US20100048557A1 - Triazolopyridine JAK Inhibitor Compounds and Methods - Google Patents

Triazolopyridine JAK Inhibitor Compounds and Methods Download PDF

Info

Publication number
US20100048557A1
US20100048557A1 US12/488,312 US48831209A US2010048557A1 US 20100048557 A1 US20100048557 A1 US 20100048557A1 US 48831209 A US48831209 A US 48831209A US 2010048557 A1 US2010048557 A1 US 2010048557A1
Authority
US
United States
Prior art keywords
alkyl
optionally substituted
compound
heterocyclyl
phenyl
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US12/488,312
Other languages
English (en)
Inventor
Bing-Yan Zhu
Michael Siu
Steven R. Magnuson
Richard Pastor
He HAIYING
Xiao YISONG
Zheng JIFU
Xu XING
Zhao JUNPING
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Genentech Inc
Original Assignee
Genentech Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Genentech Inc filed Critical Genentech Inc
Priority to US12/488,312 priority Critical patent/US20100048557A1/en
Assigned to GENENTECH, INC. reassignment GENENTECH, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: PASTOR, RICHARD, HAIYING, HE, JIFU, ZHENG, JUNPING, ZHAO, XING, XU, YISONG, XIAO, ZHU, BING-YAN, MAGNUSON, STEVEN R., SIU, MICHAEL
Publication of US20100048557A1 publication Critical patent/US20100048557A1/en
Priority to US13/471,269 priority patent/US8609687B2/en
Priority to US14/049,458 priority patent/US9434732B2/en
Priority to US15/220,087 priority patent/US20160333011A1/en
Abandoned legal-status Critical Current

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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/437Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene or sparfloxacin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/04Antipruritics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems

Definitions

  • Triazolopyridine compounds of Formula I which are inhibitors of Janus kinases, for example JAK2 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.
  • Phen MPD Myeloproliferative disorders originate in hematopoietic stem cells and primarily manifest in elevated counts of mostly normal cells of the myeloid lineage.
  • Ph+ MPD results in chronic myelogenous leukemia and is driven by a bcr-abl fusion protein that drives hematopoietic cell proliferation.
  • Ph ⁇ MPD can be further subclassified into three distinct disorders by related varieties, namely polycythemia vera (PV), essential thrombocythemia (ET) and idiopathic myelofibrosis (IMF).
  • PV polycythemia vera
  • ET essential thrombocythemia
  • IMF idiopathic myelofibrosis
  • JAK2 tyrosine kinase JAK2
  • V617F mature human JAK2 protein
  • a T875N mutation was associated with megakaryoblastic leukemia.
  • JAK2 fusion proteins have been identified in acute leukemias.
  • JAK2 functions to activate JAK2, which leads to MPD.
  • JAK2 is linked to cytokine receptors (i.e. EPO-R, TPO-R and others) and only gets activated if the receptor itself is activated by stimulation with the cognate cytokine ligand. Hematopoiesis as a whole is then regulated through the availability of ligands.
  • EPO cytokine erythropoietin
  • a mutation that uncouples JAK2 activation from EPO therefore, leads to elevated levels of red blood cells.
  • thrombopoietin (TPO) regulates platelet growth by binding to the TPO-R, which in turn also signals through JAK2.
  • TPO thrombopoietin
  • JAK2 JAK2 driven myeloproliferative disorders
  • diseases include both diseases in which JAK2 is activated by mutation or amplification, as well as, diseases in which JAK2 activation is a part of the oncogenic cascade.
  • Numerous tumor cell lines and tumor samples have high levels of phospho-STAT3, which is a JAK2 target gene.
  • 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.
  • 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.
  • JAK1 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 JAK1 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).
  • JAK3 associates exclusively with the gamma common cytokine receptor chain, which is present in the IL-2, IL-4, IL-7, IL-9, IL-15 and IL-21 cytokine receptor complexes. JAK3 is critical for lymphoid cell development and proliferation and mutations in JAK3 result in severe combined immunodeficiency (SCID) (O'Shea et al., 2002, Cell, 109 (suppl.): S121-S131).
  • SCID severe combined immunodeficiency
  • JAK3 and JAK3-mediated pathways have been targeted for immunosuppressive indications (e.g., transplantation rejection and rheumatoid arthritis) (Baslund et al., 2005, Arthritis & Rheumatism 52:2686-2692; Changelian et al., 2003, Science 302: 875-878).
  • immunosuppressive indications e.g., transplantation rejection and rheumatoid arthritis
  • TYK2 associates with the type I interferon (e.g., IFNalpha), IL-6, IL-10, IL-12 and IL-23 cytokine receptor complexes (Kisseleva et al., 2002, gene 285:1-24; Watford, W. T. & O'Shea, J. J., 2006, Immunity 25:695-697). Consistent with this, primary cells derived from a TYK2 deficient human are defective in type I interferon, IL-6, IL-10, IL-12 and IL-23 signaling.
  • a fully human monoclonal antibody targeting the shared p40 subunit of the IL-12 and 11-23 cytokines was recently approved by the European Commission for the treatment of moderate-to-severe plaque psoriasis (Krueger et al., 2007, N. Engl. J. Med. 356:580-92; Reich et al., 2009, Nat. Rev. Drug Discov. 8:355-356).
  • an antibody targeting the IL-12 and IL-23 pathways underwent clinical trials for treating Crohn's Disease (Mannon et al., 2004, N. Engl. J. Med. 351:2069-79).
  • R 1 , R 2 , R 3 , R 4 and R 5 are defined herein.
  • Another embodiment includes a pharmaceutical composition that includes a compound of Formula I and a pharmaceutically acceptable carrier, adjuvant or vehicle.
  • Another embodiment includes a method of treating or lessening the severity of a disease or condition responsive to the inhibition of one or more Janus kinase activity, selected from JAK1, JAK2, JAK3 and TYK2, in a patient.
  • the method includes administering to the patient a therapeutically effective amount of a compound of Formula I.
  • Another embodiment includes a method of treating or lessening the severity of a disease or condition responsive to the inhibition of JAK2 kinase activity in a patient.
  • the method includes administering to the patient a therapeutically effective amount of a compound of Formula I.
  • kits for treating a disease or disorder responsive to the inhibition of a JAK kinase includes a first pharmaceutical composition comprising a compound of Formula I and instructions for use
  • 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 (C 1 -C 18 ).
  • the alkyl radical is C 0 -C 6 , C 0 -C 5 , C 0 -C 3 , C 1 -C 12 , C 1 -C 10 , C 1 -C 8 , C 1 -C 6 , C 1 -C 5 , C 1 -C 4 , or C 1 -C 3 .
  • alkyl groups include C 1 -C 8 hydrocarbon moieties such as 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-1-propyl (1-Bu, i-butyl, —CH 2 CH(CH 3 ) 2 ), 2-butyl (s-Bu, s-butyl, —CH(CH 3 )CH 2 CH 3 ), 2-methyl-2-propyl (t-Bu, t-butyl, —C(CH 3 ) 3 ), 1-pentyl (n-pentyl, —CH 2 CH 2 CH 2 CH 3 ), 2-pentyl
  • alkenyl refers to linear or branched-chain monovalent hydrocarbon radical with at least one site of unsaturation, i.e., a carbon-carbon double bond, wherein the alkenyl radical may be optionally substituted independently with one or more substituents described herein, and includes radicals having “cis” and “trans” orientations, or alternatively, “E” and “Z” orientations.
  • the alkenyl radical is two to eighteen carbon atoms (C 2 -C 18 ).
  • the alkenyl radical is C 2 -C 12 , C 2 -C 10 , C 2 -C 8 , C 2 -C 6 or C 2 -C 3 .
  • Examples include, but are not limited to, ethenyl or vinyl (—CH ⁇ CH 2 ), prop-1-enyl (—CH ⁇ CHCH 3 ), prop-2-enyl (—CH 2 CH ⁇ CH 2 ), 2-methylprop-1-enyl, but-1-enyl, but-2-enyl, but-3-enyl, buta-1,3-dienyl, 2-methylbuta-1,3-diene, hex-1-enyl, hex-2-enyl, hex-3-enyl, hex-4-enyl, hexa-1,3-dienyl.
  • 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 -C 8 , C 2 -C 6 or C 2 -C 3 .
  • Examples include, but are not limited to, ethynyl (—C ⁇ CH), prop-1-ynyl (—C ⁇ CCH 3 ), prop-2-ynyl (propargyl, —CH 2 C ⁇ CH), but-1-ynyl, but-2-ynyl and but-3-ynyl.
  • 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 -C 10 or C 5 -C 10 .
  • the cycloalkyl group, as a monocycle is C 3 -C 6 or C 5 -C 6 .
  • the cycloalkyl group, as a bicycle is C 7 -C 12 .
  • Examples of monocyclic cycloalkyl include cyclopropyl, cyclobutyl, cyclopentyl, 1-cyclopent-1-enyl, 1-cyclopent-2-enyl, 1-cyclopent-3-enyl, cyclohexyl, 1-cyclohex-1-enyl, 1-cyclohex-2-enyl, 1-cyclohex-3-enyl, cyclohexadienyl, cycloheptyl, 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.
  • Aryl refers to a cyclic aromatic hydrocarbon group optionally substituted independently with one or more substituents described herein.
  • the aryl group is 6-20 carbon atoms (C 6 -C 20 ).
  • the aryl group is C 6 -C 9 .
  • the aryl group is a C 6 aryl group.
  • Aryl groups may be represented in the exemplary structures as “Ar”.
  • Aryl includes a bicyclic group comprising an aromatic ring with a fused non-aromatic or partially saturated ring.
  • Example aryl groups include, but are not limited to, phenyl, naphthalenyl, anthracenyl, indenyl, indanyl, 1,2-dihydronapthalenyl, 1,2,3,4-tetrahydronapthyl, and the like.
  • Halo refers to F, Cl, Br or I.
  • Heterocyclyl refers to a saturated or a partially unsaturated (i.e., having one or more double and/or triple bonds within the ring) cyclic group in which at least one ring atom is a heteroatom independently selected from nitrogen, oxygen, phosphorus and sulfur, the remaining ring atoms being carbon.
  • the heterocyclyl group may be optionally substituted with one or more substituents described below.
  • heterocyclyl includes monocycles or bicycles having 1 to 9 carbon ring members (C 1 -C 9 ).
  • heterocyclyl includes monocycles or bicycles having C 1 -C 5 , C 3 -C 5 or C 4 -C 5 .
  • heterocyclyl includes bridged ring systems having [2.2.1], [2.2.2], [3.2.2] and [4.1.0] arrangements, and having 1 to 3 heteroatoms selected from N, O, S and P.
  • heterocyclyl includes spiro groups having 1 to 3 heteroatoms selected from N, O, S and P.
  • the heterocyclyl group may be a carbon-linked group or heteroatom-linked group.
  • “Heterocyclyl” includes a heterocyclyl group fused to a cycloalkyl group.
  • heterocyclyl groups include, but are not limited to, oxiranyl, aziridinyl, thiiranyl, azetidinyl, oxetanyl, thietanyl, 1,2-dithietanyl, 1,3-dithietanyl, pyrrolidinyl, piperidinyl, morpholinyl, thiomorpholinyl, thioxanyl, piperazinyl, homopiperazinyl, homopiperidinyl, oxepanyl, thiepanyl, oxazepinyl, diazepinyl, thiazepinyl, dihydrothienyl, dihydropyranyl, dihydrofuranyl, tetrahydrofuranyl, tetrahydrothienyl, tetrahydropyranyl, tetrahydrothiopyranyl, 1-pyrrolinyl, 2-pyrrolinyl, 3-pyrrol
  • heterocyclyl groups wherein a ring atom is substituted with oxo ( ⁇ O) are indolinonyl, pyrimidinonyl and 1,1-dioxo-thiomorpholinyl.
  • the heterocyclyl groups herein are optionally substituted independently with one or more substituents described herein.
  • Heterocycles are described in Paquette, Leo A.; “Principles of Modern Heterocyclic Chemistry” (W. A. Benjamin, New York, 1968), particularly Chapters 1, 3, 4, 6, 7, and 9; “The Chemistry of Heterocyclic Compounds, A series of Monographs” (John Wiley & Sons, New York, 1950 to present), in particular Volumes 13, 14, 16, 19, and 28; and J. Am. Chem. Soc. (1960) 82:5566.
  • heteroaryl refers to an aromatic carbocyclic radical in which at least one ring atom is a heteroatom independently selected from nitrogen, oxygen and sulfur, the remaining ring atoms being carbon. Heteroaryl groups may be optionally substituted with one or more substituents described herein. In one example, the heteroaryl group contains 1 to 9 carbon ring atoms (C 1 -C 9 ). In other examples, the heteroaryl group is C 1 -C 5 , C 3 -C 5 or C 5 -C 10 . In one embodiment, exemplary heteroaryl groups include monocyclic aromatic 5-, 6- and 7-membered rings containing one or more heteroatoms independently selected from nitrogen, oxygen, and sulfur.
  • exemplary heteroaryl groups include fused ring systems of 8 to 20 atoms wherein at least one aromatic ring contains one or more heteroatoms independently selected from nitrogen, oxygen, and sulfur.
  • “Heteroaryl” includes heteroaryl groups fused with an aryl, cycloalkyl or heterocyclyl group.
  • heteroaryl groups include, but are not limited to, pyridinyl, imidazolyl, imidazopyridinyl, pyrimidinyl, pyrazolyl, triazolyl, pyrazinyl, tetrazolyl, furyl, thienyl, isoxazolyl, thiazolyl, oxazolyl, isothiazolyl, pyrrolyl, quinolinyl, isoquinolinyl, indolyl, benzimidazolyl, benzofuranyl, cinnolinyl, indazolyl, indolizinyl, phthalazinyl, pyridazinyl, triazinyl, isoindolyl, pteridinyl, purinyl, oxadiazolyl, triazolyl, thiadiazolyl, thiadiazolyl, furazanyl, benzofurazanyl, benzothiophen
  • the heterocyclyl or heteroaryl group is C-attached.
  • carbon bonded heterocyclyls include bonding arrangements at position 2, 3, 4, 5, or 6 of a pyridine, position 3, 4, 5, or 6 of a pyridazine, position 2, 4, 5, or 6 of a pyrimidine, position 2, 3, 5, or 6 of a pyrazine, position 2, 3, 4, or 5 of a furan, tetrahydrofuran, thiofuran, thiophene, pyrrole or tetrahydropyrrole, position 2, 4, or 5 of an oxazole, imidazole or thiazole, position 3, 4, or 5 of an isoxazole, pyrazole, or isothiazole, position 2 or 3 of an aziridine, position 2, 3, or 4 of an azetidine, position 2, 3, 4, 5, 6, 7, or 8 of a quinoline or position 1, 3, 4, 5, 6, 7, or 8 of an isoquinoline.
  • the heterocyclyl or heteroaryl 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, 1H-indazole, position 2 of a isoindole, or isoindoline, position 4 of a morpholine, and position 9 of a carbazole, or ⁇ -carboline.
  • alkyl, alkenyl and alkynyl are optionally substituted by 1 to 5 substituents independently selected from by one or more OR a , NR c R d , oxo and halo, and aryl, heterocyclyl, heteroaryl and cycloalkyl are optionally substituted by 1 to 5 substituents independently selected from OR a , oxo, halo, CF 3 , NR c R d , C 1 -C 4 alkyl and C(O)(C 1 -C 4 alkyl), wherein R a , R c and R d are defined below for Formula I.
  • alkyl, alkenyl and alkynyl are optionally substituted by 1 to 3 substituents independently selected from by one or more OR a , NR c R d , oxo and halo, and aryl, heterocyclyl, heteroaryl and cycloalkyl are optionally substituted by 1 to 3 substituents independently selected from OR a , oxo, halo, CF 3 , NR c R d , C 1 -C 4 alkyl and C(O)(C 1 -C 4 alkyl), wherein R a , R c and R d are defined below for Formula I.
  • alkyl, alkenyl and alkynyl are optionally substituted by 1 to 5 substituents independently selected from oxo and halo
  • aryl is optionally substituted by 1 to 5 substituents independently selected from OR a , halo, CF 3 , NR c R d and C 1 -C 4 alkyl, wherein R a , R c and R d are defined below for Formula I.
  • alkyl, alkenyl and alkynyl are optionally substituted by 1 to 3 substituents independently selected from oxo and halo
  • aryl is optionally substituted by 1 to 3 substituents independently selected from OR a , halo, CF 3 , NR c R d and C 1 -C 4 alkyl, wherein R a , R c and R d are defined below for Formula I.
  • alkyl, aryl and cycloalkyl are optionally substituted by 1 to 5 substituents independently selected from C 1 -C 4 alkyl, (C 0 -C 3 alkyl)OR c , oxo, halo, NR c R d and C 4 -C 5 heterocyclyl, wherein R c and R d are defined below for Formula I.
  • alkyl, aryl and cycloalkyl are optionally substituted by 1 to 3 substituents independently selected from C 1 -C 4 alkyl, (C 0 -C 3 alkyl)OR c , oxo, halo, NR c R d and C 4 -C 5 heterocyclyl, wherein R c and R d are defined below for Formula I.
  • alkyl, cycloalkyl and phenyl are optionally substituted by 1 to 5 substituents independently selected from halo, CH 3 OH, NH 2 , C(O)O(C 1 -C 6 alkyl) and C(O)NH(C 1 -C 6 alkyl).
  • alkyl, cycloalkyl and phenyl are optionally substituted by 1 to 3 substituents independently selected from halo, CH 3 OH, NH 2 , C(O)O(C 1 -C 6 alkyl) and C(O)NH(C 1 -C 6 alkyl).
  • Treating” and “treatment” includes both therapeutic treatment and prophylactic or preventative measures, wherein the object is to prevent or slow down (lessen) an undesired physiological change or disorder, such as the development or spread of cancer.
  • beneficial or desired clinical results include, but are not limited to, alleviation of symptoms, diminishment of extent of disease, stabilized (i.e., not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, and remission (whether partial or total), whether detectable or undetectable.
  • Treatment can also mean prolonging survival as compared to expected survival if not receiving treatment.
  • 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.
  • terapéuticaally 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).
  • bioavailability refers to the systemic availability (i.e., blood/plasma levels) of a given amount of drug administered to a patient. Bioavailability is an absolute term that indicates measurement of both the time (rate) and total amount (extent) of drug that reaches the general circulation from an administered dosage form.
  • cancer and “cancerous” 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, but are not limited to, 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, as well as head and neck cancer.
  • NSCLC non-small cell lung cancer
  • adenocarcinoma of the lung and squamous carcinoma of the lung cancer of the peritoneum, hepatocellular cancer, gastric or stomach cancer including gastrointestinal cancer
  • a “chemotherapeutic agent” is a chemical compound useful in the treatment of cancer.
  • chemotherapeutic agents include Erlotinib (TARCEVA®, Genentech, Inc./OSI Pharm.), Trastuzumab (HERCEPTIN®, Genentech, Inc.); bevacizumab (AVASTIN®, Genentech, Inc.); Rituximab (RITUXAN®, Genentech, Inc./Biogen Idec, Inc.), Bortezomib (VELCADE®, Millennium Pharm.), Fulvestrant (FASLODEX®, AstraZeneca), Sutent (SU11248, Pfizer), Letrozole (FEMARA®, Novartis), Imatinib mesylate (GLEEVEC®, Novartis), PTK787/ZK 222584 (Novartis), Oxaliplatin (Eloxatin®, Sanofi), 5-FU.
  • dynemicin including dynemicin A; bisphosphonates, such as clodronate; an esperamicin; as well as neocarzinostatin chromophore and related chromoprotein enediyne antibiotic chromophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, carabicin, caminomycin, carzinophilin, chromomycinis, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, ADRIAMYCIN® (doxorubicin), morpholino-doxorubicin, cyanomorpholino-doxorubicin, 2-pyrrolino-doxorubicin and deoxydoxorubicin), epirubicin,
  • chemotherapeutic agent include: (i) anti-hormonal agents that act to regulate or inhibit hormone action on tumors such as anti-estrogens and selective estrogen receptor modulators (SERMs), including, for example, tamoxifen (including NOLVADEX®; tamoxifen citrate), raloxifene, droloxifene, 4-hydroxytamoxifen, trioxifene, keoxifene, LY117018, onapristone, and FARESTON® (toremifine citrate); (ii) aromatase inhibitors that inhibit the enzyme aromatase, which regulates estrogen production in the adrenal glands, such as, for example, 4(5)-imidazoles, aminoglutethimide, MEGASE® (megestrol acetate), AROMASIN® (exemestane; Pfizer), formestanie, fadrozole, RIVISOR® (vorozole), FEMARA® (letrozole),
  • SERMs
  • Humanized monoclonal antibodies with therapeutic potential as agents in combination with the Janus kinase inhibitors of the invention include: alemtuzumab, apolizumab, aselizumab, atlizumab, bapineuzumab, bevacizumab, 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, motovizuma
  • prodrug refers to a precursor or derivative form of a pharmaceutically active substance that is less efficacious to the patient or cytotoxic to tumor cells compared to the parent drug and is capable of being enzymatically or hydrolytically activated or converted into the more active parent form. See, e.g., Wilman, “Prodrugs in Cancer Chemotherapy” Biochemical Society Transactions, 14, pp. 375-382, 615th Meeting Harbor (1986) and Stella et al., “Prodrugs: A Chemical Approach to Targeted Drug Delivery,” Directed Drug Delivery , Borchardt et al., (ed.), pp. 247-267, Humana Press (1985).
  • the prodrugs of this invention include, but are not limited to, phosphate-containing prodrugs, thiophosphate-containing prodrugs, sulfate-containing prodrugs, peptide-containing prodrugs, D-amino acid-modified prodrugs, glycosylated prodrugs, ⁇ -lactam-containing prodrugs, optionally substituted phenoxyacetamide-containing prodrugs or optionally substituted phenylacetamide-containing prodrugs, 5-fluorocytosine and other 5-fluorouridine prodrugs which can be converted into the more active cytotoxic free drug.
  • cytotoxic drugs that can be derivatized into a prodrug form for use in this invention include, but are not limited to, those chemotherapeutic agents described above.
  • a “liposome” is a small vesicle composed of various types of lipids, phospholipids and/or surfactant which is useful for delivery of a drug (such as a compound of Formula I and, optionally, a chemotherapeutic agent) to a mammal.
  • a drug such as a compound of Formula I and, optionally, a chemotherapeutic agent
  • the components of the liposome are commonly arranged in a bilayer formation, similar to the lipid arrangement of biological membranes.
  • 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.
  • 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.
  • stereoisomers refers to compounds which have identical chemical constitution, but differ with regard to the arrangement of the atoms or groups in space.
  • 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, and reactivities. Mixtures of diastereomers may separate under high resolution analytical procedures such as electrophoresis and chromatography.
  • Enantiomers refer to two stereoisomers of a compound which are non-superimposable mirror images of one another.
  • d and l or (+) and ( ⁇ ) are employed to designate the sign of rotation of plane-polarized light by the compound, with ( ⁇ ) or l 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.
  • the terms “racemic mixture” and “racemate” refer to an equimolar mixture of two enantiomeric species, devoid of optical activity.
  • phrases “pharmaceutically acceptable salt,” as used herein, refers to pharmaceutically acceptable organic or inorganic salts of a compound of Formula I.
