WO2013007768A1 - Composés hétérocycliques tricycliques, compositions et procédés d'utilisation de ces composés comme inhibiteurs des jak - Google Patents

Composés hétérocycliques tricycliques, compositions et procédés d'utilisation de ces composés comme inhibiteurs des jak 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
English (en)
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/fr

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

L'invention concerne de nouveaux composés de la formule I ayant la formule générale : (I) dans laquelle Rl s R2, R3, X et Y sont tels que décrits dans le descriptif. En conséquence, lesdits composés peuvent être présentés dans des compositions pharmaceutiquement acceptables et utilisés pour le traitement de troubles immunologiques ou hyperprolifératifs.
PCT/EP2012/063632 2011-07-13 2012-07-12 Composés hétérocycliques tricycliques, compositions et procédés d'utilisation de ces composés comme inhibiteurs des jak WO2013007768A1 (fr)

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 (fr) 2013-01-17

Family

ID=46506425

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2012/063632 WO2013007768A1 (fr) 2011-07-13 2012-07-12 Composés hétérocycliques tricycliques, compositions et procédés d'utilisation de ces composés comme inhibiteurs des jak

Country Status (1)

Country Link
WO (1) WO2013007768A1 (fr)

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 (fr) * 2013-05-01 2014-11-06 Piramal Enterprises Limited Multi-thérapie anti-cancéreuse utilisant des dérivés de imidazo[4,5-c]quinoline
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 (fr) * 2015-09-14 2017-03-23 Pfizer Inc. Nouveaux dérivés imidazo [4,5-c] quinoline et imidazo [4,5-c] [1,5] naphthyridine utilisés comme inhibiteurs de lrrk2
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 (zh) * 2017-03-31 2017-08-04 泰州新威生物科技有限公司 一种反式‑4‑Boc‑氨基环己烷乙酸的制备方法
CN107089929A (zh) * 2017-06-22 2017-08-25 广西科技大学 反式‑(4‑氰甲基)环己基氨基甲酸叔丁酯的制备方法
US9802957B2 (en) 2014-04-30 2017-10-31 Incyte Corporation Processes of preparing a JAK1 inhibitor and new forms thereto
WO2018067422A1 (fr) 2016-10-03 2018-04-12 Congxin Liang Nouveaux inhibiteurs sélectifs de jak1 et leurs utilisations
WO2018130563A1 (fr) 2017-01-11 2018-07-19 Leo Pharma A/S Nouveaux dérivés d'amino-imidazopyridine en tant qu'inhibiteurs de janus kinase et leur utilisation pharmaceutique
US10166191B2 (en) 2012-11-15 2019-01-01 Incyte Corporation Sustained-release dosage forms of ruxolitinib
KR20190138793A (ko) * 2017-03-10 2019-12-16 화이자 인코포레이티드 Lrrk2 억제제로서의 신규 이미다조[4,5-c]퀴놀린 유도체
WO2020007698A1 (fr) 2018-07-06 2020-01-09 Leo Pharma A/S Nouveaux dérivés d'amino-imidazopyrimidine utilisés en tant qu'inhibiteurs de janus kinase et leur utilisation pharmaceutique
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 (fr) 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 et procedes permettant d'inhiber la desoxyhypusine-synthase et la croissance cellulaire
WO1994017090A1 (fr) 1993-01-20 1994-08-04 Glaxo Group Limited Derives de la 2,6-diaminopurine
EP0659439A2 (fr) 1993-12-24 1995-06-28 MERCK PATENT GmbH Immunoconjugués
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 (fr) 1994-07-21 1996-02-08 Akzo Nobel N.V. Formulations de peroxides cetoniques cycliques
WO1996015111A1 (fr) 1994-11-15 1996-05-23 Regents Of The University Of Minnesota Procede et intermediaires de la synthese de korupensamines
WO1996030347A1 (fr) 1995-03-30 1996-10-03 Pfizer Inc. Derives de quinazoline
WO1996033978A1 (fr) 1995-04-27 1996-10-31 Zeneca Limited Derives de quinazoline
WO1996033980A1 (fr) 1995-04-27 1996-10-31 Zeneca Limited Derives de quinazoline
WO1996040210A1 (fr) 1995-06-07 1996-12-19 Imclone Systems Incorporated Anticorps et fragments d'anticorps inhibant la croissance des tumeurs
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 (fr) 1996-04-12 1997-10-23 Warner-Lambert Company Inhibiteurs irreversibles de tyrosine kinases
WO1998014451A1 (fr) 1996-10-02 1998-04-09 Novartis Ag Derive de pyrazole condense et procede pour sa 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 (fr) 1997-04-03 1998-10-08 American Cyanamid Company 3-cyano quinolines substituees
WO1998050433A2 (fr) 1997-05-05 1998-11-12 Abgenix, Inc. Anticorps monoclonaux humains contre le recepteur du facteur de croissance epidermique
WO1998050038A1 (fr) 1997-05-06 1998-11-12 American Cyanamid Company Utilisation de composes de la quinazoline dans le traitement de la maladie polykystique des reins
US5866572A (en) 1996-02-14 1999-02-02 Zeneca Limited Quinazoline derivatives
WO1999006396A1 (fr) 1997-07-29 1999-02-11 Warner-Lambert Company Inhibiteurs bicycliques irreversibles de tyrosine kinases
WO1999006378A1 (fr) 1997-07-29 1999-02-11 Warner-Lambert Company Inhibiteurs irreversibles de tyrosines kinases
WO1999009016A1 (fr) 1997-08-01 1999-02-25 American Cyanamid Company Derives de quinazoline substitues et leur utilisation en tant qu'inhibiteurs de la tyrosine kinase
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 (fr) 1997-11-06 1999-05-20 American Cyanamid Company Traitement des polypes du colon par des inhibiteurs de la tyrosine kinase a base de derives de quinazoline
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 (fr) * 2002-05-21 2003-11-27 Novartis Ag Derives 1h-imidazo[4,5-c]quinoline servant au traitement de maladies dependant de proteines kinases
WO2006040625A1 (fr) 2004-10-12 2006-04-20 Glenmark Pharmaceuticals S.A. Nouveaux inhibiteurs de dipeptidyle peptidase iv, compositions pharmaceutiques en contenant, et leur procede de preparation
WO2006062063A1 (fr) 2004-12-08 2006-06-15 Astellas Pharma Inc. Dérivé de la pipéridine et procédé servant à produire celui-ci
WO2006071862A2 (fr) 2004-12-30 2006-07-06 Takeda Pharmaceutical Company Limited Polymorphe de 1-(2-methylpropyl)-1h-imidazo[4,5-c][1,5]naphthyridin-4-amine ethanesulfonate
WO2008065021A1 (fr) 2006-11-30 2008-06-05 F. Hoffmann-La Roche Ag Dérivés de diaminocyclohexane et de diaminocyclopentane
WO2008121687A2 (fr) 2007-03-28 2008-10-09 Array Biopharma Inc. Composés imidazo[1,2-a]pyridine en tant qu'inhibiteurs de la tyrosine kinase récepteur
WO2009145719A1 (fr) 2008-05-30 2009-12-03 Astrazeneca Ab Dérivés d’iso-indoline comprenant un groupe cyano et leur utilisation dans le traitement de troubles de la douleur
WO2009152133A1 (fr) * 2008-06-10 2009-12-17 Abbott Laboratories Nouveaux composés tricycliques
WO2010038165A1 (fr) * 2008-09-30 2010-04-08 Pfizer Inc. Composés d'imidazo[1,5]naphtyridine, leur utilisation pharmaceutique et compositions associées

