US20120053345A1 - Indazole Compounds - Google Patents

Indazole Compounds Download PDF

Info

Publication number
US20120053345A1
US20120053345A1 US13/198,363 US201113198363A US2012053345A1 US 20120053345 A1 US20120053345 A1 US 20120053345A1 US 201113198363 A US201113198363 A US 201113198363A US 2012053345 A1 US2012053345 A1 US 2012053345A1
Authority
US
United States
Prior art keywords
phenyl
optionally substituted
group
thienyl
och
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US13/198,363
Inventor
Anna M. Ericson
Andrew Burchat
Kristine E. Frank
David J. Calderwood
Lily K. Abbott
Maria A. Argiriadi
David W. Borhani
Kevin P. Cusack
Richard W. Dixon
Thomas D. Gordon
Kelly D. Mullen
Robert V. Talanian
Xiaoyun Wu
Xiaolei ZHANG
Lu Wang
Biqin Li
Claude E. Barberis
Neil Wishart
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Abbott Laboratories
Original Assignee
Abbott Laboratories
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Abbott Laboratories filed Critical Abbott Laboratories
Priority to US13/198,363 priority Critical patent/US20120053345A1/en
Publication of US20120053345A1 publication Critical patent/US20120053345A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/02Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D409/04Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/14Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing three or more hetero rings

Definitions

  • Protein phosphorylation is important for the regulation of many cellular processes including cell cycle progression and division, signal transduction, and apoptosis.
  • the phosphorylation is usually a transfer reaction of the terminal phosphate group from ATP to the protein substrate.
  • the specific structure in the target substrate to which the phosphate is transferred is a tyrosine, serine or threonine residue. Since these amino acid residues are the target structures for the phosphoryl transfer, and since most kinases target either tyrosine or both serine and threonine, these protein kinase enzymes are commonly referred to as tyrosine kinases or serine/threonine (S/T) kinases.
  • the phosphorylation reactions, and counteracting phosphatase reactions, on the tyrosine, serine and threonine residues are involved in many cellular processes that underlie responses to diverse intracellular signals, regulation of cellular functions, and activation or deactivation of cellular processes.
  • a cascade of protein kinases often function in intracellular signal transduction. Protein kinases can be found integrated into the plasma membrane, as cytoplasmic enzymes or localized in the nucleus, often as components of enzyme complexes. In many instances, these protein kinases are an essential element of enzyme and structural protein complexes that determine where and when a cellular process occurs within a cell. Given the importance and diversity of protein kinase function, it is not surprising that phosphorylation events are required in cellular processes associated with many diseases such as cancer, diabetes, inflammation, and hypertension.
  • the identification of effective small molecules that specifically inhibit protein kinases involved in abnormal or inappropriate cell proliferation, signaling, differentiation, protein production, or metabolism is therefore desirable.
  • the identification of methods and compounds that specifically inhibit the function of kinases that are involved in immune modulation or proliferative disorders is particularly desirable.
  • the present invention provides novel compounds that inhibit one or more receptor, or non-receptor, tyrosine or S/T kinase.
  • the present invention provides a compound of Formula (I)
  • R 3 is selected from the group consisting of I, Br, COOH, NH 2 , thienyl, pyridinyl, pyrrolyl, —C(O)—NH—CH—(CH 2 OH) 2 , —C(O)—NH—CH 2 —X 100 ,
  • the invention provides compounds or pharmaceutically acceptable salts thereof according to any of the foregoing inventions wherein
  • the invention provides compounds or pharmaceutically acceptable salts thereof according to any of the foregoing inventions wherein R 1 is H or pyrimidinyl substituted with NH 2 ;
  • the invention provides compounds or pharmaceutically acceptable salts thereof according to any of the foregoing inventions wherein R 1 and R 4 are H;
  • the invention provides compounds or pharmaceutically acceptable salts thereof according to any of the foregoing inventions wherein
  • the invention provides compounds or pharmaceutically acceptable salts thereof according to any of the foregoing inventions wherein R 1 , R 3 , R 4 and R 6 are H;
  • the invention provides compounds or pharmaceutically acceptable salts thereof according to any of the foregoing inventions wherein R 1 , R 3 , R 4 and R 6 are H;
  • the invention provides compounds or pharmaceutically acceptable salts thereof according to any of the foregoing inventions wherein R 1 , R 3 , R 4 and R 6 are H;
  • the invention provides compounds or pharmaceutically acceptable salts thereof according to any of the foregoing inventions wherein R 1 , R 3 , R 4 and R 6 are H;
  • Protein kinases are a broad and diverse class, of over 500 enzymes, that include growth factors receptors, signal transduction intermediates, apoptosis related kinases and cyclin dependent kinases. Many can function as oncogenes. They are responsible for the transfer of a phosphate group to specific tyrosine, serine or threonine amino acid residues, and are broadly classified as tyrosine and S/T kinases as a result of their substrate specificity.
  • S/T kinases are a large sub-family of protein kinases that specifically transfer a phosphate group to a terminal hydroxyl moiety of specific serine or threonine residues (Hanks et al., (1988) Science, 241: 42-52).
  • a number of S/T kinase family members are involved in inflammatory signaling, tumor growth or cellular transformation.
  • the mitogen-activated protein kinases are S/T kinases that act as intermediates within the signaling cascades of Toll like receptors (TLRs), such as TLR4, growth/survival factors, such as EGF, and death receptors, such as the TNF receptor.
  • MAPKs such as extracellular signal-regulated kinases (ERK1-2), p38a, c-Jun N-terminal kinase (INK) or MAPKAP-K2 (MK2) have been shown to transduce signaling in cells, such as macrophages, resulting in the production and secretion of pro-inflammatory cytokines, such as TNF.
  • ERK1-2 extracellular signal-regulated kinases
  • p38a p38a
  • INK c-Jun N-terminal kinase
  • MK2 MAPKAP-K2
  • TPL-2 is a S/T kinase which is homologous to a subfamily of kinases called MAP kinase kinase kinases (MAP3K) in its catalytic domain (Salmeron, et al., (1996) EMBO J., 15, 817-826) and is >90% identical to the proto-oncogene product of human COT (Aoki et al., (1993) J. Biol. Chem., 268, 22723-22732).
  • MAP3K MAP kinase kinase kinases
  • TPL-2 was originally identified, in a C-terminally deleted form, as the product of an oncogene associated with Moloney murine leukemia virus-induced T cell lymphomas in rats (Patriotis, et al., (1993) Proc. Natl. Acad. Sci. USA 90, 2251-2255). TPL-2 is also highly homologous to the kinase NIK, which has been shown to regulate the inducible degradation of I ⁇ B- ⁇ (Malinin et al., (1997) Nature, 385, 540-544; WO 97/37016; May and Ghosh, (1998) Immunol. Today, 19, 80-88).
  • TPL-2 is essential for the activation of a MAP2K (MEK1-2), which in turn activate a MAPK (extracellular signal-regulated kinase, ERK1-2) in macrophages stimulated by TLR agonists, such as lipopolysachharide (LPS).
  • MAPK extracellular signal-regulated kinase
  • ERK1-2 extracellular signal-regulated kinase
  • TLR agonists such as lipopolysachharide (LPS).
  • LPS lipopolysachharide
  • TPL-2 plays a crucial role in the regulation of LPS-induced TNF, IL-1 ⁇ and COX-2 induced prostaglandin-E2 production in macrophages (Tsichlis et al, (2000), Cell, 103, 1071; Tsichlis et al, (2002), EMBO J, 21, 4831-4840).
  • COT/TPL-2 in various tumors (Tsanisi et al., (2000), Int J Mol Med, 5, 583) and the defect in TNF production observed in COT knockout mice (Tsichlis et al, (2000), Cell, 103, 1071) suggests that inhibition of COT may be a useful approach in the treatment of cancer, inflammation or other diseases mediated by pro-inflammatory cytokines.
  • MK2 (MAPKAP-K2) is an S/T kinase critically involved in inflammatory processes. MK2 is a substrate for the MAPK p38 (Stokoe et al., (1992), EMBO J., 11, 3985-3994; Ben-Levy et al., (1995), EMBO J., 14, 5920-5930). Activation of MK2 in immune cells results in an array of cellular responses including cytokine production, proliferation and activation. Knockout mice defective in MK2 production are healthy and fertile but fail to produce cytokines such as tumor necrosis factor (TNF) in response to inflammatory stimuli (Kotlyarov et al., (1999), Nat.
  • TNF tumor necrosis factor
  • MK2 may alter gene expression by phosphorylation of mRNA-binding proteins (Winzen et al., (1999), EMBO J., 18, 4969-4980; Lasa et al., (2000), Mol. Cell. Biol., 20, 4265-4274; Rousseau et al., (2002), EMBO J., 21, 6505-6514; Bollig et al., (2003), Biochem. Biophys. Res. Commun, 301, 665-670; Tran et al., (2003), Mol. Cell. Biol., 23, 7177-7188.), Chrestensen, C. A. et al. J. Biol. Chem.
  • MK2 knockouts The defect in TNF production in MK2 knockouts suggests that the antiinflammatory effect of p38 MAPK inhibitors may be largely due to blockade of activation of MK2 Inhibitors of MK2 may be effective treatments of inflammation or other diseases mediated by pro-inflammatory cytokines.
  • PTKs Protein tyrosine kinases
  • endothelial-cell specific receptor PTKs such as KDR and Tie-2 mediate the angiogenic process, and are thus involved in supporting the progression of cancers and other diseases involving inappropriate vascularization (e.g., diabetic retinopathy, choroidal neovascularization due to age-related macular degeneration, psoriasis, arthritis, retinopathy of prematurity, and infantile hemangiomas).
  • inappropriate vascularization e.g., diabetic retinopathy, choroidal neovascularization due to age-related macular degeneration, psoriasis, arthritis, retinopathy of prematurity, and infantile hemangiomas.
  • Tyrosine kinases can be of the receptor-type (having extracellular, transmembrane and intracellular domains) or the non-receptor type (being wholly intracellular).
  • RTKs Receptor Tyrosine Kinases
  • the RTKs comprise a large family of transmembrane receptors with diverse biological activities. At present, at least nineteen (19) distinct RTK subfamilies have been identified.
  • the receptor tyrosine kinase (RTK) family includes receptors that are crucial for the growth and differentiation of a variety of cell types (Yarden and Ullrich, Ann. Rev. Biochem. 57:433-478, 1988; Ullrich and Schlessinger, Cell 61:243-254, 1990).
  • RTKs The intrinsic function of RTKs is activated upon ligand binding, which results in phosphorylation of the receptor and multiple cellular substrates, and subsequently in a variety of cellular responses (Ullrich & Schlessinger, 1990, Cell 61:203-212).
  • receptor tyrosine kinase mediated signal transduction is 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 trans-phosphorylation.
  • 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 (e.g., cell division, differentiation, metabolic effects, and changes in the extracellular microenvironment; see Schlessinger and Ullrich, 1992, Neuron 9:1-20).
  • Non-Receptor Tyrosine Kinases represent a collection of cellular enzymes that lack extracellular and transmembrane sequences. Over twenty-four individual non-receptor tyrosine kinases, comprising eleven (11) subfamilies (Src, Frk, Btk, Csk, Abl, Zap70, Fes/Fps, Fak, Jak, Ack and LIMK) have been identified. The Src subfamily of non-receptor tyrosine kinases is comprised of the largest number of PTKs and include Src, Yes, Fyn, Lyn, Lck, Blk, Hck, Fgr and Yrk.
  • kinases whether a receptor or non-receptor tyrosine kinase or a S/T kinase have been found to be involved in cellular signaling pathways involved in numerous pathogenic conditions, including immunomodulation, inflammation, or proliferative disorders such as cancer.
  • the invention provides a method for inhibiting COT in a human subject suffering from a disorder in which COT activity is detrimental, comprising administering to the human subject a compound of Formula (I) such that COT activity in the human subject is inhibited and treatment is achieved.
  • the invention provides a method for inhibiting MK2 in a human subject suffering from a disorder in which MK2 activity is detrimental, comprising administering to the human subject a compound of Formula (I) such that MK2 activity in the human subject is inhibited and treatment is achieved.
  • a compound of formula (I) or a salt thereof or pharmaceutical compositions containing a therapeutically effective amount thereof is useful in the treatment of a disorder selected from the group comprising rheumatoid arthritis, osteoarthritis, juvenile chronic arthritis, Lyme arthritis, psoriatic arthritis, reactive arthritis, and septic arthritis, spondyloarthropathy, systemic lupus erythematosus, Crohn's disease, ulcerative colitis, inflammatory bowel disease, insulin dependent diabetes mellitus, thyroiditis, asthma, allergic diseases, psoriasis, dermatitis scleroderma, graft versus host disease, organ transplant rejection (including but not limited to bone marrow and solid organ rejection), acute or chronic immune disease associated with organ transplantation, sarcoidosis, atherosclerosis, disseminated intravascular coagulation, Kawasaki's disease, Grave's disease, nephrotic syndrome, chronic fatigue syndrome, Wegener's granulomatosis, Henoch-
  • such compounds may be useful in the treatment of disorders such as edema, ascites, effusions, and exudates, including for example macular edema, cerebral edema, acute lung injury, adult respiratory distress syndrome (ARDS), proliferative disorders such as restenosis, fibrotic disorders such as hepatic cirrhosis and atherosclerosis, mesangial cell proliferative disorders such as glomerulonephritis, diabetic nephropathy, malignant nephrosclerosis, thrombotic microangiopathy syndromes, and glomerulopathies, myocardial angiogenesis, coronary and cerebral collaterals, ischemic limb angiogenesis, ischemia/reperfusion injury, peptic ulcer Helicobacter related diseases, virally-induced angiogenic disorders, Crow-Fukase syndrome (POEMS), preeclampsia, menometrorrhagia, cat scratch fever, rubeosis, neovascular glaucoma and
  • these compounds can be used as active agents against solid tumors, malignant ascites, von Hippel Lindau disease, hematopoietic cancers and hyperproliferative disorders such as thyroid hyperplasia (especially Grave's disease), and cysts (such as hypervascularity of ovarian stroma characteristic of polycystic ovarian syndrome (Stein-Leventhal syndrome) and polycystic kidney disease since such diseases require a proliferation of blood vessel cells for growth and/or metastasis.
  • thyroid hyperplasia especially Grave's disease
  • cysts such as hypervascularity of ovarian stroma characteristic of polycystic ovarian syndrome (Stein-Leventhal syndrome) and polycystic kidney disease since such diseases require a proliferation of blood vessel cells for growth and/or metastasis.
  • Compounds of formula (I) of the invention can be used alone or in combination with another therapeutic agent to treat such diseases.
  • an additional agent e.g., a therapeutic agent, said additional agent being selected by the skilled artisan for its intended purpose.
  • the additional agent can be a therapeutic agent art-recognized as being useful to treat the disease or condition being treated by the compound of the present invention.
  • the additional agent also can be an agent that imparts a beneficial attribute to the therapeutic composition e.g., an agent which effects the viscosity of the composition.
  • the combinations that are to be included within this invention are those combinations useful for their intended purpose.
  • the agents set forth below are illustrative for purposes and not intended to be limited.
  • the combinations, which are part of this invention can be the compounds of the present invention and at least one additional agent selected from the lists below.
  • the combination can also include more than one additional agent, e.g., two or three additional agents if the combination is such that the formed composition can perform its intended function.
  • Preferred combinations are non-steroidal anti-inflammatory drug(s) also referred to as NSAIDS which include drugs like ibuprofen.
  • Other preferred combinations are corticosteroids including prednisolone; the well known side-effects of steroid use can be reduced or even eliminated by tapering the steroid dose required when treating patients in combination with the anti-IL-18 antibodies of this invention.
  • Non-limiting examples of therapeutic agents for rheumatoid arthritis with which a compound of formula (I) of the invention can be combined include the following: cytokine suppressive anti-inflammatory drug(s) (CSAIDs); antibodies to or antagonists of other human cytokines or growth factors, for example, TNF, LT, IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-12, IL-15, IL-16, IL-21, IL-23, interferons, EMAP-II, GM-CSF, FGF, and PDGF.
  • CSAIDs cytokine suppressive anti-inflammatory drug
  • Antibodies of the invention, or antigen binding portions thereof, can be combined with antibodies to cell surface molecules such as CD2, CD3, CD4, CD8, CD25, CD28, CD30, CD40, CD45, CD69, CD80 (B7.1), CD86 (B7.2), CD90, CTLA or their ligands including CD154 (gp39 or CD40L).
  • cell surface molecules such as CD2, CD3, CD4, CD8, CD25, CD28, CD30, CD40, CD45, CD69, CD80 (B7.1), CD86 (B7.2), CD90, CTLA or their ligands including CD154 (gp39 or CD40L).
  • Preferred combinations of therapeutic agents may interfere at different points in the autoimmune and subsequent inflammatory cascade; preferred examples include TNF antagonists like chimeric, humanized or human TNF antibodies, D2E7 (HUMIRATM), (PCT Publication No. WO 97/29131), CA2 (REMICADETM), CDP 571, and soluble p55 or p75 TNF receptors, derivatives, thereof, (p75TNFR1gG (ENBRELTM) or p55TNFR1gG (Lenercept), and also TNF ⁇ converting enzyme (TACE) inhibitors; similarly IL-1 inhibitors (Interleukin-1-converting enzyme inhibitors, IL-1RA etc.) may be effective for the same reason.
  • TNF antagonists like chimeric, humanized or human TNF antibodies, D2E7 (HUMIRATM), (PCT Publication No. WO 97/29131), CA2 (REMICADETM), CDP 571, and soluble p55 or p75 TNF receptors, derivatives, thereof, (
  • Yet another preferred combination are other key players of the autoimmune response which may act parallel to, dependent on or in concert with IL-18 function; especially preferred are IL-12 antagonists including IL-12 antibodies or soluble IL-12 receptors, or IL-12 binding proteins. It has been shown that IL-12 and IL-18 have overlapping but distinct functions and a combination of antagonists to both may be most effective. Yet another preferred combination are non-depleting anti-CD4 inhibitors. Yet other preferred combinations include antagonists of the co-stimulatory pathway CD80 (B7.1) or CD86 (B7.2) including antibodies, soluble receptors or antagonistic ligands.
  • a compound of formula (I) of the invention may also be combined with agents, such as methotrexate, 6-MP, azathioprine sulphasalazine, mesalazine, olsalazine chloroquinine/hydroxychloroquine, pencillamine, aurothiomalate (intramuscular and oral), azathioprine, cochicine, corticosteroids (oral, inhaled and local injection), beta-2 adrenoreceptor agonists (salbutamol, terbutaline, salmeteral), xanthines (theophylline, aminophylline), cromoglycate, nedocromil, ketotifen, ipratropium and oxitropium, cyclosporin, FK506, rapamycin, mycophenolate mofetil, leflunomide, NSAIDs, for example, ibuprofen, corticosteroids such as prednisolone, phosphodieste
  • IRAK family NIK, IKK family, p38 or other MAP kinase inhibitors
  • IL-1 ⁇ converting enzyme inhibitors IL-1 ⁇ converting enzyme inhibitors
  • TACE TNF ⁇ converting enzyme
  • T-cell signalling inhibitors such as kinase inhibitors, metalloproteinase inhibitors, sulfasalazine, azathioprine, 6-mercaptopurines, angiotensin converting enzyme inhibitors, soluble cytokine receptors and derivatives thereof (e.g.
  • soluble p55 or p75 TNF receptors and the derivatives p75TNFRIgG EnbrelTM and p55TNFRIgG (Lenercept)
  • sIL-1RI sIL-1RII
  • sIL-6R antiinflammatory cytokines
  • IL-4, IL-10, IL-11, IL-13 and TGF ⁇ celecoxib, folic acid, hydroxychloroquine sulfate, rofecoxib, etanercept, infliximab, naproxen, valdecoxib, sulfasalazine, methylprednisolone, meloxicam, methylprednisolone acetate, gold sodium thiomalate, aspirin, triamcinolone acetonide, propoxyphene napsylate/apap, folate, nabumetone, diclofenac, piroxicam, etodolac, diclofenac sodium, oxaprozin, oxycodone HCl, hydrocodone bitartrate/apap, diclofenac sodium/misoprostol, fentanyl, anakinra, human recombinant, tramadol HCl, salsalate
  • Non-limiting examples of therapeutic agents for inflammatory bowel disease with which a compound of formula (I) of the invention can be combined include the following: budenoside; epidermal growth factor; corticosteroids; cyclosporin, sulfasalazine; aminosalicylates; 6-mercaptopurine; azathioprine; metronidazole; lipoxygenase inhibitors; mesalamine; olsalazine; balsalazide; antioxidants; thromboxane inhibitors; IL-1 receptor antagonists; anti-IL-1 ⁇ monoclonal antibodies; anti-IL-6 monoclonal antibodies; growth factors; elastase inhibitors; pyridinyl-imidazole compounds; antibodies to or antagonists of other human cytokines or growth factors, for example, TNF, LT, IL-1, IL-2, IL-6, IL-7, IL-8, IL-12, IL-15, IL-16, EMAP-II,
  • IL-1 ⁇ converting enzyme inhibitors IL-1 ⁇ converting enzyme inhibitors
  • TNF ⁇ converting enzyme inhibitors T-cell signalling inhibitors such as kinase inhibitors; metalloproteinase inhibitors; sulfasalazine; azathioprine; 6-mercaptopurines; angiotensin converting enzyme inhibitors; soluble cytokine receptors and derivatives thereof (e.g. soluble p55 or p75 TNF receptors, sIL-1RI, sIL-1RII, sIL-6R) and antiinflammatory cytokines (e.g. IL-4, IL-10, IL-11, IL-13 and TGF ⁇ ).
  • IL-4, IL-10, IL-11, IL-13 and TGF ⁇ antiinflammatory cytokines
  • TNF antagonists for example, anti-TNF antibodies, D2E7 (PCT Publication No. WO 97/29131; HUMIRATM), CA2 (REMICADETM), CDP 571, TNFR-Ig constructs, (p75TNFRIgG (ENBRELTM) and p55TNFRIgG (LENERCEPTTM)) inhibitors and PDE4 inhibitors.
  • a compound of formula (I) can be combined with corticosteroids, for example, budenoside and dexamethasone; sulfasalazine, 5-aminosalicylic acid; olsalazine; and agents which interfere with synthesis or action of proinflammatory cytokines such as IL-1, for example, IL-1 ⁇ converting enzyme inhibitors and IL-1ra; T cell signaling inhibitors, for example, tyrosine kinase inhibitors 6-mercaptopurines; IL-11; mesalamine; prednisone; azathioprine; mercaptopurine; infliximab; methylprednisolone sodium succinate; diphenoxylate/atrop sulfate; loperamide hydrochloride; methotrexate; omeprazole; folate; ciprofloxacin/dextrose-water; hydrocodone bitartrate/apap; tetracycline hydrochloride; fluo
  • Non-limiting examples of therapeutic agents for multiple sclerosis with which a compound of formula (I) can be combined include the following: corticosteroids; prednisolone; methylprednisolone; azathioprine; cyclophosphamide; cyclosporine; methotrexate; 4-aminopyridine; tizanidine; interferon- ⁇ 1a (AVONEX; Biogen); interferon- ⁇ 1b (BETASERON; Chiron/Berlex); interferon ⁇ -n3) (Interferon Sciences/Fujimoto), interferon- ⁇ (Alfa Wassermann/J&J), interferon ⁇ 1A-IF (Serono/Inhale Therapeutics), Peginterferon ⁇ 2b (Enzon/Schering-Plough), Copolymer 1 (Cop-1; COPAXONE; Teva Pharmaceutical Industries, Inc.); hyperbaric oxygen; intravenous immunoglobulin; clabribine; antibodies to or antagonists of
  • a compound of formula (I) can be combined with antibodies to cell surface molecules such as CD2, CD3, CD4, CD8, CD19, CD20, CD25, CD28, CD30, CD40, CD45, CD69, CD80, CD86, CD90 or their ligands.
  • cell surface molecules such as CD2, CD3, CD4, CD8, CD19, CD20, CD25, CD28, CD30, CD40, CD45, CD69, CD80, CD86, CD90 or their ligands.
  • a compound of formula (I) may also be combined with agents, such as methotrexate, cyclosporine, FK506, rapamycin, mycophenolate mofetil, leflunomide, NSAIDs, for example, ibuprofen, corticosteroids such as prednisolone, phosphodiesterase inhibitors, adensosine agonists, antithrombotic agents, complement inhibitors, adrenergic agents, agents which interfere with signalling by proinflammatory cytokines such as TNF ⁇ or IL-1 (e.g.
  • IL-1 ⁇ converting enzyme inhibitors TACE inhibitors
  • T-cell signaling inhibitors such as kinase inhibitors, metalloproteinase inhibitors, sulfasalazine, azathioprine, 6-mercaptopurines, angiotensin converting enzyme inhibitors, soluble cytokine receptors and derivatives thereof (e.g. soluble p55 or p75 TNF receptors, sIL-1RI, sIL-1RII, sIL-6R) and antiinflammatory cytokines (e.g. IL-4, IL-10, IL-13 and TGF ⁇ ).
  • IL-4, IL-10, IL-13 and TGF ⁇ antiinflammatory cytokines
  • interferon- ⁇ for example, IFN ⁇ 1a and IFN ⁇ 1b
  • copaxone corticosteroids
  • caspase inhibitors for example inhibitors of caspase-1, IL-1 inhibitors, TNF inhibitors, and antibodies to CD40 ligand and CD80.
  • a compound of formula (I) may also be combined with agents, such as alemtuzumab, dronabinol, Unimed, daclizumab, mitoxantrone, xaliproden hydrochloride, fampridine, glatiramer acetate, natalizumab, sinnabidol, a-immunokine NNSO3, ABR-215062, AnergiX.MS, chemokine receptor antagonists, BBR-2778, calagualine, CPI-1189, LEM (liposome encapsulated mitoxantrone), THC.CBD (cannabinoid agonist) MBP-8298, mesopram (PDE4 inhibitor), MNA-715, anti-IL-6 receptor antibody, neurovax, pirfenidone allotrap 1258 (RDP-1258), sTNF-R1, talampanel, teriflunomide, TGF-beta2, tiplimotide, VLA-4 antagonist
  • Non-limiting examples of therapeutic agents for Angina with which a compound of formula (I) of the invention can be combined include the following: aspirin, nitroglycerin, isosorbide mononitrate, metoprolol succinate, atenolol, metoprolol tartrate, amlodipine besylate, diltiazem hydrochloride, isosorbide dinitrate, clopidogrel bisulfate, nifedipine, atorvastatin calcium, potassium chloride, furosemide, simvastatin, verapamil HCl, digoxin, propranolol hydrochloride, carvedilol, lisinopril, spironolactone, hydrochlorothiazide, enalapril maleate, nadolol, ramipril, enoxaparin sodium, heparin sodium, valsartan, sotalol hydrochloride, fenofib
  • Non-limiting examples of therapeutic agents for Ankylosing Spondylitis with which a compound of formula (I) can be combined include the following: ibuprofen, diclofenac and misoprostol, naproxen, meloxicam, indomethacin, diclofenac, celecoxib, rofecoxib, Sulfasalazine, Methotrexate, azathioprine, minocyclin, prednisone, etanercept, infliximab.
  • Non-limiting examples of therapeutic agents for Asthma with which a compound of formula (I) can be combined include the following: albuterol, salmeterol/fluticasone, montelukast sodium, fluticasone propionate, budesonide, prednisone, salmeterol xinafoate, levalbuterol HCl, albuterol sulfate/ipratropium, prednisolone sodium phosphate, triamcinolone acetonide, beclomethasone dipropionate, ipratropium bromide, azithromycin, pirbuterol acetate, prednisolone, theophylline anhydrous, methylprednisolone sodium succinate, clarithromycin, zafirlukast, formoterol fumarate, influenza virus vaccine, methylprednisolone, amoxicillin trihydrate, flunisolide, allergy injection, cromolyn sodium, fexofenadine
  • Non-limiting examples of therapeutic agents for COPD with which a compound of formula (I) can be combined include the following: albuterol sulfate/ipratropium, ipratropium bromide, salmeterol/fluticasone, albuterol, salmeterol xinafoate, fluticasone propionate, prednisone, theophylline anhydrous, methylprednisolone sodium succinate, montelukast sodium, budesonide, formoterol fumarate, triamcinolone acetonide, levofloxacin, guaifenesin, azithromycin, beclomethasone dipropionate, levalbuterol HCl, flunisolide, ceftriaxone sodium, amoxicillin trihydrate, gatifloxacin, zafirlukast, amoxicillin/clavulanate, flunisolide/menthol, chlorpheniramine/hydrocodone, metaproter
  • Non-limiting examples of therapeutic agents for HCV with which a compound of formula (I) can be combined include the following: Interferon-alpha-2a, Interferon-alpha-2b, Interferon-alpha con1, Interferon-alpha-n1, Pegylated interferon-alpha-2a, Pegylated interferon-alpha-2b, ribavirin, Peginterferon alfa-2b+ribavirin, Ursodeoxycholic Acid, Glycyrrhizic Acid, Thymalfasin, Maxamine, VX-497 and any compounds that are used to treat HCV through intervention with the following targets: HCV polymerase, HCV protease, HCV helicase, HCV IRES (internal ribosome entry site).
  • Non-limiting examples of therapeutic agents for Idiopathic Pulmonary Fibrosis with which a compound of formula (I) can be combined include the following: prednisone, azathioprine, albuterol, colchicine, albuterol sulfate, digoxin, gamma interferon, methylprednisolone sod succ, lorazepam, furosemide, lisinopril, nitroglycerin, spironolactone, cyclophosphamide, ipratropium bromide, actinomycin d, alteplase, fluticasone propionate, levofloxacin, metaproterenol sulfate, morphine sulfate, oxycodone HCl, potassium chloride, triamcinolone acetonide, tacrolimus anhydrous, calcium, interferon-alpha, methotrexate, mycophenolate mofetil, Interferon-gamma-1
  • Non-limiting examples of therapeutic agents for Myocardial Infarction with which a compound of formula (I) can be combined include the following: aspirin, nitroglycerin, metoprolol tartrate, enoxaparin sodium, heparin sodium, clopidogrel bisulfate, carvedilol, atenolol, morphine sulfate, metoprolol succinate, warfarin sodium, lisinopril, isosorbide mononitrate, digoxin, furosemide, simvastatin, ramipril, tenecteplase, enalapril maleate, torsemide, retavase, losartan potassium, quinapril HCl/mag carb, bumetanide, alteplase, enalaprilat, amiodarone hydrochloride, tirofiban HCl m-hydrate, diltiazem hydrochloride, captopril
  • Non-limiting examples of therapeutic agents for Psoriasis with which a compound of formula (I) can be combined include the following: calcipotriene, clobetasol propionate, triamcinolone acetonide, halobetasol propionate, tazarotene, methotrexate, fluocinonide, betamethasone diprop augmented, fluocinolone acetonide, acitretin, tar shampoo, betamethasone valerate, mometas one furoate, ketoconazole, pramoxine/fluocinolone, hydrocortisone valerate, flurandrenolide, urea, betamethasone, clobetasol propionate/emoll, fluticasone propionate, azithromycin, hydrocortisone, moisturizing formula, folic acid, desonide, pimecrolimus, coal tar, diflorasone diacetate, etanercept folate
  • Non-limiting examples of therapeutic agents for Psoriatic Arthritis with which a compound of formula (I) can be combined include the following: methotrexate, etanercept, rofecoxib, celecoxib, folic acid, sulfasalazine, naproxen, leflunomide, methylprednisolone acetate, indomethacin, hydroxychloroquine sulfate, prednisone, sulindac, betamethasone diprop augmented, infliximab, methotrexate, folate, triamcinolone acetonide, diclofenac, dimethylsulfoxide, piroxicam, diclofenac sodium, ketoprofen, meloxicam, methylprednisolone, nabumetone, tolmetin sodium, calcipotriene, cyclosporine, diclofenac sodium/misoprostol, fluocinonide,
  • Non-limiting examples of therapeutic agents for Restenosis with which a compound of formula (I) can be combined include the following: sirolimus, paclitaxel, everolimus, tacrolimus, ABT-578, acetaminophen.
  • Non-limiting examples of therapeutic agents for Sciatica with which a compound of formula (I) can be combined include the following: hydrocodone bitartrate/apap, rofecoxib, cyclobenzaprine HCl, methylprednisolone, naproxen, ibuprofen, oxycodone HCl/acetaminophen, celecoxib, valdecoxib, methylprednisolone acetate, prednisone, codeine phosphate/apap, tramadol HCl/acetaminophen, metaxalone, meloxicam, methocarbamol, lidocaine hydrochloride, diclofenac sodium, gabapentin, dexamethasone, carisoprodol, ketorolac tromethamine, indomethacin, acetaminophen, diazepam, nabumetone, oxycodone HCl, tizanidine
  • Preferred examples of therapeutic agents for SLE (Lupus) in which a compound of formula (I) include the following: NSAIDS, for example, diclofenac, naproxen, ibuprofen, piroxicam, indomethacin; COX2 inhibitors, for example, Celecoxib, rofecoxib, valdecoxib; anti-malarials, for example, hydroxychloroquine; Steroids, for example, prednisone, prednisolone, budenoside, dexamethasone; Cytotoxics, for example, azathioprine, cyclophosphamide, mycophenolate mofetil, methotrexate; inhibitors of PDE4 or purine synthesis inhibitor, for example Cellcept.
  • NSAIDS for example, diclofenac, naproxen, ibuprofen, piroxicam, indomethacin
  • COX2 inhibitors for example, Celecoxib, rof
  • a compound of formula (I) may also be combined with agents such as sulfasalazine, 5-aminosalicylic acid, olsalazine, Imuran and agents which interfere with synthesis, production or action of proinflammatory cytokines such as IL-1, for example, caspase inhibitors like IL-1 ⁇ converting enzyme inhibitors and IL-1ra.
  • agents such as sulfasalazine, 5-aminosalicylic acid, olsalazine, Imuran and agents which interfere with synthesis, production or action of proinflammatory cytokines such as IL-1, for example, caspase inhibitors like IL-1 ⁇ converting enzyme inhibitors and IL-1ra.
  • a compound of formula (I) may also be used with T cell signaling inhibitors, for example, tyrosine kinase inhibitors; or molecules that target T cell activation molecules, for example, CTLA-4-IgG or anti-B7 family antibodies, anti-PD-1 family antibodies.
  • a compound of formula (I) can be combined with IL-11 or anti-cytokine antibodies, for example, fonotolizumab (anti-IFNg antibody), or anti-receptor receptor antibodies, for example, anti-IL-6 receptor antibody and antibodies to B-cell surface molecules.
  • a compound of formula (I) may also be used with LJP 394 (abetimus), agents that deplete or inactivate B-cells, for example, Rituximab (anti-CD20 antibody), lymphostat-B (anti-BlyS antibody), TNF antagonists, for example, anti-TNF antibodies, D2E7 (PCT Publication No.
  • WO 97/29131 HUMIRATM
  • CA2 REMICADETM
  • CDP 571 TNFR-Ig constructs, (p75TNFRIgG (ENBRELTM) and p55TNFRIgG (LENERCEPTTM)).
  • a “therapeutically effective amount” is an amount of a compound of Formula I or a combination of two or more such compounds, which inhibits, totally or partially, the progression of the condition or alleviates, at least partially, one or more symptoms of the condition.
  • a therapeutically effective amount can also be an amount which is prophylactically effective. The amount which is therapeutically effective will depend upon the patient's size and gender, the condition to be treated, the severity of the condition and the result sought. For a given patient, a therapeutically effective amount can be determined by methods known to those of skill in the art.
  • “Physiologically acceptable salts” refers to those salts which retain the biological effectiveness and properties of the free bases and which are obtained by reaction with inorganic acids, for example, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, and phosphoric acid or organic acids such as sulfonic acid, carboxylic acid, organic phosphoric acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, citric acid, fumaric acid, maleic acid, succinic acid, benzoic acid, salicylic acid, lactic acid, tartaric acid (e.g. (+) or ( ⁇ )-tartaric acid or mixtures thereof), amino acids (e.g. (+) or ( ⁇ )-amino acids or mixtures thereof), and the like.
  • These salts can be prepared by methods known to those skilled in the art.
  • Certain compounds of formula I which have acidic substituents may exist as salts with pharmaceutically acceptable bases.
  • the present invention includes such salts.
  • Examples of such salts include sodium salts, potassium salts, lysine salts and arginine salts. These salts may be prepared by methods known to those skilled in the art.
  • Certain compounds of formula I and their salts may exist in more than one crystal form and the present invention includes each crystal form and mixtures thereof.
  • Certain compounds of formula I and their salts may also exist in the form of solvates, for example hydrates, and the present invention includes each solvate and mixtures thereof.
  • Certain compounds of formula I may contain one or more chiral centers, and exist in different optically active forms.
  • compounds of formula I may contain one chiral center, the compounds exist in two enantiomeric forms and the present invention includes both enantiomers and mixtures of enantiomers, such as racemic mixtures.
  • the enantiomers may be resolved by methods known to those skilled in the art, for example by formation of diastereoisomeric salts which may be separated, for example, by crystallization; formation of diastereoisomeric derivatives or complexes which may be separated, for example, by crystallization, gas-liquid or liquid chromatography; selective reaction of one enantiomer with an enantiomer-specific reagent, for example enzymatic esterification; or gas-liquid or liquid chromatography in a chiral environment, for example on a chiral support for example silica with a bound chiral ligand or in the presence of a chiral solvent.
  • enantiomers may be synthesized by asymmetric synthesis using optically active reagents, substrates, catalysts or solvents, or by converting one enantiomer into the other by asymmetric transformation.
  • a compound of formula I When a compound of formula I contains more than one chiral center it may exist in diastereoisomeric forms.
  • the diastereoisomeric pairs may be separated by methods known to those skilled in the art, for example chromatography or crystallization and the individual enantiomers within each pair may be separated as described above.
  • the present invention includes each diastereoisomer of compounds of formula I and mixtures thereof.
  • Certain compounds of formula I may exist in different tautomeric forms or as different geometric isomers, and the present invention includes each tautomer and/or geometric isomer of compounds of formula I and mixtures thereof.
  • Certain compounds of formula I may exist in different stable conformational forms which may be separable. Torsional asymmetry due to restricted rotation about an asymmetric single bond, for example because of steric hindrance or ring strain, may permit separation of different conformers.
  • the present invention includes each conformational isomer of compounds of formula I and mixtures thereof.
  • Certain compounds of formula I may exist in zwitterionic form and the present invention includes each zwitterionic form of compounds of formula I and mixtures thereof.
  • pro-drug refers to an agent which is converted into the parent drug in vivo by some physiological chemical process (e.g., a prodrug on being brought to the physiological pH is converted to the desired drug form).
  • Pro-drugs are often useful because, in some situations, they may be easier to administer than the parent drug. They may, for instance, be bioavailable by oral administration whereas the parent drug is not.
  • the prodrug may also have improved solubility in pharmacological compositions over the parent drug.
  • pro-drug a compound of the present invention wherein it is administered as an ester (the “pro-drug”) to facilitate transmittal across a cell membrane where water solubility is not beneficial, but then it is metabolically hydrolyzed to the carboxylic acid once inside the cell where water solubility is beneficial
  • Pro-drugs have many useful properties. For example, a pro-drug may be more water soluble than the ultimate drug, thereby facilitating intravenous administration of the drug. A pro-drug may also have a higher level of oral bioavailability than the ultimate drug. After administration, the prodrug is enzymatically or chemically cleaved to deliver the ultimate drug in the blood or tissue.
  • Exemplary pro-drugs upon cleavage release the corresponding free acid, and such hydrolyzable ester-forming residues of the compounds of this invention include but are not limited to carboxylic acid substituents (e.g., —(CH 2 )C(O)H or a moiety that contains a carboxylic acid) wherein the free hydrogen is replaced by (C 1 -C 4 )alkyl, (C 2 -C 12 )alkanoyloxymethyl, (C 4 -C 9 )1-(alkanoyloxy)ethyl, 1-methyl-1-(alkanoyloxy)-ethyl having from 5 to 10 carbon atoms, alkoxycarbonyloxymethyl having from 3 to 6 carbon atoms, 1-(alkoxycarbonyloxy)ethyl having from 4 to 7 carbon atoms, 1-methyl-1-(alkoxycarbonyloxy)ethyl having from 5 to 8 carbon atoms, N-(alkoxycarbonyl)aminomethyl having
  • exemplary pro-drugs release an alcohol of Formula I wherein the free hydrogen of the hydroxyl substituent (e.g., R 1 contains hydroxyl) is replaced by (C 1 -C 6 )alkanoyloxymethyl, 1-((C 1 -C 6 )alkanoyloxy)ethyl, 1-methyl-1-((C 1 -C 6 )alkanoyloxy)ethyl, (C 1 -C 6 )alkoxycarbonyloxymethyl, N—(C 1 -C 6 )alkoxycarbonylamino-methyl, succinoyl, (C 1 -C 6 )alkanoyl, ⁇ -amino(C 1 -C 4 )alkanoyl, arylactyl and ⁇ -aminoacyl, or ⁇ -aminoacyl- ⁇ -aminoacyl wherein said ⁇ -aminoacyl moieties are independently any of the naturally occurring L-amino acids found in proteins, P(O
  • Heteroaromatic groups include heteroaryl ring systems (e.g., for purposes of exemplification, which should not be construed as limiting the scope of this invention: thienyl, pyridyl, pyrazole, isoxazolyl, thiadiazolyl, oxadiazolyl, indazolyl, furans, pyrroles, imidazoles, pyrazoles, triazoles, pyrimidines, pyrazines, thiazoles, isothiazoles, oxazolyl or tetrazoles) and heteroaryl ring systems in which a carbocyclic aromatic ring, carbocyclic non-aromatic ring or heteroaryl ring is fused to one or more other heteroaryl rings (e.g., for purposes of exemplification, which should not be construed as limiting the scope of this invention: benzo[b]thienyl, benzimidazolyl, benzo
  • Substituted heteroaryl groups are preferably substituted with one or more substituents each independently selected from the group consisting of halogen, hydroxy, alkyl, alkoxy, alkyl-O—C(O)—, alkoxyalkyl, a heterocycloalkyl group, optionally substituted phenyl, nitro, amino, mono-substituted amino or di-substituted amino
  • heterocyclic or “heterocyclyl”, as used herein, include aromatic and non-aromatic, ring systems, including, but not limited to, monocyclic, bicyclic and tricyclic rings, which can be completely saturated or which can contain one or more units of unsaturation and have 3 to 12 atoms including at least one heteroatom, such as nitrogen, oxygen, or sulfur.
  • azaindole azetidinyls furans, imidazoles, imidazopyridine, indole, isoxazoles, isothiazoles, oxadiazoles, oxazoles, piperazines, piperidines, pyrans, pyrazines, pyrazoles, pyridines, pyrimidines, pyrroles, pyrrolidines, quinolines, quinazolines, triazoles, thiazoles, tetrahydroindole, tetrazoles, thiadiazoles, thienyls, thiomorpholinos or triazles.
  • substituted heterocyclic (or heterocyclyl) is used, what is meant is that the heterocyclic group is substituted with one or more substituents that can be made by one of ordinary skill in the art and results in a molecule that is a kinase inhibitor.
  • preferred substituents for the heterocyclyls of this invention are each independently selected from the optionally substituted group consisting of alkenyl, alkoxy, alkoxyalkoxy, alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylheterocycloalkoxy, alkyl, alkylcarbonyl, alkylester, alkyl-O—C(O)—, alkyl-heterocyclyl, alkyl-cycloalkyl, alkyl-nitrile, alkynyl, amido groups, amino, aminoalkyl, aminocarbonyl, carbonitrile, carbonylalkoxy, carboxamido, CF 3 , CN, —C(O)OH, —C(O)H, —C(O)—)(CH 3 ) 3 , —OH, —C(O)O-alkyl, —C(O)
  • R c for each occurrence is independently hydrogen, optionally substituted alkyl, optionally substituted aryl, —(C 1 -C 6 )—NR d R e , -E-(CH 2 ) t —NR d R e , -E-(CH 2 ) t —O-alkyl, -E-(CH 2 ) t —S-alkyl, or -E-(CH 2 ) t —OH
  • heterocycloalkyl is a heterocyclic group that is linked to a compound by an aliphatic group having from one to about eight carbon atoms.
  • a preferred heterocycloalkyl group is an imidazolylethyl group.
  • aliphatic or “an aliphatic group” or notations such as “(C 0 -C 8 )” include straight chained or branched hydrocarbons which are completely saturated or which contain one or more units of unsaturation, and, thus, includes alkyl, alkenyl, alkynyl and hydrocarbons comprising a mixture of single, double and triple bonds. When the group is a C o it means that the moiety is not present or in other words, it is a bond.
  • alkyl means C 1 -C 8 and includes straight chained or branched hydrocarbons which are completely saturated.
  • alkyls are methyl, ethyl, propyl, butyl, pentyl, hexyl and isomers thereof.
  • alkenyl and alkynyl means C 2 -C 8 and includes straight chained or branched hydrocarbons which contain one or more units of unsaturation, one or more double bonds for alkenyl and one or more triple bonds for alkynyl.
  • aromatic groups include aromatic carbocyclic ring systems (e.g. phenyl and cyclopentyldienyl) and fused polycyclic aromatic ring systems (e.g. naphthyl, biphenylenyl and 1,2,3,4-tetrahydronaphthyl).
  • cycloalkyl means C 3 -C 12 monocyclic or multicyclic (e.g., bicyclic, tricyclic, etc.) hydrocarbons which is completely saturated or has one or more unsaturated bonds but does not amount to an aromatic group.
  • Preferred examples of a cycloalkyl group are cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl and cyclohexenyl.
  • amido group means —NHC( ⁇ O)—.
  • acyloxy groups are —OC(O)R.
  • alkenyl groups alkoxy group (which itself can be substituted, such as —O—C 1 -C 6 -alkyl-OR, —O—C 1 -C 6 -alkyl-N(R) 2 , and OCF 3 ), alkoxyalkoxy, alkoxycarbonyl, alkoxycarbonylpiperidinylalkoxy, alkyl groups (which itself can also be substituted, such as —C 1 -C 6 -alkyl-OR, —C 1 -C 6 -alkyl-N(R) 2 , and —CF 3 ), alkylamino, alkylcarbonyl, alkylester, alkylnitrile, alkylsulfonyl, amino, aminoalkoxy, CF 3 , COH, COOH, CN, cycloalkyl, dialky
  • compositions suitable for use in the present invention include compositions wherein the active ingredients are contained in an effective amount to achieve its intended purpose. More specifically, a therapeutically effective amount means an amount effective to prevent development of or to alleviate the existing symptoms of the subject being treated. Determination of the effective amounts is well within the capability of those skilled in the art.
  • the therapeutically effective dose can be estimated initially from cellular assays.
  • a dose can be formulated in cellular and animal models to achieve a circulating concentration range that includes the IC 50 as determined in cellular assays (i.e., the concentration of the test compound which achieves a half-maximal inhibition of a given protein kinase activity).
  • the IC 50 as determined in cellular assays (i.e., the concentration of the test compound which achieves a half-maximal inhibition of a given protein kinase activity).
  • Such information can be used to more accurately determine useful doses in humans.
  • the most preferred compounds for systemic administration effectively inhibit protein kinase signaling in intact cells at levels that are safely achievable in plasma.
  • a therapeutically effective dose refers to that amount of a compound of Formula I or a combination of two or more such compounds, which inhibits, totally or partially, the progression of a condition or alleviates, at least partially, one or more symptoms of the condition.
  • a therapeutically effective amount can also be an amount which is prophylactically effective.
  • Toxicity and therapeutic efficacy of such compounds can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the maximum tolerated dose (MTD) and the ED 50 (effective dose for % maximal response).
  • the dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio between MTD and ED 50 .
  • Compounds which exhibit high therapeutic indices are preferred.
  • the data obtained from these cell culture assays and animal studies can be used in formulating a range of dosage for use in humans.
  • the dosage of such compounds lies preferably within a range of circulating concentrations that include the ED 50 with little or no toxicity.
  • the dosage may vary within this range depending upon the dosage form employed and the route of administration utilized.
  • a therapeutically effective amount can also be an amount which is prophylactically effective.
  • the amount which is therapeutically effective will depend upon the patient's size and gender, the condition to be treated, the severity of the condition and the result sought. For a given patient, a therapeutically effective amount can be determined by methods known to those of skill in the art.
  • the exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient's condition. (See e.g. Fingl et al., 1975, in “The Pharmacological Basis of Therapeutics”, Ch. 1 p 1). In the treatment of crises, the administration of an acute bolus or an infusion approaching the MTD may be required
  • Dosage amount and interval may be adjusted individually to provide plasma levels of the active moiety which are sufficient to maintain the kinase modulating effects, or minimal effective concentration (MEC).
  • MEC minimal effective concentration
  • the MEC will vary for each compound but can be estimated from in vitro data; e.g. the concentration necessary to achieve 50-90% inhibition of protein kinase using the assays described herein. Dosages necessary to achieve the MEC will depend on individual characteristics and route of administration. However, HPLC assays or bioassays can be used to determine plasma concentrations.
  • Dosage intervals can also be determined using the MEC value.
  • Compounds should be administered using a regimen which maintains plasma levels above the MEC for 10-90% of the time, preferably between 30-90% and most preferably between 50-90% until the desired amelioration of symptoms is achieved.
  • the effective local concentration of the drug may not be related to plasma concentration.
  • composition administered will, of course, be dependent on the subject being treated, on the subject's weight, the severity of the affliction, the manner of administration and the judgment of the prescribing physician.
  • compositions may, if desired, be presented in a pack or dispenser device which may contain one or more unit dosage forms containing the active ingredient.
  • the pack may for example comprise metal or plastic foil, such as a blister pack.
  • the pack or dispenser device may be accompanied by instructions for administration.
  • Compositions comprising a compound of the invention formulated in a compatible pharmaceutical carrier may also be prepared, placed in an appropriate container, and labeled for treatment of an indicated condition.
  • the compounds of the present invention in the form of particles of very small size, for example as obtained by fluid energy milling.
  • active compound denotes any compound of the invention but particularly any compound which is the final product of one of the preceding Examples.
  • capsules 10 parts by weight of active compound and 240 parts by weight of lactose can be de-aggregated and blended. The mixture can be filled into hard gelatin capsules, each capsule containing a unit dose or part of a unit dose of active compound.
  • Tablets can be prepared, for example, from the following ingredients.
  • Active compound 10 Lactose 190 Maize starch 22 Polyvinylpyrrolidone 10 Magnesium stearate 3
  • the active compound, the lactose and some of the starch can be de-aggregated, blended and the resulting mixture can be granulated with a solution of the polyvinyl-pyrrolidone in ethanol.
  • the dry granulate can be blended with the magnesium stearate and the rest of the starch.
  • the mixture is then compressed in a tabletting machine to give tablets each containing a unit dose or a part of a unit dose of active compound.
  • Tablets can be prepared by the method described in (b) above.
  • the tablets can be enteric coated in a conventional manner using a solution of 20% cellulose acetate phthalate and 3% diethyl phthalate in ethanol:dichloromethane (1:1).
  • suppositories for example, 100 parts by weight of active compound can be incorporated in 1300 parts by weight of triglyceride suppository base and the mixture formed into suppositories each containing a therapeutically effective amount of active ingredient.
  • the active compound may, if desired, be associated with other compatible pharmacologically active ingredients.
  • the compounds of this invention can be administered in combination with another therapeutic agent that is known to treat a disease or condition described herein.
  • additional pharmaceutical agents that inhibit or prevent the production of VEGF or angiopoietins, attenuate intracellular responses to VEGF or angiopoietins, block intracellular signal transduction, inhibit vascular hyperpermeability, reduce inflammation, or inhibit or prevent the formation of edema or neovascularization.
  • the compounds of the invention can be administered prior to, subsequent to or simultaneously with the additional pharmaceutical agent, whichever course of administration is appropriate.
  • the additional pharmaceutical agents include, but are not limited to any of the agents, for examples, described in pages 20-28.
  • the compounds of the invention and the additional pharmaceutical agents act either additively or synergistically.
  • the administration of such a combination of substances that inhibit angiogenesis, vascular hyperpermeability and/or inhibit the formation of edema can provide greater relief from the deletrious effects of a hyperproliferative disorder, angiogenesis, vascular hyperpermeability or edema than the administration of either substance alone.
  • combinations with antiproliferative or cytotoxic chemotherapies or radiation are included in the scope of the present invention.
  • the present invention also comprises the use of a compound of formula I as a medicament.
  • a further aspect of the present invention provides the use of a compound of formula I or a salt thereof in the manufacture of a medicament for treating vascular hyperpermeability, angiogenesis-dependent disorders, proliferative diseases and/or disorders of the immune system in mammals, particularly human beings.
  • the present invention also provides a method of treating vascular hyperpermeability, inappropriate neovascularization, proliferative diseases and/or disorders of the immune system which comprises the administration of a therapeutically effective amount of a compound of formula Ito a mammal, particularly a human being, in need thereof.
  • the potency of compounds can be determined by the amount of inhibition of the phosphorylation of an exogenous substrate (e.g., a synthetic peptide (Z. Songyang et al., Nature. 373:536-539) by a test compound relative to control.
  • an exogenous substrate e.g., a synthetic peptide (Z. Songyang et al., Nature. 373:536-539)
  • HTRF Homogenous time-resolved fluorescence
  • Purified enzymes (available from commercial sources) were mixed with different amounts of N-biotinylated substrates or GST-tagged substrates (see table) at varying concentrations of inhibitor in different reaction buffers (40 ⁇ L final volume, see table).
  • the kinase reaction was initiated by addition of ATP (0.01-0.1 mM final conc.) in a black 96-half-well plate (Perkin Elmer). After 50-60 minutes incubation at room temperature, the reaction was quenched by addition of EDTA (final conc.
  • THP-1 cells were serum starved (0.5% FBS) for about 24 hours and seeded to 96 well plates at a density of 2 ⁇ 10 5 cells/well in 100 ul of low serum media.
  • Test compounds were solubilized in DMSO and added to cells over the range of 25 uM-8 nM (final DMSO conc 0.5%).
  • Compounds were pre-incubated for about 30 mins before the addition of 1 ug/ml LPS.
  • Cells were stimulated for about 45 mins., washed and lysed in 100 ul of Biorad cell lysis buffer.
  • Level of HSP27 phosphorylation was measured via Bio-Plex phosphoprotein assay utilizing pHSP27 Beadmates from Upstate.
  • Thp-1 cells were serum starved (0.5% FBS) for about 24 hours and seeded to 96 well plates at a density of 2 ⁇ 10 5 cells/well in 100 ul of low serum media.
  • Test compounds were solubilized in DMSO and added to cells over the range of 25 uM-8 nM (final DMSO conc 0.5%).
  • Compounds were pre-incubated for 30 mins before the addition of 1 ug/ml LPS.
  • Cells were stimulated for about 3 hrs. Supernatent media was removed and TNF release was quantified by ELISA. Cellular toxicity was determined by the addition of MTT to the remaining cells.
  • PBMC Peripheral Blood Mononuclear Cell
  • PBMC's from leukopak's by Ficoll separation. Adjust the cell density to 1 ⁇ 10 7 cells/ml in media.
  • Media used is RPMI Medium 1640 (Gibco BRL, Grand Island, N.Y., Catalog Number 31800)+2% human AB sera (Sigma Chemical Company, St. Louis, Mo., Catalog Number S7148, heat inactivated) with 100 U/ml penicillin (Gibco BRL, Catalog Number 15140), 2 mM L-glutamine (Gibco BRL, Catalog Number 25030), 1 ⁇ MEM Non-Essential Amino Acids (Gibco BRL, Catalog Number 11140), and 10 mM pH 7.3 Hepes. Media is filtered through a 0.2-micron filter unit.
  • Lipopolysaccharide Escherichia coli (Calbiochem, La Jolla, Calif., Catalog Number 437625) and incubate plate(s) overnight (about 16 hours) in a 37° C. CO 2 incubator to stimulate cytokine production.
  • Harvest supernates for cytokine analysis Spin plate(s) in a centrifuge at 180 g for about 10 minutes with no brake to pellet cells (we used a Beckman GPKR centrifuge and spin at 1,000 rpm.) Remove 100 uL/well supernate for cytokine analysis.
  • MTT is converted into a colored product when it is cleaved by the mitochondrial reductase system, which is present in metabolically active cells.
  • the MTT Assay can be used as a measure of cellular viability.
  • PBMC Peripheral Blood Mononuclear Cell
  • OD570/630 of sample/OD570/630 of 100% viable control ⁇ 100 % viability of sample.
  • PBMC Peripheral Blood Mononuclear Cells
  • PBMCs are prepared from leukopaks and stored frozen in vials in liquid nitrogen freezer.
  • Compounds of formula I may have therapeutic utility in the treatment of diseases involving both identified, including those not mentioned herein, and as yet unidentified protein tyrosine kinases which are inhibited by compounds of formula I. All compounds exemplified herein significantly inhibit either COT or MK2 at concentrations of 50 micromolar or below.
  • mice are injected i.v. with LPS (from Escherichia coli Serotype 0111:B4, Sigma #L-4130), dissolved in saline.
  • LPS from Escherichia coli Serotype 0111:B4, Sigma #L-4130
  • 0.1 mpk LPS is given and to measure IFN- ⁇ , IL-1 ⁇ , IL-18, IL-6, and IL-12
  • 5mpk LPS is given.
  • the mice are then cardiac bled for serum at the appropriate time points listed below.
  • the animals are bled at 90 minutes for TNF- ⁇ or at 4 hours for IFN- ⁇ , IL-1 ⁇ , IL-18, IL-6, IL-12, then the serum cytokine levels are measured by ELISA.
  • the compound is dosed either p.o. or i.p. one hour prior to the LPS injection and the levels of target cytokines are measured and compared with those obtained for the control group in order to calculate ED 50 levels.
  • mice or rats are immunized with an emulsion of myelin basic protein (MBP), or neurogenic peptide derivatives thereof, and CFA.
  • MBP myelin basic protein
  • Acute disease can be induced with the addition of bacterial toxins such as bordetella pertussis . Relapsing/remitting disease is induced by adoptive transfer of T-cells from MBP/peptide immunized animals.
  • CIA may be induced in DBA/1 mice by immunization with type II collagen (J. Immunol:142(7):2237-2243). Mice will develop signs of arthritis as early as ten days following antigen challenge and may be scored for as long as ninety days after immunization. In both the EAE and CIA models, a compound may be administered either prophylactically or at the time of disease onset. Efficacious drugs should reduce severity and/or incidence.
  • Certain compounds of this invention which inhibit one or more angiogenic receptor PTK, and/or a protein kinase such as lck involved in mediating inflammatory responses can reduce the severity and incidence of arthritis in these models.
  • Example #G.19 was prepared from 2-(3- ⁇ 2-(dimethylamino-methyleneamino)-7-[7-benzo[b]thiophen-2-yl-1-(2-trimethylsilanyl-ethoxymethyl)-1H-indazol-5-yl]-pyrrolo[3,2-d]pyrimidin-5-yl ⁇ -propan-1-oxy)tetrahydropyran using general procedure G, as represented in the following synthetic scheme:
  • an organic solvent for example, methylene chloride or ethanol, preferably ethanol
  • an amine 2 to 4 equivalents, preferably 3 equivalents.
  • the reaction mixture is stirred at about 40-70° C. (preferably about 50° C.) for about 3-24 hours (preferably, about 5-10 hours).
  • the solvent is removed under reduced pressure to afford the crude product.
  • the crude product can be further purified by crystallization or chromatography.
  • a carboxylic acid (1-2.5 equivalents, preferably 1-1.5 equivalents) and an amine (1-2.5 equivalents, preferably 1-1.5 equivalents) in an organic solvent for example, THF, EtOAc, Et 2 O, or DMF, preferably DMF
  • an organic solvent for example, THF, EtOAc, Et 2 O, or DMF, preferably DMF
  • the coupling reagent for example, DCC, DIC, EDC, HBTU, HATU or TFFH, preferably HBTU
  • a coupling additive for example, HOBT or HOAT, preferably HOBT
  • DIEA 0.1-25 equivalents, preferably 3 equivalents.
  • the reaction mixture is stirred at about 20-70° C. (preferably about 50° C.) for about 5-40 hours (preferably about 35 hours) and then cooled to ambient temperature.
  • the reaction mixture can be purified in three different ways: 1).
  • the reaction mixture is treated with MP-carbonate (3-10 equivalents, preferably 5 equivalents) with or without methanol. After about 4-48 hours (preferably about 14 hours), the reaction solution is separated from the resin by filtration and concentrated under reduced pressure to afford the crude product that can be further purified by crystallization or chromatography. If the product precipitates prior to filtration of the resin, the suspension containing the product is separated from the resin via pipette and filtered to afford the crude product that can be further purified by crystallization or chromatography. 2).
  • the reaction mixture is partitioned between water and an organic solvent (for example, CH 2 Cl 2 , EtOAc or Et 2 O, preferably CH 2 Cl 2 ).
  • organic layer is separated and the aqueous layer is further extracted with organic solvent.
  • the combined organic extracts are dried over a desiccant and evaporated under reduced pressure to afford the product that can be further purified by crystallization or chromatography. 3).
  • the reaction mixture is directly concentrated under reduced pressure and the residue is purified by crystallization or chromatography.
  • the reaction was concentrated under reduced pressure and the residue was purified by preparative HPLC (Waters Symmetry C8 column (25 ⁇ 100 mm, 7 ⁇ m particle size) using a gradient of 10%-100% CH 3 CN/0.1% aqueous TFA over 8 min (10 min run time) at a flow rate of 40 mL/min).
  • a solution of N-methylmorpholine (2-5 equivalents, preferably 4 equivalents) in an organic solvent for example, THF, CH 2 Cl 2 , CH 3 CN, or CH 3 CN/CH 2 Cl 2 1:1
  • Si-DCT Si-dichlorotriazine
  • a solution of the carboxylic acid (1.1-2.5 equivalents, preferably 1.25 equivalents) in an organic solvent for example, THF, CH 2 Cl 2 , CH 3 CN, or 1:1 CH 3 CN/CH 2 Cl 2 , preferably 1:1 CH 3 CN/CH 2 Cl 2 ) is added and mixed for about 1 min.
  • a solution of the amine (1 equivalent) in an organic solvent for example, THF, CH 2 Cl 2 , CH 3 CN, or 1:1 CH 3 CN/CH 2 Cl 2 , preferably 1:1 CH 3 CN/CH 2 Cl 2 ) is then added.
  • the reaction is shaken at about 10-60° C., preferably about ambient temperature, for about 10-24 hours, preferably about 16 hours.
  • the crude reaction solution is mixed with DMF, eluted through a Si-carbonate (1 g, 6 mL) cartridge with additional organic solvent (for example, MeOH), and concentrated in vacuo.
  • the crude product can then be further purified by crystallization or chromatography.
  • the crude reaction solution was mixed with DMF (5 mL) to aid in solubility, eluted through a Si-carbonate (SiliCycle, Inc; 1 g, 6 mL) cartridge using MeOH (approx 3 mL), and concentrated in vacuo.
  • a solvent for example, DME or CH 2 Cl 2 , preferably CH 2 Cl 2
  • an anhydrous solvent for example, DMF or DMA, preferably DMF
  • the residue is dissolved in an organic solvent (CH 2 Cl 2 or EtOAc, preferably EtOAc) and washed with water.
  • the organic layer is dried over a desiccant (for example, magnesium sulfate or sodium sulfate, preferably magnesium sulfate) and further purified by crystallization or chromatography.
  • a desiccant for example, magnesium sulfate or sodium sulfate, preferably magnesium sulfate
  • a boronating reagent bis(pinacolato)diboron or pinacolatoborane, preferably bis(pinacolato)diboron)(1-1.5 equivalents, preferably 1.3 equivalents
  • an aryl halide for example, an aryl bromide or an aryl iodide, preferably an aryl iodide
  • a palladium catalyst for example tris(benzylideneacetone)dipalladium (0), tetrakis(triphenylphosphine)palladium(0), bis(acetato)triphenylphosphinepalladium(II) ( ⁇ 5% Pd) polymer-bound FibreCatTM or [1,1′-bis(diphenylphosphino)ferrocene] dichloropalladium(II), complex with dichloromethane) (preferably dichloro[1,1′-bis(dipheny
  • the mixture is heated at about 50-100° C. (preferably about 80° C.) for about 1-24 hours (preferably about 15 hours) under an inert atmosphere.
  • the mixture is allowed to cool to ambient temperature, and the solvent is removed under reduced pressure.
  • the residue can then be further purified by chromatography or crystallization.
  • a boronate ester or a boronic acid (1-5 equivalents, preferably 2 equivalents), an aryl halide (for example, an aryl bromide, aryl chloride or an aryl iodide, preferably an aryl iodide) (0.7-3 equivalents, preferably 1 equivalent) and an inorganic base (for example, KF, Na 2 CO 3 or Cs 2 CO 3 , preferably Cs 2 CO 3 ) (2-16 equivalents, preferably 2.5 equivalents) in a degassed organic solvent (for example THF, DME, DMF, 1,4-dioxane, DME/water or toluene, preferably DMF or DME/water) is added a palladium catalyst (for example tris(benzylideneacetone)dipalladium (0), tetrakis(triphenylphosphine)palladium(0), bis(acetato)triphenylphosphinepalladium(I
  • tributylphosphinetetrafluoroborate (0.01 to 0.20 equivalents, preferably 0.05 equivalents) is also added.
  • the reaction mixture is heated at about 40-150° C. (preferably about 80° C.) for about 2-24 hours (preferably about 18 hours) or at about 100-200° C. (preferably 150° C.) for about 5-60 minutes (preferably about 15 minutes) in a microwave under an inert atmosphere.
  • the reaction mixture is allowed to cool to ambient temperature. Subsequently, the solvents are removed under reduced pressure and the residue is suspended in a mixture of EtOAc and water; the mixture is stirred for 30 minutes and the resulting solid is collected by filtration; the product can be further purified by chromatography or crystallization.
  • the cooled reaction mixture is diluted with water or an aqueous basic solution (such as saturated aqueous NaHCO 3 ) and extracted (1-5 times, preferably 3 times) with a suitable solvent (such as EtOAc or CH 2 Cl 2 ) then the combined organic extracts are dried (for example, over Na 2 SO 4 or MgSO 4 ), decanted or filtered, and concentrated under reduced pressure to afford the product that can be further purified by chromatography or crystallization.
  • a tert-butoxycarbonyl (Boc) protected amine is used, then the material is subsequently suspended in a mixture of methanol/6 N HCl and heated to about 65° C. for about one hour then cooled, concentrated and purified by chromatography or crystallization.
  • 5-(2-Aminopyrimidin-4-yl)amino-3-chloro-7-iodo-1H-indazole was prepared from 3-Chloro-5-nitro-1H-indazole ( J. Med. Chem., 46(26); 2003; 5663-5673, via halogenation conditions used in the synthesis of Preparation #22a, and general procedure N (using 2-amino-4-chloro-pyrimidine)).
  • a mixture of a SEM protected indazole in a solvent for example MeOH, EtOH, i-PrOH, CH 2 Cl 2 or dioxane, preferably MeOH
  • a solvent for example MeOH, EtOH, i-PrOH, CH 2 Cl 2 or dioxane, preferably MeOH
  • a mineral acid for example HCl, HF or TFA, preferably HCl
  • the solvent is evaporated and the product isolated and further purified by crystallization or chromatography.
  • a TBDMS protected substrate in an organic solvent for example methanol, ethanol, ispropanol, CH 2 Cl 2 , or dioxane, preferably methanol
  • an organic solvent for example methanol, ethanol, ispropanol, CH 2 Cl 2 , or dioxane, preferably methanol
  • an acid for example HCl, or TFA, preferably HCl
  • the reaction mixture is then stirred at about 25-85° C. (preferably about 65° C.) for about 0.5-24 hours (preferably about 1 hour).
  • the solvent is evaporated and the product is isolated by crystallization or by chromatography.
  • a mixture of a benzyl protected ether (1 equivalent) and a Pd catalyst (for example, 10 wt. % Pd on carbon or palladium (II) oxide, preferably 10 wt. % Pd on carbon) (5-10 mol %, preferably 7 mol %) in a degassed organic solvent (for example MeOH or EtOH, preferably MeOH) is shaken under an atmosphere of hydrogen gas at about 30-50 psi (preferably 40 psi) at about 10-50° C. (preferably about 25° C.) for about 2-24 hours (preferably about 18 hours).
  • the reaction mixture is evacuated, flushed with an inert gas (for example nitrogen or argon, preferably nitrogen) and filtered.
  • the solvents are removed under reduced pressure to afford the product that can be further purified by chromatography or crystallization.
  • a reducing agent for example borane, borane-pyridine, borane-diemethylsulfide, LiBH 4 or NaBH 4 , preferably NaBH 4
  • an organic solvent for example, DMF, dioxane, THF, MeOH
  • aryl sulfone is treated with a large excess of a nucleophile (for example an amine or an alcohol, preferably an amine) (100-500 equivalents, preferably 250 equivalents) in the absence of an organic solvent.
  • a nucleophile for example an amine or an alcohol, preferably an amine
  • the mixture is stirred for 5-60 hours (preferably about 16 hours) at about 0-100° C. (preferably about 20° C.). If heated, the mixture is allowed to cool to ambient temperature, filtered, and the solids are further purified by crystallization or chromatography if necessary.
  • a nitroaromatic compound preferably 1 equivalent
  • a solvent for example, EtOAc, EtOH, HOAc, 9M NH 4 OH, preferably EtOAc
  • a reducing reagent for example, hydriodic acid, iron powder, iron sulfate hydrate, tin chloride dehydrate, or palladium on carbon
  • the reaction mixture is stirred at about 20-100° C. (preferably about 90° C.) for about 1-20 hours (preferably about 2 hours).
  • a hydrogen atmosphere about 15-60 psi, preferably about 40 psi is when a palladium catalyst is employed.
  • an organic solvent for example, EtOAc or CH 2 Cl 2 , preferably CH 2 Cl 2 .
  • an ester 1.0 equivalent
  • an amine for example, ammonia, a primary amine or a second amine
  • an alcoholic solvent MeOH, EtOH or i-PrOH, preferably i-PrOH
  • an amine 1.0 equivalent
  • an organic solvent for example, EtOH, MeOH, or dioxane, preferably EtOH
  • an aromatic halide 1.0-10 equivalents, preferably 1.0 equivalent.
  • the reaction mixture is heated at about 25-160° C. (preferably about 80° C.) with stirring for about 20 minutes-4 hours (preferably about 30 minutes) or heated in a microwave reactor at about 120-180° C. (preferably about 150° C.) for about 5-30 minutes (preferably about 10 minutes).
  • the mixture is allowed to cool to ambient temperature.
  • the solvent is removed under reduced pressure or the mixture is filtered to give crude product that is then purified directly by chromatography or crystallization or submitted to the following aqueous work up.
  • the crude material is suspended in a basic solution (for example, NH 4 OH in MeOH, saturated aqueous NaHCO 3 or 2M aqueous Na 2 CO 3 , preferably saturated aqueous NaHCO 3 ) then filtered and washed with an organic solvent (for example MeOH, EtOAc, or CH 2 Cl 2 ). If needed, the resulting solid can then be purified by chromatography or crystallization.
  • a basic solution for example, NH 4 OH in MeOH, saturated aqueous NaHCO 3 or 2M aqueous Na 2 CO 3 , preferably saturated aqueous NaHCO 3
  • an organic solvent for example MeOH, EtOAc, or CH 2 Cl 2
  • a tert-butoxycarbonyl (Boc) protected diamine is used then the material is suspended in a mixture of methanol/6 N hydrochloric acid and heated to about 65° C. for about one hour then cooled, concentrated and purified by chromatography or crystallization
  • an organic solvent for example 1,2-dichloroethane, THF, CH 2 Cl 2 , DMF, or EtOAc, preferably 1,2-dichloroethane
  • a reducing agent for example, sodium triacetoxyborohydride or sodium cyanoborohydride, preferably sodium triacetoxyborohydride
  • Additional acetic acid (1-10 equivalents, preferably 3 equivalents) is added to progress the reaction when necessary.
  • the resulting mixture is allowed to stir at room temperature for 2-112 hours (preferably 24 hours).
  • the reaction mixture is purified in one of two ways: 1).
  • the reaction solution is treated with an aqueous solution of an appropriate base (for example NaOH, NaHCO 3 , or Na 2 CO 3 , preferably NaHCO 3 ) and an organic solvent (for example, CH 2 Cl 2 or EtOAc, preferably CH 2 Cl 2 ).
  • an organic solvent for example, CH 2 Cl 2 or EtOAc, preferably CH 2 Cl 2
  • the two layers are stirred for about 15 minutes then separated.
  • the organic phase is concentrated under reduced pressure.
  • the resulting crude product can be purified by trituration with an appropriate solvent (water, EtOH, toluene, or EtOAc, preferably EtOH) or by chromatography, or 2).
  • the reaction mixture is directly concentrated under reduced pressure and purified by chromatography, trituration, or crystallization.
  • aqueous solution for example, saturated aqueous NaHCO 3 or Na 2 CO 3 , preferably NaHCO 3
  • organic solvent EtOAc, Et 2 O, or CH 2 Cl 2 , preferably CH 2 Cl 2
  • the residue is partitioned between water and an organic solvent.
  • the organic layer is separated and the aqueous layer further extracted with the organic solvent.
  • the combined organic extracts are dried over a desiccant and the solvent removed under reduced pressure. 2).
  • the reaction mixture can be cooled down to room temperature and diluted with MeOH then the solvents are removed under reduced pressure.
  • the compound can be further purified by chromatography or crystallization.
  • An acid (HCl or TFA, preferably HCl) was added to an amine protected with a carbamate or amidine moiety optionally dissolved in an organic solvent (MeOH, CH 2 Cl 2 or dioxane, preferably MeOH) at about 0-100° C., preferably 40° C.
  • the reaction mixture is stirred for about 5 minutes to 24 hours (preferably 16 h) until the reaction had proceeded to completion as judged by TLC or HPLC analysis. Where necessary, additional acid is added to achieve complete conversion.
  • the solvents are then removed under reduced pressure.
  • the crude product can be treated in two different ways. 1) The material is purified by chromatography, trituration, or crystallization.
  • TFA (0.078 g, 0.68 mmol) was added to a solution of 2-4-[(7-benzo[b]thiophen-2-yl-1H-indazole-5-carbonyl)-amino]-phenyl-indole-1-carboxylic acid tert-butyl ester (Preparation #7 then B, 0.100 g, 0.137 mmol) in CH 2 Cl 2 (2.5 mL, 0.039 mol) at about 0° C. The reaction mixture was stirred at about 0° C. for about 30 min then additional TFA (80 uL) was added. After about 30 min the reaction was allowed to warm to room temperature and stirred for about 16 hours, prior to the addition of more TFA (150 uL).
  • a mixture of an amine substrate and a base (for example, NaH, K 2 CO 3 , Cs 2 CO 3 , t-BuOK or Na 2 CO 3 , preferably Na 2 CO 3 ) (2-6 equivalents, preferably 2 equivalents) in an organic solvent (for example, THF, DME or DMF, preferably DMF) is stirred at about ⁇ 10-25° C. (preferably about 0° C.), for about 0-60 minutes (preferably 40 minutes) under an inert atmosphere.
  • An organic halide for example an organic bromide or an organic chloride, preferably an organic bromide
  • the reaction is stirred for about 4-96 hours (preferably about 24 hours) at about 20-100° C. (preferably about 60° C.).
  • the solvent is removed under reduced pressure and the crude product can be purified by chromatography, crystallization or used in the next step without further purification.
  • an amine and a base for example, pyridine, Et 3 N, or ethyl-diisopropyl-amine, preferably ethyl-diisopropyl-amine
  • a base for example, pyridine, Et 3 N, or ethyl-diisopropyl-amine, preferably ethyl-diisopropyl-amine
  • organic solvent for example, CH 2 Cl 2 , DME, or DMF, preferably DMF
  • a pyrrolo[3,2-d]pyrimidine (preferably 1 equivalent) followed by slow addition of a Mitsunobu coupling reagent (for example, diisopropyl azodicarboxylate, 1,1′-azobis(N,N-dimethylformamide) or diethyl azodicarboxylate, preferably diisopropyl azodicarboxylate) (2-4 equivalents, preferably 2 equivalents).
  • the reaction mixture is stirred at about 0 to 60° C. (preferably at 20° C.) for about 1-6 hours (preferably 2 hours).
  • the reaction mixture is then concentrated under reduced pressure and the residual crude product is purified by chromatography, crystallization or can be used in the next step without further purification.
  • a halide preferably 1.0 equivalent
  • an organic solvent for example, DMF or piperidine, preferably DMF
  • a terminal acetylene 1.0-12.0 equivalents, preferably 1.2 equivalent
  • a palladium catalyst for example, dichlorobis(triphenylphosphine) palladium(II), tetrakis (triphenylphosphine) palladium(0), preferably dichlorobis(triphenylphosphine) palladium(II)
  • a base for example, piperidine, triethylamine, preferably triethylamine
  • copper salt for example copper (I) bromide, copper (I) iodide, preferably copper (I) iodide) (0.03-0.15 equivalent, preferably 0.10 equivalent).
  • the reaction mixture is heated in a microwave reactor at about 80-130° C. (preferably about 120° C.) for about 2-40 minutes (preferably about 5 minutes).
  • the mixture is allowed to cool to ambient temperature.
  • the insoluble residue is removed by filtration, and the filtrate is concentrated under reduced pressure.
  • the residual crude product is purified by chromatography or crystallization.
  • N′4′-(7-bromo-1H-indazol-5-yl)-pyrimidine-2,4-diamine (Example #N.2.7, 0.060 g, 0.197 mmol) in DMF (1 mL) in a microwave tube was added phenylacetylene (0.028 ml, 0.256 mmol), copper (I) iodide (0.004 g, 0.020 mmol) and tetrakis (triphenylphosphine) palladium(0) (0.011 g, 0.009 mmol).
  • the tube was sealed and the reaction mixture was heated in a microwave at about 120° C. for about 5 minutes. The mixture was allowed to cool to ambient temperature.
  • an ester in an organic solvent for example, THF, MeOH, or 1,4-dioxane, preferably 1,4-dioxane
  • an organic solvent for example, THF, MeOH, or 1,4-dioxane, preferably 1,4-dioxane
  • an aqueous base solution for example, Na 2 CO 3 , NaOH or KOH, preferably Na 2 CO 3
  • the mixture is stirred at about 10-80° C. (preferably about 25° C.) for about 0.5-2 hours (preferably about 1 hour).
  • the mixture is acidified with an acid (preferably 1M HCl solution) to about pH 3. If a precipitation is formed it is filtered off and washed with water.
  • the solvent is removed under reduced pressure and the residue is partitioned between an aqueous acidic solution (for example HCl) and an organic solvent (for example EtOAc or CH 2 Cl 2 , preferable CH 2 Cl 2 ).
  • an aqueous acidic solution for example HCl
  • an organic solvent for example EtOAc or CH 2 Cl 2 , preferable CH 2 Cl 2 .
  • the organic layer is separated and the aqueous layer is further extracted with an organic solvent.
  • the combined organic extracts are dried over a desiccant.
  • the solvents are removed under reduced pressure to afford the product which can be further purified by crystallization or chromatography.
  • a solution of ⁇ [5-(2-amino-pyrimidin-4-ylamino)-1H-indazole-3-carbonyl]-amino ⁇ acetic acid methyl ester was prepared from Example N.2.8, B, 0.014 g, 0.043 mmol) and 2M aqueous NaOH solution (1.0 mL, 2.0 mmol) in 1,4-dioxane (2 mL) was stirred at ambient temperature for about 30 minutes. The reaction mixture was acidified with 1M aqueous HCl solution until pH 3.
  • an acid chloride and an amine preferably 1 equivalent
  • an organic solvent for example, pyridine, CH 2 Cl 2 or 1,2-dichloroethane, preferably CH 2 Cl 2
  • a base for example Et 3 N or pyridine, preferably pyridine.
  • the reaction mixture is stirred at about 0-50° C. (preferably about 25° C.) for about 0.5-20 hours (preferably about 1 hour).
  • the mixture is diluted with an organic solvent (for example, EtOAc or CH 2 Cl 2 , preferably CH 2 Cl 2 ) and washed with water, then dried under reduced pressure.
  • the product can be further purified by crystallization or chromatography.
  • a mixture of an ortho-halobenzonitrile (1 equivalent) in 30% aqueous hydrazine (1-10 equivalents, preferably about 3 equivalents) is heated in a sealed tube to about 50-200° C. (preferably about 120° C.) for about 30-120 minutes (preferably about 45 minutes).
  • the mixture is cooled to ambient temperature and the precipitation is filtered off, washed with water and dried under reduced pressure to yield the desired indazole.
  • an aryl halide for example, an aryl bromide, aryl chloride or an aryl iodide, preferably an aryl iodide
  • an inorganic base for example, KF, Na 2 CO 3 or Cs 2 CO 3 , preferably Cs 2 CO 3
  • a degassed organic solvent for example THF, DME, DMF, 1,4-dioxane, toluene, preferably 1,4-dioxane
  • a palladium catalyst for example tris(benzylideneacetone)dipalladium (0) and XANTPHOS, tetrakis(triphenylphosphine)palladium(0), bis(acetato)triphenylphosphinepalladium(II) ( ⁇ 5% Pd
  • the reaction mixture is heated at about 40-150° C. (preferably about 95° C.) for about 0.5-24 hours (preferably about 2 hours) or at about 100-200° C. (preferably 150° C.) for about 5-60 minutes (preferably about 15 minutes) in a microwave under an inert atmosphere.
  • the reaction mixture is allowed to cool and the crude product is treated with an acid (preferably trifluoroacetic acid) (5-80 M solution, preferably 20M solution) optionally containing a cation scavenger (preferably triisopropyl silane) (0.7-2.5 equivalents, preferably 1.2 equivalents) for about 10-150 minutes (preferably about 15 minutes) at about 70-150° C. (preferably about 100° C.) in a sealed tube, either by conventional heating or by microwave heating. Solvents are removed under reduced pressure to give the product that can be further purified by crystallization or chromatography.
  • a mixture of 5-bromo-2-(4-methoxy-benzyl)-2H-indazole-7-carboxylic acid, phenylamide (prepared from preparation #12a using general procedures V and B, 75.0 mg, 0.20 mmol), 4-aminopyridine (24.0 mg, 0.25 mmol), cesium carbonate (195 mg, 0.60 mmol), and XANTPHOS (11.6 mg, 0.02 mmol) was suspended in 1,4-dioxane (2.5 mL) at ambient temperature under an inert atmosphere. Tris(dibenzylideneacetone)dipalladium(0) (9.2 mg, 0.01 mmol) was added and nitrogen gas was bubbled through the resulting suspension for about 5 minutes.
  • the reaction mixture was heated at about 95° C. for about 2 hours.
  • the resulting mixture was allowed to cool to ambient temperature and filtered through a celite pad and the solvent was removed under reduced pressure.
  • the residue was dissolved in trifluoroacetic acid (4.0 mL) containing triisopropylsilane (51.0 uL, 0.25 mmol) and the mixture was heated at about 100° C. in a microwave reactor for about 15 minutes.
  • an organic solvent for example MeOH, EtOH, i-PrOH or dioxane, preferably MeOH
  • an acid for example aqueous HCl or PPTS, preferably PPTS
  • the reaction mixture is stirred at about 10-85° C. (preferably about 25° C.) for about 1-24 hours (preferably about 6 hours).
  • the reaction mixture was diluted with an organic solvent (for example EtOAc, Et 2 O, or CH 2 Cl 2 , preferably EtOAc) and was washed with an aqueous basic solution (for example aqueous Na 2 CO 3 or NaOH, preferably Na 2 CO 3 ).
  • the solvent is evaporated and the product can be further purified by crystallization or chromatography.
  • PPTS (0.019 g, 0.075 mmol) was added to a mixture of 4- ⁇ 7-[(E)-4-(tetrahydropyran-2-yloxy)-but-1-enyl]-1H-indazol-5-ylamino ⁇ -pyrimidine-2-amine (prepared from Example #N.2.7 reacted with trans-2-[3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)allyoxy]-tetrahydropyran using general procedure F, 0.037 g, 0.100 mmol) in MeOH (3.0 mL). The reaction mixture was stirred at about 25° C. for about 5 hours.
  • a solution of a Cbz protected amine substrate and formic acid (5-20 equivalents, preferably 10 equivalents) in an organic solvent (for example methanol or ethanol, preferably methanol) is added dropwise to a suspension of Palladium black (1-10 equivalents, preferably 2 equivalents) in an organic solvent (for example methanol or ethanol, preferably methanol).
  • the resulting mixture is allowed to stir at room temperature for about 2-20 hours (preferably 16 hours).
  • the suspension is filtered though a plug of celite and the solvent is removed under reduced pressure.
  • the resulting crude product can be purified by titration with an appropriate solvent (water, ethanol, toluene or ethyl acetate, preferably ethanol) or by chromatography.
  • Benzyl isocyanate (0.25 mL, 2.0 mmol) was added to 3-iodo-1H-indazol-5-ylamine (Preparation #28, 0.50 g, 1.9 mmol) in THF (10 mL). After stirring at ambient temperature for about 2.5 hours, the mixture was filtered and washed with Et 2 O to give 1-benzyl-3-(3-iodo-1H-indazol-5-yl)-urea (0.41 g, 55%) as an ivory solid; RP-HPLC (Table 1, Method e) R t 0.98 min, m/z (ESI + ) 393.0 (M+H) + .
  • the resulting solid was purified by preparative HPLC (Thermo Hypersil-Keystone 250 ⁇ 21.2 mm 8 ⁇ Hypersil® HS C18 column; 5% CH 3 CN/50 mM aqueous ammonium acetate hold for 5 min, 5-100% CH 3 CN/50 mM aqueous ammonium acetate over 30 min, hold at 100% CH 3 CN for 5 minutes, 21 mL/min) to give an impure solid that was triturated with heptane and filtered to give 2-amino-4-(7-benzo[b]thiophen-2-yl-1H-indazol-5-ylamino)-7H-pyrrolo[2,3-d]pyrimidine-5-carboxamide (6.9 mg, 7%); RP-HPLC (Table 1, Method e) R t 1.64 min; m/z (ESI + ): 441.0 (M+H) + .
  • Phosphoryl chloride 43 ⁇ L, 0.46 mmol was added dropwise to DMF (0.17 mL) at about 0° C. After about 20 min, a solution of 5-(4,4,5,5-tetramethyl-[1,3,2]dioxaborolan-2-yl)-1H-indole (Aldrich, 0.10 g, 0.42 mmol) in DMF (0.70 mL) was added dropwise. The reaction was allowed to warm slowly to ambient temperature.
  • reaction mixture was concentrated under reduced pressure to give a crude mixture containing 5-(4,4,5,5-tetramethyl-[1,3,2]dioxaborolan-2-yl)-1H-indole-3-carboxaldehyde.
  • the mixture was dissolved in DME (3.0 mL) and transferred to a microwave vial.
  • N 4 -(7-bromo-1H-indazol-5-yl)-pyrimidine-2,4-diamine (Example #N.2.7, 0.10 g, 0.34 mmol), Pd(PPh 3 ) 4 (0.039 g, 0.034 mmol), and Na 2 CO 3 (2.0 M in water, 2.1 mL, 4.2 mmol) were added and the vial was heated in CEM microwave at about 150° C. for about 10 min. The reaction was diluted with water then extracted with EtOAc (3 ⁇ 15 mL). The precipitate present at the layer interface was kept with the organics. The combined organic layers were washed with brine, dried over Na 2 SO 4 , decanted, and concentrated.
  • a microwave vial containing N 4 -(7-5-[(allylamino)methyl]-1-benzothien-2-yl-1H-indazol-5-yl)pyrimidine-2,4-diamine triacetate (Example #0.3.8, 0.022 g, 0.036 mmol), chlorotris(triphenylphosphine)-rhodium(I) (8.4 mg, 0.0090 mmol), CH 3 CN (0.84 mL), and H 2 O (0.16 mL) was heated in a CEM microwave at about 120° C. for about 30 min.
  • N-Iodosuccinimide (3.2 g, 14.2 mmol) was added portionwise to an ice-cold stirred suspension of N,N-dimethyl-N′-(5H-pyrrolo[3,2-d]pyrimidin-2-yl)-formamidine ( Journal of Medicinal Chemistry, 2003, 46(14), 3060-3071, 2.68 g, 14.2 mmol) in DMF (50 mL). After stirring at ambient temperature overnight, the reaction was concentrated, dissolved in DCM (40 mL) and the product was precipitated using EtOAc (250 mL).
  • reaction mixture was concentrated under reduced pressure.
  • the resulting solid was triturated with MeOH/H 2 O (1:1, 1 L) then filtered, washed with additional MeOH/H 2 O (1:1, 500 mL), and dried in a vacuum oven at about 50-60° C.
  • Lithium hydroxide monohydrate (2.25 g, 53.5 mmol) was added to a suspension of methyl-7-benzo[b]thiophen-2-yl-1H-indazole-5-methanoate (3.37 g, 10.71 mmol) in a mixture of 1,4-dioxane (65 mL) and water (9 mL). The mixture was heated to about 65° C. for about 18 hours then cooled to about 40° C. and filtered.
  • the mixture was sealed and heated at about 80° C. for about 1 hour then cooled to ambient temperature, filtered, and concentrated.
  • the reaction mixture was heated at about 90° C. for about 4 hours.
  • the reaction was cooled to ambient temperature and the layers separated.
  • the organic layer was filtered through a silica gel pad, eluting with 5% MeOH/EtOAc and the filtrate was concentrated under reduced pressure.
  • the residue was dissolved in trifluoroacetic acid (50 mL) containing triisopropylsilane (2.05 ml, 10.0 mmol) and the mixture was heated in a sealed tube at about 100° C. for about 30 minutes then cooled to ambient temperature and concentrate under reduced pressure.
  • the residue was triturated with ether (100 mL) and the resulting solid was collected by filtration.
  • the solid was then dissolved in methanol (75 mL), treated with aqueous sodium hydroxide (2N, 50 ml, 100 mmol) and heated at reflux for about 6 hours.
  • the reaction was filtered hot, then cooled and acidified with acetic acid to about pH 5.5.
  • the product was filtered off, washed with water (2 ⁇ 10 mL) and dried to yield 5-(5-amino-1H-pyrrolo[2,3-c]pyridin-3-yl)-1H-indazole-7-carboxylic acid (2.60 g, 78%) as the acetate salt.
  • the acetate salt was dissolved in methanol (300 mL) and aqueous HCl (2N, 25 mL) was added.
  • aqueous layer was acidified using aqueous hydrochloric acid solution (1 N) and the resulting solid was collected by filtration, washed with water (3 ⁇ 5 mL), and dried to yield the N-(7-benzo[b]thiophen-2-yl-1H-indazol-5-yl)-oxamate (76 mg, 99%); LC/MS (Table 1, Method e) R t 0.89 min; m/z: [M ⁇ H] ⁇ 336.
  • a slurry containing (5-bromo-2-fluoro-phenyl)-(1H-pyrrol-2-yl)-methanone (Preparation #16b, 2.40 g, 8.96 mmol) and hydrazine hydrate (35 mL) was sealed and heated by microwave at about 150° C. for about 1 hour.
  • the reaction was cooled to ambient temperature and diluted with water (150 mL).
  • the product was extracted with EtOAc (150 mL), washed with 2N HCl solution (50 mL), saturated NaHCO 3 solution (50 mL) and finally a saturated NaCl solution.
  • Di-tert-butyldicarbonate (0.068 g, 0.31 mmol) was added to a solution of 1H-indole-4-carboxylic acid methyl ester (Aldrich, 0.050 g, 28 mmol) and 4-dimethylaminopyridine (0.7 mg, 0.006 mmol) in methylene chloride (1.0 mL, 0.016 mol). After 45 minutes, the mixture was quenched and acidified with 1N HCl to about a pH 4. Methylene chloride was added and the layers were separated.
  • Di-tert-butyldicarbonate (16 g, 0.071 mol) was added to a solution of 1H-indole-7-carboxylic acid methyl ester (Acros chemicals, 5.0 g, 0.028 mol) and 4-dimethylaminopyridine (0.3 g, 0.003 mol) in methylene chloride (100 mL). The mixture was heated at about 45° C. for about 16 hours. The mixture was quenched and acidified with 1N HCl to about a pH 4. Methylene chloride was added and the layers were separated.
  • 1H-indole-7-carboxylic acid methyl ester Acros chemicals, 5.0 g, 0.028 mol
  • 4-dimethylaminopyridine 0.3 g, 0.003 mol
  • Methylene chloride was added and the layers were separated.
  • Triisopropyl borate (870 ⁇ L, 0.0038 mol) was added to a solution of indole-1,4-dicarboxylic acid 1-tert-butyl ester 4-methyl ester (Preparation #18, 0.70 g, 0.0025 mol) in THF (36 mL). The solution was cooled to about 0-5° C. and lithium diisopropylamide in tetrahydrofuran (2 M, 2.8 mL) was added slowly over about 1 hour. After about another 1.5 hours, the mixture was quenched with 1N HCl, and the layers were separated.
  • reaction mixture was diluted with water (30 mL) and then filtered, washed with additional water to give (2-amino-6-chloro-5-formyl-pyrimidin-4-ylsulfanyl)-acetic acid methyl ester (1.801 g, 66%) as a yellow solid; RP-HPLC (Table 1, Method a) R t 1.85 min, m/z (ESI+) 262.0 (M+H) + .
  • the crude reaction mixture was purified by reverse phase chromatography (RP-HPLC (Rainin C18, 8 mm, 300 ⁇ , 35 cm; 5-100% acetonitrile/0.1 M ammonium acetate over 20 min, 100% acetonitrile hold 10 minutes, 21 mL/min) to afford 2-(7-bromo-1H-indazol-5-ylamino)-2-methyl-propan-1-ol (0.033 g, 0.116 mmol).
  • RP-HPLC Table 1, Method e

