WO2012152763A1 - Substituted indazole derivatives active as kinase inhibitors - Google Patents

Substituted indazole derivatives active as kinase inhibitors Download PDF

Info

Publication number
WO2012152763A1
WO2012152763A1 PCT/EP2012/058389 EP2012058389W WO2012152763A1 WO 2012152763 A1 WO2012152763 A1 WO 2012152763A1 EP 2012058389 W EP2012058389 W EP 2012058389W WO 2012152763 A1 WO2012152763 A1 WO 2012152763A1
Authority
WO
WIPO (PCT)
Prior art keywords
formula
compound
indazol
methyl
branched
Prior art date
Application number
PCT/EP2012/058389
Other languages
French (fr)
Inventor
Andrea Lombardi Borgia
Marina Ciomei
Daniele Donati
Marcella Nesi
Original Assignee
Nerviano Medical Sciences S.R.L.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=46025764&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2012152763(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to US14/116,512 priority Critical patent/US9408850B2/en
Priority to KR1020137033115A priority patent/KR101953272B1/en
Priority to EA201391682A priority patent/EA023579B1/en
Priority to AU2012252468A priority patent/AU2012252468B2/en
Priority to ES12718275.6T priority patent/ES2611779T3/en
Priority to CA2835478A priority patent/CA2835478C/en
Priority to CN201280023142.9A priority patent/CN103534239B/en
Priority to BR112013028733-0A priority patent/BR112013028733B1/en
Priority to DK12718275.6T priority patent/DK2707359T3/en
Application filed by Nerviano Medical Sciences S.R.L. filed Critical Nerviano Medical Sciences S.R.L.
Priority to SG2013083696A priority patent/SG194911A1/en
Priority to SI201230838A priority patent/SI2707359T1/en
Priority to EP12718275.6A priority patent/EP2707359B1/en
Priority to MX2013012981A priority patent/MX342509B/en
Priority to JP2014509702A priority patent/JP6396210B2/en
Publication of WO2012152763A1 publication Critical patent/WO2012152763A1/en
Priority to IL229252A priority patent/IL229252A/en
Priority to ZA2013/09345A priority patent/ZA201309345B/en
Priority to HK14107159.6A priority patent/HK1193812A1/en
Priority to US15/203,087 priority patent/US9597317B2/en
Priority to US15/203,092 priority patent/US10028934B2/en
Priority to US16/013,019 priority patent/US10478423B2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4151,2-Diazoles
    • A61K31/4161,2-Diazoles condensed with carbocyclic ring systems, e.g. indazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/454Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. pimozide, domperidone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D231/00Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings
    • C07D231/54Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings condensed with carbocyclic rings or ring systems
    • C07D231/56Benzopyrazoles; Hydrogenated benzopyrazoles
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D295/00Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms
    • C07D295/04Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms
    • C07D295/14Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D295/155Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals with the ring nitrogen atoms and the carbon atoms with three bonds to hetero atoms separated by carbocyclic rings or by carbon chains interrupted by carbocyclic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D295/00Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms
    • C07D295/16Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms acylated on ring nitrogen atoms
    • C07D295/18Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms acylated on ring nitrogen atoms by radicals derived from carboxylic acids, or sulfur or nitrogen analogues thereof
    • C07D295/182Radicals derived from carboxylic acids
    • C07D295/185Radicals derived from carboxylic acids from aliphatic carboxylic acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • 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
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/12Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/12Transferases (2.) transferring phosphorus containing groups, e.g. kinases (2.7)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y207/00Transferases transferring phosphorus-containing groups (2.7)
    • C12Y207/10Protein-tyrosine kinases (2.7.10)
    • C12Y207/10001Receptor protein-tyrosine kinase (2.7.10.1)

Definitions

  • the present invention relates to certain substituted indazole compounds, which modulate the activity of protein kinases.
  • the compounds of this invention are therefore useful in treating diseases caused by deregulated protein kinase activity.
