HUE028588T2 - Heteroaril-szubsztituált pirrolo[2,3-b]piridinek és pirrolo[2,3-b]pirimidinek mint Janus-kináz inhibitorok - Google Patents

Heteroaril-szubsztituált pirrolo[2,3-b]piridinek és pirrolo[2,3-b]pirimidinek mint Janus-kináz inhibitorok Download PDF

Info

Publication number
HUE028588T2
HUE028588T2 HUE11152677A HUE11152677A HUE028588T2 HU E028588 T2 HUE028588 T2 HU E028588T2 HU E11152677 A HUE11152677 A HU E11152677A HU E11152677 A HUE11152677 A HU E11152677A HU E028588 T2 HUE028588 T2 HU E028588T2
Authority
HU
Hungary
Prior art keywords
pyrazol
pyrrolo
mol
methyl
pyrrole
Prior art date
Application number
HUE11152677A
Other languages
English (en)
Inventor
James D Rodgers
Stacey Shepard
Thomas P Maduskuie
Haisheng Wang
Nikoo Falahatpisheh
Maria Rafalski
Argyrios G Arvanitis
Louis Storace
Ravi Kumar Jalluri
Jordan S Fridman
Krishna Vaddi
Original Assignee
Incyte Holdings Corp
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=37903501&utm_source=***_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=HUE028588(T2) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Incyte Holdings Corp filed Critical Incyte Holdings Corp
Publication of HUE028588T2 publication Critical patent/HUE028588T2/hu

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
    • A61K31/495Heterocyclic 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
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/437Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/57Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone
    • A61K31/573Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone substituted in position 21, e.g. cortisone, dexamethasone, prednisone or aldosterone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • 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
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/04Antipruritics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B59/00Introduction of isotopes of elements into organic compounds ; Labelled organic compounds per se
    • C07B59/002Heterocyclic compounds
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • 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
    • 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
    • C07B2200/05Isotopically modified compounds, e.g. labelled