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Heterocyclic Carbon Compounds Containing A Hetero Ring Having Oxygen Or Sulfur (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Nitrogen And Oxygen Or Sulfur-Condensed Heterocyclic Ring Systems (AREA)

Abstract

Novel compounds of Formula (I) or pharmaceutically acceptable salts, prodrugs and biologically active metabolites thereof of Formula (I)
Figure US20120053345A1-20120301-C00001
wherein the substituents are as defined herein, which are useful as therapeutic agents.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application claims priority to U.S. NonProvisional application Ser. No. 11/731,950 filed Apr. 2, 2007, which claims priority to U.S. Provisional Application Ser. No. 60/788,553 filed Mar. 31, 2006, all of which is incorporated by reference in its entirety.
  • BACKGROUND OF THE INVENTION
  • Protein phosphorylation, at specific amino acid residues, is important for the regulation of many cellular processes including cell cycle progression and division, signal transduction, and apoptosis. The phosphorylation is usually a transfer reaction of the terminal phosphate group from ATP to the protein substrate. The specific structure in the target substrate to which the phosphate is transferred is a tyrosine, serine or threonine residue. Since these amino acid residues are the target structures for the phosphoryl transfer, and since most kinases target either tyrosine or both serine and threonine, these protein kinase enzymes are commonly referred to as tyrosine kinases or serine/threonine (S/T) kinases. The phosphorylation reactions, and counteracting phosphatase reactions, on the tyrosine, serine and threonine residues are involved in many cellular processes that underlie responses to diverse intracellular signals, regulation of cellular functions, and activation or deactivation of cellular processes. A cascade of protein kinases often function in intracellular signal transduction. Protein kinases can be found integrated into the plasma membrane, as cytoplasmic enzymes or localized in the nucleus, often as components of enzyme complexes. In many instances, these protein kinases are an essential element of enzyme and structural protein complexes that determine where and when a cellular process occurs within a cell. Given the importance and diversity of protein kinase function, it is not surprising that phosphorylation events are required in cellular processes associated with many diseases such as cancer, diabetes, inflammation, and hypertension.
  • The identification of effective small molecules that specifically inhibit protein kinases involved in abnormal or inappropriate cell proliferation, signaling, differentiation, protein production, or metabolism is therefore desirable. The identification of methods and compounds that specifically inhibit the function of kinases that are involved in immune modulation or proliferative disorders is particularly desirable.
  • The present invention provides novel compounds that inhibit one or more receptor, or non-receptor, tyrosine or S/T kinase.
  • SUMMARY OF THE INVENTION
  • Figure US20120053345A1-20120301-C00002
  • The present invention provides a compound of Formula (I)
  • wherein
    • R1 is selected from the group consisting of H, benzyl substituted with OCH3, optionally substituted (C1-C3)alkyl, pyrimidine substituted with NH2 and amino(C1-C3)alkyl;
    • R3 is selected from the group consisting of H, halogen, NH2, OH, COOH, —C(O)—NH—CH2—C(O)—OCH3, —NH—CH2-phenyl, —C(O)-pyridinyl, —NH—C(O)-cyclobutyl and —NH—C(O)-phenyl wherein phenyl is optionally substituted with either N(CH3)2 or OCH3; or
    • R3 is selected from the optionally substituted group consisting of (C1-C6)alkyl, benzo[b]thienyl, 2,3-dihydrobenzofuranyl, indolyl, isoquinolinyl, morpholinyl, naphthyl, phenyl, piperazinyl, pyrazolyl, pyridinyl, pyrimidinyl, pyrrolyl, quinolinyl and thienyl;
      • wherein the substituent is selected from one or more CH3, NH2, Cl, F, dimethylamino, OH, CH2OH, —C(O)NH2, COOH, CF3, isopropyl, OCF3, OCH3, —O—CH2-phenyl, CN, OCH2CH3, —NH—C(O)-cyclobutyl, —NH—C(O)-phenyl, NH—C(O)—CH3, NHC(O)CH3, N(CH3)2, S(O)2CH3 and C(O)NH-phenyl; or
    • R3 is —C(O)—NY100—(C(Y100)2)x—Ra wherein
      • x is 0, 1, 2 or 3;
      • Y100 is independently H or (C1-C3)alkyl; and
      • Ra is —C(O)—CH3 or is selected from the optionally substituted group (C1-C3)alkyl, amino, aminoalkyl, benzimidazolyl, benzo[b]thienyl, benzotriazolyl, biphenyl, 1,3-dihydrobenzimidazolyl, 1,3-dihydrobenzimidazolyl-2-one, imidazolyl, indolyl, naphthyl, phenyl, pyrazolyl, pyridinyl, pyrimidinyl, tetrahydropyranyl and thiazolyl; or
    • R3 is A-B wherein A is connected to the indazole and
      • A is selected from the group consisting of —C≡C, —C≡C-phenyl, indazolyl, phenyl, pyridinyl and thienyl;
      • B is selected from the group consisting of benzyloxy, morpholinyl, phenyl, thienyl, t-butyl, —NH—C(O)-cyclobutyl and —NH—C(O)-phenyl;
    • R4 is H or NH2;
    • R5 is selected from the group consisting of H, NH2, NO2, halo; or
    • R5 is selected from the optionally substituted group consisting of benzimidazolyl, 3,4-dihydrobenzo[1,4]thiazinyl, benzyloxyphenyl, furo[3,2-c]pyridine, indazolyl, indolyl, isoquinolinyl, phenyl, pyrazolo[3,4-d]pyrimidine, pyrazinyl, pyrazolyl, pyridinyl, pyrrolo[2,3-b]pyridinyl, pyrrolo[2,3-c]pyridinyl, pyrrolo[2,3-d]pyridinyl, pyrimidinyl, pyrrolo[3,2-d]pyridine, pyrrolo[2,3-d]pyrimidinyl, pyridinyl, pyrrolyl, quinolinyl, quinazolinyl, thienyl, thieno[2,3-c]pyridinyl, thieno[2,3-d]pyrimidine, thieno[3,2-c]pyridine, 7-azaindolinyl and 7-azaindolyl; or
    • R5 is —C(O)—Rb; wherein
      • Rb is selected from the group consisting of OH, (C1-C3)alkoxy, phenyl, optionally substituted piperidinyl, optionally substituted pyridinyl and optionally substituted pyrrolidinyl; or
      • Rb is D-E wherein D is attached to the C(O) and
        • D is selected from the group consisting of piperidinyl and pyrrolidinyl;
        • E is selected from the group consisting of pyridinyl and pyrimidinylaminemethyl;
    • R5 is —C(O)—NH—(CH2)a—Rc; wherein
      • a is 0, 1, 2 or 3;
      • Rc is —CONH2 or
      • Rc is selected from the optionally substituted group consisting of benzimidazolyl, benzothiazolyl, benzo[b]thiophenyl, dimethylamino, fluorene, imidazolyl, indanyl, indazolyl, isoxazolyl, oxazolyl, phenyl, piperidinyl, pyrazolyl, pyridinyl, quinazolinyl, thiadiazolyl and 1,2,4-triazolyl; or
      • Rc is J100-J200 wherein J100 is attached to (CH2)x and
        • J100 is selected from the optionally substituted group consisting of isoxazolyl, piperazinyl, pyrazolyl pyridinyl, and phenyl; and
        • J200 is selected from the group consisting of benzimidazolyl, benzoxazolyl, cyclohexyl, furanyl, imidazo[1,2-a]pyridinyl, indolyl, isoxazolyl, —NH—C(O)-phenyl, phenoxy and optionally substituted phenyl;
    • R5 is —NH—C(O)—(CH2)n—Rd; wherein
      • n is 0 to 3;
      • Rd is —C(CH3)2—CH2—C(O)—CH3; or
      • Rd is selected from the optionally substituted group of (C1-C2)alkoxy, alkylamino, benzimidazolyl, benzo[1,3]dioxazolyl, benzo[1,2,5]oxadiazolyl, benzotriazolyl, benzo[b]thienyl, benzofuranyl, benzyloxy, cyclopropyl, cyclohexyl, chromenyl, dimethylamino, furanyl, hexahydropyrimidinyl, imidazolyl, imidazo[2,1-b]thiazolyl, imidazo[2,1-b]thiazolyl, imidazolidinyl, indolyl, isoxazolyl, morpholinyl, phenyl, piperazinyl, piperidinyl, pyrazinyl, pyrazolyl, pyrazolo[1,5-a]pyrimidinyl, pyridazinyl, pyridinyl, pyrimidinyl, pyrrolidinyl, pyrrolyl, quinolinyl, quinoxalinyl, tetrahydrobenzofuranyl, tetrahydrofuranyl, thiazolyl, thieno[2,3-d]pyrimidinyl, thienyl, 2,3-dihydrothiazolo[3,2-a]pyrimidine, —S-pyrimidinyl, —O-phenyl, —O—Si(CH3)2—C(CH3)3, —NH—S(O)2-phenyl, —NH—C(O)—NH2, 1,2,3,4-tetrahydronaphthyridinyl or N(CH3)2; or
      • Rd is M-Q wherein M is attached to the (CH2)n and
      • M is selected from the group consisting of optionally substituted methylene, cyclopropylidene, optionally substituted isoxazolyl, phenyl, pyrazolyl, —NH—C(O) and optionally substituted 1,3,5-triazinyl;
      • Q is selected from the group consisting of furanyl, morpholinyl, phenyl, phenylamine and optionally substituted 1,3,4-thiadiazolyl;
    • R5 is —NH—CH2—C(Y200)2—Re wherein Y200 is independently H or (C1-C3)alkyl and Re is selected from the optionally substituted group of (C1-C6)alkoxy, imidazolyl, phenyl, piperidinyl, pyrrolidinyl and 1,2,3,4-tetrahydro[1,8]naphthyridine; or
    • R5 is —NH—C(O)—N(Rf)2 wherein Rf is independently H or optionally substituted (C1-C3)alkyl; or
    • R5 is —NH—(C(O))m—NY300—(CH2)p—Rg; wherein
      • m is 0, 1 or 2;
      • Y300 is H or optionally substituted (C1-C3)alkyl;
      • p is 1 or 2; and
      • Rg is selected from the optionally substituted group of amino, (C1-C2)alkoxy, benzo[1,3]dioxazolyl, benzothiazolyl, benzo[1,4]oxazinyl, benzo[1,2,5]thiadiazolyl, imidazol[1,2-a]pyridinyl, imidazolyl, isoxazolyl, morpholinyl, oxazolyl, phenyl, piperidinyl, pyrazinyl, pyrazolyl, pyridinyl, pyrrolyl, tetrahydropyranyl, thiazolyl and triazolyl; or
      • Rg is W—X wherein W is attached to the (CH2)p and
        • W is thiazolyl and X is thienyl;
    • R5 is —NH—S(O)2Rh wherein Rh is selected from the group of optionally substituted benzo[1,2,5]oxodiazolyl, benzo[1,2,5]thiadiazolyl, imidazolyl, isoxazolyl, oxazolyl, benzo[1,4]oxazinyl, pyrazolyl, phenyl, quinolinyl, thiazolyl, thienyl and thienyl; or
      • Rh is T-U wherein T is attached to the S(O)2 and
        • T is phenyl or thienyl;
        • U is selected from the group consisting of pyridinyl, optionally substituted thiazolyl and —NH-pyrimidinyl wherein the pyrimidinyl can be optionally substituted; or
    • R5 is —N(Y400)—Ri wherein
      • Y400 is H or Y400 is selected from the optionally substituted group consisting of (C1-C3)alkyl, amino(C1-C3)alkyl and pyridinylmethyl; and
      • Ri is selected from the optionally substituted group of (C1-C3)alkyl, 6-azaindolyl cyclobutenyl, phenyl, purinyl, pyrazinyl, pyrazolo[1,5-a]pyrimidine, pyridazinyl, pyridinyl, pyrimidinyl, pyrrolo[2,3-c]pyridinyl, pyrrolo[2,3-b]pyrimidinyl, pyrrolo[3,2-d]pyrimidinyl, pyrrolo[3,4-b]pyrimidinyl, pyrrolo[2,3-d]pyrimidinyl, quinazolinyl, thieno[3,2-d]pyrimidinyl, thieno[3,2-b]pyridinyl, and triazinyl; or
      • Ri is V—W wherein V is attached to the nitrogen and
        • V is a bond or is selected from the optionally substituted group consisting of isoquinolinyl, pyridinyl, pyrimidinyl and pyrrolo[3,2-d]pyrimidinyl;
        • W is selected from the optionally substituted group consisting of (C1-C3)alkyl, alkoxyalkyl, cyclopentyl, morpholinyl, phenyl, pyrimidinyl, pyrrolidinyl, thieno[3,2-b]pyridinyl, —NH-phenyl, —NH—CH2—CH2—N(CH3)2, —NH—NH2, NH—C(O)—CH3, CH2-phenyl, NH—C(O)-furanyl, S-isopropyl, S-naphthyl, S-phenyl and S—CH2—CH2—NH2; or
    • R5 is Z100—Z200 wherein Z100 is attached to the indazole and
      • Z100 is selected from the group consisting of butyl, ethyl, indazolyl, optionally substituted phenyl, optionally substituted pyridinyl, optionally substituted pyrimidinyl, pyrrolo[3,2-d]pyrimidinyl and optionally substituted thienyl;
      • Z200 is selected from the group consisting of —C(O)—NH—CH2CH2—NH2, NH2, —NH—C(O)-thienyl, —NH—C(O)—CH(CH3)2, —NH—C(O)—CH3, —NH—C(O)C(CH3)3, —NH—C(O)—NH-furanyl, —NH—C(O)—NH-phenyl, benzo[b]thienyl, morpholinylethyl, phenyl, piperazinyl, piperidinyl, pyrazolyl and tetrazolyl; or
    • R5 is —NH—C(O)—Y500—C(O)—Rk; wherein
      • Y500 is optionally substituted (C1-C3)alkyl; and
      • Rk is H or Rk is selected from the optionally substituted group consisting of phenyl, phenylamino and thienyl; or
    • R5 is —NH—C(O)—(CH2)y—NH-T-Rm; wherein
      • y is 1 or 2;
      • T is C(O) or S(O)2; and
      • Rm is selected from the group consisting of furanyl, phenyl and thienyl;
    • R6 is H or R6 is selected from the optionally substituted group consisting of (C1)alkoxy, (C1-C3)alkyl, benzo[b]thienyl, —NH-pyrimidinyl, —NH—S(O)2-phenyl-NH-pyrimidinyl, —NH—C(O)-benzo[b]thienyl, pyrrolo[2,3-b]pyrimidinyl and pyridinyl; and
    • R7 is selected from the group consisting of H, halo, NH2, or R7 is selected from optionally substituted group consisting of (C2-C5)alkenyl, (C2-C5)alkynyl, aminoalkynyl, benzofuranyl, benzothiazolyl, benzo[b]thienyl, furanyl, indolyl, isoquinolinyl, naphthyl, phenyl, phenylalkyl, phenyl(C2-C5)alkenyl, phenyl(C2-C5)alkynyl, piperidinyl, pyridinyl, pyrimidinyl, pyrrolyl, quinolinyl, quinoxalinyl, thieno[2,3-b]pyridinyl, thienyl, —NH—S(O)2—CH3, —NH—C(O)—CH3, —NH—C(O)-phenyl, —C(O)—NH—CH2-phenyl and —C(O)—NH-phenyl; or
    • R7 is Y—Z wherein Y is attached to the indazole; and
      • Y is benzo[b]thienyl or thienyl; and
      • Z is selected from the group consisting of phenyl, thienyl, CH2NHCH2CH2-morpholinyl and substituted piperazinyl; or
    • R7 is —C(O)—NH—(CH2)r-phenyl wherein r is 0 or 1 and the phenyl is optionally substituted;
      provided that the compound is not
  • Figure US20120053345A1-20120301-C00003
  • wherein
    • R3 is selected from the group consisting of H, OH and COOH;
    • R5 is H or NO2;
    • R7 is H or NH2
    • R100 is OCH3 and
    • R200 is H or —C(O)—OCH3;
      provided that the compound is not
  • Figure US20120053345A1-20120301-C00004
  • wherein
    • R3 is NH2 or phenyl; and
    • R5 is H or NO2;
      provided that the compound is not
  • Figure US20120053345A1-20120301-C00005
  • wherein
    • R1 is H, methyl or propyl;
    • R7 is H, F or methyl; and
    • R is H, methyl, OH, NH, or OCH3;
      provided that the compound is not
  • Figure US20120053345A1-20120301-C00006
  • wherein R3 is selected from the group consisting of I, Br, COOH, NH2, thienyl, pyridinyl, pyrrolyl, —C(O)—NH—CH—(CH2OH)2, —C(O)—NH—CH2—X100,
  • Figure US20120053345A1-20120301-C00007
      • wherein X100 is pyridinyl or phenyl optionally substituted with methyl;
        provided that the compound is not
  • Figure US20120053345A1-20120301-C00008
  • wherein
    • R3 is morpholinyl or 4-methylpiperazinyl;
    • Rc is H, C1 or NO2;
    • R6 is H or C1 and
    • R7 is H, C1 or NO2;
      provided that the compound is not
  • Figure US20120053345A1-20120301-C00009
  • wherein
    • R6 is H, methyl or OCH3;
    • R6 is H or OCH3;
    • L100 is H or isopropyl; and
    • X200 is phenyl or 4-chlorophenyl;
      provided that the compound is not
  • Figure US20120053345A1-20120301-C00010
  • wherein
    • R1 is H or CH3 and X300 is benzyl, phenyl, 2-aminophenyl or 2-hydroxyphenyl; and
      provided that the compound is not
  • Figure US20120053345A1-20120301-C00011
  • wherein
    • R1 is H, CH2OH, methyl, phenyl or 4-methoxybenzyl;
    • R3 is H, I, pyridinyl or
  • Figure US20120053345A1-20120301-C00012
  • and
    • R5 is H, Br, F, C(OH), —C(O)—OCH2CH3 or —NH—C(O)—NH-benzyl.
  • In a second embodiment, the invention provides compounds or pharmaceutically acceptable salts thereof according to any of the foregoing inventions wherein
    • R1 is H or pyrimidinyl substituted with NH2;
    • R3 is selected from the group consisting of H, CH3, OH, Cl, benzo[b]thienyl, 2,3-dihydrobenzofuranyl, indolyl, naphthyl, phenyl, pyrazolyl, pyridinyl, pyrimidinyl, pyrrolyl, quinolinyl, thienyl, —NH—C(O)-cyclobutyl and —NH—C(O)-phenyl; wherein
      • the indolyl is optionally substituted with CH3;
      • the naphthyl is optionally substituted with OCH3 or OH; and
      • the phenyl optionally substituted with one or more substituents selected from the group consisting of CH3, NH2, Cl, F, N(CH3)2, OH, CH2OH, C(O)NH2, COOH, CF3, OCF3, OCH3, CN, OCH2CH3, NHC(O)CH3, —S(O)2CH3 and —C(O)—NH-pheny; or
    • R3 is —C(O)—NY100—(C(Y100)2)x—Ra wherein
      • x is 0 or 1;
      • Y100 is H;
      • Ra is selected from the optionally substituted group consisting of benzo[b]thienyl, benzimidazolyl, 1,3-dihydrobenzimidazolyl-2-one, benzotriazolyl, biphenyl, 1,3-dihydrobenzimidazolyl, indolyl, naphthyl and phenyl; wherein
        • the naphthyl is substituted with OH or OCH3;
        • the phenyl is optionally substituted with one or more Cl, F, OH, CH2OH, CH2CH2OH, COOH, C(O)NH2, N(CH3)2 or methyl; or
    • R3 is A-B wherein
      • A is selected from the group consisting of —C≡C, —C≡C-phenyl, phenyl and thienyl; and
      • B is selected from the group consisting of benzyloxy, phenyl, thienyl, —NH—C(O)-cyclobutyl and —NH—C(O)-phenyl;
    • R4 is H;
    • R5 is pyridinyl substituted with C(O)H, CH2OH, SCH2CH2NH2 or NH2; or
    • R5 is selected from the group consisting of
  • Figure US20120053345A1-20120301-C00013
    •  wherein
      • E is selected from the group consisting of H, OH, CH3, —CH2CH2NH2, CH2CH2CH2OH, CH2CH2OCH3, CH2CH2CH2NH2, CH2C(O)OH, CH2CH2C(O)OH, CH2CH2C(O)NH2, CH2CH2C(O)OCH3, CH2CH2CH2OCH3, NHCH2CH2CH3, CH2CH2C(O)NH(CH3), CH2CH2C(O)N(CH3)2, C(O)NHCH2CH2NH(CH3), NHCH2CH2OCH3, NHCH2CH2OH, NHCH2CH2N(CH3)2, isopropyl, CH2C(O)NH2, CH2CH(CH3)C(O)OH, CH2CH2CH2C(O)OH, CH2CH(CH3)C(O)OCH3, CH2CH2CH2CH2NH2, CH2CH2CH2C(O)NH2, N(CH3)2, morpholinylethyl, piperidinylethyl,
  • Figure US20120053345A1-20120301-C00014
      •  and 4-methylpiperazinylcyclohexyl;
      • E300 is H or CH2CH2OCH3;
      • G is selected from the group consisting of H, C1, NH2, CH2CH2C(O)NHCH2CH2NH2, C(O)NH2, C(O)NHCH2CH2NH2, C(O)NHCH2CH2N(CH3)2, CH2CH2NH2, CH2CH2CH2NH2, CH2CH2C(O)OH, CH2CH2C(O)NH2, NHCH2CH2N(CH3)2, NHCH2CH2-pyridinyl and NHCH2CH2NH2; or
    • R5 is —C(O)—NH—(CH2)a—Rc wherein
      • a is 0
      • Rc is benzimidazolyl or fluorene substituted with oxo; or
      • Rc is J100-J200 wherein
        • J100 is selected from the group consisting of pyrazolyl, pyridinyl, piperazinyl and phenyl; wherein
          • the phenyl is optionally substituted with one or more substituents selected from the group consisting of F, OH and OCH3;
          • the piperazinyl is substituted with methyl; and
        • J200 is selected from the group consisting of benzoxazolyl, benzimidazolyl, furanyl, imidazo[1,2-a]pyridinyl and 1,8a-dihydroimidazo[1,2-a]pyridinyl; or
    • R5 is —NH—C(O)—(CH2)n—Rd wherein
      • n is 0, 1 or 2; and
      • Rd is benzimidazolyl, benzo[b]thienyl, imidazolyl, phenyl or pyrazolo[1,5-a]pyrimidinyl;
      • wherein
        • the phenyl is substituted with NO2;
    • R5 is —NH—(C(O))m—NY300—(CH2)p—Rg wherein
      • m is 2;
      • Y300 is H;
      • p is 1; and
      • Rg is benzo[1,3]dioxazolyl; or
    • R5 is —N(Y400)—Ri wherein
      • Y400 is selected from H, CH2CH2CH2OH, CH2CH2NH2 or pyridinylmethyl;
      • Ri is V—W wherein
      • V is a bond or pyrimidinyl; and
      • W is pyrimidinyl substituted with NH2 or pyrrolidinyl substituted with OH; or
    • R5 is Z100—Z200 wherein
      • Z100 is thienyl or pyridinyl substituted with CH3 and Z200 is thienyl or NH—C(O)-furanyl;
    • R6 is selected from the group consisting of H, pyrrolo[2,3-b]pyrimidinyl and
  • Figure US20120053345A1-20120301-C00015
  • and
    • R7 is selected from the optionally substituted group consisting of H, Br, Cl, I, benzofuranyl, benzo[b]thienyl, furanyl, indolyl, naphthyl, phenyl, pyridinyl, pyrrolyl, quinolinyl, quinoxalinyl, thieno[2,3-b]pyridinyl, thienyl, —CH═CH-phenyl, —C≡C-phenyl, —C(O)—NH—CH2-phenyl and —C(O)—NH-phenyl wherein
      • the naphthyl is optionally substituted with OH or OCH3;
      • the benzo[b]thienyl is optionally substituted with OH, CH3, OCH3, N(CH3)2, OH, CH2═CHNHCH3, CH2NH2, CH2CH2NH2 or CH2NHCH2CH2N(CH3)2;
      • the indolyl is substituted with C(O)N(CH(CH3)2)2, CH2OH, CH2C(O)NH2, COOH, C(O)NH2, N(CH3)2 or S(O)2CH3; and
      • the phenyl is optionally substituted with one or more substituents selected from the group consisting of Cl, F, CH3, CH2OH, CN, —C(O)NH2, OH, OCH3, N(CH3)2, NH—C(O)CH3, —NH—S(O)2—CH3;
      • the thienyl is substituted with CH2OH; or
    • R7 is Y—Z wherein
      • Y is benzo[b]thienyl or thienyl; and
      • Z is selected from the optionally substituted group consisting of CH═CHNHCH3, NHCH3, CH2NH2, CH2CH2NH2, CH2NHCH3, CH2NHCH2CH2N(CH3)2, N(CH3)2, CH2NHCH2CH2-morpholinyl, benzo[b]thienyl, morpholinylmethyl, piperazinylmethylphenyl and thienyl; or
    • R7 is —C(O)—NH—(CH2)r-phenyl wherein
      • r is 0 or 1;
      • the phenyl is optionally substituted with NH2.
  • In a third embodiment, the invention provides compounds or pharmaceutically acceptable salts thereof according to any of the foregoing inventions wherein R1 is H or pyrimidinyl substituted with NH2;
    • R3 is selected from the group consisting of H, CH3, OH, Cl, benzo[b]thienyl, 2,3-dihydrobenzofuranyl, indolyl, naphthyl, phenyl, pyrazolyl, pyridinyl, pyrimidinyl, pyrrolyl, quinolinyl, thienyl, —NH—C(O)-cyclobutyl and —NH—C(O)-phenyl; wherein
      • the indolyl is optionally substituted with CH3;
      • the naphthyl is optionally substituted with OH; and
      • the phenyl optionally substituted with one or more substituents selected from the group consisting of OH, F, CH3, CF3, CN, —C(O)NH2, NH2, NHC(O)CH3, OCH3, OCF3, OCH2CH3, N(CH3)2, —C(O)—NH-phenyl and —S(O)2CH3; or
    • R3 is —C(O)—NY100—(C(Y100)2)x—Ra wherein
      • Y100 is H;
      • x is 0;
      • Ra is selected from the optionally substituted group consisting of benzimidazolyl, 1,3-dihydrobenzimidazolyl-2-one, benzotriazolyl, biphenyl, indolyl, naphthyl and phenyl; wherein
        • the naphthyl is substituted with OH or OCH3;
        • the phenyl is optionally substituted with one or more Cl, F, OH, CH2OH, CH2CH2OH, C(O)NH2, N(CH3)2 or methyl; or
    • R3 is A-B wherein
      • A is selected from the group consisting of phenyl and thienyl; and
      • B is selected from the group consisting of benzyloxy, phenyl, thienyl, —NH—C(O)-cyclobutyl and —NH—C(O)-phenyl;
    • R5 is pyridinyl substituted with NH2; or
    • R5 is selected from the group consisting of
  • Figure US20120053345A1-20120301-C00016
    •  wherein
      • E is selected from the group consisting of H, CH3, CH2C(O)OH, CH2CH2CH2OH, CH2CH2CH2NH2, CH2CH2C(O)OH, CH2CH2C(O)NH2, CH2CH2C(O)OCH3, CH2CH2CH2OCH3, NHCH2CH2CH3, CH2CH2C(O)NH(CH3), NHCH2CH2OCH3, NHCH2CH2OH, isopropyl, CH2C(O)NH2, CH2CH(CH3)C(O)OH, morpholinylethyl, piperidinylethyl, CH2CH2CH2C(O)OH, CH2CH(CH3)C(O)OCH3, CH2CH2NH2, CH2CH2CH2CH2NH2, CH2CH2CH2C(O)NH2, CH2CH2C(O)N(CH3)2, and N(CH3)2; and
      • G is selected from the group consisting of H, NH2, Cl, CH2CH2C(O)NHCH2CH2NH2, C(O)NHCH2CH2NH2, C(O)NHCH2CH2N(CH3)2, NHCH2CH2N(CH3)2, NHCH2CH2NH2, CH2CH2NH2, CH2CH2CH2NH2, CH2CH2C(O)OH, CH2CH2C(O)NH2 and NHCH2CH2-pyridinyl;
    • R5 is —C(O)—NH—(CH2)a—Rc wherein
      • a is 0;
      • Rc is J100-J200 wherein
        • J100 is pyrazolyl or phenyl wherein the phenyl is optionally substituted with OCH3; and
        • J200 is benzoxazolyl, benzimidazolyl, furanyl, imidazo[1,2-a]pyridinyl or 1,8a-dihydroimidazo[1,2-a]pyridinyl; or
    • R5 is —NH—C(O)—(CH2)n—Rd wherein
      • n is 0, 1 or 2; and
      • Rd is selected from the group consisting of benzimidazolyl, benzo[b]thieny, imidazolyl and pyrazolo[1,5-a]pyrimidinyl;
    • R5 is —N(Y400)—Ri wherein
      • Y400 is selected from H, CH2CH2CH2OH, CH2CH2NH2 or pyridinylmethyl;
      • Ri is V—W wherein
      • V is a bond or pyrimidinyl and W is pyrimidinyl substituted with NH2 or pyrrolidinyl substituted with OH;
    • R5 is Z100—Z200 wherein
      • Z100 is thienyl;
      • Z200 is thienyl;
    • R6 is H or
  • Figure US20120053345A1-20120301-C00017
  • and
    • R7 is selected from the optionally substituted group consisting of H, benzofuranyl, benzo[b]thienyl, furanyl, indolyl, naphthyl, quinolinyl, phenyl, pyrrolyl, quinoxalinyl, thienyl, thieno[2,3-b]pyridinyl, —CH═CH-phenyl, —C≡C-phenyl, —C(O)—NH—CH2-phenyl and —C(O)—NH-phenyl wherein
      • the benzo[b]thienyl is optionally substituted with OH, CH3, OCH3, N(CH3)2, CH2═CHNHCH3, CH2NH2, CH2CH2NH2 or CH2NHCH2CH2N(CH3)2;
      • the indolyl is substituted with methyl, CN, C(O)H, CH2CH2CH2NH2, CH2NHCH2CH═CH2, C(O)CH3, C(O)OCH3, OCH3, C(O)N(CH(CH3)2)2, CH2OH, CH2C(O)NH2, C(O)NH2, CH2NHCH2CH2N(CH3)2 or piperidinylmethyl; and
      • the phenyl is optionally substituted with one or more substituents selected from the group consisting of Cl, F, CH3, CH2OH, CN, —C(O)NH2, OH, OCH3, N(CH3)2 and —NH—S(O)2—CH3; or
    • R7 is Y—Z wherein
      • Y is benzo[b]thienyl or thienyl; and
      • Z is selected from the optionally substituted group consisting of CH═CHNHCH3, NHCH3, CH2NH2, CH2CH2NH2, CH2NHCH3, CH2NHCH2CH2N(CH3)2, N(CH3)2, CH2NHCH2CH2-morpholinyl, benzo[b]thienyl, morpholinylmethyl and piperazinylmethyl; wherein
        • the piperazinyl is optionally substituted with methyl.
  • In a fourth embodiment, the invention provides compounds or pharmaceutically acceptable salts thereof according to any of the foregoing inventions wherein R1 and R4 are H;
    • R3 is selected from the optionally substituted group consisting of H, OH, 2,3-dihydrobenzofuranyl, naphthyl, pyrazolyl and pyrrolyl; wherein
    • R3 is —C(O)—NY100—(C(Y100)2)x—Ra wherein
      • Y100 is H;
      • x is 0;
      • Ra is selected from the optionally substituted group consisting of naphthyl and phenyl; wherein
        • the naphthyl is optionally substituted with OH;
        • the phenyl is optionally substituted with OH; or
    • R3 is selected from the group consisting of —NH—C(O)-cyclobutyl and —NH—C(O)-phenyl;
    • R3 is A-B wherein
      • A is selected from the group consisting of phenyl and thienyl; and
      • B is selected from the group consisting of benzyloxy, phenyl and thienyl;
    • R5 is selected from the group consisting of
  • Figure US20120053345A1-20120301-C00018
    •  wherein
      • E is selected from the group consisting of H, CH2C(O)NH2, CH2CH(CH3)C(O)OCH3, CH2CH2CH2OH, CH2CH2C(O)OCH3, CH2CH2CH2OCH3, CH2CH2NH2, CH2CH2CH2NH2, CH2CH2C(O)OH, CH2CH2C(O)NH2, CH2CH2CH2CH2NH2, CH2CH2CH2OCH3, CH2CH2CH2C(O)OH, CH2CH2CH2C(O)NH2, CH2CH(CH3)C(O)OH, CH2CH2C(O)NH(CH3), CH2CH2C(O)N(CH3)2, N(CH3)2, isopropyl, morpholinylethyl and piperidinylethyl; and
      • G is H, NH2 or NHCH2CH2-pyridinyl or
    • R5 is —C(O)—NH—(CH2)a—Rc wherein
      • a is 0
      • Rc is J100-J200 wherein
        • J100 is phenyl optionally substituted with OCH3 and
        • J200 is imidazo[1,2-a]pyridinyl or 1,8a-dihydroimidazo[1,2-a]pyridinyl; or
    • R5 is —NH—C(O)—(CH2)n—Rd wherein
      • n is 2 and Rd is imidazolyl; or
    • R5 is Z100—Z200 wherein
      • Z100 is thienyl;
      • Z200 is thienyl;
    • R6 is H or
  • Figure US20120053345A1-20120301-C00019
  • and
    • R7 is selected from the optionally substituted group consisting of H, benzofuranyl, benzo[b]thienyl, indolyl, naphthyl, quinolinyl, CH═CH-phenyl, —C≡C-phenyl, —C(O)—NH—CH2-phenyl and —C(O)—NH-phenyl wherein
      • the benzo[b]thienyl is optionally substituted with OH, CH3, OCH3, N(CH3)2, CH2═CH2NHCH3, CH2NH2, CH2CH2NH2, CH2NHCH2CH2N(CH3)2, piperidinylmethyl or CH2NHCH2N(CH3)2; and
      • the indolyl is optionally substituted with methyl, CN, C(O)H, CH2CH2CH2NH2, CH2NHCH2CH═CH2, C(O)CH3, C(O)OCH3, or OCH3; methyl, CN, C(O)H, CH2CH2CH2NH2, CH2NHCH2CH═CH2, C(O)CH3, C(O)OCH3, or OCH3
      • the phenyl is optionally substituted with one or more substituents selected from the group consisting of OH and OCH3; or
    • R7 is Y—Z wherein
      • Y is benzo[b]thienyl or thienyl; and
      • Z is selected from the group consisting of CH═CHNHCH3, NHCH3, CH2NH2, CH2CH2NH2, CH2NHCH3, CH2NHCH2CH2N(CH3)2, N(CH3)2, CH2NHCH2CH2-morpholinyl, benzo[b]thienyl, morpholinylmethyl and piperazinylmethyl;
        • wherein the piperazinyl is optionally substituted with methyl.
  • In a fifth embodiment, the invention provides compounds or pharmaceutically acceptable salts thereof according to any of the foregoing inventions wherein
    • R1 and R4 are H;
    • R3 is selected from the group consisting of H, OH, 2,3-dihydrobenzofuranyl, pyrrolyl and optionally substituted napthyl; or
    • R3 is —C(O)—NY100—(C(Y100)2)x—Ra wherein
      • Y100 is H;
      • x is 0; and
      • Ra is phenyl substituted with OH;
    • R5 is
  • Figure US20120053345A1-20120301-C00020
    •  wherein
      • E is selected from the group consisting of H, CH2C(O)NH2, CH2CH2NH2, CH2CH2CH2NH2, CH2CH2CH2CH2NH2, CH2CH2CH2OCH3, CH2CH2CH2OH, CH2CH2C(O)OH, CH2CH2CH2C(O)OH, CH2CH2C(O)NH2, CH2CH2CH2C(O)NH2, CH2CH(CH3)C(O)OCH3, CH2CH(CH3)C(O)OH, CH2CH2C(O)OCH3, CH2CH2C(O)NH(CH3), CH2CH2C(O)N(CH3)2, N(CH3)2, isopropyl, morpholinylethyl and piperidinylethyl; and
      • G is H, NH2 or NHCH2CH2-pyridinyl or
    • R5 is —C(O)—NH—(CH2)a—Rc wherein
      • a is 0; and
      • Rc is J100-J200 wherein
      • J100 is phenyl and J200 is 1,8a-dihydroimidazo[1,2a]pyridinyl; or
    • R5 is —NH—C(O)—(CH2)n—Rd wherein
      • n is 2 and Rd is imidazolyl; or
    • R5 is Z100—Z200 wherein
      • Z100 is thienyl and Z200 is thienyl;
    • R6 is H or
  • Figure US20120053345A1-20120301-C00021
  • and
    • R7 is selected from the optionally substituted group consisting of H, —CH═CH-phenyl, —C≡C-phenyl, benzofuranyl, benzo[b]thienyl, indolyl, quinolinyl, naphthyl, —C(O)—NH—CH2-phenyl and —C(O)—NH-phenyl;
      • wherein the naphthyl is optionally substituted with OH, C(O)H or OCH3;
      • the benzo[b]thienyl optionally substituted with OH, CH3, OCH3, CH2═CH3—NHCH3, CH2NH2, CH2CH2NH2, CH2NHCH2CH2N(CH3)2, N(CH3)2 or piperidinylmethyl;
      • the indolyl is optionally substituted with methyl, CN, C(O)H, CH2CH2CH2NH2, CH2NHCH2CH═CH2, C(O)CH3, C(O)OCH3, or OCH3;
      • the phenyl is optionally substituted with OH or OCH3; or
    • R7 is Y—Z wherein
      • Y is benzo[b]thienyl; and
      • Z is selected from the group consisting of CH2NHCH2CH2-morpholinyl, morpholinylmethyl and piperazinylmethyl wherein the piperazinyl is optionally substituted with methyl.
  • In a sixth embodiment, the invention provides compounds or pharmaceutically acceptable salts thereof according to any of the foregoing inventions wherein R1, R3, R4 and R6 are H;
    • R5 is
  • Figure US20120053345A1-20120301-C00022
    •  wherein
      • E is selected from the group consisting of H, —CH2CH2NH2, CH2CH2CH2NH2, CH2CH2CH2OH, CH2CH2C(O)OH and CH2CH2C(O)NH2; and
    • R7 is selected from the group consisting of benzo[b]thienyl, indolyl, —C(O)—NH—CH2-phenyl and —C(O)—NH-phenyl wherein
      • the benzo[b]theinyl is optionally substituted by piperidinylmethyl;
      • the indolyl is optionally substituted by CN, methyl or C(O)H.
  • In a seventh embodiment, the invention provides compounds or pharmaceutically acceptable salts thereof according to any of the foregoing inventions wherein R1, R3, R4 and R6 are H;
    • R5 is
  • Figure US20120053345A1-20120301-C00023
    •  wherein
      • E is H; and
    • R7 is —C(O)—NH—CH2-phenyl or —C(O)—NH-phenyl.
  • In an eighth embodiment, the invention provides compounds or pharmaceutically acceptable salts thereof according to any of the foregoing inventions wherein R1, R3, R4 and R6 are H;
    • R5 is
  • Figure US20120053345A1-20120301-C00024
    •  wherein
      • E is selected from the group consisting of H, —CH2CH2NH2, CH2CH2CH2NH2, CH2CH2CH2OH, CH2CH2C(O)OH and CH2CH2C(O)NH2; and
    • R7 is benzo[b]thienyl or indolyl wherein
      • the benzo[b]theinyl is optionally substituted by piperidinylmethyl;
      • the indolyl is optionally substituted by CN, methyl or C(O)H;
  • In a ninth embodiment, the invention provides compounds or pharmaceutically acceptable salts thereof according to any of the foregoing inventions wherein R1, R3, R4 and R6 are H;
    • R5 is
  • Figure US20120053345A1-20120301-C00025
    •  wherein
      • E is selected from the group consisting of —CH2CH2NH2, CH2CH2CH2NH2, CH2CH2CH2OH, CH2CH2C(O)OH and CH2CH2C(O)NH2; and
    • R7 is benzo[b]thienyl.
    DETAILED DESCRIPTION OF THE INVENTION Protein Kinases
  • Protein kinases are a broad and diverse class, of over 500 enzymes, that include growth factors receptors, signal transduction intermediates, apoptosis related kinases and cyclin dependent kinases. Many can function as oncogenes. They are responsible for the transfer of a phosphate group to specific tyrosine, serine or threonine amino acid residues, and are broadly classified as tyrosine and S/T kinases as a result of their substrate specificity.
  • Serine/Threonine Kinases
  • S/T kinases are a large sub-family of protein kinases that specifically transfer a phosphate group to a terminal hydroxyl moiety of specific serine or threonine residues (Hanks et al., (1988) Science, 241: 42-52). A number of S/T kinase family members are involved in inflammatory signaling, tumor growth or cellular transformation. For example, the mitogen-activated protein kinases (MAPKs) are S/T kinases that act as intermediates within the signaling cascades of Toll like receptors (TLRs), such as TLR4, growth/survival factors, such as EGF, and death receptors, such as the TNF receptor. Activation of MAPKs, such as extracellular signal-regulated kinases (ERK1-2), p38a, c-Jun N-terminal kinase (INK) or MAPKAP-K2 (MK2) have been shown to transduce signaling in cells, such as macrophages, resulting in the production and secretion of pro-inflammatory cytokines, such as TNF.
  • TPL-2 is a S/T kinase which is homologous to a subfamily of kinases called MAP kinase kinase kinases (MAP3K) in its catalytic domain (Salmeron, et al., (1996) EMBO J., 15, 817-826) and is >90% identical to the proto-oncogene product of human COT (Aoki et al., (1993) J. Biol. Chem., 268, 22723-22732). TPL-2 was originally identified, in a C-terminally deleted form, as the product of an oncogene associated with Moloney murine leukemia virus-induced T cell lymphomas in rats (Patriotis, et al., (1993) Proc. Natl. Acad. Sci. USA 90, 2251-2255). TPL-2 is also highly homologous to the kinase NIK, which has been shown to regulate the inducible degradation of IκB-α (Malinin et al., (1997) Nature, 385, 540-544; WO 97/37016; May and Ghosh, (1998) Immunol. Today, 19, 80-88). TPL-2 is essential for the activation of a MAP2K (MEK1-2), which in turn activate a MAPK (extracellular signal-regulated kinase, ERK1-2) in macrophages stimulated by TLR agonists, such as lipopolysachharide (LPS). TPL-2 plays a crucial role in the regulation of LPS-induced TNF, IL-1β and COX-2 induced prostaglandin-E2 production in macrophages (Tsichlis et al, (2000), Cell, 103, 1071; Tsichlis et al, (2002), EMBO J, 21, 4831-4840). The expression of COT/TPL-2 in various tumors (Tsanisi et al., (2000), Int J Mol Med, 5, 583) and the defect in TNF production observed in COT knockout mice (Tsichlis et al, (2000), Cell, 103, 1071) suggests that inhibition of COT may be a useful approach in the treatment of cancer, inflammation or other diseases mediated by pro-inflammatory cytokines.
  • MK2 (MAPKAP-K2) is an S/T kinase critically involved in inflammatory processes. MK2 is a substrate for the MAPK p38 (Stokoe et al., (1992), EMBO J., 11, 3985-3994; Ben-Levy et al., (1995), EMBO J., 14, 5920-5930). Activation of MK2 in immune cells results in an array of cellular responses including cytokine production, proliferation and activation. Knockout mice defective in MK2 production are healthy and fertile but fail to produce cytokines such as tumor necrosis factor (TNF) in response to inflammatory stimuli (Kotlyarov et al., (1999), Nat. Cell Biol, 1, 94-97.). MK2 may alter gene expression by phosphorylation of mRNA-binding proteins (Winzen et al., (1999), EMBO J., 18, 4969-4980; Lasa et al., (2000), Mol. Cell. Biol., 20, 4265-4274; Rousseau et al., (2002), EMBO J., 21, 6505-6514; Bollig et al., (2003), Biochem. Biophys. Res. Commun, 301, 665-670; Tran et al., (2003), Mol. Cell. Biol., 23, 7177-7188.), Chrestensen, C. A. et al. J. Biol. Chem. 2004, 279, 10176-10184 and Stoecklin, G. et al. EMBO J. 2004, 23, 1313-1324) transcription factors (Heidenreich et al., (1999), J. Biol. Chem., 274, 14434-14443) or other proteins (Stokoe et al., (1992), FEBS Lett., 313, 307-313; Sutherland et al., (1993), Eur. J. Biochem., 217, 715-722; Werz et al, (2000), Proc. Natl. Acad. Sci. USA, 97, 5261-5266). The defect in TNF production in MK2 knockouts suggests that the antiinflammatory effect of p38 MAPK inhibitors may be largely due to blockade of activation of MK2 Inhibitors of MK2 may be effective treatments of inflammation or other diseases mediated by pro-inflammatory cytokines.
  • Protein Tyrosine Kinases.
  • Protein tyrosine kinases (PTKs) are enzymes that catalyse the phosphorylation of specific tyrosine residues in cellular proteins. This post-translational modification of these substrate proteins, often enzymes themselves, acts as a molecular switch regulating cell proliferation, activation or differentiation (for review, see Schlessinger and Ulrich, 1992, Neuron 9:383-391). Aberrant or excessive PTK activity has been observed in many disease states including benign and malignant proliferative disorders as well as diseases resulting from inappropriate activation of the immune system (e.g. autoimmune disorders), allograft rejection, and graft vs. host disease. In addition, endothelial-cell specific receptor PTKs such as KDR and Tie-2 mediate the angiogenic process, and are thus involved in supporting the progression of cancers and other diseases involving inappropriate vascularization (e.g., diabetic retinopathy, choroidal neovascularization due to age-related macular degeneration, psoriasis, arthritis, retinopathy of prematurity, and infantile hemangiomas).
  • Tyrosine kinases can be of the receptor-type (having extracellular, transmembrane and intracellular domains) or the non-receptor type (being wholly intracellular).
  • Receptor Tyrosine Kinases (RTKs). The RTKs comprise a large family of transmembrane receptors with diverse biological activities. At present, at least nineteen (19) distinct RTK subfamilies have been identified. The receptor tyrosine kinase (RTK) family includes receptors that are crucial for the growth and differentiation of a variety of cell types (Yarden and Ullrich, Ann. Rev. Biochem. 57:433-478, 1988; Ullrich and Schlessinger, Cell 61:243-254, 1990). The intrinsic function of RTKs is activated upon ligand binding, which results in phosphorylation of the receptor and multiple cellular substrates, and subsequently in a variety of cellular responses (Ullrich & Schlessinger, 1990, Cell 61:203-212). Thus, receptor tyrosine kinase mediated signal transduction is 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 trans-phosphorylation. 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 (e.g., cell division, differentiation, metabolic effects, and changes in the extracellular microenvironment; see Schlessinger and Ullrich, 1992, Neuron 9:1-20).
  • Non-Receptor Tyrosine Kinases. Non-receptor tyrosine kinases represent a collection of cellular enzymes that lack extracellular and transmembrane sequences. Over twenty-four individual non-receptor tyrosine kinases, comprising eleven (11) subfamilies (Src, Frk, Btk, Csk, Abl, Zap70, Fes/Fps, Fak, Jak, Ack and LIMK) have been identified. The Src subfamily of non-receptor tyrosine kinases is comprised of the largest number of PTKs and include Src, Yes, Fyn, Lyn, Lck, Blk, Hck, Fgr and Yrk. The Src subfamily of enzymes has been linked to oncogenesis and immune responses. A more detailed discussion of non-receptor tyrosine kinases is provided in Bohlen, 1993, Oncogene 8:2025-2031, which is incorporated herein by reference.
  • Many of the kinases, whether a receptor or non-receptor tyrosine kinase or a S/T kinase have been found to be involved in cellular signaling pathways involved in numerous pathogenic conditions, including immunomodulation, inflammation, or proliferative disorders such as cancer.
  • In a related aspect the invention provides a method for inhibiting COT in a human subject suffering from a disorder in which COT activity is detrimental, comprising administering to the human subject a compound of Formula (I) such that COT activity in the human subject is inhibited and treatment is achieved.
  • In another related aspect the invention provides a method for inhibiting MK2 in a human subject suffering from a disorder in which MK2 activity is detrimental, comprising administering to the human subject a compound of Formula (I) such that MK2 activity in the human subject is inhibited and treatment is achieved.
  • A compound of formula (I) or a salt thereof or pharmaceutical compositions containing a therapeutically effective amount thereof is useful in the treatment of a disorder selected from the group comprising rheumatoid arthritis, osteoarthritis, juvenile chronic arthritis, Lyme arthritis, psoriatic arthritis, reactive arthritis, and septic arthritis, spondyloarthropathy, systemic lupus erythematosus, Crohn's disease, ulcerative colitis, inflammatory bowel disease, insulin dependent diabetes mellitus, thyroiditis, asthma, allergic diseases, psoriasis, dermatitis scleroderma, graft versus host disease, organ transplant rejection (including but not limited to bone marrow and solid organ rejection), acute or chronic immune disease associated with organ transplantation, sarcoidosis, atherosclerosis, disseminated intravascular coagulation, Kawasaki's disease, Grave's disease, nephrotic syndrome, chronic fatigue syndrome, Wegener's granulomatosis, Henoch-Schoenlein purpurea, microscopic vasculitis of the kidneys, chronic active hepatitis, uveitis, septic shock, toxic shock syndrome, sepsis syndrome, cachexia, infectious diseases, parasitic diseases, acquired immunodeficiency syndrome, acute transverse myelitis, Huntington's chorea, Parkinson's disease, Alzheimer's disease, stroke, primary biliary cirrhosis, hemolytic anemia, malignancies, heart failure, myocardial infarction, Addison's disease, sporadic, polyglandular deficiency type I and polyglandular deficiency type II, Schmidt's syndrome, adult (acute) respiratory distress syndrome, alopecia, alopecia greata, seronegative arthopathy, arthropathy, Reiter's disease, psoriatic arthropathy, ulcerative colitic arthropathy, enteropathic synovitis, chlamydia, yersinia and salmonella associated arthropathy, atheromatous disease/arteriosclerosis, atopic allergy, autoimmune bullous disease, pemphigus vulgaris, pemphigus foliaceus, pemphigoid, linear IgA disease, autoimmune haemolytic anaemia, Coombs positive haemolytic anaemia, acquired pernicious anaemia, juvenile pernicious anaemia, myalgic encephalitis/Royal Free Disease, chronic mucocutaneous candidiasis, giant cell arteritis, primary sclerosing hepatitis, cryptogenic autoimmune hepatitis, Acquired Immunodeficiency Disease Syndrome, Acquired Immunodeficiency Related Diseases, Hepatitis B, Hepatitis C, common varied immunodeficiency (common variable hypogammaglobulinaemia), dilated cardiomyopathy, female infertility, ovarian failure, premature ovarian failure, fibrotic lung disease, chronic wound healing, cryptogenic fibrosing alveolitis, post-inflammatory interstitial lung disease, interstitial pneumonitis, connective tissue disease associated interstitial lung disease, mixed connective tissue disease associated lung disease, systemic sclerosis associated interstitial lung disease, rheumatoid arthritis associated interstitial lung disease, systemic lupus erythematosus associated lung disease, dermatomyositis/polymyositis associated lung disease, Sjögren's disease associated lung disease, ankylosing spondylitis associated lung disease, vasculitic diffuse lung disease, haemosiderosis associated lung disease, drug-induced interstitial lung disease, radiation fibrosis, bronchiolitis obliterans, chronic eosinophilic pneumonia, lymphocytic infiltrative lung disease, postinfectious interstitial lung disease, gouty arthritis, autoimmune hepatitis, type-1 autoimmune hepatitis (classical autoimmune or lupoid hepatitis), type-2 autoimmune hepatitis (anti-LKM antibody hepatitis), autoimmune mediated hypoglycaemia, type B insulin resistance with acanthosis nigricans, hypoparathyroidism, acute immune disease associated with organ transplantation, chronic immune disease associated with organ transplantation, osteoarthrosis, primary sclerosing cholangitis, psoriasis type 1, psoriasis type 2, idiopathic leucopaenia, autoimmune neutropaenia, renal disease NOS, glomerulonephritides, microscopic vasulitis of the kidneys, Lyme disease, discoid lupus erythematosus, male infertility idiopathic or NOS, sperm autoimmunity, multiple sclerosis (all subtypes), sympathetic ophthalmia, pulmonary hypertension secondary to connective tissue disease, Goodpasture's syndrome, pulmonary manifestation of polyarteritis nodosa, acute rheumatic fever, rheumatoid spondylitis, Still's disease, systemic sclerosis, Sjögren's syndrome, Takayasu's disease/arteritis, autoimmune thrombocytopaenia, idiopathic thrombocytopaenia, autoimmune thyroid disease, hyperthyroidism, goitrous autoimmune hypothyroidism (Hashimoto's disease), atrophic autoimmune hypothyroidism, primary myxoedema, phacogenic uveitis, primary vasculitis, vitiligo, acute liver disease, chronic liver diseases, alcoholic cirrhosis, alcohol-induced liver injury, choleosatatis, idiosyncratic liver disease, Drug-Induced hepatitis, Non-alcoholic Steatohepatitis, allergy and asthma, group B streptococci (GBS) infection, mental disorders (e.g., depression and schizophrenia), Th2 Type and Th1 Type mediated diseases, and cancers such as lung, breast, stomach, bladder, colon, pancreas, ovarian, prostate and rectal cancer and hematopoietic malignancies (leukemia and lymphoma), and hematopoietic malignancies (leukemia and lymphoma), and diseases involving inappropriate vascularization for example diabetic retinopathy, retinopathy of prematurity, choroidal neovascularization due to age-related macular degeneration, and infantile hemangiomas in human beings. In addition, such compounds may be useful in the treatment of disorders such as edema, ascites, effusions, and exudates, including for example macular edema, cerebral edema, acute lung injury, adult respiratory distress syndrome (ARDS), proliferative disorders such as restenosis, fibrotic disorders such as hepatic cirrhosis and atherosclerosis, mesangial cell proliferative disorders such as glomerulonephritis, diabetic nephropathy, malignant nephrosclerosis, thrombotic microangiopathy syndromes, and glomerulopathies, myocardial angiogenesis, coronary and cerebral collaterals, ischemic limb angiogenesis, ischemia/reperfusion injury, peptic ulcer Helicobacter related diseases, virally-induced angiogenic disorders, Crow-Fukase syndrome (POEMS), preeclampsia, menometrorrhagia, cat scratch fever, rubeosis, neovascular glaucoma and retinopathies such as those associated with diabetic retinopathy, retinopathy of prematurity, or age-related macular degeneration. In addition, these compounds can be used as active agents against solid tumors, malignant ascites, von Hippel Lindau disease, hematopoietic cancers and hyperproliferative disorders such as thyroid hyperplasia (especially Grave's disease), and cysts (such as hypervascularity of ovarian stroma characteristic of polycystic ovarian syndrome (Stein-Leventhal syndrome) and polycystic kidney disease since such diseases require a proliferation of blood vessel cells for growth and/or metastasis.
  • Compounds of formula (I) of the invention can be used alone or in combination with another therapeutic agent to treat such diseases. It should be understood that the compounds of the invention can be used alone or in combination with an additional agent, e.g., a therapeutic agent, said additional agent being selected by the skilled artisan for its intended purpose. For example, the additional agent can be a therapeutic agent art-recognized as being useful to treat the disease or condition being treated by the compound of the present invention. The additional agent also can be an agent that imparts a beneficial attribute to the therapeutic composition e.g., an agent which effects the viscosity of the composition.
  • It should further be understood that the combinations that are to be included within this invention are those combinations useful for their intended purpose. The agents set forth below are illustrative for purposes and not intended to be limited. The combinations, which are part of this invention, can be the compounds of the present invention and at least one additional agent selected from the lists below. The combination can also include more than one additional agent, e.g., two or three additional agents if the combination is such that the formed composition can perform its intended function.
  • Preferred combinations are non-steroidal anti-inflammatory drug(s) also referred to as NSAIDS which include drugs like ibuprofen. Other preferred combinations are corticosteroids including prednisolone; the well known side-effects of steroid use can be reduced or even eliminated by tapering the steroid dose required when treating patients in combination with the anti-IL-18 antibodies of this invention. Non-limiting examples of therapeutic agents for rheumatoid arthritis with which a compound of formula (I) of the invention can be combined include the following: cytokine suppressive anti-inflammatory drug(s) (CSAIDs); antibodies to or antagonists of other human cytokines or growth factors, for example, TNF, LT, IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-12, IL-15, IL-16, IL-21, IL-23, interferons, EMAP-II, GM-CSF, FGF, and PDGF. Antibodies of the invention, or antigen binding portions thereof, can be combined with antibodies to cell surface molecules such as CD2, CD3, CD4, CD8, CD25, CD28, CD30, CD40, CD45, CD69, CD80 (B7.1), CD86 (B7.2), CD90, CTLA or their ligands including CD154 (gp39 or CD40L).
  • Preferred combinations of therapeutic agents may interfere at different points in the autoimmune and subsequent inflammatory cascade; preferred examples include TNF antagonists like chimeric, humanized or human TNF antibodies, D2E7 (HUMIRA™), (PCT Publication No. WO 97/29131), CA2 (REMICADE™), CDP 571, and soluble p55 or p75 TNF receptors, derivatives, thereof, (p75TNFR1gG (ENBREL™) or p55TNFR1gG (Lenercept), and also TNFα converting enzyme (TACE) inhibitors; similarly IL-1 inhibitors (Interleukin-1-converting enzyme inhibitors, IL-1RA etc.) may be effective for the same reason. Other preferred combinations include Interleukin 11. Yet another preferred combination are other key players of the autoimmune response which may act parallel to, dependent on or in concert with IL-18 function; especially preferred are IL-12 antagonists including IL-12 antibodies or soluble IL-12 receptors, or IL-12 binding proteins. It has been shown that IL-12 and IL-18 have overlapping but distinct functions and a combination of antagonists to both may be most effective. Yet another preferred combination are non-depleting anti-CD4 inhibitors. Yet other preferred combinations include antagonists of the co-stimulatory pathway CD80 (B7.1) or CD86 (B7.2) including antibodies, soluble receptors or antagonistic ligands.
  • A compound of formula (I) of the invention may also be combined with agents, such as methotrexate, 6-MP, azathioprine sulphasalazine, mesalazine, olsalazine chloroquinine/hydroxychloroquine, pencillamine, aurothiomalate (intramuscular and oral), azathioprine, cochicine, corticosteroids (oral, inhaled and local injection), beta-2 adrenoreceptor agonists (salbutamol, terbutaline, salmeteral), xanthines (theophylline, aminophylline), cromoglycate, nedocromil, ketotifen, ipratropium and oxitropium, cyclosporin, FK506, rapamycin, mycophenolate mofetil, leflunomide, NSAIDs, for example, ibuprofen, corticosteroids such as prednisolone, phosphodiesterase inhibitors, adensosine agonists, antithrombotic agents, complement inhibitors, adrenergic agents, agents which interfere with signalling by proinflammatory cytokines such as TNFα or IL-1 (e.g. IRAK family, NIK, IKK family, p38 or other MAP kinase inhibitors), IL-1β converting enzyme inhibitors, TNFα converting enzyme (TACE) inhibitors, T-cell signalling inhibitors such as kinase inhibitors, metalloproteinase inhibitors, sulfasalazine, azathioprine, 6-mercaptopurines, angiotensin converting enzyme inhibitors, soluble cytokine receptors and derivatives thereof (e.g. soluble p55 or p75 TNF receptors and the derivatives p75TNFRIgG (Enbrel™ and p55TNFRIgG (Lenercept)), sIL-1RI, sIL-1RII, sIL-6R), antiinflammatory cytokines (e.g. IL-4, IL-10, IL-11, IL-13 and TGFβ), celecoxib, folic acid, hydroxychloroquine sulfate, rofecoxib, etanercept, infliximab, naproxen, valdecoxib, sulfasalazine, methylprednisolone, meloxicam, methylprednisolone acetate, gold sodium thiomalate, aspirin, triamcinolone acetonide, propoxyphene napsylate/apap, folate, nabumetone, diclofenac, piroxicam, etodolac, diclofenac sodium, oxaprozin, oxycodone HCl, hydrocodone bitartrate/apap, diclofenac sodium/misoprostol, fentanyl, anakinra, human recombinant, tramadol HCl, salsalate, sulindac, cyanocobalamin/fa/pyridoxine, acetaminophen, alendronate sodium, prednisolone, morphine sulfate, lidocaine hydrochloride, indomethacin, glucosamine sulf/chondroitin, amitriptyline HCl, sulfadiazine, oxycodone HCl/acetaminophen, olopatadine HCl, misoprostol, naproxen sodium, omeprazole, cyclophosphamide, rituximab, IL-1 TRAP, MRA, CTLA4-IG, IL-18 BP, anti-IL-12, Anti-IL15, BIRB-796, SCIO-469, VX-702, AMG-548, VX-740, Roflumilast, IC-485, CDC-801, and Mesopram. Preferred combinations include methotrexate or leflunomide and in moderate or severe rheumatoid arthritis cases, cyclosporine and anti-TNF antibodies as noted above.
