WO2011112662A1 - Piperidin-4-yl azetidine derivatives as jak1 inhibitors - Google Patents

Piperidin-4-yl azetidine derivatives as jak1 inhibitors Download PDF

Info

Publication number
WO2011112662A1
WO2011112662A1 PCT/US2011/027665 US2011027665W WO2011112662A1 WO 2011112662 A1 WO2011112662 A1 WO 2011112662A1 US 2011027665 W US2011027665 W US 2011027665W WO 2011112662 A1 WO2011112662 A1 WO 2011112662A1
Authority
WO
WIPO (PCT)
Prior art keywords
pyrrolo
pyrazol
pyrimidin
azetidin
piperidin
Prior art date
Application number
PCT/US2011/027665
Other languages
French (fr)
Inventor
Taisheng Huang
Chu-Biao Xue
Anlai Wang
Ling Kong
Hai Fen Ye
Wenqing Yao
James D. Rodgers
Stacey Shepard
Haisheng Wang
Lixin Shao
Hui-Yin Li
Qun Li
Original Assignee
Incyte Corporation
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=44065466&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2011112662(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to SI201130747A priority Critical patent/SI2545045T1/en
Priority to MX2016001708A priority patent/MX347851B/en
Priority to KR1020127026216A priority patent/KR101857680B1/en
Priority to MX2012010344A priority patent/MX336932B/en
Priority to PL17211097T priority patent/PL3354652T3/en
Priority to MEP-2016-41A priority patent/ME02386B/en
Priority to KR1020217023315A priority patent/KR102354472B1/en
Priority to KR1020207030868A priority patent/KR102283091B1/en
Priority to KR1020227001841A priority patent/KR20220015492A/en
Priority to EP11711709.3A priority patent/EP2545045B1/en
Priority to NZ602313A priority patent/NZ602313A/en
Priority to JP2012557194A priority patent/JP5858484B2/en
Priority to MX2018002077A priority patent/MX364636B/en
Priority to ES11711709T priority patent/ES2569539T3/en
Priority to SG2012064457A priority patent/SG183551A1/en
Priority to BR112012022513A priority patent/BR112012022513A2/en
Priority to DK11711709.3T priority patent/DK2545045T3/en
Priority to KR1020207008092A priority patent/KR102172742B1/en
Priority to MX2017006392A priority patent/MX354212B/en
Priority to AU2011224484A priority patent/AU2011224484A1/en
Priority to RS20160205A priority patent/RS54823B1/en
Application filed by Incyte Corporation filed Critical Incyte Corporation
Priority to KR1020187013055A priority patent/KR101911697B1/en
Priority to EA201290894A priority patent/EA030376B1/en
Priority to EP20158754.0A priority patent/EP3715347B1/en
Priority to PL11711709T priority patent/PL2545045T3/en
Priority to KR1020187030015A priority patent/KR20180117206A/en
Priority to EP17211097.5A priority patent/EP3354652B1/en
Priority to EP21213769.9A priority patent/EP4036088B1/en
Priority to CN201180023757.7A priority patent/CN102985417B/en
Priority to CA2792508A priority patent/CA2792508C/en
Priority to MYPI2012003994A priority patent/MY175156A/en
Priority to UAA201211696A priority patent/UA110108C2/en
Publication of WO2011112662A1 publication Critical patent/WO2011112662A1/en
Priority to IL221823A priority patent/IL221823A/en
Priority to ZA2012/07420A priority patent/ZA201207420B/en
Priority to HK13104880.0A priority patent/HK1177741A1/en
Priority to AU2015205858A priority patent/AU2015205858B2/en
Priority to PH12015502575A priority patent/PH12015502575B1/en
Priority to SM201600085T priority patent/SMT201600085B/en
Priority to HRP20160326TT priority patent/HRP20160326T1/en
Priority to AU2017201801A priority patent/AU2017201801B2/en
Priority to AU2019257385A priority patent/AU2019257385B2/en
Priority to CY20201100671T priority patent/CY1123175T1/en
Priority to AU2021212085A priority patent/AU2021212085B2/en

Links

Classifications

    • 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
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/4545Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring hetero atom, e.g. pipamperone, anabasine
    • 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
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0014Skin, i.e. galenical aspects of topical compositions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • A61K9/0024Solid, semi-solid or solidifying implants, which are implanted or injected in body tissue
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0048Eye, e.g. artificial tears
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • 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
    • 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
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • 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
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • 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
    • 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
    • C07D401/14Heterocyclic 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 three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/13Crystalline forms, e.g. polymorphs
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00

Definitions

  • the present invention provides piperidin-4-yl azetidine derivatives, as well as their compositions and methods of use, that modulate the activity of Janus kinase 1 (JAK1) and are useful in the treatment of diseases related to the activity of JAK1 including, for example, inflammatory disorders, autoimmune disorders, cancer, and other diseases.
  • JAK1 Janus kinase 1
  • Protein kinases regulate diverse biological processes including cell growth, survival, differentiation, organ formation, morphogenesis, neovascularization, tissue repair, and regeneration, among others. Protein kinases also play specialized roles in a host of human diseases including cancer. Cytokines, low-molecular weight polypeptides or glycoproteins, regulate many pathways involved in the host inflammatory response to sepsis. Cytokines influence cell differentiation, proliferation and activation, and can modulate both pro-inflammatory and anti-inflammatory responses to allow the host to react appropriately to pathogens.
  • JAKs Janus kinase family
  • JAK2 Janus kinase- 1
  • JAK2 JAK2
  • JAK3 also known as Janus kinase, leukocyte
  • JAKL protein-tyrosine kinase 2
  • TYK2 protein-tyrosine kinase 2
  • autoimmune diseases e.g., asthma, systemic lupus erythematosus, thyroiditis, myocarditis
  • illnesses such as scleroderma and osteoarthritis (Ortmann, R. A., T. Cheng, et al. (2000) Arthritis Res 2(1): 16-32).
  • JAKs Deficiencies in expression of JAKs are associated with many disease states. For example, Jakl-/- mice are runted at birth, fail to nurse, and die perinatally (Rodig, S. J., M. A. Meraz, et al. (1998) Cell 93(3): 373-83). Jak2-/- mouse embryos are anemic and die around day 12.5 postcoitum due to the absence of definitive erythropoiesis.
  • the JAK/STAT pathway and in particular all four JAKs, are believed to play a role in the pathogenesis of asthmatic response, chronic obstructive pulmonary disease, bronchitis, and other related inflammatory diseases of the lower respiratory tract.
  • cytokines that signal through JAKs have been linked to inflammatory diseases/conditions of the upper respiratory tract, such as those affecting the nose and sinuses ⁇ e.g., rhinitis and sinusitis) whether classically allergic reactions or not.
  • the JAK/STAT pathway has also been implicated in inflammatory diseases/conditions of the eye and chronic allergic responses.
  • JAK STAT Activation of JAK STAT in cancers may occur by cytokine stimulation ⁇ e.g. IL-6 or GM-CSF) or by a reduction in the endogenous suppressors of JAK signaling such as SOCS (suppressor or cytokine signaling) or PIAS (protein inhibitor of activated STAT) (Boudny, V., and Kovarik, J., Neoplasm. 49:349-355, 2002).
  • SOCS suppressor or cytokine signaling
  • PIAS protein inhibitor of activated STAT
  • Activation of STAT signaling, as well as other pathways downstream of JAKs ⁇ e.g., Akt has been correlated with poor prognosis in many cancer types (Bowman, T., et al. Oncogene 19:2474-2488, 2000). Elevated levels of circulating cytokines that signal through JAK/STAT play a causal role in cachexia and/or chronic fatigue. As such, JAK inhibition may be beneficial
  • JAK2 tyrosine kinase can be beneficial for patients with myeloproliferative disorders, e.g., polycythemia vera (PV), essential thrombocythemia (ET), myeloid metaplasia with myelofibrosis (MMM) (Levin, et al, Cancer Cell, vol. 7, 2005: 387- 397).
  • PV polycythemia vera
  • ET essential thrombocythemia
  • MMM myeloid metaplasia with myelofibrosis
  • JAKs may benefit patients suffering from skin immune disorders such as psoriasis, and skin sensitization.
  • skin immune disorders such as psoriasis, and skin sensitization.
  • the maintenance of psoriasis is believed to depend on a number of inflammatory cytokines in addition to various chemokines and growth factors (JCI, 113: 1664-1675), many of which signal through JAKs ⁇ Adv
  • new or improved agents which inhibit kinases such as JAKs are continually needed for developing new and more effective pharmaceuticals that are aimed at augmentation or suppression of the immune and inflammatory pathways (such as immunosuppressive agents for organ transplants), as well as agents for the prevention and treatment of autoimmune diseases, diseases involving a hyperactive inflammatory response (e.g., eczema), allergies, cancer (e.g., prostate, leukemia, multiple myeloma), and some immune reactions (e.g. , skin rash or contact dermatitis or diarrhea) caused by other therapeutics.
  • the compounds of the invention, as well as its compositions and methods described herein are directed toward these needs and other ends.
  • the present invention provides, inter alia, compounds of Formula (I):
  • the present invention further provides compositions comprising a compound of Formula I, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
  • the present invention further provides methods of modulating an activity of JAK1 comprising contacting JAK1 with a compound of Formula I, or a pharmaceutically acceptable salt thereof.
  • the present invention further provides methods of treating a disease or a disorder associated with abnormal kinase expression or activity in a patient by administering to a patient a therapeutically effective amount of a compound of Formula I, or a
  • the present invention further provides methods of treating an autoimmune disease, a cancer, a myeloproliferative disorder, an inflammatory disease, a bone resorption disease, or organ transplant rejection in a patient in need thereof, comprising administering to said patient a therapeutically effective amount of a compound of Formula I, or a pharmaceutically acceptable salt thereof.
  • the present invention also provides compounds of Formula I, or pharmaceutically acceptable salts thereof, as described herein for use in treatment of autoimmune diseases, cancer, myeloproliferative disorders, inflammatory diseases, a bone resorption disease, or organ transplant rejection.
  • the present invention further provides compounds of Formula I as described herein, or pharmaceutically acceptable salts thereof, for use in modulating JAK1.
  • the present invention also provides uses of compounds of Formula I as described herein, or pharmaceutically acceptable salts thereof, for the preparation of medicaments for use in methods of modulating JAK1.
  • FIG. 1 depicts the DSC thermogram for the product of Example 358.
  • FIG. 2 depicts the TGA thermogram for the product of Example 358.
  • FIG. 3 depicts the XRPD pattern for the product of Example 358.
  • the present invention provides, inter alia, a compound of Formula (I):
  • X is N or CR 2 ;
  • Y is N or CR 3 ;
  • Z is H, cyano, halo, Ci_ 3 alkyl, or Ci_ 3 haloalkyl;
  • each R 1 is, independently, Ci_ 4 alkyl, hydroxyl, Ci_ 4 alkoxy, fluoro, hydroxyl-Ci_ 4 alkyl, or Ci_ 4 alkoxy-Ci_ 4 alkyl; or
  • R 2 is H, halo, hydroxyl, cyano, Ci_ 4 alkyl, Ci_ 4 haloalkyl, or Ci_ 4 alkoxy;
  • R 3 is H, cyano, nitro, halo, hydroxyl, Ci_ 6 alkyl, C 2 _ 6 alkenyl, C 2 _ 6 alkynyl, C 3 _ 7 cycloalkyl, Ci_ 6 haloalkyl, Ci_ 6 alkoxy, Ci_ 6 haloalkoxy, amino, Ci_ 6 alkylamino, di-Ci_ 6 alkylamino, Ci_ 6 alkylsulfonyl, carbamyl, Ci_ 6 alkylcarbamyl, di-Ci_ 6 alkylcarbamyl, carboxy, Ci_ 6 alkylcarbonyl, or Ci_ 6 alkoxycarbonyl;
  • each R 4 is, independently, H or Ci_ 4 alkyl; or two R 4 groups, together with the carbon atom to which they are attached, form a 3-, 4-, 5-, or 6-membered cycloalkyl ring;
  • R 4a is H or Ci_4 alkyl
  • each R 4b is, independently, H or Ci_ 4 alkyl
  • each R 5 is, independently, halo, cyano, nitro, Ci_ 6 alkyl, C 2 _ 6 alkenyl, C 2 _ 6 alkynyl, Ci_6 haloalkyl, halosulfanyl, C3-10 cycloalkyl, C3-10 cycloalkyl-Ci_3 alkyl, C2-10
  • heterocycloalkyl C 2 _io heterocycloalkyl-Ci_3 alkyl, C 6 -io aryl, C 6 -io aryl-Ci_3 alkyl, Ci_io heteroaryl, C M0 heteroaryl-Ci_ 3 alkyl, OR a , SR a , C(0)R b , C(0)NR c R d , C(0)OR a ,
  • each R 6 is, independently, halo, cyano, nitro, Ci_ 6 alkyl, C 2 _ 6 alkenyl, C 2 _ 6 alkynyl, Ci_6 haloalkyl, halosulfanyl, C 3 _i 0 cycloalkyl, C 3 _i 0 cycloalkyl-Ci_ 3 alkyl, C 2 _i 0
  • each R a , R b , R c , and R d is, independently, H, Ci_ 6 alkyl, C 2 _ 6 alkenyl, C 2 _ 6 alkynyl, Ci_6 haloalkyl, C 3-10 cycloalkyl, C 3-10 cycloalkyl-Ci_3 alkyl, C 2 _io heterocycloalkyl, C 2 _io heterocycloalkyl-Ci_3 alkyl, C 6 _io aryl, C 6 _io aryl-Ci_ 3 alkyl, C 1-10 heteroaryl, or C 1-10 heteroaryl-Ci_3 alkyl; wherein said Ci_ 6 alkyl, C 2 _ 6 alkenyl, C 2 _ 6 alkynyl, Ci_ 6 haloalkyl, halosulfanyl, C 3 _i 0 cycloalkyl, C 3 _i 0 cycloalkyl-C
  • R c and R d together with the N atom to which they are attached form a 3-, 4- , 5-, 6-, or 7-membered heterocycloalkyl group or a heteroaryl group, each optionally substituted with 1 , 2, 3 or 4 substituents independently selected from halo, cyano, nitro, hydroxyl, Ci_ 6 alkyl, C 2 _ 6 alkenyl, C 2 _ 6 alkynyl, C 3 _ 7 cycloalkyl, Ci_ 6 haloalkyl, Ci_ 6 alkoxy, Ci_ 6 haloalkoxy, amino, Ci_ 6 alkylamino, di-Ci_ 6 alkylamino, thio, Ci_ 6 alkylthio, Ci_6 alkylsulfinyl, Ci_ 6 alkylsulfonyl, carbamyl, Ci_ 6 alkylcarbamyl, di-Ci_ 6 alkylcarbamyl, carboxy, Ci_ 6 alkyl
  • each R e is, independently, H, Ci_ 6 alkyl, CN, hydroxyl, Ci_ 6 alkoxy, Ci_ 6 alkylsulfonyl, carboxy, Ci_ 6 alkylcarbonyl, amino sulfonyl, Ci_ 6 alkylaminosulfonyl, di-Ci_ 6 alkylaminosulfonyl, carbamyl, Ci_ 6 alkylcarbamyl, or di-Ci_ 6 alkylcarbamyl;
  • each R al , R bl , R cl , and R dl is, independently, H, Ci_ 6 alkyl, C 2 _ 6 alkenyl, C 2 _ 6 alkynyl, Ci_ 6 haloalkyl, C 3 _io cycloalkyl, C 3 _io cycloalkyl-Ci_ 3 alkyl, C 2 _io heterocycloalkyl, C 2 _io heterocycloalkyl-Ci_ 3 alkyl, C 6 _io aryl, C 6 _io aryl-Ci_ 3 alkyl, Ci_i 0 heteroaryl, or Ci_i 0 heteroaryl-Ci_ 3 alkyl; wherein said Ci_ 6 alkyl, C 2 _ 6 alkenyl, C 2 _ 6 alkynyl, Ci_ 6 haloalkyl, halosulfanyl, C 3 _io cycloalkyl, C 3 _
  • R cl and R dl together with the N atom to which they are attached form a 3-, 4-, 5-, 6-, or 7-membered heterocycloalkyl group or a heteroaryl group, each optionally substituted with 1 , 2, 3 or 4 substituents independently selected from halo, cyano, nitro, hydroxyl, Ci_ 6 alkyl, C 2 _ 6 alkenyl, C 2 _ 6 alkynyl, C 3 _ 7 cycloalkyl, Ci_ 6 haloalkyl, Ci_ 6 alkoxy, Ci_ 6 haloalkoxy, amino, Ci_ 6 alkylamino, di-Ci_ 6 alkylamino, thio, Ci_ 6 alkylthio, Ci_6 alkylsulfinyl, Ci_ 6 alkylsulfonyl, carbamyl, Ci_ 6 alkylcarbamyl, di-Ci_ 6 alkylcarbamyl, carboxy, Ci_ 6 alkyl
  • each R g , R g , and R h is, independently, halo, cyano, nitro, hydroxyl, Ci_ 6 alkyl, C 2 _ 6 alkenyl, C 2 _ 6 alkynyl, C3-7 cycloalkyl, Ci_ 6 haloalkyl, Ci_ 6 alkoxy, Ci_ 6 haloalkoxy, amino, Ci_6 alkylamino, di-Ci_ 6 alkylamino, thio, Ci_ 6 alkylthio, Ci_ 6 alkylsulfmyl, Ci_ 6 alkylsulfonyl, carbamyl, Ci_ 6 alkylcarbamyl, di-Ci_ 6 alkylcarbamyl, carboxy, Ci_ 6 alkylcarbonyl, Ci_ 6 alkoxycarbonyl, or Ci_ 6 alkylcarbonylamino; and
  • n 0, 1, 2, 3, or 4.
  • X is N or CR 2 ;
  • Y is N or CR 3 ;
  • Z is H, cyano, halo, Ci_ 3 alkyl, or Ci_ 3 haloalkyl;
  • each R 1 is, independently, Ci_ 4 alkyl
  • R 2 is H, halo, hydroxyl, cyano, Ci_ 4 alkyl, Ci_ 4 haloalkyl, or Ci_ 4 alkoxy;
  • R 3 is H, cyano, nitro, halo, hydroxyl, Ci_ 6 alkyl, C 2 _ 6 alkenyl, C 2 _ 6 alkynyl, C 3 _ 7 cycloalkyl, Ci_ 6 haloalkyl, Ci_ 6 alkoxy, Ci_ 6 haloalkoxy, amino, Ci_ 6 alkylamino, di-Ci_ 6 alkylamino, Ci_ 6 alkylsulfonyl, carbamyl, Ci_ 6 alkylcarbamyl, di-Ci_ 6 alkylcarbamyl, carboxy, Ci_ 6 alkylcarbonyl, or Ci_ 6 alkoxycarbonyl;
  • each R 4 is, independently, H or Ci_ 4 alkyl
  • R 4a is H or Ci_ 4 alkyl
  • each R 4b is, independently, H or Ci_ 4 alkyl; or two R groups, together with the carbon atom to which they are attached, form a 3-, 4-, 5-, or 6-membered cycloalkyl ring;
  • each R 5 is, independently, halo, cyano, nitro, Ci_ 6 alkyl, C 2 _ 6 alkenyl, C 2 _ 6 alkynyl, Ci_6 haloalkyl, halosulfanyl, C3-10 cycloalkyl, C3-10 cycloalkyl-Ci_3 alkyl, C2-10
  • heterocycloalkyl C 2 _io heterocycloalkyl-Ci_3 alkyl, C 6 -io aryl, C 6 -io aryl-Ci_3 alkyl, Ci_io heteroaryl, C M0 heteroaryl-Ci_ 3 alkyl, OR a , SR a , C(0)R b , C(0)NR c R d , C(0)OR a ,
  • each R 6 is, independently, halo, cyano, nitro, Ci_ 6 alkyl, C 2 _ 6 alkenyl, C 2 _ 6 alkynyl, Ci_6 haloalkyl, halosulfanyl, C3-10 cycloalkyl, C3-10 cycloalkyl-Ci_3 alkyl, C 2 _io
  • each R a , R b , R c , and R d is, independently, H, Ci_ 6 alkyl, C 2 _ 6 alkenyl, C 2 _ 6 alkynyl, Ci_6 haloalkyl, C 3-10 cycloalkyl, C 3-10 cycloalkyl-C 1-3 alkyl, C 2 _io heterocycloalkyl, C 2 _io heterocycloalkyl-Ci_3 alkyl, C 6 _io aryl, C 6 _io aryl-Ci_ 3 alkyl, C 1-10 heteroaryl, or C 1-10 heteroaryl-Ci_3 alkyl; wherein said Ci_ 6 alkyl, C 2 _ 6 alkenyl, C 2 _ 6 alkynyl, Ci_ 6 haloalkyl, halosulfanyl, C 3-10 cycloalkyl, C 3-10 cycloalkyl-C 1-3 alkyl, C 2
  • R c and R d together with the N atom to which they are attached form a 3-, 4- , 5-, 6-, or 7-membered heterocycloalkyl group or a heteroaryl group, each optionally substituted with 1 , 2, 3 or 4 substituents independently selected from halo, cyano, nitro, hydroxyl, Ci_ 6 alkyl, C 2 _6 alkenyl, C 2 _ 6 alkynyl, C 3 _ 7 cycloalkyl, Ci_ 6 haloalkyl, Ci_ 6 alkoxy, Ci_ 6 haloalkoxy, amino, Ci_ 6 alkylamino, di-Ci_ 6 alkylamino, thio, Ci_ 6 alkylthio, Ci_6 alkylsulfinyl, Ci_ 6 alkylsulfonyl, carbamyl, Ci_ 6 alkylcarbamyl, di-Ci_ 6 alkylcarbamyl, carboxy, Ci_ 6 alkyl
  • each R e is, independently, H, Ci_ 6 alkyl, CN, hydroxyl, Ci_ 6 alkoxy, Ci_ 6 alkylsulfonyl, carboxy, Ci_ 6 alkylcarbonyl, amino sulfonyl, Ci_ 6 alkylaminosulfonyl, di-Ci_ 6 alkylaminosulfonyl, carbamyl, Ci_ 6 alkylcarbamyl, or di-Ci_ 6 alkylcarbamyl;
  • each R al , R bl , R cl , and R dl is, independently, H, Ci_ 6 alkyl, C 2 _ 6 alkenyl, C 2 _ 6 alkynyl, Ci_ 6 haloalkyl, C 3 _i 0 cycloalkyl, C 3 _i 0 cycloalkyl-Ci_ 3 alkyl, C 2 _i 0 heterocycloalkyl, C 2 _io heterocycloalkyl-Ci_ 3 alkyl, C 6 -io aryl, C 6 -io aryl-Ci_ 3 alkyl, Ci_io heteroaryl, or Ci_io heteroaryl-Ci_ 3 alkyl; wherein said Ci_ 6 alkyl, C 2 _ 6 alkenyl, C 2 _ 6 alkynyl, Ci_ 6 haloalkyl, halosulfanyl, C 3 _io cycloalkyl, C 3
  • R cl and R dl together with the N atom to which they are attached form a 3-, 4-, 5-, 6-, or 7-membered heterocycloalkyl group or a heteroaryl group, each optionally substituted with 1 , 2, 3 or 4 substituents independently selected from halo, cyano, nitro, hydroxyl, Ci_ 6 alkyl, C 2 _ 6 alkenyl, C 2 _ 6 alkynyl, C 3 _ 7 cycloalkyl, Ci_ 6 haloalkyl, Ci_ 6 alkoxy, Ci_ 6 haloalkoxy, amino, Ci_ 6 alkylamino, di-Ci_ 6 alkylamino, thio, Ci_ 6 alkylthio, Ci_6 alkylsulfinyl, Ci_ 6 alkylsulfonyl, carbamyl, Ci_ 6 alkylcarbamyl, di-Ci_ 6 alkylcarbamyl, carboxy, Ci_ 6 alkyl
  • n 0, 1, 2, 3, or 4.
  • X is N.
  • X is CR 2 .
  • X is C(H), C(F), or C(CN).
  • X is CH.
  • Y is N.
  • Y is CR 3 .
  • Y is CH.
  • Z is cyano
  • L is C(R 4 ) 2 .
  • L is CH 2 .
  • n 0, 1, or 2.
  • n 0.
  • n 1
  • n is 2.
  • R 1 is Ci_ 4 alkyl.
  • R 1 is methyl
  • two R 1 groups form a 2-carbon bridge.
  • A is C 6-14 aryl, which is optionally substituted with 1, 2, 3, 4, 5, or 6 independently selected R 5 groups.
  • A is phenyl, which is optionally substituted with 1, 2, 3, 4, or 5 independently selected R 5 groups.
  • A is monocyclic C 3 _g cycloalkyl or bicyclic C 3 _g cycloalkyl, each of which is optionally substituted with 1, 2, 3, 4, 5, or 6 independently selected R 5 groups.
  • A is monocyclic C 2-10 heterocycloalkyl or bicyclic C 2-10 heterocycloalkyl, each of which is optionally substituted with 1, 2, 3, 4, 5, or 6 independently selected R 5 groups.
  • A is monocyclic C 1-10 heteroaryl or bicyclic C 1-10 heteroaryl, each of which is optionally substituted with 1, 2, 3, 4, 5, or 6 independently selected R 5 groups.
  • A is Ci_ 6 alkyl, which is optionally substituted with 1, 2, 3, 4, 5, or 6 independently selected R 5 groups.
  • A is Ci_ 6 alkyl.
  • A is Ci_ 6 alkyl, phenyl, a naphthyl ring, monocyclic or bicyclic C 3 _i 0 cycloalkyl, monocyclic or bicyclic C 2-10 heterocycloalkyl, or monocyclic or bicyclic C 1-10 heteroaryl; each of which is optionally substituted with 1, 2, 3, 4, or 5 independently selected R 5 groups.
  • A is phenyl, a naphthyl ring, monocyclic C 3 _io cycloalkyl, bicyclic C 3 _i 0 cycloalkyl, monocyclic C 2-10 heterocycloalkyl, bicyclic C 2-10 heterocycloalkyl, monocyclic C 1-10 heteroaryl, or bicyclic C 1-10 heteroaryl; each of which is optionally substituted with 1, 2, 3, 4, or 5 independently selected R 5 groups.
  • A is methyl, ethyl, isopropyl, phenyl, a naphthalene ring, a pyridine ring, a pyrimidine ring, a thiophene ring, a pyrazine ring, an oxazole ring, an isoxazole ring, an imidazole ring, a thiazole ring, a furan ring, a pyrazole ring, a quinoline ring, a benzothiophene ring, a benzothiazole ring, a benzoimidazole ring, a benzofuran ring, cyclopropyl, cyclopentyl, cyclohexyl, cycloheptyl, an indene ring, a tetrahydronaphthalene ring, dihydro-l,4-benzodioxoxine ring, or a piperidine ring
  • A is phenyl, a naphthalene ring, a pyridine ring, a pyrimidine ring, a thiophene ring, a pyrazine ring, an oxazole ring, an isoxazole ring, an imidazole ring, a thiazole ring, a furan ring, a pyrazole ring, a quinoline ring, a benzothiophene ring, a benzothiazole ring, a benzoimidazole ring, a benzofuran ring, cyclopropyl, cyclopentyl, cyclohexyl, cycloheptyl, an indene ring, a
  • tetrahydronaphthalene ring dihydro-l,4-benzodioxoxine ring, or a piperidine ring; each of which is optionally substituted with 1, 2, 3, 4, or 5 independently selected R 5 groups, as permitted by valency.
  • A is phenyl or a pyridine ring; each of which are optionally substituted with 1, 2, 3, or 4 independently selected R 5 groups.
  • A is pyridin-4-yl; which is optionally substituted with 1, 2, 3, or 4 independently selected R 5 groups.
  • A is a pyridine ring; which is optionally substituted with 1 , 2, 3, or 4 independently selected R 5 groups.
  • each R 5 is, independently, halo, cyano, Ci_ 6 alkyl, C 2 _ 6 alkenyl, C 2 _ 6 alkynyl, Ci_ 6 haloalkyl, C 3-10 cycloalkyl, C 3-10 cycloalkyl-Ci_3 alkyl, C 2 _io heterocycloalkyl, C 2 _i 0 heterocycloalkyl-Ci_ 3 alkyl, C 6 _io aryl, C 6 _io aryl-Ci_ 3 alkyl, C 1-10 heteroaryl, Ci_i 0 heteroaryl-Ci_ 3 alkyl, OR a , SR a , C(0)R b , C(0)NR c R d , C(0)OR a , OC(0)R b , OC(0)NR c R d , NR c R d , NR c C(0)R b , NR c C(0)OR a ,
  • each R 5 is, independently, halo, cyano, Ci_ 6 alkyl, Ci_ 6 haloalkyl, C 3 _i 0 cycloalkyl, C 2 _io heterocycloalkyl, C 6 _io aryl, C 6 _io aryl-Ci_ 3 alkyl, Ci_i 0 heteroaryl, OR a , SR a , C(0)R b , C(0)NR c R d , C(0)OR a , OC(0)R b , OC(0)NR c R d , NR c R d , NR c C(0)R b , NR c C(0)OR a , NR c C(0)NR c R d , NR c S(0) 2 R b , NR c S(0) 2 NR c R d , S(0) 2 R b , or S(0)2NR c R d ; wherein Ci_ 6 alkyl, Ci_
  • heterocycloalkyl-Ci_3 alkyl, C 6 -io aryl, C 6 -io aryl-Ci_3 alkyl, Ci_io heteroaryl, and Ci_io heteroaryl-Ci_3 alkyl are each optionally substituted with 1, 2, 3, 4, or 5 independently selected R 6 groups.
  • each R 5 is, independently, halo, cyano, Ci_ 6 alkyl, Ci_ 6 haloalkyl, C 6 -i 0 aryl, C M0 heteroaryl, OR a , SR a , C(0)OR a , NR c R d , or NR c C(0)R b ;
  • Ci_ 6 alkyl, Ci_ 6 haloalkyl, C 6 -io aryl, and Ci_io heteroaryl are each optionally substituted with 1, 2, 3, 4, or 5 independently selected R 6 groups.
  • each R 5 is, independently, halo, cyano, Ci_ 6 alkyl, Ci_ 6 haloalkyl, C 6 _i 0 aryl, C M0 heteroaryl, OR a , SR a , C(0)OR a , NR c R d , or NR c C(0)R b ;
  • Ci_ 6 alkyl, Ci_ 6 haloalkyl, C 6 -io aryl, and Ci_io heteroaryl are each optionally substituted with 1, 2, 3, 4, or 5 independently selected R 6 groups;
  • each R a , R b , R c , and R d is, independently, H, Ci_ 6 alkyl, Ci_ 6 haloalkyl, or C 6 -io aryl; wherein said Ci_ 6 alkyl and C 6 _io aryl is optionally substituted with 1, 2, 3, 4, or 5 independently selected R g groups.
  • each R 5 is, independently, halo, cyano, Ci_ 6 alkyl, Ci_ 6 haloalkyl, hydroxyl, Ci_ 6 alkoxy, Ci_ 6 haloalkoxy, C 6 -i2 aryloxy, amino, Ci_ 6 alkylamino, di-Ci_6 alkylamino, Ci_ 6 alkylcarbonylamino, Ci_ 6 alkoxycarbonyl, Ci_ 6 alkylthio, C 6 -i2 aryl, or Ci_9 heteroaryl; wherein said C 6 -i2 aryl or Ci_9 heteroaryl are each optionally substituted with 1, 2, 3, 4, or 5 independently selected R 6 groups.
  • each R 5 is, independently, chloro, fluoro, bromo, cyano, methyl, ethyl, trifluoromethyl, hydroxyl, methoxy, trifluoromethoxy, difluoromethoxy, phenoxy, dimethylamino, t-butylcarbonylamino, methoxycarbonyl, methylthio, phenyl, a pyridine ring, a thiazole ring, a quinoline ring, an isoquinoline ring, an imidazo[l ,2- ajpyrimidine ring, a benzoxazole ring, or an oxadiazole ring; wherein said phenyl, pyridine ring, thiazole ring, quinoline ring, isoquinoline ring, imidazo[l ,2-a]pyrimidine ring, benzoxazole ring, and oxadiazole ring are each optionally substituted with 1 , 2, 3, 4,
  • each R 5 is, independently, halo or Ci_ 6 haloalkyl.
  • each R 5 is, independently, fluoro or trifluoromethyl.
  • each R 6 is, independently, halo, cyano, nitro, hydroxyl, Ci_ 6 alkyl, C 2 _ 6 alkenyl, C 2 _ 6 alkynyl, C 3 _ 7 cycloalkyl, Ci_ 6 haloalkyl, Ci_ 6 alkoxy, Ci_ 6 haloalkoxy, amino, Ci_ 6 alkylamino, di-Ci_ 6 alkylamino, thio, Ci_ 6 alkylthio, Ci_ 6 alkylsulfinyl, Ci_ 6 alkylsulfonyl, carbamyl, Ci_ 6 alkylcarbamyl, di-Ci_ 6 alkylcarbamyl, carboxy, Ci_ 6 alkylcarbonyl, Ci_ 6 alkoxycarbonyl, or Ci_ 6 alkylcarbonylamino.
  • each R 6 is, independently, halo, cyano, Ci_ 6 alkyl, Ci_ 6 haloalkyl, OR al , NR cl R dl , or OC(0)R bl ;
  • each R al , R bl , R cl , and R dl is, independently, H or Ci_ 6 alkyl; wherein said Ci_ 6 alkyl is optionally substituted with a substituent independently selected from Ci_ 4 alkoxy and hydroxyl;
  • R cl and R dl together with the N atom to which they are attached form a 4-, 5-, 6-, or 7-membered heterocycloalkyl group, optionally substituted with 1 , 2, 3 or 4 substituents independently selected from halo.
  • each R 5 is, independently, halo, cyano, Ci_ 6 alkyl, Ci_ 6 haloalkyl, C 6 _i 0 aryl, Ci_i 0 heteroaryl, OR a , SR a , C(0)OR a , NR c R d , or NR c C(0)R b ;
  • Ci_ 6 alkyl, Ci_ 6 haloalkyl, C 6 -io aryl, and Ci_io heteroaryl are each optionally substituted with 1 , 2, 3, 4, or 5 independently selected R 6 groups;
  • each R a , R b , R c , and R d is, independently, H, Ci_ 6 alkyl, Ci_ 6 haloalkyl, or C 6 -io aryl; wherein said Ci_ 6 alkyl and C 6 _io aryl is optionally substituted with 1 , 2, 3, 4, or 5 independently selected R g groups;
  • each R 6 is, independently, halo, cyano, Ci_ 6 alkyl, Ci_ 6 haloalkyl, OR al , NR cl R dl , or OC(0)R bl ; and each R al , R bl , R cl , and R dl is, independently, H or Ci_ 6 alkyl; wherein said Ci_ 6 alkyl is optionally substituted with a substituent independently selected from Ci_ 4 alkoxy and hydroxyl;
  • R cl and R dl together with the N atom to which they are attached form a 4-, 5-, 6-, or 7-membered heterocycloalkyl group, optionally substituted with 1 , 2, 3 or 4 substituents independently selected from halo.
  • each R 6 is, independently, halo, cyano, or Ci_ 6 alkyl.
  • each R 6 is, independently, chloro, fluoro, cyano, or methyl.
  • R 2 is H, halo, or cyano.
  • R 2 is H, F, or cyano.
  • R 3 is H.
  • the compound is a compound of Formula (II):
  • the compound is a compound of Formula (Ha):
  • the compound is a compound of Formula (III):
  • the compound is a compound of Formula
  • X is N or CR 2 ;
  • Y is N or CR 3 ;
  • each R 1 is, independently, Ci_ 4 alkyl
  • R 2 is H, halo, or cyano
  • R 3 is H
  • Z is cyano
  • A is Ci_6 alkyl, phenyl, a naphthyl ring, monocyclic C3-10 cycloalkyl, bicyclic C3-10 cycloalkyl, monocyclic C 2 _io heterocycloalkyl, bicyclic C 2 _io heterocycloalkyl, monocyclic Ci_io heteroaryl, or bicyclic Ci_io heteroaryl; each of which is optionally substituted with 1, 2, 3, 4, or 5 independently selected R 5 groups;
  • each R 5 is, independently, halo, cyano, Ci_ 6 alkyl, C 2 _ 6 alkenyl, C 2 _ 6 alkynyl, Ci_ 6 haloalkyl, C3-10 cycloalkyl, C3-10 cycloalkyl-Ci_3 alkyl, C 2 _io heterocycloalkyl, C 2 _io heterocycloalkyl-Ci_3 alkyl, C 6 -io aryl, C 6 -io aryl-Ci_3 alkyl, Ci_io heteroaryl, Ci_io heteroaryl-Ci_ 3 alkyl, OR a , SR a , C(0)R b , C(0)NR c R d , C(0)OR a , OC(0)R b , OC(0)NR c R d , NR c R d , NR c C(0)R b , NR c C(0)OR a , NR
  • n 0, 1, or 2.
  • X is N or CR 2 ;
  • Y is N or CR 3 ;
  • each R 1 is, independently, Ci_ 4 alkyl
  • R 2 is H, halo, or cyano
  • R 3 is H
  • Z is cyano
  • A is Ci_6 alkyl, phenyl, a naphthyl ring, monocyclic C 3 _io cycloalkyl, bicyclic C 3 _io cycloalkyl, monocyclic C 2 _i 0 heterocycloalkyl, bicyclic C 2 _i 0 heterocycloalkyl, monocyclic Ci_io heteroaryl, or bicyclic Ci_io heteroaryl; each of which is optionally substituted with 1, 2, 3, 4, or 5 independently selected R 5 groups;
  • each R 5 is, independently, halo, cyano, Ci_ 6 alkyl, Ci_ 6 haloalkyl, C 3 _io cycloalkyl, C 2 _io heterocycloalkyl, C 6 -io aryl, C 6 -io aryl-Ci_ 3 alkyl, Ci_io heteroaryl, OR a , SR a , C(0)R b , C(0)NR c R d , C(0)OR a , OC(0)R b , OC(0)NR c R d , NR c R d , NR c C(0)R b , NR c C(0)OR a , NR c C(0)NR c R d , NR c S(0) 2 R b , NR c S(0) 2 NR c R d , S(0) 2 R b , or S(0) 2 NR c R d ; wherein Ci_ 6 alkyl, C 2
  • n 0, 1, or 2.
  • X is N or CR 2 ;
  • Y is N or CR 3 ;
  • each R 1 is, independently, Ci_ 4 alkyl
  • R 2 is H, halo, or cyano
  • R 3 is H
  • Z is cyano
  • A is phenyl, a naphthyl ring, monocyclic C3-10 cycloalkyl, bicyclic C3-10 cycloalkyl, monocyclic C 2 _i 0 heterocycloalkyl, bicyclic C 2 _i 0 heterocycloalkyl, monocyclic Ci_io heteroaryl, or bicyclic Ci_io heteroaryl; each of which is optionally substituted with 1, 2, 3, 4, or 5 independently selected R 5 groups;
  • each R 5 is, independently, halo, cyano, Ci_ 6 alkyl, Ci_ 6 haloalkyl, C3-10 cycloalkyl, C 2 _io heterocycloalkyl, C 6 _io aryl, C 6 _io aryl-Ci_ 3 alkyl, Ci_i 0 heteroaryl, OR a , SR a , C(0)R b , C(0)NR c R d , C(0)OR a , OC(0)R b , OC(0)NR c R d , NR c R d , NR c C(0)R b , NR c C(0)OR a , NR c C(0)NR c R d , NR c S(0) 2 R b , NR c S(0) 2 NR c R d , S(0) 2 R b , or S(0) 2 NR c R d ; wherein Ci_ 6 alkyl, C 2 _ 6
  • n 0, 1, or 2.
  • X is N or CR 2 ;
  • Y is N or CR 3 ;
  • each R 1 is, independently, Ci_ 4 alkyl
  • R 2 is H, halo, or cyano
  • R 3 is H
  • Z is cyano
  • each R 5 is, independently, halo, cyano, Ci_ 6 alkyl, Ci_ 6 haloalkyl, C 6 -io aryl, C 1-10 heteroaryl, OR a , SR a , C(0)OR a , NR c R d , NR c C(0)R b ; wherein said Ci_6 alkyl, Ci_ 6 haloalkyl, C 6 -io aryl, and C 1-10 heteroaryl are each optionally substituted with 1, 2, 3, 4, or 5 independently selected R 6 groups;
  • n 0, 1, or 2.
  • X is N or CR 2 ;
  • Y is N or CR 3 ;
  • each R 1 is, independently, Ci_ 4 alkyl
  • R 2 is H, halo, or cyano
  • R 3 is H
  • Z is cyano
  • A is phenyl, a naphthyl ring, monocyclic C 3-10 cycloalkyl, bicyclic C 3-10 cycloalkyl, monocyclic C 2 _io heterocycloalkyl, bicyclic C 2 _io heterocycloalkyl, monocyclic C 1-10 heteroaryl, or bicyclic C 1-10 heteroaryl; each of which is optionally substituted with 1, 2, 3, 4, or 5 independently selected R 5 groups;
  • each R 5 is, independently, halo, cyano, Ci_ 6 alkyl, Ci_ 6 haloalkyl, C 6 -io aryl, C 1-10 heteroaryl, OR a , SR a , C(0)OR a , NR c R d , NR c C(0)R b ; wherein said Ci_ 6 alkyl, Ci_ 6 haloalkyl, C 6 _io aryl, and C 1-10 heteroaryl are each optionally substituted with 1, 2, 3, 4, or
  • n 0, 1, or 2.
  • X is N or CR 2 ;
  • Y is N or CR 3 ;
  • each R 1 is, independently, methyl
  • R 2 is H, halo, or cyano
  • R 3 is H
  • Z is cyano
  • A is methyl, ethyl, isopropyl, phenyl, a naphthalene ring, a pyridine ring, a pyrimidine ring, a thiophene ring, a pyrazine ring, an oxazole ring, an isoxazole ring, an imidazole ring, a thiazole ring, a furan ring, a pyrazole ring, a quinoline ring, a benzothiophene ring, a benzothiazole ring, a benzoimidazole ring, a benzofuran ring, cyclopropyl, cyclopentyl, cyclohexyl, cycloheptyl, an indene ring, a
  • tetrahydronaphthalene ring dihydro-l,4-benzodioxoxine ring, or a piperidine ring; each of which are optionally substituted with 1, 2, 3, 4, 5, or 6 independently selected R 5 groups, as permitted by valency;
  • each R 5 is, independently, chloro, fluoro, bromo, cyano, methyl, ethyl, trifluoromethyl, hydroxyl, methoxy, trifluoromethoxy, difluoromethoxy, phenoxy, dimethylamino, t-butylcarbonylamino, methoxycarbonyl, methylthio, phenyl, a pyridine ring, a thiazole ring, a quinoline ring, an isoquinoline ring, an imidazo[l,2-a]pyrimidine ring, a benzoxazole ring, or an oxadiazole ring; wherein said phenyl, pyridine ring, thiazole ring, quinoline ring, isoquinoline ring, imidazo[l,2-a]pyrimidine ring, benzoxazole ring, and oxadiazole ring are each optionally substituted with 1, 2, 3, 4, or 5 independently selected R 6 groups; and
  • n 0, 1, or 2.
  • X is N or CR 2 ;
  • Y is N or CR 3 ;
  • each R 1 is, independently, methyl
  • R 2 is H, halo, or cyano
  • R 3 is H
  • Z is cyano
  • A is phenyl, a naphthalene ring, a pyridine ring, a pyrimidine ring, a thiophene ring, a pyrazine ring, an oxazole ring, an isoxazole ring, an imidazole ring, a thiazole ring, a furan ring, a pyrazole ring, a quinoline ring, a benzothiophene ring, a
  • benzothiazole ring a benzoimidazole ring, a benzofuran ring, cyclopropyl, cyclopentyl, cyclohexyl, cycloheptyl, an indene ring, a tetrahydronaphthalene ring, dihydro-1,4- benzodioxoxine ring, or a piperidine ring; each of which are optionally substituted with 1, 2, 3, 4, 5, or 6 independently selected R 5 groups;
  • each R 5 is, independently, chloro, fluoro, bromo, cyano, methyl, ethyl,
  • n 0, 1, or 2.
  • X is N or CR 2 ;
  • Y is N or CR 3 ;
  • each R 1 is, independently, Ci_ 4 alkyl
  • R 2 is H, halo, or cyano
  • R 3 is H
  • Z is cyano
  • A is Ci_6 alkyl; and n is 0, 1, or 2.
  • X is N or CH
  • Y is N
  • Z is cyano
  • A is phenyl or a pyridine ring, each of which is optionally substituted with 1, 2, 3, or 4 independently selected R 5 groups;
  • each R 5 is, independently, halo or Ci_ 6 haloalkyl
  • n O.
  • X is N or CH
  • Y is N
  • Z is cyano
  • A is phenyl or pyridin-4-yl, each of which is optionally substituted with 1 or 2 independently selected R 5 groups;
  • each R 5 is, independently, fluoro or trifluoromethyl
  • n 0.
  • X is N or CR 2 ;
  • Y is N or CR 3 ;
  • each R 1 is, independently, Ci_ 4 alkyl, hydroxyl, Ci_ 4 alkoxy, or fluoro;
  • R 2 is H, halo, or cyano
  • R 3 is H
  • Z is cyano
  • A is Ci_6 alkyl, phenyl, a naphthyl ring, monocyclic C 3-10 cycloalkyl, bicyclic C 3-10 cycloalkyl, monocyclic C 2-10 heterocycloalkyl, bicyclic C 2-10 heterocycloalkyl, monocyclic C 1-10 heteroaryl, or bicyclic C 1-10 heteroaryl; each of which is optionally substituted with 1, 2, 3, 4, or 5 independently selected R 5 groups;
  • each R 5 is, independently, halo, cyano, Ci_ 6 alkyl, Ci_ 6 haloalkyl, C 6 -io aryl, C 1-10 heteroaryl, OR a , SR a , C(0)OR a , NR c R d , NR c C(0)R b ; wherein said Ci_6 alkyl, Ci_ 6 haloalkyl, C 6 -io aryl, and C 1-10 heteroaryl are each optionally substituted with 1, 2, 3, 4, or 5 independently selected R 6 groups;
  • n 0, 1, or 2;
  • each R 5 is, independently, halo, cyano, Ci_ 6 alkyl, Ci_ 6 haloalkyl, C 6 _io aryl, C 1-10 heteroaryl, OR a , SR a , C(0)OR a , NR c R d , or NR c C(0)R b ; wherein said Ci_ 6 alkyl, Ci_ 6 haloalkyl, C 6 -io aryl, and C 1-10 heteroaryl are each optionally substituted with 1, 2, 3, 4, or 5 independently selected R 6 groups;
  • each R a , R b , R c , and R d is, independently, H, Ci_ 6 alkyl, Ci_ 6 haloalkyl, or C 6 _io aryl; wherein said Ci_ 6 alkyl and C 6 -io aryl is optionally substituted with 1, 2, 3, 4, or 5 independently selected R g groups;
  • each R 6 is, independently, halo, cyano, Ci_ 6 alkyl, Ci_ 6 haloalkyl, OR al , NR cl R dl , or OC(0)R bl ;
  • each R al , R bl , R cl , and R dl is, independently, H or Ci_ 6 alkyl; wherein said Ci_ 6 alkyl is optionally substituted with a substituent independently selected from Ci_ 4 alkoxy and hydroxyl;
  • R cl and R dl together with the N atom to which they are attached form a 4-, 5-, 6-, or 7-membered heterocycloalkyl group, optionally substituted with 1, 2, 3 or 4 substituents independently selected from halo.
  • the compound is selected from:
  • the compound is selected from:
  • the salt is a l- ⁇ l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile adipic acid salt.
  • the salt is a 1 : 1 l- ⁇ l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile : adipic acid salt.
  • the salt is that described in Example 358.
  • the salt is characterized by a melting point of about 178 °C.
  • the salt has a differential scanning calorimetry thermogram which is characterized by an endothermic peak with an onset temperature of about 176 °C. In some embodiments, the salt has a differential scanning calorimetry thermogram substantially as shown in Figure 1.
  • the salt has a thermal gravimetric analysis thermogram substantially as shown in Figure 2.
  • the salt has an X-ray powder diffraction pattern comprising a characteristic peak expressed in degrees 2 ⁇ at about 10.4.
  • the salt has an X-ray powder diffraction pattern comprising a characteristic peak expressed in degrees 2 ⁇ at about 6.9.
  • the salt has an X-ray powder diffraction pattern comprising a characteristic peak expressed in degrees 2 ⁇ at about 21.0.
  • the salt has an X-ray powder diffraction pattern comprising a characteristic peak expressed in degrees 2 ⁇ at about 23.3.
  • the salt has an X-ray powder diffraction pattern comprising a
  • the salt has an X-ray powder diffraction pattern substantially as shown in Figure 3.
  • An XRPD pattern of reflections is typically considered a fingerprint of a particular crystalline form. It is well known that the relative intensities of the XRPD peaks can widely vary depending on, inter alia, the sample preparation technique, crystal size distribution, various filters used, the sample mounting procedure, and the particular instrument employed. In some instances, new peaks may be observed or existing peaks may disappear, depending on the type of the machine or the settings (for example, whether a Ni filter is used or not). As used herein, the term "peak” refers to a reflection having a relative height/intensity of at least about 4% of the maximum peak
  • substituents of compounds of the invention are disclosed in groups or in ranges. It is specifically intended that the invention include each and every individual subcombination of the members of such groups and ranges.
  • the term "Ci_ 6 alkyl” is specifically intended to individually disclose methyl, ethyl, C 3 alkyl, C 4 alkyl, C 5 alkyl, and C 6 alkyl.
  • linking substituents are described. It is specifically intended that each linking substituent include both the forward and backward forms of the linking substituent.
  • -NR(CR'R") n - includes both - NR(CR'R") n - and -(CR'R") n NR-.
  • the Markush variables listed for that group are understood to be linking groups. For example, if the structure requires a linking group and the Markush group definition for that variable lists "alkyl” or "aryl” then it is to be understood that the "alkyl” or “aryl” represents a linking alkylene group or arylene group, respectively.
  • rings are described (e.g., "a piperidine ring”). Unless otherwise specified, these rings can be attached to the rest of the molecule at any ring member as permitted by valency.
  • a pyridine ring may refer to a pyridin-2-yl, pyridin-3-yl, or pyridin-4-yl ring.
  • n-membered where n is an integer typically describes the number of ring-forming atoms in a moiety where the number of ring-forming atoms is n.
  • piperidinyl is an example of a 6-membered heterocycloalkyl ring
  • pyrazolyl is an example of a 5-membered heteroaryl ring
  • pyridyl is an example of a 6-membered heteroaryl ring
  • 1 ,2,3,4-tetrahydro-naphthalene is an example of a 10-membered cycloalkyl group.
  • each variable can be a different moiety independently selected from the group defining the variable.
  • the two R groups can represent different moieties independently selected from the group defined for R.
  • an optionally multiple substituent is designated in the form:
  • substituent R can occur p number of times on the ring, and R can be a different moiety at each occurrence. It is to be understood that each R group may replace any hydrogen atom attached to a ring atom, including one or both of the (CH 2 ) n hydrogen atoms. Further, in the above example, should the variable Q be defined to include hydrogens, such as when Q is said to be CH 2 , NH, etc., any floating substituent such as R in the above example, can replace a hydrogen of the Q variable as well as a hydrogen in any other non-variable component of the ring.
  • the phrase "optionally substituted” means unsubstituted or substituted.
  • substituted means that a hydrogen atom is removed and replaced by a substituent. It is to be understood that substitution at a given atom is limited by valency.
  • C n _ m alkyl refers to a saturated hydrocarbon group that may be straight-chain or branched, having n to m carbon atoms.
  • the alkyl group contains 1 to 6, 1 to 4 or 1 to 3 carbon atoms.
  • alkyl moieties include, but are not limited to, chemical groups such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, 2 -methyl- 1 -butyl, 3-pentyl, n-hexyl, 1,2,2-trimethylpropyl, n-heptyl, n-octyl, and the like.
  • chemical groups such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, 2 -methyl- 1 -butyl, 3-pentyl, n-hexyl, 1,2,2-trimethylpropyl, n-heptyl, n-octyl, and the like.
  • C n _ m alkenyl refers to an alkyl group having one or more carbon-carbon double bonds and n to m carbon atoms.
  • the alkenyl moiety contains 2 to 6, or 2 to 4 carbon atoms.
  • Example alkenyl groups include, but are not limited to, ethenyl, n- propenyl, isopropenyl, n-butenyl, sec-butenyl, and the like.
  • C n _ m alkynyl refers to an alkyl group having one or more carbon-carbon triple bonds and n to m carbon atoms.
  • Example alkynyl groups include, but are not limited to, ethynyl, propyn-1- yl, propyn-2-yl, and the like.
  • the alkynyl moiety contains 2 to 6 or 2 to 4 carbon atoms.
  • halo or halogen, employed alone or in combination with other terms, includes fluoro, chloro, bromo, and iodo.
  • hydroxyl or “hydroxy” refer to a group of formula -OH.
  • C n _ m haloalkyl refers to an C n _ m alkyl group having up to ⁇ 2(n to m)+l ⁇ halogen atoms which may either be the same or different.
  • the halogen atoms are fluoro atoms.
  • the alkyl group has 1 to 6 or 1 to 4 carbon atoms.
  • Example haloalkyl groups include CF 3 , C2F5, CHF 2 , CC1 3 , CHC1 2 , C2CI5, and the like.
  • the haloalkyl group is a fluoroalkyl group.
  • C n _ m fluoroalkyl employed alone or in combination with other terms, refers to a C n _ m haloalkyl wherein the halogen atoms are selected from fluorine.
  • C n _ m fluroalkyl is fluoromethyl, difluoromethyl, or trifluoromethyl.
  • C n _ m alkoxy refers to an group of formula -O-alkyl, wherein the alkyl group has n to m carbon atoms.
  • Example alkoxy groups include methoxy, ethoxy, propoxy (e.g., n- propoxy and isopropoxy), t-butoxy, and the like.
  • the alkyl group has 1 to 6 or 1 to 4 carbon atoms.
  • C n _ m haloalkoxy refers to a group of formula -O-(haloalkyl), wherein the haloalkyl group has n to m carbon atoms.
  • the alkyl group has 1 to 6 or 1 to 4 carbon atoms.
  • An example haloalkoxy group is -OCF 3 .
  • the haloalkoxy group is a fluoroalkoxy group.
  • C n _ m fluoroalkoxy employed alone or in combination with other terms, refers to a C n _ m alkoxy group, wherein the halogen atoms are selected from fluorine.
  • amino employed alone or in combination with other terms, refers to -NH 2 .
  • C n _ m alkylamino refers to a group of formula -NH(alkyl), wherein the alkyl group has n to m carbon atoms. In some embodiments, alkyl group has 1 to 6 or 1 to 4 carbon atoms.
  • Example C n _ m alkylamino groups include methylamino, ethylamino, propylamino (e.g., n- propylamino and isopropylamino), and the like.
  • di-C n _ m -alkylamino refers to a group of formula -N(alkyl) 2 , wherein each alkyl group has independently n to m carbon atoms.
  • Example di-C n _ m -alkylamino groups include dimethylamino, diethylamino, dipropylamino (e.g., di(n-propyl)amino and
  • each alkyl group independently has 1 to 6 or 1 to 4 carbon atoms.
  • the alkyl group has n to m carbon atoms. In some embodiments, the alkyl group has 1 to 6 or 1 to 4 carbon atoms.
  • C n _ m alkylcarbonyl refers to a group of formula -C(0)-alkyl, wherein the alkyl group has n to m carbon atoms. In some embodiments, the alkyl group has 1 to 6 or 1 to 4 carbon atoms.
  • C n _ m alkylcarbonylamino employed alone or in combination with other terms, refers to a group of formula -NHC(0)-alkyl, wherein the alkyl group has n to m carbon atoms. In some embodiments, the alkyl group has 1 to 6 or 1 to 4 carbon atoms.
  • carbamyl employed alone or in combination with other terms, refers to a group of formula -C(0)-NH 2 .
  • C n _ m alkylcarbamyl refers to a group of formula -C(0)-NH(alkyl), wherein the alkyl group has n to m carbon atoms. In some embodiments, the alkyl group has 1 to 6 or 1 to 4 carbon atoms.
  • di-C n _ m alkylcarbamyl refers to a group of formula -C(0)-N(alkyl) 2 , wherein the each alkyl group independently has n to m carbon atoms. In some embodiments, the alkyl group independently has 1 to 6 or 1 to 4 carbon atoms.
  • thio refers to a group of formula -SH.
  • C n _ m alkylthio employed alone or in combination with other terms, refers to a group of formula -S-alkyl, wherein the alkyl group has n to m carbon atoms. In some embodiments, the alkyl group has 1 to 6 or 1 to 4 carbon atoms.
  • C n _ m alkylsulfmyl employed alone or in combination with other terms, refers to a group of formula -S(0)-alkyl, wherein the alkyl group has n to m carbon atoms. In some embodiments, the alkyl group has 1 to 6 or 1 to 4 carbon atoms.
  • C n _ m alkylsulfonyl refers to a group of formula -S(0) 2 -alkyl, wherein the alkyl group has n to m carbon atoms. In some embodiments, the alkyl group has 1 to 6 or 1 to 4 carbon atoms.
  • halosulfanyl refers to a sulfur group having one or more halogen substituents.
  • Example halosulfanyl groups include pentahalosulfanyl groups such as SF 5 .
  • 2- or 3-carbon bridge means that two different R groups on different ring member atoms form a bridge (-CH 2 -CH 2 - or -CH 2 -CH 2 -CH 2 -) between the two ring member atoms, wherein the two or three carbons does not include the ring member atoms.
  • a non- limiting examples see Example 138, where two R 1 groups form a 2-carbon bridge.
  • bridge of formula -CH 2 -O-CH 2 - means that two different R groups on different ring member atoms form a bridge between the two ring member atoms of formula -CH 2 -O-CH 2 -, where the ring member atoms are not part of formula -CH 2 -0-CH 2 -.
  • hydroxyl-Ci_4 alkyl refers to a group of formula -C 1-4 alkylene-OH.
  • Ci_ 4 alkoxy-Ci_ 4 alkyl refers to a group of formula -Ci_ 4 alkylene-0-(Ci_4 alkyl).
  • C n _ m cycloalkyl refers to a non-aromatic cyclic hydrocarbon including cyclized alkyl, alkenyl, and alkynyl groups, and which has n to m ring member carbon atoms.
  • Cycloalkyl groups can include mono- or polycyclic (e.g., having 2, 3, or 4 fused, bridged, or spiro rings) ring systems. Also included in the definition of cycloalkyl are moieties that have one or more aromatic rings (e.g., aryl or heteroaryl rings) fused (i.e., having a bond in common with) to the cycloalkyl ring, for example, benzo derivatives of cyclopentane, cyclopentene, cyclohexane, and the like. Ring-forming carbon atoms of a cycloalkyl group can be optionally substituted by oxo. Cycloalkyl groups also include cycloalkylidenes.
  • cycloalkyl also includes bridgehead cycloalkyl groups and spirocycloalkyl groups.
  • bridgehead cycloalkyl groups refers to non- aromatic cyclic hydrocarbon moieties containing at least one bridgehead carbon, such as admantan-l-yl.
  • spirocycloalkyl groups refers to non-aromatic hydrocarbon moieties containing at least two rings fused at a single carbon atom, such as spiro[2.5]octane and the like.
  • the cycloalkyl group has 3 to 14 ring members, 3 to 10 ring members, or 3 to 7 ring members.
  • the cycloalkyl group is monocyclic, bicyclic or tricyclic. In some embodiments, the cycloalkyl group is monocyclic. In some embodiments, the cycloalkyl group is a C3-7 monocyclic cycloalkyl group. One or more ring-forming carbon atoms of a cycloalkyl group can be oxidized to form carbonyl linkages.
  • Example cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclopentenyl,
  • C n _ m cycloalkyl-C 0 _ p alkyl refers to a group of formula -alkylene-cycloalkyl, wherein the cycloalkyl portion has n to m carbon atoms and the alkylene portion has o to p carbon atoms.
  • the alkylene portion has 1 to 4, 1 to 3, 1 to 2, or 1 carbon atom(s).
  • the alkylene portion is methylene.
  • the cycloalkyl portion has 3 to 14 ring members, 3 to 10 ring members, or 3 to 7 ring members.
  • the cycloalkyl group is monocyclic or bicyclic.
  • the cycloalkyl portion is monocyclic.
  • the cycloalkyl portion is a C3_ 7 monocyclic cycloalkyl group.
  • C n _ m heterocycloalkyl As used herein, the term "C n _ m heterocycloalkyl”, “C n _ m heterocycloalkyl ring”, or
  • C n _ m heterocycloalkyl group refers to non-aromatic ring or ring system, which may optionally contain one or more alkenylene or alkynylene groups as part of the ring structure, which has at least one heteroatom ring member independently selected from nitrogen, sulfur oxygen and phosphorus, and which has n to m ring member carbon atoms.
  • Heterocycloalkyl groups can include mono- or polycyclic (e.g., having 2, 3 or 4 fused, bridged, or spiro rings) ring systems.
  • the heterocycloalkyl group is a monocyclic or bicyclic group having 1, 2, 3, or 4 hetereoatoms independently selected from nitrogen, sulfur and oxygen.
  • moieties that have one or more aromatic rings (e.g., aryl or heteroaryl rings) fused (i.e., having a bond in common with) to the non-aromatic ring, for example, 1,2,3,4-tetrahydro-quinoline and the like.
  • Heterocycloalkyl groups can also include bridgehead heterocycloalkyl groups and spiroheterocycloalkyl groups.
  • bridgehead heterocycloalkyl group refers to a heterocycloalkyl moiety containing at least one bridgehead atom, such as
  • spiroheterocycloalkyl group refers to a heterocycloalkyl moiety containing at least two rings fused at a single atom, such as [1,4- dioxa-8-aza-spiro[4.5]decan-N-yl] and the like.
  • the amino acids such as [1,4- dioxa-8-aza-spiro[4.5]decan-N-yl] and the like.
  • heterocycloalkyl group has 3 to 20 ring-forming atoms, 3 to 14 ring-forming atoms, 3 to 10 ring-forming atoms, or about 3 to 8 ring forming atoms. In some embodiments, the heterocycloalkyl group has 2 to 20 carbon atoms, 2 to 15 carbon atoms, 2 to 10 carbon atoms, or about 2 to 8 carbon atoms. In some embodiments, the heterocycloalkyl group has 1 to 5 heteroatoms, 1 to 4 heteroatoms, 1 to 3 heteroatoms, or 1 to 2 heteroatoms.
  • the carbon atoms or hetereoatoms in the ring(s) of the heterocycloalkyl group can be oxidized to form a carbonyl, an N-oxide, or a sulfonyl group (or other oxidized linkage) or a nitrogen atom can be quaternized.
  • the heterocycloalkyl portion is a C 2 -7 monocyclic heterocycloalkyl group.
  • heterocycloalkyl groups include 1, 2,3, 4-tetrahydro-quino line, azetidine, azepane, pyrrolidine, piperidine, piperazine, morpholine, thiomorpholine, and pyran.
  • C n _ m heterocycloalkyl-C 0 _ p alkyl refers to a group of formula -alkylene-heterocycloalkyl, wherein the heterocycloalkyl portion has n to m carbon atoms and the alkylene portion has o to p carbon atoms.
  • the alkylene portion has 1 to 4, 1 to 3, 1 to 2, or 1 carbon atom(s).
  • the alkylene portion is methylene.
  • the heterocycloalkyl portion has 3 to 14 ring members, 3 to 10 ring members, or 3 to 7 ring members.
  • the heterocycloalkyl group is monocyclic or bicyclic. In some embodiments, the heterocycloalkyl portion is monocyclic. In some embodiments, the heterocycloalkyl portion is a C 2 -7 monocyclic heterocycloalkyl group.
  • C n _ m aryl refers to a monocyclic or polycyclic (e.g., having 2, 3 or 4 fused rings) aromatic hydrocarbon moiety having n to m ring member carbon atoms, such as, but not limited to, phenyl, 1-naphthyl, 2-naphthyl, anthracenyl, phenanthrenyl, and the like.
  • aryl groups have from 6 to 20 carbon atoms, from 6 to 14 carbon atoms, from 6 to 10 carbon atoms, or 6 carbon atoms.
  • the aryl group is a monocyclic or bicyclic group.
  • C n _ m aryl-C 0 _ p -alkyl refers to a group of formula -alkylene-aryl, wherein the aryl portion has n to m ring member carbon atoms and the alkylene portion has o to p carbon atoms.
  • the alkylene portion has 1 to 4, 1 to 3, 1 to 2, or 1 carbon atom(s).
  • the alkylene portion is methylene.
  • the aryl portion is phenyl.
  • the aryl group is a monocyclic or bicyclic group.
  • the arylalkyl group is benzyl.
  • the term "C n _ m heteroaryl”, “C n _ m heteroaryl ring”, or “C n _ m heteroaryl group”, employed alone or in combination with other terms refers to a monocyclic or polycyclic (e.g., having 2, 3 or 4 fused rings) aromatic hydrocarbon moiety, having one or more heteroatom ring members independently selected from nitrogen, sulfur and oxygen and having n to m ring member carbon atoms.
  • the heteroaryl group is a monocyclic or bicyclic group having 1, 2, 3, or 4 hetereoatoms independently selected from nitrogen, sulfur and oxygen.
  • Example heteroaryl groups include, but are not limited to, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazinyl, furyl, thienyl, imidazolyl, thiazolyl, indolyl, pyrryl, oxazolyl, benzthiazolyl, isoxazolyl, pyrazolyl, triazolyl, tetrazolyl, indazolyl, 1,2,4-thiadiazolyl, isothiazolyl, benzothienyl, purinyl, carbazolyl, benzimidazolyl, pyrrolyl, azolyl, oxazolyl, quinolinyl, isoquinolinyl, indolyl, benzothienyl, benzofuranyl, benzisoxazolyl, imidazo[l,2-b]thiazolyl or the like.
  • the carbon atoms or hetereoatoms in the ring(s) of the heteroaryl group can be oxidized to form a carbonyl, an N-oxide, or a sulfonyl group (or other oxidized linkage) or a nitrogen atom can be quaternized, provided the aromatic nature of the ring is preserved.
  • the heteroaryl group has from 1 to 20 carbon atoms, from 3 to 20 carbon atoms, from 3 to 15 carbon atoms, from 3 to 10 carbon atoms, from 3 to 8 carbon atoms, from 3 to 5 carbon atoms, from 1 to 5 carbon atoms, or from 5 to 10 carbon atoms.
  • the heteroaryl group contains 3 to 14, 4 to 12, 4 to 8, 9 to 10, or 5 to 6 ring-forming atoms.
  • the heteroaryl group has 1 to 4, 1 to 3, or 1 to 2 heteroatoms.
  • C n _ m heteroaryl-C 0 _ p -alkyl refers to a group of formula -alkylene-heteroaryl, wherein the heteroaryl portion has n to m ring member carbon atoms and the alkylene portion has o to p carbon atoms.
  • the alkylene portion has 1 to 4, 1 to 3, 1 to 2, or 1 carbon atom(s).
  • the alkylene portion is methylene.
  • the heteroaryl portion is a monocyclic or bicyclic group having 1, 2, 3, or 4 hetereoatoms independently selected from nitrogen, sulfur and oxygen. In some embodiments, the heteroaryl portion has 5 to 10 carbon atoms.
  • C n _ m aryloxy refers to a moiety of formula -O-aryl, wherein the aryl ring has n to m carbon atoms.
  • the appearance of the term "bicyclic" before the name of a moiety indicates that the moiety has two fused rings.
  • the compounds described herein can be asymmetric (e.g., having one or more stereocenters). All stereoisomers, such as enantiomers and diastereomers, are intended unless otherwise indicated.
  • asymmetrically substituted carbon atoms can be isolated in optically active or racemic forms.
  • Methods on how to prepare optically active forms from optically inactive starting materials are known in the art, such as by resolution of racemic mixtures or by stereoselective synthesis.
  • Cis and trans geometric isomers of the compounds of the present invention are described and may be isolated as a mixture of isomers or as separated isomeric forms.
  • Resolution of racemic mixtures of compounds can be carried out by any of numerous methods known in the art.
  • An example method includes fractional
  • Suitable resolving agents for fractional recrystallization methods are, for example, optically active acids, such as the D and L forms of tartaric acid, diacetyltartaric acid, dibenzoyltartaric acid, mandelic acid, malic acid, lactic acid or the various optically active camphorsulfonic acids such as ⁇ -camphorsulfonic acid.
  • optically active acids such as the D and L forms of tartaric acid, diacetyltartaric acid, dibenzoyltartaric acid, mandelic acid, malic acid, lactic acid or the various optically active camphorsulfonic acids such as ⁇ -camphorsulfonic acid.
  • resolving agents suitable for fractional crystallization methods include stereoisomerically pure forms of a- methylbenzylamine (e.g., S and R forms, or diastereomerically pure forms), 2- phenylglycinol, norephedrine, ephedrine, N-methylephedrine, cyclohexylethylamine, 1 ,2- diaminocyclohexane, and the like.
  • Resolution of racemic mixtures can also be carried out by elution on a column packed with an optically active resolving agent (e.g., dinitrobenzoylphenylglycine).
  • an optically active resolving agent e.g., dinitrobenzoylphenylglycine
  • Suitable elution solvent composition can be determined by one skilled in the art.
  • Tautomeric forms result from the swapping of a single bond with an adjacent double bond together with the concomitant migration of a proton.
  • Tautomeric forms include prototropic tautomers which are isomeric protonation states having the same empirical formula and total charge.
  • Example prototropic tautomers include ketone - enol pairs, amide - imidic acid pairs, lactam - lactim pairs, enamine - imine pairs, and annular forms where a proton can occupy two or more positions of a heterocyclic system, for example, 1H- and 3H- imidazole, 1H-, 2H- and 4H- 1,2,4-triazole, 1H- and 2H- isoindole, and 1H- and 2H- pyrazole.
  • Tautomeric forms can be in equilibrium or sterically locked into one form by appropriate substitution.
  • Compounds of the invention can also include all isotopes of atoms occurring in the intermediates or final compounds.
  • Isotopes include those atoms having the same atomic number but different mass numbers.
  • isotopes of hydrogen include tritium and deuterium.
  • 1, 2, or 3 CH 2 groups in the azetidine ring of Formula I are replaced by a CHD or CD 2 group.
  • 1, 2, or 3 CH 2 or CH groups in the piperidine ring of Formula I are replaced by a CHD, CD 2 or CD group, respectively.
  • 1, 2, 3, 4, or 5 CH 2 or CH groups in the piperidine ring of Formula I are replaced by a CHD, CD 2 or CD group, respectively.
  • compound as used herein is meant to include all stereoisomers, geometric iosomers, tautomers, and isotopes of the structures depicted.
  • All compounds, and pharmaceutically acceptable salts thereof, can be found together with other substances such as water and solvents (e.g., hydrates and solvates) or can be isolated.
  • the compounds of the invention, or salts thereof are substantially isolated.
  • substantially isolated is meant that the compound is at least partially or substantially separated from the environment in which it was formed or detected.
  • Partial separation can include, for example, a composition enriched in the compounds of the invention.
  • Substantial separation can include compositions containing at least about 50%, at least about 60%>, at least about 70%>, at least about 80%>, at least about 90%), at least about 95%, at least about 97%, or at least about 99% by weight of the compounds of the invention, or salt thereof. Methods for isolating compounds and their salts are routine in the art.
  • phrases "pharmaceutically acceptable” is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • ambient temperature and “room temperature,” as used herein, are understood in the art, and refer generally to a temperature, e.g. a reaction temperature, that is about the temperature of the room in which the reaction is carried out, for example, a temperature from about 20 °C to about 30 °C.
  • the present invention also includes pharmaceutically acceptable salts of the compounds described herein.
  • pharmaceutically acceptable salts refers to derivatives of the disclosed compounds wherein the parent compound is modified by converting an existing acid or base moiety to its salt form.
  • examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like.
  • the pharmaceutically acceptable salts of the present invention include the non-toxic salts of the parent compound formed, for example, from non-toxic inorganic or organic acids.
  • the pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods.
  • such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, non-aqueous media like ether, ethyl acetate, alcohols (e.g., methanol, ethanol, iso-propanol, or butanol) or acetonitrile (ACN) are preferred.
  • non-aqueous media like ether, ethyl acetate, alcohols (e.g., methanol, ethanol, iso-propanol, or butanol) or acetonitrile (ACN) are preferred.
  • ACN acetonitrile
  • Suitable solvents can be substantially non-reactive with the starting materials (reactants), the intermediates, or products at the temperatures at which the reactions are carried out, e.g., temperatures which can range from the solvent's freezing temperature to the solvent's boiling temperature.
  • a given reaction can be carried out in one solvent or a mixture of more than one solvent.
  • suitable solvents for a particular reaction step can be selected by the skilled artisan.
  • Preparation of compounds of the invention can involve the protection and deprotection of various chemical groups.
  • the need for protection and deprotection, and the selection of appropriate protecting groups, can be readily determined by one skilled in the art.
  • the chemistry of protecting groups can be found, for example, in Wuts and Greene, Protective Groups in Organic Synthesis, 4th ed., John Wiley & Sons: New Jersey, (2007), which is incorporated herein by reference in its entirety.
  • Reactions can be monitored according to any suitable method known in the art.
  • product formation can be monitored by spectroscopic means, such as nuclear magnetic resonance spectroscopy (e.g., 1H or 13 C), infrared spectroscopy, spectrophotometry (e.g., UV-visible), mass spectrometry, or by chromatographic methods such as high performance liquid chromatography (HPLC) or thin layer chromatography (TLC).
  • spectroscopic means such as nuclear magnetic resonance spectroscopy (e.g., 1H or 13 C), infrared spectroscopy, spectrophotometry (e.g., UV-visible), mass spectrometry, or by chromatographic methods such as high performance liquid chromatography (HPLC) or thin layer chromatography (TLC).
  • HPLC high performance liquid chromatography
  • TLC thin layer chromatography
  • Suzuki coupling of 1-2 with a boronic acid of pyrazole such as l-(l-ethoxyethyl)-4-(4,4,5,5-tetramethyl- l,3,2-dioxaborolan-2-yl)-lH-pyrazole (1-3)
  • a palladium reagent such as tetrakis(triphenylphosphine)palladium(0)
  • Intermediates of fomula 2-3 can be synthesized according the sequence depicted in Scheme 2.
  • the SEM-protected intermediate 1-2 is subjected to a Suzuki coupling with a boronic acid of a protected pyrrole, such as l-(triisopropylsilyl)pyrrole-3-boronic acid (2-1), using a palladium reagent, such as tetrakis(triphenylphosphine)palladium(0), in the presence of a base.
  • a palladium reagent such as tetrakis(triphenylphosphine)palladium(0)
  • Intermediates of formula 3-7 can be derivatized at the piperidine nitrogen to produce a series of compounds of Formula I as depicted in Scheme 4.
  • Reaction of the compound of formula 3-7 with a sulfonyl chloride followed by treating first with TFA and and then with ethylenediamine to remove the SEM group yields sulfonamide derivatives of formula 4-1.
  • Coupling of the compound of formula 3-7 with a carboxylic acid using a coupling agent such as BOP or with an acyl chloride, followed by removal of the SEM group provides amide compounds of formula 4-2.
  • Reductive amination of the compound of formula 3-7 with an aldehyde using a reducing agent, such as sodium triacetoxyborohydride, followed by removal of the SEM group gives rise to the N-alkyl derivatives of formula 4-3.
  • Reaction of the compound of formula 3-7 with an isocyanate, followed by removing the SEM group affords the urea compounds of formula 4-4.
  • JAK inhibitors are JAK inhibitors, and the majority of the compounds of the invention, are JAKl selective inhibitors.
  • a JAKl selective inhibitor is a compound that inhibits JAKl activity preferentially over other Janus kinases.
  • the compounds of the invention preferentially inhibit JAKl over one or more of JAK2, JAK3, and TYK2.
  • the compounds inhibit JAKl preferentially over JAK2 (e.g., have a JAK1/JAK2 IC 50 ratio >1).
  • the compounds are about 10-fold more selective for JAKl over JAK2.
  • the compounds are about 3 -fold, about 5 -fold, about 10-fold, about 15- fold, or about 20-fold more selective for JAKl over JAK2 as calculated by measuring IC 50 at 1 mM ATP (e.g., see Example A).
  • JAKl plays a central role in a number of cytokine and growth factor signaling pathways that, when dysregulated, can result in or contribute to disease states. For example, IL-6 levels are elevated in rheumatoid arthritis, a disease in which it has been suggested to have detrimental effects (Fonesca, J.E. et al, Autoimmunity Reviews, 8:538-42, 2009). Because IL-6 signals, at least in part, through JAKl, antagonizing IL-6 directly or indirectly through JAKl inhibition is expected to provide clinical benefit (Guschin, D., N., et al Embo J 14: 1421, 1995; Smolen, J. S., et al. Lancet 371 :987, 2008).
  • JAKl is mutated resulting in constitutive undesirable tumor cell growth and survival (Mullighan CG, Proc Natl Acad Sci U S A.106:9414-8, 2009; Flex E., et al.J Exp Med. 205:751-8, 2008).
  • JAKl inhibition In other autoimmune diseases and cancers elevated systemic levels of inflammatory cytokines that activate JAKl may also contribute to the disease and/or associated symptoms. Therefore, patients with such diseases may benefit from JAKl inhibition.
  • Selective inhibitors of JAKl may be efficacious while avoiding unnecessary and potentially undesirable effects of inhibiting other JAK kinases.
  • JAKl erythropoietin
  • Tpo thrombopoietin
  • Epo is a key growth factor for red blood cells production; hence a paucity of Epo-dependent signaling can result in reduced numbers of red blood cells and anemia (Kaushansky K, NEJM 354:2034-45, 2006).
  • Tpo another example of a JAK2-dependent growth factor, plays a central role in controlling the proliferation and maturation of megakaryocytes - the cells from which platelets are produced (Kaushansky K, NEJM 354:2034-45, 2006). As such, reduced Tpo signaling would decrease megakaryocyte numbers (megakaryocytopenia) and lower circulating platelet counts (thrombocytopenia). This can result in undesirable and/or uncontrollable bleeding.
  • JAK3 and Tyk2 Reduced inhibition of other JAKs, such as JAK3 and Tyk2, may also be desirable as humans lacking functional version of these kinases have been shown to suffer from numerous maladies such as severe-combined immunodeficiency or
  • JAK1 inhibitor with reduced affinity for other JAKs would have significant advantages over a less-selective inhibitor with respect to reduced side effects involving immune suppression, anemia and thrombocytopenia.
  • a JAK-associated disease can include any disease, disorder or condition that is directly or indirectly linked to expression or activity of the JAK, including overexpression and/or abnormal activity levels.
  • a JAK-associated disease can also include any disease, disorder or condition that can be prevented, ameliorated, or cured by modulating JAK activity.
  • JAK-associated diseases include diseases involving the immune system including, for example, organ transplant rejection (e.g., allograft rejection and graft versus host disease).
  • organ transplant rejection e.g., allograft rejection and graft versus host disease.
  • JAK-associated diseases include autoimmune diseases such as multiple sclerosis, rheumatoid arthritis, juvenile arthritis, psoriatic arthritis, type I diabetes, lupus, psoriasis, inflammatory bowel disease, ulcerative colitis, Crohn's disease, myasthenia gravis, immunoglobulin nephropathies, myocarditis, autoimmune thyroid disorders, chronic obstructive pulmonary disease (COPD), and the like.
  • the autoimmune disease is an autoimmune bullous skin disorder such as pemphigus vulgaris (PV) or bullous pemphigoid (BP).
  • JAK-associated diseases include allergic conditions such as asthma, food allergies, eszematous dermatitis, contact dermatitis, atopic dermatitis (atropic eczema), and rhinitis.
  • JAK-associated diseases include viral diseases such as Epstein Barr Virus (EBV), Hepatitis B, Hepatitis C, HIV, HTLV 1 , Varicella-Zoster Virus (VZV) and Human Papilloma Virus (HPV).
  • EBV Epstein Barr Virus
  • Hepatitis B Hepatitis C
  • HIV HTLV 1
  • VZV Varicella-Zoster Virus
  • HPV Human Papilloma Virus
  • JAK-associated disease examples include diseases associated with cartilage turnover, for example, gouty arthritis, septic or infectious arthritis, reactive arthritis, reflex sympathetic dystrophy, algodystrophy, Tietze syndrome, costal athropathy, osteoarthritis deformans endemica, Mseleni disease, Handigodu disease, degeneration resulting from fibromyalgia, systemic lupus erythematosus, scleroderma, or ankylosing spondylitis.
  • diseases associated with cartilage turnover for example, gouty arthritis, septic or infectious arthritis, reactive arthritis, reflex sympathetic dystrophy, algodystrophy, Tietze syndrome, costal athropathy, osteoarthritis deformans endemica, Mseleni disease, Handigodu disease, degeneration resulting from fibromyalgia, systemic lupus erythematosus, scleroderma, or ankylosing spondylitis.
  • JAK-associated disease examples include congenital cartilage malformations, including hereditary chrondro lysis, chrondrodysplasias, and
  • pseudochrondrodysplasias e.g., microtia, enotia, and metaphyseal chrondrodysplasia.
  • JAK-associated diseases or conditions include skin disorders such as psoriasis (for example, psoriasis vulgaris), atopic dermatitis, skin rash, skin irritation, skin sensitization (e.g., contact dermatitis or allergic contact dermatitis).
  • skin disorders such as psoriasis (for example, psoriasis vulgaris), atopic dermatitis, skin rash, skin irritation, skin sensitization (e.g., contact dermatitis or allergic contact dermatitis).
  • certain substances including some pharmaceuticals when topically applied can cause skin sensitization.
  • co-administration or sequential administration of at least one JAK inhibitor of the invention together with the agent causing unwanted sensitization can be helpful in treating such unwanted sensitization or dermatitis.
  • the skin disorder is treated by topical administration of at least one JAK inhibitor of the invention.
  • the JAK-associated disease is cancer including those characterized by solid tumors (e.g., prostate cancer, renal cancer, hepatic cancer, pancreatic cancer, gastric cancer, breast cancer, lung cancer, cancers of the head and neck, thyroid cancer, glioblastoma, Kaposi's sarcoma, Castleman's disease, uterine leiomyosarcoma, melanoma etc.), hematological cancers (e.g., lymphoma, leukemia such as acute lymphoblastic leukemia (ALL), acute myelogenous leukemia (AML) or multiple myeloma), and skin cancer such as cutaneous T-cell lymphoma (CTCL) and cutaneous B-cell lymphoma.
  • CTCLs include Sezary syndrome and mycosis fungoides.
  • the JAK inhibitors described herein, or in combination with other JAK inhibitors, such as those reported in U.S. Ser. No. 1 1/637,545, which is incorporated herein by reference in its entirety, can be used to treat inflammation- associated cancers.
  • the cancer is associated with inflammatory bowel disease.
  • the inflammatory bowel disease is ulcerative colitis.
  • the inflammatory bowel disease is Crohn's disease.
  • the inflammation-associated cancer is colitis-associated cancer.
  • the inflammation-associated cancer is colon cancer or colorectal cancer.
  • the cancer is gastric cancer, gastrointestinal carcinoid tumor, gastrointestinal stromal tumor (GIST), adenocarcinoma, small intestine cancer, or rectal cancer.
  • JAK-associated diseases can further include those characterized by expression of: JAK2 mutants such as those having at least one mutation in the pseudo-kinase domain (e.g., JAK2V617F); JAK2 mutants having at least one mutation outside of the pseudo- kinase domain; JAK1 mutants; JAK3 mutants; erythropoietin receptor (EPOR) mutants; or deregulated expression of CRLF2.
  • JAK2 mutants such as those having at least one mutation in the pseudo-kinase domain (e.g., JAK2V617F); JAK2 mutants having at least one mutation outside of the pseudo- kinase domain; JAK1 mutants; JAK3 mutants; erythropoietin receptor (EPOR) mutants; or deregulated expression of CRLF2.
  • JAK-associated diseases can further include myeloproliferative disorders (MPDs) such as polycythemia vera (PV), essential thrombocythemia (ET), myelofibrosis with myeloid metaplasia (MMM), primary myelofibrosis (PMF), chronic myelogenous leukemia (CML), chronic myelomonocytic leukemia (CMML), hypereosinophilic syndrome (HES), systemic mast cell disease (SMCD), and the like.
  • MPDs myeloproliferative disorders
  • PV polycythemia vera
  • E essential thrombocythemia
  • MMM myelofibrosis with myeloid metaplasia
  • PMF primary myelofibrosis
  • CML chronic myelogenous leukemia
  • CMML chronic myelomonocytic leukemia
  • HES hypereosinophilic syndrome
  • SMCD systemic mast cell disease
  • the myeloproliferative disorder is myelofibrosis (e.g., primary
  • the myeloproliferative disorder is post- essential thrombocythemia myelofibrosis (Post-ET). In some embodiments, the myeloproliferative disorder is post polycythemia vera myelofibrosis (Post-PV MF).
  • the present invention further provides methods of treating psoriasis or other skin disorders by administration of a topical formulation containing a compound of the invention.
  • JAK inhibitors described herein can be used to treat pulmonary arterial hypertension.
  • the present invention further provides a method of treating dermatological side effects of other pharmaceuticals by administration of the compound of the invention.
  • numerous pharmaceutical agents result in unwanted allergic reactions which can manifest as acneiform rash or related dermatitis.
  • Example pharmaceutical agents that have such undesirable side effects include anti-cancer drugs such as gefitinib, cetuximab, erlotinib, and the like.
  • the compounds of the invention can be administered systemically or topically (e.g., localized to the vicinity of the dermatitis) in combination with (e.g., simultaneously or sequentially) the pharmaceutical agent having the undesirable dermatological side effect.
  • the compound of the invention can be administered topically together with one or more other pharmaceuticals, where the other pharmaceuticals when topically applied in the absence of a compound of the invention cause contact dermatitis, allergic contact sensitization, or similar skin disorder.
  • compositions of the invention include topical formulations containing the compound of the invention and a further pharmaceutical agent which can cause dermatitis, skin disorders, or related side effects.
  • JAK-associated diseases include inflammation and inflammatory diseases.
  • Example inflammatory diseases include sarcoidosis, inflammatory diseases of the eye (e.g., crizis, uveitis, scleritis, conjunctivitis, or related disease), inflammatory diseases of the respiratory tract (e.g. , the upper respiratory tract including the nose and sinuses such as rhinitis or sinusitis or the lower respiratory tract including bronchitis, chronic obstructive pulmonary disease, and the like), inflammatory myopathy such as
  • the inflammation disease of the eye is blepharitis.
  • the JAK inhibitors described herein can further be used to treat ischemia reperfusion injuries or a disease or condition related to an inflammatory ischemic event such as stroke or cardiac arrest.
  • the JAK inhibitors described herein can further be used to treat endotoxin-driven disease state (e.g., complications after bypass surgery or chronic endotoxin states contributing to chronic cardiac failure).
  • the JAK inhibitors described herein can further be used to treat anorexia, cachexia, or fatigue such as that resulting from or associated with cancer.
  • the JAK inhibitors described herein can further be used to treat restenosis, sclerodermitis, or fibrosis.
  • the JAK inhibitors described herein can further be used to treat conditions associated with hypoxia or astrogliosis such as, for example, diabetic retinopathy, cancer, or neurodegeneration. See, e.g., Dudley, A.C. et al. Biochem. J. 2005, 390(Pt 2):427-36 and Sriram, K. et al. J. Biol. Chem. 2004, 279(19): 19936-47. Epub 2004 Mar 2, both of which are incorporated herein by reference in their entirety.
  • the JAK inhibitors described herein can be used to treat Alzheimer's disease.
  • JAK inhibitors described herein can further be used to treat other
  • SIRS systemic inflammatory response syndrome
  • JAK inhibitors described herein can further be used to treat gout and increased prostate size due to, e.g., benign prostatic hypertrophy or benign prostatic hyperplasia.
  • JAK-associated diseases include bone resorption diseases such as osteoporosis, osteoarthritis. Bone resorption can also be associated with other conditions such as hormonal imbalance and/or hormonal therapy, autoimmune disease (e.g. osseous sarcoidosis), or cancer (e.g. myeloma).
  • the reduction of the bone resorption due to the JAK inhibitors can be about 10%, about 20%, about 30%>, about 40%>, about 50%>, about 60%, about 70%, about 80%, or about 90%.
  • JAK inhibitors described herein can further be used to treat a dry eye disorder.
  • dry eye disorder is intended to encompass the disease states summarized in a recent official report of the Dry Eye Workshop (DEWS), which defined dry eye as "a multifactorial disease of the tears and ocular surface that results in symptoms of discomfort, visual disturbance, and tear film instability with potential damage to the ocular surface. It is accompanied by increased osmolality of the tear film and inflammation of the ocular surface.” Lemp, "The Definition and
  • the dry eye disorder is selected from aqueous tear-deficient dry eye (ADDE) or evaporative dry eye disorder, or appropriate combinations thereof.
  • the dry eye disorder is Sjogren syndrome dry eye (SSDE).
  • the dry eye disorder is non-Sjogren syndrome dry eye (NSSDE).
  • the present invention provides a method of treating conjunctivitis, uveitis (including chronic uveitis), chorioditis, retinitis, cyclitis, sclieritis, episcleritis, or ulceris; treating inflammation or pain related to corneal transplant, LASIK (laser assisted in situ keratomileusis), photorefractive keratectomy, or LASEK (laser assisted sub-epithelial keratomileusis); inhibiting loss of visual acuity related to corneal transplant, LASIK, photorefractive keratectomy, or LASEK; or inhibiting transplant rejection in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of the compound of the invention, or a pharmaceutically acceptable salt thereof.
  • the compounds of the invention can be used to treat respiratory dysfunction or failure associated wth viral infection, such as influenza and SARS.
  • the present invention provides a compound of Formula I, pharmaceutically acceptable salt thereof, as described in any of the embodiments herein, for use in a method of treating any of the diseases or disorders described herein.
  • the present invention provides the use of a compound of Formula I as described in any of the embodiments herein, for the preparation of a medicament for use in a method of treating any of the diseases or disorders described herein.
  • the present invention provides a compound of Formula I as described herein, or a pharmaceutically acceptable salt thereof, for use in a method of modulating JAK1. In some embodiments, the present invention also provides use of a compound of Formula I as described herein, or a pharmaceutically acceptable salt thereof, for the preparation of a medicament for use in a method of modulating JAK1.
  • contacting refers to the bringing together of indicated moieties in an in vitro system or an in vivo system.
  • "contacting" a JAK with a compound of the invention includes the administration of a compound of the present invention to an individual or patient, such as a human, having a JAK, as well as, for example, introducing a compound of the invention into a sample containing a cellular or purified preparation containing the JAK.
  • the phrase "therapeutically effective amount” refers to the amount of active compound or pharmaceutical agent that elicits the biological or medicinal response that is being sought in a tissue, system, animal, individual or human by a researcher, veterinarian, medical doctor or other clinician. In some embodiments, the therapeutically effective amount is about 5 mg to about 1000 mg, or about 10 mg to about 500 mg.
  • the term "treating" or “treatment” refers to one or more of (1) preventing the disease; for example, preventing a disease, condition or disorder in an individual who may be predisposed to the disease, condition or disorder but does not yet experience or display the pathology or symptomatology of the disease; (2) inhibiting the disease; for example, inhibiting a disease, condition or disorder in an individual who is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e., arresting further development of the pathology and/or symptomatology); and (3) ameliorating the disease; for example, ameliorating a disease, condition or disorder in an individual who is experiencing or displaying the pathology or
  • symptomatology of the disease, condition or disorder i.e., reversing the pathology and/or symptomatology
  • decreasing the severity of disease i.e., decreasing the severity of disease.
  • One or more additional pharmaceutical agents such as, for example,
  • chemotherapeutics anti-inflammatory agents, steroids, immunosuppressants, as well as Bcr-Abl, Flt-3, RAF and FAK kinase inhibitors such as, for example, those described in WO 2006/056399, which is incorporated herein by reference in its entirety, or other agents can be used in combination with the compounds described herein for treatment of JAK-associated diseases, disorders or conditions.
  • additional agents such as, for example, those described in WO 2006/056399, which is incorporated herein by reference in its entirety, or other agents can be used in combination with the compounds described herein for treatment of JAK-associated diseases, disorders or conditions.
  • additional agents such as, for example, those described in WO 2006/056399, which is incorporated herein by reference in its entirety, or other agents can be used in combination with the compounds described herein for treatment of JAK-associated diseases, disorders or conditions.
  • pharmaceutical agents can be administered to a patient simultaneously or sequentially.
  • Example chemotherapeutics include proteosome inhibitors ⁇ e.g., bortezomib), thalidomide, revlimid, and DNA-damaging agents such as melphalan, doxorubicin, cyclophosphamide, vincristine, etoposide, carmustine, and the like.
  • Example steroids include coriticosteroids such as dexamethasone or prednisone.
  • Example Bcr-Abl inhibitors include the compounds, and pharmaceutically acceptable salts thereof, of the genera and species disclosed in U.S. Pat. No. 5,521,184, WO 04/005281, and U.S. Ser. No. 60/578,491, all of which are incorporated herein by reference in their entirety.
  • Example suitable Flt-3 inhibitors include compounds, and their pharmaceutically acceptable salts, as disclosed in WO 03/037347, WO 03/099771, and WO 04/046120, all of which are incorporated herein by reference in their entirety.
  • Example suitable RAF inhibitors include compounds, and their pharmaceutically acceptable salts, as disclosed in WO 00/09495 and WO 05/028444, both of which are incorporated herein by reference in their entirety.
  • Example suitable FAK inhibitors include compounds, and their pharmaceutically acceptable salts, as disclosed in WO 04/080980, WO 04/056786, WO 03/024967, WO 01/064655, WO 00/053595, and WO 01/014402, all of which are incorporated herein by reference in their entirety.
  • one or more of the compounds of the invention can be used in combination with one or more other kinase inhibitors including imatinib, particularly for treating patients resistant to imatinib or other kinase inhibitors.
  • one or more JAK inhibitors of the invention can be used in combination with a chemotherapeutic in the treatment of cancer, such as multiple myeloma, and may improve the treatment response as compared to the response to the chemotherapeutic agent alone, without exacerbation of its toxic effects.
  • additional pharmaceutical agents used in the treatment of multiple myeloma can include, without limitation, melphalan, melphalan plus prednisone [MP],
  • doxorubicin doxorubicin
  • dexamethasone dexamethasone
  • Velcade bortezomib
  • Further additional agents used in the treatment of multiple myeloma include Bcr-Abl, Flt-3, RAF and FAK kinase inhibitors. Additive or synergistic effects are desirable outcomes of combining a JAK inhibitor of the present invention with an additional agent.
  • resistance of multiple myeloma cells to agents such as dexamethasone may be reversible upon treatment with a JAK inhibitor of the present invention.
  • the agents can be combined with the present compounds in a single or continuous dosage form, or the agents can be administered simultaneously or sequentially as separate dosage forms.
  • a corticosteroid such as dexamethasone is administered to a patient in combination with at least one JAK inhibitor where the dexamethasone is administered intermittently as opposed to continuously.
  • combinations of one or more JAK inhibitors of the invention with other therapeutic agents can be administered to a patient prior to, during, and/or after a bone marrow transplant or stem cell transplant.
  • the additional therapeutic agent is fluocinolone acetonide (Retisert®), or rimexolone (AL-2178, Vexol, Alcon).
  • the additional therapeutic agent is cyclosporine
  • the additional therapeutic agent is a corticosteroid.
  • the corticosteroid is triamcinolone, dexamethasone, fluocinolone, cortisone, prednisolone, or flumetholone.
  • the additional therapeutic agent is selected from:
  • dehydroepiandrosterone anakinra, efalizumab, mycophenolate sodium, etanercept (Embrel®), hydroxychloroquine, NGX267 (TorreyPines Therapeutics), actemra, gemcitabine, oxaliplatin, L-asparaginase, or thalidomide.
  • the additional therapeutic agent is an anti-angiogenic agent, cholinergic agonist, TRP-1 receptor modulator, a calcium channel blocker, a mucin secretagogue, MUC 1 stimulant, a calcineurin inhibitor, a corticosteroid, a P2Y2 receptor agonist, a muscarinic receptor agonist, an mTOR inhibitor, another JAK inhibitor, Bcr-Abl kinase inhibitor, Flt-3 kinase inhibitor, RAF kinase inhibitor, and FAK kinase inhibitor such as, for example, those described in WO 2006/056399, which is incorporated herein by reference in its entirety.
  • the additional therapeutic agent is a tetracycline derivative (e.g., minocycline or doxycline).
  • the additional therapeutic agent binds to FKBP12.
  • the additional therapeutic agent is an alkylating agent or DNA cross-linking agent; an anti-metabolite/demethylating agent (e.g., 5-flurouracil, capecitabine or azacitidine); an anti-hormone therapy (e.g., hormone receptor antagonists, SERMs, or aromotase inhibitor); a mitotic inhibitor (e.g. vincristine or paclitaxel); an topoisomerase (I or II) inhibitor (e.g. mitoxantrone and irinotecan); an apoptotic inducers (e.g. ABT-737); a nucleic acid therapy (e.g.
  • an anti-metabolite/demethylating agent e.g., 5-flurouracil, capecitabine or azacitidine
  • an anti-hormone therapy e.g., hormone receptor antagonists, SERMs, or aromotase inhibitor
  • a mitotic inhibitor e.g. vincristine or paclitaxel
  • RNAi nuclear receptor ligands
  • nuclear receptor ligands e.g., agonists and/or antagonists: all-trans retinoic acid or bexarotene
  • epigenetic targeting agents such as histone deacetylase inhibitors (e.g. vorinostat), hypomethylating agents (e.g. decitabine); regulators of protein stability such as Hsp90 inhibitors, ubiquitin and/or ubiquitin like conjugating or deconjugating molecules; or an EGFR inhibitor (erlotinib).
  • the additional therapeutic agent(s) are demulcent eye drops (also known as "artificial tears"), which include, but are not limited to, compositions containing polyvinylalcohol, hydroxypropyl methylcellulose, glycerin, polyethylene glycol (e.g. PEG400), or carboxymethyl cellulose. Artificial tears can help in the treatment of dry eye by compensating for reduced moistening and lubricating capacity of the tear film.
  • the additional therapeutic agent is a mucolytic drug, such as N-acetyl-cysteine, which can interact with the mucoproteins and, therefore, to decrease the viscosity of the tear film.
  • the additional therapeutic agent includes an antibiotic, antiviral, antifungal, anesthetic, anti-inflammatory agents including steroidal and nonsteroidal anti-inflammatories, and anti-allergic agents.
  • suitable medicaments include aminoglycosides such as amikacin, gentamycin, tobramycin, streptomycin, netilmycin, and kanamycin; fluoroquinolones such as ciprofloxacin, norfloxacin, ofloxacin, trovafloxacin, lomefloxacin, levofloxacin, and enoxacin; naphthyridine;
  • rifampins sulfonamides
  • polymyxin chloramphenicol; neomycin; paramomycin; colistimethate; bacitracin; vancomycin; tetracyclines; rifampin and its derivatives ("rifampins");
  • cycloserine beta-lactams; cephalosporins; amphotericins; fluconazole; flucytosine; natamycin; miconazole; ketoconazole; corticosteroids; diclofenac; flurbiprofen;
  • ketorolac ketorolac
  • suprofen cromolyn
  • lodoxamide levocabastin
  • naphazoline antazoline
  • pheniramine or azalide antibiotic.
  • the compounds of the invention can be administered in the form of pharmaceutical compositions.
  • These compositions can be prepared in a manner well known in the pharmaceutical art, and can be administered by a variety of routes, depending upon whether local or systemic treatment is desired and upon the area to be treated. Administration may be topical (including transdermal, epidermal, ophthalmic and to mucous membranes including intranasal, vaginal and rectal delivery), pulmonary ⁇ e.g., by inhalation or insufflation of powders or aerosols, including by nebulizer; intratracheal or intranasal), oral or parenteral.
  • Parenteral administration includes intravenous, intraarterial, subcutaneous, intraperitoneal intramuscular or injection or infusion; or intracranial, e.g., intrathecal or intraventricular, administration.
  • Parenteral administration can be in the form of a single bolus dose, or may be, for example, by a continuous perfusion pump.
  • formulations for topical administration may include transdermal patches, ointments, lotions, creams, gels, drops, suppositories, sprays, liquids and powders.
  • Conventional pharmaceutical carriers, aqueous, powder or oily bases, thickeners and the like may be necessary or desirable.
  • This invention also includes pharmaceutical compositions which contain, as the active ingredient, the compound of the invention or a pharmaceutically acceptable salt thereof, in combination with one or more pharmaceutically acceptable carriers
  • composition is suitable for topical
  • the active ingredient is typically mixed with an excipient, diluted by an excipient or enclosed within such a carrier in the form of, for example, a capsule, sachet, paper, or other container.
  • an excipient serves as a diluent, it can be a solid, semi-solid, or liquid material, which acts as a vehicle, carrier or medium for the active ingredient.
  • compositions can be in the form of tablets, pills, powders, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, aerosols (as a solid or in a liquid medium), ointments containing, for example, up to 10% by weight of the active compound, soft and hard gelatin capsules, suppositories, sterile injectable solutions, and sterile packaged powders.
  • the active compound can be milled to provide the appropriate particle size prior to combining with the other ingredients. If the active compound is substantially insoluble, it can be milled to a particle size of less than 200 mesh. If the active compound is substantially water soluble, the particle size can be adjusted by milling to provide a substantially uniform distribution in the formulation, e.g. about 40 mesh.
  • the compounds of the invention may be milled using known milling procedures such as wet milling to obtain a particle size appropriate for tablet formation and for other formulation types.
  • Finely divided (nanoparticulate) preparations of the compounds of the invention can be prepared by processes known in the art, e.g., see International App. No. WO 2002/000196.
  • excipients include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, tragacanth, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, water, syrup, and methyl cellulose.
  • the formulations can additionally include: lubricating agents such as talc, magnesium stearate, and mineral oil; wetting agents; emulsifying and suspending agents; preserving agents such as methyl- and propylhydroxy-benzoates; sweetening agents; and flavoring agents.
  • the compositions of the invention can be formulated so as to provide quick, sustained or delayed release of the active ingredient after administration to the patient by employing procedures known in the art.
  • the pharmaceutical composition comprises silicified microcrystallme cellulose (SMCC) and at least one compound described herein, or a pharmaceutically acceptable salt thereof.
  • SMCC silicified microcrystallme cellulose
  • microcrystallme cellulose comprises about 98% microcrystallme cellulose and about 2% silicon dioxide w/w.
  • the composition is a sustained release composition comprising at least one compound described herein, or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable carrier.
  • the composition comprises at least one compound described herein, or a pharmaceutically acceptable salt thereof, and at least one component selected from microcrystallme cellulose, lactose monohydrate, hydroxypropyl methylcellulose, and polyethylene oxide.
  • the composition comprises at least one compound described herein, or a pharmaceutically acceptable salt thereof, and microcrystallme cellulose, lactose monohydrate, and hydroxypropyl methylcellulose.
  • the composition comprises at least one compound described herein, or a pharmaceutically acceptable salt thereof, and microcrystallme cellulose, lactose monohydrate, and polyethylene oxide.
  • the composition further comprises magnesium stearate or silicon dioxide.
  • the microcrystallme cellulose is Avicel PHI 02TM.
  • the lactose monohydrate is Fast-flo 316TM.
  • the hydroxypropyl methylcellulose is hydroxypropyl methylcellulose 2208 K4M (e.g., Methocel K4 M PremierTM) and/or hydroxypropyl methylcellulose 2208 K100LV (e.g., Methocel K00LVTM).
  • the polyethylene oxide is polyethylene oxide WSR 1105 (e.g., Polyox WSR 1105TM).
  • a wet granulation process is used to produce the composition. In some embodiments, a dry granulation process is used to produce the composition.
  • compositions can be formulated in a unit dosage form, each dosage containing from about 5 to about 1,000 mg (1 g), more usually about 100 mg to about 500 mg, of the active ingredient. In some embodiments, each dosage contains about 10 mg of the active ingredient. In some embodiments, each dosage contains about 50 mg of the active ingredient. In some embodiments, each dosage contains about 25 mg of the active ingredient.
  • unit dosage forms refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient.
  • the compositions of the invention contain from about 5 mg to about 50 mg of the active ingredient.
  • the active ingredient contains from about 5 mg to about 50 mg.
  • One having ordinary skill in the art will appreciate that this embodies compounds or compositions containing about 5 mg to about 10 mg, about 10 mg to about 15 mg, about 15 mg to about 20 mg, about 20 mg to about 25 mg, about 25 mg to about 30 mg, about 30 mg to about 35 mg, about 35 mg to about 40 mg, about 40 mg to about 45 mg, or about 45 mg to about 50 mg of the active ingredient.
  • the compositions of the invention contain from about 50 mg to about 500 mg of the active ingredient.
  • the active ingredient contains from about 50 mg to about 500 mg of the active ingredient.
  • One having ordinary skill in the art will appreciate that this embodies compounds or compositions containing about 50 mg to about 100 mg, about 100 mg to about 150 mg, about 150 mg to about 200 mg, about 200 mg to about 250 mg, about 250 mg to about 300 mg, about 350 mg to about 400 mg, or about 450 mg to about 500 mg of the active ingredient.
  • the compositions of the invention contain from about 500 mg to about 1 ,000 mg of the active ingredient.
  • the active ingredient One having ordinary skill in the art will appreciate that this embodies compounds or compositions containing about 500 mg to about 550 mg, about 550 mg to about 600 mg, about 600 mg to about 650 mg, about 650 mg to about 700 mg, about 700 mg to about 750 mg, about 750 mg to about 800 mg, about 800 mg to about 850 mg, about 850 mg to about 900 mg, about 900 mg to about 950 mg, or about 950 mg to about 1,000 mg of the active ingredient.
  • the active compound may be effective over a wide dosage range and is generally administered in a pharmaceutically effective amount. It will be understood, however, that the amount of the compound actually administered will usually be determined by a physician, according to the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound administered, the age, weight, and response of the individual patient, the severity of the patient's symptoms, and the like.
  • the principal active ingredient is mixed with a pharmaceutical excipient to form a solid preformulation composition containing a homogeneous mixture of a compound of the present invention.
  • a solid preformulation composition containing a homogeneous mixture of a compound of the present invention.
  • the active ingredient is typically dispersed evenly throughout the composition so that the composition can be readily subdivided into equally effective unit dosage forms such as tablets, pills and capsules.
  • This solid preformulation is then subdivided into unit dosage forms of the type described above containing from, for example, about 0.1 to about 1000 mg of the active ingredient of the present invention.
  • the tablets or pills of the present invention can be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action.
  • the tablet or pill can comprise an inner dosage and an outer dosage component, the latter being in the form of an envelope over the former.
  • the two components can be separated by an enteric layer which serves to resist disintegration in the stomach and permit the inner component to pass intact into the duodenum or to be delayed in release.
  • enteric layers or coatings such materials including a number of polymeric acids and mixtures of polymeric acids with such materials as shellac, cetyl alcohol, and cellulose acetate.
  • liquid forms in which the compounds and compositions of the present invention can be incorporated for administration orally or by injection include aqueous solutions, suitably flavored syrups, aqueous or oil suspensions, and flavored emulsions with edible oils such as cottonseed oil, sesame oil, coconut oil, or peanut oil, as well as elixirs and similar pharmaceutical vehicles.
  • compositions for inhalation or insufflation include solutions and suspensions in pharmaceutically acceptable, aqueous or organic solvents, or mixtures thereof, and powders.
  • the liquid or solid compositions may contain suitable pharmaceutically acceptable excipients as described supra.
  • the compositions are administered by the oral or nasal respiratory route for local or systemic effect.
  • compositions in can be nebulized by use of inert gases. Nebulized solutions may be breathed directly from the nebulizing device or the nebulizing device can be attached to a face masks tent, or intermittent positive pressure breathing machine. Solution, suspension, or powder compositions can be administered orally or nasally from devices which deliver the formulation in an appropriate manner.
  • Topical formulations can contain one or more conventional carriers.
  • ointments can contain water and one or more hydrophobic carriers selected from, for example, liquid paraffin, polyoxyethylene alkyl ether, propylene glycol, white Vaseline, and the like.
  • Carrier compositions of creams can be based on water in combination with glycerol and one or more other components, e.g.
  • topical formulations contain at least about 0.1, at least about 0.25, at least about 0.5, at least about 1 , at least about 2, or at least about 5 wt % of the compound of the invention.
  • the topical formulations can be suitably packaged in tubes of, for example, 100 g which are optionally associated with instructions for the treatment of the select indication, e.g., psoriasis or other skin condition.
  • compositions can be administered to a patient already suffering from a disease in an amount sufficient to cure or at least partially arrest the symptoms of the disease and its complications. Effective doses will depend on the disease condition being treated as well as by the judgment of the attending clinician depending upon factors such as the severity of the disease, the age, weight and general condition of the patient, and the like.
  • compositions administered to a patient can be in the form of pharmaceutical compositions described above. These compositions can be sterilized by conventional sterilization techniques, or may be sterile filtered. Aqueous solutions can be packaged for use as is, or lyophilized, the lyophilized preparation being combined with a sterile aqueous carrier prior to administration.
  • the pH of the compound preparations typically will be between 3 and 11, more preferably from 5 to 9 and most preferably from 7 to 8. It will be understood that use of certain of the foregoing excipients, carriers, or stabilizers will result in the formation of pharmaceutical salts.
  • the therapeutic dosage of a compound of the present invention can vary according to, for example, the particular use for which the treatment is made, the manner of administration of the compound, the health and condition of the patient, and the judgment of the prescribing physician.
  • the proportion or concentration of a compound of the invention in a pharmaceutical composition can vary depending upon a number of factors including dosage, chemical characteristics (e.g., hydrophobicity), and the route of administration.
  • the compounds of the invention can be provided in an aqueous physiological buffer solution containing about 0.1 to about 10% w/v of the compound for parenteral administration. Some typical dose ranges are from about 1 ⁇ g/kg to about 1 g/kg of body weight per day.
  • the dose range is from about 0.01 mg/kg to about 100 mg/kg of body weight per day.
  • the dosage is likely to depend on such variables as the type and extent of progression of the disease or disorder, the overall health status of the particular patient, the relative biological efficacy of the compound selected, formulation of the excipient, and its route of administration. Effective doses can be extrapolated from dose-response curves derived from in vitro or animal model test systems.
  • compositions of the invention can further include one or more additional pharmaceutical agents such as a chemotherapeutic, steroid, anti-inflammatory compound, or immunosuppressant, examples of which are listed hereinabove.
  • additional pharmaceutical agents such as a chemotherapeutic, steroid, anti-inflammatory compound, or immunosuppressant, examples of which are listed hereinabove.
  • the compound, or pharmaceutically acceptable salt thereof is administered as an ophthalmic composition.
  • the methods comprise administration of the compound, or pharmaceutically acceptable salt thereof, and an ophthalmically acceptable carrier.
  • the ophthalmic composition is a liquid composition, semi-solid composition, insert, film, microparticles or nanoparticles.
  • the ophthalmic composition is a liquid composition. In some embodiments, the ophthalmic composition is a semi-solid composition. In some embodiments, the ophthalmic composition is a topical composition.
  • the topical compositions include, but are not limited to liquid and semi-solid compositions. In some embodiments, the ophthalmic composition is a topical composition.
  • the topical composition comprises aqueous solution, an aqueous suspension, an ointment or a gel.
  • the ophthalmic composition is topically applied to the front of the eye, under the upper eyelid, on the lower eyelid and in the cul-de-sac.
  • the ophthalmic composition is sterilized. The sterilization can be accomplished by known techniques like sterilizing filtration of the solution or by heating of the solution in the ampoule ready for use.
  • the ophthalmic compositions of the invention can further contain pharmaceutical excipients suitable for the preparation of ophthalmic formulations. Examples of such excipients are preserving agents, buffering agents, chelating agents, antioxidant agents and salts for regulating the osmotic pressure.
  • the term "ophthalmically acceptable carrier” refers to any material that can contain and release the compound, or pharmaceutically acceptable salt thereof, and that is compatible with the eye.
  • the ophthalmically acceptable carrier is water or an aqueous solution or suspension, but also includes oils such as those used to make ointments and polymer matrices such as used in ocular inserts.
  • the composition may be an aqueous suspension comprising the compound, or pharmaceutically acceptable salt thereof.
  • Liquid ophthalmic compositions, including both ointments and suspensions may have a viscosity that is suited for the selected route of administration. In some embodiments, the ophthalmic composition has a viscosity in the range of from about 1,000 to about 30,000 centipoise.
  • the ophthalmic compositions may further comprise one or more of surfactants, adjuvants, buffers, antioxidants, tonicity adjusters, preservatives (e.g., EDTA, BAK (benzalkonium chloride), sodium chlorite, sodium perborate, polyquaterium-1), thickeners or viscosity modifiers (e.g., carboxymethyl cellulose, hydroxymethyl cellulose, polyvinyl alcohol, polyethylene glycol, glycol 400, propylene glycol hydroxymethyl cellulose, hydroxpropyl-guar, hyaluronic acid, and hydroxypropyl cellulose) and the like.
  • surfactants e.g., EDTA, BAK (benzalkonium chloride), sodium chlorite, sodium perborate, polyquaterium-1
  • thickeners or viscosity modifiers e.g., carboxymethyl cellulose, hydroxymethyl cellulose, polyvinyl alcohol, polyethylene glycol, glycol 400, propylene glycol hydroxymethyl cellulose
  • Additives in the formulation may include, but are not limited to, sodium chloride, sodium bicarbonate, sorbic acid, methyl paraben, propyl paraben, chlorhexidine, castor oil, and sodium perborate.
  • Aqueous ophthalmic compositions (solutions or suspensions) generally do not contain physiologically or ophthalmically harmful constituents.
  • purified or deionized water is used in the composition.
  • the pH may be adjusted by adding any physiologically and ophthalmically acceptable pH adjusting acids, bases or buffers to within the range of about 5.0 to 8.5.
  • Ophthalmically acceptable examples of acids include acetic, boric, citric, lactic, phosphoric, hydrochloric, and the like
  • bases include sodium hydroxide, sodium phosphate, sodium borate, sodium citrate, sodium acetate, sodium lactate, tromethamine, trishydroxymethylamino-methane, and the like.
  • Salts and buffers include citrate/dextrose, sodium bicarbonate, ammonium chloride and mixtures of the aforementioned acids and bases.
  • the methods involve forming or supplying a depot of the therapeutic agent in contact with the external surface of the eye.
  • a depot refers to a source of therapeutic agent that is not rapidly removed by tears or other eye clearance mechanisms. This allows for continued, sustained high concentrations of therapeutic agent to be present in the fluid on the external surface of the eye by a single application.
  • absorption and penetration may be dependent on both the dissolved drug concentration and the contact duration of the external tissue with the drug containing fluid. As the drug is removed by clearance of the ocular fluid and/or absorption into the eye tissue, more drug is provided, e.g.
  • the use of a depot may more easily facilitate loading of the ocular tissue for more insoluble therapeutic agents.
  • the depot can remain for up to eight hours or more.
  • the ophthalmic depot forms includes, but is not limited to, aqueous polymeric suspensions, ointments, and solid inserts.
  • the ophthalmic composition is an ointment or gel.
  • the ophthalmic composition is an oil-based delivery vehicle.
  • the composition comprises a petroleum or lanolin base to which is added the active ingredient, usually as 0.1 to 2%, and excipients. Common bases may include, but are not limited to, mineral oil, petrolatum and combinations thereof.
  • the ointment is applied as a ribbon onto the lower eyelid.
  • the ophthalmic composition is an ophthalmic insert.
  • the ophthalmic insert is biologically inert, soft, bio-erodible, viscoelastic, stable to sterilization after exposure to therapeutic agents, resistant to infections from air borne bacteria, bio- erodible, biocompatible, and/or viscoelastic.
  • the insert comprises an ophthalmically acceptable matrix, e.g., a polymer matrix.
  • the matrix is typically a polymer and the therapeutic agent is generally dispersed therein or bonded to the polymer matrix.
  • the therapeutic agent may be slowly released from the matrix through dissolution or hydrolysis of the covalent bond.
  • the polymer is bioerodible (soluble) and the dissolution rate thereof can control the release rate of the therapeutic agent dispersed therein.
  • the polymer matrix is a biodegradable polymer that breaks down such as by hydrolysis to thereby release the therapeutic agent bonded thereto or dispersed therein.
  • the matrix and therapeutic agent can be surrounded with an additional polymeric coating to further control release.
  • the insert comprises a biodegradable polymer such as polycaprolactone (PCL), an
  • the therapeutic agent is dispersed into the matrix material or dispersed amongst the monomer composition used to make the matrix material prior to
  • the amount of therapeutic agent is from about 0.1 to about 50%, or from about 2 to about 20%.
  • the biodegradable or bioerodible polymer matrix is used so that the spent insert does not have to be removed. As the biodegradable or bioerodible polymer is degraded or dissolved, the therapeutic agent is released.
  • the ophthalmic insert comprises a polymer, including, but are not limited to, those described in Wagh, et al, "Polymers used in ocular dosage form and drug delivery systems", Asian J. Pharm., pages 12-17 (Jan. 2008), which is incorporated herein by reference in its entirety.
  • the insert comprises a polymer selected from polyvinylpyrrolidone (PVP), an acrylate or methacrylate polymer or copolymer (e.g., Eudragit® family of polymers from Rohm or Degussa), hydroxymethyl cellulose, polyacrylic acid, poly(amidoamine) dendrimers, poly(dimethyl siloxane), polyethylene oxide, poly(lactide-co-glycolide), poly(2- hydroxyethylmethacrylate), poly( vinyl alcohol), or poly(propylene fumarate).
  • the insert comprises Gelfoam® R.
  • the insert is a polyacrylic acid of 450 kDa-cysteine conjugate.
  • the ophthalmic composition is a ophthalmic film.
  • the film is a soft-contact lens, such as ones made from copolymers of ⁇ , ⁇ -diethylacrylamide and methacrylic acid crosslinked with ethyleneglycol dimethacrylate.
  • the ophthalmic compositon comprises microspheres or nanoparticles.
  • the microspheres comprise gelatin.
  • the microspheres are injected to the posterior segment of the eye, in the chroroidal space, in the sclera, intravitreally or sub-retinally.
  • the microspheres or nanoparticles comprises a polymer including, but not limited to, those described in Wagh, et al. (ibid), which is incorporated herein by reference in its entirety.
  • the polymer is chitosan, a polycarboxylic acid such as polyacrylic acid, albumin particles, hyaluronic acid esters, polyitaconic acid,
  • the microspheres or nanoparticles comprise solid lipid particles.
  • the ophthalmic composition comprises an ion-exchange resin.
  • the ion-exchange resin is an inorganic zeolite or synthetic organic resin.
  • the ion-exchange resin includes, but is not limited to, those described in Wagh, et al. (ibid), which is incorporated herein by reference in its entirety.
  • the ion-exhange resin is a partially neutralized polyacrylic acid.
  • the ophthalmic composition is an aqueous polymeric suspension.
  • the therapeutic agent or a polymeric suspending agent is suspended in an aqueous medium.
  • the aqueous polymeric suspensions may be formulated so that they retain the same or substantially the same viscosity in the eye that they had prior to administration to the eye. In some embodiments, they may be formulated so that there is increased gelation upon contact with tear fluid.
  • Another aspect of the present invention relates to labeled compounds of the invention (radio-labeled, fluorescent-labeled, etc.) that would be useful not only in imaging techniques but also in assays, both in vitro and in vivo, for localizing and quantitating JAK in tissue samples, including human, and for identifying JAK ligands by inhibition binding of a labeled compound. Accordingly, the present invention includes JAK assays that contain such labeled compounds.
  • the present invention further includes isotopically-labeled compounds of the invention.
  • An “isotopically” or “radio-labeled” compound is a compound of the invention where one or more atoms are replaced or substituted by an atom having an atomic mass or mass number different from the atomic mass or mass number typically found in nature (i.e., naturally occurring). Suitable radionuclides that may be
  • incorporated in compounds of the present invention include but are not limited to 3 H (also written as T for tritium), U C, 13 C, 14 C, 13 N, 15 N, 15 0, 17 0, 18 0, 18 F, 35 S, 36 C1, 82 Br, 75 Br,
  • radionuclide that is incorporated in the instant radio-labeled compounds will depend on the specific application of that radio-labeled compound. For example, for in vitro JAK labeling and competition assays, compounds
  • a “radio-labeled” or “labeled compound” is a compound that has incorporated at least one radionuclide. In some embodiments the
  • radionuclide is selected from the group consisting of H, C, I , S and Br.
  • the compound incorporates 1, 2, or 3 deuterium atoms.
  • the present invention can further include synthetic methods for incorporating radio-isotopes into compounds of the invention.
  • Synthetic methods for incorporating radio-isotopes into organic compounds are well known in the art, and an ordinary skill in the art will readily recognize the methods applicable for the compounds of invention.
  • a labeled compound of the invention can be used in a screening assay to identify/evaluate compounds. For example, a newly synthesized or identified compound (i.e., test compound) which is labeled can be evaluated for its ability to bind a JAK by monitoring its concentration variation when contacting with the JAK, through tracking of the labeling.
  • a test compound can be evaluated for its ability to reduce binding of another compound which is known to bind to a JAK (i.e., standard compound). Accordingly, the ability of a test compound to compete with the standard compound for binding to the JAK directly correlates to its binding affinity. Conversely, in some other screening assays, the standard compound is labeled and test compounds are unlabeled. Accordingly, the concentration of the labeled standard compound is monitored in order to evaluate the competition between the standard compound and the test compound, and the relative binding affinity of the test compound is thus ascertained.
  • kits useful for example, in the treatment or prevention of JAK-associated diseases or disorders, such as cancer, which include one or more containers containing a pharmaceutical composition comprising a therapeutically effective amount of a compound of the invention.
  • kits can further include, if desired, one or more of various conventional pharmaceutical kit components, such as, for example, containers with one or more pharmaceutically acceptable carriers, additional containers, etc., as will be readily apparent to those skilled in the art.
  • kits can also be included in the kit.
  • Step A tert-Butyl 3-Oxoazetidine-l-carboxylate
  • Step B tert-Butyl 3-(Cyanomethylene)azetidine-l-carboxylate
  • the reaction was diluted with 200 mL of 20% brine and 250 mL of EtOAc. The solution was partitioned and the aqueous phase was extracted with 250 mL of EtOAc. The combined organic phase was dried over MgSC ⁇ and filtered, evaporated under reduced pressure, and purified by flash chromatography ( 0% to 20% EtOAc/hexanes, 150 g flash column) to give the desired product, tert-butyl 3-(cyanomethylene)azetidine-l-carboxylate (15 g, 66.1% yield).
  • Step C 4-Chloro-7- 2-(trimethylsilyl)ethoxyJmethyl ⁇ -7H ⁇ yrrolo[2 -dJpyrimidine
  • tetrakis(triphenylphosphine)palladium(0) (4.071 g, 3.523 mmol).
  • the solution was degased 4 times, filling with nitrogen each time.
  • the mixture was stirred overnight at 100 °C. After being cooled to room temperature, the mixture was filtered through a bed of celite and the celite was rinsed with ethyl acetate (42 mL). The filtrate was combined, and the organic layer was separated. The aqueous layer was extracted with ethyl acetate.
  • Step F ⁇ 3-f 4- (7 - ⁇ [2- (Trimethylsilyl) ethoxy] methyl ⁇ -7 H-pyrrolo[ 2, 3-d] pyrimidin-4-yl)- lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile dihydrochloride
  • Step G tert-Butyl 4- ⁇ 3-(Cyanomethyl)-3-[4-(7- ⁇ [2-(trimethylsilyl) ethoxy] methyl ⁇ -7 H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-l-yl ⁇ piperidine-l-carb
  • Step H ⁇ l-Piperidin-4-yl-3-[4-(7- ⁇ [2-(trimethylsilyl)ethoxy]methyl ⁇ -7H-pyrrolo[2,3- d] pyrimidin-4-yl)- lH-pyrazol- 1-yl] azetidin-3-yl ⁇ acetonitrile trih drochloride
  • Step I ⁇ l- ⁇ l-[ 3-Fluoro-2-(trifluoromethyl)isonicotinoyl] piperidin-4-yl ⁇ -3- [ 4-(7- ⁇ [ 2- (trimethylsilyl)ethoxy]methyl ⁇ -7H-pyrrolo[2,3-d]pyrimidin-4-yl)
  • Step J ⁇ l- ⁇ l-[3-Fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile
  • Step A ⁇ 4-[(4- ⁇ 3-(Cyanomethyl)-3-[4-(7- ⁇ [2-(trimethylsilyl)ethoxy]methyl ⁇ -7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-l-yl ⁇ pipe
  • Step B ⁇ l-[l-(3-Fluoro-4-quinolin-6-ylbenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile
  • Example 1 The following compounds were prepared by a method analogous to that for Example 1 or Example 2.