Description

Description
FIELD OF THE INVENTION
[0001] The present invention provides heteroaryl substituted pyrrolo[2,3-b]pyridines and heteroaryl substituted pyrro-lo[2,3-b]pyrimidines that modulate the activity of Janus kinases and are useful in the treatment of diseases related to activity of Janus kinases including, for example, immune-related diseases, skin disorders, myeloid proliferative disorders, cancer, and other diseases.
BACKGROUND OF THE INVENTION
[0002] Protein kinases (PKs) are a group of enzymes that regulate diverse, important biological processes including cell growth, survival and differentiation, organ formation and morphogenesis, neovascularization, tissue repair and regeneration, among others. Protein kinases exert their physiological functions through catalyzing the phosphorylation of proteins (or substrates) and thereby modulating the cellular activities of the substrates in various biological contexts. In addition to the functions in normal tissues/organs, many protein kinases also play more specialized roles in a host of human diseases including cancer. A subset of protein kinases (also referred to as oncogenic protein kinases), when dysregulated, can cause tumor formation and growth, and further contribute to tumor maintenance and progression (Blume-Jensen P et al, Nature 2001, 411(6835):355-365). Thus far, oncogenic protein kinases represent one of the largest and most attractive groups of protein targets for cancer intervention and drug development.
[0003] Protein kinases can be categorized as receptor type and non-receptor type. Receptor tyrosine kinases (RTKs) have an extracellular portion, a transmembrane domain, and an intracellular portion, while non-receptor tyrosine kinases are entirely intracellular. RTK mediated signal transduction is typically initiated by extracellular interaction with a specific growth factor (ligand), typically followed by receptor dimerization, stimulation of the intrinsic protein tyrosine kinase activity, and receptor transphosphorylation. Binding sites are thereby created for intracellular signal transduction molecules and lead to the formation of complexes with a spectrum of cytoplasmic signaling molecules that facilitate the appropriate cellular response such as cell division, differentiation, metabolic effects, and changes in the extracellular microenvironment [0004] At present, at least nineteen (19) distinct RTK subfamilies have been identified. One RTK subfamily, designated the HER subfamily, includes EGFR, HER2, HER3 and HER4, and bind such ligands as epithelial growth factor (EGF), TGF-a, amphiregulin, ΗΒ-EGF, betacellulin and heregulin. A second family of RTKs, designated the insulin subfamily, includes the INS-R, the IGF-1R and the IR-R. A third family, the "PDGF" subfamily, includes the PDGF alpha and beta receptors, CSFIR, c-kit and FLK-II. Another subfamily of RTKs, referred to as the FLK subfamily, encompasses the Kinase insert Domain-Receptor fetal liver kinase-1 (KDR/FLK-1), the fetal liver kinase 4 (FLK-4) and the fins-like tyrosine kinase 1 (fit-1 ). Two other subfamilies of RTKs have been designated as the FGF receptor family (FGFR1, FGFR2, FGFR3 and FGFR4) and the Met subfamily (c-Met, Ron and Sea). For a detailed discussion of protein kinases, see for example, Blume-Jensen, P. et al., Nature. 2001, 411(6835):355-365, and Manning, G. et al., Science. 2002, 298(5600):1912-1934.
[0005] The non-receptor type of tyrosine kinases is also composed of numerous subfamilies, including Src, Btk, Abl, Fak, and Jak. Each of these subfamilies can be further subdivided into multiple members that have been frequently linked to oncogenesis. The Src family, for example, is the largest and includes Src, Fyn, Lck and Fgr among others. For a detailed discussion of these kinases, see Bolen JB. Nonreceptor tyrosine protein kinases. Oncogene. 1993, 8(8):2025-31.
[0006] A significant number of tyrosine kinases (both receptor and nonreceptor) are associated with cancer (see Madhusudan S, Ganesan TS. Tyrosine kinase inhibitors in cancer therapy. Clin Biochem. 2004, 37(7):618-35.). Clinical studies suggest that overexpression or dysrégulation of tyrosine kinases may also be of prognostic value. For example, members of the HER family of RTKs have been associated with poor prognosis in breast, colorectal, head and neck and lung cancer. Mutation of c-Kit tyrosine kinase is associated with decreased survival in gastrointestinal stromal tumors. In acute myelogenous leukemia, Flt-3 mutation predicts shorter disease free survival. VEGFR expression, which is important for tumor angiogenesis, is associated with a lower survival rate in lung cancer. Tie-1 kinase expression inversely correlates with survival in gastric cancer. BCR-Abl expression is an important predictor of response in chronic myelogenous leukemia and Src tyrosine kinase is an indicator of poor prognosis in all stages of colorectal cancer.
[0007] The immune system responds to injury and threats from pathogens. Cytokines are low-molecular weight polypeptides or glycoproteins that stimulate biological responses in virtually all cell types. For example, cytokines regulate many of the pathways involved in the host inflammatory response to sepsis. Cytokines influence cell differentiation, proliferation and activation, and they can modulate both proinflammatory and anti-inflammatory responses to allow the host to react appropriately to pathogens.
[0008] Binding of a cytokine to its cell surface receptor initiates intracellular signaling cascades that transduce the extracellular signal to the nucleus, ultimately leading to changes in gene expression. The pathway involving the Janus kinase family of protein tyrosine kinases (JAKs) and Signal Transducers and Activators of Transcription (STATs) is engaged in the signaling of a wide range of cytokines. Generally, cytokine receptors do not have intrinsic tyrosine kinase activity, and thus require receptor-associated kinases to propagate a phosphorylation cascade. JAKs fulfill this function. Cytokines bind to their receptors, causing receptor dimerization, and this enables JAKs to phosphorylate each other as well as specific tyrosine motifs within the cytokine receptors. STATs that recognize these phosphotyrosine motifs are recruited to the receptor, and are then themselves activated by a JAK-dependent tyrosine phosphorylation event. Upon activation, STATs dissociate from the receptors, dimerize, and translocate to the nucleus to bind to specific DNA sites and alter transcription (Scott, M. J., C. J. Godshall, et al. (2002). "Jaks, STATs, Cytokines, and Sepsis." Clin Diagn Lab Immunol 9(6): 1153-9).
[0009] The JAK family plays a role in the cytokine-dependent regulation of proliferation and function of cells involved in immune response. Currently, there are four known mammalian JAK family members: JAK1 (also known as Janus kinase-1 ), JAK2 (also known as Janus kinase-2), JAK3 (also known as Janus kinase, leukocyte; JAKL; L-JAK and Janus kinase-3) and TYK2 (also known as protein-tyrosine kinase 2). The JAK proteins range in size from 120 to 140 kDa and comprise seven conserved JAK homology (JH) domains; one of these is a functional catalytic kinase domain, and another is a pseudokinase domain potentially serving a regulatory function and/or serving as a docking site for STATs (Scott, Godshall et al. 2002, supra).
[0010] While JAKI, JAK2 and TYK2 are ubiquitously expressed, JAK3 is reported to be preferentially expressed in natural killer (NK) cells and not resting T cells, suggesting a role in lymphoid activation (Kawamura, M., D. W. McVicar, et al. (1994). "Molecular cloning of L-JAK, a Janus family protein-tyrosine kinase expressed in natural killer cells and activated leukocytes." Proc Natl Acad Sei USA 91(14): 6374-8).
[0011] Not only do the cytokine-stimulated immune and inflammatory responses contribute to normal host defense, they also play roles in the pathogenesis of diseases: pathologies such as severe combined immunodeficiency (SCID) arise from hypoactivity and suppression of the immune system, and a hyperactive or inappropriate immune/inflammatory response contributes to the pathology of autoimmune diseases such as rheumatoid and psoriatic arthritis, asthma and systemic lupus erythematosus, inflammatory bowel disease, multiple sclerosis, type I diabetes mellitus, myasthenia gravis, thyroiditis, immunoglobulin nephropathies, myocarditis as well as illnesses such as scleroderma and osteoarthritis (Ortmann, R. A., T. Cheng, et al. (2000). "Janus kinases and signal transducers and activators of transcription: their roles in cytokine signaling, development and immunoregulation." Arthritis Res 2(1): 16-32). Furthermore, syndromes with a mixed presentation of autoimmune and immunodeficiency disease are quite common (Candotti, F., L. Notarangelo, et al. (2002). "Molecular aspects of primary immunodeficiencies: lessons from cytokine and other signaling pathways." J Clin Invest 109(10): 1261-9). Thus, therapeutic agents are typically aimed at augmentation or suppression of the immune and inflammatory pathways, accordingly.
[0012] Deficiencies in expression of JAK family members are associated with disease states. Jaki-/- mice are runted at birth, fail to nurse, and die perinatally (Rodig, S. J., M. A. Meraz, etal. (1998). "Disruption of the Jaki gene demonstrates obligatory and nonredundant roles of the Jaks in cytokine-induced biologic responses." Cell 93(3): 373-83). Jak2-/-mouse embryos are anemic and die around day 12.5 postcoitum due to the absence of definitive erythropoiesis. JAK2-deficient fibroblasts do not respond to IFN gamma, although responses to IFNalpha/beta and IL-6 are unafFected. JAK2 functions in signal transduction of a specific group of cytokine receptors required in definitive erythropoiesis (Neubauer, H., A. Cumano, et al. (1998). Cell 93(3): 397-409; Parganas, E., D. Wang, et al. (1998). Cell 93(3): 385-95.). JAK3 appears to play a role in normal development and function of B and T lymphocytes. Mutations of JAK3 are reported to be responsible for autosomal recessive severe combined immunodeficiency (SCID) in humans (Candotti, F., S. A. Oakes, et al. (1997). "Structural and functional basis for JAK3-deficient severe combined immunodeficiency." Blood 90(10): 3996-4003).
[0013] The JAK/STAT pathway, and in particular all four members of the JAK family, are believed to play a role in the pathogenesis of the asthmatic response, chronic obstructive pulmonary disease, bronchitis, and other related inflammatory diseases of the lower respiratory tract. For instance, the inappropriate immune responses that characterize asthma are orchestrated by a subset of CD4+T helper cells termed T helper 2 (Th2) cells. Signaling through the cytokine receptor IL-4 stimulates JAK1 and JAK3 to activate STAT6, and signaling through IL-12 stimulates activation of JAK2 and TYK2, and subsequent phosphorylation of STAT4. STAT4 and STAT6 control multiple aspects of CD4+ T helper cell differentiation (Pernis, A. B. and P. B. Rothman (2002). "JAK-STAT signaling in asthma." J Clin Invest 109(10): 1279-83). Furthermore, TYK2-deficient mice were found to have enhanced Th2 cell-mediated allergic airway inflammation (Seto, Y., H. Nakajima, et al. (2003). "Enhanced Th2 cell-mediated allergic inflammation in Tyk2-deficient mice." J Immunol 170(2): 1077-83). Moreover, multiple cytokines that signal through JAK kinases have been linked to inflammatory diseases or conditions of the upper respiratory tract such as those affecting the nose and sinuses (e.g. rhinitis, sinusitis) whether classically allergic reactions or not.
[0014] The JAK/STAT pathway has also been implicated to play a role in inflammatory diseases/conditions of the eye including, but not limited to, iritis, uveitis, scleritis, conjunctivitis, as well as chronic allergic responses. Therefore, inhibition of JAK kinases may have a beneficial role in the therapeutic treatment of these diseases.
[0015] The JAK/STAT pathway, and in particular, JAK3, also plays a role in cancers of the immune system. In adult T cell leukemia/lymphoma (ATLL), human CD4+ T cells acquire a transformed phenotype, an event that correlates with acquisition of constitutive phosphorylation of JAKs and STATs. Furthermore, an association between JAK3 and STAT-1, STAT-3, and STAT-5 activation and cell-cycle progression was demonstrated by both propidium iodide staining and bromodeoxyuridine incorporation in cells of four ATLL patients tested. These results imply that JAK/STAT activation is associated with replication of leukemic cells and that therapeutic approaches aimed at JAK/STAT inhibition may be considered to halt neoplastic growth (Takemoto, S., J. C. Mulloy, et al. (1997). "Proliferation of adult T cell leukemia/lymphoma cells is associated with the constitutive activation of JAK/STAT proteins." Proc Natl Acad Sei USA 94(25): 13897-902).
[0016] Blocking signal transduction at the level of the JAK kinases holds promise for developing treatments for human cancers. Cytokines of the interleukin 6 (IL-6) family, which activate the signal transducer gp130, are major survival and growth factors for human multiple myeloma (MM) cells. The signal transduction of gp130 is believed to involve JAK1, JAK2 and Tyk2 and the downstream effectors STAT3 and the mitogen-activated protein kinase (MAPK) pathways. In IL-6-dependent MM cell lines treated with the JAK2 inhibitor tyrphostin AG490, JAK2 kinase activity and ERK2 and STAT3 phosphorylation were inhibited. Furthermore, cell proliferation was suppressed and apoptosis was induced (De Vos, J., M. Jourdan, et al. (2000). "JAK2 tyrosine kinase inhibitor tyrphostin AG490 downregulates the mitogen-activated protein kinase (MAPK) and signal transducer and activator of transcription (STAT) pathways and induces apoptosis in myeloma cells." BrJ Haematol 109(4): 823-8). However, in some cases, AG490 can induce dormancy of tumor cells and actually then protect them from death.
[0017] Activation of JAK/STAT in cancers may occur by multiple mechanisms including 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). Importantly, 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). Moreover, elevated levels of circulating cytokines that signal through JAK/STAT may adversely impact patient health as they are thought to play a causal role in cachexia and/or chronic fatigue. As such, JAK inhibition may be therapeutic for the treatment of cancer patients for reasons that extend beyond potential anti-tumor activity. The cachexia indication may gain further mechanistic support with realization that the satiety factor leptin signals through JAKs.
[0018] Pharmacological targeting of Janus kinase 3 (JAK3) has been employed successfully to control allograft rejection and graft versus host disease (GVHD). In addition to its involvement in signaling of cytokine receptors, JAK3 is also engaged in the CD40 signaling pathway of peripheral blood monocytes. During CD40-induced maturation of myeloid dendritic cells (DCs), JAK3 activity is induced, and increases in costimulatory molecule expression, IL-12 production, and potent allogeneic stimulatory capacity are observed. A rationally designed JAK3 inhibitor WHI-P-154 prevented these effects arresting the DCs at an immature level, suggesting that immunosuppressive therapies targeting the tyrosine kinase JAK3 may also affect the function of myeloid cells (Saemann, M. D., C. Diakos, et al. (2003). "Prevention of CD40-triggered dendritic cell maturation and induction of T-cell hyporeactivity by targeting of Janus kinase 3." Am J Transplant 3(11): 1341-9). In the mouse model system, JAK3 was also shown to be an important molecular target for treatment of autoimmune insulin-dependent (type 1) diabetes mellitus. The rationally designed JAK3 inhibitor JANEX-1 exhibited potent immunomodulatory activity and delayed the onset of diabetes in the NOD mouse model of autoimmune type 1 diabetes (Cetkovic-Cvrlje, M., A. L. Dragt, etal. (2003). "Targeting JAK3 with JANEX-1 for prevention of autoimmune type 1 diabetes in NOD mice." Clin Immunol 106(3): 213-25).
[0019] It has been suggested that inhibition of JAK2 tyrosine kinase can be beneficial for patients with myeloproliferative disorder. (Levin, etal., Cancer Cell, vol. 7,2005: 387-397) Myeloproliferative disorder (MPD) includes polycythemia vera (PV), essential thrombocythemia (ET), myeloid metaplasia with myelofibrosis (MMM), chronic myelogenous leukemia (CML), chronic myelomonocytic leukemia (CMML), hyperéosinophilie syndrome (HES) and systemic mast cell disease (SMCD). Although the myeloproliferative disorder (such as PV, ET and MMM) are thought to be caused by acquired somatic mutation in hematopoietic progenitors, the genetic basis for these diseases has not been known. However, it has been reported that hematopoietic cells from a majority of patients with PV and a significant number of patients with ET and MMM possess a recurrent somatic activating mutation in the JAK2 tyrosine kinase. It has also been reported that inhibition of the JAK2V617F kinase with a small molecule inhibitor leads to inhibition of proliferation of hematopoietic cells, suggesting that the JAK2 tyrosine kinase is a potential target for pharmacologic inhibition in patients with PV, ET and MMM.
[0020] Inhibition of the JAK kinases is also envisioned to have therapeutic benefits in patients suffering from skin immune disorders such as psoriasis, and skin sensitization. In psoriasis vulgaris, the most common form of psoriasis, it has been generally accepted that activated T lymphocytes are important for the maintenance of the disease and its associated psoriatic plaques (Gottlieb, A.B., et al, Nat Rev Drug Disc., 4:19-34). Psoriatic plaques contain a significant immune infiltrate, including leukocytes and monocytes, as well as multiple epidermal layers with increased kératinocyte proliferation. While the initial activation of immune cells in psoriasis occurs by an ill defined mechanism, the maintenance is believed to be dependent on a number of inflammatory cytokines, in addition to various chemokines and growth factors (JCI, 113:1664-1675). Many of these, including interleukins -2, -4, -6, -7, -12, -15, -18, and -23 as well as GM-CSF and IFNg, signal through the Janus (JAK) kinases (Adv Pharmacol. 2000;47:113-74). As such, blocking signal transduction at the level of JAK kinases may result in therapeutic benefits in patients suffering from psoriasis or other immune disorders of the skin.
[0021] It has been known that certain therapeutics can cause immune reactions such as skin rash or diarrhea in some patients. For instance, administration of some of the new targeted anti-cancer agents such as Iressa, Erbitux, and Tarceva has induced acneiform rash with some patients. Another example is that some therapeutics used topically induce skin irritation, skin rash, contact dermatitis or allergic contact sensitization. For some patients, these immune reactions may be bothersome, but for others, the immune reactions such as rash or diarrhea may result in inability to continue the treatment. Although the driving force behind these immune reactions has not been elucidated completely at the present time, these immune reactions are likely linked to immune infiltrate.
[0022] Inhibitors of Janus kinases or related kinases are widely sought and several publications report effective classes of compounds. For example, certain inhibitors are reported in WO 99/65909, US 2004/0198737; WO 2004/099204; W02004/099205; and WO 01/42246. Heteroaryl substituted pyrroles and other compounds are reported in WO 2004/72063 and WO 99/62908.
[0023] WO 2006/127587 discloses pyrrolopyridines useful as inhibitors of protein kinase. WO 2006/096270 discloses pyrrolopyrimidines useful as inhibitors of protein kinase. WO 2005/013986 discloses pyridylpyrrole derivatives active as kinase inhibitors. US 2005/153989 discoses pyrrolopyrimidine derivatives and their use in the treatment and prevention of diseases. WO 02/00661 discloses pyrrolo[2,3-d]pyrimidine compounds as immunosuppressive agents.
[0024] Thus, new or improved agents which inhibit kinases such as Janus kinases are continually needed that act as immunosuppressive agents for organ transplants, as well as agents for the prevention and treatment of autoimmune diseases (e.g., multiple sclerosis, rheumatoid arthritis, asthma, type 1 diabetes, inflammatory bowel disease, Crohn’s disease, autoimmune thyroid disorders, Alzheimer’s disease), 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, to name a few. The compounds, compositions and methods described herein are directed toward these needs and other ends.
SUMMARY OF THE INVENTION
[0025] The present invention provides compounds of Formula I:
or pharmaceutically acceptable salt forms thereof, wherein constituent members are defined herein, for use in a method of treating a disease selected from cachexia, a myeloproliferative disorder, and cancer in a patient in need thereof, in combination with an additional pharmaceutical agent.
DETAILED DESCRIPTION
[0026] The present invention provides, inter alia, compounds that modulate the activity of one or more JAKs and are useful, for example, in the treatment of diseases associated with JAK expression or activity. The compounds of the invention have Formula I:
including pharmaceutically acceptable salt forms thereof, wherein the compounds are defined as set out in the claims.
[0027] In the claimed compounds A1 is C, A2 is N, and R1, R2 and R3 are each H. Accordingly, the compounds of
Formula I correspond to compounds of formula:
[0028] In some embodiments, when X is N, the 5-membered rina formed hv δι „o..~ .... mnom i , ,. , v. ., yA , A2, U, T, and V is other than pyrrolyl.
[0029] In some embodiments, X is N. 1 [0030] In some embodiments, X is CR4.
[0031]
In some embodiments, the 5-membered ring formed by A1, A2 U T
[0032] In some embodiments, the 5-membered ring formed by A1 A2 U T ,, 'S ^rro^’ Py1"320^’ or imidazolyl. a y ’ A ’ u’ r' and V is selected from:
wherein: a designates the site of attachment of moiety -(Y)n-Z; b designates the site of attachment to the core moiety:
[0033] In some embodiments, n is 0.
[0034] In some embodiments, n is 1.
[0035] In some embodiments, n is 1 and Y is C-^g alkylene, (CR11R12)pC(0)(CR11R12)q, (CR11R12)pC(0)NRc(CR11R12)q, (CR11R12)pC(0)0(CR11R12)q, wherein said C^g alkylene is optionally substituted with 1,2, or 3 halo, OH, CN, amino, alkylamino, or C2_8 dialkylamino.
[0036] In some embodiments, n is 1 and Y is C^g alkylene optionally substituted with 1, 2, or 3 halo, OH, CN, amino, 0^4 alkylamino, or C2_8 dialkylamino.
[0037] In some embodiments, n is 1 and Y is ethylene optionally substituted with 1,2, or 3 halo, OH, CN, amino, C^ alkylamino, or C2_g dialkylamino.
[0038] In some embodiments, n is 1 and Y is (CR11R12)pC(0)(CR11R12)q (CR11R12)pC(0)NRc-(CR11R12)q, or (CR11 R12)pC(0)0(CR11 R12)q.
[0039] In some embodiments, p is 0.
[0040] In some embodiments, p is 1.
[0041] In some embodiments, p is 2.
[0042] In some embodiments, q is 0.
[0043] In some embodiments, q is 1.
[0044] In some embodiments, q is 2.
[0045] In some embodiments, one of p and q is 0 and the other of p and q is 1,2, or 3.
[0046] In some embodiments, Z is aryl, cycloalkyl, heteroaryl, or heterocycloalkyl, each optionally substituted with 1, 2, 3, 4, 5, or 6 substituents selected from halo, Cf.4 alkyl, C2^ alkenyl, C2^ alkynyl, C-^ haloalkyl, C1-4 hydroxyalkyl, C.|_4 cyanoalkyl, Cy1, CN, N02, ORa, SRa, C(0)Rb, C(0)NRcRd, C(0)ORa, OC(0)Rb, OC(0)NRcRd, NRcRd, NRcC(0)Rb, NRcC(0)NRcRd, NRcC(0)ORa, C(=NR')NRcRd, NRcC(=NR')NRcRd, S(0)Rb, S(0)NRcRd, S(0)2Rb, NRcS(0)2Rb, and S(0)2NRcRd.
[0047] In some embodiments, Z is aryl or heteroaryl, each optionally substituted with 1, 2, 3, 4, 5, or 6 substituents independently selected from halo, Ολ_4 alkyl, C2^ alkenyl, C2_4 alkynyl, C1_4 haloalkyl, halosulfanyl, C1-4 hydroxyalkyl, C1_4 cyanoalkyl, Cy1, CN, N02, ORa, SRa, C(0)Rb, C(0)NRcRd, C(0)ORa, OC(0)Rb, OC(0)NRcRd, NRcRd, NRcC(0)Rb, NRcC(0)NRcRd, NRcC(0)ORa, S(0)Rb, S(0)NRcRd, S(0)2Rb, NRcS(0)2Rb, and S(0)2NRcRd.
[0048] In some embodiments, Z is aryl or heteroaryl, each optionally substituted with 1, 2, 3, 4, 5, or 6 substituents independently selected from halo, C1.4 alkyl, C2^ alkenyl, C2_4 alkynyl, C1-4 haloalkyl, C1-4 hydroxyalkyl, C1-4 cyanoalkyl, Cy1, CN, N02, ORa, SRa, C(0)Rb, C(0)NRcRd, C(0)ORa, OC(0)Rb, OC(0)NRcRd, NRcRd, NRcC(0)Rb, NRcC(0)NRcRd, NRcC(0)ORa, S(0)Rb, S(0)NRcRd, S(0)2Rb, NRcS(0)2Rb, and S(0)2NRcRd.
[0049] In some embodiments, Z is phenyl or 5- or 6-membered heteroaryl, each optionally substituted with 1,2, 3, 4, 5, or 6 substituents independently selected from halo, alkyl, C2_4 alkenyl, C2_4 alkynyl, C1_4 haloalkyl, Ci_4 hydroxyalkyl, C.|_4 cyanoalkyl, Cy1, CN, N02, ORa, SRa, C(0)Rb, C(0)NRcRd, C(0)ORa, OC(0)Rb, OC(0)NRcRd, NRcRd, NRcC(0)Rb, NRcC(0)NRcRd, NRcC(0)ORa, S(0)Rb, S(0)NRcRd, S(0)2Rb, NRcS(0)2Rb, and S(0)2NRcRd.
[0050] In some embodiments, Z is phenyl optionally substituted with 1, 2, 3, 4, 5, or 6 substituents independently selected from halo, C^ alkyl, C2.4 alkenyl, C2.4 alkynyl, C^ haloalkyl, C^ hydroxyalkyl, C^ cyanoalkyl, Cy1, CN, N02, ORa, SRa, C(0)Rb, C(0)NRcRd, C(0)ORa, OC(0)Rb, OC(0)NRcRd, NRcRd, NRcC(0)Rb, NRcC(0)NRcRd, NRcC(0)ORa, S(0)Rb, S(0)NRcRd, S(0)2Rb, NRcS(0)2Rb, and S(0)2NRcRd.
[0051] In some embodiments, Z is cycloalkyl or heterocycloalkyl, each optionally substituted with 1, 2, 3, 4, 5, or 6 substituents independently selected from halo, C^ alkyl, C2^ alkenyl, C2_4 alkynyl, C^ haloalkyl, C1-4 hydroxyalkyl, C.,.4 cyanoalkyl, Cy1, CN, N02, ORa, SRa, C(0)Rb, C(0)NRcRd, C(0)ORa, OC(0)Rb, OC(0)NRcRd, NRcRd, NRcC(0)Rb, NRcC(0)NRcRd, NRcC(0)ORa, S(0)Rb, S(0)NRcRd, S(0)2Rb, NRcS(0)2Rb, and S(0)2NRcRd.
[0052] In some embodiments, Z is substituted with at least one substituent comprising at least one CN group.
[0053] In some embodiments, Cy1 is independently selected from cycloalkyl and heterocycloalkyl, each optionally substituted by 1, 2, 3, 4 or 5 substituents independently selected from halo, C1_4 alkyl, C2_4 alkenyl, C2.4 alkynyl, C^4 haloalkyl, CN, N02, ORa", SRa”, C(0)Rb”, C(0)NRc”Rd", C(0)ORa”, OC(0)Rb", OC(0)NRc"Rd", NRc"Rd", NRc”C(0)Rb", NRc"C(0)ORa", S(0)Rb", S(0)NRc"Rd", S(0)2Rb”, and S(0)2NRc”Rd”.
[0054] In some embodiments, Cy1 is independently selected from cycloalkyl optionally substituted by 1, 2, 3,4 or 5 substituents independently selected from halo, C^ alkyl, C2_4 alkenyl, C2^ alkynyl, C^ haloalkyl, CN, N02, ORa", SRa ", C(0)Rb", C(0)NRc"Rd", C(0)0Ra", 0C(0)Rb”, 0C(0)NRc"Rd", NRc"Rd", NRc"C(0)Rb", NRc”C(0)ORa", S(0)Rb", S(0)NRc"Rd", S(0)2Rb", and S(0)2NRc"Rd".
[0055] In some embodiments, R5 is H, halo, Ci_4 alkyl, C-|_4 haloalkyl, halosulfanyl, CN, or NR9R10.
[0056] In some embodiments, R5 is H, halo, Ci_4 alkyl, C^_4 haloalkyl, CN, or NR9R10.
[0057] In some embodiments, R5 is H.
[0058] In some embodiments, R6 is H or alkyl.
[0059] In some embodiments, R6 is H.
[0060] In some embodiments, the compound has Formula la or lb:
[0061] In some embodiments, the compound has Formula II:
[0062] In the claimed compounds R1, R2 and R3 are each H. Accordingly, the compounds of Formula II correspond to compounds of formula:
[0063] In some embodiments, the compound has Formula Ilia or lllb:
[0064] In some embodiments, the compound has Formula IV:
[0065] In some embodiments, the compound has Formula Va:
[0066] In some embodiments, the compound has Formula Vb:
[0067] In some embodiments, the compound has Formula Via:
[0068] In some embodiments, the compound has Formula Vlb:
[0069] 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 "C^g alkyl" is specifically intended to individually disclose methyl, ethyl, C3 alkyl, C4 alkyl, C5 alkyl, and C6 alkyl.
[0070] 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.
[0071] 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 understood that the "alkyl" or "aryl" represents a linking alkylene group or arylene group, respectively.
[0072] 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 and 1,2,3,4-tetrahydro-naphthalene is an example of a 10-membered cycloalkyl group.
[0073] As used herein, the term "alkyl" is meant to refer to a saturated hydrocarbon group which is straight-chained or branched. Example alkyl groups include methyl (Me), ethyl (Et), propyl (e.g., n-propyl and isopropyl), butyl (e.g., n-butyl, isobutyl, t-butyl), pentyl (e.g., n-pentyl, isopentyl, neopentyl), and the like. An alkyl group can contain from 1 to about 20, from 2 to about 20, from 1 to about 10, from 1 to about 8, from 1 to about 6, from 1 to about 4, or from 1 to about 3 carbon atoms. A linking alkyl group is referred to herein as "alkylene." [0074] As used herein, "alkenyl" refers to an alkyl group having one or more double carbon-carbon bonds. Example alkenyl groups include ethenyl, propenyl, cyclohexenyl, and the like. A linking alkenyl group is referred to herein as "alkenylene." [0075] As used herein, "alkynyl" refers to an alkyl group having one or more triple carbon-carbon bonds. Example alkynyl groups include ethynyl, propynyl, and the like. A linking alkynyl group is referred to herein as "alkynylene." [0076] As used herein, "haloalkyl" refers to an alkyl group having one or more halogen substituents. Example haloalkyl groups include CF3, C2F5, CFIF2, CCI3, CFICI2, C2CI5, and the like.
[0077] As used herein, "halosulfanyl" refers to a sulfur group having one or more halogen substituents. Example halosulfanyl groups include pentahalosulfanyl groups such as SF5.
[0078] As used herein, "aryl" refers to monocyclic or polycyclic (e.g., having 2,3 or 4 fused rings) aromatic hydrocarbons such as, for example, phenyl, naphthyl, anthracenyl, phenanthrenyl, indanyl, indenyl, and the like. In some embodiments, aryl groups have from 6 to about 20 carbon atoms. A linking aryl group is referred to herein as "arylene." [0079] As used herein, "cycloalkyl" refers to non-aromatic cyclic hydrocarbons including cyclized alkyl, alkenyl, and alkynyl groups. Cycloalkyl groups can include mono- or polycyclic (e.g., having 2, 3 or 4 fused rings) groups and spiro-cycles. Ring-forming carbon atoms of a cycloalkyl group can be optionally substituted byoxo orsulfido. Cycloalkyl groups also include cycloalkylidenes. Example cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cy-cloheptyl, cyclopentenyl, cyclohexenyl, cyclohexadienyl, cycloheptatrienyl, norbornyl, norpinyl, norcarnyl, adamantyl, and the like. Also included in the definition of cycloalkyl are moieties that have one or more aromatic rings fused (i.e., having a bond in common with) to the cycloalkyl ring, for example, benzo or thienyl derivatives of pentane, pentene, hexane, and the like. A cycloalkyl group containing a fused aromatic ring can be attached through any ring-forming atom including a ring-forming atom of the fused aromatic ring. A linking cycloalkyl group is referred to herein as "cycloalkylene." [0080] As used herein, "heteroaryl" refers to an aromatic heterocycle having at least one heteroatom ring member such as sulfur, oxygen, or nitrogen. Heteroaryl groups include monocyclic and polycyclic (e.g., having 2, 3 or 4 fused rings) systems. Examples of heteroaryl groups include without limitation, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazinyl, furyl, quinolyl, isoquinolyl, thienyl, imidazolyl, thiazolyl, indolyl, pyrryl, oxazolyl, benzofuryl, benzothienyl, ben-zthiazolyl, isoxazolyl, pyrazolyl, triazolyl, tetrazolyl, indazolyl, 1,2,4-thiadiazolyl, isothiazolyl, benzothienyl, purinyl, car-bazolyl, benzimidazolyl, indolinyl, and the like. In some embodiments, the heteroaryl group has from 1 to about 20 carbon atoms, and in further embodiments from about 3 to about 20 carbon atoms. In some embodiments, the heteroaryl group contains 3 to about 14, 4 to about 14, 3 to about 7, or 5 to 6 ring-forming atoms. In some embodiments, the heteroaryl group has 1 to about 4, 1 to about 3, or 1 to 2 heteroatoms. A linking heteroaryl group is referred to herein as "heter-oarylene." [0081] As used herein, "heterocycloalkyl" refers to non-aromatic heterocycles including cyclized alkyl, alkenyl, and alkynyl groups where one or more of the ring-forming carbon atoms is replaced by a heteroatom such as an Ο, N, or S atom. Heterocycloalkyl groups include monocyclic and polycyclic (e.g., having 2, 3 or 4 fused rings) systems as well as spirocycles. Example "heterocycloalkyl" groups include morpholino, thiomorpholino, piperazinyl, tetrahydrofuranyl, tet-rahydrothienyl, 2,3-dihydrobenzofuryl, 1,3-benzodioxole, benzo-1,4-dioxane, piperidinyl, pyrrolidinyl, isoxazolidinyl, iso-thiazolidinyl, pyrazolidinyl, oxazolidinyl, thiazolidinyl, imidazolidinyl, and the like. Ring-forming carbon atoms and heteroatoms of a heterocycloalkyl group can be optionally substituted by oxo or sulfido. Also included in the definition of heterocycloalkyl are moieties that have one or more aromatic rings fused (i.e., having a bond in common with) to the nonaromatic heterocyclic ring, for example phthalimidyl, naphthalimidyl, and benzo derivatives of heterocycles. The heterocycloalkyl group can be attached through a ring-forming carbon atom or a ring-forming heteroatom. The heterocycloalkyl group containing a fused aromatic ring can be attached through any ring-forming atom including a ring-forming atom of the fused aromatic ring. In some embodiments, the heterocycloalkyl group has from 1 to about 20 carbon atoms, and in further embodiments from about 3 to about 20 carbon atoms. In some embodiments, the heterocycloalkyl group contains 3 to about 14,4 to about 14, 3 to about 7, or 5 to 6 ring-forming atoms. In some embodiments, the heterocycloalkyl group has 1 to about 4, 1 to about 3, or 1 to 2 heteroatoms. In some embodiments, the heterocycloalkyl group contains 0 to 3 double or triple bonds. In some embodiments, the heterocycloalkyl group contains 0 to 2 double or triple bonds. A linking heterocycloalkyl group is referred to herein as "heterocycloalkylene." [0082] As used herein, "halo" or "halogen" includes fluoro, chloro, bromo, and iodo.
[0083] As used herein, "arylalkyl" refers to alkyl substituted by aryl and "cycloalkylalkyl" refers to alkyl substituted by cycloalkyl. An example arylalkyl group is benzyl.
[0084] As used herein, "heteroarylalkyl" refers to alkyl substituted by heteroaryl and "heterocycloalkylalkyl" refers to alkyl substituted by heterocycloalkyl.
[0085] As used herein, "amino" refers to NH2.
[0086] As used herein, "alkylamino" refers to an amino group substituted by an alkyl group.
[0087] As used herein, "dialkylamino" refers to an amino group substituted by two alkyl groups.
[0088] As used herein, "hydroxylalkyl" refers to an alkyl group substituted by hydroxyl.
[0089] As used herein, "cyanoalkyl" refers to an alkyl group substituted by cyano. The carbon of the cyano group is typically not counted if a carbon count precedes the term. For example, cyanomethyl is considered herein to be a C1 cyanoalkyl group.
[0090] 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 active 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.
[0091] Resolution of racemic mixtures of compounds can be carried out by any of numerous methods known in the art. An example method includes fractional recrystal I iza ion using a chiral resolving acid which is an optically active, saltforming 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 ß-camphorsulfonic acid. Other resolving agents suitable forfractional 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, cyclohexy-lethylamine, 1,2-diaminocyclohexane, and the like.
[0092] 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.
[0093] 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, amide - imidic acid 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 1Hand 2H-pyrazole. Tautomeric forms can be in equilibrium or sterically locked into one form by appropriate substitution.
[0094] Compounds of the invention further include hydrates and solvates, as well as anhydrous and non-solvated forms.
[0095] 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.
[0096] In some embodiments, the compounds of the invention, and 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 is was formed or detected. Partial separation can include, for example, a composition enriched in the compound 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 compound of the invention, or salt thereof. Methods for isolating compounds and their salts are routine in the art.
[0097] 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.
[0098] 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.
[0099] 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 conventional 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, nonaqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile (MeCN) 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).
[0100] As used herein, "prodrugs" refer to any covalently bonded carriers which release the active parent drug when administered to a mammalian subject. Prodrugs can be prepared by modifying functional groups present in the compounds in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent compounds. Prodrugs include compounds wherein hydroxyl, amino, sulfhydryl, or carboxyl groups are bonded to any group that, when administered to a mammalian subject, cleaves to form a free hydroxyl, amino, sulfhydryl, or carboxyl group respectively. Examples of prodrugs include, but are not limited to, acetate, formate and benzoate derivatives of alcohol and amine functional groups in the compounds of the invention. Preparation and use of prodrugs is discussed in T.
Higuchi and V. Stella, "Pro-drugs as Novel Delivery Systems," Vol. 14 of the A.C.S. Symposium Series, and in Biore-versible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987.
Synthesis [0101] Compounds of the invention, including salts thereof, can be prepared using known organic synthesis techniques and can be synthesized according to any of numerous possible synthetic routes.
[0102] 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 nonreactive 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.
[0103] 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 T.W. Green and P.G.M. Wuts, Protective Groups in Organic Synthesis, 3rd. Ed., Wiley & Sons, Inc., New York (1999).
[0104] 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), or mass spectrometry, or by chromatography such as high performance liquid chromatography (HPLC) or thin layer chromatography.
[0105] Compounds of the invention can be prepared according to numerous preparatory routes known in the literature. Example synthetic methods for preparing compounds of the invention are provided in the Schemes below.
[0106] As shown in Scheme 1, pyrazole-containing cores 1-9 and 1-6 can be synthesized starting with pyrrolo[2,3-bjpyridine or pyrrolo[2,3-b]pyrim idine 1-1. The compound 1-1 can be converted to an active species such as an N-oxide analog (1 -2) by using an oxidant such as m-CPBA. The N-oxide 1-2 can be halogenated with a halogenating agent such as a combination of tétraméthylammonium bromide and methanesulfonic anhydride to form a 4-halo compound 1-3 such as a 4-bromo compound while the N-oxide is reduced at the same time. The amine group of the compound 1-3 can be protected by a suitable amine protecting group to afford the protected compound 1-7, which subsequently undergoes a Suzuki coupling with a boric acid 1-8 to afford the pyrazole-containing cores 1-9a which can be further reacted with reagent L-(Y)n-Z (where L is a leaving group) to give compounds of the invention 1-9b. Alternatively, the N-oxide 1-2 can be halogenated with a halogenating agent such as MeS02CI to form a 4-halo compound 1-4 such as a 4-chloro compound while the N-oxide is reduced at the same time. The 4-halo compound 1-4 can be coupled to a bromo-substituted pyrazole compound 1-5 under suitable conditions such as heating to afford the pyrazole-containing core 1-6, which may contain some functional groups such as bromo or cyano suitable for further chemical modification.
[0107] Similarly, an imidazole core 1-11 can be synthesized by coupling of the 4-halo compound 1-4 to an imidazole derivative 1-10 under suitable conditions such as heating to afford the imidazole-containing core 1-11, which may contain some functional groups such as bromo or cyano suitable for further chemical modification.
[0108] As shown in Scheme 2, pyrazole-containing cores 2-3, 2-5 and 2-6 can be synthesized starting with a bromo-substituted pyrazole derivative 2-1 (a compound 1-6 in Scheme 1 wherein one of R5 is Br). The bromo-substituted pyrazole derivative 2-1 can be coupled to boron-containing aromatic species such as an aromatic boric acid 2-2 using Suzuki coupling wherein Ar is aryl or heteroaryl, each of which can be optionally substituted by one or more substituents such as alky, aryl, CN, nitro, alkoxy, etc. Alternatively, an alkene- or alkyne-containing compound such as an alkene-containing 2-5 can be obtained by coupling the bromo-substituted pyrazole derivative 2-1 to an unsaturated compound such as an alkene 2-4 in the presence of a metal catalyst such as bis(triphenylphosphine)palladium (II) chloride wherein t can be 0, 1,2, and the like; and R can be a substituent such as alkyl, aryl, CN, nitro, alkoxy, etc. The alkene group of compound 2-5 can be reduced by hydrogenation to afford the corresponding compound 2-6.
[0109] As shown in Scheme 3, imidazole-containing cores 3-7 can be synthesized starting with an N-protected 4-bromo-pyrrolo[2,3-b]pyridine or an N-protected 4-bromo-pyrrolo[2,3-b]pyrimidine 3-1 wherein P is a suitable amine protecting group such as {[2-(trimethylsilyl)ethoxy]methyl} (SEM). Compound 3-1 can be reacted with a Grignard reagent such as isopropyl magnesium chloride to generate an aromatic anion through ion exchange. The subsequent addition of a chloroacetyl-containing compound such as 2-chloro-N-methoxy-N-methylacetamide 3-2 to the anion will typically afford the chloroacetyl derivative 3-3. The derivative 3-3 can be reacted with an organic acid salt such as a cesium salt R5C02Cs to afford a compound 3-4. In the presence of a suitable ammonia source such as ammonium acetate, the compound 3-4 can react with ammonia under suitable conditions such as at a high temperature to form the imidazole ring of the compound 3-5. The free amine nitrogen of the imidazole derivative 3-5 can undergo further modification such as reacting with a compound X-(Y)n-Z where X is a leaving group such as chloro, bromo or iodo so as to afford compound 3-6. The protecting group of compound 3-6 can be removed by an appropriate method according to the nature of the protecting group to yield compound 3-7. It should be noted that if there are functional groups present within the R, R5, and -(Y)n-Z group, further modification can be made. For example, a CN group can be hydrolyzed to afford an amide group; a carboxylic acid can be converted to a ester, which in turn can be further reduced to an alcohol, which in turn can be further modified. One skilled in the art will recognize appropriate further modifications.
[0110] As shown in Scheme 4, thiazole-containing cores 4-3 can be synthesized starting with an N-protected chloro-acetyl derivative 4-1 wherein P is a suitable amine protecting group such as SEM. Compound 4-1 can be reacted with a thioamide 4-2 to form the thiazole ring, followed by deprotection of the amine nitrogen of the pyrrole ring by removal of the P group to afford the compound 4-3. Various thioureas 4-5 (equivalent to compound 4-2 wherein -(Y)n-Z is NR’R"; and R’ and R" are H, alkyl, aryl or the like; or R’ and R" together with the N atom to which they are attached form a heterocycloalkyl) useful in preparing the thiazole compounds 4-3 can be made from secondary amines 4-4. A secondary amine 4-4 can be reacted with 1,1’-thiocarbonyldiimidazole; and the resulting intermediate can further be reacted with ammonia to afford a thiourea 4-5.
[0111] As shown in Scheme 5, thiazole-containing cores 5-5 can be synthesized starting with a thiazole compound 5-1. The compound 5-1 can be reacted with a metal alkyl such as n-butyl lithium via ion exchange to generate an aromatic anion in situ. The subsequent addition of boric acid trimethyl ester followed by hydrolysis will typically afford the boric acid 5-2. The boric acid 5-2 can undergo Suzuki coupling with an N-protected 4-bromo-pyrrolo[2,3-b]pyridine or an N-protected 4-bromo-pyrrolo[2,3-b]pyrimidine 5-3 wherein P is a suitable amine protecting group such as SEM. The protecting group P of the coupling product 5-4 can be removed by an appropriate method according to the nature of the protecting group to yield the compound of the invention 5-5.
[0112] As shown in Scheme 6, pyrazole-containing compounds 6-1 can further be modified by substitution on the pyrazole NH group with appropriate reagents. For example, a compound 6-1 wherein P is a suitable amine protecting group such as SEM can be reacted with L-(Y)n-Z where L represents a leaving group such as halo, triflate or the like to afford compound 6-2 under basic condition. If there are some functional groups present within the Y and/or Z group, further modification can be made. For example, a CN group can be hydrolyzed to afford an amide group; a carboxylic acid can be converted to a ester, which in turn can be further reduced to alcohol. One skilled in the art will recognize the further modifications if appropriate.
[0113] Additionally, compound 6-1 can be reacted with alkene 6-3 (wherein R’ and R" can be H, alkyl, cycloalkyl and the like; and Z’ can be an electron withdrawing group such as an ester or CN) to afford the compound 6-4. Further, substitution can be made on alkene 6-3 at the alpha position (alpha to Z’) to generate a substituted derivatives of product, 6-4 (see, e.g., Example 68).
[0114] Compounds 6-2 and 6-4 can be deprotected by appropriate methods according to the nature of the protecting group used to afford their corresponding de-protected counterpart.
[0115] As shown in Scheme 7, bromo pyrazole containing compounds 7-1 can be further modified by metallation with reagents like butyl lithium and reaction with electrophiles like aldehydes to give the alcohol containing compounds 7-2 which can be deprotected to yield compounds of the invention having formula 7-3. One skilled in the art will recognize the further modifications where appropriate.
[0116] As shown in Scheme 8, pyrazole-containing compounds 8-4 and 8-5 can be prepared by reaction of the N-protected bromo compound 8-1 with hydrazine in an appropriate solvent such as /\/,/\/-dimethylformamide (DMF) to give the hydrazine intermediate 8-2. The hydrazino intermediate 8-2 is reacted with an appropriately substituted 1,3 bis-aldehyde like 8-3 to give the pyrazole containing compound 8-4. If there are some functional groups present within the Y and/or Z group, further modification can be made. For example, a CN group can be hydrolyzed to afford an amide group; a carboxylic acid can be converted to a ester, which in turn can be further reduced to alcohol. One skilled in the art will recognize further potential modifications.
[0117] As shown in Scheme 9, the 1,2,4-oxadiazole compound 9-6 can prepared from the N-protected bromo compound 9- 1 by treatment with zinc cyanide in DMF in the presence of a catalyst like bis(tributyl) palladium to give the N-protected cyano compound 9-2. The N-hydroxy carboximidamide compound 9-3 can be prepared by heating the N-protected cyano compound 9-2 with hydroxylamine hydrochloride in an appropriate solvent like ethanol and a base like potassium carbonate at a temperature below the boiling point of the solvent. The N-protected 1,2,4-oxadiazole compound can be prepared by treating the N-hydroxy carboximidamide compound 9-3 with an appropriately substituted acid chloride compound 9-4 in a solvent like pyridine at a sufficient temperature to complete the ring closure. If there are some functional groups present within the Y and/or Z group, further modification can be made. For example, a CN group can be hydrolyzed to afford an amide group; a carboxylic acid can be converted to an ester, which in turn can be further reduced to alcohol. One skilled in the art will recognize further modifications where appropriate.
[0118] As shown in Scheme 10, the 3- and 4-aryl pyrazolo compounds 10-9 can be prepared by reaction of the respective 3-arylpyrazolo compound 10-4 or 4-aryl pyrazolo compound 10-7 with an appropriately substituted bromo compound 10- 8 as previously described. The 3-aryl pyrazolo compound 10-4 can be prepared by reacting an appropriately substituted aryl group containing a halogen like bromo or a triflate with the N-protected boronic acid or boronic acid ester pyrazole compound 10-2 under Suzuki-like conditions known in the literature. The N-protecting group of 10-3 can be removed by conditions previously described and known in the literature for removing groups like SEM.
[0119] The 4-arylpyrazolo compounds 10-7 can be prepared by reacting the appropriately substituted acetophenone compound 10-5 with DMF acetal in DMF at elevated temperatures to give the dimethylamino compound 10-6. The 4-arylpyrazolo compounds 10-7 can be prepared by treating the dimethylamino compound 10-6 with hydrazine in a solvent such as ethanol.
[0120] As shown in Scheme 11 the substituted pyrazole compound 11-5 can be prepared by a variety of methods, such as by removing the protecting group e.g., SEM from compound 11-4 under conditions previously described. For example the substituted pyrazole N-protected compound 11-4 can be prepared by reaction of the intermediate pyrazole N-protected compound 11-3 with an appropriately substituted alkyl halide, benzyl halide, alkyl sulfonates, e.g., mesylate ortosylate, or other suitable leaving group L, in an appropriate solvent such as MeCN, DMF or tetrahydrofuran (THF), in the presence of a base such a sodium hydride or cesium carbonate. The N-aryl pyrazole 11-4 (wherein Y is aromatic) may be prepared by reacting the intermediate pyrazole 11-3 with an appropriately substituted aryl boronic acid in a solvent such as dichloromethane (DCM) with copper acetate and pyridine. Alternatively the N-aryl pyrazole 11-4 (wherein Y is aromatic) can be prepared by reacting the intermediate pyrazole 11-3 with an appropriately substituted aryl-fluoride in a solvent such as DMF at elevated temperature. Or, the substituted pyrazole compounds 11-4 (wherein Z is a group such as nitrile or ester and Y is at least two carbons) can be prepared by the reaction of intermediate pyrazole 11-3 with an appropriately substituted acrylate, acrylonitrile or other Michael-like acceptors in a solvent such as DMF in the presence of a base such as 1,8-diazabicyclo[5.4.0]undec-7-ene (DBU) or triethylamine (TEA) and at a temperature below the boiling point of the solvent. If there are some functional groups present within the Y and/or Z group, further modification can be made. For example, a CN group can be hydrolyzed to afford an amide group; a carboxylic acid can be converted to a ester, which in turn can be further reduced to alcohol. One skilled in the art will recognize the further modifications if appropriate.
[0121] As shown in Scheme 12, pyrazole 12-1 wherein P is a suitable amine protecting group such as SEM can be reacted with an alkyne-containing conjugate acceptor such as 12-2, wherein Z is an electron-withdrawing group (for example, -CN) optionally in the presence of a base (DBU or K2C03 and the like) in a solvent such as DMF or MeCN for variable lengths of time to provide olefin-containing adducts 12-3. Compounds represented by the formula 12-3 can be deprotected by appropriate methods according to the nature of the protecting group used to afford compounds of the invention 12-4.
[0122] As shown in Scheme 13, oxazole- or thiazole-containing compounds 13-6 can be prepared starting with N-protected 4-chloro-pyrrolo[2,3-b]pyrimidine 13-1 wherein P is a suitable amine protecting group such as SEM. Oxazole-or thiazole-containing products of formula 13-2 can be prepared by palladium-catalyzed coupling of 13-1 with oxazole orthiazole. The compound 13-2 can be reacted with a metal alkyl such as n-butyllithium to generate the aromatic anion in situ to which can be added at low temperatures (preferably between -78°C and 0°C) derivatives of carboxylic acids 13-3 (wherein W = N(Me)(OMe) when X1=S; and W = Cl when X1=0), in the presence of other additives such as zinc chloride and copper(l) iodide when X1=0, in a suitable solvent such as THF to generate a variety of ketones 13-4. Ketones 13-4 can be caused to react with a variety of reagents such as diethyl (cyanomethyl)phosphonate or triethyl-phosphonoacetate in the presence of a base like potassium tert-butoxide followed by reduction (including hydrogenation ora copper-hydride catalyzed conjugate reduction), or with reagents such as tosylmethyl isocyanide to provide products of formula 13-5 wherein Z is an electron-withdrawing group such as ester or-CN. If there are functional groups present within the R group or encompassed by the Z group, further modification can be made, and such appropriate further modifications will be recognized by one skilled in the art. Compounds 13-5 can be deprotected by appropriate methods according to the nature of the protecting group used to afford their corresponding deprotected counterparts 13-6.
[0123] As shown in Scheme 14, aminothiazole-containing cores 14-5 can be synthesized starting with thiazole-con-taining core 14-1 wherein P is a suitable amine protecting group such as SEM. The compound 14-1 can be treated with a metal alkyl such as n-butyllithium to generate the aromatic anion in situ to which can be added a suitable source of electrophilic halogen such as carbon tetrabromide to afford the halogenated derivative 14-2. The protecting group P of 14-2 can be removed by an appropriate method according to the nature of the protecting group to yield product 14-3. The compound 14-3 can be reacted with amines 14-4 at elevated temperatures in a suitable solvent such as DMF to afford the compound of the invention, 14-5.
[0124] As shown in Scheme 15, pyrrole-containing cores 15-4 can be synthesized starting with N-protected 4-chloro-pyrrolo[2,3-b]pyrimidine 15-1 wherein P is a suitable amine protecting group such as DEM (diethoxymethyl). The compound 15-1 can be reacted with 1-(triisopropylsilyl)pyrrole-3-boronic acid under Suzuki coupling conditions to afford the simultaneously pyrrole-deprotected core 15-2. Pyrrole-containing compounds 15-2 can be reacted with alkenes 15-3 containing an electron-withdrawing group Z (such as -CN) in the presence of an appropriate base (such as DBU) at various temperatures (e.g., between room temperature and 40° C) followed by an in situ or separate deprotection step that is suitable for the selected protecting group to afford compounds of the invention 15-4.
[0125] As shown in Scheme 16, a substituted pyrazole compound containing a sulfone or sulfoxide functionality as in 16-6 can be prepared by a variety of methods, such as starting with an appropriately substituted bromo thiophenyl ether 16-2. Thioether 16-2 may be readily prepared by alkylation of the thiophenol 16-1 with an alkyl halide, mesylate or the like using a base like DBU, potassium carbonate or sodium hydride. The cinnamyl nitrile 16-3 may be prepared by Heck chemistry and the like, using palladium acetate and triphenylphosphine in DMF at an appropriate temperature with acrylonitrile. The SEM protected intermediate 16-4 may be prepared by methods previously described for performing the Michael like addition of the pyrazole core to an appropriately substituted α-β unsaturated nitrile like 16-3. The sulfoxide 16-5, where n=1, and sulfone 16-5, where n=2, may be prepared by methods well known in the literature for the oxidation of the thio ether 16-4 like m-chloroperbenzoic acid (MCPBA) in DCM. The final compounds 16-6, where n= 0, 1 or 2, may be prepared by methods previously described for the removal of the SEM protecting group. Alternatively, the sulfur oxidation may be performed on compounds 16-2 or 16-3 depending on the compatibility of the substitution in the synthetic scheme.
[0126] Also, as shown in Scheme 17, substituted pyrazole compounds containing a sulfonamide functionality, such as 17-6 can be prepared by a variety of methods. For example, one may start with an appropriately substituted bromo phenyl sulfonamide 17-2, where Rc and Rd are suitable substituents. A compound 17-2 may be readily prepared by reaction of the bromo phenyl sulfonyl chloride 17-1 and an appropriately substituted amine such as an aniline, or a primary or secondary amine in a suitable solvent such as DCM, THF or pyridine. The cinnamyl nitrile 17-3 may be prepared by Heck chemistry or the like, using palladium acetate and triphenylphosphine in DMF at an appropriate temperature with acrylonitrile. The final compounds 17-6 where Rc and Rd are part of the sulfonamide functional group may be prepared by methods analogous to those described in Scheme 16 starting with the cinnamyl nitrile 17-3.
[0127] Also, as shown in Scheme 18, substituted pyrazole compounds containing an alpha-allyl cyclopentylmethylene functionality, such as 18-8, can be prepared by, for example, reacting a pyrazole 18-3, wherein P is a suitable amine protecting group such as SEM and X is N or C, with a cyclopentylacrylate ester 18-4 to form the ester 18-5. The ester 18-5 may then be reduced to the corresponding aldehyde, 18-6, for example, by the two-step procedure of reducing to the alcohol and selectively oxidizing the intermediate alcohol to the aldehyde, e.g., via a Swem oxidation.. The aldehyde, 18-6, may then be converted to the corresponding olefin, 18-7, for example by reaction with a Wittig reagent. The olefin 18-7, may then be deprotected, as described earlier, to produce the formula 18-7 compound. The intermediate, 18-4, may be prepared, for example as shown in Scheme 18, stearting with cyclopentylaldehyde.
[0128] Also, as shown in Scheme 19, the cyanoguanidine derivative 19-6 can be prepared starting from substituted pyrazole compounds such as pyrazole 18-3, wherein P is a suitable protecting group such as SEM and X is N or C. A compound 18-3 may, for example, be reacted with olefin 19-1, prepared by Horner-Wadsworth Emmons reaction of the corresponding Boc-protected piperidone, in the presence of a suitable basic catalyst, in a suitable solvent, to form 19-2. The intermediate 19-2 is deprotected using a suitable deprotection reaction, to provide the amine compound 19-3, which then reacts selectively with a cyanoimidocarbonate reagent such as 19-4, in a polar solvent at a suitable temperature, for example, about 20 °C to give a cyanoimidocarbamate such as 19-5, which can then be reacted with any of a variety of amines at elevated temperature to give product 19-6.
[0129] The intermediate compounds 20-5 and 20-6 may be prepared by a variety of methods in the literature, for example, methods such as are outlined in Scheme 20. The intermediate compound 20-3 may be prepared by reaction of the aldehyde compound 20-1 with an appropriately substituted Wittig reagent or Horner Emmons reagents to give the α-β unsubstituted ester 20-3. Alternatively, 20-3 may be prepared by a Heck-like reaction with an appropriately substituted aryl bromide 20-2 and an acrylic ester in the presence of a palladium reagent at elevated temperatures. The compound 20-4 may be prepared by methods previously described for the Michael-like addition of an appropriately substituted pyrrole 18-3 on the α-β unsaturated ester compound 20-3. The aldehyde compound 20-5 may be prepared by reduction of the ester compound 20-4 with reagents such as diisobutyl aluminium hydride at low temperatures such as about -78 °C in an appropriate solvent. The aldehyde compound 20-5 can be further reduced to the corresponding alcohol compound 20-6 with reagents such as sodium borohydride in methanol. Alternatively the alcohol compound 20-6 may be prepared directly by reduction of the ester 20-4 with reagents such as lithium aluminium hydride in appropriate solvent and at appropriate temperatures.
[0130] The compounds 21-2 and 21-3 may be prepared by using a variety of methods in the literature, such as, for example, methods outlined in Scheme 21. The olefin compound 21-1 may be prepared by the reaction of aldehyde compound 20-5 with an appropriately substituted Wittig reagent or Horner Emmons reagents using a base such as sodium hydride or potassium t-butoxide in an appropriate solvent and conducted at temperature. The olefin compound compound 21-1 may be reduced to the saturated compound 21-2, for example, using hydrogenation conditions well known in the literature, e.g., hydrogen in the presence of palladium on carbon in a solvent such as methanol. The acetylenic compound 21-3 may be prepared by methods previously described, or by reaction of the aldehyde 20-5 with Bestmann-Ohira reagent (E. Quesada et al, Tetrahedron, 62 (2006) 6673-6680) as described in the literature. Alternatively the alcohol compound 20-6 in Scheme 20 may be oxidized to the aldehyde 20-5 with methods well known in the literature, e.g., Swern oxidation conditions, followed by reaction with the Bestmann-Ohira reagent, wherein this reaction sequence may be carried out either as a one pot two-step reaction sequence, or in two separate reaction steps.
[0131] The compounds 22-1 and 22-3 may be prepared by using a variety of methods in the literature, for example, via methods outlined in Scheme 22. The oxygen-substituted compound 22-1 may be prepared, for example, by reaction of an appropriately substituted alcohol 20-6 (in Scheme 20), wherein X is N or C, and P is a protecting group, with a base such as sodium hydride and an appropriate agent such as an alkyl iodide, carbonate, or isocyanate, carried out in a suitable solvent and at a suitable temperature. Alternatively, the alcohol group on the compound 20-6 may be converted to a leaving group LG, as in compound 22-2, where the leaving group can be, for example, bromide or mesylate. The compound 22-2 serves as a substrate for subsequent reaction with a nucleophile, such as, for example, sodium ethoxide (Nuc = ethoxy).
[0132] It should noted that in all of the Schemes described herein, if there are functional groups present on a substituent group such as Y, Z, R, R1, R2, R5, etc., further modification can be made if appropriate and desired. For example, a CN group can be hydrolyzed to afford an amide group; a carboxylic acid can be converted to a ester, which in turn can be reduced to an alcohol, which in turn can be further modified. In another example, an OH group can be converted into a better leaving group such as mesylate, which in turn is suitable for nucleophilic substitution, such as by CN. One skilled in the art will recognize such further modifications.
Methods [0133] Compounds of the invention can modulate activity of one or more Janus kinases (JAKs). The term "modulate" is meant to refer to an ability to increase or decrease the activity of one or more members of the JAK family of kinases. Accordingly, compounds of the invention can be used in methods of modulating a JAK by contacting the JAK with any one or more of the compounds or compositions described herein. In some embodiments, compounds of the present invention can act as inhibitors of one or more JAKs. In some embodiments, compounds of the present invention can act to stimulate the activity of one or more JAKs. In further embodiments, the compounds of the invention can be used to modulate activity of a JAK in an individual in need of modulation of the receptor by administering a modulating amount of a compound of Formula la, lb, or Ic.
[0134] JAKs to which the present compounds bind and/or modulate include any member of the JAK family. In some embodiments, the JAK is JAKI, JAK2, JAK3 or TYK2. In some embodiments, the JAK is JAK1 or JAK2. In some embodiments, the JAK is JAK2. In some embodiments, the JAK is JAK3.
[0135] The compounds of the invention can be selective. By "selective" is meant that the compound binds to or inhibits a JAK with greater affinity or potency, respectively, compared to at least one other JAK. In some embodiments, the compounds of the invention are selective inhibitors of JAK1 or JAK2 over JAK3 and/or TYK2. In some embodiments, the compounds of the invention are selective inhibitors of JAK2 (e.g., over JAK1, JAK3 and TYK2). Without wishing to be bound by theory, because inhibitors of JAK3 can lead to immunosuppressive effects, a compound which is selective for JAK2 over JAK3 and which is useful in the treatment of cancer (such as multiple myeloma, for example) can offer the additional advantage of having fewer immunosuppressive side effects. Selectivity can be at least about 5-fold, 10-fold, at least about 20-fold, at least about 50-fold, at least about 100-fold, at least about 200-fold, at least about 500-fold or at least about 1000-fold. Selectivity can be measured by methods routine in the art. In some embodiments, selectivity can be tested at the Km of each enzyme. In some embodiments, selectivity of compounds of the invention for JAK2 over JAK3 can be determined by the cellular ATP concentration.
[0136] 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 over-expression 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.
[0137] 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).
[0138] Further examples of JAK-associated diseases include autoimmune diseases such as multiple sclerosis, rheumatoid arthritis juvenile arthritis, type I diabetes, lupus, psoriasis, inflammatory bowel disease, ulcerative colitis, Crohn’s disease, myasthenia gravis, immunoglobulin nephropathies, autoimmune thyroid disorders, and the like. In some embodiments, the autoimmune disease is an autoimmune bullous skin disordersuch as pemphigus vulgaris (PV) or bullous pemphigoid (BP).
[0139] Further examples of JAK-associated diseases include allergic conditions such as asthma, food allergies, atopic dermatitis and rhinitis. Further examples of JAK-associated diseases include viral diseases such as Epstein Barr Virus (EBV), Hepatitis B, Hepatitis C, HIV, FITLV 1, Varicella-Zoster Virus (VZV) and Fluman Papillomavirus (HPV).
[0140] 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 togetherwith 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.
[0141] 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, melanoma etc.), hematological cancers (e.g., lymphoma, leukemia such as acute lymphoblastic leukemia, or multiple myeloma), and skin cancer such as cutaneous T-cell lymphoma (CTCL) and cutaneous B-cell lymphoma. Example cutaneous T-cell lymphomas include Sezary syndrome and mycosis fungoides.
[0142] JAK-associated diseases can further include those characterized by expression of a mutant JAK2 such as those having at least one mutation in the pseudo-kinase domain (e.g., JAK2V617F).
[0143] JAK-associated diseases can further include myeloproliferative disorders (MPDs) such as polycythemia vera (PV), essential thrombocythemia (ET), myeloid metaplasia with myelofibrosis (MMM), chronic myelogenous leukemia (CML), chronic myelomonocytic leukemia (CMML), hyperéosinophilie syndrome (HES), systemic mast cell disease (SM-CD), and the like.
[0144] Further JAK-associated diseases include inflammation and inflammatory diseases. Example inflammatory diseases include 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.
[0145] 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 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.
[0146] 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.
[0147] 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.
[0148] 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 in a tissue, system, animal, individual or human that is being sought by a researcher, veterinarian, medical doctor or other clinician, which includes one or more of the following: (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 (/.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 (/.e., reversing the pathology and/or symptomatology).
Combination Therapies [0149] 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, or other agents can be used in combination with the compounds of the present invention 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.
[0150] Example chemotherapeutic include proteosome inhibitors (e.g., bortezomib), thalidomide, revlimid, and DNA-damaging agents such as melphalan, doxorubicin, cyclophosphamide, vincristine, etoposide, carmustine, and the like.
[0151] Example steroids include coriticosteroids such as dexamethasone or prednisone.
[0152] 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, EP2005/009967, EP2005/010408, and U.S. Ser. No. 60/578,491.
[0153] 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.
[0154] Example suitable RAF inhibitors include compounds, and their pharmaceutically acceptable salts, as disclosed in WO 00/09495 and WO 05/028444.
[0155] 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.
[0156] 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.
[0157] 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.
[0158] 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.
Pharmaceutical Formulations and Dosage Forms [0159] 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 fortopical 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. Coated condoms, gloves and the like may also be useful.
[0160] This invention also includes pharmaceutical compositions which contain, as the active ingredient, one or more of the compounds of the invention above in combination with one or more pharmaceutically acceptable carriers (excipients). 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.
[0161] 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.
[0162] 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.
[0163] The compositions can be formulated in a unit dosage form, each dosage containing from about 5 to about 1000 mg (1 g), more usually about 100 to about 500 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.
[0164] The active compound can 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.
[0165] 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.
[0166] 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.
[0167] 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.
[0168] 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.
[0169] 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.
[0170] 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.
[0171] The therapeutic dosage of the compounds 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.
[0172] 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.
Labeled Compounds and Assay Methods [0173] 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.
[0174] 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 (/. e., naturally occurring). Suitable radionuclides that may be incorporated in compounds of the present invention include but are not limited to 2H (also written as D for deuterium), 3H (also written as T for tritium), 11C, 13C, 14C, 13N, 15N, 150, 170,180, 18F, 35S, 36CI, 82Br, 75Br, 76Br, 77Br, 123l, 124l, 125l and 1311. The radionuclide that is incorporated in the instant radio-labeled compounds will depend on the specific application ofthat radio-labeled compound. For example, for in vitro metalloprotease labeling and competition assays, compounds that incorporate 3H, 14C, 82Br, 125l, 131l, 35S or will generally be most useful. For radio-imaging applications 11C, 18F, 125l, 123l, 124l, 131l, 75Br, 76Bror 77Br will generally be most useful.
[0175] It is understood that 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 3H, 14C, 125l, 35S and 82Br.
[0176] 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.
[0177] 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 [0178] The present invention also includes pharmaceutical kits useful, for example, in the treatment or prevention of J AK-associated diseases ordisorders, 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.
[0179] The invention will be described in greater detail byway 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 noncritical 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 not falling within the scope of the claims are described for reference purposes.
EXAMPLES
Example 1: 3-[3-Methyl-1-(1 H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-4-yl]benzonitrile [0180]
Step 1. 1 H-Pyrrolo[2,3-b]pyridine 7-oxide [0181] To a solution of 1 H-pyrrolo[2,3-b]pyridine (4.90 g, 0.0415 mol) in ethyl acetate (41 mL, 0.42 mol) was added a solution of meta-chloroperbenzoicacid (MCPBA; 9.3 g, 0.054 mol) in ethyl acetate (27 mL, 0.28 mol) atO °C. The reaction mixture was solidified when ~20 mL solution of MCPBA was added. An additional ~10 mL of ethyl acetate was added so that a solution resulted. The reaction mixture was allowed to warm to room temperature (rt) and stirred overnight, then was cooled at 0 °C, filtered and washed with ethyl acetate three times to give 10.94 g wet solid. The wet solid (8.45 g) was then suspended in water (35 mL), and to the suspension was added 13 mL of sat. Na2C03 dropwise, and the resulting mixture was stirred at room temperature overnight. The mixture was then cooled at 0° C, filtered and washed with water (x4) to give 3.55 g of pale purple solid which was dried at 40° C overnight to give the desired product (2.47 g, 44.4% yield). 1H NMR (400 MHz, CD3OD): δ: 8.2 (1H, d); 7.95 (1H, d); 7.5 (1H, d); 7.2 (1H, m); 6.65 (1H, d). MS (M+H)+: 136.
Step 2. 4-Chloro-1H-pyrrolo[2,3-b]pyridine [0182] To a pink solution of 1H-pyrrolo[2,3-b]pyridine 7-oxide (2.47 g, 0.0184 mol) in dimethylformamide (DMF) (13.3 mL, 0.172 mol) was added methanesulfonyl chloride (4.0 mL, 0.052 mol) at 50 °C, and the pink color changed to orange. The reaction mixture was heated at 73 °C for 2h, then cooled to 40 °C. Water (35 mL) was added, and the resulting suspension was cooled at 0 °C. NaOH was added to adjust the pH of the mixture to about 7. The mixture was filtered and washed with water (x3) to give 3.8 g of a wet pale orange solid that was dried at 40 °C overnight to give the product (2.35 g, 82.2% yield). 1H NMR (400 MHz, CDCI3): δ 10.8 (1H, br); 8.21 (1H, d); 7.41(1 H, d); 7.18 (1H, d); 6.61 (1H, d). MS (M+H)+: 153.
Step 3. 4-(4-Bromo-3-methyl-1H-pyrazol-1-yl)-1H-pyrrolo[2,3-b]pyridine [0183]
[0184] A mixture of 4-chloro-1 H-pyrrolo[2,3-b]pyridine (0.050 g, 0.00033 mol) and 4-bromo-3-methyl-1H-pyrazole (0.10 g, 0.00066 mol) was heated at 130 °C overnight. The reaction mixture then was subjected to column chromatography (eluting with 5% MeOH/DCM, 0.5% NH4OH, on silica gel) to give 80 mg pale yellow solid which was triturated with MeOH (1.5 mL) to yield the product as a pale yellow solid (44 mg, 44% yield). 1H NMR (400 MHz, CD3OD): δ 8.32 (1H, s); 8.25 (1H, d); 7.6 (1H, s); 7.45 (1H, d); 7.37 (1H, d); 6.96 (1H, d); 2.4 (3H, s). MS (M+H)+: 276.
Step 4. 3-[3-Methyl-1-( 1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-4-yl]benzonitrile [0185] A mixture of 4-(4-bromo-3-methyl-1H-pyrazol-1-yl)-1H-pyrrolo[2,3-b]pyridine (0.032 g, 0.00012 mol), (3-cyan-ophenyl)boronic acid (0.027 g, 0.00018 mol), sodium carbonate (0.032 g, 0.00030 mol) and tetrakis(triphenylphos-phine)palladium(O) (7.0 mg, 0.0000060 mol) in 1,2-dimethoxyethane (0.3 mL, 0.003 mol) and water (0.3 mL, 0.02 mol) was heated at 130 °C (a liquid resulted, but with two layers) for 4 h. The reaction mixture then was cooled to room temperature (rt), filtered and was washed with water (x2) and dimethyl ether (DME) (x2) to give the product as a pale orange solid (15 mg, 44% yield). 1H NMR (400 MHz, CD3OD): δ 8.57 (1H, s); 8.31 (1H, d); 7.8 (2H, m); 7.75 (2H, m); 7.55 (1H, s); 7.45 (2H, m); 7.01 (1H, d); 2.6 (3H, s). MS (M+H)+: 299.
Example 2: (2E)-3-[3-Methyl-1-(1H-pyrrolo[2,3-6]pyridin-4-yl)-1H-pyrazol-4-yl]acrylonitrile trifluoroacetate salt [0186]
Step 1. 4-Bromo-1H-pyrrolo[2,3-b]pyridine [0187] To a solution of 1 H-pyrrolo[2,3-b]pyridine 7-oxide (8.0 g, 0.060 mol), prepared by the procedure outlined in Example 1, Step 1 in DMF (100 mL, 1 mol) was added methanesulphonic anhydride (20.8 g, 0.119 mol, in four portions) at 0 °C. The mixture was stirred at 0 °Cforan additional 20 min followed by an addition of tétraméthylammonium bromide (23.0 g, 0.149 mol). The resulting mixture was stirred overnight. Water (0.1 L) was added, and a slight exotherm was observed. A solution of sodium hydroxide in water (12.5 M, 12 mL) was added to adjust the pH of the mixture to about 8, followed by an addition of -0.25 L of water. The resulting mixture was stirred for additional 2 h then filtered. The solid obtained was washed with water x3 to give 6.72 g of a reddish solid which was dried at 50 °C over a weekend to give the product (5.75 g, 49% yield). 1H NMR (400 MHz, CDCI3): Ô10.8 (1H, br); 8.2 (1H, d); 7.41 (1H, d); 7.19 (1H, d); 6.61 (1H, d). MS (M+H)+: 196.
Step 2. 4-Bromo-1 -(2-(trimethylsilyl)ethoxy]methyl-1 Hpyrrolo[2,3-b]pyridine [0188] To a solution of4-bromo-1H-pyrrolo[2,3-b]pyridine (6.2 g, 0.031 mol) and [ß-(trimethylsilyl)ethoxy]methyl chloride (6.7 mL, 0.038 mol) in DMF (62 mL, 0.80 mol) was added sodium hydride (1.5 g, 0.038 mol) at 0 °C, and the resulting solution turned opaque. The mixture was stirred for additional 4 h, then diluted with methyl ferf-butyl ether (MTBE). The organic layer was separated and washed with water (x2) and brine aqueous solution successively. The organic phase was dried and concentrated in vacuo to give 14.1 g of a product as a pale orange oil. The oil was purified by column chromatography eluting with 5-20% ethyl acetate/hexanes to give the purified product as a colorless oil (9.66 g , 94% yield). 1H NMR (400 MHz, CDCI3): δ 8.2 (1H, d); 7.49 (1H, d); 7.19 (1H, d); 6.62 (1H, d); 5.78 (2H, s); 3.6 (2H, t); 0.98 (2H, t); 0.0 (9H, s). MS (M+H)+: 326.
Step 3. (2E)-3-(3-Methyl-1-( 1 H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-4-yl]acrylonitrile [0189] A solution of 2-propenenitrile (0.043 mL, 0.00065 mol), bis(triphenylphosphine)palladium(ll) chloride (0.0091 g, 0.000013 mol), 4-(4-bromo-3-methyl-1H-pyrazol-1-yl)-1H-pyrrolo[2,3-b]pyridine (0.036 g, 0.00013 mol), and tetrae-thylamine (TEA) (0.15 mL, 0.0011 mol) in DMF (0.15 mL, 0.0019 mol) was microwaved at 120 °C for 2 h. The solution was then diluted with ethyl acetate and washed with water (x2) and brine successively. The organic phase was dried and concentrated in vacuo to give 62 mg of the product as an orange solid. The orange solid was purified by prep-LCMS to give 12 mg of an off-white solid as a trifluoroacetic acid (TFA) salt which was triturated with MTBE (1 mL) to provide the purified product as a pale green solid, (dried at 60 °C for 4 h, 9 mg , 28% yield). 1H NMR (400 MHz, CD3OD): 2 :1 oftrans : cis isomers. Fortrans: δ 8.95 (NH,1H, s); 7.75 (olefin, 1H, d); 6.1 (olefin, 1H, d); 2.45 (Me, 3H, s). MS (M+H)+: 249.
Example 3: 3-[3-Methyl-1-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-4-yl]propanenitrile, trifluoroacetate salt [0190]
[0191] A mixture of (2E)-3-[3-methyl-1-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-4-yl]acrylo-nitrile, TFA salt, (0.0050 g, 0.000020 mol, prepared according to Example 2) and palladium (5.8 mg, 0.0000054 mol) in methanol (1 mL, 0.02 mol) and 1,2-dichloroethane (1 mL, 0.01 mol) was degassed and then was stirred under an atmosphere of hydrogen for 3 h. The reaction mixture then was filtered and the filtrate was concentrated in vacuo to give 8 mg of the product as an off-white solid. The crude material was purified by prep-LCMS to give 5.1 mg of a white solid as a TFA salt which was triturated with MTB (1 mL) to give the product as a white solid (1.7 mg, 34% yield). 1H NMR (400 MHz, CD3OD): δ 8.52 (1H, s); 8.35 (1H, d); 7.72(1H, d); 7.6 (1H, s); 7.38 (1H, d); 6.96 (1H, d); 2.7-2.9 (4H, m); 2.4 (3H, s). MS (M+H)+: 251.
Example 13: 4-(4-Phenyl-1 H-imidazol-1-yl)-1 H-pyrrolo[2,3-b]pyridine [0192]
[0193] A melt of 4-chloro-1H-pyrrolo[2,3-b]pyridine (0.050 g, 0.00033 mol) in 4-phenyl-1 H-imidazole (0.24 g, 0.0016 mol) was heated at 200 °C overnight. The reaction was partitioned between ethyl acetate and saturated NaHCOs, separated and the organic phase was washed with brine. The organic layer was then dried and evaporated to give 250 mg of an orange oil. The oil was chromatographed with 7% MeOH/DCM, 0.7% NH4OH, sample in solvent system. Collected 74 mg of the product as an orange glass. The glass was triturated with hot DCE (1.5 mL) to give 51 mg of a brown solid which was dried at 60 °C for 4 h to afford the desired product (50 mg, 59 yield). 1H NMR (400 MHz, dimethylsulxoxide (DMSO)): δ 12.5 (1H, s); 8.5 (1H, s); 8.4 (1H, s); 8.38 (1H, d); 7.8 (2H, m); 7.62 (1H, d); 7.4 (3H, m); 7.3 (1H, m); 6.81 (1H, d). MS (M+H)+: 260
Example 14: [3-Methyl-1-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-4-yl]-piperidin-1-yl-methanone [0194]
Step 1. 3-Methyl-1-( 1 -[2-(trimethylsilyl)ethoxy]methyl-1 H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazole-4-carboxylic acid [0195] Toa-70°Csolutionof4-(4-bromo-3-methyl-1H-pyrazol-1-yl)-1-[2-(trimethylsilyl)ethoxy]-methyl-1H-pyrrolo[2,3-b]pyridine (0.107 g, 0.000263 mol) in THF (1 mL, 0.01 mol), and n-butyllithium in hexane (0.23 mL of 1.6M), 0.5g of C02 solid was added. After 15 min, the reaction was quenched with NH4CI. Ethyl acetate and water were added. The organic phase was washed with brine, and was evaporated to give 84 mg of an off-white glass/solid. The solid was chromatographed with 50% ethyl acetate/hexanes, 0.5% AcOH, sample on silica gel to give 40 mg of a purified product as a white solid (37% yield). 1H NMR (400 MHz, CDCI3): δ 8.5 (1H, d); 7.45 (1H, d); 7.25 (1H, d); 7.02 (1H, s); 6.6 (1H, d); 5.75 (2H, s); 3.6 (2H, t); 2.48 (3H, s); 0.98 (3H, t); 0.0 (9H, s). MS (M+H)+: 372.
Step 2. 4-[3-Methyl-4-(piperidin-1 -ylcarbonyl)-1 H-pyrazol-1 -yl]-1 -[2-(trimethylsilyl)ethoxy]methyl-1 H-pyrrolo[2,3-b]pyrid-ine [0196] A solution of 3-methyl-1-(1-[2-(trimethylsilyl)ethoxy]methyl-1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazole4-car-boxylic acid (0.040 g, 0.00011 mol) (1:1 of AcOH) and Ν,Ν-carbonyldiimidazole (0.035 g, 0.00021 mol) in THF (1 mL, 0.01 mol) was stirred for 1.2h, after which time piperidine (32 μί, 0.00032 mol) was added. After another 2h, another portion of piperidine (15 μί) was added and the resulting mixture was stirred overnight. The reaction mixture was then partitioned between ethyl acetate and water, and washed sequentially with sat. NaHC03 and brine. The organic phase was dried and evaporated to give 49 mg of the crude product as an orange oil/glass. The crude product was chromatographed with 75-100% ethyl acetate/hexanes, sample in DCM. Collected 25 mg of the purified product as a colorless glass/oil (50% yield). 1H NMR (400 MHz, CDCI3): δ 8.45 (1H, d); 8.23 (1H, s); 7.5 (1H, d); 7.4 (1H, d); 7.05 (1H, d); 5.8 (2H, s); 3.7 (4H, br); 3.6 (2H, t); 2.55 (3H, s); 1.7 (6H, br); 1.0 (3H, t); 0.0 (9H, s). MS (M+H)+: 439.
Step 3. 3-Methyl-1-( 1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-4-yl]-piperidin-1-yl-methanone [0197] A solution of 4-[3-methyl-4-(piperidin-1-ylcarbonyl)-1 H-pyrazol-1 -yl]-1-[2-(trimethylsilyl)-ethoxy]methyl-1H-pyr-rolo[2,3-b]pyridine (0.025 g, 0.000057 mol) in TFA (1 mL, 0.01 mol) was stirred for 1.5 h. The reaction mixture was then concentrated and partitioned between DCM and sat. NaHC03 x2, and brine. The organic layer was then dried and concentrated to give 28 mg of the product as a white foam. The foam was dissolved in methanol (1 mL, 0.02 mol) and treated with ammonium hydroxide in water (8.0M, 1 mL) for 1.5h. The reaction was concentrated using a rotary evaporator to give 24 mg of a pale yellow glass. The glass was triturated with methyl t-butyl ether (MTBE) to give 13 mg of a white solid which was dried at rt over a weekend. A total of 8 mg of the product was obtained after drying (45% yield). 1H NMR (400 MHz, CDCI3): δ 9.7 (1H, s); 8.4 (1H, d); 8.2 (1H, s); 7.42 (1H, d); 7.4 (1H, d); 6.99 (1H, d); 3.4-3.8 (4H, br); 2.47 (3H, s); 1.5-1.8 (6H, br). MS (M+H)+: 309.
Example 15: [3-Methyl-1-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-4-ylmethyl]-phenyl-amine [0198]
Step 1. 3-Methyl-1-( 1 -[2-(trimethylsilyl)ethoxy]methyl-1H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazole-4-carbaldehyde [0199] Toa-70°Csolutionof4-(4-bromo-3-methyl-1H-pyrazol-1-yl)-1-[2-(trimethylsilyl)ethoxy]-methyl-1H-pyrrolo[2,3-b]pyridine (0.25 g, 0.00061 mol) in THF (2 mL, 0.03 mol), 1.6 M n-butyllithium in hexane (0.54 mL). After 10 min, DMF (120 μΙ_, 0.0015 mol) was added. The reaction was allowed to warm to rt and stirred overnight. The reaction was then quenched with NH4CI. Ethyl acetate/water was added. The organic phase was separated and washed with brine, then dried and concentrated to give 180 mg of an orange oil. The crude product was chromatographed with 25% ethyl acetate/hexanes, sample in DCM. Collected 40 mg of a pale yellow oil (18% yield). 1H NMR (400 MHz, CDCI3): δ 10.15 (1H, s); 8.7(1H,s); 8.47 (1H,d); 7.58 (1H, d); 7.5 (1H, d); 7.05 (1H, d); 5.8 (2H, s); 3.63 (2H, t); 2.7 (3H, s); 0.98 (3H, t); 0.0 (9H, s). MS (M+H)+: 356.
Step 2. N-[3-Methyl-1-( 1 -[2-(trimethylsilyl)ethoxy]methyl-1 H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-4-yl] methylaniline [0200] A solution of 3-methyl-1-(1-[2-(trimethylsilyl)ethoxy]methyl-1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazole-4-car-baldehyde (0.025 g, 0.000070 mol) and aniline (1M in DCM, 0.070 mL), in DCM (1 mL, 0.02 mol) was stirred for 1 min. Acetic acid (20 μί, 0.0004 mol), aniline (1M in DCM, 140 μί) and sodium triacetoxyborohydride (0.022 g, 0.00010 mol) were added. The reaction was stirred overnight and partitioned between DCM and sat. NaHCOs, washed with brine. The organic phase was dried and evaporated to give 21 mg of a product as a pale orange glass (70% yield). 1H NMR (400 MHz, CDCI3): δ: 8.4 (1H, d); 8.15 (1H, s); 7.65 (1H, d); 7.35 (3H, m); 7.09 (1H, d); 6.82 (1H, m); 6.89 (2H, m); 5.8 (2H, s); 4.35 (2H, s); 3.6 (2H, t); 2.5 (3H, s); 0.99 (3H, t); 0.0 (9H, s). MS (M+M+: 433.
Step 3. [3-Methyl-1-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-4-ylmethyl]-phenyl-amine [0201] Deprotection of N-[3-methyl-1-(1-[2-(trimethylsilyl)ethoxy]methyl-1 H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-4-yljmethylaniline was carried out according to the procedures of Exam pie 14, Step 3 togive the desired product (58% yield). 1H NMR (400 MHz, CDCI3): δ: 9.9 (1H, s); 8.38 (1H, d); 8.1 (1H, s); 7.4 (1H, d); 7.35 (1H, d); 7.3 (2H, m); 7.0 (1H, d); 6.79 (1H, m); 6.77 (2H, m); 4.25 (2H, s); 3.81 (1H, s); 2.41 (3H, s). MS (M+H)+: 303.
Example 25: 3-[3-Methyl-1-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-4-yl]-cyclohexanol [0202]
Step 1. 3-Ethoxy-1-[3-methyl-1-(1-[2-(trimethylsilyl)ethoxy]methyl-1Hpyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-4-yl]cy-clohex-2-en-1-ol [0203]
[0204] Toa-75°Csolutionof4-(4-bromo-3-methyl-1H-pyrazol-1-yl)-1-[2-(trimethylsilyl)ethoxy]-methyl-1H-pyrrolo[2,3-b]pyridine (0.11 g, 0.00027 mol) in THF (1.5 mL, 0.018 mol) was added 1.6 M n-butyllithium in hexane (0.22 mL). The reaction mixture turned dark orange. After ~10 min, 1.0 M magnesium dibromide in ether (0.35 mL) was added. After another 50 min, a solution of 3-ethoxy-2-cyclohexen-1-one (41.5 μί, 0.000308 mol) in THF (-0.3 mL) was added. The resulting mixture was warmed to -40 °C over -1 h and quenched with NH4CI. Then ethyl acetate/water was added. The organic phase was washed with brine, and concentrated to give 145 mg of an orange oil. The crude product was chromatographed with 0-50% ethyl acetate/hexane gradient, sample in DCM. Collected 35 mg of the produce as an oil (30% yield). 1H NMR (400 MHz, CDCI3): δ 8.49 (1H, d); 8.38 (1H, s); 7.55 (1H, d); 7.4 (1H, d); 7.1 (1H, d); 6.0 (2H, s); 3.6 (2H, t); 2.81 (2H, m); 2.62 (3H, s); 2.58 (2H, m); 2.27 (2H, m); 1.0 (3H, t); 0.0 (9H, s). MS (M+H)+: 422.
Step 2. 3-[3-Methyl-1-(1-[2-(trimethylsilyl)ethoxy]methyl-1Hpyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-4-yl]cyclohexanol [0205] A mixture of 3-[3-methyl-1-(1-[2-(trimethylsilyl)ethoxy]methyl-1 H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol4-yl]cy-clohex-2-en-1-one (0.019 g, 0.000045 mol) and palladium on carbon (Pd/C) (0.018 g, 0.000017 mol) in methanol (2 mL, 0.05 mol) was degassed and was stirred under a hydrogen atmosphere overnight. An additional 48 mg of 10% Pd/C was added and stirred under a hydrogen atmosphere for 8h. The palladium was filtered and the filtrate was stirred with sodium tetrahydroborate (0.032 g, 0.00084 mol) for 5h. The reaction was purified by prep-HPLC to give 5 mg of the desired product. MS (M+H)+: 426.
Step 3. 3-(3-Methyl-1-( 1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-4-yl]-cyclohexanol [0206] Deprotection of 3-[3-methyl-1-(1-[2-(trimethylsilyl)ethoxy]methyl-1 H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-4-yl]cyclohexanol was carried out according to the procedures of Example 14, Step 3 to give the desired product (40% yield). 1H NMR (400 MHz, CDCI3): δ 9.72 (1H, s); 8.35 (1H, d); 7.95 (1H, s); 7.41 (1H, d); 7.35 (1H, d); 7.02 (1H, d); 3.78 (1H, m); 2.6 (1H, m); 2.4 (3H, s); 1.2-2.4 (8H, m). MS (M+H)+: 296.
Example 40: 4-[1-(3-Methoxy-1-methyl-propyl)-1 H-pyrazol-4-yl]-1 H-pyrrolo[2,3-h]pyridine [0207]
Step 1. 4-[1-(3-Methoxy-1 -methylpropyl)-1 H-pyrazol-4-yl]-1 -[2-(trimethylsilyl)ethoxy]-methyl-1 H-pyrrolo[2,3-b]pyridine [0208] To a 0 °C solution of 3-[4-(1-[2-(trimethylsilyl)ethoxy]methyl-1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]bu-tan-1-ol (the alcohol was made by DIBAL reduction of the ester in Example 58) (0.056 g, 0.00014 mol)) in DMF (1 mL, 0.01 mol), was added sodium hydride (0.0107 g, 0.000268 mol). After 5 min, methyl iodide (18 μΙ_, 0.00029 mol) was added and the resulting mixture was stirred over a weekend. The mixture was then partitioned between ethyl acetate and water, separated and the organic phase was washed with brine. The organic phase was concentrated to give a pale orange oil. 1H NMR (400 MHz, CDCI3): δ 8.4 (1H, d); 8.3 (1H, s); 8.0 (1H, s); 7.65 (1H, d); 7.27 (1H, d); 6.8 (1H, d); 5.8 (2H, s); 4.7 (1H, m); 3.63 (2H, t); 3.2-3.4 (2H, m); 3.38 (3H, s); 2.1-2.3 (2H, m); 1.7 (3H, d); 1.0 (2H, t); 0.0 (9H, s). MS (M+H)+: 400.
Step2. 4-[1 -(3-Methoxy-1 -methyl-propyl) -1 H-pyrazol-4-yl]-1 H-pyrrolo[2,3-b]pyridine [0209] Deprotection of 4-[1-(3-methoxy-1-methylpropyl)-1 H-pyrazol-4-yl]-1-[2-(trimethylsilyl)-ethoxy]-methyl-1 H-pyr-rolo[2,3-b]pyridine was carried out according to the procedures of Example 14, Step 3 to give the desired product (25% yield). 1H NMR (400 MHz, CDCI3): δ 10.0 (1H, s); 8.35 (1H, d); 8.18 (1H, s); 7.95 (1H, s); 7.41 (1H, d); 7.21 (1H, d); 6.75 (1H, d); 4.63 (1H, m); 3.15-3.4 (2H, m); 3.35 (3H, s); 2.21-2.05 (2H, m); 1.6 (3H, d). MS (M+H)+: 270.
Example 42: 4-[1-(1-Methyl-3-pyrazol-1-yl-propyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine [0210]
Step 1. 4-1-[1 -Methyl-3-( 1 H-pyrázol-1 y I) propyl-1 H-pyrazol-4-yl-1 -[2-(trimethylsilyl)ethoxy]methyl-1 H-pyrrolo[2,3-b]pyridine [0211] To a 0°C solution of 3-[4-(1-[2-(trimethylsilyl)ethoxy]methyl-1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]butyl methanesulfonate (prepared by mesylation of the alcohol as in Example 59, Step 1) (0.055 g, 0.00012 mol) and 1H-pyrazole (0.025 g, 0.00036 mol) in DMF (1 mL, 0.01 mol) was added sodium hydride (0.014 g, 0.00036 mol). The resulting solution was stirred overnight and then partitioned between ethyl acetate and 0.1 N HCI, water, the organic phase was separated and washed with brine. The organic layer was then concentrated to give 49 mg of a pale orange glass (87% yield). 1H NMR (400 MHz, CDCI3): δ 8.4 (1H, d); 8.18 (1H, s); 7.99 (1H, s); 7.6 (1H, t); 7.5 (1H, d); 7.4 (1 H, t); 7.27 (1H, d); 6.8 (1H, d); 6.3 (1H, m); 5.8 (2H, s); 4.2 (1H, m); 4.0-4.2 (2H, m); 3.61 (2H, t); 2.58 (2H, m); 1.65 (3H, d); 1.0 (2H, t); 0.0 (9H, s). MS (M+H)+: 436.
Step 2. 4-[1-(1-Methyl-3-pyrazol-1-yl-propyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine [0212] Deprotection of 4-1-[1-methyl-3-(1 H-pyrazol-1-yl)propyl]-1 H-pyrazol-4-yl-1-[2-(trimethyl-silyl)ethoxy]methyl-1H-pyrrolo[2,3-b]pyridine was carried out according to the procedures of Example 14, Step 3 to give the desired product (38% yield). 1H NMR (400 MHz, CDCI3): δ 9.7 (1H, s); 8.38 (1H, d); 8.1 (1H, s); 7.7(1H, s); 7.59 (1H, t); 7.4 (1H, d); 7.35 (1H, t); 7.21 (1H, d); 6.75 (1H, d); 6.25 (1H, m); 4.4 (1H, m); 3.9-4.15 (2H, m); 2.55 (2H, m); 1.63 (3H, d). MS (M+H)+: 306.
[0213] The following compounds in Table 1 were made by methods analogous to the procedures above as indicated. "Purification A" indicates that the product following deprotection was purified by preparative-HPLC under the following conditions: C18 eluting with a gradient of MeCN/H20 containing 0.15% NH4OH.
Table 1
Example 46: 4-(2-tert-Butyl-1-methyl-1H-imidazol-4-yl)-1H-pyrrolo[2,3-b]pyridine trifluoro-acetate salt [0214]
Step 1.4-(2-tert-butyl-1 H-imidazol-5-yl)-1 -[2-(trimethylsilyl) ethoxyjmethyl-1 H-pyrrolo[2,3-b]pyridine [0215] To a solution of trimethylacetic acid (0.169 mL, 0.00147 mol) in ethanol (6 mL, 0.1 mol) was added cesium carbonate (0.24 g, 0.00073 mol), and the resulting mixture was stirred for 2 hours. The solvent was removed in vacuo to afford cesium pivalate.
[0216] To a solution of 2-chloro-1-(1-[2-(trimethylsilyl)ethoxy]methyl-1 H-pyrrolo[2,3-b]pyridin-4-yl)ethanone (prepared, e.g., as in Ex. 50, Step 1) (0.054 g, 0.00017 mol) in DMF (1.8 mL, 0.023 mol) was added cesium pivalate (0.0389 g, 0.000166 mol) and the reaction was stirred at room temperature for 16 hours. Ammonium acetate (0.45 g, 0.0058 mol) was added, and the reaction was heated in the microwave to 170 °C for 5 minutes. Water was added and the product was extracted with MTBE. The combined organic extracts were dried over sodium sulfate, then filtered and concentrated. The crude residue was purified by flash column chromatography (2.5% MeOH/DCM) to yield 4-(2-tert-butyl-1H-imidazol-5-yl)-1-[2-(trimethylsilyl)ethoxy]methyl-1 H-pyrrolo[2,3-b]pyridine (32 mg, 52%). 1H NMR (400 MHz, CDCI3): δ 8.31 (d, 1H), 7.50 (s, 1H), 7.40 (d, 1H), 7.37 (d, 1H), 6.94 (d, 1H), 5.69 (s, 2H), 3.52 (dd, 2H), 1.46 (s, 9H), 0.90 (dd, 2H), -0.08 (s, 9H); MS(ES):371(M+1).
Step 2. 4-(2-tert-butyl-1-methyl-1 H-imidazol-4-yl)-1-[2-(trimethylsilyl)ethoxy]methyl-1 H-pyrrolo-[2,3-b]pyridine [0217] To a mixture of 4-(2-tert-butyl-1 H-imidazol-5-yl)-1-[2-(trimethylsilyl)ethoxy]methyl-1 H-pyrrolo[2,3-b]pyridine (0.019 g, 0.000051 mol) and potassium carbonate (0.15 g, 0.0011 mol) in DMF (3 mL, 0.04 mol) was added methyl iodide (0.01 mL, 0.00015 mol) in two portions over 48 hours. Water was then added and the product was extracted with MTBE. The combined extracts were dried with sodium sulfate, filtered, and concentrated in vacuo, then purified by silica gel chromatography (20% ethyl acetate/hexanes) to afford 4-(2-tert-butyl-1 -methyl-1 H-imidazol-4-yl)-1 -[2-(trimethylsi-lyl)ethoxy]methyl-1H-pyrrolo[2,3-b]pyridine (10 mg, 51%). 1H NMR (400 MHz, CDCI3): δ 8.37 (d, 1H), 7.54 (d, 1H), 7.44-7.22 (m, 2H), 7.19 (d, 1H), 5.78 (s, 2H), 3.93 (s, 3H), 3.60 (dd, 2H), 1.61 (s, 9H), 0.98 (dd, 2H), 0.00 (s, 9H); MS(ES):385(M+1).
Step 3.
[0218] Asolution or 4-(2-tert-butyl-1-methyl-1 H-imidazol-4-yl)-1-[2-(trimethylsilyl)-ethoxy]-methyl-1 H-pyrrolo[2,3-b]py-ridine (0.010 g, 0.000026 mol) in TFA (3 mL, 0.04 mol) was stirred for 2 hours. Then the excess TFA was evaporated and the residue was stirred in methanol (3 mL, 0.07 mol) and NH4OH (1 mL) for 16 hours. The solvents were removed and the product was purified by preparative-HPLC (C18 eluting with a gradient of ACN/H20 containing 0.1% TFA) to afford 4-(2-tert-butyl-1-methyl-1 H-imidazol-4-yl)-1H-pyrrolo[2,3-b]pyridine, trifluoroacetate salt (9 mg, 90%). 1H NMR (400 MHz, d6-DMSO): δ 12.24 (s, 1H), 8.38 (brs, 1H), 8.24 (s, 1H), 7.70-7.63 (m, 2H), 7.08 (brs, 1H), 2.55 (s, 3H), 1.51 (s, 9H); MS(ES):255(M+1 ).
[0219] Additional analogs were prepared as shown in Table 2 using analogous procedures to those described in Example 46 with different starting materials such as alternative carboxylic acids in Step 1. When the analogs were obtained as the free base, the product was obtained by preparative-HPLC (C18 eluting with a gradient of ACN/H20 containing 0.15% NH4OH). The results are summarized in Table 2 according to the following structure:
Example 50: 4-(2-Phenyl-1,3-thiazol-4-yl)-1H-pyrrolo[2,3-b]pyridine trifluoroacetate salt [0220]
Step 1.2-Chloro-1-(1-[2-(trimethylsilyl)ethoxy]methyl-1H-pyrrolo[2,3-b]pyridin-4-yl)ethanone [0221] To a solution of 4-bromo-1-[2-(trimethylsilyl)ethoxy]methyl-1H-pyrrolo[2,3-b]pyridine (2.05 g, 0.00626 mol) in THF (10 mL, 0.123 mol) at 0 °C was added dropwise a solution of isopropylmagnesium chloride in ether (2.0 M, 9.4 mL). The mixture was allowed to warm to room temperature and stirred for 4 hours. This mixture was then transferred via cannula to a solution of 2-chloro-N-methoxy-N-methylacetamide (2.84 g, 0.0207 mol) in THF (10 ml). After 30 minutes reaction time, the solution was quenched by the addition of saturated ammonium chloride aqueous solution. The product was extracted with ethyl acetate, the combined organic extracts were washed with brine, dried over Na2S04, filtered and concentrated. The crude residue was purified by flash column chromatography (0-20% ethyl acetate/hexanes) to afford 2-chloro-1-(1-[2-(trimethylsilyl)-ethoxy]methyl-1H-pyrrolo[2,3-b]pyridin-4-yl)ethanone (711 mg, 35%). 1H NMR (400 MHz, CDCI3): δ 8.56 (d, 1H), 7.66 (d, 1H), 7.60 (d, 1H), 7.23 (d, 1H), 5.80 (s, 2H), 4.91 (s, 2H), 3.60 (dd, 2H), 0.98 (dd, 2H), 0.01 (s, 9H); MS(ES):325(M+1).
Step 2. 4-(2-Phenyl-1,3-thiazol-4-yl)-1H-pyrrolo[2,3-b]pyridine trifluoroacetate salt [0222] A solution of 2-chloro-1-(1-[2-(trimethylsilyl)ethoxy]methyl-1H-pyrrolo[2,3-b]pyridin-4-yl)-ethanone (0.050 g, 0.00015 mol) and benzenecarbothioamide (0.031 g, 0.00022 mol) in ethanol (2 mL, 0.03 mol) was heated to reflux for 1 hour. The solvent was removed in vacuo. Ethyl acetate was added, and the resulting solid was isolated by filtration. The crude solid was stirred with TFA for 1 hour, then excess TFA was removed in vacuo. The crude residue was then stirred with aq. NH4OH and MeOH for 16 hours. The solvent was removed and the product was purified by preparative-HPLC (C18 eluting with a gradient of ACN/H20 containing 0.1% TFA) to afford 4-(2-phenyl-1,3-thiazol-4-yl)-1 H-pyrro-lo[2,3-b]pyridine as the trifluoroacetate salt (11 mg, 18%). 1H NMR (400 MHz, d6-DMSO): δ 12.01 (s, 1H), 8.58 (s, 1H), 8.39 (brs, 1H), 8.13-8.07 (m, 2H), 7.81 (d, 1H), 7.67-7.64 (m, 1H), 7.62-7.52 (m, 3H), 7.22 (d, 1H); MS(ES):278(M+1).
Example 51: N-Methyl-N-propyl-4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1,3-thiazol-2-amine, trifluoroacetate salt [0223]
Step 1. N-Methyl-N-propylthiourea [0224] N-Methyl-N-propylamine (0.501 mL, 0.00488 mol) was added to a solution of 1,1’-thiocarbonyldiimidazole (0.957 g, 0.00537 mol) in THF (9 mL, 0.1 mol), and the resulting solution was stirred for 16 hours. The intermediate from the reaction mixture was isolated by silica gel chromatography (5% MeOH in DCM) and this intermediate was stirred with ammonia (7M solution in MeOH) (6 mL)for48 hours. The solvent was removed In vacuo. N-methyl-N-propylthiourea was obtained after flash column chromatography (4% MeOH in DCM).
Step 2.
[0225] A solution of 2-chloro-1-(1-[2-(trimethylsilyl)ethoxy]methyl-1 H-pyrrolo[2,3-b]pyridin-4-yl)-ethanone (0.050 g, 0.00015 mol) and N-methyl-N-propylthiourea (0.030 g, 0.00022 mol) in ethanol (2 mL, 0.03 mol) was heated to reflux for 2 hours. Then, the ethanol was removed In vacuo and the residue was dissolved in 2 mL TFA and stirred for 40 minutes. The excess TFA was removed In vacuo and the residue was dissolved in 3 mL of MeOH. To this was added 0.5 mL of NHgOH and 100 μί of ethylenediamine, and the resulting solution was stirred for 16 hours. Solvent was removed, then water was added to give a white precipitate which was purified by preparative-HPLC (C18 eluting with a gradient of ACN/H20 containing 0.1% TFA) to afford N-methyl-N-propyl-4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1,3-thiazol-2-amine as the trifluoroacetate salt (39 mg, 67%). 1H NMR (300 MHz, CD3OD): δ 8.46-8.12 (br s, 1H), 7.92 (br s, 1H), 7.72 (s, 1H), 7.63 (d, 1H), 7.45 (brs, 1H), 3.56 (t, 2H), 3.20 (s, 3H), 1.78 (dq, 2H), 1.00 (t, 3H); MS(ES):273(M+1).
[0226] Additional aminothiazole analogs were prepared by procedures analogous to those described in Example 51, using different starting materials such as alternative thioureas in Step 2. In Examples 52 and 53, the white precipitate obtained by the procedure of Example 51 was isolated by filtration, washed with water and dried under high vacuum to afford the analogs as the free amine. The results are summarized in Table 3 according to the following structure:
Example 54: 4-(2-Phenyl-1,3-thiazol-5-yl)-1H-pyrrolo[2,3-b]pyridine trifluoroacetate salt [0227]
Step 1. (2-Phenyl-1,3-thiazol-5-yl)boronic acid [0228] To a solution of n-butyllithium in hexane (1.6 M, 2.1 mL) in ether (20 mL) at -78 °C, a solution of 2-phenyl-1,3-thiazole (449 mg, 0.00278 mol) in ether (5 mL) was added dropwise. The mixture was stirred for one hour at -78 °C followed by the addition of boric acid trimethyl ester (0.949 mL, 0.00835 mol). The mixture was stirred at -78 °C for 15 minutes, then was allowed to warm to room temperature and stirred for an additional 40 minutes. Saturated NH4CI aqueous solution was added, followed by 1.0 N aqueous HCI. The acidified mixture was stirred for 15 minutes, and the desired product was extracted with four portions of DCM containing 15% isopropanol. The combined organic extracts were dried over sodium sulfate and concentrated to give 566 mg of a white solid containing the desired (2-phenyl-1,3-thiazol-5-yl)boronic acid as a mixture with 2-phenyl-1,3-thiazole. This mixture was used in Step 2 without further purification. MS(ES):206(M+1).
Step 2.
[0229] To a mixture of (2-phenyl-1,3-thiazol-5-yl)boronic acid (75.0 mg, 0.000366 mol) and 4-bromo-1-[2-(trimethyls-ilyl)ethoxy]methyl-1H-pyrrolo[2,3-b]pyridine (80 mg, 0.000244 mol) in DMF (4 mL, 0.0516 mol) was added a solution of potassium carbonate (101 mg, 0.000732 mol) in water (1 mL, 0.0555 mol). The mixture was purged with a steady stream of nitrogen for 15 minutes.
[0230] Tetrakis(triphenylphosphine)palladium(0) (20 mg, 0.000018 mol) was added and the resulting mixture was heated to 125 °Cfor 30 minutes. The product was purified by preparative-HPLC (C18 eluting with a gradient of ACN/H20 containing 0.1% TFA) to afford 12 mg of a yellow solid containing the desired product as the major component. The mixture was stirred in TFA (1 mL) for 1 hour. Then excess TFA was removed in vacuo and the resulting residue was stirred with 2 mL MeOFI, 0.5 mL NFi4OH and 100 μί ethylenediamine for 16 hours. The product was isolated by pre-parative-FIPLC (C18 eluting with a gradient of ACN/Fi20 containing 0.1% TFA) to afford 4-(2-phenyl-1,3-thiazol-5-yl)-1 FI-pyrrolo[2,3-b]pyridine trifluoroacetate salt (5 mg, 5%). 1H NMR (400 MFIz, CD3OD): δ 8.64 (s, 1H), 8.34 (d, 1H), 8.10-8.04 (m, 2H), 7.73 (d, 1H), 7.71 (d, 1H), 7.56-7.51 (m, 3H), 7.14 (d, 1H); MS(ES):278(M+1).
Example 55: Ethyl 2-methyl-2-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]propanoate trifluoroacetate salt (55a) AND 2-Methyl-2-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]propanoic acid (55b) [0231]
[0232] 4-(1 H-Pyrazol-4-yl)-1 -[2-(trimethylsilyl)ethoxy]methyl-1 H-pyrrolo[2,3-b]pyridine (60 mg, 0.00019 mol) was dissolved in DMF (1.5 mL), and the solution was cooled to 0 °C with a cold bath. Sodium hydride (15 mg, 0.00038 mol) was added. After stirring for 10 min, 2-bromo-2-methyl-propanoic acid ethyl ester (42 μΙ-, 0.00028 mol) was added. The cold bath was then removed and the reaction mixture was allowed to warm to room temperature over 1 hour. The reaction mixture was quenched with saturated ammonium chloride solution. More water was added, and the product was extracted with MTBE. The combined extracts were dried over sodium sulfate, filtered and concentrated. The residue was dissolved in 2 mL TFA and stirred for 1 h. Then excess TFA was removed in vacuo and the resulting residue was stirred in 2 mL EtOFI containing 0.6 mL NFI4OH solution for 16 hours. Volatiles were removed, and purification of the mixture was carried out via preparative-HPLC (C18 eluting with a gradient of ACN/H20 containing 0.1 % TFA) afforded ethyl 2-methyl- 2-[4-(1H-pyrrolo(2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]propanoate trifluoroacetate salt (13 mg, 17%): 1FINMR (300 MHz, d6-DMSO): δ 12.03 (s, 1H), 8.67 (s, 1H), 8.31-8.19 (m, 2H), 7.59 (t, 1H), 7.48 (d, 1H), 6.98 (brs, 1H), 4.10 (q, 2H), 1.84 (s, 6H), 1.12 (t, 3H); MS(ES):299(M+1) and 2-methyl-2-[4-(1H-pyrrolo[2,3-blpyridin-4-yl)-1H-pyrazol-1-yl]propanoic acid (27 mg, 53%): 1H NMR (300 MHz, d6-DMSO): δ 12.04 (s, 1H), 8.64 (s, 1H), 8.26 (s, 2H), 7.59 (brs, 1 H), 7.48 (d, 1H), 6.99 (brs, 1H), 1.83 (s, 6H); MS(ES):271(M+H).
Example 56: 2-Methyl-2-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl)propanamide [0233]
[0234] A mixture of 2-methyl-2-[4-(1 H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]propanoicacid (23 mg, 0.000085 mol) and Ν,Ν-carbonyldiimidazole (CDI) (21 mg, 0.00013 mol) in 2 mL of DMF was stirred for 3 hours. An excess of solid NH4CI and TEA was added to the mixture and this was stirred for 3 hours. The majority of solvent was removed in vacuo, and the crude residue was purified by preparative-HPLC (C18 eluting with a gradient of ACN/H20 containing 0.1 % TFA) followed by re-purification via preparative-HPLC (C18 eluting with a gradient of ACN/H20 containing 0.15% NH4OH) to afford 2-methyl-2-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]propanamide (6 mg, 26%). 1H NMR (400 MHz, d6-DMSO): δ 11.63 (s, 1H), 8.44 (s, 1H), 8.16 (s, 1H), 8.14 (s, 1H), 7.47 (t, 1H), 7.29 (d, 1H), 7.21 (s, 1H), 6.93 (s, 1H), 6.80 (dd, 1H), 1.77 (s, 6H); MS(ES):270(M+1).
Example 57: Ethyl 3-methyl-3-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]butanoate trifluoroacetate salt [0235]
Step 1. Ethyl 3-methyl-3-[4-(1 -[2-(trimethylsilyl)ethoxy]methyl-1 H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1 -yl]butanoate [0236] 4-(1H-Pyrazol-4-yl)-1-[2-(trimethylsilyl)ethoxy]methyl-1H-pyiTolo[2,3-b]pyridine (220 mg, 0.0006996 mol) and 3-methyl-2-butenoic acid ethyl ester (292 μί, 0.00210 mol) were dissolved in DMF (10 mL). Cesium carbonate (912 mg, 0.00280 mol) was added and the resulting mixture was stirred at room temperature for 3 hours. The reaction mixture was diluted with water, and the product was extracted with MTBE several times. The combined extracts were dried over sodium sulfate and concentrated. The crude residue was purified by flash column chromatography (0-60% EtOAc/Hex-anes) to afford ethyl 3-methyl-3-[4-(1-[2-(trimethylsilyl)ethoxy]methyl-1H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1 -yl]bu-tanoate (244 mg, 79%). 1H NMR(300MHz, CDCI3): δ 8.37 (d, 1H), 8.11 (s, 1H),8.09 (s, 1H), 7.45 (d, 1H),7.24(d, 1H), 6.79 (d, 1H), 5.77 (s, 2H), 4.10 (q,2H), 3.62 (dd, 2H), 3.04 (s, 2H), 1.88 (s, 6H), 1.20 (t, 3H), 0.98 (dd,2H),0.00 (s, 9H); MS(ES):443(M+1).
Step 2.
[0237] Ethyl 3-methyl-3-[4-(1-[2-(trimethylsilyl)ethoxy]methyl-1H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]bu-tanoate (20 mg, 0.0000452 mol) was stirred in 1 mL TFA for 1 hour. Then excess TFA was removed in vacuo. The residue was stirred for 16 hours in 2 mL MeOH containing 0.5 mL NH4OH. Evaporation of the volatiles was followed by purification by preparative-HPLC (C 18 eluting with a gradient of ACN/H20 containing 0.1% TFA) to afford ethyl 3-methyl-3-(4-(1 H-pyrrolo[2,3-b]-pyridin-4-yl)-1 H-pyrazol-1 -yl]butanoate, trifluoroacetate salt (5 mg, 26%). 1H NMR (400 MHz, dg-DMSO): δ 12.19 (s, 1H), 8.61 (br s, 1H), 8.34-8.22 (br m, 2H), 7.62 (brs, 1H), 7.51 (brd, 1H), 7.02 (brs, 1H), 3.91 (q, 2H), 2.96 (s, 2H), 1.70 (s, 6H), 1.02 (t, 3H); MS(ES):313(M+1).
Example 58: 3-Methyl-3-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]butan-1-ol trifluoroacetate salt [0238]
[0239] To a solution of ethyl 3-methyl-3-[4-(1-[2-(trimethylsilyl)ethoxy]methyl-1 H-pyrrolo[2,3-b]-pyridin-4-yl)-1 H-pyra-zol-1-yl]butanoate (213 mg, 0.000481 mol) in THF (5 mL, 0.0616 mol) at -78 °C was added diisobutylaluminum hydride in DCM (1.00 M, 1.1 mL) dropwise. The reaction mixture was stirred for 3 hours during which time the reaction slowly warmed to -10 °C. To the mixture at -10 °C was carefully added K/Na tartrate tetrahydrate in water. The mixture was stirred for 2 hours, then was extracted with three portions of ethyl acetate. The combined organic extracts were washed with two portions of water and one portion of brine, then dried over sodium sulfate, filtered and concentrated to afford 3-methyl-3-[4-(1-[2-(trimethylsilyl)ethoxy]methyl-1H-pyrrolo[2,3-b]-pyridin-4-yl)-1 H-pyrazol-1-yl]butan-1-ol (185 mg, 96%), which was used without further purification. A portion of the alcohol so obtained (15 mg, 0.000037 mol) was stirred in TFA (1 mL) for 2 hours. The TFA was removed in vacuo and the residue was stirred with 2 mL MeOH containing 0.5 mL NH4OH for 16 hours. Volatiles were removed and the product was purified by preparative-HPLC (C18 eluting with a gradient of ACN/H20 containing 0.1% TFA) to afford 3-methyl-3-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]bu- tan-1-ol as the trifluoroacetate salt (8.0 mg, 57%). 1H NMR (300 MHz, d6-DMSO): δ 12.17 (s, 1H), 8.58 (br s, 1H), 8.32-8.22 (br m, 2H), 7.62 (br s, 1H), 7.53 (br d, 1H), 7.03 (br s, 1H), 3.25 (t, 2H), 2.07 (t, 2H), 1.62 (s, 6H); MS(ES):271(M+1).
