WO2013007768A1 - Tricyclic heterocyclic compounds, compositions and methods of use thereof as jak inhibitors - Google Patents

Tricyclic heterocyclic compounds, compositions and methods of use thereof as jak inhibitors Download PDF

Info

Publication number
WO2013007768A1
WO2013007768A1 PCT/EP2012/063632 EP2012063632W WO2013007768A1 WO 2013007768 A1 WO2013007768 A1 WO 2013007768A1 EP 2012063632 W EP2012063632 W EP 2012063632W WO 2013007768 A1 WO2013007768 A1 WO 2013007768A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkylene
halogen
optionally substituted
oxo
alkyl
Prior art date
Application number
PCT/EP2012/063632
Other languages
French (fr)
Inventor
Christopher Hurley
Janusz Kulagowski
Mark Zak
Original Assignee
F. Hoffmann-La Roche Ag
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 F. Hoffmann-La Roche Ag filed Critical F. Hoffmann-La Roche Ag
Publication of WO2013007768A1 publication Critical patent/WO2013007768A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/12Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains three hetero rings
    • C07D471/14Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems

Definitions

  • JAK Janus kinases
  • STAT signal transducer and activator of transcription
  • JAK1, JAK2 and TYK2 exhibit broad patterns of gene expression, while JAK3 expression is limited to leukocytes.
  • Cytokine receptors are typically functional as heterodimers, and as a result, more than one type of JAK kinase is usually associated with cytokine receptor complexes.
  • the specific JAKs associated with different cytokine receptor complexes have been determined in many cases through genetic studies and corroborated by other experimental evidence.
  • JAK1 was initially identified in a screen for novel kinases (Wilks A.F., 1989, Proc. Natl. Acad. Sci. U.S.A. 86: 1603-1607). Genetic and biochemical studies have shown that JAK1 is functionally and physically associated with the type I interferon (e.g., IFNalpha), type II interferon (e.g., IFNgamma), IL-2 and IL-6 cytokine receptor complexes (Kisseleva et al, 2002, gene 285: 1-24; Levy et al, 2005, Nat. Rev. Mol. Cell Biol. 3:651-662; O'Shea et al, 2002, Cell, 109 (suppl): S121-S131).
  • type I interferon e.g., IFNalpha
  • type II interferon e.g., IFNgamma
  • IL-2 and IL-6 cytokine receptor complexes Kerisseleva et al, 2002,
  • JAKl knockout mice die perinatally due to defects in LIF receptor signaling (Kisseleva et al, 2002, gene 285: 1-24; O'Shea et al, 2002, Cell, 109 (suppl): S121- S131). Characterization of tissues derived from JAKl knockout mice demonstrated critical roles for this kinase in the IFN, IL-10, IL-2/IL-4, and IL-6 pathways.
  • a humanized monoclonal antibody targeting the IL-6 pathway was recently approved by the European Commission for the treatment of moderate-to-severe rheumatoid arthritis (Scheinecker et al, 2009, Nat. Rev. Drug Discov. 8:273-274).
  • JAK2 knockout mice die of anemia (O'Shea et al, 2002, Cell, 109 (suppl): S121-S131).
  • JAK2 V617F myeloproliferative disorders
  • JAK3 associates exclusively with the gamma common cytokine receptor chain, which is present in the IL-2, IL-4, IL-7, IL-9, IL-15 and IL-21 cytokine receptor complexes. JAK3 is critical for lymphoid cell development and proliferation and mutations in JAK3 result in severe combined immunodeficiency (SCID) (O'Shea et al, 2002, Cell, 109 (suppl): S121-S131).
  • SCID severe combined immunodeficiency
  • JAK3 and JAK3-mediated pathways have been targeted for immunosuppressive indications (e.g., transplantation rejection and rheumatoid arthritis) (Baslund et al, 2005, Arthritis & Rheumatism 52:2686-2692; Changelian et al, 2003, Science 302: 875-878).
  • immunosuppressive indications e.g., transplantation rejection and rheumatoid arthritis
  • TYK2 associates with the type I interferon (e.g., IFNalpha), IL-6, IL-10, IL-12 and IL-23 cytokine receptor complexes (Kisseleva et al, 2002, gene 285: 1-24; Watford, W.T. & O'Shea, J. J., 2006, Immunity 25:695-697). Consistent with this, primary cells derived from a TYK2 deficient human are defective in type I interferon, IL-6, IL-10, IL-12 and IL-23 signaling.
  • a fully human monoclonal antibody targeting the shared p40 subunit of the IL-12 and 11-23 cytokines was recently approved by the European Commission for the treatment of moderate-to-severe plaque psoriasis (Krueger et al, 2007, N. Engl. J. Med. 356:580-92; Reich et al, 2009, Nat. Rev. Drug Discov. 8:355-356).
  • an antibody targeting the IL-12 and IL-23 pathways underwent clinical trials for treating Crohn's Disease (Mannon et al, 2004, N. Engl. J. Med. 351 :2069-79).
  • One aspect includes a compound of formula I: stereoisomers, tautomers or pharmaceutically acceptable salts thereof, wherein X, Y, R 1 , R 2 and R 3 are defined herein.
  • Another aspect includes a pharmaceutical composition that includes a compound of formula I and a pharmaceutically acceptable carrier, adjuvant or vehicle.
  • Another aspect includes a method of treating or lessening the severity of a disease or condition responsive to the inhibition of JAK1 kinase activity in a patient.
  • the method includes administering to the patient a therapeutically effective amount of a compound of formula I.
  • Another aspect includes a compound of formula I, a stereoisomer, tautomer, prodrug or pharmaceutically acceptable salt thereof, for use in therapy.
  • Another aspect includes the use of a compound of formula I, a stereoisomer, tautomer, prodrug or pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of a disease responsive to the inhibition of JAK1 kinase activity.
  • kits for treating a disease or disorder responsive to the inhibition of JAK1 kinase includes a first pharmaceutical composition comprising a compound of formula I and instructions for use
  • Acyl means a carbonyl containing substituent represented by the formula -C(0)-R in which R is hydrogen, alkyl, a cycloalkyl, a heterocyclyl, cycloalkyl -substituted alkyl or heterocyclyl-substituted alkyl wherein the alkyl, alkoxy, cycloalkyl and heterocyclyl are as defined herein.
  • Acyl groups include alkanoyl (e.g. acetyl), aroyl (e.g. benzoyl), and heteroaroyl (e.g. pyridinoyl).
  • alkyl refers to a saturated linear or branched-chain monovalent hydrocarbon radical, wherein the alkyl radical may be optionally substituted independently with one or more substituents described herein.
  • the alkyl radical is one to eighteen carbon atoms (Ci-Cis).
  • the alkyl radical is Co-C 6 , C0-C5, C0-C3, C1-C12, C1-C10, Ci-Cs, Ci- C 6 , C1-C5, C1-C4, or C1-C3.
  • Co alkyl refers to a bond.
  • alkyl groups include methyl (Me, -CH 3 ), ethyl (Et, -CH 2 CH 3 ), 1 -propyl (n-Pr, n-propyl, -CH 2 CH 2 CH 3 ), 2-propyl (i-Pr, i- propyl, -CH(CH 3 ) 2 ), 1 -butyl (n-Bu, n-butyl, -CH 2 CH 2 CH 2 CH 3 ), 2-methyl-l -propyl (i-Bu, i-butyl, -CH 2 CH(CH 3 ) 2 ), 2-butyl (s-Bu, s-butyl, -CH(CH 3 )CH 2 CH 3 ), 2-methyl-2-propyl (t-Bu, t-butyl, - C(CH 3 ) 3 ), 1-pentyl (n-pentyl, -CH 2 CH 2 CH 2 CH 2 CH 3 ), 2-pentyl (-CH(CH 3
  • alkenyl refers to linear or branched- chain monovalent hydrocarbon radical with at least one site of unsaturation, i.e., a carbon-carbon double bond, wherein the alkenyl radical may be optionally substituted independently with one or more substituents described herein, and includes radicals having "cis” and “trans” orientations, or alternatively, "E” and "Z” orientations.
  • the alkenyl radical is two to eighteen carbon atoms (C 2 -Cis).
  • the alkenyl radical is C 2 -C 12 , C 2 -C 10 , C 2 -Cs, C 2 -C 6 or C 2 -C 3 .
  • alkoxy refers to a linear or branched monovalent radical represented by the formula -OR in which R is alkyl, alkenyl, alkynyl or cycloalkyl, which can be further optionally substituted as defined herein.
  • Alkoxy groups include methoxy, ethoxy, propoxy, isopropoxy, mono-, di- and tri-fluoromethoxy and cyclopropoxy.
  • alkynyl refers to a linear or branched monovalent hydrocarbon radical with at least one site of unsaturation, i.e., a carbon-carbon, triple bond, wherein the alkynyl radical may be optionally substituted independently with one or more substituents described herein.
  • the alkynyl radical is two to eighteen carbon atoms (C 2 -C 18 ).
  • the alkynyl radical is C 2 -C 12 , C 2 -C 10 , C 2 -Cs, C 2 -C 6 or C 2 -C 3 .
  • Examples include, but are not limited to, ethynyl (-C ⁇ CH), prop-l-ynyl (-C ⁇ CCH 3 ), prop-2-ynyl (propargyl, -CH 2 C ⁇ CH), but-l-ynyl, but-2-ynyl and but-3-ynyl.
  • Alkylene refers to a saturated, branched or straight chain hydrocarbon group having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent alkane.
  • the divalent alkylene group is one to eighteen carbon atoms (Ci-Cis).
  • the divalent alkylene group is Co-C 6 , C0-C5, C0-C3, C1-C12, C1-C10, Ci-Cs, Ci-C 6 , C1-C5, C1-C4, or Ci-C 3 .
  • the group Co alkylene refers to a bond.
  • Example alkylene groups include methylene (-CH 2 -), 1,1 -ethyl (-CH(CH 3 )-), (1,2- ethyl (-CH 2 CH 2 -), 1,1-propyl (-CH(CH 2 CH 3 )-), 2,2-propyl (-C(CH 3 ) 2 -), 1,2-propyl (-CH(CH 3 )CH 2 -), 1,3-propyl (-CH 2 CH 2 CH 2 -), l,l-dimethyleth-l,2-yl (-C(CH 3 ) 2 CH 2 -), 1,4-butyl (-CH 2 CH 2 CH 2 CH 2 -), and the like.
  • Alkenylene refers to an unsaturated, branched or straight chain hydrocarbon group having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent alkene.
  • the alkenylene group is two to eighteen carbon atoms (C 2 -Cis).
  • the alkenylene group is C 2 -C 12 , C 2 - C10, C 2 -Cs, C 2 -C 6 or C 2 -C 3 .
  • Alkynylene refers to an unsaturated, branched or straight chain hydrocarbon group having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent alkyne.
  • the alkynylene radical is two to eighteen carbon atoms (C 2 -Cis).
  • the alkynylene radical is C 2 -C 12 , C 2 - C10, C 2 -Cs, C 2 -C 6 or C 2 -C 3 .
  • Example alkynylene radicals include: acetylene (-C ⁇ C-), propargyl (-CH 2 C ⁇ C-), and 4-pentynyl (-CH 2 CH 2 CH 2 C ⁇ C-).
  • Amidine means the group -C(NH)-NHR in which R is hydrogen, alkyl, a cycloalkyl, a heterocyclyl, cycloalkyl-substituted alkyl or heterocyclyl-substituted alkyl wherein the alkyl, alkoxy, cycloalkyl and heterocyclyl are as defined herein.
  • a particular amidine is the group - NH-C(NH)-NH 2 .
  • Amino means primary (i.e., -NH 2 ) , secondary (i.e., -NRH) and tertiary (i.e., -NRR) amines, that are optionally substituted, in which R is alkyl, alkoxy, a cycloalkyl, a heterocyclyl, cycloalkyl-substituted alkyl or heterocyclyl-substituted alkyl wherein the alkyl, alkoxy, cycloalkyl and heterocyclyl are as defined herein
  • Particular secondary and tertiary amines are alkylamine, dialkylamine, arylamine, diarylamine, aralkylamine and diaralkylamine wherein the alkyl is as herein defined and optionally substituted.
  • Particular secondary and tertiary amines are methylamine, ethylamine, propylamine, isopropylamine, phenylamine, benzylamine dimethylamine, diethylamine, dipropylamine and diisopropylamine.
  • amino -protecting group refers to a derivative of the groups commonly employed to block or protect an amino group while reactions are carried out on other functional groups on the compound.
  • protecting groups include carbamates, amides, alkyl and aryl groups, imines, as well as many N-heteroatom derivatives which can be removed to regenerate the desired amine group.
  • Particular amino protecting groups are Pmb (p- Methoxybenzyl), Boc (tert-Butyloxycarbonyl), Fmoc (9-Fluorenylmethyloxycarbonyl) and Cbz (Carbobenzyloxy). Further examples of these groups are found in T. W. Greene and P. G. M.
  • Aryl when used alone, or as part of another term, means a carbocyclic aromatic group, whether or not fused to one or more groups, having the number of carbon atoms designated, or if no number is designated, up to 14 carbon atoms.
  • One example includes aryl groups having 6-14 carbon atoms.
  • Another example inlcudes aryl groups having 6-10 carbon atoms. Examples of aryl groups include phenyl, naphthyl, biphenyl, phenanthrenyl, naphthacenyl, 1 ,2,3,4- tetrahydronaphthalenyl, lH-indenyl, 2,3-dihydro-lH-indenyl, and the like (see e.g.
  • aryl is phenyl.
  • Substituted phenyl or substituted aryl means a phenyl group or aryl group substituted with one, two, three, four or five, for example 1-2, 1-3 or 1-4 substituents chosen from groups specified herein.
  • optional substituents on aryl are selected from halogen (F, CI, Br, I), hydroxy, protected hydroxy, cyano, nitro, alkyl (for example Ci-C 6 alkyl), alkoxy (for example Ci-C 6 alkoxy), benzyloxy, carboxy, protected carboxy, carboxymethyl, protected carboxymethyl, hydroxymethyl, protected hydroxymethyl, aminomethyl, protected aminomethyl, trifluoromethyl, alkylsulfonylamino, alkylsulfonylaminoalkyl, arylsulfonylamino, arylsulfonylaminoalkyl, heterocyclylsulfonylamino, heterocyclylsulfonylaminoalkyl, heterocyclyl, aryl, or other groups specified.
  • halogen F, CI, Br, I
  • alkyl for example Ci-C 6 alkyl
  • alkoxy for
  • One or more methyne (CH) and/or methylene (CH 2 ) groups in these substituents may in turn be substituted with a similar group as those denoted above.
  • substituted phenyl include a mono- or di(halo)phenyl group such as 2-chlorophenyl, 2-bromophenyl, 4-chlorophenyl, 2,6-dichlorophenyl, 2,5- dichlorophenyl, 3,4-dichlorophenyl, 3-chlorophenyl, 3-bromophenyl, 4-bromophenyl, 3,4- dibromophenyl, 3-chloro-4-fluorophenyl, 2-fluorophenyl and the like; a mono- or di(hydroxy)phenyl group such as 4-hydroxyphenyl, 3-hydroxyphenyl, 2,4-dihydroxyphenyl, the protected-hydroxy derivatives thereof and the like; a nitrophenyl group such as 3- or 4-
  • substituted phenyl represents disubstituted phenyl groups where the substituents are different, for example, 3-methyl-4-hydroxyphenyl, 3- chloro-4-hydroxyphenyl, 2-methoxy-4-bromophenyl, 4-ethyl-2-hydroxyphenyl, 3-hydroxy-4- nitrophenyl, 2-hydroxy-4-chlorophenyl, and the like, as well as trisubstituted phenyl groups where the substituents are different, for example 3-methoxy-4-benzyloxy-6-methyl sulfonylamino, 3-methoxy-4-benzyloxy-6-phenyl sulfonylamino, and tetrasubstituted phenyl groups where the substituents are different such as 3-methoxy-4-benzyloxy-5-methyl-6-phenyl sulfonylamino.
  • Particular substituted phenyl groups include the 2-chlorophenyl, 2-aminophenyl, 2-bromophenyl, 3-methoxyphenyl, 3-ethoxy-phenyl, 4-benzyloxyphenyl, 4-methoxyphenyl, 3- ethoxy-4-benzyloxyphenyl, 3,4-diethoxyphenyl, 3-methoxy-4-benzyloxyphenyl, 3-methoxy-4- (l-chloromethyl)benzyloxy-6-methyl sulfonyl aminophenyl groups.
  • Fused aryl rings may also be substituted with any, for example 1 , 2 or 3, of the substituents specified herein in the same manner as substituted alkyl groups.
  • cancer and “cancerous”, “neoplasm”, “tumor” refer to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth.
  • a “tumor” comprises one or more cancerous cells. Examples of cancer include carcinoma, lymphoma, blastoma, sarcoma, and leukemia or lymphoid malignancies.
  • squamous cell cancer e.g., epithelial squamous cell cancer
  • lung cancer including small- cell lung cancer, non-small cell lung cancer ("NSCLC"), adenocarcinoma of the lung and squamous carcinoma of the lung, cancer of the peritoneum, hepatocellular cancer, gastric or stomach cancer including gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatoma, breast cancer, colon cancer, rectal cancer, colorectal cancer, endometrial or uterine carcinoma, salivary gland carcinoma, kidney or renal cancer, prostate cancer, vulval cancer, thyroid cancer, hepatic carcinoma, anal carcinoma, penile carcinoma, melanoma, multiple myeloma and B-cell lymphoma, brain, as well as head and neck cancer, and associated metastases.
  • NSCLC non-small cell lung cancer
  • chemotherapeutic agent is an agent useful in the treatment of a given disorder, for example, cancer or inflammatory disorders.
  • chemotherapeutic agents include NSAIDs; hormones such as glucocorticoids; corticosteroids such as hydrocortisone, hydrocortisone acetate, cortisone acetate, tixocortol pivalate, prednisolone, methylprednisolone, prednisone, triamcinolone acetonide, triamcinolone alcohol, mometasone, amcinonide, budesonide, desonide, fluocinonide, fluocinolone acetonide, halcinonide, betamethasone, betamethasone sodium phosphate, dexamethasone, dexamethasone sodium phosphate, fluocortolone, hydrocortisone- 17-butyrate, hydrocortisone- 17-valerate, aclometasone dipropionate
  • radioactive isotopes e.g., At , 1 , 1 , Y 90 , Re 186 , Re 188 , Sm 153 , Bi 212 , P 32 , Pb 212 and radioactive isotopes of Lu
  • miscellaneous investigational agents such as thioplatin, PS-341, phenylbutyrate, ET-18- OCH 3 , or farnesyl transferase inhibitors (L-739749, L-744832); polyphenols such as quercetin, resveratrol, piceatannol, epigallocatechine gallate, theaflavins, flavanols, procyanidins, betulinic acid and derivatives thereof; autophagy inhibitors such as chloroquine; alkylating agents such as thiotepa and cyclosphosphamide (CYTOXAN®);
  • calicheamicin especially calicheamicin gammall and calicheamicin omegall (see, e.g., Nicolaou et al., Angew. Chem Intl. Ed. Engl., 33: 183-186 (1994)); CDP323, an oral alpha-4 integrin inhibitor; dynemicin, including dynemicin A; an esperamicin; as well as neocarzino statin chromophore and related chromoprotein enediyne antibiotic chromophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, carabicin, carminomycin, carzinophilin, chromomycins, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, doxorubicin (
  • celecoxib or etoricoxib proteosome inhibitor
  • proteosome inhibitor e.g. PS341
  • bortezomib VELCADE®
  • CCI-779 tipifarnib (R1 1577); orafenib, ABT510
  • Bcl-2 inhibitor such as oblimersen sodium (GENASENSE®)
  • pixantrone EGFR inhibitors (see definition below); farnesyltransferase inhibitors such as lonafarnib (SCH 6636, SARASARTM); and pharmaceutically acceptable salts, acids or derivatives of any of the above; as well as combinations of two or more of the above such as CHOP, an abbreviation for a combined therapy of cyclophosphamide, doxorubicin, vincristine, and prednisolone; and FOLFOX, an abbreviation for a treatment regimen with oxaliplatin (ELOXATINTM) combined with 5-FU and leucovor
  • endocrine therapeutics which act to regulate, reduce, block, or inhibit the effects of hormones that can promote the growth of cancer. They may be hormones themselves, including, but not limited to: anti-estrogens with mixed agonist/antagonist profile, including, tamoxifen (NOLVADEX®), 4-hydroxytamoxifen, toremifene (FARESTON®), idoxifene, droloxifene, raloxifene (EVISTA®), trioxifene, keoxifene, and selective estrogen receptor modulators (SERMs) such as SERM3; pure anti-estrogens without agonist properties, such as fulvestrant (FASLODEX®), and EM800 (such agents may block estrogen receptor (ER) dimerization, inhibit DNA binding, increase ER turnover, and/or suppress ER levels); aromatase inhibitors, including steroidal aromatase inhibitors such as formestane and exemestane (AROMASIN®), and nonsteroidal aroma
  • Additional chemotherapeutic agents include therapeutic antibodies such as alemtuzumab (Campath), bevacizumab (AVASTIN®, Genentech); cetuximab (ERBITUX®, Imclone); panitumumab (VECTIBIX®, Amgen), rituximab (RITUXAN®, Genentech/Biogen pie), pertuzumab (OMNITARG®, 2C4, Genentech), trastuzumab (HERCEPTIN®, Genentech), tositumomab (Bexxar, Corixia), and the antibody drug conjugate, gemtuzumab ozogamicin (MYLOTARG®, Wyeth).
  • therapeutic antibodies such as alemtuzumab (Campath), bevacizumab (AVASTIN®, Genentech); cetuximab (ERBITUX®, Imclone); panitumumab (VECTIBIX®, Amgen), rituximab
  • Additional humanized monoclonal antibodies with therapeutic potential as agents in combination with the compounds of the invention include: apolizumab, aselizumab, atlizumab, bapineuzumab, bivatuzumab mertansine, cantuzumab mertansine, cedelizumab, certolizumab pegol, cidfusituzumab, cidtuzumab, daclizumab, eculizumab, efalizumab, epratuzumab, erlizumab, felvizumab, fontolizumab, gemtuzumab ozogamicin, inotuzumab ozogamicin, ipilimumab, labetuzumab, lintuzumab, matuzumab, mepolizumab, motavizumab, motovizumab, natalizumab, nimotuzumab, nolovizum
  • Chemotherapeutic agents also include "EGFR inhibitors,” which refers to compounds that bind to or otherwise interact directly with EGFR and prevent or reduce its signaling activity, and is alternatively referred to as an "EGFR antagonist.”
  • EGFR inhibitors refers to compounds that bind to or otherwise interact directly with EGFR and prevent or reduce its signaling activity
  • Examples of such agents include antibodies and small molecules that bind to EGFR.
  • antibodies which bind to EGFR include MAb 579 (ATCC CRL HB 8506), MAb 455 (ATCC CRL HB8507), MAb 225 (ATCC CRL 8508), MAb 528 (ATCC CRL 8509) (see, US Patent No.
  • EMD 55900 Stragliotto et al. Eur. J. Cancer 32A:636-640 (1996)
  • EMD7200 (matuzumab) a humanized EGFR antibody directed against EGFR that competes with both EGF and TGF-alpha for EGFR binding (EMD/Merck); human EGFR antibody, HuMax-EGFR (GenMab); fully human antibodies known as El . l, E2.4, E2.5, E6.2, E6.4, E2. l l, E6. 3 and E7.6.
  • the anti-EGFR antibody may be conjugated with a cytotoxic agent, thus generating an immuno conjugate (see, e.g., EP659,439A2, Merck Patent GmbH).
  • EGFR antagonists include small molecules such as compounds described in US Patent Nos: 5,616,582, 5,457,105, 5,475,001, 5,654,307, 5,679,683, 6,084,095, 6,265,410, 6,455,534, 6,521,620, 6,596,726, 6,713,484, 5,770,599, 6,140,332, 5,866,572, 6,399,602, 6,344,459, 6,602,863, 6,391,874, 6,344,455, 5,760,041, 6,002,008, and 5,747,498, as well as the following PCT publications: W098/14451, WO98/50038, WO99/09016, and WO99/24037.
  • EGFR antagonists include OSI-774 (CP-358774, erlotinib, TARCEVA ® Genentech/OSI Pharmaceuticals); PD 183805 (CI 1033, 2-propenamide, N-[4-[(3-chloro-4-fluorophenyl)amino]- 7-[3-(4-morpholinyl)propoxy]-6-quinazolinyl]-, dihydrochloride, Pfizer Inc.); ZD 1839, gefitinib (IRESSAJ) 4-(3'-Chloro-4'-fluoroanilino)-7-methoxy-6-(3-morpholinopropoxy)quinazoline, AstraZeneca); ZM 105180 ((6-amino-4-(3-methylphenyl-amino)-quinazoline, Zeneca); BIBX- 1382 (N8-(3-chloro-4-fluoro-phenyl)-N2-( 1 -
  • Chemotherapeutic agents also include "tyrosine kinase inhibitors" including the EGFR- targeted drugs noted in the preceding paragraph; small molecule HER2 tyrosine kinase inhibitor such as TAK165 available from Takeda; CP-724,714, an oral selective inhibitor of the ErbB2 receptor tyrosine kinase (Pfizer and OSI); dual-HER inhibitors such as EKB-569 (available from Wyeth) which preferentially binds EGFR but inhibits both HER2 and EGFR-overexpressing cells; lapatinib (GSK572016; available from Glaxo-SmithKline), an oral HER2 and EGFR tyrosine kinase inhibitor; PKI-166 (available from Novartis); pan-HER inhibitors such as canertinib (CI-1033; Pharmacia); Raf-1 inhibitors such as antisense agent ISIS-5132 available from ISIS Pharmaceuticals which inhibit Raf-1 signaling; non-HER targeted TK inhibitor
  • NSAID non-steroidal anti- inflammatory drug
  • NSAIDs include non-selective inhibitors of the enzyme cyclooxygenase.
  • NSAIDs include aspirin, propionic acid derivatives such as ibuprofen, fenoprofen, ketoprofen, flurbiprofen, oxaprozin and naproxen, acetic acid derivatives such as indomethacin, sulindac, etodolac, diclofenac, enolic acid derivatives such as piroxicam, meloxicam, tenoxicam, droxicam, lornoxicam and isoxicam, fenamic acid derivatives such as mefenamic acid, meclofenamic acid, flufenamic acid, tolfenamic acid, and COX-2 inhibitors such as celecoxib, etoricoxib, lumiracoxib, parecoxib, rofecoxib, rofecoxib, and valdecoxib.
  • acetic acid derivatives such as indomethacin
  • sulindac sulindac
  • NSAIDs can be indicated for the symptomatic relief of conditions such as rheumatoid arthritis, osteoarthritis, inflammatory arthropathies, ankylosing spondylitis, psoriatic arthritis, Reiter's syndrome, acute gout, dysmenorrhoea, metastatic bone pain, headache and migraine, postoperative pain, mild-to- moderate pain due to inflammation and tissue injury, pyrexia, ileus, and renal colic.
  • conditions such as rheumatoid arthritis, osteoarthritis, inflammatory arthropathies, ankylosing spondylitis, psoriatic arthritis, Reiter's syndrome, acute gout, dysmenorrhoea, metastatic bone pain, headache and migraine, postoperative pain, mild-to- moderate pain due to inflammation and tissue injury, pyrexia, ileus, and renal colic.
  • chemotherapeutic agents include pharmaceutically acceptable salts, acids or derivatives of any of chemotherapeutic agents, described herein, as well as combinations of two or more of them.
  • Cycloalkyl refers to a non-aromatic, saturated or partially unsaturated hydrocarbon ring group wherein the cycloalkyl group may be optionally substituted independently with one or more substituents described herein. In one example, the cycloalkyl group is 3 to 12 carbon atoms (C 3 -C12). In other examples, cycloalkyl is C 3 -C8, C 3 -Cio or C 5 -C 10 .
  • the cycloalkyl group, as a monocycle is C 3 -C8, C3-C6 or C5-C6.
  • the cycloalkyl group, as a bicycle is C 7 -C 12 .
  • the cycloalkyl group, as a spiro system is C 5 - C12.
  • Examples of monocyclic cycloalkyl include cyclopropyl, cyclo butyl, cyclopentyl, 1- cyclopent-l-enyl, l-cyclopent-2-enyl, l-cyclopent-3-enyl, cyclo hexyl, perdeuteriocyclohexyl, 1- cyclohex-l-enyl, l-cyclohex-2-enyl, 1 -cyclo hex-3-enyl, cyclo hexadienyl, cyclo heptyl, cyclooctyl, cyclononyl, cyclodecyl, cycloundecyl and cyclododecyl.
  • Exemplary arrangements of bicyclic cycloalkyls having 7 to 12 ring atoms include, but are not limited to, [4,4], [4,5], [5,5], [5,6] or [6,6] ring systems.
  • Exemplary bridged bicyclic cycloalkyls include, but are not limited to, bicyclo[2.2.1]heptane, bicyclo[2.2.2]octane and bicyclo[3.2.2]nonane.
  • Examples of spiro cycloalkyl include, spiro[2.2]pentane, spiro[2.3]hexane, spiro[2.4]heptane, spiro[2.5]octane and spiro[4.5]decane.
  • Carboxy-protecting group refers to those groups that are stable to the conditions of subsequent reaction(s) at other positions of the molecule, which may be removed at the appropriate point without disrupting the remainder of the molecule, to give the unprotected carboxy-group.
  • carboxy protecting groups include, ester groups and heterocyclyl groups. Ester derivatives of the carboxylic acid group may be employed to block or protect the carboxylic acid group while reactions are carried out on other functional groups on the compound.
  • ester groups include substituted arylalkyl, including substituted benzyls, such as 4-nitrobenzyl, 4-methoxybenzyl, 3,4-dimethoxybenzyl, 2,4-dimethoxybenzyl, 2,4,6-trimethoxybenzyl, 2,4,6-trimethylbenzyl, pentamethylbenzyl, 3,4-methylenedioxybenzyl, benzhydryl, 4,4'-dimethoxybenzhydryl, 2,2',4,4'-tetramethoxybenzhydryl, alkyl or substituted alkyl esters such as methyl, ethyl, t-butyl allyl or t-amyl, triphenylmethyl (trityl), 4- methoxytrityl, 4,4'-dimethoxytrityl, 4,4',4"-trimethoxytrityl, 2-phenylprop-2-yl, thioesters such as t-butyl thio
  • carboxy-protecting groups are heterocyclyl groups such as 1,3-oxazolinyl. Further examples of these groups are found in T. W. Greene and P. G. M. Wuts, "Protective Groups in Organic Synthesis", 2 nd ed., John Wiley & Sons, Inc., New York, N.Y., 1991, chapter 5; E. Haslam, "Protective Groups in Organic Chemistry", J. G. W. McOmie, Ed., Plenum Press, New York, N.Y., 1973, Chapter 5, and T.W. Greene, “Protective Groups in Organic Synthesis", John Wiley and Sons, New York, NY, 1981, Chapter 5.
  • protected carboxy refers to a carboxy group substituted with one of the above carboxy- protecting groups.
  • "Guanidine” means the group -NH-C(NH)-NHR in which R is hydrogen, alkyl, alkoxy, a cycloalkyl, a heterocyclyl, cycloalkyl -substituted alkyl or heterocyclyl-substituted alkyl wherein the alkyl, alkoxy, cycloalkyl and heterocyclyl are as defined herein.
  • a particular guanidine is the group -NH-C(NH)-NH 2 .
  • “Hydroxy-protecting group” refers to a derivative of the hydroxy group commonly employed to block or protect the hydroxy group while reactions are carried out on other functional groups on the compound.
  • protecting groups include tetrahydropyranyloxy, benzoyl, acetoxy, carbamoyloxy, benzyl, and silylethers (e.g. TBS, TBDPS) groups. Further examples of these groups are found in T. W. Greene and P. G. M. Wuts, "Protective Groups in Organic Synthesis", 2 nd ed., John Wiley & Sons, Inc., New York, NY, 1991, chapters 2-3; E. Haslam, "Protective Groups in Organic Chemistry", J.
  • protected hydroxy refers to a hydroxy group substituted with one of the above hydroxy- protecting groups.
  • Heterocyclic group “heterocyclic”, “heterocycle”, “heterocyclyl”, or “heterocyclo” alone, and when used as a moiety in a complex group such as a heterocycloalkyl group, are used interchangeably and refer to any mono-, bi-, tricyclic or spiro, saturated or unsaturated, aromatic (heteroaryl) or non-aromatic, ring system, having 3 to 20 ring atoms, where the ring atoms are carbon, and at least one atom in the ring or ring system is a heteroatom selected from nitrogen, sulfur or oxygen.
  • heterocyclyl includes 3-12 ring atoms and includes monocycles, bicycles, tricycles and spiro ring systems, wherein the ring atoms are carbon, and at least one atom in the ring or ring system is a heteroatom selected from nitrogen, sulfur or oxygen.
  • heterocyclyl includes 1 to 4 heteroatoms.
  • heterocyclyl includes 3- to 7-membered monocycles having one or more heteroatoms selected from nitrogen, sulfur or oxygen.
  • heterocyclyl includes 4- to 6-membered monocycles having one or more heteroatoms selected from nitrogen, sulfur or oxygen.
  • heterocyclyl includes 3-membered monocycles.
  • heterocyclyl includes 4-membered monocycles.
  • heterocyclyl includes 5-6- membered monocycles.
  • the heterocyclyl group includes 0 to 3 double bonds.
  • Any nitrogen or sulfur heteroatom may optionally be oxidized (e.g. NO, SO, S0 2 ), and any nitrogen heteroatom may optionally be quaternized (e.g. [ ⁇ ] + 0 ⁇ , [NPv 4 ] + OFf).
  • Example heterocycles are oxiranyl, aziridinyl, thiiranyl, azetidinyl, oxetanyl, thietanyl, 1 ,2-dithietanyl, 1,3-dithietanyl, pyrrolidinyl, dihydro-lH-pyrrolyl, dihydrofuranyl, tetrahydrofuranyl, dihydrothienyl, tetrahydrothienyl, imidazolidinyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, 1,1-dioxo-thiomorpholinyl, dihydropyranyl, tetrahydropyranyl, hexahydrothiopyranyl, hexahydropyrimidinyl, oxazinanyl, thiazinanyl, thioxanyl, homopiperazinyl, homopiperidinyl,
  • Examples of 5-membered heterocycles containing a sulfur or oxygen atom and one to three nitrogen atoms are thiazolyl, including thiazol-2-yl and thiazol-2-yl N-oxide, thiadiazolyl, including l,3,4-thiadiazol-5-yl and 1,2,4- thiadiazol-5-yl, oxazolyl, for example oxazol-2-yl, and oxadiazolyl, such as l,3,4-oxadiazol-5-yl, and l,2,4-oxadiazol-5-yl.
  • Example 5-membered ring heterocycles containing 2 to 4 nitrogen atoms include imidazolyl, such as imidazol-2-yl; triazolyl, such as l,3,4-triazol-5-yl; 1,2,3- triazol-5-yl, l,2,4-triazol-5-yl, and tetrazolyl, such as lH-tetrazol-5-yl.
  • Example benzo-fused 5- membered heterocycles are benzoxazol-2-yl, benzthiazol-2-yl and benzimidazol-2-yl.
  • Example 6-membered heterocycles contain one to three nitrogen atoms and optionally a sulfur or oxygen atom, for example pyridyl, such as pyrid-2-yl, pyrid-3-yl, and pyrid-4-yl; pyrimidyl, such as pyrimid-2-yl and pyrimid-4-yl; triazinyl, such as l,3,4-triazin-2-yl and l,3,5-triazin-4-yl; pyridazinyl, in particular pyridazin-3-yl, and pyrazinyl.
  • pyridyl such as pyrid-2-yl, pyrid-3-yl, and pyrid-4-yl
  • pyrimidyl such as pyrimid-2-yl and pyrimid-4-yl
  • triazinyl such as l,3,4-triazin-2-yl and l,3,5-tria
  • pyridine N-oxides and pyridazine N-oxides and the pyridyl, pyrimid-2-yl, pyrimid-4-yl, pyridazinyl and the l,3,4-triazin-2-yl groups are other example heterocycle groups.
  • Substituents for "optionally substituted heterocycles" include hydroxyl, alkyl, alkoxy, acyl, halogen, mercapto, oxo, carboxyl, halo- substituted alkyl, amino, cyano, nitro, amidino, guanidino.
  • Heteroaryl alone and when used as a moiety in a complex group such as a heteroaralkyl group, refers to any mono-, bi-, or tricyclic ring system where at least one ring is a 5- or, 6- membered aromatic ring containing from 1 to 4 heteroatoms selected from nitrogen, oxygen, and sulfur, and in an example embodiment, at least one heteroatom is nitrogen. See, for example, Lang's Handbook of Chemistry, supra. Included in the definition are any bicyclic groups where any of the above heteroaryl rings are fused to an aryl ring.
  • heteroaryl includes 4-6 membered monocyclic aromatic groups where one or more ring atoms is nitrogen, sulfur or oxygen.
  • heteroaryl includes 5-6 membered monocyclic aromatic groups where one or more ring atoms is nitrogen, sulfur or oxygen.
  • Example heteroaryl groups include thienyl, furyl, imidazolyl, pyrazolyl, thiazolyl, isothiazolyl, oxazolyl, isoxazolyl, triazolyl, thiadiazolyl, oxadiazolyl, tetrazolyl, thiatriazolyl, oxatriazolyl, pyridyl, pyrimidyl, pyrazinyl, pyridazinyl, triazinyl, tetrazinyl, tetrazolo[l,5-b]pyridazinyl, imidazol[l,2-a]pyrimidinyl and purinyl, as well as benzo-fused derivatives, for example benzoxazolyl, benzofu
  • heteroaryl groups are: 1 ,3-thiazol-2-yl, 4-(carboxymethyl)-5 -methyl- 1 ,3-thiazol-2-yl, 4-(carboxymethyl)-5 -methyl- 1,3- thiazol-2-yl sodium salt, l,2,4-thiadiazol-5-yl, 3-methyl-l,2,4-thiadiazol-5-yl, l,3,4-triazol-5-yl, 2-methyl-l,3,4-triazol-5-yl, 2-hydroxy-l,3,4-triazol-5-yl, 2-carboxy-4-methyl-l,3,4-triazol-5-yl sodium salt, 2-carboxy-4-methyl-l,3,4-triazol-5-yl, l,3-oxazol-2-yl, l,3,4-oxadiazol-5-yl, 2- methyl- 1 ,3,4-oxadiazol-5-yl, 2-(hydroxymethyl)- 1 ,3,3,4-oxadia
  • a heterocyclyl group is attached at a carbon atom of the heterocyclyl group.
  • carbon bonded heterocyclyl groups include bonding arrangements at position 2, 3, 4, 5, or 6 of a pyridine ring, position 3, 4, 5, or 6 of a pyridazine, position 2, 4, 5, or 6 of a pyrimidine ring, position 2, 3, 5, or 6 of a pyrazine ring, position 2, 3, 4, or 5 of a furan, tetrahydroiuran, thiofuran, thiophene, pyrrole or tetrahydropyrrole ring, position 2, 4, or 5 of an oxazole, imidazole or thiazole ring, position 3, 4, or 5 of an isoxazole, pyrazole, or isothiazole ring, position 2 or 3 of an aziridine ring, position 2, 3, or 4 of an azetidine ring, position 2, 3, 4, 5, 6, 7, or 8 of a
  • the heterocyclyl group is N-attached.
  • the nitrogen bonded heterocyclyl or heteroaryl group include bonding arrangements at position 1 of an aziridine, azetidine, pyrrole, pyrrolidine, 2-pyrroline, 3-pyrroline, imidazole, imidazolidine, 2- imidazoline, 3 -imidazoline, pyrazole, pyrazoline, 2-pyrazoline, 3-pyrazoline, piperidine, piperazine, indole, indoline, lH-indazole, position 2 of a isoindole, or isoindoline, position 4 of a morpholine, and position 9 of a carbazole, or ⁇ -carboline.
  • leaving group refers to a portion of a first reactant in a chemical reaction that is displaced from the first reactant in the chemical reaction.
  • Examples of leaving groups include, but are not limited to, halogen atoms, alkoxy and sulfonyloxy groups.
  • Example sulfonyloxy groups include, but are not limited to, alkylsulfonyloxy groups (for example methyl sulfonyloxy (mesylate group) and trifluoromethylsulfonyloxy (triflate group)) and arylsulfonyloxy groups (for example /?-toluenesulfonyloxy (tosylate group) and /?-nitrosulfonyloxy (nosylate group)).
  • alkylsulfonyloxy groups for example methyl sulfonyloxy (mesylate group) and trifluoromethylsulfonyloxy (triflate group)
  • arylsulfonyloxy groups for example /?-toluenesulfonyloxy (tosylate group) and /?-nitrosulfonyloxy (nosylate group)
  • Optionally substituted unless otherwise specified means that a group may be unsub
  • an optionally substituted group has 1 substituent. In another embodiment an optionally substituted group has 2 substituents. In another embodiment an optionally substituted group has 3 substituents.
  • divalent groups are described generically without specific bonding configurations, for example in the group -CH 2 C(0)-. It is understood that the generic description is meant to include both bonding configurations, unless specified otherwise. For example, in the group R'-R ⁇ R 3 , if the group R 2 is described as -CH 2 C(0)-, then it is understood that this group can be bonded both as R -CH 2 C(0)-R 3 , and as R -C(0)CH 2 -R 3 , unless specified otherwise.
  • Package insert is used to refer to instructions customarily included in commercial packages of therapeutic products that contain information about the indications, usage, dosage, administration, contraindications and/or warnings concerning the use of such therapeutic products.
  • “Pharmaceutically acceptable salts” include both acid and base addition salts.
  • “Pharmaceutically acceptable acid addition salt” refers to those salts which retain the biological effectiveness and properties of the free bases and which are not biologically or otherwise undesirable, formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, carbonic acid, phosphoric acid and the like, and organic acids may be selected from aliphatic, cycloaliphatic, aromatic, araliphatic, heterocyclic, carboxylic, and sulfonic classes of organic acids such as formic acid, acetic acid, propionic acid, glycolic acid, gluconic acid, lactic acid, pyruvic acid, oxalic acid, malic acid, maleic acid, maloneic acid, succinic acid, fumaric acid, tartaric acid, citric acid, aspartic acid, ascorbic acid, glutamic acid, anthranilic acid, benzoic acid, cinnamic acid, mandelic acid, embonic acid, phenylacetic acid, methanes
  • “Pharmaceutically acceptable base addition salts” include those derived from inorganic bases such as sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum salts and the like. Particularly base addition salts are the ammonium, potassium, sodium, calcium and magnesium salts.
  • Salts derived from pharmaceutically acceptable organic nontoxic bases includes salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, 2-diethylaminoethanol, tromethamine, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, ethylenediamine, glucosamine, methylglucamine, theobromine, purines, piperizine, piperidine, N-ethylpiperidine, polyamine resins and the like.
  • Particularly organic non-toxic bases are isopropylamine, diethylamine, ethanolamine, tromethamine, dicyclohexylamine, choline, and caffeine.
  • a “sterile” formulation is aseptic or free from all living microorganisms and their spores.
  • “Stereoisomers” refers to compounds which have identical chemical constitution, but differ with regard to the arrangement of the atoms or groups in space. Stereoisomers include diastereomers, enantiomers, conformers and the like.
  • “Chiral” refers to molecules which have the property of non-superimposability of the mirror image partner, while the term “achiral” refers to molecules which are superimposable on their mirror image partner.
  • Diastereomer refers to a stereoisomer with two or more centers of chirality and whose molecules are not mirror images of one another. Diastereomers have different physical properties, e.g. melting points, boiling points, spectral properties or biological activities. Mixtures of diastereomers may separate under high resolution analytical procedures such as electrophoresis and chromatography such as HPLC.
  • Enantiomers refer to two stereoisomers of a compound which are non-superimposable mirror images of one another.
  • d and 1 or (+) and (-) are employed to designate the sign of rotation of plane-polarized light by the compound, with (-) or 1 meaning that the compound is levorotatory.
  • a compound prefixed with (+) or d is dextrorotatory.
  • these stereoisomers are identical except that they are mirror images of one another.
  • a specific stereoisomer may also be referred to as an enantiomer, and a mixture of such isomers is often called an enantiomeric mixture.
  • a 50:50 mixture of enantiomers is referred to as a racemic mixture or a racemate, which may occur where there has been no stereoselection or stereospecificity in a chemical reaction or process.
  • racemic mixture and racemate refer to an equimolar mixture of two enantiomeric species, devoid of optical activity.
  • tautomer or "tautomeric form” refers to structural isomers of different energies which are interconvertible via a low energy barrier.
  • proton tautomers also known as prototropic tautomers
  • Valence tautomers include interconversions by reorganization of some of the bonding electrons.
  • a “solvate” refers to an association or complex of one or more solvent molecules and a compound of the present invention. Examples of solvents that form solvates include water, isopropanol, ethanol, methanol, DMSO, ethyl acetate, acetic acid, and ethanolamine.
  • hydrate refers to the complex where the solvent molecule is water.
  • a "subject,” “individual,” or “patient” is a vertebrate. In certain embodiments, the vertebrate is a mammal. Mammals include, but are not limited to, farm animals (such as cows), sport animals, pets (such as cats, dogs, and horses), primates, mice and rats. In certain embodiments, a mammal is a human.
  • “Therapeutically effective amount” means an amount of a compound of the present invention that (i) treats or prevents the particular disease, condition or disorder, (ii) attenuates, ameliorates or eliminates one or more symptoms of the particular disease, condition, or disorder, or (iii) prevents or delays the onset of one or more symptoms of the particular disease, condition or disorder described herein.
  • the therapeutically effective amount of the drug may reduce the number of cancer cells; reduce the tumor size; inhibit (i.e., slow to some extent and preferably stop) cancer cell infiltration into peripheral organs; inhibit (i.e., slow to some extent and preferably stop) tumor metastasis; inhibit, to some extent, tumor growth; and/or relieve to some extent one or more of the symptoms associated with the cancer.
  • the drug may prevent growth and/or kill existing cancer cells, it may be cytostatic and/or cytotoxic.
  • efficacy can, for example, be measured by assessing the time to disease progression (TTP) and/or determining the response rate (RR).
  • the therapeutic effective amount is an amount sufficient to decrease or alleviate an allergic disorder, the symptoms of an autoimmune and/or inflammatory disease, or the symptoms of an acute inflammatory reaction (e.g. asthma).
  • a therapeutically effective amount is an amount of a chemical entity described herein sufficient to significantly decrease the activity or number of B-cells.
  • Treatment refers to clinical intervention in an attempt to alter the natural course of the individual or cell being treated, and can be performed either for prophylaxis or during the course of clinical pathology. Desirable effects of treatment include preventing occurrence or recurrence of disease, alleviation of symptoms, diminishment of any direct or indirect pathological consequences of the disease, stabilized (i.e., not worsening) state of disease, preventing metastasis, decreasing the rate of disease progression, amelioration or palliation of the disease state, prolonging survival as compared to expected survival if not receiving treatment and remission or improved prognosis.
  • compounds of the invention are used to delay development of a disease or disorder or to slow the progression of a disease or disorder.
  • Those in need of treatment include those already with the condition or disorder as well as those prone to have the condition or disorder, (for example, through a genetic mutation) or those in which the condition or disorder is to be prevented.
  • the terms "compound(s) of this invention," and “compound(s) of the present invention”, unless otherwise indicated, include compounds of formula I and stereoisomers, tautomers, solvates, metabolites, salts (e.g., pharmaceutically acceptable salts), and prodrugs thereof.
  • structures depicted herein are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms.
  • compounds of formulas I, II and III, wherein one or more hydrogen atoms are replaced by deuterium or tritium, or one or more carbon atoms are replaced by 13 C- or 14 C-enriched carbon are within the scope of this invention.
  • One aspect of the invention provides compounds of formula I:
  • R 2 is absent, Ci_ 6 alkyl, C 2 - 6 alkenyl, C 2 - 6 alkynyl, -(Ci_ 6 alkylene)-, -(C 2 - 6 alkenylene)-, -(C 2 -6 alkynylene)-, -(C 0 _6 alkylene)CN, -(C 0-3 alkylene)NR a (C 0 _ 3 alkylene)-, -(C 0-3 alkylene)0(Co_ 3 alkylene)-, -(C0-3 alkylene)C(O)(C 0 _ 3 alkylene)-, -(C0-3 alkylene)NR a C(O)(C 0 _ 3 alkylene)-, -(C 0 _ 3 alkylene)C(O)NR a (C 0 _ 3 alkylene)-, -(C 0 _ 3 alkylene)C(O)NR a (C 0
  • alkylene)S(0)i_ 2 (Co_ 3 alkylene)-, -(C 0 - 3 alkylene)NR a S(O)i_ 2 (C 0 - 3 alkylene)-, -(C 0 - 3 alkylene)S(O)i_ 2 NR a (C 0 - 3 alkylene)- or -(C 0 _ 3 alkylene)NR a S(O)i_ 2 NR b (C 0 - 3 alkylene)-, wherein said alkyl, alkyenyl, alkynyl, alkylene, alkenylene and alkynylene are independently optionally substituted by halogen, oxo, -CN, -OR c , -SR C , -NR c R d or Ci_ 3 alkyl optionally substituted by halogen;
  • R 3 is absent, hydrogen, Ci_ 6 alkyl, C 2 _ 6 alkenyl, C 2 _ 6 alkynyl, C 3 _ 7 cycloalkyl, C 6-14 aryl or 3-20 membered heterocyclyl, wherein R 3 is independently optionally substituted by R 6 ;
  • R 4 is hydrogen, halogen or Ci_ 3 alkyl;
  • R 5 is halogen, C 1-12 alkyl, C 2-12 alkenyl, C 2-12 alkynyl, -(C 0-3 alkylene)CN, -(C 0-3 alkylene)NR a R b , -(C 0 . 3 alkylene)OR a , -(C 0 . 3 alkylene)SR a , -(C 0 . 3 alkylene)C(0)R a , -(C 0 . 3 alkylene)NR a C(0)R b , -(C 0 . 3 alkylene)C(0)NR a R b , -(C 0 . 3 alkylene)C(0)OR a , -(C 0 . 3 alkylene)C(0)OR a , -(C 0 . 3
  • alkylene)OC(0)R a , -(C 0 . 3 alkylene)NR a C(0)NR a R b , -(C 0 . 3 alkylene)OC(0)NR a R b , -(C 0 . 3 alkylene)NR a C(0)OR b , -(C 0 . 3 alkylene) S(0)i_ 2 R a , -(C 0 . 3 alkylene)NR a S(0)i_ 2 R b , -(C 0 . 3 alkylene)S(0)i_ 2 NR a R b , -(C 0 . 3 alkylene)S(0)i_ 2 NR a R b , -(C 0 .
  • alkylene)C(0)3-12 membered heterocyclyl wherein said C 2 _i 2 alkyl, alkenyl, alkynyl, alkylene, cycloalkyl, aryl and heterocyclyl are independently optionally substituted by halogen, oxo, -(C 0- 3 alkylene)CN, -(C 0 . 3 alkylene)OR c , -(C 0 . 3 alkylene)NR c R d , -(C 0 . 3 alkylene)C(0)R c , -(C 0 . 3 alkylene)C(0)OR c , -(C 0 . 3 alkylene)C(0)NR c R d , -(C 0 . 3 alkylene)NR c C(0)R d , -(C 0 . 3 alkylene)NR c C(0)R d , -(C 0 . 3 alkylene)NR c C(0)R
  • Ci_ 6 alkyl optionally substituted by oxo, -CN or halogen; and wherein said Ci alkyl is independently optionally substituted by halogen, oxo, -(C 0-3
  • alkylene)CN -(Ci_ 3 alkylene)OR c , -(C 0 . 3 alkylene)NR c R d , -(C 0 . 3 alkylene)C(0)R c , -(C 0 . 3 alkylene)C(0)OR c , -(C 0 . 3 alkylene)C(0)NR c R d , -(C 0 . 3 alkylene)NR c C(0)R d , -(C 0 . 3
  • R 6 is independently oxo, halogen, -CN, -C(0)R a , -C(0)OR a , -NR a C(0)R b , -C(0)NR a R b , -NR a C(0)NR a R b , -OC(0)NR a R b , -NR a C(0)OR b , -S(0)i_ 2 R a , -NR a S(0) 2 R a , -S(0) 2 NR a R b , -OR a , -SR a , -NR a R b , Ci_ 6 alkyl, C 3 _ 6 cycloalkyl, C 2 _ 6 alkenyl, C 2 _ 6 alkynyl, 3-7 membered heterocycly or C 6 -i4 aryl, and wherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl and aryl
  • R 1 , R 2 and R 3 are not absent at the same time.
  • R 5 is other than OH.
  • X is CR 4 . In another embodiment, X is N. In one embodiment, Y is CR 5 . In another embodiment, Y is N.
  • X is CR 4 and Y is CR 5 .
  • X is CR 4 and Y is N.
  • X is N and Y is CR 5 .
  • X is N and Y is N.
  • R 1 is absent. In one embodiment, R 1 is absent with the proviso that
  • R 1 , R 2 and R 3 are not all absent at the same time.
  • R 1 is a 3-20 membered heterocyclyl, wherein R 1 is independently optionally substituted by halogen, oxo, -CN, -OR a , -SR a , -NR a R b , or Ci_ 6 alkyl optionally substituted by oxo, -CN or halogen.
  • R 1 is a 3-12 membered heterocyclyl optionally substituted by halogen, oxo, -CN, -OR a , -NR a R b , or Ci_ 6 alkyl optionally substituted by halogen.
  • R 1 is a 4-7 membered heterocyclyl optionally substituted by halogen, oxo, or Ci_ 6 alkyl optionally substituted by halogen, wherein said heterocyclyl is selected from oxetanyl, azetidinyl, thietanyl, tetrahydrofuranyl, 2,3-dihydrofuranyl, tetrahydrothienyl, 2,3-dihydrothienyl, pyrrolidinyl, 2,3-dihydro-lH-pyrrolyl, imidazolidinyl, 2H- pyranyl, tetrahydropyranyl, morpholinyl, piperazinyl, hexahydropyrimidinyl, oxazinanyl, thiazinanyl, piperidinyl, 8-azabicyclo[3.2.1]octanyl, 2-azabicyclo[2.2.2]octanyl, oxe
  • R 1 is azetidinyl, pyrrolidinyl, imidazolidinyl, morpholinyl, piperazinyl, hexahydropyrimidinyl, or piperidinyl, wherein R 1 is optionally substituted by halogen, oxo, or Ci_ 6 alkyl optionally substituted by halogen.
  • R 1 is 4 , 5 , 6, 7-tetrahydrobenzo imidazo ly 1, 4,5,6 , 7-tetrahydro [2H] indazo ly 1, oxazo lidiny 1, thiazo lidiny 1, isothiazolidinyl, 1 , 1-dioxoisothiazolidinyl, oxazolidinonyl, 3-azabicyclo[3.1.0]hexanyl or imidazo lidinonyl, wherein R 1 is independently optionally substituted by halogen, oxo, -CN, -OR a , -SR a , -NR a R b , or Ci_ 6 alkyl optionally substituted by oxo, -CN or halogen.
  • R 1 is piperidinyl or tetrahydropyranyl wherein R 1 is independently optionally substituted by halogen, oxo, -CN, -OR a , -SR a , -NR a R b , or Ci_ 6 alkyl optionally substituted by oxo, -CN or halogen.
  • R 1 is a 3-20 membered heterocyclyl, wherein R 1 is independently optionally substituted by halogen, oxo, -CN, -OR a , -SR a , -NR a R b , or Ci_ 6 alkyl optionally substituted by oxo, -CN or halogen, with the proviso that R 5 is other than hydrogen or -OH.
  • R 1 is a 3-12 membered heterocyclyl, wherein R 1 is independently optionally substituted by halogen, oxo, -CN, -OR a , -SR a , -NR a R b , or Ci_ 6 alkyl optionally substituted by oxo, -CN or halogen.
  • R 1 is a 3-12 membered heterocyclyl optionally substituted by halogen, oxo, -CN, -OR a , -NR a R b , or Ci_ 6 alkyl, wherein said heterocyclyl is selected from oxetanyl, azetidinyl, thietanyl, tetrahydrofuranyl, 2,3- dihydrofuranyl, tetrahydrothienyl, 2,3-dihydrothienyl, pyrrolidinyl, 2,3-dihydro-lH-pyrrolyl, imidazo lidiny 1, 2H-pyranyl, tetrahydropyranyl, morpholinyl, piperazinyl, hexahydropyrimidinyl, oxazinanyl, thiazinanyl, piperidinyl, 8-azabicyclo[3.2.1]octanyl, 2-azabicyclo[2.2.2
  • R 1 represents the point of attachment
  • R 1 is azetidinyl, pyrrolidinyl or piperidinyl, optionally substituted by 1 or 2 halogen, oxo, or Ci_ 6 alkyl optionally substituted by halogen.
  • R 1 is azetidinyl, pyrrolidinyl or piperidinyl, optionally substituted by 1 or 2 halogen, oxo, -CN, -OR a , -NR a R b , or Ci_ 6 alkyl optionally substituted by halogen halogen, oxo, -CN, -OR a , -NR a R b , or Ci_ 6 alkyl optionally substituted by halogen
  • R 1 is morpholinyl, piperazinyl, 2-azabicyclo[2.2.2]octanyl, 8- azabicyclo[3.2.1]octanyl or piperidinyl, optionally substituted by 1 or 2 halogen, oxo, -CN, -OR a , -SR a , -NR a R b , or Ci_ 6 alkyl optionally substituted by oxo, -CN or halogen.
  • R 1 is morpholinyl, piperazinyl, 2-azabicyclo[2.2.2]octanyl, 8- azabicyclo[3.2.1]octanyl or piperidinyl, optionally substituted by 1 or 2 halogen, oxo or Ci_ 6 alkyl optionally substituted by halogen.
  • R 1 is piperidinyl optionally substituted by halogen, oxo, -CN, -OR a , -SR a , -NR a R b , or Ci_ 6 alkyl optionally substituted by oxo, -CN or halogen.
  • R 1 is piperidinyl optionally substituted by 1 or 2 halogen, oxo, -CN, -OR a , -NR a R b , or Ci_6 alkyl optionally substituted by halogen.
  • R 1 is piperidinyl optionally substituted by 1 or 2 halogen, oxo or Ci_ 6 alkyl.
  • R 1 is piperidinyl optionally substituted by methyl, oxo, fluoro or methoxy.
  • R 1 is piperidin-3-yl, piperidin-4-yl, 2-methylpiperidin-3-yl or 2-methylpiperidin-4- yl. In another embodiment, R 1 is (R)-piperidin-3-yl. In another embodiment, R 1 is (S)- piperidin-3-yl. In another embodiment, R 1 is substituted (R)-piperidin-4-yl, wherein said piperidinyl is substituted by 1-3 groups selected from oxo, Ci_ 3 alkyl, halogen or -OR a .
  • R 1 is substituted (S)-piperidin-4-yl, wherein said piperidinyl is substituted by 1-3 groups selected from oxo, Ci_ 3 alkyl, halogen or -OR a .
  • R 1 is (R)- (R)-2-methylpiperidin-4-yl, (R)-(S)-2-methylpiperidin-4-yl, (S)-(R)-2-methylpiperidin-4-yl or (S)-(S)-2-methylpiperidin-4-yl.
  • R 1 is (R)-(R)-3-fluoropiperidin-4-yl, (R)-(S)-3-fluoropiperidin-4-yl, (S)-(R)-3-fluoropiperidin-4-yl or (S)-(S)-3-fluoropiperidin-4-yl.
  • R 1 is piperidinonyl, 2-methylpiperidin-4-yl, 3-methylpiperidin-4-yl, 4- methylpiperidin-4-yl, 2-fluoropiperidinyl, 3-fluoropiperidin-4-yl, 3,3-difluoropiperidin-4-yl, 3-
  • R 1 is piperidinyl optionally substituted by halogen, oxo, -CN, -OR a , -SR a , -NR a R b , or Ci_ 6 alkyl optionally substituted by oxo, -CN or halogen, with the proviso that R 5 is other than hydrogen or -OH.
  • R 1 is tetrahydropyranyl optionally substituted by halogen, oxo, -CN, -OR a , -SR a , -NR a R b , or Ci_ 6 alkyl optionally substituted by oxo, -CN or halogen, with the proviso that R 5 is other than hydrogen or -OH.
  • R 1 is tetrahydropyranyl optionally substituted by halogen, oxo, -CN, -OR a , -SR a , -NR a R b , or Ci_ 6 alkyl optionally substituted by oxo, -CN or halogen, and R 2 and R 3 are both absent, with the proviso that R 5 is other than hydrogen or -OH.
  • R 1 is (R)-pyrrolidin-3-yl. In another embodiment, R 1 is (S)- pyrrolidin-3-yl.
  • R 1 is a C 4 _7 cycloalkyl, wherein R 1 is independently optionally substituted by halogen, oxo, -CN, -OR a , -SR a , -NR a R b , or Ci_ 6 alkyl optionally substituted by oxo, -CN or halogen.
  • R 1 is a C 4 -7 cycloalkyl optionally substituted by halogen, oxo, -CN, -OR a , -NR a R b , or Ci_ 6 alkyl optionally substituted by halogen.
  • R 1 is a C 4 -7 cycloalkyl optionally substituted by halogen, oxo, or Ci_ 6 alkyl optionally substituted by halogen.
  • said cycloalkyl is cyclopropyl, eye lo butyl, cyclopentyl or cyclohexyl.
  • R 1 is selected from
  • R 1 is cyclohexyl optionally substituted by halogen, oxo, -CN,
  • R 1 is cyclohexyl optionally substituted by halogen, oxo, -CN, -OR a , -NR a R b , or Ci_6 alkyl optionally substituted by halogen.
  • R 1 is cyclohexyl optionally substituted by halogen, oxo, or Ci_ 6 alkyl optionally substituted by halogen.
  • R 1 is cyclohexyl optionally substituted by halogen, oxo, or Ci_ 6 alkyl optionally substituted by halogen.
  • R 1 is cyclohexyl.
  • R 1 is selected from cyclohexyl, 2- hydroxycyclohexyl, 3-hydroxycyclohexyl, 4-hydroxycyclohexyl, bicyclo[2.2.1]heptanyl, 2- methylcyclohexyl or 4,4-difluorocyclohexyl,
  • R is optionally substituted by halogen, oxo, -CN, -OR a , -NR a R , or Ci_ 6 alkyl optionally substituted by halogen, and wherein the wavy line represents the point of attachment in formula I.
  • R 1 is selected from
  • R 2 -R 3 is selected from
  • R 1 is selected from r 2 -* 3 and ? wherein the wavy line represents the point of attachment in formula I.
  • R 2 and R 3 are absent, and R 1 is selected from ? wherein R i o is halogen, oxo, -CN, -OR a , -SR a , -NR a R b , or
  • Ci_6 alkyl optionally substituted by oxo, -CN or halogen, and wherein the wavy line represents the point of attachment in formula I.
  • R 5 is other than hydrogen or -OH.
  • R 2 and R 3 are absent, and R 1 is selected from , wherein R 10 is -OH, -NH(CH 2 CF 3 ), -CN, -CH 2 CN, -CH 2 CH 2 CN or halogen, and wherein the wavy line represents the point of attachment in formula I.
  • R 5 is other than hydrogen or -OH.
  • R 1 is cyclopentyl optionally substituted by halogen, oxo, -CN,
  • R 1 is cyclopentyl optionally substituted by halogen, oxo, -CN, -OR a , -NR a R b , or Ci_6 alkyl optionally substituted by halogen.
  • R 1 is cyclopentyl.
  • R 1 is C 5 _6 cycloalkyl or 5-6 membered heterocyclyl, wherein R 1 is independently optionally substituted by halogen, oxo, -CN, -OR a , -SR a , -NR a R b , Ci_ 3 alkylene, -S(0) 2 R a , or Ci_6 alkyl optionally substituted by oxo, -CN or halogen.
  • R 1 is cyclohexyl, cyclopentyl, piperidinyl, or tetrahydropyranyl, wherein R 1 is independently optionally substituted by halogen, oxo, -CN, -OR a , -SR a , -NR a R b , Ci_ 3 alkylene, -S(0) 2 R a , or Ci_ 6 alkyl optionally substituted by oxo, -CN or halogen.
  • R 1 is selected from:
  • -R 1 -R 2 -R 3 taken together are selected from:
  • R 1 is C 3 _8 cycloalkyl, or 4-10 membered heterocyclyl, wherein R 1 is independently optionally substituted by halogen, oxo, -CN, -OR a , -SR a , -NR a R b , Ci_ 3 alkylene, -C(0)OR a , -S(0) 2 R a , or Ci_ 6 alkyl optionally substituted by oxo, -CN, -S(0) 2 R a , C 6 _i 4 aryl, or halogen.
  • R 1 is C 5 _6 cycloalkyl or 5-6 membered heterocyclyl, wherein R 1 is independently optionally substituted by halogen, oxo, -CN, -OR a , -SR a , -NR a R b , Ci_ 3 alkylene, -C(0)OR a , -S(0) 2 R a , or Ci_ 6 alkyl optionally substituted by oxo, -CN, -S(0) 2 R a , C 6 _i 4 aryl, or halogen.
  • R 1 is cyclohexyl, cyclopentyl, piperidinyl, or tetrahydropyranyl, wherein R 1 is independently optionally substituted by halogen, oxo, -CN, -OR a , -SR a , -NR a R b , Ci_ 3 alkylene, -C(0)OR a , -S(0) 2 R a , or Ci_ 6 alkyl optionally substituted by oxo, -CN, -S(0) 2 R a , C 6-14 aryl, or halogen.
  • R 2 is absent. In one embodiment, R 2 is absent with the proviso that R 1 , R 2 and R 3 are not all absent at the same time. In one embodiment, R 2 and R 3 are absent. . In one embodiment, R 2 and R 3 are absent with the proviso that R 1 , R 2 and R 3 are not all absent at the same time.
  • R 2 is Ci_ 6 alkyl, C 2 _ 6 alkenyl, C 2 _ 6 alkynyl, wherein said alkyl, alkenyl or alkynyl are independently optionally substituted by halogen, oxo, -CN, -OR c , -SR C , -NR c R d or Ci_3 alkyl optionally substituted by halogen, and R 3 is absent.
  • R 2 is Ci_6 alkyl, C 2 _ 6 alkenyl, C 2 _ 6 alkynyl, wherein said alkyl, alkenyl or alkynyl are independently optionally substituted by halogen, oxo, -CN, -OR c , -NR c R d or Ci_ 3 alkyl optionally substituted by halogen, and R 3 is absent.
  • R 2 is selected from -CH 2 CF 3 -CH 2 CH 2 CF 3 , -
  • R 2 is -(Ci_ 6 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN, -OR c , -SR C , -NR c R d or Ci_ 3 alkyl optionally substituted by halogen.
  • R 2 is -(Ci_ 6 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN, -OR c , -NR c R d or Ci_ 3 alkyl optionally substituted by halogen.
  • R 2 is -(Ci_ 6 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN or Ci_ 3 alkyl.
  • R 2 is methylene, ethylene, -CH(CH 3 )-, -C(CH 3 ) 2 -, propylene or butylene, optionally substituted by halogen, oxo, -CN, -OR c , -NR c R d
  • R 2 is selected from methylene, ethylene,
  • R 2 is -(Co- 6 alkylene)CN, wherein said alkylene is optionally substituted by halogen, oxo, -CN, -OR c , -SR C , -NR c R d or Ci_ 3 alkyl optionally substituted by halogen, and R 3 is absent.
  • R 2 is -(Ci_ 6 alkylene)CN, wherein said alkylene is optionally substituted by halogen, oxo, -CN, -OR c , -NR c R d or Ci_ 3 alkyl optionally substituted by halogen, and R 3 is absent.
  • R 2 is -CH 2 CN, -CH 2 CH 2 CN, -CH(CH 3 )CN or -CH(CH 3 )CH 2 CN and R 3 is absent.
  • R 1 is a 3-20 membered heterocyclyl or C 3 _i 2 cycloalkyl, wherein R 1 is independently optionally substituted by halogen, oxo, -CN, -OR a , -SR a , -NR a R b , Ci_ 3 alkylene or Ci_ 6 alkyl optionally substituted by oxo, -CN or halogen, R 2 is -CN, -CH 2 CN, -CH2CH2CN, -CH(CH 3 )CN or -CH(CH 3 )CH 2 CN, and R 3 is absent, with the proviso that R 5 is other than hydrogen or -OH.
  • R 2 is -(C 0-3 alkylene)NR a (Co- 3 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN, -OR c , -SR C , -NR c R d or Ci_ 3 alkyl optionally substituted by halogen.
  • R 2 is -(C 0-3 alkylene)NR a (Co_ 3 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN, -OR c , -NR c R d or Ci_ 3 alkyl optionally substituted by halogen.
  • R 2 is -(C 0-3 alkylene)NR a (Co_ 3 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN or Ci_ 3 alkyl.
  • R 2 is - H-, -NHCH 2 - or -NHCH 2 CH 2 -
  • R 2 is -(C 0-3 alkylene)0(Co_ 3 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN, -OR c , -SR C , -NR c R d or Ci_ 3 alkyl optionally substituted by halogen.
  • R 2 is -(C 0-3 alkylene)0(Co_ 3 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN, -OR c , -NR c R d or Ci_ 3 alkyl optionally substituted by halogen.
  • R 2 is -(C 0-3 alkylene)0(Co_ 3 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN or Ci_ 3 alkyl.
  • R 2 is -CH 2 0- -CH 2 C(CH 2 ) 2 0- or -(CH 2 ) 2 0-
  • R 2 is -(Co_ 3 alkylene)NR a C(0)(Co_ 3 alkylene)- or -(Co_ 3 alkylene)C(0)NR a (Co_ 3 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN, -OR c , -SR C , -NR c R d or Ci_ 3 alkyl optionally substituted by halogen.
  • R 2 is -(C 0 _ 3 alkylene)NR a C(O)(C 0 _ 3 alkylene)- or -(C 0 _ 3 alkylene)C(O)NR a (C 0 _ 3 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN, -OR c , -NR c R d or Ci_ 3 alkyl optionally substituted by halogen.
  • R 2 is -(Co_ 3 alkylene)NR a C(0)(Co_ 3 alkylene)- or -(C 0 _ 3 alkylene)C(O)NR a (C 0 _ 3 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN or Ci_ 3 alkyl.
  • R 2 is -C(0)NH- -CH 2 C(0)NH- or -CH 2 C(0)N(CH 3 )-.
  • R 2 is -NHC(O)- or -NHC(0)CH 2 -.
  • R 2 is -(C 0-3 alkylene)OC(O)NR a (C 0 _ 3 alkylene)- or -(C 0-3 alkylene)NR a C(0)0(Co_ 3 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN, -OR c , -SR C , -NR c R d or Ci_ 3 alkyl optionally substituted by halogen.
  • R 2 is -(C0-3 alkylene)OC(O)NR a (C 0 _ 3 alkylene)- or -(C 0 _ 3 alkylene)NR a C(O)O(C 0 _ 3 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN, -OR c , -NR c R d or Ci_ 3 alkyl optionally substituted by halogen.
  • R 2 is -(Co_ 3 alkylene)OC(0)NR a (Co-3 alkylene)- or -(C 0 _ 3 alkylene)NR a C(O)O(C 0 - 3 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN or Ci_ 3 alkyl.
  • R 2 is -NHC(0)0- -N(CH 3 )C(0)0-, -NHC(0)OCH 2 - or -NHC(0)OCH 2 CH 2 -.
  • R 2 is -(Co_ 3 alkylene)C(0)(Co_ 3 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN, -OR c , -SR C , -NR c R d or Ci_ 3 alkyl optionally substituted by halogen.
  • R 2 is -(Co_ 3 alkylene)C(0)(Co_ 3 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN, -OR c , -NR c R d or Ci_ 3 alkyl optionally substituted by halogen.
  • R 2 is -(Co_ 3 alkylene)C(0)(Co_ 3 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN or Ci_ 3 alkyl. In another embodiment, R 2 is selected from:
  • R 2 is -(Co_ 3 alkylene)C(0)0(Co_ 3 alkylene)- or -(Co_ 3 alkylene)OC(0)(Co_ 3 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN, -OR c , -SR C , -NR c R d or Ci_ 3 alkyl optionally substituted by halogen.
  • R 2 is -(Co- 3 alkylene)C(0)0(Co_ 3 alkylene)- or -(Co_ 3 alkylene)OC(0)(Co_ 3 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN, -OR c , -NR c R d or Ci_ 3 alkyl optionally substituted by halogen.
  • R 2 is -(Co_ 3 alkylene)C(0)0(Co_ 3 alkylene)- or -(Co_ 3 alkylene)OC(0)(Co_ 3 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN or Ci_ 3 alkyl.
  • R 2 is selected from -C(0)0-
  • R 2 is -(Co_ 3 alkylene)S(0)i_ 2 (Co_ 3 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN, -OR c , -SR C , -NR c R d or Ci_ 3 alkyl optionally substituted by halogen.
  • R 2 is -(Co_ 3 alkylene)S(0)i_ 2 (Co_ 3 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN, -OR c , -NR c R d or Ci_ 3 alkyl optionally substituted by halogen.
  • R 2 is -(Co_ 3 alkylene)S(0)i_ 2 (Co_ 3 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN or Ci_ 3 alkyl.
  • R 2 is selected from -C(0)CH 2 S(0)2,
  • R 2 is -(Co_ 3 alkylene)NR a S(0)i_ 2 (Co- 3 alkylene)- or -(Co_ 3 alkylene)S(0)i_ 2 NR a (Co- 3 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN, -OR c , -SR C , -NR c R d or Ci_ 3 alkyl optionally substituted by halogen.
  • R 2 is -(C 0 - 3 alkylene)NR a S(O)i_ 2 (C 0 - 3 alkylene)- or -(C 0 _ 3 alkylene)S(O)i_ 2 NR a (C 0 _ 3 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN, -OR c , -NR c R d or Ci_ 3 alkyl optionally substituted by halogen.
  • R 2 is -(C 0- 3 alkylene)NR a S(0)i_ 2 (Co_3 alkylene)- or -(C 0 - 3 alkylene)S(O)i_ 2 NR a (C 0 - 3 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN or Ci_3 alkyl.
  • R 2 is -NHS(0) 2 - -N(CH 3 )S(0) 2 - or -NHS(0) 2 CH 2 -.
  • R 2 is selected from absent, -NHS(0) 2 -, -N(CH 3 )S(0) 2 - , -NHS(0) 2 CH 2 - -C(0)CH 2 S(0) 2 , "C(0)0-, -NHC(0)0-, -N(CH 3 )C(0)0-, -NHC(0)OCH 2 -, -NHC(0)OCH 2 CH 2 -, -C(0)NH-, -CH 2 C(0)NH-, -CH 2 C(0)N(CH 3 )-, -NHC(O)-, -NHC(0)CH 2 -, -CH 2 0-, -CH 2 C(CH 2 ) 2 0-, -(CH 2 ) 2 0- -NH-, -NHCH 2 -, -NHCH 2 CH 2 -, -CH 2 CN, -CH 2 CH 2 CN, -CH(CH 3 )CN , -CH(CH 3 )CH 2 CN, methylene, ethylene,
  • R 2 is absent, methylene, ethylene, -CH(CH 3 )-, -NH-, - -,
  • R 2 is absent, methylene, ethylene, ? J Ol 0 r wherein the wavy line represents the point of attachment in formula I.
  • R 3 is absent.
  • R 3 is hydrogen
  • -R 2 -R 3 is -CHO.
  • R 2 is absent and R 3 is hydrogen.
  • R 1 and R 2 are absent.
  • R 3 is Ci_ 6 alkyl optionally substituted by 1 to 3 R 6 .
  • R 3 is Ci_ 6 alkyl optionally substituted by 1 to 3 oxo, Ci_ 6 alkyl, halogen, -CN, -S(0)i_ 2 (Ci_6 alkyl), -OR a , -SR a or -NR a R b .
  • R 3 is Ci_ 6 alkyl optionally substituted by 1 to 3 oxo, Ci_ 6 alkyl, halogen, -CN, -S(0) 2 (Ci_ 6 alkyl), -OR a or -NR a R b .
  • R 3 is methyl, ethyl, propyl, isopropyl, butyl, isobutyl or t-butyl optionally substituted by oxo, Ci_ 6 alkyl, halogen, -CN, -S(0) 2 (Ci_ 6 alkyl), -OR a or -NR a R b .
  • R 3 is selected from methyl, ethyl, n-butyl, sec-butyl, t-butyl, -CF 3 , -CH 2 CF 3 , -CH 2 CH 2 F, -CH 2 CH 2 CF 3 , -CH 2 OCH 3 , -CH 2 CH 2 OCH 3 , -CH(CH 2 CH 3 )CH 2 OCH 3 , -CH(CH 3 )CH 2 CH 2 OH, -CH 2 C(CH 3 ) 2 OH, -CH 2 C(CF 3 ) 2 OH, -CH 2 CH 2 OH, -C(CH 3 ) 2 OH, -CH 2 CN, -(CH 2 ) 2 CN, -(CH 2 ) 3 CN, -CH(CH 3 )CH 2 CN, -C(CH 3 ) 2 CN, -CH(CH 3 )CN, -CH 2 NH 2 , -CH(CH 3 )N
  • R 3 is C 3 _ 7 cycloalkyl optionally substituted by 1 to 3 R 6 . In one embodiment, R 3 is C 3 _ 7 cycloalkyl optionally substituted by 1 to 3 oxo, halogen, -CN, -S(0)i_ 2 (Ci_6 alkyl), -OR a , -SR a , -NR a R b or Ci_ 6 alkyl optionally substituted by oxo or halogen.
  • R 3 is C 3 _ 7 cycloalkyl optionally substituted by 1 to 3 oxo, halogen, -CN, -S(0) 2 (Ci_6 alkyl), -OR a , -NR a R b or Ci_ 6 alkyl optionally substituted by halogen.
  • R 3 is cyclopropyl optionally substituted by 1 to 3 oxo, halogen, -CN, -S(0) 2 (Ci_ 6 alkyl), -OR a , -NR a R b or Ci_ 6 alkyl optionally substituted by halogen.
  • R 3 is selected from cyclopropyl, 1-cyanocycloprop-l-yl, 1-trifluoromethylcycloprop-l-yl, 1- methylcycloprop-l-yl, 2-fluorocyclopyrop-l-yl, 2,2-dimethylcycloprop-l-yl, 2- cyanocyclopropyl, eye lo butyl, 4-carboxyclo butyl, 1-cyanocyclobut-l-yl, 4-aminocyclo butyl, cyclopentyl, 3-aminocyclohexyl, 4-aminocyclohexyl, 2-hydroxycyclohexyl, 3- hydroxycyclohexyl, 4-hydroxycyclohexyl and 2-hydroxycyclohexyl.
  • R 3 is C 6 -i4 aryl optionally substituted by 1 to 3 R 6 .
  • R 3 is C 6 -i4 aryl optionally substituted by 1 to 3 Ci_ 6 alkyl, C 2 _ 6 alkenyl, C 2 _ 6 alkynyl, halogen, -CN, -S(0)i_ 2 (Ci_ 6 alkyl), -OR a , -SR a or -NR a R b .
  • R 3 is phenyl optionally substituted by 1 to 3 R 6 .
  • R 3 is phenyl optionally substituted by 1 to 3 Ci_6 alkyl, C 2 _ 6 alkenyl, C 2 _ 6 alkynyl, halogen, -CN, -S(0) 2 (Ci_ 6 alkyl), -OR a or -NR a R b .
  • R 3 is phenyl, 2-chloro-4-cyanophenyl, 2-cyanophenyl, 3-cyanophenyl, 4-cyanophenyl, 3-methylsulfonylphenyl, 3 -fluorophenyl or 4-methoxyphenyl.
  • R 3 is 5-6 membered heteroaryl optionally substituted by 1 to 3 R 6 . In one embodiment, R 3 is 5-6 membered heteroaryl optionally substituted by 1 to 3 oxo, Ci_ 6 alkyl, C 2 _ 6 alkenyl, C 2 _ 6 alkynyl, halogen, -CN, -S(0)i_ 2 (Ci_ 6 alkyl), -OR a , -SR a or -NR a R b .
  • R 3 is 5-6 membered heteroaryl optionally substituted by 1 to 3 oxo, Ci_ 6 alkyl, C 2 -6 alkenyl, C 2 _ 6 alkynyl, halogen, -CN, -S(0) 2 (Ci_ 6 alkyl), -OR a or -NR a R b .
  • R 3 is pyridinyl, thiazolyl, pyrimidinyl, pyrazinyl, oxazolyl, pyrazolyl, imidazolyl, optionally substituted by 1 to 3 oxo, Ci_ 6 alkyl, C 2 _ 6 alkenyl, C 2 _ 6 alkynyl, halogen, -CN, -S(0) 2 (Ci_6 alkyl), -OR a or -NR a R b .
  • R 3 is selected from pyridinyl, pyridin- 3-yl, 6-cyanopyridinyl, 6-trifluoromethylpyridinyl, 2-cyanopyridin-4-yl, 4-cyanopyridin-2-yl, 5- cyanopyridin-2-yl, 3-fluoropyridin-5-yl, thiazol-5-yl, pyrimidin-2-yl, pyrimidin-4-yl, pyrimidin- 5-yl, pyrazin-2-yl, oxazol-2-yl, oxazol-4-yl, l-methylpyrazol-5-yl, l-methylpyrazo -
  • R 3 is selected from thiazol-5-yl and isothiazol-5-yl.
  • R 3 is 3-12 membered heterocyclyl optionally substituted by 1 to 3 R 6 . In one embodiment, R 3 is 4-7 membered heterocyclyl optionally substituted by 1 to 3 R 6 . In one embodiment, R 3 is 4-7 membered heterocyclyl optionally substituted by 1 to 3 oxo, Ci_ 6 alkyl, C 2 _ 6 alkenyl, C 2 _ 6 alkynyl, halogen, -CN, -S(0)i_ 2 R a , -C(0)OR a , -OR a , -SR a or -NR a R b , wherein said alkyl, alkenyl and alkynyl are optionally substituted by oxo, halogen, -CN, -OR c or -NR c R d In one embodiment, R 3 is 4-7 membered heterocyclyl optionally substituted by 1 to 3 oxo, Ci_ 6 alkyl, C 2 _
  • R 3 is selected from oxetan-3-yl, piperidin-3-yl, piperidin-4-yl, N-methylpiperidin-2-yl, N-methylmorpholin-2-yl, 1- methylpyrrolidin-2-yl, pyrrolidinyl, pyrrolidinonyl, piperidinonyl, 3,3-difluoropyrrolidin-2-yl, 1- isopropylpyrrolidin-2-yl, 2-methylpyrrolidin-2-yl, l-methylcyanopyrrolidin-2-yl, 1-
  • R 3 is (S)-l-methylpyrrolidin-2-yl. In one embodiment, R 3 is selected from N-ethylpiperidin-2-yl, N-
  • R is absent, hydrogen, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, t-butyl, cyclopropyl, phenyl, pyridinyl, thiazolyl, pyrimidinyl, pyrazinyl, oxazolyl, pyrazolyl, imidazolyl, oxetanyl, pyrrolidinyl, piperidinyl, pyranyl or morpholinyl, optionally substituted by oxo, Ci_ 6 alkyl, halogen, -CN, -S(0) 2 (Ci_ 6 alkyl), -OR a or -NR a R b .
  • R 3 is absent, hydrogen, methyl, -CF 3 , -CH 2 CN, -(CH 2 ) 2 CN, 1- cyanocycloprop-l-yl, cyclopropyl, phenyl, 3-cyanophenyl, 4-cyanophenyl, 3- methylsulfonylphenyl, 3 -fluorophenyl, 6-cyanopyridinyl, 4-cyanopyridin-2-yl, pyridin-3-yl, pyrazin-2-yl, pyrimidin-5-yl, thiazol-5-yl or oxazol-4-yl.
  • R is selected from absent, hydrogen, methyl, ethyl, n-butyl, sec- butyl, t-butyl, -CF 3 , -CH 2 CF 3 , -CH 2 CH 2 F, -CH 2 CH 2 CF 3 , -CH 2 OCH 3 , -CH 2 CH 2 OCH 3 , -CH(CH 2 CH 3 )CH 2 OCH 3 , -CH(CH 3 )CH 2 CH 2 OH, -CH 2 C(CH 3 ) 2 OH, -CH 2 C(CF 3 ) 2 OH, - CH 2 CH 2 OH, -C(CH 3 ) 2 OH, -CH 2 CN, -(CH 2 ) 2 CN, -(CH 2 ) 3 CN, -CH(CH 3 )CH 2 CN, - C(CH 3 ) 2 CN, -CH(CH 3 )CN, -CH 2 NH 2 , -CH(CH 3 )CH
  • methylmorpholin-2-yl l-methylpyrrolidin-2-yl, pyrrolidinyl, pyrrolidinonyl, piperidinonyl, 3,3- difluoropyrrolidin-2-yl, l-isopropylpyrrolidin-2-yl, 2-methylpyrrolidin-2-yl, 1- methylcyanopyrrolidin-2-yl, l-cyclobutylpyrrolidin-2-yl, morpholinyl, pyran-4-yl, N-
  • R 4 is hydrogen, methyl or F. In another embodiment, R 4 is hydrogen.
