H), 1 1.58 (br. s., 1 H).
Example 105
1 -Cyclobutyl-6-cyclopropyl-A -[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-3- methyl-1 H-pyr -carboxamide
Figure imgf000150_0002
The title compound was prepared in the same manner as described for
intermediate 56 using 1-cyclobutyl-6-cyclopropyl-3-methyl-1 H-pyrazolo[3,4-b]pyridine-4- carboxylic acid (164 mg, 0.604 mmol), 3-(aminomethyl)-4,6-dimethyl-2(1 H)-pyridinone (129 mg, 0.846mmol), 1-hydroxy-7-azabenzotriazole (165 mg, 1.209 mmol), EDC (232 mg, 1 .209 mmol), N-methylmorpholine (0.266 mL, 2.418 mmol), and DMSO(10 mL). The final product was collected as 0.240 g (98%). LCMS E-S (M+H) = 406.3. 1H NMR (400 MHz, DMSO-d6) δ ppm 1 .03 (d, J=6.06 Hz, 4 H), 1.85 (dd, J=9.35, 5.05 Hz, 3 H), 2.12 (s, 4 H), 2.16 - 2.28 (m, 6 H), 2.30 - 2.44 (m, 7 H), 2.63 (d, J=19.96 Hz, 3 H), 4.34 (d, J=4.29 Hz, 2 H), 5.31 (t, J=8.34 Hz, 1 H), 5.87 (s, 1 H), 7.01 (s, 1 H), 8.59 (br. s., 1 H), 1 1 .52 (br. s., 1 H).
Example 106
1 -Cyclopentyl-6-cyclopropyl-A -[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3^yridinyl)methyl]- 3-methyl-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000151_0001
The title compound was prepared in the same manner as described for intermediate 56 using 1-cyclopentyl-6-cyclopropyl-3-methyl-1 H-pyrazolo[3,4-b]pyridine-4- carboxylic acid (160 mg, 0.561 mmol), 3- (aminomethyl)-4,6-dimethyl-2(1 H)-pyridinone (1 19 mg, 0.785 mmol), 1-hydroxy-7-azabenzotriazole (153 mg, 1 .121 mmol), EDC (215 mg, 1 .121 mmol), N-methylmorpholine (0.247 mL, 2.243 mmol) and DMSO(10 mL). The final product was collected as 0.205 g (87%). LCMS E-S (M+H) = 420.0. 1H NMR (400 MHz, DMSO-de) δ ppm 1 .03 (d, J=6.06 Hz, 4 H), 1.85 (dd, J=9.35, 5.05 Hz, 3 H), 2.12 (s, 4 H), 2.16 - 2.28 (m, 6 H), 2.30 - 2.44 (m, 7 H), 2.63 (d, J=19.96 Hz, 3 H), 4.34 (d, J=4.29 Hz, 2 H), 5.31 (t, J=8.34 Hz, 1 H), 5.87 (s, 1 H), 7.01 (s, 1 H), 8.59 (br. s., 1 H), 1 1 .52 (br. s., 1 H).
Example 107
6-Cyclopropyl-1 -(1 ,1 -dimethylethyl)-W-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3- pyridinyl)methyl]-3-methyl-1H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000152_0001
The title compound was prepared in the same manner as described for
intermediate 56 using 6-cyclopropyl-1-(1 , 1 -dimethylethyl)-3-methyl-1 H-pyrazolo[3,4- b]pyridine-4-carboxylic acid (210 mg, 0.768 mmol), 3-(aminomethyl)-4,6-dimethyl-2(1 H)- pyridinone (158 mg, 1.037 mmol), 1-hydroxy-7-azabenzotriazole (209 mg, 1.537 mmol), EDC (295 mg, 1 .537 mmol), N-methylmorpholine (0.338 ml_, 3.07 mmol), and DMSO(10 ml_). The final product was collected as 0.280 g (89%). LCMS E-S (M+H) = 408.2. 1H NMR (400 MHz, DMSO-d6) δ ppm 1.02 (dddd, 4 H), 1.69 (m, 9 H), 2.12 (s, 3 H), 2.22 (m, 4 H), 2.33 (s, 3 H), 4.34 (d, J=4.80 Hz, 2 H), 5.87 (s, 1 H), 7.04 (s, 1 H), 8.55 (m, 1 H), 1 1 .51 (s, 1 H). Example 108
6-Cyclopropyl-A -[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(4-pyridinyl)-
1H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000152_0002
The title compound was prepared in the same manner as described for
intermediate 56 using 6-cyclopropyl-3-methyl-1-(4-pyridinyl)-1 H-pyrazolo[3,4-b]pyridine-4- carboxylic acid (89 mg, 0.302 mmol), 3-(aminomethyl)-4,6-dimethyl-2(1 H)-pyridinone (69.0 mg, 0.454 mmol), 1 -hydroxy-7-azabenzotriazole (82 mg, 0.605 mmol), DMSO(20 mL), EDC (1 16 mg, 0.605 mmol), and N-methylmorpholine (0.133 mL, 1 .210 mmol). The final product was collected as 0.1 10 g (80%). LCMS E-S (M+H) = 429.1 1H NMR (400 MHz, DMSO-d6) δ ppm 1 .15 (br. s., 4 H), 2.13 (br. s., 3H), 2.25 (br. s., 4 H), 3.34 (br. s., 3 H), 4.37 (br. s., 2 H), 5.89 (br. s., 1 H), 7.30 (br. s., 1 H), 8.33 (br. s., 2 H), 8.67 (br. s., 3 H), 1 1 .54 (br. s., 1 H).
Example 109
6-Cyclopropyl-1 -(1 -cyclopropylethyl)-A -[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3- pyridinyl)methyl]-3-methyl-1H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000153_0001
6-Cyclopropyl-1 -(1 -cyclopropylethyl)-3-methyl-1 H-pyrazolo[3,4-b]pyridine-4- carboxylic acid (186 mg, 0.652 mmol), 3-(aminomethyl)-4,6-dimethyl-2(1 H)-pyridinone (160 mg, 0.847 mmol), 1-hydroxy-7-azabenzotriazole (177 mg, 1 .304 mmol), EDC (250 mg, 1 .304 mmol) and N-methylmorpholine (0.287 mL, 2.61 mmol) were dissolved in DMSO (10 mL) and stirred at room temperature for 3 days. The reaction mixture was diluted with water (25 mL), stirred, and filtered. The product was dried and collected as a solid, 0.200 g (73%). LCMS E-S (M+H) = 420.0. 1 H NMR (400 MHz, DMSO-de) δ ppm 0.14 - 0.30 (m, 2 H), 0.32 - 0.43 (m, 1 H), 0.48 - 0.64 (m, 1 H), 0.89 - 1 .1 1 (m, 4 H), 1 .22 - 1.43 (m, 1 H), 1.55 (d, J=6.82 Hz, 3 H), 2.1 1 (s, 3 H), 2.18 - 2.28 (m, 4 H), 2.37 (s, 3 H), 4.05 - 4.21 (m, 1 H), 4.34 (d, J=4.80 Hz, 2 H), 5.87 (s, 1 H), 6.99 (s, 1 H), 8.59 (t, J=4.67 Hz, 1 H), 1 1.51 (br. s., 1 H).
Example 110
1 -Cyclohexyl-6-cyclopropyl-A -[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3^yridinyl)methyl]- 3-methyl-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000154_0001
The title compound was prepared in the same manner as described in example
109 using 6-cyclopropyl-1 -(1 -cyclohexyl)-3-methyl-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid (150 mg, 0.501 mmol), and 3-(aminomethyl)-4,6-dimethyl-2(1 H)-pyridinone (123 mg, 0.651 mmol). The product was collected as a solid, 0.190 g (87%). LCMS E-S (M+H) = 434.2. 1 H NMR (400 MHz, DMSO-de) δ ppm 0.95 - 1.07 (m, 4 H), 1.15 - 1.31 (m, 1 H), 1.36 - 1.52 (m, 2 H), 1.68 (br. s., 1 H), 1.77 - 1.99 (m, 6 H), 2.1 1 (s, 3 H), 2.18 - 2.29 (m, 4 H), 2.35 (s, 3 H), 4.33 (d, J=4.80 Hz, 2 H), 4.53 - 4.73 (m, 1 H), 5.87 (s, 1 H), 6.96 (s, 1 H), 8.58 (t, J=4.93 Hz, 1 H), 1 1.51 (s, 1 H).
Example 111
6-Cyclopropyl-W-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-3-methyl-1 -(1 - methyl-4-piperidinyl)-1 H-pyrazolo[3,4-fo]pyridine-4-carboxamide
Figure imgf000154_0002
6-Cyclopropyl-3-methyl-1 -(1 -methyl-4-piperidinyl)-1 H-pyrazolo[3,4-b]pyridine-4- carboxylic acid (150 mg, 0.477 mmol), 3-(aminomethyl)-4,6-dimethyl-2(1 H)-pyridinone (1 17 mg, 0.620 mmol), 1-hydroxy-7-azabenzotriazole (130 mg, 0.954 mmol), EDC (183 mg, 0.954 mmol) and N-methylmorpholine (0.210 ml_, 1 .909 mmol) were suspended in DMSO(10 ml.) and stirred at room temperature for 16 h. Next was added 25 ml. of water, stirred for 10 minutes, and then extracted with EtOAc (5x). The organic layers were concentrated in vacuo to afford 20 mg product. The aqueous phase was concentrated in vacuo and the crude residue purified by reverse phase HPLC (mobile phase : 10 - 60% ACN in H20, 0.1 %NH4OH) to afford additional product. The final product was collected as a solid, 0.100 g (47%). LCMS E-S (M+H) = 449.0. 1H NMR (400 MHz, DMSO-de) δ ppm 0.97 - 1.05 (m, 4 H), 1.79 (d, J=9.60 Hz, 2 H), 2.1 1 (s, 3 H), 2.15 (d, J=8.34 Hz, 3 H), 2.21 (s, 3 H), 2.23 - 2.27 (m, 3 H), 2.35 (s, 3 H), 2.91 (d, J=8.08 Hz, 2 H), 4.33 (d, J=4.80 Hz, 2 H), 4.49 - 4.72 (m, 1 H), 5.86 (s, 1 H), 6.97 (s, 1 H), 6.92 - 7.07 (m, 1 H), 8.58 (t, J=4.93 Hz, 1 H), 1 1 .50 (s, 1 H).
Example 112
6-Cyclopropyl-W-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-3-methyl-1 - (tetrahydro-2H-pyran-4-yl)-1H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000155_0001
The title compound was prepared in the same manner as described in example 109 using 6-cyclopropyl-3-methyl-1 -(tetrahydro-2H-pyran-4-yl)-1 H-pyrazolo[3,4-b]pyridine- 4-carboxylic acid (150 mg, 0.498 mmol), and 3-(aminomethyl)-4,6-dimethyl-2(1 H)- pyridinone (98 mg, 0.647 mmol). The product was collected as a solid, 0.170 g (78%). LCMS E-S (M+H) = 436.1 . 1H NMR (400 MHz, DMSO-de) δ ppm 0.95 - 1.14 (m, 4 H), 1.79 (dd, J=12.63, 2.27 Hz, 2 H), 2.05 - 2.18 (m, 5 H), 2.19 - 2.27 (m, 4 H), 2.36 (s, 3 H), 3.53 (t, J=1 1 .12 Hz, 2 H), 3.98 (dd, J=1 1.49, 3.41 Hz, 2 H), 4.33 (d, J=4.80 Hz, 2 H), 4.87 (m, J=1 1.53, 1 1 .53, 4.1 1 , 3.92 Hz, 1 H), 5.87 (s, 1 H), 6.99 (s, 1 H), 8.59 (t, J=4.93 Hz, 1 H), 1 1 .51 (s, 1 H).
Intermediate 57
Ethyl 1 -(1 -methyleth l)-6-propyl-1 H-pyrazolo[3,4-b]pyridine-4-carboxylate
Figure imgf000156_0001
To a solution of ethyl 2,4-dioxoheptanoate (446 mg, 2.397 mmol) in benzene (5 mL) was added 1 -(1 -methylethyl)-1 H-pyrazol-5-amine (300 mg, 2.397 mmol), and the mixture was stirred at 62 °C for 18 h. The reaction mixture was concentrated in vacuo and the residue purified by column chromatography (Silica gel, gradient of 0 to 100%
EtOAc/hexanes) to give 370 mg (56%) of product. LCMS E-S (M+H) = 276.1. 1H NMR (400 MHz, CHLOROFORM-d) δ ppm 0.99 - 1.12 (m, 3 H), 1.46 - 1.54 (m, 3 H), 1.58 - 1.70 (m, 6 H), 1.82 - 1 .96 (m, 2 H), 2.91 - 3.02 (m, 2 H), 4.51 (q, J = 7.2 Hz, 2 H), 5.28 - 5.44 (m, 1 H), 7.58 - 7.68 (s, 1 H), 8.27 - 8.40 (s, 1 H)
Intermediate 58
-(1 -Methylethyl)-6-propyl-1H-pyrazolo[3,4-b]pyridine-4-carboxylic acid
Figure imgf000156_0002
To a solution of ethyl 1 -(1 -methylethyl)-6-propyl-1 H-pyrazolo[3,4-b]pyridine-4- carboxylate (180 mg, 0.654 mmol) in ethanol (5 mL) and THF (1 mL) was added sodium hydroxide (1.090 mL, 3.27 mmol), and the mixture was stirred at 40 °C for 1 h. The volatiles were removed under reduced pressure. The aqueous phase was acidified to pH 3 with aq. 1 N HCI. The solid precipitate formed was collected by filtration and dried under high vacuum to give 144 mg (89%) of product, which was used for next reaction without further purification. LCMS E-S (M+H) = 248.1 1H NMR (400 MHz, DMSO-d6) δ ppm 0.82 - 1.12 (m, 3 H), 1.50 (d, J = 6.6 Hz, 6 H), 1.78 (m, 2 H), 2.80 - 3.1 1 (m, 2 H), 5.09 - 5.38 (m, 1 H), 7.47 - 7.72 (s, 1 H), 8.18 - 8.46 (s, 1 H), 13.82 (s, 1 H). Intermediate 60
Ethyl 4-[4-(methyloxy)phenyl]-2,4-dioxobutanoate
Figure imgf000157_0001
The title compound was prepared in the same manner as intermediate 29 using sodium metal (0.168 g, 7.32 mmol), 1-[4-(methyloxy)phenyl]ethanone (1 g, 6.66 mmol), and diethyl ethanedioate (0.903 ml_, 6.66 mmol). The crude product was purified using column
chromatography (0 to 100 % EtOAc/hexanes) to give 0.95 g of product (57%). LCMS E-S (M+H) = 250.9. 1H NMR (400 MHz, CHLOROFORM-d) δ ppm 1.43 (m, 3 H), 3.91 (s, 3 H), 4.41 (q, J = 6.65 Hz, 2 H), 6.94 - 7.12 (m, 3 H), 8.01 (d, J = 8.59 Hz, 2 H).
Intermediate 61
Ethyl 1 -(1 -methylethyl)-6-[4-(methyloxy)phenyl]-1H-pyrazolo[3,4-b]pyridine-4-carboxylate
Figure imgf000157_0002
To a solution of ethyl 4-[4-(methyloxy)phenyl]-2,4-dioxobutanoate (600 mg, 2.397 mmol) in benzene (5 mL) was added 1 -(1 -methylethyl)-1 H-pyrazol-5-amine (300 mg,
2.397 mmol), and the reaction mixture was stirred at 62 °C overnight. The mixture was concentrated in vacuo and the residue was dissolved into acetic acid (3 mL). The solution was stirred at reflux for 1 h and concentrated in vacuo. The residue was purified using
column chromatography (Silica gel, 0 to 100% EtOAc/hexanes) to give 530 mg (65%) of product. LCMS E-S (M+H) = 340.1 1 H NMR (400 MHz, CHLOROFORM-d) δ ppm 1 .47 - 1.57 (m, 3 H), 1.61 - 1.71 (m, 6 H), 3.88 - 3.96 (s, 3 H), 4.55 (q, J = 7.2 Hz, 2 H), 5.46 (m, 1 H), 7.03 - 7.12 (m, 2 H), 8.13 - 8.25 (m, 3 H), 8.36 - 8.43 (m, 1 H).
Intermediate 62
1 -(1 -Methylethyl)-6-[4-(methyloxy)phenyl]-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid
Figure imgf000157_0003
The title compound was prepared in the same manner as described for
intermediate 58 using ethyl 1-(1-methylethyl)-6-[4-(methyloxy)phenyl]-1 H-pyrazolo[3,4- b]pyridine-4-carboxylate (200 mg, 0.589 mmol) , Ethanol (4 mL) and THF (1 mL), and sodium hydroxide (0.982 mL, 2.95 mmol). The final product was collected as181 mg (98%). LCMS E-S (M+H) = 312.3. 1 H NMR (400 MHz, DMSO-d6) δ ppm 1 .51 - 1 .63 (m, 6 H), 3.85 (s, 3 H), 5.35 (m, 1 H), 7.1 1 (d, J = 9.1 Hz, 2 H), 8.15 (s, 1 H), 8.18 - 8.25 (m, 2 H), 8.33 (s, 1 H) , 13.95 (br. s., 1 H).
Example 113
W-[(4,6-Dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-6-[4-
(methyloxy)phen l]-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000158_0001
The title compound was prepared in the same manner as described in example 109 using 1-(1-methylethyl)-6-[4-(methyloxy)phenyl]-1 H-pyrazolo[3,4-b]pyridine-4- carboxylic acid (70 mg, 0.225 mmol), DMSO(3 mL), 3-(aminomethyl)-4,6-dimethyl-2(1 H)- pyridinone (63.6 mg, 0.337 mmol) HCI salt, N-methylmorpholine (0.099 mL, 0.899 mmol), 1-hydroxy-7-azabenzotriazole (61 .2 mg, 0.450 mmol), and EDC (86 mg, 0.450 mmol). The final product was collected as 89 mg (89%). LCMS E-S (M+H) = 446.5. 1H NMR (400 MHz, DMSO-d6) 5 ppm 1.54 (d, J = 6.8 Hz, 6 H), 2.13 (s, 3 H), 2.23 (s, 3 H), 3.85 (s, 3 H), 4.41 (d, J = 4.8 Hz, 2 H), 5.20 - 5.48 (m, 1 H), 5.91 (s, 1 H), 7.1 1 (d, J = 8.8 Hz, 2 H), 8.1 1 (s, 1 H), 8.24 (d, J = 8.8 Hz, 2 H), 8.34 (s, 1 H), 8.95 (m, 1 H), 1 1.58 (s, 1 H).
Intermediate 63
Ethyl 2,4-dioxo-4-(4-pyridinyl)butanoate
Figure imgf000158_0002
The title compound was prepared in the same manner as intermediate 29 using sodium metal (0.466 g, 20.26 mmol), 1-(4-pyridinyl)ethanone (1 g, 6.66 mmol), and diethyl ethanedioate (2.388 mL, 17.62 mmol). The crude product was purified using column chromatography (0 to 100 % EtOAc/hexanes) to give 0.95 g of product (24%). LCMS E-S (M+H) = 222.0. 1H NMR (400 MHz, CHLOROFORM-d) δ ppm 1.37 - 1.55 (m, 3 H), 4.43 (q, J = 7.07 Hz, 2 H), 7.06 - 7.16 (m, 1 H), 7.72 - 7.92 (m, 2 H), 8.82 - 8.94 (m, 2 H). Intermediate 64
Ethyl 1 -(1 -methylethyl)-6-(4- ridinyl)-1H-pyrazolo[3,4-b]pyridine-4-carboxylate
Figure imgf000159_0001
To a suspension of ethyl 2,4-dioxo-4-(4-pyridinyl)butanoate (442 mg, 1 .997 mmol) in benzene (5 mL) was added 1 -(1 -methylethyl)-1 H-pyrazol-5-amine (250 mg, 1.997 mmol), and the reaction mixture was stirred at 62 °C overnight. The reaction mixture was concentrated in vacuo and the residue was purified using column chromatography (Silica gel, 0 to 100% EtOAc/hexanes) to give 270 mg (43%) of product. LCMS E-S (M+H) = 31 1.3 1 H NMR (400 MHz, CHLOROFORM-d) δ ppm 1 .54 (t, J = 7.2 Hz, 3 H), 1 .68 (d, J = 6.6 Hz, 6 H), 4.58 (q, J = 7.1 Hz, 2 H), 5.41 - 5.60 (m, 1 H), 8.04 - 8.19 (m, 2 H), 8.29 (s, 1 H), 8.44 - 8.55 (m, 2 H), 8.76 - 8.91 (m, 1 H).
Intermediate 65
1 -(1 -Methylethyl)-6-(4-pyridinyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid
Figure imgf000159_0002
The title compound was prepared in the same manner as described for
intermediate 58 using ethyl 1-(1-methylethyl)-6-(4-pyridinyl)-1 H-pyrazolo[3,4-b]pyridine-4- carboxylate (100 mg, 0.322 mmol), ethanol (4 mL), THF (0.8 mL), and sodium hydroxide (0.537 mL, 1.61 1 mmol). The final product was collected as 88 mg (97%). LCMS E-S (M+H) = 283.2. 1H NMR (400 MHz, DMSO-d6) δ ppm 1 .58 (d, J = 6.4 Hz, 6 H), 5.40 (m, 1 H), 8.25 (d, J = 5.6 Hz, 2 H), 8.32 (s, 1 H), 8.44 (s, 1 H), 8.79 (d, J = 5.6 Hz, 2 H), 14.12 (br. s., 1 H). Example 114
W-[(4,6-Dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-6-(4-pyridinyl)-1 H- p razolo[3,4-b]pyridine-4-carboxamide
Figure imgf000160_0001
The title compound was prepared in the same manner as described in example
109 using 1-(1-methylethyl)-6-(4-pyridinyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid
(60 mg, 0.213 mmol), DMSO(2 mL), 3-(aminomethyl)-4,6-dimethyl-2(1 H)-pyridinone (60.1 mg, 0.319 mmol) HCI salt, N-methylmorpholine (0.093 mL, 0.850 mmol), 1-hydroxy-7- azabenzotriazole (57.9 mg, 0.425 mmol), and EDC (81 mg, 0.425 mmol). The final
product was collected as 66 mg (74%). LCMS E-S (M+H) = 446.5. 1H NMR (400 MHz,
DMSO-d6) δ ppm 1 .56 (d, J = 6.0 Hz, 6 H), 2.13 (br. s., 3 H), 2.23 (br. s., 3 H), 4.43 (m, 2 H), 5.37 (m, 1 H), 5.91 (br. s., 1 H), 8.15 - 8.34 (m, 3 H), 8.44 (s, 1 H), 8.78 (br. s., 2 H),
9.00 (br. s., 1 H), 1 1.58 (br. s., 1 H). Intermediate 66
Ethyl 4- -chlorophenyl)-2,4-dioxobutanoate
Figure imgf000160_0002
Sodium metal (0.143 g, 6.21 mmol) was dissolved into ethanol (3 mL) and the
resulting solution was cooled with an ice bath. A mixture of 1 -(4-chlorophenyl)ethanone
(0.756 mL, 5.65 mmol) and diethyl ethanedioate (0.766 mL, 5.65 mmol) were added
dropwise, and the reaction mixture was stirred for 16h. The reaction mixture was
quenched with ice water (5 mL) and acidified to ~pH 3 with 1 N HCI. The precipitate was collected by filtration and dried under high vacuum to give 1.39 g (97%) of product. LCMS E-S (M+H) = 255.1 . 1H NMR (400 MHz, DMSO-de) δ ppm 1.29 (m, 3 H), 4.27 (d, J = 6.57 Hz, 2 H) ,7.60 (d, J = 7.58 Hz, 2 H), 8.02 (d, J = 6.57 Hz, 2 H). Intermediate 67