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 (fr) 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 et procedes permettant d'inhiber la desoxyhypusine-synthase et la croissance cellulaire
WO1994017090A1 (fr) 1993-01-20 1994-08-04 Glaxo Group Limited Derives de la 2,6-diaminopurine
US5475001A (en) 1993-07-19 1995-12-12 Zeneca Limited Quinazoline derivatives
EP0659439A2 (fr) 1993-12-24 1995-06-28 MERCK PATENT GmbH Immunoconjugués
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 (fr) 1994-07-21 1996-02-08 Akzo Nobel N.V. Formulations de peroxides cetoniques cycliques
US5804396A (en) 1994-10-12 1998-09-08 Sugen, Inc. Assay for agents active in proliferative disorders
WO1996015111A1 (fr) 1994-11-15 1996-05-23 Regents Of The University Of Minnesota Procede et intermediaires de la synthese de korupensamines
WO1996030347A1 (fr) 1995-03-30 1996-10-03 Pfizer Inc. Derives de quinazoline
US5770599A (en) 1995-04-27 1998-06-23 Zeneca Limited Quinazoline derivatives
WO1996033978A1 (fr) 1995-04-27 1996-10-31 Zeneca Limited Derives de quinazoline
WO1996033980A1 (fr) 1995-04-27 1996-10-31 Zeneca Limited Derives de quinazoline
WO1996040210A1 (fr) 1995-06-07 1996-12-19 Imclone Systems Incorporated Anticorps et fragments d'anticorps inhibant la croissance des tumeurs
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 (fr) 1996-04-12 1997-10-23 Warner-Lambert Company Inhibiteurs irreversibles de 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 (fr) 1996-10-02 1998-04-09 Novartis Ag Derive de pyrazole condense et procede pour sa preparation
US6002008A (en) 1997-04-03 1999-12-14 American Cyanamid Company Substituted 3-cyano quinolines
WO1998043960A1 (fr) 1997-04-03 1998-10-08 American Cyanamid Company 3-cyano quinolines substituees
US6235883B1 (en) 1997-05-05 2001-05-22 Abgenix, Inc. Human monoclonal antibodies to epidermal growth factor receptor
WO1998050433A2 (fr) 1997-05-05 1998-11-12 Abgenix, Inc. Anticorps monoclonaux humains contre le recepteur du facteur de croissance epidermique
WO1998050038A1 (fr) 1997-05-06 1998-11-12 American Cyanamid Company Utilisation de composes de la quinazoline dans le traitement de la maladie polykystique des reins
WO1999006396A1 (fr) 1997-07-29 1999-02-11 Warner-Lambert Company Inhibiteurs bicycliques irreversibles de tyrosine kinases
WO1999006378A1 (fr) 1997-07-29 1999-02-11 Warner-Lambert Company Inhibiteurs irreversibles de tyrosines kinases
WO1999009016A1 (fr) 1997-08-01 1999-02-25 American Cyanamid Company Derives de quinazoline substitues et leur utilisation en tant qu'inhibiteurs de la tyrosine kinase
WO1999024037A1 (fr) 1997-11-06 1999-05-20 American Cyanamid Company Traitement des polypes du colon par des inhibiteurs de la tyrosine kinase a base de derives de quinazoline
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 (fr) * 2002-05-21 2003-11-27 Novartis Ag Derives 1h-imidazo[4,5-c]quinoline servant au traitement de maladies dependant de proteines kinases
WO2006040625A1 (fr) 2004-10-12 2006-04-20 Glenmark Pharmaceuticals S.A. Nouveaux inhibiteurs de dipeptidyle peptidase iv, compositions pharmaceutiques en contenant, et leur procede de preparation
WO2006062063A1 (fr) 2004-12-08 2006-06-15 Astellas Pharma Inc. Dérivé de la pipéridine et procédé servant à produire celui-ci
WO2006071862A2 (fr) 2004-12-30 2006-07-06 Takeda Pharmaceutical Company Limited Polymorphe de 1-(2-methylpropyl)-1h-imidazo[4,5-c][1,5]naphthyridin-4-amine ethanesulfonate
WO2008065021A1 (fr) 2006-11-30 2008-06-05 F. Hoffmann-La Roche Ag Dérivés de diaminocyclohexane et de diaminocyclopentane
WO2008121687A2 (fr) 2007-03-28 2008-10-09 Array Biopharma Inc. Composés imidazo[1,2-a]pyridine en tant qu'inhibiteurs de la tyrosine kinase récepteur
WO2009145719A1 (fr) 2008-05-30 2009-12-03 Astrazeneca Ab Dérivés d’iso-indoline comprenant un groupe cyano et leur utilisation dans le traitement de troubles de la douleur
WO2009152133A1 (fr) * 2008-06-10 2009-12-17 Abbott Laboratories Nouveaux composés tricycliques
WO2010038165A1 (fr) * 2008-09-30 2010-04-08 Pfizer Inc. Composés d'imidazo[1,5]naphtyridine, leur utilisation pharmaceutique et compositions associées

