WO2013066839A2 - Compounds and methods - Google Patents

Compounds and methods Download PDF

Info

Publication number
WO2013066839A2
WO2013066839A2 PCT/US2012/062514 US2012062514W WO2013066839A2 WO 2013066839 A2 WO2013066839 A2 WO 2013066839A2 US 2012062514 W US2012062514 W US 2012062514W WO 2013066839 A2 WO2013066839 A2 WO 2013066839A2
Authority
WO
WIPO (PCT)
Prior art keywords
pyrimidin
trifluoromethyl
oxadiazol
pyridin
amine
Prior art date
Application number
PCT/US2012/062514
Other languages
French (fr)
Other versions
WO2013066839A3 (en
Inventor
Shomir Ghosh
Mercedes Lobera
Ryan Paul Trump
Cunyu Zhang
Original Assignee
Glaxosmithkline Llc
Tempero Pharmaceuticals, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Glaxosmithkline Llc, Tempero Pharmaceuticals, Inc. filed Critical Glaxosmithkline Llc
Publication of WO2013066839A2 publication Critical patent/WO2013066839A2/en
Publication of WO2013066839A3 publication Critical patent/WO2013066839A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/4841Filling excipients; Inactive ingredients
    • A61K9/4866Organic macromolecular compounds

Definitions

  • the present invention relates to compounds that inhibit histone deacetylase (HDAC) enzymes, the preparation of these compounds, the use of these compounds in the treatment of diseases or conditions ameliorated by inhibition of HDAC activity and pharmaceutical compositions comprising these compounds.
  • HDAC histone deacetylase
  • HDACs Histone deacetylases
  • HDAC1 1 are mainly localized to the nucleus; class II HDACs (HDAC4, HDAC5, HDAC6, HDAC7, HDAC9 and HDAC10), which shuttle between the nucleus and the cytoplasm; and class III HDACs (SIRT1-7), whose cellular localization includes various organelles.
  • Class II HDACs are further characterized as class lla HDACs and class lib HDACs.
  • HDAC9 is a class lla histone deacetylase highly expressed in human Tregs.
  • HDAC9 deficiency 1 ) increases Foxp3 expression (and other Treg markers), 2) increases Foxp3 and histone 3 acetylation, 3) increases Foxp3 DNA binding, 4) increases Treg numbers, 5) increases suppressive activity in vitro and in vivo, and 6) ameliorates murine colitis.
  • Tregs which are deficient in HDAC9 induce permanent tolerance of fully mismatched cardiac allografts.
  • HDAC9 inhibitors maybe useful for treatment of diseases and disorders associated with abnormal cell proliferation, differentiation and survival, e.g. breast and prostate tumors.
  • HDAC7 a class lla histone deacetylase
  • HDAC7 enhances Treg suppression in vitro and in vivo.
  • HDAC7 enhances FOXP3+ Treg function and induces long-term allograft survival.
  • HDAC6 inhibition in vivo decreased severity of colitis in the dextran sodium sulphate-induced colitis model and the
  • CD4 + CD62Lhigh adoptive transfer model of colitis In addition, inhibition of HDAC6 with a subtherapeutic dose of rapamycin led to prolonged cardiac allograft survival.
  • an orally available small molecule selective inhibitor of Class II HDAC activity (more specifically HDAC9 or HDAC7 or HDAC6) is expected to modulate autoimmune diseases through expansion and enhancement of Treg activity.
  • HDAC4 and 5 impair myogenesis by modulating the stability and activity of HDAC-MEF2 complexes and maybe potentially useful for the treatment of muscle and heart diseases including cardiac hypertrophy and heart failure. Also, inhibition of Class II HDAC activity represents a novel approach for disrupting or intervening in cell cycle regulation.
  • HDAC9 is also highly expressed in human B cells. Relative to normal B cells, expression of HDAC9 is deregulated in cell lines derived from B cell tumours and HDAC9 is highly overexpressed in cells derived from patients with non-Hodgkin's lymphoma (http://icr.ac.uk research/team_leaders/Zelent_Arthur/Zelent_Arthur_RI/index.shtml). HDAC4 and HDAC9 have both been reported to be overexpressed in CD19+ cells from patients with Waldenstrom Macroglobulinemia (Sun et al., Clinical Lymphoma, Myeloma & Leukemia, 201 1 , p. 152).
  • Class Ma HDACs (HDAC4, HDAC5, HDAC7 and HDAC9) have been reported to associate with Bcl-6, a transcription factor implicated in the pathogenesis of B-cell malignancies (Lemercier et al, Journal of Biological Chemistry, 2002, p. 22045, and Petrie et al, Journal of Biological Chemistry, 2003, p. 16059). Due to these interactions class lla HDACs have been suggested to modulate the transcriptional repression of BCL6 and participate in its role in B-cell activation and differentiation, inflammation, and cell-cycle regulation (Verdin et al. TRENDS in Genetics, 2003, p. 286).
  • HDAC6 a class lib HDAC, has been reported to play an important role in aggresomal protein degradation, making it a target for the treatment of B cell
  • HDAC4 small molecule selective inhibitor of HDAC4, HDAC5, HDAC6, HDAC7, HDAC8 and/or HDAC9 is expected to be beneficial in the treatment of B-cell malignancies by targeting one or several of the above enzymes.
  • HDAC4 has been linked to a variety of neurodegenerative disorders: it is a downstream target of Parkin (associating it to Parkinson's disease), it is a major component of intranuclear inclusions produced in NIIND. HDAC4 also contains a conserved glutamine rich domain, such domain has been observed to increase susceptibility to amyloid formation
  • HDAC4 and HDAC5 localization are regulated by neuronal activity, and HDAC5 nuclear import is increased in diseased neurons of Huntington's disease patients.
  • HDAC7 has been implicated in regulating ataxin-7 turnover in a SCA-7 model (Mookerjee S et al., J Neurosci., 2009, p. 15134).
  • HDAC6 is expressed in most neurons and most abundantly in cerebellar Purkinje cells, the degeneration of this type of neurons is observed in patients with spinocerebellar ataxia type 1 (SCA1 ) or SCA7. HDAC6 is involved in regulating microtubule dynamics and protein degradation and a defect in microtubule-based transport may contribute to the neuronal toxicity observed in Huntington's disease (Kazantsev et al. Nature Reviews Drug Discovery, 2008, p. 854). Additionally, HDAC6 activity has been shown to be required for autophagic degradation of aggregated huntingtin, suggesting a role in protecting cells from polyQ toxicity (Iwata, et al., J. Biol. Chem., 2005, p. 40282).
  • HDAC4 or HDAC5 or HDAC6 or HDAC7or HDAC9 is expected to be beneficial in the treatment of neurodegenerative diseases.
  • Class II HDAC inhibitors have therapeutic potential in the study and/or treatment of the various diseases or conditions described herein.
  • HDAC inhibitors Many of the known small-molecule HDAC inhibitors, however, inhibit all HDAC isoforms. It would be advantageous to identify HDAC inhibitors that inhibited one or more, but not all HDAC isoforms.
  • the invention is directed to a compound according to Formula (I):
  • A is C 3-6 cycloalkyl (C 3-6 cycloalkylenyl), 4-7 membered heterocycloalkyl (4-7 membered heterocycloalkylenyl), phenyl (phenylenyl), naphthyl (naphthylenyl), 5-6 membered heteroaryl ( 5-6 membered heteroarylenyl), or 9-10 membered heteroaryl (9-10 membered heteroarylenyl), wherein said C 3 - 6 cycloalkyl, 4-7 membered
  • heterocycloalkyl phenyl, naphthyl, 5-6 membered heteroaryl, or 9-10 membered heteroaryl is optionally substituted by 1-3 groups each independently selected from the group consisting of Ci -4 alkyl, aryl, arylCi -4 alkyl-, halogen, cyano, haloCi -4 alkyl, d ⁇ alkoxy, haloCi -4 alkoxy, -NR'R" and -Ci -4 alkylNR'R", where R' and R" are independently selected from the group consisting of H and
  • n 0, 1 , 2, 3 or 4;
  • R 1 is H or C 1-4 alkyl
  • n 0, 1 , 2, 3 or 4;
  • B is C 3-6 cycloalkyl, 4-7 membered heterocycloalkyl, phenyl, naphthyl, 5-6 membered heteroaryl, or 9-10 membered heteroaryl,
  • R B groups each independently selected from the group consisting of Ci -4 alkoxy, aryl, arylC 1-4 alkyl-, 4-7 membered heterocycloalkyl, 4-7 membered
  • any of said aryl that is the aryl group or the aryl moiety of the
  • heterocycloalkyl that is the 4-7 membered heterocycloalkyl group or the heterocycloalkyl moiety of the 4-7 membered
  • heterocycloalkylCi -4 alkyl- group of R B , R Ba , and R Bb ) or heteroaryl that is the 5-6 or 9-10 membered heteroaryl group or the heteroaryl moiety of the 5-6 or 9-10 membered heteroarylCi -4 alkyl- group of R B , R Ba , and R Bb
  • any heterocycloalkyl (that is, the 4-7 membered heterocycloalkyl group or the heterocycloalkyl moiety of the heterocycloalkylCi -4 alkoxy, or heterocycloalkyl-carbonyl groups) is optionally substituted by 1 , 2 or 3 groups each independently selected from the group consisting of Ci -4 alkyl and Ci -4 alkyl-CO-, R' and R" are independently selected from the group consisting of H and Ci -4 alkyl, and p is 0-4.
  • compositions comprising compounds of the invention and a pharmaceutical carrier are also provided.
  • the invention further encompasses a method of treating a subject having a disease or disorder mediated by inhibition of a HDAC comprising administering the compound of Formula (I), or a salt thereof.
  • alkyl represents a saturated, straight, or branched hydrocarbon moiety.
  • exemplary alkyls include, but are not limited to methyl (Me), ethyl (Et), n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, f-butyl, n-pentyl, / ' so-pentyl (3-methyl- butyl), neo-pentyl (2,2-dimethylpropyl), etc.
  • Cr 4 alkyl refers to an alkyl containing from 1 to 4 carbon atoms.
  • any substituent term containing a "C 0 alkyl” moiety indicates that the substituent term excludes the presence of an alkyl moiety in that substituent.
  • the term “-C 0 - 4 alkyl-NR Ba R Bb” is intended to describe both "- NR Ba R Bb " and “-Ci.4alkyl-NR Ba R Bb " substituents.
  • alkyl When the term “alkyl” is used in combination with other substituent groups, such as “haloalkyl” or “cycloalkyl-alkyl” or “arylalkyl”, the term “alkyl” is intended to encompass a divalent straight or branched-chain hydrocarbon radical.
  • arylalkyl is intended to mean the radical -alkylaryl, wherein the alkyl moiety thereof is a divalent straight or branched-chain carbon radical and the aryl moiety thereof is as defined herein, and is represented by the bonding arrangement present in a benzyl group (-CH 2 -phenyl).
  • cycloalkyl refers to a non-aromatic, saturated, cyclic hydrocarbon ring.
  • C 3-8 cycloalkyl refers to a non-aromatic cyclic hydrocarbon ring having from three to eight ring carbon atoms.
  • Exemplary "C 3-8 cycloalkyl” groups useful in the present invention include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl.
  • Alkoxy refers to a group containing an alkyl radical attached through an oxygen linking atom.
  • C 1-4 alkoxy refers to a straight- or branched-chain hydrocarbon radical having at least 1 and up to 4 carbon atoms attached through an oxygen linking atom.
  • Exemplary "(Ci -4 )alkoxy” groups useful in the present invention include, but are not limited to, methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, s-butoxy, and f-butoxy.
  • Aryl represents a group or moiety comprising an aromatic, monocyclic or bicyclic hydrocarbon radical containing from 6 to 10 carbon ring atoms and to which may be fused one or more cycloalkyl rings.
  • Heterocycloalkyl represents a group or moiety comprising a stable, non- aromatic, monocyclic or bicyclic radical, which is saturated or partially unsaturated, containing 3 to 10 ring atoms, which includes 1 to 4 heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur.
  • the heterocycloalkyl may be attached by any atom of the monocyclic or bicyclic radical which results in the creation of a stable structure.
  • This term encompasses bicyclic heterocycloalkyl moieties where the rings are joined at two atoms per ring, as exemplified by the bonding arrangement in 2,5- diazabicyclo[2.2.1 ]heptyl, 2-azabicyclo[2.2.1 ]heptyl, 2-oxa-5-azabicyclo[2.2.1 ]heptyl, 7- oxa-2-azabicyclo[2.2.1 ]heptyl, 2-thia-5-azabicyclo[2.2.1 ]heptyl,7-azabicyclo[2.2.1 ]heptyl, 2,6-diazatricyclo[3.3.1 .13,7]decyl, 2-azatricyclo[3.3.1 .13,7]decyl, 2,4,9- triazatricyclo[3.3.1 .13,7]decyl, 8-azabicyclo[3.2.1 ]octyl, 2,5-diazabicyclo[2.2.2]octyl,
  • This term specifically excludes bicyclic heterocycloalkyl moieties where the rings are joined at a single atom per ring (spiro), as exemplified by the bonding arrangement in a 1 -oxa-2-azaspiro[4.5]dec-2-en-3-yl group.
  • heterocycloalkyls include, but are not limited to, azetidinyl, pyrrolidyl (or pyrrolidinyl), piperidinyl, piperazinyl, morpholinyl, tetrahydro-2H-1 ,4-thiazinyl, tetrahydrofuranyl, dihydrofuranyl, oxazolinyl, thiazolinyl, pyrazolinyl, tetrahydropyranyl, dihydropyranyl, 1 ,3- dioxolanyl, 1 ,3-dioxanyl, 1 ,4-dioxanyl, 1 ,3-oxathiolanyl, 1 ,3-oxathianyl, 1 ,3-dithianyl, azabicylo[3.2.1 ]octyl, azabicylo[3.3.1 ]nonyl, azabicyl
  • heterocycloalkyl groups are 5-membered and/or 6-membered heterocycloalkyl groups, such as pyrrolidyl (or pyrrolidinyl), tetrahydrofuranyl, tetrahydrothienyl, dihydrofuranyl, oxazolinyl, thiazolinyl or pyrazolinyl, piperidyl (or piperidinyl), piperazinyl, morpholinyl, tetrahydropyranyl, dihydropyranyl, 1 ,3-dioxanyl, tetrahydro-2H-1 ,4-thiazinyl, 1 ,4-dioxanyl, 1 ,3-oxathianyl, and 1 ,3-dithianyl.
  • pyrrolidyl or pyrrolidinyl
  • tetrahydrofuranyl tetrahydrothienyl
  • dihydrofuranyl
  • Heteroaryl represents a group or moiety comprising an aromatic monocyclic or bicyclic radical, containing 5 to 10 ring atoms, including 1 to 4 heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur. This term also encompasses bicyclic heterocyclic-aryl compounds containing an aryl ring moiety fused to a heterocycloalkyl ring moiety, containing 5 to 10 ring atoms, including 1 to 4 heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur. This term is also intended to encompass heterocyclic groups containing nitrogen and/or sulfur where the nitrogen or sulfur heteroatoms are optionally oxidized.
  • heteroaryls include, but are not limited to, thienyl, pyrrolyl, imidazolyl, pyrazolyl, furanyl, isothiazolyl, furazanyl, isoxazolyl, oxazolyl, oxadiazolyl, thiazolyl, pyridinyl, pyridinyl-N-oxide, pyrazinyl, pyrimidinyl, pyridazinyl, triazinyl, tetrazinyl, triazolyl, tetrazolyl, benzo[b]thienyl, isobenzofuranyl, 2,3-dihydrobenzofuranyl, chromenyl, chromanyl, indolizinyl, isoindolyl, indolyl, indazolyl, purinyl, isoquinolyl, quinolyl, phthalazinyl, naphthridinyl
  • heteroaryl groups present in the compounds of this invention are 5-6 membered monocyclic heteroaryl groups.
  • Selected 5-membered heteroaryl groups contain one nitrogen, oxygen or sulfur ring heteroatom, and optionally contain 1 , 2 or 3 additional nitrogen ring atoms.
  • Selected 6-membered heteroaryl groups contain 1 , 2, 3 or 4 nitrogen ring heteroatoms.
  • Selected 5- or 6-membered heteroaryl groups include thienyl, pyrrolyl, imidazolyl, pyrazolyl, furanyl, isothiazolyl, furazanyl, isoxazolyl, oxazolyl, oxadiazolyl, thiazolyl, triazolyl, and tetrazolyl or pyridinyl, pyrazinyl, pyrimidinyl, pyridazinyl, triazinyl and thiadiazolyl.
  • heteroaryl groups present in the compounds of this invention are 9-10 membered bicyclic heteroaryl groups.
  • Selected 9-membered heteroaryl groups contain one nitrogen, oxygen or sulfur ring heteroatom, and optionally contain 1 , 2 or 3 additional nitrogen ring atoms.
  • Selected 10-membered heteroaryl groups contain one nitrogen, oxygen or sulfur ring heteroatom, and optionally contain 1 , 2, 3 or 4 additional nitrogen ring atoms.
  • Selected 9-10 membered heteroaryl groups include benzo[b]thienyl, isobenzofuranyl, 2,3-dihydrobenzofuranyl, chromenyl, chromanyl, indolizinyl, isoindolyl, indolyl, indazolyl, purinyl, isoquinolyl, quinolyl, phthalazinyl, naphthridinyl, quinzolinyl, benzothiazolyl, benzimidazolyl, tetrahydroquinolinyl, cinnolinyl, pteridinyl.
  • alkyl, aryl, cycloalkyl, heteroaryl, etc. may be used to define a divalent substituent, such as a group bonded to two other groups. In this instance, such terms are intended to encompass divalent moieties.
  • pentyl is intended to represent a pentylene diradical -wherein the pentyl moiety is any one of a divalent straight (e.g. -CH 2 CH 2 CH 2 CH 2 CH 2 -) or branched (e.g. -CH 2 CH(CH 3 )CH 2 CH 2 -,
  • A is a phenyl group optionally substituted by 1 -2 groups independently selected from the group consisting of Ci -4 alkyl, halogen, cyano, haloCi -4 alkyl, Ci -4 alkoxy, haloC ⁇ alkoxy, -NR'R" and -C ⁇ alkylNR'R", where R' and R" are independently selected from the group consisting of H and Ci -4 alkyl.
  • A is a phenyl group optionally substituted by 1 group selected from the group consisting of methyl, ethyl, fluoro, chloro, trifluoromethyl, methoxy, ethoxy, trifluoromethoxy, cyano, -NR'R" and -Ci -4 alkylN R'R", where R' and R" are independently selected from the group consisting of H and methyl.
  • A is an unsubstituted phenyl group or a phenyl group substituted by an ethyl, fluoro, cyano or methoxy group.
  • A is a cyclopropyl, cyclopentyl or cyclohexyl group, optionally substituted by 1-2 groups independently selected from the group consisting of C 1-4 alkyl, C 1-4 alkoxy, -NR'R" and -C 1-4 alkylNR'R".
  • A is a cyclopropyl, cyclopentyl or cyclohexyl group, optionally substituted by 1-2 groups independently selected from the group consisting of methyl, ethyl, tert-butyl, methoxy, ethoxy, -NR'R" and where each R' and R" are independently selected from the group consisting of H and methyl.
  • A is a cyclopropyl, cyclopentyl or cyclohexyl group.
  • A is naphthyl, optionally substituted by 1-2 groups independently selected from the group consisting of halogen, cyano, haloCi -4 alkyl, Ci -4 alkoxy, halo Ci -4 alkoxy, -NR'R" and
  • A is a 4-7 membered heterocycloalkyl group optionally substituted by 1-3 groups independently selected from the group consisting of halo Ci -4 alkoxy,oxo, - NR'R" and -C ⁇ alkylNR'R".
  • A is a 9-10 membered heteroaryl optionally substituted by 1-2 groups independently selected from the group consisting of C 1-4 alkyl, halogen, cyano, haloC 1-4 alkyl, C 1-4 alkoxy, halo C 1-4 alkoxy, oxo, -NR'R" and - C 1-4 alkylNR'R".
  • A is isoquinolyl, indazolyl,
  • A is a 5-6 membered heteroaryl optionally substituted by 1-2 groups independently selected from the group consisting of halogen, cyano, haloCi -4 alkyl, Ci -4 alkoxy, halo Ci -4 alkoxy, -NR'R" and in still further embodiments, A is a 5-6 membered heteroaryl optionally substituted by 1 group selected from the group consisting of methyl, ethyl, fluoro, trifluoromethyl, -NR'R" and
  • each R' and R" are independently selected from the group consisting of H and methyl and the 5-6 membered heteroaryl contains 1 ring heteroatom selected form N, O and S and optionally contains 1 additional ring nitrogen atom.
  • A is oxazolyl, pyrazolyl, or thienyl optionally substituted by a methyl group.
  • A is pyrazolyl or thienyl, optionally substituted by a methyl group.
  • A is thienyl.
  • A is oxazolyl.
  • A is a pyrimidinyl or pyridinyl group optionally substituted by 1 -2 groups independently selected from the group consisting of halogen, cyano, haloCi -4 alkyl, C- ⁇ alkoxy, halo C- ⁇ alkoxy, -NR'R" and -Ci -4 alkylNR'R".
  • A is a pyrimidinyl, pyridinyl or pyridinyl-N-oxide group optionally substituted by 1 group selected from the group consisting of methyl, ethyl, fluoro, chloro, trifluoromethyl, methoxy, ethoxy, trifluoromethoxy, cyano, -NR'R" and -(C 1-4 alkyl)NR'R", where each R' and R" are independently selected from the group consisting of H and methyl.
  • A is pyridinyl.
  • A is unsubstituted pyrimidinyl.
  • n is 0. In another embodiment, n is 1 . In a further embodiment,
  • n is 2. In still further embodiment, n is 3. In an alternate embodiment, n is 4. In particular embodiments, n is 0 or 1 .
  • m is 0. In another embodiment, m is 1 . In an alternate embodiment, m is 2. In a further embodiment, m is 3. In a still further embodiment, m is 4. In particular embodiments, m is 0 or 1.
  • n is 0 and m is 1 or n is 1 and m is 0.
  • R 1 is H. In another embodiment, R 1 is Ci -3 alkyl. In a further embodiment, R 1 is Ci -2 alkyl. In yet another embodiment, R 1 is methyl.
  • B is a cyclohexyl, phenyl, pyridinyl, pyrimidinyl, imidazolyl, thiazolyl, oxadiazolyl, quinolyl, pyrazinyl, or fluorenyl (particularly, B is a phenyl, pyridinyl, pyrimidinyl, or pyrazinyl), wherein said pyridinyl, pyrimidinyl, or pyrazinyl wherein said cyclohexyl, phenyl, pyridinyl, pyrimidinyl, imidazolyl, thiazolyl, oxadiazolyl, quinolyl, pyrazinyl, or fluorenyl is optionally substituted by 1 or 2 R groups, each independently selected from the group consisting of C 1-4 alkyl, C 1-4 alkylsulfanyl, phenyl, 5-6 membered heteroaryl
  • B is a 4, 5, 6 or 7 membered heterocycloalkyi containing 1 or 2 nitrogen atoms, optionally substituted by 1 or 2 R B groups, each independently selected from the group consisting of Ci -4 alkyl, C-M alkylsulfanyl, phenyl, 5-6 membered heteroaryl, and -NR Ba R Bb (where R Ba and R Bb are independently selected from the group consisting of H, Ci -4 alkyl, phenyl, and 5-6 membered heteroaryl), wherein said phenyl or 5-6 membered heteroaryl of R B , R Ba and R Bb is optionally substituted with a substituent selected from the group consisting of 4-7 membered heterocycloalkyi,
  • heterocycloalkylC 1-4 alkoxy 4-7 membered heterocycloalkyl-carbonyl
  • B is pyrimidinyl, phenyl, pyridinyl, or pyrizinyl, wherein said pyrimidinyl, phenyl, pyridinyl, or pyrizinyl is optionally substituted with 1 or 2 R B groups, each independently selected from the group consisting of Ci -4 alkylsulfanyl, -NH 2 , phenyl, pyrimindinyl, thienyl, thiazolyl, pyrazolyl, and pyridinyl, wherein said phenyl, pyrimindinyl, thiophenyl, thiazolyl, pyrazolyl, or pyridinyl is optionally substituted with morpholinyl, methylpiperazinyl, Ci -4 alkylsulfanyl, cyano, methyoxymethoxypropan-2-yl,
  • B is:
  • D, E, and F is CH and each remaining group is independently selected from the group consisting of CH and N,
  • R 1 is selected from the group consisting of H, methylsulfanyl, and NH 2 ,
  • G and H are each independently selected from the group consisting of CH and N, and R 2 is selected from the group consisting of H, methylsulfanyl, cyano, methyoxymethyoxypropan-2-yl, 1 -hydroxy-1 -methylethyl, morpholinyl, methylpiperazinyl, morpholinylcarbonyl, morpholinylmethyl, morpholinylethyloxy, dimethylaminocarbonyl, and dimethylaminoethoxy.
  • B is phenyl, pyridinyl, pyrazinyl, or pyrimidinyl substituted by 1 ,2-oxazol-3-yl, 1 H-pyrazol-5-yl, 1 ,3-thiazol-2-yl, thiophen-3-yl, wherein said
  • 1 ,2-oxazol-3-yl, 1 H-pyrazol-5-yl, 1 ,3-thiazol-2-yl, thiophen-3-yl is optionally substituted with C 1-4 alkyl or hydroxyC 1-4 alkyl, such as hydroxymethyl.
  • D and E are each independently selected from the group consisting of NH and CH, and n, m and B are as defined herein.
  • the terms "compound(s) of the invention” or “compound(s) of this invention” mean a compound of Formula (I), as defined above, in any form, i.e., any salt or non-salt form (e.g., as a free acid or base form, or as a salt, particularly a
  • any physical form thereof e.g., including non-solid forms (e.g., liquid or semi-solid forms), and solid forms (e.g., amorphous or crystalline forms, specific polymorphic forms, solvate forms, including hydrate forms (e.g., mono-, di- and hemi- hydrates)), and mixtures of various forms.
  • non-solid forms e.g., liquid or semi-solid forms
  • solid forms e.g., amorphous or crystalline forms, specific polymorphic forms, solvate forms, including hydrate forms (e.g., mono-, di- and hemi- hydrates)
  • optionally substituted means unsubstituted groups or rings (e.g., cycloalkyl, heterocycle, and heteroaryl rings) and groups or rings substituted with one or more specified substituents.
  • the compounds according to Formula (I) may contain one or more asymmetric center (also referred to as a chiral center) and may, therefore, exist as individual enantiomers, diastereomers, or other stereoisomeric forms, or as mixtures thereof.
  • Chiral centers such as chiral carbon atoms, may also be present in a substituent such as an alkyl group.
  • the stereochemistry of a chiral center present in Formula (I), or in any chemical structure illustrated herein, is not specified the structure is intended to encompass all individual stereoisomers and all mixtures thereof.
  • compounds according to Formula (I) containing one or more chiral centers may be used as racemic mixtures, scalemic mixtures, or as diaseteromerically or enantiomerically pure materials.
  • diastereoisomeric salts, complexes or other derivatives (2) by selective reaction with a stereoisomer-specific reagent, for example by enzymatic oxidation or reduction; or (3) by gas-liquid or liquid chromatography in a chiral environment, for example, on a chiral support such as silica with a bound chiral ligand or in the presence of a chiral solvent.
  • a stereoisomer-specific reagent for example by enzymatic oxidation or reduction
  • gas-liquid or liquid chromatography in a chiral environment, for example, on a chiral support such as silica with a bound chiral ligand or in the presence of a chiral solvent.
  • specific stereoisomers may be synthesized by asymmetric synthesis using optically active reagents, substrates, catalysts or solvents, or by converting one enantiomer to the other by asymmetric transformation.
  • polymorphism i.e. the capacity to occur in different crystalline forms. These different crystalline forms are typically known as "polymorphs.” It is to be understood that when named or depicted by structure, the disclosed compound, or solvates (particularly, hydrates) thereof, also include all polymorphs thereof. Polymorphs have the same chemical composition but differ in packing, geometrical arrangement, and other descriptive properties of the crystalline solid state. Polymorphs, therefore, may have different physical properties such as shape, density, hardness, deformability, stability, and dissolution properties. Polymorphs typically exhibit different melting points, IR spectra, and X-ray powder diffraction patterns, which may be used for identification. One of ordinary skill in the art will appreciate that different polymorphs may be produced, for example, by changing or adjusting the conditions used in crystallizing/recrystallizing the compound.
  • Formula (I) are preferably pharmaceutically acceptable salts.
  • Suitable pharmaceutically acceptable salts include those described by Berge, Bighley and Monkhouse,
  • salts encompassed within the term “pharmaceutically acceptable salts” refer to non-toxic salts of the compounds of this invention.
  • a salt may be readily prepared by using a desired acid or base as appropriate.
  • the salt may precipitate from solution and be collected by filtration or may be recovered by evaporation of the solvent.
  • a desired salt form may be prepared by any suitable method known in the art, including treatment of the free base with an inorganic acid, such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like, or with an organic acid, such as acetic acid, trifluoroacetic acid, maleic acid, succinic acid, mandelic acid, fumaric acid, malonic acid, pyruvic acid, oxalic acid, glycolic acid, salicylic acid, and the like, or with a pyranosidyl acid, such as glucuronic acid or galacturonic acid, or with an alpha-hydroxy acid, such as citric acid or tartaric acid, or with an amino acid, such as aspartic acid or glutamic acid, or with an aromatic acid, such as benzoic acid or cinnamic acid, or with a sulfonic acid, such as