  • Non-limiting examples of therapeutic agents for inflammatory bowel disease with which a compound of formula (I) of the invention can be combined include the following: budenoside; epidermal growth factor; corticosteroids; cyclosporin, sulfasalazine; aminosalicylates; 6-mercaptopurine; azathioprine; metronidazole; lipoxygenase inhibitors; mesalamine; olsalazine; balsalazide; antioxidants; thromboxane inhibitors; IL-1 receptor antagonists; anti-IL-1β monoclonal antibodies; anti-IL-6 monoclonal antibodies; growth factors; elastase inhibitors; pyridinyl-imidazole compounds; antibodies to or antagonists of other human cytokines or growth factors, for example, TNF, LT, IL-1, IL-2, IL-6, IL-7, IL-8, IL-12, IL-15, IL-16, EMAP-II, GM-CSF, FGF, and PDGF; cell surface molecules such as CD2, CD3, CD4, CD8, CD25, CD28, CD30, CD40, CD45, CD69, CD90 or their ligands; methotrexate; cyclosporine; FK506; rapamycin; mycophenolate mofetil; leflunomide; NSAIDs, for example, ibuprofen; corticosteroids such as prednisolone; phosphodiesterase inhibitors; adenosine agonists; antithrombotic agents; complement inhibitors; adrenergic agents; agents which interfere with signalling by proinflammatory cytokines such as TNFα or IL-1 (e.g. IRAK, NIK, IKK, p38 or MAP kinase inhibitors); IL-1β converting enzyme inhibitors; TNFα converting enzyme inhibitors; T-cell signalling inhibitors such as kinase inhibitors; metalloproteinase inhibitors; sulfasalazine; azathioprine; 6-mercaptopurines; angiotensin converting enzyme inhibitors; soluble cytokine receptors and derivatives thereof (e.g. soluble p55 or p75 TNF receptors, sIL-1RI, sIL-1RII, sIL-6R) and antiinflammatory cytokines (e.g. IL-4, IL-10, IL-11, IL-13 and TGFβ). Preferred examples of therapeutic agents for Crohn's disease in which a compound of formula (I) can be combined include the following: TNF antagonists, for example, anti-TNF antibodies, D2E7 (PCT Publication No. WO 97/29131; HUMIRA™), CA2 (REMICADE™), CDP 571, TNFR-Ig constructs, (p75TNFRIgG (ENBREL™) and p55TNFRIgG (LENERCEPT™)) inhibitors and PDE4 inhibitors. A compound of formula (I) can be combined with corticosteroids, for example, budenoside and dexamethasone; sulfasalazine, 5-aminosalicylic acid; olsalazine; and agents which interfere with synthesis or action of proinflammatory cytokines such as IL-1, for example, IL-1β converting enzyme inhibitors and IL-1ra; T cell signaling inhibitors, for example, tyrosine kinase inhibitors 6-mercaptopurines; IL-11; mesalamine; prednisone; azathioprine; mercaptopurine; infliximab; methylprednisolone sodium succinate; diphenoxylate/atrop sulfate; loperamide hydrochloride; methotrexate; omeprazole; folate; ciprofloxacin/dextrose-water; hydrocodone bitartrate/apap; tetracycline hydrochloride; fluocinonide; metronidazole; thimerosal/boric acid; cholestyramine/sucrose; ciprofloxacin hydrochloride; hyoscyamine sulfate; meperidine hydrochloride; midazolam hydrochloride; oxycodone HCl/acetaminophen; promethazine hydrochloride; sodium phosphate; sulfamethoxazole/trimethoprim; celecoxib; polycarbophil; propoxyphene napsylate; hydrocortisone; multivitamins; balsalazide disodium; codeine phosphate/apap; colesevelam HCl; cyanocobalamin; folic acid; levofloxacin; methylprednisolone; natalizumab and interferon-gamma.
  • Non-limiting examples of therapeutic agents for multiple sclerosis with which a compound of formula (I) can be combined include the following: corticosteroids; prednisolone; methylprednisolone; azathioprine; cyclophosphamide; cyclosporine; methotrexate; 4-aminopyridine; tizanidine; interferon-β1a (AVONEX; Biogen); interferon-β1b (BETASERON; Chiron/Berlex); interferon α-n3) (Interferon Sciences/Fujimoto), interferon-α (Alfa Wassermann/J&J), interferon β1A-IF (Serono/Inhale Therapeutics), Peginterferon α 2b (Enzon/Schering-Plough), Copolymer 1 (Cop-1; COPAXONE; Teva Pharmaceutical Industries, Inc.); hyperbaric oxygen; intravenous immunoglobulin; clabribine; antibodies to or antagonists of other human cytokines or growth factors and their receptors, for example, TNF, LT, IL-1, IL-2, IL-6, IL-7, IL-8, IL-12, IL-23, IL-15, IL-16, EMAP-II, GM-CSF, FGF, and PDGF. A compound of formula (I) can be combined with antibodies to cell surface molecules such as CD2, CD3, CD4, CD8, CD19, CD20, CD25, CD28, CD30, CD40, CD45, CD69, CD80, CD86, CD90 or their ligands. A compound of formula (I) may also be combined with agents, such as methotrexate, cyclosporine, FK506, rapamycin, mycophenolate mofetil, leflunomide, NSAIDs, for example, ibuprofen, corticosteroids such as prednisolone, phosphodiesterase inhibitors, adensosine agonists, antithrombotic agents, complement inhibitors, adrenergic agents, agents which interfere with signalling by proinflammatory cytokines such as TNFα or IL-1 (e.g. IRAK, NIK, IKK, p38 or MAP kinase inhibitors), IL-1β converting enzyme inhibitors, TACE inhibitors, T-cell signaling inhibitors such as kinase inhibitors, metalloproteinase inhibitors, sulfasalazine, azathioprine, 6-mercaptopurines, angiotensin converting enzyme inhibitors, soluble cytokine receptors and derivatives thereof (e.g. soluble p55 or p75 TNF receptors, sIL-1RI, sIL-1RII, sIL-6R) and antiinflammatory cytokines (e.g. IL-4, IL-10, IL-13 and TGFβ).
  • Preferred examples of therapeutic agents for multiple sclerosis in which a compound of formula (I) can be combined to include interferon-β, for example, IFNβ1a and IFNβ1b; copaxone, corticosteroids, caspase inhibitors, for example inhibitors of caspase-1, IL-1 inhibitors, TNF inhibitors, and antibodies to CD40 ligand and CD80.
  • A compound of formula (I) may also be combined with agents, such as alemtuzumab, dronabinol, Unimed, daclizumab, mitoxantrone, xaliproden hydrochloride, fampridine, glatiramer acetate, natalizumab, sinnabidol, a-immunokine NNSO3, ABR-215062, AnergiX.MS, chemokine receptor antagonists, BBR-2778, calagualine, CPI-1189, LEM (liposome encapsulated mitoxantrone), THC.CBD (cannabinoid agonist) MBP-8298, mesopram (PDE4 inhibitor), MNA-715, anti-IL-6 receptor antibody, neurovax, pirfenidone allotrap 1258 (RDP-1258), sTNF-R1, talampanel, teriflunomide, TGF-beta2, tiplimotide, VLA-4 antagonists (for example, TR-14035, VLA4 Ultrahaler, Antegran-ELAN/Biogen), interferon gamma antagonists, IL-4 agonists.
  • Non-limiting examples of therapeutic agents for Angina with which a compound of formula (I) of the invention can be combined include the following: aspirin, nitroglycerin, isosorbide mononitrate, metoprolol succinate, atenolol, metoprolol tartrate, amlodipine besylate, diltiazem hydrochloride, isosorbide dinitrate, clopidogrel bisulfate, nifedipine, atorvastatin calcium, potassium chloride, furosemide, simvastatin, verapamil HCl, digoxin, propranolol hydrochloride, carvedilol, lisinopril, spironolactone, hydrochlorothiazide, enalapril maleate, nadolol, ramipril, enoxaparin sodium, heparin sodium, valsartan, sotalol hydrochloride, fenofibrate, ezetimibe, bumetanide, losartan potassium, lisinopril/hydrochlorothiazide, felodipine, captopril, bisoprolol fumarate.
  • Non-limiting examples of therapeutic agents for Ankylosing Spondylitis with which a compound of formula (I) can be combined include the following: ibuprofen, diclofenac and misoprostol, naproxen, meloxicam, indomethacin, diclofenac, celecoxib, rofecoxib, Sulfasalazine, Methotrexate, azathioprine, minocyclin, prednisone, etanercept, infliximab.
  • Non-limiting examples of therapeutic agents for Asthma with which a compound of formula (I) can be combined include the following: albuterol, salmeterol/fluticasone, montelukast sodium, fluticasone propionate, budesonide, prednisone, salmeterol xinafoate, levalbuterol HCl, albuterol sulfate/ipratropium, prednisolone sodium phosphate, triamcinolone acetonide, beclomethasone dipropionate, ipratropium bromide, azithromycin, pirbuterol acetate, prednisolone, theophylline anhydrous, methylprednisolone sodium succinate, clarithromycin, zafirlukast, formoterol fumarate, influenza virus vaccine, methylprednisolone, amoxicillin trihydrate, flunisolide, allergy injection, cromolyn sodium, fexofenadine hydrochloride, flunisolide/menthol, amoxicillin/clavulanate, levofloxacin, inhaler assist device, guaifenesin, dexamethasone sodium phosphate, moxifloxacin HCl, doxycycline hyclate, guaifenesin/d-methorphan, p-ephedrine/cod/chlorphenir, gatifloxacin, cetirizine hydrochloride, mometasone furoate, salmeterol xinafoate, benzonatate, cephalexin, pe/hydrocodone/chlorphenir, cetirizine HCl/pseudoephed, phenylephrine/cod/promethazine, codeine/promethazine, cefprozil, dexamethasone, guaifenesin/pseudoephedrine, chlorpheniramine/hydrocodone, nedocromil sodium, terbutaline sulfate, epinephrine, methylprednisolone, metaproterenol sulfate.
  • Non-limiting examples of therapeutic agents for COPD with which a compound of formula (I) can be combined include the following: albuterol sulfate/ipratropium, ipratropium bromide, salmeterol/fluticasone, albuterol, salmeterol xinafoate, fluticasone propionate, prednisone, theophylline anhydrous, methylprednisolone sodium succinate, montelukast sodium, budesonide, formoterol fumarate, triamcinolone acetonide, levofloxacin, guaifenesin, azithromycin, beclomethasone dipropionate, levalbuterol HCl, flunisolide, ceftriaxone sodium, amoxicillin trihydrate, gatifloxacin, zafirlukast, amoxicillin/clavulanate, flunisolide/menthol, chlorpheniramine/hydrocodone, metaproterenol sulfate, methylprednisolone, mometasone furoate, p-ephedrine/cod/chlorphenir, pirbuterol acetate, p-ephedrine/loratadine, terbutaline sulfate, tiotropium bromide, (R,R)-formoterol, TgAAT, Cilomilast, Roflumilast.
  • Non-limiting examples of therapeutic agents for HCV with which a compound of formula (I) can be combined include the following: Interferon-alpha-2a, Interferon-alpha-2b, Interferon-alpha con1, Interferon-alpha-n1, Pegylated interferon-alpha-2a, Pegylated interferon-alpha-2b, ribavirin, Peginterferon alfa-2b+ribavirin, Ursodeoxycholic Acid, Glycyrrhizic Acid, Thymalfasin, Maxamine, VX-497 and any compounds that are used to treat HCV through intervention with the following targets: HCV polymerase, HCV protease, HCV helicase, HCV IRES (internal ribosome entry site).
  • Non-limiting examples of therapeutic agents for Idiopathic Pulmonary Fibrosis with which a compound of formula (I) can be combined include the following: prednisone, azathioprine, albuterol, colchicine, albuterol sulfate, digoxin, gamma interferon, methylprednisolone sod succ, lorazepam, furosemide, lisinopril, nitroglycerin, spironolactone, cyclophosphamide, ipratropium bromide, actinomycin d, alteplase, fluticasone propionate, levofloxacin, metaproterenol sulfate, morphine sulfate, oxycodone HCl, potassium chloride, triamcinolone acetonide, tacrolimus anhydrous, calcium, interferon-alpha, methotrexate, mycophenolate mofetil, Interferon-gamma-1β.
  • Non-limiting examples of therapeutic agents for Myocardial Infarction with which a compound of formula (I) can be combined include the following: aspirin, nitroglycerin, metoprolol tartrate, enoxaparin sodium, heparin sodium, clopidogrel bisulfate, carvedilol, atenolol, morphine sulfate, metoprolol succinate, warfarin sodium, lisinopril, isosorbide mononitrate, digoxin, furosemide, simvastatin, ramipril, tenecteplase, enalapril maleate, torsemide, retavase, losartan potassium, quinapril HCl/mag carb, bumetanide, alteplase, enalaprilat, amiodarone hydrochloride, tirofiban HCl m-hydrate, diltiazem hydrochloride, captopril, irbesartan, valsartan, propranolol hydrochloride, fosinopril sodium, lidocaine hydrochloride, eptifibatide, cefazolin sodium, atropine sulfate, aminocaproic acid, spironolactone, interferon, sotalol hydrochloride, potassium chloride, docusate sodium, dobutamine HCl, alprazolam, pravastatin sodium, atorvastatin calcium, midazolam hydrochloride, meperidine hydrochloride, isosorbide dinitrate, epinephrine, dopamine hydrochloride, bivalirudin, rosuvastatin, ezetimibe/simvastatin, avasimibe, cariporide.
  • Non-limiting examples of therapeutic agents for Psoriasis with which a compound of formula (I) can be combined include the following: calcipotriene, clobetasol propionate, triamcinolone acetonide, halobetasol propionate, tazarotene, methotrexate, fluocinonide, betamethasone diprop augmented, fluocinolone acetonide, acitretin, tar shampoo, betamethasone valerate, mometas one furoate, ketoconazole, pramoxine/fluocinolone, hydrocortisone valerate, flurandrenolide, urea, betamethasone, clobetasol propionate/emoll, fluticasone propionate, azithromycin, hydrocortisone, moisturizing formula, folic acid, desonide, pimecrolimus, coal tar, diflorasone diacetate, etanercept folate, lactic acid, methoxsalen, hc/bismuth subgal/znox/resor, methylprednisolone acetate, prednisone, sunscreen, halcinonide, salicylic acid, anthralin, clocortolone pivalate, coal extract, coal tar/salicylic acid, coal tar/salicylic acid/sulfur, desoximetasone, diazepam, emollient, fluocinonide/emollient, mineral oil/castor oil/na lact, mineral oil/peanut oil, petroleum/isopropyl myristate, psoralen, salicylic acid, soap/tribromsalan, thimerosal/boric acid, celecoxib, infliximab, cyclosporine, alefacept, efalizumab, tacrolimus, pimecrolimus, PUVA, UVB, sulfasalazine.
  • Non-limiting examples of therapeutic agents for Psoriatic Arthritis with which a compound of formula (I) can be combined include the following: methotrexate, etanercept, rofecoxib, celecoxib, folic acid, sulfasalazine, naproxen, leflunomide, methylprednisolone acetate, indomethacin, hydroxychloroquine sulfate, prednisone, sulindac, betamethasone diprop augmented, infliximab, methotrexate, folate, triamcinolone acetonide, diclofenac, dimethylsulfoxide, piroxicam, diclofenac sodium, ketoprofen, meloxicam, methylprednisolone, nabumetone, tolmetin sodium, calcipotriene, cyclosporine, diclofenac sodium/misoprostol, fluocinonide, glucosamine sulfate, gold sodium thiomalate, hydrocodone bitartrate/apap, ibuprofen, risedronate sodium, sulfadiazine, thioguanine, valdecoxib, alefacept, efalizumab.
  • Non-limiting examples of therapeutic agents for Restenosis with which a compound of formula (I) can be combined include the following: sirolimus, paclitaxel, everolimus, tacrolimus, ABT-578, acetaminophen.
  • Non-limiting examples of therapeutic agents for Sciatica with which a compound of formula (I) can be combined include the following: hydrocodone bitartrate/apap, rofecoxib, cyclobenzaprine HCl, methylprednisolone, naproxen, ibuprofen, oxycodone HCl/acetaminophen, celecoxib, valdecoxib, methylprednisolone acetate, prednisone, codeine phosphate/apap, tramadol HCl/acetaminophen, metaxalone, meloxicam, methocarbamol, lidocaine hydrochloride, diclofenac sodium, gabapentin, dexamethasone, carisoprodol, ketorolac tromethamine, indomethacin, acetaminophen, diazepam, nabumetone, oxycodone HCl, tizanidine HCl, diclofenac sodium/misoprostol, propoxyphene napsylate/apap, asa/oxycod/oxycodone ter, ibuprofen/hydrocodone bit, tramadol HCl, etodolac, propoxyphene HCl, amitriptyline HCl, carisoprodol/codeine phos/asa, morphine sulfate, multivitamins, naproxen sodium, orphenadrine citrate, temazepam.
  • Preferred examples of therapeutic agents for SLE (Lupus) in which a compound of formula (I) include the following: NSAIDS, for example, diclofenac, naproxen, ibuprofen, piroxicam, indomethacin; COX2 inhibitors, for example, Celecoxib, rofecoxib, valdecoxib; anti-malarials, for example, hydroxychloroquine; Steroids, for example, prednisone, prednisolone, budenoside, dexamethasone; Cytotoxics, for example, azathioprine, cyclophosphamide, mycophenolate mofetil, methotrexate; inhibitors of PDE4 or purine synthesis inhibitor, for example Cellcept. A compound of formula (I) may also be combined with agents such as sulfasalazine, 5-aminosalicylic acid, olsalazine, Imuran and agents which interfere with synthesis, production or action of proinflammatory cytokines such as IL-1, for example, caspase inhibitors like IL-1β converting enzyme inhibitors and IL-1ra. A compound of formula (I) may also be used with T cell signaling inhibitors, for example, tyrosine kinase inhibitors; or molecules that target T cell activation molecules, for example, CTLA-4-IgG or anti-B7 family antibodies, anti-PD-1 family antibodies. A compound of formula (I) can be combined with IL-11 or anti-cytokine antibodies, for example, fonotolizumab (anti-IFNg antibody), or anti-receptor receptor antibodies, for example, anti-IL-6 receptor antibody and antibodies to B-cell surface molecules. A compound of formula (I) may also be used with LJP 394 (abetimus), agents that deplete or inactivate B-cells, for example, Rituximab (anti-CD20 antibody), lymphostat-B (anti-BlyS antibody), TNF antagonists, for example, anti-TNF antibodies, D2E7 (PCT Publication No. WO 97/29131; HUMIRA™), CA2 (REMICADE™), CDP 571, TNFR-Ig constructs, (p75TNFRIgG (ENBREL™) and p55TNFRIgG (LENERCEPT™)).
  • In this invention, the following definitions are applicable:
  • A “therapeutically effective amount” is an amount of a compound of Formula I or a combination of two or more such compounds, which inhibits, totally or partially, the progression of the condition or alleviates, at least partially, one or more symptoms of the condition. A therapeutically effective amount can also be an amount which is prophylactically effective. The amount which is therapeutically effective will depend upon the patient's size and gender, the condition to be treated, the severity of the condition and the result sought. For a given patient, a therapeutically effective amount can be determined by methods known to those of skill in the art.
  • “Physiologically acceptable salts” refers to those salts which retain the biological effectiveness and properties of the free bases and which are obtained by reaction with inorganic acids, for example, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, and phosphoric acid or organic acids such as sulfonic acid, carboxylic acid, organic phosphoric acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, citric acid, fumaric acid, maleic acid, succinic acid, benzoic acid, salicylic acid, lactic acid, tartaric acid (e.g. (+) or (−)-tartaric acid or mixtures thereof), amino acids (e.g. (+) or (−)-amino acids or mixtures thereof), and the like. These salts can be prepared by methods known to those skilled in the art.
  • Certain compounds of formula I which have acidic substituents may exist as salts with pharmaceutically acceptable bases. The present invention includes such salts. Examples of such salts include sodium salts, potassium salts, lysine salts and arginine salts. These salts may be prepared by methods known to those skilled in the art.
  • Certain compounds of formula I and their salts may exist in more than one crystal form and the present invention includes each crystal form and mixtures thereof.
  • Certain compounds of formula I and their salts may also exist in the form of solvates, for example hydrates, and the present invention includes each solvate and mixtures thereof.
  • Certain compounds of formula I may contain one or more chiral centers, and exist in different optically active forms. When compounds of formula I contain one chiral center, the compounds exist in two enantiomeric forms and the present invention includes both enantiomers and mixtures of enantiomers, such as racemic mixtures. The enantiomers may be resolved by methods known to those skilled in the art, for example by formation of diastereoisomeric salts which may be separated, for example, by crystallization; formation of diastereoisomeric derivatives or complexes which may be separated, for example, by crystallization, gas-liquid or liquid chromatography; selective reaction of one enantiomer with an enantiomer-specific reagent, for example enzymatic esterification; or gas-liquid or liquid chromatography in a chiral environment, for example on a chiral support for example silica with a bound chiral ligand or in the presence of a chiral solvent. It will be appreciated that where the desired enantiomer is converted into another chemical entity by one of the separation procedures described above, a further step is required to liberate the desired enantiomeric form. Alternatively, specific enantiomers may be synthesized by asymmetric synthesis using optically active reagents, substrates, catalysts or solvents, or by converting one enantiomer into the other by asymmetric transformation.
  • When a compound of formula I contains more than one chiral center it may exist in diastereoisomeric forms. The diastereoisomeric pairs may be separated by methods known to those skilled in the art, for example chromatography or crystallization and the individual enantiomers within each pair may be separated as described above. The present invention includes each diastereoisomer of compounds of formula I and mixtures thereof.
  • Certain compounds of formula I may exist in different tautomeric forms or as different geometric isomers, and the present invention includes each tautomer and/or geometric isomer of compounds of formula I and mixtures thereof.
  • Certain compounds of formula I may exist in different stable conformational forms which may be separable. Torsional asymmetry due to restricted rotation about an asymmetric single bond, for example because of steric hindrance or ring strain, may permit separation of different conformers. The present invention includes each conformational isomer of compounds of formula I and mixtures thereof.
  • Certain compounds of formula I may exist in zwitterionic form and the present invention includes each zwitterionic form of compounds of formula I and mixtures thereof.
  • As used herein the term “pro-drug” refers to an agent which is converted into the parent drug in vivo by some physiological chemical process (e.g., a prodrug on being brought to the physiological pH is converted to the desired drug form). Pro-drugs are often useful because, in some situations, they may be easier to administer than the parent drug. They may, for instance, be bioavailable by oral administration whereas the parent drug is not. The prodrug may also have improved solubility in pharmacological compositions over the parent drug. An example, without limitation, of a pro-drug would be a compound of the present invention wherein it is administered as an ester (the “pro-drug”) to facilitate transmittal across a cell membrane where water solubility is not beneficial, but then it is metabolically hydrolyzed to the carboxylic acid once inside the cell where water solubility is beneficial
  • Pro-drugs have many useful properties. For example, a pro-drug may be more water soluble than the ultimate drug, thereby facilitating intravenous administration of the drug. A pro-drug may also have a higher level of oral bioavailability than the ultimate drug. After administration, the prodrug is enzymatically or chemically cleaved to deliver the ultimate drug in the blood or tissue.
  • Exemplary pro-drugs upon cleavage release the corresponding free acid, and such hydrolyzable ester-forming residues of the compounds of this invention include but are not limited to carboxylic acid substituents (e.g., —(CH2)C(O)H or a moiety that contains a carboxylic acid) wherein the free hydrogen is replaced by (C1-C4)alkyl, (C2-C12)alkanoyloxymethyl, (C4-C9)1-(alkanoyloxy)ethyl, 1-methyl-1-(alkanoyloxy)-ethyl having from 5 to 10 carbon atoms, alkoxycarbonyloxymethyl having from 3 to 6 carbon atoms, 1-(alkoxycarbonyloxy)ethyl having from 4 to 7 carbon atoms, 1-methyl-1-(alkoxycarbonyloxy)ethyl having from 5 to 8 carbon atoms, N-(alkoxycarbonyl)aminomethyl having from 3 to 9 carbon atoms, 1-(N-(alkoxycarbonyl)amino)ethyl having from 4 to 10 carbon atoms, 3-phthalidyl, 4-crotonolactonyl, gamma-butyrolacton-4-yl, di-N,N—(C1-C2)alkylamino(C2-C3)alkyl (such as β-dimethylaminoethyl), carbamoyl-(C1-C2)alkyl, N,N-di(C1-C2)-alkylcarbamoyl-(C1-C2)alkyl and piperidino-, pyrrolidino- or morpholino(C2-C3)alkyl.
  • Other exemplary pro-drugs release an alcohol of Formula I wherein the free hydrogen of the hydroxyl substituent (e.g., R1 contains hydroxyl) is replaced by (C1-C6)alkanoyloxymethyl, 1-((C1-C6)alkanoyloxy)ethyl, 1-methyl-1-((C1-C6)alkanoyloxy)ethyl, (C1-C6)alkoxycarbonyloxymethyl, N—(C1-C6)alkoxycarbonylamino-methyl, succinoyl, (C1-C6)alkanoyl, α-amino(C1-C4)alkanoyl, arylactyl and α-aminoacyl, or α-aminoacyl-α-aminoacyl wherein said α-aminoacyl moieties are independently any of the naturally occurring L-amino acids found in proteins, P(O)(OH)2, —P(O)(O(C1-C6)alkyl)2 or glycosyl (the radical resulting from detachment of the hydroxyl of the hemiacetal of a carbohydrate).
  • Heteroaromatic groups, as used herein, include heteroaryl ring systems (e.g., for purposes of exemplification, which should not be construed as limiting the scope of this invention: thienyl, pyridyl, pyrazole, isoxazolyl, thiadiazolyl, oxadiazolyl, indazolyl, furans, pyrroles, imidazoles, pyrazoles, triazoles, pyrimidines, pyrazines, thiazoles, isothiazoles, oxazolyl or tetrazoles) and heteroaryl ring systems in which a carbocyclic aromatic ring, carbocyclic non-aromatic ring or heteroaryl ring is fused to one or more other heteroaryl rings (e.g., for purposes of exemplification, which should not be construed as limiting the scope of this invention: benzo[b]thienyl, benzimidazolyl, benzoxazolyl, benzothiazolyl, benzothiadiazolyl, benzoxadiazolyl, indole, tetrahydroindole, azaindole, indazole, quinoline, imidazopyridine, quinazoline purine, pyrrolo[2,3-d]pyrimidine, pyrazolo[3,4-d]pyrimidine) and their N-oxides. Substituted heteroaryl groups are preferably substituted with one or more substituents each independently selected from the group consisting of halogen, hydroxy, alkyl, alkoxy, alkyl-O—C(O)—, alkoxyalkyl, a heterocycloalkyl group, optionally substituted phenyl, nitro, amino, mono-substituted amino or di-substituted amino
  • The term “heterocyclic” or “heterocyclyl”, as used herein, include aromatic and non-aromatic, ring systems, including, but not limited to, monocyclic, bicyclic and tricyclic rings, which can be completely saturated or which can contain one or more units of unsaturation and have 3 to 12 atoms including at least one heteroatom, such as nitrogen, oxygen, or sulfur. For purposes of exemplification, which should not be construed as limiting the scope of this invention: azaindole, azetidinyls furans, imidazoles, imidazopyridine, indole, isoxazoles, isothiazoles, oxadiazoles, oxazoles, piperazines, piperidines, pyrans, pyrazines, pyrazoles, pyridines, pyrimidines, pyrroles, pyrrolidines, quinolines, quinazolines, triazoles, thiazoles, tetrahydroindole, tetrazoles, thiadiazoles, thienyls, thiomorpholinos or triazles.
  • When the term “substituted heterocyclic” (or heterocyclyl) is used, what is meant is that the heterocyclic group is substituted with one or more substituents that can be made by one of ordinary skill in the art and results in a molecule that is a kinase inhibitor. For purposes of exemplification, which should not be construed as limiting the scope of this invention, preferred substituents for the heterocyclyls of this invention are each independently selected from the optionally substituted group consisting of alkenyl, alkoxy, alkoxyalkoxy, alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylheterocycloalkoxy, alkyl, alkylcarbonyl, alkylester, alkyl-O—C(O)—, alkyl-heterocyclyl, alkyl-cycloalkyl, alkyl-nitrile, alkynyl, amido groups, amino, aminoalkyl, aminocarbonyl, carbonitrile, carbonylalkoxy, carboxamido, CF3, CN, —C(O)OH, —C(O)H, —C(O)—)(CH3)3, —OH, —C(O)O-alkyl, —C(O)β-cycloalkyl, —C(O)O-heterocyclyl, —C(O)-alkyl, —C(O)-cycloalkyl, —C(O)-heterocyclyl, cycloalkyl, dialkylaminoalkoxy, dialkylaminocarbonylalkoxy, dialkylaminocarbonyl, halogen, heterocyclyl, a heterocycloalkyl group, heterocyclyloxy, hydroxy, hydroxyalkyl, nitro, NO2, OCF3, oxo, phenyl, —SO2CH3, —SO2CR3, tetrazolyl, thienylalkoxy, trifluoromethylcarbonylamino, trifluoromethylsulfonamido, heterocyclylalkoxy, heterocyclyl-S(O)p, cycloalkyl-S(O)p, alkyl-S—, heterocyclyl-S, heterocycloalkyl, cycloalkylalkyl, heterocycolthio, cycloalkylthio, —Z105—C(O)N(R)2, —Z105—N(R)—C(O)—Z200, —Z105—N(R)—S(O)2—Z200, —Z105—N(R)—C(O)—N(R)—Z200, —N(R)—C(O)R, —N(R)—C(O)OR, OR—C(O)-heterocyclyl-OR, Rc and —CH2ORc;
  • where Rc for each occurrence is independently hydrogen, optionally substituted alkyl, optionally substituted aryl, —(C1-C6)—NRdRe, -E-(CH2)t—NRdRe, -E-(CH2)t—O-alkyl, -E-(CH2)t—S-alkyl, or -E-(CH2)t—OH
      • wherein t is an integer from about 1 to about 6;
        Z105 for each occurrence is independently a covalent bond, alkyl, alkenyl or alkynyl; and
        Z200 for each occurrence is independently selected from an optionally substituted group selected from the group consisting of alkyl, alkenyl, alkynyl, phenyl, alkyl-phenyl, alkenyl-phenyl or alkynyl-phenyl;
        E is a direct bond, O, S, S(O), S(O)2, or NRf, wherein Rf is H or alkyl and Rd and Re are independently H, alkyl, alkanoyl or SO2-alkyl; or Rd, Re and the nitrogen atom to which they are attached together form a five- or six-membered heterocyclic ring.
  • An “heterocycloalkyl” group, as used herein, is a heterocyclic group that is linked to a compound by an aliphatic group having from one to about eight carbon atoms. For example, a preferred heterocycloalkyl group is an imidazolylethyl group.
  • As used herein, “aliphatic” or “an aliphatic group” or notations such as “(C0-C8)” include straight chained or branched hydrocarbons which are completely saturated or which contain one or more units of unsaturation, and, thus, includes alkyl, alkenyl, alkynyl and hydrocarbons comprising a mixture of single, double and triple bonds. When the group is a Co it means that the moiety is not present or in other words, it is a bond. As used herein, “alkyl” means C1-C8 and includes straight chained or branched hydrocarbons which are completely saturated. Preferred alkyls are methyl, ethyl, propyl, butyl, pentyl, hexyl and isomers thereof. As used herein, “alkenyl” and “alkynyl” means C2-C8 and includes straight chained or branched hydrocarbons which contain one or more units of unsaturation, one or more double bonds for alkenyl and one or more triple bonds for alkynyl.
  • As used herein, aromatic groups (or aryl groups) include aromatic carbocyclic ring systems (e.g. phenyl and cyclopentyldienyl) and fused polycyclic aromatic ring systems (e.g. naphthyl, biphenylenyl and 1,2,3,4-tetrahydronaphthyl).
  • As used herein, cycloalkyl means C3-C12 monocyclic or multicyclic (e.g., bicyclic, tricyclic, etc.) hydrocarbons which is completely saturated or has one or more unsaturated bonds but does not amount to an aromatic group. Preferred examples of a cycloalkyl group are cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl and cyclohexenyl.
  • As used herein, amido group means —NHC(═O)—.
  • As used herein, acyloxy groups are —OC(O)R.
  • As used herein, many moieties or substituents are termed as being either “substituted” or “optionally substituted”. When a moiety is modified by one of these terms, it denotes that any portion of the moiety that is known to one skilled in the art as being available for substitution can be substituted, which includes one or more substituents, where if more than one substituent then each substituent is independently selected. Such means for substitution are well-known in the art and/or taught by the instant disclosure. For purposes of exemplification, which should not be construed as limiting the scope of this invention, some examples of groups that are substituents are: alkenyl groups, alkoxy group (which itself can be substituted, such as —O—C1-C6-alkyl-OR, —O—C1-C6-alkyl-N(R)2, and OCF3), alkoxyalkoxy, alkoxycarbonyl, alkoxycarbonylpiperidinylalkoxy, alkyl groups (which itself can also be substituted, such as —C1-C6-alkyl-OR, —C1-C6-alkyl-N(R)2, and —CF3), alkylamino, alkylcarbonyl, alkylester, alkylnitrile, alkylsulfonyl, amino, aminoalkoxy, CF3, COH, COOH, CN, cycloalkyl, dialkylamino, dialkylaminoalkoxy, dialkylaminocarbonyl, dialkylaminocarbonylalkoxy, dialkylaminosulfonyl, esters (—C(O)—OR, where R is groups such as alkyl, heterocycloalkyl (which can be substituted), heterocyclyl, etc., which can be substituted), halogen or halo group (F, Cl, Br, I), hydroxy, morpholinoalkoxy, morpholinoalkyl, nitro, oxo, OCF3, optionally substituted phenyl, S(O)2CH3, S(O)2CF3, and sulfonyl, N-alkylamino or N,N-dialkylamino (in which the alkyl groups can also be substituted).
  • Effective Dosage
  • Pharmaceutical compositions suitable for use in the present invention include compositions wherein the active ingredients are contained in an effective amount to achieve its intended purpose. More specifically, a therapeutically effective amount means an amount effective to prevent development of or to alleviate the existing symptoms of the subject being treated. Determination of the effective amounts is well within the capability of those skilled in the art.
  • For any compound used in a method of the present invention, the therapeutically effective dose can be estimated initially from cellular assays. For example, a dose can be formulated in cellular and animal models to achieve a circulating concentration range that includes the IC50 as determined in cellular assays (i.e., the concentration of the test compound which achieves a half-maximal inhibition of a given protein kinase activity). In some cases it is appropriate to determine the IC50 in the presence of 3 to 5% serum albumin since such a determination approximates the binding effects of plasma protein on the compound. Such information can be used to more accurately determine useful doses in humans. Further, the most preferred compounds for systemic administration effectively inhibit protein kinase signaling in intact cells at levels that are safely achievable in plasma.
  • A therapeutically effective dose refers to that amount of a compound of Formula I or a combination of two or more such compounds, which inhibits, totally or partially, the progression of a condition or alleviates, at least partially, one or more symptoms of the condition. A therapeutically effective amount can also be an amount which is prophylactically effective. Toxicity and therapeutic efficacy of such compounds can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the maximum tolerated dose (MTD) and the ED50 (effective dose for % maximal response). The dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio between MTD and ED50. Compounds which exhibit high therapeutic indices are preferred. The data obtained from these cell culture assays and animal studies can be used in formulating a range of dosage for use in humans. The dosage of such compounds lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity. The dosage may vary within this range depending upon the dosage form employed and the route of administration utilized. A therapeutically effective amount can also be an amount which is prophylactically effective. The amount which is therapeutically effective will depend upon the patient's size and gender, the condition to be treated, the severity of the condition and the result sought. For a given patient, a therapeutically effective amount can be determined by methods known to those of skill in the art. The exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient's condition. (See e.g. Fingl et al., 1975, in “The Pharmacological Basis of Therapeutics”, Ch. 1 p 1). In the treatment of crises, the administration of an acute bolus or an infusion approaching the MTD may be required to obtain a rapid response.
  • Dosage amount and interval may be adjusted individually to provide plasma levels of the active moiety which are sufficient to maintain the kinase modulating effects, or minimal effective concentration (MEC). The MEC will vary for each compound but can be estimated from in vitro data; e.g. the concentration necessary to achieve 50-90% inhibition of protein kinase using the assays described herein. Dosages necessary to achieve the MEC will depend on individual characteristics and route of administration. However, HPLC assays or bioassays can be used to determine plasma concentrations.
  • Dosage intervals can also be determined using the MEC value. Compounds should be administered using a regimen which maintains plasma levels above the MEC for 10-90% of the time, preferably between 30-90% and most preferably between 50-90% until the desired amelioration of symptoms is achieved. In cases of local administration or selective uptake, the effective local concentration of the drug may not be related to plasma concentration.
  • The amount of composition administered will, of course, be dependent on the subject being treated, on the subject's weight, the severity of the affliction, the manner of administration and the judgment of the prescribing physician.
  • Packaging
  • The compositions may, if desired, be presented in a pack or dispenser device which may contain one or more unit dosage forms containing the active ingredient. The pack may for example comprise metal or plastic foil, such as a blister pack. The pack or dispenser device may be accompanied by instructions for administration. Compositions comprising a compound of the invention formulated in a compatible pharmaceutical carrier may also be prepared, placed in an appropriate container, and labeled for treatment of an indicated condition.
  • In some formulations it may be beneficial to use the compounds of the present invention in the form of particles of very small size, for example as obtained by fluid energy milling.
  • The use of compounds of the present invention in the manufacture of pharmaceutical compositions is illustrated by the following description. In this description the term “active compound” denotes any compound of the invention but particularly any compound which is the final product of one of the preceding Examples.
  • a) Capsules
  • In the preparation of capsules, 10 parts by weight of active compound and 240 parts by weight of lactose can be de-aggregated and blended. The mixture can be filled into hard gelatin capsules, each capsule containing a unit dose or part of a unit dose of active compound.
  • b) Tablets
  • Tablets can be prepared, for example, from the following ingredients.
  • Parts by weight
    Active compound 10
    Lactose 190
    Maize starch 22
    Polyvinylpyrrolidone 10
    Magnesium stearate 3
  • The active compound, the lactose and some of the starch can be de-aggregated, blended and the resulting mixture can be granulated with a solution of the polyvinyl-pyrrolidone in ethanol. The dry granulate can be blended with the magnesium stearate and the rest of the starch. The mixture is then compressed in a tabletting machine to give tablets each containing a unit dose or a part of a unit dose of active compound.
  • c) Enteric Coated Tablets
  • Tablets can be prepared by the method described in (b) above. The tablets can be enteric coated in a conventional manner using a solution of 20% cellulose acetate phthalate and 3% diethyl phthalate in ethanol:dichloromethane (1:1).
  • d) Suppositories
  • In the preparation of suppositories, for example, 100 parts by weight of active compound can be incorporated in 1300 parts by weight of triglyceride suppository base and the mixture formed into suppositories each containing a therapeutically effective amount of active ingredient.
  • In the compositions of the present invention the active compound may, if desired, be associated with other compatible pharmacologically active ingredients. For example, the compounds of this invention can be administered in combination with another therapeutic agent that is known to treat a disease or condition described herein. For example, with one or more additional pharmaceutical agents that inhibit or prevent the production of VEGF or angiopoietins, attenuate intracellular responses to VEGF or angiopoietins, block intracellular signal transduction, inhibit vascular hyperpermeability, reduce inflammation, or inhibit or prevent the formation of edema or neovascularization. The compounds of the invention can be administered prior to, subsequent to or simultaneously with the additional pharmaceutical agent, whichever course of administration is appropriate. The additional pharmaceutical agents include, but are not limited to any of the agents, for examples, described in pages 20-28. The compounds of the invention and the additional pharmaceutical agents act either additively or synergistically. Thus, the administration of such a combination of substances that inhibit angiogenesis, vascular hyperpermeability and/or inhibit the formation of edema can provide greater relief from the deletrious effects of a hyperproliferative disorder, angiogenesis, vascular hyperpermeability or edema than the administration of either substance alone. In the treatment of malignant disorders combinations with antiproliferative or cytotoxic chemotherapies or radiation are included in the scope of the present invention.
  • The present invention also comprises the use of a compound of formula I as a medicament.
  • A further aspect of the present invention provides the use of a compound of formula I or a salt thereof in the manufacture of a medicament for treating vascular hyperpermeability, angiogenesis-dependent disorders, proliferative diseases and/or disorders of the immune system in mammals, particularly human beings.
  • The present invention also provides a method of treating vascular hyperpermeability, inappropriate neovascularization, proliferative diseases and/or disorders of the immune system which comprises the administration of a therapeutically effective amount of a compound of formula Ito a mammal, particularly a human being, in need thereof.
  • The contents of all references, patents and published patent applications, in their entirety, cited throughout this application are incorporated herein by reference.
  • Assays for Screening Compounds of Formula (I) Enzyme Assays
  • The in vitro potency of compounds in inhibiting one or more of the protein kinases discussed herein or described in the art may be determined by the procedures detailed below.
  • The potency of compounds can be determined by the amount of inhibition of the phosphorylation of an exogenous substrate (e.g., a synthetic peptide (Z. Songyang et al., Nature. 373:536-539) by a test compound relative to control.
  • Homogenous time-resolved fluorescence (HTRF) in vitro kinase assay (see Mathis, G., HTRF(R) Technology. J Biomol Screen, 1999. 4(6): p. 309-314, the contents of which are incorporated in its entirety herein by reference):
  • Purified enzymes (available from commercial sources) were mixed with different amounts of N-biotinylated substrates or GST-tagged substrates (see table) at varying concentrations of inhibitor in different reaction buffers (40 μL final volume, see table). The kinase reaction was initiated by addition of ATP (0.01-0.1 mM final conc.) in a black 96-half-well plate (Perkin Elmer). After 50-60 minutes incubation at room temperature, the reaction was quenched by addition of EDTA (final conc. 100 mM) and developed by addition of revelation reagents (final approximate concentrations: 30 mM HEPES, pH7.0, 0.06% BSA, 0.006% Tween-20, 0.24 M KF, varying amounts of donor europium labeled antibodies and acceptor streptavidin labeled allophycocyanin (SAXL) or anti-GST-XL which are specific to the enzyme reactions. (see table). The developed reaction was incubated in the dark either at room temperature for 10 min, or at 4° C. overnight (see table), then read in a time-resolved fluorescence detector (Discovery, Perkin Elmer or Rubystar, BMG) at 620 nm and 665 nm simultaneously. A 337 nm nitrogen laser was used for excitation. The ratio between the signal of 620 nm and 665 nm was used to calculate the IC50.
  • Specific detailed reaction conditions for the various enzymes are included below:
  • Enzyme Substrate ATP DMSO Reaction
    Assay Conc. Conc. Conc. Conc. Time Detection
    Enzyme Construct/Mw Substrate Buffer (ng/well) (μM) (mM) (%) (min) condition Comments
    Akt1 NA/56 Biotin- Akt 0.12 4 0.1 5 50 13.6 Develop
    kD Bad- Buffer ng/well at 4° C.
    peptide Anti-P- overnight
    BAD-
    Eu; 0.17
    μg/well
    SAXL
    B-Raf Flag-B- GST COT 30 0.15 0.1 5 60 14 Develop
    Raf Unactive Buffer ng/well at 4° C.
    (446-766)/ MEK 1 Anti-P- overnight
    37.3 kD (UBI) MEK
    Eu; 0.75
    μg/well
    Anti-
    GST-XL
    Casine Human Biotin CKII 60 0.5 0.1 5 60 13.6 Develop
    Kinase II recominant/ IκBα- buffer ng/well at 4° C.
    (Calbiochem) 130 kDa peptide Anti-P- overnight
    IκBα-
    Eu; 0.34
    μg/well
    SAXL
    CDK2/Cyclin A C-His CDK2, Biotin- MK2 1.335 0.1 0.1 5 60 15 Develop
    (UBI) N-GST Cyclin MBP buffer ng/well at 4° C.
    A/110 kD protein Anti-P- overnight
    (UBI) MBP-
    Eu; 0.34
    μg/well
    SAXL
    CHK1 CHK1(1-289)- Biotin- PKA 0.6 4 0.1 5 60 1.8 Develop
    His6/33.8 kD cdc25- bufffer ng/well at RT
    peptide Anti-P- 10 min
    14-3-3
    binding
    motif-
    Eu; 0.11
    μg/well
    CR130-100
    COT Flag-COT30- Biotin- COT 25 0.5 0.1 5 60 8.6 Develop
    397/45 kD MEK- buffer ng/well at 4° C.
    peptide Anti-P- overnight
    MEK-
    Eu; 0.34
    μg/well
    SAXL
    Erk2 GST- Biotin- COT 1 0.05 0.1 5 60 15 Develop
    (UBI) Erk2/68 kB MBP Buffer ng/well at 4° C.
    protein Anti-P- overnight
    (UBI) MBP-
    Eu; 0.34
    μg/well
    SAXL
    IKK1 His-IKK1/80 kD Biotin COT 60 0.5 0.1 5 50 13.6 Develop
    IκBα- buffer ng/well at 4° C.
    peptide Anti-P- overnight
    IκBα-
    Eu; 0.34
    μg/well
    SAXL
    IKK2 His-IKK2/80 kD Biotin COT 60 0.5 0.1 5 50 13.6 Develop
    IκBα- buffer ng/well at 4° C.
    peptide Anti-P- overnight
    IκBα-
    Eu; 0.34
    μg/well
    SAXL
    JNK1 His-JNK1/45 kD Biotin- MK2 40 2 0.1 5 60 15 Develop
    (UBI) MBP buffer ng/well at 4° C.
    protein Anti-P- overnight
    (UBI) MBP-
    Eu; 0.34
    μg/well
    SAXL
    MAPKA GST-MK2 Biotin- MK2 1.5 1 0.1 5 60 1.8 Develop
    PK2 (36-400)/68 kD cdc25- buffer ng/well at RT
    peptide Anti-P- 10 min
    14-3-3
    binding
    motif-
    Eu; 0.11
    μg/well
    CR130-100
    MAPKA GST-MK3 Biotin- MK2 3 1 0.1 5 60 1.8 Develop
    PK3 (35-382)/ cdc25- buffer ng/well at RT
    66.9 kD peptide Anti-P- 10 min
    14-3-3
    binding
    motif-
    Eu; 0.11
    μg/well
    CR130-100
    MEK1 GST- unactive COT 3 0.1 0.1 5 60 15 Develop
    (UBI) MEK1-His6/ Erk2 Buffer ng/well at 4° C.
    71 kDa (UBI) Anti-P- overnight
    Erk-
    Eu; 0.39
    μg/well
    Anti-
    GST-XL
    MEKK3 Flag- Unactive COT 85 40 0.1 5 50 15 Develop
    MEKK3 MEK1 Buffer ng/well ng/well at 4° C.
    (2-627)/73 kDa Unactive MEK1 Anti-P- overnight
    Erk2 250 MBP-
    Biotin ng/well Eu; 0.34
    MBP Erk2 μg/well
    protein 0.06 uM SAXL
    (all from Biotin-
    UBI) MBP
    NIK/p100 Flag- Biotin COT 8.5 0.5 0.1 5 60 13.6 Develop
    NIK; IκBα- Buffer ng/well at 4° C.
    HA-p100/ peptide Anti-P- overnight
    203.3 kD IκBα-
    Eu; 0.34
    μg/well
    SAXL
    p38- GST- Biotin COT 1.5 0.1 0.1 5 60 15 Develop
    alpha p38α/64 kD MBP Buffer ng/well at 4° C.
    (UBI) protein Anti-P- overnight
    (UBI) MBP
    Eu; 0.34
    μg/well
    SAXL
    PKA Catalytic Biotin PKA 1.6 1 0.1 5 60 13.6 Develop
    (Invitrogen) subunit, Bad- buffer ng/well at 4° C.
    human peptide Anti-P- overnight
    recimbinant/ BAD-
    43 kDA Eu; 0.17
    μg/well
    SAXL
    PKC His-PKCα/78 kD Biotin PKA 0.4 2 0.1 5 60 1.8 Develop
    alpha cdc25- buffer ng/well at RT
    (UBI) peptide Anti-P- 10 min
    binding
    motif-
    Eu; 0.11
    μg/well
    CR130-100
    PKC His-PKCδ/ Biotin PKA 1.5 1 0.1 5 60 1.8 Develop
    delta 77.5 kD cdc25- buffer ng/well at RT
    (UBI) peptide Anti-P- 10 min
    14-3-3
    binding
    motif-
    Eu; 0.11
    μg/well
    CR130-100
    PRAK His-PRAK/54 kD Biotin MK2 40 1 0.1 5 60 29.2 Develop
    (UBI) MBP buffer ng/well at 4° C.
    protein Anti-P- overnight
    (UBI) MBP-
    Eu; 0.67
    μg/well
    SAXL
    Raf-1 Full length Unactive COT 0.1 7.5 0.1 5 60 15 Develop
    (UBI) human MEK1 buffer U/well ng/well ng/well at 4° C.
    recombinant Unactive MEK1 Anti-P- overnight
    Raf-1/74 kD Erk2 60 MBP-
    Biotin ng/well Eu; 0.34
    MBP Erk2 μg/well
    protein 0.12 μM SAXL
    (all from Biotin-
    UBI) MBP
    SGK1 His- Biotin MK2 0.3 4 0.1 5 60 13.6 Develop
    (UBI) SGK1 Bad- buffer ng/well at 4° C.
    (1-60 peptide Anti-P- overnight
    a.a. BAD-
    deleted, Eu; 0.17
    S422D) μg/well
    SAXL
    cTAK1 Full length/ Biotin COT 30.5 4 0.1 5 60 1.8 Develop
    (UBI) 90 kDa cdc25- Buffer ng/well at RT
    peptide Anti-P- 10 min
    14-3-3
    binding
    motif-
    Eu; 0.11
    μg/well
    CR130-100
  • Reaction Buffers: COT Buffer: 50 mM Tris-HCl, pH7.5; 10 mM MgCl2; 1 mM EGTA; 2 mM DTT; 0.01% Brij; 5 mM Beta-phosphoglycerol MK2 Buffer: 20 mM MOPS, pH7.2; 10 mM MgCl2; 5 mM EGTA; 5 mM Beta-phosphoglycerol; 1 mM Na3VO4; 0.01% Triton-X-100; 1 mM DTT Akt Buffer: 20 mM HEPES, pH7.5; 10 mM MgCl2; 0.01% Triton X-100; 1 mM DTT CKII Buffer: 20 mM Tris, pH7.5; 10 mM MgCl2; 10 mM KCl; 0.01% Triton-X-100; 1 mM DTT; 0.5 mM Na3VO4 PKA Buffer: 25 mM HEPES, pH7.4; 10 mM MgCl2; 0.01% Triton-X-100; 0.5 mM DTT; 0.1 mM Na3VO4 PKC Buffer:
  • 20 mM MOPS, pH7.2; 10 mM MgCl2; 5 mM EGTA; 1.2 mM DTT; 0.01% Triton-X-100; 10 mM Beta-phosphoglycerol; 1.2 mM Na3VO4; 0.1 mg/mL phosphatidylserine; 0.01 mg/mL diacylglycerol; 0.5 mM CaCl2
  • Substrates:
    • Biotin-IκBα-peptide: Biotin-Ahx-LDDRHDSGLDSMKDC-amide
    • Biotin-Bad-peptide: Biotin-EELSPFRGRSRSAPPNLWAAQR-amide
    • Biotin-CKII-substrate-peptide: Biotin-Ahx-RRADDSDDDDD-amide
    • Biotin-cdc25-peptide: Biotin-Ahx-AKVSRSGLYRSPSMPENLNRPR
    • Biotin-MEK-peptide: biotin-AGAGSGQLIDSMANSFVGTR
    • Biotin-MBP protein, GST-unactive MEK1, unactive Erk2 were all purchased from UBI
    Detection Reagents:
    • Anti-P-MBP was purchased from UBI, labeled by Cis-Bio International
    • Anti-P-MEK, Anti-P-BAD, Anti-P-IκBα, Anti-P-Erk were all purchased from Cell-Signaling, and labeled by Cis-Bio International
    • Anti-P-14-3-3 Binding Motif was purchased from Cell-Signaling, labeled by Perkin Elmer
    • SAXL was purchased from Prozyme
    • CR130-100 was purchased from Perkin Elmer
    • Anti-GST-XL was purchased from Cis-Bio International
    Cellular Assays HSP27 Cellular Assay in THP-1 Cells
  • THP-1 cells were serum starved (0.5% FBS) for about 24 hours and seeded to 96 well plates at a density of 2×105 cells/well in 100 ul of low serum media. Test compounds were solubilized in DMSO and added to cells over the range of 25 uM-8 nM (final DMSO conc 0.5%). Compounds were pre-incubated for about 30 mins before the addition of 1 ug/ml LPS. Cells were stimulated for about 45 mins., washed and lysed in 100 ul of Biorad cell lysis buffer. Level of HSP27 phosphorylation was measured via Bio-Plex phosphoprotein assay utilizing pHSP27 Beadmates from Upstate.
  • LPS Induced TNF in THP-1 Cells
  • Thp-1 cells were serum starved (0.5% FBS) for about 24 hours and seeded to 96 well plates at a density of 2×105 cells/well in 100 ul of low serum media. Test compounds were solubilized in DMSO and added to cells over the range of 25 uM-8 nM (final DMSO conc 0.5%). Compounds were pre-incubated for 30 mins before the addition of 1 ug/ml LPS. Cells were stimulated for about 3 hrs. Supernatent media was removed and TNF release was quantified by ELISA. Cellular toxicity was determined by the addition of MTT to the remaining cells.
  • LPS Induced TNF in Peripheral Blood Mononuclear Cell (PBMC) Assay Protocol:
  • Prepare PBMC's from leukopak's by Ficoll separation. Adjust the cell density to 1×107 cells/ml in media.
  • Media used is RPMI Medium 1640 (Gibco BRL, Grand Island, N.Y., Catalog Number 31800)+2% human AB sera (Sigma Chemical Company, St. Louis, Mo., Catalog Number S7148, heat inactivated) with 100 U/ml penicillin (Gibco BRL, Catalog Number 15140), 2 mM L-glutamine (Gibco BRL, Catalog Number 25030), 1×MEM Non-Essential Amino Acids (Gibco BRL, Catalog Number 11140), and 10 mM pH 7.3 Hepes. Media is filtered through a 0.2-micron filter unit.
  • To the wells of 96 well plate(s) apply: 100 uL/well inhibitors (at 2× concentrations) in 1% Dimethyl Sulphoxide, 99% media+100 uL/well PBMC's (1E6 cells/well.)
  • Pre-incubate cells and inhibitors (test compounds) in 37° C. CO2 incubator for about 30 minutes.
  • Apply 10 ng/ml Lipopolysaccharide Escherichia coli (Calbiochem, La Jolla, Calif., Catalog Number 437625) and incubate plate(s) overnight (about 16 hours) in a 37° C. CO2 incubator to stimulate cytokine production.
  • Harvest supernates for cytokine analysis: Spin plate(s) in a centrifuge at 180 g for about 10 minutes with no brake to pellet cells (we used a Beckman GPKR centrifuge and spin at 1,000 rpm.) Remove 100 uL/well supernate for cytokine analysis.
  • For hTNF ELISA, use R&D Systems Catalog Number DTA50 kits and dilute samples about 1/20.
  • After supernates are harvested, cells are used for MTT Assay to assess compound toxicity.
  • PBMC MTT Assay to Assess Cellular Toxicity:
  • MTT is converted into a colored product when it is cleaved by the mitochondrial reductase system, which is present in metabolically active cells. The MTT Assay can be used as a measure of cellular viability.
  • Follow the LPS induced TNF Peripheral Blood Mononuclear Cell (PBMC) Assay Protocol and harvest supernates for cytokine analysis. Use the remaining PBMC's in 96-well plates for the MTT Assay.
  • To cells (in about 1×106 cells/100 μL/well) apply 50 μL/well MTT (2.5 mg/ml in D-PBS, 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl-tetrazolium bromide, Sigma Chemical Company, Catalog Number M-2128) and incubate for 4 hours in a 37° C. CO2 incubator.
  • Apply 50 μL/well of 20% Sodium Dodecyl Sulfate (Natriumlauryl-sulfat, BioRad, Hercules, Calif., Catalog Number 161-0301) and incubate in a 37° C. CO2 incubator overnight. Read the absorbance at 570 nM-630 nM in an ELISA plate reader. The percent viability of cells is then calculated. Toxicity from putative inhibitor(s) is determined by comparison to a control without inhibitor. This is the 100% viable control (1% DMSO/media+cells+MTT+SDS.)