  • Exemplary salts include, but are not limited, to sulfate, citrate, acetate, oxalate, chloride, bromide, iodide, nitrate, bisulfate, phosphate, acid phosphate, isonicotinate, lactate, salicylate, acid citrate, tartrate, oleate, tannate, pantothenate, bitartrate, ascorbate, succinate, maleate, gentisinate, fumarate, gluconate, glucuronate, saccharate, formate, benzoate, glutamate, methanesulfonate, ethanesulfonate, benzenesulfonate, p-toluenesulfonate, and pamoate (i.e., 1,1′-methylene-bis-(2-hydroxy-3
  • a pharmaceutically acceptable salt may involve the inclusion of another molecule such as an acetate ion, a succinate ion or other counter ion.
  • the counter ion may be any organic or inorganic moiety that stabilizes the charge on the parent compound.
  • a pharmaceutically acceptable salt may have more than one charged atom in its structure. Instances where multiple charged atoms are part of the pharmaceutically acceptable salt can have multiple counter ions. Hence, a pharmaceutically acceptable salt can have one or more charged atoms and/or one or more counter ion.
  • a “solvate” refers to an association or complex of one or more solvent molecules and a compound of Formula I.
  • solvents that form solvates include, but are not limited to, water, isopropanol, ethanol, methanol, DMSO, ethyl acetate, acetic acid, and ethanolamine.
  • hydrate refers to the complex where the solvent molecule is water.
  • protecting group refers to a substituent that is commonly employed to block or protect a particular functionality while reacting other functional groups on the compound.
  • an “amino-protecting group” is a substituent attached to an amino group that blocks or protects the amino functionality in the compound. Suitable amino-protecting groups include acetyl, trifluoroacetyl, phthalimido, t-butoxycarbonyl (BOC), benzyloxycarbonyl (CBz) and 9-fluorenylmethylenoxycarbonyl (Fmoc).
  • a “hydroxy-protecting group” refers to a substituent of a hydroxy group that blocks or protects the hydroxy functionality.
  • Suitable hydroxy-protecting groups include acetyl, trialkylsilyl, dialkylphenylsilyl, benzoyl, benzyl, benzyloxymethyl, methyl, methoxymethyl, triarylmethyl, and tetrahydropyranyl.
  • a “carboxy-protecting group” refers to a substituent of the carboxy group that blocks or protects the carboxy functionality.
  • Common carboxy-protecting groups include —CH 2 CH 2 SO 2 Ph, cyanoethyl, 2-(trimethylsilyl)ethyl, 2-(trimethylsilyl)ethoxymethyl, 2-(p-toluenesulfonyl)ethyl, 2-(p-nitrophenylsulfenyl)ethyl, 2-(diphenylphosphino)-ethyl, nitroethyl and the like.
  • protecting groups and their use see T. W. Greene and P. Wuts, Protective Groups in Organic Synthesis, Third Ed., John Wiley & Sons, New York, 1999; and P. Kocienski, Protecting Groups, Third Ed., Verlag, 2003.
  • patient includes human patients and animal patients.
  • animal includes companion animals (e.g., dogs, cats and horses), food-source animals, zoo animals, marine animals, birds and other similar animal species.
  • phrases “pharmaceutically acceptable” indicates that the substance or composition must be compatible chemically and/or toxicologically, with the other ingredients comprising a formulation, and/or the mammal being treated therewith.
  • compound of this invention and “compounds of the present invention”, and “compounds of Formula I”, unless otherwise indicated, include compounds of Formula I and stereoisomers, tautomers, solvates, metabolites, salts (e.g., pharmaceutically acceptable salts) and prodrugs thereof.
  • structures depicted herein are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms.
  • compounds of Formula I wherein one or more hydrogen atoms are replaced deuterium or tritium, or one or more carbon atoms are replaced by a 13 C- or 14 C-enriched carbon are within the scope of this invention.
  • Inflammatory disorder can refer to any disease, disorder, or syndrome in which an excessive or unregulated inflammatory response leads to excessive inflammatory symptoms, host tissue damage, or loss of tissue function. “Inflammatory disorder” also refers to a pathological state mediated by influx of leukocytes and/or neutrophil chemotaxis.
  • Inflammation refers to a localized, protective response elicited by injury or destruction of tissues, which serves to destroy, dilute, or wall off (sequester) both the injurious agent and the injured tissue. Inflammation is notably associated with influx of leukocytes and/or neutrophil chemotaxis. Inflammation can result from infection with pathogenic organisms and viruses and from noninfectious means such as trauma or reperfusion following myocardial infarction or stroke, immune response to foreign antigen, and autoimmune responses. Accordingly, inflammatory disorders amenable to treatment with Formula I compounds encompass disorders associated with reactions of the specific defense system as well as with reactions of the nonspecific defense system.
  • Specific defense system refers to the component of the immune system that reacts to the presence of specific antigens. Examples of inflammation resulting from a response of the specific defense system include the classical response to foreign antigens, autoimmune diseases, and delayed type hypersensitivity response mediated by T-cells. Chronic inflammatory diseases, the rejection of solid transplanted tissue and organs, e.g., kidney and bone marrow transplants, and graft versus host disease (GVHD), are further examples of inflammatory reactions of the specific defense system.
  • GVHD graft versus host disease
  • nonspecific defense system refers to inflammatory disorders that are mediated by leukocytes that are incapable of immunological memory (e.g., granulocytes, and macrophages).
  • inflammation that result, at least in part, from a reaction of the nonspecific defense system include inflammation associated with conditions such as adult (acute) respiratory distress syndrome (ARDS) or multiple organ injury syndromes; reperfusion injury; acute glomerulonephritis; reactive arthritis; dermatoses with acute inflammatory components; acute purulent meningitis or other central nervous system inflammatory disorders such as stroke; thermal injury; inflammatory bowel disease; granulocyte transfusion associated syndromes; and cytokine-induced toxicity.
  • ARDS adult (acute) respiratory distress syndrome
  • multiple organ injury syndromes reperfusion injury
  • acute glomerulonephritis reactive arthritis
  • dermatoses with acute inflammatory components acute purulent meningitis or other central nervous system inflammatory disorders such as stroke; thermal injury; inflammatory bowel disease; granulocyte transfusion associated syndromes; and
  • Autoimmune disease refers to any group of disorders in which tissue injury is associated with humoral or cell-mediated responses to the body's own constituents.
  • Allergic disease refers to any symptoms, tissue damage, or loss of tissue function resulting from allergy.
  • Arthritic disease refers to any disease that is characterized by inflammatory lesions of the joints attributable to a variety of etiologies.
  • Dermatis refers to any of a large family of diseases of the skin that are characterized by inflammation of the skin attributable to a variety of etiologies.
  • Transplant rejection refers to any immune reaction directed against grafted tissue, such as organs or cells (e.g., bone marrow), characterized by a loss of function of the grafted and surrounding tissues, pain, swelling, leukocytosis, and thrombocytopenia.
  • the therapeutic methods of the present invention include methods for the treatment of disorders associated with inflammatory cell activation.
  • “Inflammatory cell activation” refers to the induction by a stimulus (including, but not limited to, cytokines, antigens or auto-antibodies) of a proliferative cellular response, the production of soluble mediators (including but not limited to cytokines, oxygen radicals, enzymes, prostanoids, or vasoactive amines), or cell surface expression of new or increased numbers of mediators (including, but not limited to, major histocompatability antigens or cell adhesion molecules) in inflammatory cells (including but not limited to monocytes, macrophages, T lymphocytes, B lymphocytes, granulocytes (i.e., polymorphonuclear leukocytes such as neutrophils, basophils, and eosinophils), mast cells, dendritic cells, Langerhans cells, and endothelial cells). It will be appreciated by persons skilled in the art that the activation of one or a combination of these phenotypes in these cells can contribute to the initiation
  • NSAID is an acronym for “non-steroidal anti-inflammatory drug” and is a therapeutic agent with analgesic, antipyretic (lowering an elevated body temperature and relieving pain without impairing consciousness) and, in higher doses, with anti-inflammatory effects (reducing inflammation).
  • non-steroidal is used to distinguish these drugs from steroids, which (among a broad range of other effects) have a similar eicosanoid-depressing, anti-inflammatory action.
  • analgesics NSAIDs are unusual in that they are non-narcotic. NSAIDs include aspirin, ibuprofen, and naproxen. NSAIDs are usually indicated for the treatment of acute or chronic conditions where pain and inflammation are present.
  • NSAIDs are generally indicated for the symptomatic relief of the following conditions: rheumatoid arthritis, osteoarthritis, inflammatory arthropathies (e.g. 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. Most NSAIDs act as non-selective inhibitors of the enzyme cyclooxygenase, inhibiting both the cyclooxygenase-1 (COX-1) and cyclooxygenase-2 (COX-2) isoenzymes.
  • COX-1 cyclooxygenase-1
  • COX-2 cyclooxygenase-2
  • Cyclooxygenase catalyzes the formation of prostaglandins and thromboxane from arachidonic acid (itself derived from the cellular phospholipid bilayer by phospholipase A 2 ).
  • Prostaglandins act (among other things) as messenger molecules in the process of inflammation.
  • COX-2 inhibitors include celecoxib, etoricoxib, lumiracoxib, parecoxib, rofecoxib, rofecoxib, and valdecoxib.
  • JAK kinase and “Janus kinase” refer to the JAK1, JAK2, JAK3 and TYK2 protein kinases.
  • a compound of Formula I and pharmaceutical formulations thereof, are provided that are useful in the treatment of diseases, conditions and/or disorders responsive to the inhibition of JAK kinases.
  • R 1 is H, C(O)OR a , phenyl, C 1 -C 9 heterocyclyl or C 1 -C 9 heteroaryl, wherein said phenyl and heteroaryl are optionally substituted by 1 to 5 R 6 ;
  • R 2 is phenyl, C 1 -C 9 heteroaryl or C 1 -C 9 heterocyclyl, wherein the phenyl, heteroaryl and heterocyclyl are optionally substituted by 1 to 5 R 7 ;
  • R 3 , R 4 and R 5 are independently H, CH 3 , CH 2 CH 3 , OCH 3 , CF 3 , F or Cl;
  • R 6 is independently H, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, (C 0 -C 6 alkyl)OR a , (C 0 -C 6 alkyl)NR a R b , halo, CN, CF 3 , S(O) 1-2 NR a R b , C(O)R a , NR a C(O)OR b , NR a S(O) 1-2 NR b , (C 0 -C 6 alkyl)C 1 -C 5 heteroaryl, (C 0 -C 6 alkyl)C 1 -C 5 heterocyclyl, (C 0 -C 6 alkyl)C 3 -C 6 cycloalkyl, (C 0 -C 6 alkyl)C 6 -C 9 aryl, (C 0 -C 6 alkyl)C(O)OR a ,
  • R 7 is independently H, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, (C 0 -C 6 alkyl)OR a , (C 0 -C 6 alkyl)NR a R b , (C 0 -C 6 alkyl)(C 6 -C 9 aryl), halo, C(O)NR a R b , NR a C(O)R b , SO 2 (C 1 -C 6 alkyl), SO 2 NR a R b , CN, CF 3 , CH 2 CF 3 , nitro, S(O)(C 1 -C 6 alkyl), S(O)NR a R b , NR a S(O) 1-2 R b , C(O)R a , C(O)OR a , (C 0 -C 6 alkyl)C 1 -C 5 heteroaryl, (C
  • R a and R b are independently H, OR c , C(O)O(C 1 -C 6 alkyl), C 1 -C 6 alkyl, C 6 aryl or C 3 -C 6 cycloalkyl, wherein said alkyl, aryl and cycloalkyl are optionally substituted by 1 to 5 substituents independently selected from C 1 -C 4 alkyl, (C 0 -C 3 alkyl)OR c , oxo, halo, NR c R d and C 4 -C 5 heterocyclyl; or
  • R a and R b together with the atom to which they are attached form a C 1 -C 5 heterocyclyl
  • R c and R d are independently H, C 1 -C 3 alkyl, C 3 -C 6 cycloalkyl or phenyl, wherein said alkyl, cycloalkyl and phenyl are optionally substituted by 1 to 5 substituents independently selected from halo, CH 3 OH or NH 2 , C(O)O(C 1 -C 6 alkyl) and C(O)NH(C 1 -C 6 alkyl).
  • R 1 is H, C(O)OR a , phenyl or C 1 -C 9 heteroaryl, wherein said phenyl and heteroaryl are optionally substituted by 1 to 6 R 6
  • R 2 is phenyl, C 1 -C 9 heteroaryl or C 1 -C 9 heterocyclyl, wherein the phenyl, heteroaryl and heterocyclyl are optionally substituted by 1 to 6 R 7 ;
  • R 3 , R 4 and R 5 are independently H, CH 3 , CH 2 CH 3 , CF 3 , F or Cl;
  • R 6 is independently H, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, (C 0 -C 6 alkyl)OR a , (C 0 -C 6 alkyl)NR a R b , halo, CN, C 1 -C 5 heteroaryl, C 1 -C 5 heterocyclyl, C 3 -C 6 cycloalkyl, C 6 -C 9 aryl, C(O)OR a , C(O)(C 0 -C 5 alkyl)NR a R b , C(O)(C 0 -C 5 alkyl)(C 1 -C 5 heterocyclyl), C(O)NR a (C 0 -C 5 alkyl)(C 1 -C 5 heterocyclyl), C(O)NR a (C 0 -C 5 alkyl)(C 1 -C 5 heterocyclyl), C(O)
  • R 7 is independently H, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, (C 0 -C 6 alkyl)OR a , (C 0 -C 6 alkyl)NR a R b , (C 0 -C 6 alkyl)(C 6 -C 9 aryl), halo, C(O)NR a R b , NR a C(O)R b , SO 2 (C 1 -C 6 alkyl), SO 2 NR a R b , CN, nitro, wherein said alkyl, alkenyl and alkynyl are optionally substituted by 1 to 5 substituents independently selected from oxo and halo, and said and said aryl is optionally substituted by 1 to 5 substituents independently selected from OR a , halo, CF 3 , NR c R d and C 1 -C 4 alkyl;
  • R a and R b are independently H, OR c , C(O)O(C 1 -C 6 alkyl), C 1 -C 6 alkyl, C 6 aryl or C 3 -C 6 cycloalkyl, wherein said alkyl, aryl and cycloalkyl are optionally substituted by 1 to 5 substituents independently selected from C 1 -C 4 alkyl, (C 0 -C 3 alkyl)OR c , oxo, halo, NR c R d and C 4 -C 5 heterocyclyl; or
  • R a and R b together with the atom to which they are attached form a C 1 -C 5 heterocyclyl
  • R c and R d are independently H, C 1 -C 3 alkyl, C 3 -C 6 cycloalkyl or phenyl, wherein said alkyl, cycloalkyl and phenyl are optionally substituted by 1 to 5 substituents independently selected from halo, CH 3 OH, NH 2 , C(O)O(C 1 -C 6 alkyl) and C(O)NH(C 1 -C 6 alkyl).
  • R 1 is phenyl or C 1 -C 9 heteroaryl, wherein said phenyl and heteroaryl are optionally substituted by 1 to 5 R 6 .
  • R 1 is phenyl optionally substituted by 1 to 5 R 6 .
  • R 1 is phenyl optionally substituted by 1 to 3 R 6 .
  • R 1 is phenyl optionally substituted by 2 R 6 .
  • R 1 is phenyl optionally substituted by 1 R 6 .
  • R 1 is C 1 -C 9 heteroaryl optionally substituted by 1 to 5 R 6 .
  • C 1 -C 9 heteroaryl is pyridinyl, imidazolyl, imidazopyridinyl, pyrimidinyl, pyrazolyl, triazolyl, pyrazinyl, tetrazolyl, furyl, thienyl, isoxazolyl, thiazolyl, oxazolyl, isothiazolyl, pyrrolyl, quinolinyl, isoquinolinyl, indolyl, benzimidazolyl, benzofuranyl, cinnolinyl, indazolyl, indolizinyl, phthalazinyl, pyridazinyl, triazinyl, isoindolyl, pteridinyl, purinyl, oxadiazolyl, triazolyl, thiadiazolyl, thiadiazolyl, furazanyl, benzofurazanyl, benzothiophen
  • R 1 is pyridinyl optionally substituted by 1 to 4 R 6 .
  • R 1 is pyridinyl optionally substituted by 2 R 6 .
  • R 1 is pyridinyl optionally substituted by 1 R 6 .
  • R 1 is H.
  • R 1 is C(O)OR a , wherein R a is independently H, OR c , C(O)O(C 1 -C 6 alkyl), C 1 -C 6 alkyl, C 6 aryl or C 3 -C 6 cycloalkyl, wherein said alkyl, aryl and cycloalkyl are optionally substituted by 1 to 5 substituents independently selected from C 1 -C 4 alkyl, (C 0 -C 3 alkyl)OR c , Oxo, halo, NR c R d and C 4 -C 5 heterocyclyl.
  • R 1 is phenyl or C 1 -C 9 heteroaryl, optionally substituted by 1 to 3 R 6 .
  • R 1 is phenyl, pyrazolyl, benzimidazolyl or pyridyl, optionally substituted by 1 to 3 R 6 .
  • R 1 is phenyl, pyrazolyl or pyridyl, optionally substituted by 1 to 3 R 6 , wherein R 6 is independently C 1 -C 6 alkyl, (C 0 -C 6 alkyl)OR a , (C 0 -C 6 alkyl)NR a R b , halo, CF 3 , C(O)OR a , C(O)(C 0 -C 5 alkyl)NR a R b , C(O)(C 0 -C 5 alkyl)(C 1 -C 5 heterocyclyl), C(O)NR a (C 0 -C 5 alkyl)(C 1 -C 5 heterocyclyl), C(O)NR a (C 0 -C 5 alkyl)(C 3 -C 6 cycloalkyl), C(O)NR a (C 0 -C 5 alkyl)(C 1 -C 5 hetero
  • R 1 is phenyl, pyrazolyl or pyridyl, optionally substituted by 1 R 6 , wherein R 6 is independently C(O)OR a , C(O)(C 0 -C 5 alkyl)NR a R b , C(O)(C 0 -C 5 alkyl)(C 1 -C 5 heterocyclyl), C(O)NR a (C 0 -C 5 alkyl)(C 1 -C 5 heterocyclyl), C(O)NR a (C 0 -C 5 alkyl)(C 3 -C 6 cycloalkyl), C(O)NR a (C 0 -C 5 alkyl)(C 1 -C 5 heteroaryl), C(O)NR a (C 1 -C 5 alkyl)NR a R b , C(O)NR a (C 0 -C 5 alkyl)(C 6 aryl), wherein said alkyl is
  • R 1 is phenyl or pyridinyl, optionally substituted by 1 to 3 R 6 , wherein R 6 is independently C 1 -C 3 alkyl, (C 0 -C 6 alkyl)OR a , (C 0 -C 6 alkyl)NR a R b , halo, CF 3 , S(O) 2 R a , S(O) 1-2 NR a R b , NR a S(O) 1-2 R b , (C 0 -C 5 alkyl)C 1 -C 5 heterocyclyl or C(O)OR a , wherein said alkyl is optionally substituted by 1 to 5 substituents independently selected from OR a , NR c R d , OXO, S(O), 2R a , S(O) 1-2 NR a R b and halo, and said heterocyclyl is optionally substituted by 1 to 3 substituents independently selected from (C
  • R 1 is phenyl or pyridinyl, optionally substituted by 1 R 6 , wherein R 6 is independently C(O)(C 0 -C 5 alkyl)NR a R b , C(O)NR a (C 0 -C 5 alkyl)(C 1 -C 5 heterocyclyl), C(O)NR a (C 0 -C 5 alkyl)(C 3 -C 6 cycloalkyl), C(O)NR a (C 0 -C 5 alkyl)(C 1 -C 5 heteroaryl), C(O)NR a (C 1 -C 5 alkyl)NR a R b , C(O)NR a (C 0 -C 5 alkyl)(C 6 aryl), wherein said aryl, heterocyclyl, heteroaryl and cycloalkyl are optionally substituted by 1 to 5 substituents independently selected from OR a , oxo, halo
  • R 1 is C 1 -C 5 heteroaryl, optionally substituted by 1 to 3 R 6 .
  • R 1 is pyrazolyl or thiazolyl, optionally substituted by 1 or 2 R 6 , wherein R 6 is C 1 -C 6 alkyl or (C 0 -C 6 alkyl)C 1 -C 5 heterocyclyl, wherein said alkyl is optionally substituted by 1 to 5 substituents independently selected from OR a , NR c R d , oxo and halo, and said heterocyclyl is optionally substituted by 1 to 5 substituents independently selected from OR a , oxo, halo, CF 3 , NR c R d , C 1 -C 4 alkyl, (C 0 -C 6 alkyl)C 1 -C 5 heterocyclyl and C(O)(C 1 -C 4 alkyl).
  • said heterocyclyl is pyrrolidinyl, tetrahydrofuranyl, tetrahydropyranyl, piperidinyl, piperazinyl or morpholinyl.
  • R 1 is selected from the following:
  • R 1 is C 8 -C 9 bicyclic heterocyclyl, optionally substituted by 1 to 5 R 6 .
  • R 1 is indolinyl or isoindolinyl, optionally substituted by 1 to 3 R 6 .
  • R 1 is 3,3-dimethylindolin-2-only or 3,3-dimethylisoindolin-1-onyl.
  • R 1 is C 1 -C 5 heterocyclyl, optionally substituted by 1 to 5 R 6 . In one example, R 1 is oxetanyl, optionally substituted by 1 to 3 R 6 .
  • R 6 is independently H, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, (C 0 -C 6 alkyl)OR a , (C 0 -C 6 alkyl)NR a R b , halo, CN, CF 3 , S(O) 1-2 NR a R b , C(O)R a , NR a C(O)OR b , NR a S(O) 1-2 NR b , (C 0 -C 6 alkyl)C 1 -C 5 heteroaryl, (C 0 -C 6 alkyl)C 1 -C 5 heterocyclyl, (C 0 -C 6 alkyl)C 3 -C 6 cycloalkyl, (C 0 -C 6 alkyl)C 6 -C 9 aryl, (C 0 -C 6 alkyl)C(O)OR
  • R 6 is independently C 1 -C 6 alkyl, (C 0 -C 6 alkyl)OR a , (C 0 -C 6 alkyl)NR a R b , halo, CN, C 1 -C 5 heteroaryl, C 4 -C 5 heterocyclyl, C 3 -C 6 cycloalkyl, C 6 aryl, C(O)OR a , C(O)(C 0 -C 5 alkyl)NR a R b , C(O)(C 0 -C 5 alkyl)(C 1 -C 5 heterocyclyl), C(O)NR a (C 0 -C 5 alkyl)(C 1 -C 5 heterocyclyl), C(O)NR a (C 0 -C 5 alkyl)(C 3 -C 6 cycloalkyl), C(O)NR a (C 0 -C 5 alkyl)(C 1 -C 5 heteroary
  • R 6 is C 4 -C 5 heterocyclyl optionally substituted by 1 to 5 substituents independently selected from OH, oxo, halo, CF 3 , NR c R d , C 1 -C 4 alkyl and C(O)(C 1 -C 4 alkyl).
  • heterocyclyl is pyrrolidinyl, tetrahydrofuranyl, tetrahydrothiophenyl, 1,1-dioxotetrahydrothiophenyl, piperidinyl, piperizinyl, tetrahydropyranyl, thianyl, morpholinyl, pyridizinyl or hexahydropyrimidinyl.
  • heterocyclyl is piperidinyl, piperizinyl or morpholinyl.
  • R 6 is (C 0 -C 6 alkyl)OR a or (C 0 -C 6 alkyl)NR a R b .
  • R 6 is (C 0 -C 3 alkyl)OR a or (C 0 -C 3 alkyl)NR a R b .
  • R 6 is halo
  • R 6 is F or Cl.
  • R 6 is S(O) 1-2 NR a R b . In one example, R 6 is S(O) 2 NH 2 .
  • R 6 is NR a C(O)R b . In one example, R 6 is NHCOCH 3 .
  • R 6 is C(O)NR a (C 0 -C 5 alkyl)(C 1 -C 5 heterocyclyl), C(O)NR a (C 0 -C 5 alkyl)(C 3 -C 6 cycloalkyl), C(O)NR a (C 0 -C 5 alkyl)(C 1 -C 5 heteroaryl), C(O)NR a (C 1 -C 5 alkyl)NR a R b , C(O)NR a (C 1 -C 5 alkyl)(C 6 aryl), wherein said alkyl is optionally substituted by 1 to 5 substituents independently selected from OR a , NR c R d , oxo and halo, and said aryl, heterocyclyl, heteroaryl and cycloalkyl are optionally substituted by 1 to 5 substituents independently selected from OR a , oxo, halo, CF 3 , NR c
  • R 6 is C(O)OR a , C(O)(C 0 -C 5 alkyl)NR a R b or C(O)(C 0 -C 5 alkyl)(C 1 -C 5 heterocyclyl), wherein said alkyl is optionally substituted by 1 to 5 substituents independently selected from OR a , NR c R d , oxo and halo, and said aryl, heterocyclyl, heteroaryl and cycloalkyl are optionally substituted by 1 to 5 substituents independently selected from OR a , oxo, halo, CF 3 , NR c R d , C 1 -C 4 alkyl and C(O)(C 1 -C 4 alkyl).