  • Suitable addition salts are formed from acids which form non-toxic salts and examples include acetate, p-aminobenzoate, ascorbate, aspartate, benzenesulfonate, benzoate, bicarbonate, bismethylenesalicylate, bisulfate, bitartrate, borate, calcium edetate, camsylate, carbonate, clavulanate, citrate, cyclohexylsulfamate, edetate, edisylate, estolate, esylate, ethanedisulfonate, ethanesulfonate, formate, fumarate, gluceptate, gluconate, glutamate, glycollate, glycollylarsanilate, hexylresorcinate, hydrabamine, hydrobromide, hydrochloride, dihydrochloride, hydrofumarate, hydrogen phosphate, hydroiodide, hydromaleate, hydrosuccinate, hydroxyn
  • exemplary acid addition salts include pyrosulfate, sulfite, bisulfite, decanoate, caprylate, acrylate, isobutyrate, caproate, heptanoate, propiolate, oxalate, malonate, suberate, sebacate, butyne-1 ,4-dioate, hexyne-1 ,6-dioate, chlorobenzoate, methylbenzoate, dinitrobenzoate, hydroxybenzoate, methoxybenzoate, phthalate, phenylacetate, phenylpropionate, phenylbutrate, lactate, ⁇ -hydroxybutyrate, mandelate, and sulfonates, such as xylenesulfonate, propanesulfonate, naphthalene-1 -sulfonate and naphthalene-2-sulfonate.
  • an inventive basic compound is isolated as a salt
  • the corresponding free base form of that compound may be prepared by any suitable method known to the art, including treatment of the salt with an inorganic or organic base, suitably an inorganic or organic base having a higher pK a than the free base form of the compound.
  • a desired salt may be prepared by any suitable method known to the art, including treatment of the free acid with an inorganic or organic base, such as an amine (primary, secondary, or tertiary), an alkali metal or alkaline earth metal hydroxide, or the like.
  • an inorganic or organic base such as an amine (primary, secondary, or tertiary), an alkali metal or alkaline earth metal hydroxide, or the like.
  • suitable salts include organic salts derived from amino acids such as glycine and arginine, ammonia, primary, secondary, and tertiary amines, and cyclic amines, such as N-methyl-D-glucamine, diethylamine, isopropylamine, trimethylamine, ethylene diamine, dicyclohexylamine, ethanolamine, piperidine, morpholine, and piperazine, as well as inorganic salts derived from sodium, calcium, potassium, magnesium, manganese, iron, copper, zinc, aluminum, and lithium.
  • amino acids such as glycine and arginine
  • ammonia such as glycine and arginine
  • primary, secondary, and tertiary amines such as N-methyl-D-glucamine, diethylamine, isopropylamine, trimethylamine, ethylene diamine, dicyclohexylamine, ethanolamine, piperidine, morpholine, and piperazine
  • Certain of the compounds of this invention may form salts with one or more equivalents of an acid (if the compound contains a basic moiety) or a base (if the compound contains an acidic moiety).
  • the present invention includes within its scope all possible stoichiometric and non-stoichiometric salt forms.
  • Compounds of the invention having both a basic and acidic moiety may be in the form of zwitterions, acid-addition salt of the basic moiety or base salts of the acidic moiety.
  • This invention also provides for the conversion of one pharmaceutically acceptable salt of a compound of this invention, e.g., a hydrochloride salt, into another
  • a pharmaceutically acceptable salt of a compound of this invention e.g., a sulfate salt.
  • solvates of the compounds of Formula (I), or salts thereof that are in crystalline form may be formed wherein solvent molecules are incorporated into the crystalline lattice during crystallization.
  • Solvates may involve nonaqueous solvents such as ethanol, isopropanol, DMSO, acetic acid, ethanolamine, and ethyl acetate, or they may involve water as the solvent that is incorporated into the crystalline lattice.
  • Solvates wherein water is the solvent that is incorporated into the crystalline lattice are typically referred to as
  • Hydrates include stoichiometric hydrates as well as compositions containing variable amounts of water.
  • the invention includes all such solvates.
  • the compounds of Formula (I) are intended for use in pharmaceutical compositions it will readily be understood that they are each preferably provided in substantially pure form, for example at least 60% pure, more suitably at least 75% pure and preferably at least 85%, especially at least 98% pure (% are on a weight for weight basis). Impure preparations of the compounds may be used for preparing the more pure forms used in the pharmaceutical compositions.
  • the compounds of this invention may be obtained by using synthetic procedures illustrated in the Schemes below or by drawing on the knowledge of a skilled organic chemist.
  • the synthesis provided in these Schemes are applicable for producing compounds of the invention having a variety of different substituent groups employing appropriate precursors, which are suitably protected if needed, to achieve compatibility with the reactions outlined herein. Subsequent deprotection, where needed, affords compounds of the nature generally disclosed. While the Schemes are shown with compounds only of Formula (I), they are illustrative of processes that may be used to make the compounds of the invention.
  • salts particularly pharmaceutically acceptable salts, thereof.
  • ChemDraw 1 1.0 available from CambridgeSoft Corporation, 100 CambridgePark Drive, Cambridge, MA 02140, USA (http://www.cambridgesoft.com).
  • the compounds of Formula (I) can be prepared according to the methods outlined below.
  • the invention also includes various deuterated forms of the compounds of Formula (I). Each available hydrogen atom attached to a carbon atom may be
  • deuterated alkyl groups e.g., /V-(deutero-methyl) amines
  • deuterated alkyl groups e.g., /V-(deutero-methyl) amines
  • the present invention is directed to a method of inhibiting an HDAC which comprises contacting the acetylase with a compound of Formula (I) or a salt thereof, particularly a pharmaceutically acceptable salt thereof. More specifically, this invention is directed to a method of inhibiting HDAC comprising contacting a cell with an effective amount of a compound of Formula (I) or a salt thereof, particularly a pharmaceutically acceptable salt thereof. This invention is also directed to a method of treatment of an HDAC-mediated disease or disorder comprising administering a therapeutically effective amount of the compound of Formula (I) or a salt thereof, particularly a pharmaceutically acceptable salt thereof, to a patient, specifically a human, in need thereof.
  • patient refers to a mammal, specifically, a human.
  • a therapeutically "effective amount” is intended to mean that amount of a compound that, when administered to a patient in need of such treatment, is sufficient to effect treatment, as defined herein.
  • a therapeutically effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt thereof is a quantity of an inventive agent that, when administered to a human in need thereof, is sufficient to inhibit the activity of HDAC such that a disease condition which is mediated by that activity is reduced, alleviated or prevented.
  • the amount of a given compound that will correspond to such an amount will vary depending upon factors such as the particular compound (e.g., the potency (pXC 5 o), efficacy (EC5 0 ), and the biological half-life of the particular compound), disease condition and its severity, the identity (e.g., age, size and weight) of the patient in need of treatment, but can nevertheless be routinely determined by one skilled in the art.
  • duration of treatment and the time period of administration (time period between dosages and the timing of the dosages, e.g., before/with/after meals) of the compound will vary according to the identity of the mammal in need of treatment (e.g., weight), the particular compound and its properties (e.g., pharmaceutical characteristics), disease or condition and its severity and the specific composition and method being used, but can
  • Treating or “treatment” is intended to mean at least the mitigation of a disease condition in a patient, where the disease condition is caused or mediated by HDAC.
  • the methods of treatment for mitigation of a disease condition include the use of the compounds in this invention in any conventionally acceptable manner, for example for prevention, retardation, prophylaxis, therapy or cure of a disease.
  • this invention is directed to a method of treating, ameliorating, or preventing an autoimmune disorder, an immunological disease, an inflammatory disorder, transplant/graft rejection (e.g., allograft), lymphopenia, or graft-versus-host disease (GvHD) in a patient, specifically in a human, comprising administering to the patient a compound of this invention, in an amount sufficient to increase the level and/or activity of a Treg cell or a population of Treg cells in the patient, thereby treating, ameliorating, or preventing the autoimmune disorder, inflammatory disorder,
  • transplant/graft rejection e.g., allograft
  • lymphopenia e.g., allograft
  • GvHD graft-versus-host disease
  • transplant/graft rejection transplant/graft rejection, lymphopenia, or GvHD in the patient.
  • diseases and conditions that may be treated by the compounds of this invention include but not limited to type II diabetes mellitus, coronary artery disease, alopecia, allergies and allergic reactions, and sepsis/toxic shock.
  • Exemplary autoimmune disorders include, but are not limited to, multiple sclerosis, juvenile idiopathic arthritis, psoriatic arthritis, hepatitis C virus-associated mixed cryoglobulinemia, polymyositis, dermatomyositis, polyglandular syndrome type II, autoimmune liver disease, Kawasaki disease, myasthenia gravis, immunodysregulation polyendocrinopathy enteropathy X-linked syndrome (IPEX (syndrome)), type I diabetes, psoriasis, hypothyroidism, hemolytic anemia, autoimmune polyendocrinopathy- candidiasis-ectodermal dystrophy (APECED), thrombocytopenia, spondyloarthritis, Sjogren's syndrome, rheumatoid arthritis, inflammatory bowel disease (IBD), Crohn's disease, ulcerative colitis, eczema, gastritis, or thyroiditis.
  • the inflammatory bowel disease IBD
  • autoimmune diseases include osteoarthritis, systemic sclerosis, sarcoidosis, insulin dependent diabetes mellitus (IDDM, type I diabetes), reactive arthritis, scleroderma, vasculitis, Wegener's granulomatosis, Hashimoto's disease, scleroderma, oophoritis, Lupus (SLE), Grave's disease, asthma, cryoglobulinemia, primary biliary sclerosis, pemphigus vulgaris, hemolytic anemia and pernicious anemia.
  • IDDM insulin dependent diabetes mellitus
  • SLE oophoritis
  • Grave's disease asthma, cryoglobulinemia, primary biliary sclerosis, pemphigus vulgaris, hemolytic anemia and pernicious anemia.
  • transplant/graft rejection e.g., allograft
  • lymphopenia e.g., lymphopenia
  • graft- versus-host disease e.g., graft- versus-host disease
  • transplant/graft rejection e.g., allograft
  • lymphopenia e.g., lymphopenia
  • graft- versus-host disease e.g., graft- versus-host disease
  • transplant/graft rejection e.g., allograft
  • lymphopenia e.g., lymphopenia
  • GvHD graft- versus-host disease
  • this invention is directed to a method of treating an HDAC-mediated neurodegenerative disease or disorder which comprises administering to a patient in need thereof, a compound of Formula I or a salt thereof, particularly a pharmaceutically acceptable salt thereof.
  • This invention is also directed to a method of treatment of a neurodegenerative disease or disorder associated with deacetylases, such as, Alzheimer's disease, Parkinson's disease, neuronal intranuclear inclusion disease (NMD), and polyglutamine disorders, such as Huntington's disease and spinocerebellar ataxia (SCA), comprising administering a therapeutically effective amount of the compound of Formula I or a salt thereof, particularly a pharmaceutically acceptable salt thereof, to a patient, specifically a human, in need thereof.
  • deacetylases such as, Alzheimer's disease, Parkinson's disease, neuronal intranuclear inclusion disease (NMD), and polyglutamine disorders, such as Huntington's disease and spinocerebellar ataxia (SCA)
  • diseases and conditions include but are not limited to cystic fibrosis, osteoporosis, obesity, epilepsy, depression, thalassemia, sickle cell anemia, amyotrophic lateral sclerosis (ALS) and hyperalgesia, cardiac disease (e.g., stroke, hypertension, atherothrombotic diseases, artherosclerosis or limitation of infarct size in acute coronary syndrome), diseases or disorders involving muscular atrophy, gentamicin-induced hearing loss, drug resistance (e.g., drug resistance in osteosarcoma and colon cancer cells), infectious diseases, and immune deficiency/immunocompromised patients.
  • cystic fibrosis e.g., osteoporosis
  • obesity e.g., epilepsy, depression, thalassemia, sickle cell anemia, amyotrophic lateral sclerosis (ALS) and hyperalgesia
  • cardiac disease e.g., stroke, hypertension, atherothrombotic diseases, artherosclerosis or limitation
  • infectious diseases relate to various pathogen infections such as viral, fungal, bacterial, mycoplasm, and infections by unicellular and multicellular eukaryotic organisms.
  • pathogens include but are not limited to HIV, HSV, HPV, Hepatitis A, B and C viruses, influenza, denge, zostrella, rubella, RSV, rotavirus, gram positive, gram negative, streptococcus, tetanus, staphalococcus, tuberculosis, listeria, and malaria.
  • the compounds of the invention may be employed alone or in combination with standard anti-cancer regimens for neoplastic cell, e.g., tumor cell and cancer cell, treatments.
  • this invention is directed to inhibitors of HDAC and their use to stop or reduce the growth of neoplastic cells, e.g., cancer cells and tumor cells.
  • carcinoma e.g., adenocarcinoma
  • heptaocellular carcinoma e.g., sarcoma
  • myeloma e.g., multiple myeloma
  • treating bone disease in multiple myeloma leukemia, childhood acute lymphoblastic leukemia and lymphoma (e.g., cutaneous cell lymphoma)
  • mixed types of cancers such as adenosquamous carcinoma, mixed mesodermal tumor, carcinosarcoma, and teratocarcinoma.
  • breast or prostate cancers or tumors are treated using the HDAC inhibitors of this invention.
  • Other cancers that may be treated using the compounds of this invention include, but are not limited to, bladder cancer, breast cancer, prostate cancer, stomach cancer, lung cancer, colon cancer, rectal cancer, colorectal cancer, liver cancer, endometrial cancer, pancreatic cancer, cervical cancer, ovarian cancer; head and neck cancer, and melanoma.
  • the present invention is further directed to a method of treating a B-cell lymphoma, particularly a B-cell lymphoma associated with deacetylases, which comprises administering to a patient in need thereof, a compound of Formula I or a salt thereof, particularly a pharmaceutically acceptable salt thereof.
  • B-cell lymphomas associated with deacetylases include Burkitt lymphoma, chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL/SLL), diffuse large B-cell lymphoma, follicular lymphoma, immunoblastic large cell lymphoma, precursor B-lymphoblastic lymphoma, mantle cell lymphoma, and
  • this invention is directed to a method of treatment of Burkitt lymphoma, chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL/SLL), diffuse large B-cell lymphoma, follicular lymphoma, immunoblastic large cell lymphoma, precursor B-lymphoblastic lymphoma, mantle cell lymphoma, and Waldenstrom Macroglobulinemia (lymphoplasmacytic lymphoma), comprising administering a therapeutically effective amount of the compound of Formula I or a salt thereof, particularly a pharmaceutically acceptable salt thereof, to a patient, specifically a human, in need thereof.
  • CLL/SLL chronic lymphocytic leukemia/small lymphocytic lymphoma
  • follicular lymphoma immunoblastic large cell lymphoma
  • precursor B-lymphoblastic lymphoma precursor B-lymphoblastic lymphoma
  • mantle cell lymphoma mantle cell lymphoma
  • the compounds of the invention may be administered by any suitable route of administration, including both systemic administration and topical administration.
  • Systemic administration includes oral administration, parenteral administration, transdermal administration, rectal administration, and administration by inhalation.
  • Parenteral administration refers to routes of administration other than enteral
  • transdermal, or by inhalation and is typically by injection or infusion.
  • Parenteral administration includes intravenous, intramuscular, and subcutaneous injection or infusion.
  • Inhalation refers to administration into the patient's lungs whether inhaled through the mouth or through the nasal passages.
  • Topical administration includes application to the skin.
  • the compounds of the invention may be administered once or according to a dosing regimen wherein a number of doses are administered at varying intervals of time for a given period of time. For example, doses may be administered one, two, three, or four times per day. Doses may be administered until the desired therapeutic effect is achieved or indefinitely to maintain the desired therapeutic effect. Suitable dosing regimens for a compound of the invention depend on the pharmacokinetic properties of that compound, such as absorption, distribution, and half-life, which can be determined by the skilled artisan.
  • suitable dosing regimens including the duration such regimens are administered, for a compound of the invention depend on the condition being treated, the severity of the condition being treated, the age and physical condition of the patient being treated, the medical history of the patient to be treated, the nature of concurrent therapy, the desired therapeutic effect, and like factors within the knowledge and expertise of the skilled artisan. It will be further understood by such skilled artisans that suitable dosing regimens may require adjustment given an individual patient's response to the dosing regimen or over time as individual patient needs change.
  • Treatment of HDAC-mediated disease conditions may be achieved using the compounds of this invention as a monotherapy, or in dual or multiple combination therapy, such as in combination with other agents, for example, in combination with one or more of the following agents: DNA methyltransferase inhibitors, acetyl transferase enhancers, proteasome or HSP90 inhibitors, and one or more immunosuppressants that do not activate the T suppressor cells including but are not limited to corticosteroids, rapamycin, Azathioprine, Mycophenolate, Cyclosporine, Mercaptopurine (6-MP), basiliximab, daclizumab, sirolimus, tacrolimus, Muromonab-CD3, cyclophosphamide, and
  • methotrexate which are administered in effective amounts as is known in the art.
  • the compounds of the invention will normally, but not necessarily, be formulated into a pharmaceutical composition prior to administration to a patient. Accordingly, in another aspect the invention is directed to pharmaceutical compositions comprising a compound of the invention and a pharmaceutically-acceptable excipient.
  • compositions of the invention may be prepared and packaged in bulk form wherein an effective amount of a compound of the invention can be extracted and then given to the patient such as with powders, syrups, and solutions for injection.
  • the pharmaceutical compositions of the invention may be prepared and packaged in unit dosage form.
  • a dose of the pharmaceutical composition contains at least a therapeutically effective amount of a compound of this invention (i.e., a compound of Formula (I) or a salt, particularly a pharmaceutically acceptable salt, thereof).
  • the pharmaceutical compositions may contain from 1 mg to 1000 mg of a compound of this invention.
  • compositions of the invention typically contain one compound of the invention. However, in certain embodiments, the pharmaceutical compositions of the invention contain more than one compound of the invention. In addition, the pharmaceutical compositions of the invention may optionally further comprise one or more additional pharmaceutically active compounds.
  • pharmaceutically-acceptable excipient means a material, composition or vehicle involved in giving form or consistency to the composition.
  • Each excipient must be compatible with the other ingredients of the pharmaceutical composition when commingled such that interactions which would substantially reduce the efficacy of the compound of the invention when administered to a patient and interactions which would result in pharmaceutical compositions that are not pharmaceutically-acceptable are avoided.
  • each excipient must of course be of sufficiently high purity to render it pharmaceutically-acceptable.
  • the compounds of the invention and the pharmaceutically-acceptable excipient or excipients will typically be formulated into a dosage form adapted for administration to the patient by the desired route of administration.
  • Conventional dosage forms include those adapted for (1 ) oral administration such as tablets, capsules, caplets, pills, troches, powders, syrups, elixirs, suspensions, solutions, emulsions, sachets, and cachets; (2) parenteral administration such as sterile solutions, suspensions, and powders for reconstitution; (3) transdermal administration such as transdermal patches; (4) rectal administration such as suppositories; (5) inhalation such as aerosols and solutions; and (6) topical administration such as creams, ointments, lotions, solutions, pastes, sprays, foams, and gels.
  • Suitable pharmaceutically-acceptable excipients will vary depending upon the particular dosage form chosen.
  • suitable pharmaceutically-acceptable excipients may be chosen for a particular function that they may serve in the composition.
  • certain pharmaceutically-acceptable excipients may be chosen for their ability to facilitate the production of uniform dosage forms.
  • Certain pharmaceutically- acceptable excipients may be chosen for their ability to facilitate the production of stable dosage forms.
  • Certain pharmaceutically-acceptable excipients may be chosen for their ability to facilitate the carrying or transporting the compound or compounds of the invention once administered to the patient from one organ, or portion of the body, to another organ, or portion of the body.
  • Certain pharmaceutically-acceptable excipients may be chosen for their ability to enhance patient compliance.
  • Suitable pharmaceutically-acceptable excipients include the following types of excipients: diluents, fillers, binders, disintegrants, lubricants, glidants, granulating agents, coating agents, wetting agents, solvents, co-solvents, suspending agents, emulsifiers, sweeteners, flavoring agents, flavor masking agents, coloring agents, anti-caking agents, humectants, chelating agents, plasticizers, viscosity increasing agents, antioxidants, preservatives, stabilizers, surfactants, and buffering agents.
  • excipients include the following types of excipients: diluents, fillers, binders, disintegrants, lubricants, glidants, granulating agents, coating agents, wetting agents, solvents, co-solvents, suspending agents, emulsifiers, sweeteners, flavoring agents, flavor masking agents, coloring agents, anti-caking agents, humectants,
  • Skilled artisans possess the knowledge and skill in the art to enable them to select suitable pharmaceutically-acceptable excipients in appropriate amounts for use in the invention.
  • resources that are available to the skilled artisan which describe pharmaceutically-acceptable excipients and may be useful in selecting suitable pharmaceutically-acceptable excipients. Examples include Remington's Pharmaceutical Sciences (Mack Publishing Company), The Handbook of Pharmaceutical Additives (Gower Publishing Limited), and The Handbook of Pharmaceutical Excipients (the American Pharmaceutical Association and the Pharmaceutical Press).
  • compositions of the invention are prepared using techniques and methods known to those skilled in the art. Some of the methods commonly used in the art are described in Remington's Pharmaceutical Sciences (Mack Publishing
  • the invention is directed to a solid oral dosage form such as a tablet or capsule comprising an effective amount of a compound of the invention and a diluent or filler.
  • Suitable diluents and fillers include lactose, sucrose, dextrose, mannitol, sorbitol, starch (e.g. corn starch, potato starch, and pre-gelatinized starch), cellulose and its derivatives (e.g. microcrystalline cellulose), calcium sulfate, and dibasic calcium phosphate.
  • the oral solid dosage form may further comprise a binder. Suitable binders include starch (e.g.
  • the oral solid dosage form may further comprise a disintegrant. Suitable disintegrants include crospovidone, sodium starch glycolate, croscarmelose, alginic acid, and sodium carboxymethyl cellulose.
  • the oral solid dosage form may further comprise a lubricant. Suitable lubricants include stearic acid, magnesium stearate, calcium stearate, and talc.
  • Capsules are prepared using conventional methods and are formulated as follows:
  • ferf-Butyl 4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)benzylcarbamate To a suspension of ferf-butyl 4-cyanobenzylcarbamate (1.32 g, 5.70 mmol), / ' -Pr 2 NEt (2.99 mL, 17.10 mmol) in ethanol (26 mL) was added hydroxylamine hydrochloride (380 mg, 1 1.4 mmol). The reaction mixture was stirred at 90 °C for 4 h. The solvents were removed under reduced pressure, and the residue was dissolved in THF (26.0 mL).
  • TFAA (2.38 mL, 17.1 mmol) was added carefully to the reaction under stirring followed by solid DMAP (139 mg, 1.140 mmol). The reaction mixture was stirred at rt for 3 h. After concentrating under reduced pressure, the reaction mixture was dissolved in DCM and quenched with satd. NH 4 CI. The organic layer was separated, dried over MgS0 4 , filtered, and
  • HDAC9 Histone Deacetylase 9
  • Novel histone deacetylase 9 (HDAC9) inhibitors were characterized in an in vitro biochemical functional assay.
  • the assay measures the increased fluorescent signal due to deacetylation, by HDAC9, of a fluorogenic substrate.
  • the commercial available substrate is Class lla HDAC-specific and contains an acetylated lysine residue and would releases the fluorescent signal upon trypsin cleavage after deacetylation.
  • test compounds diluted to various concentrations in 100% DMSO are first dispensed into 384-well assay plates.
  • Recombinant HDAC9 isoform 4 (purchased from BPS Bioscience) in complete assay buffer (50 mM Tris-HCI, pH 8.0, 137 mM NaCI, 2.7 mM KCI, 1 mM MgCI 2 , 0.05% BSA & 0.005% Tween 20) were then added to each well (5uL/well) using Multidrop Combi (Thermo Scientific), followed by 5 uL/well substrate (purchased from BPS Bioscience, 4.5 uM final).
  • HDAC7 targeting enhances FOXP3+ Treg function and induces long-term allograft survival L. Wang, et al., Am. J. Transplant 9, S621 (2009).
  • HDAC-MEF2 complexes A. Nebbioso, F. Manzo, M. Miceli, M. Conte, L. Manente, A. Baldi, A. De Luca, D. Rotili, S. Valente, A. Mai, A. Usiello, H. Gronemeyer, L. Altucci, EMBO reports 10 (7), 776-782, 2009, and references therein.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)