  • the present invention also provides methods for preparing these compounds, pharmaceutical compositions comprising these compounds, and methods of treating diseases utilizing pharmaceutical compositions comprising these compounds.
  • PKs protein kinases
  • a large share of the oncogenes and proto-oncogenes involved in human cancers encode for PKs.
  • the enhanced activities of PKs are also implicated in many non-malignant diseases, such as benign prostate hyperplasia, familial adenomatosis, polyposis, neurofibromatosis, psoriasis, vascular smooth cell proliferation associated with atherosclerosis, pulmonary fibrosis, arthritis glomerulonephritis and post-surgical stenosis and restenosis.
  • PKs are also implicated in inflammatory conditions and in the multiplication of viruses and parasites. PKs may also play a major role in the pathogenesis and development of neurodegenerative disorders.
  • PK is a group of membrane receptors with intrinsic protein-tyrosine kinase activity (RPTK). Upon binding of growth factors, RPTKs become activated and phosphorylate themselves and a series of substrates in the cytoplasm. Through this mechanism, they can transduce intracellular signallings for proliferation, differentiation or other biological changes. Structural abnormalities, over-expression and activation of RTPKs are frequently observed in human tumors, suggesting that constitutive ignition of the signal transduction leading to cell proliferation can result in malignant transformation.
  • RPTK protein-tyrosine kinase activity
  • FMS-like tyrosine kinase 3 (FLT3) and KIT are both members of the PDGFR family class III receptor tyrosine kinases characterized by an extracellular domain with 5 immunoglobulin-like loops, a transmembrane region and a cytoplasmic domain containing not only the kinase domain (divided in two regions) but also an autoinhibitory juxtamembrane (JM) domain that docks with the kinase domain to stabilize a catalytically inactive conformation.
  • JM autoinhibitory juxtamembrane
  • FLT3 has a crucial role in normal haematopoiesis and its expression is restricted to CD34+ hematopoietic stem/progenitor cells, brain, placenta, and gonads. Activation of FLT3 by FLT3-ligand promotes the normal growth of early progenitor cells.
  • FLT3 mutations of the FLT3 gene have been found to be one of the most common acquired genetic lesions. FLT3 mutations can be detected in 30% of acute myeloid leukemia (AML) patients (Nakao M, et al. Leukemia. 1996 Dec; 10(12): 1911-8), and also in 5-10% of patients with myelodisplastic syndrome (Horiike S, et al. Leukemia. 1997 Sep; 11(9): 1442-6).
  • AML acute myeloid leukemia
  • ITDs internal tandem duplications
  • TKD point mutations in the activation loop of the tyrosine kinase domain
  • ITD mutations are any elongation or shortening of the JM domain of FLT3 due to additions or deletions of amino acids that result in the constitutive activation of FLT3.
  • the presence of FLT3/ITD mutations is associated with a poor clinical outcome in both pediatric and adult patients with AML.
  • Point mutations in the activation loop of the kinase domain involve the aspartic acid, D835 residue, which leads to an activated configuration and transformation of myeloid cells.
  • D835 mutations are missense mutations that result in substitution of tyrosine, histidine, valine, glutamic acid or asparagine for aspartatic acid at amino acid 835 of FLT3. These mutations have been reported in 7% of patients with AML.
  • TKD mutations unlike ITD mutations, have not been shown to have any prognostic significance in AML patients. Both types of FLT3 mutation cause ligand-independent activation of the receptor and activation of downstream signalling pathways. Mutant FLT3 provides survival advantage to leukemic cells because it causes activation of three major intracellular signalling pathways: PI3K/AKT; RAS/RAF/MAPK and JAK/STAT (Masson K, Ronnstrand L. Cell Signal. 2009 Dec; 21(12): 1717-26).
  • interfering with the FLT3 signalling likely represents a specific and effective way to block tumor cell proliferation in AML and possibly other indications.