  • R 6 is independently C 1 -C 6 alkyl, (C 0 -C 6 alkyl)NR a R b , (C 0 -C 6 alkyl)C 1 -C 5 heterocyclyl, (C 0 -C 6 alkyl)C 3 -C 6 cycloalkyl, or C(O)(C 0 -C 5 alkyl)(C 1 -C 5 heterocyclyl), wherein said alkyl is optionally substituted by 1 to 5 substituents independently selected from OR a , NR c R d , oxo and halo, and said heterocyclyl and cycloalkyl are optionally substituted by 1 to 5 substituents independently selected from OR a , oxo, halo, CF 3 , NR c R d , C 1 -C 4 alkyl, (C 0 -C 6 alkyl)C 1 -C 5 heterocyclyl and C(O)(C 1
  • R 6 is (C 0 -C 6 alkyl)C 1 -C 5 heteroaryl, optionally substituted by 1 to 5 substituents independently selected from OR a , halo, CF 3 , NR c R d and C 1 -C 4 alkyl.
  • R 7 is pyridinyl, optionally substituted by 1 to 5 substituents independently selected from OR a , halo, CF 3 , NR c R d and C 1 -C 4 alkyl.
  • R 6 is selected from the following:
  • R 3 , R 4 and R 5 are independently H, CH 3 , CF 3 , or F.
  • R 3 , R 4 and R 5 are independently H or F.
  • R 3 , R 4 and R 5 are H.
  • R 3 is OCH 3 .
  • R 2 is phenyl, C 1 -C 9 heteroaryl or C 3 -C 5 heterocyclyl,
  • phenyl, heteroaryl and heterocyclyl are optionally substituted by 1 to 5 R 7 .
  • R 2 is phenyl optionally substituted by 1 to 5 R 7 .
  • R 2 is phenyl optionally substituted by 1 to 3 R 7 .
  • R 2 is phenyl optionally substituted by 2 R 7 .
  • R 2 is phenyl or pyridinyl, optionally substituted by 2 R 7 , wherein R 7 is independently C 1 -C 6 alkyl, OCF 3 , OCH 3 , NH 2 , NO 2 , CH 2 NH 2 , F, Cl, C(O)NR a R b , NR AC(O)R b , SO 2 (C 1 -C 3 alkyl), SO 2 NR a R b , CN, CF 3 , OCF 3 , C(O)R a , C(O)OR a , (C 0 -C 6 alkyl)C 1 -C 5 heterocyclyl, C(O)(C 0 -C 6 alkyl)C 1 -C 5 heterocyclyl or C(O)NR a (C 0 -C 6 alkyl)C 1 -C 5 heterocyclyl, wherein said heterocyclyl is optionally substituted by C 1 -C 4 alkyl
  • R 2 is pyrazolyl, optionally substituted by 1 or 2 R 7 ,
  • R 7 is independently R 7 is independently C 1 -C 6 alkyl, (C 0 -C 6 alkyl)OR a , (C 0 -C 6 alkyl)NR a R b , (C 0 -C 6 alkyl)(C 6 -C 9 aryl), halo, (C 0 -C 6 alkyl)C 1 -C 5 heteroaryl, (C 0 -C 6 alkyl)C 1 -C 5 heterocyclyl or (C 0 -C 6 alkyl)C 3 -C 6 cycloalkyl, wherein said alkyl, is optionally substituted by 1 to 5 substituents independently selected from oxo, NR a R b , OR a , and halo, and said aryl, heteroaryl, heterocyclyl and cycloalkyl are optionally substituted by 1 to 5 substituents independently selected from OR a , halo, CF 3 , NR c R
  • R 2 is phenyl optionally substituted by 1 R 7 .
  • R 7 is independently C 1 -C 6 alkyl, (C 0 -C 6 alkyl)OR a , (C 0 -C 6 alkyl)NR a R b , (C 0 -C 6 alkyl)(C 6 -C 9 aryl), halo, C(O)NR a R b , NR a C(O)R b , SO 2 (C 1 -C 6 alkyl), SO 2 NR a R b , CN, nitro, wherein said alkyl is optionally substituted by 1 to 5 substituents independently selected from oxo and halo, and said and said aryl is optionally substituted by 1 to 5 substituents independently selected from OR a , halo, CF 3 , NR c R d and C 1 -C 4 alkyl.
  • R 7 is independently C 1 -C 4 alkyl, (C 0 -C 6 alkyl)OR a , (C 0 -C 6 alkyl)NR a R b , halo, NR a C(O)R b , SO 2 (C 1 -C 6 alkyl), SO 2 NR a R b , CN or nitro.
  • R 7 is independently C 3 -C 6 cycloalkyl.
  • R 7 is independently NH 2 , OCH 3 , CH 3 , CH 2 CH 3 , CH(CH 3 ) 2 , NO 2 , OCF 3 , S(O) 2 N(CH 3 ) 2 , S(O) 2 NH(CH(CH 3 ) 2 ), S(O) 2 NH(C(CH 3 ) 3 ), CN, CF 3 , F, Cl, NHC(O)CH 3 or S(O) 2 CH 3 .
  • R 7 is independently NH 2 , OCH 3 , CH 3 , CH 2 CH 3 , CH(CH 3 ) 2 , NO 2 , OCF 3 , S(O) 2 N(CH 3 ) 2 , S(O) 2 NH(CH(CH 3 ) 2 ), S(O) 2 NH(C(CH 3 ) 3 ), CN, CF 3 , F, Cl, NHC(O)CH 3 , S(O) 2 CH 3 , CO 2 H, S(O)CH 3 , cyclopentyl, 1-hydroxyethyl, 1-aminoethyl or CH 2 CF 3 .
  • R 2 is C 1 -C 9 heteroaryl optionally substituted by 1 to 5 R 7 .
  • C 1 -C 9 heteroaryl is pyridinyl, imidazolyl, imidazopyridinyl, pyrimidinyl, pyrazolyl, triazolyl, pyrazinyl, tetrazolyl, furyl, thienyl, isoxazolyl, thiazolyl, oxazolyl, isothiazolyl, pyrrolyl, quinolinyl, isoquinolinyl, indolyl, benzimidazolyl, benzofuranyl, cinnolinyl, indazolyl, indolizinyl, phthalazinyl, pyridazinyl, triazinyl, isoindolyl, pteridinyl, purinyl, oxadiazolyl, triazolyl, thiadiazolyl, thiadiazolyl, furazanyl, benzofurazanyl, benzothiophen
  • R 2 is pyridinyl or pyrazolyl optionally substituted by 1 to 5 R 7 .
  • R 2 is pyridinyl or pyrazolyl optionally substituted by 1 to 3 R 7 .
  • R 2 is pyridinyl or pyrazolyl optionally substituted by 1 R 7 .
  • R 7 is independently CH 3 , CH 2 (phenyl), CH 2 CH(CH 3 ) 2 , or CF 3 .
  • R 2 is C 3 -C 5 heterocyclyl optionally substituted by 1 to 5 R 7 .
  • R 2 is piperidinyl, morpholinyl or piperizinyl optionally substituted by 1 to 5 R 7 .
  • R 2 is piperidinyl, morpholinyl or piperizinyl optionally substituted by 1 to 3 R 7 .
  • R 2 is piperidinyl, morpholinyl or piperizinyl optionally substituted by 2 R 7 .
  • R 2 is piperidinyl, morpholinyl or piperizinyl optionally substituted by 1 R 7 .
  • R 7 is independently CH 3 , CH 2 CH 3 , OH or OCH 3 .
  • R 1 is phenyl, optionally substituted by 1 to 5 R 6 ; and R 2 is phenyl, optionally substituted by 1 to 5 R 7 .
  • R 1 is phenyl, optionally substituted by 1 to 5 R 6 ;
  • R 2 is heterocyclyl, optionally substituted by 1 to 5 R 7 .
  • heterocyclyl is piperidinyl, morpholinyl or piperizinyl.
  • R 1 is pyridyl, optionally substituted by 1 to 4 R 6 ; and R 2 is phenyl, optionally substituted by 1 to 5 R 7 .
  • R 1 is pyridyl, optionally substituted by 1 to 4 R 6 ; and R 2 is heterocyclyl, optionally substituted by 1 to 5 R 7 .
  • heterocyclyl is piperidinyl, morpholinyl or piperizinyl.
  • R 1 is phenyl, optionally substituted by 1 to 5 R 6 ;
  • R 2 is pyridyl, optionally substituted by 1 to 4 R 7 .
  • R 1 is pyridyl, optionally substituted by 1 to 4 R 6 ; and R 2 is pyridyl, optionally substituted by 1 to 4 R 7 .
  • R 1 is phenyl or pyridinyl, optionally substituted by 1 to 3 R 6 , wherein R 6 is independently C 1 -C 3 alkyl, halo, CF 3 or C(O)OR a ; and R 2 is phenyl or pyridinyl, optionally substituted by 2 R 7 , wherein R 7 is independently C 1 -C 6 alkyl, OCF 3 , OCH 3 , NH 2 , CH 2 NH 2 , F, Cl, C(O)NR a R b , NR a C(O)R b , SO 2 (C 1 -C 3 alkyl), SO 2 NR a b, CN, CF 3 , OCF 3 , C(O)R a , C(O)OR a .
  • Another embodiment includes compounds of Formula I, selected from the compounds of Examples 1-312.
  • Another embodiment includes a compound of Formula I that has K i and/or EC 50 that is at least 15 fold, alternatively 10 fold, or 5 fold or more selective in inhibiting one Janus kinase activity over inhibiting each of the other Janus kinase activities.
  • the compounds of Formula I may contain asymmetric or chiral centers, and, therefore, exist in different stereoisomeric forms. It is intended that all stereoisomeric forms of the compounds of Formula I, including but not limited to: diastereomers, enantiomers, and atropisomers as well as mixtures thereof such as racemic mixtures, form part of the present invention.
  • the present invention embraces all geometric and positional isomers. For example, if a compound of Formula I incorporates a double bond or a fused ring, both the cis- and trans-forms, as well as mixtures, are embraced within the scope of the invention.
  • stereochemistry of any particular chiral atom is not specified, then all stereoisomers are contemplated and included as the compounds of the invention. Where stereochemistry is specified by a solid wedge or dashed line representing a particular configuration, then that stereoisomer is so specified and defined.
  • the compounds of the present invention may exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like, and it is intended that the invention, as defined by the claims, embrace both solvated and unsolvated forms.
  • 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.
  • the present invention also embraces isotopically-labeled compounds of Formula I, which are identical to those recited herein, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature. All isotopes of any particular atom or element as specified are contemplated within the scope of the invention.
  • Exemplary isotopes that can be incorporated into compounds of Formula I include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, chlorine, and iodine, such as 2 H, 3 H, 11 C, 13 C, 14 C, 13 N, 15 N, 15 O, 17 O, 18 O, 32 P, 33 P, 35 S, 18 F, 36 Cl, 123 I, and 125 I, respectively.
  • Certain isotopically-labeled compounds of Formula I e.g., those labeled with 3 H and 14 C are useful in compound and/or substrate tissue distribution assays.
  • Tritiated (i.e., 3 H) and carbon-14 (i.e., 14 C) isotopes are useful for their ease of preparation and detectability. Further, substitution with heavier isotopes such as deuterium (i.e., 2 H) may afford certain therapeutic advantages resulting from greater metabolic stability (e.g., increased in vivo half-life or reduced dosage requirements) and hence may be preferred in some circumstances.
  • Positron emitting isotopes such as 15 O, 13 N, 11 C, and 18 F are useful for positron emission tomography (PET) studies to examine substrate receptor occupancy.
  • Isotopically labeled compounds of Formula I can generally be prepared by following procedures analogous to those disclosed in the Schemes and/or in the Examples herein below, by substituting an isotopically labeled reagent for a non-isotopically labeled reagent.
  • Compounds of Formula I may be synthesized by synthetic routes described herein. In certain embodiments, processes well-known in the chemical arts can be used, in addition to, or in light of, the description contained herein.
  • the starting materials are generally available from commercial sources such as Aldrich Chemicals (Milwaukee, Wis.) or are readily prepared using methods well known to those skilled in the art (e.g., prepared by methods generally described in Louis F. Fieser and Mary Fieser, Reagents for Organic Synthesis , v. 1-19, Wiley, N.Y. (1967-1999 ed.), Beilsteins Handbuch der organischen Chemie, 4, Aufl. ed.
  • 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, diasteriomers, tautomers or pharmaceutically acceptable salts thereof.
  • reaction schemes 1-6 depicted below provide routes for synthesizing the compounds of the present invention as well as key intermediates. For a more detailed description of the individual reaction steps, see the Examples section below. Those skilled in the art will appreciate that other synthetic routes may be used to synthesize the inventive compounds. Although specific starting materials and reagents are depicted in the Schemes and discussed below, other starting materials and reagents can be easily substituted to provide a variety of derivatives and/or reaction conditions. In addition, many of the compounds prepared by the methods described below can be further modified in light of this disclosure using conventional chemistry well known to those skilled in the art.
  • Suitable amino-protecting groups include acetyl, trifluoroacetyl, t-butoxycarbonyl (BOC), benzyloxycarbonyl (CBz) and 9-fluorenylmethyleneoxycarbonyl (Fmoc).
  • NH-Pg amino-protecting groups
  • BOC t-butoxycarbonyl
  • CBz benzyloxycarbonyl
  • Fmoc 9-fluorenylmethyleneoxycarbonyl
  • compounds of Formula I can be synthesized as shown in Reaction Scheme 1.
  • a common protected intermediate 5, which is available from 2-amino-3-bromopyridine (2), can be subjected to palladium-catalyzed coupling reactions such as the Suzuki reaction with boronic acids or boronate esters to form compounds of Formula 6 after protective group removal.
  • Palladium-catalyzed amination of aryl or heteroaryl halides with triazolopyridinamine 6 provides compounds of Formula 1a.
  • Palladium-catalyzed amination of methoxycarbonyl substituted phenyl halides or methoxycarbonyl substituted heteroaryl halides 7 with triazolopyridinamine 6 provides compounds of Formula 1b.
  • Hydrolysis of the corresponding methyl esters 1b affords carboxylic acids 1c, which can be subjected to standard amide formation methods to yield amides 1d.
  • primary amine or secondary amine groups may be converted into amide groups (—NHCOR′ or —NRCOR′) by acylation.
  • Acylation may be achieved by reaction with an appropriate acid chloride in the presence of a base, such as triethylamine, in a suitable solvent, such as dichloromethane, or by reaction with an appropriate carboxylic acid in the presence of a suitable coupling agent such HATU (O-(7-azabenzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate) in a suitable solvent such as dichloromethane.
  • HATU O-(7-azabenzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate
  • amine groups may be converted into sulphonamide groups (—NHSO 2 R′ or —NR′′SO 2 R′) groups by reaction with an appropriate sulphonyl chloride in the presence of a suitable base, such as triethylamine, in a suitable solvent such as dichloromethane.
  • Primary or secondary amine groups can be converted into urea groups (—NHCONR′R′′ or —NRCONR′R′′) by reaction with an appropriate isocyanate in the presence of a suitable base such as triethylamine, in a suitable solvent, such as dichloromethane.
  • An amine (—NH 2 ) may be obtained by reduction of a nitro (—NO 2 ) group, for example by catalytic hydrogenation, using for example hydrogen in the presence of a metal catalyst, for example palladium on a support such as carbon in a solvent such as ethyl acetate or an alcohol e.g. methanol.
  • a metal catalyst for example palladium on a support such as carbon in a solvent such as ethyl acetate or an alcohol e.g. methanol.
  • the transformation may be carried out by chemical reduction using for example a metal, e.g. tin or iron, in the presence of an acid such as hydrochloric acid.
  • amine (—CH 2 NH 2 ) groups may be obtained by reduction of nitriles (—CN), for example by catalytic hydrogenation using for example hydrogen in the presence of a metal catalyst, for example palladium on a support such as carbon, or Raney nickel, in a solvent such as an ether e.g. a cyclic ether such as tetrahydrofuran, at an appropriate temperature, for example from about ⁇ 78° C. to the reflux temperature of the solvent.
  • a metal catalyst for example palladium on a support such as carbon, or Raney nickel
  • amine (—NH 2 ) groups may be obtained from carboxylic acid groups (—CO 2 H) by conversion to the corresponding acyl azide (—CON 3 ), Curtius rearrangement and hydrolysis of the resultant isocyanate (—N ⁇ C ⁇ O).
  • Aldehyde groups may be converted to amine groups (—CH 2 NR′R′′)) by reductive amination employing an amine and a borohydride, for example sodium triacetoxyborohydride or sodium cyanoborohydride, in a solvent such as a halogenated hydrocarbon, for example dichloromethane, or an alcohol such as ethanol, where necessary in the presence of an acid such as acetic acid at around ambient temperature.
  • a borohydride for example sodium triacetoxyborohydride or sodium cyanoborohydride
  • a solvent such as a halogenated hydrocarbon, for example dichloromethane, or an alcohol such as ethanol
  • aldehyde groups may be converted into alkenyl groups (—CH ⁇ CHR′) by the use of a Wittig or Wadsworth-Emmons reaction using an appropriate phosphorane or phosphonate under standard conditions known to those skilled in the art.
  • Aldehyde groups may be obtained by reduction of ester groups (such as —CO 2 Et) or nitriles (—CN) using diisobutylaluminium hydride in a suitable solvent such as toluene.
  • ester groups such as —CO 2 Et
  • —CN nitriles
  • aldehyde groups may be obtained by the oxidation of alcohol groups using any suitable oxidising agent known to those skilled in the art.
  • Ester groups (—CO 2 R′) may be converted into the corresponding acid group (—CO 2 H) by acid- or base-catalysed hydrolysis, depending on the nature of R. If R is t-butyl, acid-catalysed hydrolysis can be achieved for example by treatment with an organic acid such as trifluoroacetic acid in an aqueous solvent, or by treatment with an inorganic acid such as hydrochloric acid in an aqueous solvent.
  • Carboxylic acid groups may be converted into amides (CONHR′ or —CONR′R′′) by reaction with an appropriate amine in the presence of a suitable coupling agent, such as HATU, in a suitable solvent such as dichloromethane.
  • a suitable coupling agent such as HATU
  • carboxylic acids may be homologated by one carbon (i.e —CO 2 H to —CH 2 CO 2 H) by conversion to the corresponding acid chloride (—COCl) followed by Arndt-Eistert synthesis.
  • —OH groups may be generated from the corresponding ester (e.g. —CO 2 R′), or aldehyde (—CHO) by reduction, using for example a complex metal hydride such as lithium aluminium hydride in diethyl ether or tetrahydrofuran, or sodium borohydride in a solvent such as methanol.
  • a complex metal hydride such as lithium aluminium hydride in diethyl ether or tetrahydrofuran
  • sodium borohydride in a solvent such as methanol.
  • an alcohol may be prepared by reduction of the corresponding acid (—CO 2 H), using for example lithium aluminium hydride in a solvent such as tetrahydrofuran, or by using borane in a solvent such as tetrahydrofuran.
  • Alcohol groups may be converted into leaving groups, such as halogen atoms or sulfonyloxy groups such as an alkylsulfonyloxy, e.g. trifluoromethylsulfonyloxy or arylsulfonyloxy, e.g. p-toluenesulfonyloxy group using conditions known to those skilled in the art.
  • halogen atoms or sulfonyloxy groups such as an alkylsulfonyloxy, e.g. trifluoromethylsulfonyloxy or arylsulfonyloxy, e.g. p-toluenesulfonyloxy group using conditions known to those skilled in the art.
  • an alcohol may be reacted with thioyl chloride in a halogenated hydrocarbon (e.g. dichloromethane) to yield the corresponding chloride.
  • a base e.g. triethylamine
  • alcohol, phenol or amide groups may be alkylated by coupling a phenol or amide with an alcohol in a solvent such as tetrahydrofuran in the presence of a phosphine, e.g. triphenylphosphine and an activator such as diethyl-, diisopropyl, or dimethylazodicarboxylate.
  • a phosphine e.g. triphenylphosphine and an activator such as diethyl-, diisopropyl, or dimethylazodicarboxylate.
  • alkylation may be achieved by deprotonation using a suitable base e.g. sodium hydride followed by subsequent addition of an alkylating agent, such as an alkyl halide.
  • Aromatic halogen substituents in the compounds may be subjected to halogen-metal exchange by treatment with a base, for example a lithium base such as n-butyl or t-butyl lithium, optionally at a low temperature, e.g. around ⁇ 78° C., in a solvent such as tetrahydrofuran, and then quenched with an electrophile to introduce a desired substituent.
  • a base for example a lithium base such as n-butyl or t-butyl lithium
  • a solvent such as tetrahydrofuran
  • an electrophile to introduce a desired substituent.
  • a formyl group may be introduced by using N,N-dimethylformamide as the electrophile.
  • Aromatic halogen substituents may alternatively be subjected to metal (e.g.
  • Aromatic halogen substituents may also undergo nucleophilic displacement following reaction with an appropriate nucleophile such as an amine or an alcohol.
  • an appropriate nucleophile such as an amine or an alcohol.
  • such a reaction may be carried out at elevated temperature in the presence of microwave irradiation.
  • 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 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; 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.
  • 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
  • converting e.g., hydrolyzing
  • some of the compounds of the present invention may be atropisomers (e.g., substituted biaryls) and are considered as part of this invention.
  • 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, ⁇ -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 (Eliel, E. and Wilen, S., Stereochemistry of Organic Compounds , John Wiley & Sons, Inc., New York, 1994, p. 322).
  • Diastereomeric compounds can be formed by reacting asymmetric compounds with enantiomerically pure chiral derivatizing reagents, such as menthyl derivatives, followed by separation of the diastereomers and hydrolysis to yield the pure or enriched enantiomer.
  • a method of determining optical purity involves making chiral esters, such as a menthyl ester, e.g.
  • 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.
  • 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.
  • the catalytically active kinase domain of human JAK1, JAK2, JAK3 or TYK2 was purified from extracts of SF9 insect cells infected with a recombinant baculovirus expression vector encoding the human JAK1, JAK2, JAK3 or TYK2 kinase domains (JAK1 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 JAK1, 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 JAK1, JAK2, JAK3 or TYK2 kinase domains was measured in vitro by monitoring phosphorylation of JAK3 derived peptides using the Caliper LabChip technology (see Examples).