Ethyl 6-(4-chlorophenyl)-1 -(1 -meth lethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxylate
Figure imgf000161_0001
The title compound was prepared in the same manner as described for
intermediate 61 using ethyl 4-(4-chlorophenyl)-2,4-dioxobutanoate (610 mg, 2.397 mmol), benzene (5 mL), 1 -(1 -methylethyl)-1 H-pyrazol-5-amine (300 mg, 2.397 mmol), and acetic acid (4ml_). The crude product was purified by column chromatography (Silica gel, eluent:
0 to 100% EtOAc /hexanes) to give 621 mg (75%) of product. LCMS E-S (M+H) = 344.1 . 1 H NMR (400 MHz, CHLOROFORM-d) δ ppm 1 .48 - 1 .57 (m, 3 H), 1 .64 - 1 .74 (m, 6 H), 4.56 (q, J = 7.1 Hz, 2 H), 5.46 (m, 1 H), 7.49 - 7.55 (m, 2 H), 8.15 - 8.18 (m, 2 H), 8.21 (s,
1 H), 8.42 (s, 1 H).
Intermediate 68
6-(4-Chlorophenyl)-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid
Figure imgf000161_0002
The title compound was prepared in the same manner as described for
intermediate 58 using ethyl 6-(4-chlorophenyl)-1 -(1 -methylethyl)-1 H-pyrazolo[3,4- b]pyridine-4-carboxylate (400 mg, 1 .163 mmol), Ethanol (6 mL), THF (1 mL), and added sodium hydroxide (1 .939 mL, 5.82 mmol) wherein the reaction time was 2h. The final product was collected as 330 mg (90%). LCMS E-S (M+H) = 315.8. 1 H NMR (400 MHz, DMSO-d6) δ ppm 1 .56 (d, J = 6.8 Hz, 6 H), 5.26 - 5.47 (m, 1 H), 7.62 (d, J = 8.6 Hz, 2 H) ,8.20 (s, 1 H), 8.28 (d, J = 8.6 Hz , 2 H), 8.38 (s, 1 H), 14.03 (s, 1 H). Example 115
6-(4-Chlorophenyl)-A -[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 methyleth l)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000162_0001
The title compound was prepared in the same manner as described in example 109 using 6-(4-chlorophenyl)-1-(1-methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid (70 mg, 0.222 mmol), DMSO(2 mL), 3-(aminomethyl)-4,6-dimethyl-2(1 H)-pyridinone HCI salt (62.7 mg, 0.333 mmol), N-methylmorpholine (0.097 mL, 0.887 mmol), 1 -hydroxy- 7-azabenzotriazole (60.3 mg, 0.443 mmol) and EDC (85 mg, 0.443 mmol). The final product was collected as 77 mg (78%). LCMS E-S (M+H) = 450.3. 1H NMR (400 MHz, DMSO-de) δ ppm 1 .55 (d, J = 6.6 Hz, 6 H), 2.13 (s, 3 H), 2.22 (s, 3 H), 4.41 (d, J = 4.6 Hz, 2 H), 5.34 (quin, J = 6.7 Hz, 1 H), 5.91 (s, 1 H), 7.64 (d, J = 8.6 Hz, 2 H), 8.19 (s, 1 H), 8.31 (d, J = 8.6 Hz, 2 H), 8.39 (s, 1 H), 8.97 (t, J = 4.7 Hz, 1 H), 1 1.58 (s, 1 H).
Intermediate 69
Ethy -dioxo-4-(2-thienyl)butanoate
Figure imgf000162_0002
The title compound was prepared in the same manner as for intermediate 66 using sodium metal (0.180 g, 7.85 mmol), 1-(3-thienyl)ethanone (0.9 g, 7.13 mmol) and diethyl ethanedioate (0.967 mL, 7.13 mmol). The product was collected as 1 .41 g (87%). LCMS E-S (M+H) = 226.9. 1H NMR (400 MHz, DMSO-de) δ ppm 1.30 (m, 3 H), 4.29 (q, J = 6.82 Hz, 2 H), 6.96 (br. s., 1 H), 7.60 - 7.81 (m, 2 H), 8.74 (br. s., 1 H). Intermediate 70
Ethyl 1 -(1 -methylethyl)-6-(3-thien l)-1 H-pyrazolo[3,4-b]pyridine-4-carboxylate
Figure imgf000163_0001
The title compound was prepared in the same manner as described for intermediate 61 using ethyl 2,4-dioxo-4-(3-thienyl)butanoate (542 mg, 2.397 mmol), benzene (5 mL), 1 -(1 -methylethyl)-1 H-pyrazol-5-amine (300 mg, 2.397 mmol), and acetic acid (5 ml). The crude product was purified using column chromatography (silica gel, eluent: 0 to 100% EtOAc/hexanes) to afford the product as 590 mg (78%). LCMS E-S (M+H) = 315.8. 1 H NMR (400 MHz, CHLOROFORM-d) δ ppm 1 .53 (t, J = 6.8 Hz, 3 H), 1 .66 (d, J = 6.8 Hz, 6 H), 4.56 (q, J = 7.1 Hz, 2 H), 5.43 (spt, J = 6.7 Hz, 1 H), 7.47 (dd, J = 5.1 , 3.0 Hz, 1 H), 7.84 - 7.95 (m, 1 H), 8.08 - 8.18 (m, 2 H), 8.32 - 8.43 (m, 1 H).
Intermediate 71
1 -(1 -Methylethyl)-6-(3-thienyl)- -pyrazolo[3,4-b]pyridine-4-carboxylic acid
Figure imgf000163_0002
The title compound was prepared in the same manner as described for intermediate 58 using ethyl 1 -(1 -methylethyl)-6-(3-thienyl)-1 H-pyrazolo[3,4-b]pyridine-4- carboxylate (400 mg, 1 .268 mmol), Ethanol (5 mL) THF (1 mL), and sodium hydroxide (2.1 14 mL, 6.34 mmol). The final product was collected as 370 mg (100%). LCMS E-S (M+H) = 287.9. 1 H NMR (400 MHz, DMSO-d6) δ ppm 1 .55 (d, J = 6.8 Hz, 6 H), 5.34 (quin, J = 6.6 Hz, 1 H), 7.72 (dd, J = 5.0, 3.0 Hz, 1 H), 7.93 (dd, J = 5.0, 1 .3 Hz, 1 H), 8.15 (s, 1 H), 8.34 (s, 1 H), 8.49 (dd, J = 3.0, 1 .3 Hz, 1 H), 13.97 (s, 1 H). Example 1 16
methyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-6-(3- thienyl)-1 H-pyrazolo 3,4-b]pyridine-4-carboxamide
Figure imgf000164_0001
The title compound was prepared in the same manner as described in example
109 using 1 -(1 -methylethyl)-6-(3-thienyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid (70 mg, 0.244 mmol), DMSO(2 mL), 3-(aminomethyl)-4,6-dimethyl-2(1 H)-pyridinone (68.9 mg, 0.365 mmol) HCI salt, N-methylmorpholine (0.107 mL, 0.974 mmol), 1 -hydroxy-7- azabenzotriazole (33.2 mg, 0.244 mmol) and EDC (93 mg, 0.487 mmol). The final
product was collected as 75 mg (73%). LCMS E-S (M+H) = 422.2. 1 H NMR (400 MHz,
DMSO-d6) δ ppm 1 .53 (d, J = 6.8 Hz, 6 H), 2.13 (s, 3 H), 2.23 (s, 3 H), 4.41 (d, J = 4.8 Hz, 2 H), 5.30 (quin, J = 6.6 Hz, 1 H), 5.91 (s, 1 H), 7.71 (dd, J = 5.0, 3.0 Hz, 1 H), 7.92 (d, J =
5.0 Hz, 1 H), 8.09 (s, 1 H), 8.34 (s, 1 H), 8.37 (d, J = 1 .8 Hz, 1 H), 8.79 - 8.95 (m, 1 H),
1 1 .58 (s, 1 H).
Intermediate 72
Ethyl 3-methyl-1 -(1 -methylethyl)-6- 4-pyridinyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxylate
Figure imgf000164_0002
The title compound was prepared in the same manner as described for intermediate 64 using ethyl 2,4-dioxo-4-(4-pyridinyl)butanoate (766 mg, 2.87 mmol), benzene (6 mL), and 3- methyl-1 -(1 -methylethyl)-1 H-pyrazol-5-amine (400 mg, 2.87 mmol). The crude product was purified using column chromatography (Silica gel ,eluent: 0 to 30% MeOH/DCM) to give 95 mg (10%) of product. LCMS E-S (M+H) = 325.3. 1 H NMR (400 MHz, CHLOROFORM-d) δ ppm 1 .48 - 1 .56 (m, 3 H), 1 .61 - 1 .73 (m, 6 H), 2.77 (s, 3 H), 4.56 (q, J =7.2 Hz, 2 H), 5.37 - 5.50 (m, 1 H), 8.02 - 8.16 (m, 3 H), 8.76 - 8.83 (m, 2 H). Intermediate 73
3-Methyl-1 -(1 -methylethyl)-6-(4-pyridinyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic
acid
Figure imgf000165_0001
The title compound was prepared in the same manner as described for
intermediate 58 using ethyl 3-methyl-1 -(1 -methylethyl)-6-(4-pyridinyl)-1 H-pyrazolo[3,4- b]pyridine-4-carboxylate (36 mg, 0.1 1 1 mmol), Ethanol (2 ml) and sodium hydroxide
(0.185 mL, 0.555 mmol). The final product was collected as 26 mg 79%). LCMS E-S
(M+H) = 297.2. 1 H NMR (400 MHz, DMSO-d6) δ ppm 1 .54 (d, J = 6.4 Hz, 6 H), 2.65 (s, 3 H), 5.35 (m, 1 H), 8.17 (s, 1 H) 8.24 (m, 2 H), 8.78 (m, 2 H).
Example 1 17
A -[(4,6-Dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-3-methyl-1 -(1 -methylethyl)-6-(4- ridinyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000165_0002
The title compound was prepared in the same manner as described in example
109 using 3-methyl-1 -(1 -methylethyl)-6-(4-pyridinyl)-1 H-pyrazolo[3,4-b]pyridine-4- carboxylic acid (20 mg, 0.067 mmol), DMSO(1 mL), 3-(aminomethyl)-4,6-dimethyl-2(1 H)- pyridinone (19.10 mg, 0.101 mmol), N-methylmorpholine (0.030 mL, 0.270 mmol), 1 - hydroxy-7-azabenzotriazole (18.37 mg, 0.135 mmol) and EDC (25.9 mg, 0.135 mmol).
The final product was collected as 29 mg (100%). LCMS E-S (M+H) = 431 .2. 1 H NMR (400 MHz, DMSO-de) δ ppm 1 .52 (d, J = 6.57 Hz, 6 H), 2.12 (s, 3 H), 2.25 (s, 3 H), 2.46 (s, 3H), 4.40 (d, J = 5.0 Hz, 2 H), 5.29 (quin, J = 6.7 Hz, 1 H), 5.89 (s, 1 H), 7.83 (s, 1 H), 8.19 (d, J = 6.1 Hz, 2 H), 8.64 - 8.89 (m, 3 H), 1 1 .55 (br. s., 1 H). Intermediate 74
Ethyl 1 ,6-bis(1 ,1 -dimethyleth l)-3-methyl-1 H-pyrazolo[3,4-b]pyridine-4-carboxylate
Figure imgf000166_0001
The title compound was prepared in the same manner as described for intermediate 61 using ethyl 5,5-dimethyl-2,4-dioxohexanoate (392 mg, 1.958 mmol), benzene (10 mL), 1 -(1 ,1 - dimethylethyl)-3-methyl-1 H-pyrazol-5-amine (300 mg, 1 .958 mmol), and acetic acid (2 mL). The crude product was purified by column chromatography (Silica gel, eluent: 0 to 100% EtOAc /hexanes) to give 530 mg (85%) of product. LCMS E-S (M+H) = 318.3. 1H NMR (400 MHz, CHLOROFORM-d) δ ppm 1.43 - 1.52 (m, 12 H), 1 .84 (s, 9 H), 2.67 (s, 3 H), 4.50 (q, J = 7.07 Hz, 2 H), 7.59 (s, 1 H).
Intermediate 75
1 ,6-Bis(1 ,1 -dimethylethyl)-3-meth -1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid
Figure imgf000166_0002
To an EtOH solution (8 mL) of ethyl 1 ,6-bis(1 , 1 -dimethylethyl)-3-methyl-1 H-pyrazolo[3,4- b]pyridine-4-carboxylate (528 mg, 1 .663 mmol) was added sodium hydroxide (2.77 mL, 8.32 mmol), and the mixture was stirred for at room temperature for 2h. The volatiles were removed under reduced pressure and the aqueous phase was acidified using 1 H HCI to ~pH 3. The precipitate was filtered, washed with water, and dried under high vacuum to give the product as 445 mg (92%). LCMS E-S (M+H) = 289.4. 1H NMR (400 MHz, DMSO-d6) δ ppm 1.49 (s, 9 H), 1.76 (s, 9 H), 2.51 (s, 3 H), 7.51 (s, 1 H).
Example 118
1 ,6-Bis(1 ,1 -dimethylethyl)-W-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-3- methyl-1 H- razolo[3,4-b]pyridine-4-carboxamide
Figure imgf000167_0001
The title compound was prepared in the same manner as described in example 109 using
1 ,6-bis(1 , 1-dimethylethyl)-3-methyl-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid (70 mg, 0.242 mmol), DMSO (2 mL), 3-(aminomethyl)-4,6-dimethyl-2(1 H)-pyridinone (68.5 mg, 0.363 mmol), N- methylmorpholine (0.106 mL, 0.968 mmol), 1-hydroxy-7-azabenzotriazole (65.9 mg, 0.484 mmol) and EDC (93 mg, 0.484 mmol). The final product was collected as 91 mg (87%). LCMS E-S (M+H) = 424.0. 1H NMR (400 MHz, DMSO-d6) δ ppm 1 H NMR (400 MHz, DMSO-d6) δ ppm 1 .38 (s, 9 H), 1.74 (s, 9 H), 2.12 (s, 3 H), 2.23 (s, 3 H), 2.35 (s, 3 H), 4.35 (d, J = 4.80 Hz, 2 H), 5.88 (s, 1 H), 7.15 (s, 1 H), 8.61 (t, J = 4.93 Hz, 1 H), 1 1.52 (s, 1 H).
Intermediate 76
Ethyl 4-[4-(methylthio)phenyl]-2,4-dioxobutanoate
The title compound was prepared in the same manner as for intermediate 66 using sodium metal (0.160 g, 6.95 mmol), 1-[4-(methylthio)phenyl]ethanone (1 .05 g, 6.32 mmol) and diethyl ethanedioate (0.856 mL, 6.32 mmol). The product was collected as 1 .58 g (59%). LCMS E-S (M+H) = 266.9. 1H NMR (400 MHz, CHLOROFORM-d) δ ppm 1 .29 - 1.50 (m, 3 H), 2.41 - 2.62 (m, 3 H), 4.23 - 4.49 (m, 2 H), 7.15 - 7.38 (m, 2 H), 7.89 (dd, J = 8.72, 2.15 Hz, 2 H). Intermediate 77
Ethyl 1 -(1 -methylethyl)-6-[4-(methylthio)phenyl]-1 H-pyrazolo[3,4-b]pyridine-4-carboxylate
Figure imgf000168_0001
The title compound was prepared in the same manner as described for
intermediate 61 using ethyl 4-[4-(methylthio)phenyl]-2,4-dioxobutanoate (851 mg, 3.20 mmol), benzene (6 mL), 1 -(1 -methylethyl)-1 H-pyrazol-5-amine (400 mg, 3.20 mmol), and acetic acid (3 mL). The crude product was purified by column chromatography (Silica gel, eluent; 0 to 100% EtOAc/hexanes) to give 870 mg (63%) of product. LCMS E-S (M+H) = 433.2. 1H NMR (400 MHz, CHLOROFORM-d) δ ppm 1 .47 - 1.57 (m, 3 H), 1.66 (d, J = 6.8 Hz, 6 H), 2.58 (s, 3 H), 4.56 (q, J = 7.2 Hz, 2 H), 5.38 - 5.55 (m, 1 H), 7.36 - 7.46 (m, 2 H), 8.12 - 8.19 (m, 2 H), 8.22 (s, 1 H), 8.40 (s, 1 H).
Intermediate 78
Ethyl 1 -(1 -methylethyl)-6-[4-(methylsulfonyl)phenyl]-1 H-pyrazolo[3,4-b]pyridine-4- carbox late
Figure imgf000168_0002
Oxone (1712 mg, 2.79 mmol) was added to a solution of ethyl 1 -(1 -methylethyl)-6- [4-(methylthio)phenyl]-1 H-pyrazolo[3,4-b]pyridine-4-carboxylate (330mg, 0.928 mmol) in acetone (6 mL) and water (2 mL), and the reaction mixture was stirred for 6 h. The
reaction mixture was quenched with water (10 mL) and then neutralized with NaHC03 solution. The mixture was extracted with EtOAc (3x) and the combined extracts dried over Na2S04, filtered and concentrated in vacuo. The residue was purified using column
chromatography (Silica gel, 0 to 100% EtOAc/hexanes) to give 295 mg (82%) of product.
LCMS E-S (M+H) = 388.1 . 1H NMR (400 MHz, CHLOROFORM-d) δ ppm 1.54 (t, J = 7.2 Hz, 3 H), 1.67 - 1.69 (d, J = 6.8 Hz, 6 H), 3.14 (s, 3 H), 4.58 (q, J = 7.2 Hz, 2 H), 5.48 (spt, J = 6.7 Hz, 1 H), 8.06 - 8.18 (m, 2 H), 8.29 (s, 1 H), 8.39 - 8.44 (m, 2 H), 8.47 (s, 1 H).
Intermediate 79
1 -(1 -Methylethyl)-6-[4-(methylsulfonyl)phenyl]-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid
Figure imgf000169_0001
The title compound was prepared in the same manner as described for
intermediate 58 using ethyl 1 -(1 -methylethyl)-6-[4-(methylsulfonyl)phenyl]-1 H- pyrazolo[3,4-b]pyridine-4-carboxylate (260 mg, 0.671 mmol), Ethanol (4 mL), THF (1 mL) and sodium hydroxide (2N, 1.1 18 mL, 3.36 mmol). The final product was collected as 231 mg (96%). LCMS E-S (M+H) = 360.1 . 1H NMR (400 MHz, DMSO-d6) δ ppm 1 .58 (d, J =
6.8 Hz , 6 H), 3.30 (s, 3H), 5.34 - 5.49 (m, 1 H), 8.1 1 (d, J= 8.6 Hz, 2 H), 8.30 (s, 1 H),
8.44 (s, 1 H), 8.51 (d, J = 8.6 Hz, 2 H).
Example 119
A -[(4,6-Dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-6-[4- (methylsulfonyl) henyl]-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000169_0002
The title compound was prepared in the same manner as described in example
109 using 1-(1-methylethyl)-6-[4-(methylsulfonyl)phenyl]-1 H-pyrazolo[3,4-b]pyridine-4- carboxylic acid (90 mg, 0.250 mmol), DMSO(2 mL), 3-(aminomethyl)-4,6-dimethyl-2(1 H)- pyridinone (70.9 mg, 0.376 mmol), N-methylmorpholine (0.1 10 mL, 1 .002 mmol), 1 - hydroxy-7-azabenzotriazole (68.2 mg, 0.501 mmol) and EDC (96 mg, 0.501 mmol). The final product was collected as 106 mg (85%). LCMS E-S (M+H) = 494.2. 1H NMR (400
MHz, DMSO-de) δ ppm 1.56 (d, J = 5.6 Hz, 6 H), 2.13 (br. s., 3 H), 2.23 (br. s., 3 H), 4.42 (m, 2 H), 5.36 (br. s., 1 H), 5.91 (br. s., 1 H), 8.1 1 (d, J = 7.3 Hz, 2 H), 8.27 (br. s., 1 H), 8.39 - 8.64 (m, 3 H), 9.01 (br. s., 1 H), 1 1 .58 (br. s., 1 H).
Intermediate 80
Ethyl 4- -bromophenyl)-2,4-dioxobutanoate
Figure imgf000170_0001
The title compound was prepared in the same manner as for intermediate 66 using sodium metal (0.404 g, 17.58 mmol), 1-(4-bromophenyl)ethanone (3.5 g, 17.58 mmol), and diethyl ethanedioate (2.384 mL, 17.58 mmol). The product was collected as 5.1 g (97%). LCMS E-S (M+H) = 299.1 . 1H NMR (400 MHz, DMSO-d6) δ ppm 1.22 - 1.37 (m, 3 H), 4.25 (d, J = 6.57 Hz, 2 H), 7.72 (m, 2 H), 7.92 (m, 2 H).
Intermediate 81
Ethyl 6-(4-bromophenyl)-1 -(1 -meth lethyl)-1H-pyrazolo[3,4-b]pyridine-4-carboxylate
Figure imgf000170_0002
The title compound was prepared in the same manner as described for
intermediate 61 using ethyl 4-(4-bromophenyl)-2,4-dioxobutanoate (956 mg, 3.20 mmol), benzene (10 mL), 1-(1-methylethyl)-1 H-pyrazol-5-amine (400 mg, 3.20 mmol), and acetic acid (4 mL). The crude product was purified using column chromatography (Silica gel, eluent: 0 to 100% EtOAc/hexanes) to give 1 .1 g (89%) of product. LCMS E-S (M+H) =
388.1 . 1H NMR (400 MHz, CHLOROFORM-d) δ ppm 1 .49 - 1.57 (t, J = 67.2 Hz , 3 H), 1.63 - 1.74 (d, J = 6.8 Hz 6 H), 4.56 (q, J=7.2 Hz, 2 H), 5.46 (spt, J=6.7 Hz, 1 H), 7.65 - 7.71 (m, 2 H), 8.04 - 8.15 (m, 2 H), 8.21 (s, 1 H), 8.42 (s, 1 H). Intermediate 82
1 -(1 -Methylethyl)-6-[4-(1 H^yrazol-4-yl)phenyl]-1H^yrazolo[3,4-b]pyridine-4-carboxylic acid
Figure imgf000171_0001
To a 5-mL microwave vial were added ethyl 6-(4-bromophenyl)-1-(1-methylethyl)- 1 H-pyrazolo[3,4-b]pyridine-4-carboxylate (80 mg, 0.206 mmol), 4-(4,4,5,5-tetramethyl- 1 ,3,2-dioxaborolan-2-yl)-1 H-pyrazole (56.0 mg, 0.288 mmol), 1 ,4-dioxane (2 ml.) and
sodium carbonate (0.206 ml_, 0.412 mmol), and the mixture was degassed with nitrogen for 10 min. Next added PdP(Ph3)4 (19.05 mg, 0.016 mmol) and the vial was sealed. The reaction mixture was irradiated (microwave) at 120 °C overnight. The reaction mixture was filtered and concentrated in vacuo. The residue was purified using reverse-phase HPLC to give 25 mg (35%) of product. LCMS E-S (M+H) = 348.1. 1H NMR (400 MHz, DMSO- cfe) δ ppm 1.58 (d, J = 6.8 Hz, 6 H), 5.38 (quin, J = 6.6 Hz, 1 H), 7.81 (d, J = 8.3 Hz, 2 H), 8.14 - 8.30 (m, 4H), 8.36 (s, 1 H). Example 120