Example 59: 4-Methyl-4-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]pentanenitrile trifluoroacetate salt [0240]
Step 1. 4-Methyl-4-[4-(1-[2-(trimethylsilyl)ethoxy]methyl-1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]pentanenitrile [0241] TEA (38.0 μΙ_, 0.000273 mol) and methanesulfonyl chloride (21.1 μΙ_, 0.000273 mol) were added sequentially to a solution of 3-methyl-3-[4-(1-[2-(trimethylsilyl)ethoxy]methyl-1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]butan-1-ol (prepared as in Example 58) (81 mg, 0.00020 mol) in DCM (4 mL, 0.05 mol) at 0° C. The reaction mixture was held at this temperature for 1.5 hours, then was quenched by the addition of water. The reaction mixture was extracted with DCM four times. The combined extracts were dried over sodium sulfate, filtered and concentrated to afford crude 3-methyl-3-[4-(1-[2-(trimethylsilyl)ethoxy]methyl-1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]butyl methanesulfonate (87 mg). MS(ES):479(M+1).
[0242] A mixture of 3-methyl-3-[4-(1-[2-(trimethylsilyl)ethoxy]methyl-1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]butyl methanesulfonate (42 mg, 0.000088 mol) and potassium cyanide (46 mg, 0.000702 mol) in DMF (1 mL) was heated in the microwave reactor for 30 min at 125 °C followed by additional 30 min at 135 °C. The mixture was then diluted with water, and the product was extracted with three portions of MTBE. The combined extracts were dried over sodium sulfate, filtered and concentrated to give 61 mg of crude 4-methyl-4-[4-(1-[2-(trimethylsilyl)ethoxy]methyl-1H-pyrrolo-[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]pentanenitrile, which was used without further purification. MS(ES):410(M+1).
Step 2.
[0243] 4-Methyl-4-[4-(1-[2-(trimethylsilyl)ethoxy]methyl-1H-pyrrolo [2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]pentanenitrile (57 mg, 0.00014 mol) was stirred in DCM (4 ml) and TFA (1 mL) for 2 hours. The solvents were removed in vacuo and the residue was stirred in 2 mL MeOH containing 0.2 mL ethylenediamine for 16 hours. The volatiles were evaporated and the product was isolated from the reaction mixture by preparative-HPLC (C18 eluting with a gradient of ACN/H2Ocontaining 0.1% TFA) affording 4-methyl-4-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]pentanenitrile as the trifluoroacetate salt (10 mg, 18%). 1H NMR (400 MHz, d6-DMSO): δ 12.09 (s, 1H), 8.58 (s, 1H), 8.29 (s, 1H), 8.25 (d, 1H), 7.60 (t, 1H), 7.48 (d, 1H), 7.00 (brs, 1H), 2.33-2.21 (m, 4H), 1.61 (s, 6H); MS(ES):280(M+1).
Example 60: 4-Methyl-4-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]pentanamide trifluoroacetate salt [0244]
[0245] The crude 4-methyl-4-[4-(1-[2-(trimethylsilyl)ethoxy]methyl-1 H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]pen-tanenitrile (36 mg, 0.000088 mol, see preparation in Example 59), was stirred in TFA (2 mL) for 1 hour. The mixture was concentrated to remove excess TFA, and the resulting residue was stirred in 2 mL methanol containing 0.5 mL NH4OH for 16 hours. The product was purified by preparative-HPLC (C18 eluting with a gradient of ACN/H20 containing 0.1% TFA) to afford 4-methyl-4-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]pentanamide as the trifluoroacetate salt (21 mg, 58%). 1H NMR(400 MHz, d6-DMSO): δ 12.18 (s, 1H), 8.60 (s, 1H), 8.33-8.21 (m, 2H), 7.62 (brs, 1H), 7.53 (d, 1H), 7.22 (brs, 1H), 7.04 (brs, 1H), 6.71 (brs, 1H), 2.14-2.07 (m, 2H), 1.86-1.79 (m, 2H), 1.58 (s, 6H); MS(ES):298(M+1).
Example 61: (3S)-3-[4-(1H-Pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]butanenitrile trifluoro-acetate salt, AND (3R)-3-[4-(1H-Pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]butanenitrile trifluoroacetate salt [0246]
[0247] To a solution of 4-(1H-pyrazol-4-yl)-1-[2-(trimethylsilyl)ethoxy]methyl-1H-pyrrolo[2,3-b]pyridine (0.050 g, 0.00016 mol) in ACN were added 2-butenenitrile (0.014 mL, 0.00017 mol) and DBU (0.029 mL, 0.00020 mol). The resulting mixture was stirred for 16 hours. Then the volatiles were evaporated and the residue was dissolved in ethyl acetate. The resulting solution was washed successively with 1.0 N HCI, water, and brine, then was dried over sodium sulfate, filtered and concentrated. To obtain the enantiomers in substantially pure form, Method A (vide infra) was used.
[0248] The crude residue was dissolved in TFA (7 mL, 0.09 mol) and the solution was stirred for 1 hour. Then excess TFA was evaporated and the residue was then stirred with ethylenediamine (0.1 mL, 0.001 mol) in methanol (4 mL, 0.09 mol) for 16 hours. The mixture was concentrated, and the product was purified by preparative-HPLC (C18 eluting with a gradient of ACN/H20 containing 0.1% TFA) to afford 3-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]butanenitrile trifluoroacetate salt (35 mg, 61%). 1H NMR (300 MHz, d6-DMSO): δ 12.16 (s, 1H), 8.73 (s, 1H), 8.32 (s, 1H), 8.28 (d, 1H), 7.65-7.61 (m, 1H), 7.48 (d, 1H), 6.99 (d, 1H), 4.86 (q, 1H), 3.17 (d, 2H), 1.57 (d, 3H); MS(ES):252(M+1).
[0249] Additional analogs were prepared by procedures analogous to those described in Example 61 using different starting materials for alkylation of the pyrazole ring. For example, the α,β-unsaturated nitriles were prepared by procedures analogous to the following, illustrated for (2E)- and (2Z)-hexenenitrile: To a solution of 1.00 M potassium tert-butoxide in THF at 0 °C (24.2 mL) was added a solution of diethyl cyanomethylphosphonate (4.10 mL, 0.025 mol) in THF (30 mL) dropwise. The bath was removed and the solution was allowed to warm to room temperature. After reaching room temperature, the solution was re-cooled to 0° C and a solution of butánál (2.00 mL, 0.023 mol) in THF (7 mL) was added dropwise. The reaction mixture was allowed to warm to room temperature and stir overnight. The mixture was diluted with ethyl acetate and water. The layers were separated and the aqueous layer was extracted with three portions of ethyl acetate. The combined organic extracts were washed with brine, dried over sodium sulfate, filtered and concentrated. This afforded 1.6 g of a crude mixture containing both (2E)- and (2Z)-hexenenitrile, which was used without further purification in the subsequent alkylation step. 1H NMR (400 MHz, CDCI3): δ 6.72 (dt, 1H trans olefin), 6.48 (dt, 1H cis olefin), 5.34 (dt, 1H trans olefin), 5.31-5.30 (m, 1H cis olefin).
[0250] Where it was desirable to obtain the enantiomers in substantially pure form, chiral separation was performed by one of the following methods: A) The separation was performed on the SEM-protected intermediate after silica gel chromatography (ethyl acetate/hex-anes) by preparative chiral HPLC (OD-H column, eluting with 15% ethanol in hexanes); B) The separation was performed on the deprotected free base by preparative chiral HPLC (OD-H column, eluting with 15% ethanol in hexanes); C) The separation was performed on the SEM-protected intermediate after silica gel chromatography (ethyl acetate/hex-anes) by preparative chiral HPLC (AD-H column, eluting with 10% ethanol in hexanes); D) The separation was performed on the SEM-protected intermediate after silica gel chromatography (ethyl acetate/hex-anes) by preparative chiral HPLC (AD-H column, eluting with 15% ethanol in hexanes); E) The separation was performed on the SEM-protected intermediate after silica gel chromatography (ethyl acetate/hex-anes) by preparative chiral HPLC (OD-H column, eluting with 20% ethanol in hexanes; or F) The separation was performed on the SEM-protected intermediate after silica gel chromatography (ethyl acetate/hex-anes) by preparative chiral HPLC (OD-H column, eluting with 30% ethanol in hexanes. An OD-H column refers to Chiralcel OD-H from Chiral Technologies, Inc 3x25 cm, 5 μΓΤΐ. An AD-H column refers to ChiralPak AD-H from Chiral Technologies, Inc. 2x25 cm, 5 μίτι. The results are summarized for compounds in Table 4 below.
Table 4
Example 65: (3R)-3-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]hexanenitrile trifluoroacetate salt and (3S)3-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]hexanenitrile trifluoroacetate salt [0251]
Step 1. 4-Chloro-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidine [0252] To a solution of 4-chloropyrrolo[2,3-d]pyrimidine (0.86 g, 0.0056 mol) in DMF (20 mL, 0.2 mol) at 0 °C was added sodium hydride (0.27 g, 0.0067 mol) in several portions. The reaction mixture was stirred for an additional 45 minutes followed by a dropwise addition of ß-(trimethylsilyl)ethoxy]-methyl chloride (1.2 mL, 0.0067 mol). The resulting reaction mixture was stirred at 0 °C for 45 min, then was quenched with water and extracted with ethyl acetate. The organic extract was washed with water, brine, dried over sodium sulfate, filtered and concentrated to give an oil. The crude residue was purified by flash column chromatography (0-15% ethyl acetate/hexanes) to yield 4-chloro-7-[2-(tri-methylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidine (1.40 g, 88%). 1HNMR (400 MHz, CDCI3): δ 8.71 (s, 1H), 7.46 (d, 1H), 6.72 (d, 1H), 5.71 (s, 2H), 3.59 (dd, 2H), 0.97 (dd, 2H), 0.00 (s, 9H); MS(ES):284(M+1).
Step 2. 4-( 1H-Pyrazol-4-yl)-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidine [0253] To a mixture of 4-chloro-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidine (1.4 g, 0.0049 mol) and 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (1.4 g, 0.0074 mol) in DMF (40 mL, 0.5 mol) was added potassium carbonate (2.0 g, 0.015 mol) in 15 mL of water. The mixture was purged with a steady stream of nitrogen for 15 minutes. Tetrakis(triphenyiphosphine)pailadium(0) (0.41 g, 0.00036 mol) was added and the reaction was heated to 125 °C for 30 min. The mixture was allowed to cool then diluted with ethyl acetate. The diluted reaction mixture was washed with water, brine, dried over Na2S04 and concentrated to give a solution in a small volume of DMF (about 2-3 mL). Water was added, causing the material to form a gum on the walls of the flask. Then water was decanted, and the solids were dissolved in ethyl acetate. The solution was dried over Na2S04, and concentrated in vacuo to afford a yellow solid. The product was triturated with ethyl ether to yield 4-(1H-pyrazol-4-yl)-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyr-rolo[2,3-d]pyrimidine as a white powder which was dried under vacuum (1 g, 60%). 1H NMR (300 MHz, CDCI3): δ 10.80 (brs, 1H), 8.93 (s, 1H), 8.46 (s, 2H), 7.46 (d, 1H), 6.88 (d, 1H), 5.73 (s, 2H), 3.61 (dd, 2H), 0.98 (dd, 2H), 0.00 (s, 9H); MS(ES):316(M+1).
Step 3.
[0254] To a solution of 4-(1H-pyrazol-4-yl)-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidine (0.050 g, 0.00016 mol) in ACN (1 mL, 0.02 mol) was added hex-2-enenitrile (0.100 g, 0.00105 mol) (as a mixture of cis and trans isomers), followed by DBU(60 μί, 0.0004 mol). The resulting mixture was stirred at room temperature for 16 hours. The ACN was removed in vacuo. The crude residue was dissolved in ethyl acetate, and was washed with 1.0 N HCI, brine, dried over Na2S04 and concentrated. The crude residue was purified by flash column chromatography (0-70% EtOAc/Hexane) to afford 56 mg of product, which was stirred with 1:1 TFA/DCM for 1 hour and the solvents were evaporated. The resulting product was stirred with methanol (4 mL, 0.1 mol) containing ethylenediamine (0.1 mL, 0.001 mol) overnight. The solvent was evaporated and the product was purified by preparative-HPLC (C18 eluting with a gradient of ACN/H20 containing 0.1 % TFA) to afford 3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1 -yl]hexanenitrile as the trifluroacetate salt. Where desired, the enantiomers were isolated in substantially pure form by Method A described above for Example 61.1H NMR (300 MHz, CD3OD): δ 8.93 (s, 1H), 8.88 (s, 1H), 8.52 (s, 1H), 7.85 (d, 1H), 7.28 (d, 1H), 4.87-4.77 (m, 1H), 3.26-3.05 (m, 2H), 2.20-2.05 (m, 1H), 2.00-1.86 (m, 1H), 1.40-1.10 (m, 2H), 0.95 (t, 3H); MS(ES):281(M+1).
Example 67: (3R)- and (3S)-3-Cyclopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile [0255]
Step 1. (2E)- and (2Z)-3-Cyclopentylacrylonitrile [0256] To a solution of 1.0 M potassium tert-butoxide in THF (235 mL) at 0°C was added dropwise a solution of diethyl cyanomethylphosphonate (39.9 mL, 0.246 mol) in THF (300 mL). The cold bath was removed and the reaction was warmed to room temperature followed by recooling to 0 °C, at which time a solution of cyclopentanecarbaldehyde (22.0 g, 0.224 mol) in THF (60 mL) was added dropwise. The bath was removed and the reaction warmed to ambient temperature and stirred for 64 hours. The mixture was partitioned between diethyl ether and water, the aqueous was extracted with three portions of ether, followed by two portions of ethyl acetate. The combined extracts were washed with brine, then dried over sodium sulfate, filtered and concentrated in vacuo to afford a mixture containing 24.4 g of olefin isomers which was used without further purification (89%). 1H NMR (400 MHz, CDCI3): δ 6.69 (dd, 1H, trans olefin), 6.37 (t, 1H, cis olefin), 5.29 (dd, 1H, trans olefin), 5.20 (d, 1H, cis olefin), 3.07-2.95 (m, 1 H, cis product), 2.64-2.52 (m, 1H, trans product), 1.98-1.26 (m, 16H).
Step 2. (3R)- and (3S)-3-Cyclopentyl-3-[4-(7-[2-(trimethylsilyl)ethoxy\methyl-7H-pyrrolo[2,3-d]-pyrimittin-4-yl)-1H-pyra-zol-1 -yljpropanenitrile [0257] To a solution of 4-(1H-pyrazol-4-yl)-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]-pyrimidine (15.0 g, 0.0476 mol) in ACN (300 mL)was added 3-cyclopentylacrylonitrile(15g, 0.12 mol) (as a mixture of cis and trans isomers), followed by DBU (15 mL, 0.10 mol). The resulting mixture was stirred at room temperature overnight. The ACN was evaporated. The mixture was diluted with ethyl acetate, and the solution was washed with 1.0 N HCI. The aqueous layer was back-extracted with three portions of ethyl acetate. The combined organic extracts were washed with brine, dried over sodium sulfate, filtered and concentrated. The crude product was purified by silica gel chromatography (gradient of ethyl acetate/hexanes) to yield a viscous clear syrup, which was dissolved in ethanol and evaporated several times to remove ethyl acetate, to afford 19.4 g of racemic adduct (93%). The enantiomers were separated by preparative-HPLC, (OD-H, 15% ethanol/hexanes) and used separately in the next step to generate their corresponding final product. The final products (see Step 3) stemming from each of the separated enantiomers were found to be active JAK inhibitors; however, the final product stemming from the second peak to elute from the preparative-HPLC was more active than its enantiomer. 1H NMR (300 MHz, CDCI3): δ: 8.85 (s, 1H), 8.32 (s, 2H), 7.39 (d, 1H), 6.80 (d, 1H), 5.68 (s, 2H), 4.26 (dt, 1H), 3.54 (t, 2H), 3.14 (dd, 1H), 2.95 (dd, 1H), 2.67-2.50 (m, 1H), 2.03-1.88 (m, 1H), 1.80-1.15 (m, 7H), 0.92 (t, 2H), -0.06 (s, 9H); MS(ES):437 (M+1).
Step 3.
[0258] To a solution of 3-cyclopentyl-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]-pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile (6.5 g, 0.015 mol, R or S enantiomer as isolated above) in DCM (40 mL) was added TFA (16 mL) and this was stirred for 6 hours. The solvent and TFA were removed in vacuo. The residue was dissolved in DCM and concentrated using a rotary evaporator two further times to remove as much as possible of the TFA. Following this, the residue was stirred with ethylenediamine (4 mL, 0.06 mol) in methanol (30 mL) overnight. The solvent was removed in vacuo, water was added and the product was extracted into three portions of ethyl acetate. The combined extracts were washed with brine, dried over sodium sulfate, decanted and concentrated to afford the crude product which was purified by flash column chromatography (eluting with a gradient of methanol/DCM). The resulting mixture was further purified by preparative-HPLC/MS (C18 eluting with a gradient of ACN/H20 containing 0.15% NH4OH) to afford product (2.68 g, 58%). 1H NMR (400 MHz, D6-dmso): δ 12.11 (brs, 1H), 8.80 (s, 1H), 8.67 (s, 1H), 8.37 (s, 1H), 7.60 (d, 1H), 6.98 (d, 1H), 4.53 (dt, 1H), 3.27 (dd, 1H), 3.19 (dd, 1H), 2.48-2.36 (m, 1H), 1.86-1.76 (m, 1H), 1.68-1.13 (m, 7H); MS(ES):307(M+1).
[0259] Additional analogs provided in the following Tables were prepared by procedures analogous to those described in, for example, Examples 61 and 65, using different starting materials such as different α,β-unsaturated nitriles in Step 3. Isolation of the enantiomers in substantially pure form was achieved by the indicated chiral separation method described above (A-F) preceding Table 4. Where the product was isolated as the free amine, the product following deprotection was purified by preparative-HPLC (C18 eluting with a gradient of ACN/H20 containing 0.15% NH4OH) instead of pre-parative-HPLC (C18 eluting with a gradient of ACN/H20 containing 0.1% TFA). This is referred to as "modification G". The results are summarized in Table 5 according to the following structure:
Table 5
(continued)
Example 69: 4-{1-[(1S)-1-Methylbutyl]-1H-pyrazol-4-yl}-7H-pyrrolo[2,3-d]pyrimidine trifluoroacetate salt and 4-{1-[(1R)-1-Methylbutyl]-1H-pyrazol-4-yl}-7H-pyrrolo[2,3-d]pyrimidine trifluoroacetate salt [0260]
[0261] A solution of 4-(1H-pyrazol-4-yl)-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]-pyrimidine (0.050 g, 0.00016 mol) in DMF (2 mL, 0.02 mol) was cooled in an ice bath and to this was added sodium hydride (0.013 g, 0.00032 mol). The resulting mixture was stirred for 10 minutes, followed by an addition of 2-bromopentane (0.030 mL, 0.00024 mol). The cooling bath was then removed and the reaction was stirred at room temperature for 3 hours, at which time a further portion of 2-bromopentane (0.015 mL, 0.00012 mol) was added. After 45 minutes, water was added and the reaction mixture was extracted with three portions of ethyl acetate. The combined extracts were washed with brine, dried over sodium sulfate, filtered, and concentrated. The residue was stirred with TFA(3 mL, 0.04 mol) and DCM (3 mL, 0.05 mol) for 3.5 hours, then the solvent was removed in vacuo. The residue was then stirred with NH4OH (1.5 mL) in MeOH (4 mL) for 16 hours. The solvent was evaporated and the product was purified by preparative-HPLC (C18 eluting with a gradient of ACN/H20 containing 0.1 % TFA) to afford 4-[1-(1-methylbutyl)-1 H-pyrazol-4-yl]-7H- pyrrolo[2,3-d]pyrimidine as the trifluoroacetate salt (25 mg, 44%). 1H NMR (300 MHz, CD3OD): δ 8.83 (s, 1H), 8.75 (s, 1H), 8.43 (s, 1H), 7.77 (d, 1H), 7.24 (d, 1H), 4.63-4.50 (m, 1H), 2.07-1.91 (m, 1H), 1.88-1.74 (m, 1H), 1.58 (d, 3H), 1.38-1.09 (m, 2H), 0.93 (t, 3H); MS(ES):256(M+1).
[0262] Isolation of the enantiomers in substantially pure form was achieved by separation of the racemic free base (isolated by flash column chromatography after deprotection, eluting with a MeOH/DCM gradient) using HPLC (OD-H, eluting with 5% isopropanol/hexanes).
Example 69a: 4-Methyl-4-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]pentanenitrile [0263]
Step 1. Ethyl 3-methyl-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]bu-tanoate [0264] Asolution of 4-(1 H-pyrazol-4-yl)-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]-pyrimidine (12.1 g, 0.0384 mol), 2-butenoic acid, 3-methyl-, ethyl ester (16.0 mL, 0.115 mol) and DBU (14.3 mL, 0.0959 mol) in ACN (100 mL) was heated at reflux for 3.5 hours. The solvent was removed In vacuo. The residue was diluted with water, extracted with ethyl acetate, and the combined organic extracts were washed with saturated ammonium chloride, dried over sodium sulfate, and concentrated. The crude residue was purified by flash column chromatography (ethyl acetate/hexanes) to yield the desired product (15.5 g, 91%). 1H NMR (400 MHz, CDCI3): δ 8.83 (s, 1H), 8.36 (s, 1H), 8.27 (s, 1H), 7.37 (d, 1H), 6.80 (d, 1H), 5.66 (s, 2H), 4.03 (q, 2H), 3.54 (dd, 2H), 2.98 (s, 2H), 1.80 (s, 6H), 1.13 (t, 3H), 0.91 (dd, 2H), -0.07 (s, 9H); MS(ES):444(M+1).
Step 2. 3-Methyl-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]butan-1-ol [0265] To a solution of ethyl 3-methyl-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]-pyrimidin-4-yl)-1H-pyrazol-1-yl]butanoate (15.4 g, 0.0347 mol) in THF (151 mL) at -78 °C was added 1.00 M diisobutylaluminum hydride in DCM (84.5 mL) dropwise. The reaction was stirred for 2 hours with slow warming to-10 °C. The mixture was quenched with water, then was treated with potassium sodium tartrate tetrahydrate and water. The mixture was stirred for 1 hour, then was extracted with ethyl acetate. The extracts were washed with water and brine, then dried with sodium sulfate, filtered, and concentrated in vacuo. The crude residue was purified by flash column chromatography to yield the desired product (13.8 g, 99%). 1H NMR (300 MHz, CDCI3): δ 8.83 (s, 1H), 8.38 (s, 1H), 8.26 (s, 1H), 7.38 (d, 1H), 6.80 (d, 1H), 5.67 (s, 2H), 3.65 (dd, 2H), 3.54 (dd, 2H), 2.21 (t, 2H), 1.72 (s, 6H), 0.91 (dd, 2H), -0.07 (s, 9H); MS(ES):402(M+1).
Step 3. 3-Methyl-3-[4-(7H-pyrrolo[2,3-d]pyrlmidin-4-yl)-1 H-pyrazol-1-yl]butan-1 -ol [0266] A solution of 3-methyl-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]butan-1-ol (13.8 g, 0.0344 mol) in TFA (20 mL) was stirred for 1 hour. The mixture was then concentrated in vacuo and the residue was stirred for 2 hours in a mixture of methanol (30 mL), ammonium hydroxide (30 mL), and ethylene-diamine (8 mL). The mixture was then concentrated, and the residue was diluted with water and extracted with several portions of 15% IPA/CH2CI2. The combined extracts were dried over sodium sulfate and concentrated in vacuo to give 20 g of white solid. The solid was triturated with ether and the product was isolated by filtration to give the product as a white solid (7.75 g, 83%). 1H NMR (400 MHz, CDCI3): δ 9.99 (s, 1H), 8.83 (s, 1H), 8.39 (s, 1H), 8.28 (s, 1H), 7.38 (dd, 1H), 6.80 (dd, 1H), 3.66 (t, 2H), 2.72 (br s, 1H), 2.22 (t, 2H), 1.74 (s, 6H); MS(ES):272(M+1).
Step 4. 3-Methyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]butyl methanesulfonate [0267] A solution of 3-methyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]butan-1-ol (6.61 g, 0.0244 mol) in DCM (300 mL) at 0 °C was treated with TEA (3.74 mL, 0.0268 mol), followed by methanesulfonyl chloride (2.07 mL, 0.0268 mol). The reaction was stirred for 1 hour, and was then concentrated in vacuo. The crude residue was purified by flash column chromatography to afford the desired product (4.9 g, 57%). 1H NMR (400 MHz, d6-dmso): δ 12.45 (s, 1H), 9.50 (s, 1H), 9.35 (s, 1H), 8.83 (s, 1H), 7.79 (dd, 1H), 7.11 (dd, 1H), 4.75 (t, 1H), 3.30 (s, 3H), 2.85 (t, 1H), 1.75 (s, 6H); MS(ES):254(M-CH3S03H+1).
Step 5. 4 Methyl-4-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1Hpyrazol-1-yl]pentanenitrile [0268] 3-methyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]butyl methanesulfonate (2.97 g, 8.50 mmol), DMF (120 mL) and sodium cyanide (6.21 g, 0.127 mol) were distributed evenly into six 20 mL microwavable vessels, each of which was heated in the microwave reactor for 4000 seconds at 125 °C. The contents of the vials were combined and were diluted with 400 mL water and extracted with five 150 mL portions of ethyl acetate. The combined extracts were dried over sodium sulfate, and the solvent was removed in vacuo. The crude residue was purified by flash column chromatography to yield the desired product (1.40 g, 59%). 1H NMR (400 MHz, CDCI3): δ 9.52 (br s, 1 H), 8.83 (s, 1H), 8.34 (s, 1 H), 8.29 (s, 1H), 7.39 (dd, 1H), 6.81 (dd, 1H), 2.38 (dd, 2H), 2.16 (dd, 2H), 1.73 (s, 6H); MS(ES):281(M+1).
[0269] The analogs in Table 5a were prepared according to the above method described for Example 69a. For Example 69b, a conjugate acceptor was used and prepared as described in Perkin Trans. 1, 2000, (17), 2968-2976, and Steps 4&5 were performed before Step 3.
Table 5a
Example 69d: 3-Methyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]butanenitrile [0270]
Step 1. Seneclonltrlle [0271] To a solution of 1.0 M potassium tert-butoxide in THF (2.0 mL) at 0 °C was added a solution of diethyl cy-anomethylphosphonate (0.33 mL, 2.06 mmol) in THF (4 mL) dropwise. The cold bath was removed and the reaction was warmed to room temperature. The reaction was then re-cooled to 0 °C and acetone (0.20 mL, 2.81 mmol) was added dropwise. The cooling bath was then removed and the reaction was allowed to warm to room temperature and stir overnight. The reaction was diluted with water, the layers separated, and the aqueous extracted with ethyl acetate. The extracts were washed with brine, dried over sodium sulfate, filtered and concentrated. The product was used without further purification (339 mg, 67%). 1H NMR (300 MHz, CDCI3): δ 5.10 (br s, 1H),2.05(s, 3H), 1.92 (s, 3H).
Step 2. 3-Methyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]butanenitrile [0272] To a solution of 4-(1H-pyrazol-4-yl)-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]-pyrimidine (0.216 g, 0.684 mmol) in ACN (4 mL, 0.08 mol) was added crude senecionitrile (0.111 g, 1.37 mmol), followed by DBU (200 μί, 0.002 mol) and the resulting mixture was heated to 60 °C for 23 hours. The mixture was cooled to room temperature and the ACN was evaporated. The mixture was diluted with ethyl acetate and washed with dilute HCI and brine. The organic solution was dried over sodium sulfate, filtered and concentrated. Purification by silica gel chromatography (ethyl acetate/hexanes) afforded the desired product. 1H NMR (300 MHz, d6-dmso): δ 8.83 (s, 1H), 8.38 (s, 1H), 8.28 (s, 1H), 7.39 (d, 1H), 6.80 (d, 1H), 5.66 (s, 2H), 3.54 (dd, 2H), 3.08 (s, 2H), 1.84 (s, 6H), 0.91 (dd, 2H), -0.07 (s, 9H); MS(ES):397(M+1).
[0273] To a solution of this product in DCM at 0 °C was added TFA sufficient to comprise 20% of the total volume. The solution was stirred at this temperature for 30 min, then at ambient temperature for 2 hours and 15 minutes. The solvents were removed In vacuo and the residue was stirred with methanol (10 mL) and ethylenediamine (0.4 mL, 0.006 mol) overnight. The solvent was evaporated and the productwas purified by preparative-HPLC/MS (C18 column eluting with a gradient of ACN/H20 containing 0.15% NH4OH) to afford the product (25 mg, 14%). 1H NMR (300 MHz, d6-dmso): δ 12.08 (s, 1H), 8.68 (s, 2H), 8.39 (s, 1H), 7.59 (d, 1H), 7.05 (d, 1H, 3.32 (s, 2H), 1.73 (s, 6H); MS(ES):267(M+1).
[0274] Examples 69e and 69f in Table 5b were prepared by a method analogous to that described above for Example 69d, with unsaturated nitriles prepared either according to published literature procedures, or by the method in Step 1.
Table 5b
[0275] Additional analogs were prepared by procedures analogous to those described in Example 69, using different starting materials such as alternative bromide or mesylate compounds for the nucleophilic substitution step. Where the free amine was obtained as the product, the product was purified after deprotection either by silica gel chromatography (eluting with 5% methanol in DCM) or by preparative-HPLC (C18 eluting with a gradient of ACN/H20 containing 0.15% NH4OH). The results are summarized for compounds listed in Table 6.
Table 6
Example 74: (2Z)-3-Cyclopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-acrylonitrile [0276]
Step 1. 3-Cyclopentylprop-2-ynenitrile [0277] To a solution of cyclopentylacetylene (0.50 g, 5.3 mmol) in THF (5 mL) at-78 °C was added 2.5 M n-butyllithium in hexane (2.23 mL). The mixture was stirred for 15 min followed by the dropwise addition of phenyl cyanate (0.70 g, 5.8 mmol) in THF (3 mL). The reaction was warmed to room temperature. Into the reaction mixture was poured 6 N NaOH, and the mixture was stirred for 5 minutes. The product was extracted with diethyl ether. The extracts were washed with 6 N NaOH and with brine, then dried over sodium sulfate, decanted and the solvent was removed in vacuo to afford product (600 mg, 95%). 1H NMR (300 MHz, CDCI3): δ 2.81-2.68 (m, 1H), 2.07-1.54 (m, 8H).
Step 2. (2Z)-3-Cyclopentyl-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]acrylonitrile [0278] To a mixture of 4-(1 H-pyrazol-4-yl)-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]-pyrimidine (0.40 g, 1.2 mmol) and 3-cyclopentylprop-2-ynenitrile (0.30 g, 2.5 mmol) in DMF (8 mL) was added potassium carbonate (0.09 g, 0.6 mmol). The mixture was stirred for 35 min. The reaction was diluted with ethyl acetate and brine, and the aqueous portion extracted with three volumes of ethyl acetate. The combined organic extracts were washed with brine again, then were dried over sodium sulfate, decanted and concentrated in vacuo. The crude residue was purified by flash column chromatography (ethyl acetate/hexanes) to yield the desired product (290 mg, 53%). 1H NMR (400 MHz, CDCI3): δ. 8.98 (s, 1H), 8.87 (s, 1H), 8.46 (s, 1H), 7.42 (d, 1H), 6.84 (d, 1H), 5.67 (s, 2H), 5.21 (s, 1H), 3.64-3.55 (m, 1H), 3.53 (t, 2H), 2.13-2.01 (m, 2H), 1.83-1.66 (m, 4H), 1.57-1.46 (m, 2H), 0.91 (t, 2H), -0.07 (s, 9H); MS(ES):435(M+1).
Step 3. (2Z)-3-Cyclopenlyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1 -yl]acrylonitrile [0279] A solution of (2Z)-3-cyclopentyl-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]-pyrimidin-4-yl)-1H-pyrazol-1-yl]acrylonitrile (0.030 g, 0.069 mol) in DCM (3 mL) and TFA (2 mL) was stirred for 1 hour. The solvents were removed in vacuo and the product was stirred with THF (1.5 mL), sodium hydroxide, 50% aqueous solution (0.75 mL) and water (0.75 mL)for2 hours. The reaction mixture was neutralized by the dropwise addition of cone. HCI. The product was extracted with ethyl acetate. The combined organics were dried over sodium sulfate, filtered and concentrated in vacuo. The crude residue was purified by preparative-HPLC/MS (C18 column eluting with a gradient of ACN/H20 containing 0.15% NH4OH) to afford the desired product (16 mg, 76%). 1H NMR (400 MHz, d6-dmso): δ 9.08 (s, 1H), 8.74 (s, 1H), 8.63 (s, 1H), 7.66 (d, 1H), 7.05 (d, 1H), 5.82 (d, 1H), 3.62-3.54 (m, 1H), 2.00-1.90 (m, 2H), 1.76-1.48 (m, 6H); MS(ES):305(M+1).
Example 75 : 3-Cyclopentylidene-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]-propanenitrile [0280]
Step 1.3-Cyclopentylidene-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]pro-panenitrile [0281] To a suspension of 3-cyclopentylprop-2-ynenitrile (0.4 g, 0.003 mol) in ACN (10 mL) was added 4-(1H-pyrazol-4-yl)-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidine (0.53 g, 1.7 mmol) and DBU (0.33 mL, 2.2 mmol). This mixture was stirred at room temperature for 50 minutes. The reaction mixture was partitioned between ethyl acetate and dilute HCI. The aqueous portion was separated and extracted with ethyl acetate. The combined organic extracts were washed with dilute HCI and brine, were dried over sodium sulfate, filtered and concentrated in vacuo. The crude residue was purified by flash column chromatography (ethyl acetate/hexanes) to yield the desired product (540 mg, 74%). 1H NMR (300 MHz, CDCI3): δ 8.85 (s, 1H), 8.36 (s, 1H), 8.35 (s, 1H), 7.40 (d, 1H), 6.78 (d, 1H), 5.67 (s, 2H), 3.70 (s, 2H), 3.54 (dd, 2H), 2.55 (t, 2H), 2.45 (t, 2h), 1.85 (dddd, 2H), 1.73 (dddd, 2H), 0.91 (dd, 2H), -0.06 (s, 9H); MS(ES):435(M+1).
Step 2. 3-Cyclopentylidene-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propanenitrile [0282] A solution of 3-cyclopentylidene-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]-pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile (0.030 g, 0.069 mmol) in DCM (3 mL) and TFA (2 mL) was stirred for 1 hour. The solvents were evaporated in vacuo and the product was stirred with sodium hydroxide, 50% aqueous solution (0.75 mL) and water (0.75 mL) and THF (1.5 mL) for 2 hours. The reaction mixture was neutralized by dropwise addition of concentrated HCI. The product was extracted with ethyl acetate. The combined organic extracts were dried over sodium sulfate, filtered and concentrated in vacuo. The crude residue was purified by preparative-HPLC/MS (C18 column eluting with a gradient of ACN/H20 containing 0.15% NH4OH) to afford the desired product (7 mg, 33%). 1H NMR (400 MHz, d6-dmso): δ 12.23-12.01 (brs, 1H), 8.78 (s, 1H), 8.69 (s, 1H), 8.46 (s, 1H), 7.60 (d, 1H), 7.04 (d, 1H), 3.95 (s, 2H), 2.53 (t, 2H), 2.42 (t, 2H), 1.76 (dddd, 2H), 1.65 (dddd, 2H); MS(ES):305(M+1).
Example 76: 3-Methyl[5-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1,3-thiazol-2-yl]aminopropane-nitrile trifluoroacetate salt [0283]
Step 1. 4-(1,3-Thiazol-5-yl)-7-[2-(trimethylsHyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidine [0284] 4-Chloro-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidine (3.00 g, 0.0106 mol), and 1,3-thiazole (7.50 mL, 0.106 mol) were dissolved in Ν,Ν-dimethylacetamide (40.0 mL). The solution was distributed in equal portions into four 20 mL microwavable vessels. Into each reaction vessel was then added potassium acetate (0.777 g, 7.93 mmol) followed by tetrakis(triphenyl-phosphine)palladium(0) (0.60 g, 2.1 mmol). Each reaction vessel was heated at 200 °C in the microwave reactor for 30 minutes. The reactions were combined and most of the solvent was removed in vacuo. The residue was diluted with DCM, filtered and concentrated. Purification by flash column chromatography (ethyl ace-tate/hexanes) afforded the desired product (2.25 g, 64%). 1H NMR (300 MHz, CDCI3): δ 8.99 (s, 1H), 8.90 (s, 1H), 8.72 (s, 1H), 7.49 (d, 1H), 6.91 (d, 1H), 5.70 (s, 2H), 3.56 (dd, 2H), 0.93 (dd, 2H), -0.05 (s, 9H); MS(ES):333(M+1).
Step 2. 4-(2-Bromo-1,3-thiazol-5-yl)-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidine [0285] 2.5 M n-Butyllithium in hexane (0.860 mL) was added dropwise to a -78 °C solution of 4-(1,3-thiazol-5-yl)-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidine (550 mg, 0.0016 mol) in THF (20 mL). The mixture was stirred for 30 minutes at-78 °C, followed by the slow addition of carbon tetrabromide (658 mg, 0.00198 mol) as a solution in THF (10 mL). After 30 minutes, the mixture was quenched with a small amount of saturated ammonium chloride, diluted with ether, and dried over sodium sulfate. The residue obtained after filtration and concentration was purified by flash column chromatography (ethyl acetate/hexanes) to afford the desired product (387 mg, 57%). 1H NMR (300 MHz, CDCI3): δ 8.85 (s, 1H), 8.33 (s, 1H), 7.49 (d, 1H), 6.83 (d, 1H), 5.69 (s, 2H), 3.55 (dd, 2H), 0.92 (dd, 2H), -0.05 (s, 9H); MS(ES):411,413(M+1).
Step 3. 4-(2-Bromo-1,3-thiazol-5-yl)-7H-pyrrolo[2,3-d]pyrimidine [0286] A solution of 4-(2-bromo-1,3-thiazol-5-yl)-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo-[2,3-d]pyrimidine (370 mg, 0.90 mmol) in DCM (5.0 mL) and TFA (1.0 mL) was stirred at room temperature for 7 hours. The mixture was then concentrated, re-dissolved in methanol (2 mL), and ethylenediamine (0.5 mL) was added. The mixture was stirred for 6 hours at room temperature. The mixture was diluted with DCM (10 mL), and the precipitate was isolated by filtration and washed with a small amount of DCM to afford desired product (182 mg, 72%). 1H NMR (300 MHz, d6-dmso): δ. 8.74 (s, 1H), 8.70 (s, 1 H), 7.76 (d, 1 H), 7.15 (d, 1H); MS(ES):281,283(M+1).
Step 4. 3-Methyl[5-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1,3-thiazol-2-yl]aminopropanenitrile [0287] A solution of 4-(2-bromo-1,3-thiazol-5-yl)-7H-pyrrolo[2,3-d]pyrimidine (31 mg, 0.11 mmol) and 3-(methylami-no)propionitrile (103 μί, 0.00110 mol) in DMF (1.0 mL, 0.013 mol) was stirred at 90 °C for 2 hours. The crude reaction mixture was purified by preparative-HPLC/MS (C18 column eluting with a gradient of ACN/H20 containing 0.15% NH4OH and again by preparative-HPLC/MS (C18 column eluting with a gradient of ACN/H20 containing 0.1 % TFA) to yield the desired product as the trifluoroacetate salt (30 mg, 68%). 1H NMR (300 MHz, d6-DMSO):ô 12.25 (s, 1H), 8.60 (s, 1H), 8.31 (s, 1H), 7.60 (dd, 1H), 7.00 (dd, 1H), 3.89 (t, 2H), 3.20 (s, 3H), 2.94 (t, 2H); MS(ES):285(M+1).
Example 77: (3S)- and (3R)-3-[5-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1,3-thiazol-2-yl]hexane-nitrile [0288]
Step 1. N-Methoxy-N-methylbutanamide [0289] To a mixture of butanoic acid (1.01 g, 0.0115 mol) and Ν,Ο-dimethylhydroxylamine hydrochloride (1.12 g, 0.0115 mol) in DCM (50 mL) was added benzotriazol-1-yloxytris(dimethylamino)-phosphonium hexafluorophosphate (5.6 g, 0.013 mol) and TEA (3.2 mL, 0.023 mol). The mixture was stirred overnight at room temperature. The solution was then washed with water and brine, dried over sodium sulfate, and concentrated in vacuo. The crude product was purified by flash column chromatography (ether/hexanes). The solvent was removed (235 mbar/40 °C) to afford the product (1,33g, 88%). 1H NMR (300 MHz, CDCI3): δ 3.68 (s, 3H), 3.18 (s, 3H), 2.40 (t, 2H), 1.74-1.59 (m, 2H), 0.96 (t, 3H).
Step 2. 1-[5-(7-[2-(TrimethylsHyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1,3-thiazol-2-yl]-butan-1-one [0290] 2.5 M n-Butyllithium in hexane (878 μί) was added slowly dropwise to a -78 °C solution of 4-(1,3-thiazol-5-yl)-7-[2-(trimethylsilyl)ethoxyjmethyl-7H-pyrrolo[2,3-d]pyrimidine (501 mg, 1.37 mmol) in THF (20 mL). After 45 minutes, N-methoxy-N-methylbutanamide (0.360 g, 2.74 mmol) was added. The reaction was continued at -78 °C for 30 min, and was then allowed to reach room temperature. The reaction was quenched with saturated ammonium chloride, and was extracted with ethyl acetate. The extracts were washed with water and brine, dried over sodium sulfate and concentrated in vacuo. Flash column chromatography (ethyl acetate/hexanes) afforded the product (235 mg, 42%). 1H NMR (300 MHz, CDCI3): δ 8.93 (s, 1H), 8.76 (s, 1H), 7.52 (d, 1H), 6.92 (d, 1H), 5.71 (s, 2H), 3.56 (dd, 2H), 3.19 (t, 2H), 1.92-1.77 (m, 2H), 1.05 (t, 3H), 0.93 (dd, 2H), -0.05 (s, 9H); MS(ES):403(M+1).
Step 3. (2E)- and (2Z)-3-[5-(7-[2-(Trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1,3-thiazol-2-yl]hex-2-enenitrile [0291] To a solution of 1.0 M potassium tert-butoxide in THF (0.605 mL) in THF (4.0 mL) at 0° C was added diethyl cyanomethylphosphonate (0.102 mL, 0.634 mmol) dropwise. The cooling bath was removed and the reaction was warmed to room temperature. After 30 minutes, a solution of 1-[5-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yI)-1,3-thiazol-2-yl]butan-1-one (232 mg, 0.576 mmol) in THF (3.0 mL) was added dropwise. The reaction was stirred for 2 hours, and the crude mixture was then adsorbed onto silica gel and purified by flash column chromatography (ethyl acetate/hexanes) to afford the product as a mixture of olefin isomers (225 mg, 92%). 1H NMR (300 MHz, CDCI3), major isomer: δ 8.89 (s, 1H), 8.65 (s, 1H), 7.52 (d, 1H), 6.89 (d, 1H), 6.35 (s, 1H), 5.70 (s, 2H), 3.56 (dd, 2H), 2.96 (t, 2H), 1.88-1.72 (m, 2H), 1.08 (t, 3H), 0.93 (dd, 2H), - 0.07 (s, 9H); MS(ES):426(M+1).
Step 4. (3S)- and (3R)-3-[5-(7-[2-(Trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1,3-thiazol-2-yl]hex-anenitrile [0292] Cupric acetate, monohydrate (0.7 mg, 0.004 mmol) and (oxydi-2,1-phenylene)bis(diphenyl-phosphine) (2 mg, 0.004 mol) was mixed in toluene (0.24 mL). PMHS (30 μί) was added. The mixture was stirred for 25 minutes at room temperature followed by the addition of(2E)-3-[5-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1,3-thiazol-2-yl]hex-2-enenitrile (51 mg, 0.12 mol) in toluene (0.24 mL) and finally, tert-butyl alcohol (0.043 mL). The resulting mixture was stirred overnight. The crude mixture was purified directly by flash column chromatography (ethyl acetate/hexanes) to afford the desired product (39 mg, 76%). 1H NMR (300 MHz, CDCI3): δ 8.87 (s, 1H), 8.52 (s, 1H), 7.48 (d, 1H), 6.87 (d, 1H), 5.69 (s, 2H), 3.60-3.46 (m, 3H), 2.99-2.82 (m, 2H), 2.05-1.89 (m, 2H), 1.50-1.34 (m, 2H), 0.97 (t, 3H), 0.92 (t, 2H), -0.06 (s, 9H); MS(ES):428(M+1).
Step 5. (3S)- and (3R)-3-[5-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1,3-thiazol-2-yl]hexanenitrile [0293] TFA (1.0 mL) was added to a solution of 3-[5-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4- yl)-1,3-thiazol-2-yl]hexanenitrile (36 mg, 0.084 mmol) in DCM (4.0 mL) and the mixture was stirred at room temperature for 3 hours. The mixture was concentrated, and re-dissolved in methanol (3 mL), to which ethylenediamine (0.1 mL) was added. After 2 hours reaction time, the mixture was concentrated and directly purified by preparative-HPLC/MS (C18 column eluting with a gradient of ACN/H20 containing 0.15% NH4OH) to afford the desired product (10 mg, 40%). 1H NMR (300 MHz, CDCI3):Ô9.96 (brs, 1H), 8.87 (s, 1H), 8.54 (s, 1H), 7.51-7.45 (m, 1H), 6.90-6.86 (m, 1H), 3.59-3.44 (m, 1H), 3.01-2.82 (m, 2H), 2.06-1.87 (m, 2H), 1.51-1.34 (m, 2H), 0.98 (t, 3H); MS(ES):298(M+1).
Example 78: (3R)- and (3S)-3-Cyclopentyl-3-[5-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1,3-thiazol-2-yl]propanenitrile [0294]
[0295] To a solution of (2E)- and (2Z)-3-cyclopentyl-3-[5-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1,3-thiazol-2-yl]acrylonitrile (199 mg, 0.440 mmol) (prepared, for example, as in Example 77, steps 1 through 3) in a mixture of ethanol (10 mL) and ethyl acetate (10 mL) was added a catalytic amount of 10% palladium on carbon. The mixture was stirred at room temperature under one atmosphere of hydrogen overnight. It was then subjected to 50 PSI H2 until the reaction was complete. Filtration and removal of solvent afforded an oil which was dissolved in DCM (4 mL) and TFA (1 mL). The solution was stirred until starting material was consumed and the mixture was then concentrated and re-dissolved in methanol (3 mL), to which ethylenediamine (0.4 mL) was added. The solution was stirred at room temperature for one hour, and was concentrated in vacuo. The crude mixture was purified by preparative-HPLC/MS (C18 column eluting with a gradient of ACN/H20 containing 0.15% NH4OH) to afford the desired product (36 mg, 25%). 1H NMR (400 MHz, CDCI3): δ 10.44 (brs, 1H), 8.89 (s, 1H), 8.56 (s, 1H), 7.50 (dd, 1H), 6.89 (dd, 1H), 3.34 (dt, 1H), 2.98 (dd, 1H), 2.89 (dd, 1H), 2.44-2.31 (m, 1H), 2.07-1.96 (m, 1H), 1.80-1.52 (m, 5H), 1.40-1.24 (m, 2H); MS(ES):324(M+1).
[0296] The following compounds of Table 5c were prepared (as racemic mixtures) as described by Example 77, 78 or 86, as indicated in the following table, by using different Weinreb amides (as prepared in Example 77, Step 1):
Example 84: (2S)- and (2R)-2-[5-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1,3-thiazol-2-yl]pentane-nitrile [0297]
Step 1. (2S)- and (2R)-2-[5-(7-[2-(Trimethyisilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1,3-thiazol-2-yl]pen- tanenitrile [0298] To a mixture of 1-[5-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1,3-thiazol-2-yl)butan- 1-one (prepared as in Example 77) (101 mg, 0.251 mmol) and p-tolylsulfonylmethyl isocyanide (147 mg, 0.753 mmol) in a mixture of DMSO (5.0 mL) and ethanol (61 μι) was added 1.0 M potassium tert-butoxide in THF (753 μΙ_). The mixture was then heated to 45 °C for 2 hours. Upon cooling to room temperature, the mixture was quenched by the addition of saturated ammonium chloride, followed by water. The product was extracted with ether, and the extracts were washed with water and brine, dried over sodium sulfate, filtered and concentrated in vacuo. Flash column chromatography (ethyl acetate/hexanes) afforded the product (39 mg, 25%). 1H NMR (400 MHz, CDCI3): δ 8.88 (s, 1H),8.52 (s, 1H), 7.50 (d, 11-1),6.87 (d, 1H), 5.70 (s, 2H), 4.32 (dd, 1H), 3.55 (dd, 2H), 2.20-2.11 (m, 2H), 1.71-1.57 (m, 2H), 1.03 (t, 3H), 0.93 (dd, 2H); MS(ES):414(M+1).
Step 2. (2S)- and (2R)-2-[5-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1,3-thiazol-2-yl]pentanenitrile [0299] A solution of 2-[5-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1,3-thiazol-2-yl]pentanen-itrile (59 mg, 0.093 mmol) in DCM (3 mL) and TFA (0.5 mL) was stirred at room temperature for 4 hours. The mixture was then concentrated, and the residue was then dissolved in methanol (3 mL) to which ethylenediamine (0.3 mL) was then added. The solution was stirred at room temperature for 40 minutes. The solvent was removed in vacuo, and the crude mixture was purified by preparative-HPLC/MS (C18 column eluting with a gradient of ACN/H20 containing 0.15% NH4OH) to afford the desired product (20 mg, 76%). 1H NMR (400 MHz, CDCI3): δ 9.66 (br s, 1H), 8.88 (s, 1H), 8.54 (s, 1H), 7.49 (dd, 1H), 6.88 (dd, 1H), 4.33 (dd, 1H), 2.23-2.12 (m, 2H), 1.75-1.60 (m,2H), 1.04 (t, 3H); MS(ES):284(M+1).
Example 85: (4S)- and (4R)-4-(5-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1,3-thiazol-2-yl]heptane-nitrile [0300]
[0301] To a solution of triethyl phosphonoacetate (188 mg, 0.838 mmol) in THF (6.0 mL) at 0 °C was added 1.0 M potassium tert-butoxide in THF (840 μί). The mixture was then allowed to warm to room temperature followed by recooling to 0 °C, at which time 1 -[5-(7-[2-(trimethylsilyl)ethoxy]-methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1,3-thiazol-2-yl]bu-tan-1-one (prepared as in Example 77) (225 mg, 0.559 mmol) in THF (4.0 mL) was added. The mixture was stirred at room temperature for 1.5 hours, at which time it was quenched with water and extracted with ethyl acetate. The combined extracts were washed with water and brine, dried over sodium sulfate and concentrated in vacuo. Flash column chromatography afforded the product as a mixture of olefm isomers (222 mg, 84%). MS(ES):473(M+1).
[0302] Ethyl 3-[5-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1,3-thiazol-2-yl]hex-2-enoate as a mixture of (2E)- and (2Z)- isomers (222 mg, 0.470 mmol) was dissolved in ethanol (10 mL), and a catalytic amount of 10% Pd-C was added. The mixture was stirred under an atmosphere of hydrogen, provided by a balloon, for 16 hours. Filtration and concentration in vacuo afforded the desired product (201 mg, 90%). MS(ES):475(M+1).
[0303] To a solution of ethyl 3-[5-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1,3-thiazol-2-yl]hexanoate (201 mg, 0.423 mmol) in THF (5.0 mL) at -78 °C was added 1.00 M diisobutylaluminum hydride in DCM (1.06 mL). The mixture was allowed to warm to -10 °C slowly over 1.5 hours, followed by the addition of potassium sodium tartrate tetrahydrate, water, and ether. The mixture was stirred for 1 hour, then layers were separated, and the aqueous layer was extracted further with ethyl acetate. The organic extracts were washed with water and brine, dried over sodium sulfate and concentrated in vacuo to afford desired product (176 mg, 96%). MS(ES):433(M+1).
[0304] A solution of 3-[5-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1,3-thiazol-2-yl]hexan-1-ol (88 mg, 0.20 mmol) in TFA (2 mL) was stirred for 30 minutes. The TFA was then evaporated and the residue was stirred in methanol (2 mL) containing ethylenediamine (0.2 mL) and a drop of water for 30 minutes. Purification via preparative-HPLC/MS (C18 eluting with a gradient of ACN/H20 containing 0.15% NAOH) afforded the desired product (36 mg, 58%). MS(ES):303(M+1).
[0305] To a mixture of 3-[5-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1,3-thiazol-2-yl]hexan-1-ol (36 mg, 0.12 mmol) and TEA (19.9 μί, 0.143 mmol) in DCM (5 mL) at 0 °C was added methanesulfonyl chloride (11.0 μί, 0.143 mmol). After stirring for 10 minutes, the solution was concentrated and dissolved in DMSO (1.6 mL) and sodium cyanide (23 mg, 0.48 mmol) was added. The mixture was then heated at 125 °C in the microwave for 30 minutes. The mixture was then purified directly using preparative-HPLC/MS (C18 eluting with a gradient of ACN/H20 containing 0-15% NH4OH) to afford the desired product (10 mg, 27%). 1H NMR (400 MHz, CDCI3): δ 9.37 (br s, 1H), 8.86 (s, 1H), 8.52 (s, 1H), 7.46 (dd, 1H), 6.88 (dd, 1H), 3.34-3.25 (m, 1H), 2.47-2.30 (m, 2H), 2.22-2.12 (m, 2H), 1.95-1.71 (m, 2H), 1.44-1.31 (m, 2H), 0.94 (t, 3H); MS(ES):312(M+1).
Example 86: 3-[5-(7H-Pyrrolo[2,3-d]pyrimidm-4-yl)-1,3-thiazol-2-yl)pentanedinitrile [0306]
Step 1. N-Methoxy-2-[(4-methoxybenzyl)oxy]-N-methylacetamide [0307] To a mixture of [(4-methoxybenzyl)oxy]acetic acid (Bioorganic and Medicinal Chemistry Letters, 2001, pp. 2837-2841) (6.86 g, 0.0350 mol) and Ν,Ο-dimethylhydroxylamine hydrochloride (3.41 g, 0.0350 mol) in DCM (100 mL) was added benzotriazol-1-yloxytris(dimethylamino)-phosphonium hexafluorophosphate (17 g, 0.038 mol) followed by TEA (9.7 mL, 0.070 mol). The resulting mixture was stirred overnight at room temperature. The solution was then washed with water, 0.5 M HCI, saturated NaHC03, and brine, then was dried over sodium sulfate, filtered and concentrated in vacuo. Flash column chromatography (ether/hexanes) afforded the desired product (5.75 g, 69%).
Step 2.2-[(4-Methoxybenzyl)oxyJ-1-[5-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]-pyrimidin-4-yl)-1,3-thiazol-2- yljethanone [0308] To a solution of 4-(1,3-thiazol-5-yl)-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]-pyrimidine (2.12 g, 6.38 mmol) in THF (70 mL) at -78 °C was added 2.5 M n-butyllithium in hexane (3.06 mL) slowly dropwise. After stirring for 30 minutes, N-methoxy-2-[(4-methoxybenzyl)oxy]-N-methylacetamide (2.29 g, 9.56 mmol) was added. The reaction was continued for 30 minutes following the addition, at -78 °C, then the cooling bath was removed and the reaction was quenched with saturated ammonium chloride and extracted with ether. The extracts were dried with sodium sulfate and concentrated in vacuo. The crude mixture was purified by flash column chromatography (ethyl acetate/hexanes) to afford desired product (2.16 g, 66%). 1H NMR (300 MHz, CDCI3): δ 8.93 (s, 1H), 8.72 (s, 1H), 7.53 (d, 1H), 7.37 (d, 2H), 6.91 (d, 2H), 6.89 (d, 1H), 5.70 (s, 2H), 5.00 (s, 2H), 4.70 (s, 2H), 3.81 (s, 3H), 3.56 (dd, 2h), 0.93 (dd, 2H), -0.05 (s, 9H); MS(ES):511(M+1).
Step 3. (2E)- and (2Z)-4-[(4-Methoxybenzyl)oxy]-3-[5-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1,3-thiazol-2-yl]but-2-enenitrile [0309] To a solution of 1 M potassium tert-butoxide in THF (4.44 mL) in THF (30 mL) at 0° C was added diethyl cyanomethylphosphonate (820 mg, 0.0046 mol) dropwise. The bath was removed and the reaction was warmed to room temperature. After 30 minutes, a solution of 2-[(4-methoxybenzyl)-oxy]-1-[5-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyr-rolo[2,3-d]pyrimidin-4-yl)-1,3-thiazol-2-yl]-ethanone (2.16 g, 0.00423 mol) in THF (20 mL) was added dropwise. The reaction was stirred for 1 hour, and was then quenched with a small amount of saturated ammonium chloride, diluted with ether, dried over sodium sulfate and concentrated in vacuo. Purification by flash column chromatography, eluting with a gradient of 0-35% ethyl acetate/hexanes afforded the desired product as a mixture of olefin isomers in nearly equal amounts (1.76 g, 78%). 1H NMR (400 MHz, CDCI3): δ 8.90 (s, 1H), 8.88 (s, 1H), 8.71 (s, 1H), 8.67 (s, 1H), 7.50 (d, 2H), 7.35 (dd,2H), 7.31 (dd, 2H), 6.92 (dd, 2H), 6.90 (dd, 2H), 6.86 (d, 2H), 6.62 (s, 1H), 6.10 (t, 1H), 5.70 (s, 4H), 4.75 (s, 2H), 4.72 (d, 2H), 4.64 (s, 4H), 3.82 (s, 3H), 3.81 (s, 3H), 3.56 (dd, 2H), 3.55 (dd, 2H), 0.96-0.90 (m, 4H), -0.05 (s, 9H), -0.054 (s, 9H); MS(ES):534(M+1).
Step 4. 4-[(4-Methoxybenzyl)oxy]-3-[5-(7-[2-(trimethylsHyl)ethoxy]methyl-7H-pyrrolo[2,3-d]-pyrimidin-4-yl)-1,3-thiazol- 2-yl]butanenitrile [0310] (2E)- and (2Z)-4-[(4-Methoxybenzyl)oxy]-3-[5-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1,3-thiazol-2-yl]but-2-enenitrile (880 mg, 1.6 mmol) was dissolved in a mixture of ethanol (20 mL) and ethyl acetate (20 mL). A catalytic amount of 10% Pd-C was added. The mixture was shaken under 50 PSI of hydrogen. The mixture was filtered and concentrated in vacuo to afford the desired product (0.85 g, 99%). MS(ES):536(M+1).
Step 5. 3-[5-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1,3-thiazol-2-yl]pentanedinitrile [0311] 4-[(4-Methoxybenzyl)oxy]-3-[5-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]-pyrimidin-4-yl)-1,3-thiazol- 2- yl]butanenitrile (251 mg, 0.468 mmol) in DCM (10 mL) was treated with dichlorodicyanoquinone (DDQ) (434 mg, 1.87 mmol), followed by water (376 μί). After 1.5 hours, saturated sodium bicarbonate and water were added, and the reaction was extracted with ethyl acetate three times. The extracts were washed with water, brine, dried over sodium sulfate, filtered and concentrated in vacuo to afford the crude product which was used without further purification.
[0312] Asolution of the above prepared 4-hydroxy-3-[5-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1,3-thiazol-2-yl]butanenitrile in DCM (12 mL) at 0 °C was treated sequentially with TEA (130 μί, 0.94 mmol) and methanesulfonyl chloride (73 μί, 0.94 mmol). After 1 hour reaction time, the mixture was diluted with water and extracted with ethyl acetate three times. The extracts were washed with water and brine, dried over sodium sulfate, filtered and concentrated in vacuo. The residue was then dissolved in DMSO (5 mL) and sodium cyanide (110 mg, 2.3 mmol) was added. After 30 minutes, the mixture was diluted with water, extracted with ether, washed with water, brine and dried over sodium sulfate. Concentration and purification by flash column chromatography (ethyl acetate/hexanes) afforded the desired product (14 mg, 7%). MS(ES):425(M+1).
[0313] Asolution of 3-[5-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1,3-thiazol-2-yl]pentaned-initrile (14 mg, 0.033 mmol) in DCM (3 mL) containing TFA (0.6 mL) was stirred for 4 hours. The mixture was then concentrated and the residue was redissolved in methanol (2 mL) to which ethylenediamine (0.4 mL) was then added. After 1 hour reaction time, the product was purified by preparative-HPLC/MS (C18 eluting with a gradient of ACN/H20 containing 0.15% NH4OH to afford the desired product (6 mg, 62%). 1H NMR (400 MHz, d6-dmso): δ 12.27 (brs, 1H), 8.84 (s, 1H), 8.76 (s, 1H), 7.75 (d, 1H), 7.14 (d, 1H), 4.14 (m, 1H), 3.17 (d, 4H); MS(ES):295(M+1).
Example 87: (3R)-3-Cyclopentyl-3-[5-(7H-pyrrolo[2,3-d]pyrimidln-4-yl)-1,3-oxazol-2-yl]-propanenitrile, and (3S)- 3- Cyclopentyl-3-[5-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1,3-oxazol-2-yl]propanenitrile [0314]
Step 1. 4-( 1,3-Oxazol-5-yl)-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidine [0315] A mixture of 4-chloro-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidine (0.440 g, 1.55 mmol), 1,3-oxazole (0.306 mL, 4.65 mmol), potassium acetate (0.456 g, 4.65 mmol) and tetrakis(triphenylphosphine)palladium(0) (0.179 g, 0.155 mmol) in Ν,Ν-dimethylacetamide (8.0 mL) was heated to 200 °C in the microwave reactor for 30 minutes. Most of the solvent was removed in vacuo. The resulting residue was diluted with DCM, and was filtered and concentrated. Flash column chromatography (ethyl acetate/hexanes) afforded the product (330 mg, 67%). 1H NMR (400 MHz, CDCI3): δ 8.96 (s, 1H), 8.21 (s, 1H), 8.08 (s, 1H), 7.54 (d, 1H), 7.08 (d, 1H), 5.76 (s, 2H), 3.60 (t, 2H), 0.98 (t, 2H), 0.00 (s, 9H); MS(ES):317(M+1).
Step 2. Cyclopentyl[5-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1,3-oxazol-2-yl]methanone [0316] n-Butyllithium in hexane (1.6 M, 0.30 mL) was added slowly dropwise to a -78 °C solution of 4-(1,3-oxazol-5-yl)-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidine (140.0 mg, 0.44 mmol) in THF (10.0 mL). After 20 minutes, 1.0 M zinc dichloride in ether (0.53 mL) was added. The reaction mixture was then stirred for 60 min at 0 °C. Following this, copper(l) iodide (84 mg, 0.44 mmol) was added, and this mixture was allowed to stir for 10 minutes. Cyclopentanecarbonyl chloride (108 pL, 0.885 mmol) was then added. The reaction was stirred at 0 °C for a further 1 hour, at which time it was allowed to warm to room temperature. The reaction was quenched by the addition of saturated NH4CI solution, and was extracted with ethyl acetate. The extracts were washed with water and brine, dried over sodium sulfate, filtered and concentrated in vacuo. Flash column chromatography (ethyl acetate/hexanes) afforded the product (97 mg, 53%). 1H NMR (400 MHz, CDCI3): δ 8.96 (s, 1H), 8.21 (s, 1H), 7.56 (d, 1H), 7.22 (d, 1H), 5.76 (s, 2H), 3.60 (t, 2H), 3.56 (t, 1H), 2.23-1.56 (m, 8H), 0.98 (t, 2H), 0.00 (s, 9H); MS(ES):413(M+1).
Step 3. (3R)- and (3S)-3-Cyclopentyl-3-[5-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1,3-oxa-zol-2-yl]propanenitrile [0317] To a solution of 1.0 M potassium tert-butoxide in THF (0.355 mL) and THF (3 mL) at 0° C was added diethyl cyanomethylphosphonate (66 mg, 0.37 mmol) dropwise. The cold bath was removed and the reaction was warmed to room temperature. After 30 minutes, a solution of cyclopentyl[5-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]py-rimidin-4-yl)-1,3-oxazol-2-yl]methanone (1.40E2 mg, 0.338 mmol) in THF (2.0 mL) was added dropwise. After 3 hours reaction time, the mixture was adsorbed onto silica gel, and flash column chromatography (ethyl acetate/hexanes) afforded the desired product as a mixture of olefin isomers (89 mg, 60%). MS(ES):436(M+1).
[0318] To a mixture of cupric acetate, monohydrate (4.0 mg, 0.020 mmol) and (oxydi-2,1-phenylene)bis(diphenylphos-phine) (11 mg, 0.020 mmol) in toluene (0.40 mL, 0.0038 mol) was added PMHS (50 μί). The resulting mixture was stirred for 25 minutes at room temperature, followed by the addition of (2E)- and (2Z)-3-cyclopentyl-3-[5-(7-[2-(trimeth-ylsilyl)ethoxy]methyl-7H-pyrrolo-[2,3-d]pyrimidin-4-yl)-1,3-oxazol-2-yl]acrylonitrile (88 mg, 0.20 mmol) in toluene (0.40 mL), and then tert-butyl alcohol (0.072 mL). After failure to react at room temperature over 16 hours, additional cupric acetate, monohydrate and (oxydi-2,1-phenylene)bis(diphenylphosphine) (0.10 mol equivalent each) were added and the reaction mixture was heated at 60 °C for 16 hours. The crude mixture was subjected to flash column chromatography (ethyl acetate/hexanes) to afford the desired product (21 mg, 23%). 1H NMR (400 MHz, CDCI3): δ 8.96 (s, 1H), 8.02 (s, 1H), 7.56 (d, 1H), 7.10 (d, 1H), 5.76 (s, 2H), 3.60 (t, 2H), 3.38-3.30 (m, 1H), 3.03 (dd, 1H), 2.95 (dd, 1H), 2.60-2.40 (m, 1H), 2.10-2.00 (m, 1H), 1.85-1.15 (m, 7H), 0.98 (t, 2H), 0.00 (s,9H); MS(ES):438(M+1).
Step 4. (3R)- and (3S)-3-Cyclopentyl-3-[5-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1,3-oxazol-2-yl]-propanenitrile [0319] Asolutionof3-cyclopentyl-3-[5-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]-pyrimidin-4-yl)-1,3-oxazol-2-yl]propanenitrile (20.0 mg, 0.0457 mmol) was stirred with TFA (0.1 mL) in DCM (0.2 mL) for 6 hours. The solvent was removed, and the resulting residue was stirred overnight with ethylenediamine (0.1 mL) in methanol (0.2 mL). The solvent was removed in vacuo. The desired product was obtained via preparative-HPLC/MS (C18 column eluting with a gradient of ACN/H20 containing 0.15% NH4OH) (5.3 mg, 38%). 1H NMR (400 MHz, CDCI3): δ 10.25 (brs, 1H), 8.90 (s, 1H), 8.00 (s, 1H), 7.50 (d, 1H), 7.06 (d, 1H), 3.36-3.28 (m, 1H), 2.98 (dd, 1H), 2.90 (dd, 1H), 2.51-2.38 (m, 1H), 2.08-1.96 (m, 1H), 1.80-1.51 (m, 5H), 1.44-1.30 (m, 2H); MS(ES):308(M+1).
[0320] The following compound of Table 5d was also prepared as a racemic mixture, according to the procedure of the above Example 87.
Example 90: 5-(Methylthio)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]pentane-nitrile [0321]
Step 1. (2E)-5-(Methylthio)pent-2-enenitrile [0322] To a 0 °C mixture of [chloro(triphenyl)phosphoranyl]ACN (2.5 g, 0.0073 mol) in THF (10 mL, 0.1 mol) was added TEA (2.0 mL, 0.014 mol), and the resulting mixture was stirred for 30 min. The ice bath was removed for 30 min, then the mixture was re-cooled back to 0 °C, A solution of 3-(methylthio)-propanol (0.68 mL, 0.0072 mol) in THF (1 mL, 0.02 mol) was added and the mixture was stirred overnight. Water was added and the mixture was filtered. The filtrate was washed with water x3 and brine. The organic phase was dried and the solvent was removed by rotary evaporation to give 2.1 g of an off-white solid. The solid was triturated with MTBE and was filtered. The filtrate was washed with 1N HCI, water, sat. NaHCOs and brine. The organic phase was dried and was concentrated using a rotary evaporator to give 0.62 g orange oil (44% yield, trans : cis ~ 2 : 1). 1H NMRfortrans (400 MHz, CDCI3): δ 6.68 (1H, m); 5.14 (1H, d); 2.6 (2H, m); 2.55 (2H, t); 2.1 (3H,s).
Step 2. 5-(Methylthio)-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazot-1 -ytjpen-tanenitrile [0323] A mixture of 4-(1H-pyrazol-4-yl)-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]-pyrimidine (0.30 g, 0.00095 mol), (2E)-5-(methylthio)pent-2-enenitrile (0.28 g, 0.0016 mol) and DBU (45 μί, 0.00030 mol) in ACN (3 mL, 0.06 mol) was stirred at rt for 5 days. The solvent was removed by rotary evaporation to give an orange oil. The crude oil was chromatographed with 30-70 ethyl acetate/hex, to give 0.35 g of a colorless oil (83% yield). 1H NMR (400 MHz, CDCI3): δ 8.95 (1H, s); 8.41 (1H, s); 8.4 (1H, s); 7.48 (1H, d); 6.84 (1H, d); 5.75 (2H, s); 4.95 (1H, br); 3.6 (2H, t); 3.1 (2H, m); 2.58 (2H, m); 2.28 (2H, m); 2.1 (3H, s); 1.99 (2H, t); 0.0 (9H, s). MS (M+H): 443.
Step 3. 5-(Methythio)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]pentanenitrile [0324] A solution of 5-(methylthio)-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]-pyrimidin-4-yl)-1H-pyra-zol-1-yl]pentanenitrile (0.35 g, 0.00079 mol) in THF (4 mL, 0.05 mol) and 3.0 M HCI (HCI) in water (4 mL) was heated to reflux overnight. The solvent was removed by rotary evaporation to give a pale orange oil. The oil was stirred in ethanol (3 mL, 0.05 mol) and 8.0 M ammonium hydroxide in water (1 mL) overnight. The reaction was concentrated and purified by prep LCMS (C18 column eluting with a gradient of ACN/H20 containing 0.15% NH4OH) to give 125 mg of a white foam. The white foam was triturated with MTBE (~1.5 mL). The resulting solid was filtered, washed and dried to give 80 mg of the product (32% yield). 1H NMR (400 MHz, CDCI3): δ 10.38 (1H, s); 8.88 (1H, s); 8.39 (1H, s); 8.38 (1H, s); 7.44 (1H, d); 6.8 (1H, d); 5.75 (2H, s); 4.9 (1H, br); 3.05 (2H, m); 2.5 (2H, m); 2.23 (2H, d); 2.1 (3H, s). MS (M+H): 313.
Example 91: 5-(Methylsulfinyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazo1-yl]-pentanenitrile [0325]
[0326] A solution of 5-(methylthio)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-pentanenitrile (0.065 g, 0.00021 mol) and hydrogen peroxide (0.022 mL, 0.00023 mol) in ACN (1 mL, 0.02 mol) was stirred overnight. The reaction was concentrated and purified by HPLC to give 21 mg of a solid. The solid was triturated with MTBE (1 mL)/DCM (10 drops). The solid was filtered and washed to give 13 mg of a white solid (20% yield) which was dried rt to 50 °Cfor2 h. 1H NMR (400 MHz, CDCI3): δ 9.95 (1H, s); 8.85 (1H, s); 8.4 (2H, m); 7.4 (1H,d); 6.8 (1H, s); 4.9 (1H, br); 3.15 (2H, m); 3.0 (2H, m); 2.8-2.5 (2H, m); 2.6 (3H, s). MS (M+H): 329.
Example 92: 5-(Methylsulfonyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-pentanenitrile [0327]
[0328] A solution of 5-(methylthio)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-l-yl]-pentanenitrile (0.040 g, 0.00013 mol) and hydrogen peroxide (0.5 mL, 0.005 mol) in ACN (1 mL, 0.02 mol) was refluxed overnight. The reaction was purified by HPLC to give 16 mg of a white glass/solid which was triturated with EtOH (~0.8 mL) to give 13 mg of a white solid (30% yield). 1H NMR (400 MHz, CDCI3): δ 8.75 (1H, s); 8.48 (1H, d); 8.4 (1H, d); 7.43 (1H, d); 6.8 (1H, s); 5.0 (1H, br); 3.4 (2H, m); 3.2-3.0 (2H, m); 2.8-2.5 (2H, m); 2.95 (3H, s). MS (M+H): 345.
Example 93: 4,4,4-Trifluoro-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-pyrazol-1-yl]-butyronitrile [0329]
Step 1.4,4,4-Trifluoro-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-y]butaneni-trite [0330] A mixture of 4-(1H-pyrazol-4-yl)-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]-pyrimidine (6.9 g, 0.022 mol), (2E)-4,4,4-trifluorobut-2-enenitrile (2.8 g, 0.023 mol) and DBU (0.18 mL, 0.0012 mol) in ACN (70 mL, 1 mol) was stirred for 20 min. The reaction was filtered and filtrate was removed by rotary evaporation to give an orange oil. The crude oil was chromatographed with 20-50% ethyl acetate/hex to give to give 9.1 g of a solid/oil (96% yield). A single enantiomer (peak 2) was separated by chiral column chromatography (OD-H column, 30%EtOH/hex) as a greenish solid/glass (3.3 g, 32% yield). 1H NMR (400 MHz, CDCI3): δ 8.93 (1H, s); 8.46 (1H, s); 8.45 (1H, s); 7.5 (1H, d); 6.85 (1H, d); 5.75 (2H, s); 5.2 (1H, m); 3.6 (2H, t); 3.7-3.3 (2H, m); 1.99 (2H, t); 0.0 (9H, s). MS (M+H): 438.
Step 2. 4,4,4-Trifluoro-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)pyrazol-1-yl]-butyronitrile [0331] A solution of 4,4,4-trifluoro-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]-pyrimidin-4-yl)-1H-pyra-zol-1 -yl]butanenitrile (3.1 g, 0.0071 mol) from Step 1 in THF (35 mL, 0.43 mol) and 3.0 M HCI in water (35 mL) was heated to reflux overnight. The solvent was removed by rotary evaporation to give a greenish orange oil/glass. The oil was stirred with ethyl acetate and sat. NaHC03 (50 mL). The aqueous phase was extracted with ethyl acetate. The organic layers were washed with brine and reduced by rotary evaporation to give an oil/glass residue. The residue was stirred in ethanol (20 mL, 0.3 mol) and 8.0 M ammonium hydroxide in water (10 mL) over a weekend. The solvent was removed by rotary evaporation to give a pale orange foam/solid. The crude was chromatographed with 0-7% MeOH/DCM, 0.0.7% NH40H to give 3 g of a pale orange paste/solid. The solid was recrystallized from EtOH to give 1.6 g of an off-white crystals (74% yield). 1H NMR (400 MHz, DMSO): δ 12.2 (1H, s); 8.95 (1H, s); 8.7 (1H, s); 8.5 (1H, s); 7.63 (1H, d); 6.96 (1H, d); 6.01 (1H, m); 3.7 (2H, m). MS (M+H): 306.
[0332] The following compounds of Table 5e were prepared as indicated in the column labeled "Prep. Ex. No."
Example 97:3-(2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl)-cyclopentane-carbonitrile trifluoroacetate [0333]
Step 1: 3-(Dimethoxymethyl)cyclopentanecarbaldehyde.
[0334] Into a 3-neck round bottom flask 2-norbornene (5.500 g, 0.05841 mol) was dissolved in DCM (198.0 mL,) and methanol (38.5 mL) and was cooled at -78 °C. Ozone was passed through the reaction until it turned blue and was stirred at -78 °C for 30 minutes. Then nitrogen was passed through for 20 minutes and p-toluenesulfonic acid (0.95 g, 0.0055 mol) was added The reaction was allowed to warm at 20 °C and was stirred for 90 minutes. Into the reaction was added sodium bicarbonate (1.67 g, 0.0199 mol) and the resulting mixture was stirred at 20 °C for 30 minutes and dimethyl sulfide (9.4 mL, 0.13 mol) was added. The reaction was stirred for 16 hours and was reduced by rotary evaporation to ~50 mL The reaction was extracted with DCM and the organic extracts were washed with water and brine, dried (MgS04), and stripped in vacuo. The reaction was distilled at 135 °C (bath temperature) at high pump vacuum to give the product (7.5 g) as a ~2:1 mixture of diastereomers. 1H NMR (300 MHz, CDCI3): 9.64 & 9.62 (d, 1H), 4.15 & 4.12 (s, 1H), 3.35 & 3.34 (s, 6H), 2.77 m, 1H), 2.34 (m, 1H), 1.35-2.00 (m, 6H).
Step 2. (2E,Z)-3-[3-(Dimethoxymethyl)cyclopentyl]acrylonitrile.
[0335] Into a flask containing a 0 °C solution of t-BuOK in THF (1.0 M, 6,10 mL) was added a solution of diethyl cyanomethylphosphonate (1.1 g, 6.4 mmol) in THF (8 mL). The cooling bath was removed and the reaction was warmed to ambient temperature, then a solution of 3-(dimethoxymethyl)cyclopentanecarbaldehyde (1.00 g, 5.81 mmol) in THF (2 mL) was added dropwise. Shortly after completion of the addition orange gel-like particulates began to form, after approximately 1 hour the reaction was gelatinous. The reaction was held with stirring at ambient temperature for 16 hours at which time tic indicated complete reaction. The reaction was partitioned between water and EtOAc and the aqueous phase was washed with additional EtOAc. The combined organic phase was washed with water, then sat’d NaCI, and then was dried over MgS04 and reduced In vacuo, and the resulting residue was purified by column chromatography with 6:1 hexanes:EtOAc + 1% TEA to obtain the product as a 1:1 mixture of E/Z isomers (760 mg, 61%). 1H NMR (400 MHz, CDCI3): δ vinylic protons at 6.69 (m, 0.5H), 6.37 (m, 0.5H), 5.32 (m, 0.5H), 5.23 (m, 0.5H), acetal methine proton at 4.14 (m, 1H), methyl protons at 3.34 (s, 6H).
Step 3. 3-[3-(Dimethoxymethyl)cyclopentyl]-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1 -yl]propanenltrlle.
[0336] To a solution of 4-(1 H-pyrazol-4-yl)-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidine (230 mg, 0.74 mmol) in ACN (5 mL) was added (2E,Z)-3-[3-(dimethoxymethyl)cyclopentyl]acrylonitrile (289 mg, 1.48 mmol), followed by DBU (300 μί, 2.0 mmol). The mixture was stirred at ambient temperature for 16 hours, at which point LCMS and TLC indicated complete reaction. The reaction was reduced to dryness In vacuo, and the residue was purified by column chromatography to obtain the product as a mixture of diastereomers (293 mg, 77%). 1H NMR (400 MHz, CDCI3): δ 8.85 (s, 1H), 8.31 (s, 2H), 7.40 (d, 1H), 6.80 (d, 1H), 5.68 (s, 2H), 4.28 (m, 1H), 4.11 (m, 1H), 3.54 (t, 2H), 3.36 (s, 1,5H), 3.34 (s, 1,5H), 3.30 (s, 1,5H), 3.26 (s, 1,5H), 3.12 (m, 1H),2.94(m, 1H),2.65(m, 1H),2.34 (m, 1H), 2.0-1.0 (m, 6H), 0.92 (t, 2H), -0.56 (s, 9H). MS (El) mlz = 511.3 (M+H).
Step 4. 3-(3-Formylcyclopentyl)-3-[4-(7-(2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]-pyrimidin-4-yl)-1 H-pyrazol-1-yljpropanenitrile.
[0337] To a solution of 3-[3-(dimethoxymethyl)cyclopentyl]-3-[4-(7-[2-(trimethylsilyl)ethoxy]-methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yI)-1 H-pyrazol-1 -yl]propanenitrile (293 mg, 0.574 mmol) in THF (4.5 mL) was added aqueous HCI (1.0 M, 1.5 mL). The reaction was held at ambient temperature for 2.5 hours at which point TLC and LCMS indicated complete deprotection to the corresponding aldehyde. The reaction was partitioned between water and EtOAc and the aqueous phase was extracted with additional EtOAc. The combined organic phase was washed with water, then sat’d NaHC03, then sat’d NaCI, and then was dried over MgS04 and filtered and stripped to dryness to leave the crude product as a mixture of diastereomers. 1H NMR (400 MHz, CDCI3): δ 9.69 (d, 0.5H), 9.64 (d, 0.5H), 8.85 (s, 0.5H), 8.84 (s, 0.5H), 8.35 (s, 0.5H), 8.34 (s, 0.5H), 8.32 (s, 0.5H), 8.30 (s, 0.5H), 7.41 (d, 0.5H), 7.40 (d, 0.5H), 6.80 (d, 0.5H), 6.79 (d, 0.5H), 5.68 (s, 1H), 5.67 (s, 1H), 4.32 (m, 1H), 3.54 (m, 2H), 3.14 (m, 1H), 2.96 (m, 2H), 2.76 (m, 1H), 2.1-1.1 (m, 6H), 0.92 (m, 2H), -0.058 (s, 9H). MS (El) mlz= 465.1 (M+H).
Step 5. 3-3-[(E,Z)-(Hydroxyimino)methyl]cyclopentyl-3-[4-(7-[2-(trimethylsHyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yljpropanenitrile.
[0338] To a solution of 3-(3-formylcyclopentyl)-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile (336 mg, 0.000723 mol) in CH3OH (5.0 mL, 0.12 mol) was added hydroxylamine hydrochloride (60 mg, 0.00087 mol) and KHCO3(110 mg, 0.0011 mol) and the reaction was held at ambient temperature for 16 hours, at which point LCMS indicated complete reaction. The reaction was reduced to dryness in vacuo and the residue was partitioned between water and EtOAc, and the aqueous phase was extracted with additional EtOAc. The combined organic phase was washed with water, then sat’d NaCI, then was dried over MgS04 and concentrated to leave the crude product, which was carried forward to the subsequent reaction without purification. NMR indicated disappearance of aldehydic protons. MS (El) mlz = 480.2 (M+H).
Step 6. 3-(2-Cyano-1-[4-(7-[2-(trimethylsHyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yljethyljcy-clopentanecarbonitrile.
[0339] To a solution of 3-3-[(E,Z)-(hydroxyimino)methyl]cyclopentyl-3-[4-(7-[2-(trimethylsilyl)-ethoxy]-methyl-7H-pyr-rolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile (324 mg, 0.67 mmol) in pyridine (1.2 mL), was added meth-anesulfonyl chloride (210 μί., 2.7 mmol) dropwise. The reaction was heated to 60 °C for 2.5 hours, at which point LCMS indicated complete reaction. The reaction was partitioned between water and EtOAc, and the aqueous phase was extracted with additional EtOAc. The combined organic phase was washed with water, then 0.1 N HCI, then sat’d NaCI, and then was dried over MgS04. The crude product was purified by column chromatography to obtain the product as a mixture of diastereomers (164 mg, 52%). The diastereomers were then separated by chiral HPLCto provide four distinct diastereomers, which were taken directly on to the deprotection step. MS (El) mlz = 462.1 (M+H).
Step 7. 3-(2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl)-cyclopentane-carbonitrile trifluoroace-tate.
[0340] The four diastereomers were then separately deprotected in this representative manner. To 3-2-cyano-1-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethylcyclopentanecarbonitrile (35 mg, 0.076 mmol) dissolved in CH2CI2 (2.0 mL) was added TFA (1.0 mL) and the reaction was stirred for 2 hours at ambient temperature at which point LCMS indicated complete cleavage to the N-hydroxymethyl intermediate. The solvent was removed and to the residue was added methanol (1.0 mL) followed by ethylenediamine (40 μί, 0.61 mmol), the reaction was stirred for 16 hours at which point LCMS indicated complete reaction. The solvent was removed and the residue was purified by preparative LCMS to provide the product as a TFA salt. NOE experiments confirm that all isomers have cis geometry on cyclopentyl ring. Isomers 1 and 2: 1H NMR (400 MHz, CD3OD): δ 8.95 (s, 1H), 8.89 (s, 1H), 8.54 (s, 1H), 7.86 (d, 1H), 7.29 (d, 1H), 4.72 (m, 1H), 3.27 (m, 1H), 3.19 (m, 1H), 2.95 (m, 1H), 2.72 (m, 1H), 2.2-1.9 (m, 4H), 1.67 (m, 2H). Isomers 3 and 4:1H NMR (400 MHz, CD3OD): δ 8.95 (s, 1H), 8.88 (s, 1H), 8.52 (s, 1H), 7.85 (d, 1H), 7.28 (d, 1H), 4.72 (m, 1H), 3.27 (m, 1H), 3.19 (m, 1H), 3.05 (m, 1H), 2.71 (m, 1H), 2.44 (m, 1H), 2.05 (m, 1H), 1.92 (m, 1H), 1.72 (m, 1H), 1.58 (m, 2H).MS (El) mlz = 332.2 (M+H).
Example 98: 3-[3-(Hydroxymethyl)cyclopentyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]pro-panenitrile [0341]
Step 1: 3-[3-(Hydroxymethyl)cyclopentyl]-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1 -yl]propanenitrile [0342] A solution of 3-(3-formylcyclopentyl)-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo-[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile (50.0 mg, 0.108 mmol) in methanol (280 μί) was cooled to 0 °C, then sodium tetrahyd-roborate (14 mg, 0.37 mmol) was added. The reaction was held at 0 °C for 10 minutes, at which point LCMS and TLC indicated complete reaction. The reaction was quenched by cautious addition of 1N HCI (3 drops) and methanol (1 mL), followed by addition of aqueous NaHC03 and CHCI3. The phases were separated and the aqueous phase was washed with additional CHCI3. The combined organic phase was washed with sat’d NaCI, dried over MgS04 and reduced to dryness. The residue was purified by column chromatography to obtain the product as a mixture of diastereomers (37.4 mg, 74%). 1H NMR (400 MHz, CDCI3): δ 8.84 (s, 1H), 8.31 (s, 2H), 7.40 (d, 1H), 6.80 (d, 1H), 5.67 (s, 2H), 4.29 (m, 1H), 3.53 (m, 1H), 3.53 (t, 2H), 3.14 (m, 1H), 2.95 (m, 1H), 2.68 (m, 1H), 2.2-1.0 (m, 9H), 0.92 (t, 2H), -0.059 (s, 9H). MS (El) mlz = 467.2 (M+H).
Step 2. 3-[3-(Hydroxymethyl)cyclopentyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1 -yl]propanenitrile [0343] To 3-[3-(hydroxymethyl)cyclopentyl]-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo-[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile (60.4 mg, 0.129 mmol) dissolved in CH2CI2 (2.0 mL) was added TFA (1.0 mL) and the reaction was stirred for 1 hour at which point LCMS indicated complete cleavage to the N-hydroxymethyl intermediate (m/z = 367). The trifluoroacetate ester of the hydroxymethyl of the cyclopentyl ring was also observed (m/z = 463). The solvent was removed and to the residue was added methanol (1.0 mL) followed by ethylenediamine (80 μΙ_, 1.19 mmol). The resulting mixture was stirred for 16 hours at which point LCMS indicated complete reaction to the desired product. The solvent was removed and the residue was purified by chiral HPLC to provide four distinct diastereomers (20.2 mg total of four isomers, 46%). NOE experiments suggest that all isomers have c/'s geometry on the cyclopentyl ring. Isomers 1 and 2: 1H NMR (400 MHz, CD3OD): δ 8.65 (s, 1H), 8.62 (s, 1H), 8.38 (s, 1H), 7.50 (d, 1H), 6.95 (d, 1H), 4.51 (m, 1H), 3.40 (m, 2H), 3.22 (m, 1H), 3.11 (m, 1H), 2.61 (m, 1H), 2.10 (m, 1H), 1.94 (m, 1H), 1.82 (m, 1H), 1.6-1.4 (m, 3H), 1.03 (m, 1H). Isomers 3 and 4: 1H NMR (400 MHz, CD3OD): δ 8.66 (s, 1H), 8.62 (s, 1H), 8.37 (s, 1H), 7.50 (d, 1H), 6.95 (d, 1H), 4.51 (m, 1H), 3.46 (m, 2H), 3.21 (m, 1H), 3.11 (m, 1H), 2.61 (m, 1H), 2.22 (m, 1H), 2.09 (m, 1H), 1.71 (m, 1H), 1.55-1.25 (m, 3H), 1.04 (m, 1H). MS (El) mlz = 337.1 (M+H).
Example 100: 1-(1H-Pyrrolo[2,3-b]pyridin-4-yl)-1H-indazole (100a) and 2-(1H-pyrrolo[2,3-b]-pyridin-4-yl)-2H-in-dazole (100b) [0344]
[0345] 4-Bromo-1 H-pyrrolo[2,3-b]pyridine (0.078 g, 0.00040 mol) and 1 H-indazole (0.283 g, 0.00240 mol) was heated neat in a sealed tube at 200 °C (an oil bath) overnight with stirring. The reaction was allowed to cool to rt and the crude product was purified by prep LC-MS on a C-18 column eluting with a water/ACN gradient containing 0.2% TFA to give the title compound (0.015 gm, 15%), as an amorphous white solid, LC /MS (M+H)+ 235, 1H NMR (DMSO-d6) δ 12.01 (bs, 1H), 9.17(s, 1H), 8.31(s, 1H), 7.73(d, 1H, J=9.0), 7.67(m, 2H), 7.58(m, 1H), 7.28(m, 1H), 7.07(m, 2H).
Example 106: 3-[3-(1H-Pyrrolo[2,3-b]pyridin-4-yl)-1,2,4-oxadiazol-5-yl]benzonitrile [0346]
Step 1. 1-[2-Trimethylsilyl)ethoxy]methyl-1H-pyrrolo[2,3-b]pyridine-4-carbonitrile [0347]
[0348] 4-Bromo-1-[2-(trimethylsilyl)ethoxy]methyl-1H-pyrrolo[2,3-b]pyridine (0.300 g, 0.000917 mol) was dissolved in DMF (6.5 mL, 0.084 mol) and then zinc cyanide (0.30 g, 0.0026 mol) was added. The solution was degassed with nitrogen and then bis(tri-t-butylphosphine)palladium (0.1 g, 0.0002 mol) was added. The reaction was sealed and heated in the microwave to 100 °C for 30 minutes. The reaction was allowed to cool to rt, taken up in ethyl acetate and washed with water saturated sodium carbonate, brine, dried over magnesium sulfate and concentrated to give an oil. The crude product was purified by flash column chromatography (FCC) on silica gel, eluting with a hexane: ethyl acetate gradient to give the product (0.25 gm) as a colorless oil. LC/M S (M+H)+ 274,1H NMR (CDCI3) δ 8.22 (d, 1H), 7.53(d, 1H), 7.40(d, 1H), 6.73(d, 1H), 5.65(s, 2H), 3.50(m, 2H), 0.90(m, 2H), 0.0(s, 9H).
Step 2. N-Hydroxy-1-[2-(trimethylsilyl)ethoxy]methyl-1H-pyrrolo[2,3-b]pyridine-4-carboximidamide [0349]
[0350] 1-[2-(Trimethylsilyl)ethoxy]methyl-1FI-pyrrolo[2,3-b]pyridine-4-carbonitrile (0.05 g, 0.0002 mol) was dissolved in ethanol (2.0 mL, 0.034 mol), and then hydroxylamine hydrochloride (0.023 g, 0.00033 mol) and potassium carbonate (0.10 g, 0.00073 mol) were added. The reaction was heated to reflux for 5 h, and the reaction was then allowed to cool to rt and filtered to remove the solids. The filtrate was concentrated to give the product 0.06 g as yellow oily residue, LC/MS (M+H)+ 307.
Step 3. 3-[3-(1-[2-(Trimethylsilyl)ethoxy]methyl-1H-pyrrolo[2,3-b]pyridin-4-yl)-1,2,4-oxadiazol-5-yl]benzonitrile [0351]
[0352] The crude product N-hydroxy-1-[2-(trimethylsilyl)ethoxy]methyl-1 H-pyrrolo[2,3-b]pyridine-4-carboximidamide (0.06 gm, 0.0002 mol) was dissolved in pyridine (1.0 mL, 0.012 mol) and then 3-cyanobenzoyl chloride (0.040 g, 0.00024 mol) was added at rt. This mixture was stirred for 1 h and heated to 80 °C in an oil bath. After heating for 18 h the reaction was allowed to cool tort and then diluted with ACN and concentrated in vacuo to give 3-[3-(1-[2-(trimethylsilyl)ethoxy]me-thyl-1 H-pyrrolo[2,3-b]pyridin-4-yl)-1,2,4-oxadiazol-5-yl]benzonitrile 0.08 gm as an off white residue, LC/M S (M+H)+ 418.
Step 4. 3-[3-( 1H-Pyrrolo[2,3-b]pyridin-4-yl)-1,2,4-oxadiazol-5-yl]benzonitrile [0353] The crude 3-[3-(1-[2-(trimethylsilyl)ethoxy]methyl-1 H-pyrrolo[2,3-b]pyridin-4-yl)-1,2,4-oxa-diazol-5-yl]benzoni-trile (0.08 g, 0.0002 mol) was dissolved in TFA (3.0 mL, 0.039 mol) under nitrogen and then heated to 60 °C. After heating for 2 h the reaction was allowed to cool to rt and concentrated in vacuo. The resulting residue was taken up in methanol and concentrated to remove as much of the TFA as possible. The residue was taken up in methanol (2.0 mL, 0.049 mol) and ammonium hydroxide (1 mL). This mixture was stirred at rt for 2 h and the reaction was then complete. The reaction was concentrated in vacuo to give the crude product which was purified by prep HPLC on a C-18 column eluting with a ACN:water gradient with 0.2% TFA to give the title compound (0.025 gm, 43%) (M+H)+ 288. 1H NMR (DMSO-d6) δ 12.1 (bs, 1H), 8.65(s, 1H), 8.48(d, 1H,J=6.4), 8.39(d, 1H, J=4.8), 8.16(d, 1H, J=6.4), 7.84(t, 1H, J=6.4), 7.75(d, 1 H, J=4.8), 7.68(m, 1H), 6.99 (m, 1H).
Example 107: 4-(1-Benzothien-2-yl)-1H-pyrrolo[2,3-b]pyridine [0354]
Step 1. 4-(1 -Benzothien-1 -yl)-1 -[2-(trimethylsilyl)ethoxy]methyl-1 Hpyrrolo[2,3-b]pyridine [0355]
[0356] 1-Benzothien-2-ylboronic acid (0.05 g, 0.0003 mol) and 4-bromo-1-[2-(trimethylsilyl)-ethoxy]methyl-1H-pyrro-lo[2,3-b]pyridine (0.10 g, 0.00031 mol) were combined in toluene (3.0 mL, 0.028 mol) and ethanol (1.0 mL, 0.017 mol). Potassium carbonate (0.085 g, 0.00062 mol) dissolved in water (1.0 mL) then was added and the reaction was degassed with nitrogen. Then tetrakis(triphenylphosphine)paliadium(0) (0.05 g, 0.00004 mol) was added and the reaction was heated to 120 °C in a sealed tube in the microwave for 60 minutes. This was allowed to cool to rt, taken up in ethyl acetate and washed with water 2X, brine, dried over magnesium sulfate and concentrated to give 4-(1-benzothien-2-yl)-1-[2-(trimethylsilyl)ethoxy]methyl-1H-pyrrolo[2,3-b]-pyridine (0.10 gm) as an oil, LC /MS (M+H)+ 381.
Step 2. 4-(1-Benzothien-2-yl)-1H-pyrrolo[2,3-b]pyridine [0357] Using a procedure analogous to Example 106, Step 4, but using 4-(1-benzothien-2-yl)-1-[2-(trimethylsi-lyl)ethoxy]methyl-1H-pyrrolo[2,3-b]pyridine, the title compound was prepared as a yellow powder (0.015 g, 18%), LC /MS (M+H)+: 251, 1H NMR (DMSO-d6) δ 11.95 (bs, 1H), 8.28(d, 1H, J=5.4), 8.15(s, 1H), 8.03(m, 1H), 7.96(m, 1H), 7.64(m, 1H), 7.42(m, 2H), 7.39(d, 1H, J=5.4), 6.95(m, 1H).
Example 120: 4-Fluoro-2-[1-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-3-yl]phenol [0358]
[0359] 4-Bromo-1H-pyiTolo[2,3-b]pyridine (0.050 g, 0.00025 mol) and 4-fluoro-2-(1H-pyrazol-3-yl)phenol (0.150 g, 0.000842 mol) were heated neat to 160 °C for 5 h. The reaction was allowed to cool to rt and the residue was purified by prep LC-MS on a C-18 column eluting with a water/ACN gradient containing 0.2% TFA to give the title compound, (0.052 g, 20%, as an amorphous white solid, LC/MS (M+H)+295,1H NMR (DMSO-d6) δ 12.01 (bs, 1H), 10.25(bs, 1H), 8.81(s,H), 8.35(d, 1H, J= 5.5), 7.77(d, 1H, J=9.5), 7.64(m, 1H), 7.59(d, 1H, J=5.5), 7.32(s, 1H), 7.09(m, 1H), 7.05(m, 1H), 7.01(m, 1H).
Example 127: 4-3-[3-(Trifluoromethyl)phenyl]-1H-pyrazol-1-yl-1H-pyrrolo[2,3-b]pyridine [0360]
Step 1. (2E)-3-(Dimethylamino)-1-[3-(trifluoromethyl)phenyl]prop-2-en-1-one [0361]
[0362] 1-[5-(Trifluoromethyl)phenyl]ethanone (0.20 mL, 0.0013 mol) and 1,1-dimethoxy-N,N-dimethylmethanamine (0.17 mL, 0.0013 mol) were combined in a sealed tube and heated in a microwave to 120 °Cfor 15 minutes, the reaction was allowed to cool and was concentrated to remove the residual DMF acetal, to give (2E)-3-(dimethylamino)-1-[3-(tri-fluoromethyl)phenyl]prop-2-en-1-one, 0.32 gm, as a red oil, LC /MS (M+H)+: 244.
Step 2: 3-[3-(Trifluoromethyl)phenyl]-1 H-pyrazole [0363]
[0364] The (2E)-3-(dimethylamino)-1-[3-(trifluoromethyl)phenyl]prop-2-en-1-one (0.32 g, 0.0013 mol) was dissolved in ethanol (10.0 mL, 0.171 mol) and hydrazine (0.24 mL, 0.0078 mol) under nitrogen and heated to reflux. The reaction was monitored by HPLC and was complete almost immediately. The mixture was allowed to cool to rt and concentrated to give the crude product as an oil. The product was purified by FCC on silica gel eluting with a hexane: ethyl acetate gradient to give 3-[3-(trifluoromethyl)phenyl]-1 H-pyrazole as an oil (0.25 g, 89%), LC /MS (M+H)+: 213,1H NMR (CDCI3) δ 8.06 (s, 1H), 7.99(d, 1H, J=7.5), 7.66(d, 1H, J= 2.4), 7.57(m, 1H), 7.55(d, 1H, J=7.5), 6.69(d, 1H, J= 2.4).
Step 3. 4-3-[3-(Trifluoromethyl)phenyl]-1H-pyrazol-1-yl-1H-pyrrolo[2,3-b]pyridine [0365] 4-Bromo-1 H-pyrrolo[2,3-b]pyridine (0.028 g, 0.00014 mol) and 3-[3-(trifluoromethyl)-phenyl]-1 H-pyrazole (0.03 g, 0.0001 mol) were combined neat. The reaction was heated in a sealed tube in an oil bath to 175 °Cfor20 to produce a crude product that was a black viscous gum. The crude product was purified by HPLC on a C-18 column eluting with a watenACN gradient with 0.2% TFA to give the title product (0.025 gm, 50%) as a white amorphous solid, LC /MS (M+H)+: 329, 1H NMR (DMSO-d6) δ 11.95 (bs, 1H), 8.83(d, 1H, J=2.7), 8.31(m, 3H), 7.75(m, 2H), 7.60(m, 2H), 7.35(d, 1H, J=2.7), 7.14(m, 1H).
Example 128: 3-[1-(1H-Pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-3-yl]benzonitrile [0366]
Step 1. 3-[(2E)-3-(Dimethylamino)prop-2-enoyl]benzonitrile [0367] 3-Acetylbenzonitrile (0.435 g, 0.00300 mol) and 1,1-dimethoxy-N,N-dimethylmethanamine (0.400 mL, 0.00301 mol) were combined and heated in sealed tube to 120 °C in the microwave for 15 min. The reaction was then allowed to cool to rt giving the 3-[(2E)-3-(dimethylamino)prop-2-enoyl]-benzonitrile as a red-orange crystalline material, LC /MS (M+H)+: 201.
Step 2. 3-(1 H-Pyrazol-3-yl)benzonitrile [0368] The 3-[(2E)-3-(dimethylamino)prop-2-enoyl]benzonitrile (0.600 g, 0.00300 mol) was dissolved in ethanol (20.0 mL, 0.342 mol) and hydrazine (0.56 mL, 0.018 mol) under an atmosphere of nitrogen was stirred at room temperature for 1.5 h. The reaction was concentrated in vacuo to give a dark product which was purified by FCC on silica gel, eluting with ethyl acetate-hexane 1:1 to give 3-(1 H-pyrazol-3-yl)benzonitrile as an oil (0.430g, 84%), LC/MS (M+H)+: 170.
Step 3. 3-[1-( 1H-Pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-3-yl]benzonitrile [0369] 4-Bromo-1H-pyrrolo[2,3-b]pyridine (0.075 g, 0.00038 mol) and 3-(1H-pyrazol-3-yl)benzo-nitrile (0.161 g, 0.000952 mol) were heated in sealed tube to 160 °C for 18 h. The resulting product, dark viscous gum, was purified by HPLC on a C-18 column eluting with a watenACN gradient with 0.2% TFA to give the title product (0.030 g, 27%) as a white amorphous solid, LC /MS (M+H)+: 286, 1H NMR (DMSO-d6) δ 11.95 (bs, 1H), 8.76(s, 1H), 8.36(s, 1H), 8.29(d, 1H, J=7.5), 8.25(d, 1H, J=5.0), 7.79(d, 1H, J= 7.5), 7.62(t, 1H, J= 7.5), 7.53(m, 2H), 7.25(s, 1H), 7.11(m, 1H).
Example 153: 3-[1-(1H-Pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-4-yl]benzonitrile [0370]
Step 1. 4-(4,4,5,5-Tetramethyl-1,3,2-dioxaborolan-2-yl)-1-[2-(trimethylsilyl)ethoxy]methyl-1 H-pyrazole [0371] A solution of4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1 H-pyrazole (2.0 g, 0.010 mol) and DMF (30.0 mL, 0.387 mol) was cooled to 0 °C. Sodium hydride (320 mg, 0.013 mol) (60% in oil) was added and the mixture was stirred for 10 min. [ß-(Trimethylsilyl)ethoxy]methyl chloride (2.4 mL, 0.013 mol) was added and the resulting mixture was stirred for 20 min at 0° C and 2 h at room temperature. The reaction was partitioned between water and ethyl acetate. The organic layer was washed with brine, dried over MgS04 and concentrated to give 4-(4,4,5,5-tetramethyl-1,3,2-dioxaboro-lan-2-yl)-1-[2-(trimethylsilyl)ethoxy]methyl-1 H-pyrazole as a crude material. LC/MS (M+H)+: 325, 1H NMR (CDCI3) δ 7.85 (s, 1H), 7.80(s, 1H), 5.45(s, 2H), 3.55(t, 2H), 1.35(s, 12H), 0.95(t, 2H), 0.0(s, 9H).
Step 2. 3-(-[2-(Trimethylsilyl)ethoxy]methyl-1H-pyrazol-4-yl)benzonitrile [0372] A mixture of 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1-[2-(trimethylsilyl)ethoxy]-methyl-1 H-pyrazole (150.0 mg, 0.0004625 mol) and 3-bromobenzonitrile (0.10 g, 0.00056 mol) in toluene (2.0 mL, 0.019 mol) and ethanol (0.3 mL, 0.005 mol) was treated with sodium carbonate (98 mg, 0.00092 mol) in water (0.5 mL, 0.03 mol). The mixture was degassed by bubbling nitrogen. Tetrakis(triphenylphosphine)palladium(0) (53 mg, 0.000046 mol) was added and nitrogen was bubbled for 3 min. The reaction was heated in a microwave at 80 °C for 30 min, then allowed to cool to rt and taken up in water and ethyl acetate. The organic layer was dried over MgS04, filtered and concentrated to give a crude product, which was purified by FCC on silica gel, eluting with EtOAc/Hexanes (1:5) to give 3-(1-[2-(trimethylsi-lyl)ethoxy]methyl-1H-pyrazol-4-yl)benzonitrile, as an oil, LC /MS (M+H)+: 300.
Step 3. 3-(1H-Pyrazol-4-yl)benzonitrile trifluoroacetate [0373]
[0374] A solution of 3-(1-[2-(trimethylsilyl)ethoxy]methyl-1 H-pyrazol-4-yl)benzonitrile (110.0 mg, 0.0003673 mol) was taken up in TFA (3.0 mL, 0.039 mol) and the mixture was heated in microwave at 120 °C for 3 min. The reaction mixture was allowed to cool to rt, and then concentrated to give a crude residue. The product was purified by HPLC on a C-18 column eluting with a water/ACN gradient containing 0.2% TFA to give 3-(1 H-pyrazol-4-yl)benzonitrile trifluoroacetate as an amorphous white solid, LC /MS (M+H)+: 170.
Step 4. 3-[1-( 1H-Pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-4-yl]benzonitrile [0375] A mixture of 4-bromo-1H-pyrrolo[2,3-b]pyridine (25.0 mg, 0.000127 mol) and 3-(1 H-pyrazol-4-yl)benzonitrile trifluoroacetate (23.6 mg, 0.0000833 mol) was heated at 180 °C, neat overnight. The crude residue was purified by HPLC on a C-18 column eluting with a water; ACN gradient containing 0.2% TFA to give the title compound as an amorphous white solid, LC/MS (M+H)+: 286,1H NMR (DMSO-d6) δ 11.85 (bs, 1H), 9.18(s, 1H), 8.42(s, 1H), 8.28(s, 1H), 8.25(d, 1H, J=5.0), 8.07(d, 1H, J=7.0), 7.64(d, 1 H, J=7.0), 7.56(t, 1H, J= 7.0), 7.51 (m, 1H), 7.47(d, 1 H, J=5.0), 7.03(m,1 H).
Example 170: 2-[1-(1H-Pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-4-yl]-1,3-benzoxazole [0376]
Step 1. 4-Hydrazino-1 -/2-(trimethylsilyl)ethoxy]methyl-1 H-pyrrolo[2,3-b]pyridine [0377]
[0378] To 4-bromo-1-[2-(trimethylsilyl)ethoxy]methyl-1H-pyrrolo[2,3-b]pyridine (1.98 g, 0.00605 mol) was added hydrazine (11.0 mL, 0.350 mol) followed by addition of methanol (1.0 mL, 0.025 mol) (to improve solubility). The reaction mixture was heated in a sealed tube at 97 °C (an oil bath) for 18 h. The reaction mixture was cooled to rt and formed an off-white solid precipitate. The solid was filtered off and rinsed with cold water and dried to give 4-hydrazino-1-[2-(tri-methylsilyl)ethoxy]-methyl-1H-pyrrolo[2,3-b]pyridine (1,55gm) as a light yellow solid, LC/MS (M+H)+:279,1H NMR (DM-SO-d6) δ 7.98(d, 1H), 7.91(s, 1H), 7.28(d, 1H), 6.69(s, 1H), 6.61(d, 1H), 5.58(s, 2H), 4.37(s, 2H), 3.56(t, 2H), 0.90(t, 2H), 0.0(s, 9H).
Step 2. 2-[ 1 -( 1-[2-(Trimethylsilyl)ethoxyjmethyl-1 H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-4-yl]-1,3-benzoxazole [0379]
[0380] To 4-hydrazino-1-[2-(trimethylsilyl)ethoxy]methyl-1H-pyrrolo[2,3-b]pyridine (0.083 g, 0.00030 mol) 3782-117-1 and 1,3-benzoxazol-2-ylmalonaldehyde (0.056 g, 0.00030 mol) in toluene (1.5 mL, 0.014 mol) was added molecular sieves. The mixture was heated in a sealed tube at 70 °C (an oil bath) with stirring for 2 h. The solvent was removed in vacuo and the crude product was purified by FCC on silica using ethyl acetate:hexanes 3:7 to give 2-[1-(1-[2-(trimeth-ylsilyl)ethoxy]-methyl-1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-4-yl]-1,3-benzoxazole (0.090gm) as an oil, LC/MS (M+H)+: 432.
Step 3.
[0381] Using a procedure analogous to Example 106, Step 4, but using 2-[1-(1-[2-(trimethylsilyl)-ethoxy]methyl-1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-4-yl]-1,3-benzoxazole, the title compound was prepared as a white amorphous powder (0.015 gm, 18%), LC /MS (M+H)+:302, 1H NMR (DMSO-d6) δ 11.85 (bs, 1H), 9.45(s,1H), 8.53(s, 1H), 8.36(bs, 1H), 7.7-7.6(m, 2H), 7.65(d, 1H), 7.56(bs, 1H), 7.38-7.34(m, 2H), 7.01(d,1H).
Example 172: Cyclohexyl[1-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-4-yl]methanol [0382]
Step 1. 4-(4-Bromo-1 H-pyrazol-1 -yl)-1 H-pyrrolo[2,3-b]pyridine [0383]
[0384] A mixture of 4-bromo-1 H-pyrrolo[2,3-b]pyridine (1.10 g, 0.00558 mol) and 4-bromo-1H-pyrazole (1.2 g, 0.0084 mol) was heated neat to 150 °C for 2 h. DMF was added to dissolve the crude residue. This residue was taken up in EtOAc and washed with 1N NaOH. The organic layer was washed with brine, dried over MgS04, filtered and concentrated to give a crude 4-(4-bromo-1 H-pyrazol-1-yl)-1H-pyrrolo[2,3-b]pyridine residue, LC /MS (M+H)+: 263,265.
Step 2. 4-(4-Bromo-1 H-pyrazol-1 -yl)-1-[2-(trimethylsilyl)ethoxy]methyl-1H-pyrrolo[2,3-b]pyridine [0385]
[0386] A solution of 4-(4-bromo-1 H-pyrazol-1-yl]-1-[2-(trimethylsilyl)ethoxy]methyl chloride (1.4 mL, 0.0079 mol) was added and stirred for 20 min at 0 °C. The reaction was partitioned between ethyl acetate and water. The organic layer was washed with brine, dried over MgS04 and concentrated to give the crude material. The product was purified by FCC on silica gel (EtOAc/Hexanes, 1/10) to give 4-(4-bromo-1 H-pyrazol-1-yl)-1-[2-(trimethylsilyl)ethoxy]methyl-1H-pyr-ro!o[2,3-b]pyridine as a solid product, LC /MS (M+H)+: 393, 394, 1H NMR (CDCI3) δ 8.47(d, 1H, J=7.0), 8.27(s, 1 H), 7.88(s, 1H), 7.52(d, 1H, J=4.5), 7.39(d, 1H, J=7.0), 7.069(d, 1H, J=4.5), 5.80(s, 2H), 3.6(t, 2H), 1.95(t, 2H), 0.0(s, 9H). Step 3. Cyclohexyl[1-(1-[2-(trimethylsilyl)ethoxy]methyl-1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-4-yl]methanol [0387]
[0388] A mixture of 4-(4-bromo-1H-pyrazol-1-yl)-1-[2-(trimethylsilyl)ethoxy]methyl-1H-pyrrolo[2,3-b]pyridine (50.0 mg, 0.000127 mol) in THF (2.0 mL, 0.025 mol) under a nitrogen atmosphere was cooled to -78 °C and 1.6 M n-butyllithium in water (1.00 mL, 0.0555 mol). The mixture was stirred for 3 min. The reaction was partitioned between water and EtOAc. The organic layer was dried over MgS04, filtered and concentrated to give the cyclohexyl[1-(1:5) to give 4-yl)-1H-pyrazol-4-yl]methanol as a crude residue, LC /MS (M+H)+: 417.
Step 4. Cyclohexyl[1-phenylvinyl)-1H-pyrazol-4-yl]methanol [0389] Using a procedure analogous to Example 106, Step 4, butusing cyclohexyl[1-(1-[2-(trimethylsilyl)ethoxy]methyl-1H-pyrrolo[2,3-b]pyridine, the title compound was prepared as a white amorphous powder (0.015 gm, 18%), LC /MS (M+H)+: 297. 1H NMR (DMSO-d6) δ 11.85 (bs, 1H), 8.44(s, 1H), 7.74(s, 1H), 7.50(m, 1H), 7.44(d, 1H, J=6.5.70(s, 1H), 5.37(s, 1H).
Example 173: 4-[4-(1-Phenylvinyl)-1H-pyrazol-1-yl]-1H-pyrrolo[2,3-b]pyridine [0390]
Step 1. 4-[4-(1-Phenylvinyl)-1H-pyrazol-1-yl]-1-[2-(trimethylsilyl)ethoxy]-methyl-1Hpyrrolo[2,3-b]pyridine [0391]
[0392] A mixture of (l-phenylvinyl)boronic acid (24.0 mg, 0.000162 mol) and 4-(4-bromo-1H-pyrazol-1-yl)-1-[2-(tri-methylsilyl)ethoxy]methyl-1H-pyrrolo[2,3-b]pyridine (50.0 mg, 0.000127 mol) in toluene (2.00 mL, 0.0188 mol) and ethanol (0.50 mL, 0.0086 mol) was treated with potassium carbonate (35 mg, 0.00025 mol) in water (1.00 mL, 0.0555 mol). The mixture was degassed by bubbling nitrogen. Tetrakis(triphenylphosphine)palladium(0) (10 mg, 0.00001 mol) was added and nitrogen was bubbled for 3 min. The reaction was heated in a sealed tube in the microwave at 100 °C for 30 min. The reaction was allowed to cool to rt and partitioned between ethyl acetate and water. The combined organic layer was dried over MgS04, filtered and concentrated to give the crude material The crude product was purified by FCC on silica gel eluting with EtOAc/Hexanes (1:5) to give 4-[4-(1-phenylvinyl)-1H-pyrazol-1-yl]-1-[2-(trimethylsilyl)ethoxy]me-thyl-1H-pyrrolo[2,3-b]-pyridine as a solid residue, LC /MS (M+H)+: 417.
Step 2.
[0393] Using a procedure analogous to Example 106, Step 4, but using 4-[4-(1-phenylvinyl)-1 H-pyrazol-1-yl]-1-[2-(tri-methylsilyl)ethoxy]methyl-1H-pyrrolo[2,3-b]pyridine, the title compound was prepared as an white amorphous powder (0.015 gm, 31%), LC /MS (M+H)+: 287, 1H NMR (DMSO-d6) δ 11.85 (bs, 1H), 8.63(s, 1H), 7.99(s, 1H), 7.55(bs, 1H), 7.48(m, 2H), 7.43-7.37(m, 5H), 7.01(m,1H), 5.70(s, 1H), 5.37(s, 1H).
Example 200: 4-(1-Benzyl-1 H-pyrazol-4-yl)-1 H-pyrrolo[2,3-b]pyridine [0394]
Step 1. 4-(1 -Benzyl-1 H-pyrazol-4-yl)-1 -[2-(trimethylsilyl)ethoxy]methyl-1 H-pyrrolo[2,3-b]pyridine [0395]
[0396] 4-Bromo-1-[2-(trimethylsilyl)ethoxy]methyl-1H-pyrrolo[2,3-b]pyridine (0.100 g, 0.000306 mol) was combined with 1-benzyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (0.113 g, 0.000398 mol) in toluene (3.0 mL, 0.028 mol) and ethanol (0.5 mL, 0.008 mol). Potassium carbonate (0.084 g, 0.00061 mol) dissolved in water (1.0 mL, 0.056 mol) was added and the reaction mixture was degassed with nitrogen. Tetrakis(triphenylphosphine)paliadium(0) (0.080 g, 0.000069 mol) was added, and again the mixture was degassed with nitrogen for 5 min. The reaction was heated in sealed tube to 100 °C in a microwave for 30 minutes. The reaction was partitioned between ethyl acetate and water. The organic layer was washed with water, brine, dried over magnesium sulfate and concentrated to give a crude residue. The product was purified by FCC on silica gel using ethyl acetate:hexane 3:7, to give 4-(1-benzyl-1 H-pyrazol-4-yl)-1-[2-(trimethylsilyl)ethoxy]methyl-1H-pyrrolo[2,3-b]pyridine 0.092g as a semisolid residue, LC /MS (M+H)+: 405.
Step 2. 4-(1 -Benzyl-1 H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine [0397] Using a procedure analogous to Example 106, Step 4, but using 4-(1-benzyl-1H-pyrazol-4-yl)-1-[2-(trimethyl-silyl)ethoxy]methyl-1H-pyrrolo[2,3-b]pyridine, the title compound was prepared as a white amorphous powder (0.054 gm), LC /MS (M+H)+: 275, 1H NMR (DMSO-d6) δ 12.21 (bs, 1H), 8.80(s, 1H), 8.25(vbs, 1H), 8.23(s, 1H), 7.63(s, 1H), 7.49(bs, 1H), 7.4-7.2(m, 5H), 6.99(s, 1H), 5.42(s, 2H).
Example 201 : 4-[1-(2-Naphthylmethyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine [0398]
Step 1. 1-(2-Naphthylmethyl)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole [0399] The 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (0.10 g, 0.00052 mol) was combined with naphthalene, 2-(bromomethyl)- (0.12 g, 0.00057 mol) in ACN (3.0 mL, 0.057 mol) under nitrogen at rt. Then cesium carbonate (0.50 g, 0.0015 mol) was added and the reaction was complete after stirring for 1 h. This was partitioned between ethyl acetate and brine. The organic layer was washed with brine, dried over magnesium sulfate and concentrated to give 1-(2-naphthylmethyl)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole 0.17 gm, as an oil, LC/MS (M+H)+: 335, 1H NMR (CDCI3) δ 7.89 (s, 1H), 7.79-7.84(m, 3H), 7.69(bs, 2H), 7.49-7.4(m, 2H), 7.46-7.33(m, 1 H), 5.47(s, 2H), 1.31 (s, 12H).
Step 2. 4-[1-(2-Naphthylmethyl)-1 H-pyrazol-4-yl]-1-[2-(trimethylsilyl)ethoxy]methyl-1 H-pyrrolo[2,3-b]pyridine [0400]
[0401] 4-Bromo-1-[2-(trimethylsilyl)ethoxy]methyl-1H-pyrrolo[2,3-b]pyridine (0.06 g, 0.0002 mol) and 1-(2-naphthyl-methyl)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (0.074 g, 0.00022 mol) were combined in toluene (2.0 mL, 0.019 mol) and ethanol (1.0 mL, 0.017 mol), and then potassium carbonate (0.063 g, 0.00046 mol, in 1 mL water) was added. The reaction mixture was degassed with nitrogen, then tetrakis(triphenylphosphine)palladium(0) (0.02 g, 0.00002 mol) was added, sealed in a tube and heated to 120 °C in a microwave for 30 minutes. This was allowed to cool and then partitioned between ethyl acetate and brine. The organic layer was dried over magnesium sulfate and concentrated to give 4-[1-(2-naphthylmethyl)-1H-pyrazol-4-yl]-1-[2-(trimethylsilyl)ethoxy]methyl-1H-pyrrolo[2,3-b]pyrid-ine 0.08 g, as an oily residue, LC /MS (M+H)+: 455.
Step 3 [0402] Using a procedure analogous to Example 106, Step 4, but using 4-[1-(2-naphthylmethyl)-1H-pyrazol-4-yl]-1-[2-(trimethylsilyl)ethoxy]methyl-1H-pyrrolo[2,3-b]pyridine, the title compound was prepared as a white amorphous powder (0.053 g, 88%), LC /MS (M+H)+: 325, 1H NMR (DMSO-d6) δ 12.0(bs, 1H), 8.79(s, 1H), 8.24(s, 1H), 8.19(d, 1H, J=5.7), 7.82(m, 4H), 7.56(m, 1H), 7.43(m, 4H), 6.92(m, 1H), 5.54(s, 2H).
Example 219: 4-(1-Phenyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine [0403]
Step 1. 1-phenyl-4-(4,4,5,5-Tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole [0404] 4-(4,4,5,5-Tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (0.07 g, 0.0003 mol) and phenylboronic acid (0.083 g, 0.00068 mol) were combined in DMF (1.50 mL, 0.0194 mol). Then copper(ll) diacetate (0.010 g, 0.000055 mol) and pyridine (0.069 mL, 0.00085 mol) were added. The reaction was heated in an open tube to 80 °C for40 minutes. The reaction was complete by HPLC, allowed to cool to rt, taken up in ethyl acetate, and washed with water saturated with sodium carbonate. The organic layer was washed with brine, dried over magnesium sulfate and concentrated to give 1-phenyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazo, 0.09 gm as an oily residue, LC/MS (M+H)+: 271.
Step 2. 4-(1 -Phenyl-1 H-pyrazol-4-yl)-1 -[2-(trimethylsilyl)ethoxyjmethyl-1 H-pyrrolo[2,3-b]pyridine [0405] Using a procedure analogous to Example 201, Steps B and C, but using 1-phenyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazo, the title compound was prepared as an white amorphous powder (0.015 gm, 18%), LC/MS (M+H)+: 261, 1H NMR (DMSO-d6) δ 12.05 (bs, 1H), 9.23(s, 1H), 8.53(s, 1H), 8.31(m, 1H), 8.01(m, 2H), 7.63(m, 1H), 7.57-7.52 (m, 3H), 7.36(m, 1H), 7.13(m, 1H).
Example 231: 3-[4-(1H-Pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]benzonitrile [0406]
Step 1. 4-(1H-Pyrazol-4-yl)-1-[2-(trimethylsilyl)ethoxy]methyl-1H-pyrrolo[2,3-b]pyridine [0407]
[0408] 4-Bromo-1-[2-(trimethylsilyl)ethoxy]methyl-1H-pyrrolo[2,3-b]pyridine (0.20 g, 0.00061 mol) and 4-(4,4,5,5-te-tramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (0.15 g, 0.00079 mol) were combined in DMF (5.0 mL, 0.064 mol) and then potassium carbonate (0.25 g, 0.0018 mol) in 1 mL water was added. The reaction was degassed with nitrogen, then tetrakis(triphenylphosphine)-palladium(0) (0.08 g, 0.00007 mol) was added and in a sealed tube the reaction was heated to 120 °C oil bath. The reaction was heated for 30 minutes, allowed to cool and then taken up in ethyl acetate. The reaction mixture was washed with brine, dried over magnesium sulfate and concentrated to give an oil. The product was purified by FCC on silica gel eluting with a hexane:ethyl acetate gradient to give 4-(1H-pyrazol-4-yl)-1-[2-(trimeth-ylsilyl)ethoxy]methyl-1H-pyrrolo[2,3-b]pyridine (0.13 gm, 70%) as a crystalline white powder, LC /MS (M+H)+: 315, 1H NMR (DMSO-d6) δ 13.35 (bs, 1H), 8.59(bs, 1H), 8.32(d, 1H, J=8.5), 8.26(bs, 1H), 7.76(d, 1H, J=6.0), 7.45(d, 1H, J=8.5), 7.01(d, 1H, J=6.0), 5.73(s, 2H), 3.61(t, 2H), 0.92(t, 2H), 0.0(s, 9H).
Step 2. 3-[4-( 1-[2-(Trimethylsilyl)ethoxy]methyl-1 H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1 -yljbenzonitrile [0409]
[0410] 4-(1 H-Pyrazol-4-yl)-1-[2-(trimethylsilyl)ethoxy]methyl-1 H-pyrrolo[2,3-b]pyridine (0.025 g, 0.000080 mol) and (3-cyanophenyl)boronicacid (0.023 g, 0.00016 mol) were combined in DMF(1.50 mL, 0.0194 mol). Then copper(ll) diacetate (0.002 g, 0.00001 mol) and pyridine (0.019 mL, 0.00024 mol) were added. The reaction was heated in an open tube to 125 °C for 40 minutes, allowed to cool to rt, taken up in ethyl acetate, and washed with water saturated with sodium carbonate. The organic layer was washed with brine, dried over magnesium sulfate and concentrated to give 3-[4-(1-[2-(tri-methylsilyl)ethoxy]methyl-1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]-benzonitrile(0.025gm, 92%) as an oily residue, LC /MS (M+H)+: 316.
Step 3 [0411] Using a procedure analogous to Example 106, Step 4, but using 3-[4-(1-[2-(trimethylsilyl)-ethoxy]methyl-1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]benzonitrile, the title compound was prepared as an white crystalline powder (0.012 gm, 60%), LC/MS (M+H)+: 286, 1H NMR (DMSO-d6) δ 12.05 (bs, 1H), 9.32(s, 1H), 8.59(m, 1H), 8.55(m, 1H), 8.36(m, 1H), 8.30(d, 1H, J=5.2), 7.83(m, 1H), 7.75(m, 1 H), 7.63(m, 1H), 7.51 (d, 1 H, J=5.2), 7.12(m, 1 H).
Example 250: 4-{1-[(f/?)-1-Methylbutyl]-1H-pyrazol-4-yl}-1H-pyrrolo[2,3-b]pyridine (250a) and 4-{1-[(fS)-1-Meth-ylbutyl]-1H-pyrazol-4-yl}-1H-pyrrolo[2,3-b]pyridine (250b) [0412]
Step 1. 4-[1-( 1-Methylbutyl)-1H-pyrazol-4-yl]~ 1 -[2-(trimethylsilyl)ethoxy]-methyl-1 H-pyrrolo[2,3-b]pyridine [0413] 4-(1H-Pyrazol-4-yl)-1-[2-(trimethylsilyl)ethoxy]methyl-1H-pyirolo[2,3-b]pyridine (50 mg, 0.0002 mol) (see, Example 231, Step 1) was dissolved in DMF (2 mL, 0.02 mol) and cooled at 0 °C. This solution was treated with sodium hydride (7.0 mg, 0.00029 mol) (60% in oil) and stirred for 15 min. The mixture was then treated with 2-bromopentane (40 mg, 0.0002 mol) and was stirred for 5 h. The reaction was partitioned between ethyl acetate and water. The organic layer was washed with brine, dried over MgS04, filtered and concentrated to give the crude product 4-[1-(1-methylbutyl)-1H-pyrazol-4-yl]-1-[2-(trimethylsilyl)ethoxy]methyl-1H-pyrrolo[2,3-b]pyridine as an oil, LC/MS (M+H)+: 286.
Step 2. 4-[1-(1 -Methylbutyl) -1 H-pyrazol-4-yl]-1 H-pyrrolo[2,3-b]pyridine [0414] Using a procedure analogous to Example 106, Step 4, but using 4-[1-(1-methylbuty 1)-1 H-pyrazol-4-yl]-1-[2-(tri-methylsilyl)ethoxy]methyl-1H-pyrrolo[2,3-b]pyridine, the title compound was prepared as an white amorphous powder (0.025 gm, 60%), LC /MS (M+H)+: 255, 1H NMR (DMSO-d6) δ 12.21 (bs, 1H), 8.66(s, 1H), 8.27(bs, 1H), 8.25(s, 1H), 7.62(m, 1H), 7.49(m, 1H), 7.02(m, 1H),4.46(m, 1H), 1.9-1.8(m, 1H), 1.7-1.6(m, 1H), 1.47(d, 3H), 1.2-1,0(m, 2H), 0.83(t, 3H).
Step 3. Separation of Enantiomers [0415] The separation of the enantiomers of 4-[1-(1-methylbutyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine from Step 2 was performed by chiral column preparative HPLC separation using an OD-H column eluting with an isopropanobhexane gradient to give the title compounds as amorphous white residues, LC /MS (M+H)+: 255, 1H NMR (DMSO-d6) δ 12.21 (bs, 1H), 8.66(s, 1H), 8.27(bs, 1H), 8.25(s, 1H), 7.62(m, 1H), 7.49(m, 1H), 7.02(m, 1H), 4.46(m, 1H), 1.9-1.8(m, 1H), 1.7-1.6(m, 1H), 1.47(d, 3H), 1.2-1,0(m, 2H), 0.83(t, 3H).
Example 286: 4-Methyl-3-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]benzonitrile [0416]
Step 1. 4-Methyl-3-[4-( 1-[2-(trimethylsilyl)ethoxy]methyl-1 H-pyrrolo[2,3-b]pyridin-4yl)-1 H-pyrazol-1 -yl]benzonitrile [0417]
[0418] To a mixture of 4-(1H-pyrazol-4-yl)-1-[2-(trimethylsilyl)ethoxy]methyl-1H-pyrrolo[2,3-b]-pyridine (0.050 g, 0.00016 mol) (see, Example 231, Step 1) and cesium carbonate (0.10 g, 0.00032 mol) in dry DMF (1.0 mL, 0.013 mol) was added 3-fluoro-4-methylbenzonitrile (0.043 g, 0.00032 mol). The reaction mixture was heated in sealed tube to 120 °C for 5.5 hours. The reaction was allowed to cool and partitioned between ethyl acetate and water. The organic layer was washed with water, brine, dried over magnesium sulfate, filtered, and concentrated to give 4-methyl-3-[4-(l-[2-(tri-methylsilyl)ethoxy]methyl-1 H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1 -yl]benzonitrile as a crude product, LC/MS (M+H)+: 430.
Step 2. 4-Methyl-3-[4-( 1H-pyrrolo[2,3-bJpyridin-4-yl)-1 H-pyrázol-1 -yljbenzonitrile [0419] Using a procedure analogous to Example 106, Step 4, but using 4-methyl-3-[4-(1-[2-(trimethylsilyl)ethoxy]me-thyl-1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]benzonitrile, the title compound was prepared as a white amorphous powder (0.037gm, 88%), LC/MS (M+H)+: 300, 1H NMR (DMSO-d6) δ 12.19 (bs, 1H), 8.98(s, 1H), 8.57(s, 1H), 8.31(d, 1H, J=7.0), 8.08(s, 1H), 7.89(d, 1H, J=10), 7.66(d, 1H, J=10), 7.63(m, 1H), 7.55(d, 1H), 7.07(m, 1H), 2.4(s, 3H).
[0420] Further example compounds of the invention are provided in Tables 7, 8, 9,10, and 11 below. The compounds listed in Tables 7, 8, 9, 10 and 11 are racemic unless the enantiomers are indicated separately.
Example 407: 3-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]pentanedinitrile [0421]
Step 1: Dimethyl 3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyf}-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]pentanedio-ate [0422] 4-(1H-Pyrazol-4-yl)-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidine (31.0 g, 0.0983 mol) was suspended in ACN (620 mL, 12 mol), and DBU (9.3 mL, 0.062 mol) was added under nitrogen. The reaction was heated to 65 °C and dimethyl (2E)-pent-2-enedioate (16 mL, 0.12 mol) was added in 5 mL portions over 2 h. After stirring overnight, the reaction was complete. The reaction was allowed to cool to room temperature and was concentrated in vacuo to give a dark oil. The oil was partitioned between ethyl acetate and water. The organic layer was washed with 1.0 N HCI, brine, dried over magnesium sulfate, and then concentrated to give a dark oil. The viscous oil was triturated with ethyl ether 3X 500 mL to give a dark precipitate. The oil was taken up in ethyl acetate to form a solid. The solids were collected, washed with ethyl ether and dried to give dimethyl 3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrro-lo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]pentanedioate as a white powder (29.5 gm, 64%), LC/MS (M+H)+: 474,1H NMR (DMSO-d6) δ 9.1 (s, 1H), 9.02 (s, 1H), 8.65 (s, 1H), 8.11 (d, 1H), 7.42(d, 1H), 5.78(s, 2H), 5.27(m, 1H), 3.65(m, 8H), 3.15(m, 4H), 0.95(t, 2H), 0.1(s, 9H).
