WO2013007765A1 - Composés tricycliques fusionnés utilisés en tant qu'inhibiteurs des janus kinases - Google Patents

Composés tricycliques fusionnés utilisés en tant qu'inhibiteurs des janus kinases Download PDF

Info

Publication number
WO2013007765A1
WO2013007765A1 PCT/EP2012/063621 EP2012063621W WO2013007765A1 WO 2013007765 A1 WO2013007765 A1 WO 2013007765A1 EP 2012063621 W EP2012063621 W EP 2012063621W WO 2013007765 A1 WO2013007765 A1 WO 2013007765A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkylene
tetraaza
indacen
dihydro
ethyl
Prior art date
Application number
PCT/EP2012/063621
Other languages
English (en)
Inventor
Philippe Bergeron
Monique Bodil Van Niel
Peter Dragovich
Christopher Hurley
Janusz Kulagowski
Sharada Labadie
Neville James Mclean
Rohan Mendonca
Rebecca Pulk
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 WO2013007765A1 publication Critical patent/WO2013007765A1/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
    • C07D471/02Heterocyclic 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders

Definitions

  • Compounds of formula I which are inhibitors of a Janus kinase, as well as compositions containing these compounds, and methods of use including, but not limited to, in vitro, in situ and in vivo diagnosis or treatment of mammalian cells.
  • 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).
  • 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.
  • One aspect includes a compound of formula I:
  • 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, -CH2CH 2 CH2CH 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 (C2-C18). In other examples, 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 .
  • Alkylene refers to a saturated, branched or straight chain hydrocarbon group having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent alkane. In one example, the divalent alkylene group is one to eighteen carbon atoms (d-C 18 ).
  • 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 C1-C3.
  • 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 - Cio, C 2 -Cs, C 2 -C 6 or C 2 -C3.
  • 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 -C 18 ).
  • the alkynylene radical is C 2 -C 12 , C 2 - Cio, C 2 -Cs, C 2 -C 6 or C 2 -C3.
  • 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, fiuocinonide, fluocinolone acetonide, halcinonide, betamethasone, betamethasone sodium phosphate, dexamethasone, dexamethasone sodium phosphate, fluocortolone, hydrocortisone- 17-butyrate, hydrocortisone- 17-valerate, aclometasone dipropionat
  • 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, theafiavins, fiavanols, 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 include 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 leucovorin.
  • proteosome inhibitor
  • Additional chemotherapeutic agents as defined herein include “anti-hormonal agents” or “endocrine therapeutics” which act to regulate, reduce, block, or inhibit the effects of hormones that can promote the growth of cancer. They may be hormones themselves, including, but not limited to: anti-estrogens with mixed agonist/antagonist profile, including, tamoxifen (NOLVADEX®), 4-hydroxytamoxifen, toremifene (FARESTON®), idoxifene, droloxifene, raloxifene (EVISTA®), trioxifene, keoxifene, and selective estrogen receptor modulators (SERMs) such as SERM3; pure anti-estrogens without agonist properties, such as fulvestrant (FASLODEX®), and EM800 (such agents may block estrogen receptor (ER) dimerization, inhibit DNA binding, increase ER turnover, and/or suppress ER levels); aromatase inhibitors, including steroidal aromatase inhibitors such as
  • 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-l inhibitors such as antisense agent ISIS-5132 available from ISIS Pharmaceuticals which inhibit Raf-1 signaling; non-HER targeted TK
  • 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.
  • the cycloalkyl group is 3 to 12 carbon atoms (C 3 -C 12 ).
  • cycloalkyl is C 3 -Cs, C 3 -Cio or C 5 -C 10 .
  • the cycloalkyl group, as a monocycle is C 3 -Cs, C 3 -C 6 or C5-C6.
  • the cycloalkyl group, as a bicycle is C 7 -C 12 .
  • the cycloalkyl group is C 5 - C12.
  • 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” as used herein refers to a derivative of the hydroxy group commonly employed to block or protect the hydroxy group while reactions are carried out on other functional groups on the compound.
  • 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. G. W. McOmie, Ed., Plenum Press, New York, NY, 1973, Chapter 5, and T.W. Greene, “Protective Groups in Organic Synthesis", John Wiley and Sons, New York, NY, 1981.
  • the term "protected hydroxy” refers to a hydroxy group substituted with one of the above hydroxy- protecting groups.
  • Heterocyclic group “heterocyclic”, “heterocycle”, “heterocyclyl”, or “heterocyclo” alone, and when used as a moiety in a complex group such as a heterocycloalkyl group, are used interchangeably and refer to any mono-, bi-, tricyclic or spiro, saturated or unsaturated, aromatic (heteroaryl) or non-aromatic, ring system, having 3 to 20 ring atoms, where the ring atoms are carbon, and at least one atom in the ring or ring system is a heteroatom selected from nitrogen, sulfur or oxygen.
  • 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. [NR 4 ] C1 " , [NR 4 ] OH " ).
  • 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, thioiuran, 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 unsubstituted or substituted by one or more (e.g. 0, 1, 2, 3 or 4) of the substituents listed for that group in which said substituents may be the same or different. In an embodiment an optionally substituted group has 1 substituent. In another embodiment an optionally substituted group has 2 substituents. In another embodiment an optionally substituted group has 3 substituents.
  • 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.
  • R w -R x -R y if the group R x is described as -CH 2 C(0)-, then it is understood that this group can be bonded both as R w -CH 2 C(0)-R y , and as R w -C(0)CH 2 -R y , 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
  • “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.
  • 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.
  • 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.
  • 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.
  • compound(s) of this invention 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:
  • V is CR 4 or N
  • W is C
  • X and Z are N
  • Y is CR 5 or N
  • V is CR 4 or N
  • W is N
  • X is CH
  • Y is CR 5
  • Z is C
  • R 1 is C ? _i 2 cycloalkyl or 3-12 membered heterocyclyl, wherein R 1 is independently optionally substituted by halogen, oxo, -CN, -OR a , -S(O) 0 - 2 R a , -NR a R b , Ci_ 3 alkylene, Ci_ 6 alkyl, -(C 0-3 alkylene)C 3 _ 6 cycloalkyl, C 2 _ 6 alkenyl, C 2 _ 6 alkynyl, -(C 0-3 alkylene)phenyl, or -(C 0- 3 alkylene)3-6 membered heterocyclyl, wherein said alkyl, alkylene, alkenyl and alkynyl are independently optionally substituted by oxo, -NR c R d , -OR c , -CN or halogen and said C 3 _ 6 cycloalkyl, phenyl and
  • R 4 is hydrogen, halogen, CN or Ci_ 3 alkyl
  • R 5 is Ci-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 , -(Co-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 , -(Co-3 alkylene)C(0)NR a R b , -(C 0 . 3 alkylene)C(0)OR a , -(C 0 .
  • alkylene)NR a S(0)i_ 2 NR a R b -(C 0 - 3 alkylene)C 3 _i 2 cycloalkyl, -(C 0-3 alkylene)C 6 _i 4 aryl, -(C 0 _ 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, -(Co_ 3 alkylene)CN, -(C 0-3 alkylene)OR c , -(Co-3 alkylene)NR c R d , -(C 0 .
  • each 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) !
  • each R c and R d are independently hydrogen, C e alkyl, C 2 _6 alkenyl, C 2-6 alkynyl, -(Co -3 alkylene)C 3 _ 6 cycloalkyl, -(C 0-3 alkylene)3-12 membered
  • each R e , R f , R g , R h , R are independently hydrogen or Ci_6 alkyl optionally substituted by halogen or oxo.
  • Formula I includes compounds other than: N-[(l S,3S,4R)-3-(2-aminoimidazo[4,5-d]pyrrolo[2,3-b]pyridin- l(6H)-yl)-4- ethylcyclopentyl] -cyclopropanesulfonamide ; N-((lR,3S,4R)-3-ethyl-4-(pyrrolo[2,3-b][l,2,3]triazolo[4,5-d]pyridin-l(6H)- yl)cyclopentyl)cyclopropanesulfonamide;
  • R 5 is other than NH 2 , OH, methyl, difluoromethyl, trifluoromethyl or cyclopropyl. In certain embodiments, R 5 is other than OH.
  • V is CR 4 or N
  • W is N
  • X is CH
  • Y is CR 5
  • Z is C.
  • V is CR 4
  • W is N
  • X is CH
  • Y is CR 5
  • Z is C.
  • V is CR 4 or N
  • W is C
  • X and Z are N
  • Y is CR 5 or N.
  • V is CR 4
  • W is C
  • X and Z are N
  • Y is CR 5 or N.
  • V is N
  • W is C
  • X and Z are N
  • Y is CR 5 or N.
  • V is CR 4
  • W is C
  • X and Z are N
  • Y is CR 5 .
  • V is CR 4 , W is C, X and Z are N, and Y is N. In certain embodiments, V is N, W is C, X and Z are N, and Y is N.
  • V is CR 4
  • W is C
  • X and Z are N
  • Y is N.
  • R 1 is 3-12 membered heterocyclyl, wherein R 1 is independently optionally substituted by halogen, oxo, -CN, -OR a , -S(O) 0 - 2 R a , -NR a R b , Ci_ 3 alkylene, Ci_ 6 alkyl, C 3 _ 6 cycloalkyl, C 2 _ 6 alkenyl, C 2 _ 6 alkynyl, phenyl, or 3-6 membered heterocyclyl, wherein said alkyl, alkenyl and alkynyl are independently optionally substituted by oxo,
  • R 1 is 6-membered heterocyclyl, optionally substituted by halogen, oxo, -CN, -OR a , -S(O) 0 _ 2 R a , -NR a R b , Ci_ 3 alkylene, Ci_ 6 alkyl, C 3 _ 6 cycloalkyl, C 2 _ 6 alkenyl, C 2 _ 6 alkynyl, phenyl, or 3-6 membered heterocyclyl, wherein said alkyl, alkenyl and alkynyl are independently optionally substituted by oxo, -NR c R d , -OR c , -CN or halogen and said C 3 _6 cycloalkyl, phenyl and 3-6 membered heterocyclyl are independently optionally substituted by R 6 .