  • R 5 is halogen, C 1-12 alkyl, C 2-12 alkenyl, C 2-12 alkynyl, -(C 0 _6 alkylene)CN, -(C 0 . 3 alkylene)NR a R b , -(C 0 . 3 alkylene)OR a , -(C 0 . 3 alkylene)SR a , -(C 0 . 3 alkylene)C(0)R a , -(C 0 . 3 alkylene)NR a C(0)R b , -(C 0 . 3 alkylene)C(0)NR a R b , -(C 0 . 3 alkylene)C(0)NR a R b , -(C 0 .
  • alkylene)NR a S(0)i_ 2 NR a R b -(C 0 . 3 alkylene)C3-6 cycloalkyl, -(C 0-3 alkylene)C6_i4 aryl, -(C 0-3 alkylene)3-12 membered heterocyclyl or -(Co-3 alkylene)C(0)3-12 membered heterocyclyl, wherein said alkyl, alkenyl, alkynyl, alkylene, cycloalkyl, aryl and heterocyclyl are independently optionally substituted by halogen, oxo, -(Co-3 alkylene)CN, -(C 0 .
  • R 5 is halogen, C 1-12 alkyl, C 2 _i 2 alkenyl, C 2 _i 2 alkynyl, -(C 0 _6 alkylene)CN, -(C 0 . 3 alkylene)NR a R b , -(C 0 . 3 alkylene)OR a , -(C 0 . 3 alkylene)SR a , -(C 0 . 3 alkylene)C(0)R a , -(C 0 . 3 alkylene)NR a C(0)R b , -(C 0 . 3 alkylene)C(0)NR a R b , -(C 0 . 3 alkylene)C(0)NR a R b , -(C 0 .
  • alkylene)NR a S(0)i_ 2 NR a R b -(C 0 . 3 alkylene)C3_6 cycloalkyl, -(C 0-3 alkylene)C6_i4 aryl, -(C 0-3 alkylene)3-12 membered heterocyclyl or -(Co-3 alkylene)C(0)3-12 membered heterocyclyl, wherein said alkyl, alkenyl, alkynyl, alkylene, cycloalkyl, aryl and heterocyclyl are independently optionally substituted by halogen, oxo, -(Co-3 alkylene)CN, -(C 0 .
  • R 5 is halogen. In one embodiment, R 5 is F. In one embodiment, R 5 is C 1-12 alkyl, C 2 _i 2 alkenyl, C 2 _i 2 alkynyl, wherein said alkyl, alkenyl and alkynyl are independently optionally substituted by halogen, oxo, -(G 0-3 alkylene)CN, -(C 0 . 3 alkylene)OR c , -(C 0 . 3 alkylene)NR c R d , -(C 0 . 3 alkylene)C(0)R c , -(C 0 . 3 alkylene)C(0)OR c , -(C 0 .
  • R 5 is Ci_ 6 alkyl, C 2 _ 6 alkenyl, C 2 _ 6 alkynyl, wherein said alkyl, alkenyl and alkynyl are independently optionally substituted by halogen, oxo, -CN, -OR c , -NR c R d , -C(0)OR c , or Ci_6 alkyl optionally substituted by oxo, -CN or halogen.
  • R 5 is selected from methyl, ethyl, 1 -hydro xyethyl, 2-hydroxyethyl, propyl, isopropyl, butyl, 2-methylbutyl, 3,3-difluorobut-l-yl, isobutyl, -CH 2 F, -CHF 2 , -CF 3 , -CH 2 OH, -C(CH 3 ) 2 OH,
  • R 5 is C 1-12 alkyl independently optionally substituted by halogen, oxo, -(Co-3 alkylene)CN, -(C 0 . 3 alkylene)OR c , -(C 0 . 3 alkylene)NR c R d , -(C 0 . 3 alkylene)C(0)R c , -(Co-3 alkylene)C(0)OR c , -(C 0 . 3 alkylene)C(0)NR c R d , -(C 0 . 3 alkylene)NR c C(0)R d , -(C 0 .
  • R 5 is Ci_ 6 alkyl optionally substituted by halogen, oxo, -CN, -OR a or -NR a R b .
  • R 5 is methyl, ethyl, propyl, isopropyl or 2-methylpropyl, optionally substituted by halogen, oxo, -CN, -OR a or -NR a R b , wherein R a and R b are independently hydrogen, Ci_ 6 alkyl, C 3 _6 cycloalkyl, 4-6 membered heterocyclyl or taken together with the atom to which they are attached to form a pyrrolidinyl, piperidinyl, piperazinyl or morpholinyl group.
  • R 5 is methyl, hydro xymethyl, 1 -hydro xyethyl, 2-hydroxyethyl, isopropyl or 2-methylpropyl.
  • R 5 is -(C 0-3 alkylene)CN, wherein said alkylene is independently optionally substituted by halogen, oxo, -(C 0-3 alkylene)CN, -(C 0-3 alkylene)OR c , -(C 0-3 alkylene)NR c R d , -(C 0 . 3 alkylene)C(0)R c , -(C 0 . 3 alkylene)C(0)OR c , -(C 0 . 3 alkylene)C(0)NR c R d , -(Co-3 alkylene)NR c C(0)R d , -(C 0 .
  • R 5 is -(C 0-3 alkylene)CN, wherein said alkylene is optionally substituted by halogen, oxo, -CN, -OR c , -NR c R d , -C(0)OR c , or Ci_ 6 alkyl optionally substituted by oxo, -CN or halogen.
  • R 5 is -(Ci_ 6 alkylene)CN, wherein said alkylene is optionally substituted by halogen, oxo, -CN, -OR c , -NR c R d , -C(0)OR c , or Ci_ 6 alkyl optionally substituted by oxo, -CN or halogen.
  • R 5 is selected from - CN, -C(CH 3 ) 2 CN.
  • R 5 is -CN.
  • R 5 is -(C 0-3 alkylene)OR a or -(C 0-3 alkylene)SR a , wherein said alkylene is independently optionally substituted by halogen, oxo, -(C 0-3 alkylene)CN, -(C 0-3 alkylene)OR c , -(C 0 . 3 alkylene)NR c R d , -(C 0 . 3 alkylene)C(0)R c , -(C 0 . 3 alkylene)C(0)OR c , -(C 0 . 3 alkylene)C(0)NR c R d , -(C 0 .
  • R 5 is -(C 0-3 alkylene)OR a , wherein said alkylene is independently optionally substituted by halogen, oxo, -(Co-3 alkylene)CN, -(C 0 . 3 alkylene)OR c , -(C 0 . 3 alkylene)NR c R d , -(C 0 . 3 alkylene)C(0)OR c , -(Co-3 alkylene)C(0)NR c R d , -(C 0 .
  • R 5 is -(C 0-3 alkylene)OR a , wherein said alkylene is optionally substituted by halogen, oxo, -CN, -OR c , -NR c R d , -C(0)OR c , or Ci_ 6 alkyl optionally substituted by oxo, -CN or halogen.
  • R 5 is selected from -CH 2 OH, -CH 2 CH 2 OH, -C(CH 3 ) 2 OH, -CH 2 C(CH 3 ) 2 OH, -
  • R 5 is -(C 0-3 alkylene)NR a R b , wherein said alkylene is independently optionally substituted by halogen, oxo, -(C 0-3 alkylene)CN, -(C 0-3 alkylene)OR c 0-3 alkylene)NR c R d , -(C 0 . 3 alkylene)C(0)R c , -(C 0 . 3 alkylene)C(0)OR c , -(C 0 . 3 alkylene)C(0)NR c R d , -(Co-3 alkylene)NR c C(0)R d , -(C 0 .
  • R 5 is -(C 0-3 alkylene)NR a R b , wherein said alkylene is optionally substituted by halogen, oxo, -CN, -OR c , -NR c R d , -C(0)OR c , or Ci_ 6 alkyl optionally substituted b oxo -CN or halo en.
  • R 5 is selected from -NHCH 2 CH 2 OH, -
  • R 5 is -(C 0-3 alkylene)C3_i 2 cycloalkyl, wherein said alkylene and cycloalkyl are independently optionally substituted by halogen, oxo, -(C 0-3 alkylene)CN, -(C 0-3 alkylene)OR c , -(C 0 . 3 alkylene)NR c R d , -(C 0 . 3 alkylene)C(0)R c , -(C 0 . 3 alkylene)C(0)OR c , -(C 0 .
  • R 5 is -(C 0-3 alkylene)C 3 _6 cycloalkyl, wherein said alkylene and cycloalkyl are independently optionally substituted by halogen, oxo, -CN, -OR c , -NR c R d , -C(0)OR c , or Ci_ 6 alkyl optionally substituted by oxo, -CN or halogen.
  • R 5 is selected from -CH 2 cyclopentyl, -CH 2 cyclopropyl, -CH 2 CH 2 cyclopropyl, cyclopropyl, 2,2-difluorocyclopropyl and cyclobutyl.
  • R 5 is -(C 0-3 alkylene)C 3 _7 cycloalkyl. In another embodiment, R 5 is cyclopropyl or cyclobutyl.
  • R 5 is -(C 0-3 alkylene)C(0)NR a R b , wherein said alkylene is independently optionally substituted by halogen, oxo, -(C 0-3 alkylene)CN, -(C 0-3 alkylene)OR c , -(Co-3 alkylene)NR c R d , -(C 0 . 3 alkylene)C(0)R c , -(C 0 . 3 alkylene)C(0)OR c , -(C 0 . 3 alkylene)C(0)NR c R d , -(C 0 . 3 alkylene)NR c C(0)R d , -(C 0 .
  • R 5 is -(C 0-3 alkylene)C(0)NR a R b , wherein said alkylene is optionally substituted by halogen, oxo, -CN, -OR c , -NR c R d , -C(0)OR c , or Ci_ 6 alkyl optionally substituted by oxo, -CN or halogen.
  • R 5 is selected from -CH 2 C(0)NH 2, -CH 2 C(0)NHcyclopentyl -CH 2 C(0)N(CH 3 )(cyclopentyl), -CH 2 C(0)NHCH 3 , -CH(CH 3 )C(0)NHCH(CH 3 ) 2 ,
  • R 5 is -(C 0-3 alkylene)C(0)NR a R b , wherein said alkylene is independently optionally substituted by halogen, oxo, -(C 0-3 alkylene)CN, -(C 0-3 alkylene)OR c , -(Co- 3 alkylene)NR c R d , -(C 0 . 3 alkylene)C(0)R c , -(C 0 . 3 alkylene)C(0)OR c , -(C 0 . 3 alkylene)C(0)NR c R d , -(C 0 . 3 alkylene)NR c C(0)R d , -(C 0 .
  • R 5 is -(C 0-3 alkylene)C(0)NR a R b , wherein said alkylene is optionally substituted by oxo or halogen; and R a and R b are independently hydrogen, Ci_ 6 alkyl, C 3 _ 6 cycloalkyl, 4-6 membered heterocyclyl, wherein said alkyl, cycloalkyl and heterocyclyl are independently optionally substituted by halogen, oxo, -CN, -OR e or -NR e R f , or taken together with the atom to which they are attached to form a 3-6 membered heterocyclyl optionally substituted by oxo, halogen, -C(0)Ci_ 6 alkyl or Ci_6 alkyl.
  • R 5 is -CH 2 C(0)NR a R b , -CH 2 C(0)NHR a
  • R a and R b are independently hydrogen, Ci_ 6 alkyl, C3-6 cycloalkyl, 4-6 membered heterocyclyl, wherein said alkyl, cycloalkyl and heterocyclyl are independently optionally substituted by halogen, oxo, -CN, -OR e or -NR e R f , or taken together with the atom to which they are attached to form a 3-6 membered heterocyclyl optionally substituted by oxo, halogen, -C(0)Ci_ 6 alkyl or Ci_ 6 alkyl.
  • R 5 is -CH 2 C(0)NHCH 3 , -CH 2 C(0)N(CH 3 )(cyclopentyl), -CH 2 C(0)NH(cyclopentyl), -CH 2 C(0)NH(isopropyl), -CH 2 C(0)(pyrrolidin- 1 -yl),
  • R 5 is -(C 0-3 alkylene)NR a C(0)R b , wherein said alkylene is independently optionally substituted by halogen, oxo, -(C 0-3 alkylene)CN, -(C 0-3 alkylene)OR c , -(Co-3 alkylene)NR c R d , -(C 0 . 3 alkylene)C(0)R c , -(C 0 .
  • R 5 is -(C 0-3 alkylene)NR a C(0)R b , wherein said alkylene is optionally substituted by halogen, oxo, -CN, -OR c , -NR c R d , -C(0)OR c , or Ci_ 6 alkyl optionally substituted by oxo, -CN or halogen.
  • R 5 is selected from -CH 2 NHC(0)CH 3 , -CH 2 NHC(0)CH(CH 3 ) 2 , - CH 2 NHC(0)CH 2 CH 3 , -CH 2 NHC(0)CH 2 OCH 3 , -CH 2 NHC(0)pyridin-3-yl,
  • R 5 is -(C 0- 3 alkylene)NR a S(0)i_2R b , wherein said alkylene is independently optionally substituted by halogen, oxo, -(C 0- 3 alkylene)CN, -(C 0- 3 alkylene)OR c ,
  • R 5 is -(C 0-3 alkylene)NR a S(0)i_ 2 R b , wherein said alkylene is optionally substituted by halogen, oxo, -CN, -OR c , -NR c R d , -C(0)OR c , or Ci_ 6 alkyl optionally substituted by oxo, -CN or halogen.
  • R 5 is selected from -CH 2 NHS(0) 2 CH 3 , -CH 2 NHS(0) 2 CH 2 CH 3 , - CH 2 NHS(0) 2 CH 2 CH(CH 3 ) 2 , -CH 2 NHS(0) 2 CH(CH 3 ) 2 , -CH 2 NHS(0) 2 CH(CH 3 )CH 2 CH3, - CH 2 NHS(0) 2 cyclopropyl, -CH 2 NHS(0) 2 cyclopentyl, -CH 2 N(CH 3 ) 2 S(0) 2 CH 3 ,
  • R 5 is -(C 0-3 alkylene)5-12 membered heteroaryl, wherein said alkylene and heteroaryl are independently optionally substituted by halogen, oxo, -(C 0-3 alkylene)CN, -(C 0 . 3 alkylene)OR c , -(C 0 . 3 alkylene)NR c R d , -(C 0 . 3 alkylene)C(0)R c , -(C 0 . 3 alkylene)C(0)OR c , -(C 0 . 3 alkylene)C(0)NR c R d , -(C 0 .
  • R 5 is -(C 0-3 alkylene)5-6 membered heteroaryl, wherein said alkylene and heteroaryl are independently optionally substituted by halogen, oxo, -CN, -OR c , -NR c R d , -C(0)OR c , or Ci_6 alkyl optionally substituted by oxo, -CN or halogen.
  • R 5 is selected from -CH 2 CH 2 triazolyl, triazolyl, pyridinyl, -CH 2 pyrazolyl, -CH 2 pyridinyl, , wherein the wavy line represents the point of attachment in formula I.
  • R 5 is -(C 0 -3 alkylene)4-6 membered heteroaryl, wherein said alkylene is independently optionally substituted by halogen, oxo, -(C 0 -3 alkylene)CN, -(C 0 -3 alkylene)OR c , -(C 0 . 3 alkylene)NR c R d , -(C 0 . 3 alkylene)C(0)R c , -(C 0 . 3 alkylene)C(0)OR c , -(C 0 . 3 alkylene)C(0)NR c R d , -(C 0 .
  • R 5 is -(C 0-3 alkylene)4-6 membered heteroaryl, wherein said alkylene is optionally substituted by oxo or halogen, and said heteroaryl is optionally substituted by oxo, halogen, Ci_ 3 alkyl, -OR c or -NR c R d .
  • R 5 is pyridinyl.
  • R 5 is -(C 0-3 alkylene)3-12 membered heterocyclyl, wherein said alkylene and heterocyclyl are independently optionally substituted by halogen, oxo, -(C 0-3 alkylene)CN, -(C 0 . 3 alkylene)OR c , -(C 0 . 3 alkylene)NR c R d , -(C 0 . 3 alkylene)C(0)R c , -(C 0 . 3 alkylene)C(0)OR c , -(C 0 . 3 alkylene)C(0)NR c R d , -(C 0 .
  • R 5 is -(C 0-3 alkylene)3-7 membered heterocyclyl, wherein said alkylene and heterocyclyl are independently optionally substituted by halogen, oxo, -CN, -OR c , -NR c R d , -C(0)OR c , or Ci_ 6 alkyl optionally substituted by oxo, -CN or halogen.
  • R 5 is selected from oxetanyl, 1,1-dioxothiomorpholinyl, -CH 2 CH 2 (l,l-dioxothiomorpholinyl), - CH 2 CH 2 triazolyl, triazolyl, -CH 2 pyrazolyl, -CH 2 pyridinyl, pyridinyl, pyrrolidinyl, piperidinyl, -CH 2 (4-hydroxypiperidin-l-yl), morpholinyl, azetidinyl, 2-acetylpyrrolidin-3-yl, -CH 2 tetrahydropyranyl, -CH 2 tetrahydropyran-4-yl, tetrahydropyranyl, tetrahydrofuranyl, -CH 2 tetrahydrofuran-2-yl, -CH 2 CH 2 tetrahydrofuranyl, -CH 2 morpholinyl, l-
  • R 5 is -(C 0- 3 alkylene)4-6 membered heterocyclyl, wherein said alkylene is independently optionally substituted by halogen, oxo, -(C 0- 3 alkylene)CN, -(C 0- 3 alkylene)OR c , -(C 0 . 3 alkylene)NR c R d , -(C 0 . 3 alkylene)C(0)R c , -(C 0 . 3 alkylene)C(0)OR c , -(C 0 . 3 alkylene)C(0)NR c R d , -(C 0 .
  • R 5 is -(C 0-3 alkylene)4-6 membered heterocyclyl, wherein said alkylene is optionally substituted by oxo or halogen, and said heterocyclyl is optionally substituted by oxo, halogen, Ci_ 3 alkyl, -OR c or -NR c R d .
  • R 5 is -CH 2 C(0)(4-6 membered heterocyclyl) or -CH 2 (4-6 membered heterocyclyl), wherein said heterocyclyl is optionally substituted by oxo, halogen, Ci_ 3 alkyl, -OR c or -NR c R d .
  • said heterocyclyl is oxetanyl, pyridinyl, pyrrolindinyl, pyranyl, piperidinyl, morpholinyl or
  • R 5 is pyridin-3-yl, pyrrolidin-l-yl, pyran-4-yl, -CH 2 C(0)(pyrrolidin-l-yl), -CH 2 C(0)(4,4-difiuorpiperidin-l-yl), -CH 2 (morpholinyl), -CH 2 C(0)(morpholinyl), -CH 2 (pyrrolidin-2-on-l-yl) or , wherein the wavy line represents the point of attachment in formula I.
  • R 5 is -(C0-3 alkylene)S(0)i_2R a , wherein said alkylene is independently optionally substituted by halogen, oxo, -(C 0 -3 alkylene)CN, -(C 0 -3 alkylene)OR c , -(Co-3 alkylene)NR c R d , -(C 0 . 3 alkylene)C(0)R c , -(C 0 . 3 alkylene)C(0)OR c , -(C 0 . 3 alkylene)C(0)NR c R d , -(C 0 . 3 alkylene)NR c C(0)R d , -(C 0 .
  • R 5 is -(C 0-3 alkylene) S(0)i_ 2 R a , wherein said alkylene is optionally substituted by halogen, oxo, -CN, -OR c , -NR c R d , -C(0)OR c , or Ci_ 6 alkyl optionally substituted by oxo, -CN or halogen.
  • R 5 is selected from -CH 2 S(0) 2 CH 3 .
  • R 5 is -(C 0-3 alkylene)C6_i 2 aryl, wherein said alkylene and aryl are independently optionally substituted by halogen, oxo, -(C 0-3 alkylene)CN, -(C 0-3 alkylene)OR c , -(Co-3 alkylene)NR c R d , -(C 0 . 3 alkylene)C(0)R c , -(C 0 . 3 alkylene)C(0)OR c , -(C 0 . 3 alkylene)C(0)NR c R d , -(C 0 .
  • R 5 is -(C 0-3 alkylene)phenyl, wherein said alkylene and phenyl are independently optionally substituted by halogen, oxo, -CN, -OR c , -NR c R d , -C(0)OR c , or Ci_ 6 alkyl optionally substitu tteedd 1 by oxo, -CN cted from -CH 2 phenyl, wherein the wavy line represents the point of attachment in formula I.
  • R 5 is -(C 0-3 alkylene)phenyl, wherein said alkylene is optionally substituted by oxo or halogen, and said phenyl is optionally substituted by halogen, Ci_3 alkyl, -OR c or -NR c R d .
  • R 5 is -(C0-3 alkylene)NR a C(0)OR b , wherein said alkylene is independently optionally substituted by halogen, oxo, -(C0-3 alkylene)CN, -(C0-3 alkylene)OR c ,
  • R 5 is -(C 0-3 alkylene)NR a C(0)OR b , wherein said alkylene is optionally substituted by halogen, oxo, -CN, -OR c , -NR c R d , -C(0)OR c , or Ci_ 6 alkyl optionally substituted by oxo, -CN or halogen.
  • R 5 is selected from -CH 2 NHC(0)OCH 2 CH 3 and -CH 2 NHC(0)OCH 3 .
  • R 5 is selected from hydrogen, fluro, methyl, ethyl, 1 -hydro xyethyl,
  • -CH 2 CH 2 (l,l-dioxothiomorpholinyl), -CH 2 CH 2 triazolyl, triazolyl, -CH 2 pyrazolyl, - CH 2 pyridinyl, pyridinyl, pyrrolidinyl, piperidinyl, morpholinyl, azetidinyl, 2-acetylpyrrolidin-3- yl, -CH 2 tetrahydropyranyl, tetrahydropyranyl, tetrahydrofuranyl, -CH 2 CH 2 tetrahydrofuranyl, -CH 2 morpholinyl, l-acetylpiperidin-4-yl, -C(0)morpholi -CH 2 C(0)morpholinyl,
  • Y is CR 5 and R 5 is methyl, ethyl, propyl, isopropyl, cyclopropyl, eye lo butyl, cyano, 2-methylbutyl, N-(2-hydroxyethyl)amino, N-(2-methoxyethyl)amino, methylsulfonylamino methyl, 2-(methylsulfonylamino)ethyl, cyclopropylmethyl, 2-[N-(2- propylsulfonyl)amino]ethyl, 2-[N-(cyclopropylsulfonyl)-amino]ethyl, 2- (cyclopropylcarbonylamino)ethyl, 2-(acetylamino)ethyl, 2-(methoxymethyl- carbonylamin
  • R 5 is methyl, 1 -hydro xyethyl, hydro xymethyl,
  • R 5 is (R)-l -hydro xyethyl.
  • R 5 is (S)-l -hydro xyethyl.
  • R 5 is C 3-12 alkyl, wherein said C 3-12 alkyl, is independently optionally substituted by halogen, oxo, -(C 0-3 alkylene)CN, -(C 0-3 alkylene)OR c , -(C 0-3 alkylene)NR c R d , -(Co-3 alkylene)C(0)R c , -(C 0 . 3 alkylene)C(0)OR c , -(C 0 . 3 alkylene)C(0)NR c R d , -(C 0 .
  • R 5 is C 1-12 alkyl, wherein said alkyl, alkenyl, alkynyl, alkylene, cycloalkyl, aryl and heterocyclyl are independently optionally substituted by halogen, oxo, -(C 0- 3 alkylene)CN, -(C 2 _ 3 alkylene)OR c , -(C 0 . 3 alkylene)NR c R d , -(C 0 . 3 alkylene)C(0)R c , -(C 0 . 3 alkylene)C(0)OR c , -(C 0 . 3 alkylene)C(0)NR c R d , -(C 0 .
  • R 5 is C 1-12 alkyl, wherein said alkyl, alkenyl, alkynyl, alkylene, cycloalkyl, aryl and heterocyclyl are independently optionally substituted by halogen, oxo, -(Co-3 alkylene)CN, -(C 0 . 3 alkylene)NR c R d , -(C 0 . 3 alkylene)C(0)R c , -(C 0 . 3 alkylene)C(0)OR c , -(C 0 . 3 alkylene)C(0)NR c R d , -(C 0 .
  • R 5 is halogen, C 1-12 alkyl, C 2 _i 2 alkenyl, C 2 _i 2 alkynyl, -(C 0-3 alkylene)CN, -(C 0 . 3 alkylene)NR a R b , -(C 0 . 3 alkylene)OR a , -(C 0 . 3 alkylene)SR a , -(C 0 . 3 alkylene)C(0)R a , -(C 0 . 3 alkylene)NR a C(0)R b , -(C 0 . 3 alkylene)C(0)NR a R b , -(C 0 . 3 alkylene)C(0)NR a R b , -(C 0 .
  • alkylene)NR a S(0)i_ 2 NR a R b -(C 0 . 3 alkylene)C3_i 2 cycloalkyl, -(C 0-3 alkylene)C6_i4 aryl, -(C 0-3 alkylene)3-12 membered heterocyclyl or -(C 0-3 alkylene)C(0)3-12 membered heterocyclyl, wherein said alkyl, alkenyl, alkynyl, alkylene, cycloalkyl, aryl and heterocyclyl are independently optionally substituted by halogen, oxo, -(C 0 . 3 alkylene)CN, -(C 0 .
  • each R k is independently hydrogen, C 3 -6 alkyl, C 2 _ 6 alkenyl, C 2 _ 6 alkynyl, -(C 0-3 alkylene)C 3 - 6 cycloalkyl, -(C 0-3 alkylene)3-12 membered heterocyclyl, -(C 0-3 alkylene)C(0)3-12 membered heterocyclyl or -(C 0-3 alkylene)C 6 _i4 aryl, wherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl and aryl are independently optionally substituted by halogen, oxo, -CN, -OR g , -NR g R h , -C(0)R g , -C(0)
  • R 5 is halogen, C 1-12 alkyl, C 2-12 alkenyl, C 2-12 alkynyl, -(C 0 -3 alkylene)CN, -(C 0 . 3 alkylene)NR a R b , -(C 0 . 3 alkylene)OR a , -(C 0 . 3 alkylene)SR a , -(C 0 . 3 alkylene)C(0)R a , -(C 0 . 3 alkylene)NR a C(0)R b , -(C 0 . 3 alkylene)C(0)NR a R b , -(C 0 . 3 alkylene)C(0)NR a R b , -(C 0 .
  • alkylene)NR a S(0)i_ 2 NR a R b -(C 0 . 3 alkylene)C3_i 2 cycloalkyl, -(C 0-3 alkylene)C6_i4 aryl, -(C 0-3 alkylene)3-12 membered heterocyclyl or -(C 0-3 alkylene)C(0)3-12 membered heterocyclyl, wherein said alkyl, alkenyl, alkynyl, alkylene, cycloalkyl, aryl and heterocyclyl are independently optionally substituted by halogen, oxo, -(C 0 . 3 alkylene)CN, -(C 0 .
  • each R k is independently hydrogen, C 2 _ 6 alkenyl, C 2 _ 6 alkynyl, -(Co-3 alkylene)C3_6 cycloalkyl, -(C 0-3 alkylene)3-12 membered heterocyclyl, -(C 0-3 alkylene)C(0)3-12 membered heterocyclyl or -(C 0-3 alkylene)C 6 _i4 aryl, wherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl and aryl are independently optionally substituted by halogen, oxo, -CN, -OR g , -NR g R h , -C(0)R g , -C(0)OR g , -C(0)NR
  • R 6 is independently oxo, halogen, -CN, -C(0)R a , -C(0)OR a , -NR a C(0)R b , -C(0)NR a R b , -NR a C(0)NR a R b , -OC(0)NR a R b , -NR a C(0)OR b , -S(0)i_ 2 R a , -NR a S(0) 2 R b , -S(0) 2 NR a R b , -OR a , -SR a , -NR a R b , Ci_ 6 alkyl, C 3 _ 6 cycloalkyl, C 2 _ 6 alkenyl, C 2 _ 6 alkynyl, 3-7 membered heterocycly or C 6-14 aryl, and wherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl and aryl, and where
  • R 6 is independently oxo, halogen, -CN, -C(0)(Ci_ 6 alkyl), -C(0)0(d_ 6 alkyl), -S(0) 2 (d_ 6 alkyl), -NR a S(0) 2 (d_ 6 alkyl), -0(d_ 6 alkyl), d_ 6 alkyl, C 3 _ 6 cycloalkyl or 3-7 membered heterocyclyl, wherein said alkyl, cycloalkyl and heterocyclyl are independently optionally substituted by halogen, oxo, -CN, -OR c , -NR c R d or Ci_ 6 alkyl optionally substituted by halogen.
  • R 6 is independently oxo, F, CI, -CN, -OH, -C(0)CH 3 , -CH 2 CN, -CH 2 CH 2 CN, cyclopropyl, cyclobutyl, -CF 3 , -NHS(0) 2 CH 3 , -S(0) 2 CH 3 , -C(0)OCH 3 , pyrrolidinyl or pyrrolidinonyl.
  • R 6 is independently oxo, halogen, -CN, -C(0)(Ci_ 6 alkyl), -S(0) 2 (Ci_6 alkyl), -OR a , -NR a R b , Ci_6 alkyl or C 3 _ 6 cycloalkyl, and wherein said alkyl, alkenyl and alkynyl are independently optionally substituted by halogen, oxo, -CN, -OR c or -NR c R d .
  • R 6 is halogen, -S(0) 2 CH 3 or -CN.
  • R 3 is optionally substituted by 1 to 3 R 6 independently selected from oxo, halogen, -CN, -S(0) 2 (Ci_6 alkyl), -OR a , -NR a R b and Ci_6 alkyl, and wherein said alkyl, alkenyl and alkynyl are independently optionally substituted by halogen, oxo, -CN, -OR c or -NR c R d .
  • R 3 is optionally substituted by 1 to 3 R 6 independently selected from oxo, halogen, -CN, -C(0)(Ci_ 6 alkyl), -C(0)0(Ci_6 alkyl), -S(0) 2 (C ⁇ alkyl), -NR a S(0) 2 (Ci_6 alkyl), -0(Ci_ 6 alkyl), Ci_ 6 alkyl, C 3 _ 6 cycloalkyl or 3-7 membered heterocyclyl, wherein said alkyl, cycloalkyl and heterocyclyl are independently optionally substituted by halogen, oxo, -CN, -OR c , -NR c R d or Ci_ 6 alkyl optionally substituted by halogen.
  • each R a and R b are independently hydrogen, Ci_ 6 alkyl, C 2 _ 6 alkenyl, C 2 _ 6 alkynyl, -C 3 _ 6 cycloalkyl, -3-12 membered heterocyclyl, -C(0)3-12 membered heterocyclyl or -C 6 -i4 aryl, wherein said alkyl, cycloalkyl, heterocyclyl and aryl are independently optionally substituted by halogen, oxo, -CN, -OR e , -NR e R f , -C(0)R g , -C(0)OR g , -C(0)NR g R h , -NR g C(0)R h , -OC(0)NR g R h , -NR g C(0)NR g R h , -NR g C(0)OR h , -S(0)i_ 2 R g , -NR g S(0)
  • each R a and R b are independently hydrogen, Ci_ 6 alkyl, C3-6 cycloalkyl, 3-6 membered heterocyclyl, -C(0)3-6 membered heterocyclyl or phenyl, wherein said alkyl, cycloalkyl, heterocyclyl and phenyl are independently optionally substituted by halogen, oxo, -CN, -OR e , -NR e R f , -C(0)R g , -C(0)OR g , -C(0)NR g R h , -NR g C(0)R h , -OC(0)NR g R h , -NR g C(0)NR g R h , -NR g C(0)OR h , -S(0)i_ 2 R g , -NR g S(0)i_ 2 R h , -S(0)i_ 2 NR g R h , -NR g S(0)i_ 2 NR h
  • each R a and R b are independently hydrogen, Ci_ 6 alkyl, C 3 _ 6 cycloalkyl, 3-6 membered heterocyclyl, 5-6 membered heteroaryl or phenyl, wherein said alkyl, cycloalkyl, heterocyclyl, heteroaryl and phenyl are independently optionally substituted by halogen, oxo, -CN, -OR e , -NR e R f or Ci_ 3 alkyl optionally substituted by halogen.
  • each R a and R b are independently selected from hydrogen, methyl, ethyl, propyl, isopropyl, butyl, t-butyl, sec-butyl, -CF 3 , -CH 2 CF 3 , -CH 2 F, -CHF 2 , -CH 2 OH, -CH 2 CH 2 OH, -CH 2 NH 2 , -CH 2 CH 2 NH 2 , -CH 2 CH 2 N(CH 3 ) 2 , -CH 2 N(CH 3 ) 2 , cyclopropyl, 2,2- difluorocyclopropyl, 2-fluorocyclopropyl, 2-methylcyclopropyl, eye lo butyl, cyclopentyl, cyclohexyl, piperidinyl, morpholinyl, piperazinyl, N-methylpiperazinyl, pyrazolyl, N- methylpyrazolyl, azetidinyl,
  • a R a and a R b are independently taken together with the atom to which they are attached to form a 3-6 membered heterocyclyl optionally substituted by oxo, halogen, -C(0)Ci_ 6 alkyl or Ci_ 6 alkyl optionally substituted by oxo, halogen, OR g or NR g NR h .
  • a R a and a R b are independently taken together with the atom to which they are attached to form a 3-6 membered heterocyclyl optionally substituted by oxo, halogen, -C(0)Ci_ 6 alkyl or Ci_ 6 alkyl optionally substituted by halogen.
  • said heterocyclyl is azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, piperidinonyl, morpholinyl and 1,1-dioxomorpholinyl.
  • R a and R b are taken together with the atom to which they are attached to form a 4-6 membered heterocyclyl selected from azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl and morpholinyl, optionally substituted by oxo, halogen, -C(0)Ci_ 6 alkyl or Ci_ 6 alkyl.
  • R a and R b are independently hydrogen, methyl, isopropyl, cyclopropyl or cyclopentyl.
  • R a and R b are taken together with the atom to which they are attached to form a 4-6 membered heterocyclyl selected from azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl and morpholinyl, optionally substituted by oxo, halogen, -C(0)Ci_ 6 alkyl or Ci_ 6 alkyl.
  • each R c and R d are independently hydrogen, Ci_ 6 alkyl, C 2 _ 6 alkenyl, C 2 _ 6 alkynyl, -C3-6 cycloalkyl, -3-12 membered heterocyclyl, -C(0)3-12 membered heterocyclyl or -C 6-14 aryl, wherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl and aryl are independently optionally substituted by halogen, oxo, -CN, -OR g , -NR g R h , -C(0)R g , -C(0)OR g , -C(0)NR g R h , -NR g C(0)R h , -OC(0)NR g R h , -NR g C(0)NR g R h , -NR g C(0)OR h , -S(0)i_ 2 R g , -
  • each R c and R d are independently hydrogen, Ci_ 6 alkyl, C 2 _ 6 alkenyl, C 2 _ 6 alkynyl, -C 3 _ 6 cycloalkyl, -3-6 membered heterocyclyl, -C(0)3-6 membered heterocyclyl or phenyll, wherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl and phenyl are independently optionally substituted by halogen, oxo, -CN, -OR g , -NR g R h , -C(0)R g , -C(0)OR g , -C(0)NR g R h , -NR g C(0)R h , -OC(0)NR g R h , -NR g C(0)NR g R h , -NR g C(0)OR h , -S(0)i_ 2 R g , -NR
  • each R c and R d are independently hydrogen, Ci_ 6 alkyl, C 3 _ 6 cycloalkyl, 3-6 membered heterocyclyl, 5-6 membered heteroaryl or phenyl, wherein said alkyl, cycloalkyl, heterocyclyl, heteroaryl and phenyl are independently optionally substituted by halogen, oxo, -CN, -OR g , -NR g R h or Ci_ 6 alkyl optionally substituted by halogen.
  • each R c and R d are independently hydrogen, methyl, ethyl, isopropyl, butyl, t-butyl, sec-butyl, -CF 3 , "CH 2 CF 3 , -CH 2 F, -CHF 2 , -CH 2 OH, -CH 2 CH 2 OH, -CH 2 NH 2 , -CH 2 CH 2 NH 2 , -CH 2 CH 2 N(CH 3 ) 2 , -CH 2 N(CH 3 ) 2 , cyclopropyl, 2,2- difluorocyclopropyl, 2-fluorocyclopropyl, 2-methylcyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, piperidinyl, morpholinyl, piperazinyl, N-methylpiperazinyl, pyrazolyl, N- methylpyrazolyl, azetidinyl, 1,1-dio
  • a R c and a R d are independently taken together with the atom to which they are attached to form a 3-6 membered heterocyclyl optionally substituted by oxo, halogen, -C(0)Ci_ 6 alkyl or Ci_ 6 alkyl optionally substituted by oxo or halogen.
  • each R c and R d are independently hydrogen, methyl or ethyl, optionally substituted by fluoro or oxo. In one embodiment, each R c and R d are independently hydrogen, methyl or ethyl. In one embodiment, a R c and a R d are taken together with the atom to which they are attached to form a 3-6 membered heterocyclyl optionally substituted by oxo, halogen, -C(0)Ci_ 6 alkyl or Ci_ 6 alkyl optionally substituted by halogen.
  • said heterocyclyl is azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, piperidinonyl, morpholinyl and 1,1- dioxomorpholinyl.
  • R c , R d , R e , R f , R g and R h are independently hydrogen or methyl.
  • each R e , R f , R g , R h are independently hydrogen, methyl, ethyl, propyl or isopropyl, optionally substituted by halogen or oxo. In one embodiment, each R e , R f , R g , R h are independently hydrogen, methyl or ethyl.
  • X is CR 4 ;
  • Y is N or CR 5 ;
  • R 1 is azetidinyl, piperidinyl, pyrrolidinyl or cyclohexyl, optionally substituted by Ci_ 3 alkylene or Ci_ 3 alkyl;
  • R 2 is absent, Ci_ 3 alkyl, - H-, -NHCH 2 - -CH 2 0- -(CH 2 ) 2 0-, -C(0)NH- -CH 2 C(0)NH-, -CH 2 C(0)N(CH 3 )-, -NHC(0)CH 2 -, - HC(0)0-, -C(0)0-, -C(0)CH 2 S(0) 2 , -NHS(0) 2 -, -NHS(0) 2 CH 2 -, -CH 2 C(0)-, -(CH 2 ) 2 C(0)-, -S(0) 2 - -CH 2 S(0) 2 -, -S(0) 2 (CH 2 ) 2 -;
  • R 3 is absent, Ci_6 alkyl optionally substituted by oxo, halogen, -CN, -S(0) 2 (Ci_6 alkyl), -OR a or -NR a R b ;
  • R 4 is hydrogen, F or methyl
  • R 5 is Ci_6 alkyl or C 3 _ 6 cycloalkyl optionally substituted by halogen, oxo, -CN, -OR a or -NR a R b ,
  • each R a and R b are independently hydrogen, Ci_ 3 alkyl or C 3 _ 6 cycloalkyl, wherein said alkyl and cycloalkyl are independently optionally substituted by oxo or halogen; or are taken together with the atom to which they are attached to form a 3-6 membered heterocyclyl optionally substituted by oxo, halogen or Ci_ 3 alkyl; and each R c and R d are independently hydrogen or Ci_ 6 alkyl; or are taken together with the atom to which they are attached to form a 3-6 membered heterocyclyl optionally substituted by oxo, halogen or Ci_ 3 alkyl; and each R c and R d are independently hydrogen or Ci_ 6 alkyl; or are taken together with the atom to which they are attached to form a 3-6 membered heterocyclyl optionally substituted by oxo, halogen or Ci_ 3 alkyl; and each R c and R d are independently hydrogen or
  • X is CR 4 ; Y is N or CR 5 ; R 1 is azetidinyl, piperidinyl, pyrrolidinyl, tetrahydropyranyl, cyclopentyl or cyclohexyl, optionally substituted by Ci_ 3 alkylene, -CN, -OR a or Ci_ 3 alkyl; R 2 is absent, Ci_ 3 alkyl, - H-, -NHCH 2 -, -CH 2 0-, -(CH 2 ) 2 0- -C(0)NH-, -CH 2 C(0)NH-, -CH 2 C(0)N(CH 3 )-, -NHC(0)CH 2 -, - HC(0)0-, -C(0)0-, -C(0)CH 2 S(0) 2 , -NHS(0) 2 -, -NHS(0) 2 CH 2 -, -CH 2 C(0)-, -(CH 2 ) 2 C(0)-, -S(0) 2 -
  • R 3 is absent, hydrogen
  • Ci_6 alkyl optionally substituted by oxo, halogen, -CN, -S(0) 2 (Ci_ 6 alkyl), -OR a or -NR a R b ;
  • R 4 is hydrogen, F or methyl
  • R 5 is C 1-12 alkyl or C 3 _i 2 cycloalkyl optionally substituted by halogen, oxo, -CN, -OR a or -NR a R b ,
  • R 1 is azetidinyl, piperidinyl, pyrrolidinyl or cyclohexyl, optionally substituted by Ci_ 3 alkylene or Ci_ 3 alkyl; R 2 is absent; and R 3 is hydrogen.
  • R 1 is azetidinyl, piperidinyl, pyrrolidinyl or cyclohexyl, optionally substituted by Ci_ 3 alkylene or Ci_ 3 alkyl; R 2 is absent; R 3 is hydrogen; and R 5 is -CN or Ci_ 3 alkyl optionally substituted by halogen or oxo.
  • X is CH; Y is CR 5 ; R 1 is piperidinyl, tetrahydropyranyl, cyclopentyl or cyclohexyl, wherein R 1 is optionally substituted by Ci_ 3 alkylene, halogen, -OR a , -CN, -NR a R b or Ci_ 6 alkyl optionally substituted by oxo, -OR a , -CN, -NR a R b or halogen; R 2 is absent; R 3 is absent; R 5 is C 1-12 alkyl optionally substituted by halogen, oxo, -CN, -OH, -OCH 3 , -NH 2 or -N(CH 3 ) 2 ; and each R a and R b are independently selected from Ci_ 3 alkyl optionally substituted by oxo or halogen.
  • X is CH; Y is CR 5 ; R 1 is piperidinyl, tetrahydropyranyl or cyclohexyl, wherein R 1 is optionally substituted by Ci_ 3 alkylene, halogen, -OH, -NH 2 or Ci_ 3 alkyl optionally substituted by oxo, -CN or halogen; R 2 is absent; R 3 is absent; and R 5 is Ci_ 6 alkyl optionally substituted by halogen, oxo, -CN, -OH, -OCH 3 , -NH 2 or -N(CH 3 ) 2 .
  • X is CH; Y is CR 5 ; R 1 is piperidinyl, tetrahydrofuranyl, tetrahydropyranyl, cyclopentyl or cyclohexyl, wherein R 1 is optionally substituted by Ci_ 3 alkylene, halogen, -OH, -NH 2 or Ci_ 3 alkyl optionally substituted by oxo, -CN or halogen; R 2 is
  • R 5 is selected from
  • R 1 is piperidinyl optionally substituted by Ci_ 3 alkylene or Ci_ 3 alkyl;
  • R 2 is Ci_ 3 alkyl optionally substituted by oxo;
  • R 3 is Ci_ 6 alkyl optionally substituted by oxo, halogen or -CN, phenyl or pyridinyl, wherein said phenyl and pyridinyl are independently optionally substituted by halogen or -CN.
  • Another embodiment includes a compound selected from Examples 1-56.
  • the compound of formula I is not:
  • -R'-R ⁇ R 3 taken together are not morpholino. In one embodiment -R'-R ⁇ R 3 taken together are not morpholino when R 5 is ethoxymethyl.
  • R 5 is not ethoxymethyl.
  • Compounds of the invention may contain one or more asymmetric carbon atoms. Accordingly, the compounds may exist as diastereomers, enantiomers or mixtures thereof.
  • the syntheses of the compounds may employ racemates, diastereomers or enantiomers as starting materials or as intermediates. Mixtures of particular diastereomeric compounds may be separated, or enriched in one or more particular diastereomers, by chromatographic or crystallization methods. Similarly, enantiomeric mixtures may be separated, or enantiomerically enriched, using the same techniques or others known in the art.
  • Each of the asymmetric carbon or nitrogen atoms may be in the R or S configuration and both of these configurations are within the scope of the invention.
  • prodrugs of the compounds of formula I including known amino -protecting and carboxy-protecting groups which are released, for example hydrolyzed, to yield the compound of formula I under physiologic conditions.
  • a particular class of prodrugs are compounds in which a nitrogen atom in an amino, amidino, amino alky leneamino, iminoalkyleneamino or guanidino group is substituted with a hydroxy (OH) group, an alkylcarbonyl (-CO-R) group, an alkoxycarbonyl (-CO-OR), an acyloxyalkyl-alkoxycarbonyl (- CO-O-R-O-CO-R) group where R is a monovalent or divalent group, for example alkyl, alkylene or aryl, or a group having the formula -C(0)-0-CPlP2-haloalkyl, where PI and P2 are the same or different and are hydrogen, alkyl, alkoxy, cyano, halogen, alkyl or
  • the nitrogen atom is one of the nitrogen atoms of the amidino group of the compounds of formula I.
  • Prodrugs may be prepared by reacting a compound of formula I with an activated group, such as acyl groups, to bond, for example, a nitrogen atom in the compound of formula I to the exemplary carbonyl of the activated acyl group.
  • activated carbonyl compounds are those containing a leaving group bonded to the carbonyl group, and include, for example, acyl halides, acyl amines, acyl pyridinium salts, acyl alkoxides, acyl phenoxides such as p-nitrophenoxy acyl, dinitrophenoxy acyl, fluorophenoxy acyl, and difluorophenoxy acyl.
  • the reactions are generally carried out in inert solvents at reduced temperatures such as -78 to about 50°C.
  • the reactions may also be carried out in the presence of an inorganic base, for example potassium carbonate or sodium bicarbonate, or an organic base such as an amine, including pyridine, trimethylamine, triethylamine, triethanolamine, or the like.
  • an inorganic base for example potassium carbonate or sodium bicarbonate
  • an organic base such as an amine, including pyridine, trimethylamine, triethylamine, triethanolamine, or the like.
  • Compounds of formula I may be synthesized by synthetic routes described herein.
  • processes well-known in the chemical arts can be used, in addition to, or in light of, the description contained herein.
  • the starting materials are generally available from commercial sources such as Aldrich Chemicals (Milwaukee, Wis.) or are readily prepared using methods well known to those skilled in the art (e.g., prepared by methods generally described in Louis F. Fieser and Mary Fieser, Reagents for Organic Synthesis, v. 1-19, Wiley, N.Y. (1967- 1999 ed.), Beilsteins Handbuch der organischen Chemie, 4, Aufl. ed. Springer- Verlag, Berlin, including supplements (also available via the Beilstein online database)), or Comprehensive Heterocyclic Chemistry, Editors Katrizky and Rees, Pergamon Press, 1984.
  • Compounds of formula I may be prepared singly or as compound libraries comprising at least 2, for example 5 to 1,000 compounds, or 10 to 100 compounds of formula I.
  • Libraries of compounds of formula I may be prepared by a combinatorial "split and mix” approach or by multiple parallel syntheses using either solution phase or solid phase chemistry, by procedures known to those skilled in the art.
  • a compound library comprising at least 2 compounds of formula I, enantiomers, diastereomers, tautomers or pharmaceutically acceptable salts thereof.
  • reaction schemes 1-10 depicted below provide routes for synthesizing the compounds of the present invention as well as key intermediates. For a more detailed description of the individual reaction steps, see the Examples section below. Those skilled in the art will appreciate that other synthetic routes may be used to synthesize the inventive compounds. Although specific starting materials and reagents are depicted in the Schemes and discussed below, other starting materials and reagents can be easily substituted to provide a variety of derivatives and/or reaction conditions. In addition, many of the compounds prepared by the methods described below can be further modified in light of this disclosure using conventional chemistry well known to those skilled in the art.
  • Suitable amino -protecting groups include acetyl, trifluoro acetyl, benzyl, phenylsulfonyl, t-butoxycarbonyl (BOC), benzyloxycarbonyl (CBz) and 9-fluorenylmethyleneoxycarbonyl (Fmoc).
  • Suitable amino -protecting groups include acetyl, trifluoro acetyl, benzyl, phenylsulfonyl, t-butoxycarbonyl (BOC), benzyloxycarbonyl (CBz) and 9-fluorenylmethyleneoxycarbonyl (Fmoc).
  • n 0 or 1
  • reaction products from one another and/or from starting materials.
  • the desired products of each step or series of steps is separated and/or purified (hereinafter separated) to the desired degree of homogeneity by the techniques common in the art.
  • separations involve multiphase extraction, crystallization or trituration from a solvent or solvent mixture, distillation, sublimation, or chromatography.
  • Chromatography can involve any number of methods including, for example: reverse-phase and normal phase; size exclusion; ion exchange; supercritical fluid; high, medium, and low pressure liquid chromatography methods and apparatus; small scale analytical; simulated moving bed (SMB) and preparative thin or thick layer chromatography, as well as techniques of small scale thin layer and flash chromatography.
  • SMB simulated moving bed
  • Another class of separation methods involves treatment of a mixture with a reagent selected to bind to or render otherwise separable a desired product, unreacted starting material, reaction by product, or the like.
  • reagents include adsorbents or absorbents such as activated carbon, molecular sieves, ion exchange media, or the like.
  • the reagents can be acids in the case of a basic material, bases in the case of an acidic material, binding reagents such as antibodies, binding proteins, selective chelators such as crown ethers, liquid/liquid ion extraction reagents (LIX), or the like. Selection of appropriate methods of separation depends on the nature of the materials involved.
  • Example separation methods include boiling point, and molecular weight in distillation and sublimation, presence or absence of polar functional groups in chromatography, stability of materials in acidic and basic media in multiphase extraction, and the like.
  • One skilled in the art will apply techniques most likely to achieve the desired separation.
  • Diastereomeric mixtures can be separated into their individual diastereo isomers on the basis of their physical chemical differences by methods well known to those skilled in the art, such as by chromatography and/or fractional crystallization.
  • Enantiomers can be separated by converting the enantiomeric mixture into a diastereomeric mixture by reaction with an appropriate optically active compound (e.g., chiral auxiliary such as a chiral alcohol or Mosher's acid chloride), separating the diastereoisomers and converting (e.g., hydrolyzing) the individual diastereoisomers to the corresponding pure enantiomers. Also, some of the compounds of the present invention may be atropisomers (e.g., substituted biaryls) and are considered as part of this invention. Enantiomers can also be separated by use of a chiral HPLC column or supercritical fluid chromatography.
  • an appropriate optically active compound e.g., chiral auxiliary such as a chiral alcohol or Mosher's acid chloride
  • converting e.g., hydrolyzing
  • some of the compounds of the present invention may be atropisomers (e.g., substituted biaryls) and are considered as
  • a single stereoisomer, e.g. an enantiomer, substantially free of its stereoisomer may be obtained by resolution of the racemic mixture using a method such as formation of diastereomers using optically active resolving agents (Eliel, E. and Wilen, S., Stereochemistry of Organic Compounds, John Wiley & Sons, Inc., New York, 1994; Lochmuller, C. H., J. Chromatogr., 113(3):283-302 (1975)).
  • Racemic mixtures of chiral compounds of the invention can be separated and isolated by any suitable method, including: (1) formation of ionic, diastereomeric salts with chiral compounds and separation by fractional crystallization or other methods, (2) formation of diastereomeric compounds with chiral derivatizing reagents, separation of the diastereomers, and conversion to the pure stereoisomers, and (3) separation of the substantially pure or enriched stereoisomers directly under chiral conditions.
  • suitable method including: (1) formation of ionic, diastereomeric salts with chiral compounds and separation by fractional crystallization or other methods, (2) formation of diastereomeric compounds with chiral derivatizing reagents, separation of the diastereomers, and conversion to the pure stereoisomers, and (3) separation of the substantially pure or enriched stereoisomers directly under chiral conditions.
  • Diastereomeric salts can be formed by reaction of enantiomerically pure chiral bases such as brucine, quinine, ephedrine, strychnine, a-methyl- -phenylethylamine (amphetamine), and the like with asymmetric compounds bearing acidic functionality, such as carboxylic acid and sulfonic acid.
  • the diastereomeric salts may be induced to separate by fractional crystallization or ionic chromatography.
  • addition of chiral carboxylic or sulfonic acids such as camphorsulfonic acid, tartaric acid, mandelic acid, or lactic acid can result in formation of the diastereomeric salts.
  • the substrate to be resolved is reacted with one enantiomer of a chiral compound to form a diastereomeric pair
  • Diastereomeric compounds can be formed by reacting asymmetric compounds with enantiomerically pure chiral derivatizing reagents, such as menthyl derivatives, followed by separation of the diastereomers and hydrolysis to yield the pure or enriched enantiomer.
  • a method of determining optical purity involves making chiral esters, such as a menthyl ester, e.g.
  • a racemic mixture of two enantiomers can be separated by chromatography using a chiral stationary phase ⁇ Chiral Liquid Chromatography W. J. Lough, Ed., Chapman and Hall, New York, (1989); Okamoto, J. of Chromatogr. 513:375-378 (1990)).
  • Enriched or purified enantiomers can be distinguished by methods used to distinguish other chiral molecules with asymmetric carbon atoms, such as optical rotation and circular dichroism.
  • the absolute stereochemistry of chiral centers and enatiomers can be determined by x-ray crystallography.
  • Positional isomers for example E and Z forms, of compounds of formula I, and intermediates for their synthesis, may be observed by characterization methods such as NMR and analytical HPLC.
  • the E and Z isomers may be separated, for example by preparatory HPLC.
  • compositions or medicaments containing the compounds of the invention and a therapeutically inert carrier, diluent or excipient, as well as methods of using the compounds of the invention to prepare such compositions and medicaments.
  • compounds of formula I may be formulated by mixing at ambient temperature at the appropriate pH, and at the desired degree of purity, with physiologically acceptable carriers, i.e., carriers that are non-toxic to recipients at the dosages and concentrations employed into a galenical administration form.
  • the pH of the formulation depends mainly on the particular use and the concentration of compound, but preferably ranges anywhere from about 3 to about 8.
  • a compound of formula I is formulated in an acetate buffer, at pH 5.
  • the compounds of formula I are sterile.
  • the compound may be stored, for example, as a solid or amorphous composition, as a lyophilized formulation or as an aqueous solution.
  • compositions are formulated, dosed, and administered in a fashion consistent with good medical practice.
  • Factors for consideration in this context include the particular disorder being treated, the particular mammal being treated, the clinical condition of the individual patient, the cause of the disorder, the site of delivery of the agent, the method of administration, the scheduling of administration, and other factors known to medical practitioners.
  • the therapeutically effective amount of the compound of the invention administered parenterally per dose will be in the range of about 0.01-100 mg/kg, alternatively about 0.1 to 20 mg/kg of patient body weight per day, with the typical initial range of compound used being 0.3 to 15 mg/kg/day.
  • oral unit dosage forms such as tablets and capsules, contain from about 5 to about 100 mg of the compound of the invention.
  • the compounds of the invention may be administered by any suitable means, including oral, topical (including buccal and sublingual), rectal, vaginal, transdermal, parenteral, subcutaneous, intraperitoneal, intrapulmonary, intradermal, intrathecal, inhaled and epidural and intranasal, and, if desired for local treatment, intralesional administration.
  • Parenteral infusions include intramuscular, intravenous, intraarterial, intraperitoneal, or subcutaneous administration.
  • the compounds of the present invention may be administered in any convenient administrative form, e.g., tablets, powders, capsules, solutions, dispersions, suspensions, syrups, sprays, vapors, suppositories, gels, emulsions, patches, etc.
  • Such compositions may contain components conventional in pharmaceutical preparations, e.g., diluents, carriers, pH modifiers, sweeteners, bulking agents, and further active agents.
  • a typical formulation is prepared by mixing a compound of the present invention and a carrier or excipient.
  • Suitable carriers and excipients are well known to those skilled in the art and are described in detail in, e.g., Ansel, Howard C, et al, Ansel's Pharmaceutical Dosage Forms and Drug Delivery Systems. Philadelphia: Lippincott, Williams & Wilkins, 2004; Gennaro, Alfonso R., et al. Remington: The Science and Practice of Pharmacy. Philadelphia: Lippincott, Williams & Wilkins, 2000; and Rowe, Raymond C. Handbook of Pharmaceutical Excipients. Chicago, Pharmaceutical Press, 2005.
  • the formulations may also include one or more buffers, stabilizing agents, surfactants, wetting agents, lubricating agents, emulsifiers, suspending agents, preservatives, antioxidants, opaquing agents, glidants, processing aids, colorants, sweeteners, perfuming agents, flavoring agents, diluents and other known additives to provide an elegant presentation of the drug (i.e., a compound of the present invention or pharmaceutical composition thereof) or aid in the manufacturing of the pharmaceutical product (i.e., medicament).
  • buffers stabilizing agents, surfactants, wetting agents, lubricating agents, emulsifiers, suspending agents, preservatives, antioxidants, opaquing agents, glidants, processing aids, colorants, sweeteners, perfuming agents, flavoring agents, diluents and other known additives to provide an elegant presentation of the drug (i.e., a compound of the present invention or pharmaceutical composition thereof) or aid in the manufacturing
  • An example of a suitable oral dosage form is a tablet containing about 2 mg, 5 mg, 25mg, 50mg, lOOmg, 250mg, or 500mg of the compound of the present invention compounded with about 95-30 mg anhydrous lactose, about 5-40 mg sodium croscarmellose, about 5-30mg polyvinylpyrrolidone (PVP) K30, and about e.g., 1-10 mg magnesium stearate.
  • the powdered ingredients are first mixed together and then mixed with a solution of the PVP.
  • the resulting composition can be dried, granulated, mixed with the magnesium stearate and compressed to tablet form using conventional equipment.
  • An example of an aerosol formulation can be prepared by dissolving the compound of the present invention, for example 5-400 mg, in a suitable buffer solution, e.g. a phosphate buffer, adding a tonicifier, e.g. a salt such sodium chloride, if desired.
  • a suitable buffer solution e.g. a phosphate buffer
  • a tonicifier e.g. a salt such sodium chloride
  • the solution may be filtered, e.g. using a 0.2 micron filter, to remove impurities and contaminants.
  • An embodiment therefore, includes a pharmaceutical composition comprising a compound of formula I, stereoisomers, tautomers or pharmaceutically acceptable salts thereof.
  • a pharmaceutical composition comprising a compound of formula I, or stereoisomers, tautomers or pharmaceutically acceptable salts thereof, together with a pharmaceutically acceptable carrier or excipient.
  • Another embodiment includes a pharmaceutical composition comprising a compound of formula I stereoisomers, tautomers or pharmaceutically acceptable salts thereof for use in the treatment of a hyperproliferative disease. Another embodiment includes a pharmaceutical composition comprising a compound of formula I stereoisomers, tautomers or pharmaceutically acceptable salts thereof for use in the treatment of cancer. Another embodiment includes a pharmaceutical composition comprising a compound of formula I stereoisomers, tautomers or pharmaceutically acceptable salts thereof for use in the treatment of an immunological disorder.
  • Another embodiment includes a pharmaceutical composition comprising a compound of formula I stereoisomers, tautomers or pharmaceutically acceptable salts thereof for use in the treatment of rheumatoid arthritis, psoriasis, inflammatory bowel disease (IBD) or asthma.
  • Another embodiment includes a pharmaceutical composition comprising a compound of formula I stereoisomers, tautomers or pharmaceutically acceptable salts thereof for use in the treatment of rheumatoid arthritis, asthma, systemic lupus erythematosus, psoriasis, IBD and transplant rejection.
  • the compounds of Formula I inhibit the activity of JAK1 kinase. Accordingly, the compounds of Formula I inhibit the phosphorylation of signal transducers and activators of transcription (STATs) by JAK1 kinase as well as STAT mediated cytokine production.
  • STATs signal transducers and activators of transcription
  • Compounds of Formula I are useful for inhibiting JAK1 kinase activity in cells through cytokine pathways, such as IL-6, IL-15, IL-7, IL-2, IL-4, IL-9, IL-10, IL-13, IL-21, G-CSF, IFNalpha, IFNbeta or IFNgamma pathways.
  • the compounds of Formula I can be used for the treatment of immunological disorders driven by aberrant IL-6, IL-15, IL-7, IL-2, IL-4, IL9, IL-10, IL-13, IL- 21, G-CSF, IFNalpha, IFNbeta or IFNgamma cytokine signaling.
  • Another embodiment includes a method of treating or lessening the severity of a disease or condition responsive to the inhibition of JAK1 kinase activity in a patient.
  • the method includes the step of administering to a patient a therapeutically effective amount of a compound of the present invention.
  • the disease or condition is cancer, stroke, diabetes, hepatomegaly, cardiovascular disease, multiple sclerosis, Alzheimer's disease, cystic fibrosis, viral disease, autoimmune diseases, atherosclerosis, restenosis, psoriasis, rheumatoid arthritis, inflammatory bowel disease, asthma, allergic disorders, inflammation, neurological disorders, a hormone- related disease, conditions associated with organ transplantation, immunodeficiency disorders, destructive bone disorders, proliferative disorders, infectious diseases, conditions associated with cell death, thrombin-induced platelet aggregation, liver disease, pathologic immune conditions involving T cell activation, CNS disorders or a myeloproliferative disorder.
  • the disease or condition is cancer.
  • the disease is a myeloproliferative disorder.
  • the myeloproliferative disorder is polycythemia vera, essential thrombocytosis, myelofibrosis or chronic myelogenous leukemia (CML).
  • the cancer is breast, ovary, cervix, prostate, testis, penile, genitourinary tract, seminoma, esophagus, larynx, gastric, stomach, gastrointestinal, skin, kerato acanthoma, follicular carcinoma, melanoma, lung, small cell lung carcinoma, non-small cell lung carcinoma (NSCLC), lung adenocarcinoma, squamous carcinoma of the lung, colon, pancreas, thyroid, papillary, bladder, liver, biliary passage, kidney, bone, myeloid disorders, lymphoid disorders, hairy cells, buccal cavity and pharynx (oral), lip, tongue, mouth, salivary gland, pharynx, small intestine, colon, rectum, anal
  • the cardiovascular disease is restenosis, cardiomegaly, atherosclerosis, myocardial infarction or congestive heart failure.
  • the neurodegenerative disease is Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, Huntington's disease, and cerebral ischemia, and neurodegenerative disease caused by traumatic injury, glutamate neurotoxicity or hypoxia.
  • the inflammatory diseases is rheumatoid arthritis, psoriasis, asthma, inflammatory bowel disease, contact dermatitis or delayed hypersensitivity reactions.
  • the autoimmune disease is lupus or multiple sclerosis.
  • the disease or condition responsive to the inhibition of JAK1 kinase is rheumatoid arthritis.
  • the disease or condition responsive to the inhibition of JAK1 kinase is rheumatoid arthritis, asthma, systemic lupus erythematosus, psoriasis, IBD or transplant rejection.
  • Another embodiment includes a method of treating cancer in a mammal in need of such treatment, wherein the method comprises administering to said mammal a therapeutically effective amount of a compound of formula I, a stereoisomer, tautomer, prodrug or pharmaceutically acceptable salt thereof.
  • Another embodiment includes compounds of formula I, a stereoisomer, tautomer, prodrug or pharmaceutically acceptable salt thereof, for use in therapy.
  • the therapy is the treatment of an immunological disorder, for example rheumatoid arthritis.
  • the therapy is the treatment of cancer.
  • Another embodiment includes compounds of formula I, a stereoisomer, tautomer, prodrug or pharmaceutically acceptable salt thereof, for use in treating a disease selected from rheumatoid arthritis, asthma, systemic lupus erythematosus, psoriasis, IBD and transplant rejection.
  • Another embodiment includes the use of a compound of formulas I, a stereoisomer, tautomer, prodrug or pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of a disease described herein (e.g., cancer or immunological disorder).
  • the compounds of formula I may be employed alone or in combination with other chemotherapeutic agents for treatment.
  • the compounds of the present invention can be used in combination with one or more additional drugs, for example an anti-hyperproliferative, anticancer, cytostatic, cytotoxic, anti-inflammatory or chemotherapeutic agent.
  • the second compound of the pharmaceutical combination formulation or dosing regimen preferably has complementary activities to the compound of this invention such that they do not adversely affect each other.
  • agents are suitably present in combination in amounts that are effective for the purpose intended.
  • the compounds may be administered together in a unitary pharmaceutical composition or separately and, when administered separately this may occur simultaneously or sequentially. Such sequential administration may be close or remote in time.
  • compounds of the present invention are coadministered with a cytostatic compound selected from the group consisting of cisplatin, doxorubicin, taxol, taxotere and mitomycin C.
  • the cytostatic compound is doxorubicin.
  • compounds of the present invention are coadministered with an anti-inflammatory agent selected from a NSAID and corticosteroid.
  • compounds of the present invention are coadministered with an anti-rheumatoid agent, in one example, RITUXAN®.
  • compounds of the present invention are coadministered with a chemotherapeutic agent selected from etanercept (Enbrel), infliximab (Remicade), adalimumab (Humira), certolizumab pegol (Cimzia), golimumab (Simponi), Interleukin 1 (IL-1) blockers such as anakinra (Kineret), monoclonal antibodies against B cells such as rituximab (RITUXAN®), T cell costimulation blockers such as abatacept (Orencia), Interleukin 6 (IL-6) blockers such as tocilizumab (ACTEMERA®); Interleukin 13 (IL-13) blockers such as lebrikizumab; Interferon alpha (IFN) blockers such as Rontalizumab; Beta 7 integrin blockers such as rhuMAb Beta7; IgE pathway blockers such as Anti-Mi prime; Secreted homotrimeric IL-1
  • the compounds of the present invention can be also used in combination with radiation therapy.
  • radiation therapy refers to the use of electromagnetic or particulate radiation in the treatment of neoplasia. Radiation therapy delivers doses of radiation sufficiently high to a target area to cause death of reproducing cells, in both tumor and normal tissues.
  • the radiation dosage regimen is generally defined in terms of radiation absorbed dose (rad), time and fractionation, and must be carefully defined by the oncologist. The amount of radiation a patient receives will depend on various considerations but two of the most important considerations are the location of the tumor in relation to other critical structures or organs of the body, and the extent to which the tumor has spread.
  • radiotherapeutic agents are provided in Hellman, Principles of Radiation Therapy, Cancer, in Principles I and Practice of Oncology, 24875 (Devita et al, 4th ed., vol 1, 1993).
  • Alternative forms of radiation therapy include three- dimensional conformal external beam radiation, intensity modulated radiation therapy (IMRT), stereotactic radiosurgery and brachytherapy (interstitial radiation therapy), the latter placing the source of radiation directly into the tumor as implanted "seeds".
  • IMRT intensity modulated radiation therapy
  • stereotactic radiosurgery stereotactic radiosurgery
  • brachytherapy interstitial radiation therapy
  • These alternative treatment modalities deliver greater doses of radiation to the tumor, which accounts for their increased effectiveness when compared to standard external beam radiation therapy.
  • Another embodiment includes a method of preparing a compound of formula I or a pharmaceutically acceptable salt thereof comprising: a, for a compound of formula I wherein Y is CR 5 , cyclizing a correspond
  • kits for treating a disease or disorder responsive to the inhibition of JAK1 kinase includes: (a) a first pharmaceutical composition comprising a compound of formula I; and
  • the kit further includes:
  • a second pharmaceutical composition which includes a chemotherapeutic agent.
  • the instructions describe the simultaneous, sequential or separate administration of said first and second pharmaceutical compositions to a patient in need thereof.
  • the first and second compositions are contained in separate containers.
  • the first and second compositions are contained in the same container.
  • Containers for use include, for example, bottles, vials, syringes, blister pack, etc.
  • the containers may be formed from a variety of materials such as glass or plastic.
  • the container includes a compound of formula I or formulation thereof which is effective for treating the condition and may have a sterile access port (for example the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
  • the container includes a composition comprising at least one compound of formula I.
  • the label or package insert indicates that the composition is used for treating the condition of choice, such as cancer.
  • the label or package inserts indicates that the composition comprising the compound of formula I can be used to treat a disorder.
  • the label or package insert may indicate that the patient to be treated is one having a disorder characterized by overactive or irregular kinase activity.
  • the label or package insert may also indicate that the composition can be used to treat other disorders.
  • the article of manufacture may comprise (a) a first container with a compound of formula I contained therein; and (b) a second container with a second pharmaceutical formulation contained therein, wherein the second pharmaceutical formulation comprises a chemotherapeutic agent.
  • the article of manufacture in this embodiment of the invention may further comprise a package insert indicating that the first and second compounds can be used to treat patients at risk of stroke, thrombus or thrombosis disorder.
  • the article of manufacture may further comprise a second (or third) container comprising a pharmaceutically-acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer's solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes.
  • BWFI bacteriostatic water for injection
  • phosphate-buffered saline such as bacteriostatic water for injection (BWFI),
  • HM-N Isolute® HM-N is a modified form of diatomaceous earth HOBt Hydro xybenzotriazole
  • the solvents used in preparing the example compounds were commercial anhydrous grade and were used without further drying or purification.
  • Method A Experiments performed on a Waters Micromass ZQ2000 quadrupole mass spectrometer linked to a Waters Acquity UPLC system with a PDA UV detector.
  • the spectrometer has an electrospray source operating in positive and negative ion mode.
  • This system uses an Acquity BEH CI 8 1.7um 100 x 2.1mm column, maintained at 40°C or an Acquity BEH Shield RP18 1.7 ⁇ 100 x 2.1mm column, maintained at 40°C and a 0.4 ml / minute flow rate.
  • the initial solvent system was 95% water containing 0.1% formic acid (solvent A) and 5% acetonitrile containing 0.1% formic acid (solvent B) for the first 0.4 minute followed by a gradient up to 5% solvent A and 95% solvent B over the next 5.6 minutes. This was maintained for 0.8 minute before returning to 95% solvent A and 5% solvent B over the next 1.2 minutes. Total run time was 8 minutes.
  • Method B Experiments performed on a Finnigan AQA single quadrupole mass spectrometer linked to a Hewlett Packard 1050 LC system with UV diode array detector and autosampler.
  • the spectrometer has an electrospray source operating in positive ion mode. Additional detection is achieved using a Sedex 65 evaporative light scattering detector.
  • This system uses a Luna 3 micron CI 8(2) 30 x 4.6mm column at ambient temperature and a 2.0 ml / minute flow rate.
  • the initial solvent system was 95% water containing 0.1% formic acid (solvent A) and 5% acetonitrile containing 0.1% formic acid (solvent B) for the first 0.5 minute followed by a gradient up to 5% solvent A and 95% solvent B over the next 4.0 minutes. This was maintained for 1.0 minute before returning to 95% solvent A and 5% solvent B over the next 0.5 minute. Total run time was 6 minutes.
  • Method C The system consists of a Waters Quattro Micro triple quadrupole mass spectrometer linked to a Hewlett Packard HP 1100 LC system with a PDA UV detector. Sample injection is done by a CTC HTS PAL autosampler.
  • the spectrometer has an electrospray source operating in positive and negative ion mode. This system uses a Higgins Clipeus 5micron CI 8 100 x 3.0mm column at ambient temperature and a 2.0 mL / minute flow rate.
  • the initial solvent system was 95% water containing 0.1% formic acid (solvent A) and 5% acetonitrile containing 0.1% formic acid (solvent B) for the first 0.5 minute followed by a gradient up to 5% solvent A and 95% solvent B over the next 14 minutes. This was maintained for 5 minutes before returning to 95% solvent A and 5% solvent B over the next 2 minutes. Total run time was 25 minutes.
  • Scan range 100-800amu
  • Method E Compounds were analysed using the following conditions: Experiments were performed on a The system consists of a Waters ZMD single quadrupole mass spectrometer linked to a Hewlett Packard HP 1100 LC system with UV diode array detector and 100 position autosampler. The spectrometer has an electrospray source operating in positive and negative ion mode. This system uses an Phenomenex Luna 3micron CI 8(2) 30 x 4.6mm column at ambient temperature, and a 2.0 ml / minute flow rate.
  • the initial solvent system was 95% water containing 0.1% formic acid (solvent A) and 5% acetonitrile containing 0.1% formic acid (solvent B) for the first 0.5 minute followed by a gradient up to 5%> solvent A and 95%> solvent B over the next 4 minutes. This was maintained for 1 minute before returning to 95% solvent A and 5% solvent B over the next 0.5 minute. Total run time was 6 minutes.
  • Method F Experiments were performed on a Waters Platform LC quadrupole mass spectrometer linked to a Hewlett Packard HP 1100 LC system with diode array detector and 100 position autosampler.
  • the spectrometer has an electrospray source operating in positive and negative ion mode. Additional detection is achieved using a Sedex 85 evaporative light scattering detector.
  • This system uses an Phenomenex Luna 3micron CI 8(2) 30 x 4.6mm column at ambient temperature, and a 2.0 ml / minute flow rate.
  • the initial solvent system was 95% water containing 0.1% formic acid (solvent A) and 5% acetonitrile containing 0.1% formic acid (solvent B) for the first 0.5 minute followed by a gradient up to 5% solvent A and 95% solvent B over the next 4 minutes. This was maintained for 1 minute before returning to 95% solvent A and 5% solvent B over the next 0.5 minute. Total run time was 6 minutes.
  • Method G Experiments were performed on a Waters ZMD quadrupole mass spectrometer linked to a Waters 1525 LC system with Waters 996 diode array detector.
  • the spectrometer has an electrospray source operating in positive and negative ion mode. Additional detection is achieved using a Sedex 85 evaporative light scattering detector.
  • This system uses an Luna 3micron CI 8(2) 30 x 4.6mm column at ambient temperature, and a 2.0 ml / minute flow rate.
  • the initial solvent system was 95% water containing 0.1% formic acid (solvent A) and 5% acetonitrile containing 0.1% formic acid (solvent B) for the first 0.5 minute followed by a gradient up to 5% solvent A and 95% solvent B over the next 4 minutes. This was maintained for 1 minute before returning to 95% solvent A and 5% solvent B over the next 0.5 minute. Total run time was 6 minutes.
  • Reverse Phase High Performance Liquid Chromatography was used to purify compounds where indicated. Unless otherwise indicated, the conditions were: elution on a Phenomenex Gemini CI 8 column (250 x 21.2 mm, 5 micron) as stationary phase and using mobile phase indicated, operating at a 18 mL/min flow rate using a Gilson UV/Vis -155 dual channel detector and Gilson GX-271 automated liquid handler.
  • Microwave experiments were carried out using a Biotage Initiator 2.0 (400 W MAGNETRON ® ) which uses a single-mode resonator and dynamic field tuning. Temperature from 40-250°C can be achieved, and pressures of up to 20 bar can be reached.
  • Trans-4-Amino-3-fluoro-piperidine-l-carboxylic acid tert-butyl ester A stirred solution of trans-4-benzylamino-3-fluoro-piperidine-l-carboxylic acid tert-butyl ester (0.17 g, 0.55 mmol) in methanol (2 mL) and was treated with ammonium formate (139 mg, 2.20 mmol) and 10% palladium on activated carbon (59.0 mg, 55.0 ⁇ ) and stirred at 50 °C for 1 hour.
  • the aqueous phase was extracted with ethyl acetate (x2) and the combined organic phase washed with 10% aqueous citric acid solution, saturated sodium hydrogen carbonate solution, and brine, dried with sodium sulfate and concentrated under vacuum to give 12.6 g of crude trans [4-(2-cyano-vinyl)- cyclohexyl]-carbamic acid tert-butyl ester as a ⁇ 1 :2 mixture of E- and Z- isomers which was used without further purification.
  • the golden oil crystallized almost immediately and was used in the next step without further purification.
  • Racemic trans (3-amino-cyclopentyl)-carbamic acid tert-butyl ester was prepared following the methods outlined in J. Org. Chem. 2004, 69(13), 4538; Tetrahedron 1997, 53(9), 3347; WO94/17090 and Org. Lett. 2000, 2, 4169.
  • Racemic cis (3-Amino-cyclopentyl)-carbamic acid tert-butyl ester Racemic cis (3-amino-cyclopentyl)-carbamic acid tert-butyl ester was prepared following the methods outlined in J. Org. Chem. 2004, 69, 4538; Tetrahedron 1997, 53, 3347; WO2008/065021 ; WO94/17090; and Org Lett 2000, 2, 4169.
  • Trans 3-(4-Amino-cyclohexylamino)-propionitrile can be prepared following the methods outlined in WO 1994/15596 or using the route described below.
  • the titled compound was prepared following the methods outlined in S. Fixon-Owoo et al. Phytochemistry 63 (2003) 315-334.
  • WO 2006071862 (0.98 g, 4.69 mmol) in propan-2-ol (12 mL) was treated with trans (4-amino-cyclohexyl)-acetonitrile (0.71 g, 5.14 mmol) and N,N- diisopropylethyl amine (DIPEA, 1.22 mL, 7.02 mmol) and the reaction mixture was heated at 120 °C using microwave irradiation for 20 minutes. On cooling, a solid precipitated out of solution.
  • DIPEA N,N- diisopropylethyl amine
  • the iron residue was collected on a pad of Celite® and washed several times with ethanol (IMS grade) and water (3: 1). The filtrate was concentrated in vacuo and the aqueous residue was treated with saturated sodium hydro gencarbonate solution and extracted with ethyl acetate (EtOAc) (3x). The combined organic phases were washed (saturated sodium hydro gencarbonate solution and brine), dried (sodium sulfate) and concentrated in vacuo to give 0.66 g of a brown gummy solid.
  • N-tert-Butylcarbamate-N- ⁇ l-[(lS,3R)-3-(2,2,2-trifluoro-ethylamino)-cyclopentyl]-lH- imidazo[4,5-c]quinolin-2-ylmethyl ⁇ -methanesulfonamide A suspension of ⁇ l-[(lS,3R)-3-(2,2,2-trifluoro-ethylamino)-cyclopentyl]-lH-imidazo[4,5- c]quinolin-2-yl ⁇ -methanol (139 mg, 0.38 mmol) in anhydrous THF (3.8 mL), under an atmosphere of nitrogen was treated with tert-butyl N-methanesulfonylcarbamate (164 mg, 0.84 mmol), followed by triphenylphosphine (200 mg, 0.76 mmol) and then diisopropyl azodicarboxylate (150 ⁇ , 0.76 mmol).
  • N- ⁇ l-[(lS,3R)-3-(2,2,2-Trifluoro-ethylamino)-cyclopentyl]-lH-imidazo[4,5-c]quinolin-2- ylmethyl ⁇ -methanesulfonamide A solution of N-tert-butylcarbamate-N- ⁇ l-[(lS,3R)-3-(2,2,2-trifluoro-ethylamino)-cyclopentyl]- lH-imidazo[4,5-c]quinolin-2-ylmethyl ⁇ -methanesulfonamide (165 mg, 0.30 mmol) in TFA (3 mL) was stirred at room temperature for 10 minutes.
  • Racemic cis [3 -(3 -Nitro-quinolin-4-ylamino)-cyclopentyl] -carbamic acid tert-butyl ester A mixture of racemic cis (3-amino-cyclopentyl)-carbamic acid tert-butyl ester (prepared following the methods outlined in J. Org. Chem. 2004, 69, 4538; Tetrahedron 1997, 53, 3347; WO2008/065021 ; WO94/17090; and Org. Lett.
  • a stirred suspension of triethyloxonium tetrafluoroborate (1.14 g, 6.01 mmol) in DCM (5 mL) was treated with (R)-(+)-lactamide (547 mg, 6.14 mmol) at room temperature for 2 hours. The volatiles were removed in vacuo and the resulting residue dissolved in ethanol (absolute grade, 5 mL) and stirred for 30 minutes.
  • DMSO-d6 DMSO-d6: ⁇ 9.19 (s, 1 H), 8.71 (t, 1 H), 8.17 (dd, 1 H), 7.74 (m, 2 H), 5.79 (t, 1 H), 5.60 (m, 1 H), 5.25 (m, 1 H), 3.40 (m, 1 H), 2.82 (m, 2 H), 2.66 (m, 2 H), 2.45 (m, 2 H), 2.15 (m, 3 H), 1.96 (m, l H), 1.68 (dd, 3 H).
  • the filtrate was concentrated under vacuum and the residue partitioned between saturated sodium hydro gencarbonate solution and ethyl acetate.
  • the aqueous phase was extracted with ethyl acetate (3 x) and the combined organic phases washed (brine), dried (sodium sulfate) and concentrated.
  • the mixture was purified by column chromatography using an Isolute® SCX-2 cartridge (gradient: MeOH to 2M NH 3 in MeOH) and then by column chromatography on silica gel (gradient: 0 to 15% [2M NH 3 in MeOH] in ethyl acetate). Further purification by column chromatography on silica gel (gradient: 0 to 10% MeOH in DCM) provided a pale yellow glass.