Abstract

The present invention provides piperidin-4-yl azetidine derivatives, as well as their compositions and methods of use, that modulate the activity of Janus kinase 1 (JAK1) and are useful in the treatment of diseases related to the activity of JAK1 including, for example, inflammatory disorders, autoimmune disorders, cancer, and other diseases.

Description

PIPERIDIN-4-YL AZETIDINE DERIVATIVES AS JAK1 INHIBITORS
TECHNICAL FIELD
The present invention provides piperidin-4-yl azetidine derivatives, as well as their compositions and methods of use, that modulate the activity of Janus kinase 1 (JAK1) and are useful in the treatment of diseases related to the activity of JAK1 including, for example, inflammatory disorders, autoimmune disorders, cancer, and other diseases.
BACKGROUND
Protein kinases (PKs) regulate diverse biological processes including cell growth, survival, differentiation, organ formation, morphogenesis, neovascularization, tissue repair, and regeneration, among others. Protein kinases also play specialized roles in a host of human diseases including cancer. Cytokines, low-molecular weight polypeptides or glycoproteins, regulate many pathways involved in the host inflammatory response to sepsis. Cytokines influence cell differentiation, proliferation and activation, and can modulate both pro-inflammatory and anti-inflammatory responses to allow the host to react appropriately to pathogens. Signaling of a wide range of cytokines involves the Janus kinase family (JAKs) of protein tyrosine kinases and Signal Transducers and Activators of Transcription (STATs). There are four known mammalian JAKs: JAK1 (Janus kinase- 1), JAK2, JAK3 (also known as Janus kinase, leukocyte; JAKL; and L- JAK), and TYK2 (protein-tyrosine kinase 2).
Cytokine-stimulated immune and inflammatory responses contribute to pathogenesis of diseases: pathologies such as severe combined immunodeficiency (SCID) arise from suppression of the immune system, while a hyperactive or
inappropriate immune/inflammatory response contributes to the pathology of
autoimmune diseases (e.g., asthma, systemic lupus erythematosus, thyroiditis, myocarditis), and illnesses such as scleroderma and osteoarthritis (Ortmann, R. A., T. Cheng, et al. (2000) Arthritis Res 2(1): 16-32).
Deficiencies in expression of JAKs are associated with many disease states. For example, Jakl-/- mice are runted at birth, fail to nurse, and die perinatally (Rodig, S. J., M. A. Meraz, et al. (1998) Cell 93(3): 373-83). Jak2-/- mouse embryos are anemic and die around day 12.5 postcoitum due to the absence of definitive erythropoiesis.
The JAK/STAT pathway, and in particular all four JAKs, are believed to play a role in the pathogenesis of asthmatic response, chronic obstructive pulmonary disease, bronchitis, and other related inflammatory diseases of the lower respiratory tract.
Multiple cytokines that signal through JAKs have been linked to inflammatory diseases/conditions of the upper respiratory tract, such as those affecting the nose and sinuses {e.g., rhinitis and sinusitis) whether classically allergic reactions or not. The JAK/STAT pathway has also been implicated in inflammatory diseases/conditions of the eye and chronic allergic responses.
Activation of JAK STAT in cancers may occur by cytokine stimulation {e.g. IL-6 or GM-CSF) or by a reduction in the endogenous suppressors of JAK signaling such as SOCS (suppressor or cytokine signaling) or PIAS (protein inhibitor of activated STAT) (Boudny, V., and Kovarik, J., Neoplasm. 49:349-355, 2002). Activation of STAT signaling, as well as other pathways downstream of JAKs {e.g., Akt), has been correlated with poor prognosis in many cancer types (Bowman, T., et al. Oncogene 19:2474-2488, 2000). Elevated levels of circulating cytokines that signal through JAK/STAT play a causal role in cachexia and/or chronic fatigue. As such, JAK inhibition may be beneficial to cancer patients for reasons that extend beyond potential anti-tumor activity.
JAK2 tyrosine kinase can be beneficial for patients with myeloproliferative disorders, e.g., polycythemia vera (PV), essential thrombocythemia (ET), myeloid metaplasia with myelofibrosis (MMM) (Levin, et al, Cancer Cell, vol. 7, 2005: 387- 397). Inhibition of the JAK2V617F kinase decreases proliferation of hematopoietic cells, suggesting JAK2 as a potential target for pharmacologic inhibition in patients with PV, ET, and MMM.
Inhibition of the JAKs may benefit patients suffering from skin immune disorders such as psoriasis, and skin sensitization. The maintenance of psoriasis is believed to depend on a number of inflammatory cytokines in addition to various chemokines and growth factors (JCI, 113: 1664-1675), many of which signal through JAKs {Adv
Pharmacol. 2000;47: 113-74). Thus, new or improved agents which inhibit kinases such as JAKs are continually needed for developing new and more effective pharmaceuticals that are aimed at augmentation or suppression of the immune and inflammatory pathways (such as immunosuppressive agents for organ transplants), as well as agents for the prevention and treatment of autoimmune diseases, diseases involving a hyperactive inflammatory response (e.g., eczema), allergies, cancer (e.g., prostate, leukemia, multiple myeloma), and some immune reactions (e.g. , skin rash or contact dermatitis or diarrhea) caused by other therapeutics. The compounds of the invention, as well as its compositions and methods described herein are directed toward these needs and other ends.
SUMMARY
The present invention provides, inter alia, compounds of Formula (I):
Figure imgf000004_0001
I
or pharmaceutically acceptable salts thereof; wherein the variables are defined infra.
The present invention further provides compositions comprising a compound of Formula I, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
The present invention further provides methods of modulating an activity of JAK1 comprising contacting JAK1 with a compound of Formula I, or a pharmaceutically acceptable salt thereof.
The present invention further provides methods of treating a disease or a disorder associated with abnormal kinase expression or activity in a patient by administering to a patient a therapeutically effective amount of a compound of Formula I, or a
pharmaceutically acceptable salt thereof.
The present invention further provides methods of treating an autoimmune disease, a cancer, a myeloproliferative disorder, an inflammatory disease, a bone resorption disease, or organ transplant rejection in a patient in need thereof, comprising administering to said patient a therapeutically effective amount of a compound of Formula I, or a pharmaceutically acceptable salt thereof.
The present invention also provides compounds of Formula I, or pharmaceutically acceptable salts thereof, as described herein for use in treatment of autoimmune diseases, cancer, myeloproliferative disorders, inflammatory diseases, a bone resorption disease, or organ transplant rejection.
The present invention further provides compounds of Formula I as described herein, or pharmaceutically acceptable salts thereof, for use in modulating JAK1.
The present invention also provides uses of compounds of Formula I as described herein, or pharmaceutically acceptable salts thereof, for the preparation of medicaments for use in methods of modulating JAK1.
The details of one or more embodiments of the invention are set forth in the accompanying drawings and the description below. Other features, objects, and advantages of the invention will be apparent from the description and drawings, and from the claims.
BRIEF DESCRIPTION OF THE FIGURES
FIG. 1 depicts the DSC thermogram for the product of Example 358.
FIG. 2 depicts the TGA thermogram for the product of Example 358.
FIG. 3 depicts the XRPD pattern for the product of Example 358.
DETAILED DESCRIPTION
The present invention provides, inter alia, a compound of Formula (I):
Figure imgf000006_0001
I
or a pharmaceutically acceptable salt thereof; wherein:
X is N or CR2;
Y is N or CR3;
Z is H, cyano, halo, Ci_3 alkyl, or Ci_3 haloalkyl;
L is C(R4)2, C(=0), C(=0)N(R4a), C(=0)C(R4b)2, S(=0)2, C(=0)0,
C(=0)OC(R4b)2 or C(=0)N(R4a)C(R4b)2;
A is Ci_6 alkyl, C3_i4 cycloalkyl, C2_i3 heterocycloalkyl, C6-i4 aryl, or Ci_i4 heteroaryl; wherein said Ci_6 alkyl, C3_i4 cycloalkyl, C2_i3 heterocycloalkyl, C6_i4 aryl, and Ci_i4 heteroaryl are each optionally substituted with 1, 2, 3, 4, 5, or 6 independently selected R5 groups;
each R1 is, independently, Ci_4 alkyl, hydroxyl, Ci_4 alkoxy, fluoro, hydroxyl-Ci_4 alkyl, or Ci_4 alkoxy-Ci_4 alkyl; or
two R1 groups together form a 2- or 3 -carbon bridge or a bridge of formula -CH2- 0-CH2-;
R2 is H, halo, hydroxyl, cyano, Ci_4 alkyl, Ci_4 haloalkyl, or Ci_4 alkoxy;
R3 is H, cyano, nitro, halo, hydroxyl, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, C3_7 cycloalkyl, Ci_6 haloalkyl, Ci_6 alkoxy, Ci_6 haloalkoxy, amino, Ci_6 alkylamino, di-Ci_6 alkylamino, Ci_6 alkylsulfonyl, carbamyl, Ci_6 alkylcarbamyl, di-Ci_6 alkylcarbamyl, carboxy, Ci_6 alkylcarbonyl, or Ci_6 alkoxycarbonyl;
each R4 is, independently, H or Ci_4 alkyl; or two R4 groups, together with the carbon atom to which they are attached, form a 3-, 4-, 5-, or 6-membered cycloalkyl ring;
R4a is H or Ci_4 alkyl;
each R4b is, independently, H or Ci_4 alkyl; or
two R4b groups, together with the carbon atom to which they are attached, form a 3-, 4-, 5-, or 6-membered cycloalkyl ring;
each R5 is, independently, halo, cyano, nitro, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, Ci_6 haloalkyl, halosulfanyl, C3-10 cycloalkyl, C3-10 cycloalkyl-Ci_3 alkyl, C2-10
heterocycloalkyl, C2_io heterocycloalkyl-Ci_3 alkyl, C6-io aryl, C6-io aryl-Ci_3 alkyl, Ci_io heteroaryl, CM0 heteroaryl-Ci_3 alkyl, ORa, SRa, C(0)Rb, C(0)NRcRd, C(0)ORa,
OC(0)Rb, OC(0)NRcRd, NRcRd, NRcC(0)Rb, NRcC(0)ORa, NRcC(0)NRcRd,
C(=NRe)Rb, C(=NRe)NRcRd, NRcC(=NRe)NRcRd, NRcS(0)Rb, NRcS(0)2Rb,
NRcS(0)2NRcRd, S(0)Rb, S(0)NRcRd, S(0)2Rb, or S(0)2NRcRd; wherein Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, Ci_6 haloalkyl, C3_i0 cycloalkyl, C3_i0 cycloalkyl-Ci_3 alkyl, C2_i0 heterocycloalkyl, C2_io heterocycloalkyl-Ci_3 alkyl, C6-io aryl, C6-io aryl-Ci_3 alkyl, Ci_io heteroaryl, and Ci_io heteroaryl-Ci_3 alkyl are each optionally substituted with 1, 2, 3, 4, or 5 independently selected R6 groups;
each R6 is, independently, halo, cyano, nitro, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, Ci_6 haloalkyl, halosulfanyl, C3_i0 cycloalkyl, C3_i0 cycloalkyl-Ci_3 alkyl, C2_i0
heterocycloalkyl, C2_io heterocycloalkyl-Ci_3 alkyl, C6-io aryl, C6-io aryl-Ci_3 alkyl, Ci_io heteroaryl, CM0 heteroaryl-Ci_3 alkyl, ORal, SRal, C(0)Rbl, C(0)NRclRdl, C(0)ORal, OC(0)Rbl, OC(0)NRclRdl, C(=NRel)NRclRdl, NRclC(=NRel)NRclRdl, NRclRdl, NRclC(0)Rbl, NRclC(0)ORal, NRclC(0)NRclRdl, NRclS(0)Rbl, NRclS(0)2Rbl,
NRclS(0)2NRclRdl, S(0)Rbl, S(0)NRclRdl, S(0)2Rbl, or S(0)2NRclRdl; wherein said Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, Ci_6 haloalkyl, C3-10 cycloalkyl, C3-10 cycloalkyl-Ci_3 alkyl, C2_io heterocycloalkyl, C2_io heterocycloalkyl-Ci_3 alkyl, C6-io aryl, C6-io aryl-Ci_3 alkyl, C1-10 heteroaryl, and Ci_i0 heteroaryl-Ci_3 alkyl are each optionally substituted by 1, 2, 3, 4, or 5 independently selected Rh groups;
each Ra, Rb, Rc, and Rd is, independently, H, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, Ci_6 haloalkyl, C3-10 cycloalkyl, C3-10 cycloalkyl-Ci_3 alkyl, C2_io heterocycloalkyl, C2_io heterocycloalkyl-Ci_3 alkyl, C6_io aryl, C6_io aryl-Ci_3 alkyl, C1-10 heteroaryl, or C1-10 heteroaryl-Ci_3 alkyl; wherein said Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, Ci_6 haloalkyl, halosulfanyl, C3_i0 cycloalkyl, C3_i0 cycloalkyl-Ci_3 alkyl, C2_i0 heterocycloalkyl, C2_i0 heterocycloalkyl-Ci_3 alkyl, C6-io aryl, C6-io aryl-Ci_3 alkyl, Ci_io heteroaryl, and Ci_io heteroaryl-Ci_3 alkyl is optionally substituted with 1 , 2, 3, 4, or 5 independently selected Rg groups;
or any Rc and Rd together with the N atom to which they are attached form a 3-, 4- , 5-, 6-, or 7-membered heterocycloalkyl group or a heteroaryl group, each optionally substituted with 1 , 2, 3 or 4 substituents independently selected from halo, cyano, nitro, hydroxyl, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, C3_7 cycloalkyl, Ci_6 haloalkyl, Ci_6 alkoxy, Ci_6 haloalkoxy, amino, Ci_6 alkylamino, di-Ci_6 alkylamino, thio, Ci_6 alkylthio, Ci_6 alkylsulfinyl, Ci_6 alkylsulfonyl, carbamyl, Ci_6 alkylcarbamyl, di-Ci_6 alkylcarbamyl, carboxy, Ci_6 alkylcarbonyl, and Ci_6 alkoxycarbonyl;
each Re is, independently, H, Ci_6 alkyl, CN, hydroxyl, Ci_6 alkoxy, Ci_6 alkylsulfonyl, carboxy, Ci_6 alkylcarbonyl, amino sulfonyl, Ci_6 alkylaminosulfonyl, di-Ci_ 6 alkylaminosulfonyl, carbamyl, Ci_6 alkylcarbamyl, or di-Ci_6 alkylcarbamyl;
each Ral, Rbl, Rcl, and Rdl is, independently, H, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, Ci_6 haloalkyl, C3_io cycloalkyl, C3_io cycloalkyl-Ci_3 alkyl, C2_io heterocycloalkyl, C2_io heterocycloalkyl-Ci_3 alkyl, C6_io aryl, C6_io aryl-Ci_3 alkyl, Ci_i0 heteroaryl, or Ci_i0 heteroaryl-Ci_3 alkyl; wherein said Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, Ci_6 haloalkyl, halosulfanyl, C3_io cycloalkyl, C3_io cycloalkyl-Ci_3 alkyl, C2_io heterocycloalkyl, C2_io heterocycloalkyl-Ci_3 alkyl, C6-io aryl, C6-io aryl-Ci_3 alkyl, Ci_io heteroaryl, and Ci_io heteroaryl-Ci_3 alkyl is optionally substituted with 1 , 2, 3, 4, or 5 independently selected Rg groups;
or any Rcl and Rdl together with the N atom to which they are attached form a 3-, 4-, 5-, 6-, or 7-membered heterocycloalkyl group or a heteroaryl group, each optionally substituted with 1 , 2, 3 or 4 substituents independently selected from halo, cyano, nitro, hydroxyl, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, C3_7 cycloalkyl, Ci_6 haloalkyl, Ci_6 alkoxy, Ci_6 haloalkoxy, amino, Ci_6 alkylamino, di-Ci_6 alkylamino, thio, Ci_6 alkylthio, Ci_6 alkylsulfinyl, Ci_6 alkylsulfonyl, carbamyl, Ci_6 alkylcarbamyl, di-Ci_6 alkylcarbamyl, carboxy, Ci_6 alkylcarbonyl, Ci_6 alkoxycarbonyl, and Ci_6 alkylcarbonylamino; each Rel is, independently, H, Ci_6 alkyl, CN, hydroxyl, Ci_6 alkoxy, Ci_6 alkylsulfonyl, carboxy, Ci_6 alkylcarbonyl, amino sulfonyl, Ci_6 alkylaminosulfonyl, di-Ci_ 6 alkylaminosulfonyl, carbamyl, Ci_6 alkylcarbamyl, or di-Ci_6 alkylcarbamyl;
each Rg, Rg , and Rh is, independently, halo, cyano, nitro, hydroxyl, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, C3-7 cycloalkyl, Ci_6 haloalkyl, Ci_6 alkoxy, Ci_6 haloalkoxy, amino, Ci_6 alkylamino, di-Ci_6 alkylamino, thio, Ci_6 alkylthio, Ci_6 alkylsulfmyl, Ci_6 alkylsulfonyl, carbamyl, Ci_6 alkylcarbamyl, di-Ci_6 alkylcarbamyl, carboxy, Ci_6 alkylcarbonyl, Ci_6 alkoxycarbonyl, or Ci_6 alkylcarbonylamino; and
n is 0, 1, 2, 3, or 4.
In some embodiments:
X is N or CR2;
Y is N or CR3;
Z is H, cyano, halo, Ci_3 alkyl, or Ci_3 haloalkyl;
L is C(R4)2, C(=0), C(=0)N(R4a), C(=0)C(R4b)2, or S(=0)2;
A is Ci_6 alkyl, C3_i4 cycloalkyl, C2_i3 heterocycloalkyl, C6-i4 aryl, or Ci_i4 heteroaryl; wherein said Ci_6 alkyl, C3_i4 cycloalkyl, C2_i3 heterocycloalkyl, C6-i4 aryl, and Ci_i4 heteroaryl are each optionally substituted with 1, 2, 3, 4, 5, or 6 independently selected R5 groups;
each R1 is, independently, Ci_4 alkyl; or
two R1 groups together form a 2- or 3 -carbon bridge;
R2 is H, halo, hydroxyl, cyano, Ci_4 alkyl, Ci_4 haloalkyl, or Ci_4 alkoxy;
R3 is H, cyano, nitro, halo, hydroxyl, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, C3_7 cycloalkyl, Ci_6 haloalkyl, Ci_6 alkoxy, Ci_6 haloalkoxy, amino, Ci_6 alkylamino, di-Ci_6 alkylamino, Ci_6 alkylsulfonyl, carbamyl, Ci_6 alkylcarbamyl, di-Ci_6 alkylcarbamyl, carboxy, Ci_6 alkylcarbonyl, or Ci_6 alkoxycarbonyl;
each R4 is, independently, H or Ci_4 alkyl; or
two R4 groups, together with the carbon atom to which they are attached, form a 3-, 4-, 5-, or 6-membered cycloalkyl ring;
R4a is H or Ci_4 alkyl;
each R4b is, independently, H or Ci_4 alkyl; or two R groups, together with the carbon atom to which they are attached, form a 3-, 4-, 5-, or 6-membered cycloalkyl ring;
each R5 is, independently, halo, cyano, nitro, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, Ci_6 haloalkyl, halosulfanyl, C3-10 cycloalkyl, C3-10 cycloalkyl-Ci_3 alkyl, C2-10
heterocycloalkyl, C2_io heterocycloalkyl-Ci_3 alkyl, C6-io aryl, C6-io aryl-Ci_3 alkyl, Ci_io heteroaryl, CM0 heteroaryl-Ci_3 alkyl, ORa, SRa, C(0)Rb, C(0)NRcRd, C(0)ORa,
OC(0)Rb, OC(0)NRcRd, NRcRd, NRcC(0)Rb, NRcC(0)ORa, NRcC(0)NRcRd,
C(=NRe)Rb, C(=NRe)NRcRd, NRcC(=NRe)NRcRd, NRcS(0)Rb, NRcS(0)2Rb,
NRcS(0)2NRcRd, S(0)Rb, S(0)NRcRd, S(0)2Rb, or S(0)2NRcRd; wherein Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, Ci_6 haloalkyl, C3_i0 cycloalkyl, C3_i0 cycloalkyl-Ci_3 alkyl, C2_i0 heterocycloalkyl, C2_io heterocycloalkyl-Ci_3 alkyl, C6-io aryl, C6-io aryl-Ci_3 alkyl, Ci_io heteroaryl, and Ci_io heteroaryl-Ci_3 alkyl are each optionally substituted with 1, 2, 3, 4, or 5 independently selected R6 groups;
each R6 is, independently, halo, cyano, nitro, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, Ci_6 haloalkyl, halosulfanyl, C3-10 cycloalkyl, C3-10 cycloalkyl-Ci_3 alkyl, C2_io
heterocycloalkyl, C2_io heterocycloalkyl-Ci_3 alkyl, C6-io aryl, C6-io aryl-Ci_3 alkyl, Ci_io heteroaryl, CM0 heteroaryl-Ci_3 alkyl, ORal, SRal, C(0)Rbl, C(0)NRclRdl, C(0)ORal, OC(0)Rbl, OC(0)NRclRdl, C(=NRel)NRclRdl, NRclC(=NRel)NRclRdl, NRclRdl, NRclC(0)Rbl, NRclC(0)ORal, NRclC(0)NRclRdl, NRclS(0)Rbl, NRclS(0)2Rbl,
NRclS(0)2NRclRdl, S(0)Rbl, S(0)NRclRdl, S(0)2Rbl, or S(0)2NRclRdl; wherein said Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, Ci_6 haloalkyl, C3-10 cycloalkyl, C3-10 cycloalkyl-Ci_3 alkyl, C2_io heterocycloalkyl, C2_io heterocycloalkyl-Ci_3 alkyl, C6-io aryl, C6-io aryl-Ci_3 alkyl, C1-10 heteroaryl, and Ci_i0 heteroaryl-Ci_3 alkyl are each optionally substituted by 1, 2, 3, 4, or 5 independently selected Rh groups;
each Ra, Rb, Rc, and Rd is, independently, H, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, Ci_6 haloalkyl, C3-10 cycloalkyl, C3-10 cycloalkyl-C 1-3 alkyl, C2_io heterocycloalkyl, C2_io heterocycloalkyl-Ci_3 alkyl, C6_io aryl, C6_io aryl-Ci_3 alkyl, C1-10 heteroaryl, or C1-10 heteroaryl-Ci_3 alkyl; wherein said Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, Ci_6 haloalkyl, halosulfanyl, C3-10 cycloalkyl, C3-10 cycloalkyl-C 1-3 alkyl, C2_io heterocycloalkyl, C2_io heterocycloalkyl-Ci_3 alkyl, C6-io aryl, C6-io aryl-Ci_3 alkyl, C1-10 heteroaryl, and C1-10 heteroaryl-Ci_3 alkyl is optionally substituted with 1 , 2, 3, 4, or 5 independently selected Rg groups;
or any Rc and Rd together with the N atom to which they are attached form a 3-, 4- , 5-, 6-, or 7-membered heterocycloalkyl group or a heteroaryl group, each optionally substituted with 1 , 2, 3 or 4 substituents independently selected from halo, cyano, nitro, hydroxyl, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, C3_7 cycloalkyl, Ci_6 haloalkyl, Ci_6 alkoxy, Ci_6 haloalkoxy, amino, Ci_6 alkylamino, di-Ci_6 alkylamino, thio, Ci_6 alkylthio, Ci_6 alkylsulfinyl, Ci_6 alkylsulfonyl, carbamyl, Ci_6 alkylcarbamyl, di-Ci_6 alkylcarbamyl, carboxy, Ci_6 alkylcarbonyl, and Ci_6 alkoxycarbonyl;
each Re is, independently, H, Ci_6 alkyl, CN, hydroxyl, Ci_6 alkoxy, Ci_6 alkylsulfonyl, carboxy, Ci_6 alkylcarbonyl, amino sulfonyl, Ci_6 alkylaminosulfonyl, di-Ci_ 6 alkylaminosulfonyl, carbamyl, Ci_6 alkylcarbamyl, or di-Ci_6 alkylcarbamyl;
each Ral, Rbl, Rcl, and Rdl is, independently, H, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, Ci_6 haloalkyl, C3_i0 cycloalkyl, C3_i0 cycloalkyl-Ci_3 alkyl, C2_i0 heterocycloalkyl, C2_io heterocycloalkyl-Ci_3 alkyl, C6-io aryl, C6-io aryl-Ci_3 alkyl, Ci_io heteroaryl, or Ci_io heteroaryl-Ci_3 alkyl; wherein said Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, Ci_6 haloalkyl, halosulfanyl, C3_io cycloalkyl, C3_io cycloalkyl-Ci_3 alkyl, C2_io heterocycloalkyl, C2_io heterocycloalkyl-Ci_3 alkyl, C6_io aryl, C6_io aryl-Ci_3 alkyl, Ci_i0 heteroaryl, and Ci_i0 heteroaryl-Ci_3 alkyl is optionally substituted with 1 , 2, 3, 4, or 5 independently selected Rg groups;
or any Rcl and Rdl together with the N atom to which they are attached form a 3-, 4-, 5-, 6-, or 7-membered heterocycloalkyl group or a heteroaryl group, each optionally substituted with 1 , 2, 3 or 4 substituents independently selected from halo, cyano, nitro, hydroxyl, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, C3_7 cycloalkyl, Ci_6 haloalkyl, Ci_6 alkoxy, Ci_6 haloalkoxy, amino, Ci_6 alkylamino, di-Ci_6 alkylamino, thio, Ci_6 alkylthio, Ci_6 alkylsulfinyl, Ci_6 alkylsulfonyl, carbamyl, Ci_6 alkylcarbamyl, di-Ci_6 alkylcarbamyl, carboxy, Ci_6 alkylcarbonyl, Ci_6 alkoxycarbonyl, and Ci_6 alkylcarbonylamino;
each Rel is, independently, H, Ci_6 alkyl, CN, hydroxyl, Ci_6 alkoxy, Ci_6 alkylsulfonyl, carboxy, Ci_6 alkylcarbonyl, amino sulfonyl, Ci_6 alkylaminosulfonyl, di-Ci_ 6 alkylaminosulfonyl, carbamyl, Ci_6 alkylcarbamyl, or di-Ci_6 alkylcarbamyl; each Rg, Rg , and Rh is, independently, halo, cyano, nitro, hydroxyl, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, C3_7 cycloalkyl, Ci_6 haloalkyl, Ci_6 alkoxy, Ci_6 haloalkoxy, amino, Ci_6 alkylamino, di-Ci_6 alkylamino, thio, Ci_6 alkylthio, C1-6 alkylsulfinyl, Ci_6 alkylsulfonyl, carbamyl, C1-6 alkylcarbamyl, di-Ci_6 alkylcarbamyl, carboxy, Ci_6 alkylcarbonyl, Ci_6 alkoxycarbonyl, or Ci_6 alkylcarbonylamino; and
n is 0, 1, 2, 3, or 4.
In some embodiments, X is N.
In some embodiments, X is CR2.
In some embodiments, X is C(H), C(F), or C(CN).
In some embodiments, X is CH.
In some embodiments, Y is N.
In some embodiments, Y is CR3.
In some embodiments, Y is CH.
In some embodiments, Z is cyano.
In some embodiments, L is C(=0)NH, C(=0), S(=0)2, CH2, C(=0)CH2, or
Figure imgf000012_0001
In some embodiments, L is C(=0)NH, C(=0), S(=0)2, CH2, C(=0)CH2,
Figure imgf000012_0002
In some embodiments, L is C(=0).
In some embodiments, L is C(=0)0.
In some embodiments, L is C(=0)OCH2.
In some embodiments, L is C(=0)NH.
In some embodiments, L is S(=0)2.
In some embodiments, L is C(R4)2.
In some embodiments, L is CH2.
In some embodiments, L is C(=0)CH2 or
Figure imgf000012_0003
In some embodiments, L is C(=0)CH2. In some embodiments, L is
Figure imgf000013_0001
In some embodiments, n is 0, 1, or 2.
In some embodiments, n is 0.
In some embodiments, n is 1.
In some embodiments, n is 2.
In some embodiments, R1 is Ci_4 alkyl.
In some embodiments, R1 is methyl.
In some embodiments, two R1 groups form a 2-carbon bridge.
In some embodiments, A is C6-14 aryl, which is optionally substituted with 1, 2, 3, 4, 5, or 6 independently selected R5 groups.
In some embodiments, A is phenyl, which is optionally substituted with 1, 2, 3, 4, or 5 independently selected R5 groups.
In some embodiments, A is monocyclic C3_g cycloalkyl or bicyclic C3_g cycloalkyl, each of which is optionally substituted with 1, 2, 3, 4, 5, or 6 independently selected R5 groups.
In some embodiments, A is monocyclic C2-10 heterocycloalkyl or bicyclic C2-10 heterocycloalkyl, each of which is optionally substituted with 1, 2, 3, 4, 5, or 6 independently selected R5 groups.
In some embodiments, A is monocyclic C1-10 heteroaryl or bicyclic C1-10 heteroaryl, each of which is optionally substituted with 1, 2, 3, 4, 5, or 6 independently selected R5 groups.
In some embodiments, A is Ci_6 alkyl, which is optionally substituted with 1, 2, 3, 4, 5, or 6 independently selected R5 groups.
In some embodiments, A is Ci_6 alkyl.
In some embodiments, A is Ci_6 alkyl, phenyl, a naphthyl ring, monocyclic or bicyclic C3_i0 cycloalkyl, monocyclic or bicyclic C2-10 heterocycloalkyl, or monocyclic or bicyclic C1-10 heteroaryl; each of which is optionally substituted with 1, 2, 3, 4, or 5 independently selected R5 groups.
In some embodiments, A is phenyl, a naphthyl ring, monocyclic C3_io cycloalkyl, bicyclic C3_i0 cycloalkyl, monocyclic C2-10 heterocycloalkyl, bicyclic C2-10 heterocycloalkyl, monocyclic C1-10 heteroaryl, or bicyclic C1-10 heteroaryl; each of which is optionally substituted with 1, 2, 3, 4, or 5 independently selected R5 groups.
In some embodiments, A is methyl, ethyl, isopropyl, phenyl, a naphthalene ring, a pyridine ring, a pyrimidine ring, a thiophene ring, a pyrazine ring, an oxazole ring, an isoxazole ring, an imidazole ring, a thiazole ring, a furan ring, a pyrazole ring, a quinoline ring, a benzothiophene ring, a benzothiazole ring, a benzoimidazole ring, a benzofuran ring, cyclopropyl, cyclopentyl, cyclohexyl, cycloheptyl, an indene ring, a tetrahydronaphthalene ring, dihydro-l,4-benzodioxoxine ring, or a piperidine ring; each of which is optionally substituted with 1, 2, 3, 4, or 5 independently selected R5 groups, as permitted by valency.
In some embodiments, A is phenyl, a naphthalene ring, a pyridine ring, a pyrimidine ring, a thiophene ring, a pyrazine ring, an oxazole ring, an isoxazole ring, an imidazole ring, a thiazole ring, a furan ring, a pyrazole ring, a quinoline ring, a benzothiophene ring, a benzothiazole ring, a benzoimidazole ring, a benzofuran ring, cyclopropyl, cyclopentyl, cyclohexyl, cycloheptyl, an indene ring, a
tetrahydronaphthalene ring, dihydro-l,4-benzodioxoxine ring, or a piperidine ring; each of which is optionally substituted with 1, 2, 3, 4, or 5 independently selected R5 groups, as permitted by valency.
In some embodiments, A is phenyl or a pyridine ring; each of which are optionally substituted with 1, 2, 3, or 4 independently selected R5 groups.
In some embodiments, A is pyridin-4-yl; which is optionally substituted with 1, 2, 3, or 4 independently selected R5 groups.
In some embodiments, A is a pyridine ring; which is optionally substituted with 1 , 2, 3, or 4 independently selected R5 groups.
In some embodiments, each R5 is, independently, halo, cyano, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, Ci_6 haloalkyl, C3-10 cycloalkyl, C3-10 cycloalkyl-Ci_3 alkyl, C2_io heterocycloalkyl, C2_i0 heterocycloalkyl-Ci_3 alkyl, C6_io aryl, C6_io aryl-Ci_3 alkyl, C1-10 heteroaryl, Ci_i0 heteroaryl-Ci_3 alkyl, ORa, SRa, C(0)Rb, C(0)NRcRd, C(0)ORa, OC(0)Rb, OC(0)NRcRd, NRcRd, NRcC(0)Rb, NRcC(0)ORa, NRcC(0)NRcRd,
NRcS(0)Rb, NRcS(0)2Rb, NRcS(0)2NRcRd, S(0)Rb, S(0)NRcRd, S(0)2Rb, or
S(0)2NRcRd; wherein Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, Ci_6 haloalkyl, halosulfanyl, C3-10 cycloalkyl, C3-10 cycloalkyl-Ci_3 alkyl, C2-10 heterocycloalkyl, C2-10 heterocycloalkyl-Ci_3 alkyl, C6_io aryl, C6_io aryl-Ci_3 alkyl, Ci_i0 heteroaryl, and Ci_i0 heteroaryl-Ci_3 alkyl are each optionally substituted with 1, 2, 3, 4, or 5 independently selected R6 groups.
In some embodiments, each R5 is, independently, halo, cyano, Ci_6 alkyl, Ci_6 haloalkyl, C3_i0 cycloalkyl, C2_io heterocycloalkyl, C6_io aryl, C6_io aryl-Ci_3 alkyl, Ci_i0 heteroaryl, ORa, SRa, C(0)Rb, C(0)NRcRd, C(0)ORa, OC(0)Rb, OC(0)NRcRd, NRcRd, NRcC(0)Rb, NRcC(0)ORa, NRcC(0)NRcRd, NRcS(0)2Rb, NRcS(0)2NRcRd, S(0)2Rb, or S(0)2NRcRd; wherein Ci_6 alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci_6 haloalkyl, halosulfanyl, C3_io cycloalkyl, C3_i0 cycloalkyl-Ci_3 alkyl, C2_io heterocycloalkyl, C2_io
heterocycloalkyl-Ci_3 alkyl, C6-io aryl, C6-io aryl-Ci_3 alkyl, Ci_io heteroaryl, and Ci_io heteroaryl-Ci_3 alkyl are each optionally substituted with 1, 2, 3, 4, or 5 independently selected R6 groups.
In some embodiments, each R5 is, independently, halo, cyano, Ci_6 alkyl, Ci_6 haloalkyl, C6-i0 aryl, CM0 heteroaryl, ORa, SRa, C(0)ORa, NRcRd, or NRcC(0)Rb;
wherein said Ci_6 alkyl, Ci_6 haloalkyl, C6-io aryl, and Ci_io heteroaryl are each optionally substituted with 1, 2, 3, 4, or 5 independently selected R6 groups.
In some embodiments, each R5 is, independently, halo, cyano, Ci_6 alkyl, Ci_6 haloalkyl, C6_i0 aryl, CM0 heteroaryl, ORa, SRa, C(0)ORa, NRcRd, or NRcC(0)Rb;
wherein said Ci_6 alkyl, Ci_6 haloalkyl, C6-io aryl, and Ci_io heteroaryl are each optionally substituted with 1, 2, 3, 4, or 5 independently selected R6 groups; and
each Ra, Rb, Rc, and Rd is, independently, H, Ci_6 alkyl, Ci_6 haloalkyl, or C6-io aryl; wherein said Ci_6 alkyl and C6_io aryl is optionally substituted with 1, 2, 3, 4, or 5 independently selected Rg groups.
In some embodiments, each R5 is, independently, halo, cyano, Ci_6 alkyl, Ci_6 haloalkyl, hydroxyl, Ci_6 alkoxy, Ci_6 haloalkoxy, C6-i2 aryloxy, amino, Ci_6 alkylamino, di-Ci_6 alkylamino, Ci_6 alkylcarbonylamino, Ci_6 alkoxycarbonyl, Ci_6 alkylthio, C6-i2 aryl, or Ci_9 heteroaryl; wherein said C6-i2 aryl or Ci_9 heteroaryl are each optionally substituted with 1, 2, 3, 4, or 5 independently selected R6 groups.
In some embodiments, each R5 is, independently, chloro, fluoro, bromo, cyano, methyl, ethyl, trifluoromethyl, hydroxyl, methoxy, trifluoromethoxy, difluoromethoxy, phenoxy, dimethylamino, t-butylcarbonylamino, methoxycarbonyl, methylthio, phenyl, a pyridine ring, a thiazole ring, a quinoline ring, an isoquinoline ring, an imidazo[l ,2- ajpyrimidine ring, a benzoxazole ring, or an oxadiazole ring; wherein said phenyl, pyridine ring, thiazole ring, quinoline ring, isoquinoline ring, imidazo[l ,2-a]pyrimidine ring, benzoxazole ring, and oxadiazole ring are each optionally substituted with 1 , 2, 3, 4, or 5 independently selected R6 groups.
In some embodiments, each R5 is, independently, halo or Ci_6 haloalkyl.
In some embodiments, each R5 is, independently, fluoro or trifluoromethyl.
In some embodiments, each R6 is, independently, halo, cyano, nitro, hydroxyl, Ci_ 6 alkyl, C2_6 alkenyl, C2_6 alkynyl, C3_7 cycloalkyl, Ci_6 haloalkyl, Ci_6 alkoxy, Ci_6 haloalkoxy, amino, Ci_6 alkylamino, di-Ci_6 alkylamino, thio, Ci_6 alkylthio, Ci_6 alkylsulfinyl, Ci_6 alkylsulfonyl, carbamyl, Ci_6 alkylcarbamyl, di-Ci_6 alkylcarbamyl, carboxy, Ci_6 alkylcarbonyl, Ci_6 alkoxycarbonyl, or Ci_6 alkylcarbonylamino.
In some embodiments, each R6 is, independently, halo, cyano, Ci_6 alkyl, Ci_6 haloalkyl, ORal, NRclRdl, or OC(0)Rbl; and
each Ral, Rbl, Rcl, and Rdl is, independently, H or Ci_6 alkyl; wherein said Ci_6 alkyl is optionally substituted with a substituent independently selected from Ci_4 alkoxy and hydroxyl;
or any Rcl and Rdl together with the N atom to which they are attached form a 4-, 5-, 6-, or 7-membered heterocycloalkyl group, optionally substituted with 1 , 2, 3 or 4 substituents independently selected from halo.
In some embodiments, each R5 is, independently, halo, cyano, Ci_6 alkyl, Ci_6 haloalkyl, C6_i0 aryl, Ci_i0 heteroaryl, ORa, SRa, C(0)ORa, NRcRd, or NRcC(0)Rb;
wherein said Ci_6 alkyl, Ci_6 haloalkyl, C6-io aryl, and Ci_io heteroaryl are each optionally substituted with 1 , 2, 3, 4, or 5 independently selected R6 groups;
each Ra, Rb, Rc, and Rd is, independently, H, Ci_6 alkyl, Ci_6 haloalkyl, or C6-io aryl; wherein said Ci_6 alkyl and C6_io aryl is optionally substituted with 1 , 2, 3, 4, or 5 independently selected Rg groups;
each R6 is, independently, halo, cyano, Ci_6 alkyl, Ci_6 haloalkyl, ORal, NRclRdl, or OC(0)Rbl; and each Ral, Rbl, Rcl, and Rdl is, independently, H or Ci_6 alkyl; wherein said Ci_6 alkyl is optionally substituted with a substituent independently selected from Ci_4 alkoxy and hydroxyl;
or any Rcl and Rdl together with the N atom to which they are attached form a 4-, 5-, 6-, or 7-membered heterocycloalkyl group, optionally substituted with 1 , 2, 3 or 4 substituents independently selected from halo.
In some embodiments, each R6 is, independently, halo, cyano, or Ci_6 alkyl.
In some embodiments, each R6 is, independently, chloro, fluoro, cyano, or methyl.
In some embodiments, wherein R2 is H, halo, or cyano.
In some embodiments, R2 is H, F, or cyano.
In some embodiments, R3 is H.
In some embodiments, the compound is a compound of Formula (II):
Figure imgf000017_0001
II
or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound is a compound of Formula (Ha):
Figure imgf000018_0001
Ila
or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound is a compound of Formula (III):
Figure imgf000018_0002
III
or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound is a compound of Formula
Figure imgf000019_0001
Ilia
or a pharmaceutically acceptable salt thereof.
In some embodiments:
X is N or CR2;
Y is N or CR3;
each R1 is, independently, Ci_4 alkyl; or
two R1 groups form a 2-carbon bridge.
R2 is H, halo, or cyano;
R3 is H;
Z is cyano;
L is C(=0)NH, C(=0), S(=0)2, CH2, C(=0)CH2, or
Figure imgf000019_0002
A is Ci_6 alkyl, phenyl, a naphthyl ring, monocyclic C3-10 cycloalkyl, bicyclic C3-10 cycloalkyl, monocyclic C2_io heterocycloalkyl, bicyclic C2_io heterocycloalkyl, monocyclic Ci_io heteroaryl, or bicyclic Ci_io heteroaryl; each of which is optionally substituted with 1, 2, 3, 4, or 5 independently selected R5 groups;
each R5 is, independently, halo, cyano, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, Ci_6 haloalkyl, C3-10 cycloalkyl, C3-10 cycloalkyl-Ci_3 alkyl, C2_io heterocycloalkyl, C2_io heterocycloalkyl-Ci_3 alkyl, C6-io aryl, C6-io aryl-Ci_3 alkyl, Ci_io heteroaryl, Ci_io heteroaryl-Ci_3 alkyl, ORa, SRa, C(0)Rb, C(0)NRcRd, C(0)ORa, OC(0)Rb, OC(0)NRcRd, NRcRd, NRcC(0)Rb, NRcC(0)ORa, NRcC(0)NRcRd, NRcS(0)Rb, NRcS(0)2Rb, NRcS(0)2NRcRd, S(0)Rb, S(0)NRcRd, S(0)2Rb, or S(0)2NRcRd; wherein Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, Ci_6 haloalkyl, halosulfanyl, C3_i0 cycloalkyl, C3_i0 cycloalkyl-Ci_3 alkyl, C2_io heterocycloalkyl, C2_io heterocycloalkyl-Ci_3 alkyl, C6-io aryl, C6-io aryl-Ci_3 alkyl, Ci_io heteroaryl, and Ci_io heteroaryl-Ci_3 alkyl are each optionally substituted with 1, 2, 3, 4, or 5 independently selected R6 groups; and
n is 0, 1, or 2.
In some embodiments:
X is N or CR2;
Y is N or CR3;
each R1 is, independently, Ci_4 alkyl; or
two R1 groups form a 2-carbon bridge.
R2 is H, halo, or cyano;
R3 is H;
Z is cyano;
L is C(=0)NH, C(=0), S(=0)2, CH2, C(=0)CH2, or
Figure imgf000020_0001
A is Ci_6 alkyl, phenyl, a naphthyl ring, monocyclic C3_io cycloalkyl, bicyclic C3_io cycloalkyl, monocyclic C2_i0 heterocycloalkyl, bicyclic C2_i0 heterocycloalkyl, monocyclic Ci_io heteroaryl, or bicyclic Ci_io heteroaryl; each of which is optionally substituted with 1, 2, 3, 4, or 5 independently selected R5 groups;
each R5 is, independently, halo, cyano, Ci_6 alkyl, Ci_6 haloalkyl, C3_io cycloalkyl, C2_io heterocycloalkyl, C6-io aryl, C6-io aryl-Ci_3 alkyl, Ci_io heteroaryl, ORa, SRa, C(0)Rb, C(0)NRcRd, C(0)ORa, OC(0)Rb, OC(0)NRcRd, NRcRd, NRcC(0)Rb, NRcC(0)ORa, NRcC(0)NRcRd, NRcS(0)2Rb, NRcS(0)2NRcRd, S(0)2Rb, or S(0)2NRcRd; wherein Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, Ci_6 haloalkyl, halosulfanyl, C3_io cycloalkyl, C3_io cycloalkyl-Ci_3 alkyl, C2_io heterocycloalkyl, C2_io heterocycloalkyl-Ci_3 alkyl, C6-io aryl, C6-io aryl-Ci_3 alkyl, Ci_io heteroaryl, and Ci_io heteroaryl-Ci_3 alkyl are each optionally substituted with 1, 2, 3, 4, or 5 independently selected R6 groups; and
n is 0, 1, or 2.
In some embodiments:
X is N or CR2; Y is N or CR3;
each R1 is, independently, Ci_4 alkyl; or
two R1 groups form a 2-carbon bridge.
R2 is H, halo, or cyano;
R3 is H;
Z is cyano;
L is C(=0)NH, C(=0), S(=0)2, CH2, C(=0)CH2, or
Figure imgf000021_0001
A is phenyl, a naphthyl ring, monocyclic C3-10 cycloalkyl, bicyclic C3-10 cycloalkyl, monocyclic C2_i0 heterocycloalkyl, bicyclic C2_i0 heterocycloalkyl, monocyclic Ci_io heteroaryl, or bicyclic Ci_io heteroaryl; each of which is optionally substituted with 1, 2, 3, 4, or 5 independently selected R5 groups;
each R5 is, independently, halo, cyano, Ci_6 alkyl, Ci_6 haloalkyl, C3-10 cycloalkyl, C2_io heterocycloalkyl, C6_io aryl, C6_io aryl-Ci_3 alkyl, Ci_i0 heteroaryl, ORa, SRa, C(0)Rb, C(0)NRcRd, C(0)ORa, OC(0)Rb, OC(0)NRcRd, NRcRd, NRcC(0)Rb, NRcC(0)ORa, NRcC(0)NRcRd, NRcS(0)2Rb, NRcS(0)2NRcRd, S(0)2Rb, or S(0)2NRcRd; wherein Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, Ci_6 haloalkyl, halosulfanyl, C3-10 cycloalkyl, C3_io cycloalkyl-Ci_3 alkyl, C2_i0 heterocycloalkyl, C2_i0 heterocycloalkyl-Ci_3 alkyl, C6_io aryl, C6-io aryl-Ci_3 alkyl, Ci_io heteroaryl, and Ci_io heteroaryl-Ci_3 alkyl are each optionally substituted with 1, 2, 3, 4, or 5 independently selected R6 groups; and
n is 0, 1, or 2.
In some embodiments:
X is N or CR2;
Y is N or CR3;
each R1 is, independently, Ci_4 alkyl; or
two R1 groups form a 2-carbon bridge.
R2 is H, halo, or cyano;
R3 is H;
Z is cyano;
L is C(=0)NH, C(=0), S(=0)2, CH2, C(=0)CH2, or
Figure imgf000021_0002
A is Ci_6 alkyl, phenyl, a naphthyl ring, monocyclic C3-10 cycloalkyl, bicyclic C3-10 cycloalkyl, monocyclic C2-10 heterocycloalkyl, bicyclic C2-10 heterocycloalkyl, monocyclic C1-10 heteroaryl, or bicyclic C1-10 heteroaryl; each of which is optionally substituted with 1, 2, 3, 4, or 5 independently selected R5 groups;
each R5 is, independently, halo, cyano, Ci_6 alkyl, Ci_6 haloalkyl, C6-io aryl, C1-10 heteroaryl, ORa, SRa, C(0)ORa, NRcRd, NRcC(0)Rb; wherein said Ci_6 alkyl, Ci_6 haloalkyl, C6-io aryl, and C1-10 heteroaryl are each optionally substituted with 1, 2, 3, 4, or 5 independently selected R6 groups;
and
n is 0, 1, or 2.
In some embodiments:
X is N or CR2;
Y is N or CR3;
each R1 is, independently, Ci_4 alkyl; or
two R1 groups form a 2-carbon bridge.
R2 is H, halo, or cyano;
R3 is H;
Z is cyano;
L is C(=0)NH, C(=0), S(=0)2, CH2, C(=0)CH2, or
Figure imgf000022_0001
A is phenyl, a naphthyl ring, monocyclic C3-10 cycloalkyl, bicyclic C3-10 cycloalkyl, monocyclic C2_io heterocycloalkyl, bicyclic C2_io heterocycloalkyl, monocyclic C1-10 heteroaryl, or bicyclic C1-10 heteroaryl; each of which is optionally substituted with 1, 2, 3, 4, or 5 independently selected R5 groups;
each R5 is, independently, halo, cyano, Ci_6 alkyl, Ci_6 haloalkyl, C6-io aryl, C1-10 heteroaryl, ORa, SRa, C(0)ORa, NRcRd, NRcC(0)Rb; wherein said Ci_6 alkyl, Ci_6 haloalkyl, C6_io aryl, and C1-10 heteroaryl are each optionally substituted with 1, 2, 3, 4, or
5 independently selected R6 groups;
and
n is 0, 1, or 2.
In some embodiments: X is N or CR2;
Y is N or CR3;
each R1 is, independently, methyl; or
two R1 groups form a 2-carbon bridge.
R2 is H, halo, or cyano;
R3 is H;
Z is cyano;
L is C(=0)NH, C(=0), S(=0)2, CH2, C(=0)CH2, or
Figure imgf000023_0001
A is methyl, ethyl, isopropyl, phenyl, a naphthalene ring, a pyridine ring, a pyrimidine ring, a thiophene ring, a pyrazine ring, an oxazole ring, an isoxazole ring, an imidazole ring, a thiazole ring, a furan ring, a pyrazole ring, a quinoline ring, a benzothiophene ring, a benzothiazole ring, a benzoimidazole ring, a benzofuran ring, cyclopropyl, cyclopentyl, cyclohexyl, cycloheptyl, an indene ring, a
tetrahydronaphthalene ring, dihydro-l,4-benzodioxoxine ring, or a piperidine ring; each of which are optionally substituted with 1, 2, 3, 4, 5, or 6 independently selected R5 groups, as permitted by valency;
each R5 is, independently, chloro, fluoro, bromo, cyano, methyl, ethyl, trifluoromethyl, hydroxyl, methoxy, trifluoromethoxy, difluoromethoxy, phenoxy, dimethylamino, t-butylcarbonylamino, methoxycarbonyl, methylthio, phenyl, a pyridine ring, a thiazole ring, a quinoline ring, an isoquinoline ring, an imidazo[l,2-a]pyrimidine ring, a benzoxazole ring, or an oxadiazole ring; wherein said phenyl, pyridine ring, thiazole ring, quinoline ring, isoquinoline ring, imidazo[l,2-a]pyrimidine ring, benzoxazole ring, and oxadiazole ring are each optionally substituted with 1, 2, 3, 4, or 5 independently selected R6 groups; and
n is 0, 1, or 2.
In some embodiments:
X is N or CR2;
Y is N or CR3;
each R1 is, independently, methyl; or
two R1 groups form a 2-carbon bridge. R2 is H, halo, or cyano;
R3 is H;
Z is cyano;
L is C(=0)NH, C(=0), S(=0)2, CH2, C(=0)CH2, or
Figure imgf000024_0001
A is phenyl, a naphthalene ring, a pyridine ring, a pyrimidine ring, a thiophene ring, a pyrazine ring, an oxazole ring, an isoxazole ring, an imidazole ring, a thiazole ring, a furan ring, a pyrazole ring, a quinoline ring, a benzothiophene ring, a
benzothiazole ring, a benzoimidazole ring, a benzofuran ring, cyclopropyl, cyclopentyl, cyclohexyl, cycloheptyl, an indene ring, a tetrahydronaphthalene ring, dihydro-1,4- benzodioxoxine ring, or a piperidine ring; each of which are optionally substituted with 1, 2, 3, 4, 5, or 6 independently selected R5 groups;
each R5 is, independently, chloro, fluoro, bromo, cyano, methyl, ethyl,
trifluoromethyl, hydroxyl, methoxy, trifluoromethoxy, difluoromethoxy, phenoxy, dimethylamino, t-butylcarbonylamino, methoxycarbonyl, methylthio, phenyl, a pyridine ring, a thiazole ring, a quinoline ring, an isoquinoline ring, an imidazo[l,2-a]pyrimidine ring, a benzoxazole ring, or an oxadiazole ring; wherein said phenyl, pyridine ring, thiazole ring, quinoline ring, isoquinoline ring, imidazo[l,2-a]pyrimidine ring, benzoxazole ring, and oxadiazole ring are each optionally substituted with 1, 2, 3, 4, or 5 independently selected R6 groups; and
n is 0, 1, or 2.
In some embodiments:
X is N or CR2;
Y is N or CR3;
each R1 is, independently, Ci_4 alkyl; or
two R1 groups form a 2-carbon bridge.
R2 is H, halo, or cyano;
R3 is H;
Z is cyano;
L is S(=0)2;
A is Ci_6 alkyl; and n is 0, 1, or 2.
In some embodiments:
X is N or CH;
Y is N;
Z is cyano;
L is C(=0) or C(=0)NH;
A is phenyl or a pyridine ring, each of which is optionally substituted with 1, 2, 3, or 4 independently selected R5 groups;
each R5 is, independently, halo or Ci_6 haloalkyl; and
n is O.
In some embodiments:
X is N or CH;
Y is N;
Z is cyano;
L is C(=0) or C(=0)NH;
A is phenyl or pyridin-4-yl, each of which is optionally substituted with 1 or 2 independently selected R5 groups;
each R5 is, independently, fluoro or trifluoromethyl; and
n is 0.
In some embodiments:
X is N or CR2;
Y is N or CR3;
each R1 is, independently, Ci_4 alkyl, hydroxyl, Ci_4 alkoxy, or fluoro;
two R1 groups together form a 2- or 3 -carbon bridge or a bridge of formula -CH2-
R2 is H, halo, or cyano;
R3 is H;
Z is cyano;
L is C(=0)NH, C(=0), S(=0)2, CH2, C(=0)CH2,
Figure imgf000025_0001
C(=0)0, or
C(=0)OCH2; A is Ci_6 alkyl, phenyl, a naphthyl ring, monocyclic C3-10 cycloalkyl, bicyclic C3-10 cycloalkyl, monocyclic C2-10 heterocycloalkyl, bicyclic C2-10 heterocycloalkyl, monocyclic C1-10 heteroaryl, or bicyclic C1-10 heteroaryl; each of which is optionally substituted with 1, 2, 3, 4, or 5 independently selected R5 groups;
each R5 is, independently, halo, cyano, Ci_6 alkyl, Ci_6 haloalkyl, C6-io aryl, C1-10 heteroaryl, ORa, SRa, C(0)ORa, NRcRd, NRcC(0)Rb; wherein said Ci_6 alkyl, Ci_6 haloalkyl, C6-io aryl, and C1-10 heteroaryl are each optionally substituted with 1, 2, 3, 4, or 5 independently selected R6 groups;
n is 0, 1, or 2;
each R5 is, independently, halo, cyano, Ci_6 alkyl, Ci_6 haloalkyl, C6_io aryl, C1-10 heteroaryl, ORa, SRa, C(0)ORa, NRcRd, or NRcC(0)Rb; wherein said Ci_6 alkyl, Ci_6 haloalkyl, C6-io aryl, and C1-10 heteroaryl are each optionally substituted with 1, 2, 3, 4, or 5 independently selected R6 groups;
each Ra, Rb, Rc, and Rd is, independently, H, Ci_6 alkyl, Ci_6 haloalkyl, or C6_io aryl; wherein said Ci_6 alkyl and C6-io aryl is optionally substituted with 1, 2, 3, 4, or 5 independently selected Rg groups;
each R6 is, independently, halo, cyano, Ci_6 alkyl, Ci_6 haloalkyl, ORal, NRclRdl, or OC(0)Rbl; and
each Ral, Rbl, Rcl, and Rdl is, independently, H or Ci_6 alkyl; wherein said Ci_6 alkyl is optionally substituted with a substituent independently selected from Ci_4 alkoxy and hydroxyl;
or any Rcl and Rdl together with the N atom to which they are attached form a 4-, 5-, 6-, or 7-membered heterocycloalkyl group, optionally substituted with 1, 2, 3 or 4 substituents independently selected from halo.
In some embodiments, the compound is selected from:
{ 1 - { 1 -[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl} -3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{l-[l-(3-fluoro-4-quinolin-6-ylbenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-[l-(3,5-difluorobenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)- 1 H-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile; {3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 -yl]- 1 -[1 -(3,4,5- trifluorobenzoyl)piperidin-4-yl]azetidin-3-yl} acetonitrile;
{l-[l-(3-fluoro-4-methoxybenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-[l-(3-fluoro-4-hydroxybenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{ 1 - { 1 -[2-fluoro-3-(trifluoromethyl)benzoyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{ 1 - [ 1 -(cyclohexylcarbonyl)piperidin-4-yl] -3 - [4-(7H-pyrrolo [2,3 -d]pyrimidin-4- yl)- 1 H-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{l-(l-benzoylpiperidin-4-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
2- [(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin- 1 -yl}piperidin-l -yl)carbonyl]benzonitrile;
3- [(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin- 1 -yl}piperidin-l -yl)carbonyl]benzonitrile;
4- [(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin- 1 -yl}piperidin-l -yl)carbonyl]benzonitrile;
{ 1 - { 1 -[(6-chloropyridin-2-yl)carbonyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-[l-(pyrazin-2-lcarbonyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)- 1 H-pyrazol- 1 -yl]azetidin-3-l} acetonitrile;
{3 - [4-(7H-pyrrolo [2,3 -d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl]- 1 - [ 1 -(3 - thienylcarbonyl)piperidin-4-yl]azetidin-3-yl} acetonitrile;
{l-[l-(l,3-oxazol-2-ylcarbonyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin- 4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{ 1 -( 1 - { [2-methyl-5 -(trifluoromethyl)- 1 ,3 -oxazol-4-yl] carbonyl} piperidin-4-y l)-3 - [4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
3-[(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin- 1 -yl} piperidin- 1 -yl)carbonyl] -5 -fluorobenzonitrile; {l-[l-(3-chlorobenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-[l-(3-bromobenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
(3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol- 1 -yl] - 1 - { 1 -[3- (trifluoromethoxy)benzoyl]piperidin-4-yl}azetidin-3-yl)acetonitrile;
(3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol- 1 -yl] - 1 - { 1 -[3- (trifluoromethyl)benzoyl]piperidin-4-yl}azetidin-3-yl)acetonitrile;
{ 1 - { 1 -[3-fluoro-5-(trifluoromethyl)benzoyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{ 1 - [ 1 -(3 ,5 -dichlorobenzoyl)piperidin-4-yl] -3 - [4-(7H-pyrrolo [2,3 -d]pyrimidin-4- yl)- 1 H-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{ 1 - [ 1 -(3 -fluorobenzoyl)piperidin-4-yl] -3 - [4-(7H-pyrrolo [2,3 -d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-[l-(4-fluoro-3-methoxybenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-[l-(2-fluoro-5-methoxybenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{ 1 - [ 1 -(3 -chloro-5 -fluorobenzoyl)piperidin-4-yl] -3 - [4-(7H-pyrrolo [2,3 - d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{ 1 - [ 1 -(3 -bromo-5 -fluorobenzoyl)piperidin-4-yl] -3 - [4-(7H-pyrrolo [2,3 - d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{ 1 - { 1 -[(2,5-dichloro-3-thienyl)carbonyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{ 1 - [ 1 -(3 -methoxybenzoyl)piperidin-4-yl] -3 - [4-(7H-pyrrolo [2,3 -d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]-l-[l-(2,4,5-trifluoro-3- methoxybenzoyl)piperidin-4-yl]azetidin-3-yl} acetonitrile;
{l-[l-(3,5-dimethoxybenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin- 4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile; { 1 - [ 1 -(3 -chloro-4-fluorobenzoyl)piperidin-4-yl] -3 - [4-(7H-pyrrolo [2,3 - d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-[l-(3,4-difluorobenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)- 1 H-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{l-[l-(3-fluoro-5-methoxybenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-[l-(2-chloro-6-methoxyisonicotinoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-[l-(5-fluoro-2-methoxybenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-[l-(2-fluoro-6-methoxybenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-[l-(4-fluoro-2-methoxybenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-[l-(2,3-difluorobenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)- 1 H-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{l-[l-(2,4-difluorobenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)- 1 H-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{l-[l-(2,5-difluorobenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)- 1 H-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{l-[l-(2,6-difluorobenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)- 1 H-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[2-fluoro-6-(trifluoromethyl)benzoyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 -yl]- 1 -[1 -(2,3,4- trifluorobenzoyl)piperidin-4-yl]azetidin-3-yl} acetonitrile;
{3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 -yl]- 1 -[1 -(2,3,6- trifluorobenzoyl)piperidin-4-yl]azetidin-3-yl} acetonitrile;
{3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 -yl]- 1 -[1 -(2,4,5- trifluorobenzoyl)piperidin-4-yl]azetidin-3-yl} acetonitrile; {3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 -yl]- 1 -[1 -(2,4,6- trifluorobenzoyl)piperidin-4-yl]azetidin-3-yl}acetonitrile;
{l-[l-(3,5-dibromo-4-methoxybenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
3-[(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-l-yl}piperidin-l-yl)carbonyl]-6-(dimethylamino)-2-fluorobenzonitrile;
{ 1 - { 1 -[3-fluoro-4-(trifluoromethyl)benzoyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{ 1 -( 1 - { [4-chloro-6-(trifluoromethyl)pyridin-2-yl] carbonyl} piperidin-4-yl)-3 - [4- (7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]-l-[l-(2,3,4,5- tetrafluorobenzoyl)piperidin-4-yl]azetidin-3-yl} acetonitrile;
5-[(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin- 1 -yl} piperidin- 1 -yl)carbonyl] -2-methoxybenzonitrile;
{3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]-l-[l-(2,3,5,6- tetrafluorobenzoyl)piperidin-4-yl]azetidin-3-yl} acetonitrile;
(3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol- 1 -yl]- 1 - { 1 -[2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl}azetidin-3-yl)acetonitrile;
{l-[l-(4-fluoro-3-hydroxybenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
5-[(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin- 1 -yl} piperidin- 1 -yl)carbonyl] -2-(dimethylamino)benzonitrile;
{ 1 - { 1 -[4-(dimethylamino)-2,3 ,5 ,6-tetrafluorobenzoyl]piperidin-4-yl} -3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{l-[l-(3,5-difluoroisonicotinoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{ 1 - { 1 -[3-fluoro-4-(methylthio)benzoyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{ 1 - [ 1 -(4-chloro-3 -fluorobenzoyl)piperidin-4-yl] -3 - [4-(7H-pyrrolo [2,3 - d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile; {l-[l-(3-fluoro-4-methylbenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-[l-(2,5-dimethyl-3-furoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)- 1 H-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
4-[(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin- 1 -yl} piperidin- 1 -yl)carbonyl] -2-fluorobenzonitrile;
{l-[l-(2-fluorobenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-[l-(4-fluorobenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-[l-(2-thienylcarbonyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{ 1 - { 1 -[3-methoxy-5-(trifluoromethyl)-2-thienylcarbonyl]piperidin-4-yl} -3-[4- (7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[3-hydroxy-5-(trifluoromethyl)-2-thienylcarbonyl]piperidin-4-yl} -3-[4- (7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[(4-methoxy-3-thienyl)carbonyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{ 1 - { 1 -[(5-methyl-3-thienyl)carbonyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{ 1 - { 1 -[(5-chloro-4-methoxy-3-thienyl)carbonyl]piperidin-4-yl} -3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[(2-bromo-3-thienyl)carbonyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{ 1 - { 1 -[(3-chloro-2-thienyl)carbonyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{ 1 - { 1 -[(5-chloro-2-thienyl)carbonyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{ 1 - { 1 -[(3-methyl-2-thienyl)carbonyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile; { 1 - { 1 -[(4-methyl-2-thienyl)carbonyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{ 1 - { 1 -[(5-methyl-2-thienyl)carbonyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{ 1 - { 1 -[(3-methoxy-2-thienyl)carbonyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{ 1 - { 1 -[2-fluoro-4-(trifluoromethyl)benzoyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
4-[(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin- 1 -yl} piperidin- 1 -yl)carbonyl] -3 ,5 -difluorobenzonitrile;
{l-[l-(3-chloro-4-hydroxybenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
[3 -[4-(7H-pyrrolo [2,3 -d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] - 1 -( 1 - { [2- (trifluoromethyl)pyrimidin-4-yl]carbonyl}pipe
[3 -[4-(7H-pyrrolo [2,3 -d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] - 1 -( 1 - { [6- (trifluoromethyl)pyrazin-2-yl]carbonyl}piperidin-4-yl)azetidin-3-yl]acetonitrile;
{l-[l-(l-naphthoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH- pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 -yl]- 1 -[1 -(quinolin-3- ylcarbonyl)piperidin-4-yl]azetidin-3-yl} acetonitrile;
{3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 -yl]- 1 -[1 -(quinolin-6- ylcarbonyl)piperidin-4-yl]azetidin-3-yl} acetonitrile;
{ 1 - [ 1 -( 1 -benzothien-2-ylcarbonyl)piperidin-4-yl] -3 - [4-(7H-pyrrolo [2,3 - d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{ 1 - { 1 -[(3-chloro-6-fluoro-l -benzothien-2-yl)carbonyl]piperidin-4-yl} -3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[(3-chloro-4-fluoro-l -benzothien-2-yl)carbonyl]piperidin-4-yl} -3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
[3 -[4-(7H-pyrrolo [2,3 -d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] - 1 -( 1 - { [4- (trifluoromethyl)-l-benzothien-2-yl]carbonyl}piperidin-4-yl)azetidin-3-yl]acetonitrile; [3 -[4-(7H-pyrrolo [2,3 -d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] - 1 -( 1 - { [6- (trifluoromethyl)-l-benzothien-2-yl]carbonyl}piperidin-4-yl)azetidin-3-yl]acetonitrile;
[3 -[4-(7H-pyrrolo [2,3 -d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] -l-(l-{[7- (trifluoromethyl)-l-benzothien-2-yl]carbonyl}piperidin-4-yl)azetidin-3-yl]acetonitri
{ 1 - [ 1 -( 1 -benzothien-3 -ylcarbonyl)piperidin-4-yl] -3 - [4-(7H-pyrrolo [2,3 - d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]-l-[l-(l,2,3,4- tetrahydronaphthalen-2-ylcarbonyl)piperidin-4-yl]azetidin-3-yl} acetonitrile;
[3 -[4-(7H-pyrrolo [2,3 -d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] - 1 -( 1 - { [4- (trifluoromethyl)cyclohexyl]carbonyl}piperidin-4-yl)azetidin-3-yl]acetonitrile;
{l-[l-(2,3-dihydro-lH-inden-2-ylcarbonyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{ 1 - { 1 -[(4,4-difluorocyclohexyl)carbonyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-[l-(cyclopentylcarbonyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)- 1 H-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{l-[l-(cycloheptylcarbonyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)- 1 H-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[(3-methoxycyclohexyl)carbonyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{ 1 - { 1 -[(4-phenylcyclohexyl)carbonyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{ 1 -(1 - {[4-(4-chlorophenyl)cyclohexyl]carbonyl}piperidin-4-yl)-3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
6-{4-[(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin- 1 -yl}piperidin-l -yl)carbonyl]piperidin- 1 -yl}nicotinonitrile;
{ 1 -(1 - {[ 1 -(5-chloro-3-fluoropyridin-2-yl)piperidin-4-yl]carbonyl}piperidin-4-yl)- 3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
2- {4-[(4- {3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 - yl] azetidin- 1 -yl} piperidin- 1 -yl)carbonyl]piperidin- 1 -yl} -6-methylnicotinonitrile; {l-[l-(phenylacetyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH- pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{ 1 - { 1 -[(1 -phenylcyclopropyl)carbonyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{ 1 -(1 - {[ 1 -(4-chlorophenyl)cyclopropyl]carbonyl}piperidin-4-yl)-3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[(2,6-dichlorophenyl)acetyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-[l-(mesitylacetyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH- pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-[l-(biphenyl-4-ylcarbonyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)- 1 H-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{l-[l-(3-fluoro-4-isoquinolin-6-ylbenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-[l-(2,6-difluoro-4-pyridin-3-ylbenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{ 1 - [ 1 -(3 -fluoro-4-pyridin-4-ylbenzoyl)piperidin-4-yl] -3 - [4-(7H-pyrrolo [2,3 - d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
4'-[(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin- 1 -yl} piperidin- 1 -yl)carbonyl] -2'-fluorobipheny 1-4-carbonitrile;
4'-[(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin- 1 -yl} piperidin- 1 -yl)carbonyl] -2',3 -difluorobiphenyl-4-carbonitrile;
{ 1 - [ 1 -(2-fluoro-4-pyridin-3 -ylbenzoyl)piperidin-4-yl] -3 - [4-(7H-pyrrolo [2,3 - d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-{l-[4-fluoro-3-(l,3-thiazol-2-yl)benzoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-{l-[3-fluoro-4-(l,3-thiazol-2-yl)benzoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{ 1 - [ 1 -(3 -fluoro-4-pyridin-3 -ylbenzoyl)piperidin-4-yl] -3 - [4-(7H-pyrrolo [2,3 - d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile; 4'-[(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin- 1 -yl} piperidin- 1 -yl)carbonyl] -2'-fluorobipheny 1-2-carbonitrile;
4'-[(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin- 1 -yl} piperidin- 1 -yl)carbonyl] -2'-fluorobipheny 1-3 -carbonitrile;
4'-[(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin- 1 -yl} piperidin- 1 -yl)carbonyl]biphenyl-4-carbonitrile;
(3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol- 1 -yl]- 1 - { 1 -[(2,3',4'- trifluorobiphenyl-4-yl)carbonyl]piperidin-4-yl}azetidin-3-yl)acetonitrile;
4'-[(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin- 1 -yl} piperidin- 1 -yl)carbonyl] -2',5 -difluorobiphenyl-3 -carbonitrile;
{l-[l-(3-fluoro-4-quinolin-5-ylbenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-[l-(3-fluoro-4-isoquinolin-5-ylbenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-[l-(3-fluoro-4-isoquinolin-8-ylbenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-[l-(3-fluoro-4-quinolin-8-ylbenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-[l-(3-fluoro-4-isoquinolin-7-ylbenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-[l-(3-fluoro-4-quinolin-7-ylbenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-[l-(3-fluoro-4-imidazo[l,2-a]pyridin-6-ylbenzoyl)piperidin-4-yl]-3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{l-{l-[4-(l,3-benzoxazol-2-yl)benzoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-{l-[4-(l,3-benzoxazol-2-yl)-3-fluorobenzoyl]piperidin-4-yl}-3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
3-[(3-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin- 1-yl} -8-azabicyclo[3.2.1 ]oct-8-yl)carbonyl]-5-fluorobenzonitrile; {l-[8-(3,4-difluorobenzoyl)-8-azabicyclo[3.2.1]oct-3-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
4-[(3-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yljazetidin- 1-yl} -8-azabicyclo[3.2.1 ]oct-8-yl)carbonyl]-2-fluorobenzonitrile;
{l-[8-(4-chloro-3-fluorobenzoyl)-8-azabicyclo[3.2.1]oct-3-yl]-3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{ 1 - {8-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]-8-azabicyclo[3.2.1 ]oct-3-yl} -3- [4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
[3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]-l-(8-{[6- (trifluoromethyl)pyridin-3-yl]carbonyl} -8-azabicyclo[3.2.1 ]oct-3-yl)azetidin-3- yl] acetonitrile;
(3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol- 1 -yl]- 1 - {8-[2- (trifluoromethyl)isonicotinoyl]-8-azabicyclo[3.2.1 ]oct-3-yl} azetidin-3 -yl)acetonitrile;
{l-[8-(cyclopentylcarbonyl)-8-azabicyclo[3.2.1]oct-3-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 -yl]- 1 -[8-(tetrahydro-2H- pyran-4-ylcarbonyl)-8-azabicyclo[3.2.1 ]oct-3-yl]azetidin-3-yl} acetonitrile;
{l-[8-(cyclohexylcarbonyl)-8-azabicyclo[3.2.1]oct-3-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-{8-[(4,4-difluorocyclohexyl)carbonyl]-8-azabicyclo[3.2.1]oct-3-yl}-3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{ 1 - [ 1 -(3 -fluorobenzoyl)-2-methylpiperidin-4-yl] -3 - [4-(7H-pyrrolo [2,3 - d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{ 1 - [ 1 -(3 -fluorobenzoyl)-2-methylpiperidin-4-yl] -3 - [4-(7H-pyrrolo [2,3 - d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{ 1 - { 1 -[(4,4-difluorocyclohexyl)carbonyl]-2-methylpiperidin-4-yl} -3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{ 1 - [ 1 -(3 -fluorobenzoyl)-4-methylpiperidin-4-yl] -3 - [4-(7H-pyrrolo [2,3 - d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-[l-(cyclohexylcarbonyl)-4-methylpiperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile; 4- {3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 - yljazetidin- 1 -yl} -N-[4-fluoro-2-(trifluoromethyl)phenyl]piperidine- 1 -carboxamide;
4- {3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 - yl]azetidin-l-yl}-N-[3-(trifluoromethyl)pyridin-4-yl]piperidine-l -carboxamide;
4- {3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 - yljazetidin- 1 -yl} -N-(2,6-difluorophenyl)piperidine- 1 -carboxamide;
4- {3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 - yljazetidin- 1 -yl} -N-[2-(trifluoromethyl)phenyl]piperidine- 1 -carboxamide;
4- {3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 - yljazetidin- 1 -yl} -N-[2-(trifluoromethoxy)phenyl]piperidine- 1 -carboxamide;
N-(4-bromo-3-thienyl)-4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)- 1 H-pyrazol- 1 -yl] azetidin- 1 -yl} piperidine- 1 -carboxamide;
4- {3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 - yljazetidin- 1 -yl} -N-(2,6-dichlorophenyl)piperidine- 1 -carboxamide;
N-(2-chloro-6-methylphenyl)-4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yljazetidin- 1 -yl}piperidine- 1 -carboxamide;
N-(2-chloro-4-fluorophenyl)-4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yljazetidin- 1 -yl}piperidine- 1 -carboxamide;
N-(2-chlorophenyl)-4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yljazetidin- 1 -yl} piperidine- 1 -carboxamide;
4- {3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 - yljazetidin- 1 -yl} -N-[2-(difluoromethoxy)phenyl]piperidine- 1 -carboxamide;
4- {3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 - yl]azetidin-l-yl}-N-[4-(trifluoromethyl)pyridin-3-yl]piperidine-l -carboxamide;
4- {3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 - yljazetidin- 1 -yl} -N-[3-(trifluoromethyl)phenyl]piperidine- 1 -carboxamide;
4- {3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 - yljazetidin- 1 -yl} -N-[4-(trifluoromethyl)phenyl]piperidine- 1 -carboxamide;
N-(5-chloro-2-methylphenyl)-4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin- 1 -yl} piperidine- 1 -carboxamide; 4- {3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 - yljazetidin- 1 -yl} -N-(2-fluorophenyl)piperidine- 1 -carboxamide;
4- {3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 - yl]azetidin-l-yl}-N-[2-fluoro-3-(trifluoromethyl)phenyl]piperidine-l -carboxamide;
4- {3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 - yljazetidin- 1 -yl} -N-(2,4-difluorophenyl)piperidine- 1 -carboxamide;
4- {3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 - yljazetidin- 1 -yl} -N-(2,3 ,4-trifluorophenyl)piperidine- 1 -carboxamide;
4- {3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 - yljazetidin- 1 -yl} -N-(2,3 ,5-trifluorophenyl)piperidine- 1 -carboxamide;
4- {3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 - yljazetidin- 1 -yl} -N-(2,5-difluorophenyl)piperidine- 1 -carboxamide;
4- {3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 - yljazetidin- 1 -yl} -N-(3,5-difluorophenyl)piperidine- 1 -carboxamide;
4- {3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 - yljazetidin- 1 -yl} -N-(3 ,4-difluorophenyl)piperidine- 1 -carboxamide;
N-(3-chloro-2-fluorophenyl)-4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yljazetidin- 1 -yl}piperidine- 1 -carboxamide;
N-(4-chloro-2-fluorophenyl)-4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yljazetidin- 1 -yl}piperidine- 1 -carboxamide;
4- {3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 - yl] azetidin- 1 -yl} -N-3 -thienylpiperidine- 1 -carboxamide;
4- {3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 - yljazetidin- 1 -yl} -N-(2-methoxyphenyl)piperidine- 1 -carboxamide;
4- {3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 - yl] azetidin- 1 -yl} -N-(3 -methoxyphenyl)piperidine- 1 -carboxamide;
4- {3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 - yljazetidin- 1 -yl} -N-[4-(trifluoromethoxy)phenyl]piperidine- 1 -carboxamide;
N-(3-chlorophenyl)-4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yljazetidin- 1 -yl}piperidine- 1 -carboxamide; N-(4-chlorophenyl)-4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 -yljazetidin- 1 -yl}piperidine- 1 -carboxamide;
4- {3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 - yljazetidin- 1 -yl} -N-(2-methylphenyl)piperidine- 1 -carboxamide;
4- {3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 - yljazetidin- 1 -yl} -N-(2,5-dimethoxyphenyl)piperidine-l -carboxamide;
4- {3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 - yl] azetidin- 1 -yl} -N-(4-fluoro-2-methylphenyl)piperidine- 1 -carboxamide;
4- {3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 - yljazetidin- 1 -yl} -N-[6-(trifluoromethyl)pyridin-2-yl]piperidine- 1 -carboxamide;
4- {3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 - yljazetidin- 1 -yl} -N-(2,6-dimethylpyridin-3-yl)piperidine- 1 -carboxamide;
4- {3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 - yljazetidin- 1 -yl} -N- 1 ,3 -thiazol-2 -ylpiperi dine- 1 -carboxamide;
4- {3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 - yljazetidin- 1 -yl} -N-(4-methyl- 1 ,3-thiazol-2-yl)piperidine- 1 -carboxamide;
4- {3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 - yljazetidin- 1 -yl} -N-(4,5-dimethyl- 1 ,3 -thiazol-2-yl)piperidine-l -carboxamide;
N- 1 ,3-benzothiazol-2-yl-4- {3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin- 4-yl)- 1 H-pyrazol- 1 -yljazetidin- 1 -yl}piperidine- 1 -carboxamide;
4- {3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 - yljazetidin- 1 -yl} -N-( 1 -methyl- 1 H-benzimidazol-2-yl)piperidine- 1 -carboxamide;
N- [4-(4-chlorophenyl)- 1 ,3 -thiazol-2-yl] -4- {3 -(cyanomethyl)-3 - [4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 -yljazetidin- 1 -yl}piperidine- 1 -carboxamide;
4- {3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 - yl] azetidin- 1 -yl} -N-( 1 -ethyl- 1 H-pyrazol-5 -yl)piperidine- 1 -carboxamide;
4- {3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 - yljazetidin- 1 -yl} -N-(l ,3-dimethyl- 1 H-pyrazol-5 -yl)piperidine- 1 -carboxamide;
4- {3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 - yljazetidin- 1 -yl} -N-(2-methylpyridin-3-yl)piperidine- 1 -carboxamide; 4- {3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 - yl]azetidin-l -yl} -N-(6-fluoro-2-methylpyridin-3-yl)piperidine- 1 -carboxamide;
4- {3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 - yljazetidin- 1 -yl} -N-(2-fluoro-6-methylpyridin-3-yl)piperidine- 1 -carboxamide;
4- {3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 - yl]azetidin-l-yl}-N-[3-(trifluoromethyl)pyridin-2-yl]piperidine-l -carboxamide;
4- {3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 - yljazetidin- 1 -yl} -N-(3-fluoropyridin-2-yl)piperidine- 1 -carboxamide;
4- {3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 - yljazetidin- 1 -yl} -N-(3,5-difluoropyridin-2-yl)piperidine- 1 -carboxamide;
4- {3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 - yljazetidin- 1 -yl} -N-(2-methoxypyridin-3-yl)piperidine- 1 -carboxamide;
4- {3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 - yljazetidin- 1-yl} -N-[2-methyl-6-(trifluoromethyl)pyridin-3-yl]piperidine- 1 -carboxamide; methyl 2- {[(4- {3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH- pyrazol- 1 -yl] azetidin- 1 -yl} piperidin- 1 -yl)carbonyl] amino } benzoate;
methyl 2- {[(4- {3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH- pyrazol- 1 -yljazetidin- 1 -yl}piperidin- 1 -yl)carbonyl]amino} -5-fluorobenzoate;
methyl 4- {[(4- {3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH- pyrazol- 1 -yljazetidin- 1 -yl}piperidin- 1 -yl)carbonyl]amino} -3-fluorobenzoate;
3-[(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin- 1 -yl} piperidin- 1 -yl)methyl] -6-(dimethylamino)-2-fluorobenzonitrile;
{l-[l-(3,5-dichlorobenzyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)- 1 H-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[2-chloro-5-(trifluoromethyl)benzyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{ 1 - { 1 -[2-fluoro-3-(trifluoromethyl)benzyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{ 1 - { 1 -[2-fluoro-6-(trifluoromethyl)benzyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile; { 1 - { 1 -[(2-chloroquinolin-3-yl)methyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-[l-(3,5-difluorobenzyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{ 1 - { 1 -[2-fluoro-4-(trifluoromethyl)benzyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-[l-(2,4-difluorobenzyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-[l-(2-fluoro-6-methoxybenzyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-[l-(2,3-dichlorobenzyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)- 1 H-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
5- [(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin- 1 -yl} piperidin- 1 -yl)methyl] -2-fluorobenzonitrile;
{l-{l-[4-(l,2,3-oxadiazol-4-yl)benzyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
2- [(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin- 1 -yl} piperidin- 1 -yl)methyl]benzonitrile;
3- [(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin- 1 -yl} piperidin- 1 -yl)methyl]benzonitrile;
6- [(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin- 1 -yl} piperidin- 1 -yl)methyl] -2-methoxynicotinonitrile;
{ 1 - { 1 -[(2,6-dibromopyridin-4-yl)methyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{ 1 - { 1 -[(2-bromopyridin-4-yl)methyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-[l-(2-chloro-6-fluorobenzyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin- 4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-[l-(3-chloro-2,6-difluorobenzyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile; 4-[(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin- 1 -yl} piperidin- 1 -yl)methyl] -2-fluorobenzonitrile;
{ 1 - { 1 -[(5-methyl-3-phenylisoxazol-4-yl)methyl]piperidin-4-yl} -3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{ 1 -( 1 - { [3 -fluoro-2-(trifluoromethyl)pyridin-4-yl]methyl} piperidin-4-yl)-3 -[4- (7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{ 1 - [ 1 -( 1 -benzofuran-2-ylmethyl)piperidin-4-yl]-3 - [4-(7H-pyrrolo [2,3 - d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-[l-(3-phenoxybenzyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{ 1 -[1 -(2,3-dihydro- 1 ,4-benzodioxin-6-ylmethyl)piperidin-4-yl]-3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
N-{4-[(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol- 1 -yl]azetidin- 1 -yl}piperidin- 1 -yl)methyl]pyridin-2-yl} -2,2-dimethylpropanamide;
{ 1 - { 1 -[3-chloro-2-fluoro-6-(trifluoromethyl)benzyl]piperidin-4-yl} -3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[(3,5-dichloropyridin-4-yl)methyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{ 1 - { 1 -[(2-chloro-6-methoxyquinolin-3-yl)methyl]piperidin-4-yl} -3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{l-[l-(2-chloro-3,4-dimethoxybenzyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
3-[(3-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin- 1-yl} -8-azabicyclo[3.2.1 ]oct-8-yl)methyl]-6-(dimethylamino)-2- fluorobenzonitrile;
{l-[8-(2-chloro-3,6-difluorobenzyl)-8-azabicyclo[3.2.1]oct-3-yl]-3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
3-[(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin- 1 -yl} -2-methylpiperidin- 1 -yl)methyl]-6-(dimethylamino)-2- fluorobenzonitrile; 3-[(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yljazetidin- 1 -yl} -2-methylpiperidin- 1 -yl)methyl]-6-(dimethylamino)-2- fluorobenzonitrile;
{l-[l-(2-chloro-6-fluorobenzyl)-2-methylpiperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-[l-(2-chloro-6-fluorobenzyl)-2-methylpiperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{ 1 - { 1 -[(1 -methyl- lH-pyrazol-5-yl)sulfonyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
2- [(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin- 1 -yl}piperidin-l -yl)sulfonyl]benzonitrile;
3- [(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin- 1 -yl}piperidin-l -yl)sulfonyl]benzonitrile;
4- [(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin- 1 -yl}piperidin-l -yl)sulfonyl]benzonitrile;
5- [(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin- 1 -yl} piperidin- 1 -yl)sulfonyl] -2-(dimethylamino)benzonitrile;
{ 1 - { 1 -[(1 -methyl- lH-pyrazol-3-yl)sulfonyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{ 1 - [ 1 -(cyclohexylsulfonyl)piperidin-4-yl] -3 - [4-(7H-pyrrolo [2,3 -d]pyrimidin-4- yl)- 1 H-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{l-[l-(cyclopentylsulfonyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)- 1 H-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{ 1 - [ 1 -(methylsulfonyl)piperidin-4-yl] -3 - [4-(7H-pyrrolo [2,3 -d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{ 1 - [ 1 -(ethylsulfonyl)piperidin-4-yl] -3 - [4-(7H-pyrrolo [2,3 -d]pyrimidin-4-yl)- 1 H- pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-[l-(cyclopropylsulfonyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)- 1 H-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{ 1 - [ 1 -(isopropylsulfonyl)piperidin-4-yl]-3 - [4-(7H-pyrrolo [2 ,3 -d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile; { 1 - { 1 -[( 1 -methyl- 1 H-imidazol-4-yl)sulfonyl]piperidin-4-yl} -3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[( 1 ,2-dimethyl- 1 H-imidazol-4-yl)sulfonyl]piperidin-4-yl} -3 - [4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[(3,5-dimethylisoxazol-4-yl)sulfonyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{ 1 - { 1 -[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl} -3-[3-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrrol-l-yl]azetidin-3-yl} acetonitrile;
3-[(4-{3-(cyanomethyl)-3-[3-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrrol-l- yl] azetidin- 1 -yl}piperidin-l -yl)carbonyl]benzonitrile;
3- [(4-{3-(cyanomethyl)-3-[3-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrrol-l- yl] azetidin- 1 -yl} piperidin- 1 -yl)carbonyl] -5 -fluorobenzonitrile;
4- [(4-{3-(cyanomethyl)-3-[3-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrrol-l- yl] azetidin- 1 -yl} piperidin- 1 -yl)carbonyl] -3 -fluorobenzonitrile;
4-[(4-{3-(cyanomethyl)-3-[3-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrrol-l- yl] azetidin- 1 -yl} piperidin- 1 -yl)carbonyl] -3 ,5 -difluorobenzonitrile;
{ 1 - { 1 -[5-fluoro-2-(trifiuoromethyl)benzoyl]piperidin-4-yl} -3-[3-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrrol- 1 -yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[3-fluoro-4-(trifiuoromethyl)benzoyl]piperidin-4-yl} -3-[3-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrrol- 1 -yl]azetidin-3-yl} acetonitrile;
(3-[3-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrrol- 1 -yl]- 1 - { 1 -[3- (trifiuoromethyl)benzoyl]piperidin-4-yl}azetidin-3-yl)acetonitrile;
{ 1 - { 1 -[2-fluoro-5-(trifiuoromethoxy)benzoyl]piperidin-4-yl} -3-[3-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrrol-l-yl]azetidin-3-yl} acetonitrile;
{3-[3-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrrol-l-yl]-l-[l-(2,3,6- trifluorobenzoyl)piperidin-4-yl]azetidin-3-yl} acetonitrile;
{3-[3-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrrol-1 -yl]-l -[ 1 -(2- thienylcarbonyl)piperidin-4-yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[2-fluoro-4-(trifiuoromethyl)benzoyl]piperidin-4-yl} -3-[3-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrrol- 1 -yl]azetidin-3-yl} acetonitrile; { 1 - { 1 -[2-fluoro-5-(trifluoromethyl)benzoyl]piperidin-4-yl} -3-[3-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrrol- 1 -yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[3-fluoro-5-(trifluoromethyl)benzoyl]piperidin-4-yl} -3-[3-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrrol- 1 -yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[4-fluoro-3-(trifluoromethyl)benzoyl]piperidin-4-yl} -3-[3-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrrol- 1 -yl]azetidin-3-yl} acetonitrile;
{l-[l-(2,3-difluorobenzoyl)piperidin-4-yl]-3-[3-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)- 1 H-pyrrol- 1 -yl]azetidin-3-yl} acetonitrile;
{l-[l-(3,4-difluorobenzoyl)piperidin-4-yl]-3-[3-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)- 1 H-pyrrol- 1 -yl]azetidin-3-yl} acetonitrile;
{l-[l-(2,5-difluorobenzoyl)piperidin-4-yl]-3-[3-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)- 1 H-pyrrol- 1 -yl]azetidin-3-yl} acetonitrile;
{l-[l-(2,6-difluorobenzoyl)piperidin-4-yl]-3-[3-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)- 1 H-pyrrol- 1 -yl]azetidin-3-yl} acetonitrile;
{3-[3-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrrol-l-yl]-l-[l-(2,3,4- trifluorobenzoyl)piperidin-4-yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[2-fluoro-3-(trifluoromethoxy)benzoyl]piperidin-4-yl} -3-[3-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrrol-l-yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[4-hydroxy-3-(trifluoromethyl)benzoyl]piperidin-4-yl} -3-[3-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrrol-l-yl]azetidin-3-yl} acetonitrile;
4- {3-(cyanomethyl)-3-[3-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrrol- 1 - yljazetidin- 1 -yl} -N-[4-fluoro-2-(trifluoromethyl)phenyl]piperidine- 1 -carboxamide;
4- {3-(cyanomethyl)-3-[3-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrrol- 1 - yl] azetidin- 1 -yl} -N-(2-methoxypyridin-3 -yl)piperidine- 1 -carboxamide;
4- {3-(cyanomethyl)-3-[3-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrrol- 1 - yljazetidin- 1-yl} -N-[2-methyl-6-(trifluoromethyl)pyridin-3-yl]piperidine- 1 -carboxamide;
4- {3-(cyanomethyl)-3-[3-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrrol- 1 - yljazetidin- 1 -yl} -N-(2,4-difluorophenyl)piperidine- 1 -carboxamide;
4- {3-(cyanomethyl)-3-[3-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrrol- 1 - yljazetidin- 1 -yl} -N-(2-cyanophenyl)piperidine- 1 -carboxamide; 4- {3-(cyanomethyl)-3-[3-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrrol- 1 - yljazetidin- 1 -yl} -N-(2-methoxyphenyl)piperidine- 1 -carboxamide;
N-(2-chloro-4-fluorophenyl)-4-{3-(cyanomethyl)-3-[3-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrrol- 1 -yljazetidin- 1 -yl}piperidine- 1 -carboxamide;
4- {3-(cyanomethyl)-3-[3-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrrol- 1 - yl]azetidin-l-yl}-N-[3-(trifluoromethyl)pyridin-2-yl]piperidine-l -carboxamide;