W-[(4,6-Di methyl -2 -oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-6-[4-(1 H-pyrazol-4- yl)phenyl -1H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000171_0002
The title compound was prepared in the same manner as described in example
109 using 1-(1-methylethyl)-6-[4-(1 H-pyrazol-4-yl)phenyl]-1 H-pyrazolo[3,4-b]pyridine-4- carboxylic acid (20 mg, 0.058 mmol), DMSO(1 ml_), 3-(aminomethyl)-4,6-dimethyl-2(1 H)- pyridinone (16.29 mg, 0.086 mmol), N-methylmorpholine (0.025 ml_, 0.230 mmol), 1- hydroxy-7-azabenzotriazole (15.67 mg, 0.1 15 mmol) and EDC (22.07 mg, 0.1 15 mmol). The final product was collected as 12 mg (43%). LCMS E-S (M+H) = 482.0. 1H NMR (400 MHz, DMSO-d6) δ ppm 1.56 (d, J = 6.4 Hz, 6 H), 2.14 (s, 3 H), 2.24 (s, 3 H), 4.42 (d, J =
4.8 Hz, 2 H) 5.27 - 5.47 (m, 1 H), 5.91 (s, 1 H), 7.79 (d, J = 8.3 Hz, 2 H), 8.04 (br. s., 1
H), 8.15 - 8.21 (m, 1 H), 8.27 (d, J = 8.3 Hz, 2 H), 8.35 - 8.42 (m, 1 H), 8.99 (t, J = 4.8
Hz, 1 H), 1 1.58 (br. s., 3 H).
Intermediate 83
Ethyl 1 -(1 ,1 -dimethylethyl)-3-methyl-6-[4-(methyloxy)phenyl]-1 H-pyrazolo[3,4- b py ri d i ne-4-carboxy I ate
Figure imgf000172_0001
The title compound was prepared in the same manner as described for
intermediate 61 using ethyl 4-[4-(methyloxy)phenyl]-2,4-dioxobutanoate (327 mg, 1.305 mmol), benzene (8 mL), 1 -(1 ,1 -dimethylethyl)-3-methyl-1 H-pyrazol-5-amine (200 mg,
1.305 mmol), and acetic acid (2 mL). The crude product was purified using column
chromatography (Silica gel, eluent: 0 to 100% EtOAc/hexanes) to give 0.41 g (85%) of product. LCMS E-S (M+H) = 368.1 . 1H NMR (400 MHz, DMSO-d6) δ ppm 1.81 (s, 9 H), 2.55 (s, 3 H), 7.1 1 (d, J = 8.84 Hz, 2 H), 7.89 (s, 1 H), 8.16 (d, J = 8.84 Hz, 2 H).
Intermediate 84
1 -(1 ,1 -Dimethylethyl)-3 -methyl -6-[4-(methyloxy)phenyl]-1 H-pyrazolo[3,4-b]pyridine- 4-carboxylic acid
Figure imgf000172_0002
The title compound was prepared in the same manner as described for intermediate 75 using ethyl 1 -(1 , 1-dimethylethyl)-3-methyl-6-[4-(methyloxy)phenyl]-1 H-pyrazolo[3,4-b]pyridine-4- carboxylate (400 mg, 1 .089 mmol), 3N sodium hydroxide (1 .814 mL, 5.44 mmol), and EtOH (6 mL). The product was collected as 347 mg (94%). LCMS E-S (M+H) = 340.2. 1H NMR (400 MHz, DMSO-de) δ ppm 1 .81 (s, 9 H), 2.55 (s, 3 H), 7.1 1 (d, J = 8.84 Hz, 2 H), 7.89 (s, 1 H), 8.16 (d, J=8.84 Hz, 2 H). Example 121
1 -(1 ,1 -Dimethylethyl)-A -[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-3- methyl-6-[4-(meth loxy)phenyl]-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000173_0001
To a solution of 1 -(1 , 1 -dimethylethyl)-3-methyl-6-[4-(methyloxy)phenyl]-1 H- pyrazolo[3,4-b]pyridine-4-carboxylic acid (70 mg, 0.206 mmol) in DMSO (1 mL) were added 3-(aminomethyl)-4,6-dimethyl-2(1 H)-pyridinone (19.10 mg, 0.101 mmol), N- methylmorpholine (0.030 mL, 0.270 mmol), 1 -hydroxy-7-azabenzotriazole (18.37 mg, 0.135 mmol) and EDC (25.9 mg, 0.135 mmol), and the mixture was stirred overnight at room temperature. The reaction mixture was quenched with water (10 mL), stirred for 10 min.,and filtered. The contents were dried under high vacuum and collected as 71 mg (71 %). LCMS E-S (M+H) = 474.1 . 1 H NMR (400 MHz, DMSO-d6) δ ppm 1 .79 (s, 9 H), 2.12 (s, 3 H), 2.25 (s, 3 H), 2.39 (s, 3 H), 3.33 (s, 1 H), 4.38 (d, J = 4.80 Hz, 2 H), 5.88 (s, 1 H), 7.10 (d, J = 8.84 Hz, 2 H), 7.61 (s, 1 H), 8.16 (d, J = 8.84 Hz, 2 H), 8.70 (s, 1 H), 1 1 .53 (s, 1 H).
Intermediate 85
Ethyl 1 -(1 ,1 -dimethylethyl)-3-methyl-6-{[(trifluoromethyl)sulfonyl]oxy}-1 H- pyrazolo[3,4-b]pyridine-4-carboxylate
Figure imgf000173_0002
Trifluoromethanesulfonic anhydride (0.457 mL, 2.70 mmol) was added dropwise to a solution of ethyl 1 -(1 , 1 -dimethylethyl)-3-methyl-6-oxo-6,7-dihydro-1 H-pyrazolo[3,4-b]pyridine-4 carboxylate (500 mg, 1 .803 mmol) in pyridine (10 mL), and the reaction mixture was stirred at room temperature for 16 h. The contents were diluted with EtOAc and washed with saturated NaHC03, and then brine. The organic layer was dried over MgS04, filtered, and concentrated vacuo. The crude residue was purified via silica gel chromatography (0% to 100% DCM:Hex) to afford the final product as a solid, 670 mg (91 %). LCMS E-S (M+H) = 410.0. 1H NMR (400 MHz, DMSO-d6) δ ppm 1.38 (t, J = 7.07 Hz, 3 H), 1.71 (s, 9 H), 2.58 (s, 3 H), 4.46 (q, J = 7.07 Hz, 2 H), 7.63 (s, 1 H).
Intermediate 86
Ethyl 1 -(1 -dimethylethyl)-3-methyl-6-(3^yri
car box I ate
Figure imgf000174_0001
Ethyl 1-(1 , 1-dimethylethyl)-3-methyl-6-{[(trifluoromethyl)sulfonyl]oxy}-1 H-pyrazolo[3,4- b]pyridine-4-carboxylate (100 mg, 0.244 mmol), 3-pyridinylboronic acid (39.0 mg, 0.318 mmol), aq. saturated NaHC03 (1 mL) and 1 ,4-dioxane (3 mL) were degassed with nitrogen (10 min) followed by addition of PdCI2(dppf)-CH2CI2 adduct (9.97 mg, 0.012 mmol). The sealed mixture was irradiated (microwave) at 120 °C for 40 min. The reaction mixture was diluted with EtOAc and filtered. The filtrate was concentrated in vacuo and the crude residue was purified by silica gel chromatography (0 to 100% EtOAc/hexanes). The final product was collected as a solid, 72 mg (87%). LCMS E-S (M+H) = 339.3. 1 H NMR (400 MHz, DMSO-d6) δ ppm 1.46 - 1.57 (m, 3 H), 1.90 (s, 9 H), 2.72 (s, 3 H), 4.45 - 4.64 (m, 2 H), 7.47 (dd, J=8.08, 4.80 Hz, 1 H), 8.03 (s, 1 H), 8.46 (dt, J= 8.08, 2.02 Hz, 1 H), 8.72 (dd, J=4.80, 1 .52 Hz, 1 H), 9.43 (d, J=1.52 Hz, 1 H).
Intermediate 87
1 -(1 ,1 -Dimethylethyl)-3-methyl-6-(3-pyridinyl)-1 H-pyrazolo[3,4-b]pyridine-4- carboxylic acid
Figure imgf000174_0002
Ethyl 1-(1 , 1-dimethylethyl)-3-methyl-6-(3-pyridinyl)-1 H-pyrazolo[3,4-b]pyridine-4- carboxylate (68 mg, 0.201 mmol) was suspended in ethanol (2 mL) followed by addition of sodium hydroxide (0.335 mL, 1.005 mmol). The mixture was stirred at room temperature for 2. 5h. The mixture was concentrated and the residue was diluted with water (1 mL) and acidified to pH 3 using 1 N HCI. The precipitate was collected by filtration and further dried under high vacuum to give the product as an HCI salt, 60 mg (86%). LCMS E-S (M+H) = 31 1.5. 1H NMR (400 MHz, DMSO-d6) δ ppm 1 .82 (s, 9 H), 2.60 (s, 3 H), 7.60 (dd, J = 7.96, 4.67 Hz, 1 H), 8.08 (s, 1 H), 8.57 (dt, J = 8.34, 1.89 Hz, 1 H), 8.70 (dd, J = 4.80, 1.52 Hz, 1 H), 9.39 (d, J = 1 .52 Hz, 1 H), 14.03 (br. s., 1 H).
Example 122
1 -(1 ,1 -Dimethylethyl)-A -[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-3-methyl-6-(3- pyridinyl)-1 H- razolo[3,4-b]pyridine-4-carboxamide.
Figure imgf000175_0001
The title compound was prepared in the same manner as described in example 121 using 1-(1 , 1 -dimethylethyl)-3-methyl-6-[3-pyridinyl]-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid (58 mg, 0.167 mmol), 3-(aminomethyl)-4,6-dimethyl-2(1 H)-pyridinone (47 mg, 0.251 mmol), N- methylmorpholine (0.092 ml_, 0.836 mmol), 1 -hydroxy-7-azabenzotriazole (46 mg, 0.334 mmol), EDC (64 mg, 0.334 mmol), and DMSO (1 ml_). The product was collected as 73 mg (96%).
LCMS E-S (M+H) = 445.1 . 1H NMR (400 MHz, DMSO-d6) δ ppm 1 .81 (s, 9 H), 2.12 (s, 3 H), 2.25 (s, 3 H), 2.42 (s, 3 H), 4.39 (d, J = 5.05 Hz, 2 H), 5.89 (s, 1 H), 7.59 (dd, J = 8.08, 4.80 Hz, 1 H), 7.78 (s, 1 H), 8.55 (dt, J = 8.15, 1 .86 Hz, 1 H), 8.68 (dd, J = 4.80, 1 .52 Hz, 1 H), 8.74 (t, J = 5.05 Hz, 1 H) 9.38 (d, J = 1.77 Hz, 1 H), 1 1.54 (s, 1 H).
Example 123
3-Bromo-A -[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-6- phenyl-1H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000175_0002
3-Bromo-1-(1-methylethyl)-6-phenyl-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid (93 mg, 0.258 mmol), 3-(aminomethyl)-4,6-dimethyl-2(1 H)-pyridinone hydrochloride (64 mg, 0.336 mmol), HOBT (60 mg, 0.387 mmol) and EDC (74 mg, 0.387 mmol) were suspended in DMSO (14 ml.) and stirred at room temperature for 10 minutes, after which time DIEA (0.9 ml, 5.16 mmol) was added. After stirring for 2 h, 4-methylmorpholine (1 ml, 9.04 mmol) was added. The reaction mixture was stirred first at room temperature for 21 h, and then at 80 °C (aluminum heating block) for 31 h. The reaction mixture was cooled to room temperature and then added dropwise to a cold, slightly basic solution (pH ~ 8-10) of water (100 ml.) and 1 N Na2C03 (8 ml_). After stirring for 20 min., the contents were extracted with EtOAc (2 x 100 ml_). The combined organic layers are washed with brine, dried over Na2S04, filtered, and concentrated to a residue. The crude residue was dissolved in 10%MeOH/DCM and purified by silica gel chromatography (eluent: 10-95% gradient EtOAc/Hexanes and then 10% (5%NH4OH/MeOH)/DCM and EtOAc, 10-90% gradient). The product was collected as an off-white solid. (35 mg, 27%). LCMS E-S (M+H) = 494.1 / 496.0. 1H N MR (400 MHz, CHLOROFORM-d) δ 1 1 .58 (br. s., 1 H), 8.1 1 (dd, J = 1.89, 7.71 Hz, 2H), 7.68 (br. s., 1 H), 7.60 (br. s., 1 H), 7.44 - 7.53 (m, 3H), 5.92 (s, 1 H), 5.42 (quin, J = 6.69 Hz, 1 H), 4.63 (br. s., 1 H), 2.41 (br. s., 3H), 2.1 1 (br. s., 3H), 1 .62 (d, J = 6.57 Hz, 6H). Intermediate 88
3-Bromo-1 -(1 -methylethyl)-6-phenyl-1H-pyrazolo[3,4-b]pyridine-4-carboxylic acid
Figure imgf000176_0001
Ethyl 3-bromo-1 -(1 -methylethyl)-6-phenyl-1 H-pyrazolo[3,4-b]pyridine-4-carboxylate (1 12 mg, 0.288 mmol) was suspended in THF (0.5 ml.) and ethanol (1.5 ml_), followed by addition of 3N NaOH (150 μΙ, 0.433 mmol). The reaction mixture was stirred at 55 °C for 3 hours, and then allowed to cool to room temperature. The reaction mixture was diluted with water (1 .5 ml_), cooled in an ice bath, and acidified with 1 N HCI in a dropwise manner. The mixture was extracted with EtOAc. The organic layer was separated and concentrated in vacuo to afford a solid that dried on the high vacuum 3 hours. The product was collected as a pale yellow solid (93.6 mg, 90%). LCMS E-S (M+H) = 360.0 / 362.2. 1H NMR (400 MHz, MeOD) 58.22 (dd, J = 1 .52, 8.08 Hz, 2H), 8.05 (s, 1 H), 7.48 - 7.59 (m, 3H), 5.47 (quin, J= 6.76 Hz, 1 H), 1.63 (d, J = 6.82 Hz, 6H) Intermediate 89
Ethyl 3-bromo-1 -(1 -methyle pyrazolo[3,4-b]pyridine-4-carboxylate
Figure imgf000177_0001
Ethyl 1 -(1 -methylethyl)-6-phenyl-1 H-pyrazolo[3,4-b]pyridine-4-carboxylate (120 mg, 0.388 mmol), was suspended in acetic acid (2 ml.) followed by addition of bromine (26 μΙ, 0.504 mmol). The reaction mixture was stirred with heating at 80 °C. After 1 h, a second portion of bromine was added (26 μΙ, 0.504 mmol) and the reaction mixture heated at 80 °C for an additional 2h. After cooling to room temperature, the solution was added to a saturated aqueous solution of sodium bicarbonate (6 ml.) and extracted with
dichloromethane (2 x 10 ml.) The combined organic layers were concentrated in vacuo. The crude product was purified by silica gel chromatography (eluent: EtOAc/Hexanes, 0- 50% gradient). The product was collected as a white powder (1 12 mg, 74%). LCMS E-S (M+H) = 388.0 / 390.2. 1 H N MR (400 MHz, CHLOROFORM-d) δ 8.18 (dd, J = 1 .52, 8.08 Hz, 2H), 8.00 (s, 1 H), 7.48 - 7.58 (m, 3H), 5.47 (quin, J = 6.76 Hz, 1 H), 4.58 (q, J = 7.16 Hz, 2H), 1 .65 (d, J = 6.82 Hz, 6H), 1 .52 (t, J = 7.20 Hz, 3H).
Intermediate 90
Ethyl 1 -(1 -methylethyl)- -phenyl-1 H-pyrazolo[3,4-b]pyridine-4-carboxylate
Figure imgf000177_0002
The title compound was prepared in the same manner as described for intermediate 61 using ethyl 2,4-dioxo-4-phenylbutanoate (5 g, 22.7 mmol) , 1 -(1 - methylethyl)-1 H-pyrazol-5-amine (2.84 g, 22.7 mmol) , benzene (70 ml_), and acetic Acid (44 ml.) The crude product was purified by silica gel chromatography (eluent: 0 to 100% EtOAc/hexanes) to give 6.72 g (95%) as a pale yellow solid. LCMS E-S (M+H) = 310.5. 1 H NMR (400 MHz, DMSO-d6) δ 8.39 (s, 1 H), 8.17 - 8.28 (m, 3H), 7.49 - 7.64 (m, 3H), 5.38 (quin, J = 6.69 Hz, 1 H), 4.49 (q, J = 7.07 Hz, 2H), 1 .57 (d, J = 6.57 Hz, 6H), 1 .44 (t, 3H). Example 124
W-[(4,6-Dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-6-(2- nitrophenyl)-1 H-pyr carboxamide
Figure imgf000178_0001
1 -(1 -Methylethyl)-3-nitro-6-phenyl-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid (24 mg, 0.074 mmol), 3-(aminomethyl)-4,6-dimethyl-2(1 H)-pyridinone hydrochloride (18 mg, 0.096 mmol), HOAT (17 mg, 0.1 10 mmol) and EDC (21 mg, 0.1 10 mmol) were suspended in DMSO (800 μΙ_) and the reaction mixture stirred at room temperature. Next added 4- methylmorpholine (41 μΙ, 0.368 mmol), and the reaction was stirred at room temperature for 16 h. The reaction mixture was added dropwise to cold, slightly basic solution of water (3 ml.) and 1 N Na2C03 (0.5 ml_). After stirring for 20 min., the precipitated solids were collected via vacuum filtration and washed with water. The solid was dried on the high vacuum (72 h). The product, was collected as an off-white powder (28 mg, 82%). LCMS E-S (M+H) = 461 .1 . 1 H NMR (400 MHz, DMSO-d6) δ 9.01 (br. s., 1 H), 8.42 (s, 1 H), 8.02 (ddd, J = 1 .14, 4.99, 7.77 Hz, 2H), 7.99 (s, 1 H), 7.86 (td, J = 1 .26, 7.58 Hz, 1 H), 7.72 - 7.77 (m, 1 H), 5.90 (s, 1 H), 5.04 (quin, J = 6.69 Hz, 1 H), 4.41 (d, J = 4.55 Hz, 1 H), 3.34 (s, 2H), 2.22 (s, 3H), 2.12 (s, 3H), 1 .49 (d, J = 6.57 Hz, 6H).
Intermediate 91
1 -(1 -Methylethyl)-6-(2-nitrophenyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid
Figure imgf000178_0002
Ethyl 1 -(1 -methylethyl)-3-nitro-6-phenyl-1 H-pyrazolo[3,4-i)]pyridine-4-carboxylate was suspended in THF (0.5 ml) and ethanol (1 ml_), followed by addition of 3N NaOH aq (50 μΙ_). The reaction mixture was heated at 55 °C for 1 h, and then allowed to cool to room temperature. The volatiles were removed in vacuo and the residue was dissolved in water (1 .5 ml_). The reaction mixture was cooled in an ice bath and acidified by dropwise addition of 1 N HCI. After stirring for 20 minutes, the precipitated solids were collected by vacuum filtration dried under high vacuum overnight for 16 h. The product was collected as a white solid (24.5 mg, 76%). LCMS E-S (M+H) = 327.2. 1H NMR (400 MHz, DMSO-d6) δ 14.14 (br. s., 1 H), 8.43 (s, 1 H), 8.04 (dd, J = 1.14, 7.96 Hz, 1 H), 7.98 - 8.02 (m, J = 1.26 Hz, 2H), 7.86 (td, J = 1.26, 7.58 Hz, 1 H), 7.72 - 7.80 (m, 1 H), 5.09 (qd, J = 6.57, 6.74 Hz, 1 H), 1 .51 (d, J = 6.57 Hz, 6H).
Intermediate 92
Ethyl 1 -(1 -methylethyl)-6-(2-nitrophen l)-1 H-pyrazolo[3,4-b]pyridine-4-carboxylate
Figure imgf000179_0001
Ethyl 1-(1-methylethyl)-6-phenyl-1 H-pyrazolo[3,4-b]pyridine-4-carboxylate (120 mg, 0.388 mmol) was dissolved in concentrated nitric acid (1.7 mL) followed by addition of concentrated sulfuric acid (0.200 mL), and the reaction mixture was stirred at room temperature. After 40 min., the reaction mixture was cooled in an ice bath and with stirring was diluted with water (3 mL) and then slowly with saturated Na2C03 (1 mL). The reaction mixture was extracted with EtOAc (2 x 6 mL). The combined organic layers were concentrated in vacuo and the crude product purified by silica gel chromatography (eluent: EtOAc/Hexanes, 0-70% gradient). The product, was collected as a white solid (35 mg, 26%). LCMS E-S (M+H) = 355.2. 1H NMR (400 MHz, CHLOROFORM-d) δ 8.46 (s, 1 H), 7.99 (s, 1 H), 7.94 (dd, J = 1.14, 7.96 Hz, 1 H), 7.76 - 7.81 (m, 1 H), 7.73 (td, J = 1 .26, 7.58 Hz, 1 H), 7.59 - 7.66 (m, 1 H), 5.25 (ddd, J = 6.57, 6.69, 13.52 Hz, 1 H), 4.51 - 4.59 (m, 2H), 1.61 (d, J = 6.82 Hz, 6H), 1.52 (t, J = 7.07 Hz, 3H).
Example 125
W-[(4,6-Dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-1 -(1 -methylethyl)-6-propyl-1 H- p razolo[3,4-b]pyridine-4-carboxamide
Figure imgf000180_0001
The title compound was prepared in the same manner as described in example
109 using 1 -(1 -methylethyl)-6-propyl-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid (70 mg, 0.283 mmol), DMSO(3 ml_), 3-(aminomethyl)-4,6-dimethyl-2(1 H)-pyridinone (80 mg, 0.425 mmol), N-methylmorpholine (0.124 ml_, 1 .132 mmol), 1 -hydroxy-7-azabenzotriazole (77 mg, 0.566 mmol), and EDC (109 mg, 0.566 mmol). The collected solid was washed with water and methanol and dried under high vacuum to give 73 mg (68%) of product. LCMS E-S (M+H) = 382.3 1 H NMR (400 MHz, DMSO-d6) δ ppm 0.82 - 1 .10 (m, 3 H), 1 .48 (d, J = 6.8 Hz, 6 H), 1 .77 (m, 2 H), 2.13 (s, 3 H), 2.21 (s, 3 H), 2.85 (t, J = 7.6 Hz, 2 H), 4.36 (d, J = 4.8 Hz, 2 H), 5.20 (m, 1 H), 5.90 (s, 1 H), 7.38 - 7.58 (m, 1 H), 8.26 (s, 1 H), 8.73 (t, J = 4.7 Hz, 1 H), 1 1.57 (br. s., 1 H).