  • R 1 is tetrahydropyranyl, tetrahydro-2H-thiopyranyl or piperidinyl, optionally substituted by halogen, oxo, -CN, -OR a , -S(O) 0 _ 2 R a , -NR a R b , Ci_ 3 alkylene, Ci_ 6 alkyl, C 3 _ 6 cycloalkyl, C 2 _ 6 alkenyl, C 2 _ 6 alkynyl, phenyl, or 3-6 membered heterocyclyl, wherein said alkyl, alkenyl and alkynyl are independently optionally substituted by oxo, -NR c R d , -OR c , -CN or halogen and said C 3 _ 6 cycloalkyl, phenyl and 3-6 membered heterocyclyl are independently optionally substituted by R 6 .
  • R 1 is C 3 _i 2 cycloalkyl, wherein R 1 is independently optionally substituted by halogen, oxo, -CN, -OR a , -S(O) 0 _ 2 R a , -NR a R b , Ci_ 3 alkylene, Ci_ 6 alkyl, C 3 _ 6 cycloalkyl, C 2 _ 6 alkenyl, C 2 _ 6 alkynyl, phenyl, or 3-6 membered heterocyclyl, wherein said alkyl, alkenyl and alkynyl are independently optionally substituted by oxo, -NR c R d , -OR c , -CN or halogen and said C 3 _ 6 cycloalkyl, phenyl and 3-6 membered heterocyclyl are independently optionally substituted by R 6 .
  • R 1 is cyclopentyl or cyclohexyl, wherein R 1 is independently optionally substituted by halogen, oxo, -CN, -OR a , -S(O) 0 _ 2 R a , -NR a R b , Ci_ 3 alkylene, Ci_ 6 alkyl, C 3 _ 6 cycloalkyl, C 2 _ 6 alkenyl, C 2 _ 6 alkynyl, phenyl, or 3-6 membered heterocyclyl, wherein said alkyl, alkenyl and alkynyl are independently optionally substituted by oxo, -NR c R d , -OR c , -CN or halogen and said C 3 _ 6 cycloalkyl, phenyl and 3-6 membered heterocyclyl are independently optionally substituted by R 6 .
  • R 1 is cyclohexyl optionally substituted by halogen, oxo, -CN, -OR a , -S(O) 0 - 2 R a , -NR a R b , Ci_3 alkylene, Ci_ 6 alkyl, C 3 _ 6 cycloalkyl, C 2 _ 6 alkenyl, C 2 _ 6 alkynyl, phenyl, or 3-6 membered heterocyclyl, wherein said alkyl, alkenyl and alkynyl are independently optionally substituted by oxo, -NR c R d , -OR c , -CN or halogen and said C 3 _ 6 cycloalkyl, phenyl and 3-6 membered heterocyclyl are independently optionally substituted by R 6 .
  • R 1 is cyclohexyl optionally substituted by halogen, oxo, -CN, -OR a , -NR a R b , Ci_ 3 alkylene or Ci_ 6 alkyl optionally substituted by halogen.
  • R 1 is cyclohexyl optionally substituted by halogen, oxo, Ci_ 3 alkylene 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, wherein R 1 is optionally substituted by halogen, oxo, -CN, -OR a , -NR a R b , Ci_ 3 alkylene or Ci_ 6 alkyl optionally substituted by halogen, and wherein the wavy line represents the point of attachment in formula I.
  • R 1 is cyclopentyl halogen, oxo, -CN, -OR a , -S(0)o_ 2 R a ,
  • R 1 is cyclopentyl optionally substituted by halogen, oxo, -CN, -OR a , -NR a R b , Ci_ 3 alkylene or Ci_6 alkyl optionally substituted by halogen.
  • R 1 is cyclopentyl.
  • R 1 is tetrahydropyranyl, tetrahydro-2H-thiopyranyl, piperidinyl, cyclopentyl or cyclohexyl, wherein R 1 is independently optionally substituted by halogen, oxo, -CN, -OR a , -S(0)o- 2 R a , -NR a R b , Ci_ 3 alkylene, Ci_ 6 alkyl, C 3 _ 6 cycloalkyl, C 2 _ 6 alkenyl, C 2 _ 6 alkynyl, phenyl, or 3-6 membered heterocyclyl, wherein said alkyl, alkenyl and alkynyl are independently optionally substituted by oxo, -NR c R d , -OR c , -CN or halogen and said C 3 _ 6 cycloalkyl, phenyl and 3-6 membered heterocyclyl are independently optionally substituted by
  • -R 1 is selected from:
  • R 4 is hydrogen, methyl, CN In another embodiment, R 4 is hydrogen or CN. In certain embodiments, R 4 is hydrogen.
  • R 5 is 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 , -(C0-3 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 C 1 -12 alkyl, C 2 _i 2 alkenyl, C 2 _i 2 alkynyl, -(C 1 -3 alkylene)CN, -(Ci_ 3 alkylene)NR a R b , -(Ci_ 3 alkylene)OR a , -(Ci_ 3 alkylene)SR a , -(Ci_ 3 alkylene)C(0)R a , -(Ci_ 3 alkylene)NR a C(0)R b , -(Ci_ 3 alkylene)C(0)NR a R b , -(Ci_ 3 alkylene)C(0)OR a , -(Ci_ 3 alkylene)OC(0)R a , -(Ci_ 3 alkylene)NR a C(0)NR a R b , -(Ci_ 3 alkylene)OC(0)NR a R b , -(Ci_ 3 alkylene)-(Ci
  • R 5 is -(C 1-3 alkylene)CN, -(C 1-3 alkylene)NR a R b , -(C 1 -3 alkylene)OR a , -(Ci_ 3 alkylene)SR a , -(Ci_ 3 alkylene)C(0)R a , -(Ci_ 3 alkylene)NR a C(0)R b , -(Ci_ 3 alkylene)C(0)NR a R b , -(Ci_ 3 alkylene)C(0)OR a , -(Ci_ 3 alkylene)OC(0)R a , -(Ci_ 3 alkylene)NR a C(0)NR a R b , -(Ci_ 3 alkylene)OC(0)NR a R b , -(Ci_ 3 alkylene)NR a C(0)OR b , -(Ci_ 3 alkylene)S(0)i_ 2 R a , -(Ci_
  • 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, -(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 selected from methyl, ethyl, -CH 2 OH, wherein the wavy line represents the point of attachment in formula I.
  • 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 substituted by one or more of 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 methyl or ethyl substituted by one or more OH.
  • 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 -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 .
  • R 5 is selected
  • 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 , -(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)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 -NHCH 2 CH 2 OH, -CF 2 CH 2 NH 2 , -CH 2 C(0)NH 2 , wherein the wavy line represents the point of attachment in formula I.
  • 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 . 3 alkylene)C(0)NR c R d , -(C 0 .
  • R 5 is -(C 0-3 alkylene)C3_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 other than cyclopropyl.
  • R 5 is ⁇ ⁇
  • R 5 is -(C 0-3 alkylene)C3_7 cycloalkyl. In another embodiment, R 5 is cyclopropyl or eye lo butyl.
  • 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)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 . 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)NR a C(0)R , 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 is -(Co_ 3 alkylene)C(0)OR a , wherein said alkylene is independently optionally substituted by halogen, oxo, -(Co_ 3 alkylene)CN, -(Co_ 3 alkylene)OR c , alkylene)NR -(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 , alkylene)NR c C(0)R d , -(C 0 .
  • R 5 is -(C 0-3 alkylene)C(0)OR a , wherein said alkylene is optionally substituted by halogen, oxo, -CN, -OR c ,
  • R is selected from
  • R 5 is -(C 0-3 alkylene)NR a S(0)i_ 2 R b , wherein said alkylene is independently optionally substituted by halogen, oxo, -(C 0-3 alkylene)CN, -(C 0-3 alkylene)OR c , alkylene)NR -(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 , alkylene)NR c C(0)R d , -(C 0 .
  • R 5 is -(C0-3 alkylene)NR a S(0)i_2R 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 CH 3 , - CH 2 NHS(0) 2 cyclopropyl, -CH 2 NHS(0) 2 cyclopentyl, -CH 2 N(CH 3 )S(0) 2 CH 3 ,
  • R 5 is -(C0-3 alkylene)5-12 membered heteroaryl, wherein said alkylene and heteroaryl are independently optionally substituted by halogen, oxo, -(C 0-3 alkylene)CN, -(C0-3 alkylene)OR c , -(C0-3 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.
  • 5 is selected from -CH 2 CH 2 triazolyl, triazolyl, pyridinyl, -CH 2 pyrazolyl, -CH 2 pyridinyl,
  • 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 ( 1,1-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 , wherein the wavy line represents the point of attachment in formula I.
  • 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 -(C 0-3 alkylene) S(0)i_ 2 R 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 -(C0-3 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 , -(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)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 substituted by oxo, -CN or halo en.
  • R 5 is selected from -CH 2 phenyl, phenyl,
  • 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 -(C 0-3 alkylene)NR a C(0)OR 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)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 , -CH 2 NHC(0)OCH 3 ,
  • R 5 is Ci_i 2 alkyl optionally substituted by OR c , -(Ci_ 3 alkylene)NR a R b , -(Ci_ 3 alkylene)OR a , -(Ci_ 3 alkylene)NR a C(0)R b , -(Ci_ 3 alkylene)NR a C(0)NR a R b , -(Ci_ 3 alkylene)NR a C(0)OR b , -(Ci_ 3 alkylene)NR a C(0)OR b , -(Ci_ 3 alkylene)NR a S(0)i_ 2 R b or ⁇ (Ci_ 3 alkylene)3-12 membered heterocyclyl, wherein said alkylene and heterocyclyl are independently optionally substituted by halogen, oxo or Ci_ 6 alkyl optionally substituted by halogen.
  • R 5 is ethyl substituted by OH, -(Ci_ 3 alkylene)NR a R b , -(Ci_ 3 alkylene)OR a , -(Ci_ 3 alkylene)NR a C(0)R b , -(Ci_ 3 alkylene)NR a C(0)NR a R b , -(Ci_ 3 alkylene)NR a C(0)OR b , -(Ci_ 3 alkylene)NR a S(0)i_ 2 R b or -(Ci_ 3 alkylene)3-12 membered heterocyclyl, wherein said alkylene and heterocyclyl are independently optionally substituted by halogen, oxo or Ci_ 6 alkyl optionally substituted by halogen.
  • R is selected from methyl, ethyl,
  • 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.
  • each R a and R b are independently hydrogen, Ci_ 6 alkyl, C 2 _ 6 alkenyl, C 2 _ 6 alkynyl, -S(0)i_ 2 R', -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
  • 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, -S(0) 2 CH 3 , -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
  • 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.
  • one R a is H and one R b is Ci_ 6 alkyl 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 g R h , C3-6 cycloalkyl, 3-6 membered heterocyclyl, phenyl or Ci_ 3 alkyl optionally substituted by oxo or halogen, or taken together with the atom to which they