Step 2: 3-[4-(7-[2-(Trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-pentanedioic acid [0423] Dimethyl 3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]pentanedi-oate (43.0 g, 0.0908 mol) was dissolved in methanol (271.2 mL, 6.695 mol) and lithium hydroxide monohydrate (15 g, 0.36 mol) dissolved in water (125 mL) was added. The reaction was stirred at rt for 2 h. The methanol was removed in vacuo and a resulting aqueous layer was cooled in an ice bath. The solution was made acidic pH~4 with 1N HCI to give a white precipitate. The solid precipitate was collected, washed with water, dried to give 3-[4-(7-[2-(trimethylsi-lyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]pentanedioicacid as a white crystalline powder (31.8 gm, 80%), LC /MS (M+H)+: 446, 1H NMR (DMSO-d6) δ 8.85s 1H), 8.75(s, 1H), 8.42(s, 1H), 7.85(d, 1H), 7.17(d, 1H), 5.71(s, 2H), 5.18(m,1H), 3.65(t, 2H), 3.05(m,4H), 0.92(t, 2H), 0.1(s, 9H).
Step 3: 3-[4-(7-[2-(Trimethylsilyl)ethoxyJmethyl-7H-pyrrolo[2,3-dJpyrimidin-4-yl)-1H-pyrazol-1-yl]-pentanediamide [0424] 3-[4-(7-[2-(Trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]pentanedioic acid (31.80 g, 0.07137 mol) was dissolved in DMF (636 mL, 8.21 mol) under nitrogen cooled in an ice bath and CDI (34.7 g, 0.214 mol) was added. This mixture was allowed to stir for 30 minutes and then allowed to warm to rt. After stirring for 2 h ammonia (12.2 g, 0.714 mol) was bubbled through the solution for 30 minutes giving a cloudy suspension. The reaction mixture was concentrated to remove some of the DMF (~200 mL) and then water was added slowly to give a white precipitate. This mixture was cooled in an ice bath and the solid precipitate was collected, washed with water and dried in vacuo to give 3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]pentane-diamide as a white powder (29.0 gm, 91 %), LC /MS (M+H)+: 444,1H NMR (DMSO-d6) δ 8.85(s, 1H), 8.59(s, 1H), 8.40(s, 1H), 7.87(d,1H), 7.75(s,2H), 7.15(d, 1H), 6.95(s, 2H), 5.73(s, 2H), 5.29(m,1H), 3.63(t, 2H), 2.82(m, 2H), 2.73(m, 2H), 0.90(t, 2H), 0.1(s, 9H).
Step 4:3-[4-(7-[2-(TrimethylsHyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-pentanedinitrHe [0425] 3-[4-(7-[2-(Trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]pentanediamide (29.0 g, 0.0654 mol) was partially dissolved in DMF (200 mL, 2 mol), DCM (200 mL, 3 mol) and TEA (36 mL, 0.26 mol) and cooled in an ice bath under nitrogen atmosphere. The trichloroacetyl chloride (15 mL, 0.14 mol) was added dropwise turning the reaction to a dark solution. This was stirred at 0 °C for 1/2 h. The reaction was then concentrated to remove the DCM and the resulting DMF solution was diluted with water to precipitate the product. The solid precipitate was collected and washed with water to give a dark solid. The solid was then dissolved in DCM and washed with brine, dried over magnesium sulfate and concentrated to give a very dark oily residue. The residue was taken up in DCM, and hexane was added until the solution became slightly cloudy. This was stirred at rtto precipitate 3-[4-(7-[2-(trimethylsilyl)ethoxy]me-thyl-7H-pyrrolo[2,3-d]-pyrimidin-4-yl)-1 H-pyrazol-1 -yl]pentanedinitrile as white needle-like crystals (22.7 gm, 85%), LC /MS (M+H)+: 408, 1H NMR (DMSO-d6) δ 9.07(s, 1H), 8.87(s, 1H), 8.59(s, 1H), 7.88(d, 1H), 7.19(d, 1H), 5.75(s, 2H), 5.30(m,1 H), 3.62(t, 2H), 3.40(m, 4H), 0.91 (t, 2H), 0.10(s, 9H).
Step 5: 3-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1 -yljpentanedinitrile [0426] 3-[4-(7-[2-(Trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yljpentanedinitrile (10.0 g, 0.0245 mol) was dissolved in ACN (200 mL, 3.83 mol) and water (20 g, 1.1 mol) at rt. To this lithium tetrafluoroborate (23.0 g, 0.245 mol) was added giving a cloudy solution. The reaction was heated to reflux and monitored by HPLC. After heating for 24 h the reaction was allowed to cool to rt and then cooled in an ice bath. To this, ammonium hydroxide (23 mL, 0.59 mol) was added slowly. The reaction was allowed to warm to rt. After stirring for 18 hs the reaction was diluted with water and concentrated in vacuo to remove the ACN, giving a precipitate. The solids were collected, washed with water and dried to give the title compound as an off-white solid (6. 2 gm, 91%), LC /MS (M+H)+: 278,1H NMR (DMSO-d6) δ 8.9(s, 1H), 8.72(s,1H), 8.43(s, 1 H), 7.59(d, 1H), 6.92(d, 1H), 5.21(m,1H), 3.25(m, 4H).
Example 421: 5-{2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}-pyridine-2-carbonitrile trifluoroacetate [0427]
Step 1: 3-(6-Chloropyridin-3-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yljpropane-nitrile [0428] 3-(6-Chloropyridin-3-yl)-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]-pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile (prepared by methods analogous to those described for Example 61) (0.070 g, 0.00014 mol) in TFA (3.0 mL, 0.039 mol) and DCM (3.0 mL, 0.047 mol) was stirred at room temperature for 1 hour. Solvent was removed in vacuo, and the residue was dissolved in methanol (4.0 mL, 0.099 mol) and ethylenediamine (0.07 mL, 0.001 mol). The reaction mixture was stirred at room temperature overnight. Solvent was removed in vacuo, the crude product was purified by preparative HPLC eluting with an ACN; water gradient buffered with ammonium hydroxide to pH=10, to give 3-(6-chloropyridin-3-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile as a white powder (35mg,69%), LCMS (M+1)+:350,1H NMR (DMSO-d6) δ 12.21 (b,1H), 9.00 (s,1H), 8.78 (s,1H), 8.62 (s,1H), 8.58 (s,1H), 8.00(m,1H), 7.70(m,2H), 7.00(s,1H), 6.22(m,1H), 3.90(m,1H), 3.78(m,1H)
Step 2: 5-2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1 -yl]ethylpyridine-2-carbonitrile trifluoroacetate [0429] A mixture of 3-(6-chloropyridin-3-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile (0.025 g, 0.000071 mol) and zinc cyanide (0.08 g, 0.0007 mol) in DMF (1.0 mL, 0.013 mol) was degassed with nitrogen. To this mixture, tetrakis(triphenylphosphine)palladium(0) (0.04 g, 0.00004 mol) was added and the resulting mixture degassed again with dinitrogen. The reaction mixture was heated in a sealed tube at 170 °C for 15 minutes in a microwave (Personal Chemistry). After cooling to room temperature, the solids were filtered, rinsed with DMF and the combined solvent was concentrated in vacuo. The residue was triturated with hexanes (3x), and hexanes washes were discarded. The crude product was purified by preparative HPLC eluting with an ACN; water gradient containing 0.2% TFA to give the title compound as a white powder (16 mg, 49.27%), LCMS (M+1)+: 341, 1H NMR (DMSO-d6) δ 12.50(b,1H), 9.05(s,1H), 8,89(s,1H), 8,80(s,1H), 8.58(s,1H), 8.18(m,2H), 7.78(s,1H), 7.05(s,1H), 6.20(m,1H), 3.90(m,1H), 3.77(m,1H).
Example 428: 4-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]heptanedinitrile [0430]
Step 1: 3-[4-(7-[2-(Trimethylsilyl)ethoxy]methyl-7Hpyrrolo[2,3-d]pyrimidin-4-yl)-1 Hpyrazol-1-yl]pentane-1,5-diol [0431] Diethyl 3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]pentanedioate, prepared substantially as described in Example 407 (0.80 g, 0.0016 mol), was dissolved in THF (40 mL, 0.49 mol) and cooled in an ice bath under a nitrogen atmosphere. To this mixture, 1.0 M lithium tetrahydroaluminate in THF (3.2 mL) was added slowly. The reaction was stirred for 1 h, quenched with ice and partitioned between ethyl acetate and 1 N HCI. The organic layer was washed with brine, dried over magnesium sulfate and concentrated to give an amber oil. The product was purified by FCC on silica gel eluting with an ethyl acetate: methanol gradient to give 3-[4-(7-[2-(trimeth-ylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]pentane-1,5-diol as a clear viscous oil (0.51 gm, 76%), LC/MS (M+H)+: 418,1H NMR(DMSO-d6) δ, 8.85(s, 1H), 8.41(s, 1H), 8.37(s, 1H), 7.45(d,1H), 6.83(d, 1H), 5.73(s, 2H), 4.91(m, 1H), 3.75(m,2H), 3.59(m, 2H), 3.45(m,2H), 2.18(m, 4H), 0.95(m,2H), 0.1(s, 9H).
Step 2: 3-[4-(7-[2-(Trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]pentane-1,5-diyl dimethanesulfonate [0432] A mixture of 3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]pentane-1,5-diol (50 mg, 0.0001 mol) in DCM (2 mL, 0.03 mol) was cooled at 0 °C. To this mixture, TEA (50 μί, 0.0004 mol) was added. The reaction was stirred for 15 minutes. Methanesulfonyl chloride (23 μί, 0.00030 mol) was added and the resulting mixture was stirred for 1 hour. Water was added and the product was extracted with ethyl acetate. The combined extracts were washed with saturated sodium chloride, dried over magnesium sulfate, filtered and concentrated to give 3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-pentane-1,5-diyl dimethanesulfonate (57 mg, 80 %) as an oil. MS(ES): 574 (M+1).
Step 3: 4-[4-(7-[2-(Trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2.3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]heptanedinitrile [0433] To a mixture of 3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]pen-tane-1,5-diyl dimethanesulfonate (57 mg, 0.000099 mol) in DMSO (1 mL, 0.01 mol), sodium cyanide (10 mg, 0.0003 mol) was added and the mixture was stirred for 2 hours. The mixture was heated at 60 °C for 1 hour. Water was added and the product was extracted with ethyl acetate. The combined extracts were washed with saturated sodium chloride, dried over magnesium sulfate, filtered and concentrated to give 4-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]heptanedinitrile (40 mg, 90 %) as an oil. MS(ES): 436 (M+1).
Step 4:4-[4-(7H-Pyrrolo[2,3-d]pynmidin-4-yl)-1H-pyrazol-1-yl]heptanedinitrile [0434] Using a procedure analogous to Example 61 for the removal of the SEM protecting group, the title compound was prepared as a white amorphous solid, (17 mg, 60%) 1H NMR (400 MHz, DMSO): δ 8.75 (s, 1H), 8.65 (s, 1H), 8.4 (s, 1H), 7.6 (d, 1H), 7.0 (d, 1H), 4.5 (m, 1H), 2.35 (m, 4 H), 2.2 (m, 4H). MS(ES): 306 (M+1).
Example 429: 3-(5-Bromopyridin-3-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl] propanenitrile [0435]
Step 1: (2Z&E)-3-(5-Bromopyridin-3-yl)acrylonitrile [0436]
[0437] To a mixture of 1.0 M potassium tert-butoxide in THF (2.7 mL) at 0 °C (water-ice bath, under an atmosphere of nitrogen) was added diethyl cyanomethylphosphonate (0.48 mL, 0.0030 mol) in THF (4.0 mL, 0.049 mol), dropwise. The reaction mixture was warmed to room temperature, and then was cooled to 0 °C, followed by dropwise addition of 5-bromonicotinaldehyde (0.5 g, 0.003 mol) in THF (1.0 mL, 0.012 mol). After stirring at room temperature for 20 hours, the reaction mixture was quenched with water and extracted with ethyl acetate. The organic layer was washed with brine, dried over anhydrous magnesium sulfate, filtered, and concentrated to give a crude product as a dark oil. The crude product was purified by flash chromatography on silica gel using ethyl acetate-hexanes 3:7 as eluent to give a mixture of cis and trans isomers (2)-3-(5-bromopyridin-3-yl)acrylonitrile as an off-white solid (268 mg, 47.69%). LCMS (M+1)+: 209,211, 1H NMR(400 MHz, CDCI3): δ 8.75(s,1H), 8.62(s,1H), 7.90(s,1H), 7.40(d,1H), 6.00(d, 1H).
Step 2: 3-(5-Bromopyridin-3-yl)-3-[4-(7-[2-(trimethylsilyl)ethoxy]melhyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yljpropanenitrile [0438] To 4-(1H-pyrazol-4-yl)-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidine (0.200 g, 0.000634 mol) in 1.0 mL of dry ACN was added DBU (0.10 mL, 0.00067 mol), followed by the addition of (2Z&E)-3-(5-bromopyridin-3-yl)acrylonitrile (0.234 g, 0.00112 mol) in 1.0 mL of ACN. The reaction mixture was stirred at 67 °C for 4 hours. Upon cooling, the mixture was partitioned between dilute hydrochloric acid and ethyl acetate. The organic layer was washed with saturated sodium chloride, dried over anhydrous sodium sulfate, and concentrated. The crude product was purified by flash chromatography on silica gel using ethyl acetate : hexanes (7:3) to give 3-(5-bromopyridin-3-yl)-3-[4-(7-[2-(tri-methylsilyl)-ethoxy]-methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propanenitrile as an off-white solid (225 mg, 67.66%). LCMS (M+1 )+:524,526:1H NMR (400 MHz, CDCI3): δ 8.90(s, 1H), 8.80(s, 1H), 8.70(s, 1H), 8.42(s, 1H), 8.40(s, 1H), 8.00(s, 1H), 7.50(d, 1H), 6.82(d, 1H), 5.81(m, 1H), 5.75(s, 2H), 3.70(m,1H), 3.60(m, 2H), 3.42(m, 1H), 1.00(m, 2H), 0.08(s, 9H).
Step 3: 3-(5-Bromopyridin-3-yl)-3-[4-(7H-pyrrolo[1,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yljpropanenitrile [0439] The 3-(5-bromopyridin-3-yl)-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]-pyrimidin-4-yl)-1H-pyra-zol-1-yljpropanenitrile (0.220 g, 0.000419 mol) in DCM (9.0 mL, 0.14 mol) and TFA (9.0 mL, 0.12 mol) was stirred at room temperature for 1 hour. The reaction was concentrated in to give a residue. This crude intermediate was dissolved in methanol (12 mL, 0.30 mol) and ethylenediamine (0.2 mL, 0.003 mol) and was stirred overnight at room temperature. The reaction was concentrated in vacuo to give the crude product which was purified by preparative HPLC eluting with a water : ACN gradient buffered with ammonium hydroxide (p=10) to give 3-(5-bromopyridin-3-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yljpropanenitrile as an amorphous white powder (118 mg, 71.36%). LCMS (M+1)+:394,396, 1H NMR (400 MHz, DMSO-d6): δ 12.05(bs,1H), 8.98(s, 1H), 7.0(s, 1H), 6.50(m, 2H), 8.50(s, 1H), 8.10(s, 1H), 7.80(s, 1H), 6.98(s, 1H), 6.21(m, 1H), 3.90(m, 1H), 3.70(m, 1H).
Example 430: 3-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]pentane-1,5-diol [0440]
[0441] Using a procedure analogous to Example 61 forthe removal ofthe SEM protecting group but using 3-[4-(7-[2-(tri-methylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]-pentane-1,5-diol from Example 428, the title compound was prepared as a white amorphous solid, (25 mg, 70%) 1H NMR (400 MHz, DMSO): δ 8.65 (s, 1H), 8.6 (s, 1H), 8.25 (s, 1H), 7.6 (d, 1H), 6.0 (d, 1H),4.6(m, 1H), 3.3 (m, 2H), 3.2 (m, 2H), 2.1 (m, 2H), 1,9(m,2H). MS(ES): 288 (M+1).
Example 431: 5-(2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl)-nicotinonitrile bis(trif-luoroacetate) [0442]
[0443] A slurry of 3-(5-bromopyridin-3-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propanenitrile (0.050 g, 0.00013 mol) (from Example 429), DMF (2.0 mL, 0.026 mol) and zinc cyanide (0.1 g, 0.001 mol) was degassed by purging with nitrogen. Then tetrakis(triphenylphosphine)palladium(0) (0.07 g, 0.00006 mol) was added and the resulting slurry again was degassed with nitrogen. The reaction was sealed and heated at 170 °C for 15 minutes in a microwave (Personal Chemistry). The reaction was allowed to cool and the solids were filtered off. The combined DMF fractions were concentrated in vacuo. The residue was triturated with ethyl acetate-hexanes 2:8, then with ethyl ether to removed by-products. The crude product was purified by preparative HPLC eluting with a water : acetontrile gradient containing 0.2% TFA to give the racemic title compound (43 mg, 59.65%). LCMS (M+1)+:341, 1H NMR (400 MHz, DMSO-d6): δ 12.60(bs, 1H), 9.10(s, 1H), 8.90(s, 1H), 8.80(s, 1H), 8.50(s, 1H), 8.42(s, 1H), 7.78(s, 1H), 7.10(s, 1H), 6.30(m, 1H), 3.90(m, 1H), 3.70(m,1H).
Example 431R and Example 431S
[0444] The enantiomers R-5-(2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-ethyl)nicotinonitrile and S-5-(2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-ethyl)nicotinonitrile were separated by chiral column HPLC.
Example 467: 3-(3-Aminophenyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propanenitrile bis(trif-luoroacetate) [0445]
Step 1: 3-(3-Nitrophenyl)-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]pro-panenitrile [0446] To 4-(1H-pyrazol-4-yl)-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidine (0.500 g, 0.00158 mol) in 8.0 mL of dry ACN was added DBU (0.24 mL, 0.0016 mol) followed by addition of (2Z)-3-(3-nitrophenyl)acrylonitrile (0.36 g, 0.0021 mol) in 2.0 mL of ACN. The reaction mixture was heated at 67 °C for 18 hours. This was cooled to room temperature, and the mixture was partitioned between diluted hydrochloric acid and ethyl acetate. The organic layer was washed with saturated sodium chloride, dried over anhydrous magnesium sulfate, and concentrated. The crude product was purified by flash chromatography on silica gel using ethyl acetate-hexanes 6:4, to give 3-(3-nitrophenyl)-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile as a dark orange oil, (688 mg, 85.65%). LCMS (M+1 )+:490
Step 2. 3-(3-Aminophenyl)-3-(4-7-[2-(trimethylsilyl)ethoxy]-7H-pyrrolo[2,3-d]pyrimidin-4-yl-1H-pyrazol-1-yl)propaneni-trite [0447] The 3-(3-nitrophenyl)-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile (0.630 g, 0.00129 mol) was dissolved in ethanol (65 mL, 1.1 mol), degassed with nitrogen, and then palladium (0.55 g, 0.0052 mol) (10% on carbon) was added. The reaction mixture was again purged with nitrogen, and it was then charged at 50 psi hydrogen in a Parr shaker for 60 minutes. The reaction mixture was filtered and concentrated to give 3-(3-amino-phenyl)-3-(4-7-[2-(trimethylsilyl)ethoxy]-7H-pyrrolo[2,3-d]pyrimidin-4-yl-1H-pyrazol-1-yl)propane-ni-trile as a colorless oil (550 mg, 95.92%), LCMS (M+1 )+=460,
Step 3. 3-(3-Aminophenyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile bis(trifluoroacetate) [0448] Using a procedure analogous to that of Example 61 for the removal of the SEM protecting group, the title compound was prepared as a white amorphous solid (18 mg, 38%), LCMS (M+1)+=329: 1H NMR (DMSO-d6) δ 12.61 (b, 1H),9.00(s,1H),8.80(s,1H),8.50(s,1H),7.78(m,1H),7.25( m,1H),7.18(m,1H),6.85(m,2H),6.02(m.1H),3.78(m,1H), 3.60 (m,1H).
Example 468: N-(3-(2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl)-phenyl)acetamide trif-luoroacetate [0449]
Step 1 -(3-2-Cyano-1-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethylphe-nyl)acetamide [0450] To 3-(3-aminophenyl)-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile (0.070 g, 0.00015 mol) (from Example 467) in dry DCM (1.0 mL, 0.016 mol) was added TEA (0.042 mL, 0.00030 mol). The reaction was cooled in an ice bath and acetyl chloride (0.016 mL, 0.00023 mol) was added. The reaction mixture stirred for 30 minutes and was diluted with water and extracted with ethyl acetate (2x). The combined organic layers were washed with saturated sodium chloride, dried over anhydrous magnesium sulfate, filtered, and concentrated in vacuo to give N-(3-2-cyano-1-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethylphenyl)acetamide as a colorless oil, (65 mg, 85.08%), LCMS(M+1)+= 502.
Step 2 N-(3-2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethylphenyl)acelamide trifluoroacetate [0451] Using a procedure analogous to that of Example 61 for the removal of the SEM protecting group, the title compound was prepared as a white amorphous solid (40 mg, 68.9%), LCMS(M+1 )+=372,1H NMR (DMSO-dg) δ 12.61 (b,1 H), 9.05 (s,1 H), 8.79 (s,1H), 8.44 (s,1H), 7.85 (s,1H), 7.55 (s,1H), 7.48 (d,1H), 7.24 (m,1H), 7.10 (m,2H)), 6.05 (m,1 H), 3.70 (m,1H), 3.48 (m,1H), 1.98 (s,3H).
Example 470: 4-(2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl)-thiophene-2-carbonitrile trifluoroacetate [0452]
Step 1 4-Bromo-2-(diethoxymethyl)thiophene [0453] A mixture of 4-bromothiophene-2-carbaldehyde (1.2 g, 0.0063 mol) in ethanol (10 mL, 0.2 mol) was treated with ammonium chloride (0.42 g, 0.0078 mol) and ethyl orthoformate (1.2 g, 0.0078 mol). The mixture was stirred at 60 °C for 2 hours. The reaction was quenched with water and extracted with ethyl acetate. The combined organic layer was washed with saturated sodium chloride, dried over magnesium sulfate, filtered and concentrated to give 4-bromo-2-(diethoxymethyl)thiophene as an oil (1.3 g, 81%). 1H NMR (400 MHz, CDCI3): δ 7.22 (s, 1H), 6.99 (s, 1H), 5.68 (s, 1H), 3.63 (q, 4H) 1.24 (t, 6H).
Step 2 5-(Diethoxymethyl)thiophene-3-carbaldehyde [0454] A solution of 4-bromo-2-(diethoxymethyl)thiophene (500 mg, 0.002 mol) in ether (5 mL, 0.05 mol) was cooled at -78 °C. To this solution, 2.5 M n-butyllithium in hexane (0.83 mL) was added dropwise. The reaction was stirred at -78 °Cfor 1 hour. To the reaction was added DMF (0.4 g, 0.006 mol) at-78 °C and the mixture was stirred for 30 minutes. The reaction was quenched with waterand extracted with ethyl acetate. The combined organic layers were washed with saturated sodium chloride, dried over magnesium sulfate, filtered and concentrated. The crude residue was purified by flash column chromatography to yield the 5-(diethoxymethyl)thiophene-3-carbaldehyde as an oil (170 mg, 42.0%). By 1H NMR two different regioisomers of aldehydes were formed and were not separated; (note: NMR shifts are for the major isomer only) 1H NMR (400 MHz, CDCI3): δ 9.85 (s, 1H), 8.05, 7.7 (s, 1H), 7.45, 7.15 (s, 1H), 5.7 (s, 1H), 3.65 (m, 2H), 1.25 (m, 2H).
Step 3 (2E)-3-[5-(Diethoxymethyl)-3-thienyl]acrylonitrile [0455] To a solution of diethyl cyanomethylphosphonate (100 mg, 0.0008 mol) in THF (2 mL, 0.02 mol) cooled at 0 °C and 1.0 M potassium tert-butoxide in THF (0.8 mL) was added dropwise. The bath was removed and the reaction was warmed to room temperature for 30 minutes. The reaction was cooled to 0 °C and a solution of 5-(diethoxyme-thyl)thiophene-3-carbaldehyde (170 mg, 0.00079 mol) in THF (2 mL, 0.02 mol) was added drop wise. The reaction was stirred overnight at room temperature. The reaction was partitioned between water and ethyl acetate. The combined extracts were washed with saturated sodium chloride, dried over magnesium sulfate, filtered and concentrated. The crude residue was purified by flash column chromatography on silica gel eluting (ethyl acetate:hexane, 1:5) to give (2E)-3-[5-(diethoxymethyl)-3-thienyl]acrylonitrile as an oil (160 mg, 84.9%). 1H NMR (300 MHz, CDCI3): δ 7.4-7.0 (m, 3H), 5.65 (m 1H), 4.2 (m, 1H), 3.65 (m, 4H), 1.25 (m, 6H).
Step 4 3-[5-(Diethoxymethyl)-3-thienyl]-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1 -yl]propanenltrlle [0456] To a solution of 4-(1H-pyrazol-4-yl)-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]-pyrimidine (200 mg, 0.0007 mol) in ACN (2 mL, 0.04 mol) and (2E)-3-[5-(diethoxymethyl)-3-thienyl]acrylonitrile(160 mg, 0.00067 mol) (mixture of regioisomers) DBU (80 μι, 0.0005 mol) was added. The reaction was stirred overnight than water was added and the product was extracted with ethyl acetate. The combined extracts were washed with saturated sodium chloride, dried over magnesium sulfate, filtered and concentrated. The crude residue was purified by flash column chromatography on silica gel eluting (50% EtOAc/Hexane) to give 3-[5-(diethoxymethyl)-3-thienyl]-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile (160 mg, 43%). 1H NMR (400 MHz, CDCI3): δ 8.92 (s, 1H), 8.41 (s, 1H), 8.29 (b, 1H), 7.45(d, 1H), 7.41(d, 1H), 7.15 (s, 1H), 7.05 (d, 1H), 6.82 (m, 1H), 5.74 (d, 2H), 3.74 (m, 2H), 3.71 (m, 8H), 3.59 (m, 1H), 1.32 (m, 4H), 0.95 (m, 2H), -0.08 (s, 9H); MS(ES):553 (M+1).
Step 5 3-(5-Formyl-3-thienyl)-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yljpropanenitrile [0457] A solution of 3-[5-(diethoxymethyl)-3-thienyl]-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimi-din-4-yl)-1H-pyrazol-1-yl]propanenitrile (70 mg, 0.0001 mol) in THF (1 mL, 0.01 mol) was treated with 1 M HCI in water (400 μί). The reaction was stirred at room temperature. Water was added and the product was extracted with ethyl acetate. The combined extracts were washed with saturated sodium chloride, dried over magnesium sulfate, filtered and concentrated to give 3-(5-formyl-3-thienyl)-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile as a semisolid residue (60 mg, 98%). 1H NMR (400 MHz, CDCI3): δ 9.96 (s, 1H), 8.89 (s, 1H), 8.44 (m,2H), 7.46 (1H), 5.73 (s, 2H), 4.15 (m, 1H), 3.73-3.43 (m, 3H), 1.35 (m, 1H), 1.01 (m, 2H), 0.03 (s, 9H); MS(ES): 479 (M+1).
Step 6: 5-[(E)-(Hydroxyimino)methyl]-3-thienyl-3-[4-(7-[2-(trimethylsHyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1 -yljpropanenitrile [0458] A solution of 3-(5-formyl-3-thienyl)-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo-[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile (65 mg, 0.00014 mol) in methanol (2 mL, 0.05 mol) was treated with hydroxylamine hydrochloride (11 mg, 0.00016 mol) and potassium bicarbonate (23 mg, 0.00023 mol). The reaction was stirred at room temperature for 4 hours. Water was added and the product was extracted with ethyl acetate. The combined extracts were washed with saturated sodium chloride, dried over magnesium sulfate, filtered and concentrated to give 3-5-[(E)-(hy-droxyimino)methyl]-3-thienyl-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]-pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile as a semisolid oil (60 mg, 89.5%). (The crude product contained both isomers of oxime and also both regioisomers of thiophene). MS (ES): 494 (M+1).
Step 7: 4-(2-Cyano-1 -[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1 -yljethyl)thi-ophene-2-carbonitrile [0459] To a mixture of 3-5-[(E)-(hydroxyimino)methyl]-3-thienyl-3-[4-(7-[2-(trimethylsilyl)ethoxy]-methyl-7H-pyrro-lo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile (70 mg, 0.0001 mol) in pyridine (1 mL, 0.01 mol), methanesulfonyl chloride (100 μΙ_, 0.001 mol) was added. The mixture was stirred at 60 °Cfor2 hours. Water was added and the product was extracted with ethyl acetate. The combined extracts were washed with 0.1 N HCI, brine, dried over magnesium sulfate, filtered and concentrated to give 4-(2-cyano-1-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]-pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl)thiophene-2-carbonitrile as a crude product (30 mg, 44%). MS (ES): 476 (M+1).
Step 8:4-(2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl)thiophene-2-carbonitrile trifluoroacetate [0460] A mixture of 4-(2-cyano-1-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]-pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl)thiophene-2-carbonitrile (50 mg, 0.0001 mol) in DCM (2 mL, 0.03 mol) and TFA (1 mL, 0.01 mol) was stirred for 1 hour. The starting material was consumed and the desired methyl hydroxy compound was formed. The mixture was concentrated in vacuo to remove TFA. The crude intermediate was dissolved in methanol (3 mL, 0.07 mol) and was treated with ethylenediamine(1 mL, 0.01 mol). The mixture was stirred overnight and concentrated in vacuo. The products were purified by preparative FIPLC eluting with ACN: water with 0.2% TFA to give two regioisomers, the title compound as an amorphous white solid (30 mg, 60 %). 1H NMR (500 MHz, DMSO): δ 8.95 (s, 1H), 8.76 (s, 1H), 8.48 (s, 1H), 8.06 (s, 1H), 8.04 (s, 1H), 7.70 (d, 1H), 7.05 (d, 1H), 6.25 (m, 1H), 3.80-3.60 (m, 2H); MS (ES): 346 (M+1).
Example 471 :5-(2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl)-thiophene-2-carbonitrile trifluoroacetate [0461]
[0462] Isolated as the second regioisomer from Example 470, the title compound was isolated as an amorphous white solid (4 mg, 8%). 1H NMR (500 MHz, DMSO): δ. 9.0 (s, 1H0, 8.75 (s, 1H), 8.50 (s, 1H), 7.95 (s, 1H), 7.65 (s, 1H), 7.45 (s, 1H), 7.0 (d, 1H), 6.45 (m, 1H), 3.8 (dd, 2 H); MS (ES): 346 (M+1).
Example 472 : 3-[3-(Morpholin4-ylcarbonyl)phenyl]-3-[4-(7H-pyrrolo[2,3-dipyrimidin4-yl)-1H-pyrazol-1-yl]pro-panenitrile trifluoroacetate [0463]
Step 1: 3-(2-cyano-1-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7Hpyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrázol-1 -yl]ethyl)benzoic acid [0464] To a solution of methyl 3-2-cyano-1-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethylbenzoate (50 mg, 0.0001 mol) (prepared as in Example 61) in methanol (2 mL, 0.05 mol), lithium hydroxide (1 mg, 0.0001 mol) in water (1 mL, 0.06 mol) was added slowly. Water was added and also some 1N HCI was added until the solution was slightly acidic. The aqueous layer was extracted with ethyl acetate. The combined extracts were dried over magnesium sulfate, filtered and concentrated to give 3-(2-cyano-1-[4-(7-[2-(trimethylsi-lyl)ethoxy]-methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl)benzoic acid as a crude residue (35 mg, 72.0%). MS (ES): 489 (M+1).
Step 2: 3-[3-(Morpholine-1-ylcarbonyl)phenyl]-3-[4-(7-([2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidine-4-yl)-1H-pyrazole-1-yl]propanenitrHe [0465] To a solution of 3-(2-cyano-1-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]-pyrimidin-4-yl)-1 H-pyra-zol-1-yl]ethyl)benzoic acid (40 mg, 0.00008 mol) in DMF (1 mL, 0.01 mol), N,N,N’,N’-tetramethyl-0-(7-azabenzotriazol-1-yl)uronium hexafluorophosphate (36 mg, 0.000095 mol) and DIPEA (30 μί, 0.0002 mol) were added. The reaction was stirred for 10 minutes and then morpholine (10 mg, 0.00012 mol) was added and the resulting mixture was stirred for 3 hours. Water was added and the product was extracted with ethyl acetate. The combined organic extracts were washed with 1N HCI, brine, dried over magnesium sulfate, filtered and concentrated to give 3-[3-(morpholine-1-ylcarb-onyl)phenyl]-3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidine-4-yl)-1H-pyrazole-1-yl]propanenitrile as a crude (40 mg, 88%) product. MS (ES): 558 (M+1).
Step 3: 3-[3-(Morpholin-4-ylcarbonyl)phenyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propanenitrile trif-luoroacetate [0466] Using a procedure analogous to that of Example 61 for the removal of the SEM protecting group, the title compound was isolated as an amorphous white solid (18 mg, 50 %). 1H NMR (400 MHz, DMSO): δ. 9.05 (s, 1H), 8.75 (s, 1H), 8.44 (s, 1H), 7.85 (b, 1H), 7.665 (s, 1H), 7.55- 7.35 (m, 3H), 7.15 (s, 1H), 6.15 (m, 1H), 3.85 (m, 1H), 3.65-3.4 (m, 6H), 3.25 (m, 2H), 3.05 (m, 1H); MS(ES): 428 (M+1).
Example 482: 3-(5-Phenylpyridin-3-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile trif-luoroacetate [0467]
Step 1: 3-(5-Phenylpyridin-3-yl)-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]-pyrimidin-4-yl)-1H-pyrazol-1-yljpropanenitrile [0468] To a solution of 3-(5-bromopyridin-3-yl)-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile(from Example429) (60 mg, 0.0001 mol) in 1,4-dioxane(2 mL, 0.02 mol), phenylboronic acid (15 mg, 0.00012 mol) and sodium bicarbonate (30 mg, 0.0003 mol) in water (0.5 mL, 0.03 mol) were added. The resulting mixture was degassed using nitrogen. Tetrakis(triphenylphosphine)palladium(0) (10 mg, 0.00001 mol) was added and nitrogen was bubbled through the reaction again. The reaction was heated at 80 °C in oil bath for 1hour. Water was added and the product was extracted with ethyl acetate. The combined extracts were washed with saturated sodium chloride, dried over magnesium sulfate, filtered and concentrated to give 3-(5-phenylpyridin-3-yl)-3-[4-(7-[2-(tri-methylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]-pyrimidin-4-yl)-1 H-pyrazol-1-yl]propanenitrile (50 mg, 80%) as a crude prod- uct. MS (ES): 522 (M+1).
Step 2: 3-(5-Phenylpyridln-3-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 Hpyrazol-1 -yljpropane-nitrile trifluoroacetate [0469] Using a procedure analogous to that of Example 61 for the removal of the SEM protecting group, the title compound was isolated as an amorphous white solid (20 mg, 40 %). 1H NMR (400 MHz, DMSO): δ 9.15 (s, 1H), 8.85 (s, 1H), 8.80 (s, 1H), 8.65 (s, 1H), 8.45 (s, 1H), 8.22 (s,1H), 7.85 (b, 1H), 7.67 (m, 2H), 7.45(m 2 H), 7.43 (m, 1H), 7.15 (s, 1H), 6.25 (m 1H), 3.95 (dd, 1H), 3.80 (dd, 1H), 3.0 (m, 1H); MS (ES): 392.1 (M+1)
Example 486: 3-(5-Ethynylpyridin-3-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile trifluoroacetate [0470]
Step 1: 3-[4-(7-[2-(Trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-3-5-[(trimethylsi-lyl)ethynyl]pyridin-3-ylpropanenitrile [0471] To a solution of 3-(5-bromopyridin-3-yl)-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propanenitrile (from Example 429) (0.080 g, 0.00015 mol) in TEA (0.300 mL, 0.00215 mol) was degassed with nitrogen, and then copper(l) iodide (0.005 g, 0.00003 mol), (trimethylsilyl)acetylene, and bis(triphenyl-phosphine)palladium(ll)chloride were added. The reaction mixture was sealed in a tube and stirred at room temperature overnight. The resulting black solution was partitioned between water (10 mL) and ethyl ether. The organic layer was washed with saturated sodium chloride, dried over magnesium sulfate and concentrated in vacuo to give 3-[4-(7-[2-(tri-methylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-3-5-[(trimethylsilyl)ethynyl]pyridin-3-ylpro-panenitrile as a yellow oil (60 mg,72.6), LCMS (M+1)+:542).
Step2: 3-(5-Ethynylpyridin-3-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yljpropane-nitrile trifluoroacetate [0472] 3-[4-(7-[2-(Trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-3-5-[(trimethylsi-lyl)ethynyl]pyridin-3-ylpropanenitrile (0.050 g, 0.000092 mol) was dissolved in DCM (5.0 mL, 0.078 mol) and TFA (2.0 mL, 0.026 mol). The reaction mixture was stirred at room temperature, for 90 minutes and was concentrated in vacuo. The dry residue dissolved in methanol cooled in an ice bath and a solution of potassium hydroxide (0.482 g, 0.00859 mol) in methanol (10 mL, 0.2 mol) was added. The reaction solution was stirred for 30 min was concentrated and the crude product was purified by preparative HPLC eluting with a water: ACN gradient with 0.2% TFA, to give the title compound as a white amorphous solid (15 mg, 35.85%). LCMS (M+1)+:340, 1H NMR (400 MHz, DMSO-dg): δ 12.1 (bs, 1H), 9.02(s, 1H), 8.80(s, 1H), 8.70(m, 2H), 8.48(s, 1H), 8.00(s, 1H), 7.80(d, 1H), 7.15(d, 1H), 6.20(m, 1H), 4.82(s, 1H), 3.90(m, 1 H), 3.70(m, 1H).
Example 488: 3-[5-(Phenylthio)pyridin-3-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propaneni-trile trifluoroacetate [0473]
Step 1: 3-[5-(Phenylthio)pyridin-3-yl]-3-[4-(7-[2-(trímethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]-pyrimidin-4-yl)-1H-pyra-zol-1 -yljpropanenitrile [0474] To the 3-(5-bromopyridin-3-yl)-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]-pyrimidin-4-yl)-1H-pyrazol-1-yljpropanenitrile (0.130 g, 0.000248 mol) from Example 429 Step 2, in dry 1,4-dioxane (1.60 mL, 0.0205 mol) was added DIPEA (0.085 mL, 0.00049 mol). The solution was degassed with nitrogen, followed by addition of (9,9-dimethyl-9H-xanthene-4,5-diyl)bis(diphenylphosphine) (0.007 g, 0.00001 mol), bis(dibenzylideneacetone)palladium(0) (0.0036 g, 0.0000062 mol), and benzenethiol (0.025 mL, 0.00025 mol). Again the solution was purged with nitrogen. The reaction mixture in a sealed tube was heated to reflux for 3h. The reaction mixture was diluted with ethyl acetate, washed with water (2X), brine (1X), dried over magnesium sulfate, filtered, and the solvent was evaporated in vacuo. The crude product was triturated with hexane-ethyl acetate 9:1 to yield 3-[5-(phenylthio)pyridin-3-yl]-3-[4-(7-[2-(trimeth-ylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propanenitrile (110 mg, 80%). LC/MS (M+H)+: m/z = 554.2.
Step 2: 3-[5-(Phenylthio)pyridin-3-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]-propanenitrile trifiuoroace-tate [0475] The 3-[5-(phenylthio)pyridin-3-yl]-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile (0.110 g, 0.000199 mol) was dissolved in DCM (5.0 mL, 0.078 mol) and TFA (2.0 mL, 0.026 mol), and the mixture was stirred at room temperature for 1 hour. The solvent was removed in vacuo, and the resulting residue was dissolved in methanol (5.0 mL, 0.12 mol), and ethylenediamine(0.1 mL, 0.002 mol) was added. This reaction mixture was stirred at room temperature overnight. The mixture was concentrated in vacuo, and the crude product was purified by LCMS (pH=2) to yield the title compound as an amorphous solid (62 mg, 58.07%). 1H NMR(400 MHz, DMSO): δ. 12.80 (s), 9.10 (s) 8.87(d), 8.60 (s), 8.50 (s), 8.43 (s), 7.82 (s), 7.78 (m), 7.39 (m), 7.25 (m), 7.18 (d), 6.20 (m), 3.84 (m), 3.70 (m). LC/MS (M+H)+: m/z = 424.15
Example 491: 3-(5-Morpholin-4-ylpyridin-3-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propaneni-trile [0476]
Step 1: 4-(5-Bromopyridin-3-yl)morpholine [0477] To a solution of [3,5-dibromopyridine (1000 mg, 0.004 mol) in 1,4-dioxane (8 mL, 0.1 mol), morpholine (400 mg, 0.004 mol) and sodium ferf-butoxide (400 mg, 0.004 mol) were added. The reaction was bubbled with nitrogen. Tetrakis(triphenyiphosphine)pailadium(0) (200 mg, 0.0002 mol) was added and nitrogen was bubbled through for couple of minutes. The mixture was heated at 80 °C overnight. The reaction was allowed to cool to rt and was then partitioned between water and ethyl acetate. The organic layer was washed with saturated sodium chloride, dried over magnesium sulfate, filtered and concentrated to give a crude residue. The crude product was purified by FCC on silica gel eluting with 1:1, EtOAC:Hexane gave to give 4-(5-bromopyridin-3-yl)morpholine as a viscous oil (400 mg, 40 %). 1H NMR (400 MHz, CDCI3): δ. 8.2 (s, 1H), 8.1 (s, 1H), 7.2 (s, 1H), 3.8 (m, 4H), 3.2 (m, 4H).
Step 2: 5-Morpholln-4-ylnicotinaldehyde [0478] A solution of 4-(5-bromopyridin-3-yl)morpholine (100 mg, 0.0004 mol) in ether (2 mL, 0.02 mol) cooled at -78 °C was treated with 2.5 M n-butyllithium in hexane (0.2 mL) and was stirred for 1 h. To this mixture was added DMF (0.5 mL, 0.006 mol) dropwise. The reaction was quenched with water and extracted with ethyl acetate. The combined organic layers were washed with saturated sodium chloride, dried over magnesium sulfate, filtered and concentrated to give 5-morpholin-4-ylnicotinaldehyde (70 mg, 90%) as a crude product. 1H NMR (400 MHz, CDCI3): δ 10.1 (s, 1H), 8.0 (s, 2H), 7.6 (s, 1H), 3.8 (m, 4H), 3.2 (m, 4H).
Step 3: (2E)-3-(5-Morpholln-4-ylpyrldln-3-yl)acrylonitrile [0479] To a solution of diethyl cyanomethylphosphonate (70 mg, 0.0004 mol) in THF (2 mL, 0.02 mol) cooled at 0 °C was added 1.0 M potassium ferf-butoxide in THF (0.50 mL) dropwise. The cold bath was removed and the reaction was warmed to room temperature over 30 minutes. The reaction was cooled to 0 °C and a solution of 5-morpholin-4-ylnico-tinaldehyde (70 mg, 0.0004 mol) in THF (2 mL, 0.02 mol) was added dropwise. The reaction was stirred at room temperature for 4 h, quenched with water and extracted with ethyl acetate. The combined organic layers were washed with saturated sodium chloride, dried over magnesium sulfate, filtered and concentrated to give (2E)-3-(5-morpholin-4-ylpyridin-3-yl)acrylonitrile (75 mg, 100%) as a mixture of isomers; LC/MS: 216 (M+1).
Step 4: 3-(5-Morpholin-4-ylpyridin-3-yl)-3-[4-(7-[2-(trimethylsHyl)ethoxy]methyl-7H-pyrrolo[2,3-d]-pyrimidin-4-yl)-1 H-pyrazol-1-yl]propanenitrile [0480] To a solution of 4-(1 H-pyrazol-4-yl)-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]-pyrimidine (120 mg, 0.00038 mol) in ACN (10 mL, 0.2 mol) and (2E)-3-(5-morpholin-4-ylpyridin-3-yl)acrylonitrile (70 mg, 0.0003 mol) ( mixture of isomers), DBU (50 |xL, 0.0003 mol) was added and the resulting mixture was stirred overnight. The mixture was partitioned between water and ethyl acetate. The combined organic layers were washed with saturated sodium chloride, dried over magnesium sulfate, filtered and concentrated to give 3-(5-morpholin-4-ylpyridin-3-yl)-3-[4-(7-[2-(trimethylsi-lyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile (200 mg, 100%) as a crude product; L/MS = 531 (M+1).
Step 5: 3-(S-Morpholin-4-ylpyridin-3-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propanenitrile [0481] Using a procedure analogous to Example 61 for the removal of the SEM protecting the title compound was isolated as an amorphous white solid (18 mg, 50%). 1H NMR (400 MHz, DMSO): δ 8.8 (s, 1H), 8.6 (s, 1H), 8.4 (s, 1H), 8.2 (s, 1H), 8.0 (s, 1H), 7.6 (d, 1H), 7.4 (m, 1H), 6.9 (d, 1H), 6(m, 1H), 3.8 (dd, 1H), 3.7(m, 4H), 3.6 (dd, 1H), 3.1 (m, 4 H); LC/MS: 401(M+1).
Example 496: 3-[5-(Phenylsulfinyl)pyridin-3-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]pro-panenitrile, and Example 497: 3-[5-(Phenylsulfonyl)pyridin-3-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-.4-yl)-1H-pyra-zol-1-yl]propanenitrile [0482]
[0483] To the solution of 3-[5-(phenylthio)pyridin-3-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin4-yl)-1 H-pyrazol-1-yl]propanen-itrile trifluoroacetate (0.050 g, 0.000093 mol) from Example 488, Step 2, in THF (1.0 mL, 0.012 mol) was added MCPBA (0.022 g, 0.00013 mol) (0.031g of 77% in water), in a water ice bath. The reaction mixture was stirred for 1h at room temperature. The crude products were purified by LCMS (pH=10). Two peaks were collected : # 1 - to yield 3-[5-(phenylsulfinyl)pyridin-3-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propanenitrile (8 mg, 19.57%). 1H NMR (400 MHz, DMSO): δ. 12.1 (s), 8.89 (d), 8.80 (d), 8.70 (s), 8.62 (s), 8.40 (s), 8.19 (s), 7.70 (m), 7.58 (s), 7.42 (m), 6.90 (s), 6.20 (m), 3.82 (m), 3.65 (m). LC/MS (M+H)+: m/z = 440.0 # 2 - to yield 3-[S-(phenylsulfonyl)pyridin-3-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile (21 mg, 50%). 1H NMR (400 MHz, DMSO): δ. 12.1 (s), 9.10 (s), 8.86 (m), 8.61 (s), 8.40 (m), 7.98 (m), 7.62 (m), 7.58 (m), 6.90 (s), 6.20 (m), 3.82 (m), 3.65 (m). LC/MS (M+H)+: m/z = 456.0
Example 498: 3-[4-(7H-Pyrrolo [2,3-d]pyrimidin-4-y 1)-IH-pyrazoi-l-yl]pentan-l-ol [0484]
Step 1: 3-[4-(7-[2-(Trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]pentanal [0485] To a solution of 4-(1 H-pyrazol-4-yl)-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]-pyrimidine (100 mg, 0.0003 mol) in ACN (2 mL, 0.04 mol) and DBU (50 μί, 0.0003 mol), the (2E)-pent-2-enal (4.0E1 mg, 0.00048 mol) in 1ml ACN was added drop wise. The reaction was stirred for 1 h, and then water was added and the resulting mixture extracted with ethyl acetate. The combined organic layers were washed with saturated sodium chloride, dried over magnesium sulfate, filtered and concentrated to give the crude as the hydrated product form. LC/MS (M+H)+: m/z = 400.
Step 2: 3-[4-(7-[2-(Trimethylsllyl)ethoxy]methyl-7Hpyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1 -yl]pentan-1-ol [0486] A mixture of [3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]pentanal (50 mg, 0.0001 mol) in methanol (2 mL, 0.05 mol) was treated with sodium tetrahydroborate (8 mg, 0.0002 mol). The mixture was stirred at room temperature for 1 h, and then water was added and the product was extracted with ethyl acetate. The combined organic layers were washed with saturated sodium chloride, dried over magnesium sulfate, filtered and concentrated to give the desired product as an oil. LC/MS (N4+H)+: m/z = 402.
Step 3: [0487] Using a procedure analogous to Example 61 for the removal of the SEM protecting group the title compound was isolated as an amorphous white solid (6 mg, 20 %). 1H NMR (400 MHz, DMSO): δ 8.65 (d, 1H), 8.60 (d, 1H), 7.55 (s, 1H), 6.95 (s, 1H), 4.50 (b, 1 H),4.4(m, 1H), 3.4 (m, 1H), 3.2 (m, 1H), 2.1 (m, 1H), 1.8-2.0 (m, 3H), 0.7(t, 3H); LC/MS (M+H)+: m/z = 272.
Example 499: Methyl 3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]pentyl carbonate [0488]
Step 1 : Methyl 3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]pentyl carbonate [0489] To a solution of [3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]pen-tan-1-ol (50 mg, 0.0001 mol) from Example 498 Step 2 in pyridine (1 mL, 0.01 mol), methyl chloroformate (30 μΙ-, 0.0003 mol) was added. The reaction was stirred for 3h, water was added and the product was extracted with ethyl acetate. The combined organic layers were washed 1N HCI, brine, dried over magnesium sulfate, filtered and concentrated to give methyl 3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]pentyl carbonate as a semisolid residue (30 mg, 50%). LC/MS (M+H)+: m/z = 460.
Step 2: [0490] Using a procedure analogous to Example 61 for the removal of the SEM protecting the title compound was isolated as an amorphous white solid (8 mg, 20 %). 1H NMR (400 MHz, DMSO): δ.12.0 (b, 1H), 8.65 (d, 1H), 8.35 (s, 1H), 7.65 (b, 1H), 7.600 (s, 1H), 7.0 (s, 1H),4.4 (m, 1H),4.0 (m, 1H), 3.8 (m, 1H), 3.6 (s, 3H), 2.1 (m, 1H), 2.2 (m, 1H), 1.95 (m, 2H), 0.75 (t, 3H); LC/MS (M+H)+: m/z = 330.
Example 500(a): (1E)-3-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]pentanal oxime [0491]
Step 1: ( 1 E)-3-[4-(7-[2-(Trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazot-1 -yljpentanal oxime [0492] To a solution of 3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]pen-tanai (60 mg, 0.0002 mol)from Example 498, Step2 in methanol (2 mL, 0.05 mol) was added hydroxylamine hydrochloride (16 mg, 0.00022 mol) and potassium bicarbonate (22 mg, 0.00022 mol). The reaction was stirred at room temperature for 2h, water was added and the product was extracted with ethyl acetate. The combined extracts were washed with saturated sodium chloride, dried over magnesium sulfate, filtered and concentrated to give (1 E)-3-[4-(7-[2-(trimethyl-silyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]pentanal oxime as a semisolid residue (50 mg, 80%). LC/MS (M+H)+: m/z = 415.
Step 2: [0493] Using a procedure analogous to Example 61 for the removal of the SEM protecting the title compound was isolated as an amorphous white solid. 1H NMR (400 MHz, DMSO): δ 12.0 (b, 1H), 8.6 (m, 2H), 8.2 (m, 1H), 7.5 (d, 1H), 7.1 and 6.5 (t, 1H), 4.6 (m, 1H), 4.4 (m, 1H), 2.6-2.8 (m, 2H), 1.8 (m, 2H), 0.65 (t, 3H); LC/MS (M+H)+: m/z = 285.
Example 501(a): (1E)-3-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]pentanal O-methyloxime, and Example 502(a): (1Z)-3-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]pentanai O-methyloxime [0494]
Step 1: ( 1E)-3-[4-(7-[2-(Trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]pentanal O-meth-yloxime and ( 1Z) -3-(4-(7-(2-(Trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazot-1 -yt]pentanat O-methyloxime [0495] To a solution of 3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]pen-tanai (70 mg, 0.0002 mol) in methanol (2 mL, 0.05 mol) was added methoxylamine hydrochloride (19 mg, 0.00022 mol) and potassium bicarbonate (22 mg, 0.00022 mol). The reaction was stirred at room temperature for 2h, water was added and the product was extracted with ethyl acetate. The combined extracts were washed with saturated sodium chloride, dried over magnesium sulfate, was filtered and was concentrated to give 3-[4-(7-[2-(trimethylsilyl)-ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yljpentanal .O-methyloxime as a mixture of isomers (70 mg, 90%) crude product. LC/MS (M+H)+: m/z = 429.
Step 2: [0496] Using a procedure analogous to Example 61 for the removal of the SEM protecting the title compound was isolated as an amorphous white solid (4 mg, 25 %). Isomer 1, 1H NMR (400 MHz, DMSO): δ. 8.7 (s, 2H), 8.3 (s, 1H), 7.6 (s, 1H), 7.3 (t, 1H), 7.0 (s, 1H), 4.6(m, 1H), 3.3 (s, 3H), 2.8 (m, 2H), 1.9 (m, 2H), 0.8 (t, 3H); LC/S (M+H)+: m/z = 299.Isomer 2 (3 mg, 22%), 1H NMR (400 MHz, DMSO): δ 8.7 (s, 2H), 8.3 (s, 1H), 7.6 (s, 1H), 7.0 (s, 1H), 6.7 (t, 1H), 4.5(m, 1H), 3.3 (s, 3H), 2.8-3.0 (m, 2H), 1.9 (m, 2H), 0.8 (t, 3H); LC/MS (M+H)+: m/z = 299.
Example 503: 4-[1-(4,4-Dibromo-1-ethylbut-3-en-1-yl)-1H-pyrazol-4-yl]-7H-pyrrolo[2,3-d]-pyrimidine trifluoroac-etate [0497]
Step 1:4-[1 -(4,4-Dibromo-1 -ethylbut-3-en-1 -yl)-1 Hpyrazol-4-yl]-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyri-midine [0498] To a solution of 3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]pen-tanai (300 mg, 0.0008 mol) in DCM (4 mL, 0.06 mol) cooled at 0 °C, triphenylphosphine (800 mg, 0.003 mol) and carbon tetrabromide (500 mg, 0.002 mol) were added. The reaction was stirred at 0 °C for 10 min, waterwas added and extracted with ethyl acetate. The combined organic extracts were washed with saturated sodium chloride, dried over magnesium sulfate, filtered and concentrated. The crude product was purified by prep LC-MS (ACN, water, NH4OH) to give 4-[1-(4,4-dibromo-1-ethylbut-3-en-1-yl)-7H-pyrazol-4-yl]-7-[2-(trimethylsilyl)-ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidine as an amorphous solid (50 mg, 10%). 1H NMR (400 MHz, CDCI3): δ. 8.9 (s, 2H), 8.4 (s, 1H), 8.3 (s, 1H), 7.4 (m, 1H), 7.3 (s, 1H), 6.9 (m, 1H), 6.4 (m, 1H), 5.7 (s, 2H), 4.2 (m, 1H), 3.6 (m, 2H), 2.8 (m, 2H), 2.1 (m, 1H), 2.0 (m, 1H), 1.0 (m, 5H), LC/MS (M+H)+: m/z = 556
Step 2: [0499] Using a procedure analogous to Example 61 for the removal of the SEM protecting the title compound was isolated as an amorphous white solid (8 mg, 40 %). 1H NMR (400 MHz, DMSO): δ. 8.8 (s, 2H), 8.4 (s, 1H), 7.7 (b, 1H), 7.2 (b, 1H), 6.5 (t, 1H), 4.4 (m, 1H), 2.6 (m, 2H), 1.8 (m, 2H), 0.8 (t, 3H); LC/MS (M+H)+: m/z =: 426.
Example 506: 4-[1-(1-Ethylbut-3-yn-1-yl)-1H-pyrazol-4-yl]-7H-pyrrolo[2,3-d]pyrimidine trifluoroacetate [0500]
Step 1: 4-[1-( 1-Ethylbut-3-yn-1-yl)-1 H-pyrazol-4-yl]-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidine [0501] A solution of 4-(1-(4,4-dibromo-1-ethylbut-3-en-1-yl)-1 H-pyrazol-4-yl]-7-[2-(trimethylsilyl)-ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidine (20 mg, 0.00004 mol) (from Example 503 Step 1) in THF (1 mL, 0.01 mol) at-78 °C was treated with 2.5 M n-butyllithium in hexane (0.032 mL). The mixture was stirred at -78 °C for 1 h and then at room temperature for 1 h. The reaction was quenched with water (1 mL, 0.06 mol) and 1N HCI. The reaction was partitioned between water and ethyl acetate. The organic extract was washed with saturated sodium chloride, dried over magnesium sulfate, filtered and concentrated to give 4-[1-(1-ethylbut-3-yn-1-yl)-1 H-pyrazol-4-yl]-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidine as a semisolid (12 mg, 80%). LC/MS (M+H)+: m/z = 396.
Step 2: [0502] Using a procedure analogous to Example 61 for the removal of the SEM protecting the title compound was isolated as an amorphous white solid (4 mg, 30 %). 1H NMR (400 MHz, DMSO): δ 12.2 (b, 1H), 8.8 (s, 2H), 8.4 (s, 1H), 7.6 (s, 1H), 7.1 (s, 1H), 4.4 (m, 1H), 2.8 (m, 3H), 1.9 (m, 2H), 0.8 (t, 3H); LC/MS(M+H)+: m/z = 266.
Example 516: (R)-3-(3-(Ethylsulfonyl)phenyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propaneni-trile, and (S)-3-[3-(Ethylsulfonyl)phenyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propanenitrile [0503]
Step 1: 1-Bromo-3-(ethylthio)benzene [0504] lodoethane (0.46 mL, 0.0058 mol) was added to a suspension of 3-bromothiophenol (0.50 ml_, 0.0048 mol), ACN (7.11 mL, 0.136 mol) and potassium carbonate (2.0 g, 0.014 mol). The reaction was stirred for2 h at rt, was diluted with ethyl acetate and filtered to remove the solids. The reaction was concentrated in vacuo to give 1-bromo-3-(ethyl-thio)benzene as a colorless oil 1.0 gm, 100%
Step 2: 1-Bromo-3-(ethylsulfonyl)benzene [0505] The MCPBA (2.37 g, 10.6 mmol) was added to a solution of 1-bromo-3-(ethylthio)benzene (1.00 g, 4.80 mmol) in DCM (10 ml, 156 mmol) cooled to 0 °C. The reaction was stirred for 1 h and then was diluted with water and extracted with ethyl acetate three times. The combined organic layers were dried with magnesium sulfate, filtered, and concentrated in vacuo. The resulting crude residue was purified by flash column chromatography with a hexane: ethyl acetate gradient to give 1-bromo-3-(ethylsulfonyl)benzene as a colorless oil 1.1 gm 92%, 1H NMR (300 MHz, CDCI3): 58.09(m, 1H), 7.8S(d,1 H), 7.78(d, 1H) 7.45(t,1H), 3.14(q, 2H), 1.25(t, 3H).
Step 3: (2E & Z)-3-[3-(Ethylsulfonyl)phenyl]acrylonitrile [0506] 1-Bromo-3-(ethylsulfonyl)benzene (1.3 g, 0.0052 mol) was dissolved in the DMF (15.0 mL, 0.194 mol) and 2-propenenitrile (0.68 mL, 0.010 mol), TEA(1.4 mL, 0.010 mol) and triphenylphosphine (0.23 g, 0.00089 mol) were added. The resulting solution was degassed with nitrogen, and palladium acetate (0.07 g, 0.0003 mol) was added. Again the reaction was degassed with nitrogen and then heated to 110 °C in a sealed tube for 8 hrs. The reaction was complete by HPLC, and was then allowed to cool to rt and then partitioned between ethyl acetate and water. The organic layer was washed with brine, dried over magnesium sulfate and concentrated. The product was purified by FCC on silica gel eluting with a hexane; ethyl acetate gradient to give (2E&Z)-3-[3-(ethylsulfonyl)phenyljacrylonitrile as an amber oil (1.1 gm, 92%) LC/MS (M+H)+: m/z = 222.
Step 4: 3-[3-(Ethylsulfonyl)phenyl]-3-[4-(7-[2-(trimethylsHyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1 -yljpropanenitrile [0507] The (2E&Z)-3-[3-(ethylsulfonyl)phenyl]acrylonitrile (1.0 g, 0.0045 mol) was combined with 4-(1 H-pyrazol-4-yl)-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidine (1.3 g, 0.0041 mol) and DBU (0.61 mL, 0.0041 mol) in ACN (10.0 mL, 0.191 mol) under nitrogen at rt. The reaction was stirred at rt for 24 h. This was partitioned between ethyl acetate and water, and 0.1 N HCI was added to adjust the pH to 7. The combined organic extracts were washed with brine, dried over magnesium sulfate and concentrated to give a crude oil. The product was purified by FCC on silica gel eluting with a hexane: ethyl acetate gradient to give 3-[3-(ethylsulfonyl)phenyl]-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile as an oil (1.5 gm, 68%). LC/MS (M+H)+: m/z = 537. The oil was a racimate, which was separated by chiral column chromatography (Chiracei OD-H, eluting with ethanol: methanol: hexane 30:30:40, Rt 13.2 and 17.1 minutes) to give the two enantiomers, each as a glass (0.51 gm) LC/MS (M+H)+: m/z = 537, 1H NMR (300 MHz, CDCI3): δ 8.89(s, 1H), 8.45(s, 1H), 8.35(s,1H), 8.09(s, 1h), 8.05(d, 1H), 7.75(d, 1H), 7.71 (t, 1H), 7.45(d, 1H), 6.83(d, 1H), 5.85(t, 1H), 5.75(s, 2H), 3.78-3.42(m, 4H), 3.18(m, 2H), 1.35(t, 3H), 0.97(t, 2H), 0.05(s, 9H).
Step 5: [0508] Using a procedure analogous to Example 61 for the removal of the SEM protecting group the title compounds were prepared to give isomer #1 as an amorphous white solid (300 mg, 80 %). 1H NMR (400 MHz, DMSO): δ 9.1 (s, 1H), 8.8 (s, 1H), 8.5 (s, 1H), 8.0 (s, I H), 7.6-7.9 (m, 4H), 7.1 (s, 1H), 6.3 (m, 1H), 3.9 (m, 1H), 3.7 (m, 1H) 3.2 (q, 2H), 1.0 (t, 3H); MS(ES) (M+H)+: m/z = 407.
[0509] Using a procedure analogous to Example 61 for the removal of the SEM protecting group the title compounds were prepared to give isomer #2 as an amorphous white solid (300 mg, 80 %). 1H NMP (400 MHz, DMSO): δ. 9.1 (s, 1H), 8.8 (s, 1H), 8.5 (s, 1H), 8.0 (s, 1H), 7.6-7.9 (m, 4H), 7.1 (s, 1H), 6.3 (m, 1H), 3.9 (m, 1H), 3.7 (m, 1H) 3.2 (q, 2H), 1.0 (t, 3H); MS(ES) (M+H)+: m/z = 407.
Example 526: 4-[1-(1-Ethylbut-3-en-1-yl)-1H-pyrazol-4-yl]-7H-pyrrolo[2,3-d]pyrimidine [0510]
Step 1: 4-[1-( 1-Ethylbut-3-en-1-yl)-1 H-pyrazol-4-yl]-7-{2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo-[2,3-d]pyrimidine [0511] To an ice cooled solution of methyl triphenylphosphonium bromide (100 mg, 0.0004 mol) in THF (2 mL, 0.02 mol) was added 0.5 M potassium bis(trimethylsilyl)amide in toluene (0.8 mL). The mixture was stirred for 1 h at 0 °C ice bath, and was then cooled to -78 °C and treated with 3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]pentanal (80 mg, 0.0002 mol) (from Example 498). The reaction was stirred at-78 °C and gradually was warmed to room temperature overnight. The reaction was partitioned between water and ethyl acetate. The organic layer was washed with saturated sodium chloride, dried over magnesium sulfate, filtered and concentrated to give 4-[1-(1-ethylbut-3-en-1-yl)-1H-pyrazol-4-yl]-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidine 150 mg as a crude product. LC/MS = 398 (M+1).
Step 2: 4-[ 1-( 1-Ethylbut-3-en-1-yl)-1H-pyrazol-4-yl]-7H-pyrrolo[2,3-d]pyrimidine [0512] Using a procedure analogous to Example 61 for the removal of the SEM protecting group the title compound was isolated as an amorphous white solid (25 mg, 1%). 1H NMR (400 MHz, DMSO): δ. 8.6 (s, 2H), 8.2 (s, 1H), 7.4 (s, 1H), 6.9 (s, 1H), 5.8 (m, 1H), 5.0 (dd, 2H), 4.2 (m, 1H), 2.4-2.6 (m, 2H), 1.7-1.9 (m, 2H), 0.6 (t, 3H); LC/MS: 268 (M+1).
Example 500: (3R)- and (3S)-4,4,4-Trifluoro-3-[3-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrrol-1-yl]butanenitrile [0513]
Step 1. 4-Chloro-7-(diethoxymethyl)-7H-pyrrolo[2,3-d]pyrimidine [0514] A mixture of 4-chloropyrrolo[2,3-d]pyrimidine (2.00 g, 0.0130 mol) and ethyl orthoformate (25 mL, 0.15 mol) was heated to reflux for 2 hours. The solvent was evaporated, and the residue was purified by flash column chromatog- raphy (eluting with ethyl acetate/hexanes) to yield the desired product (1.13 g, 34%). 1H NMR (400 MHz, CDCI3): δ 8.63 (s, 1H), 7.58 (d, 1H), 6.71 (s, 1H), 6.65 (d, 1H), 3.77-3.67 (m, 2H), 3.58-3.49 (m, 2H), 1.23 (t, 3H), 1.23 (t, 3H).
Step2. 7-(Diethoxymethyl)-4-(1Hpyrrol-3-yl)-7H-pyrrolo[2,3-d]pyrimidine [0515] To a degassed solution of 4-chloro-7-(diethoxymethyl)-7H-pyrrolo[2,3-d]pyrimidine (1.13 g, 0.00442 mol) and 1-(triisopropylsilyl)-3-boronic acid (1.00 g, 0.00374 mol) and sodium carbonate (0.396 g, 0.00374 mol) in 1,2-dimethox-yethane (15 mL) and water (3 mL) was added tetrakis(triphenylphosphine)palladium(0) (0.22 g, 0.00019 mol). This mixture was stirred at ambient temperature for 2 hours, and then was heated to reflux for 4 hours. The mixture was then cooled, concentrated, and purified by flash column chromatography (eluting with ethyl acetate/hexanes) to afford a residue as an oil. ACN was added to the residue, and the product which precipitated was filtered off and washed with a small quantity of ACN (165 mg, 13%). 1H NMR (400 MHz, D6-dmso): δ 11.44 (brs, 1H), 8.66 (s, 1H), 7.80-7.78 (m, 1H), 7.58 (d, 1H), 7.03 (d, 1H), 6.94 (dd, 1H), 6.90 (dd, 1H), 6.75 (s, 1H), 3.74-3.65 (m, 2H), 3.59-3.50 (m, 2H), 1.15 (t, 6H); MS(ES): M+H = 287.
Step 3.
[0516] To a solution of 7-(diethoxymethyl)-4-(1H-pyrrol-3-yl)-7H-pyrrolo[2,3-d]pyrimidine (0.125 g, 0.436 mmol) and 4,4,4-trifluorobut-2-enenitrile (0.0476 mL, 0.480 mmol) in ACN (1 mL) was added DBU (0.0653 mL, 0.436 mmol). TFA (0.5 mL) was added and the mixture was stirred for 1 hour. The TFA and solvent was removed in vacuo. The residue was purified by preparative-HPLC/MS (C-18 eluting with a gradient of H20/ACN containing 0.15% NH4OH) to afford the product (102 mg, 76%). Where desired, the enantiomers were separated in substantially pure form by chiral HPLC (AD-H, 20% EtOH/Hexane). 1H NMR (300 MHz, D6-dmso): δ 12.05 (brs, 1H), 8.65 (s, 1H), 8.04 (s, 1H), 7.56 (dd, 1H), 7.21 (t, 1H), 7.02 (dd, 1H), 6.93 (dd, 1H), 5.89-5.74 (m, 1H), 3.95 (dd, 1H), 3.66 (dd, 1H); MS(ES): M+H = 306.
[0517] The analog in Table 12 was prepared in racemic form according to the same procedure, using a different conjugate acceptor and with the exception that in the conjugate addition in Step 3, the reaction was carried out at 40 °C for 3 days.
[0518] The following compounds in Table 13 were prepared as indicated in the column labeled "Method of Prep." and the details of certain exemplary synthetic procedures are provided following Table 13.
Example 649: 3-[3-(Morpholin-4-ylsulfonyl)phenyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]pro-panenitrile [0519]
Step 1: 4-[(3-Bromophenyl)sulfonyl]morpholine [0520] Morpholine (0.19 mL, 0.0022 mol) in 1.0 ml of THF was added dropwise to a solution of 3-bromobenzenesulfonyl chloride (0.3 mL, 0.002 mol) and TEA (0.30 mL, 0.0022 mol) in dry 4.0 mL of THF cooled in an ice bath. The reaction mixture was stirred overnight at room temperature and was then partitioned between 0.05N HCI and ethyl acetate. The organic layer was washed with water (2X), and brine (IX), and was then dried over anhydrous magnesium sulfate, filtered and then was concentrated in vacuo to give 4-[(3-bromophenyl)sulfonyl]morpholine as a white crystalline product (470 mg, 78%). LCMS (M+H)+: m/z = 306, 308.
Step 2: (2E&Z)-3-[3-(Morpholin-4-ylsulfonyl)phenyl]acrylonitrile [0521] The 4-[(3-bromophenyl)sulfonyl]morpholine (0.250 g, 0.000816 mol) was dissolved in dry DMF (2.5 mL, 0.032 mol) and the mixture was degassed using a stream of nitrogen. To this mixture was added TEA (0.23 mL, 0.0016 mol), 2-propenenitrile (0.11 mL, 0.0016 mol), palladium acetate (0.011 g, 0.000049 mol), and triphenylphosphine (0.0364 g, 0.000139 mol) and again the mixture was degassed with nitrogen. The reaction mixture in a sealed tube was heated at 110 °C for 16 hours. The reaction mixture, after cooling to room temperature, was partitioned between 0.05N HCI and ethyl acetate. The organic layer was washed with water (2X), and brine (1X), dried over anhydrous magnesium sulfate, filtered, and concentrated in vacuo, to give (2E&Z)-3-[3-(morpholin-4-yl-sulfonyl)phenyl]acrylonitrile as an oil (0.240gm, 85%) which was a mixture of cis and trans isomers. LCMS (M+H)+: m/z = 279.
Step 3: 3-[3-(Morpholin-4-ylsulfonyl)phenyl]-3-[4-(7-[2-(trimethylsHyl)ethoxy]methyl-7H-pyrrolo-[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile [0522] To a mixture of 4-(1 H-pyrazol-4-yl)-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]-pyrimidine (0.100 g, 0.000317 mol) and (2E&Z)-3-[3-(morpholin-4-ylsulfonyl)phenyl]acrylonitrile (0.097 g, 0.00035 mol) in dry ACN (2.0 mL, 0.038 mol) was added DBU (0.095 mL, 0.00063 mol), and the resulting mixture was stirred at room temperature overnight. The reaction mixture was then diluted with water and extracted with ethyl acetate. The combined organic phase was washed with water (2X), and brine (IX), dried over magnesium sulfate, filtered and then concentrated in vacuo to give the crude product. The crude product was purified by silica gel flash column chromatography using ethyl acetate-hexanes (6:4) as an eluent to give 3-[3-(morpholin-4-ylsulfonyl)phenyl]-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile as a viscous oil (62 mg, 32.94%). LCMS (M+H)+: m/z = 594
Step 4: [0523] Using a procedure analogous to Example 61 for the removal of the SEM protecting the title compound was isolated as an amorphous white solid (30 mg, 63.84%. LCMS (M+H)+: m/z = 464.1H NMR (400 MHz, DMSO-d6): δ 8.88 (s), 8.62 (s), 8.1(s), 7.78(m), 7.70(m), 7.58(m), 6.95(m), 6.20(m), 3.84(m), 3.70(m),3.45(m), 2.78(m).
Example 679: cis-4-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohexyl-acetonitrile [0524]
Step 1: 4-(Hydroxymethyl)cyclohexanol.
[0525] Ethyl 4-oxocyclohexanecarboxylate (2.0 g, 0.012 mol) was dissolved in ether (20.0 mL) and was then cooled at 0 °C Into the mixture was added 1 M lithium tetrahydroaluminate in ether (20 mL) and the resulting mixture was stirred at 0 °C for 2 hours. The reaction was quenched with water (2 mL) and 1 N NaOH (2 mL) and ether was added (100 mL). The precipitated solids were filtered off and the residue was used in the next reaction. 1H NMR(CDCI3):5 4.02 and 3.75 (m, 1H), 3.45-3.61 (m, 2H), 2.02 (m, 2H), 1.84 (m, 1H), 1.52-1.80 (m, 2H), 1.44 (m, 1H), 1,32 (m, 2H), 1.03 (m, 1H).
Step 2: 4-[(Trityloxy)methyl]cyclohexanol.
[0526] 4-(Hdroxymethyl)cyclohexanol (2.0 g, 0.015 mol) was dissolved in pyridine (15.0 mL) and the mixture was cooled to 0 °C. To the reaction was added triphenylmethyl chloride (4.7 g, 0.017 mol) and the resulting mixture was stirred at 0 °C for 2 hours and at 25 °C for 16 hours. The reaction was then concentrated using a rotory evaporator, and the concentrate was extracted with ethyl acetate. The organic extracts were washed with water, saturated NaCI, dried (MgS04) and then concentrated in vacuo. The reaction was chromatographed on silica gel using 30% EtOAc/hexanes to give the cis isomer (0.74 g) 1H NMR(CDCI3):5 7.52 (m, 6H), 7.27 (m, 9H), 3.98 (m, 1H), 2.93 (m, 2H), 1.21-1.68 (m, 9H); and the trans isomer (2.72 g) 1H NMR(CDCI3):ô 7.44 (m, 6H), 7.20-7.31 (m, 9H), 3.54 (m, 1H), 2.88 (m, 2H), 1.98 (m,2H), 1.88 (m,2H), 1.60 (m, 1H), 0.99-1.37 (m, 4H).
Step 3: trans-4-[(Trityloxy)methyl]cyclohexyl methanesulfonate.
[0527] trans-4-[(Trityloxy)methyl]cyclohexanol (2.72 g, 0.00730 mol) was dissolved in chloroform (30.0 mL) and the mixture was cooled at 0 °C To this mixture was added TEA (1.4 mL, 0.010 mol) and methanesulfonyl chloride (0.68 mL, 0.0088 mol) and the resulting mixture was stirred at 0 °C for 2 hours The reaction was then extracted with ethyl acetate and the organic extracts were washed with water, saturated NaCI, dried (MgS04) and the concentrated in vacuo. 1H NMR (CDCI3):5 7.43 (m, 6H), 7.20-7.31 (m, 9H), 4.57 (m, 1H), 3.00 (m, 3H), 2.90 (m, 2H), 2.16 (m, 2H), 1.93 (m, 2H), 1.09-1.60 (m, 5H).
Step 4: 7-[2-(Trimethylsilyl)ethoxy]methyl-4-(1-cis-4-[(trityloxy)methyl]cyclohexyl-1H-pyrazol-4-yl)-7H-pyrrolo[2,3-d]py-rimidine.
[0528] 4-(1H-Pyrazol-4-yl)-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidine (1.5 g, 0.0048 mol) was mixed with sodium hydride (0.34 g, 0.0086 mol) and trans-4-[(trityloxy)methyl]cyclohexyl methanesulfonate (3.00 g, 0.00666 mol) and the mixture was cooled to -78 °C. To this mixture was added DMF (8.3 mL) and the mixture was allowed to warm to 25 °C and was stirred for 20 minutes. The warmed mixture was stirred at 55°C for 48 hours. The reaction was extracted with ethyl acetate and the organic extracts were washed with water, saturated NaCI, dried (MgS04) and then concentrated in vacuo. The concentrate was chromatographed on silica gel using 40% EtOAc/hexanes to give the product. LC/MS (M+H)+: 670, 1H NMR(CDCI3):6 8.89 (s, 1H), 8.27 (s, 1H), 8.24 (s, 1H), 6.84-7.51 (m, 10H), 6.87 (d, 1H), 5.73 (s, 2H), 4.39 (m, 1H), 3.60 (m, 2H), 3.12 (m, 2H), 1.76-2.11 (m, 9H), 0.96 (m, 2H), 0.00 (s, 9H).
Step 5: cis-4-[4-(7-[2-(Trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]cyclohexylmetha-not.
[0529] 7-[2-(Trimethylsilyl)ethoxy]methyl-4-(1-cis-4-[(trityloxy)methyl]cyclohexyl-1H-pyrazol-4-yl)-7H-pyrrolo[2,3-d]pyrimidine (0.3 g, 0.0004 mol) was dissolved in methanol (7.0 mL) and THF (2.0 mL, 0.025 mol) and 4.0 M HCI in 1,4-dioxane (0.5 mL) was added. The reaction was then stirred at 25°C for 2 hours TLC analysis showed no starting material present and LCMS analysis showed the presence of the product. The reaction was added to a saturated NaHCOg solution and was extracted with ethyl acetate. The organic extracts were washed with water, saturated NaCI, dried (MgS04) and concentrated in vacuo. The concentrate was chromatographed on silica gel using EtOAc as eluent to give the product. LC/MS (M+H)+: 428 1H NMR (CDCI3):ô 8.89 (s, 1H),8.37(s, 1H),8.31 (s, 1H),7.44(d, 1H),6.87(d, 1H), 5.73 (d, 2H), 4.41 (m, 1H), 3.51-3.71 (m, 4H), 2.31 (m, 2H), 2.08 (m, 3H), 1.70-1.93 (m, 4H), 0.98 (m, 2H), 0.00 (s, 9H).
Step 6: cis-4-[4-(7-[2-(Trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohexylmethyl methanesulfonate.
[0530] cis-4-[4-(7-[2-(Trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohexylmetha-nol was dissolved in chloroform (3.00 mL) and was cooled to 0 °C To the reaction was added TEA (0.10 mL, 0.00072 mol) and methanesulfonyl chloride (0.05 mL, 0.0006 mol) and this mixture was stirred at 0 °C for 2 hours at which time LCMS analysis showed mainly the product present in the mixture. The reaction was extracted with ethyl acetate and the organic extracts were washed with water, saturated NaCI, dried (MgS04) and concentrated in vacuo. LC/MS (M+H)+: 506
Step 7: cis-4-[4-(7-[2-(TrimethylsHyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohexylace-tonitrile.
[0531] cis-4-[4-(7-[2-(Trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohexylmethyl methanesulfonate (0.10 g, 0.00020 mol) and sodium cyanide (0.050 g, 0.0010 mol) and DMSO (1.0 mL) were mixed. The mixture was stirred at 60 °C for 24 hours, at which time LCMS analysis showed most of the starting material had been consumed. The reaction was extracted with ethyl acetate and the organic extracts were washed with water, saturated NaCI, dried (MgS04) and concentrated in vacuo. The concentrate was chromatographed on silica gel using EtOAc as eluent to give the product. LC/MS (M+H)+: 437, 1H NMR(CDCI3):Ô 8.90 (s, 1H), 8.36 (s, 1H), 8.31 (s, 1H), 7.45 (d, 1H), 6.87 (d, 1H), 5.73 (S, 2H), 4.43 (m, 1H), 3.60 (m, 2H), 2.45(d, 2H, J = 7.6 Hz), 2.37 (m, 2H), 2.10 (m,4H), 1.70-1.93 (m, 3H), 0.98 (m, 2H), 0.00 (s, 9H).
Step 8: cis-4-[4-(7H-Pyrrolo[2,3-dJpyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohexylacetonitrile.
[0532] cis-4-[4-(7-[2-(Trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohexylace-tonitrile (0.080 g, 0.00018 mol) and TFA (0.50 mL, 0.0065 mol) were added to DCM (3.00 mL, 0.0468 mol) and the mixture was stirred at 25 °C for 16 hours. The reaction was concentrated by roto-evaporation and the concentrate was dissolved in methanol (3.0 mL, 0.074 mol) and ammonium hydroxide (0.5 mL, 0.01 mol) was added This reaction was stirred at 25 °C for 6 hours at which time LCMS analysis showed no starting material present. The reaction was chromatographed on silica gel using 5% MeOH/EtOAc to give the product. LC/MS (M+H)+:307,1H NMR(CD3OD):0 8.64 (s, 1H), 8.55 (s, 1H), 8.31 (s, 1H), 7.50 (d, 1H), 6.96 (d, 1H), 4.42 (m, 1H), 2.61 (d, 2H, J = 8.0 Hz), 2.27 (m, 2H), 1.70-2.15 (m, 7H).
Example 680: cis-4-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohexylmethyl thiocyanate [0533]
Step 1: cis-4-[4-(7-[2-(Trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohexylmethyl thiocyanate [0534] cis-4-[4-(7-[2-(Trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohexylmethyl methanesulfonate (0.1 Og, 0.00020 mol) was dissolved in DMSO(1.00 mL) with potassium thiocyanate (0.082 g, 0.00084 mol). The reaction was heated at 68 °C for 4 days at which time LCMS analysis showed -4:1 product/starting material ratio. The reaction was extracted with ethyl acetate and the organic extracts were washed with water, saturated NaCI, dried (MgS04) and concentrated in vacuo. The concentrate was chromatographed on silica gel using 1:1 EtOAc/hexanes to give the product. LC/MS (M+H)+: 469, 1H NMR(CDCI3): δ 8.89 (s, 1H), 8.36 (s, 1H), 8.31 (s, 1H), 7.45 (d, 1H), 6.87 (d, 1H), 5.73 (S, 2H), 4.45 (m, 1H), 3.60 (m, 2H), 3.05 (m, 2H), 2.37 (m, 2H), 2.10 (m, 4H), 1.70-1.93 (m, 3H), 0.98 (m, 2H), 0.00 (s, 9H).
Step 2: cis-4-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohexylmethyl thiocyanate).
[0535] cis-4-[4-(7-[2-(Trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimdin-4-yl)-1H-pyrazol-1-yl]cyclohexylmethyl thiocyanate was dissolved in methanol (2.0 mL, 0.049 mol) and DCM (2.0 mL, 0.031 mol), and TFA (0.5 mL, 0.006 mol) was added. The resulting mixture was stirred at 25 °C for 16 hours. TLC analysis showed no starting material present and LCMS analysis showed product. The reaction was concentrated using a rotary evaporator and the concentrate was chromatographed on silica gel using 2% MeOH/EtOAc to give the product. LC/MS (M+H)+:339,1H NMR(CD3OD) δ 8.65 (s, 11-1),8.55 (s, 1H), 8.31 (s, 1H), 7.50 (d, 1H), 6.96 (d, 1H), 4.43(m, 1H), 3.20 (d, 2H, J = 7.6 Hz), 2.24 (m, 2H), 1.80-2.17 (m, 7H).
Example 681: N-5-[(cis-4-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohexyl-methyl)thio]-4H-1,2,4-triazol-3-ylpyrimidin-2-amine trifluoroacetate [0536]
Step 1: 5-[(cis-4-[4-(7-[2-(Trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohexylme-thyl) thio]-4H-1,2,4-triazol-3-amin [0537] cis-4-[4-(7-[2-(Trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohexylmethyl methanesulfonate (124.56 mg, 0.00024 mol), and 5-amino-4H-1,2,4-triazole-3-thiol (43.00 mg, 0.0003702 mol) were dissolved in DMF (1.20 mL) and potassium carbonate (0.122 g, 0.000887 mol) was added. The reaction was stirred at 50 °C for 18h, at which time LCMS showed nearly complete reaction, and product present. The reaction was extracted with ethyl acetate and the organic extracts were washed with water, saturated NaCI, dried (MgS04) and concentrated in vacuo. The concentrate was chromatographed on silica gel using EtOAc as eluent to give the product. LC/MS (M+H)+: 526, 1H NMR(CDCI3):ô 8.90 (s, 1H), 8.40 (s, 1H), 8.30 (s, 1H), 7.45 (d, 1H), 6.87 (d, 1H), 5.73 (S, 2H), 4.45 (brs, 2H), 4.41 (m, 1H), 3.60 (m, 2H), 3.22 (d, 2H, J=7.2 Hz), 2.29 (m, 2H), 1.70-2.10 (m, 7H), 0.98 (m, 2H), 0.00 (s, 9H).
Step 2: 5-[(cis-4-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohexylmethyl)thio]-4H-1,2,4-triazol-3-amine [0538] 5-[(cis-4-[4-(7-[2-(Trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohexylme-thyl)thio]-4H-1,2,4-triazol-3-amine (9a) was dissolved in TFA (1 mL) and was stirred for 2h. The solution was concentrated using a rotary evaporator to remove TFA. The residue was dissolved in methanol (1 mL) and ammonium hydroxide (1 mL) added. The solution was stirred overnight. LCMS showed complete de-protection. The solution was concentrated using a rotary evaporator. The product was isolated by prep LCMS using a 30mm x 100mm C18 column; 11%CH3CN-H20 (0.1%TFA), 1.5 min, to 33% at 6 min; 60 mL/min; detector set at m/z 396; retention time, 5.5min (2 runs). The eluate was freeze dried. Yield 21 mg (di-TFAsalt). LC/MS (M+H)+:396,1H NMR(dg-DMSO) δ 12.9 (brs, 1H, NH); 8.9 (2 singlets, 2H); 8.5 (s, 1H); 7.9 (m, 1H); 7.3 (m, 1H); 4.4 (m, 1H, NCH); 3.1 (d, 2H); 2.2 (m, 2H); 1.9 (m, 3H); 1.7 (m, 2H); 1.6 (m, 2H). MS(ES) 396 (M+1).
Example 682: N-5-[(cis-4-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohexylmethyl)thio]-4H-1,2,4-triazol-3-ylpyrimidin-2-amine trifluoroacetate [0539]
Step 1: N-5-[(cis-4-[4-(7-[2-(Trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]cyclohexyl-methyl)thio]-4H-1,2,4-triazol-3-yIpyrimidin-2-amine [0540] In a vial [A] 5-[(cis-4-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cy-clohexylmethyl)thio]-4H-1,2,4-triazol-3-amine (0.047 g, 0.000089 mol) was heated with 2-chloropyrimidine (0.011 g, 0.000096 mol) in 1,4-dioxane (1.00 mL, 0.0128 mol) at 150 °C for 40 minutes in a microwave reactor. LCMS analysis showed that no reaction had taken place. To the reaction was added 2-chloropyrimidine (0.020 g, 0.00017 mol) with cesium carbonate (0.033 g, 0.00010 mol) and copper(l) iodide (4.00 mg, 0.0000210 mol) and this mixture was heated at 115 °Cfor3 hours, at which time LCMS analysis showed no starting material present and mainly product was present. The reaction was chromatographed on silica gel using 2% MeOH/EtOAc to give the product. LC/MS (M+1)+:604, 1NMR(CDCI3): 8.89 (s, 1H), 8.82 m, 2H), 8.43 (s, 1H), 8.30 (s, 1H), 7.44 (d, 1H), 7.23 (m, 1H), 7.03 (brs, 2H), 6.88 (d, 1H), 5.73 (s, 2H), 4.40 (m, 1H), 3.60 (m, 2H), 3.35 (d, 2H), 2.34 (m, 2H), 1.80-2.15 (m, 7H), 0.98 (m, 2H), 0.00 (s, 9H).
Step 2: N-5-[(cis-4-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohexylmethyl)thio]-4H-1,2,4-triazol-3-ylpyri-midin-2-amine.
[0541] N-5-[(cis-4-[4-(7-[2-(Trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohexyl-methyl)thio]-4H-1,2,4-triazol-3-ylpyrimidin-2-amine (0.024 g, 0.000040 mol) was dissolved in DCM (4.00 mL), and TFA (0.50 mL, 0.0065 mol) was added. The reaction was stirred at 25 °C for 16 hours and was concentrated in vacuo. The residue was dissolved in methanol (3.00 mL) and concentrated ammonium hydroxide (0.50 mL) was added. This reaction was stirred at 25 °C for 2 hours at which time LCMS analysis showed mostly product. The reaction was concentrated using a rotary evaporator and the concentrate was purified by prep LC to give the product as the trifluoroacetate salt. LC/MS (M+H)+:474, 1H NMR(CD3OD) δ 8.87 (s, 1H), 8.85 (s, 1H), 8.81 (s, 1H), 8.79 (s, 1H), 8.45 (s, 1H), 7.85 (d, 1H), 7.34 (m, 2H), 4.43 (m, 1H), 3.20 (d, 2H, J = 7.6 Hz), 2.24 (m, 2H), 1.80-2.17 (m, 7H).
Example 683:3-cis-4-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohexylpropane-nitrile trifluoroacetate [0542]
Step 1: 2-(1,4-Dioxaspiro[4.5]dec-8-yl)ethanol.
[0543] Ethyl 1,4-dioxaspiro[4.5]dec-8-ylacelate (3.40 g, 0.0149 mol) prepared according to the procedure of Itagaki, Noriaki; Kimura, Mari;Sugahara,Tsutomu; Iwabuchi, Yoshiharu. (OrganicLetters2005; 7(19); 4181-4183.) was dissolved in ether (30.00 mL) and the mixture was cooled to 0 °C. To the reaction was added 1.00 M lithium tetrahydroaluminate in ether (15.0 mL) and the resulting mixture was stirred at 0 °C for 60 minutes and at 25 °C for 2 hours. The reaction was cooled and water (0.40 mL, 0.022 mol) was added, followed by 1.00 M sodium hydroxide (0.40 mL). To the reaction was then added ether (100.00 mL) and the solid that precipitated was filtered off. The filtrate was concentrated using a rotary evaporator to give the product. 1H NMR(CDCI3): 3.94 (s, 4H), 3.67 (t, 2H), 1.20-1.80 (m, 11H).
Step 2: 4-(2-Hydroxyethyl)cyclohexanone.
[0544] 2-(1,4-Dioxaspiro[4.5]dec-8-yl)ethanol (2.70 g, 0.0145 mol) was dissolved in acetone (10.00 mL) and THF (10.00 mL) and 6.00 M HCI (6.00 mL) was added. The reaction was stirred at 25 °C for 16 hours, neutralized with NaHC03 solution and was then extracted with ethyl acetate. The organic extracts were washed with water, and with saturated NaCI, then dried (MgS04) and concentrated in vacuo. The crude product was used in the next reaction without further purification. 1H NMR(CDCI3): 3.75 (m, 2H), 2.36 (m, 4H), 1.20-2.13 (m, 7H).
Step 3: 4-(2-Hydroxyethyl)cyclohexanol.
[0545] 4-(2-Hydroxyethyl)cyclohexanone (2.00 g, 0.0141 mol) was dissolved in ether (30.00 mL) and was cooled at 0 °C. To the reaction was added 1.0 M lithium tetrahydroaluminate in ether (14.1 mL) and the resulting mixture was stirred at 0 °C for 2 hours and at 25 °C for 16 hours. To the reaction was added THF (20.00 mL) and this mixture was cooled at 0 °C and then water (0.40 mL, 0.022 mol) was added, followed by 1.00 M sodium hydroxide (0.40 mL). To the reaction was then added ether (100.00 mL) and the resulting mixture was stirred for 10 minutes, then was filtered and the filtrate was concentrated using a rotary evaporator to provide the crude product. The crude product was used in the next reaction without further purification. 1H NMR(CDCI3): 3.96 and 3.57 (m, 1H) minor and major CHOH (-1:5 ratio) 3.70(m, 2H), 0.94-2.02 (m, 11H).
Step 4: 4-[2-(Trityloxy)ethyl]cyclohexanol.
[0546] 4-(2-Hydroxyethyl)cyclohexanol (crude from the previous reaction) (1.88 g, 0.0130 mol) was dissolved in pyridine (20.00 mL) and was cooled at 0 °C. To the reaction was added triphenylmethyl chloride (4.0 g, 0.014 mol) and this mixture was stirred at 0 °C for 2 hours and at 25 °C for 16 hours. The reaction was concentrated using a rotary evaporator and the concentrate was extracted with ethyl acetate. The organic extracts were washed with water, and saturated NaCI, then dried (MgS04) and concentrated in vacuo. The concentrate was chromatographed on silica gel (30%EtOAc/hex-anes) to give the trans isomer (1.98 g) 1H NMR(CDCI3): 7.42-7.45 (m, 6H), 7.20-7.30 (m, 9H), 3.50 (m, 1H), 3.07 (m, 2H), 1.93 (m, 2H), 1.66 (m, 2H), 1.17-1.60 (m, 5H), 0.89 (m, 2H).
Step 5: trans-4-[2-(Trityloxy)ethyl]cyclohexyl methanesulfonate.
[0547] trans-4-[2-(Trityloxy)ethyl]cyclohexanol (1.95 g, 0.00504 mol) was dissolved in chloroform (40.00 mL) and the mixture was cooled to 0 °C. To the reaction was added TEA (0.98 mL, 0.0071 mol) and methanesulfonyl chloride (0.47 mL, 0.0060 mol) and this mixture was stirred at 0 °C for 2 hours The reaction was then extracted with ethyl acetate and the organic extracts were washed with water, and saturated NaCI, then dried (MgS04) and concentrated in vacuo. 1H NMR(CDCI3): 7.41-7.45 (m, 6H), 7.20-7.32 (m, 9H), 4.55 (m, 1H), 3.07 (m, 2H), 2.10 (m, 2H), 1.70 (m, 2H), 1.20-1.60 (m, 5H), 0.95 (m, 2H).
Step 6: 7-[2-(Trimethylsilyl)ethoxy]methyl-4-(1-cis-4-[2-(trityloxy)ethyl]cyclohexyl-1 H-pyrazol-4-yl)-7H-pyrrolo[2,3-d]py-rlmldine.
[0548] 4-(1H-Pyrazol-4-yl)-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidine (1.0 g, 0.0032 mol) was mixed with sodium hydride (0.23 g, 0.0058 mol) and trans-4-[2-(trityloxy)ethyl]cyclohexyl methanesulfonate (2.10 g, 0.00452 mol) and this mixture was cooled to -78 °C. To the reaction was added DMF (6.00 mL) and this mixture was allowed to warm to 25 °C and was then stirred for 20 minutes. The reaction was stirred at 55 °C for 48 hours at which time LCMS analysis showed mostly product. The reaction was extracted with ethyl acetate and the organic extracts were washed with water and saturated NaCI, then dried (MgS04) and concentrated in vacuo. The concentrate was chromatographed on silica gel using 40% EtOAc/hexanes to give the product. LC/MS (M+H)+:684, 1H NMR(CDCI3): 8.89 (s, 1H), 8.35 (brs, 1H), 8.30 (s, 1H), 7.50 (m, 6H), 7.44 (d, 1H), 7.27-7.32 (m, 9H), 6.87 (d, 1H), 5.73 (s, 2H), 4.33 (m, 1H), 3.60 (m, 2H), 3.17 (t, 2H), 1.50-2.25 (m, 11H). 0.98 (m, 2H), 0.00(s, 9H).
Step 7: 2-cis-4-[4-(7-[2-(Trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yljcyclohexyletha-nol (7b).
[0549] 7-[2-(Trimethylsilyl)ethoxy]methyl-4-(1-cis-4-[2-(trityloxy)ethyl]cyclohexyl-1H-pyrazol-4-yl)-7H-pyrrolo[2,3-d]pyrimidine (1.45 g, 0.00212 mol) was dissolved in methanol (30.00 mL) and THF (10.00 mL) and 4.0 M HCI in 1,4-dioxane (2.00 mL) was added. The mixture was stirred at 25 °C for 2 hours, at which time, TLC analysis showed no starting material present and LCMS analysis showed the presence of the product. The reaction was added into a saturated NaHC03 solution, and was then extracted with ethyl acetate. The organic extracts were washed with water and saturated NaCI, then dried (MgS04) and concentrated in vacuo. The concentrate was chromatographed on silica gel using EtOAc as eluent to give the product. LC/MS (M+H)+: 442
Step 8: 2-cis-4-[4-(7-[2-(TrimethylsHyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohexylethyl methanesulfonate (8b).
[0550] 2 -cis-4-[4-(7-[2-(Trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohexyleth-ano (0.89 g, 0.0020 mol) was dissolved in DCM (12.00 mL, 0.1872 mol) and was cooled at 0 °C To the reaction was added TEA (0.43 mL, 0.0031 mol) and methanesulfonyl chloride (0.19 mL, 0.0024 mol) and this mixture was stirred at 0 °C for 2 hours at which time LCMS analysis showed mainly product present. The reaction was extracted with ethyl acetate and the organic extracts were washed with water and saturated NaCI, then dried (MgS04) and concentrated in vacuo. LC/MS (M+H)+:520, 1H NMR(CDCI3): 8.90 (s, 1H), 8.38 (brs, 1H), 8.31 (s, 1H), 7.45 (d, 1H), 6.88 (d, 1H), 5.73 (s, 2H), 4.40 (m, 1H), 4.27 (t, 2H), 3.60 (m, 2H), 3.07 (s, 3H), 1.60-2.40 (m, 11H). 0.98 (m, 2H), 0.00(s, 9H)
Step 9: 3-cis-4-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohexylpropanenitrile trifluoroacetate (9b).
[0551] 2-cis-4-[4-(7-[2-(Trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohexylethyl methanesulfonate (0.075 g, 0.00014 mol) was dissolved in DMSO (1.50 mL) and sodium cyanide (0.035 g, 0.00072 mol) was added. The reaction was stirred at 40 °Cfor 16 hours at which time LCMS analysis showed no starting material present. The reaction was then extracted with ethyl acetate and the organic extracts were washed with water and saturated NaCI, then dried (MgS04) and concentrated in vacuo. The residue was dissolved in DCM (3.00 mL) and TFA (0.50 mL, 0.0065 mol) was added. This mixture was stirred at 25 °C for 16 hours at which time LCMS analysis showed mostly the hydroxymethyl intermediate. The mixture was concentrated using a rotary evaporator and the concentrate was dissolved in methanol (3.00 mL) and concentrated ammonium hydroxide (0.50 mL) was added. The reaction was stirred at 25 °C for 3 hours at which time LCMS analysis showed no starting material present. The reaction was then concentrated using a rotary evaporator and the concentrate was purified by prep LC to give the product as the TFA salt (47.8 mg). LC/MS (M+H)+:321,1H NMR(CD3OD): 8.86 (s, 1H), 8.81(s, 1H), 8.44 (s, 1H), 7.84 (d, 1H), 7.31 (d, 1H), 4.48 (m, 1H), 2.51 (m,2H), 2.28 (m,2H), 2.00 (m, 2H), 1.80 (m, 5H), 1.67 (m, 2H).
Example 684: 5-[(2-cis-4-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohexyl-ethyl)thio]-4H-1,2,4-tri-azol-3-amine trifluoroacetate [0552]
[0553] 2-cis-4-[4-(7-[2-(Trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohexylethyl methanesulfonate (0.060 g, 0.00012 mol) was dissolved in DMF (1.31 mL) with 5-amino-4H-1,2,4-triazole-3-thiol (0.020 g, 0.00017 mol) and potassium carbonate (0.024 g, 0.00017 mol). This mixture was heated at 40 °C for 18 hours at which time LCMS analysis showed no starting material present. The reaction was diluted with EtOAc, filtered and was then concentrated using a rotary evaporator. The residue was dissolved in DCM (3.60 mL) and TFA (0.60 mL, 0.0078 mol) was added. This mixture was stirred at 25 °C for 5 hours and was then concentrated using a rotary evaporator. The residue was dissolved in methanol (3.60 mL) and concentrated ammonium hydroxide (0.60 mL) was added and this mixture was stirred at 25 °C for 2 hours. The reaction was concentrated using a rotary evaporator and the concentrate was purified by prep. LC to give the product. LC/MS (M+H)+:410, 1H NMR(CD3OD): 8.85 (s, 1H), 8.80(s, 1H), 8.44 (s, 1H), 7.83 (d, 1H), 7.30 (d, 1H), 4.46 (m, 1H), 3.17 (m, 2H), 2.27 (m, 2H), 2.00 (m, 2H), 1.62-1.90 (m, 7H).
Example 685: 4-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohexylideneacetonitrile trifluoroace-tate [0554]
Step 1: 1,4-Dioxaspiro[4.5]decan-8-ol [0555] 1,4-Dioxa-spiro[4.5]decan-8-one (2.00 g, 0.0128 mol) was dissolved in ether (50 mL) and the mixture was cooled to 0 °C. To the reaction was added 1 M lithium tetrahydroaluminate in ether (7.0 mL) and this mixture was stirred at 0 °C for 2 hours at which time TLC analysis showed no starting material present. The reaction was then quenched with water and 1 N NaOH (0.5 mL of each) and then filtered. The filtered solid was washed with ether and the combined ether filtrate was concentrated using a rotary evaporator to give the product. NMR (CDCI3): 3.94 (m, 4H), 3.81 (m, 1H), 1.79-1.92 (m, 4H), 1.54-1.70 (m,4H).
Step 2: 1,4-Dioxaspiro[4.5]dec-8-yl methanesulfonate.
[0556] 1,4-Dioxaspiro[4.5]decan-8-ol (0.40 g, 0.0025 mol) was dissolved in chloroform (10.0 mL) and the resulting mixture was cooled at 0 °C. To the mixture was added TEA (0.49 mL, 0.0035 mol) and methanesulfonyl chloride (0.23 mL, 0.0030 mol) and this mixture was stirred at 0 °C for 2 hours. The reaction was extracted with ethyl acetate and the organic extracts were washed with water, and saturated NaCI, then dried (MgS04) and concentrated in vacuo. The crude product was used in the next reaction without further purification. 1H NMR(CDCI3): 4.85 (m, 1H), 3.95 (m, 4H), 3.02 (s, 3H), 1.98-2.05 (m, 4H), 1.82-1.89 (m, 2H), 1.61-1.70 (m, 2H).
Step 3: 4-[ 1 -( 1,4-Dioxaspiro[4.5]dec-8-yl)-1H-pyrazol-4-yl]-7-[2-(trimethylsilyl)ethoxy]methyl-7H pyrrolo[2,3-d]pyrimi-dine.
[0557] A mixture of 1,4-dioxaspiro[4.5]dec-8-ylmethanesulfonate (0.50 g, 0.0015 mol) with 4-(1 H-pyrazol-4-yl)-7-[2-(tri-methylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidine (0.36 g, 0.0011 mol) and sodium hydride (0.082 g, 0.0020 mol) was cooled at -78 °C and DMF (2.0 mL) was added. The reaction was allowed to warm to 25 °C and was then stirred for 20 minutes and was then heated to 55 °C for 24 hours. The reaction was then extracted with ethyl acetate. The organic extracts were washed with water and saturated NaCI, then dried (MgS04) and concentrated in vacuo. The concentrate was chromatographed on silica gel using 1:1 EtOAc/hexanes to give the product. LC/MS (M+H)+:456, 1H NMR(CDCI3): 8.89 (s, 1H), 8.35 (s, 1H), 8.30 (s, 1H),7.44(d, 1H), 6.87 (d, 1H), 5.73 (s, 2H),4.38 (m, 1H), 4.06 (s, 4H), 3.60 (m, 2H), 2.22-2.31 (m, 4H), 2.00 (m, 2H), 1.86 (m, 2H), 0.98 (m, 2H), 0.00(s, 9H)
Step 4: 4-[4-(7-[2-(Trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]cyclohexanone [0558] To 4-[1 -(1,4-dioxaspiro[4.5]dec-8-yl)-1H-pyrazol-4-yl]-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyri-midine (2.13 g, 0.00467 mol), was added acetone (85 mL) followed by 12 M HCI in water (4.0 mL). The reaction was stirred at RT. After 1 h, LCMS analysis showed 66% reaction. After 4 h, HPLC showed 80% reaction. After 20 h, HPLC showed no change (and no loss of SEM). The reaction mixture was quenched into excess sat’d NaHC03. The acetone was removed by roto-evaporation. The resulting mixture of aqueous bicarbonate and a white solid was then extracted with EtOAc. The combined organic extract was shaken with sat’d NaCI, dried over Na2S04, then concentrated to dryness to leave 2.0 g of a crude product. TLC (5% iPrOH-40% EtOAc-hexane): product Rf 0.12 (ketal 0.22). The crude product was purified by automatic flash chromatography on silica gel. Used a 40g column; flow 40 mL/min; [A= 2% iPrOH-hexane] [B= 6% iPrOH-50% EtOAc/hexane]; A, 2 min; Gradient to B in 25 min, then B for 10 min. The eluent was concentrated using a rotary evaporator to give 1.3 g of a white solid. HPLC Method: Zorbax SB C18, 5 μΐη, 15 cm, 35 °C, flow 1.2 mL/min, 10% CH3CN-H20 (0.05% TFA), to 100% CH3CN in 9.0 min; stop time 12.3 min; detector 268 nm; retention time starting material, 7.4 min; product, 6.9 min (UV max 220, 268, 300, 322 nm). 1H NMR (CDCI3) δ 8.8 (s, 1H); 8.3 (m, 2H); 7.4 (d, 1H); 7.3 (s, 1H); 6.8 (d, 1H); 5.7 (s, 2H); 4.7 (m, 1H, NCH); 3.6 (t, 2H); 2.3-2.5 (m, 8H); 0.9 (t, 2H); -0.1 (s, 9H). MS(ES) 412 (M+1).
Step 5: 4-[4-(7-[2-(Trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]cyclohexylideneace-tonitrile [0559] To a solution of 1.0 M potassium ferf-butoxide in THF (1.90 mL) at 0 °C was added a solution of diethyl cyanomethylphosphonate (321 μί, 0.00198 mol) in THF (4 mL) dropwise. The reaction was held for 10 min, then it was added to a solution of 4-[4-(7-[2-(trimethylsilyl)-ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohex-anone (743 mg, 0.00180 mol) in THF (5 mL) stirring at 0 °C under a nitrogen atmosphere. The reaction was stirred 1.5 h at rt. LCMS analysis showed clean conversion to the desired product. To the reaction mixture was then added water and EtOAc. The phases were separated and the aqueous phase was extracted with EtOAc. The combined organic extract was washed with water, then sat’d NaCI, then dried over Na2S04, and concentrated to dryness to yield 0.76 g of a white crystalline solid (TLC (EtOAc) Rf 0.33). The product was purified by automatic flash chromatography on silica gel. Used 40g column; flow 40 mL/min; [A= hexane] [B= EtOAc]; A, 2 min; Gradient to B in 20 min. Rotary evaporation yielded 0.70 g of a white crystalline solid (89% yield). 1H NMR (CDCI3) δ 8.9 (s, 1H); 8.3 (s, 2H); 7.4 (d, 1H); 7.3 (s, 1H); 6.9 (d, 1H); 5.7 (s, 2H); 5.3 (s, 1H, olefin); 4.5 (m, 1H, NCH); 3.6(m,2H); 3.2 (m, 1H);2.7(m, 1H); 2.5 (m, 4H); 2.1 (m, 2H); 1.0 (m, 2H); -0.1 (s, 9H). MS(ES) 435 (M+1).
Step 6: 4-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohexylideneacetonitrHe [0560] A solution of TFA (0.5 mL, 0.006 mol) and 4-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]cyclohexylideneacetonitrile (22.7 mg, 0.0000522 mol), was stirred for 1.5h. The solution was then concentrated using a rotary evaporator to remove TFA. LCMS analysis showed conversion to the hydroxymethyl intermediate, M+H 335. Methanol was added; and the methanol mixture was concentrated again using a rotary evaporator. The resulting residue was dissolved in methanol (1 mL) and ammonium hydroxide (0.25 mL, 0.0064 mol) was added. The resulting solution was stirred for 16 h. LCMS analysis showed complete de-protection. The solution was then concentrated using a rotary evaporator. The product was isolated by prep HPLC using a 30 mm x 100 mm C18 column; 18% CH3CN-H20 (0.1%TFA), 1min, to 35% at 6min; 60 mL/min; detector set at 254nm; retention time, 4.4min. The eluate was freeze dried, yield 7.6 mg of a white solid (TFA salt; racemic; 34.6%). 1H NMR (d6-DMSO) δ 12.9 (brs, 1H, NH); 8.9 (s, 2H); 8.5 (s, 1H); 7.8 (m, 1H); 7.3 (m, 1H);5.6(s, 1H, olefin); 4.6 (m, 1H, NCH); 2.8 (m, 1H);2.6 (m, 1H);2.5 (m, 2H); 2.3 (m, 2H) 2.0 (m, 2H). MS(ES) 305 (M+1).
Example 686: cis-4-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohexanecarbonitrile trifluoroace-tate [0561]
Step 1: cis-4-[4-(7-[2-(Trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]cyclohexanecarbal-dehyde oxime [0562] A solution of sulfur trioxide-pyridine complex (53.4 mg, 0.000336 mol) in DMSO (0.3 mL, 0.004 mol) was added to a solution of cis-4-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohexyl-methanol (57.4 mg, 0.000134 mol) and TEA (56.1 μί, 0.000403 mol) in DCM (0.3 mL, 0.004 mol) at-10 °C. The mixture was stirred vigorously at 10-20 °C for one hour. LCMS analysis showed conversion to the aldehyde. The mixture was then poured into ice-water, and extracted with DCM. The extracts were washed with 10 % citric acid, water, saturated aqueous sodium bicarbonate, water, and brine, and then dried oversodium sulfate. Concentration gave 57 mg of a residue.
[0563] To the resulting residue was added hydroxylamine-HCI (50mg), 1 mL 20% K2C03, and 3 mL MeOH and this mixture was stirred at rt until LCMS showed conversion to the corresponding oxime, M+H 441. The product was isolated by prep HPLCMS using a 30 mm x 10, 0 mm, C18 column; 30% CH3CN-H20 (0.1%TFA), 1 min, to 60% at 6 min; 60 mL/min; detector set at m/z 441 ; retention time, 6.0min. freeze-dried, yield 17.4 mg of a white solid.
Step 2: cis-4-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-ylJcyclohexanecarbonitrile [0564] [A] cis-4-[4-(7-[2-(Trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohexanecarbal-dehyde oxime (11.0 mg, 0.0000250 mol) was dissolved in pyridine (0.25 mL, 0.0031 mol), and benzenesulfonyl chloride (10.0 μί, 0.0000784 mol) was added and the resulting mixture was stirred at rt. After stirring 15 h, LCMS analysis showed formation of the product, M+H 423. The product was isolated by prep HPLCMS using a 19 mm x 100 mm C18 column; 45% CH3CN-H20 (0.1% NH4OH), 1min, to 75% at 6 min; 30 mL/min; detector set at m/z 423; retention time, 4.8 min. The eluent was concentrated using a rotary evaporator to give 8 mg of the desired product.
[0565] The product was dissolved in TFA (0.25 mL). stirred for 2h. The solution was concentrated using a rotary evaporator to remove TFA. Methanol was added and the mixture was concentrated again. LCMS showed clean conversion to the hydroxymethyl intermediate (M+H 323). The residue was dissolved in methanol (1 mL) and ammonium hydroxide (0.25 mL) was added. The solution was stirred 0.5 h, at which time, LCMS showed complete de-protection to the desired product M+H 293. The mixture was then concentrated by roto-evaporation, and the product was isolated by prep HPLCMS using a 19 mm x 100 mm C18 column; 15% CH3CN-H20 (0.1% TFA), 1.5 min, to 30% at 6 min; 30 mL/min; detector set at m/z 293; retention time, 5.2 min. The eluate was freeze dried to yield 5.5 mg of the product as a TFA salt. 1H NMR (dg-DMSO) δ 12.82 (brs, 1H, NH); 8.87 (s, 1H); 8.85 (s, 1H); 8.48 (s, 1H); 7.82 (m, 1H); 7.24 (m, 1H); 4.40 (m, 1H, NCH); 3.22 (m, 1H); 2.05 (m, 6H); 1.79 (m, 2H). MS(ES) 293 (M+1).
Example 687: 2-[(cis-4-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohexylmethyl)sulfinyl]benzoni-trile trifluoroacetate [0566]
Step 1: 4-[1-(cis-4-[(2-Bromophenyl)thio]methylcyclohexyl)-1H-pyrazol-4-yl]-7-[2-(trimethylsilyl)-ethoxy]methyl-7H-pyr-rolo[2,3-d]pyrimidine [0567] This compound was prepared from (cis-4-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohexylmethyl methanesulfonate as in Example 686[A], Yield 73%. The product was purified using the following HPLC method: Zorbax SB C18, 5 μΠΊ, 15cm, 35 C, flow 1.2 mL/min, 10% CH3CN-H20 (0.05% TFA), to 100% CH3CN in 9.0 min; stop time 12.3 min; detector 254 nm; retention time starting mesylate, 7.5 min; product, 9.9 min (UV max 215, 258, 300, & 326 nm). TLC: Rf 0.3 using 35% EtOAc/5% iPrOH/hexane. The product was purified by automated silica gel flash chromatography using 30% EtOAc/5% iPrOH/hexane. 1H NMR (CDCI3) δ 8.84 (s, 1H); 8.31 (s, 1H); 8.26 (s, 1H); 7.55 (m, 1H); 7.39 (d, 1H); 7.27 (m, 2H); 7.03 (m, 1H); 6.82 (d, 1H); 5.67 (s, 2H); 4.34 (m, 1H, NCH); 3.55 (m, 2H); 2.98 (d, 2H); 2.28 (m, 2H); 2.02 (m, 3H); 1.83 (m, 4H); 0.92 (m, 2H); -0.06 (s, 9H). MS(ES) 598/600 1:1 (M+1).
Step 2: 2-[(cis-4-[4-(7-[2-(Trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]cyclohexylme-thyl)lhio]benzonitrile [0568] 4-[1-(cis-4-[(2-Bromophenyl)thio]methylcyclohexyl)-1H-pyrazol-4-yl]-7-[2-(trimethylsilyl)-ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidine (62.7 mg, 0.000105 mol), zinc cyanide (123 mg, 0.00105 mol), and tetrakis(triphenylphos-phine)palladium(O) (30.2 mg, 0.0000262 mol) were stirred in DMF (3 mL) and the solution was flushed with nitrogen. The solution was then heated to 100 °C for 25 min in a microwave reactor. LCMS and HPLC analyses showed > 90% reaction. The product was isolated by prep HPLCMS using a 30 mm x 100 mm C18 column; 52%CH3CN-H20 (0.1 %TFA), 1.5 min, to 75% at 6 min; 60 mL/min; detector set at 545 nm. The eluent was concentrated using a rotary evaporator to give 37 mg of the 2-cyanophenylsulfide TFA salt. HPLC Method: Zorbax SB C18, 5 μ(η, 15 cm, 35 C, flow 1.2 mL/min, 10% CH3CN-H20 (0.05% TFA), to 100% CH3CN in 9.0 min; stop time 12.3 min; detector 265 nm; retention time starting material, 9.9 min; product, 8.9 min. MS(ES) 545 (M+1).
Step 3: 2-[(cis-4-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohexylmethyl)sulfinyl]-benzonitrile [0569] Asolutionof2-[(cis-4-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cy-clohexylmethyl)thio]benzonitrile (30.6 mg, 0.0000562 mol), in TFA (1 mL) was stirred for 2 h. The solution was concentrated using a rotary evaporator to remove TFA. Methanol was added, and the mixture was concentrated again. The resulting residue was dissolved in methanol (1 mL) and ammonium hydroxide (1 mL) was added. The resulting solution was stirred overnight, at which time HPLC showed complete deprotection. The product was isolated by prep HPLCMS using a 19 mm x 100 mm C18 column; 30% CH3CN-H20 (0.1% TFA), 1.5 min, to 59% at 6 min; 30 mL/min; detector set at m/z 415 nm; retention time, 4.7 min. The eluate was concentrated using a rotary evaporator to give 36 mg of the sulfide TFA salt, a colorless glassy material. NMR (dg-DMSO) δ 12.82 (brs, 1H, NH); 8.84 (2 singlets, 2H); 8.45 (s, 1H); 7.8 (m,2H); 7.64 (m,2H); 7.34 (td, 1H); 7.24 (s, 1H);4.39 (m, 1H, NCH); 3.23 (d,2H); 2.19 (m, 2H); 1.89 (m, 3H); 1.72 (m, 4H). MS(ES) 415 (M+1). This material was then dissolved in CH2CI2 and cooled to 0 °C. To the cooled mixture was added MCPBA(12.9 mg, 0.0000562 mol), and the resulting mixture was stirred for 1 h. LCMS showed conversion to the product, and no remaining sulfide. The reaction mixture was concentrated by rotovap, and the product was isolated by prep HPLCMS using a 19 mm x 100 mm C18 column; 18% CH3CN-H20 (0.1% TFA), 1. 0 min, to 35% at 6 min; 30 mL/min; detector set at m/z 431 nm; retention time, 5.6 min. The product was isolated from the eluent by freeze-drying. The yield was 27.6 mg of the TFA salt. The HPLC method was: Zorbax SB C18, 5 μίτι, 15 cm, 35 °C, flow 1.2 mL/min, 10% CH3CN-H20 (0.05% TFA), to 100% CH3CN in 9.0 min; stop time 12.3 min; detector 268 nm; retention time starting material, 5.6 min; sulfoxide, 4.8 min; sulfone, 5.2 min; MCPBA, 6.0 min. 1H NMR (CDCI3) δ 12.1 (brs, 1H, NH); 9.0 (s, 1H); 8.9 (s, 1H); 8.3 (s, 1H); 8.1 (m, 1H); 7.9 (m, 1H); 7.8 (m, 1H); 7.6 (m, 2H); 7.0 (m, 1H); 4.4 (m, 1H, NCH); 3.1 (dd, 1H); 2.9 (dd, 1H);2.5(m, 1H);2.3(m, 1H); 2.3-1.7 (m, 7H). MS(ES) 431 (M+1).
Example 688: 2-[(cis-4-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohexylmethyl)sulfonyl]benzoni-trile trifluoroacetate [0570]
[0571] 2-[(cis-4-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohexylmethyl)sulfinyl]-benzonitrile (17.2 mg, 0.0000400 mol) (21 mg TFA salt), was dissolved in DCM (10 mL) and cooled to 0 °C. To this mixture was added MCPBA (18 mg, 0.0000800 mol). The resulting mixture was stirred for 1h atO °C, and then for 16 h at rt. HPLC and LCMS showed 80 area% product, and 3 area% sulfoxide. The MCPBA was removed using a sat’d NaHC03 wash, and the resulting washed mixture was concentrated by roto-evaporation. The product was isolated by prep HPLCMS using a 19 mm x 100 mm C18 column; 23%CH3CN-H20 (0.1%TFA), 1.0 min, to 43% at 6 min; 30 mL/min; detector set at m/z 447 nm; retention time, 5.1 min. The product was isolated from the eluent by freeze-drying. The yield was 5 mg of the TFA salt. 1H NMR (dg-DMSO) δ 12.70 (brs, 1H, NH); 8.83 (s, 1H); 8.82 (s, 1H); 8.41 (s, 1H); 8.21 (dd, 1H); 8.16 (dd, 1H); 8.01 (td, 1H); 7.95 (td, 1H); 7.78 (s, 1H); 7.19 (s, 1H); 4.34 (m, 1H, NCH); 3.62 (d, 2H); 2.28 (m, 1H); 2.10 (m, 2H); 1.90 (m, 2H); 1.72 (m, 4H). MS(ES) 447 (M+1).
Example 689: 3-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohexylacetonitrile trifluoroacetate [0572]
Step 1: 3-[4-(7-[2-(Trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]-cyclohexanone [0573] To a solution of 4-(1H-pyrazol-4-yl)-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]-pyrimidine (309 mg, 0.980 mmol) in ACN (6 mL) was added 2-cyclohexen-1-one (190 μί, 01.96 mmol), followed by DBU (40 μί, 0.3 mmol). The resulting mixture was stirred for one hour at which point LCMS indicated complete addition. The mixture was reduced in vacuo and the crude product was purified by column chromatography to obtain the product (397 mg, 98%). 1H NMR (400 MHz, CDCI3): δ 8.84 (s, 1H), 8.27 (s, 1H), 8.25 (s, 1H), 7.45 (d, 1H), 6.79 (d, 1H), 5.67 (s, 2H), 4.61 (m, 1H), 3.55 (m, 2H), 3.05-2.90 (m, 2H), 2.45-2.30 (m, 4H), 2.05 (m, 1H), 1.90 (m, 1H), 0.92 (m, 2H), -0.06 (s, 9H). MS (El) mlz = 412.2 (M+H).
Step 2: (2E,Z)-3-[4-(7-[2-(Trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]cyclohexylide-neacetonitrile [0574] To a solution of t-BuOK in THF (1.0 M, 0.255 mL, 0.255 mmol) at 0 °C was added a solution of diethyl cy-anomethylphosphonate (43 μί, 0.27 mmol) in THF (0.6 mL) dropwise. The reaction was held for 10 minutes, then a solution of 3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]-pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohexanone (100.0 mg, 0.2430 mmol) in THF (0.34 mL) was added dropwise. After complete addition, the cooling bath was removed and the reaction was held at ambient temperature for 16 hours, at which point LCMS indicated complete addition to yield the desired product as a mixture of E and Z isomers (87.9 mg, 83%). 1H NMR (400 MHz, CDCI3): δ 8.84 (s, 0.5H), 8.83 (s, 0.5 H), 8.27 (d, 1H), 8.25 (s, 1H), 7.40 (s, 0.5H), 7.39 (s, 0.5H), 6.81 (d, 0.5H), 6.79 (d, 0.5H), 5.67 (s, 2H), 5.28 (s, 0.5H), 5.24 (s, 0.5H), 4.4 (m, 1H), 3.55 (m, 2H), 3.1-2.8 (m, 2H), 2.5-2.1 (m, 6H), 0.92 (m, 2H), -0.06 (s, 9H). MS (En m/z = 435.2 (M+H).
Step 3: 3-[4-(7-[2-(Trimethylsilyl)ethoxy}methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]-cyclohexylacetonitrile [0575] To (2E, Z)-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohexyli-deneacetonitrile (42.0 mg, 0.0966 mmol) was added THF (0.5 mL). The resulting solution was cooled to -78 °C, and then 1.0 M L-Selectride® in THF (120 μι, 0.12 mmol) was added dropwise. The reaction was held at -78 °C for 1 h at which point LCMS indicated complete reduction. The reaction was quenched at-78 °C by addition of saturated aqueous NH4CI and EtOAc, and was then allowed to warm to ambient temperature. The phases were separated and the aqueous phase was extracted with additional EtOAc. The combined organic phase was washed with water, then saturated NaCI, and then was dried over MgS04. The crude product was purified by column chromatography to obtain the product (26.5 mg, 63%). 1H NMR (400 MHz, CDCI3): δ 8.84 (s, 1H), 8.32 (s, 1H), 8.25 (s, 1H), 7.39 (d, 1H),6.81 (d, 1H), 5.67 (s, 2H), 4.53 (m, 1H), 3.52 (m, 2H), 2.6-1.4 (m, 11H), 0.92 (m, 2H), -0.06 (s, 9H). MS (El) m/z=437.2 (M+H).
Step 4: 3-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl) -1 H-pyrázol-1 -yljcyclohexylacetonitrile trifluoroacetate [0576] To 3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohexylace-tonitrile (30.1 mg, 0.0689 mmol) was added DCM (1.0 mL) and TFA (1.0 mL). The resulting mixture was stirred for 1 hour at ambient temperature, at which point LCMS indicated complete cleavage to the N-hydroxymethyl intermediate. The solvent was removed and to the residue was added methanol (1.0 mL) followed by ethylenediamine (37 μί, 0.55 mmol), after which the reaction was stirred for 5 hours, at which point LCMS indicated complete reaction. The solvent was removed and the residue was purified by preparative LCMS to provide the product as a TFA salt (24 mg, 83%). 1H NMR (400 MHz, CD3OD): δ 8.91 (s, 1H), 8.82 (s, 1H), 8.45 (s, 1H), 7.84 (s, 1H), 7.31 (s, 1H), 4.69 (s, 1H), 2.58 (d, 2H), 2.5-1.5 (m, 9H). MS (El) m/z = 307.10 (M+H).
Example 690: 5-({cis-4-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohexyl}thio)-1H-1,2,4-triazol-3-amine bis(trifluoroacetate) [0577]
Step 1: trans-4-[4-(7-[2-(Trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1 -yl]cyclohexanol [0578] A solution of 4-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]cyclohex-anone (662 mg, 1.61 mmol) in THF (5 mL) was cooled to 0 °C and lithium tetrahydroaluminate (2M in THF, 0.804 mL, 1.61 mmol) was added slowly. The mixture was allowed to warm slowly to ambient temperature until LCMS indicated complete reduction. The reaction was cooled to 0 °C and quenched with dropwise addition of water (0.5 mL). DCM was added, and the mixture was stirred for 1 hour at ambient temperature, after which the precipitated solids were removed by filtration. The filtrate was reduced in vacuo to leave a white solid (0.63g, 99%). HPLC of the solid showed an approximately 4:1 ratio of trans to cis product. Tic (6:3:1 EtOAc:hexanes:isopropanol) gave an Rf of 0.25 for the cis product, and 0.18 for the trans product. The product was purified by flash chromatography on silica gel to recover 230 mg of the pure trans alcohol and 25 mg pure of the cis alcohol, and 350 mg of mixed isomers. 1H NMR (400 MHz, CDCI3): δ 8.83 (s, 1H), 8.27 (s, 1H), 8.24 (s, 1H), 7.39 (d, 1H), 6.81 (d, 1H), 5.67 (s, 2H), 4.24 (m, 1 H), 3.79 (m, 1H), 3.54 (m, 2H), 2.28 (m, 2H), 2.17 (m, 2H), 1.94 (m, 2H), 1.53 (m, 2H), 0.92 (m, 2H), -0.06 (s, 9H). MS (El) mlz = 414 (M+H).
Step 2: trans-4-[4-(7-[2-(Trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohexyl meth-anesulfonate [0579] To trans-4-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohexanol (154 mg, 0.372 mmol) was added DCM (1.0 mL) and TEA (73 μΙ_, 0.52 mmol). The resulting solution was then cooled to 0 °C and methanesulfonyl chloride (34 μι, 0.45 mmol) was added. The reaction was held for 2 hours, at which point tic and LCMS indicated complete reaction. The reaction was partitioned between water and DCM, the phases were separated and the aqueous phase was extracted with additional solvent. The combined organic phase was washed with water, then saturated NaCI, then was dried over MgS04 and reduced in vacuo to give the crude product which was used without further purification (173 mg, 95%). 1H NMR (400 MHz, CDCI3): δ 8.83 (s, 1H), 8.24 (s, 2H), 8.24 (s, 1H), 7.39 (d, 1H), 6.80 (d, 1H), 5.67 (s, 2H), 4.77 (m, 1H), 4.27 (m, 1H), 3.54 (m, 2H), 3.06 (s, 3H), 2.36 (m, 4H), 2.03 (m, 2H), 1.82 (m, 2H), 1.53 (m, 2H), 0.92 (m, 2H), -0.06 (s, 9H). MS (El) mlz = 492.1 (M+H).
Step 3: 5-({cis-4-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1 -yl]cyclohexyl)thio)-1 H-1,2,4-triazo!-3-amine bis(trif-luoroacetate) [0580] To a solution of trans-4-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]cyclohexyl methanesulfonate (42 mg, 0.085 mmol) in DMF (800 μΙ_) was added 3-amino-1 H-1,2,4-triazole-5-thiol (30 mg, 0.26 mmol) and K2C03 (36 mg, 0.26 mmol). The reaction was sealed and held at 100 °C for 2 hours at which point LCMS indicated conversion to desired product. The reaction was diluted with water and extracted successively with ether, ethyl acetate, and 3:1 chloroform Isopropyl alcohol. The combined organic phase was washed with water, then saturated NaCI, dried over MgS04 and reduced in vacuo, and the crude product was purified by column chromatography to give 5-({cis-4-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cy-clohexyl}thio)-1 H-1,2,4-triazol-3-amine (27.3 mg, 63%). To the product was added DCM (0.5 mL) and TFA (0.5 mL), and the reaction was stirred for 1 hour at ambient temperature at which point LCMS indicated complete cleavage to the N-hydroxymethyl intermediate. The solvent was removed and to the residue was added methanol (1.0 mL) followed by NH4OH (0.3 mL), the reaction was stirred for 16 hours at which point LCMS indicated complete deprotection. The solvent was removed and the residue was purified by preparative LCMS to provide the product as a bis-TFA salt (15.1 mg, 29%). 1H NMR (400 MHz, CD3OD): δ 8.77 (s, 1H), 8.72 (s, 1H), 8.37 (s, 1H), 7.74 (d, 1H), 7.21 (d, 1H), 4.40 (m, 1H), 3.97 (m, 1 H), 2.25(m, 2H), 2.04 (m, 6H). MS (El)m/z = 382.2 (M+H).
Example 691: N-{5-[({cis-4-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohexyl}-methyl)thio]-4H- 1,2,4-triazol-3-yl}methanesulfonamide trifluoroacetate [0581]
Step 1. N-5-[(cis-4-[4-(7-[2-(Trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yljcyclohexyl-methyl)thio]-4H-1,2,4-triazol-3-ylmethanesulfonamide [0582] 5-[(cis-4-[4-(7-[2-(Trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidm-4-yl)-1H-pyrazol-1-yl]cyclohexylme-thyl)thio]-4H-1,2,4-triazol-3-amine (30.00 mg, 5.706E-5 mol) was dissolved in DCM (2.00 mL, 0.0312 mol) with TEA (0.024 mL, 0.00017 mol) and was cooled at 0 °C. To the reaction was added methanesulfonyl chloride (0.0066 mL, 0.000086 mol) and the resulting mixture was stirred at 0 °Cfor60 minutes, at which time LCMS analysis showed mostly product. The reaction was chromatographed on silica gel using EtOAc as eluent to give the product. LC/MS (M+1 )+:604
Step 2. N-5-[(cis-4-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]cyclohexylmethyl)thio]-4H-1,2,4-triazol-3-yl-methanesulfonamide [0583] Into a 1-neck round-bottom flask [A] N-5-[(cis-4-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimi-din-4-yl)-1H-pyrazol-1-yl]cyclohexylmethyl)thio]-4H-1,2,4-triazol-3-ylmethane-ulfonamide (0.025 g, 0.000041 mol) was dissolved in DCM (3.00 mL, 0.0468 mol) and TFA (mL, 0.006 mol) was added. The reaction was stirred at 25 °C for 16 hours at which time LCMS analysis showed no starting material present. The reaction was concentrated using a rotary evaporator and was dissolved in methanol (2.00 mL, 0.0494 mol) and 16 M ammonia in water (0.2 mL) was added. The reaction was stirred at 25 °C for 3 hours at which time LCMS analysis showed no starting material present. The reaction was concentrated using a rotary evaporator and was purified by prep LC to give the product as the trifluoroacetate salt. LC/MS (M+1)+:474, 1H NMR(CD3OD): 8.87 (s, 1H), 8.82 (s, 1H), 8.45 (s, 1H), 7.85 (d, 1H), 7.33 (d, 1H), 4.48 (m, 1H), 3.36 (s, 3H), 3.23 (d, 2H), 2.30 (m, 2H), 2.04 (m, 3H), 1.85 (m, 4H).