  • KIT is normally activated by stem cell factor. Signalling by KIT plays an important role in erythropoiesis, lymphopoiesis, mast cell development and function, megakaryopoiesis, gametogenesis and melanogenesis. Hematopoietic stem cells, multipotent progenitors and common myeloid progenitors, but also early T lineage progenitors and thymocytes express high levels of KIT. In addition, mast cells, melanocytes in the skin, and interstitial cells of Cajal in the digestive tract express KIT (Pittoni P. et al. Oncogene 2011 Feb 17; 30(7): 757-69). KIT overexpression or mutations can lead to cancer.
  • GIST gastrointestinal stromal tumors
  • KIT KIT-induced melanoma
  • melanoma Curtin JA, JCO, 2006, 24 (26): 4340-4346
  • acute myeloid leukemia Malaise M, Steinbach D, Corbacioglu S, Curr Hematol Malig Rep. 2009, 4(2): 77-82
  • primary adenoid cystic carcinoma of the salivary gland Vila L, Liu H, Al-Quran SZ, Coco DP, Dong HJ, Liu C, Mod Pathol. 2009; 22(10): 1296-302).
  • Overexpression is reported also in thymic carcinoma (Strobel P, Hohenberger P, Marx A, J Thorac Oncol.
  • KIT kinase activation appears to be the triggering factor for an important group of malignancies, both hematological and solid cancer diseases, thereby suggesting that it could represent a good therapeutic target for the treatment of these pathologies.
  • a first object of the present invention is to provide a substituted indazole compound represented by formula (I),
  • R1 is A, NR6R7, OR8, SO n R9, COR10, nitro, cyano or an optionally substituted group selected from C3-C6 cycloalkyl, heterocyclyl and heteroaryl;
  • R2, R3, R4 and R5 are independently hydrogen, halogen, nitro, cyano, SO n R9, COR10, NR11 R12, OR13 or an optionally substituted group selected from straight or branched C1-C6 alkyl, straight or branched C2-C6 alkenyl, straight or branched C2-C6 alkynyl, C3-C6 cycloalkyl and heterocyclyl wherein:
  • A is a straight or branched C1-C6 alkyl substituted with a group selected from an optionally substituted heterocyclyl, an optionally substituted heteroaryl, SO n R9, COR10, NR11 R12 and OR13;
  • R6 is hydrogen or an optionally substituted group selected from straight or branched ⁇ - ⁇ alkyl, straight or branched C2-C6 alkenyl, straight or branched C2-C6 alkynyl, C3-C6 cycloalkyl, heterocyclyl, aryl and heteroaryl;
  • R7 is hydrogen, SO n R9, COR10, a substituted straight or branched C1-C6 alkyl or an optionally substituted group selected from straight or branched C2-C6 alkenyl, straight or branched C2-C6 alkynyl, C3-C6 cycloalkyl, heterocyclyl, aryl and heteroaryl or
  • R6 and R7 taken together with the nitrogen atom to which they are bound, may form an optionally substituted heterocyclyl group
  • R8 is hydrogen, A, COR10 or an optionally substituted group selected from straight or branched C2-C6 alkenyl, straight or branched C2-C6 alkynyl, C3-C6 cycloalkyl, heterocyclyl, aryl and heteroaryl, wherein A is as defined above;
  • R9 is NR11 R12 or an optionally substituted group selected from straight or branched ⁇ - ⁇ alkyl, straight or branched C2-C6 alkenyl, straight or branched C2-C6 alkynyl, C3-C6 cycloalkyl, heterocyclyl, aryl and heteroaryl;
  • R10 is hydrogen, NR11 R12, OR13 or an optionally substituted group selected from straight or branched O- C6 alkyl, straight or branched C2-C6 alkenyl, straight or branched C2-C6 alkynyl, C3-C6 cycloalkyl, heterocyclyl, aryl and heteroaryl;
  • R11 and R12 taken together with the nitrogen atom to which they are bound, may form an optionally substituted heterocyclyl group
  • R13 is hydrogen, COR10 or an optionally substituted group selected from straight or branched ⁇ - ⁇ alkyl, straight or branched C2-C6 alkenyl, straight or branched C2-C6 alkynyl, C3-C6 cycloalkyl, heterocyclyl, aryl and heteroaryl, wherein R10 is as defined above;
  • X is a bond or an optionally substituted group selected from straight or branched ⁇ - ⁇ alkyl, heterocycly and aryl
  • Y is a bond, oxygen, or an optionally substituted group selected from straight or branched ⁇ - ⁇ alkyl, straight or branched C2-C6 alkenyl, straight or branched C2-C6 alkynyl, heterocyclyl and aryl;
  • Z is a bond, oxygen or an optionally substituted straight or branched C1-C6 alkyl
  • Ar' is an optionally substituted aryl or an optionally substituted heteroaryl
  • the present invention also provides methods of synthesizing the substituted indazole derivatives of formula (I) prepared through a process consisting of standard synthetic transformations and isomers, tautomers, hydrates, solvates, complexes, metabolites, prodrugs, carriers, N-oxides.