  • each R c and R d are independently hydrogen, Ci_ 6 alkyl, C 2 _ 6 alkenyl, C 2 _ 6 alkynyl, -C 3 _ 6 cycloalkyl, -3-12 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-d
  • 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 certain embodiments, each R c and R d are independently hydrogen, methyl or ethyl.
  • 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.
  • each R c , R d , R e , R f , R g , R h and R 1 are independently hydrogen or methyl.
  • each R e , R f , R g , R h and R 1 are independently hydrogen, methyl, ethyl, propyl or isopropyl, optionally substituted by halogen or oxo. In certain embodiments, each R e , R f , R g , R h and R' are independently hydrogen, methyl or ethyl.
  • R 1 is C5-7 cycloalkyl independently substituted by one wherein R a is H and R b is Ci_ 6 alkyl 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 (O) 1 _ 2 NR g R h , C 3 _ 6 cycloalkyl, 3-6 membered heterocyclyl, phenyl or Ci_ 3 alkyl optionally
  • Another embodiment includes a compound selected from Examples 1-146. Another embodiment includes a compound selected from Examples 1-147.
  • 2-Fluoro-cyclopropanecarboxylic acid (2- ⁇ 1 -[4-(2,2,2-trifluoro-ethylamino)-cyclohexyl]- 1 ,6-dihydro- 1 ,3,5 ,6-tetraaza-as-indacen-2-yl ⁇ -ethyl)-amide; trans N-(2- ⁇ l-[4-(2,2,2-Trifluoro-ethylamino)-cyclohexyl]-l,6-dihydro-l,3,5,6-tetraaza-as- indacen-2-yl ⁇ -ethyl)-isobutyramide; trans Cyclobutanecarboxylic acid (2- ⁇ l-[4-(2,2,2-trifluoro-ethylamino)-cyclohexyl]-l,6- dihydro-l,3,5,6-tetraaza-as-indacen-2-yl
  • Ethanesulfonic acid [ 1 -(-4,4-difluoro-tetrahydro-pyran-3-yl)- 1 ,6-dihydro- 1 ,3,5,6-tetraaza- as-indacen-2-ylmethyl]-amide; Cyclopropanesulfonic acid [l-(-4,4-difluoro-tetrahydro-pyran-3-yl)-l,6-dihydro-l, 3,5,6- tetraaza-as-indacen-2-ylmethyl]-amide;
  • Compounds of the invention may contain one or more asymmetric carbon atoms. Accordingly, the compounds may exist as diastereomers, enantiomers or mixtures thereof.
  • the syntheses of the compounds may employ racemates, diastereomers or enantiomers as starting materials or as intermediates. Mixtures of particular diastereomeric compounds may be separated, or enriched in one or more particular diastereomers, by chromatographic or crystallization methods. Similarly, enantiomeric mixtures may be separated, or enantiomerically enriched, using the same techniques or others known in the art.
  • Each of the asymmetric carbon or nitrogen atoms may be in the R or S configuration and both of these configurations are within the scope of the invention.
  • Another aspect includes prodrugs of the compounds of formula I, including known amino -protecting and carboxy-protecting groups which are released, for example hydrolyzed, to yield the compound of formula I under physiologic conditions.
  • a particular class of prodrugs are compounds in which a nitrogen atom in an amino, amidino, amino alky leneamino, iminoalkyleneamino or guanidino group is substituted with a hydroxy (OH) group, an alkylcarbonyl (-CO-R) group, an alkoxycarbonyl (-CO-OR), an acyloxyalkyl-alkoxycarbonyl (- CO-O-R-O-CO-R) group where R is a monovalent or divalent group, for example alkyl, alkylene or aryl, or a group having the formula -C(0)-0-CPlP2-haloalkyl, where PI and P2 are the same or different and are hydrogen, alkyl, alkoxy, cyano, halogen, alkyl or aryl.
  • 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 1-25 depicted below provide routes for synthesizing the compounds of the present invention as well as key intermediates.
  • Examples section below 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.
  • 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.
  • 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).
  • BOC t-butoxycarbonyl
  • Fmoc 9-fluorenylmethyleneoxycarbonyl
  • azaindole where X is CR 4
  • imidazopyridine where X is N
  • a substituted amine for example -NH 2 R 1
  • imidazole formation can be achieved using two general methods, i) Amide bond formation by treatment with an acid chloride or a carboxlic acid in the presence of a suitable coupling reagent such as EDCI or HATU will give an amide which can then be dehydratively cyclized by treating with a reagent such as glacial acetic acid. Deprotection will give compound 2.3a. ii) Treatment with an imidate, followed by deprotection will give compound 2.3a. Further independent derivatization of compound 2.3a gives compound 2.4a.
  • Compounds of formula I can also be synthesized as shown in Reaction Scheme 4.
  • compound 3.1 can be cyclized, for example with 2-chloro-2-oxoethyl acetate, deprotected and hydrolyzed to give tricyclic alcohol compounds 4.1.
  • Alcohols 4.1 can undergo amination to give compounds 4.2, or further derivatized with R a -Lg (where Lg is a leaving group) to give compounds 4.3.
  • Compound 5.2 can be derivatized with an amine having the formula H 2 NR 1 , for example with commercially available (i?)-l-benzyl-3-aminopiperidine, in the presence of base such as diisopropylethylamine.
  • Reaction Scheme 6 illustrates the synthesis of compounds of formula I, for example compounds 6.5.
  • Compound 6.1 can be prepared by treatment of compound 5.2 with a suitably protected diamine, for example commercially available l-Boc-3-aminoazetidine, in the presence of base such as diisopropylethylamine. Reduction of compound 6.1 with hydrogen in the presence of palladium on carbon gives diamine compound 6.2. Cyclization of compound 6.2 with R 5 -substituted orthoformate, for example triethyl orthoformate (where R 5 is hydrogen), in presence of /?-toluenesulfonic acid gives tricyclic compound 6.3. Hydrolysis of compound 6.3 with aqueous sodium hydroxide in methanol/tetrahydrofuran (THF) provides compound 6.4. Deprotection of 6.4 with an acid, such as trifluoro acetic acid, gives compound 6.5.
  • a suitably protected diamine for example commercially available l-Bo
  • Reaction Scheme 7 illustrates the synthesis of compounds of formula I, for example compounds 7.4.
  • Protected 3,4-diaminoazaindole 5.4 can be cyclised using n-butyl nitrite in the presence of copper (II) bromide to give l,6-dihydropyrrolo[2,3-b][l,2,3]triazolo[4,5- d]pyridine compound 7.1.
  • Deprotection of compound 7.1 using aqueous sodium hydroxide in methanol provides compounds 7.2.
  • Treatment of compound 7.2 with ammonium formate and palladium (II) hydroxide in refluxing methanol provides compounds 7.3.
  • Compounds 7.3 can be derivatized by reacting with compounds of the formula Lg-R (where Lg is a leaving group), for example, carboxylic acids of the formula RC0 2 H (where each R is independently an optionally substituted group, such as alkyl, alkenyl, alkynyl, cycloalkyl, aryl or heterocyclyl) in the presence of suitable coupling reagents such as N- hydroxybenzotriazole (HOBt), 1 -ethyl-3-(3-dimethylaminopropyl)carbodiimide hydrochloride (EDCI) and DMAP in dichloromethane (DCM).
  • Lg-R where Lg is a leaving group
  • carboxylic acids of the formula RC0 2 H where each R is independently an optionally substituted group, such as alkyl, alkenyl, alkynyl, cycloalkyl, aryl or heterocyclyl
  • suitable coupling reagents such as N-
  • Reaction Scheme 8 illustrates the synthesis of compounds 8.5.
  • protected 3,4- diaminoazaindole 5.4 can be treated with ethyl malonyl chloride in the presence of base such as triethylamine and then cyclized in the presence of acetic acid to give imidazolo compound 8.1.
  • Compound 8.1 can be reduced with a reducing agent, such as lithium aluminium hydride, to provide alcohol 8.2.
  • a reducing agent such as lithium aluminium hydride
  • Deprotection of compound 8.2 using aqueous sodium hydroxide in methanol provides compounds 8.3.
  • Treatment of compound 8.3 with ammonium formate and palladium (II) hydroxide in re fluxing methanol provides compounds 8.4.
  • Compounds 8.4 can be converted using, for example, carboxylic acids in the presence of suitable coupling reagents such as HOBt and EDCI in DCM, to provide compounds 8.5.
  • Reaction Scheme 9 illustrates the synthesis of compounds 9.4.
  • Amino compounds such as 5.7 can be alkylated to give compounds 9.1 using a 2-substituted ethene in ethanol heated under reflux.
  • Compounds 5.7 can be alkylated to give compounds 9.3 using an appropriate aldehyde or ketone, or an oxo-substitued compound such as R-C(0)-R, in the presence of a suitable reducing agent such as sodium triacetoxyborohydride either in the presence or absence of acetic acid.
  • compounds 5.7 can be alkylated with a suitable haloalkane (where Lg is a leaving group such as a halogen) in the presence of base such as potassium carbonate in THF to provide compounds of 9.4.
  • Reaction Scheme 10 illustrates the synthesis of compounds 10.2 and 10.3.
  • amino compounds 10.1 can be treated with various functionalized sulfonyl chlorides in presence of base such as triethylamine to give compounds 10.2.
  • Compounds 10.1 can be coupled to various acid chlorides in the presence of base such as triethylamine to give compounds 10.3.
  • Reaction Scheme 11 illustrates the synthesis of compounds 11.1 and 11.2.
  • Amino compounds 10.1 can be treated with a suitable heteroaryl chloride, such as 4-chloropyridine, in presence of base, such as diisopropylethylamine, to provide compounds 11.1.
  • Compounds 10.1 can be coupled to aryl or heteroaryl boronic acids in the presence of copper (II) acetate, either in the presence or absence of an oxygen atmosphere, using a suitable solvent, such as dichloromethane, to give compounds 11.2.
  • Reaction Scheme 12 illustrates the synthesis of compounds 12.5.