Intermediate 93
3-(Aminomethyl)-6-ethyl-4-methyl-2(1 H)-pyridinone
Figure imgf000180_0002
Step 1
2,4-Hexanedione (2.283 g, 20 mmol) and cyanacetamide (1 .682 g, 20.00 mmol) were added to ethanol (20 ml_). The contents were initially heterogenous, but gradually dissolved upon reaching ca. 70 °C. Next added piperidine (1.976 ml_, 20.00 mmol) and stirred with warming to reflux. After 30 min, the heterogenous contents were removed from heating and allowed to stir with cooling to room temperature. The contents were filtered in vacuo to afford a light yellowish colored solid and yellow filtrate. The collected solid filter cake was copiously washed with water which removed the yellow color. The final product was collected as fine white crystalline solid (1.66 g after drying). LCMS E-S (M+H) = 163.2. 1 H NMR (400 MHz, CHLOROFORM-d) 5ppm 1.34 (t, J=7.58 Hz, 3H) 2.47 (s, 3H) 2.71 (q, J=7.58 Hz, 2H), 6.12 (s, 1 H).
Step 2
To a stirred solution of 6-ethyl-4-methyl-2-oxo-1 ,2-dihydro-3-pyridinecarbonitrile
(1 .462 g, 9.01 mmol) in Methanol (50 mL), cooled to 0 °C were added di- tertbutoxycarbonyl anhydride (4.19 mL, 18.03 mmol) and nickel(ll) chloride*hexahydrate (0.214 g, 0.901 mmol). Sodium borohydride (2.387 g, 63.1 mmol) was added portionwise over 30 min, then the mixture was allowed to warm to room termperature and stirred overnight. Additional sodium borohydride (2.387 g, 63.1 mmol) was added and the reaction stirred overnight again. Diethylenetriamine (0.979 mL, 9.01 mmol) was added, and the reaction mixture was stirred for 30 min. at room temperature. The solvent was evaporated, and the residue was dissolved in ethyl acetate (20 mL) and extracted with sodium bicarbonate (2 x 10 mL). The organics were dried over sodium sulfate, and concentrated. The residue was dissolved in chloroform (25 mL) and TFA (3.47 mL, 45.1 mmol) was added. The reaction was heated to 50 °C for 3 h, then cooled to room temperature and concentrated. The reaction mixture was concentrated in vacuo and the crude product was purified by reverse phase HPLC (eluent; 0-30% gradient ACN in H20, 0.1 % TFA) to afford the title compound (TFA salt, 1.37 g, 54.1 % yield) as a white solid. LC-MS(ES) m/z = 167 [M+H]+. 1H NMR (400 MHz, DMSO-d) δ ppm 1 .12 (t, J=7.58 Hz, 3H), 2.20 (s, 3H), 2.44 (q, J=7.58 Hz, 2H), 3.78 - 3.83 (m, 2H), 6.01 (s, 1 H), 7.75 - 7.9 (br s, 3H), 1 1 .86 (s, 1 H).
Intermediate 94
3-(Aminomethyl)-5-fluoro-4,6-dimethyl-2(1 H)-pyridinone
Figure imgf000181_0001
Step 1
2,4-Pentanedione (12 g, 120 mmol) was suspended in deuterated MeCN (39 mL), to which was added Selectfluor (44.7 g, 120 mmol) in one portion. The resulting paste was then heated in an oil bath to 50 °C. After 5 min, an exotherm was observed, and the mixture was removed from the oil bath. The mixture was then heated at 50 °C for 1 h. The mixture was then aged at room temperature for 16 h, and then distilled under vacuum. The desired product distilled at 40-60 °C at 10-15 torr and was collected as a light yellowish liquid (1.8 mL, 1 .95 g). A less pure fraction (ca. 80%) distilled at 25-35 °C at 15- 20 torr, and was collected as a light yellowish liquid (3.33 g). 1H NMR (400 MHz,
ACETONITRILE-ds) δ ppm 2.24 (s, 3 H), 2.26 (s, 3 H), 5.46 (d, J=48.0 Hz, 1 H).
Step 2
A mixture of 3-fluoro-2,4-pentanedione (247 mg, 2.10 mmol), cyanoacetamide
(176 mg, 2.1 mmol, 1 equiv) and piperidine (0.21 mL, 2.1 mmol, 1 equiv) in 3 mL of EtOH was heated at 85 °C for 16 h, resulting in a clear but dark brown solution. The mixture was concentrated in vacuo. The residue was taken up in 2 mL of 1 N HCI to give a suspension, which was filtered. The tan solids collected were dried under vacuum to give the desired product as 80 mg. 1H NMR (400 MHz, CHLOROFORM-d) δ ppm 2.33 (s, 3H), 2.42 (s, 3H).
Step 3
5-Fluoro-4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinecarbonitrile (1 .0 g, 6.02 mmol) was suspended in 50 mL of HOAc at room temperature to give a dark brownish solution. NaOAc (1.0 g, 12.3 mmol, 2 equiv), Pt02 (20 mg) and 5% Pd/C (1.3 g) were charged into a 500 mL Parr bottle, followed by wetting with some acetic acid under nitrogen. The substrate solution was then added, and rinsed with another 50 mL of acetic acid. The mixture was hydrogenated under a pressure of 40 psi. There was initial drop of hydrogen pressure and the vessel was refilled. The mixture was hydroganted for 6 h. The mixture was filtered through Celite, and concentrated in vacuo to give a solid residue, which was suspended in 6 mL of cone. HCI to give a paste. The paste was filtered, and the cake was washed with 1 mL of cone HCI (in duplicate). The filtrate was concentrated in vacuo. The solid residue dissolved in 0.4 mL of cone. HCI and 6 mL of EtOH as a suspension, which was stored in the freezer for 2h, followed by filtration. The cake was washed with 2 mL of cold EtOH (in duplicate). The solids were dried under vacuum at room temperature for 18h to give the title compound as a cream-colored solid (1 .03 g). LC-MS (ES) m/z = 171
[M+H]+ and a dominant peak at 154. 1H NMR (400 MHz, DMSO-d6) δ ppm , 2.20 (d, J=3.0 Hz, 3 H), 2.22 (d, J=2.0 Hz, 3 H), 3.83 (q, J=5.2 Hz, 2 H), 7.99 (br. s., 3 H), 1 1.99 (br. s., 1 H). Example 126
N-((4,6-dimethyl-2-oxo-1 ,2-dihydropyridin-3-yl)methyl)-1 -isopropyl-3-methyl-6- (pyridin-3-yl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000183_0001
In a 25 ml. sealable tube under nitrogen were combined 6-chloro-N-[(4,6-dimethyl-2-oxo- 1 ,2-dihydro-3-pyridinyl)methyl]-3-methyl-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4- carboxamide (90 mg, 0.23 mmol) and 3-pyridinylboronic acid (42.8 mg, 0.35 mmol) in dioxane/water (3:1 ml_). PdCI2(dppf)-CH2CI2 adduct (9.47 mg, 0.012 mmol) was added and the resulting mixture was degassed with nitrogen for 10 min. Sodium carbonate (58.5 mg, 0.7 mmol) was added, the vessel was sealed, and the insoluble mixture was heated in a microwave to 1 10 °C for 20 min. Upon cooling, water was added and solids that precipitated were filtered, dissolved in DCM and purified by Si02 chromatography (eluent: gradient 0 to 80:20:2 DCM/MeOH/Nh^OH). The product containing fractions were evaporated and suspended in EtOH/EtOAc (1 :1 ). The contents were sonicated, solids that precipitated were filtered, washed with hexanes and dried to afford the title compound as a white solid (78 mg, 77%). 1H NMR (400 MHz, DMSO-c/6) δ ppm 1 1.53 (br. s., 1 H) 9.39 (d, J=1.52 Hz, 1 H) 8.76 (t, J=5.05 Hz, 1 H) 8.69 (dd, J=4.67, 1.64 Hz, 1 H) 8.57 (dt, J=8.27, 1 .80 Hz, 1 H) 7.78 (s, 1 H) 7.57 (dd, J=8.08, 5.56 Hz, 1 H) 5.89 (s, 1 H) 5.28 (quin, J=6.69 Hz, 1 H) 4.41 (s, 1 H) 4.39 (s, 1 H) 2.46 (s, 3 H) 2.25 (s, 3 H) 2.12 (s, 3 H) 1 .52 (s, 3 H) 1 .51 (s, 3 H); LC-MS (ES) m/z = 431.1 [M+H]+. Examples 127-129 were prepared from 6-chloro-N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3- pyridinyl)methyl]-3-methyl-1 -(1-methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide and the appropariate boronic acid reagent using the conditions described in the above example. The heating temperatures were between 100 -1 10 °C, and product triturations were performed using either EtOH/EtOAc or EtOAc/hexanes. Example 127
N-((4,6-dimethyl-2-oxo-1 ,2-dihydropyridin-3-yl)methyl)-1 -isopropyl-3-methyl-6- phenyl-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000184_0001
1H NMR (400 MHz, DMSO-d6) δ ppm 1 1.53 (s, 1 H) 8.76 (t, J=4.93 Hz, 1 H) 8.22 (d, J=1.52 Hz, 1 H) 8.20 (s, 1 H) 7.69 (s, 1 H) 7.47 - 7.57 (m, 3 H) 5.89 (s, 1 H) 5.26 (quin, J=6.69 Hz, 1 H) 4.40 (s, 1 H) 4.39 (s, 1 H) 2.45 (s, 3 H) 2.25 (s, 3 H) 2.12 (s, 3 H) 1.52 (s, 3 H) 1 .50 (s, 3H). LC-MS (ES) m/z = 429.9[M+H]+.
Example 128
N-((4,6-dimethyl-2-oxo-1 ,2-dihydropyridin-3-yl)methyl)-6-(4-
((dimethylamino)methyl)phenyl)-1 -isopropyl-3-methyl-1 H-pyrazolo[3,4-b]pyridine-4- carboxamide
Figure imgf000184_0002
1H NMR (400 MHz, DMSO-d6) δ ppm 1 1.52 (s, 1 H) 8.74 (t, J=5.05 Hz, 1 H) 8.17 (s, 1 H) 8.15 (s, 1 H) 7.67 (s, 1 H) 7.46 (s, 1 H) 7.44 (s, 1 H) 5.89 (s, 1 H) 5.26 (quin, J=6.63 Hz, 1 H) 4.40 (br. s., 1 H) 4.39 (br. s., 1 H) 3.46 (s, 2 H) 2.45 (s, 3 H) 2.25 (s, 3 H) 2.18 (s, 6 H) 2.08 - 2.14 (m, 3 H) 1.51 (s, 3 H) 1 .50 (s, 3 H). LC-MS (ES) m/z = 487.1 [M+H]+. Example 129
N-((4,6-dimethyl-2-oxo-1 ,2-dihydropyridin-3-yl)methyl)-6-(4-fluorophenyl)-1 - isopropyl-3-methyl-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000185_0001
1H NMR (400 MHz, DMSO-d6) δ ppm 1 1.53 (s, 1 H) 8.74 (t, J=4.93 Hz, 1 H) 8.25 - 8.30 (m, 2 H) 7.69 (s, 1 H) 7.34 - 7.40 (m, 2 H) 5.89 (s, 1 H) 5.25 (quin, J=6.69 Hz, 1 H) 4.40 (s, 1 H) 4.39 (s, 1 H) 2.45 (s, 3 H) 2.25 (s, 3 H) 2.12 (s, 3 H) 1.51 (s, 3 H) 1.50 (s, 3 H). LC-MS (ES) m/z = 447.8 [M+H]+.
Example 130
1 -isopropyl-N-((6-methyl-2-oxo-4-propyl-1 ,2-dihydropyridin-3-yl)methyl)-6-phenyl- 1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000185_0002
Ethyl 1-(1-methylethyl)-6-phenyl-1 H-pyrazolo[3,4-b]pyridine-4-carboxylate (270 mg, 0.873 mmol) was suspended in THF (1913 μΙ) and Methanol (6378 μΙ). To the mixture was added 3N NaOH (436 μΙ, 1 .309 mmol) and the contents heated on an aluminum block at 55 °C overnight. The solvent was removed in vacuo. The remaining orange residue was dissolved with water (10 ml.) and cooled with stirring in an ice bath. The contents were acidifed by dropwise addition 1 N HCI until precipitation ceased. The white solid was collected via vacuum filtration and the filter cake was dried open to the air, under vacuum overnight. The white solid was added to a 20 mL vial followed by 3-(aminomethyl)-6- methyl-4-propyl-2(1 H)-pyridinone (208 mg, 0.960 mmol), N-[3-(dimethylamino)propyl]-N'- ethylcarbodiimide hydrochloride (251 mg, 1.309 mmol), 3H-[1 ,2,3]triazolo[4,5-b]pyridin-3- ol hydrate (202 mg, 1.309 mmol) and DMSO (4 ml). To the same was added a magnetic stir bar and 4-methylmorpholine (618 mg, 6.1 1 mmol). The reaction was stirred at room temperature overnight. The reaction contents were slowly poured onto an aqueous solution of sat Na2C03 (6 mL) and water (20 mL) and stirred in an ice bath. The resulting suspension was stirred 20 minutes. The solid was collected by vacuum filtration and dried under high vacuum overnight. The title compound was obtained as an off-white solid (388 mg, 99%). 1 H NMR (400 MHz, DMSO-56) δ 9.00 (t, J = 3.92 Hz, 1 H), 8.38 (s, 1 H), 8.23 - 8.29 (m, 2H), 8.16 (s, 1 H), 7.46 - 7.62 (m, 3H), 5.93 (s, 1 H), 5.34 (quin, J = 6.63 Hz, 1 H), 4.45 (d, J = 4.55 Hz, 2H), 2.55 (s, 2H), 2.14 (s, 3H), 1 .56 (d, J = 6.57 Hz, 6H), 1 .45 - 1 .54 (m, 2H), 0.88 (t, J = 7.20 Hz, 3H). LCMS(ES) [M+H]+ 444.3. Example 131
1 -cyclopropyl-A -[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3^yridinyl)methyl]-3-methyl-6-(2- methyl-5-pyrimidinyl)-1H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000186_0001
a) Methyl 6-chloro-3-methyl-1 H-pyrazolo[3,4-b]pyridine-4-carboxylate
Figure imgf000186_0002
Ethyl 1-(1 , 1-dimethylethyl)-3-methyl-6-hydroxy-1 H-pyrazolo[3,4-b]pyridine-4-carboxylate (2 g, 7.21 mmol) and phosphorus oxychloride (10 ml, 107 mmol) were heated at 100 °C for 32 hours. Ice was added and the contents were extracted with EtOAc. The combined organic layers were washed with brine, dried over MgS04, filtered, and concentrated in vacuo. The crude residue was purified via silica gel chromatography (eluent: 0% to 50% gradient EtOAc:Hex). The product was collected as 310 mg. 1H NMR (400 MHz, DMSO- d6) δ ppm 1.38 (t, J=7.20 Hz, 3 H), 2.58 (s, 3 H), 4.44 (q, J=7.16 Hz, 2 H), 7.53 (s, 1 H), 13.82 (br. s., 1 H). LCMS(ES) [M+H]+ 240.1 b) Methyl 6-chloro-1 -cyclopropyl-3-methyl-1 H-pyrazolo[3,4-b]pyridine-4- carboxylate
Figure imgf000187_0001
To a suspension of methyl 6-chloro-3-methyl-1 H-pyrazolo[3,4-b]pyridine-4-carboxylate (510 mg, 2.260 mmol), cyclopropylboronic acid (388 mg, 4.52 mmol), sodium carbonate (479 mg, 4.52 mmol) in 1 ,2-Dichloroethane (DCE) (150 mL) was added a suspension of copper(ll) acetate (41 1 mg, 2.260 mmol), 2,2'-bipyridine (353 mg, 2.260 mmol) in hot 1 ,2- dichloroethane. The contents were heated at 70 °C for 16 hours. Added
cyclopropylboronic acid (388 mg, 4.52 mmol), sodium carbonate (479 mg, 4.52 mmol) followed by copper(ll) acetate (41 1 mg, 2.260 mmol), 2,2'-bipyridine (353 mg, 2.260 mmol) in hot 1 ,2-dichloroethane and heated at 70 °C for 16 hours. The contents were cooled to room temperature followed by addition of 100 mL of saturated NH4CI solution and the phases were separated. The aqueous layer was extracted with DCM twice. The combined organic layers were washed with brine, dried over MgS04, filtered, and concentrated in vacuo. The crude residue was purified via silica gel chromatography (eluent: 0% to 20% gradient EtOAc:Hex). The product was collected as 310mg. 1H NMR (400 MHz, DMSO-de) δ ppm 0.95 - 1.26 (m, 4 H), 2.53 (s, 3 H), 3.75 - 3.91 (m, 1 H), 3.96 (s, 3 H), 7.55 (s, 1 H). LCMS(ES) [M+H]+266.0 c) 6-chloro-1 -cyclopropyl-3-methyl-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid
Figure imgf000188_0001
To a solution of methyl 6-chloro-1-cyclopropyl-3-methyl-1 H-pyrazolo[3,4-b]pyridine-4- carboxylate (370 mg, 1.393 mmol) in ethanol (30 mL) was added 1 N sodium hydroxide (1 .393 mL, 1.393 mmol) and heated at reflux for 2 hours. The solvent was removed in vacuo, the residue dissolved in 20 mL of water, and acidified with acetic acid. The contents were extracted with EtOAc (4x30 mL). The combined organic layers were washed with water, brine, dried over MgS04, filtered and concentrated in vacuo. The product was collected as 297mg. 1H NMR (400 MHz, DMSO-d6) δ ppm 0.77 - 1 .43 (m, 4 H), 2.55 (s, 3 H), 3.63 - 4.15 (m, 1 H), 7.52 (s, 1 H), 14.18 (br. s., 1 H). LCMS(ES)
[M+H]+252.4 d) 6-chloro-1 -cyclopropyl-A -[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3- pyridinyl)methyl]-3-methyl-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000188_0002
6-chloro-1-cyclopropyl-3-methyl-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid (297 mg, 1.180 mmol), 3-(aminomethyl)-4,6-dimethyl-2(1 H)-pyridinone (289 mg, 1.534 mmol), 1 - hydroxy-7-azabenzotriazole (321 mg, 2.360 mmol), EDC (452 mg, 2.360 mmol) and N- methylmorpholine (0.519 mL, 4.72 mmol) were suspended in Dimethyl Sulfoxide (DMSO) (10 mL) and stirred at room temperature for 16 hours. Added 25 mL of water and let stir for 10 minutes. The contents were filtered and dried. The product was collected as 455 mg. 1H NMR (400 MHz, DMSO-de) δ ppm 1.01 - 1.22 (m, 4 H), 2.12 (s, 3 H), 2.22 (s, 3 H), 2.36 (s, 3 H), 3.74 - 3.87 (m, 1 H), 4.33 (d, J=5.05 Hz, 2 H), 5.88 (s, 1 H), 7.19 (s, 1 H), 8.79 (t, J=4.67 Hz, 1 H), 1 1.53 (br. s., 1 H). MS(ES) [M+H]+385.9 d) 1 -cyclopropyl-A -[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-3- methyl-6-(2-methyl-5^yrimidinyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
A mixture of 6-chloro-1 -cyclopropyl-N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]- 3-methyl-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide (85 mg, 0.220 mmol), 2-methyl-5- (4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)pyrimidine (58.2 mg, 0.264 mmol), sodium carbonate (0.330 ml_, 0.661 mmol), 1 ,2-Dimethoxyethane (DME) (3 ml.) and Water (1 ml.) were added to a microwave vial and degassed for 10 minutes. Next added 1 , 1 '- bis(diphenylphosphino)ferrocene-palladium(ll)dichloride dichloromethane complex (14.39 mg, 0.018 mmol). The contents were irradiated at 140 °C for 10 minutes. Water was added and insoluble material was filtered off. This insoluble material was dissolved in acetonitrile and purified by silica gel chromatography (eluent: 0% to 20%
(MeOH:NH40H/9:1 ):DCM). The product was collected as 23mg. 1 H NMR (400 MHz, DMSO-de) δ ppm 1.00 - 1.34 (m, 4 H), 2.12 (s, 3 H), 2.25 (s, 3 H), 2.42 (s, 3 H), 2.72 (s, 3 H), 3.99 (dt, J=7.26, 3.57 Hz, 1 H), 4.39 (d, J=5.05 Hz, 2 H), 5.89 (s, 1 H), 7.86 (s, 1 H), 8.74 (s, 1 H), 9.47 (s, 1 H), 1 1.53 (br. s., 1 H). LCMS(ES) [M+H]+ 444.2
Examples 132-134 were prepared using the general procedures outlined for the above compound using 6-chloro-1 -cyclopropyl-N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3- pyridinyl)methyl]-3-methyl-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide and the appropriate boronic acid reagent.