  • the present invention also provides a method of treating diseases caused by and/or associated with deregulated protein kinase activity, particularly ABL, ACK1 , AKT1 , ALK, AUR1 , AUR2, BRK, BUB1 , CDC7/DBF4, CDK2/CYCA, CHK1 , CK2, EEF2K, EGFR1 , EphA2, EphB4, ERK2, FAK, FGFR1 , FLT3, GSK3beta, Haspin, IGFR1 , IKK2, IR, JAK1 , JAK2, JAK3, KIT, LCK, LYN, MAPKAPK2, MELK, MET, MNK2, MPS1 , MST4, NEK6, NIM1 , P38alpha, PAK4, PDGFR, PDK1 , PERK, PIM1 , PIM2, PKAalpha, PKCbeta, PLK1 , RET, ROS1 , SULU1 , Syk
  • a preferred method of the present invention is to treat a disease caused by and/or associated with deregulated protein kinase activity selected from the group consisting of cancer, cell proliferation disorders and immune cell- associated diseases and disorders.
  • Another preferred method of the present invention is to treat specific types of cancer selected from the group consisting of, but not limited to, carcinoma such as bladder, breast, colon, kidney, liver, lung (including small cell lung cancer), salivary gland, esophagus, gall-bladder, ovary, pancreas, stomach, cervix, thyroid, thymus, prostate, and skin, including squamous cell carcinoma; hematopoietic tumors of lymphoid lineage, including leukemia, acute lymphocitic leukemia, acute lymphoblastic leukemia, B-cell lymphoma, T-cell-lymphoma, Hodgkin's lymphoma, non- Hodgkin's lymphoma, hairy cell lymphoma and Burkitt's lymphoma; hematopoietic tumors of myeloid lineage, including acute and chronic myelogenous leukemias, myelodysplastic syndrome and promyelocytic leukemia; tumors of me
  • Another preferred method of the present invention is to treat specific cellular proliferation disorders such as, for example, benign prostate hyperplasia, familial adenomatosis polyposis, neurofibromatosis, psoriasis, vascular smooth cell proliferation associated with atherosclerosis, pulmonary fibrosis, arthritis, glomerulonephritis and postsurgical stenosis and restenosis.
  • specific cellular proliferation disorders such as, for example, benign prostate hyperplasia, familial adenomatosis polyposis, neurofibromatosis, psoriasis, vascular smooth cell proliferation associated with atherosclerosis, pulmonary fibrosis, arthritis, glomerulonephritis and postsurgical stenosis and restenosis.
  • Another preferred method of the present invention is to treat immune cell-associated diseases and disorders, such as inflammatory and autoimmune diseases, for examples multiple sclerosis, systemic lupus erythematosis, inflammatory bowel diseases (IBD), Crohn's disease, irritable bowel syndrome, pancreatitis, ulcerative colitis, diverticulosis, myasthenia gravis, vasculitis, psoriasis, scleroderma, asthma, allergy, systemic sclerosis, vitiligo, arthritis such as osteoarthritis, juvenile rheumatoid arthritis, ankylosing spondylitis.