  • a suitably protected 3,4-diaminoazaindole such as compound 12.1 can be cyclised with a suitable amidine in the presence of a suitable solvent, such as ethanol, to give substituted imidazolo compound 12.2.
  • Compound 12.2 can be hydrolysed using an aqueous base such as lithium hydroxide in a compatible solvent, such as THF, to provide acid salt compound 12.3.
  • Compound 12.4 can be prepared from compound 12.3 using suitable primary or secondary amines in the presence of a suitable coupling reagent, such as HATU, in a compatible solvent, such as DMF.
  • a suitable coupling reagent such as HATU
  • Reaction Scheme 13 illustrates an alternative synthesis of compounds 12.5.
  • Compound 14 can be treated with suitable amines, such as methylamine, in a compatible solvent, such as ethanol, at elevated temperatures to directly provide amide compound 12.4.
  • Compound 12.4 can be hydrolysed using aqueous sodium hydroxide to give compound 12.5.
  • Reaction Scheme 14 illustrates the synthesis of compounds 14.2.
  • a protected 3,4- diaminoazaindole such as compound 12.1
  • can be cyclised with a triethyl orthoalkane such as triethyl orthoacetate, in the presence of catalytic /?-toluenesulfonic acid with prolonged heating to give substituted imidazolo compound 14.1.
  • Hydrolysis of compound 14.1 with aqueous sodium hydroxide in methanol provides compounds 14.2.
  • Reaction Scheme 15 illustrates the synthesis of compounds 14.2.
  • Reaction Scheme 15 illustrates the synthesis of compounds 15.3.
  • a suitably protected 3,4-diaminoazaindole such as compound 12.1 can be treated with acetoxyacetyl chloride in the presence of base, such as triethylamine, and then cyclized in the presence of acetic acid to give substituted imidazolo compound 15.1.
  • Compound 15.1 can be hydro lysed using an aqueous base such as lithium hydroxide in a compatible solvent, such as THF, to provide alcohol compound 15.2.
  • Hydrolysis of compound 15.2 with aqueous sodium hydroxide in methanol provides compounds 15.3.
  • Reaction Scheme 16 illustrates the synthesis of compounds 16.2.
  • alcohol compound 15.2 can be treated with methanesulfonyl chloride in the presence of a suitable base, such as triethylamine, and the resulting product can be reacted with a compatible amine or lactam, such as 2-pyrrolidinone, in the presence of a suitable base, such as sodium hydride, to provide compound 16.1.
  • a suitable base such as sodium hydride
  • Reaction Scheme 17 illustrates the synthesis of compounds 17.7.
  • the preparation of compound 17.1 has been previously described (see: Itoh et. al, J. Heterocyclic Chem., 19, 513- 517 (1982)).
  • Compound 17.1 can be treated with methanesulfonyl chloride in the presence of a suitable base, such as triethylamine, to give compound 17.2.
  • a suitable base such as triethylamine
  • Nitration of 17.2 using tetrabutylammonium nitrate in the presence of trifluoro acetic anhydride gives compound 17.3.
  • Compound 17.3 can be reacted with an appropriate primary amine to give compound 17.4, which can then be treated with a reducing reagent, such as iron, in the presence of ammonium chloride to give aniline 17.5.
  • Compound 17.5 can be cyclized to give imidazole 17.6, which can then be hydro lyzed with aqueous sodium hydroxide to give compound 17.
  • Reaction Scheme 18 illustrates the synthesis of compound 18.6.
  • Compound 5.2 can be treated with a brominating reagent such as N-bromosuccinimide to give compound 18.1, which can then be treated with an appropriate primary amine to give intermediate 18.2.
  • Compound 18.2 can be treated with a reducing agent, such as iron, in the presence of ammonium chloride to give aniline 18.3, which then can be cyclized to give imidazole 18.4.
  • Compound 18.4 can be treated with alkylating reagents, such as methyl zinc chloride and tetrakis(triphenylphosphine)palladium(0), or trimethylboroxine in the presence of [ ⁇ , - Bis(diphenylphosphino)ferrocene] dichloropalladium(II) and sodium hydrogen carbonate, to give alkylated compound 18.5.
  • alkylating reagents such as methyl zinc chloride and tetrakis(triphenylphosphine)palladium(0), or trimethylboroxine
  • [ ⁇ , - Bis(diphenylphosphino)ferrocene] dichloropalladium(II) and sodium hydrogen carbonate to give alkylated compound 18.5.
  • Compound 18.5 can then be hydro lyzed with aqueous sodium hydroxide to give compounds 18.6.
  • Reaction Scheme 19 illustrates the synthesis of compounds 19.5.
  • Compound 5.2 can be treated with a fluorinating reagent, such as Select-Fluor, to give compound 19.1, which can then be treated with an appropriate primary amine to give intermediate 19.2.
  • Compound 19.2 can then be treated with a reducing agent, such as iron in the presence of ammonium chloride, to give aniline 19.3, which then can be cyclized to give imidazole 19.4.
  • Compound 19.4 can be hydro lyzed with aqueous sodium hydroxide to give compounds 19.5.
  • Reaction Scheme 20 illustrates the synthesis of compounds 20.2.
  • compound 20.1 can be treated with a fluorinating agent, such as Select-Fluor, to directly provide compounds 20.2.
  • Reaction Scheme 21
  • Compounds of type 21.4 and 21.5 can be synthesized from compounds of type 21.1 by cyclization for example with triethyl ortho formate.
  • Halogenenation of 21.2 can be achieved for example by treating 21.2 with a suitable base such as lithium diisopropylamide then quenching with a suitable halogen source such as an N-halosuccinimide.
  • Compounds of type 21.4 can be prepared by displacing the halogen in 21.3 with a suitable nucleophile such as sodium methoxide.
  • Compounds of type 21.5 can similarly be prepared by displacement of the halogen in 21.3 with a suitable amine such as ethanolamine.
  • Compounds of type 22.3 can be prepared directly from compounds of type 22.1 by reaction with a reagent such as dichloromethylene-dimethyliminium chloride. Alternatively, compounds of type 22.3 can be prepared by first reacting compounds of type 22.1 with a reagent such as an alkyl isothiocyanate. Compounds of type 22.3 may be prepared by cyclisation of a compound of type 22.2 by reaction with a reagent such as l-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride.
  • Reaction Scheme 26 illustrates the synthesis of compounds 17.8.
  • Nitration of 26.1 provides 26.2.
  • the aniline group present in 26.2 may be converted to aryl chloride 26.3 under standard conditions.
  • Treatment of activated aryl chloride 26.3 with a primary amine in the presence of a base such as triethylamine provides anilines 26.4.
  • Reduction of the nitro group under conditions such as iron and ammonium chloride provides dianilines 26.5.
  • Cyclization of the dianilines with a reagent such as a triethyl orthoalkane provides imidazoles 26.6.
  • Displacement of the aryl halide groups of 26.6 with amino groups provides dianilines 26.7, which may be further cyclized with triethylortho acetate in the presence of formic acid to provide compounds 26.8.
  • 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
  • reagents 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.
  • 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 diastereoisomers 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.
  • an appropriate optically active compound e.g., chiral auxiliary such as a chiral alcohol or Mosher's acid chloride
  • some of the compounds of the present invention may be atropisomers (e.g., substituted biaryls) and are considered as part of this invention. Enantiomers can also be separated by use of a chiral HPLC column or supercritical fluid chromatography.
  • a single stereoisomer, e.g. an enantiomer, substantially free of its stereoisomer may be obtained by resolution of the racemic mixture using a method such as formation of diastereomers using optically active resolving agents (Eliel, E. and Wilen, S., Stereochemistry of Organic Compounds, John Wiley & Sons, Inc., New York, 1994; Lochmuller, C. H., J. Chromatogr., 113(3):283-302 (1975)).
  • Racemic mixtures of chiral compounds of the invention can be separated and isolated by any suitable method, including: (1) formation of ionic, diastereomeric salts with chiral compounds and separation by fractional crystallization or other methods, (2) formation of diastereomeric compounds with chiral derivatizing reagents, separation of the diastereomers, and conversion to the pure stereoisomers, and (3) separation of the substantially pure or enriched stereoisomers directly under chiral conditions.
  • 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, renal, prostate, vulval, thyroid, large intestine, endometrial, uterine, brain, central nervous system, cancer of the peritoneum, hepatocellular cancer, head cancer, neck
  • 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).
  • 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 (RITUXA ®), 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 homotri
  • 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
  • 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
  • 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. In certain embodiments, 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.
  • BWFI bacteriostatic water for injection
  • non-exemplified compounds according to the invention may be successfully performed by modifications apparent to those skilled in the art, e.g., by appropriately protecting interfering groups, by utilizing other suitable reagents known in the art other than those described, and/or by making routine modifications of reaction conditions.