Example 132
1 -cyclopropyl-A -[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3^yridinyl)methyl]-3-methyl-6-[4- (methyloxy)phenyl]-1 H-pyraz -b]pyridine-4-carboxamide
Figure imgf000189_0001
1H NMR (400 MHz, DMSO-de) δ ppm 0.95 - 1.34 (m, 4 H), 2.12 (s, 3 H), 2.25 (s, 3 H), 2.40 (s, 3 H), 3.85 - 4.01 (m, 4 H), 4.38 (d, J=5.05 Hz, 2 H), 5.89 (s, 1 H), 7.00 (d, J=8.59 Hz, 1 H), 7.72 (s, 1 H), 8.53 (dd, J=8.84, 2.53 Hz, 1 H), 8.72 (t, J=4.93 Hz, 1 H), 9.03 (d, J=2.27 Hz, 1 H), 1 1 .53 (s, 1 H). LCMS(ES) [M+H]+ 459.2
Example 133
1 -cyclopropyl-A -[(41 -cyclopropyl-A -[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3- pyridinyl)methyl]-3 -methyl -6-[6-(4-methyl-1 -piperazinyl)-3-pyridinyl]-1H- pyrazolo[3,4-b]pyridine-4-carboxamide.
Figure imgf000190_0001
1H NMR (400 MHz, DMSO-d6) δ ppm 0.99 - 1.24 (m, 4 H), 2.12 (s, 3 H), 2.21 - 2.29 (m, 6 H), 2.38 (s, 3 H), 2.45 (br. s., 4 H), 3.63 (br. s., 4 H), 3.93 (m, J=7.26, 7.26, 3.79, 3.66 Hz, 1 H), 4.38 (d, J=5.05 Hz, 2 H), 5.88 (s, 1 H), 6.98 (d, J=9.09 Hz, 1 H), 7.64 (s, 1 H), 8.35 (dd, J=9.09, 2.53 Hz, 1 H), 8.70 (t, J=5.05 Hz, 1 H), 8.98 (d, J=2.27 Hz, 1 H), 1 1.52 (s, 1 H). LCMS(ES) [M+H]+ 527.1
Example 134
1 -cyclopropyl-A -[(41 -cyclopropyl-A -[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3- pyridinyl)methyl]-3 -methyl -6-[6-(4-morpholinyl)-3-pyridinyl]-1 H-pyrazolo[3, 4- b]pyridine-4-carboxamide.
Figure imgf000191_0001
1H NMR (400 MHz, DMSO-de) δ ppm 1 .03 - 1.32 (m, 4 H), 2.12 (s, 3 H), 2.24 (s, 3 H), 2.38 (s, 3 H), 3.49 - 3.64 (m, 4 H), 3.68 - 3.79 (m, 4 H), 3.93 (m, J=7.29, 7.29, 3.92, 3.73 Hz, 1 H), 4.38 (d, J=5.05 Hz, 2 H), 5.88 (s, 1 H), 6.98 (d, J=8.84 Hz, 1 H), 7.65 (s, 1 H), 8.38 (dd, J=8.97, 2.40 Hz, 1 H), 8.70 (t, J=4.93 Hz, 1 H), 9.00 (d, J=2.53 Hz, 1 H), 1 1.52 (s, 1 H). LCMS(ES) [M+H]+ 514.2
Example 135
6-cyclopropyl-1 -(1 ,1 -dimethylethyl)-3-methyl-A -[(6-methyl-2-oxo-4-propyl-1 ,2- dihydro-3-pyridinyl)methyl]-1 H-pyrazolo[3,4-j ]pyridine-4-carboxamide
Figure imgf000191_0002
a) Ethyl 6-cyclopropyl-1 -(1 ,1 -dimethylethyl)-3-methyl-1 H-pyrazolo[3,4- b] py ri d i ne-4-carboxy I ate
Figure imgf000192_0001
1-(1 , 1 -dimethylethyl)-3-methyl-1 H-pyrazol-5-amine (166 mg, 1 .086 mmol), ethyl (3Z)-4- cyclopropyl-4-hydroxy-2-oxo-3-butenoate (200 mg, 1.086 mmol) and acetic acid (10 mL) were heated to reflux for 2 hours. The solvent was removed in vacuo and the residue purified by silica gel chromatography (eluent: 0% to 7 % EtOAc:Hex). The product was collected as 278 mg. 1H NMR (400 MHz, CHLOROFORM-d) δ ppm 1.07 (dt, 2 H), 1.13 - 1.20 (m, 2 H), 1.46 (t, J=7.20 Hz, 3 H), 1 .78 (s, 9 H), 2.18 (m, J=7.99, 7.99, 4.74, 4.55 Hz, 1 H), 2.65 (s, 3 H), 4.47 (q, J=7.07 Hz, 2 H), 7.43 (s, 1 H). LCMS(ES) [M+H]+ 302.2 a) 6-cyclopropyl-1 -(1 ,1 -dimethylethyl)-3-methyl-1 H-pyrazolo[3,4-b]pyridine-4- carboxylic acid
Figure imgf000192_0002
To a solution of ethyl 6-cyclopropyl-1-(1 , 1 -dimethylethyl)-3-methyl-1 H-pyrazolo[3,4- b]pyridine-4-carboxylate (278 mg, 0.922 mmol) in ethanol (30 mL) was added sodium hydroxide (3.69 mL, 3.69 mmol) and the contents heated at reflux for 2 hours. The solvent was removed in vacuo, the residue dissolved in 20 mL of water, and acidifed by addition of acetic acid. The contents were extracted with EtOAc (4x30ml). The combined organic extracts were washed with water, brine, dried over MgS04, filtered, and concentrated in vacuo. The product was collected as 233 mg. 1 H NMR (400 MHz, DMSO-de) δ ppm 0.97 - 1.12 (m, 4 H), 1 .69 (s, 9 H), 2.22 - 2.41 (m, 3 H), 2.52 (s, 3 H), 7.45 (s, 3 H). LCMS(ES) [M+H]+ 274.2 b) 6-cyclopropyl-1 -(1 ,1 -dimethylethyl)-3-methyl-A -[(6-methyl-2-oxo-4-propyl-1 ,2- dihydro-3^yridinyl)methyl]-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
6-cyclopropyl-1 -(1 , 1 -dimethylethyl)-3-methyl-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid (53 mg, 0.194 mmol), 3-(aminomethyl)-6-methyl-4-propyl-2(1 H)-pyridinone (71.3 mg, 0.242 mmol), 1-hydroxy-7-azabenzotriazole (52.8 mg, 0.388 mmol), EDC (74.3 mg, 0.388 mmol) and N-methylmorpholine (0.085 mL, 0.776 mmol) were suspended in Dimethyl Sulfoxide (DMSO) (10 mL) and stirred at room temperature for 16 hours. Added 25 mL of water and stirred for 10 minutes. The contents were filtered and dried, and collected as 70 mg. 1 H NMR (400 MHz, DMSO-de) δ ppm 0.93 (t, J=7.33 Hz, 3 H), 0.96 - 1.09 (m, 4 H), 1.54 (m, J=7.52, 7.52, 7.52, 7.52, 7.33 Hz, 2 H), 1 .68 (s, 9 H), 2.12 (s, 2 H), 2.17 - 2.27 (m, 1 H), 2.33 (s, 3 H), 2.5(2H), 4.34 (d, J=5.05 Hz, 2 H), 5.89 (s, 1 H), 7.01 (s, 1 H), 8.54 (t, J=4.80 Hz, 1 H), 1 1.50 (s, 1 H). LCMS(ES) [M+H]+ 436.1 .
Example 136
1 ,6-dicyclopropyl-3-methyl-A -[(6-methyl-2-oxo-4-propyl-1 ,2-dihydro-3- pyridinyl)methyl]-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000193_0001
a) 1 ,1 -dimethylethyl cyclopropyl(nitroso)carbamate
Figure imgf000193_0002
Nitrosonium tetrafluoroborate (2.415 g, 20.67 mmol) was added in several portions to a cooled (-30 °C) solution of 1 , 1 -dimethylethyl cyclopropylcarbamate (2.5 g, 15.90 mmol) in anhydrous Pyridine (4 mL) and Acetonitrile (40 mL). The solution was stirred at -30 °C for 30 minutes then at 0 °C for 2 hours. Ice water and EtOAc were added. The organic phase was separated and washed with 1 N HCI, 1 N NaHC03, brine, dried over MgS04, filtered and concentrated in vacuo. The product was collected as 2.66 g. 1 H NMR (400 MHz, CHLOROFORM-d) δ ppm 0.37 - 0.79 (m, 4 H), 1 .64 (s, 9 H), 2.18 - 2.41 (m, 1 H). b) Cyclopropylhydrazine
Figure imgf000194_0001
1 , 1 -dimethylethyl cyclopropyl(nitroso)carbamate (2.63 g, 14.12 mmol) was dissolved in methanol (130 mL) and cooled to -78 °C. Hydrochloric acid (12.48 mL, 150 mmol) was slowly added dropwise. Zinc was added in portions, and the contents were stirred at -78 °C for 6 hours. The contents were filtered through celite. The solvent was removed in vacuo. The residue was suspended in EtOH and concentrated in vacuo in triplicate (to help azeotrope off water). The product was obtained as a sticky semi-solid which still had EtOH in sample (5 g). 1H NMR (400 MHz, DMSO-de) δ ppm 0.34 - 0.79 (m, 5 H), 7.91 (d, 1 H), 8.87 (br. s., 2 H)
c) 1 -cyclopropyl-3-methyl-1H-pyrazol-5-amine
Figure imgf000194_0002
(2Z)-3-amino-2-butenenitrile (903 mg, 1 1 .00 mmol), cyclopropylhydrazine (1 194 mg, 1 1 mmol) and triethylamine (3.07 mL, 22.00 mmol) were suspended in ethanol (100 mL) and heated to 70 °C for 16 hours. The solvent was removed in vacuo. The residue was suspended in 50 mL of saturated NaHC03 and stirred for 10 minutes. The contents were extracted with DCM, dried over MgS04, filtered, and concentrated in vacuo. The residue was purified by silica gel chromatography (eluent: 0% to 100% gradient EtOAc:Hex). The product was collected as 310mg. 1H NMR (400 MHz, CHLOROFORM-d) δ ppm 0.95 - 1.13 (m, 4 H), 2.13 (s, 3 H), 3.06 (m, J=6.95, 6.95, 3.79, 3.54 Hz, 1 H), 3.76 - 3.88 (m, 2 H), 5.24 (s, 1 H). LCMS(ES) [M+H]+ 138.1 d) Ethyl 1 ,6-dicyclopropyl-3-methyl-1H-pyrazolo[3,4-b]pyridine-4-carboxylate
Figure imgf000195_0001
1-cyclopropyl-3-methyl-1 H-pyrazol-5-amine (300 mg, 2.187 mmol), ethyl 4-cyclopropyl- 2,4-dioxobutanoate (403 mg, 2.187 mmol) and acetic Acid (40 mL) were heated at reflux for 2 hours. The solvent was removed in vacuo and the crude residue was purified via silica gel chromatography (eluent: 0 to 1 1 % EtOAc:Hex). The product was collected as 310mg. 1 H N MR (400 MHz, CHLOROFORM-d) δ ppm 1.05 - 1 .13 (m, 4 H), 1 .16 - 1 .21 (m, 2 H), 1.25 - 1 .30 (m, 2 H), 1 .46 (t, J=7.20 Hz, 3 H), 2.16 - 2.31 (m, 1 H), 2.63 (s, 3 H), 3.78 (m, J=7.26, 7.26, 3.79, 3.66 Hz, 1 H), 4.47 (q, J=7.24 Hz, 2 H), 7.40 (s, 1 H).
LCMS(ES) [M+H]+ 286.2 e) 1 ,6-dicyclopropyl-3-methyl-1H-pyrazolo[3,4-b]pyridine-4-carboxylic acid
Figure imgf000195_0002
To a solution of ethyl 1 ,6-dicyclopropyl-3-methyl-1 H-pyrazolo[3,4-b]pyridine-4-carboxylate (310 mg, 1.086 mmol) in ethanol (30 mL) was added sodium hydroxide (4.35 mL, 4.35 mmol) and the mixture heated at reflux for 2 hours. The solvent was removed in vacuo, the residue suspended in water (20 mL), and acidified by addition of acetic acid. The contents wer extracted with EtOAc (4x30 mL). The combined EtOAc extracts were washed with water, brine, dried over MgS04, filtered, and concentrated in vacuo. The product was collected as 250 mg. 1H NMR (400 MHz, DMSO-d6) δ ppm 0.88 - 1 .31 (m, 8 H), 2.21 - 2.42 (m, 1 H), 3.83 (m, J=7.39, 7.39, 3.79, 3.66 Hz, 1 H), 7.44 (s, 1 H), 13.72 (br. s., 1 H). LCMS(ES) [M+H]+ 258.2 f) 1 ,6-dicyclopropyl-3-methyl-A -[(6-methyl-2-oxo-4-propyl-1 ,2-dihydro-3- pyridinyl)methyl]-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide 1 ,6-dicyclopropyl-3-methyl-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid (125 mg, 0.486 mmol), 3-(aminomethyl)-6-methyl-4-propyl-2(1 H)-pyridinone (126 mg, 0.583 mmol), 1- hydroxy-7-azabenzotriazole (132 mg, 0.972 mmol), EDC (186 mg, 0.972 mmol) and N- methylmorpholine (0.214 mL, 1 .943 mmol) were suspended in Dimethyl Sulfoxide (DMSO) (10 mL) and stirred at room temperature for 16 hours. Added 25 mL of water and let stir for 10 minutes. The contents were filtered and dried. The product was collected as 200 mg. 1H NMR (400 MHz, DMSO-de) δ ppm 0.92 (t, J=7.07 Hz, 3 H), 1 .04 (d, J=6.06 Hz, 8 H), 1.45 - 1 .73 (m, 2 H), 2.12 (s, 3 H), 2.24 (br. s., 1 H), 2.33 (s, 3 H), 3.79 (br. s., 1 H), 4.34 (d, J=3.79 Hz, 2 H), 5.89 (br. s., 1 H), 6.99 (s, 1 H), 8.57 (br. s., 1 H), 1 1 .51 (br. s., 1 H). MS(ES) [M+H]+ 420.2
Example 137
1 ,6-dicyclopropyl-A -[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3^yridinyl)methyl]-3-methyl- 1 H-pyrazolo[3,4-b]pyridine-4-carboxamide.
Figure imgf000196_0001
The title compound was made using the same procedure as for example 1 1 (step f) from
I , 6-dicyclopropyl-3-methyl-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid and 3- (aminomethyl)-4,6-dimethyl-2(1 H)-pyridinone. 1 H NMR (400 MHz, DMSO-de) δ ppm 0.93 - 1.18 (m, 8 H), 2.1 1 (s, 3 H), 2.21 (s, 4 H), 2.32 (s, 3 H), 3.78 (m, J=7.33, 7.33, 3.92, 3.66 Hz, 1 H), 4.33 (d, J=5.05 Hz, 2 H), 5.87 (s, 1 H), 7.01 (s, 1 H), 8.58 (t, J=4.80 Hz, 1 H),
I I .50 (s, 1 H). LCMS(ES) [M+H]+ 392. Example 138
6-(cyclopropylamino)-N-((4,6-dimethyl-2-oxo-1 ,2-dihydropyridin-3-yl)methyl)-1 - isopropyl-3-methyl-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000197_0001
6-chloro-N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-3-methyl-1 -(1 - methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxamide (100mg, 0.258 mmol) was dissolved in 1 ,4-dioxane (2.6 ml.) followed by addition of cesium carbonate (336 mg, 1.031 mmol). After degassing with nitrogen for 5 min, Xantphos (44.8 mg, 0.077 mmol) was added. The mixture was degassed for 2 min, Pd2(dba)3 (35.4 mg, 0.039 mmol) was added, and then degassed for 1 min. Cyclopropanamine (0.090 ml_, 1 .289 mmol) was added and the contents sealed. The suspension was stirred for 3h at 100 °C (heat block). The reaction mixture was poured onto water and extracted with EtOAc (2x). The combined organic layers were dried over MgS04, filtered, and concentrated in vacuo to a solid residue. The aq. layer was extracted with 10%MeOH/DCM (2x). The combined organic layers were dried over MgS04, filtered, and concentrated in vacuo to afford a solid. The combined solid residues were dissolved in DMSO and acetonitrile spiked with TFA, and purified by reverse phase HPLC (Gradient B : 10-70%. A: Water + .1 % TFA. B: CH3CN + .1 % TFA) The title compound was collected as 1 1 mg (10%) ; 1H NMR (400 MHz, DMSO-d6) δ ppm 0.39 - 0.47 (m, 2 H), 0.68 - 0.76 (m, 2 H), 1 .39 (d, J=6.57 Hz, 6 H), 2.1 1 (s, 3 H), 2.24 (s, 3 H), 2.20 (s, 3 H), 2.64 - 2.72 (m, 1 H), 4.30 (d, J=4.80 Hz, 2 H), 4.89 (quin, J=6.69 Hz, 1 H), 5.86 (s, 1 H), 6.34 (s, 1 H), 7.25 (d, J=3.03 Hz, 1 H), 8.41 (t, J=4.93 Hz, 1 H), 1 1.49 (s, 1 H). LCMS(ES) [M+H]+ 409.1 Example 139
N-((4-benzyl-6-methyl-2-oxo-1 ,2-dihydropyridin-3-yl)methyl)-1 -isopropyl-1 H- pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000198_0001
a) 1 -isopropyl-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid
Figure imgf000198_0002
To a 100 mL round bottom flask was charged 10% Pd/C(degussa) (0.213 g, 0.100 mmol), under N2 followed by addition of ca. 5 mL EtOH. The slurry was stirred followed by addition of 6-chloro-1-(1 -methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid (.48g, 2.003 mmol) and ethanol (22 mL). Next added Et3N (0.837 mL, 6.01 mmol) and stirred for 5 min under nitrogen. The contents were then stirred under an atmosphere of hydrogen (1 atm) for 6h at RT. The vessel was then flushed with nitrogen and the contents diluted with DCM (10 mL) and a small amount of celite. The contents were stirred for 10 min., filtered through analytical grade celite and washed with 10% MeOH/DCM, EtOH, then DCM. The filtrate was concentrated in vacuo to a residue and dried on hi-vacuum overnight. The solid was treated with water and adjusted to pH 3 with 1 M HCI. The contents were filtered, air-dried and then dried in hi-vacuum oven at 45 °C for 18h. The title compound was collected as 0.326g (78%). 1H NMR (400 MHz, DMSO-c/6) 5ppm 1.52 (d, J=6.82 Hz, 6 H) 5.28 (quin, J=6.69 Hz, 1 H) 7.70 (d, J=4.55 Hz, 1 H) 8.39 (s, 1 H) 8.73 (d, J=4.55 Hz, 1 H) 13.91 (br. s., 1 H). LCMS(ES) [M+H]+ 205.9 b) N-((4-benzyl-6-methyl-2-oxo-1 ,2-dihydropyridin-3-yl)methyl)-1 -isopropyl-1 H- pyrazolo[3,4-b]pyridine-4-carboxamide
The title compound was prepared in the same manner as described for example 1 1 from 1-(1-methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid (.060 g, 0.292 mmol) and 3(aminomethyl)-6-methyl-4-(phenylmethyl)-2(1 H)-pyridinone (0.081 g, 0.307 mmol) wherein the product obtained was further purified by reverse phase HPLC (Gradient B: 5- 85%. A:Dichloromethane. B: 10% (2M Ammonia in Methanol) in Chloroform). The isolated product was concentrated from MTBE (2x) to afford a white solid (93 mg, 75%). 1H NMR (400 MHz, DMSO-d6) δ ppm 1 .50 (d, J=6.57 Hz, 6 H), 2.10 (s, 3 H), 3.98 (s, 2 H), 4.45 (d, J=5.05 Hz, 2 H), 5.24 (quin, J=6.63 Hz, 1 H), 5.81 (s, 1 H), 7.12 - 7.30 (m, 5 H), 7.50 (d, J=4.55 Hz, 1 H), 8.33 (s, 1 H), 8.61 (d, J=4.55 Hz, 1 H), 8.88 (t, J=5.05 Hz, 1 H), 1 1 .63 (s, 1 H). LCMS(ES) [M+H]+ 416.0 Example 140
N-((4-benzyl-6-methyl-2-oxo-1 ,2-dihydropyridin-3-yl)methyl)-1 -isopropyl-3-methyl- 1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000199_0001
a) 1 -isopropyl-3-methyl-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid
Figure imgf000199_0002
The title compound was prepared in the same manner as described for example 139 (step a) from 10% Pd/C(degussa) (0.457 g, 0.215 mmol), and 6-chloro-3-methyl-1-(1- methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid (1 .09g, 4.30 mmol) wherein the stir time was 4h Upon acidification to pH 3 with 1 M HCI, the contents were extracted with EtOAc (2x) and the combined organic layers dried over MgS04, filtered, and concentrated in vacuo. The collected solid was dried in a vacuum oven at 45 °C for 3 h. The final product was collected as 0.826g (84%). 1H NMR (400 MHz, DMSO-d6) d ppm 1.48 (d, J=6.57 Hz, 6 H), 2.63 (s, 3 H), 5.21 (quin, J=6.69 Hz, 1 H), 7.52 (d, J=4.55 Hz, 1 H), 8.63 (d, J=4.55Hz, 1 H), 13.77 (br. s., 1 H). LCMS(ES) [M+H]+ 220.2
N-((4-benzyl-6-methyl-2-oxo-1 ,2-dihydropyridin-3-yl)methyl)-1 -isopropyl-3-methyl- 1 H-pyrazolo[3,4-b]pyridine-4-carboxamide The title compound was prepared in the same manner as described for example 1 1 from 3-methyl-1-(1-methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid (.060g, 0.274 mmol) and 3-(aminomethyl)-6-methyl-4-(phenylmethyl)-2(1 H)-pyridinone (0.076 g, 0.287 mmol). The product was collected as a white solid (92 mg, 77%). 1 H NMR (400 MHz, DMSO-d6) δ ppm 1.46 (d, J=6.82 Hz, 6 H), 2.09 (s, 3 H), 2.41 (s, 3 H), 3.98 (s, 2 H), 4.43 (d, J=5.05 Hz, 2 H), 5.16 (quin, J=6.63 Hz, 1 H), 5.79 (s, 1 H), 7.04 (d, J=4.80 Hz, 1 H), 7.16 - 7.34 (m, 5 H), 8.49 (d, J=4.55 Hz, 1 H), 8.75 (t, J=5.05 Hz, 1 H), 1 1 .59 (s, 1 H). LCMS(ES) [M+H]+ 429.9
Example 141
6-cyclopropyl-A -[(4-cyclopropyl-6-methyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-3- methyl-1 -(1 -methylethyl)-1H- razolo[3,4-b]pyridine-4-carboxamide