  • OD570/630 of sample/OD570/630 of 100% viable control×100=% viability of sample.
  • LPS Induced TNF in PECs
  • Collect PEC's (peritoneal exudated cells) by washing the peritoneal cavity of B6 mice injected 4 days prior with 2 ml of 3% thioglycollate IP.
  • Wash cells with D-PBS and plate 2.5×105/0.25 ml/well in 96 well plates in 10% FBS RPMI media supplemented with Penicillin-Streptomycin and 2 mM L-Glutamine. Grow cells overnight in 37° C. CO2 incubator. Pre-incubate cells and inhibitors in 1% DMSO/media 0.5% FBS for about 30 minutes. Apply Lipopolysaccharide Escherichia coli (1 μg/ml, Calbiochem, La Jolla, Calif., Catalog Number 437625) and stimulate cells 2 hours in 37° C. CO2 incubator.
  • Harvest Supernates for Cytokine Analysis:
      • Spin plate(s) in a centrifuge at 180 g for about 10 minutes with no brake to pellet cells.
      • Remove 50 uL/well supernate for cytokine analysis.
  • To measure mTNF cytokine levels, use R & D Systems Catalog Number MTA00 ELISA kits. Calculate TNF IC50.
  • LPS Induced TNF and IL-1β in Differentiated Human Peripheral Blood Mononuclear Cells (PBMC)
  • PBMCs are prepared from leukopaks and stored frozen in vials in liquid nitrogen freezer.
  • Thaw PBMCs and plate in 48 well plates at 2×106 cells per well in 400 μl media (RPMI+2% Hu ab serum+Penicillin/Streptomycin+L-glutamine+non-essential amino acids+Hepes+SOng/ml Recombinant Human MCSF). Incubate 24 h at 37° C. 5% CO2. Wash cells 3× with media (no MCSF). In separate 48 well plate, dilute compounds in Media+2% Hu ab serum. For compounds at 10 mM add 10 ul of the compound to 990 μl media then do 1:5 serial dilutions in Media+1% DMSO 200 μl+800 μl media.
  • Remove media from cells and add 250 μl of compound dilutions in duplicate wells of 48 well plates of cells. To negative and positive control wells, add 250 μl media+1% DMSO. Incubate for about 30 minutes 37° C. 5% CO2. Stimulate cells with 10 ng/ml LPS for 3 h 30′ at 37° C. 5% CO2. LPS stock 500 μg/ml: dilute stock 1:5000 in media then add 25 μl to each well except negative controls which get media alone. Incubate for about 3 hours 30 minutes at 37° C. 5% CO2.
  • Add Nigericin (Sigma Cat. # N-7143 FW=747):
  • (Nigericin Final concentration=20 μM: dissolve 2.7 mg in 805 μl ethanol. Dilute this 1:8 in media 250 μl to 1.75 ml. Add 10 μl/well of 48 well plates.) Incubate 30 minutes 37° C. 5% CO2. After 30 minutes, remove supernatant to 96 well plates and assay human IL-1β and human TNFα using R & D Systems ELISA Kits.
  • Compounds of formula I may have therapeutic utility in the treatment of diseases involving both identified, including those not mentioned herein, and as yet unidentified protein tyrosine kinases which are inhibited by compounds of formula I. All compounds exemplified herein significantly inhibit either COT or MK2 at concentrations of 50 micromolar or below.
  • In Vivo Models In Vivo Inhibition of LPS-Induced Cytokines
  • Mice are injected i.v. with LPS (from Escherichia coli Serotype 0111:B4, Sigma #L-4130), dissolved in saline. In order to monitor TNF-α production, 0.1 mpk LPS is given and to measure IFN-γ, IL-1β, IL-18, IL-6, and IL-12, 5mpk LPS is given. The mice are then cardiac bled for serum at the appropriate time points listed below. The animals are bled at 90 minutes for TNF-α or at 4 hours for IFN-γ, IL-1β, IL-18, IL-6, IL-12, then the serum cytokine levels are measured by ELISA. In compound efficacy studies, the compound is dosed either p.o. or i.p. one hour prior to the LPS injection and the levels of target cytokines are measured and compared with those obtained for the control group in order to calculate ED50 levels.
  • Compounds can also be tested in animal models of human disease. These are exemplified by experimental auto-immune encephalomyelitis (EAE) and collagen-induced arthritis (CIA). EAE models which mimic aspects of human multiple sclerosis have been described in both rats and mice (reviewed FASEB J. 5:2560-2566, 1991; murine model: Lab. Invest. 4(3):278, 1981; rodent model: J. Immunol. 146(4):1163-8, 1991). Briefly, mice or rats are immunized with an emulsion of myelin basic protein (MBP), or neurogenic peptide derivatives thereof, and CFA. Acute disease can be induced with the addition of bacterial toxins such as bordetella pertussis. Relapsing/remitting disease is induced by adoptive transfer of T-cells from MBP/peptide immunized animals.
  • CIA may be induced in DBA/1 mice by immunization with type II collagen (J. Immunol:142(7):2237-2243). Mice will develop signs of arthritis as early as ten days following antigen challenge and may be scored for as long as ninety days after immunization. In both the EAE and CIA models, a compound may be administered either prophylactically or at the time of disease onset. Efficacious drugs should reduce severity and/or incidence.
  • Certain compounds of this invention which inhibit one or more angiogenic receptor PTK, and/or a protein kinase such as lck involved in mediating inflammatory responses can reduce the severity and incidence of arthritis in these models.
  • The teachings of all references, including journal articles, patents and published patent applications, are incorporated herein by reference in their entirety.
  • The following examples are for illustrative purposes and are not to be construed as limiting the scope of the present invention.
  • Abbreviations
    • ACN Acetonitrile
    • Racemic-BINAP (±)-2,2′-Bis(diphenylphosphino)-1,1′-binaphthalene
    • (R)-BINAP (R)-(+)-2,2′-Bis(diphenylphosphino)-1,1′-binaphthalene
    • (S)-BINAP (S)-(+2,2′-Bis(diphenylphosphino)-1,1′-binaphthalene
    • Boc tert-Butoxycarbonyl
    • t-BuOH tert-Butyl alcohol
    • t-BuOK Potassium tert-butoxide
    • Cbz Benzyloxycarbonyl
    • DCC N,N′-Dicyclohexylcarbodiimide
    • DCM Dichloromethane
    • DIC N,N′-Diisopropylcarbodiimide
    • DIEA N,N-Diisopropylethylamine
    • DMA N,N-Dimethylacetamide
    • DME 1,2-Dimethoxyethane
    • DMF N,N-Dimethylformamide
    • DMFDMA N,N-Dimethylformamide dimethyl acetal
    • DMSO Dimethyl sulfoxide
    • DPPF 1,1′-Bis(diphenylphosphino)ferrocene
    • EDC 1-(3-Dimethylaminopropyl)-3-ethylcarbodiimide
    • Et2O Diethyl ether
    • Et3N Triethylamine
    • EtOAc Ethyl acetate
    • HATU O-(7-Azabenzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosaphate
    • HBTU O-Benzotriazol-1-yl-N,N,N′,N′-tetramethyluronium hexafluorophosaphate
    • HOAc Acetic acid
    • HOAT 1-Hydroxy-7-azabenzotriazole
    • HOBT 1-Hydroxybenzotriazole
    • HPLC High Performance Liquid Chromatography
    • KOAc Potassium acetate
    • LDA Lithium diisopropylamide
    • MP-carbonate Polymer bound tetraalkylammonium carbonate
    • PMB p-Methoxybenzyl
    • PPh3 Triphenylphosphine
    • PPTS Pyridinium p-toluenesulfonate
    • i-PrOH 2-Propanol
    • RP Reverse Phase
    • Rt Retention time
    • SEM 2-(Trimethylsilyl)ethoxymethyl
    • SEM-Cl 2-(Trimethylsilyl)ethoxymethyl chloride
    • Si-DCT Silica bound dichlorotriazine
    • TBAF tetra-n-Butylammonium fluoride
    • TBDMS tert-Butyldimethylsilane
    • TFA Trifluoroacetic acid
    • TFFH Fluoro-N,N,N′,N′-tetramethylformamidinium hexafluorophosphate
    • THF Tetrahydrofuran
    • TMS Trimethylsilyl
    • XANTPHOS 9,9-Dimethyl-4,5-bis(diphenylphosphino)xanthene
    Synthetic Details
  • Analytical data is defined either within the general procedures or in the tables of examples. Unless otherwise stated, all 1H or 13C NMR data were collected on a Varian Mercury Plus 400 MHz; chemical shifts are quoted in parts per million (ppm). High performance liquid chromatography (HPLC) analytical data are either detailed within the experimental or referenced to the table of HPLC conditions, using the lower case method letter, in Table 1.
  • TABLE 1
    List of HPLC methods
    Method HPLC Conditions
    a LC/MS (30% to 95% acetonitrile/0.01M aqueous ammonium acetate
    over 4.5 min at 0.8 mL/min; UV λ = 190-400 nm; Genesis C18, 3 μm,
    30 × 4.6 mm column; ESI +ve/−ve)
    b RP-HPLC (5% to 85% acetonitrile/0.05M aqueous ammonium
    acetate, buffered to pH 4.5, over 20 min at 1 mL/min; UV λ = 254 nm;
    Hypersil C18, 100 Å, 5 μm, 250 × 4.6 mm column)
    c LC/MS (5% to 100% acetonitrile/5 mM ammonium acetate over 5 min
    at 2.0 ml/min; UV λ = 250-380 nm; Genesis C18, 4 μm, 33 × 4.6 mm column;
    ESI +ve/−ve)
    d LC/MS (5% to 100% acetonitrile/5 mM ammonium acetate over 5 min
    at 2.0 ml/min; UV λ = 250-380 nm; Pecosphere C18, 3 μm, 33 × 4.6 mm
    column; ESI +ve/−ve)
    e LC/MS (30% to 95% acetonitrile/0.01M aqueous ammonium acetate
    over 2.0 min; 95% acetonitrile/0.01M aqueous ammonium acetate
    for 1.5 min at 1.0 mL/min; UV λ = 210-360 nm; Genesis C8, 4 μm, 30 × 4.6 mm
    column; ESI +ve/−ve)
    f LC/MS (10% to 40% acetonitrile/0.01M aqueous ammonium acetate
    over 4.0 min; 40% to 95% acetonitrile/0.01M aqueous ammonium
    acetate over 2.0 min; 95% acetonitrile/0.01M aqueous ammonium
    acetate for 1.0 min at 1.0 mL/min; UV λ = 210-360 nm; Genesis C8, 4 μm,
    30 × 4.6 mm column; ESI +ve/−ve)
    g LC/MS (5% to 95% acetonitrile/0.01M aqueous ammonium acetate
    over 2.0 min; 95% acetonitrile/0.01M aqueous ammonium acetate
    for 1.5 min at 1.4 mL/min; UV λ = 210-360 nm; Genesis C8, 4 μm, 30 × 4.6 mm
    column; ESI +ve/−ve)
    h LC/MS (30% to 95% acetonitrile/0.01M aqueous ammonium acetate
    over 2.0 min; 95% acetonitrile/0.01M aqueous ammonium acetate
    for 3.5 min at 1.0 mL/min; UV λ = 190-400 nm; Genesis C8, 4 μm, 30 × 4.6 mm
    column; ESI +ve/−ve)
    i LC/MS (5% to 35% acetonitrile/0.01M aqueous ammonium acetate
    over 4.0 min; 35%-95% acetonitrile/0.01M aqueous ammonium
    acetate over 2 min; 95% acetonitrile/0.01M aqueous ammonium
    acetate for 1.0 min at 1.0 mL/min; UV λ = 190-400 nm; Genesis C8, 4 μm, 30 × 4.6 mm
    column; ESI +ve/−ve)
    J RP-HPLC (5% to 100% acetonitrile/0.05M aqueous ammonium
    acetate, buffered to pH 4.5, over 10 min at 1 mL/min; UV λ = 254 nm;
    Hypersil C18, 100 Å, 5 μm, 250 × 4.6 mm column)
    k LC/MS (5% to 95% acetonitrile/5 mM aqueous ammonium acetate
    over 3.0 min; 95% to 100% acetonitrile/5 mM aqueous ammonium
    acetate over 0.7 min; 100% to 5% acetonitrile/5 mM aqueous
    ammonium acetate over 0.1 min; 5% acetonitrile/5 mM aqueous
    ammonium acetate for 0.2 min at 2.0 ml/min; λ = 250-380 nm;
    Pecosphere C18, 3 μm, 33 × 4.6 mm column; ESI +ve/−ve)
    l LC/MS (5% acetonitrile/0.01M aqueous ammonium acetate for 0.25 min;
    5-95% acetonitrile/0.01M aqueous ammonium acetate over 2.5 min;
    95% acetonitrile/0.01M aqueous ammonium acetate for 0.85 min;
    95-5% acetonitrile/0.01M aqueous ammonium acetate over 0.15 min;
    5% acetonitrile/0.01M aqueous ammonium acetate for 0.25 min
    at 1.0 mL/min; UV λ = 210-360 nm; Genesis C8, 4 μm, 30 × 4.6 mm column
    ESI +ve/−ve)
    m LC/MS (10% to 100% acetonitrile/0.1% aqueous trifluoroacetic acid
    over 3 min; hold at 100% acetonitrile for 1 min at 1.5 mL/min; ELSD;
    UV λ = 254 nm; Phenomenex Luna C8(2), 5 μm, 100 Å, 30 × 2.0 mm
    column; APCI +ve) 1MS data from flow injection ESI + experiment not the LC-MS
    method (ion not detected in LC-MS run)
  • General Procedures and Examples
  • The general synthetic schemes that were utilized to construct the majority of compounds disclosed in this application are described below in (Schemes 1-25).
  • Figure US20120053345A1-20120301-C00026
  • Figure US20120053345A1-20120301-C00027
  • Figure US20120053345A1-20120301-C00028
  • Figure US20120053345A1-20120301-C00029
  • Figure US20120053345A1-20120301-C00030
  • Figure US20120053345A1-20120301-C00031
  • Figure US20120053345A1-20120301-C00032
  • Figure US20120053345A1-20120301-C00033
  • Figure US20120053345A1-20120301-C00034
  • Figure US20120053345A1-20120301-C00035
  • Figure US20120053345A1-20120301-C00036
  • Figure US20120053345A1-20120301-C00037
  • Figure US20120053345A1-20120301-C00038
  • Figure US20120053345A1-20120301-C00039
  • Figure US20120053345A1-20120301-C00040
  • Figure US20120053345A1-20120301-C00041
  • Figure US20120053345A1-20120301-C00042
  • Figure US20120053345A1-20120301-C00043
  • Figure US20120053345A1-20120301-C00044
  • Figure US20120053345A1-20120301-C00045
  • Figure US20120053345A1-20120301-C00046
  • Figure US20120053345A1-20120301-C00047
  • Figure US20120053345A1-20120301-C00048
  • Figure US20120053345A1-20120301-C00049
  • Figure US20120053345A1-20120301-C00050
  • List of General Procedures
  • General Procedure A: Formation of a urea from an amine
    General Procedure B: Formation of an amide from a carboxylic acid and amine
    General Procedure C: Formation of an amide from a carboxylic acid and amine using Si-DCT
    General Procedure D: Protection of an indazole with a trimethyl-silanylethoxymethyl group
    General Procedure E: Conversion of a bromide to a boronic acid or boronate
    General Procedure F: Suzuki coupling of a boronate or boronic acid with an aryl halide substrate
    General Procedure G: Deprotection of a trimethylsilanylethoxymethyl (SEM) protected indazole
    General Procedure H: Acid cleavage of a TBDMS-protecting group
    General Procedure I: Deprotection of a benzyl ether
    General Procedure J: Reduction of an aldehyde to an alcohol
    General Procedure K: Nucleophilic substitution of an aryl sulfone
    General Procedure L: Reduction of a nitroaromatic compound to an aniline
    General Procedure M: Amide formation from an ester
    General Procedure N: Nucleophilic substitution of an aromatic halide with amine
    General Procedure O: Reductive Alkylation of an Amine with an Aldehyde or a Ketone
    General Procedure P: Deprotection of a methyl-protected alcohol using boron tribromide
    General Procedure Q: Acid catalyzed cleavage of esters, amidines, and carbamates
    General Procedure R: Base-promoted amine alkylation
    General Procedure S: Formation of a sulfonamide from an amine
    General Procedure T: Mitsunobu coupling
    General Procedure U: Sonogashira coupling of an aryl halide with an acetylene
    General Procedure V: Hydrolysis of ester to a carboxylic acid
    General Procedure W: Amide formation from an acid chloride and an amine
    General Procedure X: Indazole formation using hydrazine
    General Procedure Y: Pd mediated coupling of an aryl halide with an amine followed by acid deprotection
    General Procedure Z: Acid cleavage of a THP-protecting group
    General Procedure AA: Deprotection of a Cbz-protected amino group
  • The general procedure letter codes constitute a synthetic route to the final product. A worked example of how the route is determined is given below using the synthesis of Example #G.19 as a non-limiting illustration. Example #G.19 was prepared from 2-(3-{2-(dimethylamino-methyleneamino)-7-[7-benzo[b]thiophen-2-yl-1-(2-trimethylsilanyl-ethoxymethyl)-1H-indazol-5-yl]-pyrrolo[3,2-d]pyrimidin-5-yl}-propan-1-oxy)tetrahydropyran using general procedure G, as represented in the following synthetic scheme:
  • Figure US20120053345A1-20120301-C00051
  • The precursor to Example #G.19, 2-(3-{2-(dimethylamino-methyleneamino)-7-[7-benzo[b]thiophen-2-yl-1-(2-trimethylsilanyl-ethoxymethyl)-1H-indazol-5-yl]-pyrrolo[3,2-d]pyrimidin-5-yl}-propan-1-oxy)tetrahydropyran was prepared via the noted reaction sequence: Preparation #23, E, F (using Preparation #4), R (using 2-(3-bromo-propan-1-oxy) tetrahydropyran, which translates into the following synthetic scheme:
  • Figure US20120053345A1-20120301-C00052
  • The general synthetic methods used in each General Procedure follow, and include an illustration of a compound that was synthesized using the designated General Procedure. None of the specific conditions and reagents noted in the following are to be construed as limiting the scope of the instant invention and are provided for illustrative purposes only.
  • General Procedure A: Formation of a Urea from an Amine
  • Figure US20120053345A1-20120301-C00053
  • To a mixture of thiocarbamic acid methyl ester in an organic solvent (for example, methylene chloride or ethanol, preferably ethanol) is added an amine (2 to 4 equivalents, preferably 3 equivalents). The reaction mixture is stirred at about 40-70° C. (preferably about 50° C.) for about 3-24 hours (preferably, about 5-10 hours). The solvent is removed under reduced pressure to afford the crude product. The crude product can be further purified by crystallization or chromatography.
  • Illustration of General Procedure A Example #11 1-(7-Benzo[b]thiophen-2-yl-1H-indazol-5-yl)-3-(2-pyridin-2-yl-ethyl)-urea
  • Figure US20120053345A1-20120301-C00054
  • A mixture of 7-benzo[b]thiophen-2-yl-1H-indazole-5-yl-thiocarbamic acid methyl ester (Preparation #14, 25.0 mg, 0.074 mmol) and 2-pyridin-2-yl-ethylamine (23 mg, 0.185 mmol) in ethanol (2 mL) was stirred at about 50° C. for about 8 hours. The solvent was removed under reduced pressure and the product was purified by crystallization in ethanol to give 1-(7-benzo[b]thiophen-2-yl-1H-indazol-5-yl)-3-(2-pyridin-2-yl-ethyl)-urea (12 mg, 39% yield); RP HPLC (Table 1, Method e) Rt=1.82 min; MS m/z: (M−H) 412.
  • TABLE A
    Examples synthesized using general procedure A from Preparation #14
    Figure US20120053345A1-20120301-C00055
    Figure US20120053345A1-20120301-C00056
    Figure US20120053345A1-20120301-C00057
    HPLC Rt
    Example (min)
    Amine Product # (Method) m/z
    4-Nitroaniline
    Figure US20120053345A1-20120301-C00058
    A.1 2.21 (a) 558 (M − H)
    2-Piperidin-1-yl- ethylamine
    Figure US20120053345A1-20120301-C00059
    A.2 1.96 (a) 420 (M + H)+
    2-(1H-Imidazol- 4-yl)-ethylamine
    Figure US20120053345A1-20120301-C00060
    A.3 1.63 (a) 403 (M + H)+
    2-Methoxy- ethylamine
    Figure US20120053345A1-20120301-C00061
    A.4 1.75 (a) 367 (M + H)+
    N,N-Dimethyl- ethane-1,2- diamine
    Figure US20120053345A1-20120301-C00062
    A.5 1.75 (a) 380 (M + H)+
    Tetrahydro-pyran- 4-ylamine
    Figure US20120053345A1-20120301-C00063
    A.6 1.70 (a) 391 (M − H)
    2-Pyridin-3-yl- ethylamine
    Figure US20120053345A1-20120301-C00064
    A.7 1.60 (a) 414 (M + H)+
    3-Amino-propan- 1-ol
    Figure US20120053345A1-20120301-C00065
    A.8 1.50 (a) 367 (M + H)+
    2-Amino- propane-1,3-diol
    Figure US20120053345A1-20120301-C00066
    A.9 1.32 (a) 383 (M + H)+
    Pyridin-2-yl- methylamine
    Figure US20120053345A1-20120301-C00067
    A.10 1.75 (a) 400 (M + H)+
    Pyridin-3-yl- methylamine
    Figure US20120053345A1-20120301-C00068
    A.11 1.71 (a) 400 (M + H)+
    3-Amino- propionic acid
    Figure US20120053345A1-20120301-C00069
    A.12 0.65 (a) 381 (M + H)+
    2-Amino-ethanol
    Figure US20120053345A1-20120301-C00070
    A.13 1.42 (a) 383 (M + H)+
    1-Methoxy-but-2- ylamine
    Figure US20120053345A1-20120301-C00071
    A.14 1.93 (a) 393 (M − H)
    (5-Methyl- pyrazin-2-yl)- methylamine
    Figure US20120053345A1-20120301-C00072
    A.15 1.71 (a) 415 (M + H)+
    2-(1-Methyl-1H- pyrrol-2-yl)- ethylamine
    Figure US20120053345A1-20120301-C00073
    A.16 2.05 (a) 416 (M + H)+
    2-Pyridin-4-yl- ethylamine
    Figure US20120053345A1-20120301-C00074
    A.17 1.62 (a) 412 (M − H)
    (1,3,5-Trimethyl- 1H-pyrazol-4-yl)- methylamine
    Figure US20120053345A1-20120301-C00075
    A.18 1.61 (a) 429 (M − H)
    3-Amino- propionamide
    Figure US20120053345A1-20120301-C00076
    A.19 1.36 (a) 380 (M + H)+
    (6-Chloro- pyridin-3-yl)- methylamine
    Figure US20120053345A1-20120301-C00077
    A.20 1.92 (a) 434 (M + H)+
    1-Pyridin-3-yl- ethylamine
    Figure US20120053345A1-20120301-C00078
    A.21 1.70 (a) 412 (M − H)
    1-Methyl- piperidin-4- ylamine
    Figure US20120053345A1-20120301-C00079
    A.22 1.78 (a) 404 (M − H)
    2-Piperazin-1-yl- ethanol
    Figure US20120053345A1-20120301-C00080
    A.23 1.42 (a) 422 (M + H)+
    Bis-(2-methoxy- ethyl)-amine
    Figure US20120053345A1-20120301-C00081
    A.24 2.09 (a) 425 (M + H)+
    1-Methyl piperazine
    Figure US20120053345A1-20120301-C00082
    A.25 1.62 (a) 392 (M + H)+
    [2-(Thiophen-2- yl)-thiazol-4-yl]- methylamine
    Figure US20120053345A1-20120301-C00083
    A.26 2.09 (a) 488 (M + H)+
    Benzothiazol-2- yl-methylamine
    Figure US20120053345A1-20120301-C00084
    A.27 2.00 (a) 456 (M + H)+
    Oxazol-2-yl- methylamine
    Figure US20120053345A1-20120301-C00085
    A.28 1.65 (a) 388 (M − H)
    Imidazo[1,2- a]pyridin-2-yl- methylamine
    Figure US20120053345A1-20120301-C00086
    A.29 1.69 (a) 437 (M − H)
    Benzo[2,1,3] thiadiazol-5-yl- methylamine
    Figure US20120053345A1-20120301-C00087
    A.30 1.98 (a) 455 (M − H)
    5-Aminomethyl- 2H-[1,2,4]triazol- 3-ylamine
    Figure US20120053345A1-20120301-C00088
    A.31 1.19 (a) 403 (M − H)
    5-(2-Amino- ethyl)-2H- [1,2,4]triazol-3- ylamine
    Figure US20120053345A1-20120301-C00089
    A.32 1.22 (a) 419 (M + H)+
    Morpholine
    Figure US20120053345A1-20120301-C00090
    A.33 1.79 (a) 379 (M + H)+
    2-Methylamino- ethanol
    Figure US20120053345A1-20120301-C00091
    A.34 1.50 (a) 367 (M + H)+
    N,N,N′-Trimethyl- ethane-1,2- diamine
    Figure US20120053345A1-20120301-C00092
    A.35 1.32 (a) 394 (M + H)+
    N,N,N′-Trimethyl- propane-1,3- diamine
    Figure US20120053345A1-20120301-C00093
    A.36 1.37 (a) 408 (M + H)+
    3 -Methylamino- propane-1,2-diol
    Figure US20120053345A1-20120301-C00094
    A.37 1.38 (a) 397 (M + H)+
    Piperidin-4-yl- methanol
    Figure US20120053345A1-20120301-C00095
    A.38 1.57 (a) 407 (M + H)+
    Piperazine
    Figure US20120053345A1-20120301-C00096
    A.39 1.14 (a) 378 (M + H)+
    (2-Methoxy- ethyl)-methyl- amine
    Figure US20120053345A1-20120301-C00097
    A.40 1.92 (a) 381 (M + H)+
    Piperidin-4-ol
    Figure US20120053345A1-20120301-C00098
    A.41 1.51 (a) 393 (M + H)+
    (S)-1-Pyrrolidin- 2-yl-methanol
    Figure US20120053345A1-20120301-C00099
    A.42 1.71 (a) 393 (M + H)+
    (R)-1-Pyrrolidin- 2-yl-methanol
    Figure US20120053345A1-20120301-C00100
    A.43 1.71 (a) 393 (M + H)+
    Bis-(2- ethanol)amine
    Figure US20120053345A1-20120301-C00101
    A.44 1.46 (a) 397 (M + H)+
    Ammonia
    Figure US20120053345A1-20120301-C00102
    A.45 1.42 (a) 309 (M + H)+
    Methylamine
    Figure US20120053345A1-20120301-C00103
    A.46 1.67 (a) 323 (M + H)+
    Dimethylamine
    Figure US20120053345A1-20120301-C00104
    A.47 1.78 (a) 335 (M − H)
    Pyrid-4-yl methylamine
    Figure US20120053345A1-20120301-C00105
    A.48 1.67 (a) 400.0 (M + H)+ 398.0 (M − H)
    4-(2-Aminoethyl) morpholine
    Figure US20120053345A1-20120301-C00106
    A.49 1.57 (a) 422.0 (M + H)+ 419.5 (M − H)

    General Procedure B: Formation of an Amide from Carboxylic Acid and Amine
  • Figure US20120053345A1-20120301-C00107
  • To a mixture of a carboxylic acid (1-2.5 equivalents, preferably 1-1.5 equivalents) and an amine (1-2.5 equivalents, preferably 1-1.5 equivalents) in an organic solvent (for example, THF, EtOAc, Et2O, or DMF, preferably DMF) is added the coupling reagent (for example, DCC, DIC, EDC, HBTU, HATU or TFFH, preferably HBTU) (1-5 equivalents, preferably 1.2 equivalents), with or without a coupling additive (for example, HOBT or HOAT, preferably HOBT) (0.1-5 equivalents, preferably 0.2 equivalents) and, optionally, DIEA (0.1-25 equivalents, preferably 3 equivalents). The reaction mixture is stirred at about 20-70° C. (preferably about 50° C.) for about 5-40 hours (preferably about 35 hours) and then cooled to ambient temperature. The reaction mixture can be purified in three different ways: 1). The reaction mixture is treated with MP-carbonate (3-10 equivalents, preferably 5 equivalents) with or without methanol. After about 4-48 hours (preferably about 14 hours), the reaction solution is separated from the resin by filtration and concentrated under reduced pressure to afford the crude product that can be further purified by crystallization or chromatography. If the product precipitates prior to filtration of the resin, the suspension containing the product is separated from the resin via pipette and filtered to afford the crude product that can be further purified by crystallization or chromatography. 2). The reaction mixture is partitioned between water and an organic solvent (for example, CH2Cl2, EtOAc or Et2O, preferably CH2Cl2). The organic layer is separated and the aqueous layer is further extracted with organic solvent. The combined organic extracts are dried over a desiccant and evaporated under reduced pressure to afford the product that can be further purified by crystallization or chromatography. 3). The reaction mixture is directly concentrated under reduced pressure and the residue is purified by crystallization or chromatography.
  • Illustration of General Procedure B Example #12 N-Benzyl 5-(5-Amino-1H-pyrrolo[2,3-c]pyridin-3-yl)-1H-indazole-7-carboxamide acetate
  • Figure US20120053345A1-20120301-C00108
  • A solution of 5-(5-amino-1H-pyrrolo[2,3-c]pyridin-3-yl)-1H-indazole-7-carboxylic acid hydrochloride (Preparation #13, 32.9 mg, 0.10 mmol) and benzylamine (24.0 μL, 0.22 mmol) in DMF (1.0 mL) were treated with o-(7-azabenzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate (38.0 mg, 0.10 mmol) at ambient temperature and the reaction was stirred for about 1 hour. The reaction was concentrated under reduced pressure and the residue was purified by preparative HPLC (Waters Symmetry C8 column (25×100 mm, 7 μm particle size) using a gradient of 10%-100% CH3CN/0.1% aqueous TFA over 8 min (10 min run time) at a flow rate of 40 mL/min). Product fractions were combined and concentrated to remove organic solvents and then lyophilized to yield N-benzyl 5-(5-amino-1H-pyrrolo[2,3-c]pyridin-3-yl)-1H-indazole-7-carboxamide acetate (10 mg, 23%) as an off-white powder; RP-HPLC (Table 1, Method e) Rt 1.94 min; m/z: (M−H) 381.
  • TABLE B.1
    Examples were prepared using general procedure B
    Figure US20120053345A1-20120301-C00109
    HPLC Rt
    (min)
    Acid Amine Product Ex # (Method) m/z
    Preparation #13 Aniline
    Figure US20120053345A1-20120301-C00110
    B.1.1 1.97 (e) 367 (M − H)
    Preparation #11c, V Aniline
    Figure US20120053345A1-20120301-C00111
    B.1.2 1.63 (e) 314 and 316 (M + H+)
  • TABLE B.2
    Examples prepared using general procedure B from Example #F.8.1
    Figure US20120053345A1-20120301-C00112
    Figure US20120053345A1-20120301-C00113
    HPLC Rt
    Example (min)
    Acid Product # (Method) m/z
    1H- Benzimidazole-5- carboxylic acid
    Figure US20120053345A1-20120301-C00114
    B.2.1 1.69 (e) 409.9 (M + H)+
    Imidazo[2,1- b]thiazole-6- carboxylic acid
    Figure US20120053345A1-20120301-C00115
    B.2.2 2.02 (e) 415.9 (M + H)+
    1H-Pyrazole-4- carboxylic acid
    Figure US20120053345A1-20120301-C00116
    B.2.3 1.60 (e) 359.9 (M + H)+
    1H-Indazole-3- carboxylic acid
    Figure US20120053345A1-20120301-C00117
    B.2.4 2.14 (e) 409.9 (M + H)+
    [(2R)-3,6- Dioxopiperazin-2- yl]acetic acid
    Figure US20120053345A1-20120301-C00118
    B.2.5 1.26 (e) 419.9 (M + H)+
    3-Methyl-5-(4- methyl-1,2,3- thiadiazol-5- yl)isoxazole-4- carboxylic acid
    Figure US20120053345A1-20120301-C00119
    B.2.6 2.21 (e) 470.5 (M + H)+
    N- (Aminocarbonyl) glycine
    Figure US20120053345A1-20120301-C00120
    B.2.7 1.33 (e) 365.9 (M + H)+
    3-Amino-1- carboxymethyl- pyridin-2-one trifluoroacetate
    Figure US20120053345A1-20120301-C00121
    B.2.8 1.61 (e) 415.9 (M + H)+
    3-Aminopyrazine- 2-carboxylic acid
    Figure US20120053345A1-20120301-C00122
    B.2.9 2.24 (e) 384.5 (M − H)
    6-Aminonicotinic acid
    Figure US20120053345A1-20120301-C00123
    B.2.10 1.70 (e) 385.9 (M + H)+
    2-Hydroxy cinchoninic acid
    Figure US20120053345A1-20120301-C00124
    B.2.11 1.84 (e) 437.1 (M + H)+
    1H-Benzimidazol- 2-ylacetic acid
    Figure US20120053345A1-20120301-C00125
    B.2.12 1.87 (e) 388.0 (M + H)+
    2-(Acetylamino)- 1,3-thiazole-4- carboxylic acid
    Figure US20120053345A1-20120301-C00126
    B.2.13 1.84 (e) 433.8 (M + H)+
    2-Aminonicotinic acid
    Figure US20120053345A1-20120301-C00127
    B.2.14 1.99 (e) 385.9 (M + H)+
    3- Aminoisonicotinic acid
    Figure US20120053345A1-20120301-C00128
    B.2.15 1.85 (e) 386.0 (M + H)+
    4-(Methylamino)- 6-morpholin-4-yl- 1,3,5-triazine-2- carboxylic acid
    Figure US20120053345A1-20120301-C00129
    B.2.16 2.00 (e) 486.9 (M + H)+
    3-[(4R)-2,5- Dioxoimidazo- lidin-4- yl]propanoic acid
    Figure US20120053345A1-20120301-C00130
    B.2.17 1.45 (e) 417.9 (M − H)
    2,6- Dihydroxypyri- midine-4- carboxylic acid - lithium monohydrate
    Figure US20120053345A1-20120301-C00131
    B.2.18 1.34 (e) 401.6 (M − H)
    3-(1H-Imidazol-2- yl)propanoic acid
    Figure US20120053345A1-20120301-C00132
    B.2.19 1.34 (e) 388.0 (M + H)+
    3-(5,6,7,8- Tetrahydro-1,8- naphthyridin-2- yl)propanoic acid
    Figure US20120053345A1-20120301-C00133
    B.2.20 1.73 (e) 454.0 (M + H)+
    4- Aminopyrimidine- 5-carboxylic acid
    Figure US20120053345A1-20120301-C00134
    B.2.21 1.76 (e) 387.3 (M + H)+
    5-Methyl-4-oxo- 3,4-dihydro- thieno[2,3- d]pyrimidine-6- carboxylic acid
    Figure US20120053345A1-20120301-C00135
    B.2.22 1.78 (e) 458.3 (M + H)+
    7-Amino-2- methylpyrazolo [1,5-a]pyrimidine- 6-carboxylic acid
    Figure US20120053345A1-20120301-C00136
    B.2.23 2.00 (e) 440.3 (M + H)+
    2,1,3- Benzoxadiazole- 5-carboxylic acid
    Figure US20120053345A1-20120301-C00137
    B.2.24 2.27 (e) 406.3 (M + H)+
    1H-Imidazole-2- carboxylic acid
    Figure US20120053345A1-20120301-C00138
    B.2.25 1.82 (e) 360.2 (M + H)+
    1H- Benzimidazole-2- carboxylic acid
    Figure US20120053345A1-20120301-C00139
    B.2.26 2.19 (e) 410.2 (M + H)+
    2,7- Dimethylpyrazolo [1,5- a]pyrimidine-6- carboxylic acid
    Figure US20120053345A1-20120301-C00140
    B.2.27 1.95 (e) 439.4 (M + H)+
    5-Oxo-2,3- dihydro-5H-[1,3] thiazolo[3,2- a]pyrimidine-6- carboxylic acid
    Figure US20120053345A1-20120301-C00141
    B.2.28 1.97 (e) 446.2 (M + H)+
    3-Hydroxy quinoxaline-2- carboxylic acid
    Figure US20120053345A1-20120301-C00142
    B.2.29 1.85 (e) 438.3 (M + H)+
    5-Aminonicotinic acid
    Figure US20120053345A1-20120301-C00143
    B.2.30 1.60 (g) 386.2 (M + H)+
    6-Oxo-1,4,5,6- tetrahydro- pyridazine-3- carboxylic acid
    Figure US20120053345A1-20120301-C00144
    B.2.31 1.63 (g) 390.2 (M + H)+
    2- (Acetylamino) isonicotinic acid
    Figure US20120053345A1-20120301-C00145
    B.2.32 1.68 (g) 428.3 (M + H)+
    (3R)-5- Oxopyrrolidine-3- carboxylic acid
    Figure US20120053345A1-20120301-C00146
    B.2.33 1.49 (g) 377.2 (M + H)+
    (4R)-2,6- Dioxohexahydro- pyrimidine-4- carboxylic acid
    Figure US20120053345A1-20120301-C00147
    B.2.34 1.48 (g) 406.3 (M + H)+
    1H-Imidazole-4- carboxylic acid
    Figure US20120053345A1-20120301-C00148
    B.2.35 1.58 (g) 360.2 (M + H)+
    1H-1,2,3- Benzotriazole-5- carboxylic acid
    Figure US20120053345A1-20120301-C00149
    B.2.36 1.66 (g) 411.2 (M + H)+
    1H-Pyrazole-3- carboxylic acid
    Figure US20120053345A1-20120301-C00150
    B.2.37 1.68 (g) 360.2 (M + H)+
    1-Methyl-1H- benzimidazole-2- carboxylic acid
    Figure US20120053345A1-20120301-C00151
    B.2.38 2.24 (g) 424.3 (M + H)+
    Sodium benzothiazole-2- carboxylate
    Figure US20120053345A1-20120301-C00152
    B.2.39 2.35 (g) 427.2 (M + H)+
  • TABLE B.3
    Examples prepared using general procedure C from Preparation #7.
    Figure US20120053345A1-20120301-C00153
    Example Rt/min m/z
    Amine Precursor Product # (Method) (ESI+)
    N-(2-amino-phenyl)- acetamide
    Figure US20120053345A1-20120301-C00154
    B.3.1 1.44(e) 426.9 (M + H)+
    3-methoxymethyl- pyrrolidine
    Figure US20120053345A1-20120301-C00155
    B.3.2 1.52(e) 391.9 (M + H)+
    5-amino-piperidin- 2-one
    Figure US20120053345A1-20120301-C00156
    B.3.3 1.43(e) 391 (M + H)+
    Piperidin-4- ylmethyl-pyrimidin- 2-yl-amine
    Figure US20120053345A1-20120301-C00157
    B.3.4 1.83(e) 469.0 (M + H)+
    3-Imidazol-1- yl-propylamine
    Figure US20120053345A1-20120301-C00158
    B.3.5 1.71(e) 401 (M + H)+
    Piperidin-4- yl-methanol
    Figure US20120053345A1-20120301-C00159
    B.3.6 1.13(e) 391.9 (M + H)+
    1-Methyl-1H- benzoimidazol- 4-ylamine
    Figure US20120053345A1-20120301-C00160
    B.3.7 2.13(e) 423.9 (M + H)+
    2-Pyrrolidin- 3-yl-pyridine
    Figure US20120053345A1-20120301-C00161
    B.3.8 1.88(e) 424.9 (M + H)+
    5-Methylsulfanyl- 1H-[1,2,4]triazol- 3-ylamine
    Figure US20120053345A1-20120301-C00162
    B.3.9 1.72(e) 404.6 (M + H)+
    5-Benzooxazol-2-yl- 2-methoxy-phenyl amine
    Figure US20120053345A1-20120301-C00163
    B.3.10 2.13(e) 517.0 (M + H)+
    N-(6-Amino- benzothiazol-2-yl)- acetamide
    Figure US20120053345A1-20120301-C00164
    B.3.11 1.45(e) 483.9 (M + H)+
    1H-Indazol-6- ylamine
    Figure US20120053345A1-20120301-C00165
    B.3.12 1.47(e) 408.0 (M − H)
    1H-Indazol-5- ylamine
    Figure US20120053345A1-20120301-C00166
    B.3.13 1.37(e) 407.6 (M − H)
    1H-Benzoimidazol- 5-ylamine
    Figure US20120053345A1-20120301-C00167
    B.3.14 1.23(e) 409.9 (M + H)+
    Quinazolin-4- ylamine
    Figure US20120053345A1-20120301-C00168
    B.3.15 2.35(e) 420.2 (M − H)
    4-(4-Fluoro-phenyl)- 5-methyl-2H- pyrazol-3-ylamine
    Figure US20120053345A1-20120301-C00169
    B.3.16 2.48(e) 466.3 (M − H)
    5-Methyl-1H- [1,2,4]triazol-3- ylamine
    Figure US20120053345A1-20120301-C00170
    B.3.17 2.02(e) 375.1 (M + H)+
    4-Cyclohexyl- phenylamine
    Figure US20120053345A1-20120301-C00171
    B.3.18 2.71(e) 450.4 (M − H)
    2-Amino-4-hydroxy- benzamide
    Figure US20120053345A1-20120301-C00172
    B.3.19 1.85(e) 427.2 (M − H)
    C-Oxazol-2-yl- methylamine
    Figure US20120053345A1-20120301-C00173
    B.3.20 1.62(e) 373.2 (M − H)
    [1,3,4]Thiadiazol-2- ylamine
    Figure US20120053345A1-20120301-C00174
    B.3.21 1.79(e) 376.1 (M − H)
    4-Methyl-thiazol-2- ylamine
    Figure US20120053345A1-20120301-C00175
    B.3.22 2.11(e) 391.1 (M + H)+
    5-Furan-2-yl-1H- pyrazol-3-ylamine
    Figure US20120053345A1-20120301-C00176
    B.3.23 2.21(e) 424.2 (M − H)
    2-(1H-Indol-2-yl)- phenylamine
    Figure US20120053345A1-20120301-C00177
    B.3.24 2.35(e) 483.2 (M − H)
    Biphenyl-2-ylamine
    Figure US20120053345A1-20120301-C00178
    B.3.25 2.35(e) 444.3 (M − H)
    4-(1H-Imidazo[1,2- a]pyridin-2-yl)- phenylamine
    Figure US20120053345A1-20120301-C00179
    B.3.26 1.97(e) 486.3 (M + H)+
    5-Amino-1,3- dihydro- benzoimidazol-2-one
    Figure US20120053345A1-20120301-C00180
    B.3.27 1.49(e) 424.2 (M − H)
    2-Methyl- benzothiazol- 5-ylamine
    Figure US20120053345A1-20120301-C00181
    B.3.28 2.09(e) 439.3 (M − H)
    5-Aminomethyl-2H- [1,2,4]triazol-3- yllamine
    Figure US20120053345A1-20120301-C00182
    B.3.29 1.20(e) 388.2 (M − H)
    2-Amino-fluoren-9- one
    Figure US20120053345A1-20120301-C00183
    B.3.30 2.36(e) 470.3 (M − H)
    N-(3-Amino- phenyl)-benzamide
    Figure US20120053345A1-20120301-C00184
    B.3.31 2.10(e) 487.3 (M − H)
    C-(5-Methyl-3- phenyl-isoxazol-4- yl)-methylamine
    Figure US20120053345A1-20120301-C00185
    B.3.32 2.08(e) 2.08 (M − H)
    Indan-5-ylamine
    Figure US20120053345A1-20120301-C00186
    B.3.33 2.37(e) 408.3 (M − H)
    2-(4-Phenoxy- phenyl)-ethylamine
    Figure US20120053345A1-20120301-C00187
    B.3.34 2.38(e) 488.3 (M − H)
    4-Amino-4-methyl- pentan-2-one
    Figure US20120053345A1-20120301-C00188
    B.3.35 1.94(e) 390.2 (M − H)
    5-(2-Amino-ethyl)- 2H-[1,2,4]triazol- 3-ylamine
    Figure US20120053345A1-20120301-C00189
    B.3.36 1.21(e) 403.2 (M − H)
    Benzo[b]thiophen-5- ylamine
    Figure US20120053345A1-20120301-C00190
    B.3.37 2.26(e) 424.3 (M − H)
    3-Amino-4- methoxy-benzamide
    Figure US20120053345A1-20120301-C00191
    B.3.38 1.69(e) 443.3 (M + H)+
    2-Methoxy- phenylamine
    Figure US20120053345A1-20120301-C00192
    B.3.39 2.34(e) 400.2 (M + H)+
    4-Methoxy- biphenyl-3-ylamine
    Figure US20120053345A1-20120301-C00193
    B.3.40 2.69(e) 476.3 (M + H)+
    N-(3-Amino-4- methoxy-phenyl)- acetamide
    Figure US20120053345A1-20120301-C00194
    B.3.41 1.86(e) 457.3 (M + H)+
    2,5-Dimethoxy- phenylamine
    Figure US20120053345A1-20120301-C00195
    B.3.42 2.39(e) 430.3 (M + H)+
    3-Methoxy- phenylamine
    Figure US20120053345A1-20120301-C00196
    B.3.43 2.24(e) 400.3 (M + H)+
    4-Methoxy- phenylamine
    Figure US20120053345A1-20120301-C00197
    B.3.44 2.17(e) 400.3 (M + H)+
    3-(1H- Benzoimidazol-2- yl)-phenylamine
    Figure US20120053345A1-20120301-C00198
    B.3.45 2.11(e) 486.3 (M + H)+
    3-Amino- benzamidine
    Figure US20120053345A1-20120301-C00199
    B.3.46 1.95(e) 412.3 (M + H)+
    3-Amino-benzoic acid methyl ester
    Figure US20120053345A1-20120301-C00200
    B.3.47 2.29(e) 458.3 (M + H)+
    3-(1H-Tetrazol-5- yl)-phenylamine
    Figure US20120053345A1-20120301-C00201
    B.3.48 1.56(e) 438.3 (M + H)+
    2-Amino-phenol
    Figure US20120053345A1-20120301-C00202
    B.3.49 2.16(e) 386.2 (M + H)+
    3-Amino-4-hydroxy- benzoic acid methyl ester
    Figure US20120053345A1-20120301-C00203
    B.3.50 2.13(e) 444.2 (M + H)+
    3-Benzooxazol-2-yl- phenylamine
    Figure US20120053345A1-20120301-C00204
    B.3.51 2.54(e) 484.7 (M − H)
    Phenylamine
    Figure US20120053345A1-20120301-C00205
    B.3.52 2.27(e) 370.2 (M + H)+
    5-Amino-2-phenyl- 2,4-dihydro-pyrazol- 3-one
    Figure US20120053345A1-20120301-C00206
    B.3.53 1.77(e) 450.5 (M − H)
    4-Amino-N-[5- methyl-3H- [1,3,4]oxadiazol- (2E)-ylidene]- benzenesulfonamide
    Figure US20120053345A1-20120301-C00207
    B.3.54 1.51(e) 529.0 (M − H)
    5-Trifluoromethyl- [1,3,4]thiadiazol- 2-ylamine
    Figure US20120053345A1-20120301-C00208
    B.3.55 2.42(e) 443.8 (M − H)
    N-(3-Amino- phenyl)-acetamide
    Figure US20120053345A1-20120301-C00209
    B.3.56 1.86(e) 427.3 (M + H)+
    6-Morpholin-4-yl- pyridin-3-ylamine
    Figure US20120053345A1-20120301-C00210
    B.3.57 1.92(e) 456.3 (M + H)+
    2-Amino-5,6- dihydro-4H- benzothiazol-7-one
    Figure US20120053345A1-20120301-C00211
    B.3.58 2.12(e) 445.3 (M + H)+
    N-(4-Amino-2,5- dimethoxy-phenyl)- benzamide
    Figure US20120053345A1-20120301-C00212
    B.3.59 2.46(e) 547.0 (M + H)+
    4-Morpholin-4-yl- phenylamine
    Figure US20120053345A1-20120301-C00213
    B.3.60 2.06(e) 455.4 (M + H)+
    4-(6-Methyl- benzothiazol-2-yl)- phenylamine
    Figure US20120053345A1-20120301-C00214
    B.3.61 2.77(e) 515.5 (M − H)
    4-Benzooxazol-2-yl- phenylamine
    Figure US20120053345A1-20120301-C00215
    B.3.62 2.56(e) 487.2 (M + H)+
    4-(1H- Benzoimidazol-2- yl)-phenylamine
    Figure US20120053345A1-20120301-C00216
    B.3.63 2.0(e) 483.8 (M − H)−
    5-(1H- Benzoimidazol-2- yl)-2-methoxy- phenylamine
    Figure US20120053345A1-20120301-C00217
    B.3.64 2.1(e) 516.2 (M + H)+
    (Labotest)
    Glycinamide
    Figure US20120053345A1-20120301-C00218
    B.3.65 1.37(e) 351.2 (M + H)+
    4-Amino-N- (3,4-dimethyl- isoxazol-5-yl)- benzenesulfonamide
    Figure US20120053345A1-20120301-C00219
    B.3.66 2.04(e) 542.3 (M − H)
    4-Amino-N-thiazol- 2-yl- benzenesulfonamide
    Figure US20120053345A1-20120301-C00220
    B.3.67 1.85(e) 530.4 (M − H)
    6-(4-Methyl- piperazin-1-yl)- pyridin-3-ylamine
    Figure US20120053345A1-20120301-C00221
    B.3.68 1.35(e) 467.5 (M − H)
    4-Amino-N-(5- methyl-isoxazol-3- yl)- benzenesulfonamide
    Figure US20120053345A1-20120301-C00222
    B.3.69 2.08(e) 527.9 (M + H)+
    5-Amino-pyridine-2- sulfonic acid isopropylamide
    Figure US20120053345A1-20120301-C00223
    B.3.70 2.62(e) 489.1 (M + H)+
    6-(Propane-1- sulfonyl)-1H- benzoimidazol-2- ylamine
    Figure US20120053345A1-20120301-C00224
    B.3.71 2.07(e) 514.6 (M − H)
    4-Amino-N- (4,6-dimethyl- pyrimidin-2-yl)- benzenesulfonamide
    Figure US20120053345A1-20120301-C00225
    B.3.72 1.94(e) 553.5 (M − H)
    4-Amino-N- (2,6-dimethyl- pyrimidin-4-yl)- benzenesulfonamide
    Figure US20120053345A1-20120301-C00226
    B.3.73 1.59(e) 553.5 (M − H)
    N-(4-Amino-2- methoxy-phenyl)-2- methoxy-benzamide
    Figure US20120053345A1-20120301-C00227
    B.3.74 2.38(e) 547.2 (M − H)
    5-tert-Butyl- [1,3,4]thiadiazol- 2-yl amine
    Figure US20120053345A1-20120301-C00228
    B.3.75 2.34(e) 432.2 (M − H)
    3-Amino-benzamide
    Figure US20120053345A1-20120301-C00229
    B.3.76 1.75(e) 411.2 (M − H)
    2-(1H-Imidazol-4- yl)-ethylamine
    Figure US20120053345A1-20120301-C00230
    B.3.77 1.54(e) 388.0 (M + H)+
    Pyridin-2-yl- methylamine
    Figure US20120053345A1-20120301-C00231
    B.3.78 1.80(e) 384.0 (M + H)+
    Dimethyl-(R)- pyrrolidin-3-yl- amine
    Figure US20120053345A1-20120301-C00232
    B.3.79 1.89(e) 389.0 (M − H)
    Piperidine-3- carboxamide
    Figure US20120053345A1-20120301-C00233
    B.3.80 1.44(e) 404.9 (M + H)+
  • TABLE B.4
    Examples prepared using general procedure C from Example #N.2.8
    Figure US20120053345A1-20120301-C00234
    Example Rt/min m/z
    Amine Precursor Product # (Method) (ESI+)
    Phenylamine
    Figure US20120053345A1-20120301-C00235
    B.4.1 1.92(e) 346 (M + H)+
    Amino-acetic acid methyl ester
    Figure US20120053345A1-20120301-C00236
    B.4.2 2.63(e) 341 (M + H)+
    Benzylamine
    Figure US20120053345A1-20120301-C00237
    B.4.3 1.48(e) 360 (M + H)+
    Tetrahydro- pyran-4- ylamine
    Figure US20120053345A1-20120301-C00238
    B.4.4 0.72(e) 354 (M + H)+
    N,N,N′- Trimethyl- propane-1,3- diamine
    Figure US20120053345A1-20120301-C00239
    B.4.5 0.63(e) 369.7 (M + H)+
    2-Amino- ethanol
    Figure US20120053345A1-20120301-C00240
    B.4.6 0.53(e) 314 (M + H)+
    4-Fluoro- benzylamine
    Figure US20120053345A1-20120301-C00241
    B.4.7 1.57(e) 378 (M + H)+
    N,N-Dimethyl- benzene-1,4- diamine
    Figure US20120053345A1-20120301-C00242
    B.4.8 1.58(e) 389 (M + H)+
    4-Methoxy- benzylamine
    Figure US20120053345A1-20120301-C00243
    B.4.9 1.52(e) 390 (M + H)+
    C-Benzo[b] thiophen-3-yl- methylamine
    Figure US20120053345A1-20120301-C00244
    B.4.10 1.78(e) 416 (M + H)+
    4-Fluoro- phenylamine
    Figure US20120053345A1-20120301-C00245
    B.4.11 1.65(e) 364 (M + H)+
    3-Amino- propan-1-ol
    Figure US20120053345A1-20120301-C00246
    B.4.12 0.57(e) 328 (M + H)+
    4-Amino- phenol
    Figure US20120053345A1-20120301-C00247
    B.4.13 0.95 (e) 362 (M + H)+
    4-Chloro- phenylamine
    Figure US20120053345A1-20120301-C00248
    B.4.14 1.82(e) 380 (M + H)+
    p-Tolylamine
    Figure US20120053345A1-20120301-C00249
    B.4.15 1.72(e) 360 (M + H)+
    (4-Amino- phenyl)- methanol
    Figure US20120053345A1-20120301-C00250
    B.4.16 0.80(e) 375 (M + H)+
    1H- Benzoimidazol- 5-ylamine
    Figure US20120053345A1-20120301-C00251
    B.4.17 0.72(e) 385.7 (M + H)+
    Biphenyl-4- ylamine
    Figure US20120053345A1-20120301-C00252
    B.4.18 2.03(e) 422 (M + H)+
    Naphthalen-2- ylamine
    Figure US20120053345A1-20120301-C00253
    B.4.19 1.87(e) 396 (M + H)+
    4-Methoxy- naphthalen-2- ylamine
    Figure US20120053345A1-20120301-C00254
    B.4.20 1.98(e) 426 (M + H)+
    m-Tolylamine
    Figure US20120053345A1-20120301-C00255
    B.4.21 1.70(e) 360.4 (M + H)+
    1H-Benzotriazol- 5-ylamine
    Figure US20120053345A1-20120301-C00256
    B.4.22 0.75(e) 387 (M + H)+
    1H-Indol-6- ylamine
    Figure US20120053345A1-20120301-C00257
    B.4.23 1.42(e) 385 (M + H)+
    3-Amino-phenol
    Figure US20120053345A1-20120301-C00258
    B.4.24 1.23(e) 362 (M + H)+
    5-Amino-1,3- dihydro- benzoimidazol- 2-one
    Figure US20120053345A1-20120301-C00259
    B.4.25 0.67(e) 402 (M + H)+
    2-(4-Amino- phenyl)-ethanol
    Figure US20120053345A1-20120301-C00260
    B.4.26 1.15(e) 389.9 (M + H)+
    4-Amino-2- fluoro-phenol
    Figure US20120053345A1-20120301-C00261
    B.4.27 1.22(e) 380 (M + H)+
    2-Amino-phenol
    Figure US20120053345A1-20120301-C00262
    B.4.28 1.52(e) 362 (M + H)+

    General Procedure C: Formation of an Amide from Carboxylic Acid and Amine Using Si-DCT
  • Figure US20120053345A1-20120301-C00263
  • In a 20 mL vial, a solution of N-methylmorpholine (2-5 equivalents, preferably 4 equivalents) in an organic solvent (for example, THF, CH2Cl2, CH3CN, or CH3CN/CH2Cl2 1:1) is added to Si-dichlorotriazine (Si-DCT) (2-4 equivalents, preferably 3 equivalents). A solution of the carboxylic acid (1.1-2.5 equivalents, preferably 1.25 equivalents) in an organic solvent (for example, THF, CH2Cl2, CH3CN, or 1:1 CH3CN/CH2Cl2, preferably 1:1 CH3CN/CH2Cl2) is added and mixed for about 1 min. A solution of the amine (1 equivalent) in an organic solvent (for example, THF, CH2Cl2, CH3CN, or 1:1 CH3CN/CH2Cl2, preferably 1:1 CH3CN/CH2Cl2) is then added. The reaction is shaken at about 10-60° C., preferably about ambient temperature, for about 10-24 hours, preferably about 16 hours. The crude reaction solution is mixed with DMF, eluted through a Si-carbonate (1 g, 6 mL) cartridge with additional organic solvent (for example, MeOH), and concentrated in vacuo. The crude product can then be further purified by crystallization or chromatography.
  • Illustration of General Procedure C Example #13 Furan-2-carboxylic acid (7-benzo[b]thiophen-2-yl-1H-indazol-5-yl)-amide
  • Figure US20120053345A1-20120301-C00264
  • In a 20 mL vial, a solution of N-methylmorpholine (0.060 g, 0.60 mmol) in MeCN/CH2Cl2 (1:1, 0.688 mL) was added to Si-dichlorotriazine (SiliCycle, Inc; 0.60 mmol/g, 0.74 g, 0.44 mmol). Then, a solution of 2-furoic acid (0.021 g, 0.19 mmol) in MeCN (0.925 mL) was added and mixed for about 1 min., prior to the addition of a solution of 7-benzo[b]thiophen-2-yl-1H-indazol-5-ylamine (Example #F.8.1, 0.039 g, 0.15 mmol) in MeCN/CH2Cl2 (1:1, 0.688 mL). The reaction mixture was shaken at ambient temperature for about 16 hours. The crude reaction solution was mixed with DMF (5 mL) to aid in solubility, eluted through a Si-carbonate (SiliCycle, Inc; 1 g, 6 mL) cartridge using MeOH (approx 3 mL), and concentrated in vacuo. The crude product was then purified by preparative HPLC (Waters Symmetry C8 column (25×100 mm, 7 μm particle size) using a gradient of 10%-100% CH3CN/0.1% aqueous TFA over 8 min (10 min run time) at a flow rate of 40 mL/min) to afford furan-2-carboxylic acid (7-benzo[b]thiophen-2-yl-1H-indazol-5-yl)-amide (0.0025 g, 4.7%); RP-HPLC (Table 1, Method l) Rt 2.51 min; m/z: (M+H)+ 360.
  • TABLE C.1
    Examples prepared using general procedure C using Example #F.8.1
    Figure US20120053345A1-20120301-C00265
    HPLC Rt
    Example (min)
    Acid Product # (Method) m/z
    Tetrahydro-3- furoic acid
    Figure US20120053345A1-20120301-C00266
    C.1.1 2.31 (1) 364 (M + H)+
    2-Methoxy phenylacetic acid
    Figure US20120053345A1-20120301-C00267
    C.1.2 2.65 (1) 414 (M + H)+
    3-Methoxy phenylacetic acid
    Figure US20120053345A1-20120301-C00268
    C.1.3 2.62 (1) 414 (M + H)+
    4-Methoxy phenylacetic acid
    Figure US20120053345A1-20120301-C00269
    C.1.4 2.61 (1) 414 (M + H)+
    3- (Trifluoromethoxy) phenylacetic acid
    Figure US20120053345A1-20120301-C00270
    C.1.5 2.85 (1) 468 (M + H)+
    3,4- (Methylenedioxy) phenylacetic acid
    Figure US20120053345A1-20120301-C00271
    C.1.6 2.59 (1) 428 (M + H)+
    3- Ethoxypropionic acid
    Figure US20120053345A1-20120301-C00272
    C.1.7 2.43 (1) 366 (M + H)+
    L-Pyroglutamic acid
    Figure US20120053345A1-20120301-C00273
    C.1.8 2.07 (1) 377 (M + H)+
    D-Pyroglutamic acid
    Figure US20120053345A1-20120301-C00274
    C.1.9 2.07 (1) 377 (M + H)+
    1- (Aminocarbonyl)- 1-cyclopropane carboxylic acid
    Figure US20120053345A1-20120301-C00275
    C.1.10 2.24 (1) 377 (M + H)+
    Benzyloxyacetic acid
    Figure US20120053345A1-20120301-C00276
    C.1.11 2.72 (1) 414 (M + H)+
    4-Methoxy- cyclohexane carboxylic acid
    Figure US20120053345A1-20120301-C00277
    C.1.12 2.53 (1) 406 (M + H)+
    1-Phenyl-1- cyclopropane carboxylic acid
    Figure US20120053345A1-20120301-C00278
    C.1.13 2.85 (1) 410 (M + H)+
    (S)-(+)-2- Phenylbutyric acid
    Figure US20120053345A1-20120301-C00279
    C.1.14 2.83 (1) 412 (M + H)+
    4-Phenylbutyric acid
    Figure US20120053345A1-20120301-C00280
    C.1.15 2.78 (1) 412 (M + H)+
    N-(2-Furoyl)glycine
    Figure US20120053345A1-20120301-C00281
    C.1.16 2.63 (1) 376 (M + H)+
    4-(2- Thienyl)butyric acid
    Figure US20120053345A1-20120301-C00282
    C.1.17 2.75 (1) 418 (M + H)+
    1- Acetylpiperidine-4- carboxylic acid
    Figure US20120053345A1-20120301-C00283
    C.1.18 2.21 (1) 419 (M + H)+
    N-(N,N-Di- propyl)-L-alanine
    Figure US20120053345A1-20120301-C00284
    C.1.19 2.92 (1) 421 (M + H)+
    3- Benzoylpropionic acid
    Figure US20120053345A1-20120301-C00285
    C.1.20 2.62 (1) 426 (M + H)+
    Hippuric acid
    Figure US20120053345A1-20120301-C00286
    C.1.21 1.86 (m) 427.11 (M + H)+
    3-(3- Methoxyphenyl) propionic acid
    Figure US20120053345A1-20120301-C00287
    C.1.22 2.30 (1) 360 (M + H)+
    3- Methoxyphenoxy acetic acid
    Figure US20120053345A1-20120301-C00288
    C.1.23 2.69 (1) 430 (M + H)+
    4-Oxo-4- (2-thienyl) butyric acid
    Figure US20120053345A1-20120301-C00289
    C.1.24 2.56 (1) 432 (M + H)+
    2-(4- Methylpyrimidin- 2-ylthio) acetic acid
    Figure US20120053345A1-20120301-C00290
    C.1.25 2.48 (1) 432 (M + H)+
    Glutaranilic acid
    Figure US20120053345A1-20120301-C00291
    C.1.26 2.48 (1) 455 (M + H)+
    4-Methylsulfonyl phenylacetic acid
    Figure US20120053345A1-20120301-C00292
    C.1.27 2.40 (1) 462 (M + H)+
    N-p-Tosylglycine
    Figure US20120053345A1-20120301-C00293
    C.1.28 2.52 (1) 477 (M + H)+
    4-Acetyl benzoic acid
    Figure US20120053345A1-20120301-C00294
    C.1.29 2.56 (1) 412 (M + H)+
    4-(Methylthio) benzoic acid
    Figure US20120053345A1-20120301-C00295
    C.1.30 2.74 (1) 416 (M + H)+
    3-Fluoro-4- methoxy benzoic acid
    Figure US20120053345A1-20120301-C00296
    C.1.31 2.67 (1) 418 (M + H)+
    2-Acetamido benzoic acid
    Figure US20120053345A1-20120301-C00297
    C.1.32 2.55 (1) 425 (M − H)
    3-Acetamido benzoic acid
    Figure US20120053345A1-20120301-C00298
    C.1.33 2.35 (1) 427 (M + H)+
    4-Diethylamino benzoic acid
    Figure US20120053345A1-20120301-C00299
    C.1.34 2.85 (1) 441 (M + H)+
    N- Phenylanthranilic acid
    Figure US20120053345A1-20120301-C00300
    C.1.35 3.02 (1) 461 (M + H)+
    3-Furoic acid
    Figure US20120053345A1-20120301-C00301
    C.1.36 2.52 (1) 360 (M + H)+
    Thiophene-2- carboxylic acid
    Figure US20120053345A1-20120301-C00302
    C.1.37 2.63 (1) 374 (M + H)+
    3-Methyl-2- thiophene carboxylic acid
    Figure US20120053345A1-20120301-C00303
    C.1.38 2.70 (1) 390 (M + H)+
    Pyrrole-2- carboxylic acid
    Figure US20120053345A1-20120301-C00304
    C.1.39 2.49 (1) 359 (M + H)+
    Thiazole-4- carboxylic acid
    Figure US20120053345A1-20120301-C00305
    C.1.40 2.53 (1) 377 (M + H)+
    Thiazole-5- carboxylic acid
    Figure US20120053345A1-20120301-C00306
    C.1.41 2.43 (1) 375 (M − H)
    1H-Pyrazole-5- carboxylic acid
    Figure US20120053345A1-20120301-C00307
    C.1.42 2.30 (1) 360 (M + H)+
    Isoxazole-5- carboxylic acid
    Figure US20120053345A1-20120301-C00308
    C.1.43 2.46 (1) 361 (M + H)+
    3,5-Dimethyl isoxazole-4- carboxylic acid
    Figure US20120053345A1-20120301-C00309
    C.1.44 2.51 (1) 389 (M + H)+
    5-Methyl-3- phenylisoxazole- 4-carboxylic acid
    Figure US20120053345A1-20120301-C00310
    C.1.45 2.72 (1) 451 (M + H)+
    Picolinic acid
    Figure US20120053345A1-20120301-C00311
    C.1.46 2.68 (1) 371 (M + H)+
    2- Hydroxynicotinic acid
    Figure US20120053345A1-20120301-C00312
    C.1.47 1.73 (m) 387.51 (M + H)+
    6- Hydroxynicotinic acid
    Figure US20120053345A1-20120301-C00313
    C.1.48 2.12 (1) 387 (M + H)+
    2-Pyridylacetic acid
    Figure US20120053345A1-20120301-C00314
    C.1.49 2.37 (1) 385 (M + H)+
    3-Pyridylacetic acid
    Figure US20120053345A1-20120301-C00315
    C.1.50 2.32 (1) 385 (M + H)+
    Pyrimidine-4- carboxylic acid
    Figure US20120053345A1-20120301-C00316
    C.1.51 1.88 (m) 372.0 (M + H)+
    2- Methylpyrazine- 5-carboxylic acid
    Figure US20120053345A1-20120301-C00317
    C.1.52 2.58 (1) 386 (M + H)+
    Indole-3- carboxylic acid
    Figure US20120053345A1-20120301-C00318
    C.1.53 2.05 (m) 409.1 (M + H)+
    5-Methyl-1- phenylpyrazole-4- carboxylic acid
    Figure US20120053345A1-20120301-C00319
    C.1.54 2.68 (1) 450 (M + H)+
    4-Oxo-4,5,6,7- tetrahydrobenzo[b] furan-3- carboxylic acid
    Figure US20120053345A1-20120301-C00320
    C.1.55 2.76 (1) 428 (M + H)+
    6-Chloro-2H-1- benzopyran-3- carboxylic acid
    Figure US20120053345A1-20120301-C00321
    C.1.56 2.93 (1) 458 (M + H)+
    3- (Dimethylamino) propanoic acid
    Figure US20120053345A1-20120301-C00322
    C.1.57 2.05 (1) 365 (M + H)+
    1-Pyrrolidine propanoic acid
    Figure US20120053345A1-20120301-C00323
    C.1.58 2.12 (1) 391 (M + H)+
    1-Piperidine propionic acid
    Figure US20120053345A1-20120301-C00324
    C.1.59 2.18 (1) 405 (M + H)+
    4- Morpholinoacetic acid
    Figure US20120053345A1-20120301-C00325
    C.1.60 2.34 (1) 393 (M + H)+
    5-(5-Amino-1H- pyrrolo[2,3- c]pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00326
    C.1.61 1.894 (k)  373 (M + H)+
    5-(5-Amino-1H- pyrrolo[2,3- c]pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00327
    C.1.62 1.628 (k)  384 (M + H)+
    5-(5-Amino-1H- pyrrolo[2,3- c]pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00328
    C.1.63 1.536 (k)  384 (M + H)+
    5-(5-Amino-1H- pyrrolo[2,3- c]pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00329
    C.1.64 1.487 (k)  384 (M + H)+
    5-(5-Amino-1H- pyrrolo[2,3- c]pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00330
    C.1.65 1.301 (k)  364 (M + H)+
    5-(5-Amino-1H- pyrrolo[2,3-c] pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00331
    C.1.66 1.442 (k)  390 (M + H)+
    5-(5-Amino-1H- pyrrolo[2,3-c] pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00332
    C.1.67 1.461 (k)  404 (M + H)+
    5-(5-Amino-1H- pyrrolo[2,3- c]pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00333
    C.1.68 2.093 (k)  387 (M + H)+
    5-(5-Amino-1H- pyrrolo[2,3-c] pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00334
    C.1.69 2.201 (k)  436 (M + H)+
    5-(5-Amino-1H- pyrrolo[2,3- c]pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00335
    C.1.70 1.624 (k)  377 (M + H)+
    5-(5-Amino-1H- pyrrolo[2,3- c]pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00336
    C.1.71 1.628 (k)  377 (M + H)+
    5-(5-Amino-1H- pyrrolo[2,3- c]pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00337
    C.1.72 1.819 (k)  347 (M + H)+
    5-(5-Amino-1H- pyrrolo[2,3- c]pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00338
    C.1.73 2.007 (k)  361 (M + H)+
    5-(5-Amino-1H- pyrrolo[2,3- c]pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00339
    C.1.74 2.436 (k)  389 (M + H)+
    5-(5-Amino-1H- pyrrolo[2,3- c]pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00340
    C.1.75 1.652 (k)  361 (M + H)+
    5-(5-Amino-1H- pyrrolo[2,3- c]pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00341
    C.1.76 1.391 (k)  363 (M + H)+
    5-(5-Amino-1H- pyrrolo[2,3- c]pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00342
    C.1.77 2.101 (k)  383 (M + H)+
    5-(5-Amino-1H- pyrrolo[2,3- c]pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00343
    C.1.78 2.021 (k)  383 (M + H)+
    5-(5-Amino-1H- pyrrolo[2,3- c]pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00344
    C.1.79 2.328 (k)  399 (M + H)+
    5-(5-Amino-1H- pyrrolo[2,3- c]pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00345
    C.1.80 2.129 (k)  399 (M + H)+
    5-(5-Amino-1H- pyrrolo[2,3- c]pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00346
    C.1.81 2.555 (k)  403 (M + H)+
    5-(5-Amino-1H- pyrrolo[2,3- c]pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00347
    C.1.82 2.563 (k)  403 (M + H)+
    5-(5-Amino-1H- pyrrolo[2,3- c]pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00348
    C.1.83 2.845 (k)  461 (M + H)+
    5-(5-Amino-1H- pyrrolo[2,3- c]pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00349
    C.1.84 2.179 (k)  412 (M + H)+
    5-(5-Amino-1H- pyrrolo[2,3- c]pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00350
    C.1.85 2.142 (k)  413 (M + H)+
    5-(5-Amino-1H- pyrrolo[2,3- c]pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00351
    C.1.86 2.096 (k)  413 (M + H)+
    5-(5-Amino-1H- pyrrolo[2,3- c]pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00352
    C.1.87 2.107 (k)  413 (M + H)+
    5-(5-Amino-1H- pyrrolo[2,3- c]pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00353
    C.1.88 2.338 (k)  417 (M + H)+
    5-(5-Amino-1H- pyrrolo[2,3- c]pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00354
    C.1.89 2.37 (k) 417 (M + H)+
    5-(5-Amino-1H- pyrrolo[2,3- c]pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00355
    C.1.90 2.31 (k) 417 (M + H)+
    5-(5-Amino-1H- pyrrolo[2,3- c]pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00356
    C.1.91 2.446 (k)  451 (M + H)+
    5-(5-Amino-1H- pyrrolo[2,3- c]pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00357
    C.1.92 1.74 (k) 413 (M + H)+
    5-(5-Amino-1H- pyrrolo[2,3- c]pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00358
    C.1.93 2.232 (k)  427 (M + H)+
    5-(5-Amino-1H- pyrrolo[2,3- c]pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00359
    C.1.94 2.161 (k)  417 (M + H)+
    5-(5-Amino-1H- pyrrolo[2,3- c]pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00360
    C.1.95 1.349 (k)  406 (M + H)+
    5-(5-Amino-1H- pyrrolo[2,3- c]pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00361
    C.1.96 2.26 (k) 399 (M + H)+
    5-(5-Amino-1H- pyrrolo[2,3- c]pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00362
    C.1.97 1.994 (k)  389 (M + H)+
    5-(5-Amino-1H- pyrrolo[2,3- c]pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00363
    C.1.98 2.074 389 (M + H)+
    5-(5-Amino-1H- pyrrolo[2,3- c]pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00364
    C.1.99 2.99 (e) 426 (M + H)+
    5-(5-Amino-1H- pyrrolo[2,3- c]pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00365
    C.1.100 3.09 (e) 468 (M + H)+
    5-(5-Amino-1H- pyrrolo[2,3- c]pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00366
    C.1.101 3.28 (e) 452 (M + H)+
    5-(5-Amino-1H- pyrrolo[2,3- c]pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00367
    C.1.102 3.22 (e) 481 (M + H)+
    5-(5-Amino-1H- pyrrolo[2,3- c]pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00368
    C.1.103 0.96 (e) 381
    5-(5-Amino-1H- pyrrolo[2,3- c]pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00369
    C.1.104 1.09 (e) 395.2
    5-(5-Amino-1H- pyrrolo[2,3- c]pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00370
    C.1.105 0.77 (e) 396.2
    5-(5-Amino-1H- pyrrolo[2,3- c]pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00371
    C.1.106 0.85 (e) 399.2
    5-(5-Amino-1H- pyrrolo[2,3- c]pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00372
    C.1.107 1.17 (e) 408.2
    5-(5-Amino-1H- pyrrolo[2,3- c]pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00373
    C.1.108 1.29 (e) 466
    5-(5-Amino-1H- pyrrolo[2,3- c]pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00374
    C.1.109 0.78 (e) 462.1
    5-(5-Amino-1H- pyrrolo[2,3- c]pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00375
    C.1.110 1.30 (e) 467.1
    5-(5-Amino-1H- pyrrolo[2,3- c]pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00376
    C.1.111 1.29 (e) 426
    5-(5-Amino-1H- pyrrolo[2,3- c]pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00377
    C.1.112 0.45 (e) 398.1
    5-(5-Amino-1H- pyrrolo[2,3- c]pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00378
    C.1.113 0.61 (e) 467.2
    5-(5-Amino-1H- pyrrolo[2,3- c]pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00379
    C.1.114 1.19 (e) 397.3
    5-(2-Amino-5H- pyrrolo[3,2- d]pyrimidin-7-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00380
    C.1.115 1.33 (e) 382
    5-(2-Amino-5H- pyrrolo[3,2- d]pyrimidin-7-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00381
    C.1.116 1.30 (e) 370.1
    5-(5-Amino-1H- pyrrolo[2,3- c]pyridin-3-yl)- 1H-indazole-7- carboxylic acid
    Figure US20120053345A1-20120301-C00382
    C.1.117 1.15 (e) 408.2