  • the compounds of the present invention are tested for their capacity to inhibit a Janus kinase activity and activation (primary assays) and for their biological effects on growing cells (secondary assays) as described herein.
  • Another embodiment includes a method of treating or lessening the severity of a disease or condition responsive to the inhibition of one or more Janus kinase activity, selected from JAK1, JAK2, JAK3 and TYK2, in a patient.
  • the method includes administering to the patient a therapeutically effective amount of a compound of Formula I.
  • Another embodiment includes a method of treating or lessening the severity of a disease or condition responsive to the inhibition of JAK2 kinase activity in a patient.
  • the method includes the step of administering to a patient a therapeutically effective amount of a compound of Formula I.
  • 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, 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).
  • CML chronic myelogenous leukemia
  • the cancer is breast, ovary, cervix, prostate, testis, penile, genitourinary tract, seminoma, esophagus, larynx, gastric, stomach, gastrointestinal, skin, keratoacanthoma, follicular carcinoma, melanoma, lung, small cell lung carcinoma, non-small cell lung carcinoma (NSCLC), lung adenocarcinoma, squamous carcinoma of the lung, colon, pancreas, thyroid, papillary, bladder, liver, biliary passage, kidney, bone, myeloid disorders, lymphoid disorders, hairy cells, buccal cavity and pharynx (oral), lip, tongue, mouth, salivary gland, pharynx, small intestine, colon, rectum, anal, renal, prostate, vulval, thyroid, large intestine, endometrial, uterine, brain, central nervous system, cancer of the peritoneum, hepatocellular cancer, head cancer, neck cancer,
  • 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, contact dermatitis or delayed hypersensitivity reactions.
  • the inflammatory disease is inflammatory bowel disease
  • the autoimmune disease is lupus or multiple sclerosis.
  • Evaluation of drug-induced immunosuppression by the compounds of the invention may be performed using in vivo functional tests, such as rodent models of induced arthritis and therapeutic or prophylactic treatment to assess disease score, T cell-dependent antibody response (TDAR), and delayed-type hypersensitivity (DTH).
  • TDAR T cell-dependent antibody response
  • DTH delayed-type hypersensitivity
  • Other in vivo systems including murine models of host defense against infections or tumor resistance (Burleson G R, Dean J H, and Munson A E. Methods in Immunotoxicology, Vol. 1. Wiley-Liss, New York, 1995) may be considered to elucidate the nature or mechanisms of observed immunosuppression.
  • the in vivo test systems can be complemented by well-established in vitro or ex vivo functional assays for the assessment of immune competence.
  • These assays may comprise B or T cell proliferation in response to mitogens or specific antigens, measurement of signaling through one or more of the Janus kinase pathways in B or T cells or immortalized B or T cell lines, measurement of cell surface markers in response to B or T cell signaling, natural killer (NK) cell activity, mast cell activity, mast cell degranulation, macrophage phagocytosis or kill activity, and neutrophil oxidative burst and/or chemotaxis.
  • NK natural killer
  • NK natural killer
  • mast cell activity mast cell activity
  • mast cell degranulation macrophage phagocytosis or kill activity
  • neutrophil oxidative burst and/or chemotaxis may be included.
  • the in vitro and ex vivo assays can be applied in both preclinical and clinical testing using lymphoid tissues and/or peripheral blood (House R V. “Theory and practice of cytokine assessment in immunotoxicology” (1999) Methods 19:17-27; Hubbard A K. “Effects of xenobiotics on macrophage function: evaluation in vitro” (1999) Methods; 19:8-16; Lebrec H, et al (2001) Toxicology 158:25-29).
  • Collagen-Induced Arthritis 6-week detailed study using an autoimmune mechanism to mimic human arthritis; rat and mouse models (Example 68).
  • Collagen-induced arthritis is one of the most commonly used animal models of human rheumatoid arthritis (RA). Joint inflammation, which develops in animals with CIA, strongly resembles inflammation observed in patients with RA. Blocking tumor necrosis factor (TNF) is an efficacious treatment of CIA, just as it is a highly efficacious therapy in treatment of RA patients.
  • CIA is mediated by both T-cells and antibodies (B-cells). Macrophages are believed to play an important role in mediating tissue damage during disease development.
  • CIA is induced by immunizing animals with collagen emulsified in Complete Freund's Adjuvant (CFA). It is most commonly induced in the DBA/1 mouse strain, but the disease can also be induced in Lewis rats.
  • CFA Complete Freund's Adjuvant
  • CD69 is the early activation marker in leukocytes including T cells, thymocytes, B cells, NK cells, neutrophils, and eosinophils.
  • the CD69 human whole blood assay determines the ability of compounds to inhibit the production of CD69 by B lymphocytes in human whole blood activated by crosslinking surface IgM with goat F(ab′)2 anti-human IgM.
  • the T-cell Dependent Antibody Response is a predictive assay for immune function testing when potential immunotoxic effects of compounds need to be studied.
  • TDAR has proven to be a highly predictable assay for adult exposure immunotoxicity detection in mice based on the US National Toxicology Program (NTP) database (M. I. Luster et al (1992) Fundam. Appl. Toxicol. 18:200-210).
  • NTP National Toxicology Program
  • a TDAR is dependent on functions of the following cellular compartments: (1) antigen-presenting cells, such as macrophages or dendritic cells; (2) T-helper cells, which are critical players in the genesis of the response, as well as in isotype switching; and (3) B-cells, which are the ultimate effector cells and are responsible for antibody production.
  • antigen-presenting cells such as macrophages or dendritic cells
  • T-helper cells which are critical players in the genesis of the response, as well as in isotype switching
  • B-cells which are the ultimate effector cells and are responsible for antibody production.
  • Chemically-induced changes in any one compartment can cause significant changes in the overall TDAR (M. P. Holsapple In: G. R. Burleson, J. H. Dean and A. E. Munson, Editors, Modern Methods in Immunotoxicology, Volume 1, Wiley-Liss Publishers, New York, N.Y. (1995), pp. 71-108).
  • this assay is performed either as an ELISA for measurement of soluble antibody (R. J. Smialowizc et al (2001) Toxicol. Sci. 61:164-175) or as a plaque (or antibody) forming cell assay (L. Guo et al (2002) Toxicol. Appl. Pharmacol. 181:219-227) to detect plasma cells secreting antigen specific antibodies.
  • the antigen of choice is either whole cells (e.g. sheep erythrocytes) or soluble protein antigens (T. Miller et al (1998) Toxicol. Sci. 42:129-135).
  • a compound of Formula I may be administered by any route appropriate to the disease or condition to be treated. Suitable routes include oral, parenteral (including subcutaneous, intramuscular, intravenous, intraarterial, intradermal, intrathecal and epidural), transdermal, rectal, nasal, topical (including buccal and sublingual), vaginal, intraperitoneal, intrapulmonary, and intranasal.
  • the compounds may be administered by intralesional administration, including perfusing or otherwise contacting the graft with the inhibitor before transplantation. It will be appreciated that the preferred route may vary with for example the condition of the recipient. Where the compound of Formula I is administered orally, it may be formulated as a pill, capsule, tablet, etc. with a pharmaceutically acceptable carrier or excipient. Where the compound of Formula I is administered parenterally, it may be formulated with a pharmaceutically acceptable parenteral vehicle and in a unit dosage injectable form, as detailed below.
  • a dose to treat human patients may range from about 10 mg to about 1000 mg of a compound of Formula I.
  • a typical dose may be about 100 mg to about 300 mg of a compound of Formula I.
  • a dose may be administered once a day (QD), twice per day (BID), or more frequently, depending on the pharmacokinetic and pharmacodynamic properties, including absorption, distribution, metabolism, and excretion of the particular compound.
  • QD once a day
  • BID twice per day
  • toxicity factors may influence the dosage and administration regimen.
  • the pill, capsule, or tablet When administered orally, may be ingested daily or less frequently for a specified period of time. The regimen may be repeated for a number of cycles of therapy.
  • Another embodiment of the invention aspect of this invention provides a compound of this invention for use as a medicament in the treatment of the diseases or conditions described herein in a mammal, for example, a human, suffering from such disease or condition. Also provided is the use of a compound of this invention in the preparation of a medicament for the treatment of the diseases and conditions described herein in a warm-blooded animal, such as a mammal, for example a human, suffering from such disorder.
  • Another embodiment includes a pharmaceutical composition that includes a compound of Formula I and a pharmaceutically acceptable carrier, adjuvant or vehicle.
  • the pharmaceutical composition also includes an additional therapeutic agent selected from an anti-proliferative agent, an anti-inflammatory agent, an immunomodulatory agent, a neurotropic factor, an agent for treating cardiovascular disease, an agent for treating liver disease, an anti-viral agent, an agent for treating blood disorders, an agent for treating diabetes, or an agent for treating immunodeficiency disorders.
  • an additional therapeutic agent selected from an anti-proliferative agent, an anti-inflammatory agent, an immunomodulatory agent, a neurotropic factor, an agent for treating cardiovascular disease, an agent for treating liver disease, an anti-viral agent, an agent for treating blood disorders, an agent for treating diabetes, or an agent for treating immunodeficiency disorders.
  • a compound of Formula I is present in a pharmaceutical formulation in an amount to detectably inhibit one or more of a Janus kinase activity, selected from JAK1, JAK2, JAK3 and TYK2, and a pharmaceutically acceptable carrier, adjuvant or vehicle.
  • a compound of Formula I is present in a pharmaceutical formulation in an amount to detectably inhibit JAK2 kinase activity and a pharmaceutically acceptable carrier, adjuvant or vehicle.
  • a compound of Formula I is present in a pharmaceutical formulation in an amount to detectably inhibit JAK2 kinase activity and is at least 10 fold or more selective in inhibiting JAK2 kinase activity over inhibiting each of JAK1, JAK3 and Tyk-2 activity.
  • a compound of Formula I is present in a pharmaceutical formulation in an amount to detectably inhibit one of a Janus kinase activity and is at least 15 fold, alternatively 10 fold, or 5 fold or more selective in inhibiting one such Janus kinase activity over inhibiting each of the other Janus kinase activity.
  • a compound of Formula I is present in a pharmaceutical formulation in an amount to detectably inhibit a Janus kinase activity and is at least 15 fold, alternatively 10 fold, or 5 fold or more selective in inhibiting one Janus kinase activity over inhibiting each of the other JAK1, JAK2, JAK3 and/or Tyk-2 activity.
  • a typical formulation is prepared by mixing a compound of the present invention and a carrier, diluent or excipient.
  • Suitable carriers, diluents and excipients are well known to those skilled in the art and include materials such as carbohydrates, waxes, water soluble and/or swellable polymers, hydrophilic or hydrophobic materials, gelatin, oils, solvents, water, and the like.
  • the particular carrier, diluent or excipient used will depend upon the means and purpose for which the compound of the present invention is being applied. Solvents are generally selected based on solvents recognized by persons skilled in the art as safe (GRAS) to be administered to a mammal.
  • GRAS solvents recognized by persons skilled in the art as safe
  • safe solvents are non-toxic aqueous solvents such as water and other non-toxic solvents that are soluble or miscible in water.
  • Suitable aqueous solvents include water, ethanol, propylene glycol, polyethylene glycols (e.g., PEG400, PEG300), etc. and mixtures thereof.
  • 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 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 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).
  • the formulations may be prepared using conventional dissolution and mixing procedures.
  • the bulk drug substance i.e., compound of the present invention or stabilized form of the compound, such as a complex with a cyclodextrin derivative or other known complexation agent
  • a suitable solvent in the presence of one or more of the excipients described above.
  • the compound of the present invention is typically formulated into pharmaceutical dosage forms to provide an easily controllable dosage of the drug and to enable patient compliance with the prescribed regimen.
  • the pharmaceutical composition (or formulation) for application may be packaged in a variety of ways depending upon the method used for administering the drug.
  • an article for distribution includes a container having deposited therein the pharmaceutical formulation in an appropriate form.
  • Suitable containers are well-known to those skilled in the art and include materials such as bottles (plastic and glass), sachets, ampoules, plastic bags, metal cylinders, and the like.
  • the container may also include a tamper-proof assemblage to prevent indiscreet access to the contents of the package.
  • the container has deposited thereon a label that describes the contents of the container. The label may also include appropriate warnings.
  • formulations of a compound of Formula I may be prepared for various routes and types of administration.
  • a compound of Formula I having the desired degree of purity is optionally mixed with pharmaceutically acceptable diluents, carriers, excipients or stabilizers (Remington's Pharmaceutical Sciences (1980) 16th edition, Osol, A. Ed.), in the form of a lyophilized formulation, milled powder, or an aqueous solution.
  • Formulation may be conducted 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.
  • the pH of the formulation depends mainly on the particular use and the concentration of compound, but may range from about 3 to about 8.
  • Formulation in an acetate buffer at pH 5 is a suitable embodiment.
  • the inhibitory compound for use herein is preferably sterile.
  • the compound ordinarily will be stored as a solid composition, although lyophilized formulations or aqueous solutions are acceptable.
  • compositions of the invention will be formulated, dosed, and administered in a fashion, i.e. amounts, concentrations, schedules, course, vehicles, and route of administration, 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 to be administered will be governed by such considerations, and is the minimum amount necessary to prevent, ameliorate, or treat the disorder. Such amount is preferably below the amount that is toxic to the host.
  • the initial pharmaceutically effective amount of the inhibitor administered parenterally per dose will be in the range of about 0.01-100 mg/kg, namely 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.
  • Acceptable diluents, carriers, excipients, and stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparag
  • the active pharmaceutical ingredients may also be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methylmethacylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions.
  • colloidal drug delivery systems for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules
  • Such techniques are disclosed in Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980).
  • Sustained-release preparations may be prepared. Suitable examples of sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing a compound of Formula I, which matrices are in the form of shaped articles, e.g. films, or microcapsules. Examples of sustained-release matrices include polyesters, hydrogels (for example, poly(2-hydroxyethyl-methacrylate), or poly(vinylalcohol)), polylactides (U.S. Pat. No.
  • copolymers of L-glutamic acid and gamma-ethyl-L-glutamate non-degradable ethylene-vinyl acetate
  • degradable lactic acid-glycolic acid copolymers such as the LUPRON DEPOTTM (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), and poly-D-( ⁇ )-3-hydroxybutyric acid.
  • the formulations to be used for in vivo administration must be sterile, which is readily accomplished by filtration through sterile filtration membranes.
  • the formulations include those suitable for the administration routes detailed herein.
  • the formulations may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. Techniques and formulations generally are found in Remington's Pharmaceutical Sciences (Mack Publishing Co., Easton, Pa.). Such methods include the step of bringing into association the active ingredient with the carrier which constitutes one or more accessory ingredients. In general the formulations are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product.
  • Formulations of a compound of Formula I suitable for oral administration may be prepared as discrete units such as pills, capsules, cachets or tablets each containing a predetermined amount of the compound of Formula I.
  • Compressed tablets may be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as a powder or granules, optionally mixed with a binder, lubricant, inert diluent, preservative, surface active or dispersing agent. Molded tablets may be made by molding in a suitable machine a mixture of the powdered active ingredient moistened with an inert liquid diluent. The tablets may optionally be coated or scored and optionally are formulated so as to provide slow or controlled release of the active ingredient therefrom.
  • Tablets, troches, lozenges, aqueous or oil suspensions, dispersible powders or granules, emulsions, hard or soft capsules, e.g. gelatin capsules, syrups or elixirs may be prepared for oral use.
  • Formulations of a compound of Formula I intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents including sweetening agents, flavoring agents, coloring agents and preserving agents, in order to provide a palatable preparation. Tablets containing the active ingredient in admixture with non-toxic pharmaceutically acceptable excipient which are suitable for manufacture of tablets are acceptable.
  • excipients may be, for example, inert diluents, such as calcium or sodium carbonate, lactose, calcium or sodium phosphate; granulating and disintegrating agents, such as maize starch, or alginic acid; binding agents, such as starch, gelatin or acacia; and lubricating agents, such as magnesium stearate, stearic acid or talc. Tablets may be uncoated or may be coated by known techniques including microencapsulation to delay disintegration and adsorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a time delay material such as glyceryl monostearate or glyceryl distearate alone or with a wax may be employed.
  • inert diluents such as calcium or sodium carbonate, lactose, calcium or sodium phosphate
  • granulating and disintegrating agents such as maize starch, or alginic acid
  • binding agents such as starch, ge
  • the formulations are preferably applied as a topical ointment or cream containing the active ingredient(s) in an amount of, for example, 0.075 to 20% w/w.
  • the active ingredients may be employed with either a paraffinic or a water-miscible ointment base.
  • the active ingredients may be formulated in a cream with an oil-in-water cream base.
  • the aqueous phase of the cream base may include a polyhydric alcohol, i.e. an alcohol having two or more hydroxyl groups such as propylene glycol, butane 1,3-diol, mannitol, sorbitol, glycerol and polyethylene glycol (including PEG 400) and mixtures thereof.
  • the topical formulations may desirably include a compound which enhances absorption or penetration of the active ingredient through the skin or other affected areas. Examples of such dermal penetration enhancers include dimethyl sulfoxide and related analogs.