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 (fr) * 2013-05-01 2014-11-06 Piramal Enterprises Limited Multi-thérapie anti-cancéreuse utilisant des dérivés de imidazo[4,5-c]quinoline
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 (zh) * 2015-09-14 2018-06-08 辉瑞大药厂 作为LRRK2抑制剂的新颖咪唑并[4,5-c]喹啉和咪唑并[4,5-c][1,5]萘啶衍生物
KR20180050407A (ko) * 2015-09-14 2018-05-14 화이자 인코포레이티드 LRRK2 억제제로서 이미다조[4,5-c]퀴놀린 및 이미다조[4,5-c][1,5]나프티리딘 유도체
KR102161364B1 (ko) * 2015-09-14 2020-09-29 화이자 인코포레이티드 LRRK2 억제제로서 이미다조[4,5-c]퀴놀린 및 이미다조[4,5-c][1,5]나프티리딘 유도체
RU2722149C1 (ru) * 2015-09-14 2020-05-27 Пфайзер Инк. Новые производные имидазо[4,5-c] хинолинов и имидазо[4,5-c][1,5] нафтиридинов в качестве ингибиторов LRRK2
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 (fr) * 2015-09-14 2017-03-23 Pfizer Inc. Nouveaux dérivés imidazo [4,5-c] quinoline et imidazo [4,5-c] [1,5] naphthyridine utilisés comme inhibiteurs de lrrk2
JP2018526422A (ja) * 2015-09-14 2018-09-13 ファイザー・インク LRRK2阻害薬としての新規のイミダゾ[4,5−c]キノリンおよびイミダゾ[4,5−c][1,5]ナフチリジン誘導体
WO2018067422A1 (fr) 2016-10-03 2018-04-12 Congxin Liang Nouveaux inhibiteurs sélectifs de jak1 et leurs utilisations
KR20190103231A (ko) * 2017-01-11 2019-09-04 레오 파마 에이/에스 야누스 키나아제 억제제로서 신규한 아미노-이미다조피리딘 유도체 및 그의 약제학적 용도
JP7009504B2 (ja) 2017-01-11 2022-01-25 レオ ファーマ アクティーゼルスカブ ヤヌスキナーゼ阻害剤としての新規アミノイミダゾピリジン誘導体及びそれらの医薬としての使用
JP2020504186A (ja) * 2017-01-11 2020-02-06 レオ ファーマ アクティーゼルスカブ ヤヌスキナーゼ阻害剤としての新規アミノイミダゾピリジン誘導体及びそれらの医薬としての使用
WO2018130563A1 (fr) 2017-01-11 2018-07-19 Leo Pharma A/S Nouveaux dérivés d'amino-imidazopyridine en tant qu'inhibiteurs de janus kinase et leur utilisation pharmaceutique
US10703751B2 (en) 2017-01-11 2020-07-07 Leo Pharma A/S Amino-imidazopyridine derivatives as Janus kinase inhibitors and pharmaceutical use thereof
KR102548543B1 (ko) 2017-01-11 2023-06-29 아킬리온 에이비 야누스 키나아제 억제제로서 신규한 아미노-이미다조피리딘 유도체 및 그의 약제학적 용도
KR20190138793A (ko) * 2017-03-10 2019-12-16 화이자 인코포레이티드 Lrrk2 억제제로서의 신규 이미다조[4,5-c]퀴놀린 유도체
KR102582626B1 (ko) 2017-03-10 2023-09-22 화이자 인코포레이티드 Lrrk2 억제제로서의 신규 이미다조[4,5-c]퀴놀린 유도체
CN107011216A (zh) * 2017-03-31 2017-08-04 泰州新威生物科技有限公司 一种反式‑4‑Boc‑氨基环己烷乙酸的制备方法
CN107089929A (zh) * 2017-06-22 2017-08-25 广西科技大学 反式‑(4‑氰甲基)环己基氨基甲酸叔丁酯的制备方法
US11304949B2 (en) 2018-03-30 2022-04-19 Incyte Corporation Treatment of hidradenitis suppurativa using JAK inhibitors
WO2020007698A1 (fr) 2018-07-06 2020-01-09 Leo Pharma A/S Nouveaux dérivés d'amino-imidazopyrimidine utilisés en tant qu'inhibiteurs de janus kinase et leur utilisation pharmaceutique
US11833155B2 (en) 2020-06-03 2023-12-05 Incyte Corporation Combination therapy for treatment of myeloproliferative neoplasms