  • other reactions disclosed herein or known in the art will be recognized as having applicability for preparing other compounds of the invention.
  • HM-N Isolute® HM-N is a modified form of diatomaceous earth
  • 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% methanol 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.
  • Scan range 110-800amu
  • Scan range 110-800amu
  • Scan range 110-800amu
  • Method G HPLC instrument: Waters Acquity UPLC; mobile phase A: H 2 0 with 0.1% formic acid; mobile phase B: CH3CN with 0.1% formic acid; column: Acquity UPLC BEH C18, 1.7 um, 2.1 x 30 mm; column temperature: 80 °C; LC gradient: 5-95% B in 1.4 min, 95% in 0.3 min; LC flowrate: 800uL/min; UV wavelength: 220 nm and 254 nm; mass spectrometer: Waters SQ detector; ionization: ESI+; scan range: 100-800 amu.
  • Method H 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 I 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.
  • Scan range 100-lOOOamu
  • Scan range 100-lOOOamu
  • Scan range 100-lOOOamu
  • Scan range 150-1250amu
  • Scan range 60-1000amu
  • Method Q Experiments were performed on a 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 R Experiments were performed on a VG Platform II quadrupole spectrometer is linked to a Hewlett Packard HP 1050 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 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.
  • Mobile phase B Methanol with 0.1% ammonium hydroxide
  • Mobile phase B Methanol with 0.1% ammonium hydroxide Isocratic conditions with 30% Mobile phase B
  • Mobile phase B methanol with 0.1% ammonium hydroxide Isocratic conditions with 20% Mobile phase B
  • 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.
  • the catalytically active kinase domain of human JAKl, JAK2, JAK3 or TYK2 was purified from extracts of SF9 insect cells infected with a recombinant baculo virus expression vector encoding the human JAKl, JAK2, JAK3 or TYK2 kinase domains (JAKl amino acid residues N852-D1154 according to the numbering of GenBank sequence accession number P23458, JAK2 amino acid residues D812-G1132 according to the numbering of GenBank sequence accession number NP 004963.1; JAK3 amino acid residues S783-S1124 according to the numbering of GenBank sequence accession number P52333, and TYK2 amino acid residues N873-C1187 according to the numbering of GenBank sequence accession number P29597).
  • the activity of the JAKl, JAK2, JAK3 or TYK2 kinase domains can be measured by a number of direct and indirect methods, including quantification of phosphorylation of peptide substrates derived from the human JAK3 protein (Saltzman et al, Biochem. Biophys. Res. Commun. 246:627-633 (2004)).
  • the activity of the JAKl, JAK2, JAK3 or TYK2 kinase domains was measured in vitro by monitoring phosphorylation of JAK3 derived peptides using the Caliper LabChip technology.
  • JAK2 Inhibition Assay Protocol The activity of the isolated JAK2 kinase domain was measured by monitoring phosphorylation of a peptide derived from JAK3 (Val- Ala-Leu- Val-Asp-Gly-Tyr-Phe-Arg-Leu- Thr-Thr) fluorescent ly labelled on the N-terminus with 5-carboxyf uorescein using the Caliper LabChip technology (Caliper Life Sciences, Hopkinton, MA).
  • K inhibition constants
  • the proportion of phosphorylated product was determined as a fraction of total peptide substrate using the Caliper LabChip 3000 according to the manufacturer's specifications. K values were then determined using the Morrison tight binding model. Morrison, J.F., Biochim. Biophys. Acta. 185:269-296 (1969); William, J.W. and Morrison, J.F., Meth. Enzymol, 63:437-467 (1979).
  • JAK1 and TYK2 Inhibition Assay Protocol The activity of the isolated JAK1 or TYK2 kinase domain was measured by monitoring phosphorylation of a peptide derived from JAK3 (Val- Ala-Leu- Val-Asp-Gly-Tyr-Phe-Arg-Leu- Thr-Thr) fluorescent ly labelled on the N-terminus with 5-carboxyfluorescein using the Caliper LabChip technology (Caliper Life Sciences, Hopkinton, MA).
  • Ki inhibition constants
  • the activity of the isolated JAK3 kinase domain was measured by monitoring phosphorylation of a peptide derived from JAK3 (Leu-Pro-Leu-Asp-Lys-Asp-Tyr-Tyr-Val-Val- Arg) fluorescent ly labelled on the N-terminus with 5-carboxyfluorescein using the Caliper LabChip technology (Caliper Life Sciences, Hopkinton, MA).
  • Ki inhibition constants
  • the activities of compounds were determined in cell-based assays that are designed to measure JAK2- dependent signaling or proliferation.
  • Compounds were serially diluted in DMSO and incubated with Set-2 cells (German Collection of Microorganisms and Cell Cultures (DSMZ); Braunschweig, Germany), which express the JAK2V617F mutant protein, in 96-well microtiter plates for 1 hr at 37°C in RPMI medium at a final cell density of 100,000 cells per well and a final DMSO concentration of 0.57%.
  • Set-2 cells German Collection of Microorganisms and Cell Cultures (DSMZ); Braunschweig, Germany
  • NK92 cells American Type Culture Collection (ATCC); Manassas, VA) in 96-well microtiter plates in RPMI medium at a final cell density of 100,000 cells per well and a final DMSO concentration of 0.57%.
  • Human recombinant IL-12 R&D systems; Minneapolis, MN was then added at a final concentration of lOng/mL to the microtiter plates containing the NK92 cells and compound and the plates were incubated for 1 hr at 37 °C.
  • Compound-mediated effects on STAT4 phosphorylation were then measured in the lysates of incubated cells using the Meso Scale Discovery (MSD) technology (Gaithersburg, Maryland) according to the manufacturer's protocol and EC50 values were determined.
  • MSD Meso Scale Discovery
  • the activities of compounds were determined in cell-based assays that are designed to measure JAK1 or JAK2- dependent signaling.
  • Compounds were serially diluted in DMSO and incubated with TF-1 cells (American Type Culture Collection (ATCC); Manassas, VA) in 384- well microtiter plates in OptiMEM medium without phenol red, 1% Charcoal/Dextran stripped FBS, 0.1 mM NEAA, ImM sodium pyruvate (Invitrogen Corp.; Carlsbad, CA) at a final cell density of 100,000 cells per well and a final DMSO concentration of 0.2%.
  • ATCC American Type Culture Collection
  • VA Manassas, VA
  • OptiMEM medium without phenol red
  • Charcoal/Dextran stripped FBS 0.1 mM NEAA
  • ImM sodium pyruvate Invitrogen Corp.; Carlsbad, CA
  • IL-6 Human recombinant IL-6 (R&D systems; Minneapolis, MN) or EPO (Invitrogen Corp.; Carlsbad, CA) was then added at a final concentration of 30 ng/mL or 10 Units/mL, respectively, to the microtiter plates containing the TF-1 cells and compound and the plates were incubated for 30 min at 37 °C.
  • Compound-mediated effects on STAT3 or STAT5 phosphorylation were then measured in the lysates of cells incubated in the presence of IL-6 or EPO, respectively, using the Meso Scale Discovery (MSD) technology (Gaithersburg, Maryland) according to the manufacturer's protocol and EC50 values were determined.
  • MSD Meso Scale Discovery
  • NK92 cells American Type Culture Collection (ATCC); Manassas, VA) in 384-well microtiter plates in RPMI medium at a final cell density of 50,000 cells per well and a final DMSO concentration of 0.2%.
  • Human recombinant IL-12 R&D systems; Minneapolis, MN was then added at a final concentration of 30ng/ml to the microtiter plates containing the NK92 cells and compound and the plates were incubated for 45 min at 37°C.
  • Compound-mediated effects on STAT4 phosphorylation were then measured in the lysates of incubated cells using the Meso Scale Discovery (MSD) technology (Gaithersburg, Maryland) according to the manufacturer's protocol and EC50 values were determined.
  • MSD Meso Scale Discovery
  • H-7 H-8 H-9 H-10 To a solution of compound H-7 (31 g, 0.14 mol) in EtOH (1.5 L) at -20 °C was added
  • Dess-Martin periodinane (0.326 g, 0.761 mmol) was added to a solution of racemic- ⁇ 2-[6-benzenesulfonyl- 1 -((lR,2R,4S,6S)-6-hydroxy-bicyclo[2.2.1 ]hept-2-yl)- 1 ,6-dihydro- l,3,5,6-tetraaza-as-indacen-2-yl]-ethyl ⁇ -carbamic acid tert-butyl ester (0.210 g. 0.381 mmol) in CH 2 CI 2 at 25 °C.
  • reaction mixture was stirred for 1.5 h at 25 °C, then was partitioned between half- saturated NaHC0 3 (100 ml) and EtOAc (2 x 100 ml). The organic layers were dried over MgSC ⁇ , filtered and was concentrated under reduced pressure. Purification of the residue by column chromatography on silica gel (gradient: 0 to 7% CH 3 OH in CH 2 CI 2 ) afforded racemic- ⁇ 2-[6-benzenesulfonyl-l-((lR,2R,4S)-6-oxo-bicyclo[2.2.
  • reaction mixture was stirred for 18 h at 25 °C, then was partitioned between half- saturated NaHC0 3 (100 ml) and EtOAc (2 x 100 ml). The organic layers were dried over MgSC ⁇ , filtered and was concentrated under reduced pressure.
  • the reaction mixture was stirred at 50 °C for 26 h, then was cooled to 25 °C.
  • HCl 2.0 ml of a 1.0 M solution in water
  • saturated NaHC03 (0.50 ml) were added sequentially.
  • the resulting mixture was concentrated under reduced pressure overnight at 50 °C (GeneVacc).
  • the residue was suspended in DMF (2 ml) and was thoroughly mixed via vortexing and spatula scraping.
  • This material was prepared from racemic-(lS,2R,4S,5R)-5-(l-benzenesulfonyl-5- nitro-lH-pyrrolo[2,3-b]pyridin-4-ylamino)-bicyclo[2.2.1]heptan-2-ol in a manner analogous to that described for the conversion of racemic-(lR,2S,4S,6R)-6-(l-benzenesulfonyl-5-nitro-lH- pyrrolo[2,3-b]pyridin-4-ylamino)-bicyclo[2.2.1]heptan-2-ol to racemic- ⁇ 2- [l-(6-hydro xy- bicyclo[2.2.

Abstract

L'invention concerne des nouveaux composés représentés par la formule générale (I) dans laquelle R1, V, W, X, Y et Z sont comme représentés dans la description. Par voie de conséquence, les composés peuvent être inclus dans des nouvelles compostions de qualité pharmaceutique et utilisés pour le traitement de troubles immunologiques ou hyperprolifératifs.
PCT/EP2012/063621 2011-07-13 2012-07-12 Composés tricycliques fusionnés utilisés en tant qu'inhibiteurs des janus kinases WO2013007765A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201161507561P 2011-07-13 2011-07-13
US61/507,561 2011-07-13

Publications (1)

Publication Number Publication Date
WO2013007765A1 true WO2013007765A1 (fr) 2013-01-17

Family

ID=46651468

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2012/063621 WO2013007765A1 (fr) 2011-07-13 2012-07-12 Composés tricycliques fusionnés utilisés en tant qu'inhibiteurs des janus kinases

Country Status (1)

Country Link
WO (1) WO2013007765A1 (fr)

Cited By (49)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2014525397A (ja) * 2011-08-12 2014-09-29 日産化学工業株式会社 3環性ヘテロ環化合物及びjak阻害剤
CN104391058A (zh) * 2014-11-25 2015-03-04 广东东阳光药业有限公司 阿法替尼及其异构体的检测方法
US9079912B2 (en) 2005-12-13 2015-07-14 Incyte Corporation Heteroaryl substituted pyrrolo[2,3-B] pyridines and pyrrolo[2,3-B] pyrimidines as Janus kinase inhibitors
US9181271B2 (en) 2012-11-01 2015-11-10 Incyte Holdings Corporation Tricyclic fused thiophene derivatives as JAK inhibitors
US9193733B2 (en) 2012-05-18 2015-11-24 Incyte Holdings Corporation Piperidinylcyclobutyl substituted pyrrolopyridine and pyrrolopyrimidine derivatives as JAK 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
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
JP2017506656A (ja) * 2014-03-28 2017-03-09 ノバルティス ティーアゲズントハイト アーゲー 新規化合物
US9611269B2 (en) 2011-06-20 2017-04-04 Incyte Corporation Azetidinyl phenyl, pyridyl or pyrazinyl carboxamide derivatives as JAK inhibitors
US9655854B2 (en) 2013-08-07 2017-05-23 Incyte Corporation Sustained release dosage forms for a JAK1 inhibitor
US9802957B2 (en) 2014-04-30 2017-10-31 Incyte Corporation Processes of preparing a JAK1 inhibitor and new forms thereto
WO2018015289A1 (fr) 2016-07-19 2018-01-25 Bayer Cropscience Aktiengesellschaft Dérivés hétérocycliques bicycliques condensés utilisés comme pesticides
WO2018112382A1 (fr) 2016-12-16 2018-06-21 Janssen Pharmaceutica Nv Imidazopyrrolopyridine en tant qu'inhibiteurs de la famille jak de kinases
WO2018112379A1 (fr) 2016-12-16 2018-06-21 Janssen Pharmaceutica Nv Inhibiteurs à petites molécules de la famille jak des kinases
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
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
US10166191B2 (en) 2012-11-15 2019-01-01 Incyte Corporation Sustained-release dosage forms of ruxolitinib
CN110483514A (zh) * 2019-09-16 2019-11-22 维眸生物科技(上海)有限公司 一种氰基取代的环肼衍生物及其应用
WO2019239387A1 (fr) 2018-06-15 2019-12-19 Janssen Pharmaceutica Nv Inhibiteurs à petites molécules de la famille jak de kinases
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
CN110724142A (zh) * 2019-10-29 2020-01-24 维眸生物科技(上海)有限公司 一种作为jak激酶抑制剂的酰胺或磺酰胺取代的肼衍生物
US10640506B2 (en) 2010-11-19 2020-05-05 Incyte Holdings Corporation Cyclobutyl substituted pyrrolopyridine and pyrrolopyrimidines derivatives as JAK inhibitors
WO2020088659A1 (fr) * 2018-11-01 2020-05-07 Lynk Pharmaceuticals Co. Ltd. Inhibiteurs de janus kinase 1 tricycliques, et compositions et procédés associés
WO2020100959A1 (fr) 2018-11-15 2020-05-22 日本新薬株式会社 Composé de 1,3,4-oxadiazolone et médicament
JP2020522550A (ja) * 2017-06-09 2020-07-30 ノバルティス アーゲー 軟骨形成を誘導するための化合物及び組成物
EA037347B1 (ru) * 2017-12-08 2021-03-16 Янссен Фармацевтика Нв Низкомолекулярные ингибиторы киназ семейства jak
WO2021104488A1 (fr) * 2019-11-27 2021-06-03 正大天晴药业集团股份有限公司 Composé tri-hétérocyclique en tant qu'inhibiteur de jak et son utilisation
CN114149428A (zh) * 2020-09-08 2022-03-08 中国药科大学 吡啶并环类化合物及其制备方法、中间体、组合物和应用
WO2022074572A1 (fr) * 2020-10-08 2022-04-14 Unichem Laboratories Ltd Composés tricycliques substitués
US11304949B2 (en) 2018-03-30 2022-04-19 Incyte Corporation Treatment of hidradenitis suppurativa using JAK inhibitors
WO2022182839A1 (fr) * 2021-02-25 2022-09-01 Incyte Corporation Lactames spirocycliques utilisés comme inhibiteurs du v617f de jak2
CN115385911A (zh) * 2021-05-22 2022-11-25 南京康屹生物医药中心(有限合伙) 一种二吡咯并吡啶结构的化合物、制备方法和医药用途
WO2022247885A1 (fr) * 2021-05-26 2022-12-01 正大天晴药业集团股份有限公司 Cristal et sel de composé trihétérocyclique et leur utilisation
US11661422B2 (en) 2020-08-27 2023-05-30 Incyte Corporation Tricyclic urea compounds as JAK2 V617F inhibitors
WO2023093718A1 (fr) * 2021-11-25 2023-06-01 启元生物(杭州)有限公司 Nouvelle forme cristalline du composé, son procédé de préparation et son utilisation
WO2023115327A1 (fr) * 2021-12-21 2023-06-29 中国药科大学 Composé cyclique de pyrido, son procédé de préparation, intermédiaire, composition et application
US11691971B2 (en) 2020-06-19 2023-07-04 Incyte Corporation Naphthyridinone compounds as JAK2 V617F inhibitors
US11753413B2 (en) 2020-06-19 2023-09-12 Incyte Corporation Substituted pyrrolo[2,1-f][1,2,4]triazine compounds as JAK2 V617F inhibitors
US11767323B2 (en) 2020-07-02 2023-09-26 Incyte Corporation Tricyclic pyridone compounds as JAK2 V617F inhibitors
US11780840B2 (en) 2020-07-02 2023-10-10 Incyte Corporation Tricyclic urea compounds as JAK2 V617F inhibitors
US11833155B2 (en) 2020-06-03 2023-12-05 Incyte Corporation Combination therapy for treatment of myeloproliferative neoplasms
WO2024041586A1 (fr) * 2022-08-25 2024-02-29 启元生物(杭州)有限公司 Promédicament d'inhibiteur de kinase jak
US11919908B2 (en) 2020-12-21 2024-03-05 Incyte Corporation Substituted pyrrolo[2,3-d]pyrimidine compounds as JAK2 V617F inhibitors

Citations (34)

* 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
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
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
WO1998050433A2 (fr) 1997-05-05 1998-11-12 Abgenix, Inc. Anticorps monoclonaux humains contre le recepteur du facteur de croissance epidermique
US5866572A (en) 1996-02-14 1999-02-02 Zeneca Limited Quinazoline derivatives
WO1999006378A1 (fr) 1997-07-29 1999-02-11 Warner-Lambert Company Inhibiteurs irreversibles de tyrosines kinases
WO1999006396A1 (fr) 1997-07-29 1999-02-11 Warner-Lambert Company Inhibiteurs bicycliques irreversibles de tyrosine 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
WO2011068881A1 (fr) * 2009-12-01 2011-06-09 Abbott Laboratories Composés tricycliques innovants
WO2011086053A1 (fr) * 2010-01-12 2011-07-21 F. Hoffmann-La Roche Ag Composés hétérocycliques tricycliques, leurs compositions et procédés d'utilisation

Patent Citations (45)