Abstract

The invention provides novel compounds of formula I having the general formula:(I) wherein Rl s R2, R3, X and Y are as described herein. Accordingly, the compounds may be provided in pharmaceutically acceptable compositions and used for the treatment of immunological or hyperproliferative disorders.

Description

TRICYCLIC HETEROCYCLIC COMPOUNDS, COMPOSITIONS AND METHODS
OF USE THEREOF AS JAK INHIBITORS
FIELD OF THE INVENTION
Compounds of formula I, which are inhibitors of a Janus kinase, as well as compositions containing these compounds, and methods of use including, but not limited to, in vitro, in situ and in vivo diagnosis or treatment of mammalian cells. BACKGROUND OF INVENTION
Cytokine pathways mediate a broad range of biological functions, including many aspects of inflammation and immunity. Janus kinases (JAK), including JAK1, JAK2, JAK3 and TYK2 are cytoplasmic protein kinases that associate with type I and type II cytokine receptors and regulate cytokine signal transduction. Cytokine engagement with cognate receptors triggers activation of receptor associated JAKs and this leads to JAK-mediated tyrosine phosphorylation of signal transducer and activator of transcription (STAT) proteins and ultimately transcriptional activation of specific gene sets (Schindler et al, 2007, J Biol. Chem. 282: 20059-63). JAK1, JAK2 and TYK2 exhibit broad patterns of gene expression, while JAK3 expression is limited to leukocytes. Cytokine receptors are typically functional as heterodimers, and as a result, more than one type of JAK kinase is usually associated with cytokine receptor complexes. The specific JAKs associated with different cytokine receptor complexes have been determined in many cases through genetic studies and corroborated by other experimental evidence.
JAK1 was initially identified in a screen for novel kinases (Wilks A.F., 1989, Proc. Natl. Acad. Sci. U.S.A. 86: 1603-1607). Genetic and biochemical studies have shown that JAK1 is functionally and physically associated with the type I interferon (e.g., IFNalpha), type II interferon (e.g., IFNgamma), IL-2 and IL-6 cytokine receptor complexes (Kisseleva et al, 2002, gene 285: 1-24; Levy et al, 2005, Nat. Rev. Mol. Cell Biol. 3:651-662; O'Shea et al, 2002, Cell, 109 (suppl): S121-S131). JAKl knockout mice die perinatally due to defects in LIF receptor signaling (Kisseleva et al, 2002, gene 285: 1-24; O'Shea et al, 2002, Cell, 109 (suppl): S121- S131). Characterization of tissues derived from JAKl knockout mice demonstrated critical roles for this kinase in the IFN, IL-10, IL-2/IL-4, and IL-6 pathways. A humanized monoclonal antibody targeting the IL-6 pathway (Tocilizumab) was recently approved by the European Commission for the treatment of moderate-to-severe rheumatoid arthritis (Scheinecker et al, 2009, Nat. Rev. Drug Discov. 8:273-274).
Biochemical and genetic studies have shown an association between JAK2 and single- chain (e.g., EPO), IL-3 and interferon gamma cytokine receptor families (Kisseleva et al, 2002, gene 285: 1-24; Levy et al, 2005, Nat. Rev. Mol. Cell Biol. 3:651-662; O'Shea et al, 2002, Cell, 109 (suppl): S121-S131). Consistent with this, JAK2 knockout mice die of anemia (O'Shea et al, 2002, Cell, 109 (suppl): S121-S131). Kinase activating mutations in JAK2 (e.g., JAK2 V617F) are associated with myeloproliferative disorders (MPDs) in humans.
JAK3 associates exclusively with the gamma common cytokine receptor chain, which is present in the IL-2, IL-4, IL-7, IL-9, IL-15 and IL-21 cytokine receptor complexes. JAK3 is critical for lymphoid cell development and proliferation and mutations in JAK3 result in severe combined immunodeficiency (SCID) (O'Shea et al, 2002, Cell, 109 (suppl): S121-S131). Based on its role in regulating lymphocytes, JAK3 and JAK3-mediated pathways have been targeted for immunosuppressive indications (e.g., transplantation rejection and rheumatoid arthritis) (Baslund et al, 2005, Arthritis & Rheumatism 52:2686-2692; Changelian et al, 2003, Science 302: 875-878).
TYK2 associates with the type I interferon (e.g., IFNalpha), IL-6, IL-10, IL-12 and IL-23 cytokine receptor complexes (Kisseleva et al, 2002, gene 285: 1-24; Watford, W.T. & O'Shea, J. J., 2006, Immunity 25:695-697). Consistent with this, primary cells derived from a TYK2 deficient human are defective in type I interferon, IL-6, IL-10, IL-12 and IL-23 signaling. A fully human monoclonal antibody targeting the shared p40 subunit of the IL-12 and 11-23 cytokines (Ustekinumab) was recently approved by the European Commission for the treatment of moderate-to-severe plaque psoriasis (Krueger et al, 2007, N. Engl. J. Med. 356:580-92; Reich et al, 2009, Nat. Rev. Drug Discov. 8:355-356). In addition, an antibody targeting the IL-12 and IL-23 pathways underwent clinical trials for treating Crohn's Disease (Mannon et al, 2004, N. Engl. J. Med. 351 :2069-79).
SUMMARY OF INVENTION
One aspect includes a compound of formula I:
Figure imgf000004_0001
stereoisomers, tautomers or pharmaceutically acceptable salts thereof, wherein X, Y, R1, R2 and R3 are defined herein.
Another aspect includes a pharmaceutical composition that includes a compound of formula I and a pharmaceutically acceptable carrier, adjuvant or vehicle.
Another aspect includes a method of treating or lessening the severity of a disease or condition responsive to the inhibition of JAK1 kinase activity in a patient. The method includes administering to the patient a therapeutically effective amount of a compound of formula I.
Another aspect includes a compound of formula I, a stereoisomer, tautomer, prodrug or pharmaceutically acceptable salt thereof, for use in therapy.
Another aspect includes the use of a compound of formula I, a stereoisomer, tautomer, prodrug or pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of a disease responsive to the inhibition of JAK1 kinase activity.
Another aspect includes a kit for treating a disease or disorder responsive to the inhibition of JAK1 kinase. The kit includes a first pharmaceutical composition comprising a compound of formula I and instructions for use
DETAILED DESCRIPTION OF THE INVENTION
DEFINITIONS
"Acyl" means a carbonyl containing substituent represented by the formula -C(0)-R in which R is hydrogen, alkyl, a cycloalkyl, a heterocyclyl, cycloalkyl -substituted alkyl or heterocyclyl-substituted alkyl wherein the alkyl, alkoxy, cycloalkyl and heterocyclyl are as defined herein. Acyl groups include alkanoyl (e.g. acetyl), aroyl (e.g. benzoyl), and heteroaroyl (e.g. pyridinoyl).
The term "alkyl" refers to a saturated linear or branched-chain monovalent hydrocarbon radical, wherein the alkyl radical may be optionally substituted independently with one or more substituents described herein. In one example, the alkyl radical is one to eighteen carbon atoms (Ci-Cis). In other examples, the alkyl radical is Co-C6, C0-C5, C0-C3, C1-C12, C1-C10, Ci-Cs, Ci- C6, C1-C5, C1-C4, or C1-C3. Co alkyl refers to a bond. Examples of alkyl groups include methyl (Me, -CH3), ethyl (Et, -CH2CH3), 1 -propyl (n-Pr, n-propyl, -CH2CH2CH3), 2-propyl (i-Pr, i- propyl, -CH(CH3)2), 1 -butyl (n-Bu, n-butyl, -CH2CH2CH2CH3), 2-methyl-l -propyl (i-Bu, i-butyl, -CH2CH(CH3)2), 2-butyl (s-Bu, s-butyl, -CH(CH3)CH2CH3), 2-methyl-2-propyl (t-Bu, t-butyl, - C(CH3)3), 1-pentyl (n-pentyl, -CH2CH2CH2CH2CH3), 2-pentyl (-CH(CH3)CH2CH2CH3), 3- pentyl (-CH(CH2CH3)2), 2-methyl-2-butyl (-C(CH3)2CH2CH3), 3-methyl-2-butyl (- CH(CH3)CH(CH3)2), 3-methyl-l -butyl (-CH2CH2CH(CH3)2), 2-methyl-l -butyl (- CH2CH(CH3)CH2CH3), 1-hexyl (-CH2CH2CH2CH2CH2CH3), 2-hexyl (- CH(CH3)CH2CH2CH2CH3), 3-hexyl (-CH(CH2CH3)(CH2CH2CH3)), 2-methyl-2-pentyl (- C(CH3)2CH2CH2CH3), 3-methyl-2-pentyl (-CH(CH3)CH(CH3)CH2CH3), 4-methyl-2-pentyl (- CH(CH3)CH2CH(CH3)2), 3-methyl-3-pentyl (-C(CH3)(CH2CH3)2), 2-methyl-3-pentyl (- CH(CH2CH3)CH(CH3)2), 2,3-dimethyl-2-butyl (-C(CH3)2CH(CH3)2), 3,3-dimethyl-2-butyl (- CH(CH3)C(CH3)3, 1 -heptyl and 1 -octyl.
The term "alkenyl" refers to linear or branched- chain monovalent hydrocarbon radical with at least one site of unsaturation, i.e., a carbon-carbon double bond, wherein the alkenyl radical may be optionally substituted independently with one or more substituents described herein, and includes radicals having "cis" and "trans" orientations, or alternatively, "E" and "Z" orientations. In one example, the alkenyl radical is two to eighteen carbon atoms (C2-Cis). In other examples, the alkenyl radical is C2-C12, C2-C10, C2-Cs, C2-C6 or C2-C3. Examples include, but are not limited to, ethenyl or vinyl (-CH=CH2), prop-l-enyl (-CH=CHCH3), prop-2-enyl (- CH2CH=CH2), 2-methylprop-l-enyl, but-l-enyl, but-2-enyl, but-3-enyl, buta-l,3-dienyl, 2- methylbuta-l,3-diene, hex-l-enyl, hex-2-enyl, hex-3-enyl, hex-4-enyl and hexa-l,3-dienyl. The term "alkoxy" refers to a linear or branched monovalent radical represented by the formula -OR in which R is alkyl, alkenyl, alkynyl or cycloalkyl, which can be further optionally substituted as defined herein. Alkoxy groups include methoxy, ethoxy, propoxy, isopropoxy, mono-, di- and tri-fluoromethoxy and cyclopropoxy.
The term "alkynyl" refers to a linear or branched monovalent hydrocarbon radical with at least one site of unsaturation, i.e., a carbon-carbon, triple bond, wherein the alkynyl radical may be optionally substituted independently with one or more substituents described herein. In one example, the alkynyl radical is two to eighteen carbon atoms (C2-C18). In other examples, the alkynyl radical is C2-C12, C2-C10, C2-Cs, C2-C6 or C2-C3. Examples include, but are not limited to, ethynyl (-C≡CH), prop-l-ynyl (-C≡CCH3), prop-2-ynyl (propargyl, -CH2C≡CH), but-l-ynyl, but-2-ynyl and but-3-ynyl.
"Alkylene" refers to a saturated, branched or straight chain hydrocarbon group having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent alkane. In one example, the divalent alkylene group is one to eighteen carbon atoms (Ci-Cis). In other examples, the divalent alkylene group is Co-C6, C0-C5, C0-C3, C1-C12, C1-C10, Ci-Cs, Ci-C6, C1-C5, C1-C4, or Ci-C3. The group Co alkylene refers to a bond. Example alkylene groups include methylene (-CH2-), 1,1 -ethyl (-CH(CH3)-), (1,2- ethyl (-CH2CH2-), 1,1-propyl (-CH(CH2CH3)-), 2,2-propyl (-C(CH3)2-), 1,2-propyl (-CH(CH3)CH2-), 1,3-propyl (-CH2CH2CH2-), l,l-dimethyleth-l,2-yl (-C(CH3)2CH2-), 1,4-butyl (-CH2CH2CH2CH2-), and the like.
"Alkenylene" refers to an unsaturated, branched or straight chain hydrocarbon group having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent alkene. In one example, the alkenylene group is two to eighteen carbon atoms (C2-Cis). In other examples, the alkenylene group is C2-C12, C2- C10, C2-Cs, C2-C6 or C2-C3. Example alkenylene groups include: 1,2-ethylene (-CH=CH-).
"Alkynylene" refers to an unsaturated, branched or straight chain hydrocarbon group having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent alkyne. In one example, the alkynylene radical is two to eighteen carbon atoms (C2-Cis). In other examples, the alkynylene radical is C2-C12, C2- C10, C2-Cs, C2-C6 or C2-C3. Example alkynylene radicals include: acetylene (-C≡C-), propargyl (-CH2C≡C-), and 4-pentynyl (-CH2CH2CH2C≡C-).
"Amidine" means the group -C(NH)-NHR in which R is hydrogen, alkyl, a cycloalkyl, a heterocyclyl, cycloalkyl-substituted alkyl or heterocyclyl-substituted alkyl wherein the alkyl, alkoxy, cycloalkyl and heterocyclyl are as defined herein. A particular amidine is the group - NH-C(NH)-NH2.
"Amino" means primary (i.e., -NH2) , secondary (i.e., -NRH) and tertiary (i.e., -NRR) amines, that are optionally substituted, in which R is alkyl, alkoxy, a cycloalkyl, a heterocyclyl, cycloalkyl-substituted alkyl or heterocyclyl-substituted alkyl wherein the alkyl, alkoxy, cycloalkyl and heterocyclyl are as defined herein Particular secondary and tertiary amines are alkylamine, dialkylamine, arylamine, diarylamine, aralkylamine and diaralkylamine wherein the alkyl is as herein defined and optionally substituted. Particular secondary and tertiary amines are methylamine, ethylamine, propylamine, isopropylamine, phenylamine, benzylamine dimethylamine, diethylamine, dipropylamine and diisopropylamine.
"Amino -protecting group" as used herein refers to a derivative of the groups commonly employed to block or protect an amino group while reactions are carried out on other functional groups on the compound. Examples of such protecting groups include carbamates, amides, alkyl and aryl groups, imines, as well as many N-heteroatom derivatives which can be removed to regenerate the desired amine group. Particular amino protecting groups are Pmb (p- Methoxybenzyl), Boc (tert-Butyloxycarbonyl), Fmoc (9-Fluorenylmethyloxycarbonyl) and Cbz (Carbobenzyloxy). Further examples of these groups are found in T. W. Greene and P. G. M. Wuts, "Protective Groups in Organic Synthesis", 2nd ed., John Wiley & Sons, Inc., New York, NY, 1991 , chapter 7; E. Haslam, "Protective Groups in Organic Chemistry", J. G. W. McOmie, Ed., Plenum Press, New York, NY, 1973, Chapter 5, and T.W. Greene, "Protective Groups in Organic Synthesis", John Wiley and Sons, New York, NY, 1981. The term "protected amino" refers to an amino group substituted with one of the above amino -protecting groups.
"Aryl" when used alone, or as part of another term, means a carbocyclic aromatic group, whether or not fused to one or more groups, having the number of carbon atoms designated, or if no number is designated, up to 14 carbon atoms. One example includes aryl groups having 6-14 carbon atoms. Another example inlcudes aryl groups having 6-10 carbon atoms. Examples of aryl groups include phenyl, naphthyl, biphenyl, phenanthrenyl, naphthacenyl, 1 ,2,3,4- tetrahydronaphthalenyl, lH-indenyl, 2,3-dihydro-lH-indenyl, and the like (see e.g. Lang 's Handbook of Chemistry (Dean, J. A., ed) 13th ed. Table 7-2 [1985]). A particular aryl is phenyl. Substituted phenyl or substituted aryl means a phenyl group or aryl group substituted with one, two, three, four or five, for example 1-2, 1-3 or 1-4 substituents chosen from groups specified herein. In one example, optional substituents on aryl are selected from halogen (F, CI, Br, I), hydroxy, protected hydroxy, cyano, nitro, alkyl (for example Ci-C6 alkyl), alkoxy (for example Ci-C6 alkoxy), benzyloxy, carboxy, protected carboxy, carboxymethyl, protected carboxymethyl, hydroxymethyl, protected hydroxymethyl, aminomethyl, protected aminomethyl, trifluoromethyl, alkylsulfonylamino, alkylsulfonylaminoalkyl, arylsulfonylamino, arylsulfonylaminoalkyl, heterocyclylsulfonylamino, heterocyclylsulfonylaminoalkyl, heterocyclyl, aryl, or other groups specified. One or more methyne (CH) and/or methylene (CH2) groups in these substituents may in turn be substituted with a similar group as those denoted above. Examples of the term "substituted phenyl" include a mono- or di(halo)phenyl group such as 2-chlorophenyl, 2-bromophenyl, 4-chlorophenyl, 2,6-dichlorophenyl, 2,5- dichlorophenyl, 3,4-dichlorophenyl, 3-chlorophenyl, 3-bromophenyl, 4-bromophenyl, 3,4- dibromophenyl, 3-chloro-4-fluorophenyl, 2-fluorophenyl and the like; a mono- or di(hydroxy)phenyl group such as 4-hydroxyphenyl, 3-hydroxyphenyl, 2,4-dihydroxyphenyl, the protected-hydroxy derivatives thereof and the like; a nitrophenyl group such as 3- or 4- nitrophenyl; a cyanophenyl group, for example, 4-cyanophenyl; a mono- or di(lower alkyl)phenyl group such as 4-methylphenyl, 2,4-dimethylphenyl, 2-methylphenyl, 4- (isopropyl)phenyl, 4-ethylphenyl, 3-(n-propyl)phenyl and the like; a mono or di(alkoxy)phenyl group, for example, 3,4-dimethoxyphenyl, 3-methoxy-4-benzyloxyphenyl, 3-ethoxyphenyl, 4- (isopropoxy)phenyl, 4-(t-butoxy)phenyl, 3-ethoxy-4-methoxyphenyl and the like; 3- or 4- trifluoromethylphenyl; a mono- or dicarboxyphenyl or (protected carboxy)phenyl group such 4- carboxyphenyl, a mono- or di(hydroxymethyl)phenyl or (protected hydroxymethyl)phenyl such as 3-(protected hydroxymethyl)phenyl or 3,4-di(hydroxymethyl)phenyl; a mono- or di(aminomethyl)phenyl or (protected aminomethyl)phenyl such as 2-(aminomethyl)phenyl or 2,4-(protected aminomethyl)phenyl; or a mono- or di(N-(methylsulfonylamino))phenyl such as 3-(N-methylsulfonylamino))phenyl. Also, the term "substituted phenyl" represents disubstituted phenyl groups where the substituents are different, for example, 3-methyl-4-hydroxyphenyl, 3- chloro-4-hydroxyphenyl, 2-methoxy-4-bromophenyl, 4-ethyl-2-hydroxyphenyl, 3-hydroxy-4- nitrophenyl, 2-hydroxy-4-chlorophenyl, and the like, as well as trisubstituted phenyl groups where the substituents are different, for example 3-methoxy-4-benzyloxy-6-methyl sulfonylamino, 3-methoxy-4-benzyloxy-6-phenyl sulfonylamino, and tetrasubstituted phenyl groups where the substituents are different such as 3-methoxy-4-benzyloxy-5-methyl-6-phenyl sulfonylamino. Particular substituted phenyl groups include the 2-chlorophenyl, 2-aminophenyl, 2-bromophenyl, 3-methoxyphenyl, 3-ethoxy-phenyl, 4-benzyloxyphenyl, 4-methoxyphenyl, 3- ethoxy-4-benzyloxyphenyl, 3,4-diethoxyphenyl, 3-methoxy-4-benzyloxyphenyl, 3-methoxy-4- (l-chloromethyl)benzyloxy-6-methyl sulfonyl aminophenyl groups. Fused aryl rings may also be substituted with any, for example 1 , 2 or 3, of the substituents specified herein in the same manner as substituted alkyl groups.
The terms "cancer" and "cancerous", "neoplasm", "tumor" refer to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth. A "tumor" comprises one or more cancerous cells. Examples of cancer include carcinoma, lymphoma, blastoma, sarcoma, and leukemia or lymphoid malignancies. More particular examples of such cancers include squamous cell cancer (e.g., epithelial squamous cell cancer), lung cancer including small- cell lung cancer, non-small cell lung cancer ("NSCLC"), adenocarcinoma of the lung and squamous carcinoma of the lung, cancer of the peritoneum, hepatocellular cancer, gastric or stomach cancer including gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatoma, breast cancer, colon cancer, rectal cancer, colorectal cancer, endometrial or uterine carcinoma, salivary gland carcinoma, kidney or renal cancer, prostate cancer, vulval cancer, thyroid cancer, hepatic carcinoma, anal carcinoma, penile carcinoma, melanoma, multiple myeloma and B-cell lymphoma, brain, as well as head and neck cancer, and associated metastases.
A "chemotherapeutic agent" is an agent useful in the treatment of a given disorder, for example, cancer or inflammatory disorders. Examples of chemotherapeutic agents include NSAIDs; hormones such as glucocorticoids; corticosteroids such as hydrocortisone, hydrocortisone acetate, cortisone acetate, tixocortol pivalate, prednisolone, methylprednisolone, prednisone, triamcinolone acetonide, triamcinolone alcohol, mometasone, amcinonide, budesonide, desonide, fluocinonide, fluocinolone acetonide, halcinonide, betamethasone, betamethasone sodium phosphate, dexamethasone, dexamethasone sodium phosphate, fluocortolone, hydrocortisone- 17-butyrate, hydrocortisone- 17-valerate, aclometasone dipropionate, betamethasone valerate, betamethasone dipropionate, prednicarbate, clobetasone- 17-butyrate, clobetasol-17-propionate, fluocortolone caproate, fluocortolone pivalate and fluprednidene acetate; immune selective anti-inflammatory peptides (ImSAIDs) such as phenylalanine-glutamine-glycine (FEG) and its D-isomeric form (feG) (IMULAN BioTherapeutics, LLC); anti-rheumatic drugs such as azathioprine, ciclosporin (cyclosporine A), D-penicillamine, gold salts, hydroxychloroquine, leflunomide, methotrexate (MTX), minocycline, sulfasalazine, cyclophosphamide, tumor necrosis factor alpha (TNFa) blockers such as etanercept (Enbrel), infliximab (Remicade), adalimumab (Humira), certolizumab pegol (Cimzia), golimumab (Simponi), Interleukin 1 (IL-1) blockers such as anakinra (Kineret), monoclonal antibodies against B cells such as rituximab (RITUXAN®), T cell costimulation blockers such as abatacept (Orencia), Interleukin 6 (IL-6) blockers such as tocilizumab (ACTEMERA®); Interleukin 13 (IL-13) blockers such as lebrikizumab; Interferon alpha (IFN) blockers such as Rontalizumab; Beta 7 integrin blockers such as rhuMAb Beta7; IgE pathway blockers such as Anti-Mi prime; Secreted homotrimeric LTa3 and membrane bound heterotrimer LTal/p2 blockers such as Anti-lymphotoxin alpha (LTa); hormone antagonists,
211 131 125 such as tamoxifen, finasteride or LHRH antagonists; radioactive isotopes (e.g., At , 1 , 1 , Y90, Re186, Re188, Sm153, Bi212, P32, Pb212 and radioactive isotopes of Lu); miscellaneous investigational agents such as thioplatin, PS-341, phenylbutyrate, ET-18- OCH3, or farnesyl transferase inhibitors (L-739749, L-744832); polyphenols such as quercetin, resveratrol, piceatannol, epigallocatechine gallate, theaflavins, flavanols, procyanidins, betulinic acid and derivatives thereof; autophagy inhibitors such as chloroquine; alkylating agents such as thiotepa and cyclosphosphamide (CYTOXAN®); alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, triethylenephosphoramide, triethylenethiophosphoramide and trimethylomelamine; acetogenins (especially bullatacin and bullatacinone); delta-9-tetrahydrocannabinol (dronabinol, MARINOL®); beta-lapachone; lapachol; colchicines; betulinic acid; a camptothecin (including the synthetic analogue topotecan (HYCAMTIN®), CPT-11 (irinotecan, CAMPTOSA ®), acetylcamptothecin, scopolectin, and 9-aminocamptothecin); bryostatin; callystatin; CC-1065 (including its adozelesin, carzelesin and bizelesin synthetic analogues); podophyllotoxin; podophyllinic acid; teniposide; cryptophycins (particularly cryptophycin 1 and cryptophycin 8); dolastatin; duocarmycin (including the synthetic analogues, KW-2189 and CB1-TM1); eleutherobin; pancratistatin; a sarcodictyin; spongistatin; nitrogen mustards such as chlorambucil, chlornaphazine, chlorophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard; nitrosoureas such as carmustine, chlorozotocin, fotemustine, lomustine, nimustine, and ranimnustine; antibiotics such as the enediyne antibiotics (e. g., calicheamicin, especially calicheamicin gammall and calicheamicin omegall (see, e.g., Nicolaou et al., Angew. Chem Intl. Ed. Engl., 33: 183-186 (1994)); CDP323, an oral alpha-4 integrin inhibitor; dynemicin, including dynemicin A; an esperamicin; as well as neocarzino statin chromophore and related chromoprotein enediyne antibiotic chromophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, carabicin, carminomycin, carzinophilin, chromomycins, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, doxorubicin (including ADRIAMYCIN®, morpholino-doxorubicin, cyanomorpholino-doxorubicin, 2-pyrrolino- doxorubicin, doxorubicin HC1 liposome injection (DOXIL®), liposomal doxorubicin TLC D-99 (MYOCET®), peglylated liposomal doxorubicin (CAELYX®), and deoxydoxorubicin), epirubicin, esorubicin, idarubicin, marcellomycin, mitomycins such as mitomycin C, mycophenolic acid, nogalamycin, olivomycins, peplomycin, porfiromycin, puromycin, quelamycin, rodorubicin, streptonigrin, streptozocin, tubercidin, ubenimex, zinostatin, zorubicin; anti-metabolites such as methotrexate, gemcitabine (GEMZAR®), tegafur (UFTORAL®), capecitabine (XELODA®), an epothilone, and 5-fluorouracil (5-FU); folic acid analogues such as denopterin, methotrexate, pteropterin, trimetrexate; purine analogs such as fludarabine, 6- mercaptopurine, thiamiprine, thioguanine; pyrimidine analogs such as ancitabine, azacitidine, 6- azauridine, carmofur, cytarabine, dideoxyuridine, doxifluridine, enocitabine, fioxuridine; androgens such as calusterone, dromostanolone propionate, epitiostanol, mepitiostane, testolactone; anti-adrenals such as aminoglutethimide, mitotane, trilostane; folic acid replenisher such as frolinic acid; aceglatone; aldophosphamide glycoside; aminolevulinic acid; eniluracil; amsacrine; bestrabucil; bisantrene; edatraxate; defofamine; demecolcine; diaziquone; elfornithine; elliptinium acetate; an epothilone; etoglucid; gallium nitrate; hydroxyurea; lentinan; lonidainine; maytansinoids such as maytansine and ansamitocins; mitoguazone; mitoxantrone; mopidanmol; nitraerine; pentostatin; phenamet; pirarubicin; losoxantrone; 2-ethylhydrazide; procarbazine; PSK® polysaccharide complex (JHS Natural Products, Eugene, OR); razoxane; rhizoxin; sizofiran; spirogermanium; tenuazonic acid; triaziquone; 2,2',2'-trichlorotriethylamine; trichothecenes (especially T-2 toxin, verracurin A, roridin A and anguidine); urethan; vindesine (ELDISINE®, FILDESIN®); dacarbazine; mannomustine; mitobronitol; mitolactol; pipobroman; gacytosine; arabinoside ("Ara-C"); thiotepa; taxoid, e.g., paclitaxel (TAXOL®), albumin- engineered nanoparticle formulation of paclitaxel (ABRAXANE™), and docetaxel (TAXOTERE®); chloranbucil; 6-thioguanine; mercaptopurine; methotrexate; platinum agents such as cisp latin, oxalip latin (e.g., ELOXATIN®), and carboplatin; vincas, which prevent tubulin polymerization from forming microtubules, including vinblastine (VELBAN®), vincristine (ONCOVIN®), vindesine (ELDISINE®, FILDESIN®), and vinorelbine (NAVELBINE®); etoposide (VP- 16); ifosfamide; mitoxantrone; leucovorin; novantrone; edatrexate; daunomycin; aminopterin; ibandronate; topoisomerase inhibitor RFS 2000; difluoromethylornithine (DMFO); retinoids such as fenretinide, retinoic acid, including bexarotene (TARGRETIN®); bisphosphonates such as clodronate (for example, BONEFOS® or OSTAC®), etidronate (DIDROCAL®), NE-58095, zoledronic acid/zoledronate (ZOMETA®), alendronate (FOSAMAX®), pamidronate (AREDIA®), tiludronate (SKELID®), or risedronate (ACTONEL®); troxacitabine (a 1 ,3-dioxolane nucleoside cytosine analog); antisense oligonucleotides, particularly those that inhibit expression of genes in signaling pathways implicated in aberrant cell proliferation, such as, for example, PKC-alpha, Raf, H-Ras, and epidermal growth factor receptor (EGF-R); vaccines such as THERATOPE® vaccine and gene therapy vaccines, for example, ALLOVECTIN® vaccine, LEUVECTIN® vaccine, and VAXID® vaccine; topoisomerase 1 inhibitor (e.g., LURTOTECAN®); rmRH (e.g., ABARELIX®); BAY439006 (sorafenib; Bayer); SU-1 1248 (sunitinib, SUTENT®, Pfizer); perifosine, COX-2 inhibitor (e.g. celecoxib or etoricoxib), proteosome inhibitor (e.g. PS341); bortezomib (VELCADE®); CCI-779; tipifarnib (R1 1577); orafenib, ABT510; Bcl-2 inhibitor such as oblimersen sodium (GENASENSE®); pixantrone; EGFR inhibitors (see definition below); farnesyltransferase inhibitors such as lonafarnib (SCH 6636, SARASAR™); and pharmaceutically acceptable salts, acids or derivatives of any of the above; as well as combinations of two or more of the above such as CHOP, an abbreviation for a combined therapy of cyclophosphamide, doxorubicin, vincristine, and prednisolone; and FOLFOX, an abbreviation for a treatment regimen with oxaliplatin (ELOXATIN™) combined with 5-FU and leucovorin. Additional chemotherapeutic agents as defined herein include "anti-hormonal agents" or
"endocrine therapeutics" which act to regulate, reduce, block, or inhibit the effects of hormones that can promote the growth of cancer. They may be hormones themselves, including, but not limited to: anti-estrogens with mixed agonist/antagonist profile, including, tamoxifen (NOLVADEX®), 4-hydroxytamoxifen, toremifene (FARESTON®), idoxifene, droloxifene, raloxifene (EVISTA®), trioxifene, keoxifene, and selective estrogen receptor modulators (SERMs) such as SERM3; pure anti-estrogens without agonist properties, such as fulvestrant (FASLODEX®), and EM800 (such agents may block estrogen receptor (ER) dimerization, inhibit DNA binding, increase ER turnover, and/or suppress ER levels); aromatase inhibitors, including steroidal aromatase inhibitors such as formestane and exemestane (AROMASIN®), and nonsteroidal aromatase inhibitors such as anastrazole (ARIMIDEX®), letrozole (FEMARA®) and aminoglutethimide, and other aromatase inhibitors include vorozole (RIVISOR®), megestrol acetate (MEGASE®), fadrozole, and 4(5)-imidazoles; lutenizing hormone-releaseing hormone agonists, including leuprolide (LUPRON® and ELIGARD®), goserelin, buserelin, and tripterelin; sex steroids, including progestines such as megestrol acetate and medroxyprogesterone acetate, estrogens such as diethylstilbestrol and premarin, and androgens/retinoids such as fluoxymesterone, all transretionic acid and fenretinide; onapristone; anti-progesterones; estrogen receptor down-regulators (ERDs); anti-androgens such as flutamide, nilutamide and bicalutamide.
Additional chemotherapeutic agents include therapeutic antibodies such as alemtuzumab (Campath), bevacizumab (AVASTIN®, Genentech); cetuximab (ERBITUX®, Imclone); panitumumab (VECTIBIX®, Amgen), rituximab (RITUXAN®, Genentech/Biogen Idee), pertuzumab (OMNITARG®, 2C4, Genentech), trastuzumab (HERCEPTIN®, Genentech), tositumomab (Bexxar, Corixia), and the antibody drug conjugate, gemtuzumab ozogamicin (MYLOTARG®, Wyeth). Additional humanized monoclonal antibodies with therapeutic potential as agents in combination with the compounds of the invention include: apolizumab, aselizumab, atlizumab, bapineuzumab, bivatuzumab mertansine, cantuzumab mertansine, cedelizumab, certolizumab pegol, cidfusituzumab, cidtuzumab, daclizumab, eculizumab, efalizumab, epratuzumab, erlizumab, felvizumab, fontolizumab, gemtuzumab ozogamicin, inotuzumab ozogamicin, ipilimumab, labetuzumab, lintuzumab, matuzumab, mepolizumab, motavizumab, motovizumab, natalizumab, nimotuzumab, nolovizumab, numavizumab, ocrelizumab, omalizumab, palivizumab, pascolizumab, pecfusituzumab, pectuzumab, pexelizumab, ralivizumab, ranibizumab, reslivizumab, reslizumab, resyvizumab, rovelizumab, ruplizumab, sibrotuzumab, siplizumab, sontuzumab, tacatuzumab tetraxetan, tadocizumab, talizumab, tefibazumab, tocilizumab, toralizumab, tucotuzumab celmoleukin, tucusituzumab, umavizumab, urtoxazumab, ustekinumab, visilizumab, and the anti-interleukin-12 (ABT- 874/J695, Wyeth Research and Abbott Laboratories) which is a recombinant exclusively human- sequence, full-length IgGi λ antibody genetically modified to recognize interleukin-12 p40 protein.
Chemotherapeutic agents also include "EGFR inhibitors," which refers to compounds that bind to or otherwise interact directly with EGFR and prevent or reduce its signaling activity, and is alternatively referred to as an "EGFR antagonist." Examples of such agents include antibodies and small molecules that bind to EGFR. Examples of antibodies which bind to EGFR include MAb 579 (ATCC CRL HB 8506), MAb 455 (ATCC CRL HB8507), MAb 225 (ATCC CRL 8508), MAb 528 (ATCC CRL 8509) (see, US Patent No. 4,943, 533, Mendelsohn et al.) and variants thereof, such as chimerized 225 (C225 or Cetuximab; ERBUTIX®) and reshaped human 225 (H225) (see, WO 96/40210, Imclone Systems Inc.); IMC-11F8, a fully human, EGFR-targeted antibody (Imclone); antibodies that bind type II mutant EGFR (US Patent No. 5,212,290); humanized and chimeric antibodies that bind EGFR as described in US Patent No. 5,891,996; and human antibodies that bind EGFR, such as ABX-EGF or Panitumumab (see WO98/50433, Abgenix/Amgen); EMD 55900 (Stragliotto et al. Eur. J. Cancer 32A:636-640 (1996)); EMD7200 (matuzumab) a humanized EGFR antibody directed against EGFR that competes with both EGF and TGF-alpha for EGFR binding (EMD/Merck); human EGFR antibody, HuMax-EGFR (GenMab); fully human antibodies known as El . l, E2.4, E2.5, E6.2, E6.4, E2. l l, E6. 3 and E7.6. 3 and described in US 6,235,883; MDX-447 (Medarex Inc); and mAb 806 or humanized mAb 806 (Johns et al, J. Biol. Chem. 279(29):30375-30384 (2004)). The anti-EGFR antibody may be conjugated with a cytotoxic agent, thus generating an immuno conjugate (see, e.g., EP659,439A2, Merck Patent GmbH). EGFR antagonists include small molecules such as compounds described in US Patent Nos: 5,616,582, 5,457,105, 5,475,001, 5,654,307, 5,679,683, 6,084,095, 6,265,410, 6,455,534, 6,521,620, 6,596,726, 6,713,484, 5,770,599, 6,140,332, 5,866,572, 6,399,602, 6,344,459, 6,602,863, 6,391,874, 6,344,455, 5,760,041, 6,002,008, and 5,747,498, as well as the following PCT publications: W098/14451, WO98/50038, WO99/09016, and WO99/24037. Particular small molecule EGFR antagonists include OSI-774 (CP-358774, erlotinib, TARCEVA® Genentech/OSI Pharmaceuticals); PD 183805 (CI 1033, 2-propenamide, N-[4-[(3-chloro-4-fluorophenyl)amino]- 7-[3-(4-morpholinyl)propoxy]-6-quinazolinyl]-, dihydrochloride, Pfizer Inc.); ZD 1839, gefitinib (IRESSAJ) 4-(3'-Chloro-4'-fluoroanilino)-7-methoxy-6-(3-morpholinopropoxy)quinazoline, AstraZeneca); ZM 105180 ((6-amino-4-(3-methylphenyl-amino)-quinazoline, Zeneca); BIBX- 1382 (N8-(3-chloro-4-fluoro-phenyl)-N2-( 1 -methyl-piperidin-4-yl)-pyrimido [5 ,4-d]pyrimidine- 2,8-diamine, Boehringer Ingelheim); PKI-166 ((R)-4-[4-[(l-phenylethyl)amino]-lH-pyrrolo[2,3- d]pyrimidin-6-yl] -phenol); (R)-6-(4-hydroxyphenyl)-4-[(l-phenylethyl)amino]-7H-pyrrolo[2,3- d]pyrimidine) ; CL-387785 (N- [4- [(3 -bromophenyl)amino] -6-quinazo linyl] -2-butynamide) ; EKB-569 (N-[4-[(3-chloro-4-fluorophenyl)amino]-3-cyano-7-ethoxy-6-quinolinyl]-4- (dimethylamino)-2-butenamide) (Wyeth); AG1478 (Pfizer); AG1571 (SU 5271 ; Pfizer); dual EGFR/HER2 tyrosine kinase inhibitors such as lapatinib (TYKERB®, GSK572016 or N-[3- chloro-4- [(3 fluorophenyl)methoxy]phenyl] -6 [5 [ [ [2methylsulfonyl)ethyl] amino]methyl] -2- furanyl] -4-quinazolinamine) . Chemotherapeutic agents also include "tyrosine kinase inhibitors" including the EGFR- targeted drugs noted in the preceding paragraph; small molecule HER2 tyrosine kinase inhibitor such as TAK165 available from Takeda; CP-724,714, an oral selective inhibitor of the ErbB2 receptor tyrosine kinase (Pfizer and OSI); dual-HER inhibitors such as EKB-569 (available from Wyeth) which preferentially binds EGFR but inhibits both HER2 and EGFR-overexpressing cells; lapatinib (GSK572016; available from Glaxo-SmithKline), an oral HER2 and EGFR tyrosine kinase inhibitor; PKI-166 (available from Novartis); pan-HER inhibitors such as canertinib (CI-1033; Pharmacia); Raf-1 inhibitors such as antisense agent ISIS-5132 available from ISIS Pharmaceuticals which inhibit Raf-1 signaling; non-HER targeted TK inhibitors such as imatinib mesylate (GLEEVECJ, available from Glaxo SmithKline); multi-targeted tyrosine kinase inhibitors such as sunitinib (SUTENT®, available from Pfizer); VEGF receptor tyrosine kinase inhibitors such as vatalanib (PTK787/ZK222584, available from Novartis/Schering AG); MAPK extracellular regulated kinase I inhibitor CI- 1040 (available from Pharmacia); quinazolines, such as PD 153035,4-(3-chloroanilino) quinazoline; pyridopyrimidines; pyrimidopyrimidines; pyrrolopyrimidines, such as CGP 59326, CGP 60261 and CGP 62706; pyrazolopyrimidines, 4-(phenylamino)-7H-pyrrolo[2,3-d] pyrimidines; curcumin (diferuloyl methane, 4,5-bis (4-fluoroanilino)phthalimide); tyrphostines containing nitrothiophene moieties; PD-0183805 (Warner-Lamber); antisense molecules (e.g. those that bind to HER-encoding nucleic acid); quinoxalines (US Patent No. 5,804,396); tryphostins (US Patent No. 5,804,396); ZD6474 (Astra Zeneca); PTK-787 (Novartis/Schering AG); pan-HER inhibitors such as CI-1033 (Pfizer); Affmitac (ISIS 3521; Isis/Lilly); imatinib mesylate (GLEEVECJ); PKI 166 (Novartis); GW2016 (Glaxo SmithKline); CI- 1033 (Pfizer); EKB-569 (Wyeth); Semaxinib (Pfizer); ZD6474 (AstraZeneca); PTK-787 (Novartis/Schering AG); INC-lCl l (Imclone), rapamycin (sirolimus, RAPAMUNE®); or as described in any of the following patent publications: US Patent No. 5,804,396; WO 1999/09016 (American Cyanamid); WO 1998/43960 (American Cyanamid); WO 1997/38983 (Warner Lambert); WO 1999/06378 (Warner Lambert); WO 1999/06396 (Warner Lambert); WO 1996/30347 (Pfizer, Inc); WO 1996/33978 (Zeneca); WO 1996/3397 (Zeneca) and WO 1996/33980 (Zeneca).
The term "NSAID" and the terms "non-steroidal anti- inflammatory drug" refer to therapeutic agents with analgesic, antipyretic and anti- inflammatory effects. NSAIDs include non-selective inhibitors of the enzyme cyclooxygenase. Specific examples of NSAIDs include aspirin, propionic acid derivatives such as ibuprofen, fenoprofen, ketoprofen, flurbiprofen, oxaprozin and naproxen, acetic acid derivatives such as indomethacin, sulindac, etodolac, diclofenac, enolic acid derivatives such as piroxicam, meloxicam, tenoxicam, droxicam, lornoxicam and isoxicam, fenamic acid derivatives such as mefenamic acid, meclofenamic acid, flufenamic acid, tolfenamic acid, and COX-2 inhibitors such as celecoxib, etoricoxib, lumiracoxib, parecoxib, rofecoxib, rofecoxib, and valdecoxib. NSAIDs can be indicated for the symptomatic relief of conditions such as rheumatoid arthritis, osteoarthritis, inflammatory arthropathies, ankylosing spondylitis, psoriatic arthritis, Reiter's syndrome, acute gout, dysmenorrhoea, metastatic bone pain, headache and migraine, postoperative pain, mild-to- moderate pain due to inflammation and tissue injury, pyrexia, ileus, and renal colic.
Additionally, chemotherapeutic agents include pharmaceutically acceptable salts, acids or derivatives of any of chemotherapeutic agents, described herein, as well as combinations of two or more of them. "Cycloalkyl" refers to a non-aromatic, saturated or partially unsaturated hydrocarbon ring group wherein the cycloalkyl group may be optionally substituted independently with one or more substituents described herein. In one example, the cycloalkyl group is 3 to 12 carbon atoms (C3-C12). In other examples, cycloalkyl is C3-C8, C3-Cio or C5-C10. In other examples, the cycloalkyl group, as a monocycle, is C3-C8, C3-C6 or C5-C6. In another example, the cycloalkyl group, as a bicycle, is C7-C12. In another example, the cycloalkyl group, as a spiro system, is C5- C12. Examples of monocyclic cycloalkyl include cyclopropyl, cyclo butyl, cyclopentyl, 1- cyclopent-l-enyl, l-cyclopent-2-enyl, l-cyclopent-3-enyl, cyclo hexyl, perdeuteriocyclohexyl, 1- cyclohex-l-enyl, l-cyclohex-2-enyl, 1 -cyclo hex-3-enyl, cyclo hexadienyl, cyclo heptyl, cyclooctyl, cyclononyl, cyclodecyl, cycloundecyl and cyclododecyl. Exemplary arrangements of bicyclic cycloalkyls having 7 to 12 ring atoms include, but are not limited to, [4,4], [4,5], [5,5], [5,6] or [6,6] ring systems. Exemplary bridged bicyclic cycloalkyls include, but are not limited to, bicyclo[2.2.1]heptane, bicyclo[2.2.2]octane and bicyclo[3.2.2]nonane. Examples of spiro cycloalkyl include, spiro[2.2]pentane, spiro[2.3]hexane, spiro[2.4]heptane, spiro[2.5]octane and spiro[4.5]decane.
"Carboxy-protecting group" as used herein refers to those groups that are stable to the conditions of subsequent reaction(s) at other positions of the molecule, which may be removed at the appropriate point without disrupting the remainder of the molecule, to give the unprotected carboxy-group. Examples of carboxy protecting groups include, ester groups and heterocyclyl groups. Ester derivatives of the carboxylic acid group may be employed to block or protect the carboxylic acid group while reactions are carried out on other functional groups on the compound. Examples of such ester groups include substituted arylalkyl, including substituted benzyls, such as 4-nitrobenzyl, 4-methoxybenzyl, 3,4-dimethoxybenzyl, 2,4-dimethoxybenzyl, 2,4,6-trimethoxybenzyl, 2,4,6-trimethylbenzyl, pentamethylbenzyl, 3,4-methylenedioxybenzyl, benzhydryl, 4,4'-dimethoxybenzhydryl, 2,2',4,4'-tetramethoxybenzhydryl, alkyl or substituted alkyl esters such as methyl, ethyl, t-butyl allyl or t-amyl, triphenylmethyl (trityl), 4- methoxytrityl, 4,4'-dimethoxytrityl, 4,4',4"-trimethoxytrityl, 2-phenylprop-2-yl, thioesters such as t-butyl thioester, silyl esters such as trimethylsilyl, t-butyldimethylsilyl esters, phenacyl, 2,2,2- trichloro ethyl, beta-(trimethylsilyl)ethyl, beta-(di(n-butyl)methylsilyl)ethyl, p- toluenesulfonylethyl, 4-nitrobenzylsulfonylethyl, allyl, cinnamyl, l-(trimethylsilylmethyl)prop- l-en-3-yl, and like moieties. Another example of carboxy-protecting groups are heterocyclyl groups such as 1,3-oxazolinyl. Further examples of these groups are found in T. W. Greene and P. G. M. Wuts, "Protective Groups in Organic Synthesis", 2nd ed., John Wiley & Sons, Inc., New York, N.Y., 1991, chapter 5; E. Haslam, "Protective Groups in Organic Chemistry", J. G. W. McOmie, Ed., Plenum Press, New York, N.Y., 1973, Chapter 5, and T.W. Greene, "Protective Groups in Organic Synthesis", John Wiley and Sons, New York, NY, 1981, Chapter 5. The term "protected carboxy" refers to a carboxy group substituted with one of the above carboxy- protecting groups. "Guanidine" means the group -NH-C(NH)-NHR in which R is hydrogen, alkyl, alkoxy, a cycloalkyl, a heterocyclyl, cycloalkyl -substituted alkyl or heterocyclyl-substituted alkyl wherein the alkyl, alkoxy, cycloalkyl and heterocyclyl are as defined herein. A particular guanidine is the group -NH-C(NH)-NH2. "Hydroxy-protecting group" as used herein refers to a derivative of the hydroxy group commonly employed to block or protect the hydroxy group while reactions are carried out on other functional groups on the compound. Examples of such protecting groups include tetrahydropyranyloxy, benzoyl, acetoxy, carbamoyloxy, benzyl, and silylethers (e.g. TBS, TBDPS) groups. Further examples of these groups are found in T. W. Greene and P. G. M. Wuts, "Protective Groups in Organic Synthesis", 2nd ed., John Wiley & Sons, Inc., New York, NY, 1991, chapters 2-3; E. Haslam, "Protective Groups in Organic Chemistry", J. G. W. McOmie, Ed., Plenum Press, New York, NY, 1973, Chapter 5, and T.W. Greene, "Protective Groups in Organic Synthesis", John Wiley and Sons, New York, NY, 1981. The term "protected hydroxy" refers to a hydroxy group substituted with one of the above hydroxy- protecting groups.
"Heterocyclic group", "heterocyclic", "heterocycle", "heterocyclyl", or "heterocyclo" alone, and when used as a moiety in a complex group such as a heterocycloalkyl group, are used interchangeably and refer to any mono-, bi-, tricyclic or spiro, saturated or unsaturated, aromatic (heteroaryl) or non-aromatic, ring system, having 3 to 20 ring atoms, where the ring atoms are carbon, and at least one atom in the ring or ring system is a heteroatom selected from nitrogen, sulfur or oxygen. In one example, heterocyclyl includes 3-12 ring atoms and includes monocycles, bicycles, tricycles and spiro ring systems, wherein the ring atoms are carbon, and at least one atom in the ring or ring system is a heteroatom selected from nitrogen, sulfur or oxygen. In one example, heterocyclyl includes 1 to 4 heteroatoms. In another example, heterocyclyl includes 3- to 7-membered monocycles having one or more heteroatoms selected from nitrogen, sulfur or oxygen. In another example, heterocyclyl includes 4- to 6-membered monocycles having one or more heteroatoms selected from nitrogen, sulfur or oxygen. In another example, heterocyclyl includes 3-membered monocycles. In another example, heterocyclyl includes 4-membered monocycles. In another example, heterocyclyl includes 5-6- membered monocycles. In one example, the heterocyclyl group includes 0 to 3 double bonds. Any nitrogen or sulfur heteroatom may optionally be oxidized (e.g. NO, SO, S02), and any nitrogen heteroatom may optionally be quaternized (e.g. [ΝΡ ]+0~, [NPv4]+OFf). Example heterocycles are oxiranyl, aziridinyl, thiiranyl, azetidinyl, oxetanyl, thietanyl, 1 ,2-dithietanyl, 1,3-dithietanyl, pyrrolidinyl, dihydro-lH-pyrrolyl, dihydrofuranyl, tetrahydrofuranyl, dihydrothienyl, tetrahydrothienyl, imidazolidinyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, 1,1-dioxo-thiomorpholinyl, dihydropyranyl, tetrahydropyranyl, hexahydrothiopyranyl, hexahydropyrimidinyl, oxazinanyl, thiazinanyl, thioxanyl, homopiperazinyl, homopiperidinyl, azepanyl, oxepanyl, thiepanyl, oxazepinyl, oxazepanyl, diazepanyl, 1 ,4-diazepanyl, diazepinyl, thiazepinyl, thiazepanyl, tetrahydrothiopyranyl, oxazolidinyl, thiazolidinyl, isothiazolidinyl, 1,1-dioxoisothiazolidinonyl, oxazolidinonyl, imidazolidinonyl, 4,5,6,7-tetrahydro[2H]indazolyl, tetrahydrobenzoimidazolyl, 4,5,6,7- tetrahydrobenzo[d]imidazolyl, 1 ,6-dihydroimidazol[4,5-d]pyrrolo[2,3-b]pyridinyl, thiazinyl, oxazinyl, thiadiazinyl, oxadiazinyl, dithiazinyl, dioxazinyl, oxathiazinyl, thiatriazinyl, oxatriazinyl, dithiadiazinyl, imidazolinyl, dihydropyrimidyl, tetrahydropyrimidyl, 1-pyrrolinyl, 2-pyrrolinyl, 3-pyrrolinyl, indolinyl, thiapyranyl, 2H-pyranyl, 4H-pyranyl, dioxanyl, 1,3- dioxolanyl, pyrazolinyl, pyrazolidinyl, dithianyl, dithiolanyl, pyrimidinonyl, pyrimidindionyl, pyrimidin-2,4-dionyl, piperazinonyl, piperazindionyl, pyrazolidinylimidazolinyl, 3- azabicyclo[3.1.0]hexanyl, 3,6-diazabicyclo[3.1.1]heptanyl, 6-azabicyclo[3.1.1]heptanyl, 3- azabicyclo[3.1.1]heptanyl, 3-azabicyclo[4.1.0]heptanyl, azabicyclo[2.2.2]hexanyl, 2- azabicyclo[3.2.1]octanyl, 8-azabicyclo[3.2.1]octanyl, 2-azabicyclo[2.2.2]octanyl, 8- azabicyclo[2.2.2]octanyl, 7-oxabicyclo[2.2. ljheptane, azaspiro[3.5]nonanyl, azaspiro [2.5]octanyl, azaspiro [4.5] decanyl, 1 -azaspiro [4.5] decan-2-only, azaspiro[5.5]undecanyl, tetrahydroindolyl, octahydroindolyl, tetrahydroisoindolyl, tetrahydroindazolyl, 1,1-dioxohexahydrothiopyranyl. Examples of 5-membered heterocycles containing a sulfur or oxygen atom and one to three nitrogen atoms are thiazolyl, including thiazol-2-yl and thiazol-2-yl N-oxide, thiadiazolyl, including l,3,4-thiadiazol-5-yl and 1,2,4- thiadiazol-5-yl, oxazolyl, for example oxazol-2-yl, and oxadiazolyl, such as l,3,4-oxadiazol-5-yl, and l,2,4-oxadiazol-5-yl. Example 5-membered ring heterocycles containing 2 to 4 nitrogen atoms include imidazolyl, such as imidazol-2-yl; triazolyl, such as l,3,4-triazol-5-yl; 1,2,3- triazol-5-yl, l,2,4-triazol-5-yl, and tetrazolyl, such as lH-tetrazol-5-yl. Example benzo-fused 5- membered heterocycles are benzoxazol-2-yl, benzthiazol-2-yl and benzimidazol-2-yl. Example 6-membered heterocycles contain one to three nitrogen atoms and optionally a sulfur or oxygen atom, for example pyridyl, such as pyrid-2-yl, pyrid-3-yl, and pyrid-4-yl; pyrimidyl, such as pyrimid-2-yl and pyrimid-4-yl; triazinyl, such as l,3,4-triazin-2-yl and l,3,5-triazin-4-yl; pyridazinyl, in particular pyridazin-3-yl, and pyrazinyl. The pyridine N-oxides and pyridazine N-oxides and the pyridyl, pyrimid-2-yl, pyrimid-4-yl, pyridazinyl and the l,3,4-triazin-2-yl groups, are other example heterocycle groups. Substituents for "optionally substituted heterocycles" include hydroxyl, alkyl, alkoxy, acyl, halogen, mercapto, oxo, carboxyl, halo- substituted alkyl, amino, cyano, nitro, amidino, guanidino.
"Heteroaryl" alone and when used as a moiety in a complex group such as a heteroaralkyl group, refers to any mono-, bi-, or tricyclic ring system where at least one ring is a 5- or, 6- membered aromatic ring containing from 1 to 4 heteroatoms selected from nitrogen, oxygen, and sulfur, and in an example embodiment, at least one heteroatom is nitrogen. See, for example, Lang's Handbook of Chemistry, supra. Included in the definition are any bicyclic groups where any of the above heteroaryl rings are fused to an aryl ring. In one embodiment, heteroaryl includes 4-6 membered monocyclic aromatic groups where one or more ring atoms is nitrogen, sulfur or oxygen. In another embodiment, heteroaryl includes 5-6 membered monocyclic aromatic groups where one or more ring atoms is nitrogen, sulfur or oxygen. Example heteroaryl groups (whether substituted or unsubstituted) include thienyl, furyl, imidazolyl, pyrazolyl, thiazolyl, isothiazolyl, oxazolyl, isoxazolyl, triazolyl, thiadiazolyl, oxadiazolyl, tetrazolyl, thiatriazolyl, oxatriazolyl, pyridyl, pyrimidyl, pyrazinyl, pyridazinyl, triazinyl, tetrazinyl, tetrazolo[l,5-b]pyridazinyl, imidazol[l,2-a]pyrimidinyl and purinyl, as well as benzo-fused derivatives, for example benzoxazolyl, benzofuryl, benzo thiazolyl, benzo thiadiazolyl, benzo triazolyl, benzoimidazolyl and indolyl. Additional examples of "heteroaryl" groups are: 1 ,3-thiazol-2-yl, 4-(carboxymethyl)-5 -methyl- 1 ,3-thiazol-2-yl, 4-(carboxymethyl)-5 -methyl- 1,3- thiazol-2-yl sodium salt, l,2,4-thiadiazol-5-yl, 3-methyl-l,2,4-thiadiazol-5-yl, l,3,4-triazol-5-yl, 2-methyl-l,3,4-triazol-5-yl, 2-hydroxy-l,3,4-triazol-5-yl, 2-carboxy-4-methyl-l,3,4-triazol-5-yl sodium salt, 2-carboxy-4-methyl-l,3,4-triazol-5-yl, l,3-oxazol-2-yl, l,3,4-oxadiazol-5-yl, 2- methyl- 1 ,3,4-oxadiazol-5-yl, 2-(hydroxymethyl)- 1 ,3,4-oxadiazol-5-yl, 1 ,2,4-oxadiazol-5-yl, l,3,4-thiadiazol-5-yl, 2-thiol-l,3,4-thiadiazol-5-yl, 2-(methylthio)-l,3,4-thiadiazol-5-yl, 2- amino-l,3,4-thiadiazol-5-yl, lH-tetrazol-5-yl, l-methyl-lH-tetrazol-5-yl, 1-(1- (dimethylamino)eth-2-yl)-lH-tetrazol-5-yl, l-(carboxymethyl)-lH-tetrazol-5-yl, 1-
(carboxymethyl)-lH-tetrazol-5-yl sodium salt, l-(methylsulfonic acid)-lH-tetrazol-5-yl, 1- (methylsulfonic acid)-lH-tetrazol-5-yl sodium salt, 2-methyl-lH-tetrazol-5-yl, l,2,3-triazol-5-yl, 1 -methyl- 1 ,2,3-triazol-5-yl, 2-methyl- 1 ,2,3-triazol-5-yl, 4-methyl- 1 ,2,3-triazol-5-yl, pyrid-2-yl N-oxide, 6-methoxy-2-(n-oxide)-pyridaz-3-yl, 6-hydroxypyridaz-3-yl, l-methylpyrid-2-yl, 1- methylpyrid-4-yl, 2-hydroxypyrimid-4-yl, 1 ,4,5,6-tetrahydro-5,6-dioxo-4-methyl-as-triazin-3-yl, l,4,5,6-tetrahydro-4-(formylmethyl)-5,6-dioxo-as-triazin-3-yl, 2,5-dihydro-5-oxo-6-hydroxy- astriazin-3-yl, 2,5-dihydro-5-oxo-6-hydroxy-as-triazin-3-yl sodium salt, 2,5-dihydro-5-oxo-6- hydroxy-2-methyl-astriazin-3-yl sodium salt, 2,5-dihydro-5-oxo-6-hydroxy-2-methyl-as-triazin- 3-yl, 2,5-dihydro-5-oxo-6-methoxy-2-methyl-as-triazin-3-yl, 2,5-dihydro-5-oxo-as-triazin-3-yl, 2,5-dihydro-5-oxo-2-methyl-as-triazin-3-yl, 2,5-dihydro-5-oxo-2,6-dimethyl-as-triazin-3-yl, tetrazolo[l,5-b]pyridazin-6-yl and 8-aminotetrazolo[l,5-b]-pyridazin-6-yl. Heteroaryl groups are optionally substituted as described for heterocycles.
In particular embodiments, a heterocyclyl group is attached at a carbon atom of the heterocyclyl group. By way of example, carbon bonded heterocyclyl groups include bonding arrangements at position 2, 3, 4, 5, or 6 of a pyridine ring, position 3, 4, 5, or 6 of a pyridazine, position 2, 4, 5, or 6 of a pyrimidine ring, position 2, 3, 5, or 6 of a pyrazine ring, position 2, 3, 4, or 5 of a furan, tetrahydroiuran, thiofuran, thiophene, pyrrole or tetrahydropyrrole ring, position 2, 4, or 5 of an oxazole, imidazole or thiazole ring, position 3, 4, or 5 of an isoxazole, pyrazole, or isothiazole ring, position 2 or 3 of an aziridine ring, position 2, 3, or 4 of an azetidine ring, position 2, 3, 4, 5, 6, 7, or 8 of a quinoline ring or position 1, 3, 4, 5, 6, 7, or 8 of an isoquinoline ring.
In certain embodiments, the heterocyclyl group is N-attached. By way of example, the nitrogen bonded heterocyclyl or heteroaryl group include bonding arrangements at position 1 of an aziridine, azetidine, pyrrole, pyrrolidine, 2-pyrroline, 3-pyrroline, imidazole, imidazolidine, 2- imidazoline, 3 -imidazoline, pyrazole, pyrazoline, 2-pyrazoline, 3-pyrazoline, piperidine, piperazine, indole, indoline, lH-indazole, position 2 of a isoindole, or isoindoline, position 4 of a morpholine, and position 9 of a carbazole, or β-carboline.
"Leaving group" refers to a portion of a first reactant in a chemical reaction that is displaced from the first reactant in the chemical reaction. Examples of leaving groups include, but are not limited to, halogen atoms, alkoxy and sulfonyloxy groups. Example sulfonyloxy groups include, but are not limited to, alkylsulfonyloxy groups (for example methyl sulfonyloxy (mesylate group) and trifluoromethylsulfonyloxy (triflate group)) and arylsulfonyloxy groups (for example /?-toluenesulfonyloxy (tosylate group) and /?-nitrosulfonyloxy (nosylate group)). "Optionally substituted" unless otherwise specified means that a group may be unsubstituted or substituted by one or more (e.g. 0, 1, 2, 3 or 4) of the substituents listed for that group in which said substituents may be the same or different. In an embodiment an optionally substituted group has 1 substituent. In another embodiment an optionally substituted group has 2 substituents. In another embodiment an optionally substituted group has 3 substituents. In certain embodiments, divalent groups are described generically without specific bonding configurations, for example in the group -CH2C(0)-. It is understood that the generic description is meant to include both bonding configurations, unless specified otherwise. For example, in the group R'-R^R3, if the group R2 is described as -CH2C(0)-, then it is understood that this group can be bonded both as R -CH2C(0)-R3, and as R -C(0)CH2-R3, unless specified otherwise. "Package insert" is used to refer to instructions customarily included in commercial packages of therapeutic products that contain information about the indications, usage, dosage, administration, contraindications and/or warnings concerning the use of such therapeutic products. "Pharmaceutically acceptable salts" include both acid and base addition salts.
"Pharmaceutically acceptable acid addition salt" refers to those salts which retain the biological effectiveness and properties of the free bases and which are not biologically or otherwise undesirable, formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, carbonic acid, phosphoric acid and the like, and organic acids may be selected from aliphatic, cycloaliphatic, aromatic, araliphatic, heterocyclic, carboxylic, and sulfonic classes of organic acids such as formic acid, acetic acid, propionic acid, glycolic acid, gluconic acid, lactic acid, pyruvic acid, oxalic acid, malic acid, maleic acid, maloneic acid, succinic acid, fumaric acid, tartaric acid, citric acid, aspartic acid, ascorbic acid, glutamic acid, anthranilic acid, benzoic acid, cinnamic acid, mandelic acid, embonic acid, phenylacetic acid, methanesulfonic acid, ethanesulfonic acid, benzenesulfonic acid, p-toluenesulfonic acid, salicyclic acid and the like.
"Pharmaceutically acceptable base addition salts" include those derived from inorganic bases such as sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum salts and the like. Particularly base addition salts are the ammonium, potassium, sodium, calcium and magnesium salts. Salts derived from pharmaceutically acceptable organic nontoxic bases includes salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, 2-diethylaminoethanol, tromethamine, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, ethylenediamine, glucosamine, methylglucamine, theobromine, purines, piperizine, piperidine, N-ethylpiperidine, polyamine resins and the like. Particularly organic non-toxic bases are isopropylamine, diethylamine, ethanolamine, tromethamine, dicyclohexylamine, choline, and caffeine.
A "sterile" formulation is aseptic or free from all living microorganisms and their spores. "Stereoisomers" refers to compounds which have identical chemical constitution, but differ with regard to the arrangement of the atoms or groups in space. Stereoisomers include diastereomers, enantiomers, conformers and the like. "Chiral" refers to molecules which have the property of non-superimposability of the mirror image partner, while the term "achiral" refers to molecules which are superimposable on their mirror image partner.
"Diastereomer" refers to a stereoisomer with two or more centers of chirality and whose molecules are not mirror images of one another. Diastereomers have different physical properties, e.g. melting points, boiling points, spectral properties or biological activities. Mixtures of diastereomers may separate under high resolution analytical procedures such as electrophoresis and chromatography such as HPLC.
"Enantiomers" refer to two stereoisomers of a compound which are non-superimposable mirror images of one another.
Stereochemical definitions and conventions used herein generally follow S. P. Parker, Ed., McGraw-Hill Dictionary of Chemical Terms (1984) McGraw-Hill Book Company, New York; and Eliel, E. and Wilen, S., "Stereochemistry of Organic Compounds", John Wiley & Sons, Inc., New York, 1994. Many organic compounds exist in optically active forms, i.e., they have the ability to rotate the plane of plane-polarized light. In describing an optically active compound, the prefixes D and L, or R and S, are used to denote the absolute configuration of the molecule about its chiral center(s). The prefixes d and 1 or (+) and (-) are employed to designate the sign of rotation of plane-polarized light by the compound, with (-) or 1 meaning that the compound is levorotatory. A compound prefixed with (+) or d is dextrorotatory. For a given chemical structure, these stereoisomers are identical except that they are mirror images of one another. A specific stereoisomer may also be referred to as an enantiomer, and a mixture of such isomers is often called an enantiomeric mixture. A 50:50 mixture of enantiomers is referred to as a racemic mixture or a racemate, which may occur where there has been no stereoselection or stereospecificity in a chemical reaction or process. The terms "racemic mixture" and "racemate" refer to an equimolar mixture of two enantiomeric species, devoid of optical activity.
The term "tautomer" or "tautomeric form" refers to structural isomers of different energies which are interconvertible via a low energy barrier. For example, proton tautomers (also known as prototropic tautomers) include interconversions via migration of a proton, such as keto-enol and imine-enamine isomerizations. Valence tautomers include interconversions by reorganization of some of the bonding electrons. A "solvate" refers to an association or complex of one or more solvent molecules and a compound of the present invention. Examples of solvents that form solvates include water, isopropanol, ethanol, methanol, DMSO, ethyl acetate, acetic acid, and ethanolamine. The term "hydrate" refers to the complex where the solvent molecule is water. A "subject," "individual," or "patient" is a vertebrate. In certain embodiments, the vertebrate is a mammal. Mammals include, but are not limited to, farm animals (such as cows), sport animals, pets (such as cats, dogs, and horses), primates, mice and rats. In certain embodiments, a mammal is a human.
"Therapeutically effective amount" means an amount of a compound of the present invention that (i) treats or prevents the particular disease, condition or disorder, (ii) attenuates, ameliorates or eliminates one or more symptoms of the particular disease, condition, or disorder, or (iii) prevents or delays the onset of one or more symptoms of the particular disease, condition or disorder described herein. In the case of cancer, the therapeutically effective amount of the drug may reduce the number of cancer cells; reduce the tumor size; inhibit (i.e., slow to some extent and preferably stop) cancer cell infiltration into peripheral organs; inhibit (i.e., slow to some extent and preferably stop) tumor metastasis; inhibit, to some extent, tumor growth; and/or relieve to some extent one or more of the symptoms associated with the cancer. To the extent the drug may prevent growth and/or kill existing cancer cells, it may be cytostatic and/or cytotoxic. For cancer therapy, efficacy can, for example, be measured by assessing the time to disease progression (TTP) and/or determining the response rate (RR). In the case of immunological disorders, the therapeutic effective amount is an amount sufficient to decrease or alleviate an allergic disorder, the symptoms of an autoimmune and/or inflammatory disease, or the symptoms of an acute inflammatory reaction (e.g. asthma). In some embodiments, a therapeutically effective amount is an amount of a chemical entity described herein sufficient to significantly decrease the activity or number of B-cells.