Example 692: [cis-4-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-1-(1H-1,2,4-triazol-1-yl)cyclohexyl] acetonitrile [0584]
[0585] 1H-1,2,4-Triazole (91.0 mg, 0.00132 mol), DBU (174 μί, 0.00070 mol), [A] 4-[4-(7-[2-(trimethylsilyl)ethoxy]me-thyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohexylidene-acetonitrile (86.4 mg, 0.000199 mol), and ACN (2.0 mL) were stirred at rt. After 4d, LCMS showed about 58 area% product (two peaks, M+H 504, ratio 1:1). The DBU in the reaction was neutralized with TFA. The product was isolated by prep HPLC using a 30 mm x 100 mm C18 column; 32% CFI3CN-H2O (0.1%TFA), 1 min, to 47% at 6 min; 60 mL/min; detector set at 254 nm; retention time, 5. 1 (A) & 5.4 (B) min. The eluent was concentrated using a rotary evaporator to give 22 mg of (A) & 36 mg of (B).
[0586] Deprotection: The products were dissolved separately in TFA (0.5 mL) and stirred for 1h. LCMS showed conversion to the hydroxymethyl derivative (M+H 404). The solutions were concentrated using a rotary evaporator to remove TFA. Methanol was added, and the resulting mixtures were concentrated again. The resulting residue was dissolved in methanol (1 mL), and ammonium hydroxide (0.25 mL) added. The solution was stirred 0.5h. LCMS showed complete de-protection (M+H 374) and the mixture was then concentrated by roto-evaporation. Each isomer was isolated by prep HPLCMS using a 19 mm x 100 mm C18 column; 15% CH3CN-H20 (0.1% TFA), 1.5 min, to 32% at 6 min; 30 mL/min; detector set at m/z 374; retention time, 4.5 min (A) & 4.7 min (B). The eluates were freeze dried. Yield 13 mg isomer A and 24 mg isomer B (TFA salts, white solids). NMR analysis (including NOE & COSY) was consistent with expectation for the structures, with A=cis, and B=trans. NMR (d6-DMSO) δ cis: 12.94 (brs, 1H, NH); 8.95 (s, 1H); 8.87 (s, 1H); 8.81 (s, 1H); 8.42 (s, 1H); 8.14 (s, 1H); 7.85 (m, 1H); 7.22 (m, 1H); 4.48 (m, 1H, NCH); 3.12 (s, 2H); 2.84 (m, 2H); 2.07 (m, 4H); 1.69 (m, 2H). MS(ES) 374 (M+1). trans: 12.85 (brs, 1H, NH); 8.94 (s, 1H);8.89(s, 1H);8.84 (s, 1H); 8.47 (s, 1H); 8.11 (s, 1H); 7.84 (m, 1H); 7.26 (m, 1H); 4.50 (m, 1H, NCH); 3.48 (s, 2H); 2.42-2.10 (m, 8H). MS(ES) 374 (M+1).
Example 705:3-1-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]but-3-yn-1-yl-benzonitrile trifluoroacetate [0587]
Step 1: 3-{1-[4-(7-{[2-(Trimethylsilyl)ethoxy]methyí}-7Hpyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]but-3-yn-1-yl}ben-zonitrile [0588]
[0589] 1 M Diisobutylaluminum hydride in hexane (0.31 mL) was added dropwise to a solution of methyl 3-(3-cyano-phenyl)-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanoate (100 mg, 0.0002 mol) (prepared by using a procedure analogous to Example 712, Step 1) in DCM (3 mL, 0.05 mol) and the mixture was cooled to -78 °C. The reaction mixture was stirred at -78 °C for 4 h and was afterward quenched with cold methanol (3 mL, 0.07 mol). The reaction was allowed to warm toO °C and potassium carbonate (60 mg, 0.0004 mol) and Bestmann-Ohira reagent (1.5 eq, 57 mg) (E. Quesada et al, Tetrahedron, 62 (2006) 6673-6680) were added. The reaction was stirred at room temperature overnight, and then partitioned between ethyl acetate and water. The organic layer was washed with saturated NaCI, dried over MgS04, filtered and concentrated to give the crude product. The crude product was purified using silica gel (EtOAC/Hexane 1:3 to 1:1) to give the desired product, 3-{1-[4-(7-{[2-(trimethylsi-lyl)ethoxy]-methyl}-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]but-3-yn-1-yl}benzonitrile (40 mg of mixture), m/z = 469 (M+1).
Step 2: 3-1-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]but-3-yn-1-ylbenzonitrHe trifluoroacetate [0590] Using a procedure analogous to Example 712, Step 4, the title compound was prepared (4.5 mg, 46%) as an amorphous white solid. 1H NMR (500 MHz, DMSO): δ. 12.5 (b, 1H), 9 (s, 1H), 8.8 (s, 1H), 8.4 (s, 1H), 8 (s, 1H), 7.8 (m 2H), 7.7 (s, 1H), 7.6 (m, 1H), 7 (m, 1H), 5.9 (m, 1H), 3.4 (dd, 1H), 3.2 (dd, 1H), 2.9 (s, 1H), m/z= 339 (M+1).
Example 706: 3-{1-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]but-3-yn-1-yl}benzaldehyde trifluoroacetate [0591]
[0592] Using the procedure of Example 705, the title compound was prepared as a secondary product (4.5 mg, 46%) as an amorphous white solid. 1H NMR (400 MHz, CDCI3): δ 10 (s, 1H), 9 (s, 1H), 8.8 (s, 1H), 8.4 (s, 1 H), 8 (s, 1H), 7.9 (m 1H), 7.8 (m, 1 H), 7.7 s, 1H), 7.6 (m, 1H), 7.1 (s, 1H), 5.9 (m, 1H), 3.4 (dd, 1H), 3.2 (dd, 1H), 2.9 (s, 1H). m/z = 342.
Example 712: 4-[1-(3-Methoxy-1-phenylpropyl)-1H-pyrazol-4-yl]-7H-pyrrolo[2,3-d]pyrimidine trifluoroacetate [0593]
Step 1: Methyl 3-phenyl-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]pro-panoate [0594]
[0595] A solution of methyl (2E)-3-phenylacrylate (500 mg, 0.003 mol) in ACN (2 mL, 0.04 mol) was slowly added to a solution of 4-(1H-pyrazol-4-yl)-7-[2-(trimethylsilyl)ethoxylmethyl-7H-pyrrolo[2,3-d]pyrimidine (0.5 g, 0.002 mol) in ACN (2 mL, 0.04 mol) and DBU (500 μί, 0.003 mol). The reaction was stirred at room temperature over the weekend. The reaction was partitioned between water and EtOAc. The organic layer was washed with saturated sodium chloride, dried over MgS04, filtered and concentrated to give an oil. The product was purified by FCC on silica gel using EtOAc/Hexane (1:2 to 1:1) gave methyl 3-phenyl-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanoate (500 mg, 70%) as a semisolid residue. 1H NMR (400 MHz, CDCI3): δ 8.9 (s, 1H), 8.4 (s, 2H), 7.4 (m, 5H), 6.8 (d, 1H), 6 (m, 1H), 5.7 (s, 2H), 3.7-3.8 (m, 3H), 3.6 (m, 2H), 2.2 (m, 1H), 1.4 (m, 2H), 1.1 (m, 2H), 0.02 (s, 9H), m/z = 478 (M+1).
Step 2: 3-Phenyl-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propan-1-ol [0596]
[0597] Diisobutylaluminum hydride in hexane (1 M, 0.69 mL) was added to a solution of methyl 3-phenyl-3-[4-(7-[2-(tri-methylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propanoate (150 mg, 0.00031 mol) in DCM (3 mL, 0.05 mol) and the mixture was cooled to -78 °C under a nitrogen atmosphere. The reaction was stirred for 1 h at -78 °C and was allowed to warm to room temperature for 4 hrs. The reaction was quenched with methanol (100 μί), and saturated ammonium chloride (100 μί), and then taken up in ethyl acetate dried over MgS04 and filtered. The filtrate was concentrated to give 3-phenyl-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propan-1-ol (130 mg, 92%) as an oil. m/z = 450 (M+1).
Step 3:4-[1-(3-Methoxy-1 -phenylpropyl)-1 H-pyrazol-4-yl]-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidine [0598]
[0599] Sodium hydride (9.6 mg, 0.00040 mol) was added to a solution of 3-phenyl-3-[4-(7-[2-(trimethylsilyl)ethoxy]me-thyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]pxopan-1-ol (120 mg, 0.00027 mol) in DMF (3 mL, 0.04 mol) and the mixture was cooled to 0 °C. The reaction was stirred for 20 min and methyl iodide (22 μί, 0.00035 mol) was added. The reaction was allowed to warm to room temperature and stirred overnight. The reaction was partitioned between water and EtOAc. The organic layer was washed with saturated NaCI, dried over MgS04, filtered and concentrated to give 4-[1-(3-methoxy-1-phenylpropyl)-1H-pyrazol-4-yl]-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidine (100 mg, 88%) as a semisolid, m/z = 464 (M+1).
Step 4: 4-[ 1-(3-Methoxy-1-phenylpropyl)-1H-pyrazol-4-yl]-7H-pyrrolo[2,3-d]pyrimidine trlfluoroacetate [0600] Trifluoroacetic Acid (2 mL, 0.02 mol) was added to a mixture of 4-[1-(3-methoxy-1-phenylpropyl)-1 H-pyrazol-4-yl]-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidine (80 mg, 0.0002 mol) in DCM (3 mL, 0.05 mol) at room temperature. The starting material was consumed after stirring for 2hrs and the reaction solution was concentrated to remove the TFA. The crude reaction was diluted with methanol (3 mL, 0.07 mol) and was treated with ethylenediamine (0.3 mL, 0.004 mol) at room temperature. The reaction mixture was stirred for 18 hs and was concentrated and purified using HPLC on a C-18 column eluting with an ACN: water gradient containing 0.2% TFA, to give the title compound (43 mg, 60%) as a white amorphous solid. 1H NMR (400 MHz, CDCI3): 88.9 (s, 1H), 8.8 (s, 1H), 8.4 (s, 1H), 7.8 (s, 1H), 7.4 (m, 1H), 7.3 (m, 5H), 7.2 (b, 1H), 5.7 (m, 1 H), 3.3 (m, 1H), 3.2 (s, 3H), 2.7 (m, 1H), 2.4 (m, 1H). m/z = 334 (M+1).
Example 715: 3-1-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]but-3-en-1-ylbenzonitrile trifluoroacetate [0601]
[0602] A mixture of [4-1-[1-(3-bromophenyl)but-3-en-1-yl]-1H-pyrazol-4-yl-7H-pyrrolo[2,3-d]-pyrimidine (20 mg, 0.00005 mol) in DMF (2 mL, 0.02 mol) and zinc cyanide (60 mg, 0.0005 mol) was degassed with a nitrogen stream. The mixture was then treated with tetrakis(triphenylphosphine)palladium(0) (40 mg, 0.00003 mol), again degassed with nitrogen, and was then heated in a microwave reactor to 170 °Cfor 15 min. The reaction was allowed to cool, was filtered and purified by HPLC on a C-18 column eluting with an ACN/water/TFA gradient to give the title compound (10 mg, 40%) as a white amorphous solid. 1H NMR (400 MHz, DMSO): δ 8.9 (s, 1H), 8.8 (s, 1H), 8.4 (s, 1H), 7.9 (s, 1H), 7.8 (m, 3H), 7.6 (m, 1H), 7.1 (b, 1H), 5.6-5.8 (m, 2H), 5.1 (d, 1H), 5 (d, 1H), 3.3 (m, 1H), 3 (m, 1H). m/z = 341 (M+1).
Example 716: 4-1-[1-(3-Bromophenyl)but-3-en-1-yl]-1H-pyrazol-4-yl-7H-pyrrolo[2,3-d]-pyrimidine [0603]
Step 1:3-(3-Bromophenyl)-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]pro-panai [0604]
[0605] Diisobutylaluminum hydride in hexane (1 M, 4 mL) was added to a -78 °C solution of ethyl 3-(3-bromophenyl)-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1Hpyrazol-1-yl]propanoate (600 mg, 0.001 mol) in DCM (6 mL, 0.09 mol). After stirring, for 4 h, the reaction was quenched with cold methanol (300 μί), and then saturated ammonium chloride (500 μί) was added and the resulting solution was stirred for 1 h. The reaction was partitioned between water and EtOAc. The organic layer was washed with brine, dried over MgS04, filtered and con centrated. The product was purified by flash chromatography on silica gel eluting with hexane: EtOAc, (2:1 to 1:2), to give 3-(3-bromophenyl)-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]-pyrimidin-4-yl)-1H-pyrazol-1-yl]pro-panai (400 mg, 70%) as an oil.. 1H NMR (400 MHz, CDCI3): δ 9.9 (s, 1H), 8.9 (s, 1H), 8.4 (s, 2H), 7.6 (d, 1H), 7.5 (d, 1H), 7.4 (d, 1H), 7.3-7.4 (m, 2H), 6.8 (d, 1H), 6.1 (m, 1H), 5.7 (s, 2H), 4 (m, 1H), 3.6 (m, 2H), 3.3 (dd, 1H), 1.0 (m, 2H), 0.01 (s, 9H). m/z = 526, 528 (M+1).
Step 2: 4-1 -[1 -(3-Bromophenyl)but-3-en-1 -yl]-1 H-pyrazol-4-yl-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyri-midine [0606]
[0607] Potassium ferf-butoxide in THF (!M, 200 μΙ_) was added to a solution of methyltriphenylphosphonium iodide (80 mg, 0.0002 mol) in THF (2 mL, 0.02 mol) at 0 °C. The reaction was stirred at room temperature for 1h and then cooled to -78 °C. The 3-(3-bromophenyl)-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanal (90 mg, 0.0002 mol) in THF (2 mL, 0.02 mol) was added dropwise. The reaction was allowed to warm to room temperature gradually. The reaction was partitioned between water and EtOAc. The organic layer was washed with saturated NaCI, dried over MgS04, filtered and concentrated to give an oil. The product was purified by FCC on silicagel eluting with EtOAc:Hexane, (1:1) togive4-1-[1-(3-bromophenyl)but-3-en-1-yl]-1H-pyrazol-4-yl-7-[2-(tri-methylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidine (35 mg, 40%) as an oil. m/z = 524, 526 (M+1).
Step 3: 4-1 -[1 -(3-Bromophenyl)but-3-en-1 -yl]-1 H-pyrazol-4-yl-7H-pyrrolo[2,3-d]pyrimidine [0608] Using a procedure analogous to Example 712, Step 4, but using 4-1-[1-(3-bromophenyl)but-3-en-1-yl]-1H-pyrazol-4-yl-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidine the title compound was prepared (10 mg, 30%) as a white amorphous solid, 1H NMR (400 MHz, DMSO): δ 8.9(s,1H), 8.8(s,1H), 8.4(s,1H), 7.8(s,1H), 7.7(s,1H), 7.5 (m,2H), 7.3(m,1H), 7.1(s,1H), 5.7(m,2H), 5.2(d,1H), 5.0(d,1H), 3.2(m,1H), 3.0(m,1H). m/z = 394, 396 (M+1).
Example 717: 3-(4,4-Difluoro)-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]but-3-en-1-ylbenzonitrile [0609]
Step 1: 4-{1-[1-(3-Bromophenyl)-4,4-difluorobut-3-en-1-yl]-1 Hpyrazol-4-yl}-7-{[2-(trimethylsilyl)-ethoxy]methyl}-7H-pyr-rolo[2,3-d]pyrimidine [0610]
[0611] To a solution of3-(3-bromophenyl)-3-[4-(7-[2-(trimethylsilyl)ethoxylmethyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanal (0.05 g, 0.00009 mol) in A/,A/-dimethylacetamide (2 mL, 0.02 mol) was added triphenylphosphine (0.1 g, 0.0006 mol), dibromodifluoromethane (50 uL, 0.0006 mol) and 0.76 M zinc in THF (0.7 mL). The reaction was stirred at room temperature for 18 hs. The reaction was partitioned between water and EtOAc. The organic layer was washed with saturated NaCI, dried over MgS04, filtered and concentrated to give an oil. The product was purified by FCC on silica gel eluting with EtOAc, Hexane (1:2) to give 4-{1 -[1-(3-bromophenyl)-4,4-difluorobut-3-en-1 -yl]-1 H-pyrazol-4-yl}-7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidine (20 mg, 40%) as a clear oil. m/z = 560, 562 (M+1).
Step 2: 4-1-[1 -(3-Bromophenyl)-4,4-difluorobut-3-en-1-yl]-1H-pyrazol-4-yl-7H-pyrrolo[2,3-d]-pyrimidine [0612]
[0613] Using a procedure analogous to Example 712, Step 4, but using 4-{1-[1-(3-bromophenyl)-4,4-difluorobut-3-en-1-yl]-1H-pyrazol-4-yl}-7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidine, the compound 4-1 -[1 -(3-bromophenyl)-4,4-difluorobut-3-en-1-yl]-1H-pyrazol-4-yl-7H-pyrrolo[2,3-d]pyrimidine was prepared (30 mg, 99%) as an oil. m/z = 430, 432 (M+1).
Step 3: 3-4,4-Difluoro-1-[4-(7H-pyrrolo[2,3-d]pynmidin-4-yl)-1H-pyrazol-1-yl]but-3-en-1-yl-benzonitrile [0614]
[0615] A mixture of 4-1-[1-(3-bromophenyl)-4,4-difluorobut-3-en-1-yl]-1H-pyrazol-4-yl-7H-pyrrolo[2,3-d]pyrimidine (30 mg, 0.00007 mol) in DMF (2 mL, 0.02 mol) and zinc cyanide (80 mg, 0.0007 mol) was degassed with nitrogen. The mixture was then treated with tetrakis(triphenylphosphine)palladium(0) (50 mg, 0.00004 mol) and was degassed with nitrogen, and then was heated in microwave at 170 °C for 15 min. The reaction was then allowed to cool, filtered and purified by HPLC on a C-18 column eluting with an ACN/water/TFA gradient to give the title compound (10 mg, 30%) as a white amorphous solid. 1H NMR (400 MHz, DMSO): Ô8.9 (s, 1H), 8.7 (s, 1H), 8.4 (s, 1H), 7.9 (s, 1H), 7.7 -7.8 (m, 3H), 7.5 (m, 1H), 7.1 (m, 1H), 5.7 (m, 1H), 4.3-4.4 (m, 1H), 3.1 (m, 1H), 2.9 (m, 1H). m/z = 377 (M+1).
[0616] The following compounds in Table 14 were prepared as indicated in the column labeled "Prep. Ex. No." and the details of certain exemplary synthetic procedures are provided following Table 14.
Table 14
[0617] * Step 1 of example 731 was modified as follows: The Ph3P and CF2Br2 were combined in DMAC at 0 °C and then allowed to warm to room temperature until the ylid formation was complete as determined by LCMS. The solution of the ylid was then re-cooled to 0 °C and the aldehyde and zinc were added to the ylid solution and the reaction was slowly warmed to room temperature.
Example 727: 4-[1-(1-Cyclopentylbut-3-en-1-yl)-1H-pyrazol-4-yl]-7H-pyrrolo[2,3-d]pyrimidine trifluoroacetate salt [0618]
Step 1: (2E)-3-Cyclopentylacrylic acid [0619] To a solution of malonic acid (1.06 g, 10.2 mol) in pyridine (1.25 mL) was added piperidine (0.15 mL) and cyclopentanecarbaldehyde (1.00 g, 10.2 mmol). The mixture was heated to 40 °C for 2 hours, followed by stirring at room temperature for 16 hours. The mixture was then cooled in an ice bath and 2N HCI was added to acidify. The product was extracted with ether. The ether extract was washed with aq. HCI and brine, dried over sodium sulfate, filtered, and the solvent was removed in vacuo to afford the product (1.30 g, 77%), which was used without further purification. 1H NMR (300 MHz, CDCI3): Ô7.06 (dd, 1H), 5.80 (dd, 1H), 2.70-2.54 (m, 1H), 1.93-1.32 (m, 8H); MS(ES):141(M+H).
Step 2. Methyl (2E)-3-cyclopentylacrylate [0620] To a solution of (2E)-3-cyclopentylacrylic acid (1.3 g, 9.3 mmol) in DCM (65 mL) at 0 °C was added oxalyl chloride (3.1 mL, 37 mmol), dropwise. The resulting solution was stirred at 0 °Cfor40 minutes, then at room temperature for 2 hours. The volatiles were evaporated to afford (2E)-3-cyclopentylacryloyl chloride as a colorless liquid. A portion of this (2E)-3-cyclopentylacryloyl chloride (0.75 g, 4.7 mol) was dissolved in methanol (10 mL) and the resulting solution was stirred for 2 hours. The solvent was evaporated to afford the product (700 mg, 96%). 1H NMR (300 MHz, CDCI3): Ô6.94 (dd, 1H), 5.79 (dd, 1H), 3.71 (s, 3H), 2.66-2.50 (m, 1H), 1.92-1.27 (m, 8H).
Step 3. Methyl 3-cyclopentyl-3-[4-(7-[2-(trimethylsHyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yljpropanoate [0621] To a solution of 4-(1H-pyrazol-4-yl)-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidine (2.9 g, 9.2 mmol) and methyl (2E)-3-cyclopentylacrylate (1.70 g, 11.0 mmol) in ACN (100 mL), was added DBU (2.7 mL, 18 mmol). The resulting mixture was stirred for 96 hours. The ACN was removed in vacuo, and the resulting residue was dissolved in ethyl acetate. This solution was washed with 1.0 N HCI, followed by brine, and then dried over sodium sulfate, and the solvent removed in vacuo. Flash column chromatography (eluting with a gradientfrom 0-70% ethyl acetate in hexanes) afforded the product (2.73 g, 63%). 1H NMR (300 MHz, CDCI3): δ 8.84 (s, 1H), 8.28 (s, 2H), 7.39 (d, 1H), 6.81 (d, 1H), 5.67 (s, 2H), 4.46 (dt, 1H), 3.60 (s, 3H), 3.54 (t, 2H), 3.18 (dd, 1H), 2.89 (dd, 1H), 2.59-2.42 (m, 1H), 1.95-1.80 (m, 1H), 1.75-1.10 (m, 7H), 0.92 (t, 2H), -0.06 (s, 9H); MS(ES):470(M+H).
Step 4. 3-Cyclopentyl-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-y/)-1H-pyrazol-1-yl]propanal [0622] To a solution of methyl 3-cyclopentyl-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo-[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanoate (0.501 g, 1.07 mmol) in THF (5.0 mL) at -78 °C was added 1.00 M diisobutylaluminum hydride in DCM (2.35 mL) dropwise. The reaction was stirred with gradual warming to -10 °C over the course of 2 hours. At this temperature, a further portion of 1.0 M diisobutylaluminum hydride in DCM (1.50 mL) was added. When the reaction was determined to be complete by LCMS, a saturated solution of K/Na tartrate was added, followed by ether. The resulting mixture was stirred for two hours at room temperature. The organic layer was separated and washed with water, and brine, then dried over sodium sulfate and the solvent was removed In vacuo to give a viscous oil, which was used without further purification. MS(ES):442(M+H).
[0623] To a solution of oxalyl chloride (0.108 mL, 1.28 mmol) in DCM (10.0 mL) at -78 °C was added DMSO (151 μί, 2.13 mmol). Afterstirring for 5 minutes, 3-cyclopentyl-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propan-1-ol (471 mg, 1.07 mmol) in DCM (3.00 mL) was added. The mixture was stirred for 30 minutes at -78 °C. TEA (594 μί, 4.26 mmol) was then added. The resulting mixture was then allowed to warm to room temperature over the course of 30 minutes. Water was added, and the layers were separated. The organic layer was washed successively with 0.1 N HCI, water, saturated sodium bicarbonate solution, and brine, and was then dried over sodium sulfate and the solvent was removed in vacuo. Flash column chromatography (eluting with a gradient of 0-60% ethyl acetate in hexanes) afforded the product (384 mg, 82%). 1H NMR (300 MHz, CDCI3): δ 9.73 (s, 1H), 8.87 (s, 1H), 8.71 (br s, 1H), 8.30 (s, 1H), 7.47 (brs, 1H), 6.88 (brs, 1H), 5.69 (s, 2H), 4.66-4.49 (m, 1H), 3.54 (t, 2H), 3.40 (ddd, 1H), 2.95 (ddd, 1H), 2.55-2.44 (m, 1H), 2.01-1.21 (m, 8H), 0.98 (t, 2H), 0.00 (s, 9H); MS(ES):440(M+H).
Step 5. 4-[1-( 1-Cyclopentylbut-3-en-1-yl)-1H-pyrazol-4-yl]-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimi-dine [0624] To a solution of 1.0 M potassium ferf-butoxide in THF (0.207 mL) in THF (2.0 mL) at 0 °C was added triphe-nylmethylphosphonium bromide (77.8 mg, 0.218 mmol). The resulting mixture was warmed to room temperature and allowed to stir for 30 minutes. A solution of 3-cyclopentyl-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyri-midin-4-yl)-1H-pyrazol-1-yl]propanal (0.100 g, 0.228 mmol) in THF (2.0 mL) was added. After 30 minutes, the mixture was quenched by the addition of saturated ammonium chloride solution and the product was then extracted with ether. The ether extract was dried over sodium sulfate and the solvent was removed in vacuo. Flash column chromatography (eluting with a gradient of 0-40% ethyl acetate in hexanes) afforded the product (40 mg, 44%). 1H NMR (400 MHz, CDCI3): δ 8.84 (s, 1H), 8.26 (s, 1H), 8.19 (brs, 1H), 7.40 (s, 1H), 6.83 (brs, 1H), 5.67 (s, 2H), 5.60 (ddt, 1H), 5.01 (dq, 1H), 4.97-4.93 (m, 1H), 3.99 (dt, 1H), 3.54 (t, 2H), 2.79-2.60 (m, 2H), 2.60-2.40 (m, 1H), 1.99-1.89 (m, 1H), 1.75-1.41 (m, 5H), 1.37-1.12 (m, 2H), 0.92 (t, 2H), - 0.06 (s, 9H); MS(ES):438(M+H).
Step 6. 4-[1-(1-Cyclopentylbut-3-en-1-yl)-1Hpyrazol-4-yl]-7H-pyrrolo[2,3-d]pyrimidine trifluoroacetate salt [0625] 4-[1-(1-Cyclopentylbut-3-en-1-yl)-1H-pyrazol-4-yl]-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimi-dine (13 mg, 0.030 mmol) was dissolved in DCM (3 mL) and TFA (0.5 mL) was added. The resulting solution was stirred at room temperature for 3 hours. The solvent was removed in vacuo. The residue was dissolved in THF (2 mL), and 6 N NaOH (1 mL) was added. The mixture was stirred at room temperature for 1 hour, and then was partitioned between water and ethyl acetate. The organic layer was dried over sodium sulfate and the solvent was removed in vacuo. Purification via preparative-HPLC/MS (C18 eluting with a gradient of H20 and ACN containing 0.1% TFA) afforded the product (10 mg, 80%). 1H NMR (400 MHz, d6-DMSO): Ô12.73 (s, 1H), 8.88 (s, 2H), 8.43 (s, 1H), 7.79 (t, 1H), 7.19 (dd, 1H), 5.60 (ddt, 1H), 5.00-4.93 (m, 1H), 4.91-4.87 (m, 1H), 4.23 (dt, 1H), 2.76-2.59 (m, 2H), 2.47-2.34 (m, 1H), 1.92-1.82 (m, 1H), 1.68-1.22 (m, 6H), 1.21-1.09 (m, 1H); MS(ES):308(M+H).
Example 729: 4-[1-(1-Cyclopeutyl-2-cyclopropylethyl)-1H-pyrazol-4-yl]-7H-pyrrolo[2,3-d]-pyrimidine trifluoroacetate salt [0626]
Step 1. 4-[1-(1-Cyclopentyl-2-cyclopropylethyl)-1 H-pyrazol-4-yl]-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]py-rimidlne trlfluoroacetate salt [0627] A solution of 4-[1-(1-cyclopentylbut-3-en-1-yl)-1 H-pyrazol-4-yl]-7-[2-(trimethylsilyl)-ethoxy]methyl-7H-pyiro-lo[2,3-d]pyrimidine (prepared in Example 727, Step 5) (54.0 mg, 0.123 mmol) in DCM (1 mL) was added to a freshly prepared ethereal solution of excess CH2N2 held at 0 °C. Palladium acetate (10.0 mg, 0.044 mol) was added. After standing for 2 hours, the excess CH2N2 was quenched by the addition of acetic acid. The solution was then diluted with further DCM, washed successively with saturated sodium bicarbonate solution, water, and brine, and dried over sodium sulfate, and the solvent was removed In vacuo. Purification via preparative-HPLC/MS (C18 eluting with a gradient of H20 and ACN containing 0.1% TFA) afforded the product (13 mg, 18%). 1H NMR (300 MHz, CDCI3): δ 9.05 (s, 1H), 8.81 (d, 1H), 8.35 (s, 1H), 7.59 (t, 1H), 7.03 (t, 1H), 5.76 (s, 2H), 4.10 (t, 1H), 3.59 (t, 2H), 2.57-2.36 (m, 1H), 2.15-2.00 (m, 1H), 2.00-1.83 (m, 1H), 1.79-1.40 (m, 6H), 1.37-1.09 (m, 2H), 0.97 (t, 2H), 0.55-0.26 (m, 3H), 0.07- -0.15 (m, 11H); MS(ES):452(M+H).
Step 2. 4-[1-(1-Cyclopentyl-2-cyclopropylethyl)-1 H-pyrazol-4-yl]-7H-pyrrolo[2,3-d]pyrimidine trifluoroacetate salt [0628] 4-[1-(1-Cyclopentyl-2-cyclopropylethyl)-1 H-pyrazol-4-yl]-7-[2-(trimethylsilyl)ethoxy]-methyl-7H-pyrrolo[2,3-d]pyrimidine trifluoroacetate salt (13 mg, 0.023 mol) was stirred at room temperature in a solution of DCM (2 mL) containing TFA (1.5 mL) for two hours. The solvent was removed in vacuo. The resulting residue was redissolved in THF (3 mL), and 6N NaOH (2 mL) was added. After stirring for one hour, the mixture was partitioned between water and ethyl acetate. The organic layer was dried over sodium sulfate and the solvent was removed in vacuo. Purification via preparative-HPLC/MS (C18 eluting with a gradient of H20 and ACN containing 0.1% TFA) afforded the product (9 mg, 90%). 1H NMR (400 MHz, d6-DMSO): δ 12.75 (s, 1H), 8.90 (s, 1H), 8.84 (s, 1H), 8.47 (s, 1H), 7.81 (s, 1H), 7.22 (s, 1H), 4.19 (dt, 1H), 2.43-2.29 (m, 1H), 2.03-1.92 (m, 1H), 1.88-1.76 (m, 1H), 1.68-1.37 (m, 5H), 1.35-1.08 (m, 3H), 0.43-0.26 (m, 2H), 0.24-0.13 (m, 1H), 0.07--0.03 (m, 1H), -0.14--0.24 (m, 1H); MS(ES):322(M+H).
Example 730: 4-[1-(1-Cyclopentylbut-3-yn-1-yl)-1H-pyrazol-4-yl]-7H-pyrrolo[2,3-d]pyrimidine trifluoroacetate salt [0629]
Step 1. 4-[1-(1-Cyclopentylbut-3-yn-1-yl)-1H-pyrazol-4-yl]-7-[2-(trimethylsHyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidine [0630] To a mixture of potassium carbonate (38.4 mg, 0.278 mmol) in methanol (2.0 mL) at 0 °C was added a solution of 3-cyclopentyl-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]-pyrimidin-4-yl)-1H-pyrazol-1-yl]propanal (prepared as in Example 727, step 4) (61.0 mg, 0.139 mmol) in methanol (1.0 mL), followed by a solution of dimethyl (1-diazo-2-oxopropyl)phosphonate (40.0 mg, 0.208 mmol) in methanol (1.0 mL). The mixture was slowly warmed to ambient temperature and stirred for 16 hours. The mixture was then diluted with water and extracted with ethyl acetate. The combined extracts were washed with water, saturated ammonium chloride, and then dried over sodium sulfate and the solvent was removed in vacuo to afford the product, which was used without further purification (52 mg, 86%). 1H NMR (300 MHz, CDCI3): δ 8.85 (s, 1H), 8.47 (s, 1H), 8.29 (s, 1H), 7.41 (d, 1H), 6.84 (d, 1H), 5.67 (s, 2H), 4.14 (ddd, 1H), 3.53 (t, 2H), 2.90 (ddd, 1H), 2.79 (ddd, 1H), 2.66-2.49 (m, 1H), 1.98 (t, 1H), 2.00-1.88 (m, 1H), 1.78-1.44 (m, 5H), 1.39-1.11 (m, 2H), 0.92 (t, 2H), -0.06 (s, 9H); MS(ES):436(M+H).
Step 2. 4-[1-(1-Cyclopentylbut-3-yn-1-yl)-1H-pyrazol-4-yl]-7H-pyrrolo[2,3-d]pyrimidine trifluoroacetate salt [0631] A solution of 4-[1-(1-cyclopentylbut-3-yn-1-yl)-1H-pyrazol-4-yl]-7-[2-(trimethylsilyl)-ethoxy]methyl-7H-pyrro-lo[2,3-d]pyrimidine (52 mg, 0.12 mmol) in DCM (3 mL) and TFA (1 mL) was stirred for 2 hours. The solvents were removed in vacuo. The resulting residue was dissolved in THF (3 mL) and 6N NaOH (2 mL) was added. After stirring for 1 hour, the mixture was partitioned between water and ethyl acetate. The organic layer was dried over sodium sulfate and the solvent was removed in vacuo. Purification via preparative-HPLC/MS (C18 eluting with a gradient of H20 and ACN containing 0.1% TFA) afforded product (30 mg, 60%). 1H NMR (300 MHz, d6-DMSO): δ 12.72 (s, 1H), 8.91 (s, 1H), 8.84 (s, 1H), 8.47 (s, 1H), 7.80 (s, 1H), 7.19 (s, 1H), 4.34 (dt, 1H), 2.97-2.69 (m, 3H), 2.50-2.32 (m, 1H), 1.93-1.77 (m, 1H), 1.70-1.09 (m, 7H); MS(ES):306(M+H).
Example 731: 4-[1-(1-Cyclopentylbutyl)-1H-pyrazol-4-yl]-7H-pyrrolo[2,3-d]pyrimidine trifluoroacetate salt [0632]
[0633] 4-[1-(1-Cyclopentylbut-3-yn-1-yl)-1H-pyrazol-4-yl]-7H-pyrrolo[2,3-d]pyrimidine trifluoroacetate salt (prepared in Example 729) (20 mg, 0.048 mmol) was dissolved in methanol (2 mL) and a catalytic amount of 5% Pd-C was added. The mixture was stirred under 1 atmosphere of hydrogen via an affixed balloon. After 2 hours, the mixture was filtered and purified via preparative-HPLC/MS (C18 eluting with a gradient of H20 and ACN containing 0.1% TFA) to afford the product (14 mg, 69%). 1H NMR (400 MHz, d6-DMSO): δ 12.73 (s, 1H), 8.86 (s, 1H), 8.83 (s, 1H), 8.45 (s, 1H), 7.79 (t, 1H), 7.20 (d, 1H), 4.11 (dt, 1H), 2.43-2.26 (m, 1H), 2.02-1.70 (m, 3H), 1.68-1.35 (m, 4H), 1.33-0.89 (m, 5H), 0.83 (t, 3H); MS(ES):310(M+H).
Example 732: 4-[1-(1-Cyclopentyl-4,4-difluorobut-3-en-1-yl)-1H-pyrazol-4-yl]-7H-pyrrolo[2,3-d]pyrimidine trifluoroacetate salt [0634]
Step 1. 4-[1 -(1 -Cyclopentyl-4,4-dlfluorobut-3-en-1-yl)-1H-pyrazol-4-yl]-7-[2-(trimethylsilyl)ethoxy]-methyl-7H-pyrro-lo[2,3-d]pyrimidine [0635] To a solution of 3-cyclopentyl-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]-pyrimidin-4-yl)-1H-pyrazol-1-yl]propanal (prepared as in Example 727, Step 4) (181 mg, 0.41 mmol) in A/,A/-dimethylacetamide (3.6 mL) was added triphenylphosphine (294 mg, 1.12 mmol) followed by dibromodifluoromethane (235 mg, 1.12 mmol). Rieke® Zinc (1.8 mL of a suspension of 2.5 g in 50 ml THF) was then added in one portion. The resulting mixture was stirred at room temperature for 4.5 hours. The mixture was filtered through diatomaceous earth. The filtrate was partitioned between ether and water. The ether layer was washed with water, and brine, then dried over sodium sulfate, and the solvent was removed In vacuo. Flash column chromatography (eluting with a gradient from 0-30% ethyl acetate in hexanes) afforded product (104 mg, 53%). 1H NMR (400 MHz, CDCI3): δ 8.91 (s, 1H), 8.51 (brs, 1H), 8.34 (s, 1H), 7.51 (d, 1 H), 6.93 (d, 1H), 5.74 (s, 2H), 4.05 (ddd, 1H), 4.04-3.96 (m, 1H), 3.60 (t, 2H), 2.78-2.62 (m, 2H), 2.58-2.45 (m, 1H), 2.07-0.87 (m, 10H), 0.00 (s, 9H); MS(ES):474(M+H).
Step 2. 4-[ 1-( 1-Cyclopentyl-4,4-difluorobut-3-en-1-yl)-1H*pyrazol-4-yl]-7H-pyrrolo[2,3-d]pyrimidine trifluoroacetate salt [0636] A solution of 4-[1-(1-cyclopentyl-4,4-difluorobut-3-en-1-yl)-1H-pyrazol-4-yl]-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidine (41 mg, 0.086 mmol) in DCM (3 mL) and TFA (1.5 mL) was stirred for two hours at room temperature. The solution was then concentrated in vacuo. The resulting residue was redissolved in THF (3 mL), and 6N NaOH (2 mL) was added. After stirring for 1 hour, the mixture was partitioned between water and ethyl acetate. The organic layer was dried over sodium sulfate and the solvent was removed in vacuo. Purification via preparative-HPLC/MS (C18 eluting with a gradient of H20 and ACN containing 0.1% TFA) afforded the desired product (39 mg, 98%). 1H NMR (400 MHz, d6-DMSO): δ 12.72 (s, 1H), 8.84 (s, 1H), 8.83 (s, 1H), 8.45 (s, 1H), 7.80 (t, 1H), 7.18 (d, 1H), 4.32 (ddt 1H), 4.20 (dt, 1H), 2.72-2.37 (m, 3H), 1.95-1.81 (m, 1H), 1.69-1.06 (m, 7H); MS(ES):344(M+H).
[0637] Where conjugate acceptors, such as were used in Example 737, Step 3 were not commercially available, such compounds were generated according to the procedure provided below for ethyl (2E)-3-(tetrahydrofuran-3-yl)acrylate (toward the preparation of Example 733).
Preparation of ethyl (2E)-3-(tetrahydrofuran-3-yl)acrylate: [0638]
Step A: Tetrahydrofuran-3-carbaldehyde [0639] To a solution of Dess-Martin periodinane (3.37 g, 7.95 mmol) in DCM (20 mL) was added tetrahydrofuran-3-ylmethanol (0.701 mL, 7.23 mmol). The reaction was stirred at ambient temperature for 2 hours, and the solvent was then removed in vacuo. Flash column chromatography (using DCM as eluent) afforded the product as a clear oil, which was used without further purification. 1H NMR (400 MHz, CDCI3): δ 9.65 (d, 1H), 4.12-4.07 (m, 1H), 3.92-3.85 (m, 2H), 3.80-3.73 (m, 1H), 3.10-3.02 (m, 1H), 2.26-2.10 (m, 2H).
Step B: Ethyl (2E)-3-(tetrahydrofuran-3-yl)acrylate [0640] To a 0 °C mixture of sodium hydride (60% in mineral oil) (382 mg, 9.40 mmol) in DMF (15.0 mL) (THF may also be used) was added triethyl phosphonoacetate (1.72 mL, 8.68 mmol) dropwise. The resulting mixture was warmed to room temperature and stirred for 30 minutes, then was re-cooled to 0 °C, at which time a solution of tetrahydrofuran-3-carbaldehyde (724 mg, 7.23 mmol) in DMF (4.0 mL) was added dropwise. The resulting mixture was stirred at this temperature for 1.5 hours, at which time the mixture was diluted with water and the product was extracted with ether. The combined extracts were washed with water and brine, dried over sodium sulfate and the solvent removed in vacuo. Flash column chromatography (eluting with a gradient from 0-40% ethyl acetate in hexanes) afforded the product (640 mg, 52%). 1H NMR (400 MHz, CDCI3): δ 6.87 (dd, 1H), 5.86 (dd, 1H), 3.96-3.88 (m, 2H), 3.81 (dd, 1H), 3.53 (dd, 1H), 3.04-2.93 (m, 1H), 2.20-2.10 (m, 1H), 2.03 (s, 3H), 1.79 (dq, 1H).
Example 736: 4-[1-(1-Cyclopentyl-4,4-difluorobutyl)-1H-pyrazol-4-yl]-7H-pyrrolo[2,3-d]-pyrimidine trifluoroace-tate salt [0641]
[0642] 4-[1-(1-Cyclopentyl-4,4-difluorobut-3-en-1-yl)-1H-pyrazol-4-yl]-7H-pyrrolo[2,3-d]-pyrimidine trifluoroacetate salt (prepared as in Example 731) (20.0 mg, 0.041 mmol) was dissolved in methanol (3 mL), and a catalytic amount of 5% Pd on C was added. The mixture was stirred at room temperature for 2 hours, under an atmosphere of hydrogen provided by an affixed balloon. The mixture was filtered and purified via preparative-HPLC/MS (C18 eluting with a gradient of H20 and ACN containing 0.1% TFA) to afford product (4 mg, 21%). 1H NMR (400 MHz, d6-DMSO): δ 12.74 (s, 1H), 8.88 (s, 1H), 8.85 (s, 1H), 8.48 (s, 1H), 7.80 (t, 1H), 7.20 (dd, 1H), 6.05 (tt, 1H), 4.17 (dt, 1H), 2.47-2.34 (m, 1H), 2.14-1.08 (m, 12H); MS(ES):346(M+H).
Example 737: 3-(1-Methylcyclopentyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile trifluoroacetate salt [0643]
Step 1. 1-Methylcyclopentanecarbaldehyde [0644] To a solution of cyclopentanecarbaldehyde (1.00 mL, 9.36 mmol) in DCM (47 mL) at 0 °C was added solid potassium ferf-butoxide (1.44 g, 12.2 mmol) in one portion followed by methyl iodide (1.7 mL, 28 mmol) in one portion. After 30 minutes at 0 °C, the reaction mixture was allowed to warm to room temperature and stirred at that temperature for 16 hours. The mixture was poured into brine, and the layers were separated. The organic layer was dried over sodium sulfate, decanted and concentrated, and used without further purification in Step 2.
Step 2: (2Z)- and (2E)-3-(1-Methylcyclopentyl)acrylonitrile [0645] To a solution of 1.0 M potassium ferf-butoxide in THF (9.36 mL) at 0 °C was added a solution of diethyl cyanomethylphosphonate (1.59 mL, 9.81 mmol) in THF (10 mL) dropwise. The cooling bath was removed and the reaction was warmed to room temperature followed by re-cooling to 0 °C, at which time a solution of 1-methylcyclopen-tanecarbaldehyde (1.0 g, generated in Step 1) in THF (2 mL) was added dropwise. The bath was removed and the reaction was stirred at ambient temperature for 3 hours. To the mixture was added water and ethyl ether. The aqueous layer was further extracted with ethyl ether. The combined extracts were washed with brine, dried over sodium sulfate, filtered and adsorbed onto silica gel in vacuo. Flash column chromatography (eluting with a gradient from 0-10% ethyl acetate in hexanes) afforded product as a mixture with hexanes, which product was used without further purification in Step 3.
Step 3: 3-( 1-Methylcyclopentyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1 -yl]propane-nitrile trifiuoroacetate salt [0646] To a mixture of 4-(1H-pyrazol-4-yl)-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidine (0.134 g, 0.426 mmol) in ACN (3 mL) was added a mixture of(2Z)- and (2E)-3-(1-methylcyclopentyl)acrylonitrile(0.12g, 0.9 mmol) followed by DBU (0.13 mL, 0.90 mmol). The reaction was heated to 60 °C for 6 h. The ACN was removed in vacuo. Ethyl acetate was added, followed by 0.1 N HCI. The aqueous layer was extracted with three portions of ethyl acetate. The combined organic extracts were washed with brine, dried oversodium sulfate, filtered and the solventwas evaporated. The crude material was deprotected by stirring with TFA (2 mL) in DCM (8 mL) for 2 hours. The solvent and TFA were removed in vacuo. THF (8 mL) was used to dissolve the residue, and 6.0 M sodium hydroxide in water (8 mL) was added. The reaction was stirred in this basic mixture for 2 hours. Ethyl acetate was used to extract the product. The combined extracts were dried (Na2S04) and the solventwas removed in vacuo. Purification via preparative-HPLC/MS (C18 eluting with a gradient of H20 and ACN containing 0.1 % TFA) afforded product (44 mg, 24%). 1H NMR (400 MHz, d6-DMSO): δ 12.71 (s, 1H), 9.00 (s, 1H), 8.85 (s, 1H), 8.51 (s, 1 H), 7.81 (s, 1H), 7.18 (s, 1H), 4.72 (dd, 1H), 3.47 (dd, 1H), 3.21 (dd, 1H), 1.74-1.51 (m, 6H), 1.44-1.32 (m, 1H), 1.09-1.00 (m, 1H), 0.97 (s, 3H); MS(ES):321(M+H).
Example 739: 1-2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethylcyclo-propanecarbonitrile trifiuoroacetate salt [0647]
Stepl: 1-(Hydroxymethyl)cyclopropanecarbonitrile [0648] Ethyl 1-cyanocyclopropanecarboxylate (801 mg, 5.76 mmol) in THF (12.0 mL) was treated with lithium tetrahy-droborate (251 mg, 11.5 mmol). The solution was heated to reflux for 1.5 hours. Upon cooling to room temperature, the reaction was quenched with water, and extracted with ethyl acetate. The combined extracts were dried over MgS04, filtered and concentrated to afford a clear oil, which was used without further purification in the following step (482 mg, 86%). 1H NMR (400 MHz, CDCI3): δ 3.61 (s, 2H), 1.27 (dd, 2H), 0.98 (dd, 2H).
Step2: 1 -Formylcyclopropanecarbonitrile [0649] Dess-Martin periodinane (1.11 g, 2.62 mmol) was dissolved in DCM (12 mL) and 1-(hydroxymethyl)cyclopro-panecarbonitrile (231 mg, 2.38 mmol) was added. The reaction was stirred at ambient temperature for one hour. The mixture was then purified by flash column chromatography (eluting with a gradient from 0-80% ethyl acetate in hexanes) to afford the product (106 mg, 46%). 1H NMR (400 MHz, CDCI3): δ 9.35 (s, 1H), 1.79-1.74 (m, 4H).
Step 3: 1-[(E)-2-Cyanovinyl]cyclopropanecarbonitrile [0650] To a solution of 1.0 M potassium fe/f-butoxide in THF (1.12 mL) at 0 °C was added slowly dropwise a solution of diethyl cyanomethylphosphonate (210 mg, 1.2 mmol) in THF (2 mL). The cold bath was removed and the reaction was warmed to ambient temperature. The solution was then re-cooled to 0 °C and a solution of 1-formylcyclopropanecarbonitrile (101 mg, 1.06 mmol) in THF (1.0 mL) was added dropwise. The cold bath was removed and the reaction was stirred for 3 hours at ambient temperature. The mixture was then diluted with ether and water, the ether solution was separated, washed with brine, dried over sodium sulfate, filtered and the solvent was removed in vacuo. Flash column chromatography (eluting with a gradient from 0-60% ethyl ether in hexanes) afforded the product (24 mg, 19%). 1H NMR (400 MHz, CDCI3): δ 5.94 (d, 1H), 5.82 (d, 1H), 1.80 (dd, 2H), 1.39 (dd, 2H).
Step 4: 1-2-Cyano-1-[4-(7-[2-(trimethylsHyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1 -yl]ethylcyclo- propanecarbonitrile [0651] To a solution of 4-(1H-pyrazol-4-yl)-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]-pyrimidine (61.4 mg, 0.195 mmol) and 1-[(E)-2-cyanovinyl]cyclopropanecarbonitrile (23 mg, 0.19 mmol) in ACN (2 mL) was added DBU (58 uL, 0.39 mmol) and the resulting mixture was stirred for 16 hours. The ACN was evaporated, and the residue was dissolved in ethyl acetate. This solution was washed with 1.0 N HCI, water, and brine, and dried over sodium sulfate, and the solvent removed in vacuo. Flash column chromatography (eluting with a gradient from 0-80% ethyl acetate in hexanes) afforded the product (49 mg, 58%). 1H NMR (400 MHz, CDCI3): δ 8.85 (s, 1H), 8.43 (s, 1H), 8.34 (s, 1H), 7.43 (d, 1H), 6.80 (d, 1H), 5.68 (s, 2H), 3.54 (dd, 1H), 3.51 (dd, 1H), 3.36 (dd, 1H), 1.62 (ddd, 1H), 1.45 (ddd, 1H), 1.34 (ddd, 1H), 1.25 (ddd, 1H), 0.92 (t, 2H),-0.06 (s, 9H); MS(ES):434(M+H).
Step 5: 1 -2-Cyano-1 -[4-(7H-pyrroio[2,3-d]pyrimidin-4-yi)-1 Hpyrazoi-1 -yijethyicyciopropane-carbonitriie trifluoroacetate salt [0652] 1-2 -cyano-1-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethylcyclo-propanecarbonitrile (48 mg, 0.11 mmol) was stirred in a mixture of DCM (3 mL) and TFA(2 mL) for 3 hours. The solvents were removed in vacuo and the residue was redissolved in THF (3 mL). 6N NaOH (2 mL) was added and the resulting mixture was stirred at ambient temperature for 3 hours. The crude reaction mixture was partitioned between ethyl acetate and water. The layers were separated and the organic layer was dried over sodium sulfate and the solvent was removed in vacuo. Purification via preparative-HPLC/MS (C18 eluting with a gradient of H20 and ACN containing 0.1 % TFA) afforded product (20 mg, 43%). 1H NMR (400 MHz, d6-DMSO): 512.74 (s, 1H), 8.99 (s, 1H), 8.88 (s, 1H), 8.60 (s, 1H), 7.83 (t, 1H), 7.17 (dd, 1H), 4.55 (dd, 1H), 3.66 (dd, 1H), 3.54 (dd, 1H), 1.55-1.30 (m, 4H); MS(ES):304(M+H).
Example 740: N-[(1-2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl-cyclopentyl)me-thyl]benzamide [0653]
Step 1: Methyl 1-cyanocyclopentanecarboxylate [0654] To a solution of acetic acid, cyano-, methyl ester (2.66 mL, 30.3 mmol) and 1,4-dibromobutane, (3.62 mL, 30.3 mmol) in acetone (50 mL) was added potassium carbonate (8.37 g, 60.6 mmol). The reaction was stirred at ambient temperature for 16 hours. The reaction was filtered through diatomaceous earth and concentrated. The resulting residue was partitioned between ether and saturated NH4CI solution, and the aqueous layer was extracted with two further portions of ether. The combined ethereal extracts were washed with brine, and dried over sodium sulfate, then filtered and the solvent was removed in vacuo. Flash column chromatography (eluting with a gradient from 0-30% ethyl acetate in hexanes) afforded the product (2.92 g, 63%). 1H NMR (300 MHz, CDCI3): Ô3.82 (s, 3H), 2.30-2.21 (m, 4H), 1.93-1.82 (m, 4H).
Step 2: Methyl 1-[(tert-butoxycarbonyl)amino]methylcyclopentanecarboxylate [0655] To a solution of methyl 1-cyanocyclopentanecarboxylate (1.26 g, 8.22 mmol) in methanol (100 mL) was added cobalt dichloride (2.1 g, 16.0 mmol). The purple mixture was cooled in an ice-water bath. Sodium tetrahydroborate (3.11 g, 82.2 mmol) was added portionwise with caution (exothermic) to provide a black mixture. Upon complete addition, cooling was discontinued and the reaction was stirred for 40 minutes under nitrogen and the reaction was quenched by the careful addition of 1N HCI (700 ml). The methanol was removed in vacuo, and the solution was then made alkaline (pH ~ 9) by the addition of concentrated NH4OH(aq). The mixture was extracted with DCM (6 times), and the combined DCM extracts were dried over sodium sulfate and concentrated to afford the crude product as a light yellow oil. To this crude amine in DCM (50 ml) was added di-tert-butyldicarbonate (1.31 g, 6.01 mmol) and the reaction was stirred at 25 °C for 30 minutes. The reaction was diluted with water and extracted with ethyl acetate three times. The combined extracts were dried over sodium sulfate, filtered, and the solvent removed in vacuo. The crude residue was purified by flash column chromatography to yield the desired product (1.5 g, 71 %). 1H NMR (300 MHz, CDCI3): δ 5.03 (s, 1H), 3.69 (s, 3H), 3.26 (d, 2H), 2.02-1.33 (m, 17H).
Step 3: tert-Butyl [1-(hydroxymethyl)cyclopentyl]methylcarbamate [0656] To a solution of methyl 1-[(ferf-butoxycarbonyl)amino]methylcyclopentanecarboxylate (1.50 g, 5.83 mmol) in THF (25.0 mL) at -78 °C was added dropwise 1.0 M diisobutylaluminum hydride in DCM (17.5 mL). The reaction was stirred for 2 hours with slow warming to -10 °C. A saturated solution of K/Na tartrate was added, followed by ether. This mixture was stirred for 30 minutes at ambient temperature and the organic layer was separated and washed with water, and brine. The organic layer was then dried over sodium sulfate, and the solvent was removed in vacuo to afford the product (1.03 g, 77%). 1H NMR (300 MHz, CDCI3):ô4.90(brs, 1H),3.27 (s,2H), 3.06 (d,2H), 1.5-1.17 (m, 8H), 1.44 (s, 9H).
Step 4: tert-Butyl [(1 -formylcyclopentyl)methyl]carbamate [0657] To a solution of oxalyl chloride (456 μί, 5.38 mmol) in DCM (30.0 mL) at -78 °C was added DMSO (637 μί, 8.97 mmol) and the resulting mixture was stirred for 5 minutes. ferf-Butyl [1-(hydroxymethyl)cyclopentyl]methylcarbamate (1.03 g, 4.48 mmol) in DCM (10.0 mL) was added and the resulting mixture was stirred for 30 minutes at -78 °C. TEA (2.50 mL, 17.9 mmol) was added and the resulting mixture was allowed to warm to ambient temperature over 30 minutes. Water was added. The organic phase was washed sequentially with 0.1 N HCI, water, saturated sodium bicarbonate solution, and brine, and then dried over sodium sulfate and the solvent was removed in vacuo to afford the product (957 mg, 94%). 1H NMR (300 MHz, CDCI3): δ 9.39 (s, 1H), 4.94 (brs, 1H), 3.25 (d, 2H), 1.89-1.46 (m, 8H), 1.41 (s, 9H).
Step 5: tert-Butyl (1-[(E)-2-cyanovinyl]cyclopentylmethyl)carbamate and tert-butyl (1-[(Z)-2-cyanovinyl]cyclopentylme-thyl)carbamate [0658] To a solution of 1.0 M potassium ferf-butoxide in THF (4.4 mL) at 0 °C was added a solution of diethyl cy-anomethylphosphonate (820 mg, 4.6 mmol) in THF (6.0 mL) dropwise. The cold bath was removed and the reaction was warmed to ambient temperature. The mixture was then re-cooled to 0 °C and a solution of ferf-butyl [(1-formylcy-clopentyl)methyl]carbamate (952 mg, 4.19 mmol) in THF (4.0 mL) was added dropwise. The reaction was allowed to warm to ambient temperature and the warmed mixture was stirfor 16 hours. The reaction mixture was then diluted with ether and water. The organic layer was separated and washed sequentially with water and brine, then dried over sodium sulfate, then filtered, and the solvent was removed in vacuo to afford the product (1.05 g, 99%) as a mixture of (£) and (Z) isomers. 1H NMR (300 MHz, CDCI3): δ 6.71 (d, 1H, £), 6.46 (d, 1H, Z), 5.36 (d, 1H, Z), 5.36 (d, 1H, E), 4.70 (br s, 1H, Z), 4.51 (br s, 1H, £), 3.25 (d, 2H, Z), 3.18 (d, 2H, E), 1.88-1.48 (m, 8H (£) and 8H (Z)), 1.43 (s, 9H (£) and 9H (Z)); MS(ES):151 (M+H-Boc).
Step 6: tert-Butyl [( 1-2-cyano-1-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethylcyclopentyl)methyl]carbamate [0659] To a solution of 4-(1H-pyrazol-4-yl)-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]-pyrimidine (355 mg, 1.12 mmol) and tert-butyl (1-[(E)-2-cyanovinyl]cyclopentylmethyl)carbamate and tert-butyl (1-[(Z)-2-cyanovinyl]cy-clopentylmethyl)carbamate as a mixture of isomers (329 mg, 1.31 mmol) in ACN (10 mL) was added DBU (0.168 mL, 1.12 mmol). The resulting mixture was stirred at ambient temperature for 3 hours followed by heating to 60 °C for 2.5 hours. The ACN was removed in vacuo and the resulting residue was purified by flash column chromatography (eluting with 0-55% ethyl acetate in hexanes) to afford the product (350 mg, 55%). 1H NMR (300 MHz, CDCI3): δ 8.85 (s, 1H), 8.37 (brs, 1H), 8.34 (s, 1H), 7.41 (d, 1H), 6.82 (d, 1H), 5.68 (s, 2H), 5.37 (br s, 1H), 4.52 (dd, 1H), 3.54 (t, 2H), 3.40 (dd, 1H), 3.23 (dd, 1H), 3.08 (d, 1H),2.90 (dd, 1H), 1.84-1.47 (m, 8H), 1.45 (s, 9H), 0.92 (t, 2H), -0.06 (s, 9H); MS(ES):566(M+H).
Step 7: N-[( 1-2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethylcyclopentyl)-methyl]benzamide [0660] A solution of tert-butyl [(1-2-cyano-1-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidm-4-yl)-1H-pyrazol-1-yl]ethylcyclopentyl)methyl]carbamate (175 mg, 0.309 mmol) in DCM (5 mL) and TFA (5 mL) was stirred for 3 hours and the solvents were then removed in vacuo. The resulting residue was stirred in a mixture of THF (3 mL) and 6N NaOH (3 mL)for3 hours. The THF was removed in vacuo, and water (10 mL) was added. The mixture was extracted with several portions of DCM containing 15% isopropanol. The combined extracts were dried over sodium sulfate and the solvents were removed in vacuo to afford the product, which was used without further purification. MS(ES):336(M+H).
[0661] To a solution of 3-[1-(aminomethyl)cyclopentyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]pro-panenitrile (31 mg, 0.060 mmol) and benzoyl chloride (7.0 μί, 0.060 mol) in DCM (1.0 mL), was added TEA (17 μί, 0.12 mmol). After 15 minutes, the solvent was removed in vacuo and the mixture was purified via preparative-HPLC/MS (C18 eluting first with a gradient of H20 and ACN containing 0.1% TFA, followed by chromatographic purification, eluting with a gradient of H20 and ACN containing 0.15% NH4OH) to afford the product (7 mg, 27%). 1H NMR (400 MHz, d6-DMSO): Ô12.12 (s, 1H), 8.95 (s, 1H), 8.68 (s, 1H), 8.55 (s, 1H), 8.41 (s, 1H), 7.92-7.87 (m, 2H), 7.60 (d, 1H), 7.59-7.48 (m, 3H), 7.02 (d, 1H), 4.83 (dd, 1H), 3.52-3.45 (m, 2H), 3.42 (dd, 1H), 3.27 (dd, 1H), 2.06-1.95 (m, 1H), 1.68-1.12 (m, 7H); MS(ES):440(M+H).
Example 741: 3-1-[(Benzyloxy)methyl]cyclopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]pro-panenitrile trifluoroacetate salt [0662]
Step 1:1-(Hydroxymethyl)cyclopentanecarbonitrile [0663] A mixture of methyl 1 -cyanocyclopentanecarboxylate (prepared in Example 740, Step 1) (500 mg, 3.0 mmol) in THF (7 mL) was treated with lithium tetrahydroborate (100 mg, 6.0 mmol). The resulting solution was heated to reflux for 3 hours, then stirred at ambient temperature for 16 hours. The mixture was quenched by the addition of water, and was extracted with ethyl acetate. The combined organic extracts were dried over Na2S04, then filtered and the solvent was removed in vacuo to afford the product (387 mg, 95%). 1H NMR(300 MHz, CDCI3): 63.62 (s, 2H), 2.39-1.60 (m, 8H).
Step 2:1-[(Benzyloxy)methyl]cyclopentanecarbonitrile [0664] To a solution of 1-(hydroxymethyl)cyclopentanecarbonitrile (0.30g, 2.0 mmol) in DMF (4 mL) was added sodium hydride (60% dispersion in mineral oil, 0.101 g, 2.52 mol). The resulting mixture was stirred for 20 minutes, followed by the addition of benzyl bromide (0.28 mL, 2.4 mmol). The reaction was stirred at ambient temperature for 64 hours. Additional sodium hydride (60% dispersion in mineral oil, 0.060 g, 1.5 mmol) and benzyl bromide (0.18 mL, 1.5 mmol) were added and the reaction was stirred for an additional 30 minutes. Water was then added to the mixture, followed by brine, and the aqueous layer was extracted with ethyl acetate. The extracts were combined and dried over sodium sulfate, and the solvent was then removed in vacuo. To the resulting residue was added water. The product was isolated by extraction with diethyl ether. The ethereal extracts were dried over sodium sulfate, and the solvent was evaporated. Flash column chromatography (eluting with a gradient from 0-30% ethyl acetate in hexanes) afforded product (330 mg, 64%). 1H NMR (300 MHz, CDCI3): 67.40-7.27 (m, 5H), 4.62 (s, 2H), 3.44 (s, 2H), 2.18-2.03 (m, 2H), 1.90-1.62 (m, 6H).
Step 3: 1-[(Benzyloxy)methyl]cyclopentanecarbaldehyde [0665] To a mixture containing 1-[(benzyloxy)methyl]cyclopentanecarbonitrile (0.16 g, 0.75 mmol) in toluene (5 mL) at 0 °C was added 1.0 M diisobutylaluminum hydride in hexanes (0.8 mL). The reaction was stirred at 0 °C for 1.5 hours, during which time the starting nitrile was consumed. The reaction was cooled to -78 °C and quenched by the addition of methanol. The mixture was warmed to ambient temperature and 3 N HCI was added. Following stirring for 45 minutes, solid NaCI was added, and the mixture was extracted with three portions of ethyl acetate. The combined extracts were dried (Na2S04), and filtered, and the solvent was removed in vacuo. Flash column chromatography of the resulting residue (eluting with a gradient from 0-30% ethyl acetate in hexanes) afforded the product (20 mg, 12%). 1H NMR (300 MHz, CDCI3): δ 9.60 (s, 1H), 7.38-7.26 (m, 5H), 4.52 (s, 2H), 3.54 (s, 2H), 2.00-1.89 (m, 2H), 1.66-1.46 (m, 6H).
Step 4: (2E)- and (2Z)-3-1-[(Benzyloxy)methyl]cyclopentylacrylonitrHe [0666] To a stirred solution of diethyl cyanomethylphosphonate (18 μί, 0.11 mmol) in THF (1 mL) was added 1.0 M potassium ferf-butoxide in THF (0.10 mL). The resulting mixture was stirred 30 minutes, after which a solution of 1-[(ben-zyloxy)methyl]cyclopentanecarbaldehyde (0.020 g, 0.092 mmol) in THF (1 mL) was added. The resulting mixture was stirred for 16 hours. Water was then added to the reaction and the resulting mixture was extracted with three portions of ethyl ether. The combined extracts were washed with brine, then dried over sodium sulfate, decanted from the sodium sulfate, and the solvent was removed in vacuo to afford the product, which was used without further purification in the subsequent conjugate addition step. 1H NMR (400 MHz, CDCI3): δ 7.37-7.27 (m, 5H), 6.80 (d, 1H (E)), 6.59 (d, 1H (Z)), 5.34 (d, 1H (E)), 5.33 (d, 1H (Z)), 4.53 (s, 2H (Z)), 4.50 (s, 2H (E)), 3.45 (s, 2H (Z)), 3.31 (s, 2H (E)), 1.80-1.55 (m, 8H); MS(ES)=242(M+H).
Step 5: 3-1-[(Benzyloxy)methyl]cyclopentyl-3-[4-(7Hpyrrolo[2,3-d]pyrimidin-4-yi)-1 H-pyrazoi-1 -yijpropanenitriie trifluor-oacetate salt [0667] To a mixture of (2E)- and (2Z)-3-1-[(benzyloxy)methyl]cyclopentylacrylonitrile (generated in Step 4) and 4-(1H-pyrazol-4-yl)-7-[2-(trimethylsilyl)ethoxy]methy]-7H-pyrrolo[2,3-d]pyrimidine (0.037 g, 0.12 mmol) in ACN (1.5 mL) was added DBU (18 μι, 0.12 mmol). The resulting mixture was stirred at ambient temperature for 3 hours, and then was heated to 60 °C for 28 hours. The reaction mixture was diluted with diethyl ether and 0.1 N HCI. The layers were separated and the aqueous layer was extracted with ethyl acetate. The ethyl acetate extract was washed with brine, dried over sodium sulfate, decanted, and the solvent was removed in vacuo. The resulting residue was dissolved in DCM (3 mL) and TFA (0.75 mL), and this solution was stirred for 3 hours. The solvents were removed in vacuo, and the resulting residue was dissolved in THF (5 mL) and 6.0 M sodium hydroxide in water (3 mL) and stirred for 2 hours. The reaction mixture was extracted with three portions of ethyl acetate. The combined extracts were washed with brine, dried over sodium sulfate, decanted, and the solvent was removed in vacuo. The crude mixture was purified by preparative-HPLC/MS (C 18 eluting with a gradient of H20 and ACN containing 0.1% TFA) and lyophilized to afford the desired product (10 mg, 20% over the two steps). 1H NMR (400 MHz, d6-DMSO): Ô12.71 (br s, 1H), 8.99 (s, 1H), 8.86 (s, 1H), 8.52 (s, 1H), 7.80 (s, 1H), 7.38-7.23 (m, 5H), 7.19-7.16 (m, 1H), 4.92 (dd, 1H), 4.50 (d, 1H), 4.44 (d, 1H), 3.49 (dd, 1H), 3.35 (d, 1H), 3.23 (dd, 1H), 3.05 (d, 1H), 1.92-1.82 (m, 1H), 1.66-1.27 (m,7H); MS(ES):427(M+H).
Example 742: 3-[1-(Methylsulfonyl)pyrrolidin-3-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]pro-panenitrile trifluoroacetate salt [0668]
Step 1: Benzyl 3-(hydroxymethyl)pyrrolidine-1 -carboxylate [0669] To a solution of 1-[(benzyloxy)carbonyl]pyrrolidine-3-carboxylic acid (1.0 g, 4.0 mmol) in THF (37 mL) at 0 °C was added dropwise a solution of 1.0 M borane in THF (16.4 mL). The reaction was allowed to warm to room temperature and stir for 16 hours. The mixture was cooled to 0 °C and 10% HCI (50 mL) was added. After the addition, the mixture was extracted with DCM, and the extract was washed sequentially with saturated NaHC03 solution and brine, then dried over sodium sulfate, filtered and the solvent was removed in vacuo. The product was used without further purification in the subsequent oxidation step. 1H NMR (300 MHz, CDCI3): δ 7.39-7.26 (m, 5H), 5.11 (s, 2H), 3.61-3.31 (m, 5H), 3.18 (dt, 1H), 2.75 (brs, 0.45H), 2.59 (brs, 0.45H), 2.49-2.31 (m, 1H), 2.19 (brs, 0.1H), 2.05-1.89 (m, 1H), 1.77-1.58 (m, 1H); MS(ES):236(M+H).
Step 2: Benzyl 3-formylpyrrolidine-1-carboxylate [0670] DMSO (597 μί, 8.42 mmol) was added to a solution of oxalyl chloride (427 μί, 5.05 mmol) in DCM (25 mL) at -78 °C. After 5 minutes, benzyl 3-(hydroxymethyl)pyrrolidine-1-carboxylate (generated in Step 1) was added. The reaction was continued for 30 minutes at -78 °C. TEA (2.3 mL, 17 mmol) was then added. The resulting mixture was then allowed to warm to room temperature over the course of 30 minutes. Water was then added. The layers were separated and the organic phase was washed sequentially with 0.1 N HCI, water, saturated NaHC03, and brine. The organic phase was then dried over sodium sulfate and the solvent was removed in vacuo to afford the product (0.82 g, 88% over two steps). 1H NMR (300 MHz, CDCI3): δ 9.68 (d, 1H), 7.38-7.28 (m, 5H), 5.13 (s, 2H), 3.79 (dd, 1H), 3.65-3.35 (m, 3H), 3.11-2.99 (m, 1H), 2.32-2.04 (m,2H).
Step 3: Benzyl 3-[(E)-2-cyanovinyl]pyrrolidine-1-carboxylate and benzyl 3-[(Z)-2-cyanovinyl]-pyrrolidine-1-carboxylate [0671] To a solution of 1.0 M potassium ferf-butoxide in THF (4.40 mL) at 0 °C was added a solution of diethyl cyanomethylphosphonate (820 mg, 4.6 mmol) in THF (6.0 mL) dropwise. The cold bath was removed and the reaction was warmed to room temperature and stirred for 15 minutes. The mixture was cooled to 0 °C and a solution of benzyl 3-formylpyrrolidine-1-carboxylate (0.82 g, 2.3 mmol) in THF (4.00 mL) was added dropwise. Cooling was discontinued and the reaction stirred for 16 hours at ambient temperature. The mixture was diluted with ether and water, the layers were separated and the organic layer was washed with water, followed by brine, and then dried over sodium sulfate, filtered and the solvent was removed in vacuo. The resulting residue was purified by flash column chromatography (eluting with a gradient from 0-35% ethyl acetate in hexanes) to afford the product as a mixture of E and Z isomers (246 mg, 42%). 1H NMR (300 MHz, CDCI3): δ 7.41-7.27 (m, 5H), 6.70-6.58 (m, 0.3H (£)), 6.38 (dt, 0.7H (Z)), 5.50-5.30 (m, 1H), 5.14 (s, 2H), 3.79-3.11 (m, 5H), 2.27-2.06 (m, 1H), 1.90-1.70 (m, 1H); MS(ES):279(M+Na).
Step 4: Benzyl 3-2-cyano-1-[4-(7-[2-(trimethylsHyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl-pyrrolidine-1 -carboxyla te [0672] To a mixture of benzyl 3-[(E)-2-cyanovinyl]pyrrolidine-1-carboxylate and benzyl 3-[(Z)-2-cyanovinyl]pyrrolidine-1-carboxylate (241 mg, 0.940 mmol) and DBU (234 μι, 1.57 mmol) in ACN (13 mL) was added 4-(1 H-pyrazol-4-yl)-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidine (250 mg, 0.78 mmol). The mixture was stirred at ambient temperature for 3 hours. The solvent was removed In vacuo. The resulting residue was dissolved in ethyl acetate, and the organic layer was washed sequentially with 1N HCI, water, saturated NaHCOs, and brine. The washed solution was dried over sodium sulfate and the solvent was removed in vacuo. Purification via flash column chromatography (eluting with a gradient of 0-100% [5% MeOH/DCM] in hexanes) afforded the produce as a mixture of diastereomers (400 mg, 89%). 1H NMR (400 MHz, CDCI3 a mixture of diastereomers):ô 8.85 (s, 1H), 8.35-8.28 (m, 2H), 7.42-7.25 (m, 6H), 6.80-6.76 (m, 1H), 5.69-5.66 (m, 2H), 5.15-5.04 (m, 2H), 4.46-4.32 (m, 1H), 3.84-3.84 (m, 6H), 3.54 (t, 2H), 2.26-2.13 (m, 1H), 1.84-1.54 (m, 2H), 0.95-0.89 (m, 2H), -0.06 (s, 9H); MS(ES):572(M+H).
Step 5. 3-Pyrrolidin-3-yl-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]pro-panenitrile [0673] Benzyl 3-2-cyano-1-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-dlpyrimidin-4-yl)-1H-pyrazol-1-yl)ethylpyrrolidine-1-carboxylate (161 mg, 0.282 mmol) was dissolved in methanol (5 mL), and a catalytic amount of 5% Pd-C was added. The suspension was stirred at ambient temperature for 1 hour under an atmosphere of hydrogen provided by a balloon. A catalytic amount of 10% Pd-C was then added, and the reaction stirred for 2 hours under an atmosphere of hydrogen provided by a balloon. The mixture was then filtered, and purified via preparative-HPLC/MS (C18 eluting with a gradient of H20 and ACN containing 0.15% NH4OH) to afford the product as a mixture of diastereomers (57 mg, 46%). 1H NMR (400 MHz, CDCI3, a mixture of diastereomers): δ 8.84 (s, 1H), 8.34-8.32 (m, 2H), 7.40 (d, 1H), 6.81-6.78 (m, 1H), 5.67 (s, 2H), 4.38 (dt, 1H), 3.54 (t, 2H), 3.30-1.38 (m, 9H), 0.92 (t, 2H), -0.06 (s, 9H); MS(ES):438(M+H).
Step 6:3-[1-(Methylsulfonyl)pyrrolidin-3-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile trifluor-oacetate salt [0674] To asolution of 3-pyrrolidin-3-yl-3-[4-(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidinyl)1 H-pyra-zol-1-yl]propanenitrile (25 mg, 0.057 mmol) and TEA (10 μί, 0.074 mmol) in DCM (1.0 mL) at 0 °C was added meth-anesulfonyl chloride (6 μί, 0.074 mmol). The reaction was allowed to reach ambient temperature and stir for 16 hours. Half of the solvent was removed in vacuo and TFA (1 mL) was added to the vial. After stirring for 1 hour at room temperature, the solvents were removed in vacuo and the resulting residue reconstituted in THF (0.5 mL). To this was added 6 N NaOH (1 mL) and this solution was stirred for 2 hours. The reaction mixture was extracted with five portions of ethyl acetate. The combined extracts were dried (Na2S04), decanted and concentrated. Preparative-HPLC/MS (C18 eluting with a gradient of H20 and ACN containing 0.1 % TFA) was used to afford the product (16 mg, 57%). 1H NMR (400 MHz, d6-DMSO, a mixture of diastereomers): δ 12.69 (s, 1H), 8.98 (s, 0.5H), 8.95 (s, 0.5H), 8.84 (s, 1H), 8.53-8.51 (m, 1H), 7.80-7.77 (m, 1H), 7.16-7.13 (m, 1H), 4.86-4.75 (m, 1H), 3.55-3.48 (m, 1H), 3.42-3.08 (m, 4H), 2.99-2.91 (m, 1H), 2.90 (s, 1.5H), 2.85 (s, 1.5H), 2.16-2.07 (m, 1H), 1.82-1.70 (m, 1H), 1.64-1.48 (m, 1H); MS(ES):386(M+H).
Example 743: N’-Cyano-4-(cyanomethyl)-4-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]piperidine-1-carboximidamide [0675]
Step 1: tert-Butyl 4-(cyanomethylene)piperidine-1-carboxylate [0676] To a solution of 1.0 M potassium ferf-butoxide in THF (10.1 mL) at 0 °C was added a solution of diethyl cyanomethylphosphonate (1.66 mL, 0.0102 mol) in THF (20 mL) dropwise. The reaction was held for 10 min, then added to a solution of tert-butyl 4-oxo-1-piperidinecarboxylate (2.00 g, 0.0100 mol) in THF (30 mL) stirring at 0 °C under an atmosphere of nitrogen. After complete addition, the cold bath was removed and the reaction was allowed to stir 1.0 h at 20 °C. LCMS analysis showed the desired product and no remaining starting material. HPLC showed the product UVmax at 200 & 230 nm. Water and EtOAc were added to the reaction mixture. The phases were separated, and the aqueous phase was extracted with EtOAc. The combined organic phase was washed with water, then saturated NaCI, then dried over Na2S04, and concentrated to dryness to provide 2.5 g of the product as a yellow oil. TLC (25% EtOAc/hex-ane) Rf 0.22. The product was purified by automatic flash chromatography on silica gel. Used a 40g column; flow 40 mL/min; [A= hexane] [B= EtOAc], A, 4 min; Gradient to 20% B in 30 min. Collected 44 mL fractions. The product eluted in 21-27 min. The fractions were contrated to yield 0.67 g of a white solid. 1H NMR (CDCI3) δ 5.19 (s, 1H); 3.51 (m, 4H); 2.56 (t, 2H); 2.33 (t, 2H); 1.50 (s, 9H). MS(ES) 245 (M+Na, weak; base peak M+H-56 = 167).
Step 2: tert-Butyl 4-(cyanomethyl)-4-[4-(7-[2-(trimethylsHyl)ethoxy]methyl-7H-pyrrolo[2,3-d]-pyrimidin-4-yl)-1H-pyrazol- 1 -yljpiperidine-1 -carboxylate [0677] 4-(1H-Pyrazol-4-yl)-7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidine (0.840 g, 2.66 mmol) was slurried in a mixture of ACN (20 mL) and DBU (398 μί, 2.66 mmol), and ferf-butyl 4-(cyanomethylene)piperidine-1-carboxylate (0.651 g, 2.93 mmol) was added. The pyrazole did not dissolve at 20° C, but a solution was formed when the mixture was heated to 40 °C for 1h. LCMS and HPLC analyses showed about 20% conversion to product. The mixture was stirred at 40-45 °C overnight. HPLC showed 60area% product. The ACN was removed by retory evaporator at 20 °C. To the resulting residue was added saturated NaHCOs and EtOAc. The organic layer was shaken with more aqueous saturated NaHC03, then dried (Na2S04) and rotovaped to give 1.6g of a brown oil residue. TLC (60% EtOAc/hex-ane): product Rf = 0.25. The product was purified by automatic flash chromatography on silica gel, using a 40g column, at a flow of 40 mL/min; [A= hexane] [B= EtOAc]. A, 3min; Gradient to 100% B in 50 min. Collected 44 mL fractions. The product eluted in 24-29 min; the pyrazole in 39-46 min; and the olefin in 13-15 min. Solvent was removed in vacuo for the appropriate fractions to give 0.27 g olefin; 0.30 g pyrazole; and a yield of 0.67 g of the product, all of which were isolated as white solids. 1H NMR (CDCI3) δ 8.84 (s, 1H); 8.42 (s, 1H); 8.33 (s, 1H); 7.40 (d, 1H); 6.79 (d, 1H); 5.67 (s, 2H); 3.94 (m, 2H); 3.54 (m, 2H); 3.07 (m, 2H); 2.90 (s, 2H); 2.72 (m, 2H); 2.08 (m, 2H); 1.45 (s, 9H); 0.91 (m, 2H); -0.06 (s, 9H). MS(ES) 538 (M+H).
Step 3: 4-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]piperidin-4-ylacetonitrile [0678] tert-Butyl 4-(cyanomethyl)-4-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]-pyrimidin-4-yl)-1H-pyra-zol-1-yl]piperidine-1-carboxylate (0.670 g, 1.24 mmol) was dissolved in TFA (5.0 mL, 65 mmol) and was stirred for 1.3 h. LCMS showed conversion to the hydroxymethyl intermediate, M+H 338. The solution was concentrated to remove the TFA. Methanol was added to the resulting residue, and the resulting mixture was concentrated. The resulting residue was dissolved in methanol (10 mL) and 15.0 M ammonium hydroxide in water (1.66 mL) was added. The resulting solution was stirred for 2 h. LCMS and HPLC analyses showed complete deprotection. The mixture was concentrated. Toluene was added to the resulting residue and the resulting mixture was concentrated to provide a white semisolid. Most of this intermediate product was used for the next step. The rest was purified by prep HPLC using a 30 mm x 100 mm C18 column; 8% ACN-H20 (0.1% NH4OH), 1.0min, to 27% at 6min; 60 mL/min; detector set at m/z 308; retention time, 5.4 min. Tubes containing pure product were combined and freeze dried to give 13.6 mg of the product. 1H NMR (d6-DMSO) δ 12.07 (s, 1H);8.68 (s, 1H);8.62(s, 1H);8.36(s, 1H);7.54 (d, 1H);7.00 (d, 1H);3.16(s, 2H);2.87 (m, 2H); 2.55 (m, 4H); 1.94 (m, 2H). MS(ES) 308 (M+H).
Step 4: Methyl N-cyano-4-(cyanomethyl)-4-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-piperidine-1-carbimi-dothioate [0679] 4-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]piperidin-4-ylacetonitrile (361 mg, 1.17 mmol) and A/-cy-ano-S,S’-dimethyldithioimido carbonate (344 mg, 2.35 mmol) were dissolved in isopropyl alcohol (2.5 mL) and DMSO (2.5 mL) at 20 °C. After 16 h reaction time, LCMS analysis showed the presence of some product, M+H 406; of the reagent, M+H 147; and of the piperidine, M+H 308. HPLC analysis showed about 2% reaction. The HPLC method was: Zorbax SB C18, 5μ(η, 15 cm, 35 °C, flow 1.2 mL/min, 5% ACN-H20 (0.05% TFA), 1.5 min, to 100% ACN in 15.0 min; detector set at 324, 225, and 265 nm. The retention time of the starting material was 4.9 min (UV max 224, 262, 292, & 325 nm); of the product, 6.5 min (UV max 226, 262, 290, & 324nm); and of the reagent, 7.7min (UV max 265nm). To the product was added TEA (327 μι, 2.35 mmol), and the resulting mixture was stirred at RT. After stirring for 3 h, HPLC and LCMS analyses showed 60% reaction. The product and the unreacted piperidine were isolated by prep HPLC using a 30 mm X 100 mm C18 column; 5%ACN-H20 (0.1%TFA), 1.0min, to 35% at 6min; 60 mL/min; detector set at 326 nm. The retention time for the product was 5.9 min; and for the starting piperidine was 3.5-4.3 min. The product was freeze dried to yield 301 mg of a white solid TFA salt. 1H NMR (dg-DMSO) δ 12.85 (s, 1H);9.01 (s, 1H); 8.90 (s, 1H); 8.59 (s, 1H); 7.85 (m, 1H); 7.30 (m, 1H); 4.23 (m, 2H); 3.35 (m, 2H); 3.30 (s, 2H); 2.78 (m, 2H); 2.68 (s, 3H); 2.16 (m, 2H). MS(ES) 406 (M+H).
Step 5: N’-Cyano-4-(cyanomethyl)-4-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]piperidine-1-carboximidamide [0680] Methyl /V-cyano-4-(cyanomethyl)-4-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-piperidine-1-carbimi-dothioate (41.3 mg, 0.102 mmol) (53 mg TFA salt) was dissolved in 2.0 M ammonia in isopropyl alcohol (4.00 mL). The resulting mixture was heated to 100 °C for 1 h in a microwave reactor. Analysis by HPLC and LCMS showed 60% reaction to give the expected M+H 375 (50 area%). To this mixture was added 2 mL of 7 N NH3/MeOH. The resulting mixture was heated at 120 °C for one hour. HPLC and LCMS analyses showed no remaining starting material. The reaction mixture was concentrated on a rotory evaporator. The product was isolated by prep HPLCMS using a 30 mm X 100 mm C18 column, eluting with a solvent gradient; 10% ACN-H20 (0.1%TFA), 1.5min, to 30% at 6min; 60mL/min; detector set at m/z 375; retention time, 4.7 min. The eluate was freeze-dried to yield 11.7 mg of the product TFA salt as a white solid. 1H NMR (d6-DMSO) δ 12.69 (s, 1H, NH); 8.92 (s, 1H); 8.81 (s, 1H); 8.51 (s, 1H); 7.75 (m, 1H); 7.22 (m, 1H); 7.18 (s, 2H, NH2); 3.84 (m, 2H); 3.23 (s, 2H); 2.99 (m, 2H); 2.60 (m, 2H); 1.97 (m, 2H). MS(ES) 375 (M+H).
Example 744: 4-1-[2,2,2-Trifluoro-1-(1H-imidaxol-2-ylmethyl)ethyl]-1H-pyrazol-4-yl-7H-pyrrolo[2,3-d]pyrimidine [0681]
Stepl: (3R)-4,4,4-Trifluoro-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1Hpyrazol-1yl]bu-tanai [0682]
[0683] To a -70 °C solution of (3R)-4,4,4-trifluoro-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]butanenitrile (1.06 g, 0.00243 mol) (see, Example 93, Stepl) in DCM (10 mL, 0.2 mol) was added 1.0 M diisobutylaluminum hydride in DCM (4.8 mL). The resulting mixture was stirred for 3h and allowed to warm during this time interval from -70 to -25 °C, after which the reaction was cooled back at -70 °C. Methanol (1.5 mL, 0.037 mol) was added, followed by 2.0 M HCI in water (15 mL). Insoluble material was then filtered from the reaction mixture. The organic filtrate was washed sequentially with: 2.0 M HCI in water, water and saturated aqueous NaCI. The washed organic phase was dried over sodium sulfate and was concentrated using a rotory evaporator to give 0.58 g of the crude product as a pale yellow foam/solid. The crude product was chromatographed with 0-80% ethyl acetate/hexanes to give the purified product (0.9 g) as a pale orange oil (47% yield). 1H NMR (400 MHz, CDCI3): δ 9.85 (1H, s); 8.95 (1H, s); 8.5 (1H, s); 8.4 (1H, s); 7.5 (1H, d); 6.85 (1H, d); 5.75 (2H, s); 5.5 (1H, m); 4.0 (1H, dd); 3.6 (2H, t); 3.3 (1H, dd); 1.99 (2H, t); 0.0 (9H, s). MS (M+H): 440.
Step2: 4-1-[2,2,2-Trifluoro-1-(1H-imidazol-2-ylmethyl)ethyl]-1H-pyrazol-4-yl-7-[2-(trimethylsilyl)-ethoxy]methyl-7H-pyr-rolo[2,3-d]pyrimidine [0684]
[0685] A solution of 4,4,4-trifluoro-3-[4-(7-[2-(trimethylsilyl)ethoxy]methyl-7H-pyrrolo[2,3-d]-pyrimidin-4-yl)-1H-pyra-zol-1 -yl]butanal (0.138 g, 0.000314 mol), 7.0 M ammonia in methanol (1 mL), ethanedial (0.5 mL, 0.004 mol) and acetic acid (20 uL, 0.0004 mol) in methanol (2 mL, 0.05 mol) was microwaved on 100 watts, at 80 °Cfor 60 minutes. Following the microwave reaction, ethyl acetate/water was added. The organic phase was separated and washed with saturated NaHC03 and saturated NaCI. The washed organic phase was dried and concentrated (rotory evaporator) to give 196 mg of the crude product as an orange glass. The crude product was purified by chromatography with 0-100% ethyl acetate/hexanes to give 57 mg of purified product as an off-white solid (38% yield). 1H NMR (400 MHz, CDCI3): δ 8.91 (1H, s); 8.4 (1H, s); 8.2 (1H, s); 7.5 (1H, d); 7.0 (2H, s); 6.83 (1H, d); 5.75 (2H, s); 5.62 (1H, m); 4.15 (1H, dd); 3.8 (1H, dd); 3.6 (2H, t); 1.99 (2H, t); 0.0 (9H, s). MS (M+H): 478.
Step3: 4-1 -[2,2,2-Trifluoro-1-(1 H-imidazol-2-ylmethyl)ethyl]-1H-pyrazol-4-yl-7H-pyrrolo[2,3-d]-pyrimidine [0686] A solution of 4-1-[2,2,2-trifluoro-1-(1H-imidazol-2-ylmethyl)ethyl]-1H-pyrazol-4-yl-7-[2-(trimethylsi-lyl)ethoxy]methyl-7H-pyrrolo[2,3-d]pyrimidine (0.055 g, 0.12 mmol) in 1,2-dichloroethane (1 mL, 10 mmol) and TFA(0.5 mL, 6 mmol) was stirred overnight. The reaction was concentrated to provide an orange oil. The oil was stirred in methanol (1 mL, 20 mmol) and 8.0 M ammonium hydroxide in water (1 mL) for 4h. This mixture was then concentrated to provide a crude product as an orange glass/solid. The crude product was purified by Prep HPLC (pH10) to give 28 mg of purified product as a colorless glass, which was triturated with 2-methoxy-2-methylpropane (1 mL, 8 mmol), and then filtered and washed to provide 15 mg of the product as a white solid (38% yield) which then was dried rt-50 °C for 3h. 1H NMR (400 MHz, DMSO): δ 12.13 (1H, s); 11.89 (1H, s); 8.65 (1H, s); 8.37 (1H, s); 7.6 (1H, d); 6.95 (1H, d); 6.92 (1H, d); 5.91 (1H, m); 3.78 (1H, dd); 3.47 (H, dd). MS (M+H): 348.
Example 745: 4-(1-(1 R)-2,2,2-Trifluoro-1-[(4-methyl-1,3-thiazol-2-yl)methyl]ethyl-1 H-pyrazol-4-yl)-7H-pyrro-lo[2,3-d]pyrimidine [0687]
Step 1: (3R)-4,4,4-Trifluoro-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]butane-thioamide [0688]
[0689] A suspension of phosphorus pentasulfide (0.46 g, 1.0 mmol) in ethanol (0.5 mL, 8 mmol) was stirred for 1h. (3R)-4,4,4-Trifluoro-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]butane-nitrile (0.15 g, 0.50 mmol) (see, Example 93) was added and the resulting mixture was heated at 80 °C in a sealed vial for 0.5h, during which reaction the mixture became a yellow solution. The reaction was heated overnight. The reaction was then cooled to rt. Water (1 g, 60 mmol) and ethyl acetate were added to the mixture. The organic phase was separated and washed with saturated NaHC03 and saturated aqueous NaCI. The washed organic phase was then dried and concentrated to give 387 mg of a crude product as a white glass/oil. The crude product was chromatographed with 0-10% MeOH/DCM, 0-1%NH4OH to give 0.13 g of the purified product as a white solid (76% yield). 1H NMR (400 MHz, CDCI3): δ 8.7 (1H, s); 8.5 (1H, s); 8.3 (1H, s); 7.4 (1H, d); 7.0 6.75 (1H, d); 5.82 (1H, m); 3.75 (1H, dd); 3.2 (1H, dd). MS (M+H): 341.
[0690] A suspension of (3R)-4,4,4-trifluoro-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]butanethioamide (0.038 g, 0.00011 mol), chloroacetone (15 uL, 0.00019 mol) in ethanol (1 mL, 0.02 mol) and 1,2-dichloroethane (1 mL, 0.01 mol) was heated to reflux overnight. Following this, the reaction mixture was filtered to remove insoluble material. The filtrate was dissolved in MeOH (1 mL) and DMF (1 mL) and purified by prep HPLC at pH10 to provide 6 mg of the purified product as a colorless glass/oil, which was then triturated with MTBE/hexanes and was dried at 40 °C overnight to give 5.2 mg of the purified product as an off-white solid (13% yield). 1H NMR (400 MHz, CDCI3): δ 10.11 (1H,s); 8.88 (1H,s); 8.42 (1H, s); 8.38 (1H, s); 7.45 (1H,d); 6.79 (1H,s); 6.65 (1H, d); 5.41 (1H, m); 4.15 (1H, dd); 3.75 (H, dd); 2.18 (3H, s). MS (M+H): 379.
Example A: In vitro JAK Kinase Assay [0691] Compounds herein were tested for inhibitory activity of JAK targets according to the following in vitro assay described in Park et al., Analytical Biochemistry 1999,269,94-104. The catalytic domains of human JAK1 (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 JAK1, 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 reactions that contain the enzyme, ATP and 500 nM peptide in 50 mM Tris (pH 7.8) buffer with 100 mM NaCI, 5 mM DTT, and 0.1 mg/mL (0.01%) BSA. The ATP concentration in the reactions was 90 μΜ for Jaki, 30 μΜ for Jak2 and 3 μΜ for Jak3. Reactions were carried out at room temperature for 1 hr and then stopped with 20 μί 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). Compounds having an IC50 of 10 μΜ or less for any of the above-mentioned JAK targets were considered active.
Example B: Cellular Assays [0692] One or more compounds herein were tested for inhibitory activity of JAK targets according to at least one of the following cellular assays.
[0693] Cancer cell lines dependent on cytokines and hence JAK/STAT signal transduction, for growth, were plated at 6000 cells per well (96 well plate format) in RPMI 1640, 10% FBS, and 1 nG/mL of appropriate cytokine. Compounds were 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 was assessed using the CellTiter-Glo Luminescent Cell Viability Assay (Promega) followed by TopCount (Perkin Elmer, Boston, MA) quantitation. Potential off-target effects of compounds were measured in parallel using a non-JAK driven cell line with the same assay readout. Compounds having an IC50 of 10 μΜ or less with selectivity for JAK driven proliferation were considered active. All experiments were performed in duplicate.
[0694] 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.
[0695] 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.
[0696] Certain compounds herein have been or 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 élutriation. Freshly isolated human T-cells can be maintained in culture medium (RPM11640 supplemented with10% 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 cel ls/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, Wl).
Example C: In vivo anti-tumor efficacy [0697] 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 subcutaneously (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 overtime 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 [0698] 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(1 ):37-44). Murine DTH shares multiple characteristics with psoriasis, including the immune infiltrate, the accompanying increase in inflammatory cytokines, and kératinocyte 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(1-2):116-21).
[0699] 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 μι. (10 μι. on the internal pinna and 10 μι. 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 was 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). T reatment of the test compounds (in different concentration) was 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).
[0700] 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 kératinocytes 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 [0701] Compounds herein can be or have been 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 l-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.).
[0702] 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.
Claims 1. A compound of formula:
or a pharmaceutically acceptable salt thereof, wherein: T, U, and V are independently selected from O, S, N, CR5, and NR6; wherein the 5-membered ring formed by carbon atom, nitrogen atom, U, T, and V is aromatic; X is N or CR4; n is 0; or n is 1 and Y is C.,_8 alkylene, C2_8 alkenylene, (CR11R12)pC(0)(CR11R12)q, (CR11R12)pC(0)NRc(CR11R12)q, (CR11R12)pC(0)0(CR11R12)q, or (CR11R12)p0C(0)(CR11R12)q, wherein said C.,_8 alkylene orC2_8 alkenylene, is optionally substituted with 1,2, or 3 halo, OH, CN, amino, C1-4 alkylamino, or C2_8 dialkylamino; Z is aryl, cycloalkyl, heteroaryl, or heterocycloalkyl, each optionally substituted with 1,2, 3,4, 5, or 6 substituents independently selected from halo, C^ alkyl, C2.4 alkenyl, C2.4 alkynyl, C1-4 haloalkyl, hydroxyalkyle, 01-4 cyanoalkyl, Cy1, CN, N02, ORa, SRa, C(0)Rb, C(0)NRcRd, C(0)0Ra, 0C(0)Rb, 0C(0)NRcRd, NRcRd, NRcC(0)Rb, NRcC(0)NRcRd, NRc(0)0Ra, S(0)Rb, S(0)NRcRd, S(0)2Rb, NRcS(0)2Rb, and S(0)2NRcRd;
Cy1 is independently selected from aryl, heteroaryl, cycloalkyl, and heterocycloalkyl, each optionally substituted by 1,2,3,4 or 5 substituents independently selected from halo, C1_4 alkyl, C2-4 alkenyl, C2_4 alkynyl. C^ haloalkyl, CN, N02, OR3', SRa", C(0)Rb", C(0)NRc"Rd", C(0)0Ra", 0C(0)Rb”, 0C(0)NRc”Rd”, NRc"Rd", NRc”C(0)Rd", NRc"C(0)0Ra", S(0)Rb", S(0)NRc"Rd", S(0)2Rb", and S(0)2NRc"Rd"; R4 is H; R5 is H, halo, C^ alkyl, C2_4 alkenyl, C2_4 alkynyl, C^ haloalkyl, CN, N02, OR7, SR7, C(0)R8, C(0)NR9R10, C(0)0R7, O0(O)R8, OC(0)NR9R10, NR9R10, NR9C(0)R8, NR9C(0)0R7, S(0)R8, S(0)NR9R10, S(0)2R8, NR9S(0)2R8, or S(0)2NR9R10; R6 is H, alkyl, C2.4 alkenyl, C2_4 alkynyl, haloalkyl, OR7, C(0)R8, C(0)NR9R10, 0(O)OR7, S(0)R8, S(0)NR9R10, S(0)2R8, or S(0)2NR9R10; R7 is H, alkyl, haloalkyl, C2.6 alkenyl, C2.6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl; R8 is H, C^ alkyl, C^g haloalkyl, C2.6 alkenyl, C2.6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl; R9 and R10 are independently selected from Η, Ο1-10 alkyl, 01-6 haloalkyl, C2_6 alkenyl, C2_6 alkynyl, 01-6 alkylcarbonyl, arylcarbonyl, C.|_g alkylsulfonyl, mylsulfonyl aryl, heteroaryl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl and heterocycloalkylalkyl; or R9 and R10 together with the N atom to which they are attached form a 4-, 5-, 6-or7-membered heterocycloalkyl group; R11 and R12 are independently selected from H, halo, OH, CN, C1jt alkyl, 01-4 haloalkyl, C2_^ alkenyl, C24 alkynyl, C1-4 hydroxyalkyl, cyanoalkyl, aryl, heteroaryl, cycloalkyl, and heterocycloalkyl;
Raand Ra" are independently selected from Η, 01-6 alkyl, C^g haloalkyl, C2_6 alkenyl, C2_g alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl and heterocycloalkylalkyl, wherein said C^g alkyl, haloalkyl, C2_6 alkenyl, C2_g alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted with 1,2, or 3 substituents independently selected from OH, CN, amino, halo, C., _6 alkyl, C^g haloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, cycloalkyl and heterocycloalkyl;
Rband Rb" are independently selected from Η, 01-6 alkyl, C^g haloalkyl, C2_6 alkenyl, C2_6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl and heterocycloalkylalkyl, wherein said alkyl, haloalkyl, C2_g alkenyl, C2_g alkenyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted with 1, 2, or 3 substituents independently selected from OH, CN, amino, halo, C^galkyl, 01-6haloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, cycloalkyl and heterocycloalkyl;
Rc and Rd are independently selected from H, C.|_10 alkyl, 01-6 haloalkyl, C2.6 alkenyl, C2.6 alkynyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl and heterocycloalkylalkyl, wherein said Ci_io alkyl, C^g haloalkyl, C2_6 alkenyl, C2.6 alkenyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted with 1,2, or 3 substituents independently selected from OH, CN, amino, halo, C^galkyl, C^ghaloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, cycloalkyl or heterocycloalkyl; orR°and Rd together with the N atom to which they are attached form a 4-, 5-, 6-or7-membered heterocycloalkyl group optionally substituted with 1, 2, or 3 substituents independently selected from OH, CN, amino, halo, C^galkyl, C^ghaloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, cycloalkyl and heterocycloalkyl;
Rc" and Rd" are independently selected from H, C-i^q alkyl, 01-6 haloalkyl, C2_g alkenyl, C2_6 alkynyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl and heterocycloalkylalkyl, wherein said Ο1-10 alkyl, C^ haloalkyl, C2.6 alkenyl, C2_g alkenyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted with 1,2, or 3 substituents independently selected from OH, CN, amino, halo, 01-6 alkyl, C^ghaloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, cycloalkyl and heterocycloalkyl; or Rc" and Rd" together with the N atom to which they are attached form a 4-, 5-, 6- or 7-mem bered heterocycloalkyl group optionally substituted with 1, 2, or 3 substituents independently selected from OH, CN, amino, halo, C^galkyl, C^ghaloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, cycloalkyl and heterocycloalkyl; p is 0, 1,2, 3, 4, 5, or 6; and q is 0, 1,2, 3, 4, 5 or 6; for use in a method of treating a disease selected from cachexia, a myeloproliferative disorder, and cancer in a patient in need thereof, in combination with an additional pharmaceutical agent. 2. The compound or salt for use according to claim 1, wherein the compound is a compound of Formula II:
or a pharmaceutically acceptable salt thereof. 3. The compound or salt for use according to claims 1 or 2, wherein X is N. 4. The compound or salt for use according to any one of claims 1 to 3, wherein n is 0. 5. The compound or salt for use according to any one of claims 1 to 3, wherein n is 1 and Y is 01-8 alkylene optionally substituted with 1,2, or 3 halo, OH, CN, amino, C1jt alkylamino, or C2_g dialkylamino. 6. The compound or salt for use according to any one of claims 1 to 5, wherein Z is cycloalkyl, optionally substituted with 1, 2, 3, 4, 5, or 6 substituents independently selected from halo, alkyl, C2.4 alkenyl, C2_4 alkynyl, 01-4 haloalkyl, hydroxyalkyl, C1A cyanoalkyl, Cy1, CN, N02, ORa, SRa, C(0)Rb, C(0)NRcRd, C(0)0Ra, 0C(0)Rb, 0C(0)NRcRd, NRcRd, NRcC(0)Rb, NRcC(0)NRcRd, NRcC(0)0Ra, S(0)Rb, S(0)NRcRd, S(0)2Rb, NRcS(0)2Rb, and S(0)2NRcRd. 7. The compound or salt for use according to any one of claims 1 to 5, wherein Z is heterocycloalkyl, optionally substituted with 1,2,3,4,5, or 6 substituents independently selected from halo, C.|_4 alkyl, C2.4 alkenyl, C2.4 alkynyl, 01-4 haloalkyl, hydroxyalkyl, cyanoalkyl, Cy1, CN, N02, ORa, SRa, C(0)Rb, C(0)NRcRd, C(0)0Ra, 0C(0)Rb, 0C(0)NRcRd, NRcRd, NRcC(0)Rb, NRcC(0)NRcRd, NRcC(0)0Ra, S(0)Rb, S(0)NRcRd, S(0)2Rb, NRcS(0)2Rb, and SCO)2NRcRd. 8. The compound or salt for use according to claim 1, wherein said compound is selected from: 4-[1-(1-methyl-3-pyrazol-1-yl-propyl)-1 H-pyrazol-4-yl]-1 H-pyrrolo[2,3 b]pyridine; 4-[1-(3-imidazol-1-yl-1-methyl-propyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine; 4-cyclopentyl-4-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-pyrazol-1-yl]-butyronitrile; 3-cyclopropyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-pyrazol-1-yl]-propionitrile; 3-cyclopentyl-3-[4-(1H-pyrrolo[2,3b]pyridin-4-yl)-1H-pyrazol-1-yl]propanenitrile; 3-cyclohexyl-3-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]propanenitrile; 3-cyclopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile; 3-cyclohexyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile; 3- 1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclopropylpropanenitrile; 4- cyclopropyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]butanenitrile; 1- [4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclopropylacetonitrile; 4-1-[(pyrrolidin-2-yl)methyl]-1H-pyrazol-4-yl-7H-pyrrolo[2,3-d]pyrimidine; 4-(1-[1-(methylsulfonyl)pyrrolidin-2-yl]methyl-1H-pyrazol-4-yl)-7H-pyrrolo[2,3-d]pyrimidine; 3-cyclopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]acrylonitrile; 3-(2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl)-cyclopentane-carbonitrile; 3- [3-(hydroxymethyl)cyclopentyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propanenitrile; 4- (1-benzyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine; 4-[1-(2-naphthylmethyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine; 4-(1-phenyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine; 3- [4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]benzonitrile; 4- methyl-3-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]benzonitrile; 4-[1 -(3,5-dimethoxybenzyl)-1 H-pyrazol-4-yl]-1 H-pyrrolo[2,3-b]pyridine; 4-[1 -(1 -phenylethyl)-1 H-pyrazol-4-yl]-1 H-pyrrolo[2,3-b]pyridine; 4-[1 -(cyclohexylmethyl)-l H-pyrazol-4-yl]-1 H-pyrrolo[2,3-b]pyridine; 3- {[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]methyl}benzonitrile; 2- {[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]methyl}benzonitrile; 4- {[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]methyl}benzonitrile; 1-phenyl-2-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]ethanone; 4-{1-[(5-methylisoxazol-3-yl)methyl]-1H-pyrazol-4-yl}-1H-pyrrolo[2,3-b]pyridine; 4-[1 -(tetrahydro-2H-pyran-2-ylmethyl)-1 H-pyrazol-4-yl]-1 H-pyrrolo[2,3-b]pyridine; 4-(1-cyclohex-2-en-1-yl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine; 4-(1-cyclohexyl-1 H-pyrazol-4-yl)-1 H-pyrrolo[2,3-b]pyridine; 4’- {[4-(1-H-pyrrolo[2,3-b]pyridin-4-yl)-1-H-pyrazol-1-yl]methyl}biphenyl-2-carbonitrile; 4-[1-(2-nitrobenzyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine; 4-{1-[2,6-dichloro-4-(trifluoromethyl)phenyl]-1H-pyrazol-4-yl}-1H-pyrrolo[2,3-b]pyridine; 4-[1-(3-nitrobenzyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine; 4-[1 -(2 bramobenzyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine; N-phenyl-2-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]propanamide; 4-{1-[3-(trifluoromethoxy)benzyl]-1H-pyrazol-4-yl}-1H-pyrrolo[2,3-b]pyridine; 4-{1-[2-fluoro-5-(trifluoromethyl)benzyl]-1H-pyrazol-4-yl}-1H-pyrrolo[2,3-b]pyridine; 4-{1-[3-(trifluoromethyl)benzyl]-1H-pyrazol-4-yl}-1H-pyrrolo[2,3-b]pyridine; 4-[1-(pyridin-3-ylmethyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine; 4-{1-[1-phenylbutyl]-1H-pyrazol-4-yl}-1H-pyrrolo[2,3-b]pyridine; 1- phenyl-2-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]propan-1-one; 4-[1-(2,6-dichlorobenzyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine; 4-[1-(2,6-dimethylphenyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine; 2- (4-(1 H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]-5-(trifluoromethyl)-benzonitrile; 4-[1-(4-bromo-3,5,6-trifluoropyridin-2-yl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine; 4-[1-(cyclopropylmethyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine; 4-[1-(2,5-dimethylphenyl)-1H-pyrazolo-4-yl]-1H-pyrrolo[2,3-b]pyridine; 4-[1-(2-methylphenyl)-1 H-pyrazol-4-yl]-1 H-pyrrolo[2,3-b]pyridine; 4-[1-(2-methoxyphenyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine; 3-{1-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]ethyl}benzonitrile; 3- chloro-4-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]benzonitrile; 4- [1-(1-cyclohexylethyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine; 4-fluoro-2-[4-(1 H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]benzonitrile; 2- fluoro-4-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]benzonitrile; 3- fluoro-4-[4-(1 H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1 -yl]benzonitrile; 4- (1-{1-[3-(trifluoromethyl)phenyl]ethyl}-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine; 4-[1-(3,5-dimethylphenyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine; 4-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]benzonitrile; {4-[4-(1 H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]phenyl}acetonitrile; 4-[1-(1-phenylpropyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine; 4-(1-{1-[4-(methylsulfonyl)phenyl]ethyl}-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine; 4-{1-[1-(3-fluoro-4-methoxyphenyl)ethyl]-1H-pyrazol-4-yl}-1H-pyrrolo[2,3-b]pyridine; 4-(1-{1-[2-(trifluoromethyl)phenyl]ethyl}-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine; 4-(1-{1-[3,5-bis(trifluoromethyl)phenyl]ethyl}-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine; 4-{1 -[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1 -yl]ethyl}benzonitrile; 4-{1-[4-nitro-2-(trifluoromethyl)phenyl]-1H-pyrazol-4-yl}-1H-pyrrolo[2,3-b]pyridine; 3- methyl-4-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]benzonitrile; 4- [1-(2-chlorophenyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine; 3- bromo-4-[4-(1 H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]benzonitrile; ethyl 4-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]benzoate; 4- {1-[2-chloro-6-nitro-4-(trifluoromethyl)phenyl]-1H-pyrazol-4-yl}-1H-pyrrolo[2,3-b]pyridine; 4-(1-{1-[4-(trifluoromethyl)phenyl]ethyl}-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine; 4-[1-(2,3-dihydro-1H-inden-1-yl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine; 4-[1 -(1,2,3,4-tetrahydronaphthalen-1-yl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine; 4-(1-{1-[2-chloro-5-(trifluoromethyl)phenyl]ethyl}-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine; 4-{1-[1-(2,4-dichloro-5-fluorophenyl)ethyl]-1H-pyrazol-4-yl}-1H-pyrrolo[2,3-b]pyridine; 4-[1-(1-cyclopentylethyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine; 4-[1-(1-methyl-3-phenylpropyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine; 4-[1-(1-cyclobutylethyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine; [2-[4-(1 H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]-5-(trifluoromethyl)phenyl]-acetonitrile; [5-[4-(1 H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]-2-(trifluoromethyl)phenyl]-acetonitrile; 4- {1-[4-phenylbut-3-en-1-yl]-1H-pyrazol-4-yl}-1H-pyrrolo[2,3-b]pyridine; [3-chloro-2-[4-(1 H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]-5-(trifluoromethyl)phenyl]-acetonitrile; 5- [4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]-2-(trifluoromethyl)-benzonitrile; 4-{1-[2-chloro-4-(trifluoromethyl)phenyl]-1H-pyrazol-4-yl}-1H-pyrrolo[2,3-b]pyridine; 4-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]-2-(trifluoromethyl)-benzonitrile; 2- [4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]benzonitrile; 3- chloro-2-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1 -yl]benzonitrile; 4- amino-5,6-difluoro-2-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]isophthalonitrile; 1- {[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]methyl}-cyclopropanecarbonitrile; 5- bromo-2-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]benzonitrile; 3-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]-4-(trifluoromethyl)-benzonitrile; 2- [4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]-3-(trifluoromethyl)-benzonitrile; 3- [4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]-4-(trifluoromethyl)benzamide; 3- [4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]cyclohexanone; 2-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1 -yl]cyclohexanol; 4- (1-{[1-(methylsulfonyl)piperidin-4-yl]methyl}-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine; 2-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]cyclohexanecarbonitrile; 4-{1-[2-(trifluoromethyl)phenyl]-1H-pyrazol-4-yl}-1H-pyrrolo[2,3-b]pyridine; 4-[1-(2,6-dichlorophenyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine; (4-{[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]methyl}cyclohexyl)methanol; 4-[1-(tetrahydrofuran-2-ylmethyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine; 4-[1-(1-cyclopentylpropyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine; 4-[1-(tetrahydrofuran-3-ylmethyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine; 2- chloro-3-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]benzonitrile; 3- [4-(1 H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]-3-(1,3-thiazol-5-yl)propanenitrile; 3-(1-methyl-1H-imidazol-5-yl)-3-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl] propane-nitrile; 3- [4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]-3-(3-thienyl)propanenitrile; {1-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]cyclopentyl}acetonitrile; 4- chloro-3-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]benzonitrile; 4-[4-(1 H pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]phthalonitrile; 3- methyl-4-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]benzaldehyde; 4- [1-(2-methyl-4-nitrophenyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine; 3- [4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]cyclopentanone; 4- [1-(3-furylmethyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine; 4-[1-(2-furylmethyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine; 3-{2-cyano-1-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]ethyl}benzonitrile; {3-methyl-4-[4-(1H-pyirolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]phenyl}methanol; 3-(1-benzofuran-2-yl)-3-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]propanenitrile; 3- (3-furyl)-3-[4-(1 H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1 -yl]propanenitrile; {3-methyl-4-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]phenyl}acetonitrile; 4- methyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]benzonitrile; 4-[1-(1-cyclopentylpropyl)-1H-pyrazol-4-yl]-7H-pyrrolo[2,3-d]pyrimidine; {1-[4-(7H-pyrrolo[2,3-d] pyrimidin-4-yl)-1 H-pyrazol-1-yl]cyclopentyl}acetonitrile; 3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}benzonitrile; 3- [4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]-3-(3-thienyl)propanenitrile; 4- chloro-3-[4-(7H-pyrro lo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]benzonitrile; 3-(3-furyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propanenitrile; 3-{1 -[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1 -yl]cyclopentyl}-propanenitrile; {1 -[4-(7H-pyrrolo[2,3-d] pyrimidin-4-yl)-1 H-pyrazol-1 -yl]cyclohexyl}acetonitrile; {3-methyl-4-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]phenyl}m ethanol; 3-pyridin-4-yl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1 -yl]propanenitrile; 3-pyridin-3-yl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile; 3-[4-(methylthio)phenyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile; 3-(3-methoxyphenyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile; 3-(4-methoxyphenyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-l H-pyrazol-1 -yl]propanenitdle; {3-methyl-4-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]phenyl}acetonitrile; 3-[4-(methylsulfinyl)phenyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]-propanenitrile; 3-[4-(methylsulfonyl)phenyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propanenitrile; 3-[3-(cyanomethoxy)phenyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1 -yl]-propanenitrile; 3-(6-chloropyridin-3-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propanenitrile; 5- {2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1 -yl]ethyl}pyridine-2-carbonitrile; 3-(3,5-dimethylisoxazol-4-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrmiidin-4-yl)-1 H-pyrazol-1-yl]-propanenitrile; 3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1 -yl]-3-[6-(trifluoromethyl)pyridin-3-yl]-propanenitrile; 3-(6-methoxypyridin-3-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl] propane-nitrile; 3-pyridin-2-yl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propanenitrile; 3-(6-bromopyridin-2-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propanenitrile; 6- {2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}pyridine-2-carbonitrile; 3-(5-bromopyridin-3-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile; 5-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}nicotinonitrile; 3-(2-methoxypyridin-3-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl] propane-nitrile; 3-[4-(cyanomethoxy)phenyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl] propane-nitrile; 3-[2-(cyanomethoxy)phenyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]-propanenitrile; 3-(3,5-dibromophenyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile; 5-{2-cyano-1-[4-(7H pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}isophthalonitrile; 3-[6-(dimethylamino)pyridin-2-yl]-3-[4-(7H-pyirolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]-propanenitrile; 3-(4-bromo-2-thienyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propanenitrile; 5-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}thiophene-3-carbonitrile; 3-(5-bromo-2-fluorophenyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]-propanenitrile; 3-(3-nitrophenyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile; 3-(5-bromo-2-methoxyphenyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propanenitrile; 3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}-4-methoxybenzo-nitrile; 3-(3-bromophenyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile; 3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}-4-fluorobenzonitrile; 3-[5-bromo-2-(cyanomethoxy)phenyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propanenitrile; 3- (4-bromo-2-furyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile; 4- (cyanomethoxy)-3-{2-cyano-1-[4-(7H-pyirolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}-benzonitrile; 3-(4-bromopyridin-2-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile; 2- {2-cyano-1-[4-(7H-pyirolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}isonicotinonitrile; 5- {2-cyano-1-[4-(7H-pyirolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}-3-furonitrile; 3- [2-bromo-5-(cyanomethoxy)phenyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propanenitrile; 4- (cyanomethoxy)-2-{2-cyano-1-[4-(7H-pyirolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-ethyl}benzonitrile; 3-pyrimidin-5-yl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile; 3- (2-bromopyridin-4-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile; 4- {2-cyano-1-[4-(7H-pyirolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}pyridine-2-carbo-nitrile; 3-(5-methoxypyridin-3-yl)-3-[4-(7H-pyirolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl] propane-nitrile; 3-(3-chlorophenyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile; 3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-3-[3-(trifluoromethyl)phenyl]-propanenitrile; 3-(3-phenoxyphenyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile; 3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-3-[3-(trifluoromethoxy)-phenyl]-propanenitrile; methyl 3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}benzoate; 3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}benzoic acid; 3-[3-(1H-pyrazol-4-yl)phenyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propanenitrile; 3- (3-aminophenyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile; N-(3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}phenyl)acetamide; N-(3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}phenyl)-methanesulfonamide; 4- {2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}thiophene-2-carbonitrile; 5- {2-cyano-1-[4-(7H pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}thiophene-2-carbonitrile; 3-[3-(morpholin-4-ylcarbonyl)phenyl]-3-[4-(7H-pyrrolo[23-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propanenitrile; N-(2-aminoethyl)-3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}-benzamide; 3-(5-formyl-3-thienyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile; 3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}-N-methylbenzamide; 2- cyano-N-(3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}phenyl)-acetamide; N-(3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}phenyl)-nicotinamide; N-(3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}phenyl)-N’-isopropylurea; isopropyl (3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}phenyl)-carbamate; 3- (5-phenylpyridin-3-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile; 3-(3,3’-bipyridin-5-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile; 3-(5-pyrimidin-5-ylpyridin-3-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propanenitrile; 3-[5-(1-methyl-1 H-pyrazol-4-yl)pyridin-3-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propaneni-trile; 3-(5-ethynylpyridin-3-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propane-nitrile; 3-[5-(phenylthio)pyridin-3-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propanenitrile; 3-(2-bromo-1,3-thiazol-5-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propanenitrile; 3-(5-morpholin-4-ylpyridin-3-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propanenitrile; 3-(1-methyl-1H-pyrazol-4-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propanenitrile; 3-(3-pyridin-3-ylphenyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile; 3-[5-(phenylsulfinyl)pyridin-3-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propanenitrile; 3-[5-(phenylsulfonyl)pyridin-3-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propanenitrile; 3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-3-[5-(1,3-thiazol-2-ylthio)pyridin-3-yl]propanenitrile; 3- [5-(ethylthio)pyridin-3-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propanenitrile; 4- {1-[1-methyl-2-(1H-1,2,4-triazol-1-yl)ethyl]-1H-pyrazol-4-yl}-7H-pyrrolo[2,3-d]pyrimidine; 1-phenyl-2-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propan-1-one; 3-[5-(ethylsulfinyl)pyridin-3-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propanenitrile; 3-[5-(ethylsulfonyl)pyridin-3-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propanenitrile; 3-[5-(cyclohexylthio)pyridin-3-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propanenitrile; 1-phenyl-2-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propan-1-ol; 3-[3-(ethylthio)phenyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile; 3-[3-(ethylsulfonyl)phenyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propanenitrile; 3-[3-(ethylsulfonyl)phenyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propanenitrile; 3-[5-(cyclohexylsulfonyl)pyridin-3-yl]-3-[4-(7H-pyrrolo[2,3-d]-,yrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile; 3-[5-(cyclohexylsulfinyl)pyridin-3-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile; 4-[1-(1-methyl-2-phenylethyl)-1H-pyrazol-4-yl]-7H-pyrrolo[2,3-d]pyrimidine; 4-{1-[1 -methyl-2-(3-thienyl)ethyl]-1 H-pyrazol-4-yl}-7H-pyrrolo[2,3-d]pyrimidine; 3- {1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}benzonitrile; 4- {1-[2-(1H-imidazol-1-yl)-1-methylethyl]-1H-pyrazol-4-yl}-7H-pyrrolo[2,3-d]pyrimidine; 4- {1-[1-methyl-2-(3-methyl-1,2,4-oxadiazol-5-yl)ethyl]-1 H-pyrazol-4-yl}-7H-pyrrolo[2,3-d]-pyrimidine; 3-[3-(methylsulfonyl)phenyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]-propanenitrile; 3-(3-pyridin-4-ylphenyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile; 3-[5-(isopropylthio)pyridin-3-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]-propanenitrile; 3-[5-(isopropylsulfinyl)pyridin-3-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propanenitrile; 3-[5-(isopropylsulfonyl)pyridin-3-yl]_l-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propanenitrile; 3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]-3-[5-(trifluoromethyl)pyridin-3-yl]-propanenitrile; 2- [4-(7Hpyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1yl]-N-[3-(trifluoromethyl)phenyl]-propanamide; N-2-naphthyl-2-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanamide; N-1-naphthyl-2-[4-(7H pyrrolo[2,3-d]pyrimidin-4-yl)-1H pyrazol-1-yl]propanamide; N-(3-cyanophenyl)-2-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propanamide; N-phenyl-2-[4-(7H-pyrrolo[2,3-d]_lpyrimidin-4-yl)-1 H-pyrazol-l-yl^butanamide; N-(4-phenoxyphenyl)-2-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]butanamide; N-2-naphthyl-2-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]butanamide; N-(3-cyanophenyl)-2-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]butanamide; N-biphenyl-4-yl-2-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]butanamide; N-(biphenyl-4-ylmediyl)-2-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-IH-pyrazol-1-yl]biitanamide; N-(biphenyl-3-ylmethyl)-2-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]butanamide; N-(4-cyanophenyl)-2-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]butanamide; N-1-naphthyl-2-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]butanamide; 5- {2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}-N-phenylnicotin-amide; N-(3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}phenyl)-3-(trifluoromethyl)benza-mide; N-(3-{[4-(7H-pyirolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]methyl}phenyl)-3-(trifluoromethyl)benzamide; 3- [3-(methylsulfonyl)phenyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]-propanenitrile; N-(3-{[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]methyl}phenyl)benzenesulfonamide; 3-{[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]methyl}-N-[3-(trifluoromethyl)-phenyl]benzamide; 3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}-N,N-dimethyl-benzenesulfonamide; N-benzyl-3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}benzene-sulfonamide; N-benzyl-3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}benzamide; 3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}-N-phenylbenzamide; 3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}-N-[3-(trifluoromethyl)phenyl]benza-mide; N-(3-cyanophenyl)-3-{[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]methyl}-benzamide; N-benzyl-3-{[4-(7H-pyrrolo_l[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]methyl}benzamide; N-1-naphthyl-3-{[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]methyl}benzamide; N-2-naphthyl-3-{[4-(7H-pyrrolo[2,3-d]_lpyrimidin-4-yl)-1 H-pyrazol-1-yl]methyl}benzamide; N-(3-{[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]methyl}phenyl)-2-naphthamide; N-(3-{[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]methyl}phenyl)-1-naphthamide; 2- phenyl-N-(3-{[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]methyl}phenyl)-acetamide; 3- chloro-N-(3-{[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]methyl}phenyl)-benzamide; N-(3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}phenyl)-2-naphthamide; N-(3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}phenyl)-1-naphthamide; N-(3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-ethyl}phenyl)-2-phenylacetamide; 3-cyano-N-(3-(2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl)phenyl)-benzamide; N-(3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}phenyl)-benzamide; N-(3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}phenyl)-4-(trifluoromethyl)benza-mide; N-(3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}phenyl)-N’-phenylurea; 3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}-N-[4-(trifluoromethyl)phenyl]benza-mide; 3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}-N-(4-methylphenyl)benzamide; N-(4-cyanophenyl)-3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-ethyl}benzamide; 3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}-N-2-naphthyl-benzamide; 3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}-N-1-naphthyl-benzamide; 3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}-N,N-dimethyl-benzamide; 3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}-N-pyridin-3-yl-benzamide; 3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}-N-methyl-N-phenyl-benzamide; 3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}-N-cyclohexyl-benzamide; N-(3-cyanophenyl)-3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}-benzamide; N-biphenyl-4-yl-3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-ethyl}benzamide; N-(4-chlorophenyl)-3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]-ethyl}benzamide; 3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}-N-(3,4-dimethylphenyl)benzamide; 3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}-N-isoxazol-3-yl-benzamide; 3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}-N-methyl-N-phenylbenzenesulfona-mide; 3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}-N-propylbenzene-sulfonamide; 3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}-N-phenyl-benzenesulfonamide; 3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}-N-2-naphthyl-benzenesulfonamide; 3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}-N-cyclopropyl-benzenesulfonamide; 3-[3-(piperidin-1-ylsulfonyl)phenyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]-propanenitrile; 3-[3-(morpholin-4-ylsulfonyl)phenyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]-propanenitrile; 3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}-N-(4-methylphenyl)-benzenesulfona-mide; 3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}-N-(3,4-dimethylphenyl)benzenesul-fonamide; 3-[3-(benzylsulfonyl)henyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]-propanenitrile; 3- [3-(benzylthio)phenyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propanenitrile; 4- {[(3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}phenyl)-sulfonyl]methyl}benzoni-trile; 3-(2-cyano-l-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}-N-methylbenzene-sulfonamide; 3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}-N-1-naphthyl-benzenesulfonamide; N-biphenyl-4-yl-3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}-benzenesulfonamide; 3-[3-(benzyloxy)phenyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propanenitrile; 3-{2-cyano-1-[4-(7H pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}-N-cyclohexyl-benzenesulfonamide; 3-[3-(3,4-dihydroisoquinolin-2(1 H)-ylsulfonyl)phenyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile; 3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}-N,N-diethyl-benzenesulfonamide; 3-{3-[(4-ethylpiperazin-1-yl)sulfonyl]phenyl}-3-[4-(7H-pyrrolo[2,3-dl-pyrimidin-4-yl)-1 H-pyrazol-1-yl]propaneni-trile; N-1,3-benzodioxol-5-yl-3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]-ethyl}benzenesul-fonamide; 3-{3-[(2,6-dimethylmorpholin-4-yl)sulfonyl]phenyl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]pro-panenitrile; 3-{3-[(4-oxopiperidin-1-yl)sulfonyl]phenyl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propaneni-trile; 3-[3-(isopropylsulfonyl)phenyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propanenitrile; 3-{3-[(cyclohexylmethyl)sulfonyl]phenyl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propanenitrile; 3-[3-(octahydroisoquinolin-2(1 H)-ylsulfonyl)phenyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]pro-panenitrile; 3-{3-[(2-phenylethyl)-sulfonyl]phenyl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propanenitrile; cis-4-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohexylacetonitrile; 3-cis-4-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]cyclohexylpropanenitrile; cis-4-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]cyclohexanecarbonitrile; 3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohexylacetonitrile; 5- ({cis-4-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]cyclohexyl}thio)-1 H-1,2,4-triazol-3-amine; [cis-4-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-1-(1 H-1,2,4-triazol-1-yl)cyclo-hexyl]acetonitrile; 3-[3-(piperazin-1-ylsulfonyl)phenyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile; 3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-3-[3-(thiomorpholin-4-ylsulfonyl)-phenyl]propanenitrile; 3-{3-[(4-hydroxypiperidin-1-yl)sulfonyl]phenyl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]pro-panenitrile; 3-[3-(isobutylsulfonyl)phenyl]-3-[4-(7H-pyrrolo[2 ,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]-propanenitrile; 3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-3-{3-[(tetrahydro-2H-pyran-4-ylmethyl)sulfonyl]phe- nyl}propanenitrile; 3-{3-[(3-furylmethyl)sulfonyl]phenyl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile; 3-{3-[(1,1-dioxidothiomorpholin-4-yl)sulfonyl]phenyl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1- yl]propanenitrile; 3-{3-[(pyridin-4-ylmethyl)sulfonyl]phenyl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile; 3-(3-hydroxy-1-[4-(7H-pyirolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propyl}-A/,A/-dimethyl-benzenesulfona- mide; 3- {1-[4-(7H-pyirolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]but-3-en-1-yl}benzonitrile; 4- {1-[1-(3-bromophenyl)but-3-en-1-yl]-1H-pyrazol-4-yl}-7H-pyrrolo[2,3-d]pyrimidine; 3- {4,4-difluoro-1-[4-(7H-pyirolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]but-3-en-1-yl}-benzonitrile; 4- (1-{4,4-difluoro-1-[3-(morpholin-4-ylsulfonyl)phenyl]but-3-en-1-yl}-1H-pyrazol-4-yl)-7H-pyirolo[2,3-d]pyrimi-dine; 4-(1-{1-[3-(ethylsulfonyl)phenyl]-4,4-difluorobut-3-en-1-yl}-1H-pyrazol-4-yl)-7H-pyrrolo[2,3-d]pyrimidine; 4-(1-{1-[3-(benzyloxy)phenyl]-4,4-difluorobut-3-en-1-yl}-1H-pyrazol-4-yl)-7H-pyrrolo[2,3-d]-pyrimidine; 4-(1-{4,4-difluoro-1-[3-(methylsulfonyl)phenyl]but-3-en-1-yl}-1H-pyrazol-4-yl)-7H-pyrrolo[2,3-d]pyrimidine; 3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]methyl}benzonitrile; 3- {1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]_,butyl}benzonitrile; 4- (1-{1-[3-(ethylsulfonyl)phenyl]-4,4-difluorobutyl}-1H-pyrazol-4-yl)-7H-pyrrolo[2,3-d]-pyrimidine; 4-[1-(1-cyclopentylbut-3-en-1-yl)-1H-pyrazol-4-yl]-7H-pyrrolo[2,3-d]pyrimidineD; 4-[1-(1-cyclopentylbutyl)-1H-pyrazol-4-yl]-7H-pyrrolo[2,3-d]pyrimidineD; 4-[1-(1-cyclopentyl-4,4-difluorobut-3-en-1-yl)-1H-pyrazol-4-yl]-7H-pyrrolo[2,3-d]pyrimidineD; 4-{1-[4,4-difluoro-1-(tetrahydrofuran-3-yl)but-3-en-1-yl]-1H-pyrazol-4-yl}-7H-pyrrolo[2,3-d]pyrimidine; 4-[1-(1-cyclopropyl-4,4-difluorobut-3-en-1-yl)-1H-pyrazol-4-yl]-7H-pyrrolo[2,3-d]pyrimidine; 4-(1-(1-cyclopentyl-4,4-difluorobutyl)-1H-pyrazol-4-yl]-7H-pyrrolo[2,3-d]pyrimidine; 3-(1-methylcyclopentyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile; 1-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}cyclopropane-carbonitrile; 3- [1-(methylsulfonyl)pyrrolidin-3-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile; 4- {1-[2,2,2-trifluoro-1 -(1 H-imidazol-2-ylmethyl)ethyl]-1H-pyrazol-4-yl}-7H-pyrrolo[2,3-d]pyrimidine; 4-(1-(1R)-2,2,2-trifluoro-1-[(4-methyl-1,3-thiazol-2-yl)methyl]ethyl-1H-pyrazol-4-yl)-7H-pyrrolo[2,3-d]pyrimi-dine; 4- {1-[1-(5-bromopyridin-3-yl)-4,4-difluorobut-3-en-1-yl]-1H-pyrazol-4-yl}-7Hpyrrolo[2,3-d]pyrimidine; 5- {4,4-difluoro-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]but-3-en-1-yl}nicotinonitrile; 3-[3-(pyrrolidin-1-ylsulfonyl)-phenyl]-3-[4-(7H-pyrrolo-[2,3-d]-pyrimidin-4-yl)-1H-pyrazol-1-yl]-propanenitrile; N-benzyl-3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}-N-methylbenzenesulfona-mide; 3-{[(3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}-,phenyl)sulfonyl]methyl}-benzoni- trile; 3-{3-[(2-naphthylmethyl)-sulfonyl]-phenyl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propaneni- trile; 3-{3-[(1-phenylethyl)sulfonyl]-phenyl}-3-[4-(7H-pyrrolo[2,3-d]-pyrimidin-4-yl)-1H-pyrazol-1-yl]-propanenitrile; 3-{2-cyano-1-[4-(7H-pyrrolo-[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}-N-(2-morpholin-4-ylethyl)-benze- nesulfonamide; 3-{2-cyano-1-[4-(7H-pyrrolo-[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}-N-[(1S)-1-phenylethyl]benzenesul- fonamide; 3-{2-cyano-1-[4-(7H-pyrrolo-[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}-N-phenyl-benzamide; 3-{2-cyano-1-[4-(7H pyrrolo-[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}-N-(tetrahydrofuran-2-yl-methyl)ben-zenesulfonamide; 3-{3-[(cyclopropylmethyl)sulfonyl]phenyl}-3-[4-(7 H-pyrrolo[2,3-d]pyrimidin-4-yI)-1 H-pyrazol-1-yl]propanenitrile trifluoroacetate; 3-{3-[(4-methylpiperazin-1-yl)-sulfonyl]phenyl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]pro-panenitrile; and 3-{3-[(1-oxidothiomorpholin-4-yl)sulfonyl]phenyl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]pro- panenitrile; or a pharmaceutically acceptable salt thereof. 9. A compound which is 3-cyclopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propanenitrile; or a pharmaceutically acceptable salt thereof; for use in a method of treating a disease selected from cachexia, a myelopro- liferative disorder, and cancer in a patient in need thereof, in combination with an additional pharmaceutical agent. 10. A compound which is (R)-3-cyclopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propanenitrile; or a pharmaceutically acceptable salt thereof; for use in a method of treating a disease selected from cachexia, a myeloproliferative disorder, and cancer in a patient in need thereof, in combination with an additional pharmaceutical agent. 11. A compound which is 3-cyclopropyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-pyrazol-1-yl]-propionitrile; or a pharmaceutically acceptable salt thereof; for use in a method of treating a disease selected from cachexia, a myeloproliferative disorder, and cancer in a patient in need thereof, in combination with an additional pharmaceutical agent. 12. The compound or salt for use according to any one of claims 1 to 11, wherein said additional pharmaceutical agent is a chemotherapeutic agent. 13. The compound or salt for use according to anyone of claims 1 to 11, wherein said method comprises administering said compound or salt and said additional pharmaceutical agent simultaneously to the patient. 14. The compound or salt for use according to anyone of claims 1 to 11, wherein said method comprises administering said compound, or said salt, and said additional pharmaceutical agent sequentially to said patient. 15. The compound or salt for use according to any one of claims 1 to 11, wherein about 5 to about 1000 mg of said compound or said salt is administered to said patient. 16. The compound or salt for use according to any one of claims 1 to 15, wherein the disease is a myeloproliferative disorder. 17. The compound or salt for use according to claim 16, wherein said myeloproliferative disorder is polycythemia vera (PV), essential thrombocythemia (ET), myeloid metaplasia with myelofibrosis (MMM), chronic myelogenous leukemia (CML), chronic myelomonocytic leukemia (CMML), hyperéosinophilie syndrome (HES), or systemic mast cell disease (SMCD). 18. The compound or salt for use according to claim 16, wherein said myeloproliferative disorder is polycythemia vera (PV). 19. The compound or salt for use according to claim 16, wherein said myeloproliferative disorder is essential thrombocythemia (ET). 20. The compound or salt for use according to claim 16, wherein said myeloproliferative disorder is myeloid metaplasia with myelofibrosis (MMM). 21. The compound or salt for use according to claim 16, wherein said myeloproliferative disorder is chronic myelogenous leukemia (CML). 22. The compound or salt for use according to claim 16, wherein said myeloproliferative disorder is chronic myelomonocytic leukemia (CMML). 23. The compound or salt for use according to any one of claims 1 to 15, wherein the disease is cancer. 24. The compound or salt for use according to claim 23, wherein said cancer is multiple myeloma. 25. The compound or salt for use according to claim 23, wherein said cancer is pancreatic cancer. 26. The compound or salt for use according to claim 23, wherein said cancer is leukemia. 27. The compound or salt for use according to claim 23, wherein said cancer is lymphoma. 28. The compound or salt for use according to claim 23, wherein said cancer is breast cancer. 29. The compound or salt for use according to claim 23, wherein said cancer is acute myelogenous leukemia. 30. The compound or salt for use according to claim 23, wherein said cancer is lung cancer. 31. The compound or salt for use according to claim 23, wherein said cancer is cancer of the head and neck. 32. The compound or salt for use according to claim 23, wherein said cancer is selected from prostate cancer, renal cancer, hepatic cancer, gastric cancer, thyroid cancer, glioblastoma, Karposi’s sarcoma, Castleman’s disease, melanoma, cutaneous T-cell lymphoma (CTCL), cutaneous Bell lymphoma, Sezary syndrome, mycosis fungoides, acute lymphoblastic leukemia, and colorectal cancer. 33. The compound or salt for use according to any one of claims 1 to 15, wherein said cachexia results from or is associated with cancer. 34. The compound or salt for use according to any one of claims 1 to 11, wherein said additional pharmaceutical agent is revlimid. 35. The compound or salt for use according to any one of claims 1 to 11, wherein said method further comprises administering a corticosteroid to said patient. 36. The compound or salt for use according to claim 35, wherein said corticosteroid is prednisone. 37. The compound or salt for use according to any one of claims 1 to 11, wherein said additional pharmaceutical agent is a DNA-damaging agent. 38. The compound or salt for use according to claim 37, wherein said DNA-damaging agent is selected from melphalan, doxorubicin, cyclophosphamide, vincristine, etoposide, and carmustine. 39. The compound or salt for use according to claim 24, wherein said additional pharmaceutical agent is selected from melphalan, melphalan plus prednisone, doxorubicin, dexamethasone, and bortezomib. 40. The compound or salt for use according to any one of claims 1 to 11, wherein said additional pharmaceutical agent is an anti-inflammatory compound. 41. The compound or salt for use according to any one of claims 1 to 11, wherein said additional pharmaceutical agent is a proteosome inhibitor, bortezomib, thalidomide, a Bcr-Abl inhibitor, a Flt-3 inhibitor, a RAF inhibitor, or a FAK inhibitor.