Similar Documents

Publication Publication Date Title
WO2013007768A1 (fr) Composés hétérocycliques tricycliques, compositions et procédés d'utilisation de ces composés comme inhibiteurs des jak
US8461328B2 (en) Tricyclic heterocyclic compounds, compositions and methods of use thereof
EP2742040B1 (fr) Composes d'indazole, compositions pharmaceutiques et procedes d'utilisation de ceux-ci
WO2013007765A1 (fr) Composés tricycliques fusionnés utilisés en tant qu'inhibiteurs des janus kinases
WO2012085176A1 (fr) Composés pyrazinones tricycliques, leurs compositions et leurs procédés d'utilisation en tant qu'inhibiteurs de janus kinase
DK2989106T3 (en) CONDENSED HETEROCYCLIC COMPOUNDS AS PROTEINKINASE INHIBITORS
US20220127265A1 (en) Therapeutic compounds and uses thereof
WO2012066061A1 (fr) Pyrazolopyridines, et pyrazolopyridines et leur utilisation en tant qu'inhibiteurs de tyk2
MX2012010265A (es) Compuestos de imidazopiridina, composiciones y metodos de uso.
EP2616072A1 (fr) Composés d'azabenzothiazole, compositions et procédés d'utilisation
US20140206702A1 (en) Imidazopyridine compounds, compositions and methods of use
JP2022539259A (ja) キナーゼ阻害剤としてのヘテロ環式化合物
US20160326142A1 (en) Pyrazole carboxamide compounds, compositions and methods of use
WO2016001341A1 (fr) Composés sulfonylaminopyridine, compositions et procédés d'utilisation associés
WO2015049325A1 (fr) Inhibiteurs thérapeutiques de cdk8 et utilisations de ceux-ci
WO2016091916A1 (fr) Pyrazolylaminopurines en tant qu'inhibiteurs de la tyrosyne kinase (itk)
TW202300150A (zh) 環狀化合物及其使用方法

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