  • the oily phase of the emulsions of this invention may be constituted from known ingredients in a known manner. While the phase may comprise merely an emulsifier (otherwise known as an emulgent), it desirably comprises a mixture of at least one emulsifier with a fat or an oil or with both a fat and an oil. Preferably, a hydrophilic emulsifier is included together with a lipophilic emulsifier which acts as a stabilizer. It is also preferred to include both an oil and a fat.
  • Emulgents and emulsion stabilizers suitable for use in the formulation of the invention include Tween® 60, Span® 80, cetostearyl alcohol, benzyl alcohol, myristyl alcohol, glyceryl mono-stearate and sodium lauryl sulfate.
  • Aqueous suspensions of the invention contain the active materials in admixture with excipients suitable for the manufacture of aqueous suspensions.
  • excipients include a suspending agent, such as sodium carboxymethylcellulose, croscarmellose, povidone, methylcellulose, hydroxypropyl methylcelluose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia, and dispersing or wetting agents such as a naturally occurring phosphatide (e.g., lecithin), a condensation product of an alkyl oxide (e.g.
  • ethylene oxide, propylene oxide) with a fatty acid e.g., polyoxyethylene stearate
  • a condensation product of ethylene oxide with a long chain aliphatic alcohol e.g., heptadecaethyleneoxycetanol
  • a condensation product of ethylene oxide with a partial ester derived from a fatty acid and a hexitol anhydride e.g., polyoxyethylene sorbitan monooleate.
  • the aqueous suspension may also contain one or more preservatives such as ethyl or n-propyl p-hydroxy-benzoate, one or more coloring agents, one or more flavoring agents and one or more sweetening agents, such as sucrose or saccharin.
  • the pharmaceutical composition of a compound of Formula I may be in the form of a sterile injectable preparation, such as a sterile injectable aqueous or oleaginous suspension.
  • a sterile injectable preparation such as a sterile injectable aqueous or oleaginous suspension.
  • This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, such as a solution in 1,3-butane-diol or prepared as a lyophilized powder.
  • the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution.
  • sterile fixed oils may conventionally be employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid may likewise be used in the preparation of injectables.
  • a time-release formulation intended for oral administration to humans may contain approximately 1 to 1000 mg of active material compounded with an appropriate and convenient amount of carrier material which may vary from about 5 to about 95% of the total compositions (weight:weight).
  • the pharmaceutical composition can be prepared to provide easily measurable amounts for administration.
  • an aqueous solution intended for intravenous infusion may contain from about 3 to 500 ⁇ g of the active ingredient per milliliter of solution in order that infusion of a suitable volume at a rate of about 30 mL/hr can occur.
  • Formulations suitable for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents.
  • Formulations suitable for topical administration to the eye also include eye drops wherein the active ingredient is dissolved or suspended in a suitable carrier, especially an aqueous solvent for the active ingredient.
  • the active ingredient is preferably present in such formulations in a concentration of 0.5 to 20%, advantageously 0.5 to 10% particularly about 1.5% w/w.
  • Formulations suitable for topical administration in the mouth include lozenges comprising the active ingredient in a flavored basis, usually sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert basis such as gelatin and glycerin, or sucrose and acacia; and mouthwashes comprising the active ingredient in a suitable liquid carrier.
  • Formulations for rectal administration may be presented as a suppository with a suitable base comprising for example cocoa butter or a salicylate.
  • Formulations suitable for intrapulmonary or nasal administration have a particle size for example in the range of 0.1 to 500 microns (including particle sizes in a range between 0.1 and 500 microns in increments microns such as 0.5, 1, 30 microns, 35 microns, etc.), which is administered by rapid inhalation through the nasal passage or by inhalation through the mouth so as to reach the alveolar sacs.
  • Suitable formulations include aqueous or oily solutions of the active ingredient.
  • Formulations suitable for aerosol or dry powder administration may be prepared according to conventional methods and may be delivered with other therapeutic agents such as compounds heretofore used in the treatment or prophylaxis of HIV infections as described below.
  • Formulations suitable for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams or spray formulations containing in addition to the active ingredient such carriers as are known in the art to be appropriate.
  • the formulations may be packaged in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water, for injection immediately prior to use.
  • sterile liquid carrier for example water
  • Extemporaneous injection solutions and suspensions are prepared from sterile powders, granules and tablets of the kind previously described.
  • Preferred unit dosage formulations are those containing a daily dose or unit daily sub-dose, as herein above recited, or an appropriate fraction thereof, of the active ingredient.
  • the invention further provides veterinary compositions comprising at least one active ingredient as above defined together with a veterinary carrier therefore.
  • Veterinary carriers are materials useful for the purpose of administering the composition and may be solid, liquid or gaseous materials which are otherwise inert or acceptable in the veterinary art and are compatible with the active ingredient. These veterinary compositions may be administered parenterally, orally or by any other desired route.
  • a compound of Formula I may be combined in a pharmaceutical combination formulation, or dosing regimen as combination therapy, with a second compound that has anti-hyperproliferative or chemotherapeutic properties, that is useful for treating a disease or disorder responsive to the inhibition of a JAK kinase, for example a hyperproliferative disorder (e.g. cancer), or that is useful in treating another disorder named herein.
  • the second compound of the pharmaceutical combination formulation or dosing regimen preferably has complementary activities to a compound of Formula I of the combination such that they do not adversely affect each other.
  • Such molecules are suitably present in combination in amounts that are effective for the purpose intended.
  • a compound of Formulas I may be employed alone or in combination with other therapeutic agents for the treatment of a disease or disorder described herein, such as an immunologic disorder (e.g. psoriasis or inflammation).
  • a compound of Formula I is combined in a pharmaceutical combination formulation, or dosing regimen as combination therapy, with a second therapeutic compound that has anti-inflammatory or that is useful for treating an inflammation, immune-response disorder.
  • the second therapeutic agent may be a NSAID or other anti-inflammatory agent.
  • a composition of this invention comprises a compound of Formula I, or a stereoisomer, geometric isomer, tautomer, solvate, metabolite, or pharmaceutically acceptable salt or prodrug thereof, in combination with a therapeutic agent such as an NSAID.
  • Another embodiment includes a method of treating or lessening the severity of a disease or condition responsive to the inhibition of JAK2 kinase activity in a patient, comprising administering to said patient a therapeutically effective amount of a compound of Formula I, and further comprising, administering a second chemotherapeutic agent.
  • the combination therapy may be administered as a simultaneous or sequential regimen.
  • the combination may be administered in two or more administrations.
  • the combined administration includes coadministration, using separate formulations or a single pharmaceutical formulation, and consecutive administration in either order, wherein preferably there is a time period while both (or all) active agents simultaneously exert their biological activities.
  • Suitable dosages for any of the above coadministered agents are those presently used and may be lowered due to the combined action (synergy) of the newly identified agent and other chemotherapeutic agents or treatments.
  • the combination therapy may provide “synergy” and prove “synergistic”, i.e. the effect achieved when the active ingredients used together is greater than the sum of the effects that results from using the compounds separately.
  • a synergistic effect may be attained when the active ingredients are: (1) co-formulated and administered or delivered simultaneously in a combined, unit dosage formulation; (2) delivered by alternation or in parallel as separate formulations; or (3) by some other regimen.
  • a synergistic effect may be attained when the compounds are administered or delivered sequentially, e.g. by different injections in separate syringes.
  • an effective dosage of each active ingredient is administered sequentially, i.e. serially, whereas in combination therapy, effective dosages of two or more active ingredients are administered together.
  • Another embodiment includes in vivo metabolic products of an administered compound of Formula I. Such products may result for example from the oxidation, reduction, hydrolysis, amidation, deamidation, esterification, deesterification, enzymatic cleavage, and the like, of the administered compound.
  • Metabolite products typically are identified by preparing a radiolabelled (e.g. 14 C or 3 H) isotope of a compound of the invention, administering it parenterally in a detectable dose (e.g. greater than about 0.5 mg/kg) to an animal such as rat, mouse, guinea pig, monkey, or to man, allowing sufficient time for metabolism to occur (typically about 30 seconds to 30 hours) and isolating its conversion products from the urine, blood or other biological samples.
  • a detectable dose e.g. greater than about 0.5 mg/kg
  • kits for treating a disease or disorder responsive to the inhibition of a JAK kinase includes:
  • the kit further includes:
  • a second pharmaceutical composition which includes a chemotherapeutic agent.
  • the instructions include instructions for the simultaneous, sequential or separate administration of said first and second pharmaceutical compositions to a patient in need thereof.
  • the first and second compositions are contained in separate containers.
  • the first and second compositions are contained in the same container.
  • Containers for use include, for example, bottles, vials, syringes, blister pack, etc.
  • the containers may be formed from a variety of materials such as glass or plastic.
  • the container includes a compound of Formula I or formulation thereof which is effective for treating the condition and may have a sterile access port (for example the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
  • the container includes a composition comprising at least one compound of Formula I.
  • the label or package insert indicates that the composition is used for treating the condition of choice, such as cancer.
  • the label or package inserts indicates that the composition comprising the compound of Formula I can be used to treat a disorder.
  • the label or package insert may indicate that the patient to be treated is one having a disorder characterized by overactive or irregular kinase activity.
  • the label or package insert may also indicate that the composition can be used to treat other disorders.
  • the article of manufacture may comprise (a) a first container with a compound of Formula I contained therein; and (b) a second container with a second pharmaceutical formulation contained therein, wherein the second pharmaceutical formulation comprises a chemotherapeutic agent.
  • the article of manufacture in this embodiment of the invention may further comprise a package insert indicating that the first and second compounds can be used to treat patients at risk of stroke, thrombus or thrombosis disorder.
  • the article of manufacture may further comprise a second (or third) container comprising a pharmaceutically-acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer's solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes.
  • BWFI bacteriostatic water for injection
  • the compounds of Formula I can be used to control JAK protein kinases, tyrosine kinases, additional serine/threonine kinases, and/or dual specificity kinases.
  • they are useful as pharmacological standards for use in the development of new biological tests, assays and in the search for new pharmacological agents.
  • Compounds of Formula I may be assayed for the ability to modulate the activity of JAK protein kinases, tyrosine kinases, additional serine/threonine kinases, and/or dual specificity kinases in vitro and in vivo.
  • In vitro assays include biochemical and cell-based assays that determine inhibition of the kinase activity.
  • Alternate in vitro assays quantify the ability of the compound of Formula I to bind to kinases and may be measured either by radiolabelling the compound of Formula I prior to binding, isolating the compound of Formula I/kinase complex and determining the amount of radiolabel bound, or by running a competition experiment where a compound of Formula I is incubated with known radiolabeled ligands.
  • Compounds of Formula I may be assayed for the ability to modulate the activity of Janus protein kinases, tyrosine kinases, additional serine/threonine kinases, and/or dual specificity kinases in vitro and in vivo.
  • In vitro assays include biochemical and cell-based assays that determine inhibition of the kinase activity.
  • Alternate in vitro assays quantify the ability of the compound of Formula I to bind to kinases and may be measured either by radiolabelling the compound of Formula I prior to binding, isolating the compound of Formula I/kinase complex and determining the amount of radiolabel bound, or by running a competition experiment where a compound of Formula I is incubated with known radiolabeled ligands.
  • the catalytically active kinase domain of human JAK1, JAK2, JAK3 or TYK2 was purified from extracts of SF9 insect cells infected with a recombinant baculovirus expression vector encoding the human JAK1, JAK2, JAK3 or TYK2 kinase domains (JAK1 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 JAK1, 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 JAK1, JAK2, JAK3 or TYK2 kinase domains was measured in vitro by monitoring phosphorylation of JAK3 derived peptides using the Caliper LabChip technology (see Examples).
  • 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) fluorescently labeled on the N-terminus with 5-carboxyfluorescein using the Caliper LabChip technology (Caliper Life Sciences, Hopkinton, Mass.).
  • Examples 1-304 To determine the inhibition constants (Ki) of Examples 1-304, compounds were diluted serially in DMSO and added to 50 ⁇ L kinase reactions containing 0.2 nM purified JAK2 enzyme, 100 mM Hepes pH7.2, 0.015% Brij-35, 1.5 ⁇ M peptide substrate, 25 ⁇ M ATP, 10 mM MgCl 2 , 4 mM DTT at a final DMSO concentration of 2%. Reactions were incubated at 22° C.
  • 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) fluorescently labeled on the N-terminus with 5-carboxyfluorescein using the Caliper LabChip technology (Caliper Life Sciences, Hopkinton, Mass.).
  • Examples 1-312 To determine the inhibition constants (Ki) of Examples 1-312, compounds were diluted serially in DMSO and added to 50 uL kinase reactions containing 1.5 nM JAK1, 0.2 nM purified JAK2 or 1 nM purified TYK2 enzyme, 100 mM Hepes pH7.2, 0.015% Brij-35, 1.5 uM peptide substrate, 25 uM ATP, 10 mM MgCl 2 , 4 mM DTT at a final DMSO concentration of 2%. Reactions were incubated at 22° C.
  • 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) fluorescently labeled on the N-terminus with 5-carboxyfluorescein using the Caliper LabChip technology (Caliper Life Sciences, Hopkinton, Mass.).
  • the activities of compounds 1-312 were determined in cell-based assays that are designed to measure Janus kinase dependent signaling. 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 10 5 cells per well and a final DMSO concentration of 0.57%.
  • Set-2 cells German Collection of Microorganisms and Cell Cultures (DSMZ); Braunschweig, Germany
  • IL-12 Human recombinant IL-12 (R&D systems; Minneapolis, Minn.) was then added at a final concentration of 10 ng/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, Md.) according to the manufacturer's protocol and EC 50 values were determined.
  • MSD Meso Scale Discovery
  • HM-N Isolute® HM-N is a modified form of diatomaceous earth IMS industrial methylated spirits
  • LCMS High Pressure Liquid Chromatography-Mass Spectrometry
  • Method A Experiments performed on a Waters Micromass ZQ quadrupole mass spectrometer linked to a Hewlett Packard HP1100 LC system with diode array detector. This system uses a Higgins Clipeus 5 micron C18 100 ⁇ 3.0 mm column and a 1 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 minute followed by a gradient up to 5% solvent A and 95% solvent B over the next 14 minutes. The final solvent system was held constant for a further 5 minutes.
  • Method B Experiments performed on a Waters Platform LC quadrupole mass spectrometer linked to a Hewlett Packard HP1100 LC system with diode array detector and 100 position autosampler using a Phenomenex Luna C18(2) 30 ⁇ 4.6 mm column and a 2 ml/minute flow rate.
  • the 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 0.50 minutes followed by a gradient up to 5% solvent A and 95% solvent B over the next 4 minutes. The final solvent system was held constant for a further 0.50 minutes.
  • Microwave experiments were carried out using a Biotage Initiator 60TM or CEM Explorer®. Temperatures from 40-250° C. can be achieved, and pressures of up to 30 bar can be reached.
  • reaction flasks were typically fitted with rubber septa for the introduction of substrates and reagents via syringe. Glassware was oven dried and/or heat dried.
  • reaction mixture was heated at 110° C. by microwave for 10 min.
  • the reaction mixture was diluted with ethyl acetate (50 mL) and filtered through celite. The filtrate was then washed with brine (3 ⁇ 20 mL), dried over anhydrous sodium sulfate, filtered, and concentrated. Purification by preparative HPLC afforded 8-(3-methoxyphenyl)-N-(2-methylpyridin-4-yl)-[1,2,4]triazolo[1,5-a]pyridin-2-amine (80 mg, 39%).
  • Examples 3-126 shown in Table 1 were prepared according to the above-described methods.
  • the reaction mixture was heated at 140° C. for 30 min in the microwave.
  • LCMS of the reaction mixture showed ⁇ 60% conversion, and additional bis(diphenylphosphino)ferrocenepalladium(II) chloride (95 mg, 0.12 mmol, 0.05 equiv) and 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (225 mg, 1.16 mmol, 0.500 equiv) were added.
  • the reaction mixture was then heated at 140° C. for 30 min in the microwave.
  • the resulting residue was dissolved in methanol (4 mL) and circulated through a H-Cube® continuous-flow hydrogenation reactor (ThalesNano) fitted with a palladium on carbon catalyst cartridge at 30° C.
  • the resulting solution was concentrated in vacuo to provide product as a pink oil, which was used without further purification.
  • Examples 130-312 shown in Table 2 were prepared according to the above-described methods.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Diabetes (AREA)
  • Biomedical Technology (AREA)
  • Epidemiology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Hematology (AREA)
  • Oncology (AREA)
  • Rheumatology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Dermatology (AREA)
  • Communicable Diseases (AREA)
  • Psychology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Cardiology (AREA)
  • Endocrinology (AREA)
  • Psychiatry (AREA)
  • Pulmonology (AREA)
  • Urology & Nephrology (AREA)
  • Hospice & Palliative Care (AREA)
  • Pain & Pain Management (AREA)
  • Virology (AREA)
  • Emergency Medicine (AREA)
  • Transplantation (AREA)
US12/488,312 2008-06-20 2009-06-19 Triazolopyridine JAK Inhibitor Compounds and Methods Abandoned US20100048557A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
US12/488,312 US20100048557A1 (en) 2008-06-20 2009-06-19 Triazolopyridine JAK Inhibitor Compounds and Methods
US13/471,269 US8609687B2 (en) 2008-06-20 2012-05-14 Triazolopyridine JAK inhibitor compounds and methods
US14/049,458 US9434732B2 (en) 2008-06-20 2013-10-09 Triazolopyridine JAK inhibitor compounds and methods
US15/220,087 US20160333011A1 (en) 2008-06-20 2016-07-26 Triazolopyridine jak inhibitor compounds and methods