Figure imgf000200_0001
6-cyclopropyl-A -[(4-cyclopropyl-6-methyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-3- methyl-1 -(1 -methylethyl)-1H-pyrazolo[3,4-b]pyridine-4-carboxamide
6-cyclopropyl-1 -isopropyl-3-methyl-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid (150 mg, 0.578 mmol), 1 -hydroxy-7-azabenzotriazole (1 18 mg, 0.868 mmol) and EDC (166 mg, 0.868 mmol) were suspended in Dimethyl Sulfoxide (DMSO) (10 mL). To the solution was added in one portion N-methylmorpholine (0.191 mL, 1.735 mmol), followed by 3- (aminomethyl)-4-cyclopropyl-6-methylpyridin-2(1 H)-one (bis HCI salt) (189 mg, 0.752 mmol). and the reaction stirred at RT for 12 h. The reaction contents were poured onto ice water (200ml_) and were stirred for 20 min. The contents were filtered and washed with water (10 mL) and then methanol/ice water (10ml_/10 mL). The product was dried to afford a tan solid which was collected as 180 mg (71 %). 1H NMR (400 MHz, DMSO-c/6) d ppm 1 1 .45 (s, 1 H) 8.64 (t, J=4.93 Hz, 1 H) 6.99 (s, 1 H) 5.49 (s, 1 H) 5.04 (quin, J=6.69 Hz, 1 H) 4.52 (d, J=5.05 Hz, 2 H) 2.37 (s, 3 H) 2.21 - 2.26 (m, 1 H) 2.12 - 2.17 (m, 1 H) 2.09 (s, 3 H) 1.42 (d, J=6.57 Hz, 6 H) 1 .01 - 1 .04 (m, 4 H) 0.95 - 1.00 (m, 2 H), 0.075-
0.076 (m, 2H); LCMS: [M+H]+ = 420.3.
Examples 142-144 were prepared in the same manner as described for example 16 step c using 6-cyclopropyl-1 -isopropyl-3-methyl-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid and the appropriately substituted 3-aminomethyl-pyridone intermediate.
Example 142
6-cyclopropyl-3-methyl-1 -(1 -methylethyl)-W-{[6-methyl-4-(1 -methylethyl)-2- dihydro-3-pyridinyl]methyl}-1H- razolo[3,4-b]pyridine-4-carboxamide
Figure imgf000201_0001
1H NMR (400 MHz, DMSO-d6) δ ppm 1 1.51 (br. s., 1 H) 8.60 (t, J=4.93 Hz, 1 H) 6.97 (s, 1 H) 6.01 (s, 1 H) 5.04 (dt, J=13.39, 6.69 Hz, 1 H) 4.40 (d, J=4.80 Hz, 2 H) 3.18 - 3.27 (m, 1 H) 2.38 (s, 3 H) 2.19 - 2.26 (m, 1 H) 2.15 (s, 3 H) 1 .42 (d, J=6.57 Hz, 6 H) 1.13 (d, J=6.82 Hz, 6 H) 1.01 - 1.05 (m, 4 H); LCMS: [M+H]+= 422.3 Example 143
6-cyclopropyl-3-methyl-1 -(1 -methylethyl)-A -[(6-methyl-2-oxo-4-propyl-1 ,2-dihydro-3- pyridinyl)methyl]-1 H-pyrazolo 3,4-b]pyridine-4-carboxamide
Figure imgf000202_0001
1H NMR (400 MHz, DMSO-d6) δ ppm 1 1.52 (br. s., 1 H) 8.57 (t, J=4.80 Hz, 1 H) 6.97 (s, 1 H) 5.90 (s, 1 H) 5.00-5.07 (m, 1 H) 4.35 (d, J=5.05 Hz, 2 H) 2.37 (s, 3 H) 2.20 - 2.26 (m, 1 H) 2.13 (s, 3 H) 1.51 - 1.59 (m, 2 H) 1 .42 (d, J=6.57 Hz, 6 H) 0.99 - 1.06 (m, 5 H) 0.93 (t, J=7.33 Hz, 4 H); LCMS: [M+H]+ = 422.3
Example 144
6-cyclopropyl-3-methyl-1 -(1 -methylethyl)-A -{[6-methyl-2-oxo-4-(phenylmethyl)-1 ,2- dihydro-3-pyridinyl]methyl}-1H- razolo[3,4-b]pyridine-4-carboxamide
Figure imgf000202_0002
1H NMR (400 MHz, DMSO-d6) δ ppm 1 1.60 (br. s., 1 H) 8.66 - 8.75 (m, 1 H) 7.20 - 7.35 (m, 5 H) 6.92 (s, 1 H) 5.79 (s, 1 H) 4.90-5.04 (m, 1 H) 4.42 (d, J=4.80 Hz, 2 H) 3.97 (s, 2 H) 2.35 (s, 3 H) 2.01-2.21 (m, 1 H) 2.09 (s, 3 H) 1.42 (d, J=6.82 Hz, 6 H) 1 .00 - 1 .07 (m, 4 H); LCMS:[M+H]+= 470.3 Intermediates 95 and 96
3- (aminomethyl)-6-methyl-4-(phenylmethyl)-2(1 H)-pyridinone and 3-(aminomethyl)-
4- methyl-6-(phenylmethyl)-2(1 H)-pyridinone
Figure imgf000203_0001
a) 1 -phenyl-2,4-pentanedione
Figure imgf000203_0002
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 1 N HCI. 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 %). 1H NMR (400 MHz, CDCI3-d3) δ 15.49 (br s, 1 H), 7.33-7.45 (m, 5H), 5.53 (s, 1 H), 3.66 (s, 2H), 2.10 (s, 3H). b) 6-methyl-2-oxo-4-(phenylmethyl)-1 ,2-dihydro-3-pyridinecarbonitrile and 4-methyl- 2-oxo-6-(phenylmethyl)-1 ,2-dihydro-3-pyridinecarbonitrile
1-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)-1 ,2-dihydro-3-pyridinecarbonitrile and 4-methyl-2-oxo-6- (phenylmethyl)-1 ,2-dihydro-3-pyridinecarbonitrile (12.06 g, 52%). LCMS MH+ = 225.1 1H NMR (400 MHz, DMSO-d6) (mixture of compounds) 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). a) 3-(aminomethyl)-6-methyl-4-(phenylmethyl)-2(1 H)-pyridinone and 3- (aminomethyl)-4-methyl-6-(phenylmethyl)-2(1 H)-pyridinone
Figure imgf000204_0001
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)-1 ,2-dihydro-3-pyridinecarbonitrile and 4-methyl-2-oxo-6-(phenylmethyl)- 1 ,2-dihydro-3-pyridinecarbonitrile (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(1 H)-pyridinone and 3- (aminomethyl)-4-methyl-6-(phenylmethyl)-2(1 H)-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)-1 ,2-dihydro-3-pyridinyl]methyl}carbamate (1 H NMR (400 MHz, DMSO-d6) δ 1 1.55-1 .60 (br s, 1 H), 7.20-7.29 (m, 5H), 5.85 (s, 1 H), 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)-1 ,2-dihydro-3-pyridinyl]methyl}carbamate (1H NMR (400 MHz, DMSO-d6) 51 1.50-1 1.55 (br s, 1 H), 7.18-7.25 (m, 5H), 5.75 (s, 1 H), 4.02 (s, 2H), 3.85 (s, 2H), 2.05 (s, 3H), 1.32 (s, 9H). d) 3-(aminomethyl)-4-methyl-6-(phenylmethyl)-2(1 H)-pyridinone hydrochloride
Figure imgf000205_0001
A solution of 1 , 1-dimethylethyl {[4-methyl-2-oxo-6-(phenylmethyl)-1 ,2-dihydro-3- pyridinyl]methyl}carbamate (1.2 g, 3.66 mmol) in 4N HCI (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(1 H)-pyridinone as an HCI salt (0.725 g, 87%). LCMS MH+ = 229.1 1 H NMR (400 MHz, DMSO-d6) δ 1 1.9- 12.0 (br s, 1 H), 7.99 (br s, 3H), 7.20 (s, 5H), 5.97 (s, 1 H), 3.72-3.75 (m, 4H), 2.17 (s, 3H). e) 3-(aminomethyl)-6-methyl-4-(phenylmethyl)-2(1 H)-pyridinone hydrochloride
Figure imgf000205_0002
A solution of 1 , 1-dimethylethyl {[6-methyl-2-oxo-4-(phenylmethyl)-1 ,2-dihydro-3- pyridinyl]methyl}carbamate (1.0 g, 3.0 mmol) in 4N HCI (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(1 H)-pyridinone as an HCI salt (0.600 g, 86%). LCMS MH+ = 229.1 1 H NMR (400 MHz, DMSO-d6) δ 1 1.9- 12.0 (br s, 1 H), 8.03 (br s, 3H), 7.16-7.30 (m, 5H), 5.84 (s, 1 H), 3.91 (s, 2H), 3.81 (s, 2H), 2.10 (s, 3H).
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 1 1 -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-HCI, pH 8: Per 1 L of base buffer, combine 1 M Tris-HCI, pH 8 (50 ml.) and distilled water (950 ml_).
2. 1x Assay Buffer: Per 10 ml. of 1 x Assay Buffer, combine 50 mM Tris-HCI, pH 8 (9958 uL), 1 M MgCI2 (20 uL), 2 M DTT (20 uL), and 10% Tween-20 (2 uL) to provide a final concentration of 50 mM Tris-HCI, pH 8, 2 mM MgCI2, 4 mM DTT, 0.002% Tween-20.
3. 2x Enzyme Solution: Per 10 ml. of 2x Enzyme Solution, combine 1x 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 LEADSeeker 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 1x 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 ddH20 (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 100x 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 IC50 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 IC50 values ranged from about 1 nM to about 10 μΜ; The IC50 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 , 475; Ex 2, 806; Ex 4, 1 16; Ex 5, 705; Ex 6, 695; Ex 7, 1296; Ex 8, 167; Ex 9, 1309; Ex 10, 569; Ex 1 1 , 18; Ex 12, 55; Ex 13, 55; Ex 14, 735; Ex 15, 179; Ex 16, 105; Ex 17, 2591 ; Ex 18, 40; Ex 19, 3372; Ex 20, 4647; Ex 21 , 1040; Ex 22, 1362; Ex 23, 1428; Ex 24, 873; Ex 25, 685; Ex 26, 673; Ex 27, 24; Ex 28, 348; Ex 29, 234; Ex 30, 154; Ex 31 , 232; Ex 32, 856; Ex 33, 70; Ex 35, 673; Ex 36, 924; Ex 37, 1095; Ex 38, 392; Ex 41 , 86; Ex 42, 56; Ex 43, 204; Ex 44, 74; Ex 45, 248; Ex 46, 128; Ex 47, 88; Ex 48, 198; Ex 49, 1 15; Ex 50, 81 ; Ex 51 , 161 ; Ex 53, 436; Ex 54, 514; Ex 55, 260; Ex 56, 21 1 1 ; Ex 57, 784; Ex 58, 78; Ex 59, 155; Ex 60, 198; Ex 61 , 1 12; Ex 62, 581 ; Ex 63, 96; Ex 64, 79; Ex 65, 55; Ex 66, 81 ; Ex 67, 58; Ex 68, 76; Ex 69, 25; Ex 70, 1893; Ex 71 , 402; Ex 72, 171 ; Ex 73, 533; Ex 74, 151 ; Ex 75, 131 ; Ex 76, 82; Ex 77, 52; Ex 78, 43; Ex 79, 140; Ex 80, 71 ; Ex 81 , 30; Ex 82, 108; Ex 83, 43; Ex 84, 99; Ex 85, 31 ; Ex 86, 142; Ex 87, 18; Ex 88, 52; Ex 89, 67; Ex 90, 173; Ex 92, 76; Ex 93, 83; Ex 94, 103; Ex 95, 489; Ex 96, 57; Ex 97, 55; Ex 99, 25044; Ex 100, 5747; Ex 103, 373; Ex 105, 315; Ex 106, 1 19; Ex 107, 75; Ex 109, 207; Ex 1 10, 231 ; Ex 1 1 1 , 367; Ex 1 12, 693; Ex 1 13, 248; Ex 1 14, 199; Ex 1 17, 190; Ex 1 18, 273; Ex 1 19, 333; Ex 120, 270; Ex 121 , 407; Ex 122, 153; Ex 123, 218; Ex 124, 1052; Ex 125, 2164. Preparation of specific compounds (Compound A, B and C) that were tested in the T cell proliferation and cytokine production studies:
N-((4,6-dimethyl-2-oxo-1 ,2-dihydropyridin-3-yl)methyl)-6-(4-(2- (dimethylamino)ethoxy)phenyl)-1 -isopropyl-1 H-indazole-4-carboxamide
Figure imgf000208_0001
6-bromo-N-(4,6-dimethyl-2-oxo-1 ,2-dihydropyridin-3-yl)methyl)-1-isopropyl-1 H
carboxamide (80 mg, 0.19 mmol), N,N-dimethyl-2-(4-(4,4,5,5-tetramethyl-1 ,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:1 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/NH4OH; 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%). 1H NMR (400 MHz, DMSO-c/6) ppm 1 1.54 (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]+
Compound A can be prepared according to a procedure analogous to the above example: Compound A: 1 -(1 -methylethyl)-N-[(6-methyl-2-oxo-4-propyl-1 ,2-dihydro-3- pyridinyl)methyl]-6-[6-(4-methyl-1 -piperazinyl)-3-pyridinyl]-1 H-indazole-4- carboxamide
Figure imgf000209_0001
The title compound was prepared in a similar manner as described for the above example 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-1 ,3,2-dioxaborolan-2-yl)-2-pyridinyl]piperazine (92 mg, 0.303 mmol). The final product was collected as a light brown solid (54 mg, 49%). 1H NMR (400 MHz, DMSO-de) ppm 1 H NMR (400 MHz, DMSO-d6) ppm 1 1 .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, 1 H) 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]+
Compound B: N-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-3-methyl-1 -(1 - methylethyl)-6-[6-(4-methyl-1 ^iperazinyl)-3-pyridinyl]-1 H-indole-4-carboxamide
Figure imgf000210_0001
Compound B can be prepared according to the general procedure described in the above experimental section.
6-Cyclopropyl-1 -(1 -methylethyl)-A -[(4-methyl-2-oxo-6-propyl-1 ,2-dihydro-3- pyridinyl)methy yridine-4-carboxamide
Figure imgf000210_0002
6-Cyclopropyl-1 -(1 -methylethyl)-1 H-pyrazolo[3,4-b]pyridine-4-carboxylic acid (167 mg, 0.680 mmol), 3-(aminomethyl)-4-methyl-6-propyl-2(1 /-/)-pyridinone trifluoroacetate (200 mg, 0.680 mmol), HOAT (139 mg, 1 .019 mmol), EDC (195 mg, 1 .019 mmol), and N- methylmorpholine (0.299 mL, 2.72 mmol) were dissolved in DMF(6 mL) and stirred at 40 °C for 24 h. The reaction mixture was poured into water (20 mL) and extracted with ethyl acetate (3 x 50 mL). The combined organic layers were dried over sodium sulfate and concentrated to an orange oil. The residue was dissolved in DMSO, and purified by reverse phase HPLC (mobile phase: 40-60% ACN in H20, 0.1 % TFA). The isolated product was dried in a vacuum oven overnight and furnished the TFA salt of the title compound as a white solid, 0.1 13 g (32%). LCMS E-S (M+H) = 408.1. 1H NMR (400 MHz, DMSO-d6) δ ppm 0.80 - 0.98 (m, 3 H), 1.06 (d, J = 7.07 Hz, 4 H), 1.46 (d, J = 6.82 Hz, 6 H), 1.52 - 1 .67 (m, 2 H), 2.17 - 2.31 (m, 4 H), 2.37 (t, J = 7.58 Hz, 2 H), 4.36 (d, J = 4.80 Hz, 2 H), 5.02 - 5.27 (m, 1 H), 5.91 (s, 1 H), 7.43 (s, 1 H), 8.21 (s, 1 H), 8.62 - 8.87 (m, 1 H), 1 1 .54 (br. s., 1 H).
Compound C can be prepared according to a procedure analogous to the above example: 6-cyclopropyl-1 -(1 -methylethyl)-N-[(6-methyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]- 1 H-pyrazolo[3,4-b]pyridine-4-carboxamide
Figure imgf000211_0001
Examples
Proliferation Studies:
The EZH 1/EZH2 inhibitors, Compound A and Compound B, concentration- dependently impaired T cell receptor-induced proliferation of CD4+ T cells with a plC50 of 5.30 ± 0.06 and 4.93 ± 0.05 respectively (n=4; Figure 1 ). In contrast, Compound C had no effect on T cell receptor-induced proliferation of CD4+ T cells (n=4). Cytokine Production Studies:
The EZH 1/EZH2 inhibitors, Compound A and Compound B, concentration- dependently impaired T cell receptor-induced production of IL-10, IL-13, IL-17, IFN & TNF in CD4+ T cells with plC50 values ranging between 5.65 ± 0.06 - 5.23 ± 0.03 and 5.21 ± 0.09 - 4.94 ± 0.03 respectively (n=4; Figure 2 & Table 1 ). In contrast, Compound C had no effect on T cell receptor-induced production of IL-17 or IFN in CD4+ T cells (n=4). High concentrations of Compound C (i.e. >15 μΜ) reduced IL-10, IL-13 & TNF production (n=4).
The EZH 1/EZH2 inhibitors, Compound A and Compound B, concentration- dependently impaired T cell receptor-induced production of IL-2 in CD4+ T cells with plC50 values of 6.24 ± 0.24 and 5.76 ± 0.25 respectively (n=4; Figure 3). Compound C also impaired T cell receptor-induced production of IL-2 in CD4+ T cells, although a plC50 could not be calculated (n=4; Figure 3).
Methods: CD4 T cell isolation from human blood:
15 mis ficoll was added to 8 x 50ml_ accuspin tubes per donor and centrifuged at 2000RPM for 1 min. Whole blood samples (200 ml. + 1 % heparin) were collected from human donors. Approximately 25 ml blood was added to each ficoll pre-filled 50ml falcon tubes with filter and centrifuged at 1500 RPM for 20 min with the brake off to avoid disturbing cell separation. 45 ml with PBS was added to the PBMC fraction of each tube and centrifuged at 1500RPM for 10 min with brake. Supernatant were discarded and cell pellets resuspended in 2 ml PBS. Cell suspensions for each donor were recombined into a single falcon tube, made up to 45 ml with PBS and centrifuged at 1500RPM for 10 min. CD4+ T cells were isolated by negative depletion using a CD4 T cell Isolation Kit from Miltenyi Biotech according to manufacturers protocol.
Proliferation Studies:
96-well flat-bottom plates were coated with 10 g/mL anti-CD3 + 2 g/mL anti- CD28 at 4'C overnight. The following day, plates were washed with PBS. For proliferation studies, cells were stained with CFSE (Invitrogen) according to manufacturer's protocol. Cells were then added to anti-CD3 + anti-CD28 pre-coated 96- well plates at 0.2x106cells/well in the presence of EZH1/EZH2 inhibitors (Compound A & Compound B), an inactive control compound (Compound C) or vehicle (0.1 % DMSO) and incubated for 6 days at 37'C/ 5 % C02. Cells were then transferred to 96-well round- bottom plates, washed and then analysed by flow cytometry.
Cytokine Production Studies:
96-well flat-bottom plates were coated with 10 g/mL anti-CD3 + 2 g/mL anti- CD28 at 4'C overnight. The following day, plates were washed with PBS. For cytokine production studies, cells were added to anti-CD3 + anti-CD28 pre-coated 96-well plates at 0.4x106cells/well in the presence of EZH1/EZH2 inhibitors or vehicle and incubated at 37'C/ 5 % C02. Supernatants were taken 18 h (IL-2) or 72 h (IL-10, IL-13, IL-17, I FN, TNF) for determination of cytokine production by multiplex ELISA from Mesoscale discovery according to the manufacturers protocols. Drugs and Materials
Anti-CD3 (HU CD3 NALE MAB UCHT1 ; Cat # 555329) and anti-CD28 (HU CD28 NALE MAB CD28.2; Cat #: 555725), were obtained from BD Pharmingen and dissolved in Dulbecco's Phosphate Buffered Saline (with Ca2+/Mg2+; Gibco). Ficoll-paque (Cat # 17- 1440-03) was obtained from GE Healthcare. The CD4+ T cell isolation kit (Cat # 130-091- 155) was obtained from Miltenyi Biotec. Cells were cultured in RPMI 1640 containing 10 % FCS + 1 % pen/strep + 1 % L-glutamine. The CellTrace CFSE cell proliferation kit (Cat # C34554) was obtained from Invitrogen.
Compound A, Compound B and Compound C were all synthesised in house and dissolved in 100 % DMSO.
Data Analysis
Proliferation Studies:
Flowjo Software was used to calculate a division index using fluorescence intensity. Data are expressed as mean ± standard error of the mean; n values are the numbers of human donors used. plC50 values were generated using GraphPad Prism software and non-linear sigmoid dose curve-fit.
Cytokine Production Studies:
Data were analysed using Excel and are expressed as mean ± standard error of the mean; n values are the numbers of human donors used. plC50 values were generated using GraphPad Prism software and non-linear sigmoid dose curve-fit.