4- {3-(cyanomethyl)-3-[3-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrrol- 1 - yl]azetidin-l-yl}-N-[4-(trifluoromethyl)pyridin-3-yl]piperidine-l -carboxamide;
4- {3-(cyanomethyl)-3-[3-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrrol- 1 - yljazetidin- 1 -yl} -N-(3-fluoropyridin-2-yl)piperidine- 1 -carboxamide;
N-(4-chloro-2-cyanophenyl)-4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yljazetidin- 1 -yl}piperidine- 1 -carboxamide;
{ 1 - { 1 -[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl} -3-[4-(lH- pyrrolo[2,3-b]pyridin-4-yl)- lH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
4-[(4-{3-(cyanomethyl)-3-[4-(lH-pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol-l- yl] azetidin- 1 -yl} piperidin- 1 -yl)carbonyl] -3 -fluorobenzonitrile;
{l-[l-(3-fluoro-4-hydroxybenzoyl)piperidin-4-yl]-3-[4-(lH-pyrrolo[2,3- b]pyridin-4-yl)- 1 H-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{l-[l-(2-fluoro-4-hydroxybenzoyl)piperidin-4-yl]-3-[4-(lH-pyrrolo[2,3- b]pyridin-4-yl)- 1 H-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[5-chloro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl} -3-[4-( 1 H- pyrrolo[2,3-b]pyridin-4-yl)- lH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
[3-[4-(l H-pyrrolo [2,3-b]pyridin-4-yl)- 1 H-pyrazol- 1 -yl]- 1 -(1 - { [2- (trifluoromethyl)pyrimidin-4-yl]carbonyl}piperidin-4-yl)azetidin-3-yl]acetonitrile;
[3-[4-(lH-pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol-l-yl]-l-(l-{[6- (trifluoromethyl)pyrazin-2-yl]carbonyl}piperidin-4-yl)azetidin-3-yl]acetonitrile;
[3-[4-(lH-pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol-l-yl]-l-(l-{[5- (trifluoromethyl)pyrazin-2-yl]carbonyl}piperidin-4-yl)azetidin-3-yl]acetonitrile;
{ 1 - { 1 -[(4,4-difluorocyclohexyl)carbonyl]piperidin-4-yl} -3-[4-(lH-pyrrolo[2,3- b]pyridin-4-yl)- 1 H-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile; 4-{3-(cyanomethyl)-3-[4-(lH-pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol-l- yljazetidin- 1 -yl} -N-[4-fluoro-2-(trifluoromethyl)phenyl]piperidine- 1 -carboxamide;
4-{3-(cyanomethyl)-3-[4-(lH-pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol-l- yljazetidin- 1 -yl} -N-(2,4-difluorophenyl)piperidine- 1 -carboxamide;
N-(2-chloro-4-fluorophenyl)-4-{3-(cyanomethyl)-3-[4-(lH-pyrrolo[2,3-b]pyridin- 4-yl)- 1 H-pyrazol- 1 -yl] azetidin- 1 -yl} piperidine- 1 -carboxamide;
4-{3-(cyanomethyl)-3-[4-(lH-pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol-l- yl] azetidin- 1 -yl} -N-(2-methoxypyridin-3 -yl)piperidine- 1 -carboxamide;
4-{3-(cyanomethyl)-3-[4-(lH-pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol-l- yl]azetidin-l-yl}-N-[3-(trifluoromethyl)pyridin-4-yl]piperidine-l -carboxamide;
4-{3-(cyanomethyl)-3-[4-(lH-pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol-l- yl]azetidin-l-yl}-N-[4-(trifluoromethyl)pyridin-3-yl]piperidine-l -carboxamide;
4-{3-(cyanomethyl)-3-[4-(lH-pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol-l- yljazetidin- 1 -yl} -N-(2-fluorophenyl)piperidine- 1 -carboxamide;
N-(2-chlorophenyl)-4-{3-(cyanomethyl)-3-[4-(lH-pyrrolo[2,3-b]pyridin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin- 1 -yl} piperidine- 1 -carboxamide;
4- {3-(cyanomethyl)-3-[4-(lH-pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol-l- yl] azetidin- 1-yl} -N-[4-cyano-2-(trifluoromethyl)phenyl]piperidine- 1 -carboxamide;
N-(4-cyano-2-fluorophenyl)-4-{3-(cyanomethyl)-3-[4-(lH-pyrrolo[2,3-b]pyridin- 4-yl)- 1 H-pyrazol- 1 -yljazetidin- 1 -yl}piperidine- 1 -carboxamide;
N-(2-chloro-4-cyanophenyl)-4-{3-(cyanomethyl)-3-[4-(lH-pyrrolo[2,3-b]pyridin- 4-yl)- 1 H-pyrazol- 1 -yljazetidin- 1 -yl}piperidine- 1 -carboxamide;
(3-[4-(5-fluoro-lH-pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol-l-yl]-l-{l-[3-fluoro- 2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}azetidin-3-yl)acetonitrile;
5- [(4-{3-(cyanomethyl)-3-[4-(5-fluoro-lH-pyrrolo[2,3-b]pyridin-4-yl)-lH- pyrazol- 1 -yljazetidin- 1 -yl}piperidin- 1 -yl)carbonyl]isophthalonitrile;
3- [(4-{3-(cyanomethyl)-3-[4-(5-fluoro-lH-pyrrolo[2,3-b]pyridin-4-yl)-lH- pyrazol- 1 -yljazetidin- 1 -yl}piperidin- 1 -yl)carbonyl]-5-fluorobenzonitrile;
4- [(4-{3-(cyanomethyl)-3-[4-(5-fluoro-lH-pyrrolo[2,3-b]pyridin-4-yl)-lH- pyrazol- 1 -yljazetidin- 1 -yl}piperidin- 1 -yl)carbonyl]-3-fluorobenzonitrile; 5-[(4-{3-(cyanomethyl)-3-[4-(5-fluoro-lH-pyrrolo[2,3-b]pyridin-4-yl)-lH- pyrazol- 1 -yljazetidin- 1 -yl}piperidin- 1 -yl)carbonyl]-2-fluorobenzonitrile;
{ 1 - { 1 -[(5-fluoropyridin-2-yl)carbonyl]piperidin-4-yl} -3-[4-(5-fluoro- 1H- pyrrolo[2,3-b]pyridin-4-yl)- lH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{ 1 - [ 1 -(3 -fluoroisonicotinoyl)piperidin-4-yl] -3 - [4-(5 -fluoro- 1 H-pyrrolo [2,3 - b]pyridin-4-yl)- 1 H-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{l-[l-(3,5-difluoroisonicotinoyl)piperidin-4-yl]-3-[4-(5-fluoro-lH-pyrrolo[2,3- b]pyridin-4-yl)- 1 H-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{l-[l-(3-fluoro-4-hydroxybenzoyl)piperidin-4-yl]-3-[4-(5-fluoro-lH-pyrrolo[2,3- b]pyridin-4-yl)- 1 H-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{l-[l-(2-fluoro-4-hydroxybenzoyl)piperidin-4-yl]-3-[4-(5-fluoro-lH-pyrrolo[2,3- b]pyridin-4-yl)- 1 H-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
2-[(4-{3-(cyanomethyl)-3-[4-(5-fluoro-lH-pyrrolo[2,3-b]pyridin-4-yl)-lH- pyrazol- 1 -yl] azetidin- 1 -yl} piperidin- 1 -yl)carbonyl]terephthalonitrile;
4-[(4-{3-(cyanomethyl)-3-[4-(5-fluoro-lH-pyrrolo[2,3-b]pyridin-4-yl)-lH- pyrazol- 1 -yljazetidin- 1 -yl}piperidin- 1 -yl)carbonyl]-2-fluorobenzonitrile;
{ 1 - { 1 -[5-chloro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl} -3-[4-(5-fluoro- lH-pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[(4,4-difluorocyclohexyl)carbonyl]piperidin-4-yl} -3 -[4-(5 -fluoro- 1H- pyrrolo[2,3-b]pyridine-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
4-{3-(cyanomethyl)-3-[4-(5-fluoro-lH-pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol-l- yljazetidin- 1 -yl} -N-[4-fluoro-2-(trifluoromethyl)phenyl]piperidine- 1 -carboxamide;
4-{3-(cyanomethyl)-3-[4-(5-fluoro-lH-pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol-l- yl]azetidin-l-yl}-N-[3-(trifluoromethyl)pyridin-2-yl]piperidine-l -carboxamide;
4-{3-(cyanomethyl)-3-[4-(5-fluoro-lH-pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol-l- yl]azetidin-l-yl}-N-[4-(trifluoromethyl)pyridin-3-yl]piperidine-l -carboxamide;
4-{3-(cyanomethyl)-3-[4-(5-fluoro-lH-pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol-l- yl]azetidin-l-yl}-N-[3-(trifluoromethyl)pyridin-4-yl]piperidine-l -carboxamide;
4-[ 1 -(3-(cyanomethyl)- 1 - { 1 -[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin- 4-yl}azetidin-3-yl)-lH-pyrazol-4-yl]-lH-pyrrolo[2,3-b]pyridine-5-carbonitrile; 4- { 1 -[ 1 -[1 -(3-cyano-5-fluorobenzoyl)piperidin-4-yl]-3-(cyanomethyl)azetidin-3- yl]-lH-pyrazol-4-yl}-lH-pyrrolo[2,3-b]pyridine-5-carbonitrile;
4- { 1 -[ 1 -[1 -(4-cyano-3-fluorobenzoyl)piperidin-4-yl]-3-(cyanomethyl)azetidin-3- yl]-lH-pyrazol-4-yl}-lH-pyrrolo[2,3-b]pyridine-5-carbonitrile;
4-(l - {3-(cyanomethyl)-l -[ 1 -(2,5-dibromobenzoyl)piperidin-4-yl]azetidin-3-yl} - lH-pyrazol-4-yl)-lH-pyrrolo[2,3-b]pyridine-5-carbonitrile;
4- (l - {3-(cyanomethyl)-l -[ 1 -(3,5-dibromobenzoyl)piperidin-4-yl]azetidin-3-yl} - lH-pyrazol-4-yl)-lH-pyrrolo[2,3-b]pyridine-5-carbonitrile;
2-[(4-{3-(cyanomethyl)-3-[4-(5-cyano-lH-pyrrolo[2,3-b]pyridin-4-yl)-lH- pyrazol- 1 -yl] azetidin- 1 -yl} piperidin- 1 -yl)carbonyl]terephthalonitrile;
5- [(4-{3-(cyanomethyl)-3-[4-(5-cyano-lH-pyrrolo[2,3-b]pyridin-4-yl)-lH- pyrazol- 1 -yljazetidin- 1 -yl}piperidin- 1 -yl)carbonyl]isophthalonitrile;
4- { 1 -[ 1 -[1 -(4-cyano-2-fluorobenzoyl)piperidin-4-yl]-3-(cyanomethyl)azetidin-3- yl]-lH-pyrazol-4-yl}-lH-pyrrolo[2,3-b]pyridine-5-carbonitrile;
4- { 1 -[ 1 -[1 -(4-cyano-2,6-difluorobenzoyl)piperidin-4-yl]-3- (cyanomethyl)azetidin-3-yl]-lH-pyrazol-4-yl}-lH-pyrrolo[2,3-b]pyridine-5-carbonitrile;
4- { 1 -[ 1 - { 1 -[5-chloro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl} -3- (cyanomethyl)azetidin-3-yl]-lH-pyrazol-4-yl}-lH-pyrrolo[2,3-b]pyridine-5-carbonitrile;
{ 1 - { 1 -[5-Chloro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl} -3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[5-Fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl} -3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
[3-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol- 1 -yl] - 1 -(1 - {[4- (trifluoromethyl)- 1 ,3 -thiazol-2-yl] carbonyl} piperidin-4-yl)azetidin-3 -yl] acetonitrile;
[3-[3-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrrol- 1 -yl]- 1 -(1 - {[2- (trifluoromethyl)pyrimidin-4-yl]carbonyl}piperidin-4-yl)azetidin-3-yl]acetonitri
[3-[3-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrrol- 1 -yl]- 1 -(1 - {[4- (trifluoromethyl)- 1 ,3 -thiazol-2-yl] carbonyl} piperidin-4-yl)azetidin-3 -yl] acetonitrile;
[3-[3-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrrol- 1 -yl]- 1 -(1 - {[5- (trifluoromethyl)pyrazin-2-yl] carbonyl} piperidin-4-yl)azetidin-3 -yl] acetonitrile; {l-[l-(Methylsulfonyl)piperidin-4-yl]-3-[3-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- 1 H-pyrrol- 1 -yl] azetidin-3 -yl} acetonitrile;
[3-[4-(lH-Pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol-l-yl]-l-(l-{[5- (trifluoromethyl)pyrazin-2-yl] carbonyl} piperidin-4-yl)azetidin-3 -yl] acetonitrile;
[3-[4-(lH-Pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol-l-yl]-l-(l-{[4- (trifluoromethyl)- 1 ,3 -thiazol-2-yl] carbonyl} piperidin-4-yl)azetidin-3 -yl] acetonitrile;
{1 1-(Methylsulfonyl)piperidin-4-yl]-3-[4-(lH-pyrrolo[2,3-b]pyridin-4-yl)-lH- pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
[3-[4-(5-Fluoro-lH-pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol-l-yl]-l-(l-{[6- (trifluoromethyl)pyrazin-2-yl] carbonyl} piperidin-4-yl)azetidin-3 -yl] acetonitrile;
[3-[4-(5-Fluoro-lH-pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol-l-yl]-l-(l-{[2- (trifluoromethyl)pyrimidin-4-yl]carbonyl}piperidin-4-yl)azetidin-3-yl]acetonitri
[3-[4-(5-Fluoro-lH-pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol-l-yl]-l-(l-{[5- (trifluoromethyl)pyrazin-2-yl] carbonyl} piperidin-4-yl)azetidin-3 -yl] acetonitrile;
{3-[4-(5-Fluoro-lH-pyrrolo[2,3-b]pyridin-4-yl)-lHpyrazol-l-yl]-l-[l- (methylsulfonyl)piperidin-4-yl]azetidin-3-yl} acetonitrile;
4- [ 1 -(3 -(Cyanomethyl)- 1 - { 1 - [5 -fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin- 4-yl}azetidin-3-yl)-lH-pyrazol-4-yl]-lH-pyrrolo[2,3-b]pyridine-5-carbonitrile; and
4-(l - (3-(Cyanomethyl)- 1 -[1 -(methylsulfonyl)piperidin-4-yl]azetidin-3-yl} -1 H- pyrazol-4-yl)-lH-pyrrolo[2,3-b]pyridine-5-carbonitrile;
or a pharmaceutically acceptable salt of any of the aforementioned.
In some embodiments, the compound is selected from:
cis- { 1 - {(3-Methoxy- 1 -[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl} - 3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-l H-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{ 1 -(cis-3 -Methoxy- 1 - { [2-(trifluoromethyl)pyrimidin-4-yl] carbonyl} piperidin-4- yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{ 1 - {cis-3-Fluoro-l -[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl} -3- [4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{ 1 -(cis-3 -Fluoro- 1 - { [2-(trifluoromethyl)pyrimidin-4-yl] carbonyl} piperidin-4-yl)- 3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile; { 1 - { 1 -[3-Fluoro-2-(trifluoromethyl)isonicotinoyl]-4-deuteropiperidin-4-yl} -3-[4- (7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile;
{l-{l-[3-Fluoro-2-(trifluoromethyl)isonicotinoyl]3,3,4,5,5-pentadeuteropiperidin- 4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile;
{ 1 - {7-[3-Fluoro-2-(trifluoromethyl)isonicotinoyl]-3-oxa-7-azabicyclo[3.3.1 ]non- 9-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile;
{ 1 -(1 - { [4-[(dimethylamino)methyl]-6-(trifluoromethyl)pyridin-2- yl]carbonyl}piperidin-4-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-3-yl} acetonitrile;
4- {3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 - yljazetidin- 1 -yl} -N-isopropylpiperidine- 1 -carboxamide;
{ 1 - { 1 - [6- [(dimethylamino)methyl] -3 -fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
3-[(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin- 1 -yl} piperidin- 1 -yl)carbonyl] -5 -[(dimethylamino)methyl]benzonitrile;
{ 1 -(1 - { [6-[(dimethylamino)methyl]-2-(trifluoromethyl)pyrimidin-4- yl]carbonyl}piperidin-4-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-3-yl} acetonitrile;
{ 1 -(1 - {[6-[(methylamino)methyl]-2-(trifluoromethyl)pyrimidin-4- yl]carbonyl}piperidin-4-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-3-yl} acetonitrile;
{ 1 -( 1 - { [6-(azetidin- 1 -ylmethyl)-2-(trifluoromethyl)pyrimidin-4- yl]carbonyl}piperidin-4-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-3-yl} acetonitrile;
{ 1 -(1 - {[6-[(diethylamino)methyl]-2-(trifluoromethyl)pyrimidin-4- yl]carbonyl}piperidin-4-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-3-yl} acetonitrile;
{ 1 -(1 - { [6- { [ethyl(methyl)amino]methyl} -2-(trifluoromethyl)pyrimidin-4- yl]carbonyl}piperidin-4-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-3-yl} acetonitrile; { 1 -(1 - {3-(difluoromethyl)-5-[(dimethylamino)methyl]benzoyl}piperidin-4-yl)-3- [4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{ 1 -(1 - { [6-(pyrrolidin- 1 -ylmethyl)-2-(trifluoromethyl)pyrimidin-4- yl]carbonyl}piperidin-4-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-3-yl} acetonitrile;
{l-(l-{[6-{[(3 S)-3 -fluoropyrrolidm- 1 -yljmethyl} -2-(trifluoromethyl)pyrimidin-4- yl]carbonyl}piperidin-4-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-3-yl} acetonitrile;
{ 1 -(1 - { [6- { [(3R)-3-fluoropyrrolidin- 1 -yljmethyl} -2-(trifluoromethyl)pyrimidin-4- yl]carbonyl}piperidin-4-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-3-yl} acetonitrile;
{l-(l-{[6-[(3 ,3 -difluoropyrrolidin- 1 -yl)methyl] -2-(trifluoromethyl)pyrimidin-4- yl]carbonyl}piperidin-4-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-3-yl} acetonitrile;
{ 1 -(1 - {[6-[(tert-butylamino)methyl]-2-(trifluoromethyl)pyrimidin-4- yl]carbonyl}piperidin-4-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-3-yl} acetonitrile;
{l-(l-{[6-(hydroxymethyl)-2-(trifluoromethyl)pyrimidin-4-yl]carbonyl}piperidin- 4-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{ 1 -(1 - { [6-[(isopropylamino)methyl]-2-(trifluoromethyl)pyrimidin-4- yl]carbonyl}piperidin-4-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-3-yl} acetonitrile;
{ 1 -(1 - { [6-[(ethylamino)methyl]-2-(trifluoromethyl)pyrimidin-4- yl]carbonyl}piperidin-4-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-3-yl} acetonitrile;
{l-(l-{[6-{ [(2-methoxyethyl)(methyl)amino]methyl} -2- (trifluoromethyl)pyrimidin-4-yl] carbonyl} piperidin-4-yl)-3 -[4-(7H-pyrrolo [2,3 - d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-(l-{[6-{[(3 -hydroxypropyl)(methyl)amino]methyl} -2- (trifluoromethyl)pyrimidin-4-yl] carbonyl} piperidin-4-yl)-3 -[4-(7H-pyrrolo [2,3 - d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile; propyl 4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol- 1 -yljazetidin- 1 -yl}piperidine- 1 -carboxylate;
cyclobutylmethyl 4- { 3 -(cyanomethyl)-3 - [4-(7H-pyrrolo [2,3 -d]pyrimidin-4-yl)- lH-pyrazol- 1 -yljazetidin- 1 -yl}piperidine- 1 -carboxylate;
ethyl 4- {3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 - yljazetidin- 1 -yl}piperidine- 1 -carboxylate;
benzyl 4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol- 1 -yljazetidin- 1 -yl}piperidine- 1 -carboxylate;
isobutyl 4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol- 1 -yljazetidin- 1 -yl}piperidine- 1 -carboxylate;
cyclopropylmethyl 4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 -yljazetidin- 1 -yl}piperidine- 1 -carboxylate;
( 1 -methylcyclopropyl)methyl 4- {3 -(cyanomethyl)-3 - [4-(7H-pyrrolo [2,3 - d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin- 1 -yl} piperidine- 1 -carboxylate;
2,4-difluorobenzyl 4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yljazetidin- 1 -yl} piperidine- 1 -carboxylate;
3.4- difluorobenzyl 4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yljazetidin- 1 -yl} piperidine- 1 -carboxylate;
3.5- difluorobenzyl 4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yljazetidin- 1 -yl} piperidine- 1 -carboxylate;
cyclopentylmethyl 4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 -yljazetidin- 1 -yl}piperidine- 1 -carboxylate; and
cyclohexylmethyl 4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yljazetidin- 1 -yl} piperidine- 1 -carboxylate;
or a pharmaceutically acceptable salt of any of the aforementioned.
In some embodiments, the salt is a l-{l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile adipic acid salt. In some embodiments, the salt is a 1 : 1 l-{l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile : adipic acid salt. In some embodiments, the salt is that described in Example 358. In some embodiments, the salt is characterized by a melting point of about 178 °C. In some embodiments, the salt has a differential scanning calorimetry thermogram which is characterized by an endothermic peak with an onset temperature of about 176 °C. In some embodiments, the salt has a differential scanning calorimetry thermogram substantially as shown in Figure 1.
In some embodiments, the salt has a thermal gravimetric analysis thermogram substantially as shown in Figure 2. In some embodiments, the salt has an X-ray powder diffraction pattern comprising a characteristic peak expressed in degrees 2Θ at about 10.4. In some embodiments, the salt has an X-ray powder diffraction pattern comprising a characteristic peak expressed in degrees 2Θ at about 6.9. In some embodiments, the salt has an X-ray powder diffraction pattern comprising a characteristic peak expressed in degrees 2Θ at about 21.0. In some embodiments, the salt has an X-ray powder diffraction pattern comprising a characteristic peak expressed in degrees 2Θ at about 23.3. In some embodiments, the salt has an X-ray powder diffraction pattern comprising a
characteristic peak expressed in degrees 2Θ at about 6.9, 10.4, 21.0, and 23.3. In some embodiments, the salt has an X-ray powder diffraction pattern substantially as shown in Figure 3.
An XRPD pattern of reflections (peaks) is typically considered a fingerprint of a particular crystalline form. It is well known that the relative intensities of the XRPD peaks can widely vary depending on, inter alia, the sample preparation technique, crystal size distribution, various filters used, the sample mounting procedure, and the particular instrument employed. In some instances, new peaks may be observed or existing peaks may disappear, depending on the type of the machine or the settings (for example, whether a Ni filter is used or not). As used herein, the term "peak" refers to a reflection having a relative height/intensity of at least about 4% of the maximum peak
height/intensity. Moreover, instrument variation and other factors can affect the 2-theta values. Thus, peak assignments, such as those reported herein, can vary by plus or minus about 0.2° (2-theta), and the term "substantially" as used in the context of XRPD herein is meant to encompass the above-mentioned variations.
In the same way, temperature readings in connection with DSC, TGA, or other thermal experiments can vary about ±3 °C depending on the instrument, particular settings, sample preparation, etc. Accordingly, a crystalline form reported herein having a DSC thermogram "substantially" as shown in any of the Figures is understood to accommodate such variation.
It is further appreciated that certain features of the invention, which are, for clarity, described in the context of separate embodiments, can also be provided in combination in a single embodiment. Conversely, various features of the invention which are, for brevity, described in the context of a single embodiment, can also be provided separately or in any suitable subcombination.
At various places in the present specification, substituents of compounds of the invention are disclosed in groups or in ranges. It is specifically intended that the invention include each and every individual subcombination of the members of such groups and ranges. For example, the term "Ci_6 alkyl" is specifically intended to individually disclose methyl, ethyl, C3 alkyl, C4 alkyl, C5 alkyl, and C6 alkyl.
At various places in the present specification, linking substituents are described. It is specifically intended that each linking substituent include both the forward and backward forms of the linking substituent. For example, -NR(CR'R")n- includes both - NR(CR'R")n- and -(CR'R")nNR-. Where the structure clearly requires a linking group, the Markush variables listed for that group are understood to be linking groups. For example, if the structure requires a linking group and the Markush group definition for that variable lists "alkyl" or "aryl" then it is to be understood that the "alkyl" or "aryl" represents a linking alkylene group or arylene group, respectively.
At various places in the present specification, rings are described (e.g., "a piperidine ring"). Unless otherwise specified, these rings can be attached to the rest of the molecule at any ring member as permitted by valency. For example, the term "a pyridine ring" may refer to a pyridin-2-yl, pyridin-3-yl, or pyridin-4-yl ring.
The term "n-membered" where n is an integer typically describes the number of ring-forming atoms in a moiety where the number of ring-forming atoms is n. For example, piperidinyl is an example of a 6-membered heterocycloalkyl ring, pyrazolyl is an example of a 5-membered heteroaryl ring, pyridyl is an example of a 6-membered heteroaryl ring, and 1 ,2,3,4-tetrahydro-naphthalene is an example of a 10-membered cycloalkyl group. For compounds of the invention in which a variable appears more than once, each variable can be a different moiety independently selected from the group defining the variable. For example, where a structure is described having two R groups that are simultaneously present on the same compound, the two R groups can represent different moieties independently selected from the group defined for R. In another example, when an optionally multiple substituent is designated in the form:
Figure imgf000056_0001
then it is to be understood that substituent R can occur p number of times on the ring, and R can be a different moiety at each occurrence. It is to be understood that each R group may replace any hydrogen atom attached to a ring atom, including one or both of the (CH2)n hydrogen atoms. Further, in the above example, should the variable Q be defined to include hydrogens, such as when Q is said to be CH2, NH, etc., any floating substituent such as R in the above example, can replace a hydrogen of the Q variable as well as a hydrogen in any other non-variable component of the ring.
As used herein, the phrase "optionally substituted" means unsubstituted or substituted. As used herein, the term "substituted" means that a hydrogen atom is removed and replaced by a substituent. It is to be understood that substitution at a given atom is limited by valency.
As used herein, the term "Cn_m alkyl", employed alone or in combination with other terms, refers to a saturated hydrocarbon group that may be straight-chain or branched, having n to m carbon atoms. In some embodiments, the alkyl group contains 1 to 6, 1 to 4 or 1 to 3 carbon atoms. Examples of alkyl moieties include, but are not limited to, chemical groups such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, 2 -methyl- 1 -butyl, 3-pentyl, n-hexyl, 1,2,2-trimethylpropyl, n-heptyl, n-octyl, and the like.
As used herein, "Cn_m alkenyl", employed alone or in combination with other terms, refers to an alkyl group having one or more carbon-carbon double bonds and n to m carbon atoms. In some embodiments, the alkenyl moiety contains 2 to 6, or 2 to 4 carbon atoms. Example alkenyl groups include, but are not limited to, ethenyl, n- propenyl, isopropenyl, n-butenyl, sec-butenyl, and the like.
As used herein, "Cn_m alkynyl", employed alone or in combination with other terms, refers to an alkyl group having one or more carbon-carbon triple bonds and n to m carbon atoms. Example alkynyl groups include, but are not limited to, ethynyl, propyn-1- yl, propyn-2-yl, and the like. In some embodiments, the alkynyl moiety contains 2 to 6 or 2 to 4 carbon atoms.
As used herein, "halo" or "halogen", employed alone or in combination with other terms, includes fluoro, chloro, bromo, and iodo.
As used herein, "hydroxyl" or "hydroxy" refer to a group of formula -OH.
As used herein, the term "Cn_m haloalkyl", employed alone or in combination with other terms, refers to an Cn_m alkyl group having up to {2(n to m)+l } halogen atoms which may either be the same or different. In some embodiments, the halogen atoms are fluoro atoms. In some embodiments, the alkyl group has 1 to 6 or 1 to 4 carbon atoms. Example haloalkyl groups include CF3, C2F5, CHF2, CC13, CHC12, C2CI5, and the like. In some embodiments, the haloalkyl group is a fluoroalkyl group.
As used herein, the term "Cn_m fluoroalkyl", employed alone or in combination with other terms, refers to a Cn_m haloalkyl wherein the halogen atoms are selected from fluorine. In some embodiments, Cn_m fluroalkyl is fluoromethyl, difluoromethyl, or trifluoromethyl.
As used herein, the term "Cn_m alkoxy", employed alone or in combination with other terms, refers to an group of formula -O-alkyl, wherein the alkyl group has n to m carbon atoms. Example alkoxy groups include methoxy, ethoxy, propoxy (e.g., n- propoxy and isopropoxy), t-butoxy, and the like. In some embodiments, the alkyl group has 1 to 6 or 1 to 4 carbon atoms.
As used herein, "Cn_m haloalkoxy", employed alone or in combination with other terms, refers to a group of formula -O-(haloalkyl), wherein the haloalkyl group has n to m carbon atoms. In some embodiments, the alkyl group has 1 to 6 or 1 to 4 carbon atoms. An example haloalkoxy group is -OCF3. In some embodiments, the haloalkoxy group is a fluoroalkoxy group. As used herein, the term "Cn_m fluoroalkoxy", employed alone or in combination with other terms, refers to a Cn_m alkoxy group, wherein the halogen atoms are selected from fluorine.
As used herein, "amino", employed alone or in combination with other terms, refers to -NH2.
As used herein, the term "Cn_m alkylamino", employed alone or in combination with other terms, refers to a group of formula -NH(alkyl), wherein the alkyl group has n to m carbon atoms. In some embodiments, alkyl group has 1 to 6 or 1 to 4 carbon atoms. Example Cn_m alkylamino groups include methylamino, ethylamino, propylamino (e.g., n- propylamino and isopropylamino), and the like.
As used herein, the term "di-Cn_m-alkylamino", employed alone or in combination with other terms, refers to a group of formula -N(alkyl)2, wherein each alkyl group has independently n to m carbon atoms. Example di-Cn_m-alkylamino groups include dimethylamino, diethylamino, dipropylamino (e.g., di(n-propyl)amino and
di(isopropyl)amino), and the like. In some embodiments, each alkyl group independently has 1 to 6 or 1 to 4 carbon atoms.
As used herein, the term "carboxy", employed alone or in combination with other terms, refers to a group of formula -C(0)OH.
As used herein, the term "Cn_m alkoxycarbonyl", employed alone or in
combination with other terms, refers to a group of formula -C(0)0-alkyl, wherein the alkyl group has n to m carbon atoms. In some embodiments, the alkyl group has 1 to 6 or 1 to 4 carbon atoms.
As used herein, the term "Cn_m alkylcarbonyl", employed alone or in combination with other terms, refers to a group of formula -C(0)-alkyl, wherein the alkyl group has n to m carbon atoms. In some embodiments, the alkyl group has 1 to 6 or 1 to 4 carbon atoms.
As used herein, the term "Cn_m alkylcarbonylamino", employed alone or in combination with other terms, refers to a group of formula -NHC(0)-alkyl, wherein the alkyl group has n to m carbon atoms. In some embodiments, the alkyl group has 1 to 6 or 1 to 4 carbon atoms. As used herein, the term "carbamyl", employed alone or in combination with other terms, refers to a group of formula -C(0)-NH2.
As used herein, the term "Cn_m alkylcarbamyl", employed alone or in combination with other terms, refers to a group of formula -C(0)-NH(alkyl), wherein the alkyl group has n to m carbon atoms. In some embodiments, the alkyl group has 1 to 6 or 1 to 4 carbon atoms.
As used herein, the term "di-Cn_m alkylcarbamyl", employed alone or in combination with other terms, refers to a group of formula -C(0)-N(alkyl)2, wherein the each alkyl group independently has n to m carbon atoms. In some embodiments, the alkyl group independently has 1 to 6 or 1 to 4 carbon atoms.
As used herein, the term "thio" refers to a group of formula -SH.
As used herein, the term "Cn_m alkylthio", employed alone or in combination with other terms, refers to a group of formula -S-alkyl, wherein the alkyl group has n to m carbon atoms. In some embodiments, the alkyl group has 1 to 6 or 1 to 4 carbon atoms.
As used herein, the term "Cn_m alkylsulfmyl", employed alone or in combination with other terms, refers to a group of formula -S(0)-alkyl, wherein the alkyl group has n to m carbon atoms. In some embodiments, the alkyl group has 1 to 6 or 1 to 4 carbon atoms.
As used herein, the term "Cn_m alkylsulfonyl", employed alone or in combination with other terms, refers to a group of formula -S(0)2-alkyl, wherein the alkyl group has n to m carbon atoms. In some embodiments, the alkyl group has 1 to 6 or 1 to 4 carbon atoms.
As used herein, "halosulfanyl" refers to a sulfur group having one or more halogen substituents. Example halosulfanyl groups include pentahalosulfanyl groups such as SF5.
As used herein, the term "2- or 3-carbon bridge" means that two different R groups on different ring member atoms form a bridge (-CH2-CH2- or -CH2-CH2-CH2-) between the two ring member atoms, wherein the two or three carbons does not include the ring member atoms. For a non- limiting examples, see Example 138, where two R1 groups form a 2-carbon bridge. As used herein, the term "bridge of formula -CH2-O-CH2-" means that two different R groups on different ring member atoms form a bridge between the two ring member atoms of formula -CH2-O-CH2-, where the ring member atoms are not part of formula -CH2-0-CH2-.
As used herein, the term "hydroxyl-Ci_4 alkyl" refers to a group of formula -C1-4 alkylene-OH.
As used herein, the term "Ci_4 alkoxy-Ci_4 alkyl" refers to a group of formula -Ci_ 4 alkylene-0-(Ci_4 alkyl).
As used herein, the term "Cn_m cycloalkyl", employed alone or in combination with other terms, refers to a non-aromatic cyclic hydrocarbon including cyclized alkyl, alkenyl, and alkynyl groups, and which has n to m ring member carbon atoms.
Cycloalkyl groups can include mono- or polycyclic (e.g., having 2, 3, or 4 fused, bridged, or spiro rings) ring systems. Also included in the definition of cycloalkyl are moieties that have one or more aromatic rings (e.g., aryl or heteroaryl rings) fused (i.e., having a bond in common with) to the cycloalkyl ring, for example, benzo derivatives of cyclopentane, cyclopentene, cyclohexane, and the like. Ring-forming carbon atoms of a cycloalkyl group can be optionally substituted by oxo. Cycloalkyl groups also include cycloalkylidenes. The term "cycloalkyl" also includes bridgehead cycloalkyl groups and spirocycloalkyl groups. As used herein, "bridgehead cycloalkyl groups" refers to non- aromatic cyclic hydrocarbon moieties containing at least one bridgehead carbon, such as admantan-l-yl. As used herein, "spirocycloalkyl groups" refers to non-aromatic hydrocarbon moieties containing at least two rings fused at a single carbon atom, such as spiro[2.5]octane and the like. In some embodiments, the cycloalkyl group has 3 to 14 ring members, 3 to 10 ring members, or 3 to 7 ring members. In some embodiments, the cycloalkyl group is monocyclic, bicyclic or tricyclic. In some embodiments, the cycloalkyl group is monocyclic. In some embodiments, the cycloalkyl group is a C3-7 monocyclic cycloalkyl group. One or more ring-forming carbon atoms of a cycloalkyl group can be oxidized to form carbonyl linkages. Example cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclopentenyl,
cyclohexenyl, cyclohexadienyl, cycloheptatrienyl, norbornyl, norpinyl, norcarnyl, adamantyl, tetrahydronaphthalenyl, octahydronaphthalenyl, indanyl, and the like. As used herein, the term "Cn_m cycloalkyl-C0_p alkyl", employed alone or in combination with other terms, refers to a group of formula -alkylene-cycloalkyl, wherein the cycloalkyl portion has n to m carbon atoms and the alkylene portion has o to p carbon atoms. In some embodiments, the alkylene portion has 1 to 4, 1 to 3, 1 to 2, or 1 carbon atom(s). In some embodiments, the alkylene portion is methylene. In some
embodiments, the cycloalkyl portion has 3 to 14 ring members, 3 to 10 ring members, or 3 to 7 ring members. In some embodiments, the cycloalkyl group is monocyclic or bicyclic. In some embodiments, the cycloalkyl portion is monocyclic. In some embodiments, the cycloalkyl portion is a C3_7 monocyclic cycloalkyl group.
As used herein, the term "Cn_m heterocycloalkyl", "Cn_m heterocycloalkyl ring", or
"Cn_m heterocycloalkyl group", employed alone or in combination with other terms, refers to non-aromatic ring or ring system, which may optionally contain one or more alkenylene or alkynylene groups as part of the ring structure, which has at least one heteroatom ring member independently selected from nitrogen, sulfur oxygen and phosphorus, and which has n to m ring member carbon atoms. Heterocycloalkyl groups can include mono- or polycyclic (e.g., having 2, 3 or 4 fused, bridged, or spiro rings) ring systems. In some embodiments, the heterocycloalkyl group is a monocyclic or bicyclic group having 1, 2, 3, or 4 hetereoatoms independently selected from nitrogen, sulfur and oxygen. Also included in the definition of heterocycloalkyl are moieties that have one or more aromatic rings (e.g., aryl or heteroaryl rings) fused (i.e., having a bond in common with) to the non-aromatic ring, for example, 1,2,3,4-tetrahydro-quinoline and the like. Heterocycloalkyl groups can also include bridgehead heterocycloalkyl groups and spiroheterocycloalkyl groups. As used herein, "bridgehead heterocycloalkyl group" refers to a heterocycloalkyl moiety containing at least one bridgehead atom, such as
azaadmantan-l-yl and the like. As used herein, "spiroheterocycloalkyl group" refers to a heterocycloalkyl moiety containing at least two rings fused at a single atom, such as [1,4- dioxa-8-aza-spiro[4.5]decan-N-yl] and the like. In some embodiments, the
heterocycloalkyl group has 3 to 20 ring-forming atoms, 3 to 14 ring-forming atoms, 3 to 10 ring-forming atoms, or about 3 to 8 ring forming atoms. In some embodiments, the heterocycloalkyl group has 2 to 20 carbon atoms, 2 to 15 carbon atoms, 2 to 10 carbon atoms, or about 2 to 8 carbon atoms. In some embodiments, the heterocycloalkyl group has 1 to 5 heteroatoms, 1 to 4 heteroatoms, 1 to 3 heteroatoms, or 1 to 2 heteroatoms. The carbon atoms or hetereoatoms in the ring(s) of the heterocycloalkyl group can be oxidized to form a carbonyl, an N-oxide, or a sulfonyl group (or other oxidized linkage) or a nitrogen atom can be quaternized. In some embodiments, the heterocycloalkyl portion is a C2-7 monocyclic heterocycloalkyl group. Examples of heterocycloalkyl groups include 1, 2,3, 4-tetrahydro-quino line, azetidine, azepane, pyrrolidine, piperidine, piperazine, morpholine, thiomorpholine, and pyran.
As used herein, the term "Cn_m heterocycloalkyl-C0_p alkyl", employed alone or in combination with other terms, refers to a group of formula -alkylene-heterocycloalkyl, wherein the heterocycloalkyl portion has n to m carbon atoms and the alkylene portion has o to p carbon atoms. In some embodiments, the alkylene portion has 1 to 4, 1 to 3, 1 to 2, or 1 carbon atom(s). In some embodiments, the alkylene portion is methylene. In some embodiments, the heterocycloalkyl portion has 3 to 14 ring members, 3 to 10 ring members, or 3 to 7 ring members. In some embodiments, the heterocycloalkyl group is monocyclic or bicyclic. In some embodiments, the heterocycloalkyl portion is monocyclic. In some embodiments, the heterocycloalkyl portion is a C2-7 monocyclic heterocycloalkyl group.
As used herein, the term "Cn_m aryl", employed alone or in combination with other terms, refers to a monocyclic or polycyclic (e.g., having 2, 3 or 4 fused rings) aromatic hydrocarbon moiety having n to m ring member carbon atoms, such as, but not limited to, phenyl, 1-naphthyl, 2-naphthyl, anthracenyl, phenanthrenyl, and the like. In some embodiments, aryl groups have from 6 to 20 carbon atoms, from 6 to 14 carbon atoms, from 6 to 10 carbon atoms, or 6 carbon atoms. In some embodiments, the aryl group is a monocyclic or bicyclic group.
As used herein, the term "Cn_m aryl-C0_p-alkyl", employed alone or in combination with other terms, refers to a group of formula -alkylene-aryl, wherein the aryl portion has n to m ring member carbon atoms and the alkylene portion has o to p carbon atoms. In some embodiments, the alkylene portion has 1 to 4, 1 to 3, 1 to 2, or 1 carbon atom(s). In some embodiments, the alkylene portion is methylene. In some embodiments, the aryl portion is phenyl. In some embodiments, the aryl group is a monocyclic or bicyclic group. In some embodiments, the arylalkyl group is benzyl. As used herein, the term "Cn_m heteroaryl", "Cn_m heteroaryl ring", or "Cn_m heteroaryl group", employed alone or in combination with other terms, refers to a monocyclic or polycyclic (e.g., having 2, 3 or 4 fused rings) aromatic hydrocarbon moiety, having one or more heteroatom ring members independently selected from nitrogen, sulfur and oxygen and having n to m ring member carbon atoms. In some embodiments, the heteroaryl group is a monocyclic or bicyclic group having 1, 2, 3, or 4 hetereoatoms independently selected from nitrogen, sulfur and oxygen. Example heteroaryl groups include, but are not limited to, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazinyl, furyl, thienyl, imidazolyl, thiazolyl, indolyl, pyrryl, oxazolyl, benzthiazolyl, isoxazolyl, pyrazolyl, triazolyl, tetrazolyl, indazolyl, 1,2,4-thiadiazolyl, isothiazolyl, benzothienyl, purinyl, carbazolyl, benzimidazolyl, pyrrolyl, azolyl, oxazolyl, quinolinyl, isoquinolinyl, indolyl, benzothienyl, benzofuranyl, benzisoxazolyl, imidazo[l,2-b]thiazolyl or the like. The carbon atoms or hetereoatoms in the ring(s) of the heteroaryl group can be oxidized to form a carbonyl, an N-oxide, or a sulfonyl group (or other oxidized linkage) or a nitrogen atom can be quaternized, provided the aromatic nature of the ring is preserved. In some embodiments, the heteroaryl group has from 1 to 20 carbon atoms, from 3 to 20 carbon atoms, from 3 to 15 carbon atoms, from 3 to 10 carbon atoms, from 3 to 8 carbon atoms, from 3 to 5 carbon atoms, from 1 to 5 carbon atoms, or from 5 to 10 carbon atoms. In some embodiments, the heteroaryl group contains 3 to 14, 4 to 12, 4 to 8, 9 to 10, or 5 to 6 ring-forming atoms. In some embodiments, the heteroaryl group has 1 to 4, 1 to 3, or 1 to 2 heteroatoms.
As used herein, the term "Cn_m heteroaryl-C0_p-alkyl", employed alone or in combination with other terms, refers to a group of formula -alkylene-heteroaryl, wherein the heteroaryl portion has n to m ring member carbon atoms and the alkylene portion has o to p carbon atoms. In some embodiments, the alkylene portion has 1 to 4, 1 to 3, 1 to 2, or 1 carbon atom(s). In some embodiments, the alkylene portion is methylene. In some embodiments, the heteroaryl portion is a monocyclic or bicyclic group having 1, 2, 3, or 4 hetereoatoms independently selected from nitrogen, sulfur and oxygen. In some embodiments, the heteroaryl portion has 5 to 10 carbon atoms.
As used herein, the term "Cn_m aryloxy" refers to a moiety of formula -O-aryl, wherein the aryl ring has n to m carbon atoms. As used herein, the appearance of the term "bicyclic" before the name of a moiety indicates that the moiety has two fused rings.
As used herein, the appearance of the term "monocyclic" before the name of a moiety indicates that the moiety has a single ring.
The compounds described herein can be asymmetric (e.g., having one or more stereocenters). All stereoisomers, such as enantiomers and diastereomers, are intended unless otherwise indicated. Compounds of the present invention that contain
asymmetrically substituted carbon atoms can be isolated in optically active or racemic forms. Methods on how to prepare optically active forms from optically inactive starting materials are known in the art, such as by resolution of racemic mixtures or by stereoselective synthesis. Many geometric isomers of olefins, C=N double bonds, and the like can also be present in the compounds described herein, and all such stable isomers are contemplated in the present invention. Cis and trans geometric isomers of the compounds of the present invention are described and may be isolated as a mixture of isomers or as separated isomeric forms.
Resolution of racemic mixtures of compounds can be carried out by any of numerous methods known in the art. An example method includes fractional
recrystallizaion using a chiral resolving acid which is an optically active, salt-forming organic acid. Suitable resolving agents for fractional recrystallization methods are, for example, optically active acids, such as the D and L forms of tartaric acid, diacetyltartaric acid, dibenzoyltartaric acid, mandelic acid, malic acid, lactic acid or the various optically active camphorsulfonic acids such as β-camphorsulfonic acid. Other resolving agents suitable for fractional crystallization methods include stereoisomerically pure forms of a- methylbenzylamine (e.g., S and R forms, or diastereomerically pure forms), 2- phenylglycinol, norephedrine, ephedrine, N-methylephedrine, cyclohexylethylamine, 1 ,2- diaminocyclohexane, and the like.
Resolution of racemic mixtures can also be carried out by elution on a column packed with an optically active resolving agent (e.g., dinitrobenzoylphenylglycine). Suitable elution solvent composition can be determined by one skilled in the art.
Compounds of the invention also include tautomeric forms. Tautomeric forms result from the swapping of a single bond with an adjacent double bond together with the concomitant migration of a proton. Tautomeric forms include prototropic tautomers which are isomeric protonation states having the same empirical formula and total charge. Example prototropic tautomers include ketone - enol pairs, amide - imidic acid pairs, lactam - lactim pairs, enamine - imine pairs, and annular forms where a proton can occupy two or more positions of a heterocyclic system, for example, 1H- and 3H- imidazole, 1H-, 2H- and 4H- 1,2,4-triazole, 1H- and 2H- isoindole, and 1H- and 2H- pyrazole. Tautomeric forms can be in equilibrium or sterically locked into one form by appropriate substitution.
Compounds of the invention can also include all isotopes of atoms occurring in the intermediates or final compounds. Isotopes include those atoms having the same atomic number but different mass numbers. For example, isotopes of hydrogen include tritium and deuterium. In some embodiments, 1, 2, or 3 CH2 groups in the azetidine ring of Formula I are replaced by a CHD or CD2 group. In some embodiments, 1, 2, or 3 CH2 or CH groups in the piperidine ring of Formula I are replaced by a CHD, CD2 or CD group, respectively. In some embodiments, 1, 2, 3, 4, or 5 CH2 or CH groups in the piperidine ring of Formula I are replaced by a CHD, CD2 or CD group, respectively.
The term, "compound," as used herein is meant to include all stereoisomers, geometric iosomers, tautomers, and isotopes of the structures depicted.
All compounds, and pharmaceutically acceptable salts thereof, can be found together with other substances such as water and solvents (e.g., hydrates and solvates) or can be isolated.
In some embodiments, the compounds of the invention, or salts thereof, are substantially isolated. By "substantially isolated" is meant that the compound is at least partially or substantially separated from the environment in which it was formed or detected. Partial separation can include, for example, a composition enriched in the compounds of the invention. Substantial separation can include compositions containing at least about 50%, at least about 60%>, at least about 70%>, at least about 80%>, at least about 90%), at least about 95%, at least about 97%, or at least about 99% by weight of the compounds of the invention, or salt thereof. Methods for isolating compounds and their salts are routine in the art. The phrase "pharmaceutically acceptable" is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
The expressions, "ambient temperature" and "room temperature," as used herein, are understood in the art, and refer generally to a temperature, e.g. a reaction temperature, that is about the temperature of the room in which the reaction is carried out, for example, a temperature from about 20 °C to about 30 °C.
The present invention also includes pharmaceutically acceptable salts of the compounds described herein. As used herein, "pharmaceutically acceptable salts" refers to derivatives of the disclosed compounds wherein the parent compound is modified by converting an existing acid or base moiety to its salt form. Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like. The pharmaceutically acceptable salts of the present invention include the non-toxic salts of the parent compound formed, for example, from non-toxic inorganic or organic acids. The pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, non-aqueous media like ether, ethyl acetate, alcohols (e.g., methanol, ethanol, iso-propanol, or butanol) or acetonitrile (ACN) are preferred. Lists of suitable salts are found in Remington's Pharmaceutical Sciences, 17th ed., Mack Publishing Company, Easton, Pa., 1985, p. 1418 and Journal of Pharmaceutical Science, 66, 2 (1977), each of which is incorporated herein by reference in its entirety. In some embodiments, the compounds described herein include the N-oxide forms. Synthesis
Compounds of the invention, including salts and N-oxides thereof, can be prepared using known organic synthesis techniques and can be synthesized according to any of numerous possible synthetic routes, such as those in the Schemes below. The reactions for preparing compounds of the invention can be carried out in suitable solvents which can be readily selected by one of skill in the art of organic synthesis. Suitable solvents can be substantially non-reactive with the starting materials (reactants), the intermediates, or products at the temperatures at which the reactions are carried out, e.g., temperatures which can range from the solvent's freezing temperature to the solvent's boiling temperature. A given reaction can be carried out in one solvent or a mixture of more than one solvent. Depending on the particular reaction step, suitable solvents for a particular reaction step can be selected by the skilled artisan.
Preparation of compounds of the invention can involve the protection and deprotection of various chemical groups. The need for protection and deprotection, and the selection of appropriate protecting groups, can be readily determined by one skilled in the art. The chemistry of protecting groups can be found, for example, in Wuts and Greene, Protective Groups in Organic Synthesis, 4th ed., John Wiley & Sons: New Jersey, (2007), which is incorporated herein by reference in its entirety.
Reactions can be monitored according to any suitable method known in the art. For example, product formation can be monitored by spectroscopic means, such as nuclear magnetic resonance spectroscopy (e.g., 1H or 13C), infrared spectroscopy, spectrophotometry (e.g., UV-visible), mass spectrometry, or by chromatographic methods such as high performance liquid chromatography (HPLC) or thin layer chromatography (TLC).
Compounds of Formula I can be prepared using methods outlined in Schemes 1-4. Intermediates of formula 1-5 can be synthesized according to the methods described in Scheme 1. The commercially available starting material pyrrolo[2,3-d]pyrimidine-4- halide or 5-substituted pyrrolo[2,3-b]pyridine-4-halide (1-1) can be converted to a SEM (2-(trimethylsilyl)ethoxymethyl) protected intermediate of formula 1-2 by treating with sodium hydride followed by 2-(trimethylsilyl)ethoxymethyl chloride. Suzuki coupling of 1-2 with a boronic acid of pyrazole, such as l-(l-ethoxyethyl)-4-(4,4,5,5-tetramethyl- l,3,2-dioxaborolan-2-yl)-lH-pyrazole (1-3), using a palladium reagent, such as tetrakis(triphenylphosphine)palladium(0), gives rise to the intermediate 1-4, which in situ can be converted to the desired product 1-5 after prolongation of the reaction.
Scheme 1
Figure imgf000068_0001
Figure imgf000068_0002
Intermediates of fomula 2-3 can be synthesized according the sequence depicted in Scheme 2. The SEM-protected intermediate 1-2 is subjected to a Suzuki coupling with a boronic acid of a protected pyrrole, such as l-(triisopropylsilyl)pyrrole-3-boronic acid (2-1), using a palladium reagent, such as tetrakis(triphenylphosphine)palladium(0), in the presence of a base. The coupling product of formula 2-2 can be converted to the desired product of formula 2-3 in situ by carrying out the reaction overnight in the same media.
Scheme 2
Figure imgf000069_0001
Intermediates of formula 3-7 can be prepared according to the procedures shown in Scheme 3. A Boc-protected azetidinone of formula 3-1 is subjected to a Wittig reaction with a phosphonate, such as diethyl cyanomethylphosphonate, in the presense of a base, such as sodium hydride, to form a cyano derivative of formula 3-2. Michael addition of intermediates of formula 1-5 or 2-3 to the derivative of formula 3-2 in the presense of base, such as DBU produces the addition product of formula 3-3. Following removal of the Boc group (e.g, by using an acid such as 4 N HC1 in dioxane), reductive amination of the resulting azetidine of formula 3-4 with a N-Boc protected piperidinone of formula 3-5 using a reducing agent, such as sodium triacetoxyborohydride, gives rise to a compound of formula 3-6. Removal of the Boc group in the compound of formula 3-6 (e.g., using an acid such as 4 N HC1 in dioaxne) affords the desired intermediates of formula 3-7. Scheme 3
Figure imgf000070_0001
Intermediates of formula 3-7 can be derivatized at the piperidine nitrogen to produce a series of compounds of Formula I as depicted in Scheme 4. Reaction of the compound of formula 3-7 with a sulfonyl chloride followed by treating first with TFA and and then with ethylenediamine to remove the SEM group yields sulfonamide derivatives of formula 4-1. Coupling of the compound of formula 3-7 with a carboxylic acid using a coupling agent such as BOP or with an acyl chloride, followed by removal of the SEM group provides amide compounds of formula 4-2. Reductive amination of the compound of formula 3-7 with an aldehyde using a reducing agent, such as sodium triacetoxyborohydride, followed by removal of the SEM group gives rise to the N-alkyl derivatives of formula 4-3. Reaction of the compound of formula 3-7 with an isocyanate, followed by removing the SEM group affords the urea compounds of formula 4-4.
Scheme 4
Figure imgf000071_0001
Methods
Compounds of the invention are JAK inhibitors, and the majority of the compounds of the invention, are JAKl selective inhibitors. A JAKl selective inhibitor is a compound that inhibits JAKl activity preferentially over other Janus kinases. For example, the compounds of the invention preferentially inhibit JAKl over one or more of JAK2, JAK3, and TYK2. In some embodiments, the compounds inhibit JAKl preferentially over JAK2 (e.g., have a JAK1/JAK2 IC50 ratio >1). In some embodiments, the compounds are about 10-fold more selective for JAKl over JAK2. In some embodiments, the compounds are about 3 -fold, about 5 -fold, about 10-fold, about 15- fold, or about 20-fold more selective for JAKl over JAK2 as calculated by measuring IC50 at 1 mM ATP (e.g., see Example A).
JAKl plays a central role in a number of cytokine and growth factor signaling pathways that, when dysregulated, can result in or contribute to disease states. For example, IL-6 levels are elevated in rheumatoid arthritis, a disease in which it has been suggested to have detrimental effects (Fonesca, J.E. et al, Autoimmunity Reviews, 8:538-42, 2009). Because IL-6 signals, at least in part, through JAKl, antagonizing IL-6 directly or indirectly through JAKl inhibition is expected to provide clinical benefit (Guschin, D., N., et al Embo J 14: 1421, 1995; Smolen, J. S., et al. Lancet 371 :987, 2008). Moreover, in some cancers JAKl is mutated resulting in constitutive undesirable tumor cell growth and survival (Mullighan CG, Proc Natl Acad Sci U S A.106:9414-8, 2009; Flex E., et al.J Exp Med. 205:751-8, 2008). In other autoimmune diseases and cancers elevated systemic levels of inflammatory cytokines that activate JAKl may also contribute to the disease and/or associated symptoms. Therefore, patients with such diseases may benefit from JAKl inhibition. Selective inhibitors of JAKl may be efficacious while avoiding unnecessary and potentially undesirable effects of inhibiting other JAK kinases.
Selective inhibitors of JAKl, relative to other JAK kinases, may have multiple therapeutic advantages over less selective inhibitors. With respect to selectivity against JAK2, a number of important cytokines and growth factors signal through JAK2 including, for example, erythropoietin (Epo) and thrombopoietin (Tpo) (Parganas E, et al. Cell. 93:385-95, 1998). Epo is a key growth factor for red blood cells production; hence a paucity of Epo-dependent signaling can result in reduced numbers of red blood cells and anemia (Kaushansky K, NEJM 354:2034-45, 2006). Tpo, another example of a JAK2-dependent growth factor, plays a central role in controlling the proliferation and maturation of megakaryocytes - the cells from which platelets are produced (Kaushansky K, NEJM 354:2034-45, 2006). As such, reduced Tpo signaling would decrease megakaryocyte numbers (megakaryocytopenia) and lower circulating platelet counts (thrombocytopenia). This can result in undesirable and/or uncontrollable bleeding.
Reduced inhibition of other JAKs, such as JAK3 and Tyk2, may also be desirable as humans lacking functional version of these kinases have been shown to suffer from numerous maladies such as severe-combined immunodeficiency or
hyperimmunoglobulin E syndrome (Minegishi, Y, et al. Immunity 25 :745-55, 2006; Macchi P, et al. Nature. 377:65-8, 1995). Therefore a JAK1 inhibitor with reduced affinity for other JAKs would have significant advantages over a less-selective inhibitor with respect to reduced side effects involving immune suppression, anemia and thrombocytopenia.
Another aspect of the present invention pertains to methods of treating a JAK- associated disease or disorder in an individual (e.g. , patient) by administering to the individual in need of such treatment a therapeutically effective amount or dose of a compound of the present invention or a pharmaceutical composition thereof. A JAK- associated disease can include any disease, disorder or condition that is directly or indirectly linked to expression or activity of the JAK, including overexpression and/or abnormal activity levels. A JAK-associated disease can also include any disease, disorder or condition that can be prevented, ameliorated, or cured by modulating JAK activity.
Examples of JAK-associated diseases include diseases involving the immune system including, for example, organ transplant rejection (e.g., allograft rejection and graft versus host disease).
Further examples of JAK-associated diseases include autoimmune diseases such as multiple sclerosis, rheumatoid arthritis, juvenile arthritis, psoriatic arthritis, type I diabetes, lupus, psoriasis, inflammatory bowel disease, ulcerative colitis, Crohn's disease, myasthenia gravis, immunoglobulin nephropathies, myocarditis, autoimmune thyroid disorders, chronic obstructive pulmonary disease (COPD), and the like. In some embodiments, the autoimmune disease is an autoimmune bullous skin disorder such as pemphigus vulgaris (PV) or bullous pemphigoid (BP).
Further examples of JAK-associated diseases include allergic conditions such as asthma, food allergies, eszematous dermatitis, contact dermatitis, atopic dermatitis (atropic eczema), and rhinitis. Further examples of JAK-associated diseases include viral diseases such as Epstein Barr Virus (EBV), Hepatitis B, Hepatitis C, HIV, HTLV 1 , Varicella-Zoster Virus (VZV) and Human Papilloma Virus (HPV).
Further examples of JAK-associated disease include diseases associated with cartilage turnover, for example, gouty arthritis, septic or infectious arthritis, reactive arthritis, reflex sympathetic dystrophy, algodystrophy, Tietze syndrome, costal athropathy, osteoarthritis deformans endemica, Mseleni disease, Handigodu disease, degeneration resulting from fibromyalgia, systemic lupus erythematosus, scleroderma, or ankylosing spondylitis.
Further examples of JAK-associated disease include congenital cartilage malformations, including hereditary chrondro lysis, chrondrodysplasias, and
pseudochrondrodysplasias (e.g., microtia, enotia, and metaphyseal chrondrodysplasia).
Further examples of JAK-associated diseases or conditions include skin disorders such as psoriasis (for example, psoriasis vulgaris), atopic dermatitis, skin rash, skin irritation, skin sensitization (e.g., contact dermatitis or allergic contact dermatitis). For example, certain substances including some pharmaceuticals when topically applied can cause skin sensitization. In some embodiments, co-administration or sequential administration of at least one JAK inhibitor of the invention together with the agent causing unwanted sensitization can be helpful in treating such unwanted sensitization or dermatitis. In some embodiments, the skin disorder is treated by topical administration of at least one JAK inhibitor of the invention.
In further embodiments, the JAK-associated disease is cancer including those characterized by solid tumors (e.g., prostate cancer, renal cancer, hepatic cancer, pancreatic cancer, gastric cancer, breast cancer, lung cancer, cancers of the head and neck, thyroid cancer, glioblastoma, Kaposi's sarcoma, Castleman's disease, uterine leiomyosarcoma, melanoma etc.), hematological cancers (e.g., lymphoma, leukemia such as acute lymphoblastic leukemia (ALL), acute myelogenous leukemia (AML) or multiple myeloma), and skin cancer such as cutaneous T-cell lymphoma (CTCL) and cutaneous B-cell lymphoma. Example CTCLs include Sezary syndrome and mycosis fungoides.
In some embodiments, the JAK inhibitors described herein, or in combination with other JAK inhibitors, such as those reported in U.S. Ser. No. 1 1/637,545, which is incorporated herein by reference in its entirety, can be used to treat inflammation- associated cancers. In some embodiments, the cancer is associated with inflammatory bowel disease. In some embodiments, the inflammatory bowel disease is ulcerative colitis. In some embodiments, the inflammatory bowel disease is Crohn's disease. In some embodiments, the inflammation-associated cancer is colitis-associated cancer. In some embodiments, the inflammation-associated cancer is colon cancer or colorectal cancer. In some embodiments, the cancer is gastric cancer, gastrointestinal carcinoid tumor, gastrointestinal stromal tumor (GIST), adenocarcinoma, small intestine cancer, or rectal cancer.
JAK-associated diseases can further include those characterized by expression of: JAK2 mutants such as those having at least one mutation in the pseudo-kinase domain (e.g., JAK2V617F); JAK2 mutants having at least one mutation outside of the pseudo- kinase domain; JAK1 mutants; JAK3 mutants; erythropoietin receptor (EPOR) mutants; or deregulated expression of CRLF2.
JAK-associated diseases can further include myeloproliferative disorders (MPDs) such as polycythemia vera (PV), essential thrombocythemia (ET), myelofibrosis with myeloid metaplasia (MMM), primary myelofibrosis (PMF), chronic myelogenous leukemia (CML), chronic myelomonocytic leukemia (CMML), hypereosinophilic syndrome (HES), systemic mast cell disease (SMCD), and the like. In some
embodiments, the myeloproliferative disorder is myelofibrosis (e.g., primary
myelofibrosis (PMF) or post polycythemia vera/essential thrombocythemia myelofibrosis (Post-PV/ET MF)). In some embodiments, the myeloproliferative disorder is post- essential thrombocythemia myelofibrosis (Post-ET). In some embodiments, the myeloproliferative disorder is post polycythemia vera myelofibrosis (Post-PV MF).
The present invention further provides methods of treating psoriasis or other skin disorders by administration of a topical formulation containing a compound of the invention.
In some embodiments, JAK inhibitors described herein can be used to treat pulmonary arterial hypertension.
The present invention further provides a method of treating dermatological side effects of other pharmaceuticals by administration of the compound of the invention. For example, numerous pharmaceutical agents result in unwanted allergic reactions which can manifest as acneiform rash or related dermatitis. Example pharmaceutical agents that have such undesirable side effects include anti-cancer drugs such as gefitinib, cetuximab, erlotinib, and the like. The compounds of the invention can be administered systemically or topically (e.g., localized to the vicinity of the dermatitis) in combination with (e.g., simultaneously or sequentially) the pharmaceutical agent having the undesirable dermatological side effect. In some embodiments, the compound of the invention can be administered topically together with one or more other pharmaceuticals, where the other pharmaceuticals when topically applied in the absence of a compound of the invention cause contact dermatitis, allergic contact sensitization, or similar skin disorder.
Accordingly, compositions of the invention include topical formulations containing the compound of the invention and a further pharmaceutical agent which can cause dermatitis, skin disorders, or related side effects.
Further JAK-associated diseases include inflammation and inflammatory diseases. Example inflammatory diseases include sarcoidosis, inflammatory diseases of the eye (e.g., iritis, uveitis, scleritis, conjunctivitis, or related disease), inflammatory diseases of the respiratory tract (e.g. , the upper respiratory tract including the nose and sinuses such as rhinitis or sinusitis or the lower respiratory tract including bronchitis, chronic obstructive pulmonary disease, and the like), inflammatory myopathy such as
myocarditis, and other inflammatory diseases. In some embodiments, the inflammation disease of the eye is blepharitis.
The JAK inhibitors described herein can further be used to treat ischemia reperfusion injuries or a disease or condition related to an inflammatory ischemic event such as stroke or cardiac arrest. The JAK inhibitors described herein can further be used to treat endotoxin-driven disease state (e.g., complications after bypass surgery or chronic endotoxin states contributing to chronic cardiac failure). The JAK inhibitors described herein can further be used to treat anorexia, cachexia, or fatigue such as that resulting from or associated with cancer. The JAK inhibitors described herein can further be used to treat restenosis, sclerodermitis, or fibrosis. The JAK inhibitors described herein can further be used to treat conditions associated with hypoxia or astrogliosis such as, for example, diabetic retinopathy, cancer, or neurodegeneration. See, e.g., Dudley, A.C. et al. Biochem. J. 2005, 390(Pt 2):427-36 and Sriram, K. et al. J. Biol. Chem. 2004, 279(19): 19936-47. Epub 2004 Mar 2, both of which are incorporated herein by reference in their entirety. The JAK inhibitors described herein can be used to treat Alzheimer's disease.
The JAK inhibitors described herein can further be used to treat other
inflammatory diseases such as systemic inflammatory response syndrome (SIRS) and septic shock.
The JAK inhibitors described herein can further be used to treat gout and increased prostate size due to, e.g., benign prostatic hypertrophy or benign prostatic hyperplasia.
Further JAK-associated diseases include bone resorption diseases such as osteoporosis, osteoarthritis. Bone resorption can also be associated with other conditions such as hormonal imbalance and/or hormonal therapy, autoimmune disease (e.g. osseous sarcoidosis), or cancer (e.g. myeloma). The reduction of the bone resorption due to the JAK inhibitors can be about 10%, about 20%, about 30%>, about 40%>, about 50%>, about 60%, about 70%, about 80%, or about 90%.
In some embodiments, JAK inhibitors described herein can further be used to treat a dry eye disorder. As used herein, "dry eye disorder" is intended to encompass the disease states summarized in a recent official report of the Dry Eye Workshop (DEWS), which defined dry eye as "a multifactorial disease of the tears and ocular surface that results in symptoms of discomfort, visual disturbance, and tear film instability with potential damage to the ocular surface. It is accompanied by increased osmolality of the tear film and inflammation of the ocular surface." Lemp, "The Definition and
Classification of Dry Eye Disease: Report of the Definition and Classification
Subcommittee of the International Dry Eye Workshop", The Ocular Surface, 5(2), 75-92 April 2007, which is incorporated herein by reference in its entirety. In some
embodiments, the dry eye disorder is selected from aqueous tear-deficient dry eye (ADDE) or evaporative dry eye disorder, or appropriate combinations thereof. In some embodiments, the dry eye disorder is Sjogren syndrome dry eye (SSDE). In some embodiments, the dry eye disorder is non-Sjogren syndrome dry eye (NSSDE).
In a further aspect, the present invention provides a method of treating conjunctivitis, uveitis (including chronic uveitis), chorioditis, retinitis, cyclitis, sclieritis, episcleritis, or iritis; treating inflammation or pain related to corneal transplant, LASIK (laser assisted in situ keratomileusis), photorefractive keratectomy, or LASEK (laser assisted sub-epithelial keratomileusis); inhibiting loss of visual acuity related to corneal transplant, LASIK, photorefractive keratectomy, or LASEK; or inhibiting transplant rejection in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of the compound of the invention, or a pharmaceutically acceptable salt thereof.
Additionally, the compounds of the invention, or in combination with other JAK inhibitors, such as those reported in U.S. Ser. No. 11/637,545, which is incorporated herein by reference in its entirety, can be used to treat respiratory dysfunction or failure associated wth viral infection, such as influenza and SARS.
In some embodiments, the present invention provides a compound of Formula I, pharmaceutically acceptable salt thereof, as described in any of the embodiments herein, for use in a method of treating any of the diseases or disorders described herein. In some embodiments, the present invention provides the use of a compound of Formula I as described in any of the embodiments herein, for the preparation of a medicament for use in a method of treating any of the diseases or disorders described herein.
In some embodiments, the present invention provides a compound of Formula I as described herein, or a pharmaceutically acceptable salt thereof, for use in a method of modulating JAK1. In some embodiments, the present invention also provides use of a compound of Formula I as described herein, or a pharmaceutically acceptable salt thereof, for the preparation of a medicament for use in a method of modulating JAK1.
As used herein, the term "contacting" refers to the bringing together of indicated moieties in an in vitro system or an in vivo system. For example, "contacting" a JAK with a compound of the invention includes the administration of a compound of the present invention to an individual or patient, such as a human, having a JAK, as well as, for example, introducing a compound of the invention into a sample containing a cellular or purified preparation containing the JAK.
As used herein, the term "individual" or "patient," used interchangeably, refers to any animal, including mammals, preferably mice, rats, other rodents, rabbits, dogs, cats, swine, cattle, sheep, horses, or primates, and most preferably humans. As used herein, the phrase "therapeutically effective amount" refers to the amount of active compound or pharmaceutical agent that elicits the biological or medicinal response that is being sought in a tissue, system, animal, individual or human by a researcher, veterinarian, medical doctor or other clinician. In some embodiments, the therapeutically effective amount is about 5 mg to about 1000 mg, or about 10 mg to about 500 mg.
As used herein, the term "treating" or "treatment" refers to one or more of (1) preventing the disease; for example, preventing a disease, condition or disorder in an individual who may be predisposed to the disease, condition or disorder but does not yet experience or display the pathology or symptomatology of the disease; (2) inhibiting the disease; for example, inhibiting a disease, condition or disorder in an individual who is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e., arresting further development of the pathology and/or symptomatology); and (3) ameliorating the disease; for example, ameliorating a disease, condition or disorder in an individual who is experiencing or displaying the pathology or
symptomatology of the disease, condition or disorder (i.e., reversing the pathology and/or symptomatology) such as decreasing the severity of disease.
Combination Therapies
One or more additional pharmaceutical agents such as, for example,
chemotherapeutics, anti-inflammatory agents, steroids, immunosuppressants, as well as Bcr-Abl, Flt-3, RAF and FAK kinase inhibitors such as, for example, those described in WO 2006/056399, which is incorporated herein by reference in its entirety, or other agents can be used in combination with the compounds described herein for treatment of JAK-associated diseases, disorders or conditions. The one or more additional
pharmaceutical agents can be administered to a patient simultaneously or sequentially.
Example chemotherapeutics include proteosome inhibitors {e.g., bortezomib), thalidomide, revlimid, and DNA-damaging agents such as melphalan, doxorubicin, cyclophosphamide, vincristine, etoposide, carmustine, and the like.
Example steroids include coriticosteroids such as dexamethasone or prednisone. Example Bcr-Abl inhibitors include the compounds, and pharmaceutically acceptable salts thereof, of the genera and species disclosed in U.S. Pat. No. 5,521,184, WO 04/005281, and U.S. Ser. No. 60/578,491, all of which are incorporated herein by reference in their entirety.
Example suitable Flt-3 inhibitors include compounds, and their pharmaceutically acceptable salts, as disclosed in WO 03/037347, WO 03/099771, and WO 04/046120, all of which are incorporated herein by reference in their entirety.
Example suitable RAF inhibitors include compounds, and their pharmaceutically acceptable salts, as disclosed in WO 00/09495 and WO 05/028444, both of which are incorporated herein by reference in their entirety.
Example suitable FAK inhibitors include compounds, and their pharmaceutically acceptable salts, as disclosed in WO 04/080980, WO 04/056786, WO 03/024967, WO 01/064655, WO 00/053595, and WO 01/014402, all of which are incorporated herein by reference in their entirety.
In some embodiments, one or more of the compounds of the invention can be used in combination with one or more other kinase inhibitors including imatinib, particularly for treating patients resistant to imatinib or other kinase inhibitors.
In some embodiments, one or more JAK inhibitors of the invention can be used in combination with a chemotherapeutic in the treatment of cancer, such as multiple myeloma, and may improve the treatment response as compared to the response to the chemotherapeutic agent alone, without exacerbation of its toxic effects. Examples of additional pharmaceutical agents used in the treatment of multiple myeloma, for example, can include, without limitation, melphalan, melphalan plus prednisone [MP],
doxorubicin, dexamethasone, and Velcade (bortezomib). Further additional agents used in the treatment of multiple myeloma include Bcr-Abl, Flt-3, RAF and FAK kinase inhibitors. Additive or synergistic effects are desirable outcomes of combining a JAK inhibitor of the present invention with an additional agent. Furthermore, resistance of multiple myeloma cells to agents such as dexamethasone may be reversible upon treatment with a JAK inhibitor of the present invention. The agents can be combined with the present compounds in a single or continuous dosage form, or the agents can be administered simultaneously or sequentially as separate dosage forms. In some embodiments, a corticosteroid such as dexamethasone is administered to a patient in combination with at least one JAK inhibitor where the dexamethasone is administered intermittently as opposed to continuously.
In some further embodiments, combinations of one or more JAK inhibitors of the invention with other therapeutic agents can be administered to a patient prior to, during, and/or after a bone marrow transplant or stem cell transplant.
In some embodiments, the additional therapeutic agent is fluocinolone acetonide (Retisert®), or rimexolone (AL-2178, Vexol, Alcon).
In some embodiments, the additional therapeutic agent is cyclosporine
(Restasis®).
In some embodiments, the additional therapeutic agent is a corticosteroid. In some embodiments, the corticosteroid is triamcinolone, dexamethasone, fluocinolone, cortisone, prednisolone, or flumetholone.
In some embodiments, the additional therapeutic agent is selected from
Dehydrex™ (Holies Labs), Civamide (Opko), sodium hyaluronate (Vismed,
Lantibio/TRB Chemedia), cyclosporine (ST-603, Sirion Therapeutics), ARG101(T) (testosterone, Argentis), AGR1012(P) (Argentis), ecabet sodium (Senju-Ista), gefarnate (Santen), 15-(s)-hydroxyeicosatetraenoic acid (15(S)-HETE), cevilemine, doxycycline (ALTY-0501, Alacrity), minocycline, iDestrin™ (NP50301, Nascent Pharmaceuticals), cyclosporine A (Nova22007, Novagali), oxytetracycline (Duramycin, MOLI1901,
Lantibio), CFlOl (2S,3S,4R,5R)-3,4-dihydroxy-5-[6-[(3-iodophenyl)methylamino]purin- 9-yl]-N-methyl-oxolane-2-carbamyl, Can-Fite Biopharma), voclosporin (LX212 or LX214, Lux Biosciences), ARG103 (Agentis), RX- 10045 (synthetic resolvin analog, Resolvyx), DYN15 (Dyanmis Therapeutics), rivoglitazone (DEOl l, Daiichi Sanko), TB4 (RegeneRx), OPH-01 (Ophtalmis Monaco), PCS 101 (Pericor Science), REV 1-31
(Evolutec), Lacritin (Senju), rebamipide (Otsuka-Novartis), OT-551 (Othera), PAI-2 (University of Pennsylvania and Temple University), pilocarpine, tacrolimus, pimecrolimus (AMS981, Novartis), loteprednol etabonate, rituximab, diquafosol tetrasodium (INS365, Inspire), KLS-0611 (Kissei Pharmaceuticals),
dehydroepiandrosterone, anakinra, efalizumab, mycophenolate sodium, etanercept (Embrel®), hydroxychloroquine, NGX267 (TorreyPines Therapeutics), actemra, gemcitabine, oxaliplatin, L-asparaginase, or thalidomide.
In some embodiments, the additional therapeutic agent is an anti-angiogenic agent, cholinergic agonist, TRP-1 receptor modulator, a calcium channel blocker, a mucin secretagogue, MUC 1 stimulant, a calcineurin inhibitor, a corticosteroid, a P2Y2 receptor agonist, a muscarinic receptor agonist, an mTOR inhibitor, another JAK inhibitor, Bcr-Abl kinase inhibitor, Flt-3 kinase inhibitor, RAF kinase inhibitor, and FAK kinase inhibitor such as, for example, those described in WO 2006/056399, which is incorporated herein by reference in its entirety. In some embodiments, the additional therapeutic agent is a tetracycline derivative (e.g., minocycline or doxycline). In some embodiments, the additional therapeutic agent binds to FKBP12.
In some embodiments, the additional therapeutic agent is an alkylating agent or DNA cross-linking agent; an anti-metabolite/demethylating agent (e.g., 5-flurouracil, capecitabine or azacitidine); an anti-hormone therapy (e.g., hormone receptor antagonists, SERMs, or aromotase inhibitor); a mitotic inhibitor (e.g. vincristine or paclitaxel); an topoisomerase (I or II) inhibitor (e.g. mitoxantrone and irinotecan); an apoptotic inducers (e.g. ABT-737); a nucleic acid therapy (e.g. antisense or RNAi); nuclear receptor ligands (e.g., agonists and/or antagonists: all-trans retinoic acid or bexarotene); epigenetic targeting agents such as histone deacetylase inhibitors (e.g. vorinostat), hypomethylating agents (e.g. decitabine); regulators of protein stability such as Hsp90 inhibitors, ubiquitin and/or ubiquitin like conjugating or deconjugating molecules; or an EGFR inhibitor (erlotinib).
In some embodiments, the additional therapeutic agent(s) are demulcent eye drops (also known as "artificial tears"), which include, but are not limited to, compositions containing polyvinylalcohol, hydroxypropyl methylcellulose, glycerin, polyethylene glycol (e.g. PEG400), or carboxymethyl cellulose. Artificial tears can help in the treatment of dry eye by compensating for reduced moistening and lubricating capacity of the tear film. In some embodiments, the additional therapeutic agent is a mucolytic drug, such as N-acetyl-cysteine, which can interact with the mucoproteins and, therefore, to decrease the viscosity of the tear film. In some embodiments, the additional therapeutic agent includes an antibiotic, antiviral, antifungal, anesthetic, anti-inflammatory agents including steroidal and nonsteroidal anti-inflammatories, and anti-allergic agents. Examples of suitable medicaments include aminoglycosides such as amikacin, gentamycin, tobramycin, streptomycin, netilmycin, and kanamycin; fluoroquinolones such as ciprofloxacin, norfloxacin, ofloxacin, trovafloxacin, lomefloxacin, levofloxacin, and enoxacin; naphthyridine;
sulfonamides; polymyxin; chloramphenicol; neomycin; paramomycin; colistimethate; bacitracin; vancomycin; tetracyclines; rifampin and its derivatives ("rifampins");
cycloserine; beta-lactams; cephalosporins; amphotericins; fluconazole; flucytosine; natamycin; miconazole; ketoconazole; corticosteroids; diclofenac; flurbiprofen;
ketorolac; suprofen; cromolyn; lodoxamide; levocabastin; naphazoline; antazoline;
pheniramine; or azalide antibiotic.
Pharmaceutical Formulations and Dosage Forms