"Treatment" (and variations such as "treat" or "treating") refers to clinical intervention in an attempt to alter the natural course of the individual or cell being treated, and can be performed either for prophylaxis or during the course of clinical pathology. Desirable effects of treatment include preventing occurrence or recurrence of disease, alleviation of symptoms, diminishment of any direct or indirect pathological consequences of the disease, stabilized (i.e., not worsening) state of disease, preventing metastasis, decreasing the rate of disease progression, amelioration or palliation of the disease state, prolonging survival as compared to expected survival if not receiving treatment and remission or improved prognosis. In some embodiments, compounds of the invention are used to delay development of a disease or disorder or to slow the progression of a disease or disorder. Those in need of treatment include those already with the condition or disorder as well as those prone to have the condition or disorder, (for example, through a genetic mutation) or those in which the condition or disorder is to be prevented. The terms "compound(s) of this invention," and "compound(s) of the present invention", unless otherwise indicated, include compounds of formula I and stereoisomers, tautomers, solvates, metabolites, salts (e.g., pharmaceutically acceptable salts), and prodrugs thereof. Unless otherwise stated, structures depicted herein are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms. For example, compounds of formulas I, II and III, wherein one or more hydrogen atoms are replaced by deuterium or tritium, or one or more carbon atoms are replaced by 13C- or 14C-enriched carbon are within the scope of this invention.
INHIBITORS OF JAK1 KINASE
One aspect of the invention provides compounds of formula I:
Figure imgf000024_0001
stereoisomers, tautomers and pharmaceutically acceptable salts thereof, wherein
X is N or CR4; Y is N or CR5; R1 is C3-12 cycloalkyl, or 3-20 membered heterocyclyl, wherein R1 is independently optionally substituted by halogen, oxo, -CN, -ORa, -SRa, -NRaRb, Ci_3 -C(0)ORa, -S(0)2Ra, or Ci_6 alkyl optionally substituted by oxo, -CN, -S(0)2Ra, C6-14 aryl, or halogen;
R2 is absent, Ci_6 alkyl, C2-6 alkenyl, C2-6 alkynyl, -(Ci_6 alkylene)-, -(C2-6 alkenylene)-, -(C2-6 alkynylene)-, -(C0_6 alkylene)CN, -(C0-3 alkylene)NRa(C0_3 alkylene)-, -(C0-3 alkylene)0(Co_3 alkylene)-, -(C0-3 alkylene)C(O)(C0_3 alkylene)-, -(C0-3 alkylene)NRaC(O)(C0_3 alkylene)-, -(C0_3 alkylene)C(O)NRa(C0_3 alkylene)-, -(C0_3 alkylene)C(O)O(C0_3 alkylene)-, -(Co-3 alkylene)OC(0)(Co_3 alkylene)-, -(C0.3 alkylene)NRaC(O)NRb(C0-3 alkylene)-, -(C0.3 alkylene)OC(0)NRa(Co_3 alkylene)-, -(C0.3 alkylene)NRaC(O)O(C0_3 alkylene)-, -(C0.3 alkylene)S(0)i_2(Co_3 alkylene)-, -(C0-3 alkylene)NRaS(O)i_2(C0-3 alkylene)-, -(C0-3 alkylene)S(O)i_2NRa(C0-3 alkylene)- or -(C0_3 alkylene)NRaS(O)i_2NRb(C0-3 alkylene)-, wherein said alkyl, alkyenyl, alkynyl, alkylene, alkenylene and alkynylene are independently optionally substituted by halogen, oxo, -CN, -ORc, -SRC, -NRcRd or Ci_3 alkyl optionally substituted by halogen;
R3 is absent, hydrogen, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, C3_7 cycloalkyl, C6-14 aryl or 3-20 membered heterocyclyl, wherein R3 is independently optionally substituted by R6; R4 is hydrogen, halogen or Ci_3 alkyl;
R5 is halogen, C1-12 alkyl, C2-12 alkenyl, C2-12 alkynyl, -(C0-3 alkylene)CN, -(C0-3 alkylene)NRaRb, -(C0.3 alkylene)ORa, -(C0.3 alkylene)SRa, -(C0.3 alkylene)C(0)Ra, -(C0.3 alkylene)NRaC(0)Rb, -(C0.3 alkylene)C(0)NRaRb, -(C0.3 alkylene)C(0)ORa, -(C0.3
alkylene)OC(0)Ra, -(C0.3 alkylene)NRaC(0)NRaRb, -(C0.3 alkylene)OC(0)NRaRb, -(C0.3 alkylene)NRaC(0)ORb, -(C0.3 alkylene) S(0)i_2Ra, -(C0.3 alkylene)NRaS(0)i_2Rb, -(C0.3 alkylene)S(0)i_2NRaRb, -(C0.3 alkylene)NRaS(0)i_2NRaRb, -(C0.3 alkylene)C3_i2 cycloalkyl, -(C0-3 alkylene)C6_i4 aryl, -(C0-3 alkylene)3-12 membered heterocyclyl or -(C0-3
alkylene)C(0)3-12 membered heterocyclyl, wherein said C2_i2 alkyl, alkenyl, alkynyl, alkylene, cycloalkyl, aryl and heterocyclyl are independently optionally substituted by halogen, oxo, -(C0- 3 alkylene)CN, -(C0.3 alkylene)ORc, -(C0.3 alkylene)NRcRd, -(C0.3 alkylene)C(0)Rc, -(C0.3 alkylene)C(0)ORc, -(C0.3 alkylene)C(0)NRcRd, -(C0.3 alkylene)NRcC(0)Rd, -(C0.3
alkylene)OC(0)NRcRd, -(C0.3 alkylene)NRcC(0)NRcRd, -(C0.3 alkylene)NRcC(0)ORd, -(C0.3 alkylene)S(0)o_2Rc, -(C0.3 alkylene)NRcS(0)i_2Rd, -(C0.3 alkylene) S(0)i_2NRcRd, -(C0.3 alkylene)NRcS(0)i_2NRcRd or Ci_6 alkyl optionally substituted by oxo, -CN or halogen; and wherein said Ci alkyl is independently optionally substituted by halogen, oxo, -(C0-3
alkylene)CN, -(Ci_3 alkylene)ORc, -(C0.3 alkylene)NRcRd, -(C0.3 alkylene)C(0)Rc, -(C0.3 alkylene)C(0)ORc, -(C0.3 alkylene)C(0)NRcRd, -(C0.3 alkylene)NRcC(0)Rd, -(C0.3
alkylene)OC(0)NRcRd, -(C0.3 alkylene)NRcC(0)NRcRd, -(C0.3 alkylene)NRcC(0)ORd, -(C0.3 alkylene)S(0)o_2Rc, -(C0.3 alkylene)NRcS(0)i_2Rd, -(C0.3 alkylene) S(0)i_2NRcRd, -(C0.3 alkylene)NRcS(0)i_2NRcRd, -ORk, or Ci_6 alkyl optionally substituted by oxo, -CN or halogen;
R6 is independently oxo, halogen, -CN, -C(0)Ra, -C(0)ORa, -NRaC(0)Rb, -C(0)NRaRb, -NRaC(0)NRaRb, -OC(0)NRaRb, -NRaC(0)ORb, -S(0)i_2Ra, -NRaS(0)2Ra, -S(0)2NRaRb, -ORa, -SRa, -NRaRb, Ci_6 alkyl, C3_6 cycloalkyl, C2_6 alkenyl, C2_6 alkynyl, 3-7 membered heterocycly or C6-i4 aryl, and wherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl and aryl are independently optionally substituted by halogen, oxo, -CN, -ORc, -SRC, -NRcRd or Ci_6 alkyl optionally substituted by oxo or halogen; each Ra and Rb are independently hydrogen, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, -(Co_3 alkylene)C3_6 cycloalkyl, -(Co_3 alkylene)3-12 membered heterocyclyl, -(Co_3 alkylene)C(0)3-12 membered heterocyclyl or -(Co_3 alkylene)C6_i4 aryl, wherein said alkyl, cycloalkyl, heterocyclyl and aryl are independently optionally substituted by halogen, oxo, -CN, -ORe, -NReRf, -C(0)Rg, -C(0)ORg, -C(0)NRgRh, -NRgC(0)Rh, -OC(0)NRgRh, -NRgC(0)NRgRh, -NRgC(0)ORh, -S(0)i_2Rg, -NRgS(0)i_2Rh, -S(0)i_2NRgRh, -NRgS(0)i_2NRgRh, C3_6 cycloalkyl, 3-6 membered heterocyclyl, phenyl or Ci_3 alkyl optionally substituted by oxo or halogen, or taken together with the atom to which they are attached to form a 3-6 membered heterocyclyl optionally substituted by oxo, halogen, -C(0)Ci_6 alkyl or Ci_6 alkyl optionally substituted by oxo, halogen, ORg or NRgNRh; each Rc and Rd are independently hydrogen, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, -(Co_3 alkylene)C3_6 cycloalkyl, -(Co_3 alkylene)3-12 membered heterocyclyl, -(Co_3 alkylene)C(0)3-12 membered heterocyclyl or -(Co_3 alkylene)C6_i4 aryl, wherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl and aryl are independently optionally substituted by halogen, oxo, -CN, -ORg, -NRgRh, -C(0)Rg, -C(0)ORg, -C(0)NRgRh, -NRgC(0)Rh, -OC(0)NRgRh, -NRgC(0)NRgRh, -NRgC(0)ORh, -S(0)i_2Rg, -NRgS(0)i_2Rh, -S(0)i_2NRgRh, -NRgS(0)i_ 2NRgRh, C3_6 cycloalkyl, 3-6 membered heterocyclyl, phenyl or Ci_6 alkyl optionally substituted by oxo or halogen, or taken together with the atom to which they are attached to form a 3-6 membered heterocyclyl optionally substituted by oxo, halogen, -C(0)Ci_6 alkyl or Ci_6 alkyl optionally substituted by oxo or halogen; each Re, Rf, Rg, Rh are independently hydrogen or Ci_6 alkyl optionally substituted by halogen or oxo; and each Rk is independently hydrogen, C2_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, -(Co_3 alkylene)C3_6 cycloalkyl, -(Co_3 alkylene)3-12 membered heterocyclyl, -(Co_3 alkylene)C(0)3-12 membered heterocyclyl or -(Co_3 alkylene)C6_i4 aryl, wherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl and aryl are independently optionally substituted by halogen, oxo, -CN, -ORg, -NRgRh, -C(0)Rg, -C(0)ORg, -C(0)NRgRh, -NRgC(0)Rh, -OC(0)NRgRh,
-NRgC(0)NRgRh, -NRgC(0)ORh, -S(0)i_2Rg, -NRgS(0)i_2Rh, -S(0)i_2NRgRh, -NRgS(0)i_ 2NRgRh, C3-6 cycloalkyl, 3-6 membered heterocyclyl, phenyl or Ci_6 alkyl optionally substituted by oxo or halogen, or taken together with the atom to which they are attached to form a 3-6 membered heterocyclyl optionally substituted by oxo, halogen, -C(0)Ci_6 alkyl or Ci_6 alkyl optionally substituted by oxo or halogen. In certain embodiments, when R1 and R2 are absent, one of R3, R4 and R5 is other than hydrogen.
In certain embodiments, R1, R2 and R3 are not absent at the same time.
In certain embodiments, when R2 and R3 are absent, R5 is other than OH.
In one embodiment, X is CR4. In another embodiment, X is N. In one embodiment, Y is CR5. In another embodiment, Y is N.
In one embodiment, X is CR4 and Y is CR5.
In another embodiment, X is CR4 and Y is N.
In another embodiment, X is N and Y is CR5.
In another embodiment, X is N and Y is N. In one embodiment, R1 is absent. In one embodiment, R1 is absent with the proviso that
R1, R2 and R3 are not all absent at the same time.
In one embodiment R1 is a 3-20 membered heterocyclyl, wherein R1 is independently optionally substituted by halogen, oxo, -CN, -ORa, -SRa, -NRaRb, or Ci_6 alkyl optionally substituted by oxo, -CN or halogen. In one embodiment R1 is a 3-12 membered heterocyclyl optionally substituted by halogen, oxo, -CN, -ORa, -NRaRb, or Ci_6 alkyl optionally substituted by halogen. In one embodiment R1 is a 4-7 membered heterocyclyl optionally substituted by halogen, oxo, or Ci_6 alkyl optionally substituted by halogen, wherein said heterocyclyl is selected from oxetanyl, azetidinyl, thietanyl, tetrahydrofuranyl, 2,3-dihydrofuranyl, tetrahydrothienyl, 2,3-dihydrothienyl, pyrrolidinyl, 2,3-dihydro-lH-pyrrolyl, imidazolidinyl, 2H- pyranyl, tetrahydropyranyl, morpholinyl, piperazinyl, hexahydropyrimidinyl, oxazinanyl, thiazinanyl, piperidinyl, 8-azabicyclo[3.2.1]octanyl, 2-azabicyclo[2.2.2]octanyl, oxepanyl and azepanyl. In another embodiment, R1 is azetidinyl, pyrrolidinyl, imidazolidinyl, morpholinyl, piperazinyl, hexahydropyrimidinyl, or piperidinyl, wherein R1 is optionally substituted by halogen, oxo, or Ci_6 alkyl optionally substituted by halogen. In another embodiment, R1 is 4 , 5 , 6, 7-tetrahydrobenzo imidazo ly 1, 4,5,6 , 7-tetrahydro [2H] indazo ly 1, oxazo lidiny 1, thiazo lidiny 1, isothiazolidinyl, 1 , 1-dioxoisothiazolidinyl, oxazolidinonyl, 3-azabicyclo[3.1.0]hexanyl or imidazo lidinonyl, wherein R1 is independently optionally substituted by halogen, oxo, -CN, -ORa, -SRa, -NRaRb, or Ci_6 alkyl optionally substituted by oxo, -CN or halogen. In another embodiment, R1 is piperidinyl or tetrahydropyranyl wherein R1 is independently optionally substituted by halogen, oxo, -CN, -ORa, -SRa, -NRaRb, or Ci_6 alkyl optionally substituted by oxo, -CN or halogen. In one embodiment R1 is a 3-20 membered heterocyclyl, wherein R1 is independently optionally substituted by halogen, oxo, -CN, -ORa, -SRa, -NRaRb, or Ci_6 alkyl optionally substituted by oxo, -CN or halogen, with the proviso that R5 is other than hydrogen or -OH.
In another embodiment, R1 is a 3-12 membered heterocyclyl, wherein R1 is independently optionally substituted by halogen, oxo, -CN, -ORa, -SRa, -NRaRb, or Ci_6 alkyl optionally substituted by oxo, -CN or halogen. In another embodiment, R1 is a 3-12 membered heterocyclyl optionally substituted by halogen, oxo, -CN, -ORa, -NRaRb, or Ci_6 alkyl, wherein said heterocyclyl is selected from oxetanyl, azetidinyl, thietanyl, tetrahydrofuranyl, 2,3- dihydrofuranyl, tetrahydrothienyl, 2,3-dihydrothienyl, pyrrolidinyl, 2,3-dihydro-lH-pyrrolyl, imidazo lidiny 1, 2H-pyranyl, tetrahydropyranyl, morpholinyl, piperazinyl, hexahydropyrimidinyl, oxazinanyl, thiazinanyl, piperidinyl, 8-azabicyclo[3.2.1]octanyl, 2-azabicyclo[2.2.2]octanyl, oxepanyl, azepanyl, 7-oxabicyclo[2.2.1]heptane, octahydro-lH-indolyl, l-azaspiro[4.5]decanyl,
Figure imgf000028_0001
wherein the wav line re resents the point of attachment in formula I. In
embodiment, R1 represents the point of attachment
Figure imgf000028_0002
formula I. In one embo the wavy line represents the point of attachment in formula I.
In another embodiment, R1 is azetidinyl, pyrrolidinyl or piperidinyl, optionally substituted by 1 or 2 halogen, oxo, or Ci_6 alkyl optionally substituted by halogen. In another embodiment, R1 is azetidinyl, pyrrolidinyl or piperidinyl, optionally substituted by 1 or 2 halogen, oxo, -CN, -ORa, -NRaRb, or Ci_6 alkyl optionally substituted by halogen halogen, oxo, -CN, -ORa, -NRaRb, or Ci_6 alkyl optionally substituted by halogen
In another embodiment, R1 is morpholinyl, piperazinyl, 2-azabicyclo[2.2.2]octanyl, 8- azabicyclo[3.2.1]octanyl or piperidinyl, optionally substituted by 1 or 2 halogen, oxo, -CN, -ORa, -SRa, -NRaRb, or Ci_6 alkyl optionally substituted by oxo, -CN or halogen. In another embodiment, R1 is morpholinyl, piperazinyl, 2-azabicyclo[2.2.2]octanyl, 8- azabicyclo[3.2.1]octanyl or piperidinyl, optionally substituted by 1 or 2 halogen, oxo or Ci_6 alkyl optionally substituted by halogen.
In another embodiment, R1 is piperidinyl optionally substituted by halogen, oxo, -CN, -ORa, -SRa, -NRaRb, or Ci_6 alkyl optionally substituted by oxo, -CN or halogen. In another embodiment, R1 is piperidinyl optionally substituted by 1 or 2 halogen, oxo, -CN, -ORa, -NRaRb, or Ci_6 alkyl optionally substituted by halogen. In another embodiment, R1 is piperidinyl optionally substituted by 1 or 2 halogen, oxo or Ci_6 alkyl. In another embodiment, R1 is piperidinyl optionally substituted by methyl, oxo, fluoro or methoxy. In another
embodiment, R1 is piperidin-3-yl, piperidin-4-yl, 2-methylpiperidin-3-yl or 2-methylpiperidin-4- yl. In another embodiment, R1 is (R)-piperidin-3-yl. In another embodiment, R1 is (S)- piperidin-3-yl. In another embodiment, R1 is substituted (R)-piperidin-4-yl, wherein said piperidinyl is substituted by 1-3 groups selected from oxo, Ci_3 alkyl, halogen or -ORa. In another embodiment, R1 is substituted (S)-piperidin-4-yl, wherein said piperidinyl is substituted by 1-3 groups selected from oxo, Ci_3 alkyl, halogen or -ORa. In another embodiment, R1 is (R)- (R)-2-methylpiperidin-4-yl, (R)-(S)-2-methylpiperidin-4-yl, (S)-(R)-2-methylpiperidin-4-yl or (S)-(S)-2-methylpiperidin-4-yl. In another embodiment, R1 is (R)-(R)-3-fluoropiperidin-4-yl, (R)-(S)-3-fluoropiperidin-4-yl, (S)-(R)-3-fluoropiperidin-4-yl or (S)-(S)-3-fluoropiperidin-4-yl. In another embodiment, R1 is piperidinonyl, 2-methylpiperidin-4-yl, 3-methylpiperidin-4-yl, 4- methylpiperidin-4-yl, 2-fluoropiperidinyl, 3-fluoropiperidin-4-yl, 3,3-difluoropiperidin-4-yl, 3-
R2 R3
I
Ν,, methoxypiperidin-4-yl or , wherein the wavy line represents the point of attachment in formula I.
In another embodiment, R1 is piperidinyl optionally substituted by halogen, oxo, -CN, -ORa, -SRa, -NRaRb, or Ci_6 alkyl optionally substituted by oxo, -CN or halogen, with the proviso that R5 is other than hydrogen or -OH. In another embodiment, R1 is tetrahydropyranyl optionally substituted by halogen, oxo, -CN, -ORa, -SRa, -NRaRb, or Ci_6 alkyl optionally substituted by oxo, -CN or halogen, with the proviso that R5 is other than hydrogen or -OH. In another embodiment, R1 is tetrahydropyranyl optionally substituted by halogen, oxo, -CN, -ORa, -SRa, -NRaRb, or Ci_6 alkyl optionally substituted by oxo, -CN or halogen, and R2 and R3 are both absent, with the proviso that R5 is other than hydrogen or -OH.
In another embodiment, R1 is (R)-pyrrolidin-3-yl. In another embodiment, R1 is (S)- pyrrolidin-3-yl.
In one embodiment R1 is a C4_7 cycloalkyl, wherein R1 is independently optionally substituted by halogen, oxo, -CN, -ORa, -SRa, -NRaRb, or Ci_6 alkyl optionally substituted by oxo, -CN or halogen. In one embodiment R1 is a C4-7 cycloalkyl optionally substituted by halogen, oxo, -CN, -ORa, -NRaRb, or Ci_6 alkyl optionally substituted by halogen. In one embodiment R1 is a C4-7 cycloalkyl optionally substituted by halogen, oxo, or Ci_6 alkyl optionally substituted by halogen. In one embodiment, said cycloalkyl is cyclopropyl, eye lo butyl, cyclopentyl or cyclohexyl. In one embodiment, R1 is selected from
Figure imgf000030_0001
represents the point of attachment in formula I. In another embodiment, R1 is cyclohexyl optionally substituted by halogen, oxo, -CN,
-ORa, -SRa, -NRaRb, or Ci_6 alkyl optionally substituted by oxo, -CN or halogen. In another embodiment, R1 is cyclohexyl optionally substituted by halogen, oxo, -CN, -ORa, -NRaRb, or Ci_6 alkyl optionally substituted by halogen. In another embodiment, R1 is cyclohexyl optionally substituted by halogen, oxo, or Ci_6 alkyl optionally substituted by halogen. In another embodiment, R1 is cyclohexyl. In one embodiment, R1 is selected from cyclohexyl, 2- hydroxycyclohexyl, 3-hydroxycyclohexyl, 4-hydroxycyclohexyl, bicyclo[2.2.1]heptanyl, 2- methylcyclohexyl or 4,4-difluorocyclohexyl,
Figure imgf000031_0001
wherein R is optionally substituted by halogen, oxo, -CN, -ORa, -NRaR , or Ci_6 alkyl optionally substituted by halogen, and wherein the wavy line represents the point of attachment in formula I.
In one embodiment, R1 is selected from
Figure imgf000031_0002
and R2-R3 is selected from
Figure imgf000031_0003
wherein the wavy line represents the point of attachment in formula I.
In one embodiment, R1 is selected from r 2-*3 and
Figure imgf000031_0004
? wherein the wavy line represents the point of attachment in formula I.
In certain embodiments, R2 and R3 are absent, and R1 is selected from
Figure imgf000031_0005
? wherein R io is halogen, oxo, -CN, -ORa, -SRa, -NRaRb, or
Ci_6 alkyl optionally substituted by oxo, -CN or halogen, and wherein the wavy line represents the point of attachment in formula I. In another embodiment of this paragraph, R5 is other than hydrogen or -OH. In certain embodiments, R2 and R3 are absent, and R1 is selected from
Figure imgf000032_0001
, wherein R10 is -OH, -NH(CH2CF3), -CN, -CH2CN, -CH2CH2CN or halogen, and wherein the wavy line represents the point of attachment in formula I. In another embodiment of this paragraph, R5 is other than hydrogen or -OH. In another embodiment, R1 is cyclopentyl optionally substituted by halogen, oxo, -CN,
-ORa, -SRa, -NRaRb, or Ci_6 alkyl optionally substituted by oxo, -CN or halogen. In another embodiment, R1 is cyclopentyl optionally substituted by halogen, oxo, -CN, -ORa, -NRaRb, or Ci_6 alkyl optionally substituted by halogen. In another embodiment, R1 is cyclopentyl.
In one embodiment, R1 is C5_6 cycloalkyl or 5-6 membered heterocyclyl, wherein R1 is independently optionally substituted by halogen, oxo, -CN, -ORa, -SRa, -NRaRb, Ci_3 alkylene, -S(0)2Ra, or Ci_6 alkyl optionally substituted by oxo, -CN or halogen.
In one embodiment, R1 is cyclohexyl, cyclopentyl, piperidinyl, or tetrahydropyranyl, wherein R1 is independently optionally substituted by halogen, oxo, -CN, -ORa, -SRa, -NRaRb, Ci_3 alkylene, -S(0)2Ra, or Ci_6 alkyl optionally substituted by oxo, -CN or halogen. In one embodiment, R1 is selected from:
Figure imgf000032_0002
In one embodiment,-R1-R2-R3 taken together are selected from:
Figure imgf000033_0001
Figure imgf000034_0001
Figure imgf000034_0002
In one embodiment, R1 is C3_8 cycloalkyl, or 4-10 membered heterocyclyl, wherein R1 is independently optionally substituted by halogen, oxo, -CN, -ORa, -SRa, -NRaRb, Ci_3 alkylene, -C(0)ORa, -S(0)2Ra, or Ci_6 alkyl optionally substituted by oxo, -CN, -S(0)2Ra, C6_i4 aryl, or halogen.
In one embodiment, R1 is C5_6 cycloalkyl or 5-6 membered heterocyclyl, wherein R1 is independently optionally substituted by halogen, oxo, -CN, -ORa, -SRa, -NRaRb, Ci_3 alkylene, -C(0)ORa, -S(0)2Ra, or Ci_6 alkyl optionally substituted by oxo, -CN, -S(0)2Ra, C6_i4 aryl, or halogen.
In one embodiment, R1 is cyclohexyl, cyclopentyl, piperidinyl, or tetrahydropyranyl, wherein R1 is independently optionally substituted by halogen, oxo, -CN, -ORa, -SRa, -NRaRb, Ci_3 alkylene, -C(0)ORa, -S(0)2Ra, or Ci_6 alkyl optionally substituted by oxo, -CN, -S(0)2Ra, C6-14 aryl, or halogen.
In one embodiment, R2 is absent. In one embodiment, R2 is absent with the proviso that R1, R2 and R3 are not all absent at the same time. In one embodiment, R2 and R3 are absent. . In one embodiment, R2 and R3 are absent with the proviso that R1, R2 and R3 are not all absent at the same time.
In one embodiment, R2 is Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, wherein said alkyl, alkenyl or alkynyl are independently optionally substituted by halogen, oxo, -CN, -ORc, -SRC, -NRcRd or Ci_3 alkyl optionally substituted by halogen, and R3 is absent. In one embodiment, R2 is Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, wherein said alkyl, alkenyl or alkynyl are independently optionally substituted by halogen, oxo, -CN, -ORc, -NRcRd or Ci_3 alkyl optionally substituted by halogen, and R3 is absent. In one embodiment, R2 is selected from -CH2CF3 -CH2CH2CF3, -
CH2CH2F, -C(CH3)2OH, -CH2C(CH3)2OH, -CH2CH2OH, -CH2CH2OCH3
Figure imgf000035_0001
wherein the wavy line represents the point of attachment in formula I.
In one embodiment, R2 is -(Ci_6 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN, -ORc, -SRC, -NRcRd or Ci_3 alkyl optionally substituted by halogen. In one embodiment, R2 is -(Ci_6 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN, -ORc, -NRcRd or Ci_3 alkyl optionally substituted by halogen. In one embodiment, R2 is -(Ci_6 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN or Ci_3 alkyl. In another embodiment, R2 is methylene, ethylene, -CH(CH3)-, -C(CH3)2-, propylene or butylene, optionally substituted by halogen, oxo, -CN, -ORc, -NRcRd another embodiment, R2 is selected from methylene, ethylene,
-C(CH3)2- and
Figure imgf000035_0002
in the wavy line represents the point of attachment in formula I. In one embodiment, R2 is -(Co-6 alkylene)CN, wherein said alkylene is optionally substituted by halogen, oxo, -CN, -ORc, -SRC, -NRcRd or Ci_3 alkyl optionally substituted by halogen, and R3 is absent. In one embodiment, R2 is -(Ci_6 alkylene)CN, wherein said alkylene is optionally substituted by halogen, oxo, -CN, -ORc, -NRcRd or Ci_3 alkyl optionally substituted by halogen, and R3 is absent. In another embodiment, R2 is -CH2CN, -CH2CH2CN, -CH(CH3)CN or -CH(CH3)CH2CN and R3 is absent.
In another embodiment, R1 is a 3-20 membered heterocyclyl or C3_i2 cycloalkyl, wherein R1 is independently optionally substituted by halogen, oxo, -CN, -ORa, -SRa, -NRaRb, Ci_3 alkylene or Ci_6 alkyl optionally substituted by oxo, -CN or halogen, R2 is -CN, -CH2CN, -CH2CH2CN, -CH(CH3)CN or -CH(CH3)CH2CN, and R3 is absent, with the proviso that R5 is other than hydrogen or -OH.
In one embodiment, R2 is -(C0-3 alkylene)NRa(Co-3 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN, -ORc, -SRC, -NRcRd or Ci_3 alkyl optionally substituted by halogen. In one embodiment, R2 is -(C0-3 alkylene)NRa(Co_3 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN, -ORc, -NRcRd or Ci_3 alkyl optionally substituted by halogen. In one embodiment, R2 is -(C0-3 alkylene)NRa(Co_3 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN or Ci_3 alkyl. In another embodiment, R2 is - H-, -NHCH2- or -NHCH2CH2- In one embodiment, R2 is -(C0-3 alkylene)0(Co_3 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN, -ORc, -SRC, -NRcRd or Ci_3 alkyl optionally substituted by halogen. In one embodiment, R2 is -(C0-3 alkylene)0(Co_3 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN, -ORc, -NRcRd or Ci_3 alkyl optionally substituted by halogen. In one embodiment, R2 is -(C0-3 alkylene)0(Co_3 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN or Ci_3 alkyl. In another embodiment, R2 is -CH20- -CH2C(CH2)20- or -(CH2)20-
In one embodiment, R2 is -(Co_3 alkylene)NRaC(0)(Co_3 alkylene)- or -(Co_3 alkylene)C(0)NRa(Co_3 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN, -ORc, -SRC, -NRcRd or Ci_3 alkyl optionally substituted by halogen. In one embodiment, R2 is -(C0_3 alkylene)NRaC(O)(C0_3 alkylene)- or -(C0_3 alkylene)C(O)NRa(C0_3 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN, -ORc, -NRcRd or Ci_3 alkyl optionally substituted by halogen. In one embodiment, R2 is -(Co_3 alkylene)NRaC(0)(Co_3 alkylene)- or -(C0_3 alkylene)C(O)NRa(C0_3 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN or Ci_3 alkyl. In another embodiment, R2 is -C(0)NH- -CH2C(0)NH- or -CH2C(0)N(CH3)-. In another embodiment, R2 is -NHC(O)- or -NHC(0)CH2-.
In one embodiment R2 is -(C0-3 alkylene)OC(O)NRa(C0_3 alkylene)- or -(C0-3 alkylene)NRaC(0)0(Co_3 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN, -ORc, -SRC, -NRcRd or Ci_3 alkyl optionally substituted by halogen. In one embodiment R2 is -(C0-3 alkylene)OC(O)NRa(C0_3 alkylene)- or -(C0_3 alkylene)NRaC(O)O(C0_3 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN, -ORc, -NRcRd or Ci_3 alkyl optionally substituted by halogen. In one embodiment R2 is -(Co_3 alkylene)OC(0)NRa(Co-3 alkylene)- or -(C0_3 alkylene)NRaC(O)O(C0-3 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN or Ci_3 alkyl. In another embodiment, R2 is -NHC(0)0- -N(CH3)C(0)0-, -NHC(0)OCH2- or -NHC(0)OCH2CH2-.
In one embodiment R2 is -(Co_3 alkylene)C(0)(Co_3 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN, -ORc, -SRC, -NRcRd or Ci_3 alkyl optionally substituted by halogen. In one embodiment R2 is -(Co_3 alkylene)C(0)(Co_3 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN, -ORc, -NRcRd or Ci_3 alkyl optionally substituted by halogen. In one embodiment R2 is -(Co_3 alkylene)C(0)(Co_3 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN or Ci_3 alkyl. In another embodiment, R2 is selected from:
Figure imgf000037_0001
wherein the wavy lines represent points of attachment.
In one embodiment R2 is -(Co_3 alkylene)C(0)0(Co_3 alkylene)- or -(Co_3 alkylene)OC(0)(Co_3 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN, -ORc, -SRC, -NRcRd or Ci_3 alkyl optionally substituted by halogen. In one embodiment R2 is -(Co-3 alkylene)C(0)0(Co_3 alkylene)- or -(Co_3 alkylene)OC(0)(Co_3 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN, -ORc, -NRcRd or Ci_3 alkyl optionally substituted by halogen. In one embodiment R2 is -(Co_3 alkylene)C(0)0(Co_3 alkylene)- or -(Co_3 alkylene)OC(0)(Co_3 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN or Ci_3 alkyl. In another embodiment, R2 is selected from -C(0)0-
In one embodiment R2 is -(Co_3 alkylene)S(0)i_2(Co_3 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN, -ORc, -SRC, -NRcRd or Ci_3 alkyl optionally substituted by halogen. In one embodiment R2 is -(Co_3 alkylene)S(0)i_2(Co_3 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN, -ORc, -NRcRd or Ci_3 alkyl optionally substituted by halogen. In one embodiment R2 is -(Co_3 alkylene)S(0)i_2(Co_3 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN or Ci_3 alkyl. In another embodiment, R2 is selected from -C(0)CH2S(0)2,
Figure imgf000038_0001
wherein the wavy lines represent points of attachment. In one embodiment, R2 is -(Co_3 alkylene)NRaS(0)i_2(Co-3 alkylene)- or -(Co_3 alkylene)S(0)i_2NRa(Co-3 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN, -ORc, -SRC, -NRcRd or Ci_3 alkyl optionally substituted by halogen. In one embodiment, R2 is -(C0-3 alkylene)NRaS(O)i_2(C0-3 alkylene)- or -(C0_3 alkylene)S(O)i_2NRa(C0_ 3 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN, -ORc, -NRcRd or Ci_3 alkyl optionally substituted by halogen. In one embodiment, R2 is -(C0-3 alkylene)NRaS(0)i_2(Co_3 alkylene)- or -(C0-3 alkylene)S(O)i_2NRa(C0-3 alkylene)-, wherein said alkylene is optionally substituted by halogen, oxo, -CN or Ci_3 alkyl. In another embodiment, R2 is -NHS(0)2- -N(CH3)S(0)2- or -NHS(0)2CH2-.
In one embodiment, R2 is selected from absent, -NHS(0)2-, -N(CH3)S(0)2- , -NHS(0)2CH2- -C(0)CH2S(0)2, "C(0)0-, -NHC(0)0-, -N(CH3)C(0)0-, -NHC(0)OCH2-, -NHC(0)OCH2CH2-, -C(0)NH-, -CH2C(0)NH-, -CH2C(0)N(CH3)-, -NHC(O)-, -NHC(0)CH2-, -CH20-, -CH2C(CH2)20-, -(CH2)20- -NH-, -NHCH2-, -NHCH2CH2-, -CH2CN, -CH2CH2CN, -CH(CH3)CN , -CH(CH3)CH2CN, methylene, ethylene, -C(CH3)2- -CH2CF3, -CH2CH2CF3, -CH2CH2F, -CH2C(CH3)2OH, -CH2CH2OH, - CH2CH2OCH3,
Figure imgf000039_0001
Figure imgf000039_0002
, wherein the wavy line represents the point of attachment in formula
In one embodiment, R2 is absent, methylene, ethylene, -CH(CH3)-, -NH-, - -,
-C(0)0-, -C(0)NH- -NHC(0)0-, -CH2C(0)N(CH3)-, -NHS(0)2
Figure imgf000039_0003
^ ^ , ° , '"^ v , '"^ v ^ ,„or '""^ ^ ^ , wherein the wavy line represents the point of attachment in formula I.
In one embodiment, R 2 is absent, methylene, ethylene,
Figure imgf000039_0004
? J Ol 0r
Figure imgf000039_0005
wherein the wavy line represents the point of attachment in formula I.
In one embodiment, R3 is absent.
In one embodiment, R3 is hydrogen.
In one embodiment, -R2-R3 is -CHO.
In one embodiment, R2 is absent and R3 is hydrogen.
In one embodiment, R1 and R2 are absent. In one embodiment, R3 is Ci_6 alkyl optionally substituted by 1 to 3 R6. In another embodiment, R3 is Ci_6 alkyl optionally substituted by 1 to 3 oxo, Ci_6 alkyl, halogen, -CN, -S(0)i_2(Ci_6 alkyl), -ORa, -SRa or -NRaRb. In another embodiment, R3 is Ci_6 alkyl optionally substituted by 1 to 3 oxo, Ci_6 alkyl, halogen, -CN, -S(0)2(Ci_6 alkyl), -ORa or -NRaRb. In another embodiment, R3 is methyl, ethyl, propyl, isopropyl, butyl, isobutyl or t-butyl optionally substituted by oxo, Ci_6 alkyl, halogen, -CN, -S(0)2(Ci_6 alkyl), -ORa or -NRaRb. In another embodiment, R3 is selected from methyl, ethyl, n-butyl, sec-butyl, t-butyl, -CF3, -CH2CF3, -CH2CH2F, -CH2CH2CF3, -CH2OCH3, -CH2CH2OCH3, -CH(CH2CH3)CH2OCH3, -CH(CH3)CH2CH2OH, -CH2C(CH3)2OH, -CH2C(CF3)2OH, -CH2CH2OH, -C(CH3)2OH, -CH2CN, -(CH2)2CN, -(CH2)3CN, -CH(CH3)CH2CN, -C(CH3)2CN, -CH(CH3)CN, -CH2NH2, -CH(CH3)N(CH3)2 and-CH2CH2N(CH3)2.
In one embodiment, R3 is C3_7 cycloalkyl optionally substituted by 1 to 3 R6. In one embodiment, R3 is C3_7 cycloalkyl optionally substituted by 1 to 3 oxo, halogen, -CN, -S(0)i_ 2(Ci_6 alkyl), -ORa, -SRa, -NRaRb or Ci_6 alkyl optionally substituted by oxo or halogen. In one embodiment, R3 is C3_7 cycloalkyl optionally substituted by 1 to 3 oxo, halogen, -CN, -S(0)2(Ci_6 alkyl), -ORa, -NRaRb or Ci_6 alkyl optionally substituted by halogen. In another embodiment, R3 is cyclopropyl optionally substituted by 1 to 3 oxo, halogen, -CN, -S(0)2(Ci_6 alkyl), -ORa, -NRaRb or Ci_6 alkyl optionally substituted by halogen. In another embodiment, R3 is selected from cyclopropyl, 1-cyanocycloprop-l-yl, 1-trifluoromethylcycloprop-l-yl, 1- methylcycloprop-l-yl, 2-fluorocyclopyrop-l-yl, 2,2-dimethylcycloprop-l-yl, 2- cyanocyclopropyl, eye lo butyl, 4-carboxyclo butyl, 1-cyanocyclobut-l-yl, 4-aminocyclo butyl, cyclopentyl, 3-aminocyclohexyl, 4-aminocyclohexyl, 2-hydroxycyclohexyl, 3- hydroxycyclohexyl, 4-hydroxycyclohexyl and 2-hydroxycyclohexyl.
In one embodiment, R3 is C6-i4 aryl optionally substituted by 1 to 3 R6. In one embodiment, R3 is C6-i4 aryl optionally substituted by 1 to 3 Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, halogen, -CN, -S(0)i_2(Ci_6 alkyl), -ORa, -SRa or -NRaRb. In one embodiment, R3 is phenyl optionally substituted by 1 to 3 R6. In one embodiment, R3 is phenyl optionally substituted by 1 to 3 Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, halogen, -CN, -S(0)2(Ci_6 alkyl), -ORa or -NRaRb. In another embodiment, R3 is phenyl, 2-chloro-4-cyanophenyl, 2-cyanophenyl, 3-cyanophenyl, 4-cyanophenyl, 3-methylsulfonylphenyl, 3 -fluorophenyl or 4-methoxyphenyl.
In one embodiment, R3 is 5-6 membered heteroaryl optionally substituted by 1 to 3 R6. In one embodiment, R3 is 5-6 membered heteroaryl optionally substituted by 1 to 3 oxo, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, halogen, -CN, -S(0)i_2(Ci_6 alkyl), -ORa, -SRa or -NRaRb. In one embodiment, R3 is 5-6 membered heteroaryl optionally substituted by 1 to 3 oxo, Ci_6 alkyl, C2-6 alkenyl, C2_6 alkynyl, halogen, -CN, -S(0)2(Ci_6 alkyl), -ORa or -NRaRb. In one embodiment, R3 is pyridinyl, thiazolyl, pyrimidinyl, pyrazinyl, oxazolyl, pyrazolyl, imidazolyl, optionally substituted by 1 to 3 oxo, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, halogen, -CN, -S(0)2(Ci_6 alkyl), -ORa or -NRaRb. In one embodiment, R3 is selected from pyridinyl, pyridin- 3-yl, 6-cyanopyridinyl, 6-trifluoromethylpyridinyl, 2-cyanopyridin-4-yl, 4-cyanopyridin-2-yl, 5- cyanopyridin-2-yl, 3-fluoropyridin-5-yl, thiazol-5-yl, pyrimidin-2-yl, pyrimidin-4-yl, pyrimidin- 5-yl, pyrazin-2-yl, oxazol-2-yl, oxazol-4-yl, l-methylpyrazol-5-yl, l-methylpyrazo -
methylimidazo 1-2-yl
Figure imgf000041_0001
\ , wherein the wavy line represents the point of attachment in formula I. In one embodiment, R3 is selected from thiazol-5-yl and isothiazol-5-yl.
In one embodiment, R3 is 3-12 membered heterocyclyl optionally substituted by 1 to 3 R6. In one embodiment, R3 is 4-7 membered heterocyclyl optionally substituted by 1 to 3 R6. In one embodiment, R3 is 4-7 membered heterocyclyl optionally substituted by 1 to 3 oxo, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, halogen, -CN, -S(0)i_2Ra, -C(0)ORa, -ORa, -SRa or -NRaRb, wherein said alkyl, alkenyl and alkynyl are optionally substituted by oxo, halogen, -CN, -ORc or -NRcRd In one embodiment, R3 is 4-7 membered heterocyclyl optionally substituted by 1 to 3 oxo, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, halogen, -CN, -S(0)2Ra, -C(0)ORa, -ORa or -NRaRb, wherein said alkyl, alkenyl and alkynyl are optionally substituted by oxo, halogen, -CN, -ORc or -NRcRd In one embodiment, R3 is oxetanyl, tetrahydrofuranyl, pyrrolidinyl, piperidinyl, piperazinyl, pyranyl, tetrahydropyranyl, morpholinyl optionally substituted by 1 to 3 oxo, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, halogen, -CN, -S(0)2Ra, -C(0)ORa, -ORa or -NRaRb, wherein said alkyl, alkenyl and alkynyl are optionally substituted by oxo, halogen, -CN, -ORc or -NRcRd. In one embodiment, R3 is selected from oxetan-3-yl, piperidin-3-yl, piperidin-4-yl, N-methylpiperidin-2-yl, N-methylmorpholin-2-yl, 1- methylpyrrolidin-2-yl, pyrrolidinyl, pyrrolidinonyl, piperidinonyl, 3,3-difluoropyrrolidin-2-yl, 1- isopropylpyrrolidin-2-yl, 2-methylpyrrolidin-2-yl, l-methylcyanopyrrolidin-2-yl, 1-
cyclobutylpyrrolidin-2-yl, morpholinyl, pyran-4-yl, N-methylpiperazinyl,
Figure imgf000041_0002
,
Figure imgf000042_0001
wherein the wavy line represents the point of attachment in formula I. In one embodiment, R3 is (S)-l-methylpyrrolidin-2-yl. In one embodiment, R3 is selected from N-ethylpiperidin-2-yl, N-
(2-methoxyethyl)piperidin-2-yl, N-methylazepan-2-yl,
Figure imgf000042_0002
, wherein the wavy line represents the point of attachment in formula I.
In one embodiment, R is absent, hydrogen, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, t-butyl, cyclopropyl, phenyl, pyridinyl, thiazolyl, pyrimidinyl, pyrazinyl, oxazolyl, pyrazolyl, imidazolyl, oxetanyl, pyrrolidinyl, piperidinyl, pyranyl or morpholinyl, optionally substituted by oxo, Ci_6 alkyl, halogen, -CN, -S(0)2(Ci_6 alkyl), -ORa or -NRaRb.
In one embodiment, R3 is absent, hydrogen, methyl, -CF3, -CH2CN, -(CH2)2CN, 1- cyanocycloprop-l-yl, cyclopropyl, phenyl, 3-cyanophenyl, 4-cyanophenyl, 3- methylsulfonylphenyl, 3 -fluorophenyl, 6-cyanopyridinyl, 4-cyanopyridin-2-yl, pyridin-3-yl, pyrazin-2-yl, pyrimidin-5-yl, thiazol-5-yl or oxazol-4-yl. In one embodiment, R is selected from absent, hydrogen, methyl, ethyl, n-butyl, sec- butyl, t-butyl, -CF3, -CH2CF3, -CH2CH2F, -CH2CH2CF3, -CH2OCH3, -CH2CH2OCH3, -CH(CH2CH3)CH2OCH3, -CH(CH3)CH2CH2OH, -CH2C(CH3)2OH, -CH2C(CF3)2OH, - CH2CH2OH, -C(CH3)2OH, -CH2CN, -(CH2)2CN, -(CH2)3CN, -CH(CH3)CH2CN, - C(CH3)2CN, -CH(CH3)CN, -CH2NH2, -CH(CH3)N(CH3)2, -CH2CH2N(CH3)2, cyclopropyl, 1- cyanocycloprop-l-yl, 1-trifluoromethylcycloprop-l-yl, 1-methylcycloprop-l-yl, 2- fluorocyclopyrop-l-yl, 2,2-dimethylcycloprop-l-yl, 2-cyanocyclopropyl, eye lo butyl, 4- carboxyclo butyl, 1-cyanocyclobut-l-yl, 4-aminocyclo butyl, cyclopentyl, 3-aminocyclohexyl, 4- aminocyclohexyl, 2-hydroxycyclohexyl, 3-hydroxycyclohexyl, 4-hydroxycyclohexyl, 2- hydroxycyclohexyl, phenyl, 2-chloro-4-cyanophenyl, 2-cyanophenyl, 3-cyanophenyl, 4- cyanophenyl, 3-methylsulfonylphenyl, 3 -fluorophenyl, 4-methoxyphenyl, pyridinyl, pyridin-3- yl, 6-cyanopyridinyl, 6-trifluoromethylpyridinyl, 2-cyanopyridin-4-yl, 4-cyanopyridin-2-yl, 5- cyanopyridin-2-yl, 3-fluoropyridin-5-yl, thiazol-5-yl, pyrimidin-2-yl, pyrimidin-4-yl, pyrimidin- -yl, pyrazin-2-yl, oxazol-2-yl, oxazol-4-yl, l-methylpyrazol-5-yl, l-methylpyrazol-4-yl, 1-
Figure imgf000043_0001
methylmorpholin-2-yl, l-methylpyrrolidin-2-yl, pyrrolidinyl, pyrrolidinonyl, piperidinonyl, 3,3- difluoropyrrolidin-2-yl, l-isopropylpyrrolidin-2-yl, 2-methylpyrrolidin-2-yl, 1- methylcyanopyrrolidin-2-yl, l-cyclobutylpyrrolidin-2-yl, morpholinyl, pyran-4-yl, N-
Figure imgf000043_0002
, wherein the wavy line represents the point of attachment in formula I.
In one embodiment, R4 is hydrogen, methyl or F. In another embodiment, R4 is hydrogen.
In certain embodiments, R5 is halogen, C1-12 alkyl, C2-12 alkenyl, C2-12 alkynyl, -(C0_6 alkylene)CN, -(C0.3 alkylene)NRaRb, -(C0.3 alkylene)ORa, -(C0.3 alkylene)SRa, -(C0.3 alkylene)C(0)Ra, -(C0.3 alkylene)NRaC(0)Rb, -(C0.3 alkylene)C(0)NRaRb, -(C0.3 alkylene)C(0)ORa, -(C0.3 alkylene)OC(0)Ra, -(C0.3 alkylene)NRaC(0)NRaRb, -(C0.3 alkylene)OC(0)NRaRb, -(C0.3 alkylene)NRaC(0)ORb, -(C0.3 alkylene)S(0)i_2Ra, -(C0.3 alkylene)NRaS(0)i_2Rb, -(C0.3 alkylene)S(0)i_2NRaRb, -(C0.3 alkylene)NRaS(0)i_2NRaRb, -(C0.3 alkylene)C3-6 cycloalkyl, -(C0-3 alkylene)C6_i4 aryl, -(C0-3 alkylene)3-12 membered heterocyclyl or -(Co-3 alkylene)C(0)3-12 membered heterocyclyl, wherein said alkyl, alkenyl, alkynyl, alkylene, cycloalkyl, aryl and heterocyclyl are independently optionally substituted by halogen, oxo, -(Co-3 alkylene)CN, -(C0.3 alkylene)ORc, -(C0.3 alkylene)NRcRd, -(C0.3 alkylene)C(0)Rc, -(Co-3 alkylene)C(0)ORc, -(C0.3 alkylene)C(0)NRcRd, -(C0.3 alkylene)NRcC(0)Rd, -(C0.3 alkylene)OC(0)NRcRd, -(C0.3 alkylene)NRcC(0)NRcRd, -(C0.3 alkylene)NRcC(0)ORd, -(C0.3 alkylene)S(0)o-2Rc, -(C0.3 alkylene)NRcS(0)i_2Rd, -(C0.3 alkylene)S(0)i_2NRcRd, -(C0.3 alkylene)NRcS(0)i_2NRcRd or Ci_6 alkyl optionally substituted by oxo, -CN or halogen, with the proviso that R5 is other than OH.
In certain embodiments, R5 is halogen, C1-12 alkyl, C2_i2 alkenyl, C2_i2 alkynyl, -(C0_6 alkylene)CN, -(C0.3 alkylene)NRaRb, -(C0.3 alkylene)ORa, -(C0.3 alkylene)SRa, -(C0.3 alkylene)C(0)Ra, -(C0.3 alkylene)NRaC(0)Rb, -(C0.3 alkylene)C(0)NRaRb, -(C0.3 alkylene)C(0)ORa, -(C0.3 alkylene)OC(0)Ra, -(C0.3 alkylene)NRaC(0)NRaRb, -(C0.3 alkylene)OC(0)NRaRb, -(C0.3 alkylene)NRaC(0)ORb, -(C0.3 alkylene)S(0)i_2Ra, -(C0.3 alkylene)NRaS(0)i_2Rb, -(C0.3 alkylene)S(0)i_2NRaRb, -(C0.3 alkylene)NRaS(0)i_2NRaRb, -(C0.3 alkylene)C3_6 cycloalkyl, -(C0-3 alkylene)C6_i4 aryl, -(C0-3 alkylene)3-12 membered heterocyclyl or -(Co-3 alkylene)C(0)3-12 membered heterocyclyl, wherein said alkyl, alkenyl, alkynyl, alkylene, cycloalkyl, aryl and heterocyclyl are independently optionally substituted by halogen, oxo, -(Co-3 alkylene)CN, -(C0.3 alkylene)ORc, -(C0.3 alkylene)NRcRd, -(C0.3 alkylene)C(0)Rc, -(Co-3 alkylene)C(0)ORc, -(C0.3 alkylene)C(0)NRcRd, -(C0.3 alkylene)NRcC(0)Rd, -(C0.3 alkylene)OC(0)NRcRd, -(C0.3 alkylene)NRcC(0)NRcRd, -(C0.3 alkylene)NRcC(0)ORd, -(C0.3 alkylene)S(O)0-2Rc, -(C0.3 alkylene)NRcS(0)i-2Rd, -(C0.3 alkylene) S(0)i_2NRcRd, -(C0.3 alkylene)NRcS(0)i_2NRcRd or Ci_6 alkyl optionally substituted by oxo, -CN or halogen, with the proviso that R5 is other than OH.
In one embodiment, R5 is halogen. In one embodiment, R5 is F. In one embodiment, R5 is C1-12 alkyl, C2_i2 alkenyl, C2_i2 alkynyl, wherein said alkyl, alkenyl and alkynyl are independently optionally substituted by halogen, oxo, -(G 0-3 alkylene)CN, -(C0.3 alkylene)ORc, -(C0.3 alkylene)NRcRd, -(C0.3 alkylene)C(0)Rc, -(C0.3 alkylene)C(0)ORc, -(C0.3 alkylene)C(0)NRcRd, -(C0.3 alkylene)NRcC(0)Rd, -(C0.3 alkylene)OC(0)NRcRd, -(C0.3 alkylene)NRcC(0)NRcRd, -(C0.3 alkylene)NRcC(0)ORd, -(C0.3 alkylene)S(O)0-2Rc, -(C0.3 alkylene)NRcS(0)i-2Rd, -(C0.3 alkylene) S(0)i_2NRcRd, -(C0.3 alkylene)NRcS(0)i_2NRcRd or Ci_6 alkyl optionally substituted by oxo, -CN or halogen. In one embodiment, R5 is Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, wherein said alkyl, alkenyl and alkynyl are independently optionally substituted by halogen, oxo, -CN, -ORc, -NRcRd, -C(0)ORc, or Ci_6 alkyl optionally substituted by oxo, -CN or halogen. In one embodiment, R5 is selected from methyl, ethyl, 1 -hydro xyethyl, 2-hydroxyethyl, propyl, isopropyl, butyl, 2-methylbutyl, 3,3-difluorobut-l-yl, isobutyl, -CH2F, -CHF2, -CF3, -CH2OH, -C(CH3)2OH,
HO, HO,,.
wherein the wavy line represents the point of attachment in formula I. In one embodiment, R5 is C1-12 alkyl independently optionally substituted by halogen, oxo, -(Co-3 alkylene)CN, -(C0.3 alkylene)ORc, -(C0.3 alkylene)NRcRd, -(C0.3 alkylene)C(0)Rc, -(Co-3 alkylene)C(0)ORc, -(C0.3 alkylene)C(0)NRcRd, -(C0.3 alkylene)NRcC(0)Rd, -(C0.3 alkylene)OC(0)NRcRd, -(C0.3 alkylene)NRcC(0)NRcRd, -(C0.3 alkylene)NRcC(0)ORd, -(C0.3 alkylene)S(0)o-2Rc, -(C0.3 alkylene)NRcS(0)i_2Rd, -(C0.3 alkylene) S(0)i_2NRcRd, -(C0.3 alkylene)NRcS(0)i_2NRcRd or Ci_6 alkyl optionally substituted by oxo, -CN or halogen. In one embodiment, R5 is Ci_6 alkyl optionally substituted by halogen, oxo, -CN, -ORa or -NRaRb. In another embodiment, R5 is methyl, ethyl, propyl, isopropyl or 2-methylpropyl, optionally substituted by halogen, oxo, -CN, -ORa or -NRaRb, wherein Ra and Rb are independently hydrogen, Ci_6 alkyl, C3_6 cycloalkyl, 4-6 membered heterocyclyl or taken together with the atom to which they are attached to form a pyrrolidinyl, piperidinyl, piperazinyl or morpholinyl group. In another embodiment, R5 is methyl, hydro xymethyl, 1 -hydro xyethyl, 2-hydroxyethyl, isopropyl or 2-methylpropyl.
In one embodiment, R5 is -(C0-3 alkylene)CN, wherein said alkylene is independently optionally substituted by halogen, oxo, -(C0-3 alkylene)CN, -(C0-3 alkylene)ORc, -(C0-3 alkylene)NRcRd, -(C0.3 alkylene)C(0)Rc, -(C0.3 alkylene)C(0)ORc, -(C0.3 alkylene)C(0)NRcRd, -(Co-3 alkylene)NRcC(0)Rd, -(C0.3 alkylene)OC(0)NRcRd, -(C0.3 alkylene)NRcC(0)NRcRd, -(Co-3 alkylene)NRcC(0)ORd, -(C0.3 alkylene) S(O)0-2Rc, -(C0.3 alkylene)NRcS(0)i-2Rd, -(C0.3 alkylene)S(0)i_2NRcRd, -(C0.3 alkylene)NRcS(0)i_2NRcRd or Ci_6 alkyl optionally substituted by oxo, -CN or halogen. In one embodiment, R5 is -(C0-3 alkylene)CN, wherein said alkylene is optionally substituted by halogen, oxo, -CN, -ORc, -NRcRd, -C(0)ORc, or Ci_6 alkyl optionally substituted by oxo, -CN or halogen. In one embodiment, R5 is -(Ci_6 alkylene)CN, wherein said alkylene is optionally substituted by halogen, oxo, -CN, -ORc, -NRcRd, -C(0)ORc, or Ci_6 alkyl optionally substituted by oxo, -CN or halogen. In one embodiment, R5 is selected from - CN, -C(CH3)2CN.
In one embodiment, R5 is -CN.
In one embodiment, R5 is -(C0-3 alkylene)ORa or -(C0-3 alkylene)SRa, wherein said alkylene is independently optionally substituted by halogen, oxo, -(C0-3 alkylene)CN, -(C0-3 alkylene)ORc, -(C0.3 alkylene)NRcRd, -(C0.3 alkylene)C(0)Rc, -(C0.3 alkylene)C(0)ORc, -(C0.3 alkylene)C(0)NRcRd, -(C0.3 alkylene)NRcC(0)Rd, -(C0.3 alkylene)OC(0)NRcRd, -(C0.3 alkylene)NRcC(0)NRcRd, -(C0.3 alkylene)NRcC(0)ORd, -(C0.3 alkylene) S(O)0_2Rc, -(C0.3 alkylene)NRcS(0)i_2Rd, -(C0.3 alkylene) S(0)i_2NRcRd, -(C0.3 alkylene)NRcS(0)i_2NRcRd or Ci_6 alkyl optionally substituted by oxo, -CN or halogen. In one embodiment, R5 is -(C0-3 alkylene)ORa, wherein said alkylene is independently optionally substituted by halogen, oxo, -(Co-3 alkylene)CN, -(C0.3 alkylene)ORc, -(C0.3 alkylene)NRcRd, -(C0.3 alkylene)C(0)ORc, -(Co-3 alkylene)C(0)NRcRd, -(C0.3 alkylene)NRcC(0)Rd, -(C0.3 alkylene)OC(0)NRcRd, -(C0.3 alkylene)NRcC(0)NRcRd, -(C0.3 alkylene)NRcC(0)ORd, -(C0.3 alkylene) S(O)0-2Rc, -(C0.3 alkylene)NRcS(0)i_2Rd, -(C0.3 alkylene) S(0)i_2NRcRd, -(C0.3 alkylene)NRcS(0)i_2NRcRd or Ci_6 alkyl optionally substituted by oxo, -CN or halogen. In one embodiment, R5 is -(C0-3 alkylene)ORa, wherein said alkylene is optionally substituted by halogen, oxo, -CN, -ORc, -NRcRd, -C(0)ORc, or Ci_6 alkyl optionally substituted by oxo, -CN or halogen. In one embodiment, R5 is selected from -CH2OH, -CH2CH2OH, -C(CH3)2OH, -CH2C(CH3)2OH, -
H0*y^ H0'<.^ CH2OCH2CH(CH3)2, -CH2OCH2C(CH3)3, '
Figure imgf000046_0001
, wherein the wavy line represents the point of attachment in formula I.
In one embodiment, R5 is -(C0-3 alkylene)NRaRb, wherein said alkylene is independently optionally substituted by halogen, oxo, -(C0-3 alkylene)CN, -(C0-3 alkylene)ORc 0-3 alkylene)NRcRd, -(C0.3 alkylene)C(0)Rc, -(C0.3 alkylene)C(0)ORc, -(C0.3 alkylene)C(0)NRcRd, -(Co-3 alkylene)NRcC(0)Rd, -(C0.3 alkylene)OC(0)NRcRd, -(C0.3 alkylene)NRcC(0)NRcRd, -(Co-3 alkylene)NRcC(0)ORd, -(C0.3 alkylene) S(O)0-2Rc, -(C0.3 alkylene)NRcS(0)i_2Rd, -(C0.3 alkylene)S(0)i_2NRcRd, -(C0.3 alkylene)NRcS(0)i_2NRcRd or Ci_6 alkyl optionally substituted by oxo, -CN or halogen. In one embodiment, R5 is -(C0-3 alkylene)NRaRb, wherein said alkylene is optionally substituted by halogen, oxo, -CN, -ORc, -NRcRd, -C(0)ORc, or Ci_6 alkyl optionally substituted b oxo -CN or halo en. In one embodiment R5 is selected from -NHCH2CH2OH, -
Figure imgf000046_0002
, wherein the wavy line represents the point of attachment in formula I. In one embodiment, R5 is -(C0-3 alkylene)C3_i2 cycloalkyl, wherein said alkylene and cycloalkyl are independently optionally substituted by halogen, oxo, -(C0-3 alkylene)CN, -(C0-3 alkylene)ORc, -(C0.3 alkylene)NRcRd, -(C0.3 alkylene)C(0)Rc, -(C0.3 alkylene)C(0)ORc, -(C0.3 alkylene)C(0)NRcRd, -(C0.3 alkylene)NRcC(0)Rd, -(C0.3 alkylene)OC(0)NRcRd, -(C0.3 alkylene)NRcC(0)NRcRd, -(C0.3 alkylene)NRcC(0)ORd, -(C0.3 alkylene)S(0)o-2Rc, -(C0.3 alkylene)NRcS(0)i_2Rd, -(C0.3 alkylene)S(0)i_2NRcRd, -(C0.3 alkylene)NRcS(0)i_2NRcRd or Ci_6 alkyl optionally substituted by oxo, -CN or halogen. In one embodiment, R5 is -(C0-3 alkylene)C3_6 cycloalkyl, wherein said alkylene and cycloalkyl are independently optionally substituted by halogen, oxo, -CN, -ORc, -NRcRd, -C(0)ORc, or Ci_6 alkyl optionally substituted by oxo, -CN or halogen. In one embodiment, R5 is selected from -CH2cyclopentyl, -CH2cyclopropyl, -CH2CH2cyclopropyl, cyclopropyl, 2,2-difluorocyclopropyl and cyclobutyl.
In one embodiment, R5 is -(C0-3 alkylene)C3_7 cycloalkyl. In another embodiment, R5 is cyclopropyl or cyclobutyl.
In one embodiment, R5 is -(C0-3 alkylene)C(0)NRaRb, wherein said alkylene is independently optionally substituted by halogen, oxo, -(C0-3 alkylene)CN, -(C0-3 alkylene)ORc, -(Co-3 alkylene)NRcRd, -(C0.3 alkylene)C(0)Rc, -(C0.3 alkylene)C(0)ORc, -(C0.3 alkylene)C(0)NRcRd, -(C0.3 alkylene)NRcC(0)Rd, -(C0.3 alkylene)OC(0)NRcRd, -(C0.3 alkylene)NRcC(0)NRcRd, -(C0.3 alkylene)NRcC(0)ORd, -(C0.3 alkylene) S(O)0_2Rc, -(C0.3 alkylene)NRcS(0)i_2Rd, -(C0.3 alkylene) S(0)i_2NRcRd, -(C0.3 alkylene)NRcS(0)i_2NRcRd or Ci_6 alkyl optionally substituted by oxo, -CN or halogen. In one embodiment, R5 is -(C0-3 alkylene)C(0)NRaRb, wherein said alkylene is optionally substituted by halogen, oxo, -CN, -ORc, -NRcRd, -C(0)ORc, or Ci_6 alkyl optionally substituted by oxo, -CN or halogen. In one embodiment, R5 is selected from -CH2C(0)NH2, -CH2C(0)NHcyclopentyl -CH2C(0)N(CH3)(cyclopentyl), -CH2C(0)NHCH3, -CH(CH3)C(0)NHCH(CH3)2,
-CH2C(0)(pyrrolidin-l-yl), -CH2C(0)(4,4-difluorpiperidin-l-yl), -CH2C(0)(morpholinyl) and
Figure imgf000047_0001
, wherein the wavy line represents the point of attachment in formula I.
In one embodiment, R5 is -(C0-3 alkylene)C(0)NRaRb, wherein said alkylene is independently optionally substituted by halogen, oxo, -(C0-3 alkylene)CN, -(C0-3 alkylene)ORc, -(Co-3 alkylene)NRcRd, -(C0.3 alkylene)C(0)Rc, -(C0.3 alkylene)C(0)ORc, -(C0.3 alkylene)C(0)NRcRd, -(C0.3 alkylene)NRcC(0)Rd, -(C0.3 alkylene)OC(0)NRcRd, -(C0.3 alkylene)NRcC(0)NRcRd, -(C0.3 alkylene)NRcC(0)ORd, -(C0.3 alkylene) S(O)0_2Rc, -(C0.3 alkylene)NRcS(0)i_2Rd, -(C0.3 alkylene) S(0)i_2NRcRd, -(C0.3 alkylene)NRcS(0)i_2NRcRd or Ci_6 alkyl optionally substituted by oxo, -CN or halogen. In one embodiment, R5 is -(C0-3 alkylene)C(0)NRaRb, wherein said alkylene is optionally substituted by oxo or halogen; and Ra and Rb are independently hydrogen, Ci_6 alkyl, C3_6 cycloalkyl, 4-6 membered heterocyclyl, wherein said alkyl, cycloalkyl and heterocyclyl are independently optionally substituted by halogen, oxo, -CN, -ORe or -NReRf, or taken together with the atom to which they are attached to form a 3-6 membered heterocyclyl optionally substituted by oxo, halogen, -C(0)Ci_6 alkyl or Ci_6 alkyl. In another embodiment, R5 is -CH2C(0)NRaRb, -CH2C(0)NHRa, and Ra and Rb are independently hydrogen, Ci_6 alkyl, C3-6 cycloalkyl, 4-6 membered heterocyclyl, wherein said alkyl, cycloalkyl and heterocyclyl are independently optionally substituted by halogen, oxo, -CN, -ORe or -NReRf, or taken together with the atom to which they are attached to form a 3-6 membered heterocyclyl optionally substituted by oxo, halogen, -C(0)Ci_6 alkyl or Ci_6 alkyl. In another embodiment, R5 is -CH2C(0)NHCH3, -CH2C(0)N(CH3)(cyclopentyl), -CH2C(0)NH(cyclopentyl), -CH2C(0)NH(isopropyl), -CH2C(0)(pyrrolidin- 1 -yl),
-CH2C(0)(4,4-difiuorpiperidin-l-yl), -CH2C(0)(morpholinyl) or
Figure imgf000048_0001
, wherein the wavy line represents the point of attachment in formula I. In one embodiment, R5 is -(C0-3 alkylene)NRaC(0)Rb, wherein said alkylene is independently optionally substituted by halogen, oxo, -(C0-3 alkylene)CN, -(C0-3 alkylene)ORc, -(Co-3 alkylene)NRcRd, -(C0.3 alkylene)C(0)Rc, -(C0.3 alkylene)C(0)ORc, -(C0.3 alkylene)C(0)NRcRd, -(C0.3 alkylene)NRcC(0)Rd, -(C0.3 alkylene)OC(0)NRcRd, -(C0.3 alkylene)NRcC(0)NRcRd, -(C0.3 alkylene)NRcC(0)ORd, -(C0.3 alkylene) S(O)0_2Rc, -(C0.3 alkylene)NRcS(0)i_2Rd, -(C0.3 alkylene) S(0)i_2NRcRd, -(C0.3 alkylene)NRcS(0)i_2NRcRd or Ci_6 alkyl optionally substituted by oxo, -CN or halogen. In one embodiment, R5 is -(C0-3 alkylene)NRaC(0)Rb, wherein said alkylene is optionally substituted by halogen, oxo, -CN, -ORc, -NRcRd, -C(0)ORc, or Ci_6 alkyl optionally substituted by oxo, -CN or halogen. In one embodiment, R5 is selected from -CH2NHC(0)CH3, -CH2NHC(0)CH(CH3)2, - CH2NHC(0)CH2CH3, -CH2NHC(0)CH2OCH3, -CH2NHC(0)pyridin-3-yl,
Figure imgf000048_0002
Figure imgf000049_0001
, wherein the wavy line represents the point of attachment in formula I.
In one embodiment, R5 is -(C0-3 alkylene)NRaS(0)i_2Rb, wherein said alkylene is independently optionally substituted by halogen, oxo, -(C0-3 alkylene)CN, -(C0-3 alkylene)ORc,
-(Co-3 alkylene)NRcRd, -(C0.3 alkylene)C(0)Rc, -(C0.3 alkylene)C(0)ORc, -(C0.3 alkylene)C(0)NRcRd, -(C0.3 alkylene)NRcC(0)Rd, -(C0.3 alkylene)OC(0)NRcRd, -(C0.3 alkylene)NRcC(0)NRcRd, -(C0.3 alkylene)NRcC(0)ORd, -(C0.3 alkylene) S(O)0_2Rc, -(C0.3 alkylene)NRcS(0)i_2Rd, -(C0.3 alkylene) S(0)i_2NRcRd, -(C0.3 alkylene)NRcS(0)i_2NRcRd or Ci_6 alkyl optionally substituted by oxo, -CN or halogen. In one embodiment, R5 is -(C0-3 alkylene)NRaS(0)i_2Rb, wherein said alkylene is optionally substituted by halogen, oxo, -CN, -ORc, -NRcRd, -C(0)ORc, or Ci_6 alkyl optionally substituted by oxo, -CN or halogen. In one embodiment, R5 is selected from -CH2NHS(0)2CH3, -CH2NHS(0)2CH2CH3, - CH2NHS(0)2CH2CH(CH3)2, -CH2NHS(0)2CH(CH3)2, -CH2NHS(0)2CH(CH3)CH2CH3, - CH2NHS(0)2cyclopropyl, -CH2NHS(0)2cyclopentyl, -CH2N(CH3)2S(0)2CH3,
CH2CH2NHS(0)2CH3, -CH2CH2NHS(0)2CH2CH3,
Figure imgf000049_0002
Figure imgf000049_0003
, , wherein the wavy line represents the point of attachment in formula I.
In one embodiment, R5 is -(C0-3 alkylene)5-12 membered heteroaryl, wherein said alkylene and heteroaryl are independently optionally substituted by halogen, oxo, -(C0-3 alkylene)CN, -(C0.3 alkylene)ORc, -(C0.3 alkylene)NRcRd, -(C0.3 alkylene)C(0)Rc, -(C0.3 alkylene)C(0)ORc, -(C0.3 alkylene)C(0)NRcRd, -(C0.3 alkylene)NRcC(0)Rd, -(C0.3 alkylene)OC(0)NRcRd, -(C0.3 alkylene)NRcC(0)NRcRd, -(C0.3 alkylene)NRcC(0)ORd, -(C0.3 alkylene)S(0)o_2Rc, -(C0.3 alkylene)NRcS(0)i_2Rd, -(C0.3 alkylene) S(0)i_2NRcRd, -(C0.3 alkylene)NRcS(0)i_2NRcRd or Ci_6 alkyl optionally substituted by oxo, -CN or halogen. In one embodiment, R5 is -(C0-3 alkylene)5-6 membered heteroaryl, wherein said alkylene and heteroaryl are independently optionally substituted by halogen, oxo, -CN, -ORc, -NRcRd, -C(0)ORc, or Ci_6 alkyl optionally substituted by oxo, -CN or halogen. In one embodiment, R5 is selected from -CH2CH2triazolyl, triazolyl, pyridinyl, -CH2pyrazolyl, -CH2pyridinyl,
Figure imgf000050_0001
Figure imgf000050_0002
, wherein the wavy line represents the point of attachment in formula I.
In one embodiment, R5 is -(C0-3 alkylene)4-6 membered heteroaryl, wherein said alkylene is independently optionally substituted by halogen, oxo, -(C0-3 alkylene)CN, -(C0-3 alkylene)ORc, -(C0.3 alkylene)NRcRd, -(C0.3 alkylene)C(0)Rc, -(C0.3 alkylene)C(0)ORc, -(C0.3 alkylene)C(0)NRcRd, -(C0.3 alkylene)NRcC(0)Rd, -(C0.3 alkylene)OC(0)NRcRd, -(C0.3 alkylene)NRcC(0)NRcRd, -(C0.3 alkylene)NRcC(0)ORd, -(C0.3 alkylene) S(O)0_2Rc, -(C0.3 alkylene)NRcS(0)i_2Rd, -(C0.3 alkylene) S(0)i_2NRcRd, -(C0.3 alkylene)NRcS(0)i_2NRcRd or Ci_6 alkyl optionally substituted by oxo, -CN or halogen. In one embodiment, R5 is -(C0-3 alkylene)4-6 membered heteroaryl, wherein said alkylene is optionally substituted by oxo or halogen, and said heteroaryl is optionally substituted by oxo, halogen, Ci_3 alkyl, -ORc or -NRcRd. In one embodiment, R5 is pyridinyl.
In one embodiment, R5 is -(C0-3 alkylene)3-12 membered heterocyclyl, wherein said alkylene and heterocyclyl are independently optionally substituted by halogen, oxo, -(C 0-3 alkylene)CN, -(C0.3 alkylene)ORc, -(C0.3 alkylene)NRcRd, -(C0.3 alkylene)C(0)Rc, -(C0.3 alkylene)C(0)ORc, -(C0.3 alkylene)C(0)NRcRd, -(C0.3 alkylene)NRcC(0)Rd, -(C0.3 alkylene)OC(0)NRcRd, -(C0.3 alkylene)NRcC(0)NRcRd, -(C0.3 alkylene)NRcC(0)ORd, -(C0.3 alkylene)S(0)o_2Rc, -(C0.3 alkylene)NRcS(0)i_2Rd, -(C0.3 alkylene) S(0)i_2NRcRd, -(C0.3 alkylene)NRcS(0)i_2NRcRd or Ci_6 alkyl optionally substituted by oxo, -CN or halogen. In one embodiment, R5 is -(C0-3 alkylene)3-7 membered heterocyclyl, wherein said alkylene and heterocyclyl are independently optionally substituted by halogen, oxo, -CN, -ORc, -NRcRd, -C(0)ORc, or Ci_6 alkyl optionally substituted by oxo, -CN or halogen. In one embodiment, R5 is selected from oxetanyl, 1,1-dioxothiomorpholinyl, -CH2CH2(l,l-dioxothiomorpholinyl), - CH2CH2triazolyl, triazolyl, -CH2pyrazolyl, -CH2pyridinyl, pyridinyl, pyrrolidinyl, piperidinyl, -CH2(4-hydroxypiperidin-l-yl), morpholinyl, azetidinyl, 2-acetylpyrrolidin-3-yl, -CH2tetrahydropyranyl, -CH2tetrahydropyran-4-yl, tetrahydropyranyl, tetrahydrofuranyl, -CH2tetrahydrofuran-2-yl, -CH2CH2tetrahydrofuranyl, -CH2morpholinyl, l-acetylpiperidin-4- -C(0)morpholinyl, -CH2C(0)morpholinyl, -CH2C(0)(l,l-dioxothiomorpholin-4-yl),
Figure imgf000051_0001
, , wherein the wavy line represents the point of attachment in formula I.
In one embodiment, R5 is -(C0-3 alkylene)4-6 membered heterocyclyl, wherein said alkylene is independently optionally substituted by halogen, oxo, -(C0-3 alkylene)CN, -(C0-3 alkylene)ORc, -(C0.3 alkylene)NRcRd, -(C0.3 alkylene)C(0)Rc, -(C0.3 alkylene)C(0)ORc, -(C0.3 alkylene)C(0)NRcRd, -(C0.3 alkylene)NRcC(0)Rd, -(C0.3 alkylene)OC(0)NRcRd, -(C0.3 alkylene)NRcC(0)NRcRd, -(C0.3 alkylene)NRcC(0)ORd, -(C0.3 alkylene) S(O)0_2Rc, -(C0.3 alkylene)NRcS(0)i_2Rd, -(C0.3 alkylene) S(0)i_2NRcRd, -(C0.3 alkylene)NRcS(0)i_2NRcRd or Ci_6 alkyl optionally substituted by oxo, -CN or halogen. In one embodiment, R5 is -(C0-3 alkylene)4-6 membered heterocyclyl, wherein said alkylene is optionally substituted by oxo or halogen, and said heterocyclyl is optionally substituted by oxo, halogen, Ci_3 alkyl, -ORc or -NRcRd. In one embodiment, R5 is -CH2C(0)(4-6 membered heterocyclyl) or -CH2(4-6 membered heterocyclyl), wherein said heterocyclyl is optionally substituted by oxo, halogen, Ci_ 3 alkyl, -ORc or -NRcRd. In another embodiment, said heterocyclyl is oxetanyl, pyridinyl, pyrrolindinyl, pyranyl, piperidinyl, morpholinyl or
O \— ' , wherein the wavy line represents the point of attachment in formula I. In another embodiment, R5 is pyridin-3-yl, pyrrolidin-l-yl, pyran-4-yl, -CH2C(0)(pyrrolidin-l-yl), -CH2C(0)(4,4-difiuorpiperidin-l-yl), -CH2 (morpholinyl), -CH2C(0)(morpholinyl), -CH2(pyrrolidin-2-on-l-yl) or
Figure imgf000052_0001
, wherein the wavy line represents the point of attachment in formula I.
In one embodiment, R5 is -(C0-3 alkylene)S(0)i_2Ra, wherein said alkylene is independently optionally substituted by halogen, oxo, -(C0-3 alkylene)CN, -(C0-3 alkylene)ORc, -(Co-3 alkylene)NRcRd, -(C0.3 alkylene)C(0)Rc, -(C0.3 alkylene)C(0)ORc, -(C0.3 alkylene)C(0)NRcRd, -(C0.3 alkylene)NRcC(0)Rd, -(C0.3 alkylene)OC(0)NRcRd, -(C0.3 alkylene)NRcC(0)NRcRd, -(C0.3 alkylene)NRcC(0)ORd, -(C0.3 alkylene) S(O)0_2Rc, -(C0.3 alkylene)NRcS(0)i_2Rd, -(C0.3 alkylene) S(0)i_2NRcRd, -(C0.3 alkylene)NRcS(0)i_2NRcRd or Ci_6 alkyl optionally substituted by oxo, -CN or halogen. In one embodiment, R5 is -(C0-3 alkylene) S(0)i_2Ra, wherein said alkylene is optionally substituted by halogen, oxo, -CN, -ORc, -NRcRd, -C(0)ORc, or Ci_6 alkyl optionally substituted by oxo, -CN or halogen. In one embodiment, R5 is selected from -CH2S(0)2CH3.
In one embodiment, R5 is -(C0-3 alkylene)C6_i2 aryl, wherein said alkylene and aryl are independently optionally substituted by halogen, oxo, -(C0-3 alkylene)CN, -(C0-3 alkylene)ORc, -(Co-3 alkylene)NRcRd, -(C0.3 alkylene)C(0)Rc, -(C0.3 alkylene)C(0)ORc, -(C0.3 alkylene)C(0)NRcRd, -(C0.3 alkylene)NRcC(0)Rd, -(C0.3 alkylene)OC(0)NRcRd, -(C0.3 alkylene)NRcC(0)NRcRd, -(C0.3 alkylene)NRcC(0)ORd, -(C0.3 alkylene) S(O)0_2Rc, -(C0.3 alkylene)NRcS(0)i_2Rd, -(C0.3 alkylene) S(0)i_2NRcRd, -(C0.3 alkylene)NRcS(0)i_2NRcRd or Ci_6 alkyl optionally substituted by oxo, -CN or halogen. In one embodiment, R5 is -(C0-3 alkylene)phenyl, wherein said alkylene and phenyl are independently optionally substituted by halogen, oxo, -CN, -ORc, -NRcRd, -C(0)ORc, or Ci_6 alkyl optionally substitu tteedd 1 by oxo, -CN cted from -CH2phenyl,
Figure imgf000052_0002
wherein the wavy line represents the point of attachment in formula I.
In one embodiment, R5 is -(C0-3 alkylene)phenyl, wherein said alkylene is optionally substituted by oxo or halogen, and said phenyl is optionally substituted by halogen, Ci_3 alkyl, -ORc or -NRcRd. In one embodiment, R5 is -(C0-3 alkylene)NRaC(0)ORb, wherein said alkylene is independently optionally substituted by halogen, oxo, -(C0-3 alkylene)CN, -(C0-3 alkylene)ORc,
-(Co-3 alkylene)NRcRd, -(C0.3 alkylene)C(0)Rc, -(C0.3 alkylene)C(0)ORc, -(C0.3 alkylene)C(0)NRcRd, -(C0.3 alkylene)NRcC(0)Rd, -(C0.3 alkylene)OC(0)NRcRd, -(C0.3 alkylene)NRcC(0)NRcRd, -(C0.3 alkylene)NRcC(0)ORd, -(C0.3 alkylene)S(0)o-2Rc, -(C0.3 alkylene)NRcS(0)i_2Rd, -(C0.3 alkylene)S(0)i_2NRcRd, -(C0.3 alkylene)NRcS(0)i_2NRcRd or Ci_6 alkyl optionally substituted by oxo, -CN or halogen. In one embodiment, R5 is -(C0-3 alkylene)NRaC(0)ORb, wherein said alkylene is optionally substituted by halogen, oxo, -CN, -ORc, -NRcRd, -C(0)ORc, or Ci_6 alkyl optionally substituted by oxo, -CN or halogen. In one embodiment, R5 is selected from -CH2NHC(0)OCH2CH3 and -CH2NHC(0)OCH3.
In one embodiment, R5 is selected from hydrogen, fluro, methyl, ethyl, 1 -hydro xyethyl,
2-hydroxyethyl, propyl, isopropyl, butyl, 2-methylbutyl, isobutyl, -CH2F, -CHF2, -CF3, - CH2OH, -C(CH3)2OH, -CN, -C(CH3)2CN, -CH2CH2OH, -CH2C(CH3)2OH, -
CH
Figure imgf000053_0001
H2, -CH2C(0)NH2,
Figure imgf000053_0002
-CH2cyclopentyl,
CH2cyclopropyl, -CH2CH2cyclopropyl, cyclopropyl, 2,2-difluorocyclopropyl, cyclobutyl, - CH2C(0)NH2, -CH2C(0)NHcyclopentyl -CH2C(0)N(CH3)(cyclopentyl), -CH2C(0)NHCH3, - CH(CH3)C(0)NHCH(CH3)2 -CH2C(0)(pyrrolidin- 1 -yl), -CH2C(0)(4,4-difluorpiperidin- 1 -yl),
-CH2C(0)(morpholinyl),
Figure imgf000053_0003
-CH2NHC(0)CH3, -CH2NHC(0)CH(CH3)2, - CH2NHC(0)CH2CH3, -CH2NHC(0)CH2OCH3, -CH2NHC(0)pyridin-3-yl,
Figure imgf000053_0004
Figure imgf000054_0001
I , -CH2NHS(0)2CH3, -CH2NHS(0)2CH2CH3,
CH2NHS(0)2CH2CH(CH3)2, -CH2NHS(0)2CH(CH3)2, -CH2NHS(0)2CH(CH3)CH2CH3, CH2NHS(0)2cyclopropyl, -CH2NHS(0)2cyclopentyl, -CH2N(CH3)2S(0)2CH3,
CH2CH2NHS(0)2CH3, -CH2CH2NHS(0)2CH2CH3,
Figure imgf000054_0002
Figure imgf000054_0003
s oxetanyl, 1,1-dioxothiomorpholinyl,
-CH2CH2(l,l-dioxothiomorpholinyl), -CH2CH2triazolyl, triazolyl, -CH2pyrazolyl, - CH2pyridinyl, pyridinyl, pyrrolidinyl, piperidinyl, morpholinyl, azetidinyl, 2-acetylpyrrolidin-3- yl, -CH2tetrahydropyranyl, tetrahydropyranyl, tetrahydrofuranyl, -CH2CH2tetrahydrofuranyl, -CH2morpholinyl, l-acetylpiperidin-4-yl, -C(0)morpholi -CH2C(0)morpholinyl,
-CH2C(0)(l,l-dioxothiomorpholin-4-yl), -CH2C(0)pyrrolidinyl,
Figure imgf000054_0004
Figure imgf000054_0005
, -CH2phenyl, phenyl, , and
Figure imgf000054_0006
wherein the wavy line represents the point of attachment in formula I. In certain embodiments, Y is CR5 and R5 is methyl, ethyl, propyl, isopropyl, cyclopropyl, eye lo butyl, cyano, 2-methylbutyl, N-(2-hydroxyethyl)amino, N-(2-methoxyethyl)amino, methylsulfonylamino methyl, 2-(methylsulfonylamino)ethyl, cyclopropylmethyl, 2-[N-(2- propylsulfonyl)amino]ethyl, 2-[N-(cyclopropylsulfonyl)-amino]ethyl, 2- (cyclopropylcarbonylamino)ethyl, 2-(acetylamino)ethyl, 2-(methoxymethyl- carbonylamino)ethyl, cyclopentoxymethyl, cyclopropylmethoxymethyl, 2,2,2- trifluoroethoxymethyl, cyclohexyl, methylamino, 2-(N,N-dimethylaminocarbonyl)ethyl, 2-(N- acetyl-N-methylamino)ethyl, 2-(ethoxycarbonylamino)ethyl, 1 -hydro xyethyl, N- acylaminomethyl, 2-amino-l,l-difluoroethyl, Ν,Ν-dimethylamino, hydro xymethyl, methoxy, N- methylamino, N,N-dimethylamino,N-(2,2,2-trifluoroethyl)aminomethyl, (2- carboxycyclopropyl)(hydroxy)methyl, 2-hydro xyethyl, amino carbonylmethyl, methylammocarbonylmethyl, ethylamino carbonylmethyl, 1 -hydro xypropyl, 1 ,2-dihydro xyethyl, N-(2-methylpropyl)aminocarbonylmethyl, eye lopentylamino carbonylmethyl, 2-
(methoxycarbonylamino)ethyl, 2,2,2-trifluoro- 1 -hydro xyethyl, tert-butylaminocarbonylmethyl, cyclobutylaminocarbonylmethyl, 2-hydroxyethoxy, isopropylamino carbonylmethyl, N-(N'N'- diemthylaminocarbonylmethyl)aminocarbonylmethyl, 4,4-difluorocyclohexyl- amino carbonylmethyl, 2,2-difluoroethylaminocarbonylmethyl, N-(2-hydroxyethyl)-N- methylamino carbonylmethyl, cyclopentylmethyl, N-cyclopentyl-N-methylaminocarbonylmethyl,
2- amino-l,l-difluoroethyl, 3-pyridyl, morpho lino methyl, morpholmocarbonylmethyl, 2-cyano-2- methylethyl, trifluoromethyl, 1 -hydroxy- 1-methylethyl, l-(N-isopropylaminocarbonyl)ethyl, 2- hydroxy-2-methylpropyl, N-(methylsulfonyl)-N-methylaminomethyl, difluoromethyl, 2-(2- butylsulfonylamino)ethyl, 2-(4-fluorophenylcarbonylamino)ethyl, 2-(cyclobutylcarbonyl- amino)ethyl, 2-(2-methylbutanoylamino)ethyl, 2-(benzoylamino)ethyl, 2,2-difluorocyclopropyl,
3- cyanobenzyl, 2-methylpropoxymethyl, 2-cyclopropylethyl, 3-pyridylmethyl, methylsulfonylmethyl, ethoxycarbonylaminomethyl, 3 -pyridylcarbonylamino methyl, isopropylsulfonylaminomethyl, 2-pyridylcarbonylaminomethyl, cyclopropylsulfonyl- aminomethyl, cyclopentylsulfonylaminomethyl, 2-methylpropanoylaminomethyl, cyclopropylcarbonylaminomethyl, 2-fluorobenzoylaminomethyl, 3-fluorobenzoylaminomethyl, 1-methylpropylsulfonylaminomethyl, 2-methylpropylsulfonylaminomethyl, methoxyacetylaminomethyl, ethylsylfonylaminomethyl, 2-(3,3,3-trifluoropropylsulfonyl- amino)ethyl, 2-(2,2-difluorocyclopropylcarbonylamino)ethyl, fluoromethyl, 2- hydroxyethylamino, 2-methoxyethylamino, 1-aminoethyl, 2-(ethylsulfonylamino)ethyl, 2,2- dimethylpropo xymethyl, 1-metho xyethyl, tert-butylsulfonylaminomethyl, 2,2,2-trifluoroethyl- aminomethyl,
Figure imgf000056_0001
Figure imgf000056_0002
Figure imgf000057_0001
Figure imgf000057_0002
wherein the wavy line represents the point of attachment in formula I.
In one embodiment, R5 is methyl, 1 -hydro xyethyl, hydro xymethyl,
methylsulfonylaminomethyl, aminomethyl, or methoxycarbonyl-aminomethyl.
In one embodiment, R5 is (R)-l -hydro xyethyl.
In one embodiment, R5 is (S)-l -hydro xyethyl. In one embodiment, R5 is C3-12 alkyl, wherein said C3-12 alkyl, is independently optionally substituted by halogen, oxo, -(C0-3 alkylene)CN, -(C0-3 alkylene)ORc, -(C0-3 alkylene)NRcRd, -(Co-3 alkylene)C(0)Rc, -(C0.3 alkylene)C(0)ORc, -(C0.3 alkylene)C(0)NRcRd, -(C0.3 alkylene)NRcC(0)Rd, -(C0.3 alkylene)OC(0)NRcRd, -(C0.3 alkylene)NRcC(0)NRcRd, -(C0.3 alkylene)NRcC(0)ORd, -(C0.3 alkylene) S(O)0_2Rc, -(C0.3 alkylene)NRcS(0)i_2Rd, -(C0.3 alkylene)S(0)i_2NRcRd, -(C0.3 alkylene)NRcS(0)i_2NRcRd.
In one embodiment, R5 is C1-12 alkyl, wherein said alkyl, alkenyl, alkynyl, alkylene, cycloalkyl, aryl and heterocyclyl are independently optionally substituted by halogen, oxo, -(C0- 3 alkylene)CN, -(C2_3 alkylene)ORc, -(C0.3 alkylene)NRcRd, -(C0.3 alkylene)C(0)Rc, -(C0.3 alkylene)C(0)ORc, -(C0.3 alkylene)C(0)NRcRd, -(C0.3 alkylene)NRcC(0)Rd, -(C0.3 alkylene)OC(0)NRcRd, -(C0.3 alkylene)NRcC(0)NRcRd, -(C0.3 alkylene)NRcC(0)ORd, -(C0.3 alkylene)S(0)o-2Rc, -(C0.3 alkylene)NRcS(0)i_2Rd, -(C0.3 alkylene) S(0)i_2NRcRd, -(C0.3 alkylene)NRcS(0)i_2NRcRd
1. In one embodiment, R5 is C1-12 alkyl, wherein said alkyl, alkenyl, alkynyl, alkylene, cycloalkyl, aryl and heterocyclyl are independently optionally substituted by halogen, oxo, -(Co-3 alkylene)CN, -(C0.3 alkylene)NRcRd, -(C0.3 alkylene)C(0)Rc, -(C0.3 alkylene)C(0)ORc, -(C0.3 alkylene)C(0)NRcRd, -(C0.3 alkylene)NRcC(0)Rd, -(C0.3 alkylene)OC(0)NRcRd, -(C0.3 alkylene)NRcC(0)NRcRd, -(C0.3 alkylene)NRcC(0)ORd, -(C0.3 alkylene) S(O)0. 2RC, -(Co-3 alkylene)NRcS(0)i.2Rd, -(C0.3 alkylene) S(0)i_2NRcRd, -(C0.3 alkylene)NRcS(0)i-2NRcRd
In one embodiment, R5 is halogen, C1-12 alkyl, C2_i2 alkenyl, C2_i2 alkynyl, -(C0-3 alkylene)CN, -(C0.3 alkylene)NRaRb, -(C0.3 alkylene)ORa, -(C0.3 alkylene)SRa, -(C0.3 alkylene)C(0)Ra, -(C0.3 alkylene)NRaC(0)Rb, -(C0.3 alkylene)C(0)NRaRb, -(C0.3 alkylene)C(0)ORa, -(C0.3 alkylene)OC(0)Ra, -(C0.3 alkylene)NRaC(0)NRaRb, -(C0.3 alkylene)OC(0)NRaRb, -(C0.3 alkylene)NRaC(0)ORb, -(C0.3 alkylene) S(0)i_2Ra, -(C0.3 alkylene)NRaS(0)i_2Rb, -(C0.3 alkylene)S(0)i_2NRaRb, -(C0.3 alkylene)NRaS(0)i_2NRaRb, -(C0.3 alkylene)C3_i2 cycloalkyl, -(C0-3 alkylene)C6_i4 aryl, -(C0-3 alkylene)3-12 membered heterocyclyl or -(C0-3 alkylene)C(0)3-12 membered heterocyclyl, wherein said alkyl, alkenyl, alkynyl, alkylene, cycloalkyl, aryl and heterocyclyl are independently optionally substituted by halogen, oxo, -(C0.3 alkylene)CN, -(C0.3 alkylene)ORk, -(C0.3 alkylene)NRcRd, -(C0.3 alkylene)C(0)Rc, -(C0.3 alkylene)C(0)ORc, -(C0.3 alkylene)C(0)NRcRd, -(C0.3 alkylene)NRcC(0)Rd, -(C0.3 alkylene)OC(0)NRcRd, -(C0.3 alkylene)NRcC(0)NRcRd, -(C0.3 alkylene)NRcC(0)ORd, -(C0.3 alkylene) S(O)0_2Rc, -(C0.3 alkylene)NRcS(0)i_2Rd, -(C0.3 alkylene)S(0)i_2NRcRd, -(C0.3 alkylene)NRcS(0)i_2NRcRd or Ci_6 alkyl optionally substituted by oxo, -CN or halogen; wherein each Rk is independently hydrogen, C3-6 alkyl, C2_6 alkenyl, C2_6 alkynyl, -(C0-3 alkylene)C3-6 cycloalkyl, -(C0-3 alkylene)3-12 membered heterocyclyl, -(C0-3 alkylene)C(0)3-12 membered heterocyclyl or -(C0-3 alkylene)C6_i4 aryl, wherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl and aryl are independently optionally substituted by halogen, oxo, -CN, -ORg, -NRgRh, -C(0)Rg, -C(0)ORg, -C(0)NRgRh, -NRgC(0)Rh, -OC(0)NRgRh, -NRgC(0)NRgRh, -NRgC(0)ORh, -S(0)i_2Rg, -NRgS(0)i_2Rh, -S(0)i_2NRgRh, -NRgS(0)i_2NRgRh, C3-6 cycloalkyl, 3-6 membered heterocyclyl, phenyl or Ci_6 alkyl optionally substituted by oxo or halogen, or taken together with the atom to which they are attached to form a 3-6 membered heterocyclyl optionally substituted by oxo, halogen, -C(0)Ci_6 alkyl or alkyl optionally substituted by oxo or halogen.
In one embodiment, R5 is halogen, C1-12 alkyl, C2-12 alkenyl, C2-12 alkynyl, -(C0-3 alkylene)CN, -(C0.3 alkylene)NRaRb, -(C0.3 alkylene)ORa, -(C0.3 alkylene)SRa, -(C0.3 alkylene)C(0)Ra, -(C0.3 alkylene)NRaC(0)Rb, -(C0.3 alkylene)C(0)NRaRb, -(C0.3 alkylene)C(0)ORa, -(C0.3 alkylene)OC(0)Ra, -(C0.3 alkylene)NRaC(0)NRaRb, -(C0.3 alkylene)OC(0)NRaRb, -(C0.3 alkylene)NRaC(0)ORb, -(C0.3 alkylene)S(0)i_2Ra, -(C0.3 alkylene)NRaS(0)i_2Rb, -(C0.3 alkylene)S(0)i_2NRaRb, -(C0.3 alkylene)NRaS(0)i_2NRaRb, -(C0.3 alkylene)C3_i2 cycloalkyl, -(C0-3 alkylene)C6_i4 aryl, -(C0-3 alkylene)3-12 membered heterocyclyl or -(C0-3 alkylene)C(0)3-12 membered heterocyclyl, wherein said alkyl, alkenyl, alkynyl, alkylene, cycloalkyl, aryl and heterocyclyl are independently optionally substituted by halogen, oxo, -(C0.3 alkylene)CN, -(C0.3 alkylene)ORk, -(C0.3 alkylene)NRcRd, -(C0.3 alkylene)C(0)Rc, -(C0.3 alkylene)C(0)ORc, -(C0.3 alkylene)C(0)NRcRd, -(C0.3 alkylene)NRcC(0)Rd, -(C0.3 alkylene)OC(0)NRcRd, -(C0.3 alkylene)NRcC(0)NRcRd, -(C0.3 alkylene)NRcC(0)ORd, -(C0.3 alkylene) S(O)0_2Rc, -(C0.3 alkylene)NRcS(0)i_2Rd, -(C0.3 alkylene)S(0)i_2NRcRd, -(C0.3 alkylene)NRcS(0)i_2NRcRd or Ci_6 alkyl optionally substituted by oxo, -CN or halogen; wherein each Rk is independently hydrogen, C2_6 alkenyl, C2_6 alkynyl, -(Co-3 alkylene)C3_6 cycloalkyl, -(C0-3 alkylene)3-12 membered heterocyclyl, -(C0-3 alkylene)C(0)3-12 membered heterocyclyl or -(C0-3 alkylene)C6_i4 aryl, wherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl and aryl are independently optionally substituted by halogen, oxo, -CN, -ORg, -NRgRh, -C(0)Rg, -C(0)ORg, -C(0)NRgRh, -NRgC(0)Rh, -OC(0)NRgRh, -NRgC(0)NRgRh, -NRgC(0)ORh, -S(0)i_2Rg, -NRgS(0)i_2Rh, -S(0)i_2NRgRh, -NRgS(0)i_2NRgRh, C3-6 cycloalkyl, 3-6 membered heterocyclyl, phenyl or Ci_6 alkyl optionally substituted by oxo or halogen, or taken together with the atom to which they are attached to form a 3-6 membered heterocyclyl optionally substituted by oxo, halogen, -C(0)Ci_6 alkyl or Ci_6 alkyl optionally substituted by oxo or halogen.
In certain embodiments, R6 is independently oxo, halogen, -CN, -C(0)Ra, -C(0)ORa, -NRaC(0)Rb, -C(0)NRaRb, -NRaC(0)NRaRb, -OC(0)NRaRb, -NRaC(0)ORb, -S(0)i_2Ra, -NRaS(0)2Rb, -S(0)2NRaRb, -ORa, -SRa, -NRaRb, Ci_6 alkyl, C3_6 cycloalkyl, C2_6 alkenyl, C2_6 alkynyl, 3-7 membered heterocycly or C6-14 aryl, and wherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl and aryl are independently optionally substituted by halogen, oxo, -CN, -ORc, -SRC, -NRcRd or Ci_6 alkyl optionally substituted by oxo or halogen. In one embodiment, R6 is independently oxo, halogen, -CN, -C(0)(Ci_6 alkyl), -C(0)0(d_6 alkyl), -S(0)2(d_6 alkyl), -NRaS(0)2(d_6 alkyl), -0(d_6 alkyl), d_6 alkyl, C3_6 cycloalkyl or 3-7 membered heterocyclyl, wherein said alkyl, cycloalkyl and heterocyclyl are independently optionally substituted by halogen, oxo, -CN, -ORc, -NRcRd or Ci_6 alkyl optionally substituted by halogen. In one embodiment, R6 is independently oxo, F, CI, -CN, -OH, -C(0)CH3, -CH2CN, -CH2CH2CN, cyclopropyl, cyclobutyl, -CF3, -NHS(0)2CH3, -S(0)2CH3, -C(0)OCH3, pyrrolidinyl or pyrrolidinonyl.
In one embodiment, R6 is independently oxo, halogen, -CN, -C(0)(Ci_6 alkyl), -S(0)2(Ci_6 alkyl), -ORa, -NRaRb, Ci_6 alkyl or C3_6 cycloalkyl, and wherein said alkyl, alkenyl and alkynyl are independently optionally substituted by halogen, oxo, -CN, -ORc or -NRcRd. In one embodiment, R6 is halogen, -S(0)2CH3 or -CN.
In one embodiment, R3 is optionally substituted by 1 to 3 R6 independently selected from oxo, halogen, -CN, -S(0)2(Ci_6 alkyl), -ORa, -NRaRb and Ci_6 alkyl, and wherein said alkyl, alkenyl and alkynyl are independently optionally substituted by halogen, oxo, -CN, -ORc or -NRcRd.
In one embodiment, R3 is optionally substituted by 1 to 3 R6 independently selected from oxo, halogen, -CN, -C(0)(Ci_6 alkyl), -C(0)0(Ci_6 alkyl), -S(0)2(C^ alkyl), -NRaS(0)2(Ci_6 alkyl), -0(Ci_6 alkyl), Ci_6 alkyl, C3_6 cycloalkyl or 3-7 membered heterocyclyl, wherein said alkyl, cycloalkyl and heterocyclyl are independently optionally substituted by halogen, oxo, -CN, -ORc, -NRcRd or Ci_6 alkyl optionally substituted by halogen.
In certain embodiments, each Ra and Rb are independently hydrogen, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, -C3_6 cycloalkyl, -3-12 membered heterocyclyl, -C(0)3-12 membered heterocyclyl or -C6-i4 aryl, wherein said alkyl, cycloalkyl, heterocyclyl and aryl are independently optionally substituted by halogen, oxo, -CN, -ORe, -NReRf, -C(0)Rg, -C(0)ORg, -C(0)NRgRh, -NRgC(0)Rh, -OC(0)NRgRh, -NRgC(0)NRgRh, -NRgC(0)ORh, -S(0)i_2Rg, -NRgS(0)i_2Rh, -S(0)i_2NRgRh, -NRgS(0)i_2NRgRh, C3_6 cycloalkyl, 3-6 membered heterocyclyl, phenyl or Ci_3 alkyl optionally substituted by oxo or halogen, or taken together with the atom to which they are attached to form a 3-6 membered heterocyclyl optionally substituted by oxo, halogen, -C(0)Ci_6 alkyl or Ci_6 alkyl optionally substituted by oxo, halogen, ORg or NRgNRh. In certain embodiments, each Ra and Rb are independently hydrogen, Ci_6 alkyl, C3-6 cycloalkyl, 3-6 membered heterocyclyl, -C(0)3-6 membered heterocyclyl or phenyl, wherein said alkyl, cycloalkyl, heterocyclyl and phenyl are independently optionally substituted by halogen, oxo, -CN, -ORe, -NReRf, -C(0)Rg, -C(0)ORg, -C(0)NRgRh, -NRgC(0)Rh, -OC(0)NRgRh, -NRgC(0)NRgRh, -NRgC(0)ORh, -S(0)i_2Rg, -NRgS(0)i_2Rh, -S(0)i_2NRgRh, -NRgS(0)i_2NRgRh, C3-6 cycloalkyl, 3-6 membered heterocyclyl, phenyl or Ci_3 alkyl optionally substituted by oxo or halogen.
In one embodiment, each Ra and Rb are independently hydrogen, Ci_6 alkyl, C3_6 cycloalkyl, 3-6 membered heterocyclyl, 5-6 membered heteroaryl or phenyl, wherein said alkyl, cycloalkyl, heterocyclyl, heteroaryl and phenyl are independently optionally substituted by halogen, oxo, -CN, -ORe, -NReRf or Ci_3 alkyl optionally substituted by halogen.
In one embodiment, each Ra and Rb are independently selected from hydrogen, methyl, ethyl, propyl, isopropyl, butyl, t-butyl, sec-butyl, -CF3, -CH2CF3, -CH2F, -CHF2, -CH2OH, -CH2CH2OH, -CH2NH2, -CH2CH2NH2, -CH2CH2N(CH3)2, -CH2N(CH3)2, cyclopropyl, 2,2- difluorocyclopropyl, 2-fluorocyclopropyl, 2-methylcyclopropyl, eye lo butyl, cyclopentyl, cyclohexyl, piperidinyl, morpholinyl, piperazinyl, N-methylpiperazinyl, pyrazolyl, N- methylpyrazolyl, azetidinyl, 1,1-dioxothiomorpholinyl, pyrrolidinyl, pyrrolidinonyl, pyridinyl, cyanopyridinyl, phenyl and fluorophenyl.
In certain embodiments, a Ra and a Rb are independently taken together with the atom to which they are attached to form a 3-6 membered heterocyclyl optionally substituted by oxo, halogen, -C(0)Ci_6 alkyl or Ci_6 alkyl optionally substituted by oxo, halogen, ORg or NRgNRh.
In one embodiment, a Ra and a Rb are independently taken together with the atom to which they are attached to form a 3-6 membered heterocyclyl optionally substituted by oxo, halogen, -C(0)Ci_6 alkyl or Ci_6 alkyl optionally substituted by halogen. In one embodiment, said heterocyclyl is azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, piperidinonyl, morpholinyl and 1,1-dioxomorpholinyl.
In one embodiment, Ra and Rb are taken together with the atom to which they are attached to form a 4-6 membered heterocyclyl selected from azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl and morpholinyl, optionally substituted by oxo, halogen, -C(0)Ci_6 alkyl or Ci_6 alkyl. In one embodiment, Ra and Rb are independently hydrogen, methyl, isopropyl, cyclopropyl or cyclopentyl.
In one embodiment, Ra and Rb are taken together with the atom to which they are attached to form a 4-6 membered heterocyclyl selected from azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl and morpholinyl, optionally substituted by oxo, halogen, -C(0)Ci_6 alkyl or Ci_6 alkyl.
In certain embodiments, each Rc and Rd are independently hydrogen, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, -C3-6 cycloalkyl, -3-12 membered heterocyclyl, -C(0)3-12 membered heterocyclyl or -C6-14 aryl, wherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl and aryl are independently optionally substituted by halogen, oxo, -CN, -ORg, -NRgRh, -C(0)Rg, -C(0)ORg, -C(0)NRgRh, -NRgC(0)Rh, -OC(0)NRgRh, -NRgC(0)NRgRh, -NRgC(0)ORh, -S(0)i_2Rg, -NRgS(0)i_2Rh, -S(0)i_2NRgRh, -NRgS (O) 1 _2NRgRh, C3_6 cycloalkyl, 3-6 membered heterocyclyl, phenyl or Ci_6 alkyl optionally substituted by oxo or halogen, or taken together with the atom to which they are attached to form a 3-6 membered heterocyclyl optionally substituted by oxo, halogen, -C(0)Ci_6 alkyl or Ci_6 alkyl optionally substituted by oxo or halogen.
In certain embodiments, each Rc and Rd are independently hydrogen, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, -C3_6 cycloalkyl, -3-6 membered heterocyclyl, -C(0)3-6 membered heterocyclyl or phenyll, wherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl and phenyl are independently optionally substituted by halogen, oxo, -CN, -ORg, -NRgRh, -C(0)Rg, -C(0)ORg, -C(0)NRgRh, -NRgC(0)Rh, -OC(0)NRgRh, -NRgC(0)NRgRh, -NRgC(0)ORh, -S(0)i_2Rg, -NRgS(0)i_2Rh, -S(0)i_2NRgRh, -NRgS (O) 1 _2NRgRh, C3_6 cycloalkyl, 3-6 membered heterocyclyl, phenyl or Ci_6 alkyl optionally substituted by oxo or halogen.
In one embodiment, each Rc and Rd are independently hydrogen, Ci_6 alkyl, C3_6 cycloalkyl, 3-6 membered heterocyclyl, 5-6 membered heteroaryl or phenyl, wherein said alkyl, cycloalkyl, heterocyclyl, heteroaryl and phenyl are independently optionally substituted by halogen, oxo, -CN, -ORg, -NRgRh or Ci_6 alkyl optionally substituted by halogen.
In one embodiment, each Rc and Rd are independently hydrogen, methyl, ethyl, isopropyl, butyl, t-butyl, sec-butyl, -CF3, "CH2CF3, -CH2F, -CHF2, -CH2OH, -CH2CH2OH, -CH2NH2, -CH2CH2NH2, -CH2CH2N(CH3)2, -CH2N(CH3)2, cyclopropyl, 2,2- difluorocyclopropyl, 2-fluorocyclopropyl, 2-methylcyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, piperidinyl, morpholinyl, piperazinyl, N-methylpiperazinyl, pyrazolyl, N- methylpyrazolyl, azetidinyl, 1,1-dioxothiomorpholinyl, pyrrolidinyl, pyrrolidinonyl, pyridinyl, cyanopyridinyl, phenyl and fluorophenyl.
In certain embodiments, a Rc and a Rd are independently taken together with the atom to which they are attached to form a 3-6 membered heterocyclyl optionally substituted by oxo, halogen, -C(0)Ci_6 alkyl or Ci_6 alkyl optionally substituted by oxo or halogen.
In one embodiment, each Rc and Rd are independently hydrogen, methyl or ethyl, optionally substituted by fluoro or oxo. In one embodiment, each Rc and Rd are independently hydrogen, methyl or ethyl. In one embodiment, a Rc and a Rd are taken together with the atom to which they are attached to form a 3-6 membered heterocyclyl optionally substituted by oxo, halogen, -C(0)Ci_6 alkyl or Ci_6 alkyl optionally substituted by halogen. In one embodiment, said heterocyclyl is azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, piperidinonyl, morpholinyl and 1,1- dioxomorpholinyl. In one embodiment, Rc, Rd, Re, Rf, Rg and Rh are independently hydrogen or methyl.
In one embodiment, each Re, Rf, Rg, Rh are independently hydrogen, methyl, ethyl, propyl or isopropyl, optionally substituted by halogen or oxo. In one embodiment, each Re, Rf, Rg, Rh are independently hydrogen, methyl or ethyl.
In another embodiment, X is CR4; Y is N or CR5;
R1 is azetidinyl, piperidinyl, pyrrolidinyl or cyclohexyl, optionally substituted by Ci_3 alkylene or Ci_3 alkyl;
R2 is absent, Ci_3 alkyl, - H-, -NHCH2- -CH20- -(CH2)20-, -C(0)NH- -CH2C(0)NH-, -CH2C(0)N(CH3)-, -NHC(0)CH2-, - HC(0)0-, -C(0)0-, -C(0)CH2S(0)2, -NHS(0)2-, -NHS(0)2CH2-, -CH2C(0)-, -(CH2)2C(0)-, -S(0)2- -CH2S(0)2-, -S(0)2(CH2)2-;
R3 is absent, Ci_6 alkyl optionally substituted by oxo, halogen, -CN, -S(0)2(Ci_6 alkyl), -ORa or -NRaRb;
C3-7 cycloalkyl optionally substituted by oxo, halogen, -CN, -S(0)2(Ci_6 alkyl), -ORa, -NRaRb or Ci_6 alkyl optionally substituted by halogen, phenyl optionally substituted by Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, halogen,
-CF3, -CN, -S(0)2(Ci_6 alkyl), -ORa or -NRaRb,
5-6 membered heteroaryl optionally substituted by oxo, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, -CF3, halogen, -CN, -S(0)2(Ci_6 alkyl), -ORa or -NRaRb, or
4-7 membered heterocyclyl optionally substituted by oxo, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, -CF3, halogen, -CN, -S(0)2(Ci_6 alkyl), -ORa or -NRaRb;
R4 is hydrogen, F or methyl;
R5 is Ci_6 alkyl or C3_6 cycloalkyl optionally substituted by halogen, oxo, -CN, -ORa or -NRaRb,
-CH2C(0)NRaRb, -CH2C(0)NHRa, -CH2C(0)(4-6 membered heterocyclyl), -CH2(4-6 membered heterocyclyl), -(Co_3 alkylene)4-6 membered heteroaryl or
-(Co-3 alkylene)phenyl, wherein said alkylene is optionally substituted by oxo or halogen, and said heterocyclyl, heteroaryl and phenyl are independently optionally substituted by oxo, halogen, Ci_3 alkyl, -ORc or -NRcRd; each Ra and Rb are independently hydrogen, Ci_3 alkyl or C3_6 cycloalkyl, wherein said alkyl and cycloalkyl are independently optionally substituted by oxo or halogen; or are taken together with the atom to which they are attached to form a 3-6 membered heterocyclyl optionally substituted by oxo, halogen or Ci_3 alkyl; and each Rc and Rd are independently hydrogen or Ci_6 alkyl; or are taken together with the atom to which they are attached to form a 3-6 membered heterocyclyl optionally substituted by oxo, halogen, or Ci_3_ alkyl.
In another embodiment, X is CR4; Y is N or CR5; R1 is azetidinyl, piperidinyl, pyrrolidinyl, tetrahydropyranyl, cyclopentyl or cyclohexyl, optionally substituted by Ci_3 alkylene, -CN, -ORa or Ci_3 alkyl; R2 is absent, Ci_3 alkyl, - H-, -NHCH2-, -CH20-, -(CH2)20- -C(0)NH-, -CH2C(0)NH-, -CH2C(0)N(CH3)-, -NHC(0)CH2-, - HC(0)0-, -C(0)0-, -C(0)CH2S(0)2, -NHS(0)2-, -NHS(0)2CH2-, -CH2C(0)-, -(CH2)2C(0)-, -S(0)2-, -CH2S(0)2-, -S(0)2(CH2)2-;
R3 is absent, hydrogen,
Ci_6 alkyl optionally substituted by oxo, halogen, -CN, -S(0)2(Ci_6 alkyl), -ORa or -NRaRb;
C3_7 cycloalkyl optionally substituted by oxo, halogen, -CN, -S(0)2(Ci_6 alkyl), -ORa, -NRaRb or Ci_6 alkyl optionally substituted by halogen, phenyl optionally substituted by Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, halogen, -CF3, -CN, -S(0)2(Ci_6 alkyl), -ORa or -NRaRb,
5-6 membered heteroaryl optionally substituted by oxo, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, -CF3, halogen, -CN, -S(0)2(Ci_6 alkyl), -ORa or -NRaRb, or
4-7 membered heterocyclyl optionally substituted by oxo, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, -CF3, halogen, -CN, -S(0)2(Ci_6 alkyl), -ORa or -NRaRb;
R4 is hydrogen, F or methyl;
R5 is C1-12 alkyl or C3_i2 cycloalkyl optionally substituted by halogen, oxo, -CN, -ORa or -NRaRb,
-CH2C(0)NRaRb, -CH2C(0)NHRa, -CH2C(0)(4-6 membered heterocyclyl), -CH2(4-6 membered heterocyclyl), -(Co_3 alkylene)4-6 membered heteroaryl or -(C0-3 alkylene)phenyl, wherein said alkylene is optionally substituted by oxo or halogen, and said heterocyclyl, heteroaryl and phenyl are independently optionally substituted by oxo, halogen, Ci_3 alkyl, -ORc or -NRcRd; each Ra and Rb are independently hydrogen, Ci_3 alkyl or C3_6 cycloalkyl, wherein said alkyl and cycloalkyl are independently optionally substituted by oxo or halogen; or are taken together with the atom to which they are attached to form a 3-6 membered heterocyclyl optionally substituted by oxo, halogen or Ci_3 alkyl; and each Rc and Rd are independently hydrogen or Ci_6 alkyl; or are taken together with the atom to which they are attached to form a 3-6 membered heterocyclyl optionally substituted by oxo, halogen, or Ci_3_ alkyl.
In another embodiment, R1 is azetidinyl, piperidinyl, pyrrolidinyl or cyclohexyl, optionally substituted by Ci_3 alkylene or Ci_3 alkyl; R2 is absent; and R3 is hydrogen. In another embodiment, R1 is azetidinyl, piperidinyl, pyrrolidinyl or cyclohexyl, optionally substituted by Ci_3 alkylene or Ci_3 alkyl; R2 is absent; R3 is hydrogen; and R5 is -CN or Ci_3 alkyl optionally substituted by halogen or oxo.
In another embodiment, X is CH; Y is CR5; R1 is piperidinyl, tetrahydropyranyl, cyclopentyl or cyclohexyl, wherein R1 is optionally substituted by Ci_3 alkylene, halogen, -ORa, -CN, -NRaRb or Ci_6 alkyl optionally substituted by oxo, -ORa, -CN, -NRaRb or halogen; R2 is absent; R3 is absent; R5 is C1-12 alkyl optionally substituted by halogen, oxo, -CN, -OH, -OCH3, -NH2 or -N(CH3)2; and each Ra and Rb are independently selected from Ci_3 alkyl optionally substituted by oxo or halogen. In another embodiment, X is CH; Y is CR5; R1 is piperidinyl, tetrahydropyranyl or cyclohexyl, wherein R1 is optionally substituted by Ci_3 alkylene, halogen, -OH, -NH2 or Ci_3 alkyl optionally substituted by oxo, -CN or halogen; R2 is absent; R3 is absent; and R5 is Ci_6 alkyl optionally substituted by halogen, oxo, -CN, -OH, -OCH3, -NH2 or -N(CH3)2.
In another embodiment, X is CH; Y is CR5; R1 is piperidinyl, tetrahydrofuranyl, tetrahydropyranyl, cyclopentyl or cyclohexyl, wherein R1 is optionally substituted by Ci_3 alkylene, halogen, -OH, -NH2 or Ci_3 alkyl optionally substituted by oxo, -CN or halogen; R2 is
3 is absent; and R5 is selected from
Figure imgf000066_0001
wherein the wavy line represents the point of attachment in formula I.
In another embodiment, R1 is piperidinyl optionally substituted by Ci_3 alkylene or Ci_3 alkyl; R2 is Ci_3 alkyl optionally substituted by oxo; R3 is Ci_6 alkyl optionally substituted by oxo, halogen or -CN, phenyl or pyridinyl, wherein said phenyl and pyridinyl are independently optionally substituted by halogen or -CN.
Figure imgf000067_0001
Figure imgf000068_0001
Figure imgf000069_0001
Figure imgf000070_0001
Figure imgf000071_0001
Figure imgf000072_0001
Figure imgf000073_0001
Figure imgf000074_0001
Figure imgf000075_0001
Figure imgf000076_0001
Figure imgf000077_0001
Figure imgf000078_0001
Figure imgf000079_0001
Figure imgf000080_0001
Figure imgf000081_0001
Another embodiment includes a compound selected from Examples 1-56.
Figure imgf000082_0001
Figure imgf000083_0001
Figure imgf000084_0001
Figure imgf000085_0001
and stereoisomers, tautomers and pharmaceutically acceptable salts thereof..
In one embodiment, the compound of formula I is not:
Figure imgf000085_0002
In one embodiment -R'-R^R3 taken together are not morpholino. In one embodiment -R'-R^R3 taken together are not morpholino when R5 is ethoxymethyl.
In one embodiment R5 is not ethoxymethyl.
Compounds of the invention may contain one or more asymmetric carbon atoms. Accordingly, the compounds may exist as diastereomers, enantiomers or mixtures thereof. The syntheses of the compounds may employ racemates, diastereomers or enantiomers as starting materials or as intermediates. Mixtures of particular diastereomeric compounds may be separated, or enriched in one or more particular diastereomers, by chromatographic or crystallization methods. Similarly, enantiomeric mixtures may be separated, or enantiomerically enriched, using the same techniques or others known in the art. Each of the asymmetric carbon or nitrogen atoms may be in the R or S configuration and both of these configurations are within the scope of the invention.
Another aspect includes prodrugs of the compounds of formula I, including known amino -protecting and carboxy-protecting groups which are released, for example hydrolyzed, to yield the compound of formula I under physiologic conditions. A particular class of prodrugs are compounds in which a nitrogen atom in an amino, amidino, amino alky leneamino, iminoalkyleneamino or guanidino group is substituted with a hydroxy (OH) group, an alkylcarbonyl (-CO-R) group, an alkoxycarbonyl (-CO-OR), an acyloxyalkyl-alkoxycarbonyl (- CO-O-R-O-CO-R) group where R is a monovalent or divalent group, for example alkyl, alkylene or aryl, or a group having the formula -C(0)-0-CPlP2-haloalkyl, where PI and P2 are the same or different and are hydrogen, alkyl, alkoxy, cyano, halogen, alkyl or aryl. In a particular embodiment, the nitrogen atom is one of the nitrogen atoms of the amidino group of the compounds of formula I. Prodrugs may be prepared by reacting a compound of formula I with an activated group, such as acyl groups, to bond, for example, a nitrogen atom in the compound of formula I to the exemplary carbonyl of the activated acyl group. Examples of activated carbonyl compounds are those containing a leaving group bonded to the carbonyl group, and include, for example, acyl halides, acyl amines, acyl pyridinium salts, acyl alkoxides, acyl phenoxides such as p-nitrophenoxy acyl, dinitrophenoxy acyl, fluorophenoxy acyl, and difluorophenoxy acyl. The reactions are generally carried out in inert solvents at reduced temperatures such as -78 to about 50°C. The reactions may also be carried out in the presence of an inorganic base, for example potassium carbonate or sodium bicarbonate, or an organic base such as an amine, including pyridine, trimethylamine, triethylamine, triethanolamine, or the like. SYNTHESIS OF JAK1 INHIBITOR COMPOUNDS
Compounds of formula I may be synthesized by synthetic routes described herein. In certain embodiments, processes well-known in the chemical arts can be used, in addition to, or in light of, the description contained herein. The starting materials are generally available from commercial sources such as Aldrich Chemicals (Milwaukee, Wis.) or are readily prepared using methods well known to those skilled in the art (e.g., prepared by methods generally described in Louis F. Fieser and Mary Fieser, Reagents for Organic Synthesis, v. 1-19, Wiley, N.Y. (1967- 1999 ed.), Beilsteins Handbuch der organischen Chemie, 4, Aufl. ed. Springer- Verlag, Berlin, including supplements (also available via the Beilstein online database)), or Comprehensive Heterocyclic Chemistry, Editors Katrizky and Rees, Pergamon Press, 1984.
Compounds of formula I may be prepared singly or as compound libraries comprising at least 2, for example 5 to 1,000 compounds, or 10 to 100 compounds of formula I. Libraries of compounds of formula I may be prepared by a combinatorial "split and mix" approach or by multiple parallel syntheses using either solution phase or solid phase chemistry, by procedures known to those skilled in the art. Thus according to a further aspect of the invention there is provided a compound library comprising at least 2 compounds of formula I, enantiomers, diastereomers, tautomers or pharmaceutically acceptable salts thereof.
For illustrative purposes, reaction schemes 1-10 depicted below provide routes for synthesizing the compounds of the present invention as well as key intermediates. For a more detailed description of the individual reaction steps, see the Examples section below. Those skilled in the art will appreciate that other synthetic routes may be used to synthesize the inventive compounds. Although specific starting materials and reagents are depicted in the Schemes and discussed below, other starting materials and reagents can be easily substituted to provide a variety of derivatives and/or reaction conditions. In addition, many of the compounds prepared by the methods described below can be further modified in light of this disclosure using conventional chemistry well known to those skilled in the art.
In the preparation of compounds of the present invention, protection of remote functionality (e.g., primary or secondary amine) of intermediates may be necessary. The need for such protection will vary depending on the nature of the remote functionality and the conditions of the preparation methods. Suitable amino -protecting groups (NH-Pg) include acetyl, trifluoro acetyl, benzyl, phenylsulfonyl, t-butoxycarbonyl (BOC), benzyloxycarbonyl (CBz) and 9-fluorenylmethyleneoxycarbonyl (Fmoc). The need for such protection is readily determined by one skilled in the art. For a general description of protecting groups and their use, see T. W. Greene, Protective Groups in Organic Synthesis, John Wiley & Sons, New York, 1991.
Compounds of the invention may be prepared from readily available starting materials using the general methods illustrated in Reaction Schemes 1-10 below.
Reaction Scheme 1
General Quinoline synthesis
Figure imgf000088_0001
Reaction Scheme 2
General Naphthyridine synthesis
Figure imgf000088_0002
Reaction Scheme 3
Introducing hindered
Figure imgf000089_0001
Reaction Scheme 4
Introducing C2 amines route I
Figure imgf000089_0002
X = CorN X = CorN n = 1, 2 or 3 n = 1, 2 or 3
MeS02CI, Et3N,
THF
Figure imgf000089_0003
X = C or N
n = 1, 2 or 3 Reaction Scheme 5
Introducing C2 amines route II
Figure imgf000090_0001
X = C or N X = C or N n = 1 , 2 or 3
Figure imgf000090_0002
X = C or N X = C or N
n = 1 , 2 or 3 n = 1 , 2 or 3
Reaction Scheme 6
Introducing C2 amines route III
Figure imgf000090_0003
X = C or N X = C or N
Figure imgf000090_0004
X = C or N X = C or N X = C or N
Reaction Scheme 7 Synthesis of protected amino-cycloalkyls route I
Figure imgf000091_0001
Reaction Scheme 8
Synthesis of protected amino-cycloalkyls route II
Figure imgf000091_0002
Figure imgf000091_0003
Reaction Scheme 9
Synthesis of boc protected piperidine
Figure imgf000092_0001
X = C or N
n = 0 or 1
Reaction Scheme 10
Synthesis of benzyl protected piperidine
Figure imgf000092_0002
X = C or N X = C or N It will be appreciated that where appropriate functional groups exist, compounds of various formulae or any intermediates used in their preparation may be further derivatized by one or more standard synthetic methods employing condensation, substitution, oxidation, reduction, or cleavage reactions. Particular substitution approaches include conventional alkylation, arylation, heteroarylation, acylation, sulfonylation, halogenation, nitration, formylation and coupling procedures.
METHODS OF SEPARATION
In each of the exemplary Schemes it may be advantageous to separate reaction products from one another and/or from starting materials. The desired products of each step or series of steps is separated and/or purified (hereinafter separated) to the desired degree of homogeneity by the techniques common in the art. Typically such separations involve multiphase extraction, crystallization or trituration from a solvent or solvent mixture, distillation, sublimation, or chromatography. Chromatography can involve any number of methods including, for example: reverse-phase and normal phase; size exclusion; ion exchange; supercritical fluid; high, medium, and low pressure liquid chromatography methods and apparatus; small scale analytical; simulated moving bed (SMB) and preparative thin or thick layer chromatography, as well as techniques of small scale thin layer and flash chromatography.
Another class of separation methods involves treatment of a mixture with a reagent selected to bind to or render otherwise separable a desired product, unreacted starting material, reaction by product, or the like. Such reagents include adsorbents or absorbents such as activated carbon, molecular sieves, ion exchange media, or the like. Alternatively, the reagents can be acids in the case of a basic material, bases in the case of an acidic material, binding reagents such as antibodies, binding proteins, selective chelators such as crown ethers, liquid/liquid ion extraction reagents (LIX), or the like. Selection of appropriate methods of separation depends on the nature of the materials involved. Example separation methods include boiling point, and molecular weight in distillation and sublimation, presence or absence of polar functional groups in chromatography, stability of materials in acidic and basic media in multiphase extraction, and the like. One skilled in the art will apply techniques most likely to achieve the desired separation. Diastereomeric mixtures can be separated into their individual diastereo isomers on the basis of their physical chemical differences by methods well known to those skilled in the art, such as by chromatography and/or fractional crystallization. Enantiomers can be separated by converting the enantiomeric mixture into a diastereomeric mixture by reaction with an appropriate optically active compound (e.g., chiral auxiliary such as a chiral alcohol or Mosher's acid chloride), separating the diastereoisomers and converting (e.g., hydrolyzing) the individual diastereoisomers to the corresponding pure enantiomers. Also, some of the compounds of the present invention may be atropisomers (e.g., substituted biaryls) and are considered as part of this invention. Enantiomers can also be separated by use of a chiral HPLC column or supercritical fluid chromatography.
A single stereoisomer, e.g. an enantiomer, substantially free of its stereoisomer may be obtained by resolution of the racemic mixture using a method such as formation of diastereomers using optically active resolving agents (Eliel, E. and Wilen, S., Stereochemistry of Organic Compounds, John Wiley & Sons, Inc., New York, 1994; Lochmuller, C. H., J. Chromatogr., 113(3):283-302 (1975)). Racemic mixtures of chiral compounds of the invention can be separated and isolated by any suitable method, including: (1) formation of ionic, diastereomeric salts with chiral compounds and separation by fractional crystallization or other methods, (2) formation of diastereomeric compounds with chiral derivatizing reagents, separation of the diastereomers, and conversion to the pure stereoisomers, and (3) separation of the substantially pure or enriched stereoisomers directly under chiral conditions. See: Drug Stereochemistry, Analytical Methods and Pharmacology, Irving W. Wainer, Ed., Marcel Dekker, Inc., New York (1993).
Diastereomeric salts can be formed by reaction of enantiomerically pure chiral bases such as brucine, quinine, ephedrine, strychnine, a-methyl- -phenylethylamine (amphetamine), and the like with asymmetric compounds bearing acidic functionality, such as carboxylic acid and sulfonic acid. The diastereomeric salts may be induced to separate by fractional crystallization or ionic chromatography. For separation of the optical isomers of amino compounds, addition of chiral carboxylic or sulfonic acids, such as camphorsulfonic acid, tartaric acid, mandelic acid, or lactic acid can result in formation of the diastereomeric salts.
Alternatively, the substrate to be resolved is reacted with one enantiomer of a chiral compound to form a diastereomeric pair (Eliel, E. and Wilen, S., Stereochemistry of Organic Compounds, John Wiley & Sons, Inc., New York, 1994, p. 322). Diastereomeric compounds can be formed by reacting asymmetric compounds with enantiomerically pure chiral derivatizing reagents, such as menthyl derivatives, followed by separation of the diastereomers and hydrolysis to yield the pure or enriched enantiomer. A method of determining optical purity involves making chiral esters, such as a menthyl ester, e.g. (-) menthyl chloroformate in the presence of base, or Mosher ester, a-methoxy-a-(trifluoromethyl)phenyl acetate (Jacob, J. Org. Chem. 47:4165 (1982)), of the racemic mixture, and analyzing the NMR spectrum for the presence of the two atropisomeric enantiomers or diastereomers. Stable diastereomers of atropisomeric compounds can be separated and isolated by normal- and reverse-phase chromatography following methods for separation of atropisomeric naphthyl-isoquino lines (WO 96/15111). By method (3), a racemic mixture of two enantiomers can be separated by chromatography using a chiral stationary phase {Chiral Liquid Chromatography W. J. Lough, Ed., Chapman and Hall, New York, (1989); Okamoto, J. of Chromatogr. 513:375-378 (1990)). Enriched or purified enantiomers can be distinguished by methods used to distinguish other chiral molecules with asymmetric carbon atoms, such as optical rotation and circular dichroism. The absolute stereochemistry of chiral centers and enatiomers can be determined by x-ray crystallography.
Positional isomers, for example E and Z forms, of compounds of formula I, and intermediates for their synthesis, may be observed by characterization methods such as NMR and analytical HPLC. For certain compounds where the energy barrier for interconversion is sufficiently high, the E and Z isomers may be separated, for example by preparatory HPLC.
PHARMACEUTICAL COMPOSITIONS AND ADMINISTRATION
Another embodiment provides pharmaceutical compositions or medicaments containing the compounds of the invention and a therapeutically inert carrier, diluent or excipient, as well as methods of using the compounds of the invention to prepare such compositions and medicaments. In one example, compounds of formula I may be formulated by mixing at ambient temperature at the appropriate pH, and at the desired degree of purity, with physiologically acceptable carriers, i.e., carriers that are non-toxic to recipients at the dosages and concentrations employed into a galenical administration form. The pH of the formulation depends mainly on the particular use and the concentration of compound, but preferably ranges anywhere from about 3 to about 8. In one example, a compound of formula I is formulated in an acetate buffer, at pH 5. In another embodiment, the compounds of formula I are sterile. The compound may be stored, for example, as a solid or amorphous composition, as a lyophilized formulation or as an aqueous solution.
Compositions are formulated, dosed, and administered in a fashion consistent with good medical practice. Factors for consideration in this context include the particular disorder being treated, the particular mammal being treated, the clinical condition of the individual patient, the cause of the disorder, the site of delivery of the agent, the method of administration, the scheduling of administration, and other factors known to medical practitioners.
In one example, the therapeutically effective amount of the compound of the invention administered parenterally per dose will be in the range of about 0.01-100 mg/kg, alternatively about 0.1 to 20 mg/kg of patient body weight per day, with the typical initial range of compound used being 0.3 to 15 mg/kg/day. In another embodiment, oral unit dosage forms, such as tablets and capsules, contain from about 5 to about 100 mg of the compound of the invention.
The compounds of the invention may be administered by any suitable means, including oral, topical (including buccal and sublingual), rectal, vaginal, transdermal, parenteral, subcutaneous, intraperitoneal, intrapulmonary, intradermal, intrathecal, inhaled and epidural and intranasal, and, if desired for local treatment, intralesional administration. Parenteral infusions include intramuscular, intravenous, intraarterial, intraperitoneal, or subcutaneous administration.
The compounds of the present invention may be administered in any convenient administrative form, e.g., tablets, powders, capsules, solutions, dispersions, suspensions, syrups, sprays, vapors, suppositories, gels, emulsions, patches, etc. Such compositions may contain components conventional in pharmaceutical preparations, e.g., diluents, carriers, pH modifiers, sweeteners, bulking agents, and further active agents.
A typical formulation is prepared by mixing a compound of the present invention and a carrier or excipient. Suitable carriers and excipients are well known to those skilled in the art and are described in detail in, e.g., Ansel, Howard C, et al, Ansel's Pharmaceutical Dosage Forms and Drug Delivery Systems. Philadelphia: Lippincott, Williams & Wilkins, 2004; Gennaro, Alfonso R., et al. Remington: The Science and Practice of Pharmacy. Philadelphia: Lippincott, Williams & Wilkins, 2000; and Rowe, Raymond C. Handbook of Pharmaceutical Excipients. Chicago, Pharmaceutical Press, 2005. The formulations may also include one or more buffers, stabilizing agents, surfactants, wetting agents, lubricating agents, emulsifiers, suspending agents, preservatives, antioxidants, opaquing agents, glidants, processing aids, colorants, sweeteners, perfuming agents, flavoring agents, diluents and other known additives to provide an elegant presentation of the drug (i.e., a compound of the present invention or pharmaceutical composition thereof) or aid in the manufacturing of the pharmaceutical product (i.e., medicament). An example of a suitable oral dosage form is a tablet containing about 2 mg, 5 mg, 25mg, 50mg, lOOmg, 250mg, or 500mg of the compound of the present invention compounded with about 95-30 mg anhydrous lactose, about 5-40 mg sodium croscarmellose, about 5-30mg polyvinylpyrrolidone (PVP) K30, and about e.g., 1-10 mg magnesium stearate. The powdered ingredients are first mixed together and then mixed with a solution of the PVP. The resulting composition can be dried, granulated, mixed with the magnesium stearate and compressed to tablet form using conventional equipment. An example of an aerosol formulation can be prepared by dissolving the compound of the present invention, for example 5-400 mg, in a suitable buffer solution, e.g. a phosphate buffer, adding a tonicifier, e.g. a salt such sodium chloride, if desired. The solution may be filtered, e.g. using a 0.2 micron filter, to remove impurities and contaminants.
An embodiment, therefore, includes a pharmaceutical composition comprising a compound of formula I, stereoisomers, tautomers or pharmaceutically acceptable salts thereof. In a further embodiment includes a pharmaceutical composition comprising a compound of formula I, or stereoisomers, tautomers or pharmaceutically acceptable salts thereof, together with a pharmaceutically acceptable carrier or excipient.
Another embodiment includes a pharmaceutical composition comprising a compound of formula I stereoisomers, tautomers or pharmaceutically acceptable salts thereof for use in the treatment of a hyperproliferative disease. Another embodiment includes a pharmaceutical composition comprising a compound of formula I stereoisomers, tautomers or pharmaceutically acceptable salts thereof for use in the treatment of cancer. Another embodiment includes a pharmaceutical composition comprising a compound of formula I stereoisomers, tautomers or pharmaceutically acceptable salts thereof for use in the treatment of an immunological disorder. Another embodiment includes a pharmaceutical composition comprising a compound of formula I stereoisomers, tautomers or pharmaceutically acceptable salts thereof for use in the treatment of rheumatoid arthritis, psoriasis, inflammatory bowel disease (IBD) or asthma. Another embodiment includes a pharmaceutical composition comprising a compound of formula I stereoisomers, tautomers or pharmaceutically acceptable salts thereof for use in the treatment of rheumatoid arthritis, asthma, systemic lupus erythematosus, psoriasis, IBD and transplant rejection.
METHODS OF TREATMENT WITH AND USES OF JAK1 INHIBITORS The compounds of Formula I inhibit the activity of JAK1 kinase. Accordingly, the compounds of Formula I inhibit the phosphorylation of signal transducers and activators of transcription (STATs) by JAK1 kinase as well as STAT mediated cytokine production. Compounds of Formula I are useful for inhibiting JAK1 kinase activity in cells through cytokine pathways, such as IL-6, IL-15, IL-7, IL-2, IL-4, IL-9, IL-10, IL-13, IL-21, G-CSF, IFNalpha, IFNbeta or IFNgamma pathways. The compounds of Formula I can be used for the treatment of immunological disorders driven by aberrant IL-6, IL-15, IL-7, IL-2, IL-4, IL9, IL-10, IL-13, IL- 21, G-CSF, IFNalpha, IFNbeta or IFNgamma cytokine signaling.
Another embodiment includes a method of treating or lessening the severity of a disease or condition responsive to the inhibition of JAK1 kinase activity in a patient. The method includes the step of administering to a patient a therapeutically effective amount of a compound of the present invention.
In one embodiment, the disease or condition is cancer, stroke, diabetes, hepatomegaly, cardiovascular disease, multiple sclerosis, Alzheimer's disease, cystic fibrosis, viral disease, autoimmune diseases, atherosclerosis, restenosis, psoriasis, rheumatoid arthritis, inflammatory bowel disease, asthma, allergic disorders, inflammation, neurological disorders, a hormone- related disease, conditions associated with organ transplantation, immunodeficiency disorders, destructive bone disorders, proliferative disorders, infectious diseases, conditions associated with cell death, thrombin-induced platelet aggregation, liver disease, pathologic immune conditions involving T cell activation, CNS disorders or a myeloproliferative disorder.
In one embodiment, the disease or condition is cancer.
In one embodiment, the disease is a myeloproliferative disorder.
In one embodiment, the myeloproliferative disorder is polycythemia vera, essential thrombocytosis, myelofibrosis or chronic myelogenous leukemia (CML). In one embodiment, the cancer is breast, ovary, cervix, prostate, testis, penile, genitourinary tract, seminoma, esophagus, larynx, gastric, stomach, gastrointestinal, skin, kerato acanthoma, follicular carcinoma, melanoma, lung, small cell lung carcinoma, non-small cell lung carcinoma (NSCLC), lung adenocarcinoma, squamous carcinoma of the lung, colon, pancreas, thyroid, papillary, bladder, liver, biliary passage, kidney, bone, myeloid disorders, lymphoid disorders, hairy cells, buccal cavity and pharynx (oral), lip, tongue, mouth, salivary gland, pharynx, small intestine, colon, rectum, anal, renal, prostate, vulval, thyroid, large intestine, endometrial, uterine, brain, central nervous system, cancer of the peritoneum, hepatocellular cancer, head cancer, neck cancer, Hodgkin's or leukemia.
In one embodiment, the cardiovascular disease is restenosis, cardiomegaly, atherosclerosis, myocardial infarction or congestive heart failure. In one embodiment, the neurodegenerative disease is Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, Huntington's disease, and cerebral ischemia, and neurodegenerative disease caused by traumatic injury, glutamate neurotoxicity or hypoxia.
In one embodiment, the inflammatory diseases is rheumatoid arthritis, psoriasis, asthma, inflammatory bowel disease, contact dermatitis or delayed hypersensitivity reactions. In one embodiment, the autoimmune disease is lupus or multiple sclerosis.
In one embodiment, the disease or condition responsive to the inhibition of JAK1 kinase is rheumatoid arthritis.
In one embodiment, the disease or condition responsive to the inhibition of JAK1 kinase is rheumatoid arthritis, asthma, systemic lupus erythematosus, psoriasis, IBD or transplant rejection.
Another embodiment includes a method of treating cancer in a mammal in need of such treatment, wherein the method comprises administering to said mammal a therapeutically effective amount of a compound of formula I, a stereoisomer, tautomer, prodrug or pharmaceutically acceptable salt thereof. Another embodiment includes compounds of formula I, a stereoisomer, tautomer, prodrug or pharmaceutically acceptable salt thereof, for use in therapy. In another embodiment, the therapy is the treatment of an immunological disorder, for example rheumatoid arthritis. In another embodiment, the therapy is the treatment of cancer.
Another embodiment includes compounds of formula I, a stereoisomer, tautomer, prodrug or pharmaceutically acceptable salt thereof, for use in treating a disease selected from rheumatoid arthritis, asthma, systemic lupus erythematosus, psoriasis, IBD and transplant rejection. Another embodiment includes the use of a compound of formulas I, a stereoisomer, tautomer, prodrug or pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of a disease described herein (e.g., cancer or immunological disorder). COMBINATION THERAPY
The compounds of formula I may be employed alone or in combination with other chemotherapeutic agents for treatment. The compounds of the present invention can be used in combination with one or more additional drugs, for example an anti-hyperproliferative, anticancer, cytostatic, cytotoxic, anti-inflammatory or chemotherapeutic agent. The second compound of the pharmaceutical combination formulation or dosing regimen preferably has complementary activities to the compound of this invention such that they do not adversely affect each other. Such agents are suitably present in combination in amounts that are effective for the purpose intended. The compounds may be administered together in a unitary pharmaceutical composition or separately and, when administered separately this may occur simultaneously or sequentially. Such sequential administration may be close or remote in time. In one embodiment, compounds of the present invention are coadministered with a cytostatic compound selected from the group consisting of cisplatin, doxorubicin, taxol, taxotere and mitomycin C. In another embodiment, the cytostatic compound is doxorubicin. In another embodiment, compounds of the present invention are coadministered with an anti-inflammatory agent selected from a NSAID and corticosteroid. In another embodiment, compounds of the present invention are coadministered with an anti-rheumatoid agent, in one example, RITUXAN®. In another embodiment, compounds of the present invention are coadministered with a chemotherapeutic agent selected from etanercept (Enbrel), infliximab (Remicade), adalimumab (Humira), certolizumab pegol (Cimzia), golimumab (Simponi), Interleukin 1 (IL-1) blockers such as anakinra (Kineret), monoclonal antibodies against B cells such as rituximab (RITUXAN®), T cell costimulation blockers such as abatacept (Orencia), Interleukin 6 (IL-6) blockers such as tocilizumab (ACTEMERA®); Interleukin 13 (IL-13) blockers such as lebrikizumab; Interferon alpha (IFN) blockers such as Rontalizumab; Beta 7 integrin blockers such as rhuMAb Beta7; IgE pathway blockers such as Anti-Mi prime; Secreted homotrimeric LTa3 and membrane bound heterotrimer LTal/p2 blockers such as Anti-lymphotoxin alpha (LTa)
The compounds of the present invention can be also used in combination with radiation therapy. The phrase "radiation therapy" refers to the use of electromagnetic or particulate radiation in the treatment of neoplasia. Radiation therapy delivers doses of radiation sufficiently high to a target area to cause death of reproducing cells, in both tumor and normal tissues. The radiation dosage regimen is generally defined in terms of radiation absorbed dose (rad), time and fractionation, and must be carefully defined by the oncologist. The amount of radiation a patient receives will depend on various considerations but two of the most important considerations are the location of the tumor in relation to other critical structures or organs of the body, and the extent to which the tumor has spread. Examples of radiotherapeutic agents are provided in Hellman, Principles of Radiation Therapy, Cancer, in Principles I and Practice of Oncology, 24875 (Devita et al, 4th ed., vol 1, 1993). Alternative forms of radiation therapy include three- dimensional conformal external beam radiation, intensity modulated radiation therapy (IMRT), stereotactic radiosurgery and brachytherapy (interstitial radiation therapy), the latter placing the source of radiation directly into the tumor as implanted "seeds". These alternative treatment modalities deliver greater doses of radiation to the tumor, which accounts for their increased effectiveness when compared to standard external beam radiation therapy. Another embodiment includes a method of preparing a compound of formula I or a pharmaceutically acceptable salt thereof comprising: a, for a compound of formula I wherein Y is CR5, cyclizing a correspond
compound of formula 100:
Figure imgf000101_0001
to provide the compound of formula I; b contacting a compound of formula I with an acid or base to provide the corresponding pharmaceutically acceptable salt; c removing a protecting group from a corresponding compound bearing a protecting group to provide the compound of formula I; d, for a compound of formula I wherein R2 and R3 are not each absent, converting a compound of formula 101:
Figure imgf000102_0001
to the corresponding compound of formula I wherein R2 and R3 are not each absent; e. for a compound of formula I wherein -R1-R2-R3 taken together are:
Figure imgf000102_0002
reacting a corresponding compound of formula I wherein -R1-R2-R3 taken together are:
Figure imgf000102_0003
with a compound of formula Ra-Lg, wherein Lg is a suitable leaving group, to provide the compound of formula I; f. for a compound of formula I wherein -R1-R2-R3 taken together are:
Figure imgf000102_0004
reacting a corresponding compound of formula I wherein -R1-R2-R3 taken together are:
Figure imgf000102_0005
with a compound of formula Ra-Lg, wherein Lg is a suitable leaving group, to provide the compound of formula I; g. for a compound of formula I wherein -R1 is:
Figure imgf000102_0006
removing a protecting group Pg from a corresponding compound of formula I wherein -R1 is
Figure imgf000103_0001
to provide the compound of formula I; or h. for a compound of formula I wherein -R1 is:
Figure imgf000103_0002
removing a protecting group Pg from a corresponding compound of formula I wherein -R1 is
Figure imgf000103_0003
to provide the compound of formula I.
ARTICLES OF MANUFACTURE
Another embodiment includes a kit for treating a disease or disorder responsive to the inhibition of JAK1 kinase. The kit includes: (a) a first pharmaceutical composition comprising a compound of formula I; and
(b) instructions for use.
In another embodiment, the kit further includes:
(c) a second pharmaceutical composition, which includes a chemotherapeutic agent.
In one embodiment, the instructions describe the simultaneous, sequential or separate administration of said first and second pharmaceutical compositions to a patient in need thereof. In one embodiment, the first and second compositions are contained in separate containers.
In one embodiment, the first and second compositions are contained in the same container. Containers for use include, for example, bottles, vials, syringes, blister pack, etc. The containers may be formed from a variety of materials such as glass or plastic. The container includes a compound of formula I or formulation thereof which is effective for treating the condition and may have a sterile access port (for example the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle). The container includes a composition comprising at least one compound of formula I. The label or package insert indicates that the composition is used for treating the condition of choice, such as cancer. In one embodiment, the label or package inserts indicates that the composition comprising the compound of formula I can be used to treat a disorder. In addition, the label or package insert may indicate that the patient to be treated is one having a disorder characterized by overactive or irregular kinase activity. The label or package insert may also indicate that the composition can be used to treat other disorders.
The article of manufacture may comprise (a) a first container with a compound of formula I contained therein; and (b) a second container with a second pharmaceutical formulation contained therein, wherein the second pharmaceutical formulation comprises a chemotherapeutic agent. The article of manufacture in this embodiment of the invention may further comprise a package insert indicating that the first and second compounds can be used to treat patients at risk of stroke, thrombus or thrombosis disorder. Alternatively, or additionally, the article of manufacture may further comprise a second (or third) container comprising a pharmaceutically-acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer's solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes.
In order to illustrate the invention, the following examples are included. However, it is to be understood that these examples do not limit the invention and are only meant to suggest a method of practicing the invention. Persons skilled in the art will recognize that the chemical reactions described may be readily adapted to prepare other compounds of formula I, and alternative methods for preparing the compounds of formula I are within the scope of this invention. For example, the synthesis of non-exemplified compounds according to the invention may be successfully performed by modifications apparent to those skilled in the art, e.g., by appropriately protecting interfering groups, by utilizing other suitable reagents known in the art other than those described, and/or by making routine modifications of reaction conditions. Alternatively, other reactions disclosed herein or known in the art will be recognized as having applicability for preparing other compounds of the invention.
EXAMPLES
The invention will be more fully understood by reference to the following examples. They should not, however, be construed as limiting the scope of the invention. Abbreviations used herein are as follows:
Abbreviations:
AcOH Acetic acid
aq. Aqueous
Bn Benzyl
Boc20 Di-tert-butyl dicarbonate
Brine Saturated aqueous sodium chloride solution
CAS Chemical abstract service number
CDCI3 Deuterated chloroform
DCM Dichloromethane
DIAD Diisopropyl azodicarboxylate
DIPEA Diisopropylethylamine / iPr2EtN
DMAP 4-(Dimethylamino)pyridine
DMSO Dimethylsulfoxide
DMSO-d6 Deuterated DMSO
DMF Dimethylformamide
EDCI l-Ethyl-3-(3-dimethylaminopropyl) carbodiimide hydrochloride eq equivalents
ESI Electrospray
Et Ethyl
EtOAc Ethyl acetate EtOH Ethanol
Et3N Triethylamine
Et20 Diethyl ether
Fe Iron
h Hour
hr Hour
HC02H Formic acid
HC1 Hydrochloric acid
HM-N Isolute® HM-N is a modified form of diatomaceous earth HOBt Hydro xybenzotriazole
HPLC High performance liquid chromatography
IMS Industrial methylated spirit
IPA Isopropyl alcohol / propan-2-ol
iPr Isopropyl
Me Methyl
MeCN Acetonitrile
MeOH Methanol
MeOD Deuterated methanol
MgSC"4 Magnesium sulfate
min minute / minutes
NaOH Sodium Hydroxide
Na2S04 Sodium sulfate
NaHCC"3 Sodium bicarbonate / Sodium hydrogen carbonate
NaOH Sodium hydroxide
NEt3 Triethylamine
N¾ Ammonia
NH4C1 Ammonium chloride
Pd Palladium
Pd on C Palladium on Carbon Pr Propyl
p-TsOH /?ara-toluenesulfonic acid
RT Retention time in minutes (for LCMS)
RT Room temperature (for reaction conditions)
SCX-2 Pre-packed Isolute® silica-based sorbent with a chemically
bonded propylsulfonic acid functional group
TEA Triethylamine
TFA Trifluoro acetic acid
THF Tetrahydrofuran
TLC Thin layer chromatography
TMSC1 Trimethylsilyl chloride
General Experimental Conditions:
All temperatures are in degrees Celsius (°C). Unless otherwise stated, operations were carried out at room or ambient temperature (18-25 °C).
Unless otherwise stated, the solvents used in preparing the example compounds were commercial anhydrous grade and were used without further drying or purification.
1H NMR spectra were recorded at ambient temperature or at 80 °C where indicated using one of the following machines: Varian Unity Inova (400MHz) spectrometer with a triple resonance 5mm probe, Bruker Avance DRX400 (400MHz) spectrometer with a triple resonance 5mm probe, a Bruker Avance DPX 300 (300MHz) equipped with a standard 5mm dual frequency probe for detection of 1H and 13C, a Bruker AVIII (400 MHz) using a BBI Broad Band Inverse 5mm probe, or a Bruker AVIII (500 MHz) using a QNP ( Quad Nucleus detect) 5mm probe. Chemical shifts are expressed in ppm relative to an internal standard; tetramethylsilane (ppm = 0.00). The following abbreviations have been used: br = broad signal, s = singlet, d = doublet, dd = double doublet, t = triplet, q = quartet, m = multiplet, or any combination of. High Pressure Liquid Chromatography - Mass Spectrometry (LCMS) experiments to determine retention times (RT) and associated mass ions (m+H) were performed using one of the following methods:
Method A: Experiments performed on a Waters Micromass ZQ2000 quadrupole mass spectrometer linked to a Waters Acquity UPLC system with a PDA UV detector. The spectrometer has an electrospray source operating in positive and negative ion mode. This system uses an Acquity BEH CI 8 1.7um 100 x 2.1mm column, maintained at 40°C or an Acquity BEH Shield RP18 1.7 μηι 100 x 2.1mm column, maintained at 40°C and a 0.4 ml / minute flow rate. The initial solvent system was 95% water containing 0.1% formic acid (solvent A) and 5% acetonitrile containing 0.1% formic acid (solvent B) for the first 0.4 minute followed by a gradient up to 5% solvent A and 95% solvent B over the next 5.6 minutes. This was maintained for 0.8 minute before returning to 95% solvent A and 5% solvent B over the next 1.2 minutes. Total run time was 8 minutes.
Method B: Experiments performed on a Finnigan AQA single quadrupole mass spectrometer linked to a Hewlett Packard 1050 LC system with UV diode array detector and autosampler. The spectrometer has an electrospray source operating in positive ion mode. Additional detection is achieved using a Sedex 65 evaporative light scattering detector. This system uses a Luna 3 micron CI 8(2) 30 x 4.6mm column at ambient temperature and a 2.0 ml / minute flow rate. The initial solvent system was 95% water containing 0.1% formic acid (solvent A) and 5% acetonitrile containing 0.1% formic acid (solvent B) for the first 0.5 minute followed by a gradient up to 5% solvent A and 95% solvent B over the next 4.0 minutes. This was maintained for 1.0 minute before returning to 95% solvent A and 5% solvent B over the next 0.5 minute. Total run time was 6 minutes.
Method C: The system consists of a Waters Quattro Micro triple quadrupole mass spectrometer linked to a Hewlett Packard HP 1100 LC system with a PDA UV detector. Sample injection is done by a CTC HTS PAL autosampler. The spectrometer has an electrospray source operating in positive and negative ion mode. This system uses a Higgins Clipeus 5micron CI 8 100 x 3.0mm column at ambient temperature and a 2.0 mL / minute flow rate. The initial solvent system was 95% water containing 0.1% formic acid (solvent A) and 5% acetonitrile containing 0.1% formic acid (solvent B) for the first 0.5 minute followed by a gradient up to 5% solvent A and 95% solvent B over the next 14 minutes. This was maintained for 5 minutes before returning to 95% solvent A and 5% solvent B over the next 2 minutes. Total run time was 25 minutes. Method D:
Waters Acquity UPLC Mobile phase A H20 with 0.1%Formic Acid Mobile phase B Acetonitrile with 0.1%Formic Acid Column Acquity UPLC BEH C18, 1.7μιη, 2.1
Column temperature 40 degree C LC gradient 5-95%B in 1.4 min, 95% in 0.3 min LC Flow rate 800uL/min
UV wavelength 220nm and 254nm
Mass Spec - Waters SQ Detector Ionization ESI+
Scan range 100-800amu
Method E: Compounds were analysed using the following conditions: Experiments were performed on a The system consists of a Waters ZMD single quadrupole mass spectrometer linked to a Hewlett Packard HP 1100 LC system with UV diode array detector and 100 position autosampler. The spectrometer has an electrospray source operating in positive and negative ion mode. This system uses an Phenomenex Luna 3micron CI 8(2) 30 x 4.6mm column at ambient temperature, and a 2.0 ml / minute flow rate. The initial solvent system was 95% water containing 0.1% formic acid (solvent A) and 5% acetonitrile containing 0.1% formic acid (solvent B) for the first 0.5 minute followed by a gradient up to 5%> solvent A and 95%> solvent B over the next 4 minutes. This was maintained for 1 minute before returning to 95% solvent A and 5% solvent B over the next 0.5 minute. Total run time was 6 minutes.
Method F: Experiments were performed on a Waters Platform LC quadrupole mass spectrometer linked to a Hewlett Packard HP 1100 LC system with diode array detector and 100 position autosampler. The spectrometer has an electrospray source operating in positive and negative ion mode. Additional detection is achieved using a Sedex 85 evaporative light scattering detector. This system uses an Phenomenex Luna 3micron CI 8(2) 30 x 4.6mm column at ambient temperature, and a 2.0 ml / minute flow rate. The initial solvent system was 95% water containing 0.1% formic acid (solvent A) and 5% acetonitrile containing 0.1% formic acid (solvent B) for the first 0.5 minute followed by a gradient up to 5% solvent A and 95% solvent B over the next 4 minutes. This was maintained for 1 minute before returning to 95% solvent A and 5% solvent B over the next 0.5 minute. Total run time was 6 minutes.
Method G: Experiments were performed on a Waters ZMD quadrupole mass spectrometer linked to a Waters 1525 LC system with Waters 996 diode array detector. The spectrometer has an electrospray source operating in positive and negative ion mode. Additional detection is achieved using a Sedex 85 evaporative light scattering detector. This system uses an Luna 3micron CI 8(2) 30 x 4.6mm column at ambient temperature, and a 2.0 ml / minute flow rate. The initial solvent system was 95% water containing 0.1% formic acid (solvent A) and 5% acetonitrile containing 0.1% formic acid (solvent B) for the first 0.5 minute followed by a gradient up to 5% solvent A and 95% solvent B over the next 4 minutes. This was maintained for 1 minute before returning to 95% solvent A and 5% solvent B over the next 0.5 minute. Total run time was 6 minutes.
Reverse Phase High Performance Liquid Chromatography (HPLC) was used to purify compounds where indicated. Unless otherwise indicated, the conditions were: elution on a Phenomenex Gemini CI 8 column (250 x 21.2 mm, 5 micron) as stationary phase and using mobile phase indicated, operating at a 18 mL/min flow rate using a Gilson UV/Vis -155 dual channel detector and Gilson GX-271 automated liquid handler.
Microwave experiments were carried out using a Biotage Initiator 2.0 (400 W MAGNETRON®) which uses a single-mode resonator and dynamic field tuning. Temperature from 40-250°C can be achieved, and pressures of up to 20 bar can be reached.
Intermediate 1
Figure imgf000110_0001
H-CI
(S)-(Tetrahydro-pyran-3-yl)amine hydrochloride (S)-Dimethyl 2-(tert-butoxycarbonylamino)pentanedioate
To MeOH (7L) was added TMSC1 slowly at 0 °C, and the mixture was stirred for 30 min, then L-glutamic acid (700 g, 4.76 mol) was added to the mixture. The mixture was stirred at room temperature until complete reaction was observed (monitored by TLC). After cooling to 0 °C, triethylamine (313 g, 31.0 mol) and Boc20 (1.14 Kg, 5.23 mol) were added slowly to the reaction solution successively while keeping the internal temperature below 25 °C, and the resultant solution was stirred for 16 hours. After concentration, the residue was poured into water (5 L) and extracted with ethyl acetate (10 L). The organic phase was washed with 4L of 20% citric acid and brine, and dried over sodium sulfate. After filtration and concentration, the crude (S)-dimethyl 2-(tert-butoxycarbonylamino)pentanedioate (1.30 Kg) was obtained as pale yellow oil.
(S)-tert-Butyl 1 ,5-dihydroxypentan-2-ylcarbamate A 20 L reactor was charged with 10 L of dry THF and lithium boro hydride (400 g) and cooled to 0 °C. A solution of (S)-dimethyl 2-(tert-butoxycarbonylamino)pentanedioate (1.00 Kg , 3.63 mol) dissolved in THF (2 L) was then added dropwise while keeping the internal temp, below 15 °C. The mixture was slowly warmed to room temperature and stirred for 16 hours. Cooled to 0 °C, then MeOH (10 L) was added dropwise to the reaction mixture to quench the excess reducing reagent. Concentrated, and the residue was poured into 5L water. After extraction with ethyl acetate, the combined organic phase was dried over sodium sulfate. After filtration the solvent was removed in vacuo to give 730 g of crude (S)-tert-butyl 1,5- dihydroxypentan-2-ylcarbamate as a pale yellow oil.
(S)-tert-Butyl tetrahydro-2H-pyran-3-ylcarbamate A 20-L reactor equipped with a mechanical stirrer was charged with (S)-tert-butyl 1,5- dihydroxypentan-2-ylcarbamate (950 g, 4.33mol), triphenylphosphine (2.27 Kg, 8.66 mol) and DCM (10 L). Then DIAD (1.75 Kg, 8.66 mol) was added dropwise to the reaction solution. The solution was stirred for 48 hours at room temperature until the reaction was complete. After filtration and concentration in vacuo, the residue was purified by column chromatography with petroleum ether as eluent to give 550 g of (S)-tert-butyl tetrahydro-2H-pyran-3-ylcarbamate as white solid. 1H NMR (400 MHz, CDC13) δ 4.77 (m, 1H), 3.78 (d, J = 11.1 Hz, 1H), 3.63 (d, J = 8.8 Hz, 3H), 3.38 (m, 1H), 1.88 (td, J = 8.4, 3.8 Hz, 1H), 1.74 (ddd, J = 10.4, 9.1, 4.9 Hz, 1H), 1.65 - 1.51 (m, 2H), 1.45 (s, 9H).
(S)-(Tetrahydro-pyran-3-yl)amine hydrochloride Intermediate (S)-tert-butyl tetrahydro-2H-pyran-3-ylcarbamate (414 g, 2.06 mol) was added to a 6N HC1 solution of MeOH (4 L) at room temperature, and the reaction mixture was stirred until it was complete (monitored by TLC). After concentration in vacuo, 283 g of (S)- (tetrahydro-pyran-3-yl)amine hydrochloride was obtained as a white solid (yield, 99.8%, enantiomeric excess >99%). 1H NMR (400 MHz, DMSO-d6) δ 8.37 (s, 3H), 3.87 - 3.72 (m, 1H), 3.70 - 3.57 (m, 1H), 3.54 - 3.38 (m, 2H), 3.12 (d, J = 2.1 Hz, 1H), 2.05 - 1.89 (m, 1H), 1.81 - 1.60 (m, 2H), 1.49 (dtd, J= 12.5, 8.3, 4.3 Hz, 1H).
Intermediate 2
Figure imgf000112_0001
(lR,3R)-3-Amino-cyclohexanol
(lR,3R)-3-Amino-cyclohexanol was prepared following the procedures described in P. Bernardelli et al. / Tetrahedron: Asymmetry 15 (2004) 1451-1455.
Intermediate 3
Figure imgf000112_0002
Trans-4-Amino-3-fluoro-piperidine-l-carboxylic acid tert-butyl ester cis and trans 4-Benzylamino-3-fluoro-piperidine-l-carboxylic acid tert-butyl ester A stirred solution of 3-fluoro-4-oxo-piperidine-l-carboxylic acid tert-butyl ester (2.00 g,
9.21 mmol) in methanol (20 mL) and acetic acid (1 mL) was treated with benzyl amine (1.11 mL, 10.0 mmol) and stirred at ambient temperature for 3 hours. The mixture was then treated with sodium cyanoborohydride (868 mg, 13.8 mmol) and left to stand overnight. The reaction was quenched with a saturated solution of sodium bicarbonate (20 mL) and extracted with three portions of dichloro methane (50 mL). Combined organic layers were dried with magnesium sulfate and concentrated under vacuum to give crude product. Purification by column chromatography on silica gel (gradient: 0 to 60% ethyl acetate in heptane) gave 734 mg (26%) of trans- 4-benzylamino-3-fluoro-piperidine-l-carboxylic acid tert-butyl ester and 653 mg (23%) of cis- 4-benzylamino-3-fluoro-piperidine-l-carboxylic acid tert-butyl ester. Trans Data: LCMS (Method D, ESI): RT = 0.67 min, m+H =309.2; 1H NMR (500 MHz, DMSO-d6) δ = 7.33 (s, 5H), 4.44 - 4.31 (m, 1H), 3.84 - 3.65 (m, 3H), 3.53 - 3.41 (m, 1H), 3.17
- 3.07 (m, 1H), 2.80 - 2.68 (m, 1H), 2.34 - 2.28 (m, 1H), 1.85 - 1.74 (m, 1H), 1.39 (s, 9H), 1.36
- 1.30 (m, 1H) Cis Data: LCMS (Method D, ESI): RT = 0.63 min, m+H =309.2; 1H NMR (500 MHz, DMSO- d6) δ = 7.32 (dd, J=18.4, 7.3, 5H), 4.84 - 4.69 (m, 1H), 4.21 - 4.06 (m, 1H), 3.97 - 3.86 (m, 1H), 3.77 (s, 2H), 3.09 - 2.88 (m, 2H), 2.85 - 2.69 (m, 1H), 2.65 - 2.55 (m, 1H), 2.07 - 1.97 (m, 1H), 1.72 - 1.61 (m, 1H), 1.48 - 1.42 (m, 1H), 1.38 (s, 9H).
Trans-4-Amino-3-fluoro-piperidine-l-carboxylic acid tert-butyl ester A stirred solution of trans-4-benzylamino-3-fluoro-piperidine-l-carboxylic acid tert-butyl ester (0.17 g, 0.55 mmol) in methanol (2 mL) and was treated with ammonium formate (139 mg, 2.20 mmol) and 10% palladium on activated carbon (59.0 mg, 55.0 μιηοΐ) and stirred at 50 °C for 1 hour. The mixture was then filtered through Celite®, washed with copious methanol and concentrated under vacuum to give 118 mg (100%) of trans-4-amino-3-fluoro-piperidine-l- carboxylic acid tert-butyl ester that was used without further purification. LCMS (Method D, ESI): RT = 0.14 min, m+H =219.0; 1H NMR (400 MHz, DMSO-d6) δ 4.22 - 4.01 (m, 1H), 3.96
- 3.68 (m, 2H), 3.62 - 3.52 (m, 1H), 3.22 - 2.94 (m, 2H), 2.89 - 2.77 (m, 1H), 1.81 - 1.63 (m, 2H), 1.39 (s, 10H), 1.28 - 1.16 (m, 1H).
Intermediate 4
Figure imgf000113_0001
Trans 4-Amino-cyclohexanecarbonitrile trifluoro acetic acid salt
Following the procedure outlined in WO2009/145719 the title compound was prepared from trans-4-(t-butoxycarbonylamino)-cyclohexane carboxylic acid in three steps with isolation of the TFA salt of trans 4-amino-cyclohexanecarbonitrile via diethyl ether trituration and filtration to afford a white solid in 63% overall yield. 1H NMR (400 MHz, DMSO-d6) δ: 7.93 (s, 3 H), 3.03 (m, 1 H), 2.66 (m, 1 H), 2.05 (m, 2 H), 1.92 (dd, 2 H), 1.58 (m, 2 H), 1.31 (m, 2 H).
Intermediate 5
Figure imgf000114_0001
Trans (4-Amino-cyclohexyl)-acetonitrile
Trans Methanesulfonic acid 4-tert-butoxycarbonylamino-cyclohexylmethyl ester
Trans (4-hydroxymethyl-cyclohexyl)-carbamic acid tert-butyl ester (3.49 g, 15.0 mmol) in DCM (50 mL) was treated with pyridine (4.98 mL, 60.8 mmol). The mixture was cooled to 0 °C and methanesulfonyl chloride (2.36 mL, 30.4 mmol) was added dropwise over 5 minutes. The mixture was stirred at room temperature for 5 hours and then concentrated under vacuum. The residue was partitioned between ethyl acetate and water and the aqueous phase extracted with ethyl acetate (x2). The combined organic extracts were washed with brine, dried with sodium sulfate and concentrated under vacuum. The residue was triturated with cyclohexane then dried under vacuum to give a white solid. Further purification by column chromatography on silica gel (eluting with 1 : 1 cyclohexane/ethyl acetate) gave 4.17 g (90%) of trans methanesulfonic acid 4- tert-butoxycarbonylamino-cyclohexylmethyl ester as a white solid. 1H NMR (400 MHz, CDCI3) δ: 4.37 (br s, 1 H), 4.02 (d, 2 H), 3.39 (br s, 1 H), 3.00 (s, 3 H), 2.11-2.00 (m, 2 H), 1.91-1.81 (m, 2 H), 1.77-1.64 (m, 1 H), 1.43 (s, 9 H), 1.18-1.05 (m, 4 H).
Trans (4-Cyanomethyl-cyclohexyl)-carbamic acid tert-butyl ester
A mixture of trans methanesulfonic acid 4-tert-butoxycarbonylamino-cyclohexylmethyl ester (0.93 mg, 3.00 mmol) and sodium cyanide (0.44 g, 9.00 mmol) in DMSO (10 mL) was stirred at 90 °C for 4 hours. After cooling the mixture was partitioned between ethyl acetate and brine and the aqueous phase extracted with ethyl acetate (x2). The combined organic extracts were washed (water and brine), dried (sodium sulfate) and concentrated under vacuum to give 0.72 g (100%) of trans (4-cyanomethyl-cyclohexyl)-carbamic acid tert-butyl ester as a white solid. 1H NMR (400 MHz, CDCI3) δ: 4.38 (br s, 1 H), 3.45-3.33 (m, 1 H), 2.26 (d, 2 H), 2.10-2.02 (m, 2 H), 1.96-1.87 (m, 2 H), 1.71-1.57 (m, 1 H), 1.45 (s, 9 H), 1.26-1.03 (m, 4 H). Trans (4-Amino-cyclohexyl)-acetonitrile
A solution of trans (4-cyanomethyl-cyclohexyl)-carbamic acid tert-butyl ester (0.71 g, 3.00 mmol) in DCM (15 mL) was treated with trifluoroacetic acid (2 mL) and the mixture was stirred at ambient temperature for 2.5 hours, then concentrated under vacuum. The residue was purified by column chromatography using Isolute® SCX-2 cartridge (eluting with MeOH then 2M NH3 in MeOH) to afford 0.40 g (97%) of trans (4-amino-cyclohexyl)-acetonitrile that was used without further purification. 1H NMR (400 MHz, CDC13) δ: 3.52-3.42 (m, 2 H), 2.69-2.60 (m, 1 H), 2.26 (d, 2 H), 1.94-1.83 (m, 4 H), 1.70-1.57 (m, 1 H), 1.24-1.06 (m, 4 H).
Intermediate 6
Figure imgf000115_0001
Trans 3-(4-Amino-cyclohexyl)-propionitrile trifluoro acetic acid salt
Trans (4-Formyl-cyclohexyl)-carbamic acid tert-butyl ester
A mixture of trans (4-hydroxymethyl-cyclohexyl)-carbamic acid tert-butyl ester (10.0 g, 43.7 mmol) in DCM (225 mL) was treated with dimethylsulfoxide (DMSO) (75 mL) and then cooled to 0 °C. Diisopropylethylamine (30.4 mL, 0.17 mol) was added followed by dropwise addition of a fine suspension of sulphur trioxide pyridine complex (27.8 g, 0.17 mol) in DMSO (75 mL). The mixture was stirred at ambient temperature for 10 minutes and then diluted with diethyl ether and IN aqueous hydrochloric acid with cooling in an ice bath. The phases were separated and the aqueous layer was extracted with ether (x2) and the combined organic phases were washed with IN aqueous hydrochloric acid, and brine, dried with sodium sulfate and concentrated under vacuum. Trituration with pentane gave 9.36 g (94%) of trans (4-formyl- cyclohexyl)-carbamic acid tert-butyl ester as a white solid which was used without further purification. 1H NMR (400 MHz, CDC13) δ: 9.62 (d, 1 H), 4.40 (br s, 1 H), 3.51-3.28 (m, 1 H), 2.20-1.99 (m, 4 H), 1.44 (s, 9 H), 1.38 (m, 2 H), 1.16 (m, 2 H).
Trans E- and Z- [4-(2-Cyano-vinyl)-cyclohexyl]-carbamic acid tert-butyl ester
To a suspension of potassium tert-butoxide (5.54 g, 49.5 mmol) in dry THF (110 mL) at 0 °C, diethyl cyanomethyl phosphonate (8.00 mL, 49.5 mmol) was added slowly dropwise and the mixture was stirred at 0 °C for 1 hour. A solution of trans (4-formyl-cyclohexyl)-carbamic acid tert-butyl ester (9.36 g, 41.2 mmol) in dry THF (290 mL) was added slowly over -15 mins, then the cooling bath was removed and the mixture was stirred at ambient temperature for 1 hour. Ethyl acetate and water were added and the phases were separated. The aqueous phase was extracted with ethyl acetate (x2) and the combined organic phase washed with 10% aqueous citric acid solution, saturated sodium hydrogen carbonate solution, and brine, dried with sodium sulfate and concentrated under vacuum to give 12.6 g of crude trans [4-(2-cyano-vinyl)- cyclohexyl]-carbamic acid tert-butyl ester as a ~ 1 :2 mixture of E- and Z- isomers which was used without further purification. The golden oil crystallized almost immediately and was used in the next step without further purification.
E-isomer: 1H NMR (400 MHz, CDC13) δ: 6.64 (dd, 1 H), 5.29 (dd, 1 H), 4.39 (m, 1 H), 4.26 (m, 1 H), 3.52-3.28 (m, 1 H), 2.08 (m, 2 H), 1.83 (m, 2 H), 1.44 (s, 9 H), 1.35-1.10 (m, 4 H).
Z-isomer: 1H NMR (400 MHz, CDC13) δ: 6.28 (dd, 1 H), 5.25 (dd, 1 H), 4.39 (m, 1 H), 4.26 (m, 1 H), 3.52-3.28 (m, 1 H), 2.08 (m, 2 H), 1.83 (m, 2 H), 1.44 (s, 9 H), 1.35-1.10 (m, 4 H).
Trans [4-(2-Cyano-ethyl)-cyclohexyl]-carbamic acid tert-butyl ester
A mixture of trans E- and Z- [4-(2-cyano-vinyl)-cyclohexyl]-carbamic acid tert-butyl ester (4.00 g, 16.0 mmol) in ethanol (50 mL, IMS grade) under nitrogen was treated with 10% palladium/carbon (1.60 g, 1.50 mmol palladium). The mixture was purged with hydrogen gas and stirred under an atmosphere of hydrogen (balloon) for 64 hours. The mixture was filtered through Celite® and the filtrate was concentrated under vacuum. Purification by column chromatography on silica gel (ethyl acetate/cyclohexane 1 :3) gave 2.68 g (76% over 3 steps) of trans [4-(2-cyano-ethyl)-cyclohexyl]-carbamic acid tert-butyl ester. 1H NMR (400 MHz, CDC13) δ: 4.37 (br s, 1 H), 3.38 (br s, 1 H), 2.36 (t, 2 H), 2.03 (m, 2H), 1.79 (m, 2 H), 1.56 (q, 2 H), 1.44 (s, 9 H), 1.36 (m, 1 H), 1.19-0.98 (m, 4 H).
Trans 3-(4-Amino-cyclohexyl)-propionitrile trifluoro acetic acid salt
A solution of trans [4-(2-cyano-ethyl)-cyclohexyl]-carbamic acid tert-butyl ester (2.68 g, 10.6 mmol) in DCM (30 mL) was treated with trifluoro acetic acid (30 mL) and the mixture was stirred at ambient temperature for 30 mins then concentrated under vacuum. The residue was azeotroped with toluene (x3) and the resulting oil was triturated (diethyl ether) to give a solid, that was washed with ether and dried to give 2.75 g (97%) of trans 3-(4-amino-cyclohexyl)- propionitrile trifluoro acetic acid salt as a white solid. 1H NMR (400 MHz, CDC13) δ: 7.88 (br s, 3 H), 3.01 (m, l H), 2.37 (t, 2 H), 2.08 (m, 2 H), 1.89 (m, 2 H), 1.59 (q, 2 H), 1.49-1.35 (m, 3 H), 1.04 (m, 2 H).
Intermediate 7
Figure imgf000117_0001
Trans 4-(2-Methanesulfonyl-ethyl)-cyclohexylamine
Trans [4-(2-Methanesulfonyl-vinyl)-cyclohexyl]-carbamic acid tert-butyl ester
A solution of diethyl methanesulfonylmethyl phosphonate (900 mg, 3.90 mmol) in THF (20 mL) was treated with sodium hydride (160 mg of a 60% dispersion in mineral oil, 4.00 mmol). The mixture was stirred for 1 hour to give a thick slurry. Trans (4-formyl-cyclohexyl)-carbamic acid tert-butyl ester (975 mg, 4.30 mmol) was added and the slurry thinned. The mixture was stirred for 2 hours. A few drops of methanol were added and after 5 minutes the solvent removed under vacuum. The residue was partitioned between water (25 mL) and DCM (25 mL). The aqueous phase was washed with DCM (2 x 10 mL). The combined organic phase was dried over magnesium sulfate and concentrated under vacuum to afford 1.30 g (100%) of crude trans [4- (2-methanesulfonyl-vinyl)-cyclohexyl]-carbamic acid tert-butyl ester as a white solid (Ή NMR showed -4: 1 mixture of double bond geometries).
Major isomer Ή NMR (400 MHz, CDC13): δ 6.88 (dd, 1 H), 6.32 (dd, 1 H), 4.39 (br s, 1 H), 3.40 (br s, 1 H), 2.93 (s, 3 H), 2.21-1.98 (m, 3 H), 1.91-1.78 (m, 2 H), 1.44 (s, 9 H), 1.35-1.09 (m, 4 H).
For the minor isomer the following signals were clearly discernable δ 6.20 (d, 1 H), 6.12 (t, 1 H) and 2.96 (s, 3 H). Other signals were either coincident with those for the major isomer or ill- defined. Trans [4-(2-Methanesulfonyl-ethyl)-cyclohexyl]-carbamic acid tert-butyl ester
To trans [4-(2-methanesulfonyl-vinyl)-cyclohexyl]-carbamic acid tert-butyl ester (1.00 g, 3.30 mmol) in ethanol (15 mL, IMS grade), 10% palladium on carbon (100 mg) followed by ammonium formate (1.25 g, 19.8 mmol) were added. The mixture was heated at 90 °C for 45 min, then cooled to room temperature. A further 100 mg portion of palladium on carbon was added and the mixture was heated at 95 °C for 16 hours. The mixture was cooled to room temperature then filtered and concentrated under vacuum. The residue was taken up in water and extracted with ethyl acetate (x3). The combined organic extracts were washed (brine), dried (sodium sulfate) and concentrated under vacuum to give 928 mg (92%>) of trans [4-(2- methanesulfonyl-ethyl)-cyclohexyl]-carbamic acid tert-butyl ester. LCMS (Method E, ESI): RT = 3.35 min, m+H = 204.1 ; NMR (400 MHz, CDC13) δ: 4.36 (br s, 1 H), 3.38 (br s, 1 H), 3.01 (m, 2 H), 2.89 (s, 3 H), 2.07-1.98 (m, 2 H), 1.82-1.72 (m, 4 H), 1.44 (s, 9 H), 1.36-1.08 (m, 5 H).
Trans 4-(2-Methanesulfonyl-ethyl)-cyclohexylamine Trans [4-(2-methanesulfonyl-ethyl)-cyclohexyl]-carbamic acid tert-butyl ester (900 mg, 2.95 mmol) was dissolved in DCM (7 mL) and trifluoro acetic acid (7 mL) was added. The mixture was stirred at room temperature for 1 h, then concentrated. The residue was taken up in methanol and purified by I so lute® SCX-2 column (gradient: methanol to 2 M NH3 in methanol) to give 594 mg (98%) of trans 4-(2-methanesulfonyl-ethyl)-cyclohexylamine. LCMS (Method E, ESI): RT = 0.48 min, m+H = 206.1; NMR (400 MHz, DMSO-d6) δ: 3.09 (m, 2 H), 2.93 (s, 3 H), 2.48- 2.41 (m, 1 H), 1.78-1.64 (m, 4 H), 1.58-1.51 (m, 2 H), 1.30-1.18 (m, 1 H), 1.03-0.82 (m, 4 H).
Intermediate 8
Figure imgf000118_0001
((lR,3R)-3-Amino-cyclopentyl)-carbamic acid tert-butyl ester ((lR,3R)-3-Allyloxycarbonylamino-cyclopentyl)-carbamic acid tert-butyl ester
A mixture of (lR,3R)-3-tert-butoxycarbonylamino-cyclopentanecarboxylic acid (1.00 g, 4.36 mmol), diphenylphosphoryl azide (1.41 mL, 6.54 mmol) and triethylamine (1.21 mL, 8.72 mmol) in toluene (15 mL) was heated at 90 °C for 2 hours then cooled to ambient temperature. Allyl alcohol (1.63 mL, 24.0 mmol) and DMAP (54.0 mg, 0.44 mmol) were added and the resulting mixture heated at 90 °C for 18 hours. The cooled reaction mixture was concentrated under vacuum and the resulting residue was dissolved in ethyl acetate. The mixture was washed with 10% aqueous citric acid, saturated aqueous sodium carbonate and brine. The combined aqueous layers were back-extracted with ethyl acetate. The combined organic layers were dried (sodium sulfate) and concentrated under vacuum. The resulting residue was purified by column chromatography on silica gel (gradient: 0 to 12% ethyl acetate in cyclohexane) to afford 890 mg (72%) of ((lR,3R)-3-allyloxycarbonylamino-cyclopentyl)-carbamic acid tert-butyl ester. 1H NMR (400 MHz, DMSO-d6) δ: 7.23 (d, 1 H), 6.85 (d, 1 H), 5.90 (m, 1 H), 5.26 (dq, 1 H), 5.16 (dq, 1 H), 4.46-4.41 (m, 2 H), 3.93-3.83 (m, 2 H), 1.88 (m, 2 H), 1.65 (t, 2 H), 1.37 (s, 9 H).
((lR,3R)-3-Amino-cyclopentyl)-carbamic acid tert-butyl ester
A mixture of ((lR,3R)-3-allyloxycarbonylamino-cyclopentyl)-carbamic acid tert-butyl ester (890 mg, 3.13 mmol), 1,3-dimethylbarbituric acid (1.47 g, 9.39 mmol) and Pd(PPh3)4 (181 mg, 0.15 mmol) in DCM (30 mL) was stirred at ambient temperature for 90 minutes. The reaction mixture was concentrated under vacuum and purified by column chromatography on silica gel (gradient: 2M NH3 in methanol solution in DCM) to afford 350 mg (56%) ((lR,3R)-3-amino-cyclopentyl)- carbamic acid tert-butyl ester. LCMS (Method F, ESI): RT = 0.36 min, m+H = 200.9; 1H NMR (400 MHz, DMSO-d6) δ: 6.77 (s, 1 H), 3.90 (m, 1 H), 3.31 (m, 1 H), 1.87 (m, 2 H), 1.54 (m, 2 H), 1.37 (s, 10 H), 1.18 (m, 1 H).
Intermediate 9
Figure imgf000119_0001
((lR,3S)-3-Amino-cyclopentyl)-carbamic acid tert-butyl ester ((lR,3S)-3-Allyloxycarbonylamino-cyclopentyl)-carbamic acid tert-butyl ester
Following the procedure for ((lR,3R)-3-allyloxycarbonylamino-cyclopentyl)-carbamic acid tert- butyl ester the title compound was prepared from (lR,3S)-3-tert-butoxycarbonylamino- cyclopentanecarboxylic acid to afford 760 mg (61%) of ((lR,3S)-3-allyloxycarbonylamino- cyclopentyl)-carbamic acid tert-butyl ester as a white solid. 1H NMR (400 MHz, DMSO-d6) δ: 7.22 (d, 1 H), 6.84 (d, 1 H), 5.90 (m, 1 H), 5.27 (dq, 1 H), 5.16 (dq, 1 H), 4.47-4.42 (m, 2 H), 3.76 (m, 2 H), 2.14 (m, 1 H), 1.78-1.71 (m, 2 H), 1.50-1.44 (m, 2 H), 1.37 (s, 9 H), 1.27 (m, 1 H).
((lR,3S)-3-Amino-cyclopentyl)-carbamic acid tert-butyl ester
Following the procedure for ((lR,3R)-3-amino-cyclopentyl)-carbamic acid tert-butyl ester the title compound was prepared from ((lR,3S)-3-allyloxycarbonylamino-cyclopentyl)-carbamic acid tert-butyl ester with further purification by column chromatography on silica gel (gradient: 0 to 5% methanol in DCM then 0 to 10% 2M NH3 in methanol solution in DCM) to afford 300 mg (56%) of ((lR,3S)-3-amino-cyclopentyl)-carbamic acid tert-butyl ester. LCMS (Method G, ESI): RT = 0.32 min, m+H-Boc = 101.2; 1H NMR (400 MHz, DMSO-d6) δ: 6.84 (s, 1 H), 3.77-3.69 (m, 1 H), 3.21 (m, 2 H), 2.00 (m, 1 H), 1.78-1.63 (m, 2 H), 1.50 (m, 1 H), 1.37 (s, 9 H), 1.17 (m, 1 H).
Intermediate 10
Figure imgf000120_0001
Racemic trans (3-Amino-cyclopentyl)-carbamic acid tert-butyl ester
Racemic trans (3-amino-cyclopentyl)-carbamic acid tert-butyl ester was prepared following the methods outlined in J. Org. Chem. 2004, 69(13), 4538; Tetrahedron 1997, 53(9), 3347; WO94/17090 and Org. Lett. 2000, 2, 4169.
Intermediate 11
Figure imgf000120_0002
Racemic cis (3-Amino-cyclopentyl)-carbamic acid tert-butyl ester Racemic cis (3-amino-cyclopentyl)-carbamic acid tert-butyl ester was prepared following the methods outlined in J. Org. Chem. 2004, 69, 4538; Tetrahedron 1997, 53, 3347; WO2008/065021 ; WO94/17090; and Org Lett 2000, 2, 4169.
Intermediate 12
Figure imgf000120_0003
(( 1 R,3 S)-3-Amino-cyclopentyl)-acetonitrile ((lR,3S)-3-Amino-cyclopentyl)-acetonitrile was prepared following the methods outlined in Thomas, Abraham et al WO 2006040625.
Intermediate 13
Figure imgf000121_0001
( 1 R,3 S)-3-Amino-cyclopentanecarbonitrile
(lR,3S)-3-Amino-cyclopentanecarbonitrile was prepared following the methods outlined in Thomas, Abraham et al WO 2006040625.
Intermediate 14
Figure imgf000121_0002
Trans 3-(4-Amino-cyclohexylamino)-propionitrile
Trans 3-(4-Amino-cyclohexylamino)-propionitrile can be prepared following the methods outlined in WO 1994/15596 or using the route described below.
Trans [4-(2-Cyano-ethylamino)-cyclohexyl]-carbamic acid tert-butyl ester
The titled compound was prepared following the methods outlined in S. Fixon-Owoo et al. Phytochemistry 63 (2003) 315-334.
Trans 3-(4-Amino-cyclohexylamino)-propionitrile
Trans [4-(2-cyano-ethylamino)-cyclohexyl]-carbamic acid tert-butyl ester (5.75 g, 21.5 mmol) in DCM (50 mL) was treated dropwise with TFA (50 mL) at 0 °C. The mixture was then stirred for 30 minutes. The volatiles were removed in vacuo and the residue azeotroped (toluene) and triturated (diethyl ether) to furnish a white solid. Purification by column chromatography using an Isolute® SCX-2 cartridge (gradient: MeOH to 2M NH3 in MeOH) afforded 3.32 g (92%) of trans 3-(4-amino-cyclohexylamino)-propionitrile as a golden oil. LCMS (Method B, ESI): RT = 0.53 min.
Example 1
Figure imgf000122_0001
Trans (4-[2-((R)- 1 -Hydro xy-ethyl)- 1 ,3 ,5 ,9-tetraaza-cyclopenta[a]naphthalen- 1 -yl]-cyclohexyl} - acetonitrile
Trans [4-(3-Nitro-[ 1 ,5]naphthyridin-4-ylamino)-cyclohexyl]-acetonitrile A suspension of 4-chloro-3-nitro-[l,5]naphthyridine (prepared according to the procedures outlined in David Ach et al. WO 2006071862) (0.98 g, 4.69 mmol) in propan-2-ol (12 mL) was treated with trans (4-amino-cyclohexyl)-acetonitrile (0.71 g, 5.14 mmol) and N,N- diisopropylethyl amine (DIPEA, 1.22 mL, 7.02 mmol) and the reaction mixture was heated at 120 °C using microwave irradiation for 20 minutes. On cooling, a solid precipitated out of solution. This was filtered, washed with propan-2-ol (2x) and water and dried in vacuo, at 60°C to afford 0.88 g (60%) of trans [4-(3-nitro-[l,5]naphthyridin-4-ylamino)-cyclohexyl]-acetonitrile as a brown solid. LCMS (Method B, ESI): RT = 3.44 min, m+H = 312.2; 1H NMR (400 MHz, CDCI3): δ 9.39 (s, 1 H), 8.83 (dd, 1 H), 8.25 (dd, 1 H), 7.68 (dd, 1 H), 2.39 (m, 4 H), 2.05- 1.96 (m, 2 H), 1.89-1.76 (m, 1 H), 1.66-1.55 (m, 1 H), 1.54-1.38 (m, 4 H). Trans [4-(3-Amino-[ 1 ,5]naphthyridin-4-ylamino)-cyclohexyl]-acetonitrile
A mixture of trans [4-(3-nitro-[l,5]naphthyridin-4-ylamino)-cyclohexyl]-acetonitrile (0.88 g, 2.83 mmol) in ethanol (IMS grade, 8.80 mL) and water (2.90 mL) was treated with ammonium chloride (0.91 g, 17.0 mmol), followed by iron powder (0.63 g, 11.3 mmol) and the reaction mixture was heated at 80 °C for 8 hours. The reaction mixture was treated with further portions of ammonium chloride (0.33 g) and iron powder (0.32 g) and heating continued for 45 minutes. The iron residue was collected on a pad of Celite® and washed several times with ethanol (IMS grade) and water (3: 1). The filtrate was concentrated in vacuo and the aqueous residue was treated with saturated sodium hydro gencarbonate solution and extracted with ethyl acetate (EtOAc) (3x). The combined organic phases were washed (saturated sodium hydro gencarbonate solution and brine), dried (sodium sulfate) and concentrated in vacuo to give 0.66 g of a brown gummy solid. Purification by column chromatography on silica gel (gradient: EtOAc to 5% [2M NH3 in methanol (MeOH)] in EtOAc) afforded 0.63 g (79%) of trans [4-(3-amino- [l,5]naphthyridin-4-ylamino)-cyclohexyl]-acetonitrile as a pale brown oil, which crystallized on standing. LCMS (Method B, ESI): RT = 2.17 min, m+H = 282.2; 1H NMR (400 MHz, CDC13): δ 8.73 (dd, 1 H), 8.44 (s, 1 H), 8.21 (dd, 1 H), 7.41 (dd, 1 H), 5.71 (d, 1 H), 3.70 (s, 2 H), 3.64 (m, 1 H), 2.30 (d, 2 H), 2.11-2.05 (m, 2 H), 1.96-1.88 (m, 2 H), 1.79-1.67 (m, 1 H), 1.44-1.20 (m, 4 H). Trans (4-[2-((R)- 1 -Hydro xy-ethyl)- 1 ,3 ,5 ,9-tetraaza-cyclopenta[a]naphthalen- 1 -yl]-cyclohexyl} - acetonitrile
A solution of (R)-(+)-lactamide (0.64 g, 7.19 mmol) in anhydrous tetrahydrofuran (THF, 9.80 mL), under an atmosphere of nitrogen was treated with triethyloxonium tetrafluoroborate (1.28 g, 6.74 mmol) and the mixture was stirred at room temperature for 2 hours. The volatiles were removed in vacuo and the residue was dissolved in ethanol (absolute grade, 4.60 mL). This was added to a solution of trans [4-(3-amino-[l,5]naphthyridin-4-ylamino)-cyclohexyl]-acetonitrile (0.63 g, 2.24 mmol) in ethanol (absolute grade, 15 mL) and the reaction mixture was heated to 75 °C, under an atmosphere of nitrogen for 1 hour. The volatiles were removed in vacuo and the residue was partitioned between EtOAc and saturated sodium hydro gencarbonate solution. The phases were separated and the aqueous phase was extracted with EtOAc (2x). The combined organic phases were washed (brine), dried (sodium sulfate) and concentrated in vacuo to give 1.96 g of a yellow oil. Purification by column chromatography on silica gel (gradient: EtOAc to 7% [2M NH3 in MeOH] in EtOAc) afforded 0.62 g of a pale yellow gum, which crystallized on standing. This was triturated (diethyl ether) to afford 0.48 g, (64%) of trans {4-[2-((R)-l- hydroxy-ethyl)-l,3,5,9-tetraaza-cyclopenta[a]naphthalen-l-yl]-cyclohexyl}-acetonitrile as a white solid. LCMS (Method A, ESI): RT = 3.02 min, m+H = 336.1; 1H NMR (400 MHz, DMSO-d6): δ 9.29 (s, 1 H), 9.07 (s, 1 H), 8.53 (d, 1 H), 7.75 (dd, 1 H), 5.86 (m, 1 H), 5.26 (m, 1 H), 4.98 (m, 1 H), 3.29-3.16 (m, 1 H), 2.61 (d, 2 H), 2.15-1.92 (m, 4 H), 1.91-1.77 (m, 2 H), 1.69 (d, 3 H), 1.47-1.32 (s, 2 H).
Example 2
Figure imgf000123_0001
(R)-l-{l-[(lS,3R)-3-(2,2,2-Trifluoro-ethylamino)-cyclopentyl]-lH-l,3,5,9-tetraaza- cyclopenta[a]naphthalen-2-yl} -ethanol
[(lR,3S)-3-(3-Nitro-[l,5]naphthyridin-4-ylamino)-cyclopentyl]-carbamic acid tert-butyl ester
A suspension of 4-chloro-3-nitro-[l,5]naphthyridine (175 mg, 0.84 mmol) in propan-2-ol (5 mL) was treated with ((lR,3S)-3-amino-cyclopentyl)-carbamic acid tert-butyl ester (184 mg, 0.92 mmol) and DIPEA (219 uL, 1.26 mmol) and the reaction mixture was heated at 120 °C using microwave irradiation for 20 minutes. On cooling, a solid precipitated out of solution. This was filtered, washed with propan-2-ol (2x) and water and dried in vacuo to afford 111 mg (36%) of [(lR,3S)-3-(3-nitro-[l,5]naphthyridin-4-ylamino)-cyclopentyl]-carbamic acid tert-butyl ester as a dark yellow solid. The filtrate was re-filtered and the solid obtained was washed and dried in vacuo to produce a second crop of [(lR,3S)-3-(3-nitro-[l,5]naphthyridin-4-ylamino)- cyclopentyTJ-carbamic acid tert-butyl ester (99 mg, 32%). LCMS (Method B, ESI): RT = 3.73 min, m+H = 374.2; 1H NMR (400 MHz, CDC13): δ 9.38 (s, 1 H), 8.82 (dd, 1 H), 8.24 (dd, 1 H), 7.66 (dd, 1 H), 4.60 (m, 1 H), 4.09 (m, 1 H), 2.83-2.71 (m, 1 H), 2.38-2.26 (m, 1 H), 2.21- 2.09 (m, 1 H), 1.89-1.77 (m, 1 H), 1.75-1.64 (m, 1 H), 1.62-1.52 (m, 1 H, obscured by water), 1.45 (s, 9 H).
[(lR,3S)-3-(3-Amino-[l,5]naphthyridin-4-ylamino)-cyclopentyl]-carbamic acid tert-butyl ester
A mixture of [(lR,3S)-3-(3-nitro-[l,5]naphthyridin-4-ylamino)-cyclopentyl]-carbamic acid tert- butyl ester (196 mg, 0.53 mmol) in ethanol (IMS grade, 3 mL) and water (1 mL) was treated with ammonium chloride (169 mg, 3.2 mmol), followed by iron powder (118 mg, 2.1 mmol) and the reaction mixture was heated at 80 °C for 1.5 hours. After cooling, the iron residue was collected on a pad of Celite® and washed several times with ethanol (IMS grade) and water (3: 1). The organics were concentrated in vacuo and the aqueous residue was treated with saturated sodium hydro gencarbonate solution and extracted with EtOAc (3x). The combined organic phases were washed (saturated sodium hydro gencarbonate solution and brine), dried (sodium sulfate) and concentrated in vacuo to give a yellow gum. This was purified by column chromatography on silica gel (gradient: EtOAc to 10% [20% (2M NH3 in MeOH)] in EtOAc) to afford 157 mg (87%>) of [(lR,3S)-3-(3-amino-[l,5]naphthyridin-4-ylamino)-cyclopentyl]- carbamic acid tert-butyl ester as a pale yellow gum. LCMS (Method B, ESI): RT = 2.45 min, m+H = 344.1; 1H NMR (400 MHz, CDC13): δ 8.78 (s, 1 H), 8.48 (s, 1 H), 8.24 (dd, 1 H), 7.42 (dd, 1 H), 6.16 (s, 1 H), 5.52 (m, 1 H), 4.30-4.21(m, 1 H), 4.11-4.03 (m, 1 H), 3.84 (s, 2 H), 2.12-1.93 (m, 2 H), 1.88-1.73 (m, 2 H), 1.62-1.54 (s, 2 H), 1.46 (s, 9 H). {(1 R,3 S)-3-[2-((R)- 1 -Hydro xy-ethyl)- 1 ,3 ,5 ,9-tetraaza-cyclopenta[a]naphthalen- 1 -yl]- cyclopentyl}-carbamic acid tert-butyl ester
A solution of (R)-(+)-lactamide (133 mg, 1.50 mmol) in anhydrous dichloro methane (DCM, 2 mL), under an atmosphere of nitrogen was treated with triethyloxonium tetrafluoroborate (201 mg, 1.40 mmol) and the mixture was stirred at room temperature for 2 hours. The volatiles were removed in vacuo and the residue was dissolved in ethanol (absolute grade, 1 mL). This was added to a solution of [(lR,3S)-3-(3-amino-[l,5]naphthyridin-4-ylamino)-cyclopentyl]-carbamic acid tert-butyl ester (157 mg, 0.46 mmol) in ethanol (absolute grade, 3 mL) and the reaction mixture was heated to 75 °C, under an atmosphere of nitrogen for 1 hour. The volatiles were removed in vacuo and the residue was partitioned between EtOAc and saturated sodium hydro gencarbonate solution. The phases were separated and the aqueous phase was extracted with EtOAc (3x). The combined organic phases were washed (brine), dried (sodium sulfate) and concentrated in vacuo to give a yellow oil. This was purified by column chromatography on silica gel (gradient: EtOAc to 10% [20% (2M NH3 in MeOH)] in EtOAc) to afford 95 mg (52%) of a colourless gum. LCMS (Method B, ESI): RT = 3.03 min, m+H = 398.2; 1H NMR (400 MHz, CDC13): δ 9.37 (s, 1 H), 9.13 (m, 1 H), 8.61 (d, 1 H), 7.69 (dd, 1 H), 6.97 (m, 1 H), 5.29 (m, 1 H), 5.19 (m, 1 H), 4.35 (m, 1 H), 3.10 (d, 1 H), 3.00-2.89 (m, 1 H), 2.71-2.58 (m, 1 H), 2.55-2.45 (m, 1 H), 2.14-1.99 (m, 3 H, obscured by EtOAc), 1.80 (d, 3 H), 1.55 (s, 9 H).
(R)- 1 -[ 1 -(( 1 S,3R)-3-Amino-cyclopentyl)- 1 H- 1 ,3 ,5 ,9-tetraaza-cyclopenta[a]naphthalen-2-yl]- ethanol
{(1 R,3 S)-3-[2-((R)- 1 -Hydro xy-ethyl)- 1 ,3 ,5 ,9-tetraaza-cyclopenta[a]naphthalen- 1 -yl]- cyclopentyl}-carbamic acid tert-butyl ester (95.0 mg, 0.24 mmol) was treated with 20% trifluoro acetic acid (TFA) in DCM (5 mL) solution and the reaction mixture was stirred at room temperature for 30 minutes. The mixture was concentrated in vacuo and the residue was purified by column chromatography using an Isolute® SCX-2 cartridge (gradient: MeOH to 2M NH3 in MeOH) to afford 54 mg (76%) of (R)-l-[l-((lS,3R)-3-amino-cyclopentyl)-lH-l,3,5,9-tetraaza- cyclopenta[a]naphthalen-2-yl] -ethanol as a white solid. LCMS (Method B, ESI): RT = 0.52/1.04 min, m+H = 298.1 ; 1H NMR (400 MHz, CDC13): δ 9.35 (s, 1 H), 8.96 (dd, 1 H), 8.55 (dd, 1 H), 7.64 (dd, 1 H), 5.67 (m, 1 H), 5.37 (q, 1 H), 3.63 (m, 1 H), 2.94-2.82 (m, 1 H), 2.69-2.57 (m, 1 H), 2.47-2.38 (m, 1 H), 2.24-2.08 (m, 3 H), 1.81 (d, 3 H).
(R)-l-{l-[(lS,3R)-3-(2,2,2-Trifiuoro-ethylamino)-cyclopentyl]-lH-l,3,5,9-tetraaza- cyclopenta[a]naphthalen-2-yl} -ethanol A solution of (R)-l-[l-((lS,3R)-3-amino-cyclopentyl)-lH-l,3,5,9-tetraaza- cyclopenta[a]naphthalen-2-yl]-ethanol (51.5 mg, 0.17 mmol) in anhydrous THF (2 mL) was treated with triethylamine (48.0 μί, 0.35 mmol) and 2,2,2-trifluoroethyl trifluoromethanesulfonate (26.0 μί, 0.18 mmol) and the reaction mixture was stirred at room temperature overnight. The mixture was concentrated in vacuo and the residue purified by reverse phase high pressure liquid chromatography (HPLC) (gradient: 5 to 40% MeCN in H20 with 0.1% HC02H). The material obtained was then purified by column chromatography using an Isolute® SCX-2 cartridge (gradient: MeOH to 2M NH3 in MeOH) to give a colourless gum, which was triturated (EtOAc and diethyl ether) to afford 15.4 mg, (23%) of (R)-l-{l-[(lS,3R)-3- (2,2,2-trifluoro-ethylamino)-cyclopentyl]-lH- 1,3,5, 9-tetraaza-cyclopenta[a]naphthalen-2-yl}- ethanol as a white solid. LCMS (Method A, ESI): RT = 2.19 min, m+H = 380.1; 1H NMR (400 MHz, DMSO-d6): δ 9.30 (s, 1 H), 9.04 (dd, 1 H), 8.55 (dd, 1 H), 7.79 (dd, 1 H), 5.87 (d, 1 H), 5.64 (m, 1 H), 5.27 (m, 1 H), 3.54-3.32 (m, 2 H, obscured by water), 2.70-2.57 (m, 3 H), 2.35- 2.25 (m, 1 H), 2.21-1.97 (m, 3 H), 1.68 (d, 3 H).
Example 3
Figure imgf000126_0001
( 1 R,3R)-3-(2-Methyl- 1 ,3 ,5 ,9-tetraaza-cyclopenta[a]naphthalen- 1 -yl)-cyclohexanol
( 1 R,3R)-3 -(3 -Nitro- [ 1 ,5 ]naphthyridin-4-ylamino)-cyclohexano 1 A mixture of 4-chloro-3-nitro-[l,5]naphthyridine (100 mg, 0.48 mmol) in propan-2-ol (2 mL) was treated with (lR,3R)-3-amino-cyclohexanol [prepared following the procedures described in: P. Bernardelli et al. / Tetrahedron: Asymmetry 15 (2004) 1451-1455] (60.5 mg, 0.52 mmol) and DIPEA (207 μί, 1.19 mmol) and the reaction mixture was heated at 120 °C in the microwave for 20 minutes. After cooling, the resulting precipitate was collected and washed with propan-2-ol and air dried. (lR,3R)-3-(3-nitro-[l,5]naphthyridin-4-ylamino)-cyclohexanol (85.6 mg, 62%>) was isolated as a yellow/ green solid. LCMS (Method B, ESI): RT = 2.71 min, m+H = 289.1; 1H NMR (400 MHz, DMSO-d6): δ 9.20 (s, 1 H), 8.92 (dd, 1 H), 8.29 (dd, 1 H), 7.87 (dd, 1 H), 4.60 (d, 1 H), 3.94 (s, 1 H), 2.01-1.89 (m, 2 H), 1.84-1.71 (m, 2 H), 1.64-1.46 (m, 4 H).
(lR,3R)-3-(3-Amino-[l,5]naphthyridin-4-ylamino)-cyclohexanol
To a stirred suspension of (lR,3R)-3-(3-nitro-[l,5]naphthyridin-4-ylamino)-cyclohexanol (82.0 mg, 284 μηιοΐ) and iron powder (62.0 mg, 1.11 mmol) in ethanol (IMS grade, 2 mL) a solution of ammonium chloride (88.3 mg, 1.65 mmol) in water (0.5 mL) was added at room temperature. The mixture was then heated at 80 °C for 2 hours. After cooling, the mixture was filtered through of Celite® and the filter cake washed several times with ethanol (IMS grade) and water (4: 1). The filtrate was concentrated in vacuo and the resulting aqueous residue was treated with saturated sodium hydro gencarbonate solution and extracted with ethyl acetate (3x). The combined organic phases were washed (saturated sodium hydro gencarbonate solution and brine), dried (sodium sulfate) and concentrated in vacuo to afford 67.0 mg (91%) of (lR,3R)-3-(3- amino-[l,5]naphthyridin-4-ylamino)-cyclohexanol as a pale yellow residue. This material was used for the next step without further purification. LCMS (Method B, ESI): RT = 1.88 min, m+H = 259.1; 1H NMR (400 MHz, DMSO-d6): δ 8.71 (dd, 1 H), 8.42 (s, 1 H), 8.11 (dd, 1 H), 7.41 (dd, 1 H), 5.62 (d, 1 H), 5.01 (s, 2 H), 4.39 (d, 1 H), 4.29-4.18 (m, 1 H), 3.91-3.83 (m, 1 H), 1.73-1.23 (m, 8 H).
( 1 R,3R)-3-(2-Methyl- 1 ,3 ,5 ,9-tetraaza-cyclopenta[a]naphthalen- 1 -yl)-cyclohexanol
A stirred mixture of (lR,3R)-3-(3-amino-[l,5]naphthyridin-4-ylamino)-cyclohexanol (67.0 mg, 259 μιηοΐ) and triethyl orthoacetate (71.3 μί, 389 μιηοΐ) in acetic acid (2 mL) was heated to 120 °C for 30 minutes. After cooling, the mixture was concentrated in vacuo and the residue purified by column chromatography using an Isolute® SCX-2 cartridge (eluting: MeOH to 2M NH3 in MeOH solution). The crude product was purified by reverse phase HPLC (gradient: 5 to 98% acetonitrile (MeCN) in water, + 0.1% HC02H) and relevant fractions combined and then purified by column chromatography using an Isolute® SCX-2 cartridge (eluting with MeOH to 2M NH3 in MeOH solution) to provide 21.0 mg (29%) of (lR,3R)-3-(2-methyl-l,3,5,9-tetraaza- cyclopenta[a]naphthalen-l-yl)-cyclohexanol as a pale yellow solid. LCMS (Method A, ESI): RT = 2.46 min, m+H = 283.1; 1H NMR (400 MHz, DMSO-d6): δ 9.20 (s, 1 H), 9.03 (s, 1 H), 8.52 (dd, 1 H), 7.73 (dd, 1 H), 4.86 (br s, 1 H), 4.74 (s, 1 H), 4.21 (m, 1 H), 2.75 (s, 3 H), 2.05-1.90 (m, 2 H), 1.88-1.60 (m, 6 H).
Example 4
Figure imgf000128_0001
Trans [4-(2-Hydroxymethyl- 1 ,3 ,5 ,9-tetraaza-cyclopenta[a]naphthalen- 1 -yl)-cyclohexyl]- acetonitrile
A solution of glycolamide (52.4 mg, 698 μηιοΐ) in anhydrous THF (2 mL), under an atmosphere of nitrogen, was treated with triethyloxonium tetrafluoroborate (122 mg, 641 μιηοΐ) and stirred at room temperature for 2 hours. The volatiles were removed in vacuo and the residue was dissolved in ethanol (absolute grade, 1.5 mL). This was added to a stirred solution of trans [4-(3- amino-[l,5]naphthyridin-4-ylamino)-cyclohexyl]-acetonitrile (60.0 mg, 214 μιηοΐ) in ethanol (absolute grade, 2.5 mL) and the reaction mixture was heated to 75 °C for 2 hours. The volatiles were removed in vacuo and the residue was partitioned between EtOAc and saturated sodium hydro gencarbonate solution. The phases were separated and the aqueous phase extracted with EtOAc (2x). The combined organic phases were washed with brine, dried (sodium sulfate), filtered and concentrated in vacuo to give a gum. Purified by column chromatography on silica gel (gradient: EtOAc to 30% [2M NH3 in MeOH] in EtOAc) to afford 46.6 mg of trans [4-(2- hydroxymethyl-l,3,5,9-tetraaza-cyclopenta[a]naphthalen-l-yl)-cyclohexyl]-acetonitrile as a white solid. LCMS (Method A, ESI): RT = 2.87 min, m+H = 322.0; 1H NMR (400 MHz, DMSO): δ (400 MHz, DMSO-d6): δ 9.27 (s, 1 H), 9.07 (m, 1 H), 8.54 (dd, 1 H), 7.76 (dd, 1 H), 5.84 (t, 1 H), 4.91 (d, 2 H), 4.82 (s, 1 H), 3.22 (s, 1 H), 2.61 (d, 2 H), 2.11-1.79 (m, 6 H), 1.48-1.35 (m, 2 H).
Example 5
Figure imgf000128_0002
Trans (4-[2-((R)- 1 -Hydro xy-ethyl)-imidazo[4,5-c]quinolin- 1 -yl]-cyclohexyl} -acetonitrile Trans [4-(3-Nitro-quinolin-4-ylamino)-cyclohexyl]-acetonitrile
A solution of trans (4-amino-cyclohexyl)-acetonitrile (1.74 g) in propan-2-ol (25 mL) was treated with DIPEA (3.00 mL) and 4-chloro-3-nitro-quinoline (CAS: 39061-97-7) (3.00 g) at room temperature. The mixture was then heated to reflux for 2 hours. After cooling, the resulting yellow precipitate was collected, washed (propan-2-ol and diethyl ether) and air dried to provide 3.74 g of trans [4-(3-nitro-quinolin-4-ylamino)-cyclohexyl]-acetonitrile as a yellow solid. 1H NMR (400 MHz, CDC13): δ 9.38 (s, 1 H), 9.34 (d, 1 H), 8.14 (d, 1 H), 8.02 (dd, 1 H), 7.80 (ddd, 1 H), 7.53 (ddd, 1 H), 4.21-4.08 (m, 1 H), 2.41-2.30 (m, 4 H), 2.03 (m, 2 H), 1.88-1.77 (m, 1 H), 1.66-1.53 (m, 2 H, partially obscured by water), 1.44-1.30 (m, 2 H). Trans [4-(3-Amino-quinolin-4-ylamino)-cyclohexyl]-acetonitrile
A solution of trans [4-(3-nitro-quinolin-4-ylamino)-cyclohexyl]-acetonitrile (3.50 g) in EtOAc (75 mL) and THF (75 mL) was treated with palladium on carbon (350 mg) under a nitrogen atmosphere. The mixture was evacuated and purged with hydrogen and then stirred under an atmosphere of hydrogen for 4 hours. The mixture was filtered through Celite® and the filtrate concentrated in vacuo. The residue was triturated (ethyl acetate) and the resulting solid washed (10% ethyl acetate in diethyl ether) and air dried to provide 1.40 g (44%) of trans [4-(3-amino- quinolin-4-ylamino)-cyclohexyl]-acetonitrile as a yellow solid. The filtrate from the above trituration was concentrated in vacuo and the residue purified by column chromatography on silica gel (gradient: EtOAc to 2.5%[2M NH3 in MeOH] in EtOAc). The resulting residue was triturated (pentane) to give an additional 1.38 g (43%) of trans [4-(3-amino-quinolin-4-ylamino)- cyclohexyl]-acetonitrile as a red solid, which was analytically identical to the first batch. 1H NMR (400 MHz, CDC13): δ 8.49 (s, 1 H), 8.00-7.96 (m, 1 H), 7.80-7.76 (m, 1 H), 7.50-7.42 (m, 2 H), 3.78 (s, 2 H), 3.36-3.26 (m, 1 H), 2.26 (d, 2 H), 2.12-2.03 (m, 2 H), 1.95-1.87 (m, 2 H), 1.78-1.65 (m, 1 H), 1.41-1.29 (m, 2 H), 1.24-1.12 (m, 2 H). Trans (4-[2-((R)- 1 -Hydro xy-ethyl)-imidazo[4,5-c]quinolin- 1 -yl]-cyclohexyl} -acetonitrile
A suspension of triethyloxonium tetrafluoroborate (2.44 g) in THF (25 mL) was treated with (R)- (+)-lactamide (1.37 g) at room temperature and stirred for 2 hours. The volatiles were removed in vacuo and the residue was dissolved in ethanol (absolute grade, 25 mL) and trans [4-(3-amino- quinolin-4-ylamino)-cyclohexyl]-acetonitrile (2.40 g) was added. The mixture was heated to 75 °C for 4 hours. TLC analysis indicated remaining starting material, therefore a suspension of triethyloxonium tetrafluoroborate (2.44 g) in THF (25 mL) was treated with (R)-(+)-lactamide (1.37 g) at room temperature for 2 hours and then concentrated in vacuo. The residue was dissolved in minimal ethanol and added to the above reaction mixture at room temperature and then heated at 75 °C for 18 hours. After cooling, the solvent was removed in vacuo and the resulting residue dissolved EtOAc. The mixture was washed (saturated sodium hydro gencarbonate solution, water and brine), dried (sodium sulfate) and concentrated in vacuo. Purification by column chromatography on silica gel (gradient: 5% to 10% [2M NH3 in MeOH] in EtOAc) afforded 1.83 g (64%) trans {4-[2-((R)-l-hydroxy-ethyl)-imidazo[4,5-c]quinolin-l- yl]-cyclohexyl}-acetonitrile as a foam. LCMS (Method A, ESI): RT = 2.42 min, m+H = 335.1; 1H NMR (400 MHz, DMSO-d6, 80 °C): δ 9.13 (s, 1 H), 8.46 (d, 1 H), 8.16 (dd, 1 H), 7.76- 7.63 (m, 2 H), 5.42 (d, 1 H), 5.26-5.08 (m, 2 H), 2.60-2.52 (m, 3 H), 2.16-1.99 (m, 6 H), 1.70 (d, 3 H), 1.56-1.41 (m, 2 H).
Example 6
Figure imgf000130_0001
Trans (R)- 1 - { 1 -[4-(2,2,2-Trifluoro-ethylamino)-cyclohexyl]- lH-imidazo[4,5-c]quinolin-2-yl} - ethanol
Trans [4-(3-Nitro-quinolin-4-ylamino)-cyclohexyl]-carbamic acid tert-butyl ester
A suspension of 4-chloro-3-nitro-quinoline (800 mg, 3.80 mmol) in propan-2-ol (10 mL) was treated with (4-amino-cyclohexyl)-carbamic acid tert-butyl ester (903 mg, 4.20 mmol) and DIPEA (1.00 mL, 5.80 mmol) and the reaction mixture was heated at 120 °C using microwave irradiation for 25 minutes. After cooling, the resulting precipitate was collected by filtration, washed (propan-2-ol, water and diethyl ether) and dried between 50-70 °C under vacuum for 90 minutes. Trans [4-(3-nitro-quinolin-4-ylamino)-cyclohexyl]-carbamic acid tert-butyl ester (1.30 g, 88%) was isolated as a yellow solid. LCMS (Method B, ESI): RT = 3.17 min, m+H = 387; 1H NMR (400 MHz, CDC13): δ 9.37 (s, 1 H), 9.34 (d, 1 H), 8.14 (d, 1 H), 8.01 (dd, 1 H), 7.78 (ddd, 1 H), 7.51 (ddd, 1 H), 4.40 (s, 1 H), 4.18-4.05 (m, 1 H), 3.53 (s, 1 H), 2.28 (m, 2 H), 2.16 (m, 2 H), 1.79-1.59 (m, 2 H), 1.46 (s, 9 H), 1.37-1.24 (m, 2 H).
Trans [4-(3-Amino-quinolin-4-ylamino)-cyclohexyl]-carbamic acid tert-butyl ester A suspension of trans [4-(3-nitro-quinolin-4-ylamino)-cyclohexyl]-carbamic acid tert-butyl ester (1.30 g, 3.40 mmol) in ethanol (IMS grade, 34 mL) was treated with palladium on carbon (360 mg, 10% Pd, 0.34 mmol) under a nitrogen atmosphere. The mixture was evacuated and purged with hydrogen and then stirred under an atmosphere of hydrogen for 3 hours. The mixture was filtered through Celite® and the filtrate concentrated in vacuo. The residue purified by column chromatography on silica gel (gradient: EtOAc to 7%[2M NH3 in MeOH] in EtOAc) to give 695 mg (58%) of trans [4-(3-amino-quinolin-4-ylamino)-cyclohexyl]-carbamic acid tert-butyl ester as an orange foam. LCMS (Method B, ESI): RT = 2.49 min, m+H = 357; 1H NMR (400 MHz, DMSO-d6, 80 °C): δ 8.39 (s, 1 H), 7.95 (m, 1 H), 7.72 (m, 1 H), 7.37-7.27 (m, 2 H), 4.75 (m, 2 H), 4.23 (d, 1 H), 3.63 (s, 1 H), 3.27-3.13 (m, 2 H), 1.89-1.74 (m, 4 H), 1.48-1.37 (m, 2 H, partially obscured by water), 1.22- 1.09 (m, 2 H).
Trans {4-[2-((R)-l-Hydroxy-ethyl)-imidazo[4,5-c]quinolin-l-yl]-cyclohexyl}-carbamic acid tert- butyl ester
A solution of (R)-(+)-lactamide (257 mg, 2.90 mmol) in anhydrous DCM (4.1 mL), under an atmosphere of nitrogen was treated with triethyloxonium tetrafluoroborate (514 mg, 2.70 mmol) and the mixture was stirred at room temperature for 2 hours. The volatiles were removed in vacuo and the residue was dissolved in ethanol (absolute grade, 2.2 mL). This was added to a suspension of trans [4-(3-amino-quinolin-4-ylamino)-cyclohexyl]-carbamic acid tert-butyl ester (322 mg, 0.90 mmol) in ethanol (absolute grade, 6 mL) and the reaction mixture was heated to 75 °C for 3 hours. The volatiles were removed in vacuo and the residue was triturated (EtOAc) to provide 229 mg (62%) of trans {4-[2-((R)-l-hydroxy-ethyl)-imidazo[4,5-c]quinolin-l-yl]- cyclohexyl}-carbamic acid tert-butyl ester as a white solid. LCMS (Method B, ESI): RT = 2.54 min, m+H = 411; 1H NMR (400 MHz, DMSO-d6, 80 °C): δ 9.14 (s, 1 H), 8.44 (d, 1 H), 8.17 (dd, 1 H), 7.75-7.64 (m, 2 H), 6.43 (s, 1 H), 5.44 (m, 1 H), 5.27-5.18 (m, 1 H), 5.17-5.06 (m, 1 H), 3.68-3.54 (m, 1 H), 2.60-2.49 (m, 2 H), 2.14-2.01 (m, 4 H), 1.70 (d, 3 H), 1.65-1.51 (m, 2 H), 1.43 (s, 9 H).
Trans (R)- 1 -[ 1 -(4-Amino-cyclohexyl)- lH-imidazo[4,5-c]quinolin-2-yl]-ethanol
Trans {4-[2-((R)-l-hydroxy-ethyl)-imidazo[4,5-c]quinolin-l-yl]-cyclohexyl}-carbamic acid tert- butyl ester (224 mg, 0.55 mmol) was treated with TFA (3 mL) at room temperature for 25 minutes. The mixture was concentrated in vacuo and the resulting residue azeotroped with toluene. The residue was purified by column chromatography using an Isolute® SCX-2 cartridge (eluting with MeOH to 2M NH3 in MeOH solution) and the isolated product triturated (diethyl ether) to provide 146 mg (86%) of trans (R)-l-[l-(4-amino-cyclohexyl)-lH-imidazo[4,5- c]quinolin-2-yl] -ethanol as a white solid. LCMS (Method B, ESI): RT = 0.63 min, m+H = 311 ; 1H NMR (400 MHz, DMSO-d6, 80 °C): δ 9.15 (s, 1 H), 8.47 (d, 1 H), 8.17 (m, 1 H), 7.76- 7.65 (m, 2 H), 5.43 (s, 1 H), 5.26-5.19 (m, 1 H), 5.18-5.08 (m, 1 H), 2.61-2.50 (m, 2 H, partially obscured by DMSO), 2.09-1.96 (m, 4 H), 1.71 (d, 3 H), 1.50-1.36 (m, 2 H).
Trans (R)- 1 - { 1 -[4-(2,2,2-Trifluoro-ethylamino)-cyclohexyl]- lH-imidazo[4,5-c]quinolin-2-yl} - ethanol
A suspension of trans (R)-l-[l-(4-amino-cyclohexyl)-lH-imidazo[4,5-c]quinolin-2-yl]-ethanol (142 mg, 0.46 mmol) in anhydrous THF (4.6 mL) was treated with triethylamine (0.16 mL, 1.15 mmol) and 2,2,2-trifluoroethyl trifluoromethanesulfonate (99.0 μί, 0.69 mmol), the reaction mixture was then heated to 50 °C for 18 hours. The mixture was concentrated in vacuo and the residue dissolved in EtOAc. The mixture was washed (water and brine), dried (sodium sulfate) and concentrated in vacuo. Purification by column chromatography on silica gel (gradient: EtOAc to 8%[2M N¾ in MeOH] in EtOAc) provided a gum. The gum was azeotroped (diethyl ether) and then triturated (pentane) to provide 94.0 mg (52%) of trans (R)-l-{l-[4-(2,2,2- trifluoro-ethylamino)-cyclohexyl]-lH-imidazo[4,5-c]quinolin-2-yl}-ethanol as an off-white solid. LCMS (Method A, ESI): RT = 1.64 min, m+H = 393; 1H NMR (400 MHz, DMSO-d6, 80 °C): δ 9.14 (s, 1 H), 8.44 (d, 1 H), 8.16 (m, 1 H), 7.74-7.63 (m, 2 H), 5.39 (m, 1 H), 5.24-5.09 (m, 2 H), 3.40-3.29 (m, 2 H), 2.91-2.80 (m, 1 H), 2.59-2.49 (m, 2 H, partially obscured by DMSO), 2.20-2.01 (m, 4 H), 1.70 (d, 3 H), 1.50-1.36 (m, 2 H).
Example 7
Figure imgf000132_0001
Trans [4-(2-Hydroxymethyl-imidazo[4,5-c]quinolin-l-yl)-cyclohexyl]-acetonitrile A solution of glycolamide (167 mg, 2.20 mmol) in anhydrous THF (3.1 mL), under an atmosphere of nitrogen, was treated with triethyloxonium tetrafluoroborate (397 mg, 2.10 mmol) and stirred at room temperature for 3.5 hours. The volatiles were removed in vacuo and the residue was dissolved in ethanol (absolute grade, 1.3 mL). This was added to a stirred solution of trans [4-(3-amino-quinolin-4-ylamino)-cyclohexyl]-acetonitrile (195 mg, 0.70 mmol) in ethanol (absolute grade, 4.6 mL) and the reaction mixture was heated to 75 °C for 1 hour. The volatiles were removed in vacuo and the residue was partitioned between EtOAc and saturated sodium hydro gencarbonate solution. The phases were separated and the aqueous phase was extracted with EtOAc (2x). The combined organic phases were washed with brine, dried (sodium sulfate), filtered and concentrated in vacuo to give a brown gum. Purified by column chromatography on silica gel (gradient: EtOAc to 10% [2M NH3 in MeOH] in EtOAc) to afford a green gum. This was triturated (diethyl ether) and the solid obtained dried in vacuo, at 60 °C, to afford 136 mg (61%) of trans [4-(2-hydroxymethyl-imidazo[4,5-c]quinolin-l-yl)-cyclohexyl]-acetonitrile as a pale yellow solid. LCMS (Method A, ESI): RT = 2.32 min, m+H = 321.1 ; 1H NMR (400 MHz, DMSO-d6, 80 °C): δ 9.13 (s, 1 H), 8.42 (d, 1 H), 8.18 (dd, 1 H), 7.76-7.66 (m, 2 H), 5.49- 5.43 (t, 1 H), 5.10-4.98 (m, 1 H), 4.90 (d, 2 H), 2.56 (d, 2 H), 2.53-2.43 (m, 2 H, obscured by DMSO), 2.20-1.95 (m, 5 H), 1.59-1.46 (m, 2 H).
Example 8
Figure imgf000133_0001
(R)- 1 - { 1 -[ 1 S,3R)-3-(2,2,2-Trifluoro-ethylamino)-cyclopentyl]- lH-imidazo[4,5-c]quinolin-2-yl} - ethanol
[(lR,3S)-3-(3-Nitro-quinolin-4-ylamino)-cyclopentyl]-carbamic acid tert-butyl ester
A mixture of ((lR,3S)-3-amino-cyclopentyl)-carbamic acid tert-butyl ester (1.06 g, 5.30 mmol) in propan-2-ol (10 mL) was treated with 4-chloro-3-nitroquinoline (1.00 g, 4.80 mmol) and DIPEA (1.30 mL, 7.48 mmol) and the reaction mixture was heated at 120 °C using microwave irradiation for 25 minutes. On cooling, a solid precipitated out of solution. This was filtered, washed with propan-2-ol and water and dried in vacuo, at 60 °C, to afford 1.18 g (66%>) of [(lR,3S)-3-(3-nitro-quinolin-4-ylamino)-cyclopentyl]-carbamic acid tert-butyl ester as a bright yellow solid. LCMS (Method B, ESI): RT = 3.01 min, m+H = 373.1 ; 1H NMR (400 MHz, CDC13): δ 9.81 (d, 1 H), 9.36 (s, 1 H), 8.25 (d, 1 H), 7.99 (d, 1 H), 7.77 (t, 1 H), 7.50 (t, 1 H), 4.73-4.576 (m, 2 H), 4.13-4.01 (m, 1 H), 2.79-2.69 (dt, 1 H), 2.33-2.11 (m, 2 H), 2.02-1.90 (m, 1 H), 1.82-1.70 (m, 2 H), 1.45 (s, 9 H). (lS,3R)-N-(3-Nitro-quinolin-4-yl)-cyclopentane-l,3-diamine
A solution of [(lR,3S)-3-(3-nitro-quinolin-4-ylamino)-cyclopentyl]-carbamic acid tert-butyl ester (1.18 g, 3.17 mmol) in DCM (6 mL) was treated with TFA (6 mL) and the reaction mixture was stirred at room temperature for 25 minutes. The mixture was concentrated in vacuo and azeotroped with toluene. The residue was purified by column chromatography using an Isolute® SCX-2 cartridge (gradient: MeOH to 2M NH3 in MeOH) to afford a yellow crystalline solid. This was treated with 40-60% petroleum ether and was stirred vigorously for 10 minutes. The solid was filtered, washed with 40-60% petroleum ether and dried in vacuo, at 60 °C, to afford 0.80 g (93%>) of (lS,3R)-N-(3-nitro-quinolin-4-yl)-cyclopentane-l,3-diamine as a yellow sold. LCMS (Method B, ESI): RT = 0.56 min, m+H = 273.1 ; 1H NMR (400 MHz, CDC13): δ 10.27 (m, 1 H), 9.34 (s, 1 H), 8.25 (dd, 1 H), 7.98 (dd, 1 H), 7.75 (ddd, 1 H), 7.47 (ddd, 1 H), 4.73-4.63 (m, 1 H), 3.75-3.68 (m, 1 H), 2.44-2.34 (m, 1 H), 2.30-2.15 (m, 2 H), 2.02-1.91 (m, 1 H), 1.80-1.69 (m, 2 H), 1.44-1.25 (s, 2 H).
(lS,3R)-N-(3-Nitro-quinolin-4-yl)-N'-(2,2,2-trifluoro-ethyl)-cyclopentane-l,3-diamine A solution of (lS,3R)-N-(3-nitro-quinolin-4-yl)-cyclopentane-l,3-diamine (800 mg, 2.94 mmol) in a 1 : 1 mixture of DCM and N,N-dimethylformamide (DMF, 8mL) was treated with triethylamine (0.16 mL, 1.15 mmol) and 2,2,2-trifluoroethyl trifluoromethanesulfonate (1.02 mL, 7.33 mmol). The reaction mixture was heated to 50 °C for 3 hours. The mixture was concentrated in vacuo and the residue was diluted with water and EtOAc. The phases were separated and the aqueous phase was extracted with EtOAc (2x). The combined organic phases were washed (water, saturated sodium hydro gencarbonate solution and brine), dried (sodium sulfate) and concentrated in vacuo to afford 1.02 g (98%>) of (lS,3R)-N-(3-nitro-quinolin-4-yl)- N'-(2,2,2-trifluoro-ethyl)-cyclopentane-l,3-diamine as a yellow oil. LCMS (Method B, ESI): RT = 2.00 min, m+H = 355.1; 1H NMR (400 MHz, CDC13): δ 9.96 (m, 1 H), 9.34 (s, 1 H), 8.22 (d, 1 H), 7.99 (dd, 1 H), 7.75 (ddd, 1 H), 7.47 (ddd, 1 H), 4.75-4.66 (m, 1 H), 3.57 (m, 1 H), 3.36-3.19 (m, 2 H), 2.42-2.23 (m, 2 H), 2.16-2.07 (m, 1 H), 2.01-1.91 (m, 1 H), 1.90-1.79 (m, 2 H).