Claims

Claims
1. A method of treating a T cell mediated inflammatory immune disease or a T cell mediated hypersensitivity disease, which comprises administering to a human in need thereof an effective amount of a compound, or a pharmaceutically acceptable salt thereof, which inhibits EZH2 and/or EZH1 .
2. A method of treating T cell mediated inflammatory immune diseases, or a T cell mediated hypersensitivity disease, which comprises administering an effective amount of a compound of Formula (III), or a pharmaceutically acceptable salt thereof,
Figure imgf000214_0001
wherein
X and Z are selected independently from the group consisting of hydrogen, (C C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, unsubstituted or substituted (C3-C8)cycloalkyl, unsubstituted or substituted (C3-C8)cycloalkyl-(CrC8)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-Ci0)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 (Ci-C8)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-(C1-C8)alkyl or -(C2-C8)alkenyl, unsubstituted or substituted (C6-C10)bicycloalkyl, unsubstituted or substituted heterocycloalkyi 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-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-(d-C8)alkyl, unsubstituted or substituted (C5-C8)cycloalkenyl, unsubstituted or substituted (C5- C8)cycloalkenyl-(Ci-C8)alkyl, (C6-Ci0)bicycloalkyl, unsubstituted or substituted
heterocycloalkyi, unsubstituted or substituted heterocycloalkyl-(Ci-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, -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 (C1-C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, cycloalkyl, cycloalkenyl, bicycloalkyl, heterocycloalkyi, 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)1-2, -S(Ci-C6)alkyl(Rc)1-2, -(Ci-C6)alkyl(Rc)1-2, (Ci-C8)alkyl- heterocycloalkyl, (C3-C8)cycloalkyl-heterocycloalkyl, halo, (CrC6)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, heterocycloalkyi, 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, (CrC6)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, and -OC(0)NRaRb;
Ra and Rb are each independently hydrogen, (C1-C8)alkyl, (C2-C8)alkenyl,
(C2-C8)alkynyl, (C3-C8)cycloalkyl, (C5-C8)cycloalkenyl, (C6-Ci0)bicycloalkyl,
heterocycloalkyi, aryl, heteroaryl, wherein said (Ci-C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, cycloalkyi, cycloalkenyl, bicycloalkyi, heterocycloalkyi ,aryl or heteroaryl group is optionally substituted by 1 , 2 or 3 groups independently selected from halo, hydroxyl, (CrC4)alkoxy, amino, (C C4)alkylamino, ((Ci-C4)alkyl)((C C4)alkyl)amino, -C02H, -C02(C C4)alkyl, - CONH2,-CONH(Ci-C4)alkyl, -CON((C C4)alkyl)((Ci-C4)alkyl), -S02(C C4)alkyl, - S02NH2,-S02NH(Ci-C4)alkyl, or -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, (Ci-C4)haloalkyl, amino, (Ci-C4)alkylamino, ((Ci-C4)alkyl)((Ci-C4)alkyl)amino, hydroxyl, oxo, (Ci-C4)alkoxy, and (Ci-C4)alkoxy(CrC4)alkyl, wherein said ring is optionally fused to a (C3-C8)cycloalkyl, heterocycloalkyi, 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, heterocycloalkyi, aryl, or heteroaryl ring;
each Rc is independently (d-C^alkylamino, -NRaS02Rb, -SORa, -S02Ra, - NRaC(0)ORa, -NRaRb, or -C02Ra;
or a salt thereof.
3. A method of claim 2, wherein X and Z are selected from the group consisting of (Ci-C8)alkyl, (C3-C8)cycloalkyl, heterocycloalkyi, aryl, heteroaryl, -NRaRb, and -ORa;
Y is H or F;
R1 is selected from the group consisting of (Ci-C8)alkyl, (C3-C8)cycloalkyl, heterocycloalkyi, aryl, and heteroaryl;
R2 is hydrogen, (Ci-C8)alkyl, trifluoromethyl, alkoxy, or halo, in which said (C
C8)alkyl maybe substituted with one to two groups selected from: amino, and (C
C3)alkylamino;
R7 is hydrogen, (Ci-C3)alkyl, or alkoxy; 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, (C1-C8)alkyl, (C3-C8)cycloalkyl, aryl, heteroaryl, acylamino, (C2-C8)alkynyl, arylalkynyl, heteroarylalkynyl, -S02Ra, -S02NRaRb , and -NRaS02Rb ;
wherein any (d-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)alkyl(Rc)1-2, -S(Ci-C6)alkyl(Rc)1-2, -(CrC6)alkyl(Rc)1-2, (C C8)alkyl- heterocycloalkyl, (C3-C8)cycloalkyl-heterocycloalkyl, halo, (CrC6)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, heterocycloalkyi, aryl, heteroaryl, aryl(Ci-C4)alkyl, and heteroaryl(Ci-C4)alkyl;
each Rc is independently (Ci-C4)alkylamino, -NRaS02Rb, -SORa, -S02Ra, - NRaC(0)ORa, -NRaRb, 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,
heterocycloalkyi, aryl, heteroaryl, wherein said (Ci-C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, cycloalkyi, cycloalkenyl, bicycloalkyi, heterocycloalkyi ,aryl or heteroaryl group is optionally substituted by 1 , 2 or 3 groups independently selected from halo, hydroxyl, (d-d)alkoxy, amino, (C C4)alkylamino, ((C1-C4)alkyl)((C1-C4)alkyl)amino, -C02H, -C02(C C4)alkyl, - CONH2, -CONH(d-C4)alkyl, -CON((d-d)alkyl)((d-d)alkyl), -S02(d-d)alkyl, - S02NH2,-S02NH(d-d)alkyl, and -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-C4)alkyl)((d-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, heterocycloalkyi, 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, heterocycloalkyi, 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:
wherein in (1 ),
A is O, NH, or S; B is CH or N, and C is h drogen or CrC8 alkyl; or
Figure imgf000218_0002
wherein in (2),
D is N or C optionally substituted b hydrogen or Ci-C8 alkyl; or
Figure imgf000218_0003
wherein in (3),
E is NH or CH2; F is O or CO; and G is NH or CH
Figure imgf000218_0004
wherein in (4),
J is O, S or CO; or
Figure imgf000218_0005
wherein in (5),
Q is CH or N;
M is CH or N; and
L/(5) is hydrogen, halo, amino, cyano, (CrC8)alkyl, (C3-C8)cycloalkyl, -CORa, - C02Ra, -CONRaRb, -CONRaNRaRb, -S02Ra, -S02NRaRb, -NRaRb, -NRaC(0)Rb -
NRaS02Rb, -NRaS02NRaRb, -NRaNRaRb, -NRaNRaC(0)Rb, -N RaN RaC(0)N RaRb, -ORa, wherein any (Ci-C8)alkyl, (C3-C8)cycloalkyl, group is optionally substituted by 1 , 2 or 3 groups independently selected from (d-C6)alkyl, (C3-C8)cycloalkyl, (C5-Ce)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 imgf000219_0001
wherein in 6,
L/(6) is NH or CH2; or
Figure imgf000219_0002
wherein in 7,
M/(7) is hydrogen, halo, amino, cyano, (CrC8)alkyl, (C3-C8)cycloalkyl, heterocycloalkyi, -CORa, -C02Ra, -CONRaRb, -CONRaNRaRb, -S02Ra, - S02NRaRb, -NRaRb, -NRaC(0)Rb,-NRaS02Rb, -NRaS02NRaRb, -NRaNRaRb, - NRaNRaC(0)Rb, -NRaNRaC(0)NRaRb, -ORa,
wherein any (C1-C8)alkyl, (C3-C8)cycloalkyl, heterocycloalkyi group is optionally substituted by 1 , 2 or 3 groups independently selected from (C1-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 and Rb are defined as above; or
Figure imgf000219_0003
wherein in (8),
P is CH2, NH, O, or S; 0/(8) is CH or N; and n is 0-2; or
Figure imgf000220_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, (d-C8)alkyl, (C3-
C8)cycloalkyl, -CORa, -C02Ra, -CONRaRb, -S02Ra, -S02NRaRb, -NRaRb, - NRaC(0)Rb,-NRaS02Rb, -NRaS02NRaRb, -NRaNRaRb, -NRaNRaC(0)Rb, , -ORa, 4-(1 H- 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-Ce)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.
4. A method of claim 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 (C1-C8)alkyl, (C3-C8)cycloalkyl, or heterocycloalkyl;
R2 is hydrogen, (Ci-C3)alkyl, or halo, in which said (Ci-C3)alkyl maybe substituted with one to two groups selected from: amino, and (Ci-C3)alkylamino;
R7 is hydrogen, (Ci-C3)alkyl, or alkoxy; R3 is hydrogen, (Ci-C8)alkyl or halo;
R6 is hydrogen, halo, cyano, trifluoromethyl, amino, (Ci-C8)alkyl, (C3-C8)cycloalkyl, aryl, heteroaryl, acylamino, (C2-C8)alkynyl, arylalkynyl, heteroarylalkynyl, -S02Ra, - S02NRaRb, or -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 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, heterocycloalkyi, aryl, heteroaryl, aryl(C1-C4)alkyl, and heteroaryl(C1-C4)alkyl;
Ra and Rb are each independently hydrogen, (C1-C8)alkyl, (C2-C8)alkenyl,
(C2-C8)alkynyl, (C3-C8)cycloalkyl, (C5-C8)cycloalkenyl, (C6-Ci0)bicycloalkyl,
heterocycloalkyi, aryl, heteroaryl, wherein said (Ci-C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, cycloalkyi, cycloalkenyl, bicycloalkyi, heterocycloalkyi ,aryl or heteroaryl group is optionally substituted by 1 , 2 or 3 groups independently selected from halo, hydroxyl, (Ci-C4)alkoxy, amino, (C C4)alkylamino, ((Ci-C4)alkyl)((C C4)alkyl)amino, -C02H, -C02(C C4)alkyl, - CONH2,-CONH(Ci-C4)alkyl, -CON((C C4)alkyl)((Ci-C4)alkyl), -S02(C C4)alkyl, - S02NH2, -S02NH(CrC4)alkyl, and -S02N((Ci-C4)alkyl)((C 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, (Ci-C4)haloalkyl, amino, (Ci-C4)alkylamino, ((Ci-C4)alkyl)((Ci-C4)alkyl)amino, hydroxyl, oxo, (Ci-C4)alkoxy, and (Ci-C4)alkoxy(CrC4)alkyl, wherein said ring is optionally fused to a (C3-C8)cycloalkyl, heterocycloalkyi, 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, heterocycloalkyi, 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 imgf000221_0001
wherein in (1 ),
A is O, NH, or S; B is CH or N, and C is h drogen or Ci-C8 alkyl; or
Figure imgf000221_0002
wherein in (2),
D is N or C optionally substituted by hydrogen or Ci-C8 alkyl; or
Figure imgf000222_0001
wherein in (3),
E is NH or CH2; F is O or CO; and G is NH or CH2; or
Figure imgf000222_0002
wherein in (4),
J is O, S or CO; or
Figure imgf000222_0003
wherein in (5),
Q is CH or N;
M is CH or N; and
L/(5) is hydrogen, halo, amino, cyano, (CrC8)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 (C1-C8)alkyl, (C3-C8)cycloalkyl, group is optionally substituted by 1 , 2 or 3 groups independently selected from (C1-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 imgf000222_0004
wherein in 6,
L/(6) is NH or CH
Figure imgf000223_0001
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 (C1-C8)alkyl, (C3-C8)cycloalkyl, heterocycloalkyl group is optionally substituted by 1 , 2 or 3 groups independently selected from (d-C6)alkyl, (C3-Ce)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 imgf000223_0002
wherein in (8),
P is CH2, NH, O, or S; 0/(8) is CH or N; and n is 0-2; or
Figure imgf000223_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-(1 H- 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 -CON RaRb, -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.
5. A method of claim 2, 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, /V,/V-dimethyl-1-propanaminyl, benzyl, or 4- pyridyl;
R2 is hydrogen, (CrC3)alkyl, or halo, in which said (CrC3)alkyl may be substituted with one to two groups selected from: amino, and (Ci-C3)alkylamino;
R7 is hydrogen, (d-C3)alkyl, or alkoxy; R3 is H, methyl, or Br; and
R6 is methyl, bis(1 , 1-dimethylethyl), bis(l -methylethyl), cyclopropyl, propyl, dimethylamino, ethylamino, (2-hydroxyethyl)amino, 2-propen-1 -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-1 -yl, 4-pyridinylethynyl, phenylethynyl, 2-furanyl, 3-thienyl; 1 H-pyrazol-4-yl, 1 H-indazol-5-yl, 1 H-indazol-6-yl, 3- methyl-1 H-indazol-5-yl, 1 H-1 ,2,3-benzotriazol-5-yl, 2-oxo-2,3-dihydro-1 H-benzimidazol-5- yl, 2-0X0-2, 3-dihydro-1 H-indol-5-yl, 2-oxo-2,3-dihydro-1 H-indol-6-yl, 2,1 ,3-benzoxadiazol- 5-yl, 2-amino-6-quinazolinyl, 2,4-dioxo-1 ,2,3,4-tetrahydro-5-pyrimidinyl, 2-amino-5- pyrimidinyl, 7-oxo-1 ,5,6,7-tetrahydro-1 ,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-(1 H-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.
6. A method of any one of the above claims, wherein said T cell mediated inflammatory immune disease is selected from a group consisting of: acute disseminated alopecia universalise, Behcet's disease, Chagas' disease, chronic fatigue syndrome, dysautonomia, encephalomyelitis, ankylosing spondylitis, aplastic anemia, hidradenitis suppurativa, autoimmune hepatitis, autoimmune oophoritis, celiac disease, Crohn's disease, diabetes mellitus type 1 , giant cell arteritis, goodpasture's syndrome, Grave's disease, Guillain-Barre syndrome, Hashimoto's disease, Henoch-Schonlein purpura, Kawasaki's disease, lupus erythematosus, microscopic colitis, microscopic polyarteritis, mixed connective tissue disease, multiple sclerosis, myasthenia gravis, opsoclonus myoclonus syndrome, optic neuritis, ord's thyroiditis, pemphigus, polyarteritis nodosa, polymyalgia, Reiter's syndrome, Sjogren's syndrome, temporal arteritis, Wegener's granulomatosis, warm autoimmune haemolytic anemia, interstitial cystitis, lyme disease, morphea, sarcoidosis, scleroderma, ulcerative colitis, and vitiligo.
7. A method of any one of claims 1-5, wherein the T cell mediated hypersensitivity disease is selected from the group consisting of: contact hypersensitivity, contact dermatitis (including that due to poison ivy), uticaria, skin allergies, respiratory allergies (hayfever, allergic rhinitis) and gluten-sensitive enteropathy (Celliac disease).
8. A method of treating a T cell mediated inflammatory immune disease or a T cell mediated hypersensitivity disease, which comprises administering to a human in need thereof an effective amount of a compound, or a pharmaceutically acceptable salt thereof, which inhibits EZH2 and/or EZH1 , in a pharmaceutically acceptable composition.
9. A method of treating a T cell mediated inflammatory immune disease or a T cell mediated hypersensitivity disease, which comprises administering to a human in need thereof an effective amount of a compound, or a pharmaceutically acceptable salt thereof, as defined in any one of claims 2-5, in a pharmaceutically acceptable composition.
10. A compound or a pharmaceutically acceptable salt thereof which inhibits EZH2 and/or EZH1 for use in treating T cell mediated inflammatory immune diseases or T cell mediated hypersensitivity diseases.
1 1 . Use of a compound or a pharmaceutically acceptable salt thereof which inhibits EZH2 and/or EZH1 in the manufacture of a medicament for treating T cell mediated inflammatory immune diseases or T cell mediated hypersensitivity diseases.
12. Compound or use according to claim 10 or claim 1 1 wherein said T cell mediated inflammatory immune disease is selected from a group consisting of: acute disseminated alopecia universalise, Behcet's disease, Chagas' disease, chronic fatigue syndrome, dysautonomia, encephalomyelitis, ankylosing spondylitis, aplastic anemia, hidradenitis suppurativa, autoimmune hepatitis, autoimmune oophoritis, celiac disease, Crohn's disease, diabetes mellitus type 1 , giant cell arteritis, goodpasture's syndrome, Grave's disease, Guillain-Barre syndrome, Hashimoto's disease, Henoch-Schonlein purpura, Kawasaki's disease, lupus erythematosus, microscopic colitis, microscopic polyarteritis, mixed connective tissue disease, multiple sclerosis, myasthenia gravis, opsoclonus myoclonus syndrome, optic neuritis, ord's thyroiditis, pemphigus, polyarteritis nodosa, polymyalgia, Reiter's syndrome, Sjogren's syndrome, temporal arteritis, Wegener's granulomatosis, warm autoimmune haemolytic anemia, interstitial cystitis, lyme disease, morphea, sarcoidosis, scleroderma, ulcerative colitis, and vitiligo.
13. Compound or use according to claim 10 or claim 1 1 , wherein said T cell mediated hypersensitivity disease is selected from the group consisting of: contact hypersensitivity, contact dermatitis (including that due to poison ivy), uticaria, skin allergies, respiratory allergies (hayfever, allergic rhinitis) and gluten-sensitive enteropathy (Celliac disease).
14. Compound or use according to any one of claims 10 to 13, wherein the compound is a compound as defined in any one of claims 2 to 5.
PCT/US2012/063243 2011-11-04 2012-11-02 Method of treatment WO2013067302A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201161555671P 2011-11-04 2011-11-04
US61/555,671 2011-11-04