* 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
US5475001A (en) 1993-07-19 1995-12-12 Zeneca Limited Quinazoline derivatives
EP0659439A2 (fr) 1993-12-24 1995-06-28 MERCK PATENT GmbH Immunoconjugués
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
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
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
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
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
US6344459B1 (en) 1996-04-12 2002-02-05 Warner-Lambert Company Irreversible inhibitors of tyrosine kinases
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
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
WO1998043960A1 (fr) 1997-04-03 1998-10-08 American Cyanamid Company 3-cyano quinolines substituees
US6002008A (en) 1997-04-03 1999-12-14 American Cyanamid Company Substituted 3-cyano quinolines
WO1998050433A2 (fr) 1997-05-05 1998-11-12 Abgenix, Inc. Anticorps monoclonaux humains contre le recepteur du facteur de croissance epidermique
US6235883B1 (en) 1997-05-05 2001-05-22 Abgenix, Inc. Human monoclonal antibodies to epidermal growth factor receptor
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
WO1999006378A1 (fr) 1997-07-29 1999-02-11 Warner-Lambert Company Inhibiteurs irreversibles de tyrosines kinases
WO1999006396A1 (fr) 1997-07-29 1999-02-11 Warner-Lambert Company Inhibiteurs bicycliques irreversibles de tyrosine 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
WO2011068881A1 (fr) * 2009-12-01 2011-06-09 Abbott Laboratories Composés tricycliques innovants
WO2011086053A1 (fr) * 2010-01-12 2011-07-21 F. Hoffmann-La Roche Ag Composés hétérocycliques tricycliques, leurs compositions et procédés d'utilisation

Non-Patent Citations (41)

* Cited by examiner, † Cited by third party
Title
"Beilsteins Handbuch der organischen", SPRINGER-VERLAG
"Chiral Liquid Chromatography", 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
CHANGELIAN ET AL., SCIENCE, vol. 302, 2003, pages 875 - 878
E. HASLAM: "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 of Radiation Therapy, Cancer, in Principles I and Practice of Oncology", vol. 1, 1993, pages: 24875
ITOH, J. HETEROCYCLIC CHEM., vol. 19, 1982, pages 513 - 517
J. ORG. CHEM., vol. 74, no. 5, 2009, pages 1932 - 1938
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
MARK ZAK ET AL: "Discovery and Optimization of C -2 Methyl Imidazopyrrolopyridines as Potent and Orally Bioavailable JAK1 Inhibitors with Selectivity over JAK2", JOURNAL OF MEDICINAL CHEMISTRY, vol. 55, no. 13, 14 June 2012 (2012-06-14), pages 6176 - 6193, XP055037261, ISSN: 0022-2623, DOI: 10.1021/jm300628c *
MORRISON, J.F., BIOCHIM. BIOPHYS. ACTA, vol. 185, 1969, pages 269 - 296
NICOLAOU ET AL., ANGEW. CHEM INTL. ED. ENGL., vol. 33, 1994, pages 183 - 186
OKAMOTO, J. OF CHROMATOGR., vol. 513, 1990, pages 375 - 378
O'SHEA ET AL., CELL, vol. 109, 2002, pages S121 - S131
REICH ET AL., NAT. REV. DRUG DISCOV., vol. 8, 2009, pages 355 - 356
ROWE; RAYMOND C: "Handbook of Pharmaceutical Excipients", 2005, PHARMACEUTICAL PRESS
SALTZMAN ET AL., BIOCHEM. BIOPHYS. RES. COMMUN., vol. 246, 2004, pages 627 - 633
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
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
WILLIAM, J.W.; MORRISON, J.F., METH. ENZYMOL., vol. 63, 1979, pages 437 - 467