Patentansprüche 1. Verbindung der Formel:
oder ein pharmazeutisch unbedenkliches Salz davon; wobei: T, U und V unabhängig voneinander aus O, S, N, CR5 und NR6 ausgewählt sind; wobei derdurch das Kohlenstoffatom, das Stickstoffatom, U, T und V gebildete 5-gliedrige Ring aromatisch ist; X für N oder CR4 steht; n für 0 steht oder n für 1 steht und Y für C^g-Alkylen, C2_8-Alkenylen, (CR11R12)pC(0)(CR11R12)q, (CR11 R12)pC(0)NRc(CR11 R12)q, (CR11 R12)pC(0)0(CR11 R12)q oder(CR11R12)pOC(0)(CR11R12)q steht, wobei Ci_8-Alkylen oder C2_8-Alkenylen gegebenenfalls durch 1,2 oder 3 Halogene, OH, CN, Amino, C^-Alkylamino oder C2_8-Dialkylamino substituiert ist; Z für Aryl, Cycloalkyl, Heteroaryl oder Heterocycloalkyl steht, jeweils gegebenenfalls substituiert durch 1,2, 3, 4, 5 oder 6 Substituenten unabhängig voneinander ausgewählt aus Halogen, C1_4-Alkyl, C2.4-Alkenyl, C2.4-Al-kinyl, C^-Halogen-alkyl, C^^Hydroxyalkyl, C^-Cyanoalkyl, Cy1, CN, N02, ORa, SRa, C(0)Rb, C(0)NRcRd, C(0)0Ra, 0C(0)Rb, 0C(0)NRcRd, NRcRd, NRcC(0)Rb, NRcC(0)NRcRd; NRcC(0)0Ra, S(0)Rb, S(0)NRcRd, S(0)2Rb, NRcS(0)2Rb und S(0)2NRcRd;
Cy1 unabhängig ausgewählt ist aus Aryl, Heteroaryl, Cycloalkyl und Heterocycloalkyl, jeweils gegebenenfalls substituiert durch 1,2, 3, 4 oder 5 Substituenten unabhängig voneinander ausgewählt aus Halogen, C^^Alkyl, C2_4-Alkenyl, C^-Alkinyl, C^-Halogenalkyl, CN, N02, ORa", SRa", C(0)Rb", C(0)NRc"Rd", C(0)0Ra", 0C(0)Rb", 0C(0)NRc"Rd", NRc"Rd", NRc"C(0)Rb", NRc"C(0)0Ra", S(0)Rb", S(0)NRc"Rd", S(0)2Rb" und S(0)2NRc"Rd" ; R4 für H steht; R5 für H, Halogen, C^-Alkyl, C2.4-Alkenyl, C2_4-Alkinyl, C.|_4-Halogenalkyl, CN, N02, OR7, SR7, C(0)R8, C(0)NR9R1°, C(0)0R7, OC(0)R8, 0C(0)NR9R10, NR9R10, NR9C(0)R8, NR9C(0)0R7, S(0)R8, S(0)NR9R10, S(0)2R8, NR9S(0)2R8 oder S(0)2NR9R1° steht; R6 für H, C.|_4-Alkyl, C2_4-Alkenyl, C2.4-Alkinyl, C.,_4-Halogenalkyl, OR7, C(0)R8, C(0)NR9R10, C(0)0R7, S(0)R8, S(0)NR9R1°, S(0)2R8 oder S(0)2NR9R1° steht; R7fürH, C^g-Alkyl, C^g-Halogenalkyl, C2_6-Alkenyl, C2.6-Alkinyi, Aryl, Cycloalkyl, Heteroaryl, Heterocycloalkyl, Arylalkyl, Heteroarylalkyl, Cycloalkylalkyl oder Heterocycloalkylalkyl steht; R8fürH, C^g-Alkyl, C^g-Halogenalkyl, C2_6-Alkenyl, C2.6-Alkinyi, Aryl, Cycloalkyl, Heteroaryl, Heterocycloalkyl, Arylalkyl, Heteroarylalkyl, Cycloalkylalkyl oder Heterocycloalkylalkyl steht; R9 und R10 unabhängig voneinander aus H, C1.10-Alkyl, C^g-Halogenalkyl, C2.6-Alkenyl, C2_g-Alkinyl, C^g-Al-kylcarbonyl, Arylcarbonyl, C^g-Alkylsulfonyl, Arylsulfonyl, Aryl, Heteroaryl, Cycloalkyl, Heterocycloalkyl, Arylalkyl, Heteroarylalkyl, Cycloalkylalkyl und Heterocycloalkylalkyl ausgewählt sind; oder R9 und R10 zusammen mit dem Stickstoffatom, an das sie gebunden sind, eine 4-, 5-, 6- oder 7-gliedrige Heterocycloalkylgruppe bilden; R11 und R12 unabhängig voneinander aus H, Halogen, OH, CN, C^-Alkyl, C^^Halogenalkyl, C2_4-Alkenyl, C2_4-Alkinyi, C^-Hydroxyalkyl, C^-Cyanoalkyl, Aryl, Heteroaryl, Cycloalkyl und Heterocycloalkyl ausgewählt sind;
Ra und Ra" unabhängig voneinander aus H, C^-Alkyl, C^g-Halogenalkyl, C2.6-Alkenyl, C2_6-Alkinyl, Aryl, Cycloalkyl, Heteroaryl, Heterocycloalkyl, Arylalkyl, Heteroarylalkyl, Cycloalkylalkyl und Heterocycloalkylalkyl ausgewählt sind, wobei C^g-Alkyl, C^g-Halogenalkyl, C2.6-Alkenyl, C2_6-Alkinyl, Aryl, Cycloalkyl, Heteroaryl, Heterocycloalkyl, Arylalkyl, Heteroarylalkyl, Cycloalkylalkyl oder Heterocycloalkylalkyl gegebenenfalls substituiert istdurch 1,2 oder 3 Substituenten unabhängig voneinander ausgewählt aus OH, CN, Amino, Halogen, C^g-Alkyl, C^g-Halogenalkyl, Aryl, Arylalkyl, Heteroaryl, Heteroarylalkyl, Cycloalkyl und Heterocycloalkyl;
Rb und Rb" unabhängig voneinander aus H, C^-Alkyl, C^g-Halogenalkyl, C2_6-Alkenyl, C2.6-Alkinyl, Aryl, Cycloalkyl, Heteroaryl, Heterocycloalkyl, Arylalkyl, Heteroarylalkyl, Cycloalkylalkyl und Heterocycloalkylalkyl ausgewählt sind, wobei C^g-Alkyl, Ci_g-Halogenalkyl, C2_6-Alkenyl, C2_6-Alkinyl, Aryl, Cycloalkyl, Heteroaryl, Heterocycloalkyl, Arylalkyl, Heteroarylalkyl, Cycloalkylalkyl oder Heterocycloalkylalkyl gegebenenfalls substituiert istdurch 1,2 oder 3 Substituenten unabhängig voneinander ausgewählt aus OH, CN, Amino, Halogen, C^g-Alkyl, C^g-Halogenalkyl, Aryl, Arylalkyl, Heteroaryl, Heteroarylalkyl, Cycloalkyl und Heterocycloalkyl;
Rc und Rd unabhängig voneinander aus H, Ci_g-Alkyl, C^g-Halogenalkyl, C2_6-Alkenyl, C2_6-Alkinyl, Aryl, Heteroaryl, Cycloalkyl, Heterocycloalkyl, Arylalkyl, Heteroarylalkyl, Cycloalkylalkyl und Heterocycloalkylalkyl ausgewählt sind, wobei C1_6-Alkyl, Ci_g-Halogenalkyl, C2_6-Alkenyl, C2_6-Alkinyl, Aryl, Heteroaryl, Cycloalkyl, Heterocycloalkyl, Arylalkyl, Heteroarylalkyl, Cycloalkylalkyl oder Heterocycloalkylalkyl gegebenenfalls substituiert istdurch 1,2oder3 Substituenten unabhängig voneinander ausgewählt aus OH, CN, Amino, Halogen, C^g-Alkyl, C^g-Halogenalkyl, Aryl, Arylalkyl, Heteroaryl, Heteroarylalkyl, Cycloalkyl und Heterocycloalkyl; oder Rc und Rd zusammen mit dem Stickstoffatom, an das sie gebunden, eine 4-, 5-, 6- oder 7-gliedrige Heterocycloalkylgruppe bilden, welche gegebenenfalls durch 1,2 oder 3 Substituenten unabhängig voneinander ausgewählt aus OH, CN, Amino, Halogen, C^-Alkyl, C^g-Halogenalkyl, Aryl, Arylalkyl, Heteroaryl, Heteroarylalkyl, Cycloalkyl und Heterocycloalkyl substituiert ist;
Rc" und Rd" unabhängig voneinander ausgewählt sind aus H, C^^-Alkyl, C^g-Halogenalkyl, C2_6-Alkenyl, C2-6-Alkinyl, Aryl, Heteroaryl, Cycloalkyl, Heterocycloalkyl, Arylalkyl, Heteroarylalkyl, Cycloalkylalkyl und Hete-rocycloalkylalkyl, wobei C.,_10-Alkyl, C^g-Halogenalkyl, C2.6-Alkenyl, C2.6-Alkinyl, Aryl, Heteroaryl, Cycloalkyl, Heterocycloalkyl, Arylalkyl, Heteroarylalkyl, Cycloalkylalkyl oder Heterocycloalkylalkyl gegebenenfalls substituiert ist durch 1,2 oder 3 Substituenten, unabhängig voneinander ausgewählt aus OH, CN, Amino, Halogen, C1_6-Alkyl, C^g-Halogenalkyl, Aryl, Arylalkyl, Heteroaryl, Heteroarylalkyl, Cycloalkyl und Heterocycloalkyl; oder Rc" und Rd" zusammen mit dem Stickstoffatom, an das sie gebunden sind, eine 4-, 5-, 6- oder 7-gliedrige Heterocycloalkylgruppe bilden, welche gegebenenfalls durch 1,2 oder 3 Substituenten unabhängig voneinander ausgewählt aus OH, CN, Amino, Halogen, C^g-Alkyl, C^g-Halogenalkyl, Aryl, Arylalkyl, Heteroaryl, Heteroarylalkyl, Cycloalkyl und Heterocycloalkyl substituiert ist; p für 0, 1,2, 3, 4, 5 oder 6 steht und q für 0, 1,2, 3, 4, 5 oder 6 steht; zur Verwendung in einem Verfahren zur Behandlung einer Krankheit ausgewählt aus Kachexie, einer myeloproliferativen Erkrankung und Krebs in einem Patienten, derdies benötigt, in Kombination mit einem zusätzlichen pharmazeutischen Mittel. 2. Verbindung oder Salz zur Verwendung nach Anspruch 1, wobei die Verbindung eine Verbindung der Formel II:
oder ein pharmazeutisch unbedenkliches Salz davon ist. 3. Verbindung oder Salz zur Verwendung nach Anspruch 1 oder 2, wobei X für N steht. 4. Verbindung oder Salz zur Verwendung nach einem der Ansprüche 1 bis 3, wobei n für 0 steht. 5. Verbindung oder Salz zur Verwendung nach einem der Ansprüche 1 bis 3, wobei n für 1 steht und YfürC^g-Alkylen, gegebenenfalls substituiert durch 1,2 oder 3 Halogene, OH, CN, Amino, c1-4 -Alkylamino oder C2_8-Dialkylamino, steht. 6. Verbindung oder Salz zur Verwendung nach einem der Ansprüche 1 bis 5, wobei Z für Cycloalkyl, gegebenenfalls substituiert durch 1,2,3,4,5 oder 6 Substituenten unabhängig voneinander ausgewählt aus Halogen, C^-Alkyl, C2.4-Alkenyl, C2_4-Alkinyl, C^-Halogenalkyl, C^^Hydroxyalkyl, C^^Cyanoalkyl, Cy1, CN, N02, ORa, SRa, C(0)Rb, C(0)NRcRd, C(0)0Ra, 0C(0)Rb, 0C(0)NRcRd, NRcRd, NRcC(0)Rb, NRcC(0)NRcRd; NRcC(0)0Ra, S(0)Rb, S(0)NRcRd, S(0)2Rb, NRcS(0)2Rb und S(0)2NRcRd, steht. 7. Verbindung oder Salz zur Verwendung nach einem der Ansprüche 1 bis 5, wobei Z für Heterocycloalkyl, gegebenenfalls substituiert durch 1, 2, 3, 4, 5 oder 6 Substituenten unabhängig voneinander ausgewählt aus Halogen, C.|_4-Alkyl, C2_4-Alkenyl, C2_4-Alkinyl, C^^Halogenalkyl, C^-Hydroxyalkyl, C^-Cyanoalkyl, Cy1, CN, N02, ORa, SRa, C(0)Rb, C(0)NRcRd, C(0)0Ra, 0C(0)Rb, 0C(0)NRcRd, NRcRd, NRcC(0)Rb, NRcC(0)NRcRd; NRcC(0)0Ra, S(0)Rb, S(0)NRcRd, S(0)2Rb, NRcS(0)2Rb und S(0)2NRcRd, steht. 8. Verbindung oder Salz zur Verwendung nach Anspruch 1, wobei die Verbindung aus: 4-[1-(1-Methyl-3-pyrazol-1-ylpropyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridin; 4-[1-(3-lmidazol-1-yl-1-methylpropyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridin; 4-Cyclopentyl-4-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)pyrazol-1-yl]butyronitril; 3-Cyclopropyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)pyrazol-1-yl]-propionitril; 3-Cyclopentyl-3-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]propannitril; 3-Cyclohexyl-3-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]propannitril; 3-Cyclopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propannitril; 3-Cyclohexyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propannitril; 3- 1-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclopropylpropannitril; 4- Cyclopropyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]butannitril; 1- [4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclopropylacetonitril; 4-1-[(Pyrrolidin-2-yl)methyl]-1H-pyrazol-4-yl-7H-pyrrolo[2,3-d]pyrimidin; 4-(1-[1-(Methylsulfonyl)pyrrolidin-2-yl]methyl-1H-pyrazol-4-yl)-7H-pyrrolo[2,3-d]pyrimidin; 3-Cyclopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]acrylonitril; 3-(2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl)cyclopentancarbonitril; 3- [3-(Hydroxymethyl)cyclopentyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propannitril; 4- (1-Benzyl-1 H-pyrazol-4-yl)-1 H-pyrrolo[2,3-b]pyridin; 4-[1-(2-Naphthylmethyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridin; 4-(1-Phenyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridin; 3- [4-(1H-Pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]benzonitril; 4- Methyl-3-[4-(1 H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1 -yl]benzonitril; 4-[1-(3,5-Dimethoxybenzyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridin; 4-[1-(1-Phenylethyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridin; 4-[1-(Cyclohexylmethyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridin; 3- {[4-(IH-Pyrrolo[2,3-b]pyridin-4-yl)-IH-pyrazol-1-yl]methyl}benzonitril; 2- {[4-(1H-Pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]methyl}benzonitril; 4- {[4-(1H-Pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]methyl}benzonitril; 1-Phenyl-2-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yljethanon; 4-{1-[(5-Methylisoxazol-3-yl)methyl]-1H-pyrazol-4-yl}-1H-pyrrolo[2,3-b]pyridin; 4-[1-(Tetrahydro-2H-pyran-2-ylmethyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridin; 4-(1-Cyclohex-2-en-1-yl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridin; 4-(1-Cyclohexyl-1 H-pyrazol-4-yl)-1 H-pyrrolo[2,3-b]pyridin; 4’-{[4-(1H-Pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1 -yl]methyl}biphenyl-2-carbonitril; 4-[1-(2-Nitrobenzyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridin; 4-{1-[2,6-Dichlor-4-(trifluormethyl)phenyl]-1H-pyrazol-4-yl}-1H-pyrrolo[2,3-b]pyridin; 4-[1-(3-Nitrobenzyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridin; 4-[1-(2-Brombenzyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridin; N-Phenyl-2-[4-(1 H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]propanamid; 4-{ 1-[3-(Trifluormethoxy) benzyl]-1 H-pyrazol-4-yl}-1 H-pyrrolo[2,3-b]pyridin; 4-{1-[2-Fluor-5-(trifluormethyl)benzyl]-1H-pyrazol-4-yl}-1H-pyrrolo[2,3-b]pyridin; 4-{1-[3-(Trifluormethyl)benzyl]-1 H-pyrazol-4-yl}-1 H-pyrrolo[2,3-b]pyridin; 4-[1 -(Pyrid in-3-y Im ethyl)-1 H-pyrazol-4-yl]-1 H-pyrrolo[2,3-b]pyridin; 4-{1-[1-Phenylbutyl]-1H-pyrazol-4-yl}-1H-pyrrolo[2,3-b]pyridin; 1- Phenyl-2-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]propan-1-on; 4-[1-(2,6-Dichlorbenzyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridin; 4-[1-(2,6-Dimethylphenyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridin; 2- [4-(1H-Pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]-5-(trifluormethyl)-benzonitril; 4-[1-(4-Brom-3,5,6-trifluorpyridin-2-yl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridin; 4-[1-(Cyclopropylmethyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridin; 4-[1-(2,5-Dimethylphenyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridin; 4-[1-(2-Methylphenyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridin; 4-[1-(2-Methoxyphenyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridin; 3- {1-[4-(1H-Pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]ethyl}benzonitril; 3- Chlor-4-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]benzonitril; 4- [1-(1-Cyclohexylethyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridin; 4-Fluor-2-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]benzonitril; 2- Fluor-4-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yljbenzonitril; 3- Fluor-4-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]benzonitril; 4- (1-{1-[3-(Trifluormethyl)phenyl]ethyl}-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridin; 4-[1-(3,5-Dimethylphenyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridin; 4-[4-(1 H-Pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1 -yl]benzonitril; {4-[4-(1 H-Pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1 -yl]phenyl}acetonitril; 4-[1-(1-Phenylpropyl)-1 H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridin; 4-(1-{1-[4-(Methylsulfonyl)phenyl]ethyl}-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridin; 4-{1-[1-(3-Fluor-4-methoxyphenyl)ethyl]-1H-pyrazol-4-yl}-1H-pyrrolo[2,3-b]pyridin; 4-(1-{1-[2-(Trifluormethyl)phenyl]ethyl}-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridin; 4-(1-(1-[3,5-Bis(trifluormethyl)phenyl]ethyl}-1H-pyrazol-4-yl)-1 H-pyrrolo[2,3-b]pyridin; 4-(1-(4-(1 H-Pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]ethyl}benzonitril; 4-{1-[4-Nitro-2-(trifluormethyl)phenyl]-1H-pyrazol-4-yl}-1H-pyrrolo[2,3-b]pyridin; 3- Methyl-4-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]benzonitril; 4- [1-(2-Chlorphenyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridin; 3- Brom-4-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yljbenzonitril; 4- (4-(1 H-Pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]benzoesäureethyl; 4-{1-[2-Chlor-6-nitro-4-(trifluormethyl)phenyl]-1 H-pyrazol-4-yl}-1 H-pyrrolo[2,3-b]pyridin; 4-(1-{1-[4-(Trifluormethyl)phenyl]ethyl}-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridin; 4-(1-(2,3-Dihydro-1 H-inden-1-yl)-1 H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridin; 4-(1-(1,2,3,4-Tetrahydronaphthalen-1-yl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridin; 4-(1-{1-[2-Chlor-5-(trifluorrmethyl)phenyl]ethyl}-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridin; 4-(1-(1 -(2,4-Dichlor-5-fluorphenyl)ethyl]-1 H-pyrazol-4-yl}-1 H-pyrrolo[2,3-b]pyridin; 4-(1-(1-Cyclopentylethyl)-1 H-pyrazol-4-yl]-1 H-pyrrolo[2,3-b]pyridin; 4-(1-(1-Methyl-3-phenylpropyl)-1 H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridin; 4-(1-(1-Cyclobutylethyl)-1 H-pyrazol-4-yl]-1 H-pyrrolo[2,3-b]pyridin; [2-(4-(1 H-Pyrrolo[2,3-b] pyridin-4-yl)-1 H-pyrazol-1-yl]-5-(trifluormethyl)phenyl]acetonitril; [5-(4-(1 H-Pyrrolo[2,3-b] pyridin-4-yl)-1 H-pyrazol-1-yl]-2-(trifluormethyl)phenyl]acetonitril; 4- {1-[4-Phenylbut-3-en-1-yl]-1H-pyrazol-4-yl}-1H-pyrrolo[2,3-b]pyridin; [3-Chlor-2-[4-(1 H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1 -yl]-5-(trifluormethyl)phenyl]acetonitril; 5- (4-(1 H-Pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1 -yl]-2-(trifluormethyl)benzonitril; 4-{1-[2-Chlor-4-(trifluormethyl)phenyl]-1H-pyrazol-4-yl}-1H-pyrrolo[2,3-b]pyridin; 4-(4-(1 H-Pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1 -yl]-2-(trifluormethyl)benzonitril; 2- (4-(1 H-Pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1 -yl]benzonitril; 3- Chlor-2-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1 -yljbenzonitril; 4- Amino-5,6-difluor-2-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]isophtalonitril; 1- {[4-(1 H-Pyrrolo[2,3-b] pyridin-4-yl)-1 H-pyrazol-1-yl]methyl}cyclopropancarbonitril; 5- Brom-2-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]benzonitril; 3-(4-(1 H-Pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1 -yl]-4-(trifluormethyl)benzonitril; 2- (4-(1 H-Pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]-3-(trifluormethyl)benzonitril; 3- (4-(1 H-Pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1 -yl]-4-(trifluormethyl)benzamid; 3- (4-(1 H-Pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]cyclohexanon; 2-[4-(1 H-Pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]cyclohexanol; 4- (1-{[1-(Methylsulfonyl)piperidin-4-yl]methyl}-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridin; 2-[4-(1 H-Pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]cyclohexancarbonitril; 4-{1-[2-(Trifluormethyl)phenyl]-1H-pyrazol-4-yl}-1H-pyrrolo[2,3-b]pyridin; 4-(1 -(2,6-Dichlorphenyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridin; (4-((4-(1 H-Pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]methyl}cyclohexyl)methanol ; 4-[1-(Tetrahydrofuran-2-ylmethyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridin; 4-[1-(1-Cyclopentylpropyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridin; 4-[1-(Tetrahydrofuran-3-ylmethyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridin; 2- Chlor-3-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yljbenzonitril; 3- (4-(1 H-Pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1 -yl]-3-(1,3-thiazol-5-yl)propannitril; 3-(1-Methyl-1H-imidazol-5-yl)-3-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]propannitril; 3- (4-(1 H-Pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1 -yl]-3-(3-thienyl)propannitril; (1-[4-(1 H-Pyrrolo[2,3-b] pyridin-4-yl)-1 H-pyrazol-1 -yl]cyclopentyl}acetonitril; 4- Chlor-3-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]benzonitril; 4-(4-(1 H-Pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1 -yl]phtalonitril; 3- Methyl-4-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1 -yljbenzaldehyd; 4- [1-(2-Methyl-4-nitrophenyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridin; 3- [4-(1 H-Pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]cyclopentanon; 4- [1-(3-Furylmethyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridin; 4-[1-(2-Furylmethyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridin; 3-{2-Cyano-1-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]ethyl}benzonitril; {3-Methyl-4-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]phenyl}methanol; 3-(1-Benzofuran-2-yl)-3-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]propannitril; 3- (3-Furyl)-3-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]propannitril; {3-Methyl-4-[4-(1FI-pyrrolo[2,3-b]pyridin-4-yl)-1FI-pyrazol-1-yl]phenyl}acetonitril; 4- Methyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]benzonitril; 4-[1-(1-Cyclopentylpropyl)-1 FI-pyrazol-4-yl]-7FI-pyrrolo[2,3-d]pyrimidin; {1-[4-(7FI-Pyrrolo[2,3-d]pyrimidin-4-yl)-1 FI-pyrazol-1-yl]cyclopentyl}acetonitril; 3-{2-Cyano-1-[4-(7FI-pyrrolo[2,3-d]pyrimidin-4-yl)-1FI-pyrazol-1-yl]ethyl}benzonitril; 3- [4-(7FI-Pyrrolo[2,3-d]pyrimidin-4-yl)-1FI-pyrazol-1-yl]-3-(3-thienyl)propannitril; 4- Chlor-3-[4-(7FI-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1 -yl]benzonitril; 3-(3-Furyl)-3-[4-(7FI-pyrrolo[2,3-d]pyrimidin-4-yl)-1FI-pyrazol-1-yl]propannitril; 3-{1-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]cyclopentyl}propannitril; {1-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]cyclohexyl}acetonitril; {3-Methyl-4-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]phenyl}m ethanol; 3-Pyridin-4-yl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propannitril; 3-Pyridin-3-yl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propannitril; 3-[4-(Methylthio)phenyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1 -yl]propannitril; 3-(3-Methoxyphenyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propannitril; 3-(4-Methoxyphenyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propannitril; {3-Methyl-4-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]phenyl}acetonitril; 3-[4-(Methylsulfinyl)phenyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propannitril; 3-[4-(Methylsulfonyl)phenyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propannitril; 3-[3-(Cyanomethoxy)phenyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propannitril; 3-(6-Chlorpyridin-3-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1 -yl]propannitril ; 5- {2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}pyridin-2-carbonitril; 3-(3,5-Dim ethyl isoxazol-4-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propannitril; 3-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-3-[6-(Trifluormethyl)pyridin-3-yl]propannitril; 3-(6-Methoxypyridin-3-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propannitril; 3-Pyridin-2-yl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propannitril; 3-(6-Brompyridin-2-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propannitril; 6- {2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}pyridin-2-carbonitril; 3-(5-Brompyridin-3-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propannitril; 5-{2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}nicotinonitril; 3-(2-Methoxypyridin-3-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propannitril; 3-[4-(Cyanomethoxy)phenyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propannitril; 3-[2-(Cyanomethoxy)phenyl]-3-[4-(7H-pyrrolo)2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propannitril; 3-(3,5-Dibromphenyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propannitril; 5-{2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}isophthalonitril; 3-[6-(Dimethylamino)pyridin-2-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1 -yl]propannitril; 3-(4-Brom-2-thienyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propannitril; 5-{2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}thiophen-3-carbonitril; 3-(5-Brom-2-fluorphenyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propannitril; 3-(3-Nitrophenyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propannitril; 3-(5-Brom-2-methoxyphenyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]-propannitril; 3-{2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}-4-methoxybenzonitril; 3-(3-Bromphenyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propannitril; 3-{2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}-4-fluorbenzonitril; 3-[5-Brom-2-(cyanomethoxy)phenyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propannitril; 3- (4-Brom-2-furyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propannitril; 4- (Cyanomethoxy)-3-{2-cyano-1 -[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1 -yl]ethyl}benzonitril; 3-(4-Brompyridin-2-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propannitril; 2- {2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}isonicotinonitril; 5- {2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1 -yl]ethyl}-3-furonitril; 3- [2-Brom-5-(cyanomethoxy)phenyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propannitril; 4- (Cyanomethoxy)-2-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}benzonitril; 3-Pyrimidin-5-yl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propannitril; 3- (2-Brompyridin-4-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propannitril; 4- {2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}pyridin-2-carbonitril; 3-(5-Methoxypyridin-3-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propannitril; 3-(3-Chlorphenyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propannitril; 3-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-3-[3-(trifluormethyl)phenyl]propannitril; 3-(3-Phenoxyphenyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propannitril; 3-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-3-[3-(trifluormethoxy)phenyl]propannitril; 3-{2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}benzoesäuremethylester; 3-{2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}benzoesäure; 3-[3-(1H-Pyrazol-4-yl)phenyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-l-yl]propannitril; 3- (3-Aminophenyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propannitril; N-(3-{2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}phenyl)acetamid; N-(3-{2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}phenyl)methansulfonamid; 4- {2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}thiophen-2-carbonitril; 5- {2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}thiophen-2-carbonitril; 3-[3-(Morpholin-4-ylcarbonyl)phenyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propannitril; N-(2-Aminoethyl)-3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}benzamid; 3-(5-Formyl-3-thienyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propannitril; 3-{2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}-N-methylbenzamid; 2- Cyano-N-(3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}phenyl)acetamid; N-(3-{2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}phenyl)nicotinamid; N-(3-{2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}phenyl)-N’-isopropylharnstoff; (3-{2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}phenyl)isopropylcarbamat; 3- (5-Phenylpyridin-3-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propannitril; 3-(3,3’-Bipyridin-5-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propannitril; 3-(5- Pyrimidin-5-ylpyridin-3-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrim idin-4-yl)-1 H-pyrazol-1 -yl]-propannitril; 3-[5-(1 -Methyl-1 H-pyrazol-4-yl)pyridin-3-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propannitril; 3-(5-Ethinylpyridin-3-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propannitril; 3-[5-(Phenylth io)pyridin-3-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propannitril; 3-(2-Brom-1,3-thiazol-5-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propannitril; 3-(5-Morpholin-4-ylpyridin-3-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propannitril; 3-(1-Methyl-1H-pyrazol-4-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propannitril; 3-(3-Pyridin-3-ylphenyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propannitril; 3-[5-(Phenylsulfinyl)pyridin-3-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propannitril; 3-[5-(Phenylsulfonyl)pyridin-3-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]-propannitril; 3-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]-3-[5-(1,3-thiazol-2-ylthio)pyridin-3-yl]propannitril; 3- [5-(Ethylthio)pyridin-3-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propannitril; 4- {1-[1-Methyl-2-(1 H-1,2,4-triazol-1-yl)ethyl]-1 H-pyrazol-4-yl}-7H-pyrrolo[2,3-d]pyrimidin; 1-Phenyl-2-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propan-1-on; 3-[5-(Ethylsulfinyl)pyridin-3-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propannitril; 3-[5-(Ethylsulfonyl)pyridin-3-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propannitril; 3-[5-(Cyclohexylthio)pyridin-3-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]-propannitril; 1-Phenyl-2-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propan-1-ol; 3-[3-(Ethylthio)phenyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propannitril; 3-[3-(Ethylsulfinyl)phenyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propannitril; 3-[3-(Ethylsulfonyl)phenyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propannitril; 3-[5-(Cyclohexylsulfonyl)pyridin-3-yl]-3-[4-(7H-pyrrolo[2,3-d]-pyrimidin-4-yl)-1 H-pyrazol-1-yl]propannitril; 3- [5-(Cyclohexylsulfinyl)pyridin-3-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1 -yl]propannitril ; 4- [1-(1-Methyl-2-phenylethyl)-1H-pyrazol-4-yl]-7H-pyrrolo[2,3-d]pyrimidin; 4-{1-[1-Methyl-2-(3-thienyl)ethyl]-1H-pyrazol-4-yl}-7H-pyrrolo[2,3-d]pyrimidin; 3- {1-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}benzonitril; 4- {1-[2-(1H-lmidazol-1-yl)-1-methylethyl]-1H-pyrazol-4-yl}-7H-pyrrolo[2,3-d]pyrimidin; 4-{1-[1-Methyl-2-(3-methyl-1,2,4-oxadiazol-5-yl)ethyl]-1 H-Pyrazol-4-yl}-7H-pyrrolo[2,3-d]pyrimidin; 3-[3-(Methylsulfonyl)phenyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propannitril; 3-(3-Pyridin-4-ylphenyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propannitril; 3-[5-(lsopropylthio)pyridin-3-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propannitril; 3-[5-(lsopropylsulfinyl)pyridin-3-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1 -yl]propannitril; 3-[5-(lsopropylsulfonyl)pyridin-3-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propannitril; 3-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1 -yl]-3-[5-(trifluormethyl)pyridin-3-yl]propannitril ; 2- [4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-N-[3-(trifluormethyl)phenyl]propanamid; N-2-Naphthyl-2-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanamid; N-1-Naphthyl-2-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanamid; N-(3-Cyanophenyl)-2-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1 -yl]propanamid; N-Phenyl-2-[4-(7H-pyrrolo[2,3-d]-pyrim idin-4-yl)-1 H-pyrazol-1-yl]-butanamid; N-(4-Phenoxyphenyl)-2-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]butanamid; N-2-Naphthyl-2-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]butanamid; N-(3-Cyanophenyl)-2-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]butanamid; N-Biphenyl-4-yl-2-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]butanamid; N-(Biphenyl-4-ylmethyl)-2-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]butanamid; N-(Biphenyl-3-ylmethyl)-2-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]butanamid; N-(4-Cyanophenyl)-2-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]butanamid; N-1 -Naphthyl-2-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1 -yl]butanamid; 5-{2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}-N-phenylnicotinamid; N-(3-{2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}phenyl)-3-(trifluormethyl)benzamid; N-(3-{[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]methyl}phenyl)-3-(trifluormethyl)benzamid; 3- [3-(Methylsulfonyl)phenyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propannitril; N-(3-{[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]methyl}phenyl)benzolsulfonamid; 3-{[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]methyl}-N-[3-(trifluormethyl)phenyl]benzamid; 3-{2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}-N,N-dimethylbenzolsulfonamid; N-Benzyl-3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}benzolsulfonam id; N-Benzyl-3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}benzam id; 3-{2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}-N-phenylbenzamid; 3-{2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrim idin-4-yl)-1 H-pyrazol-1-yl]ethyl}-N-[3-(trifluormethyl)phenyl]benzamid; N-(3-Cyanophenyl)-3-{[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]methyl}benzamid; N-Benzyl-3-{[4-(7H-pyrrolo-[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]methyl}benzamid; N-1-Naphthyl-3-{[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]methyl}benzam id; N-2-Naphthyl-3-{[4-(7H-pyrrolo[2,3-d]-pyrimidin-4-yl)-1 H-pyrazol-1-yl]methyl}benzamid; N-(3-{[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]methyl}phenyl)-2-naphthamid; N-(3-{[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]methyl}phenyl)-1-naphthamid; 2- Phenyl-N-(3-{[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]methyl}phenyl)acetamid; 3- Chlor-N-(3-{[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]methyl}phenyl)benzamid; N-(3-{2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}phenyl)-2-naphtham id; N-(3-{2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}phenyl)-1-naphthamid; N-(3-{2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}phenyl)-2-phenylacetamid; 3-Cyano-N-(3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}phenyl)benzamid; N-(3-12-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}phenyl)benzamid; N-(3-{2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}phenyl)-4-(trifluormethyl)benzamid; N-(3-{2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}phenyl)-N’-phenylharnstoff; 3-{2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}-N-[4-(trifluormethyl)phenyl]benzamid; 3-{2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}-N-(4-methylphenyl)benzamid; N-(4-Cyanophenyl)-3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}benzamid; 3-{2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrim idin-4-yl)-1 H-pyrazol-1-yl]ethyl}-N-2-naphthylbenzamid; 3-{2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}-N-1-naphthylbenzamid; 3-{2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}-N,N-dimethylbenzamid; 3-{2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}-N-pyridin-3-yl-benzamid; 3-{2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}-N-methyl-N-phenylbenzamid; 3-{2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrim idin-4-yl)-1 H-pyrazol-1-yl]ethyl}-N-cyclohexylbenzamid; N-(3-Cyanophenyl)-3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}benzamid; N-Biphenyl-4-yl-3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}benzamid; N-(4-Chlorphenyl)-3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}benzamid; 3-{2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}-N-(3,4-dimethylphenyl)benzamid; 3-{2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}-N-isoxazol-3-yl-benzamid; 3-{2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}-N-methyl-N-phenylbenzolsulfonamid; 3-{2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}-N-propylbenzolsulfonam id; 3-{2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}-N-phenylbenzolsulfonamid; 3-{2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}-N-2-naphthylbenzolsulfonamid; 3-{2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}-N-cyclopropylbenzolsulfonamid; 3-[3-(Piperidin-1 -ylsulfonyl)phenyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1 -yl]-propannitril; 3-[3-(Morpholin-4-ylsulfonyl)phenyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propannitril; 3-{2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}-N-(4-methylphenyl)-benzolsulfona-mid; 3-{2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}-N-(3,4-dimethylphenyl)benzolsulfon-amid; 3-[3-(Benzylsulfonyl)phenyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propannitril; 3- [3-(Benzylthio)phenyl]-3-[4-(7H-pyrrolo[2,3-d]pyrim idin-4-yl)-1 H-pyrazol-1-yl]propannitril; 4- {[(3-{2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}phenyl)sulfonyl]methyl}benzonitril; 3-{2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}-N-methylbenzolsulfonamid; 3-{2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}-N-1-naphthylbenzolsulfonamid; N-Biphenyl-4-yl-3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}-benzolsulfonamid; 3-[3-(Benzyloxy)phenyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propannitril; 3-{2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}-N-cyclohexylbenzolsulfonamid; 3-[3-(3,4-Dihydroisochinolin-2(1H)-ylsulfonyl)phenyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propannitril; 3-{2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]ethyl}-N,N-diethylbenzolsulfonamid; 3-{3-[(4-Ethylpiperazin-1-yl)sulfony I] phenyl}-3-[4-(7H-pyrrolo[2,3-d]-pyrimidin-4-yl)-1 H-pyrazol-1-yl]propannit- ril; N-1,3-Benzodioxol-5-yl-3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]-ethyl}benzolsulfon-amid; 3-{3-[(2,6-Dimethylmorpholin-4-yl)sulfonyl]phenyl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]pro- pannitril; 3-{3-[(4-Oxopiperidin-1-yl)sulfonyl]phenyl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propannitril; 3-[3-(lsopropylsulfonyl)phenyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propannitril; 3-{3-[(Cyclohexylmethyl)sulfonyl]phenyl}-3-[4-(7H-pynOlo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propannitril; 3-[3-(Octahydroisochinolin-2(1H)-ylsulfonyl)phenyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]pro- pannitril; 3-{3-[(2-Phenylethyl)sulfonyl]phenyl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1 -yl]propannitril; cis-4-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohexylacetonitril; 3-cis-4-[4-(7H-Pyrrolo[2,3-d] pyrimidin-4-yl)-1 H-pyrazol-1 -yl]cyclohexylpropannitril; cis-4-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohexancarbonitril; 3-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]cyclohexylacetonitril; 5- ({cis-4-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]cyclohexyl}thio)-1H-1,2,4-triazol-3-amin; [cis-4-[4-(7H-Pyrrolo[2,3-d]pyrim idin-4-yl)-1 H-pyrazol-1-yl]-1-(1 H-1,2,4-triazol-1-yl)cyclohexyl]acetonitril; 3-[3-(Piperazin-1-ylsulfonyl)phenyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propannitril; 3-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-3-[3-(thiomorpholin-4-ylsulfonyl)phenyl]propannitril; 3-{3-[(4-Hydroxypiperid in-1-yl)sulfonyl]phenyl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propan-nitril; 3-[3-(lsobutylsulfonyl)phenyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propannitril; 3-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-3-{3-[(tetrahydro-2H-pyran-4-ylmethyl)sulfonyl]phe- nyljpropannitril; 3-{3-[(3-Furylmethyl)sulfonyl]phenyl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propannitril; 3-{3-[(1,1-Dioxidothiomorpholin-4-yl)sulfonyl]phenyl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propannitril; 3-{3-[(Pyridin-4-ylmethyl)sulfonyl]phenyl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propannitril; 3-{3-Hydroxy-1-[4-(7H-pyrrolo[2,3-d] pyri m id in-4-y I )-1 H-pyrazol-1 -yl]propyl}-/\/,/\/-dimethylbenzolsulfonamid; 3- {1 - [4-(7H-Pyrro lo[2,3-d] pyri m id in-4-y I )-1 H-pyrazol-1 -yl]but-3-en-1-yl}benzonitril; 4- {1-[1-(3-Bromphenyl)but-3-en-1-yl]-1H-pyrazol-4-yl}-7H-pyrrolo[2,3-d]pyrimidin; 3- {4,4-Difluor-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1 -yl]but-3-en-1 -yljbenzonitril; 4- (1-{4,4-Difluor-1-[3-(morpholin-4-ylsulfonyl)phenyl]but-3-en-1-yl}-1H-pyrazol-4-yl)-7H-pyrrolo[2,3-d]pyrimi-din; 4-(1-{1-[3-(Ethylsulfonyl)phenyl]-4,4-difluorbut-3-en-1-yl}-1H-pyrazol-4-yl)-7H-pyrrolo[2,3-d]pyrimidin; 4-(1-{1-[3-(Benzyloxy)phenyl]-4,4-difluorbut-3-en-1-yl}-1H-pyrazol-4-yl)-7H-pyrrolo[2,3-d]-pyrimidin; 4-(1-{4,4-Difluor-1-[3-(methylsulfonyl)phenyl]but-3-en-1-yl}-1H-pyrazol-4-yl)-7H-pyrrolo[2,3-d]pyrimidin; 3-{[4-(7H-Pyrrolo[2,3-d] pyrimidin-4-yl)-1 H-pyrazol-1-yl]methyl}benzonitril; 3-{1-[4-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]butyl}benzonitril; 4-(1-{1-[3-(Ethylsulfonyl)phenyl]-4,4-difluorbutyl}-1H-pyrazol-4-yl)-7H-pyiTolo[2,3-d]pyrimidin; 4-[1-(1-Cyclopentylbut-3-en-1-yl)-1H-pyrazol-4-yl]-7H-pyirolo[2,3-d]pyrimidin; 4-[1-(1-Cyclopentylbutyl)-1 H-pyrazol-4-yl]-7H-pyrrolo[2,3-d]pyrimidin; 4-[1-(1-Cyclopentyl-4,4-difluorbut-3-en-1-yl)-1H-pyrazol-4-yl]-7H-pyirolo[2,3-d]pyrimidin; 4-{1-[4,4-Difluor-1-(tetrahydrofuran-3-yl)but-3-en-1-yl]-1H-pyrazol-4-yl}-7H-pyrrolo[2,3-d]pyrimidin; 4-[1-(1-Cyclopropyl-4,4-difluorbut-3-en-1-yl)-1H-pyrazol-4-yl]-7H-pyiTolo[2,3-d]pyrimidin; 4-[1-(1-Cyclopentyl-4,4-difluorbutyl)-1H-pyrazol-4-yl]-7H-pyrrolo[2,3-d]pyrimidin; 3-(1-Methylcyclopentyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propannitril; 1-{2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}cyclopropancarbonitril; 3- [1-(Methylsulfonyl)pyrrolidin-3-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propannitril; 4- {1-[2,2,2-Trifluor-1-(1H-imidazol-2-ylmethyl)ethyl]-1H-pyrazol-4-yl}-7H-pyrrolo[2,3-d]pyrimidin; 4-(1-(1 R)-2,2,2-Trifluor-1-[(4-methyl-1,3-thiazol-2-yl)methyl]ethyl-1H-pyrazo1-4-yl)-7H-pyrrolo[2,3-d]pyrimidin; 4- {1-[1-(5-Brompyridin-3-yl)-4,4-difluorbut-3-en-1-yl]-1H-pyrazol-4-yl}-7H-pyrrolo[2,3-d]pyrimidin; 5- {4,4-Difluor-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]but-3-en-1-yl}nicotinonitril; 3-[3-(Pyrrolidin-1-ylsulfonyl)phenyl]-3-[4-(7H-pyrrolo-[2,3-d]-pyrimidin-4-yl)-1H-pyrazol-1-yl]-propannitril; N-Benzyl-3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}-N-methylbenzolsulfonamid; 3-{[(3-{2-Cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}-phenyl)sulfonyl]methyl}-benzonit-ril; 3-{3-[(2-Naphthylmethyl)sulfonyl]phenyl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-y1)-1H-pyrazol-1-yl]propannitril; 3-{3-[(1-Phenylethyl)sulfonyl]phenyl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propannitril; 3-{2-Cyano-1-[4-(7H-pyrrolo-[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}-N-(2-morpholin-4-ylethyl)-benzolsul- fonamid; 3-{2-Cyano-1-[4-(7H-pyrrolo-[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}-N-[(1S)-1-phenylethyl]benzolsulfon- amid; 3-{2-Cyano-1-[4-(7H-pyrrolo-[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}-N-phenylbenzamid; 3-{2-Cyano-1-[4-(7H-pyrrolo-[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]ethyl}-N-(tetrahydrofuran-2-yl-methyl)ben- zolsulfonamid;
Trifluoressigsäure-3-{3-[(cyclopropylmethyl)-sulfonyl]phenyl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyra-zol-1 -yl]propannitrilester; 3-,3-[(4-Methylpiperazin-1-yl)sulfonyl]phenyl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propannit- ril; und 3-{3-[(1-Oxidothiomorpholin-4-yl)sulfonyl]phenyl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]pro- pannitril; oder einem pharmazeutisch unbedenklichen Salz davon ausgewählt ist. 9. Verbindung, bei der es sich um 3-Cyclopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propannitril oderein pharmazeutisch unbedenkliches Salz davon handelt, zur Verwendung in einem Verfahren zur Behandlung einer Krankheit ausgewählt aus Kachexie, einer myeloproliferativen Erkrankung und Krebs in einem Patienten, der dies benötigt, in Kombination mit einem zusätzlichen pharmazeutischen Mittel. 10. Verbindung, bei der es sich um (R)-3-Cyclopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propannitril oderein pharmazeutisch unbedenkliches Salz davon handelt, zur Verwendung in einem Verfahren zur Behandlung einer Krankheit ausgewählt aus Kachexie, einer myeloproliferativen Erkrankung und Krebs in einem Patienten, der dies benötigt, in Kombination mit einem zusätzlichen pharmazeutischen Mittel. 11. Verbindung, bei der es sich um 3-Cyclopropyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-pyrazol-1-yl]propionitril oder ein pharmazeutisch unbedenkliches Salz davon handelt, zur Verwendung in einem Verfahren zur Behandlung einer Krankheit ausgewählt aus Kachexie, einer myeloproliferativen Erkrankung und Krebs in einem Patienten, der dies benötigt, in Kombination mit einem zusätzlichen pharmazeutischen Mittel. 12. Verbindung oder Salz zur Verwendung nach einem der Ansprüche 1 bis 11, wobei es sich bei dem zusätzlichen pharmazeutischen Mittel um ein Chemotherapeutikum handelt. 13. Verbindung oder Salz zur Verwendung nach einem der Ansprüche 1 bis 11, wobei das Verfahren das gleichzeitige Verabreichen der Verbindung bzw. des Salzes und des zusätzlichen pharmazeutischen Mittels an den Patienten umfasst. 14. Verbindung oder Salz zur Verwendung nach einem der Ansprüche 1 bis 11, wobei das Verfahren das Verabreichen der Verbindung oder des Salzes und des zusätzlichen pharmazeutischen Mittels nacheinander an den Patienten umfasst. 15. Verbindung oder Salz zur Verwendung nach einem der Ansprüche 1 bis 11, wobei etwa 5 bis etwa 1000 mg der Verbindung oder des Salzes dem Patienten verabreicht werden. 16. Verbindung oder Salz zur Verwendung nach einem der Ansprüche 1 bis 15, wobei es sich bei der Krankheit um eine myeloproliferative Erkrankung handelt. 17. Verbindung oder Salz zur Verwendung nach Anspruch 16, wobei es sich bei der myeloproliferativen Erkrankung um Polycythemia vera (PV), essenzielle Thrombozythämie (ET), myeloische Metaplasie mit Myelofibrose (MMM), chronische myeloische Leukämie (CML), chronische myelomonozytäre Leukämie (CMML), hyperäosinophiles Syndrom (HES) oder systemische Mastzellenkrankheit (SMCD) handelt. 18. Verbindung oder Salz zur Verwendung nach Anspruch 16, wobei es sich bei der myeloproliferativen Erkrankung um Polycythemia vera (PV) handelt. 19. Verbindung oder Salz zur Verwendung nach Anspruch 16, wobei es sich bei der myeloproliferativen Erkrankung um essenzielle Thrombozythämie (ET) handelt. 20. Verbindung oder Salz zur Verwendung nach Anspruch 16, wobei es sich bei der myeloproliferativen Erkrankung um myeloische Metaplasie mit Myelofibrose (MMM) handelt. 21. Verbindung oder Salz zur Verwendung nach Anspruch 16, wobei es sich bei der myeloproliferativen Erkrankung um chronische myeloische Leukämie (CML) handelt. 22. Verbindung oder Salz zur Verwendung nach Anspruch 16, wobei es sich bei der myeloproliferativen Erkrankung um chronische myelomonozytäre Leukämie (CMML) handelt. 23. Verbindung oder Salz zur Verwendung nach einem der Ansprüche 1 bis 15, wobei es sich bei der Krankheit um Krebs handelt. 24. Verbindung oder Salz zur Verwendung nach Anspruch 25, wobei es sich bei dem Krebs um multiples Myelom handelt. 25. Verbindung oder Salz zur Verwendung nach Anspruch 23, wobei es sich bei dem Krebs um Bauchspeicheldrüsenkrebs handelt. 26. Verbindung oder Salz zur Verwendung nach Anspruch 23, wobei es sich bei dem Krebs um Leukämie handelt. 27. Verbindung oder Salz zur Verwendung nach Anspruch 23, wobei es sich bei dem Krebs um Lymphom handelt. 28. Verbindung oder Salz zur Verwendung nach Anspruch 23, wobei es sich bei dem Krebs um Brustkrebs handelt. 29. Verbindung oder Salz zur Verwendung nach Anspruch 23, wobei es sich bei dem Krebs um akute myeloische
Leukämie handelt. 30. Verbindung oder Salz zur Verwendung nach Anspruch 23, wobei es sich bei dem Krebs um Lungenkrebs handelt. 31. Verbindung oder Salz zur Verwendung nach Anspruch 23, wobei es sich bei dem Krebs um Kopf- und Halskrebs handelt. 32. Verbindung oder Salz zur Verwendung nach Anspruch 23, wobei der Krebs ausgewählt ist aus Prostatakrebs, Nierenkrebs, Leberkrebs, Magenkrebs, Schilddrüsenkrebs, Glioblastom, Karposi-Sarkom, Castleman-Krankheit, Melanom, kutanem T-Zellen-Lymphom (CTCL), kutanem B-Zellen-Lymphom, Sezary-Syndrom, Mycosisfungoides, akuter lymphoblastischer Leukämie und Kolorektalkrebs. 33. Verbindung oder Salz zur Verwendung nach einem der Ansprüche 1 bis 15, wobei die Kachexie von Krebs herrührt oder mit Krebs assoziiert ist. 34. Verbindung oder Salz zur Verwendung nach einem der Ansprüche 1 bis 11, wobei es sich bei dem zusätzlichen pharmazeutischen Mittel um Revlimid handelt. 35. Verbindung oder Salz zur Verwendung nach einem der Ansprüche 1 bis 11, wobei das Verfahren ferner das Verabreichen eines Corticosteroids an den Patienten umfasst. 36. Verbindung oder Salz zur Verwendung nach Anspruch 35, wobei es sich bei dem Corticosteroid um Prednison handelt. 37. Verbindung oder Salz zur Verwendung nach einem der Ansprüche 1 bis 11, wobei es sich bei dem zusätzlichen pharmazeutischen Mittel um ein DNA schädigendes Mittel handelt. 38. Verbindung oder Salz zur Verwendung nach Anspruch 37, wobei das DNA schädigende Mittel aus Melphalan, Doxorubicin, Cyclophosphamid, Vincristin, Etoposid und Carmustin ausgewählt ist. 39. Verbindung oder Salz zur Verwendung nach Anspruch 24, wobei das zusätzliche pharmazeutische Mittel aus Melphalan, Melphalan plus Prednison, Doxorubicin, Dexamethason und Bortezomib ausgewählt ist. 40. Verbindung oder Salz zur Verwendung nach einem der Ansprüche 1 bis 11, wobei es sich bei dem zusätzlichen pharmazeutischen Mittel um eine entzündungshemmende Verbindung handelt. 41. Verbindung oder Salz zur Verwendung nach einem der Ansprüche 1 bis 11, wobei es sich bei dem zusätzlichen pharmazeutischen Mittel um einen Proteosomeninhibitor, Bortezomib, Thalidomid, einen Bcr-Abl-Inhibitor, einen Flt-3-lnhibitor, einen RAF-Inhibitoroder einen FAK-Inhibitor handelt.
Revendications 1. Composé de formule :
ou l’un de ses sels de qualité pharmaceutique ; où : T, U et V sont indépendamment choisis parmi O, S, N, CR5 et NR6 ; le cycle à 5 chaînons formé par l’atome de carbone, l’atome d’azote, U, T et V étant aromatique ; X représente N ou CR4; n est égal à 0 ; ou n est égal à 1 et Y représente un groupement alkylène en Ci_8, alcénylène en C2_8, (CR11R12)pC(0)(CR11R12)q, (CR11R12)pC(0)NRc(CR11R12)q (CR11R12)pC(0)0(CR11R12)q ou (CR11R12)p0C(0)(CRllRl2)q, ledit groupement alkylène en ΰ1-8 ou alcénylène en C2.8 étant éventuellement substitué par 1,2 ou 3 groupements halogéno, OH, CN, amino, (alkyle en C1_4)-amino ou di-(alkyle en C2_8)-amino ; Z représente un groupement aryle, cycloalkyle, hétéroaryle ou hétérocycloalkyle, chacun étant éventuellement substitué par 1, 2, 3, 4, 5 ou 6 substituants indépendants choisis parmi les groupements halogéno, alkyle en 0^4, alcényle en C2_4, alcynyle en C2-4, halogénoalkyle en Cf.4, hydroxyalkyle en C^. cyanoalkyle en C^, Cy1, CN, N02, ORa, SRa, C(0)Rb, C (0)NRcRd, C(0)0Ra, 0C(0)Rb, OC(0)NRcRd, NRcRd, NRcC(0)Rb, NRcC(0)NRcRd, NRcC(0)0Ra, S(0)Rb, S (0)NRcRd, S (0)2Rb, NRCS (0)2Rb et S (0)2NRcRd ;
Cy1 est indépendamment choisi parmi les groupements aryle, hétéroaryle, cycloalkyle et hétérocycloalkyle, chacun étant éventuellement substitué par 1, 2, 3, 4 ou 5 substituants indépendamment choisis parmi les groupements halogéno, alkyle en C^, alcényle en C2.4, alcynyle en C2_4, halogénoalkyle en C1_4, CN, N02, 0Ra , SRa", C(0)Rb", C(0)NRc"Rd", C(0)0Ra”, 0C(0)Rb”, 0C(0)NRc”Rd”, NRc"Rd", NRc"C(0)Rb”, NRc'C(0)0Ra”, S(0)Rb", S(0)NRc"Rd", S(0)2Rb" et S(0)2NRc"Rd"; R4 représente H ; R5 représente un atome H u un groupement halogéno, alkyle en C^, alcényle en C2_4, alcynyle en C2^, halogénoalkyle en C^, CN, N02, OR7, SR7, C(0)R8, C(0)NR9R10, C(0)0R7, 0C(0)R8, OC(0)NR9R10, NR9R10, NR9C(0)R8, NR9C(0)0R7, S(0)R8, S(0)NR9R1°, S(0)2R8, NR9S(0)2R8 ou S(0)2NR9R10 ; R6 représente H ou un groupement alkyle en C^, alcényle en C2^, alcynyle en C2_4, halogénoalkyle en C^, OR7, C(0)R8, C(0)NR9R10, C(0)0R7, S(0)r8, S(0)NR9R10, S(0)2R8 ou S(0)2NR9R10; R7 représente H ou un groupement alkyle en C^, halogénoalkyle en C-|_6, alcényle en C2_6, alcynyle en C2_g, aryle, cycloalkyle, hétéroaryle, hétérocycloalkyle, arylalkyle, hétéroarylalkyle, cycloalkylalkyle ou hétérocycloalkylalkyle ; R8 représente H ou un groupement alkyle en C^, halogénoalkyle en C^g, alcényle en C2_6, alcynyle en C2_g, aryle, cycloalkyle, hétéroaryle, hétérocycloalkyle, arylalkyle, hétéroarylalkyle, cycloalkylalkyle ou hétérocycloalkylalkyle ; R9 et R10 sont indépendamment choisis parmi H et les groupements alkyle en C^q, halogénoalkyle en C^, alcényle en C2_6, alcynyle en C2_6, (alkyle en C.|_6)-carbonyle, arylcarbonyle, (alkyle en C.|_6)-sulfonyle, arylsul-fonyle, aryle, hétéroaryle, cycloalkyle, hétérocycloalkyle, arylalkyle, hétéroarylalkyle, cycloalkylalkyle et hétérocycloalkylalkyle ; ou R9 et R10 forment ensemble et avec l’atome N auquel ils sont liés un groupement hétérocycloalkyle à 4, 5, 6 ou 7 chaînons ; R11 et R12 sont indépendamment choisis parmi H et les groupements halogéno, OH, CN, alkyle en C.|_4. halogénoalkyle en C-|_4, alcényle en C2^, alcynyle en 02_φ hydroxyalkyle en C.|_4, cyanoalkyle en C^, aryle, hétéroaryle, cycloalkyle et hétérocycloalkyle ;
Ra et Ra" sont indépendamment choisis parmi H et les groupements alkyle en C^g, halogénoalkyle en C^. alcényle en C2_6, alcynyle en C2.6, aryle, cycloalkyle, hétéroaryle, hétérocycloalkyle, arylalkyle, hétéroarylalkyle, cycloalkylalkyle et hétérocycloalkylalkyle, lesdits groupements alkyle en C^g, halogénoalkyle en C^g, alcényle en C2_6, alcynyle en C2_6, aryle, cycloalkyle, hétéroaryle, hétérocycloalkyle, arylalkyle, hétéroarylalkyle, cycloalkylalkyle ou hétérocycloalkylalkyle étant éventuellement substitués par 1,2 ou 3 substituants indépendamment choisis parmi les groupements OH, CN, amino, halogéno, alkyle en C^g, halogénoalkyle en C^g, aryle, arylalkyle, hétéroaryle, hétéroarylalkyle, cycloalkyle et hétérocycloalkyle ;
Rb et Rb" sont indépendamment choisis parmi H et les groupements alkyle en C^g, halogénoalkyle en C^, alcényle en C2_6, alcynyle en C2_6, aryle, cycloalkyle, hétéroaryle, hétérocycloalkyle, arylalkyle, hétéroarylalkyle, cycloalkylalkyle et hétérocycloalkylalkyle, lesdits groupements alkyle en C^g, halogénoalkyle en C^g, alcényle en C2_6, alcynyle en C2_6, aryle, cycloalkyle, hétéroaryle, hétérocycloalkyle, arylalkyle, hétéroarylalkyle, cycloalkylalkyle ou hétérocycloalkylalkyle étant éventuellement substitués par 1,2 ou 3 substituants indépendamment choisis parmi OH, CN, amino, halogéno, alkyle en C^, halogénoalkyle en C^g, aryle, arylalkyle, hétéroaryle, hétéroarylalkyle, cycloalkyle et hétérocycloalkyle ;
Rc et Rd sont indépendamment choisis parmi H et les groupements alkyle en C^g. halogénoalkyle en C^, alcényle en C2_6, alcynyle en C2_6, aryle, hétéroaryle, cycloalkyle, hétérocycloalkyle, arylalkyle, hétéroarylalkyle, cycloalkylalkyle et hétérocycloalkylalkyle, lesdits groupements alkyle en Ci_10, halogénoalkyle en C^g, alcényle en C2_6, alcynyle en C2_6, aryle, hétéroaryle, cycloalkyle, hétérocycloalkyle, arylalkyle, hétéroarylalkyle, cycloalkylalkyle ou hétérocycloalkylalkyle étant éventuellement substitués par 1,2 ou 3 substituants indépendamment choisis parmi OH, CN, amino, halogéno, alkyle en C^g, halogénoalkyle en Cr6, aryle, arylalkyle, hétéroaryle, hétéroarylalkyle, cycloalkyle ou hétérocycloalkyle ; ou Rc et Rd forment ensemble et avec l’atome N auxquels ils sont liés un groupement hétérocycloalkyle à 4, 5, 6 ou 7 chaînons éventuellement substitué par 1,2 ou 3 substituants indépendamment choisis parmi les groupements OH, CN, amino, halogéno, alkyle en 01-6, halogénoalkyle en C^g, aryle, arylalkyle, hétéroaryle, hétéroarylalkyle, cycloalkyle et hétérocycloalkyle ;
Rc" et Rd" sont indépendamment choisis parmi H et les groupements alkyle en C^o, halogénoalkyle en C^, alcényle en C2_6, alcynyle en C2_6, aryle, hétéroaryle, cycloalkyle, hétérocycloalkyle, arylalkyle, hétéroarylalkyle, cycloalkylalkyle et hétérocycloalkylalkyle, lesdits groupements alkyle en Ci_i0. halogénoalkyle en C^g, alcényle en C2_6, alcynyle en C2_6, aryle, hétéroaryle, cycloalkyle, hétérocycloalkyle, arylalkyle, hétéroarylalkyle, cy- cloalkylalkyle ou hétérocycloalkylalkyle étant éventuellement substitués par 1,2 ou 3 substituants indépendamment choisis parmi les groupements OH, CN, amino, halogéno, alkyle en C-|_6, halogénoalkyle en C-|_6, aryle, arylalkyle, hétéroaryle, hétéroarylalkyle, cycloalkyle et hétérocycloalkyle ; ou Rc" et Rd" forment ensemble et avec l’atome N auquel ils sont liés un groupement hétérocycloalkyle à 4, 5, 6 ou 7 chaînons éventuellement substitué par 1,2 ou 3 substituants indépendamment choisis parmi les groupements OH, CN, amino, halogéno, alkyle en C^g. halogénoalkyle en C^, aryle, arylalkyle, hétéroaryle, hétéroarylalkyle, cycloalkyle et hétérocycloalkyle ; p est égal à 0,1,2, 3, 4, 5 ou 6 ; et q est égal à 0,1,2, 3, 4, 5 ou 6 ; pour utilisation dans une méthode de traitement d’une maladie choisie parmi la cachexie, un trouble myélopro-liferatif et un cancer chez un patient en ayant besoin, en combinaison avec un agent pharmaceutique supplémentaire. 2. Composé ou sel pour utilisation selon la revendication 1, où le composé est un composé de Formule II :
3. Composé ou sel pour utilisation selon la revendication 1 ou 2, où X représente N. 4. Composé ou sel pour utilisation selon l’une quelconque des revendications 1 à 3, où n est égal à 0. 5. Composé ou sel pour utilisation selon l’une quelconque des revendications 1 à 3, où n est égal à 1 et Y représente un groupement alkylène en Ci_8 éventuellement substitué par 1,2 ou 3 groupements halogéno, OH, CN, amino, (alkyle en C-^-amino ou di-(alkyle en C2_8) -amino. 6. Composé ou sel pour utilisation selon l’une quelconque des revendications 1 à 5, où Z représente un groupement cycloalkyle éventuellement substitué par 1,2, 3,4, 5 ou 6 substituants indépendants choisis parmi les groupements halogéno, alkyle en C1_4, alcényle en C2_4, alcynyle en C2_4, halogénoalkyle en C^, hydroxyalkyle en C1_4, cya-noalkyle en C^, Cy1, CN, N02, ORa, SRa, C(0)Rb, C(0)NRcRd, C(0)0Ra, 0C(0)Rb, 0C(0)NRcRd, NRcRd, NRcC(0)Rb, NRcC(0)NRcRd NRcC(0)ORa, S(0)Rb, S(0)NRcRd, S(0)2Rb, NRcS(0)2Rb et S(0)2NRcRd. 7. Composé ou sel pour utilisation selon l’une quelconque des revendications 1 à 5, où Z représente un groupement hétérocycloalkyle éventuellement substitué par 1,2, 3, 4, 5 ou 6 substituants indépendants choisis parmi les groupements halogéno, alkyle en C^, alcényle en C2.4, alcynyle en C2_4, halogénoalkyle en C^, hydroxyalkyle en Ci_4, cyanoalkyle en C^, Cy1, CN, N02, ORa, SRa, C(0)Rb, C (0)NRcRd, C(0)0Ra, 0C(0)Rb, OC (0)NRcRd, NRcRd, NRcC(0)Rb, NRcC(0)NRcRd, NRcC(0)0Ra, S (0)Rb, S (0)NRcRd, S(0)2Rb, NRcS(0)2Rb et S(0)2NRcRd. 8. Composé ou sel pour utilisation selon la revendication 1, où ledit composé est choisi parmi les suivants : 4-[1-(1-méthyl-3-pyrazol-1-yl-propyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine ; 4-[1-(3-imidazol-1-yl-1-méthyl-propyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine ; 4-cyclopentyl-4-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-pyrazol-1-yl]-butyronitrile ; 3-cyclopropyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-pyrazol-1-yl]-propionitrile ; 3-cyclopentyl-3-[4-(1 H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]propanenitrile ; 3-cyclohexyl-3-[4-(1 H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]propanenitrile ; 3-cyclopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile ; 3-cyclohexyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile ; 3- 1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclopropylpropanenitrile ; 4- cyclopropyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]butanenitrile ; 1- [4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclopropylacétonitrile ; 4-1-[(pyrrolidin-2-yl)méthyl]-1H-pyrazol-4-yl-7H-pyrrolo[2,3-d]pyrimidine ; 4-(1-[1-(méthylsulfonyl)pyrrolidin-2-yl]méthyl-1H-pyrazol-4-yl)-7H-pyrrolo[2,3-d]pyrimidine ; 3-cyclopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]acrylonitrile ; 3-(2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]éthyl)-cyclopentane-carbonitrile ; 3- [3-(hydroxyméthyl)cyclopentyl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]-propanenitrile ; 4- (1-benzyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine ; 4-[1 -(2-naphtylméthyl)-1 H-pyrazol-4-yl]-1 H-pyrrolo[2,3-b]pyridine ; 4-(1-phényl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine ; 3- [4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]benzonitrile ; 4- méthyl-3-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]benzonitrile ; 4-[1-(3,5-diméthoxybenzyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine ; 4-[1 -(1 -phényléthyl)-1 H-pyrazol-4-yl]-1 H-pyrrolo[2,3-b]pyridine ; 4-[1 -(cyclohexylméthyl)-l H-pyrazol-4-yl]-1 H-pyirolo[2,3-b]pyridine ; 3- {[4-(1 H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]méthyl}benzonitrile ; 2- {[4-(1 H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]méthyl}benzonitrile ; 4- {[4-(1 H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]méthyl}benzonitrile ; 1-phényl-2-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]éthanone ; 4-{1-[(5-méthylisoxazol-3-yl)méthyl]-1H-pyrazol-4-yl}-1H-pyrrolo[2,3-b]pyridine ; 4-[1-(tétrahydro-2H-pyrann-2-ylméthyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine ; 4-(1-cyclohex-2-én-1-yl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine ; 4-(1-cyclohexyl-1 H-pyrazol-4-yl)-1 H-pyrrolo[2,3-b]pyridine ; 4’-{[4-(1 H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]méthyl}biphényl-2-carbonitrile ; 4-[1-(2-nitrobenzyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine ; 4-{1-[2,6-dichloro-4-(trifluorométhyl)phényl]-1H-pyrazol-4-yl}-1H-pyrrolo[2,3-b]pyridine ; 4-[1-(3-nitrobenzyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine ; 4-[1-(2-bromobenzyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine ; N-phényl-2-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]propanamide ; 4-{1-[3-(trifluorométhoxy)benzyl]-1 H-pyrazol-4-yl}-1 H-pyrrolo[2,3-b]pyridine ; 4-{1-[2-fluoro-5-(trifluorométhyl)benzyl]-1H-pyrazol-4-yl}-1H-pyirolo[2,3-b]pyridine ; 4-{1-[3-(trifluorométhyl)benzyl]-1H-pyrazol-4-yl}-1H-pyrrolo[2,3-b]pyridine ; 4-[1-(pyridin-3-ylméthyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine ; 4-{1-[1-phénylbutyl]-1H-pyrazol-4-yl}-1H-pyrrolo[2,3-b]pyridine ; 1- phényl-2-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]propan-1-one ; 4-[1-(2,6-dichlorobenzyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine ; 4-[1-(2,6-diméthylphényl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine ; 2- [4-(1 H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]-5-(trifluorométhyl)-benzonitrile ; 4-[1-(4-bromo-3,5,6-trifluoropyridin-2-yl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine ; 4-[1-(cyclopropylméthyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine ; 4-[1-(2,5-diméthylphényl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine ; 4-[1-(2-méthylphényl)-1 H-pyrazol-4-yl]-1 H-pyrrolo[2,3-b]pyridine ; 4-[1-(2-méthoxyphényl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine ; 3- {-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]éthyl}benzonitrile ; 3- chloro-4-[4-(1 H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]benzonitrile ; 4- [1 -(1 -cyclohexyléthyl)-1 H-pyrazol-4-yl]-1 H-pyrrolo[2,3-b]pyridine ; 4-fluoro-2-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]benzonitrile ; 2- fluoro-4-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]benzonitrile ; 3- fluoro-4-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]benzonitrile ; 4- (1-{1-[3-(trifluorométhyl)phényl]éthyl}-1H-pyrazol-4-yl)-1H-pyirolo[2,3-b]pyridine ; 4-[1-(3,5-diméthylphényl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine ; 4-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]benzonitrile ; {4-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]phényl}acétonitrile ; 4-[1-(1-phénylpropyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine ; 4-(1-{1-[4-(méthylsulfonyl)phényl]éthyl}-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine ; 4-{1-[1-(3-fluoro-4-méthoxyphényl)éthyl]-1H-pyrazol-4-yl}-1H-pyrrolo[2,3-b]pyridine ; 4-(1-{1-[2-(trifluorométhyl)phényl]éthyl}-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine ; 4-(1-{1-[3,5-bis(trifluorométhyl)phényl]éthyl}-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine ; 4-{1-[4-(1 H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]éthyl}benzonitrile ; 4-{1-[4-nitro-2-(trifluorométhyl)phényl]-1 H-pyrazol-4-yl}-1 H-pyrrolo[2,3-b]pyridine ; 3- méthyl-4-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]benzonitrile ; 4- [1-(2-chlorophényl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine ; 3- bromo-4-[4-(1 H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]benzonitrile ; 4- [4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]benzoate d’éthyle ; 4-{1-[2-chloro-6-nitro-4-(trifluorométhyl)phényl]-1H-pyrazol-4-yl}-1H-pyrrolo[2,3-b]pyridine ; 4-(1-{1-[4-(trifluorométhyl)phényl]éthyl}-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine ; 4-[1-(2,3-dihydro-1H-indén-1-yl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine ; 4-[1-(1,2,3,4-tétrahydronaphtalén-1-yl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine ; 4-(1-{1-[2-chloro-5-(trifluororméthyl)phényl]éthyl}-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine ; 4-{1-[1-(2,4-dichloro-5-fluorophényl)éthyl]-1H-pyrazol-4-yl}-1H-pyirolo[2,3-b]pyridine ; 4-[1-(1-cyclopentyléthyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine ; 4-[1-(1-méthyl-3-phénylpropyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine ; 4-[1-(1-cyclobutyléthyl)-1 H-pyrazol-4-yl]-1 H-pyirolo[2,3-b]pyridine ; [2-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]-5-(trifluorométhyl)phényl]-acétonitrile ; [5-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]-2-(trifluorométhyl)phényl]-acétonitrile ; 4- {1-[4-phénylbut-3-én-1-yl]-1H-pyrazol-4-yl}-1H-pyiTolo[2,3-b]pyridine ; [3-chloro-2-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]-5-(trifluorométhyl)phényl]-acétonitrile ; 5- [4-(1 H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]-2-(trifluorométhyl)-benzonitrile ; 4-{1-[2-chloro-4-(trifluorométhyl)phényl]-1H-pyrazol-4-yl}-1H-pyrrolo[2,3-b]pyridine ; 4-[4-(1 H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]-2-(trifluorométhyl)-benzonitrile ; 2- [4-(1 H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]benzonitrile ; 3- chloro-2-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]benzonitrile ; 4- amino-5,6-difluoro-2-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]isophtalonitrile ; 1- {[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]méthyl}-cyclopropanecarbonitrile; 5- bromo-2-[4-(1 H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]benzonitrile ; 3-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]-4-(trifluorométhyl)-benzonitrile ; 2- [4-(1 H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1 -yl]-3-(trifluorométhyl)-benzonitrile ; 3- [4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]-4-(trifluorométhyl)benzamide ; 3- [4-(1H-pyrrolo[2,3-b]pyndin-4-yl)-1 H-pyrazol-1-yljcyclohexanone ; 2-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yljcyclohexanol ; 4- (1-{[1-(méthylsulfonyl)pipéridin-4-yl]méthyl}-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine ; 2-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]cyclohexanecarbonitrile ; 4-{1-[2-(trifluorométhyl)phényl]-1H-pyrazol-4-yl}-1H-pyrrolo[2,3-b]pyridine ; 4-[1-(2,6-dichlorophényl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine ; (4-{[4-(1 H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]méthyl}cyclohexyl)méthanol ; 4-[1-(tétrahydrofurann-2-ylméthyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine ; 4-[1-(1-cyclopentylpropyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine ; 4-[1-(tétrahydrofurann-3-ylméthyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine ; 2- chloro-3-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]benzonitrile ; 3- [4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1 -yl]-3-(1,3-thiazol-5-yl)propanenitrile ; 3-(1-méthyl-1 H-imidazol-5-yl)-3-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]propane-nitrile ; 3- [4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]-3-(3-thiényl)propanenitrile ; {1-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1H-pyrazol-1-yl]cyclopentyl}acétonitrile ; 4- chloro-3-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1 -yl]benzonitrile ; 4-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1 -yl]phtalonitrile ; 3- méthyl-4-[4-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]benzaldéhyde ; 4- [1-(2-méthyl-4-nitrophényl)-1 H-pyrazol-4-yl]-1 H-pyrrolo[2,3-b]pyridine ; 3- [4-(1 H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]cyclopentanone ; 4- [1-(3-furylméthyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine ; 4-[1-(2-furylméthyl)-1H-pyrazol-4-yl]-1H-pyrrolo[2,3-b]pyridine ; 3-{2-cyano-1-[4-(1 H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]éthyl}benzonitrile ; {3-méthyl-4-[4-(1 H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]phényl}méthanol ; 3-(1-benzofurann-2-yl)-3-[4-(1 H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1 -yl]propanenitrile ; 3-(3-furyl)-3-[4-(1 H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1-yl]propanenitrile ; {3-méthyl-4-[4-(1 H-pyrrolo[2,3-b]pyridin-4-yl)-1 H-pyrazol-1 -yl]phényl}acétonitrile ; 4-méthyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]benzonitrile ; 4-[1-(1-cyclopentylpropyl)-1H-pyrazol-4-yl]-7H-pyrrolo[2,3-d]pyrimidine ; {1-[4-(7H-pyirolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1 -yl]cyclopentyl}acétonitrile ; 3-{2-cyano-1-[4-(7H-pyirolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]éthyl}benzonitrile ; 3- [4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]-3-(3-thiényl)propanenitrile ; 4- chloro-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1 -yl]benzonitrile ; 3-(3-furyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propanenitrile ; 3-{1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclopentyl}-propanenitrile ; {1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]cyclohexyl}acétonitrile ; {3-méthyl-4-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]phényl}méthanol ; 3-pyridin-4-yl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile ; 3-pyridin-3-yl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile ; 3-[4-(méthylthio)phényl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propanenitrile ; 3-(3-méthoxyphényl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propanenitrile ; 3-(4-méthoxyphényl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propanenitrile ; {3-méthyl-4-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]phényl}acétonitrile ; 3-[4-(méthylsulfinyl)phényl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propanenitrile ; 3-[4-(méthylsulfonyl)phényl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propanenitrile ; 3-[3-(cyanométhoxy)phényl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propanenitrile ; 3-(6-chloropyridin-3-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile ; 5- {2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]éthyl}pyridine-2-carbonitrile ; 3-(3,5-diméthylisoxazol-4-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]-propanenitrile ; 3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1 -yl]-3-[6-(trifluorométhyl)pyridin-3-yl]-propanenitrile ; 3-(6-méthoxypyridin-3-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propane-nitrile ; 3-pyridin-2-yl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1 -yl]propanenitrile ; 3-(6-bromopyridin-2-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile ; 6- {2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]éthyl}pyridine-2-carbonitrile ; 3-(5-bromopyridin-3-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propanenitrile ; 5-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]éthyl}nicotinonitrile ; 3-(2-méthoxypyridin-3-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propane-nitrile ; 3-[4-(cyanométhoxy)phényl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propane-nitrile ; 3-[2-(cyanométhoxy)phényl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propanenitrile ; 3-(3,5-dibromophényl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile ; 5-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]éthyl}isophtalonitrile ; 3-[6-(diméthylamino)pyridin-2-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propanenitrile ; 3-(4-bromo-2-thiényl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile ; 5-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]éthyl}thiophène-3-carbonitrile ; 3-(5-bromo-2-fluorophényl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]-propanenitrile ; 3-(3-nitrophényl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propanenitrile ; 3-(5-bromo-2-méthoxyphényl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]-propanenitrile ; 3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]éthyl}-4-méthoxybenzonitrile ; 3-(3-bromophényl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile ; 3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]éthyl}-4-fluorobenzonitrile ; 3-[5-bromo-2-(cyanométhoxy)phényl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propanenitrile ; 3- (4-bromo-2-furyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propanenitrile ; 4- (cyanométhoxy)-3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]éthyl}-benzonitrile ; 3-(4-bromopyridin-2-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1 -yl]propanenitrile ; 2- {2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]éthyl}isonicotinonitrile ; 5- {2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1 -yI]éthyI}-3-furonitriIe ; 3- [2-bromo-5-(cyanométhoxy)phényl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]-propanenitrile ; 4- (cyanométhoxy)-2-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]-éthyl}benzonitrile ; 3-pyrimidin-5-yl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile ; 3- (2-bromopyridin-4-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1 -yl]propanenitrile ; 4- {2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]éthyl}pyridine-2-carbo-nitrile ; 3-(5-méthoxypyridin-3-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propane-nitrile ; 3-(3-chlorophényl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile ; 3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-3-[3-(trifluorométhyl)phényl]-propanenitrile ; 3-(3-phénoxyphényl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1 -yl]propanenitrile ; 3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-3-[3-(trifluorométhoxy)-phényl]-propanenitrile ; 3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]éthyl}benzoate de méthyle ; acide 3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]éthyl}benzoïque ; 3-[3-(1H-pyrazol-4-yl)phényl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]-propanenitrile ; 3- (3-aminophényl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile ; N-(3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1 -yl]éthyl}phényl)acétamide ; N-(3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]éthyl}phényl)-méthanesulfonamide ; 4- {2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]éthyl}thiophène-2-carbonitrile ; 5- {2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]éthyl}thiophène-2-carbonitrile ; 3-[3-(morpholin-4-ylcarbonyl)phényl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propanenitrile ; N-(2-aminoéthyl)-3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]éthyl}-benzamide ; 3-(5-formyl-3-thiényl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile ; 3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]éthyl}-N-méthylbenzamide ; 2- cyano-N-(3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]éthyl}phényl)-acétamide ; N-(3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]éthyl}phényl)-nicotinamide ; N-(3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]éthyl}phényl)-N’-isopropylurée ; (3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]éthyl}phényl)-carbamate d’isopropyle ; 3- (5-phénylpyridin-3-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile ; 3-(3,3’-bipyridin-5-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile ; 3-(5-pyrimidin-5-ylpyridin-3-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propanenitrile ; 3-[5-(1-méthyl-1H-pyrazol-4-yl)pyridin-3-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propanenitrile ; 3-(5-éthynylpyridin-3-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propane-nitrile ; 3-[5-(phénylthio)pyridin-3-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propanenitrile ; 3-(2-bromo-1,3-thiazol-5-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]-propanenitrile ; 3-(5-morpholin-4-ylpyridin-3-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propanenitrile ; 3-(1-méthyl-1H-pyrazol-4-yl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propanenitrile ; 3-(3-pyridin-3-ylphényl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile ; 3-[5-(phénylsulfinyl)pyridin-3-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propanenitrile ; 3-[5-(phénylsulfonyl)pyridin-3-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propanenitrile ; 3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-3-[5-(1,3-thiazol-2-ylthio)pyridin-3-yl]propanenitrile ; 3- [5-(éthylthio)pyridin-3-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propanenitrile ; 4- {1-[1-méthyl-2-(1H-1,2,4-triazol-1 -yl)éthyl]-1 H-pyrazol-4-yl}-7H-pyrrolo[2,3-d]pyrimidine ; 1-phényl-2-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1 -yl]propan-1 -one ; 3-[5-(éthylsulfinyl)pyridin-3-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1 -yl]-propanenitrile ; 3-[5-(éthylsulfonyl)pyridin-3-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propanenitrile ; 3-[5-(cyclohexylthio)pyridin-3-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propanenitrile ; 1- phényl-2-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propan-1-ol ; 3-[3-(éthylthio)phényl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile ; 3-[3-(éthylsulfinyl)phényl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propanenitrile ; 3-[3-(éthylsulfonyl)phényl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propanenitrile ; 3-[5-(cyclohexylsulfonyl)pyridin-3-yl]-3-[4-(7H-pyrrolo[2,3-d]-pyrimidin-4-yl)-1 H-pyrazol-1 -yl]propanenitrile ; 3- [5-(cyclohexylsulfinyl)pyridin-3-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile ; 4- [1-(1-méthyl-2-phényléthyl)-1H-pyrazol-4-yl]-7H-pyrrolo[2,3-d]pyrimidine ; 4-{1-[1-méthyl-2-(3-thiényl)éthyl]-1H-pyrazol-4-yl}-7H-pyrrolo[2,3-d]pyrimidine ; 3- {1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1 -yl]éthyl}benzonitrile ; 4- {1-[2-(1H-imidazol-1-yl)-1-méthyléthyl]-1H-pyrazol-4-yl}-7H-pyrrolo[2J3-d]pyrimidine ; 4-{1-[1-méthyl-2-(3-méthyl-1,2,4-oxadiazol-5-yl)éthyl]-1H-pyrazol-4-yl}-7H-pyrrolo[2,3-d]-pyrimidine 3-[3-(méthylsulfonyl)phényl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propanenitrile ; 3-(3-pyridin-4-ylphényl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile ; 3-[5-(isopropylthio)pyridin-3-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1 -yl]-propanenitrile ; 3-[5-(isopropylsulfinyl)pyridin-3-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1 -yl]propanenitrile ; 3-[5-(isopropylsulfonyl)pyridin-3-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1 -yl]propanenitrile ; 3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-3-[5-(trifluorométhyl)pyridin-3-yl]-propanenitrile ; 2- [4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1 -yl]-N-[3-(trifluorométhyl)phényl]-propanamide ; N-2-naphtyl-2-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propanamide ; N-1-naphtyl-2-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propanamide ; N-(3-cyanophényl)-2-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propanamide ; N-phényl-2-[4-(7H-pyrrolo[2,3-d]-pyrimidin-4-yl)-1H-pyrazol-1-yl]-butanamide ; N-(4-phénoxyphényl)-2-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]butanamide ; N-2-naphtyl-2-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]butanamide ; N-(3-cyanophényl)-2-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]butanamide ; N-biphényl-4-yl-2-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]butanamide ; N-(biphényl-4-ylméthyl)-2-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]butanamide ; N-(biphényl-3-ylméthyl)-2-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]butanamide ; N-(4-cyanophényl)-2-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]butanamide ; N-1-naphtyl-2-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]butanamide ; 5-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]éthyl}-N-phénylnicotin-amide ; N-(3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]éthyl}phényl)- 3-(trifluorométhyl)benzamide ; N-(3-{[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]méthyl}phényl)-3-(trifluoro-méthyl)benzamide ; 3-[3-(méthylsulfonyl)phényl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]-propanenitrile ; N-(3-{[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]méthyl}phényl)benzène-sulfonamide ; 3-{[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]méthyl}-N-[3-(trifluorométhyl)-phényl]benzamide ; 3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]éthyl}-N,N-diméthyl-benzènesulfonamide ; N-benzyl-3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]éthyl}benzène-sulfonamide ; N-benzyl-3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]éthyl}benzamide ; 3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]éthyl}-N-phénylbenzamide ; 3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]éthyl}-N-[3-(trifluoro-méthyl)phényl]benzamide ; N-(3-cyanophényl)-3-{[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]méthyl}-benzamide ; N-benzyl-3-{[4-(7H-pyrrolo-[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]méthyl}benzamide ; N-1-naphtyl-3-{[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]méthyl}benzamide ; N-2-naphtyl-3-{[4-(7H-pyrrolo[2,3-d]-pyrimidin-4-yl)-1 H-pyrazol-1-yl]méthyl}benzamide ; N-(3-{[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]méthyl}phényl)-2-naphtamide ; N-(3-{[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]méthyl}phényl)-1-naphtamide ; 2- phényl-N-(3-{[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]méthyl}phényl)-acétamide ; 3- chloro-N-(3-{[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]méthyl}phényl)-benzamide ; N-(3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]éthyl}phényl)-2-naphtamide ; N-(3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]éthyl}phényl)-1-naphtamide ; N-(3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]-éthyl}phényl)-2-phénylacétamide ; 3- cyano-N-(3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]éthyl}phényl)-benzamide ; N-(3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]éthyl}phényl)-benzamide ; N-(3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]éthyl}phényl)- 4- (trifluorométhyl)benzamide ; N-(3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]éthyl}phényl)-N’-phénylurée ; 3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]éthyl}-N-[4-(trifluorométhyl)phényl]benzamide ; 3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]éthyl}-N-(4-méthyl-phényl)benzamide ; N-(4-cyanophényl)-3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-éthyl}benzamide ; 3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]éthyl}-N-2-naphtyl-benzamide ; 3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]éthyl}-N-1-naphtyl-benzamide ; 3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]éthyl}-N,N-diméthyl-benzamide ; 3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]éthyl}-N-pyridin-3-yl-benzamide ; 3-{2-cyano-1l-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]éthyl}-N-méthyl-N-phényl-benzamide ; 3-12-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]éthyl}-N-cyclohexyl-benzamide N-(3-cyanophényl)-3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]éthyl}-benzamide ; N-biphényl-4-yl-3-12-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]-éthyl}benzamide N-(4-chlorophényl)-3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]-éthyl}benzamide ; 3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]éthyl}-N-(3,4-diméthyl-phényl)benzamide ; 3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]éthyl}-N-isoxazol-3-yl-benzamide ; 3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]éthyl}-N-méthyl-phénylbenzènesulfonamide ; 3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]éthyl}-N-propylbenzène-sulfonamide ; 3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]éthyl}-N-phényl-benzènesulfonamide ; 3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]éthyl}-N-2-naphtyl-benzènesulfonamide ; 3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]éthyl}-N-cyclopropyl-benzènesulfonamide ; 3-[3-(pipéridin-1-ylsulfonyl)phényl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propanenitrile ; 3-[3-(morpholin-4-ylsulfonyl)phényl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propanenitrile ; 3-{2-cyano-1-[4-(7H-pyrrolo[2,3-dlpyrimidin-4-yl)-1H-pyrazol-1-yl]éthyl}-N-(4-méthylphényl)-benzènesulfonamide ; 3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]éthyl}-N-(3,4-diméthylphényl)benzènesulfonamide ; 3-[3-(benzylsulfonyl)phényl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propanenitrile ; 3- [3-(benzylthio)phényl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile ; 4- {[(3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]éthyl}phényl)-sulfonyl]méthyl}benzonitrile ; 3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]éthyl}-N-méthylbenzène-sulfonamide ; 3-{2-cyano-1-[4-(7H-pyirolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]éthyl}-N-1-naphtyl-benzènesulfonamide ; N-biphényl-4-yl-3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]éthyl}-benzènesulfonamide ; 3-[3-(benzyloxy)phényl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile ; 3-{2-cyano-1-[4-(7H-pyirolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]éthyl}-N-cyclohexyl-benzènesulfonamide 3-[3-(3,4-dihydroisoquinoléin-2(1H)-ylsulfonyl)phényl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile ; 3-{2-cyano-1-[4-(7H-pyirolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]éthyl}-N,N-diéthyl-benzènesulfonamide ; 3-{3-[(4-éthylpipérazin-1-yl)sulfonyl]phényl}-3-[4-(7H-pyrrolo[2,3-d]-pyrimidin-4-yl)-1 H-pyrazol-1-yl]propanenitrile ; N-1,3-benzodioxol-5-yl-3-12-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-éthyl}benzènesulfonamide ; 3-{3-[(2,6-diméthylmorpholin-4-yl)sulfonyl]phényl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile ; 3-{3-[(4-oxopipéridin-1-yl)sulfonyl]phényl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]propanenitrile ; 3-[3-(isopropylsulfonyl)phényl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propanenitrile ; 3-{3-[(cyclohexylméthyl)sulfonyl]phényl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile ; 3-[3-(octahydroisoquinoléin-2(1H)-ylsulfonyl)phényl]-3-[4-(7H-pyirolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile ; 3-{3-[(2-phényléthyl)-sulfonyl]phényl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile ; cis-4-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohexylacétonitrile ; 3-cis-4-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohexylpropanenitrile cis-4-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohexanecarbonitrile ; 3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]cyclohexylacétonitrile ; 5- ({cis-4-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]cyclohexyl}thio)-1 H-1,2,4-triazol-3-amine ; [cis-4-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1 H-pyrazol-1-yl]-1-(1 H-1,2,4-triazol-1-yl)cyclohexyl]-acétonitrile ; 3-[3-(pipérazin-1-ylsulfonyl)phényl]-3-[4-(7H-pyirolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile ; 3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-3-[3-(thiomorpholin-4-ylsulfonyl)-phényl]propanenitrile ; 3-{3-[(4-hydroxypipéridin-1-yl)sulfonyl]phényl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile ; 3-[3-(isobutylsulfonyl)phényl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-propanenitrile 3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-3-{3-[(tétrahydro-2H-pyran-4-ylméthyl)sulfonyl]-phényl}propanenitrile ; 3-{3-[(3-furylméthyl)sulfonyl]phényl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile ; 3-{3-[(1,1-dioxidothiomorpholin-4-yl)sulfonyl]phényl}-3-[4-(7H-pyiTolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile ; 3-{3-[(pyridin-4-ylméthyl)sulfonyl]phényl}-3-[4-(7H-pyirolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile ; 3-{3-hydroxy-1-[4-(7H-pyirolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propyl}-N,N-ditnéthyl-benzènesulfonamide ; 3- {1-[4-(7H-pyiTolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]but-3-én-1-yl}benzonitrile ; 4- {1-[1-(3-bromophényl)but-3-én-1-yl]-IH-pyrazol-4-yl}-7H-pyiTolo[2,3-d]pyrimidine ; 3- {4,4-difluoro-1-[4-(7H-pyirolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]but-3-én-1-yl}-benzonitrile ; 4- (1-{4,4-difluoro-1-[3-(morpholin-4-ylsulfonyl)-phényl]but-3-én-1-yl}-1H-pyrazol-4-yl)-7H-pyrrolo[2,3- d]pyrimidine ; 4-(1-{1-[3-(éthylsulfonyl)phényl]-4,4-difluorobut-3-én-1-yl}-1H-pyrazol-4-yl)-7H-pyrrolo[2,3-d]pyrimidine ; 4-(1-{1-[3-(benzyloxy)phényl]-4,4-difluorobut-3-én-1-yl}-1H-pyrazol-4-yl)-7H-pyrrolo[2,3-d]-pyrimidine ; 4-(1-{4,4-difluoro-1-[3-(méthylsulfonyl)phényl]but-3-én-1-yl}-1H-pyrazol-4-yl)-7H-pyrrolo[2,3-d]pyrimidine ; 3-{[4-(7H-pyirolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]méthyl}benzonitrile ; 3- {1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]-butyl}benzonitrile ; 4- (1-{1-[3-(éthylsulfonyl)phényl]-4,4-difluorobutyl}-1H-pyrazol-4-yl)-7H-pyrrolo[2,3-d]-pyrimidine ; 4-[1-(1-cyclopentylbut-3-én-1-yl)-1H-pyrazol-4-yl]-7H-pyirolo[2,3-d]pyrimidine ; 4-[1-(1-cyclopentylbutyl)-1H-pyrazol-4-yl]-7H-pyrrolo[2,3-d]pyrimidine ; 4-[1-(1-cyclopentyl-4,4-difluorobut-3-én-1-yl)-1H-pyrazol-4-yl]-7H-pyrrolo[2,3-d]pyrimidine ; 4-{1-[4,4-difluoro-1-(tétrahydrofurann-3-yl)but-3-én-1-yl]-1H-pyrazol-4-yl}-7H-pynOlo[2,3-d]pyrimidine ; 4-[1-(1-cyclopropyl-4,4-difluorobut-3-én-1-yl)-1H-pyrazol-4-yl]-7H-pyrrolo[2,3-d]pyrimidine ; 4-[1-(1-cyclopentyl-4,4-difluorobutyl)-1H-pyrazol-4-yl]-7H-pyrrolo[2,3-d]pyrimidine ; 3-(1-méthylcyclopentyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile ; 1-{2-cyano-1-[4-(7H-pyirolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]éthyl}cyclopropane-carbonitrile ; 3- [1-(méthylsulfonyl)pyrrolidin-3-yl]-3-[4-(7H-pyirolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile ; 4- {1-[2,2,2-trifluoro-1-(1H-imidazol-2-ylméthyl)éthyl]-1H-pyrazol-4-yl}-7H-pyiTolo[2,3-d]pyrimidine ; 4-(1-(1R)-2,2,2-trifluoro-1-[(4-méthyl-1,3-thiazol-2-yl)méthyl]éthyl-1H-pyrazol-4-yl)-7H-pyrrolo[2,3-d]pyrimidine ; 4- {1-[1-(5-bromopyridin-3-yl)-4,4-difluorobut-3-én-1-yl]-1H-pyrazol-4-yl}-7H-pyrrolo[2,3-d]pyrimidine ; 5- {4,4-difluoro-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]but-3-én-1-yl}nicotinonitrile ; 3-[3-(pyirolidin-1-ylsulfonyl)-phényl]-3-[4-(7H-pyrrolo-[2,3-d]-pyrimidin-4-yl)-1H-pyrazol-1-yl]-propanenitrile ; N-benzyl-3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]éthyl}-N-méthylbenzènesulfonamide ; 3-{[(3-{2-cyano-1-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]éthyl}-phényl)sulfonyl]méthyl}-benzonitrile ; 3-{3-[(2-naphtylméthyl)-sulfonyl]-phényl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile ; 3-{3-[(1-phényléthyl)sulfonyl]-phényl}-3-[4-(7H-pyrrolo[2,3-d]-pyrimidin-4-yl)-1H-pyrazol-1-yl]-propanenitrile ; 3-{2-cyano-1-[4-(7H-pyrrolo-[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]éthyl}-N-(2-morpholin-4-yléthyl)-benzènesulfonamide ; 3-{2-cyano-1-[4-(7H-pyrrolo-[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]éthyl}-N-[(1S)-1-phényléthyl]benzènesulfonamide ; 3-{2-cyano-1-[4-(7H-pyirolo-[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]éthyl}-N-phényl-benzamide ; 3-{2-cyano-1-[4-(7H-pyirolo-[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]éthyl}-N-(tétrahydrofurann-2-yl-méthyl)benzènesulfonamide ; trifluoroacétate de 3-{3-[(cyclopropylméthyl)sulfonyl]phényl}-3-[4-(7H-pyrrolo-[2,3-d]pyrimidin-4-yl)-1H-pyra-zol-1-yl]propanenitrile ; 3-{3-[(4-méthylpipérazin-1-yl)-sulfonyl]phényl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile ; et 3-{3-[(1-oxidothiomorpholin-4-yl)sulfonyl]phényl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile ; ou l’un de ses sels de qualité pharmaceutique. 9. Composé qui est le 3-cyclopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile ; ou l’un de ses sels de qualité pharmaceutique ; pour utilisation dans une méthode de traitement d’une maladie choisie parmi la cachexie, un trouble myéloproliferatif et un cancer chez un patient en ayant besoin, en combinaison avec un agent pharmaceutique supplémentaire. 10. Composé qui est le (R)-3-cyclopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile ; ou l’un de ses sels de qualité pharmaceutique ; pour utilisation dans une méthode de traitement d’une maladie choisie parmi la cachexie, un trouble myéloproliferatif et un cancer chez un patient en ayant besoin, en combinaison avec un agent pharmaceutique supplémentaire. 11. Composé qui est le 3-cyclopropyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-pyrazol-1-yl]-propionitrile ; ou l’un de ses sels de qualité pharmaceutique ; pour utilisation dans une méthode de traitement d’une maladie choisie parmi la cachexie, un trouble myéloproliferatif et un cancer chez un patient en ayant besoin, en combinaison avec un agent pharmaceutique supplémentaire. 12. Composé ou sel pour utilisation selon l’une quelconque des revendications 1 à 11, où ledit agent pharmaceutique supplémentaire est un agent chimiothérapeutique. 13. Composé ou sel pour utilisation selon l’une quelconque des revendications 1 à 11, où ladite méthode comprend l’administration simultanée dudit composé ou sel et dudit agent pharmaceutique supplémentaire au patient. 14. Composé ou sel pour utilisation selon l’une quelconque des revendications 1 à 11, où ladite méthode comprend l’administration séquentielle dudit composé, ou dudit sel, et dudit agent pharmaceutique supplémentaire audit patient. 15. Composé ou sel pour utilisation selon l’une quelconque des revendications 1 à 11, où environ 5 à environ 1000 mg dudit composé ou dudit sel sont administrés audit patient. 16. Composé ou sel pour utilisation selon l’une quelconque des revendications 1 à 15, où la maladie est un trouble myéloprolifératif. 17. Composé ou sel pour utilisation selon la revendication 16, où ledit trouble myéloprolifératif est la polyglobulie essentielle (PE), la thrombocythémie essentielle (TE), la splénomégalie myéloïde (MMM), la leucémie myéloïde chronique (LMC), la leucémie myélomonocytaire chronique (LMMC), le syndrome hyperéosinophile (SHE) ou la mas-tocytose systémique (MCS). 18. Composé ou sel pour utilisation selon la revendication 16, où ledit trouble myéloprolifératif est la polyglobulie essentielle (PE). 19. Composé ou sel pour utilisation selon la revendication 16, où ledit trouble myéloprolifératif est la thrombocythémie essentielle (TE). 20. Composé ou sel pour utilisation selon la revendication 16, où ledit trouble myéloprolifératif est la splénomégalie myéloïde (MMM). 21. Composé ou sel pour utilisation selon la revendication 16, où ledit trouble myéloprolifératif est la leucémie myéloïde chronique (LMC). 22. Composé ou sel pour utilisation selon la revendication 16, où ledit trouble myéloprolifératif est la leucémie myélomonocytaire chronique (LMMC). 23. Composé ou sel pour utilisation selon l’une quelconque des revendications 1 à 15, où la maladie est un cancer. 24. Composé ou sel pour utilisation selon la revendication 23, où ledit cancer est un myélome multiple. 25. Composé ou sel pour utilisation selon la revendication 23, où ledit cancer est un cancer du pancréas. 26. Composé ou sel pour utilisation selon la revendication 23, où ledit cancer est une leucémie. 27. Composé ou sel pour utilisation selon la revendication 23, où ledit cancer est un lymphome. 28. Composé ou sel pour utilisation selon la revendication 23, où ledit cancer est un cancer du sein. 29. Composé ou sel pour utilisation selon la revendication 23, où ledit cancer est une leucémie myéloïde aiguë. 30. Composé ou sel pour utilisation selon la revendication 23, où ledit cancer est un cancer du poumon. 31. Composition pharmaceutique pour utilisation selon la revendication 23, où ledit cancer est un cancer de la tête et du cou. 32. Composition pharmaceutique pour utilisation selon la revendication 23, où ledit cancer est choisi parmi les suivants : cancer de la prostate, cancer du rein, cancer du foie, cancer de l’estomac, cancer de la thyroïde, glioblastome, sarcome de Kaposi, maladie de Castleman, mélanome, lymphome cutané à cellules T (LCCT), lymphome cutané à cellules B, syndrome de Sézary, mycosis fongoïde, leucémie lymphocytaire aiguë, et cancer colorectal. 33. Composé ou sel pour utilisation selon l’une quelconque revendications 1 à 15, où ladite cachexie résulte d’un cancer ou y est associée. 34. Composé ou sel pour utilisation selon l’une quelconque des revendications 1 à 11, où ledit agent pharmaceutique supplémentaire est le revlimid. 35. Composé ou sel pour utilisation selon l’une quelconque des revendications 1 à 11, où ledit procédé comprend l’administration d’un corticostéroïde audit patient. 36. Composé ou sel pour utilisation selon la revendication 35, où ledit corticostéroïde est la prednisone. 37. Composé ou sel pour utilisation selon l’une quelconque des revendications 1 à 11, où ledit agent pharmaceutique supplémentaire est un agent endommageant l’ADN. 38. Composé ou sel pour utilisation selon la revendication 37, où ledit agent endommageant l’ADN est choisi parmi le melphalan, la doxorubicine, le cyclophosphamide, la vincristine, l’étoposide et la carmustine. 39. Composé ou sel pour utilisation selon la revendication 24, où ledit agent pharmaceutique supplémentaire est choisi parmi le melphalan, le melphalan plus la prednisone, la doxorubicine, la dexaméthasone, et le bortézomib. 40. Composé ou sel pour utilisation selon l’une quelconque des revendications 1 à 11, où ledit agent pharmaceutique supplémentaire est un composé anti-inflammatoire. 41. Composé ou sel pour utilisation selon l’une quelconque des revendications 1 à 11, où ledit agent pharmaceutique supplémentaire est un inhibiteurde protéosome, le bortézomib, le thalidomide, un inhibiteurde Bcr-Abl, un inhibiteur de Flt-3, un inhibiteurde RAF, ou un inhibiteurde FAK.