When employed as pharmaceuticals, the compounds of the invention can be administered in the form of pharmaceutical compositions. These compositions can be prepared in a manner well known in the pharmaceutical art, and can be administered by a variety of routes, depending upon whether local or systemic treatment is desired and upon the area to be treated. Administration may be topical (including transdermal, epidermal, ophthalmic and to mucous membranes including intranasal, vaginal and rectal delivery), pulmonary {e.g., by inhalation or insufflation of powders or aerosols, including by nebulizer; intratracheal or intranasal), oral or parenteral. Parenteral administration includes intravenous, intraarterial, subcutaneous, intraperitoneal intramuscular or injection or infusion; or intracranial, e.g., intrathecal or intraventricular, administration. Parenteral administration can be in the form of a single bolus dose, or may be, for example, by a continuous perfusion pump. Pharmaceutical compositions and
formulations for topical administration may include transdermal patches, ointments, lotions, creams, gels, drops, suppositories, sprays, liquids and powders. Conventional pharmaceutical carriers, aqueous, powder or oily bases, thickeners and the like may be necessary or desirable. This invention also includes pharmaceutical compositions which contain, as the active ingredient, the compound of the invention or a pharmaceutically acceptable salt thereof, in combination with one or more pharmaceutically acceptable carriers
(excipients). In some embodiments, the composition is suitable for topical
administration. In making the compositions of the invention, the active ingredient is typically mixed with an excipient, diluted by an excipient or enclosed within such a carrier in the form of, for example, a capsule, sachet, paper, or other container. When the excipient serves as a diluent, it can be a solid, semi-solid, or liquid material, which acts as a vehicle, carrier or medium for the active ingredient. Thus, the compositions can be in the form of tablets, pills, powders, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, aerosols (as a solid or in a liquid medium), ointments containing, for example, up to 10% by weight of the active compound, soft and hard gelatin capsules, suppositories, sterile injectable solutions, and sterile packaged powders.
In preparing a formulation, the active compound can be milled to provide the appropriate particle size prior to combining with the other ingredients. If the active compound is substantially insoluble, it can be milled to a particle size of less than 200 mesh. If the active compound is substantially water soluble, the particle size can be adjusted by milling to provide a substantially uniform distribution in the formulation, e.g. about 40 mesh.
The compounds of the invention may be milled using known milling procedures such as wet milling to obtain a particle size appropriate for tablet formation and for other formulation types. Finely divided (nanoparticulate) preparations of the compounds of the invention can be prepared by processes known in the art, e.g., see International App. No. WO 2002/000196.
Some examples of suitable excipients include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, tragacanth, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, water, syrup, and methyl cellulose. The formulations can additionally include: lubricating agents such as talc, magnesium stearate, and mineral oil; wetting agents; emulsifying and suspending agents; preserving agents such as methyl- and propylhydroxy-benzoates; sweetening agents; and flavoring agents. The compositions of the invention can be formulated so as to provide quick, sustained or delayed release of the active ingredient after administration to the patient by employing procedures known in the art.
In some embodiments, the pharmaceutical composition comprises silicified microcrystallme cellulose (SMCC) and at least one compound described herein, or a pharmaceutically acceptable salt thereof. In some embodiments, the silicified
microcrystallme cellulose comprises about 98% microcrystallme cellulose and about 2% silicon dioxide w/w.
In some embodiments, the composition is a sustained release composition comprising at least one compound described herein, or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable carrier. In some embodiments, the composition comprises at least one compound described herein, or a pharmaceutically acceptable salt thereof, and at least one component selected from microcrystallme cellulose, lactose monohydrate, hydroxypropyl methylcellulose, and polyethylene oxide. In some embodiments, the composition comprises at least one compound described herein, or a pharmaceutically acceptable salt thereof, and microcrystallme cellulose, lactose monohydrate, and hydroxypropyl methylcellulose. In some embodiments, the composition comprises at least one compound described herein, or a pharmaceutically acceptable salt thereof, and microcrystallme cellulose, lactose monohydrate, and polyethylene oxide. In some embodiments, the composition further comprises magnesium stearate or silicon dioxide. In some embodiments, the microcrystallme cellulose is Avicel PHI 02™. In some embodiments, the lactose monohydrate is Fast-flo 316™. In some embodiments, the hydroxypropyl methylcellulose is hydroxypropyl methylcellulose 2208 K4M (e.g., Methocel K4 M Premier™) and/or hydroxypropyl methylcellulose 2208 K100LV (e.g., Methocel K00LV™). In some embodiments, the polyethylene oxide is polyethylene oxide WSR 1105 (e.g., Polyox WSR 1105™).
In some embodiments, a wet granulation process is used to produce the composition. In some embodiments, a dry granulation process is used to produce the composition.
The compositions can be formulated in a unit dosage form, each dosage containing from about 5 to about 1,000 mg (1 g), more usually about 100 mg to about 500 mg, of the active ingredient. In some embodiments, each dosage contains about 10 mg of the active ingredient. In some embodiments, each dosage contains about 50 mg of the active ingredient. In some embodiments, each dosage contains about 25 mg of the active ingredient. The term "unit dosage forms" refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient.
In some embodiments, the compositions of the invention contain from about 5 mg to about 50 mg of the active ingredient. One having ordinary skill in the art will appreciate that this embodies compounds or compositions containing about 5 mg to about 10 mg, about 10 mg to about 15 mg, about 15 mg to about 20 mg, about 20 mg to about 25 mg, about 25 mg to about 30 mg, about 30 mg to about 35 mg, about 35 mg to about 40 mg, about 40 mg to about 45 mg, or about 45 mg to about 50 mg of the active ingredient.
In some embodiments, the compositions of the invention contain from about 50 mg to about 500 mg of the active ingredient. One having ordinary skill in the art will appreciate that this embodies compounds or compositions containing about 50 mg to about 100 mg, about 100 mg to about 150 mg, about 150 mg to about 200 mg, about 200 mg to about 250 mg, about 250 mg to about 300 mg, about 350 mg to about 400 mg, or about 450 mg to about 500 mg of the active ingredient.
In some embodiments, the compositions of the invention contain from about 500 mg to about 1 ,000 mg of the active ingredient. One having ordinary skill in the art will appreciate that this embodies compounds or compositions containing about 500 mg to about 550 mg, about 550 mg to about 600 mg, about 600 mg to about 650 mg, about 650 mg to about 700 mg, about 700 mg to about 750 mg, about 750 mg to about 800 mg, about 800 mg to about 850 mg, about 850 mg to about 900 mg, about 900 mg to about 950 mg, or about 950 mg to about 1,000 mg of the active ingredient.
The active compound may be effective over a wide dosage range and is generally administered in a pharmaceutically effective amount. It will be understood, however, that the amount of the compound actually administered will usually be determined by a physician, according to the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound administered, the age, weight, and response of the individual patient, the severity of the patient's symptoms, and the like.
For preparing solid compositions such as tablets, the principal active ingredient is mixed with a pharmaceutical excipient to form a solid preformulation composition containing a homogeneous mixture of a compound of the present invention. When referring to these preformulation compositions as homogeneous, the active ingredient is typically dispersed evenly throughout the composition so that the composition can be readily subdivided into equally effective unit dosage forms such as tablets, pills and capsules. This solid preformulation is then subdivided into unit dosage forms of the type described above containing from, for example, about 0.1 to about 1000 mg of the active ingredient of the present invention.
The tablets or pills of the present invention can be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action. For example, the tablet or pill can comprise an inner dosage and an outer dosage component, the latter being in the form of an envelope over the former. The two components can be separated by an enteric layer which serves to resist disintegration in the stomach and permit the inner component to pass intact into the duodenum or to be delayed in release. A variety of materials can be used for such enteric layers or coatings, such materials including a number of polymeric acids and mixtures of polymeric acids with such materials as shellac, cetyl alcohol, and cellulose acetate.
The liquid forms in which the compounds and compositions of the present invention can be incorporated for administration orally or by injection include aqueous solutions, suitably flavored syrups, aqueous or oil suspensions, and flavored emulsions with edible oils such as cottonseed oil, sesame oil, coconut oil, or peanut oil, as well as elixirs and similar pharmaceutical vehicles.
Compositions for inhalation or insufflation include solutions and suspensions in pharmaceutically acceptable, aqueous or organic solvents, or mixtures thereof, and powders. The liquid or solid compositions may contain suitable pharmaceutically acceptable excipients as described supra. In some embodiments, the compositions are administered by the oral or nasal respiratory route for local or systemic effect.
Compositions in can be nebulized by use of inert gases. Nebulized solutions may be breathed directly from the nebulizing device or the nebulizing device can be attached to a face masks tent, or intermittent positive pressure breathing machine. Solution, suspension, or powder compositions can be administered orally or nasally from devices which deliver the formulation in an appropriate manner.
Topical formulations can contain one or more conventional carriers. In some embodiments, ointments can contain water and one or more hydrophobic carriers selected from, for example, liquid paraffin, polyoxyethylene alkyl ether, propylene glycol, white Vaseline, and the like. Carrier compositions of creams can be based on water in combination with glycerol and one or more other components, e.g.
glycerinemonostearate, PEG-glycerinemonostearate and cetylstearyl alcohol. Gels can be formulated using isopropyl alcohol and water, suitably in combination with other components such as, for example, glycerol, hydroxyethyl cellulose, and the like. In some embodiments, topical formulations contain at least about 0.1, at least about 0.25, at least about 0.5, at least about 1 , at least about 2, or at least about 5 wt % of the compound of the invention. The topical formulations can be suitably packaged in tubes of, for example, 100 g which are optionally associated with instructions for the treatment of the select indication, e.g., psoriasis or other skin condition.
The amount of compound or composition administered to a patient will vary depending upon what is being administered, the purpose of the administration, such as prophylaxis or therapy, the state of the patient, the manner of administration, and the like. In therapeutic applications, compositions can be administered to a patient already suffering from a disease in an amount sufficient to cure or at least partially arrest the symptoms of the disease and its complications. Effective doses will depend on the disease condition being treated as well as by the judgment of the attending clinician depending upon factors such as the severity of the disease, the age, weight and general condition of the patient, and the like.
The compositions administered to a patient can be in the form of pharmaceutical compositions described above. These compositions can be sterilized by conventional sterilization techniques, or may be sterile filtered. Aqueous solutions can be packaged for use as is, or lyophilized, the lyophilized preparation being combined with a sterile aqueous carrier prior to administration. The pH of the compound preparations typically will be between 3 and 11, more preferably from 5 to 9 and most preferably from 7 to 8. It will be understood that use of certain of the foregoing excipients, carriers, or stabilizers will result in the formation of pharmaceutical salts.
The therapeutic dosage of a compound of the present invention can vary according to, for example, the particular use for which the treatment is made, the manner of administration of the compound, the health and condition of the patient, and the judgment of the prescribing physician. The proportion or concentration of a compound of the invention in a pharmaceutical composition can vary depending upon a number of factors including dosage, chemical characteristics (e.g., hydrophobicity), and the route of administration. For example, the compounds of the invention can be provided in an aqueous physiological buffer solution containing about 0.1 to about 10% w/v of the compound for parenteral administration. Some typical dose ranges are from about 1 μg/kg to about 1 g/kg of body weight per day. In some embodiments, the dose range is from about 0.01 mg/kg to about 100 mg/kg of body weight per day. The dosage is likely to depend on such variables as the type and extent of progression of the disease or disorder, the overall health status of the particular patient, the relative biological efficacy of the compound selected, formulation of the excipient, and its route of administration. Effective doses can be extrapolated from dose-response curves derived from in vitro or animal model test systems.
The compositions of the invention can further include one or more additional pharmaceutical agents such as a chemotherapeutic, steroid, anti-inflammatory compound, or immunosuppressant, examples of which are listed hereinabove.
In some embodiments, the compound, or pharmaceutically acceptable salt thereof, is administered as an ophthalmic composition. Accordingly, in some embodiments, the methods comprise administration of the compound, or pharmaceutically acceptable salt thereof, and an ophthalmically acceptable carrier. In some embodiments, the ophthalmic composition is a liquid composition, semi-solid composition, insert, film, microparticles or nanoparticles.
In some embodiments, the ophthalmic composition is a liquid composition. In some embodiments, the ophthalmic composition is a semi-solid composition. In some embodiments, the ophthalmic composition is a topical composition. The topical compositions include, but are not limited to liquid and semi-solid compositions. In some embodiments, the ophthalmic composition is a topical composition. In some
embodiments, the topical composition comprises aqueous solution, an aqueous suspension, an ointment or a gel. In some embodiments, the ophthalmic composition is topically applied to the front of the eye, under the upper eyelid, on the lower eyelid and in the cul-de-sac. In some embodiments, the ophthalmic composition is sterilized. The sterilization can be accomplished by known techniques like sterilizing filtration of the solution or by heating of the solution in the ampoule ready for use. The ophthalmic compositions of the invention can further contain pharmaceutical excipients suitable for the preparation of ophthalmic formulations. Examples of such excipients are preserving agents, buffering agents, chelating agents, antioxidant agents and salts for regulating the osmotic pressure.
As used herein, the term "ophthalmically acceptable carrier" refers to any material that can contain and release the compound, or pharmaceutically acceptable salt thereof, and that is compatible with the eye. In some embodiments, the ophthalmically acceptable carrier is water or an aqueous solution or suspension, but also includes oils such as those used to make ointments and polymer matrices such as used in ocular inserts. In some embodiments, the composition may be an aqueous suspension comprising the compound, or pharmaceutically acceptable salt thereof. Liquid ophthalmic compositions, including both ointments and suspensions, may have a viscosity that is suited for the selected route of administration. In some embodiments, the ophthalmic composition has a viscosity in the range of from about 1,000 to about 30,000 centipoise.
In some embodiments, the ophthalmic compositions may further comprise one or more of surfactants, adjuvants, buffers, antioxidants, tonicity adjusters, preservatives (e.g., EDTA, BAK (benzalkonium chloride), sodium chlorite, sodium perborate, polyquaterium-1), thickeners or viscosity modifiers (e.g., carboxymethyl cellulose, hydroxymethyl cellulose, polyvinyl alcohol, polyethylene glycol, glycol 400, propylene glycol hydroxymethyl cellulose, hydroxpropyl-guar, hyaluronic acid, and hydroxypropyl cellulose) and the like. Additives in the formulation may include, but are not limited to, sodium chloride, sodium bicarbonate, sorbic acid, methyl paraben, propyl paraben, chlorhexidine, castor oil, and sodium perborate. Aqueous ophthalmic compositions (solutions or suspensions) generally do not contain physiologically or ophthalmically harmful constituents. In some embodiments, purified or deionized water is used in the composition. The pH may be adjusted by adding any physiologically and ophthalmically acceptable pH adjusting acids, bases or buffers to within the range of about 5.0 to 8.5. Ophthalmically acceptable examples of acids include acetic, boric, citric, lactic, phosphoric, hydrochloric, and the like, and examples of bases include sodium hydroxide, sodium phosphate, sodium borate, sodium citrate, sodium acetate, sodium lactate, tromethamine, trishydroxymethylamino-methane, and the like. Salts and buffers include citrate/dextrose, sodium bicarbonate, ammonium chloride and mixtures of the aforementioned acids and bases.
In some embodiments, the methods involve forming or supplying a depot of the therapeutic agent in contact with the external surface of the eye. A depot refers to a source of therapeutic agent that is not rapidly removed by tears or other eye clearance mechanisms. This allows for continued, sustained high concentrations of therapeutic agent to be present in the fluid on the external surface of the eye by a single application. Without wishing to be bound by any theory, it is believed that absorption and penetration may be dependent on both the dissolved drug concentration and the contact duration of the external tissue with the drug containing fluid. As the drug is removed by clearance of the ocular fluid and/or absorption into the eye tissue, more drug is provided, e.g.
dissolved, into the replenished ocular fluid from the depot. Accordingly, the use of a depot may more easily facilitate loading of the ocular tissue for more insoluble therapeutic agents. In some embodiments, the depot can remain for up to eight hours or more. In some embodiments, the ophthalmic depot forms includes, but is not limited to, aqueous polymeric suspensions, ointments, and solid inserts.
In some embodiments, the ophthalmic composition is an ointment or gel. In some embodiment, the ophthalmic composition is an oil-based delivery vehicle. In some embodiments, the composition comprises a petroleum or lanolin base to which is added the active ingredient, usually as 0.1 to 2%, and excipients. Common bases may include, but are not limited to, mineral oil, petrolatum and combinations thereof. In some embodiments, the ointment is applied as a ribbon onto the lower eyelid. In some embodiment, the ophthalmic composition is an ophthalmic insert. In some embodiments, the ophthalmic insert is biologically inert, soft, bio-erodible, viscoelastic, stable to sterilization after exposure to therapeutic agents, resistant to infections from air borne bacteria, bio- erodible, biocompatible, and/or viscoelastic. In some embodiments, the insert comprises an ophthalmically acceptable matrix, e.g., a polymer matrix. The matrix is typically a polymer and the therapeutic agent is generally dispersed therein or bonded to the polymer matrix. In some embodiments, the therapeutic agent may be slowly released from the matrix through dissolution or hydrolysis of the covalent bond. In some embodiments, the polymer is bioerodible (soluble) and the dissolution rate thereof can control the release rate of the therapeutic agent dispersed therein. In another form, the polymer matrix is a biodegradable polymer that breaks down such as by hydrolysis to thereby release the therapeutic agent bonded thereto or dispersed therein. In further embodiments, the matrix and therapeutic agent can be surrounded with an additional polymeric coating to further control release. In some embodiments, the insert comprises a biodegradable polymer such as polycaprolactone (PCL), an
ethylene/vinyl acetate copolymer (EVA), polyalkyl cyanoacrylate, polyurethane, a nylon, or poly (dl-lactide-co-glycolide) (PLGA), or a copolymer of any of these. In some embodiments, the therapeutic agent is dispersed into the matrix material or dispersed amongst the monomer composition used to make the matrix material prior to
polymerization. In some embodiments, the amount of therapeutic agent is from about 0.1 to about 50%, or from about 2 to about 20%. In further embodiments, the biodegradable or bioerodible polymer matrix is used so that the spent insert does not have to be removed. As the biodegradable or bioerodible polymer is degraded or dissolved, the therapeutic agent is released.
In further embodiments, the ophthalmic insert comprises a polymer, including, but are not limited to, those described in Wagh, et al, "Polymers used in ocular dosage form and drug delivery systems", Asian J. Pharm., pages 12-17 (Jan. 2008), which is incorporated herein by reference in its entirety. In some embodiments, the insert comprises a polymer selected from polyvinylpyrrolidone (PVP), an acrylate or methacrylate polymer or copolymer (e.g., Eudragit® family of polymers from Rohm or Degussa), hydroxymethyl cellulose, polyacrylic acid, poly(amidoamine) dendrimers, poly(dimethyl siloxane), polyethylene oxide, poly(lactide-co-glycolide), poly(2- hydroxyethylmethacrylate), poly( vinyl alcohol), or poly(propylene fumarate). In some embodiments, the insert comprises Gelfoam® R. In some embodiments, the insert is a polyacrylic acid of 450 kDa-cysteine conjugate.
In some embodiments, the ophthalmic composition is a ophthalmic film.
Polymers suitable for such films include, but are not limited to, those described in Wagh, et al. (ibid), In some embodiments, the film is a soft-contact lens, such as ones made from copolymers of Ν,Ν-diethylacrylamide and methacrylic acid crosslinked with ethyleneglycol dimethacrylate.
In some embodiments, the ophthalmic compositon comprises microspheres or nanoparticles. In some embodiment, the microspheres comprise gelatin. In some embodiments, the microspheres are injected to the posterior segment of the eye, in the chroroidal space, in the sclera, intravitreally or sub-retinally. In some embodiments, the microspheres or nanoparticles comprises a polymer including, but not limited to, those described in Wagh, et al. (ibid), which is incorporated herein by reference in its entirety. In some embodiments, the polymer is chitosan, a polycarboxylic acid such as polyacrylic acid, albumin particles, hyaluronic acid esters, polyitaconic acid,
poly(butyl)cyanoacrylate, polycaprolactone, poly(isobutyl)caprolactone, poly(lactic acid- co-glycolic acid), or poly(lactic acid). In some embodiments, the microspheres or nanoparticles comprise solid lipid particles.
In some embodiments, the ophthalmic composition comprises an ion-exchange resin. In some embodiments, the ion-exchange resin is an inorganic zeolite or synthetic organic resin. In some embodiments, the ion-exchange resin includes, but is not limited to, those described in Wagh, et al. (ibid), which is incorporated herein by reference in its entirety. In some embodiments, the ion-exhange resin is a partially neutralized polyacrylic acid.
In some embodiments, the ophthalmic composition is an aqueous polymeric suspension. In some embodiments, the therapeutic agent or a polymeric suspending agent is suspended in an aqueous medium. In some embodiments, the aqueous polymeric suspensions may be formulated so that they retain the same or substantially the same viscosity in the eye that they had prior to administration to the eye. In some embodiments, they may be formulated so that there is increased gelation upon contact with tear fluid.
Labeled Compounds and Assay Methods
Another aspect of the present invention relates to labeled compounds of the invention (radio-labeled, fluorescent-labeled, etc.) that would be useful not only in imaging techniques but also in assays, both in vitro and in vivo, for localizing and quantitating JAK in tissue samples, including human, and for identifying JAK ligands by inhibition binding of a labeled compound. Accordingly, the present invention includes JAK assays that contain such labeled compounds.
The present invention further includes isotopically-labeled compounds of the invention. An "isotopically" or "radio-labeled" compound is a compound of the invention where one or more atoms are replaced or substituted by an atom having an atomic mass or mass number different from the atomic mass or mass number typically found in nature (i.e., naturally occurring). Suitable radionuclides that may be
incorporated in compounds of the present invention include but are not limited to 3H (also written as T for tritium), UC, 13C, 14C, 13N, 15N, 150, 170, 180, 18F, 35S, 36C1, 82Br, 75Br,
76 11 123 124 125 131
Br, Br, I, I, I and I. The radionuclide that is incorporated in the instant radio-labeled compounds will depend on the specific application of that radio-labeled compound. For example, for in vitro JAK labeling and competition assays, compounds
3 14 82 125 131 35
that incorporate H, C, Br, I , I, S or will generally be most useful. For radio- imaging applications nC, 18F, 125I, 123I, 124I, 131I, 75Br, 76Br or 77Br will generally be most useful.
It is to be understood that a "radio-labeled " or "labeled compound" is a compound that has incorporated at least one radionuclide. In some embodiments the
3 14 125 35 82
radionuclide is selected from the group consisting of H, C, I , S and Br. In some embodiments, the compound incorporates 1, 2, or 3 deuterium atoms.
The present invention can further include synthetic methods for incorporating radio-isotopes into compounds of the invention. Synthetic methods for incorporating radio-isotopes into organic compounds are well known in the art, and an ordinary skill in the art will readily recognize the methods applicable for the compounds of invention. A labeled compound of the invention can be used in a screening assay to identify/evaluate compounds. For example, a newly synthesized or identified compound (i.e., test compound) which is labeled can be evaluated for its ability to bind a JAK by monitoring its concentration variation when contacting with the JAK, through tracking of the labeling. For example, a test compound (labeled) can be evaluated for its ability to reduce binding of another compound which is known to bind to a JAK (i.e., standard compound). Accordingly, the ability of a test compound to compete with the standard compound for binding to the JAK directly correlates to its binding affinity. Conversely, in some other screening assays, the standard compound is labeled and test compounds are unlabeled. Accordingly, the concentration of the labeled standard compound is monitored in order to evaluate the competition between the standard compound and the test compound, and the relative binding affinity of the test compound is thus ascertained.
Kits
The present invention also includes pharmaceutical kits useful, for example, in the treatment or prevention of JAK-associated diseases or disorders, such as cancer, which include one or more containers containing a pharmaceutical composition comprising a therapeutically effective amount of a compound of the invention. Such kits can further include, if desired, one or more of various conventional pharmaceutical kit components, such as, for example, containers with one or more pharmaceutically acceptable carriers, additional containers, etc., as will be readily apparent to those skilled in the art.
Instructions, either as inserts or as labels, indicating quantities of the components to be administered, guidelines for administration, and/or guidelines for mixing the components, can also be included in the kit.
The invention will be described in greater detail by way of specific examples. The following examples are offered for illustrative purposes, and are not intended to limit the invention in any manner. Those of skill in the art will readily recognize a variety of non- critical parameters which can be changed or modified to yield essentially the same results. The compounds of the Examples have been found to be JAK inhibitors according to at least one assay described herein. EXAMPLES
The example compounds below containing one or more chiral centers were obtained in enantiomerically pure form or as scalemic mixtures, unless otherwise specified.
Unless otherwise indicated, the example compounds were purified by preparative
HPLC using acidic conditions (method A) and were obtained as a TFA salt or using basic conditions (method B) and were obtained as a free base.
Method A:
Column: Waters Sun Fire CI 8, 5 μιη particle size, 30 x 100 mm;
Mobile phase: water (0.1% TFA)/acetonitrile
Flow rate: 60 mL/min
Gradient: 5 min or 12 min from 5% acetonitrile/95% water to 100% acetonitrile Method B:
Column: Waters X Bridge CI 8, 5 μιη particle size, 30 x 100 mm;
Mobile phase: water (0.15% NH4OH)/acetonitrile
Method C:
Column: C18 column, 5 μιη OBD
Mobile phase: water + 0.05% NH4OH (A), CH3CN + 0.05% NH4OH (B)
Gradient: 5% B to 100% B in 15 min
Flow rate: 60 mL/min Example 1. {l-{l-[3-Fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4- (7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile
Figure imgf000097_0001
Step A: tert-Butyl 3-Oxoazetidine-l-carboxylate
,Boc Swern Oxidation ,Boc
Figure imgf000097_0002
To a mixture of tert-butyl 3-hydroxyazetidine-l-carboxylate (10.0 g, 57.7 mmol), dimethyl sulfoxide (24.0 mL, 338 mmol), triethylamine (40 mL, 300 mmol) and methylene chloride (2.0 mL) was added sulfur trioxide-pyridine complex (40 g, 200 mmol) portionwise at 0 °C. The mixture was stirred for 3 hours, quenched with brine, and extracted with methylene chloride. The combined extracts were dried over anhydrous Na2S04, filtered, and concentrated under reduced pressure. The residue was purified by silica gel column (0-6% ethyl acetate (EtOAc) in hexanes) to give fert-butyl 3- oxoazetidine-l-carboxylate (5.1 g, 52% yield).
Step B: tert-Butyl 3-(Cyanomethylene)azetidine-l-carboxylate
Figure imgf000097_0003
An oven-dried 1 L 4-neck round bottom flask fitted with stir bar, septa, nitrogen inlet, 250 ml addition funnel and thermocouple was charged with sodium hydride (5.6 g, 0.14 mol) and tetrahydrofuran (THF) (140 mL) under a nitrogen atmosphere. The mixture was chilled to 3 °C, and then charged with diethyl cyanomethylphosphonate (22.4 mL,
0.138 mol) drop wise via a syringe over 20 minutes. The solution became a light yellow slurry. The reaction was then stirred for 75 minutes while warming to 18.2 °C. A solution of tert-butyl 3-oxoazetidine-l-carboxylate (20 g, 0.1 mol) in tetrahydrofuran (280 mL) was prepared in an oven-dried round bottom, charged to the addition funnel via canula, then added to the reaction mixture dropwise over 25 minutes. The reaction solution became red in color. The reaction was allowed to stir overnight. The reaction was checked after 24 hours by TLC (70% hexane/EtOAc) and found to be complete. The reaction was diluted with 200 mL of 20% brine and 250 mL of EtOAc. The solution was partitioned and the aqueous phase was extracted with 250 mL of EtOAc.The combined organic phase was dried over MgSC^ and filtered, evaporated under reduced pressure, and purified by flash chromatography ( 0% to 20% EtOAc/hexanes, 150 g flash column) to give the desired product, tert-butyl 3-(cyanomethylene)azetidine-l-carboxylate (15 g, 66.1% yield).
Step C: 4-Chloro-7- 2-(trimethylsilyl)ethoxyJmethyl}-7H^yrrolo[2 -dJpyrimidine
Figure imgf000098_0001
To a suspension of sodium hydride (36.141 g, 903.62 mmol) in N,N- dimethylacetamide (118 mL) at -5 °C (ice/salt bath) was added a dark solution of 4- chloropyrrolo[2,3-d]pyrimidine (119.37 g, 777.30 mmol) in N,N-dimethylacetamide (237 mL) slowly. The flask and addition funnel were rinsed with N,N-dimethylacetamide (30 mL). A large amount of gas was evolved immediately. The mixture became a slightly cloudy orange mixture. The mixture was stirred at 0 °C for 60 min to give a light brown turbid mixture. To the mixture was slowly added [2-(trimethylsilyl)ethoxy]methyl chloride (152.40 g, 914.11 mmol) and the reaction was stirred at 0 °C for 1 h. The reaction was quenched by addition of 12 mL of H20 slowly. More water (120 mL) was added followed by methyl tert-butyl ether (MTBE) (120 mL). The mixture was stirred for 10 min. The organic layer was separated. The aqueous layer was extracted with another portion of MTBE (120 mL). The organic extracts were combined, washed with brine (120 mL x 2) and concentrated under reduced pressure to give the crude product 4- chloro-7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidine as a dark oil. Yield: 85.07 g (97%); LC-MS: 284.1 (M+H)+. It was carried to the next reaction without purification. Step D: 4-(lH-Pyrazol-4-yl)-7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3- djpyrimidine
Figure imgf000099_0001
A 1000 mL round bottom flask was charged with 4-chloro-7-{[2- (trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidine (10.00 g, 35.23 mmol), 1- butanol (25.0 mL), l-(l-ethoxyethyl)-4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)- lH-pyrazole (15.66 g, 52.85 mmol), water (25.0 mL) and potassium carbonate (12.17 g, 88.08 mmol). This solution was degased 4 times, filling with nitrogen each time. To the solution was added tetrakis(triphenylphosphine)palladium(0) (4.071 g, 3.523 mmol). The solution was degased 4 times, filling with nitrogen each time. The mixture was stirred overnight at 100 °C. After being cooled to room temperature, the mixture was filtered through a bed of celite and the celite was rinsed with ethyl acetate (42 mL). The filtrate was combined, and the organic layer was separated. The aqueous layer was extracted with ethyl acetate. The organic extracts were combined and concentrated under vacuum with a bath temerature of 30-70 °C to give the final compound 4-(lH-pyrazol-4-yl)-7-{[2- (trimethylsilyl)ethoxy]methyl} -7H-pyrrolo[2,3-d]pyrimidine. Yield: 78%. LC-MS : 316.2 (M+H)+. Step E: tert-Butyl 3-(Cyanomethyl)-3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidine-l-carboxylate
Figure imgf000100_0001
A 2 L round bottom flask fitted with overhead stirring, septa and nitrogen inlet was charged with fert-butyl 3-(cyanomethylene)azetidine-l-carboxylate (9.17 g, 0.0472 mol), 4-(lH-pyrazol-4-yl)-7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3- d]pyrimidine (14.9 g, 0.0472 mol) and acetonitrile (300 mL). The resulting solution was heterogeneous. To the solution was added l,8-diazabicyclo[5.4.0]undec-7-ene (8.48 mL, 0.0567 mol) portionwise via syringe over 3 min at room temperature. The solution slowly became homogeneous and yellow in color. The reaction was allowed to stir at room temperature for 3 h. The reaction was complete by HPLC and LC/MS and was concentrated by rotary evaporation to remove acetonitrile (-150 mL). EtOAc (100 mL) was added followed by 100 ml of 20% brine. The two phases were partitioned. The aqueous phase was extracted with 150 mL of EtOAC. The combine organic phases were dried over MgSC^, filtered and concentrated to yield an orange oil. Purification by flash chromatography (150 grams silica, 60%> EtOAc/hexanes, loaded with CH2CI2) yielded the title compound tert-butyl 3-(cyanomethyl)-3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}- 7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidine-l-carboxylate as a yellow oil (21.1 g, 88% yield). LC-MS: [M+H]+ = 510.3.
Step F: { 3-f 4- (7 -{[2- (Trimethylsilyl) ethoxy] methyl} -7 H-pyrrolo[ 2, 3-d] pyrimidin-4-yl)- lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile dihydrochloride
Figure imgf000101_0001
To a solution of tert-butyl 3-(cyanomethyl)-3-[4-(7-{[2- (trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidine-l-carboxylate (2 g, 3.9 mmol) in 10 mL of THF was added 10 mL of 4 N HCl in dioxane. The solution was stirred at room temperature for 1 hour and concentrated in vacuo to provide 1.9 g (99%) of the title compound as a white powder solid, which was used for the next reaction without purification. LC-MS: [M+H]+ = 410.3.
Step G: tert-Butyl 4-{3-(Cyanomethyl)-3-[4-(7-{[2-(trimethylsilyl) ethoxy] methyl} -7 H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-l-yl}piperidine-l-carb
Figure imgf000101_0002
Into the solution of {3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile dihydrochloride (2.6 g, 6.3 mmol), tert-butyl 4-oxo-l-piperidinecarboxylate (1.3 g, 6.3 mmol) in THF (30 mL) were added N,N-diisopropylethylamine (4.4 mL, 25 mmol) and sodium triacetoxyborohydride (2.2 g, 10 mmol). The mixture was stirred at room temperature overnight. After adding 20 mL of brine, the solution was extracted with EtOAc. The extract was dried over anhydrous Na2S04 and concentrated. The residue was purified by combiflash column eluting with 30-80 % EtOAc in hexanes to give the desired product, tert-butyl 4-{3- (cyanomethyl)-3-[4-(7-{[2-(trimethylsilyl) ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidin- 4-yl)-lH-pyrazol-l-yl]azetidin-l-yl}piperidine-l-carboxylate. Yield: 3.2 g (86%); LC- MS: [M+H]+ = 593.3.
Step H: {l-Piperidin-4-yl-3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3- d] pyrimidin-4-yl)- lH-pyrazol- 1-yl] azetidin-3-yl}acetonitrile trih drochloride
Figure imgf000102_0001
To a solution of tert-butyl 4-{3-(cyanomethyl)-3-[4-(7-{[2-(trimethylsilyl) ethoxy]methyl} -7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol- 1 -yljazetidin- 1 - yl}piperidine-l-carboxylate (3.2 g, 5.4 mmol) in 10 mL of THF was added 10 mL of 4 N HC1 in dioxane. The reaction mixture was stirred at room temperature for 2 hours.
Removing solvents under reduced pressure yielded 3.25 g (100%) of {l-piperidin-4-yl-3- [4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol- l-yl]azetidin-3-yl}acetonitrile trihydrochloride as a white powder solid, which was used directly in the next reaction. LC-MS: [M+H]+ = 493.3. 1H NMR (400 MHz, DMSO-d6): δ 9.42 (s 1H), 9.21 (s, 1H), 8.89 (s, 1H), 8.69 (s, 1H), 7.97 (s, 1H), 7.39 (d, 1H), 5.68 (s, 2H), 4.96 (d, 2H), 4.56 (m, 2H), 4.02-3.63 (m, 2H), 3.55 (s, 2H), 3.53 (t, 2H), 3.49-3.31 (3, 3H), 2.81 (m, 2H), 2.12 (d, 2H), 1.79 (m, 2H), 0.83 (t, 2H), -0.10 (s, 9H). Step I: {l-{l-[ 3-Fluoro-2-(trifluoromethyl)isonicotinoyl] piperidin-4-yl}-3- [ 4-(7-{[ 2- (trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidin-4-yl)
ylJazetidin-3-yl) acetonitrile
Figure imgf000103_0001
A mixture of {l-piperidin-4-yl-3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H- pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile trihydrochloride (1.22 g, 2.03 mmol), 3-fluoro-2-(trifluoromethyl)isonicotinic acid (460 mg, 2.2 mmol), benzotriazol-l-yloxytris(dimethylamino)phosphonium hexafluorophosphate (1.07 g, 2.42 mmol), and triethylamine (2.0 mL, 14 mmol) in dimethylformamide (DMF) (20.0 mL) was stirred at room temperature overnight. LS-MS showed the reaction was complete. EtOAc (60 mL) and saturated NaHC03 aqueous solution (60 mL) were added to the reaction mixture. After stirring at room temperature for 10 minutes, the organic phase was seperated and the aqueous layer was extracted with EtOAc three times. The combined organic phase was washed with brine, dried over anhydrous Na2S04, filtered and evaporated under reduced pressure. Purification by flash chromatography provided the desired product {l-{l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3- [4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol- l-yl]azetidin-3-yl} acetonitrile. LC-MS: 684.3 (M+H)+.
Step J: {l-{l-[3-Fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile
Figure imgf000104_0001
Into a solution of {l-{l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4- yl}-3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH- pyrazol-l-yl]azetidin-3-yl}acetonitrile (56 mg, 0.1 mmol) in methylene chloride (1.5 mL) was added trifluoroacetic acid (1.5 mL). The mixture was stirred at room temperature for 2 hours. After removing the solvents in vacuum, the residue was dissolved in a methanol solution containing 20% ethylenediamine. After being stirred at room temperature for 1 hour, the solution was purified by HPLC (method B) to give the title compound. LC-MS: 554.3 (M+H)+; 1H NMR (400 MHz, CDC13): 9.71 (s, 1H), 8.82 (s, 1H), 8.55 (d, J=4.6 Hz, 1H), 8.39 (s, 1H), 8.30 (s, 1H), 7.52 (t, J=4.6 Hz, 1H), 7.39 (dd, Ji=3.4 Hz, J2=1.5 Hz, 1H), 6.77 (dd, Ji=3.6 Hz, J2=0.7 Hz, 1H), 4.18 (m, 1H), 3.75 (m, 2H), 3.63 (dd, Ji=7.8 Hz, J2=3.7 Hz, 2H), 3.45 (m, 2H), 3.38 (s, 2H), 3.11 (m, 1H), 2.57 (m, 1H), 1.72 (m, 1H), 1.60 (m, 1H), 1.48 (m, 1H), 1.40 (m, 1H).
Example 2. {l-[l-(3-Fluoro-4-quinolin-6-ylbenzoyl)piperidin-4-yl]-3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH- razol-l-yl]azetidin-3-yl} acetonitrile
Figure imgf000105_0001
Step A: {4-[(4-{3-(Cyanomethyl)-3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-l-yl}pipe
fluorophenyljboronic acid
Figure imgf000105_0002
To a solution of {l-piperidin-4-yl-3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H- pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile trihydrochloride (1.0 g, 2.0 mmol) in methylene chloride (DCM) (10 mL) were added benzotriazol-1- yloxytris(dimethylamino)phosphonium hexafiuorophosphate (1.2 g, 2.6 mmol), N,N- diisopropylethylamine (1.1 mL, 6.1 mmol), and 4-(dihydroxyboryl)-3-fluorobenzoic acid (0.37 g, 2.0 mmol). The mixture was stirred at room temperature overnight. Solvents were removed under reduced pressure and the residue was purified using HPLC to give 0.54 g (41%) of the corresponding product {4-[(4-{3-(cyanomethyl)-3-[4-(7-{[2- (trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-l-yl}piperidin-l-yl)carbonyl]-2-fluorophenyl}boronic acid. LC-MS: 659.3 (M+H)+.
Step B: {l-[l-(3-Fluoro-4-quinolin-6-ylbenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile
To a solution of {4-[(4-{3-(cyanomethyl)-3-[4-(7-{[2- (trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-l-yl}piperidin-l-yl)carbonyl]-2-fluorophenyl}boronic acid (50 mg, 0.08 mmol) in DMF (2 mL) were added 6-bromoquinoline (15 mg, 0.076 mmol),
triethylamine (0.021 mL, 0.15 mmol) and 3 drops of 2 N K2CO3 aqueous solution. The mixture was degased and bis(triphenylphosphine)palladium(II) chloride (5.4 mg, 0.0076 mmol) was added. The reaction mixture was stirred at 140 °C in a microwave oven for 25 minutes, then cooled to room temperature and filtered. The filtrate was purified by HPLC to afford a white powder. The white powder was dissolved in a 5 mL of DCM/TFA (1 :2). After being stirred at room temperature for 1 hour, the solution was concentrated. The residue was dissolved in 5 mL of 10% ethylenediamine in THF. After being stirred at room temperature for 2 hours, the solution was concentrated. The residue was purified by HPLC (method B) to afford the title compound {l-[l-(3-fluoro-4-quinolin-6- ylbenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-3-yl}acetonitrile. LC-MS: 612.2 (M+H)+.
The following compounds were prepared by a method analogous to that for Example 1 or Example 2.
Figure imgf000107_0001
y car ony enzont e
Figure imgf000108_0001
Figure imgf000109_0001
Figure imgf000110_0001
Figure imgf000111_0001
Figure imgf000112_0001
y acetont e
Figure imgf000113_0001
1 -yl] azetidin-3-yl} acetonitrile
Figure imgf000114_0001
Example R Compound LC-MS
# (M+H)+
{ 1 - [ 1 - ( 1 -naphthoyl)piperidin-4-yl] - 3 - [4-
87 (7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH- 517.2 pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile
{3 - [4-(7H-pyrrolo [2,3 -d]pyrimidin-4-yl)-
88 yCO 1 H-pyrazol- 1 -yl] - 1 - [ 1 -(quinolin-3 - 518.2 ylcarbonyl)piperidin-4-yl] azetidin-3 - yl} acetonitrile
{3 - [4-(7H-pyrrolo [2,3 -d]pyrimidin-4-yl)-
89 1 H-pyrazol- 1 -yl] - 1 - [ 1 -(quinolin-6- 518.2 ylcarbonyl)piperidin-4-yl] azetidin-3 - yl} acetonitrile
{ 1 - [ 1 -( 1 -benzothien-2-
90 ylcarbonyl)piperidin-4-yl]-3-[4-(7H- 523.2 pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol- 1 -yl] azetidin-3-yl} acetonitrile
{ 1 - { 1 - [(3 -chloro-6-fluoro- 1 -benzothien-2-
91 yl)carbonyl]piperidin-4-yl}-3-[4-(7H- 575.1 pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol- 1 -yl] azetidin-3-yl} acetonitrile
{ 1 - { 1 - [(3 -chloro-4-fluoro- 1 -benzothien-2-
92 yl)carbonyl]piperidin-4-yl}-3-[4-(7H- 575.1 pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol- 1 -yl] azetidin-3-yl} acetonitrile
[3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-
93 lH-pyrazol-l-yl]- l-(l- {[4- 591.1
(trifluoromethyl)- 1 -benzothien-2-
CF3 yl]carbonyl}piperidin-4-yl)azetidin-3- yl] acetonitrile
[3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-
94 lH-pyrazol-l-yl]- l-(l- {[6- 591.1
(trifluoromethyl)- 1 -benzothien-2- yl]carbonyl}piperidin-4-yl)azetidin-3- yl] acetonitrile
[3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-
95 lH-pyrazol-l-yl]- l-(l- {[7- 591.1
(trifluoromethyl)- 1 -benzothien-2- yl]carbonyl}piperidin-4-yl)azetidin-3- yl] acetonitrile
{ 1 - [ 1 -( 1 -benzothien-3 -
96 ylcarbonyl)piperidin-4-yl]-3-[4-(7H- 523.2 pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol- 1 -yl] azetidin-3-yl} acetonitrile
{3 - [4-(7H-pyrrolo [2,3 -d]pyrimidin-4-yl)-
97 1 H-pyrazol- 1 -yl] - 1 - [ 1 -( 1 ,2,3 ,4- 521.2 tetrahydronaphthalen-2- ylcarbonyl)piperidin-4-yl] azetidin-3 - yl} acetonitrile
Figure imgf000116_0001
F 1 H-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile
Figure imgf000117_0001
Figure imgf000118_0001
Example R Compound LC-MS
# (M+H)+
4'-[(4- {3-(c
126 i-0-0-~ yanomethyl)-3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol- 568.2 1 -yljazetidin- 1 -yljpiperidin- 1 - yl)carbonyl]biphenyl-4-carbonitrile
(3 - [4-(7H-pyrrolo [2,3 -d]pyrimidin-4-yl)-
127 1 H-pyrazol- 1 -yl] - 1 - { 1 - [(2,3 ',4'- 597.2 trifluorobiphenyl-4-yl)carbonyl]piperidin- 4-yl} azetidin-3-yl)acetonitrile
F 4'-[(4- {3-(cyanomethyl)-3-[4-(7H-
128 pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol- 604.2
1 -yljazetidin- 1 -yljpiperidin- 1 - yl)carbonyl]-2',5-difluorobiphenyl-3- carbonitrile
{ 1 - [ 1 -(3 -fluoro-4-quinolin-5-
129 ylbenzoyl)piperidin-4-yl]-3-[4-(7H- 612.2 pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-
XXJ X 1 -yl] azetidin-3-yl} acetonitrile
{ 1 - [ 1 -(3 -fluoro-4-isoquinolin-5-
130 ylbenzoyl)piperidin-4-yl]-3-[4-(7H- 612.2 pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol- 1 -yl] azetidin-3-yl} acetonitrile
{ l-[l-(3-fluoro-4-isoquinolin-8-
131 ylbenzoyl)piperidin-4-yl]-3-[4-(7H- 612.2 pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol- 1 -yl] azetidin-3-yl} acetonitrile
{ l-[l-(3-fluoro-4-quinolin-8-
132 ylbenzoyl)piperidin-4-yl]-3-[4-(7H- 612.2 pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol- 1 -yl] azetidin-3-yl} acetonitrile
{ 1 - [ 1 -(3 -fluoro-4-isoquinolin-7-
133 ylbenzoyl)piperidin-4-yl]-3-[4-(7H- 612.2 pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol- 1 -yl] azetidin-3-yl} acetonitrile
{ 1 - [ 1 -(3 -fluoro-4-quinolin-7-
134 ylbenzoyl)piperidin-4-yl]-3-[4-(7H- 612.2 pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol- ii ΊΓ N 1 -yl] azetidin-3-yl} acetonitrile Example R Compound LC-MS
# (M+H)+
F { 1 - [ 1 -(3 -fluoro-4-imidazo [ 1 ,2-a]pyridin-6-
135 ylbenzoyl)piperidin-4-yl]-3-[4-(7H- 601.2 pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 -yl] azetidin-3-yl} acetonitrile
{ 1 - { 1 - [4-(l ,3-benzoxazol-2-
136 yl)benzoyl]piperidin-4-yl} -3-[4-(7H- 584.2 pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 -yl] azetidin-3-yl} acetonitrile
{ 1 - { 1 - [4-(l ,3-benzoxazol-2-yl)-3-
137 fluorobenzoyl]piperidin-4-yl} -3-[4-(7H- 602.2 pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 -yl] azetidin-3-yl} acetonitrile
Figure imgf000120_0001
Figure imgf000121_0001
Figure imgf000122_0001
Figure imgf000123_0001
Figure imgf000124_0001
9.05 (s, IH), 8.75 (s, IH), 8.64 (d, IH), 8.55 (s, IH), 8.51 (s, IH), 8.23 (d, IH), 8.15 (d, 2H), 7.79 (t, IH), 7.68 (t, IH), 7.45 (dd, IH), 7.38 (dd, IH), 7.11 (dd, IH), 4.94 (d, 2H), 4.69 (m, 2H), 4.55 (m, IH), 3.75 (m, 2H), 3.63 (m, 2H), 3.14 (m, IH), 2.83 (m, IH), 2.20 (m, IH), 1.96 (m, IH), 1.39 (m, 2H).
1H NMR (400 MHz, DMSO-d6) of Example 134 (TFA salt): δ 12.42 (s, IH), 9.09 (s, IH), 9.04 (dd, IH), 8.79 (s, IH), 8.58 (m, 2H), 8.26 (s, IH), 8.18 (d, IH), 7.88 (d, IH), 7.81 (t, IH), 7.73 (dd, IH), 7.68 (dd, IH), 7.45 (dd, IH), 7.38 (dd, IH), 7.15 (dd, IH), 4.98 (d, 2H), 4.73 (m, 2H), 4.56 (m, IH), 3.78 (m, 2H), 3.67 (m, 2H), 3.24 (m, IH), 2.68 (m, IH), 2.20 (m, IH), 1.98 (m, IH), 1.40 (m, 2H).
1H NMR (400 MHz, DMSO-d6) of Example 136 (TFA salt): δ 12.31 (brs, IH), 9.06 (s, IH), 8.75 (s, IH), 8.56 (s, IH), 8.27 (d, 2H), 7.78 (m, 2H), 7.69 (dd, IH), 7.53 (d, 2H), 7.46 (dd, 2H), 7.11 (dd, IH), 4.94 (m, 2H), 4.73 (m, 2H), 4.56 (m, IH), 3.74 (s, 2H), 3.67 (m, 2H), 3.13 (m, IH), 2.84 (m, IH), 2.09 (m, IH), 1.96 (m, IH), 1.37 (m, 2H).
1H NMR (400 MHz, DMSO-d6) of Example 137 (TFA salt): δ 12.32 (brs, IH), 9.07 (s, IH), 8.76 (s, IH), 8.56 (s, IH), 8.30 (dd, IH), 7.87 (dd, 2H), 7.70 (dd, IH), 7.55 (dd, 2H), 7.45 (dd, 2H), 7.12 (dd, IH), 4.94 (m, 2H), 4.73 (m, 2H), 4.54 (m, IH), 3.74 (s, 2H), 3.67 (m, 2H), 3.13 (m, IH), 2.84 (m, IH), 2.10 (m, IH), 1.96 (m, IH), 1.37 (m, 2H).
Example 138. 3- [(3-{3-(Cyanomethyl)-3- [4-(7H-pyrrolo [2,3-d] pyrimidin-4-yl)-lH- pyrazol-l-yl]azetidin-l-yl}-8-azabicyclo[3.2.1]oct-8-yl)carbonyl]-5- fluorobenzonitrile
Figure imgf000125_0001
Step A: tert-Butyl 3-{3-(Cyanomethyl)-3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H- pyrrolo[2, 3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-l-yl}-8-azabicyclo[ 3.2.1 Joctane- 8-carboxylate
Figure imgf000126_0001
To a solution of {3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile dihydrochloride (2.6 g, 6.3 mmol) in THF (30 mL) were added tert-butyl 3-oxo-8-azabicyclo[3.2.1]octane-8- carboxylate (1.3 g, 6.3 mmol), N,N-diisopropylethylamine (4.4 mL, 25 mmol) and sodium triacetoxyborohydride (2.2 g, 10 mmol). The mixture was stirred at room temperature overnight and quenched by addition of 20 mL of brine. The solution was extracted with EtOAc. The extract was dried over anhydrous Na2S04. After removing solvent, the residue was purified by combiflash column eluting with 30-80 % EtOAc in hexanes to give the desired product tert-butyl 3-{3-(cyanomethyl)-3-[4-(7-{[2- (trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-l-yl}-8-azabicyclo[3.2.1]octane-8-carboxylate. LC-MS: 619.3 (M+H)+.
Step B: {l-(8-Azabicyclo[ 3.2.1 ]oct-3-yl)-3-[ 4-(7-{[ 2-(trimethylsilyl)ethoxyJmethyl}-7H- pyrrolo[2, 3-d] pyrimidin-4-yl)- lH-pyrazol- 1-yl] azetidin-3-yl}acetonitrile
trihydrochloride
Figure imgf000127_0001
To a solution of tert-butyl 3-{3-(cyanomethyl)-3-[4-(7-{[2- (trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol- yl]azetidin-l-yl}-8-azabicyclo[3.2.1]octane-8-carboxylate (123 mg, 0.2 mmol) in THF (3 mL) was added a 4 N solution of HC1 in dioxane (3 mL). After being stirred at room temperature for 2 hours, the solution was concentrated. The residue obtained was used for the next reaction. LC-MS: 519.3 (M+H)+.
Step C: 3-[(3-{3-(Cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin- l-yl}-8-azabicyclo [3.2.1] oct-8-yl)carbonyl] -5-fluorobenzonitrile
A mixture of { l-(8-azabicyclo[3.2.1]oct-3-yl)-3-[4-(7-{[2- (trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-3-yl}acetonitrile (100.0 mg, 0.193 mmol), 3-cyano-5-fluorobenzoic acid (31.8 mg, 0.193 mmol), benzotriazol-l-yloxytris(dimethylamino)phosphonium
hexafluorophosphate (93.8 mg, 0.212 mmol), and triethylamine(0.108 mL, 0.771 mmol) in DMF (3.0 mL) was stirred at room temperature for 2 hours. Purification by HPLC afforded the coupling product as a white powder. LCMS found: 666.3 (M+l)+. The white powder was dissolved in trifluoroacetic acid (2 mL) and methylene chloride (2 mL). The resulting solution was stirred at room temperature for 1 hour. The solvents were evaporated to driness. The residue was treated with methanol (3 mL) and
ethylenediamine (0.3 mL, 4 mmol) for 1 hour at room temperature. Purification using HPLC method A gave the title compound 3-[(3-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- IH-pyrazol- 1 -yljazetidin- 1 -yl} -8-azabicyclo[3.2.1 ]oct-8-yl)carbonyl]- 5-fiuorobenzonitrile as a TFA salt. LCMS found: 536.3 (M+H)+. 1H NMR (400 MHz, DMSO-dg): δ 8.93 (s, 1H), 8.81 (s, 1H), 8.51 (s, 1H), 7.97 (s, 1H), 7.81 (s, 1H), 7.72 (m, 3H), 7.18 (s, 1H), 4.53 (m, 2H), 3.80 (m, 1H), 3.57 (m, 6H), 1.55-2.08 (m, 8H). The following compounds were prepared by a method analogous to that for
Example 138.
Figure imgf000128_0001
Figure imgf000128_0002
Figure imgf000129_0001
Examples 149 and 150. Diastereomers of {l-[l-(3-fluorobenzoyl)-2-methylpiperidin- 4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3- yl} acetonitrile
Figure imgf000130_0001
Step A: tert-Butyl 4-{3-(Cyanomethyl)-3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-l-yl}-2-m
carboxylate
Figure imgf000130_0002
To a solution of {3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile dihydrochloride (2.6 g, 6.3 mmol) and tert-butyl 2-methyl-4-oxopiperidine-l -carboxylate (1.3 g, 6.3 mmol) in THF (30 mL) were added N,N-diisopropylethylamine (4.4 mL, 25 mmol) and sodium triacetoxyborohydride (2.2 g, 10 mmol). The mixture was stirred at room temperature overnight. After addition of 20 mL of brine, the solution was extracted with EtOAc. The extract was dried over anhydrous Na2SC"4 and filtered. After removing the solvent, the residue was purified by combiflash chromatography eluting with 30-80 % EtOAc in hexanes to give 2.6 g (81%) of the desired product tert-butyl 4-{3-(cyanomethyl)-3-[4-(7- { [2-(trimethylsilyl)ethoxy]methyl} -7H-pyrrolo [2,3 -d]pyrimidin-4-yl)- 1 H-pyrazol- 1- yl]azetidin-l-yl}-2-methylpiperidine-l-carboxylate. LC-MS: 607.3 (M+H)+.
Step B: {l-(2-Methylpiperidin-4-yl)-3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H- pyrrolo 2, 3-d) 'pyrimidin-4-yl)- 1 H-pyrazol- 1-ylj 'azetidin-3-yl}acetonitrile
Figure imgf000131_0001
To a solution of tert-butyl 4-{3-(cyanomethyl)-3-[4-(7-{[2- (trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-l-yl}-2-methylpiperidine-l-carboxylate (0.5 g) in methanol (2 mL) was added 10 mL of a 4.0 N solution of hydrogen chloride in 1,4-dioxane (40 mmol). The resulting solution was stirred at room temperature for an hour. The solvents were removed under reduced pressure to give 0.5 g (99%) of {l-(2-methylpiperidin-4-yl)-3-[4-(7-{[2- (trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-3-yl}acetonitrile as a white solid. LC-MS: 507.1 (M+H)+.
Step C: {!-[!-( 3-Fluorobenzoyl)-2-methylpiperidin-4-yl]-3-[ 4- (7H-pyrrolo[ 2, 3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile
To a solution of {l-(2-methylpiperidin-4-yl)-3-[4-(7-{[2- (trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-3-yl}acetonitrile (40 mg, 0.08 mmol) in DMF (3 mL) were added 3- fluorobenzoic acid (12.51 mg, 0.0893 mmol), benzotriazol-1- yloxytris(dimethylamino)phosphonium hexafluorophosphate (53.86 mg, 0.122 mmol) and triethylamine (0.0396 mL, 0.284 mmol). The mixture was stirred at room temperature overnight and purified by prep-LC-MS to give 20 mg of {l-[l-(3- fluorobenzoyl)-2-methylpiperidin-4-yl]-3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile as a white powder. LC/MS found: 629.3 (M+H)+.
The above white powder (20 mg, 0.03 mmol) was dissolved in 2 mL of trifluoroacetic acid and 2 mL of methylene chloride. The mixture was stirred at room temperature for an hour. The solvents were removed under reduced pressure. The residue was dissolved in methanol (2 mL) and ethylenediamine (0.03 mL, 0.4 mmol). The mixture was stirred at room temperature for an hour. Purification by HPLC (method B) gave 4.5 mg of diastereomer 1 (Example 149) and 4.5 mg of diastereomer 2 (Example 150) as a white solid. Both diastereomers were mixture of 2 enantiomers. LC/MS found: 499.3 (M+H)+ for both diastereomers.
Example 151. { l-{ 1- [(4,4-difluorocyclohexyl)carbonyl] -2-methylpiperidin-4-yl}-3- [4-(7H-pyrrolo[2,3-d]pyrimidin-4- l)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile
Figure imgf000132_0001
The title compound was prepared as a mixture of 4 isomers by a method analogous to that for Examples 149 and 150. LC-MS: 523.2 (M+H)+. Example 152. {l-[l-(3-Fluorobenzoyl)-4-methylpiperidin-4-yl]-3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile
Figure imgf000133_0001
Step A: tert-Butyl 4-[3-(Cyanomethylene)azetidin-l-yl]-4-methylpiperidine-l-carboxylate
Figure imgf000133_0002
A round-bottom flask was charged with {3-[4-(7-{[2- (trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-3-yl}acetonitrile trihydrochloride (1.0 g, 2.4 mmol), tert-butyl 4-oxo-l- piperidinecarboxylate (0.49 g, 2.4 mmol), titanium tetraisopropoxide (0.72 mL, 2.4 mmol), triethylamine (1.0 mL, 7.3 mmol), and 10 mL of dichloromethane. The reaction mixture was stirred at room temperature overnight and then rotovaped to driness to give an oil residue which was used directly for the next step.
The above residue was dissolved in 25 mL of THF. To the resulting solution was added a 1.0 M solution of diethylaluminum cyanide in toluene (8.4 mL, 8.4 mmol). The mixture was stirred at 30 °C for 5 hours. The reaction was quenched with 1 mL of water and 20 mL of EtOAc, stirred for 30 min and filtered through celite. The celite was washed with 20 mL of EtOAc. The filtrate was dried over Na2S04 and concentrated to driness to give 1.3 g of the desired product as a colorless oil. MS found: 618 (M+H)+.
The colorless oil was dissolved in THF (20 mL ) and a 3 M solution of methylmagnesium bromide in THF (0.45 mL, 1.3 mmol) was added. The mixture was stirred at room temperature overnight. The reaction was quenched by addition of 15 mL of water and 25 mL of EtOAc. After being stirred for 30 min, the solution was filterred through a celite bed. The organic layer was separated, dried over anhydrous Na2S04, and evaporated under reduced pressure. Purification by HPLC afforded the desired product tert-butyl 4-[3-(cyanomethylene)azetidin-l-yl]-4-methylpiperidine-l-carboxylate. LC- MS: 292.1 (M+H)+.
Step B: tert-Butyl 4-{3-(Cyanomethyl)-3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH^yrazol-l-yl]azetidin-l-yl}-4-methylpipe
carboxylate
Boc
Figure imgf000134_0001
A 2 L round bottom flask fitted with overhead stirring, septa and nitrogen inlet was charged with tert-butyl 4-[3-(cyanomethylene)azetidin-l-yl]-4-methylpiperidine-l- carboxylate (9.17 g, 0.0472 mol), 4-(lH-pyrazol-4-yl)-7-{[2-
(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidine (14.9 g, 0.0472 mol) and acetonitrile (300 mL). The resulting solution was heterogeneous. To the solution was added l,8-diazabicyclo[5.4.0]undec-7-ene (8.48 mL, 0.0567 mol) portionwise via a syringe over 3 minutes at room temperature. The solution slowly became homogeneous and yellow in color. The reaction was allowed to stir at room temperature for 3 hours. The solution was concentrated on a rotovap to remove -150 mL of acetonitrile. After addition of 100 mL of EtOAc and 100 mL of 20% brine, the organic phase was separated. The aqueous layer was extracted with 150 mL of EtOAc. The combined organic phase was dried over MgSC^, filtered and concentrated to yield an orange oil. Purification by flash chromatography (150 grams of silica, 60% EtOAc/hexanes, loaded with CH2CI2) yielded the title compound as a white foam. LC-MS: 607.2 (M+H)+.
Step C: {l-(4-Methylpiperidin-4-yl)-3-[4-(7-{f2-(trimethylsilyl)ethoxyJmethy
pyrrolo 2, 3-d) 'pyrimidin-4-yl)- lH-pyrazol- 1-ylj azetidin-3-ylj acetonitrile
Figure imgf000135_0001
To a solution of tert-butyl 4-{3-(cyanomethyl)-3-[4-(7-{[2- (trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-l-yl}-4-methylpiperidine-l-carboxylate (30 mg, 0.05 mmol) in THF (2 mL) was added a 4 N solution of HC1 in dioxane (2 mL). After being stirred at room temperature for 2 hours, the reaction mixture was evaporated under reduced pressure to give the title compound (31 mg, 99%), which was used for the next reaction. LC-MS found: 507.2 (M+H)+. Step D: {l-[l-(3-Fluorobenzoyl)-4-methylpiperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile
To a solution of {l-(4-methylpiperidin-4-yl)-3-[4-(7-{[2- (trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-3-yl}acetonitrile (40 mg, 0.08 mmol) in DMF (3 mL) were added
benzotriazol- 1 -yloxytris(dimethylamino)phosphonium hexafluorophosphate (45 mg, 0.10 mmol), N,N-diisopropylethylamine (0.041 mL, 0.24 mmol), and 3-fluorobenzoic acid (11 mg, 0.079 mmol). The reaction mixture was stirred at room temperature overnight.
Purification by HPLC afforded the desired intermediate as a white powder, which was then treated with TFA (1 mL) and DCM (1 mL) for 1 hour at room temperature. After removing the solvents, the residue was treated with ethylenediamine (1 mL) in methanol (5 mL) for 2 hours. Purification using HPLC method A provided the final product {1-[1- (3-fluorobenzoyl)-4-methylpiperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH- pyrazol-l-yl]azetidin-3-yl}acetonitrile as a TFA salt. LC-MS found: 499.3 (M+H)+. 1H NMR (400 MHz, DMSO-d6): δ 12.36 (s, 1H), 8.81-9.11 (m, 2H), 8.77 (s, 1H), 8.57 (s, 1H), 7.71 (t, 1H), 7.31 (m, 1H), 7.25 (dd, 2H), 7.17 (s, 1H), 4.5-4.85 (m, 4H), 3.82 (m, 2H), 3.56 (s, 4H), 3.21 (m, 1H), 2.60 (m, 1H) , 1.55-1.75 (m, 2H), 1.37 (s, 3H).
Example 153. { 1- [ l-(Cyclohexylcarbonyl)-4-methylpiperidin-4-yl] -3- [4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile
Figure imgf000137_0001
The title compound was prepared by a method analogous to that for Example 152 and was obtained as a TFA salt using HPLC method A for purification. LC-MS: 487.2 (M+H)+. 1H NMR (400 MHz, DMSO-d6): δ 12.28(s, 1H), 8.81-9.1 l(m, 1H), 8.74 (s, 1H), 8.55(s, 1H), 7.68 (t, 1H), 7.11 (m, 1H), 4.40-4.85 (m, 4H), 3.80-3.95 (m, 1H), 3.56 (s, 2H), 3.15 (m, 1H), 2.60 (m, 1H) , 1.63 (m, 10H), 1.43 (s, 3H), 1.12-1.28 (m, 6H).
Example 154. 4- {3-(Cyanomethyl)-3- [4-(7H-pyrrolo [2,3-d] pyrimidin-4-yl)- 1H- pyrazol-l-yl]azetidin-l-yl}-N-[4-fluoro-2-(trifluoromethyl)phenyl]piperidine-l- carboxamide
Figure imgf000137_0002
Step A: 4-{3-(Cyanomethyl)-3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l -yl] azetidin-1 -yl}-N-[4-fluoro-2- (trifluoromethyl)phenyl]piperidine-l-carboxamide
Figure imgf000138_0001
To a solution of {l-piperidin-4-yl-3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H- pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile trihydrochloride (500 mg, 1 mmol) in tetrahydrofuran (30 mL) were added triethylamine (0.29 g, 2.8 mmol) and 4-fluoro-l-isocyanato-2-(trifluoromethyl)benzene (190 mg, 0.95 mmol). The mixture was stirred at room temperature for 1 hour. The solvent was removed under reduced pressure. Purification by combi-flash using 30-100% EtOAc/hexanes gave 4-{3- (cyanomethyl)-3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidin- 4-yl)- 1 H-pyrazol- 1 -yljazetidin- 1 -yl} -N-[4-fluoro-2-(trifluoromethyl)phenyl]piperidine- 1 - carboxamide as a powder. LC-MS: 698.1 (M+H)+. Step B: 4-{3-(Cyanomethyl)-3- [4-(7H-pyrrolo [2 ,3-d] pyrimidin-4-yl)- lH-pyrazol-1- yl] azetidin- l-yl}-N- [4-fluoro-2-(trifluoromethyl)phenyl] piperidine- 1 -carboxamide
4-{3-(Cyanomethyl)-3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yljazetidin- 1 -yl} -N-[4-fluoro-2-
(trifluoromethyl)phenyl]piperidine-l -carboxamide (210 mg, 0.3 mmol) was dissolved in a 50 M solution of trifluoroacetic acid in methylene chloride (20 mL). After being stirred at room temperature for one hour, the solvents were removed under reduced pressure. The residue was dissolved in methanol (20 mL) and ethylenediamine (1.0 g, 17 mmol). After being stirred at room temperature for one hour, the mixture was purified by HPLC (method B) to give 4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH- pyrazol- 1 -yljazetidin- 1 -yl} -N-[4-fluoro-2-(trifluoromethyl)phenyl]piperidine- 1 - carboxamide as a white powder. LC-MS: 568.1 (M+H)+. 1H NMR (400 MHz, DMSO- d6): δ 12.10 (s, 1H), 8.76 (s, 1H), 8.63 (s, 1H), 8.36 (s, 1H), 8.18 (s, 1H), 7.55 (d, J=3.6 Hz, 1H), 7.50 (m, 1H), 7.43 (m, 1H), 7.34 (m, 1H), 7.01(d, J=3.6 Hz, 1H), 3.79 (m, 2H), 3.67 (d, J=8 Hz, 2H), 3.51 (m, 4H), 2.92(m, 2H), 2.38 (m, 1H), 1.62 (m, 2H), 1.09 (m, 2H).
Example 155. 4- {3-(Cyanomethyl)-3- [4-(7H-pyrrolo [2,3-d] yrimidin-4-yl)- 1H- pyrazol-l-yl]azetidin-l-yl}-N-[3-(trifluoromethyl)pyridin-4-yl]piperidine-l- carboxamide
Figure imgf000139_0001
Step A: 4-{3-(Cyanomethyl)-3- [4-(7-{ [2-(trimethylsilyl)ethoxy] methyl}-7H- pyrrolo [2,3-d] pyrimidin-4-yl)- lH-pyrazol-l-yl] azetidin- l-yl}-N-[3- (trifluoromethyl)pyridin-4-yl]piperidine-l -carboxamide
Figure imgf000140_0001
A 20 mL vial was charged with 4-(trifluoromethyl)pyridin-3 -amine (15.6 mg, 0.0963 mmol), THF (2 mL), a 20 M solution of phosgene in toluene (0.50 mL, 1 mmol) and triethylamine (0.017 mL, 0.12 mmol). The mixture was stirred at room temperature for one hour and concentrated. To the vial were added {l-piperidin-4-yl-3-[3-(7-{[2- (trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrrol-l- yl]azetidin-3-yl}acetonitrile trihydrochloride (40 mg, 0.08 mmol), THF (2 mL) and triethylamine (0.025 g, 0.24 mmol). The mixture was stirred for two hours and purified with HPLC to give 4-{3-(cyanomethyl)-3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol- 1 -yljazetidin- 1 -yl} -N-[3- (trifluoromethyl)pyridin-4-yl]piperidine-l-carboxamide as a white solid. LC-MS: 681.3 (M+H)+.
Step B: 4-{3-(Cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- ylJazetidin-l-yl}-N-[3-(trifluoromethyl)pyridin-4-ylJpiperidine-l-carbo
A 20 mL vial was charged with 4-{3-(cyanomethyl)-3-[4-(7-{[2- (trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-l-yl}-N-[3-(trifluoromethyl)pyridin-4-yl]piperidine-l-carboxamide (56 mg, 0.1 mmol), trifluoroacetic acid (1.5 mL, 19 mmol) and methylene chloride (1.5 mL). The mixture was stirred at room temperature for 1 hour and concentrated in vacuum. The residue was dissolved in 3 mL of a methanol solution containing 20% ethylenediamine. After being stirred at room temperature for 1 hour, HPLC purification (method B) gave the title compound 4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH- pyrazol- 1 -yl] azetidin- 1 -yl} -N- [3 -(trifluoromethyl)pyridin-4-y ljpiperidine- 1 -carboxamide. LC-MS: 551.2 (M+H)+.
The following compounds were prepared by a method analogous to that for Example 154 or Example 155.
Figure imgf000141_0001
Figure imgf000141_0002
Figure imgf000142_0001
Figure imgf000143_0001
Figure imgf000144_0001
Figure imgf000145_0001
Figure imgf000146_0001
-y car ony amno - - uoro enzoate
Figure imgf000146_0002
Figure imgf000147_0001
Figure imgf000148_0001
Figure imgf000149_0001
Figure imgf000150_0001
Example 209. 3- [(4-{3-(Cyanomethyl)-3- [4-(7H-pyrrolo [2,3-d] pyrimidin-4-yl)-lH- pyrazol-l-yl]azetidin-l-yl}piperidin-l-yl)methyl]-6-(dimethylamino)-2- fluorobenzonitrile
Figure imgf000151_0001
Step A: 3- [(4-{3-(Cyanomethyl)-3- [4-(7-{ [2-(trimethylsilyl)ethoxy] methyl}-7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-l-yl}piperidin-l-yl^
(dimethylamino)-2-fluorobenzonitrile
Figure imgf000151_0002
To a solution of {l-piperidin-4-yl-3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H- pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile trihydrochloride
(200 mg, 0.4 mmol) in THF (10 mL) and triethylamine (0.1643 mL, 1.179 mmol) was added 6-(dimethylamino)-2-fiuoro-3-formylbenzonitrile (75.52 mg, 0.3929 mmol). The solution was stirred at room temperature for 30 minutes before the addition of sodium triacetoxyborohydride (249.8 mg, 1.179 mmol). The mixture was stirred at room temperature overnight. After addition of aqueous NaHC03, and EtOAc, the organic layer was separated, washed with brine, dried over anhydrous Na2S04 and concentrated. Purification by HPLC afforded 150 mg of the product 3-[(4-{3-(cyanomethyl)-3-[4-(7- { [2-(trimethylsilyl)ethoxy]methyl} -7H-pyrrolo [2,3 -d]pyrimidin-4-yl)- 1 H-pyrazol- 1 - yl] azetidin- 1 -yl} piperidin- 1 -yl)methyl] -6-(dimethy lamino)-2-fluorobenzonitrile as a white solid. LC/MS: 669.2 (M+H)+.
Step B: 3-[(4-{3-(Cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- ylj 'azetidin- l-yl}piperidin-l-yl)methyl]-6-(dimethylamino)-2-fluorobenzonitrile
Into a reaction vial were added 3-[(4-{3-(cyanomethyl)-3-[4-(7-{[2- (trimethylsilyl)ethoxy]methyl} -7H-pyrrolo[2,3-d]pyrimidin-4-yl)- 1 H-pyrazol- 1 - yl] azetidin- 1 -yl} piperidin- 1 -yl)methyl] -6-(dimethylamino)-2-fluorobenzonitrile (56 mg, 0.1 mmol), trifluoroacetic acid (1.5 mL) and methylene chloride (1.5 mL). The mixture was stirred at room temperature for 1 hour and concentrated in vacuum. The residue was dissolved in 5 mL of a methanol solution containing 20% ethylenediamine. After being stirred at room temperature for 1 hour, the mixture was purified by HPLC (method B) to give the title compound. LC-MS: 539.2 (M+H)+. 1H NMR (400 MHz, DMSO-d6): δ 12.10 (s, 1H), 8.79 (s, 1H), 8.67 (s, 1H), 8.39 (s, 1H), 7.59 (d, J=3.6 Hz, 1H), 7.42 (t, J=8.8 Hz, 1H), 7.05 (d, J=3.6 Hz, 1H), 6.78 (d, J=8.8 Hz, 1H), 3.68 (d, J=8.4 Hz, 2H), 3.51 (m, 4H), 3.38 (s, 2H), 3.01 (s, 6H), 2.67 (m, 2H), 2.17 (m, 1H), 1.97 (m, 2H), 1.63 (m, 2H), 1.15 (m, 2H).
The following compounds were prepared by a method analogous to that for Example 209.
Figure imgf000153_0001
Figure imgf000154_0001
Figure imgf000155_0001
Figure imgf000156_0001
Figure imgf000157_0001
Figure imgf000158_0001
Example 240. 3- [(3-{3-(Cyanomethyl)-3- [4-(7H-pyrrolo [2,3-d] pyrimidin-4-yl)-lH- pyrazol-l-yl]azetidin-l-yl}-8-azabicyclo[3.2.1]oct-8-yl)methyl]-6-(dimethylamino)-2- fluorobenzonitrile
\
Figure imgf000158_0002
To a solution of {l-(8-azabicyclo[3.2.1]oct-3-yl)-3-[4-(7-{[2- (trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-3-yl}acetonitrile (40 mg, 0.08mmol) in THF (2mL) were added 6- (dimethylamino)-2-fluoro-3-formylbenzonitrile (17.2 mg, 0.089 mmol), and
triethylamine (0.034 mL, 0.24 mmol). The mixture was stirred at room temperature for 30 minutes before the addition of sodium triacetoxyborohydride (51.6 mg, 0.24 mmol). The mixture was stirred at room temperature overnight. The resulting solution was worked up with aqueous NaHC03 and EtOAc. The organic layer was washed with brine, dried over Na2S04 and concentrated under reduced pressure. Purification with acidic prep-LCMS afforded 25 mg (41.6%) of the desired intermediate 3-[(3-{3-(cyanomethyl)- 3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-]pyrimidin-4-yl)-lH-pyrazol- 1 -yl]azetidin-l -yl} -8-azabicyclo[3.2.1 ]oct-8-yl)methyl]-6-(dimethylamino)-2- fluorobenzonitrile. LC/MS found: 695.3 (M+H)+.
The above white solid (25 mg, 0.036 mmol) was dissolved in a 50 M solution of trifluoroacetic acid in methylene chloride (2mL, 100 mmol). The mixture was stirred at room temperature for an hour and concentrated under reduced pressure. The residue was dissolved in methanol (2 mL, 50 mmol) and ethylenediamine (0.03 mL, 0.4 mmol). After being stirred at room temperature for an hour, the mixture was purified with HPLC (method B) to give about 10 mg (50%) of the title compound as a white solid. LC/MS found: 565.3 (M+H)+. 1H NMR (400 MHz, DMSO-d6): δ 12.20 (s, 1H), 8.90 (s, 1H), 8.79 (s, 1H), 8.50 (s, 1H), 7.71 (d, J=3.2 Hz, 1H), 7.67 (t, J=8.8 Hz, 1H), 7.15 (d, J=3.2 Hz, 1H), 6.90 (d, J=8.8 Hz, 1H), 3.65 (m, 6H), 3.48 (s, 2H), 3.12 (s, 6H), 3.07 (m, 2H), 2.60 (m, 1H), 2.05 (m, 2H), 1.96 (m, 2H), 1.80 (m, 2H), 1.57 (m, 2H).
Example 241. {l-[8-(2-Chloro-3,6-difluorobenzyl)-8-azabicyclo[3.2.1]oct-3-yl]-3-[4- (7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile
Figure imgf000160_0001
The title compound was prepared by a method analogous to that for Example 240. LC-MS: 549.1 (M+H)+. Examples 242 and 243. Diastereomers of 3-[(4-{3-(cyanomethyl)-3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-l-yl}-2-methylpiperidin-l- yl)methyl]-6-(dimethylamino)-2-fluorobenzonitrile
Figure imgf000160_0002
To a solution of {l-(2-methylpiperidin-4-yl)-3-[4-(7-{[2- (trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-3-yl}acetonitrile (40 mg, 0.08 mmol) in THF (2 mL) were added 6- (dimethylamino)-2-fluoro-3-formylbenzonitrile (17.16 mg, 0.0893 mmol) and triethylamine (0.034 mL, 0.244 mmol). The solution was stirred at room temperature for 30 min before the addition of sodium triacetoxyborohydride (51.62 mg, 0.244 mmol). The mixture was stirred at room temperature overnight. The resulting mixture was quenched with aqueous NaHC03 and EtOAc. The organic layer was separated and washed with brine, dried over Na2S04 and concentrated under reduced pressure. The residue was purified with acidic prep-LCMS to afford 25 mg (47%) of the intermediate 3- [(4-{3-(cyanomethyl)-3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yljazetidin- 1 -yl} -2-methylpiperidin- 1 -yl)methyl]-6- (dimethylamino)-2-fluorobenzonitrile as a white solid. LC/MS found: 683.2 (M+H)+.
The above white solid powder (25 mg, 0.037 mmol) was dissolved in 2 mL of trifluoroacetic acid and 2 mL of methylene chloride. The mixture was stirred at room temperature for an hour. The solvents were removed under reduced pressure. The residue was dissolved in methanol (2 mL) and ethylenediamine (0.03 mL, 0.4 mmol). After being stirred at room temperature for an hour, the mixture was purified with HPLC (method B) to give the desired two products Example 245 and Example 246 as a white solid:
Example 245 (7 mg) was the fast moving diastereomer on HPLC and Example 246 (7 mg) was the slow moving diatereomer on HPLC. LC/MS found: 553.2 (M+H)+ for both isomers. Examples 244 and 245. Distereomers of {l-[l-(2-chloro-6-fluorobenzyl)-2- methylpiperidin-4-yl] -3- [4-(7H-pyrr olo [2,3-d] pyrimidin-4-yl)- 1 H-pyrazol- 1- yl] azetidin-3-yl}acetonitrile
Figure imgf000162_0001
The title compounds were prepared by a method analogous to that for Examples 242 and 243. Example 247 was the fast moving diastereomer on HPLC and Example 248 was the slow moving diatereomer on HPLC. LC/MS found: 519.2 (M+H)+ for both isomers.
Example 246. {l-{l-[(l-Methyl-lH-pyrazol-5-yl)sulfonyl]piperidin-4-yl}-3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile
Figure imgf000162_0002
Step A: {l-{l-[(l-Methyl-lH-pyrazol-5-yl)sulfonyl]piperidin-4-yl}-3-[4-(7-{[2- (trimethylsilyl)ethoxy]methyl}-7H^yrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin-3-yl}acetonitrile
Figure imgf000163_0001
A mixture of {3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile trihydrochloride (44.4 mg, 0.108 mmol), 1 -methyl- lH-pyrazole-5-sulfonyl chloride (19.6 mg, 0.108 mmol) and triethylamine (0.0412 mL, 0.296 mmol) in THF (10.0 mL) was stirred at room
temperature for 2 hours. Purification on silica gel column afforded the desired product { 1 - { 1 -[( 1 -methyl- 1 H-pyrazol-5-yl)sulfonyl]piperidin-4-yl} -3-[4-(7- { [2- (trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-3-yl}acetonitrile. Yield: 58.8%. LC-MS: 637.3 (M+H)+.
Step B: {l-{l-[(l-Methyl-lH-pyrazol-5-yl)sulfonyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile
To a solution of {l-{l-[(l-methyl-lH-pyrazol-5-yl)sulfonyl]piperidin-4-yl}-3-[4- (7- { [2-(trimethylsilyl)ethoxy]methyl} -7H-pyrrolo[2,3-d]pyrimidin-4-yl)- 1 H-pyrazol- 1 - yl]azetidin-3-yl}acetonitrile (56 mg, 0.1 mmol) in methylene chloride (1.5 mL) was added trifluoroacetic acid (1.5 mL). The mixture was stirred at room temperature for 2 hours and concentrated in vacuo. The residue was dissolved in 2 mL of a methanol solution containing 20% ethylenediamine. After being stirred at room temperaturee for 1 hour, the mixture was purified by HPLC (method B) to give 20 mg (64.5%>) of {1-{1-[(1- methyl-lH-pyrazol-5-yl)sulfonyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile. LC-MS: 507.2 (M+H)+. 1H NMR (300 MHz, DMSO-dg): δ 12.08 (brs, 1H), 8.72 (d, 1H), 8.62 (s, 1H), 8.34 (s, 1H), 7.55 (d, 2H), 6.99 (d, IH), 6.75 (s, IH), 3.95 (s, 3H), 3.62 (dd, 2H), 3.45 (dd, 2H), 3.40 (m, 2H), 3.25 (s, 2H), 3.19 (m, IH), 2.75 (m, 2H), 1.70 (m, 2H), 1.25 (m, 2H).
The following compounds were prepared by a method analogous to that for Example 246.
Figure imgf000164_0001
Figure imgf000165_0001
pyrazo- -y azet n- -y acetont e
Figure imgf000165_0002
Figure imgf000166_0001
Example 261. {l-{l-[3-Fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[3- (7H-pyrrolo[2,3-d]pyrimidin-4-y -lH-pyrrol-l-yl]azetidin-3-yl}acetonitrile
Figure imgf000166_0002
Step A: 4-( lH-Pyrrol-3-yl)-7-{ [2-(trimethylsilyl)ethoxy] 1 methyl} -7 H-pyrrolo [2 ,3- djpyrimidine
Figure imgf000166_0003
A 100 mL round bottom flask was charged with 4-chloro-7-{[2- (trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidine (1.00 g, 3.52 mmol), 1- butanol (25.0 mL), [l-(triisopropylsilyl)-lH-pyrrol-3-yl]boronic acid (1.41 g, 5.28 mmol), water (25.0 mL) and potassium carbonate (1.27 g, 8.8 mmol). This solution was degased 4 times, filling with nitrogen each time. Tetrakis(triphenylphosphine)- palladium(O) (0.41 g, 0.35 mmol) was added and the mixture was degased 4 times, filling with nitrogen each time. The reaction was stirred overnight at 100 °C and cooled to room temperature. The mixture was filtered through a bed of celite and the celite was rinsed with ethyl acetate (42 mL). The filtrate was combined and the organic layer was separated. The aqueous layer was extracted with ethyl acetate. The organic extracts were combined and concentrated under vacuum with a bath temerature of 30-70 °C to give the title compound 4-(lH-pyrrol-3-yl)-7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3- d]pyrimidine. Yield: 83%; LC-MS: 315.2 (M+H)+.
Step B: tert-Butyl 3-(Cyanomethyl)-3-[3-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrrol-l-yl]azetidine-l-carboxylate
Figure imgf000167_0001