Abstract

Disclosed are compounds having the formula (I) and salts thereof wherein A, B, m, and n are as defined herein, and methods of making and using the same.

Description

COMPOUNDS AND METHODS
BACKGROUND OF THE INVENTION
Field of the Invention
The present invention relates to compounds that inhibit histone deacetylase (HDAC) enzymes, the preparation of these compounds, the use of these compounds in the treatment of diseases or conditions ameliorated by inhibition of HDAC activity and pharmaceutical compositions comprising these compounds.
Background of the Invention
Chromatin organization involves DNA wound around histone octamers that form nucleosomes. Core histones with N-terminal tails extending from compact nucleosomal core particles can be acetylated or deacetylated at epsilon lysine residues affecting histone-DNA and histone-non-histone protein interactions. Histone deacetylases (HDACs) catalyze the deacetylation of histone and non-histone proteins and play an important role in epigenetic regulation. There are currently 18 known HDACs that are organized into three classes: class I HDACs (HDAC1 , HDAC2, HDAC3, HDAC8 and
HDAC1 1 ) are mainly localized to the nucleus; class II HDACs (HDAC4, HDAC5, HDAC6, HDAC7, HDAC9 and HDAC10), which shuttle between the nucleus and the cytoplasm; and class III HDACs (SIRT1-7), whose cellular localization includes various organelles.
Class II HDACs are further characterized as class lla HDACs and class lib HDACs.
HDAC9 is a class lla histone deacetylase highly expressed in human Tregs. HDAC9 deficiency: 1 ) increases Foxp3 expression (and other Treg markers), 2) increases Foxp3 and histone 3 acetylation, 3) increases Foxp3 DNA binding, 4) increases Treg numbers, 5) increases suppressive activity in vitro and in vivo, and 6) ameliorates murine colitis. Tregs which are deficient in HDAC9 induce permanent tolerance of fully mismatched cardiac allografts. In addition, HDAC9 inhibitors maybe useful for treatment of diseases and disorders associated with abnormal cell proliferation, differentiation and survival, e.g. breast and prostate tumors.
Preliminary data shows that targeting HDAC7, a class lla histone deacetylase, enhances Treg suppression in vitro and in vivo. HDAC7 enhances FOXP3+ Treg function and induces long-term allograft survival.
Inhibition of HDAC6, a class lib HDAC, has been shown to increase Treg suppressive function in vitro along with increased expression of FOXP3 protein and Treg associated genes including CTLA, IL-10, TNR18. HDAC6 inhibition in vivo decreased severity of colitis in the dextran sodium sulphate-induced colitis model and the
CD4+CD62Lhigh adoptive transfer model of colitis. In addition, inhibition of HDAC6 with a subtherapeutic dose of rapamycin led to prolonged cardiac allograft survival.
Thus, an orally available small molecule selective inhibitor of Class II HDAC activity (more specifically HDAC9 or HDAC7 or HDAC6) is expected to modulate autoimmune diseases through expansion and enhancement of Treg activity.
Inhibition of other Class II HDAC's for example HDAC4 and 5 impair myogenesis by modulating the stability and activity of HDAC-MEF2 complexes and maybe potentially useful for the treatment of muscle and heart diseases including cardiac hypertrophy and heart failure. Also, inhibition of Class II HDAC activity represents a novel approach for disrupting or intervening in cell cycle regulation.
HDAC9 is also highly expressed in human B cells. Relative to normal B cells, expression of HDAC9 is deregulated in cell lines derived from B cell tumours and HDAC9 is highly overexpressed in cells derived from patients with non-Hodgkin's lymphoma (http://icr.ac.uk research/team_leaders/Zelent_Arthur/Zelent_Arthur_RI/index.shtml). HDAC4 and HDAC9 have both been reported to be overexpressed in CD19+ cells from patients with Waldenstrom Macroglobulinemia (Sun et al., Clinical Lymphoma, Myeloma & Leukemia, 201 1 , p. 152).
Class Ma HDACs (HDAC4, HDAC5, HDAC7 and HDAC9) have been reported to associate with Bcl-6, a transcription factor implicated in the pathogenesis of B-cell malignancies (Lemercier et al, Journal of Biological Chemistry, 2002, p. 22045, and Petrie et al, Journal of Biological Chemistry, 2003, p. 16059). Due to these interactions class lla HDACs have been suggested to modulate the transcriptional repression of BCL6 and participate in its role in B-cell activation and differentiation, inflammation, and cell-cycle regulation (Verdin et al. TRENDS in Genetics, 2003, p. 286).
HDAC6, a class lib HDAC, has been reported to play an important role in aggresomal protein degradation, making it a target for the treatment of B cell
malignancies (Simms-Waldrip et al., Molecular Genetics and Metabolism, 2008, p. 283) Accordingly, a small molecule selective inhibitor of HDAC4, HDAC5, HDAC6, HDAC7, HDAC8 and/or HDAC9 is expected to be beneficial in the treatment of B-cell malignancies by targeting one or several of the above enzymes.
HDAC4, HDAC5 and HDAC9 are also highly expressed in the brain. HDAC4 has been linked to a variety of neurodegenerative disorders: it is a downstream target of Parkin (associating it to Parkinson's disease), it is a major component of intranuclear inclusions produced in NIIND. HDAC4 also contains a conserved glutamine rich domain, such domain has been observed to increase susceptibility to amyloid formation
associated with Alzheimer's disease (Majdzadeh et al. Front. Biosci., 2009, p. 1072). Heterozygotes of HDAC4 knockouts crossed to R6/2 mice (Huntington's disease model) led to improved motor/behavior and reduced aggregation
(http://bmi.epfl.ch/files/content/sites/bmi/files/shared/Abstract_Gillian_Bates.pdf). HDAC4 and HDAC5 localization are regulated by neuronal activity, and HDAC5 nuclear import is increased in diseased neurons of Huntington's disease patients.
HDAC7 has been implicated in regulating ataxin-7 turnover in a SCA-7 model (Mookerjee S et al., J Neurosci., 2009, p. 15134).
HDAC6 is expressed in most neurons and most abundantly in cerebellar Purkinje cells, the degeneration of this type of neurons is observed in patients with spinocerebellar ataxia type 1 (SCA1 ) or SCA7. HDAC6 is involved in regulating microtubule dynamics and protein degradation and a defect in microtubule-based transport may contribute to the neuronal toxicity observed in Huntington's disease (Kazantsev et al. Nature Reviews Drug Discovery, 2008, p. 854). Additionally, HDAC6 activity has been shown to be required for autophagic degradation of aggregated huntingtin, suggesting a role in protecting cells from polyQ toxicity (Iwata, et al., J. Biol. Chem., 2005, p. 40282).
Accordingly, a small molecule selective inhibitor of HDAC activity (more
specifically HDAC4 or HDAC5 or HDAC6 or HDAC7or HDAC9) is expected to be beneficial in the treatment of neurodegenerative diseases.
Class II HDAC inhibitors have therapeutic potential in the study and/or treatment of the various diseases or conditions described herein.
Many of the known small-molecule HDAC inhibitors, however, inhibit all HDAC isoforms. It would be advantageous to identify HDAC inhibitors that inhibited one or more, but not all HDAC isoforms.
SUMMARY OF THE INVENTION
The invention is directed to a compound according to Formula (I):
Figure imgf000004_0001
and salts, particularly pharmaceutically-acceptable salts thereof wherein: A is C3-6cycloalkyl (C3-6cycloalkylenyl), 4-7 membered heterocycloalkyl (4-7 membered heterocycloalkylenyl), phenyl (phenylenyl), naphthyl (naphthylenyl), 5-6 membered heteroaryl ( 5-6 membered heteroarylenyl), or 9-10 membered heteroaryl (9-10 membered heteroarylenyl), wherein said C3-6cycloalkyl, 4-7 membered
heterocycloalkyl, phenyl, naphthyl, 5-6 membered heteroaryl, or 9-10 membered heteroaryl is optionally substituted by 1-3 groups each independently selected from the group consisting of Ci-4alkyl, aryl, arylCi-4alkyl-, halogen, cyano, haloCi-4alkyl, d^alkoxy, haloCi-4alkoxy, -NR'R" and -Ci-4alkylNR'R", where R' and R" are independently selected from the group consisting of H and
Figure imgf000005_0001
n is 0, 1 , 2, 3 or 4;
R1 is H or C1-4alkyl;
m is 0, 1 , 2, 3 or 4; and
B is C3-6cycloalkyl, 4-7 membered heterocycloalkyl, phenyl, naphthyl, 5-6 membered heteroaryl, or 9-10 membered heteroaryl,
wherein said C3-6cycloalkyl, 4-7 membered heterocycloalkyl, phenyl, naphthyl, 5-6 membered heteroaryl, or 9-10 membered heteroaryl is optionally substituted by 1 , 2 or 3 RB groups each independently selected from the group consisting of
Figure imgf000005_0002
Ci-4alkoxy, aryl, arylC1-4alkyl-, 4-7 membered heterocycloalkyl, 4-7 membered
heterocycloalkylC1-4alkyl-, 5-6 membered heteroaryl, 5-6 membered heteroarylC1-4alkyl-, 9-10 membered heteroaryl, 9-10 membered heteroarylC1-4alkyl-, halogen, haloC1-4alkyl, haloC1-4alkoxy, hydroxyC1-4alkyl, C1-4alkylsulfanyl, (C1-4alkyl)(C1-4alkyl)N-C2-C4alkoxy, and -C0-4alkyl-NRBaRBb, wherein RBa and RBb are independently selected from the group consisting of H, Ci-4alkyl, aryl, 4-7 membered heterocycloalkyl, 5-6 membered heteroaryl, arylCi-4alkyl-, 5-6 membered heteroarylCi-4alkyl-, and 4-7 membered
heterocycloalkylCi-4alkyl-,
wherein any of said aryl (that is the aryl group or the aryl moiety of the
arylCi-4alkyl- group of RB, RBa, and R Bb), heterocycloalkyl (that is the 4-7 membered heterocycloalkyl group or the heterocycloalkyl moiety of the 4-7 membered
heterocycloalkylCi-4alkyl- group of RB, RBa, and R Bb) or heteroaryl (that is the 5-6 or 9-10 membered heteroaryl group or the heteroaryl moiety of the 5-6 or 9-10 membered heteroarylCi-4alkyl- group of RB, RBa, and R Bb) is optionally substituted by a substituent selected from the group consisting of 4-7 membered heterocycloalkyl,
Figure imgf000005_0003
Ci-4alkyl, halogen, haloCi-4alkyl, Ci-4alkoxy, Ci-4 alkylsulfanyl, haloCi-4alkoxy, cyano, Ci-4alkoxyC2-4alkoxyCi-4 alkyl, aminoC2-6alkoxy, Ci-6alkylaminoC2-C6alkoxy,
(Ci-6alkyl)(Ci-6alkyl)aminoC2-6alkoxy, hydroxyl,
Figure imgf000005_0004
4-7 membered heterocycloalkylC1-4alkyl, 4-7 membered heterocycloalkylC1-4alkoxy, 4-7 membered heterocycloalkyl-carbonyl-, -C0-4alkyl-NR'R" and -C(0)(CH2)pNR'R",
wherein: any heterocycloalkyl (that is, the 4-7 membered heterocycloalkyl group or the heterocycloalkyl moiety of the
Figure imgf000006_0001
heterocycloalkylCi-4alkoxy, or heterocycloalkyl-carbonyl groups) is optionally substituted by 1 , 2 or 3 groups each independently selected from the group consisting of Ci-4alkyl and Ci-4alkyl-CO-, R' and R" are independently selected from the group consisting of H and Ci-4alkyl, and p is 0-4.
Pharmaceutical compositions comprising compounds of the invention and a pharmaceutical carrier are also provided.
The invention further encompasses a method of treating a subject having a disease or disorder mediated by inhibition of a HDAC comprising administering the compound of Formula (I), or a salt thereof.
DETAILED DESCRIPTION OF THE INVENTION
The alternative definitions for the various groups and substituent groups of
Formula (I) provided throughout the specification are intended to particularly describe each compound species disclosed herein, individually, as well as groups of one or more compound species. The scope of this invention includes any combination of these group and substituent group definitions.
As used herein, the term "alkyl" represents a saturated, straight, or branched hydrocarbon moiety. Exemplary alkyls include, but are not limited to methyl (Me), ethyl (Et), n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, f-butyl, n-pentyl, /'so-pentyl (3-methyl- butyl), neo-pentyl (2,2-dimethylpropyl), etc. The term "Cr4alkyl" refers to an alkyl containing from 1 to 4 carbon atoms. Any substituent term containing a "C0 alkyl" moiety indicates that the substituent term excludes the presence of an alkyl moiety in that substituent. For example, the term "-C0-4alkyl-NRBaRBb" is intended to describe both "- NRBaRBb" and "-Ci.4alkyl-NRBaRBb" substituents.
When the term "alkyl" is used in combination with other substituent groups, such as "haloalkyl" or "cycloalkyl-alkyl" or "arylalkyl", the term "alkyl" is intended to encompass a divalent straight or branched-chain hydrocarbon radical. For example, "arylalkyl" is intended to mean the radical -alkylaryl, wherein the alkyl moiety thereof is a divalent straight or branched-chain carbon radical and the aryl moiety thereof is as defined herein, and is represented by the bonding arrangement present in a benzyl group (-CH2-phenyl).
As used herein, the term "cycloalkyl" refers to a non-aromatic, saturated, cyclic hydrocarbon ring. The term "C3-8cycloalkyl" refers to a non-aromatic cyclic hydrocarbon ring having from three to eight ring carbon atoms. Exemplary "C3-8cycloalkyl" groups useful in the present invention include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl.
"Alkoxy" refers to a group containing an alkyl radical attached through an oxygen linking atom. The term "C1-4alkoxy" refers to a straight- or branched-chain hydrocarbon radical having at least 1 and up to 4 carbon atoms attached through an oxygen linking atom. Exemplary "(Ci-4)alkoxy" groups useful in the present invention include, but are not limited to, methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, s-butoxy, and f-butoxy.
"Aryl" represents a group or moiety comprising an aromatic, monocyclic or bicyclic hydrocarbon radical containing from 6 to 10 carbon ring atoms and to which may be fused one or more cycloalkyl rings.
"Heterocycloalkyl" represents a group or moiety comprising a stable, non- aromatic, monocyclic or bicyclic radical, which is saturated or partially unsaturated, containing 3 to 10 ring atoms, which includes 1 to 4 heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur. The heterocycloalkyl may be attached by any atom of the monocyclic or bicyclic radical which results in the creation of a stable structure. This term encompasses bicyclic heterocycloalkyl moieties where the rings are joined at two atoms per ring, as exemplified by the bonding arrangement in 2,5- diazabicyclo[2.2.1 ]heptyl, 2-azabicyclo[2.2.1 ]heptyl, 2-oxa-5-azabicyclo[2.2.1 ]heptyl, 7- oxa-2-azabicyclo[2.2.1 ]heptyl, 2-thia-5-azabicyclo[2.2.1 ]heptyl,7-azabicyclo[2.2.1 ]heptyl, 2,6-diazatricyclo[3.3.1 .13,7]decyl, 2-azatricyclo[3.3.1 .13,7]decyl, 2,4,9- triazatricyclo[3.3.1 .13,7]decyl, 8-azabicyclo[3.2.1 ]octyl, 2,5-diazabicyclo[2.2.2]octyl, 2- azabicyclo[2.2.2]octyl, 3-azabicyclo[3.2.1 ]octyl, 8-azabicyclo[3.2.1 ]octyl, octahydro-1 H- pyrrolo[3,2-6]pyridinyl group. This term specifically excludes bicyclic heterocycloalkyl moieties where the rings are joined at a single atom per ring (spiro), as exemplified by the bonding arrangement in a 1 -oxa-2-azaspiro[4.5]dec-2-en-3-yl group. Illustrative examples of heterocycloalkyls include, but are not limited to, azetidinyl, pyrrolidyl (or pyrrolidinyl), piperidinyl, piperazinyl, morpholinyl, tetrahydro-2H-1 ,4-thiazinyl, tetrahydrofuranyl, dihydrofuranyl, oxazolinyl, thiazolinyl, pyrazolinyl, tetrahydropyranyl, dihydropyranyl, 1 ,3- dioxolanyl, 1 ,3-dioxanyl, 1 ,4-dioxanyl, 1 ,3-oxathiolanyl, 1 ,3-oxathianyl, 1 ,3-dithianyl, azabicylo[3.2.1 ]octyl, azabicylo[3.3.1 ]nonyl, azabicylo[4.3.0]nonyl, oxabicylo[2.2.1 ]heptyl and 1 ,5,9-triazacyclododecyl.
Generally, in the compounds of this invention, heterocycloalkyl groups are 5-membered and/or 6-membered heterocycloalkyl groups, such as pyrrolidyl (or pyrrolidinyl), tetrahydrofuranyl, tetrahydrothienyl, dihydrofuranyl, oxazolinyl, thiazolinyl or pyrazolinyl, piperidyl (or piperidinyl), piperazinyl, morpholinyl, tetrahydropyranyl, dihydropyranyl, 1 ,3-dioxanyl, tetrahydro-2H-1 ,4-thiazinyl, 1 ,4-dioxanyl, 1 ,3-oxathianyl, and 1 ,3-dithianyl.
"Heteroaryl" represents a group or moiety comprising an aromatic monocyclic or bicyclic radical, containing 5 to 10 ring atoms, including 1 to 4 heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur. This term also encompasses bicyclic heterocyclic-aryl compounds containing an aryl ring moiety fused to a heterocycloalkyl ring moiety, containing 5 to 10 ring atoms, including 1 to 4 heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur. This term is also intended to encompass heterocyclic groups containing nitrogen and/or sulfur where the nitrogen or sulfur heteroatoms are optionally oxidized. Illustrative examples of heteroaryls include, but are not limited to, thienyl, pyrrolyl, imidazolyl, pyrazolyl, furanyl, isothiazolyl, furazanyl, isoxazolyl, oxazolyl, oxadiazolyl, thiazolyl, pyridinyl, pyridinyl-N-oxide, pyrazinyl, pyrimidinyl, pyridazinyl, triazinyl, tetrazinyl, triazolyl, tetrazolyl, benzo[b]thienyl, isobenzofuranyl, 2,3-dihydrobenzofuranyl, chromenyl, chromanyl, indolizinyl, isoindolyl, indolyl, indazolyl, purinyl, isoquinolyl, quinolyl, phthalazinyl, naphthridinyl, quinzolinyl, benzothiazolyl, benzimidazolyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, isoindolinyl, indolinyl, cinnolinyl, pteridinyl, isothiazolyl.
Some of the heteroaryl groups present in the compounds of this invention are 5-6 membered monocyclic heteroaryl groups. Selected 5-membered heteroaryl groups contain one nitrogen, oxygen or sulfur ring heteroatom, and optionally contain 1 , 2 or 3 additional nitrogen ring atoms. Selected 6-membered heteroaryl groups contain 1 , 2, 3 or 4 nitrogen ring heteroatoms. Selected 5- or 6-membered heteroaryl groups include thienyl, pyrrolyl, imidazolyl, pyrazolyl, furanyl, isothiazolyl, furazanyl, isoxazolyl, oxazolyl, oxadiazolyl, thiazolyl, triazolyl, and tetrazolyl or pyridinyl, pyrazinyl, pyrimidinyl, pyridazinyl, triazinyl and thiadiazolyl.
Some of the heteroaryl groups present in the compounds of this invention are 9-10 membered bicyclic heteroaryl groups. Selected 9-membered heteroaryl groups contain one nitrogen, oxygen or sulfur ring heteroatom, and optionally contain 1 , 2 or 3 additional nitrogen ring atoms. Selected 10-membered heteroaryl groups contain one nitrogen, oxygen or sulfur ring heteroatom, and optionally contain 1 , 2, 3 or 4 additional nitrogen ring atoms. Selected 9-10 membered heteroaryl groups include benzo[b]thienyl, isobenzofuranyl, 2,3-dihydrobenzofuranyl, chromenyl, chromanyl, indolizinyl, isoindolyl, indolyl, indazolyl, purinyl, isoquinolyl, quinolyl, phthalazinyl, naphthridinyl, quinzolinyl, benzothiazolyl, benzimidazolyl, tetrahydroquinolinyl, cinnolinyl, pteridinyl.
In addition, the terms alkyl, aryl, cycloalkyl, heteroaryl, etc. may be used to define a divalent substituent, such as a group bonded to two other groups. In this instance, such terms are intended to encompass divalent moieties. For example, "pentyl" is intended to represent a pentylene diradical -wherein the pentyl moiety is any one of a divalent straight (e.g. -CH2CH2CH2CH2CH2-) or branched (e.g. -CH2CH(CH3)CH2CH2-,
-CH2CH2CH(CH2CH3)-, -CH2CH2C(CH3)2-) chain 5-carbon radical.
The terms "halogen" and "halo" represent chloro, fluoro, bromo or iodo
substituents.
The compounds of the invention are only those which are contemplated to be "chemically stable" as will be appreciated by those skilled in the art.
In one embodiment, A is a phenyl group optionally substituted by 1 -2 groups independently selected from the group consisting of Ci-4alkyl, halogen, cyano, haloCi-4alkyl, Ci-4alkoxy, haloC^alkoxy, -NR'R" and -C^alkylNR'R", where R' and R" are independently selected from the group consisting of H and Ci-4alkyl. In further embodiments, A is a phenyl group optionally substituted by 1 group selected from the group consisting of methyl, ethyl, fluoro, chloro, trifluoromethyl, methoxy, ethoxy, trifluoromethoxy, cyano, -NR'R" and -Ci-4alkylN R'R", where R' and R" are independently selected from the group consisting of H and methyl. In specific embodiments, A is an unsubstituted phenyl group or a phenyl group substituted by an ethyl, fluoro, cyano or methoxy group.
In yet another embodiment, A is a cyclopropyl, cyclopentyl or cyclohexyl group, optionally substituted by 1-2 groups independently selected from the group consisting of C1-4alkyl, C1-4alkoxy, -NR'R" and -C1-4alkylNR'R". In further embodiments, A is a cyclopropyl, cyclopentyl or cyclohexyl group, optionally substituted by 1-2 groups independently selected from the group consisting of methyl, ethyl, tert-butyl, methoxy, ethoxy, -NR'R" and
Figure imgf000009_0001