N*4*-[(lS,3R)-3-(2,2,2-Trifluoro-ethylamino)-cyclopentyl]-quinoline-3,4-diamine
A solution of (lS,3R)-N-(3-nitro-quinolin-4-yl)-N'-(2,2,2-trifluoro-ethyl)-cyclopentane-l,3- diamine (1.02 g, 2.88 mmol) in ethanol (IMS grade, 10.5 mL) and water (3.5 mL) was treated with ammonium chloride (0.92 g, 17.3 mmol), followed by iron powder (0.65 g, 11.5 mmol) and the reaction mixture was heated at 80 °C overnight. The iron residue was collected on a pad of Celite® and washed several times with ethanol (IMS grade) and water (3: 1). The organics were concentrated in vacuo and the aqueous residue was treated with saturated sodium hydro gencarbonate solution and extracted with ethyl acetate (3x). The combined organic phases were washed with brine, dried (sodium sulfate), filtered and concentrated in vacuo to give 0.65 g of a brown oil. This was purified by column chromatography on silica gel (gradient: EtOAc to 15% [2M NH3 in MeOH] in EtOAc) to afford 396 mg (40%) of N*4*-[(lS,3R)-3-(2,2,2- trifluoro-ethylamino)-cyclopentyl]-quinoline-3,4-diamine as a brown oil. LCMS (Method B, ESI): RT = 1.35 min, m+H = 325.1 ; 1H NMR (400 MHz, CDC13): δ 8.45 (s, 1 H), 7.95-7.88 (m, 2 H), 7.47-7.39 (m, 2 H), 4.61 (s, 1 H), 4.23 (s, 1 H), 3.78 (s, 2 H), 3.44-3.38 (m, 1 H), 3.31- 3.20 (qd, 2 H), 2.02-1.66 (m, 6 H).
(R)- 1 - { 1 -[ 1 S,3R)-3-(2,2,2-Trifluoro-ethylamino)-cyclopentyl]- lH-imidazo[4,5-c]quinolin-2-yl} - ethanol
A solution of (R)-(+)-lactamide (257 mg, 2.90 mmol) in anhydrous THF (2.1 mL), under an atmosphere of nitrogen was treated with triethyloxonium tetrafluoroborate (264 mg, 1.39 mmol) and the mixture was stirred at room temperature for 2 hours. The volatiles were removed in vacuo and the residue was dissolved in ethanol (absolute grade, 0.8 mL). This was added to a suspension of N*4*-[(l S,3R)-3-(2,2,2-trifluoro-ethylamino)-cyclopentyl]-quinoline-3,4-diamine (150 mg, 0.47 mmol) in ethanol (absolute grade, 3.1 mL) and the reaction mixture heated to 75 °C for 5 hours. The volatiles were removed in vacuo and the residue was partitioned between EtOAc and saturated sodium hydro gencarbonate solution. The phases were separated and the aqueous phase was extracted with EtOAc (2x). The combined organic phases were washed (brine), dried (sodium sulfate) and concentrated in vacuo to afford 419 mg of a brown oil. This was purified by column chromatography on silica gel (gradient: EtOAc to 8% [2M NH3 in MeOH] in EtOAc) to afford 107 mg of a colourless gum. The gum was dissolved in acetonitrile and water and lyophilised to afford 96 mg (55%) of (R)-l-{l-[lS,3R)-3-(2,2,2-trifiuoro- ethylamino)-cyclopentyl]-lH-imidazo[4,5-c]quinolin-2-yl}-ethanol as a white solid. LCMS (Method A, ESI): RT = 2.18 min, m+H = 379.1; 1H NMR (400 MHz, CDC13): δ 9.24 (s, 1 H), 8.55 (d, 1 H), 8.27 (dd, 1 H), 7.70-7.61 (m, 2 H), 5.55-5.43 (m, 1 H), 5.37-5.29 (q, 1 H), 3.67- 3.59 (m, 1 H), 3.34-3.21 (m, 2 H), 2.74-2.61 (m, 1 H), 2.57-2.47 (m, 1 H), 2.44-2.35 (m, 1 H), 2.34-2.15 (m, 2 H), 2.08-1.98 (m, 1 H), 1.81 (d, 3 H).
Example 9
Figure imgf000136_0001
Trans N-[l-(4-Cyanomethyl-cyclohexyl)-lH-imidazo[4,5-c]quinolin-2-ylmethyl]- methanesulfonamide
N-tert-Butylcarbamate-N- [ 1 -(4-cyanomethyl-cyclohexyl)- 1 H-imidazo [4,5 -c] quino lin-2- ylmethylj-methanesulfonamide
A suspension of trans [4-(2-hydroxymethyl-imidazo[4,5-c]quinolin-l-yl)-cyclohexyl]- acetonitrile (110 mg, 0.34 mmol) in anhydrous THF (3 mL), under an atmosphere of nitrogen was treated with tert-butyl N-methanesulfonylcarbamate (147 mg, 0.76 mmol), followed by triphenylphosphine (180 mg, 0.69 mmol) and then diisopropyl azodicarboxylate (135 μί, 0.69 mmol). The reaction mixture was stirred at room temperature, under nitrogen overnight. The mixture was diluted with EtOAc and water and the phases were separated. The aqueous layer was extracted with EtOAc (2x) and the combined organic phases washed (brine), dried (sodium sulfate) and concentrated in vacuo to afford a green gel. This was purified by column chromatography on an Isolute® SCX-2 cartridge (eluting with MeOH and then 2M NH3 in MeOH) to afford 175 mg (100%) of N-tert-butylcarbamate-N-[l-(4-cyanomethyl-cyclohexyl)- lH-imidazo[4,5-c]quinolin-2-ylmethyl]-methanesulfonamide as a colourless gum. LCMS (Method B, ESI): RT = 2.85 min, m+H = 498.2; 1H NMR (400 MHz, DMSO-d6, 80 °C): δ 9.11 (s, 1 H), 8.40 (d, 1 H), 8.18 (dd, 1 H), 7.76-7.68 (m, 2 H), 5.32 (s, 2 H), 5.05 (s, 1 H), 3.70 (m, 1 H), 3.63 (s, 3 H), 2.55 (d, 2 H), 2.39-2.26 (m, 2 H), 2.20-2.11 (m, 2 H), 2.09-2.02 (m, 2 H), 1.63-1.50 (m, 2 H), 1.39 (s, 9 H).
Trans N-[l-(4-Cyanomethyl-cyclohexyl)-lH-imidazo[4,5-c]quinolin-2-ylmethyl]- methanesulfonamide A solution of N-tert-butylcarbamate-N-[l-(4-cyanomethyl-cyclohexyl)-lH-imidazo[4,5- c] quino lin-2-ylmethyl]-methanesulfonamide (166 mg, 0.33 mmol) in TFA (3 mL) was stirred at room temperature for 30 minutes. The mixture was diluted with toluene and concentrated in vacuo. The residue was azeotroped (toluene) and then purified by column chromatography using an Isolute® SCX-2 cartridge (eluting with MeOH and then 2M NH3 in MeOH) to afford a yellow glass. This was purified by column chromatography on silica gel (gradient: 0 to 10% [NH3 in MeOH] in EtOAc) to afford 121 mg (91%) of a white foam. This was dissolved in acetonitrile and water and lyophilised to afford 95.0 mg (71%) of trans N-[l-(4-cyanomethyl- cyclohexyl)-lH-imidazo[4,5-c]quinolin-2-ylmethyl]-methanesulfonamide as a white solid. LCMS (Method A, ESI): RT = 2.49 min, m+H = 398.1; 1H NMR (400 MHz, DMSO-d6, 80 °C): δ 9.16 (s, 1 H), 8.46 (d, 1 H), 8.20 (dd, 1 H), 7.79-7.68 (m, 2 H), 7.50 (s, 1 H), 5.02 (m, 1 H), 4.71 (s, 2 H), 3.03 (s, 3 H), 2.57 (d, 2 H), 2.46 (m, 1 H, partially obscured by DMSO), 2.21- 2.13 (m, 2 H), 2.12-2.03 (m, 4 H), 1.61-1.48 (m, 2 H).
Example 10
Figure imgf000137_0001
{l-[(lS,3R)-3-(2,2,2-Trifiuoro-ethylamino)-cyclopentyl]-lH-imidazo[4,5-c]quinolin-2-yl}- methanol
A suspension of glycolamide (144 mg, 1.93 mmol) in anhydrous THF (2.7 mL), under an atmosphere of nitrogen was treated with triethyloxonium tetrafluoroborate (343 mg, 1.81 mmol) and the mixture was stirred at room temperature for 2 hours. The volatiles were removed in vacuo and the residue was dissolved in ethanol (absolute grade, 1.1 mL). This was added to a suspension of N*4*-[(l S,3R)-3-(2,2,2-trifluoro-ethylamino)-cyclopentyl]-quinoline-3,4-diamine (195 mg, 0.60 mmol) in ethanol (absolute grade, 4.2 mL) and the reaction mixture was heated to 75 °C for 2 hours. The volatiles were removed in vacuo and the residue was partitioned between EtOAc and saturated sodium hydro gencarbonate solution. The phases were separated and the aqueous phase was extracted with EtOAc (2x). The combined organic phases were washed (brine), dried (sodium sulfate) and concentrated in vacuo to afford 833 mg of a brown oil. This was purified by column chromatography on silica gel (gradient: 0 to 13% [2M N¾ in MeOH] in EtOAc) to afford 180 mg of a pale brown gum. The gum was dissolved in acetonitrile and water and lyophilised to afford 161 mg (74%) of (l-[(lS,3R)-3-(2,2,2-trifluoro-ethylamino)- cyclopentyl]-lH-imidazo[4,5-c]quinolin-2-yl}-methanol as a white solid. LCMS (Method A, ESI): RT = 2.00 min, m+H = 365.0; 1H NMR (400 MHz, CDC13): δ 9.21 (s, 1 H), 8.50 (d, 1 H), 8.26 (m, 1 H), 7.65 (m, 2 H), 5.48 (m, 1 H), 5.07 (m, 2 H), 3.63 (m, 1 H), 3.28 (m, 2 H), 2.65 (m, 2 H), 2.26 (m, 3 H), 2.02 (m, 1 H).
Example 11
Figure imgf000138_0001
N-{l-[(lS,3R)-3-(2,2,2-Trifluoro-ethylamino)-cyclopentyl]-lH-imidazo[4,5-c]quinolin-2- ylmethyl} -methanesulfonamide
N-tert-Butylcarbamate-N-{l-[(lS,3R)-3-(2,2,2-trifluoro-ethylamino)-cyclopentyl]-lH- imidazo[4,5-c]quinolin-2-ylmethyl} -methanesulfonamide A suspension of {l-[(lS,3R)-3-(2,2,2-trifluoro-ethylamino)-cyclopentyl]-lH-imidazo[4,5- c]quinolin-2-yl} -methanol (139 mg, 0.38 mmol) in anhydrous THF (3.8 mL), under an atmosphere of nitrogen was treated with tert-butyl N-methanesulfonylcarbamate (164 mg, 0.84 mmol), followed by triphenylphosphine (200 mg, 0.76 mmol) and then diisopropyl azodicarboxylate (150 μί, 0.76 mmol). The reaction mixture was stirred at room temperature overnight. The mixture was diluted with EtOAc and water and the phases separated. The aqueous layer was extracted with EtOAc (2x) and the combined organic phases washed (brine), dried (sodium sulfate) and concentrated in vacuo to afford 780 mg of a yellow oil. Purification by column chromatography on an Isolute® SCX-2 cartridge (eluting with MeOH and then 2M NH3 in MeOH) afforded 192 mg of a golden glassy solid. This was purified by filtration through a pad of silica gel (gradient: 0 to 2% [2M NH3 in MeOH] in EtOAc) to afford 169 mg (82%) of N-tert-butylcarbamate-N- { 1 - [( 1 S ,3R)-3 -(2,2,2-trifluoro-ethylamino)-cyclopentyl] - 1 H- imidazo[4,5-c]quinolin-2-ylmethyl}-methanesulfonamide as a colourless gum. LCMS (Method B, ESI): RT = 2.67 min, m+H = 542.0; 1H NMR (400 MHz, CDC13): δ 9.20 (s, 1 H), 8.40 (d, 1 H), 8.28 (d, 1 H), 7.66 (m, 2 H), 5.54-5.30 (m, 3 H), 3.68 (s, 3 H), 3.60 (m, 1 H), 3.30 (m, 2 H), 2.60 (m, 2 H), 2.33 (m, 1 H), 2.15 (m, 2 H), 1.98 (m, 1 H), 1.50 (s, 9 H).
N-{l-[(lS,3R)-3-(2,2,2-Trifluoro-ethylamino)-cyclopentyl]-lH-imidazo[4,5-c]quinolin-2- ylmethyl} -methanesulfonamide A solution of N-tert-butylcarbamate-N-{l-[(lS,3R)-3-(2,2,2-trifluoro-ethylamino)-cyclopentyl]- lH-imidazo[4,5-c]quinolin-2-ylmethyl}-methanesulfonamide (165 mg, 0.30 mmol) in TFA (3 mL) was stirred at room temperature for 10 minutes. The mixture was diluted with toluene and concentrated in vacuo. The residue was azeotroped (toluene) and purified by column chromatography using an Isolute® SCX-2 cartridge (eluting with MeOH and then 2M NH3 in MeOH) to afford a yellow glass. This was purified by column chromatography on silica gel (gradient: 0 to 10% [NH3 in MeOH] in EtOAc) to afford 120 mg of a colourless gum. The residue was treated with EtOAc, which gave rise to crystallization of a white solid (33 mg), which was removed by filtration. The resultant filtrate was concentrated and purified by column chromatography on silica gel (gradient: 0 to 10% [2M NH3 in MeOH] in EtOAc) to afford 110 mg of a white solid. Trituration (diethyl ether) afforded 90.0 mg (67%) of N-{l-[(lS,3R)-3- (2,2,2-trifluoro-ethylamino)-cyclopentyl]-lH-imidazo[4,5-c]quinolin-2-ylmethyl}- methanesulfonamide as a white solid. LCMS (Method A, ESI): RT = 2.28 min, m+H = 442.0; 1H NMR (400 MHz, CDC13): δ 9.24 (s, 1 H), 8.49 (d, 1 H), 8.29 (d, 1 H), 7.68 (m, 2 H), 5.81 (m, 1 H), 5.41 (m, 1 H), 4.93-4.72 (m, 2 H), 3.62 (m, 1 H), 3.27 (m, 2 H), 3.07 (s, 3 H), 2.61 (m, 2 H), 2.30 (m, 1 H), 2.17 (m, 2 H), 2.03 (m, 1 H).
Example 12
Figure imgf000139_0001
Trans [4-(2-Aminomethyl-imidazo[4,5-c]quinolin-l-yl)-cyclohexyl]-acetonitrile Trans [4-(2-Chloromethyl-imidazo[4,5-c]quinolin-l-yl)-cyclohexyl]-acetonitrile
A solution of trans [4-(3-amino-quinolin-4-ylamino)-cyclohexyl]-acetonitrile (500 mg, 1.80 mmol) in acetic acid (1.5 mL) was treated with 2-chloro-l,l,-trimethyoxyethane (0.48 mL, 3.60 mmol). The mixture was heated to 125 °C for 30 minutes. The solvent was removed in vacuo and the residue partitioned between ethyl acetate and saturated sodium hydrogencarbonate solution. The aqueous phase was extracted with ethyl acetate (2x) and the combined organic phases washed (saturated sodium hydrogencarbonate solution and brine), dried (sodium sulfate) and concentrated. Crude trans [4-(2-chloromethyl-imidazo[4,5-c]quinolin-l-yl)-cyclohexyl]- acetonitrile was isolated as a dark green foam solid and was used without further purification. LCMS (Method B, ESI): RT = 2.61 min, m+H = 339 and 341.
Trans [4-(2-Azidomethyl-imidazo[4,5-c]quinolin-l-yl)-cyclohexyl]-acetonitrile
A solution of trans [4-(2-chloromethyl-imidazo[4,5-c]quinolin-l-yl)-cyclohexyl]-acetonitrile (assumed to be 1.80 mmol) in DMF (18 mL) was treated with sodium azide (360 mg, 5.50 mmol) and stirred at room temperature for 4 hours and then allowed to stand over night. Water was added and the mixture extracted with ethyl acetate (3x). The combined organic extracts were washed (brine), dried (sodium sulfate) and concentrated to provide crude trans [4-(2- azidomethyl-imidazo[4,5-c]quinolin-l-yl)-cyclohexyl]-acetonitrile as a brown oil, which was used for the next step without purification. LCMS (Method B, ESI): RT = 2.52 min, m+H = 346.
Trans [4-(2-Aminomethyl-imidazo[4,5-c]quinolin-l-yl)-cyclohexyl]-acetonitrile
A solution of trans [4-(2-azidomethyl-imidazo[4,5-c]quinolin-l-yl)-cyclohexyl]-acetonitrile (assumed to be 1.80 mmol) in THF (9 mL) and ethanol (9 mL, IMS grade) was treated with palladium on carbon (180 mg, 0.18 mmol, 10% Pd) under nitrogen. The mixture was purged with hydrogen and then stirred under an atmosphere of hydrogen for 90 minutes at room temperature. The mixture was filtered through a pad of Celite® and then concentrated to give a brown oil. Purification by column chromatography on silica gel (gradient: 0 to 20% [2M N¾ in MeOH] in ethyl acetate) afforded a golden gum which was dried in vacuo. Trans [4-(2- aminomethyl-imidazo[4,5-c]quinolin-l-yl)-cyclohexyl]-acetonitrile (265 mg, 46%) was isolated as a fawn coloured foam solid. LCMS (Method A, ESI): RT = 1.77 min, m+H = 320.1 ; 1H NMR (400 MHz, DMSO-d6, 80 °C): δ 9.11 (s, 1 H), 8.41 (d, 1 H), 8.16 (dd, 1 H), 7.69 (m, 2 H), 5.02 (m, 1 H), 4.19 (s, 2 H), 2.55 (d, 2 H), 2.43 (m, 1 H), 2.06 (m, 6 H), 1.53 (m, 2 H).
Example 13
Figure imgf000140_0001
Trans [l-(4-Cyanomethyl-cyclohexyl)-lH-imidazo[4,5-c]quinolin-2-ylmethyl]-carbamic acid methyl ester formic acid salt A stirred solution of trans [4-(2-aminomethyl-imidazo[4,5-c]quinolin-l-yl)-cyclohexyl]- acetonitrile (100 mg, 0.31 mmol) in THF (3 mL) was treated sequentially with DIPEA (82.0 μί, 0.47 mmol) and a solution of methyl chloroformate (29.0 μί, 0.38 mmol) in THF (0.1 mL) at 0 °C. Stirring was continued for 90 minutes. The mixture was diluted with ethyl acetate and washed (water and brine), dried (sodium sulfate) and concentrated. The residue was purified by reverse phase HPLC (5 to 95% MeCN in H20 + 0.1% HC02H) and the relevant fractions combined and lyophilised to provide 31.0 mg (26%) of trans [l-(4-cyanomethyl-cyclohexyl)-lH- imidazo[4,5-c]quinolin-2-ylmethyl]-carbamic acid methyl ester formic acid salt as a white solid. LCMS (Method C, ESI): RT = 5.22 min, m+H = 378.2; 1H NMR (400 MHz, DMSO-d6, 80 °C): δ 9.12 (s, 1 H), 8.42 (d, 1 H), 8.24 (br s, 0.4 H, formate), 8.17 (d, 1 H), 7.70 (m, 2 H), 7.41 (br s, 1 H), 4.95 (m, 1 H), 4.70 (d, 2 H), 3.62 (d, 3 H), 2.55 (d, 2 H), 2.41 (m, 2 H), 2.07 (m, 6 H), 1.52 (m, 2 H).
Example 14
Figure imgf000141_0001
4-[2-((R)- 1 -Hydro xy-ethyl)-imidazo[4,5-c]quinolin- 1 -yl]-piperidine- 1 -carboxylic acid tert-butyl ester
4-(3-Nitro-quinolin-4-ylamino)-piperidine-l -carboxylic acid tert-butyl ester
A mixture of 4-amino-l-boc-piperidine (441 mg, 2.20 mmol), DIPEA (514 μΕ, 3.00mmol) and 4-chloro-3-nitro-quinoline (500 mg, 2.00 mmol) in propan-2-ol (8 mL) was heated to reflux for 2 hours. After cooling, the resulting precipitate was collected, washed (propan-2-ol and diethyl ether) and air dried to provide 755 mg of 4-(3-nitro-quinolin-4-ylamino)-piperidine-l -carboxylic acid tert-butyl ester as a yellow solid. LCMS (Method B, ESI): RT = 3.37 min, m+H = 373.2; 1H NMR (400 MHz, CDC13): δ 9.38 (m, 2 H), 8.15 (d, 1 H), 8.03 (dd, 1 H), 7.79 (m, 1 H), 7.53 (m, 1 H), 4.35 (m, 1 H), 4.06 (m, 2 H), 3.04 (m, 2 H), 2.15 (m, 2 H), 1.71 (m, 2 H), 1.48 (s, 9 H).
4-(3-Amino-quinolin-4-ylamino)-piperidine-l -carboxylic acid tert-butyl ester A solution of 4-(3-nitro-quinolin-4-ylamino)-piperidine-l-carboxylic acid tert-butyl ester (745 mg, 2.00 mmol) in EtOAc (10 mL) and THF (10 mL) was treated with palladium on carbon (106 mg, 0.10 mmol, 10% Pd) under a nitrogen atmosphere. The mixture was evacuated and purged with hydrogen and then stirred under an atmosphere of hydrogen for 2½ hours. The mixture was filtered through Celite® and the filtrate concentrated in vacuo to provide 729 mg of 4-(3-amino- quinolin-4-ylamino)-piperidine-l-carboxylic acid tert-butyl ester as an orange residue. The product was used without further purification. LCMS (Method B, ESI): RT = 2.52 min, m+H = 343.2; 1H NMR (400 MHz, DMSO-d6): δ 8.40 (s, 1 H), 7.99 (dd, 1 H), 7.73 (dd, 1 H), 7.35 (m, 2 H), 5.09 (s, 2 H), 4.52 (d, 1 H), 3.91 (m, 2 H), 2.67 (m, 2 H), 1.75 (br d, 2 H), 1.43 (m, 2 H), 1.40 (s, 9 H).
4-[2-((R)- 1 -Hydro xy-ethyl)-imidazo[4,5-c]quinolin- 1 -yl]-piperidine- 1 -carboxylic acid tert-butyl ester
A suspension of triethyloxonium tetrafluoroborate (1.48 g, 7.80 mmol) in DCM (10 mL) was treated with (R)-(+)-lactamide (713 mg, 8.00 mmol) at room temperature and stirred for 2 hours. The volatiles were removed in vacuo and the resulting residue dissolved in ethanol (absolute grade, 5 mL) and stirred for 30 minutes. This was added to a stirred solution of 4-(3-amino- quinolin-4-ylamino)-piperidine-l -carboxylic acid tert-butyl ester (assumed to be 2.00 mmol) in ethanol (absolute grade, 10 mL) and heated to reflux for 10 minutes. After cooling using an ice bath, the solvent was removed in vacuo and the resulting residue dissolved ethyl acetate. The mixture was washed (saturated sodium hydro gencarbonate solution, water and brine), dried (sodium sulfate) and concentrated in vacuo. Purification by column chromatography on silica gel (gradient: 0 to 10% MeOH in DCM) afforded 684 mg (86%) of 4-[2-((R)-l-hydroxy-ethyl)- imidazo[4,5-c]quinolin-l-yl]-piperidine-l-carboxylic acid tert-butyl ester as an off-white foam solid. LCMS (Method A, ESI): RT = 3.07 min, m+H = 397.1; 1H NMR (400 MHz, DMSO-d6, 80 °C): δ 9.16 (s, 1 H), 8.34 (d, 1 H), 8.18 (dd, 1 H), 7.68 (m, 1 H), 7.58 (m, 1 H), 5.46 (br s, 1 H), 5.36 (m, 1 H), 5.23 (m, 1 H), 4.27 (m, 2 H), 3.07 (m, 2 H), 2.56 (m, 2 H), 2.04 (br t, 2 H), 1.71 (d, 3 H), 1.52 (s, 9 H).
Example 15
Figure imgf000143_0001
(R)- 1 -( 1 -Piperidin-4-yl- 1 H-imidazo [4,5 -c] quino lin-2-yl)-ethano 1
A solution of 4-[2-((R)-l-hydroxy-ethyl)-imidazo[4,5-c]quinolin-l-yl]-piperidine-l-carboxylic acid tert-butyl ester (650 mg, 1.64 mmol) in TFA (5 mL) was stirred at room temperature for 30 minutes. The mixture was concentrated in vacuo and the residue purified by column chromatography using an Isolute® SCX-2 cartridge (gradient: MeOH to 2M NH3 in MeOH) to provide 435 mg (90%) of (R)-l-(l-piperidin-4-yl-lH-imidazo[4,5-c]quinolin-2-yl)-ethanol as an off-white foam. LCMS (Method A, ESI): RT = 0.97 min, m+H =297.0; 1H NMR (400 MHz, DMSO-d6, 80 °C): δ 9.14 (s, 1 H), 8.80 (d, 1 H), 8.16 (m, 1 H), 7.68 (m, 2 H), 5.44 (br s, 1 H), 5.21 (m, 2 H), 3.23 (m, 3 H), 2.76 (m, 2 H), 2.57 (m, 2 H), 1.94 (m, 2 H), 1.70 (d, 3 H).
Example 16
Figure imgf000143_0002
3- (4-[2-((R)- l-Hydroxy-ethyl)-imidazo[4,5-c]quinolin- 1 -yl]-piperidin- 1 -yl} -propionitrile
A solution of (R)-l-(l-piperidin-4-yl-lH-imidazo[4,5-c]quinolin-2-yl)-ethanol (75.0 mg, 0.25 mmol) in ethanol (2 mL, IMS grade) was treated with acrylonitrile (33.0 μί, 0.51 mmol) at reflux for 2 hours. After cooling, the mixture was concentrated onto diatomaceous earth and purified by column chromatography on silica gel (gradient: 0 to 15% [2M NH3 in MeOH] in ethyl acetate) to afford a colourless residue. The residue was triturated (diethyl ether) and dried at 50 °C under vacuum to leave 69.0 mg (79%) of 3-{4-[2-((R)-l-hydroxy-ethyl)-imidazo[4,5- c]quinolin-l-yl]-piperidin-l-yl} -propionitrile as a white solid. LCMS (Method A, ESI): RT = 1.64 min, m+H = 350.1; 1H NMR (400 MHz, DMSO-d6, 80 °C): δ 9.15 (s, 1 H), 8.84 (d, 1 H), 8.16 (dd, 1 H), 7.74 (m, 1 H), 7.66 (m, 1 H), 5.47 (br d, 1 H), 5.20 (m, 2 H), 3.20 (m, 2 H), 2.75 (m, 6 H), 2.39 (m, 2 H), 2.03 (m, 2 H), 1.70 (d, 3 H).
Example 17
Figure imgf000144_0001
(R)- 1 - { 1 -[ 1 -(2,2,2-Trifluoro-ethyl)-piperidin-4-yl]- lH-imidazo[4,5-c]quinolin-2-yl} -ethanol
A mixture of (R)-l-(l-piperidin-4-yl-lH-imidazo[4,5-c]quinolin-2-yl)-ethanol (75.0 mg, 250 μιηοΐ), 2,2,2-trifluoroetfiyl trifluoromethanesulfonate (87.0 mg, 375 μιηοΐ) and triethylamine (61.0 μί, 438 μιηοΐ) in THF (3 mL) was heated to 50 °C for 18 hours. After cooling, the mixture was diluted with ethyl acetate and washed (saturated sodium hydro gencarbonate solution, water and brine), dried (sodium sulfate) and concentrated. Purification by column chromatography on silica gel (gradient: 0 to 15% [2M N¾ in MeOH] in ethyl acetate) afforded a colourless residue. The residue was triturated (pentane) to provide 61.0 mg (64%) of (R)-1-{1- [l-(2,2,2-trifluoro-ethyl)-piperidin-4-yl]-lH-imidazo[4,5-c]quinolin-2-yl}-ethanol as a white solid. LCMS (Method A, ESI): RT = 2.94 min, m+H = 379.1; 1H NMR (400 MHz, DMSO-d6, 80 °C): δ 9.15 (s, 1 H), 8.79 (d, 1 H), 8.17 (dd, 1 H), 7.66 (m, 2 H), 5.48 (br d, 1 H), 5.20 (m, 2 H), 3.32 (q, 2 H), 3.21 (m, 2 H), 2.74 (m, 4 H), 2.03 (m, 2 H), 1.70 (d, 3 H).
Example 18
Figure imgf000144_0002
Racemic cis (3-[2-((R)- 1 -Hydro xy-ethyl)-imidazo[4,5-c]quinolin- 1 -yl]-cyclopentyl} -carbamic acid tert-butyl ester
Racemic cis [3 -(3 -Nitro-quinolin-4-ylamino)-cyclopentyl] -carbamic acid tert-butyl ester A mixture of racemic cis (3-amino-cyclopentyl)-carbamic acid tert-butyl ester (prepared following the methods outlined in J. Org. Chem. 2004, 69, 4538; Tetrahedron 1997, 53, 3347; WO2008/065021 ; WO94/17090; and Org. Lett. 2000, 2, 4169) (441 mg, 2.20 mmol), DIPEA (1.03 mL, 6.00 mmol) and 4-chloro-3-nitro-quinoline (500 mg, 2.00 mmol) in propan-2-ol (8 mL) was heated to reflux for 3 hours. The resulting mixture was filtered and concentrated in vacuo. The residue was purified by column chromatography on silica gel (gradient: 0 to 50% ethyl acetate in cyclohexane) to afford 548 mg (73%) of racemic cis [3-(3-nitro-quinolin-4- ylamino)-cyclopentyl]-carbamic acid tert-butyl ester as a yellow solid. LCMS (Method B, ESI): RT = 3.07 min, m+H = 373.2; 1H NMR (400 MHz, CDC13): δ 9.83 (br d, 1 H), 9.37 (s, 1 H), 8.25 (d, 1 H), 8.01 (d, 1 H), 7.78 (m, 1 H), 7.50 (m, 1 H), 4.67 (m, 2 H), 4.10 (m, 1 H), 2.73 (m, 1 H), 2.21 (m, 2 H), 1.97 (m, 1 H), 1.76 (m, 2 H), 1.45 (s, 9 H).
Racemic cis [3-(3-Amino-quinolin-4-ylamino)-cyclopentyl]-carbamic acid tert-butyl ester
A solution of racemic cis [3-(3-nitro-quinolin-4-ylamino)-cyclopentyl]-carbamic acid tert-butyl ester (540 mg, 1.45 mmol) in ethyl acetate (10 mL) and THF (10 mL) was treated with palladium on carbon (77.0 mg, 72.0 μιηοΐ, 10%> Pd) under a nitrogen atmosphere. The mixture was evacuated and purged with hydrogen and then stirred under an atmosphere of hydrogen for 2 hours. The mixture was filtered through Celite® and the filtrate concentrated in vacuo to provide 539 mg of racemic cis [3-(3-amino-quinolin-4-ylamino)-cyclopentyl]-carbamic acid tert-butyl ester as an orange residue. The product was used without further purification. LCMS (Method B, ESI): RT = 2.47 min, m+H = 343.2; 1H NMR (400 MHz, DMSO-d6): δ 8.39 (s, 1 H), 8.01 (m, 1 H), 7.73 (m, 1 H), 7.35 (m, 2 H), 6.96 (m, 1 H), 5.02 (br s, 2 H), 4.68 (d, 1 H), 3.87 (m, 1 H), 3.70 (m, 1 H), 2.17 (m, 1 H), 1.70 (m, 2 H), 1.60 (m, 2 H), 1.41 (m, 1 H), 1.38 (s, 9 H).
Racemic cis (3-[2-((R)- 1 -Hydro xy-ethyl)-imidazo[4,5-c]quinolin- 1 -yl]-cyclopentyl} -carbamic acid tert-butyl ester A stirred suspension of triethyloxonium tetrafluoroborate (1.14 g, 6.01 mmol) in DCM (5 mL) was treated with (R)-(+)-lactamide (547 mg, 6.14 mmol) at room temperature for 2 hours. The volatiles were removed in vacuo and the resulting residue dissolved in ethanol (absolute grade, 5 mL) and stirred for 30 minutes. This was added to a stirred solution of racemic cis [3-(3-amino- quinolin-4-ylamino)-cyclopentyl]-carbamic acid tert-butyl ester (526 mg, 1.54 mmol) in ethanol (absolute grade, 10 mL) and heated to reflux for 15 minutes. After cooling using an ice bath, the solvent was removed in vacuo and the resulting residue dissolved ethyl acetate. The mixture was washed (saturated sodium hydro gencarbonate solution, water and brine), dried (sodium sulfate) and concentrated in vacuo. Purification by column chromatography on silica gel (gradient: 0 to 10% MeOH in DCM) afforded 321 mg (53%) of racemic cis (3-[2-((R)-l-hydroxy-ethyl)- imidazo[4,5-c]quinolin-l-yl]-cyclopentyl}-carbamic acid tert-butyl ester as a beige foam solid. LCMS (Method A, ESI): RT = 2.95 min, m+H = 397.1; 1H NMR (400 MHz, DMSO-d6, 80 °C): δ 9.14 (s, 1 H), 8.43 (t, 1 H), 8.16 (dd, 1 H), 7.70 (m, 2 H), 6.91 (m, 1 H), 5.63 (m, 1 H), 5.42 (m, 1 H), 5.24 (m, 1 H), 4.11 (m, 1 H), 2.40 (m, 2 H), 2.20 (m, 3 H), 1.69 (dd, 3 H), 1.42 (d, 9 H).
Example 19
Figure imgf000146_0001
Racemic cis (R)- 1 -[ 1 -(3-Amino-cyclopentyl)- lH-imidazo[4,5-c]quinolin-2-yl]-ethanol
A solution of racemic cis {3-[2-((R)-l-hydroxy-ethyl)-imidazo[4,5-c]quinolin-l-yl]- cyclopentyl}-carbamic acid tert-butyl ester (250 mg, 0.63 mmol) in TFA (5 mL) was stirred at room temperature for 30 minutes. The mixture was concentrated in vacuo and the residue purified by column chromatography using an Isolute® SCX-2 cartridge (eluting MeOH to 2M NH3 in MeOH). Subsequent purification by column chromatography on silica gel (gradient: 0 to 10% [2M NH3 in MeOH] in DCM) followed by trituration (diethyl ether) provided 172 mg (92%) of racemic cis (R)-l-[l-(3-amino-cyclopentyl)-lH-imidazo[4,5-c]quinolin-2-yl]-ethanol as an off-white solid. LCMS (Method A, ESI): RT = 1.49 min, m+H = 297.0; 1H NMR (400
MHz, DMSO-d6, 80 °C): δ 9.14 (s, 1 H), 8.64 (d, 1 H), 8.15 (m, 1 H), 7.68 (m, 2 H), 5.62 (m, 1 H), 5.43 (br s, 1 H), 5.27 (m, 1 H), 3.59 (m, 1 H), 2.55 (m, 1 H), 2.41 (m, 1 H), 2.20 (m, 3 H), 1.87 (m, 1 H), 1.69 (dd, 3 H). Example 20
Figure imgf000147_0001
Racemic cis 3 - {3 - [2-((R)- 1 -Hydro xy-ethyl)-imidazo [4,5 -c] quino lin- 1 -yl] -cyclopentylamino } - propionitrile A solution of racemic cis (R)-l-[l-(3-amino-cyclopentyl)-lH-imidazo[4,5-c]quinolin-2-yl]- ethanol (140 mg, 0.47 mmol) in ethanol (3 mL, IMS grade) was treated with acrylonitrile (62.0 μί, 0.94 mmol) and heated at reflux for 2 hours. After cooling, the mixture was concentrated onto diatomaceous earth and purified by column chromatography on silica gel (gradient: 0 to 15% [2M N¾ in MeOH] in ethyl acetate). The isolated product was re-purified by column chromatography on silica gel (gradient: 0 to 10% MeOH in DCM) to afford a colourless residue. The residue was dried at 50 °C under vacuum to leave 54.0 mg (33%) of racemic cis 3-{3-[2- ((R)-l-hydroxy-ethyl)-imidazo[4,5-c]quinolin-l-yl]-cyclopentylamino}-propionitrile as an off- white foam solid. LCMS (Method A, ESI): RT = 1.60 min, m+H = 350.0; 1H NMR (400 MHz,
DMSO-d6): δ 9.19 (s, 1 H), 8.71 (t, 1 H), 8.17 (dd, 1 H), 7.74 (m, 2 H), 5.79 (t, 1 H), 5.60 (m, 1 H), 5.25 (m, 1 H), 3.40 (m, 1 H), 2.82 (m, 2 H), 2.66 (m, 2 H), 2.45 (m, 2 H), 2.15 (m, 3 H), 1.96 (m, l H), 1.68 (dd, 3 H).
Example 21
Figure imgf000147_0002
Trans (4-[2-((R)- 1 -Hydro xy-ethyl)- 1 ,3 ,5 ,9-tetraaza-cyclopenta[a]naphthalen- 1 -yl]-cyclohexyl} - carbamic acid tert-butyl ester Trans [4-(3-Nitro-[l,5]naphthyridin-4-ylamino)-cyclohexyl]-carbamic acid tert-butyl ester
A mixture of trans (4-amino-cyclohexyl)-carbamic acid tert-butyl ester (394 mg, 1.80 mmol), DIPEA (0.44 mL, 2.50 mmol) and 4-chloro-3-nitro-[l,5]naphthyridine (350 mg, 1.67 mmol) in propan-2-ol (6 mL) was heated to 120 °C using microwave irradiation for 20 minutes. After cooling, the resulting precipitate was isolated by filtration and the solid washed (propan-2-ol) and dried to afford 233 mg (36%) of trans [4-(3-nitro-[l,5]naphthyridin-4-ylamino)-cyclohexyl]- carbamic acid tert-butyl ester as a pale yellow solid. The above filtrate was concentrated in vacuo and the residue taken up in to ethyl acetate. The mixture was washed (saturated sodium hydro gencarbonate solution and brine), dried (sodium sulfate) and concentrated to provide an additional 238 mg (37%) of trans [4-(3-nitro-[l,5]naphthyridin-4-ylamino)-cyclohexyl]-carbamic acid tert-butyl ester as a yellow solid. LCMS (Method B, ESI): RT = 3.87 min, m+H = 388; 1H NMR (400 MHz, DMSO-d6): δ 9.44 (br s, 1 H), 9.18 (s, 1 H), 8.92 (dd, 1 H), 8.29 (dd, 1 H), 7.86 (dd, 1 H), 6.78 (m, 1 H), 5.13 (br s, 1 H), 2.13 (m, 2 H), 1.85 (m, 2 H), 1.54-1.51 (m, 2 H), 1.39 (s, 9 H), 1.33 (m, 2 H). Trans [4-(3-Amino-[l,5]naphthyridin-4-ylamino)-cyclohexyl]-carbamic acid tert-butyl ester
A mixture of trans [4-(3-nitro-[l,5]naphthyridin-4-ylamino)-cyclohexyl]-carbamic acid tert-butyl ester (471 mg, 1.20 mmol) in ethanol (IMS grade, 7.3 mL) and water (2.4 mL) was treated with ammonium chloride (390 mg, 7.30 mmol), followed by iron powder (273 mg, 4.90 mmol) and the reaction mixture was heated at 80 °C for 3½ hours. After cooling, the iron residue was collected on a pad of Celite® and washed several times with ethanol (IMS grade) and water (3: 1). The filtrate was concentrated under vacuum and the residue partitioned between saturated sodium hydro gencarbonate solution and ethyl acetate. The aqueous phase was extracted with ethyl acetate (3 x) and the combined organic phases washed (brine), dried (sodium sulfate) and concentrated. Purification by column chromatography on silica gel (gradient: 0 to 20% [20%(2M NH3 in MeOH) in ethyl acetate] in ethyl acetate) afforded 291 mg (68%) of trans [4-(3-amino- [l,5]naphthyridin-4-ylamino)-cyclohexyl]-carbamic acid tert-butyl ester as a yellow solid. LCMS (Method B, ESI): RT = 2.53 min, m+H = 358; 1H NMR 400 MHz, CDC13): δ 8.72 (dd, 1 H), 8.42 (s, 1 H), 8.21 (dd, 1 H), 7.41 (dd, 1 H), 5.74 (br s, 1 H), 4.35 (br s, 1 H), 3.68 (s, 2 H), 3.48 (br s, 1 H), 2.04 (m, 2 H), 1.70 (br s, 2 H), 1.43 (m, 11 H), 1.24 (m, 2 H). Trans (4-[2-((R)- 1 -Hydro xy-ethyl)- 1 ,3,5,9-tetraaza-cyclopenta[a]naphthalen- 1 -yl]-cyclohexyl} - carbamic acid tert-butyl ester A stirred suspension of triethyloxonium tetrafluoroborate (465 mg, 2.45 mmol) in THF (5 mL) was treated with (R)-(+)-lactamide (237 mg, 2.70 mmol) at room temperature for 2 hours. The volatiles were removed in vacuo and the resulting residue dissolved in ethanol (absolute grade, 4 mL) and added to a stirred solution of trans [4-(3-amino-[l,5]naphthyridin-4-ylamino)- cyclohexyl]-carbamic acid tert-butyl ester (291 mg, 815 μιηοΐ) in ethanol (absolute grade, 6 mL) and heated to reflux for 2 hours. After cooling, the solvent was removed in vacuo and the resulting residue dissolved ethyl acetate. The mixture was washed (saturated sodium hydro gencarbonate solution and brine), dried (sodium sulfate) and concentrated in vacuo. Purification by column chromatography on silica gel (gradient: 0 to 30% [20%(2M N¾ in MeOH) in ethyl acetate] in ethyl acetate) followed by trituration (diethyl ether) provided 159 mg (47%) of trans {4-[2-((R)-l-hydroxy-ethyl)-l,3,5,9-tetraaza-cyclopenta[a]naphthalen-l-yl]- cyclohexyl}-carbamic acid tert-butyl ester as a pale green solid. LCMS (Method A, ESI): RT = 7.77 min, m+H = 412; 1H NMR (400 MHz, DMSO-d6): δ 9.29 (s, 1 H), 9.06 (br s, 1 H), 8.54 (dd, 1 H), 7.76 (dd, 1 H), 6.88 (d, 1 H), 5.87 (br d, 1 H), 5.24 (m, 1 H), 4.95 (m, 1 H), 3.68 (m, 1 H), 3.24 (m, 2 H), 1.97 (m, 2 H), 1.80 (m, 2 H), 1.69 (d, 3 H), 1.46 (m, 11 H).
Example 22
Figure imgf000149_0001
Trans (R)- 1 -[ 1 -(4-Amino-cyclohexyl)- 1 H- 1 ,3 ,5 ,9-tetraaza-cyclopenta[a]naphthalen-2-yl]- ethanol Trans (4-[2-((R)- 1 -hydro xy-ethyl)- 1 ,3 ,5 ,9-tetraaza-cyclopenta[a]naphthalen- 1 -yl]-cyclohexyl} - carbamic acid tert-butyl ester (148 mg, 360 μιηοΐ) was treated with DCM (8 mL) and TFA (2 mL) and stirred at room temperature for 45 minutes. The solvent was removed under vacuum and the resulting residue purified by column chromatography using an Isolute® SCX-2 cartridge (gradient: MeOH to 2M N¾ in MeOH). The isolated product was azeotroped (diethyl ether) to afford 104 mg (92%) of trans (R)-l-[l-(4-amino-cyclohexyl)-lH-l,3,5,9-tetraaza- cyclopenta[a]naphthalen-2-yl] -ethanol as a white solid. LCMS (Method A, ESI): RT = 1.60 min, m+H = 312.0; 1H NMR (400 MHz, DMSO-d6): δ 9.28 (s, 1 H), 9.05 (d, 1 H), 8.53 (dd, 1 H), 7.74 (dd, 1 H), 5.85 (br s, 1 H), 5.24 (m, 1 H), 4.96 (m, 1 H), 3.24 (m, obscured by water), 3.01 (m, l H), 1.94 (m, 2 H), 1.75 (m, 2 H), 1.69 (d, 3 H), 1.32 (m, 2 H).
Example 23
Figure imgf000150_0001
Trans 3-{4-[2-((R)-l-Hydroxy-ethyl)-l,3,5,9-tetraaza-cyclopenta[a]naphthalen-l-yl]- cyclohexylamino } -propionitrile
Trans (R)- 1 -[ 1 -(4-amino-cyclohexyl)- 1 H- 1 ,3 ,5 ,9-tetraaza-cyclopenta[a]naphthalen-2-yl]-ethanol (50.0 mg, 161 μηιοΐ) was treated with MeOH (2 mL), triethylamine (44.7 μί, 322 μηιοΐ) and acrylonitrile (11.6 μί, 177 μηιοΐ) and then stirred for 18 hours at room temperature. The solvent was removed in vacuo and the residue purified by reverse phase HPLC (gradient: 5 to 40% MeCN in H20 + 0.1% HC02H). The combined fractions were loaded on to an Isolute® SCX-2 cartridge and purified (gradient: MeOH to 2M N¾ in MeOH). The isolated product was triturated (diethyl ether) to provide 24.5 mg (42%) of trans 3-{4-[2-((R)-l-hydroxy-ethyl)- l,3,5,9-tetraaza-cyclopenta[a]naphthalen-l-yl]-cyclohexylamino}-propionitrile as a pale yellow solid. LCMS (Method A, ESI): RT = 1.79 min, m+H = 365.1; 1H NMR (400 MHz, DMSO-d6): δ 9.28 (s, 1 H), 9.07 (s, 1 H), 8.53 (dd, 1 H), 7.75 (dd, 1 H), 5.84 (br d, 1 H), 5.25 (m, 1 H), 4.98 (m, 1 H), 3.23 (m, 2 H), 2.87 (m, 3 H), 2.62 (t, 2 H), 2.07 (m, 2 H), 1.79 (m, 2 H), 1.69 (d, 3 H), 1.29 (m, 2 H).
Example 24
Figure imgf000150_0002
Trans (R)- 1 - { 1 -[4-(2,2,2-Trifluoro-ethylamino)-cyclohexyl]- 1 H- 1 ,3 ,5 ,9-tetraaza- cyclopenta[a]naphthalen-2-yl}-ethanol
Trans (R)- 1 -[ 1 -(4-amino-cyclohexyl)- 1 H- 1 ,3 ,5 ,9-tetraaza-cyclopenta[a]naphthalen-2-yl]-ethanol (46.0 mg, 148 μηιοΐ) was treated with THF (2 mL), triethylamine (41.0 μί, 296 μηιοΐ) and 2,2,2- trifluoro ethyl trifluoromethanesulfonate (22.4 μΐ^, 155 μηιοΐ) and then stirred at room temperature for 18 hours. The solvent was removed in vacuo and the residue purified by reverse phase HPLC (gradient: 5 to 40% MeCN in H20 + 0.1% HC02H). The combined fractions were loaded on to an Isolute® SCX-2 cartridge and purified (gradient: MeOH to 2M NH3 in MeOH). The isolated product was azeotroped (diethyl ether and ethyl acetate) to provide 16.5 mg (28%) of trans (R)-l-{l-[4-(2,2,2-trifluoro-ethylamino)-cyclohexyl]-lH-l,3,5,9-tetraaza- cyclopenta[a]naphthalen-2-yl}-ethanol. LCMS (Method A, ESI): RT = 2.15 min, m+H = 394.1 ; 1H NMR (400 MHz, DMSO-d6): δ 9.29 (s, 1 H), 9.03 (dd, 1 H), 8.53 (dd, 1 H), 7.75 (dd, 1 H), 5.85 (br d, 1 H), 5.26 (m, 1 H), 4.98 (m, 1 H), 3.33 (m, 2 H), 3.22 (m, 2 H), 2.90 (m, 1 H), 2.33 (m, l H), 2.09 (m, 2 H), 1.79 (m, 2 H), 1.69 (d, 3 H), 1.31 (m, 2 H).
Example 25
Figure imgf000151_0001
(R)-3-[2-((R)- 1 -Hydro xy-ethyl)-imidazo[4,5-c]quinolin- 1 -yl]-piperidine- 1 -carboxylic acid tert- butyl ester
(R)-3-(3-Nitro-quinolin-4-ylamino)-piperidine-l -carboxylic acid tert-butyl ester
A mixture of R-(-)-3-amino-l-boc-piperidine (441 mg, 2.20 mmol), DIPEA (514 μΕ, 3.00 mmol) and 4-chloro-3-nitro-quinoline (500 mg, 2.00 mmol) in propan-2-ol (8 mL) was heated to reflux for 2 hours. After cooling, the resulting precipitate was collected by filtration and washed with propan-2-ol and diethyl ether. The solid was air dried to provide 689 mg (92%) of (R)-3- (3-nitro-quinolin-4-ylamino)-piperidine-l -carboxylic acid tert-butyl ester as a yellow solid.
LCMS (Method B, ESI): RT = 3.42 min, m+H = 373.2; 1H NMR (400 MHz, CDC13): δ 9.48 (br d, 1 H), 9.39 (s, 1 H), 8.24 (br d, 1 H), 8.03 (d, 1 H), 7.79 (m, 1 H), 7.53 (m, 1 H), 4.33 (m, 1 H), 3.96 (br d, 1 H), 3.58 (m, 1 H), 3.37 (m, 2 H), 2.13 (m, 1 H), 1.83 (m, 2 H), 1.63 (m, 1 H), 1.45 (s, 9 H).
(R)-3-(3-Amino-quinolin-4-ylamino)-piperidine-l-carboxylic acid tert-butyl ester
A solution of (R)-3-(3-nitro-quinolin-4-ylamino)-piperidine-l-carboxylic acid tert-butyl ester (680 mg, 1.83 mmol) in ethyl acetate (10 mL) and THF (10 mL) was treated with palladium on carbon (96.0 mg, 91.0 μιηοΐ, 10% Pd) under a nitrogen atmosphere. The mixture was evacuated and purged with hydrogen and then stirred under an atmosphere of hydrogen for 2 hours. The mixture was filtered through Celite® and the filtrate concentrated in vacuo to provide 641 mg of crude (R)-3-(3-amino-quinolin-4-ylamino)-piperidine-l-carboxylic acid tert-butyl ester as an orange residue. The product was used without further purification. LCMS (Method B, ESI): RT = 2.41 min, m+H = 343.2; 1H NMR (400 MHz, CDC13): δ 8.56 (s, 1 H), 8.05 (d, 1 H), 7.86 (d, 1 H), 7.52 (m, 1 H), 7.43 (m, 1 H), 4.80 (br s, 1 H), 4.21 (br s, 1 H), 3.81 (m, 1 H), 3.74 (m, 1 H), 3.50 (m, 1 H), 3.35 (m, 2 H), 1.97 (m, 1 H), 1.78 (m, 2 H), 1.55 (m, 1 H), 1.43 (s, 9 H).
(R)-3-[2-((R)- 1 -Hydro xy-ethyl)-imidazo[4,5-c]quinolin- 1 -yl]-piperidine- 1 -carboxylic acid tert- butyl ester
A stirred suspension of triethyloxonium tetrafluoroborate (678 mg, 3.57 mmol) in DCM (5 mL) was treated with (R)-(+)-lactamide (326 mg, 3.66 mmol) at room temperature for 2 hours. The volatiles were removed in vacuo and the resulting residue dissolved in ethanol (absolute grade, 5 mL) and stirred for 30 minutes. This was added to a stirred solution of (R)-3-(3-amino-quinolin- 4-ylamino)-piperidine-l -carboxylic acid tert-butyl ester (310 mg, 0.90 mmol) in ethanol (absolute grade, 5 mL) at reflux. Heating was continued for 35 minutes. After cooling, the solvent was removed in vacuo and the resulting residue dissolved ethyl acetate. The mixture was washed (saturated sodium hydro gencarbonate solution, water and brine), dried (sodium sulfate) and concentrated in vacuo. Purification by column chromatography on silica gel (gradient: 0 to 10% MeOH in DCM) afforded 320 mg (89%) of (R)-3-[2-((R)-l-hydroxy-ethyl)-imidazo[4,5- c]quinolin-l-yl]-piperidine-l -carboxylic acid tert-butyl ester as an off-white foam solid. LCMS (Method A, ESI): RT = 3.13 min, m+H = 397.1; 1H NMR (400 MHz, DMSO-d6, 80 °C): δ 9.16 (s, 1 H), 8.48 (d, 1 H), 8.17 (m, 1 H), 7.71 (m, 2 H), 5.48 (br d, 1 H), 5.24 (m, 2 H), 4.35 (dd, 1 H), 4.11 (br d, 1 H), 3.74 (t, 1 H), 3.05 (td, 1 H), 2.63 (m, 1 H), 2.16 (m, 1 H), 1.97 (m, 1 H), 1.75 (m, 1 H), 1.71 (d, 3 H), 1.41 (s, 9 H).
Example 26
Figure imgf000153_0001
(R)- 1 -((R)- 1 -Piperidin-3 -yl- 1 H-imidazo [4,5 -c] quino lin-2-yl)-ethano 1
A solution of (R)-3-[2-((R)-l-hydroxy-ethyl)-imidazo[4,5-c]quinolin-l-yl]-piperidine-l- carboxylic acid tert-butyl ester (290 mg, 0.73 mmol) in TFA (5 mL) was stirred at room temperature for 30 minutes. The mixture was concentrated in vacuo and the residue purified by column chromatography using an Isolute® SCX-2 cartridge (gradient: MeOH to 2M NH3 in MeOH). Subsequent purification by column chromatography on silica gel (gradient: 0 to 10% [2M NH3 in MeOH] in DCM) provided 204 mg (94%) of (R)-1-((R)-1 -piperidin-3 -yl-lH- imidazo[4,5-c]quinolin-2-yl)-ethanol as a white foam solid. LCMS (Method A, ESI): RT = 1.54 min, m+H = 297.0; 1H NMR (400 MHz, DMSO-d6, 80 °C): δ 9.14 (s, 1 H), 8.60 (d, 1 H), 8.17 (dd, 1 H), 7.70 (m, 2 H), 5.38 (br s, 1 H), 5.21 (m, 2 H), 3.54 (t, 1 H), 3.26 (dd, 1 H), 3.01 (m, 2 H), 2.80 (m, 1 H), 2.58 (m, 1 H), 2.14 (br d, 1 H), 1.91 (m, 1 H), 1.79 (m, 1 H), 1.70 (d, 3 H).
Example 27
Figure imgf000153_0002
3- {(R)-3-[2-((R)- l-Hydroxy-ethyl)-imidazo[4,5-c]quinolin- 1 -yl]-piperidin- 1 -yl} -3-oxo- propionitrile
A solution of (R)-l-((R)-l-piperidin-3-yl-lH-imidazo[4,5-c]quinolin-2-yl)-ethanol (125 mg, 0.42 mmol) in MeCN (8 mL) was treated with cyanoacetic acid (43.0 mg, 0.51 mmol), N- hydroxybenzotriazole (HOBt) (79.0 mg, 0.59 mmol), 4-dimethylaminopyridine (DMAP) (77.0 mg, 0.63 mmol) and l-ethyl-3-(3-dimethylaminopropyl) carbodiimide hydrochloride (EDCI) (120 mg, 0.63 mmol) at room temperature and stirred for 18 hours. The mixture was purified by column chromatography using an Isolute® SCX-2 cartridge (gradient: MeOH to 2M NH3 in MeOH) and then by column chromatography on silica gel (gradient: 0 to 15% [2M NH3 in MeOH] in ethyl acetate). Further purification by column chromatography on silica gel (gradient: 0 to 10% MeOH in DCM) provided a pale yellow glass. Trituration (diethyl ether) and subsequent drying in vacuo at 50 °C provided 1 11 mg (73%) of 3-{(R)-3-[2-((R)-l-hydroxy- ethyl)-imidazo[4,5-c]quinolin-l-yl]-piperidin-l-yl}-3-oxo-propionitrile as a pale yellow solid. LCMS (Method C, ESI): RT = 4..31 min, m+H = 364.2; 1H NMR (400 MHz, DMSO-d6, 80 °C): δ 9.17 (s, 1 H), 8.46 (d, 1 H), 8.18 (dd, 1 H), 7.71 (m, 2 H), 5.55 (br s, 1 H), 5.22 (m, 2 H), 4.80-3.60 (br m, 6 H), 2.66 (m, 1 H), 2.16 (m, 1 H), 1.99 (m, 1 H), 1.83 (m, 1 H), 1.73 (d, 3 H).
Example 28
Figure imgf000154_0001
(R)-3-(2-Methyl-imidazo[4,5-c]quinolin-l-yl)-piperidine-l-carboxylic acid tert-butyl ester
A mixture of (R)-3-(3-amino-quinolin-4-ylamino)-piperidine-l-carboxylic acid tert-butyl ester (310 mg, 0.905 mmol) and triethyl orthoacetate (415 μί, 2.26 mmol) in acetic acid (5 mL) was heated to 90 °C for 4 hours. After cooling, the solvent was removed in vacuo and the residue was taken up in ethyl acetate. The mixture was washed (saturated sodium hydro gencarbonate solution, water and brine), dried (sodium sulfate) and concentrated to leave a yellow residue. Purification by column chromatography on silica gel (gradient: 0 to 10% MeOH in DCM) provided a gum which was azeotroped (diethyl ether) to afford 296 mg (89%) of (R)-3-(2- methyl-imidazo[4,5-c]quinolin-l-yl)-piperidine-l-carboxylic acid tert-butyl ester as a white foam solid. LCMS (Method C, ESI): RT = 6.57 min, m+H = 367.3; 1H NMR (400 MHz, DMSO- d6, 80 °C): δ 9.07 (s, 1 H), 8.39 (d, 1 H), 8.16 (m, 1 H), 7.66 (m, 2 H), 5.02 (m, 1 H), 4.31 (d, 1 H), 4.09 (d, 1 H), 3.54 (t, 1 H), 3.01 (m, 1 H), 2.78 (s, 3 H), 2.48 (m, 1 H), 2.22 (m, 1 H), 1.93 (m, 1 H), 1.76 (m, 1 H), 1.42 (s, 9 H). Example 29
Figure imgf000155_0001
2-Methyl- 1 -(R)-piperidin-3 -yl- 1 H-imidazo [4,5 -c] quino line
A solution of (R)-3-(2-methyl-imidazo[4,5-c]quinolin-l-yl)-piperidine-l-carboxylic acid tert- butyl ester (270 mg, 0.74 mmol) in TFA (5 mL) was stirred at room temperature for 30 minutes. The mixture was concentrated in vacuo and the residue purified by column chromatography using an Isolute® SCX-2 cartridge (gradient: MeOH to 2M NH3 in MeOH). Subsequent azeotroping (diethyl ether and pentane) provided 180 mg (91%) of 2-methyl-l-(R)-piperidin-3- yl-lH-imidazo[4,5-c]quinoline as a sticky solid. LCMS (Method A, ESI): RT = 1.19 min, m+H = 267.1; 1H NMR (400 MHz, DMSO-d6, 80 °C): δ 9.06 (s, 1 H), 8.51 (m, 1 H), 8.16 (m, 1 H), 7.68 (m, 2 H), 5.06 (m, 1 H), 3.28 (m, 3 H), 2.96 (m, 1 H), 2.77 (s, 3 H), 2.68 (m, 1 H), 2.38 (m, 1 H), 2.18 (m, 1 H), 1.89 (m, 1 H), 1.77 (m, 1 H).
Example 30
Figure imgf000155_0002
3-[(R)-3-(2-Methyl-imidazo[4,5-c]quinolin- 1 -yl)-piperidin- 1 -yl]-3-oxo-propionitrile
A solution of 2-methyl-l-(R)-piperidin-3-yl-lH-imidazo[4,5-c]quinoline (100 mg, 375 μιηοΐ) in MeCN (8 mL) was treated with cyanoacetic acid (38.0 mg, 451 μιηοΐ), HOBt (71.0 mg, 525 μιηοΐ), DMAP (69.0 mg, 563 μιηοΐ) and EDCI (107 mg, 563 μιηοΐ) at room temperature and stirred for 18 hours. The mixture was purified by column chromatography using an Isolute® SCX-2 cartridge (gradient: MeOH to 2M NH3 in MeOH) and then by column chromatography on silica gel (gradient: 0 to 15% [2M NH3 in MeOH] in ethyl acetate). Further purification by column chromatography on silica gel (gradient: 0 to 10% MeOH in DCM) provided a colourless glass. Trituration (diethyl ether) and subsequent drying in vacuo at 50 °C provided 64.0 mg (51%) of 3-[(R)-3-(2-methyl-imidazo[4,5-c]quinolm^ as a white solid. LCMS (Method A, ESI): RT = 2.00 min, m+H = 334.1 ; 1H NMR (400 MHz, DMSO-d6, 80 °C): δ 9.08 (s, 1 H), 8.39 (s, 1 H), 8.17 (m, 1 H), 7.66 (m, 2 H), 5.05 (br s, 1 H), 4.80-3.40 (br m, 6 H), 2.79 (s, 3 H), 2.49 (m, 1 H), 2.23 (m, 1 H), 1.96 (m, 1 H), 1.85 (m, 1 H).
Example 31
Figure imgf000156_0001
Trans N-[l-(4-Cyanomethyl-cyclohexyl)-lH- 1,3,5, 9-tetraaza-cyclopenta[a]naphthalen-2- ylmethyl] -methanesulfonamide Trans [ 1 -(4-Cyanomethyl-cyclohexyl)- 1 H- 1 ,3 ,5 ,9-tetraaza-cyclopenta[a]naphthalen-2-ylmethyl]- carbamic acid tert-butyl ester
A stirred solution of boc-glycinamide (701 mg, 4.00 mmol) in DCM (17 mL) was treated with triethyloxonium tetrafluoroborate (702 mg, 3.70 mmol) at room temperature for 2 hours. The volatiles were removed in vacuo and trans [4-(3-amino-[l,5]naphthyridin-4-ylamino)- cyclohexyl]-acetonitrile (346 mg, 1.23 mmol) dissolved in ethanol (absolute grade, 25 mL) was added to the residue. The mixture was stirred at 75 °C for 3 hours and left to stand at room temperature for 16 hours. The solvent was removed in vacuo and the resulting residue dissolved ethyl acetate. The mixture was washed (saturated sodium hydro gencarbonate solution, water and brine), dried (sodium sulfate) and concentrated in vacuo. Purification by column chromatography on silica gel (gradient: 0 to 10% [20% (2M NH3 in MeOH) in ethyl acetate] in ethyl acetate) afforded 387 mg (75%) of trans [l-(4-cyanomethyl-cyclohexyl)-lH- 1,3,5, 9-tetraaza- cyclopenta[a]naphthalen-2-ylmethyl]-carbamic acid tert-butyl ester. LCMS (Method B, ESI): RT = 3.27 min, m+H = 421.1; 1H NMR (400 MHz, DMSO-d6): δ 9.26 (s, 1 H), 9.06 (s, 1 H), 8.54 (dd, 1 H), 7.75 (dd, 1 H), 7.62 (br s, 1 H), 4.69 (m, 1 H), 4.67 (d, 2 H), 3.21 (m, 2 H), 2.60 (d, 2 H), 1.99 (m, 2 H), 1.79 (m, 1 H), 1.42 (m, 13 H).
Trans [4-(2-Aminomethyl- 1 ,3 ,5 ,9-tetraaza-cyclopenta[a]naphthalen- 1 -yl)-cyclohexyl]- acetonitrile A stirred solution of trans [ l-(4-cyanomethyl-cyclohexyl)-lH- 1,3,5, 9-tetraaza- cyclopenta[a]naphthalen-2-ylmethyl]-carbamic acid tert-butyl ester (290 mg, 690 μηιοΐ) and dioxane (7.5 mL) was treated with HCl in dioxane (4M, 7.5 mL) at room temperature for 2 hours. The solvent was removed in vacuo and the residue azeotroped (toluene). Purification by column chromatography using Isolute® SCX-2 cartridge (gradient: MeOH to 2M N¾ in MeOH) afforded trans [4-(2-aminomethyl-l,3,5,9-tetraaza-cyclopenta[a]naphthalen-l-yl)- cyclohexyl]-acetonitrile as a yellow gum. The product was used for the next step without further purification. LCMS (Method B, ESI): RT = 1.99 min, m+H = 321.
Trans N-[l-(4-Cyanomethyl-cyclohexyl)-lH- 1,3,5, 9-tetraaza-cyclopenta[a]naphthalen-2- ylmethyl] -methanesulfonamide
A stirred solution of trans [4-(2-aminomethyl-l,3,5,9-tetraaza-cyclopenta[a]naphthalen-l-yl)- cyclohexyl]-acetonitrile (114 mg, 356 μιηοΐ) in THF (3 mL) was treated with triethylamine (99.0 μί, 712 μιηοΐ) and methane sulfonylchloride (30.0 μί, 392 μιηοΐ) at 0 °C for 10 minutes. The mixture was partitioned between ethyl acetate and saturated sodium hydro gencarbonate solution. The organic phase was washed (brine), dried (sodium sulfate) and concentrated. Purification by column chromatography on silica gel (gradient: 0 to 30% [20% (2M N¾ in MeOH) in ethyl acetate] in ethyl acetate) afforded 77.1 mg (54%) of trans N-[l-(4-cyanomethyl-cyclohexyl)-lH- l,3,5,9-tetraaza-cyclopenta[a]naphthalen-2-ylmethyl]-methanesulfonamide as a white solid. LCMS (Method A, ESI): RT = 3.12 min, m+H = 399.0; 1H NMR (400 MHz, DMSO-d6): δ 9.29 (s, 1 H), 9.08 (br s, 1 H), 8.55 (dd, 1 H), 7.90 (s, 1 H), 7.77 (dd, 1 H), 4.74 (br s, 1 H), 4.71 (br s, 2 H), 3.21 (m, 2 H), 3.02 (s, 3 H), 2.60 (d, 2 H), 2.11-1.82 (m, 5 H), 1.42 (m, 2 H).
Example 32
Figure imgf000157_0001
Trans [ 1 -(4-Cyanomethyl-cyclohexyl)- 1 H- 1 ,3 ,5 ,9-tetraaza-cyclopenta[a]naphthalen-2-ylmethyl]- carbamic acid methyl ester
A stirred solution of trans [4-(2-aminomethyl-l,3,5,9-tetraaza-cyclopenta[a]naphthalen-l-yl)- cyclohexyl]-acetonitrile (114 mg, 356 μιηοΐ) in THF (3 mL) was treated with triethylamine (74.0 μΐ^, 534 μηιοΐ) and methyl chloro formate (30.0 μί, 392 μmol) at 0 °C for 10 minutes. The mixture was partitioned between ethyl acetate and saturated sodium hydro gencarbonate solution. The organic phase was washed (brine), dried (sodium sulfate) and concentrated. Purification by column chromatography on silica gel (gradient: 0 to 30% [20% (2M N¾ in MeOH) in ethyl acetate] in ethyl acetate) afforded 79.0 mg (59%) of trans [l-(4-cyanomethyl-cyclohexyl)-lH- l,3,5,9-tetraaza-cyclopenta[a]naphthalen-2-ylmethyl]-carbamic acid methyl ester as a white solid. LCMS (Method A, ESI): RT = 3.12 min, m+H = 379.1; 1H NMR (400 MHz, DMSO-d6): δ 9.27 (s, 1 H), 9.06 (br s, 1 H), 8.54 (dd, 1 H), 7.94 (br s, 1 H), 7.75 (dd, 1 H), 4.73 (d, 2 H), 4.63 (br s, 1 H), 3.60 (s, 3 H), 3.21 (m, 2 H), 2.60 (d, 2 H), 1.97 (m, 3 H), 1.80 (m, 2 H), 1.39 (m, 2 H).
Example 33
Figure imgf000158_0001
l-(l-Methanesulfonyl-4-methyl-piperidin-4-yl)-2-methyl-lH-imidazo[4,5-c]quinoline ( 1 -Methanesulfonyl-4-methyl-piperidin-4-yl)-(3 -nitro-quino lin-4-yl)-amine A mixture of l-methanesulfonyl-4-methyl-piperidin-4-ylamine hydrochloride (WO2006062063) (361 mg, 1.58 mmol), DIPEA (1.25 mL, 7.20 mmol) and 4-chloro-3-nitro-quinoline (300 mg, 1.44 mmol) in propan-2-ol (10 mL) was heated to 120 °C for 40 minutes using microwave irradiation. After cooling, the resulting precipitate was collected by filtration, washed with propan-2-ol and sucked dry to provide 352 mg (67%) of (l-methanesulfonyl-4-methyl-piperidin- 4-yl)-(3-nitro-quinolin-4-yl)-amine as a yellow solid. LCMS (Method B, ESI): RT = 2.94 min, m+H = 365.1; 1H NMR (400 MHz, DMSO-d6): δ 9.21 (s, 1 H), 8.33 (d, 1 H), 7.98 (m, 1 H), 7.89 (m, 1 H), 7.86 (s, 1 H), 7.68 (m, 1 H), 3.19 (m, 4 H), 2.87 (s, 3 H), 1.98 (m, 4 H), 1.31 (s, 3 H).
] f*4*-(l-Methanesulfonyl-4-methyl-piperidin-4-yl)-quinoline-3,4-diamine A solution of (l-methanesulfonyl-4-methyl-piperidin-4-yl)-(3-nitro-quinolin-4-yl)-amine (350 mg, 0.96 mmol) in ethyl acetate (10 mL) and THF (10 mL) was treated with palladium on carbon (50 mg, 10% Pd) under a nitrogen atmosphere. The mixture was evacuated and purged with hydrogen and then stirred under an atmosphere of hydrogen for 90 minutes. The mixture was filtered through Celite® and the filtrate concentrated in vacuo to provide 315 mg (98%>) of crude ] f*4*-(l-methanesulfonyl-4-methyl-piperidin-4-yl)-quinoline-3,4-diamine as a pale yellow foam. The product was used without further purification. LCMS (Method B, ESI): RT = 2.05 min, m+H = 335.2; 1H NMR (400 MHz, DMSO-d6): δ 8.49 (s, 1 H), 8.05 (d, 1 H), 7.74 (dd, 1 H), 7.35 (m, 2 H), 5.28 (s, 2 H), 4.12 (s, 1 H), 3.39 (m, 2 H), 2.93 (m, 2 H), 2.85 (s, 3 H), 1.88 (m, 2 H), 1.69 (m, 2 H), 1.12 (s, 3 H). l-(l-Methanesulfonyl-4-methyl-piperidin-4-yl)-2-methyl-lH-imidazo[4,5-c]quinoline A mixture of N*4!i:-(l-methanesulfonyl-4-methyl-piperidin-4-yl)-quinoline-3,4-diamine (50.0 mg, 149 μιηοΐ) and triethyl orthoacetate (274 μΐ^, 1.50 mmol) in ethanol (2 mL, IMS grade) was heated at 80 °C for 18 hours in a sealed vial. p-Toluenesulfonic acid (cat) was added and heating continued for 2 hours. After cooling, the mixture was concentrated in vacuo and the residue dissolved in ethyl acetate. The mixture was washed (saturated sodium hydro gencarbonate solution and brine), dried (sodium sulfate) and concentrated. The residue was purified by column chromatography on silica gel (gradient: 0 to 20% MeOH in ethyl acetate) and the isolated product azeotroped (diethyl ether). l-(l-Methanesulfonyl-4-methyl-piperidin-4-yl)-2-methyl-lH- imidazo[4,5-c]quinoline (28.1 mg, 53%) was isolated as a white solid. LCMS (Method C, ESI): RT = 4.76 min, m+H = 359.2; 1H NMR (400 MHz, DMSO-d6): δ 9.13 (s, 1 H), 8.63 (m, 1 H), 8.17 (m, 1 H), 7.66 (m, 2 H), 3.54 (m, 2 H), 3.26 (m, 2 H), 2.97 (s, 3 H), 2.92 (s, 3 H), 2.72 (m, 2 H), 2.50 (m, 2 H), 2.10 (s, 3 H).
Example 34
Figure imgf000159_0001
Mixture of diastereoisomers (R)-l-[l-(l-Benzyl-3,3-difluoro-piperidin-4-yl)-lH-l,3,5,9-tetraaza- eye lopenta[a]naphthalen-2-yl] -ethanol
Racemic (l-Benzyl-3,3-difiuoro-piperidin-4-yl)-(3-nitro-[l,5]naphthyridin-4-yl)-amine A mixture of racemic l-benzyl-3,3-difluoro-piperidin-4-ylamine (prepared according to the procedures outline in WO2008121687) (357 mg, 1.58 mmol), DIPEA (1.25 mL, 7.20 mmol) and 4-chloro-3-nitro-[l,5]naphthyridine (302 mg, 1.44 mmol) in propan-2-ol (10 mL) was heated to 120 °C for 40 minutes using microwave irradiation. After cooling, the resulting precipitate was collected by filtration and washed with propan-2-ol and sucked dry to provide 412 mg (72%) of racemic (l-benzyl-3,3-difluoro-piperidin-4-yl)-(3-nitro-[l,5]naphthyridin-4-yl)-amine as a brown/ green solid. LCMS (Method B, ESI): RT = 3.73 min, m+H = 400.2; 1H NMR (400 MHz, DMSO-d6): δ 9.76 (br s, 1 H), 9.28 (s, 1 H), 8.95 (dd, 1 H), 8.36 (dd, 1 H), 7.91 (dd, 1 H), 7.34 (m, 5 H), 6.07 (br s, 1 H), 3.67 (s, 2 H), 3.19 (m, 1 H), 2.91 (br d, 1 H), 2.55 (m, 1 H), 2.36 (m, 2 H), 1.83 (m, 1 H).
Racemic N*4*-(l-Benzyl-3,3-difiuoro-piperidin-4-yl)-[l,5]naphthyridine-3,4-diamine
To a stirred suspension of racemic (l-benzyl-3,3-difluoro-piperidin-4-yl)-(3-nitro- [l,5]naphthyridin-4-yl)-amine (393 mg, 985 μιηοΐ) and iron powder (215 mg, 3.84 mmol) in ethanol (IMS grade, 5 mL), a solution of ammonium chloride (305 mg, 5.71 mmol) in water (1.5 mL) was added at room temperature. The mixture was then heated at 80 °C for 2 hours. After cooling, the mixture was filtered through of Celite® and the filter cake washed several times with ethanol (IMS grade) and water (4: 1). The filtrate was concentrated in vacuo and the resulting aqueous residue was treated with saturated sodium hydro gencarbonate solution and extracted with ethyl acetate (3x). The combined organic phases were washed (saturated sodium hydro gencarbonate solution and brine), dried (sodium sulfate) and concentrated in vacuo to afford 340 mg (93%) of racemic N*4*-(l-benzyl-3,3-difluoro-piperidin-4-yl)- [l,5]naphthyridine-3,4-diamine as a light brown solid. This material was used for the next step without further purification. LCMS (Method B, ESI): RT = 2.12 min, m+H = 370.2; 1H NMR (400 MHz, DMSO-d6): δ 8.72 (dd, 1 H), 8.44 (s, 1 H), 8.11 (dd, 1 H), 7.42 (dd, 1 H), 7.32 (m, 5 H), 5.88 (d, 1 H), 5.10 (s, 2 H), 4.51 (m, 1 H), 3.61 (q, 2 H), 3.03 (m, 1 H), 2.83 (br d, 1 H), 2.34 (m, 1 H), 2.20 (m, 1 H), 2.04 (m, 1 H), 1.68 (m, 1 H).
Mixture of diastereoisomers (R)-l-[l-(l-Benzyl-3,3-difluoro-piperidin-4-yl)-lH-l,3,5,9-tetraaza- cyclopenta[a]naphthalen-2-yl] -ethanol A stirred solution of (R)-(+)-lactamide (205 mg, 2.30 mmol) in anhydrous DCM (4 mL), was treated with triethyloxonium tetrafluoroborate (410 mg, 2.16 mmol) at room temperature for 2 hours. The volatiles were removed in vacuo and the residue dissolved in ethanol (absolute grade, 1 mL). This was added to a solution of racemic N*4!i:-(l-benzyl-3,3-difluoro-piperidin-4-yl)- [l,5]naphthyridine-3,4-diamine (266 mg, 0.72 mmol) in ethanol (absolute grade, 3 mL) and the reaction mixture was heated to 75°C, in a sealed vial, for 2 hours. The volatiles were removed in vacuo and the residue was purified by column chromatography using Isolute® SCX-2 cartridge (gradient: MeOH to 1M NH3 in MeOH) followed by column chromatography on silica gel (gradient: 0 to 10% [2M NH3 in MeOH] in DCM) to afford 198 mg (65%) of a mixture of diastereoisomers of (R)- 1 -[ 1 -( 1 -benzyl-3 ,3-difluoro-piperidin-4-yl)- 1 H- 1 ,3 ,5 ,9-tetraaza- cyclopenta[a]naphthalen-2-yl] -ethanol as a light orange solid. LCMS (Method A, ESI): RT = 3.57 min, m+H = 424.0 (Diastereoisomer I); RT = 3.83 min, m+H = 424.0 (Diastereoisomer II); Ratio 1 : 1; 1H NMR (400 MHz, DMSO-d6): (mixture of diastereoisomers and rotamers) δ 9.34- 9.30 (m, 1 H), 9.09 (dd, 0.25 H), 8.97 (m, 0.75 H), 8.58-8.53 (m, 1 H), 7.80-7.73 (m, 1 H), 7.54-7.36 (m, 5 H), 7.35-7.28 (m, 1 H), 5.96 (m, 0.25 H), 5.60 (m, 0.8 H), 5.57-5.45 (m, 0.2 H), 5.36-5.24 (m, 0.75 H), 5.19-5.11 (m, 0.25 H), 4.92-4.78 (m, 0.25 H), 3.96-3.84 (m, 0.5 H), 3.82-3.69 (m, 1.5 H), 3.31 (m, 1 H), 3.20-3.11 (m, 1 H), 2.92-2.73 (m, 1 H), 2.69-2.36 (m, 3 H), 1.72 (t, 2.3 H), 1.53 (d, 0.7 H).
Example 35
Figure imgf000161_0001
Mixture of diastereoisomers (R)-l-[l-(3,3-Difluoro-piperidin-4-yl)-lH-l,3,5,9-tetraaza- cyclopenta[a]naphthalen-2-yl] -ethanol formic acid salt To a stirred solution of a mixture of diastereoisomers of (R)-l-[l-(l-benzyl-3,3-difluoro- piperidin-4-yl)-lH-l,3,5,9-tetraaza-cyclopenta[a]naphthalen-2-yl]-ethanol (165 mg, 390 μιηοΐ) in MeOH (3 mL), ammonium formate (245 mg, 3.90 mmol) and palladium hydroxide (28 mg, 20 wt%) were added. The resulting mixture was heated to 60 °C for 1 hour. After cooling, the mixture was filtered through Celite® and the filtrate concentrated. The residue was concentrated on to silica gel and purified by flash chromatography (gradient: 0 to 20% MeOH in DCM). The resulting product was azeotroped (diethyl ether) to afford 95 mg of the crude product as an off- white solid. Purification by reverse phase HPLC (gradient: 5 to 50% MeCN in ¾0 + 0.1% HC02H) and subsequent lyophilisation afforded a mixture of diastereoisomers of (R)-l-[l-(3,3- difiuoro-piperidin-4-yl)-lH-l,3,5,9-tetraaza-cyclopenta[a]naphthalen-2-yl]-ethanol as a partial formic acid salt. LCMS (Method A, ESI): RT = 1.71 min, m+H = 334.0 (Diastereoisomer I); RT = 1.86 min, m+H = 334.0 (Diastereoisomer II); Ratio 1 : 1.7; 1H NMR (400 MHz, DMSO-d6): (mixture of diastereoisomers and rotamers) δ 9.35-9.28 (m, 1 H), 9.04 (dd, 0.15 H), 9.00-8.98 (m, 0.85 H), 8.58-8.54 (m, 0.85 H), 8.53-50 (m, 0.15 H), 8.18 (s, 0.4 H, formate), 7.80-7.72 (m, 1 H), 7.61-7.45 (m, 1 H), 6.10-5.85 (br s, 0.2 H), 5.66-5.45 (m, 0.8 H), 5.42-5.30 (m, 0.85 H), 5.21-5.13 (m, 0.15 H), 4.73-4.59 (m, 0.15 H), 3.50-3.15 (m, 4.5 H), 3.06-2.90 (dd, 0.85 H), 2.85-2.70 (m, 1.5 H), 2.40-2.30 (m, 0.85 H), 2.00-1.92 (m, 0.15 H), 1.76-1.67 (m, 2.4 H), 1.52 (d, 0.6 H).
Using procedures similar to those described herein the following compounds
Examples 36-56 were prepared.
TABLE 1
Figure imgf000162_0001
Figure imgf000163_0001
Figure imgf000164_0001
Figure imgf000165_0001
0
1 -( 1 -Methanesulfonyl-4-
56 m ethyl-pi perid i n-4-yl )-2- 33 2.80 / A 360.0 methyl-1 H-1 ,3,5,9-tetraaza- cyclopenta[a]naphthalene
The corresponding JAKl, JAK2, JAK3 and TYK2 inhibitions are shown in Table 2 for Examples 1-56.
Table 4
Example JAKl Ki, μΜ JAK2 Ki, μΜ JAK3 Ki, μΜ TYK2 Ki, μΜ
1 0.00021 0.001677 0.035879 0.00045
2 0.049253 0.346492 1.928896 0.066751
3 0.0131 15 0.039727 0.258109 0.017608
4 0.000026 0.00098 0.032335 0.000327
5 0.001 184 0.015383 0.249131 0.00266
6 0.019848 0.1 18633 1.459569 0.033861
7 0.001451 0.008023 0.216225 0.00156
8 0.090101 0.590916 2.936771 0.219831
9 0.006144 0.037516 1.240689 0.016127
10 0.183261 0.743909 3.163198 0.277747
11 0.291 1 1 2.193043 4.103771 1.552186
12 0.016482 0.064725 2.275882 0.023985
13 0.01545 0.058734 0.590122 0.041 175
14 3.782298 1.629453 4.103771 2.843124
15 0.554504 3.188008 3.167651 3.201208
16 0.057019 0.099813 0.091322 0.077887 17 0.076722 0.195324 0.555883 0.073655
18 0.022435 0.388687 1.680755 0.800049
19 0.797485 3.188008 3.330969 0.782784
20 0.281887 0.814472 1.987808 0.57372
21 0.157837 0.638439 0.870449 1.130425
22 0.12546 0.877731 1.596212 0.44102
23 0.008986 0.044382 0.412137 0.015552
24 0.00295 0.015628 0.247067 0.003289
25 5.621712 3.188008 4.103771 3.472582
26 2.541901 3.188008 3.818807 2.251313
27 0.21 1091 0.693582 1.609237 0.401 197
28 1.233426 1.437416 4.103771 3.472582
29 2.330678 3.188008 1.367924 3.472582
30 0.062102 0.134127 0.908426 0.18751
31 0.00074 0.004727 0.17679 0.002435
32 0.001 183 0.004759 0.084659 0.00309
33 0.128849 0.107103 0.257002 0.10508
34 0.014907 0.022002 0.232793 0.050148
35 0.180148 0.257001 1.648133 0.254747
36 0.00096 0.003286 0.013208 0.001463
37 0.051207 0.312484 2.27131 0.1 1 1679
38 0.020639 0.226953 2.691652 0.03581 1
39 0.174564 1.151221 4.103771 0.525042
40 0.622343 1.334921 3.375212 0.665646
41 0.026162 0.059677 1.123413 0.070278
42 0.008962 0.037038 0.1 16985 0.023126
43 0.002182 0.004491 0.01841 1 0.00198 44 0.005462 0.01271 1 0.07869 0.017251
45 0.003949 0.016464 0.191833 0.00868
46 0.135401 0.728806 1.021745 0.274803
47 0.364822 0.197944 1.998834 0.747173
48 0.68203 3.063791 4.103771 1.217607
49 0.588468 0.672592 2.630907 0.522185
50 0.043841 0.199531 0.405295 0.073614
51 0.06895 0.17071 1 0.436073 0.06081 1
52 0.065437 0.372106 0.34613 0.058302
53 0.070867 0.962636 1.605636 0.197231
54 0.141201 0.736512 1.493751 0.173314
55 0.479593 1.209702 4.103771 2.807143
56 0.077149 0.1 1 1 104 0.408752 0.166796
Although the invention has been described and illustrated with a certain degree of particularity, it is understood that the present disclosure has been made only by way of example, and that numerous changes in the combination and arrangement of parts can be resorted to by those skilled in the art without departing from the spirit and scope of the invention, as defined by the claims.