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US7450608P 2008-06-20 2008-06-20
US12/488,312 US20100048557A1 (en) 2008-06-20 2009-06-19 Triazolopyridine JAK Inhibitor Compounds and Methods

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/471,269 Continuation US8609687B2 (en) 2008-06-20 2012-05-14 Triazolopyridine JAK inhibitor compounds and methods

Publications (1)

Publication Number Publication Date
US20100048557A1 true US20100048557A1 (en) 2010-02-25

Family

ID=41434469

Family Applications (4)

Application Number Title Priority Date Filing Date
US12/488,312 Abandoned US20100048557A1 (en) 2008-06-20 2009-06-19 Triazolopyridine JAK Inhibitor Compounds and Methods
US13/471,269 Expired - Fee Related US8609687B2 (en) 2008-06-20 2012-05-14 Triazolopyridine JAK inhibitor compounds and methods
US14/049,458 Expired - Fee Related US9434732B2 (en) 2008-06-20 2013-10-09 Triazolopyridine JAK inhibitor compounds and methods
US15/220,087 Abandoned US20160333011A1 (en) 2008-06-20 2016-07-26 Triazolopyridine jak inhibitor compounds and methods

Family Applications After (3)

Application Number Title Priority Date Filing Date
US13/471,269 Expired - Fee Related US8609687B2 (en) 2008-06-20 2012-05-14 Triazolopyridine JAK inhibitor compounds and methods
US14/049,458 Expired - Fee Related US9434732B2 (en) 2008-06-20 2013-10-09 Triazolopyridine JAK inhibitor compounds and methods
US15/220,087 Abandoned US20160333011A1 (en) 2008-06-20 2016-07-26 Triazolopyridine jak inhibitor compounds and methods

Country Status (15)

Country Link
US (4) US20100048557A1 (ja)
EP (1) EP2288260A4 (ja)
JP (1) JP5512665B2 (ja)
KR (1) KR20110033223A (ja)
CN (1) CN102131389A (ja)
AU (1) AU2009259853A1 (ja)
BR (1) BRPI0910021A2 (ja)
CA (1) CA2727036C (ja)
IL (1) IL209763A0 (ja)
MX (1) MX2010014005A (ja)
PE (1) PE20110545A1 (ja)
RU (1) RU2560153C2 (ja)
SG (1) SG178812A1 (ja)
WO (1) WO2009155551A1 (ja)
ZA (1) ZA201008957B (ja)

Cited By (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100035875A1 (en) * 2008-06-20 2010-02-11 Bing-Yan Zhu Triazolopyridine jak inhibitor compounds and methods
US20100311693A1 (en) * 2009-06-05 2010-12-09 Cephalon, Inc. Preparation and Uses of 1,2,4-Triazolo [1,5a] Pyridine Derivatives
US8551980B2 (en) 2009-11-30 2013-10-08 Bayer Intellectual Property Gmbh Substituted triazolopyridines
US9023851B2 (en) 2010-08-27 2015-05-05 Merck Patent Gmbh Triazolopyrazine derivatives
US9120804B2 (en) 2012-02-21 2015-09-01 Merck Patent Gmbh 8-substituted 2-amino-[1,2,4] triazolo [1, 5-A] pyrazines as Syk tryrosine kinase inhibitors and GCN2 serin kinase inhibitors
WO2016139212A1 (en) 2015-03-04 2016-09-09 F. Hoffmann-La Roche Ag Triazolopyridine compounds and methods of use thereof
WO2017089390A1 (en) 2015-11-23 2017-06-01 F. Hoffmann-La Roche Ag Janus kinases inhibitors, compositions thereof and use thereof
WO2017140825A1 (en) 2016-02-18 2017-08-24 F. Hoffmann-La Roche Ag Therapeutic compounds, compositions and methods of use thereof
WO2017191098A1 (en) 2016-05-05 2017-11-09 F. Hoffmann-La Roche Ag Pyrazole derivatives, compositions and therapeutic use thereof
WO2018046409A1 (en) 2016-09-06 2018-03-15 F. Hoffmann-La Roche Ag 8-(azetidin-1-yl)-[1,2,4]triazolo[1,5-a]pyridinyl compounds, compositions and methods of use thereof
WO2018083085A1 (en) 2016-11-02 2018-05-11 F. Hoffmann-La Roche Ag PYRAZOLO[1,5a]PYRIMIDINE DERIVATIVES AS IRAK4 MODULATORS
WO2018122212A1 (en) 2016-12-29 2018-07-05 F. Hoffmann-La Roche Ag Pyrazolopyrimidine compounds and methods of use thereof
WO2018166993A2 (en) 2017-03-14 2018-09-20 F. Hoffmann-La Roche Ag Pyrazolochlorophenyl compounds, compositions and methods of use thereof
WO2018215389A1 (en) 2017-05-22 2018-11-29 F. Hoffmann-La Roche Ag Therapeutic compounds and compositions, and methods of use thereof
WO2018215390A1 (en) 2017-05-22 2018-11-29 F. Hoffmann-La Roche Ag Therapeutic compounds and compositions, and methods of use thereof
WO2018234343A1 (en) 2017-06-21 2018-12-27 F. Hoffmann-La Roche Ag BENZOFURANES AS MODULATORS OF IRAQ4
WO2018234342A1 (en) 2017-06-21 2018-12-27 F. Hoffmann-La Roche Ag ISOINDOLINONE DERIVATIVES AS MODULATORS OF IRAK4
WO2018234345A1 (en) 2017-06-21 2018-12-27 F. Hoffmann-La Roche Ag PYRAZOLO [1,5A] PYRIMIDINE DERIVATIVES AS MODULATORS OF IRAK4
WO2020097537A2 (en) 2018-11-09 2020-05-14 Genentech, Inc. Fused ring compounds
WO2020257143A1 (en) 2019-06-18 2020-12-24 Genentech, Inc. Pyrazolopyrimidine aryl ether inhibitors of jak kinases and uses thereof
WO2020257142A1 (en) 2019-06-18 2020-12-24 Genentech, Inc. Tetrazole-substituted pyrazolopyrimidine inhibitors of jak kinases and uses thereof
WO2020257145A1 (en) 2019-06-18 2020-12-24 Genentech, Inc. Pyrazolopyrimidine sulfone inhibitors of jak kinases and uses thereof
WO2022035790A1 (en) 2020-08-12 2022-02-17 Genentech, Inc. Synthesis of quinazoline compounds
WO2023150653A1 (en) 2022-02-07 2023-08-10 Genentech, Inc. Process for synthesis of quinazoline compounds
US11760744B2 (en) 2018-08-16 2023-09-19 Genentech, Inc. Fused ring compounds

Families Citing this family (38)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101952275B (zh) 2008-02-22 2014-06-18 弗·哈夫曼-拉罗切有限公司 β-淀粉样蛋白的调节剂
BRPI0910021A2 (pt) * 2008-06-20 2015-09-01 Genentech Inc "composto, composição farmacêutica, método para tratar ou atenuar a gravidade de uma doença ou condição responsiva à inibição da atividade jak2 quinas em um paciente, kit para o tratamento de uma doença ou distúrbio responsivo à inibição da jak quinase"
TWI453207B (zh) 2008-09-08 2014-09-21 Signal Pharm Llc 胺基三唑并吡啶,其組合物及使用其之治療方法
WO2010040661A1 (en) 2008-10-09 2010-04-15 F. Hoffmann-La Roche Ag Modulators for amyloid beta
CN102209537A (zh) 2008-11-10 2011-10-05 弗·哈夫曼-拉罗切有限公司 杂环γ分泌酶调节剂
EP2396324A1 (en) * 2009-02-13 2011-12-21 Fovea Pharmaceuticals Ý1, 2, 4¨triazolo ý1, 5 -a¨pyridines as kinase inhibitors
AU2011211410B2 (en) * 2009-02-13 2013-01-31 Fovea Pharmaceuticals Sa [1,2,4] triazolo [1,5-A] pyridines as kinase inhibitors
TWI462920B (zh) * 2009-06-26 2014-12-01 葛萊伯格有限公司 用於治療退化性及發炎疾病之新穎化合物
CA2784769A1 (en) 2010-01-15 2011-07-21 Janssen Pharmaceuticals, Inc. Novel substituted triazole derivatives as gamma secretase modulators
US20110190269A1 (en) * 2010-02-01 2011-08-04 Karlheinz Baumann Gamma secretase modulators
US8486967B2 (en) 2010-02-17 2013-07-16 Hoffmann-La Roche Inc. Heteroaryl substituted piperidines
EP2588105A1 (en) 2010-07-01 2013-05-08 Cellzome Limited Triazolopyridines as tyk2 inhibitors
CN103298794A (zh) 2010-11-09 2013-09-11 塞尔卓姆有限公司 作为tyk2抑制剂的吡啶化合物及其氮杂类似物
JP6033230B2 (ja) * 2010-12-06 2016-11-30 セファロン、インク. 狼瘡の処置のためのヤーヌスキナーゼ2(jak2)インヒビタ
AR086042A1 (es) * 2011-04-28 2013-11-13 Galapagos Nv Compuesto util para el tratamiento de enfermedades degenerativas e inflamatorias y composicion farmaceutica
JP6106745B2 (ja) 2012-05-16 2017-04-05 ヤンセン ファーマシューティカルズ,インコーポレーテッド (特に)アルツハイマー病の治療に有用な置換3,4−ジヒドロ−2H−ピリド[1,2−a]ピラジン−1,6−ジオン誘導体
JP6171003B2 (ja) * 2012-05-24 2017-07-26 ノバルティス アーゲー ピロロピロリジノン化合物
EP2855451B1 (en) 2012-05-24 2017-10-04 Cellzome Limited Heterocyclyl pyrimidine analogues as tyk2 inhibitors
SG11201503141TA (en) 2012-11-01 2015-06-29 Incyte Corp Tricyclic fused thiophene derivatives as jak inhibitors
AU2013366668B2 (en) 2012-12-20 2017-07-20 Janssen Pharmaceutica Nv Novel tricyclic 3,4-dihydro-2H-pyrido[1,2-alpha]pyrazine -1,6-dione derivatives as gamma secretase modulators
WO2014111457A1 (en) 2013-01-17 2014-07-24 Janssen Pharmaceutica Nv Novel substituted pyrido-piperazinone derivatives as gamma secretase modulators
CA2922770A1 (en) * 2013-09-05 2015-03-12 F. Hoffmann-La Roche Ag Triazolopyridine compounds, compositions and methods of use thereof
BR112016012469A2 (pt) * 2014-01-10 2017-08-08 Lilly Co Eli Isopropil triazolo piridina, seus usos, seu processo de preparação e seus intermediários, composição farmacêutica
AU2015206629B2 (en) * 2014-01-14 2020-03-05 The Johns Hopkins University Cyclodextrin compositions encapsulating a selective ATP inhibitor and uses thereof
US10562897B2 (en) 2014-01-16 2020-02-18 Janssen Pharmaceutica Nv Substituted 3,4-dihydro-2H-pyrido[1,2-a]pyrazine-1,6-diones as gamma secretase modulators
CN106687462A (zh) 2014-04-30 2017-05-17 因赛特公司 Jak1抑制剂的制备方法以及其新形式
BR112017007072A2 (pt) * 2014-10-06 2018-01-30 Flatley Discovery Lab Llc compostos de triazolopiridina e métodos para o tratamento de fibrose cística
WO2017007634A1 (en) * 2015-07-06 2017-01-12 The Board Of Regents Of The University Of Texas System Benzamide or benzamine compounds useful as anticancer agents for the treatment of human cancers
EP3371186A1 (en) 2015-11-03 2018-09-12 Theravance Biopharma R&D IP, LLC Jak kinase inhibitor compounds for treatment of respiratory disease
EP3390402B1 (en) * 2015-12-18 2020-01-15 H. Hoffnabb-La Roche Ag Therapeutic compounds, compositions and methods of use thereof
CA3059785A1 (en) 2017-05-01 2018-11-08 Theravance Biopharma R&D Ip, Llc Methods of treatment using a jak inhibitor compound
EP3672971B1 (en) * 2017-12-19 2021-07-14 Boehringer Ingelheim International GmbH Triazolo pyridines as modulators of gamma-secretase
MX2020010322A (es) 2018-03-30 2022-11-30 Incyte Corp Tratamiento de la hidradenitis supurativa mediante el uso de inhibidores de actividad de la cinasa janus (jak).
US20220143049A1 (en) 2019-03-21 2022-05-12 Onxeo A dbait molecule in combination with kinase inhibitor for the treatment of cancer
EP4054579A1 (en) 2019-11-08 2022-09-14 Institut National de la Santé et de la Recherche Médicale (INSERM) Methods for the treatment of cancers that have acquired resistance to kinase inhibitors
JP7371253B2 (ja) * 2019-11-25 2023-10-30 デウン ファーマシューティカル カンパニー リミテッド 新規なトリアゾロピリジン誘導体およびこれを含む薬学組成物
WO2021148581A1 (en) 2020-01-22 2021-07-29 Onxeo Novel dbait molecule and its use
WO2023064458A1 (en) * 2021-10-13 2023-04-20 Yale University Selective jak2 inhibitors and methods of use

Citations (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4847256A (en) * 1986-10-16 1989-07-11 American Cyanamid Company 4,5-dihydro and 4,5,6,7-tetrahydropyrazolo(1,5-A)-pyrimidines
US6355653B1 (en) * 1999-09-06 2002-03-12 Hoffmann-La Roche Inc. Amino-triazolopyridine derivatives
US6514989B1 (en) * 2001-07-20 2003-02-04 Hoffmann-La Roche Inc. Aromatic and heteroaromatic substituted 1,2,4-triazolo pyridine derivatives
US6693116B2 (en) * 2001-10-08 2004-02-17 Hoffmann-La Roche Inc. Adenosine receptor ligands
US20040209878A1 (en) * 2002-09-04 2004-10-21 Schering Corporation And Pharmacopeia, Inc. Novel pyrazolopyrimidines as cyclin dependent kinase inhibitors
US20050020639A1 (en) * 2003-07-25 2005-01-27 Pfizer Inc Novel compounds
US20050288502A1 (en) * 2004-06-25 2005-12-29 Andersen Denise L Substituted heterocyclic compounds and methods of use
US20060089362A1 (en) * 2002-04-23 2006-04-27 Shionogi & Co., Ltd Pyrazolo [1,5-a] pyrimidine derivative and nad (p) h oxidase inhibitor containing the same
US20060128725A1 (en) * 2002-09-04 2006-06-15 Schering Corporation Novel pyrazolopyrimidines as cyclin dependent kinase inhibitors
US20060183761A1 (en) * 2005-02-03 2006-08-17 Mark Ledeboer Pyrrolopyrimidines useful as inhibitors of protein kinase
US20060241134A1 (en) * 2003-05-27 2006-10-26 Altana Pharma Ga Pharmaceutical combinations of a proton pump inhibitor and a compound which modifies gastrointestinal motility
US7161003B1 (en) * 2002-09-04 2007-01-09 Schering Corporation Pyrazolopyrimidines as cyclin dependent kinase inhibitors
US20070072881A1 (en) * 2002-09-04 2007-03-29 Schering Corporation Novel pyrazolopyrimidines as cyclin dependent kinase inhibitors
US20070142402A1 (en) * 2005-12-15 2007-06-21 Rigel Pharmaceuticals, Inc. Kinase Inhibitors And Their Uses
US20070270408A1 (en) * 2003-04-11 2007-11-22 Novo Nordisk A/S Pharmaceutical use of substituted pyrazolo[1,5-a]pyrimidines
US20070281951A1 (en) * 2002-09-04 2007-12-06 Schering Corporation Novel pyrazolopyrimidines as cyclin dependent kinase inhibitors
US7306631B2 (en) * 2004-03-30 2007-12-11 The Procter & Gamble Company Keratin dyeing compounds, keratin dyeing compositions containing them, and use thereof
US20080014189A1 (en) * 2002-11-15 2008-01-17 Pierce Albert C Diaminotriazoles useful as inhibitors of protein kinases
US20080050384A1 (en) * 2002-09-04 2008-02-28 Shering Corporation And Pharmacopeia, Inc. Novel pyrazolopyrimidines as cyclin dependent kinase inhibitors
US20100035875A1 (en) * 2008-06-20 2010-02-11 Bing-Yan Zhu Triazolopyridine jak inhibitor compounds and methods