where each R' and R" are independently selected from the group consisting of H and methyl. In selected embodiments of this invention, A is a cyclopropyl, cyclopentyl or cyclohexyl group.
In another embodiment of this invention, A is naphthyl, optionally substituted by 1-2 groups independently selected from the group consisting of
Figure imgf000009_0002
halogen, cyano, haloCi-4alkyl, Ci-4alkoxy, halo Ci-4alkoxy, -NR'R" and
Figure imgf000009_0003
In another embodiment of this invention, A is a 4-7 membered heterocycloalkyl group optionally substituted by 1-3 groups independently selected from the group consisting of
Figure imgf000009_0004
halo Ci-4alkoxy,oxo, - NR'R" and -C^alkylNR'R".
In another embodiment of this invention, A is a 9-10 membered heteroaryl optionally substituted by 1-2 groups independently selected from the group consisting of C1-4alkyl, halogen, cyano, haloC1-4alkyl, C1-4alkoxy, halo C1-4alkoxy, oxo, -NR'R" and - C1-4alkylNR'R". In selected embodiments, A is isoquinolyl, indazolyl,
tetrahydroisoquinolinonyl, isoindolinonyl, benzofuranyl, benzothienyl or indolinyl.
In further embodiments, A is a 5-6 membered heteroaryl optionally substituted by 1-2 groups independently selected from the group consisting of
Figure imgf000010_0001
halogen, cyano, haloCi-4alkyl, Ci-4alkoxy, halo Ci-4alkoxy, -NR'R" and
Figure imgf000010_0002
In still further embodiments, A is a 5-6 membered heteroaryl optionally substituted by 1 group selected from the group consisting of methyl, ethyl, fluoro, trifluoromethyl, -NR'R" and
Figure imgf000010_0003
where each R' and R" are independently selected from the group consisting of H and methyl and the 5-6 membered heteroaryl contains 1 ring heteroatom selected form N, O and S and optionally contains 1 additional ring nitrogen atom. In selected embodiments, A is oxazolyl, pyrazolyl, or thienyl optionally substituted by a methyl group. In other selected embodiments, A is pyrazolyl or thienyl, optionally substituted by a methyl group. In specific embodiments, A is thienyl. In other specific embodiments, A is oxazolyl.
In yet other embodiments, A is a pyrimidinyl or pyridinyl group optionally substituted by 1 -2 groups independently selected from the group consisting of
Figure imgf000010_0004
halogen, cyano, haloCi-4alkyl, C-^alkoxy, halo C-^alkoxy, -NR'R" and -Ci-4alkylNR'R". In further embodiments, A is a pyrimidinyl, pyridinyl or pyridinyl-N-oxide group optionally substituted by 1 group selected from the group consisting of methyl, ethyl, fluoro, chloro, trifluoromethyl, methoxy, ethoxy, trifluoromethoxy, cyano, -NR'R" and -(C1-4alkyl)NR'R", where each R' and R" are independently selected from the group consisting of H and methyl. In specific embodiments, A is pyridinyl. In particular embodiments, A is unsubstituted pyrimidinyl.
In one embodiment, n is 0. In another embodiment, n is 1 . In a further
embodiment, n is 2. In still further embodiment, n is 3. In an alternate embodiment, n is 4. In particular embodiments, n is 0 or 1 .
In one embodiment, m is 0. In another embodiment, m is 1 . In an alternate embodiment, m is 2. In a further embodiment, m is 3. In a still further embodiment, m is 4. In particular embodiments, m is 0 or 1.
In particular embodiments, n is 0 and m is 1 or n is 1 and m is 0.
In one embodiment, R1 is H. In another embodiment, R1 is Ci-3alkyl. In a further embodiment, R1 is Ci-2alkyl. In yet another embodiment, R1 is methyl.
In an embodiment of this invention, B is a cyclohexyl, phenyl, pyridinyl, pyrimidinyl, imidazolyl, thiazolyl, oxadiazolyl, quinolyl, pyrazinyl, or fluorenyl (particularly, B is a phenyl, pyridinyl, pyrimidinyl, or pyrazinyl), wherein said pyridinyl, pyrimidinyl, or pyrazinyl wherein said cyclohexyl, phenyl, pyridinyl, pyrimidinyl, imidazolyl, thiazolyl, oxadiazolyl, quinolyl, pyrazinyl, or fluorenyl is optionally substituted by 1 or 2 R groups, each independently selected from the group consisting of C1-4alkyl, C1-4alkylsulfanyl, phenyl, 5-6 membered heteroaryl, and -N RBaRBb (where RBa and RBb are independently selected from the group consisting of H, Ci-4alkyl, phenyl, and 5-6 membered heteroaryl), wherein any of said phenyl or 5-6 membered heteroaryl of RB, RBa and RBb is optionally substituted with a substituent selected from the group consisting of 4-7 membered heterocycloalkyi, Ci-4alkylheterocycloalkyl, Ci-4alklylsulfanyl, cyano, Ci-4alkoxyC2-4alkoxyCi-4 alkyl,
Figure imgf000011_0001
heterocycloalkylCi-4alkyl, heterocycloalkyl- carbonyl- (heterocycloalkyl-C(=0)-), and -C(0)(CH2)pNR'R" where p is 0-4 and R' and R" are independently selected from the group consisting of H and Ci-4alkyl.
In other embodiments, B is a 4, 5, 6 or 7 membered heterocycloalkyi containing 1 or 2 nitrogen atoms, optionally substituted by 1 or 2 RB groups, each independently selected from the group consisting of Ci-4alkyl, C-M alkylsulfanyl, phenyl, 5-6 membered heteroaryl, and -NRBaRBb (where RBa and RBb are independently selected from the group consisting of H, Ci-4alkyl, phenyl, and 5-6 membered heteroaryl), wherein said phenyl or 5-6 membered heteroaryl of RB, RBa and RBb is optionally substituted with a substituent selected from the group consisting of 4-7 membered heterocycloalkyi,
C1-4alkylheterocycloalkyl, C1-4alklylsulfanyl, cyano, C1-4alkoxyC2-4alkoxyC1-4 alkyl, hydroxyC1-4alkyl, 4-7 membered heterocycloalkylC1-4alkyl, 4-7 membered
heterocycloalkylC1-4alkoxy, 4-7 membered heterocycloalkyl-carbonyl, and
-C(0)(CH2)pNR'R" where p is 0-4 and R' and R" are independently selected from the group consisting of H and
Figure imgf000011_0002
In some embodiments, B is pyrimidinyl, phenyl, pyridinyl, or pyrizinyl, wherein said pyrimidinyl, phenyl, pyridinyl, or pyrizinyl is optionally substituted with 1 or 2 RB groups, each independently selected from the group consisting of Ci-4 alkylsulfanyl, -NH2, phenyl, pyrimindinyl, thienyl, thiazolyl, pyrazolyl, and pyridinyl, wherein said phenyl, pyrimindinyl, thiophenyl, thiazolyl, pyrazolyl, or pyridinyl is optionally substituted with morpholinyl, methylpiperazinyl, Ci-4alkylsulfanyl, cyano, methyoxymethoxypropan-2-yl,
Figure imgf000011_0003
morpholinylCi-4alkoxy, morpholinylcarbonyl, or - C(0)(CH2)pNR'R" where p is 0, 1 , 2, 3, or 4 and R' and R" are independently selected from the group consisting of H and
Figure imgf000011_0004
In particular embodiments, B is:
Figure imgf000012_0001
wherein at least 1 of D, E, and F is CH and each remaining group is independently selected from the group consisting of CH and N,
R1 is selected from the group consisting of H, methylsulfanyl, and NH2,
G and H are each independently selected from the group consisting of CH and N, and R2 is selected from the group consisting of H, methylsulfanyl, cyano, methyoxymethyoxypropan-2-yl, 1 -hydroxy-1 -methylethyl, morpholinyl, methylpiperazinyl, morpholinylcarbonyl, morpholinylmethyl, morpholinylethyloxy, dimethylaminocarbonyl, and dimethylaminoethoxy.
In other embodiments, B is phenyl, pyridinyl, pyrazinyl, or pyrimidinyl substituted by 1 ,2-oxazol-3-yl, 1 H-pyrazol-5-yl, 1 ,3-thiazol-2-yl, thiophen-3-yl, wherein said
1 ,2-oxazol-3-yl, 1 H-pyrazol-5-yl, 1 ,3-thiazol-2-yl, thiophen-3-yl is optionally substituted with C1-4alkyl or hydroxyC1-4alkyl, such as hydroxymethyl.
The invention specifically encompasses compounds according Formula (II):
Figure imgf000012_0002
wherein D and E are each independently selected from the group consisting of NH and CH, and n, m and B are as defined herein.
As used herein, the terms "compound(s) of the invention" or "compound(s) of this invention" mean a compound of Formula (I), as defined above, in any form, i.e., any salt or non-salt form (e.g., as a free acid or base form, or as a salt, particularly a
pharmaceutically acceptable salt thereof) and any physical form thereof (e.g., including non-solid forms (e.g., liquid or semi-solid forms), and solid forms (e.g., amorphous or crystalline forms, specific polymorphic forms, solvate forms, including hydrate forms (e.g., mono-, di- and hemi- hydrates)), and mixtures of various forms. As used herein, the term "optionally substituted" means unsubstituted groups or rings (e.g., cycloalkyl, heterocycle, and heteroaryl rings) and groups or rings substituted with one or more specified substituents.
The compounds according to Formula (I) may contain one or more asymmetric center (also referred to as a chiral center) and may, therefore, exist as individual enantiomers, diastereomers, or other stereoisomeric forms, or as mixtures thereof. Chiral centers, such as chiral carbon atoms, may also be present in a substituent such as an alkyl group. Where the stereochemistry of a chiral center present in Formula (I), or in any chemical structure illustrated herein, is not specified the structure is intended to encompass all individual stereoisomers and all mixtures thereof. Thus, compounds according to Formula (I) containing one or more chiral centers may be used as racemic mixtures, scalemic mixtures, or as diaseteromerically or enantiomerically pure materials.
Individual stereoisomers of a compound according to Formula (I) which contain one or more asymmetric center may be resolved by methods known to those skilled in the art. For example, such resolution may be carried out (1 ) by formation of
diastereoisomeric salts, complexes or other derivatives; (2) by selective reaction with a stereoisomer-specific reagent, for example by enzymatic oxidation or reduction; or (3) by gas-liquid or liquid chromatography in a chiral environment, for example, on a chiral support such as silica with a bound chiral ligand or in the presence of a chiral solvent. The skilled artisan will appreciate that where the desired stereoisomer is converted into another chemical entity by one of the separation procedures described above, a further step is required to liberate the desired form. Alternatively, specific stereoisomers may be synthesized by asymmetric synthesis using optically active reagents, substrates, catalysts or solvents, or by converting one enantiomer to the other by asymmetric transformation.
When a disclosed compound or its salt is named or depicted by structure, it is to be understood that the compound or salt, including solvates (particularly, hydrates) thereof, may exist in crystalline forms, non-crystalline forms or a mixture thereof. The compound or salt, or solvates (particularly, hydrates) thereof, may also exhibit
polymorphism (i.e. the capacity to occur in different crystalline forms). These different crystalline forms are typically known as "polymorphs." It is to be understood that when named or depicted by structure, the disclosed compound, or solvates (particularly, hydrates) thereof, also include all polymorphs thereof. Polymorphs have the same chemical composition but differ in packing, geometrical arrangement, and other descriptive properties of the crystalline solid state. Polymorphs, therefore, may have different physical properties such as shape, density, hardness, deformability, stability, and dissolution properties. Polymorphs typically exhibit different melting points, IR spectra, and X-ray powder diffraction patterns, which may be used for identification. One of ordinary skill in the art will appreciate that different polymorphs may be produced, for example, by changing or adjusting the conditions used in crystallizing/recrystallizing the compound.
Because of their potential use in medicine, the salts of the compounds of
Formula (I) are preferably pharmaceutically acceptable salts. Suitable pharmaceutically acceptable salts include those described by Berge, Bighley and Monkhouse,
J.Pharm.Sci (1977) 66, pp 1 -19. Salts encompassed within the term "pharmaceutically acceptable salts" refer to non-toxic salts of the compounds of this invention.
Typically, a salt may be readily prepared by using a desired acid or base as appropriate. The salt may precipitate from solution and be collected by filtration or may be recovered by evaporation of the solvent.
When a compound of the invention is a base (contain a basic moiety), a desired salt form may be prepared by any suitable method known in the art, including treatment of the free base with an inorganic acid, such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like, or with an organic acid, such as acetic acid, trifluoroacetic acid, maleic acid, succinic acid, mandelic acid, fumaric acid, malonic acid, pyruvic acid, oxalic acid, glycolic acid, salicylic acid, and the like, or with a pyranosidyl acid, such as glucuronic acid or galacturonic acid, or with an alpha-hydroxy acid, such as citric acid or tartaric acid, or with an amino acid, such as aspartic acid or glutamic acid, or with an aromatic acid, such as benzoic acid or cinnamic acid, or with a sulfonic acid, such as p-toluenesulfonic acid, methanesulfonic acid, ethanesulfonic acid or the like.
Suitable addition salts are formed from acids which form non-toxic salts and examples include acetate, p-aminobenzoate, ascorbate, aspartate, benzenesulfonate, benzoate, bicarbonate, bismethylenesalicylate, bisulfate, bitartrate, borate, calcium edetate, camsylate, carbonate, clavulanate, citrate, cyclohexylsulfamate, edetate, edisylate, estolate, esylate, ethanedisulfonate, ethanesulfonate, formate, fumarate, gluceptate, gluconate, glutamate, glycollate, glycollylarsanilate, hexylresorcinate, hydrabamine, hydrobromide, hydrochloride, dihydrochloride, hydrofumarate, hydrogen phosphate, hydroiodide, hydromaleate, hydrosuccinate, hydroxynaphthoate, isethionate, itaconate, lactate, lactobionate, laurate, malate, maleate, mandelate, mesylate, methylbromide, methylnitrate, methylsulfate, monopotassium maleate, mucate, napsylate, nitrate, /V-methylglucamine, oxalate, oxaloacetate, pamoate (embonate), palmate, palmitate, pantothenate, phosphate/diphosphate, pyruvate, polygalacturonate, propionate, saccharate, salicylate, stearate, subacetate, succinate, sulfate, tannate, tartrate, teoclate, tosylate, triethiodide, trifluoroacetate and valerate. Other exemplary acid addition salts include pyrosulfate, sulfite, bisulfite, decanoate, caprylate, acrylate, isobutyrate, caproate, heptanoate, propiolate, oxalate, malonate, suberate, sebacate, butyne-1 ,4-dioate, hexyne-1 ,6-dioate, chlorobenzoate, methylbenzoate, dinitrobenzoate, hydroxybenzoate, methoxybenzoate, phthalate, phenylacetate, phenylpropionate, phenylbutrate, lactate, γ-hydroxybutyrate, mandelate, and sulfonates, such as xylenesulfonate, propanesulfonate, naphthalene-1 -sulfonate and naphthalene-2-sulfonate.
If an inventive basic compound is isolated as a salt, the corresponding free base form of that compound may be prepared by any suitable method known to the art, including treatment of the salt with an inorganic or organic base, suitably an inorganic or organic base having a higher pKa than the free base form of the compound.
When a compound of the invention is an acid (contains an acidic moiety), a desired salt may be prepared by any suitable method known to the art, including treatment of the free acid with an inorganic or organic base, such as an amine (primary, secondary, or tertiary), an alkali metal or alkaline earth metal hydroxide, or the like.
Illustrative examples of suitable salts include organic salts derived from amino acids such as glycine and arginine, ammonia, primary, secondary, and tertiary amines, and cyclic amines, such as N-methyl-D-glucamine, diethylamine, isopropylamine, trimethylamine, ethylene diamine, dicyclohexylamine, ethanolamine, piperidine, morpholine, and piperazine, as well as inorganic salts derived from sodium, calcium, potassium, magnesium, manganese, iron, copper, zinc, aluminum, and lithium.
Certain of the compounds of this invention may form salts with one or more equivalents of an acid (if the compound contains a basic moiety) or a base (if the compound contains an acidic moiety). The present invention includes within its scope all possible stoichiometric and non-stoichiometric salt forms.
Compounds of the invention having both a basic and acidic moiety may be in the form of zwitterions, acid-addition salt of the basic moiety or base salts of the acidic moiety.
This invention also provides for the conversion of one pharmaceutically acceptable salt of a compound of this invention, e.g., a hydrochloride salt, into another
pharmaceutically acceptable salt of a compound of this invention, e.g., a sulfate salt.
For solvates of the compounds of Formula (I), or salts thereof that are in crystalline form, the skilled artisan will appreciate that pharmaceutically-acceptable solvates may be formed wherein solvent molecules are incorporated into the crystalline lattice during crystallization. Solvates may involve nonaqueous solvents such as ethanol, isopropanol, DMSO, acetic acid, ethanolamine, and ethyl acetate, or they may involve water as the solvent that is incorporated into the crystalline lattice. Solvates wherein water is the solvent that is incorporated into the crystalline lattice are typically referred to as
"hydrates." Hydrates include stoichiometric hydrates as well as compositions containing variable amounts of water. The invention includes all such solvates.
Because the compounds of Formula (I) are intended for use in pharmaceutical compositions it will readily be understood that they are each preferably provided in substantially pure form, for example at least 60% pure, more suitably at least 75% pure and preferably at least 85%, especially at least 98% pure (% are on a weight for weight basis). Impure preparations of the compounds may be used for preparing the more pure forms used in the pharmaceutical compositions.
The compounds of this invention may be obtained by using synthetic procedures illustrated in the Schemes below or by drawing on the knowledge of a skilled organic chemist. The synthesis provided in these Schemes are applicable for producing compounds of the invention having a variety of different substituent groups employing appropriate precursors, which are suitably protected if needed, to achieve compatibility with the reactions outlined herein. Subsequent deprotection, where needed, affords compounds of the nature generally disclosed. While the Schemes are shown with compounds only of Formula (I), they are illustrative of processes that may be used to make the compounds of the invention.
Intermediates (compounds used in the preparation of the compounds of the invention) may also be present as salts. Thus, in reference to intermediates, the phrase "compound(s) of formula (number)" means a compound having that structural formula or a pharmaceutically acceptable salt thereof.
Specific compounds of this invention are:
4-(pyridin-3-yl)-/V-(4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)benzyl)pyrimidin-2- amine,
A/-((1-methyl-1 H-indol-3-yl)methyl)-5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3- yl)pyrimidin-2-amine,
Λ/-(2-(1 -Methyl-1 H-indol-3-yl)ethyl)-5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3- yl)pyrimidin-2-amine,
and salts, particularly pharmaceutically acceptable salts, thereof.
Other compounds that may be prepared using the methods described herein include:
4-(pyridin-3-yl)-N-(4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)benzyl)pyrimidin-2- amine; 4-(pyridin-3-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyridin-2- yl)methyl)pyrimidin-2-amine;
4-(pyridin-3-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyridin-2- yl)methyl)pyrimidin-2-amine;
3-(pyridin-3-yl)-N-(4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)benzyl)aniline;
N-(4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)benzyl)-[2,3'-bipyridin]-6-amine;
2-(pyridin-3-yl)-N-(4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)benzyl)pyrimidin-4- amine;
6-(pyridin-3-yl)-N-(4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)benzyl)pyrimidin-4- amine;
6-(pyridin-3-yl)-N-(4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)benzyl)pyrazin-2- amine;
2- (methylthio)-6-(pyridin-3-yl)-N-(4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3- yl)benzyl)pyrimidin-4-amine;
6-(pyridin-3-yl)-N2-(4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)benzyl)pyrimidine- 2,4-diamine;
3- (pyridin-3-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyridin-2- yl)methyl)aniline;
N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyridin-2-yl)methyl)-[2,3'-bipyridin]-6- amine;