A 100 mL round bottom flask fitted with overhead stirring, septa and nitrogen inlet was charged with tert-butyl-3-(cyanomethylene)azetidine-l-carboxylate (1.8 g, 9.5 mmol), 4-(lH-pyrrol-3-yl)-7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3- d]pyrimidine (3.0 g, 9.5 mmol) and acetonitrile (60 mL). The resulting solution was heterogeneous. l,8-Diazabicyclo[5.4.0]undec-7-ene (1.4 mL, 9.5 mmol) was added portionwise via a syringe over 3 minutes at room temperature. The solution slowly becomes homogeneous and yellow in color. The reaction was allowed to stir at room temperature for 3 hours. The solution was concentrated by rotary evaporation to remove acetonitrile. EtOAc (100 mL) and brine (100 mL) were added. The organic phase was separated and the aqueous layer was extracted with 3><30 mL EtOAc. The combined extracts were dried over anhydrous MgSC^, filtered and concentrated in vacuo to yield an orange oil which was purified by flash chromatography (120 grams silica, 30-55% EtOAc/hexane, loaded with CH2CI2). Desired fractions were combined and concentrated to yield a yellow oil which was placed on a high vacuum pump to give 4 g (83%) of tert- butyl 3 -(cyanomethyl)-3 - [3 -(7- { [2-(trimethylsilyl)ethoxy]methyl} -7H-pyrrolo [2,3 - d]pyrimidin-4-yl)-lH-pyrrol-l-yl]azetidine-l-carboxylate as a white foam. LC-MS: [M+H]+ = 509.3.
Step C: {3-[3-(7-{[2-(Trimethylsilyl)ethoxy]methyl}-7H^yrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrrol-l- lJazetidin- 3 '-ylj acetonitrile dihydrochloride
Figure imgf000168_0001
To a solution of tert-bv&yl 3-(cyanomethyl)-3-[3-(7-{[2- (trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrrol-l- yl]azetidine-l-carboxylate (4 g, 7.87 mmol) in 20 mL of THF was added 20 mL of 4 N HC1 in dioxane. After being stirred at room temperature for 1 hour, the solvents were removed in vacuo to give 3.9 g (99%) of the desired product {3-[3-(7-{[2- (trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrrol-l- yl]azetidin-3-yl} acetonitrile dihydrochloride, which was used for the next reaction. LC- MS: [M+H]+ = 409.3.
Step D: tert-Butyl 4-{3-(Cyanomethyl)-3-f3-(7-{f2-(trimethylsilyl)ethoxyJmethyl}-7H- pyrrolo[2,3-dJpyrimidin-4-yl)-lH^yrrol-l-ylJazetidin-l-yl}piperidine-l-carboxylate Boc
Figure imgf000169_0001
To a suspension of {3-[3-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrrol-l-yl]azetidin-3-yl}acetonitrile dihydrochloride (3.0 g, 7.3 mmol) in THF (30 mL) were added tert-butyl 4-oxo-l-piperidinecarboxylate (1.4 g, 7.3 mmol), N,N-diisopropylethylamine (6.4 mL, 37 mmol) and sodium
triacetoxyborohydride (3.1 g, 15 mmol). The reaction mixture was stirred at room temperature overnight. Brine (20 mL) and EtOAc (20 mL) were added. The organic phase was separated and the aqueous layer was extracted with EtOAc. The combined extracts were dried over sodium sulfate, filtered, and evaporated in vacuo. The residue was purified using combiflash column eluting with 20-50 % EtOAc in hexanes to generate tert-butyl 4-{3-(cyanomethyl)-3-[3-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H- pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrrol- 1 -yljazetidin- 1 -yl}piperidine- 1 -carboxylate as an oil. Yield: 3.37 g (78%); LC-MS: [M+H]+ = 592.3.
Step E: {l-Piperidin-4-yl-3-[ 3-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[ 2, 3- d]pyrimidin-4-yl)-lH-pyrrol-l-yl]azetidin-3-yl}acetonitrile trihydrochloride
Figure imgf000170_0001
To a solution of tert-butyl 4-{3-(cyanomethyl)-3-[3-(7-{[2- (trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrr^
yl]azetidin-l-yl}piperidine-l-carboxylate (3.3 g, 5.6 mmol) in THF (17 mL) was added a 4 N solution of HC1 in dioxane (17 mL). The mixture was stirred at room temperature for 2 hours and concentrated to afford {l-piperidin-4-yl-3-[3-(7-{[2- (trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrrol-l- yl]azetidin-3-yl}acetonitrile trihydrochloride as a white powder solid, which was used for the next reaction. Yield: 99%; LC-MS: [M+H]+ = 492.3.
Step F: {l-{l-[ 3-Fluoro-2-(trifluoromethyl)isonicotinoyl] piperidin-4-yl}-3- [ 3-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrrol-l-yl]azetidin-3-yl}acetonitrile
A mixture of {l-piperidin-4-yl-3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H- pyrrolo [2,3 -d]pyrimidin-4-yl)- 1 H-pyrrol- 1 -yl] azetidin-3 -yl} acetonitrile trihydrochloride (1.22 g, 2.03 mmol), 3-fluoro-2-(trifluoromethyl)isonicotinic acid (460 mg, 2.2 mmol), benzotriazol-l-yloxytris(dimethylamino)phosphonium hexafluorophosphate (1.07 g, 2.42 mmol) and triethylamine (2.0 mL, 14 mmol) in DMF (20.0 mL) was stirred at room temperature overnight. LS-MS showed the reaction was complete. EtOAc (60 mL) and saturated NaHCC"3 aqueous solution (60 mL) were added to the reaction mixture. After stirring at room temperature for 10 minutes, the organic phase was seperated and the aqueous layer was extracted with EtOAc three times. The combined extracts were washed with brine, dried over anhydrous Na2S04, filtered and evaporated under reduced pressure. Purification by flash chromatography provided {l-{l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7-{[2- (trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrrol-l- yl]azetidin-3-yl}acetonitrile as a white powder.
The white powder was dissolved in trifluoroacetic acid (5 mL) and methylene chloride (5 mL). The mixture was stirred at room temperature for 2 hours and concentrated in vacuo. The residue was dissolved in 10 mL of a methanol solution containing 20% ethylenediamine. After being stirred at room temperature for 1 hour, HPLC purification (method B) gave the title compound {l-{l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[3-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrrol-l-yl]azetidin-3-yl}acetonitrile. LC-MS: 553.3 (M+H)+. 1H NMR (400 MHz, CDC13): δ 9.10 (m, 1H), 8.77 (s, 1H), 8.57 (d, J=4.8 Hz, 1H), 7.57 (s, 1H), 7.53 (t, J=4.7 Hz, 1H), 7.31 (dd, Ji=3.6 Hz, J2=2.2 Hz, 1H), 6.97 (dd, Ji=2.9 Hz, J2=1.5 Hz, 1H), 6.83 (dd, Ji=3.8 Hz, J2=2.1 Hz, 1H), 6.81 (t, J=2.6 Hz, 1H), 4.17 (m, 1H), 3.76 (m, 2H), 3.50 (dd, Ji=9.1 Hz, J2=7.4 Hz, 2H), 3.44 (m, 2H), 3.21 (s, 2H), 3.09 (m, 1H), 2.52 (m, 1H), 1.81 (m, 1H), 1.69 (m, 1H), 1.51 (m, 1H), 1.23 (m, 1H).
The following compounds were prepared by a method analogous to that for Example 261.
Figure imgf000171_0001
Figure imgf000171_0002
Figure imgf000172_0001
Figure imgf000173_0001
1H NMR (400 MHz, DMSO-d6) of Example 262: δ 11.98 (s, IH), 8.60 (s, IH), 7.90 (m, IH), 7.86 (m, IH), 7.82 (m, IH), 7.69 (m, IH), 7.63 (t, IH), 7.50 (d, IH), 7.07 (t, IH), 6.93 (m, 2H), 4.10 (m, IH), 3.56 (m, 5H), 3.45 (m, 3H), 3.17 (m, IH), 3.05 (m, IH), 1.74 (m, IH), 1.62 (m, IH), 1.25 (m, 2H).
1H NMR (400 MHz, DMSO-d6) of Example 263: δ 11.97 (s, IH), 8.60 (s, IH), 7.95 (m, IH), 7.82 (t, IH), 7.76 (t, IH), 7.68 (m, IH), 7.50 (d, IH), 7.07 (t, IH), 6.93 (m, 2H),
Figure imgf000174_0001
Example 283. 4- {3-(Cyanomethyl)-3- [3-(7H-pyrrolo [2,3-d] pyrimidin-4-yl)- 1H- pyrrol-l-yl]azetidin-l-yl}-N-[4-fluoro-2-(trifluoromethyl)phenyl]piperidine-l- carboxamide
Figure imgf000175_0001
To a solution of {l-piperidin-4-yl-3-[3-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H- pyrrolo [2,3 -d]pyrimidin-4-yl)- 1 H-pyrrol- 1 -yl] azetidin-3 -yl} acetonitrile trihydrochloride (500 mg, 1 mmol) in THF (30 mL) were added triethylamine (0.29 g, 2.8 mmol), and 4- fluoro-l-isocyanato-2-(trifiuoromethyl)benzene (190 mg, 0.95 mmol). The mixture was stirred at room temperature for one hour. The solvent was removed under reduced pressure. Purification with combi-fiash using 30-100% EtOAc/hexanes gave the product as a powder. LC-MS: 697.1 (M+H)+.
Into the above solid was added a 50 M solution of trifluoroacetic acid in methylene chloride (20 mL, 1000 mmol). After being stirred at room temperature for one hour, the solvent was removed. The residue was dissolved in methanol (20 mL) and ethylenediamine (1.0 g, 17 mmol). After being stirred at room temperature for one hour, the mixture was purified with HPLC (method B) to give 4-{3-(cyanomethyl)-3-[3-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrrol- 1 -yljazetidin- 1 -yl} -N-[4-fluoro-2- (trifluoromethyl)phenyl]piperidine-l-carboxamide as a white powder. LC-MS: 567.2
(M+H)+. 1H NMR (400 MHz, DMSO-d6): δ 11.95 (s, 1H), 8.60 (s, 1H), 8.22 (s, 1H), 7.82 (s, 1H), 7.55 (m, 1H), 7.49 (m, 2H), 7.40 (m, 1H), 7.07 (m, 1H), 6.94 (m, 2H), 3.83 (m, 2H), 3.60 (d, J=8.0 Hz, 2H), 3.54 (d, J=8.0 Hz, 2H), 3.47 (s, 2H), 2.97 (t, J=10.4 Hz, 2H), 2.39 (m, 1H), 1.65 (m, 2H), 1.14 (m, 2H). The following compounds were prepared by a method analogous to that for Example 283.
R
Figure imgf000176_0001
Figure imgf000176_0002
Figure imgf000177_0001
Figure imgf000178_0001
Example 294. {l-{l-[3-Fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[3- (7H-pyrrolo[2,3-d]pyridin-4-yl)- -pyrazol-l-yl]azetidin-3-yl}acetonitrile
Figure imgf000178_0002
Step A: 4-Bromo- 1-{ [2-(trimethylsilyl)ethoxy]methyl}- lH-pyrrolo [2 , 3-b] pyridine
Figure imgf000178_0003
A solution of 4-bromo-lH-pyrrolo[2,3-b]pyridine (10.0 g, 0.0508 mol) in DMF (40 mL) was cooled under nitrogen to 0 °C. Sodium hydride (3.0 g, 0.075 mol) was added portionwise. The reaction was stirred for 1 hour. To this mixture, [2-
(trimethylsilyl)ethoxy]methyl chloride (10.8 mL, 0.061 mol) was added slowly. After being stirred at 0 °C for 1 hour, the reaction was quenched with water and extracted with EtOAc twice. The combined extracts were washed with water, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography, eluting with 0-25% EtOAc/hexanes to afford 15.7 g (94.5%) of the desired product as a yellowish oil. LC/MS found: 327.1, 329.1 (M+H)+.
Step B: 4-(lH-Pyrazol-4-yl)-l-{[ 2-(trimethylsilyl)ethoxy] methyl}- lH-pyrrolo[ 2, 3- b] pyridine
Figure imgf000179_0001
A mixture of 4-bromo-l-{[2-(trimethylsilyl)ethoxy]methyl}-lH-pyrrolo[2,3- b]pyridine (15.70 g, 47.97 mmol), l-(l-ethoxyethyl)-4-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)-lH-pyrazole (14.04 g, 52.77 mmol),
tetrakis(triphenylphosphine)palladium(0) (2.772 g, 2.398 mmol) and sodium carbonate (15.25 g, 143.9 mmol) in 1,4-dioxane (150 mL) and water (75 mL) was stirred at 110 °C for 1 hour. After being cooled to room temperature, the mixture was diluted with EtOAc, and washed with water and brine, dried over Na2S04 and concentrated. The residue was purified by flash chromatograpgy on silica gel eluting with 10-30% EtOAc/hexanes. The purified intermediate was dissolved in THF (21 mL), water (90 mL) and hydrogen chloride (75 mL, 240 mmol). The resulting suspension was stirred at room temperature for 2 hours. The mixture was adjusted to pH = 9-10 with 6 N NaOH. Hexanes (150 mL) were added. The solids formed was filtered and washed with water (3x) to provide 12.9 g (85 %) of 4-( 1 H-pyrazol-4-yl)- 1 - { [2-(trimethylsilyl)ethoxy]methyl} - 1 H-pyrrolo [2,3 - b]pyridine as a white solid. LC/MS found: 315.2 (M+H)+. Step C: tert-Butyl 3-(Cyanomethyl)-3-[4-(l-{[2-(trimethylsilyl)ethoxy]methyl}-lH- pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol-l-yl]azetidine-l-carboxylate
Figure imgf000180_0001
To a solution of 4-(lH-pyrazol-4-yl)-l-{[2-(trimethylsilyl)ethoxy]methyl}-lH- pyrrolo[2,3-b]pyridine (230 mg, 0.73 mmol) and tert-bvXy\ 3-(cyanomethylene)azetidine- 1-carboxylate (142 mg, 0.73 mmol) in acetonitrile (5 mL) was added 1,8- diazabicyclo[5.4.0]undec-7-ene (0.11 mL, 0.73 mmol). After being stirred for 5 minutes at room temperature, the mixture became a solution. LC-MS indicated that the reaction was complete. Acetonitrile was evaporated and ethyl acetate was added. The mixture was washed with 1 N HC1, brine, dried over sodium sulfate, filtered and concentrated. The residue was purified by silica gel chromatography (0-80% EtOAc/hexanes) to give 341 mg of tert-butyl 3-(cyanomethyl)-3-[4-(l-{[2-(trimethylsilyl)ethoxy]methyl}-lH- pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol-l-yl]azetidine-l-carboxylate as a colorless oil. LC/MS found: 509.2 (M+H)+.
Step D: {3-[ 4-(l-{ [2-(Trimethylsilyl)ethoxy] methyl}- lH-pyrrolo [2, 3-b] pyridin pyrazol- 1 -yl azetidin-3-yl} acetonitrile '2[HCl]
Figure imgf000180_0002
A solution of tert-butyl 3-(cyanomethyl)-3-[4-(l-{[2- (trimethylsilyl)ethoxy]methyl} - 1 H-pyrrolo [2,3 -b]pyridin-4-yl)- 1 H-pyrazol- 1 - yl]azetidine-l-carboxylate (341 mg, 0.67 mmol) in THF (5 mL) and methanol (5 mL) was added a 4.0 M solution of hydrogen chloride in 1,4-dioxane (5 mL, 20 mmol). The mixture was stirred at room temperature for 2 hours and concentrated to give 347 mg (100%) of {3-[4-(l-{[2-(trimethylsilyl)ethoxy]methyl}-lH-pyrrolo[2,3-b]pyridin-4-yl)- 1 H-pyrazol- l-yl]azetidin-3-yl}acetonitrile»2 [HCl] as a yellowish solid. LC/MS found: 409.2 (M+H)+.
Step E: {l-Piperidin-4-yl-3-[ 4-(l-{[2-(trimethylsilyl)ethoxy]methyl}-lH-pyrrolo[2,3- bj 'pyridin-4-yl)- 1 H-pyrazol- 1-ylj ' azetidin-3-yl}acetonitrile trihydrochloride
Figure imgf000181_0001
To a mixture of {3-[4-(l-{[2-(trimethylsilyl)ethoxy]methyl}-lH-pyrrolo[2,3- b]pyridin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile»2[HCl] (347 mg, 0.70 mmol), tert-butyl 4-oxo-l-piperidinecarboxylate (134 mg, 0.70 mmol) and N,N- diisopropylethylamine (0.467 mL, 2.68 mmol) in THF (10.0 mL) was added sodium triacetoxyborohydride (284 mg, 1.34 mmol). The mixture was stirred at room
temperature for 2 hours and quenched with brine. The resulting solution was extracted with EtOAc (2 times). The combined extracts were washed with water, brine and dried over Na2S04. After filtration and evaporation, the residue was purified by flash chromatography on silica gel eluting with 50-100% EtOAc/hexanes. The purified intermediate (MS: [M+Hf = 592.3) was dissolved in THF (6 mL). To the solution was added a 4.0 M solution of HCl in 1,4-dioxane (6 mL, 24 mmol). The mixture was stirred at room temperature for 2 hours and concentrated to give 260 mg of {l-piperidin-4-yl-3- [4-( 1 - { [2-(trimethylsilyl)ethoxy]methyl} - 1 H-pyrrolo [2,3 -b]pyridin-4-yl)- 1 H-pyrazol- 1 - yl]azetidin-3-yl}acetonitrile trihydrochloride as a yellowish solid. LC/MS found: 492.0 (M+H)+. 1H NMR (400 MHz, DMSO-d6), δ 9.45 (s, 1H), 8.96 (s, 1H), 8.73 (s, 1H), 8.35 (d, 1H), 8.46 (s, 1H), 8.33 (d, 1H), 7.81 (d, 1H), 7.50 (d, 1H), 7.10 (m, 1H), 5.69 (s, 2H), 4.93 (d, 2H), 4.54 (d, 2H), 3.75-3.60 (m, 1H), 3.55 (s, 2H), 3.55 (s, 2H), 3.52 (t, 2H), 3.49-3.37 (m, 2H), 2.81 (m, 2H), 2.12 (d, 2H), 1.80 (m, 2H), 0.82 (t, 2H), -0.11 (s, 9H).
Step F: {l-{l-[ 3-Fluoro-2- ( trifluoromethyl) ison icotinoyl] piperidin-4-yl} -3-[4-(lH- pyrrolo[2, 3-b]pyridin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile
A mixture of {l-piperidin-4-yl-3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H- pyrrolo[2,3-d]pyridin-4-yl)- lH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile trihydrochloride (1.22 g, 2.03 mmol), 3-fluoro-2-(trifluoromethyl)isonicotinic acid (460 mg, 2.2 mmol), benzotriazol-l-yloxytris(dimethylamino)phosphonium hexafluorophosphate (1.07 g, 2.42 mmol) and triethylamine (2.0 mL, 14 mmol) in DMF (10.0 mL) was stirred at room temperature overnight. LS-MS showed the reaction was complete. EtOAc (60 mL) and saturated NaHC03 aqueous solution (60 mL) were added to the reaction mixture. After stirring at room temperature for 10 minutes, the organic phase was separated and the aqueous layer was extracted with EtOAc three times. The combined extracts were washed with brine, dried over anhydrous Na2S04, filtered and evaporated under reduced pressure. The residue was purified by flash chromatography to provide {l-{l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7-{[2- (trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrrol-l- yl]azetidin-3-yl} acetonitrile as a white powder. The powder was dissolved in 10 mL of TFA/DCM (1 : 1). After being stirred at room temperature for 2 hours, the solution was concentrated. The residue was dissolved in 10 mL of a solution of 20%
ethylenediamine/MeOH. After being stirred at room temperature for 2 hours, the solution was concentrated. Purification by HPLC (method B) provided the final compound {1-{1- [3 -fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl} -3 - [3 -(7H-pyrrolo [2,3 - d]pyridin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile. LC-MS: 553.3 (M+H)+. 1H NMR (400 MHz, CDC13): δ 9.42 (s, 1H), 8.82 (s, 1H), 8.57 (d, J=4.7 Hz, 1H), 8.29 (d, J=5.2 Hz, IH), 8.06 (d, J=5.8 Hz, IH), 7.53 (t, J=4.5 Hz, IH), 7.38 (dd, Ji=3.6 Hz, J2=2.4 Hz, IH), 7.16 (d, J=5.1 Hz, IH), 6.69 (dd, Ji=3.7 Hz, J2=2.1 Hz, IH), 4.21 (m, IH), 3.76 (m, 2H), 3.63 (dd, Ji=7.4 Hz, J2=5.8 Hz, 2H), 3.46(m, 2H), 3.35 (s, 2H), 3.1 l(m, IH), 2.56 (m, IH), 1.84 (m, IH), 1.71(m, IH), 1.49 (m, IH), 1.40 (m, IH).
The following compounds were prepared by a method analogous to that for Example 294.
Figure imgf000183_0001
Figure imgf000184_0001
H-pyrazo- -y azet n- -y acetontr e
Figure imgf000184_0002
Example 303. 4-{3-(Cyanomethyl)-3-[4-(lH-pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol- l-yl]azetidin-l-yl}-N-[4-fluoro-2-(trifluoromethyl)phenyl]piperidine-l-carboxamide
Figure imgf000185_0001
To a solution of {l-piperidin-4-yl-3-[4-(l-{[2-(trimethylsilyl)ethoxy]methyl}-lH- pyrrolo[2,3-b]pyridin-4-yl)- IH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile trihydrochloride (40 mg, 0.08 mmol) in THF (8 mL) were added triethylamine (0.025 g, 0.24 mmol), and 4-fluoro-l-isocyanato-2-(trifluoromethyl)benzene (18 mg, 0.086 mmol). The mixture was stirred at room temperature for one hour and concentrated. Purification with combi-fiash chromatography using 30-100% EtOAc/hexanes gave a powder product. LC-MS: 697.1
(M+H)+.
The above solid was dissolved in a 50 M solution of trifluoroacetic acid in methylene chloride (2 mL, 100 mmol). After being stirred at room temperature for one hour, the solvents were removed. The residue was dissolved in methanol (2 mL) and ethylenediamine (0.024 g, 0.40 mmol). After being stirred at room temperature for one hour, the solution was purified by HPLC (method B) to give 4-{3-(cyanomethyl)-3-[4- ( 1 H-pyrrolo[2,3-b]pyridin-4-yl)- 1 H-pyrazol- 1 -yljazetidin- 1 -yl} -N-[4-fluoro-2- (trifluoromethyl)phenyl]piperidine-l-carboxamide as a white powder. LC-MS: 567.2
(M+H). 1H NMR (400 MHz, DMSO-d6): δ 11.70 (s, 1H), 8.68 (s, 1H), 8.26 (m, 1H), 8.23 (s, 1H), 8.18 (d, J=4.8 Hz, 1H), 7.57 (dd, Ji=7.2 Hz, J2=3.2 Hz, 1H), 7.50(m, 2H), 7.41(m, 1H), 7.32 (d, J=7.2 Hz, 1H), 6.86 (d, J=3.6 Hz, 1H), 3.86 (m, 2H), 3.73 (d, J=8.0 Hz, 2H), 3.58 (d, J=8.4 Hz, 2H), 3.52 (s, 2H), 2.99 (t, J=10.6 Hz, 2H), 2.43 (m, 1H), 1.68 (m, 2H), 1.12 (m, 2H). The following compounds were prepared by a method analogous to that for Example 303.
Figure imgf000186_0001
Figure imgf000186_0002
Figure imgf000187_0001
1H), 8.12 (m, 2H), 7.46 (m, 1H), 7.38 (m, 2H), 7.27 (d, 1H), 7.21 (t, 1H), 7.04 (t, 1H), 6.81 (m, 1H), 3.83 (m, 2H), 3.68 (d, 2H), 3.53 (d, 2H), 3.48 (m, 2H), 2.99 (t, 2H), 2.38 (m, 1H), 1.64 (m, 2H), 1.14 (m, 2H).
1H NMR (400 MHz, DMSO-d6) of Example 312: δ 11.65 (br, 1H), 8.65 (br, 1H), 8.62 (s, 1H), 8.21 (s, 1H), 8.12 (m, 1H), 7.67 (m, 2H), 7.51 (d, 1H), 7.46 (m, 1H), 7.27 (d, 1H), 6.81 (m, 1H), 3.83 (m, 2H), 3.68 (d, 2H), 3.53 (d, 2H), 3.48 (m, 2H), 2.98 (t, 2H), 2.39 (m, 1H), 1.64 (m, 2H), 1.14 (m, 2H).
1H NMR (400 MHz, DMSO-d6) of Example 313: δ 11.65 (br, 1H), 8.63 (s, 1H), 8.36 (br, 1H), 8.21 (s, 1H), 8.12 (m, 1H), 7.95 (s, 1H), 7.75 (d, 1H), 7.64 (m, 1H), 7.46 (m, 1H), 7.27 (d, 1H), 6.81 (m, 1H), 3.83 (m, 2H), 3.68 (d, 2H), 3.53 (d, 2H), 3.48 (m, 2H), 3.00 (t, 2H), 2.39 (m, 1H), 1.66 (m, 2H), 1.15 (m, 2H).
Example 314. (3-[4-(5-Fluoro-lH-pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol-l-yl]-l-{l- [3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}azetidin-3-yl)acetonitrile
Figure imgf000188_0001
Step A: 5-fluoro-4-iodo- 1-{ [2-(trimethylsilyl)ethoxyj 'methyl}- lH-pyrrolo [2, 3-bj 'pyridine
Figure imgf000188_0002
To a solution of 5-fluoro-4-iodo-lH-pyrrolo[2,3-b]pyridine (5.0 g, 0.019 mol) in DMF (30.0 mL) cooled at 0 °C under nitrogen was portionwise added sodium hydride (1.13 g, 0.0282 mol). The reaction was stirred for 1 hour. To the mixture was slowly added [P-(trimethylsilyl)ethoxy]methyl chloride (4.05 mL, 0.0229 mol). The reaction was stirred at 0 °C for 1 hour and quenched with water. The resulting solution was extracted with EtOAc (2 times). The combined extracts were washed with water, brine, dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel chromatography, eluting with 0-25% EtOAc/hexanes to afford 7.1 g (95%) of 5-fluoro-4-iodo- 1 - {[2-(trimethylsilyl)ethoxy]methyl} - lH-pyrrolo[2,3-b]pyridine as a yellowish oil. LC/MS found: 393.0 (M+H)+.
Step B: 4-(lH-Pyrazol-4-yl)-5-fluoro-l-{[2-(trimethylsilyl)ethoxy]methyl}-lH- pyrrolof 2, 3-b ] pyridine
Figure imgf000189_0001
A mixture of 5-fluoro-4-iodo-l-{[2-(trimethylsilyl)ethoxy]methyl}-lH- pyrrolo[2,3-b]pyridine (7.20 g, 18.4 mmol), l-(l-ethoxyethyl)-4-(4,4,5,5-tetramethyl- l ,3,2-dioxaborolan-2-yl)-lH-pyrazole (5.36 g, 20.1 mmol),
tetrakis(triphenylphosphine)palladium(0) (1.06 g, 0.918 mmol) and sodium carbonate (5.84 g, 55.1 mmol) in 1 ,4-dioxane (50 mL) and water (25 mL) was stirred at 1 10 °C under nitrogen for 1 hour. After being cooled to room temperature, the mixture was diluted with EtOAc, washed with water and brine, dried over Na2S04 and concentrated under reduced pressure. The residue was purified by flash chromatograpgy on silica gel eluting with 10-30% EtOAc/hexanes. The purifed intermediate was added into a mixture solution of THF (8.0 mL), water (30 mL) and hydrogen chloride (30 mL, 100 mmol). The resulting suspension was stirred at room temperature for 2 hours. The mixture was adjusted to pH = 9-10 with 6 N NaOH and extracted with EtOAc (2x). The combined extracts were washed with water, brine and dried over anhydrous Na2S04. After filtration and concentration, the residue was added into a mixture solvents of hexane and EtOAc (9/1 , 50 mL). The solid formed was filtered to give 4.2 g (69%>) of 4-(lH-pyrazol-4-yl)-5- fluoro-l-{[2-(trimethylsilyl)ethoxy]methyl}-lH-pyrrolo[2,3-b]pyridine as a light green solid. LC/MS found: 333.2 (M+H)+.
Step C: {3- [4-(5-Fluoro- 1-{ [2-(trimethylsilyl)ethoxy] 'methyl}- lH-pyrrolo [2, 3-bj 'pyridin- 4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile'2[HCl]
Figure imgf000190_0001
To a solution of 5-fluoro-4-(lH-pyrazol-4-yl)-l-{[2- (trimethylsilyl)ethoxy]methyl}-lH-pyrrolo[2,3-b]pyridine (2.00 g, 6.02 mmol) and tert- butyl 3-(cyanomethylene)azetidine-l-carboxylate (1.168 g, 6.02 mmol) in acetonitrile (20 mL) was added l,8-diazabicyclo[5.4.0]undec-7-ene (0.8996 mL, 6.02 mmol). The mixture was stirred at room temperature overnight and concentrated. The residue was purified by silica gel chromatography (0-80% EtOAc/hexanes) to give 3.05 g (96.3%>) of tert-butyl 3 -(cyanomethyl)-3 - [4-(5 -fluoro- 1 - { [2-(trimethylsily l)ethoxy]methyl} - 1 H- pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol-l-yl]azetidine-l-carboxylate as a colorless oil. LC/MS found: 527.3 (M+H)+.
To a solution of tert-bvXy\ 3-(cyanomethyl)-3-[4-(5-fluoro-l-{[2- (trimethylsilyl)ethoxy]methyl} - 1 H-pyrrolo [2,3 -b]pyridin-4-yl)- 1 H-pyrazol- 1 - yl]azetidine-l-carboxylate (3.05 g, 5.79 mmol) in THF (40 mL) was added a 4.0 M solution of hydrogen chloride in 1,4-dioxane (70 mL, 280 mmol). The mixture was stirred at room temperature for 2 hours and concentrated to give 3.08 g (99.2%) of {3-[4- (5 -fluoro- 1 - { [2-(trimethylsilyl)ethoxy]methyl} - 1 H-pyrrolo [2,3 -b]pyridin-4-yl)- 1 H- pyrazol-l-yl]azetidin-3-yl}acetonitrile»2[HCl] as a yellowish solid. LC/MS found: 427.2 (M+H)+.
Step D: {3 -[4- (5 -Fluoro- 1 -{[2-(trimethylsilyl)ethoxy]methyl}-l H-pyrrolo [2, 3-bJpyridin- 4-yl)-lH-pyrazol-l-yl]-l-piperidin-4-ylazetidin-3-yl}acetonitrile'3[HCl]
Figure imgf000191_0001
To a mixture of {3-[4-(5-fluoro-l-{[2-(trimethylsilyl)ethoxy]methyl}-lH- pyrrolo[2,3-b]pyridin-4-yl)- lH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile»2[HCl] (3.10 g,
5.78 mmol), tert-butyl 4-oxo-l-piperidinecarboxylate (1.152 g, 5.784 mmol) and N,N- diisopropylethylamine (3.022 mL, 17.35 mmol) in THF (100.0 mL) was added sodium triacetoxyborohydride (2.452 g, 11.57 mmol). The mixture was stirred at room temperature for 2 hours and quenched with brine. The resulting solution was extracted with EtOAc (2 times). The combined extracts were washed with water, brine and dried over Na2SC"4. After filtration and evaporation, the residue was purified by flash chromatography on silica gel eluting with 50-100% EtOAc/hexanes. The purified intermediate ([M+H]+ = 610.3) was dissolved in THF (50 mL). To the solution at 10 °C was added a 4.0 M solution of hydrogen chloride in 1,4-dioxane (50.0 mL, 200 mmol). The mixture was stirred at room temperature for 2 hours and concentrated to give 3.57 g of {3-[4-(5-fluoro-l-{[2-(trimethylsilyl)ethoxy]methyl}-lH-pyrrolo[2,3-b]pyridin-4-yl)- lH-pyrazol-l-yl]-l-piperidin-4-ylazetidin-3-yl}acetonitrile»3[HCl] as an off-white solid. LC/MS found: 510.3 (M+H)+. 1H NMR (400 MHz, DMSO-d6), δ 9.41 (s, 1H), 8.81 (s, 1H), 8.72 (s, 1H), 8.35 (d, 1H), 8.32 (s, 1H), 7.85 (d, 1H), 7.00 (d, 1H), 5.63 (s, 2H), 4.93 (d, 2H), 4.55 (d, 2H), 3.78-3.60 (m, 1H), 3.55 (s, 2H), 3.51 (t, 2H), 3.47-3.37 (m, 2H),
2.79 (m, 2H), 2.11 (m, 2H), 1.80 (m, 2H), 0.81 (t, 2H), -0.12 (s, 9H).
Step E: (3-[4-(5-Fluoro-lH-pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol-l-yl]-l-{l-[3-fluoro- 2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}azetidin- A solution of 3-fluoro-2-(trifluoromethyl)isonicotinic acid (70.0 mg, 0.335 mmol), {3-[4-(5-fluoro-l-{[2-(trimethylsilyl)ethoxy]methyl}-lH-pyrrolo[2,3-b]pyridin- 4-yl)-lH-pyrazol-l-yl]-l-piperidin-4-ylazetidin-3-yl}acetonitrile»3[HCl] (207 mg, 0.335 mmol), benzotriazol-l-yloxytris(dimethylamino)phosphonium hexafluorophosphate (148 mg, 0.335 mmol) and triethylamine (0.234 mL, 1.68 mmol) in methylene chloride (2 mL) was stirred at room temperature for 1 hour. To the mixture was added trifluoroacetic acid (2 mL, 20 mmol). The resulting solution was stirred at room temperature for 1 hour and concentrated. The residue was dissolved in methanol (2 mL) and ethylenediamine (0.5 mL, 7 mmol). After being stirred at room temperature for 2 hours, the mixture was purified by HPLC (method B) to give 11.2 mg of the title compound as a white solid.
LC/MS found: 571.3 (M+H)+. 1H NMR (400 MHz, CDC13): δ 11.85 (brs, 1H), 8.65 (d, J = 4.4 Hz, 1H), 8.59 (s, 1H), 8.23 (d, J = 3.2 Hz, 1H), 8.20 (s, 1H), 7.89 (s, 1H), 7.61 (s, 1H), 6.85 (s, 1H), 4.04 (m,l H), 3.72 (m, 2H), 3.58 (m, 2H), 3.53 (m, 2H), 3.53 (m, 2H), 3.40 (m, 1H), 3.25 (m, 1H), 3.06 (t, J = 9.2 Hz, 1H), 2.54 (m, 1H), 1.75 (m, 1H), 1.62 (m, 1H), 1.26 (m, 1H), 1,21 (m, 1H).
The following compounds were prepared following the procedures described for Example 314.
Figure imgf000192_0001
Figure imgf000192_0002
Figure imgf000193_0001
fluorobenzonitrile
Figure imgf000194_0001
y aceton t e
1H NMR (300 MHz, DMSO-d6) of Example 316: δ 11.81 (brs,lH), 8.55 (s, IH), 8.18 (d, J = 3.00 Hz, IH), 8.15 (s, IH), 7.88 (d, J = 8.1 Hz, IH), 7.70 (s, IH), 7.61 (d, J = 7.80 Hz, IH), 7.56 (d, J = 3.30 Hz, IH), 6.81 (d, J = 3.30 Hz, IH), 3.97 (m, IH), 3.68 (m, 2H), 3.52 (m, 2H), 3.47 (s, 2H), 3.27 (m, IH), 3.11 (m, IH), 2.99 (m, IH), 2.43 (m, IH), 1.68 (m, IH), 1.56 (m, IH), 1.20 (m, 2H).
1H NMR (300 MHz, DMSO-d6) of Example 317: δ 11.81 (brs, IH), 8.54 (s, IH), 8.18 (d, J = 3.30 Hz, IH), 8.15 (d, J = 2.1 Hz, IH), 7.93 (dd, Ji = 9.30 Hz, J2 = 1.20 Hz, IH), 7.72 (dd, Ji = 7.80 Hz, J2 = 1.20 Hz, IH), 7.56 (m, 2H), 6.80 (d, J = 3.30 Hz, IH), 4.02 (m, IH), 3.67 (m, 2H), 3.53 (m, 2H), 3.48 (s, 2H), 3.16 (m, 2H), 2.96 (m, IH), 2.48 (m, IH), 1.70 (m, IH), 1.55 (m, IH), 1.15 (m, 2H).
1H NMR (300 MHz, DMSO-d6) of Example 318: δ 11.81 (brs, IH), 8.54 (s, IH), 8.18 (d, J = 3.30 Hz, IH), 8.15 (d, J = 2.1 Hz, IH), 7.95 (dd, Ji = 6.30 Hz, J2 = 2.10 Hz, IH), 7.74 (m, IH), 7.54 (m, 2H), 6.81 (m, IH), 4.00 (m, IH), 3.68 (m, 2H), 3.52 (m, 2H), 3.47 (s, 2H), 3.01 (m, 3H), 2.45 (m, IH), 1.64 (m, 2H), 1.20 (m, 2H).
1H NMR (300 MHz, DMSO-d6) of Example 319: δ 11.81 (brs, IH), 8.54 (s, IH), 8.52 (d, J = 2.70 Hz, IH), 8.18 (d, J = 3.60, IH) 8.15 (d, J = 1.80 Hz, IH), 7.78 (m, IH), 7.58 (m, 2H), 6.81 (d, J = 3.60 Hz, IH), 4.04 (m, IH), 3.67 (m, 2H), 3.52 (m, 2H), 3.50 (s, 2H), 3.08 (m, 3H), 2.46 (m, IH), 1.70 (m, IH), 1.58 (m, IH), 1.16 (m, 2H).
1H NMR (400 MHz, DMSO-d6) of Example 321 : δ 11.86 (brs, IH), 8.65 (s, 2H), 8.59 (s, IH), 8.23 (d, J = 3.20 Hz, IH), 8.20 (d, J = 2.0 Hz, IH), 7.61 (d, J = 3.20 Hz, IH), 6.86 (d, J = 3.60 Hz, IH), 4.07 (m, IH), 3.72 (m, 2H), 3.58 (m, 2H), 3.54 (s, 2H), 3.42 (m,lH), 3.28 (m, IH), 3.10 (m, IH), 2.54 (m, IH), 1.76 (m, IH), 1.64 (m, IH), 1.20 (m, 2H).
1H NMR (400 MHz, DMSO-d6) of Example 322: δ 11.86 (brs, IH), 8.59 (s, IH), 8.23 (d, J = 3.60 Hz, IH), 8.20 (d, J = 2.00 Hz, IH), 7.61 (d, J = 3.20 Hz, IH), 7.14 (dd, Ji = 11.60 Hz, J2 = 1.60 Hz, IH), 7.00 (d, J = 8.00 Hz, IH), 6.92 (t, J = 8.00 Hz, IH), 6.86 (d, J = 3.60 Hz, IH), 3.80 (m,l H), 3.72 (m, 2H), 3.56 (m, 2H), 3.52 (s, 2H), 3.08 (m, 3H), 2.51 (m IH), 1.67 (m, 2H), 1.19 (m, 2H). 1H NMR (300 MHz, DMSO-d6) of Example 323: δ 11.82 (brs, 1H), 8.54 (, s, 1H), 8.18 (d, J = 3.60 Hz, 1H), 8.15 (d, J = 1.80 Hz, 1H), 7.56 (d, J = 3.90 Hz, 1H), 7.09 (t, J = 8.40 Hz, 1H), 6.81 (d, J = 3.60 Hz, 1H), 6.54 (m, 2H),4.00 (m, 1H), 3.66 (m, 2H), 3.51 (m, 2H), 3.48 (s, 2H), 3.35 (2H), 3.01 (m, 1H), 2.47 (m, 1H), 1.61 (m, 2H), 1.11 (m, 2H).
1H NMR (300 MHz, DMSO-d6) of Example 325: δ 11.81 (brs, 1H), 8.54 (s, 1H), 8.18 (d, J = 3.30 Hz, 1H), 8.15 (d, J = 2.1 Hz, 1H), 7.94 (t, J = 6.90 Hz, 1H), 7.56 (m, 2H), 7.34 (dd, Ji = 8.10 Hz, = 1.20 Hz, 1H), 6.81 (m, 1H),3.99 (m, 1H), 3.68 (m, 2H), 3.52 (m, 2H), 3.47 (s, 2H), 3.05 (m, 3H), 2.47 (m, 1H), 1.68 (m, 1H), 1.16 (m, 1H), 1.18 (m, 2H).
1H NMR (300 MHz, DMSO-d6) of Example 326: δ 11.82 (brs, 1H), 8.88 (s, 1H), 8.54 (s, 1H), 8.18 (d, J = 3.60 Hz, 1H), 8.15 (d, J = 2.10 Hz, 1H), 8.06 (d, J = 13.20 Hz, 1H), 7.56 (s, 1H), 6.81 (d, J = 3.30 Hz, 1H), 4.02 (m,lH), 3.68 (m, 2H), 3.50 (m, 4H), 3.19 (m, 2H), 2.94 (m, 1H), 2.48 (m, 1H), 1.67 (m, 2H), 1.20 (m, 2H).
1H NMR (300 MHz, DMSO-d6) of Example 327: δ 11.81 (brs, 1H), 8.54 (s, 1H), 8.18 (d, J = 3.30 Hz, 1H), 8.15 (d, J = 1.80 Hz, 1H), 7.56 (d, J = 3.30 Hz, 1H), 6.81 (d, J = 3.60 Hz, 1H), 3.93 (m, 1H), 3.75 (m, 1H), 3.66 (m, 2H), 3.52 (m, 2H), 3.48 (s, 2H), 3.09 (m, 1H), 2.83 (m, 1H), 2.72 (m, 1H), 2.42 (m, lH), 1.90 (m, 4H), 1.61 (m, 6H), 1.06 (m, 2H).
Example 328. 4- {3-(Cyanomethyl)-3- [4-(5-fluoro- IH-pyrrolo [2,3-b] pyridin-4-yl)- lH-pyrazol-l-yl]azetidin-l-yl}-N-[4-fluoro-2-(trifluoromethyl)phenyl]piperidine-l- carboxamide
Figure imgf000195_0001
To a mixture of {3-[4-(5-fluoro-l-{[2-(trimethylsilyl)ethoxy]methyl}-lH- pyrrolo[2,3-b]pyridin-4-yl)- lH-pyrazol- 1 -yl]- 1 -piperidin-4-ylazetidin-3- l}acetonitrile»3[HCl] (70.0 mg, 0.113 mmol) and triethylamine (41.3 uL, 0.296 mmol) in THF (4 mL) was added 4-fluoro-l-isocyanato-2-(trifluoromethyl)benzene (23.1 mg, 0.113 mmol). The mixture was stirred at room temperature for 1 hour and concentrated. The residue was diluted with acetonitrile (2 mL) and water (2 mL). The mixture was submitted to purification by HPLC to give 34 mg (49%) of the desired intermediate. LC- MS found: 715.3 (M+H)+.
The purified intermediate was dissolved in methylene chloride (1 mL) and trifluoroacetic acid (1 mL, 10 mmol). The solution was stirred at room temperature for 1 hour and concentrated. The residue was treated with methanol (1 mL) and
ethylenediamine (0.2 mL, 3 mmol). The solution was stirred at room temperature for 1 hour and purified by HPLC (method B) to give the title compound. LC-MS found: 585.1 (M+H)+. 1H NMR (300 MHz, DMSO-d6): δ 11.86 (brs, 1H), 8.60 (s, 1H), 8.24 (d, J = 3.30 Hz, 1H), 8.21 (d, J = 2.40 Hz, 1H), 7.61 (t, J = 3.30 Hz, 1H), 7.49 (m, 3H), 6.86 (m, 1H), 3.83 (m, 2H), 3.72 (m, 2H), 3.58 (m, 2H), 3.54 (s, 2H), 2.97 (m, 2H), 2.44 (m, 1H), 1.66 (m, 2H), 1.13 (m, 2H).
The following compounds were prepared following the procedures described for Example 328.
Figure imgf000196_0001
Figure imgf000196_0002
Figure imgf000197_0001
Example 332. 4-[l-(3-(Cyanomethyl)-l-{l-[3-fluoro-2-
(trifluoromethyl)isonicotinoyl] piperidin-4-yl} azetidin-3-yl)- lH-pyrazol-4-yl] - 1H- pyrrolo [2,3-b] pyridine-5-carbonitrile
Figure imgf000198_0001
Step A: 4-Chloro-5-cyano-l-{[ 2 -(trimethylsilyl)ethoxy] methyl} -lH-pyrrolo[ 2, 3- b] pyridine
Figure imgf000198_0002
To a suspension of sodium hydride (1.8 g, 45.1 mmol) in N,N-dimethylacetamide (10 mL) at -5 °C (ice/salt bath) was added a dark solution of 4-chloro-5-cyano- pyrrolo[2,3-d]pyridine (6.0 g, 39 mmol) in N,N-dimethylacetamide (10 mL) slowly. The flask and addition funnel were rinsed with N,N-dimethylacetamide (5 mL). A large amount of gas was evolved immediately. The mixture turned to a slightly cloudy orange mixture and was stirred at 0 °C for 1 hour to give a light brown turbid mixture. To the mixture was slowly added [ -(trimethylsilyl)ethoxy]methyl chloride (7.6 g, 45 mmol). The reaction was stirred at 0 °C for 1 hour and quenched by addition of 12 mL of H20. After the reaction was quenched, H20 (120 mL) was added. It was followed by MTBE (120 mL). The mixture was stirred for 10 minutes. The organic layer was separated. The aqueous layer was extracted with another portion of MTBE (120 mL). The organic extracts were combined, washed with brine, dried over sodium sulfate and concentrated under reduced pressure to give 10 g (95%) of the crude product 4-chloro-5-cyano-l-{[2- (trimethylsilyl)ethoxy]methyl}-lH-pyrrolo[2,3-b]pyridine as a dark oil. LC-MS: 308.1 (M+H)+. The crude product was carried over to the next reaction without further purification. Step B: 4-(lH-Pyrazol-4-yl)-5-cyano-l-{[2-(trimethylsilyl)ethoxy]methyl}-lH- pyrrolof 2, 3-b ] pyridine
Figure imgf000199_0001
A 250 mL round bottom flask was charged with 4-chloro-5-cyano-l-{[2-
(trimethylsilyl)ethoxy]methyl}-lH-pyrrolo[2,3-d]pyridine (5.00 g, 17.6 mmol), 1-butanol (25.0 mL), l-(l-ethoxyethyl)-4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-lH- pyrazole (7.06 g, 26.4 mmol), water (25.0 mL) and potassium carbonate (6.17 g, 44.08 mmol). This solution was degased 4 times with filling with nitrogen each time. To it was added tetrakis(triphenylphosphine)palladium(0) (2.071 g, 1.773 mmol). The solution was degassed 4 times, filling with nitrogen each time, and stirred at 100 °C for 3 hours. After being cooled to room temperature, the mixture was filtered through a bed of celite and the celite was rinsed with ethyl acetate (42 mL). The filtrate was combined and the organic layer was separated. The aqueous layer was extracted with ethyl acetate. The combined extracts were dried over anhydrous Na2S04, filtered, evaporated under reduced pressure to give an oil residue which was purified by combiflash column to generate 3.8 g (53%) of the desired intermediate. LC-MS: 412.2 (M+H)+.
A mixture of 3.8 g of the above intermediate in 20 mL of 2 N HC1 aqueous solution and 20 mL of CHCI3 was stirred ar room temperature over weekend. The organic layer was separated and the aqueous phase was extracted with EtOAc (3x50 mL). The combined extracts were dried over anhydrous Na2S04, filtered, evaporated under reduced pressure to give 2.9 g (97%) of 4-(lH-pyrazol-4-yl)-5-cyano-l-{[2- (trimethylsilyl)ethoxy]methyl}-lH-pyrrolo[2,3-b]pyridine. LC-MS: 340.2 (M+H)+.
Step C: 4-{l-[3-(Cyanomethyl)azetidin-3-yl]-lH-pyrazol-4-yl}-l-{[2- (trimethylsilyl)ethoxyj 1 methyl} -IH-pyrrolo '[2, 3-bj 1 pyridine-5-carbonitrile'2 [HCl]
Figure imgf000200_0001
To a solution of 4-(lH-pyrazol-4-yl)-l-{[2-(trimethylsilyl)ethoxy]methyl}-lH- pyrrolo[2,3-b]pyridine-5-carbonitrile (2.26 g, 6.66 mmol) and tert-butyl 3- (cyanomethylene)azetidine-l-carboxylate (1.293 g, 6.66 mmol) in acetonitrile (40 mL) was added l,8-diazabicyclo[5.4.0]undec-7-ene (0.996 mL, 6.66 mmol). The mixture was stirred at room temperature overnight and concentrated. Purification by silica gel chromatography (0-80% EtOAc/hexanes) gave 2.20 g (62%) of the intermediate tert- butyl 3 -(cyanomethyl)-3 - [4-(5 -cyano- 1 - { [2-(trimethylsilyl)ethoxy]methyl} - 1 H- pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol-l-yl]azetidine-l-carboxylate as a colorless oil. LC-MS found: 534.3 (M+H)+.
To a solution of the above oil intermediate (2.20 g, 4.12 mmol) in THF (40 mL) was added a 4.0 M solution of hydrogen chloride in 1,4-dioxane (70 mL, 280 mmol). The mixture was stirred at room temperature for 2 hours and concentrated to give 2.23 g (99.6%) of the desired product as a yellowish solid. LC/MS found: 434.2 (M+H)+.
Step D: 4-{l-[3-(Cyanomethyl)-l-piperidin-4-ylazetidin-3-yl]-lH-pyrazol-4-yl}-l-{[2- (trimethylsilyl)ethoxy]methyl}-lH-pyrrolo[2,3-b]pyridine-5-ca^
Figure imgf000200_0002
To a mixture of 4-{l-[3-(cyanomethyl)azetidin-3-yl]-lH-pyrazol-4-yl}-l-{[2- (trimethylsilyl)ethoxy]methyl} - 1 H-pyrrolo [2,3 -b]pyridine-5 -carbonitrile»2 [HC1] (2.0 g, 3.68 mmol), tert-butyl 4-oxo-l-piperidinecarboxylate (0.734 g, 3.68 mmol) and N,N- diisopropylethylamine (3.21 mL, 18.4 mmol) in THF (70.0 mL) was added sodium triacetoxyborohydride(l .56 g, 7.37 mmol). The mixture was stirred at room temperature for 2 hours and quenched with brine. The resulting solution was extracted with EtOAc (2 times). The combined extracts were washed with water, brine and dried over Na2S04. After filtration and evaporation, the residue was purified by flash chromatography on silica gel eluting with 50-100% EtOAc/Hexanes. The purified intermediate was dissolved in THF (30 mL). To the solution at 10 °C was added a 4.0 M solution of hydrogen chloride in 1,4-dioxane (30.0 mL, 1.20 mmol). The mixture was stirred at room temperature for 2 hours and concentrated to give 2.01 g (87.2%) of the desired product as an off-white solid. LC/MS found: 517.3 (M+H)+. 1H NMR (400 MHz, DMSO-d6): δ 9.29 (s, 1H), 8.88 (s, 1H), 8.74 (s, 1H), 8.37 (s, 1H), 7.97 (d, 1H), 6.97 (s, 1H), 5.69 (s, 2H), 5.94 (m, 2H), 4.55 (m, 2H), 3.73-3.56 (m, 2H), 3.63 (t, 2H), 3.52 (m, 2H), 3.47-3.35 (m, 3H), 2.81 (m, 2H), 2.10 (m, 2H), 1.75 (m, 2H), 0.82 (t, 2H), -0.11 (s, 9H).
Step E: 4-f l-(3-(Cyanomethyl)-l-{l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin- 4-yl}azetidin-3-yl)-lH^yrazol-4-yl]-lH^yrrolo[2,3-b]pyridine-5-carbonitrile
A mixture of 3-fluoro-2-(trifluoromethyl)isonicotinic acid (70.0mg, 0.335mmol),
4- { 1 -[3-(cyanomethyl)- 1 -piperidin-4-ylazetidin-3-yl]-lH-pyrazol-4-yl} - 1 - { [2- (trimethylsilyl)ethoxy]methyl} -lH-pyrrolo[2,3-b]pyridine-5-carbonitrile»3 [HC1] (210 mg, 0.335 mmol), benzotriazol-l-yloxytris(dimethylamino)phosphonium
hexafluorophosphate (149 mg, 0.335 mmol) and triethylamine (0.234 mL, 1.68 mmol) in DMF (2.0 mL) was stirred at room temperature for 2 hours. The mixture was diluted with water, then extracted with EtOAc (2 times). The combined extracts were washed with saturated NaHC03, water, brine, dried over Na2S04. After filtration and evaporation, the residue was purified by flash chromatography on silica gel eluting with 0-10%
MeOH/EtOAc to give 143 mg of the intermediate. LC-MS found: 708.1 (M+H)+.
The purified above intermediate (143 mg) was dissolved in methylene chloride
(10 mL) and trifluoroacetic acid (10 mL, 100 mmol). The resulting solution was stirred at room temperature for 1 hour and concentrated. The residue was treated with methanol (10 mL) and ethylenediamine (5 mL, 70 mmol). The solution was stirred at room temperature for 1 hour. Purification with flash chromatography on silica gel eluting with 5-15% MeOH/EtOAc gave 63 mg (55%) of the title compound as an off-white solid. LC/MS found: 578.2 (M+l)+. 1H NMR (400 MHz, CDC13): δ 12.34 (brs, 1H), 8.66 (s, 1H), 8.60 (d, J = 4.80 Hz, 1H), 8.56 (s, 1H), 8.19 (s, 1H), 7.84 (t, J = 4.80 Hz, 1H), 7.68 (d, J = 3.60 Hz, 1H), 6.79 (d, J = 3.60 Hz, 1H), 4.01 (m, 1H), 3.68 (m, 2H), 3.54 (m, 2H), 3.49 (m, 2H), 3.36 (m, 1H), 3.22 (m, 1H), 3.03 (m, 1H), 2.50 (m, 1H), 1.69 (m, 1H), 1.57 (m, 1H), 1.16 (m, 2H).
The following compounds were prepared following the procedures described for Example 332.
Figure imgf000202_0001
Figure imgf000202_0002
Figure imgf000203_0001
1H NMR (300 MHz, DMSO-d6) of Example 339: δ 12.28 (brs, IH), 8.66 (s, IH), 8.55 (s, IH), 8.19 (s, IH), 7.92 (dd, Ji = 9.30 Hz, J2 = 1.20 Hz, IH), 7.72 (dd, Ji = 7.80 Hz, J2 = 1.20 Hz, IH), 7.67 (d, J = 3.90 Hz, IH), 7.56 (t, J = 7.20 Hz, IH), 6.78 (d, J = 3.60 Hz, IH), 4.00 (m, IH), 3.67 (m, 2H), 3.54 (m, 2H), 3.49 (s, 2H), 3.21 (m, 2H), 2.96 (m, IH), 2.49 (m, IH), 1.63 (m, 2H), 1.18 (m, 2H).
1H NMR (300 MHz, DMSO-d6) of Example 340: δ 8.66 (s, IH), 8.55 (s, IH), 8.19 (s, IH), 7.89 (d, J = 7.80 Hz, IH), 7.67 (d, J = 3.90 Hz, IH), 6.78 (d, J = 3.60 Hz, IH), 4.01 (m, IH), 3.71 (m, 2H), 3.57 (m, 2H), 3.50 (s, 2H), 3.38 (m, 2H), 3.03 (m, IH), 2.46 (m, IH), 1.68 (m, IH), 1.55 (m, IH), 1.14 (m, 2H). 1H NMR (300 MHz, DMSO-d6) of example 341 : δ 12.26 (brs, 1H), 8.87 (s, 1H), 8.66 (s, 1H), 8.56 (s, 1H), 8.19 (s, 1H), 8.06 (d, J = 10.5 Hz, 1H), 7.67 (d, J = 3.90 Hz, 1H), 6.79 (d, J = 3.60 Hz, 1H), 4.02 (m, 1H), 3.68 (m, 2H), 3.55 (m, 2H), 3.47 (s, 2H), 3.23 (m, 2H), 2.95 (m, 1H), 2.49 (m, 1H), 1.60 (m, 2H), 1.21 (m, 2H).
Example 342. {l-{l-[5-Chloro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}- (7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH- razol-l-yl]azetidin-3-yl}acetonitrile
Figure imgf000204_0001
Reaction of {l-piperidin-4-yl-3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H- pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile trihydrochloride with 5-chloro-2-trifluoromethylisonicotinoic acid following the procedure described for Example 1 , followed by HPLC purification (method B) provided the title compound. LC- MS: 570.2 (M+H)+. 1H NMR (300 MHz, DMSO-d6): δ 12.08 (brs, 1H),8.92 (, 1H), 8.80 (s, 1H), 8.67 (s, 1H), 8.40 (s, 1H), 8.11 (d, J = 14.09 Hz, 1H), 7.59 (d, J = 3.30 Hz, 1H), 7.04 (d, J = 3.30 Hz, 1H), 4.07 (m, 1H), 3.73 (m, 2H), 3.56 (m, 2H), 3.52 (s, 2H), 3.26 (m, 2H), 2.98 (m, 1H), 2.53 (m, 1H), 1.69 (m, 2H), 1.25 (m, 2H).
Example 343. {l-{l-[5-Fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4- (7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile
Figure imgf000205_0001
Reaction of { 1 -piperidin-4-yl-3-[4-(7- {[2-(trimethylsilyl)ethoxy]methyl} -7H- pyrrolo [2,3 -d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile trihydrochloride with 5-fluoro-2-trifluoromethylisonicotinoic acid following the procedure described for Example 1 , followed by HPLC purification (method B) provided the title compound. LC- MS: 554.2 (M+H)+. 1H NMR (400 MHz, DMSO-d6): δ 12.08 (brs, 1H), 8.84 (s, 1H), 8.76 (s, 1H), 8.63 (s, 1H), 8.36 (s, 1H), 8.09 (d, J = 4.80 Hz, 1H), 7.55 (d, J = 3.60 Hz, 1H), 7.00 (d, J = 3.60 Hz, 1H), 4.02 (m, 1H), 3.69 (m, 2H), 3.52 (m, 2H), 3.49 (s, 2H), 3.34 (m, 1H), 3.20 (m, 1H), 3.00 (m, 1H), 2.49 (m, 1H), 1.70 (m, 1H), 1.57 (m, 1H), 1.22 (m, 2H).
Example 344. [3-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]-l-(l-{[4- (trifluoromethyl)-l,3-thiazol-2-yl]carbonyl}piperidin-4-yl)azetidin-3-yl]acetonitrile
Figure imgf000206_0001
Figure imgf000206_0002
Reaction of { 1 -piperidin-4-yl-3-[4-(7- {[2-(trimethylsilyl)ethoxy]methyl} -7H- pyrrolo [2,3 -d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile trihydrochloride with 4-trifluoromethylthiazol-2-ylcarboxylic acid following the procedure described for Example 1 , followed by HPLC purification (method B) provided the title compound. LC- MS: 542.2 (M+H)+.
Example 345. [3-[3-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrrol-l-yl]-l-(l-{[2- (trifluoromethyl)pyrimidin-4-yl]carbonyl}piperidin-4-yl)azetidin-3-yl]acetonitrile
Figure imgf000206_0003
Reaction of { 1 -piperidin-4-yl-3-[3-(7- {[2-(trimethylsilyl)ethoxy]methyl} -7H- pyrrolo [2,3 -d]pyrimidin-4-yl)- 1 H-pyrrol- 1 -yl] azetidin-3 -yl} acetonitrile trihydrochloride with 2-trifluoromethylpyrimidin-4-carboxylic acid following the procedure described for
Example 261, followed by purification with HPLC (method B) provided the title compound. LC-MS: 536.2 (M+H)+. 1H NMR (300 MHz, DMSO-d6): δ 11.93 (brs, 1H), 9.18 (d, 1H), 8.59 (s, 1H), 7.95 (d, 1H), 7.79 (s, 1H), 7.48 (d, 1H), 7.05 (d, 1H), 6.91 (d, 2H), 4.00 (m, 1H), 3.55 (d, 2H), 3.40 (d, 2H), 3.35 (m, 1H), 3.30 (s 2H), 3.23 (m, 2H), 3.11 (m, 1H), 2.58 (m, 1H), 1.80-1.52 (m, 2H), 1.22 (m, 2H).
Example 346. [3-[3-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrrol-l-yl]-l-(l-{[4- (trifluoromethyl)-l,3-thiazol-2-yl]carbonyl}piperidin-4-yl)azetidin-3-yl]acetonitrile
Figure imgf000207_0001
Reaction of { 1 -piperidin-4-yl-3-[3-(7- {[2-(trimethylsilyl)ethoxy]methyl} -7H- pyrrolo [2,3 -d]pyrimidin-4-yl)- 1 H-pyrrol- 1 -yl] azetidin-3 -yl} acetonitrile trihydrochloride with 4-trifluoromethylthiazol-2-ylcarboxylic acid following the procedure described for Example 261, followed by purification with HPLC (method B) provided the title compound. LC-MS: 541.2 (M+H)+. 1H NMR (300 MHz, DMSO-d6): δ 11.92 (brs, 1H), 8.75 (s, 1H), 8.57 (s, 1H), 7.78 (s, 1H), 7.43 (d, 2H), 7.02 (s, 1H), 6.90 (s, 1H), 4.45 (m, 1H), 3.95 (m, 1H), 3.70 (m, 1H), 3.55 (d, 2H), 3.45 (d, 2H), 3.37 (s, 2H), 3.20 (m, 2H), 1.80-1.60 (m, 2H), 1.25 (m, 2H).
Example 347. [3-[3-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrrol-l-yl]-l-(l-{[5- (trifluoromethyl)pyrazin-2-yl]carbonyl}piperidin-4-yl)azetidin-3-yl]acetonitrile
Figure imgf000208_0001
Reaction of { 1 -piperidin-4-yl-3-[3-(7- {[2-(trimethylsilyl)ethoxy]methyl} -7H- pyrrolo [2,3 -d]pyrimidin-4-yl)- 1 H-pyrrol- 1 -yl] azetidin-3 -yl} acetonitrile trihydrochloride with 5-trifiuoromethylpyrazin-2-ylcarboxylic acid following the procedure described for Example 261 , followed by purification with HPLC (method B) provided the title compound. LC-MS: 536.1 (M+H)+. 1H NMR (300 MHz, DMSO-d6): δ 11.99 (brs, 1H), 9.23 (s, 1H), 9.07 (s, 1H), 8.62 (s, 1H), 7.84 (s, 1H), 7.55 (d, 1H), 7.11 (s, 1H), 6.98 (d, 2H), 4.12 (m 1H), 3.65 (d, 2H), 3.55 (d, 2H), 3.41 (m, 1H), 3.31 (s, 2H), 3.15 (m, 2H), 2.60 (m, 1H), 1.90-1.60 (m, 2H), 1.30 (m, 2H).
Example 348. { 1- [ l-(Methylsulfonyl)piperidin-4-yl] -3- [3-(7H-pyr rolo [2,3- d]pyrimidin-4-yl)-lH-pyrrol-l-yl]azetidin-3- l} acetonitrile
Figure imgf000208_0002
Reaction of { 1 -piperidin-4-yl-3-[3-(7- {[2-(trimethylsilyl)ethoxy]methyl} -7H- pyrrolo [2,3 -d]pyrimidin-4-yl)- 1 H-pyrrol- 1 -yl] azetidin-3 -yl} acetonitrile trihydrochloride with methanesulfonyl chloride following the procedure described for Example 246, followed by purification with HPLC (method B) provided the title compound. LC-MS:
440.1 (M+H)+. 1H NMR (300 MHz, DMSO-d6): δ 11.91 (brs, 1H), 8.55 (s, 1H), 7.76 (s, 1H), 7.43 (d, 1H), 7.01 (d, 1H), 6.88 (d, 2H), 3.52 (d, 2H), 3.47 (d, 2H), 3.30 (m, 2H), 3.27 (s, 2H), 2.80 (m, 2H), 2.75 (s, 3H), 2.27 (m, 1H), 1.65 (m, 2H), 1.23 (m, 2H).
Example 349. [3-[4-(lH-Pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol-l-yl]-l-(l-{[5- (trifluoromethyl)pyrazin-2-yl]carbon l}piperidin-4-yl)azetidin-3-yl]acetonitrile
Figure imgf000209_0001
Reaction of { 1 -piperidin-4-yl-3-[4-(7- {[2-(trimethylsilyl)ethoxy]methyl} -7H- pyrrolo[2,3-d]pyridin-4-yl)- IH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile trihydrochloride with 5-trifluoromethylpyrazin-2-carboxylic acid following the procedure described for Example 294, followed by HPLC purification (method B) provided the title compound. LC-MS: 536.2 (M+H)+. 1H NMR (300 MHz, DMSO-d6): δ 12.07 (brs, 1H), 8.82 (s, 1H), 8.76 (s, 1H), 8.68 (s, 2H), 8.41 (s, 2H), 7.61 (d, 1H), 7.06 (d, 1H), 4.55 (m, 1H), 4.05 (m, 1H), 3.75 (dd, 2H), 3.59 (dd, 2H), 3.55 (m, 2H), 3.32 (s, 2H), 3.30 (m, 1H), 1.80 (m, 2H), 1.30 (m, 2H).
Example 350. [3-[4-(lH-Pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol-l-yl]-l-(l-{[4- (trifluoromethyl)-l,3-thiazol-2-yl]carbonyl}piperidin-4-yl)azetidin-3-yl]acetonitrile
Figure imgf000210_0001
Reaction of { 1 -piperidin-4-yl-3-[4-(7- {[2-(trimethylsilyl)ethoxy]methyl} -7H- pyrrolo[2,3-d]pyridin-4-yl)- IH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile trihydrochloride with 4-trifluoromethylthiazol-2-carboxylic acid following the procedure described for Example 294, followed by HPLC purification (method B) provided the title compound. LC-MS: 541.2 (M+H)+. 1H NMR (300 MHz, DMSO-d6): δ 12.01 (brs, 1H), 9.09 (s, 1H), 8.99 (s, 1H), 8.59 (s, 1H), 8.48 (d, 1H), 7.82 (dd, 1H), 7.62 (d, 1H), 7.16 (d, 1H), 4.85 (m, 1H), 4.35 (m, 1H), 4.05 (d, 2H), 4.03 (m, 1H), 3.90 (d, 1H), 3.80 (m, 1H), 3.62 (s, 2H), 3.58 (m, 1H), 2.90 (m, 1H), 2.08 (m, 2H), 1.62 (m, 2H).
Example 351. {l-[l-(Methylsulfonyl)piperidin-4-yl]-3-[4-(lH-pyrrolo[2,3-b]pyridin- 4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile
Figure imgf000210_0002
Reaction of { 1 -piperidin-4-yl-3-[4-(7- {[2-(trimethylsilyl)ethoxy]methyl} -7H- pyrrolo[2,3-d]pyridin-4-yl)- IH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile trihydrochloride with methanesulfonyl chloride following the procedure described for Example 246, followed by HPLC purification (method B) provided the title compound. LC-MS: 440.1
(M+H)+. 1H NMR (300 MHz, DMSO-d6): δ 11.91 (brs, IH), 8.55 (s, IH), 7.76 (s, IH), 7.43 (d, IH), 7.01 (d, IH), 6.88 (d, 2H), 3.52 (d, 2H), 3.47 (d, 2H), 3.30 (m, 2H), 3.27 (s, 2H), 2.80 (m, 2H), 2.75 (s, 3H), 2.27 (m, IH), 1.65 (m, 2H), 1.23 (m, 2H).
Example 352. [3-[4-(5-Fluoro-lH-pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol-l-yl]-l-(l- { [6-(trifluoromethyl)pyr azin-2-yl] carbon l}piperidin-4-yl)azetidin-3-yl] acetonitrile
Figure imgf000211_0001
Reaction of {3-[4-(5-fluoro-l-{[2-(trimethylsilyl)ethoxy]methyl}-lH-pyrrolo[2,3- b]pyridin-4-yl)- 1 H-pyrazol- 1 -yl]- 1 -piperidin-4-ylazetidin-3-yl} acetonitrile»3 [HC1] with 6-trifluoromethylpyrazin-2-carboxylic acid following the procedure described for Example 314, followed by HPLC purification (method B) provided the title compound.
LC-MS: 554.1 (M+H)+. 1H NMR (300 MHz, DMSO-d6): δ 12.11 (brs, IH), 9.23 (s, IH), 9.12 (s, IH), 8.56 (s, IH), 8.19 (d, IH), 8.15 (d, IH), 7.58 (d, IH), 6.82 (d, IH), 4.03 (m, IH), 3.68 (d, 2H), 3.55 (d, 2H), 3.45 (m, IH), 3.26 (s 2H), 3.24 (m, IH), 3.11 (m, IH), 2.55 (m, IH), 1.80-1.57 (m, 2H), 1.25 (m, 2H). Example 353. [3-[4-(5-Fluoro-lH-pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol-l-yl]-l-(l-
{[2-(trifluoromethyl)pyrimidin-4-yl]carbonyl}piperidin-4-yl)azetidin-3- yl]acetonitrile
Figure imgf000212_0001
Reaction of {3-[4-(5-fluoro-l-{[2-(trimethylsilyl)ethoxy]methyl}-lH-pyrrolo[2,3- b]pyridin-4-yl)- 1 H-pyrazol- 1 -yl]- 1 -piperidin-4-ylazetidin-3-yl} acetonitrile»3 [HC1] with 2-trifluoromethylpyrimidin-4-carboxylic acid following the procedure described for Example 314, followed by HPLC purification (method B) provided the title compound. LC-MS: 554.1 (M+H)+. 1H NMR (300 MHz, DMSO-d6): δ 11.83 (brs, 1H), 9.15 (d, 1H), 8.56 (s, 1H), 8.21 (d, 1H), 8.18 (d, 1H), 7.94 (d, 1H), 7.58 (d, 1H), 6.82 (d, 1H), 4.00 (m, 1H), 3.70 (d, 2H), 3.55 (d, 2H), 3.45 (m, 1H), 3.40 (s, 2H), 3.15 (m, 1H), 3.08 (m, 1H), 2.52 (m, 1H), 1.80 -1.52 (m, 2H), 1.23 (m, 2H).
Example 354. [3-[4-(5-Fluoro-lH-pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol-l-yl]-l-(l- { [5-(trifluoromethyl)pyrazin-2-yl] carbonyl}piperidin-4-yl)azetidin-3-yl] acetonitrile
Figure imgf000213_0001
Reaction of {3-[4-(5-fluoro-l-{[2-(trimethylsilyl)ethoxy]methyl}-lH-pyrrolo[2,3- b]pyridin-4-yl)- 1 H-pyrazol- 1 -yl]- 1 -piperidin-4-ylazetidin-3-yl} acetonitrile»3 [HC1] with 5-trifluoromethylpyrazin-2-carboxylic acid following the procedure described for Example 314, followed by HPLC purification (method B) provided the title compound. LC-MS: 554.1 (M+H)+. 1H NMR (300 MHz, DMSO-d6): δ 11.82 (brs, 1H), 9.16 (s, 1H), 8.99 (s, 1H), 8.59 (s, 1H), 8.20 (d, 2H), 7.59 (s, 1H), 6.82 (s, 1H), 4.05 (m, 1H), 3.70 (d, 2H), 3.55 (d, 2H), 3.45 (m, 1H), 3.35 (s, 2H), 3.15 (m, 2H), 2.59 (m, 1H), 1.80-1.55 (m, 2H), 1.22 (m, 2H).
Example 355. {3-[4-(5-Fluoro-lH-pyrrolo[2,3-b]pyridin-4-yl)-lHpyrazol-l-yl]-l-[l- (methylsulfonyl)piperidin-4-yl] azetidin-3- l} acetonitrile
Figure imgf000213_0002
Reaction of {3-[4-(5-fluoro-l-{[2-(trimethylsilyl)ethoxy]methyl}-lH-pyrrolo[2,3- b]pyridin-4-yl)- 1 H-pyrazol- 1 -yl]- 1 -piperidin-4-ylazetidin-3-yl} acetonitrile»3 [HC1] with methanesulfonyl chloride following the procedure described for Example 246, followed by HPLC purification (method B) provided the title compound. LC-MS: 458.1 (M+H)+. 1H NMR (300 MHz, DMSO-d6): δ 11.87 (brs, 1H), 8.59 (s, 1H), 8.23 (d, J = 3.60 Hz, 1H), 8.20 (d, J = 2.10 Hz, 1H), 7.61 (d, J = 3.60 Hz, 1H), 6.86 (d, J = 3.30 Hz, 1H), 3.71 (m, 2H), 3.57 (m, 2H), 3.54 (s, 2H), 3.36 (m, 2H), 2.86 (m, 2H), 2.83 (s, 3H), 2.37 (m, 1H), 1.73 (m, 2H), 1.30 (m, 2H).
Example 356. 4-[l-(3-(Cyanomethyl)-l-{l-[5-fluoro-2-
(trifluoromethyl)isonicotinoyl] piperidin-4-yl} azetidin-3-yl)- lH-pyrazol-4-yl] - 1H- pyrrolo [2,3-b] pyridine-5-carbonitrile
Figure imgf000214_0001
Reaction of 4-{l-[3-(cyanomethyl)-l-piperidin-4-ylazetidin-3-yl]-lH-pyrazol-4- yl} - 1 - { [2-(trimethylsilyl)ethoxy]methyl} - 1 H-pyrrolo[2,3-b]pyridine-5- carbonitrile»3[HCl] with 5-fluoro-2-trifluoromethylisonicotinoic acid following the procedure described for Example 332, followed by HPLC purification (method B) provided the title compound. LC-MS: 578.2 (M+H)+. 1H NMR (300 MHz, DMSO-d6) : δ 8.84 (s, 1H), 8.67 (s, 1H), 8.55 (s, 1H), 8.19 (s, 1H), 8.09 (d, J = 4.80 Hz, 1H), 7.68 (d, J = 3.60 Hz, 1H), 6.79 (d, J = 3.60 Hz, 1H), 3.99 (m, 1H), 3.69 (m, 2H), 3.54 (m, 2H), 3.49 (s, 2H), 3.42 (m, 2H), 3.00 (m, 1H), 2.51 (m, 1H), 1.69 (m, 1H), 1.57 (m, 1H), 1.20 (m, 2H). Example 357. 4-(l-{3-(Cyanomethyl)-l-[l-(methylsulfonyl)piperidin-4-yl]azetidin-3- yl}-lH-pyrazol-4-yl)-lH-pyrrolo[2,3-b ridine-5-carbonitrile
Figure imgf000215_0001
Reaction of 4-{l-[3-(cyanomethyl)-l-piperidin-4-ylazetidin-3-yl]-lH-pyrazol-4- yl} - 1 - { [2-(trimethylsilyl)ethoxy]methyl} - 1 H-pyrrolo[2,3-b]pyridine-5- carbonitrile»3[HCl] with methanesulfonyl chloride following the procedure described for Example 246, followed by HPLC purification (method B) provided the title compound. LC-MS: 465.1 (M+H)+. 1H NMR (300 MHz, DMSO-d6): δ 12.28 (brs, 1H), 8.66 (s, 1H), 8.55 (s, 1H), 8.20 (s, 1H), 7.68 (d, J = 3.60 Hz, 1H), 6.79 (d, J = 3.90 Hz, 1H), 3.67 (m, 2H), 3.53 (m, 2H), 3.50 (s, 2H), 3.31 (m, 2H), 2.81 (m, 2H), 2.78 (s, 3H), 2.35 (m, 1H), 1.68 (m, 2H), 1.26 (m, 2H).
Example 358. {l-{l-[3-Fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4- (7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile adipic acid salt
Figure imgf000216_0001
Screening: 1 - { 1 -[3-Fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl} -3-[4- (7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile (Example 1) free base is an amorphous substance. A salt screening study was performed using pharmaceutically acceptable acids for the formation of crystalline Example 1 salts.
Adipic acid was identified to give a crystalline Example 1 adipic acid salt. The initially obtained solid form (Form I) of Example 1 adipic acid salt with a melting point of 178 °C has been selected for optimization. This form is a crystalline form as verified by X-ray powder diffractometry (XRPD), differential scanning calorimetry (DSC), and thermal gravimetric analysis (TGA). The stoichiometry of the salt was determined to be 1 : 1 (freebase to adipic acid) by 1H NMR spectroscopy and elemental analysis.
A polymorph screening study was performed on Example 1 adipate salt. Phase equilibration studies were carried out by slurrying the Form I crystals in a variety of solvents (MeCN, CHC13, CH2C12, MIBK, MEK, acetone, toluene, hexane, heptane, THF, MTBE, EtOH, z-PrOH, rc-BuOH, EtOAc, z-PrOAc at 25°C or 50°C. At 25°C, all the solvents tested gave the same crystalline Form I after slurrying. At 50°C, the same results were observed with the exception of ethanol. The XRPD pattern of the solid obtained from ethanol slurry showed the presence of free base which can be explained by a simple salt dissociation. The results of the phase equilibration studies suggest Form I is a stable crystalline form. Moreover, multiple lots (from gram to kilogram scales) of Example 1 adipic acid salt made to date have been determined to be the same crystalline form (Form I). Seeding had been used to induce the formation of Form I in the crystallization. However, it was also observed that Form I was obtained even without seeding in the crystallization.
Preparation: Adipic acid (790 g, 5.406 mol) was dissolved in methanol (29 L) at 16 °C. 2-(3-(4-(7H-Pyrrolo[2,3- ]pyrimidin-4-yl)- lH-pyrazol- 1 -yl)- 1 -(1 -(3-fiuoro-2- (trifiuoromethyl)-isonicotinoyl)piperidin-4-yl)azetidin-3-yl)acetonitrile free base (2.85 kg, 5.149 mol) was added to the adipic acid solution in methanol at 16 °C. The reaction mixture was heated under reflux for 2 hours. The resulting reaction mixture was cooled to ambient temperature and the solvent was removed by distillation under reduced pressure to give the crude adipic acid salt. The crude adipic acid salt was dissolved in acetone (14 L) at ambient temperature. n-Heptane (20 L) was added over 2 hours to the crude adipic acid salt solution in acetone at 18°C to precipitate the salt. The resultant slurry was stirred at 18°C for 1 hour. The salt was isolated by filtration. The wet cake was washed with n- heptane (6 L). The product was dried on the filter funnel under suction for 18 hours to afford the crude 2-(3-(4-(7H-pyrrolo[2,3-<i]pyrimidin-4-yl)-lH-pyrazol-l-yl)-l-(l-(3- fluoro-2-(trifluoromethyl)-isonicotinoyl)piperidin-4-yl)azetidin-3-yl)acetonitrile adipic acid salt (3.572 kg , 5.105 mol, 99.2% yield) as a white crystalline solid.
The crude adipic acid salt can be further purified by recrystallization. The crude adipic acid salt (3.378 kg, 4.828 mol) was suspended in acetone (24 L) at ambient temperature. The resulting suspension was heated to 55°C and stirred at 50-60°C to give a clear solution. The solution was filtered through an in-line filter to remove particulates, n- Heptane (24 L) was added to the solution at 55°C over 2 hours to precipitate the salt. Upon complete addition of /? -heptane, the slurry was cooled to 30°C over 3 hours. The pure adipic acid salt was isolated by filtration. The wet cake was washed with a mixture of n-heptane and acetone (2: 1 v/v, 6.8 L). The product was dried on the filter funnel under suction for 15 hours and was further dried in a vacuum oven at 55 °C for 42 hours to give pure 2-(3-(4-(7H-pyrrolo[2,3-<i]pyrimidin-4-yl)-lH-pyrazol-l-yl)-l-(l-(3-fiuoro- 2-(trifluoromethyl)isonicotinoyl)piperidin-4-yl)azetidin-3-yl)acetonitrile adipic acid salt, 3.116 kg, 92.2% yield) as a white crystalline solid.
For adipic acid salt: mp 178°C; 1H NMR (400 MHz, DMSO-<¾) δ 12.14 (s, 1H), 12.02 (br s, 2H), 8.81 (s, 1H), 8.69 (s, 1H), 8.66 (d, J= 4.7 Hz, 1H), 8.42 (s, 1H), 7.90 (dd, J= 4.7, 4.7, 1H), 7.60 (dd, J= 2.3, 3.5 Hz, 1H), 7.06 (dd, J= 1.8, 3.6 Hz, 1H), 4.08 (m, 1H), 3.74 (m, 2H), 3.57 (m, 2H), 3.55 (m, 2H), 3.39 (m, 1H), 3.25 (m, 1H), 3.07 (m, 1H), 2.56 (m, 1H), 2.19 (m, 4H), 1.77 (m, 1H), 1.62 (m, 1H), 1.48 (m, 4H), 1.36 - 1.12 (m, 2H); 13C NMR (100 MHz, OMSO-d6) δ 174.4, 160.3, 152.2 ( ¥ = 265 Hz), 152.2, 150.9, 149.6, 146.3 (4JCF = 5.8 Hz), 139.5, 135.0 (2JCF = 17.3 Hz), 134.5 (2JCF = 35.3, 1 1.9Hz), 129.2, 127.6, 126.8, 121.7, 120.6 (\½ = 274.0 Hz, 3JCF = 4.8 Hz), 1 17.4, 1 13.0, 100.0, 61.4, 60.5 57.0, 44.2, 33.4, 28.6, 27.9, 27.2, 24.0; 19F NMR (376 MHz, OMSO-d6) δ -64.54 (d, J= 15.8 Hz, 3F), -129.34 (m, IF); Anal. Calcd for: C32H33F4N905: C, 54.93; H, 4.75; F, 10.86; N, 18.02; found: C, 54.68; H, 4.56; F, 10.94; N, 17.90. LCMS calculated for C26H24F4N90 (M + H)+ for the free base: m/z 554.2; found: 554.2.
The chemical purity was determined by reverse-phase HPLC to be 99.57 area%; the DSC thermogram revealed one major endothermic event with an onset of the peak at 175.9°C which is believed to relate to the compound melting with the peak at 177.9°C (see Figure 1). The DSC was scanned from an initial temperature of 30°C to a final temperature of 280°C using a heating rate of 10°C/min. The TGA thermogram showed a small weight loss of 0.29% observed from 20°C to 100°C, and a significant weight loss of 62%o was observed upon further heating from 100°C to 600°C (see Figure 2). The TGA thermogram was obtained when the sample was heated from 20°C to 600°C at a heating rate of 20°C/min. The XRPD pattern indicated crystalline nature of the adipic acid salt (see Figure 3). The DSC, TGA, and XRPD data were consistent with those of Form I.
DSC parameters: Mettler Toledo Differential Scanning Calorimetry (DSC) instrument, Model No. 822; Aluminum sample pan (40 μί); general condition: 30-280 °C at 10 °C/min.
TGA parameters: TA Instrument, Model No. Q500. The general starting method condition is: ramp at 20 °C/min. to 600 °C.
XRPD conditions: Rigaku MiniFlex X-ray Powder Diffractometer (XRPD) instrument; X-ray radiation is from Copper Cu at 1.054056A with Κβ filter; sample powder is dispersed on a zero-background sample holder; and general measurement conditions are:
Start Angle - 3
Stop Angle - 45 Sampling - 0.02
Scan speed - 2
Table 1. XRPD Data
-Theta (°) d(A) BG Height H% Area A% FWHM.84 22.9919 7 341 24.4 19142 100 0.955.92 12.7638 169 461 33 6796 35.5 0.25.78 10.0638 164 52 3.7 1571 8.2 0.513.28 9.5227 161 47 3.4 760 4 0.2750.4 8.4994 203 1399 100 15230 79.6 0.1850.981 8.0506 208 135 9.6 3688 19.3 0.4651.74 7.532 179 302 21.6 6396 33.4 0.364.92 5.933 165 723 51.7 15980 83.5 0.3765.4 5.7492 179 377 27 6733 35.2 0.3036.859 5.2547 295 123 8.8 843 4.4 0.1 177.52 5.058 249 316 22.6 12179 63.6 0.6558.68 4.7463 238 482 34.4 7294 38.1 0.2579.861 4.4668 240 361 25.8 5072 26.5 0.2390.98 4.2309 261 547 39.1 1 1823 61.8 0.3682.12 4.0153 267 273 19.5 6037 31.5 0.3772.46 3.9553 280 414 29.6 8893 46.5 0.3653.28 3.8178 300 546 39 10395 54.3 0.3243.74 3.7449 254 216 15.5 9220 48.2 0.7254.38 3.6481 270 256 18.3 2926 15.3 0.1945.062 3.5503 219 54 3.9 791 4.1 0.2495.979 3.427 247 212 15.1 3384 17.7 0.2726.901 3.31 16 241 60 4.3 1 124 5.9 0.327.76 3.21 1 1 213 78 5.6 1985 10.4 0.4318.839 3.0933 203 170 12.1 2489 13 0.2499.841 2.9917 205 98 7 1 1 15 5.8 0.1940.94 2.8879 184 127 9.1 5062 26.4 0.6771.562 2.8324 184 66 4.7 623 3.3 0.1612.92 2.7185 181 125 8.9 3846 20.1 0.5225.14 2.5518 182 147 10.5 4215 22 0.4885.62 2.5185 173 83 6 5361 28 1.093 2-Theta (°) d(A) BG Height H% Area A% FWHM
36.96 2.4302 178 52 3.7 1724 9 0.559
37.359 2.4051 178 89 6.4 2358 12.3 0.45
38.86 2.3156 173 72 5.2 3599 18.8 0.846
39.279 2.2918 177 77 5.5 2214 1 1.6 0.486
Example 359. c/s-{l-{(3-Methoxy-l-[3-fluoro-2-
(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile
Figure imgf000220_0001
Step 1. tert-Butyl cis-4-[3-(cyanomethylene)azetidin-l-yl]-3-methoxypiperidine-l- carboxylate
Figure imgf000220_0002
To a solution of tert-butyl 3-(cyanomethylene)azetidine-l-carboxylate (3.0 g, 15 mmol) in tetrahydrofuran (8 niL) was added a 4.0 M solution of hydrogen chloride in 1,4-dioxane (10 mL, 40 mmol). After being stirred at room temperature for two hours, the reaction mixture was concentrated under reduced pressure. The residue was dissolved in tetrahydrofuran (20 mL), and tert-butyl 3-methoxy-4-oxopiperidine-l-carboxylate (3.58 g, 15.62 mmol) and triethylamine (3.126 g, 30.89 mmol) were added. After being stirred at room temperature for 30 minutes, sodium triacetoxyborohydride (8.184 g, 38.61 mmol) was added. The mixture was stirred at room temperature overnight. The resulting solution was diluted with aqueous NaHC03 and EtOAc. The organic layer was separated and washed with brine, dried over Na2SC>4 and concentrated. Purification with combi- flash (20-100% EtOAc in hexanes) afforded 2.8 g (60% yield) of the desired product. LC/MS found: 308.1 (M-56)+.
Step 2. cis-{l-[ 3-Methoxypiperidin-4-yl] -3- [ 4-(7-{[ 2-(trimethylsilyl)ethoxyJmethyl}-7H- pyrrolo [2, 3-d) 'pyrimidin-4-yl)- lH-pyrazol- 1-ylj 'azetidin-3-yl}acetonitrile
Figure imgf000221_0001
To a solution of 4-(lH-pyrazol-4-yl)-7-{[2-
(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidine (0.256 g, 0.812 mmol) in acetonitrile (10 mL) were added tert-butyl cis-4-[3-(cyanomethylene)azetidin-l-yl]-3- methoxypiperidine-l-carboxylate (0.20 g, 0.68 mmol) and l,8-diazabicyclo[5.4.0]undec- 7-ene (0.152 mL, 1.02 mmol ). After being stirred at room temperature for 5 minutes, the reaction mixture became a clear solution. Stirring was continued at room temperature overnight. LC-MS indicated the reaction was complete. The solution was concentrated under reduced pressure and ethyl acetate was added. The resulting solution was washed with 1 N HCl and brine, dried over sodium sulfate, filtered and concentrated. Purification by silica gel chromatography (20-100% EtOAc/hexanes) afforded an oily product. The product was dissolved in THF (5 mL). To it was added 4 N HCl in dioxane (5 mL). After being stirred at room temperature for 2 hours, the solution was concentrated under reduced pressure to give 0.30 g (86%) of the title compound as a solid. LCMS: 511.1 (M+l)+.
Step 3. cis-{l-{(3-Methoxy-l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin^ 3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetom
Into a 20 mL vial were added 3-fluoro-2-(trifluoromethyl)isonicotinic acid (18.67 mg, 0.0893 mmol) in DMF (3 mL), {l-[cis-3-methoxypiperidin-4-yl]-3-[4-(7-{[2- (trimethylsilyl)ethoxy]methyl} -7H-pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 - yl]azetidin-3-yl}acetonitrile (50.17 mg, 0.098 mmol), benzotriazol-1- yloxytris(dimethylamino)phosphonium hexafluorophosphate (59.25 mg, 0.134 mmol) and triethylamine (0.037 mL, 0.268 mmol). The mixture was stirred at room temperature overnight and purified with prep-LCMS to give 20 mg of the desired intermediate as a white solid. The white solid was dissolved in methylene chloride (1 mL) and
trifluoroacetic acid (1 mL) was added. After being stirred at room teprature for one hour, tthe solution was concentrated. The residue was dissolved in methanol (2 mL) and ethylene diamine (0.1 g, 2 mmol) was added. The mixture was stirred at room
temperature for two hours. Purification by prep-LCMS (pH=10, Method C) gave the title compound as a white powder. LCMS: 584.3 (M+l)+. 1H NMR (400 MHz, CD3OD): δ 12.93 (s, 1H), 9.60 (d, J=2 Hz, 1H), 9.48 (d, J=7.2 Hz, 1H), 9.46 (d, J=4.4 Hz, 1H), 9.21 (d, J=1.2 Hz, 1H), 8.64 (s, 1H), 8.40 (d, J=3.6 Hz, 1H), 7.85 (d, J=3.2 Hz, 1H), 4.81 (m, 1H), 4.60 (m, 1H), 4.55 (m, 1H), 4.40 (m, 4H), 4.28 (m, 1H), 4.10 (m, 1H), 3.95 (m, 1H), 3.90 (s, 3H), 3.56 (m, 1H), 2.40 (m, 2H), 2.25 (m, 1H).
Example 360. {l-(cis-3-Methoxy-l-{[2-(trifluoromethyl)pyrimidin-4- yl]carbonyl}piperidin-4-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin-3-yl}acetonitrile
Figure imgf000223_0001
Into a 20 mL vial were added {l-[cis-3-methoxypiperidin-4-yl]-3-[4-(7-{[2- (trimethylsilyl)ethoxy]methyl} -7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 - yl]azetidin-3-yl}acetonitrile (660 mg, 1.3 mmol) in DMF (10 mL), 2- (trifluoromethyl)pyrimidine-4-carboxylic acid (270 mg, 1.4 mmol), benzotriazol-1- yloxytris(dimethylamino)phosphonium hexafiuorophosphate (610 mg, 1.4 mmol) and triethylamine (0.48 mL, 3.4 mmol). The mixture was stirred at room temperature overnight and purified with combi-flash using 5% methanol (MeOH)/50%
EtOAc/hexanes to give 400 mg of the desired intermediate as a white solid. LC/MS found: 697.2 (M+l)+.
The above residue was dissolved in methylene chloride (2 mL) and trifluoroacetic acid (2 mL) was added. After being stirred at room temperature for one hour, the solution was concentrated. The residue was dissolved in methanol (5 mL) and ethylenediamine (0.4 g, 7 mmol) was added. The resulting mixture was stirred at room temperarure for two hours. Purification by prep-LCMS (pH=10, Method C) gave 210 mg of the title compound as a white powder. LCMS found: 567.2 (M+l)+. 1H NMR (400 MHz, CD3OD): δ 12.93 (s, IH), 10.00 (dd, J=5.6 & 5.2 Hz, IH), 9.60 (s, IH), 9.48 (s, IH), 9.21 (s, IH), 8.73 (dd, J=5.6 & 5.6 Hz, IH), 8.40 (d, J=3.6 Hz, IH), 7.85 (d, J=3.2 Hz, IH), 4.78 (m, IH), 4.60 (m, IH), 4.56 (m, IH), 4.40 (m, 4H), 4.25 (m, IH), 4.08 (m, IH), 3.95 (m, IH), 3.90 (s, 3H), 3.60 (m, IH), 2.40 (m, 2H), 2.25 (m, IH). Example 361. {l-{cis-3-Fluoro-l-[3-fluoro-2-
(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile
Figure imgf000224_0001
Step 1. tert-Butyl cis-4-[3-(Cyanomethylene)azetidin-l-yl]-3-fluoropiperidine-l- carboxylate
Figure imgf000224_0002
To a solution of tert-butyl 3-(cyanomethylene)azetidine-l-carboxylate (3.0 g, 15 mmol) in tetrahydrofuran (8 mL) was added a 4.0 M solution of hydrogen chloride in 1,4- dioxane (10 mL, 40 mmol). After being stirred at room temperature for two hours, the solution was concentrated under reduced pressure. The residue was dissolved in tetrahydrofuran (20 mL). To it were added tert-butyl 3-fluoro-4-oxopiperidine-l- carboxylate (3.392 g, 15.62 mmol) and triethylamine (3.126 g, 30.89 mmol). After being stirred at room temperature for 30 minutes, sodium triacetoxyborohydride (8.184 g, 38.61 mmol) was added. The mixture was stirred at room temperature overnight and diluted with aqueous NaHC03 and EtOAc. The organic layer was separated and washed with brine, dried over Na2S04 and concentrated under reduced pressure. Purification with combi-fiash (20-100% EtOAc in hexanes) afforded 0.5 g (66% yield) of the desired product. LC/MS found: 240.1 (M-56)+.
Step 2. { 1- [cis-3-Fluoropiperidin-4-yl] -3- [4-(7-{ [2-(trimethylsilyl)ethoxy
pyrrolo [2, 3-d) 'pyrimidin-4-yl)- lH-pyrazol- 1-ylj 'azetidin-3-yl}acetonitrile
Figure imgf000225_0001
To a solution of tert-butyl cis-4-[3-(cyanomethylene)azetidin-l-yl]-3- fluoropiperidine-l-carboxylate (0.24 g, 0.81 mmol) and 4-(lH-pyrazol-4-yl)-7-{[2- (trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidine (0.31 g, 0.98
mmol) in acetonitrile (8 mL) was added l,8-diazabicyclo[5.4.0]undec-7-ene (0.15 g, 0.98 mmol) by syringe. The resulting solution was stirred at room temperature for 6 hours and concentrated. Purification with combi-fiash using 40-100% EtOAc/hexanew as eluent gave 0.30 g (61%> yield) of the desired compound as a solid. LCMS found: 611.1 (M+l)+.
The above solid was dissolved in tetrahydrofuran (4 mL). To it was added a solution of 4.0 M HC1 in dioxane (4 mL). The solution was stirred at room temperature for 2 hours and concentrated to give the title compound: LCMS: 511.1 (M+l)+.
Step 3. {l-{cis-3-Fluoro-l-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}- [4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}a^
Into a 20 mL vial were added {l-[cis-3-fluoropiperidin-4-yl]-3-[4-(7-{[2- (trimethylsilyl)ethoxy]methyl} -7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 - yl]azetidin-3-yl} acetonitrile (200 mg, 0.4 mmol) in DMF(3 mL), 3-fluoro-2- (trifluoromethyl)isonicotinic acid (99 mg, 0.47 mmol), benzotriazol-1- yloxytris(dimethylamino)phosphonium hexafluorophosphate (225 mg, 0.509 mmol) and triethylamine (0.14 mL, 1.0 mmol). The mixture was stirred at room temperature overnight and purified with prep-LCMS to give 50 mg of the desired compound as a white solid. LC/MS found: 702.2 (M+l)+.
The above product was dissolved in methylene chloride (2 mL). To it was added trifluoroacetic acid (2 mL). The resulting mixture was stirred at room temperature for an hour and concentrated. The residue was taken up in methanol (5 mL) and
ethylenediamine (0.5 g, 8 mmol) was added. The mixture was stirred at room temperature for two hours. Purification with prep-LCMS (pH=10, Method C) gave the title compound as a white powder. LCMS found: 572.2 (M+l)+. 1H NMR (400 MHz, CD3OD): δ 12.93 (s, 1H), 9.62 (s, 1H), 9.49 (s, 1H), 9.48 (d, J=4.8 Hz, 1H), 9.22 (s, 1H), 8.60 (m, 1H), 8.40 (d, J=4.0 Hz, 1H), 7.85 (d, J=3.2 Hz, 1H), 5.60 (m, 1H), 5.20 (m, 1H), 4.60 (m, 2H), 4.42 (m, 2H), 4.38 (m, 2H), 4.20 (m, 1H), 3.90 (m, 1H), 3.58 (m, 1H), 2.52 (m, 1H), 2.35 (m, 2H).
Example 362. {l-(cis-3-Fluoro-l-{[2-(trifluoromethyl)pyrimidin-4- yl]carbonyl}piperidin-4-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin-3-yl}acetonitrile
Figure imgf000226_0001
To a solution of {l-[cis-3-fluoropiperidin-4-yl]-3-[4-(7-{[2- (trimethylsilyl)ethoxy]methyl} -7H-pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 - yl]azetidin-3-yl}acetonitrile (200 mg, 0.4 mmol) and 2-(trifluoromethyl)pyrimidine-4- carboxylic acid (83 mg, 0.43 mmol) in DMF (10 mL) were added benzotriazol-1- yloxytris(dimethylamino)phosphonium hexafluorophosphate (5.20E2 mg, 1.17 mmol) and triethylamine (0.14 mL, 0.98 mmol). The mixture was stirred at room temperature overnight and purified with prep-LCMS to give 100 mg (36% yield) of the desired compound as a white solid.
The above residue was dissolved in methylene chloride (2 mL). To it was added trifluoroacetic acid (2 mL). After being stirred at room temperature for an hour, the solution was concentrated. The residue was dissolved in methanol (5 mL) and
ethylenediamine (1 mL) was added. After being stirred at room temperature for two hours, the mixture was purified with prep-LCMS (pH=10, Method C) to give 41 mg (51% yield) of the title compound as a white powder. LCMS found: 555.3 (M+H)+. 1H NMR (400 MHz, CD3OD): δ 12.93 (s, 1H), 10.01 (dd, J=5.2 & 1.2 Hz, 1H), 9.62 (s, 1H), 9.49 (s, 1H), 9.22 (s, 1H), 8.71 (dd, J=22.4 & 4.8 Hz, 1H), 8.40 (dd, J=3.4 & 2.4 Hz, 1H), 7.85 (dd, J=3.6 & 1.2 Hz, 1H), 5.60 (m, 1H), 5.20 (m, 1H), 4.62 (m, 2H), 4.42 (m, 2H), 4.38 (m, 2H), 4.20 (m, 1H), 3.96 (m, 1H), 3.60 (m, 1H), 2.56 (m, 1H), 2.40 (m, 2H).
Example 363. {l-{l-[3-Fluoro-2-(trifluoromethyl)isonicotinoyl]-4-deuteropiperidin- 4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3- yl}acetonitrile
Figure imgf000227_0001
Step 1. l-(3-Fluoro-2-(trifluoromethyl)isonicotinoyl)piperidin-4-one
Figure imgf000228_0001
To a solution of piperidin-4-one (1 g, 10 mmol) and 3-fluoro-2- (trifluoromethyl)isonicotinic acid (2.32 g, 11.1 mmol) in DMF (20 mL) were added benzotriazol-l-yloxytris(dimethylamino)phosphonium hexafluorophosphate (5.62 g, 12.7 mmol) and triethylamine (4.43 mL, 31.8 mmol). The mixture was stirred at room temperature overnight and diluted with EtOAc. The solution was washed with brine, dried with sodium sulfate, filtered and concentrated. Purification on silica gel with 50- 100% EtOAc/hexanes gave 2.1 g (70% yield) of the title compound. LC/MS found: 291.2 (M+l)+.
Step 2. {l-{l-[3-Fluoro-2-(trifluoromethyl)isonicotinoylJ-4-deuteropiperidin-4-y (7H^yrrolof2 -dJpyrimidin-4-yl)-lH^yrazol-l-ylJazetidin-3-yl}acetonitrile
To a solution of {3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile (50 mg, 0.1 mmol) and 1- (3-fluoro-2-(trifluoromethyl)isonicotinoyl)piperidin-4-one (39 mg, 0.13 mmol) in tetrahydrofuran (3 mL, 40 mmol) was added sodium cyanoborodeuteride (0.024 g, 0.37 mmol). The mixture was stirred at room temperature overnight. The solvent was removed under reduced pressure. The resulting residue was taken up in EtOAc. The solution was washed with aqueous NaHC03 and brine, dried over Na2S04 and
concentrated under reduced pressure. Purification with prep-LCMS afforded 25 mg (36% yield) of the desired product as a solid. LC/MS found: 685.1 (M+l)+.
The above solid was dissolved in CH2C12 (2 mL). To it was added trifluoroacetic acid (2 mL). After being stirred at room temperature for 1 hour, the solution was concentrated. The residue was taken up in methanol (2 mL). Ethylenediamine (0.1 g, 2 mmol) was added. The mixture was stirred at room temperature for two hours. Direct purification with prep-LCMS (Method C) gave the title compound as a white powder. LCMS found: 555.1 (M+l)+. 1H NMR (400 MHz, CD3OD): δ 12.93 (s, 1H), 9.61 (s, 1H),
9.49 (s, 1H), 9.48 (d, J=4.4 Hz, 1H), 9.21 (s, 1H), 8.70 (dd, J=4.8 & 4.8 Hz, 1H), 8.40 (dd, J=3.2 & 2.4 Hz, 1H), 7.86 (d, J=2.8 Hz, 1H), 4.85 (m, 1H), 4.58 (m, 2H), 4.38 (m, 4H), 4.21 (m, 1H), 4.08 (m, 1H), 3.90 (m, 1H), 2.58 (m, 1H), 2.40 (m, 1H), 2.02 (m, 2H).
Example 364. {l-{l-[3-Fluoro-2-(trifluoromethyl)isonicotinoyl]3,3,4,5,5- pentadeuteropiperidin-4-yl}-3- [4-(7H-pyrrolo [2,3-d] pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin-3-yl}acetonitrile
Figure imgf000229_0001
A solution of l-(3-fluoro-2-(trifluoromethyl)isonicotinoyl)piperidin-4-one (0.66 g, 2.3 mmol) and triethylamine (0.45 g, 4.4 mmol) in methanol-d4 was stirred at room temperature overnight. To it were added {3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile (1.1 g, 2.7 mmol) and sodium cyanoborodeuteride (0.45 g, 6.8 mmol). The mixture was stirred at room temperature for 4 hours and concentrated. The residue was taken up in EtOAc. The solution was washed with aqueous NaHC03 and brine, dried over Na2S04 and concentrated under reduced pressure. Purification with prep-LCMS afforded 80 mg (5% yield) of the desired compound. LC/MS found: 589.1, (M+l)+.
The above product was dissolved in CH2C12 (2 mL). To it was added
trifluoroacetic acid (2 mL). After being stirred for 1 hour, the solution was concentrated. The residue was dissolved in methanol (3 mL). To it was added ethylenediamine (0.5 g, 8 mmol). After being stirred for 2 hours, the mixture was separated by prep-HPLC (Method
C) to give the title compound. LCMS found: 559.1 (M+l)+. 1H NMR (400 MHz, CD3OD): d 12.94 (s, 1H), 9.61 (s, 1H), 9.49 (s, 1H), 9.46 (d, J=4.4 Hz, 1H), 9.21 (s, 8.70 (dd, J=4.4 & 4.4 Hz, IH), 8.40 (d, J=3.6 Hz, IH), 7.86 (d, J=3.6 Hz, IH), 4.82 (d, J=13.2 Hz, IH), 4.54 (d, J=7.6 Hz, 2H), 4.39 (m, 4H), 4.20 (d, J=13.6 Hz, IH), 4.05 (d, J=132. Hz, IH), 3.85 (d, J=13.6 Hz, IH).
Example 365. {l-{7-[3-Fluoro-2-(trifluoromethyl)isonicotinoyl]-3-oxa-7- azabicyclo[3.3.1]non-9-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin-3-yl}acetonitrile
Figure imgf000230_0001
Step 1. {l-(3-Oxa-7-azabicyclo[3.3.1]non-9-yl)-3-[4-(7-{[2-
(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrim
ylJazetidin-3-yl}
Figure imgf000230_0002
To a mixture of {3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile (0.80 g, 1.95 mmol) and tert-butyl 9-oxo-3-oxa-7-azabicyclo[3.3.1]nonane-7-carboxylate (0.518 g, 2.148 mmol) in tetrahydrofuran (20.0 mL) were added triethylamine (1.62 mL, 11.7 mmol) and sodium triacetoxyborohydride (0.828 g, 3.91 mmol). After being stirred at room temperature for 2 hours, the reaction was quenched with 20 mL of water and 100 mL of EtOAc. The organic layer was washed with brine, dried over Na2S04, filtered and concentrated under reduced pressure. Purification with combi-fiash (5% MeOH/CH2Cl2) gave 0.8 g (65% yield) of the desired intermediate. LCMS found: 635.3 (M+l)+.
The above product was dissolved in THF (5 mL) and a 4 M solution of HC1 in dioxane (5 mL) was added. After being stirred for 1 hour, the solution was concentrated under reduced pressure to give the title compound. LCMS found: 535.2 (M+l)+.
Step 2. {l-{7-[ 3-Fluoro-2- (rifluoromethyl) isonicotinoyl] -3-oxa- 7-azabicyclof 3.3.1 ]non- 9-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}aceto
To a mixture of 3-fluoro-2-(trifluoromethyl)isonicotinic acid (50.0 mg, 0.239 mmol) and {l-(3-oxa-7-azabicyclo[3.3.1]non-9-yl)-3-[4-(7-{[2- (trimethylsilyl)ethoxy]methyl} -7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 - yl]azetidin-3-yl}acetonitrile (128 mg, 0.239 mmol) in DMF (2 mL) were added triethylamine (0.100 mL, 0.717 mmol) and benzotriazol-1- yloxytris(dimethylamino)phosphonium hexafluorophosphate (127 mg, 0.287 mmol). The mixture was stirred at room temperature for 2 hours. Direct purification with prep-LCMS (pH=10) afforded 50 mg (30% yield) of the desired product. LCMS found: 696.3 (M+l)+.
The about product was dissolved in methylene chloride (2 mL). To it was added TFA (2 mL). After being stirred for 1 hour, the solution was concentrated. The residue was taken up in a solution of 20% ethylenediamine in MeOH (2 mL). After being stirred for 2 hours, the mixture was separated with prep-HPLC (Method C) to give the title compound. LCMS found: 596.1 (M+l)+. 1HNMR (300 MHz, DMSO-d6): δ 12.10 (s, 1H), 8.82 (s, 1H), 8.68 (s, 1H), 8.66 (d, J=6.1 Hz, 1H), 8.41 (s, 1H), 7.63 (s, 1H), 7.59 (d, J=7.1 Hz, 1H), 7.05 (d, J=6.3 Hz, 1H), 4.79 (d, J=9.1 Hz, 1H), 4.44 (d, J=10.1 Hz, 1H), 4.00-3.80 (m, 2H), 3.77-3.41 (m, 4H), 3.60 (s, 2H), 3.39-3.18 (m, 4H), 3.01 (m, 1H), 1.60 (m, 2H).