Family Cites Families (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6579857B1 (en) 1999-06-11 2003-06-17 Evanston Northwestern Healthcare Research Institute Combination cancer therapy comprising adenosine and deaminase enzyme inhibitors
MXPA04003277A (es) * 2001-10-08 2004-07-23 Hoffmann La Roche Amida del acido 8-amino-?1,2,4?triazolo?1,5-a?piridina-6-carboxilico.
GB2423083B (en) 2003-12-03 2007-07-11 Cytopia Res Pty Ltd Azole-based kinase inhibitors
ZA200602666B (en) 2004-01-12 2007-09-26 Cytopia Res Pty Ltd Selective kinase inhibitors
BRPI0516819A (pt) 2004-10-07 2008-09-23 Warner Lambert Co derivados de triazolopiridina e agentes antibacterianos
AR054416A1 (es) * 2004-12-22 2007-06-27 Incyte Corp Pirrolo [2,3-b]piridin-4-il-aminas y pirrolo [2,3-b]pirimidin-4-il-aminas como inhibidores de las quinasas janus. composiciones farmaceuticas.
CA2604551A1 (en) 2005-05-03 2007-03-08 Rigel Pharmaceuticals, Inc. Jak kinase inhibitors and their uses
EP1899370A4 (en) 2005-05-12 2009-11-11 Cytopia Res Pty Ltd CRYSTALLINE STRUCTURE AND USE THEREOF
EP1904457B1 (en) 2005-06-08 2017-09-06 Rigel Pharmaceuticals, Inc. Compositions and methods for inhibition of the jak pathway
US20100216798A1 (en) 2005-07-29 2010-08-26 Astellas Pharma Inc Fused heterocycles as lck inhibitors
LT2474545T (lt) * 2005-12-13 2017-02-27 Incyte Holdings Corporation Heteroarilu pakeisti pirolo[2,3-b]piridinai ir pirolo[2,3-b]pirimidinai kaip janus kinazės inhibitoriai
PE20070978A1 (es) 2006-02-14 2007-11-15 Novartis Ag COMPUESTOS HETEROCICLICOS COMO INHIBIDORES DE FOSFATIDILINOSITOL 3-QUINASAS (PI3Ks)
WO2007103760A2 (en) 2006-03-02 2007-09-13 Smithkline Beecham Corporation Thiazolones for use as pi3 kinase inhibitors
EP1873157A1 (en) 2006-06-21 2008-01-02 Bayer Schering Pharma Aktiengesellschaft Pyrazolopyrimidines and salts thereof, pharmaceutical compositions comprising same, methods of preparing same and uses of same
PE20080403A1 (es) 2006-07-14 2008-04-25 Amgen Inc Derivados heterociclicos fusionados y metodos de uso
WO2008025821A1 (en) * 2006-08-30 2008-03-06 Cellzome Limited Triazole derivatives as kinase inhibitors
WO2008052734A1 (en) 2006-10-30 2008-05-08 Novartis Ag Heterocyclic compounds as antiinflammatory agents
JP2010524911A (ja) * 2007-04-18 2010-07-22 アストラゼネカ アクチボラグ 5−アミノピラゾール−3−イル−3H−イミダゾ[4,5−b]ピリジン誘導体と癌の治療のためのその使用
WO2009017954A1 (en) * 2007-08-01 2009-02-05 Phenomix Corporation Inhibitors of jak2 kinase
WO2009027283A1 (en) 2007-08-31 2009-03-05 Merck Serono S.A. Triazolopyridine compounds and their use as ask inhibitors
GB0719803D0 (en) * 2007-10-10 2007-11-21 Cancer Rec Tech Ltd Therapeutic compounds and their use
DK2288610T3 (en) * 2008-03-11 2016-11-28 Incyte Holdings Corp Azetidinesulfonic AND CYCLOBUTANDERIVATER AS JAK INHIBITORS
JP2011518219A (ja) * 2008-04-22 2011-06-23 ポートラ ファーマシューティカルズ, インコーポレイテッド タンパク質キナーゼの阻害剤
BRPI0910021A2 (pt) * 2008-06-20 2015-09-01 Genentech Inc "composto, composição farmacêutica, método para tratar ou atenuar a gravidade de uma doença ou condição responsiva à inibição da atividade jak2 quinas em um paciente, kit para o tratamento de uma doença ou distúrbio responsivo à inibição da jak quinase"
WO2010010184A1 (en) 2008-07-25 2010-01-28 Galapagos Nv [1, 2, 4] triazolo [1, 5-a] pyridines as jak inhibitors
WO2010010189A1 (en) 2008-07-25 2010-01-28 Galapagos Nv Novel compounds useful for the treatment of degenerative and inflammatory diseases
WO2010010186A1 (en) 2008-07-25 2010-01-28 Galapagos Nv Novel compounds useful for the treatment of degenerative and inflammatory diseases
WO2010010188A1 (en) 2008-07-25 2010-01-28 Galapagos Nv Novel compounds useful for the treatment of degenerative and inflammatory diseases.
BRPI1009637A2 (pt) 2009-06-05 2019-04-30 Cephalon, Inc composto, composição e uso de um composto
US8993756B2 (en) 2011-12-06 2015-03-31 Merck Sharp & Dohme Corp. Pyrrolopyrimidines as janus kinase inhibitors

Patent Citations (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4847256A (en) * 1986-10-16 1989-07-11 American Cyanamid Company 4,5-dihydro and 4,5,6,7-tetrahydropyrazolo(1,5-A)-pyrimidines
US6355653B1 (en) * 1999-09-06 2002-03-12 Hoffmann-La Roche Inc. Amino-triazolopyridine derivatives
US6514989B1 (en) * 2001-07-20 2003-02-04 Hoffmann-La Roche Inc. Aromatic and heteroaromatic substituted 1,2,4-triazolo pyridine derivatives
US6693116B2 (en) * 2001-10-08 2004-02-17 Hoffmann-La Roche Inc. Adenosine receptor ligands
US20060089362A1 (en) * 2002-04-23 2006-04-27 Shionogi & Co., Ltd Pyrazolo [1,5-a] pyrimidine derivative and nad (p) h oxidase inhibitor containing the same
US20070054925A1 (en) * 2002-09-04 2007-03-08 Schering Corporation Novel pyrazolopyrimidines as cyclin dependent kinase inhibitors
US7161003B1 (en) * 2002-09-04 2007-01-09 Schering Corporation Pyrazolopyrimidines as cyclin dependent kinase inhibitors
US20040209878A1 (en) * 2002-09-04 2004-10-21 Schering Corporation And Pharmacopeia, Inc. Novel pyrazolopyrimidines as cyclin dependent kinase inhibitors
US20060128725A1 (en) * 2002-09-04 2006-06-15 Schering Corporation Novel pyrazolopyrimidines as cyclin dependent kinase inhibitors
US20080050384A1 (en) * 2002-09-04 2008-02-28 Shering Corporation And Pharmacopeia, Inc. Novel pyrazolopyrimidines as cyclin dependent kinase inhibitors
US7119200B2 (en) * 2002-09-04 2006-10-10 Schering Corporation Pyrazolopyrimidines as cyclin dependent kinase inhibitors
US20070281951A1 (en) * 2002-09-04 2007-12-06 Schering Corporation Novel pyrazolopyrimidines as cyclin dependent kinase inhibitors
US20070072881A1 (en) * 2002-09-04 2007-03-29 Schering Corporation Novel pyrazolopyrimidines as cyclin dependent kinase inhibitors
US20070037824A1 (en) * 2002-09-04 2007-02-15 Schering Corporation Novel pyrazolopyrimidines as cyclin dependent kinase inhibitors
US20070054906A1 (en) * 2002-09-04 2007-03-08 Schering Corporation Novel pyrazolopyrimidines as cyclin dependent kinase inhibitors
US7196078B2 (en) * 2002-09-04 2007-03-27 Schering Corpoartion Trisubstituted and tetrasubstituted pyrazolopyrimidines as cyclin dependent kinase inhibitors
US20080014189A1 (en) * 2002-11-15 2008-01-17 Pierce Albert C Diaminotriazoles useful as inhibitors of protein kinases
US20070270408A1 (en) * 2003-04-11 2007-11-22 Novo Nordisk A/S Pharmaceutical use of substituted pyrazolo[1,5-a]pyrimidines
US20060241134A1 (en) * 2003-05-27 2006-10-26 Altana Pharma Ga Pharmaceutical combinations of a proton pump inhibitor and a compound which modifies gastrointestinal motility
US20050020639A1 (en) * 2003-07-25 2005-01-27 Pfizer Inc Novel compounds
US7306631B2 (en) * 2004-03-30 2007-12-11 The Procter & Gamble Company Keratin dyeing compounds, keratin dyeing compositions containing them, and use thereof
US20050288502A1 (en) * 2004-06-25 2005-12-29 Andersen Denise L Substituted heterocyclic compounds and methods of use
US20060183761A1 (en) * 2005-02-03 2006-08-17 Mark Ledeboer Pyrrolopyrimidines useful as inhibitors of protein kinase
US20070142402A1 (en) * 2005-12-15 2007-06-21 Rigel Pharmaceuticals, Inc. Kinase Inhibitors And Their Uses
US20100035875A1 (en) * 2008-06-20 2010-02-11 Bing-Yan Zhu Triazolopyridine jak inhibitor compounds and methods

Cited By (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100035875A1 (en) * 2008-06-20 2010-02-11 Bing-Yan Zhu Triazolopyridine jak inhibitor compounds and methods
US8889673B2 (en) 2008-06-20 2014-11-18 Genentech, Inc. Triazolopyridine JAK inhibitor compounds and methods
US20100311693A1 (en) * 2009-06-05 2010-12-09 Cephalon, Inc. Preparation and Uses of 1,2,4-Triazolo [1,5a] Pyridine Derivatives
US8501936B2 (en) * 2009-06-05 2013-08-06 Cephalon, Inc. Preparation and uses of 1,2,4-triazolo [1,5a] pyridine derivatives
US8633173B2 (en) 2009-06-05 2014-01-21 Cephalon, Inc Preparation and uses of 1,2,4-triazolo [1,5a] pyridine derivatives
US8551980B2 (en) 2009-11-30 2013-10-08 Bayer Intellectual Property Gmbh Substituted triazolopyridines
US9023851B2 (en) 2010-08-27 2015-05-05 Merck Patent Gmbh Triazolopyrazine derivatives
US9120804B2 (en) 2012-02-21 2015-09-01 Merck Patent Gmbh 8-substituted 2-amino-[1,2,4] triazolo [1, 5-A] pyrazines as Syk tryrosine kinase inhibitors and GCN2 serin kinase inhibitors
WO2016139212A1 (en) 2015-03-04 2016-09-09 F. Hoffmann-La Roche Ag Triazolopyridine compounds and methods of use thereof
WO2017089390A1 (en) 2015-11-23 2017-06-01 F. Hoffmann-La Roche Ag Janus kinases inhibitors, compositions thereof and use thereof
WO2017140825A1 (en) 2016-02-18 2017-08-24 F. Hoffmann-La Roche Ag Therapeutic compounds, compositions and methods of use thereof
WO2017191098A1 (en) 2016-05-05 2017-11-09 F. Hoffmann-La Roche Ag Pyrazole derivatives, compositions and therapeutic use thereof
WO2018046409A1 (en) 2016-09-06 2018-03-15 F. Hoffmann-La Roche Ag 8-(azetidin-1-yl)-[1,2,4]triazolo[1,5-a]pyridinyl compounds, compositions and methods of use thereof
EP4198036A1 (en) 2016-09-06 2023-06-21 F. Hoffmann-La Roche AG 8-(azetidin-1-yl)-[1,2,4]triazolo[1,5-a] pyridinyl compounds, compositions and methods of use thereof
WO2018083085A1 (en) 2016-11-02 2018-05-11 F. Hoffmann-La Roche Ag PYRAZOLO[1,5a]PYRIMIDINE DERIVATIVES AS IRAK4 MODULATORS
WO2018122212A1 (en) 2016-12-29 2018-07-05 F. Hoffmann-La Roche Ag Pyrazolopyrimidine compounds and methods of use thereof
WO2018166993A2 (en) 2017-03-14 2018-09-20 F. Hoffmann-La Roche Ag Pyrazolochlorophenyl compounds, compositions and methods of use thereof
WO2018215389A1 (en) 2017-05-22 2018-11-29 F. Hoffmann-La Roche Ag Therapeutic compounds and compositions, and methods of use thereof
WO2018215390A1 (en) 2017-05-22 2018-11-29 F. Hoffmann-La Roche Ag Therapeutic compounds and compositions, and methods of use thereof
WO2018234343A1 (en) 2017-06-21 2018-12-27 F. Hoffmann-La Roche Ag BENZOFURANES AS MODULATORS OF IRAQ4
WO2018234342A1 (en) 2017-06-21 2018-12-27 F. Hoffmann-La Roche Ag ISOINDOLINONE DERIVATIVES AS MODULATORS OF IRAK4
WO2018234345A1 (en) 2017-06-21 2018-12-27 F. Hoffmann-La Roche Ag PYRAZOLO [1,5A] PYRIMIDINE DERIVATIVES AS MODULATORS OF IRAK4
US11760744B2 (en) 2018-08-16 2023-09-19 Genentech, Inc. Fused ring compounds
US11236068B2 (en) 2018-11-09 2022-02-01 Genentech, Inc. Fused ring compounds
WO2020097537A2 (en) 2018-11-09 2020-05-14 Genentech, Inc. Fused ring compounds
WO2020257145A1 (en) 2019-06-18 2020-12-24 Genentech, Inc. Pyrazolopyrimidine sulfone inhibitors of jak kinases and uses thereof
WO2020257142A1 (en) 2019-06-18 2020-12-24 Genentech, Inc. Tetrazole-substituted pyrazolopyrimidine inhibitors of jak kinases and uses thereof
WO2020257143A1 (en) 2019-06-18 2020-12-24 Genentech, Inc. Pyrazolopyrimidine aryl ether inhibitors of jak kinases and uses thereof
WO2022035790A1 (en) 2020-08-12 2022-02-17 Genentech, Inc. Synthesis of quinazoline compounds
WO2023150653A1 (en) 2022-02-07 2023-08-10 Genentech, Inc. Process for synthesis of quinazoline compounds

Also Published As

Publication number Publication date
CA2727036C (en) 2017-03-21
PE20110545A1 (es) 2011-08-18
MX2010014005A (es) 2011-02-15
CA2727036A1 (en) 2009-12-23
RU2011101898A (ru) 2012-07-27
US20140038939A1 (en) 2014-02-06
AU2009259853A1 (en) 2009-12-23
EP2288260A4 (en) 2013-10-23
US8609687B2 (en) 2013-12-17
BRPI0910021A2 (pt) 2015-09-01
RU2560153C2 (ru) 2015-08-20
CN102131389A (zh) 2011-07-20
IL209763A0 (en) 2011-02-28
JP2011525192A (ja) 2011-09-15
EP2288260A1 (en) 2011-03-02
KR20110033223A (ko) 2011-03-30
ZA201008957B (en) 2014-03-26
JP5512665B2 (ja) 2014-06-04
US20120225855A1 (en) 2012-09-06
US20160333011A1 (en) 2016-11-17
WO2009155551A1 (en) 2009-12-23
SG178812A1 (en) 2012-03-29
US9434732B2 (en) 2016-09-06

Similar Documents

Publication Publication Date Title
US9434732B2 (en) Triazolopyridine JAK inhibitor compounds and methods
US8889673B2 (en) Triazolopyridine JAK inhibitor compounds and methods
US8637526B2 (en) Pyrazolopyrimidine JAK inhibitor compounds and methods
US20240150364A1 (en) Pyrazolopyrimidine jak inhibitor compounds and methods
JP5763057B2 (ja) ヤーヌスキナーゼ阻害剤化合物および方法

Legal Events

Date Code Title Description
AS Assignment

Owner name: GENENTECH, INC.,CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:ZHU, BING-YAN;SIU, MICHAEL;MAGNUSON, STEVEN R.;AND OTHERS;SIGNING DATES FROM 20091008 TO 20091013;REEL/FRAME:023433/0713

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION