WO2013067302A1 - Méthode de traitement - Google Patents

Méthode de traitement 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
English (en)
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/fr

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

La présente invention concerne une méthode de traitement de maladies immunes inflammatoires médiées par les lymphocytes T ou de maladies d'hypersensibilité médiées par les lymphocytes T, la méthode consistant à administrer à un être humain nécessitant ce traitement une quantité efficace d'un composé qui inhibe EZH2 et/ou EZH1, ou un sel pharmaceutiquement acceptable de ce composé.
PCT/US2012/063243 2011-11-04 2012-11-02 Méthode de traitement WO2013067302A1 (fr)

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 (fr) 2013-05-10

Family

ID=48192804

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2012/063243 WO2013067302A1 (fr) 2011-11-04 2012-11-02 Méthode de traitement

Country Status (1)

Country Link
WO (1) WO2013067302A1 (fr)

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2015506985A (ja) * 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. メチル基変更酵素の調節物質、組成物及びその使用
US9051269B2 (en) 2011-11-18 2015-06-09 Constellation Pharmaceuticals, Inc. Modulators of methyl modifying enzymes, compositions and uses thereof
WO2015141616A1 (fr) * 2014-03-17 2015-09-24 第一三共株式会社 Dérivé 1,3-benzodioxole
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 (zh) * 2014-11-06 2017-08-29 达纳-法伯癌症研究所股份有限公司 Ezh2抑制剂及其用途
JP2017533922A (ja) * 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 移植片対宿主病(gvhd)のためのクロマチン構造を調節する組成物の使用
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 (fr) * 2018-05-02 2019-11-07 제이더블유중외제약 주식회사 Nouveau dérivé hétérocyclique
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 (fr) * 2018-10-30 2020-05-07 Repare Therapeutics Inc. Composés, compositions pharmaceutiques, procédés de préparation de composés et leur utilisation en tant qu'inhibiteurs de kinase atr
JP2020521775A (ja) * 2017-05-31 2020-07-27 ノバルティス アーゲー 寄生虫症の治療のための5,6−縮合二環式化合物及び組成物
WO2020171606A1 (fr) * 2019-02-19 2020-08-27 한미약품 주식회사 Nouveau composé hétérocyclique et utilisation associée
WO2020179859A1 (fr) 2019-03-06 2020-09-10 第一三共株式会社 Dérivé de pyrrolopyrazole
CN113292489A (zh) * 2021-06-16 2021-08-24 泓博智源(开原)药业有限公司 二氯代二烷基烟腈的制备方法
WO2022035303A1 (fr) * 2020-08-13 2022-02-17 Hanmi Pharmaceutical Co., Ltd. Nouveaux dérivés de dioxoloisoquinolinone et leur utilisation
CN114174292A (zh) * 2019-07-22 2022-03-11 修复治疗公司 作为atr激酶抑制剂的取代的2-吗啉代吡啶衍生物
US11535629B2 (en) 2020-08-13 2022-12-27 Hanmi Pharmaceutical Co., Ltd. Dioxoloisoquinolinone derivatives and use thereof
RU2792163C2 (ru) * 2018-05-02 2023-03-17 ДжейВ ФАРМАСЬЮТИКАЛ КОРПОРЕЙШН Новое гетероциклическое производное

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005034845A2 (fr) * 2003-09-12 2005-04-21 Supergen, Inc. Compositions et methodes de traitement du cancer
WO2007053114A1 (fr) * 2005-10-31 2007-05-10 S*Bio Pte Ltd Méthode permettant de prévoir une réponse aux inhibiteurs hdac
WO2010090723A2 (fr) * 2009-02-04 2010-08-12 University Of Georgia Research Foundation, Inc. Procédés d'inhibition de fibrogenèse et de traitement de maladie fibrotique
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 (fr) * 2003-09-12 2005-04-21 Supergen, Inc. Compositions et methodes de traitement du cancer
WO2007053114A1 (fr) * 2005-10-31 2007-05-10 S*Bio Pte Ltd Méthode permettant de prévoir une réponse aux inhibiteurs hdac
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 (fr) * 2009-02-04 2010-08-12 University Of Georgia Research Foundation, Inc. Procédés d'inhibition de fibrogenèse et de traitement de maladie fibrotique

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 (ja) * 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. メチル基変更酵素の調節物質、組成物及びその使用
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 (ru) * 2014-03-17 2019-02-06 Дайити Санкио Компани, Лимитед Производное 1,3-бензодиоксола
US10017500B2 (en) 2014-03-17 2018-07-10 Daiichi Sankyo Company, Limited 1,3-benzodioxole derivative
WO2015141616A1 (fr) * 2014-03-17 2015-09-24 第一三共株式会社 Dérivé 1,3-benzodioxole
EP3214935A4 (fr) * 2014-11-06 2018-08-29 Dana-Farber Cancer Institute, Inc. Inhibiteurs d'ezh2 et leurs utilisations
JP2017533922A (ja) * 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 移植片対宿主病(gvhd)のためのクロマチン構造を調節する組成物の使用
CN107105651A (zh) * 2014-11-06 2017-08-29 达纳-法伯癌症研究所股份有限公司 Ezh2抑制剂及其用途
US11236082B2 (en) 2014-11-06 2022-02-01 Dana-Farber Cancer Institute, Inc. EZH2 inhibitors and uses thereof
EP3885343A1 (fr) 2014-11-06 2021-09-29 Dana-Farber Cancer Institute, Inc. Composés d'indole comme inhibiteurs de ezh2 et utilisations associées
AU2015342774B2 (en) * 2014-11-06 2020-01-30 Dana-Farber Cancer Institute, Inc. EZH2 inhibitors and uses thereof
EP3215160A4 (fr) * 2014-11-06 2018-08-08 Dana-Farber Cancer Institute, Inc. Utilisation de compositions modulant la structure de la chromatine contre la maladie du greffon contre l'hôte (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 (ja) 2017-05-31 2022-12-12 ノバルティス アーゲー 寄生虫症の治療のための5,6-縮合二環式化合物及び組成物
JP2020521775A (ja) * 2017-05-31 2020-07-27 ノバルティス アーゲー 寄生虫症の治療のための5,6−縮合二環式化合物及び組成物
WO2019212256A1 (fr) * 2018-05-02 2019-11-07 제이더블유중외제약 주식회사 Nouveau dérivé hétérocyclique
KR20200139171A (ko) * 2018-05-02 2020-12-11 제이더블유중외제약 주식회사 신규한 헤테로 사이클 유도체
CN112313232A (zh) * 2018-05-02 2021-02-02 Jw中外制药公司 新型杂环衍生物
KR102653799B1 (ko) 2018-05-02 2024-04-03 제이더블유중외제약 주식회사 신규한 헤테로 사이클 유도체
CN112313232B (zh) * 2018-05-02 2024-03-08 Jw中外制药公司 新型杂环衍生物
RU2792163C2 (ru) * 2018-05-02 2023-03-17 ДжейВ ФАРМАСЬЮТИКАЛ КОРПОРЕЙШН Новое гетероциклическое производное
CN113454080A (zh) * 2018-10-30 2021-09-28 修复治疗公司 化合物、药物组合物和制备化合物的方法以及其使用方法
WO2020087170A1 (fr) * 2018-10-30 2020-05-07 Repare Therapeutics Inc. Composés, compositions pharmaceutiques, procédés de préparation de composés et leur utilisation en tant qu'inhibiteurs de kinase atr
RU2806857C2 (ru) * 2018-10-30 2023-11-08 Рипэйр Терапьютикс Инк. Соединения, фармацевтические композиции, способы приготовления соединений и их применение в качестве ингибиторов киназы atr
AU2020226042B2 (en) * 2019-02-19 2022-07-14 Hanmi Pharmaceutical Co., Ltd. Novel heterotricyclic derivative compound and use of same
WO2020171606A1 (fr) * 2019-02-19 2020-08-27 한미약품 주식회사 Nouveau composé hétérocyclique et utilisation associée
CN113423710A (zh) * 2019-02-19 2021-09-21 韩美药品株式会社 新型杂环三环衍生物化合物及其用途
WO2020179859A1 (fr) 2019-03-06 2020-09-10 第一三共株式会社 Dérivé de pyrrolopyrazole
CN114174292A (zh) * 2019-07-22 2022-03-11 修复治疗公司 作为atr激酶抑制剂的取代的2-吗啉代吡啶衍生物
WO2022035303A1 (fr) * 2020-08-13 2022-02-17 Hanmi Pharmaceutical Co., Ltd. Nouveaux dérivés de dioxoloisoquinolinone et leur utilisation
US11535629B2 (en) 2020-08-13 2022-12-27 Hanmi Pharmaceutical Co., Ltd. Dioxoloisoquinolinone derivatives and use thereof
JP2023517272A (ja) * 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 新規なジオキソロイソキノリノン誘導体化合物及びその用途{novel dioxoloisoquinolinone derivatives and use thereof}
JP7387907B2 (ja) 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 新規なジオキソロイソキノリノン誘導体化合物及びその用途{novel dioxoloisoquinolinone derivatives and use thereof}
CN113292489A (zh) * 2021-06-16 2021-08-24 泓博智源(开原)药业有限公司 二氯代二烷基烟腈的制备方法

Similar Documents

Publication Publication Date Title
WO2013067302A1 (fr) Méthode de traitement
US9446041B2 (en) Method of treatment
US9649307B2 (en) Indoles
US9073924B2 (en) Azaindazoles
US8637509B2 (en) Azaindazoles
CA2933480C (fr) Inhibiteur de la demethylase-1 specifique de la lysine
EP2566328B1 (fr) Indazoles
WO2013067300A1 (fr) Méthode de traitement
WO2024083773A1 (fr) Imidazo[1,2-a]pyrimidines substituées par triazole en tant qu'inhibiteurs de cgas

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