Example 366. {l-(l-{[4-[(dimethylamino)methyl]-6-(trifluoromethyl)pyridin-2- yl] carbonyl}piperidin-4-yl)-3- [4-(7H-pyrrolo [2,3-d] pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin-3-yl}acetonitrile
\
Figure imgf000232_0001
Step A. 4-[(dimethylamino)methyl]-6-(trifluoromethyl)pyridine-2-carboxy acid
Figure imgf000232_0002
A mixture of 4-bromo-6-(trifluoromethyl)pyridine-2-carboxylic acid (200 mg, 0.7 mmol, Anichem), cesium carbonate (724 mg, 2.22 mmol), dicyclohexyl(2',4',6'- triisopropylbiphenyl-2-yl)phosphine (42 mg, 0.089 mmol, Aldrich), potassium
[(dimethylamino)methyl](trifluoro)borate(l-) (147 mg, 0.889 mmol, Aldrich), palladium acetate (10. mg, 0.044 mmol, Sigma-Aldrich) and THF:H20 (10: 1, 4.6 mL) was degassed by bubbling a stream of nitrogen through the solution for 15 minutes. The reaction vial was sealed and heated at 80 °C overnight. The crude mixture was purified via HPLC (Waters XBridge CI 8, 5um particle size, 30 x 100 mm; 5 to 25 % MeCN/H20 containing 0.15 % NH4OH over 5 minutes) and the product was detected by UV absorbance and collected. Fractions containing the desired product were rotovapped and azeotroped once with methanol to afford product. Yield: 0.029 g (20%); LC-MS: 249.1 (M+H)
Step B. {l-(l-{[4-[ (dimethylamino)methyl]-6-(trifluoromethyl)pyridin-2- yl]carbonyl}piperidin-4-yl)-3-[4-(7H^yrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin-3-yl}acetonitrile
To a solution of 4-[(dimethylamino)methyl]-6-(trifluoromethyl)pyridine-2- carboxylic acid (0.015 g, 0.060 mmol, from Step A) in Ν,Ν-dimethylformamide (1 mL) was added N,N,N',N'-tetramethyl-0-(7-azabenzotriazol-l-yl)uronium
hexafluorophosphate (0.028 g, 0.072 mmol, Aldrich) and N,N-diisopropylethylamine (0.042 mL, 0.24 mmol) . This mixture was prestirred for 15 minutes, followed by the addition of {l-piperidin-4-yl-3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile (0.030 g, 0.061 mmol, prepared as described in Example 1, Step H, except worked up to provide the free base) . The reaction was stirred overnight. Additional N,N-diisopropylethylamine (0.042 mL, 0.24 mmol), 4-[(dimethylamino)methyl]-6-(trifluoromethyl)pyridine-2-carboxylic acid (0.014 g, 0.056 mmol) in DMF (1 mL), and N,N,N*,N*-tetramethyl-0-(7- azabenzotriazol-l-yl)uronium hexafluorophosphate (0.018 g, 0.048 mmol) were added and the reaction was stirred over an additional 48 hours. The reaction mixture was then partitioned between ethyl acetate and water and the layers were separated. Solid NaCl was added to saturate the aqueous layer and this was extracted with two further portions of ethyl acetate. The combined organic extracts were dried over sodium sulfate, decanted and concentrated. The crude product was deprotected by stirring with a 1 : 1 mixture of TFA:DCM for 1.5 hours, followed by evaporation and then stirring the resulting residue with ethylenediamine (0.2 mL) in methanol (4 mL). The mixture was filtered and purified by preparative HPLC-MS, (C 18 eluting with a gradient of MeCN/H20 containing 0.15 % NH4OH). Fractions containing product were frozen and lyophilized. Yield: 0.015 g (42%); LC-MS: 593.2 (M+H)+. 1H NMR (400 MHz, CD3OD): δ 8.70 (s, 1H), 8.67 (s, 1H), 8.40 (s, 1H), 7.88 (s, 1H), 7.80 (s, 1H), 7.51 (d, 1H), 6.99 (d, 1H), 4.32 (ddd, 1H), 3.86-3.70 (m, 5H), 3.64 (s, 2H), 3.50 (s, 2H), 3.36-3.16 (m, 2H), 2.69-2.61 (m, 1H), 2.28 (s, 6H), 1.98-1.88 (m, 1H), 1.86-1.77 (m, 1H), 1.51-1.39 (m, 2H). 19F NMR (400 MHz, CD3OD): δ -69.82 (s, 3F). Example 367. 4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH- pyrazol-l-yl]azetidin-l-yl}-N-isopropylpiperidine-l-carboxamide
Figure imgf000234_0001
To a solution of {l-piperidin-4-yl-3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-
7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile (0.075 g, 0.15 mmol, prepared as described in Example 1, Step H, except worked up to provide the free base) in methylene chloride (2 mL) was added N,N-diisopropylethylamine (0.026 mL, 0.15 mmol) followed by 2-isocyanatopropane (20 μί, 0.2 mmol, Aldrich). The reaction was continued for 2 hours. The crude product was deprotected by the addition of 1 mL TFA to the solution, which was stirred for 1 hour and evaporated. The deprotection was completed by stirring with ethylenediamine (0.2 mL) in methanol for 30 minutes. The product was purified by preparative HPLC-MS (CI 8 eluting with a gradient of MeCN/LLO containing 0.15 % ΝΗ4ΟΗ). Fractions containing desired product were frozen and lyophilized. Yield: 0.025 g (37%); LC-MS: 448.2 (M+H)+. 1H NMR (300
MHz, CDCI3): δ 10.21 (br s, 1H), 8.85 (s, 1H), 8.41 (s, 1H), 8.32 (s, 1H), 7.41 (dd, 1H), 6.79 (dd, 1H), 4.22 (d, 1H), 4.04-3.87 (m, 1H), 3.81-3.69 (m, 4H), 3.61 (d, 2H), 3.40 (s, 2H), 3.00-2.86 (dq, 2H), 2.44-2.31 (m, 1H), 1.76-1.64 (m, 2H), 1.40-1.22 (m, 2H), 1.15 (d, 6H).
Example 368. {l-{l-[6-[(dimethylamino)methyl]-3-fluoro-2-
(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile
Figure imgf000235_0001
Step A. 3-fluoro-2-(trifluoromethyl)isonicotinic acid 1 -oxide
Figure imgf000235_0002
To a solution of 3-fluoro-2-(trifluoromethyl)isonicotinic acid (2.00 g, 9.56 mmol, Oakwood) and urea hydrogen peroxide addition compound (5.00 g, 53.2 mmol,
Aldrich) in methylene chloride (50 mL) at 0 °C was added trifluoroacetic anhydride (7.51 mL, 53.2 mmol). The bath was removed and the reaction was allowed to warm to room temperature and stir overnight. The precipitate was filtered off. The filtrate was diluted with a small amount of water and ethyl acetate, the layers were separated and the aqueous was extracted with two further portions of ethyl acetate. The combined extracts were dried over sodium sulfate, filtered and concentrated. The solvent was removed in vacuo to afford a yellow solid which was used without further purification. LC-MS: 225.9 (M+H)+. 1H NMR (300 MHz, CD3OD): δ 8.20 (d, 1H), 8.04 (dd, 1H). Step B. 6-chloro-3-fluoro-2-(trifluoromethyl)isonicotinic acid
Figure imgf000235_0003
A solution of 3-fluoro-2-(trifluoromethyl)isonicotinic acid 1 -oxide (1.0 g, 4.4 mmol from Step A) in phosphoryl chloride (4 mL, 40 mmol) was heated to 110 °C for 2 hours, then was left to stir at ambient temperature overnight. The POCI3 was evaporated, the residue was treated with sodium bicarbonate solution, which was stirred for 1 hour. To this mixture was added tetrahydrofuran (20 mL) and lithium hydroxide monohydrate (0.24 g, 5.7 mmol). This was stirred for 3 hours. The pH of the mixture was then adjusted to the range of pH 4-5 by the addition of c.HCl. The product was extracted with three portions of ethyl acetate. The combined extracts were dried over sodium sulfate, filtered and concentrated. Yield: 0.54 g (50%); LC-MS: 244.1/245.9 (M+H)+. 1H NMR (300 MHz, CDCI3): δ 8.07 (d, 1H).
Step C. 6-[(dimethylamino)methylJ-3- uoro-2-(trifluoromethyl)isonicotinic acid
Figure imgf000236_0001
To a sealable vial was added 6-chloro-3-fluoro-2-(trifluoromethyl)isonicotinic acid (0.200 g, 0.821 mmol, from Step B), Palladium acetate (0.13 g, 0.57 mmol,
Aldrich), dicyclohexyl(2',4',6'-triisopropylbiphenyl-2-yl)phosphine (0.55 g, 1.1 mmol, Aldrich), Cesium Carbonate (0.803 g, 2.46 mmol) and potassium
[(dimethylamino)methyl](trifluoro)borate(l-) (0.163 g, 0.985 mmol, Aldrich) and THF:H20 (10: 1, 15 mL). The reaction was degassed by alternating vacuum and N2 in three cycles. The vial was sealed and heated to 80 °C overnight. The mixture was filtered and the solvent was removed in vacuo. The product was purified by preparative HPLC- MS (CI 8 eluting with a gradient of MeCN/H20 containing 0.1 % TFA). Fractions containing product were rotovapped to afford a residue comprising product and DMF. The weight percent of each component was determined by NMR and the product used without further purification. Yield: 0.077 g (35%); 1H NMR (400 MHz, CD3OD): δ 8.26 (d, 1H), 4.60 (s, 2H), 2.98 (s, 6H).
Step D. {l-{l-[6-[(dimethylamino)methyl]-3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo
lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile
A solution of 6-[(dimethylamino)methyl]-3-fluoro-2-(trifluoromethyl)isonicotinic acid (0.0776 g, 0.291 mmol, from Step C) in Ν,Ν-dimethylformamide (1 mL) was treated with N,N-diisopropylethylamine (0.3 mL, 2 mmol) and benzotriazol-1- yloxytris(dimethylamino)phosphonium hexafluorophosphate (0.15 g, 0.35 mmol) . This mixture was pre-stirred for 1 hour, followed by the addition of {l-piperidin-4-yl-3-[4-(7- { [2-(trimethylsilyl)ethoxy]methyl} -7H-pyrrolo [2,3 -d]pyrimidin-4-yl)- 1 H-pyrazol- 1 - yl]azetidin-3-yl}acetonitrile (0.158 g, 0.320 mmol, prepared as described in Example 1, Step H, except worked up to provide the free base). After 3 hours, additional N,N- diisopropylethylamine (0.507 mL, 2.91 mmol) and benzotriazol-1- yloxytris(dimethylamino)phosphonium hexafluorophosphate (0.26 g, 0.58 mmol) were added and the mixture was stirred for 48 hours. The SEM-protected product was purified by preparative HPLC-MS (CI 8 eluting with a gradient of MeCN/H20 containing 0.15 % NH4OH). Solvent was removed from fractions containing desired product by rotary evaporation. The product was deprotected by stirring with a 1 : 1 mixture of TFA:DCM for 1 hour, followed by evaporation, and stirring with ethylenediamine (0.2 mL) in methanol for 30 minutes. The product was purified by preparative HPLC-MS (CI 8 eluting with a gradient of MeCN/H20 containing 0.15 % NH4OH). Fractions containing desired product were frozen and lyophilized. Yield: 0.006 g (3%); LC-MS: 611.2 (M+H)+. 1H NMR (300 MHz, CD3OD): δ 8.69 (s, 1H), 8.67 (s, 1H), 8.40 (s, 1H), 7.77 (d, 1H), 7.51 (d, 1H), 6.98 (d, 1H), 4.33-4.21 (m, 1H), 3.87-3.71 (m, 4H), 3.68 (s, 2H), 3.59-3.45 (m, 3H), 3.44-3.10 (m, 2H), 2.71-2.58 (m, 1H), 2.31 (s, 6H), 1.97-1.85 (m, 1H), 1.85-1.72 (m, 1H), 1.56- 1.24 (m, 2H). 19F NMR (300 MHz, CD3OD): δ -67.26 (d, 3F), -132.8 (m, IF).
Example 369. 3- [(4- {3-(cyanomethyl)-3- [4-(7H-pyrrolo [2,3-d] yrimidin-4-yl)- 1H- pyrazol-l-yl]azetidin-l-yl}piperidin-l-yl)carbonyl]-5-
[(dimethylamino)methyl]benzonitrile
Figure imgf000238_0001
Step A. methyl 3-bromo-5-[(dimeth lamino)methyl]benzoate
Figure imgf000238_0002
To a solution of methyl 3-bromo-5-formylbenzoate (1.8 g, 7.4 mmol, prepared as described in WO 2003048111 from dimethyl 5-bromoisophthalate, Alfa Aesar) in methylene chloride (20 mL) was added a solution of 2.0 M dimethylamine in
tetrahydrofuran (7.4 mL, 15 mmol). This mixture was stirred for 15 minutes, followed by the addition of sodium triacetoxyborohydride (4.7 g, 22 mmol). The resulting mixture was stirred overnight. Saturated sodium bicarbonate solution was added and the product was extracted with ethyl acetate. The combined organic extracts were washed twice with water, once with brine, dried over sodium sulfate, filtered and concentrated to afford a light yellow oil. Yield: 1.87 g (93%); LC-MS: 272.0, 274.0 (M+H)+. 1H NMR (400 MHz, CDCI3): δ 8.06 (dd, 1H), 7.89 (dd, 1H), 7.69 (dd, 1H), 3.91 (s, 3H), 3.42 (s, 2H), 2.24 (s, 6H).
Step B. 3-bromo-5-[(dimethylamino)methyl]benzoic
Figure imgf000239_0001
Methyl 3-bromo-5-[(dimethylamino)methyl]benzoate (0.30 g, 1.1 mmol, from Step A) was dissolved in tetrahydrofuran (20 mL) and lithium hydroxide monohydrate (0.555 g, 13.2 mmol) in water (6 mL) was added. After stirring for 3 hours, the mixture was rotovapped to remove THF and reduce the volume of water. The mixture was diluted with an equivalent volume of acetonitrile and filtered. The product was purified via preparative HPLC-MS (CI 8 eluting with a gradient of MeCN/H20 containing 0.15 % NH4OH). Solvent was removed from fractions containing desired product by rotary evaporation to afford a white solid. Yield: 0.26 g (91%); LC-MS: 258.0, 260.0 (M+H)+. 1H NMR (300 MHz, DMSO-d6): δ 7.86 (dd, 1H), 7.76 (dd, 1H), 7.43 (dd, 1H), 3.38 (s, 2H), 2.14 (s, 6H).
Step C. 3-[(4-{3-(cyanomethyl)-3-[ 4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-l-yl}piperidin-l-yl)carbonyl]-5-[(dimethylamino)meth^
To a solution of 3-bromo-5-[(dimethylamino)methyl]benzoic acid (31.4 mg, 0.122 mmol, from Step B) in tetrahydrofuran (1.0 mL) was added triethylamine (0.045 mL, 0.32 mmol) and N,N,N',N'-tetramethyl-0-(7-azabenzotriazol-l-yl)uronium hexafluorophosphate (43.2 mg, 0.114 mmol). The mixture was pre-stirred for 15 minutes, followed by the addition of {l-piperidin-4-yl-3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}- 7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile (40. mg, 0.081 mmol, prepared as described in Example 1, Step H, except worked up to provide the free base). After stirring for two hours, the reaction mixture was partitioned between ethyl acetate and water. The organic layer was washed successively with water, 0.1N NaOH and sat. NaCl, dried over sodium sulfate and concentrated. The residue was dissolved in N,N-dimethylformamide (1.50 mL) and zinc cyanide (57 mg, 0.49 mmol) was added. The solution was degassed via a stream of nitrogen through the mixture for 10 minutes. Tetrakis(triphenylphosphine)palladium(0) (19 mg, 0.016 mmol) was added, and the reaction was heated to 120 °C in the microwave for 30 minutes. The reaction mixture was worked up by partition between water and ethyl acetate. The ethyl acetate layer was washed twice with water, once with brine, dried over sodium sulfate and concentrated. The residue was stirred in a 1 : 1 mixture of DCM:TFA for one hour, then concentrated. The residue was redissolved in methanol (1 mL), and ethylenediamine (0.2 mL) was added. Upon complete deprotection, the product was isolated via preparative HPLC-MS (CI 8 eluting with a gradient of MeCN/H20
containing 0.15 % NH4OH). Yield: 11.7 mg (26%); LC-MS: 549.3 (M+H)+. 1H NMR (400 MHz, DMSO-dg): δ 12.15 (br s, 1H), 8.82 (s, 1H), 8.69 (s, 1H), 8.42 (s, 1H), 7.85 (s, 1H), 7.82 (s, 1H), 7.66 (s, 1H), 7.61 (dd, 1H), 7.07 (dd, 1H), 4.14-4.03 (m, 1H), 3.80-3.28 (m, 9H), 3.22-3.02 (m, 2H), 2.59-2.51 (m, 1H), 1.83-1.71 (m, 1H), 1.69-1.58 (m, 1H), 1.35-1.15 (m, 2H).
Example 370. {l-(l-{[6-[(dimethylamino)methyl]-2-(trifluoromethyl)pyrimidin-4- yl] carbonyl}piperidin-4-yl)-3- [4-(7H-pyrrolo [2,3-d] pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-3-yl}acetonitrile
Figure imgf000240_0001
Step A. ethyl 6-(bromomethyl)-2-(tri uoromethyl)pyrimidine-4-carboxylate
Figure imgf000240_0002
A mixture of ethyl 6-methyl-2-(trifluoromethyl)pyrimidine-4-carboxylate (1.1 g, 4.5 mmol, prepared as described in WO 2007/090748), N-bromosuccinimide (2.86 g, 16.1 mmol) and benzoyl peroxide (0.21 g, 0.9 mmol) in carbon tetrachloride (9 mL) was heated in a sealed vessel to 100 °C overnight. The mixture was diluted with
dichloromethane (DCM), filtered and the solvent was removed in vacuo. Purification via preparative HPLC-MS (CI 8 eluting with a gradient of MeCN/H20 containing 0.15 % NH4OH) afforded the product, which on removal of solvent, was an oil.
Yield: 0.34 g (24%); LC-MS: 313.0, 315.0 (M+H)+. 1H NMR (400 MHz, DMSO-d6): δ 8.48 (s, 1H), 4.91 (s, 2H), 4.44 (q, 2H), 1.36 (t, 3H).
Step B. 6-[(dimethylamino)methylJ-2- trifluoromethyl)pyrimidine-4-carboxylic acid
Figure imgf000241_0001
To a solution of 2.0 M dimethylamine in THF (5.27 mL, 10.5 mmol) was added a solution of ethyl 6-(bromomethyl)-2-(trifluoromethyl)pyrimidine-4-carboxylate (0.33 g, 1.0 mmol, from Step A) in methylene chloride (5.0 mL). The reaction was stirred at room temperature for 2 hours, then concentrated. The residue was dissolved in tetrahydrofuran (20 mL), water (6 mL) was added, followed by lithium hydroxide monohydrate (0.4 g, 10 mmol). The mixture was stirred for one hour, then was rotovapped to remove most of the THF. The pH was adjusted to 7 by the addition of cone. HC1. Acetonitrile (10 mL) was added, the mixture was filtered and then purified via preparative HPLC-MS (CI 8 eluting with a gradient of MeCN/H20 containing 0.15 % NH4OH) to afford product as a light yellow solid. Yield: 0.153 g (58%); LC-MS: 250.1 (M+H)+. 1H NMR (400 MHz, DMSO-de): δ 8.02 (s, 1H), 3.70 (s, 2H), 2.27 (s, 6H).
Step C. {1 -(!-{[ 6-f (dimethylamino)methyl]-2-(trifluoromethyl)pyrimidin-4- yl]carbonyl}piperidin-4-yl)-3-[4-(7H^yrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- ylJazetidin-3-yl) acetonitrile 6-[(Dimethylamino)methyl]-2-(trifluoromethyl)pyrimidine-4-carboxylic acid (22.8 mg, 0.0913 mmol, from Step B) was dissolved in tetrahydrofuran (0.67 mL), and triethylamine (33.9 μί, 0.244 mmol) and N,N,N',N'-tetramethyl-0-(7-azabenzotriazol-l- yl)uronium hexafluorophosphate (32.4 mg, 0.0852 mmol) were added. The mixture was stirred for 15 minutes, followed by the addition of {l-piperidin-4-yl-3-[4-(7-{[2- (trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-3-yl}acetonitrile (30.0 mg, 0.0609 mmol, prepared as described in Example 1, Step H, except worked up to provide the free base). After two hours, the reaction mixture was partitioned between ethyl acetate and water. The organic layer was washed with water, 0. IN NaOH and sat. NaCl, dried over sodium sulfate and concentrated. The residue was stirred in a 1 : 1 mixture of DCM:TFA for 1 hour, concentrated, and stirred in methanol (1 mL) containing ethylenediamine (0.2 mL) until the deprotection was complete. Purification via preparative HPLC-MS (CI 8 eluting with a gradient of
MeCN/H20 containing 0.15 % NH4OH), afforded the desired compound as a white powder. Yield: 0.014 g (39%); LC-MS: 594.4 (M+H)+. 1H NMR (400 MHz, DMSO-d6): δ 12.14 (br s, 1H), 8.82 (s, 1H), 8.69 (s, 1H), 8.42 (s, 1H), 7.90 (s, 1H), 7.61 (dd, 1H), 7.07 (dd, 1H), 4.07 (ddd, 1H), 3.77-3.72 (m, 2H), 3.70 (s, 2H), 3.63-3.48 (m, 5H), 3.31- 3.22 (m, 1H), 3.16-3.07 (m, 1H), 2.60-2.53 (m, 1H), 2.25 (s, 6H), 1.85-1.73 (m, 1H), 1.71-1.59 (m, 1H), 1.37-1.18 (m, 2H). 19F NMR (400 MHz, DMSO-d6): δ -69.45 (s, 3F).
Example 371. {l-(l-{[6-[(methylamino)methyl]-2-(trifluoromethyl)pyrimidin-4- yl] carbonyl}piperidin-4-yl)-3- [4-(7H-pyrrolo [2,3-d] pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-3-yl}acetonitrile trifluoroacetate salt
Figure imgf000243_0001
Step A. 6-[(methylamino)methylJ-2- trifluoromethyl)pyrimidine-4-carboxylic acid
Figure imgf000243_0002
A solution of methylamine (33 wt% in Ethanol, 1.12 mmol, Aldrich) was added portionwise to a solution of ethyl 6-(bromomethyl)-2-(trifluoromethyl)pyrimidine-4- carboxylate (0.150 g, 0.321 mmol, Example 5, Step A) in methylene chloride (3.0 mL) , until reaction was complete. Solvent was removed in vacuo. The residue was dissolved in tetrahydrofuran (7.0 mL) and water (2.0 mL), and lithium hydroxide monohydrate (0.135 g, 3.21 mmol) was added. After a 5 minute reaction time, the mixture was treated with IN HCl to adjust the pH to 7, then was purified using preparative HPLC-MS (CI 8 eluting with a gradient of MeCN/H20 containing 0.15% NH4OH). LC-MS: 236.1 (M+H)+.
Step B. {l-(l-{[ 6-f (methylamino)methyl]-2-(trifluoromethyl)pyrimidin-4- yl]carbonyl}piperidin-4-yl)-3-[4-(7H^yrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- ylJazetidin-3-yl}acetonitrile
N,N,N',N'-Tetramethyl-0-(7-azabenzotriazol- 1 -yl)uranium hexafluorophosphate (32.4 mg, 0.0852 mmol) was added to a mixture of 6-[(methylamino)methyl]-2- (trifluoromethyl)pyrimidine-4-carboxylic acid (21.5 mg, 0.0913 mmol, from Step A), {1- piperidin-4-yl-3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile (30.0 mg, 0.0609 mmol, prepared as described in Example 1 , Step H, except worked up to provide the free base) and triethylamine (33.9 μί, 0.244 mmol) in acetonitrile (0.30 mL) and THF (0.67 mL) and the reaction was stirred overnight. The reaction mixture was partitioned between ethyl acetate and water. The organic layer was separated and was washed successively with water, 0.1N NaOH and sat. NaCl, dried over sodium sulfate, filtered and concentrated. The residue was then stirred in a 1 : 1 mixture of DCM:TFA for 1 hour, concentrated, and subsequently stirred with ethylenediamine (0.2 mL) in methanol (1 mL) until
deprotection was complete. Purification first via preparative HPLC-MS (CI 8 eluting with a gradient of MeCN/H20 containing 0.15% NH4OH) then again eluting with MeCN/H20 containing 0.1% TFA afforded desired product as the trifluoroacetate salt. Yield: 0.0015 g (3%); LC-MS: 580.4 (M+H)+.
Example 372. {l-(l-{[6-(azetidin-l-ylmethyl)-2-(trifluoromethyl)pyrimidin-4- yl] carbonyl}piperidin-4-yl)-3- [4-(7H-pyrrolo [2,3-d] pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin-3-yl} acetonitrile
Figure imgf000244_0001
Step A. Ethyl 6-(azetidin-l-ylmeth l)-2-(trifluoromethyl)pyrimidine-4-carboxy
Figure imgf000244_0002
O Azetidine (0.110 mL, 1.6 mmol, Aldrich) was added to a solution of ethyl 6- (bromomethyl)-2-(trifluoromethyl)pyrimidine-4-carboxylate (0.78 g, 1.1 mmol, prepared as in Example 376, Step A) in DCM (11 mL). After stirring for 20 minutes, additional azetidine (0.10 mL, 1 mmol) was added. After 10 minutes, excess reagents and solvent were removed in vacuo. Flash chromatography, eluting with a gradient of 0-5% MeOH in DCM afforded purified product. Yield: 0.29 g (87%); LC-MS: 290.1 (M+H)+. 1H NMR (400 MHz, CDCls): δ 8.24 (s, 1H), 4.52 (q, 2H), 3.90 (s, 2H), 3.39 (t, 4H), 2.19 (quin, 2H), 1.45 (t, 3H).
Step B. 6-(azetidin-l-ylmethyl)-2-(trifluoromethyl)pyrimidine-4-carboxylic acid hydrochloric acid salt
Figure imgf000245_0001
HC1
Ethyl 6-(azetidin- 1 -ylmethyl)-2-(trifluoromethyl)pyrimidine-4-carboxylate (0.34 g, 1.2 mmol, from Step A) was dissolved in THF (6.0 mL) and water (1.5 mL) and lithium hydroxide monohydrate (0.108 g, 2.57 mmol) was added. After 15 minutes, THF was removed in vacuo and the mixture was treated with a solution of 1 N HC1 (5.3 mL) and acetonitrile (7.0 mL). The mixture was then filtered and concentrated to afford a yellow solid, theoretical yield assumed. LC-MS: 262.1 (M+H)+.
Step C. {1 -(!-{[ 6-(azetidin-l-ylmethyl)-2-(trifluoromethyl)pyrimidin-4- yl]carbonyl}piperidin-4-yl)-3-[4-(7H^yrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- ylJazetidin-3-yl) acetonitrile
{l-Piperidin-4-yl-3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile (0.150 g, 0.30 mmol, prepared as described in Example 1 , Step H, except worked up to provide the free base) was added to a mixture of 6-(azetidin-l-ylmethyl)-2-(trifluoromethyl)pyrimidine-4- carboxylic acid (0.20 g, 0.46 mmol, as the hydrochloride salt from Step B), triethylamine (0.255 mL, 1.83 mmol) and N,N,N',N'-tetramethyl-0-(7-azabenzotriazol-l-yl)uronium hexafluorophosphate (0.150 g, 0.396 mmol) in tetrahydrofuran (3.0 mL) and DCM (3.0 mL) that was pre-stirred for 30 minutes. After stirring for 2 hours, the reaction mixture was diluted with ethyl acetate and washed successively with water, 0.1 N NaOH, and brine. The organic phase was dried over sodium sulfate, filtered and concentrated. The residue was dissolved in a 1 : 1 mixture of DCM:TFA, stirred for 1 hour, concentrated again, then stirred with methanol (3 mL) containing ethylenediamine (0.2 mL). After complete deprotection, the product was purified via preparative HPLC-MS (CI 8 eluting with a gradient of MeCN/H20 containing 0.1 % TFA) then again eluting with
MeCN/H20 containing 0.15% NH4OH to afford desired product. Yield: 0.043 g (23%); LC-MS: 606.2 (M+H)+. 1H NMR (400 MHz, DMSO-d6): δ 12.15 (br s, 1H), 8.82 (s, 1H), 8.69 (s, 1H), 8.42 (s, 1H), 7.81 (s, 1H), 7.61 (d, 1H), 7.06 (d, 1H), 4.10-4.01 (m, 1H), 3.81 (s, 2H), 3.75 (dd, 2H), 3.62-3.44 (m, 5H), 3.31-3.21 (m, 5H), 3.14-3.05 (m, 1H), 2.60-2.52 (m, 1H), 2.04 (quin, 2H), 1.83-1.73 (m, 1H), 1.69-1.60 (m, 1H), 1.37-1.18 (m, 2H). 19F NMR (400 MHz, CD3OD): δ -72.38 (s, 3F).
Example 373. {l-(l-{[6-[(diethylamino)methyl]-2-(trifluoromethyl)pyrimidin-4- yl] carbonyl}piperidin-4-yl)-3- [4-(7H-pyrrolo [2,3-d] pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin-3-yl}acetonitrile
Figure imgf000246_0001
Step A. 6-[(diethylamino)methyl]-2-(trifluoromethyl)pyrimidine-4-carboxylic acid
Figure imgf000247_0001
To a solution of ethyl 6-(bromomethyl)-2-(trifluoromethyl)pyrimidine-4- carboxylate (0.15 g, 0.32 mmol, prepared as in Example 5, Step A) in methylene chloride (3.0 mL) was added N-ethylethanamine (0.13 mL, 1.3 mmol). After 30 minutes, the solvent was removed in vacuo. The ester was hydro lyzed by stirring with lithium hydroxide monohydrate (0.12 g, 3.0 mmol) in a mixture of tetrahydrofuran (5 mL) and water (2 mL). After 1 hour, IN HC1 was added dropwise to neutralize. Purification via preparative HPLC-MS (CI 8 eluting with a gradient of MeCN/H20 containing 0.15% NH4OH) afforded product as a light yellow solid. Yield: 0.050 g (60%); LC-MS: 278.0 (M+H)+. 1H NMR (400 MHz, DMSO-d6): δ 7.97 (s, 1H), 3.74 (s, 2H), 2.52 (q, 4H), 0.99 (t, 6H).
Step B. {l-(l-{[ 6-f (diethylamino)methyl]-2-(trifluoromethyl)pyrimidin-4- yl]carbonyl}piperidin-4-yl)-3-[4-(7H^yrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin-3-yl}acetonitrile
{l-Piperidin-4-yl-3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile (30.0 mg, 0.0609 mmol, prepared as described in Example 1 , Step H, except worked up to provide the free base) was added to a mixture of 6-[(diethylamino)methyl]-2-(trifluoromethyl)pyrimidine-4- carboxylic acid (22.8 mg, 0.0822 mmol, from Step A), triethylamine (33.9
Figure imgf000247_0002
0.244 mmol) and N,N,N',N'-tetramethyl-0-(7-azabenzotriazol- 1 -yl)uronium
hexafluorophosphate (28.9 mg, 0.0761 mmol) in tetrahydrofuran (0.67 mL), that was prestirred for 30 minutes. After stirring overnight, the reaction mixture was partitioned between ethyl acetate and water. The organic layer was washed successively with water, 0.1N NaOH and sat. NaCl, dried over sodium sulfate, filtered and concentrated. The residue was stirred in a 1 :1 mixture of DCM:TFA for 1 hour, and solvents were removed in vacuo. The residue was then stirred with ethylenediamine (0.2 mL) in methanol (1 mL) until deprotection was complete. Purification via preparative HPLC-MS (MeCN/H20 containing 0.15% NH4OH) afforded product. Yield: 0.0156 g (41%); LC-MS: 622.2 (M+H)+. 1H NMR (400 MHz, DMSO-d6): δ 12.14 (br s, 1H), 8.82 (s, 1H), 8.69 (s, 1H), 8.42 (s, 1H), 7.91 (s, 1H), 7.61 (d, 1H), 7.06 (d, 1H), 4.06 (ddd, 1H), 3.82 (s, 2H), 3.75 (d, 2H), 3.62-3.46 (m, 5H), 3.31-3.21 (m, 1H), 3.16-3.06 (m, 1H), 2.55 (q, 4H), 1.85-1.74 (m, 1H), 1.70-1.60 (m, 1H), 1.37-1.18 (m, 2H), 0.98 (t, 6H). 19F NMR (400 MHz, DMSO-de): δ -69.46 (s, 3F).
Example 374. {l-(l-{[6-{[ethyl(methyl)amino]methyl}-2-(trifluoromethyl)pyrimidin- 4-yl]carbonyl}piperidin-4-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin-3-yl}acetonitrile
Figure imgf000248_0001
Step A. 6-{fethyl(methyl)aminoJmeth l}-2-(trifluoromethyl)pyrimidine-4-carboxylw acid
Figure imgf000248_0002
N-methylethanamine (96 μί, 1.1 mmol) was added to a solution of ethyl 6- (bromomethyl)-2-(trifluorometliyl)pyrimidine-4-carboxylate (0.13 g, 0.28 mmol, prepared as in Example 5, Step A) in methylene chloride (2.6 mL). After stirring for 30 minutes, solvent was removed in vacuo. The ester was hydrolyzed by stirring with lithium hydroxide monohydrate (0.12 g, 2.8 mmol) in tetrahydrofuran (4 mL) and water (2 mL) for 1 hour. IN HC1 was added dropwise to adjust the pH to 7. Purifiation via preparative HPLC-MS (CI 8 eluting with a gradient of MeCN/H20 containing 0.15% NH4OH) afforded product. Yield: 0.043 g (58%); LC-MS: 264.1 (M+H)+. 1H NMR (300 MHz, DMSO-dg): δ 8.03 (s, 1H), 3.75 (s, 2H), 2.51 (q, 2H), 2.22 (s, 3H), 1.05 (t, 3H). Step B. {l-(l-{[6-{[ethyl(methyl)amino]methyl}-2-(trifluoromethyl)pyrimidin-4- yl]carbonyl}piperidin-4-yl)-3-[4-(7H^yrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin-3-yl}acetonitrile
After prestirring a mixture of 6-{[ethyl(methyl)amino]methyl}-2- (trifluoromethyl)pyrimidine-4-carboxylic acid (40.1 mg, 0.152 mmol, from Step A), triethylamine (56.6
Figure imgf000249_0001
0.406 mmol) and N,N,N',N'-tetramethyl-0-(7-azabenzotriazol-l- yl)uronium hexafluorophosphate (54.0 mg, 0.142 mmol) in tetrahydrofuran (1.1 mL) for 30 minutes, {l-piperidin-4-yl-3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile (50.0 mg, 0.101 mmol, prepared as described in Example 1, Step H, except worked up to provide the free base) was added and the reaction stirred overnight. The reaction mixture was partitioned between ethyl acetate and water. The organic layer was successively washed with water, 0.1N NaOH and sat. NaCl, dried over sodium sulfate, filtered and concentrated. The product was deprotected by stirring first in a 1 : 1 mixture of DCM:TFA for 1 hour, followed by evaporation and stirring with ethylenediamine (0.2 mL) in methanol (1 mL) until deprotection was complete. Purification via preparative HPLC-MS (CI 8 eluting with a gradient of MeCN/H20 containing 0.15% NH4OH) afforded product. Yield: 0.025 g (41%); LC-MS: 608.2 (M+H)+. 1H NMR (400 MHz, DMSO-d6): δ 12.14 (br s, 1H), 8.82 (s, 1H), 8.69 (s, 1H), 8.42 (s, 1H), 7.89 (s, 1H), 7.61 (d, 1H), 7.06 (d, 1H), 4.11-4.02 (m, 1H), 3.78-3.71 (m, 4H), 3.63-3.48 (m, 5H), 3.32-3.21 (m, 1H), 3.17-3.07 (m, 1H), 2.62-2.53 (m, 1H), 2.48 (q, 2H), 2.21 (s, 3H), 1.84-1.74 (m, 1H), 1.70-1.61 (m, 1H), 1.37- 1.18 (m, 2H), 1.03 (t, 3H). 19F NMR (400 MHz, DMSO-d6): δ -69.45 (s, 3F).
Example 375. {l-(l-{3-(difluoromethyl)-5-
[(dimethylamino)methyl]benzoyl}piperidin-4-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin- 4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile
Figure imgf000250_0001
Step A. methyl 3- [(dimethylamino)methyl] -5-(hydroxymethyl)benzoate
Figure imgf000250_0002
To a reaction vial was added methyl 3-bromo-5-(hydroxymethyl)benzoate (1.2 g, 4.9 mmol, prepared as described in WO 2003048111 from dimethyl 5-bromoisophthalate, Alfa Aesar), cesium carbonate (4.79 g, 14.7 mmol), dicyclohexyl(2',4',6'- triisopropylbiphenyl-2-yl)phosphine (280 mg, 0.59 mmol, Aldrich), potassium
[(dimethylamino)methyl](trifluoro)borate(l-) (0.970 g, 5.88 mmol, Aldrich), palladium acetate (66 mg, 0.29 mmol) and THF:H20 (10: 1, 30 mL). The reaction mixture was degassed by purging with a stream of nitrogen for 10 minutes. The vial was sealed and heated at 80 °C for 17 hours. The reaction mixture was partitioned between water and ethyl acetate. The organic layer was was washed twice with water. The combined aqueous portions were then saturated with NaCl, and the product was extracted with eight portions of DCM. The extracts were dried over sodium sulfate, filtered and concentrated to afford product as a colorless oil. Yield: 0.37 g (34%); LC-MS: 224.1 (M+H)+. 1H
NMR (300 MHz, CDC13): δ 7.94 (s, 1H), 7.88 (s, 1H), 7.56 (s, 1H), 4.74 (s, 2H), 3.91 (s, 3H), 3.46 (s, 2H), 2.24 (s, 6H).
Step B. methyl 3-[(dimethylamino)methyl]-5-formylbenzoate
Figure imgf000251_0001
Manganese(IV) oxide (0.72 g, 8.3 mmol) was added to methyl 3- [(dimethylamino)methyl]-5-(hydroxymethyl)benzoate (0.37 g, 1.6 mmol, from Step A) in Toluene (15 mL). The mixture was heated to 105 °C for 2 hours, then was cooled to room temperature and filtered. Solvent was removed from the filtrate in vacuo to afford the product as a colorless oil. Yield: 0.30 g (82%); LC-MS: 222.1 (M+H)+. 1H NMR (400 MHz, CDCI3): δ 10.07 (s, 1H), 8.43 (dd, 1H), 8.25 (dd, 1H), 8.05 (dd, 1H), 3.96 (s, 3H), 3.54 (s, 2H), 2.26 (s, 6H).
Step C. methyl 3-(difluoromethyl)-5-[(dimethylamino)methyl]benzoate
Figure imgf000251_0002
Methyl 3-[(dimethylamino)methyl]-5-formylbenzoate (99 mg, 0.45 mmol, from Step B), was stirred in DeoxoFluor® (495 μί, 2.69 mmol) containing ethanol (5 μί, 0.09 mmol) for 24 hours. The mixture was quenched by dropwise addition into ice-cold saturated NaHCC solution. The product was isolated by extraction using DCM. The organic extract was washed twice with water, once with brine, was dried over sodium sulfate, filtered and concentrated to afford product as a light yellow oil which was used without further purification. Yield: 0.046 g (30%); LC-MS: 244.1 (M+H)+. 1H NMR (400 MHz, CDCI3): δ 8.09 (s, 2H), 7.69 (s, 1H), 6.68 (t, 1H), 3.94 (s, 3H), 3.36 (s, 2H), 2.25
(s, 6H).
Step D. 3-(difluoromethyl)-5-[(dimethylamino)methyl]b
Figure imgf000252_0001
Lithium hydroxide monohydrate (65.2 mg, 1.55 mmol) in water (0.7 mL) was added to a solution of methyl 3-(difluoromethyl)-5-[(dimethylamino)methyl]benzoate (45 mg, 0.13 mmol, from Step C) in tetrahydrofuran (2 mL). Upon stirring for 3.5 hours, the mixture was treated with IN HCl to adjust the pH to 7, then THF was removed by rotary evaporation. Acetonitrile was added to make a 1 : 1 ACN:water mixture, the mixture was filtered, and the filtrate was purified via preparative HPLC-MS (CI 8 eluting with a gradient of MeCN/H20 containing 0.15% NH4OH) to afford product as a white solid. Yield: 0.030 g (100%); LC-MS: 230.1 (M+H)+. 1H NMR (300 MHz, DMSO-d6): δ 7.95 (s, 2H), 7.50 (s, 1H), 7.05 (t, 1H), 3.44 (s, 2H), 2.15 (s, 6H).
Step E. {l-(l-{3-(dtfluoromethyl)-5-[ (dimethylamino)methyl]benzoyl}piperidin-4-yl)-3- [4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}a^
A mixture of 3-(difluoromethyl)-5-[(dimethylamino)methyl]benzoic acid (14.0 mg, 0.0609 mmol, from Step D), Triethylamine (28.3 L, 0.203 mmol) and Ν,Ν,Ν',Ν'- tetramethyl-0-(7-azabenzotriazol-l-yl)uronium hexafluorophosphate (21.2 mg, 0.0558 mmol) in tetrahydrofuran (0.56 mL) was stirred for 15 minutes. {l-piperidin-4-yl-3-[4- (7- { [2-(trimethylsilyl)ethoxy]methyl} -7H-pyrrolo[2,3-d]pyrimidin-4-yl)- 1 H-pyrazol- 1 - yl]azetidin-3-yl} acetonitrile (25.0 mg, 0.0507 mmol, prepared as described in Example 1, Step H, except worked up to provide the free base) was added and the reaction was stirred for two hours. The reaction mixture was partitioned between ethyl acetate and water. The organic portion was washed with water, 0.1N NaOH and sat. NaCl, dried over sodium sulfate, filtered and concentrated. The residue was stirred in 1 : 1 DCM:TFA for 1 hour, solvents were removed in vacuo, and the resulting residue stirred in methanol (1 mL) containing ethylenediamine (0.2 mL) until deprotection was complete. Purification via preparative HPLC-MS (C18 eluting with a gradient of MeCN/H20 containing 0.15% NH4OH) afforded product as a white powder. Yield: 0.012 g (40%); LC-MS: 574.3 (M+H)+. 1H NMR (400 MHz, DMSO-d6): δ 12.09 (br s, 1H), 8.82 (s, 1H), 8.69 (s, 1H), 8.42 (s, 1H), 7.61 (d, 1H), 7.56 (s, 1H), 7.46-7.41 (m, 2H), 7.07 (t, 1H), 7.07 (d, 1H), 4.17-4.03 (m, 1H), 3.75 (d, 2H), 3.62-3.25 (m, 7H), 3.22-3.03 (m, 2H), 2.58-2.51 (m, 1H), 2.15 (s, 6H), 1.85-1.55 (m, 2H), 1.33-1.12 (m, 2H).
Example 376. {l-(l-{[6-(pyrrolidin-l-ylmethyl)-2-(trifluoromethyl)pyrimidin-4- yl] carbonyl}piperidin-4-yl)-3- [4-(7H-pyrrolo [2,3-d] pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-3-yl}acetonitrile (trifluoroacetate salt: 4 TFA)
Figure imgf000253_0001
Step A. ethyl 6-(bromomethyl)-2-(tri uoromethyl)pyrimidine-4-carboxylate
Figure imgf000253_0002
A solution of ethyl 6-methyl-2-(trifluoromethyl)pyrimidine-4-carboxylate (2.00 g, 8.54 mmol, prepared as described in WO2007/090748) in acetic acid (12 mL) was treated with bromine (1.36 g, 8.54 mmol) and the reaction was heated to 80 °C in a sealed vial for 30 minutes, at which time it was decolorized. The mixture containing unreacted starting material, desired product, and over brominated product, was rotovapped and azeotroped once with toluene. The percent by weight of desired component was determined by NMR and the mixture used without further purification. Yield: 1.62 g (61%); LC-MS: 313.0, 315.0 (M+H)+. 1H NMR (300 MHz, CDC13): δ 8.32 (s, 1H), 4.60 (s, 2H), 4.54 (q, 2H), 1.46 (t, 3H). Step B. ethyl 6-[(acetyloxy)methyl -2-(trifluoromethyl)pyrimidine-4-carboxylate
Figure imgf000254_0001
Ethyl 6-(bromomethyl)-2-(trifluoromethyl)pyrimidine-4-carboxylate (1.62 g, 5.17 mmol, from Step A) was dissolved in acetonitrile (15 mL) and sodium acetate (2.8 g, 34 mmol) was added. The mixture was heated to 80 °C for 4 hours, then stood at room temperature overnight. Acetonitrile was removed in vacuo. The residue was partitioned between water and ethyl acetate and the aqueous layer was extracted with two further portions of ethyl acetate. The combined extracts were washed with water, then brine, dried over sodium sulfate, filtered and concentrated. Flash chromatography, eluting with a gradient from 0-60% ethyl acetate/hexane afforded purified product. Yield: 0.95 g (63%); LC-MS: 293.0 (M+H)+. 1H NMR (300 MHz, CDC13): δ 8.15 (s, 1H), 5.36 (s, 2H), 4.53 (q, 2H), 2.25 (s, 3H), 1.46 (t, 3H). Step C. 6-[(acetyloxy)methyl]-2-(trifluoromethyl)pyrimidine-4-carboxylic acid
Figure imgf000254_0002
A solution of ethyl 6-[(acetyloxy)methyl]-2-(trifluoromethyl)pyrimidine-4- carboxylate (0.95 g, 3.2 mmol, from Step B) in tetrahydrofuran (8.7 mL) at 0 °C was treated with Lithium hydroxide, monohydrate (140 mg, 3.2 mmol) in water (1.3 mL). The reaction was stirred for 15 minutes, then was treated with IN HC1 to pH~4 while still in the ice bath. THF was removed from the mixture in vacuo. The product was extracted first with ethyl acetate, then with several portions of 10% isopropanol in CHC13, including periodic adjustment of pH as necessary. The extracts were combined and dried over sodium sulfate, filtered and concentrated to afford a yellow oil, which was used without further purification. Yield: 0.86 g (100%); LC-MS: 265.0 (M+H)+. 1H NMR (300 MHz, CDC13): δ 8.25 (s, 1H), 5.35 (s, 2H), 2.23 (s, 3H).
Step D. [ 6-[(4-{3-(cyanomethyl)-3-[ 4-(7-{ [2-(trimethylsilyl)ethoxy] methyl}-7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH^yrazol-l-yl]azetidin-l-yl}piperidin- ( trifluoromethyl)pyrimidin-4-yl] meth l acetate
Figure imgf000255_0001
{l-Piperidin-4-yl-3-[4-(7-{[2-(trimethylsilyl)etlioxy]rnetliyl}-7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile (0.89 g, 1.8 mmol, prepared as described in Example 1, Step H, except worked up to provide the free base) and 6- [(acetyloxy)methyl]-2-(trifluoromethyl)pyrimidine-4-carboxylic acid (0.572 g, 2.16 mmol, from Step C) were dissolved in N,N-dimethylformamide (18 mL). Triethylamine (1.2 mL, 9.0 mmol) and benzotriazol-l-yloxytris(dimethylamino)phosphonium
hexafluorophosphate (958 mg, 2.16 mmol) were added. The reaction was stirred overnight. The reaction mixture was partitioned between ethyl acetate and water. The aqueous was extracted with three portions of ethyl acetate. The combined extracts were washed twice with water and once with brine, dried over sodium sulfate, filtered and concentrated. Flash chromatography, eluting with a gradient from 0-5% methanol in ethyl acetate, afforded desired product. Yield: 0.85 g (64%); LC-MS: 739.2 (M+H)+. 1H NMR (300 MHz, CDC13): δ 8.85 (s, 1H), 8.40 (s, 1H), 8.31 (s, 1H), 7.82 (s, 1H), 7.41 (d, 1H), 6.80 (d, 1H), 5.68 (s, 2H), 5.32 (s, 2H), 4.26-4.16 (m, 1H), 3.87-3.72 (m, 3H), 3.64 (dd, 2H), 3.55 (dd, 2H), 3.48-3.35 (m, 3H), 3.29 (ddd, 1H), 2.64-2.54 (m, 1H), 2.04 (s, 3H), 1.92-1.73 (m, 2H), 1.61-1.43 (m, 2H), 0.92 (dd, 2H), -0.06 (s, 9H).
Step E. {l-(l-{[ 6-(hydroxymethyl)-2-(trifluoromethyl)pyrimidin-4-ylJcarbonyl}piperidin- 4-yl)-3-[ 4-(7-{[ 2-(trimethylsilyl)ethoxy) methyl} -7 H-pyrrolo [ 2, 3-d]pyrimidin-4-yl)-lH- pyrazol-l-yl]azetidin-3-yl}acetonitrile
Figure imgf000256_0001
To a solution of [6-[(4-{3-(cyanomethyl)-3-[4-(7-{[2- trimethylsilyl)ethoxy]methyl} -7H-pyrrolo[2,3-d]pyrimidin-4-yl)- 1 H-pyrazol- 1 - yl]azetidin-l-yl)carbonyl]-2-(trifluoromethyl)pyrimidin-4-yl]metliyl acetate (0.85 g, 1.15 mmol, from Step D) in THF (16 mL) at room temperature was added a solution of lithium hydroxide, monohydrate (0.072 g, 1.7 mmol) in water (4 mL). The reaction was stirred for 45 minutes, then was neutralized by the addition of IN HCl and the product was extracted with ethyl acetate. The extracts were combined and dried over sodium sulfate, decanted and concentrated to afford a yellow foam, which was used without further purification. Yield: 0.72 g (90%); LC-MS: 697.2 (M+H)+. 1H NMR (300 MHz, CDCI3): δ 8.86 (s, 1H), 8.44 (s, 1H), 8.31 (s, 1H), 7.92 (s, 1H), 7.42 (d, 1H), 6.81 (d, 1H), 5.68 (s, 2H), 4.92 (s, 2H), 4.28-4.17 (m, 1H), 3.89-3.71 (m, 3H), 3.71-3.61 (m, 2H), 3.55 (dd, 2H), 3.47-3.34 (m, 3H), 3.33-3.21 (m, 1H), 2.68-2.53 (m, 1H), 1.93-1.68 (m, 2H), 1.60-1.41 (m, 2H), 0.92 (dd, 2H), -0.06 (s, 9H).
Step F. {1 -(!-{[ 6-(pyrrolidin-l-ylmethyl)-2-(trifluoromethyl)pyrimidin-4- yl]carbonyl}piperidin-4-yl)-3-[4-(7H^yrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin-3-yl}acetonitrile (trifluoroacetate salt: 4 TFA)
To a solution of {l-(l-{[6-(hydroxymethyl)-2-(trifluorometliyl)pyrimidin-4- yl]carbonyl}piperidin-4-yl)-3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile (0.030 g, 0.043 mmol, from Step E) and triethylamine (0.015 mL, 0.11 mmol) in methylene chloride (1 mL) at 0 °C was added methanesulfonyl chloride (0.006 mL, 0.08 mmol). The reaction was allowed to warm to room temperature immediately after addition of mesyl chloride (MsCl). When mesylate formation was confirmed to be complete, the solution of mesylate was added to a mixture of pyrrolidine (0.017 mL, 0.20 mmol, Aldrich) and a few drops of
triethylamine in DCM (0.2 mL). The displacement reaction was heated to 40 °C for 30 minutes. The reaction was cooled to room temperature and trifluoroacetic acid (1 mL) was added. After stirring for 1 hour, the solvents were removed in vacuo and replaced with methanol (1 mL) and ethylenediamine (0.2 mL). After stirring for 30 minutes, the product was purified by preparative HPLC-MS (Waters SunFire CI 8, 5 um particle size, 30 x 100 mm, eluting with a gradient of 5-23% MeCN in H20 containing 0.1% TFA over 12 minutes). Yield: 0.012 g (25%); LC-MS: 620.2 (M+H)+. 1H NMR (300 MHz, CD3OD): δ 9.10 (s, 1H), 8.90 (s, 1H), 8.59 (s, 1H), 8.01 (s, 1H), 7.83 (d, 1H), 7.29 (d, 1H), 4.95 (d, 2H), 4.85 (s, 2H), 4.77-4.66 (m, 3H), 4.02 (d, 1H), 3.95-3.15 (m, 8H), 3.12-2.99 (m, 1H), 2.31-2.03 (m, 6H), 1.77-1.51 (m, 2H). 19F NMR (300 MHz, CD3OD): δ -72.21 (s, 3F), -77.61 (s, 12F).
Example 377. {l-(l-{[6-{[(3S)-3-fluoropyrrolidin-l-yl]methyl}-2- (trifluoromethyl)pyrimidin-4-yl] carbonyl}piperidin-4-yl)-3- [4-(7H-pyrrolo [2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile (trifluoroacetate salt: 4 TFA)
Figure imgf000258_0001
Prepared as in the manner of Example 376, using (3S)-3-fluoropyrrolidine hydrochloride (0.050 g, 0.40 mmol, Aldrich) and excess triethylamine in Step F, and was heated for 24 hours at 40 °C. Yield: 0.012 g (26%); LC-MS: 638.3 (M+H)+. 1H NMR (300 MHz, CD3OD): δ 9.05 (s, IH), 8.88 (s, IH), 8.57 (s, IH), 8.01 (s, IH), 7.79 (d, IH), 7.25 (d, IH), 5.50 (d, IH), 4.93 (s, 2H), 4.85 (d, 2H), 4.74-4.57 (m, 3H), 4.06-3.65 (m, 7H), 3.60-3.45 (m, IH), 3.29-3.17 (m, IH), 3.15-3.02 (m, IH), 2.58-2.36 (m, 2H), 2.22 (d, IH), 2.10 (d, IH), 1.75-1.49 (m, 2H). 19F NMR (300 MHz, CD3OD): δ -72.24 (s, 3F), -77.59 (s, 12F), -175.45 (br, IF).
Example 378. {l-(l-{[6-{[(3R)-3-fluoropyrrolidin-l-yl]methyl}-2- (trifluoromethyl)pyrimidin-4-yl] carbonyl}piperidin-4-yl)-3- [4-(7H-pyrrolo [2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile (trifluoroacetate salt: 4 TFA)
Figure imgf000259_0001
Prepared as in the manner of Example 376, using (3R)-3-fluoropyrrolidine hydrochloride (0.025 g, 0.20 mmol, Oakwood). Yield: 0.012 g (26%); LC-MS: 638.3 (M+H)+. 1H NMR (300 MHz, CD3OD): δ 9.04 (s, 1H), 8.87 (s, 1H), 8.57 (s, 1H), 8.01 (s, 1H), 7.78 (d, 1H), 7.25 (d, 1H), 5.50 (d, 1H), 4.93 (s, 2H), 4.84 (d, 2H), 4.74-4.54 (m, 3H), 4.07-3.62 (m, 7H), 3.59-3.44 (m, 1H), 3.29-3.20 (m, 1H), 3.15-3.03 (m, 1H), 2.58- 2.37 (m, 2H), 2.21 (d, 1H), 2.09 (d, 1H), 1.75-1.49 (m, 2H). 19F NMR (300 MHz, CD3OD): δ -72.24 (s, 3F), -77.55 (s, 12F), -175.47 (br, IF). Example 379. {l-(l-{[6-[(3,3-difluoropyrrolidin-l-yl)methyl]-2-
(trifluoromethyl)pyrimidin-4-yl] carbonyl}piperidin-4-yl)-3- [4-(7H-pyrrolo [2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile (trifluoroacetate salt: 4 TFA)
Figure imgf000260_0001
Prepared as in the manner of Example 376, using 3,3-difluoropyrrolidine hydrochloride (0.050 g, 0.40 mmol, Matrix) and excess triethylamine in Step F, and was heated for 24 hours at 40 °C. Yield: 0.012 g (25%); LC-MS: 656.2 (M+H)+. 1H NMR (300 MHz, CD3OD): δ 9.07 (s, 1H), 8.89 (s, 1H), 8.58 (s, 1H), 7.99 (s, 1H), 7.81 (d, 1H), 7.26 (d, 1H), 4.95-4.87 (m, 2H), 4.76-4.62 (m, 3H), 4.24 (s, 2H), 4.04-3.92 (m, 1H), 3.71 (s, 2H), 3.65-3.50 (m, 1H), 3.40-3.14 (m, 5H), 3.12-2.99 (m, 1H), 2.44 (tt, 2H), 2.10 (d, 1H), 1.62 (dddd, 2H). 19F NMR (300 MHz, CD3OD): δ -72.33 (s, 3F), -77.64 (s, 12F), - 95.48 (tt, 2F).
Example 380. {l-(l-{[6-[(tert-butylamino)methyl]-2-(trifluoromethyl)pyrimidin-4- yl] carbonyl}piperidin-4-yl)-3- [4-(7H-pyrrolo [2,3-d] pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-3-yl}acetonitrile (trifluoroacetate salt: 3 TFA)
Figure imgf000261_0001
Prepared as in the manner of Example 376, using tert-butylamine (0.050 mL, 0.48 mmol, Aldrich) and excess triethylamine in Step F, and was heated for 24 hours at 40 °C. Yield: 0.012 g (28%); LC-MS: 622.3 (M+H)+. 1H NMR (300 MHz, CD3OD): δ 9.01 (s, IH), 8.85 (s, IH), 8.56 (s, IH), 8.03 (s, IH), 7.75 (d, IH), 7.22 (d, IH), 4.77 (d, 2H), 4.71-4.61 (m, 3H), 4.56 (d, 2H), 3.95 (br d, IH), 3.69 (s, 2H), 3.56-3.39 (m, IH), 3.30- 3.20 (m, IH), 3.17-3.04 (m, IH), 2.19 (br d, IH), 2.07 (br d, IH), 1.73-1.52 (m, 2H), 1.49 (s, 9H). 19F NMR (300 MHz, CD3OD): δ -72.02 (s, 3F), -77.47 (s, 9F).
Example 381. {l-(l-{[6-(hydroxymethyl)-2-(trifluoromethyl)pyrimidin-4- yl] carbonyl}piperidin-4-yl)-3- [4-(7H-pyrrolo [2,3-d] pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin-3-yl}acetonitrile
Lithium hydroxide monohydrate (3.6 mg, 0.085 mmol) in water (0.10 mL) was added to a solution of [6-[(4-{3-(cyanomethyl)-3-[4-(7-{[2- (trimethylsilyl)ethoxy]methyl} -7H-pyrrolo[2,3-d]pyrimidin-4-yl)- 1 H-pyrazol- 1 - yl] azetidin- 1 -yl} piperidin- 1 -yl)carbonyl] -2-(trifluoromethyl)pyrimidin-4-yl]methyl acetate (21 mg, 0.028 mmol, from Example 376, Step D) in tetrahydrofuran (0.40 mL). The mixture was stirred for 5 minutes and was then treated with IN HC1 to neutralize. Solvent was then removed in vacuo. The residue was stirred in a solution of 1 : 1
TFA/DCM for one hour, then solvents were removed in vacuo again. The residue was redissolved in MeOH (1 mL) and ethylenediamine (0.2 mL) was added. When
deprotection was complete, the product was purified via preparative HPLC-MS (CI 8 eluting with a gradient of MeCN/H20 containing 0.15% NH4OH). Yield: 0.004 g (25%); LC-MS: 567.3 (M+H)+. 1H NMR (400 MHz, DMSO-d6): δ 12.14 (br s, 1H), 8.82 (s, 1H), 8.69 (s, 1H), 8.42 (s, 1H), 7.90 (s, 1H), 7.61 (d, 1H), 7.06 (d, 1H), 5.94 (br s, 1H), 4.71 (s, 2H), 4.12-4.02 (m, 1H), 3.75 (dd, 2H), 3.63-3.46 (m, 5H), 3.31-3.22 (m, 1H), 3.17-3.07 (m, lh), 2.61-2.53 (m, 1H), 1.84-1.75 (m, 1H), 1.71-1.61 (m, 1H), 1.37-1.18 (m, 2H). 19F NMR (400 MHz, DMSO-d6): δ -69.46 (s, 3F).
Example 382. {l-(l-{[6-[(isopropylamino)methyl]-2-(trifluoromethyl)pyrimidin-4- yl] carbonyl}piperidin-4-yl)-3- [4-(7H-pyrrolo [2,3-d] pyrimidin-4-yl)-l H-pyrazol- 1- yl] azetidin-3-yl}acetonitrile
Figure imgf000263_0001
Methanesulfonyl chloride (2.8 μί, 0.036 mmol) in methylene chloride (0.20 mL) was added to a mixture of {l-(l-{[6-(hydroxymethyl)-2-(trifluoromethyl)pyrimidin- 4-yl]carbonyl}piperidin-4-yl)-3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile (21 mg, 0.030 mmol, prepared as in Example 376, Step E) and triethylamine (8.4 μΕ, 0.060 mmol) in methylene chloride (1.0 mL) at 0 °C. After 15 minutes, 2-propanamine (20 μί, 0.3 mmol) was added. The mixture was then heated to 40 °C. After 1.5 hours, additional 2- Propanamine (20 μΐ^, 0.3 mmol, Aldrich) was added and the reaction heated for a total of 3 hours at this temperature. The mixture was concentrated in vacuo. The residue was stirred for 1 hour in a 1 : 1 mixture of TFA/DCM, then concentrated again. The residue was redissolved in MeOH (1.0 mL) and ethylenediamine (0.2 mL) was added. The product was purified via preparative HPLC-MS (CI 8 eluting with a gradient of
MeCN/H20 containing 0.15% NH4OH). Yield: 11.6 mg (63%); LC-MS: 608.4 (M+H)+. 1H NMR (400 MHz, DMSO-d6): δ 12.15 (br s, 1H), 8.82 (s, 1H), 8.69 (s, 1H), 8.42 (s, 1H), 7.97 (s, 1H), 7.61 (d, 1H), 7.07 (d, 1H), 41.2-4.02 (m, 1H), 3.94 (s, 2H), 3.75 (dd, 2H), 3.63-3.45 (m, 5H), 3.31-3.22 (m, 1H), 3.16-3.06 (m, 1H), 2.75 (septet, 1H), 2.61- 2.53 (m, 1H), 1.84-1.75 (m, 1H), 1.70-1.60 (m, 1H), 1.36-1.19 (m, 2H), 1.01 (d, 6H). 19F NMR (400 MHz, DMSO-d6): δ -69.46 (s, 3F).
Example 383. {l-(l-{[6-[(ethylamino)methyl]-2-(trifluoromethyl)pyrimidin-4- yl] carbonyl}piperidin-4-yl)-3- [4-(7H-pyrrolo [2,3-d] pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin-3-yl}acetonitrile
Figure imgf000264_0001
Prepared by the method of Example 382, using Ethylamine (0.10 mL, 1.8 mmol, Aldrich) and carrying out the substitution reaction at room temperature for 1 hour.
Yield: 8.4 mg (47%); LC-MS: 594.2 (M+H)+. 1H NMR (400 MHz, DMSO-d6): δ 12.13 (br s, 1H), 8.82 (s, 1H), 8.69 (s, 1H), 8.42 (s, 1H), 7.95 (s, 1H), 7.61 (d, 1H), 7.06 (d, 1H), 4.13-4.02 (m, 1H), 3.93 (s, 2H), 3.75 (dd, 2H), 3.62-3.46 (m, 5H), 3.31-3.22 (m, 1H), 3.15-3.06 (m, 1H), 2.61-2.53 (m, 3H), 1.84-1.74 (m, 1H), 1.71-1.60 (m, 1H), 1.37-1.18 (m, 2H), 1.04 (t, 3H). 19F NMR (400 MHz, DMSO-d6): δ -69.45 (s, 3F).
Example 384. {l-(l-{[6-{[(2-methoxyethyl)(methyl)amino]methyl}-2- (trifluoromethyl)pyrimidin-4-yl] carbonyl}piperidin-4-yl)-3- [4-(7H-pyrrolo [2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile
Figure imgf000265_0001
Prepared in the manner of Example 382, using 2-methoxy-N-methylethaneamine (0.077 g, 0.86 mmol, Oakwood) and the substitution carried out in a sealed vial at 60 °C for 7 hours. Yield: 0.007 g (26%); LC-MS: 638.3 (M+H)+. 1H NMR (400 MHz, CD3OD): δ 8.69 (s, 1H), 8.67 (s, 1H), 8.40 (s, 1H), 8.01 (s, 1H), 7.51 (d, 1H), 6.98 (d, 1H), 4.32- 4.23 (m, lh), 3.88 (s, 2H), 3.85-3.73 (m, 4H), 3.72-3.64 (m, 1H), 3.54 (t, 2H), 3.50 (s, 2H), 3.38-3.31 (m, 1H), 3.31 (s, 3H), 3.27-3.17 (m, 1H), 2.71 (t, 2H), 2.69-2.61 (m, 1H), 2.37 (s, 3H), 1.97-1.87 (m, 1H), 1.86-1.76 (m, 1H), 1.52-1.38 (m, 2H). 19F NMR (300 MHz, CD3OD): δ -72.34 (s, 3F).
Example 385. {l-(l-{[6-{[(3-hydroxypropyl)(methyl)amino]methyl}-2- (trifluoromethyl)pyrimidin-4-yl] carbonyl}piperidin-4-yl)-3- [4-(7H-pyrrolo [2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile
Figure imgf000266_0001
Prepared in the manner of Example 382, using 3-(methylamino)propanol (0.038 g, 0.43 mmol, TCI America) and the substitution carried out at 40 °C for 1 hour.
Yield: 0.007 g (26%); LC-MS: 638.3 (M+H)+. 1H NMR (400 MHz, CD3OD): δ 8.69 (s, 1H), 8.67 (s, 1H), 8.40 (s, 1H), 7.94 (s, 1H), 7.51 (d, 1H), 6.99 (d, 1H), 4.33-4.23 (m,
1H), 3.86-3.74 (m, 6H), 3.74-3.66 (m, 1H), 3.63 (t, 2H), 3.50 (s, 2H), 3.38-3.18 (m, 2H), 2.70-2.62 (m, 1H), 2.59 (t, 2H), 2.32 (s, 3H), 1.97-1.88 (m, 1H), 1.86-1.79 (m, 1H), 1.75 (tt, 2H), 1.52-1.39 (m, 2H). 19 F NMR (300 MHz, CD3OD): δ -71.88 (s, 3F). Example 386. Propyl 4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-
4-yl)-lH-pyrazol-l-yl]azetidin-l-yl}piperidine-l-carboxylate
Figure imgf000266_0002
A solution of { l-piperidin-4-yl-3-[4-(7- {[2-(trimethylsilyl)ethoxy]methyl}-7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile (0.010 g, 0.020 mmol) and propyl chloroformate (3.0 μί, 0.026 mmol) in methylene chloride (1 mL, 20 mmol) was stirred for 1.5 h. TFA, 1 mL, was added. After 1 hour, the solvent was removed by rotary evaporation to give an oil. The oil was dissolved in lmL MeOH and 50 microL ethylenediamine was added. After 1 hour, the reaction was purified by prep- HPLC (pHIO) using a Waters XBridge CI 8, 5 μιη particle size, 19x100mm; mobile phase system: aqueous (0.1 % NH4OH)/acetonitrile; flow rate: 30 mL/min; separation gradient: 40-60% B in 5 min to give 5.3 mg white solid (58%). 1H NMR (400 MHz,
DMSO): δ 12.13 (1H, br); 8.83 (1H, s); 8.7 (1H, s); 8.42 (1H, s); 7.61 (1H, m); 7.05 (1H, m); 3.95 (2H, t); 3.75 (4H, m); 3.55 (4H, m); 3.0 (2H, br); 2.43 (1H, m); 1.65 (2H, m); 1.58 (2H, m); 1.15 (2H, m); 0.95 (3H, t). LCMS (M+l): 449.
Example 387. Cyclobutylmethyl 4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3- d] py r imidin-4-yl)- 1 H-py r azol- 1 -yl] azetidin- 1 -yl} piperidine- 1 -carboxylate
Figure imgf000267_0001
To a solution of cyclobutylmethanol (1 1 μΙ^, 0Α2 mmol) in methylene chloride (1 mL, 20 mmol) was added 2.02 M phosgene in toluene (0.045 mL, 0.091 mmol). After stirred for 2 hours, { l-piperidin-4-yl-3-[4-(7- {[2-(trimethylsilyl)ethoxy]methyl}-7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile (0.020 g, 0.040 mmol) and N,N-diisopropylethylamine (0.040 mL, 0.23 mmol) were added and was stirred overnight. The solvent was removed by rotary evaporation to give an oil. The oil was then dissolved in 1 mL methanol and 100 microL ethylenediamine was added. After 1 hour, the reaction was purified by prep-HPLC (pHIO) using a Waters XBridge CI 8, 5 μιη particle size, 19x100mm; mobile phase system: aqueous (0.1 % NH4OH)/acetonitrile; flow rate: 30 mL/min; separation gradient: 40-60% B in 5 min to give 15 mg white solid (78%). 1H NMR (400 MHz, DMSO): δ 12.08 (1H, br); 8.75 (1H, s); 8.62 (1H, s); 8.35 (1H, s); 7.55 (1H, m); 7.0 (1H, m); 3.9 (2H, d); 3.65 (4H, m); 3.5 (4H, m); 2.9 (2H, br); 2.38 (1H, m); 1.91 (2H, m); 1.77 (2H, m); 1.61 (5H, m); 1.03 (2H, m). LCMS (M+l): 475.
The following compounds were prepared by methods analogous to those
Examples 386-387.
Figure imgf000268_0001
Figure imgf000269_0001
Figure imgf000270_0001
Figure imgf000271_0001
Figure imgf000271_0002
Example 398. Crystalline salts of {l-{l-[3-Fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile
A. Glutarate salt: A flask was charged with { 1 - { 1 -[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile free base (37.85 mg, 0.068 mmol, 1 eq) and 2- propanol (0.6 mL). The reaction mixture was stirred for 15 min to give a clear solution followed by adding of glutaric acid (12.1 mg, 0.092 mmol, 1.34 eq, Aldrich, Cat G3407). The reaction mixture was stirred for about 8 min to give a thick slurry, and continuously stirred for 5 h. The solid was collected by filtration, washed with heptane and dried to provide glutarate salt as an off- white crystal (39.9 mg, 85%, 1796-108).
The stoichiometric ratio of free base to glutaric acid was determined by 1H NMR as 1 : 1. The crystallinity of the glutarate salt was confirmed by XRPD. The DSC thermogram exhibited a melting endotherms, with an initial Tonset at 206.26 °C and Tpeak at 207.63 °C. The TGA showed a weight loss of 0.037% up to approximately 100 °C. SEM image indicated that the glutarate salt has a rod-like crystal shape.
B. Citrate salt: A reactor was charged with { 1 - { 1 - [3 -fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile free base (30.97 mg, 0.056 mmol, 1 eq) and ethanol (0.5 mL). To the clear solution was added citric acid (11.92 mg, 0.062 mmol, 1.1 equiv.). After the reaction mixture was stirred for 60 min to give a slurry, the slurry was heated at about 75 °C for 80 min and stirred at room temperature for 4 h. The precipitate was collected by filtration, washed with heptane and dried under vacuum overnight to provide the citrate salt (38.6 mg, 91.9%) as off-white solid.
The stoichiometric ratio of the salt between free base and citric acid was determined by 1H NMR as 1 : 1. The crystallinity of the salt was confirmed by XRPD and further supported by DSC. The TGA showed about 0.57%> weight loss up to about 100 °C. SEM image indicated that the salt has a plane-like crystal shape.
C. Benzoate salt: To the solution of { 1 - { 1 -[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile free base (31.41 mg, 0.0.057 mmol, 1 eq) in 2- propanol (0.5 mL) was added benzoic acid (16.54 mg, 0.135 mmol, 2.39 eq). The colorless solution turns to a slurry after stirring for 20 min. The mixture was stirred at room temperature overnight. The solid was collected by filtration, washed by heptane (1.5 mL) and dried overnight under vacuum to afford the benzoate salt (35 mg, 91.3%) as off-white solid.
The stoichiometric ratio of free base to benzoic acid was determined by 1H NMR as 1 : 1. The crystallinity of the benzoate salt was confirmed by XRPD. The DSC thermogram exhibited melting endotherms. The TGA showed a weight loss of 0.080% up to approximately 100 °C. The SEM image showed that the benzoate salt was a plane-like crystal.
Using similar procedures to those described above, the maleate, salicylate, saccharin, camsylate, and nicotinate salts of {l-{l-[3-fluoro-2-
(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile were also found to be good crystalline salts.
Example 399. Pharmaceutical Compositions of {l-{l-[3-Fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile Adipic Acid Salt
Prototype capsules were manufactured using a conventional dry blending process. The initial prototype capsules was performed on a 200 mg weight blend, for both the 10 mg and 50 mg capsules. Silicified microcrystalline cellulose formulation was selected based on manufacturability, dissolution and content uniformity data obtained on development batches . The 1 - { 1 - [3 -fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4- yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile adipic acid salt ("adipic acid salt") could be formed as shown in Example 358. The composition of the silicified microcrystalline cellulose prototype capsule formulation is listed in Tables A and B below. Table A. Com onents and Com osition of 10 m Ca sule
Figure imgf000274_0001
*Salt conversion factor is 0.7911
**Comprised of 98% Microcrystalline Cellulose NF and 2 % Colloidal Silicon Dioxide NF
Batch formula for 10 mg and 50 mg capsules are shown in Tables C and D. The capsules are made by the steps below:
1. The required amount of adipic acid salt and an approximately equal amount of silicified microcrystalline cellulose (SMCC) are pre -mixed.
2. The mixture from step 1 is passed through a suitable screen (e.g., 40 mesh).
3. The remaining SMCC is screened through the same screen used in step 2.
4. The screened SMCC from step 3 is blended along with the mixture from step 2 in a suitable blender (e.g., a Turbula blender) for approximately 5 minutes.
5. The blend is filled into capsules to the desired fill weight.
Table C. Batch Formula for 225 g Blend for 10 mg Capsules
Figure imgf000274_0002
Table D. Batch Formula for 936 g Blend for 50 mg Capsules
Figure imgf000275_0001
Example A: In vitro JAK Kinase Assay
Compounds herein were tested for inhibitory activity of JAK targets according to the following in vitro assay described in Park et ah, Analytical Biochemistry 1999, 269, 94-104. The catalytic domains of human JAKl (a.a. 837-1142), Jak2 (a.a. 828-1132) and Jak3 (a.a. 781-1124) with an N-terminal His tag were expressed using baculovirus in insect cells and purified. The catalytic activity of JAKl, JAK2 or JAK3 was assayed by measuring the phosphorylation of a biotinylated peptide. The phosphorylated peptide was detected by homogenous time resolved fluorescence (HTRF). IC50S of compounds were measured for each kinase in the 40 microL reactions that contain the enzyme, ATP and 500 nM peptide in 50 mM Tris (pH 7.8) buffer with 100 mM NaCl, 5 mM DTT, and 0.1 mg/mL (0.01%) BSA. For the 1 mM IC50 measurements, ATP concentration in the reactions was 1 mM. Reactions were carried out at room temperature for 1 hr and then stopped with 20 45 mM EDTA, 300 nM SA-APC, 6 nM Eu-Py20 in assay buffer (Perkin Elmer, Boston, MA). Binding to the Europium labeled antibody took place for 40 minutes and HTRF signal was measured on a Fusion plate reader (Perkin Elmer, Boston, MA). See Table 1 for data related to compounds of the invention.
Table 1. IC50 data for JAK enzyme assay (measured at 1 mM ATP)
Example JAKl JAK2 IC50 Ratio
ICso iM)1 IC50 (nM)2 JAK2/JAK1
1 + ++++ 24.5
2 + +++ 12.3
3 + ++ 11.0
4 ++ +++ 10.2
5 + ++ 12.0
6 + ++
14.3
7 + + 10.8
8 ++ +++ 11.0 Example JAK1 JAK2 IC5o Ratio IQo iM)1 IC50 (nM)2 JAK2/JAK1
9 + ++ 3.2
10 ++ ++ 4.4
11 ++ ++ 7.2
12 +++ ++++ 5.2
13 + ++ 5.3
14 ++ ++ 3.2
15 ++ ++ 5.3
16 ++ ++ 2.5
17 +++ +++ 2.6
18 ++ +++ 8.8
19 + + 3.8
20 + + 4.2
21 ++ ++ 3.2
22 ++ ++ 3.2
23 + ++ 6.7
24 + + 6.6
25 + ++ 5.8
26 + + 3.3
27 ++ +++ 5.1
28 + + 8.5
29 + + 7.2
30 + + 2.8
31 + + 1.9
32 + ++ 4.4
33 + + 0.5
34 ++ ++ 6.0
35 + ++ 6.9
36 + + 2.3
37 + + 2.1
38 + + 5.7
39 + + 3.2
40 + + 4.8
41 + + 4.9
42 + + 6.7
43 + ++ 5.0
44 + + 2.9
45 + ++ 3.1
46 + ++ 6.3
47 + + 3.7
48 + ++ 6.5
49 + + 6.5
50 + + 6.9
51 + + 6.3
52 ++ +++ 4.4
53 + ++ 9.4
54 ++ +++ 9.1
55 + ++ 7.5 Example JAK1 JAK2 IC5o Ratio IQo iM)1 IC50 (nM)2 JAK2/JAK1
56 + + 8.8
57 ++++ ++++ 3.3
58 + ++ 5.0
59 + + 4.6
60 + + 1.8
61 ++ ++ 4.2
62 ++ +++ 6.6
63 + ++ 6.1
64 + + 2.7
65 + ++ 4.3
66 ++ +++ 6.2
67 + + 4.7
68 ++ ++ 5.3
69 ++ ++ 5.3
70 + + 3.8
71 +++ +++ 4.4
72 + + 6.6
73 + + 4.2
74 + ++ 3.8
75 + + 3.4
76 + + 5.5
77 + + 5.5
78 + ++ 5.4
79 + + 3.9
80 + + 4.1
81 + + 4.2
82 + + 5.3
83 + ++ 5.8
84 + ++ 6.5
85 ++ ++++ 15.9
86 + +++ 19.4
87 + + 0.3
88 ++ + 0.9
89 + + 2.6
90 + + 1.5
91 + + 1.7
92 + + 2.8
93 + + 1.5
94 ++ ++ 1.0
95 + + 2.7
96 + + 0.5
97 ++ ++ 2.2
98 ++ +++ 3.7
99 ++ ++ 5.6
100 ++ +++ 6.6
101 ++ ++++ 8.3
102 ++ ++ 4.2 Example JAK1 JAK2 IC5o Ratio IQo iM)1 IC50 (nM)2 JAK2/JAK1
103 ++ ++ 2.5
104 ++ ++ 6.3
105 ++ ++++ 7.2
106 ++ ++ 4.2
107 ++ +++ 4.1
108 ++ ++ 1.4
109 +++ ++ 1.1
110 ++ ++ 1.6
111 ++ ++ 2.6
112 ++ +++ 8.2
113 +++ +++ 4.2
114 ++ ++ 1.5
115 + ++ 15.7
116 + ++ 10.6
117 + ++ 10.0
118 + ++ 21.5
119 + ++ 14.0
120 + + 2.1
121 + + 3.8
122 ++ ++ 2.5
123 + + 3.8
124 + + 8.4
125 + ++ 8.1
126 + ++ 6.3
127 ++ ++ 4.3
128 + ++ 3.5
129 + + 7.9
130 + + 7.1
131 + ++ 8.1
132 + + 5.8
133 + ++ 7.9
134 + ++ 6.7
135 + ++ 7.1
136 ++ ++++ 5.6
137 +++ ++++ 6.0
138 + + 13.9
139 + + 4.4
140 ++ ++ 5.4
141 + + 6.2
142 + + 7.4
143 ++ ++ 3.5
144 ++ ++ 3.9
145 ++ ++ 3.6
146 ++ ++ 5.8
147 + + 6.1
148 + + 5.7
149 + ++ 5.0 Example JAK1 JAK2 IC5o Ratio IQo iM)1 IC50 (nM)2 JAK2/JAK1
150 + + 7.0
151 ++ +++ 5.8
152 ++ ++ 4.0
153 +++ ++ 1.2
154 + ++ 22.0
155 ++ +++ 12.2
156 + ++ 11.2
157 + +++ 14.8
158 + ++ 12.0
159 + ++ 15.0
160 + ++ 12.1
161 + ++ 12.1
162 + ++ 13.6
163 + ++ 12.0
164 + ++ 13.3
165 ++ +++ 10.4
166 + + 5.5
167 + + 2.0
168 + + 2.6
169 + ++ 5.5
170 + ++ 6.9
171 + ++ 7.5
172 ++ ++ 4.2
173 ++ ++ 5.7
174 + + 7.1
175 + + 3.6
176 + + 5.1
177 + + 8.5
178 + + 6.5
179 ++ +++ 3.8
180 + ++ 4.0
181 + ++ 4.9
182 ++ ++ 3.7
183 + + 3.8
184 + + 3.9
185 + ++ 9.5
186 + ++ 5.5
187 + ++ 8.8
188 ++ ++ 2.3
189 ++ ++ 3.5
190 +++ +++ 2.1
191 +++ ++ 1.4
192 ++ ++ 3.0
193 + + 0.5
194 + + 2.2
195 +++ +++ 3.2
196 ++ +++ 9.9 Example JAK1 JAK2 IC5o Ratio IQo iM)1 IC50 (nM)2 JAK2/JAK1
197 ++ ++ 3.2
198 + ++ 4.4
199 + ++ 10.0
200 + ++ 6.7
201 ++ ++ 3.6
202 + ++ 6.4
203 ++ +++ 8.6
204 + ++ 4.0
205 ++ ++ 4.4
206 + + 1.6
207 + + 2.5
208 + ++ 4.8
209 + ++ 19.0
210 + + 4.3
211 ++ ++ 5.0
212 ++ +++ 7.8
213 + ++ 9.7
214 ++ ++ 2.5
215 + ++ 5.7
216 ++ ++ 4.0
217 ++ ++ 5.8
218 +++ +++ 2.6
219 ++ ++ 2.4
220 ++ +++ 4.4
221 ++ ++ 1.3
222 ++ ++ 2.4
223 ++ ++ 5.9
224 +++ +++ 2.4
225 + ++ 3.8
226 ++ ++ 1.9
227 + + 7.9
228 + + 3.9
229 ++ +++ 6.4
230 ++ ++++ 9.3
231 ++ +++ 5.1
232 ++ + 0.7
233 +++ +++ 2.2
234 + ++ 4.2
235 ++ ++ 2.5
236 + + 5.5
237 ++ ++ 2.3
238 + ++ 6.8
239 ++ ++ 2.3
240 + ++ 8.7
241 + + 3.8
242 + ++ 8.3
243 +++ ++++ 6.4 Example JAK1 JAK2 IC5o Ratio IQo iM)1 IC50 (nM)2 JAK2/JAK1
244 ++ ++ 1.8
245 ++ ++ 1.5
246 + ++ 7.5
247 + + 5.1
248 +++ ++++ 5.2
249 ++ ++ 2.1
250 + ++ 4.7
251 + + 4.9
252 ++ ++ 2.3
253 + + 5.8
254 + ++ 3.8
255 ++ ++ 3.2
256 + ++ 8.2
257 ++ ++ 4.4
258 + + 5.0
259 + + 4.6
260 + ++ 7.1
261 ++ ++++ 20.8
262 ++ +++ 7.4
263 + +++ 12.2
264 ++ +++ 9.5
265 ++ +++ 6.6
266 ++ +++ 6.6
267 ++ +++ 6.6
268 ++ ++ 3.1
269 + ++ 5.0
270 + + 6.1
271 + ++ 7.8
272 ++ ++ 6.5
273 + + 4.4
274 + ++ 9.0
275 ++ ++ 3.1
276 + + 4.0
277 + ++ 9.8
278 + + 5.6
279 + + 4.1
280 + + 7.5
281 + ++ 10.0
282 ++ ++ 3.4
283 + +++ 35.6
284 + ++ 5.8
285 ++ ++ 5.7
286 + ++ 13.1
287 ++ +++ 6.7
288 + + 9.0
289 + ++ 14.0
290 +++ ++++ 3.9 Example JAK1 JAK2 IC5o Ratio IQo iM)1 IC50 (nM)2 JAK2/JAK1
291 + +++ 20.3
292 + ++ 9.1
293 + ++ 3.4
294 ++ ++++ 21.4
295 ++ ++ 5.1
296 + ++ 4.6
297 + + 10.0
298 +++ +++ 2.8
299 +++ ++++ 6.6
300 ++ ++++ 11.7
301 +++ +++ 3.3
302 +++ ++++ 6.5
303 + +++ 19.7
304 + ++ 10.3
305 + +++ 16.0
306 ++ ++ 3.5
307 ++ +++ 8.8
308 + +++ 10.4
309 ++ ++ 5.3
310 + ++ 9.0
311 ++ ++++ 11.1
312 ++ +++ 5.3
313 + ++ 14.3
314 + ++ 11.0
315 ++ +++ 16.2
316 + ++ 9.5
317 + ++ 4.5
318 ++ ++ 4.7
319 + ++ 4.0
320 + ++ 2.8
321 + ++ 3.0
322 + + 4.7
323 + + 5.0
324 + ++ 5.6
325 ++ +++ 4.3
326 ++ ++ 1.7
327 ++ +++ 4.1
328 + ++ 12.9
329 ++ +++ 4.5
330 + ++ 6.7
331 + ++ 10.8
332 + +++ 19.3
333 ++ +++ 8.8
334 +++ ++++ 4.3
335 + + 2.4
336 + + 2.9
337 ++ ++ 6.0 Example JAK1 JAK2 IC5o Ratio
IQo iM)1 IC50 (nM)2 JAK2/JAK1
338 ++ +++ 7.4
339 ++ ++ 4.1
340 +++ ++ 1.4
341 ++ +++ 3.2
342 + ++ 5.7
343 ++ +++ 5.5
344 + +++ 18.9
345 ++ +++ 11.9
346 + ++ 5.9
347 ++ ++ 2.7
348 + ++ 3.2
349 +++ +++ 3.3
350 ++ ++++ 15.0
351 + ++ 3.8
352 + ++ 18.8
353 ++ ++ 5.0
354 ++ ++ 2.1
355 + + 3.1
356 ++ +++ 5.8
357 + ++ 3.8
358 + +++ 49.2
359 + ++++ 59.2
360 + ++++ 56.4
361 + +++ 20.9
362 + +++ 22.4
363 + +++ 32.9
364 + +++ 37.8
365 + ++ 15.5
366 + ++ 68.4
367 + ++ 33.8
368 + ++ 54.2
369 + ++++ 43.8
370 + +++ 55
371 + +++ 65.3
372 + ++ 67.3
373 + ++ 36.8
374 + ++ 50
375 + ++ 12.7
376 + ++ 69.2
377 + ++ 35.7
378 + ++ 32.7
379 + +++ 36.9
380 + ++ 15.9
381 + ++ 20
382 + ++ 26.7 Example JAK1 JAK2 IC5o Ratio
IQo iM)1 IC50 (nM)2 JAK2/JAK1
383 + ++ 30.5
384 + +++ 29.6
385 + +++ 28.9
386 + ++ 7.7
387 + ++ 9.7
388 ++ ++ 7.6
389 + ++ 6.5
390 + ++ 8.3
391 ++ +++ 10.6
392 ++ ++ 5.6
393 + ++ 7.1
394 ++ +++ 11.1
395 + +++ 24.4
396 + ++ 7.1
397 + ++ 21.7
^or JAK1 : 5 nM or less (+); > 5 nM to 20 nM (++); > 20 nM to 30 nM (+++); and > 30 nM (++++)
2For JAK2: 10 nM or less (+); > 10 nM to 50 nM (++); > 50 nM to 100 nM (+++); and > 100 nM (++++)
Example B: Cellular Assays
Cancer cell lines dependent on cytokines and hence JAK/STAT signal transduction, for growth, can be plated at 6000 cells per well (96 well plate format) in RPMI 1640, 10% FBS, and 1 nG/mL of appropriate cytokine. Compounds can be added to the cells in DMSO/media (final concentration 0.2% DMSO) and incubated for 72 hours at 37 °C, 5%> C02. The effect of compound on cell viability is assessed using the CellTiter-Glo Luminescent Cell Viability Assay (Promega) followed by TopCount (Perkin Elmer, Boston, MA) quantitation. Potential off-target effects of compounds are measured in parallel using a non-JAK driven cell line with the same assay readout. All experiments are typically performed in duplicate.
The above cell lines can also be used to examine the effects of compounds on phosphorylation of JAK kinases or potential downstream substrates such as STAT proteins, Akt, Shp2, or Erk. These experiments can be performed following an overnight cytokine starvation, followed by a brief preincubation with compound (2 hours or less) and cytokine stimulation of approximately 1 hour or less. Proteins are then extracted from cells and analyzed by techniques familiar to those schooled in the art including Western blotting or ELISAs using antibodies that can differentiate between phosphorylated and total protein. These experiments can utilize normal or cancer cells to investigate the activity of compounds on tumor cell survival biology or on mediators of inflammatory disease. For example, with regards to the latter, cytokines such as IL-6, IL- 12, IL-23, or IFN can be used to stimulate JAK activation resulting in phosphorylation of STAT protein(s) and potentially in transcriptional profiles (assessed by array or qPCR technology) or production and/or secretion of proteins, such as IL-17. The ability of compounds to inhibit these cytokine mediated effects can be measured using techniques common to those schooled in the art.
Compounds herein can also be tested in cellular models designed to evaluate their potency and activity against mutant JAKs, for example, the JAK2V617F mutation found in myeloid proliferative disorders. These experiments often utilize cytokine dependent cells of hematological lineage (e.g. BaF/3) into which the wild-type or mutant JAK kinases are ectopically expressed (James, C, et al. Nature 434:1144-1148; Staerk, J., et al. JBC 280:41893-41899). Endpoints include the effects of compounds on cell survival, proliferation, and phosphorylated JAK, STAT, Akt, or Erk proteins.
Certain compounds herein can be evaluated for their activity inhibiting T-cell proliferation. Such as assay can be considered a second cytokine (i.e. JAK) driven proliferation assay and also a simplistic assay of immune suppression or inhibition of immune activation. The following is a brief outline of how such experiments can be performed. Peripheral blood mononuclear cells (PBMCs) are prepared from human whole blood samples using Ficoll Hypaque separation method and T-cells (fraction 2000) can be obtained from PBMCs by elutriation. Freshly isolated human T-cells can be maintained in culture medium (RPMI 1640 supplemented with 10% fetal bovine serum, 100 U/ml penicillin, 100 μg/ml streptomycin) at a density of 2 x 106 cells/ml at 37 °C for up to 2 days. For IL-2 stimulated cell proliferation analysis, T-cells are first treated with Phytohemagglutinin (PHA) at a final concentration of 10 μg/mL for 72h. After washing once with PBS, 6000 cells/well are plated in 96-well plates and treated with compounds at different concentrations in the culture medium in the presence of 100 U/mL human IL- 2 (ProSpec-Tany TechnoGene; Rehovot, Israel). The plates are incubated at 37 °C for 72h and the proliferation index is assessed using CellTiter-Glo Luminescent reagents following the manufactory suggested protocol (Promega; Madison, WI).
Example C: In vivo anti-tumor efficacy
Compounds herein can be evaluated in human tumor xenograft models in immune compromised mice. For example, a tumorigenic variant of the INA-6 plasmacytoma cell line can be used to inoculate SCID mice subcutaneous ly (Burger, R., et al. Hematol J. 2:42-53, 2001). Tumor bearing animals can then be randomized into drug or vehicle treatment groups and different doses of compounds can be administered by any number of the usual routes including oral, i.p., or continuous infusion using implantable pumps. Tumor growth is followed over time using calipers. Further, tumor samples can be harvested at any time after the initiation of treatment for analysis as described above (Example B) to evaluate compound effects on JAK activity and downstream signaling pathways. In addition, selectivity of the compound(s) can be assessed using xenograft tumor models that are driven by other know kinases (e.g. Bcr-Abl) such as the K562 tumor model.
Example D: Murine Skin Contact Delayed Hypersensitivity Response Test
Compounds herein can also be tested for their efficacies (of inhibiting JAK targets) in the T-cell driven murine delayed hypersensitivity test model. The murine skin contact delayed-type hypersensitivity (DTH) response is considered to be a valid model of clinical contact dermatitis, and other T-lymphocyte mediated immune disorders of the skin, such as psoriasis (Immunol Today. 1998 Jan;19(l):37-44). Murine DTH shares multiple characteristics with psoriasis, including the immune infiltrate, the accompanying increase in inflammatory cytokines, and keratinocyte hyperproliferation. Furthermore, many classes of agents that are efficacious in treating psoriasis in the clinic are also effective inhibitors of the DTH response in mice (Agents Actions. 1993 Jan;38(l-2): 116- 21).
On Day 0 and 1, Balb/c mice are sensitized with a topical application, to their shaved abdomen with the antigen 2,4,dinitro-fluorobenzene (DNFB). On day 5, ears are measured for thickness using an engineer's micrometer. This measurement is recorded and used as a baseline. Both of the animals' ears are then challenged by a topical application of DNFB in a total of 20 μΐ^ (10 μΐ^ on the internal pinna and 10 μΐ^ on the external pinna) at a concentration of 0.2%. Twenty-four to seventy-two hours after the challenge, ears are measured again. Treatment with the test compounds is given throughout the sensitization and challenge phases (day -1 to day 7) or prior to and throughout the challenge phase (usually afternoon of day 4 to day 7). Treatment of the test compounds (in different concentration) is administered either systemically or topically (topical application of the treatment to the ears). Efficacies of the test compounds are indicated by a reduction in ear swelling comparing to the situation without the treatment. Compounds causing a reduction of 20% or more were considered efficacious. In some experiments, the mice are challenged but not sensitized (negative control).
The inhibitive effect (inhibiting activation of the JAK-STAT pathways) of the test compounds can be confirmed by immunohistochemical analysis. Activation of the JAK- STAT pathway(s) results in the formation and translocation of functional transcription factors. Further, the influx of immune cells and the increased proliferation of
keratinocytes should also provide unique expression profile changes in the ear that can be investigated and quantified. Formalin fixed and paraffin embedded ear sections
(harvested after the challenge phase in the DTH model) are subjected to
immunohistochemical analysis using an antibody that specifically interacts with phosphorylated STAT3 (clone 58E12, Cell Signaling Technologies). The mouse ears are treated with test compounds, vehicle, or dexamethasone (a clinically efficacious treatment for psoriasis), or without any treatment, in the DTH model for comparisons. Test compounds and the dexamethasone can produce similar transcriptional changes both qualitatively and quantitatively, and both the test compounds and dexamethasone can reduce the number of infiltrating cells. Both systemically and topical administration of the test compounds can produce inhibitive effects, i.e., reduction in the number of infiltrating cells and inhibition of the transcriptional changes. Example E: In vivo anti-inflammatory activity
Compounds herein can be evaluated in rodent or non-rodent models designed to replicate a single or complex inflammation response. For instance, rodent models of arthritis can be used to evaluate the therapeutic potential of compounds dosed
preventatively or therapeutically. These models include but are not limited to mouse or rat collagen-induced arthritis, rat adjuvant-induced arthritis, and collagen antibody- induced arthritis. Autoimmune diseases including, but not limited to, multiple sclerosis, type I-diabetes mellitus, uveoretinitis, thyroditis, myasthenia gravis, immunoglobulin nephropathies, myocarditis, airway sensitization (asthma), lupus, or colitis may also be used to evaluate the therapeutic potential of compounds herein. These models are well established in the research community and are familiar to those schooled in the art (Current Protocols in Immunology, Vol 3., Coligan, J.E. et al, Wiley Press.; Methods in Molecular Biology: Vol. 225, Inflammation Protocols., Winyard, P.G. and Willoughby, D.A., Humana Press, 2003.).
Example F: Animal Models for the Treatment of Dry Eye, Uveitis, and Conjunctivitis
Agents may be evaluated in one or more preclinical models of dry eye known to those schooled in the art including, but not limited to, the rabbit concanavalin A (ConA) lacrimal gland model, the scopolamine mouse model (subcutaneous or transdermal), the Botulinumn mouse lacrimal gland model, or any of a number of spontaneous rodent autoimmune models that result in ocular gland dysfunction (e.g. NOD-SCID, MRL/lpr, or NZB/NZW) (Barabino et al, Experimental Eye Research 2004, 79, 613-621 and
Schrader et al., Developmental Opthalmology, Karger 2008, 41, 298-312, each of which is incorporated herein by reference in its entirety). Endpoints in these models may include histopathology of the ocular glands and eye (cornea, etc.) and possibly the classic Schirmer test or modified versions thereof (Barabino et al.) which measure tear production. Activity may be assessed by dosing via multiple routes of administration (e.g. systemic or topical) which may begin prior to or after measurable disease exists.
Agents may be evaluated in one or more preclinical models of uveitis known to those schooled in the art. These include, but are not limited to, models of experimental autoimmune uveitis (EAU) and endotoxin induced uveitis (EIU). EAU experiements may be performed in the rabbit, rat, or mouse and may involve passive or activate immunization. For instance, any of a number or retinal antigens may be used to sensitize animals to a relevant immunogen after which animals may be challenged ocuarly with the same antigen. The EIU model is more acute and involves local or systemic
administration of lipopolysaccaride at sublethal doses. Endpoints for both the EIU and EAU models may include fundoscopic exam, histopathology amongst others. These models are reviewed by Smith et al. (Immunology and Cell Biology 1998, 76, 497-512, which is incorporated herein by reference in its entirety). Activity is assessed by dosing via multiple routes of administration (e.g. systemic or topical) which may begin prior to or after measurable disease exists. Some models listed above may also develop scleritis/episcleritis, chorioditis, cyclitis, or iritis and are therefore useful in investigating the potential activity of compounds for the therapeutic treatment of these diseases.
Agents may also be evaluated in one or more preclinical models of conjunctivitis known those schooled in the art. These include, but are not limited to, rodent models utilizing guinea-pig, rat, or mouse. The guinea-pig models include those utilizing active or passive immunization and/or immune challenge protocols with antigens such as ovalbumin or ragweed (reviewed in Groneberg, D.A., et al, Allergy 2003, 58, 1101- 1113, which is incorporated herein by reference in its entirety). Rat and mouse models are similar in general design to those in the guinea-pig (also reviewed by Groneberg). Activity may be assessed by dosing via multiple routes of administration (e.g. systemic or topical) which may begin prior to or after measurable disease exists. Endpoints for such studies may include, for example, histological, immunological, biochemical, or molecular analysis of ocular tissues such as the conjunctiva.
Example G: In vivo protection of bone
Compounds may be evaluated in various preclinical models of osteopenia, osteoporosis, or bone resorption known to those schooled in the art. For example, ovariectomized rodents may be used to evaluate the ability of compounds to affect signs and markers of bone remodeling and/or density (W.S.S. Jee and W. Yao, J Musculoskel. Nueron. Interact., 2001, 1(3), 193-207, which is incorporated herein by reference in its entirety). Alternatively, bone density and architecture may be evaluated in control or compound treated rodents in models of therapy (e.g. glucocorticoid) induced osteopenia (Yao, et al. Arthritis and Rheumatism, 2008, 58(6), 3485-3497; and id. 58(11), 1674- 1686, both of which are incorporated herein by reference in its entirety). In addition, the effects of compounds on bone resorption and density may be evaluable in the rodent models of arthritis discussed above (Example E). Endpoints for all these models may vary but often include histological and radiological assessments as well as
immunohisotology and appropriate biochemical markers of bone remodeling.
Various modifications of the invention, in addition to those described herein, will be apparent to those skilled in the art from the foregoing description. Such modifications are also intended to fall within the scope of the appended claims. Each reference, including all patent, patent applications, and publications, cited in the present application is incorporated herein by reference in its entirety.