Publications (1)

Publication Number Publication Date
WO2013067302A1 true WO2013067302A1 (en) 2013-05-10

Family

ID=48192804

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2012/063243 WO2013067302A1 (en) 2011-11-04 2012-11-02 Method of treatment

Country Status (1)

Country Link
WO (1) WO2013067302A1 (en)

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
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
US9051269B2 (en) 2011-11-18 2015-06-09 Constellation Pharmaceuticals, Inc. Modulators of methyl modifying enzymes, compositions and uses thereof
WO2015141616A1 (en) * 2014-03-17 2015-09-24 第一三共株式会社 1,3-benzodioxole derivative
US20150344459A1 (en) * 2012-12-21 2015-12-03 Epizyme, Inc. 1,4-pyridone bicyclic heteroaryl 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
US9745305B2 (en) 2013-03-15 2017-08-29 Constellation Pharmaceuticals, Inc. Modulators of methyl modifying enzymes, compositions and uses thereof
CN107105651A (en) * 2014-11-06 2017-08-29 达纳-法伯癌症研究所股份有限公司 Ezh2 inhibitor and application thereof
JP2017533922A (en) * 2014-11-06 2017-11-16 ダナ−ファーバー キャンサー インスティテュート, インコーポレイテッド Use of compositions that modulate chromatin structure for graft-versus-host disease (GVHD)
US9969716B2 (en) 2013-08-15 2018-05-15 Constellation Pharmaceuticals, Inc. Indole derivatives as modulators of methyl modifying enzymes, compositions and uses thereof
US10174019B2 (en) 2013-03-15 2019-01-08 Epizyme, Inc. Substituted 6,5-fused bicyclic heteroaryl compounds
US10457640B2 (en) 2016-10-19 2019-10-29 Constellation Pharmaceuticals, Inc. Synthesis of inhibitors of EZH2
WO2019212256A1 (en) * 2018-05-02 2019-11-07 제이더블유중외제약 주식회사 Novel heterocycle derivative
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
US10633371B2 (en) 2016-04-22 2020-04-28 Dana-Farber Cancer Institute, Inc. EZH2 inhibitors and uses thereof
WO2020087170A1 (en) * 2018-10-30 2020-05-07 Repare Therapeutics Inc. Compounds, pharmaceutical compositions, and methods of preparing compounds and of their use as atr kinase inhibitors
JP2020521775A (en) * 2017-05-31 2020-07-27 ノバルティス アーゲー 5,6-Fused bicyclic compounds and compositions for the treatment of parasitic disease
WO2020171606A1 (en) * 2019-02-19 2020-08-27 한미약품 주식회사 Novel heterotricyclic derivative compound and use of same
WO2020179859A1 (en) 2019-03-06 2020-09-10 第一三共株式会社 Pyrrolopyrazole derivative
CN113292489A (en) * 2021-06-16 2021-08-24 泓博智源(开原)药业有限公司 Preparation method of dichlorodialkyl nicotinonitrile
WO2022035303A1 (en) * 2020-08-13 2022-02-17 Hanmi Pharmaceutical Co., Ltd. Novel dioxoloisoquinolinone derivatives and use thereof
CN114174292A (en) * 2019-07-22 2022-03-11 修复治疗公司 Substituted 2-morpholinopyridine derivatives as ATR kinase inhibitors
US11535629B2 (en) 2020-08-13 2022-12-27 Hanmi Pharmaceutical Co., Ltd. Dioxoloisoquinolinone derivatives and use thereof
RU2792163C2 (en) * 2018-05-02 2023-03-17 ДжейВ ФАРМАСЬЮТИКАЛ КОРПОРЕЙШН New heterocyclic derivative

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005034845A2 (en) * 2003-09-12 2005-04-21 Supergen, Inc. Compositions and methods for treatment of cancer
WO2007053114A1 (en) * 2005-10-31 2007-05-10 S*Bio Pte Ltd Method of predicting a response to hdac inhibitors
WO2010090723A2 (en) * 2009-02-04 2010-08-12 University Of Georgia Research Foundation, Inc. Methods of inhibiting fibrogenesis and treating fibrotic disease
US20110064664A1 (en) * 2007-10-08 2011-03-17 The Board Of Regents Of The University Of Texas System Methods and compositions involving chitosan nanoparticles

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005034845A2 (en) * 2003-09-12 2005-04-21 Supergen, Inc. Compositions and methods for treatment of cancer
WO2007053114A1 (en) * 2005-10-31 2007-05-10 S*Bio Pte Ltd Method of predicting a response to hdac inhibitors
US20110064664A1 (en) * 2007-10-08 2011-03-17 The Board Of Regents Of The University Of Texas System Methods and compositions involving chitosan nanoparticles
WO2010090723A2 (en) * 2009-02-04 2010-08-12 University Of Georgia Research Foundation, Inc. Methods of inhibiting fibrogenesis and treating fibrotic disease

Cited By (53)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9051269B2 (en) 2011-11-18 2015-06-09 Constellation Pharmaceuticals, Inc. 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
US9980952B2 (en) 2012-02-10 2018-05-29 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
US9371331B2 (en) 2012-02-10 2016-06-21 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
USRE47428E1 (en) 2012-02-10 2019-06-11 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
US20150344459A1 (en) * 2012-12-21 2015-12-03 Epizyme, Inc. 1,4-pyridone bicyclic heteroaryl compounds
US9701666B2 (en) * 2012-12-21 2017-07-11 Epizyme, Inc. 1,4-pyridone bicyclic heteroaryl compounds
US10150759B2 (en) 2012-12-21 2018-12-11 Epizyme, Inc. 1,4-pyridone bicycic heteroaryl compounds
US10174019B2 (en) 2013-03-15 2019-01-08 Epizyme, Inc. Substituted 6,5-fused bicyclic heteroaryl compounds
US9745305B2 (en) 2013-03-15 2017-08-29 Constellation Pharmaceuticals, Inc. Modulators of methyl modifying enzymes, compositions and uses thereof
US9969716B2 (en) 2013-08-15 2018-05-15 Constellation Pharmaceuticals, Inc. Indole derivatives as modulators of methyl modifying enzymes, compositions and uses thereof
US10954219B2 (en) 2014-03-17 2021-03-23 Daiichi Sankyo Company, Limited 1,3-benzodioxole derivative
RU2679131C2 (en) * 2014-03-17 2019-02-06 Дайити Санкио Компани, Лимитед 1,3-benzodioxole derivative
US10017500B2 (en) 2014-03-17 2018-07-10 Daiichi Sankyo Company, Limited 1,3-benzodioxole derivative
WO2015141616A1 (en) * 2014-03-17 2015-09-24 第一三共株式会社 1,3-benzodioxole derivative
EP3214935A4 (en) * 2014-11-06 2018-08-29 Dana-Farber Cancer Institute, Inc. Ezh2 inhibitors and uses thereof
JP2017533922A (en) * 2014-11-06 2017-11-16 ダナ−ファーバー キャンサー インスティテュート, インコーポレイテッド Use of compositions that modulate chromatin structure for graft-versus-host disease (GVHD)
CN107105651A (en) * 2014-11-06 2017-08-29 达纳-法伯癌症研究所股份有限公司 Ezh2 inhibitor and application thereof
US11236082B2 (en) 2014-11-06 2022-02-01 Dana-Farber Cancer Institute, Inc. EZH2 inhibitors and uses thereof
EP3885343A1 (en) 2014-11-06 2021-09-29 Dana-Farber Cancer Institute, Inc. Indole compounds as ezh2 inhibitors and uses thereof
AU2015342774B2 (en) * 2014-11-06 2020-01-30 Dana-Farber Cancer Institute, Inc. EZH2 inhibitors and uses thereof
EP3215160A4 (en) * 2014-11-06 2018-08-08 Dana-Farber Cancer Institute, Inc. Use of compositions modulating chromatin structure for graft versus host disease (gvhd)
AU2015342774C1 (en) * 2014-11-06 2021-08-05 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
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
US10633371B2 (en) 2016-04-22 2020-04-28 Dana-Farber Cancer Institute, Inc. EZH2 inhibitors and uses thereof
US10457640B2 (en) 2016-10-19 2019-10-29 Constellation Pharmaceuticals, Inc. Synthesis of inhibitors of EZH2
JP7187490B2 (en) 2017-05-31 2022-12-12 ノバルティス アーゲー 5,6-Fused Bicyclic Compounds and Compositions for Treatment of Parasitosis
JP2020521775A (en) * 2017-05-31 2020-07-27 ノバルティス アーゲー 5,6-Fused bicyclic compounds and compositions for the treatment of parasitic disease
WO2019212256A1 (en) * 2018-05-02 2019-11-07 제이더블유중외제약 주식회사 Novel heterocycle derivative
KR20200139171A (en) * 2018-05-02 2020-12-11 제이더블유중외제약 주식회사 Novel heterocycle derivatives
CN112313232A (en) * 2018-05-02 2021-02-02 Jw中外制药公司 Novel heterocyclic derivatives
KR102653799B1 (en) 2018-05-02 2024-04-03 제이더블유중외제약 주식회사 Novel heterocycle derivatives
CN112313232B (en) * 2018-05-02 2024-03-08 Jw中外制药公司 Novel heterocyclic derivatives
RU2792163C2 (en) * 2018-05-02 2023-03-17 ДжейВ ФАРМАСЬЮТИКАЛ КОРПОРЕЙШН New heterocyclic derivative
CN113454080A (en) * 2018-10-30 2021-09-28 修复治疗公司 Compounds, pharmaceutical compositions and methods of making compounds and methods of use thereof
WO2020087170A1 (en) * 2018-10-30 2020-05-07 Repare Therapeutics Inc. Compounds, pharmaceutical compositions, and methods of preparing compounds and of their use as atr kinase inhibitors
RU2806857C2 (en) * 2018-10-30 2023-11-08 Рипэйр Терапьютикс Инк. Compounds, pharmaceutical compositions, methods for preparing the compounds and their use as atr kinase inhibitors
AU2020226042B2 (en) * 2019-02-19 2022-07-14 Hanmi Pharmaceutical Co., Ltd. Novel heterotricyclic derivative compound and use of same
WO2020171606A1 (en) * 2019-02-19 2020-08-27 한미약품 주식회사 Novel heterotricyclic derivative compound and use of same
CN113423710A (en) * 2019-02-19 2021-09-21 韩美药品株式会社 Novel heterocyclic tricyclic derivative compound and use thereof
WO2020179859A1 (en) 2019-03-06 2020-09-10 第一三共株式会社 Pyrrolopyrazole derivative
CN114174292A (en) * 2019-07-22 2022-03-11 修复治疗公司 Substituted 2-morpholinopyridine derivatives as ATR kinase inhibitors
WO2022035303A1 (en) * 2020-08-13 2022-02-17 Hanmi Pharmaceutical Co., Ltd. Novel dioxoloisoquinolinone derivatives and use thereof
US11535629B2 (en) 2020-08-13 2022-12-27 Hanmi Pharmaceutical Co., Ltd. Dioxoloisoquinolinone derivatives and use thereof
JP2023517272A (en) * 2020-08-13 2023-04-25 ハンミ ファーマシューティカル カンパニー リミテッド NOVEL DIOXOLOISOQUINOLINONE DERIVATIVES AND USE THEREOF
JP7387907B2 (en) 2020-08-13 2023-11-28 ハンミ ファーマシューティカル カンパニー リミテッド Novel dioxoloisoquinolinone derivative compounds and their uses {NOVEL DIOXOLOISOQUINOLINONE DERIVATIVES AND USE THEREOF}
CN113292489A (en) * 2021-06-16 2021-08-24 泓博智源(开原)药业有限公司 Preparation method of dichlorodialkyl nicotinonitrile

Similar Documents

Publication Publication Date Title
WO2013067302A1 (en) Method of treatment
US9446041B2 (en) Method of treatment
US9649307B2 (en) Indoles
US9073924B2 (en) Azaindazoles
US8637509B2 (en) Azaindazoles
CA2933480C (en) Inhibitors of lysine specific demethylase-1
EP2566328B1 (en) Indazoles
WO2013067300A1 (en) Method of treatment
WO2024083773A1 (en) Triazole-substituted imidazo[1,2-a]pyrimidines as cgas inhibitors

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: 12846342

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 12846342

Country of ref document: EP

Kind code of ref document: A1