Claims (4)

  1. .:. A
    képletö vegplet vagy gyögyászatlikg élPgípkáíé séj8£ klai:: T, ïi és V, egymástól föggeiienß^ é;Ie8sÄ8Ä' W van kiválasztva:: €k 2¾¾ OR? ös MiR-; afeot a szss-aient, nilTogenaîam, U, T és V áltól &amp;11Ä$ j$ls§à g^Éf X jelentése N vagy CR*; n értéke ö; vagy a értéke: ! és Y jelentése G}.g: alkiteesöport, Cj.* elkeni iéneseport, CCR:iR^}fC(öjiiG^i!R.!%5: (CR:iRi%C(0)OÍCR!iR!2}<. vagy í CR1 !R^OCiÖXCR5’&amp;% ahol az említet? € î .8 áld lásesoport vagy afeRéncsopaí adott esettet sadíssatoáíva van 1,2 vagy l halögéngtömstóG Ä csoporttal, CM esoportkd, amiooesoporítak 0M alkí:{"atnte--€söpte:al vagy G** ciaMl-amÓKv-csoperJtal; % jeleíRése miesoport. oifeloalfcilesepM, teeroadiesoport vagy tesroctkloatecsopep melyek misá-egyte adott esstiea saábsatitaaíva van 1,2,.3,4, 5 vagy ö szabszt&amp;uenssek mely szafessittoess{ek| egyöíástól iiiggeienii a követkeÄ feSsíd sanCnak): kiválasztva: WögénattmvCj* aífciesüporti: C2* álként lesöpört,Cv* alkmtlesttpört, CS4 Mog&amp;v-atktSi-osoport, Cj4 tddroxl-aMGesogiStj; €** stettektl-esöpot't, Cy’, CN, MOj, OR\ SRI C(0}Rb, CpSjMR^R* C(Ö}ÖR:\ ÖCÍÖp.1', OQGjNR'R*, NR"Rd, NRcC(0)Rs>, MRÍCföjMR^, NRcC(0)ÖR3, SÍÖ)R\ S(ö}NRcRá, SppR*, MR* S(í%ifm SPjjMSRK* Cy! a MvetteÄ közül w egymástól íllggotíésöl kiválasztva: aritesoport, heteroarilesopört. dkloaiktleseport és htetetktelkitcsopori:, melyek mindegyike adott esettet szatedtelva vart 1,2, 3, 4 vagy f szithsteoenssel, mely :skahsztáaeásiek> egymástól figgstléöüí a következők közűi väsptRd kiválasztva: lta~ íogéoatom, C;aíkilcsopori, C*.., aíkenilcsoport, €3.4 alkiniíesoport, C|.4 teogéo-aikil-esppínt, CM, M02, OR*, $R4\ C(0}Rk\ CiOprV", €(ö)ORs X 0C{0}Rk\ OCíOMR^rC NRC* ÜA'\ NR* €(0¾^ NR^CÍOJOR*' S{Ö)R1,, 8(0)ΝΚ*Ε,Γ, S(0)jRh és S{%MRc"Rtf ; R* jelentése M; R5 jefeíése H, halogénatom, C(j! afkt lesöpört, CM alteslesopon, Qm alkimicsoport, $%á Mogén-alkik csoport, CM, MOj, ÖR\ SR7, C{OjR*. CiO}NR<fRi0, C<OK>Rr,öaö}R*, OCíOtMR*R5í>, Itóé6, MlYXOiR*, MR?QÔ)OR; WÏ0, S<0>NRV0, S(0);.R4, NrS(0)jR8 vagy S(0)PRItg;: R*Jefetéss H, slkicsoport, C74 alkentksppoti:, '€%$ alkmilossport, €y; Mögéü-aikil-esopork OR', C(0}Rs, CiepÄ^ C{0)0R?. S{0)R*. :Sp>PR!íRíaa SjCps Väf SP$Ä®5 R? jetetése H, c.^ alkilcsoport, GU kalpgfedktNsöpört, C34 alkenjiesrtpork €34 alRykisopotk anlesogert, eikioalkilcsopotí, tteteroanlcsopors,. hsamæik:loa&amp;ifcso|>iÎ5^: arii-aikil-esopod:,: íssítercíariialk^I-csep^l,. gMoa&amp;H-alkH-csopesit'ii^'' Imew^fesMMkS^csí^órí; ^jelentése H. C;.(, alkilesopott, Cj.í( fesS:ógfe%a-a!kí!-<sopo«s. Ç$* '63* -aiMlfes^pœt*.. articsopon. etidualkllcsoporti bstetwi lesöpört, beteroeikloalkiksoport, ról-aíkí^&amp;Épíí^. ttetsroaril^lfel-esoport, cíkkíaikil-aikil'-csopon: vagy heitTOctxbalkil-aíkil-csoport; R!> ás R1®, egymástól i|§geilenöi, a kővetkezők közöl vannak kiválasztva:: H, Cnts alkilcsöpörk,¾¾ baltt-gésmlklbesopott, C2.* alkerölesoporl, slktttiicsopon, aMl-karbosPl-esoporí, mi-kasőonil-esoporC, G*.* alÜ-sail&amp;nil-esöpori, aêteuifosi-csopea^ srüesoport, beteroan lesöpört, clkloalkilcsopfM, heiprocikteaikitt csoport, anl-aikií-cssípeíftv betezoaril-slkilHssoport, cikloalkilmlkit-csoposà és ksîemcikfealkil^&amp;iklkfiîsspôil:;: vagy R8 és R;fö a nítrogésaloittmal egyik!, amelyte kspesolődaak, 4>»: 5-, 6» vagy ï-iagà östemsíkioalkitesopott alkot; Ru és Ru, egymástól: függetlenül, a következők közöl van kiválasztva: II, balogénak:«»,: 0¾.Cm alfcilesopork Cm hukigéo-alkJÍ-esoport. Cm alkemfcsoport.
  2. Cm atkinilcsoport, í.\_. hiífcóxi-stkikesöpöfí, Cm elano-alkil-esopon, anlesupon, heteroariiesopost. cisloaíkücsoporí es hcterocikloatkilcsopött:;: R* és R*, egymástól függetleníti, a következők közül va» kiválasztva: H, Cm alkilesppprí, Cm halőgén-alkil-cäijport, Cj..í; alkensksoport, €*<. aíkinilcsoport, árucsoport dkloaikiksoport heteröarifössparí, beteto-Ctkioalkifcsoport, anl-aikű-csoperi, Iteteroarskalkibcsoport, eiklöÄi^aMMpöpM: és csoport, aha: az említeti C« alkiesoport, Cm hafögén-alkfl-csoporl, €>.<> aikeoiksoport, Cj* álkiniteoport, atiesopOft, ctkloaíkílesoport, heteroanlesopott, kÂpaîMMkcso- fórt*. plkloalklkalkiksopoít vagy heteroctkloaöN®iN^»SÍ;Äfű 1* % «agy 3 sipttsztítöéáSséi, «ïy s^ÉSátteSHs(ek} egytplstöf filggetienöl a következók Ägl variénak) kiválasztva! M, CÍM, amíoöosöpörk Miog&amp;*á:íoj% CK aikltesőpött, €W: halogé»-alkílxtsspp!í,: ánksopork animiktkesopott, heleroatiicsopott:, letctajarílmikil-ekoijort, eifeleÄilesopsst és ket^ociklaälÄstspt»! Rs és: Ri5k ogytoasíól íüggeieail :a következek közöl vm. kiválasztva! M, (¾ alkilcsoport, Cm kilogéé-alkil-csepoti, G**: álkesztfcsopört, €3.* slkínfeap^ anksoporî, dkl^sÉfc^i^'iüetetwâpSQpitst» beîeroeîic tealkiksopott, akíbsiklksopött, IkleKtadkalkükese^öst, eiklöalk!l--aMkesöpmt: és hekroctkloalkikaikil^so-pori, attól az erttlltetí G(.«; áíRtlesoport, Cj.s ösfegétt-aikil-csoporp €2?«. áfóttfeöpötf, áru csoport, eikioáMfesoport, ttetoroariicsopott, heieroelfcloalki lesöpört, ari~siikil-csope?% heieroarälmikil-csopötl, cik!oslkii~aifeÍÍ-osö|ssÉ vagy tóerodkloalktl-alki-csoporí adott esetije» szabsatitusiva van 1, 2 vagy 3 mb* sztttoenssel, mely sztibsztittísosfek) egymástól függetleníti: &amp; kővetkezők köböl Vánpak) kiválásává: Oki, CN, atttinöcsopptt, lálögénálÖtn, Gtz alkiiesopor·, C..„ bslógÉi^ilkli^Söpert, Om bálögép-alktGcsoptMt, artiesoporí, arilmikiimsppott, ittteroanicsópött, betsroartl-alkil-es^jpík cíkloáikilcsopott és tassőeikioalktiesígsork R‘ és R\ egyrtjástól ítlggeilenöi, a kövelltezők Äö| vaó ktvalaszíva: H. alklksoport, balogén·· alki-csopop,: Cmí á&amp;ettilésópótt, aiktttticsöport, arilíísopott, |^ear®®ntkssep«»t, cikloálkllesoport, leteti· Cíklbalkifesóprk: anl^lkilrCSópori, ttetemartl-stkil-e»opprtS; Ciklpälkil-alktt-csopott: és ketemetklPalki-alkil-esopDít. ahol az ernlnett C;..^ alkiicsojxn·?, €.„> hakigén-alkil-esopott, C>.(> aiketttksep^t,. 'alSkSi^gpife· aííiosöpott,. betermttáiéscpsttj cikioalldlosopott, beietőeikioaÉicsoport, ari-alkll-csopori, beterosriittiiki*: csoport, cikloalktlmlkli-esopott vagy ΐ!θ!0Γ£χ:&amp;1οο10ΐ1··οΙ1ί11^5θροϊ?1 adott eselbsn szabszailoáiva van: t, 2: yagy .3 sz»bsttiíaenssei.; :mely :sz»bszíít8et!s(ek) egynráslól töggsíienöl: a ksvsttkezök közöl vae(oak) kiválasztva:: Oll, Ä. antlnoesoport,: lÄgloattmt, Cä,* aikik:sof5ort, C&amp;&amp; haiogémalkti-osöport, Cu. lattogén-alkiNköpött, adiesepart, ^l^fkltei^îÎSîÎ,: hertroadfesppert, he^o^ï^ylilHçss^«^» ieiktelkiiesopost és beietoeiklpalkib sssapsst; vagy 1*' #s W* -W$ a ndrtígénakípntal egyi&amp;Ç apéijiléz kápéspíMik, 4-, 3-, tk vagy ?4&amp;gú !5«ieroçâkïiôa|k|icsç^ort® ájkot, amely ailed mákm. szobszíitiátvá: VaR L 2 vagy 2 sirtfezdiöeßssel, jttsïy sâgÉHS3®ia®RS(ék) egyshâsièi függetlenül a kivetkezők febzn! vanfaak:) kiválasztva: 0¾. CM, makm$ô$&amp;t%i àa» fegémitoit·, €),&amp; alkiksopon, Cm. halogén-alkll-esopork adlcsoporg aril-alkil-esoport, heteroanicsoport, heieryanl-alkil-csoport, elkMalkilésogort és heôaéîkleâlkllésoport; R* és Rà, apkiásíáí iiggÂaik a kövÄsÄ közül van kMasztva:: il* €].» sikíiestjpork CU balogé»-' Äkcseport, Cm alk«nilcs8|pft, €** dkkilssoport,. anksoport, isieroadfesoport« eikloalMlcsoport, kPemcikí<^!k3les8p85í> edkálkil^soport, hert?oartt-alkil-s:Mp8rt- eikioalkil-alki-esoport és feetertteikloalkil-· elkii-esépMC aboi az említed Cr,^ aikiíksoperí, C).„ haíogén^aikíl-esoport, Ck* alképilcsoport, Cj.* siklnii-csoport, laAsoport, keterösrilesoport, eiklealkilcsoport. heteroesklrtslkifcsoport, adbaíkil-esopsrt, heteroari-aM"éSö:pórk Cîklôélkil-âikiMcsïipoîî és ÍveterocikioaÍkii •alkiR'spport, amelyek átfed esetbe» szsbsziiiíálva vannak I, 2 vagy 3 smbsdfeíensséi;, mely szabsdiéaens(ek) egymásáéi függetlenül a következik közül vanfaski kiválasztva: Ód, CM, anrtnoesopert., hafegénatoss, C>:.Ä aikilesoport: Cm Megén^Idt-csopert, C*.* .balogért-· aiklteoport, .ariíesöjprf, ani-stlkl-esöport, beteawfespod, keProarikalkll-cssport, eikioaikiksoport és kére í oc ik hal k ; 1 esope rt ;. vagy Re> és %? azzá! a nkrogénatonmtid együtt, amelyhez kapcsnfedik, 4-, 5-, 'é~ vagy ?-tagü asierscíkloaSkikSPpöítik alkot, atndy öáoh esetben. azobszfirtdlva van 11:, 2 vagy 3 szobsztitoenssel., mely , szubsdlmensfeíC egymástól: fläggelpaäl a következők közöl vatinaR) kiválasztva: CS, CN, ambsöcsoporg ba-fegéptipk C ; &amp; stlkiksoport, €*,S;Sialo^a-sIkll-ssoperE, C< ,ft halogeo-alkskesoport., sdlesoport, odMalkö-esoport, kPlpl'öadlfesppöö, teprpavi Wkikcsoport, eMrtaMlesoport és lieterociklosikticsoport, p értéke ö, 1,2, 3,4, 5 vagy é; és q értéke 0, I« 2, 3,4, .5 vagy é; saches la, mídoptoliiérativ resdéiénesség és rák közül választod betegség páciensben, vali kezelési eljárásába» történő aicalmazésra, kpÂbïleiéiban további gyógyászati szertől, % Ag; :L ígéiypáüí szedőd vegyűtet vagy ;si alkalmazásra., aboi a vegyüld egy, a íi képlet szedni? vegyi-let:
  3. 3-Pretm?ô-1-[4^7H-pirf«?ô|2,3'd]pMmâdâ^4-4:î|-lî3^àfa2DÎ-i-âl|-etiî|-M-ei:kMex5^bg8s^sza!ibîiSiï5Sd; iij-propânîdïril; 3-|2-ciônc-!-(4<7H-»lm>!0[22)Kï]pbirfikîÙ5-4-il}-Uipi!«2<iS-î-â!)-stii}-H>N-dfeti!-ben2ôte»ijroRaîni<l; propânnfPd; K-I,345S8^sjxö3^S4ir3-p-daB8-í^4d!?^*!>wMl>3’djpifímídd^4-tl)“íB~p|papp^;í-4í:]"etí^-4>eö20^ sxulforsamki; 3-|3-[p,^dí8íeíií:-8S8:MíK’44á^S28lfe(í|~feHSl|>'3'p'(7?3'-pírr0Íö|23"d|pfeííni4í^4d!í-i^pipi2oPl--Il:j- pmpdssidHl; 3·· (3-((4 -ôxo ·ρ iperiÄ-1 ·4}-ϊ£ΐΓ(ίΙνπ4]-ΓίΐΒ!}-3-ΐ4-(73Ι-ρίΐτο(<>!'2,3'£ΐ|ρϊη:Βΐΐά!«-4-;1}~ fH-píraxol-I-dj-prppájmitní; 3:-p-|^csklöi'íexl(-8)CBÍ}"^/:ülfbn!y-(Vn4 ^3-(4-( ?I4-pííK4<42J-d]pirimidm-4-H}-íH-pirö2oW:#|-ppepia- niírii; 3*i3-(oktahid^tí7^»kmoIííí^^l(fl:)Hí^á^ö|^n:tÍi)Hfsn:ú^^ -I^^TH-^ppioilóiÍS^^^Jpíí'ímidso-^^íi^ Î H~p£?-í«®öl-l-(ί|Η|5Γ4ρδ88ϊΐΓθ:; 3-{3· [p- fersH-ei il *-szu}fi>nil j-fenil} -3-f4-(7H-pirroio(23 -<l |pirmadm-4 4!lHM«pk8^l*^Íl-|Ésg#ÉÉfií| cisx-b-p-i 7H-pmO[op,3“d]pirinísdin-4-il)-ni-pirBí.oi~]-4j-cikk4;<;xil-ae8íOB4ril; 3-c(s/--!<-p47H-pin-oU5{’2:!3-d|p!n84>ásR-4-(iV(H'pír;izo(-i-H(-cik(o(íexil-propáö:M(rH; cisz>4-|4'>(,7H»pÍTTOk)P.3'djjpí«ra5dm-4*iÍ}»lH*p{fazol“i4l]-cikbhsxáyfka?t«m:itó; 3-(4-(7B-p(rroiol2,3-d|pá'Piúdín-4-(l)' 1 H-pirasoi-1 -irpc&amp;iohexd-acetositbl; 5-{ (o3S2~4-[447H-píri'<>]o] 2.3-4|pírim{din-4-il>-n-{-píniícol-l-jl i-cíkíohe>íil}4 ios- S H-1 P,-l*tri3iOi-3^^É^· jp^2-4-[4-C7H-pirro}8pJ-dipamdäui-4-jPr1H-pirü2i4-[~iä]-t~(lH~!p/7-«ia/oM-ii}-ükl8hexiä j-aepäp* mim; 3:«[3*(plpéra2;d5~!>i!-s2ü:l1©Bi!>fcd|-3-|4“í73'l“píí^top;j3-d|pmmym4-p)~iH''Pir£Uíobid!J--prapáí(nitrll; 3:-|4^7}|-pÍ5-r8bpj-djpida^ídin-4~ií}-IH-pK'a:2dl-l43J-3-|3-(tóó:íp0i:ípIm-4-í(-ii;íi!fbn;!>íeí;i!]-pí'Opáí!·· odri); ^3~|4^C?;ir3-piî'rôJôg2:ÿ3^ài|ip:3ïiwMliï-44i;ï^lM^pîrâ propâamïnj; 3-|3-|l20böíd;-sxöl^íi)-il^d|-3-(4^(:3B-ípirr83e|2f3-dlpíímldk-^-J:!|-l7!-p5:íaKöl~:!:-il|-p!ri>pá:«í:öif:íl5 3-:(3-(3M-p3iTc48P?3-d]p!ysasd3n>4-i3|>-l44-jdra2öl-l-sl|-3-{3-|(teiididdifö:-234-pbaB4"|biSsb||-szui:feid|· ί eïi ϊ {} - ρ ropis nr: n r * 1 ; df-'ppppâpôipï!; nitrií; asniíi; 3- { l-Í4-(7H-pirroilí5P3-d|pldmid:5R-4-il}- llü-pira^ol-1 -ijbup3-ss-l~i4f-benzomíril; 4- {1: -í i-(3 -bmss-iswíi-tyut-^es-1. -íi] -1 Hi^toaW-i) -7}4^iiTo!ö|2;3-il|pMmidi«; % {4,4-dïfiaor-1 -toeíízoííiöii; 4-C ] - {4,4-dí%Kir-1 -p-Cmorroisn^'d-äzuUmsf Î )-í«nsi Jbui-3-es-1 -ü*- ! Η'ρ?Γ^.οΙ~;}--!!>7Η-ρί jTok)|'2,3-d}pl-nroidm; 4 -( Hí -J3-{eíü- ^ÍIbhíI)- feHRJ-d^-difiuoy-hut-a-aí· Î -Ü}--í H-pin^b4-41}-7H-pt*Töbí2P-^ Ή í - (4.4-difkior- i -[j-tmetíi-sjaaj i főni D-f«ai I j-biít-3 -en-141}4Η'ρίΓΰζο!^-Ι!}-71Ι~ρίπ·οΐ0Ρ,3-^ρ^β;: 3- {[4-t7H-pjrjOÍop,.>-d|pirfmidm*4-iS}-1 H-pírazol-1 --l'j-meUi] -bsm'onitrii: .>·{ í-[4-i 7ΐ-ί-ρΐΓΓθϊοΐ 2,3-d.1píríjnid!ri-4--í!'V- iH-piraïoi- !-il j-bisul!· -heïœimtrii, 4- f l-{l-p-feiïl-sa«!fon}lH«niî}-4.4-diniiOf-bîîtilj- [H^lnyjoM-iSVTn-pirroloPJ^pmmidb; Ή ! ~( S -clkkî|x*otü»bui-.î-e«- i -i’b- I H-pli-a*oi~+-ii]-?H-psrrol^[2,3-d jpsnrmdm; 4-j.1 -{I -clkiopcmlbbrnsi)- 5 H-pirazo! ·44!1·?Η^>ύτο4«233κ!1ί>«1κΐίί1ίη; 4i|l^4J4-difl««^l><t«5rald;^Äba&amp;*34b^böb3”en>Nb!-b4i»Faäoi“4di]-7öbpädr<dd|3^3-dpä«myift; 4-[|4l-Äl opeÄ-4 ,4-<b$«Pb<hsMb' l wSio|2 J~d|psf imife; 34d:-mdii-dkâope£td;}-4r[4-if7ï4^5îrobp,3~d|pir}«î:ybs44!b14bpîïaxôl4-4l|-pri)jîâïJâïiô4l; î-|2-eîâJio- i 4:H®-pins»bp,3-d|py sddb-4-di)~ í H-pIraæe b í - i f-s?tü}“C;kfepröpäri-karfeosbn I; 34H^eíil--S2ulfonüVpirrolbin--3:4]|--3-|4<7|l--3>tolöp;Jb]p«iís!diív4-!l)-:iíl-|5ífSZOI-l-íÍbpröpéR«3ö^; 4-1 41 Id~in:sd;i?o!-2-d”í!«íÍí|--sÍ!bi-l43í-p!föaöb4-4Jl-2í:b^d&amp;ídteC2i3*d3|íirimiá«i:; 44Η^·^)"Ι^»|4«ΠϋθΓ-ΐ-[{4-«ΐ!;!Π-ί J-íim;öN24b-saöl:d|-edl-3H^raxol-44]h7Hibrroioi2!3~djpirhii5dfej 4-| J>fi-($4#b-pÍridin· 3-413--+,4- tHJtBor-huí-3-eo-1 il] - i ïi-pii<&amp;«)M“il}*‘?H-'pirr^o{2 3~djpmniidh; 5.f4j4Hlif]uof-Í-[4-{7H-pirrou>]2.3-'d!pirÍ!'í-idin~-4-i!}-lH~píraí’BÍ-l-ji]but-3-en-?-il}-nikoímoniínl; 3--[3-vj>irroHdb£* I -jbszujftmilV bíd;|-3-{4-i 7H-pírr<4o-l2,3*djpíÉnddl«-4-i)'ÍH+p£ra20·-3 +ll|-pöpáíísiín!; N-benzí I-3- {2-efe^-Í:»|4-{7H -pim>l5>f2,3^)p}riînid]n4-il>4îî^il^j^l-il|^l)-N“®ssîü-|ii8âÂ^lfôa-: amid; 3-]í:|:3- {2^:|äfip-4 -[d^PII-pprofepJ ^Ipin^áÉrHÍ-i?)« 3-!%ik4äsßMk||^|+3e«il-sz8lfeaslbrpssid ] ~ bessösifel; 3-p-f (2«ii-Pieblbs^lfesd3'|-fe d} P-|4--p5:bpiÍFr(deP:,3~d]píFMd»4'-g>3 8-pim^ib-£Î|-pïppâ3æifeiik: 3“|3--Í{l'-lfedÍ-8íd>amÍlbni!j"íenii}“3-'|4*C7i1'-piríok>[2,3-cyp!ripi}dIí^4:4i;k4b'-pteol-]-4l|“pPípá®d8'í:i; 3+p+c iaap+ |-Í4+pl3^|frplo-[2,3 -dip sr»dip--4-ä I b U f-pirazo -etil } - N ^moboJbHbbëb ψ berizotezulfonamld; szsdíönannd;; ! · [<M TH^ânRSîo-p^-dJpirnRidin-^-a j-1 H^raz»W4|^il|*M«É»lS(4>öiípía^ $>':|:^c:iarsd-14;N(?Hi5ST?ole42J'djpírimíd?n-«-il)4]^pim^^I“íiNíl^^^^afe3É'et3ran“3-!;l:-ís:ea!>· benzölszuifonamid; 3 - {3 {( c îk bpïopl-mets ll-szal foasi l#ni| j- r3*Í4-(ÍMkp«?rWfe[23-d j:pÍFteiá^:-4rÍ)-|>Í-p5raml- i>||-propámsitn i-íri íksoraeelát; propásmitril; és 3*|3··|Π-ox k!mripmsrlblâî-4dl>-saïdÂI!|‘^sd1 i H-pjrazal-1-d^- propánnitril: vagy gyógyász&amp;tilág elfogadható sója.
    9. A következő vegyület - ?-cikbpeaîa*5^4^7H‘fî«;ôè3|2>:3"4lp^®»àfe'i^“â|·Ä'”pißä2«3|-:N?]>-p^toiW. vagy gyógyászaübg. dagadható sója; kachesla, rnlekiproitjíímíív resdelitessség és rák közéi választött betegség páciensben való kezelési eljárásába»történő&amp;t WmSzisfS*fc«a&amp;felÄ»8 ip^abfergydgj'ászaíls^sel. 1(1. à következő vegyüiet: {EkS-ciklopsntl-d-ld^/H'pimiktp^-d^risayjd-d^l-ikl-pfeaiol-i'ttj-pmpánmlril; vagy gyâ^teb:i;feg.eÂdg8âhÂs^fî'^ibs8a5.:«ifeIé^6ÏiâiÂf:Â&amp;iïlæ^ség és rák fem ! választott betegség pácíerssben velő kezelést efdrásában történő alkalmazásra, komkisdctobantovábbi gyógyászati szemel. Il* As kővetkező vegytilet: Ι^ΐΕ{ρ|^^)Ι>3-|4^?0^^οΐΘ|2ν3'4^Λ^^·'·8>ρ.ΐ*»20Ν··ί8|ρ0ά|Ϊ0^^ΐ' vagy gydgyässattfeg ellögsdhdtó sója;: kaetaäa, mietppxdiflsrattv rersdellenésség és rák közti! vÄszÄdsdfegv ség pác-znsben való kezelést eljárásában történő alkalmazásra, kombinációban tovább; gyógyászati szerrel.
    13. Az Ml, ígéaypöíapk; báppejyite szerinti vegyfiet yagf- g#tÉ«$ A tovább* szer kemoterápiás szer, !3 Az í i $, j^^y^MÉdk.'Ni^pk« szériák vegyßM vagy só alksloiázásira, aboli az e|iárás a vegybtdi vagy se és a to vábbi gyógyászati szer a páciensnek történd egykiejti beadását Ibgtatja nragábáp.
    14. At $4 3Í. igénypontok bán»e?ylke szerinti vegyöiai vagy só alkalmazásra, abö! m epzás a vegyillel vágy sé és a további gyógyászati szer a páciensnek történó egymást .IcävÄi^^Äi^pipt magiban.
    15. Az 1-11, igénypontok bármelyike szerinti vepdtet vagy sé alkalmazása, abet ikdrtMbelöl 5 - körbibe-Ä i ÜOÖ mg vegyidét: vagy só vaa beadva a páciensnek,
    16. Az 1-15; igénypontok bármelyike szerinti vegyüiet vagy só alkalmazásra,; abet a betegség mieioproblcratív rendellenesség. 17. A 16. igénypont szerinti vegyüiet vagy só alkalmazásra, ahol a. miatapmiiierativ rendellenesség a: poliebémla vera esszeneiális tromhoeitémla 0Q, nsteloid metspMzaa/mteioIirrozissal pábábá|; ikrónikns: mieipgén teakémia: $CMCk krónikás mlelomosmcitlksp leokámia í(MMLj,: Mpereozinoill szindróma (PtaSjí vagy szisztémás hízósejt betegség tjSMCD}. II. A. 16. igénypont szerinti vsgvöfet vagy só alkalmazásra, ahöl az említett mleioproiSraliv heíógSág a poiicítémia vera (FV). 19. A 16. igénypont szerinti vegydiel vagy só alkalmazásra, abet az említett mieioproHferatív betegség tsz esszenciális trombochémia t;ET>. 2!), ,4 16- igénypsni szerinti vegyidet «agy sö alkalmazásra, #ö! sa 'miW&amp;Ü mtelopmliia-abv betegség a ntieloki metaplázia mtefeibrbässaä (MMM). 2:1,. à. líh igénypont szerinti vegyidet vagy só alkaknagásra, aboi a nneloprolilerariv rendellenesség króni-kns: mielogén: leukémia (CML ). 22, A 16. igénypont szerinti vegyidet vagy só aikataazásta, aboi a mielogrolifeealiv resdaiteesség krimi-kw:mwfammmM&amp;· lénké sa la (CMML),
    23, Az 1:-15. igénypontok bármelyike perinii vegyidet vagy só alkalmazásra, ahöl a betegség rák. 24, A 23, igénypont szerinti: yegytilet vagy só alkAmzásra. ahol á rák mieióma multiplex·,. 23, A 23,: igénypont szerinti: yegyulet vagy só alktíte&amp;zásra, alsói .s rák kssnyáhnirigyrák. 26, A 23. Igénypont s/ermri vég; ukn vagy só alkalmazásra, ahoi a rák leukémia. '21. A 21 igeos pont szerinti vesyülvt s agy .sé síkátázásra, ahol a rák lánfóros. 2k, A 2.1 igénypont szerinti veg> ölet vagy só alkalmazásra, ahoi a rák emlőrák. 29. ,4 23, Igénypont szerinti yegyalet vagy só alkalmazásra, ahol a rák akut mléfegén lenkémla. 30. A 23. igénypont szerinti vegyidet vagy só alkaltnazásra, ahol a rák tüdőrák. 31., A 23 . igénypont szerinti: vegyülni vagy só alkalmazásra* ahol a rák S$-sygkí rák. 32. A 28. igénypont szerinti vágyóiét vagy só alkalmazásra, ahol a rák a következők közöl vart kiválasztva: prosztatarák, veserâk, májrák^ gyomornak, pajzsmlrigyrák., glioblssztojna, Laposi-szarkóma^ C’astleman-bétégség, meteőms* bőr f-seft ImriSma. (iCXCLy Mr S-sgp Ihnfóma, Sezany szindróma. mpesis hmgoídes, :$kul lonfoblasztox leukémia és kok>rektábs rák,
    33. Μ I -15. só aitalnázgsíi, aboi; a kaehexig rák miatt van vagy azzal fcapexoia»·,.
    34. Az i-i i. igénypontok bármelyike szerinti vegyidet vagy só alkalmazásra, ahol a további gyógyászati szer revíimki.
    35. Az ML igénypontok bármelyike szedeti vegyöiet vagy soaiknipaMkMnhril^k : eljárás továbbá ram gában foglalja koriikoszíeroid páciensnek történő beadását is, 36. Λ 3 5 igénypont szerijatl szerinti vegyidet vagy só aikabnazásín, ahol a kmtíkoszföfoid prédákon, 3?. Az-1 -1:1. igénypontok bármelyike szerinti vágyóiét vagy só .aikalms&amp;bsza, ahol: a tövaMf gyógyászati szer D'NS4: árusító szer. 3k. Â 32. igónypont szerinti szerinti vegyidet vagy SÓ alkalmazásra, ahol a ÖNS-károsÓó szer a: soel&amp;ián. doxombieln, elkloíbsxíámid. vinkrhzitn, etopozid és: kámms/tin közül választott. 39. A 24. Igénypont szerinti szerinti vegyüleí vágy só alkalmazásra, ahol a további gyógyászati szer # melfalan, melÄp plusz prednizon, doxorubicin, dexamótazon «s bopexotnib közit! választott
    10. Az ML igénypontok bármelyiké szerinti vagy SÓ álknlmazlsrá, ahol: a további: gyógyászati szer gyalladásedienes vegyidet,
    3-Cïa30-H..(3-|2-aia8O'î-p-{7y-ptFíx>Sop3-4|[4riiï^i(kí^4-U!-II'^'pirazc>í-:;-äíl-^ti}}-ieπil;-feδn^a8îîd; N4.í-{2-ciarto-k'f4-(7H-p:rrolop>3'dipirin»dií;-4-'2V-LH-pira2ol- -íefB:!}-4--(triíl8ör~85«íH}- ben2ss»»d; Í2-Cíam>-!-|44 7H-p5;rmíoP J-dlpirôHïdfet-d^^-M-pfeiZôl-l-ill-etlJ-feiiï^-N'-fe^d-kaïlÂM; J- p-<3ä8o-l-|4~(7;ii-pirFö]423^1pi0i8äd:tS'4-äi:p I H-p&amp;aæed-l benaaroid; ;JrP<iafa>^44.'W^Î'£ro^2,3Hlïpk8æ^fe^41HM^Iïmlr-î-îî|*etR}-H44'®^MeR*li>bma»H8^ M-C4-ciaße-te:i)-3- |2-oiasí5- ! -p-(7H-f irrolopp-d|psi8Mdm-4-5l> líi-pÉppöl~1 -il j-eíi 1} 45«82anddj 3-{2-cias5^l-|4-{3P-pía-öfep>3-d|ptónidi8-4-íl}-ÍI4-|»razol-lai:Íi|-etll}-N-2-nsfti-feóPzamí4; 3-p4ÍS8ö-4-p-{7^pimsbp J-d|p3riR5iáinP4l}~íí3-pk3xol-I-i]-«í5l) N-Hsaftil-beíízamííí: 3- {2-cisßo- J -|'4-(7il-plrrofoP3-d|pîd5sidf8-4-:îlHi4-pira5»i-l-il-etïlj -N^-diitiefil-beKsamid;; 3-<2-ciärtö-]-|4-i 7H-pim5fep,3-d|jpààï8ldi:ô-44l|-lii-pir3Âd-l4l|-*Pi|-N-fiàiidâK-34l-be8î;a3sld:; 3- f 2 -c saso- 1-(4-47 Η44«·ρίο[23·-4|ΐ858:ίί4ίϊί-4--ί||-1 ^-piraastí 4i|-etä ) -N-í»öí:íl-bl-lei33l-bes5asffiid;; 3-{2-c;afiô-i-|-H 7)-^ä«^8{23-4|pWmldin-4-i|-iH-pä£^irMi}^tiil}-N*äÄ»l»«ü-beaMmldi: N-P-eiapö· feuO'3-|2:-da»ö-4-p~p^~p|08:iöp,3-äjpkli8:idS!5-4-||^- IH-pdiBöS- Nlj-edil-bda^oki:;; l444Ælôr-iesïï|P-j2-eiaïiô-4-[4-(7i-|-pàa®fep3-45pMï83d!iï-4-3l}-IM^ïi82ôl-l:4lj-«i|-beîS^nâîÉ 3-p-etan3>'i--p-í3l4-pÍ0föto|23-d|pinníyí8-4-i:i|-lbl-p^8oi-44í|-Píílí-ll33>4-dÍ3n^I-:fe8lÍ)-beííZ8í8Íd; s^ulfo.níisttid; a-p-c-ancs- i-[<b'{7H--pirroäs>|2,3<l{piri;-iridt!?-4-n.S· I il-piraxoi-l-iil'-eidi-N-priipfl-bößircfisxuddnamiid; 3-{2-c:axü>-1· j4-<?í{-p!Ffoio[23-djpiFÍ!53:da5-4-il|-LH-pkaa4-!-it]-^tí|pl4-:3-Fa5lSIAeimds28ÍildPa:mkl; 3-12-ciamv- i 444 ?H-pirmlop.3-dipinmÂ-4-ü3" IM-pamd i Ul-aídplkcífelopöpd-benzolszölfeöamíd; •Ηί^1^^4^^4Ι·^^Μ^8)^^8|Ι]Ηΐ4442ϊ4ρ^0ΐΙβΡ^4^ρίΛΙ0^··-4ΐ#-ΙΒ·^®τ»»θ!ΐ>-ϊ4^-ρΓ©ϊΛ8^| 343Hn.orb;>Í!n-r-d-h/.uífo nlHa ssj] I [t j?H proiyfV djpmmädr-4 ιΠ liî-p uayl-i-d'-prôjsâsîwîm; 3-|2-63iS8^l4447l3-:pifroiop,3-d]p?r.H8(di8'4-ii)-i3i-piraaoi-l-ili“t4n}-N44-mpdi-lesiO~bana0lsi;idfo55^ amid; 3-:p-eía8ö-Í-p42di3lFFob[23-íÍ3:piíáínMfP-4-ilp|:ií-pda2®l-l:4l]-dtd|-N43^-d5P5öi:dr|eai7H><sss>i“ s/ujfona-rfki; 3-[3^berssil-satjlfoi»l.>-fcnilj-J'[4'(,'?n-p*n'olol2.3'^]pirirnidin-il-n)-UI~p{ra2öl-]-li]-js«pääaiir;i; 3434^^^“tíi>)--fei5^j-3-|4^7H^TOö:íiáÍ23^J!>®^Ms8^^:ÍÍ-líÍ^síraz8^1-i}-pFopáx«5:tírí| ise«20i)i5ri:{; 3 -12-ciäse-1 -p^M-pííT ötepp-dlpimpyiB-d-i)- llí-pirassoi- 7 ~íi|-esM| -K^ssííMs&amp;bxoíszu íiíbnaasjd; 3:-|2^»ΐ^:ΐ4^^»ρίπ·0ΐ0Γ23·^]^ΓΜ^^ίΗ^Γ^^^®Ι*^Ι*^·ΐ”®8^^β^δ3®Μΐ»δΚϊ8ί^. 3r|3^to^i|0i3ii)^fea|^^4H(7il^»r«l^|j^|p#^ï»î4-ÎlVlHiîiï^M.-ï3'-|®opteî»i;
    3-PdsfdÉ>-i34:iS2öS~S-ii)~3-f4#ll-fS:m5h>í2j-íbplr!í{dd:P544í^?i;bpkss^b2-Iji5mp&amp;5öMk 3-[4H7H.-pirr<:<k)|24^á]piriínkljn-4-4V!5-|'pir£Kobí~sIj-3-[5-(i44iazol-24i-tío}-pi:ndin'3-il]-propá;®í.t;lS:; 3- |540ίίϊ4ίιβ^«ΰ48ΐ-3'4]~34447Η-ρχπ;ο]ο(2,3~4]ρίϊί«ϊ«ίΐη-4-·ίΙ)-!}·ί··ρίΓΑ?.ο1-ΐ·ΐ!|'·ρΐ'ορ&amp;ΐΐηΐίΓίΐ', 4- 1 J-P^ns&amp;d-S-PI-M Ρ2ΐ-^8^ί-Ρ0;Ν^1·4^·ρ!Γ32θ^4··ί1}-7Η-ρίΓΓρΙρΡ3-ΰ|ρ5Γίί0®!8ϊ' ;i:4æîîip4#^'bpîîi^lôp2-djpirîR5MÈ8-4-:il)-&amp;pÎî-â^~l-ii]'p{X>pa8-l-8«:;: 3-p4gM^^lfei^>pÂiP-3Hî3j-344#k!^imsl8p3-d||#imidi8~4-(l3-:kl:34s8^8l-i~H^pbôpàHo4riI; 3-í5-ú8d-»zxd1b8d)-pírPÍ8~3-Hp34447I-I-pín-oiof32'^^pírímídÍ8~4-y}-IFI-pira^óí-!-íl|-propáí588ríí; 34S4câfâÂxiI-ife)-pkMÎKv3-M|-3-p-{7M^îf^iop,3-d]pM8itÂ>44î}-i;M-fià'SiKô!-:S4!j-p?opàoriiiâ;rib î i: M-fâiaz#-:WI|>fS0pas· 1 -ol;; :3>|3-pîâ-mdSp:i:l|-;fead|'3-p#H-p!næloP:2:-d|pâriînâdœ:-4i-d;^::kkl4pir8^S-i-ïiJ*pi^pâa8iir|l:; 3-p#b:l»ssd:Sï8i^4M^3'P<7H-p8:mlop J^i|pÍ!'8pídÍ8-4- Π>· \ H-piraasM 4i|*pri>pà8îÂI;: 3i-{Hfe^fciíési-S28Í!ön4|'p3fláí8-3'|3^3~|4-(70-fín'oio[2 J-djpirimidin^i'lt)- ? PÍÉÉÉ 345-(cíklobex!Í-szuÍfÍ8ÍÍ}-!>ií'ídíx:-3··!: j- 3-f4~(7H-pirmlof23 3jpirÍK8<lm4M3J“3 fi^teob'l 4IJ-pi®fsáí8dírd; 4-|^ί^ί00^2·<3^^ίΪ|^ΐϊ]-ΪΒ-|>'8·3/ο{~Ι-Η>-7Η>·{>ίΐΤθΐο|2,·5*ϋ|ρ»πϊηκ]}η, *1]-i H 'ρΐΓ82Χ>1'-4-ΙΙ>~?Ι·;Ϊ -pírrolo(^,3“dl|>ír»«id«íí. ·4>{;ΐ4;ΐ:^ο{|ίΙ~2^-η^|ΐ.-.'|,|ί4-οχ^ί;«2θ}*5'Α)·>©ΐ1Ι)*{Η'ρΐΓ8Λ)Ϊ~4··ΐΐ>··7Ι!·ρΐιτοΙο(2,3·^||ί^8^Ιβί 3-|l-fmetii-s:2iÂfirl)-ffi®l|-344“(7Hi>inokîi'2.3-d]pirim^in^-î0'HI-pirs£ol-i'ilj-pre|5£m»iîrsi; 3<3-p)íidin-4-!^fenii)-3-:4-(?H-pim>b|2>3-d|piri?«fdín'4-ü}'-IH-pir3XoKí'ií]-propárinü:nl; 3-jS-|!Xí>|mípíí-mdflnil}-pirid!n--Í~íj J--.>-|4-i ?5-l-pirrolo| 2^3-<ÍJpmmtdin-4-íl)- 3 l-i-pifa2f»î- i -IlJ-propádSitrib -H4-(7H-plm>4ös2J-d]plrirnsdin-4dlHH-pirazd~l-ií]-3-[5-(írit]uor-metií>-pÉrídÍ8-3dlj-pfopánn?lri3; H-plr apő3-l-:dJ-Nd3 ^Âub-meil^feÂl-pfôpàrmmkl ; N-2-oafdi2>.d4"(7Hpùy<>kif2s2-ii)pîr;<n«iin-4-iâ}-H3-pi:nii'oi-l'ii!'propàn»ïî?id; NdS-cbnO'féÎïdl'SdMJH-pdrolôP^^piibïMb-dd í)-lld-pdaïsi-bâlj-prôpàisajïïid;: î^“bRM-2-|4-^7l4^bebp3'd|p!dœyb^457-lM^lm2eP3-!l|-fe8iâHâî8ïd; ^{4--f<uKí>;(-btid}-2-f4477H-pirn>io(2í3-<F|phimk3H^4-i!)-2H-pB'azc3-3-il]-b«íá«íÍKí!d; t3»2”|447Îi“piffi4oPP^pïriinidm^-il~I !!-p3molrl 4114Hiiaîï30)34;: 3:~c iss0rbsin^2-|4-P^-pini03pP54-dlpfe4a5ab"4-ii^ 3 M-p3îaKfô?-i-dj^aÂpaid·; Mbsfedl;4-i3^2^[4d3i4dîàïc4ôpp-d||dïb«db^-j§'4ïl>|i!îaKoi:“3--iï|>ibabB3:mïd;: M-Cbî3ssj3b-i-5Kôtin-2-|4d3;iri-plrTô3d[2pb|piÂidis^-i^:îH-piîSïîôl-:i-sll-byîâii!};oïid; K>^i:ls{di-d-43-n-^d;i~2-{4d3:M-p«¥o3df2p--4|pkb?:i4i®-4-i!^d4lvpäräK©^3-4ll4iäbdäb:idi; ^-#--üäiöKi-fe«l])-2d447l;i“p3Ä!ö|2pp|plrävfs4b-4-d)-l!3“pl?a2ö|*|^i3|b^ä»abi4;:: M-3^palfc3i-2-f4-C7M-pîbKîlt^2^rd^Mlinid!n-4-nViH-pirsâZs>î- l-síJ-tnUánainld; 5K'2<lasô-l’i4*(7H*pJn-ob[2,3-à.lpïrfinidm~4-H>-tH~p!r8zoM-îl|-e{}U-N“f«otl-n}kotin8nîid; M-{3- ! 2-eiímo·- ? -)4-(73 ä-pirrolop J-d]pinî5îidin-4-îl s- ! H-pÈrazoi-ïdi}*euij dem0~3dtnilisor-rasdl§-beiizamid; N-Ü-J^ldTH-pifmlopj-dlpjrimidio-d-iJhlH-pirazoî-lbâJriS^l^irenîOO-Ctriîluor-jaèÉÎ^feéiSîsaïôidij M H«íed!"S..tii]í;){!Í0"ret»!j“3-44-(?H-pírr(>Í(>p..3-d|pírÍHtídS«-4-í3}-íí{-piríi2oí-í~in-pnapáJííiiíri3: N-f3-^4d7n-pifrolop>3“dj^irirRidin*4-n>*lH-plra2oi-'Nii^Ä|:*fep»t>'benK(}-S2u}ib‘ß»m{d; 3-{[4~(7H-!dm>lo[23'~d]pirbnidiB^-ii)-3i i-piníXol-!di|dpPdi|#P4tníb.tor-!netii}-feri:Íj-ben;^índd; 3-{2~daR^:-pd7^pirroiop,3-d]pínmidsn--4-5n-n]'psráí;o!-t~s]J-er;ií-M.N-dsmc'dl-heim>!síu!foriam;d; $4rl«tóJ~3-f 2-ciano~ i -|4-{ 7H-pirrote[2J*á^rw\iáin~4*ji)» 3 H-piraxol- {di j-e? jjj »her>2ols.culfonar«id, I4-bem:§-3 - {2-ε v&amp;m~ f '|4-{7H-p:irrö3ő|2,3 -d)p:rábdb-4-íl)-- I H-psax»!·- 3 dl)-őíi] -hem&amp;idid ; 3-Ρ·'ΡΜβρ··^|447ΐΙ·ρ^'ό1οΡΡ^|ρ3Γίπΐ54Ιη-4-4Ι)-1Η-ρ:^όΙ-·Ι'·ί1|'«ΰΐρΝ-Γΐ;ηΐ143δβ^τί4; 3-|2^spe-1-|447 B-pb'^iöp Jrd|piriisid i n b dl)-í H-pÍraxel-1-dped S PM d 34 tri teor-medí Vfeb% benz&amp;m.id; N4'3-csaB<?-bîiil)-3-?[4-(i7I-l"pfîT<>3of2-3-d]pïriï»idîri-4-îi)-4H-ptraïol-d dlj-metil}~benz£jrrnd; M-ben;íil-3- {14-1 ^H-pi5Tcdo-p,3-djplîîîn|d|n~4-il)- IM-pírsxpi-1 -Üj-meíö) -beaïaœid; W~ 1 -·ϊϊ»14;ϊ ! ~3 - {Η ^ Τϊί ~|rf í3-roíö|2 3'^J» * ídmí;í$ifí '4^-d331-¾¾^ í~ 3 - ti -feeissa^íá^ H-2-na ftî ί-3 - i 14 -< 7 î ï-fi5rî€>lo| 2 3 -<î j p Iri itï sain-i- î l> î B-pimol- Mlj-roeün-bmramîd; W^3-f4>tmpl»1op,3-djpminidin-4*iO-îH-pirazo}-l-U|-met!i}-tèn!IV^.-8âftamïd; M43'li4^(?H-pml:kv[2,3'-d|piri!nidii;-4H-pirazol-\ -üj-meti; pferd !)- i -o&amp;ftamidi H~p3f3^oi - î -il'j-mei il>-fersil .s-acelarok!; 3-klór-N-O- (|4^H^^o^^|pHtíi^44i)rlP-]>im2sol· S -il] -rncui} -feöjl>«beaz^l^: NH'k·i^c!aîκ^-^d;447H-p!m>^2>3κ!jp!rbtPdin~44r)kl·^pím^ok^dl-ePi)^eπi!)-·2-naítaink1; M-í3· {2-ciaîiO-1 '|447^^-ρΪΓΓθϊθ|2:3κ:|[^ΐπΐχ·Μίΰ^-4·ίΐ!--1Η*ρίΓΚΖ0ΐ- I-ii]~edlf-4sfdi)-l -nabainM; P-C?~ P-piaßö-1 414- lld-piraadl- !
    ;3> Az i. vagy sgénypopí; szeriül vegyidet vagy sé alkalpgrtásra, áhel M jelentése N.
    4, .Az 1-3, igéayponiok báddéiyike sgerimi vegs-ülel ysigy so aikalaiasdsta*. #el :b értéke 0,
    5. Az; M3;, igépypooiök Msspelylks: szerinti vegyölét vagy sé s&amp;aipazásfa, ;iéol: n értéke t, ás'Ï jeleaiése ;C!:.sj: aikiléncsopört,. tanely aáot esélbéé szubszbíualva VSA % % vagy 3dp|pgéhat8stmaÇ üd-esepöiltál, 04-:rt^örtts|,antisöcsoperrtal, CU alkiikartitao-eseporttal vágy Cgsiialkil-ártvtik-űséperial,
    6. Az i-S. igénypontok fcânnelyik* szerinti vegyidet vagy so a&amp;aitoszáxta, ahol 2. jelentése cikloaikilcsoporL amely adott esetben szubsztiritálva van I, 2, 3, 4, $ vagy 6 szubszrituenssel, mely sanbszbtuenspk) egymástól taggsienil a következők köaül van(nak) kiválasztva: haktgénafóot, CM atkilesopöíC Ci« &amp;4to8pbfÄ ft.* aíkmilcseipork Cm. báktgén-alksi-esoport, Cm hidroxi-aikil-esopott. Cm-ciaoívaikík-esepork €y\ CR NO,, ÖR‘, SR\ C(ö)R\ Γ(0)ΝΠΐ<, CfOjORl 0C<0#b, 0C(0}NRcRy I'M*, NraoÄ prcxoiNr»“, nrtcoxjr*, stOiiiy síOímrí**, síO),r\ NKcs(OhRb a showím*.
    7. Az igénypontok bármelyike szerinti vegyület vagy só alkalmazásra, ahoi Z jelentése hetetosi&amp;iO' klkilesopeíí,; amely adott esetben sznbsztitgáíva van i, 2, 3.4,,5 vagy € mrbsztitae^sek mely sz*íbsz&amp;séns(ök4 egynvástdS ibgpîlepbl a kővetkezők köpi vadnak} kiválasztva: fedogexatom, €04 alkifesopott, €*4 »Ikentl-csopon, Cm alkiniiesoport, Cm balogéB-aikIl-csoport, Cm mdtsrivalkibcsoporí:., Çj.,î elsno-alkií-osopert, Cy·. CN, NOj, OR3, SR\ CiOjR*', CtO)NRsR'í , C(0}ÖR'\ ÓCf®|R* OCtOpÄ, ;»%C0)Rhf NR*C<0)NR'R's, NR*C(O)QR\ S{0}R\ $(0}NRsR\ Wl és SfOfeNKfe*. 8. A?. L igénypont szerinti vegyük;!1 vagy só alkalmazásra, abbl m vegyüet a következők közi! vág kiválasztva: 4-{M i-toetsk3:»pstoaskI'il“ptopil)-lil-plrazoM4l:)-l:Hi-pin^0loP:3'-blbltidm; 4-[ .1-p-ifâMassal- ! 41-1 -metikpropil^ l fí-p ksml-d-ill- lll^iiMfeppdjlpsIditß;: :3--öíklopropil:3>p42íi":Rkt^fcp,3^i:Jptomidái-4dl|rpkazoi-l4Ij-proptoakriii:;: 3-eikïppeitt};l-3-P-( 1ri-pbrofcpj -bip:iriÂ-4-ïl}*ll4rpkto:oi-l411-prefémdtrii:; 3:"eiki8bex:ik3v|44îi4:-ptottiô|2,3-b]pirsdto,4,5Ï}^lîkpi:makOîl|-pï^É!H5iyi:; 3-cék':0()éatÍ5-:v44-3?H-triíT<vi()[2;3-d]pír!írtidirv-4'íí}-tl\i'ptrazokiéi}-pKvpámikrí!; 3-ciik I ohejtil-3-[2,3 -dJpiFtïsî ïd:in-4ΐ jpapaaxxl ~ î - iP-psryxpámtMrí I; 3- i-j4'{7H'pirrokt[2,3xi|pirirnidin~4-tl)'{H~piraz<)i-i-iSj-cik.li>pri>pil-pr<späixiisril; 4- t:i.kl<ï|>r<5pii-3--['4-<?s;i-ptrt<il<ïpï,3-<rjpmmj<lîXi-4-il>- IH-píra^-ol-1 -il j-buí:£m£íítrii; i · f4-(7H-pifro!<f[2,3-d)ptrijn idirt-d-tí}- i M-psrazoW-ti j-ciklopropi'l-acetonitril; 4-14Cpim>lidm-2dí)~meííij-Í H'pirazx)M'ií*7H-pímdo|2<3-d]pir)tstátn; 44.1414met!Í'SZulfon!lVp!:n-olidin-2-if.|-metll' H'pín52ol-4-í!)-?H-pt:m:>kfj253-d]pí.di»iáin; 3- cikiî^ptii-3'[44?I4sp4ttô4o|23“d|plitnidin^-il)-tM^kaaôkl4||,ÂaisarMlj 342r-ciasô^k{4-i7kktxddfe#3-4]p5d4Ân-44l)-îlI^inapki-î4toiil"p|fciopeïa:àî5,kijrkaîpSîrtk 444 4>ensid M-tosxKoM4|), 4 H-pirnikî|â,3 ·:0|ρίνΙ4ΐ«; 4- f i -{2-nafti Hnetil}- I íbpib^kddil'^iS^inroIppj-bJpiiÄy 44 4déni I-tkÎ-^îtotaoi-ddkl-ÎH-pâtïiikïlSiS-blpô'fdia:; 4-meül-3 4441 H-pirrolePp-blpmd ίη-4~ί|- ÍH-pirazob1 dij-benvomtrii; 4d I -C3 J-dbsetoxi-bearikb í:M-pir3Zöl-4-il|- i M-pIrrotoP^-blpíddjö i 4414i~ferb!~eti| H H-pirazoi~4-tlHH4àiro.h^^ 3-{|44 Î:^piÂteÎ2»5^|pièiilfe-4-ft^ i B-prazol-l 4í|-^etil}-bítóimWÍ|? 2-(144 Äpityei<423-b]pkjdla-4-äi:}-113-pàtmd-]-íll-toetíil-ben^aitó;: 1·:-β?η·Ιΐτ·^ϊ-1··< I ϊί-^ΓΓ«Ϊ^2»3^||>^ϊ^*-^*Ϊ^-·-::ϊΜ-ρ»ΤΒΕΐΕ0ί^- l:.-£ijH^Ëa^€)^;-4-(]-[(5-aK^íyxoxazo!-3~íi)-m<;{ii]-ü^pkaíoí-4-ii)-}H-pííT<ík{[2,3-blptTÍd:in: ^fï-C:teimhy?0'2tl--plra» ·2-0-ΐϊ^ΐΗ)'Ι H^5räzoM-5!|~iH~p5rrö]öP,3-fe|p!rk!s:ß: 44 ? -cikksfcex -2-es?~ i 41- I !3^ka2si-4-P)4 M-pimdsf2p-bsplndsn; 44 i •äkksbexil · I H'pirazüí-44l}~1 M-pIrroiopjB'b jpindin; Î -<2-ß ÈôÎï^be332;t Íí- iíí^jím^ööí--4--1^--1 H*pin®lpp3“b]|5íri(Í5P; 4- {l-(2,6Hiikiôr-4<tn0ifôr-n^îil)-feniilj- ?H-p:!ra?.o.M-ä;} - ík^md<42J4>jpmdm; 44 Î 43-niPö-ber.zi!}~1 H-pirazoM-ilj-1 H”pimîkï[2,3-¥}|sk«iitpi 441 k2“bi'0S)-beßi'ii}-1 ίΐ - pirazo l·-4-4Γ|-1 íkpirrokíp J-hJpîridm; í H"pfc'<?feii3*b|pÄin~4-iä HÍR *p iramok \ -i|--prit>pása^áí 4-! !-ipkírílkior-ííisíoxi)-hínz:íllkM-pk2LZölk-ílh!H-pín-olo|2p-b|pírídl8; 4-1 H-pirasßN^iii J - ! BÎp!pk4ô|2p 'b|p^Âi 4-{ 4e-pi»!op^S3|pirldip &amp;Í I“(pÄöp3kkpie{|}~ l SpifazoM-iij- I Hi>i^|«|2p4>lplriÄi 4~{ î i l'fenäkbuulj-1 H-psraz«M4ïH B-pimbopJ4>)pkkkn; 1- fsn]k2-f'4-{ 1 li-pmokt! 2,34>]pkkih'4-ii>4 H-pirazoi- 1 · tíj-prcspan· Î -or; 4-|: î djpn ïîI}« i {l-pirazö1-441]"l Ji-ptrr«]io|2P-b}pk5Ä;
    4-Γ i -(2,,D-dirastii- feta I)· i î 5--püa?ök4> ii ; i H-pirroio: pJ-bjpirÄp: 24'443H4)mdof22kb]píridií544I^IH-pirazol44|]-g-(tTÍSöia^meöi)-bföa205aiín:1:; 4-( 14'4%àîK4444riÉtâiïr~py d^r2rílk^kpífazek4-d j-1 :B-]pm3fo(23 -blp&amp;Ä; 4-p: -(2,5-dííösil-i:«ik- i N-'pi*d*bl44î]- i :H-piirpb5|2:,3--b!pkä4iS; 4-|J-(2-s\eös-ib:ak3-líkptrazsM-dj-3íf-pÍTí®bp;,3>bpi:ddin:; 4-\ 1 -p~a:aa;öxkleml}~ ÎM-pkszôbd-s !j-1 M-pk®löP(34i|pkidik;: 3- { !:-[444l3-pbiölöPs3*blpaksk:4-:4 $? 5 pira/oM 4!(sid]4>enzon!iri!; lV.klér4-:f4-<!M-pkrok)f2 J - í ipse kJtn-4-d p i H-pírasKd-1 -ij-böszök iírfí: 4~( 1 4 i -ciktobexíketii}-1} ï-pîrs/oM-H !-H4-pirrok>f2,3-b]pkid5b' 442»»r-2{4-(fg-pfe>k42-.3 4^)p)Tub«-4-d)4H-paaz<4;-l:4í:j-beiHOíú4'iI; 2- -Íuöí'-4-|4-Clbkpíafole|23“b]píryk“4-d)”Ííl-f?tm2®b?41|-fegrm>íík?:ik 3- 0aor-4.(4..(i;iI.p;m^b:fi2>3-bJpir5difi-4-ii}-3H-pj:razokl4lj-benzor;!tdi; 4- { I --{k[3-{i:fifiixir-in8i:il>fenl1|--3ik} ·· {H-pinîzoî-4-is)·· · H-pim>k>(23-bjpksdm; 4- í l-<3.:v<btîxail··5¾ní!)·4H·ρk3.^ok4-·!l|4l·!-pkrob>p3”bjpif'id«'4 444-( I H-pirroiof2J4^irida>4-il}- ! H-psrazokl -iSj-benzonitrib {4-[4-bbï3~p}n-(ipp,3-b]ptbïdki-44 ΓΗ4 ïkpkazoî- í -M j-fedi j -aceíoaM:; 4db(renii-pfopii>iH-piraz<:4-4-il)-SH-pim>io(2,3--bjpirldis; 441 -11 -p4in«bkszidfbaiO-Sbníl]-ePlf-^ H-pirazol-4 41)-1 H-pkrok>[2,3~b]piridisK 441 -j 1 -(3 -líaoí-4 -r»«to>; t-femlkeíil]·· ! H-pirazol-4-il} -1 H“pirrok>(2J-b|piridisp 4-( 1--( I -p'#íSööf-ía«íd}:-lbíd íj-etil} -1 íí-pi razek4d}|- î H^irrok)|2J45|paádÍ!p;: 4-(1-( 1 îi-pi3*blpp3-b|plriÂ;: 4 -(144-(1 H-p àrolop, 34>(pmdto-4 -s!)-1 li-piraz<>!~ 1 -llj-eS il1 -heræomtnl; 4* {Í -[4-ai?ro-2-(tn lluor-œcd!}-fedi j -1 H-p:frazo]-4rii| - î : S~pirîefep,3'b|p5nà:% S4)ïpfölop3d?Ípy ^if^44 i)-í 1-il j-besxoi5isdl| 4-11~-p4;iér-:lenií)- 1 B-plraaol-d-Hj- 1 H-plm4o[2 J-hjpbidín; 3- bíóín-4'[4<IH~pimsk-42 J-b!p:4ídiíí-4~i?)-il4-piraxo!-i -dl-beozoohdS; etil 4”[44114-f feolof2y3-blpiddr»-44ö-4:^^}:f&amp;2fti-í-íil-beívxoáí; 4- |í~|2-yöf-6-:mö:^4MÍö^ífeííÍ)-lfe®ll-l B-píís^M-íQ-di-pÍTmlöp^dsIpiíM®; 4-P- p I 113-plmdoP Jdlfpßils}:; 4-(l-(23-dihidro- í H-indeo-1 -il}- ! B-pirazoM-d]- i }ï«pim)!o|2,3- bjplrkinp 4-f í -( 1,2,3,4^(^4)(4(0-(:3 ífeHa· 1 il}-1B -pinizol-4-ií) · i B-ptrrc4o(2,3-bjpiridm; 4-( I i1 -[2«fei:édS44ríÍdöí-TaeÍi Ι}-&amp;ΐΗΐ|·δίΐΙ|-!fí-piriföö3-4- Ifi-1 M-prmM3f Mslplri 44M l-(24<]!k]0r~5-nuor4dni!)-eti]|dH--päfäZ0i--441dlHi>(rre4<423-b|p4idm; 4-{ i -( ! -cikiopentH-äidV 4 i-p!öiXö^44ä|-ffi-pädo1e|2,$-l>|pmdmj 4-í Hi-fneul-3-fbnil-propdV i} l-piraioM-il]- i B3piíB4op3-l>!plridöfe 4-| 1 -( -cikiobidil-etif}- ! B-pirazoi-4-d]~tB-pirrolo[2 J-blpiridís; p-p-î Bl-pirroioP^ 5-bjplndm-4-il;-!:d~píraxól-í4l|-54|d4ÍH^medl^&amp;si|-«eeifö(ídrd; [5-[4-(lH-pidök>|24~líjpu]dii)~4-ilí-]íí-pin3»;o!- :f4!]H2-(ifcllteor-î»^iî)-iœÂ|«^iirt; 4- P -|4-fe«llkd-3-ea- i 41 j-U i-pirazül-441}-kí4-:pirrofe|2,3-b:|pirídia; [ 3-k Ι<5τ-2-{4·< i B-p]rB4dp3-bJpíBdia-4-df- tlírpírszsl-l -d]~S4trii]aer-medî)-:isa4|-^tonM1:; 5..[4.( {H-pÍBBÍo|24-l|p5ridib-4~d;)-IH'p]raxad]-^l-Sdfeifldör-'mfiiilJ-ijeözaßddk: 4-(l-f2-k Iór-4-(tri Üoöf-oíoííÍI-ísoI!}- lB-plBszol-4-d J - 1 li-p|í'rcdöp,,3~dtp»'Μϊκ; 4-(4-0 H-prnrol of 2,3-b}p!r id 1 t--4 4 :l)-IS-p«aaol-141j^2-(feíiöar^B^:í)-b«8míW8í: 2- [4-f t ϊϊ-ρί:Γτθϊθ|:25.3-4ΐ3ρΐ3'ίί.ϊ3ϊΐ-4·4 ]}-3ϊ4-ρ:ίί·«^:ί:}?·-ί--ΐί[-4ΐθίΐ^:<>πΐ5:Γί]; 3>kl4r-2-j4-(314^pfcf«tof2P-fe]pirife:-4-d])-ii^plra2ol-l-$]:j-beim5a3irii; 4- 8^^0-5,6-4111^0^-2-14-(] B-pirro lof 2,3 1- ((4-(:1 B-pÍBt4bp:,3-bjpiridin-441}- ! B-pÓ4^1-l-d|-n3«ril 1 5- bróo5P-p-(4 íi-psfrolofd^-bjpíridío-d-i^lö^ia^ol-]-d]45doÂ5îEi;: 3- |4-(;ít~pin'oló|23-b]pir!(líR"Í-ii)-rH-pírszo]-l-!S:l--1-(i(\Ífluor-m<ítí]3-berímr!Íirb; 2- f4~(ie--pfeo3<4;2J~b|piF(ddB44i)-IP“pir8^4-f4i3-34trif3«OF-m^d^bsn2öoiÄ; 3- (4-( í iI-pin:okt|24-b|p:iri<;b(i-4-i])-iri--pfi'a^<il·· i -11 j-4 (Srifeor-fr.eíil i-beazmnd ; 2-ρ-(111-ρί:ηίο1ορ3-^ρ3ρί61ο-4~113-134>-ρ3ο^1·-:1-llf-eíítlobe^asöl;: 2^4-(ili>fiRef6f2,3MI^Mdfe-^8|4iE-pteiàP4l3^Âkpxà^fe»teWî; 4- (1 -(2-(iriÔaBO-oïstlli-l®Ml]- 1 B-forazoM-i) -lM-pltTOÍoP3-b|psridlö:; (4-{(4-(St!--piro4o|2:3-b|p3ridiii-4-ilpil4-pÍFaz(tf-l-ijj-niet!Í}-cik;ohex!l)-53it4oool; 4414tebrahídn>í\irsiS3-2-il-roed]}-· 1 !.Bp]ra?ol-4-il|·· 3 H-pirr<ibP,3-b]-pmdi?í; 4-| b{ b<db^sPd-|)o^ll}-4 H-plmK0]-4-41-! P-ptebf23:-b]plrid:]bi 2- kför^44'nH-pírí-í>lo[2,í-b)p!íidm-4-il)· 1 f-I-ptra^ol-14í]-btíi«»fHtriU :34^i-i.Îisb>-rxîl<){2(3-b|pind'm-4-ïl)-HÎ-pirazo!-î-il]»3-{? J-bszol-S-ilvpriïpàpskri!; :3<;l~ftusl-í H- irakla7x>l44I}^4<iH^jiTolop.3-b]p»idin-4~a hUi-piya^^l^ii]-prppfeiST|;: 3,|4,;;}|.ρ;πΌΐο|2,3-Η]ρ;ΓΜΙκ-4-ίΐ)-ΙΒ'ρΐΓ3ίθ1-1-ΐ1]-3-(3-ΐί£Γ:ίΙ}-ρΐ·0ράβΗ5ί:ΓΪ1; f 1 -[4-( I H-p-irnïtop J~b]ps:íÉíi8-4--tí)-íí i-pbazo!-14§^ί6ρέ8ΐίΙ} -acskdßM; 4- kJ0r-344-0 H-p:«:K4<:<f2,<-b|pmdi»-4-ii>· f R-pirazol- i ~H}-ben:sonkrik 4~|4;^í 5- metiM-^pll^fek^^SdjlpiMsn-d^lH&amp;piisæbi-îO-bea^MÂd:; 4-(4 -£2-í«s£k4-o«rö~feií S)-3 H-pirasa>i-4~5«3··1M-pi rrc<lo[2 3--b|píf idœ; 3- f44i H~piiroΫt2,3-l>|fpîrî<]m^4-Îâ>~ ï i^ks^bl-ill-ciktepeïttâîïôs; 4- ( ä 43-ΐ\ίΠΐ-?Λ5·«ί3ίΐ-ί íí-píra>:oM~dj-1 H-píFT0fóf2J~b’jpLbdin; 4-Íi-{2-&amp;r5l^®íj^-IB-pímöb-4-ü|-lM-píffídí|234^pK5diöi 3- f 2-c «mo- ! -|44144-p5írP?pp,3-bJpííMkí-4-}:í3- ί: H-psraxcd-í -Mj-gdî4-hfi8Zôïdt dk {3 nu;iii-4-?4-( nbpirroÍ0[2)3"blpirífíHV-4-d}·'í Ibp«azoi-- í-i!|-i«s):Ü)-metaíioI; 3-( ] -bensö^^-2-íl4-344-( I i-iK-praf átóíd| 3- (3-:ásri!}-3 -|4-<1 FÍ-psn'ölc4'2.3-b|pirídf:rí-4~ií)-IH-p4^azöI-l-ii|-propás0Íb;i:; f3-!sedi-4444^^5sm4<d2,344psidiJ5:“4-tä:|-iH~pir8^-14j|-Äi3b-^ÄrdtFi; 4· f-íetil-3 -{4 •(TH'-plmdop.j · d)pirlmi(Kn-4-t! ,i-1H-pirazc*Í~ I -Uj-feerreofmríÉ; 4- [ i-( i-dklopeiPü-propíO-1 H-p!mzol-4-Hj-7H-pinroío[2,3-d]p5ritnídm; i ;444^^pí^'í>^2v>-dspií?midb}-4-ír!-!H-p3'3^ol-l"íl|-cÍKí!>5í<:í)tíl}~3Cvíkmlírih 3- ;2<iaíK5-i~[4-(7H-pliTOÍo|2.3-d jphiîrskiÎH-4-iî}- i H-pirazoi- I -d]-eíü} 4x-^z>.>min.i; 3- Γ4-! 71 í “pssTO 1 ű| 2,3-djpiruík á i v4-d V i H-pirazoí-l -íij-3-{3-lfóTis{}*propánnidsU 4- kk)r-3-!447H-pirroS0[2f3-djpírhrr:din-4-ii)-1H'piraíöi-l-i]|-bes^.omtnS:; 3-ü~tdriI)-3~[4~(7H-plrrolö(2 J-dipirímidia-d-ilMB-pirazol-í-dj-propánminl; 3. i.|a47H-píxröÍD[2b3-djpirí}'5íidbi-4"i^}4H-pÍ!'a.i'í)i- :-íl]-<IkiopeiníSJ -propdAíJSiriii;: {\ -{4-i 7Ï bp;rwloí2J-d]p!rmddin-4-ií}-1 Η-ρ?τ&amp;&amp;ΐΙ~ϊ -H j-ciklohcxü] -sc«îoni5rîS; >3-meul-i44-vH-pkToio|2,3-d]pirirekbn-4-äi M H-pirazol-i 41]-!taii}-nae(«iti&amp; 3-p[ddh)'4-ii- >-[44 71 {-p«to]o[2,3-d3pjrimidín-4-ín- ) S í-pirscKd-1 -il l-propatpitrii; 3"piridÍ3x-3-ií-3-[4-(7i i-pirrok)[23-d|pdln;!dúí-44i>l H-pirazííl-ä-sl j-pföpátólFik 3~[4-(metíí-tro>-f«iii3*3-(4-<7H-p<rro1«P<3-d]pa4mldm-4-HVlH'pá«2oM;‘'ií|-própfejBÍ^Í. 3“(3“íxísíPxXí -Ibix i:S) ·3 -(|44:7Η-ρΰτοΙρ|2 j-dlplrimídm-d-í ÍJ-1 il-ppaíök l-ä3|rp-opanxmdl; í 3-fíí«íi*4-|4-(7M“p3«öl^23”á3pMtód3p“4“ílk IR-pirazol- 1 ”iI3-fcil}«efötxMI; 3-[4-(mePkszi;lnn!!)-f<;ai!j-3-[447H-p!rroío|2,3'd]p4Í!«!dÍF-4~ílj-iil-pirá5s>i~!-íI]-propáiií'.iö'i:>; 3^4dSítóks»4^&amp;p®-%plik3-|4-{7l3"ptalof23 ·43ρ3ρβ»30ΐρ-4-1|-1] -dl-pfopáppirí I; 3-[3'(cíapo-t3^etpxd-fe^il|-3~[4'(?H-pín'olo|2J3-d|pí:rímids53~4-d}-IH~p3ra2(3Í-í-iI]-p!'0psí3íi3tr;d; 5- 42^ciar}(>-l-[4-{?y-psTOíof2J-d3pMmidm^-ij-4&amp;pM^}y~j:l|-ed!|-pkidin-2-kaifee8Stól; 3 -í JjS-diîaedl-âîxJsa^d^-ljj-l-p^^^àTfigop^-dlpsriPï&amp;srd-â^-l 14-pF33sd-1 -d|-prőpfeadrd; 3-Ρ-{?Η-ρί(το]οί ? 3-d)pírím«d}n-4-ii)- i 3.^ίδΗ»εΐθλΐ-»ιΗΛη-3-Η)ο-[4^71·ί^πΌΪο{2,3^]ρ}Πίη^?η^ιΐν?Η>'0ρ»ΐ-.Ι4?|.-ρ·«^Ββ|Λΐ :J-|»#«P-->ii“344^7H-pKTolo[2,3-djpir5midin~-i-H)-IH-p>ra?ol-J*H]''P^P^iK^U ;3^^05η^ιπί!ΐη»2-Ιΐν344Η7Η-ρ{ΓΤ<»Ιορ.3^]ρΐ{«ηΐ0κί-4->Ί)-1ί{-ρ«·«2θ}·Ι·{Π“ΡΡορ«ί*0ϊίΗ| áhf2-da»o-1 *(4-{7H-pjjTok>p, J-dJpírhdd m-4-iiíh i H-piræad· î U -p*Trtdi»-2-katííe®íö?IÍJ 3-(5->ÍS'óir:"piridiíi- 3-ii}-3 ~(4~f 7 H-pirroloj 2,3-d}pmsnidin-4-H>· 1 s 1-pân.uol· 1 ~d|-pFppâsd»$; iS- p*ékdô-1 Í 43 J-et si} -o^oîtaîïfei: ;^[4-(ckÄK)-ffieiöx5}-:feni|k3^4K7!-I-pirrokv[2;3-dlpirimkiiis-4-v|}-n-!-pirazc4-i~i!]-pröpä®i5;tri;; 3-P^!anö-meí0d>^ní]j-3-|447H-pjrm!oP,3-á;|pkímíd:i^4-í^rH-pfeazo!4~M|^ft^áíaí4d]:; 3-|;S41t£isepka^3Sö}-pá4dfe-2-:i?]!-3~p^'7Prpírrölöp3-d]p4s83:y^4^d)-4;M-p3raK0kl-:i3|-píopápsiMI; 5- l -|44'7H-pí:s'olo|2,3-dlp3?'s^idiirí^-?l)^l34-píra2ú;!-i 4]|-ei3} -d^fiäa^S-karböaätdl; S^d-d^s-Äd}·##-(7!Í-pirí'G4öP .,3 “d3pk»iÄ5~44|)~ UI-plœol-λii|<pr<^^dpikîl; 1- p-dano- ! -{4-(7Η-ρ«τοΐ0{ 2P~d'jpirimídit-~441)- i I l-pirazol- \ -il ;-etli}~4-rodoxiba5«>sd&amp;i;: 3-(3-brô{n-rei«iV3-Î4-(7H-pâiToîoP,3-ti]pirimidin-4-ii}-3ii-pirazo!-i-il}-pfopàniJrtrâl; 3-52-ciano-1 -{44?H-pirroksp23-d]p3ri5nidin-44iH H-pírazol-l 4 VJ -ets s} -4-4 i-or-bena« sisri f; 3-P43íx>n-'24cia30-metoxi)'íe33Íi|-34447H^irro]<í[2,3-#ptí:mfdWd:|-lM3píía2öÍÍ-í''Í||-propá!3mtn]:; 344-bmm~p!fi3i;a-2-3}'3-[<|-(7l3-pirrí>k>|2,3'(l|pi!'ín3ÍdÍ8~4'!Í}-S B-psraxd-l-iO-píopámPdl; 2- k2<33n0vk[4d'7H-pirrok!{23-d|pirm>sdißd-i!VlH-pira2cd-!4]J<dii}-köi;dkofeön:äft'sl; 5d:2-edno-4-l447H^lrrsfepp^pinmy } B4>Brasöl-4:4l|-etíl|-·3"&amp;®)Μ;; 3'[2-0Γ0!ΐΐ-$~(€ί3ί3ο-πί«ιοχί}-ί3Γ3Ι1^-|447Η-ρίΓτο!0ί23'·4|ρ3Γ58ΐ!<ΐίη-4-!ΐ)-1Ι-1"ρίΓ«χ;οΐ-Ι-·5!]-ρΚ!ρίΐηβϊΐΓί1; 4^Âs0Tis^ôsï^2^p^t-œ^i4447^-p^ô1or^3«à|piôm^feHÎ^I)ri;H^«2ôy>Si^!|4i^£îî^æsU 3^irmsMtï3-S-îÎd3>-H-ï 7^ί -îslrmÎ0|2 ï ί ΐ-ρ tra^cïl- r-il]-|y^äftöät^ü •k(.: bróí3)-p:n<.{)Xi ·4·4)· > ·|4··(7Η-ρ0τοΙο[2.3-<:ί|ρ(ϊΐ)3··κΐ4ΐ··4·)ΐ)· lH- pirax:«l-5- H)-propikioikíS; *» l#fei^4-TÍí>- í M^f»ír»zoI-l -* 1 j- 3- 0 -r»*ioxî--pàkiîî>:-3-'di--3-| 4-(7 H-pki'ok43,3~d}pin;ddiii-4~i 1)-3 H-ph'a>'d~3 -d:]~pi'ôpàîïs4ïiü; l il-fsttszpí- l^il|-<propâjMîôFil; B-l^-íTH-pirroíoíSjS-dipínmídín-d-iíí'-í H-p5m;ii>l-l-ü.l-23-13'(ir{{:luor-mefy}--fen.y'|-'propäimätfi3; ;3HE3-fé8dx^É*KÎ^34^<7^w{àÂdP'J«4^ÂMIM4l>i4H-jpi^M4l|>prë§pmÂ% 3~|3î ^ ; 3 -^1 Jp iîi sí Mïsr^-àl>-4 ï -iî]-J Hp -<i ïiïilMor-Tîï <;îos .(>-l^34|-^öp^^:ítri 1 ί 3-p-cïa0e-ï-|4--(7l3-piisrof0[23-dÿjFimy30^d3-i33-piïd2;0l-i-ïi|-«ts(j-&amp;eiix®ssav:: 3-(34314-^íí^XföM-i!Vfens|^3-p^7;íí.piyFö|öp3-djpkimy(ö-44l3-:lH-piíSZö3-Í33|'psípátinMl; 1*1#' iS^sN^' 1 #-( W-psproifop ,|-d|py mPîsi-44 i}~ ï M-pirszföl-1 -Ί1|-«ΐϋ| -fesilJiiieiäaszulÄiÄUid; 5* > 3-ciiiîïô·· 1IH-phm»)·- Ν-(2·*!Βο·«ίΐΡ· I H^4m>y 4i^l 3-<5-ife^EÔ-3[-tÊa*£ii>-3-|4-*{7l-î*phErt>lc^2>3-ü ipirimídm-^-i] >~Oi~ptra^ol- Î-il}~»>ropáafí «ríl; î-|>e»&amp;-144#ïri-pàTôloï2,3-djpîrimi<lin~l>il)-1 H-ptra/oi- í -Hj-etíl) N'-metU-benzarnid; 2^cjasö-Hn<:l'i2-ciia:aö-]^"(?^b-pírroUíP J-d]pïriîrsS<SÎTi-4-;i)-3H"pirazol~î~il]-<îtn}-fenii McetaiSÎâ; N-^3-|2 ^íaíí©-1-P-(TH-pírroio[2f3-djplnmidin-4'-il}-ÍH-píra?cíí-í'ilj-etü )~fe«i3}-mkon88ip:|d; H^'3"{2'eias©~l-|4“(7il-pjrroio[2,3~djpirimídí;>4~íi)-1H"píra;Kd-^ü|-edl}-fcnii)"N'd?Á>p5'op3ll--ltafÍm5d; feopropii (M2-ciano- i - (4 -* 7H-púro}af2 J-d jpirús i-jin4-H)- î ï l-pira-ioM-iiHtjH-fenüVkadbïa^p 3'4i5-iefi!l-piridin-3“i!>-3-|-}'(7H-pitro!i>(2i.3-djp5rif!)idin-4-il)-i B-pir&amp;«>i“MÍ|~propára4Snl; ;3-(3,3‘-bipiridnf-S-í!V j"i-í-t'?H-piíTokip,3--dijpíxitnídirí-4-íí)-íH-pífa20Í-Í-ilj-í«f0p>ánnitrü; ;^i5-pärimidiß-54j-piridin-3*il>-3#^7H-pirro!<^2,3‘>djpinm5dfö“4''n}»Ui-pjt«aol'i-t})-pfpjpteM% ;3#4 l-metil-ï 1 J-pjra2oM-ííVpiridin45-ilj-3-[4-í7H«p»m>löP43-d}pÍrúnidiíx-4··}!)·· ! H -praz©l~I-t|-prppäöriiiöl; 325^pM3-pikidln4-d|:2i-f427l-l"pfe^dii|2:3'dlpÄ:idmP-'iii)~ IB^iraxö^l^ll-propäaminl; 3454Í8ail4:iiií3-pi?ídifi^3-:il|'3#27:H-pkroloP:>3~d|pa4;KÍd|®-4-i}-lB-piía2©l“l:40"p^p4íJfií^s!;
  4. 4). Az i-ΓΙ, igénypontok bánneiylfceszerinti: vegyiléf vagy só alkálmagásra, álmí á továbbilyégyászati, szer proteoszóms inhibitor. ixmezoíoib, riilklothith: Bez-Ahl lohlöhöh, filial inhibbpr, LÁk inhibitor vagy |Aik inhibitor.
HUE11152677A 2005-12-13 2006-12-12 Heteroaril-szubsztituált pirrolo[2,3-b]piridinek és pirrolo[2,3-b]pirimidinek mint Janus-kináz inhibitorok HUE028588T2 (hu)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US74990505P 2005-12-13 2005-12-13
US81023106P 2006-06-02 2006-06-02
US85062506P 2006-10-10 2006-10-10
US85687206P 2006-11-03 2006-11-03
US85940406P 2006-11-16 2006-11-16