Claims

WHAT IS CLAIMED IS:
1. A compound of formula I:
Figure imgf000169_0001
stereoisomers, tautomers and pharmaceutically acceptable salts thereof, wherein
X is N or CR4; Y is N or CR5;
R1 is C3-12 cycloalkyl, or 3-20 membered heterocyclyl, wherein R1 is independently optionally substituted by halogen, oxo, -CN, -ORa, -SRa, -NRaRb, Ci_3 -C(0)ORa, -S(0)2Ra, or Ci_6 alkyl optionally substituted by oxo, -CN, -S(0)2Ra, C6-14 aryl, or halogen;
R2 is absent, Ci_6 alkyl, C2-6 alkenyl, C2-6 alkynyl, -(Ci_6 alkylene)-, -(C2_6 alkenylene)-, -(C2-6 alkynylene)-, -(C0_6 alkylene)CN, -(C0-3 alkylene)NRa(C0-3 alkylene)-, -(C0-3 alkylene)0(Co_3 alkylene)-, -(C0.3 alkylene)C(O)(C0-3 alkylene)-, -(C0.3 alkylene)NRaC(O)(C0-3 alkylene)-, -(C0.3 alkylene)C(O)NRa(C0-3 alkylene)-, -(C0.3 alkylene)C(O)O(C0-3 alkylene)-, -(Co-3 alkylene)OC(0)(Co_3 alkylene)-, -(C0.3 alkylene)NRaC(O)NRb(C0-3 alkylene)-, -(C0.3 alkylene)OC(0)NRa(Co_3 alkylene)-, -(C0.3 alkylene)NRaC(O)O(C0-3 alkylene)-, -(C0.3 alkylene)S(O)i_2(C0-3 alkylene)-, -(C0.3 alkylene)NRaS(O)i_2(C0-3 alkylene)-, -(C0.3 alkylene)S(O)i_2NRa(C0-3 alkylene)- or -(C0.3 alkylene)NRaS(O)i_2NRb(C0-3 alkylene)-, wherein said alkyl, alkyenyl, alkynyl, alkylene, alkenylene and alkynylene are independently optionally substituted by halogen, oxo, -CN, -ORc, -SRC, -NRcRd or Ci_3 alkyl optionally substituted by halogen;
R3 is absent, hydrogen, Ci_6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-7 cycloalkyl, C6-14 aryl or 3-20 membered heterocyclyl, wherein R3 is independently optionally substituted by R6;
R4 is hydrogen, halogen or Ci_3 alkyl; WO 2013/007768 " i ov " PCT/EP2012/063632
R5 is halogen, C1-12 alkyl, C2-12 alkenyl, C2-12 alkynyl, -(C0-3 alkylene)CN, -(C0-3 alkylene)NRaRb, -(C0.3 alkylene)ORa, -(C0.3 alkylene)SRa, -(C0.3 alkylene)C(0)Ra, -(C0.3 alkylene)NRaC(0)Rb, -(C0.3 alkylene)C(0)NRaRb, -(C0.3 alkylene)C(0)ORa, -(C0.3
alkylene)OC(0)Ra, -(C0.3 alkylene)NRaC(0)NRaRb, -(C0.3 alkylene)OC(0)NRaRb, -(C0.3 alkylene)NRaC(0)ORb, -(C0.3 alkylene)S(0)i_2Ra, -(C0.3 alkylene)NRaS(0)i_2Rb, -(C0.3 alkylene)S(0)i_2NRaRb, -(C0.3 alkylene)NRaS(0)i_2NRaRb, -(C0.3 alkylene)C3_i2 cycloalkyl, -(Co-3 alkylene)C6-i4 aryl, -(C0-3 alkylene)3-12 membered heterocyclyl or -(C0-3
alkylene)C(0)3-12 membered heterocyclyl, wherein said C2_i2 alkyl, alkenyl, alkynyl, alkylene, cycloalkyl, aryl and heterocyclyl are independently optionally substituted by halogen, oxo, -(C0- 3 alkylene)CN, -(C0.3 alkylene)ORc, -(C0.3 alkylene)NRcRd, -(C0.3 alkylene)C(0)Rc, -(C0.3 alkylene)C(0)ORc, -(C0.3 alkylene)C(0)NRcRd, -(C0.3 alkylene)NRcC(0)Rd, -(C0.3
alkylene)OC(0)NRcRd, -(C0.3 alkylene)NRcC(0)NRcRd, -(C0.3 alkylene)NRcC(0)ORd, -(C0.3 alkylene)S(0)o_2Rc, -(C0.3 alkylene)NRcS(0)i_2Rd, -(C0.3 alkylene) S(0)i_2NRcRd, -(C0.3 alkylene)NRcS(0)i_2NRcRd or Ci_6 alkyl optionally substituted by oxo, -CN or halogen; and wherein said Ci alkyl is independently optionally substituted by halogen, oxo, -(C0-3
alkylene)CN, -(Ci_3 alkylene)ORc, -(C0.3 alkylene)NRcRd, -(C0.3 alkylene)C(0)Rc, -(C0.3 alkylene)C(0)ORc, -(C0.3 alkylene)C(0)NRcRd, -(C0.3 alkylene)NRcC(0)Rd, -(C0.3
alkylene)OC(0)NRcRd, -(C0.3 alkylene)NRcC(0)NRcRd, -(C0.3 alkylene)NRcC(0)ORd, -(C0.3 alkylene)S(0)o_2Rc, -(C0.3 alkylene)NRcS(0)i_2Rd, -(C0.3 alkylene) S(0)i_2NRcRd, -(C0.3 alkylene)NRcS(0)i_2NRcRd, -ORk, or Ci_6 alkyl optionally substituted by oxo, -CN or halogen;
R6 is independently oxo, halogen, -CN, -C(0)Ra, -C(0)ORa, -NRaC(0)Rb, -C(0)NRaRb, -NRaC(0)NRaRb, -OC(0)NRaRb, -NRaC(0)ORb, -S(0)i_2Ra, -NRaS(0)2Ra, -S(0)2NRaRb, -ORa, -SRa, -NRaRb, Ci_6 alkyl, C3_6 cycloalkyl, C2_6 alkenyl, C2_6 alkynyl, 3-7 membered heterocycly or C6-14 aryl, and wherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl and aryl are independently optionally substituted by halogen, oxo, -CN, -ORc, -SRC, -NRcRd or Ci_6 alkyl optionally substituted by oxo or halogen; each Ra and Rb are independently hydrogen, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, -(C0-3 alkylene)C3-6 cycloalkyl, -(C0-3 alkylene)3-12 membered heterocyclyl, -(C0-3 alkylene)C(0)3-12 membered heterocyclyl or -(C0-3 alkylene)C6_i4 aryl, wherein said alkyl, cycloalkyl, heterocyclyl and aryl are independently optionally substituted by halogen, oxo, -CN, -ORe, -NReRf, -C(0)Rg, -C(0)ORg, -C(0)NRgRh, -NRgC(0)Rh, -OC(0)NRgRh, -NRgC(0)NRgRh, -NRgC(0)ORh, -S(0)i_2Rg, -NRgS(0)i_2Rh, -S(0)i_2NRgRh, -NRgS (O) 1 _2NRgRh, C3_6 cycloalkyl, 3-6 membered heterocyclyl, phenyl or Ci_3 alkyl optionally substituted by oxo or halogen, or taken together with the atom to which they are attached to form a 3-6 membered heterocyclyl optionally substituted by oxo, halogen, -C(0)Ci_6 alkyl or Ci_6 alkyl optionally substituted by oxo, halogen, ORg or NRgNRh; each Rc and Rd are independently hydrogen, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, -(Co_3 alkylene)C3_6 cycloalkyl, -(Co_3 alkylene)3-12 membered heterocyclyl, -(Co_3 alkylene)C(0)3-12 membered heterocyclyl or -(Co_3 alkylene)C6_i4 aryl, wherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl and aryl are independently optionally substituted by halogen, oxo, -CN, -ORg, -NRgRh, -C(0)Rg, -C(0)ORg, -C(0)NRgRh, -NRgC(0)Rh, -OC(0)NRgRh, -NRgC(0)NRgRh, -NRgC(0)ORh, -S(0)i_2Rg, -NRgS(0)i_2Rh, -S(0)i_2NRgRh, -NRgS(0)i_ 2NRgRh, C3_6 cycloalkyl, 3-6 membered heterocyclyl, phenyl or Ci_6 alkyl optionally substituted by oxo or halogen, or taken together with the atom to which they are attached to form a 3-6 membered heterocyclyl optionally substituted by oxo, halogen, -C(0)Ci_6 alkyl or Ci_6 alkyl optionally substituted by oxo or halogen; each Re, Rf, Rg, Rh are independently hydrogen or Ci_6 alkyl optionally substituted by halogen or oxo; and each Rk is independently hydrogen, C2_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, -(Co_3 alkylene)C3_6 cycloalkyl, -(Co_3 alkylene)3-12 membered heterocyclyl, -(Co_3 alkylene)C(0)3-12 membered heterocyclyl or -(Co_3 alkylene)C6_i4 aryl, wherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl and aryl are independently optionally substituted by halogen, oxo, -CN, -ORg, -NRgRh, -C(0)Rg, -C(0)ORg, -C(0)NRgRh, -NRgC(0)Rh, -OC(0)NRgRh,
-NRgC(0)NRgRh, -NRgC(0)ORh, -S(0)i_2Rg, -NRgS(0)i_2Rh, -S(0)i_2NRgRh, -NRgS(0)i_ 2NRgRh, C3_6 cycloalkyl, 3-6 membered heterocyclyl, phenyl or Ci_6 alkyl optionally substituted by oxo or halogen, or taken together with the atom to which they are attached to form a 3-6 membered heterocyclyl optionally substituted by oxo, halogen, -C(0)Ci_6 alkyl or Ci_6 alkyl optionally substituted by oxo or halogen.
2. The compound of claim 1, wherein X is CR4 and Y is CR5.
3. The compound of any one of claims 1-2, wherein R1 is C3_8 cycloalkyl, or 4-10 membered heterocyclyl, wherein R1 is independently optionally substituted by halogen, oxo, -CN, -ORa, -SRa, -NRaRb, -C(0)ORa, -S(0)2Ra, or Ci_6 alkyl optionally substituted by oxo, -CN, -S(0)2Ra, C6-i4 aryl, or halogen. WO 2013/007768 " 1 ' 1 " PCT/EP2012/063632
4. The compound of any one of claims 1-2, wherein R1 is C5_6 cycloalkyl or 5-6 membered heterocyclyl, wherein R1 is independently optionally substituted by halogen, oxo, -CN, -ORa, -SRa, -NRaRb, -C(0)ORa, -S(0)2Ra, or Ci_6 alkyl optionally substituted by oxo, -CN, -S(0)2Ra, C6-i4 aryl, or halogen. 5. The compound of any one of claims 1-2, wherein R1 is cyclohexyl, cyclopentyl, piperidinyl, or tetrahydropyranyl, wherein R1 is independently optionally substituted by halogen, oxo, -CN, -ORa, -SRa, -NRaRb, -C(0)ORa, -S(0)2Ra, or Ci_6 alkyl optionally substituted by oxo, -CN, -S(0)2Ra, C6-i4 aryl, or halogen.
6. The compound of any one of claims 1-2, wherein R1 is selected from:
Figure imgf000172_0001
7. The compound of any one of claims 1-2 wherein -R1-R2-R3 taken together are selected from:
Figure imgf000172_0002
Figure imgf000173_0001
3632
Figure imgf000174_0001
9. The compound of any one of claims 1-8 wherein R4 is hydrogen, methyl or F. 10. The compound of any one of claims 1-9, wherein R5 is halogen, C1-12 alkyl, -(C0-3 alkylene)CN, -(C0.3 alkylene)ORa, -(C0.3 alkylene)NRaRb, -(C0.3 alkylene)C(0)NRaRb, -(C0.3 alkylene)C3-i2 cycloalkyl, -(C0.3 alkylene)C(0)NRaRb, -(C0.3 alkylene)NRaC(0)Rb, -(C0.3 alkylene)NRaS(0)i_2Rb, -(C0.3 alkylene)NRaS(0)i_2NRaRb, -(C0.3 alkylene)S(0)i_2NRaRb, -(C0.3 alkylene)NRaC(0)ORb, -(C0.3 alkylene)S(0)i_2Ra, wherein R5 is independently optionally substituted by halogen, oxo, -(C0.3 alkylene)CN, -(C0.3 alkylene)ORc, -(C0.3 alkylene)NRcRd, -(Co-3 alkylene)C(0)Rc, -(C0.3 alkylene)C(0)ORc, -(C0.3 alkylene)C(0)NRcRd, -(C0.3 alkylene)NRcC(0)Rd, -(C0.3 alkylene)OC(0)NRcRd, -(C0.3 alkylene)NRcC(0)NRcRd, -(C0.3 alkylene)NRcC(0)ORd, -(C0.3 alkylene)S(O)0-2Rc, -(C0.3 alkylene)NRcS(0)i_2Rd, -(C0.3 alkylene)S(0)i_2NRcRd, -(C0.3 alkylene)NRcS(0)i_2NRcRd or Ci_6 alkyl optionally substituted by oxo, -CN or halogen.
11. The compound of any one of claims 1-10, wherein R5 is methyl, ethyl, propyl, isopropyl, cyclopropyl, eye lo butyl, cyano, 2-methylbutyl, N-(2-hydroxyethyl)amino, N-(2- methoxyethyl)amino , methylsulfonylamino methyl, 2-(methylsulfonylamino)ethyl,
cyclopropylmethyl, 2- [N-(2-propylsulfonyl)amino ] ethyl, 2- [N-(cyclopropylsulfonyl)- amino] ethyl, 2-(cyclopropylcarbonylamino)ethyl, 2-(acetylamino)ethyl, 2-(methoxymethyl- carbonylamino)ethyl, cyclopentoxymethyl, cyclopropylmethoxymethyl, 2,2,2- trifluoroethoxymethyl, cyclohexyl, methylamino, 2-(N,N-dimethylaminocarbonyl)ethyl, 2-(N- acetyl-N-methylamino)ethyl, 2-(ethoxycarbonylamino)ethyl, 1 -hydro xyethyl, N- WO 2013/007768 ~ L I^ ~ PCT/EP2012/063632 acylaminomethyl, 2-amino-l,l-difluoroethyl, Ν,Ν-dimethylamino, hydro xymethyl, methoxy, N- methylamino, N,N-dimethylamino,N-(2,2,2-trifluoroethyl)aminomethyl, (2- carboxycyclopropyl)(hydroxy)methyl, 2-hydro xyethyl, amino carbonylmethyl,
methylammocarbonylmethyl, ethylamino carbonylmethyl, 1 -hydro xypropyl, 1 ,2-dihydro xyethyl, N-(2-methylpropyl)aminocarbonylmethyl, cyclopentylaminocarbonylmethyl, 2-
(methoxycarbonylamino)ethyl, 2,2,2-trifluoro- 1 -hydro xyethyl, tert-butylamino carbonylmethyl, cyclobutylaminocarbonylmethyl, 2-hydroxyethoxy, isopropylamino carbonylmethyl, N-(N'N'- diemthylaminocarbonylmethyl)aminocarbonylmethyl, 4,4-difluorocyclohexyl- amino carbonylmethyl, 2,2-difluoroethylaminocarbonylmethyl, N-(2-hydroxyethyl)-N- methylammocarbonylmethyl, cyclopentylmethyl, N-cyclopentyl-N-methylaminocarbonylmethyl,
2- amino-l,l-difluoroethyl, 3-pyridyl, morpho lino methyl, morpholmocarbonylmethyl, 2-cyano-2- methylethyl, trifluoromethyl, 1 -hydroxy- 1-methylethyl, l-(N-isopropylaminocarbonyl)ethyl, 2- hydroxy-2-methylpropyl, N-(methylsulfonyl)-N-methylaminomethyl, difluoromethyl, 2-(2- butylsulfonylamino)ethyl, 2-(4-fluorophenylcarbonylamino)ethyl, 2-(cyclobutylcarbonyl- amino)ethyl, 2-(2-methylbutanoylamino)ethyl, 2-(benzoylamino)ethyl, 2,2-difluorocyclopropyl,
3- cyanobenzyl, 2-methylpropo xymethyl, 2-cyclopropylethyl, 3-pyridylmethyl,
methylsulfonylmethyl, ethoxycarbonylamino methyl, 3 -pyridylcarbonylamino methyl,
isopropylsulfonylaminomethyl, 2-pyridylcarbonylaminomethyl, cyclopropylsulfonyl- aminomethyl, cyclopentylsulfonylaminomethyl, 2-methylpropanoylaminomethyl,
cyclopropylcarbonylaminomethyl, 2-fluorobenzoylaminomethyl, 3-fluorobenzoylaminomethyl, 1-methylpropylsulfonylaminomethyl, 2-methylpropylsulfonylaminomethyl,
methoxyacetylaminomethyl, ethylsylfonylaminomethyl, 2-(3,3,3-trifluoropropylsulfonyl- amino)ethyl, 2-(2,2-difluorocyclopropylcarbonylamino)ethyl, fluoromethyl, 2- hydroxyethylamino, 2-methoxyethylamino, 1-aminoethyl, 2-(ethylsulfonylamino)ethyl, 2,2- dimethylpropo xymethyl, 1-metho xyethyl, tert-butylsulfonylaminomethyl, 2,2,2-trifluoroethyl- aminomethyl,
Figure imgf000175_0001
Figure imgf000175_0002
Figure imgf000176_0001
W
Figure imgf000177_0001
, wherein the wavy line represents the point of attachment in formula I.
12. The compound of any one of claims 1-11, wherein R6 is independently oxo, halogen, -CN, -C(0)Ra, -C(0)ORa, -NRaC(0)Rb, -C(0)NRaRb, -NRaC(0)NRaRb,
-OC(0)NRaRb, -NRaC(0)ORb, -S(0)i_2Ra, -NRaS(0)2Rb, -S(0)2NRaRb, -ORa, -SRa,
-NRaRb, Ci_6 alkyl, C3-6 cycloalkyl, C2_6 alkenyl, C2_6 alkynyl, 3-7 membered heterocycly or C6-14 aryl, and wherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl and aryl are
independently optionally substituted by halogen, oxo, -CN, -ORc, -SRC, -NRcRd or Ci_6 alkyl optionally substituted by oxo or halogen.
13. The compound of any one of claims 1-12, wherein each Ra and Rb are
independently hydrogen, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, -C3-6 cycloalkyl, -3-12 membered heterocyclyl, -C(0)3-12 membered heterocyclyl or -C6-14 aryl, wherein said alkyl, cycloalkyl, heterocyclyl and aryl are independently optionally substituted by halogen, oxo, -CN, -ORe, -NReRf, -C(0)Rg, -C(0)ORg, -C(0)NRgRh, -NRgC(0)Rh, -OC(0)NRgRh, -NRgC(0)NRgRh, -NRgC(0)ORh, -S(0)i_2Rg, -NRgS(0)i_2Rh, -S(0)i_2NRgRh, -NRgS (O) 1 _2NRgRh, C3_6 cycloalkyl, 3-6 membered heterocyclyl, phenyl or Ci_3 alkyl optionally substituted by oxo or halogen, or taken together with the atom to which they are attached to form a 3-6 membered heterocyclyl optionally substituted by oxo, halogen, -C(0)Ci_6 alkyl or Ci_6 alkyl optionally substituted by oxo, halogen, ORg or NRgNRh.
14. The compound of any one of claims 1-13, wherein each Rc and Rd are
independently hydrogen, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, -C3_6 cycloalkyl, -3-12 membered heterocyclyl, -C(0)3-12 membered heterocyclyl or -C6-14 aryl, wherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl and aryl are independently optionally substituted by halogen, oxo, -CN, -ORg, -NRgRh, -C(0)Rg, -C(0)ORg, -C(0)NRgRh, -NRgC(0)Rh, -OC(0)NRgRh, -NRgC(0)NRgRh, -NRgC(0)ORh, -S(0)i_2Rg, -NRgS(0)i_2Rh, -S(0)i_2NRgRh, -NRgS(0)i_ 2NRgRh, C3_6 cycloalkyl, 3-6 membered heterocyclyl, phenyl or Ci_6 alkyl optionally substituted by oxo or halogen, or taken together with the atom to which they are attached to form a 3-6 WO 2013/007768 " 1 " " PCT/EP2012/063632 membered heterocyclyl optionally substituted by oxo, halogen, -C(0)Ci_6 alkyl or Ci_6 alkyl optionally substituted by oxo or halogen.
15. The compound of any one of claims 1-14, wherein each Re, Rf, Rg, Rh are independently hydrogen, methyl, ethyl, propyl or isopropyl, optionally substituted by halogen oxo.
A compound of claim 1, selected from
Figure imgf000178_0001
Figure imgf000179_0001
Figure imgf000180_0001
Figure imgf000181_0001
and stereoisomers, tautomers and pharmaceutically acceptable salts thereof.
17. A pharmaceutical composition comprising a compound of any one of claims 1-16, a stereoisomer, tautomer or pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, adjuvant or vehicle.
18. A compound of any one of claims 1-16, a stereoisomer, tautomer, prodrug or pharmaceutically acceptable salt thereof, for use in therapy.
19. A compound of any one of claims 1-16, a stereoisomer, tautomer, prodrug or pharmaceutically acceptable salt thereof, for use in treating an immunological disease.
20. A compound of any one of claims 1-16, a stereoisomer, tautomer, prodrug or pharmaceutically acceptable salt thereof, for use in treating an immunological disease selected from rheumatoid arthritis, asthma, systemic lupus erythematosus, psoriasis, IBD and transplant rejection.
21. The use of a compound of any one of claims 1-16, a stereoisomer, tautomer, prodrug or pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of a disease responsive to the inhibition of JAKl kinase activity.
22. A method for preparing a compound of formula I or a pharmaceutically acceptable salt thereof as described in claim 1 comprising: a. for a compound of formula I wherein Y is CR5, cyclizing a corresponding compound of formula 100:
Figure imgf000182_0001
to provide the compound of formula I; b. contacting a compound of formula I with an acid or base to provide the corresponding pharmaceutically acceptable salt; c. removing a protecting group from a corresponding compound bearing a protecting group to provide the compound of formula I; d. for a compound of formula I wherein R2 and R3 are not each absent, converting a compound of formula 101:
Figure imgf000183_0001
to the corresponding compound of formula I wherein R2 and R3 are not each absent; for a compound of formula I wherein -R1-R2-R3 taken together are:
Figure imgf000183_0002
reacting a corresponding compound of formula I wherein -R1-R2-R3 taken together are:
Figure imgf000183_0003
with a compound of formula Ra-Lg, wherein Lg is a suitable leaving group, to provide the compound of formula I; for a compound of formula I wherein -R1-R2-R3 taken together are:
Figure imgf000183_0004
reacting a corresponding compound of formula I wherein -R1-R2-R3 taken together are: with a compound of formula Ra-Lg, wherein Lg is a suitable leaving group, to provide the compound of formula I;
g. for a compound of formula I wherein -R1 is:
Figure imgf000184_0001
removing a protecting group Pg from a corresponding compound of formula I wherein -R1 is
Figure imgf000184_0002
to provide the compound of formula I; or for a compound of formula I wherein -R1
Figure imgf000184_0003
removing a protecting group Pg from a corresponding compound of formula I wherein -R1 is
Figure imgf000184_0004
to provide the compound of formula I.
PCT/EP2012/063632 2011-07-13 2012-07-12 Tricyclic heterocyclic compounds, compositions and methods of use thereof as jak inhibitors WO2013007768A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201161507528P 2011-07-13 2011-07-13
US61/507,528 2011-07-13

Publications (1)

Publication Number Publication Date
WO2013007768A1 true WO2013007768A1 (en) 2013-01-17

Family

ID=46506425

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2012/063632 WO2013007768A1 (en) 2011-07-13 2012-07-12 Tricyclic heterocyclic compounds, compositions and methods of use thereof as jak inhibitors

Country Status (1)

Country Link
WO (1) WO2013007768A1 (en)

Cited By (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140121198A1 (en) * 2012-11-01 2014-05-01 Incyte Corporation Tricyclic fused thiophene derivatives as jak inhibitors
WO2014177915A1 (en) * 2013-05-01 2014-11-06 Piramal Enterprises Limited Cancer combination therapy using imidazo[4,5-c]quinoline derivatives
US9193733B2 (en) 2012-05-18 2015-11-24 Incyte Holdings Corporation Piperidinylcyclobutyl substituted pyrrolopyridine and pyrrolopyrimidine derivatives as JAK inhibitors
US9206187B2 (en) 2005-12-13 2015-12-08 Incyte Holdings Corporation Heteroaryl substituted pyrrolo[2,3-B] pyridines and pyrrolo[2,3-B] pyrimidines as Janus kinase
US9216984B2 (en) 2009-05-22 2015-12-22 Incyte Corporation 3-[4-(7H-pyrrolo[2,3-D]pyrimidin-4-yl)-1H-pyrazol-1-yl]octane—or heptane-nitrile as JAK inhibitors
US9221845B2 (en) 2013-03-06 2015-12-29 Incyte Holdings Corporation Processes and intermediates for making a JAK inhibitor
US9249145B2 (en) 2009-09-01 2016-02-02 Incyte Holdings Corporation Heterocyclic derivatives of pyrazol-4-yl-pyrrolo[2,3-d]pyrimidines as janus kinase inhibitors
US9334274B2 (en) 2009-05-22 2016-05-10 Incyte Holdings Corporation N-(hetero)aryl-pyrrolidine derivatives of pyrazol-4-yl-pyrrolo[2,3-d]pyrimidines and pyrrol-3-yl-pyrrolo[2,3-d]pyrimidines as janus kinase inhibitors
US9359358B2 (en) 2011-08-18 2016-06-07 Incyte Holdings Corporation Cyclohexyl azetidine derivatives as JAK inhibitors
US9464088B2 (en) 2010-03-10 2016-10-11 Incyte Holdings Corporation Piperidin-4-yl azetidine derivatives as JAK1 inhibitors
US9487521B2 (en) 2011-09-07 2016-11-08 Incyte Holdings Corporation Processes and intermediates for making a JAK inhibitor
US9498467B2 (en) 2014-05-30 2016-11-22 Incyte Corporation Treatment of chronic neutrophilic leukemia (CNL) and atypical chronic myeloid leukemia (aCML) by inhibitors of JAK1
WO2017046675A1 (en) * 2015-09-14 2017-03-23 Pfizer Inc. Novel imidazo [4,5-c] quinoline and imidazo [4,5-c][1,5] naphthyridine derivatives as lrrk2 inhibitors
US9611269B2 (en) 2011-06-20 2017-04-04 Incyte Corporation Azetidinyl phenyl, pyridyl or pyrazinyl carboxamide derivatives as JAK inhibitors
US9642855B2 (en) 2012-06-29 2017-05-09 Pfizer Inc. Substituted pyrrolo[2,3-d]pyrimidines as LRRK2 inhibitors
US9655854B2 (en) 2013-08-07 2017-05-23 Incyte Corporation Sustained release dosage forms for a JAK1 inhibitor
US9695171B2 (en) 2013-12-17 2017-07-04 Pfizer Inc. 3,4-disubstituted-1 H-pyrrolo[2,3-b]pyridines and 4,5-disubstituted-7H-pyrrolo[2,3-c]pyridazines as LRRK2 inhibitors
CN107011216A (en) * 2017-03-31 2017-08-04 泰州新威生物科技有限公司 A kind of preparation method of trans 4 Boc aminocyclohexane acetic acid
CN107089929A (en) * 2017-06-22 2017-08-25 广西科技大学 It is trans(4 cyanogen methyl)The preparation method of Cyclohexylamino t-butyl formate
US9802957B2 (en) 2014-04-30 2017-10-31 Incyte Corporation Processes of preparing a JAK1 inhibitor and new forms thereto
WO2018067422A1 (en) 2016-10-03 2018-04-12 Congxin Liang Novel jak1 selective inhibitors and uses thereof
WO2018130563A1 (en) 2017-01-11 2018-07-19 Leo Pharma A/S Novel amino-imidazopyridine derivatives as janus kinase inhibitors and pharmaceutical use thereof
US10166191B2 (en) 2012-11-15 2019-01-01 Incyte Corporation Sustained-release dosage forms of ruxolitinib
KR20190138793A (en) * 2017-03-10 2019-12-16 화이자 인코포레이티드 Novel imidazo [4,5-C] quinoline derivatives as LRRK2 inhibitors
WO2020007698A1 (en) 2018-07-06 2020-01-09 Leo Pharma A/S Novel amino-imidazopyrimidine derivatives as janus kinase inhibitors and pharmaceutical use thereof
US10640506B2 (en) 2010-11-19 2020-05-05 Incyte Holdings Corporation Cyclobutyl substituted pyrrolopyridine and pyrrolopyrimidines derivatives as JAK inhibitors
US11304949B2 (en) 2018-03-30 2022-04-19 Incyte Corporation Treatment of hidradenitis suppurativa using JAK inhibitors
US11833155B2 (en) 2020-06-03 2023-12-05 Incyte Corporation Combination therapy for treatment of myeloproliferative neoplasms

Citations (43)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4943533A (en) 1984-03-01 1990-07-24 The Regents Of The University Of California Hybrid cell lines that produce monoclonal antibodies to epidermal growth factor receptor
US5212290A (en) 1989-09-08 1993-05-18 The Johns Hopkins University Antibodies specific for type II mutant EGTR
WO1994015596A1 (en) 1992-12-30 1994-07-21 The Government Of The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Compositions and methods for inhibiting deoxyhypusine synthase and the growth of cells
WO1994017090A1 (en) 1993-01-20 1994-08-04 Glaxo Group Limited 2,6-diaminopurine derivatives
EP0659439A2 (en) 1993-12-24 1995-06-28 MERCK PATENT GmbH Immunoconjugates
US5457105A (en) 1992-01-20 1995-10-10 Zeneca Limited Quinazoline derivatives useful for treatment of neoplastic disease
US5475001A (en) 1993-07-19 1995-12-12 Zeneca Limited Quinazoline derivatives
WO1996003397A1 (en) 1994-07-21 1996-02-08 Akzo Nobel N.V. Cyclic ketone peroxide formulations
WO1996015111A1 (en) 1994-11-15 1996-05-23 Regents Of The University Of Minnesota Method and intermediates for the synthesis of korupensamines
WO1996030347A1 (en) 1995-03-30 1996-10-03 Pfizer Inc. Quinazoline derivatives
WO1996033980A1 (en) 1995-04-27 1996-10-31 Zeneca Limited Quinazoline derivatives
WO1996033978A1 (en) 1995-04-27 1996-10-31 Zeneca Limited Quinazoline derivative
WO1996040210A1 (en) 1995-06-07 1996-12-19 Imclone Systems Incorporated Antibody and antibody fragments for inhibiting the growth of tumors
US5654307A (en) 1994-01-25 1997-08-05 Warner-Lambert Company Bicyclic compounds capable of inhibiting tyrosine kinases of the epidermal growth factor receptor family
WO1997038983A1 (en) 1996-04-12 1997-10-23 Warner-Lambert Company Irreversible inhibitors of tyrosine kinases
WO1998014451A1 (en) 1996-10-02 1998-04-09 Novartis Ag Fused pyrazole derivative and process for its preparation
US5747498A (en) 1996-05-28 1998-05-05 Pfizer Inc. Alkynyl and azido-substituted 4-anilinoquinazolines
US5760041A (en) 1996-02-05 1998-06-02 American Cyanamid Company 4-aminoquinazoline EGFR Inhibitors
US5804396A (en) 1994-10-12 1998-09-08 Sugen, Inc. Assay for agents active in proliferative disorders
WO1998043960A1 (en) 1997-04-03 1998-10-08 American Cyanamid Company Substituted 3-cyano quinolines
WO1998050433A2 (en) 1997-05-05 1998-11-12 Abgenix, Inc. Human monoclonal antibodies to epidermal growth factor receptor
WO1998050038A1 (en) 1997-05-06 1998-11-12 American Cyanamid Company Use of quinazoline compounds for the treatment of polycystic kidney disease
US5866572A (en) 1996-02-14 1999-02-02 Zeneca Limited Quinazoline derivatives
WO1999006396A1 (en) 1997-07-29 1999-02-11 Warner-Lambert Company Irreversible bicyclic inhibitors of tyrosine kinases
WO1999006378A1 (en) 1997-07-29 1999-02-11 Warner-Lambert Company Irreversible inhibitors of tyrosine kinases
WO1999009016A1 (en) 1997-08-01 1999-02-25 American Cyanamid Company Substituted quinazoline derivatives and their use as tyrosine kinase inhibitors
US5891996A (en) 1972-09-17 1999-04-06 Centro De Inmunologia Molecular Humanized and chimeric monoclonal antibodies that recognize epidermal growth factor receptor (EGF-R); diagnostic and therapeutic use
WO1999024037A1 (en) 1997-11-06 1999-05-20 American Cyanamid Company Use of quinazoline derivatives as tyrosine kinase inhibitors for treating colonic polyps
US6002008A (en) 1997-04-03 1999-12-14 American Cyanamid Company Substituted 3-cyano quinolines
US6084095A (en) 1994-01-25 2000-07-04 Warner-Lambert Company Substituted pyrido[3,2-d]pyrimidines capable of inhibiting tyrosine kinases of the epidermal growth factor receptor family
US6140332A (en) 1995-07-06 2000-10-31 Novartis Ag Pyrrolopyrimidines and processes for the preparation thereof
US6344455B1 (en) 1998-11-19 2002-02-05 Warner-Lambert Company N-[4-(3-chloro-4-fluoro-phenylamino)-7-(3-morpholin-4-yl-propoxy)-quinazolin-6-yl]-acrylamide, and irreversible inhibitor of tyrosine kinases
US6391874B1 (en) 1996-07-13 2002-05-21 Smithkline Beecham Corporation Fused heterocyclic compounds as protein tyrosine kinase inhibitors
US6596726B1 (en) 1994-01-25 2003-07-22 Warner Lambert Company Tricyclic compounds capable of inhibiting tyrosine kinases of the epidermal growth factor receptor family
WO2003097641A2 (en) * 2002-05-21 2003-11-27 Novartis Ag 1h-imidazo[4,5-c] quinoline derivatives in the treatment of protein kinase dependent diseases
WO2006040625A1 (en) 2004-10-12 2006-04-20 Glenmark Pharmaceuticals S.A. Novel dipeptidyl peptidase iv inhibitors, pharmaceutical compositions containing them, and process for their preparation
WO2006062063A1 (en) 2004-12-08 2006-06-15 Astellas Pharma Inc. Piperidine derivative and process for producing the same
WO2006071862A2 (en) 2004-12-30 2006-07-06 Takeda Pharmaceutical Company Limited Polymorphs of 1-(2-methylpropyl)-1h-imidazo[4,5-c][1,5]naphthyridin-4-amine ethanesulfonate
WO2008065021A1 (en) 2006-11-30 2008-06-05 F. Hoffmann-La Roche Ag Diaminocyclohexane and diaminocyclopentane derivatives
WO2008121687A2 (en) 2007-03-28 2008-10-09 Array Biopharma Inc. Imidazo[1,2-a]pyridine compounds as receptor tyrosine kinase inhibitors
WO2009145719A1 (en) 2008-05-30 2009-12-03 Astrazeneca Ab Isoindoline derivatives comprising a cyano group and their use in the treatment of pain disorders
WO2009152133A1 (en) * 2008-06-10 2009-12-17 Abbott Laboratories Novel tricyclic compounds
WO2010038165A1 (en) * 2008-09-30 2010-04-08 Pfizer Inc. Imidazo[1,5]naphthyridine compounds, their pharmaceutical use and compositions

Patent Citations (54)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5891996A (en) 1972-09-17 1999-04-06 Centro De Inmunologia Molecular Humanized and chimeric monoclonal antibodies that recognize epidermal growth factor receptor (EGF-R); diagnostic and therapeutic use
US4943533A (en) 1984-03-01 1990-07-24 The Regents Of The University Of California Hybrid cell lines that produce monoclonal antibodies to epidermal growth factor receptor
US5212290A (en) 1989-09-08 1993-05-18 The Johns Hopkins University Antibodies specific for type II mutant EGTR
US5616582A (en) 1992-01-20 1997-04-01 Zeneca Limited Quinazoline derivatives as anti-proliferative agents
US5457105A (en) 1992-01-20 1995-10-10 Zeneca Limited Quinazoline derivatives useful for treatment of neoplastic disease
WO1994015596A1 (en) 1992-12-30 1994-07-21 The Government Of The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Compositions and methods for inhibiting deoxyhypusine synthase and the growth of cells
WO1994017090A1 (en) 1993-01-20 1994-08-04 Glaxo Group Limited 2,6-diaminopurine derivatives
US5475001A (en) 1993-07-19 1995-12-12 Zeneca Limited Quinazoline derivatives
EP0659439A2 (en) 1993-12-24 1995-06-28 MERCK PATENT GmbH Immunoconjugates
US6521620B1 (en) 1994-01-25 2003-02-18 Warner-Lambert Company Bicyclic compounds capable of inhibiting tyrosine kinases of the epidermal growth factor receptor family
US6455534B2 (en) 1994-01-25 2002-09-24 Warner-Lambert Company Bicyclic compounds capable of inhibiting tyrosine kinases of the epidermal growth factor receptor family
US6084095A (en) 1994-01-25 2000-07-04 Warner-Lambert Company Substituted pyrido[3,2-d]pyrimidines capable of inhibiting tyrosine kinases of the epidermal growth factor receptor family
US6265410B1 (en) 1994-01-25 2001-07-24 Warner-Lambert Company Bicyclic compounds capable of inhibiting tyrosine kinases of the epidermal growth factor receptor family
US6596726B1 (en) 1994-01-25 2003-07-22 Warner Lambert Company Tricyclic compounds capable of inhibiting tyrosine kinases of the epidermal growth factor receptor family
US5654307A (en) 1994-01-25 1997-08-05 Warner-Lambert Company Bicyclic compounds capable of inhibiting tyrosine kinases of the epidermal growth factor receptor family
US5679683A (en) 1994-01-25 1997-10-21 Warner-Lambert Company Tricyclic compounds capable of inhibiting tyrosine kinases of the epidermal growth factor receptor family
US6713484B2 (en) 1994-01-25 2004-03-30 Warner-Lambert Company Bicyclic compounds capable of inhibiting tyrosine kinases of the epidermal growth factor receptor family
WO1996003397A1 (en) 1994-07-21 1996-02-08 Akzo Nobel N.V. Cyclic ketone peroxide formulations
US5804396A (en) 1994-10-12 1998-09-08 Sugen, Inc. Assay for agents active in proliferative disorders
WO1996015111A1 (en) 1994-11-15 1996-05-23 Regents Of The University Of Minnesota Method and intermediates for the synthesis of korupensamines
WO1996030347A1 (en) 1995-03-30 1996-10-03 Pfizer Inc. Quinazoline derivatives
US5770599A (en) 1995-04-27 1998-06-23 Zeneca Limited Quinazoline derivatives
WO1996033980A1 (en) 1995-04-27 1996-10-31 Zeneca Limited Quinazoline derivatives
WO1996033978A1 (en) 1995-04-27 1996-10-31 Zeneca Limited Quinazoline derivative
WO1996040210A1 (en) 1995-06-07 1996-12-19 Imclone Systems Incorporated Antibody and antibody fragments for inhibiting the growth of tumors
US6140332A (en) 1995-07-06 2000-10-31 Novartis Ag Pyrrolopyrimidines and processes for the preparation thereof
US5760041A (en) 1996-02-05 1998-06-02 American Cyanamid Company 4-aminoquinazoline EGFR Inhibitors
US6399602B1 (en) 1996-02-14 2002-06-04 Zeneca Limited Quinazoline derivatives
US5866572A (en) 1996-02-14 1999-02-02 Zeneca Limited Quinazoline derivatives
WO1997038983A1 (en) 1996-04-12 1997-10-23 Warner-Lambert Company Irreversible inhibitors of tyrosine kinases
US6602863B1 (en) 1996-04-12 2003-08-05 Warner-Lambert Company Irreversible inhibitors of tyrosine kinases
US6344459B1 (en) 1996-04-12 2002-02-05 Warner-Lambert Company Irreversible inhibitors of tyrosine kinases
US5747498A (en) 1996-05-28 1998-05-05 Pfizer Inc. Alkynyl and azido-substituted 4-anilinoquinazolines
US6391874B1 (en) 1996-07-13 2002-05-21 Smithkline Beecham Corporation Fused heterocyclic compounds as protein tyrosine kinase inhibitors
WO1998014451A1 (en) 1996-10-02 1998-04-09 Novartis Ag Fused pyrazole derivative and process for its preparation
US6002008A (en) 1997-04-03 1999-12-14 American Cyanamid Company Substituted 3-cyano quinolines
WO1998043960A1 (en) 1997-04-03 1998-10-08 American Cyanamid Company Substituted 3-cyano quinolines
US6235883B1 (en) 1997-05-05 2001-05-22 Abgenix, Inc. Human monoclonal antibodies to epidermal growth factor receptor
WO1998050433A2 (en) 1997-05-05 1998-11-12 Abgenix, Inc. Human monoclonal antibodies to epidermal growth factor receptor
WO1998050038A1 (en) 1997-05-06 1998-11-12 American Cyanamid Company Use of quinazoline compounds for the treatment of polycystic kidney disease
WO1999006396A1 (en) 1997-07-29 1999-02-11 Warner-Lambert Company Irreversible bicyclic inhibitors of tyrosine kinases
WO1999006378A1 (en) 1997-07-29 1999-02-11 Warner-Lambert Company Irreversible inhibitors of tyrosine kinases
WO1999009016A1 (en) 1997-08-01 1999-02-25 American Cyanamid Company Substituted quinazoline derivatives and their use as tyrosine kinase inhibitors
WO1999024037A1 (en) 1997-11-06 1999-05-20 American Cyanamid Company Use of quinazoline derivatives as tyrosine kinase inhibitors for treating colonic polyps
US6344455B1 (en) 1998-11-19 2002-02-05 Warner-Lambert Company N-[4-(3-chloro-4-fluoro-phenylamino)-7-(3-morpholin-4-yl-propoxy)-quinazolin-6-yl]-acrylamide, and irreversible inhibitor of tyrosine kinases
WO2003097641A2 (en) * 2002-05-21 2003-11-27 Novartis Ag 1h-imidazo[4,5-c] quinoline derivatives in the treatment of protein kinase dependent diseases
WO2006040625A1 (en) 2004-10-12 2006-04-20 Glenmark Pharmaceuticals S.A. Novel dipeptidyl peptidase iv inhibitors, pharmaceutical compositions containing them, and process for their preparation
WO2006062063A1 (en) 2004-12-08 2006-06-15 Astellas Pharma Inc. Piperidine derivative and process for producing the same
WO2006071862A2 (en) 2004-12-30 2006-07-06 Takeda Pharmaceutical Company Limited Polymorphs of 1-(2-methylpropyl)-1h-imidazo[4,5-c][1,5]naphthyridin-4-amine ethanesulfonate
WO2008065021A1 (en) 2006-11-30 2008-06-05 F. Hoffmann-La Roche Ag Diaminocyclohexane and diaminocyclopentane derivatives
WO2008121687A2 (en) 2007-03-28 2008-10-09 Array Biopharma Inc. Imidazo[1,2-a]pyridine compounds as receptor tyrosine kinase inhibitors
WO2009145719A1 (en) 2008-05-30 2009-12-03 Astrazeneca Ab Isoindoline derivatives comprising a cyano group and their use in the treatment of pain disorders
WO2009152133A1 (en) * 2008-06-10 2009-12-17 Abbott Laboratories Novel tricyclic compounds
WO2010038165A1 (en) * 2008-09-30 2010-04-08 Pfizer Inc. Imidazo[1,5]naphthyridine compounds, their pharmaceutical use and compositions

Non-Patent Citations (45)

* Cited by examiner, † Cited by third party
Title
"Beilsteins Handbuch der organischen Chemie", SPRINGER-VERLAG
"Chiral Liquid Chromatography W.", 1989, CHAPMAN AND HALL
"Comprehensive Heterocyclic Chemistry", 1984, PERGAMON PRESS
"Drug Stereochemistry, Analytical Methods and Pharmacology", 1993, MARCEL DEKKER, INC.
"Lang's Handbook ø/ Chemistrv", 1985
"McGraw-Hill Dictionary of Chemical Terms", 1984, MCGRAW-HILL BOOK COMPANY
ANSEL, HOWARD C. ET AL.: "Ansel's Pharmaceutical Dosage Forms and Drug Delivery Systems", 2004, LIPPINCOTT, WILLIAMS & WILKINS
BASLUND ET AL., ARTHRITIS & RHEUMATISM, vol. 52, 2005, pages 2686 - 2692
BERNARDELLI ET AL., TETRAHEDRON: ASYMMETRY, vol. 15, 2004, pages 1451 - 1455
CHANGELIAN ET AL., SCIENCE, vol. 302, 2003, pages 875 - 878
E. HASLAM: "Protective Groups in Organic Chemistry", 1973, PLENUM PRESS
E. HASLAM; J. G. W. MCOMIE: "Protective Groups in Organic Chemistry", 1973, PLENUM PRESS
ELIEL, E.; WILEN, S.: "Stereochemistry of Organic Compounds", 1994, JOHN WILEY & SONS, INC.
ELIEL, E.; WILEN, S.: "Stereochemistry of Organic Compounds", 1994, JOHN WILEY & SONS, INC., pages: 322
GENNARO, ALFONSO R. ET AL.: "Remington: The Science and Practice of Pharmacy", 2000, LIPPINCOTT, WILLIAMS & WILKINS
HELLMAN ET AL.: "Principles I and Practice of Oncology", vol. 1, 1993, article "Principles of Radiation Therapy, Cancer", pages: 24875
J. ORG. CHEM., vol. 69, 2004, pages 4538
J. ORG. CHEM., vol. 69, no. 13, 2004, pages 4538
JACOB, J. ORG. CHEM., vol. 47, 1982, pages 4165
JOHNS ET AL., J. BIOL. CHEM., vol. 279, no. 29, 2004, pages 30375 - 30384
KISSELEVA ET AL., GENE, vol. 285, 2002, pages 1 - 24
KRUEGER ET AL., N. ENGL. J. MED., vol. 356, 2007, pages 580 - 92
LEVY ET AL., NAT. REV. MOL. CELL BIOL., vol. 3, 2005, pages 651 - 662
LOCHMULLER, C. H., J. CHROMATOGR., vol. 113, no. 3, 1975, pages 283 - 302
LOUIS F. FIESER; MARY FIESER: "Reagents for Organic Synthesis", vol. 1-19, 1967, WILEY
MANNON ET AL., N. ENGL. J. MED., vol. 351, 2004, pages 2069 - 79
NICOLAOU ET AL., ANGEW. CHEM INTL. ED. ENGL., vol. 33, 1994, pages 183 - 186
OKAMOTO, J. OF CHROMATOGR., vol. 513, 1990, pages 375 - 378
ORG LETT, vol. 2, 2000, pages 4169
ORG. LETT., vol. 2, 2000, pages 4169
O'SHEA ET AL., CELL, vol. 109, 2002, pages S121 - S131
P. BERNARDELLI ET AL., TETRAHEDRON: ASYMMETRY, vol. 15, 2004, pages 1451 - 1455
REICH ET AL., NAT. REV. DRUG DISCOV., vol. 8, 2009, pages 355 - 356
ROWE, RAYMOND C.: "Handbook of Pharmaceutical Excipients. Chicago", 2005, PHARMACEUTICAL PRESS
S. FIXON-OWOO ET AL., PHYTOCHEMISTRY, vol. 63, 2003, pages 315 - 334
SCHEINECKER ET AL., NAT. REV. DRUG DISCOV., vol. 8, 2009, pages 273 - 274
SCHINDLER ET AL., J BIOL. CHEM., vol. 282, 2007, pages 20059 - 63
STRAGLIOTTO ET AL., EUR. J. CANCER, vol. 32A, 1996, pages 636 - 640
T. W. GREENE: "Protective Groups in Organic Synthesis", 1991, JOHN WILEY & SONS
T. W. GREENE; P. G. M. WUTS: "Protective Groups in Organic Synthesis", 1991, JOHN WILEY & SONS, INC.
T.W. GREENE: "Protective Groups in Organic Synthesis", 1981, JOHN WILEY AND SONS
TETRAHEDRON, vol. 53, 1997, pages 3347
TETRAHEDRON, vol. 53, no. 9, 1997, pages 3347
WATFORD, W.T.; O'SHEA, J.J., IMMUNITY, vol. 25, 2006, pages 695 - 697
WILKS A.F., PROC. NATL. ACAD. SCI. U.S.A., vol. 86, 1989, pages 1603 - 1607

Cited By (67)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9814722B2 (en) 2005-12-13 2017-11-14 Incyte Holdings Corporation Heteroaryl substituted pyrrolo[2,3-B] pyridines and pyrrolo[2,3-B] pyrimidines as janus kinase inhibitors
US11744832B2 (en) 2005-12-13 2023-09-05 Incyte Corporation Heteroaryl substituted pyrrolo[2,3-b]pyridines and pyrrolo[2,3-b]pyrimidines as Janus kinase inhibitors
US9662335B2 (en) 2005-12-13 2017-05-30 Incyte Holdings Corporation Heteroaryl substituted pyrrolo[2,3-B] pyridines and pyrrolo[2,3-B] pyrimidines as janus kinase inhibitors
US10398699B2 (en) 2005-12-13 2019-09-03 Incyte Holdings Corporation Heteroaryl substituted pyrrolo[2,3-b]pyridines and pyrrolo[2,3-b]pyrimidines as janus kinase inhibitors
US10639310B2 (en) 2005-12-13 2020-05-05 Incyte Corporation Heteroaryl substituted pyrrolo[2,3-b]pyridines and pyrrolo[2,3-b]pyrimidines as Janus kinase inhibitors
US11331320B2 (en) 2005-12-13 2022-05-17 Incyte Holdings Corporation Heteroaryl substituted pyrrolo[2,3-b]pyridines and pyrrolo[2,3-b]pyrimidines as Janus kinase inhibitors
US9206187B2 (en) 2005-12-13 2015-12-08 Incyte Holdings Corporation Heteroaryl substituted pyrrolo[2,3-B] pyridines and pyrrolo[2,3-B] pyrimidines as Janus kinase
US9334274B2 (en) 2009-05-22 2016-05-10 Incyte Holdings Corporation N-(hetero)aryl-pyrrolidine derivatives of pyrazol-4-yl-pyrrolo[2,3-d]pyrimidines and pyrrol-3-yl-pyrrolo[2,3-d]pyrimidines as janus kinase inhibitors
US9216984B2 (en) 2009-05-22 2015-12-22 Incyte Corporation 3-[4-(7H-pyrrolo[2,3-D]pyrimidin-4-yl)-1H-pyrazol-1-yl]octane—or heptane-nitrile as JAK inhibitors
US9623029B2 (en) 2009-05-22 2017-04-18 Incyte Holdings Corporation 3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]octane- or heptane-nitrile as JAK inhibitors
US9249145B2 (en) 2009-09-01 2016-02-02 Incyte Holdings Corporation Heterocyclic derivatives of pyrazol-4-yl-pyrrolo[2,3-d]pyrimidines as janus kinase inhibitors
US11285140B2 (en) 2010-03-10 2022-03-29 Incyte Corporation Piperidin-4-yl azetidine derivatives as JAK1 inhibitors
US9464088B2 (en) 2010-03-10 2016-10-11 Incyte Holdings Corporation Piperidin-4-yl azetidine derivatives as JAK1 inhibitors
US10640506B2 (en) 2010-11-19 2020-05-05 Incyte Holdings Corporation Cyclobutyl substituted pyrrolopyridine and pyrrolopyrimidines derivatives as JAK inhibitors
US11214573B2 (en) 2011-06-20 2022-01-04 Incyte Holdings Corporation Azetidinyl phenyl, pyridyl or pyrazinyl carboxamide derivatives as JAK inhibitors
US9611269B2 (en) 2011-06-20 2017-04-04 Incyte Corporation Azetidinyl phenyl, pyridyl or pyrazinyl carboxamide derivatives as JAK inhibitors
US9359358B2 (en) 2011-08-18 2016-06-07 Incyte Holdings Corporation Cyclohexyl azetidine derivatives as JAK inhibitors
US9718834B2 (en) 2011-09-07 2017-08-01 Incyte Corporation Processes and intermediates for making a JAK inhibitor
US9487521B2 (en) 2011-09-07 2016-11-08 Incyte Holdings Corporation Processes and intermediates for making a JAK inhibitor
US9193733B2 (en) 2012-05-18 2015-11-24 Incyte Holdings Corporation Piperidinylcyclobutyl substituted pyrrolopyridine and pyrrolopyrimidine derivatives as JAK inhibitors
US9642855B2 (en) 2012-06-29 2017-05-09 Pfizer Inc. Substituted pyrrolo[2,3-d]pyrimidines as LRRK2 inhibitors
US10370387B2 (en) 2012-11-01 2019-08-06 Incyte Holdings Corporation Tricyclic fused thiophene derivatives as JAK inhibitors
US20160015695A1 (en) * 2012-11-01 2016-01-21 Incyte Corporation Tricyclic fused thiophene derivatives as jak inhibitors
US11851442B2 (en) 2012-11-01 2023-12-26 Incyte Corporation Tricyclic fused thiophene derivatives as JAK inhibitors
US11161855B2 (en) 2012-11-01 2021-11-02 Incyte Corporation Tricyclic fused thiophene derivatives as JAK inhibitors
US9777017B2 (en) * 2012-11-01 2017-10-03 Incyte Holdings Corporation Tricyclic fused thiophene derivatives as JAK inhibitors
US9181271B2 (en) * 2012-11-01 2015-11-10 Incyte Holdings Corporation Tricyclic fused thiophene derivatives as JAK inhibitors
US20160024109A1 (en) * 2012-11-01 2016-01-28 Incyte Corporation Tricyclic fused thiophene derivatives as jak inhibitors
US9908895B2 (en) * 2012-11-01 2018-03-06 Incyte Corporation Tricyclic fused thiophene derivatives as JAK inhibitors
US20140121198A1 (en) * 2012-11-01 2014-05-01 Incyte Corporation Tricyclic fused thiophene derivatives as jak inhibitors
US11896717B2 (en) 2012-11-15 2024-02-13 Incyte Holdings Corporation Sustained-release dosage forms of ruxolitinib
US11576865B2 (en) 2012-11-15 2023-02-14 Incyte Corporation Sustained-release dosage forms of ruxolitinib
US10166191B2 (en) 2012-11-15 2019-01-01 Incyte Corporation Sustained-release dosage forms of ruxolitinib
US11337927B2 (en) 2012-11-15 2022-05-24 Incyte Holdings Corporation Sustained-release dosage forms of ruxolitinib
US11576864B2 (en) 2012-11-15 2023-02-14 Incyte Corporation Sustained-release dosage forms of ruxolitinib
US10874616B2 (en) 2012-11-15 2020-12-29 Incyte Corporation Sustained-release dosage forms of ruxolitinib
US9221845B2 (en) 2013-03-06 2015-12-29 Incyte Holdings Corporation Processes and intermediates for making a JAK inhibitor
WO2014177915A1 (en) * 2013-05-01 2014-11-06 Piramal Enterprises Limited Cancer combination therapy using imidazo[4,5-c]quinoline derivatives
US11045421B2 (en) 2013-08-07 2021-06-29 Incyte Corporation Sustained release dosage forms for a JAK1 inhibitor
US10561616B2 (en) 2013-08-07 2020-02-18 Incyte Corporation Sustained release dosage forms for a JAK1 inhibitor
US9655854B2 (en) 2013-08-07 2017-05-23 Incyte Corporation Sustained release dosage forms for a JAK1 inhibitor
US9695171B2 (en) 2013-12-17 2017-07-04 Pfizer Inc. 3,4-disubstituted-1 H-pyrrolo[2,3-b]pyridines and 4,5-disubstituted-7H-pyrrolo[2,3-c]pyridazines as LRRK2 inhibitors
US10450325B2 (en) 2014-04-30 2019-10-22 Incyte Corporation Processes of preparing a JAK1 inhibitor and new forms thereto
US9802957B2 (en) 2014-04-30 2017-10-31 Incyte Corporation Processes of preparing a JAK1 inhibitor and new forms thereto
US9498467B2 (en) 2014-05-30 2016-11-22 Incyte Corporation Treatment of chronic neutrophilic leukemia (CNL) and atypical chronic myeloid leukemia (aCML) by inhibitors of JAK1
US10039753B2 (en) 2015-09-14 2018-08-07 Pfizer Inc. Imidazo[4,5-c]quinoline and imidazo[4,5-c][1,5]naphthyridine derivatives as LRRK2 inhibitors
CN108137586A (en) * 2015-09-14 2018-06-08 辉瑞大药厂 As novel imidazo [4,5-c] quinoline of LRRK2 inhibitor and imidazo [4,5-c] [1,5] 7-naphthyridine derivatives
KR20180050407A (en) * 2015-09-14 2018-05-14 화이자 인코포레이티드 Imidazo [4,5-c] quinoline and imidazo [4,5-c] [1,5] naphthyridine derivatives as LRRK2 inhibitors
KR102161364B1 (en) * 2015-09-14 2020-09-29 화이자 인코포레이티드 Imidazo[4,5-c]quinoline and imidazo[4,5-c][1,5]naphthyridine derivatives as LRRK2 inhibitors
RU2722149C1 (en) * 2015-09-14 2020-05-27 Пфайзер Инк. New derivatives of imidazo [4,5-c] quinolines and imidazo [4,5-c] [1,5] naphthyridines as lrrk2 inhibitors
AU2016322813B2 (en) * 2015-09-14 2021-04-01 Pfizer Inc. Novel imidazo (4,5-c) quinoline and imidazo (4,5-c)(1,5) naphthyridine derivatives as LRRK2 inhibitors
WO2017046675A1 (en) * 2015-09-14 2017-03-23 Pfizer Inc. Novel imidazo [4,5-c] quinoline and imidazo [4,5-c][1,5] naphthyridine derivatives as lrrk2 inhibitors
JP2018526422A (en) * 2015-09-14 2018-09-13 ファイザー・インク Novel imidazo [4,5-c] quinoline and imidazo [4,5-c] [1,5] naphthyridine derivatives as LRRK2 inhibitors
WO2018067422A1 (en) 2016-10-03 2018-04-12 Congxin Liang Novel jak1 selective inhibitors and uses thereof
KR20190103231A (en) * 2017-01-11 2019-09-04 레오 파마 에이/에스 Novel amino-imidazopyridine derivatives as Janus kinase inhibitors and pharmaceutical uses thereof
JP7009504B2 (en) 2017-01-11 2022-01-25 レオ ファーマ アクティーゼルスカブ Novel aminoimidazolipyridine derivatives as Janus kinase inhibitors and their pharmaceutical use
JP2020504186A (en) * 2017-01-11 2020-02-06 レオ ファーマ アクティーゼルスカブ Novel aminoimidazopyridine derivatives as Janus kinase inhibitors and their use as medicaments
WO2018130563A1 (en) 2017-01-11 2018-07-19 Leo Pharma A/S Novel amino-imidazopyridine derivatives as janus kinase inhibitors and pharmaceutical use thereof
US10703751B2 (en) 2017-01-11 2020-07-07 Leo Pharma A/S Amino-imidazopyridine derivatives as Janus kinase inhibitors and pharmaceutical use thereof
KR102548543B1 (en) 2017-01-11 2023-06-29 아킬리온 에이비 Novel amino-imidazopyridine derivatives as Janus kinase inhibitors and their pharmaceutical uses
KR20190138793A (en) * 2017-03-10 2019-12-16 화이자 인코포레이티드 Novel imidazo [4,5-C] quinoline derivatives as LRRK2 inhibitors
KR102582626B1 (en) 2017-03-10 2023-09-22 화이자 인코포레이티드 Novel imidazo[4,5-C]quinoline derivatives as LRRK2 inhibitors
CN107011216A (en) * 2017-03-31 2017-08-04 泰州新威生物科技有限公司 A kind of preparation method of trans 4 Boc aminocyclohexane acetic acid
CN107089929A (en) * 2017-06-22 2017-08-25 广西科技大学 It is trans(4 cyanogen methyl)The preparation method of Cyclohexylamino t-butyl formate
US11304949B2 (en) 2018-03-30 2022-04-19 Incyte Corporation Treatment of hidradenitis suppurativa using JAK inhibitors
WO2020007698A1 (en) 2018-07-06 2020-01-09 Leo Pharma A/S Novel amino-imidazopyrimidine derivatives as janus kinase inhibitors and pharmaceutical use thereof
US11833155B2 (en) 2020-06-03 2023-12-05 Incyte Corporation Combination therapy for treatment of myeloproliferative neoplasms

Similar Documents

Publication Publication Date Title
WO2013007768A1 (en) Tricyclic heterocyclic compounds, compositions and methods of use thereof as jak inhibitors
US8461328B2 (en) Tricyclic heterocyclic compounds, compositions and methods of use thereof
EP2742040B1 (en) Indazole compounds, compositions and methods of use
WO2013007765A1 (en) Fused tricyclic compounds for use as inhibitors of janus kinases
DK2989106T3 (en) CONDENSED HETEROCYCLIC COMPOUNDS AS PROTEINKINASE INHIBITORS
WO2012085176A1 (en) Tricyclic pyrazinone compounds, compositions and methods of use thereof as janus kinase inhibitors
US20220127265A1 (en) Therapeutic compounds and uses thereof
WO2012066061A1 (en) Pyrazolopyridines and pyrazolopyridines and their use as tyk2 inhibitors
MX2012010265A (en) Imidazopyridine and purine compounds, compositions and methods of use.
EP2616072A1 (en) Azabenzothiazole compounds, compositions and methods of use
US20140206702A1 (en) Imidazopyridine compounds, compositions and methods of use
JP2022539259A (en) Heterocyclic compounds as kinase inhibitors
US20160326142A1 (en) Pyrazole carboxamide compounds, compositions and methods of use
WO2016001341A1 (en) Sulfonylaminopyridine compounds, compositions and methods of use
WO2015049325A1 (en) Therapeutic inhibitors of cdk8 and uses thereof
WO2016091916A1 (en) Pyrazolylaminopurines as itk inhibitors
TW202300150A (en) Cyclic compounds and methods of using same

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: 12733745

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 12733745

Country of ref document: EP

Kind code of ref document: A1