    General Procedure D: Protection of an Indazole with a trimethyl-silanylethoxymethyl Group
  • Figure US20120053345A1-20120301-C00383
  • A mixture of an indazole (1 equivalent) and a base (for example, Na2CO3, NaOH, Cs2CO3 or t-BuOK, preferably Na2CO3) (1-10 equivalents, preferably 1-2 equivalents) and SEMCl (1-2 equivalents, preferably 1.2 equivalents) in a solvent (for example, DME or CH2Cl2, preferably CH2Cl2) mixed with either water or in an anhydrous solvent, (for example, DMF or DMA, preferably DMF) is stirred at about 10-40° C. (preferably about 20-25° C.) for about 0.5-24 hours (preferably about 1-2 hours) under an inert atmosphere. Saturated aqueous NH4Cl is added and the solvents are removed under reduced pressure. The residue is dissolved in an organic solvent (CH2Cl2 or EtOAc, preferably EtOAc) and washed with water. The organic layer is dried over a desiccant (for example, magnesium sulfate or sodium sulfate, preferably magnesium sulfate) and further purified by crystallization or chromatography.
  • Illustration of General Procedure D Preparation #23 7-Benzo[b]thiophen-2-yl-5-bromo-1-(2-trimethylsilanyl-ethoxymethyl)-1H-indazole, and Preparation #24 7-Benzo[b]thiophen-2-yl-5-bromo-2-(2-trimethylsilanyl-ethoxymethyl)-2H-indazole
  • Figure US20120053345A1-20120301-C00384
  • To a mixture of 7-benzo[b]thiophen-2-yl-5-bromo-1H-indazole (Preparation #26, 38.0 g, 0.116 mol) in DMF (570 mL) at about 5° C. was added t-BuOK (15.5 g, 0.139 mmol). The mixture was stirred for about 30 minutes then SEM-Cl (23.2 g, 0.139 mmol) was added over the course of about 5 minutes while maintaining the temperature between about 0-5° C. After warming to ambient temperature and stirring the solution for about 30 minutes the solution was treated with saturated aqueous NH4Cl (approximately 5 mL). The solvents were removed under reduced pressure and the residue was partitioned between EtOAc:water (1:1, 700 mL). The organic layer was further washed with water (150 mL) and brine (150 mL), dried over anhydrous MgSO4, filtered and concentrated to give an oil that solidified upon standing. Heptane (500 mL) was added and the mixture was heated to about 100° C. to dissolve all of the solids. The mixture was cooled to ambient temperature and the resulting solids were collected by filtration to afford 7-benzo[b]thiophen-2-yl-5-bromo-2-(2-trimethylsilanyl-ethoxymethyl)-2H-indazole (33 g, 62%); (DMSO-d6, 400 MHz) δ 8.73 (s, 1H), 8.61 (s, 1H), 8.11 (d, 1H), 8.04 (m, 1H), 7.95 (m, 1H), 7.79 (d, 1H), 7.45 (m, 2H), 5.89 (s, 2H), 3.79 (m, 2H), 0.96 (m, 2H), 0.00 (s, 9H); RP-HPLC (Table 1, Method e) Rt 3.63 min; m/z: (M+H)+. 461. The filtrate was concentrated and purified by flash chromatography over silica gel using heptane/EtOAc (9:1) as an eluent to give 7-benzo[b]thiophen-2-yl-5-bromo-1-(2-trimethylsilanyl-ethoxymethyl)-1H-indazole 7.8 g (15%) as an oil which solidified upon standing; (DMSO-d6, 400 MHz) g 8.50 (s, 1H), 8.40 (s, 1H), 8.26 (m, 1H), 8.12 (m, 1H), 7.89 (s, 1H), 7.80 (d, 1H), 7.67 (m, 2H), 5.64 (s, 2H), 3.34 (m, 2H), 0.81 (m, 2H), 0.00 (s, 9H); RP-HPLC (Table 1, Method e) Rt 3.63 min; m/z: (M+H)+. 461.
  • General Procedure E: Conversion of an Aryl Halide to a Boronic Acid or Boronate
  • Figure US20120053345A1-20120301-C00385
  • To a mixture of a boronating reagent (bis(pinacolato)diboron or pinacolatoborane, preferably bis(pinacolato)diboron)(1-1.5 equivalents, preferably 1.3 equivalents), an aryl halide (for example, an aryl bromide or an aryl iodide, preferably an aryl iodide) (0.5-3 equivalents, preferably 1 equivalent), a palladium catalyst (for example tris(benzylideneacetone)dipalladium (0), tetrakis(triphenylphosphine)palladium(0), bis(acetato)triphenylphosphinepalladium(II) (˜5% Pd) polymer-bound FibreCat™ or [1,1′-bis(diphenylphosphino)ferrocene] dichloropalladium(II), complex with dichloromethane) (preferably dichloro[1,1′-bis(diphenylphosphino)ferrocene]-palladium (II) dichloromethane adduct) (0.03-0.15 equivalent, preferably 0.10 equivalents) and a base (for example, NaOAc or KOAc, preferably KOAc) (1.5-3.0 equivalents, preferably 2.5 equivalents) is added an organic solvent (for example, DMF, dioxane, or THF, preferably DMF). The mixture is heated at about 50-100° C. (preferably about 80° C.) for about 1-24 hours (preferably about 15 hours) under an inert atmosphere. The mixture is allowed to cool to ambient temperature, and the solvent is removed under reduced pressure. The residue can then be further purified by chromatography or crystallization.
  • Illustration of General Procedure E Preparation #25 7-Benzo[b]thiophen-2-yl-5-(4,4,5,5-tetramethyl-[1,3,2]dioxaborolan-2-yl)-1H-indazole
  • Figure US20120053345A1-20120301-C00386
  • A mixture of 7-benzo[b]thiophen-2-yl-5-bromo-1H-indazole (Preparation #26, 5.0 g, 15.2 mmol), bis(pinacolato)diboron (5.78, 22.8 mmol), KOAc (3.72 g, 38 mmol) and [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium(II), complex with dichloromethane (1:1) (0.99 g, 1.22 mmol) in DMF (125 mL) was heated at about 100° C. under an atmosphere of nitrogen for about 18 hours. The dark reaction solution was cooled to ambient temperature, diluted with CH2Cl2 (25 mL) then washed with water (2×20 mL). The reaction mixture was cooled, concentrated under reduced pressure, triturated with CH2Cl2 (175 mL), filtered and the filtrate concentrated under reduced pressure. The resulting material was purified by flash chromatography over silica gel using CH2Cl2/EtOAc (97:3) as the eluent and the material was triturated with heptane (25 mL) to give 7-benzo[b]thiophen-2-yl-5-(4,4,5,5-tetramethyl-[1,3,2]dioxaborolan-2-yl)-1H-indazole as a white solid (2.23 g, 39%); (DMSO-d6, 400 MHz) δ 13.5 (s, 1H), 8.31 (s, 1H), 8.22 (s, 1H), 8.06 (s, 1H), 8.02 (d, 1H), 7.89 (d, 1H), 7.83 (s, 1H), 7.43 (m, 2H), 1.35 (s, 12H); RP-HPLC (Table 1, Method e) Rt 2.77 min; m/z (M−H) 374.5.
  • General Procedure F: Suzuki Coupling of a Boronate or Boronic Acid with an Aryl Halide Substrate
  • Figure US20120053345A1-20120301-C00387
  • To a mixture of a boronate ester or a boronic acid (1-5 equivalents, preferably 2 equivalents), an aryl halide (for example, an aryl bromide, aryl chloride or an aryl iodide, preferably an aryl iodide) (0.7-3 equivalents, preferably 1 equivalent) and an inorganic base (for example, KF, Na2CO3 or Cs2CO3, preferably Cs2CO3) (2-16 equivalents, preferably 2.5 equivalents) in a degassed organic solvent (for example THF, DME, DMF, 1,4-dioxane, DME/water or toluene, preferably DMF or DME/water) is added a palladium catalyst (for example tris(benzylideneacetone)dipalladium (0), tetrakis(triphenylphosphine)palladium(0), bis(acetato)triphenylphosphinepalladium(II) (−5% Pd) polymer-bound FibreCat™ or [1,1′-bis(diphenylphosphino)ferrocene] dichloropalladium(II), complex with dichloromethane, preferably tetrakis(triphenylphosphine)palladium(0)) (0.01-0.10 equivalents, preferably 0.05 equivalents). If necessary, tributylphosphinetetrafluoroborate (0.01 to 0.20 equivalents, preferably 0.05 equivalents) is also added. The reaction mixture is heated at about 40-150° C. (preferably about 80° C.) for about 2-24 hours (preferably about 18 hours) or at about 100-200° C. (preferably 150° C.) for about 5-60 minutes (preferably about 15 minutes) in a microwave under an inert atmosphere. The reaction mixture is allowed to cool to ambient temperature. Subsequently, the solvents are removed under reduced pressure and the residue is suspended in a mixture of EtOAc and water; the mixture is stirred for 30 minutes and the resulting solid is collected by filtration; the product can be further purified by chromatography or crystallization. Alternatively, the cooled reaction mixture is diluted with water or an aqueous basic solution (such as saturated aqueous NaHCO3) and extracted (1-5 times, preferably 3 times) with a suitable solvent (such as EtOAc or CH2Cl2) then the combined organic extracts are dried (for example, over Na2SO4 or MgSO4), decanted or filtered, and concentrated under reduced pressure to afford the product that can be further purified by chromatography or crystallization. If a tert-butoxycarbonyl (Boc) protected amine is used, then the material is subsequently suspended in a mixture of methanol/6 N HCl and heated to about 65° C. for about one hour then cooled, concentrated and purified by chromatography or crystallization.
  • Illustrations of General Procedure F Example #13a 7-Benzo[b]thiophen-2-yl-5-bromo-1H-indazole
  • Figure US20120053345A1-20120301-C00388
  • A mixture of 5-bromo-7-iodo-1H-indazole (Preparation #22a, 30.0 g, 92.9 mmol) and thianapthene-2-boronic acid (21.5 g, 120.7 mmol), DME (480 mL), water (48 mL), Na2CO3 (29.5 g, 279 mmol) and tetrakis triphenylphosphine palladium (0) (8.6 g, 7.43 mmol) was heated at about 90° C. in an oil bath under an atmosphere of nitrogen for about 15 hours. The solvent was removed under reduced pressure and the residue was suspended in a mixture of ethyl acetate (600 mL) and water (300 mL). The mixture was stirred for about 30 minutes and the resulting solid was collected by filtration and dried to yield 7-Benzo[b]thiophen-2-yl-5-bromo-1H-indazole (21.4 g, 70%); (DMSO-d6, 400 MHz) δ 13.63 (s, 1H), 8.24 (s, 1H), 8.05-8.09 (m, 3H), 7.92 (d, 1H), 7.65 (s, 1H), 7.16 (m, 2H); RP-HPLC (Table 1, Method e) Rt=2.69 min; m/z: (M−H) 328.4.
  • Example #13c N-Phenyl-5-pyridin-3-yl-1H-indazole-7-carboxamide
  • Figure US20120053345A1-20120301-C00389
  • A mixture of N-phenyl-5-bromo-JH-indazole-7-carboxamide (Example #B.1.2, 0.05 g, 0.16 mmol) and pyridine-3-yl-boronic acid (0.11 g, 0.8 mmol), 1,2-dimethoxyethane (1.3 mL), water (0.7 mL), Cs2CO3 (0.16 g, 0.48 mmol) and [1,1′-bis(diphenylphosphino)ferrocene] dichloropalladium(II), complex with dichloromethane (0.013 g, 0.016 mmol) was heated at about 150° C. in the microwave under an atmosphere of nitrogen for about 15 minutes. The crude product was filtered and the solvent was removed under reduced pressure. The residue was dissolved in DMSO then purified by reverse phase HPLC (Waters Symmetry C8 column (25×100 mm, 7 μm particle size) using a gradient of 10%-100% CH3CN/0.1% aqueous TFA over 8 min (10 min run time) at a flow rate of 40 mL/min) to yield N-phenyl-5-pyridin-3-yl-1H-indazole-7-carboxamide (0.014 g, 6%); (DMSO-d6, 400 MHz) δ 13.4 (bs, 1H), 10.5 (bs, 1H), 9.12 (d, 1H), 8.62 (d, 1H), 8.48 (s, 1H), 8.34 (s, 1H), 8.28 (m, 2H), 7.84 (d, 2H), 7.56 (m, 1H), 7.42 (m, 2H), 7.16 (m, 2H); RP-HPLC (Table 1, Method e) Rt=1.81 min; m/z: (M+H)+ 315.3.
  • TABLE F.1
    Examples prepared using general procedure F from Preparation #25
    Figure US20120053345A1-20120301-C00390
    HPLC Rt
    Example (min)
    Aryl Halide Product # (method) m/z
    2-Chloro-4-iodo- pyridine
    Figure US20120053345A1-20120301-C00391
    F.1.1 2.63 (e) 359.5 (M − H)
    5-Bromo- nicotinotrile
    Figure US20120053345A1-20120301-C00392
    F.1.2 2.28 (e) 350.8 (M − H)
    5-Bromo- nicotinamide
    Figure US20120053345A1-20120301-C00393
    F.1.3 1.70 (e) 368.7 (M − H)
    (5-Bromo-pyridin- 2-yl)-methanol
    Figure US20120053345A1-20120301-C00394
    F.1.4 1.93 (e) 355.8 (M − H)
    1-(5-Bromo- pyridin-2-yl)- ethanone
    Figure US20120053345A1-20120301-C00395
    F.1.5 2.44 (e) 368.1 (M − H)
    5-Bromo-nicotinic acid hydrazide
    Figure US20120053345A1-20120301-C00396
    F.1.6 1.55 (e) 384.0 (M − H)
    5-Bromo-pyridine- 2-carbonitrile
    Figure US20120053345A1-20120301-C00397
    F.1.7 2.39 (e) 351.1 (M − H)
    5-Bromo-pyridine- 2-carboxylic acid
    Figure US20120053345A1-20120301-C00398
    F.1.8 1.55 (e) 369.9 (M − H)
    5-Bromo-pyridine- 3-carboxaldehyde
    Figure US20120053345A1-20120301-C00399
    F.1.9 2.26 (e) 354.2 (M − H)
    (3-Bromo-phenyl)- urea
    Figure US20120053345A1-20120301-C00400
    F.1.10 1.94 (e) 382.7 (M − H)
    3-Bromo- thieno[3,2- c]pyridin-4-yl amine
    Figure US20120053345A1-20120301-C00401
    F.1.11 2.49 (e) 399.0 (M − H)
    3-Bromo-furo[3,2- c]pyridin-4-ylamine
    Figure US20120053345A1-20120301-C00402
    F.1.12 2.39 (e) 383.0 (M − H)
    Cis-{3-iodo-1-[4- (4-methyl- piperazin-1-yl)- cyclohexyl]-1H- pyrazolo[3,4-d] pyrimidin-4- ylamine}
    Figure US20120053345A1-20120301-C00403
    F.1.13 1.79 (e) 564.1 (M + H)+
    3-Iodo-pyridin-4- ylamine
    Figure US20120053345A1-20120301-C00404
    F.1.14 1.88 (e) 343.0 (M + H)+
    3-Iodo-pyridin-2- ylamine
    Figure US20120053345A1-20120301-C00405
    F.1.15 2.30 (e) 343.0 (M + H)+
    4-(3-Iodo-pyridin- 2-yl)-morpholine
    Figure US20120053345A1-20120301-C00406
    F.1.16 2.41 (e) 412.9 (M + H)+
    3-Iodo-1-methyl- 1H-pyrazolo[3,4- d]pyrimidin-4- ylamine
    Figure US20120053345A1-20120301-C00407
    F.1.17 1.96 (e) 398.0 (M + H)+
    3-Iodo-2- isopropoxy-pyridine
    Figure US20120053345A1-20120301-C00408
    F.1.18 2.79 (e) 383.7 (M + H)+
    3-Iodo-2-methoxy- pyridine
    Figure US20120053345A1-20120301-C00409
    F.1.19 2.56 (e) 357.9 (M + H)+
    [2-(6-Iodo-pyridin- 2-ylamino)-ethyl]- carbamic acid tert- butyl ester
    Figure US20120053345A1-20120301-C00410
    F.1.20 2.10 (e) 386.0 (M + H)+
    [2-(6-Iodo-pyridin- 2-yloxy)-ethyl]- carbamic acid tert- butyl ester
    Figure US20120053345A1-20120301-C00411
    F.1.21 2.02 (e) 387.0 (M + H)+
    Thiophene-2- carboxylic acid (5- bromo-4,6- dimethyl-pyrimidin- 2-yl)-amide
    Figure US20120053345A1-20120301-C00412
    F.1.22 2.02 (e) 481.9 (M + H)+
    N-(5-Bromo-3- methyl-pyridin-2- yl)-acetamide
    Figure US20120053345A1-20120301-C00413
    F.1.23 1.83 (e) 399.0 (M + H)+
    N-(2-Bromo- pyridin-3-yl)- acetamide
    Figure US20120053345A1-20120301-C00414
    F.1.24 1.70 (e) 385.0 (M + H)+
    5-Iodo-6-methyl- pyrimidin-4- ylamine
    Figure US20120053345A1-20120301-C00415
    F.1.25 2.16 (e) 358.0 (M + H)+
    N-(5-Iodo-6- methyl-pyridin-2- yl)-2,2- dimethyl- propionamide
    Figure US20120053345A1-20120301-C00416
    F.1.26 2.42 (e) 441.0 (M + H)+
    5-Bromo-4-methyl- pyridin-2-ylamine
    Figure US20120053345A1-20120301-C00417
    F.1.27 2.14 (e) 354.6 (M − H)
    3-Bromo-5-(1H- tetrazol-5-yl)- pyridine
    Figure US20120053345A1-20120301-C00418
    F.1.28 1.30 (e) 441.0 (M + H)+
    2-Bromo-pyridin-3- ylamine
    Figure US20120053345A1-20120301-C00419
    F.1.29 1.92 (e) 342.9 (M + H)+
    4-Bromo-6- trifluoromethyl-1H- benzoimidazole
    Figure US20120053345A1-20120301-C00420
    F.1.30 2.39 (e) 432.4 (M − H)
    1-(5-Bromo-2- chloro-pyridin-3- yl)-3-phenyl-urea
    Figure US20120053345A1-20120301-C00421
    F.1.31 2.56 (e) 493.9 (M − H)
    3-Iodo-indazole-1- carboxylic acid tert- butyl ester
    Figure US20120053345A1-20120301-C00422
    F.1.32 2.27 (e) 364.6 (M − H)
    2-Cyclopropyl methoxy-3-iodo- pyridine
    Figure US20120053345A1-20120301-C00423
    F.1.33 2.81 (e) 396.0 (M − H)
    6′-Bromo-3,4,5,6- tetrahydro-2H- [1,2′]bipyridinyl-3′- ylamine
    Figure US20120053345A1-20120301-C00424
    F.1.34 2.84 (e) 426 (M + H)+
    4-Bromo-quinolin- 3-ylamine
    Figure US20120053345A1-20120301-C00425
    F.1.35 2.44 (e) 393 (M + H)+
    4-(3-Iodo-pyridin- 2-yl)-piperazine-1- carboxylic acid tert- butyl ester
    Figure US20120053345A1-20120301-C00426
    F.1.36 2.29 (e) 412 (M + H)+
    6-Bromo-pyridin-2- ylamine
    Figure US20120053345A1-20120301-C00427
    F.1.37 2.19 (e) 341 (M + H)+
    3-(5-Bromo- thiophen-2-yl)-1H- pyrazole
    Figure US20120053345A1-20120301-C00428
    F.1.38 2.42 (e) 399 (M + H)+
    3-Bromo-5-methyl- pyridin-2-ylamine
    Figure US20120053345A1-20120301-C00429
    F.1.39 2.50 (e) 357 (M + H)+
    7-Bromo-4H- benzo[1,4]thiazin-3- one
    Figure US20120053345A1-20120301-C00430
    F.1.40 2.30 (e) 412 (M − H)
    [2-(3-Iodo-pyridin- 2-ylsulfanyl)-ethyl]- carbamic acid tert- butyl ester
    Figure US20120053345A1-20120301-C00431
    F.1.41 2.30 (e) 401 (M − H)
    5-Bromo- pyrazin-2-yl amine
    Figure US20120053345A1-20120301-C00432
    F.1.42 1.98 (e) 342 (M − H)
    5-Bromo-2- methoxy-nicotinic acid methyl ester
    Figure US20120053345A1-20120301-C00433
    F.1.43 1.21 (e) 400 (M − H)
    3-Fluoro-4-iodo- pyridine
    Figure US20120053345A1-20120301-C00434
    F.1.44 2.78 (e) 344 (M − H)
    3-Fluoro-2-iodo- pyridine
    Figure US20120053345A1-20120301-C00435
    F.1.45 2.48 (e) 344 (M − H)
    2-Chloro-5-iodo- pyridine
    Figure US20120053345A1-20120301-C00436
    F.1.46 2.53 (e) 360 (M − H)
    N-(5-iodo-3- methyl-pyridin-2- yl) thiophene-2- carboxamide
    Figure US20120053345A1-20120301-C00437
    F.1.47 2.2 (e) 466.8 (M + H)+
    N-(5-Iodo-6- methyl-pyridin-2- yl)-isobutyramide
    Figure US20120053345A1-20120301-C00438
    F.1.48 2.3 (e) 427.0 (M + H)+
    4-Bromo-1H- indazole
    Figure US20120053345A1-20120301-C00439
    F.1.49 2.4 (e) 364.5 (M − H)
    1-Bromo-4-(4- fluorophenyl)- isoquinolin-3-yl amine
    Figure US20120053345A1-20120301-C00440
    F.1.50 2.9 (e) 486.9 (M + H)+
    N-(5-Iodo-3- methyl-pyridin-2- yl)-furan-2- carboxamide
    Figure US20120053345A1-20120301-C00441
    F.1.51 2.1 (e) 450.9 (M + H)+
    N-(5-Iodo-pyridin- 2-yl)-acetamide
    Figure US20120053345A1-20120301-C00442
    F.1.52 2.1 (e) 383.2 (M − H)
    4-Bromo-quinoline- 2-carboxamide
    Figure US20120053345A1-20120301-C00443
    F.1.53 2.5 (e) 420.9 (M + H)+
    5-Bromo-1H- pyrrolo[2,3- b]pyridine
    Figure US20120053345A1-20120301-C00444
    F.1.54 2.5 (e) 365.0 (M − H)
    5-Iodo-3-methyl- pyridin-2-ylamine
    Figure US20120053345A1-20120301-C00445
    F.1.55 2.4 (e) 354.6 (M − H)
    5-Bromo-1H- indazole
    Figure US20120053345A1-20120301-C00446
    F.1.56 2.3 (e) 364.6 (M − H)
    6-Iodo-1H- quinazolin-4-one
    Figure US20120053345A1-20120301-C00447
    F.1.57 2.1 (e) 392.6 (M − H)
  • TABLE F.2
    Examples prepared using general procedure F from Example #N.2.2
    Figure US20120053345A1-20120301-C00448
    HPLC Rt
    Boronates/Boronic Example (min) m/z
    Acids Product # (method) (ESI+)
    4-Hydroxyphenyl boronic acid
    Figure US20120053345A1-20120301-C00449
    F.2.1 1.1 (e) 317.6 (M − H)
    2-Hydroxyphenyl boronic acid
    Figure US20120053345A1-20120301-C00450
    F.2.2 1.7 (e) 317.6 (M − H)
    3-Hydroxyphenyl boronic acid
    Figure US20120053345A1-20120301-C00451
    F.2.3 1.1 (e) 317.4 (M − H)
    4-Hydroxymethyl phenyl boronic acid
    Figure US20120053345A1-20120301-C00452
    F.2.4 0.98 (e) 333.2 (M + H)+
    6-Methoxy-2- naphthalene boronic acid
    Figure US20120053345A1-20120301-C00453
    F.2.5 1.87 (e) 383.8 (M + H)+
    Benzamide-3- boronic acid
    Figure US20120053345A1-20120301-C00454
    F.2.6 0.93 (e) 346.1 (M + H)+
    Benzamide 4- boronic acid
    Figure US20120053345A1-20120301-C00455
    F.2.7 0.67 (e) 346.8 (M + H)+
    3-(Trifluoromethyl) phenylboronic acid
    Figure US20120053345A1-20120301-C00456
    F.2.8 1.93 (e) 369.6 (M − H)
    3-(Isopropyl) phenylboronic acid
    Figure US20120053345A1-20120301-C00457
    F.2.9 1.97 (e) 343.8 (M − H)
    3-(Trifluoro methoxy) phenylboronic acid
    Figure US20120053345A1-20120301-C00458
    F.2.10 2.02 (e) 385.4 (M − H)
    3-(Methoxy) phenylboronic acid
    Figure US20120053345A1-20120301-C00459
    F.2.11 1.58 (e) 331.6 (M − H)
    3-(Benzyloxy) phenylboronic acid
    Figure US20120053345A1-20120301-C00460
    F.2.12 2.05 (e) 409.2 (M + H)+
    3-Biphenyl boronic acid
    Figure US20120053345A1-20120301-C00461
    F.2.13 2.02 (e) 377.6 (M − H)
    3-Cyanophenyl boronic acid
    Figure US20120053345A1-20120301-C00462
    F.2.14 1.52 (e) 328.1 (M + H)+
    3-Ethoxyphenyl boronic acid
    Figure US20120053345A1-20120301-C00463
    F.2.15 1.82 (e) 347.3 (M + H)+
    3-Aminophenyl boronic acid
    Figure US20120053345A1-20120301-C00464
    F.2.16 0.93 (e) 318.7 (M + H)+
    3,5- Dimethylphenyl boronic acid
    Figure US20120053345A1-20120301-C00465
    F.2.17 1.75 (e) 331.0 (M + H)+
    3-Methyl-4- methoxyphenyl boronic acid
    Figure US20120053345A1-20120301-C00466
    F.2.18 1.55 (e) 347.0 (M + H)+
    3- (Hydroxymethyl) phenyl boronic acid
    Figure US20120053345A1-20120301-C00467
    F.2.19 0.93 (e) 333.3 (M + H)+
    3,4- Dimethylphenyl boronic acid
    Figure US20120053345A1-20120301-C00468
    F.2.20 1.65 (e) 331.0 (M + H)+
    3,4- Dimethoxyphenyl boronic acid
    Figure US20120053345A1-20120301-C00469
    F.2.21 1.30 (e) 362.0 (M + H)+
    3-Methyl-4-fluoro phenyl boronic acid
    Figure US20120053345A1-20120301-C00470
    F.2.22 1.24 (e) 335.3 (M + H)+
    Thianaphthene-2- boronic acid
    Figure US20120053345A1-20120301-C00471
    F.2.23 1.59 (e) 359.3 (M + H)+
    5- (Dimethylamino)- 1-benzothiophen- 2-yl boronic acid
    Figure US20120053345A1-20120301-C00472
    F.2.24 1.67 (e) 402.3 (M + H)+
    5-Methoxy-1- benzothiophen-2- yl boronic acid
    Figure US20120053345A1-20120301-C00473
    F.2.25 1.15 (e) 346.2 (M + H)+
    4-Carboxyphenyl boronic acid
    Figure US20120053345A1-20120301-C00474
    F.2.26 0.5 (e) 345.5 (M − H)
  • TABLE F.3
    Examples prepared using general procedure F using 6-bromo-1H-indazole
    Figure US20120053345A1-20120301-C00475
    Example Rt (min) m/z
    Boronic acid Product # (method) (ESI+)
    Benzothiophene- 2-boronic acid
    Figure US20120053345A1-20120301-C00476
    F.3.1 2.34 (e) 248.8 (M − H)
    2-Methoxy-5- pyridineboronic acid
    Figure US20120053345A1-20120301-C00477
    F.3.2 1.70 (e) 224.1 (M − H)
  • TABLE F.4
    Examples prepared using general procedure F using 5-(2-aminopyrimidin-
    4-yl)amino-3-chloro-7-iodo-1H-indazole
    Figure US20120053345A1-20120301-C00478
    Figure US20120053345A1-20120301-C00479
  • 5-(2-Aminopyrimidin-4-yl)amino-3-chloro-7-iodo-1H-indazole was prepared from 3-Chloro-5-nitro-1H-indazole (J. Med. Chem., 46(26); 2003; 5663-5673, via halogenation conditions used in the synthesis of Preparation #22a, and general procedure N (using 2-amino-4-chloro-pyrimidine)).
  • Example Rt (min) m/z
    Boronic Acid Product # (method) (ESI+)
    Benzothiophene- 2-boronic acid
    Figure US20120053345A1-20120301-C00480
    F.4.1 1.89 (e) 393.2 (M + H)+
    3-Quinoline boronic acid
    Figure US20120053345A1-20120301-C00481
    F.4.2 1.42 (e) 388.3 (M + H)+
  • TABLE F.5
    Examples prepared using general procedure F from Example #N.2.7
    Figure US20120053345A1-20120301-C00482
    HPLC Rt
    Example (min) m/z
    Boronate Product # (method) (ESI+)
    4,4,5,5- Tetramethyl-2- vinyl-[1,3,2] dioxaborolane
    Figure US20120053345A1-20120301-C00483
    F.5.1 0.93 (e) 250.9 (M − H)
    Indole-6-boronic acid
    Figure US20120053345A1-20120301-C00484
    F.5.2 1.38 (es) 342.3 (M + H)+
    2-[5-(4,4,5,5- Tetramethyl-[1,3,2] dioxaborolan-2-yl)- indol-1-yl]- acetamide (5-(4,4,5,5- Tetramethyl-[1,3,2] dioxaborolan-2-yl)- indole, R)
    Figure US20120053345A1-20120301-C00485
    F.5.3 1.18 (e) 399.3 (M + H)+
    5-Formylbenzo[b] thiophene-2-boronic acid pinacol
    Figure US20120053345A1-20120301-C00486
    F.5.4 1.41 (e) 387.3 (M + H)+
    3-Pyridylboronic acid
    Figure US20120053345A1-20120301-C00487
    F.5.5 1.10 (e) 304.3 (M + H)+
    N,N-dimethyl-1-[5- (4,4,5,5- tetramethyl-1,3,2- dioxaborolan-2-yl)- 1H-indole-3- yl]methanamine (Preparation #22b)
    Figure US20120053345A1-20120301-C00488
    F.5.6 1.43 (e) 399.0 (M + H)+
    Thieno[2,3- b]pyridin-2-yl boronic acid (Preparation #22c)
    Figure US20120053345A1-20120301-C00489
    F.5.7 1.25 (e) 360.3 (M + H)+
    2-[6-(4,4,5,5- Tetramethyl-1,3,2- dioxaborolan-2-yl)- 1H-indol-1- yl]acetamide (6- (4,4,5,5- Tetramethyl-1,3,2- dioxaborolan-2-yl)- 1H-indole, R)
    Figure US20120053345A1-20120301-C00490
    F.5.8 1.19 (e) 399.4 (M + H)+
    Indole-2-boronic- 1,4-dicarboxylic acid 1-tert-butyl ester 4-methyl ester (Preparation #21)
    Figure US20120053345A1-20120301-C00491
    F.5.9 1.2 (e) 400.3 (M + H)+
    4-Diisopropyl carbamoyl-indole- 2-boronic-1- carboxylic acid 1- tert-butyl ester (Preparation #22)
    Figure US20120053345A1-20120301-C00492
    F.5.10 1.5 (e) 467 (M − H)
    Indole-2-boronic- 1,7-dicarboxylic acid 1-tert-butyl ester 7-methyl ester (Preparation #20)
    Figure US20120053345A1-20120301-C00493
    F.5.11 1.4 (e) 400.3 (M + H)+
    5-Cyano-indole-2- boronic -1- carboxylic acid 1- tert-butyl ester
    Figure US20120053345A1-20120301-C00494
    F.5.12 1.0 (e) 367.4 (M + H)+
    2-[(E)-4-(4,4,5,5- Tetramethyl-[1,3, 2]dioxaborolan-2- yl)-but-3-enyloxy]- tetrahydro-pyran
    Figure US20120053345A1-20120301-C00495
    F.5.13 1.47 (e) 381.2 (M + H)+
    2-((E)-2- Trimethylsilanyl- vinyl)- [1,3,2]dioxaborolane
    Figure US20120053345A1-20120301-C00496
    F.5.14 1.95 (e) 325.1 (M + H)+
    5-Boronic acid- quinoline
    Figure US20120053345A1-20120301-C00497
    F.5.15 0.95 (e) 354.1 (M + H)+
    Thiophen-2-yl- boronic acid
    Figure US20120053345A1-20120301-C00498
    F.5.16 1.15 (e) 309.0 (M + H)+
    5-Boronic acid- pyrimidine
    Figure US20120053345A1-20120301-C00499
    F.5.17 0.98 (e) 304.9 (M + H)+
    8-Boronic acid- quinoline
    Figure US20120053345A1-20120301-C00500
    F.5.18 1.17 (e) 354.1 (M + H)+
    6-Boronic acid- naphthalen-2-ol
    Figure US20120053345A1-20120301-C00501
    F.5.19 1.43 (e) 369.1 (M + H)+
    6-Methoxy- naphthalen-2-yl- boronic acid
    Figure US20120053345A1-20120301-C00502
    F.5.20 1.80 (e) 383.1 (M + H)+
    5-Boronic acid- isoquinoline
    Figure US20120053345A1-20120301-C00503
    F.5.21 1.25 (e) 354.1 (M + H)+
    Furan-2-yl-boronic acid
    Figure US20120053345A1-20120301-C00504
    F.5.22 0.75 (e) 302.9 (M + H)+
    (5-Boronic acid- thiophen-2-yl)- methanol
    Figure US20120053345A1-20120301-C00505
    F.5.23 0.68 (e) 339.1 (M + H)+
    5-Phenyl-thiophen- 2-yl-boronic acid
    Figure US20120053345A1-20120301-C00506
    F.5.24 1.92 (e) 385.1 (M + H)+
    6-Boronic acid- quinoxaline
    Figure US20120053345A1-20120301-C00507
    F.5.25 0.97 (e) 355.1 (M + H)+
    6-Boronic acid- quinoline
    Figure US20120053345A1-20120301-C00508
    F.5.26 1.23 (e) 354.1 (M + H)+
    2- [2,2′]Bithiophenyl- 5-yl-4,4,5,5- tetramethyl- [1,3,2]dioxaborolane
    Figure US20120053345A1-20120301-C00509
    F.5.27 2.00 (e) 391.1 (M + H)+
    5-Methyl- benzo[b]thiophen- 2-yl-boronic acid
    Figure US20120053345A1-20120301-C00510
    F.5.28 1.94 (e) 373.1 (M + H)+
    (E)-2-(3-Methoxy- phenyl)-vinyl- boronic acid
    Figure US20120053345A1-20120301-C00511
    F.5.29 1.77 (e) 359.1 (M + H)+
    (E)-2-(4-Methoxy- phenyl)-vinyl- boronic acid
    Figure US20120053345A1-20120301-C00512
    F.5.30 1.76 (e) 359.1 (M + H)+
    4-Boronic acid- pyridine
    Figure US20120053345A1-20120301-C00513
    F.5.31 0.79 (e) 304.0 (M + H)+
    2-Boronic acid-1H- indole
    Figure US20120053345A1-20120301-C00514
    F.5.32 1.21 (e) 342.0 (M + H)+
    (3-Boronic acid- phenyl)-methanol
    Figure US20120053345A1-20120301-C00515
    F.5.33 0.56 (e) 333.3 (M + H)+
    N-(3-Boronic acid- phenyl)- methanesulfonamide
    Figure US20120053345A1-20120301-C00516
    F.5.34 0.63 (e) 396.3 (M + H)+
    [3-(4,4,5,5- Tetramethyl- [1,3,2]dioxaborolan- 2-yl)-phenyl]- acetonitrile
    Figure US20120053345A1-20120301-C00517
    F.5.35 0.84 (e) 342.3 (M + H)+
    3-Boronic acid- benzylamine
    Figure US20120053345A1-20120301-C00518
    F.5.36 0.38 (e) 332.3 (M + H)+
    3-Boronic acid- benzylamine
    Figure US20120053345A1-20120301-C00519
    F.5.37 0.63 (e) 381.3 (M + H)+
    3-(4,4,5,5- Tetramethyl- [1,3,2]dioxaborolan- 2-yl)-benzonitrile
    Figure US20120053345A1-20120301-C00520
    F.5.38 0.87 (e) 328.3 (M + H)+
    2-Boronic acid-6- methyl-1H-indole
    Figure US20120053345A1-20120301-C00521
    F.5.39 1.46 (e) 356.6 (M + H)+
    2-Boronic acid-5- methoxy-1H-indole
    Figure US20120053345A1-20120301-C00522
    F.5.40 1.18 (e) 372.3 (M + H)+
    2-Boronic acid-5- methyl-1H-indole
    Figure US20120053345A1-20120301-C00523
    F.5.41 1.45 (e) 356.3 (M + H)+
  • TABLE F.6
    Examples prepared using general procedure F using 3-iodo-5-(2-aminopyrimidin-4-yl)amino-1H-indazole
    Figure US20120053345A1-20120301-C00524
    HPLC Rt
    Example (min)
    Boronate Product # (method) m/z
    4-Methylphenyl boronic acid
    Figure US20120053345A1-20120301-C00525
    F.6.1 5.80 (a) 317 (M + H)+
    4-[5-(4,4,5,5-Tetramethyl-[1,3,2] dioxaborolan-2-yl)-pyridin-2-yl]- morpholine
    Figure US20120053345A1-20120301-C00526
    F.6.2 4.40 (a) 389 (M + H)+
    4-(4,4,5,5-Tetramethyl-1,3,2- dioxaborolan-2-yl)-1H-pyrazole
    Figure US20120053345A1-20120301-C00527
    F.6.3 0.60 (a) 293 (M + H)+
    4-(4,4,5,5-Tetramethyl-1,3,2- dioxaborolan-2-yl)aniline
    Figure US20120053345A1-20120301-C00528
    F.6.4 0.80 (a) 318 (M + H)+
    2-Methylphenyl boronic acid
    Figure US20120053345A1-20120301-C00529
    F.6.5 1.70 (a) 317 (M + H)+
    Phenylboronic acid
    Figure US20120053345A1-20120301-C00530
    F.6.6 1.70 (a) 303 (M + H)+
    3-Fluorophenyl boronic acid
    Figure US20120053345A1-20120301-C00531
    F.6.7 1.90 (a) 321 (M + H)+
    2-Fluorophenyl boronic acid
    Figure US20120053345A1-20120301-C00532
    F.6.8 1.60 (a) 321 (M + H)+
    4-Fluorophenyl boronic acid
    Figure US20120053345A1-20120301-C00533
    F.6.9 1.80 (a) 321 (M + H)+
    Pyridin-3-yl boronic acid
    Figure US20120053345A1-20120301-C00534
    F.6.10 0.80 (a) 304 (M + H)+
    Thiophen-3-yl boronic acid
    Figure US20120053345A1-20120301-C00535
    F.6.11 1.50 (a) 309 (M + H)+
    Indol-5-yl boronic acid
    Figure US20120053345A1-20120301-C00536
    F.6.12 1.40 (a) 342 (M + H)+
    1-(Tert-butoxycarbonyl)-1H- pyrrol-2-yl boronic acid
    Figure US20120053345A1-20120301-C00537
    F.6.13 1.10 (a) 292 (M + H)+
    4-(Methane sulfonyl)phenyl boronic acid
    Figure US20120053345A1-20120301-C00538
    F.6.14 1.20 (a) 381 (M + H)+
    Pyrimidin-5-yl boronic acid
    Figure US20120053345A1-20120301-C00539
    F.6.15 1.80 (a) 347 (M + H)+
    3-Methylphenyl boronic acid
    Figure US20120053345A1-20120301-C00540
    F.6.16 2.00 (a) 317 (M + H)+
    3-(N,N-Dimethylamino) phenylboronic acid
    Figure US20120053345A1-20120301-C00541
    F.6.17 1.90 (a) 346 (M + H)+
    4-Fluoro-3-methylphenyl boronic acid
    Figure US20120053345A1-20120301-C00542
    F.6.18 2.00 (a) 335 (M + H)+
    3,4-Difluorophenyl boronic acid
    Figure US20120053345A1-20120301-C00543
    F.6.19 2.10 (a) 339 (M + H)+
    2-Methoxy-5-methylphenyl boronic acid
    Figure US20120053345A1-20120301-C00544
    F.6.20 1.80 (a) 347 (M + H)+
    1-Naphthalene boronic acid
    Figure US20120053345A1-20120301-C00545
    F.6.21 2.10 (a) 353 (M + H)+
    3-Quinoline boronic acid
    Figure US20120053345A1-20120301-C00546
    F.6.22 1.40 (a) 354 (M + H)+
    2,3-Dihydro-1-benzofuran-5-yl boronic acid
    Figure US20120053345A1-20120301-C00547
    F.6.23 1.70 (a) 345 (M + H)+
    4-Isoquinoline boronic acid
    Figure US20120053345A1-20120301-C00548
    F.6.24 1.40 (a) 354 (M + H)+
    Benzo[b]thiophene-2-boronic acid
    Figure US20120053345A1-20120301-C00549
    F.6.25 2.40 (a) 359 (M + H)+
    2-Naphthalene boronic acid
    Figure US20120053345A1-20120301-C00550
    F.6.26 2.30 (a) 353 (M + H)+
    2,2′-Bithiophene-5-boronic acid
    Figure US20120053345A1-20120301-C00551
    F.6.27 2.60 (a) 390 (M + H)+
  • TABLE F.7
    Examples prepared using general procedure F from 3-iodo-5-(6-
    aminopyrrolo[2,3-d]pyrimidin-4-yl)amino-1H-indazole (prepared from Preparation #28
    using general procedure N starting with 6-amino-4-chloropyrrolo[2,3-d]pyrimidine.)
    Figure US20120053345A1-20120301-C00552
    Rt (min)
    Boronate Product Ex # (method) m/z
    Benzo[b]thiophene- 2-boronic acid
    Figure US20120053345A1-20120301-C00553
    F.7.1 2.40 (a) 398 (M + H)+
  • TABLE F.8
    Examples prepared using general procedure F from Preparation #6
    Figure US20120053345A1-20120301-C00554
    Boronic Example Rt (min) m/z
    Acid Product # (method) (ESI+)
    Benzo- thiophene- 2-boronic acid
    Figure US20120053345A1-20120301-C00555
    F.8.1 1.82 (e) 266.0 (M + H)+
  • TABLE F.9
    Examples prepared using general procedure F from Example #2
    Figure US20120053345A1-20120301-C00556
    Rt (min)
    Boronic Acid Product Example # (method) m/z (ESI+)
    Phenylboronic acid
    Figure US20120053345A1-20120301-C00557
    F.9.1 1.85 (e) 343.1 (M + H)+
  • TABLE F.10
    Examples prepared using general procedure F from 7-(1H-inden-2-yl)-5-(4,4,5,5-tetramethyl-[1,3,2]dioxaborolan-2-yl)-1-(2-
    trimethylsilanyl-ethoxymethyl)-1H-indazole (Preparation #23 then E)
    Figure US20120053345A1-20120301-C00558
    Example Rt (min)
    Aryl Halide Product # (method) m/z
    7-Benzo[b] thiophen-2-yl-5- bromo-1- (2-trimethylsilanyl- ethoxymethyl)-1H- indazole (Preparation #23)
    Figure US20120053345A1-20120301-C00559
    F.10.1 4.9 (h) 759 (M + H)+
  • TABLE F.11
    Examples prepared using general procedure F from Example #N.2.2
    Figure US20120053345A1-20120301-C00560
    m/z or 1H NMR
    Rt/min (d6 DMSO,
    Boronate/boronic acid Precursor Product Example # (method) 400 MHz)
    Thiophen-3-yl-boronic acid
    Figure US20120053345A1-20120301-C00561
    F.11.1 1.13 (e) 309.3 (M + H)+
    Naphthalen-2-yl-boronic acid
    Figure US20120053345A1-20120301-C00562
    F.11.2 1.59 (e) 351.3 (M + H)+
    p-Tolyl-boronic acid
    Figure US20120053345A1-20120301-C00563
    F.11.3 1.36 (e) 317 (M + H)+
    Phenyl-boronic acid
    Figure US20120053345A1-20120301-C00564
    F.11.4 1.18 (e) 303 (M + H)+
    5-Boronic acid-1H-indole
    Figure US20120053345A1-20120301-C00565
    F.11.5 0.84 (e) 342.3 (M + H)+
    Benzofuran-2-yl-boronic acid
    Figure US20120053345A1-20120301-C00566
    F.11.6 1.52 (e) 343.3 (M + H)+
    4-Chloro-phenyl-boronic acid
    Figure US20120053345A1-20120301-C00567
    F.11.7 1.58 (e) 337.2 (M + H)+
    3-Boronic acid-quinoline
    Figure US20120053345A1-20120301-C00568
    F.11.8 1.09 (e) 354 (M + H)+
    4-Phenol-boronic acid
    Figure US20120053345A1-20120301-C00569
    F.11.9 0.73 (e) 319.3 (M + H)+
    N-(4-boronic acid-phenyl)-acetamide
    Figure US20120053345A1-20120301-C00570
    F.11.10 0.67 (e) 360 (M + H)+
    Furan-3-yl-boronic acid
    Figure US20120053345A1-20120301-C00571
    F.11.11 1.12 (e) 293.2 (M + H)+
    2-Boronic acid-1H-pyrrole
    Figure US20120053345A1-20120301-C00572
    F.11.12 0.85 (e) 292 (M + H)+
    3-Methoxy-phenyl-boronic acid
    Figure US20120053345A1-20120301-C00573
    F.11.13 1.47 (e) 333.2 (M + H)+
    (4-Boronic acid-phenyl)-dimethyl- amine
    Figure US20120053345A1-20120301-C00574
    F.11.14 1.53 (e) 346 (M + H)+
    (3-Boronic acid-phenyl)-dimethyl- amine
    Figure US20120053345A1-20120301-C00575
    F.11.15 1.57 (e) 346 (M + H)+
    3-Boronic acid-benzamide
    Figure US20120053345A1-20120301-C00576
    F.11.16 0.65 (e) 346 (M + H)+
    (E)-Styryl-boronic acid
    Figure US20120053345A1-20120301-C00577
    F.11.17 1.50 (e) 329.2 (M + H)+
    4-Fluoro-3-methyl-phenyl-boronic acid
    Figure US20120053345A1-20120301-C00578
    F.11.18 1.67 (e) 335.2 (M + H)+
  • TABLE F.12
    Examples prepared using general procedure F from Example X.1.2
    Figure US20120053345A1-20120301-C00579
    Example Rt/min m/z
    Halide Precursor Product # (method) (ESI+)
    3-Hydroxybenzeneboronic acid
    Figure US20120053345A1-20120301-C00580
    F.12.1 1.07 (e) 226.3 (M + H)+
    Biphenyl-2-ol-5-(4,4,5,5- tetramethyl-1,3,2- dioxaborolan-2-yl) (E)
    Figure US20120053345A1-20120301-C00581
    F.12.2 1.59 (e) 302.3 (M + H)+
    1H-indazole-5- (4,4,5,5- tetramethyl-1,3,2- dioxaborolan-2-yl) (E)
    Figure US20120053345A1-20120301-C00582
    F.12.3 1.03 (e) 250.3 (M + H)+
    5-indolylboronic acid
    Figure US20120053345A1-20120301-C00583
    F.12.4 1.38 (e) 249.3 (M + H)+
    2-chloro-phenylamine-4- (4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl) (E)
    Figure US20120053345A1-20120301-C00584
    F.12.5 1.40 (e) 259.2 (M + H)+
    2-Benzo[b]thiophen-2-yl-4- (4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl) phenol (E)
    Figure US20120053345A1-20120301-C00585
    F.12.6 1.84 (e) 358.3 (M + H)+
    1H-indazol-3-ylamine-5- (4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl) (E)
    Figure US20120053345A1-20120301-C00586
    F.12.7 0.64 (e) 265.3 (M + H)+
    4-hydroxybenzeneboronic acid
    Figure US20120053345A1-20120301-C00587
    F.12.8 0.97 (e) 226.2 (M + H)+
    2-chloro-4-(4,4,5,5- tetramethyl-1,3,2- dioxaborolan-2-yl) phenol (E)
    Figure US20120053345A1-20120301-C00588
    F.12.9 1.30 (e) 260.2 (M + H)+
    4-Benzyloxy-3-chloro-phenyl- boronic acid
    Figure US20120053345A1-20120301-C00589
    F.12.10 2.08 (e) 350.2 (M + H)+
    2-Benzo[b]thiophen-2-yl- boronic acid*
    Figure US20120053345A1-20120301-C00590
    F.12.11 1.93 (e) 266.3 (M + H)+
    5-(4,4,5,5-Tetramethyl- [1,3,2]dioxaborolan-2-yl)- indazole-1-carboxylic acid tert-butyl ester*
    Figure US20120053345A1-20120301-C00591
    F.12.12 1.01 (e) 250.3 (M + H)+
    Pyridine-4-yl-boronic acid*
    Figure US20120053345A1-20120301-C00592
    F.12.13 0.82 (e) 211 (M + H)+
    Pyridine-3-yl-boronic acid
    Figure US20120053345A1-20120301-C00593
    F.12.14 1.03 (e) 211 (M + H)+
    Pyridine-3-yl-boronic acid**
    Figure US20120053345A1-20120301-C00594
    F.12.15 0.90 (e) 211 (M + H)+
    *Reacted with 3-Amino-4-bromo-indazole
    **Reacted with 3-Amino-6-bromo-indazole
  • TABLE F.13
    Examples prepared using general procedure F from Example #B.1.2
    Figure US20120053345A1-20120301-C00595
    m/z
    or 1H NMR
    Rt/min (d6 DMSO,
    Boronic acid Product Example # (method) 400 MHz)
    Pyridine-3-yl-boronic acid
    Figure US20120053345A1-20120301-C00596
    F.13.1 1.81 (e) 313.2 (M + H)+
    Benzonitrile-3-yl-boronic acid
    Figure US20120053345A1-20120301-C00597
    F.13.2 2.08 (e) 337.3 (M + H)+
    Benzonitrile-2-yl-boronic acid
    Figure US20120053345A1-20120301-C00598
    F.13.3 1.98 (e) 337.2 (M + H)+
    Benzamide-2-yl-boronic acid
    Figure US20120053345A1-20120301-C00599
    F.13.4 1.49 (e) 357.1 (M + H)+
    Benzamide-3-yl-boronic acid
    Figure US20120053345A1-20120301-C00600
    F.13.5 1.51 (e) 355 (M + H)+
    5-(boronic acid-2-yl)-isoquinoline
    Figure US20120053345A1-20120301-C00601
    F.13.6 1.86 (e) 363.2 (M + H)+
    4-(boronic acid-2-yl)-benzoic acid methyl ester
    Figure US20120053345A1-20120301-C00602
    F.13.7 2.31 (e) 370 (M + H)+
  • TABLE F.14
    Examples prepared using General Procedure F from Preparation #56
    Figure US20120053345A1-20120301-C00603
    m/z or 1H NMR
    Rt/min (d6 DMSO,
    Halide Product Example # (method) 400 MHz)
    4-Iodo-pyridine
    Figure US20120053345A1-20120301-C00604
    F.14.1 2.18 (e) 261 (M + H)+
    2-Bromo-pyridine
    Figure US20120053345A1-20120301-C00605
    F.14.2 2.53 (e) 261 (M + H)+
    3-Benzyloxy-5-bromo-pyridine
    Figure US20120053345A1-20120301-C00606
    F.14.3 2.98 (e) 367 (M + H)+
  • TABLE F.15
    Examples prepared using general procedure F using 5-(Dimethylamino-methyleneamino)-3-iodo-pyrrolo[2,3-c]pyridine-
    1-carboxylic acid tert-butyl ester followed by deprotection (outlined in Procedure #5, step 1)
    Figure US20120053345A1-20120301-C00607
    Rt/min
    Boronate Product Example # (method) m/z
    2-Chloro-4-(4,4,5,5-tetramethyl- [1,3,2]dioxaborolan-2-yl)-phenol
    Figure US20120053345A1-20120301-C00608
    F.15.1 1.16 (e) 260.2 (M + H)+
    2-(3-Benzo[b]thiophen-2-yl-4- methoxy-phenyl)-4,4,5,5- tetramethyl-[1,3,2]dioxaborolane
    Figure US20120053345A1-20120301-C00609
    F.15.2 1.89 (e) 358.3 (M + H)+
    4-Boronic acid-N-cyclopropyl- benzamide
    Figure US20120053345A1-20120301-C00610
    F.15.3 0.61 (e) 293.2 (M + H)+
    3-boronic acid-benzamide
    Figure US20120053345A1-20120301-C00611
    F.15.4 0.46 (e) 253 (M + H)+
    Indole-6-boronic acid
    Figure US20120053345A1-20120301-C00612
    F.15.5 1.14 (e) 249.2 (M + H)+
    4-boronic acid-benzoic acid
    Figure US20120053345A1-20120301-C00613
    F.15.6 0.44 (e) 254.2 (M + H)+
    4-Boronic acid-N-methyl- benzenesulfonamide
    Figure US20120053345A1-20120301-C00614
    F.15.7 0.69 (e) 303.2 (M + H)+
    4-Boronic acid- benzenesulfonamide
    Figure US20120053345A1-20120301-C00615
    F.15.8 0.56 (e) 289.2 (M + H)+
    4-Boronic acid-benzylamine
    Figure US20120053345A1-20120301-C00616
    F.15.9 1.44 (e) 239.2 (M + H)+
    3-boronic acid-N,N-dimethyl- benzamide
    Figure US20120053345A1-20120301-C00617
    F.15.10 0.60 (e) 281.2 (M + H)+
    4-boronic acid-N,N-dimethyl- benzamide
    Figure US20120053345A1-20120301-C00618
    F.15.11 0.56 (e) 281.2 (M + H)+
    3-boronic acid-benzamide
    Figure US20120053345A1-20120301-C00619
    F.15.12 0.48 (e) 253.2 (M + H)+
  • General Procedure G: Deprotection of a SEM-Protected Indazole
  • Figure US20120053345A1-20120301-C00620
  • A mixture of a SEM protected indazole in a solvent (for example MeOH, EtOH, i-PrOH, CH2Cl2 or dioxane, preferably MeOH) is treated with an excess amount of a mineral acid (for example HCl, HF or TFA, preferably HCl) and then stirred at about 25-85° C. (preferably about 65° C.) for about 1-24 hours (preferably about 1 hour). The solvent is evaporated and the product isolated and further purified by crystallization or chromatography.
  • Illustration of General Procedure G Example #14 5-(7-Benzo[b]thiophen-2-yl-1H-indazol-5-yl)-pyrimidin-2-ylamine
  • Figure US20120053345A1-20120301-C00621
  • A mixture of 6N HCl (1 mL) and 5-[7-benzo[b]thiophen-2-yl-2-(2-trimethylsilanyl-ethoxymethyl)-2H-indazol-5-yl]-pyrimidin-2-ylamine (0.063 g, 0.133 mmol) (prepared from 7-Benzo[b]thiophen-2-yl-5-(4,4,5,5-tetramethyl-[1,3,2]dioxaborolan-2-yl)-2-(2-trimethylsilanyl-ethoxymethyl)-2H-indazole (Preparation #23, E) and 5-iodo-pyrimidin-2-ylamine according to general procedure F) in MeOH (2 mL) was heated to about 65° C. for about 1 hour. The solvents were removed under reduced pressure and the residue treated with NaHCO3 (4 mL) and EtOAc (2 mL). The insoluble product was collected by filtration then dried to give 5-(7-benzo[b]thiophen-2-yl-1H-indazol-5-yl)-pyrimidin-2-ylamine (27 mg, 60%) as an off white solid; (DMSO-d6, 400 MHz) δ13.56 (bs, 1H), 8.66 (s, 2H), 8.29 (s, 1H), 8.15 (s, 1H), 8.04 (m, 2H), 7.93 (m, 1H), 7.79 (d, 1H), 7.43 (m, 2H), 7.76 (bs, 2H); RP-HPLC (Table 1, Method e) Rt=2.03 MS m/z: (M−H) 341.7.
  • TABLE G
    Examples prepared using general procedure G
    Figure US20120053345A1-20120301-C00622
    HPLC Rt
    Example (min)
    Precursor Product # (method) m/z
    Preparation #5
    Figure US20120053345A1-20120301-C00623
    G.1  1.68 (e) 383.0 (M + H)+
    {3-[7-(7- Benzo[b]thiophen-2- yl-1-(2- trimethylsilanyl- ethoxymethyl)-1H- indazol-5-yl)-2- (dimethylamino- methyleneamino)- pyrrolo[3,2- d]pyrimidin-5-yl]- propyl}-carbamic acid tert-butyl ester (Preparation #5, R (using tert-butyl 3- bromopropyl carbamate)
    Figure US20120053345A1-20120301-C00624
    G.2  1.29 (e) 440.3 (M + H)+
    {2-[7-(7- Benzo[b]thiophen-2- yl-1-(2- trimethylsilanyl- ethoxymethyl)-1H- indazol-5-yl)-2- (dimethylamino- methyleneamino)- pyrrolo[3,2- d]pyrimidin-5-yl]- ethyl}-carbamic acid tert-butyl ester (Preparation #5, R (using tert-butyl 2- bromoethyl carbamate))
    Figure US20120053345A1-20120301-C00625
    G.3  1.30 (e) 426.3 (M + H)+
    {2-Amino-7-[7- benzo[b]thiophen-2- yl-1-(2- trimethylsilanyl- ethoxymethyl)-1H- indazol-5-yl]- pyrrolo[3,2- d]pyrimidin-5-yl}- acetic acid (Preparation #5, R (using methyl 2- bromoacetate), V)
    Figure US20120053345A1-20120301-C00626
    G.4  0.97 (e) 441.3 (M + H)+
    2-{2-Amino-7-[7- benzo[b]thiophen-2- yl-1-(2- trimethylsilanyl- ethoxymethyl)-1H- indazol-5-yl]- pyrrolo[3,2- d]pyrimidin-5- yl}propionic acid (Preparation #5, R (using tert-butyl 2- bromopropionoate), Q, V)
    Figure US20120053345A1-20120301-C00627
    G.5  1.02 (e) 455.2 (M + H)+
    7-[7- Benzo[b]thiophen-2- yl-1-(2- trimethylsilanyl- ethoxymethyl)-1H- indazol-5-yl]-5- cyclopentyl-5H- pyrrolo[3,2-d] pyrimidin-2- ylamine (Preparation #5a, T (using cyclopentanol)
    Figure US20120053345A1-20120301-C00628
    G.6  2.15 (e) 451.3 (M + H)+
    7-[7- Benzo[b]thiophen-2- yl-1-(2- trimethylsilanyl- ethoxymethyl)-1H- indazol-5-yl]-5- isopropyl-5H- pyrrolo[3, 2-d]pyrimidin-2- ylamine (Preparation #5a, T (isopropanol))
    Figure US20120053345A1-20120301-C00629
    G.7  1.96 (e) 425.3 (M + H)+
    3-{2-Amino-7-[7- benzo[b]thiophen-2- yl-1-(2- trimethylsilanyl- ethoxymethyl)-1H- indazol-5-yl]- pyrrolo[3,2- d]pyrimidin-5-yl}- propionic acid (Preparation #5, R (3-bromo ethyl propionate), V)
    Figure US20120053345A1-20120301-C00630
    G.8  1.16 (e) 453.1 (M + H)+
    7-[7- Benzo[b]thiophen- 2-yl-1-(2- trimethylsilanyl- ethoxymethyl)-1H- indazol-5-yl]-5-(2- morpholin-4-yl- ethyl)-5H- pyrrolo[3,2- d]pyrimidin-2- ylamine N,N- dimethyl- formamidine (Preparation #5, R (using 1-(2- bromoethyl) morpholine)
    Figure US20120053345A1-20120301-C00631
    G.9  1.85 (e) 494.2 (M − H)+
    3-{2-Amino-7-[7- benzo[b]thiophen- 2-yl-1-(2- trimethylsilanyl- ethoxymethyl)-1H- indazol-5-yl]-pyrrolo [3,2-d]pyrimidin-5- yl{propionamide (Preparation #5, R (using methyl 3- bromopropionate, M)
    Figure US20120053345A1-20120301-C00632
    G.10 1.46 (e) 454.3 (M + H)+
    4-{2-[7-[7- Benzo[b]thiophen- 2-yl-1-(2- trimethylsilanyl- ethoxymethyl)-1H- indazol-5-yl]-2- (dimethylamino- methyleneamino)- pyrrolo[3,2- d]pyrimidin-5-yl]- ethyl}-piperidine-1- carboxylic acid tert- butyl ester (Preparation #5, R (using 4-(2-bromo- ethyl)-piperidine-1- carboxylic acid tert butyl ester)
    Figure US20120053345A1-20120301-C00633
    G.11 1.58 (e) 494.3 (M + H)+
    N-Methyl-3-{5- amino-3-[7- benzo[b]thiophen- 2-yl-1-(2- trimethylsilanyl- ethoxymethyl)- 1H-indazol-5- yl]-pyrrolo[3,2- d]pyrimidin-1-yl}- propionamide (Preparation #5, R (3-bromo ethyl propionate), M)
    Figure US20120053345A1-20120301-C00634
    G.12 1.56 (e) 468.4 (M + H)+
    3-{2- (Dimethylamino- methyleneamino)- 7-[7- benzo[b]thiophen-2- yl-1-(2- trimethylsilanyl- ethoxymethyl)-1H- indazol-5-yl]- pyrrolo[3,2-d] pyrimidin-5-yl}- propionic acid methyl ester (Preparation #5, R (using 3-bromo methyl propionate))
    Figure US20120053345A1-20120301-C00635
    G.13 1.87 (e) 469.0 (M + H)+
    3-{2-Amino-7-[7- benzo[b]thiophen- 2-yl-1-(2- trimethylsilanyl- ethoxymethyl)- 1H-indazol-5- yl]-pyrrolo[3,2-d] pyrimidin-5-yl}- N,N-dimethyl propionamide (Preparation #5, R (3-bromo methyl propionate), M)
    Figure US20120053345A1-20120301-C00636
    G.14 1.73 (e) 482.3 (M + H)+
    7-[7- Benzo[b]thiophen-2-yl-1-(2- trimethylsilanyl- ethoxymethyl)-1H- indazol-5-yl]-5-(2- piperidin-3-yl-ethyl)- 5H-pyrrolo[3,2- d]pyrimidin-2-yl- N,N-dimethylamino- methyleneamine (Preparation #5, R (using 3-(2-bromo- ethyl)-piperidine-1- carboxylic acid tert- butyl ester)
    Figure US20120053345A1-20120301-C00637
    G.15 1.58 (e) 494.0 (M + H)+
    3-{2-Amino-7-[7- benzo[b]thiophen- 2-yl-1-(2- trimethylsilanyl- ethoxymethyl)- 1H-indazol-5- yl]-pyrrolo[3,2-d] pyrimidin-5-yl}-N- (2-amino-ethyl)- propionamide (Preparation #5, R (3-bromo methyl propionate), M)
    Figure US20120053345A1-20120301-C00638
    G.16 1.31 (e) 497.0 (M + H)+
    {3-[7-(7- Benzo[b]thiophen- 2-yl-1-(2- trimethylsilanyl- ethoxymethyl)-1H- indazol-5-yl)-2- (dimethylamino- methyleneamino)- pyrrolo[3,2- d]pyrimidin-5-yl]- butyl}-carbamic acid tert-butyl ester (Preparation #5, R (using tert-butyl 4- bromo-butyl carbamate)
    Figure US20120053345A1-20120301-C00639
    G.17 1.40 (e) 454.1 (M + H)+
    7-[7- Benzo[b]thiophen- 2-yl-1-(2- trimethylsilanyl- ethoxymethyl)-1H- indazol-5-yl]-5-(3- methoxy-propyl)- 5H-pyrrolo[3,2- d]pyrimidin-2-yl- N,N-dimethylamino- methyleneamine (Preparation #5, R (using 1-bromo-3- methoxypropane))
    Figure US20120053345A1-20120301-C00640
    G.18 2.24 (e) 455.0 (M + H)+
    2-(3-{2- (Dimethylamino- methyleneamino)- 7-[7- benzo[b]thiophen- 2-yl-1-(2- trimethylsilanyl- ethoxymethyl)-1H- indazol-5-yl]- pyrrolo[3,2-d] pyrimidin-5-yl}- propan-1- oxy)tetrahydropyran (Preparation #5, R (using 2-(3-bromo- propan-1-oxy) tetrahydropyran))
    Figure US20120053345A1-20120301-C00641
    G.19 1.64 (e) 441.3 (M + H)+
    2-{2-Amino-7-[7- benzo[b]thiophen- 2-yl-1-(2- trimethylsilanyl- ethoxymethyl)- 1H-indazol-5- yl]-pyrrolo[3,2-d] pyrimidin-5-yl}- acetamide (Preparation #5, R (using 2-bromo methyl acetate), M)
    Figure US20120053345A1-20120301-C00642
    G.20 1.49 (e) 440.0 (M + H)+
    4-{2-Amino-7-[7- benzo[b]thiophen- 2-yl-1-(2- trimethylsilanyl- ethoxymethyl)- 1H-indazol-5- yl]-pyrrolo[3,2- d]pyrimidin-5-yl}- butyric acid (Preparation #5, R (using methyl 4- bromo-butanoate), V)
    Figure US20120053345A1-20120301-C00643
    G.21 1.61 (e) 467.0 (M − H)+
    4-{2-Amino-7-[7- benzo[b]thiophen- 2-yl-1-(2- trimethylsilanyl- ethoxymethyl)- 1H-indazol-5- yl]-pyrrolo[3,2- d]pyrimidin-5-yl}- butyramide (Preparation #5, R (using methyl 4- bromo-butanoate), M)
    Figure US20120053345A1-20120301-C00644
    G.22 1.49 (e) 468.2 (M + H)+
    3-{2-Amino-7-[7- benzo[b]thiophen- 2-yl-1-(2- trimethylsilanyl- ethoxymethyl)- 1H-indazol-5- yl]-pyrrolo[3,2-d] pyrimidin-5-yl}-2- methyl-propionic acid (Preparation #5, R (methyl 3-bromo 2- methyl propionoate), V)
    Figure US20120053345A1-20120301-C00645
    G.23 1.63 (e) 469.2 (M + H)+
    3-{2-Amino-7-[7- benzo[b]thiophen- 2-yl-1-(2- trimethylsilanyl- ethoxymethyl)- 1H-indazol-5- yl]-pyrrolo[3,2-d] pyrimidin-5-yl}-2- methyl-propionic acid methyl ester (Preparation #5, R (using methyl 3- bromo 2-methyl propionoate))
    Figure US20120053345A1-20120301-C00646
    G.24 1.96 (e) 483.3 (M + H)+
    2-Amino-7-[7- benzo[b]thiophen- 2-yl-1-(2- trimethylsilanyl- ethoxymethyl)- 1H-indazol-5-yl]- pyrrolo[3,2- d]pyrimidine-5- carboxylic acid isopropyl ester (Preparation #5, T (using diisopropyl azodicarboxylate)
    Figure US20120053345A1-20120301-C00647
    G.25 2.22 (e) 469.1 (M + H)+
  • General Procedure H: Acid Cleavage of a TBDMS-Protecting Group
  • Figure US20120053345A1-20120301-C00648
  • To a mixture of a TBDMS protected substrate in an organic solvent (for example methanol, ethanol, ispropanol, CH2Cl2, or dioxane, preferably methanol) is added an excess amount of an acid (5-50 equivalents, preferably 20 equivalents) (for example HCl, or TFA, preferably HCl). The reaction mixture is then stirred at about 25-85° C. (preferably about 65° C.) for about 0.5-24 hours (preferably about 1 hour). The solvent is evaporated and the product is isolated by crystallization or by chromatography.
  • Illustration of General Procedure H Example #30 (3-[(2-Amino-pyrimidin-4-yl)-(7-benzo[b]thiophen-2-yl-1H-indazol-5-yl)-amino]-propan-1-ol
  • Figure US20120053345A1-20120301-C00649
  • 6N hydrochloric acid (1 mL) was added to a mixture of N4-(7-benzo[b]thiophen-2-yl-1H-indazol-5-yl)-N4-[3-(tert-butyl-dimethyl-silanyloxy)-propyl]-pyrimidine-2,4-diamine (prepared from example #F.8.1, via general procedures O then N, 0.179 g, 0.339 mmol) in ethanol (2 mL). The reaction mixture was heated to about 65° C. for about 1 hour then cooled to about ambient temperature. The solvents were evaporated and the residue was purified by reverse phase chromatography (Thermo Hypersil-Keystone 250×21.2 mm 8μ Hypersil® HS C18 column; 5% acetonitrile/0.1 M aqueous ammonium acetate-100% acetonitrile over 20 min, 100% acetonitrile hold 5 minutes, 21 mL/min) to afford (3-[(2-amino-pyrimidin-4-yl)-(7-benzo[b]thiophen-2-yl-1H-indazol-5-yl)-amino]-propan-1-ol (0.022 g, 0.053 mmol) as an off white solid; RP-HPLC (Table 1, Method e) Rt 1.77 min.; m/z: (M+H)+ 417.1.
  • TABLE H.1
    Examples prepared using general procedure H
    Figure US20120053345A1-20120301-C00650
    TBDMS Example Rt/min m/z
    protected alcohol Produce # (method) (ESI+)
    N′4′-(7- Benzo[b]thiophen-2- yl-1H-indazol-5-yl)- N′4′-[2-(tert-butyl- dimethyl-silanyloxy)- ethyl]-pyrimidine- 2,4-diamine (Example #F.8.1, O then N)
    Figure US20120053345A1-20120301-C00651
    H.1.1 1.52 (e) 390 (M + H)+
  • General Procedure I: Deprotection of a Benzyl Ether
  • Figure US20120053345A1-20120301-C00652
  • A mixture of a benzyl protected ether (1 equivalent) and a Pd catalyst (for example, 10 wt. % Pd on carbon or palladium (II) oxide, preferably 10 wt. % Pd on carbon) (5-10 mol %, preferably 7 mol %) in a degassed organic solvent (for example MeOH or EtOH, preferably MeOH) is shaken under an atmosphere of hydrogen gas at about 30-50 psi (preferably 40 psi) at about 10-50° C. (preferably about 25° C.) for about 2-24 hours (preferably about 18 hours). The reaction mixture is evacuated, flushed with an inert gas (for example nitrogen or argon, preferably nitrogen) and filtered. The solvents are removed under reduced pressure to afford the product that can be further purified by chromatography or crystallization.
  • Illustration of General Procedure I Example #15 5-[3-(1H-Pyrrol-2-yl)-1H-indazol-5-yl]-pyridin-3-ol
  • Figure US20120053345A1-20120301-C00653
  • A mixture of 5-(5-benzyloxy-pyridin-3-yl)-3-(1H-pyrrol-2-yl)-1H-indazole (Preparation #16c, E and F (using 5-benzyloxy-3-bromopyridine), 0.015 g, 0.4 mmol) in MeOH (5 mL) and 10 wt. % Pd/C (3 mg) was shaken under an atmosphere of hydrogen (about 40 psi) for about 24 hours. The crude product was filtered and the solvent was removed under reduced pressure to yield 5-[3-(1H-pyrrol-2-yl)-1H-indazol-5-yl]-pyridin-3-ol (0.088 g, 80%); RP-HPLC (Table 1, Method e) Rt=1.78 min; m/z: (M+H)+277, (M−H) 275.
  • General Procedure J: Reduction of an Aldehyde to an Alcohol
  • Figure US20120053345A1-20120301-C00654
  • A mixture of an aldehyde (1-1.2 equivalents, preferably 1 equivalent) and a reducing agent (for example borane, borane-pyridine, borane-diemethylsulfide, LiBH4 or NaBH4, preferably NaBH4) (1.0-3.0 equivalents, preferably 2.0 equivalents) in an organic solvent (for example, DMF, dioxane, THF, MeOH or EtOH, preferably MeOH) is stirred at about 5-65° C. (preferably about 25° C.) for about 0.5-24 hours (preferably about 1 hour) under an inert atmosphere. If heated, the mixture is allowed to cool to ambient temperature, and the solvent is removed under reduced pressure. The solid residue can then be purified by chromatography or crystallization.
  • Illustration of General Procedure J Example #16 [3-(7-Benzo[b]thiophen-2-yl-1H-indazol-5-yl)-phenyl]-methanol
  • Figure US20120053345A1-20120301-C00655
  • A mixture of 3-(7-benzo[b]thiophen-2-yl-1H-indazol-5-yl)-benzaldehyde (Preparation #25, F (using 3-bromobenzaldehyde), 50 mg, 0.14 mmol), and NaBH4 (0.01 g, 0.26 mmol) in MeOH (2 mL) was stirred at ambient temperature for about 1 hour. The reaction mixture was concentrated under reduced pressure then the residue was purified by reverse phase preparative HPLC (Thermo Hypersil-Keystone 250×21.2 mm 8μ Hypersil® HS C18 column; 5% CH3CN/50 mM aqueous ammonium acetate hold for 5 min; 5-50% CH3CN/50 mM aqueous ammonium acetate over 20 min; 50-100% CH3CN/50 mM aqueous ammonium acetate over 1 min; hold at 100% CH3CN for 5 minutes, 21 mL/min) to yield [3-(7-benzo[b]thiophen-2-yl-1H-indazol-5-yl)-phenyl]-methanol (6 mg, 12%); (DMSO-d6, 400 MHz) 8.8 (d, 1H), 8.55 (d, 1H), 8.37 (s, 1H), 8.19 (d, 1H), 8.18 (s, 1H), 8.10 (t, 1H), 8.04 (dd, 1H), 7.93 (dd, 1H), 7.89 (d, 1H, 7.45 (m, 2H), 5.40 (t, 1H), 4.6 (d, 2H); RP-HPLC (Table 1, Method e) Rt=1.85 min; m/z: (M−H) 355.8.
  • General Procedure K: Nucleophilic Displacement of an Aryl Sulfone
  • Figure US20120053345A1-20120301-C00656
  • An aryl sulfone is treated with a large excess of a nucleophile (for example an amine or an alcohol, preferably an amine) (100-500 equivalents, preferably 250 equivalents) in the absence of an organic solvent. The mixture is stirred for 5-60 hours (preferably about 16 hours) at about 0-100° C. (preferably about 20° C.). If heated, the mixture is allowed to cool to ambient temperature, filtered, and the solids are further purified by crystallization or chromatography if necessary.
  • Illustration of General Procedure K Example #17 (7-Benzo[b]thiophen-2-yl-1H-indazol-5-yl)-(2-hydrazino-pyrimidin-4-yl)-amine
  • Figure US20120053345A1-20120301-C00657
  • (7-Benzo[b]thiophen-2-yl-1H-indazol-5-yl)-(2-methanesulfonyl-pyrimidin-4-yl)-amine (Preparation #8, 20 mg, 0.048 mmol) was treated with hydrazine (0.4 mL, 12.2 mmol) at ambient temperature for about 5 hours. A solid was filtered off and was further purified by trituration in dichloromethane followed by filtration to yield (7-benzo[b]thiophen-2-yl-1H-indazol-5-yl)-(2-hydrazino-pyrimidin-4-yl)-amine (4 mg, 23% yield); LC/MS (method f) Rt 6.0 min; m/z (ESI−): (M−H)+ 371.7.
  • TABLE K
    Examples using general procedure K prepared from (7-benzo[b]thiophen-2-
    yl-1H-indazol-5-yl)-(2-methanesulfonyl-pyrimidin-4-yl)-amine (Preparation #8)
    Figure US20120053345A1-20120301-C00658
    Rt m/z
    Example (min) (ESI−):
    Nucleophile Product # (method) (M − H)+
    N′,N′-Dimethyl- ethane-1,2-diamine
    Figure US20120053345A1-20120301-C00659
    K.1.1 1.7 (e) 430.1 (M − H)+
    Pyrrolidin-3-ol
    Figure US20120053345A1-20120301-C00660
    K.1.2 1.7 (e) 429.1 (M − H)+
    2-Amino-ethanol
    Figure US20120053345A1-20120301-C00661
    K.1.3 1.6 (e) 403.1 (M − H)+
    2-Methoxy- ethylamine
    Figure US20120053345A1-20120301-C00662
    K.1.4 1.8 (e) 417.1 (M − H)+
    2-Dimethylamino- ethanol
    Figure US20120053345A1-20120301-C00663
    K.1.5 1.9 (e) 431.1 (M − H)+
    2-(2-Hydroxy- ethylamino)-ethanol
    Figure US20120053345A1-20120301-C00664
    K.1.6 1.6 (e) 447.1 (M − H)+
  • General Procedure L: Reduction of a Nitroaromatic Compound to an Aniline
  • To a solution of a nitroaromatic compound (preferably 1 equivalent) in a solvent (for example, EtOAc, EtOH, HOAc, 9M NH4OH, preferably EtOAc) is added a reducing reagent (for example, hydriodic acid, iron powder, iron sulfate hydrate, tin chloride dehydrate, or palladium on carbon) (0.2-10 equivalents, preferably 3 equivalents). The reaction mixture is stirred at about 20-100° C. (preferably about 90° C.) for about 1-20 hours (preferably about 2 hours). A hydrogen atmosphere (about 15-60 psi, preferably about 40 psi) is when a palladium catalyst is employed. If iron sulfate hydrate is used as the reducing reagent the mixture is filtered hot and then acidified with HOAc to about pH=4, then the product is filtered off and rinsed with water. If tin chloride or iron powder are used the mixture is allowed to cool to ambient temperature and then diluted with an organic solvent (for example, EtOAc or CH2Cl2, preferably CH2Cl2). If palladium is used as the reducing reagent the reaction mixture is filtered through celite and rinsed with organic solvent. If hydriodic acid is used, the organic layer is washed with saturated aqueous sodium thiosulfate. The organic layer is then washed with an aqueous base solution (for example, Na2CO3 or NaOH, preferably NaOH). The organic layer is separated, dried over a desiccant, and concentrated to give the aniline compound.
  • Illustration of General Procedure L Preparation #28 3-Iodo-1H-indazol-5-ylamine
  • Figure US20120053345A1-20120301-C00665
  • A suspension of 3-iodo-5-nitro-1H-indazole (2.0 g, 6.92 mmol) in stabilized hydroiodic acid (57% wt aqueous solution, 21 mL) was heated at about 90° C. for about 2 hours. The reaction mixture became homogeneous as the reaction progressed. After cooling to ambient temperature, the dark purple mixture was diluted with EtOAc (500 mL) and washed successively with saturated aqueous sodium thiosulfate (200 mL), saturated aqueous NaHCO3 (200 mL) and brine (200 mL). The colorless organic layer was dried over anhydrous magnesium sulfate, and concentrated to dryness to give 3-iodo-1H-indazol-5-ylamine: 1H NMR (DMSO-d6, 400 MHz) δ 13.03 (s, 1H), 7.26 (d, 1H), 6.85 (m, 1H), 6.44 (d, 1H), 5.03 (s, 2H); LC/MS (30% to 95% acetonitrile/0.01M aqueous ammonium acetate over 4.5 min at 0.8 mL/min; λ=190-700 nm; Genesis C18, 3 μm, 30×4.6 mm column; Electrospray ionization method observing both +ve and −ve ions) Rt 1.25 min.; m/z: 260 (M+H)+.
  • General Procedure M: Amide Formation from an Ester
  • Figure US20120053345A1-20120301-C00666
  • At room temperature, a mixture of an ester (1.0 equivalent) and an amine (for example, ammonia, a primary amine or a second amine) (10-500 equivalents, preferably 20 equivalents) optionally diluted with an alcoholic solvent (MeOH, EtOH or i-PrOH, preferably i-PrOH) is heated at about 60-140° C. (preferably about 100° C.) in a sealed vessel with stirring for about 0.5-7 days (preferably about 1 day). The mixture is cooled to ambient temperature, concentrated and is further purified by chromatography or crystallization, or used in a subsequent step without further purification.
  • Illustration of General Procedure M:
  • See Example #1.
  • General Procedure N: Nucleophilic Substitution of Aromatic Halide with an Amino Compound
  • Figure US20120053345A1-20120301-C00667
  • To a solution of an amine (1.0 equivalent) with or without a base (Et3N or DIEA, preferably Et3N) in an organic solvent (for example, EtOH, MeOH, or dioxane, preferably EtOH) is added an aromatic halide (1.0-10 equivalents, preferably 1.0 equivalent). The reaction mixture is heated at about 25-160° C. (preferably about 80° C.) with stirring for about 20 minutes-4 hours (preferably about 30 minutes) or heated in a microwave reactor at about 120-180° C. (preferably about 150° C.) for about 5-30 minutes (preferably about 10 minutes). The mixture is allowed to cool to ambient temperature. Then either the solvent is removed under reduced pressure or the mixture is filtered to give crude product that is then purified directly by chromatography or crystallization or submitted to the following aqueous work up. The crude material is suspended in a basic solution (for example, NH4OH in MeOH, saturated aqueous NaHCO3 or 2M aqueous Na2CO3, preferably saturated aqueous NaHCO3) then filtered and washed with an organic solvent (for example MeOH, EtOAc, or CH2Cl2). If needed, the resulting solid can then be purified by chromatography or crystallization. If a tert-butoxycarbonyl (Boc) protected diamine is used then the material is suspended in a mixture of methanol/6 N hydrochloric acid and heated to about 65° C. for about one hour then cooled, concentrated and purified by chromatography or crystallization
  • Illustration of General Procedure N Preparation #46 (7-Benzo[b]thiophen-2-yl-1H-indazol-5-yl)-(2-methanesulfonyl-pyrimidin-4-yl)-amine
  • Figure US20120053345A1-20120301-C00668
  • To a mixture of 4-chloro-2-methanesulfonyl-pyrimidine (2.54 g, 13.2 mmol) in DME (20 mL) was added a mixture of 7-benzo[b]thiophen-2-yl-1H-indazol-5-ylamine (Example #F.8.1, 1.95 g, 7.35 mmol) and Et3N (1.43 mL, 10.3 mmol) in DME (160 mL). The reaction mixture was stirred at room temperature for about 16 hours then filtered, then filtrate was adsorbed onto alumina. The mixture was purified by flash column chromatography over alumina using CH2Cl2/MeOH (99:1) as an eluent to yield the initial crop of product. A second crop was obtained by triturating the alumina in CH2Cl2/MeOH (8:2) and combined with the first to afford 7-benzo[b]thiophen-2-yl-1H-indazol-5-yl)-(2-methanesulfonyl-pyrimidin-4-yl)-amine (1.09 g, 36% yield): LC/MS (method e) Rt 1.8 min; m/z (ESI−): (M−H)+ 419.9.
  • TABLE N.1
    Examples prepared using general procedure N from Example #F.8.1
    Figure US20120053345A1-20120301-C00669
    m/z or
    1H NMR
    Rt (min) (d6 DMSO,
    Aryl halide Product Ex. # (method) 400 MHz)
    7-Chloro-1H- pyrrolo[2,3- c]pyridine
    Figure US20120053345A1-20120301-C00670
    N.1.1  2.2 min (e) 382.0 (M + H)+
    4-Chloro- pyridin-3- ylamine
    Figure US20120053345A1-20120301-C00671
    N.1.2   14 min (b) 1.87 (s, 3 H), 4.85 (bs, 2 H), 6.85 (d, 1 H), 7.41 (m, 3 H), 7.44 (s, 1 H), 7.57 (s, 1 H), 7.61 (m, 1 H), 7.88 (m, 2 H), 8.00 (m, 1 H), 8.15 (m, 2 H)
    2-Chloro-4,6- dimethoxy- 1,3,5-triazine
    Figure US20120053345A1-20120301-C00672
    N.1.3  2.1 min (e) 403.0 (M + H)+
    2-Amino-6- chloropurine
    Figure US20120053345A1-20120301-C00673
    N.1.4  1.6 min (e) 399.0 (M + H)+
    6-Chloro- pyrimidine- 2,4-diamine
    Figure US20120053345A1-20120301-C00674
    N.1.5  1.5 min (e) 373.9 (M + H)+
    4-Chloro- thieno[3,2- c]pyridine
    Figure US20120053345A1-20120301-C00675
    N.1.6  2.5 min (e) 398.9 (M + H)+
    6-Chloro- pyridazin-3- ylamine
    Figure US20120053345A1-20120301-C00676
    N.1.7  1.7 min (e) 358.9 (M + H)+
    2-Chloro- pyrimidine- 4,5-diamine
    Figure US20120053345A1-20120301-C00677
    N.1.8  1.6 min (e) 374.0 (M + H)+
    6-Chloro- [1,3,5]triazine- 2,4-diamine
    Figure US20120053345A1-20120301-C00678
    N.1.9  1.5 min (e) 373.1 (M − H)
    4-Chloro- thieno[3,2- d]pyrimidine
    Figure US20120053345A1-20120301-C00679
    N.1.10 2.2 min (e) 399.8 (M + H)+
    2-Chloro-6,7- dimethoxy- quinazolin-4- ylamine
    Figure US20120053345A1-20120301-C00680
    N.1.11 2.1 min (e) 468.9 (M + H)+
    1H- Pyrrolo[2,3- d]pyrimidin- 4-ylamine
    Figure US20120053345A1-20120301-C00681
    N.1.12 2.0 min (e) 383.0 (M + H)+
    6-Chloro- nicotinamide
    Figure US20120053345A1-20120301-C00682
    N.1.13 2.4 min (e) 7.10 (d, 1 H), 7.34 (m, 3 H), 7.37 (s, 1 H), 7.91 (d, 1 H), 7.97 (d, 2 H), 8.08 (m, 3 H), 8.22 (bs, 1 H), 8.58 (s, 1 H), 9.19 (s, 1 H), 13.27 (bs, 1H)
    2-Acetamido- 4-chloro pyrimidine
    Figure US20120053345A1-20120301-C00683
    N.1.14 2.50 (e) 353.0 (M − H)
  • TABLE N.2
    Examples using general procedure N from 4-chloro-2-aminopyrimidine
    Figure US20120053345A1-20120301-C00684
    Rt (min)
    Amine Product Ex. # (method) m/z
    (7-Benzo[b] thiophen-2-yl- 1H-indazol-5- yl)-(4- imidazo[1,2- a]pyridin-2-yl- benzyl)- amine (Example # F.8.1, then O)
    Figure US20120053345A1-20120301-C00685
    N.2.1  1.6 min (e) 565.4 (M + H)+
    3-Iodo-1H- indazol-5- ylamine (FR2836914)
    Figure US20120053345A1-20120301-C00686
    N.2.2   0.8 (e) 261.2 (M − H)
    5-Amino-1H- indazol-3-ol (FR2836914)
    Figure US20120053345A1-20120301-C00687
    N.2.3  0.96 (f) 241.2 (M − H)
    5-Amino-7- benzo[b]thiophen- 2-yl-1H- indazol-3-ol (Preparation #50, N, L)
    Figure US20120053345A1-20120301-C00688
    N.2.4  1.26 (e) 375.3 (M − H)
    N-(5-Amino-3- chloro-1H- indazol-7-yl)- acetamide (J. Med. Chem., 2003, 46(26), 5663-5673)
    Figure US20120053345A1-20120301-C00689
    N.2.5  0.62 (e) 318.0 (M + H)+
    3-Iodo-1H- indazol-5- ylamine (Preparation #28)
    Figure US20120053345A1-20120301-C00690
    N.2.6  1.22 (a) 352.8 (M + H)+
    7-Bromo-1H- indazol-5- ylamine (Preparation #6)
    Figure US20120053345A1-20120301-C00691
    N.2.7  1.13 (e) 305.1 (M + H)+
    5-Amino-1H- indazole-3- carboxylic acid
    Figure US20120053345A1-20120301-C00692
    N.2.8  0.40 (e) 270.9 (M + H)+
    N-(5-Amino- 1H-indazol-3- yl)-benzamide (W,L)
    Figure US20120053345A1-20120301-C00693
    N.2.9  1.08 (e) 346.1 (M + H)+
    N-(5-Amino- 1H-indazol-3- yl)-3-methoxy- benzamide (W,L)
    Figure US20120053345A1-20120301-C00694
    N.2.10 0.93 (e) 376.4 (M + H)+
    N-(5-Amino- 1H-indazol-3- yl)-3- dimethylamino- benzamide (W,L)
    Figure US20120053345A1-20120301-C00695
    N.2.11 1.13 (e) 389.2 (M + H)+
    N′3′-Benzyl- 1H-indazole- 3,5-diamine (O,L)
    Figure US20120053345A1-20120301-C00696
    N.2.12 1.61 (e) 425.3 (M + H)+
    1H-Indazol-4- ylamine (L)
    Figure US20120053345A1-20120301-C00697
    N.2.13 0.48 (e) 226.8 (M + H)+
    6-Methoxy- 1H-indazol-5- ylamine (According to Preparation #6)
    Figure US20120053345A1-20120301-C00698
    N.2.14 0.40 (e) 257.3 (M + H)+
    7-Chloro-1H- indazol-5- ylamine (According to Preparation #6)
    Figure US20120053345A1-20120301-C00699
    N.2.15 1.58 (e) 389   (M + H)+
    (7-Benzo[b] thiophen-2-yl- 1H-indazol-5- yl)-(tetrahydro- pyran-4- ylmethyl)- amine (Example #F.8.1, O)
    Figure US20120053345A1-20120301-C00700
    N.2.16 2.17 (e) 457.1 (M + H)+
    3-Chloro-1H- indazol-5- ylamine (J. Med. Chem. (2003), 46(26), 5663- 5673)
    Figure US20120053345A1-20120301-C00701
    N.2.17 3.03 (f) 261.2 (M + H)+
    3-Morpholin- 4-yl-1H- indazol-5- ylamine (Pharmazie, 33(6), 377-8, 1978, N, L)
    Figure US20120053345A1-20120301-C00702
    N.2.18 2.43 (f) 311.9 (M + H)+
    N-(5-Amino-3- chloro-1H- indazol-7-yl)- benzamide (J. Med. Chem., 46(26), 5663- 5673, 2003, W, L)
    Figure US20120053345A1-20120301-C00703
    N.2.19 1.47 (e) 380.3 (M + H)+
    (5-Amino-3- chloro-1H- indazol-7-yl)- carbamic acid tert-butyl ester (J. Med. Chem., 46(26), 5663-5673, 2003, W, L)
    Figure US20120053345A1-20120301-C00704
    N.2.20 0.52 (e) 276.0 (M + H)+
    N-(5-Amino-3- chloro-1H- indazol-7-yl)- methane- sulfonamide (J. Med. Chem., 46(26), 5663-5673, 2003, S, L)
    Figure US20120053345A1-20120301-C00705
    N.2.21 2.82 (f) 354.2 (M + H)+
  • Representative NMR Data of Example #N.2.8
  • 1H NMR (DMSO-d6, 400 MHz); 8.28 (br, 1H), 8.10 (m, 1H), 8.03 (d, 1H), 7.92 (d, 1H), 7.78 (s, 1H), 7.61 (d, 1H), 7.43 (m, 3H), 6.07 (m, 2H), 5.42 (d, 1H), 3.92 (d, 2H), 3.82 (m, 2H), 3.21 (t, 2H), 2.33 (m, 1H), 1.91 (s, 3H), 1.60 (m, 2H), 1.29 (m, 2H).
  • TABLE N.3
    Examples prepared using general procedure N from Preparation #3
    Figure US20120053345A1-20120301-C00706
    Rt (min)
    Precursor Product Ex# (method) m/z
    1H-Indazol-5- ylamine
    Figure US20120053345A1-20120301-C00707
    N.3.1 3.38(a) 266(M + H)+
  • TABLE N.4
    Examples prepared using general procedure N from Example #F.8.1
    Figure US20120053345A1-20120301-C00708
    Rt (min)
    Aryl halide Product Ex# (method) m/z
    2,4,6- Trichloro- [1,3,5]triazine
    Figure US20120053345A1-20120301-C00709
    N.4.1 2.35(e) 411(M − H)
    2-Amino-6- chloro- pyrimidine-4- carboxylic acid methyl ester (J. Org. Chem., 1961, 26, 2755- 2763)
    Figure US20120053345A1-20120301-C00710
    N.4.2 1.77(e) 417(M + H)+
    4,6-Dichloro- pyrimidin-2- ylamine
    Figure US20120053345A1-20120301-C00711
    N.4.3 2.02(e) 391(M − H)
    4-Chloro-6- isopropyl- pyrimidin-2- yl amine
    Figure US20120053345A1-20120301-C00712
    N.4.4 1.72(e) 401.3(M + H)+ 398.9(M − H)
  • TABLE N.5
    Examples prepared using general procedure N from Example #N.4.3
    Figure US20120053345A1-20120301-C00713
    Rt (min)
    Amine Product Ex# (method) m/z
    N′,N′- Dimethyl- ethane-1,2- diamine
    Figure US20120053345A1-20120301-C00714
    N.5.1 1.23(e) 446 (M + H)+
    Diethyl-amine
    Figure US20120053345A1-20120301-C00715
    N.5.2 2.21(e) 431 (M + H)+
  • TABLE N.6
    Examples prepared using general procedure N from Example #N.4.1
    Figure US20120053345A1-20120301-C00716
    Rt (min)
    Precursor Product Ex# (method) m/z
    Diethylamine
    Figure US20120053345A1-20120301-C00717
    N.6.1 3.23(a) 485(M − H)−
  • TABLE N.7
    Examples prepared using general procedure N from Preparation #22d
    Figure US20120053345A1-20120301-C00718
    Example Rt (min) m/z
    Amine Product # (method) (ESI+)
    1H-Indazol-5- ylamine
    Figure US20120053345A1-20120301-C00719
    N.7.1 1.82(a) 340.9 (M + H)+
  • TABLE N.8
    Examples prepared using general procedure N from Preparation #3
    Figure US20120053345A1-20120301-C00720
    Example Rt (min) m/z
    Amine Precursor Product # (method) (ESI+)
    7- Benzo[b]thiphen- 2-yl-1H-indazol- 5-ylamine (Example #F.8.1)
    Figure US20120053345A1-20120301-C00721
    N.8.1 1.80(e) 398.1 (M + H)+
  • TABLE N.9
    Examples prepared using general procedure N from Example N.4.3
    Figure US20120053345A1-20120301-C00722
    Amine Example Rt (min) m/z
    Precursor Product # (method) (ESI+)
    2-Methoxy- ethylamine
    Figure US20120053345A1-20120301-C00723
    N.9.1 1.54(e) 432(M + H)+ 430(M − H)
    2-Morpholin-4- yl-ethylamine
    Figure US20120053345A1-20120301-C00724
    N.9.2 1.03(e) 487(M + H)+ 485(M − H)
    N*1*, N*1*- Dimethyl- ethane-1,2- diamine
    Figure US20120053345A1-20120301-C00725
    N.9.3 1.11(e) 445(M + H)+ 443(M − H)
    Diethyl-amine
    Figure US20120053345A1-20120301-C00726
    N.9.4 1.93(e) 430(M + H)+ 428(M − H)
    Isopropyl- amine
    Figure US20120053345A1-20120301-C00727
    N.9.5 1.69(e) 416(M + H)+ 414(M − H)
    2-(1H- Imidazol-4-yl)- ethylamine
    Figure US20120053345A1-20120301-C00728
    N.9.6 1.10(e) 468(M + H)+ 466(M − H)
    2-Amino- ethanol
    Figure US20120053345A1-20120301-C00729
    N.9.7 1.28(e) 418(M + H)+ 416(M − H)
    3-Amino- propanol
    Figure US20120053345A1-20120301-C00730
    N.9.8 1.44(e) 432(M + H)+ 430(M − H)
    Ethane-1,2- diamine
    Figure US20120053345A1-20120301-C00731
    N.9.9 1.31(e) 417(M + H)+ 415(M − H)
    2-Pyridin-3-yl- ethylamine
    Figure US20120053345A1-20120301-C00732
    N.9.10 1.53(e) 479(M + H)+ 477(M − H)
    Propyl-amine
    Figure US20120053345A1-20120301-C00733
    N.9.11
    Phenethylamine
    Figure US20120053345A1-20120301-C00734
    N.9.12 1.78(e) 478(M + H)+ 476(M − H)

    General Procedure O: Reductive Alkylation of an Amine with an Aldehyde or a Ketone.
  • Figure US20120053345A1-20120301-C00735
  • To a suspension of an amine and an aldehyde or a ketone (1-10 equivalents, preferably 1-2 equivalents) in an organic solvent (for example 1,2-dichloroethane, THF, CH2Cl2, DMF, or EtOAc, preferably 1,2-dichloroethane) with or without acetic acid (1-10 equivalents, preferably 1 equivalent) is added a reducing agent (for example, sodium triacetoxyborohydride or sodium cyanoborohydride, preferably sodium triacetoxyborohydride) (1-10 equivalents, preferably about 2 equivalents). Additional acetic acid (1-10 equivalents, preferably 3 equivalents) is added to progress the reaction when necessary. The resulting mixture is allowed to stir at room temperature for 2-112 hours (preferably 24 hours). The reaction mixture is purified in one of two ways: 1). The reaction solution is treated with an aqueous solution of an appropriate base (for example NaOH, NaHCO3, or Na2CO3, preferably NaHCO3) and an organic solvent (for example, CH2Cl2 or EtOAc, preferably CH2Cl2). The two layers are stirred for about 15 minutes then separated. The organic phase is concentrated under reduced pressure. The resulting crude product can be purified by trituration with an appropriate solvent (water, EtOH, toluene, or EtOAc, preferably EtOH) or by chromatography, or 2). The reaction mixture is directly concentrated under reduced pressure and purified by chromatography, trituration, or crystallization.
  • Illustration of General Procedure O Example #19a N-Benzyl-(5-nitro-1H-indazol-3-yl)-amine
  • Figure US20120053345A1-20120301-C00736
  • To a suspension of 5-nitro-1H-indazol-3-ylamine (0.025 g, 0.140 mmol) and benzaldehyde (0.067 g, 0.60 mmol) in CH2Cl2 (3.0 mL) was added acetic acid (0.024 mL, 0.42 mmol) followed by sodium triacetoxyborohydride (0.059 g, 0.28 mmol). The resulting solution was allowed to stir at room temperature for about 19 hours. The reaction solution was diluted with CH2Cl2 (5 mL) and treated with an aqueous solution of NaOH (1N, 5 mL). The two phases were separated and the organic solvent was removed under reduced pressure. The residue was further purified by preparative HPLC (Thermo Hypersil-Keystone 250×21.2 mm 8 Hypersil® HS C18 column; 5% acetonitrile/0.1 M aqueous ammonium acetate-100% acetonitrile over 20 min, 100% acetonitrile hold 5 minutes, 21 mL/min) to give benzyl-(5-nitro-1H-indazol-3-yl)-amine (0.012 g, 0.046 mmol) as an orange solid. RP-HPLC (Table 1, Method e) Rt 2.15 min.; m/z: (M+H)+ 269.
  • TABLE O.1
    Examples prepared from using general procedure O from Example #F.1.9
    Figure US20120053345A1-20120301-C00737
    Example Rt/min m/z
    Amine Product # (method) (ESI+)
    Dimethylamine
    Figure US20120053345A1-20120301-C00738
    O.1.1 1.36(e) 383.4 (M − H)
  • TABLE O.2
    Examples prepared using general procedure O from Example #F.8.1
    Figure US20120053345A1-20120301-C00739
    Example Rt/min m/z
    Aldehyde/Ketone Product # (method) (ESI+)
    4-(Imidazolo[1,2- a]pyridin-4-yl) benzaldehyde
    Figure US20120053345A1-20120301-C00740
    O.2.1 2.1(e) 472.4 (M + H)+
  • TABLE O.3
    Examples prepared using general procedure O from Example #F.5.4
    Figure US20120053345A1-20120301-C00741
    Example Rt/min
    Amine Precursor Product # (method) m/z
    Piperidine
    Figure US20120053345A1-20120301-C00742
    O.3.1 1.24(g) 456.4 (M + H)+
    N,N-Dimethyl- 1,2-ethanediamine
    Figure US20120053345A1-20120301-C00743
    O.3.2 1.06(g) 459.3 (M + H)+
    2-Methoxyethyl amine
    Figure US20120053345A1-20120301-C00744
    O.3.3 1.12(g) 446.3 (M + H)+
    N-(2- Aminoethyl) morpholine
    Figure US20120053345A1-20120301-C00745
    O.3.4 1.12(g) 501.4 (M + H)+
    Piperazine
    Figure US20120053345A1-20120301-C00746
    O.3.5 1.13(g) 457.3 (M + H)+
    1- Methylpiperazine
    Figure US20120053345A1-20120301-C00747
    O.3.6 1.14(g) 471.4 (M + H)+
    Morpholine
    Figure US20120053345A1-20120301-C00748
    O.3.7 1.13(g) 458.3 (M + H)+
    2-Propen-1-amine
    Figure US20120053345A1-20120301-C00749
    O.3.8 1.13(g) 428.4 (M + H)+
  • TABLE O.4
    Examples prepared using general procedure O from Example #5
    Figure US20120053345A1-20120301-C00750
    Example Rt/min
    Amine Precursor Product # (method) m/z
    2-Propen-1-amine
    Figure US20120053345A1-20120301-C00751
    O.4.1 1.05(g) 409.1 (M − H)
  • General Procedure P. Deprotection of a Methyl-Protected Alcohol Using Boron Tribromide
  • To a suspension of a methyl-protected alcohol in an organic solvent (1,2-dichloroethane or CH2Cl2, preferably 1,2-dichloroethane) is added a solution of BBr3 (8-20 equivalents, preferably 10 equivalents) in CH2Cl2. The reaction mixture is stirred at about −10 to 5° C. (preferably about 0° C.) for about 0.5-3 hours (preferably about 2 hours), then heated at about 30-100° C. (preferably about 80° C.) for about 2-8 hours (preferably about 3 hours). The reaction mixture can be treated in two different ways: 1). The reaction mixture is allowed to cool to about −10-10° C. (preferably about 0° C.) and quenched with an aqueous solution (for example, saturated aqueous NaHCO3 or Na2CO3, preferably NaHCO3) and extracted with an organic solvent (EtOAc, Et2O, or CH2Cl2, preferably CH2Cl2). The residue is partitioned between water and an organic solvent. The organic layer is separated and the aqueous layer further extracted with the organic solvent. The combined organic extracts are dried over a desiccant and the solvent removed under reduced pressure. 2). The reaction mixture can be cooled down to room temperature and diluted with MeOH then the solvents are removed under reduced pressure. The compound can be further purified by chromatography or crystallization.
  • Illustration of General Procedure P Example #19b 7-[5-(2-Amino-pyrimidin-4-ylamino)-1H-indazol-3-yl]-naphthalen-2-ol diacetate
  • Figure US20120053345A1-20120301-C00752
  • To a suspension of 4-[3-(7-methoxy-naphthalen-2-yl)-1H-indazol-5-ylamino]-pyrimidine-2-amine (Preparation #28 followed by general procedure N and F, 20 mg, 0.05 mmol) in CH2Cl2 (1 mL) was added 1M BBr3 in THF (1 mL). The mixture was stirred for about 2 hours at room temperature then methanol (1 mL) was added and the mixture concentrated under reduced pressure. The residue was purified by preparative reverse phase HPLC to give 7-[5-(2-amino-pyrimidin-4-ylamino)-1H-indazol-3-yl]-naphthalen-2-ol diacetate (12 mg, 60%); (DMSO-d6, 400 MHz) δ 9.08 (s, 1H), 8.35 (s, 1H), 8.30 (s, 1H), 8.03 (d, 1H), 7.89 (d, 1H), 7.80 (m, 2H), 7.72 (d, 1H), 7.53 (d, 1H), 7.16 (s, 1H), 7.11 (d, 1H), 6.06 (bs, 2H), 5.99 (d, 1H), 1.87 (s, 6H); RP-HPLC (Table 1, Method e) Rt 1.43 min; m/z: (M+H)+ 369.2.
  • TABLE P.1
    Examples synthesized using general procedure P
    Example Rt/min m/z
    Precursor Product # (method) (ESI+)
    4-[3-(5-Methoxy- benzo[b]thiophen- 2-yl)-1H-indazol- 5-ylamino]- pyrimidine-2- amine (Preparation #28, N, F)
    Figure US20120053345A1-20120301-C00753
    P.1.1 1.19(e) 375.3 (M + H)+
  • General Procedure Q: Acid Catalyzed Cleavage of Esters, Amidines, and Carbamates
  • An acid (HCl or TFA, preferably HCl) was added to an amine protected with a carbamate or amidine moiety optionally dissolved in an organic solvent (MeOH, CH2Cl2 or dioxane, preferably MeOH) at about 0-100° C., preferably 40° C. The reaction mixture is stirred for about 5 minutes to 24 hours (preferably 16 h) until the reaction had proceeded to completion as judged by TLC or HPLC analysis. Where necessary, additional acid is added to achieve complete conversion. The solvents are then removed under reduced pressure. The crude product can be treated in two different ways. 1) The material is purified by chromatography, trituration, or crystallization. 2) The material is treated with an aqueous solution of an appropriate base (for example NaOH, NaHCO3, or Na2CO3, preferably NaHCO3) and either collected by filtration or extracted into an organic solvent (for example, CH2Cl2, CH2Cl2/MeOH (9:1), or EtOAc, preferably CH2Cl2). The resulting crude product can be purified by chromatography, trituration, or crystallization.
  • Illustration of General Procedure Q Example 19c #48 7-Benzo[b]thiophen-2-yl-1H-indazole-5-carboxylic acid [4-(1H-indol-2-yl)phenyl]-amide
  • Figure US20120053345A1-20120301-C00754
  • TFA (0.078 g, 0.68 mmol) was added to a solution of 2-4-[(7-benzo[b]thiophen-2-yl-1H-indazole-5-carbonyl)-amino]-phenyl-indole-1-carboxylic acid tert-butyl ester (Preparation #7 then B, 0.100 g, 0.137 mmol) in CH2Cl2 (2.5 mL, 0.039 mol) at about 0° C. The reaction mixture was stirred at about 0° C. for about 30 min then additional TFA (80 uL) was added. After about 30 min the reaction was allowed to warm to room temperature and stirred for about 16 hours, prior to the addition of more TFA (150 uL). After 2 hours the mixture was concentrated under reduced pressure and the crude product was purified by RP-HPLC to yield 7-benzo[b]thiophen-2-yl-1H-indazole-5-carboxylic acid [4-(1H-indol-2-yl)-phenyl]-amide (13 mg, 20% yield): RP-HPLC (Table 1, Method e) Rt=2.4 min MS m/z: (M−H) 483.2.
  • TABLE Q.1
    Examples prepared using general procedure Q
    Ester/Carbamate Example Rt/min m/z
    Precursor Product # (method) (ESI+)
    4-{[(2-Amino- pyrimidin-4-yl)-(7- benzo[b]thiophen-2- yl-1H-indazol-5-yl)- amino]-methyl}- piperidine-1-carboxylic acid tert-butyl ester (Example #F.8.1, O, N)
    Figure US20120053345A1-20120301-C00755
    Q.1.1 1.78(e) 416 (M + H)+
    7-(1-tert- Butoxycarbonyl-1H- indazol-5-yl)-4- dimethylamino-2- (dimethylamino- methyleneamino)- pyrrolo[3,2- d]pyrimidine-5- carboxylic acid tert- butyl ester (J. Med. Chem., 2003, 46, 3060-3071, F)
    Figure US20120053345A1-20120301-C00756
    Q.1.2 0.82(e) 294.1 (M + H)+
    4-(7- Benzo[b]thiophen-2- yl-1H-indazol-5-yl)- pyrazole-1-carboxylic acid tert-butyl ester (Preparation #26, F)
    Figure US20120053345A1-20120301-C00757
    Q.1.3 2.00(e) 315.1 (M + H)+
    (3-{5-[5-(2- Aminopyrimidin-4- ylamino)-1H-indazol- 7-yl]-indol-1-yl}- propyl)-carbamic acid tert-butyl ester (Example# F.11.5, R)
    Figure US20120053345A1-20120301-C00758
    Q.1.4 1.15(g) 399.3 (M + H)+
    (2-{5-[5-(2- Aminopyrimidin-4- ylamino)-1H-indazol- 7-yl]-indol-1-yl}- ethyl)-carbamic acid tert-butyl ester (Example# F.11.5, R)
    Figure US20120053345A1-20120301-C00759
    Q.1.5 1.10(g) 385.3 (M + H)+
    (3-{6-[5-(2- Aminopyrimidin-4- ylamino)-1H-indazol- 7-yl] indol-1-yl}- propyl)-carbamic acid tert-butyl ester (Example# F.5.2, R)
    Figure US20120053345A1-20120301-C00760
    Q.1.6 1.09(g) 399.5 (M + H)+
  • General Procedure R: Base-Promoted Amine Alkylation
  • Figure US20120053345A1-20120301-C00761
  • A mixture of an amine substrate and a base (for example, NaH, K2CO3, Cs2CO3, t-BuOK or Na2CO3, preferably Na2CO3) (2-6 equivalents, preferably 2 equivalents) in an organic solvent (for example, THF, DME or DMF, preferably DMF) is stirred at about −10-25° C. (preferably about 0° C.), for about 0-60 minutes (preferably 40 minutes) under an inert atmosphere. An organic halide (for example an organic bromide or an organic chloride, preferably an organic bromide) (2-6 equivalents, preferably 3 equivalents) is added and the reaction is stirred for about 4-96 hours (preferably about 24 hours) at about 20-100° C. (preferably about 60° C.). The solvent is removed under reduced pressure and the crude product can be purified by chromatography, crystallization or used in the next step without further purification.
  • Illustration of General Procedure R Example #20 3-[7-[7-Benzo[b]thiophen-2-yl-1-(2-trimethylsilanyl-ethoxymethyl)-1H-indazol-5-yl]-2-(dimethylamino-methyleneamino)-pyrrolo[3,2-d]pyrimidin-5-yl]-propionic acid methyl ester
  • Figure US20120053345A1-20120301-C00762
  • To a mixture of {7-[7-benzo[b]thiophen-2-yl-1-(2-trimethylsilanyl-ethoxymethyl)-1H-indazol-5-yl]-5H-pyrrolo[3,2-d]pyrimidin-2-yl}-N,N-dimethyl-formamidine (Preparation #5, 240 mg, 0.42 mmol) in DMF (3 mL) at about 0° C. and under an inert atmosphere was added K2CO3 (140 mg, 1.02 mmol). The reaction mixture was stirred at about 0° C. for about 40 minutes, then 3-bromo-propionic acid methyl ester (0.12 mL, 1.02 mmol) was added. The reaction mixture was stirred at about 60° C. for about 24 hours, then the solvent was removed under reduced pressure and the residual material was used in the next step without further purification. LC/MS (Table 1, Method h) Rt 3.29 min; ESI-MS [M+H]+=654.4.
  • General Procedure S: Formation of a Sulfonamide from an Amine:
  • Figure US20120053345A1-20120301-C00763
  • To a mixture of an amine and a base (for example, pyridine, Et3N, or ethyl-diisopropyl-amine, preferably ethyl-diisopropyl-amine) (1-5 equivalents, preferably 1.5 equivalents) in an organic solvent (for example, CH2Cl2, DME, or DMF, preferably DMF) at about 0-50° C. (preferably about 20° C.) and under an inert atmosphere is added a sulfonylchloride (1-5 equivalents, preferably 1.05 equivalents). The reaction mixture is stirred for about 1-6 hours (preferably 2 hours), then concentrated under reduced pressure and purified by chromatography, crystallization or it can be used in the next step without further purification.
  • Illustration of General Procedure S Example #21 3-Oxo-3,4-dihydro-2H-benzo[1,4]oxazine-6-sulfonic acid (7-benzo[b]thiophen-2-yl-1H-indazol-5-yl)-amide
  • Figure US20120053345A1-20120301-C00764
  • To a mixture of 7-benzo[b]thiophen-2-yl-1H-indazol-5-ylamine (Example #F.8.1, 32 mg, 0.12) and ethyl-diisopropylamine (0.032 mL, 0.18 mmol) in DMF (1 mL) at room temperature and under an inert atmosphere was added 3-oxo-3,4-dihydro-2H-benzo[1,4]oxazine-6-sulfonyl chloride (33 mg, 0.13 mmol). The reaction mixture was stirred at room temperature for 2 hours then concentrated under reduced pressure. The crude material was purified by preparative HPLC (20% to 80% acetonitrile/0.05 M aqueous ammonium acetate, buffered to pH 4.5 over 30 min at 20 mL/min; Hyperprep C18, 300 Å, 8 μm, 250×21.1 mm column) to afford 3-oxo-3,4-dihydro-2H-benzo[1,4]oxazine-6-sulfonic acid (7-benzo[b]thiophen-2-yl-1H-indazol-5-yl)-amide as a white solid (34 mg, 58%). LC/MS (Table 1, Method e) Rt 1.75 min; ESI-MS [M+H]+=475.3.
  • TABLE S.1
    Examples synthesized using general procedure S from Example #F.8.1
    Figure US20120053345A1-20120301-C00765
    Figure US20120053345A1-20120301-C00766
    HPLC Rt m/z
    Precursor Product Example # (Method) (ESI+)
    Benzo[1,2,5] thiadiazole- 4-sulfonyl chloride
    Figure US20120053345A1-20120301-C00767
    S.1.1 2.00 (e) 362.1 (M − H)+
    5-Pyridin-2- yl- thiophene- 2-sulfonyl chloride
    Figure US20120053345A1-20120301-C00768
    S.1.2 2.11 (e) 487.2 (M − H)+
    Thiophene- 2-sulfonyl chloride
    Figure US20120053345A1-20120301-C00769
    S.1.3 1.98 (e) 410.2 (M − H)+
    5-Chloro- 1,3- dimethyl- 1H- pyrazole-4- sulfonyl chloride
    Figure US20120053345A1-20120301-C00770
    S.1.4 1.89 (e) 456.2 (M − H)+
    4-Methoxy- benzenesul- fonyl chloride
    Figure US20120053345A1-20120301-C00771
    S.1.5 2.07 (e) 134.2 (M − H)+
    1-Methyl- 1H- imidazole- 4-sulfonyl chloride
    Figure US20120053345A1-20120301-C00772
    S.1.6 1.57 (e) 408.0 (M − H)+
    Benzo[1,2,5] oxadiazole- 4-sulfonyl chloride
    Figure US20120053345A1-20120301-C00773
    S.1.7 2.01 (e) 446.0 (M − H)+
    Benzo[1,2,5] thiadiazole- 5-sulfonyl chloride
    Figure US20120053345A1-20120301-C00774
    S.1.8 2.06 (e) 462.2 (M − H)+
    Quinoline- 8-sulfonyl chloride
    Figure US20120053345A1-20120301-C00775
    S.1.9 2.05 (e) 455.1 (M − H)+
    4-Amino- benzenesul- fonyl chloride
    Figure US20120053345A1-20120301-C00776
    S.1.10 1.80 (e) 419.2 (M − H)+
    Isoquinoline- 5-sulfonyl chloride
    Figure US20120053345A1-20120301-C00777
    S.1.11 1.89 (e) 455.2 (M − H)+
    2-Amino-4- methyl- thiazole-5- sulfonyl chloride
    Figure US20120053345A1-20120301-C00778
    S.1.12 1.77 (e) 439.4 (M − H)+
    3,5- Dimethyl- isoxazole-4- sulfonyl chloride
    Figure US20120053345A1-20120301-C00779
    S.1.13 2.00 (e) 423.0 (M − H)+
    5-(2- Methyl- thiazol-4- yl)- thiophene- 2-sulfonyl chloride
    Figure US20120053345A1-20120301-C00780
    S.1.14 2.13 (e) 507.2 (M − H)+
    3-Oxo-3,4- dihydro-2H- benzo[1,4] oxazine-6- sulfonyl chloride
    Figure US20120053345A1-20120301-C00781
    S.1.15 1.75 (e) 475.3 (M − H)+
    1,2- Dimethyl- 1H- imidazole- 4-sulfonyl chloride
    Figure US20120053345A1-20120301-C00782
    S.1.16 1.59 (e) 422.0 (M − H)+
    4- Acetylamino- benzenesul- fonyl chloride
    Figure US20120053345A1-20120301-C00783
    S.1.17 1.72 (e) 461.3 (M − H)+
    2- Acetylamino- 4-methyl- thiazole-5- sulfonyl chloride
    Figure US20120053345A1-20120301-C00784
    S.1.18 1.75 (e) 482.1 (M − H)+
  • General Procedure T: Mitsunobu Coupling
  • Figure US20120053345A1-20120301-C00785
  • To a mixture of an alcohol (1-5 equivalents, preferably 3 equivalents) and PPh3 (1-5 equivalents, preferably 2 equivalents) in an organic solvent (for example, THF, DME or CH2Cl2, preferably THF) at about −10 to 20° C. (preferably about 0° C.) under an inert atmosphere is added a pyrrolo[3,2-d]pyrimidine (preferably 1 equivalent) followed by slow addition of a Mitsunobu coupling reagent (for example, diisopropyl azodicarboxylate, 1,1′-azobis(N,N-dimethylformamide) or diethyl azodicarboxylate, preferably diisopropyl azodicarboxylate) (2-4 equivalents, preferably 2 equivalents). The reaction mixture is stirred at about 0 to 60° C. (preferably at 20° C.) for about 1-6 hours (preferably 2 hours). The reaction mixture is then concentrated under reduced pressure and the residual crude product is purified by chromatography, crystallization or can be used in the next step without further purification.
  • Illustration of General Procedure T Example #23 N′-{7-[7-Benzo[b]thiophen-2-yl-1-(2-trimethylsilanyl-ethoxymethyl)-1H-indazol-5-yl]-5-isopropyl-5H-pyrrolo[3,2-d]pyrimidin-2-yl}-N,N-dimethylformamidine
  • Figure US20120053345A1-20120301-C00786
  • To a mixture of i-PrOH (0.023 uL, 0.3 mmol) and PPh3 (52.4 mg, 0.2 mmol) in THF at about 0° C. under an inert atmosphere was added {7-[7-benzo[b]thiophen-2-yl-1-(2-trimethylsilanyl-ethoxymethyl)-1H-indazol-5-yl]-5H-pyrrolo[3,2-d]pyrimidin-2-yl}-N,N-dimethylformamidine (Preparation 5a, 50 mg, 0.1 mmol) followed by the slow addition of diisopropyl azodicarboxylate (0.04 mL, 0.2 mmol). The reaction mixture was stirred at about 20° C. for about 4 hours. The reaction mixture was then concentrated, and utilized in the next step without further purification. LC/MS (Table 1, Method e) Rt 2.80 min; ESI-MS [M+H]+=555.4.
  • General Procedure U: Sonogashira Coupling of a Halide with an Acetylene Compound
  • Figure US20120053345A1-20120301-C00787
  • To a solution of a halide (preferably 1.0 equivalent) in an organic solvent (for example, DMF or piperidine, preferably DMF) is added a terminal acetylene (1.0-12.0 equivalents, preferably 1.2 equivalent), a palladium catalyst (for example, dichlorobis(triphenylphosphine) palladium(II), tetrakis (triphenylphosphine) palladium(0), preferably dichlorobis(triphenylphosphine) palladium(II)) (0.02-0.05 equivalent, preferably 0.05 equivalent), a base (for example, piperidine, triethylamine, preferably triethylamine) (0.03-0.06 equivalent, preferably 0.05 equivalent), and copper salt (for example copper (I) bromide, copper (I) iodide, preferably copper (I) iodide) (0.03-0.15 equivalent, preferably 0.10 equivalent). The reaction mixture is heated in a microwave reactor at about 80-130° C. (preferably about 120° C.) for about 2-40 minutes (preferably about 5 minutes). The mixture is allowed to cool to ambient temperature. The insoluble residue is removed by filtration, and the filtrate is concentrated under reduced pressure. The residual crude product is purified by chromatography or crystallization.
  • Illustration of General Procedure U Example #24 4-(7-Phenylethynyl-1H-indazol-5-ylamino)-pyrimidine-2-amine
  • Figure US20120053345A1-20120301-C00788
  • To a solution of N′4′-(7-bromo-1H-indazol-5-yl)-pyrimidine-2,4-diamine (Example #N.2.7, 0.060 g, 0.197 mmol) in DMF (1 mL) in a microwave tube was added phenylacetylene (0.028 ml, 0.256 mmol), copper (I) iodide (0.004 g, 0.020 mmol) and tetrakis (triphenylphosphine) palladium(0) (0.011 g, 0.009 mmol). The tube was sealed and the reaction mixture was heated in a microwave at about 120° C. for about 5 minutes. The mixture was allowed to cool to ambient temperature. The insoluble residue was removed by filtration and washed with DMF (2 mL). The filtrate was purified by preparative RP-HPLC (Rainin C18, 8 mm, 300 Å, 35 cm; 5-100% acetonitrile/0.1 M ammonium acetate over 20 min, 100% acetonitrile hold 10 minutes, 21 mL/min) to obtain 4-(7-phenylethynyl-1H-indazol-5-ylamino)-pyrimidine-2-amine as a white solid (0.015 g, 0.046 mmol); RP-HPLC (Table 1, Method e) Rt 1.70 min; m/z: (M+H)+327.2.
  • TABLE U.1
    Examples prepared using General Procedure U using 3-iodo-5-(2-
    aminopyrimidin-4-yl)amino-1H-indazole
    Figure US20120053345A1-20120301-C00789
    Figure US20120053345A1-20120301-C00790
    3-Iodo-5-(2-aminopyrimidin-4-yl)amino-1H-indazole was prepared from Preparation #28 via
    general procedure N using 2-amino-4-chloropyrimdine.
    Example HPLC Rt
    Acetylene Product # (Method) m/z
    Ethynyl- benzene
    Figure US20120053345A1-20120301-C00791
    U.1.1 2.20 (a) 327 (M + H)+
    3,3- Dimethyl- but-1-yne
    Figure US20120053345A1-20120301-C00792
    U.1.2 2.10 (a) 307 (M + H)+
  • TABLE U.2
    Examples prepared using General Procedure U from Example #N.2.7
    Figure US20120053345A1-20120301-C00793
    Figure US20120053345A1-20120301-C00794
    Example Rt/min m/z
    Acetylene Precursor Product # (method) (ESI+)
    Hex-5-ynenitrile
    Figure US20120053345A1-20120301-C00795
    U.2.1 1.27 (e) 318.2 (M + H)+
    Pent-4-yn-1-ol
    Figure US20120053345A1-20120301-C00796
    U.2.2 0.70 (e) 309.0 (M + H)+
    Dimethyl-prop-2- ynyl-amine
    Figure US20120053345A1-20120301-C00797
    U.2.3 1.05 (e) 308.0 (M + H)+
    2-Methyl-but-3-yn-2- ol
    Figure US20120053345A1-20120301-C00798
    U.2.4 0.73 (e) 307.0 (M + H)+
    Prop-2-yn-1-ol
    Figure US20120053345A1-20120301-C00799
    U.2.5 0.53 (e) 278.9 (M + H)+
    Ethynyl-benzene
    Figure US20120053345A1-20120301-C00800
    U.2.6 1.70 (e) 327.2 (M + H)+
    Methyl-prop-2-ynyl- amine
    Figure US20120053345A1-20120301-C00801
    U.2.7 0.53 (e) 294.0 (M + H)+
    3-Ethynyl-phenol
    Figure US20120053345A1-20120301-C00802
    U.2.8 1.20 (e) 343.0 (M + H)+
  • General Procedure V: Hydrolysis of an Ester to a Carboxylic Acid
  • Figure US20120053345A1-20120301-C00803
  • To a solution of an ester in an organic solvent (for example, THF, MeOH, or 1,4-dioxane, preferably 1,4-dioxane) at about 10-50° C. (preferably about 25° C.) is added an aqueous base solution (for example, Na2CO3, NaOH or KOH, preferably Na2CO3) (3-20 equivalents, preferably 5 equivalents). The mixture is stirred at about 10-80° C. (preferably about 25° C.) for about 0.5-2 hours (preferably about 1 hour). The mixture is acidified with an acid (preferably 1M HCl solution) to about pH 3. If a precipitation is formed it is filtered off and washed with water. If no precipitation is formed the solvent is removed under reduced pressure and the residue is partitioned between an aqueous acidic solution (for example HCl) and an organic solvent (for example EtOAc or CH2Cl2, preferable CH2Cl2). The organic layer is separated and the aqueous layer is further extracted with an organic solvent. The combined organic extracts are dried over a desiccant. The solvents are removed under reduced pressure to afford the product which can be further purified by crystallization or chromatography.
  • Illustration of General Procedure V Example #25 {[5-(2-Amino-pyrimidin-4-ylamino)-1H-indazole-3-carbonyl]-amino}-acetic acid
  • Figure US20120053345A1-20120301-C00804
  • A solution of {[5-(2-amino-pyrimidin-4-ylamino)-1H-indazole-3-carbonyl]-amino}acetic acid methyl ester (was prepared from Example N.2.8, B, 0.014 g, 0.043 mmol) and 2M aqueous NaOH solution (1.0 mL, 2.0 mmol) in 1,4-dioxane (2 mL) was stirred at ambient temperature for about 30 minutes. The reaction mixture was acidified with 1M aqueous HCl solution until pH 3. The white precipitation was filtered off, washed with water (3 mL) and dried under reduced pressure to give {[5-(2-amino-pyrimidin-4-ylamino)-1H-indazole-3-carbonyl]-amino}-acetic acid (0.001 g, 0.003 mmol); RP HPLC (Table 1, Method a) Rt=0.42 min; m/z: (M+H)+ 328.2.
  • TABLE V.1
    Examples prepared using general procedure V
    m/z
    (ESI+) or
    NMR (400
    Example Rt/min MHz) (d6-
    Ester Precursor Product # (method) DMSO)
    3-{[5-(2-Amino- pyrimidin-4-ylamino)- 1H-indazole-3- carbonyl]-amino}- benzoic acid ethyl ester (Example #N.2.8, B)
    Figure US20120053345A1-20120301-C00805
    V.1.1 6.90 (j) 13.8 (br, 1H), 10.51 (s, 1H), 9.67 (m, 1H), 8.58 (s, 1H), 8.22 (m, 1H), 8.08 (d, 1H), 7.89 (d, 1H), 7.82 (d, 1H), 7.64 (m, 2H), 7.47 (t, 1H), 6.62 (m, 2H), 6.11 (d, 1H).
    5-Bromo-1H- indazole-7-carboxylic acid methyl ester
    Figure US20120053345A1-20120301-C00806
    V.1.2 0.48 (e) 239/241 (M − H)
    2-[5-(2-Amino- pyrimidin-4- ylamino)-1H-indazol- 7-yl]-1H-indole-7- carboxylic acid methyl ester (Example #F.5.11)
    Figure US20120053345A1-20120301-C00807
    V.1.3 0.8 (e) 383.9 (M − H)−
    2-Amino-6-(7- benzo[b]thiophen-2- yl-1H-indazol-5- ylamino)-pyrimidine- 4-carboxylic acid methyl ester (Example #N.4.2 )
    Figure US20120053345A1-20120301-C00808
    V.1.4 1.32 (e) 403 (M + H)+

    General Procedure W: Amide Formation from an Acid Chloride and an Amine
  • Figure US20120053345A1-20120301-C00809
  • To a solution of an acid chloride and an amine (preferably 1 equivalent) in an organic solvent (for example, pyridine, CH2Cl2 or 1,2-dichloroethane, preferably CH2Cl2) is added a base (for example Et3N or pyridine, preferably pyridine). The reaction mixture is stirred at about 0-50° C. (preferably about 25° C.) for about 0.5-20 hours (preferably about 1 hour). The mixture is diluted with an organic solvent (for example, EtOAc or CH2Cl2, preferably CH2Cl2) and washed with water, then dried under reduced pressure. The product can be further purified by crystallization or chromatography.
  • Illustration of General Procedure W Example #26 3-Dimethylamino-N-(5-nitro-1H-indazol-3-yl)-benzamide
  • Figure US20120053345A1-20120301-C00810
  • To a solution of 5-nitro-1H-indazol-3-ylamine in pyridine (Ryan Scientific, 0.150 g, 0.838 mmol) cooled to about 0° C. in an ice bath, was added 3-dimethylamino-benzoyl chloride (0.154 g, 0.838 mmol). The reaction mixture was stirred as it warmed to ambient temperature. After about 1 hour it was poured into crushed ice, then extracted with EtOAc (10 mL). The organic solvent portion was washed with a 1M aqueous HCl solution, then concentrated and dried under reduced pressure. The residue was further purified via preparative RP-HPLC (Rainin C18, 8 mm, 300 Å, 35 cm; 5-100% acetonitrile/0.1 M ammonium acetate over 20 min, 100% acetonitrile hold 10 minutes, 21 mL/min) to obtain 3-dimethylamino-N-(5-nitro-1H-indazol-3-yl)-benzamide as a white solid (0.002 g, 0.006 mmol); RP-HPLC (Table 1, Method e) Rt 1.95 min; m/z: (M+H)+ 326.4.
  • TABLE W.1
    Examples prepared from 5-nitro-1H-indazol-3-ylamine using General
    Procedure W
    Figure US20120053345A1-20120301-C00811
    Figure US20120053345A1-20120301-C00812
    Acid chloride Example Rt/min m/z
    Precursor Product # (method) (ESI+)
    3-Dimethylamino- benzoyl chloride
    Figure US20120053345A1-20120301-C00813
    W.1 1.95 (e) 326.4 (M + H)+
    Benzoyl chloride
    Figure US20120053345A1-20120301-C00814
    W.2 1.83 (e) 281 (M − H)
  • TABLE W.2
    Examples prepared from Example # F.2.16 using general procedure W
    Figure US20120053345A1-20120301-C00815
    Figure US20120053345A1-20120301-C00816
    Example Rt/min m/z
    Acid chloride Product # (method) (ESI+)
    Acetyl chloride
    Figure US20120053345A1-20120301-C00817
    W.2.1 0.92 (e) 360.6 (M + H)+
    Benzoyl chloride
    Figure US20120053345A1-20120301-C00818
    W.2.2 1.65 (e) 422.1 (M + H)+
    Cyclobutane carbonyl chloride
    Figure US20120053345A1-20120301-C00819
    W.2.3 1.55 (e) 400.3 (M + H)+
  • TABLE W.3
    Preparation of oxalates from Preparation #15 using general procedure W
    Figure US20120053345A1-20120301-C00820
    Figure US20120053345A1-20120301-C00821
    HPLC m/z or 1H NMR
    Example Rt (d6-DMSO, 400
    Amine Product # (Method) MHz)
    2-Methoxy- ethylamine
    Figure US20120053345A1-20120301-C00822
    W.3.1 1.84 (e) 395 (M + H)+
    N,N,N′-Trimethyl- ethane-1,2-diamine
    Figure US20120053345A1-20120301-C00823
    W.3.2 1.34 (a) 422 (M + H)+
    Pyridin-2-ylamine
    Figure US20120053345A1-20120301-C00824
    W.3.3 2.23 (a) 414 (M + H)+
    Aniline
    Figure US20120053345A1-20120301-C00825
    W.3.4 2.44 (a) 411 (M − H)
    Ammonia
    Figure US20120053345A1-20120301-C00826
    W.3.5 1.66 (a) 337 (M + H)+
    Morpholine
    Figure US20120053345A1-20120301-C00827
    W.3.6 1.80 (a) 407 (M + H)+
    Methylamine
    Figure US20120053345A1-20120301-C00828
    W.3.7 1.86 (a) 351 (M + H)+
    Dimethyl-amine
    Figure US20120053345A1-20120301-C00829
    W.3.8 1.74 (a) 363 (M − H)
    1-[2-(1-piperidine)- 1-oxo-ethyl] piperazine
    Figure US20120053345A1-20120301-C00830
    W.3.9 NMR (DMSO): 1.58-1.41 (m, 8H), 3.21 (s, 2H), 3.40-3.55 (m, 10H), 7.44 (m, 2H), 7.91-8.25(m, 6H).
    Cyclohexylamine
    Figure US20120053345A1-20120301-C00831
    W.3.10 2.98 (k) 417 (M − H)
    1-(3-Amino- propyl)-pyrrolidin- 2-one
    Figure US20120053345A1-20120301-C00832
    W.3.11 2.28 (k) 460 (M − H)
    Cyclopropyl- methylamine
    Figure US20120053345A1-20120301-C00833
    W.3.12 2.65 (k) 389 (M − H)
    Tetrahydro-pyran- 4-ylamine
    Figure US20120053345A1-20120301-C00834
    W.3.13 2.35 (k) 419 (M − H)
    [2,2′]-Bithiophenyl- 5-yl-methylamine
    Figure US20120053345A1-20120301-C00835
    W.3.14 3.09 (k) 513 (M − H)
    Pyridin-4-ylamine
    Figure US20120053345A1-20120301-C00836
    W.3.15 2.48 (k) 412 (M − H)
    Pyridin-3-ylamine
    Figure US20120053345A1-20120301-C00837
    W.3.16 2.42 (k) 412 (M − H)
    Naphthalen-2-yl- methylamine
    Figure US20120053345A1-20120301-C00838
    W.3.17 3.02 (k) 475 (M − H)
    Pyrimidin-5- ylamine
    Figure US20120053345A1-20120301-C00839
    W.3.18 2.27 (k) 413 (M − H)
    2-Ethoxy- ethylamine
    Figure US20120053345A1-20120301-C00840
    W.3.19 1.93 (k) 407 (M − H)
    1-Pyrazin-2-yl- piperazine
    Figure US20120053345A1-20120301-C00841
    W.3.20 2.28 (k) 482 (M − H)
    1-Isopropyl- piperazin-2-one
    Figure US20120053345A1-20120301-C00842
    W.3.21 2.22 (k) 460 (M − H)
    4-Pyrimidin-2- yloxy-piperidine
    Figure US20120053345A1-20120301-C00843
    W.3.22 2.62 (k) 497 (M − H)
    3-Imidazol-1-yl- propylamine
    Figure US20120053345A1-20120301-C00844
    W.3.23 2.45 (k) 443 (M − H)
    (1-Methyl-1H- pyrrol-2-yl)- methylamine
    Figure US20120053345A1-20120301-C00845
    W.3.24 2.63 (k) 428 (M − H)
    1-Methyl- piperazine
    Figure US20120053345A1-20120301-C00846
    W.3.25 2.19 (k) 418 (M − H)
    N-Methyl-(2- pyridin-2- yl)ethylamine
    Figure US20120053345A1-20120301-C00847
    W.3.26 2.28 (k) 454 (M − H)
    N,N-Diethyl- piperidine-3- carboxamide
    Figure US20120053345A1-20120301-C00848
    W.3.27 2.32 (k) 502 (M − H)
    2-(2-Methyl- benzimidazol-1-yl)- ethylamine
    Figure US20120053345A1-20120301-C00849
    W.3.28 2.35 (k) 493 (M − H)
    5-Methyl-pyrazin- 2-ylmethylamine
    Figure US20120053345A1-20120301-C00850
    W.3.29 2.27 (k) 441 (M − H)
    Pyrazin-2-ylamine
    Figure US20120053345A1-20120301-C00851
    W.3.30 2.48 (k) 413 (M − H)
    N,N-Dimethyl- ethane-1,2-diamine
    Figure US20120053345A1-20120301-C00852
    W.3.31 2.36 (k) 406 (M − H)
    1-Morpholin-4-yl- 2-piperazin-1-yl- ethanone
    Figure US20120053345A1-20120301-C00853
    W.3.32 2.09 (k) 531 (M − H)
    2-(1-Methyl-1H- pyrrol-2-yl)- ethylamine
    Figure US20120053345A1-20120301-C00854
    W.3.33 2.69 (k) 442 (M − H)
    Pyridin-2-yl- methylamine
    Figure US20120053345A1-20120301-C00855
    W.3.34 2.37 (k) 426 (M − H)
    Pyridin-3-yl- methylamine
    Figure US20120053345A1-20120301-C00856
    W.3.35 2.28 (k) 426 (M − H)
    Benzo[1,3]dioxol- 5-yl-methylamine
    Figure US20120053345A1-20120301-C00857
    W.3.36 2.67 (k) 469 (M − H)
    Benzothiazol-6- ylamine
    Figure US20120053345A1-20120301-C00858
    W.3.37 2.70 (k) 468 (M − H)
    2,5-Dimethyl-2H- pyrazol-3-ylamine
    Figure US20120053345A1-20120301-C00859
    W.3.38 2.33 (k) 429 (M − H)
    Pyridin-4-yl- methylamine
    Figure US20120053345A1-20120301-C00860
    W.3.39 2.28 (k) 426 (M − H)
  • General Procedure X: Indazole Formation Using Hydrazine
  • Figure US20120053345A1-20120301-C00861
  • A mixture of an ortho-halobenzonitrile (1 equivalent) in 30% aqueous hydrazine (1-10 equivalents, preferably about 3 equivalents) is heated in a sealed tube to about 50-200° C. (preferably about 120° C.) for about 30-120 minutes (preferably about 45 minutes). The mixture is cooled to ambient temperature and the precipitation is filtered off, washed with water and dried under reduced pressure to yield the desired indazole.
  • Illustration of General Procedure X Preparation #49 7-Nitro-1H-indazol-3-ylamine
  • Figure US20120053345A1-20120301-C00862
  • A mixture of 5-nitro-2-fluoro-benzonitrile (Aldrich, 1.00 g, 6.02 mmol) in 30% aqueous hydrazine (5.00 mL, 18.1 mmol) was heated to about 120° C. in a sealed tube for about 40 minutes. The mixture was cooled to ambient temperature and the precipitate was filtered off, washed with water (10 mL) and dried under reduced pressure to yield 7-nitro-1H-indazol-3-ylamine (0.470 g, 26.4 mmol); (DMSO-d6, 400 MHz) δ 12.16 (br, 1H), 8.90 (m, 1H), 8.05 (dd 1H), 7.35 (d, 1H), 5.98 (br, 2H); RP-HPLC (Table 1, Method j) Rt 8.07 min.
  • TABLE X.1
    Examples prepared using general procedure X
    Figure US20120053345A1-20120301-C00863
    Figure US20120053345A1-20120301-C00864
    m/z
    or 1H
    NMR (d6
    DMSO,
    Benzonitrile Rt/min 400
    Precursor Product Example # (method) MHz)
    2-Fluoro-6- iodo- benzonitrile
    Figure US20120053345A1-20120301-C00865
    X.1.1 1.24 (e) 260 (M + H)+
    2-Fluoro-5- bromo- benzonitrile
    Figure US20120053345A1-20120301-C00866
    X.1.2 1.78 (e) 210/212 (M − H)
    2-Fluoro-6- bromo- benzonitrile
    Figure US20120053345A1-20120301-C00867
    X.1.3 1.82 (e) 210/212 (M − H)

    General Procedure Y: Pd Mediated Coupling of an Aryl Halide with an Amine Followed by Acid Deprotection
  • To a mixture of an amine (1-5 equivalents, preferably 1.25 equivalents), an aryl halide (for example, an aryl bromide, aryl chloride or an aryl iodide, preferably an aryl iodide) (0.7-3 equivalents, preferably 1 equivalent) and an inorganic base (for example, KF, Na2CO3 or Cs2CO3, preferably Cs2CO3) (2-16 equivalents, preferably 2.5 equivalents) in a degassed organic solvent (for example THF, DME, DMF, 1,4-dioxane, toluene, preferably 1,4-dioxane) is added a palladium catalyst (for example tris(benzylideneacetone)dipalladium (0) and XANTPHOS, tetrakis(triphenylphosphine)palladium(0), bis(acetato)triphenylphosphinepalladium(II) (˜5% Pd) polymer-bound FibreCat™ or [1,1′-bis(diphenylphosphino)ferrocene] dichloropalladium(II), complex with dichloromethane, preferably tris(benzylideneacetone)dipalladium (0) and XANTPHOS) (0.01-0.10 equivalents, preferably 0.05 equivalents). The reaction mixture is heated at about 40-150° C. (preferably about 95° C.) for about 0.5-24 hours (preferably about 2 hours) or at about 100-200° C. (preferably 150° C.) for about 5-60 minutes (preferably about 15 minutes) in a microwave under an inert atmosphere. The reaction mixture is allowed to cool and the crude product is treated with an acid (preferably trifluoroacetic acid) (5-80 M solution, preferably 20M solution) optionally containing a cation scavenger (preferably triisopropyl silane) (0.7-2.5 equivalents, preferably 1.2 equivalents) for about 10-150 minutes (preferably about 15 minutes) at about 70-150° C. (preferably about 100° C.) in a sealed tube, either by conventional heating or by microwave heating. Solvents are removed under reduced pressure to give the product that can be further purified by crystallization or chromatography.
  • Illustration of General Procedure Y Example #27 N-Phenyl-5-(pyridin-4-ylamino)-1H-indazole-7-carboxamide
  • Figure US20120053345A1-20120301-C00868
  • A mixture of 5-bromo-2-(4-methoxy-benzyl)-2H-indazole-7-carboxylic acid, phenylamide (prepared from preparation #12a using general procedures V and B, 75.0 mg, 0.20 mmol), 4-aminopyridine (24.0 mg, 0.25 mmol), cesium carbonate (195 mg, 0.60 mmol), and XANTPHOS (11.6 mg, 0.02 mmol) was suspended in 1,4-dioxane (2.5 mL) at ambient temperature under an inert atmosphere. Tris(dibenzylideneacetone)dipalladium(0) (9.2 mg, 0.01 mmol) was added and nitrogen gas was bubbled through the resulting suspension for about 5 minutes. The reaction mixture was heated at about 95° C. for about 2 hours. The resulting mixture was allowed to cool to ambient temperature and filtered through a celite pad and the solvent was removed under reduced pressure. The residue was dissolved in trifluoroacetic acid (4.0 mL) containing triisopropylsilane (51.0 uL, 0.25 mmol) and the mixture was heated at about 100° C. in a microwave reactor for about 15 minutes. Solvents were removed under reduced pressure and the residue was purified preparative RP-HPLC (Rainin C18, 8 mm, 300 Å, 35 cm; 5-100% acetonitrile/0.1 M ammonium acetate over 20 min, 100% acetonitrile hold 10 minutes, 21 mL/min) to afford N-phenyl-5-(pyridin-4-ylamino)-1H-indazole-7-carboxamide (30 mg, 46%) as an off-white solid, RP HPLC (Table 1, Method e) Rt=1.32; MS m/z: (MH)+ 330.
  • TABLE Y.1
    Examples synthesized using general procedure Y
    m/z or 1H
    NMR (d6
    Example HPLC Rt DMSO, 400
    Amine Product # (Method) MHz)
    5-Bromo-1- (4-methoxy- benzyl)-1H- indazole-7- carboxylic acid phenylamide (preparation #11d, V and B)
    Figure US20120053345A1-20120301-C00869
    Y.1.1 1.37 (e) 345 (MH)+
  • TABLE Y.2
    Examples prepared using general procedure Y from Preparation #F.8.1 (4-
    methoxybenzylated using the conditions used to prepare Prepn. 11d and 12a)
    Figure US20120053345A1-20120301-C00870
    Figure US20120053345A1-20120301-C00871
    m/z or 1H
    NMR (d6
    Heterocyclic Example HPLC Rt DMSO, 400
    halide Product # (Method) MHz)
    4-Chloro-5- methyl- isoxazole
    Figure US20120053345A1-20120301-C00872
    Y.2.1 2.1 min (c) 347.2 (M + H)+
  • General Procedure Z: Acid Cleavage of a THP-Protecting Group
  • Figure US20120053345A1-20120301-C00873
  • To a mixture of a THP protected alcohol in an organic solvent (for example MeOH, EtOH, i-PrOH or dioxane, preferably MeOH) is added an acid (for example aqueous HCl or PPTS, preferably PPTS) (0.1-1.0 equivalents, preferably 0.5 equivalents). The reaction mixture is stirred at about 10-85° C. (preferably about 25° C.) for about 1-24 hours (preferably about 6 hours). The reaction mixture was diluted with an organic solvent (for example EtOAc, Et2O, or CH2Cl2, preferably EtOAc) and was washed with an aqueous basic solution (for example aqueous Na2CO3 or NaOH, preferably Na2CO3). The solvent is evaporated and the product can be further purified by crystallization or chromatography.
  • Illustration of General Procedure Z Example #28 (E)-4-[5-(2-Amino-pyrimidin-4-ylamino)-1H-indazol-7-yl]-but-3-en-1-ol
  • Figure US20120053345A1-20120301-C00874
  • PPTS (0.019 g, 0.075 mmol) was added to a mixture of 4-{7-[(E)-4-(tetrahydropyran-2-yloxy)-but-1-enyl]-1H-indazol-5-ylamino}-pyrimidine-2-amine (prepared from Example #N.2.7 reacted with trans-2-[3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)allyoxy]-tetrahydropyran using general procedure F, 0.037 g, 0.100 mmol) in MeOH (3.0 mL). The reaction mixture was stirred at about 25° C. for about 5 hours. The reaction mixture was diluted with EtOAc (5 mL) and was washed with aqueous Na2CO3 solution (5 mL). The organic portion was evaporated under reduced pressure to afford (E)-4-[5-(2-aminopyrimidin-4-ylamino)-1H-indazol-7-yl]-but-3-en-1-ol (0.015 g, 0.051 mmol) as an off white solid; RP-HPLC (Table 1, Method e) Rt 0.62 min; m/z: (M+H)+297.0.
  • General Procedure AA: Deprotection of a Cbz-Protected Amino Group
  • Figure US20120053345A1-20120301-C00875
  • A solution of a Cbz protected amine substrate and formic acid (5-20 equivalents, preferably 10 equivalents) in an organic solvent (for example methanol or ethanol, preferably methanol) is added dropwise to a suspension of Palladium black (1-10 equivalents, preferably 2 equivalents) in an organic solvent (for example methanol or ethanol, preferably methanol). The resulting mixture is allowed to stir at room temperature for about 2-20 hours (preferably 16 hours). The suspension is filtered though a plug of celite and the solvent is removed under reduced pressure. The resulting crude product can be purified by titration with an appropriate solvent (water, ethanol, toluene or ethyl acetate, preferably ethanol) or by chromatography.
  • Illustration of General Procedure AA Example #29 N4-(3-amino-propyl)-N4-(7-benzo[b]thiophen-2-yl-1H-indazol-5-yl)-pyrimidine-2,4-diamine
  • Figure US20120053345A1-20120301-C00876
  • A solution of {3-[(2-amino-pyrimidin-4-yl)-(7-benzo[b]thiophen-2-yl-1H-indazol-5-yl)-amino]-propyl}-carbamic acid benzyl ester (prepared from Example #F.8.1 using general procedures O and N, 0.169 g, 0.308 mmol) and formic acid (0.116 mL, 3.08 mmol) in methanol (2 mL) was added dropwise to a suspension of Palladium black (0.080 g, 0.752 mmol) in methanol (1.5 mL). The resulting mixture was allowed to stir at ambient temperature for about 19 hours. The resulting suspension was filtered though celite and the solvent was removed under reduced pressure. The resulting crude product was purified by reverse chromatography (Thermo Hypersil-Keystone 250×21.2 mm 8μ Hypersil® HS C18 column; 5% acetonitrile/0.1 M aqueous ammonium acetate-100% acetonitrile over 20 min, 100% acetonitrile hold 5 minutes, 21 mL/min) to afford N4-(3-amino-propyl)-N4-(7-benzo[b]thiophen-2-yl-1H-indazol-5-yl)-pyrimidine-2,4-diamine (0.021 g, 17% yield) as a green solid. RP-HPLC (Table 1, Method e) Rt 1.62 min.; m/z: (M+H)+ 416.1.
  • SPECIFIC EXAMPLES AND INTERMEDIATE PREPARATIONS Example #1 2-Amino-4-(1H-indazol-5-ylamino)-thieno[2,3-d]pyrimidine-6-carboxylic acid amide
  • Figure US20120053345A1-20120301-C00877
  • A mixture of 2-amino-4-(1H-indazol-5-ylamino)-thieno[2,3-d]pyrimidine-6-carboxylic acid methyl ester (Example #N.7.1, 0.050 g, 0.15 mmol) and ammonia (ca. 7N in methanol, 1.0 mL) was heated at about 60° C. in a sealed vessel for about 1.5 hours. The mixture was allowed to cool to ambient temperature and additional ammonia (ca. 7N in methanol, 5.0 mL) was added. The reaction mixture was then heated at about 75° C. for about 18 hours, cooled to ambient temperature, additional ammonia (ca. 7N in methanol, 2.0 mL) was added, and heating at about 75° C. was continued for about another 3 days. The mixture was concentrated under reduced pressure and the residue was purified by preparative HPLC (Thermo Hypersil-Keystone 250×21.2 mm 8μ Hypersil® HS C18 column; 5% CH3CN/50 mM aqueous ammonium acetate hold for 5 min, 5-100% CH3CN/50 mM aqueous ammonium acetate over 30 min, hold at 100% CH3CN for 5 minutes, 21 mL/min) to give 2-amino-4-(1H-indazol-5-ylamino)-thieno[2,3-d]pyrimidine-6-carboxylic acid amide (0.015 g, 31%) as a beige solid; RP-HPLC (Table 1, Method a) Rt 0.68 min, m/z (ESI+) 325.9 (M+H)+.
  • Example #2 1-Benzyl-3-(3-iodo-1H-indazol-5-yl)-urea
  • Figure US20120053345A1-20120301-C00878
  • Benzyl isocyanate (0.25 mL, 2.0 mmol) was added to 3-iodo-1H-indazol-5-ylamine (Preparation #28, 0.50 g, 1.9 mmol) in THF (10 mL). After stirring at ambient temperature for about 2.5 hours, the mixture was filtered and washed with Et2O to give 1-benzyl-3-(3-iodo-1H-indazol-5-yl)-urea (0.41 g, 55%) as an ivory solid; RP-HPLC (Table 1, Method e) Rt 0.98 min, m/z (ESI+) 393.0 (M+H)+.
  • Example #3 2-Amino-4-(7-benzo[b]thiophen-2-yl-1H-indazol-5-ylamino)-7H-pyrrolo[2,3-d]pyrimidine-5-carboxamide
  • Figure US20120053345A1-20120301-C00879
  • Sodium percarbonate (0.040 g, 0.25 mmol) was added to a mixture of 2-amino-4-(7-benzo[b]thiophen-2-yl-1H-indazol-5-ylamino)-7H-pyrrolo[2,3-d]pyrimidine-5-carbonitrile (prepared via General Procedure N from the reaction of Example #F.8.1 with 6-amino-4-chloro-3-cyano-pyrrolo[2,3-d]pyrimidine [prepared by chlorination of the 4-oxo derivative (J. Med. Chem., 2001, 44(12), 1993-2003) using POCl3 (Tet., 2004, 60, 943-959), 0.10 g, 0.24 mmol) in 1N KOH (2.4 mL) at ambient temperature. After about 21 hours, the reaction mixture was acidified with 5N HCl and filtered. The resulting solid was purified by preparative HPLC (Thermo Hypersil-Keystone 250×21.2 mm 8μ Hypersil® HS C18 column; 5% CH3CN/50 mM aqueous ammonium acetate hold for 5 min, 5-100% CH3CN/50 mM aqueous ammonium acetate over 30 min, hold at 100% CH3CN for 5 minutes, 21 mL/min) to give an impure solid that was triturated with heptane and filtered to give 2-amino-4-(7-benzo[b]thiophen-2-yl-1H-indazol-5-ylamino)-7H-pyrrolo[2,3-d]pyrimidine-5-carboxamide (6.9 mg, 7%); RP-HPLC (Table 1, Method e) Rt 1.64 min; m/z (ESI+): 441.0 (M+H)+.
  • Example #4 3-(7-Benzo[b]thiophen-2-yl-1H-indazol-5-ylamino)-4-methoxy-cyclobut-3-ene-1,2-dione
  • Figure US20120053345A1-20120301-C00880
  • A vial containing a solution of 7-benzo[b]thiophen-2-yl-1H-indazol-5-ylamine (Example #F.8.1, 0.020 g, 0.075 mmol), 3,4-dimethoxy-3-cyclobutene-1,2-dione (0.011 g, 0.077 mmol), N,N-diisopropylethylamine (14 μL, 0.080 mmol) and MeOH (1.5 mL) was shaken at ambient temperature for about 16 hours then filtered to give 3-(7-benzo[b]thiophen-2-yl-1H-indazol-5-ylamino)-4-methoxy-cyclobut-3-ene-1,2-dione (0.022 g, 78%) as a tan solid; RP-HPLC (Table 1, Method e) Rt 1.80 min, m/z (ESI) 375.9 (M+H)+.
  • Example #5 5-[5-(2-Amino-pyrimidin-4-ylamino)-1H-indazol-7-yl]-1H-indole-3-carboxaldehyde diacetate
  • Figure US20120053345A1-20120301-C00881
  • Phosphoryl chloride (43 μL, 0.46 mmol) was added dropwise to DMF (0.17 mL) at about 0° C. After about 20 min, a solution of 5-(4,4,5,5-tetramethyl-[1,3,2]dioxaborolan-2-yl)-1H-indole (Aldrich, 0.10 g, 0.42 mmol) in DMF (0.70 mL) was added dropwise. The reaction was allowed to warm slowly to ambient temperature. After about 7.5 hours, the reaction mixture was concentrated under reduced pressure to give a crude mixture containing 5-(4,4,5,5-tetramethyl-[1,3,2]dioxaborolan-2-yl)-1H-indole-3-carboxaldehyde. The mixture was dissolved in DME (3.0 mL) and transferred to a microwave vial. N4-(7-bromo-1H-indazol-5-yl)-pyrimidine-2,4-diamine (Example #N.2.7, 0.10 g, 0.34 mmol), Pd(PPh3)4 (0.039 g, 0.034 mmol), and Na2CO3 (2.0 M in water, 2.1 mL, 4.2 mmol) were added and the vial was heated in CEM microwave at about 150° C. for about 10 min. The reaction was diluted with water then extracted with EtOAc (3×15 mL). The precipitate present at the layer interface was kept with the organics. The combined organic layers were washed with brine, dried over Na2SO4, decanted, and concentrated. The crude product was triturated with CH2Cl2 and filtered to give a solid which was dissolved in DMSO and purified by preparative HPLC (Thermo Hypersil-Keystone 250×21.2 mm 8μ Hypersil® HS C18 column; 5% CH3CN/50 mM aqueous ammonium acetate hold for 5 min; 5-50% CH3CN/50 mM aqueous ammonium acetate over 15 min; 50-100% CH3CN/50 mM aqueous ammonium acetate over 1 min; hold at 100% CH3CN for 5 minutes, 21 mL/min). The combined fractions containing impure product were concentrated under reduced pressure, lyophilized, dissolved in DMSO, and further purified by preparative HPLC (Thermo Hypersil-Keystone 250×21.2 mm 8μ Hypersil® HS C18 column; 5% CH3CN/50 mM aqueous ammonium acetate hold for 5 min; 5-50% CH3CN/50 mM aqueous ammonium acetate over 20 min; 50-100% CH3CN/50 mM aqueous ammonium acetate over 1 min; hold at 100% CH3CN for 5 minutes, 21 mL/min) to afford 5-[5-(2-amino-pyrimidin-4-ylamino)-1H-indazol-7-yl]-1H-indole-3-carbaldehyde diacetate (12.3 mg, 6%); RP-HPLC (Table 1, Method e) Rt 1.18 min, m/z (ESI+) 370.3 (M+H)+.
  • Example #6 N4-(7-Piperidin-3-yl-1H-indazol-5-yl)pyrimidine-2,4-diamine diacetate
  • Figure US20120053345A1-20120301-C00882
  • To a vial containing N4-7-pyridin-3-yl-1H-indazol-5-yl)-pyrimidine-2,4-diamine diacetate (Example #F.5.5, 0.040 g, 0.090 mmol) was added L-Selectride® (1.0 M in THF, 0.45 mL, 0.45 mmol). Heated at about 130° C. for about 16 hours Cooled to room temperature and added L-Selectride® (1.0 M in THF, 1.0 mL, 1.0 mmol). Heating at about 130° C. was resumed for about 24 hours. Cooled to room temperature and added L-Selectride® (1.0 M in THF, 0.5 mL, 0.5 mmol). Heating at about 130° C. was resumed for about another 24 hours. The reaction was then cooled to room temperature, quenched with MeOH, and concentrated. The crude product was dissolved in DMSO and purified by preparative HPLC (Thermo Hypersil-Keystone 250×21.2 mm 8 μl Hypersil® HS C18 column; 5% CH3CN/50 mM aqueous ammonium acetate hold for 5 min; 5-50% CH3CN/50 mM aqueous ammonium acetate over 20 min; 50-100% CH3CN/50 mM aqueous ammonium acetate over 1 min; hold at 100% CH3CN for 5 minutes, 21 mL/min) to give N4-(7-Piperidin-3-yl-1H-indazol-5-yl)pyrimidine-2,4-diamine diacetate (9.1 mg, 23%) as a white solid; RP-HPLC (Table 1, Method g) Rt 0.94 min, m/z (ESL) 310.3 (M+H)+.
  • Example #7 N4-{7-[5-(Aminomethyl)-1-benzothien-2-yl]-1H-indazol-5-yl}pyrimidine-2,4-diamine acetate
  • Figure US20120053345A1-20120301-C00883
  • A microwave vial containing N4-(7-5-[(allylamino)methyl]-1-benzothien-2-yl-1H-indazol-5-yl)pyrimidine-2,4-diamine triacetate (Example #0.3.8, 0.022 g, 0.036 mmol), chlorotris(triphenylphosphine)-rhodium(I) (8.4 mg, 0.0090 mmol), CH3CN (0.84 mL), and H2O (0.16 mL) was heated in a CEM microwave at about 120° C. for about 30 min. The reaction was diluted with CH3CN (5 mL), filtered, and washed with CH3CN and EtOAc, to give N4-7-[5-(aminomethyl)-1-benzothien-2-yl]-1H-indazol-5-yl}pyrimidine-2,4-diamine acetate (12.8 mg, 76%) as a tan solid; RP-HPLC (Table 1, Method g) Rt 1.05 min, m/z (ESI+) 388.3 (M+H)+.
  • Preparation #1 4-Chloro-2-methanesulfonyl-pyrimidine
  • Figure US20120053345A1-20120301-C00884
  • A solution of 4-chloro-2-methylsulfanyl-pyrimidine (Aldrich, 41.37 g, 0.252 mol) in DCM (800 mL) at about 0° C. was treated with 3-chloroperbenzoic acid (75% by weight, 127.76 g, 0.555 mol) in small portions. The reaction mixture was warmed up to ambient temperature and stirred for about 2.5 hours. The insoluble residue was collected by filtration and washed with dichloromethane (100 mL). The filtrate and washings were washed with saturated aqueous sodium bicarbonate solution (3×150 mL) and saturated aqueous sodium chloride solution (200 mL), dried over anhydrous magnesium sulfate, and concentrated to dryness to give 4-chloro-2-methanesulfonyl-pyrimidine (41.83 g, 86%): 1H NMR (DMSO-d6, 400 MHz) δ 9.08 (d, 1H), 8.10 (d, 1H), 3.44 (s, 3H); LC/MS (Table 1, Method a) Rt 0.70 min.; m/z: (M+H)+193.
  • Preparation #2 4-Chloro-pyrimidin-2-ylamine
  • Figure US20120053345A1-20120301-C00885
  • A suspension of 4-chloro-2-methanesulfonyl-pyrimidine (Preparation #1, 41.83 g, 0.217 mol) in ethanol (30 mL) was treated with a solution of saturated ammonia gas in ethanol (300 mL). It was stirred at ambient temperature for about 2 hours, and the white precipitate was collected, washed with methanol (100 mL), and dried under reduced pressure to give 4-chloro-pyrimidin-2-ylamine (15.99 g, 57%): 1H NMR (DMSO-d6, 400 MHz) δ8.18 (d, 1H), 7.13 (br, 2H), 6.65 (d, 1H); LC/MS (Table 1, Method a) Rt 1.18 min.
  • Preparation #3 4-Chloro-7H-pyrrolo[2,3-d]pyrimidin-2-ylamine
  • Figure US20120053345A1-20120301-C00886
  • A mixture of 2-amino-7H-pyrrolo[2,3-d]pyrimidin-4-ol (Sigma, 150 mg, 1.0 mmol) in phosphorus oxychloride (1.5 mL) was heated at about 110° C. for about 30 minutes. Phosphorus oxychloride was carefully removed under reduced pressure and the reaction mixture was quenched by slow addition of ice water (10 mL). The resulting mixture was neutralized with saturated aqueous sodium carbonate (about 5 mL) to pH 7. The crude product was extracted into dichloromethane (20 mL) and washed with water (15 mL). The organic layer was separated and the aqueous phase was further extracted with dichloromethane (3×30 mL). The combined organic extracts were dried over anhydrous sodium sulfate and concentrated to give 4-chloro-7H-pyrrolo[2,3-d]pyrimidin-2-ylamine (0.036 g, 0.21 mmol) as a light yellow solid; LC/MS (Table 1, Method a) Rt 1.17 min; MS m/z: (M+H)+169.
  • Preparation #4 tert-Butyl-1,2-(dimethylaminomethyleneamino)-7-iodo-pyrrolo[3,2-d]pyrimidine-5-methanoate
  • Figure US20120053345A1-20120301-C00887
  • Step 1 Preparation #4a N′-(7-iodo-5H-pyrrolo[3,2-d]pyrimidin-2-yl)-N,N-dimethyl-formamidine
  • N-Iodosuccinimide (3.2 g, 14.2 mmol) was added portionwise to an ice-cold stirred suspension of N,N-dimethyl-N′-(5H-pyrrolo[3,2-d]pyrimidin-2-yl)-formamidine (Journal of Medicinal Chemistry, 2003, 46(14), 3060-3071, 2.68 g, 14.2 mmol) in DMF (50 mL). After stirring at ambient temperature overnight, the reaction was concentrated, dissolved in DCM (40 mL) and the product was precipitated using EtOAc (250 mL). After filtration, the pale yellow powder was collected, washed with EtOAc (2×20 mL) and dried under a vacuum overnight to afford N′-(7-iodo-5H-pyrrolo[3,2-d]pyrimidin-2-yl)-N,N-dimethyl-formamidine (3.8 g, 85%) as a yellow powder; 1H NMR (DMSO-d6, 400 MHz) δ 12.38 (s, 1H), 8.74 (s, 1 H), 8.04 (s, 1H), 7.93 (s, 1H), 3.29 (s, 3H), 3.19 (s, 3H); ESI-MS [M+H]+=316.1.
  • Step 2 Preparation #4 tert-Butyl-1,2-(dimethylaminomethyleneamino)-7-iodo-pyrrolo[3,2-d]pyrimidine-5-methanoate
  • Na2CO3 (1.27 g, 12 mmol) was added to a stirred suspension of N′-(7-iodo-5H-pyrrolo[3,2-d]pyrimidin-2-yl)-N,N-dimethyl-formamidine (Preparation #4a, 1 g, 3.17 mmol) in anhydrous THF (20 mL). Di-tert-butyl dicarbonate (2.76 g, 12 mmol) was added portionwise at ambient temperature and the mixture was stirred at ambient temperature for about 16 hours prior to diluting with DCM (150 mL), filtering and concentrating the filtrate to dryness. The product precipitated from the residue using EtOAc/heptane (50:50, 100 mL). The precipitate was filtered and dried to afford tert-butyl-1,2-(dimethylamino-methyleneamino)-7-iodo-pyrrolo[3,2-d]pyrimidine-5-methanoate (0.95 g, 72%) as a pale yellow solid; 1H NMR (DMSO-d6, 400 MHz) δ 9.10 (s, 1H), 8.74 (s, 1H), 7.96 (s, 1H), 3.23 (s, 3H), 3.18 (s, 3H), 1.68 (s, 9H); ESI-MS [M+H]+=416.2.
  • Preparation #5 N′-{7-[7-Benzo[b]thiophen-2-yl-1-(2-trimethylsilanyl-ethoxymethyl)-1H-indazol-5-yl]-5H-pyrrolo[3,2-d]pyrimidin-2-yl}-N,N-dimethyl-formamidine
  • Figure US20120053345A1-20120301-C00888
  • Step 1 Preparation #5a 7-[7-benzo[b]thiophen-2-yl-1-(2-trimethylsilanyl-ethoxymethyl)-1H-indazol-5-yl]-5H-pyrrolo[3,2-d]pyrimidin-2-ylamine
  • 2-(Dimethylamino-methyleneamino)-7-iodo-pyrrolo[3,2-d]pyrimidine-5-carboxylic acid tert-butyl ester (Preparation #4, 0.415 g, 1.0 mmoles) was added to a mixture of cesium carbonate (977 mg, 3.0 mmoles), Pd(PPh3)4 (0.12 g, 0.1 mmoles) and 7-(1H-inden-2-yl)-5-(4,4,5,5-tetramethyl-[1,3,2]dioxaborolan-2-yl)-1-(2-trimethylsilanyl-ethoxymethyl)-1H-indazole (Preparation #23 then E, 0.7 g, 1.1 mmoles) in DME (10 mL) and water (0.9 mL) at ambient temperature. The reaction was heated at about 65° C. for about 16 hours then the solvent was evaporated under reduced pressure. EtOH (4 mL) and 30% aqueous NaOH (4 mL) were added and the mixture was heated to about 85° C. for about 1 hour before the EtOH was removed under reduced pressure. The mixture was diluted with EtOAc (150 mL) and washed with water (50 mL) and brine (50 mL), dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by flash column chromatography (0-5% MeOH in CH2Cl2) to afford 7-[7-benzo[b]thiophen-2-yl-1-(2-trimethylsilanyl-ethoxymethyl)-1H-indazol-5-yl]-5H-pyrrolo[3,2-d]pyrimidin-2-ylamine (0.3 g, 60%) as a pale brown solid; LC/MS (Table 1, Method e) Rt 2.66 min; ESI-MS [M+H]+=511.3.
  • Step 2 Preparation #5 N′-{7-[7-Benzo[b]thiophen-2-yl-1-(2-trimethylsilanyl-ethoxymethyl)-1H-indazol-5-yl]-5H-pyrrolo[3,2-d]pyrimidin-2-yl}-N,N-dimethyl-formamidine
  • 7-[7-Benzo[b]thiophen-2-yl-1-(2-trimethylsilanyl-ethoxymethyl)-1H-indazol-5-yl]-5H-pyrrolo[3,2-d]pyrimidin-2-ylamine (Preparation #5a, 490 mg, 0.96 mmol) was added to a solution of N,N-dimethylformamide dimethyl acetal (1.27 mL, 9.6 mmol) in CH2Cl2/CHCl3 (3:2, 5 mL) at ambient temperature. The mixture was heated to about 45° C. for about 16 hours. After concentration, the crude product was purified by flash column chromatography (5-10% MeOH in CH2Cl2) to afford 7′-{7-[7-benzo[b]thiophen-2-yl-1-(2-trimethylsilanyl-ethoxymethyl)-1H-indazol-5-yl]-5H-pyrrolo[3,2-d]pyrimidin-2-yl}-N,N-dimethyl-formamidine (0.4 g, 73%) as a pale brown solid; LC/MS (Table 1, Method h) Rt 2.97 min; ESI-MS [M+H]+=568.4.
  • Preparation #6 7-Bromo-1H-indazol-5-ylamine
  • Figure US20120053345A1-20120301-C00889
  • Step 1 Preparation #6a 2-Bromo-4-nitro-6-methyl-aniline
  • Bromine (34.0 mL, 662 mmol) was added dropwise from an addition funnel to a suspension of 2-methyl-4-nitroaniline (100 g, 657 mmol) in glacial HOAc (1 L) at ambient temperature over about 40 min. The reaction mixture was stirred for about an additional 30 min at ambient temperature then diluted with H2O (1 L). The resulting solid was filtered, washed with additional H2O (1 L), and then dried in a vacuum oven overnight (at about 50-60° C.) to give the first crop of material (138.5 g, 91%). Additional precipitate, formed in the filtrate, was collected, washed with H2O (300 mL) then dried in a vacuum oven overnight (at about 50-60° C.) to give an additional batch of 2-bromo-6-methyl-4-nitroaniline (11.4 g, 7%); 1H NMR (DMSO-d6, 400 MHz) δ 8.14 (d, J=2.4 Hz, 1H), 7.92 (d, J=2.4 Hz, 1H), 6.49 (br s, 2H), 2.23 (s, 3H); LC/MS (Table 1, Method e) Rt 2.02 min; m/z (ESI) 230.7.
  • Step 2 Preparation #6b 7-Bromo-5-nitro-1H-indazole
  • NaNO2 (65.00 g, 942.0 mmol) in H2O (140 mL) was added via addition funnel to a 5 L 3-neck round bottom flask, equipped with a mechanical stirrer and a thermometer, containing a mixture of crude 2-bromo-6-methyl-4-nitroaniliine (145.0 g, 627.6 mmol) and glacial HOAc (2.5 L). During the addition, the reaction mixture was cooled with an ice bath to maintain the internal reaction temperature below 25° C. About 1 hour after the addition was complete, additional NaNO2 (21.65 g, 313.8 mmol) in H2O (50 mL) was added to the reaction mixture relatively rapidly (<5 min) with no evidence of an exotherm. After an additional 1 hour, the reaction mixture was concentrated under reduced pressure. The resulting solid was triturated with MeOH/H2O (1:1, 1 L) then filtered, washed with additional MeOH/H2O (1:1, 500 mL), and dried in a vacuum oven at about 50-60° C. for about 16 hours to give 7-bromo-5-nitro-1H-indazole (109.4 g, 72%, 90% pure by HPLC); 1H NMR (DMSO-d6, 400 MHz) δ 14.28 (br s, 1H), 8.87 (d, J=1.9 Hz, 1H), 8.57 (s, 1H), 8.40 (d, J=1.9 Hz, 1H); LC/MS (Table 1, Method e) Rt 1.75 min; m/z (ESI) 241.7.
  • Step 3 Preparation #6 7-Bromo-1H-indazol-5-ylamine
  • A mixture of iron (Aldrich 99.99+%, 13.85 g, 248.0 mmol) and crude 7-bromo-5-nitro-1H-indazole (20.00 g, 82.63 mmol) in glacial acetic acid (100 mL) was heated at about 80° C. in a 2-neck round bottom flask equipped with a mechanical stirrer and a nitrogen line with a bubbler. After about 2.5 hours, MeOH (100 mL) was added, warmed, and filtered hot through Celite®. The Celite® pad was washed with additional MeOH (4×100 mL) and the combined organic layers were concentrated under reduced pressure. The residues were basified to pH ˜7 using aqueous Na2CO3 (2M) and the product was extracted into EtOAc (1 L) over about 16 hours. The layers were separated and the aqueous layer was further extracted with additional EtOAc (3×500 mL). The combined organic extracts were washed with brine, dried over anhydrous Na2SO4, decanted, and concentrated. The resulting solid was pre-adsorbed onto silica and purified by silica gel chromatography using heptane/EtOAc (1:1) as the eluent to afford 7-bromo-1H-indazol-5-ylamine (6.92 g, 40%); 1H NMR (DMSO-d6, 400 MHz) δ 12.92 (br s, 1H), 7.87 (s, 1H), 7.03 (d, J=1.6 Hz, 1H), 6.77 (d, J=1.2 Hz, 1H) 4.96 (br s, 2H); LC-MS (Table 1, Method e) Rt 0.83.
  • Preparation #7 7-Benzo[b]thiophen-2-yl-1H-indazole-5-carboxylic acid
  • Figure US20120053345A1-20120301-C00890
  • Step 1 Preparation #7a Methyl 7-benzo[b]thiophen-2-yl-1H-indazol-5-yl carboxylate
  • A solution of 7-benzo[b]thiophen-2-yl-5-bromo-1H-indazole (Preparation #26, 4.5 g, 13.7 mmol) in N,N-dimethylformamide (100 mL) was degassed and purged with carbon monoxide. Triethylamine (4.1 g, 41 mmol), methanol (13.1 g, 410 mmol) and dichloro[bis(triphenylphosphine)]palladium (II) (1.45 g, 2.05 mmol) were added and the mixture was heated to about 90° C. for about 15 hours. The mixture was cooled and the solvent was removed under reduced pressure. The residue was triturated with ethyl acetate (75 mL) and water (35 mL) then further purified by flash chromatography over silica gel using dichloromethane/ethyl acetate (95:5) as the eluent to give methyl-7-benzo[b]thiophen-2-yl-1H-indazole-5-methanoate (3.37 g, 80%).
  • Step 2 Preparation #7b 7-Benzo[b]thiophen-2-yl-1H-indazole-5-carboxylic acid
  • Lithium hydroxide monohydrate (2.25 g, 53.5 mmol) was added to a suspension of methyl-7-benzo[b]thiophen-2-yl-1H-indazole-5-methanoate (3.37 g, 10.71 mmol) in a mixture of 1,4-dioxane (65 mL) and water (9 mL). The mixture was heated to about 65° C. for about 18 hours then cooled to about 40° C. and filtered. The filtrate was acidified with aqueous hydrochloric acid (1N, 54 mL) and the resulting precipitate was collected by filtration and dried under reduced pressure to give 7-benzo[b]thiophen-2-yl-1H-indazole-5-carboxylic acid (3.16 g, 100%); 1H NMR (DMSO-d6, 400 MHz) δ 13.56 (bs, 1H), 8.51 (d, 1H), 8.47 (s, 1H), 8.14 (d, 1H), 8.04 (d, 1H), 7.94 (d, 1H), 7.45 (d, 2H); RP-HPLC (Table 1, Method e) Rt=1.06, m/z: (M−H) 292.8.
  • Preparation #8 (7-Benzo[b]thiophen-2-yl-1H-indazol-5-yl)-(2-methanesulfonyl-pyrimidin-4-yl)-amine Preparation #9 4-(7-Benzo[b]thiophen-2-yl-1H-indazol-5-ylamino)-pyrimidin-2-ol
  • Figure US20120053345A1-20120301-C00891
  • To a mixture of 4-chloro-2-methanesulfonyl-pyrimidine (Preparation #1, 2.54 g, 13.2 mmol) in DME (20 mL) was added 7-benzo[b]thiophen-2-yl-1H-indazol-5-ylamine (Example #F.8.1, 1.95 g, 7.35 mmol) and triethylamine (1.43 mL, 10.3 mmol) in DME (160 mL). After about 16 hours, the mixture was filtered, and the filtrate evaporated and purified by flash column chromatography over alumina using dichloromethane/methanol (99:1) as the eluent to yield product (190 mg, 6% yield). Additional product was obtained by washing the alumina with dichloromethane/methanol (8:2) and concentrating the solution to yield (7-benzo[b]thiophen-2-yl-1H-indazol-5-yl)-(2-methanesulfonyl-pyrimidin-4-yl)-amine (900 mg, 30% yield); LC/MS (Method e) Rt 1.8 min; m/z (ESI−): (M−H)+ 419.9.
  • In an alternate work up, the crude reaction mixture was filtered and the solid was purified by RP-HPLC to yield 4-(7-benzo[b]thiophen-2-yl-1H-indazol-5-ylamino)-pyrimidin-2-ol (2 mg, 0.006 mmol, 0.1% yield): LC/MS Rt (Table 1, Method e) Rt 1.3 min; m/z (ESI−): (M−H)+ 358.3.
  • Preparation #10 N′-(1-Benzenesulfonyl-3-bromo-1H-pyrrolo[2,3-c]pyridin-5-yl)-N,N-dimethyl-formamidine
  • Figure US20120053345A1-20120301-C00892
  • Step 1 Preparation #10a N′-[4-((E)-2-Dimethylamino-vinyl)-5-nitro-pyridin-2-yl]-N,N-dimethylformamidine
  • Figure US20120053345A1-20120301-C00893
  • 4-Methyl-5-nitro-pyridin-2-ylamine (Aldrich, 4.80 g, 31.4 mmol) was suspended in dimethoxymethyl-dimethylamine (50 mL) and the mixture was heated at about 120° C. in a sealed vessel with stirring for about 4 days. The reaction was cooled and concentrated to afford N′-[4-((E)-2-Dimethylamino-vinyl)-5-nitro-pyridin-2-yl]-N,N-dimethyl-formamidine (8.36 g, 101%) as a dark solid, RP HPLC (Table 1, Method e) Rt=1.62, m/z: (MH)+ 264. The crude product was then used in the next step without further purification.
  • Step 2 Preparation #10b N,N-Dimethyl-N′-(1H-pyrrolo[2,3-c]pyridin-5-yl)-formamidine
  • Figure US20120053345A1-20120301-C00894
  • A suspension of N′-[4-((E)-2-dimethylamino-vinyl)-5-nitro-pyridin-2-yl]-N,N-dimethyl-formamidine (Preparation #10a, 8.36 g, 31.4 mmol) and 10% Pd on carbon (800 mg) in ethanol (120 mL) was shaken in a hydrogenation vessel under an atmosphere of 10-55 psi H2 for about 24 hours. The reaction was filtered through Celite® and concentrated to yield N,N-dimethyl-N′-(1H-pyrrolo[2,3-d]pyridin-5-yl)-formamidine (6.19 g, 105%) as a dark solid. RP HPLC (Table 1, Method e) Rt=0.84, m/z: (MH)+ 189. The crude product was used in the next step without further purification.
  • Step 3 Preparation #10 N′-(1-Benzenesulfonyl-3-bromo-1H-pyrrolo[2,3-c]pyridin-5-yl)-N,N-dimethyl-formamidine
  • Figure US20120053345A1-20120301-C00895
  • 1-Bromo-pyrrolidine-2,5-dione (5.34 g, 30 mmol) was added to a stirred solution of N,N-dimethyl-N′-(1H-pyrrolo[2,3-c]pyridin-5-yl)-formamidine (Preparation #10b, 6.00 g, 31.9 mmol) in DMF (200 mL) at about 0° C. The reaction was allowed to warm to ambient temperature then stirred for about 1 hour. Benzenesulfonyl chloride (3.83 mL, 30.0 mmol) and Na2CO3 (6.36 g, 60.0 mmol) were added and the mixture was warmed at about 60° C. for about 1 hour. The reaction mixture was concentrated under reduced pressure and the dark solids were triturated with EtOAc (2×100 mL) and filtered. The combined EtOAc layers were passed through a 6 inch silica gel column, eluting with EtOAc. The product fractions were combined and concentrated to yield N′-(1-benzenesulfonyl-3-bromo-1H-pyrrolo[2,3-d]pyridin-5-yl)-N,N-dimethylformamidine (4.29 g, 35%) as a tan solid. RP HPLC (Table 1, Method e) Rt=2.18, m/z: (MH)+ 407/409.
  • Preparation #11 Methyl 1-(4-Methoxy-benzyl)-5-(4,4,5,5-tetramethyl-[1,3,2]dioxaborolan-2-yl)-1H-indazole-7-carboxylate Preparation #12 Methyl 2-(4-Methoxy-benzyl)-5-(4,4,5,5-tetramethyl-[1,3,2]dioxaborolan-2-yl)-2H-indazole-7-carboxylate
  • Figure US20120053345A1-20120301-C00896
  • Step 1 Preparation #11a Methyl 2-amino-5-bromo-3-iodo-benzoate
  • Figure US20120053345A1-20120301-C00897
  • 1-Iodo-pyrrolidine-2,5-dione (10.35 g, 46.0 mmol) was added in one portion to a solution of methyl 2-amino-5-bromo-benzoate (10.35 g, 45.0 mmol) in trifluoroacetic acid (90 mL) at ambient temperature. The reaction was stirred for about 1 hour at ambient temperature then concentrated under reduced pressure. The residue was dissolved in CH2Cl2 (150 mL), washed with saturated Na2CO3 solution (2×150 mL) and a 10% aqueous solution of Na2S2O4 (2×100 mL), dried over anhydrous MgSO4, filtered and concentrated to yield methyl 2-amino-5-bromo-3-iodo-benzoate (15.5 g, 97%) as a yellow solid; RP HPLC (Table 1, Method e) Rt=2.45, 1H NMR (DMSO-d6, 400 MHz) δ 3.83 (s, 3H), 7.74 (broad s), 2H), 7.86-7.88 (d, 2H), 8.00-8.02 (d, 1H).
  • Step 2 Preparation #11b Methyl 2-amino-5-bromo-3-methylbenzoate
  • Figure US20120053345A1-20120301-C00898
  • 2,4,6-Trimethyl-cyclotriboroxane (6.99 ml, 50.0 mmol) was added to a stirred suspension of methyl 2-amino-5-bromo-3-iodo-benzoate (Preparation #11a, 15.5 g, 43.6 mmol), [1,1′-bis(diphenylphosphino)-ferrocene)dichloropalladium(II) 1:1 complexed with dichloromethane (1.67 g, 2.05 mmol) and cesium carbonate (42.9 g, 132 mmol) in 1,4-dioxane (200 mL) under N2. After about 4 hours at about 90° C. additional 2,4,6-trimethyl-cyclotriboroxane (1.00 ml, 7.15 mmol) was added and the reaction was continued with heating for about 2 more hours. The reaction was cooled, filtered through a short pad of silica gel and concentrated to afford an oil. The crude product was purified by flash chromatography over silica gel using heptane:EtOAc (92:8) as the eluent to yield methyl 2-amino-5-bromo-3-methyl-benzoate (5.9 g, 55%) as a pale yellow solid; RP HPLC (Table 1, Method e) Rt=2.72, m/z: (MH)+ 244/246.
  • Step 3 Preparation #11c Methyl 5-bromo-1H-indazole-7-carboxylate
  • Figure US20120053345A1-20120301-C00899
  • 3-Methyl-1-nitrosooxy-butane (3.39 ml, 25.2 mmol) was added to a solution of 2-methyl amino-5-bromo-3-methyl-benzoate (5.38 g, 22.0 mmol) in glacial acetic acid (300 mL) cooled to about 17° C. The reaction was stirred for about 10 minutes at about 17° C. then transferred over about 1 hour to a mixture of acetic acid (60 mL) and potassium acetate (20.0 g, 0.20 mol) held at 60° C. with stirring. The mixture was stirred about 1 hour then cooled to ambient temperature and concentrated under reduced pressure. The residue was dissolved in EtOAc (about 350 mL) and washed with water (3×150 mL) and saturated aqueous NaCl solution (100 mL), then dried over anhydrous MgSO4. The residues were triturated with ether (50 mL), filtered, washed with additional ether (2×10 mL), and dried to yield 5-bromo-1H-indazole-7-carboxylic acid methyl ester (4.42 g, 79%) as a tan solid, RP HPLC (Table 1, Method e) Rt=2.43, m/z: (MH)+ 255/257.
  • Step 4 Preparation #11d Methyl 1-(4-methoxy-benzyl)-5-bromo-1H-indazole-7-carboxylate, and, Preparation #12a Methyl 2-(4-methoxy-benzyl)-5-bromo-2H-indazole-7
  • Figure US20120053345A1-20120301-C00900
  • A mixture of methyl 5-bromo-1H-indazole-7-carboxylate (Preparation #11c, 2.60 g, 10.2 mmol), potassium carbonate 3.31 g, 24.0 mmol) and 4-methoxy-benzyl chloride (1.62 ml, 12.0 mmol) in N,N-dimethylformamide (40 mL) was warmed at about 60° C. for about 4 hours. The reaction was filtered, concentrated under reduced pressure and the residue was partitioned between EtOAc (100 mL) and water (50 mL). The layers were separated and the organic layer was washed with water (2×50 mL), dried over anhydrous MgSO4, filtered and concentrated to yield a mixture of methyl 1-(4-methoxy-benzyl)-5-bromo-1H-indazole-7-carboxylate and methyl 2-(4-methoxy-benzyl)-5-2H-indazole-7-carboxylate as an oil (4.07 g, 106%). RP HPLC (Table 1, Method e) Rt=2.17, m/z: (MH)+ 244/246, and Rt=2.46, m/z: (MH)+ 244/246. The crude product was used without further purification.
  • Step 5 Preparation #11 Methyl 1-(4-methoxy-benzyl)-5-(4,4,5,5-tetramethyl-[1,3,2]dioxaborolan-2-yl)-1H-indazole-7-carboxylate Preparation #12 Methyl 2-(4-methoxy-benzyl)-5-(4,4,5,5-tetramethyl-[1,3,2]dioxaborolan-2-yl)-2H-indazole-7-carboxylate
  • Figure US20120053345A1-20120301-C00901
  • [1,1′-Bis(diphenylphosphino)-ferrocene)dichloropalladium(II) 1:1 complex with dichloromethane (408 mg, 0.50 mmol) was added to a mixture of methyl 1-(4-methoxy-benzyl)-5-bromo-1H-indazole-7-carboxylate and methyl 2-(4-methoxy-benzyl)-5-2H-indazole-7-carboxylate (Preparations #11d and 12a, 3.70 g, 9.87 mmol), 4,4,5,5,4′,4′,5′,5′-octamethyl-[2,2′]bi[[1,3,2]dioxaborolanyl] (7.62 g, 30.0 mmol) and potassium acetate (5.88 g, 60.0 mmol) in dimethylformamide (50 mL) under a nitrogen atmosphere. The mixture was sealed and heated at about 80° C. for about 1 hour then cooled to ambient temperature, filtered, and concentrated. The mixture was purified on a silica gel column using CH2Cl2 as the eluent to elute fractions containing the first isomer. These combined fractions were triturated with heptane (about 15 mL), filtered and dried to yield isomer A (2.06 g, 94% purity, 46% yield), RP HPLC (Table 1, Method e) Rt=2.50, m/z: (MH)+ 423. The second isomer was eluted using EtOAc and again the fractions were combined, concentrated, and dried to yield isomer B (2.31 g, 55%), RP HPLC (Table 1, Method e) Rt=2.24, m/z: (MH)+ 423.
  • Preparation #13 5-(5-Amino-1H-pyrrolo[2,3-c]pyridin-3-yl)-1H-indazole-7-carboxylic acid monohydrochloride
  • Figure US20120053345A1-20120301-C00902
  • [1,1′-Bis(diphenylphosphino)-ferrocene)dichloropalladium(II) 1:1 complex with dichloromethane (408 mg, 0.50 mmol) was added to a mixture of methyl 5-bromo-1-(4-methoxy-benzyl)-1H-indazole-7-carboxylate ester and methyl 5-bromo-2-(4-methoxy-benzyl)-2H-indazole-7-carboxylate (Preparations #11 and 12, 1:1, 3.99 g, 9.45 mmol), cesium carbonate (9.75 g, 30.0 mmol) and N′-(1-benzenesulfonyl-3-bromo-1H-pyrrolo[2,3-c]pyridin-5-yl)-N,N-dimethyl-formamidine (Preparation #10, 3.85 g, 9.45 mmol) in 1,2-dimethoxyethane (100 mL) and water (25 mL) under an atmosphere of N2. The reaction mixture was heated at about 90° C. for about 4 hours. The reaction was cooled to ambient temperature and the layers separated. The organic layer was filtered through a silica gel pad, eluting with 5% MeOH/EtOAc and the filtrate was concentrated under reduced pressure. The residue was dissolved in trifluoroacetic acid (50 mL) containing triisopropylsilane (2.05 ml, 10.0 mmol) and the mixture was heated in a sealed tube at about 100° C. for about 30 minutes then cooled to ambient temperature and concentrate under reduced pressure. The residue was triturated with ether (100 mL) and the resulting solid was collected by filtration. The solid was then dissolved in methanol (75 mL), treated with aqueous sodium hydroxide (2N, 50 ml, 100 mmol) and heated at reflux for about 6 hours. The reaction was filtered hot, then cooled and acidified with acetic acid to about pH 5.5. The product was filtered off, washed with water (2×10 mL) and dried to yield 5-(5-amino-1H-pyrrolo[2,3-c]pyridin-3-yl)-1H-indazole-7-carboxylic acid (2.60 g, 78%) as the acetate salt. The acetate salt was dissolved in methanol (300 mL) and aqueous HCl (2N, 25 mL) was added. The mixture was concentrated and dried to yield 5-(5-amino-1H-pyrrolo[2,3-c]pyridin-3-yl)-1H-indazole-7-carboxylic acid monohydrochloride (2.35 g, 75%) as a pale yellow solid. RP HPLC (Table 1, Method e) Rt=1.31, m/z: (MH)+ 294.
  • Preparation #14 (7-Benzo[b]thiophen-2-yl-1H-indazol-5-yl)-thiocarbamic acid S-methyl ester
  • Figure US20120053345A1-20120301-C00903
  • To a solution of 7-benzo[b]thiophen-2-yl-1H-indazol-5-ylamine (Example #F.8.1, 530 mg, 2.0 mmol) and methyl chlorothioformate (0.25 mL, 2.8 mmol) in ethanol (10 mL) was added triethylamine (0.56 mL, 4.0 mmol). The reaction mixture was stirred at ambient temperature for about 16 hours and the resulting solid was collected by filtration, washed with water (3×50 mL), and dried to yield (7-benzo[b]thiophen-2-yl-1H-indazol-5-yl)-thiocarbamic acid S-methyl ester (448 mg, 66%); LC/MS (Table 1, Method e) Rt 2.14 min; m/z: [M−H] 338.
  • Preparation #15 2-(7-Benzo[b]thiophen-2-yl-1H-indazol-5-ylamino)-2-oxalyl chloride
  • Figure US20120053345A1-20120301-C00904
  • Step 1 Preparation #15a Ethyl-N-(7-benzo[b]thiophen-2-yl-1H-indazol-5-yl)-oxamate
  • To a suspension of 7-benzo[b]thiophen-2-yl-1H-indazol-5-ylamine (Example #F.8.1, 151 mg, 0.57 mmol) in dichloromethane (6 mL) was added triethylamine (0.088 mL, 0.63 mmol). The reaction mixture was stirred at about −5° C. for about 10 minutes and ethyl oxalyl chloride (0.064 mL, 0.57 mmol) was added dropwise. The reaction mixture was stirred at about −5° C. for about 90 minutes prior to quenching the reaction with water (about 10 mL). The suspension was extracted with EtOAc (3×50 mL), washed with brine solution (100 mL), and dried over anhydrous magnesium sulfate. The solvent was removed in vacuo and the crude product was purified by flash chromatography over silica gel using heptane:EtOAc (30:70) as the eluent to yield ethyl-N-(7-benzo[b]thiophen-2-yl-1H-indazol-5-yl)-oxamate (169 mg, 81%) as a pale yellow solid, RP HPLC (Table 1, Method e) Rt=2.03, m/z: (M−H) 364.
  • Step 2 Preparation #15b N-(7-benzo[b]thiophen-2-yl-1H-indazol-5-yl)-oxamate
  • Sodium hydroxide solution (2% aqueous solution, 2 mL) was added to a suspension of ethyl-N-(7-benzo[b]thiophen-2-yl-1H-indazol-5-yl)-oxamate (Preparation #15a, 83 mg, 0.23 mmol) in methanol (3 mL) and the reaction mixture was refluxed at 100° C. for about 1 hour. The solvent was removed in vacuo, and water was added to the residue. The aqueous layer was acidified using aqueous hydrochloric acid solution (1 N) and the resulting solid was collected by filtration, washed with water (3×5 mL), and dried to yield the N-(7-benzo[b]thiophen-2-yl-1H-indazol-5-yl)-oxamate (76 mg, 99%); LC/MS (Table 1, Method e) Rt 0.89 min; m/z: [M−H] 336.
  • Step 3 Preparation #15 2-(7-Benzo[b]thiophen-2-yl-1H-indazol-5-ylamino)-2-oxalyl chloride
  • A mixture of N-(7-benzo[b]thiophen-2-yl-1H-indazol-5-yl)-oxamate (Preparation #15b, 30 mg, 0.089 mmol) and thionyl chloride (0.5 mL, 6.7 mmol) was stirred at about 80° C. for about 1 hour. The excess thionyl chloride was removed in vacuo and the 2-(7-benzo[b]thiophen-2-yl-1H-indazol-5-ylamino)-2-oxalyl chloride (31 mg, 98%) was used without further purification.
  • Preparation #16 5-Pyridin-3-yl-3-(1H-pyrrol-2-yl)-1H-indazole
  • Figure US20120053345A1-20120301-C00905
  • Step 1 Preparation #16a 5-Bromo-2-fluoro-N,N-dimethylbenzamide
  • Figure US20120053345A1-20120301-C00906
  • Thionyl chloride (19.8 mL, 228 mmol) was added to a mixture of 5-bromo-2-fluoro-benzoic acid (Aldrich, 5.0 g, 22.8 mmol) in toluene (45 mL). The resulting mixture was heated at reflux for about 3 hours, cooled to ambient temperature and the solvents were removed under reduced pressure. The residue was stirred in THF (40 mL) at about 0° C. and then the reaction was treated with gaseous dimethylamine. The mixture was stirred for about 15 min, concentrated, dissolved in EtOAc (100 mL), washed with 2N HCl solution (2×50 mL), 2N NaOH solution (2×50 mL), and finally with saturated NaCl solution (50 mL). The organic layer was dried over anhydrous MgSO4, filtered, concentrated, and dried to yield 5-bromo-2-fluoro-N,N-dimethyl-benzamide (4.82 g, 86%) of as an oil, RP HPLC (Table 1, Method e) Rt=2.33, m/z: (MH)+ 246/248
  • Step 2 Preparation #16b (5-Bromo-2-fluoro-phenyl)-(1H-pyrrol-2-yl)-methanone
  • Figure US20120053345A1-20120301-C00907
  • 5-Bromo-2-fluoro-N,N-dimethyl-benzamide (Preparation #16a, 4.75 g, 19.3 mmol) and phosphorus oxychloride (5.46 ml, 40.5 mmol) were combined and warmed at about 35° C. for about 28 hours. The mixture was cooled to ambient temperature and a mixture of pyrrole (1.38 mL, 20.0 mmol) in CH2Cl2 (40 mL) was added and the reaction was stirred for about 3 hours at ambient temperature. The reaction was quenched by cautious addition of aqueous Na2CO3 solution (10% w/v, ˜150 mL) and the resulting mixture was heated at reflux for about 2 hours. The product was extracted with CH2Cl2 (3×50 mL), dried over anhydrous MgSO4, filtered and concentrated. The crude product was further purified on silica gel using EtOAc:heptane (1:1) as the eluent to yield 5-bromo-2-fluoro-N,N-dimethyl-benzamide (2.57 g, 50%) as an oil which solidifies on standing. RP HPLC (Table 1, Method e) Rt=2.57; m/z: (M−H) 266/268
  • Step 3 Preparation #16c 5-Bromo-3-(1H-pyrrol-2-yl)-1H-indazole
  • Figure US20120053345A1-20120301-C00908
  • A slurry containing (5-bromo-2-fluoro-phenyl)-(1H-pyrrol-2-yl)-methanone (Preparation #16b, 2.40 g, 8.96 mmol) and hydrazine hydrate (35 mL) was sealed and heated by microwave at about 150° C. for about 1 hour. The reaction was cooled to ambient temperature and diluted with water (150 mL). The product was extracted with EtOAc (150 mL), washed with 2N HCl solution (50 mL), saturated NaHCO3 solution (50 mL) and finally a saturated NaCl solution. The EtOAc layer was dried over anhydrous MgSO4, filtered and concentrated under reduced pressure to yield 5-bromo-3-(1H-pyrrol-2-yl)-1H-indazole (2.38 g, 99%) as an oil. RP HPLC (Table 1, Method e) Rt=2.95; m/z: (MH)+ 262/264
  • Step 4 Preparation #16 5-Pyridin-3-yl-3-(1H-pyrrol-2-yl)-1H-indazole
  • Figure US20120053345A1-20120301-C00909
  • [1,1′-Bis(diphenylphosphino)-ferrocene)dichloropalladium(II) 1:1 complex with dichloromethane (40.8 mg, 0.05 mmol) was added to a mixture containing 5-bromo-3-(1H-pyrrol-2-yl)-1H-indazole (Preparation #16c, 105 mg, 0.40 mmol), aqueous pyridine-3-boronic acid (1M, 1.0 mL), and Cs2CO3 (391 mg, 1.20 mmol) in DMF (2.0 mL) under a nitrogen atmosphere was added and the mixture was purged with N2 again, then heated at reflux for about 3 days. The reaction was cooled to ambient temperature and the organic layer was filtered through silica gel and concentrated under reduced pressure. The residue was further purified by preparative HPLC (Thermo Hypersil-Keystone 250×21.2 mm 8μ Hypersil® HS C18 column; 5% CH3CN/50 mM aqueous ammonium acetate hold for 5 min; 5-50% CH3CN/50 mM aqueous ammonium acetate over 20 min; 50-100% CH3CN/50 mM aqueous ammonium acetate over 1 min; hold at 100% CH3CN for 5 minutes, 21 mL/min). Product fractions were combined and concentrated to remove organic solvents and the lyophilized to yield 5-pyridin-3-yl-3-(1H-pyrrol-2-yl)-1H-indazole (44 mg, 42%) as an off-white powder. RP HPLC (Table 1, Method e) Rt=2.23; m/z: (MH)+ 261.
  • Preparation #17 5-Pyridin-3-yl-1H-indazole
  • Figure US20120053345A1-20120301-C00910
  • Step 1 Preparation #17a 5-Iodo-1H-indazole
  • Figure US20120053345A1-20120301-C00911
  • A solution of NaNO2 (6.24 g, 90.0 mmol) in water (36 mL) was added dropwise to a mixture of 1H-indazol-5-ylamine (Aldrich, 12.0 g, 90.1 mmol), water (36 mL), acetonitrile (12 mL) and concentrated HCl (14.4 mL) that was pre-cooled to about −10° C., maintaining the reaction temperature below about −5° C. during the addition. The mixture was stirred for about 10 minutes at about −5° C. after the addition was complete then a solution of KI (15.0 g, 90.0 mmol) in cold water (30 mL) was added, followed by cold acetonitrile (50 mL). The cold bath was removed and the reaction was allowed to warm to ambient temperature with stirring overnight. The organic solvent was removed under reduced pressure and the solid product was filtered off, then re-dissolved in acetonitrile (125 mL) and stirred in the presence of charcoal (about 5 g) for about 30 min. The mixture was filtered and concentrated to yield 5-iodo-1H-indazole (13.1 g, 60%) as a light brown solid. RP HPLC (Table 1, Method e) Rt=2.38; m/z: (MH)+245.
  • Step 2 Preparation #17b 5-Iodo-1-(2-trimethylsilanyl-ethoxymethyl)-1H-indazole
  • Figure US20120053345A1-20120301-C00912
  • 1H-Indazol-5-ylamine (Preparation #17a, 12.9 g, 52.9 mmol) was dissolved in CHCl3 (66 mL), cooled to about 0° C. and treated with a solution of KOH (66.0 g, 1.18 mol) in water (66 mL) and tetrabutylammonium bromide (175 mg, 0.54 mmol). (2-chloromethoxy-ethyl)-trimethyl-silane (10.5 ml, 59.3 mmol) was added over about 5 minutes at about 0° C. with vigorous stirring. The reaction was then stirred for about 30 minutes at about 0° C., diluted with water (100 mL) and extracted with CHCl3 (3×100 mL). The CHCl3 layer was dried over anhydrous MgSO4, filtered, and concentrated to give an oil. The crude product was further purified over silica gel using EtOAc:heptane (95:5) as the eluent to yield 5-iodo-1-(2-trimethylsilanyl-ethoxymethyl)-1H-indazole (11.2 g, 56%) as an oil. RP HPLC (Table 1, Method e) Rt=3.04; NMR (d6-DMSO, 400 MHz) δ −0.12 (s, 9H), 0.77-0.80 (t, 2H), 3.47-3.51 (t, 2H), 5.74 (s, 2H), 7.60-7.63 (d, 1H), 7.67-7.70 (d,d 1H), 8.09-8.10 (d, 1H), 8.21-8.22 (m, 1H).
  • Step 3 Preparation #17c 5-Pyridin-3-yl-1-(2-trimethylsilanyl-ethoxymethyl)-1H-indazole
  • Figure US20120053345A1-20120301-C00913
  • [1,1′-Bis(diphenylphosphino)-ferrocene)dichloropalladium(II) 1:1 complex with dichloromethane (204 mg, 0.25 mmol) was added to a mixture of 5-iodo-1-(2-trimethylsilanyl-ethoxymethyl)-1H-indazole (Preparation #17a, 1.87 g, 5.00 mmol), pyridine-3-boronic acid (737 mg, 6.00 mmol), and Cs2CO3 (4.89 g, 15.0 mmol) in 1,2-dimethoxy-ethane (16 mL) and water (3.2 mL) under a nitrogen atmosphere. The reaction was heated at about 100° C. overnight, cooled, concentrated and extracted with CH2Cl2 (about 30 mL). The extracts were adsorbed onto silica and purified over a silica gel column eluting first with heptane:EtOAc (85:15) and then with heptane:EtOAc (3:2) to yield 5-pyridin-3-yl-1-(2-trimethylsilanyl-ethoxymethyl)-1H-indazole (857 mg, 53%) as an oil. RP HPLC (Table 1, Method e) Rt=3.04, m/z: (MH)+ 326.
  • Step 4 Preparation #17 5-Pyridin-3-yl-1H-indazole
  • Figure US20120053345A1-20120301-C00914
  • 5-Pyridin-3-yl-1-(2-trimethylsilanyl-ethoxymethyl)-1H-indazole (Preparation #17c, 100 mg, 0.31 mmol) and ethylenediamine (133 μL, 2.00 mmol) were dissolved in 1M tetrabutylammonium fluoride in THF (4.0 mL) and the mixture was heated at reflux for about 2 hours. The reactions was cooled to ambient temperature and concentrated under reduced pressure. The residue was dissolved in EtOAc (25 mL), washed with water (2×10 mL) and concentrated under reduced pressure. The crude product was further purified by reverse phase preparative HPLC (Thermo Hypersil-Keystone 250×21.2 mm 8μ Hypersil® HS C18 column; 5% CH3CN/50 mM aqueous ammonium acetate hold for 5 min; 5-50% CH3CN/50 mM aqueous ammonium acetate over 20 min; 50-100% CH3CN/50 mM aqueous ammonium acetate over 1 min; hold at 100% CH3CN for 5 minutes, 21 mL/min) to yield 5-pyridin-3-yl-1H-indazole (61 mg, 64%) as an off-white solid. RP HPLC (Table 1, Method e) Rt=1.65, m/z: (MH)+ 196.
  • Preparation #18 Indole-1,4-dicarboxylic acid 1-tert-butyl ester 4-methyl ester
  • Figure US20120053345A1-20120301-C00915
  • Di-tert-butyldicarbonate (0.068 g, 0.31 mmol) was added to a solution of 1H-indole-4-carboxylic acid methyl ester (Aldrich, 0.050 g, 28 mmol) and 4-dimethylaminopyridine (0.7 mg, 0.006 mmol) in methylene chloride (1.0 mL, 0.016 mol). After 45 minutes, the mixture was quenched and acidified with 1N HCl to about a pH 4. Methylene chloride was added and the layers were separated. The aqueous layer was further extracted with methylene chloride and the combined organics were dried over anhydrous MgSO4 and concentrated to yield indole-1,4-dicarboxylic acid 1-tert-butyl ester 4-methyl ester (0.072 g, 92% yield); (DMSO-d6, 400 MHz) δ 8.37 (d, 1H), 7.91 (d, 1H), 7.86 (d, 1H), 7.46 (t, 1H), 7.21 (d, 1H), 3.92 (s, 3H), 1.65 (s, 10H); RP-HPLC (Table 1, Method e) Rt=2.7, m/z: (M+H)+ 276.3.
  • Preparation #19 Indole-1,7-dicarboxylic acid 1-tert-butyl ester 7-methyl ester
  • Figure US20120053345A1-20120301-C00916
  • Di-tert-butyldicarbonate (16 g, 0.071 mol) was added to a solution of 1H-indole-7-carboxylic acid methyl ester (Acros chemicals, 5.0 g, 0.028 mol) and 4-dimethylaminopyridine (0.3 g, 0.003 mol) in methylene chloride (100 mL). The mixture was heated at about 45° C. for about 16 hours. The mixture was quenched and acidified with 1N HCl to about a pH 4. Methylene chloride was added and the layers were separated. The aqueous layer was further extracted with methylene chloride and the combined organics were dried over anhydrous MgSO4 and concentrated to yield indole-1,7-dicarboxylic acid 1-tert-butyl ester 7-methyl ester (9.5 g, 97% yield); RP-HPLC (Table 1, Method e) Rt=2.5, m/z: (M+H)+ 276.2.
  • Preparation #20 Indole-2-boronic-1,7-dicarboxylic acid 1-tert-butyl ester 7-methyl ester
  • Figure US20120053345A1-20120301-C00917
  • To a mixture of indole-1,7-dicarboxylic acid 1-tert-butyl ester 7-methyl ester (Preparation #19, 9.5 g, 0.028 mol) in tetrahydrofuran (39 mL, 0.48 mol) was added triisopropyl borate (9.5 mL, 0.041 mol). The heterogeneous mixture was cooled to about 0-5° C. and lithium diisopropylamide in tetrahydrofuran (2.0M, 25 mL) was added slowly. After about 2 hours the mixture was quenched with 1N HCl and the layers were separated. The aqueous layer was washed with DCM and the combined organics were dried over anhydrous MgSO4 and concentrated. Trituration in ether/heptane gave 3 crops of indole-2-boronic-1,7-dicarboxylic acid 1-tert-butyl ester 7-methyl ester (4.10 g, 1.6 g and 3.0 g respectively) as a yellow solid; RP-HPLC (Table 1, Method e) Rt 1.7 min; m/z (M+H)+ 378.4.
  • Preparation #21 Indole-2-boronic-1,4-dicarboxylic acid 1-tert-butyl ester 4-methyl ester
  • Figure US20120053345A1-20120301-C00918
  • To a solution of indole-1,4-dicarboxylic acid 1-tert-butyl ester 4-methyl ester (Preparation #18, 0.207 g, 0.752 mmol) in THF (1.1 mL) was added triisopropyl borate (260 μL, 0.0011 mol). The solution was cooled to about 0-5° C. and lithium diisopropylamide in tetrahydrofuran (2.0M, 600 μL) was added slowly over about 1 hour. After about 30 minutes, the mixture was quenched with 1N HCl, and the layers were separated. The aqueous layer was washed with DCM and the combined organics were dried over anhydrous MgSO4 and concentrated. The mixture was triturated in heptane (50 mL) and ether (5 mL) and filtered to yield indole-2-boronic-1,4-dicarboxylic acid 1-tert-butyl ester 4-methyl ester (175 mg, 51% yield): RP-HPLC (Table 1, Method e) Rt 1.84 min; m/z 377.9 (M−H).
  • Preparation #22 4-Diisopropylcarbamoyl-indole-2-boronic-1-carboxylic acid 1-tert-butyl ester
  • Figure US20120053345A1-20120301-C00919
  • Triisopropyl borate (870 μL, 0.0038 mol) was added to a solution of indole-1,4-dicarboxylic acid 1-tert-butyl ester 4-methyl ester (Preparation #18, 0.70 g, 0.0025 mol) in THF (36 mL). The solution was cooled to about 0-5° C. and lithium diisopropylamide in tetrahydrofuran (2 M, 2.8 mL) was added slowly over about 1 hour. After about another 1.5 hours, the mixture was quenched with 1N HCl, and the layers were separated. The aqueous layer was extracted with DCM, and the combined organics were dried, concentrated, triturated in heptane (50 mL) and ether (5 mL) and filtered. The material was further purified by flash column chromatography using DCM/MeOH (98:2) as the eluent to afford 4-diisopropylcarbamoyl-indole-2-boronic-1-carboxylic acid 1-tert-butyl ester (20 mg, 2% yield): RP-HPLC (Table 1, Method e) Rt 2.0 min; m/z 389.4 (M+H)+.
  • Example #8 4-(7-Benzo[b]thiophen-2-yl-1H-indazol-5-ylamino)-pyrimidin-2-ol
  • Figure US20120053345A1-20120301-C00920
  • To a mixture of 4-chloro-2-methanesulfonyl-pyrimidine (Preparation #1, 1.35 g, 7.0 mmol) in DME (10 mL) was added a mixture of 7-benzo[b]thiophen-2-yl-1H-indazol-5-ylamine (Example #F.8.1, 1.03 g, 3.9 mmol) and triethylamine (0.76 mL, 5.4 mmol) in DME (70 mL).
  • After about 16 hours at ambient temperature the mixture was filtered, and the solid was purified by reverse phase preparative HPLC (Thermo Hypersil-Keystone 250×21.2 mm 8μ Hypersil® HS C18 column; 5% CH3CN/50 mM aqueous ammonium acetate hold for 5 min; 5-50% CH3CN/50 mM aqueous ammonium acetate over 20 min; 50-100% CH3CN/50 mM aqueous ammonium acetate over 1 min; hold at 100% CH3CN for 5 minutes, 21 mL/min) to yield 4-(7-Benzo[b]thiophen-2-yl-1H-indazol-5-ylamino)-pyrimidin-2-ol (2 mg, 0.006 mmol, 0.1% yield): LC/MS Rt (Method e) 1.3 min; m/z (M−H) 358.3.
  • Example #9 {2-[5-(2-Amino-pyrimidin-4-ylamino)-1H-indazol-7-yl]-1H-indol-7-yl}-methanol
  • Figure US20120053345A1-20120301-C00921
  • 2-[5-(2-Amino-pyrimidin-4-ylamino)-1H-indazol-7-yl]-1H-indole-7-carboxylic acid methyl ester (Preparation #F.8.1, 0.100 g, 0.000250 mol) was added to a solution of lithium aluminium hydride in tetrahydrofuran (1.0M, 1.0 mL) at about 0° C. in 3 portions. Additional THF (1.0 mL) was added after about 10 min., and after about 40 min, the mixture was allowed to warm to ambient temperature. After about a further 60 min, lithium aluminium hydride in THF (1.0M, 1 mL) was added. About 16 hours later, the mixture was quenched with water and neutralized with a saturated aqueous solution of ammonium chloride. The mixture was diluted with EtOAc, and the layers were separated. The aqueous layer was further extracted with EtOAc, and the combined organics were dried over anhydrous MgSO4 and concentrated to yield {2-[5-(2-amino-pyrimidin-4-ylamino)-1H-indazol-7-yl]-1H-indol-7-yl}-methanol (28 mg, 30% yield); LC/MS Rt (Method e) 0.8 min; m/z (M−H) 370.5.
  • Example #10 2-[5-(2-Amino-pyrimidin-4-ylamino)-1H-indazol-7-yl]-1H-indole-7-carboxylic acid amide
  • Figure US20120053345A1-20120301-C00922
  • A solution of 2-[5-(2-amino-pyrimidin-4-ylamino)-1H-indazol-7-yl]-1H-indole-7-carboxylic acid (Example #F.5.11, 0.100 g, 0.000259 mol) and N,N-carbonyldiimidazole (0.0842 g, 0.000519 mol) in N,N-dimethylformamide (2.5 mL, 0.032 mol) was heated at about 55° C. for about 45 min. The mixture was cooled with an ice bath and treated with ammonia gas for about 15 min at about 0° C. The mixture was sealed and allowed to warm to ambient temperature. After about 1 hour the mixture was opened to atmosphere, and a 1.2 mL portion of the mixture was purified by reverse phase preparative HPLC (Thermo Hypersil-Keystone 250×21.2 mm 8μ Hypersil® HS C18 column; 5% CH3CN/50 mM aqueous ammonium acetate hold for 5 min; 5-50% CH3CN/50 mM aqueous ammonium acetate over 20 min; 50-100% CH3CN/50 mM aqueous ammonium acetate over 1 min; hold at 100% CH3CN for 5 minutes, 21 mL/min) then filtered to remove the remaining acid. The filtrate was then lyophilized to yield 2-[5-(2-amino-pyrimidin-4-ylamino)-1H-indazol-7-yl]-1H-indole-7-carboxylic acid amide (2 mg, 2% yield): LC/MS Rt (Method e) 0.7 min; m/z (M+H)+ 385.4.
  • Preparation #22a 5-Bromo-7-iodo-1H-indazole
  • Figure US20120053345A1-20120301-C00923
  • A 1 L round bottom flask equipped with a stir bar was charged with 4-bromo-2-methylaniline (Aldrich, 24.3 g, 0.130 mmol) and dichloromethane (250 mL). bis(pyridine)iodonium(I)tetrafluoroborate (50.0 g, 0.13 mmol) was added in four portions over approximately 10 minutes. The mixture was stirred for about 30 min at ambient temperature then two additional portions of the bis(pyridine)iodonium(I)tetrafluoroborate (2.5 g each) were added and the reaction was continued for about 1 h. Water (150 mL) was added and after stirring for about 10 minutes the mixture was transferred to a separatory funnel and the layers separated. The organic solution was then washed with saturated aqueous Na2S2O3 (100 mL) then water (50 mL). The organic solution was dried over anhydrous magnesium sulfate then filtered and evaporated to give an oil (51.4 g) which was applied to a silica gel column (400 g) and eluted with heptane/dichloromethane/ethyl acetate (12:7:1) to give 4-bromo-2-iodo-6-methyl-phenylamine (31.08 g, 76.6%) as a dark solid.
  • 4-bromo-2-iodo-6-methyl-phenylamine (31.08 g, 99.6 mmol) was dissolved in glacial acetic acid (400 mL) in a 1 L round bottom flask equipped with a stir bar. The flask was then charged in one portion with sodium nitrite (7.56 g, 109.6 mmol) dissolved in water (18.7 mL). The mixture was stirred for about 15 minutes then the solvents were removed by evaporation on a rotovap at a bath temperature of about 35° C. The residue was stirred with water (200 mL) for 15 minutes and the resulting solid was collected by filtration and dried to yield 5-bromo-7-iodo-1H-indazole (30.6 g, 95%) as a brownish-tan solid; (DMSO-d6, 400 MHz) δ 13.6 (bs, 1H), 8.24 (s, 1H), 8.04 (d, J=1.54 Hz, 1H), and 7.88 (d, J=1.54 Hz, 1H); RP-HPLC (Table 1, Method e) Rt=3.63 min; m/z: (M−H) 322.8.
  • Preparation #22b N,N-Dimethyl-1-[5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-indol-3-yl]methanamine
  • Figure US20120053345A1-20120301-C00924
  • To a suspension of Eschenmoser's salt (0.91 g, 4.9 mmol) in CH3CN (3 mL) and HOAc (1.5 mL) at rt was added a solution of 5-(4,4,5,5-tetramethyl-[1,3,2]dioxaborolan-2-yl)-1H-indole (1.0 g, 4.1 mmol) in CH3CN (3 mL) dropwise over about 10 min. After about 2 hours, the reaction was quenched with saturated aqueous NaHCO3 and extracted with CH2Cl2 (3×15 mL). The combined organic layers were washed with brine, dried over Na2SO4, decanted, and concentrated to give crude N,N-Dimethyl-1-[5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-indol-3-yl]methanamine (0.60 g, 50%); RP-HPLC (Table 1, Method g) Rt 1.39 min; 1H NMR (d6-DMSO, 400 MHz) δ11.26 (br s, 1H), 8.07 (s, 1H), 7.44 (d, J=8.2 Hz, 1H), 7.37 (m, 2H), 3.92 (s, 2H), 2.40 (s, 6H), 1.31 (s, 12H).
  • Preparation #22c Thieno[2,3-b]pyridin-2-ylboronic acid
  • Figure US20120053345A1-20120301-C00925
  • To a solution of thieno[2,3-b]pyridine (0.50 g, 3.7 mmol) in THF (25 mL) at about −78° C. was added n-BuLi (1.6 M in hexane, 2.5 mL, 4.1 mmol) dropwise over about 10 min. After about 45 min, added triisopropyl borate (0.93 mL, 4.1 mol). Stirred for about 1 hour at about −78° C. then at about 0° C. for about 30 min. 10% aqueous HCl (10 mL) was added and the ice bath was removed. After about 30 min, EtOAc (30 mL) was added. The reaction mixture was filtered to remove a precipitate that was washed with additional water and EtOAc. The combined layers were separated and the aqueous layer was extracted with additional EtOAc (2×30 mL). The combined organic layers were washed with brine, dried over anhydrous Na2SO4, decanted, and concentrated to get a solid that was triturated with EtOAc and heptane then filtered to give thieno[2,3-h]pyridin-2-ylboronic acid (0.043 g, 6%); RP-HPLC (Table 1, Method g) Rt 1.67 min, m/z (ESI+) 180.1 (M+H)+.
  • Preparation #22d 2-Amino-4-chloro-thieno[2,3-d]pyrimidine-6-carboxylic acid methyl ester
  • Figure US20120053345A1-20120301-C00926
  • Step 1 Preparation #22e (2-amino-6-chloro-5-formyl-pyrimidin-4-ylsulfanyl)-acetic acid methyl ester
  • To a mixture of 2-amino-4,6-dichloro-5-pyrimidinecarbaldehyde (Apin, 2.00 g, 10.4 mmol) in dioxane (20 mL) was added triethylamine (1.60 mL, 11.5 mmol) and methyl thioglycolate (1.90 mL, 21.2 mmol). After about 3 hours, the reaction mixture was diluted with water (30 mL) and then filtered, washed with additional water to give (2-amino-6-chloro-5-formyl-pyrimidin-4-ylsulfanyl)-acetic acid methyl ester (1.801 g, 66%) as a yellow solid; RP-HPLC (Table 1, Method a) Rt 1.85 min, m/z (ESI+) 262.0 (M+H)+.
  • Step 2 Preparation #22d 2-Amino-4-chloro-thieno[2,3-d]pyrimidine-6-carboxylic acid methyl ester
  • To a mixture of (2-amino-6-chloro-5-formyl-pyrimidin-4-ylsulfanyl)-acetic acid methyl ester (Preparation #22e, 0.20 g, 0.76 mmol) in dioxane (7.6 mL) was added K2CO3 (0.21 g, 1.5 mmol). The resulting mixture was heated at about 100° C. overnight, cooled to room temperature and water (1 mL) added to dissolve base. The remaining solid was filtered and washed with MeOH to give 2-amino-4-chloro-thieno[2,3-d]pyrimidine-6-carboxylic acid methyl ester (0.16 g, 84%) as a pale yellow solid; RP-HPLC (Table 1, Method a) Rt 2.38 min; 1H NMR (d6-DMSO, 400 MHz) δ7.82 (s, 1H), 7.72 (br s, 2H), 3.86 (s, 3H).
  • Preparation #50 7-Benzo[b]thiophen-2-yl-5-nitro-1H-indazol-3-ol
  • Figure US20120053345A1-20120301-C00927
  • A mixture of 3-Benzo[b]thiophen-2-yl-2-chloro-5-nitro-benzoic acid (0.11 g, 0.33 mmol), hydrazine (0.35 mL) and isopropyl alcohol (3 mL) was heated to about 90° C. for about 1 hour. The mixture was cooled then the solvents were evaporated under reduced pressure. The residue was treated with 6N aqueous hydrochloric acid (5 mL) the mixture was extracted with ethyl acetate (3×30 mL). The organic extracts were combined then concentrated to give 7-Benzo[b]thiophen-2-yl-5-nitro-1H-indazol-3-ol RP-HPLC (Table 1, Method e) Rt 1.41 min; m/z: (M−H) 310.24.
  • Example #51 2-(7-Bromo-1H-indazol-5-ylamino)-2-methyl-propan-1-ol
  • Figure US20120053345A1-20120301-C00928
  • To a solution of 2,2-dimethyl-oxirane (0.100 mL, 1.13 mmol) in THF (5.0 mL) was added 7-bromo-1H-indazol-5-ylamine (Preparation #6, 0.100 g, 0.472 mmol) and samarium chloride (0.012 g, 0.047 mmol). The reaction mixture was stirred at ambient temperature for about 20 hours followed by removal of THF in vacuo. The crude reaction mixture was purified by reverse phase chromatography (RP-HPLC (Rainin C18, 8 mm, 300 Å, 35 cm; 5-100% acetonitrile/0.1 M ammonium acetate over 20 min, 100% acetonitrile hold 10 minutes, 21 mL/min) to afford 2-(7-bromo-1H-indazol-5-ylamino)-2-methyl-propan-1-ol (0.033 g, 0.116 mmol). RP-HPLC (Table 1, Method e) Rt 1.18 min; m/z: (M+H)+ 286.1.
  • Preparation #52 (7-Benzo[b]thiophen-2-yl-1H-indazol-5-yl)-phenyl-methanone
  • Figure US20120053345A1-20120301-C00929
  • A mixture of 7-benzo[b]thiophen-2-yl-5-bromo-1H-indazole (Preparation #26, 0.25 g, 0.76 mol), tributylphenyltin (0.56 g, 1.52 mmol), triethylamine (0.23 g, 2.28 mmol) and trans-dichloro[bis(triphenylphosphine)]palladium (II) (0.08 g, 0.114 mmol) in N,N-dimethylformamide (4 mL) was purged with carbon monoxide then stirred under an atmosphere of carbon monoxide at about 90° C. in an oil bath for about 2 hours. The solvent was removed in vacuo and the residue was applied to a silica gel column then eluted with dichloromethane/methanol (95:5). The fractions that contained product were combined and triturated with heptane then the solids were collected by filtration and purified by preparative reverse phase HPLC to give (7-Benzo[b]thiophen-2-yl-1H-indazol-5-yl)-phenyl-methanone (23 mg, 8.5%); (DMSO-d6, 400 MHz) δ 13.85 (bs, 1H), 8.49 (bs, 1H), 8.27 (s, 1H), 8.18 (bs, 1H), 8.06 (m, 2H), 7.93 (m, 1H), 7.81 (m, 2H), 7.71 (m, 1H), 7.61 (m, 2H), 7.45 (m, 2H); RP-HPLC (Table 1, Method e) Rt 2.50 min; m/z: (M+H)+353.0.
  • Preparation #53 (7-Benzo[b]thiophen-2-yl-1H-indazol-3-yl)-pyridin-2-yl-methanone
  • Figure US20120053345A1-20120301-C00930
  • A mixture of 7-benzo[b]thiophen-2-yl-5-bromo-2-(2-trimethylsilanyl-ethoxymethyl)-2H-indazole (Preparation #23, 0.15 g, 0.326 mmol) in tetrahydrofuran (2 mL) was cooled to about −65° C. then a solution of n-butyllithium (1.6 M, 0.36 mL, 0.36 mmol) was added dropwise. After about 30 minutes, 2-pyridinecarboxaldehyde (0.042 g, 0.39 mmol) was added. The mixture was quenched with methanol (1 mL) then warmed to room temperature. The solvent was removed in vacuo and the residue was purified by preparative reverse phase HPLC chromatography (RP-HPLC (Rainin C18, 8 mm, 300 Å, 35 cm; 5-100% acetonitrile/0.1 M ammonium acetate over 20 min, 100% acetonitrile hold 10 minutes, 21 mL/min) to give the intermediate [7-benzo[b]thiophen-2-yl-2-(2-trimethylsilanyl-ethoxymethyl)-2H-indazol-3-yl]-pyridin-2-yl-methanol (50 mg) which was suspended in a mixture of 1,4-dioxane (2 mL) and manganese dioxide (90 mg, 31%). The mixture was heated to about 90° C. for 15 minutes then filtered and evaporated. The residue was purified by reverse phase HPLC(RP-HPLC (Rainin C18, 8 mm, 300 Å, 35 cm; 5-100% acetonitrile/0.1 M ammonium acetate over 20 min, 100% acetonitrile hold 10 minutes, 21 mL/min) to give (7-Benzo[b]thiophen-2-yl-1H-indazol-3-yl)-pyridin-2-yl-methanone (22 mg, 44%); (DMSO-d6, 400 MHz) δ 8.8 (bs, 1H), 8.31 (d, 1H), 8.09 (m, 4H), 7.93 (d, 1H), 7.78 (bs, 1H), 6.96 (bs, 1H), 7.51 (t, 1H), 7.43 (m, 2H); RP-HPLC (Table 1, Method e) Rt 2.60 min; m/z: (M−H) 353.8.
  • Preparation #54 (7-Benzo[b]thiophen-2-yl-1H-indazol-5-yl)-pyridin-2-yl-methanone
  • Figure US20120053345A1-20120301-C00931
  • Starting with 7-benzo[b]thiophen-2-yl-5-bromo-1-(2-trimethylsilanyl-ethoxymethyl)-1H-indazole (Preparation #23, 0.2 g, 0.435 mmol) and using the procedure for the preparation of (7-benzo[b]thiophen-2-yl-1H-indazol-3-yl)-pyridin-2-yl-methanone (Preparation #52 above) provided (7-benzo[b]thiophen-2-yl-1H-indazol-5-yl)-pyridin-2-yl-methanone (17 mg, 11%); (DMSO-d6, 400 MHz) δ 8.79 (d, 1H), 8.65 (s, 1H), 8.54 (s, 1H), 8.27 (s, 1H), 8.24 (s, 1H), 8.11 (m, 1H), 8.05 (m, 2H), 7.95 (m, 1H), 7.72 (m, 1H), 7.45 (m, 2H); RP-HPLC (Table 1, Method e) Rt 2.28 min; m/z: (M−H) 354.1.
  • Figure US20120053345A1-20120301-C00932
  • Preparation #55 7-(7-Benzo[b]thiophen-2-yl-3-methyl-1H-indazol-5-yl)-5H-pyrrolo[3,2-d]pyrimidin-2-ylamine
  • Figure US20120053345A1-20120301-C00933
  • A mixture of 7-benzo[b]thiophen-2-yl-5-bromo-2-(2-trimethylsilanyl-ethoxymethyl)-2H-indazole (Preparation #24, 1.0 g, 2.18 mmol) in tetrahydrofuran (15 mL) was cooled to about −65° C. then a solution of n-butyllithium (1.6 M, 1.5 mL, 2.4 mmol) was added dropwise. After about 5 minutes methyliodide (0.37 g, 2.6 mmol) was added then the solution was warmed to about −20° C. Saturated ammonium chloride (1 mL) was added then the mixture was diluted with ethyl acetate (25 mL) and water (10 mL). The layers were separated then the organic layer was dried over magnesium sulfate, filtered and evaporated to give the crude 7-benzo[b]thiophen-2-yl-5-bromo-3-methyl-2-(2-trimethylsilanyl-ethoxymethyl)-2H-indazole which was converted to 7-benzo[b]thiophen-2-yl-3-methyl-5-(4,4,5,5-tetramethyl-[1,3,2]dioxaborolan-2-yl)-2-(2-trimethylsilanyl-ethoxymethyl)-2H-indazole (0.29 g, 25% over 2 steps) using general procedure E. The boronate (200 mg, 0.385 mmol) was coupled with 2-(dimethylamino-methyleneamino)-7-iodo-pyrrolo[3,2-d]pyrimidine-5-carboxylic acid tert-butyl ester (0.135 g, 0.32 mmol) using general procedure F and the resulting product deprotected using general procedure G to give 7-(7-benzo[b]thiophen-2-yl-3-methyl-1H-indazol-5-yl)-5H-pyrrolo[3,2-d]pyrimidin-2-ylamine (24 mg, 19%); (DMSO-d6, 400 MHz) δ 12.48 (bs, 1H), 11.44 (s, 1H), 8.50 (m, 3H), 8.18 (d, 1H), 8.07 (bs, 1H), 8.02 (d, 1H), 7.90 (d, 1H), 7.43 (m, 2H), 5.94 (bs, 2H), 2.61 (s, 3H); RP-HPLC (Table 1, Method e) Rt 1.76 min; m/z: (M−H) 394.9.

Claims (9)

What is claimed is:
1. A compound of Formula (I)
Figure US20120053345A1-20120301-C00934
wherein
R1 is selected from the group consisting of H, benzyl substituted with OCH3, optionally substituted (C1-C3)alkyl, pyrimidine substituted with NH2 and amino(C1-C3)alkyl;
R3 is selected from the group consisting of H, halogen, NH2, OH, COOH, —C(O)—NH—CH2—C(O)—OCH3, —NH—CH2-phenyl, —C(O)-pyridinyl, —NH—C(O)-cyclobutyl and —NH—C(O)-phenyl wherein phenyl is optionally substituted with either N(CH3)2 or OCH3; or
R3 is selected from the optionally substituted group consisting of (C1-C6)alkyl, benzo[b]thienyl, 2,3-dihydrobenzofuranyl, indolyl, isoquinolinyl, morpholinyl, naphthyl, phenyl, piperazinyl, pyrazolyl, pyridinyl, pyrimidinyl, pyrrolyl, quinolinyl and thienyl;
wherein the substituent is selected from one or more CH3, NH2, Cl, F, dimethylamino, OH, CH2OH, —C(O)NH2, COOH, CF3, isopropyl, OCF3, OCH3, —O—CH2-phenyl, CN, OCH2CH3, —NH—C(O)-cyclobutyl, —NH—C(O)-phenyl, NH—C(O)—CH3, NHC(O)CH3, N(CH3)2, S(O)2CH3 and C(O)NH-phenyl; or
R3 is —C(O)—NY100—(C(Y100)2)x—Ra wherein
x is 0, 1, 2 or 3;
Y100 is independently H or (C1-C3)alkyl; and
Ra is —C(O)—CH3 or is selected from the optionally substituted group (C1-C3)alkyl, amino, aminoalkyl, benzimidazolyl, benzo[b]thienyl, benzotriazolyl, biphenyl, 1,3-dihydrobenzimidazolyl, 1,3-dihydrobenzimidazolyl-2-one, imidazolyl, indolyl, naphthyl, phenyl, pyrazolyl, pyridinyl, pyrimidinyl, tetrahydropyranyl and thiazolyl; or
R3 is A-B wherein A is connected to the indazole and
A is selected from the group consisting of —C≡C, —C≡C-phenyl, indazolyl, phenyl, pyridinyl and thienyl;
B is selected from the group consisting of benzyloxy, morpholinyl, phenyl, thienyl, t-butyl, —NH—C(O)-cyclobutyl and —NH—C(O)-phenyl;
R4 is H or NH2;
R5 is selected from the group consisting of H, NH2, NO2, halo; or
R5 is selected from the optionally substituted group consisting of benzimidazolyl, 3,4-dihydrobenzo[1,4]thiazinyl, benzyloxyphenyl, furo[3,2-c]pyridine, indazolyl, indolyl, isoquinolinyl, phenyl, piperidinyl, pyrazolo[3,4-d]pyrimidine, pyrazinyl, pyrazolyl, pyridinyl, pyrrolo[2,3-b]pyridinyl, pyrrolo[2,3-c]pyridinyl, pyrrolo[2,3-d]pyridinyl, pyrimidinyl, pyrrolo[3,2-d]pyridine, pyrrolo[2,3-d]pyrimidinyl, pyridinyl, pyrrolyl, quinolinyl, quinazolinyl, thienyl, thieno[2,3-c]pyridinyl, thieno[2,3-d]pyrimidine, thieno[3,2-c]pyridine, 7-azaindolinyl and 7-azaindolyl; or
R5 is —C(O)—Rb; wherein
Rb is selected from the group consisting of OH, (C1-C3)alkoxy, phenyl, optionally substituted piperidinyl, optionally substituted pyridinyl and optionally substituted pyrrolidinyl; or
Rb is D-E wherein D is attached to the C(O) and
D is selected from the group consisting of piperidinyl and pyrrolidinyl;
E is selected from the group consisting of pyridinyl and pyrimidinylaminemethyl;
R5 is —C(O)—NH—(CH2)a—Rc; wherein
a is 0, 1, 2 or 3;
Rc is —CONH2 or
Rc is selected from the optionally substituted group consisting of benzimidazolyl, benzothiazolyl, benzo[b]thiophenyl, dimethylamino, fluorene, imidazolyl, indanyl, indazolyl, isoxazolyl, oxazolyl, phenyl, piperidinyl, pyrazolyl, pyridinyl, quinazolinyl, thiadiazolyl and 1,2,4-triazolyl; or
Rc is J100-J200 wherein J100 is attached to (CH2)x and
J100 is selected from the optionally substituted group consisting of isoxazolyl, piperazinyl, pyrazolyl pyridinyl, and phenyl; and
J200 is selected from the group consisting of benzimidazolyl, benzoxazolyl, cyclohexyl, furanyl, imidazo[1,2-a]pyridinyl, indolyl, isoxazolyl, —NH—C(O)-phenyl, phenoxy and optionally substituted phenyl;
R5 is —NH—C(O)—(CH2)n—Rd; wherein
n is 0 to 3;
Rd is —C(CH3)2—CH2—C(O)—CH3; or
Rd is selected from the optionally substituted group of (C1-C2)alkoxy, alkylamino, benzimidazolyl, benzo[1,3]dioxazolyl, benzo[1,2,5]oxadiazolyl, benzotriazolyl, benzo[b]thienyl, benzofuranyl, benzyloxy, cyclopropyl, cyclohexyl, chromenyl, dimethylamino, furanyl, hexahydropyrimidinyl, imidazolyl, imidazo[2,1-b]thiazolyl, imidazo[2,1-b]thiazolyl, imidazolidinyl, indolyl, isoxazolyl, morpholinyl, phenyl, piperazinyl, piperidinyl, pyrazinyl, pyrazolyl, pyrazolo[1,5-a]pyrimidinyl, pyridazinyl, pyridinyl, pyrimidinyl, pyrrolidinyl, pyrrolyl, quinolinyl, quinoxalinyl, tetrahydrobenzofuranyl, tetrahydrofuranyl, thiazolyl, thieno[2,3-d]pyrimidinyl, thienyl, 2,3-dihydrothiazolo[3,2-a]pyrimidine, —S-pyrimidinyl, —O-phenyl, —O—Si(CH3)2—C(CH3)3, —NH—S(O)2-phenyl, —NH—C(O)—NH2, 1,2,3,4-tetrahydronaphthyridinyl or N(CH3)2; or
Rd is M-Q wherein M is attached to the (CH2)n and
M is selected from the group consisting of optionally substituted methylene, cyclopropylidene, optionally substituted isoxazolyl, phenyl, pyrazolyl, —NH—C(O) and optionally substituted 1,3,5-triazinyl;
Q is selected from the group consisting of furanyl, morpholinyl, phenyl, phenylamine and optionally substituted 1,3,4-thiadiazolyl;
R5 is —NH—CH2—C(Y200)2—Re wherein Y200 is independently H or (C1-C3)alkyl and Re is selected from the optionally substituted group of (C1-C6)alkoxy, imidazolyl, phenyl, piperidinyl, pyrrolidinyl and 1,2,3,4-tetrahydro[1,8]naphthyridine; or
R5 is —NH—C(O)—N(Rf)2 wherein Rf is independently H or optionally substituted (C1-C3)alkyl; or
R5 is —NH—(C(O))m—NY300—(CH2)p—Rg; wherein
m is 0, 1 or 2;
Y300 is H or optionally substituted (C1-C3)alkyl;
p is 1 or 2; and
Rg is selected from the optionally substituted group of amino, (C1-C2)alkoxy, benzo[1,3]dioxazolyl, benzothiazolyl, benzo[1,4]oxazinyl, benzo[1,2,5]thiadiazolyl, imidazol[1,2-a]pyridinyl, imidazolyl, isoxazolyl, morpholinyl, oxazolyl, phenyl, piperidinyl, pyrazinyl, pyrazolyl, pyridinyl, pyrrolyl, tetrahydropyranyl, thiazolyl and triazolyl; or
Rg is W—X wherein W is attached to the (CH2)p and
W is thiazolyl and X is thienyl;
R5 is —NH—S(O)2Rh wherein Rh is selected from the group of optionally substituted benzo[1,2,5]oxodiazolyl, benzo[1,2,5]thiadiazolyl, imidazolyl, isoxazolyl, oxazolyl, benzo[1,4]oxazinyl, pyrazolyl, phenyl, quinolinyl, thiazolyl, thienyl and thienyl; or
Rh is T-U wherein T is attached to the S(O)2 and
T is phenyl or thienyl;
U is selected from the group consisting of pyridinyl, optionally substituted thiazolyl and —NH-pyrimidinyl wherein the pyrimidinyl can be optionally substituted; or
R5 is —N(Y400)—Ri wherein
Y400 is H or Y400 is selected from the optionally substituted group consisting of (C1-C3)alkyl, amino(C1-C3)alkyl and pyridinylmethyl; and
Ri is selected from the optionally substituted group of (C1-C3)alkyl, 6-azaindolyl cyclobutenyl, phenyl, purinyl, pyrazinyl, pyrazolo[1,5-a]pyrimidine, pyridazinyl, pyridinyl, pyrimidinyl, pyrrolo[2,3-c]pyridinyl, pyrrolo[2,3-b]pyrimidinyl, pyrrolo[3,2-d]pyrimidinyl, pyrrolo[3,4-b]pyrimidinyl, pyrrolo[2,3-d]pyrimidinyl, quinazolinyl, thieno[3,2-d]pyrimidinyl, thieno[3,2-b]pyridinyl, and triazinyl; or
Ri is V—W wherein V is attached to the nitrogen and
V is a bond or is selected from the optionally substituted group consisting of isoquinolinyl, pyridinyl, pyrimidinyl and pyrrolo[3,2-d]pyrimidinyl;
W is selected from the optionally substituted group consisting of (C1-C3)alkyl, alkoxyalkyl, cyclopentyl, morpholinyl, phenyl, pyrimidinyl, pyrrolidinyl, thieno[3,2-b]pyridinyl, —NH-phenyl, —NH—CH2—CH2—N(CH3)2, —NH—NH2, NH—C(O)—CH3, CH2-phenyl, NH—C(O)-furanyl, S-isopropyl, S-naphthyl, S-phenyl and S—CH2—CH2—NH2; or
R5 is Z100—Z200 wherein Z100 is attached to the indazole and
Z100 is selected from the group consisting of butyl, ethyl, indazolyl, optionally substituted phenyl, optionally substituted pyridinyl, optionally substituted pyrimidinyl, pyrrolo[3,2-d]pyrimidinyl and optionally substituted thienyl;
Z200 is selected from the group consisting of —C(O)—NH—CH2CH2—NH2, NH2, —NH—C(O)-thienyl, —NH—C(O)—CH(CH3)2, —NH—C(O)—CH3, —NH—C(O)C(CH3)3, —NH—C(O)—NH-furanyl, —NH—C(O)—NH-phenyl, benzo[b]thienyl, morpholinylethyl, phenyl, piperazinyl, piperidinyl, pyrazolyl and tetrazolyl; or
R5 is —NH—C(O)—Y500—C(O)—Rk; wherein
Y500 is optionally substituted (C1-C3)alkyl; and
Rk is H or Rk is selected from the optionally substituted group consisting of phenyl, phenylamino and thienyl; or
R5 is —NH—C(O)—(CH2)y—NH-T-Rm; wherein
y is 1 or 2;
T is C(O) or S(O)2; and
Rm is selected from the group consisting of furanyl, phenyl and thienyl;
R6 is H or R6 is selected from the optionally substituted group consisting of (Ci)alkoxy, (C1-C3)alkyl, benzo[b]thienyl, —NH-pyrimidinyl, —NH—S(O)2-phenyl-NH-pyrimidinyl, —NH—C(O)-benzo[b]thienyl, pyrrolo[2,3-b]pyrimidinyl and pyridinyl; and
R7 is selected from the group consisting of H, halo, NH2, or R7 is selected from optionally substituted group consisting of (C2-C5)alkenyl, (C2-C5)alkynyl, aminoalkynyl, benzofuranyl, benzothiazolyl, benzo[b]thienyl, furanyl, indolyl, isoquinolinyl, naphthyl, phenyl, phenylalkyl, phenyl(C2-C5)alkenyl, phenyl(C2-C5)alkynyl, pyridinyl, pyrimidinyl, pyrrolyl, quinolinyl, quinoxalinyl, thieno[2,3-b]pyridinyl, thienyl, —NH—S(O)2—CH3, —NH—C(O)—CH3, —NH—C(O)-phenyl, —C(O)—NH—CH2-phenyl and —C(O)—NH-phenyl; or
R7 is Y—Z wherein Y is attached to the indazole; and
Y is benzo[b]thienyl or thienyl; and
Z is selected from the group consisting of phenyl, thienyl, CH2NHCH2CH2-morpholinyl and substituted piperazinyl; or
R7 is —C(O)—NH—(CH2)r-phenyl wherein r is 0 or 1 and the phenyl is optionally substituted;
provided that the compound is not
Figure US20120053345A1-20120301-C00935
wherein
R3 is selected from the group consisting of H, OH and COOH;
R5 is H or NO2;
R7 is H or NH2
R100 is OCH3 and
R200 is H or —C(O)—OCH3;
provided that the compound is not
Figure US20120053345A1-20120301-C00936
wherein
R3 is NH2 or phenyl; and
R5 is H or NO2;
provided that the compound is not
Figure US20120053345A1-20120301-C00937
wherein
R1 is H, methyl or propyl;
R7 is H, F or methyl; and
R is H, methyl, OH, NH, or OCH3;
provided that the compound is not
Figure US20120053345A1-20120301-C00938
wherein R3 is selected from the group consisting of I, Br, COOH, NH2, thienyl, pyridinyl, pyrrolyl, —C(O)—NH—CH—(CH2OH)2, —C(O)—NH—CH2—X100,
Figure US20120053345A1-20120301-C00939
wherein X100 is pyridinyl or phenyl optionally substituted with methyl;
provided that the compound is not
Figure US20120053345A1-20120301-C00940
wherein
R3 is morpholinyl or 4-methylpiperazinyl;
Rc is H, C1 or NO2;
R6 is H or C1 and
R7 is H, C1 or NO2;
provided that the compound is not
Figure US20120053345A1-20120301-C00941
wherein
R6 is H, methyl or OCH3;
R6 is H or OCH3;
L100 is H or isopropyl; and
X200 is phenyl or 4-chlorophenyl;
provided that the compound is not
Figure US20120053345A1-20120301-C00942
wherein
R1 is H or CH3 and X300 is benzyl, phenyl, 2-aminophenyl or 2-hydroxyphenyl; and
provided that the compound is not
Figure US20120053345A1-20120301-C00943
wherein
R1 is H, CH2OH, methyl, phenyl or 4-methoxybenzyl;
R3 is H, I, pyridinyl or
Figure US20120053345A1-20120301-C00944
and
R5 is H, Br, F, C(OH), —C(O)—OCH2CH3 or —NH—C(O)—NH-benzyl.
2. The compound of claim 1 wherein
R1 is H or pyrimidinyl substituted with NH2;
R3 is selected from the group consisting of H, CH3, OH, Cl, benzo[b]thienyl, 2,3-dihydrobenzofuranyl, indolyl, naphthyl, phenyl, pyrazolyl, pyridinyl, pyrimidinyl, pyrrolyl, quinolinyl, thienyl, —NH—C(O)-cyclobutyl and —NH—C(O)-phenyl; wherein
the indolyl is optionally substituted with CH3;
the naphthyl is optionally substituted with OCH3 or OH; and
the phenyl optionally substituted with one or more substituents selected from the group consisting of CH3, NH2, Cl, F, N(CH3)2, OH, CH2OH, C(O)NH2, COOH, CF3, OCF3, OCH3, CN, OCH2CH3, NHC(O)CH3, —S(O)2CH3 and —C(O)—NH-pheny; or
R3 is —C(O)—NY100—(C(Y100)2 ) x—Ra wherein
x is 0 or 1;
Y100 is H;
Ra is selected from the optionally substituted group consisting of benzo[b]thienyl, benzimidazolyl, 1,3-dihydrobenzimidazolyl-2-one, benzotriazolyl, biphenyl, 1,3-dihydrobenzimidazolyl, indolyl, naphthyl and phenyl; wherein
the naphthyl is substituted with OH or OCH3;
the phenyl is optionally substituted with one or more Cl, F, OH, CH2OH, CH2CH2OH, COOH, C(O)NH2, N(CH3)2 or methyl; or
R3 is A-B wherein
A is selected from the group consisting of —C≡C, —C≡C-phenyl, phenyl and thienyl; and
B is selected from the group consisting of benzyloxy, phenyl, thienyl, —NH—C(O)-cyclobutyl and —NH—C(O)-phenyl;
R4 is H;
R5 is pyridinyl substituted with C(O)H, CH2OH, SCH2CH2NH2 or NH2; or
R5 is selected from the group consisting of
Figure US20120053345A1-20120301-C00945
 wherein
E is selected from the group consisting of H, OH, CH3, —CH2CH2NH2, CH2CH2CH2OH, CH2CH2OCH3, CH2CH2CH2NH2, CH2C(O)OH, CH2CH2C(O)OH, CH2CH2C(O)NH2, CH2CH2C(O)OCH3, CH2CH2CH2OCH3, NHCH2CH2CH3, CH2CH2C(O)NH(CH3), CH2CH2C(O)N(CH3)2, C(O)NHCH2CH2NH(CH3), NHCH2CH2OCH3, NHCH2CH2OH, NHCH2CH2N(CH3)2, isopropyl, CH2C(O)NH2, CH2CH(CH3)C(O)OH, CH2CH2CH2C(O)OH, CH2CH(CH3)C(O)OCH3, CH2CH2CH2CH2NH2, CH2CH2CH2C(O)NH2, N(CH3)2, morpholinylethyl, piperidinylethyl,
Figure US20120053345A1-20120301-C00946
 and 4-methylpiperazinylcyclohexyl;
E300 is H or CH2CH2OCH3;
G is selected from the group consisting of H, C1, NH2, CH2CH2C(O)NHCH2CH2NH2, C(O)NH2, C(O)NHCH2CH2NH2, C(O)NHCH2CH2N(CH3)2, CH2CH2NH2, CH2CH2CH2NH2, CH2CH2C(O)OH, CH2CH2C(O)NH2, NHCH2CH2N(CH3)2, NHCH2CH2-pyridinyl and NHCH2CH2NH2; or
R5 is —C(O)—NH—(CH2)a—Rc wherein
a is 0
Rc is benzimidazolyl or fluorene substituted with oxo; or
Rc is J100-J200 wherein
J100 is selected from the group consisting of pyrazolyl, pyridinyl, piperazinyl and phenyl; wherein
the phenyl is optionally substituted with one or more substituents selected from the group consisting of F, OH and OCH3;
the piperazinyl is substituted with methyl; and
J200 is selected from the group consisting of benzoxazolyl, benzimidazolyl, furanyl, imidazo[1,2-a]pyridinyl and 1,8a-dihydroimidazo[1,2-a]pyridinyl; or
R5 is —NH—C(O)—(CH2)n—Rd wherein
n is 0, 1 or 2; and
Rd is benzimidazolyl, benzo[b]thienyl, imidazolyl, phenyl or pyrazolo[1,5-a]pyrimidinyl; wherein
the phenyl is substituted with NO2;
R5 is —NH—(C(O))m—NY300—(CH2)p—Rg wherein
m is 2;
Y300 is H;
p is 1; and
Rg is benzo[1,3]dioxazolyl; or
R5 is N(Y400)—Ri wherein
Y400 is selected from H, CH2CH2CH2OH, CH2CH2NH2 or pyridinylmethyl;
Ri is V—W wherein
V is a bond or pyrimidinyl; and
W is pyrimidinyl substituted with NH2 or pyrrolidinyl substituted with OH; or
R5 is Z100—Z200 wherein
Z100 is thienyl or pyridinyl substituted with CH3 and Z200 is thienyl or NH—C(O)-furanyl;
R6 is selected from the group consisting of H, pyrrolo[2,3-b]pyrimidinyl and
Figure US20120053345A1-20120301-C00947
and
R7 is selected from the optionally substituted group consisting of H, Br, Cl, I, benzofuranyl, benzo[b]thienyl, furanyl, indolyl, naphthyl, phenyl, pyridinyl, pyrrolyl, quinolinyl, quinoxalinyl, thieno[2,3-b]pyridinyl, thienyl, —CH═CH-phenyl, —C≡C-phenyl, —C(O)—NH—CH2-phenyl and —C(O)—NH-phenyl wherein
the naphthyl is optionally substsituted with OH or OCH3;
the benzo[b]thienyl is optionally substituted with OH, CH3, OCH3, N(CH3)2, OH, CH2═CHNHCH3, CH2NH2, CH2CH2NH2 or CH2NHCH2CH2N(CH3)2;
the indolyl is substituted with C(O)N(CH(CH3)2)2, CH2OH, CH2C(O)NH2, COOH, C(O)NH2, N(CH3)2 or S(O)2CH3; and
the phenyl is optionally substituted with one or more substituents selected from the group consisting of Cl, F, CH3, CH2OH, CN, —C(O)NH2, OH, OCH3, N(CH3)2, NH—C(O)CH3, —NH—S(O)2—CH3;
the thienyl is substituted with CH2OH; or
R7 is Y—Z wherein
Y is benzo[b]thienyl or thienyl; and
Z is selected from the optionally substituted group consisting of CH═CHNHCH3, NHCH3, CH2NH2, CH2CH2NH2, CH2NHCH3, CH2NHCH2CH2N(CH3)2, N(CH3)2, CH2NHCH2CH2-morpholinyl, benzo[b]thienyl, morpholinylmethyl, piperazinylmethylphenyl and thienyl; or
R7 is —C(O)—NH—(CH2)r-phenyl wherein
r is 0 or 1;
the phenyl is optionally substituted with NH2.
3. The compound of claim 2 wherein
R1 is H or pyrimidinyl substituted with NH2;
R3 is selected from the group consisting of H, CH3, OH, Cl, benzo[b]thienyl, 2,3-dihydrobenzofuranyl, indolyl, naphthyl, phenyl, pyrazolyl, pyridinyl, pyrimidinyl, pyrrolyl, quinolinyl, thienyl, —NH—C(O)-cyclobutyl and —NH—C(O)-phenyl; wherein
the indolyl is optionally substituted with CH3;
the naphthyl is optionally substituted with OH; and
the phenyl optionally substituted with one or more substituents selected from the group consisting of OH, F, CH3, CF3, CN, —C(O)NH2, NH2, NHC(O)CH3, OCH3, OCF3, OCH2CH3, N(CH3)2, —C(O)—NH-phenyl and —S(O)2CH3; or
R3 is —C(O)—NY100—(C(Y100)2)x—Ra wherein
Y100 is H;
x is 0;
Ra is selected from the optionally substituted group consisting of benzimidazolyl, 1,3-dihydrobenzimidazolyl-2-one, benzotriazolyl, biphenyl, indolyl, naphthyl and phenyl; wherein
the naphthyl is substituted with OH or OCH3;
the phenyl is optionally substituted with one or more Cl, F, OH, CH2OH, CH2CH2OH, C(O)NH2, N(CH3)2 or methyl; or
R3 is A-B wherein
A is selected from the group consisting of phenyl and thienyl; and
B is selected from the group consisting of benzyloxy, phenyl, thienyl, —NH—C(O)-cyclobutyl and —NH—C(O)-phenyl;
R5 is pyridinyl substituted with NH2; or
R5 is selected from the group consisting of
Figure US20120053345A1-20120301-C00948
 wherein
E is selected from the group consisting of H, CH3, CH2C(O)OH, CH2CH2CH2OH, CH2CH2CH2NH2, CH2CH2C(O)OH, CH2CH2C(O)NH2, CH2CH2C(O)OCH3, CH2CH2CH2OCH3, NHCH2CH2CH3, CH2CH2C(O)NH(CH3), NHCH2CH2OCH3, NHCH2CH2OH, isopropyl, CH2C(O)NH2, CH2CH(CH3)C(O)OH, morpholinylethyl, piperidinylethyl, CH2CH2CH2C(O)OH, CH2CH(CH3)C(O)OCH3, CH2CH2NH2, CH2CH2CH2CH2NH2, CH2CH2CH2C(O)NH2, CH2CH2C(O)N(CH3)2, and N(CH3)2; and
G is selected from the group consisting of H, NH2, Cl, CH2CH2C(O)NHCH2CH2NH2, C(O)NHCH2CH2NH2, C(O)NHCH2CH2N(CH3)2, NHCH2CH2N(CH3)2, NHCH2CH2NH2, CH2CH2NH2, CH2CH2CH2NH2, CH2CH2C(O)OH, CH2CH2C(O)NH2 and NHCH2CH2-pyridinyl;
R5 is —C(O)—NH—(CH2)a—Re wherein
a is 0;
Rc is J100-J200 wherein
J100 is pyrazolyl or phenyl wherein the phenyl is optionally substituted with OCH3; and
J200 is benzoxazolyl, benzimidazolyl, furanyl, imidazo[1,2-a]pyridinyl or 1,8a-dihydroimidazo[1,2-a]pyridinyl; or
R5 is —NH—C(O)—(CH2)n—Rd wherein
n is 0, 1 or 2; and
Rd is selected from the group consisting of benzimidazolyl, benzo[b]thieny, imidazolyl and pyrazolo[1,5-a]pyrimidinyl;
R5 is —N(Y400)—Ri wherein
Y400 is selected from H, CH2CH2CH2OH, CH2CH2NH2 or pyridinylmethyl;
Ri is V—W wherein
V is a bond or pyrimidinyl and W is pyrimidinyl substituted with NH2 or pyrrolidinyl substituted with OH;
R5 is Z100—Z200 wherein
Z100 is thienyl;
Z200 is thienyl;
R6 is H or
Figure US20120053345A1-20120301-C00949
and
R7 is selected from the optionally substituted group consisting of H, benzofuranyl, benzo[b]thienyl, furanyl, indolyl, naphthyl, quinolinyl, phenyl, pyrrolyl, quinoxalinyl, thienyl, thieno[2,3-b]pyridinyl, —CH═CH-phenyl, —C≡C-phenyl, —C(O)—NH—CH2-phenyl and —C(O)—NH-phenyl wherein
the benzo[b]thienyl is optionally substituted with OH, CH3, OCH3, N(CH3)2, CH2═CHNHCH3, CH2NH2, CH2CH2NH2 or CH2NHCH2CH2N(CH3)2;
the indolyl is substituted with methyl, CN, C(O)H, CH2CH2CH2NH2, CH2NHCH2CH═CH2, C(O)CH3, C(O)OCH3, OCH3, C(O)N(CH(CH3)2)2, CH2OH, CH2C(O)NH2, C(O)NH2, CH2NHCH2CH2N(CH3)2 or piperidinylmethyl; and
the phenyl is optionally substituted with one or more substituents selected from the group consisting of Cl, F, CH3, CH2OH, CN, —C(O)NH2, OH, OCH3, N(CH3)2 and —NH—S(O)2—CH3; or
R7 is Y—Z wherein
Y is benzo[b]thienyl or thienyl; and
Z is selected from the optionally substituted group consisting of CH═CHNHCH3, NHCH3, CH2NH2, CH2CH2NH2, CH2NHCH3, CH2NHCH2CH2N(CH3)2, N(CH3)2, CH2NHCH2CH2-morpholinyl, benzo[b]thienyl, morpholinylmethyl and piperazinylmethyl; wherein
the piperazinyl is optionally substituted with methyl.
4. The compound of claim 3 wherein
R1 and R4 are H;
R3 is selected from the optionally substituted group consisting of H, OH, 2,3-dihydrobenzofuranyl, naphthyl, pyrazolyl and pyrrolyl; wherein
R3 is —C(O)—NY100—(C(Y100)2)x—Ra wherein
Y100 is H;
x is 0;
Ra is selected from the optionally substituted group consisting of naphthyl and phenyl; wherein
the naphthyl is optionally substituted with OH;
the phenyl is optionally substituted with OH; or
R3 is selected from the group consisting of —NH—C(O)-cyclobutyl and —NH—C(O)-phenyl;
R3 is A-B wherein
A is selected from the group consisting of phenyl and thienyl; and
B is selected from the group consisting of benzyloxy, phenyl and thienyl;
R5 is selected from the group consisting of
Figure US20120053345A1-20120301-C00950
 wherein
E is selected from the group consisting of H, CH2C(O)NH2, CH2CH(CH3)C(O)OCH3, CH2CH2CH2OH, CH2CH2C(O)OCH3, CH2CH2CH2OCH3, CH2CH2NH2, CH2CH2CH2NH2, CH2CH2C(O)OH, CH2CH2C(O)NH2, CH2CH2CH2CH2NH2, CH2CH2CH2OCH3, CH2CH2CH2C(O)OH, CH2CH2CH2C(O)NH2, CH2CH(CH3)C(O)OH, CH2CH2C(O)NH(CH3), CH2CH2C(O)N(CH3)2, N(CH3)2, isopropyl, morpholinylethyl and piperidinylethyl; and
G is H, NH2 or NHCH2CH2-pyridinyl or
R5 is —C(O)—NH—(CH2)a—Rc wherein
a is 0
Rc is J100-J200 wherein
J100 is phenyl optionally substituted with OCH3 and
J200 is imidazo[1,2-a]pyridinyl or 1,8a-dihydroimidazo[1,2-a]pyridinyl; or
R5 is —NH—C(O)—(CH2)n—Rd wherein
n is 2 and Rd is imidazolyl; or
R5 is Z100—Z200 wherein
Z100 is thienyl;
Z200 is thienyl;
R6 is H or
Figure US20120053345A1-20120301-C00951
and
R7 is selected from the optionally substituted group consisting of H, benzofuranyl, benzo[b]thienyl, indolyl, naphthyl, quinolinyl, CH═CH-phenyl, —C≡C-phenyl, —C(O)—NH—CH2-phenyl and —C(O)—NH-phenyl wherein
the benzo[b]thienyl is optionally substituted with OH, CH3, OCH3, N(CH3)2, CH2═CH2NHCH3, CH2NH2, CH2CH2NH2, CH2NHCH2CH2N(CH3)2, piperidinylmethyl or CH2NHCH2N(CH3)2; and
the indolyl is optionally substituted with methyl, CN, C(O)H, CH2CH2CH2NH2, CH2NHCH2CH═CH2, C(O)CH3, C(O)OCH3, or OCH3; methyl, CN, C(O)H, CH2CH2CH2NH2, CH2NHCH2CH═CH2, C(O)CH3, C(O)OCH3, or OCH3
the phenyl is optionally substituted with one or more substituents selected from the group consisting of OH and OCH3; or
R7 is Y—Z wherein
Y is benzo[b]thienyl or thienyl; and
Z is selected from the group consisting of CH═CHNHCH3, NHCH3, CH2NH2, CH2CH2NH2, CH2NHCH3, CH2NHCH2CH2N(CH3)2, N(CH3)2, CH2NHCH2CH2-morpholinyl, benzo[b]thienyl, morpholinylmethyl and piperazinylmethyl;
wherein the piperazinyl is optionally substituted with methyl.
5. The compound of claim 4 wherein
R1 and R4 are H;
R3 is selected from the group consisting of H, OH, 2,3-dihydrobenzofuranyl, pyrrolyl and optionally substituted napthyl; or
R3 is —C(O)—NY100—(C(Y100)2)x—Ra wherein
Y100 is H;
x is 0; and
Ra is phenyl substituted with OH;
R5 is
Figure US20120053345A1-20120301-C00952
 wherein
E is selected from the group consisting of H, CH2C(O)NH2, CH2CH2NH2, CH2CH2CH2NH2, CH2CH2CH2CH2NH2, CH2CH2CH2OCH3, CH2CH2CH2OH, CH2CH2C(O)OH, CH2CH2CH2C(O)OH, CH2CH2C(O)NH2, CH2CH2CH2C(O)NH2, CH2CH(CH3)C(O)OCH3, CH2CH(CH3)C(O)OH, CH2CH2C(O)OCH3, CH2CH2C(O)NH(CH3), CH2CH2C(O)N(CH3)2, N(CH3)2, isopropyl, morpholinylethyl and piperidinylethyl; and
G is H, NH2 or NHCH2CH2-pyridinyl or
R5 is —C(O)—NH—(CH2)a—Rc wherein
a is 0; and
Rc is J100-J200 wherein
J100 is phenyl and J200 is 1,8a-dihydroimidazo[1,2a]pyridinyl; or
R5 is —NH—C(O)—(CH2)n—Rd wherein
n is 2 and Rd is imidazolyl; or
R5 is Z100—Z200 wherein
Z100 is thienyl and Z200 is thienyl;
R6 is H or
Figure US20120053345A1-20120301-C00953
and
R7 is selected from the optionally substituted group consisting of H, —CH═CH-phenyl, —C≡C-phenyl, benzofuranyl, benzo[b]thienyl, indolyl, quinolinyl, naphthyl, —C(O)—NH—CH2-phenyl and —C(O)—NH-phenyl;
wherein the naphthyl is optionally substituted with OH, C(O)H or OCH3;
the benzo[b]thienyl optionally substituted with OH, CH3, OCH3, CH2═CH3—NHCH3, CH2NH2, CH2CH2NH2, CH2NHCH2CH2N(CH3)2, N(CH3)2 or piperidinylmethyl;
the indolyl is optionally substituted with methyl, CN, C(O)H, CH2CH2CH2NH2, CH2NHCH2CH═CH2, C(O)CH3, C(O)OCH3, or OCH3;
the phenyl is optionally substituted with OH or OCH3; or
R7 is Y—Z wherein
Y is benzo[b]thienyl; and
Z is selected from the group consisting of CH2NHCH2CH2-morpholinyl, morpholinylmethyl and piperazinylmethyl wherein the piperazinyl is optionally substituted with methyl.
6. The compound of claim 5 wherein
R1, R3, R4 and R6 are H;
R5 is
Figure US20120053345A1-20120301-C00954
 wherein
E is selected from the group consisting of H, —CH2CH2NH2, CH2CH2CH2NH2, CH2CH2CH2OH, CH2CH2C(O)OH and CH2CH2C(O)NH2; and
R7 is selected from the group consisting of benzo[b]thienyl, indolyl, —C(O)—NH—CH2-phenyl and —C(O)—NH-phenyl wherein
the benzo[b]theinyl is optionally substituted by piperidinylmethyl;
the indolyl is optionally substituted by CN, methyl or C(O)H.
7. The compound of claim 6 wherein
R1, R3, R4 and R6 are H;
R5 is
Figure US20120053345A1-20120301-C00955
 wherein
E is H; and
R7 is —C(O)—NH—CH2-phenyl or —C(O)—NH-phenyl.
8. The compound of claim 6 wherein
R1, R3, R4 and R6 are H;
R5 is
Figure US20120053345A1-20120301-C00956
 wherein
E is selected from the group consisting of H, —CH2CH2NH2, CH2CH2CH2NH2, CH2CH2CH2OH, CH2CH2C(O)OH and CH2CH2C(O)NH2; and
R7 is benzo[b]thienyl or indolyl wherein
the benzo[b]theinyl is optionally substituted by piperidinylmethyl;
the indolyl is optionally substituted by CN, methyl or C(O)H.
9. The compound of claim 8 wherein R1, R3, R4 and R6 are H;
R5 is
Figure US20120053345A1-20120301-C00957
 wherein
E is selected from the group consisting of —CH2CH2NH2, CH2CH2CH2NH2, CH2CH2CH2OH, CH2CH2C(O)OH and CH2CH2C(O)NH2; and
R7 is benzo[b]thienyl.
US13/198,363 2006-03-31 2011-08-04 Indazole Compounds Abandoned US20120053345A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US13/198,363 US20120053345A1 (en) 2006-03-31 2011-08-04 Indazole Compounds

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US78855306P 2006-03-31 2006-03-31
US11/731,950 US8008481B2 (en) 2006-03-31 2007-04-02 Indazole compounds
US13/198,363 US20120053345A1 (en) 2006-03-31 2011-08-04 Indazole Compounds

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US11/731,950 Continuation US8008481B2 (en) 2006-03-31 2007-04-02 Indazole compounds

Publications (1)

Publication Number Publication Date
US20120053345A1 true US20120053345A1 (en) 2012-03-01

Family

ID=38581581

Family Applications (2)

Application Number Title Priority Date Filing Date
US11/731,950 Active 2029-09-21 US8008481B2 (en) 2006-03-31 2007-04-02 Indazole compounds
US13/198,363 Abandoned US20120053345A1 (en) 2006-03-31 2011-08-04 Indazole Compounds

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US11/731,950 Active 2029-09-21 US8008481B2 (en) 2006-03-31 2007-04-02 Indazole compounds

Country Status (7)

Country Link
US (2) US8008481B2 (en)
EP (1) EP2001480A4 (en)
JP (1) JP5255559B2 (en)
CN (1) CN101437519A (en)
CA (1) CA2644910C (en)
MX (1) MX2008012482A (en)
WO (1) WO2007117465A2 (en)

Cited By (41)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9029536B2 (en) 2010-08-04 2015-05-12 Takeda Pharmaceutical Company Limited Fused heterocyclic compounds
WO2017009798A1 (en) * 2015-07-15 2017-01-19 Aurigene Discovery Technologies Limited Indazole and azaindazole compounds as irak-4 inhibitors
WO2017024021A1 (en) * 2015-08-03 2017-02-09 Samumed, Llc 3-(1h-pyrrolo[2,3-b]pyridin-2-yl)-1h-indazoles and therapeutic uses thereof
US9603836B2 (en) 2014-05-15 2017-03-28 Iteos Therapeutics Pyrrolidine-2, 5-dione derivatives, pharmaceutical compositions and methods for use as IDO1 inhibitors
US20170158642A1 (en) * 2014-07-01 2017-06-08 Gwangju Institute Of Science And Technology Composition for inducing cell reprogramming
US9873690B2 (en) 2015-03-17 2018-01-23 Pfizer Inc 3-indol substituted derivatives, pharmaceutical compositions and methods for use
US10131677B2 (en) 2014-09-08 2018-11-20 Samumed, Llc 3-(3H-imidazo[4,5-B]pyridin-2-yl)-1H-pyrazolo[3,4-B]pyridine and therapeutic uses thereof
US10160753B2 (en) 2014-01-10 2018-12-25 Aurigene Discovery Technologies Limited Indazole compounds as IRAK4 inhibitors
US10166218B2 (en) 2015-08-03 2019-01-01 Samumed, Llc 3-(1H-indol-2-yl)-1H-pyrazolo[3,4-C]pyridines and therapeutic uses thereof
US10183929B2 (en) 2013-01-08 2019-01-22 Samumed, Llc 3-(benzoimidazol-2-yl)-indazole inhibitors of the Wnt signaling pathway and therapeutic uses thereof
US10188634B2 (en) 2015-08-03 2019-01-29 Samumed, Llc 3-(3H-imidazo[4,5-C]pyridin-2-yl)-1 H-pyrazolo[4,3-B]pyridines and therapeutic uses thereof
US10195185B2 (en) 2015-08-03 2019-02-05 Samumed, Llc 3-(1H-imidazo[4,5-C]pyridin-2-yl)-1H-pyrazolo[4,3-B]pyridines and therapeutic uses thereof
US10202377B2 (en) 2014-09-08 2019-02-12 Samumed, Llc 3-(1H-benzo[D]imidazol-2-yl)-1H-pyrazolo[3,4-C]pyridine and therapeutic uses thereof
US10206929B2 (en) 2014-09-08 2019-02-19 Samumed, Llc 3-(1H-imidazo[4,5-c]pyridin-2-yl)-1H-pyrazolo[3,4-b]pyridine and therapeutic uses thereof
US10206909B2 (en) 2015-08-03 2019-02-19 Samumed, Llc 3-(1H-pyrrolo[2,3-B]pyridin-2-yl)-1H-pyrazolo[4,3-B]pyridines and therapeutic uses thereof
US10206908B2 (en) 2015-08-03 2019-02-19 Samumed, Llc 3-(1H-pyrrolo[3,2-C]pyridin-2-YL)-1H-pyrazolo[3,4-C]pyridines and therapeutic uses thereof
US10226448B2 (en) 2015-08-03 2019-03-12 Samumed, Llc 3-(1H-pyrrolo[3,2-C]pyridin-2-yl)-1H-pyrazolo[3,4-B]pyridines and therapeutic uses thereof
US10226453B2 (en) 2015-08-03 2019-03-12 Samumed, Llc 3-(1H-indol-2-yl)-1H-pyrazolo[4,3-B]pyridines and therapeutic uses thereof
US10231956B2 (en) 2015-08-03 2019-03-19 Samumed, Llc 3-(1H-pyrrolo[3,2-C]pyridin-2-YL)-1 H-pyrazolo[4,3-B]pyridines and therapeutic uses thereof
US10280166B2 (en) 2014-09-08 2019-05-07 Samumed, Llc 2-(1H-indazol-3-yl)-3H-imidazo[4,5-B]pyridine and therapeutic uses thereof
US10285983B2 (en) 2015-08-03 2019-05-14 Samumed, Llc 3-(1H-pyrrolo[2,3-B]pyridin-2-yl)-1H-pyrazolo[3,4-B] pyridines and therapeutic uses thereof
US10285982B2 (en) 2015-08-03 2019-05-14 Samumed, Llc 3-(1H-pyrrolo[2,3-B]pyridin-2-yl)-1H-pyrazolo[3,4-C]pyridines and therapeutic uses thereof
US20190142799A1 (en) * 2016-05-12 2019-05-16 The Regents Of The University Of Michigan Ash1l inhibitors and methods of treatment therewith
US10329309B2 (en) 2015-08-03 2019-06-25 Samumed, Llc 3-(3H-imidazo[4,5-B]pyridin-2-yl)-1H-pyrazolo[4,3-B]pyridines and therapeutic uses thereof
US10383861B2 (en) 2015-08-03 2019-08-20 Sammumed, LLC 3-(1H-pyrrolo[2,3-C]pyridin-2-yl)-1H-pyrazolo[3,4-C]pyridines and therapeutic uses thereof
US10392383B2 (en) 2015-08-03 2019-08-27 Samumed, Llc 3-(1H-benzo[d]imidazol-2-yl)-1H-pyrazolo[4,3-b]pyridines and therapeutic uses thereof
US10407425B2 (en) 2012-04-04 2019-09-10 Samumed, Llc Indazole inhibitors of the Wnt signal pathway and therapeutic uses thereof
US10463651B2 (en) 2015-08-03 2019-11-05 Samumed, Llc 3-(1H-pyrrolo[3,2-C]pyridin-2-YL)-1H-indazoles and therapeutic uses thereof
US10464924B2 (en) 2011-09-14 2019-11-05 Samumed, Llc Indazole-3-carboxamides and their use as Wnt/β-catenin signaling pathway inhibitors
US10519169B2 (en) 2015-08-03 2019-12-31 Samumed, Llc 3-(1H-pyrrolo[2,3-C]pyridin-2-yl)-1 H-pyrazolo[4,3-B]pyridines and therapeutic uses thereof
US10533020B2 (en) 2014-09-08 2020-01-14 Samumed, Llc 3-(3H-imidazo[4,5-B]pyridin-2-yl)-1 H-pyrazolo[3,4-C]pyridine and therapeutic uses thereof
US10544139B2 (en) 2015-11-06 2020-01-28 Samumed, Llc Treatment of osteoarthritis
US10544095B2 (en) 2015-08-10 2020-01-28 Pfizer Inc. 3-indol substituted derivatives, pharmaceutical compositions and methods for use
US10596154B2 (en) 2014-09-08 2020-03-24 Samumed, Llc 3-(1H-imidazo[4,5-C]pyridin-2-yl)-1H-pyrazolo[3,4-C]pyridine and therapeutic uses thereof
US10604512B2 (en) 2015-08-03 2020-03-31 Samumed, Llc 3-(1H-indol-2-yl)-1H-indazoles and therapeutic uses thereof
US10758518B2 (en) 2017-10-31 2020-09-01 Curis, Inc. Compounds and compositions for treating hematological disorders
US10758523B2 (en) 2016-11-07 2020-09-01 Samumed, Llc Single-dose, ready-to-use injectable formulations
US10806726B2 (en) 2016-10-21 2020-10-20 Samumed, Llc Methods of using indazole-3-carb oxamides and their use as Wnt/B-catenin signaling pathway inhibitors
US10995100B2 (en) 2014-01-13 2021-05-04 Aurigene Discovery Technologies Limited Bicyclic heterocyclyl derivatives as IRAK4 inhibitors
US11110177B2 (en) 2017-11-10 2021-09-07 The Regents Of The University Of Michigan ASH1L degraders and methods of treatment therewith
US11419875B2 (en) 2017-03-31 2022-08-23 Aurigene Discovery Technologies Limited Compounds and compositions for treating hematological disorders

Families Citing this family (74)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB0400895D0 (en) * 2004-01-15 2004-02-18 Smithkline Beecham Corp Chemical compounds
AR050253A1 (en) * 2004-06-24 2006-10-11 Smithkline Beecham Corp COMPOSITE DERIVED FROM INDAZOL CARBOXAMIDE, COMPOSITION THAT INCLUDES IT AND ITS USE FOR THE PREPARATION OF A MEDICINAL PRODUCT
US8063071B2 (en) 2007-10-31 2011-11-22 GlaxoSmithKline, LLC Chemical compounds
EP2125793B1 (en) 2006-12-21 2010-09-01 Eli Lilly & Company Imidazolidinonyl aminopyrimidine compounds for the treatment of cancer
ES2351371T3 (en) * 2006-12-21 2011-02-03 ELI LILLY &amp; COMPANY IMIDAZOLIDINONIL AMINOPIRIMIDINE COMPOUNDS FOR THE TREATMENT OF CANCER.
PE20081889A1 (en) 2007-03-23 2009-03-05 Smithkline Beecham Corp INDOL CARBOXAMIDES AS INHIBITORS OF IKK2
WO2008144222A2 (en) 2007-05-16 2008-11-27 Eli Lilly And Company Triazolyl aminopyrimidine compounds
BRPI0811433A2 (en) 2007-05-16 2016-03-22 Lilly Co Eli triazolyl aminopyrimidine-like compounds
CN101790526A (en) * 2007-06-08 2010-07-28 雅培制药有限公司 5-heteroaryl substituted indazoles as kinase inhibitors
EP2195328A4 (en) 2007-08-15 2011-06-15 Cytokinetics Inc Certain chemical entities, compositions, and methods
US7705004B2 (en) 2007-08-17 2010-04-27 Portola Pharmaceuticals, Inc. Protein kinase inhibitors
US9149463B2 (en) 2007-09-18 2015-10-06 The Board Of Trustees Of The Leland Standford Junior University Methods and compositions of treating a Flaviviridae family viral infection
US9061010B2 (en) 2007-09-18 2015-06-23 The Board Of Trustees Of The Leland Stanford Junior University Methods of treating a Flaviviridae family viral infection and compositions for treating a Flaviviridae family viral infection
US8940730B2 (en) 2007-09-18 2015-01-27 The Board Of Trustees Of The Leland Stanford Junior University Methods and compositions of treating a Flaviviridae family viral infection
WO2010107742A2 (en) * 2009-03-18 2010-09-23 The Board Of Trustees Of The Leland Stanford Junior University Methods and compositions of treating a flaviviridae family viral infection
US9101628B2 (en) 2007-09-18 2015-08-11 The Board Of Trustees Of The Leland Stanford Junior University Methods and composition of treating a flaviviridae family viral infection
RU2364597C1 (en) * 2007-12-14 2009-08-20 Андрей Александрович Иващенко HETEROCYCLIC INHIBITORS OF Hh-SYGNAL CASCADE, BASED ON THEM MEDICINAL COMPOSITIONS AND METHOD OF TREATING DISEASES INDUCED BY ABBARANT ACTIVITY OF Hh-SIGNAL SYSTEM
FR2938340A1 (en) * 2008-11-13 2010-05-14 Galderma Res & Dev MODULATORS OF CARNITINE OCTANOYLTRANSFERASE IN THE TREATMENT OF ACNE, SEBORRHEIC DERMATITIS OR HYPERSEBORRHEA
EP2346508B1 (en) 2008-09-26 2016-08-24 Intellikine, LLC Heterocyclic kinase inhibitors
JP2012520884A (en) 2009-03-18 2012-09-10 ザ ボード オブ トラスティーズ オブ ザ リランド スタンフォード ジュニア ユニバーシティー Methods and compositions for treating flaviviridae viral infections
SG10201401169QA (en) * 2009-04-02 2014-05-29 Merck Serono Sa Dihydroorotate dehydrogenase inhibitors
JP5756457B2 (en) 2009-04-06 2015-07-29 アジオス ファーマシューティカルズ, インコーポレイテッド Pyruvate kinase M2 modulators, therapeutic compositions and related methods of use
US8575162B2 (en) 2009-04-30 2013-11-05 Glaxosmithkline Intellectual Property Development Limited Compounds
AU2010259023A1 (en) * 2009-06-08 2012-01-12 Nantbioscience, Inc. Triazine derivatives and their therapeutical applications
SG10201403696UA (en) 2009-06-29 2014-10-30 Agios Pharmaceuticals Inc Therapeutic compounds and compositions
CA2770116C (en) 2009-08-10 2018-03-13 Epitherix, Llc Indazole inhibitors of the wnt signal pathway and therapeutic uses thereof
EP2464231A4 (en) * 2009-08-10 2013-02-06 Samumed Llc Indazoles as wnt/b-catenin signaling pathway inhibitors and therapeutic uses thereof
US8299070B2 (en) * 2009-11-25 2012-10-30 Japan Tobacco Inc. Indole compounds and pharmaceutical use thereof
PT3001903T (en) * 2009-12-21 2017-12-18 Samumed Llc 1h-pyrazolo[3,4-b]pyridines and therapeutic uses thereof
JP5745501B2 (en) * 2010-03-25 2015-07-08 国立大学法人 岡山大学 Benzanilide derivatives
JP2013526542A (en) 2010-05-12 2013-06-24 アッヴィ・インコーポレイテッド Indazole inhibitor of kinase
EP2593453B1 (en) 2010-07-15 2014-09-17 Bristol-Myers Squibb Company Azaindazole compounds
MX336022B (en) 2010-12-21 2016-01-06 Agios Pharmaceuticals Inc Bicyclic pkm2 activators.
US9199918B2 (en) 2011-02-15 2015-12-01 Georgetown University Small molecule inhibitors of AGBL2
CN102675286B (en) * 2011-03-07 2015-08-19 中国科学院上海药物研究所 One class indazole compounds and preparation method thereof, purposes and pharmaceutical composition
US9181231B2 (en) 2011-05-03 2015-11-10 Agios Pharmaceuticals, Inc Pyruvate kinase activators for use for increasing lifetime of the red blood cells and treating anemia
SI2704721T1 (en) 2011-05-03 2018-08-31 Agios Pharmaceuticals, Inc. Pyruvate kinase activators for use in therapy
KR20140063605A (en) 2011-07-19 2014-05-27 인피니티 파마슈티칼스, 인코포레이티드 Heterocyclic compounds and uses thereof
CA2860672A1 (en) 2012-01-06 2013-07-11 University Of South Florida Compositions, methods of use, and methods of treatment
US9745339B2 (en) 2012-02-08 2017-08-29 Novalix Deutschland Gmbh Ligands for antibody and Fc-fusion protein purification by affinity chromotography IV
US8889730B2 (en) 2012-04-10 2014-11-18 Pfizer Inc. Indole and indazole compounds that activate AMPK
PT2770994T (en) 2012-05-04 2019-11-04 Samumed Llc 1h-pyrazolo[3,4-b]pyridines and therapeutic uses thereof
JP2015531361A (en) 2012-09-26 2015-11-02 バイエル・ファルマ・アクティエンゲゼルシャフト Substituted indazole-pyrrolopyrimidines useful for the treatment of hyperproliferative diseases
EP2900671A1 (en) 2012-09-26 2015-08-05 Bayer Pharma Aktiengesellschaft Substituted indazol-pyrrolopyrimidines useful in the treatment of hyperproliferative diseases
DK2935302T3 (en) 2012-12-18 2018-08-13 Vertex Pharma MANNOS DERIVATIVES FOR TREATING BACTERIAL INFECTIONS
WO2014139144A1 (en) 2013-03-15 2014-09-18 Agios Pharmaceuticals, Inc. Therapeutic compounds and compositions
JP6064062B2 (en) 2013-03-15 2017-01-18 ファイザー・インク Indazole compounds that activate AMPK
WO2015004534A2 (en) * 2013-06-21 2015-01-15 Zenith Epigenetics Corp. Novel substituted bicyclic compounds as bromodomain inhibitors
EP3183239B1 (en) * 2014-08-22 2018-09-26 Merck Patent GmbH Indazoles
WO2016040188A1 (en) 2014-09-08 2016-03-17 Samumed, Llc 3-(3h-imidazo[4,5-c]pyridin-2-yl)-1h-pyrazolo[3,4-c]pyridine and therapeutic uses thereof
WO2016040182A1 (en) 2014-09-08 2016-03-17 Samumed, Llc 2-(1h-indazol-3-yl)-1h-imidazo[4,5-c]pyridine and therapeutic uses thereof
SG11201701960XA (en) 2014-10-13 2017-04-27 Yuhan Corp Compounds and compositions for modulating egfr mutant kinase activities
AU2016276951B2 (en) 2015-06-11 2020-10-08 Agios Pharmaceuticals, Inc. Methods of using pyruvate kinase activators
AU2016275361B2 (en) 2015-06-12 2020-11-19 Vertex Pharmaceuticals Incorporated Mannose derivatives for treating bacterial infections
SI3319955T1 (en) 2015-07-06 2021-01-29 Gilead Sciences, Inc. 6-amino-quinoline-3-carbonitrils as cot modulators
SI3191470T1 (en) 2015-07-06 2019-04-30 Gilead Sciences, Inc. Cot modulators and methods of use thereof
EP3333157B1 (en) * 2015-08-07 2020-12-23 Harbin Zhenbao Pharmaceutical Co., Ltd. Vinyl compounds as fgfr and vegfr inhibitors
CN107021963A (en) * 2016-01-29 2017-08-08 北京诺诚健华医药科技有限公司 Pyrazole fused ring analog derivative, its preparation method and its application in treating cancer, inflammation and immunity disease
WO2017175842A1 (en) * 2016-04-07 2017-10-12 国立大学法人京都大学 Compound and pharmaceutical composition pertaining to modification of splicing
JP7010851B2 (en) 2016-06-01 2022-02-10 バイオスプライス セラピューティクス インコーポレイテッド N- (5- (3- (7- (3-Fluorophenyl) -3H-imidazole [4,5-c] pyridin-2-yl) -1H-indazole-5-yl) pyridin-3-yl)- Method for preparing 3-methylbutaneamide
ES2800339T3 (en) 2016-06-30 2020-12-29 Gilead Sciences Inc 4,6-diaminoquinazolines as cradle modulators and methods of using them
WO2018043674A1 (en) 2016-08-31 2018-03-08 国立大学法人京都大学 Composition for activating neurogenesis
CN110139862B (en) * 2016-11-03 2024-01-16 百时美施贵宝公司 Substituted bicyclic heterocyclic derivatives useful as ROMK channel inhibitors
CN106674132B (en) * 2016-11-22 2019-04-26 上海书亚医药科技有限公司 A kind of preparation method and application of miazines herbicide
PL3580220T3 (en) * 2017-02-13 2022-02-07 Bristol-Myers Squibb Company Aminotriazolopyridines as kinase inhibitors
TW202043205A (en) 2018-12-31 2020-12-01 美商拜歐米富士恩有限公司 Inhibitors of menin-mll interaction
EP3906026A4 (en) 2018-12-31 2022-10-19 Biomea Fusion, LLC Irreversible inhibitors of menin-mll interaction
TW202235416A (en) 2019-06-14 2022-09-16 美商基利科學股份有限公司 Cot modulators and methods of use thereof
CN114072387B (en) * 2019-11-06 2024-03-08 暨南大学 Indazole compound, pharmaceutical composition and application thereof
AU2021249010A1 (en) 2020-03-30 2022-10-06 Gilead Sciences, Inc. Solid forms of (S)-6-(((1-(bicyclo[1.1.1]pentan-1-yl)-1H-1,2,3-triazol-4-yl)2-methyl-1-oxo-1,2- dihydroisoquinolin-5-yl)methyl)))amino)8-chloro-(neopentylamino)quinoline-3-carb onitrile a cot inhibitor compound
CN115397824A (en) 2020-04-02 2022-11-25 吉利德科学公司 Process for preparing COT inhibitor compounds
CN112457235B (en) * 2020-12-02 2023-04-25 烟台凯博医药科技有限公司 Preparation method of 7-methylindole
CN114605329B (en) * 2022-03-28 2024-01-26 河南中医药大学 Substituted indazole carboxamides or substituted azaindazole carboxamides FLT3 inhibitors and uses thereof
WO2023217232A1 (en) * 2022-05-13 2023-11-16 上海湃隆生物科技有限公司 Kinesin kif18a inhibitor and use thereof

Family Cites Families (52)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US544038A (en) * 1895-08-06 Draft-gate for fire-doors
US4999356A (en) * 1987-08-07 1991-03-12 Hoechst-Roussel Pharmaceuticals, Inc. 1-phenyl-3-(1-piperazinyl)-1H-indazoles
US5444038A (en) * 1992-03-09 1995-08-22 Zeneca Limited Arylindazoles and their use as herbicides
CA2223081C (en) * 1995-06-07 2001-03-06 Pfizer Inc. Heterocyclic ring-fused pyrimidine derivatives
EP1028964A1 (en) 1997-11-11 2000-08-23 Pfizer Products Inc. Thienopyrimidine and thienopyridine derivatives useful as anticancer agents
EE200100102A (en) 1998-08-20 2002-06-17 Agouron Pharmaceuticals, Inc. Non-peptide GnRH Agents, Methods of Preparation, and Intermediates
GB9918035D0 (en) * 1999-07-30 1999-09-29 Novartis Ag Organic compounds
MXPA02003436A (en) * 1999-10-07 2002-08-20 Amgen Inc Triazine kinase inhibitors.
US6897231B2 (en) 2000-07-31 2005-05-24 Signal Pharmaceuticals, Inc. Indazole derivatives as JNK inhibitors and compositions and methods related thereto
US20040007787A1 (en) * 2000-12-05 2004-01-15 The Procter & Gamble Company Articles, systems, and methods for dispensing volatile materials
PE20020776A1 (en) * 2000-12-20 2002-08-22 Sugen Inc INDOLINONES 4-ARIL SUBSTITUTED
KR100947185B1 (en) * 2000-12-21 2010-03-15 버텍스 파마슈티칼스 인코포레이티드 Pyrazole compounds useful as protein kinase inhibitors and compositions comprising said compounds
PT1370553E (en) * 2001-03-23 2006-09-29 Bayer Corp RHOQUINASE INHIBITORS
EP1463505A2 (en) * 2001-12-13 2004-10-06 Abbott Laboratories 3-(phenyl-alkoxy)-5-(phenyl)-pyridine derivatives and related compounds as kinase inhibitors for the treatment of cancer
US7201963B2 (en) * 2002-01-15 2007-04-10 Gentex Corporation Pre-processed workpiece having a surface deposition of absorber dye rendering the workpiece weld-enabled
KR20050004214A (en) 2002-05-31 2005-01-12 에자이 가부시키가이샤 Pyrazole compound and medicinal composition containing the same
CA2488699A1 (en) 2002-06-12 2003-12-24 Sumitomo Pharmaceuticals Co., Ltd. Indole, indazole, and benzazole derivative
GB0218625D0 (en) * 2002-08-10 2002-09-18 Astex Technology Ltd Pharmaceutical compounds
EP1545596A4 (en) 2002-09-06 2005-12-28 Janssen Pharmaceutica Nv Use of histamine h4 receptor modulators for the treatment of allergy and asthma
FR2845382A1 (en) * 2002-10-02 2004-04-09 Sanofi Synthelabo INDAZOLECARBOXAMIDE DERIVATIVES, THEIR PREPARATION AND THEIR USE IN THERAPEUTICS
US7196082B2 (en) * 2002-11-08 2007-03-27 Merck & Co. Inc. Ophthalmic compositions for treating ocular hypertension
US7488823B2 (en) * 2003-11-10 2009-02-10 Array Biopharma, Inc. Cyanoguanidines and cyanoamidines as ErbB2 and EGFR inhibitors
KR20050101551A (en) * 2003-02-14 2005-10-24 와이어쓰 Heterocyclyl-3-sulfonylindazoles as 5-hydroxytryptamine-6 ligands
AU2004215481B2 (en) * 2003-02-28 2010-11-11 Teijin Pharma Limited Pyrazolo(1,5-A)pyrimidine derivatives
US7191964B2 (en) * 2003-04-02 2007-03-20 Elkhart Brass Manufacturing Company, Inc. Fire-fighting monitor with remote control
ES2337150T3 (en) 2003-07-21 2010-04-21 Merck Serono Sa ALQUINILARIL CARBOXAMIDAS.
JP2007500700A (en) 2003-07-29 2007-01-18 スミスクライン ビーチャム コーポレーション Compound
US20050090529A1 (en) * 2003-07-31 2005-04-28 Pfizer Inc 3,5 Disubstituted indazole compounds with nitrogen-bearing 5-membered heterocycles, pharmaceutical compositions, and methods for mediating or inhibiting cell proliferation
AR045445A1 (en) 2003-08-05 2005-10-26 Vertex Pharma IONINIBID COMPOUNDS OF IONIC CHANNELS REGULATED BY VOLTAGE
WO2005014554A1 (en) 2003-08-08 2005-02-17 Astex Therapeutics Limited 1h-indazole-3-carboxamide compounds as mapkap kinase modulators
WO2005016862A1 (en) 2003-08-14 2005-02-24 Asahi Kasei Pharma Corporation Substituted arylalkanoic acid derivative and use thereof
RU2376292C2 (en) 2003-08-14 2009-12-20 Ф.Хоффманн-Ля Рош Аг Gaba-ergic modulators
US7378409B2 (en) * 2003-08-21 2008-05-27 Bristol-Myers Squibb Company Substituted cycloalkylamine derivatives as modulators of chemokine receptor activity
US8362264B2 (en) * 2003-08-22 2013-01-29 Dendreon Corporation Compositions and methods for the treatment of disease associated with Trp-p8 expression
MXPA06002296A (en) 2003-08-29 2006-05-22 Pfizer Thienopyridine-phenylacet amides and their derivatives useful as new anti-angiogenic agents.
US6984652B2 (en) * 2003-09-05 2006-01-10 Warner-Lambert Company Llc Gyrase inhibitors
BRPI0414313A (en) 2003-09-11 2006-11-07 Kemia Inc cytokine inhibitors
US8084457B2 (en) 2003-09-15 2011-12-27 Lead Discovery Center Gmbh Pharmaceutically active 4,6-disubstituted aminopyrimidine derivatives as modulators of protein kinases
JP2007505858A (en) 2003-09-18 2007-03-15 ノバルティス アクチエンゲゼルシャフト 2,4-Di (phenylamino) pyrimidine useful for the treatment of proliferative disorders
US7399765B2 (en) * 2003-09-19 2008-07-15 Abbott Laboratories Substituted diazabicycloalkane derivatives
GB0322016D0 (en) 2003-09-19 2003-10-22 Merck Sharp & Dohme New compounds
US20050119324A1 (en) * 2003-09-24 2005-06-02 Wyeth Methods of treating atherosclerosis using NF-kB inhibitors
WO2005030206A1 (en) 2003-09-24 2005-04-07 Imclone Systems Incorporated Aryl-1,3-azole derivatives and methods for inhibiting heparnase activity
US20050119276A1 (en) * 2003-09-24 2005-06-02 Wyeth Methods of treating inflammatory bowel disease using NF-kB inhibitors
CA2541949A1 (en) 2003-10-07 2005-05-26 Renovis, Inc. Amide derivatives as ion-channel ligands and pharmaceutical compositions and methods of using the same
RU2006115602A (en) 2003-10-08 2007-11-20 Вертекс Фармасьютикалз Инкорпорейтед (Us) ATF-BINDING CASSETTE CARRIER MODULATORS
TWI344961B (en) * 2003-10-15 2011-07-11 Ube Industries Novel indazole derivative
JP4537407B2 (en) 2003-10-30 2010-09-01 ベーリンガー インゲルハイム (カナダ) リミテッド RSV polymerase inhibitor
AU2004293035A1 (en) * 2003-11-19 2005-06-09 Signal Pharmaceuticals, Llc Methods of treating diseases and disorders by targeting multiple kinases
GB0409080D0 (en) * 2004-04-23 2004-05-26 Biofocus Discovery Ltd Compounds which interact with protein kinases
MXPA06013250A (en) * 2004-05-14 2007-02-28 Abbott Lab Kinase inhibitors as therapeutic agents.
DE102004028862A1 (en) * 2004-06-15 2005-12-29 Merck Patent Gmbh 3-aminoindazoles

Cited By (64)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9029536B2 (en) 2010-08-04 2015-05-12 Takeda Pharmaceutical Company Limited Fused heterocyclic compounds
US11066388B2 (en) 2011-09-14 2021-07-20 Biosplice Therapeutics, Inc. Indazole-3-carboxamides and their use as WNT/B-catenin signaling pathway inhibitors
US11780823B2 (en) 2011-09-14 2023-10-10 Biosplice Therapeutics, Inc. Indazole-3-carboxamides and their use as Wnt/β-catenin signaling pathway inhibitors
US10464924B2 (en) 2011-09-14 2019-11-05 Samumed, Llc Indazole-3-carboxamides and their use as Wnt/β-catenin signaling pathway inhibitors
US10407425B2 (en) 2012-04-04 2019-09-10 Samumed, Llc Indazole inhibitors of the Wnt signal pathway and therapeutic uses thereof
US11697649B2 (en) 2012-04-04 2023-07-11 Biosplice Therapeutics, Inc. Indazole inhibitors of the Wnt signal pathway and therapeutic uses thereof
US10947228B2 (en) 2012-04-04 2021-03-16 Samumed, Llc Indazole inhibitors of the Wnt signal pathway and therapeutic uses thereof
US10654832B2 (en) 2013-01-08 2020-05-19 Samumed, Llc 3-(benzoimidazol-2-YL)-indazole inhibitors of the Wnt signaling pathway and therapeutic uses thereof
US10183929B2 (en) 2013-01-08 2019-01-22 Samumed, Llc 3-(benzoimidazol-2-yl)-indazole inhibitors of the Wnt signaling pathway and therapeutic uses thereof
US10160753B2 (en) 2014-01-10 2018-12-25 Aurigene Discovery Technologies Limited Indazole compounds as IRAK4 inhibitors
US10995100B2 (en) 2014-01-13 2021-05-04 Aurigene Discovery Technologies Limited Bicyclic heterocyclyl derivatives as IRAK4 inhibitors
US11691987B2 (en) 2014-01-13 2023-07-04 Aurigene Discovery Technologies Limited Bicyclic heterocyclyl derivatives as IRAK4 inhibitors
US11981685B2 (en) 2014-01-13 2024-05-14 Aurigene Oncology Limited Bicyclic heterocyclyl derivatives as IRAK4 inhibitors
US10398679B2 (en) 2014-05-15 2019-09-03 Iteos Therapeutics Treatment method utilizing pyrrolidine-2, 5-dione derivatives as IDO1 inhibitors
US9949951B2 (en) 2014-05-15 2018-04-24 Iteos Therapeutics Pyrrolidine-2, 5-dione derivatives, pharmaceutical compositions and methods for use as IDO1 inhibitors
US9603836B2 (en) 2014-05-15 2017-03-28 Iteos Therapeutics Pyrrolidine-2, 5-dione derivatives, pharmaceutical compositions and methods for use as IDO1 inhibitors
US10858321B2 (en) * 2014-07-01 2020-12-08 Gwangju Institute Of Science And Technology Composition for inducing cell reprogramming
US20170158642A1 (en) * 2014-07-01 2017-06-08 Gwangju Institute Of Science And Technology Composition for inducing cell reprogramming
US10526347B2 (en) 2014-09-08 2020-01-07 Samumed, Llc 3-(3H-imidazo[4,5-B]pyridin-2-yl)-1H-pyrazolo[3,4-B]pyridine and therapeutic uses thereof
US10533020B2 (en) 2014-09-08 2020-01-14 Samumed, Llc 3-(3H-imidazo[4,5-B]pyridin-2-yl)-1 H-pyrazolo[3,4-C]pyridine and therapeutic uses thereof
US10280166B2 (en) 2014-09-08 2019-05-07 Samumed, Llc 2-(1H-indazol-3-yl)-3H-imidazo[4,5-B]pyridine and therapeutic uses thereof
US10131677B2 (en) 2014-09-08 2018-11-20 Samumed, Llc 3-(3H-imidazo[4,5-B]pyridin-2-yl)-1H-pyrazolo[3,4-B]pyridine and therapeutic uses thereof
US10202377B2 (en) 2014-09-08 2019-02-12 Samumed, Llc 3-(1H-benzo[D]imidazol-2-yl)-1H-pyrazolo[3,4-C]pyridine and therapeutic uses thereof
US10206929B2 (en) 2014-09-08 2019-02-19 Samumed, Llc 3-(1H-imidazo[4,5-c]pyridin-2-yl)-1H-pyrazolo[3,4-b]pyridine and therapeutic uses thereof
US10596154B2 (en) 2014-09-08 2020-03-24 Samumed, Llc 3-(1H-imidazo[4,5-C]pyridin-2-yl)-1H-pyrazolo[3,4-C]pyridine and therapeutic uses thereof
US9873690B2 (en) 2015-03-17 2018-01-23 Pfizer Inc 3-indol substituted derivatives, pharmaceutical compositions and methods for use
WO2017009798A1 (en) * 2015-07-15 2017-01-19 Aurigene Discovery Technologies Limited Indazole and azaindazole compounds as irak-4 inhibitors
US10226453B2 (en) 2015-08-03 2019-03-12 Samumed, Llc 3-(1H-indol-2-yl)-1H-pyrazolo[4,3-B]pyridines and therapeutic uses thereof
US10604512B2 (en) 2015-08-03 2020-03-31 Samumed, Llc 3-(1H-indol-2-yl)-1H-indazoles and therapeutic uses thereof
US10226448B2 (en) 2015-08-03 2019-03-12 Samumed, Llc 3-(1H-pyrrolo[3,2-C]pyridin-2-yl)-1H-pyrazolo[3,4-B]pyridines and therapeutic uses thereof
US10463651B2 (en) 2015-08-03 2019-11-05 Samumed, Llc 3-(1H-pyrrolo[3,2-C]pyridin-2-YL)-1H-indazoles and therapeutic uses thereof
US10206908B2 (en) 2015-08-03 2019-02-19 Samumed, Llc 3-(1H-pyrrolo[3,2-C]pyridin-2-YL)-1H-pyrazolo[3,4-C]pyridines and therapeutic uses thereof
US10519169B2 (en) 2015-08-03 2019-12-31 Samumed, Llc 3-(1H-pyrrolo[2,3-C]pyridin-2-yl)-1 H-pyrazolo[4,3-B]pyridines and therapeutic uses thereof
US10206909B2 (en) 2015-08-03 2019-02-19 Samumed, Llc 3-(1H-pyrrolo[2,3-B]pyridin-2-yl)-1H-pyrazolo[4,3-B]pyridines and therapeutic uses thereof
US10392383B2 (en) 2015-08-03 2019-08-27 Samumed, Llc 3-(1H-benzo[d]imidazol-2-yl)-1H-pyrazolo[4,3-b]pyridines and therapeutic uses thereof
WO2017024021A1 (en) * 2015-08-03 2017-02-09 Samumed, Llc 3-(1h-pyrrolo[2,3-b]pyridin-2-yl)-1h-indazoles and therapeutic uses thereof
US10285983B2 (en) 2015-08-03 2019-05-14 Samumed, Llc 3-(1H-pyrrolo[2,3-B]pyridin-2-yl)-1H-pyrazolo[3,4-B] pyridines and therapeutic uses thereof
US10383861B2 (en) 2015-08-03 2019-08-20 Sammumed, LLC 3-(1H-pyrrolo[2,3-C]pyridin-2-yl)-1H-pyrazolo[3,4-C]pyridines and therapeutic uses thereof
US10285982B2 (en) 2015-08-03 2019-05-14 Samumed, Llc 3-(1H-pyrrolo[2,3-B]pyridin-2-yl)-1H-pyrazolo[3,4-C]pyridines and therapeutic uses thereof
US10350199B2 (en) 2015-08-03 2019-07-16 Samumed, Llc 3-(1h-pyrrolo[2,3-b]pyridin-2-yl)-1h-indazoles and therapeutic uses thereof
US10329309B2 (en) 2015-08-03 2019-06-25 Samumed, Llc 3-(3H-imidazo[4,5-B]pyridin-2-yl)-1H-pyrazolo[4,3-B]pyridines and therapeutic uses thereof
US10231956B2 (en) 2015-08-03 2019-03-19 Samumed, Llc 3-(1H-pyrrolo[3,2-C]pyridin-2-YL)-1 H-pyrazolo[4,3-B]pyridines and therapeutic uses thereof
US10166218B2 (en) 2015-08-03 2019-01-01 Samumed, Llc 3-(1H-indol-2-yl)-1H-pyrazolo[3,4-C]pyridines and therapeutic uses thereof
US10188634B2 (en) 2015-08-03 2019-01-29 Samumed, Llc 3-(3H-imidazo[4,5-C]pyridin-2-yl)-1 H-pyrazolo[4,3-B]pyridines and therapeutic uses thereof
US10195185B2 (en) 2015-08-03 2019-02-05 Samumed, Llc 3-(1H-imidazo[4,5-C]pyridin-2-yl)-1H-pyrazolo[4,3-B]pyridines and therapeutic uses thereof
US10544095B2 (en) 2015-08-10 2020-01-28 Pfizer Inc. 3-indol substituted derivatives, pharmaceutical compositions and methods for use
US10899757B2 (en) 2015-11-06 2021-01-26 Samumed, Llc 2-(1H-indazol-3-yl)-3H-imidazo[4,5-C]pyridines and their anti-inflammatory uses thereof
US10882860B2 (en) 2015-11-06 2021-01-05 Samumed, Llc Treatment of osteoarthritis
US11560378B2 (en) 2015-11-06 2023-01-24 Biosplice Therapeutics, Inc. Treatment of osteoarthritis
US11667632B2 (en) 2015-11-06 2023-06-06 Biosplice Therapeutics, Inc. 2-(1H-indazol-3-yl)-3H-imidazo[4,5-C]pyridines and their anti-inflammatory uses thereof
US10544139B2 (en) 2015-11-06 2020-01-28 Samumed, Llc Treatment of osteoarthritis
US11883381B2 (en) * 2016-05-12 2024-01-30 The Regents Of The University Of Michigan ASH1L inhibitors and methods of treatment therewith
AU2017263574B2 (en) * 2016-05-12 2022-11-17 The Regents Of The University Of Michigan ASH1L inhibitors and methods of treatment therewith
US20190142799A1 (en) * 2016-05-12 2019-05-16 The Regents Of The University Of Michigan Ash1l inhibitors and methods of treatment therewith
US11684615B2 (en) 2016-10-21 2023-06-27 Biosplice Therapeutics, Inc. Methods of using indazole-3-carboxamides and their use as Wnt/β-catenin signaling pathway inhibitors
US10806726B2 (en) 2016-10-21 2020-10-20 Samumed, Llc Methods of using indazole-3-carb oxamides and their use as Wnt/B-catenin signaling pathway inhibitors
US10758523B2 (en) 2016-11-07 2020-09-01 Samumed, Llc Single-dose, ready-to-use injectable formulations
US11446288B2 (en) 2016-11-07 2022-09-20 Biosplice Therapeutics, Inc. Single-dose, ready-to-use injectable formulations
US11819499B2 (en) 2016-11-07 2023-11-21 Biosplice Therapeutics, Inc. Single-dose, ready-to-use injectable formulations
US11419875B2 (en) 2017-03-31 2022-08-23 Aurigene Discovery Technologies Limited Compounds and compositions for treating hematological disorders
US10758518B2 (en) 2017-10-31 2020-09-01 Curis, Inc. Compounds and compositions for treating hematological disorders
US11147885B2 (en) 2017-11-10 2021-10-19 The Regents Of The University Of Michigan ASH1L inhibitors and methods of treatment therewith
US11786602B2 (en) 2017-11-10 2023-10-17 The Regents Of The University Of Michigan ASH1L degraders and methods of treatment therewith
US11110177B2 (en) 2017-11-10 2021-09-07 The Regents Of The University Of Michigan ASH1L degraders and methods of treatment therewith

Also Published As

Publication number Publication date
WO2007117465A3 (en) 2008-08-28
WO2007117465A2 (en) 2007-10-18
EP2001480A4 (en) 2011-06-15
CA2644910A1 (en) 2007-10-18
CA2644910C (en) 2014-01-28
JP5255559B2 (en) 2013-08-07
US8008481B2 (en) 2011-08-30
EP2001480A2 (en) 2008-12-17
JP2009532370A (en) 2009-09-10
MX2008012482A (en) 2008-10-10
CN101437519A (en) 2009-05-20
US20070282101A1 (en) 2007-12-06

Similar Documents

Publication Publication Date Title
US8008481B2 (en) Indazole compounds
US20060074102A1 (en) Kinase inhibitors as therapeutic agents
US9567339B2 (en) Primary carboxamides as BTK inhibitors
US7709468B2 (en) Imidazo based heterocycles
US8962596B2 (en) 5,7-substituted-imidazo[1,2-C]pyrimidines as inhibitors of JAK kinases
US8637529B2 (en) Pyrazolo[3,4-d]pyrimidine compounds
US7790741B2 (en) Imidazothiazoles and imidazoxazoles
KR101661383B1 (en) 8-methyl-1-phenyl-imidazol[1,5-a]pyrazine compounds
US10160748B2 (en) Indazolones as modulators of tnf signaling
US20080076924A1 (en) Piperazines as P2X7 antagonists
AU2015246482A1 (en) Heterocyclic kinase inhibitors
US9150592B2 (en) Heterocyclic nuclear hormone receptor modulators
EA037563B1 (en) New (hetero)aryl-substituted piperidinyl derivatives, process for their preparation and pharmaceutical compositions containing them
US20140315883A1 (en) 6H-IMIDAZO[1,5-a]PYRROLO[2,3-e]PYRAZINE COMPOUNDS

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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