2-(pyridin-3-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyridin-2- yl)methyl)pyrimidin-4-amine;
6-(pyridin-3-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyridin-2- yl)methyl)pyrimidin-4-amine;
6-(pyridin-3-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyridin-2- yl)methyl)pyrazin-2-amine;
2- (methylthio)-6-(pyridin-3-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3- yl)pyridin-2-yl)methyl)pyrimidin-4-amine;
6-(pyridin-3-yl)-N2-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyridin-2- yl)methyl)pyrimidine-2,4-diamine;
3- (pyridin-3-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyrimidin-2- yl)methyl)aniline;
N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyrimidin-2-yl)methyl)-[2,3'-bipyridin]- 6-amine;
2-(pyridin-3-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyrimidin-2- yl)methyl)pyrimidin-4-amine; 6-(pyridin-3-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyrimidin-2- yl)methyl)pyrimidin-4-amine;
6-(pyridin-3-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyrimidin-2- yl)methyl)pyrazin-2-amine;
2-(methylthio)-6-(pyridin-3-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3- yl)pyrimidin-2-yl)methyl)pyrimidin-4-amine;
6-(pyridin-3-yl)-N2-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyrimidin-2- yl)methyl)pyrimidine-2,4-diamine;
4-(6-morpholinopyridin-3-yl)-N-(4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3- yl)benzyl)pyrimidin-2-amine;
4-(6-(4-methylpiperazin-1-yl)pyridin-3-yl)-N-(4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol- 3-yl)benzyl)pyrimidin-2-amine;
4- (6-(methylthio)pyridin-3-yl)-N-(4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3- yl)benzyl)pyrimidin-2-amine;
5- (2-((4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)benzyl)amino)pyrimidin-4- yl)picolinonitrile;
4-(6-(2-(methoxymethoxy)propan-2-yl)pyridin-3-yl)-N-(4-(5-(trifluoromethyl)-1 ,2,4- oxadiazol-3-yl)benzyl)pyrimidin-2-amine;
2-(5-(2-((4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)benzyl)amino)pyrimidin-4- yl)pyridin-2-yl)propan-2-ol;
N,N-dimethyl-5-(2-((4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3- yl)benzyl)amino)pyrimidin-4-yl)picolinamide;
morpholino(5-(2-((4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3- yl)benzyl)amino)pyrimidin-4-yl)pyridin-2-yl)methanone;
4-(6-morpholinopyridin-3-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyridin-
2- yl)methyl)pyrimidin-2-amine;
4-(6-(4-methylpiperazin-1-yl)pyridin-3-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-
3- yl)pyridin-2-yl)methyl)pyrimidin-2-amine;
4- (6-(methylthio)pyridin-3-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyridin- 2-yl)methyl)pyrimidin-2-amine;
5- (2-(((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyridin-2- yl)methyl)amino)pyrimidin-4-yl)picolinonitrile;
4-(6-(2-(methoxymethoxy)propan-2-yl)pyridin-3-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4- oxadiazol-3-yl)pyridin-2-yl)methyl)pyrimidin-2-amine;
2-(5-(2-(((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyridin-2- yl)methyl)amino)pyrimidin-4-yl)pyridin-2-yl)propan-2-ol; N,N-dimethyl-5-(2-(((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyridin-2- yl)methyl)amino)pyrimidin-4-yl)picolinamide;
morpholino(5-(2-(((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyridin-2- yl)methyl)amino)pyrimidin-4-yl)pyridin-2-yl)methanone;
4-(6-morpholinopyridin-3-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3- yl)pyrimidin-2-yl)methyl)pyrimidin-2-amine;
4-(6-(4-methylpiperazin-1-yl)pyridin-3-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol- 3-yl)pyrimidin-2-yl)methyl)pyrimidin-2-amine;
4- (6-(methylthio)pyridin-3-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3- yl)pyrimidin-2-yl)methyl)pyrimidin-2-amine;
5- (2-(((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyrimidin-2- yl)methyl)amino)pyrimidin-4-yl)picolinonitrile;
4-(6-(2-(methoxymethoxy)propan-2-yl)pyridin-3-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4- oxadiazol-3-yl)pyrimidin-2-yl)methyl)pyrimidin-2-amine;
2-(5-(2-(((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyrimidin-2- yl)methyl)amino)pyrimidin-4-yl)pyridin-2-yl)propan-2-ol;
N,N-dimethyl-5-(2-(((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyrimidin-2- yl)methyl)amino)pyrimidin-4-yl)picolinamide;
morpholino(5-(2-(((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyrimidin-2- yl)methyl)amino)pyrimidin-4-yl)pyridin-2-yl)methanone;
4-(pyrazin-2-yl)-N-(4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)benzyl)pyrimidin-2- amine;
4-(thiophen-3-yl)-N-(4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)benzyl)pyrimidin-2- amine;
4-(thiazol-2-yl)-N-(4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)benzyl)pyrimidin-2- amine;
4-(1 H-pyrazol-5-yl)-N-(4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)benzyl)pyrimidin- 2-amine;
(5-(2-((4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)benzyl)amino)pyrimidin-4- yl)isoxazol-3-yl)methanol;
4-(4-(morpholinomethyl)phenyl)-N-(4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3- yl)benzyl)pyrimidin-2-amine;
4-(4-(2-morpholinoethoxy)phenyl)-N-(4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3- yl)benzyl)pyrimidin-2-amine;
4-(3-(2-morpholinoethoxy)phenyl)-N-(4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3- yl)benzyl)pyrimidin-2-amine; 4-(4-(2-(dimethylamino)ethoxy)phenyl)-N-(4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3- yl)benzyl)pyrimidin-2-amine;
4-(3-(2-(dimethylamino)ethoxy)phenyl)-N-(4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3- yl)benzyl)pyrimidin-2-amine;
4-(pyrazin-2-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyridin-2- yl)methyl)pyrimidin-2-amine;
4-(thiophen-3-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyridin-2- yl)methyl)pyrimidin-2-amine;
4-(thiazol-2-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyridin-2- yl)methyl)pyrimidin-2-amine;
4-(1 H-pyrazol-5-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyridin-2- yl)methyl)pyrimidin-2-amine;
(5-(2-(((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyridin-2- yl)methyl)amino)pyrimidin-4-yl)isoxazol-3-yl)methanol;
4-(4-(morpholinomethyl)phenyl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3- yl)pyridin-2-yl)methyl)pyrimidin-2-amine;
4-(4-(2-morpholinoethoxy)phenyl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3- yl)pyridin-2-yl)methyl)pyrimidin-2-amine;
4-(3-(2-morpholinoethoxy)phenyl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3- yl)pyridin-2-yl)methyl)pyrimidin-2-amine;
4-(4-(2-(dimethylamino)ethoxy)phenyl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3 yl)pyridin-2-yl)methyl)pyrimidin-2-amine;
4-(3-(2-(dimethylamino)ethoxy)phenyl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3 yl)pyridin-2-yl)methyl)pyrimidin-2-amine;;
4-(pyrazin-2-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyrimidin-2- yl)methyl)pyrimidin-2-amine;
4-(thiophen-3-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyrimidin-2- yl)methyl)pyrimidin-2-amine;
4-(thiazol-2-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyrimidin-2- yl)methyl)pyrimidin-2-amine;
4-(1 H-pyrazol-5-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyrimidin-2- yl)methyl)pyrimidin-2-amine;
(5-(2-(((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyrimidin-2- yl)methyl)amino)pyrimidin-4-yl)isoxazol-3-yl)methanol;
4-(4-(morpholinomethyl)phenyl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3- yl)pyrimidin-2-yl)methyl)pyrimidin-2-amine; 4-(4-(2-morpholinoethoxy)phenyl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3- yl)pyrimidin-2-yl)methyl)pyrimidin-2-amine;
4-(3-(2-morpholinoethoxy)phenyl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3- yl)pyrimidin-2-yl)methyl)pyrimidin-2-amine;
4-(4-(2-(dimethylamino)ethoxy)phenyl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3 yl)pyrimidin-2-yl)methyl)pyrimidin-2-amine;
4-(3-(2-(dimethylamino)ethoxy)phenyl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-^ yl)pyrimidin-2-yl)methyl)pyrimidin-2-amine; and
salts, particularly pharmaceutically acceptable salts, thereof.
Compound names were generated using the software naming program
ChemDraw 1 1.0 available from CambridgeSoft Corporation, 100 CambridgePark Drive, Cambridge, MA 02140, USA (http://www.cambridgesoft.com).
The compounds of Formula (I) can be prepared according to the methods outlined below.
Scheme 1
Figure imgf000021_0001
The invention also includes various deuterated forms of the compounds of Formula (I). Each available hydrogen atom attached to a carbon atom may be
independently replaced with a deuterium atom. A person of ordinary skill in the art will know how to synthesize deuterated forms of the compounds of Formula (I). For example, deuterated alkyl groups (e.g., /V-(deutero-methyl) amines) may be prepared by
conventional techniques (see for example: methyl-c/3-amine available from Aldrich
Chemical Co., Milwaukee, Wl, Cat. No.489, 689-2). Employing such compounds will allow for the preparation of compounds of Formula (I) in which various hydrogen atoms of the /V-methyl groups are replaced with a deuterium atom.
The present invention is directed to a method of inhibiting an HDAC which comprises contacting the acetylase with a compound of Formula (I) or a salt thereof, particularly a pharmaceutically acceptable salt thereof. More specifically, this invention is directed to a method of inhibiting HDAC comprising contacting a cell with an effective amount of a compound of Formula (I) or a salt thereof, particularly a pharmaceutically acceptable salt thereof. This invention is also directed to a method of treatment of an HDAC-mediated disease or disorder comprising administering a therapeutically effective amount of the compound of Formula (I) or a salt thereof, particularly a pharmaceutically acceptable salt thereof, to a patient, specifically a human, in need thereof. As used herein, "patient" refers to a mammal, specifically, a human. A therapeutically "effective amount" is intended to mean that amount of a compound that, when administered to a patient in need of such treatment, is sufficient to effect treatment, as defined herein.
Thus, e.g., a therapeutically effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, is a quantity of an inventive agent that, when administered to a human in need thereof, is sufficient to inhibit the activity of HDAC such that a disease condition which is mediated by that activity is reduced, alleviated or prevented. The amount of a given compound that will correspond to such an amount will vary depending upon factors such as the particular compound (e.g., the potency (pXC5o), efficacy (EC50), and the biological half-life of the particular compound), disease condition and its severity, the identity (e.g., age, size and weight) of the patient in need of treatment, but can nevertheless be routinely determined by one skilled in the art. Likewise, the duration of treatment and the time period of administration (time period between dosages and the timing of the dosages, e.g., before/with/after meals) of the compound will vary according to the identity of the mammal in need of treatment (e.g., weight), the particular compound and its properties (e.g., pharmaceutical characteristics), disease or condition and its severity and the specific composition and method being used, but can
nevertheless be determined by one of skill in the art. "Treating" or "treatment" is intended to mean at least the mitigation of a disease condition in a patient, where the disease condition is caused or mediated by HDAC. The methods of treatment for mitigation of a disease condition include the use of the compounds in this invention in any conventionally acceptable manner, for example for prevention, retardation, prophylaxis, therapy or cure of a disease.
In one embodiment, this invention is directed to a method of treating, ameliorating, or preventing an autoimmune disorder, an immunological disease, an inflammatory disorder, transplant/graft rejection (e.g., allograft), lymphopenia, or graft-versus-host disease (GvHD) in a patient, specifically in a human, comprising administering to the patient a compound of this invention, in an amount sufficient to increase the level and/or activity of a Treg cell or a population of Treg cells in the patient, thereby treating, ameliorating, or preventing the autoimmune disorder, inflammatory disorder,
transplant/graft rejection, lymphopenia, or GvHD in the patient.
Additional examples of diseases and conditions that may be treated by the compounds of this invention include but not limited to type II diabetes mellitus, coronary artery disease, alopecia, allergies and allergic reactions, and sepsis/toxic shock.
Exemplary autoimmune disorders include, but are not limited to, multiple sclerosis, juvenile idiopathic arthritis, psoriatic arthritis, hepatitis C virus-associated mixed cryoglobulinemia, polymyositis, dermatomyositis, polyglandular syndrome type II, autoimmune liver disease, Kawasaki disease, myasthenia gravis, immunodysregulation polyendocrinopathy enteropathy X-linked syndrome (IPEX (syndrome)), type I diabetes, psoriasis, hypothyroidism, hemolytic anemia, autoimmune polyendocrinopathy- candidiasis-ectodermal dystrophy (APECED), thrombocytopenia, spondyloarthritis, Sjogren's syndrome, rheumatoid arthritis, inflammatory bowel disease (IBD), Crohn's disease, ulcerative colitis, eczema, gastritis, or thyroiditis. As part of a nonlimiting list, the inflammatory disorder can be contact hypersensitivity, atopic dermatitis or Still disease.
Additional examples of autoimmune diseases include but are not limited to autoimmune diseases include osteoarthritis, systemic sclerosis, sarcoidosis, insulin dependent diabetes mellitus (IDDM, type I diabetes), reactive arthritis, scleroderma, vasculitis, Wegener's granulomatosis, Hashimoto's disease, scleroderma, oophoritis, Lupus (SLE), Grave's disease, asthma, cryoglobulinemia, primary biliary sclerosis, pemphigus vulgaris, hemolytic anemia and pernicious anemia.
Examples of transplant/graft rejection (e.g., allograft), lymphopenia, or graft- versus-host disease (GvHD) are those arising from cell, tissue and organ transplantation procedures, such as therapeutic cell transplants such as stem cells, muscle cells such as cardiac cells, islet cells, liver cells, bone marrow transplants, skin grafts, bone grafts, lung transplants, kidney transplants, liver transplants, and heart transplants.
In another embodiment, this invention is directed to a method of treating an HDAC-mediated neurodegenerative disease or disorder which comprises administering to a patient in need thereof, a compound of Formula I or a salt thereof, particularly a pharmaceutically acceptable salt thereof. This invention is also directed to a method of treatment of a neurodegenerative disease or disorder associated with deacetylases, such as, Alzheimer's disease, Parkinson's disease, neuronal intranuclear inclusion disease (NMD), and polyglutamine disorders, such as Huntington's disease and spinocerebellar ataxia (SCA), comprising administering a therapeutically effective amount of the compound of Formula I or a salt thereof, particularly a pharmaceutically acceptable salt thereof, to a patient, specifically a human, in need thereof.
Other examples of diseases and conditions that may be treated by the compounds of this invention include but are not limited to cystic fibrosis, osteoporosis, obesity, epilepsy, depression, thalassemia, sickle cell anemia, amyotrophic lateral sclerosis (ALS) and hyperalgesia, cardiac disease (e.g., stroke, hypertension, atherothrombotic diseases, artherosclerosis or limitation of infarct size in acute coronary syndrome), diseases or disorders involving muscular atrophy, gentamicin-induced hearing loss, drug resistance (e.g., drug resistance in osteosarcoma and colon cancer cells), infectious diseases, and immune deficiency/immunocompromised patients. Examples of infectious diseases relate to various pathogen infections such as viral, fungal, bacterial, mycoplasm, and infections by unicellular and multicellular eukaryotic organisms. Common human pathogens include but are not limited to HIV, HSV, HPV, Hepatitis A, B and C viruses, influenza, denge, zostrella, rubella, RSV, rotavirus, gram positive, gram negative, streptococcus, tetanus, staphalococcus, tuberculosis, listeria, and malaria.
The compounds of the invention may be employed alone or in combination with standard anti-cancer regimens for neoplastic cell, e.g., tumor cell and cancer cell, treatments. Thus, in another embodiment, this invention is directed to inhibitors of HDAC and their use to stop or reduce the growth of neoplastic cells, e.g., cancer cells and tumor cells. The growth of cancer cells and/or tumor cells that are found in the following cancer types may be reduced by treatment with a compound of this invention: carcinoma (e.g., adenocarcinoma), heptaocellular carcinoma, sarcoma, myeloma (e.g., multiple myeloma), treating bone disease in multiple myeloma, leukemia, childhood acute lymphoblastic leukemia and lymphoma (e.g., cutaneous cell lymphoma), and mixed types of cancers, such as adenosquamous carcinoma, mixed mesodermal tumor, carcinosarcoma, and teratocarcinoma. In one aspect of the invention, breast or prostate cancers or tumors are treated using the HDAC inhibitors of this invention. Other cancers that may be treated using the compounds of this invention include, but are not limited to, bladder cancer, breast cancer, prostate cancer, stomach cancer, lung cancer, colon cancer, rectal cancer, colorectal cancer, liver cancer, endometrial cancer, pancreatic cancer, cervical cancer, ovarian cancer; head and neck cancer, and melanoma.
The present invention is further directed to a method of treating a B-cell lymphoma, particularly a B-cell lymphoma associated with deacetylases, which comprises administering to a patient in need thereof, a compound of Formula I or a salt thereof, particularly a pharmaceutically acceptable salt thereof. Examples of B-cell lymphomas associated with deacetylases that may be treated using the method of this invention include Burkitt lymphoma, chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL/SLL), diffuse large B-cell lymphoma, follicular lymphoma, immunoblastic large cell lymphoma, precursor B-lymphoblastic lymphoma, mantle cell lymphoma, and
Waldenstrom Macroglobulinemia (lymphoplasmacytic lymphoma). More specifically, this invention is directed to a method of treatment of Burkitt lymphoma, chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL/SLL), diffuse large B-cell lymphoma, follicular lymphoma, immunoblastic large cell lymphoma, precursor B-lymphoblastic lymphoma, mantle cell lymphoma, and Waldenstrom Macroglobulinemia (lymphoplasmacytic lymphoma), comprising administering a therapeutically effective amount of the compound of Formula I or a salt thereof, particularly a pharmaceutically acceptable salt thereof, to a patient, specifically a human, in need thereof.
The compounds of the invention may be administered by any suitable route of administration, including both systemic administration and topical administration.
Systemic administration includes oral administration, parenteral administration, transdermal administration, rectal administration, and administration by inhalation.
Parenteral administration refers to routes of administration other than enteral,
transdermal, or by inhalation, and is typically by injection or infusion. Parenteral administration includes intravenous, intramuscular, and subcutaneous injection or infusion. Inhalation refers to administration into the patient's lungs whether inhaled through the mouth or through the nasal passages. Topical administration includes application to the skin.
The compounds of the invention may be administered once or according to a dosing regimen wherein a number of doses are administered at varying intervals of time for a given period of time. For example, doses may be administered one, two, three, or four times per day. Doses may be administered until the desired therapeutic effect is achieved or indefinitely to maintain the desired therapeutic effect. Suitable dosing regimens for a compound of the invention depend on the pharmacokinetic properties of that compound, such as absorption, distribution, and half-life, which can be determined by the skilled artisan. In addition, suitable dosing regimens, including the duration such regimens are administered, for a compound of the invention depend on the condition being treated, the severity of the condition being treated, the age and physical condition of the patient being treated, the medical history of the patient to be treated, the nature of concurrent therapy, the desired therapeutic effect, and like factors within the knowledge and expertise of the skilled artisan. It will be further understood by such skilled artisans that suitable dosing regimens may require adjustment given an individual patient's response to the dosing regimen or over time as individual patient needs change.
Treatment of HDAC-mediated disease conditions may be achieved using the compounds of this invention as a monotherapy, or in dual or multiple combination therapy, such as in combination with other agents, for example, in combination with one or more of the following agents: DNA methyltransferase inhibitors, acetyl transferase enhancers, proteasome or HSP90 inhibitors, and one or more immunosuppressants that do not activate the T suppressor cells including but are not limited to corticosteroids, rapamycin, Azathioprine, Mycophenolate, Cyclosporine, Mercaptopurine (6-MP), basiliximab, daclizumab, sirolimus, tacrolimus, Muromonab-CD3, cyclophosphamide, and
methotrexate, which are administered in effective amounts as is known in the art.
The compounds of the invention will normally, but not necessarily, be formulated into a pharmaceutical composition prior to administration to a patient. Accordingly, in another aspect the invention is directed to pharmaceutical compositions comprising a compound of the invention and a pharmaceutically-acceptable excipient.
The pharmaceutical compositions of the invention may be prepared and packaged in bulk form wherein an effective amount of a compound of the invention can be extracted and then given to the patient such as with powders, syrups, and solutions for injection. Alternatively, the pharmaceutical compositions of the invention may be prepared and packaged in unit dosage form. For oral application, for example, one or more tablets or capsules may be administered. A dose of the pharmaceutical composition contains at least a therapeutically effective amount of a compound of this invention (i.e., a compound of Formula (I) or a salt, particularly a pharmaceutically acceptable salt, thereof). When prepared in unit dosage form, the pharmaceutical compositions may contain from 1 mg to 1000 mg of a compound of this invention.
The pharmaceutical compositions of the invention typically contain one compound of the invention. However, in certain embodiments, the pharmaceutical compositions of the invention contain more than one compound of the invention. In addition, the pharmaceutical compositions of the invention may optionally further comprise one or more additional pharmaceutically active compounds.
As used herein, "pharmaceutically-acceptable excipient" means a material, composition or vehicle involved in giving form or consistency to the composition. Each excipient must be compatible with the other ingredients of the pharmaceutical composition when commingled such that interactions which would substantially reduce the efficacy of the compound of the invention when administered to a patient and interactions which would result in pharmaceutical compositions that are not pharmaceutically-acceptable are avoided. In addition, each excipient must of course be of sufficiently high purity to render it pharmaceutically-acceptable.
The compounds of the invention and the pharmaceutically-acceptable excipient or excipients will typically be formulated into a dosage form adapted for administration to the patient by the desired route of administration. Conventional dosage forms include those adapted for (1 ) oral administration such as tablets, capsules, caplets, pills, troches, powders, syrups, elixirs, suspensions, solutions, emulsions, sachets, and cachets; (2) parenteral administration such as sterile solutions, suspensions, and powders for reconstitution; (3) transdermal administration such as transdermal patches; (4) rectal administration such as suppositories; (5) inhalation such as aerosols and solutions; and (6) topical administration such as creams, ointments, lotions, solutions, pastes, sprays, foams, and gels.
Suitable pharmaceutically-acceptable excipients will vary depending upon the particular dosage form chosen. In addition, suitable pharmaceutically-acceptable excipients may be chosen for a particular function that they may serve in the composition. For example, certain pharmaceutically-acceptable excipients may be chosen for their ability to facilitate the production of uniform dosage forms. Certain pharmaceutically- acceptable excipients may be chosen for their ability to facilitate the production of stable dosage forms. Certain pharmaceutically-acceptable excipients may be chosen for their ability to facilitate the carrying or transporting the compound or compounds of the invention once administered to the patient from one organ, or portion of the body, to another organ, or portion of the body. Certain pharmaceutically-acceptable excipients may be chosen for their ability to enhance patient compliance.
Suitable pharmaceutically-acceptable excipients include the following types of excipients: diluents, fillers, binders, disintegrants, lubricants, glidants, granulating agents, coating agents, wetting agents, solvents, co-solvents, suspending agents, emulsifiers, sweeteners, flavoring agents, flavor masking agents, coloring agents, anti-caking agents, humectants, chelating agents, plasticizers, viscosity increasing agents, antioxidants, preservatives, stabilizers, surfactants, and buffering agents. The skilled artisan will appreciate that certain pharmaceutically-acceptable excipients may serve more than one function and may serve alternative functions depending on how much of the excipient is present in the formulation and what other ingredients are present in the formulation.
Skilled artisans possess the knowledge and skill in the art to enable them to select suitable pharmaceutically-acceptable excipients in appropriate amounts for use in the invention. In addition, there are a number of resources that are available to the skilled artisan which describe pharmaceutically-acceptable excipients and may be useful in selecting suitable pharmaceutically-acceptable excipients. Examples include Remington's Pharmaceutical Sciences (Mack Publishing Company), The Handbook of Pharmaceutical Additives (Gower Publishing Limited), and The Handbook of Pharmaceutical Excipients (the American Pharmaceutical Association and the Pharmaceutical Press).
The pharmaceutical compositions of the invention are prepared using techniques and methods known to those skilled in the art. Some of the methods commonly used in the art are described in Remington's Pharmaceutical Sciences (Mack Publishing
Company).
In one aspect, the invention is directed to a solid oral dosage form such as a tablet or capsule comprising an effective amount of a compound of the invention and a diluent or filler. Suitable diluents and fillers include lactose, sucrose, dextrose, mannitol, sorbitol, starch (e.g. corn starch, potato starch, and pre-gelatinized starch), cellulose and its derivatives (e.g. microcrystalline cellulose), calcium sulfate, and dibasic calcium phosphate. The oral solid dosage form may further comprise a binder. Suitable binders include starch (e.g. corn starch, potato starch, and pre-gelatinized starch), gelatin, acacia, sodium alginate, alginic acid, tragacanth, guar gum, povidone, and cellulose and its derivatives (e.g. microcrystalline cellulose). The oral solid dosage form may further comprise a disintegrant. Suitable disintegrants include crospovidone, sodium starch glycolate, croscarmelose, alginic acid, and sodium carboxymethyl cellulose. The oral solid dosage form may further comprise a lubricant. Suitable lubricants include stearic acid, magnesium stearate, calcium stearate, and talc.
EXAMPLES
The following examples illustrate the invention. These examples are not intended to limit the scope of the present invention, but rather to provide guidance to the skilled artisan to prepare and use the compounds, compositions, and methods of the present invention. While particular embodiments of the present invention are described, the skilled artisan will appreciate that various changes and modifications can be made without departing from the spirit and scope of the invention.
In the following experimental descriptions, the following abbreviations may be used:
Figure imgf000029_0001
Capsules are prepared using conventional methods and are formulated as follows:
Inqredient Amount per tablet
Compound 15mg
Dried starch 178mg
Maqnesium stearate 2mq
Total 195mg EXAMPLE 1
Figure imgf000030_0001
2-Amino-A -hydroxypyrimidine-5-carboximidamide: Hydroxylamine hydrochloride (1 .464 g, 21.06 mmol) and potassium carbonate (2.91 g, 21.06 mmol) were added to a suspension of 2-aminopyrimidine-5-carbonitrile (1 .265 g, 10.53 mmol) in ethanol (60 ml.) with a few milligrams of 8-hydroxyquinoline. The resulting mixture was heated under reflux overnight, cooled, and filtered. The solid obtained was suspended in water (30ml), stirred thoroughly, filtered, washed with water and isopropanol, and then dried in air to afford the title product as a beige powder (0.95 g, 56%). 1H NMR (400 MHz, DMSO-d6) δ 9.47 (s, 1 H), 8.46 (s, 2H),
Figure imgf000030_0002
5-(5-(Trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyrimidin-2-amine: TFAA (2.59 ml_, 18.32 mmol) was slowly added to a suspension of 2-amino-/V-hydroxypyrimidine-5- carboximidamide (0.935 g, 6.1 1 mmol) in pyridine (10 ml_)at 0 °C. The reaction mixture was warmed to rt, heated to 50 °C, concentrated under reduced pressure, and partitioned between EtOAc and satd. NH4CI. The organic phase was separated, and the aqueous was extracted with EtOAc. The combined organic phases were washed with water and brine, dried over MgS04, filtered, and concentrated under reduced pressure. The residue was recrystallized from DCM/hexanes to afford the title compound as an off-white solid (1 .0 g). The filtrate was further purified by silica gel chromatography (0% - 50% EtOAc in hexanes) to obtain a second batch of the product (0.28 g). Total yield: 91 %. 1H NMR (400 MHz, DMSO-de) δ 8.84 (s, 2H), 7.58 (s, 2H). 19F NMR (376 MHz, DMSO-d6) δ -65.1. LCMS: fR = 0.65 +H)+.
Figure imgf000030_0003
3-(2-Chloropyrimidin-5-yl)-5-(trifluoromethyl)-1 ,2,4-oxadiazole: Copper(ll) chloride (0.785 g, 5.84 mmol) and ferf-butyl nitrite (0.695 ml_, 5.84 mmol) were added to a suspension of 5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyrimidin-2-amine (0.9 g, 3.89 mmol) in MeCN (50 ml.) . The reaction mixture was heated at 60 °C under nitrogen for 2 h, cooled, diluted with ether, and filtered. The filtrate was concentrated under reduced pressure and purified by silica gel chromatography (0% - 15% EtOAc in hexanes) to afford the title compound as an off-white solid (0.32 g, 32.5%). 1H NMR (400 MHz,
CHLOROFORM-d) δ 9.35 (s, 2H). 19F NMR (376 MHz, CHLOROFORM-d)
δ -6 = 0.84 min, 96%. MS (ESI): m/z 250 (M+H)+.
Figure imgf000031_0001
Λ -(2-(1 -Methyl-1 H-indol-3-yl)ethyl)-5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3- yl)pyrimidin-2-amine: 2-(1 -Methyl-1 H-indol-3-yl)ethanamine (0.063 g, 0.359 mmol) was added to a solution of 3-(2-chloropyrimidin-5-yl)-5-(trifluoromethyl)-1 ,2,4-oxadiazole (0.045 g, 0.180 mmol) and /'-Pr2NEt (0.063 mL, 0.359 mmol) in NMP (2 mL) . The reaction mixture was heated at 70 °C overnight, cooled, poured into water, and extracted with EtOAc three times. The combined organic phases were washed with water/brine, dried over MgS04, filtered, and concentrated under reduced pressure. The residue was purified by silica gel chromatography (0% - 40% EtOAc in hexanes) to afford the title compound as a white solid (21 mg, 30%). 1H NMR (400 MHz, CHLOROFORM-d) δ 8.82 - 9.10 (m, 2H), 7.68 (d, J = 7.83 Hz, 1 H), 7.33 - 7.41 (m, 1 H), 7.30 (d, J = 7.05 Hz, 1 H), 7.13 - 7.22 (m, 1 H), 6.97 (s, 1 H), 5.83 (br. s., 1 H), 3.89 (q, J = 6.46 Hz, 2H), 3.81 (s, 3H), 3.15 (t, J = 6.66 Hz, 2H). LCMS: fR = 1 .06 min, 100%. MS m/z = 389 (M+H)+.
EXAMPLE 2
Figure imgf000031_0002
Λ -((1 -Methyl-1 H-indol-3-yl)methyl)-5-(5-(trifluoromethyl)-1 , 2,4-oxadiazol-3- yl)pyrimidin-2 -amine: A procedure similar to the one used in the synthesis of Λ/-(2-(1 - methyl-1 /-/-indol-3-yl)ethyl)-5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyrimidin-2-amine (example 1 ) but using (1 -methyl-1 H-indol-3-yl)methanamine was used to prepare the title compound as a white solid. 1H NMR (400 MHz, CHLOROFORM-d) δ 9.06 (br. s., 1 H), 7.65 (d, J = 7.83 Hz, 1 H), 7.30 - 7.40 (m, 1 H), 7.23 - 7.31 (m, 2H), 7.08 - 7.20 (m, 2H), 5.92 (br. s., 1 H), 4.86 (d, J = 5.09 Hz, 2H), 3.80 (s, 3H). 19F NMR (376 MHz,
CHLOROFORM-d) δ -65.7. LCMS: fR = 1.00 min, 100%. MS m/z = 375 (M+H)+.
Figure imgf000032_0001
2-Chloro-4-(pyridin-3-yl)pyrimidine: Pyridin-3-ylboronic acid (263 mg, 2.14 mmol), PEPPSI - IPr (132 mg, 0.195 mmol), and potassium carbonate (2.4 ml of 2 M aqueous solution, 4.8 mmol) were added to 2,4-dichloropyrimidine (290 mg, 1 .95 mmol) in 1 ,2- dimethoxyethane (8 mL) . The reaction mixture was stirred at 100 °C for 1 h. The solvents were removed under reduced pressure, and the residue was dissolved in DCM and filtered. The filtrate was purified with silica gel chromatography (20% - 60% EtOAc in hexanes) to yield 150 mg, 40.2 % yield of the title compound. 1H NMR (400 MHz,
CHLOROFORM-d) δ 1 H NMR (400 MHz, CHLOROFORM-d) δ 9.28 (br. s., 1 H), 8.79 (d, J = 3.92 Hz, 1 H), 8.73 (d, J = 5.09 Hz, 1 H), 8.46 (d, J = 8.03 Hz, 1 H), 7.72 (d, J = 5.29 Hz, 1 H), 7.49 (dd, 1 H). LCMS: fR = 0.47 min, 93%. MS m/z = 192 (M+H)+.
Figure imgf000032_0002
ferf-Butyl 4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)benzylcarbamate: To a suspension of ferf-butyl 4-cyanobenzylcarbamate (1.32 g, 5.70 mmol), /'-Pr2NEt (2.99 mL, 17.10 mmol) in ethanol (26 mL) was added hydroxylamine hydrochloride (380 mg, 1 1.4 mmol). The reaction mixture was stirred at 90 °C for 4 h. The solvents were removed under reduced pressure, and the residue was dissolved in THF (26.0 mL). TFAA (2.38 mL, 17.1 mmol) was added carefully to the reaction under stirring followed by solid DMAP (139 mg, 1.140 mmol). The reaction mixture was stirred at rt for 3 h. After concentrating under reduced pressure, the reaction mixture was dissolved in DCM and quenched with satd. NH4CI. The organic layer was separated, dried over MgS04, filtered, and
concentrated under reduced pressure. The obtained residue was dissolved in MeOH and purified via reverse phase prep-HPLC (10% - 100% MeCN in water (0.05% TFA) to give the title compound (620 mg, 32 % yield). 1 H NMR (400 MHz, CHLOROFORM-d) δ 8.09 (d, J = 8.22 Hz, 2H), 7.45 (d, J = 8.22 Hz, 2H), 4.93 (br. s., 1 H), 4.41 (d, J = 5.68 Hz, 2H), 1.35 - 1.57 (m, 9H). = 1.01 min, 92%. MS m/z = 344 (M+H)+.
Figure imgf000033_0001
(4-(5-(Trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)phenyl)methanamine: To a suspension of ferf-butyl 4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)benzylcarbamate (98 mg, 0.28 mmol) was dissolved in a solution of HCI 4 M in 1 ,4-dioxane (2.2 ml). The reaction mixture was stirred for 1 h at rt. The solvents were removed under reduced pressure and the crude was dissolved in DCM and concentrated down again to give the title compound (75 mg, 94 % yield). 1 H NMR (400 MHz, DMSO-d6) δ 8.55 (br. s., 3H), 8.1 1 (d, J = 8.22 Hz, 2H), 7.74 (d, J = 8.22 Hz, 2H), 3.62 - 3.78 (m, 1 H), 3.43 - 3.53 (m, 1 H). LCMS: fR = 0.52 min, 98 = 244 (M+H)+.
Figure imgf000033_0002
4-(Pyridin-3-yl)-A -(4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)benzyl)pyrimidin-2- amine, trifluroacetic acid salt: To a suspension of 4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol- 3-yl)phenyl)methanamine (70 mg, 0.29 mmol), 2-chloro-4-(pyridin-3-yl)pyrimidine (50 mg, 0.26 mmol) in A/JV-dimethylacetamide (2.5 ml.) was added a solution of Et3N (0.091 ml_, 0.652 mmol) in one charge. The reaction vessel was sealed and heated to 160 °C under microwave irradiation for 10 min. After cooling the reaction, the solvent was removed under reduced pressure, and the mixture was dissolved in MeOH and purified via reverse phase prep-HPLC (10% - 100% MeCN in water with TFA 0.05%) to give a solid. The solid obtained was triturated with Et20 to provide the title compound (24 mg, 0.047 mmol, 17.95 % yield) as a white powder. 1H NMR (400 MHz, METHANOL-^) δ ppm 9.31 (br. s., 1 H), 8.76 (d, J = 5.1 Hz, 2 H), 8.45 (d, J = 5.3 Hz, 1 H), 8.09 (d, J = 8.2 Hz, 2 H), 7.74 - 7.89 (m, 1 H), 7.62 (d, J = 8.0 Hz, 2 H), 7.30 (d, J = 5.3 Hz, 1 H), 4.80 (s, 2 H). LCMS: f, = 0.82 min, 98%. MS m/z = 399 (M+H)+.
Histone Deacetylase 9 (HDAC9) Inhibition Assay:
Novel histone deacetylase 9 (HDAC9) inhibitors were characterized in an in vitro biochemical functional assay. The assay measures the increased fluorescent signal due to deacetylation, by HDAC9, of a fluorogenic substrate. The commercial available substrate is Class lla HDAC-specific and contains an acetylated lysine residue and would releases the fluorescent signal upon trypsin cleavage after deacetylation.
Specifically, test compounds diluted to various concentrations in 100% DMSO are first dispensed into 384-well assay plates. Recombinant HDAC9 isoform 4 (purchased from BPS Bioscience) in complete assay buffer (50 mM Tris-HCI, pH 8.0, 137 mM NaCI, 2.7 mM KCI, 1 mM MgCI2, 0.05% BSA & 0.005% Tween 20) were then added to each well (5uL/well) using Multidrop Combi (Thermo Scientific), followed by 5 uL/well substrate (purchased from BPS Bioscience, 4.5 uM final). After 45 minutes incubation at room temperature, 10uL 2x developer solution (assay buffer with 40 uM Trypsin and 20 uM Trichostatin A) was added. The plates were then incubated 1 hour at room temperature before reading in fluorescent intensity mode at 450nm in an Envision (Perkin Elmer) plate reader. Percent Inhibition of HDAC9 activity by compounds in each test wells was calculated by normalizing to fluorescent signal in control wells containing DMSO only. The plC50s value of test compounds were calculated from non-linear curve fitting, using ActivityBase5 data analysis tool (IDBS), from 1 1 point 3x dilution series starting from 100 uM final compound concentration.
For concentration/dose response experiments, normalized data were fit and plC50s determined using conventional techniques. The plC50s are averaged to determine a mean value, for a minimum of 2 experiments. As determined using the above method, the compounds of Examples 1-3 exhibited a plC5o between 5.0 and 9.0 e.g., for example, the compound of Example 2 inhibited HDAC9 in the above method with a mean plC50 >6. References:
US 20060269559, US Patent No. 7,521 ,044, WO2007084775
"Deacetylase inhibition promotes the generation and function of regulatory T cells," R.Tao, E. F. de Zoeten, E. O' zkaynak, C. Chen, L. Wang, P. M. Porrett, B. Li, L. A. Turka, E. N. Olson, M. I. Greene, A. D. Wells, W. W. Hancock, Nature Medicine, 13 (1 1 ), 2007.
"Expression of HDAC9 by T Regulatory Cells Prevents Colitis in Mice," E. F. de
Zoeten, L. Wang, H. Sai, W. H. Dillmann, W. W. Hancock, Gastroenterology. 2009 Oct 28. "Immunomodulatory effects of deacetylase inhibitors: therapeutic targeting of FOXP3+ regulatory T cells," L. Wang, E. F. de Zoeten, M. I. Greene and W. W. Hancock, Nature Review Drug Discovery. 8(12):969-81 , 2009 and references therein.
"HDAC7 targeting enhances FOXP3+ Treg function and induces long-term allograft survival," L. Wang, et al., Am. J. Transplant 9, S621 (2009).
"Selective class II HDAC inhibitors impair myogenesis by modulating the stability and activity of HDAC-MEF2 complexes," A. Nebbioso, F. Manzo, M. Miceli, M. Conte, L. Manente, A. Baldi, A. De Luca, D. Rotili, S. Valente, A. Mai, A. Usiello, H. Gronemeyer, L. Altucci, EMBO reports 10 (7), 776-782, 2009, and references therein.
"Myocyte Enhancer Factor 2 and Class II Histone Deacetylases Control a Gender-
Specific Pathway of Cardioprotection Mediated by the Estrogen Receptor," E. van Rooij, J. Fielitz, L. B. Sutherland, V. L. Thijssen, H. J. Crijns, M. J. Dimaio, J. Shelton, L. J. De Windt, J. A. Hill, E.N. Olson, Circulation Research, Jan 2010.

Claims

CLAIMS What is claimed is:
1. A compound according to Formula (I):
Figure imgf000036_0001
wherein:
-6cycloalkyl, 4-7 membered heterocycloalkyi, phenyl, naphthyl, 5-6 membered heteroaryl, or 9-10 membered heteroaryl, wherein said C3-6cycloalkyl, 4-7 membered heterocycloalkyi, phenyl, naphthyl, 5-6 membered heteroaryl, or 9-10 membered heteroaryl is optionally substituted by 1-3 groups each independently selected from the group consisting of
Figure imgf000036_0002
Ci-4alkoxy, halo
Figure imgf000036_0003
where R' and R" are
independently selected from the group consisting of H and Ci-4alkyl;
n is 0, 1 , 2, 3 or 4;
R1 is H or C1-4alkyl;
m is 0, 1 , 2, 3 or 4; and
B is C3-6cycloalkyl, 4-7 membered heterocycloalkyi, phenyl, naphthyl, 5-6 membered heteroaryl, or 9-10 membered heteroaryl,
wherein said C3-6cycloalkyl, 4-7 membered heterocycloalkyi, phenyl, naphthyl, 5-6 membered heteroaryl, or 9-10 membered heteroaryl is optionally substituted by 1 , 2 or 3 RB groups each independently selected from the group consisting of C1-4alkyl, C1-4alkoxy, aryl, arylC1-4alkyl-, 4-7 membered heterocycloalkyi, 4-7 membered
heterocycloalkylCi-4alkyl-, 5-6 membered he
9-10 membered heteroaryl, 9-10 membered
Figure imgf000036_0004
haloCi-4alkoxy, hydroxyCi-4alkyl, Ci-4alkylsulfanyl, (Ci-4alkyl)(Ci-4alkyl)N-C2-C4alkoxy, and-C0-4alkyl-NRBaRBb, wherein RBa and R Bb are independently selected from the group consisting of H, Ci-4alkyl, aryl, 4-7 membered heterocycloalkyi, 5-6 membered heteroaryl, arylCi-4alkyl-, 5-6 membered heteroarylCi-4alkyl-, and 4-7 membered
heterocycloalkylCi-4alkyl-, wherein any of said aryl, heterocycloalkyl or heteroaryl is optionally substituted by a substituent selected from the group consisting of 4-7 membered heterocycloalkyl, C1-4alklylsulfanyl, Ci-4alkyl, halogen, haloCi-4alkyl, C-|-4alkoxy, C1-4 alkylsulfanyl, haloCr4alkoxy, cyano, Ci-4alkoxyC2-4alkoxyCi-4 alkyl, aminoC2-6alkoxy,
Ci-C6alkylaminoC2-6alkoxy, (Ci-6alkyl)(Ci-6alkyl)aminoC2-6alkoxy, hydroxyl,
Figure imgf000037_0001
4-7 membered
heterocycloalkylCi-4alkoxy, 4-7 membered heterocycloalkyl-carbonyl, -C0-4alkyl-NR'R" and -C(0)(CH2)pNR'R",
wherein: any heterocycloalkyl is optionally substituted by 1 , 2 or 3 groups each independently selected from the group consisting of Ci-4alkyl and Ci-4alkyl-CO-, R' and R" are independently selected from the group consisting of H and Ci-4alkyl, and p is 0-4; or a pharmaceutically-acceptable salt thereof
2. The compound, or salt thereof, according to claim 1 wherein A is a phenyl group optionally substituted by 1-2 groups independently selected from the group consisting of
Ci-4alkyl, halogen, cyano, haloC-M alkyl, C-^alkoxy, halo C-^alkoxy, -NR'R" and
-C^alkylNR'R".
3. The compound, or salt thereof, according to claim 1 wherein A is a phenyl group optionally substituted by 1 group selected from the group consisting of methyl, ethyl, fluoro, chloro, trifluoromethyl, methoxy, ethoxy, trifluoromethoxy, cyano, -NR'R" and
Figure imgf000037_0002
where R' and R" are independently selected from the group consisting of H and methyl.
4. The compound, or salt thereof, according to claim 1 wherein A is an unsubstituted phenyl group or a phenyl group substituted by an ethyl, fluoro, cyano or methoxy group.
5. The compound, or salt thereof, according to claim 1 wherein A is a cyclopropyl, cyclopentyl or cyclohexyl group, optionally substituted by 1-2 groups independently selected from the group consisting of Ci-4alkyl,
Figure imgf000037_0003
6. The compound, or salt thereof, according to claim 1 wherein A is naphthyl optionally substituted by 1-2 groups independently selected from the group consisting of Ci-4alkyl, halogen, cyano,
Figure imgf000037_0004
Ci-4alkoxy, haloCi-4alkoxy, -NR'R" and - Ci-4alkylNR'R".
7. The compound, or salt thereof, according to claim 1 wherein A is a 4-7 membered heterocycloalkyi group optionally substituted by 1-3 groups independently selected from the group consisting of C1-4alkyl, halogen, cyano, haloC1-4alkyl, C1-4alkoxy, haloC1-4alkoxy, oxo, -NR'R" and -Ci-4NR'R".
8. The compound, or salt thereof, according to claim 1 wherein A is a 9-10 membered heteroaryl optionally substituted by 1 -2 groups independently selected from the group consisting of Ci-4alkyl, halogen, cyano, halo d^alkyl, d^alkoxy, halo
Ci-4alkoxy, oxo, -NR'R" and -Ci-4alkylNR'R".
9. The compound, or salt thereof, according to claim 1 wherein A is a 5-6 membered heteroaryl optionally substituted by 1 -2 groups independently selected from the group consisting of
Figure imgf000038_0001
haloCi-4alkoxy, -NR'R" and -Ci-4alkylNR'R".
10. The compound, or salt thereof, according to claim 1 wherein A is a pyrimidinyl or group optionally substituted by 1-2 groups independently selected from the group consisting of C1-4alkyl, halogen, cyano, halo C1-4alkyl, C1-4alkoxy, halo C1-4alkoxy, -NR'R" and -C1-4alkylNR'R".
1 1 . The compound, or salt thereof, according to claim 1 wherein A is a pyridinyl or group optionally substituted by 1-2 groups independently selected from the group consisting of
Figure imgf000038_0002
halo Ci-4alkoxy, -NR'R" and -C^alkylNR'R".
12. The compound, or salt thereof, according to claim 1 having the Formula (II):
Figure imgf000038_0003
wherein D and E are each independently selected from the group consisting of NH and CH.
13. The compound, or salt thereof, according to any one of claims 1 -12 wherein n is 0.
14. The compound, or salt thereof, according to any one of claims 1 -12 wherein n is 1.
15. The compound, or salt thereof, according to any one of claims 1 -12 wherein n is 2.
16. The compound, or salt thereof, according to any one of claims 1 -12 wherein n is 3.
17. The compound, or salt thereof, according to any one of claims 1 -12 wherein n is 4.
18. The compound, or salt thereof, according to any one of claims 1-17 wherein m is 0.
19. The compound, or salt thereof, according to any one of claims 1-17 wherein m is 1.
20. The compound, or salt thereof, according to any one of claims 1-17 wherein m is 2.
21 . The compound, or salt thereof, according to any one of claims 1-17 wherein m is 3.
22. The compound, or salt thereof, according to any one of claims 1-17 wherein m is 4.
23. The compound, or salt thereof, according to any one of claims 1-22, wherein
B is a cyclohexyl, phenyl, pyridinyl, pyrimidinyl, imidazolyl, thiazolyl, oxadiazolyl, quinolyl, pyrazinyl, or fluorenyl, wherein said cyclohexyl, phenyl, pyridinyl, pyrimidinyl, imidazolyl, thiazolyl, oxadiazolyl, quinolyl, pyrazinyl, or fluorenyl is optionally substituted by 1 or 2 RB groups, each independently selected from the group consisting of d^alkyl,
Ci-4alkylsulfanyl, phenyl, 5-6 membered heteroaryl, and -N RBaRBb (where RBa and RBb are independently selected from the group consisting of H, d^alkyl, phenyl, and 5-6 membered heteroaryl), wherein any of said phenyl or 5-6 membered heteroaryl of RB, RBa and RBb is optionally substituted with a substituent selected from the group consisting of 4-7 membered heterocycloalkyl,
Figure imgf000039_0001
Ci-4alklylsulfanyl, cyano, C1-4alkoxyC2-4alkoxyC1-4alkyl, hydroxyC1-4alkyl, heterocycloalkylC1-4alkyl, heterocycloalkylC1-4alkoxy, heterocycloalkyl-carbonyl-, and -C(0)(CH2)pNR'R" where p is 0-4 and R' and R" are independently selected from the group consisting of H and
C1-4alkyl.
24. The compound, or salt thereof, according to any one of claims 1 -22, wherein B is a 4, 5, 6 or 7 membered heterocycloalkyl containing 1 or 2 nitrogen atoms, optionally substituted by 1 or 2 RB groups, each independently selected from the group consisting of Ci-4alkyl, C1-4 alkylsulfanyl, phenyl, 5-6 membered heteroaryl, and -NRBaRBb (where RBa and RBb are independently selected from the group consisting of H, Ci-4alkyl, phenyl, and 5-6 membered heteroaryl), wherein said phenyl or 5-6 membered heteroaryl of RB, RBa and RBb is optionally substituted with a substituent selected from the group consisting of 4-7 membered heterocycloalkyl,
Figure imgf000040_0001
cyano, Ci-4alkoxyC2-4alkoxyCi-4alkyl, hydroxyCi-4alkyl, 4-7 membered
Figure imgf000040_0002
4-7 membered heterocycloalkylCi-4alkoxy, 4-7 membered heterocycloalkyl-carbonyl, and -C(0)(CH2)pNR'R" where p is 0-4 and R' and R" are independently selected from the group consisting of H and
Figure imgf000040_0003
25. The compound, or salt thereof, according to any one of claims 1 -22 wherein B is pyrimidinyl, phenyl, pyridinyl, or pyrizinyl, wherein said pyrimidinyl, phenyl, pyridinyl, or pyrizinyl is optionally substituted with 1 or 2 RB groups, each independently selected from the group consisting of C1-4 alkylsulfanyl, -NH2, phenyl, pyrimindinyl, thienyl, thiazolyl, pyrazolyl, and pyridinyl, wherein said phenyl, pyrimindinyl, thiophenyl, thiazolyl, pyrazolyl, or pyridinyl is optionally substituted with morpholinyl, methylpiperazinyl, Ci-4alkylsulfanyl, cyano, methyoxymethoxypropan-2-yl, hydroxyCi-4alkyl, morpholinylCi-4alkyl,
Figure imgf000040_0004
morpholinylcarbonyl, or -C(0)(CH2) NR'R" where p is 0, 1 , 2, 3, or 4 and R' and R" are independently selected from the group consisting of H and Ci-4alkyl.
26. The compound, or salt thereof, according to any one of claims 1 -22 wherein B is phenyl, pyridinyl, pyrazinyl, or pyrimidinyl substituted by 1 ,2-oxazol-3-yl, 1 H-pyrazol-5-yl, 1 ,3-thiazol-2-yl, thiophen-3-yl, wherein said 1 ,2-oxazol-3-yl, 1 H-pyrazol-5-yl,
1 ,3-thiazol-2-yl, thiophen-3-yl may be optionally substituted with Ci-4alkyl or
Figure imgf000040_0005
27. The compound, or salt thereof, according to any one of claims 1 -22 wherein B is:
Figure imgf000041_0001
wherein at least 1 of D, E, and F is CH and each remaining group is independently selected from the group consisting of CH and N,
R1 is selected from the group consisting of H, methylsulfanyl, and NH2,
G and H are each independently selected from the group consisting of CH and N, and R2 is selected from the group consisting of H, methylsulfanyl, cyano, methoxymethyoxypropan-2-yl, 1 -hydroxy-1-methylethyl, morpholinyl, methylpiperazinyl, morpholinylcarbonyl, morpholinylmethyl, morpholinylethyloxy, dimethylaminocarbonyl, and dimethylaminoethoxy.
28. The compound according to claim 1 , selected from the group consisting of:
Λ/-(2-(1 -Methyl-1 H-indol-3-yl)ethyl)-5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3- yl)pyrimidin-2-amine;
Λ/-((1 -Methyl-1 H-indol-3-yl)methyl)-5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3- yl)pyrimidin-2-amine;
4-(Pyridin-3-yl)-/V-(4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)benzyl)pyrimidin-2- amine;
and a pharmaceutically acceptable salt thereof.
29. The compound according to claim 1 selected from the group consisting of:
4-(pyridin-3-yl)-N-(4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)benzyl)pyrimidin-2- amine;
4-(pyridin-3-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyridin-2- yl)methyl)pyrimidin-2-amine;
4-(pyridin-3-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyridin-2- yl)methyl)pyrimidin-2-amine;
3-(pyridin-3-yl)-N-(4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)benzyl)aniline;
N-(4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)benzyl)-[2,3'-bipyridin]-6-amine;
2-(pyridin-3-yl)-N-(4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)benzyl)pyrimidin-4- amine; 6-(pyridin-3-yl)-N-(4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)benzyl)pyrimidin-4- amine;
6-(pyridin-3-yl)-N-(4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)benzyl)pyrazin-2- amine;
2- (methylthio)-6-(pyridin-3-yl)-N-(4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3- yl)benzyl)pyrimidin-4-amine;
6-(pyridin-3-yl)-N2-(4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)benzyl)pyrimidine- 2,4-diamine;
3- (pyridin-3-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyridin-2- yl)methyl)aniline;
N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyridin-2-yl)methyl)-[2,3'-bipyridin]-6- amine;
2-(pyridin-3-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyridin-2- yl)methyl)pyrimidin-4-amine;
6-(pyridin-3-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyridin-2- yl)methyl)pyrimidin-4-amine;
6-(pyridin-3-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyridin-2- yl)methyl)pyrazin-2-amine;
2- (methylthio)-6-(pyridin-3-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3- yl)pyridin-2-yl)methyl)pyrimidin-4-amine;
6-(pyridin-3-yl)-N2-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyridin-2- yl)methyl)pyrimidine-2,4-diamine;
3- (pyridin-3-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyrimidin-2- yl)methyl)aniline;
N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyrimidin-2-yl)methyl)-[2,3'-bipyridin]- 6-amine;
2-(pyridin-3-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyrimidin-2- yl)methyl)pyrimidin-4-amine;
6-(pyridin-3-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyrimidin-2- yl)methyl)pyrimidin-4-amine;
6-(pyridin-3-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyrimidin-2- yl)methyl)pyrazin-2-amine;
2-(methylthio)-6-(pyridin-3-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3- yl)pyrimidin-2-yl)methyl)pyrimidin-4-amine;
6-(pyridin-3-yl)-N2-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyrimidin-2- yl)methyl)pyrimidine-2,4-diamine; 4-(6-morpholinopyridin-3-yl)-N-(4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3- yl)benzyl)pyrimidin-2-amine;
4-(6-(4-methylpiperazin-1-yl)pyridin-3-yl)-N-(4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol- 3-yl)benzyl)pyrimidin-2-amine;
4- (6-(methylthio)pyridin-3-yl)-N-(4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3- yl)benzyl)pyrimidin-2-amine;
5- (2-((4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)benzyl)amino)pyrimidin-4- yl)picolinonitrile;
4-(6-(2-(methoxymethoxy)propan-2-yl)pyridin-3-yl)-N-(4-(5-(trifluoromethyl)-1 ,2,4- oxadiazol-3-yl)benzyl)pyrimidin-2-amine;
2-(5-(2-((4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)benzyl)amino)pyrimidin-4- yl)pyridin-2-yl)propan-2-ol;
N,N-dimethyl-5-(2-((4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3- yl)benzyl)amino)pyrimidin-4-yl)picolinamide;
morpholino(5-(2-((4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3- yl)benzyl)amino)pyrimidin-4-yl)pyridin-2-yl)methanone;
4-(6-morpholinopyridin-3-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyridin-
2- yl)methyl)pyrimidin-2-amine;
4-(6-(4-methylpiperazin-1-yl)pyridin-3-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol
3- yl)pyridin-2-yl)methyl)pyrimidin-2-amine;
4- (6-(methylthio)pyridin-3-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyridin
2- yl)methyl)pyrimidin-2-amine;
5- (2-(((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyridin-2- yl)methyl)amino)pyrimidin-4-yl)picolinonitrile;
4-(6-(2-(methoxymethoxy)propan-2-yl)pyridin-3-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4- oxadiazol-3-yl)pyridin-2-yl)methyl)pyrimidin-2-amine;
2-(5-(2-(((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyridin-2- yl)methyl)amino)pyrimidin-4-yl)pyridin-2-yl)propan-2-ol;
N,N-dimethyl-5-(2-(((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyridin-2- yl)methyl)amino)pyrimidin-4-yl)picolinamide;
morpholino(5-(2-(((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyridin-2- yl)methyl)amino)pyrimidin-4-yl)pyridin-2-yl)methanone;
4-(6-morpholinopyridin-3-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3- yl)pyrimidin-2-yl)methyl)pyrimidin-2-amine;
4-(6-(4-methylpiperazin-1-yl)pyridin-3-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol
3- yl)pyrimidin-2-yl)methyl)pyrimidin-2-amine; 4- (6-(methylthio)pyridin-3-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3- yl)pyrimidin-2-yl)methyl)pyrimidin-2-amine;
5- (2-(((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyrimidin-2- yl)methyl)amino)pyrimidin-4-yl)picolinonitrile;
4-(6-(2-(methoxymethoxy)propan-2-yl)pyridin-3-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4 oxadiazol-3-yl)pyrimidin-2-yl)methyl)pyrimidin-2-amine;
2-(5-(2-(((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyrimidin-2- yl)methyl)amino)pyrimidin-4-yl)pyridin-2-yl)propan-2-ol;
N,N-dimethyl-5-(2-(((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyrimidin-2- yl)methyl)amino)pyrimidin-4-yl)picolinamide;
morpholino(5-(2-(((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyrimidin-2- yl)methyl)amino)pyrimidin-4-yl)pyridin-2-yl)methanone;
4-(pyrazin-2-yl)-N-(4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)benzyl)pyrimidin-2- amine;
4-(thiophen-3-yl)-N-(4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)benzyl)pyrimidin-2- amine;
4-(thiazol-2-yl)-N-(4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)benzyl)pyrimidin-2- amine;
4-(1 H-pyrazol-5-yl)-N-(4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)benzyl)pyrimidin 2-amine;
(5-(2-((4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)benzyl)amino)pyrimidin-4- yl)isoxazol-3-yl)methanol;
4-(4-(morpholinomethyl)phenyl)-N-(4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3- yl)benzyl)pyrimidin-2-amine;
4-(4-(2-morpholinoethoxy)phenyl)-N-(4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3- yl)benzyl)pyrimidin-2-amine;
4-(3-(2-morpholinoethoxy)phenyl)-N-(4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3- yl)benzyl)pyrimidin-2-amine;
4-(4-(2-(dimethylamino)ethoxy)phenyl)-N-(4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3 yl)benzyl)pyrimidin-2-amine;
4-(3-(2-(dimethylamino)ethoxy)phenyl)-N-(4-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3 yl)benzyl)pyrimidin-2-amine;
4-(pyrazin-2-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyridin-2- yl)methyl)pyrimidin-2-amine;
4-(thiophen-3-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyridin-2- yl)methyl)pyrimidin-2-amine; 4-(thiazol-2-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyridin-2- yl)methyl)pyrimidin-2-amine;
4-(1 H-pyrazol-5-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyridin-2- yl)methyl)pyrimidin-2-amine;
(5-(2-(((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyridin-2- yl)methyl)amino)pyrimidin-4-yl)isoxazol-3-yl)methanol;
4-(4-(morpholinomethyl)phenyl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3- yl)pyridin-2-yl)methyl)pyrimidin-2-amine;
4-(4-(2-morpholinoethoxy)phenyl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3- yl)pyridin-2-yl)methyl)pyrimidin-2-amine;
4-(3-(2-morpholinoethoxy)phenyl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3- yl)pyridin-2-yl)methyl)pyrimidin-2-amine;
4-(4-(2-(dimethylamino)ethoxy)phenyl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3 yl)pyridin-2-yl)methyl)pyrimidin-2-amine;
4-(3-(2-(dimethylamino)ethoxy)phenyl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3 yl)pyridin-2-yl)methyl)pyrimidin-2-amine;;
4-(pyrazin-2-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyrimidin-2- yl)methyl)pyrimidin-2-amine;
4-(thiophen-3-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyrimidin-2- yl)methyl)pyrimidin-2-amine;
4-(thiazol-2-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyrimidin-2- yl)methyl)pyrimidin-2-amine;
4-(1 H-pyrazol-5-yl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyrimidin-2- yl)methyl)pyrimidin-2-amine;
(5-(2-(((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3-yl)pyrimidin-2- yl)methyl)amino)pyrimidin-4-yl)isoxazol-3-yl)methanol;
4-(4-(morpholinomethyl)phenyl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3- yl)pyrimidin-2-yl)methyl)pyrimidin-2-amine;
4-(4-(2-morpholinoethoxy)phenyl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3- yl)pyrimidin-2-yl)methyl)pyrimidin-2-amine;
4-(3-(2-morpholinoethoxy)phenyl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3- yl)pyrimidin-2-yl)methyl)pyrimidin-2-amine;
4-(4-(2-(dimethylamino)ethoxy)phenyl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3 yl)pyrimidin-2-yl)methyl)pyrimidin-2-amine;
4-(3-(2-(dimethylamino)ethoxy)phenyl)-N-((5-(5-(trifluoromethyl)-1 ,2,4-oxadiazol-3 yl)pyrimidin-2-yl)methyl)pyrimidin-2-amine; and a pharmaceutically acceptable salt, thereof.
30. A pharmaceutical composition comprising the compound, or salt thereof, according to any one of claims 1-29 and a pharmaceutical carrier.
31 . A method of treating a subject having a disease or disorder mediated by HDAC, wherein said method comprises comprising administering to the subject the compound, or salt thereof, according to any one of claims 1-29.
32. Use of the compound, or salt thereof, according to any one of claims 1 -29 in the preparation of a medicament for the treatment of an HDAC-mediated disease or disorder.
PCT/US2012/062514 2011-10-31 2012-10-30 Compounds and methods WO2013066839A2 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201161553757P 2011-10-31 2011-10-31
US61/553,757 2011-10-31
US201261596269P 2012-02-08 2012-02-08
US61/596,269 2012-02-08

Publications (2)

Publication Number Publication Date
WO2013066839A2 true WO2013066839A2 (en) 2013-05-10
WO2013066839A3 WO2013066839A3 (en) 2015-06-25

Family

ID=48193011

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2012/062514 WO2013066839A2 (en) 2011-10-31 2012-10-30 Compounds and methods

Country Status (1)

Country Link
WO (1) WO2013066839A2 (en)

Cited By (48)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140221366A1 (en) * 2011-07-07 2014-08-07 Merck Patent Gmbh Substituted Azaheterocycles for the Treatment of Cancer
US9056843B2 (en) 2011-07-08 2015-06-16 Novartis Ag Trifluoromethyl-oxadiazole derivatives and their use in the treatment of disease
WO2016031815A1 (en) * 2014-08-26 2016-03-03 武田薬品工業株式会社 Heterocyclic compound
CN106243083A (en) * 2016-07-12 2016-12-21 浙江宏冠生物药业有限公司 A kind of imatinib preparation method
WO2017014170A1 (en) * 2015-07-17 2017-01-26 武田薬品工業株式会社 Heterocyclic compound
KR20170013187A (en) 2015-07-27 2017-02-06 주식회사 종근당 1,3,4-Oxadiazole Amide Derivative Compounds as Histone Deacetylase 6 Inhibitor, and the Pharmaceutical Composition Comprising the same
KR20170013186A (en) 2015-07-27 2017-02-06 주식회사 종근당 1,3,4-Oxadiazole Sulfamide Derivative Compounds as Histone Deacetylase 6 Inhibitor, and the Pharmaceutical Composition Comprising the same
KR20170013836A (en) 2015-07-27 2017-02-07 주식회사 종근당 1,3,4-Oxadiazole Sulfonamide Derivative Compounds as Histone Deacetylase 6 Inhibitor, and the Pharmaceutical Composition Comprising the same
KR20170017792A (en) 2015-08-04 2017-02-15 주식회사 종근당 1,3,4-Oxadiazole Derivative Compounds as Histone Deacetylase 6 Inhibitor, and the Pharmaceutical Composition Comprising the same
WO2017055473A1 (en) 2015-10-02 2017-04-06 Syngenta Participations Ag Microbiocidal oxadiazole derivatives
WO2017055469A1 (en) * 2015-10-02 2017-04-06 Syngenta Participations Ag Microbiocidal oxadiazole derivatives
WO2017103219A1 (en) * 2015-12-17 2017-06-22 Syngenta Participations Ag Microbiocidal oxadiazole derivatives
WO2017103223A1 (en) * 2015-12-18 2017-06-22 Syngenta Participations Ag Microbiocidal oxadiazole derivatives
WO2017178245A1 (en) 2016-04-11 2017-10-19 Basf Se Substituted oxadiazoles for combating phytopathogenic fungi
WO2017222951A1 (en) 2016-06-23 2017-12-28 Merck Sharp & Dohme Corp. 3-aryl and heteroaryl substituted 5-trifluoromethyl oxadiazoles as histone deacetylase 6 (hdac6) inhibitors
US10064869B2 (en) 2014-06-04 2018-09-04 Thomas Helledays Stiftelse For Medicinsk Forskning MTH1 inhibitors for treatment of inflammatory and autoimmune conditions
WO2018177880A1 (en) 2017-03-31 2018-10-04 Syngenta Participations Ag Fungicidal compositions
WO2018177894A1 (en) 2017-03-31 2018-10-04 Syngenta Participations Ag Fungicidal compositions
WO2018184985A1 (en) 2017-04-05 2018-10-11 Syngenta Participations Ag Microbiocidal oxadiazole derivatives
WO2018184986A1 (en) 2017-04-05 2018-10-11 Syngenta Participations Ag Microbiocidal oxadiazole derivatives
WO2018184984A1 (en) 2017-04-05 2018-10-11 Syngenta Participations Ag Microbiocidal oxadiazole derivatives
WO2018184982A1 (en) 2017-04-05 2018-10-11 Syngenta Participations Ag Microbiocidal oxadiazole derivatives
WO2018184988A1 (en) 2017-04-05 2018-10-11 Syngenta Participations Ag Microbiocidal oxadiazole derivatives
WO2018187553A1 (en) * 2017-04-06 2018-10-11 Fmc Corporation Fungicidal oxadiazoles
WO2018184987A1 (en) 2017-04-05 2018-10-11 Syngenta Participations Ag Microbiocidal oxadiazole derivatives
WO2018185013A1 (en) 2017-04-03 2018-10-11 Syngenta Participations Ag Microbiocidal oxadiazole derivatives
CN109071522A (en) * 2016-04-12 2018-12-21 先正达参股股份有限公司 Kill the oxadiazole derivatives of microorganism
US10179790B2 (en) 2014-06-04 2019-01-15 Thomas Helledays Stiftelse For Medicinsk Forskning MTH1 inhibitors for treatment of cancer
US10308643B2 (en) 2015-07-17 2019-06-04 Takeda Pharmaceutical Company Limited Heterocyclic compound
RU2695133C1 (en) * 2015-10-12 2019-07-22 Чонг Кун Данг Фармасьютикал Корп. Oxadiazolamine derivatives as histone deacetylase 6 inhibitor and pharmaceutical composition containing them
US10406146B2 (en) 2015-08-25 2019-09-10 Takeda Pharmaceutical Company Limited Heterocyclic compound
US10435399B2 (en) 2017-07-31 2019-10-08 Takeda Pharmaceutical Company Limited HDAC6 inhibitory heterocyclic compound
WO2019207062A1 (en) 2018-04-26 2019-10-31 Syngenta Participations Ag Microbiocidal oxadiazole derivatives
US10499644B2 (en) 2015-11-19 2019-12-10 Basf Se Substituted oxadiazoles for combating phytopathogenic fungi
US10555526B2 (en) 2015-11-05 2020-02-11 Basf Se Substituted oxadiazoles for combating phytopathogenic fungi
US10640497B2 (en) 2015-12-02 2020-05-05 Syngenta Participations Ag Microbiocidal oxadiazole derivatives
US10674727B2 (en) 2015-11-19 2020-06-09 Basf Se Substituted oxadiazoles for combating phytopathogenic fungi
CN111285861A (en) * 2018-12-10 2020-06-16 浙江省化工研究院有限公司 Trifluoromethyl oxadiazole substituted pyrimidinamine compound, and preparation method and application thereof
US10785980B2 (en) 2016-06-09 2020-09-29 Basf Se Substituted oxadiazoles for combating phytopathogenic fungi
US10798941B2 (en) 2016-01-08 2020-10-13 Syngenta Participations Ag Microbiocidal oxadiazole derivatives
WO2020212513A1 (en) 2019-04-18 2020-10-22 Syngenta Crop Protection Ag Process for the preparation of microbiocidal oxadiazole derivatives
WO2020240493A1 (en) 2019-05-31 2020-12-03 Chong Kun Dang Pharmaceutical Corp. 1,3,4-oxadiazole homophthalimide derivative compounds as histone deacetylase 6 inhibitor, and the pharmaceutical composition comprising the same
WO2020254494A1 (en) * 2019-06-21 2020-12-24 Bayer Aktiengesellschaft Fungicidal oxadiazoles
JP2021505684A (en) * 2017-12-05 2021-02-18 オリソン ヘノミクス,ソシエダ アノニマ 1,2,4-oxadiazole derivative as a histone deacetylase 6 inhibitor
US11083196B2 (en) 2016-03-24 2021-08-10 Syngenta Participations Ag Microbiocidal oxadiazole derivatives
US11453661B2 (en) 2019-09-27 2022-09-27 Takeda Pharmaceutical Company Limited Heterocyclic compound
US11938134B2 (en) 2017-03-10 2024-03-26 Eikonizo Therapeutics, Inc. Metalloenzyme inhibitor compounds
US11958844B2 (en) 2018-07-26 2024-04-16 Chong Kun Dang Pharmaceutical Corp. 1,3,4-oxadiazole derivative compounds as histone deacetylase 6 inhibitor, and pharmaceutical composition comprising the same

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP4383177B2 (en) * 2002-03-01 2009-12-16 スミスクライン ビーチャム コーポレーション hPPAR activator
JP2009509987A (en) * 2005-09-29 2009-03-12 サノフィ−アベンティス 2-Aminothiazole and 2-aminooxazole derivatives, processes for their preparation and their use as pharmaceuticals
ES2748599T3 (en) * 2007-08-27 2020-03-17 Dart Neuroscience Cayman Ltd Isoxazole therapeutic compounds
WO2011088192A1 (en) * 2010-01-13 2011-07-21 Tempero Pharmaceuticals, Inc. Compounds and methods

Cited By (88)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2012280725B2 (en) * 2011-07-07 2017-02-02 Merck Patent Gmbh Substituted azaheterocycles for the treatment of cancer
US9199962B2 (en) * 2011-07-07 2015-12-01 Merck Patent Gmbh Substituted azaheterocycles for the treatment of cancer
US20140221366A1 (en) * 2011-07-07 2014-08-07 Merck Patent Gmbh Substituted Azaheterocycles for the Treatment of Cancer
US9056843B2 (en) 2011-07-08 2015-06-16 Novartis Ag Trifluoromethyl-oxadiazole derivatives and their use in the treatment of disease
US10632125B2 (en) 2014-06-04 2020-04-28 Thomas Helledays Stiftelse For Medicinsk Forskning MTH1 inhibitors for treatment of inflammatory and autoimmune conditions
US10179790B2 (en) 2014-06-04 2019-01-15 Thomas Helledays Stiftelse For Medicinsk Forskning MTH1 inhibitors for treatment of cancer
US10064869B2 (en) 2014-06-04 2018-09-04 Thomas Helledays Stiftelse For Medicinsk Forskning MTH1 inhibitors for treatment of inflammatory and autoimmune conditions
JPWO2016031815A1 (en) * 2014-08-26 2017-06-15 武田薬品工業株式会社 Heterocyclic compounds
US10081624B2 (en) 2014-08-26 2018-09-25 Takeda Pharmaceutical Company Limited Heterocyclic compound
WO2016031815A1 (en) * 2014-08-26 2016-03-03 武田薬品工業株式会社 Heterocyclic compound
WO2017014170A1 (en) * 2015-07-17 2017-01-26 武田薬品工業株式会社 Heterocyclic compound
US10308643B2 (en) 2015-07-17 2019-06-04 Takeda Pharmaceutical Company Limited Heterocyclic compound
US10357484B2 (en) 2015-07-17 2019-07-23 Takeda Pharmaceutical Company Limited Heterocyclic compound
KR20170013186A (en) 2015-07-27 2017-02-06 주식회사 종근당 1,3,4-Oxadiazole Sulfamide Derivative Compounds as Histone Deacetylase 6 Inhibitor, and the Pharmaceutical Composition Comprising the same
KR20170013836A (en) 2015-07-27 2017-02-07 주식회사 종근당 1,3,4-Oxadiazole Sulfonamide Derivative Compounds as Histone Deacetylase 6 Inhibitor, and the Pharmaceutical Composition Comprising the same
KR20170013187A (en) 2015-07-27 2017-02-06 주식회사 종근당 1,3,4-Oxadiazole Amide Derivative Compounds as Histone Deacetylase 6 Inhibitor, and the Pharmaceutical Composition Comprising the same
US10584117B2 (en) 2015-07-27 2020-03-10 Chong Kun Dang Pharmaceutical Corp. 1,3,4-oxadiazole amide derivative compound as histone deacetylase 6 inhibitor, and pharmaceutical composition containing same
US10538498B2 (en) 2015-07-27 2020-01-21 Chong Kun Dang Pharmaceutical Corp. 1,3,4-oxadiazole sulfonamide derivative compounds as histone deacetylase 6 inhibitor, and the pharmaceutical composition comprising the same
US10464911B2 (en) 2015-07-27 2019-11-05 Chong Kun Dang Pharmaceutical Corp. 1,3,4-oxadiazole sulfamide derivative compounds as histone deacetylase 6 inhibitor, and the pharmaceutical composition comprising the same
KR20170017792A (en) 2015-08-04 2017-02-15 주식회사 종근당 1,3,4-Oxadiazole Derivative Compounds as Histone Deacetylase 6 Inhibitor, and the Pharmaceutical Composition Comprising the same
KR101799010B1 (en) * 2015-08-04 2017-11-17 주식회사 종근당 1,3,4-Oxadiazole Derivative Compounds as Histone Deacetylase 6 Inhibitor, and the Pharmaceutical Composition Comprising the same
JP2018523663A (en) * 2015-08-04 2018-08-23 チョン クン ダン ファーマシューティカル コーポレーション 1,3,4-oxadiazole derivative compound as histone deacetylase 6 inhibitor and pharmaceutical composition containing the same
RU2709207C2 (en) * 2015-08-04 2019-12-17 Чонг Кун Данг Фармасьютикал Корп. 1,3,4-oxadiazole derivatives of compound as inhibitor of histone deacetylase 6 and pharmaceutical composition containing same
WO2017023133A3 (en) * 2015-08-04 2017-03-23 Chong Kun Dang Pharmaceutical Corp. 1,3,4-oxadiazole derivative compounds as histone deacetylase 6 inhibitor, and the pharmaceutical composition comprising the same
US10717716B2 (en) 2015-08-04 2020-07-21 Chong Kun Dang Pharmaceutical Corp. 1,3,4-oxadiazole derivative compounds as histone deacetylase 6 inhibitor, and the pharmaceutical composition comprising the same
US10406146B2 (en) 2015-08-25 2019-09-10 Takeda Pharmaceutical Company Limited Heterocyclic compound
CN108137517A (en) * 2015-10-02 2018-06-08 先正达参股股份有限公司 Kill microorganism oxadiazole derivatives
WO2017055473A1 (en) 2015-10-02 2017-04-06 Syngenta Participations Ag Microbiocidal oxadiazole derivatives
WO2017055469A1 (en) * 2015-10-02 2017-04-06 Syngenta Participations Ag Microbiocidal oxadiazole derivatives
US11180462B2 (en) 2015-10-02 2021-11-23 Syngenta Participations Ag Microbiocidal oxadiazole derivatives
US10501425B2 (en) 2015-10-02 2019-12-10 Syngenta Participations Ag Microbiocidal oxadiazole derivatives
US11066375B2 (en) 2015-10-02 2021-07-20 Syngenta Participations Ag Microbiocidal oxadiazole derivatives
US10899724B2 (en) 2015-10-02 2021-01-26 Syngenta Participations Ag Microbiocidal oxadiazole derivatives
JP2018535935A (en) * 2015-10-02 2018-12-06 シンジェンタ パーティシペーションズ アーゲー Microbicidal oxadiazole derivatives
RU2695133C1 (en) * 2015-10-12 2019-07-22 Чонг Кун Данг Фармасьютикал Корп. Oxadiazolamine derivatives as histone deacetylase 6 inhibitor and pharmaceutical composition containing them
US10494355B2 (en) 2015-10-12 2019-12-03 Chong Kun Dang Pharmaceutical Corp. Oxadiazole amine derivative compounds as histone deacetylase 6 inhibitor, and the pharmaceutical composition comprising the same
US10555526B2 (en) 2015-11-05 2020-02-11 Basf Se Substituted oxadiazoles for combating phytopathogenic fungi
US10499644B2 (en) 2015-11-19 2019-12-10 Basf Se Substituted oxadiazoles for combating phytopathogenic fungi
US10674727B2 (en) 2015-11-19 2020-06-09 Basf Se Substituted oxadiazoles for combating phytopathogenic fungi
US10640497B2 (en) 2015-12-02 2020-05-05 Syngenta Participations Ag Microbiocidal oxadiazole derivatives
WO2017103219A1 (en) * 2015-12-17 2017-06-22 Syngenta Participations Ag Microbiocidal oxadiazole derivatives
WO2017103223A1 (en) * 2015-12-18 2017-06-22 Syngenta Participations Ag Microbiocidal oxadiazole derivatives
CN108368098A (en) * 2015-12-18 2018-08-03 先正达参股股份有限公司 Kill microorganism oxadiazole derivatives
US10798941B2 (en) 2016-01-08 2020-10-13 Syngenta Participations Ag Microbiocidal oxadiazole derivatives
US11259524B2 (en) 2016-01-08 2022-03-01 Syngenta Participations Ag Microbiocidal oxadiazole derivatives
US11083196B2 (en) 2016-03-24 2021-08-10 Syngenta Participations Ag Microbiocidal oxadiazole derivatives
WO2017178245A1 (en) 2016-04-11 2017-10-19 Basf Se Substituted oxadiazoles for combating phytopathogenic fungi
US10986839B2 (en) 2016-04-11 2021-04-27 Basf Se Substituted oxadiazoles for combating phytopathogenic fungi
CN109071522A (en) * 2016-04-12 2018-12-21 先正达参股股份有限公司 Kill the oxadiazole derivatives of microorganism
US10785980B2 (en) 2016-06-09 2020-09-29 Basf Se Substituted oxadiazoles for combating phytopathogenic fungi
WO2017222951A1 (en) 2016-06-23 2017-12-28 Merck Sharp & Dohme Corp. 3-aryl and heteroaryl substituted 5-trifluoromethyl oxadiazoles as histone deacetylase 6 (hdac6) inhibitors
EP3475275A4 (en) * 2016-06-23 2020-01-22 Merck Sharp & Dohme Corp. 3-aryl and heteroaryl substituted 5-trifluoromethyl oxadiazoles as histone deacetylase 6 (hdac6) inhibitors
CN106243083A (en) * 2016-07-12 2016-12-21 浙江宏冠生物药业有限公司 A kind of imatinib preparation method
CN106243083B (en) * 2016-07-12 2018-10-30 浙江宏冠生物药业有限公司 A kind of Imatinib preparation method
US11938134B2 (en) 2017-03-10 2024-03-26 Eikonizo Therapeutics, Inc. Metalloenzyme inhibitor compounds
WO2018177880A1 (en) 2017-03-31 2018-10-04 Syngenta Participations Ag Fungicidal compositions
US11974572B2 (en) 2017-03-31 2024-05-07 Sygenta Participations Ag Fungicidal compositions
WO2018177894A1 (en) 2017-03-31 2018-10-04 Syngenta Participations Ag Fungicidal compositions
WO2018185013A1 (en) 2017-04-03 2018-10-11 Syngenta Participations Ag Microbiocidal oxadiazole derivatives
WO2018184982A1 (en) 2017-04-05 2018-10-11 Syngenta Participations Ag Microbiocidal oxadiazole derivatives
WO2018184987A1 (en) 2017-04-05 2018-10-11 Syngenta Participations Ag Microbiocidal oxadiazole derivatives
WO2018184986A1 (en) 2017-04-05 2018-10-11 Syngenta Participations Ag Microbiocidal oxadiazole derivatives
WO2018184984A1 (en) 2017-04-05 2018-10-11 Syngenta Participations Ag Microbiocidal oxadiazole derivatives
WO2018184985A1 (en) 2017-04-05 2018-10-11 Syngenta Participations Ag Microbiocidal oxadiazole derivatives
WO2018184988A1 (en) 2017-04-05 2018-10-11 Syngenta Participations Ag Microbiocidal oxadiazole derivatives
CN110650956B (en) * 2017-04-06 2024-03-08 Fmc公司 Fungicidal oxadiazoles
US11708358B2 (en) 2017-04-06 2023-07-25 Fmc Corporation Fungicidal oxadiazoles
IL269786B2 (en) * 2017-04-06 2023-07-01 Fmc Corp Fungicidal oxadiazoles
WO2018187553A1 (en) * 2017-04-06 2018-10-11 Fmc Corporation Fungicidal oxadiazoles
KR102660243B1 (en) * 2017-04-06 2024-04-25 에프엠씨 코포레이션 Fungicide oxadiazole
CN110650956A (en) * 2017-04-06 2020-01-03 Fmc公司 Fungicidal oxadiazoles
JP7235672B2 (en) 2017-04-06 2023-03-08 エフ エム シー コーポレーション fungicidal oxadiazole
IL269786B1 (en) * 2017-04-06 2023-03-01 Fmc Corp Fungicidal oxadiazoles
EP3957636A1 (en) * 2017-04-06 2022-02-23 FMC Corporation Fungicidal oxadiazoles
JP2020516598A (en) * 2017-04-06 2020-06-11 エフ エム シー コーポレーションFmc Corporation Fungicidal oxadiazole
AU2018249537B2 (en) * 2017-04-06 2022-03-31 Fmc Corporation Fungicidal oxadiazoles
US10435399B2 (en) 2017-07-31 2019-10-08 Takeda Pharmaceutical Company Limited HDAC6 inhibitory heterocyclic compound
JP2021505684A (en) * 2017-12-05 2021-02-18 オリソン ヘノミクス,ソシエダ アノニマ 1,2,4-oxadiazole derivative as a histone deacetylase 6 inhibitor
JP7369458B2 (en) 2017-12-05 2023-10-26 オリソン ヘノミクス,ソシエダ アノニマ 1,2,4-oxadiazole derivatives as histone deacetylase 6 inhibitors
WO2019207062A1 (en) 2018-04-26 2019-10-31 Syngenta Participations Ag Microbiocidal oxadiazole derivatives
US11958844B2 (en) 2018-07-26 2024-04-16 Chong Kun Dang Pharmaceutical Corp. 1,3,4-oxadiazole derivative compounds as histone deacetylase 6 inhibitor, and pharmaceutical composition comprising the same
CN111285861B (en) * 2018-12-10 2021-08-17 浙江省化工研究院有限公司 Trifluoromethyl oxadiazole substituted pyrimidinamine compound, and preparation method and application thereof
CN111285861A (en) * 2018-12-10 2020-06-16 浙江省化工研究院有限公司 Trifluoromethyl oxadiazole substituted pyrimidinamine compound, and preparation method and application thereof
WO2020212513A1 (en) 2019-04-18 2020-10-22 Syngenta Crop Protection Ag Process for the preparation of microbiocidal oxadiazole derivatives
WO2020240493A1 (en) 2019-05-31 2020-12-03 Chong Kun Dang Pharmaceutical Corp. 1,3,4-oxadiazole homophthalimide derivative compounds as histone deacetylase 6 inhibitor, and the pharmaceutical composition comprising the same
WO2020254494A1 (en) * 2019-06-21 2020-12-24 Bayer Aktiengesellschaft Fungicidal oxadiazoles
US11453661B2 (en) 2019-09-27 2022-09-27 Takeda Pharmaceutical Company Limited Heterocyclic compound
US11958845B2 (en) 2019-09-27 2024-04-16 Takeda Pharmaceutical Company Limited Heterocyclic compound

Also Published As

Publication number Publication date
WO2013066839A3 (en) 2015-06-25

Similar Documents

Publication Publication Date Title
WO2013066839A2 (en) Compounds and methods
WO2013066835A2 (en) Compounds and methods
WO2013066838A1 (en) Compounds and methods
US8981084B2 (en) Oxadiazole HDAC inhibitors
WO2013006408A1 (en) Compounds and methods
WO2013066831A1 (en) Compounds and methods
AU2011205283B2 (en) Compounds and methods
WO2013066833A1 (en) Compounds and methods to inhibit histone deacetylase (hdac) enzymes
AU2002220195B2 (en) Anilinopyrimidine derivatives as IKK inhibitors and compositions and methods related thereto
AU2018202568A1 (en) Heterocyclyl compounds as MEK inhibitors
CN109563057B (en) Sulfonamide compound or salt thereof
EP3740481A1 (en) Dihydrobenzofuran and inden analogs as cardiac sarcomere inhibitors
WO2011088187A1 (en) Compounds and methods
WO2009006389A2 (en) Pyrimidine derivatives useful as raf kinase inhibitors
EP2552214A1 (en) Pyrazolyl-pyrimidines as kinase inhibitors
CN105793262B (en) Two cycloalkyne derivatives and application thereof
JP6846574B2 (en) Sulfonamide compounds and their use
US20210276991A1 (en) Cardiac sarcomere inhibitors
KR20140117651A (en) Isoquinoline and naphthyridine derivatives
IL274973B1 (en) 1,2,4-oxadiazole derivatives as histone deacetylase 6 inhibitors
CN103917530A (en) Morpholino substituted urea or carbamate derivatives as MTOR inhibitors
WO2017073743A1 (en) Tricyclic compound
WO2013066836A1 (en) Compounds and methods
WO2013066834A1 (en) Compounds and methods
WO2013066832A1 (en) Compounds and methods

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: 12844814

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase in:

Ref country code: DE

32PN Ep: public notification in the ep bulletin as address of the adressee cannot be established

Free format text: NOTING OF LOSS OF RIGHTS PURSUANT TO RULE 112(1) EPC (EPO FORM 1205A DATED 27/08/2014)

122 Ep: pct app. not ent. europ. phase

Ref document number: 12844814

Country of ref document: EP

Kind code of ref document: A2