Claims

WHAT IS CLAIMED IS:
1. A compound of Formula (I) :
Figure imgf000291_0001
I
or a pharmaceutically acceptable salt thereof; wherein:
X is N or CR2;
Y is N or CR3;
Z is H, cyano, halo, Ci_3 alkyl, or Ci_3 haloalkyl;
L is C(R4)2, C(=0), C(=0)N(R4a), C(=0)C(R4b)2, S(=0)2, C(=0)0,
C(=0)OC(R4b)2 or C(=0)N(R4a)C(R4b)2;
A is Ci_6 alkyl, C3_i4 cycloalkyl, C2_i3 heterocycloalkyl, C6-i4 aryl, or Ci_i4 heteroaryl; wherein said Ci_6 alkyl, C3_i4 cycloalkyl, C2_i3 heterocycloalkyl, C6-i4 aryl, and Ci_i4 heteroaryl are each optionally substituted with 1, 2, 3, 4, 5, or 6 independently selected R5 groups;
each R1 is, independently, Ci_4 alkyl, hydroxyl, Ci_4 alkoxy, fluoro, hydroxyl-Ci_4 alkyl, or Ci_4 alkoxy-Ci_4 alkyl; or
two R1 groups together form a 2- or 3 -carbon bridge or a bridge of formula -CH2- 0-CH2-;
R2 is H, halo, hydroxyl, cyano, Ci_4 alkyl, Ci_4 haloalkyl, or Ci_4 alkoxy;
R3 is H, cyano, nitro, halo, hydroxyl, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, C3_7 cycloalkyl, Ci_6 haloalkyl, Ci_6 alkoxy, Ci_6 haloalkoxy, amino, Ci_6 alkylamino, di-Ci_6 alkylamino, Ci_6 alkylsulfonyl, carbamyl, Ci_6 alkylcarbamyl, di-Ci_6 alkylcarbamyl, carboxy, Ci_6 alkylcarbonyl, or Ci_6 alkoxycarbonyl; each R4 is, independently, H or Ci_4 alkyl; or
two R4 groups, together with the carbon atom to which they are attached, form a 3-, 4-, 5-, or 6-membered cycloalkyl ring;
R4a is H or Ci_4 alkyl;
each R4b is, independently, H or Ci_4 alkyl; or
two R4b groups, together with the carbon atom to which they are attached, form a 3-, 4-, 5-, or 6-membered cycloalkyl ring;
each R5 is, independently, halo, cyano, nitro, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, Ci_6 haloalkyl, halosulfanyl, C3-10 cycloalkyl, C3-10 cycloalkyl-Ci_3 alkyl, C2_io
heterocycloalkyl, C2_i0 heterocycloalkyl-Ci_3 alkyl, C6_io aryl, C6_io aryl-Ci_3 alkyl, C1-10 heteroaryl, CM0 heteroaryl-Ci_3 alkyl, ORa, SRa, C(0)Rb, C(0)NRcRd, C(0)ORa,
OC(0)Rb, OC(0)NRcRd, NRcRd, NRcC(0)Rb, NRcC(0)ORa, NRcC(0)NRcRd,
C(=NRe)Rb, C(=NRe)NRcRd, NRcC(=NRe)NRcRd, NRcS(0)Rb, NRcS(0)2Rb,
NRcS(0)2NRcRd, S(0)Rb, S(0)NRcRd, S(0)2Rb, or S(0)2NRcRd; wherein Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, Ci_6 haloalkyl, C3_io cycloalkyl, C3_io cycloalkyl-Ci_3 alkyl, C2_io heterocycloalkyl, C2_io heterocycloalkyl-Ci_3 alkyl, C6-io aryl, C6-io aryl-Ci_3 alkyl, C1-10 heteroaryl, and C1-10 heteroaryl-Ci_3 alkyl are each optionally substituted with 1, 2, 3, 4, or 5 independently selected R6 groups;
each R6 is, independently, halo, cyano, nitro, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, Ci_6 haloalkyl, halosulfanyl, C3_io cycloalkyl, C3_io cycloalkyl-Ci_3 alkyl, C2_io
heterocycloalkyl, C2_io heterocycloalkyl-Ci_3 alkyl, C6-io aryl, C6-io aryl-Ci_3 alkyl, C1-10 heteroaryl, CM0 heteroaryl-Ci_3 alkyl, ORal, SRal, C(0)Rbl, C(0)NRclRdl, C(0)ORal, OC(0)Rbl, OC(0)NRclRdl, C(=NRel)NRclRdl, NRclC(=NRel)NRclRdl, NRclRdl, NRclC(0)Rbl, NRclC(0)ORal, NRclC(0)NRclRdl, NRclS(0)Rbl, NRclS(0)2Rbl,
NRclS(0)2NRclRdl, S(0)Rbl, S(0)NRclRdl, S(0)2Rbl, or S(0)2NRclRdl; wherein said Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, Ci_6 haloalkyl, C3_io cycloalkyl, C3_io cycloalkyl-Ci_3 alkyl, C2_i0 heterocycloalkyl, C2_i0 heterocycloalkyl-Ci_3 alkyl, C6_io aryl, C6_io aryl-Ci_3 alkyl, C1-10 heteroaryl, and C1-10 heteroaryl-Ci_3 alkyl are each optionally substituted by 1, 2, 3, 4, or 5 independently selected Rh groups;
each Ra, Rb, Rc, and Rd is, independently, H, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, Ci_6 haloalkyl, C3_i0 cycloalkyl, C3_i0 cycloalkyl-Ci_3 alkyl, C2_i0 heterocycloalkyl, C2_i0 heterocycloalkyl-Ci_3 alkyl, C6-io aryl, C6-io aryl-Ci_3 alkyl, Ci_io heteroaryl, or Ci_io heteroaryl-Ci_3 alkyl; wherein said Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, Ci_6 haloalkyl, halosulfanyl, C3-10 cycloalkyl, C3-10 cycloalkyl-Ci_3 alkyl, C2_io heterocycloalkyl, C2_io heterocycloalkyl-Ci_3 alkyl, C6-io aryl, C6-io aryl-Ci_3 alkyl, Ci_io heteroaryl, and Ci_io heteroaryl-Ci_3 alkyl is optionally substituted with 1 , 2, 3, 4, or 5 independently selected Rg groups;
or any Rc and Rd together with the N atom to which they are attached form a 3-, 4- , 5-, 6-, or 7-membered heterocycloalkyl group or a heteroaryl group, each optionally substituted with 1 , 2, 3 or 4 substituents independently selected from halo, cyano, nitro, hydroxyl, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, C3_7 cycloalkyl, Ci_6 haloalkyl, Ci_6 alkoxy, Ci_6 haloalkoxy, amino, Ci_6 alkylamino, di-Ci_6 alkylamino, thio, Ci_6 alkylthio, Ci_6 alkylsulfmyl, Ci_6 alkylsulfonyl, carbamyl, Ci_6 alkylcarbamyl, di-Ci_6 alkylcarbamyl, carboxy, Ci_6 alkylcarbonyl, and Ci_6 alkoxycarbonyl;
each Re is, independently, H, Ci_6 alkyl, CN, hydroxyl, Ci_6 alkoxy, Ci_6 alkylsulfonyl, carboxy, Ci_6 alkylcarbonyl, aminosulfonyl, Ci_6 alkylaminosulfonyl, di-Ci_ 6 alkylaminosulfonyl, carbamyl, Ci_6 alkylcarbamyl, or di-Ci_6 alkylcarbamyl;
each Ral, Rbl, Rcl, and Rdl is, independently, H, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, Ci_6 haloalkyl, C3_i0 cycloalkyl, C3_i0 cycloalkyl-Ci_3 alkyl, C2_i0 heterocycloalkyl, C2_io heterocycloalkyl-Ci_3 alkyl, C6_io aryl, C6_io aryl-Ci_3 alkyl, Ci_i0 heteroaryl, or Ci_i0 heteroaryl-Ci_3 alkyl; wherein said Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, Ci_6 haloalkyl, halosulfanyl, C3-10 cycloalkyl, C3-10 cycloalkyl-Ci_3 alkyl, C2_io heterocycloalkyl, C2_io heterocycloalkyl-Ci_3 alkyl, C6-io aryl, C6-io aryl-Ci_3 alkyl, Ci_io heteroaryl, and Ci_io heteroaryl-Ci_3 alkyl is optionally substituted with 1 , 2, 3, 4, or 5 independently selected Rg groups;
or any Rcl and Rdl together with the N atom to which they are attached form a 3-, 4-, 5-, 6-, or 7-membered heterocycloalkyl group or a heteroaryl group, each optionally substituted with 1 , 2, 3 or 4 substituents independently selected from halo, cyano, nitro, hydroxyl, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, C3_7 cycloalkyl, Ci_6 haloalkyl, Ci_6 alkoxy, Ci_6 haloalkoxy, amino, Ci_6 alkylamino, di-Ci_6 alkylamino, thio, Ci_6 alkylthio, Ci_6 alkylsulfmyl, Ci_6 alkylsulfonyl, carbamyl, Ci_6 alkylcarbamyl, di-Ci_6 alkylcarbamyl, carboxy, Ci_6 alkylcarbonyl, Ci_6 alkoxycarbonyl, and Ci_6 alkylcarbonylamino; each Rel is, independently, H, Ci_6 alkyl, CN, hydroxyl, Ci_6 alkoxy, Ci_6 alkylsulfonyl, carboxy, Ci_6 alkylcarbonyl, aminosulfonyl, Ci_6 alkylaminosulfonyl, di-Ci_ 6 alkylaminosulfonyl, carbamyl, Ci_6 alkylcarbamyl, or di-Ci_6 alkylcarbamyl;
each Rg, Rg , and Rh is, independently, halo, cyano, nitro, hydroxyl, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, C3-7 cycloalkyl, Ci_6 haloalkyl, Ci_6 alkoxy, Ci_6 haloalkoxy, amino, Ci_6 alkylamino, di-Ci_6 alkylamino, thio, Ci_6 alkylthio, Ci_6 alkylsulfmyl, Ci_6 alkylsulfonyl, carbamyl, Ci_6 alkylcarbamyl, di-Ci_6 alkylcarbamyl, carboxy, Ci_6 alkylcarbonyl, Ci_6 alkoxycarbonyl, or Ci_6 alkylcarbonylamino; and
n is 0, 1, 2, 3, or 4.
2. A compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein:
X is N or CR2;
Y is N or CR3;
Z is H, cyano, halo, Ci_3 alkyl, or Ci_3 haloalkyl;
L is C(R4)2, C(=0), C(=0)N(R4a), C(=0)C(R4b)2, or S(=0)2;
A is Ci_6 alkyl, C3_i4 cycloalkyl, C2_i3 heterocycloalkyl, C6-i4 aryl, or Ci_i4 heteroaryl; wherein said Ci_6 alkyl, C3_i4 cycloalkyl, C2_i3 heterocycloalkyl, C6-i4 aryl, and Ci_i4 heteroaryl are each optionally substituted with 1, 2, 3, 4, 5, or 6 independently selected R5 groups;
each R1 is, independently, Ci_4 alkyl; or
two R1 groups together form a 2- or 3-carbon bridge;
R2 is H, halo, hydroxyl, cyano, Ci_4 alkyl, Ci_4 haloalkyl, or Ci_4 alkoxy;
R3 is H, cyano, nitro, halo, hydroxyl, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, C3_7 cycloalkyl, Ci_6 haloalkyl, Ci_6 alkoxy, Ci_6 haloalkoxy, amino, Ci_6 alkylamino, di-Ci_6 alkylamino, Ci_6 alkylsulfonyl, carbamyl, Ci_6 alkylcarbamyl, di-Ci_6 alkylcarbamyl, carboxy, Ci_6 alkylcarbonyl, or Ci_6 alkoxycarbonyl;
each R4 is, independently, H or Ci_4 alkyl; or
two R4 groups, together with the carbon atom to which they are attached, form a 3-, 4-, 5-, or 6-membered cycloalkyl ring;
R4a is H or Ci_4 alkyl;
each R4b is, independently, H or Ci_4 alkyl; or two R groups, together with the carbon atom to which they are attached, form a 3-, 4-, 5-, or 6-membered cycloalkyl ring;
each R5 is, independently, halo, cyano, nitro, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, Ci_6 haloalkyl, halosulfanyl, C3-10 cycloalkyl, C3-10 cycloalkyl-Ci_3 alkyl, C2-10
heterocycloalkyl, C2-10 heterocycloalkyl-Ci_3 alkyl, C6-io aryl, C6-io aryl-Ci_3 alkyl, C1-10 heteroaryl, CM0 heteroaryl-Ci_3 alkyl, ORa, SRa, C(0)Rb, C(0)NRcRd, C(0)ORa,
OC(0)Rb, OC(0)NRcRd, NRcRd, NRcC(0)Rb, NRcC(0)ORa, NRcC(0)NRcRd,
C(=NRe)Rb, C(=NRe)NRcRd, NRcC(=NRe)NRcRd, NRcS(0)Rb, NRcS(0)2Rb,
NRcS(0)2NRcRd, S(0)Rb, S(0)NRcRd, S(0)2Rb, or S(0)2NRcRd; wherein Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, Ci_6 haloalkyl, C3_i0 cycloalkyl, C3_i0 cycloalkyl-Ci_3 alkyl, C2_i0 heterocycloalkyl, C2_io heterocycloalkyl-Ci_3 alkyl, C6-io aryl, C6-io aryl-Ci_3 alkyl, C1-10 heteroaryl, and C1-10 heteroaryl-Ci_3 alkyl are each optionally substituted with 1, 2, 3, 4, or 5 independently selected R6 groups;
each R6 is, independently, halo, cyano, nitro, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, Ci_6 haloalkyl, halosulfanyl, C3_io cycloalkyl, C3_io cycloalkyl-Ci_3 alkyl, C2_io
heterocycloalkyl, C2_io heterocycloalkyl-Ci_3 alkyl, C6-io aryl, C6-io aryl-Ci_3 alkyl, C1-10 heteroaryl, CM0 heteroaryl-Ci_3 alkyl, ORal, SRal, C(0)Rbl, C(0)NRclRdl, C(0)ORal, OC(0)Rbl, OC(0)NRclRdl, C(=NRel)NRclRdl, NRclC(=NRel)NRclRdl, NRclRdl, NRclC(0)Rbl, NRclC(0)ORal, NRclC(0)NRclRdl, NRclS(0)Rbl, NRclS(0)2Rbl,
NRclS(0)2NRclRdl, S(0)Rbl, S(0)NRclRdl, S(0)2Rbl, or S(0)2NRclRdl; wherein said Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, Ci_6 haloalkyl, C3_io cycloalkyl, C3_io cycloalkyl-Ci_3 alkyl, C2_io heterocycloalkyl, C2_io heterocycloalkyl-Ci_3 alkyl, C6-io aryl, C6-io aryl-Ci_3 alkyl, C1-10 heteroaryl, and C1-10 heteroaryl-Ci_3 alkyl are each optionally substituted by 1, 2, 3, 4, or 5 independently selected Rh groups;
each Ra, Rb, Rc, and Rd is, independently, H, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, Ci_6 haloalkyl, C3_io cycloalkyl, C3_io cycloalkyl-C i_3 alkyl, C2_io heterocycloalkyl, C2_io heterocycloalkyl-Ci_3 alkyl, C6_io aryl, C6_io aryl-Ci_3 alkyl, C1-10 heteroaryl, or C1-10 heteroaryl-Ci_3 alkyl; wherein said Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, Ci_6 haloalkyl, halosulfanyl, C3_io cycloalkyl, C3_io cycloalkyl-C i_3 alkyl, C2_io heterocycloalkyl, C2_io heterocycloalkyl-Ci_3 alkyl, C6-io aryl, C6-io aryl-Ci_3 alkyl, C1-10 heteroaryl, and C1-10 heteroaryl-Ci_3 alkyl is optionally substituted with 1 , 2, 3, 4, or 5 independently selected Rg groups;
or any Rc and Rd together with the N atom to which they are attached form a 3-, 4- , 5-, 6-, or 7-membered heterocycloalkyl group or a heteroaryl group, each optionally substituted with 1 , 2, 3 or 4 substituents independently selected from halo, cyano, nitro, hydroxyl, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, C3_7 cycloalkyl, Ci_6 haloalkyl, Ci_6 alkoxy, Ci_6 haloalkoxy, amino, Ci_6 alkylamino, di-Ci_6 alkylamino, thio, Ci_6 alkylthio, Ci_6 alkylsulfmyl, Ci_6 alkylsulfonyl, carbamyl, Ci_6 alkylcarbamyl, di-Ci_6 alkylcarbamyl, carboxy, Ci_6 alkylcarbonyl, and Ci_6 alkoxycarbonyl;
each Re is, independently, H, Ci_6 alkyl, CN, hydroxyl, Ci_6 alkoxy, Ci_6 alkylsulfonyl, carboxy, Ci_6 alkylcarbonyl, aminosulfonyl, Ci_6 alkylaminosulfonyl, di-Ci_ 6 alkylaminosulfonyl, carbamyl, Ci_6 alkylcarbamyl, or di-Ci_6 alkylcarbamyl;
each Ral, Rbl, Rcl, and Rdl is, independently, H, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, Ci_6 haloalkyl, C3_i0 cycloalkyl, C3_i0 cycloalkyl-Ci_3 alkyl, C2_i0 heterocycloalkyl, C2_io heterocycloalkyl-Ci_3 alkyl, C6-io aryl, C6-io aryl-Ci_3 alkyl, Ci_io heteroaryl, or Ci_io heteroaryl-Ci_3 alkyl; wherein said Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, Ci_6 haloalkyl, halosulfanyl, C3_io cycloalkyl, C3_io cycloalkyl-Ci_3 alkyl, C2_io heterocycloalkyl, C2_io heterocycloalkyl-Ci_3 alkyl, C6_io aryl, C6_io aryl-Ci_3 alkyl, Ci_i0 heteroaryl, and Ci_i0 heteroaryl-Ci_3 alkyl is optionally substituted with 1 , 2, 3, 4, or 5 independently selected Rg groups;
or any Rcl and Rdl together with the N atom to which they are attached form a 3-, 4-, 5-, 6-, or 7-membered heterocycloalkyl group or a heteroaryl group, each optionally substituted with 1 , 2, 3 or 4 substituents independently selected from halo, cyano, nitro, hydroxyl, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, C3_7 cycloalkyl, Ci_6 haloalkyl, Ci_6 alkoxy, Ci_6 haloalkoxy, amino, Ci_6 alkylamino, di-Ci_6 alkylamino, thio, Ci_6 alkylthio, Ci_6 alkylsulfmyl, Ci_6 alkylsulfonyl, carbamyl, Ci_6 alkylcarbamyl, di-Ci_6 alkylcarbamyl, carboxy, Ci_6 alkylcarbonyl, Ci_6 alkoxycarbonyl, and Ci_6 alkylcarbonylamino;
each Rel is, independently, H, Ci_6 alkyl, CN, hydroxyl, Ci_6 alkoxy, Ci_6 alkylsulfonyl, carboxy, Ci_6 alkylcarbonyl, aminosulfonyl, Ci_6 alkylaminosulfonyl, di-Ci_ 6 alkylaminosulfonyl, carbamyl, Ci_6 alkylcarbamyl, or di-Ci_6 alkylcarbamyl; each Rg, Rg , and Rh is, independently, halo, cyano, nitro, hydroxyl, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, C3_7 cycloalkyl, Ci_6 haloalkyl, Ci_6 alkoxy, Ci_6 haloalkoxy, amino, Ci_6 alkylamino, di-Ci_6 alkylamino, thio, Ci_6 alkylthio, Ci_6 alkylsulfmyl, Ci_6 alkylsulfonyl, carbamyl, Ci_6 alkylcarbamyl, di-Ci_6 alkylcarbamyl, carboxy, Ci_6 alkylcarbonyl, Ci_6 alkoxycarbonyl, or Ci_6 alkylcarbonylamino; and
n is 0, 1, 2, 3, or 4.
3. The compound according to claim 1 or 2, or a pharmaceutically acceptable salt thereof, wherein X is N.
4. The compound according to claim 1 or 2, or a pharmaceutically acceptable salt thereof, wherein X is CR2.
5. The compound according to claim 1 or 2, or a pharmaceutically acceptable salt thereof, wherein X is C(H), C(F), or C(CN).
6. The compound according to claim 1 or 2, or a pharmaceutically acceptable salt thereof, wherein X is CH.
7. The compound according to any one of claims 1 to 6, or a pharmaceutically
acceptable salt thereof, wherein Y is N.
8. The compound according to any one of claims 1 to 6, or a pharmaceutically
acceptable salt thereof, wherein Y is CR3.
9. The compound according to any one of claims 1 to 6, or a pharmaceutically
acceptable salt thereof, wherein Y is CH.
10. The compound according to any one of claims 1 to 9, or a pharmaceutically
acceptable salt thereof, wherein Z is cyano. compound according to any one of claims 1 to 10, or a pharmaceutically acceptable salt thereof, wherein L is C(=0)NH, C(=0), S(=0)2, CH2, C(=0)CH2,
Figure imgf000298_0001
compound according to any one of claims 1 to 10, or a pharmaceutically acceptable salt thereof, wherein L is C(=0)NH, C(=0), S(=0)2, CH2, C(=0)CH2,
Figure imgf000298_0002
13. The compound according to any one of claims 1 to 12, or a pharmaceutically acceptable salt thereof, wherein n is 0, 1, or 2.
14. The compound according to any one of claims 1 to 12, or a pharmaceutically acceptable salt thereof, wherein n is 0.
15. The compound according to any one of claims 1 to 14, or a pharmaceutically acceptable salt thereof, wherein R1 is Ci_4 alkyl.
16. The compound according to any one of claims 1 to 14, or a pharmaceutically acceptable salt thereof, wherein R1 is methyl.
17. The compound according to any one of claims 1 to 14, or a pharmaceutically acceptable salt thereof, wherein two R1 groups form a 2-carbon bridge.
18. The compound according to any one of claims 1 to 17, or a pharmaceutically acceptable salt thereof, wherein A is C6_i4 aryl, which is optionally substituted with 1, 2, 3, 4, 5, or 6 independently selected R5 groups.
19. The compound according to any one of claims 1 to 17, or a pharmaceutically acceptable salt thereof, wherein A is phenyl, which is optionally substituted with 1, 2, 3, 4, or 5 independently selected R5 groups.
20. The compound according to any one of claims 1 to 17, or a pharmaceutically acceptable salt thereof, wherein A is monocyclic C3_g cycloalkyl or bicyclic C3_g cycloalkyl, each of which is optionally substituted with 1, 2, 3, 4, 5, or 6 independently selected R5 groups.
21. The compound according to any one of claims 1 to 17, or a pharmaceutically acceptable salt thereof, wherein A is monocyclic C2-10 heterocycloalkyl or bicyclic C2-10 heterocycloalkyl, each of which is optionally substituted with 1, 2, 3, 4, 5, or 6 independently selected R5 groups.
22. The compound according to any one of claims 1 to 17, or a pharmaceutically acceptable salt thereof, wherein A is monocyclic C1-10 heteroaryl or bicyclic C1-10 heteroaryl, each of which is optionally substituted with 1, 2, 3, 4, 5, or 6 independently selected R5 groups.
23. The compound according to any one of claims 1 to 17, or a pharmaceutically acceptable salt thereof, wherein A is Ci_6 alkyl.
24. The compound according to any one of claims 1 to 17, or a pharmaceutically acceptable salt thereof, wherein A is Ci_6 alkyl, phenyl, a naphthyl ring, monocyclic or bicyclic C3_io cycloalkyl, monocyclic or bicyclic C2-10
heterocycloalkyl, or monocyclic or bicyclic C1-10 heteroaryl; each of which is optionally substituted with 1, 2, 3, 4, or 5 independently selected R5 groups.
25. The compound according to any one of claims 1 to 17, or a pharmaceutically acceptable salt thereof, wherein A is phenyl, a naphthyl ring, monocyclic C3_io cycloalkyl, bicyclic C3_i0 cycloalkyl, monocyclic C2-10 heterocycloalkyl, bicyclic C2-10 heterocycloalkyl, monocyclic Ci_io heteroaryl, or bicyclic Ci_io heteroaryl; each of which is optionally substituted with 1, 2, 3, 4, or 5 independently selected R5 groups.
26. The compound according to any one of claims 1 to 17, or a pharmaceutically acceptable salt thereof, wherein A is methyl, ethyl, isopropyl, phenyl, a naphthalene ring, a pyridine ring, a pyrimidine ring, a thiophene ring, a pyrazine ring, an oxazole ring, an isoxazole ring, an imidazole ring, a thiazole ring, a furan ring, a pyrazole ring, a quinoline ring, a benzothiophene ring, a
benzothiazole ring, a benzoimidazole ring, a benzofuran ring, cyclopropyl, cyclopentyl, cyclohexyl, cycloheptyl, an indene ring, a tetrahydronaphthalene ring, dihydro-l,4-benzodioxoxine ring, or a piperidine ring; each of which is optionally substituted with 1, 2, 3, 4, or 5 independently selected R5 groups, as permitted by valency.
27. The compound according to any one of claims 1 to 17, or a pharmaceutically acceptable salt thereof, wherein A is phenyl or a pyridine ring; each of which are optionally substituted with 1, 2, 3, or 4 independently selected R5 groups.
28. The compound according to any one of claims 1 to 17, or a pharmaceutically acceptable salt thereof, wherein A is pyridin-4-yl; which is optionally substituted with 1, 2, 3, or 4 independently selected R5 groups.
29. The compound according to any one of claims 1 to 28, or a pharmaceutically acceptable salt thereof, wherein wherein each R5 is, independently, halo, cyano, Ci_6 alkyl, Ci_6 haloalkyl, C6_io aryl, CM0 heteroaryl, ORa, SRa, C(0)ORa, NRcRd, NRcC(0)Rb; wherein said Ci_6 alkyl, Ci_6 haloalkyl, C6_io aryl, and Ci_i0 heteroaryl are each optionally substituted with 1, 2, 3, 4, or 5 independently selected R6 groups.
30. The compound according to any one of claims 1 to 28, or a pharmaceutically acceptable salt thereof, wherein each R5 is, independently, chloro, fluoro, bromo, cyano, methyl, ethyl, trifluoromethyl, hydroxyl, methoxy, trifluoromethoxy, difluoromethoxy, phenoxy, dimethylamino, t-butylcarbonylamino,
methoxycarbonyl, methylthio, phenyl, a pyridine ring, a thiazole ring, a quinoline ring, an isoquinoline ring, an imidazo[l,2-a]pyrimidine ring, a benzoxazole ring, or an oxadiazole ring; wherein said phenyl, pyridine ring, thiazole ring, quinoline ring, isoquinoline ring, imidazo[l,2-a]pyrimidine ring, benzoxazole ring, and oxadiazole ring are each optionally substituted with 1, 2, 3, 4, or 5 independently selected R6 groups.
31. The compound according to any one of claims 1 to 28, or a pharmaceutically acceptable salt thereof, wherein:
each R5 is, independently, halo, cyano, Ci_6 alkyl, Ci_6 haloalkyl, C6_io aryl, Ci_i0 heteroaryl, ORa, SRa, C(0)ORa, NRcRd, or NRcC(0)Rb; wherein said Ci_6 alkyl, Ci_6 haloalkyl, C6-io aryl, and Ci_io heteroaryl are each optionally substituted with 1, 2, 3, 4, or 5 independently selected R6 groups;
each Ra, Rb, Rc, and Rd is, independently, H, Ci_6 alkyl, Ci_6 haloalkyl, or C6_io aryl; wherein said Ci_6 alkyl and C6_io aryl is optionally substituted with 1, 2, 3, 4, or 5 independently selected Rg groups;
each R6 is, independently, halo, cyano, Ci_6 alkyl, Ci_6 haloalkyl, ORal, NRclRdl, or OC(0)Rbl; and
each Ral, Rbl, Rcl, and Rdl is, independently, H or Ci_6 alkyl; wherein said Ci_6 alkyl is optionally substituted with a substituent independently selected from Ci_4 alkoxy and hydroxyl;
or any Rcl and Rdl together with the N atom to which they are attached form a 4-, 5-, 6-, or 7-membered heterocycloalkyl group, optionally substituted with 1, 2, 3 or 4 substituents independently selected from halo.
32. The compound according to any one of claims 1 to 28, or a pharmaceutically acceptable salt thereof, wherein each R5 is, independently, halo or Ci_6 haloalkyl. The compound according to any one of claims 1 to 28, or a pharmaceutically acceptable salt thereof, wherein each R5 is, independently, fluoro or trifluoromethy 1.
The compound according to any one of claims 1 to 9 and 18 to 33, having Formula (II):
Figure imgf000302_0001
II
or a pharmaceutically acceptable salt thereof.
35. The compound according to any one of claims 1 to 9 and 18 to 33, having Formula (III):
Figure imgf000302_0002
III or a pharmaceutically acceptable salt thereof.
36. The compound according to claim 1 or 2, or a pharmaceutically acceptable salt thereof, wherein:
X is N or CR2;
Y is N or CR3;
each R1 is, independently, Ci_4 alkyl; or
two R1 groups form a 2-carbon bridge.
R2 is H, halo, or cyano;
R3 is H;
Z is cyano;
L is C(=0)NH, C(=0), S(=0)2, CH2, C(=0)CH2, or
Figure imgf000303_0001
A is Ci_6 alkyl, phenyl, a naphthyl ring, monocyclic C3_i0 cycloalkyl, bicyclic C3_i0 cycloalkyl, monocyclic C2_io heterocycloalkyl, bicyclic C2_io heterocycloalkyl, monocyclic Ci_io heteroaryl, or bicyclic Ci_io heteroaryl; each of which is optionally substituted with 1, 2, 3, 4, or 5 independently selected R5 groups;
each R5 is, independently, halo, cyano, Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, Ci_6 haloalkyl, C3_io cycloalkyl, C3_io cycloalkyl-Ci_3 alkyl, C2_io heterocycloalkyl, C2_io heterocycloalkyl-Ci_3 alkyl, C6-io aryl, C6-io aryl-Ci_3 alkyl, Ci_io heteroaryl, Ci_io heteroaryl-Ci_3 alkyl, ORa, SRa, C(0)Rb, C(0)NRcRd, C(0)ORa, OC(0)Rb, OC(0)NRcRd, NRcRd, NRcC(0)Rb, NRcC(0)ORa, NRcC(0)NRcRd, NRcS(0)Rb, NRcS(0)2Rb,
NRcS(0)2NRcRd, S(0)Rb, S(0)NRcRd, S(0)2Rb, or S(0)2NRcRd; wherein Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, Ci_6 haloalkyl, halosulfanyl, C3_io cycloalkyl, C3_io cycloalkyl-Ci_3 alkyl, C2_io heterocycloalkyl, C2_io heterocycloalkyl-Ci_3 alkyl, C6-io aryl, C6-io aryl-Ci_3 alkyl, Ci_io heteroaryl, and Ci_io heteroaryl-Ci_3 alkyl are each optionally substituted with 1, 2, 3, 4, or 5 independently selected R6 groups; and
n is 0, 1, or 2.
37. The compound according to claim 1 or 2, or a pharmaceutically acceptable salt thereof, wherein: X is N or CR2;
Y is N or CR3;
each R1 is, independently, Ci_4 alkyl; or
two R1 groups form a 2-carbon bridge.
R2 is H, halo, or cyano;
R3 is H;
Z is cyano;
L is C(=0)NH, C(=0), S(=0)2, CH2, C(=0)CH2, or
Figure imgf000304_0001
A is Ci_6 alkyl, phenyl, a naphthyl ring, monocyclic C3_i0 cycloalkyl, bicyclic C3_i0 cycloalkyl, monocyclic C2_io heterocycloalkyl, bicyclic C2_io heterocycloalkyl, monocyclic Ci_io heteroaryl, or bicyclic Ci_io heteroaryl; each of which is optionally substituted with 1, 2, 3, 4, or 5 independently selected R5 groups;
each R5 is, independently, halo, cyano, Ci_6 alkyl, Ci_6 haloalkyl, C3_i0 cycloalkyl, C2_io heterocycloalkyl, C6-io aryl, C6-io aryl-Ci_3 alkyl, Ci_io heteroaryl, ORa, SRa, C(0)Rb, C(0)NRcRd, C(0)ORa, OC(0)Rb, OC(0)NRcRd, NRcRd, NRcC(0)Rb, NRcC(0)ORa, NRcC(0)NRcRd, NRcS(0)2Rb, NRcS(0)2NRcRd, S(0)2Rb, or S(0)2NRcRd; wherein Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, Ci_6 haloalkyl, halosulfanyl, C3_i0 cycloalkyl, C3_i0 cycloalkyl-Ci_3 alkyl, C2_io heterocycloalkyl, C2_io heterocycloalkyl-Ci_3 alkyl, C6-io aryl, C6-io aryl-Ci_3 alkyl, Ci_io heteroaryl, and Ci_io heteroaryl-Ci_3 alkyl are each optionally substituted with 1, 2, 3, 4, or 5 independently selected R6 groups; and
n is 0, 1, or 2.
38. The compound according to claim 1 or 2, or a pharmaceutically acceptable salt thereof, wherein:
X is N or CR2;
Y is N or CR3;
each R1 is, independently, Ci_4 alkyl; or
two R1 groups form a 2-carbon bridge.
R2 is H, halo, or cyano;
R3 is H; Z is cyano;
L is C(=0)NH, C(=0), S(=0)2, CH2, C(=0)CH2, or
Figure imgf000305_0001
A is phenyl, a naphthyl ring, monocyclic C3-10 cycloalkyl, bicyclic C3-10 cycloalkyl, monocyclic C2_io heterocycloalkyl, bicyclic C2_io heterocycloalkyl, monocyclic Ci_io heteroaryl, or bicyclic Ci_io heteroaryl; each of which is optionally substituted with 1, 2, 3, 4, or 5 independently selected R5 groups;
each R5 is, independently, halo, cyano, Ci_6 alkyl, Ci_6 haloalkyl, C3-10 cycloalkyl, C2_io heterocycloalkyl, C6-io aryl, C6-io aryl-Ci_3 alkyl, Ci_io heteroaryl, ORa, SRa, C(0)Rb, C(0)NRcRd, C(0)ORa, OC(0)Rb, OC(0)NRcRd, NRcRd, NRcC(0)Rb, NRcC(0)ORa, NRcC(0)NRcRd, NRcS(0)2Rb, NRcS(0)2NRcRd, S(0)2Rb, or S(0)2NRcRd; wherein Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, Ci_6 haloalkyl, halosulfanyl, C3-10 cycloalkyl, C3-10 cycloalkyl-Ci_3 alkyl, C2_io heterocycloalkyl, C2_io heterocycloalkyl-Ci_3 alkyl, C6-io aryl, C6-10 aryl-Ci_3 alkyl, Ci_io heteroaryl, and Ci_io heteroaryl-Ci_3 alkyl are each optionally substituted with 1, 2, 3, 4, or 5 independently selected R6 groups; and
n is 0, 1, or 2.
39. The compound according to claim 1 or 2, or a pharmaceutically acceptable salt thereof, wherein:
X is N or CR2;
Y is N or CR3;
each R1 is, independently, Ci_4 alkyl; or
two R1 groups form a 2-carbon bridge.
R2 is H, halo, or cyano;
R3 is H;
Z is cyano;
L is C(=0)NH, C(=0), S(=0)2, CH2, C(=0)CH2, or
Figure imgf000305_0002
A is Ci_6 alkyl, phenyl, a naphthyl ring, monocyclic C3-10 cycloalkyl, bicyclic C3-10 cycloalkyl, monocyclic C2_io heterocycloalkyl, bicyclic C2_io heterocycloalkyl, monocyclic C1-10 heteroaryl, or bicyclic C1-10 heteroaryl; each of which is optionally substituted with 1, 2, 3, 4, or 5 independently selected R5 groups;
each R5 is, independently, halo, cyano, Ci_6 alkyl, Ci_6 haloalkyl, C6-io aryl, C1-10 heteroaryl, ORa, SRa, C(0)ORa, NRcRd, NRcC(0)Rb; wherein said Ci_6 alkyl, Ci_6 haloalkyl, C6-io aryl, and C1-10 heteroaryl are each optionally substituted with 1, 2, 3, 4, or 5 independently selected R6 groups;
and
n is 0, 1, or 2.
40. The compound according to claim 1 or 2, or a pharmaceutically acceptable salt thereof, wherein:
X is N or CR2;
Y is N or CR3;
each R1 is, independently, Ci_4 alkyl; or
two R1 groups form a 2-carbon bridge.
R2 is H, halo, or cyano;
R3 is H;
Z is cyano;
L is C(=0)NH, C(=0), S(=0)2, CH2, C(=0)CH2, or
Figure imgf000306_0001
A is phenyl, a naphthyl ring, monocyclic C3-10 cycloalkyl, bicyclic C3-10 cycloalkyl, monocyclic C2_io heterocycloalkyl, bicyclic C2_io heterocycloalkyl, monocyclic C1-10 heteroaryl, or bicyclic C1-10 heteroaryl; each of which is optionally substituted with 1, 2, 3, 4, or 5 independently selected R5 groups;
each R5 is, independently, halo, cyano, Ci_6 alkyl, Ci_6 haloalkyl, C6-io aryl, C1-10 heteroaryl, ORa, SRa, C(0)ORa, NRcRd, NRcC(0)Rb; wherein said Ci_6 alkyl, Ci_6 haloalkyl, C6_io aryl, and C1-10 heteroaryl are each optionally substituted with 1, 2, 3, 4, or
5 independently selected R6 groups;
and
n is 0, 1, or 2. The compound according to claim 1 or 2, or a pharmaceutically acceptable thereof, wherein:
X is N or CR2;
Y is N or CR3;
each R is, independently, methyl; or
two R1 groups form a 2-carbon bridge.
R2 is H, halo, or cyano;
R3 is H;
Z is cyano;
L is C(=0)NH, C(=0), S(=0)2, CH2, C(=0)CH2, or
Figure imgf000307_0001
A is methyl, ethyl, isopropyl, phenyl, a naphthalene ring, a pyridine ring, a pyrimidine ring, a thiophene ring, a pyrazine ring, an oxazole ring, an isoxazole ring, an imidazole ring, a thiazole ring, a furan ring, a pyrazole ring, a quinoline ring, a benzothiophene ring, a benzothiazole ring, a benzoimidazole ring, a benzofuran ring, cyclopropyl, cyclopentyl, cyclohexyl, cycloheptyl, an indene ring, a
tetrahydronaphthalene ring, dihydro-l,4-benzodioxoxine ring, or a piperidine ring; each of which are optionally substituted with 1, 2, 3, 4, 5, or 6 independently selected R5 groups, as permitted by valency;
each R5 is, independently, chloro, fluoro, bromo, cyano, methyl, ethyl, trifluoromethyl, hydroxyl, methoxy, trifluoromethoxy, difluoromethoxy, phenoxy, dimethylamino, t-butylcarbonylamino, methoxycarbonyl, methylthio, phenyl, a pyridine ring, a thiazole ring, a quinoline ring, an isoquinoline ring, an imidazo[l,2-a]pyrimidine ring, a benzoxazole ring, or an oxadiazole ring; wherein said phenyl, pyridine ring, thiazole ring, quinoline ring, isoquinoline ring, imidazo[l,2-a]pyrimidine ring, benzoxazole ring, and oxadiazole ring are each optionally substituted with 1, 2, 3, 4, or 5 independently selected R6 groups; and
n is 0, 1, or 2.
42. The compound according to claim 1 or 2, or a pharmaceutically acceptable salt thereof, wherein: X is N or CR2;
Y is N or CR3;
each R1 is, independently, methyl; or
two R1 groups form a 2-carbon bridge.
R2 is H, halo, or cyano;
R3 is H;
Z is cyano;
L is C(=0)NH, C(=0), S(=0)2, CH2, C(=0)CH2, or
Figure imgf000308_0001
A is phenyl, a naphthalene ring, a pyridine ring, a pyrimidine ring, a thiophene ring, a pyrazine ring, an oxazole ring, an isoxazole ring, an imidazole ring, a thiazole ring, a furan ring, a pyrazole ring, a quinoline ring, a benzothiophene ring, a
benzothiazole ring, a benzoimidazole ring, a benzofuran ring, cyclopropyl, cyclopentyl, cyclohexyl, cycloheptyl, an indene ring, a tetrahydronaphthalene ring, dihydro-1,4- benzodioxoxine ring, or a piperidine ring; each of which are optionally substituted with 1 , 2, 3, 4, 5, or 6 independently selected R5 groups;
each R5 is, independently, chloro, fluoro, bromo, cyano, methyl, ethyl,
trifluoromethyl, hydroxyl, methoxy, trifluoromethoxy, difluoromethoxy, phenoxy, dimethylamino, t-butylcarbonylamino, methoxycarbonyl, methylthio, phenyl, a pyridine ring, a thiazole ring, a quinoline ring, an isoquinoline ring, an imidazo[l,2-a]pyrimidine ring, a benzoxazole ring, or an oxadiazole ring; wherein said phenyl, pyridine ring, thiazole ring, quinoline ring, isoquinoline ring, imidazo[l,2-a]pyrimidine ring, benzoxazole ring, and oxadiazole ring are each optionally substituted with 1, 2, 3, 4, or 5 independently selected R6 groups; and
n is 0, 1, or 2.
43. The compound according to claim 1 or 2, or a pharmaceutically acceptable salt thereof, wherein:
X is N or CR2;
Y is N or CR3;
each R1 is, independently, Ci_4 alkyl; or two R1 groups form a 2-carbon bridge.
R2 is H, halo, or cyano;
R3 is H;
Z is cyano;
L is S(=0)2;
A is Ci_6 alkyl; and
n is 0, 1, or 2.
44. The compound according to claim 1 or 2, or a pharmaceutically acceptable salt thereof, wherein:
X is N or CH;
Y is N;
Z is cyano;
L is C(=0) or C(=0)NH;
A is phenyl or a pyridine ring, each of which is optionally substituted with 1, 2, 3, or 4 independently selected R5 groups;
each R5 is, independently, halo or Ci_6 haloalkyl; and
n is 0.
45. The compound according to claim 1 or 2, or a pharmaceutically acceptable salt thereof, wherein:
X is N or CH;
Y is N;
Z is cyano;
L is C(=0) or C(=0)NH;
A is phenyl or pyridin-4-yl, each of which is optionally substituted with 1 or 2 independently selected R5 groups;
each R5 is, independently, fluoro or trifluoromethyl; and
n is 0.
46. The compound according to claim 1 or 2, or a pharmaceutically acceptable salt thereof, wherein:
X is N or CR2;
Y is N or CR3;
each R1 is, independently, Ci_4 alkyl, hydroxyl, Ci_4 alkoxy, or fluoro;
two R1 groups together form a 2- or 3 -carbon bridge or a bridge of formula -CH2- 0-CH2-;
R2 is H, halo, or cyano;
R3 is H;
Z is cyano;
O
L is C(=0)NH, C(=0), S(=0)2, CH2, C(=0)CH2, Δ C(=0)0, or
C(=0)OCH2;
A is Ci_6 alkyl, phenyl, a naphthyl ring, monocyclic C3_i0 cycloalkyl, bicyclic C3_i0 cycloalkyl, monocyclic C2_io heterocycloalkyl, bicyclic C2_io heterocycloalkyl, monocyclic Ci_io heteroaryl, or bicyclic Ci_io heteroaryl; each of which is optionally substituted with 1, 2, 3, 4, or 5 independently selected R5 groups;
each R5 is, independently, halo, cyano, Ci_6 alkyl, Ci_6 haloalkyl, C6_io aryl, Ci_i0 heteroaryl, ORa, SRa, C(0)ORa, NRcRd, NRcC(0)Rb; wherein said Ci_6 alkyl, Ci_6 haloalkyl, C6-io aryl, and Ci_io heteroaryl are each optionally substituted with 1, 2, 3, 4, or 5 independently selected R6 groups;
n is 0, 1, or 2;
each R5 is, independently, halo, cyano, Ci_6 alkyl, Ci_6 haloalkyl, C6_io aryl, Ci_i0 heteroaryl, ORa, SRa, C(0)ORa, NRcRd, or NRcC(0)Rb; wherein said Ci_6 alkyl, Ci_6 haloalkyl, C6-io aryl, and Ci_io heteroaryl are each optionally substituted with 1, 2, 3, 4, or 5 independently selected R6 groups;
each Ra, Rb, Rc, and Rd is, independently, H, Ci_6 alkyl, Ci_6 haloalkyl, or C6_io aryl; wherein said Ci_6 alkyl and C6-io aryl is optionally substituted with 1, 2, 3, 4, or 5 independently selected Rg groups;
each R6 is, independently, halo, cyano, Ci_6 alkyl, Ci_6 haloalkyl, ORal, NRclRdl, or OC(0)Rbl; and each Ral, Rbl, Rcl, and Rdl is, independently, H or Ci_6 alkyl; wherein said Ci_6 alkyl is optionally substituted with a substituent independently selected from Ci_4 alkoxy and hydroxyl;
or any Rcl and Rdl together with the N atom to which they are attached form a 4-, 5-, 6-, or 7-membered heterocycloalkyl group, optionally substituted with 1, 2, 3 or 4 substituents independently selected from halo.
47. The compound according to claim 1, wherein the compound is selected from:
{ 1 - { 1 -[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl} -3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{l-[l-(3-fluoro-4-quinolin-6-ylbenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{l-[l-(3,5-difluorobenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl]- 1 -[ 1 -(3 ,4,5- trifluorobenzoyl)piperidin-4-yl]azetidin-3-yl} acetonitrile;
{l-[l-(3-fluoro-4-methoxybenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{l-[l-(3-fluoro-4-hydroxybenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[2-fluoro-3-(trifluoromethyl)benzoyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{l-[l-(cyclohexylcarbonyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-(l-benzoylpiperidin-4-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
2-[(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin- 1 -yl}piperidin-l -yl)carbonyl]benzonitrile;
3 - [(4- {3 -(cyanomethyl)-3 - [4-(7H-pyrrolo[2,3 -d]pyrimidin-4-yl)- 1 H-pyrazol- 1 - yl]azetidin- 1 -yl}piperidin-l -yl)carbonyl]benzonitrile; 4-[(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yljazetidin- 1 -yl}piperidin-l -yl)carbonyl]benzonitrile;
{ 1 - { 1 -[(6-chloropyridin-2-yl)carbonyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-[l-(pyrazin-2-lcarbonyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)- 1 H-pyrazol- 1 -yl]azetidin-3-l} acetonitrile;
{3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol-1 -yl]- 1 -[1 -(3- thienylcarbonyl)piperidin-4-yl]azetidin-3-yl} acetonitrile;
{l-[l-(l,3-oxazol-2-ylcarbonyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin- 4-yl)- 1 H-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{ 1 -( 1 - { [2-methyl-5-(trifluoromethyl)- 1 ,3 -oxazol-4-yl] carbonyl} piperidin-4-yl)-3 - [4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
3 - [(4- {3 -(cyanomethyl)-3 - [4-(7H-pyrrolo[2,3 -d]pyrimidin-4-yl)- 1 H-pyrazol- 1 - yl]azetidin- 1 -yl}piperidin-l -yl)carbonyl]-5-fluorobenzonitrile;
{l-[l-(3-chlorobenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -y 1] azetidin-3 -y 1 } acetonitrile;
{ 1 - [ 1 -(3 -bromobenzoyl)piperidin-4-yl] -3 - [4-(7H-pyrrolo[2,3 -d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -y 1] azetidin-3 -y 1 } acetonitrile;
(3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol- 1 -yl] - 1 - { 1 -[3- (trifluoromethoxy)benzoyl]piperidin-4-yl}azetidin-3-yl)acetonitrile;
(3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol- 1 -yl] - 1 - { 1 -[3- (trifluoromethyl)benzoyl]piperidin-4-yl}azetidin-3-yl)acetonitrile;
{ 1 - { 1 -[3-fluoro-5-(trifluoromethyl)benzoyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{l-[l-(3,5-dichlorobenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-[l-(3-fluorobenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -y 1] azetidin-3 -y 1 } acetonitrile;
{l-[l-(4-fluoro-3-methoxybenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile; {l-[l-(2-fluoro-5-methoxybenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{l-[l-(3-chloro-5-fluorobenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{ 1 - [ 1 -(3 -bromo-5-fluorobenzoyl)piperidin-4-yl] -3 - [4-(7H-pyrrolo[2,3 - d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[(2,5-dichloro-3-thienyl)carbonyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{ 1 - [ 1 -(3 -methoxybenzoyl)piperidin-4-yl] -3 - [4-(7H-pyrrolo[2,3 -d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -y 1] azetidin-3 -y 1 } acetonitrile;
{3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol- 1 -yl]- 1 -[1 -(2,4,5-trifluoro-3- methoxybenzoyl)piperidin-4-yl]azetidin-3-yl} acetonitrile;
{l-[l-(3,5-dimethoxybenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin- 4-yl)- 1 H-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{l-[l-(3-chloro-4-fluorobenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{l-[l-(3,4-difluorobenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-[l-(3-fluoro-5-methoxybenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{ 1 - [ 1 -(2-chloro-6-methoxy isonicotinoyl)piperidin-4-yl] -3 - [4-(7H-pyrrolo[2,3 - d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{l-[l-(5-fluoro-2-methoxybenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{l-[l-(2-fluoro-6-methoxybenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{l-[l-(4-fluoro-2-methoxybenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{l-[l-(2,3-difluorobenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile; {l-[l-(2,4-difluorobenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-[l-(2,5-difluorobenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-[l-(2,6-difluorobenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{ 1 - { 1 -[2-fluoro-6-(trifluoromethyl)benzoyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]-l-[l-(2,3,4- trifluorobenzoyl)piperidin-4-yl]azetidin-3-yl} acetonitrile;
{3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]-l-[l-(2,3,6- trifluorobenzoyl)piperidin-4-yl]azetidin-3-yl} acetonitrile;
{3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]-l-[l-(2,4,5- trifluorobenzoyl)piperidin-4-yl]azetidin-3-yl} acetonitrile;
{3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]-l-[l-(2,4,6- trifluorobenzoyl)piperidin-4-yl]azetidin-3-yl} acetonitrile;
{l-[l-(3,5-dibromo-4-methoxybenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
3 - [(4- {3 -(cyanomethyl)-3 - [4-(7H-pyrrolo[2,3 -d]pyrimidin-4-yl)- 1 H-pyrazol- 1 - yl]azetidin-l-yl}piperidin-l-yl)carbonyl]-6-(dimethylamino)-2-fluorobenzonitrile;
{ 1 - { 1 -[3-fluoro-4-(trifluoromethyl)benzoyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{ 1 -( 1 - { [4-chloro-6-(trifluoromethyl)pyridin-2-yl] carbonyl} piperidin-4-yl)-3 - [4- (7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]-l-[l-(2,3,4,5- tetrafluorobenzoyl)piperidin-4-yl]azetidin-3-yl} acetonitrile;
5-[(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin- 1 -yl} piperidin- 1 -yl)carbonyl] -2-methoxybenzonitrile;
{3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]-l-[l-(2,3,5,6- tetrafluorobenzoyl)piperidin-4-yl]azetidin-3-yl} acetonitrile; (3 -[4-(7H-pyrrolo [2,3 -d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] - 1 - { 1 - [2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl}azetidin-3-yl)acetonitrile;
{l-[l-(4-fluoro-3-hydroxybenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
5-[(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin- 1 -yl} piperidin- 1 -yl)carbonyl] -2-(dimethylamino)benzonitrile;
{ 1 - { 1 -[4-(dimethylamino)-2,3,5,6-tetrafluorobenzoyl]piperidin-4-yl} -3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-[l-(3,5-difluoroisonicotinoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[3-fluoro-4-(methylthio)benzoyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{l-[l-(4-chloro-3-fluorobenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{l-[l-(3-fluoro-4-methylbenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{l-[l-(2,5-dimethyl-3-furoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
4-[(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin- 1 -yl} piperidin- 1 -yl)carbonyl] -2-fluorobenzonitrile;
{l-[l-(2-fluorobenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -y 1] azetidin-3 -y 1 } acetonitrile;
{l-[l-(4-fluorobenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-[l-(2-thienylcarbonyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{ 1 - { 1 -[3-methoxy-5-(trifluoromethyl)-2-thienylcarbonyl]piperidin-4-yl} -3-[4- (7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[3-hydroxy-5-(trifluoromethyl)-2-thienylcarbonyl]piperidin-4-yl} -3-[4- (7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile; { 1 - { 1 -[(4-methoxy-3-thienyl)carbonyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[(5-methyl-3-thienyl)carbonyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[(5-chloro-4-methoxy-3-thienyl)carbonyl]piperidin-4-yl} -3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[(2-bromo-3-thienyl)carbonyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[(3-chloro-2-thienyl)carbonyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[(5-chloro-2-thienyl)carbonyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[(3-methyl-2-thienyl)carbonyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[(4-methyl-2-thienyl)carbonyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[(5-methyl-2-thienyl)carbonyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[(3-methoxy-2-thienyl)carbonyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[2-fluoro-4-(trifluoromethyl)benzoyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
4- [(4- {3 -(cyanomethyl)-3 - [4-(7H-pyrrolo[2 ,3 -d]pyrimidin-4-yl)- 1 H-pyrazol- 1 - yl]azetidin-l-yl}piperidin-l-yl)carbonyl]-3,5-difluorobenzonitrile;
{l-[l-(3-chloro-4-hydroxybenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
[3 -[4-(7H-pyrrolo[2,3 -d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] - 1 -( 1 - { [2- (trifluoromethyl)pyrimidin-4-yl]carbonyl}piperidin-4-yl)azetidin-3-yl]acetonitrile;
[3 -[4-(7H-pyrrolo[2,3 -d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] - 1 -( 1 - { [6- (trifluoromethyl)pyrazin-2-yl]carbonyl}piperidin-4-yl)azetidin-3-yl]acetonitrile; { 1 - [ 1 -( 1 -naphthoyl)piperidin-4-yl] -3 - [4-(7H-pyrrolo[2,3 -d]pyrimidin-4-yl)- 1 H- pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol-1 -yl]- 1 -[1 -(quinolin-3- ylcarbonyl)piperidin-4-yl]azetidin-3-yl} acetonitrile;
{3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol-1 -yl]- 1 -[1 -(quinolin-6- ylcarbonyl)piperidin-4-yl]azetidin-3-yl} acetonitrile;
{l-[l-(l-benzothien-2-ylcarbonyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[(3-chloro-6-fluoro-l -benzothien-2-yl)carbonyl]piperidin-4-yl} -3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[(3-chloro-4-fluoro-l -benzothien-2-yl)carbonyl]piperidin-4-yl} -3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
[3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl]- 1 -( 1 - { [4- (trifluoromethyl)-l-benzothien-2-yl]carbonyl}piperidin-4-yl)azetidin-3-yl]acetonitrile;
[3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl]- 1 -( 1 - { [6- (trifluoromethyl)-l-benzothien-2-yl]carbonyl}piperidin-4-yl)azetidin-3-yl]acetonitrile;
[3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl]- 1 -( 1 - { [7- (trifluoromethyl)-l-benzothien-2-yl]carbonyl}piperidin-4-yl)azetidin-3-yl]acetonitrile;
{l-[l-(l-benzothien-3-ylcarbonyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]-l-[l-(l ,2,3,4- tetrahydronaphthalen-2-ylcarbonyl)piperidin-4-yl]azetidin-3-yl} acetonitrile;
[3 -[4-(7H-pyrrolo[2,3 -d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] - 1 -( 1 - { [4- (trifluoromethyl)cyclohexyl]carbonyl}piperidin-4-yl)azetidin-3-yl]acetonitrile;
{l-[l-(2,3-dihydro-lH-inden-2-ylcarbonyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[(4,4-difluorocyclohexyl)carbonyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{l-[l-(cyclopentylcarbonyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile; { 1 - [ 1 -(cycloheptylcarbonyl)piperidin-4-yl] -3 - [4-(7H-pyrrolo[2,3 -d]pyrimidin-4- yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{ 1 - { 1 -[(3-methoxycyclohexyl)carbonyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[(4-phenylcyclohexyl)carbonyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{ 1 -(1 - {[4-(4-chlorophenyl)cyclohexyl]carbonyl}piperidin-4-yl)-3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
6-{4-[(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yljazetidin- 1 -yl}piperidin-l -yl)carbonyl]piperidin- 1 -yl}nicotinonitrile;
{ 1 -(1 - {[ 1 -(5-chloro-3-fluoropyridin-2-yl)piperidin-4-yl]carbonyl}piperidin-4-yl)- 3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
2-{4-[(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin- 1 -yl} piperidin- 1 -yl)carbonyl]piperidin- 1 -yl} -6-methylnicotinonitrile;
{l-[l-(phenylacetyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH- pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[(1 -phenylcyclopropyl)carbonyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{ 1 -(1 - {[ 1 -(4-chlorophenyl)cyclopropyl]carbonyl}piperidin-4-yl)-3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{ 1 - { 1 -[(2,6-dichlorophenyl)acetyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{l-[l-(mesitylacetyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH- pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{l-[l-(biphenyl-4-ylcarbonyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-[l-(3-fluoro-4-isoquinolin-6-ylbenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{l-[l-(2,6-difluoro-4-pyridin-3-ylbenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile; {l-[l-(3-fluoro-4-pyridin-4-ylbenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
4'-[(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin- 1 -yl} piperidin- 1 -yl)carbonyl] -2'-fluorobiphenyl-4-carbonitrile;
4'-[(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-l-yl}piperidin-l-yl)carbonyl]-2',3-difluorobiphenyl-4-carbonitrile;
{l-[l-(2-fluoro-4-pyridin-3-ylbenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-{l-[4-fluoro-3-(l,3-thiazol-2-yl)benzoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{l-{l-[3-fluoro-4-(l,3-thiazol-2-yl)benzoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{ 1 - [ 1 -(3 -fluoro-4-pyridin-3 -ylbenzoyl)piperidin-4-yl] -3 -[4-(7H-pyrrolo[2,3 - d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
4'-[(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin- 1 -yl} piperidin- 1 -yl)carbonyl] -2'-fluorobiphenyl-2-carbonitrile;
4'-[(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin- 1 -yl} piperidin- 1 -yl)carbonyl] -2'-fluorobiphenyl-3 -carbonitrile;
4'-[(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin- 1 -yl} piperidin- 1 -yl)carbonyl]biphenyl-4-carbonitrile;
(3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]-l-{l-[(2,3,,4,- trifluorobiphenyl-4-yl)carbonyl]piperidin-4-yl}azetidin-3-yl)acetonitrile;
4'-[(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-l-yl}piperidin-l-yl)carbonyl]-2',5-difluorobiphenyl-3-carbonitrile;
{l-[l-(3-fluoro-4-quinolin-5-ylbenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{l-[l-(3-fluoro-4-isoquinolin-5-ylbenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{l-[l-(3-fluoro-4-isoquinolin-8-ylbenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile; {l-[l-(3-fluoro-4-quinolin-8-ylbenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-[l-(3-fluoro-4-isoquinolin-7-ylbenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{l-[l-(3-fluoro-4-quinolin-7-ylbenzoyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{ 1 -[1 -(3-fluoro-4-imidazo[ 1 ,2-a]pyridin-6-ylbenzoyl)piperidin-4-yl]-3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{l-{l-[4-(l,3-benzoxazol-2-yl)benzoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[4-(l ,3-benzoxazol-2-yl)-3-fluorobenzoyl]piperidin-4-yl} -3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
3 - [(3 - {3 -(cyanomethyl)-3 - [4-(7H-pyrrolo[2,3 -d]pyrimidin-4-yl)- 1 H-pyrazol- 1 - yl]azetidin- 1-yl} -8-azabicyclo[3.2.1 ]oct-8-yl)carbonyl]-5-fluorobenzonitrile;
{l-[8-(3,4-difluorobenzoyl)-8-azabicyclo[3.2.1]oct-3-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
4- [(3 - {3 -(cyanomethyl)-3 - [4-(7H-pyrrolo[2,3 -d]pyrimidin-4-yl)- 1 H-pyrazol- 1 - yl]azetidin- 1-yl} -8-azabicyclo[3.2.1 ]oct-8-yl)carbonyl]-2-fluorobenzonitrile;
{l-[8-(4-chloro-3-fluorobenzoyl)-8-azabicyclo[3.2.1]oct-3-yl]-3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{ 1 - {8-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]-8-azabicyclo[3.2.1 ]oct-3-yl} -3- [4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
[3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl]- 1 -(8- { [6- (trifluoromethyl)pyridin-3-yl]carbonyl} -8-azabicyclo[3.2.1 ]oct-3-yl)azetidin-3- yl] acetonitrile;
(3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]-l-{8-[2- (trifluoromethyl)isonicotinoyl]-8-azabicyclo[3.2.1 ]oct-3-yl} azetidin-3-yl)acetonitrile;
{l-[8-(cyclopentylcarbonyl)-8-azabicyclo[3.2.1]oct-3-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
(3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol-1 -yl]- 1 -[8-(tetrahydro-2H- pyran-4-ylcarbonyl)-8-azabicyclo[3.2.1 ]oct-3-yl]azetidin-3-yl} acetonitrile; {l-[8-(cyclohexylcarbonyl)-8-azabicyclo[3.2.1]oct-3-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-{8-[(4,4-difluorocyclohexyl)carbonyl]-8-azabicyclo[3.2.1]oct-3-yl}-3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-[l-(3-fluorobenzoyl)-2-methylpiperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{l-[l-(3-fluorobenzoyl)-2-methylpiperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[(4,4-difluorocyclohexyl)carbonyl]-2-methylpiperidin-4-yl} -3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-[l-(3-fluorobenzoyl)-4-methylpiperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{l-[l-(cyclohexylcarbonyl)-4-methylpiperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin- 1 -yl} -N-[4-fluoro-2-(trifluoromethyl)phenyl]piperidine- 1 -carboxamide;
4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin- 1 -yl} -N- [3 -(trifluoromethyl)pyridin-4-yl]piperidine-l -carboxamide;
4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin- 1 -yl} -N-(2,6-difluorophenyl)piperidine- 1 -carboxamide;
4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin- 1 -yl} -N-[2-(trifluoromethyl)phenyl]piperidine- 1 -carboxamide;
4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin- 1 -yl} -N-[2-(trifluoromethoxy)phenyl]piperidine- 1 -carboxamide;
N-(4-bromo-3-thienyl)-4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)- 1 H-pyrazol- 1 -yl] azetidin- 1 -y 1} piperidine- 1 -carboxamide;
4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin- 1 -yl} -N-(2,6-dichlorophenyl)piperidine- 1 -carboxamide;
N-(2-chloro-6-methylphenyl)-4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl]azetidin- 1 -yl}piperidine- 1 -carboxamide; N-(2-chloro-4-fluorophenyl)-4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yljazetidin- 1 -yl}piperidine- 1 -carboxamide;
N-(2-chlorophenyl)-4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yljazetidin- 1 -yl}piperidine- 1 -carboxamide;
4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yljazetidin- 1 -yl} -N-[2-(difluoromethoxy)phenyl]piperidine- 1 -carboxamide;
4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-l-yl}-N-[4-(trifluoromethyl)pyridin-3-yl]piperidine-l -carboxamide;
4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-l -yl} -N-[3-(trifluoromethyl)phenyl]piperidine- 1 -carboxamide;
4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yljazetidin- 1 -yl} -N-[4-(trifluoromethyl)phenyl]piperidine- 1 -carboxamide;
N-(5-chloro-2-methylphenyl)-4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yljazetidin- 1 -yl}piperidine- 1 -carboxamide;
4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yljazetidin- 1 -yl} -N-(2-fluorophenyl)piperidine- 1 -carboxamide;
4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-l-yl}-N-[2-fluoro-3-(trifluoromethyl)phenyl]piperidine-l -carboxamide;
4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yljazetidin- 1 -yl} -N-(2,4-difluorophenyl)piperidine- 1 -carboxamide;
4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yljazetidin- 1 -yl} -N-(2,3 ,4-trifluorophenyl)piperidine- 1 -carboxamide;
4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yljazetidin- 1 -yl} -N-(2,3 ,5-trifluorophenyl)piperidine- 1 -carboxamide;
4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yljazetidin- 1 -yl} -N-(2,5-difluorophenyl)piperidine- 1 -carboxamide;
4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yljazetidin- 1 -yl} -N-(3 ,5-difluorophenyl)piperidine- 1 -carboxamide;
4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yljazetidin- 1 -yl} -N-(3 ,4-difluorophenyl)piperidine- 1 -carboxamide; N-(3-chloro-2-fluorophenyl)-4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yljazetidin- 1 -yl}piperidine- 1 -carboxamide;
N-(4-chloro-2-fluorophenyl)-4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yljazetidin- 1 -yl}piperidine- 1 -carboxamide;
4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yljazetidin- 1 -yl} -N-3-thienylpiperidine- 1 -carboxamide;
4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yljazetidin- 1 -yl} -N-(2-methoxyphenyl)piperidine- 1 -carboxamide;
4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin- 1 -yl} -N-(3 -methoxyphenyl)piperidine- 1 -carboxamide;
4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yljazetidin- 1 -yl} -N-[4-(trifluoromethoxy)phenyl]piperidine- 1 -carboxamide;
N-(3-chlorophenyl)-4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yljazetidin- 1 -yl}piperidine- 1 -carboxamide;
N-(4-chlorophenyl)-4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yljazetidin- 1 -yl}piperidine- 1 -carboxamide;
4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yljazetidin- 1 -yl} -N-(2-methylphenyl)piperidine- 1 -carboxamide;
4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yljazetidin- 1 -yl} -N-(2,5-dimethoxyphenyl)piperidine- 1 -carboxamide;
4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yljazetidin- 1 -yl} -N-(4-fluoro-2-methylphenyl)piperidine- 1 -carboxamide;
4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yljazetidin- 1 -yl} -N-[6-(trifluoromethyl)pyridin-2-yl]piperidine- 1 -carboxamide;
4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yljazetidin- 1 -yl} -N-(2,6-dimethylpyridin-3-yl)piperidine- 1 -carboxamide;
4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin- 1 -yl} -N- 1 ,3 -thiazol-2 -yip iperidine-1 -carboxamide;
4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yljazetidin- 1 -yl} -N-(4-methyl-l ,3-thiazol-2-yl)piperidine- 1 -carboxamide; 4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-l-yl}-N-(4,5-dimethyl-l,3-thiazol-2-yl)piperidine-l -carboxamide;
N- 1 ,3-benzothiazol-2-yl-4- {3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin- 4-yl)- 1 H-pyrazol- 1 -yl] azetidin- 1 -yl} piperidine- 1 -carboxamide;
4- {3 -(cyanomethyl)-3 - [4-(7H-pyrrolo[2 ,3 -d]pyrimidin-4-yl)- 1 H-pyrazol- 1 - yl] azetidin- 1 -yl} -N-( 1 -methyl- 1 H-benzimidazol-2-yl)piperidine- 1 -carboxamide;
N-[4-(4-chlorophenyl)-l,3-thiazol-2-yl]-4-{3-(cyanomethyl)-3-[4-(7H- pyrrolo [2, 3 -d]pyrimidin-4-yl)-l H-pyrazol- 1 -yl] azetidin- 1 -yl} piperidine- 1 -carboxamide;
4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin- 1 -yl} -N-( 1 -ethyl- 1 H-pyrazol-5-yl)piperidine- 1 -carboxamide;
4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin- 1 -yl} -N-( 1 ,3-dimethyl- 1 H-pyrazol-5-yl)piperidine- 1 -carboxamide;
4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin- 1 -yl} -N-(2-methylpyridin-3-yl)piperidine- 1 -carboxamide;
4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin- 1 -yl} -N-(6-fluoro-2-methylpyridin-3-yl)piperidine- 1 -carboxamide;
4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin- 1 -yl} -N-(2-fluoro-6-methylpyridin-3-yl)piperidine- 1 -carboxamide;
4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin- 1 -yl} -N- [3 -(trifluoromethyl)pyridin-2-yl]piperidine-l -carboxamide;
4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin- 1 -yl} -N-(3-fluoropyridin-2-yl)piperidine- 1 -carboxamide;
4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin- 1 -yl} -N-(3 ,5-difluoropyridin-2-yl)piperidine- 1 -carboxamide;
4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin- 1 -yl} -N-(2-methoxypyridin-3-yl)piperidine- 1 -carboxamide;
4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin- 1-yl} -N-[2-methyl-6-(trifluoromethyl)pyridin-3-yl]piperidine- 1 -carboxamide; methyl 2-{[(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH- pyrazol- 1 -yl] azetidin- 1 -yl} piperidin- 1 -yl)carbonyl] amino} benzoate; methyl 2-{[(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH- pyrazol- 1 -yljazetidin- 1 -yl}piperidin- l-yl)carbonyl]amino} -5-fluorobenzoate;
methyl 4-{[(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH- pyrazol- 1 -yljazetidin- 1 -yl}piperidin- l-yl)carbonyl]amino} -3-fluorobenzoate;
3 - [(4- {3 -(cyanomethyl)-3 - [4-(7H-pyrrolo[2,3 -d]pyrimidin-4-yl)- 1 H-pyrazol- 1 - yl] azetidin- 1 -yl} piperidin- 1 -yl)methyl] -6-(dimethylamino)-2-fluorobenzonitrile;
{l-[l-(3,5-dichlorobenzyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{ 1 - { 1 -[2-chloro-5-(trifluoromethyl)benzyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[2-fluoro-3-(trifluoromethyl)benzyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[2-fluoro-6-(trifluoromethyl)benzyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[(2-chloroquinolin-3-yl)methyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{l-[l-(3,5-difluorobenzyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -y 1] azetidin-3 -y 1 } acetonitrile;
{ 1 - { 1 -[2-fluoro-4-(trifluoromethyl)benzyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{ 1 -[ 1 -(2,4-difluorobenzyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-[l-(2-fluoro-6-methoxybenzyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{l-[l-(2,3-dichlorobenzyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
5-[(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin- 1 -yl} piperidin- 1 -yl)methyl] -2-fluorobenzonitrile;
{l-{l-[4-(l,2,3-oxadiazol-4-yl)benzyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile; 2-[(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin- 1 -yl} piperidin- 1 -yl)methyl]benzonitrile;
3 - [(4- {3 -(cyanomethyl)-3 - [4-(7H-pyrrolo[2,3 -d]pyrimidin-4-yl)- 1 H-pyrazol- 1 - yl] azetidin- 1 -yl} piperidin- 1 -yl)methyl]benzonitrile;
6-[(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin- 1 -yl} piperidin- 1 -yl)methyl] -2-methoxynicotinonitrile;
{ 1 - { 1 -[(2,6-dibromopyridin-4-yl)methyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile;
{ 1 - { 1 -[(2-bromopyridin-4-yl)methyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile;
{l-[l-(2-chloro-6-fluorobenzyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin- 4-yl)- 1 H-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{l-[l-(3-chloro-2,6-difluorobenzyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
4-[(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin- 1 -yl} piperidin- 1 -yl)methyl] -2-fluorobenzonitrile;
{ 1 - { 1 -[(5-methyl-3-phenylisoxazol-4-yl)methyl]piperidin-4-yl} -3 -[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{ 1 -( 1 - { [3 -fluoro-2-(trifluoromethyl)pyridin-4-yl]methyl} piperidin-4-yl)-3 -[4- (7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{l-[l-(l-benzofuran-2-ylmethyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{l-[l-(3-phenoxybenzyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -y 1] azetidin-3 -y 1 } acetonitrile;
{ 1 -[1 -(2,3-dihydro- 1 ,4-benzodioxin-6-ylmethyl)piperidin-4-yl]-3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
N-{4-[(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol- 1 -yl]azetidin- 1 -yl}piperidin- 1 -yl)methyl]pyridin-2-yl} -2,2-dimethylpropanamide;
{ 1 - { 1 -[3-chloro-2-fluoro-6-(trifluoromethyl)benzyl]piperidin-4-yl} -3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile; { 1 - { 1 -[(3,5-dichloropyridin-4-yl)methyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{ 1 - { 1 -[(2-chloro-6-methoxyquinolin-3-yl)methyl]piperidin-4-yl} -3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-[l-(2-chloro-3,4-dimethoxybenzyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
3 - [(3 - {3 -(cyanomethyl)-3 - [4-(7H-pyrrolo[2,3 -d]pyrimidin-4-yl)- 1 H-pyrazol- 1 - yljazetidin- 1-yl} -8-azabicyclo[3.2.1 ]oct-8-yl)methyl]-6-(dimethylamino)-2- fluorobenzonitrile;
{l-[8-(2-chloro-3,6-difluorobenzyl)-8-azabicyclo[3.2.1]oct-3-yl]-3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
3 - [(4- {3 -(cyanomethyl)-3 - [4-(7H-pyrrolo[2,3 -d]pyrimidin-4-yl)- 1 H-pyrazol- 1 - yljazetidin- 1 -yl} -2-methylpiperidin- 1 -yl)methyl]-6-(dimethylamino)-2- fluorobenzonitrile;
3 - [(4- {3 -(cyanomethyl)-3 - [4-(7H-pyrrolo[2,3 -d]pyrimidin-4-yl)- 1 H-pyrazol- 1 - yljazetidin- 1 -yl} -2-methylpiperidin- 1 -yl)methyl]-6-(dimethylamino)-2- fluorobenzonitrile;
{l-[l-(2-chloro-6-fluorobenzyl)-2-methylpiperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{l-[l-(2-chloro-6-fluorobenzyl)-2-methylpiperidin-4-yl]-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{1-{1-[(1 -methyl- 1 H-pyrazol-5-yl)sulfonyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
2-[(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yljazetidin- 1 -yl}piperidin-l -yl)sulfonyl]benzonitrile;
3 - [(4- {3 -(cyanomethyl)-3 - [4-(7H-pyrrolo[2,3 -d]pyrimidin-4-yl)- 1 H-pyrazol- 1 - yljazetidin- 1 -yl}piperidin-l -yl)sulfonyl]benzonitrile;
4- [(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yljazetidin- 1 -yl}piperidin-l -yl)sulfonyl]benzonitrile;
5- [(4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl] azetidin- 1 -yl} piperidin- 1 -yl)sulfonyl] -2-(dimethylamino)benzonitrile; { 1 - { 1 -[(1 -methyl- lH-pyrazol-3-yl)sulfonyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile;
{l-[l-(cyclohexylsulfonyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-[l-(cyclopentylsulfonyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-[l-(methylsulfonyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -y 1] azetidin-3 -y 1 } acetonitrile;
{ 1 - [ 1 -(ethylsulfonyl)piperidin-4-yl] -3 - [4-(7H-pyrrolo[2,3 -d]pyrimidin-4-yl)- 1 H- pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{l-[l-(cyclopropylsulfonyl)piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)- 1 H-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{ 1 - [ 1 -(isopropylsulfonyl)piperidin-4-yl] -3 - [4-(7H-pyrrolo[2,3 -d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -y 1] azetidin-3 -y 1 } acetonitrile;
{l-{l-[(l-methyl-lH-imidazol-4-yl)sulfonyl]piperidin-4-yl}-3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{l-{l-[(l,2-dimethyl-lH-imidazol-4-yl)sulfonyl]piperidin-4-yl}-3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 -yl] azetidin-3 -yl} acetonitrile;
{ 1 - { 1 -[(3,5-dimethylisoxazol-4-yl)sulfonyl]piperidin-4-yl} -3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl} -3-[3-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrrol- 1 -yl]azetidin-3-yl} acetonitrile;
3 - [(4- { 3 -(cy anomethy l)-3 - [3 -(7H-pyrrolo [2 , 3 -d]pyrimidin-4-y 1)- 1 H-pyrrol- 1 - yl]azetidin- 1 -yl}piperidin-l -yl)carbonyl]benzonitrile;
3 - [(4- { 3 -(cy anomethy l)-3 - [3 -(7H-pyrrolo [2 , 3 -d]pyrimidin-4-y 1)- 1 H-pyrrol- 1 - yl]azetidin- 1 -yl}piperidin-l -yl)carbonyl]-5-fluorobenzonitrile;
4-[(4-{3-(cyanomethyl)-3-[3-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrrol-l- yl] azetidin- 1 -yl} piperidin- 1 -yl)carbonyl] -3 -fluorobenzonitrile;
4-[(4-{3-(cyanomethyl)-3-[3-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrrol-l- yl]azetidin-l-yl}piperidin-l-yl)carbonyl]-3,5-difluorobenzonitrile; { 1 - { 1 -[5-fluoro-2-(trifluoromethyl)benzoyl]piperidin-4-yl} -3-[3-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrrol- 1 -yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[3-fluoro-4-(trifluoromethyl)benzoyl]piperidin-4-yl} -3-[3-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrrol- 1 -yl]azetidin-3-yl} acetonitrile;
(3-[3-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrrol- 1 -yl]- 1 - { 1 -[3- (trifluoromethyl)benzoyl]piperidin-4-yl}azetidin-3-yl)acetonitrile;
{ 1 - { 1 -[2-fluoro-5-(trifluoromethoxy)benzoyl]piperidin-4-yl} -3-[3-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)-l H-pyrrol- 1 -yl]azetidin-3-yl} acetonitrile;
{3-[3-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrrol-l-yl]-l-[l-(2,3,6- trifluorobenzoyl)piperidin-4-yl]azetidin-3-yl} acetonitrile;
{3-[3-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrrol- 1 -yl]-l -[ 1 -(2- thienylcarbonyl)piperidin-4-yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[2-fluoro-4-(trifluoromethyl)benzoyl]piperidin-4-yl} -3-[3-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrrol- 1 -yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[2-fluoro-5-(trifluoromethyl)benzoyl]piperidin-4-yl} -3-[3-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrrol- 1 -yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[3-fluoro-5-(trifluoromethyl)benzoyl]piperidin-4-yl} -3-[3-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrrol- 1 -yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[4-fluoro-3-(trifluoromethyl)benzoyl]piperidin-4-yl} -3-[3-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrrol- 1 -yl]azetidin-3-yl} acetonitrile;
{l-[l-(2,3-difluorobenzoyl)piperidin-4-yl]-3-[3-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)- 1 H-pyrrol- 1 -yl]azetidin-3-yl} acetonitrile;
{l-[l-(3,4-difluorobenzoyl)piperidin-4-yl]-3-[3-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)- 1 H-pyrrol- 1 -yl]azetidin-3-yl} acetonitrile;
{l-[l-(2,5-difluorobenzoyl)piperidin-4-yl]-3-[3-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)- 1 H-pyrrol- 1 -yl]azetidin-3-yl} acetonitrile;
{l-[l-(2,6-difluorobenzoyl)piperidin-4-yl]-3-[3-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)- 1 H-pyrrol- 1 -yl]azetidin-3-yl} acetonitrile;
{3-[3-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrrol- 1 -yl]-l -[ 1 -(2,3,4- trifluorobenzoyl)piperidin-4-yl]azetidin-3-yl} acetonitrile; { 1 - { 1 -[2-fluoro-3-(trifluoromethoxy)benzoyl]piperidin-4-yl} -3-[3-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)-l H-pyrrol- 1 -yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[4-hydroxy-3-(trifluoromethyl)benzoyl]piperidin-4-yl} -3-[3-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrrol- 1 -yl]azetidin-3-yl} acetonitrile;
4-{3-(cyanomethyl)-3-[3-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrrol-l- yljazetidin- 1 -yl} -N-[4-fluoro-2-(trifluoromethyl)phenyl]piperidine- 1 -carboxamide;
4-{3-(cyanomethyl)-3-[3-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrrol-l- yl]azetidin-l -yl} -N-(2-methoxypyridin-3-yl)piperidine- 1 -carboxamide;
4-{3-(cyanomethyl)-3-[3-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrrol-l- yljazetidin- 1-yl} -N-[2-methyl-6-(trifluoromethyl)pyridin-3-yl]piperidine- 1 -carboxamide;
4-{3-(cyanomethyl)-3-[3-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrrol-l- yljazetidin- 1 -yl} -N-(2,4-difluorophenyl)piperidine- 1 -carboxamide;
4-{3-(cyanomethyl)-3-[3-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrrol-l- yljazetidin- 1 -yl} -N-(2-cyanophenyl)piperidine- 1 -carboxamide;
4-{3-(cyanomethyl)-3-[3-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrrol-l- yljazetidin- 1 -yl} -N-(2-methoxyphenyl)piperidine- 1 -carboxamide;
N-(2-chloro-4-fluorophenyl)-4-{3-(cyanomethyl)-3-[3-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrrol- 1 -yljazetidin- 1 -yl}piperidine- 1 -carboxamide;
4-{3-(cyanomethyl)-3-[3-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrrol-l- yl]azetidin-l-yl}-N-[3-(trifluoromethyl)pyridin-2-yl]piperidine-l -carboxamide;
4-{3-(cyanomethyl)-3-[3-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrrol-l- yljazetidin- 1 -yl} -N-[4-(trifluoromethyl)pyridin-3-yl]piperidine-l -carboxamide;
4-{3-(cyanomethyl)-3-[3-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrrol-l- yljazetidin- 1 -yl} -N-(3-fluoropyridin-2-yl)piperidine- 1 -carboxamide;
N-(4-chloro-2-cyanophenyl)-4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yljazetidin- 1 -yl}piperidine- 1 -carboxamide;
{ 1 - { 1 -[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl} -3-[4-(lH- pyrrolo[2,3-b]pyridin-4-yl)- lH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
4-[(4-{3-(cyanomethyl)-3-[4-(lH-pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol-l- yl] azetidin- 1 -yl} piperidin- 1 -yl)carbonyl] -3 -fluorobenzonitrile; {l-[l-(3-fluoro-4-hydroxybenzoyl)piperidin-4-yl]-3-[4-(lH-pyrrolo[2,3- b]pyridin-4-yl)- 1 H-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{l-[l-(2-fluoro-4-hydroxybenzoyl)piperidin-4-yl]-3-[4-(lH-pyrrolo[2,3- b]pyridin-4-yl)- 1 H-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[5-chloro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl} -3-[4-( 1 H- pyrrolo[2,3-b]pyridin-4-yl)- lH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
[3-[4-(lH-pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol-l-yl]-l-(l-{[2- (trifluoromethyl)pyrimidin-4-yl]carbonyl}piperidin-4-yl)azetidin-3-yl]acetonitrile;
[3-[4-(lH-pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol-l-yl]-l-(l-{[6- (trifluoromethyl)pyrazin-2-yl]carbonyl}piperidin-4-yl)azetidin-3-yl]acetonitrile;
[3-[4-(lH-pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol-l-yl]-l-(l-{[5- (trifluoromethyl)pyrazin-2-yl]carbonyl}piperidin-4-yl)azetidin-3-yl]acetonitrile;
{ 1 - { 1 -[(4,4-difluorocyclohexyl)carbonyl]piperidin-4-yl} -3-[4-(lH-pyrrolo[2,3- b]pyridin-4-yl)- 1 H-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
4-{3-(cyanomethyl)-3-[4-(lH-pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol-l- yljazetidin- 1 -yl} -N-[4-fluoro-2-(trifluoromethyl)phenyl]piperidine- 1 -carboxamide;
4-{3-(cyanomethyl)-3-[4-(lH-pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol-l- yljazetidin- 1 -yl} -N-(2,4-difluorophenyl)piperidine- 1 -carboxamide;
N-(2-chloro-4-fluorophenyl)-4-{3-(cyanomethyl)-3-[4-(lH-pyrrolo[2,3-b]pyridin- 4-yl)- 1 H-pyrazol- 1 -yl] azetidin- 1 -yl} piperidine- 1 -carboxamide;
4-{3-(cyanomethyl)-3-[4-(lH-pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol-l- yljazetidin- 1 -yl} -N-(2-methoxypyridin-3-yl)piperidine- 1 -carboxamide;
4-{3-(cyanomethyl)-3-[4-(lH-pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol-l- yl]azetidin-l-yl}-N-[3-(trifluoromethyl)pyridin-4-yl]piperidine-l -carboxamide;
4-{3-(cyanomethyl)-3-[4-(lH-pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol-l- yl]azetidin-l-yl}-N-[4-(trifluoromethyl)pyridin-3-yl]piperidine-l -carboxamide;
4-{3-(cyanomethyl)-3-[4-(lH-pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol-l- yl] azetidin- 1 -yl} -N-(2-fluorophenyl)piperidine- 1 -carboxamide;
N-(2-chlorophenyl)-4-{3-(cyanomethyl)-3-[4-(lH-pyrrolo[2,3-b]pyridin-4-yl)- 1 H-pyrazol- 1 -yljazetidin- 1 -yl}piperidine- 1 -carboxamide; 4- {3-(cyanomethyl)-3-[4-(lH-pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol-l- yl] azetidin- 1-yl} -N-[4-cyano-2-(trifluoromethyl)phenyl]piperidine- 1 -carboxamide;
N-(4-cyano-2-fluorophenyl)-4-{3-(cyanomethyl)-3-[4-(lH-pyrrolo[2,3-b]pyridin- 4-yl)- 1 H-pyrazol- 1 -yl] azetidin- 1 -yl} piperidine- 1 -carboxamide;
N-(2-chloro-4-cyanophenyl)-4-{3-(cyanomethyl)-3-[4-(lH-pyrrolo[2,3-b]pyridin- 4-yl)- 1 H-pyrazol- 1 -yl] azetidin- 1 -yl} piperidine- 1 -carboxamide;
(3-[4-(5-fluoro- lH-pyrrolo[2,3-b]pyridin-4-yl)- IH-pyrazol- 1 -yl]-l - { 1 -[3-fluoro- 2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}azetidin-3-yl)acetonitrile;
5- [(4-{3-(cyanomethyl)-3-[4-(5-fluoro-lH-pyrrolo[2,3-b]pyridin-4-yl)-lH- pyrazol- 1 -yljazetidin- 1 -yl}piperidin- 1 -yl)carbonyl]isophthalonitrile;
3- [(4-{3-(cyanomethyl)-3-[4-(5-fluoro-lH-pyrrolo[2,3-b]pyridin-4-yl)-lH- pyrazol- 1 -yljazetidin- 1 -yl}piperidin- 1 -yl)carbonyl]-5-fluorobenzonitrile;
4- [(4-{3-(cyanomethyl)-3-[4-(5-fluoro-lH-pyrrolo[2,3-b]pyridin-4-yl)-lH- pyrazol- 1 -yljazetidin- 1 -yl}piperidin- 1 -yl)carbonyl]-3-fluorobenzonitrile;
5- [(4-{3-(cyanomethyl)-3-[4-(5-fluoro-lH-pyrrolo[2,3-b]pyridin-4-yl)-lH- pyrazol- 1 -yljazetidin- 1 -yl}piperidin- 1 -yl)carbonyl]-2-fluorobenzonitrile;
{ 1 - { 1 -[(5-fluoropyridin-2-yl)carbonyl]piperidin-4-yl} -3-[4-(5-fluoro- 1 H- pyrrolo[2,3-b]pyridin-4-yl)- IH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{l-[l-(3-fluoroisonicotinoyl)piperidin-4-yl]-3-[4-(5-fluoro-lH-pyrrolo[2,3- b]pyridin-4-yl)- 1 H-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{l-[l-(3,5-difluoroisonicotinoyl)piperidin-4-yl]-3-[4-(5-fluoro-lH-pyrrolo[2,3- b]pyridin-4-yl)- 1 H-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{l-[l-(3-fluoro-4-hydroxybenzoyl)piperidin-4-yl]-3-[4-(5-fluoro-lH-pyrrolo[2,3- b]pyridin-4-yl)- 1 H-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{l-[l-(2-fluoro-4-hydroxybenzoyl)piperidin-4-yl]-3-[4-(5-fluoro-lH-pyrrolo[2,3- b]pyridin-4-yl)- 1 H-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
2-[(4-{3-(cyanomethyl)-3-[4-(5-fluoro-lH-pyrrolo[2,3-b]pyridin-4-yl)-lH- pyrazol- 1 -yl] azetidin- 1 -yl} piperidin- 1 -yl)carbonyl]terephthalonitrile;
4-[(4-{3-(cyanomethyl)-3-[4-(5-fluoro-lH-pyrrolo[2,3-b]pyridin-4-yl)-lH- pyrazol- 1 -yljazetidin- 1 -yl} piperidin- 1 -yl)carbonyl]-2-fluorobenzonitrile; { 1 - { 1 -[5-chloro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl} -3-[4-(5-fluoro- lH-pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile;
{ 1 - { 1 -[(4,4-difluorocyclohexyl)carbonyl]piperidin-4-yl} -3-[4-(5-fluoro- 1H- pyrrolo[2,3-b]pyridine-4-yl)- 1 H-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
4-{3-(cyanomethyl)-3-[4-(5-fluoro-lH-pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol-l- yljazetidin- 1 -yl} -N-[4-fluoro-2-(trifluoromethyl)phenyl]piperidine- 1 -carboxamide;
4-{3-(cyanomethyl)-3-[4-(5-fluoro-lH-pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol-l- yljazetidin- 1 -yl} -N- [3 -(trifluoromethyl)pyridin-2-yl]piperidine-l -carboxamide;
4-{3-(cyanomethyl)-3-[4-(5-fluoro-lH-pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol-l- yljazetidin- 1 -yl} -N-[4-(trifluoromethyl)pyridin-3-yl]piperidine-l -carboxamide;
4-{3-(cyanomethyl)-3-[4-(5-fluoro-lH-pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol-l- yljazetidin- 1 -yl} -N- [3 -(trifluoromethyl)pyridin-4-yl]piperidine-l -carboxamide;
4-[ 1 -(3-(cyanomethyl)- 1 - { 1 -[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin- 4-yl}azetidin-3-yl)-lH-pyrazol-4-yl]-lH-pyrrolo[2,3-b]pyridine-5-carbonitrile;
4- { 1 -[ 1 -[1 -(3-cyano-5-fluorobenzoyl)piperidin-4-yl]-3-(cyanomethyl)azetidin-3- yl]-lH-pyrazol-4-yl}-lH-pyrrolo[2,3-b]pyridine-5-carbonitrile;
4- { 1 -[ 1 -[1 -(4-cyano-3-fluorobenzoyl)piperidin-4-yl]-3-(cyanomethyl)azetidin-3- yl]-lH-pyrazol-4-yl}-lH-pyrrolo[2,3-b]pyridine-5-carbonitrile;
4-(l - (3-(cyanomethyl)- 1 -[1 -(2,5-dibromobenzoyl)piperidin-4-yl]azetidin-3-yl} - 1 H-pyrazol-4-yl)- 1 H-pyrrolo[2,3-b]pyridine-5-carbonitrile;
4- (l - (3-(cyanomethyl)- 1 - [1 -(3,5-dibromobenzoyl)piperidin-4-yl]azetidin-3-yl} - 1 H-pyrazol-4-yl)- 1 H-pyrrolo[2,3-b]pyridine-5-carbonitrile;
2-[(4-{3-(cyanomethyl)-3-[4-(5-cyano-lH-pyrrolo[2,3-b]pyridin-4-yl)-lH- pyrazol- 1 -yljazetidin- 1 -yl}piperidin- 1 -yl)carbonyl]terephthalonitrile;
5- [(4-{3-(cyanomethyl)-3-[4-(5-cyano-lH-pyrrolo[2,3-b]pyridin-4-yl)-lH- pyrazol- 1 -yljazetidin- 1 -yl}piperidin- 1 -yl)carbonyl]isophthalonitrile;
4- { 1 -[ 1 -[1 -(4-cyano-2-fluorobenzoyl)piperidin-4-yl]-3-(cyanomethyl)azetidin-3- yl]-lH-pyrazol-4-yl}-lH-pyrrolo[2,3-b]pyridine-5-carbonitrile;
4- { 1 -[ 1 -[1 -(4-cyano-2,6-difluorobenzoyl)piperidin-4-yl]-3- (cyanomethyl)azetidin-3-yl]-lH-pyrazol-4-yl}-lH-pyrrolo[2,3-b]pyridine-5-carbonitrile; 4- { 1 -[ 1 - { 1 -[5-chloro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl} -3- (cyanomethyl)azetidin-3-yl]-lH-pyrazol-4-yl}-lH-pyrrolo[2,3-b]pyridine-5-carbonitrile;
{ 1 - { 1 -[5-Chloro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl} -3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[5-Fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl} -3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
[3 - [4-(7H-Pyrrolo[2,3 -d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yl] - 1 -( 1 - { [4- (trifluoromethyl)-l,3-thiazol-2-yl]carbonyl}piperidin-4-yl)azetidin-3-yl]acetonitrile;
[3-[3-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrrol-l-yl]-l-(l-{[2- (trifluoromethyl)pyrimidin-4-yl]carbonyl}piperidin-4-yl)azetidin-3-yl]acetonitrile;
[3-[3-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrrol-l-yl]-l-(l-{[4- (trifluoromethyl)-l,3-thiazol-2-yl]carbonyl}piperidin-4-yl)azetidin-3-yl]acetonitrile;
[3-[3-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrrol-l-yl]-l-(l-{[5- (trifluoromethyl)pyrazin-2-yl] carbonyl} piperidin-4-yl)azetidin-3 -yl] acetonitrile;
{l-[l-(Methylsulfonyl)piperidin-4-yl]-3-[3-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- 1 H-pyrrol- 1 -yl] azetidin-3 -yl} acetonitrile;
[3-[4-( 1 H-Pyrrolo[2,3-b]pyridin-4-yl)- 1 H-pyrazol- 1 -yl]- 1 -( 1 - { [5- (trifluoromethyl)pyrazin-2-yl] carbonyl} piperidin-4-yl)azetidin-3 -yl] acetonitrile;
[3 - [4-( 1 H-Pyrrolo[2,3 -b]pyridin-4-yl)- 1 H-pyrazol- 1 -yl]- 1 -( 1 - { [4- (trifluoromethyl)-l,3-thiazol-2-yl]carbonyl}piperidin-4-yl)azetidin-3-yl]acetonitrile;
{l-[l-(Methylsulfonyl)piperidin-4-yl]-3-[4-(lH-pyrrolo[2,3-b]pyridin-4-yl)-lH- pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
[3-[4-(5-Fluoro-lH-pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol-l-yl]-l-(l-{[6- (trifluoromethyl)pyrazin-2-yl] carbonyl} piperidin-4-yl)azetidin-3 -yl] acetonitrile;
[3-[4-(5-Fluoro-lH-pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol-l-yl]-l-(l-{[2- (trifluoromethyl)pyrimidin-4-yl]carbonyl}piperidin-4-yl)azetidin-3-yl]acetonitrile;
[3-[4-(5-Fluoro-lH-pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol-l-yl]-l-(l-{[5- (trifluoromethyl)pyrazin-2-yl] carbonyl} piperidin-4-yl)azetidin-3 -yl] acetonitrile;
{3-[4-(5-Fluoro-lH-pyrrolo[2,3-b]pyridin-4-yl)-lHpyrazol-l-yl]-l-[l- (methylsulfonyl)piperidin-4-yl]azetidin-3-yl} acetonitrile; 4- [ 1 -(3 -(Cyanomethyl)- 1 - { 1 - [5-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin- 4-yl} azetidin-3-yl)- 1 H-pyrazol-4-yl]- 1 H-pyrrolo[2,3 -b]pyridine-5-carbonitrile; and
4-(l - (3-(Cyanomethyl)- 1 -[1 -(methylsulfonyl)piperidin-4-yl]azetidin-3-yl} -1 H- pyrazol-4-yl)-lH-pyrrolo[2,3-b]pyridine-5-carbonitrile;
or a pharmaceutically acceptable salt of any of the aforementioned.
48. The compound according to claim 1, wherein the compound is selected from: cis- { 1 - {(3-Methoxy- 1 -[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl} - 3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{ 1 -(cis-3 -Methoxy- 1 - { [2-(trifluoromethyl)pyrimidin-4-yl] carbonyl} piperidin-4- yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{ 1 - {cis-3-Fluoro- 1 -[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl} -3- [4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{ 1 -(cis-3 -Fluoro- 1 - { [2-(trifluoromethyl)pyrimidin-4-yl] carbonyl} piperidin-4-yl)-
3- [4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{ 1 - { 1 -[3-Fluoro-2-(trifluoromethyl)isonicotinoyl]-4-deuteropiperidin-4-yl} -3-[4- (7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{l-{l-[3-Fluoro-2-(trifluoromethyl)isonicotinoyl]3,3,4,5,5-pentadeuteropiperidin-
4- yl} -3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{ 1 - {7-[3-Fluoro-2-(trifluoromethyl)isonicotinoyl]-3-oxa-7-azabicyclo[3.3.1 ]non- 9-yl} -3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{ 1 -(1 - {[4-[(dimethylamino)methyl]-6-(trifluoromethyl)pyridin-2- yl]carbonyl}piperidin-4-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-3-yl} acetonitrile;
4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yljazetidin- 1 -yl} -N-isopropylpiperidine- 1 -carboxamide;
{ 1 - { 1 - [6- [(dimethylamino)methyl] -3 -fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -y 1] azetidin-3 -y 1 } acetonitrile;
3 - [(4- {3 -(cyanomethyl)-3 - [4-(7H-pyrrolo[2 ,3 -d]pyrimidin-4-yl)- 1 H-pyrazol- 1 - yl]azetidin-l-yl}piperidin-l-yl)carbonyl]-5-[(dimethylamino)methyl]benzonitrile; { 1 -(1 - { [6-[(dimethylamino)methyl]-2-(trifluoromethyl)pyrimidin-4- yl]carbonyl}piperidin-4-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-3-yl} acetonitrile;
{ 1 -(1 - {[6-[(methylamino)methyl]-2-(trifluoromethyl)pyrimidin-4- yl]carbonyl}piperidin-4-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-3-yl} acetonitrile;
{ 1 -( 1 - { [6-(azetidin- 1 -ylmethyl)-2-(trifluoromethyl)pyrimidin-4- yl]carbonyl}piperidin-4-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-3-yl} acetonitrile;
{ 1 -(1 - { [6-[(diethylamino)methyl]-2-(trifluoromethyl)pyrimidin-4- yl]carbonyl}piperidin-4-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-3-yl} acetonitrile;
{ 1 -(1 - { [6- { [ethyl(methyl)amino]methyl} -2-(trifluoromethyl)pyrimidin-4- yl]carbonyl}piperidin-4-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-3-yl} acetonitrile;
{ 1 -(1 - {3-(difluoromethyl)-5-[(dimethylamino)methyl]benzoyl}piperidin-4-yl)-3- [4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- IH-pyrazol- 1 -yl]azetidin-3-yl} acetonitrile;
{ 1 -( 1 - { [6-(pyrrolidin- 1 -ylmethyl)-2-(trifluoromethyl)pyrimidin-4- yl]carbonyl}piperidin-4-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-3-yl} acetonitrile;
{l-(l-{[6-{[(3 S)-3 -fluoropyrrolidin- 1 -yljmethyl} -2-(trifluoromethy l)pyrimidin-4- yl]carbonyl}piperidin-4-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-3-yl} acetonitrile;
{ 1 -(1 - { [6- { [(3R)-3-fluoropyrrolidin- 1 -yljmethyl} -2-(trifluoromethyl)pyrimidin-4- yl]carbonyl}piperidin-4-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-3-yl} acetonitrile;
{l-(l-{[6-[(3 ,3 -difluoropyrrolidin- 1 -yl)methyl] -2-(trifluoromethyl)pyrimidin-4- yl]carbonyl}piperidin-4-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-3-yl} acetonitrile;
{ 1 -(1 - {[6-[(tert-butylamino)methyl]-2-(trifluoromethyl)pyrimidin-4- yl]carbonyl}piperidin-4-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-3-yl} acetonitrile;
{l-(l-{[6-(hydroxymethyl)-2-(trifluoromethyl)pyrimidin-4-yl]carbonyl}piperidin- 4-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{ 1 -(1 - {[6-[(isopropylamino)methyl]-2-(trifluoromethyl)pyrimidin-4- yl]carbonyl}piperidin-4-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-3-yl} acetonitrile;
{ 1 -(1 - { [6-[(ethylamino)methyl]-2-(trifluoromethyl)pyrimidin-4- yl]carbonyl}piperidin-4-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yl]azetidin-3-yl} acetonitrile;
{l-(l-{[6-{ [(2-methoxyethy l)(methyl)amino]methyl} -2- (trifluoromethyl)pyrimidin-4-yl]carbonyl}piperidin-4-yl)-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
{l-(l-{[6-{[(3 -hydroxypropyl)(methyl)amino]methyl} -2- (trifluoromethyl)pyrimidin-4-yl]carbonyl}piperidin-4-yl)-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl} acetonitrile;
propyl 4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol- 1 -yljazetidin- 1 -yl}piperidine- 1 -carboxylate;
cyclobutylmethyl 4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 -yljazetidin- 1 -yl}piperidine- 1 -carboxylate;
ethyl 4- {3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- 1 - yljazetidin- 1 -yl}piperidine- 1 -carboxylate;
benzyl 4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol- 1 -yljazetidin- 1 -yl}piperidine- 1 -carboxylate;
isobutyl 4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol- 1 -yljazetidin- 1 -yl}piperidine- 1 -carboxylate;
cyclopropylmethyl 4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yljazetidin- 1 -yl}piperidine- 1 -carboxylate;
(l-methylcyclopropyl)methyl 4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yljazetidin- 1 -yl}piperidine- 1 -carboxylate;
2,4-difluorobenzyl 4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- 1 H-pyrazol- 1 -yljazetidin- 1 -yl}piperidine- 1 -carboxylate;
3.4- difluorobenzyl 4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- H-pyrazol- 1 -yljazetidin- 1 -yl}piperidine- 1 -carboxylate;
3.5- difluorobenzyl 4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- H-pyrazol- 1 -yljazetidin- 1 -yl}piperidine- 1 -carboxylate;
cyclopentylmethyl 4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- H-pyrazol- 1 -yl] azetidin- 1 -y 1} piperidine- 1 -carboxylate; and
cyclohexylmethyl 4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- H-pyrazol- 1 -yljazetidin- 1 -yl}piperidine- 1 -carboxylate;
or a pharmaceutically acceptable salt of any of the aforementioned.
49. A compound according to claim 1 , which is { 1 - { 1 -[3-Fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin- 4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof.
50. A salt according to claim 49, which is {l-{l-[3-Fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin- 4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile adipic acid salt.
51. The salt according to claim 50, which is characterized by a melting point of about 178 °C.
52. The salt according to claim 50 or 51 having a differential scanning calorimetry thermogram which is characterized by an endothermic peak with an onset temperature of about 176 °C.
53. The salt according to claim 50 or 51, having a differential scanning calorimetry thermogram substantially as shown in Figure 1.
54. The salt according to any one of claims 50 to 53, having a thermal gravimetric analysis thermogram substantially as shown in Figure 2. The salt according to any one of claims 50 to 54 having an X-ray powder diffraction pattern comprising a characteristic peak expressed in degrees 2Θ about 10.4.
The salt according to any one of claims 50 to 54 having an X-ray powder diffraction pattern comprising a characteristic peak expressed in degrees 2Θ about 6.9.
The salt according to any one of claims 50 to 54 having an X-ray powder diffraction pattern comprising a characteristic peak expressed in degrees 2Θ about 21.0.
The salt according to any one of claims 50 to 54 having an X-ray powder diffraction pattern comprising a characteristic peak expressed in degrees 2Θ about 23.3.
The salt according to any one of claims 50 to 54 having an X-ray powder diffraction pattern comprising a characteristic peak expressed in degrees 2Θ about 6.9, 10.4, 21.0, and 23.3.
60. The salt according to any one of claims 50 to 54 having an X-ray powder
diffraction pattern substantially as shown in Figure 3.
A composition comprising a compound or salt according to any one of claims 1 to 60 and a pharmaceutically acceptable carrier.
A method of modulating an activity of JAK1 comprising contacting JAK1 with compound or salt according to any one of claims 1 to 60.
63. A method according to claim 62, wherein said compound, or pharmaceutically acceptable salt thereof, is selective for JAK1 over JAK2.
64. A method of treating an autoimmune disease, a cancer, a myeloproliferative disorder, an inflammatory disease, a bone resorption disease, or organ transplant rejection in a patient in need thereof, comprising administering to said patient a therapeutically effective amount of a compound or salt according to any one of claims 1 to 60.
65. A method according to claim 64, wherein said autoimmune disease is a skin disorder, multiple sclerosis, rheumatoid arthritis, psoriatic arthritis, juvenile arthritis, type I diabetes, lupus, inflammatory bowel disease, Crohn's disease, myasthenia gravis, immunoglobulin nephropathies, myocarditis, or autoimmune thyroid disorder.
66. A method according to claim 64, wherein said autoimmune disease is rheumatoid arthritis.
67. A method according to claim 64, wherein said autoimmune disease is a skin disorder.
68. A method according to claim 67, wherein said skin disorder is atopic dermatitis, psoriasis, skin sensitization, skin irritation, skin rash, contact dermatitis or allergic contact sensitization.
69. A method according to claim 64, wherein said cancer is a solid tumor.
70. A method according to claim 64, wherein said cancer is prostate cancer, renal cancer, hepatic cancer, breast cancer, lung cancer, thyroid cancer, Kaposi's sarcoma, Castleman's disease or pancreatic cancer.
71. A method according to claim 64, wherein said cancer is lymphoma, leukemia, or multiple myeloma.
72. A method according to claim 64, wherein said myeloproliferative disorder is polycythemia vera (PV), essential thrombocythemia (ET), myeloid metaplasia with myelofibrosis (MMM), primary myelofibrosis (PMF), chronic myelogenous leukemia (CML), chronic myelomonocytic leukemia (CMML),
hypereosinophilic syndrome (HES), idiopathic myelofibrosis (IMF), or systemic mast cell disease (SMCD).
73. A method according to claim 64, wherein said myeloproliferative disorder is myelofibrosis.
74. A method according to claim 64, wherein said myeloproliferative disorder is primary myelofibrosis (PMF).
75. A method according to claim 64, wherein said bone resorption disease is
osteoporosis, osteoarthritis, bone resorption associated with hormonal imbalance, bone resorption associated with hormonal therapy, bone resorption associated with autoimmune disease, or bone resorption associated with cancer.
PCT/US2011/027665 2010-03-10 2011-03-09 Piperidin-4-yl azetidine derivatives as jak1 inhibitors WO2011112662A1 (en)

Priority Applications (43)

Application Number Priority Date Filing Date Title
MYPI2012003994A MY175156A (en) 2010-03-10 2011-03-09 Piperidin-4-yl azetidine derivatives as jak1 inhibitors
EA201290894A EA030376B1 (en) 2010-03-10 2011-03-09 Piperidin-4-yl azetidine derivatives as jak1 inhibitors
KR1020127026216A KR101857680B1 (en) 2010-03-10 2011-03-09 Piperidin-4-yl azetidine derivatives as jak1 inhibitors
KR1020187013055A KR101911697B1 (en) 2010-03-10 2011-03-09 Piperidin-4-yl azetidine derivatives as jak1 inhibitors
PL17211097T PL3354652T3 (en) 2010-03-10 2011-03-09 Piperidin-4-yl azetidine derivatives as jak1 inhibitors
MEP-2016-41A ME02386B (en) 2010-03-10 2011-03-09 Piperidin-4-yl azetidine derivatives as jak1 inhibitors
KR1020217023315A KR102354472B1 (en) 2010-03-10 2011-03-09 Piperidin-4-yl azetidine derivatives as jak1 inhibitors
KR1020207030868A KR102283091B1 (en) 2010-03-10 2011-03-09 Piperidin-4-yl azetidine derivatives as jak1 inhibitors
KR1020227001841A KR20220015492A (en) 2010-03-10 2011-03-09 Piperidin-4-yl azetidine derivatives as jak1 inhibitors
EP11711709.3A EP2545045B1 (en) 2010-03-10 2011-03-09 Piperidin-4-yl azetidine derivatives as jak1 inhibitors
NZ602313A NZ602313A (en) 2010-03-10 2011-03-09 Piperidin-4-yl azetidine derivatives as jak1 inhibitors
JP2012557194A JP5858484B2 (en) 2010-03-10 2011-03-09 Piperidin-4-ylazetidine derivatives as JAK1 inhibitors
MX2018002077A MX364636B (en) 2010-03-10 2011-03-09 Piperidin-4-yl azetidine derivatives as jak1 inhibitors.
ES11711709T ES2569539T3 (en) 2010-03-10 2011-03-09 Piperidin-4-yl azetidine derivatives as JAK1 inhibitors
SG2012064457A SG183551A1 (en) 2010-03-10 2011-03-09 Piperidin-4-yl azetidine derivatives as jak1 inhibitors
BR112012022513A BR112012022513A2 (en) 2010-03-10 2011-03-09 piperidin-4-yl azetidine derivatives as jak1 inhibitors
DK11711709.3T DK2545045T3 (en) 2010-03-10 2011-03-09 PIPERIDINE-4-YL-azetidine derivatives AS JAK1 INHIBITORS
EP20158754.0A EP3715347B1 (en) 2010-03-10 2011-03-09 Piperidin-4-yl azetidine derivatives as jak1 inhibitors
MX2017006392A MX354212B (en) 2010-03-10 2011-03-09 Piperidin-4-yl azetidine derivatives as jak1 inhibitors.
AU2011224484A AU2011224484A1 (en) 2010-03-10 2011-03-09 Piperidin-4-yl azetidine derivatives as JAK1 inhibitors
RS20160205A RS54823B1 (en) 2010-03-10 2011-03-09 Piperidin-4-yl azetidine derivatives as jak1 inhibitors
SI201130747A SI2545045T1 (en) 2010-03-10 2011-03-09 Piperidin-4-yl azetidine derivatives as jak1 inhibitors
MX2012010344A MX336932B (en) 2010-03-10 2011-03-09 Piperidin-4-yl azetidine derivatives as jak1 inhibitors.
KR1020207008092A KR102172742B1 (en) 2010-03-10 2011-03-09 Piperidin-4-yl azetidine derivatives as jak1 inhibitors
MX2016001708A MX347851B (en) 2010-03-10 2011-03-09 Piperidin-4-yl azetidine derivatives as jak1 inhibitors.
PL11711709T PL2545045T3 (en) 2010-03-10 2011-03-09 Piperidin-4-yl azetidine derivatives as jak1 inhibitors
KR1020187030015A KR20180117206A (en) 2010-03-10 2011-03-09 Piperidin-4-yl azetidine derivatives as jak1 inhibitors
EP17211097.5A EP3354652B1 (en) 2010-03-10 2011-03-09 Piperidin-4-yl azetidine derivatives as jak1 inhibitors
EP21213769.9A EP4036088B1 (en) 2010-03-10 2011-03-09 Piperidin-4-yl azetidine derivatives as jak1 inhibitors
CN201180023757.7A CN102985417B (en) 2010-03-10 2011-03-09 Piperidin-4-yl azetidine derivatives as JAK1 inhibitors
CA2792508A CA2792508C (en) 2010-03-10 2011-03-09 Piperidin-4-yl azetidine derivatives as jak1 inhibitors
UAA201211696A UA110108C2 (en) 2010-03-10 2011-09-03 Azetidine derivatives of piperidine-4-yl as JAK1 inhibitors
IL221823A IL221823A (en) 2010-03-10 2012-09-06 Piperidin-4-yl-azetidine derivatives as jak1 inhibitors
ZA2012/07420A ZA201207420B (en) 2010-03-10 2012-10-03 Piperidin-4-yl azetidine derivatives as jak1 inhibitors
HK13104880.0A HK1177741A1 (en) 2010-03-10 2013-04-23 Piperidin-4-yl azetidine derivatives as jak1 inhibitors jak1 -4-
AU2015205858A AU2015205858B2 (en) 2010-03-10 2015-07-21 Piperidin-4-yl azetidine derivatives as jak1 inhibitors
PH12015502575A PH12015502575B1 (en) 2010-03-10 2015-11-13 Piperidin-4-yl azetidine derivatives as jak1 inhibitors
SM201600085T SMT201600085B (en) 2010-03-10 2016-03-24 PIPERIDIN-4-AZETIDIN DERIVED AS INHIBITORS OF JAK1
HRP20160326TT HRP20160326T1 (en) 2010-03-10 2016-04-01 Piperidin-4-yl azetidine derivatives as jak1 inhibitors
AU2017201801A AU2017201801B2 (en) 2010-03-10 2017-03-16 Piperidin-4-yl azetidine derivatives as jak1 inhibitors
AU2019257385A AU2019257385B2 (en) 2010-03-10 2019-10-29 Piperidin-4-yl azetidine derivatives as jak1 inhibitors
CY20201100671T CY1123175T1 (en) 2010-03-10 2020-07-22 PIPERIDIN-4-YL AZETIDINE DERIVATIVES AS JAK1 INHIBITORS
AU2021212085A AU2021212085B2 (en) 2010-03-10 2021-08-05 Piperidin-4-yl azetidine derivatives as jak1 inhibitors

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US31258810P 2010-03-10 2010-03-10
US61/312,588 2010-03-10
US41560210P 2010-11-19 2010-11-19
US61/415,602 2010-11-19

Publications (1)

Publication Number Publication Date
WO2011112662A1 true WO2011112662A1 (en) 2011-09-15

Family

ID=44065466

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2011/027665 WO2011112662A1 (en) 2010-03-10 2011-03-09 Piperidin-4-yl azetidine derivatives as jak1 inhibitors

Country Status (38)

Country Link
US (6) US8765734B2 (en)
EP (5) EP3715347B1 (en)
JP (6) JP5858484B2 (en)
KR (7) KR20220015492A (en)
CN (1) CN102985417B (en)
AR (1) AR081315A1 (en)
AU (1) AU2011224484A1 (en)
BR (1) BR112012022513A2 (en)
CA (1) CA2792508C (en)
CL (1) CL2012002501A1 (en)
CO (1) CO6602158A2 (en)
CR (1) CR20120510A (en)
CY (2) CY1120154T1 (en)
DK (3) DK3354652T3 (en)
EA (1) EA030376B1 (en)
EC (1) ECSP12012218A (en)
ES (4) ES2569539T3 (en)
HK (2) HK1177741A1 (en)
HR (3) HRP20160326T1 (en)
HU (3) HUE037077T2 (en)
IL (1) IL221823A (en)
LT (2) LT3050882T (en)
ME (2) ME02386B (en)
MX (5) MX364636B (en)
MY (2) MY175156A (en)
NO (1) NO3050882T3 (en)
NZ (1) NZ602313A (en)
PE (1) PE20130038A1 (en)
PH (1) PH12015502575B1 (en)
PL (3) PL3354652T3 (en)
PT (2) PT3354652T (en)
RS (3) RS54823B1 (en)
SG (1) SG183551A1 (en)
SI (3) SI3354652T1 (en)
SM (1) SMT201600085B (en)
TW (6) TWI592413B (en)
WO (1) WO2011112662A1 (en)
ZA (1) ZA201207420B (en)

Cited By (55)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013026025A1 (en) * 2011-08-18 2013-02-21 Incyte Corporation Cyclohexyl azetidine derivatives as jak inhibitors
WO2013041042A1 (en) * 2011-09-22 2013-03-28 Merck Sharp & Dohme Corp. Pyrazole carboxamides as janus kinase inhibitors
WO2013173720A1 (en) * 2012-05-18 2013-11-21 Incyte Corporation Piperidinylcyclobutyl substituted pyrrolopyridine and pyrrolopyrimidine derivatives as jak inhibitors
US8691807B2 (en) 2011-06-20 2014-04-08 Incyte Corporation Azetidinyl phenyl, pyridyl or pyrazinyl carboxamide derivatives as JAK inhibitors
US8722693B2 (en) 2007-06-13 2014-05-13 Incyte Corporation Salts of the Janus kinase inhibitor (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile
WO2014138168A1 (en) * 2013-03-06 2014-09-12 Incyte Corporation Processes and intermediates for making a jak inhibitor
WO2014146246A1 (en) * 2013-03-19 2014-09-25 Merck Sharp & Dohme Corp. Cycloalkyl nitrile pyrazolo pyridones as janus kinase inhibitors
WO2014161046A1 (en) * 2013-04-04 2014-10-09 The Walter And Eliza Hall Institute Of Medical Research Methods of treating diseases characterized by excessive wnt signalling
US8933086B2 (en) 2005-12-13 2015-01-13 Incyte Corporation Heteroaryl substituted pyrrolo[2,3-B]pyridines and pyrrolo[2,3-B]pyrimidines as Janus kinase inhibitors
US8933085B2 (en) 2010-11-19 2015-01-13 Incyte Corporation Cyclobutyl substituted pyrrolopyridine and pyrrolopyrimidine derivatives as JAK inhibitors
WO2015021153A1 (en) * 2013-08-07 2015-02-12 Incyte Corporation Sustained release dosage forms for a jak1 inhibitor
US9034884B2 (en) 2010-11-19 2015-05-19 Incyte Corporation Heterocyclic-substituted pyrrolopyridines and pyrrolopyrimidines as JAK inhibitors
US9216984B2 (en) 2009-05-22 2015-12-22 Incyte Corporation 3-[4-(7H-pyrrolo[2,3-D]pyrimidin-4-yl)-1H-pyrazol-1-yl]octane—or heptane-nitrile as JAK inhibitors
US9249145B2 (en) 2009-09-01 2016-02-02 Incyte Holdings Corporation Heterocyclic derivatives of pyrazol-4-yl-pyrrolo[2,3-d]pyrimidines as janus kinase inhibitors
US9334274B2 (en) 2009-05-22 2016-05-10 Incyte Holdings Corporation N-(hetero)aryl-pyrrolidine derivatives of pyrazol-4-yl-pyrrolo[2,3-d]pyrimidines and pyrrol-3-yl-pyrrolo[2,3-d]pyrimidines as janus kinase inhibitors
US9464088B2 (en) 2010-03-10 2016-10-11 Incyte Holdings Corporation Piperidin-4-yl azetidine derivatives as JAK1 inhibitors
US9487521B2 (en) 2011-09-07 2016-11-08 Incyte Holdings Corporation Processes and intermediates for making a JAK inhibitor
US9498467B2 (en) 2014-05-30 2016-11-22 Incyte Corporation Treatment of chronic neutrophilic leukemia (CNL) and atypical chronic myeloid leukemia (aCML) by inhibitors of JAK1
US9505776B2 (en) 2013-03-14 2016-11-29 Alkermes Pharma Ireland Limited Prodrugs of fumarates and their use in treating various diseases
WO2016196244A1 (en) 2015-05-29 2016-12-08 Incyte Corporation Pyridineamine compounds useful as pim kinase inhibitors
KR20160146778A (en) * 2014-04-08 2016-12-21 인사이트 코포레이션 Treatment of b-cell malignancies by a combination jak and pi3k inhibitor
US9550765B2 (en) 2013-01-15 2017-01-24 Incyte Holdings Corporation Thiazolecarboxamides and pyridinecarboxamide compounds useful as Pim kinase inhibitors
WO2017013270A1 (en) 2015-07-23 2017-01-26 Universite De Strasbourg Use of leptin signaling inhibitor for protecting kidneys from patients having ciliopathy
WO2017044730A1 (en) 2015-09-09 2017-03-16 Incyte Corporation Salts of a pim kinase inhibitor
US9604922B2 (en) 2014-02-24 2017-03-28 Alkermes Pharma Ireland Limited Sulfonamide and sulfinamide prodrugs of fumarates and their use in treating various diseases
WO2017059251A1 (en) 2015-10-02 2017-04-06 Incyte Corporation Heterocyclic compounds useful as pim kinase inhibitors
US9676750B2 (en) 2013-01-14 2017-06-13 Incyte Corporation Bicyclic aromatic carboxamide compounds useful as pim kinase inhibitors
CN106905322A (en) * 2016-01-26 2017-06-30 杭州华东医药集团新药研究院有限公司 Pyrrolopyrimidine penta azacyclo derivative and its application
WO2017162604A1 (en) 2016-03-21 2017-09-28 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for diagnosis and treatment of solar lentigo
US9822124B2 (en) 2014-07-14 2017-11-21 Incyte Corporation Bicyclic heteroaromatic carboxamide compounds useful as Pim kinase inhibitors
US9890162B2 (en) 2014-07-14 2018-02-13 Incyte Corporation Bicyclic aromatic carboxamide compounds useful as pim kinase inhibitors
US9957265B2 (en) 2013-03-19 2018-05-01 Merck Sharp & Dohme Corp. N-(2-cyano heterocyclyl) pyrazolo pyridones as janus kinase inhibitors
US10000507B2 (en) 2013-08-23 2018-06-19 Incyte Corporation Furo- and thieno-pyridine carboxamide compounds useful as pim kinase inhibitors
US10059705B2 (en) 2013-03-19 2018-08-28 Merck Sharp & Dohme Corp. Acyclic cyanoethylpyrazolo pyridones as janus kinase inhibitors
WO2018167283A1 (en) 2017-03-17 2018-09-20 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for the diagnosis and treatment of pancreatic ductal adenocarcinoma associated neural remodeling
US10166191B2 (en) 2012-11-15 2019-01-01 Incyte Corporation Sustained-release dosage forms of ruxolitinib
WO2019152374A1 (en) * 2018-01-30 2019-08-08 Incyte Corporation Processes for preparing (1 -(3-fluoro-2-(trifluoromethyl)isonicotinyl)piperidine-4-one)
WO2019171326A1 (en) 2018-03-08 2019-09-12 Novartis Ag Use of an anti-p-selectin antibody
WO2020039401A1 (en) 2018-08-24 2020-02-27 Novartis Ag Treatment comprising il-1βeta binding antibodies and combinations thereof
US10596161B2 (en) 2017-12-08 2020-03-24 Incyte Corporation Low dose combination therapy for treatment of myeloproliferative neoplasms
WO2020092015A1 (en) 2018-11-02 2020-05-07 University Of Rochester Therapeutic mitigation of epithelial infection
US10758543B2 (en) 2010-05-21 2020-09-01 Incyte Corporation Topical formulation for a JAK inhibitor
WO2020182159A1 (en) 2019-03-14 2020-09-17 上海华汇拓医药科技有限公司 Jak kinase inhibitor, preparation method for same, and applications thereof in field of medicine
WO2020256739A1 (en) 2019-06-21 2020-12-24 Constellation Pharmaceuticals, Inc. Methods of treating myeloproliferative disorders
WO2020257644A1 (en) 2019-06-21 2020-12-24 Constellation Pharmaceuticals, Inc. Methods of treating myeloproliferative disorders
WO2021053490A1 (en) 2019-09-16 2021-03-25 Novartis Ag Use of high-affinity, ligand-blocking, humanized anti-t-cell immunoglobulin domain and mucin domain-3 (tim-3) igg4 antibody for the treatment of myelofibrosis
WO2021053489A1 (en) 2019-09-16 2021-03-25 Novartis Ag Use of an mdm2 inhibitor for the treatment of myelofibrosis
WO2021091535A1 (en) 2019-11-05 2021-05-14 Constellation Pharmaceuticals, Inc. Treating myeloproliferative disorders with cpi-0610 and a jak inhibitor
US11045455B2 (en) 2015-12-11 2021-06-29 Sichuan Kelun-Biotech Biopharmaceutical Co., Ltd. Azetidine derivative, preparation method therefor, and use thereof
US11230548B2 (en) 2013-03-14 2022-01-25 Alkermes Pharma Ireland Limited Prodrugs of fumarates and their use in treating various diseases
WO2022074600A1 (en) 2020-10-08 2022-04-14 Novartis Ag Use of an erk inhibitor for the treatment of myelofibrosis
US11304949B2 (en) 2018-03-30 2022-04-19 Incyte Corporation Treatment of hidradenitis suppurativa using JAK inhibitors
US11384064B2 (en) 2018-01-26 2022-07-12 Rapt Therapeutics, Inc. Chemokine receptor modulators and uses thereof
US11833155B2 (en) 2020-06-03 2023-12-05 Incyte Corporation Combination therapy for treatment of myeloproliferative neoplasms
US11878976B2 (en) 2021-03-04 2024-01-23 Eli Lilly And Company FGFR3 inhibitor compounds