Cited By (122)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
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
US9079912B2 (en) 2005-12-13 2015-07-14 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
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
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
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
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
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
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
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
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
JP2014525397A (ja) * 2011-08-12 2014-09-29 日産化学工業株式会社 3環性ヘテロ環化合物及びjak阻害剤
US9359358B2 (en) 2011-08-18 2016-06-07 Incyte Holdings Corporation Cyclohexyl azetidine derivatives as JAK inhibitors
US9487521B2 (en) 2011-09-07 2016-11-08 Incyte Holdings Corporation Processes and intermediates for making a JAK inhibitor
US9718834B2 (en) 2011-09-07 2017-08-01 Incyte 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
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
US10370387B2 (en) 2012-11-01 2019-08-06 Incyte Holdings 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
US9181271B2 (en) 2012-11-01 2015-11-10 Incyte Holdings 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
US11576865B2 (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
US11337927B2 (en) 2012-11-15 2022-05-24 Incyte Holdings Corporation Sustained-release dosage forms of ruxolitinib
US10166191B2 (en) 2012-11-15 2019-01-01 Incyte Corporation Sustained-release dosage forms of ruxolitinib
US11896717B2 (en) 2012-11-15 2024-02-13 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
US9221845B2 (en) 2013-03-06 2015-12-29 Incyte Holdings Corporation Processes and intermediates for making a JAK inhibitor
US11045421B2 (en) 2013-08-07 2021-06-29 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
US10561616B2 (en) 2013-08-07 2020-02-18 Incyte Corporation Sustained release dosage forms for a JAK1 inhibitor
US10059709B2 (en) 2014-03-28 2018-08-28 Elanco Tiergesundheit Ag Compounds
US10370375B2 (en) 2014-03-28 2019-08-06 Elanco Tiergesundheit Ag Heterocyclyl-substituted cyclohexylsulfonamides as JAK inhibitors
JP2017506656A (ja) * 2014-03-28 2017-03-09 ノバルティス ティーアゲズントハイト アーゲー 新規化合物
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
CN104391058A (zh) * 2014-11-25 2015-03-04 广东东阳光药业有限公司 阿法替尼及其异构体的检测方法
CN104391058B (zh) * 2014-11-25 2015-12-02 广东东阳光药业有限公司 阿法替尼及其异构体的检测方法
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
WO2018015289A1 (fr) 2016-07-19 2018-01-25 Bayer Cropscience Aktiengesellschaft Dérivés hétérocycliques bicycliques condensés utilisés comme pesticides
KR102352462B1 (ko) 2016-12-16 2022-01-17 얀센 파마슈티카 엔.브이. Jak 패밀리의 키나제의 소분자 억제제
AU2017377072B2 (en) * 2016-12-16 2021-08-26 Janssen Pharmaceutica Nv Imidazopyrrolopyridine as inhibitors of the JAK family of kinases
JP2020502113A (ja) * 2016-12-16 2020-01-23 ヤンセン ファーマシューティカ エヌ.ベー. Jakファミリーのキナーゼの低分子阻害物質
US10294226B2 (en) 2016-12-16 2019-05-21 Janssen Pharmaceutica Nv Small molecule inhibitors of the JAK family of kinases
IL267215B (en) * 2016-12-16 2022-09-01 Janssen Pharmaceutica Nv Imidazopyrrolopyridines as inhibitors of the jak family of kinases
US11827638B2 (en) 2016-12-16 2023-11-28 Janssen Pharmaceutica Nv Imidazopyrrolopyridine as inhibitors of the JAK family of kinases
AU2022204240B2 (en) * 2016-12-16 2023-10-05 Janssen Pharmaceutica Nv Small molecule inhibitors of the JAK family of kinases
JP7153647B2 (ja) 2016-12-16 2022-10-14 ヤンセン ファーマシューティカ エヌ.ベー. Jakファミリーのキナーゼの阻害剤としてのイミダゾピロロピリジン
AU2017377069B2 (en) * 2016-12-16 2020-07-23 Janssen Pharmaceutica Nv Small molecule inhibitors of the JAK family of kinases
EP4086256A1 (fr) 2016-12-16 2022-11-09 Janssen Pharmaceutica NV Inhibiteurs à petites molécules de la famille de kinases jak
US20180170931A1 (en) * 2016-12-16 2018-06-21 Janssen Pharmaceutica Nv Small Molecule Inhibitors of the JAK Family of Kinases
WO2018112379A1 (fr) 2016-12-16 2018-06-21 Janssen Pharmaceutica Nv Inhibiteurs à petites molécules de la famille jak des kinases
CN114685505A (zh) * 2016-12-16 2022-07-01 詹森药业有限公司 作为jak家族激酶抑制剂的咪唑并吡咯并吡啶
US10364246B2 (en) 2016-12-16 2019-07-30 Janssen Pharmaceutica Nv Small molecule inhibitors of the JAK family of kinases
US10981911B2 (en) 2016-12-16 2021-04-20 Janssen Pharmaceutica Nv Imidazopyrrolopyridine as inhibitors of the JAK family of kinases
WO2018112382A1 (fr) 2016-12-16 2018-06-21 Janssen Pharmaceutica Nv Imidazopyrrolopyridine en tant qu'inhibiteurs de la famille jak de kinases
TWI777997B (zh) * 2016-12-16 2022-09-21 比利時商健生藥品公司 Jak激酶家族之小分子抑制劑
US11059823B2 (en) 2016-12-16 2021-07-13 Janssen Pharmaceutica Nv Small molecule inhibitors of the JAK family of kinases
KR20190086779A (ko) * 2016-12-16 2019-07-23 얀센 파마슈티카 엔.브이. Jak 패밀리의 키나제의 소분자 억제제
JP2020502141A (ja) * 2016-12-16 2020-01-23 ヤンセン ファーマシューティカ エヌ.ベー. Jakファミリーのキナーゼの阻害剤としてのイミダゾピロロピリジン
JP7291077B2 (ja) 2016-12-16 2023-06-14 ヤンセン ファーマシューティカ エヌ.ベー. Jakファミリーのキナーゼの低分子阻害物質
AU2021273566B2 (en) * 2016-12-16 2023-04-13 Janssen Pharmaceutica Nv Imidazopyrrolopyridine as inhibitors of the JAK family of kinases
CN110312719B (zh) * 2016-12-16 2022-04-15 詹森药业有限公司 作为jak家族激酶抑制剂的咪唑并吡咯并吡啶
US10487083B2 (en) 2016-12-16 2019-11-26 Janssen Pharmaceutica Nv Small molecule inhibitors of the JAK family of kinases
CN110088105A (zh) * 2016-12-16 2019-08-02 詹森药业有限公司 Jak家族激酶的小分子抑制剂
CN110312719A (zh) * 2016-12-16 2019-10-08 詹森药业有限公司 作为jak家族激酶抑制剂的咪唑并吡咯并吡啶
CN110088105B (zh) * 2016-12-16 2022-03-18 詹森药业有限公司 Jak家族激酶的小分子抑制剂
JP7441264B2 (ja) 2016-12-16 2024-02-29 ヤンセン ファーマシューティカ エヌ.ベー. Jakファミリーのキナーゼの阻害剤としてのイミダゾピロロピリジン
KR20190103231A (ko) * 2017-01-11 2019-09-04 레오 파마 에이/에스 야누스 키나아제 억제제로서 신규한 아미노-이미다조피리딘 유도체 및 그의 약제학적 용도
KR102548543B1 (ko) 2017-01-11 2023-06-29 아킬리온 에이비 야누스 키나아제 억제제로서 신규한 아미노-이미다조피리딘 유도체 및 그의 약제학적 용도
US10703751B2 (en) 2017-01-11 2020-07-07 Leo Pharma A/S Amino-imidazopyridine derivatives as Janus kinase inhibitors and pharmaceutical use thereof
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
JP7229945B2 (ja) 2017-06-09 2023-02-28 ノバルティス アーゲー 軟骨形成を誘導するための化合物及び組成物
JP7432778B2 (ja) 2017-06-09 2024-02-16 ノバルティス アーゲー 軟骨形成を誘導するための化合物及び組成物
US11753416B2 (en) 2017-06-09 2023-09-12 Novartis Ag Compounds and compositions for inducing chondrogenesis
JP2020522550A (ja) * 2017-06-09 2020-07-30 ノバルティス アーゲー 軟骨形成を誘導するための化合物及び組成物
EA037347B1 (ru) * 2017-12-08 2021-03-16 Янссен Фармацевтика Нв Низкомолекулярные ингибиторы киназ семейства jak
US11304949B2 (en) 2018-03-30 2022-04-19 Incyte Corporation Treatment of hidradenitis suppurativa using JAK inhibitors
CN112313233A (zh) * 2018-06-15 2021-02-02 詹森药业有限公司 Jak家族激酶的小分子抑制剂
JP2021528391A (ja) * 2018-06-15 2021-10-21 ヤンセン ファーマシューティカ エヌ.ベー. Jakファミリーのキナーゼの低分子阻害物質
US11066406B2 (en) 2018-06-15 2021-07-20 Janssen Pharmaceutica Nv Small molecule inhibitors of the JAK family of kinases
US11787807B2 (en) 2018-06-15 2023-10-17 Janssen Pharmaceutica Nv Small molecule inhibitors of the JAK family of kinases
WO2019239387A1 (fr) 2018-06-15 2019-12-19 Janssen Pharmaceutica Nv Inhibiteurs à petites molécules de la famille jak de kinases
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
US11858933B2 (en) 2018-11-01 2024-01-02 Lynk Pharmaceuticals Co. Ltd. Tricyclic Janus kinase 1 inhibitors, and compositions and methods thereof
US11851431B2 (en) 2018-11-01 2023-12-26 Lynk Pharmaceuticals Co. Ltd. Tricyclic Janus kinase 1 inhibitors, and compositions and methods thereof
WO2020088659A1 (fr) * 2018-11-01 2020-05-07 Lynk Pharmaceuticals Co. Ltd. Inhibiteurs de janus kinase 1 tricycliques, et compositions et procédés associés
CN113396150A (zh) * 2018-11-01 2021-09-14 凌科药业(杭州)有限公司 三环janus激酶1抑制剂及其组合物和方法
CN113396150B (zh) * 2018-11-01 2024-03-12 凌科药业(杭州)有限公司 三环janus激酶1抑制剂及其组合物和方法
WO2020100959A1 (fr) 2018-11-15 2020-05-22 日本新薬株式会社 Composé de 1,3,4-oxadiazolone et médicament
CN110483514B (zh) * 2019-09-16 2022-06-14 启元生物(杭州)有限公司 一种氰基取代的环肼衍生物及其应用
CN110483514A (zh) * 2019-09-16 2019-11-22 维眸生物科技(上海)有限公司 一种氰基取代的环肼衍生物及其应用
WO2021051899A1 (fr) * 2019-09-16 2021-03-25 维眸生物科技(上海)有限公司 Dérivé d'hydrazine cyclique cyano-substitué et son application
CN110724142B (zh) * 2019-10-29 2022-09-02 启元生物(杭州)有限公司 一种作为jak激酶抑制剂的酰胺或磺酰胺取代的肼衍生物
CN110724142A (zh) * 2019-10-29 2020-01-24 维眸生物科技(上海)有限公司 一种作为jak激酶抑制剂的酰胺或磺酰胺取代的肼衍生物
WO2021104488A1 (fr) * 2019-11-27 2021-06-03 正大天晴药业集团股份有限公司 Composé tri-hétérocyclique en tant qu'inhibiteur de jak et son utilisation
CN114728967A (zh) * 2019-11-27 2022-07-08 正大天晴药业集团股份有限公司 作为jak抑制剂的三并杂环类化合物及其应用
US11833155B2 (en) 2020-06-03 2023-12-05 Incyte Corporation Combination therapy for treatment of myeloproliferative neoplasms
US11691971B2 (en) 2020-06-19 2023-07-04 Incyte Corporation Naphthyridinone compounds as JAK2 V617F inhibitors
US11753413B2 (en) 2020-06-19 2023-09-12 Incyte Corporation Substituted pyrrolo[2,1-f][1,2,4]triazine compounds as JAK2 V617F inhibitors
US11767323B2 (en) 2020-07-02 2023-09-26 Incyte Corporation Tricyclic pyridone compounds as JAK2 V617F inhibitors
US11780840B2 (en) 2020-07-02 2023-10-10 Incyte Corporation Tricyclic urea compounds as JAK2 V617F inhibitors
US11661422B2 (en) 2020-08-27 2023-05-30 Incyte Corporation Tricyclic urea compounds as JAK2 V617F inhibitors
CN114149428B (zh) * 2020-09-08 2023-06-13 中国药科大学 吡啶并环类化合物及其制备方法、中间体、组合物和应用
CN114149428A (zh) * 2020-09-08 2022-03-08 中国药科大学 吡啶并环类化合物及其制备方法、中间体、组合物和应用
WO2022074572A1 (fr) * 2020-10-08 2022-04-14 Unichem Laboratories Ltd Composés tricycliques substitués
US11919908B2 (en) 2020-12-21 2024-03-05 Incyte Corporation Substituted pyrrolo[2,3-d]pyrimidine compounds as JAK2 V617F inhibitors
WO2022182839A1 (fr) * 2021-02-25 2022-09-01 Incyte Corporation Lactames spirocycliques utilisés comme inhibiteurs du v617f de jak2
US11958861B2 (en) 2021-02-25 2024-04-16 Incyte Corporation Spirocyclic lactams as JAK2 V617F inhibitors
CN115385911A (zh) * 2021-05-22 2022-11-25 南京康屹生物医药中心(有限合伙) 一种二吡咯并吡啶结构的化合物、制备方法和医药用途
WO2022247885A1 (fr) * 2021-05-26 2022-12-01 正大天晴药业集团股份有限公司 Cristal et sel de composé trihétérocyclique et leur utilisation
WO2023093718A1 (fr) * 2021-11-25 2023-06-01 启元生物(杭州)有限公司 Nouvelle forme cristalline du composé, son procédé de préparation et son utilisation
WO2023115327A1 (fr) * 2021-12-21 2023-06-29 中国药科大学 Composé cyclique de pyrido, son procédé de préparation, intermédiaire, composition et application
WO2024041586A1 (fr) * 2022-08-25 2024-02-29 启元生物(杭州)有限公司 Promédicament d'inhibiteur de kinase jak

Similar Documents

Publication Publication Date Title
WO2013007765A1 (fr) Composés tricycliques fusionnés utilisés en tant qu'inhibiteurs des janus kinases
US8461328B2 (en) Tricyclic heterocyclic compounds, compositions and methods of use thereof
WO2013007768A1 (fr) Composés hétérocycliques tricycliques, compositions et procédés d'utilisation de ces composés comme inhibiteurs des jak
US9309240B2 (en) Pyrazolopyridine compounds, compositions and methods of use
WO2012085176A1 (fr) Composés pyrazinones tricycliques, leurs compositions et leurs procédés d'utilisation en tant qu'inhibiteurs de janus kinase
JP5822934B2 (ja) アザベンゾチアゾール化合物、組成物及び使用方法
EP2356117B1 (fr) Composés inhibiteurs de pi3k de type pyrazolopyridine et leurs procédés d'utilisation
MX2012010265A (es) Compuestos de imidazopiridina, composiciones y metodos de uso.
EP2989106A1 (fr) Composés hétérocycliques fusionnés en tant qu'inhibiteurs de protéine kinase
MX2011004494A (es) Compuestos de pirazolopirimidina inhibidores de jak y metodos.
EP2742040A1 (fr) Composés d'indazole, compositions et procédés d'utilisation
WO2009155565A1 (fr) Composés triazolopyridine inhibiteurs de jak kinase et procédés
US20140206702A1 (en) Imidazopyridine compounds, compositions and methods of use
US20160326142A1 (en) Pyrazole carboxamide compounds, compositions and methods of use
WO2016001341A1 (fr) Composés sulfonylaminopyridine, compositions et procédés d'utilisation associés
WO2016091916A1 (fr) Pyrazolylaminopurines en tant qu'inhibiteurs de la tyrosyne kinase (itk)
NZ624021B2 (en) Heteroaryl pyridone and aza-pyridone compounds as inhibitors of btk activity

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: 12746296

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: 12746296

Country of ref document: EP

Kind code of ref document: A1