Publications (1)

Publication Number Publication Date
HUE028588T2 true HUE028588T2 (hu) 2016-12-28

Family

ID=37903501

Family Applications (7)

Application Number Title Priority Date Filing Date
HUE11152677A HUE028588T2 (hu) 2005-12-13 2006-12-12 Heteroaril-szubsztituált pirrolo[2,3-b]piridinek és pirrolo[2,3-b]pirimidinek mint Janus-kináz inhibitorok
HUE11152730A HUE030418T2 (hu) 2005-12-13 2006-12-12 Heteroaril-szubsztituált pirrolo[2,3-b]piridinek és pirrolo[2,3-b]pirimidinek mint Janus-kináz inhibitorok
HUE11152708A HUE025173T2 (hu) 2005-12-13 2006-12-12 Heteroaril-szubsztituált pirrollo[2,3-b]piridinek és pirrolo[2,3-b]pirimidinek mint Janus-kináz inhibitorok
HUE16197502A HUE041382T2 (hu) 2005-12-13 2006-12-12 Pirrolo[2,3-d]pirimidin származékok janus-kináz inhibitorként
HUE11152714A HUE032337T2 (hu) 2005-12-13 2006-12-12 Heteroaril-szubsztituált pirrolo[2,3-b]piridinek és pirrolo[2,3-b]pirimidinek mint Janus-kináz inhibitorok
HUE11152723A HUE030235T2 (hu) 2005-12-13 2006-12-12 Heteroaril-szubsztituált pirrolo[2,3-b]piridinek és pirrolo[2,3-b]pirimidinek mint Janus-kináz inhibitorok
HUS1700017C HUS1700017I1 (hu) 2005-12-13 2017-04-25 Heteroaril-szubsztituált pirrolo[2,3-b]piridinek és pirrolo[2,3-b]pirimidinek mint Janus-kináz inhibitorok

Family Applications After (6)

Application Number Title Priority Date Filing Date
HUE11152730A HUE030418T2 (hu) 2005-12-13 2006-12-12 Heteroaril-szubsztituált pirrolo[2,3-b]piridinek és pirrolo[2,3-b]pirimidinek mint Janus-kináz inhibitorok
HUE11152708A HUE025173T2 (hu) 2005-12-13 2006-12-12 Heteroaril-szubsztituált pirrollo[2,3-b]piridinek és pirrolo[2,3-b]pirimidinek mint Janus-kináz inhibitorok
HUE16197502A HUE041382T2 (hu) 2005-12-13 2006-12-12 Pirrolo[2,3-d]pirimidin származékok janus-kináz inhibitorként
HUE11152714A HUE032337T2 (hu) 2005-12-13 2006-12-12 Heteroaril-szubsztituált pirrolo[2,3-b]piridinek és pirrolo[2,3-b]pirimidinek mint Janus-kináz inhibitorok
HUE11152723A HUE030235T2 (hu) 2005-12-13 2006-12-12 Heteroaril-szubsztituált pirrolo[2,3-b]piridinek és pirrolo[2,3-b]pirimidinek mint Janus-kináz inhibitorok
HUS1700017C HUS1700017I1 (hu) 2005-12-13 2017-04-25 Heteroaril-szubsztituált pirrolo[2,3-b]piridinek és pirrolo[2,3-b]pirimidinek mint Janus-kináz inhibitorok

Country Status (36)

Country Link
US (16) US7598257B2 (hu)
EP (10) EP3838903B1 (hu)
JP (4) JP5017278B2 (hu)
KR (4) KR101218214B1 (hu)
CN (4) CN103214484B (hu)
AR (1) AR057995A1 (hu)
AT (1) ATE525374T1 (hu)
AU (1) AU2006326548B2 (hu)
BR (1) BRPI0619817B8 (hu)
CA (1) CA2632466C (hu)
CR (2) CR10065A (hu)
CY (8) CY1112762T1 (hu)
DK (7) DK2474545T3 (hu)
EA (3) EA019504B1 (hu)
EC (2) ECSP088540A (hu)
ES (9) ES2373688T3 (hu)
FR (1) FR17C1013I2 (hu)
HK (5) HK1124840A1 (hu)
HR (7) HRP20110903T1 (hu)
HU (7) HUE028588T2 (hu)
IL (3) IL192019A (hu)
LT (5) LT2455382T (hu)
LU (1) LU92137I2 (hu)
ME (1) ME01312B (hu)
MX (1) MX346183B (hu)
MY (2) MY162590A (hu)
NZ (2) NZ569015A (hu)
PL (7) PL3184526T3 (hu)
PT (7) PT2474545T (hu)
RS (7) RS58113B1 (hu)
SG (3) SG179430A1 (hu)
SI (7) SI2426129T1 (hu)
TW (6) TWI664182B (hu)
UA (2) UA116187C2 (hu)
WO (1) WO2007070514A1 (hu)
ZA (1) ZA200805165B (hu)

Families Citing this family (372)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060106020A1 (en) * 2004-04-28 2006-05-18 Rodgers James D Tetracyclic inhibitors of Janus kinases
AR054416A1 (es) 2004-12-22 2007-06-27 Incyte Corp Pirrolo [2,3-b]piridin-4-il-aminas y pirrolo [2,3-b]pirimidin-4-il-aminas como inhibidores de las quinasas janus. composiciones farmaceuticas.
JP2009508832A (ja) * 2005-09-16 2009-03-05 アストラゼネカ アクチボラグ グルコキナーゼ活性化剤としてのヘテロ二環式化合物
EP1926735A1 (en) 2005-09-22 2008-06-04 Incyte Corporation Tetracyclic inhibitors of janus kinases
DK1951684T3 (en) * 2005-11-01 2016-10-24 Targegen Inc BIARYLMETAPYRIMIDIN kinase inhibitors
US8133900B2 (en) * 2005-11-01 2012-03-13 Targegen, Inc. Use of bi-aryl meta-pyrimidine inhibitors of kinases
US8604042B2 (en) * 2005-11-01 2013-12-10 Targegen, Inc. Bi-aryl meta-pyrimidine inhibitors of kinases
MY162590A (en) 2005-12-13 2017-06-30 Incyte Holdings Corp Heteroaryl substituted pyrrolo[2,3-b] pyridines and pyrrolo[2,3-b] pyrimidines as janus kinase inhibitors
KR20080083680A (ko) * 2005-12-23 2008-09-18 스미스클라인 비참 코포레이션 오로라 키나제의 아자인돌 억제제
WO2007116866A1 (ja) * 2006-04-03 2007-10-18 Astellas Pharma Inc. ヘテロ化合物
MX2008012860A (es) * 2006-04-05 2009-01-07 Vertex Pharma Desazapurinas de utilidad como inhibidores de janus cinasas.
US20100099772A1 (en) 2006-11-20 2010-04-22 Bean Bruce P Methods, compositions, and kits for treating pain and pruritis
US8513270B2 (en) * 2006-12-22 2013-08-20 Incyte Corporation Substituted heterocycles as Janus kinase inhibitors
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 (es) * 2007-06-13 2008-11-03 Incyte Corp Compuestos derivados de pirrolo [2,3-b]pirimidina, moduladores de quinasas jak; composicion farmaceutica; y uso en el tratamiento de enfermedades tales como cancer, psoriasis, artritis reumatoide, entre otras.
WO2009003998A2 (en) * 2007-07-02 2009-01-08 Boehringer Ingelheim International Gmbh Antiproliferative compounds based on 5-membered heterocycles
WO2009032338A1 (en) * 2007-09-09 2009-03-12 University Of Florida Research Foundation Apratoxin therapeutic agents: mechanism and methods of treatment
WO2009049028A1 (en) * 2007-10-09 2009-04-16 Targegen Inc. Pyrrolopyrimidine compounds and their use as janus kinase modulators
WO2009054941A1 (en) * 2007-10-25 2009-04-30 Merck & Co., Inc. Therapeutic compounds
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
CA2714177A1 (en) * 2008-02-06 2009-08-13 Novartis Ag Pyrrolo [2,3-d] pyridines and use thereof as tyrosine kinase inhibitors
US20110105436A1 (en) * 2008-03-10 2011-05-05 Auckland Uniservices Limited Heteroaryl compounds, compositions, and methods of use in cancer treatment
SI2288610T1 (sl) * 2008-03-11 2016-11-30 Incyte Holdings Corporation Derivati azetidina in ciklobutana kot inhibitorji jak
EP2278969B1 (en) * 2008-04-21 2013-02-13 Merck Sharp & Dohme Corp. Inhibitors of Janus kinases
CA2722326A1 (en) * 2008-04-24 2009-10-29 Incyte Corporation Macrocyclic compounds and their use as kinase inhibitors
CL2009001152A1 (es) 2008-05-13 2009-10-16 Array Biopharma Inc Compuestos derivados de n-(4-(cicloalquilo nitrogenado-1-il)-1h-pirrolo[2,3-b]piridin-3-il)amida, inhibidores de cinasa; proceso de preparacion; composicion farmaceutica; y su uso para el tratamiento de una enfermedad proliferativa.
US8344144B2 (en) * 2008-06-18 2013-01-01 Merck Sharp & Dohme Corp. Inhibitors of Janus kinases
RU2561104C2 (ru) * 2008-06-20 2015-08-20 Дженентек, Инк. Триазолопиридиновые соединения-ингибиторы jak и способы
RU2560153C2 (ru) * 2008-06-20 2015-08-20 Дженентек, Инк. Триазолпиридиновые соединения, ингибирующие jak, и способы
CL2009001884A1 (es) * 2008-10-02 2010-05-14 Incyte Holdings Corp Uso de 3-ciclopentil-3-[4-(7h-pirrolo[2,3-d]pirimidin-4-il)-1h-pirazol-1-il)propanonitrilo, inhibidor de janus quinasa, y uso de una composición que lo comprende para el tratamiento del ojo seco.
JOP20190230A1 (ar) * 2009-01-15 2017-06-16 Incyte Corp طرق لاصلاح مثبطات انزيم jak و المركبات الوسيطة المتعلقة به
WO2010085597A1 (en) * 2009-01-23 2010-07-29 Incyte Corporation Macrocyclic compounds and their use as kinase inhibitors
MX2011012262A (es) 2009-05-22 2012-01-25 Incyte Corp 3-[4-(7h-pirrolo[2,3-d]pirimidin-4-il)-1h-pirazol-1-il] octano-o heptano-nitrilo como inhibidores de cinasas janus (jak).
EA025520B1 (ru) * 2009-05-22 2017-01-30 Инсайт Холдингс Корпорейшн N-(ГЕТЕРО)АРИЛПИРРОЛИДИНОВЫЕ ПРОИЗВОДНЫЕ ПИРАЗОЛ-4-ИЛ-ПИРРОЛО[2,3-d]ПИРИМИДИНОВ И ПИРРОЛ-3-ИЛ-ПИРРОЛО[2,3-d]ПИРИМИДИНОВ В КАЧЕСТВЕ ИНГИБИТОРОВ ЯНУС-КИНАЗЫ
ES2540964T3 (es) 2009-06-08 2015-07-15 Takeda Pharmaceutical Company Limited Compuestos de dihidropirrolonaftiridinona como inhibidores de JAK
AR077280A1 (es) 2009-06-29 2011-08-17 Incyte Corp Pirimidinonas como inhibidores de pi3k, y composiciones farmaceuticas que los comprenden
EP3485881B1 (en) 2009-07-10 2024-03-13 President and Fellows of Harvard College Permanently charged sodium and calcium channel blockers as anti-inflammatory agents
TWI466885B (zh) 2009-07-31 2015-01-01 Japan Tobacco Inc 含氮螺環化合物及其醫藥用途
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
US9029359B2 (en) 2009-09-04 2015-05-12 Biogen Idec Ma, Inc. Heteroaryl Btk inhibitors
JP5658756B2 (ja) 2009-09-10 2015-01-28 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft Jakの阻害剤
KR101907535B1 (ko) 2009-10-02 2018-10-15 아벡신 에이에스 소염제 2-옥소티아졸 및 2-옥소옥사졸
PT2486041E (pt) 2009-10-09 2013-11-14 Incyte Corp Derivados hidroxilo, ceto e glucuronido de 3-(4-(7h-pirrolo[2,3-d]pirimidin-4-il)-1h-pirazol-1-il)-3-ciclopentil-propanonitrilo
US8389728B2 (en) * 2009-11-06 2013-03-05 The Arizona Board Of Regents Pollen tube stimulants from Arabidopsis pistils
EA023444B1 (ru) * 2010-02-18 2016-06-30 Инсайт Холдингс Корпорейшн Циклобутановые и метилциклобутановые производные, композиции на их основе и способы их применения
WO2011109217A2 (en) * 2010-03-02 2011-09-09 Immunodiagnostics, Inc. Methods of treating or preventing rna polymerase dependent viral disorders by administration of jak2 kinase inhibitors
SI3354652T1 (sl) * 2010-03-10 2020-08-31 Incyte Holdings Corporation Derivati piperidin-4-il azetidina kot inhibitorji JAK1
AU2015205858B2 (en) * 2010-03-10 2017-04-13 Incyte Holdings Corporation Piperidin-4-yl azetidine derivatives as jak1 inhibitors
KR20130094710A (ko) * 2010-04-14 2013-08-26 어레이 바이오파마 인크. Jak 키나아제의 억제제로서 5,7-치환된-이미다조[1,2-c]피리미딘
US9133123B2 (en) 2010-04-23 2015-09-15 Cytokinetics, Inc. Certain amino-pyridines and amino-triazines, compositions thereof, and methods for their use
AR081626A1 (es) 2010-04-23 2012-10-10 Cytokinetics Inc Compuestos amino-piridazinicos, composiciones farmaceuticas que los contienen y uso de los mismos para tratar trastornos musculares cardiacos y esqueleticos
AR081331A1 (es) 2010-04-23 2012-08-08 Cytokinetics Inc Amino- pirimidinas composiciones de las mismas y metodos para el uso de los mismos
SG10201910912TA (en) * 2010-05-21 2020-01-30 Incyte Corp Topical Formulation for a JAK Inhibitor
SG187742A1 (en) * 2010-08-20 2013-03-28 Hutchison Medipharma Ltd Pyrrolopyrimidine compounds and uses thereof
WO2012040527A2 (en) * 2010-09-24 2012-03-29 The Regents Of The University Of Michigan Deubiquitinase inhibitors and methods for use of the same
CA2816088A1 (en) 2010-10-28 2012-05-03 Viamet Pharmaceuticals, Inc. Metalloenzyme inhibitor compounds
WO2012060847A1 (en) 2010-11-07 2012-05-10 Targegen, Inc. Compositions and methods for treating myelofibrosis
BR112013012502A2 (pt) * 2010-11-19 2019-03-06 Incyte Corporation pirrolopiridina ciclobutil substituída e derivados de pirrolopirimidina derivativos como inibidores de jak
EA036970B1 (ru) * 2010-11-19 2021-01-21 Инсайт Холдингс Корпорейшн Применение {1-{1-[3-фтор-2-(трифтометил)изоникотиноил] пиперидин-4-ил}-3-[4-(7h-пирроло[2,3-d]пиримидин-4-ил)-1н-пиразол-1-ил]азетидин-3-ил}ацетонитрила для лечения заболеваний, связанных с активностью jak1
US9034884B2 (en) 2010-11-19 2015-05-19 Incyte Corporation Heterocyclic-substituted pyrrolopyridines and pyrrolopyrimidines as JAK inhibitors
EP2651930B1 (en) * 2010-12-16 2015-10-28 Boehringer Ingelheim International GmbH Biarylamide inhibitors of leukotriene production
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
WO2012116151A2 (en) * 2011-02-24 2012-08-30 Cephalon, Inc. Substituted aromatic sulfur compounds and methods of their use
AU2012232658B2 (en) 2011-03-22 2016-06-09 Advinus Therapeutics Limited Substituted fused tricyclic compounds, compositions and medicinal applications thereof
US8759380B2 (en) 2011-04-22 2014-06-24 Cytokinetics, Inc. Certain heterocycles, compositions thereof, and methods for their use
EP2710006A1 (en) * 2011-05-17 2014-03-26 Principia Biopharma Inc. Azaindole derivatives as tyrosine kinase inhibitors
MX344580B (es) 2011-06-14 2016-12-20 Novartis Ag Combinacion de panobinostat y ruxolitinib en el tratamiento del cancer tal como una neoplasia mieloproliferativa.
MY165963A (en) 2011-06-20 2018-05-18 Incyte Holdings Corp Azetidinyl phenyl, pyridyl or pyrazinyl carboxamide derivatives as jak inhibitors
CN103732596B (zh) 2011-07-08 2016-06-01 诺华股份有限公司 吡咯并嘧啶衍生物
EP2741747A1 (en) 2011-08-10 2014-06-18 Novartis Pharma AG JAK P13K/mTOR COMBINATION THERAPY
TW201313721A (zh) 2011-08-18 2013-04-01 Incyte Corp 作為jak抑制劑之環己基氮雜環丁烷衍生物
SI2751109T1 (sl) 2011-09-02 2017-03-31 Incyte Holdings Corporation Heterociklilamini kot inhibitorji pi3k
UA111854C2 (uk) 2011-09-07 2016-06-24 Інсайт Холдінгс Корпорейшн Способи і проміжні сполуки для отримання інгібіторів jak
WO2013041042A1 (en) 2011-09-22 2013-03-28 Merck Sharp & Dohme Corp. Pyrazole carboxamides as janus kinase inhibitors
KR20140076619A (ko) * 2011-10-12 2014-06-20 어레이 바이오파마 인크. 5,7-치환된-이미다조[1,2-c]피리미딘
US10821111B2 (en) 2011-11-30 2020-11-03 Emory University Antiviral JAK inhibitors useful in treating or preventing retroviral and other viral infections
US9089574B2 (en) 2011-11-30 2015-07-28 Emory University Antiviral JAK inhibitors useful in treating or preventing retroviral and other viral infections
US8993756B2 (en) * 2011-12-06 2015-03-31 Merck Sharp & Dohme Corp. Pyrrolopyrimidines as janus kinase inhibitors
AR090548A1 (es) 2012-04-02 2014-11-19 Incyte Corp Azaheterociclobencilaminas biciclicas como inhibidores de pi3k
US20130310340A1 (en) 2012-05-16 2013-11-21 Rigel Pharmaceuticals, Inc. Method of treating muscular degradation
US9193733B2 (en) 2012-05-18 2015-11-24 Incyte Holdings Corporation Piperidinylcyclobutyl substituted pyrrolopyridine and pyrrolopyrimidine derivatives as JAK inhibitors
EP3450434B1 (en) * 2012-06-15 2021-02-24 CoNCERT Pharmaceuticals, Inc. Deuterated derivatives of ruxolitinib
US20150197525A1 (en) 2012-06-15 2015-07-16 Concert Pharmaceuticals, Inc. Deuterated derivatives of ruxolitinib
CN104797267A (zh) 2012-06-26 2015-07-22 德玛医药 使用卫康醇、二乙酰二脱水卫矛醇、二溴卫矛醇或类似物或其衍生物治疗具有基因多型性或ahi1失调或突变患者的抗酪氨酸激酶抑制剂的恶性肿瘤的方法
EA201590272A1 (ru) * 2012-07-27 2015-05-29 Рациофарм Гмбх Пероральные дозированные формы для модифицированного высвобождения, содержащие руксолитиниб
JP6276762B2 (ja) 2012-08-02 2018-02-07 ネルビアーノ・メデイカル・サイエンシーズ・エツセ・エルレ・エルレ キナーゼ阻害剤として活性な置換ピロール類
JP2015529242A (ja) 2012-09-21 2015-10-05 アドヴィヌス セラピューティクス リミテッドAdvinus Therapeutics Limited 置換された縮合三環式化合物、組成物およびその医薬用途
US20150284394A1 (en) * 2012-10-26 2015-10-08 Hoffmann-La Roche Inc. Inhibitors of bruton's tyrosine kinase
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
US9310374B2 (en) 2012-11-16 2016-04-12 Redwood Bioscience, Inc. Hydrazinyl-indole compounds and methods for producing a conjugate
WO2014085154A1 (en) 2012-11-27 2014-06-05 Beth Israel Deaconess Medical Center, Inc. Methods for treating renal disease
NZ708392A (en) * 2012-12-06 2020-05-29 Baruch S Blumberg Inst Functionalized benzamide derivatives as antiviral agents against hbv infection
US9260426B2 (en) * 2012-12-14 2016-02-16 Arrien Pharmaceuticals Llc Substituted 1H-pyrrolo [2, 3-b] pyridine and 1H-pyrazolo [3, 4-b] pyridine derivatives as salt inducible kinase 2 (SIK2) inhibitors
WO2014118195A1 (en) 2013-01-29 2014-08-07 Avexxin As Antiinflammatory and antitumor 2-oxothiazoles and 2-oxothiophenes compounds
KR101828662B1 (ko) * 2013-02-12 2018-02-12 코니카 미놀타 가부시키가이샤 유기 일렉트로루미네센스 소자 및 조명 장치
EA030705B1 (ru) 2013-03-06 2018-09-28 Инсайт Холдингс Корпорейшн Способы и промежуточные соединения при получении ингибитора jak
US20140343034A1 (en) 2013-04-25 2014-11-20 Japan Tobacco Inc. Skin barrier function improving agent
PE20160126A1 (es) 2013-05-17 2016-02-24 Incyte Corp Derivados del bipirazol como inhibidores jak
ES2792549T3 (es) 2013-08-07 2020-11-11 Incyte Corp Formas de dosificación de liberación sostenida para un inhibidor de JAK1
CN105555313A (zh) 2013-08-20 2016-05-04 因赛特公司 在c-反应蛋白水平较高的实体肿瘤患者中的存活益处
EP3049442A4 (en) 2013-09-26 2017-06-28 Costim Pharmaceuticals Inc. Methods for treating hematologic cancers
WO2015054283A1 (en) * 2013-10-08 2015-04-16 Calcimedica, Inc. Compounds that modulate intracellular calcium
WO2015057695A1 (en) * 2013-10-15 2015-04-23 Bohan Jin Novel compositions, uses and methods for their preparation
US20160375024A1 (en) 2013-11-27 2016-12-29 Novartis Ag Combination therapy comprising an inhibitor of jak, cdk, and pim
SI3318565T1 (sl) 2013-12-05 2021-07-30 Pfizer Inc. Pirolo(2,3-D)pirimidinil, pirolo(2,3-B)pirazinil in pirolo(2,3-D)piridinil akrilamidi
JP6367545B2 (ja) * 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 ルキソリチニブの重水素化誘導体
KR102261733B1 (ko) * 2013-12-18 2021-06-04 콘서트 파마슈티컬즈, 인크. 룩소리티닙의 중수소화된 유도체
CN104725380B (zh) * 2013-12-18 2019-06-28 康塞特医药品有限公司 卢索替尼的氘代衍生物
JOP20200094A1 (ar) 2014-01-24 2017-06-16 Dana Farber Cancer Inst Inc جزيئات جسم مضاد لـ pd-1 واستخداماتها
JOP20200096A1 (ar) 2014-01-31 2017-06-16 Children’S Medical Center Corp جزيئات جسم مضاد لـ tim-3 واستخداماتها
CN106456773A (zh) 2014-02-28 2017-02-22 因赛特公司 用于治疗骨髓增生异常综合征的jak1抑制剂
CA2940666C (en) 2014-02-28 2022-08-23 Nimbus Lakshmi, Inc. Tyk2 inhibitors and uses thereof
CN106103484B (zh) 2014-03-14 2021-08-20 诺华股份有限公司 针对lag-3的抗体分子及其用途
KR20220066179A (ko) 2014-04-08 2022-05-23 인사이트 코포레이션 Jak 및 pi3k 억제제 조합에 의한 b-세포 악성종양의 치료
AU2015253192B2 (en) 2014-04-30 2019-05-16 Incyte Holdings Corporation Processes of preparing a JAK1 inhibitor and new forms thereto
RU2564891C1 (ru) * 2014-05-27 2015-10-10 Александр Александрович Кролевец Способ получения нанокапсул цитокининов
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
CN105218548A (zh) * 2014-06-09 2016-01-06 上海海和药物研究开发有限公司 一种新型杂环化合物及其制备方法和作为激酶抑制剂的用途
WO2015191677A1 (en) 2014-06-11 2015-12-17 Incyte Corporation Bicyclic heteroarylaminoalkyl phenyl derivatives as pi3k inhibitors
NZ629796A (en) * 2014-07-14 2015-12-24 Signal Pharm Llc Amorphous form of 4-((4-(cyclopentyloxy)-5-(2-methylbenzo[d]oxazol-6-yl)-7h-pyrrolo[2,3-d]pyrimidin-2-yl)amino)-3-methoxy-n-methylbenzamide, compositions thereof and methods of their use
US9623028B2 (en) * 2014-07-14 2017-04-18 Signal Pharmaceuticals, Llc Methods of treating a cancer using substituted pyrrolopyrimidine compounds, compositions thereof
GB201413695D0 (en) 2014-08-01 2014-09-17 Avexxin As Compound
TW201618773A (zh) 2014-08-11 2016-06-01 艾森塔製藥公司 Btk抑制劑、pi3k抑制劑、jak-2抑制劑、及/或cdk4/6抑制劑的治療組合物
ES2921875T3 (es) 2014-08-11 2022-09-01 Acerta Pharma Bv Combinaciones terapéuticas de un inhibidor BTK, un inhibidor PD-1 y/o un inhibidor PD-L1
HRP20211813T1 (hr) 2014-08-11 2022-03-04 Acerta Pharma B.V. Terapeutske kombinacije inhibitora btk i inhibitora bcl-2
US11311512B2 (en) 2014-08-12 2022-04-26 Monash University Lymph directing prodrugs
WO2016026974A1 (en) * 2014-08-21 2016-02-25 Ratiopharm Gmbh Oxalate salt of ruxolitinib
US9993551B2 (en) 2014-09-13 2018-06-12 Novartis Ag Combination therapies of EGFR inhibitors
CN105524067A (zh) * 2014-09-28 2016-04-27 江苏柯菲平医药股份有限公司 4-取代吡咯并[2,3-d]嘧啶化合物及其用途
KR20170066546A (ko) 2014-10-03 2017-06-14 노파르티스 아게 조합 요법
MA41044A (fr) 2014-10-08 2017-08-15 Novartis Ag Compositions et procédés d'utilisation pour une réponse immunitaire accrue et traitement contre le cancer
CU20170052A7 (es) 2014-10-14 2017-11-07 Dana Farber Cancer Inst Inc Moléculas de anticuerpo que se unen a pd-l1
WO2016063294A2 (en) * 2014-10-20 2016-04-28 Msn Laboratories Private Limited Process for the preparation of (r)-3-(4-(7h-pyrrolo[2,3-d], pyrimidin-4-yl)-1 h-pyrazol-1-yl)-3-cyclopentylpropanenitrile phosphate and its polymorphs thereof
GB2535427A (en) 2014-11-07 2016-08-24 Nicoventures Holdings Ltd Solution
CZ2014773A3 (cs) 2014-11-10 2016-05-18 Zentiva, K.S. Soli (3R)-3-cyklopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)pyrazol-1-yl]propannitrilu
CN105777754B (zh) * 2014-12-16 2019-07-26 北京赛林泰医药技术有限公司 吡咯并嘧啶化合物
CN117800973A (zh) 2015-02-27 2024-04-02 因赛特控股公司 Pi3k抑制剂的盐及其制备方法
TWI788655B (zh) 2015-02-27 2023-01-01 美商林伯士拉克許米公司 酪胺酸蛋白質激酶2(tyk2)抑制劑及其用途
MY190404A (en) 2015-03-10 2022-04-21 Aduro Biotech Inc Compositions and methods for activating "stimulator of interferon gene"-dependent signalling
KR101859170B1 (ko) * 2015-04-17 2018-05-17 광주과학기술원 트리아졸 화합물 및 이의 용도
JP6600365B2 (ja) * 2015-04-29 2019-10-30 无▲錫▼福祈制▲薬▼有限公司 Jak阻害剤
WO2016183060A1 (en) 2015-05-11 2016-11-17 Incyte Corporation Process for the synthesis of a phosphoinositide 3-kinase inhibitor
US9840503B2 (en) 2015-05-11 2017-12-12 Incyte Corporation Heterocyclic compounds and uses thereof
US9988401B2 (en) 2015-05-11 2018-06-05 Incyte Corporation Crystalline forms of a PI3K inhibitor
WO2017004134A1 (en) 2015-06-29 2017-01-05 Nimbus Iris, Inc. Irak inhibitors and uses thereof
CZ2015496A3 (cs) 2015-07-14 2017-01-25 Zentiva, K.S. Krystalické formy solí (3R)-3-cyklopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)pyrazol-1-yl]propannitrilu a jejich příprava
WO2017011720A1 (en) * 2015-07-16 2017-01-19 Signal Pharmaceuticals, Llc Solod forms 4-((4-(cyclopentyloxy)-5-(2-methylbenzo[d] oxazol-6-yl)17h-pyrrolo[2,3-d]pyrimidin-2-yl)amino)-3-methoxy-n-methylbenzamide, compositions thereof and methods of their use
SI3317301T1 (sl) 2015-07-29 2021-10-29 Novartis Ag Kombinirane terapije, ki obsegajo molekule protitelesa na LAG-3
EP3316902A1 (en) 2015-07-29 2018-05-09 Novartis AG Combination therapies comprising antibody molecules to tim-3
WO2017024037A1 (en) 2015-08-03 2017-02-09 President And Fellows Of Harvard College Charged ion channel blockers and methods for use
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
US10023571B2 (en) 2015-09-02 2018-07-17 Nimbus Lakshimi, Inc. TYK2 inhibitors and uses thereof
AU2016318229A1 (en) 2015-09-08 2018-03-29 Monash University Lymph directing prodrugs
US10683308B2 (en) 2015-09-11 2020-06-16 Navitor Pharmaceuticals, Inc. Rapamycin analogs and uses thereof
JP6800968B2 (ja) 2015-10-23 2020-12-16 ナビター ファーマシューティカルズ, インコーポレイテッド セストリン−gator2相互作用のモジュレーターおよびその使用
BR112018008904A2 (pt) 2015-11-03 2018-11-27 Janssen Biotech Inc anticorpos que se ligam especificamente a tim-3 e seus usos
TWI744256B (zh) 2015-11-06 2021-11-01 美商英塞特公司 作為PI3K-γ抑制劑之雜環化合物
RU2601410C1 (ru) * 2015-11-13 2016-11-10 ЗАО "Р-Фарм" {3-[(7H-ПИРРОЛО[2,3-d]ПИРИМИДИН-4-ИЛ)АЗОЛИЛ]АЗЕТИДИН-3-ИЛ}АЦЕТОНИТРИЛЫ В КАЧЕСТВЕ ИНГИБИТОРОВ ЯНУС КИНАЗ
EP3389664A4 (en) 2015-12-14 2020-01-08 Raze Therapeutics Inc. MTHFD2 CAFFEIN INHIBITORS AND USES THEREOF
CN108368115B (zh) * 2015-12-15 2020-01-03 正大天晴药业集团股份有限公司 吡咯并嘧啶化合物的盐
JP2019502695A (ja) 2015-12-17 2019-01-31 ノバルティス アーゲー PD−1に対する抗体分子とC−Met阻害剤との組合せおよびその使用
CA3007671A1 (en) 2015-12-17 2017-06-22 Novartis Ag Antibody molecules to pd-1 and uses thereof
US9630968B1 (en) 2015-12-23 2017-04-25 Arqule, Inc. Tetrahydropyranyl amino-pyrrolopyrimidinone and methods of use thereof
WO2017114461A1 (zh) * 2015-12-31 2017-07-06 正大天晴药业集团股份有限公司 一种芦可替尼的合成工艺
WO2017123741A1 (en) 2016-01-12 2017-07-20 Institute For Myeloma & Bone Cancer Research Improved methods for monitoring immune status of a subject
CZ201629A3 (cs) 2016-01-22 2017-08-02 Zentiva, K.S. Krystalické modifikace solí (3R)-3-cyklopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)pyrazol-1-yl]propannitrilu a způsoby jejich přípravy
JP6770580B2 (ja) * 2016-01-26 2020-10-14 杭州華東医薬集団生物医薬有限公司Hangzhou Huadong Medicine Group Biopharmaceutical Co., Ltd. ピロロピリミジン5員環アザ環状誘導体およびその利用
CN105541891B (zh) * 2016-02-04 2017-11-28 东南大学 巴瑞替尼的中间体及其制备方法及由该中间体制备巴瑞替尼的方法
PT3426243T (pt) 2016-03-09 2021-08-19 Raze Therapeutics Inc Inibidores de 3-fosfoglicerato desidrogenase e utilizações dos mesmos
US11014882B2 (en) 2016-03-09 2021-05-25 Raze Therapeutics, Inc. 3-phosphoglycerate dehydrogenase inhibitors and uses thereof
GB201604318D0 (en) 2016-03-14 2016-04-27 Avexxin As Combination therapy
CA3019145A1 (en) 2016-03-28 2017-10-05 Incyte Corporation Pyrrolotriazine compounds as tam inhibitors
JP2019510785A (ja) 2016-04-08 2019-04-18 エックス4 ファーマシューティカルズ, インコーポレイテッド 癌を処置する方法
BR112018072339A2 (pt) 2016-05-04 2019-02-19 Concert Pharmaceuticals, Inc. tratamento de distúrbios de perda de cabelo com inibidores de jak deuterados
CN107513069A (zh) * 2016-06-16 2017-12-26 正大天晴药业集团股份有限公司 手性吡咯并嘧啶化合物的制备方法
CN107759600A (zh) * 2016-06-16 2018-03-06 正大天晴药业集团股份有限公司 作为jak抑制剂的吡咯并嘧啶化合物的结晶
CN107513067A (zh) * 2016-06-16 2017-12-26 北京赛林泰医药技术有限公司 含有取代环戊基的吡咯并嘧啶化合物
US10759796B2 (en) 2016-06-21 2020-09-01 X4 Pharmaceuticals, Inc. CXCR4 inhibitors and uses thereof
EP3471726A4 (en) 2016-06-21 2019-10-09 X4 Pharmaceuticals, Inc. CXCR4 INHIBITORS AND USES THEREOF
WO2017223243A1 (en) 2016-06-21 2017-12-28 X4 Pharmaceuticals, Inc. Cxcr4 inhibitors and uses thereof
EP3950691A1 (en) 2016-06-30 2022-02-09 Daewoong Pharmaceutical Co., Ltd. Pyrazolopyrimidine derivatives as kinase inhibitor
EP3507367A4 (en) 2016-07-05 2020-03-25 Aduro BioTech, Inc. CYCLIC DINUCLEOTID COMPOUNDS WITH INCLUDED NUCLEIC ACIDS AND USES THEREOF
CN109843883B (zh) * 2016-07-26 2022-01-14 苏州隆博泰药业有限公司 作为选择性jak抑制剂化合物,该化合物的盐类及其治疗用途
JP2019530650A (ja) 2016-08-24 2019-10-24 アークル インコーポレイテッド アミノ−ピロロピリミジノン化合物およびその使用方法
JOP20190024A1 (ar) 2016-08-26 2019-02-19 Gilead Sciences Inc مركبات بيروليزين بها استبدال واستخداماتها
IT201600092051A1 (it) * 2016-09-13 2018-03-13 Alessandro Antonelli Composto medicale per il trattamento di tumori della tiroide
EP3526222B1 (en) 2016-10-14 2022-08-17 Nimbus Lakshmi, Inc. Tyk2 inhibitors and uses thereof
EP3528816A4 (en) 2016-10-21 2020-04-08 Nimbus Lakshmi, Inc. TYK2 INHIBITORS AND USES THEREOF
WO2018089499A1 (en) 2016-11-08 2018-05-17 Navitor Pharmaceuticals, Inc. PHENYL AMINO PIPERIDINE mTORC INHIBITORS AND USES THEREOF
CA3043768A1 (en) 2016-11-29 2018-06-07 PureTech Health LLC Exosomes for delivery of therapeutic agents
US11091451B2 (en) 2016-12-05 2021-08-17 Raze Therapeutics, Inc. SHMT inhibitors and uses thereof
RU2644155C1 (ru) * 2016-12-12 2018-02-08 Закрытое акционерное общество "Р-Фарм" (ЗАО "Р-Фарм") 2-(3-(4-(7H-пирроло[2,3-d]пиримидин-4-ил)-1H-пиразол-1-ил)-1-(этилсульфонил)азетидин-3-ил)ацетонитрила геминафтилдисульфонат в качестве ингибитора Янус киназ
JP2020502238A (ja) 2016-12-23 2020-01-23 バイスクルアールディー・リミテッド 新規連結構造を有するペプチド誘導体
EP3565638B8 (en) 2017-01-06 2024-04-10 BicycleRD Limited Bicycle conjugate for treating cancer
EA039352B1 (ru) * 2017-01-19 2022-01-17 Сучжоу Лонгбайотек Фармасьютикалз Ко., Лтд. Соединение в качестве селективного ингибитора jak и его соли и терапевтическое применение
TWI783978B (zh) 2017-03-08 2022-11-21 美商林伯士拉克許米公司 Tyk2抑制劑、其用途及生產方法
EP3375778A1 (en) 2017-03-14 2018-09-19 Artax Biopharma Inc. Aryl-piperidine derivatives
EP3375784A1 (en) 2017-03-14 2018-09-19 Artax Biopharma Inc. Aza-dihydro-acridone derivatives
WO2018191146A1 (en) 2017-04-10 2018-10-18 Navitor Pharmaceuticals, Inc. Heteroaryl rheb inhibitors and uses thereof
JOP20180036A1 (ar) 2017-04-18 2019-01-30 Vifor Int Ag أملاح لمثبطات فروبورتين جديدة
WO2018200625A1 (en) 2017-04-26 2018-11-01 Navitor Pharmaceuticals, Inc. Modulators of sestrin-gator2 interaction and uses thereof
US10857196B2 (en) 2017-04-27 2020-12-08 Bicycletx Limited Bicyclic peptide ligands and uses thereof
UY37695A (es) 2017-04-28 2018-11-30 Novartis Ag Compuesto dinucleótido cíclico bis 2’-5’-rr-(3’f-a)(3’f-a) y usos del mismo
CA3063963A1 (en) 2017-05-23 2018-11-29 Theravance Biopharma R&D Ip, Llc Glucuronide prodrugs of janus kinase inhibitors
WO2018231944A1 (en) 2017-06-13 2018-12-20 Berenson James R Diagnostic, prognostic, and monitoring methods for solid tumor cancers
JP7433910B2 (ja) 2017-06-22 2024-02-20 ノバルティス アーゲー Cd73に対する抗体分子及びその使用
WO2018237173A1 (en) 2017-06-22 2018-12-27 Novartis Ag ANTIBODY MOLECULES DIRECTED AGAINST CD73 AND CORRESPONDING USES
WO2019002842A1 (en) 2017-06-26 2019-01-03 Bicyclerd Limited BICYCLIC PEPTIDE LIGANDS WITH DETECTABLE FRACTIONS AND USES THEREOF
CN107298680A (zh) * 2017-07-12 2017-10-27 海门华祥医药科技有限公司 一种4‑氯‑7‑氮杂吲哚的生产工艺
JP7216705B2 (ja) 2017-07-28 2023-02-02 ニンバス ラクシュミ, インコーポレイテッド Tyk2阻害剤およびその使用方法
EP3668887A1 (en) 2017-08-14 2020-06-24 Bicyclerd Limited Bicyclic peptide ligand sting conjugates and uses thereof
WO2019034868A1 (en) 2017-08-14 2019-02-21 Bicyclerd Limited CONJUGATES PEPTIDE BICYCLIC-LIGAND PPR-A AND USES THEREOF
US11883497B2 (en) 2017-08-29 2024-01-30 Puretech Lyt, Inc. Lymphatic system-directing lipid prodrugs
EP4306524A2 (en) 2017-08-29 2024-01-17 PureTech LYT, Inc. Lymphatic system-directing lipid prodrugs
US11358948B2 (en) 2017-09-22 2022-06-14 Kymera Therapeutics, Inc. CRBN ligands and uses thereof
IL307995A (en) 2017-09-22 2023-12-01 Kymera Therapeutics Inc Protein compounds and their uses
RS62872B1 (sr) 2017-09-27 2022-02-28 Incyte Corp Soli derivata pirrolotriazina korisne kao tam inhibitori
CN109651424B (zh) * 2017-10-11 2021-01-22 新发药业有限公司 一种7-保护基-4-(1-氢-吡唑-4-基)吡咯[2,3-d]嘧啶的合成方法
US10800775B2 (en) 2017-11-03 2020-10-13 Aclaris Therapeutics, Inc. Pyrazolyl pyrrolo[2,3-b]pyrmidine-5-carboxylate analogs and methods of making the same
BR112020008850A2 (pt) 2017-11-03 2020-10-20 Aclaris Therapeutics, Inc. composto, composição farmacêutica e método para tratar uma doença mediada por jak1 e jak3
KR102034538B1 (ko) 2017-11-28 2019-10-21 주식회사한국파마 Jak 저해제 화합물, 및 이의 제조방법
US10596161B2 (en) 2017-12-08 2020-03-24 Incyte Corporation Low dose combination therapy for treatment of myeloproliferative neoplasms
GB201721265D0 (en) 2017-12-19 2018-01-31 Bicyclerd Ltd Bicyclic peptide ligands specific for EphA2
TWI825046B (zh) 2017-12-19 2023-12-11 英商拜西可泰克斯有限公司 Epha2特用之雙環胜肽配位基
US11608345B1 (en) 2017-12-19 2023-03-21 Puretech Lyt, Inc. Lipid prodrugs of rapamycin and its analogs and uses thereof
WO2019126378A1 (en) 2017-12-19 2019-06-27 Ariya Therapeutics, Inc. Lipid prodrugs of mycophenolic acid and uses thereof
US11304954B2 (en) 2017-12-19 2022-04-19 Puretech Lyt, Inc. Lipid prodrugs of mycophenolic acid and uses thereof
MX2020006812A (es) 2017-12-26 2020-11-06 Kymera Therapeutics Inc Degradadores de cinasas asociadas al receptor de interleucina-1 (irak) y usos de los mismos.
US11512080B2 (en) 2018-01-12 2022-11-29 Kymera Therapeutics, Inc. CRBN ligands and uses thereof
EP3737666A4 (en) 2018-01-12 2022-01-05 Kymera Therapeutics, Inc. PROTEIN DEGRADANTS AND USES THEREOF
KR20200115620A (ko) 2018-01-29 2020-10-07 메르크 파텐트 게엠베하 Gcn2 억제제 및 이의 용도
EP3746075A4 (en) 2018-01-29 2021-09-08 Merck Patent GmbH GCN2 INHIBITORS AND THEIR USES
PL3746429T3 (pl) 2018-01-30 2022-06-20 Incyte Corporation Procesy do otrzymywania (1-(3-fluoro-2-(trifluorometylo)izonikotynoilo)piperydyn-4-onu)
SG11202007805SA (en) 2018-02-16 2020-09-29 Incyte Corp Jak1 pathway inhibitors for the treatment of cytokine-related disorders
EP3759109B1 (en) 2018-02-26 2023-08-30 Gilead Sciences, Inc. Substituted pyrrolizine compounds as hbv replication inhibitors
WO2019169001A1 (en) 2018-02-27 2019-09-06 Artax Biopharma Inc. Chromene derivatives as inhibitors of tcr-nck interaction
KR20210003086A (ko) 2018-03-08 2021-01-11 노파르티스 아게 항-p-셀렉틴 항체의 용도
CN110357887B (zh) * 2018-03-26 2022-09-16 武汉誉祥医药科技有限公司 取代的7H-吡咯并[2,3-d]嘧啶衍生物及其制备方法和用途
WO2019191684A1 (en) 2018-03-30 2019-10-03 Incyte Corporation Treatment of hidradenitis suppurativa using jak inhibitors
US11584961B2 (en) 2018-03-30 2023-02-21 Incyte Corporation Biomarkers for inflammatory skin disease
MX2020010815A (es) 2018-04-13 2020-12-11 Incyte Corp Biomarcadores para enfermedad de injerto contra hospedero.
CA3097774A1 (en) 2018-04-24 2019-10-31 Vertex Pharmaceuticals Incorporated Pteridinone compounds and uses thereof
ES2919572T3 (es) 2018-04-24 2022-07-27 Merck Patent Gmbh Compuestos antiproliferación y usos de los mismos
TW202015726A (zh) 2018-05-30 2020-05-01 瑞士商諾華公司 Entpd2抗體、組合療法、及使用該等抗體和組合療法之方法
US20210214459A1 (en) 2018-05-31 2021-07-15 Novartis Ag Antibody molecules to cd73 and uses thereof
WO2019233434A1 (zh) * 2018-06-06 2019-12-12 杭州澳津生物医药技术有限公司 一种吡唑嘧啶衍生物及其用途和药物组合物
JP7382353B2 (ja) 2018-06-15 2023-11-16 ヤンセン ファーマシューティカ エヌ.ベー. ラパマイシン類似体およびその使用
US11180531B2 (en) 2018-06-22 2021-11-23 Bicycletx Limited Bicyclic peptide ligands specific for Nectin-4
GB201810316D0 (en) 2018-06-22 2018-08-08 Bicyclerd Ltd Peptide ligands for binding to EphA2
JP2021529765A (ja) 2018-06-29 2021-11-04 インサイト・コーポレイションIncyte Corporation Axl/mer阻害剤の製剤
JP6830460B2 (ja) * 2018-07-05 2021-02-17 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 ルキソリチニブの重水素化誘導体
WO2020010177A1 (en) 2018-07-06 2020-01-09 Kymera Therapeutics, Inc. Tricyclic crbn ligands and uses thereof
KR102653681B1 (ko) 2018-07-31 2024-04-03 록쏘 온콜로지, 인코포레이티드 (s)-5-아미노-3-(4-((5-플루오로-2-메톡시벤즈아미도)메틸)페닐)-1-(1,1,1-트리플루오로프로판-2-일)-1h-피라졸-4-카르복스아미드의분무-건조된 분산물 및 제제
EP3833350A4 (en) 2018-08-10 2022-05-18 Aclaris Therapeutics, Inc. PYRROLOPYRIMIDINE TKI INHIBITORS
WO2020039401A1 (en) 2018-08-24 2020-02-27 Novartis Ag Treatment comprising il-1βeta binding antibodies and combinations thereof
US10548889B1 (en) 2018-08-31 2020-02-04 X4 Pharmaceuticals, Inc. Compositions of CXCR4 inhibitors and methods of preparation and use
EP3846793B1 (en) 2018-09-07 2024-01-24 PIC Therapeutics, Inc. Eif4e inhibitors and uses thereof
WO2020081508A1 (en) 2018-10-15 2020-04-23 Nimbus Lakshmi, Inc. Tyk2 inhibitors and uses thereof
CN112955459A (zh) 2018-10-23 2021-06-11 拜斯科技术开发有限公司 双环肽配体和其用途
AU2019364542A1 (en) 2018-10-24 2021-06-10 Navitor Pharmaceuticals, Inc. Polymorphic compounds and uses thereof
US11324749B2 (en) 2018-10-31 2022-05-10 Incyte Corporation Combination therapy for treatment of hematological diseases
AU2019389025A1 (en) 2018-11-30 2021-06-17 Takeda Pharmaceutical Company Limited TYK2 inhibitors and uses thereof
US11352350B2 (en) 2018-11-30 2022-06-07 Kymera Therapeutics, Inc. IRAK degraders and uses thereof
CN109394768B (zh) * 2018-12-10 2019-08-23 牡丹江医学院 一种治疗湿疹的药物及其制备方法
CN111320633B (zh) * 2018-12-14 2022-09-27 中国医药研究开发中心有限公司 吡咯/咪唑并六元杂芳环类化合物及其制备方法和医药用途
EP3898615A1 (en) 2018-12-19 2021-10-27 Array Biopharma, Inc. 7-((3,5-dimethoxyphenyl)amino)quinoxaline derivatives as fgfr inhibitors for treating cancer
CN113490666A (zh) 2018-12-19 2021-10-08 奥瑞生物药品公司 作为fgfr酪氨酸激酶的抑制剂的取代的吡唑并[1,5-a]吡啶化合物
EP3670659A1 (en) 2018-12-20 2020-06-24 Abivax Biomarkers, and uses in treatment of viral infections, inflammations, or cancer
BR112021011894A2 (pt) 2018-12-21 2021-09-08 Daiichi Sankyo Company, Limited Composição farmacêutica
CN113348021A (zh) 2019-01-23 2021-09-03 林伯士拉克许米公司 Tyk2抑制剂和其用途
WO2020165600A1 (en) 2019-02-14 2020-08-20 Bicycletx Limited Bicyclic peptide ligand sting conjugates and uses thereof
CN111620873B (zh) * 2019-02-28 2021-12-28 沈阳药科大学 一类含哌啶的吡咯并[2,3-d]嘧啶衍生物及其制备和用途
KR20210137087A (ko) 2019-03-05 2021-11-17 인사이트 코포레이션 만성 폐 동종이식 기능장애의 치료를 위한 jak1 경로 억제제
US10786485B1 (en) 2019-03-11 2020-09-29 Nocion Therapeutics, Inc. Charged ion channel blockers and methods for use
EP3937945A4 (en) 2019-03-11 2023-01-04 Nocion Therapeutics, Inc. CHARGED ION CHANNEL BLOCKERS AND METHODS OF USE
US10780083B1 (en) 2019-03-11 2020-09-22 Nocion Therapeutics, Inc. Charged ion channel blockers and methods for use
CA3129117A1 (en) 2019-03-11 2020-09-17 Bridget Mccarthy Cole Charged ion channel blockers and methods for use
JP2022527731A (ja) 2019-03-11 2022-06-06 ノシオン セラピューティクス,インコーポレイテッド エステル置換イオンチャンネル遮断薬および使用方法
TW202102222A (zh) 2019-03-19 2021-01-16 美商英塞特公司 白斑病之生物標記物
AU2020242287A1 (en) 2019-03-21 2021-09-02 INSERM (Institut National de la Santé et de la Recherche Médicale) A Dbait molecule in combination with kinase inhibitor for the treatment of cancer
JP2022528887A (ja) 2019-04-02 2022-06-16 バイスクルテクス・リミテッド バイシクルトキシンコンジュゲートおよびその使用
KR20220006139A (ko) 2019-04-05 2022-01-14 카이메라 쎄라퓨틱스 인코포레이티드 Stat 분해제 및 이의 용도
CN114466681A (zh) 2019-04-24 2022-05-10 礼蓝美国公司 7h-吡咯并[2,3-d]嘧啶jak抑制剂
JP2022531088A (ja) 2019-05-02 2022-07-06 アクラリス セラピューティクス,インコーポレイテッド Jak阻害剤としての置換ピロロピリジン
KR102286372B1 (ko) 2019-05-27 2021-08-05 주식회사한국파마 Jak 저해제 화합물, 및 이를 포함하는 의약 조성물
CN110028509B (zh) * 2019-05-27 2020-10-09 上海勋和医药科技有限公司 作为选择性jak2抑制剂的吡咯并嘧啶类化合物、其合成方法及用途
JP2022534425A (ja) 2019-05-31 2022-07-29 イケナ オンコロジー, インコーポレイテッド Tead阻害剤およびその使用
CA3150975A1 (en) 2019-06-10 2020-12-17 Incyte Corporation Topical treatment of vitiligo by a jak inhibitor
MX2022000016A (es) 2019-06-27 2022-02-24 Crispr Therapeutics Ag Uso de linfocitos t con receptor de antigeno quimerico e inhibidores de linfocitos citoliticos naturales para el tratamiento del cancer.
CN110305140B (zh) 2019-07-30 2020-08-04 上海勋和医药科技有限公司 二氢吡咯并嘧啶类选择性jak2抑制剂
WO2021022178A1 (en) * 2019-07-31 2021-02-04 Aclaris Therapeutics, Inc. Substituted sulfonamide pyrrolopyridines as jak inhibitors
WO2021022076A1 (en) * 2019-08-01 2021-02-04 St. Jude Children's Research Hospital Molecules and methods related to treatment of uncontrolled cellular proliferation
AU2020345962A1 (en) 2019-09-11 2022-03-31 Vincere Biosciences, Inc. USP30 inhibitors and uses thereof
JP2022547719A (ja) 2019-09-13 2022-11-15 ニンバス サターン, インコーポレイテッド Hpk1アンタゴニストおよびその使用
TW202124443A (zh) 2019-09-16 2021-07-01 瑞士商諾華公司 高親和力的、配位基阻斷性、人源化的抗T細胞免疫球蛋白結構域和黏蛋白結構域3(TIM-3)IgG4抗體用於治療骨髓纖維化之用途
WO2021053489A1 (en) 2019-09-16 2021-03-25 Novartis Ag Use of an mdm2 inhibitor for the treatment of myelofibrosis
JP2022548881A (ja) 2019-09-18 2022-11-22 ノバルティス アーゲー Entpd2抗体、組合せ療法並びに抗体及び組合せ療法を使用する方法
CN110538183B (zh) * 2019-10-09 2021-05-04 吉林大学 一种预防和治疗小儿湿疹的组合物及其制备方法
US20210123930A1 (en) 2019-10-10 2021-04-29 Incyte Corporation Biomarkers for graft-versus-host disease
US20210123931A1 (en) 2019-10-10 2021-04-29 Incyte Corporation Biomarkers for graft-versus-host disease
JP2023506118A (ja) 2019-10-16 2023-02-15 インサイト・コーポレイション 皮膚エリテマトーデス及び扁平苔癬(lp)の治療のためのjak1阻害剤の使用
US11723890B2 (en) 2019-11-01 2023-08-15 Navitor Pharmaceuticals, Inc. Methods of treatment using an mTORC1 modulator
EP4054559A4 (en) 2019-11-06 2023-11-29 Nocion Therapeutics, Inc. CHARGED ION CHANNEL BLOCKERS AND METHODS OF USE
US10933055B1 (en) 2019-11-06 2021-03-02 Nocion Therapeutics, Inc. Charged ion channel blockers and methods for use
US20220401436A1 (en) 2019-11-08 2022-12-22 INSERM (Institute National de la Santé et de la Recherche Médicale) Methods for the treatment of cancers that have acquired resistance to kinase inhibitors
KR20220107213A (ko) 2019-11-22 2022-08-02 인사이트 코포레이션 Alk2 억제제 및 jak2 억제제를 포함하는 병용 요법
CN115209897A (zh) 2019-12-05 2022-10-18 安纳库利亚治疗公司 雷帕霉素类似物及其用途
US11591332B2 (en) 2019-12-17 2023-02-28 Kymera Therapeutics, Inc. IRAK degraders and uses thereof
IL293917A (en) 2019-12-17 2022-08-01 Kymera Therapeutics Inc Iraq joints and their uses
CA3162502A1 (en) 2019-12-23 2021-07-01 Yi Zhang Smarca degraders and uses thereof
US11091447B2 (en) 2020-01-03 2021-08-17 Berg Llc UBE2K modulators and methods for their use
WO2021148581A1 (en) 2020-01-22 2021-07-29 Onxeo Novel dbait molecule and its use
CN111728975A (zh) * 2020-02-25 2020-10-02 广东省检迅检测科技有限公司 用于减少运动损伤和促进运动损伤修复的组合物
TW202146393A (zh) 2020-03-03 2021-12-16 美商皮克醫療公司 Eif4e抑制劑及其用途
WO2021183639A1 (en) 2020-03-11 2021-09-16 Nocion Therapeutics, Inc. Charged ion channel blockers and methods for use
EP4121043A1 (en) 2020-03-19 2023-01-25 Kymera Therapeutics, Inc. Mdm2 degraders and uses thereof
US11324750B2 (en) 2020-04-09 2022-05-10 Children's Hospital Medical Center Compositions and methods for the treatment of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection
WO2021206766A1 (en) 2020-04-09 2021-10-14 Children's Hospital Medical Center Sars-cov-2 infection biomarkers and uses thereof
EP3892280A3 (en) 2020-04-09 2022-01-12 Children's Hospital Medical Center Sars-cov-2 infection biomarkers and uses thereof
WO2021209563A1 (en) 2020-04-16 2021-10-21 Som Innovation Biotech, S.A. Compounds for use in the treatment of viral infections by respiratory syndrome-related coronavirus
WO2021236139A1 (en) 2020-05-21 2021-11-25 Concert Pharmaceuticals, Inc. Novel deuterated jak inhibitor and uses thereof
US11685731B2 (en) 2020-06-02 2023-06-27 Incyte Corporation Processes of preparing a JAK1 inhibitor
US11833155B2 (en) 2020-06-03 2023-12-05 Incyte Corporation Combination therapy for treatment of myeloproliferative neoplasms
IL298200A (en) 2020-06-03 2023-01-01 Incyte Corp Combined therapy for the treatment of myeloproliferative neoplasms
TW202210483A (zh) 2020-06-03 2022-03-16 美商凱麥拉醫療公司 Irak降解劑之結晶型
KR20230035031A (ko) 2020-06-05 2023-03-10 킨네이트 바이오파마 인크. 섬유아세포 성장 인자 수용체 키나아제의 억제제
US20220023344A1 (en) 2020-06-26 2022-01-27 Crispr Therapeutics Ag Allogeneic cell therapy of acute lymphoblastic leukemia using genetically engineered t cells targeting cd19
EP3944859A1 (en) 2020-07-30 2022-02-02 Assistance Publique Hôpitaux de Paris Method for treating immune toxicities induced by immune checkpoint inhibitors
US11751108B2 (en) * 2020-08-05 2023-09-05 Qualcomm Incorporated Execution of reduced signaling handover
CA3194469A1 (en) * 2020-08-12 2022-02-17 Concert Pharmaceuticals, Inc. Process for preparing enantiomerically enriched jak inhibitors
CN116234931A (zh) 2020-08-17 2023-06-06 拜斯科技术开发有限公司 对nectin-4具有特异性的双环缀合物及其用途
EP4200278A1 (en) 2020-08-18 2023-06-28 Incyte Corporation Process and intermediates for preparing a jak1 inhibitor
CR20230129A (es) 2020-08-18 2023-07-13 Incyte Corp Proceso e intermediarios para preparar un inhibidor de jak
US20230364095A1 (en) 2020-09-16 2023-11-16 Incyte Corporation Topical treatment of vitiligo
EP4221711A1 (en) 2020-10-02 2023-08-09 Incyte Corporation Topical ruxolitinib for treating lichen planus
EP4225317A1 (en) 2020-10-08 2023-08-16 Novartis AG Use of an erk inhibitor for the treatment of myelofibrosis
EP4225316A1 (en) 2020-10-08 2023-08-16 Novartis AG Use of an erk inhibitor for the treatment of myelofibrosis
CN114437079A (zh) * 2020-10-30 2022-05-06 杭州邦顺制药有限公司 吡咯嘧啶五元氮杂环化合物的晶型
KR102551758B1 (ko) 2020-11-30 2023-07-05 주식회사한국파마 신규한 jak 특이 저해제 화합물, 및 이의 제조방법
WO2022120353A1 (en) 2020-12-02 2022-06-09 Ikena Oncology, Inc. Tead inhibitors and uses thereof
EP4255895A1 (en) 2020-12-02 2023-10-11 Ikena Oncology, Inc. Tead inhibitors and uses thereof
KR20230128472A (ko) 2020-12-04 2023-09-05 인사이트 코포레이션 피부 질환의 치료를 위한 비타민 d 유사체를 함유하는jak 억제제
KR20230118118A (ko) 2020-12-08 2023-08-10 인사이트 코포레이션 백반증의 치료를 위한 jak1 경로 저해제
AU2021401834A1 (en) 2020-12-18 2023-07-06 Boehringer Ingelheim Animal Health USA Inc. Boron containing pyrazole compounds, compositions comprising them, methods and uses thereof
KR20230157307A (ko) 2021-01-11 2023-11-16 인사이트 코포레이션 Jak 경로 억제제 및 rock 억제제를 포함하는 병용 요법
EP4284802A1 (en) * 2021-02-01 2023-12-06 Janssen Biotech, Inc. Small molecule inhibitors of salt inducible kinases
US20230113202A1 (en) 2021-02-02 2023-04-13 Liminal Biosciences Limited Gpr84 antagonists and uses thereof
CN116888116A (zh) 2021-02-02 2023-10-13 里米诺生物科学有限公司 Gpr84拮抗剂和其用途
CN116981669A (zh) * 2021-02-05 2023-10-31 上海齐鲁制药研究中心有限公司 嘧啶或吡啶并杂环类腺苷受体抑制剂及其制备方法和用途
TW202245789A (zh) 2021-02-15 2022-12-01 美商凱麥拉醫療公司 Irak4降解劑及其用途
EP4297750A1 (en) 2021-02-25 2024-01-03 Impact Biomedicines, Inc. Use of a bet inhibitor alone or in combination with fedratinib or ruxolitinib for treating a hematological malignancy such as myelofibrosis
EP4301756A1 (en) 2021-03-05 2024-01-10 Nimbus Saturn, Inc. Hpk1 antagonists and uses thereof
AU2022258968A1 (en) 2021-04-16 2023-10-19 Ikena Oncology, Inc. Mek inhibitors and uses thereof
US20230134932A1 (en) 2021-08-25 2023-05-04 PIC Therapeutics, Inc. Eif4e inhibitors and uses thereof
WO2023028235A1 (en) 2021-08-25 2023-03-02 PIC Therapeutics, Inc. Eif4e inhibitors and uses thereof
CA3231996A1 (en) * 2021-09-18 2023-03-23 Satya Srinivas HANUMARA An improved process for the preparation of ruxolitinib phosphate
WO2023076161A1 (en) 2021-10-25 2023-05-04 Kymera Therapeutics, Inc. Tyk2 degraders and uses thereof
WO2023102559A1 (en) 2021-12-03 2023-06-08 Incyte Corporation Topical formulations of ruxolitinib with an organic amine ph adjusting agent for treatment of skin diseases
WO2023114984A1 (en) 2021-12-17 2023-06-22 Ikena Oncology, Inc. Tead inhibitors and uses thereof
CN114044777B (zh) * 2022-01-10 2022-04-19 南京佰麦生物技术有限公司 一种磷酸芦可替尼的制备方法
CN114456181A (zh) * 2022-02-21 2022-05-10 浙江乐普药业股份有限公司 一种芦可替尼的制备方法
WO2023173053A1 (en) 2022-03-10 2023-09-14 Ikena Oncology, Inc. Mek inhibitors and uses thereof
WO2023173057A1 (en) 2022-03-10 2023-09-14 Ikena Oncology, Inc. Mek inhibitors and uses thereof
WO2023211889A1 (en) 2022-04-25 2023-11-02 Ikena Oncology, Inc. Polymorphic compounds and uses thereof
US11878958B2 (en) 2022-05-25 2024-01-23 Ikena Oncology, Inc. MEK inhibitors and uses thereof
WO2024028364A1 (en) 2022-08-02 2024-02-08 Liminal Biosciences Limited Aryl-triazolyl and related gpr84 antagonists and uses thereof
WO2024028365A1 (en) 2022-08-02 2024-02-08 Liminal Biosciences Limited Substituted pyridone gpr84 antagonists and uses thereof
WO2024028363A1 (en) 2022-08-02 2024-02-08 Liminal Biosciences Limited Heteroaryl carboxamide and related gpr84 antagonists and uses thereof
WO2024028193A1 (en) 2022-08-03 2024-02-08 Medichem, S.A. Stable oral pharmaceutical formulation containing ruxolitinib hemifumarate

Family Cites Families (311)

* 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 (de) 1980-09-26 1982-05-13 Troponwerke GmbH & Co KG, 5000 Köln Neue pyrrolo-pyrimidine, verfahren zu ihrer herstellung und ihre verwendung bei der herstellung von biologischen wirkstoffen
DE3220113A1 (de) * 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 (nl) 1984-10-24 1986-05-16 Oce Andeno Bv Dioxafosforinanen, de bereiding ervan en de toepassing voor het splitsen van optisch actieve verbindingen.
CA1306260C (en) 1985-10-18 1992-08-11 Shionogi & Co., Ltd. Condensed imidazopyridine derivatives
US4921947A (en) 1986-03-31 1990-05-01 Eli Lilly And Company Process for preparing macrolide derivatives
JPH0710876Y2 (ja) 1989-08-31 1995-03-15 石垣機工株式会社 スクリュープレスにおける脱水筒の洗浄装置
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 (it) * 1992-01-16 1996-02-29 Zambon Spa Composizione farmaceutica oftalmica contenente n-acetilcisteina e polivinilalcol
US5521184A (en) 1992-04-03 1996-05-28 Ciba-Geigy Corporation Pyrimidine derivatives and processes for the preparation thereof
FR2695126B1 (fr) 1992-08-27 1994-11-10 Sanofi Elf Dérivés d'acide thiényl ou pyrrolyl carboxyliques, leur préparation et médicaments les contenant.
AU671491B2 (en) 1992-12-18 1996-08-29 F. Hoffmann-La Roche Ag N-oxycarbonyl substituted 5'-deoxy-5-fluorcytidines
JPH0710876A (ja) * 1993-06-24 1995-01-13 Teijin Ltd 4位に環状アミノ基を有するピロロ[2,3―d]ピリミジン
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 and cosmetic compositions containing God-type ellagitannin as an 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 (zh) 1995-07-05 2003-04-09 纳幕尔杜邦公司 杀真菌嘧啶酮
SI9620103A (sl) 1995-07-06 1998-10-31 Novartis Ag Pirolopirimidini in postopki za njihovo pripravo
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 (ja) 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 癌治療方法
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 (ja) 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 癌の治療方法
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 (ja) * 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−ヘテロアリールジピリド[2,3−b:3’,2’−f]アゼピン類及びHIV感染の予防及び治療におけるそれらの使用
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
SE9800729L (sv) 1998-03-06 1999-09-07 Scotia Lipidteknik Ab Ny topikal formulering I
US6025366A (en) 1998-04-02 2000-02-15 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6232320B1 (en) * 1998-06-04 2001-05-15 Abbott Laboratories Cell adhesion-inhibiting antiinflammatory compounds
SK18542000A3 (sk) 1998-06-04 2001-12-03 Abbott Laboratories Protizápalové zlúčeniny inhibujúce bunkovú adhéziu
PA8474101A1 (es) * 1998-06-19 2000-09-29 Pfizer Prod Inc Compuestos de pirrolo [2,3-d] pirimidina
ID26698A (id) 1998-06-19 2001-02-01 Pfizer Prod Inc SENYAWA-SENYAWA PIROLO [2,3-d] PIRIMIDINA
ATE459616T1 (de) 1998-08-11 2010-03-15 Novartis Ag Isochinoline derivate mit angiogenesis-hemmender wirkung
JP2000119271A (ja) 1998-08-12 2000-04-25 Hokuriku Seiyaku Co Ltd 1h―イミダゾピリジン誘導体
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
US6413419B1 (en) * 1998-10-29 2002-07-02 Institut Francais Du Petrole Process and device for separation with variable-length chromatographic
FR2785196B1 (fr) * 1998-10-29 2000-12-15 Inst Francais Du Petrole Procede et dispositif de separation avec des zones chromatographiques a longueur variable
US6133031A (en) 1999-08-19 2000-10-17 Isis Pharmaceuticals Inc. Antisense inhibition of focal adhesion kinase expression
WO2000051614A1 (en) 1999-03-03 2000-09-08 Merck & Co., Inc. Inhibitors of prenyl-protein transferases
GB9905075D0 (en) 1999-03-06 1999-04-28 Zeneca Ltd Chemical compounds
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
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 (da) * 1999-12-10 2004-03-29 Pfizer Prod Inc Pyrrolo[2,3-d]pyrimidin-forbindelser som proteinkinaseinhibitorer
CN1615873A (zh) * 1999-12-24 2005-05-18 阿文蒂斯药物有限公司 氮杂吲哚类化合物
GB0004890D0 (en) 2000-03-01 2000-04-19 Astrazeneca Uk Ltd Chemical compounds
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 (da) * 2000-04-07 2004-02-09 Medidom Lab Oftalmologiske formuleringer på basis af ciclosporin, hyaluronsyre og polysorbat
WO2001081345A1 (fr) 2000-04-20 2001-11-01 Mitsubishi Pharma Corporation Composes d'amides aromatiques
SI2223922T1 (sl) * 2000-04-25 2016-04-29 Icos Corporation Inhibitorji humane fosfatidil-inositol 3-kinazne delta izoforme
JP4954426B2 (ja) 2000-06-16 2012-06-13 キュリス,インコーポレイテッド 血管形成調節組成物及び利用
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 (de) * 2000-06-23 2010-05-15 Mitsubishi Tanabe Pharma Corp Antitumoreffekt-verstärker
EE200200711A (et) * 2000-06-26 2004-06-15 Pfizer Products Inc. Pürrolo[2,3-d]pürimidiinühendid kui immunosupressiivsed vahendid
CN1321628C (zh) 2000-06-28 2007-06-20 史密斯克莱·比奇曼公司 湿磨方法
AU2001278790A1 (en) 2000-08-22 2002-03-04 Hokuriku Seiyaku Co. Ltd 1h-imidazopyridine derivatives
MXPA03005001A (es) * 2000-12-05 2003-09-05 Vertex Pharma Inhibidores de n- terminal c-jun cinasas (jnk) y otras proteinas cinasas.
GB0100622D0 (en) 2001-01-10 2001-02-21 Vernalis Res Ltd Chemical compounds V111
JP2004520347A (ja) * 2001-01-15 2004-07-08 グラクソ グループ リミテッド Ldl−受容体発現のインデューサーとしてのアリールピペリジンおよびピペラジン誘導体
EP1363702A4 (en) 2001-01-30 2007-08-22 Cytopia Pty Ltd PROCESS FOR INHIBITING KINASES
JP4316893B2 (ja) 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 Srcおよび他のプロテインキナーゼのインヒビター
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
DE60230890D1 (de) 2001-09-19 2009-03-05 Aventis Pharma Sa Indolizine als kinaseproteinhemmer
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
IL161156A0 (en) 2001-10-30 2004-08-31 Novartis Ag Staurosporine derivatives as inhibitors of flt3 receptor tyrosine kinase activity
JP2003155285A (ja) 2001-11-19 2003-05-27 Toray Ind Inc 環状含窒素誘導体
AU2002224131A1 (en) * 2001-11-30 2003-06-17 Teijin Limited Process for producing 5-(3-cyanophenyl)-3-formylbenzoic acid compound
GT200200234A (es) 2001-12-06 2003-06-27 Compuestos cristalinos novedosos
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
PE20040522A1 (es) 2002-05-29 2004-09-28 Novartis Ag Derivados de diarilurea dependientes de la cinasa de proteina
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
GB0215676D0 (en) 2002-07-05 2002-08-14 Novartis Ag Organic compounds
GB0215844D0 (en) * 2002-07-09 2002-08-14 Novartis Ag Organic compounds
WO2004007472A1 (ja) * 2002-07-10 2004-01-22 Ono Pharmaceutical Co., Ltd. Ccr4アンタゴニストおよびその医薬用途
JP2006502183A (ja) 2002-09-20 2006-01-19 アルコン,インコーポレイテッド ドライアイ障害の処置のためのサイトカイン合成インヒビターの使用
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
AR042052A1 (es) 2002-11-15 2005-06-08 Vertex Pharma Diaminotriazoles utiles como inhibidores de proteinquinasas
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
UA80767C2 (en) 2002-12-20 2007-10-25 Pfizer Prod Inc Pyrimidine derivatives for the treatment of abnormal cell growth
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
GB0305929D0 (en) 2003-03-14 2003-04-23 Novartis Ag Organic compounds
US7547794B2 (en) 2003-04-03 2009-06-16 Vertex Pharmaceuticals Incorporated Compositions useful as inhibitors of protein kinases
SE0301373D0 (sv) * 2003-05-09 2003-05-09 Astrazeneca Ab Novel compounds
SE0301372D0 (sv) 2003-05-09 2003-05-09 Astrazeneca Ab Novel compounds
FR2857454B1 (fr) 2003-07-08 2006-08-11 Aventis Pasteur Dosage des acides techoiques des bacteries 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
PE20050952A1 (es) 2003-09-24 2005-12-19 Novartis Ag Derivados de isoquinolina como inhibidores de b-raf
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 (pt) 2003-11-25 2007-01-16 Pfizer Prod Inc método de tratamento de aterosclerose
CA2549485A1 (en) 2003-12-17 2005-07-07 Pfizer Products Inc. Pyrrolo [2,3-d] pyrimidine compounds for treating transplant rejection
EP1696920B8 (en) 2003-12-19 2015-05-06 Plexxikon Inc. Compounds and methods for development of ret modulators
CA2550189A1 (en) * 2003-12-19 2005-07-21 Schering Corporation Thiadiazoles as cxc- and cc- chemokine receptor ligands
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 (pl) 2004-03-30 2013-03-29 Vertex Pharma Azaindole użyteczne jako inhibitory JAK i innych kinaz białkowych
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 (ja) 2004-05-03 2007-12-13 ノバルティス アクチエンゲゼルシャフト S1p受容体アゴニストおよびjak3キナーゼ阻害剤を含む、組合せ剤
WO2005110410A2 (en) 2004-05-14 2005-11-24 Abbott Laboratories Kinase inhibitors as therapeutic agents
PE20060426A1 (es) 2004-06-02 2006-06-28 Schering Corp DERIVADOS DE ACIDO TARTARICO COMO INHIBIDORES DE MMPs, ADAMs, TACE Y TNF-alfa
TW200610762A (en) 2004-06-10 2006-04-01 Irm Llc Compounds and compositions as protein kinase inhibitors
JP5315611B2 (ja) * 2004-06-23 2013-10-16 小野薬品工業株式会社 S1p受容体結合能を有する化合物およびその用途
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 (fr) 2004-07-29 2006-10-06 Sanofi Synthelabo Derives d'amino-piperidine, leur preparation et leur application en therapeutique
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
GB0421525D0 (en) 2004-09-28 2004-10-27 Novartis Ag Inhibitors of protein kineses
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 (es) 2004-10-25 2006-05-31 Astex Therapeutics Ltd Derivados sustituidos de purina, purinona y deazapurina, composiciones que los contienen métodos para su preparación y sus usos
US7528138B2 (en) 2004-11-04 2009-05-05 Vertex Pharmaceuticals Incorporated Pyrazolo[1,5-a]pyrimidines useful as inhibitors of protein kinases
AU2005309019A1 (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
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
AR054416A1 (es) 2004-12-22 2007-06-27 Incyte Corp Pirrolo [2,3-b]piridin-4-il-aminas y pirrolo [2,3-b]pirimidin-4-il-aminas como inhibidores de las quinasas janus. composiciones farmaceuticas.
WO2006067445A2 (en) 2004-12-22 2006-06-29 Astrazeneca Ab Csf-1r kinase inhibitors
US20090124635A1 (en) 2005-01-20 2009-05-14 Pfizer Inc. Chemical compounds
KR20070104641A (ko) * 2005-02-03 2007-10-26 버텍스 파마슈티칼스 인코포레이티드 단백질 키나제의 억제제로 유용한 피롤로피리미딘
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 (pt) 2005-03-15 2010-01-05 Irm Llc compostos e composições como inibidores da proteìna quinase
BRPI0610514A2 (pt) 2005-04-05 2016-11-16 Pharmacopeia Inc composto, composição farmacêutica, e, método de tratamento de um distúrbio
GB0510139D0 (en) 2005-05-18 2005-06-22 Addex Pharmaceuticals Sa Novel compounds B1
MX2007014619A (es) * 2005-05-20 2009-02-13 Vertex Pharma Pirrolopiridinas de utilidad como inhibidores de proteina quinasa.
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 (zh) 2005-06-22 2015-06-24 普莱希科公司 作为蛋白质激酶抑制剂的吡咯并[2,3-b]吡啶衍生物
CN102127078A (zh) 2005-07-14 2011-07-20 安斯泰来制药株式会社 Janus激酶3的杂环类抑制剂
FR2889662B1 (fr) 2005-08-11 2011-01-14 Galderma Res & Dev Emulsion de type huile-dans-eau pour application topique en dermatologie
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 (pt) 2005-10-28 2011-08-16 Astrazeneca Ab composto ou um sal farmaceuticamente aceitável do mesmo, processo para a preparação do mesmo, composição farmacêutica, uso de um composto ou um sal farmaceuticamente aceitável do mesmo, e, métodos para produzir um efeito anti-proliferativo e um efeito pró-apoptósico em um animal de sangue quente, para tratar doença e para produzir um efeito inibidor de jak em um animal de sangue quente
DK1951684T3 (en) * 2005-11-01 2016-10-24 Targegen Inc BIARYLMETAPYRIMIDIN kinase inhibitors
WO2007062459A1 (en) 2005-11-29 2007-06-07 Cytopia Research Pty Ltd Selective kinase inhibitors based on pyridine scaffold
MY162590A (en) * 2005-12-13 2017-06-30 Incyte Holdings Corp Heteroaryl substituted pyrrolo[2,3-b] pyridines and pyrrolo[2,3-b] pyrimidines as janus kinase inhibitors
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 (ja) 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活性阻害剤
KR20080083680A (ko) * 2005-12-23 2008-09-18 스미스클라인 비참 코포레이션 오로라 키나제의 아자인돌 억제제
JP4643455B2 (ja) 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 遊技システム
BRPI0706537A2 (pt) 2006-01-17 2011-03-29 Vertex Pharma azaindóis úteis como inibidores de janus cinases
EP1979353A2 (en) * 2006-01-19 2008-10-15 OSI Pharmaceuticals, Inc. Fused heterobicyclic kinase inhibitors
JP2009525350A (ja) 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 Rafキナーゼ阻害薬として有用なピロロ[2,3,b]ピリジン誘導体
US7745477B2 (en) 2006-02-07 2010-06-29 Hoffman-La Roche Inc. Heteroaryl and benzyl amide compounds
PL1842534T3 (pl) 2006-02-24 2012-05-31 Teva Pharma Tabletki bursztynianu metoprololu o przedłużonym uwalnianiu oraz sposoby ich wytwarzania
EP1995246A4 (en) * 2006-03-10 2010-11-17 Ono Pharmaceutical Co NITROGENATED HETEROCYCLIC DERIVATIVE AND MEDICAMENT WITH DERIVATIVE ACTIVE SUBSTANCE
WO2007116866A1 (ja) * 2006-04-03 2007-10-18 Astellas Pharma Inc. ヘテロ化合物
MX2008012860A (es) 2006-04-05 2009-01-07 Vertex Pharma Desazapurinas de utilidad como inhibidores de janus cinasas.
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 (zh) 2006-05-26 2013-06-11 Novartis Ag 吡咯并嘧啶化合物及其用途
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 (fr) * 2006-08-03 2008-02-07 Takeda Pharmaceutical Company Limited INHIBITEUR DE LA GSK-3β
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 (es) 2006-10-04 2008-12-30 Pharmacopeia Inc Derivados de 2-(bencimidazolil) purina y purinonas 6-sustituidas utiles como inmunosupresores,y composiciones farmaceuticas que los contienen.
AR063141A1 (es) * 2006-10-04 2008-12-30 Pharmacopeia Inc Derivados de 2- ( benzimidazolil ) purina 8- sustituida para inmunosupresion
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 (ja) 2006-11-22 2014-08-13 インサイト・コーポレイション キナーゼ阻害剤としてのイミダゾトリアジンおよびイミダゾピリミジン
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
CA2667072C (en) 2006-12-22 2015-11-24 Sigma-Tau Industrie Farmaceutiche Riunite S.P.A. Gel useful for the delivery of ophthalmic drugs
US8513270B2 (en) * 2006-12-22 2013-08-20 Incyte Corporation Substituted heterocycles as Janus kinase inhibitors
WO2008082840A1 (en) 2006-12-29 2008-07-10 Abbott Laboratories Pim kinase inhibitors as cancer chemotherapeutics
KR20080062876A (ko) 2006-12-29 2008-07-03 주식회사 대웅제약 신규한 항진균성 트리아졸 유도체
WO2008082839A2 (en) 2006-12-29 2008-07-10 Abbott Laboratories Pim kinase inhibitors as cancer chemotherapeutics
BRPI0808523A2 (pt) 2007-03-01 2014-08-19 Novartis Vaccines & Diagnostic Inibidores de pim cinase e métodos de seu uso
BRPI0809998B8 (pt) 2007-04-03 2021-05-25 Array Biopharma Inc composto de imidazo[1,2-a] piridina como inibidores do receptor tirosina quinase, seus usos, seus processos de preparação e composições farmacêuticas
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 (es) 2007-06-13 2008-11-03 Incyte Corp Compuestos derivados de pirrolo [2,3-b]pirimidina, moduladores de quinasas jak; composicion farmaceutica; y uso en el tratamiento de enfermedades tales como cancer, psoriasis, artritis reumatoide, entre otras.
KR20120115413A (ko) 2007-07-11 2012-10-17 화이자 인코포레이티드 안구 건조증 치료용 약학 조성물 및 방법
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 (ru) 2008-02-04 2014-02-28 Меркьюри Терапьютикс, Инк. Модуляторы ampk (амф-активируемой протеинкиназы)
UY31679A1 (es) 2008-03-03 2009-09-30 Inhibidores de cinasa pim y metodos para su uso
SI2288610T1 (sl) 2008-03-11 2016-11-30 Incyte Holdings Corporation Derivati azetidina in ciklobutana kot inhibitorji jak
JP5384611B2 (ja) 2008-03-21 2014-01-08 ノバルティス アーゲー 新規ヘテロ環式化合物およびそれらの使用
US8344144B2 (en) 2008-06-18 2013-01-01 Merck Sharp & Dohme Corp. Inhibitors of Janus kinases
BRPI0914630A2 (pt) 2008-06-26 2019-09-24 Anterios Inc liberação dérmica
TWI461423B (zh) 2008-07-02 2014-11-21 Astrazeneca Ab 用於治療Pim激酶相關病狀及疾病之噻唑啶二酮化合物
FR2933409B1 (fr) 2008-07-03 2010-08-27 Centre Nat Rech Scient NOUVEAUX PYRROLO °2,3-a! CARBAZOLES ET LEUR UTILISATION COMME INHIBITEURS DES KINASES PIM
TWI496779B (zh) 2008-08-19 2015-08-21 Array Biopharma Inc 作為pim激酶抑制劑之***吡啶化合物
WO2010022081A1 (en) 2008-08-19 2010-02-25 Array Biopharma Inc. Triazolopyridine compounds as pim kinase inhibitors
JP4884570B2 (ja) 2008-08-20 2012-02-29 ファイザー・インク ピロロ[2,3−d]ピリミジン化合物
EP2342190A1 (en) 2008-09-02 2011-07-13 Novartis AG Bicyclic kinase inhibitors
PT2344474E (pt) 2008-09-02 2015-12-28 Novartis Ag Derivados de picolinamida como inibidores de cinase
BRPI0918846A2 (pt) 2008-09-02 2019-09-24 Novartis Ag inibidores de cinase heterocíclica
CL2009001884A1 (es) 2008-10-02 2010-05-14 Incyte Holdings Corp Uso de 3-ciclopentil-3-[4-(7h-pirrolo[2,3-d]pirimidin-4-il)-1h-pirazol-1-il)propanonitrilo, inhibidor de janus quinasa, y uso de una composición que lo comprende para el tratamiento del ojo seco.
CA2739466A1 (en) 2008-10-17 2010-04-22 Merck Frosst Canada Ltd. Azetidine derivatives as inhibitors of stearoyl-coenzyme a delta-9 desaturase
JOP20190230A1 (ar) 2009-01-15 2017-06-16 Incyte Corp طرق لاصلاح مثبطات انزيم jak و المركبات الوسيطة المتعلقة به
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
EA025520B1 (ru) 2009-05-22 2017-01-30 Инсайт Холдингс Корпорейшн N-(ГЕТЕРО)АРИЛПИРРОЛИДИНОВЫЕ ПРОИЗВОДНЫЕ ПИРАЗОЛ-4-ИЛ-ПИРРОЛО[2,3-d]ПИРИМИДИНОВ И ПИРРОЛ-3-ИЛ-ПИРРОЛО[2,3-d]ПИРИМИДИНОВ В КАЧЕСТВЕ ИНГИБИТОРОВ ЯНУС-КИНАЗЫ
MX2011012262A (es) 2009-05-22 2012-01-25 Incyte Corp 3-[4-(7h-pirrolo[2,3-d]pirimidin-4-il)-1h-pirazol-1-il] octano-o heptano-nitrilo como inhibidores de cinasas janus (jak).
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 (ja) 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−置換ピリジン−3−イル−カルボキサミド化合物及び使用方法
EP2305660A1 (en) 2009-09-25 2011-04-06 Almirall, S.A. New thiadiazole derivatives
PT2486041E (pt) 2009-10-09 2013-11-14 Incyte Corp Derivados hidroxilo, ceto e glucuronido de 3-(4-(7h-pirrolo[2,3-d]pirimidin-4-il)-1h-pirazol-1-il)-3-ciclopentil-propanonitrilo
MX2012004020A (es) 2009-10-20 2012-05-08 Cellzome Ltd Analogos de heterociclilo pirazolopirimidina como inhibidores de jak.
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 (zh) 2009-11-24 2016-10-12 奥尔德生物制药公司 Il-6抗体及其用途
EP2506852A4 (en) 2009-12-04 2013-06-19 Univ Texas INTERFERONTHERAPIES IN COMBINATION WITH BLOCKING OF STAT3 ACTIVATION
JP5739446B2 (ja) 2009-12-18 2015-06-24 ファイザー・インク ピロロ[2,3−d]ピリミジン化合物
CN102712640A (zh) 2010-01-12 2012-10-03 弗·哈夫曼-拉罗切有限公司 三环杂环化合物、其组合物和应用方法
JP2013518882A (ja) 2010-02-05 2013-05-23 ファイザー・インク JAK阻害剤としてのピロロ[2,3−d]ピリミジン尿素化合物
SA111320200B1 (ar) 2010-02-17 2014-02-16 ديبيوفارم اس ايه مركبات ثنائية الحلقة واستخداماتها كمثبطات c-src/jak مزدوجة
EA023444B1 (ru) 2010-02-18 2016-06-30 Инсайт Холдингс Корпорейшн Циклобутановые и метилциклобутановые производные, композиции на их основе и способы их применения
SI3354652T1 (sl) * 2010-03-10 2020-08-31 Incyte Holdings Corporation Derivati piperidin-4-il azetidina kot inhibitorji JAK1
KR20130094710A (ko) 2010-04-14 2013-08-26 어레이 바이오파마 인크. Jak 키나아제의 억제제로서 5,7-치환된-이미다조[1,2-c]피리미딘
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 (pt) * 2010-11-19 2019-03-06 Incyte Corporation pirrolopiridina ciclobutil substituída e derivados de pirrolopirimidina derivativos como inibidores de jak
US9034884B2 (en) * 2010-11-19 2015-05-19 Incyte Corporation Heterocyclic-substituted pyrrolopyridines and pyrrolopyrimidines as JAK inhibitors
CN103370068A (zh) 2010-12-03 2013-10-23 Ym生物科学澳大利亚私人有限公司 Jak-2 介导的病症的治疗
WO2012112847A1 (en) * 2011-02-18 2012-08-23 Novartis Pharma Ag mTOR/JAK INHIBITOR COMBINATION THERAPY
CN102247368B (zh) 2011-05-19 2013-05-29 安徽永生堂药业有限责任公司 一种复方阿伐斯汀缓释片及其制备方法
CN102218042A (zh) 2011-05-26 2011-10-19 青岛黄海制药有限责任公司 富马酸喹硫平组合物的缓释片剂及其制备方法
MY165963A (en) 2011-06-20 2018-05-18 Incyte Holdings Corp Azetidinyl phenyl, pyridyl or pyrazinyl carboxamide derivatives 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
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
EP2741747A1 (en) * 2011-08-10 2014-06-18 Novartis Pharma AG JAK P13K/mTOR COMBINATION THERAPY
TW201313721A (zh) 2011-08-18 2013-04-01 Incyte Corp 作為jak抑制劑之環己基氮雜環丁烷衍生物
UA111854C2 (uk) * 2011-09-07 2016-06-24 Інсайт Холдінгс Корпорейшн Способи і проміжні сполуки для отримання інгібіторів jak
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 (ru) 2012-07-27 2015-05-29 Рациофарм Гмбх Пероральные дозированные формы для модифицированного высвобождения, содержащие руксолитиниб
CN102772384A (zh) 2012-08-07 2012-11-14 四川百利药业有限责任公司 一种盐酸米诺环素缓释片及其制备方法
JP2015526520A (ja) 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 Itk阻害剤としてのベンズイミダゾール誘導体
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 (ru) 2013-03-06 2018-09-28 Инсайт Холдингс Корпорейшн Способы и промежуточные соединения при получении ингибитора jak
PE20160126A1 (es) 2013-05-17 2016-02-24 Incyte Corp Derivados del bipirazol como inhibidores jak
ES2792549T3 (es) 2013-08-07 2020-11-11 Incyte Corp Formas de dosificación de liberación sostenida para un inhibidor de JAK1
CN105555313A (zh) 2013-08-20 2016-05-04 因赛特公司 在c-反应蛋白水平较高的实体肿瘤患者中的存活益处
CN106456773A (zh) 2014-02-28 2017-02-22 因赛特公司 用于治疗骨髓增生异常综合征的jak1抑制剂
KR20220066179A (ko) 2014-04-08 2022-05-23 인사이트 코포레이션 Jak 및 pi3k 억제제 조합에 의한 b-세포 악성종양의 치료
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

Also Published As

Publication number Publication date
ES2611588T3 (es) 2017-05-09
CY1118724T1 (el) 2017-07-12
LTPA2017012I1 (lt) 2017-05-10
US20140018374A1 (en) 2014-01-16
ECSP12008540A (es) 2012-04-30
HUE030418T2 (hu) 2017-05-29
ECSP088540A (es) 2008-07-30
LU92137I9 (hu) 2019-01-04
SI2426129T1 (sl) 2017-02-28
PT2343299E (pt) 2016-02-26
RS55632B1 (sr) 2017-06-30
EP2343298A1 (en) 2011-07-13
LU92137I2 (fr) 2014-01-18
DK3184526T3 (en) 2019-01-14
TWI468162B (zh) 2015-01-11
ES2543904T3 (es) 2015-08-25
JP5710430B2 (ja) 2015-04-30
SI2455382T1 (sl) 2017-03-31
ES2700433T3 (es) 2019-02-15
EP2343299B1 (en) 2015-11-04
PT1966202E (pt) 2012-01-03
CY1118607T1 (el) 2017-07-12
CN101448826A (zh) 2009-06-03
US8415362B2 (en) 2013-04-09
JP2011252024A (ja) 2011-12-15
US9079912B2 (en) 2015-07-14
ES2612196T3 (es) 2017-05-12
EP1966202A1 (en) 2008-09-10
KR101324737B1 (ko) 2013-11-05
HRP20160112T1 (hr) 2016-02-26
IL248938A0 (en) 2017-01-31
EP2455382A1 (en) 2012-05-23
NZ569015A (en) 2011-06-30
US11744832B2 (en) 2023-09-05
HRP20170090T1 (hr) 2017-03-24
MY162590A (en) 2017-06-30
BRPI0619817B8 (pt) 2021-05-25
EA200870048A1 (ru) 2009-02-27
CR20130506A (es) 2013-10-30
ES2373688T3 (es) 2012-02-07
KR20110137406A (ko) 2011-12-22
CY1121202T1 (el) 2020-05-29
EP2426129B1 (en) 2016-11-02
US20220395506A1 (en) 2022-12-15
ES2561507T3 (es) 2016-02-26
CA2632466A1 (en) 2007-06-21
EP2455382B1 (en) 2016-10-26
HK1160137A1 (en) 2012-08-10
IL231992A0 (en) 2014-05-28
US20190125750A1 (en) 2019-05-02
ES2543903T3 (es) 2015-08-25
HUE041382T2 (hu) 2019-05-28
BRPI0619817B1 (pt) 2020-03-17
FR17C1013I1 (fr) 2017-06-02
DK2426129T3 (en) 2017-01-16
RS58113B1 (sr) 2019-02-28
TW200728275A (en) 2007-08-01
US20200338077A1 (en) 2020-10-29
UA116187C2 (uk) 2018-02-26
US7598257B2 (en) 2009-10-06
LT2474545T (lt) 2017-02-27
CN103214484A (zh) 2013-07-24
EA019504B1 (ru) 2014-04-30
BRPI0619817A8 (pt) 2018-01-23
EP2474545B1 (en) 2016-11-09
DK2474545T3 (en) 2017-01-23
HUS1700017I1 (hu) 2017-05-29
CY2017015I2 (el) 2017-09-13
EA201200132A1 (ru) 2017-01-30
RS52101B (en) 2012-06-30
PL2474545T3 (pl) 2017-04-28
CY1118506T1 (el) 2017-07-12
KR20120120462A (ko) 2012-11-01
US10398699B2 (en) 2019-09-03
EP2348023B9 (en) 2017-03-08
HK1171023A1 (zh) 2013-03-15
EA035795B1 (ru) 2020-08-11
EP2343298B9 (en) 2020-05-06
SI2474545T1 (sl) 2017-03-31
EP3838903A1 (en) 2021-06-23
KR20120120463A (ko) 2012-11-01
PL2343299T3 (pl) 2016-09-30
PL2426129T3 (pl) 2017-04-28
JP6138865B2 (ja) 2017-05-31
HK1160115A1 (zh) 2012-08-10
HRP20170200T1 (hr) 2017-04-07
HRP20150837T2 (hr) 2017-04-07
CN103214484B (zh) 2016-07-06
HRP20150837T1 (hr) 2015-09-11
UA98449C2 (en) 2012-05-25
EP3838903B1 (en) 2023-11-22
RS54181B1 (en) 2015-12-31
DK1966202T3 (da) 2012-01-16
BRPI0619817A2 (pt) 2011-11-22
EP2348023B1 (en) 2015-05-06
CN103254190B (zh) 2016-12-07
CY2013006I2 (el) 2015-10-07
US20150238492A1 (en) 2015-08-27
TW201831490A (zh) 2018-09-01
KR101218214B1 (ko) 2013-01-04
PL2455382T3 (pl) 2017-04-28
US9814722B2 (en) 2017-11-14
AU2006326548A1 (en) 2007-06-21
NZ778831A (en) 2022-12-23
KR20080079677A (ko) 2008-09-01
ES2867505T3 (es) 2021-10-20
PT3184526T (pt) 2018-12-19
TWI410407B (zh) 2013-10-01
US8933086B2 (en) 2015-01-13
US20140243360A1 (en) 2014-08-28
US20170071947A1 (en) 2017-03-16
EP1966202B1 (en) 2011-09-21
PL1966202T3 (pl) 2012-02-29
HUE025173T2 (hu) 2016-01-28
US20180338978A1 (en) 2018-11-29
EP3184526A1 (en) 2017-06-28
HRP20110903T1 (hr) 2012-01-31
EP2348023A1 (en) 2011-07-27
US9662335B2 (en) 2017-05-30
RS55634B1 (sr) 2017-06-30
IL231992A (en) 2016-11-30
EP2343299B9 (en) 2017-03-08
EP2474545A1 (en) 2012-07-11
HK1160111A1 (en) 2012-08-10
RS54683B1 (en) 2016-08-31
US20110224157A1 (en) 2011-09-15
CY1116574T1 (el) 2018-03-07
IL192019A0 (en) 2008-12-29
KR101216055B1 (ko) 2012-12-27
EP3184526B1 (en) 2018-10-03
HRP20170162T1 (hr) 2017-03-24
US20140005210A1 (en) 2014-01-02
RS55576B1 (sr) 2017-05-31
DK2455382T3 (da) 2017-01-02
US20160272648A1 (en) 2016-09-22
EA036785B1 (ru) 2020-12-21
US20160346286A1 (en) 2016-12-01
KR101391900B1 (ko) 2014-05-02
PL2348023T3 (pl) 2015-11-30
EA201200132A8 (ru) 2018-10-31
TW201704235A (zh) 2017-02-01
CN103214483A (zh) 2013-07-24
HRP20181912T1 (hr) 2019-03-22
US8541425B2 (en) 2013-09-24
EA201691294A2 (ru) 2018-11-30
ME01312B (me) 2013-12-20
US20110223210A1 (en) 2011-09-15
US20090181959A1 (en) 2009-07-16
US20100022522A1 (en) 2010-01-28
WO2007070514A1 (en) 2007-06-21
AU2006326548B2 (en) 2012-04-05
SI1966202T1 (sl) 2012-01-31
EP2426129A1 (en) 2012-03-07
US9206187B2 (en) 2015-12-08
LT2455382T (lt) 2017-02-10
ATE525374T1 (de) 2011-10-15
US10639310B2 (en) 2020-05-05
DK2348023T5 (da) 2017-05-15
LT2426129T (lt) 2017-02-10
SI2348023T1 (sl) 2015-10-30
MY159449A (en) 2017-01-13
PL3184526T3 (pl) 2019-04-30
JP5876026B2 (ja) 2016-03-02
HUE032337T2 (hu) 2017-09-28
US8530485B2 (en) 2013-09-10
CY2013006I1 (el) 2015-10-07
JP2009519340A (ja) 2009-05-14
CR10065A (es) 2008-07-10
ES2612489T3 (es) 2017-05-17
ES2543904T9 (es) 2017-05-29
PT2348023E (pt) 2015-09-15
RS54181B9 (sr) 2020-01-31
CY1112762T1 (el) 2015-10-07
EP2343298B1 (en) 2015-05-06
JP2015193641A (ja) 2015-11-05
TWI664182B (zh) 2019-07-01
US8946245B2 (en) 2015-02-03
EP3466953A1 (en) 2019-04-10
FR17C1013I2 (fr) 2018-05-04
ZA200805165B (en) 2012-05-30
TWI630207B (zh) 2018-07-21
SI3184526T1 (sl) 2019-03-29
CN103214483B (zh) 2014-12-17
HUE030235T2 (hu) 2017-04-28
DK2348023T3 (da) 2015-06-22
HK1124840A1 (en) 2009-07-24
SI2343299T1 (sl) 2016-06-30
SG179430A1 (en) 2012-04-27
PT2455382T (pt) 2017-01-31
EP2343299A1 (en) 2011-07-13
EP3466953B1 (en) 2021-02-03
SG10201506912RA (en) 2015-10-29
TWI553008B (zh) 2016-10-11
US20070135461A1 (en) 2007-06-14
TW201522344A (zh) 2015-06-16
CY2017015I1 (el) 2017-09-13
US11331320B2 (en) 2022-05-17
DK2343299T3 (en) 2016-01-18
TW201240663A (en) 2012-10-16
JP5017278B2 (ja) 2012-09-05
MX346183B (es) 2017-03-10
TW201434835A (zh) 2014-09-16
IL192019A (en) 2014-04-30
PT2474545T (pt) 2017-02-14
SG10202003901UA (en) 2020-05-28
PT2426129T (pt) 2017-02-10
CN103254190A (zh) 2013-08-21
JP2014051531A (ja) 2014-03-20
AR057995A1 (es) 2008-01-09
CA2632466C (en) 2013-09-24
LT3184526T (lt) 2019-02-25
US9974790B2 (en) 2018-05-22

Similar Documents

Publication Publication Date Title
US11744832B2 (en) Heteroaryl substituted pyrrolo[2,3-b]pyridines and pyrrolo[2,3-b]pyrimidines as Janus kinase inhibitors
US20130137681A1 (en) HETEROARYL SUBSTITUTED PYRROLO[2,3-b]PYRIDINES AND PYRROLO[2,3-b]PYRIMIDINES AS JANUS KINASE INHIBITORS
AU2017200685B2 (en) Heteroaryl substituted pyrrolo[2,3-b]pyridines and pyrrolo[2,3-b]pyrimidines as Janus kinase inhibitors