Families Citing this family (64)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CL2008001709A1 (en) 2007-06-13 2008-11-03 Incyte Corp Compounds derived from pyrrolo [2,3-b] pyrimidine, jak kinase modulators; pharmaceutical composition; and use in the treatment of diseases such as cancer, psoriasis, rheumatoid arthritis, among others.
CL2009001884A1 (en) * 2008-10-02 2010-05-14 Incyte Holdings Corp Use of 3-cyclopentyl-3- [4- (7h-pyrrolo [2,3-d] pyrimidin-4-yl) -1h-pyrazol-1-yl) propanonitrile, janus kinase inhibitor, and use of a composition that understands it for the treatment of dry eye.
JOP20190230A1 (en) 2009-01-15 2017-06-16 Incyte Corp Processes for preparing jak inhibitors and related intermediate compounds
AR077280A1 (en) 2009-06-29 2011-08-17 Incyte Corp PYRIMIDINONES AS PI3K INHIBITORS, AND PHARMACEUTICAL COMPOSITIONS THAT UNDERSTAND THEM
TW201249844A (en) 2010-12-20 2012-12-16 Incyte Corp N-(1-(substituted-phenyl)ethyl)-9H-purin-6-amines as PI3K inhibitors
WO2012112847A1 (en) 2011-02-18 2012-08-23 Novartis Pharma Ag mTOR/JAK INHIBITOR COMBINATION THERAPY
EP2741747A1 (en) 2011-08-10 2014-06-18 Novartis Pharma AG JAK P13K/mTOR COMBINATION THERAPY
SI2751109T1 (en) 2011-09-02 2017-03-31 Incyte Holdings Corporation Heterocyclylamines as pi3k inhibitors
AR090548A1 (en) 2012-04-02 2014-11-19 Incyte Corp BICYCLIC AZAHETEROCICLOBENCILAMINS AS PI3K INHIBITORS
MD20150043A2 (en) 2012-10-02 2015-08-31 Epitherapeutics Aps Inhibitors of histone demethylases
SG10201703533VA (en) 2012-11-01 2017-06-29 Incyte Corp Tricyclic fused thiophene derivatives as jak inhibitors
EP3170811A1 (en) 2013-02-27 2017-05-24 Gilead Sciences, Inc. Ethyl pyridine-4-carboxylate compound as inhibitor of histone demethylases
WO2014146249A1 (en) * 2013-03-19 2014-09-25 Merck Sharp & Dohme Corp. Geminally substituted cyanoethylpyrazolo pyridones as janus kinase inhibitors
PE20160126A1 (en) 2013-05-17 2016-02-24 Incyte Corp DERIVATIVES OF BIPIRAZOLE AS JAK INHIBITORS
CN105555313A (en) 2013-08-20 2016-05-04 因赛特公司 Survival benefit in patients with solid tumors with elevated c-reactive protein levels
JP6367545B2 (en) * 2013-12-17 2018-08-01 コンサート ファーマシューティカルズ インコーポレイテッド Deuterated derivatives of ruxolitinib
CN106456773A (en) 2014-02-28 2017-02-22 因赛特公司 Jak1 inhibitors for the treatment of myelodysplastic syndromes
AU2015253192B2 (en) 2014-04-30 2019-05-16 Incyte Holdings Corporation Processes of preparing a JAK1 inhibitor and new forms thereto
WO2015191677A1 (en) 2014-06-11 2015-12-17 Incyte Corporation Bicyclic heteroarylaminoalkyl phenyl derivatives as pi3k inhibitors
CN107074807A (en) * 2014-08-27 2017-08-18 吉利德科学公司 Composition and method for inhibition of histone demethylase
WO2016130501A1 (en) 2015-02-09 2016-08-18 Incyte Corporation Aza-heteroaryl compounds as pi3k-gamma inhibitors
CN117800973A (en) 2015-02-27 2024-04-02 因赛特控股公司 Salts of PI3K inhibitors and methods of making the same
US9840503B2 (en) 2015-05-11 2017-12-12 Incyte Corporation Heterocyclic compounds and uses thereof
WO2016183060A1 (en) 2015-05-11 2016-11-17 Incyte Corporation Process for the synthesis of a phosphoinositide 3-kinase inhibitor
US9988401B2 (en) 2015-05-11 2018-06-05 Incyte Corporation Crystalline forms of a PI3K inhibitor
WO2017027717A1 (en) 2015-08-12 2017-02-16 Incyte Corporation Bicyclic fused pyrimidine compounds as tam inhibitors
US10053465B2 (en) 2015-08-26 2018-08-21 Incyte Corporation Pyrrolopyrimidine derivatives as TAM inhibitors
TWI744256B (en) 2015-11-06 2021-11-01 美商英塞特公司 HETEROCYCLIC COMPOUNDS AS PI3K-γ INHIBITORS
ES2833955T3 (en) 2016-01-05 2021-06-16 Incyte Corp Pyridines substituted with pyrazole / imidazole as PI3K-Gamma inhibitors
CA3019145A1 (en) 2016-03-28 2017-10-05 Incyte Corporation Pyrrolotriazine compounds as tam inhibitors
WO2017223414A1 (en) 2016-06-24 2017-12-28 Incyte Corporation HETEROCYCLIC COMPOUNDS AS PI3K-γ INHIBITORS
AU2017316997B2 (en) * 2016-08-24 2022-06-30 Royal Melbourne Institute Of Technology Diagnostic methods and device
GB201617871D0 (en) * 2016-10-21 2016-12-07 Sareum Limited Pharmaceutical compounds
US20180153922A1 (en) 2016-12-06 2018-06-07 New York Society For The Ruptured And Crippled Maintaining The Hospital For Special Surgery Inhibition of expansion and function of pathogenic age-associated b cells and use for the prevention and treatment of autoimmune disease
KR102585048B1 (en) 2017-01-17 2023-10-05 아스트라제네카 아베 JAK1 selective inhibitor
CN109422753B (en) * 2017-09-03 2021-12-31 上海美志医药科技有限公司 Compounds capable of inhibiting and degrading tyrosine protein kinase JAK1 or JAK2 activity
RS62872B1 (en) 2017-09-27 2022-02-28 Incyte Corp Salts of pyrrolotriazine derivatives useful as tam inhibitors
SG11202003428VA (en) 2017-10-18 2020-05-28 Incyte Corp Condensed imidazole derivatives substituted by tertiary hydroxy groups as pi3k-gamma inhibitors
CN109867676B (en) * 2017-12-01 2020-10-30 北京普祺医药科技有限公司 Pyrrolopyrimidine derivative compound, pharmaceutical composition and application thereof
SG11202007805SA (en) * 2018-02-16 2020-09-29 Incyte Corp Jak1 pathway inhibitors for the treatment of cytokine-related disorders
US11584961B2 (en) 2018-03-30 2023-02-21 Incyte Corporation Biomarkers for inflammatory skin disease
MX2020010815A (en) 2018-04-13 2020-12-11 Incyte Corp Biomarkers for graft-versus-host disease.
SG11202011680YA (en) 2018-06-01 2020-12-30 Incyte Corp Dosing regimen for the treatment of pi3k related disorders
JP2021529765A (en) 2018-06-29 2021-11-04 インサイト・コーポレイションIncyte Corporation Formulation of AXL / MER inhibitor
CR20210165A (en) 2018-09-05 2021-10-01 Incyte Corp Crystalline forms of a phosphoinositide 3-kinase (pi3k) inhibitor
US11324749B2 (en) 2018-10-31 2022-05-10 Incyte Corporation Combination therapy for treatment of hematological diseases
KR101992148B1 (en) 2018-11-30 2019-06-25 한승훈 Seat belt anti-twist system
CN113692278A (en) 2018-12-19 2021-11-23 因赛特公司 JAK1 pathway inhibitors for the treatment of gastrointestinal diseases
KR20210137087A (en) 2019-03-05 2021-11-17 인사이트 코포레이션 JAK1 pathway inhibitors for the treatment of chronic lung allograft dysfunction
TW202102222A (en) 2019-03-19 2021-01-16 美商英塞特公司 Biomarkers for vitiligo
US20210123931A1 (en) 2019-10-10 2021-04-29 Incyte Corporation Biomarkers for graft-versus-host disease
US20210123930A1 (en) 2019-10-10 2021-04-29 Incyte Corporation Biomarkers for graft-versus-host disease
US20210113566A1 (en) * 2019-10-16 2021-04-22 Incyte Corporation Use of jak1 inhibitors for the treatment of cutaneous lupus erythematosus and lichen planus (lp)
JP2023506118A (en) * 2019-10-16 2023-02-15 インサイト・コーポレイション Use of JAK1 inhibitors for the treatment of cutaneous lupus erythematosus and lichen planus (LP)
KR20220107213A (en) 2019-11-22 2022-08-02 인사이트 코포레이션 Combination therapy comprising an ALK2 inhibitor and a JAK2 inhibitor
US11685731B2 (en) 2020-06-02 2023-06-27 Incyte Corporation Processes of preparing a JAK1 inhibitor
CR20230129A (en) 2020-08-18 2023-07-13 Incyte Corp Process and intermediates for preparing a jak inhibitor
EP4200278A1 (en) 2020-08-18 2023-06-28 Incyte Corporation Process and intermediates for preparing a jak1 inhibitor
KR20230128472A (en) 2020-12-04 2023-09-05 인사이트 코포레이션 JAK Inhibitors Containing Vitamin D Analogs for Treatment of Skin Diseases
KR20230118118A (en) 2020-12-08 2023-08-10 인사이트 코포레이션 JAK1 pathway inhibitors for the treatment of vitiligo
KR20230157307A (en) 2021-01-11 2023-11-16 인사이트 코포레이션 Combination therapy involving JAK pathway inhibitors and ROCK inhibitors
KR20240009964A (en) 2021-05-03 2024-01-23 인사이트 코포레이션 JAK1 pathway inhibitors for the treatment of prurigo nodosum
CN114149437A (en) * 2021-12-24 2022-03-08 安徽大学 Pyrrolopyrimidine five-membered nitrogen heterocyclic derivative and preparation method and application thereof
US20240058343A1 (en) 2022-08-05 2024-02-22 Incyte Corporation Treatment of urticaria using jak inhibitors

Citations (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5521184A (en) 1992-04-03 1996-05-28 Ciba-Geigy Corporation Pyrimidine derivatives and processes for the preparation thereof
WO2000009495A1 (en) 1998-08-11 2000-02-24 Novartis Ag Isoquinoline derivatives with angiogenesis inhibiting activity
WO2000053595A1 (en) 1999-03-06 2000-09-14 Astrazeneca Ab Pyrimidine compounds
WO2001014402A1 (en) 1999-08-19 2001-03-01 Isis Pharmaceuticals, Inc. Antisense modulation of focal adhesion kinase expression
WO2001064655A1 (en) 2000-03-01 2001-09-07 Astrazeneca Ab 2, 4-di(hetero-)arylamino (-oxy)-5-substituted pyrimidines as antineoplastic agents
WO2002000196A2 (en) 2000-06-28 2002-01-03 Smithkline Beecham P.L.C. Wet milling process
WO2003024967A2 (en) 2001-09-19 2003-03-27 Aventis Pharma S.A. Indolizines as kinase protein inhibitors
WO2003037347A1 (en) 2001-10-30 2003-05-08 Novartis Ag Staurosporine derivatives as inhibitors of flt3 receptor tyrosine kinase activity
WO2003048111A1 (en) 2001-11-30 2003-06-12 Teijin Limited Process for producing 5-(3-cyanophenyl)-3-formylbenzoic acid compound
WO2003099771A2 (en) 2002-05-29 2003-12-04 Novartis Ag Diaryl urea derivatives useful for the treatment of protein kinase dependent diseases
WO2004005281A1 (en) 2002-07-05 2004-01-15 Novartis Ag Inhibitors of tyrosine kinases
WO2004046120A2 (en) 2002-11-15 2004-06-03 Vertex Pharmaceuticals Incorporated Diaminotriazoles useful as inhibitors of protein kinases
WO2004056786A2 (en) 2002-12-20 2004-07-08 Pfizer Products Inc. Pyrimidine derivates for the treatment of abnormal cell growth
WO2004080980A1 (en) 2003-03-14 2004-09-23 Novartis Ag 2, 4- di (phenylamino) pyrimidines useful in the treatment of neoplastic diseases, inflammatory and immune system disorders
WO2005028444A1 (en) 2003-09-24 2005-03-31 Novartis Ag 1,4-disubstituted isoquinilone derivatives as raf-kinase inhibitors useful for the treatment of proliferative diseases
WO2006056399A2 (en) 2004-11-24 2006-06-01 Novartis Ag Combinations of jak inhibitors and at least one of bcr-abl, flt-3, fak or raf kinase inhibitors
WO2007070514A1 (en) * 2005-12-13 2007-06-21 Incyte Corporation Heteroaryl substituted pyrrolo[2,3-b]pyridines and pyrrolo[2,3-b]pyrimidines as janus kinase inhibitors
WO2007090748A1 (en) 2006-02-07 2007-08-16 F. Hoffmann-La Roche Ag Benzamide and heteroarene derivatives
WO2007117494A1 (en) * 2006-04-05 2007-10-18 Vertex Pharmaceuticals Incorporated Deazapurines useful as inhibitors of janus kinases
US20090233903A1 (en) * 2008-03-11 2009-09-17 Incyte Corporation Azetidine and cyclobutane derivatives as jak inhibitors
WO2010039939A1 (en) * 2008-10-02 2010-04-08 Incyte Corporation Janus kinase inhibitors for treatment of dry eye and other eye related diseases

Family Cites Families (288)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2985589A (en) 1957-05-22 1961-05-23 Universal Oil Prod Co Continuous sorption process employing fixed bed of sorbent and moving inlets and outlets
US3632836A (en) 1968-10-25 1972-01-04 Dow Chemical Co Solid curable polyepoxides modified with hydrolyzed liquid polyepoxides
US3832460A (en) 1971-03-19 1974-08-27 C Kosti Anesthetic-vasoconstrictor-antihistamine composition for the treatment of hypertrophied oral tissue
US4140755A (en) 1976-02-13 1979-02-20 Hoffmann-La Roche Inc. Sustained release tablet formulations
DE3036390A1 (en) 1980-09-26 1982-05-13 Troponwerke GmbH & Co KG, 5000 Köln Antiinflammatory intermediate 7H-pyrrolo-(2,3-D)-pyrimidine derivs. - prepd. by dealkylation of 7-phenyl:ethyl derivs. by reaction with hydrochloric, phosphoric or poly:phosphoric acid
DE3220113A1 (en) 1982-05-28 1983-12-01 Basf Ag, 6700 Ludwigshafen DIFLUORMETHOXIPHENYLTHIOPHOSPHORSAEUREESTER
US4402832A (en) 1982-08-12 1983-09-06 Uop Inc. High efficiency continuous separation process
US4404335A (en) 1982-08-16 1983-09-13 The Dow Chemical Company Hydrolyzing epoxy resins in absence of solvent and in presence of oxalic acid and a phosphonium compound
US4548990A (en) 1983-08-15 1985-10-22 Ciba-Geigy Corporation Crosslinked, porous polymers for controlled drug delivery
US4498991A (en) 1984-06-18 1985-02-12 Uop Inc. Serial flow continuous separation process
NL8403224A (en) 1984-10-24 1986-05-16 Oce Andeno Bv DIOXAPHOSPHORINANS, THEIR PREPARATION AND THE USE FOR SPLITTING OF OPTICALLY ACTIVE COMPOUNDS.
CA1306260C (en) 1985-10-18 1992-08-11 Shionogi & Co., Ltd. Condensed imidazopyridine derivatives
JPH0710876Y2 (en) 1989-08-31 1995-03-15 石垣機工株式会社 Cleaning device for dehydration cylinder in screw press
AU645504B2 (en) 1989-10-11 1994-01-20 Teijin Limited Bicyclic pyrimidine derivative, method of producing the same, and pharmaceutical preparation containing the same as active ingredient
US5403593A (en) 1991-03-04 1995-04-04 Sandoz Ltd. Melt granulated compositions for preparing sustained release dosage forms
IT1258781B (en) 1992-01-16 1996-02-29 Zambon Spa OPHTHALMIC PHARMACEUTICAL COMPOSITION CONTAINING N-ACETYLCISTEIN AND POLYVINYL ALCOHOL
FR2695126B1 (en) 1992-08-27 1994-11-10 Sanofi Elf Thienyl or pyrrolyl carboxylic acid derivatives, their preparation and medicaments containing them.
AU671491B2 (en) 1992-12-18 1996-08-29 F. Hoffmann-La Roche Ag N-oxycarbonyl substituted 5'-deoxy-5-fluorcytidines
JPH0710876A (en) 1993-06-24 1995-01-13 Teijin Ltd Pyrrolo(2,3-d)pyrimidine having cyclic amino group at 4-position
USH1439H (en) 1993-10-18 1995-05-02 The Dow Chemical Company Method to increase the level of α-glycol in liquid epoxy resin
EP0727217A3 (en) 1995-02-10 1997-01-15 Suntory Ltd Pharmaceutical composition containing god-type ellagitannin as active ingredient
US5856326A (en) 1995-03-29 1999-01-05 Merck & Co., Inc. Inhibitors of farnesyl-protein transferase
IL117580A0 (en) 1995-03-29 1996-07-23 Merck & Co Inc Inhibitors of farnesyl-protein transferase and pharmaceutical compositions containing them
CN1105113C (en) 1995-07-05 2003-04-09 纳幕尔杜邦公司 Fungicidal pyrimidinones
SI9620103A (en) 1995-07-06 1998-10-31 Novartis Ag Pyrrolopyrimidines and processes for the preparation thereof
US5630943A (en) 1995-11-30 1997-05-20 Merck Patent Gesellschaft Mit Beschrankter Haftung Discontinuous countercurrent chromatographic process and apparatus
GB9604361D0 (en) 1996-02-29 1996-05-01 Pharmacia Spa 4-Substituted pyrrolopyrimidine compounds as tyrosine kinase inhibitors
JP2000504023A (en) 1996-04-03 2000-04-04 メルク エンド カンパニー インコーポレーテッド Cancer treatment methods
AU2802297A (en) 1996-04-18 1997-11-07 Merck & Co., Inc. A method of treating cancer
US5795909A (en) 1996-05-22 1998-08-18 Neuromedica, Inc. DHA-pharmaceutical agent conjugates of taxanes
JP2000508335A (en) 1996-05-30 2000-07-04 メルク エンド カンパニー インコーポレーテッド How to treat cancer
US6624138B1 (en) 2001-09-27 2003-09-23 Gp Medical Drug-loaded biological material chemically treated with genipin
WO1998044797A1 (en) 1997-04-07 1998-10-15 Merck & Co., Inc. A method of treating cancer
US6060038A (en) 1997-05-15 2000-05-09 Merck & Co., Inc. Radiolabeled farnesyl-protein transferase inhibitors
US6063284A (en) 1997-05-15 2000-05-16 Em Industries, Inc. Single column closed-loop recycling with periodic intra-profile injection
JP2001513502A (en) 1997-08-11 2001-09-04 ベーリンガー インゲルハイム ファーマシューティカルズ インコーポレイテッド 5,6-Heteroaryldipyrido [2,3-b: 3 ', 2'-f] azepines and their use in the prevention and treatment of HIV infection
US7153845B2 (en) 1998-08-25 2006-12-26 Columbia Laboratories, Inc. Bioadhesive progressive hydration tablets
US6075056A (en) 1997-10-03 2000-06-13 Penederm, Inc. Antifungal/steroid topical compositions
US6025366A (en) 1998-04-02 2000-02-15 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
SK18542000A3 (en) 1998-06-04 2001-12-03 Abbott Laboratories Cell adhesion-inhibiting anti-inflammatory compounds
US6232320B1 (en) 1998-06-04 2001-05-15 Abbott Laboratories Cell adhesion-inhibiting antiinflammatory compounds
ID26698A (en) 1998-06-19 2001-02-01 Pfizer Prod Inc PIROLO COMPOUNDS [2,3-d] PYRIMIDINE
PA8474101A1 (en) 1998-06-19 2000-09-29 Pfizer Prod Inc PYROLEUM [2,3-D] PIRIMIDINE COMPOUNDS
JP2000119271A (en) 1998-08-12 2000-04-25 Hokuriku Seiyaku Co Ltd 1h-imidazopyridine derivative
CA2343148C (en) 1998-09-10 2005-11-15 Nycomed Danmark A/S Quick release pharmaceutical compositions of drug substances
US6375839B1 (en) 1998-10-29 2002-04-23 Institut Francais Du Petrole Process and device for separation with variable-length chromatographic zones
FR2785196B1 (en) 1998-10-29 2000-12-15 Inst Francais Du Petrole METHOD AND DEVICE FOR SEPARATION WITH VARIABLE LENGTH CHROMATOGRAPHIC AREAS
US6413419B1 (en) 1998-10-29 2002-07-02 Institut Francais Du Petrole Process and device for separation with variable-length chromatographic
WO2000051614A1 (en) 1999-03-03 2000-09-08 Merck & Co., Inc. Inhibitors of prenyl-protein transferases
US6217895B1 (en) 1999-03-22 2001-04-17 Control Delivery Systems Method for treating and/or preventing retinal diseases with sustained release corticosteroids
US6239113B1 (en) 1999-03-31 2001-05-29 Insite Vision, Incorporated Topical treatment or prevention of ocular infections
AU3565999A (en) 1999-04-16 2000-11-02 Coelacanth Chemical Corporation Synthesis of azetidine derivatives
US6915718B2 (en) * 1999-09-02 2005-07-12 United Parts Fhs Automobil Systeme Gmbh Selector handle in a motor vehicle
US6921763B2 (en) * 1999-09-17 2005-07-26 Abbott Laboratories Pyrazolopyrimidines as therapeutic agents
CA2387535A1 (en) * 1999-10-13 2001-04-19 Banyu Pharmaceutical Co., Ltd. Substituted imidazolidinone derivatives
US7235258B1 (en) 1999-10-19 2007-06-26 Nps Pharmaceuticals, Inc. Sustained-release formulations for treating CNS-mediated disorders
DK1235830T3 (en) 1999-12-10 2004-03-29 Pfizer Prod Inc Pyrrolo [2,3-d] pyrimidine compounds as protein kinase inhibitors
CN1615873A (en) 1999-12-24 2005-05-18 阿文蒂斯药物有限公司 Azaindoles compound
US7235551B2 (en) 2000-03-02 2007-06-26 Smithkline Beecham Corporation 1,5-disubstituted-3,4-dihydro-1h-pyrimido[4,5-d]pyrimidin-2-one compounds and their use in treating csbp/p38 kinase mediated diseases
DK1142566T3 (en) 2000-04-07 2004-02-09 Medidom Lab Ophthalmological formulations based on cyclosporine, hyaluronic acid and polysorbate
WO2001081345A1 (en) 2000-04-20 2001-11-01 Mitsubishi Pharma Corporation Aromatic amide compounds
SI2223922T1 (en) 2000-04-25 2016-04-29 Icos Corporation Inhibitors of human phosphatidyl-inositol 3-kinase delta
JP4954426B2 (en) 2000-06-16 2012-06-13 キュリス,インコーポレイテッド Angiogenesis regulating composition and use
US7498304B2 (en) 2000-06-16 2009-03-03 Curis, Inc. Angiogenesis-modulating compositions and uses
US6335342B1 (en) 2000-06-19 2002-01-01 Pharmacia & Upjohn S.P.A. Azaindole derivatives, process for their preparation, and their use as antitumor agents
ATE465756T1 (en) 2000-06-23 2010-05-15 Mitsubishi Tanabe Pharma Corp ANTITUMOR EFFECT AMPLIFIER
EE200200711A (en) 2000-06-26 2004-06-15 Pfizer Products Inc. Pyrrolo [2,3-d] pyrimidine compounds as immunosuppressive agents
AU2001278790A1 (en) 2000-08-22 2002-03-04 Hokuriku Seiyaku Co. Ltd 1h-imidazopyridine derivatives
MXPA03005001A (en) 2000-12-05 2003-09-05 Vertex Pharma Inhibitors of c-jun n-terminal kinases (jnk) and other protein kinases.
GB0100622D0 (en) 2001-01-10 2001-02-21 Vernalis Res Ltd Chemical compounds V111
JP2004520347A (en) 2001-01-15 2004-07-08 グラクソ グループ リミテッド Arylpiperidine and piperazine derivatives as inducers of LDL-receptor expression
EP1363702A4 (en) 2001-01-30 2007-08-22 Cytopia Pty Ltd Methods of inhibiting kinases
JP4316893B2 (en) 2001-05-16 2009-08-19 バーテックス ファーマシューティカルズ インコーポレイテッド Inhibitors of Src and other protein kinases
US7301023B2 (en) 2001-05-31 2007-11-27 Pfizer Inc. Chiral salt resolution
GB0115109D0 (en) 2001-06-21 2001-08-15 Aventis Pharma Ltd Chemical compounds
GB0115393D0 (en) 2001-06-23 2001-08-15 Aventis Pharma Ltd Chemical compounds
US6852727B2 (en) 2001-08-01 2005-02-08 Merck & Co., Inc. Benzimisazo[4,5-f]isoquinolinone derivatives
US6429231B1 (en) 2001-09-24 2002-08-06 Bradley Pharmaceuticals, Inc. Compositions containing antimicrobials and urea for the treatment of dermatological disorders and methods for their use
JP2003155285A (en) 2001-11-19 2003-05-27 Toray Ind Inc Cyclic nitrogen-containing derivative
GT200200234A (en) 2001-12-06 2003-06-27 NEW CRYSTAL COMPOUNDS
US6995144B2 (en) 2002-03-14 2006-02-07 Eisai Co., Ltd. Nitrogen containing heterocyclic compounds and medicines containing the same
WO2003088952A1 (en) 2002-04-15 2003-10-30 Adams Laboratories, Inc. Sustained release of guaifenesin combination drugs
TW200403058A (en) 2002-04-19 2004-03-01 Bristol Myers Squibb Co Heterocyclo inhibitors of potassium channel function
US7304061B2 (en) 2002-04-26 2007-12-04 Vertex Pharmaceuticals Incorporated Heterocyclic inhibitors of ERK2 and uses thereof
CA2483084A1 (en) 2002-05-02 2003-11-13 Merck & Co., Inc. Tyrosine kinase inhibitors
CA2484632C (en) 2002-05-07 2012-12-11 Control Delivery Systems, Inc. Processes for forming a drug delivery device
CA2486183C (en) 2002-05-23 2012-01-10 Cytopia Pty Ltd. Protein kinase inhibitors
US7385018B2 (en) 2002-06-26 2008-06-10 Idemitsu Kosan Co., Ltd. Hydrogenated copolymer, process for producing the same, and hot-melt adhesive composition containing the same
GB0215844D0 (en) 2002-07-09 2002-08-14 Novartis Ag Organic compounds
WO2004007472A1 (en) 2002-07-10 2004-01-22 Ono Pharmaceutical Co., Ltd. Ccr4 antagonist and medicinal use thereof
JP2006502183A (en) 2002-09-20 2006-01-19 アルコン,インコーポレイテッド Use of cytokine synthesis inhibitors for the treatment of dry eye disorders
US20040204404A1 (en) 2002-09-30 2004-10-14 Robert Zelle Human N-type calcium channel blockers
CA2506773A1 (en) 2002-11-04 2004-05-21 Vertex Pharmaceuticals Incorporated Heteroaryl-pyramidine derivatives as jak inhibitors
US8034831B2 (en) 2002-11-06 2011-10-11 Celgene Corporation Methods for the treatment and management of myeloproliferative diseases using 4-(amino)-2-(2,6-Dioxo(3-piperidyl)-isoindoline-1,3-dione in combination with other therapies
US20040099204A1 (en) 2002-11-25 2004-05-27 Nestor John J. Sheet, page, line, position marker
EP1572213A1 (en) 2002-11-26 2005-09-14 Pfizer Products Inc. Method of treatment of transplant rejection
TWI335819B (en) 2002-12-24 2011-01-11 Alcon Inc Use of oculosurface selective glucocorticoid in the treatment of dry eye
TW200418806A (en) 2003-01-13 2004-10-01 Fujisawa Pharmaceutical Co HDAC inhibitor
US7167750B2 (en) 2003-02-03 2007-01-23 Enteromedics, Inc. Obesity treatment with electrically induced vagal down regulation
US7407962B2 (en) 2003-02-07 2008-08-05 Vertex Pharmaceuticals Incorporated Heteroaryl compounds useful as inhibitors or protein kinases
US7547794B2 (en) 2003-04-03 2009-06-16 Vertex Pharmaceuticals Incorporated Compositions useful as inhibitors of protein kinases
SE0301372D0 (en) 2003-05-09 2003-05-09 Astrazeneca Ab Novel compounds
SE0301373D0 (en) 2003-05-09 2003-05-09 Astrazeneca Ab Novel compounds
FR2857454B1 (en) 2003-07-08 2006-08-11 Aventis Pasteur DOSAGE OF TECHIC ACIDS OF BACTERIA GRAM +
US20050043346A1 (en) 2003-08-08 2005-02-24 Pharmacia Italia S.P.A. Pyridylpyrrole derivatives active as kinase inhibitors
WO2005020921A2 (en) 2003-08-29 2005-03-10 Exelixis, Inc. C-kit modulators and methods of use
EP1678147B1 (en) 2003-09-15 2012-08-08 Lead Discovery Center GmbH Pharmaceutically active 4,6-disubstituted aminopyrimidine derivatives as modulators of protein kinases
EP1679074B1 (en) 2003-10-24 2010-12-08 Santen Pharmaceutical Co., Ltd. Therapeutic agent for keratoconjunctive disorder
US7387793B2 (en) 2003-11-14 2008-06-17 Eurand, Inc. Modified release dosage forms of skeletal muscle relaxants
MY141220A (en) 2003-11-17 2010-03-31 Astrazeneca Ab Pyrazole derivatives as inhibitors of receptor tyrosine kinases
BRPI0416909A (en) 2003-11-25 2007-01-16 Pfizer Prod Inc atherosclerosis treatment method
CA2549485A1 (en) 2003-12-17 2005-07-07 Pfizer Products Inc. Pyrrolo [2,3-d] pyrimidine compounds for treating transplant rejection
CA2550189A1 (en) 2003-12-19 2005-07-21 Schering Corporation Thiadiazoles as cxc- and cc- chemokine receptor ligands
EP1696920B8 (en) 2003-12-19 2015-05-06 Plexxikon Inc. Compounds and methods for development of ret modulators
CA2548374C (en) 2003-12-23 2014-05-27 Astex Therapeutics Limited Pyrazole derivatives as protein kinase modulators
US20050165029A1 (en) 2004-01-13 2005-07-28 Ambit Biosciences Corporation Pyrrolopyrimidine derivatives and analogs and their use in the treatment and prevention of diseases
US20050277629A1 (en) 2004-03-18 2005-12-15 The Brigham And Women's Hospital, Inc. Methods for the treatment of synucleinopathies (Lansbury)
PL2332940T3 (en) 2004-03-30 2013-03-29 Vertex Pharma Azaindoles useful as inhibitors of JAK and other protein kinases
WO2005117909A2 (en) 2004-04-23 2005-12-15 Exelixis, Inc. Kinase modulators and methods of use
US20060106020A1 (en) 2004-04-28 2006-05-18 Rodgers James D Tetracyclic inhibitors of Janus kinases
US7558717B2 (en) 2004-04-28 2009-07-07 Vertex Pharmaceuticals Incorporated Crystal structure of human JAK3 kinase domain complex and binding pockets thereof
JP2007536310A (en) 2004-05-03 2007-12-13 ノバルティス アクチエンゲゼルシャフト Combination comprising S1P receptor agonist and JAK3 kinase inhibitor
WO2005110410A2 (en) 2004-05-14 2005-11-24 Abbott Laboratories Kinase inhibitors as therapeutic agents
PE20060426A1 (en) 2004-06-02 2006-06-28 Schering Corp TARTARIC ACID DERIVATIVES AS INHIBITORS OF MMPs, ADAMs, TACE AND TNF-alpha
TW200610762A (en) 2004-06-10 2006-04-01 Irm Llc Compounds and compositions as protein kinase inhibitors
JP5315611B2 (en) 2004-06-23 2013-10-16 小野薬品工業株式会社 Compound having S1P receptor binding ability and use thereof
EP1765819B1 (en) 2004-06-30 2014-03-12 Vertex Pharmaceuticals Inc. Azaindoles useful as inhibitors of protein kinases
US7138423B2 (en) 2004-07-20 2006-11-21 Bristol-Myers Squibb Company Arylpyrrolidine derivatives as NK-1 /SSRI antagonists
FR2873691B1 (en) 2004-07-29 2006-10-06 Sanofi Synthelabo AMINO-PIPERIDINE DERIVATIVES, THEIR PREPARATION AND THEIR THERAPEUTIC APPLICATION
WO2006013114A1 (en) 2004-08-06 2006-02-09 Develogen Aktiengesellschaft Use of a timp-2 secreted protein product for preventing and treating pancreatic diseases and/or obesity and/or metabolic syndrome
WO2006022459A1 (en) 2004-08-23 2006-03-02 Mogam Biotechnology Institute Primer and probe for detection of sars coronavirus, kit comprising the primer and/or the probe, and detection method thereof
US20070054916A1 (en) 2004-10-01 2007-03-08 Amgen Inc. Aryl nitrogen-containing bicyclic compounds and methods of use
CA2582985A1 (en) 2004-10-13 2006-04-20 Jin-Jun Liu Disubstituted pyrazolobenzodiazepines useful as inhibitors for cdk2 and angiogesis, and for the treatment of breast, colon, lung and prostate cancer
MY179032A (en) 2004-10-25 2020-10-26 Cancer Research Tech Ltd Ortho-condensed pyridine and pyrimidine derivatives (e.g.purines) as protein kinase inhibitors
UY29177A1 (en) 2004-10-25 2006-05-31 Astex Therapeutics Ltd SUBSTITUTED DERIVATIVES OF PURINA, PURINONA AND DEAZAPURINA, COMPOSITIONS THAT CONTAIN METHODS FOR THEIR PREPARATION AND ITS USES
US7528138B2 (en) 2004-11-04 2009-05-05 Vertex Pharmaceuticals Incorporated Pyrazolo[1,5-a]pyrimidines useful as inhibitors of protein kinases
US7517870B2 (en) 2004-12-03 2009-04-14 Fondazione Telethon Use of compounds that interfere with the hedgehog signaling pathway for the manufacture of a medicament for preventing, inhibiting, and/or reversing ocular diseases related with ocular neovascularization
US20060128803A1 (en) 2004-12-14 2006-06-15 Alcon, Inc. Method of treating dry eye disorders using 13(S)-HODE and its analogs
WO2006067445A2 (en) 2004-12-22 2006-06-29 Astrazeneca Ab Csf-1r kinase inhibitors
AR054416A1 (en) 2004-12-22 2007-06-27 Incyte Corp PIRROLO [2,3-B] PIRIDIN-4-IL-AMINAS AND PIRROLO [2,3-B] PIRIMIDIN-4-IL-AMINAS AS INHIBITORS OF THE JANUS KINASES. PHARMACEUTICAL COMPOSITIONS.
US20090124635A1 (en) 2005-01-20 2009-05-14 Pfizer Inc. Chemical compounds
KR20070104641A (en) 2005-02-03 2007-10-26 버텍스 파마슈티칼스 인코포레이티드 Pyrrolopyrimidines useful as inhibitors of protein kinase
WO2007044050A2 (en) 2005-02-04 2007-04-19 Bristol-Myers Squibb Company 1h-imidazo[4,5-d]thieno[3,2-b]pyridine based tricyclic compounds and pharmaceutical compositions comprising same
BRPI0608513A2 (en) 2005-03-15 2010-01-05 Irm Llc compounds and compositions as protein kinase inhibitors
BRPI0610514A2 (en) 2005-04-05 2016-11-16 Pharmacopeia Inc compound, pharmaceutical composition, and method of treating a disorder
GB0510139D0 (en) 2005-05-18 2005-06-22 Addex Pharmaceuticals Sa Novel compounds B1
MX2007014619A (en) 2005-05-20 2009-02-13 Vertex Pharma Pyrrolopyridines useful as inhibitors of protein kinase.
GB0510390D0 (en) 2005-05-20 2005-06-29 Novartis Ag Organic compounds
WO2006133426A2 (en) 2005-06-08 2006-12-14 Rigel Pharmaceuticals, Inc. Compositions and methods for inhibition of the jak pathway
WO2006136823A1 (en) 2005-06-21 2006-12-28 Astex Therapeutics Limited Heterocyclic containing amines as kinase b inhibitors
CN102603581B (en) 2005-06-22 2015-06-24 普莱希科公司 Pyrrolo[2,3-b] pyridine derivatives as protein kinase inhibitors
CN102127078A (en) 2005-07-14 2011-07-20 安斯泰来制药株式会社 Heterocyclic janus kinase 3 inhibitors
FR2889662B1 (en) 2005-08-11 2011-01-14 Galderma Res & Dev OIL-IN-WATER EMULSION FOR TOPICAL APPLICATION IN DERMATOLOGY
US20070049591A1 (en) 2005-08-25 2007-03-01 Kalypsys, Inc. Inhibitors of MAPK/Erk Kinase
EP1926735A1 (en) 2005-09-22 2008-06-04 Incyte Corporation Tetracyclic inhibitors of janus kinases
AU2006297351A1 (en) 2005-09-30 2007-04-12 Vertex Pharmaceuticals Incorporated Deazapurines useful as inhibitors of janus kinases
WO2007044894A2 (en) 2005-10-11 2007-04-19 Chembridge Research Laboratories, Inc. Cell-free protein expression systems and methods of use thereof
EP1937664B1 (en) 2005-10-14 2011-06-15 Sumitomo Chemical Company, Limited Hydrazide compound and pesticidal use of the same
BRPI0618011A2 (en) 2005-10-28 2011-08-16 Astrazeneca Ab compound or a pharmaceutically acceptable salt thereof, process for the preparation thereof, pharmaceutical composition, use of a compound or a pharmaceutically acceptable salt thereof, and methods for producing an antiproliferative effect and a pro-apoptotic effect in an animal warm-blooded to treat disease and to produce a jak inhibiting effect on a warm-blooded animal
DK1951684T3 (en) 2005-11-01 2016-10-24 Targegen Inc BIARYLMETAPYRIMIDIN kinase inhibitors
CA2629245C (en) 2005-11-21 2016-07-12 Novartis Ag Neuroendocrine tumor treatment
WO2007062459A1 (en) 2005-11-29 2007-06-07 Cytopia Research Pty Ltd Selective kinase inhibitors based on pyridine scaffold
US20130137681A1 (en) 2005-12-13 2013-05-30 Incyte Corporation HETEROARYL SUBSTITUTED PYRROLO[2,3-b]PYRIDINES AND PYRROLO[2,3-b]PYRIMIDINES AS JANUS KINASE INHIBITORS
JP2009521504A (en) 2005-12-22 2009-06-04 スミスクライン・ビーチャム・コーポレイション Akt activity inhibitor
KR20080083680A (en) 2005-12-23 2008-09-18 스미스클라인 비참 코포레이션 Azaindole inhibitors of aurora kinases
JP4643455B2 (en) 2006-01-12 2011-03-02 株式会社ユニバーサルエンターテインメント Game system
BRPI0706537A2 (en) 2006-01-17 2011-03-29 Vertex Pharma azaindoles useful as janus kinases inhibitors
EP1979353A2 (en) 2006-01-19 2008-10-15 OSI Pharmaceuticals, Inc. Fused heterobicyclic kinase inhibitors
JP2009525350A (en) 2006-02-01 2009-07-09 スミスクライン ビーチャム コーポレーション Pyrrolo [2,3, B] pyridine derivatives useful as RAF kinase inhibitors
PL1842534T3 (en) 2006-02-24 2012-05-31 Teva Pharma Metoprolol succinate extended release tablets and methods for their preparation
EP1995246A4 (en) 2006-03-10 2010-11-17 Ono Pharmaceutical Co Nitrogenated heterocyclic derivative, and pharmaceutical agent comprising the derivative as active ingredient
WO2007116866A1 (en) 2006-04-03 2007-10-18 Astellas Pharma Inc. Hetero compound
WO2007116313A2 (en) 2006-04-12 2007-10-18 Pfizer Limited Pyrrolidine derivatives as modulators of chemokine ccr5 receptors
WO2007129195A2 (en) 2006-05-04 2007-11-15 Pfizer Products Inc. 4-pyrimidine-5-amino-pyrazole compounds
WO2007135461A2 (en) 2006-05-18 2007-11-29 Bayer Healthcare Ag Pharmaceutical compositions comprising implitapide and methods of using same
US7691811B2 (en) 2006-05-25 2010-04-06 Bodor Nicholas S Transporter-enhanced corticosteroid activity and methods and compositions for treating dry eye
TWI398252B (en) 2006-05-26 2013-06-11 Novartis Ag Pyrrolopyrimidine compounds and their uses
NZ573174A (en) 2006-06-01 2012-01-12 Msd Consumer Care Inc Sustained release pharmaceutical dosage form containing phenylephrine
US20080021217A1 (en) 2006-07-20 2008-01-24 Allen Borchardt Heterocyclic inhibitors of rho kinase
WO2008013622A2 (en) 2006-07-27 2008-01-31 E. I. Du Pont De Nemours And Company Fungicidal azocyclic amides
WO2008016123A1 (en) 2006-08-03 2008-02-07 Takeda Pharmaceutical Company Limited GSK-3β INHIBITOR
CA2660560A1 (en) 2006-08-16 2008-02-21 Boehringer Ingelheim International Gmbh Pyrazine compounds, their use and methods of preparation
WO2008028937A1 (en) 2006-09-08 2008-03-13 Novartis Ag N-biaryl (hetero) arylsulphonamide derivatives useful in the treatment of diseases mediated by lymphocytes interactions
WO2008035376A2 (en) 2006-09-19 2008-03-27 Council Of Scientific & Industrial Research A novel bio-erodible insert for ophthalmic applications and a process for the preparation thereof
AR063142A1 (en) 2006-10-04 2008-12-30 Pharmacopeia Inc DERIVATIVES OF 2- (BENCIMIDAZOLIL) PURINE AND PURINONES 6-USEFUL SUBSTITUTES AS IMMUNOSUPPRESSORS, AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM.
AR063141A1 (en) 2006-10-04 2008-12-30 Pharmacopeia Inc DERIVATIVES OF 2- (BENZIMIDAZOLIL) PURINA 8- REPLACED FOR IMMUNOSUPPRESSION
US20120225057A1 (en) 2006-10-11 2012-09-06 Deciphera Pharmaceuticals, Llc Methods and compositions for the treatment of myeloproliferative diseases and other proliferative diseases
NZ576234A (en) 2006-11-06 2011-06-30 Supergen Inc Imidazo[1,2-b]pyridazine and pyrazolo[1,5-a]pyrimidine derivatives and their use as protein kinase inhibitors
US20080119496A1 (en) 2006-11-16 2008-05-22 Pharmacopeia Drug Discovery, Inc. 7-Substituted Purine Derivatives for Immunosuppression
JP5572388B2 (en) 2006-11-22 2014-08-13 インサイト・コーポレイション Imidazotriazines and imidazopyrimidines as kinase inhibitors
WO2008067119A2 (en) 2006-11-27 2008-06-05 Smithkline Beecham Corporation Novel compounds
SG177221A1 (en) 2006-12-15 2012-01-30 Abbott Lab Novel oxadiazole compounds
CA2672903C (en) 2006-12-20 2012-10-23 Amgen Inc. Heterocyclic compounds and their use in treating inflammation, angiogenesis and cancer
AU2007338792B2 (en) 2006-12-20 2012-05-31 Amgen Inc. Substituted heterocycles and methods of use
US8513270B2 (en) 2006-12-22 2013-08-20 Incyte Corporation Substituted heterocycles as Janus kinase inhibitors
CA2667072C (en) 2006-12-22 2015-11-24 Sigma-Tau Industrie Farmaceutiche Riunite S.P.A. Gel useful for the delivery of ophthalmic drugs
KR20080062876A (en) 2006-12-29 2008-07-03 주식회사 대웅제약 Novel antifungal triazole derivatives
WO2008082840A1 (en) 2006-12-29 2008-07-10 Abbott Laboratories Pim kinase inhibitors as cancer chemotherapeutics
WO2008082839A2 (en) 2006-12-29 2008-07-10 Abbott Laboratories Pim kinase inhibitors as cancer chemotherapeutics
BRPI0808523A2 (en) 2007-03-01 2014-08-19 Novartis Vaccines & Diagnostic PIM KINASE INHIBITORS AND METHODS OF USE
BRPI0809998B8 (en) 2007-04-03 2021-05-25 Array Biopharma Inc imidazo[1,2-a]pyridine compound as receptor tyrosine kinase inhibitors, their uses, their preparation processes and pharmaceutical compositions
US8188178B2 (en) 2007-05-07 2012-05-29 3M Innovative Properties Company Cold shrinkable article including an epichlorohydrin composition
GB0709031D0 (en) 2007-05-10 2007-06-20 Sareum Ltd Pharmaceutical compounds
EP2155689B1 (en) 2007-05-31 2015-07-08 Boehringer Ingelheim International GmbH Ccr2 receptor antagonists and uses thereof
GB0710528D0 (en) 2007-06-01 2007-07-11 Glaxo Group Ltd Novel compounds
EP3495369B1 (en) 2007-06-13 2021-10-27 Incyte Holdings Corporation Use of salts of the janus kinase inhibitor (r)-3-(4-(7h-pyrrolo[2,3-d]pyrimidin-4-yl)-1h- pyrazol-1-yl)-3- cyclopentylpropanenitrile
CL2008001709A1 (en) 2007-06-13 2008-11-03 Incyte Corp Compounds derived from pyrrolo [2,3-b] pyrimidine, jak kinase modulators; pharmaceutical composition; and use in the treatment of diseases such as cancer, psoriasis, rheumatoid arthritis, among others.
KR20120115413A (en) 2007-07-11 2012-10-17 화이자 인코포레이티드 Pharmaceutical compositions and methods of treating dry eye disorders
AP2010005167A0 (en) 2007-08-01 2010-02-28 Pfizer Pyrazole compounds and their use as RAF inhibitors
WO2009049028A1 (en) 2007-10-09 2009-04-16 Targegen Inc. Pyrrolopyrimidine compounds and their use as janus kinase modulators
CA2743756A1 (en) 2007-11-15 2009-05-22 Musc Foundation For Research Development Inhibitors of pim protein kinases, compositions, and methods for treating cancer
CA2704599C (en) 2007-11-16 2015-05-12 Incyte Corporation 4-pyrazolyl-n-arylpyrimidin-2-amines and 4-pyrazolyl-n-heteroarylpyrimidin-2-amines as janus kinase inhibitors
GB0723815D0 (en) 2007-12-05 2008-01-16 Glaxo Group Ltd Compounds
CA2711384C (en) 2008-01-18 2016-07-26 Institute Of Organic Chemistry And Biochemistry As Cr, V.V.I. Cytostatic 7-deazapurine nucleosides
EA019309B1 (en) 2008-02-04 2014-02-28 Меркьюри Терапьютикс, Инк. Ampk (amp-activated protein kinase) modulators
UY31679A1 (en) 2008-03-03 2009-09-30 PIM KINASE INHIBITORS AND METHODS FOR USE
JP5384611B2 (en) 2008-03-21 2014-01-08 ノバルティス アーゲー Novel heterocyclic compounds and their use
US8344144B2 (en) 2008-06-18 2013-01-01 Merck Sharp & Dohme Corp. Inhibitors of Janus kinases
BRPI0914630A2 (en) 2008-06-26 2019-09-24 Anterios Inc dermal release
TWI461423B (en) 2008-07-02 2014-11-21 Astrazeneca Ab Thiazolidinedione compounds useful in the treatment of pim kinase related conditions and diseases
FR2933409B1 (en) 2008-07-03 2010-08-27 Centre Nat Rech Scient NEW PYRROLO ° 2,3-a! CARBAZOLES AND THEIR USE AS INHIBITORS OF PIM KINASES
WO2010022081A1 (en) 2008-08-19 2010-02-25 Array Biopharma Inc. Triazolopyridine compounds as pim kinase inhibitors
TWI496779B (en) 2008-08-19 2015-08-21 Array Biopharma Inc Triazolopyridine compounds as pim kinase inhibitors
JP4884570B2 (en) 2008-08-20 2012-02-29 ファイザー・インク Pyrrolo [2,3-d] pyrimidine compound
BRPI0918846A2 (en) 2008-09-02 2019-09-24 Novartis Ag heterocyclic kinase inhibitors
EP2342190A1 (en) 2008-09-02 2011-07-13 Novartis AG Bicyclic kinase inhibitors
PT2344474E (en) 2008-09-02 2015-12-28 Novartis Ag Picolinamide derivatives as kinase inhibitors
CA2739466A1 (en) 2008-10-17 2010-04-22 Merck Frosst Canada Ltd. Azetidine derivatives as inhibitors of stearoyl-coenzyme a delta-9 desaturase
JOP20190230A1 (en) 2009-01-15 2017-06-16 Incyte Corp Processes for preparing jak inhibitors and related intermediate compounds
EP2210890A1 (en) 2009-01-19 2010-07-28 Almirall, S.A. Oxadiazole derivatives as S1P1 receptor agonists
US8263601B2 (en) 2009-02-27 2012-09-11 Concert Pharmaceuticals, Inc. Deuterium substituted xanthine derivatives
MX2011012262A (en) 2009-05-22 2012-01-25 Incyte Corp 3-[4-(7h-pyrrolo[2,3-d]pyrimidin-4-yl)-1h-pyrazol-1-yl]octane- or heptane-nitrile as jak inhibitors.
EA025520B1 (en) 2009-05-22 2017-01-30 Инсайт Холдингс Корпорейшн N-(HETERO)ARYL-PYRROLIDINE DERIVATIVES OF PYRAZOL-4-YL-PYRROLO[2,3-d]PYRIMIDINES AND PYRROL-3-YL-PYRROLO[2,3-d]PYRIMIDINES AS JANUS KINASE INHIBITORS
UA110324C2 (en) 2009-07-02 2015-12-25 Genentech Inc Jak inhibitory compounds based on pyrazolo pyrimidine
CA2767079A1 (en) 2009-07-08 2011-01-13 Leo Pharma A/S Heterocyclic compounds as jak receptor and protein tyrosine kinase inhibitors
US20120157500A1 (en) 2009-08-24 2012-06-21 Weikang Tao Jak inhibition blocks rna interference associated toxicities
TW201111385A (en) 2009-08-27 2011-04-01 Biocryst Pharm Inc Heterocyclic compounds as janus kinase inhibitors
TW201113285A (en) 2009-09-01 2011-04-16 Incyte Corp Heterocyclic derivatives of pyrazol-4-yl-pyrrolo[2,3-d]pyrimidines as janus kinase inhibitors
JP5567136B2 (en) 2009-09-08 2014-08-06 エフ・ホフマン−ラ・ロシュ・アクチェンゲゼルシャフト 4-Substituted pyridin-3-yl-carboxamide compounds and methods of use
EP2305660A1 (en) 2009-09-25 2011-04-06 Almirall, S.A. New thiadiazole derivatives
PT2486041E (en) 2009-10-09 2013-11-14 Incyte Corp Hydroxyl, keto, and glucuronide derivatives of 3-(4-(7h-pyrrolo[2,3-d]pyrimidin-4-yl)-1h-pyrazol-1-yl)-3-cyclopentylpropanenitrile
MX2012004020A (en) 2009-10-20 2012-05-08 Cellzome Ltd Heterocyclyl pyrazolopyrimidine analogues as jak inhibitors.
US8671402B2 (en) 2009-11-09 2014-03-11 Bank Of America Corporation Network-enhanced control of software updates received via removable computer-readable medium
EP2332917B1 (en) 2009-11-11 2012-08-01 Sygnis Bioscience GmbH & Co. KG Compounds for PIM kinase inhibition and for treating malignancy
CN102740888B (en) 2009-11-24 2016-10-12 奥尔德生物制药公司 IL-6 antibody and application thereof
EP2506852A4 (en) 2009-12-04 2013-06-19 Univ Texas Interferon therapies in combination with blockade of stat3 activation
JP5739446B2 (en) 2009-12-18 2015-06-24 ファイザー・インク Pyrrolo [2,3-d] pyrimidine compound
CN102712640A (en) 2010-01-12 2012-10-03 弗·哈夫曼-拉罗切有限公司 Tricyclic heterocyclic compounds, compositions and methods of use thereof
SA111320200B1 (en) 2010-02-17 2014-02-16 ديبيوفارم اس ايه Bicyclic Compounds and their Uses as Dual C-SRC / JAK Inhibitors
EA023444B1 (en) 2010-02-18 2016-06-30 Инсайт Холдингс Корпорейшн Cyclobutane and methylcyclobutane derivatives, composition based thereon and methods of use thereof
SI3354652T1 (en) 2010-03-10 2020-08-31 Incyte Holdings Corporation Piperidin-4-yl azetidine derivatives as jak1 inhibitors
KR20130094710A (en) 2010-04-14 2013-08-26 어레이 바이오파마 인크. 5,7-substituted-imidazo[1,2-c]pyrimidines as inhibitors of jak kinases
EP2390252A1 (en) 2010-05-19 2011-11-30 Almirall, S.A. New pyrazole derivatives
SG10201910912TA (en) 2010-05-21 2020-01-30 Incyte Corp Topical Formulation for a JAK Inhibitor
US8637529B2 (en) 2010-06-11 2014-01-28 AbbYie Inc. Pyrazolo[3,4-d]pyrimidine compounds
US9351943B2 (en) 2010-07-01 2016-05-31 Matthew T. McLeay Anti-fibroblastic fluorochemical emulsion therapies
US20130237493A1 (en) 2010-09-30 2013-09-12 Portola Pharmaceuticals, Inc. Combination therapy of 4-(cyclopropylamino)-2-(4-(4-(ethylsulfonyl)piperazin-1-yl)phenylamino)pyrimidine-5-carboxamide and fludarabine
BR112013012502A2 (en) 2010-11-19 2019-03-06 Incyte Corporation substituted cyclobutyl pyrrolopyridine and derivative pyrrolopyrimidine derivatives as jak inhibitors
US9034884B2 (en) 2010-11-19 2015-05-19 Incyte Corporation Heterocyclic-substituted pyrrolopyridines and pyrrolopyrimidines as JAK inhibitors
CN103370068A (en) 2010-12-03 2013-10-23 Ym生物科学澳大利亚私人有限公司 Treatment of JAK2-mediated conditions
WO2012112847A1 (en) 2011-02-18 2012-08-23 Novartis Pharma Ag mTOR/JAK INHIBITOR COMBINATION THERAPY
CN102247368B (en) 2011-05-19 2013-05-29 安徽永生堂药业有限责任公司 Compound acrivastine sustained release tablets, and preparation method thereof
CN102218042A (en) 2011-05-26 2011-10-19 青岛黄海制药有限责任公司 Sustained release tablet of quetiapine fumarate composition and preparation method of sustained release tablet
MY165963A (en) 2011-06-20 2018-05-18 Incyte Holdings Corp Azetidinyl phenyl, pyridyl or pyrazinyl carboxamide derivatives as jak inhibitors
WO2013007768A1 (en) 2011-07-13 2013-01-17 F. Hoffmann-La Roche Ag Tricyclic heterocyclic compounds, compositions and methods of use thereof as jak inhibitors
WO2013007765A1 (en) 2011-07-13 2013-01-17 F. Hoffmann-La Roche Ag Fused tricyclic compounds for use as inhibitors of janus kinases
EP2741747A1 (en) 2011-08-10 2014-06-18 Novartis Pharma AG JAK P13K/mTOR COMBINATION THERAPY
TW201313721A (en) 2011-08-18 2013-04-01 Incyte Corp Cyclohexyl azetidine derivatives as JAK inhibitors
UA111854C2 (en) 2011-09-07 2016-06-24 Інсайт Холдінгс Корпорейшн METHODS AND INTERMEDIATE COMPOUNDS FOR JAK INHIBITORS
US9193733B2 (en) 2012-05-18 2015-11-24 Incyte Holdings Corporation Piperidinylcyclobutyl substituted pyrrolopyridine and pyrrolopyrimidine derivatives as JAK inhibitors
US10155987B2 (en) 2012-06-12 2018-12-18 Dana-Farber Cancer Institute, Inc. Methods of predicting resistance to JAK inhibitor therapy
EP3450434B1 (en) 2012-06-15 2021-02-24 CoNCERT Pharmaceuticals, Inc. Deuterated derivatives of ruxolitinib
EA201590272A1 (en) 2012-07-27 2015-05-29 Рациофарм Гмбх ORAL DOSED FORMS FOR MODIFIED SURVENT CONTAINING RUXOLITINIB
CN102772384A (en) 2012-08-07 2012-11-14 四川百利药业有限责任公司 Minocycline hydrochloride sustained release tablet and preparation method thereof
JP2015526520A (en) 2012-08-31 2015-09-10 プリンシピア バイオファーマ インコーポレイテッド Benzimidazole derivatives as ITK inhibitors
SG10201703533VA (en) 2012-11-01 2017-06-29 Incyte Corp Tricyclic fused thiophene derivatives as jak inhibitors
SG11201503695XA (en) 2012-11-15 2015-06-29 Incyte Corp Sustained-release dosage forms of ruxolitinib
EA030705B1 (en) 2013-03-06 2018-09-28 Инсайт Холдингс Корпорейшн Processes and intermediates for making a jak inhibitor
PE20160126A1 (en) 2013-05-17 2016-02-24 Incyte Corp DERIVATIVES OF BIPIRAZOLE AS JAK INHIBITORS
ES2792549T3 (en) 2013-08-07 2020-11-11 Incyte Corp Sustained-release dosage forms for a JAK1 inhibitor
CN105555313A (en) 2013-08-20 2016-05-04 因赛特公司 Survival benefit in patients with solid tumors with elevated c-reactive protein levels
CN106456773A (en) 2014-02-28 2017-02-22 因赛特公司 Jak1 inhibitors for the treatment of myelodysplastic syndromes
KR20220066179A (en) 2014-04-08 2022-05-23 인사이트 코포레이션 Treatment of b-cell malignancies by a combination jak and pi3k inhibitor
AU2015253192B2 (en) 2014-04-30 2019-05-16 Incyte Holdings Corporation Processes of preparing a JAK1 inhibitor and new forms thereto
EP4233870A3 (en) 2014-05-28 2024-01-24 Onco Tracker, Inc. Anti-cancer effects of jak2 inhibitors in combination with thalidomide derivatives and glucocorticoids
WO2015184305A1 (en) 2014-05-30 2015-12-03 Incyte Corporation TREATMENT OF CHRONIC NEUTROPHILIC LEUKEMIA (CNL) AND ATYPICAL CHRONIC MYELOID LEUKEMIA (aCML) BY INHIBITORS OF JAK1
US10766900B2 (en) 2017-12-29 2020-09-08 Formosa Laboratories, Inc. Baricitinib intermediate, method for forming Baricitinib intermediate, and method for preparing Baricitinib or pharmaceutically acceptable salt thereof
WO2020163653A1 (en) 2019-02-06 2020-08-13 Concert Pharmaceuticals, Inc. Process for preparing enantiomerically enriched jak inhibitors

Patent Citations (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5521184A (en) 1992-04-03 1996-05-28 Ciba-Geigy Corporation Pyrimidine derivatives and processes for the preparation thereof
WO2000009495A1 (en) 1998-08-11 2000-02-24 Novartis Ag Isoquinoline derivatives with angiogenesis inhibiting activity
WO2000053595A1 (en) 1999-03-06 2000-09-14 Astrazeneca Ab Pyrimidine compounds
WO2001014402A1 (en) 1999-08-19 2001-03-01 Isis Pharmaceuticals, Inc. Antisense modulation of focal adhesion kinase expression
WO2001064655A1 (en) 2000-03-01 2001-09-07 Astrazeneca Ab 2, 4-di(hetero-)arylamino (-oxy)-5-substituted pyrimidines as antineoplastic agents
WO2002000196A2 (en) 2000-06-28 2002-01-03 Smithkline Beecham P.L.C. Wet milling process
WO2003024967A2 (en) 2001-09-19 2003-03-27 Aventis Pharma S.A. Indolizines as kinase protein inhibitors
WO2003037347A1 (en) 2001-10-30 2003-05-08 Novartis Ag Staurosporine derivatives as inhibitors of flt3 receptor tyrosine kinase activity
WO2003048111A1 (en) 2001-11-30 2003-06-12 Teijin Limited Process for producing 5-(3-cyanophenyl)-3-formylbenzoic acid compound
WO2003099771A2 (en) 2002-05-29 2003-12-04 Novartis Ag Diaryl urea derivatives useful for the treatment of protein kinase dependent diseases
WO2004005281A1 (en) 2002-07-05 2004-01-15 Novartis Ag Inhibitors of tyrosine kinases
WO2004046120A2 (en) 2002-11-15 2004-06-03 Vertex Pharmaceuticals Incorporated Diaminotriazoles useful as inhibitors of protein kinases
WO2004056786A2 (en) 2002-12-20 2004-07-08 Pfizer Products Inc. Pyrimidine derivates for the treatment of abnormal cell growth
WO2004080980A1 (en) 2003-03-14 2004-09-23 Novartis Ag 2, 4- di (phenylamino) pyrimidines useful in the treatment of neoplastic diseases, inflammatory and immune system disorders
WO2005028444A1 (en) 2003-09-24 2005-03-31 Novartis Ag 1,4-disubstituted isoquinilone derivatives as raf-kinase inhibitors useful for the treatment of proliferative diseases
WO2006056399A2 (en) 2004-11-24 2006-06-01 Novartis Ag Combinations of jak inhibitors and at least one of bcr-abl, flt-3, fak or raf kinase inhibitors
WO2007070514A1 (en) * 2005-12-13 2007-06-21 Incyte Corporation Heteroaryl substituted pyrrolo[2,3-b]pyridines and pyrrolo[2,3-b]pyrimidines as janus kinase inhibitors
WO2007090748A1 (en) 2006-02-07 2007-08-16 F. Hoffmann-La Roche Ag Benzamide and heteroarene derivatives
WO2007117494A1 (en) * 2006-04-05 2007-10-18 Vertex Pharmaceuticals Incorporated Deazapurines useful as inhibitors of janus kinases
US20090233903A1 (en) * 2008-03-11 2009-09-17 Incyte Corporation Azetidine and cyclobutane derivatives as jak inhibitors
WO2010039939A1 (en) * 2008-10-02 2010-04-08 Incyte Corporation Janus kinase inhibitors for treatment of dry eye and other eye related diseases

Non-Patent Citations (37)

* Cited by examiner, † Cited by third party
Title
"Remington's Pharmaceutical Sciences", 1985, MACK PUBLISHING COMPANY, pages: 1418
ADV PHARMACOL., vol. 47, 2000, pages 113 - 74
AGENTS ACTIONS., vol. 38, no. 1-2, January 1993 (1993-01-01), pages 116 - 21
BARABINO ET AL., EXPERIMENTAL EYE RESEARCH, vol. 79, 2004, pages 613 - 621
BOUDNY, V., KOVARIK, J., NEOPLASM., vol. 49, 2002, pages 349 - 355
BOWMAN, T. ET AL., ONCOGENE, vol. 19, 2000, pages 2474 - 2488
BURGER, R. ET AL., HEMATOL J., vol. 2, 2001, pages 42 - 53
COLIGAN, J.E. ET AL.: "Current Protocols in Immunology", vol. 3, WILEY PRESS
DUDLEY, A.C. ET AL., BIOCHEM. J., vol. 390, 2005, pages 427 - 36
FLEX E. ET AL., J EXP MED., vol. 205, 2008, pages 751 - 8
FONESCA, J.E. ET AL., AUTOIMMUNITY REVIEWS, vol. 8, 2009, pages 538 - 42
GRONEBERG, D.A. ET AL., ALLERGY, vol. 58, 2003, pages 1101 - 1113
GUSCHIN, D., N. ET AL., EMBO J, vol. 14, 1995, pages 1421
IMMUNOL TODAY., vol. L9, no. 1, January 1998 (1998-01-01), pages 37 - 44
JAMES, C. ET AL., NATURE, vol. 434, pages 1144 - 1148
JCI, vol. 113, pages 1664 - 1675
JOURNAL OFPHARMACEUTICAL SCIENCE, vol. 66, 1977, pages 2
KAUSHANSKY K, NEJM, vol. 354, 2006, pages 2034 - 45
LEMP: "The Definition and Classification of Dry Eye Disease: Report of the Defmition and Classification Subcommittee of the International Dry Eye Workshop", THE OCULAR SURFACE, vol. 5, no. 2, April 2007 (2007-04-01), pages 75 - 92
LEVIN ET AL., CANCER CELL, vol. 7, 2005, pages 387 - 397
MACCHI P ET AL., NATURE, vol. 377, 1995, pages 65 - 8
MINEGISHI, Y ET AL., IMMUNITY, vol. 25, 2006, pages 745 - 55
MULLIGHAN CG, PROC NATL ACAD SCI U S A., vol. 106, 2009, pages 9414 - 8
ORTMANN, R. A., T. CHENG ET AL., ARTHRITIS RES, vol. 2, no. 1, 2000, pages 16 - 32
PARGANAS E ET AL., CELL, vol. 93, 1998, pages 385 - 95
PARK ET AL., ANALYTICAL BIOCHEMISTRY, vol. 269, 1999, pages 94 - 104
RODIG, S. J., M. A. MERAZ ET AL., CELL, vol. 93, no. 3, 1998, pages 373 - 83
SCHRADER ET AL.: "Developmental Opthalmology", vol. 41, 2008, KARGER, pages: 298 - 312
SMITH ET AL., IMMUNOLOGY AND CELL BIOLOGY, vol. 76, 1998, pages 497 - 512
SMOLEN, J. S. ET AL., LANCET, vol. 371, 2008, pages 987
SRIRAM, K. ET AL., J. BIOL. CHEM., vol. 279, no. 19, 2 March 2004 (2004-03-02), pages 19936 - 47
STAERK, J. ET AL., JBC, vol. 280, pages 41893 - 41899
W.S.S. JEE, W. YAO, J MUSCULOSKEL. NUERON. INTERACT., vol. 1, no. 3, 2001, pages 193 - 207
WAGH ET AL.: "Polymers used in ocular dosage form and drug delivery systems", ASIAN J. PHARM., January 2008 (2008-01-01), pages 12 - 17
WINYARD, P.G., WILLOUGHBY, D.A.: "Methods in Molecular Biology: Vol. 225, Inflammation Protocols.", vol. 225, 2003, HUMANA PRESS
WUTS, GREENE: "Protective Groups in Organic Synthesis", 2007, JOHN WILEY & SONS
YAO ET AL., ARTHRITIS AND RHEUMATISM, vol. 58, no. 6, 2008, pages 3485 - 3497

Cited By (148)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10639310B2 (en) 2005-12-13 2020-05-05 Incyte Corporation Heteroaryl substituted pyrrolo[2,3-b]pyridines and pyrrolo[2,3-b]pyrimidines as Janus kinase inhibitors
US9974790B2 (en) 2005-12-13 2018-05-22 Incyte Corporation Heteroaryl substituted pyrrolo[2,3-B] pyridines and pyrrolo[2,3-B] pyrimidines as janus kinase inhibitors
US8933086B2 (en) 2005-12-13 2015-01-13 Incyte Corporation Heteroaryl substituted pyrrolo[2,3-B]pyridines and pyrrolo[2,3-B]pyrimidines as Janus kinase inhibitors
US10398699B2 (en) 2005-12-13 2019-09-03 Incyte Holdings Corporation Heteroaryl substituted pyrrolo[2,3-b]pyridines and pyrrolo[2,3-b]pyrimidines as janus kinase inhibitors
US9206187B2 (en) 2005-12-13 2015-12-08 Incyte Holdings Corporation Heteroaryl substituted pyrrolo[2,3-B] pyridines and pyrrolo[2,3-B] pyrimidines as Janus kinase
US9079912B2 (en) 2005-12-13 2015-07-14 Incyte Corporation Heteroaryl substituted pyrrolo[2,3-B] pyridines and pyrrolo[2,3-B] pyrimidines as Janus kinase inhibitors
US11744832B2 (en) 2005-12-13 2023-09-05 Incyte Corporation Heteroaryl substituted pyrrolo[2,3-b]pyridines and pyrrolo[2,3-b]pyrimidines as Janus kinase inhibitors
US9662335B2 (en) 2005-12-13 2017-05-30 Incyte Holdings Corporation Heteroaryl substituted pyrrolo[2,3-B] pyridines and pyrrolo[2,3-B] pyrimidines as janus kinase inhibitors
US9814722B2 (en) 2005-12-13 2017-11-14 Incyte Holdings Corporation Heteroaryl substituted pyrrolo[2,3-B] pyridines and pyrrolo[2,3-B] pyrimidines as janus kinase inhibitors
US8946245B2 (en) 2005-12-13 2015-02-03 Incyte Corporation Heteroaryl substituted pyrrolo[2,3-b]pyridines and pyrrolo[2,3-b]pyrimidines as Janus kinase inhibitors
US11331320B2 (en) 2005-12-13 2022-05-17 Incyte Holdings Corporation Heteroaryl substituted pyrrolo[2,3-b]pyridines and pyrrolo[2,3-b]pyrimidines as Janus kinase inhibitors
US8829013B1 (en) 2007-06-13 2014-09-09 Incyte Corporation Salts of the Janus kinase inhibitor (R)-3-(4-(7H-pyrrolo[2,3-D]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile
US11213528B2 (en) 2007-06-13 2022-01-04 Incyte Holdings Corporation Salts of the janus kinase inhibitor (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile
US10610530B2 (en) 2007-06-13 2020-04-07 Incyte Corporation Salts of the Janus kinase inhibitor (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile
US9376439B2 (en) 2007-06-13 2016-06-28 Incyte Corporation Salts of the janus kinase inhibitor (R)-3(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile
US8822481B1 (en) 2007-06-13 2014-09-02 Incyte Corporation Salts of the janus kinase inhibitor (R)-3-(4-(7H-pyrrolo[2,3-d] pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile
US8722693B2 (en) 2007-06-13 2014-05-13 Incyte Corporation Salts of the Janus kinase inhibitor (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile
US10016429B2 (en) 2007-06-13 2018-07-10 Incyte Corporation Salts of the janus kinase inhibitor (R)-3-(4-(7H-pyrrolo[2,3-D]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile
US9623029B2 (en) 2009-05-22 2017-04-18 Incyte Holdings Corporation 3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]octane- or heptane-nitrile as JAK inhibitors
US9216984B2 (en) 2009-05-22 2015-12-22 Incyte Corporation 3-[4-(7H-pyrrolo[2,3-D]pyrimidin-4-yl)-1H-pyrazol-1-yl]octane—or heptane-nitrile as JAK inhibitors
US9334274B2 (en) 2009-05-22 2016-05-10 Incyte Holdings Corporation N-(hetero)aryl-pyrrolidine derivatives of pyrazol-4-yl-pyrrolo[2,3-d]pyrimidines and pyrrol-3-yl-pyrrolo[2,3-d]pyrimidines as janus kinase inhibitors
US9249145B2 (en) 2009-09-01 2016-02-02 Incyte Holdings Corporation Heterocyclic derivatives of pyrazol-4-yl-pyrrolo[2,3-d]pyrimidines as janus kinase inhibitors
US11285140B2 (en) 2010-03-10 2022-03-29 Incyte Corporation Piperidin-4-yl azetidine derivatives as JAK1 inhibitors
US9464088B2 (en) 2010-03-10 2016-10-11 Incyte Holdings Corporation Piperidin-4-yl azetidine derivatives as JAK1 inhibitors
US10695337B2 (en) 2010-03-10 2020-06-30 Incyte Holdings Corporation Piperidin-4-yl azetidine derivatives as JAK1 inhibitors
US9999619B2 (en) 2010-03-10 2018-06-19 Incyte Holdings Corporation Piperidin-4-yl azetidine derivatives as JAK1 inhibitors
US10758543B2 (en) 2010-05-21 2020-09-01 Incyte Corporation Topical formulation for a JAK inhibitor
US11590136B2 (en) 2010-05-21 2023-02-28 Incyte Corporation Topical formulation for a JAK inhibitor
US11219624B2 (en) 2010-05-21 2022-01-11 Incyte Holdings Corporation Topical formulation for a JAK inhibitor
US10869870B2 (en) 2010-05-21 2020-12-22 Incyte Corporation Topical formulation for a JAK inhibitor
US11571425B2 (en) 2010-05-21 2023-02-07 Incyte Corporation Topical formulation for a JAK inhibitor
US10640506B2 (en) 2010-11-19 2020-05-05 Incyte Holdings Corporation Cyclobutyl substituted pyrrolopyridine and pyrrolopyrimidines derivatives as JAK inhibitors
US8933085B2 (en) 2010-11-19 2015-01-13 Incyte Corporation Cyclobutyl substituted pyrrolopyridine and pyrrolopyrimidine derivatives as JAK inhibitors
US9034884B2 (en) 2010-11-19 2015-05-19 Incyte Corporation Heterocyclic-substituted pyrrolopyridines and pyrrolopyrimidines as JAK inhibitors
US9023840B2 (en) 2011-06-20 2015-05-05 Incyte Corporation Azetidinyl phenyl, pyridyl or pyrazinyl carboxamide derivatives as JAK inhibitors
US8691807B2 (en) 2011-06-20 2014-04-08 Incyte Corporation Azetidinyl phenyl, pyridyl or pyrazinyl carboxamide derivatives as JAK inhibitors
US11214573B2 (en) 2011-06-20 2022-01-04 Incyte Holdings Corporation Azetidinyl phenyl, pyridyl or pyrazinyl carboxamide derivatives as JAK inhibitors
US9611269B2 (en) 2011-06-20 2017-04-04 Incyte Corporation Azetidinyl phenyl, pyridyl or pyrazinyl carboxamide derivatives as JAK inhibitors
US10513522B2 (en) 2011-06-20 2019-12-24 Incyte Corporation Azetidinyl phenyl, pyridyl or pyrazinyl carboxamide derivatives as JAK inhibitors
US9359358B2 (en) 2011-08-18 2016-06-07 Incyte Holdings Corporation Cyclohexyl azetidine derivatives as JAK inhibitors
WO2013026025A1 (en) * 2011-08-18 2013-02-21 Incyte Corporation Cyclohexyl azetidine derivatives as jak inhibitors
US9487521B2 (en) 2011-09-07 2016-11-08 Incyte Holdings Corporation Processes and intermediates for making a JAK inhibitor
US9718834B2 (en) 2011-09-07 2017-08-01 Incyte Corporation Processes and intermediates for making a JAK inhibitor
US9394282B2 (en) 2011-09-22 2016-07-19 Merck Sharp & Dohme Corp. Pyrazole carboxamides as Janus kinase inhibitors
JP2014526525A (en) * 2011-09-22 2014-10-06 メルク・シャープ・アンド・ドーム・コーポレーション Pyrazole carboxamides as Janus kinase inhibitors
WO2013041042A1 (en) * 2011-09-22 2013-03-28 Merck Sharp & Dohme Corp. Pyrazole carboxamides as janus kinase inhibitors
US9193733B2 (en) 2012-05-18 2015-11-24 Incyte Holdings Corporation Piperidinylcyclobutyl substituted pyrrolopyridine and pyrrolopyrimidine derivatives as JAK inhibitors
WO2013173720A1 (en) * 2012-05-18 2013-11-21 Incyte Corporation Piperidinylcyclobutyl substituted pyrrolopyridine and pyrrolopyrimidine derivatives as jak inhibitors
US11576864B2 (en) 2012-11-15 2023-02-14 Incyte Corporation Sustained-release dosage forms of ruxolitinib
US10166191B2 (en) 2012-11-15 2019-01-01 Incyte Corporation Sustained-release dosage forms of ruxolitinib
US11896717B2 (en) 2012-11-15 2024-02-13 Incyte Holdings Corporation Sustained-release dosage forms of ruxolitinib
US11576865B2 (en) 2012-11-15 2023-02-14 Incyte Corporation Sustained-release dosage forms of ruxolitinib
US10874616B2 (en) 2012-11-15 2020-12-29 Incyte Corporation Sustained-release dosage forms of ruxolitinib
US11337927B2 (en) 2012-11-15 2022-05-24 Incyte Holdings Corporation Sustained-release dosage forms of ruxolitinib
US9676750B2 (en) 2013-01-14 2017-06-13 Incyte Corporation Bicyclic aromatic carboxamide compounds useful as pim kinase inhibitors
US9849120B2 (en) 2013-01-15 2017-12-26 Incyte Holdings Corporation Thiazolecarboxamides and pyridinecarboxamide compounds useful as Pim kinase inhibitors
US10265307B2 (en) 2013-01-15 2019-04-23 Incyte Corporation Thiazolecarboxamides and pyridinecarboxamide compounds useful as Pim kinase inhibitors
US11229631B2 (en) 2013-01-15 2022-01-25 Incyte Corporation Thiazolecarboxamides and pyridinecarboxamide compounds useful as Pim kinase inhibitors
US10828290B2 (en) 2013-01-15 2020-11-10 Incyte Corporation Thiazolecarboxamides and pyridinecarboxamide compounds useful as pim kinase inhibitors
US10517858B2 (en) 2013-01-15 2019-12-31 Incyte Holdings Corporation Thiazolecarboxamides and pyridinecarboxamide compounds useful as PIM kinase inhibitors
US9550765B2 (en) 2013-01-15 2017-01-24 Incyte Holdings Corporation Thiazolecarboxamides and pyridinecarboxamide compounds useful as Pim kinase inhibitors
EP3985008A1 (en) * 2013-03-06 2022-04-20 Incyte Holdings Corporation Processes and intermediates for making a jak inhibitor
KR20150140287A (en) * 2013-03-06 2015-12-15 인사이트 코포레이션 Processes and intermediates for making a jak inhibitor
WO2014138168A1 (en) * 2013-03-06 2014-09-12 Incyte Corporation Processes and intermediates for making a jak inhibitor
JP2016512196A (en) * 2013-03-06 2016-04-25 インサイト・コーポレイションIncyte Corporation Method for producing JAK inhibitor and intermediate thereof
US9221845B2 (en) 2013-03-06 2015-12-29 Incyte Holdings Corporation Processes and intermediates for making a JAK inhibitor
CN105189509A (en) * 2013-03-06 2015-12-23 因赛特公司 Processes and intermediates for making a JAK inhibitor
TWI634121B (en) * 2013-03-06 2018-09-01 英塞特控股公司 Processes and intermediates for making a jak inhibitor
KR20210038705A (en) * 2013-03-06 2021-04-07 인사이트 홀딩스 코포레이션 Processes and intermediates for making a jak inhibitor
EA030705B1 (en) * 2013-03-06 2018-09-28 Инсайт Холдингс Корпорейшн Processes and intermediates for making a jak inhibitor
US9714233B2 (en) 2013-03-06 2017-07-25 Incyte Corporation Processes and intermediates for making a JAK inhibitor
JP2019023190A (en) * 2013-03-06 2019-02-14 インサイト・ホールディングス・コーポレイションIncyte Holdings Corporation Processes and intermediates for making jak inhibitor
US8987443B2 (en) 2013-03-06 2015-03-24 Incyte Corporation Processes and intermediates for making a JAK inhibitor
EP3489239A1 (en) * 2013-03-06 2019-05-29 Incyte Holdings Corporation Processes and intermediates for making a jak inhibitor
KR102366356B1 (en) 2013-03-06 2022-02-23 인사이트 홀딩스 코포레이션 Processes and intermediates for making a jak inhibitor
KR102236232B1 (en) * 2013-03-06 2021-04-05 인사이트 홀딩스 코포레이션 Processes and intermediates for making a jak inhibitor
US11230548B2 (en) 2013-03-14 2022-01-25 Alkermes Pharma Ireland Limited Prodrugs of fumarates and their use in treating various diseases
US10406133B2 (en) 2013-03-14 2019-09-10 Alkermes Pharma Ireland Limited Prodrugs of fumarates and their use in treating various diseases
US11083703B2 (en) 2013-03-14 2021-08-10 Alkermes Pharma Ireland Limited Prodrugs of fumarates and their use in treating various diseases
US11905298B2 (en) 2013-03-14 2024-02-20 Alkermes Pharma Ireland Limited Prodrugs of fumarates and their use in treating various diseases
US9505776B2 (en) 2013-03-14 2016-11-29 Alkermes Pharma Ireland Limited Prodrugs of fumarates and their use in treating various diseases
US11679092B2 (en) 2013-03-14 2023-06-20 Alkermes Pharma Ireland Limited Prodrugs of fumarates and their use in treating various diseases
US10596140B2 (en) 2013-03-14 2020-03-24 Alkermes Pharma Ireland Limited Prodrugs of fumarates and their use in treating various diseases
US9957265B2 (en) 2013-03-19 2018-05-01 Merck Sharp & Dohme Corp. N-(2-cyano heterocyclyl) pyrazolo pyridones as janus kinase inhibitors
US10059705B2 (en) 2013-03-19 2018-08-28 Merck Sharp & Dohme Corp. Acyclic cyanoethylpyrazolo pyridones as janus kinase inhibitors
US9725445B2 (en) 2013-03-19 2017-08-08 Merck Sharp & Dohme Corp. Cycloalkyl nitrile pyrazolo pyridones as Janus kinase inhibitors
WO2014146246A1 (en) * 2013-03-19 2014-09-25 Merck Sharp & Dohme Corp. Cycloalkyl nitrile pyrazolo pyridones as janus kinase inhibitors
WO2014161046A1 (en) * 2013-04-04 2014-10-09 The Walter And Eliza Hall Institute Of Medical Research Methods of treating diseases characterized by excessive wnt signalling
US11045421B2 (en) 2013-08-07 2021-06-29 Incyte Corporation Sustained release dosage forms for a JAK1 inhibitor
WO2015021153A1 (en) * 2013-08-07 2015-02-12 Incyte Corporation Sustained release dosage forms for a jak1 inhibitor
TWI822248B (en) * 2013-08-07 2023-11-11 美商英塞特控股公司 Sustained release dosage forms for a jak1 inhibitor
US9655854B2 (en) 2013-08-07 2017-05-23 Incyte Corporation Sustained release dosage forms for a JAK1 inhibitor
TWI789516B (en) * 2013-08-07 2023-01-11 美商英塞特公司 Sustained release dosage forms for a jak1 inhibitor
KR102419714B1 (en) 2013-08-07 2022-07-13 인사이트 코포레이션 Sustained release dosage forms for a jak1 inhibitor
US10561616B2 (en) * 2013-08-07 2020-02-18 Incyte Corporation Sustained release dosage forms for a JAK1 inhibitor
EP3721873A1 (en) * 2013-08-07 2020-10-14 Incyte Corporation Sustained release dosage forms for a jak1 inhibitor
IL277554B1 (en) * 2013-08-07 2023-11-01 Incyte Corp Sustained release dosage forms for a jak1 inhibitor
KR20210076186A (en) * 2013-08-07 2021-06-23 인사이트 코포레이션 Sustained release dosage forms for a jak1 inhibitor
IL277554B2 (en) * 2013-08-07 2024-03-01 Incyte Corp Sustained release dosage forms for a jak1 inhibitor
US20170319487A1 (en) * 2013-08-07 2017-11-09 Incyte Corporation Sustained release dosage forms for a jak1 inhibitor
AU2014305989B2 (en) * 2013-08-07 2019-11-28 Incyte Holdings Corporation Sustained release dosage forms for a JAK1 inhibitor
TWI730935B (en) * 2013-08-07 2021-06-21 美商英塞特公司 Sustained release dosage forms for a jak1 inhibitor
AU2019257368B2 (en) * 2013-08-07 2021-02-25 Incyte Holdings Corporation Sustained release dosage forms for a jak1 inhibitor
US10000507B2 (en) 2013-08-23 2018-06-19 Incyte Corporation Furo- and thieno-pyridine carboxamide compounds useful as pim kinase inhibitors
US9604922B2 (en) 2014-02-24 2017-03-28 Alkermes Pharma Ireland Limited Sulfonamide and sulfinamide prodrugs of fumarates and their use in treating various diseases
KR102396717B1 (en) * 2014-04-08 2022-05-11 인사이트 코포레이션 Treatment of b-cell malignancies by a combination jak and pi3k inhibitor
KR20160146778A (en) * 2014-04-08 2016-12-21 인사이트 코포레이션 Treatment of b-cell malignancies by a combination jak and pi3k inhibitor
US9498467B2 (en) 2014-05-30 2016-11-22 Incyte Corporation Treatment of chronic neutrophilic leukemia (CNL) and atypical chronic myeloid leukemia (aCML) by inhibitors of JAK1
US9822124B2 (en) 2014-07-14 2017-11-21 Incyte Corporation Bicyclic heteroaromatic carboxamide compounds useful as Pim kinase inhibitors
US9890162B2 (en) 2014-07-14 2018-02-13 Incyte Corporation Bicyclic aromatic carboxamide compounds useful as pim kinase inhibitors
US9540347B2 (en) 2015-05-29 2017-01-10 Incyte Corporation Pyridineamine compounds useful as Pim kinase inhibitors
WO2016196244A1 (en) 2015-05-29 2016-12-08 Incyte Corporation Pyridineamine compounds useful as pim kinase inhibitors
US9802918B2 (en) 2015-05-29 2017-10-31 Incyte Corporation Pyridineamine compounds useful as Pim kinase inhibitors
WO2017013270A1 (en) 2015-07-23 2017-01-26 Universite De Strasbourg Use of leptin signaling inhibitor for protecting kidneys from patients having ciliopathy
US10336728B2 (en) 2015-09-09 2019-07-02 Incyte Corporation Salts of a Pim kinase inhibitor
US11066387B2 (en) 2015-09-09 2021-07-20 Incyte Corporation Salts of a Pim kinase inhibitor
US11505540B2 (en) 2015-09-09 2022-11-22 Incyte Corporation Salts of a Pim kinase inhibitor
US9862705B2 (en) 2015-09-09 2018-01-09 Incyte Corporation Salts of a pim kinase inhibitor
WO2017044730A1 (en) 2015-09-09 2017-03-16 Incyte Corporation Salts of a pim kinase inhibitor
WO2017059251A1 (en) 2015-10-02 2017-04-06 Incyte Corporation Heterocyclic compounds useful as pim kinase inhibitors
US9920032B2 (en) 2015-10-02 2018-03-20 Incyte Corporation Heterocyclic compounds useful as pim kinase inhibitors
US10450296B2 (en) 2015-10-02 2019-10-22 Incyte Corporation Heterocyclic compounds useful as Pim kinase inhibitors
US11053215B2 (en) 2015-10-02 2021-07-06 Incyte Corporation Heterocyclic compounds useful as Pim kinase inhibitors
US11045455B2 (en) 2015-12-11 2021-06-29 Sichuan Kelun-Biotech Biopharmaceutical Co., Ltd. Azetidine derivative, preparation method therefor, and use thereof
CN106905322A (en) * 2016-01-26 2017-06-30 杭州华东医药集团新药研究院有限公司 Pyrrolopyrimidine penta azacyclo derivative and its application
CN106905322B (en) * 2016-01-26 2019-10-15 杭州华东医药集团新药研究院有限公司 Pyrrolopyrimidine penta azacyclo derivative and its application
WO2017162604A1 (en) 2016-03-21 2017-09-28 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for diagnosis and treatment of solar lentigo
WO2018167283A1 (en) 2017-03-17 2018-09-20 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for the diagnosis and treatment of pancreatic ductal adenocarcinoma associated neural remodeling
US11278541B2 (en) 2017-12-08 2022-03-22 Incyte Corporation Low dose combination therapy for treatment of myeloproliferative neoplasms
US10596161B2 (en) 2017-12-08 2020-03-24 Incyte Corporation Low dose combination therapy for treatment of myeloproliferative neoplasms
US11384064B2 (en) 2018-01-26 2022-07-12 Rapt Therapeutics, Inc. Chemokine receptor modulators and uses thereof
EP4086245A1 (en) * 2018-01-30 2022-11-09 Incyte Corporation Processes for preparing intermediates for the synthesis of a jak inhibitor
US10899736B2 (en) 2018-01-30 2021-01-26 Incyte Corporation Processes and intermediates for making a JAK inhibitor
TWI797242B (en) * 2018-01-30 2023-04-01 美商英塞特公司 Processes and intermediates for making a jak inhibitor
WO2019152374A1 (en) * 2018-01-30 2019-08-08 Incyte Corporation Processes for preparing (1 -(3-fluoro-2-(trifluoromethyl)isonicotinyl)piperidine-4-one)
WO2019171326A1 (en) 2018-03-08 2019-09-12 Novartis Ag Use of an anti-p-selectin antibody
US11304949B2 (en) 2018-03-30 2022-04-19 Incyte Corporation Treatment of hidradenitis suppurativa using JAK inhibitors
WO2020039401A1 (en) 2018-08-24 2020-02-27 Novartis Ag Treatment comprising il-1βeta binding antibodies and combinations thereof
WO2020092015A1 (en) 2018-11-02 2020-05-07 University Of Rochester Therapeutic mitigation of epithelial infection
WO2020182159A1 (en) 2019-03-14 2020-09-17 上海华汇拓医药科技有限公司 Jak kinase inhibitor, preparation method for same, and applications thereof in field of medicine
WO2020256739A1 (en) 2019-06-21 2020-12-24 Constellation Pharmaceuticals, Inc. Methods of treating myeloproliferative disorders
WO2020257644A1 (en) 2019-06-21 2020-12-24 Constellation Pharmaceuticals, Inc. Methods of treating myeloproliferative disorders
WO2021053490A1 (en) 2019-09-16 2021-03-25 Novartis Ag Use of high-affinity, ligand-blocking, humanized anti-t-cell immunoglobulin domain and mucin domain-3 (tim-3) igg4 antibody for the treatment of myelofibrosis
WO2021053489A1 (en) 2019-09-16 2021-03-25 Novartis Ag Use of an mdm2 inhibitor for the treatment of myelofibrosis
WO2021091535A1 (en) 2019-11-05 2021-05-14 Constellation Pharmaceuticals, Inc. Treating myeloproliferative disorders with cpi-0610 and a jak inhibitor
US11833155B2 (en) 2020-06-03 2023-12-05 Incyte Corporation Combination therapy for treatment of myeloproliferative neoplasms
WO2022074600A1 (en) 2020-10-08 2022-04-14 Novartis Ag Use of an erk inhibitor for the treatment of myelofibrosis
US11878976B2 (en) 2021-03-04 2024-01-23 Eli Lilly And Company FGFR3 inhibitor compounds

Also Published As

Publication number Publication date
TW201206923A (en) 2012-02-16
EP4036088B1 (en) 2024-04-03
AU2011224484A1 (en) 2012-09-27
ECSP12012218A (en) 2012-11-30
PH12015502575A1 (en) 2017-04-24
TWI592413B (en) 2017-07-21
KR102283091B1 (en) 2021-07-30
KR20200033996A (en) 2020-03-30
JP2013522214A (en) 2013-06-13
EP3715347B1 (en) 2021-12-22
JP6158282B2 (en) 2017-07-05
MX354212B (en) 2018-02-19
EP3050882A1 (en) 2016-08-03
CN102985417B (en) 2015-01-28
ZA201207420B (en) 2014-03-26
US20230043959A1 (en) 2023-02-09
MX2019005232A (en) 2019-08-05
LT3354652T (en) 2020-09-25
PT3050882T (en) 2018-04-16
US20110224190A1 (en) 2011-09-15
HRP20201086T1 (en) 2020-10-30
ES2662588T3 (en) 2018-04-09
RS60680B1 (en) 2020-09-30
TWI643857B (en) 2018-12-11
NO3050882T3 (en) 2018-06-30
HUE028723T2 (en) 2016-12-28
SI3050882T1 (en) 2018-06-29
HK1258929A1 (en) 2019-11-22
IL221823A (en) 2016-10-31
JP5858484B2 (en) 2016-02-10
KR20200124776A (en) 2020-11-03
TW201734014A (en) 2017-10-01
SI2545045T1 (en) 2016-05-31
US11285140B2 (en) 2022-03-29
US10695337B2 (en) 2020-06-30
US20180353499A1 (en) 2018-12-13
ES2796377T3 (en) 2020-11-26
EP2545045B1 (en) 2016-01-06
SI3354652T1 (en) 2020-08-31
SG183551A1 (en) 2012-10-30
MX347851B (en) 2017-05-16
PE20130038A1 (en) 2013-01-28
KR20220015492A (en) 2022-02-08
EP3354652B1 (en) 2020-05-06
US20200397774A1 (en) 2020-12-24
US20170246157A1 (en) 2017-08-31
TW201906613A (en) 2019-02-16
NZ602313A (en) 2014-08-29
JP6318291B2 (en) 2018-04-25
PT3354652T (en) 2020-07-20
HUE037077T2 (en) 2018-08-28
JP2018118992A (en) 2018-08-02
TWI766281B (en) 2022-06-01
PL2545045T3 (en) 2016-08-31
CO6602158A2 (en) 2013-01-18
CN102985417A (en) 2013-03-20
KR102172742B1 (en) 2020-11-02
KR101911697B1 (en) 2018-10-25
EP3715347A1 (en) 2020-09-30
CY1123175T1 (en) 2021-10-29
HRP20180552T1 (en) 2018-06-15
BR112012022513A2 (en) 2016-08-30
MX364636B (en) 2019-05-03
TW202102223A (en) 2021-01-16
PL3354652T3 (en) 2020-11-16
ME03002B (en) 2018-10-20
TWI531572B (en) 2016-05-01
ES2908412T3 (en) 2022-04-29
EP2545045A1 (en) 2013-01-16
CR20120510A (en) 2013-01-11
PH12015502575B1 (en) 2017-04-24
KR20180054890A (en) 2018-05-24
KR20210095229A (en) 2021-07-30
CA2792508C (en) 2018-01-16
KR20130038834A (en) 2013-04-18
RS54823B1 (en) 2016-10-31
US20140275031A1 (en) 2014-09-18
JP2017149779A (en) 2017-08-31
KR20180117206A (en) 2018-10-26
EP3050882B1 (en) 2018-01-31
LT3050882T (en) 2018-06-11
ME02386B (en) 2016-09-20
JP2016094443A (en) 2016-05-26
MX2012010344A (en) 2012-11-16
DK3354652T3 (en) 2020-05-18
JP2023099660A (en) 2023-07-13
EA201290894A1 (en) 2013-04-30
KR101857680B1 (en) 2018-05-14
MY192255A (en) 2022-08-11
RS57219B1 (en) 2018-07-31
EP3354652A1 (en) 2018-08-01
HRP20160326T1 (en) 2016-05-20
EP4036088A1 (en) 2022-08-03
EA030376B1 (en) 2018-07-31
US9999619B2 (en) 2018-06-19
ES2569539T3 (en) 2016-05-11
HUE049914T2 (en) 2020-11-30
AR081315A1 (en) 2012-08-08
CA2792508A1 (en) 2011-09-15
US8765734B2 (en) 2014-07-01
HK1177741A1 (en) 2013-08-30
TWI694826B (en) 2020-06-01
JP2020114868A (en) 2020-07-30
DK2545045T3 (en) 2016-01-25
KR102354472B1 (en) 2022-01-21
MX336932B (en) 2016-02-08
CY1120154T1 (en) 2018-12-12
PL3050882T3 (en) 2018-08-31
MY175156A (en) 2020-06-11
SMT201600085B (en) 2016-07-01
US9464088B2 (en) 2016-10-11
TW201638088A (en) 2016-11-01
CL2012002501A1 (en) 2012-12-14
TW202304922A (en) 2023-02-01
DK3050882T3 (en) 2018-03-19

Similar Documents

Publication Publication Date Title
US11285140B2 (en) Piperidin-4-yl azetidine derivatives as JAK1 inhibitors
AU2021212085B2 (en) Piperidin-4-yl azetidine derivatives as jak1 inhibitors

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 201180023757.7

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 11711709

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 12012501768

Country of ref document: PH

WWE Wipo information: entry into national phase

Ref document number: 221823

Country of ref document: IL

Ref document number: 2011224484

Country of ref document: AU

Ref document number: MX/A/2012/010344

Country of ref document: MX

ENP Entry into the national phase

Ref document number: 2792508

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2012557194

Country of ref document: JP

Ref document number: 001467-2012

Country of ref document: PE

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 1201004553

Country of ref document: TH

WWE Wipo information: entry into national phase

Ref document number: 12158921

Country of ref document: CO

WWE Wipo information: entry into national phase

Ref document number: 8434/DELNP/2012

Country of ref document: IN

ENP Entry into the national phase

Ref document number: 2011224484

Country of ref document: AU

Date of ref document: 20110309

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2011711709

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 20127026216

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: CR2012-000510

Country of ref document: CR

WWE Wipo information: entry into national phase

Ref document number: 201290894

Country of ref document: EA

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112012022513

Country of ref document: BR

WWE Wipo information: entry into national phase

Ref document number: 12015502575

Country of ref document: PH

WWE Wipo information: entry into national phase

Ref document number: P-2016/0205

Country of ref document: RS

ENP Entry into the national phase

Ref document number: 112012022513

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20120906