  • IBD inflammatory bowel diseases
  • Crohn's disease irritable bowel syndrome
  • pancreatitis ulcerative colitis
  • diverticulosis myasthenia gravis
  • vasculitis vasculitis
  • psoriasis scleroderma
  • asthma allergy
  • systemic sclerosis vitiligo
  • arthritis such
  • Another preferred method of the present invention is to treat FLT3 mutated cancers, such as acute myeloid leukemia or myelodisplastic syndrome.
  • Another preferred method of the present invention is to treat KIT mutated cancers, such as gastrointestinal stromal tumors, melanoma, acute myeloid leukemia, primary adenoid cystic carcinoma of the salivary gland, thymic carcinoma, glioma, testicular seminoma, small cell lung cancers, mast cell disease or piebaldism.
  • KIT mutated cancers such as gastrointestinal stromal tumors, melanoma, acute myeloid leukemia, primary adenoid cystic carcinoma of the salivary gland, thymic carcinoma, glioma, testicular seminoma, small cell lung cancers, mast cell disease or piebaldism.
  • the method of the present invention also provides tumor angiogenesis and metastasis inhibition.
  • the method of the present invention further comprises subjecting the mammal in need thereof to a radiation therapy or chemotherapy regimen in combination with at least one cytostatic or cytotoxic agent.
  • the invention provides an in vitro method for inhibiting FLT3 or KIT protein kinase activity which comprises contacting the said protein with an effective amount of a compound of formula (I).
  • the present invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof and at least one pharmaceutically acceptable excipient, carrier and/or diluent.
  • the present invention further provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of formula (I) in combination with one or more chemotherapeutic - e.g. cytostatic or cytotoxic - agents, antibiotic-type agents, alkylating agents, antimetabolite agents, hormonal agents, immunological agents, interferon-type agents, cyclooxygenase inhibitors (e.g. COX-2 inhibitors), matrixmetalloprotease inhibitors, telomerase inhibitors, tyrosine kinase inhibitors, anti-growth factor receptor agents, anti-HER agents, anti-EGFR agents, anti-angiogenesis agents (e.g.
  • chemotherapeutic e.g. cytostatic or cytotoxic - agents, antibiotic-type agents, alkylating agents, antimetabolite agents, hormonal agents, immunological agents, interferon-type agents, cyclooxygenase inhibitors (e.g. COX-2 inhibitors), matrixmetalloprotease inhibitors, telomerase inhibitors
  • angiogenesis inhibitors farnesyl transferase inhibitors, ras-raf signal transduction pathway inhibitors, cell cycle inhibitors, other cdks inhibitors, tubulin binding agents, topoisomerase I inhibitors, topoisomerase II inhibitors, and the like.
  • the invention provides a product comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof, as defined above, and one or more chemotherapeutic agents, as a combined preparation for simultaneous, separate or sequential use in anticancer therapy.
  • the invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof, as defined above, for use as a medicament.
  • the invention provides the use of a compound of formula (I) or a pharmaceutically acceptable salt thereof, as defined above, in the manufacture of a medicament with antitumor activity.
  • the compounds of formula (I) may have one or more asymmetric centres, and may therefore exist as individual optical isomers or racemic mixtures. Accordingly, all the possible isomers, and their mixtures, of the compounds of formula (I) are within the scope of the present invention.
  • N-oxides are compounds of formula (I) wherein nitrogen and oxygen are tethered through a dative bond.
  • heterocyclyl refers to a 3- to 7-membered, saturated or partially unsaturated carbocyclic ring where one or more carbon atoms are replaced by heteroatoms such as nitrogen, oxygen and sulfur.
  • heterocyclyl groups are, for instance, oxiranyl, aziridinyl, oxetanyl, azetidinyl, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothiophenyl, dihydrothiophenyl, pyrrolidinyl, dihydropyrrolyl, pyranyl, dihydropyranyl, tetrahydropyranyl, tetrahydrothiopyranyl, piperidinyl, pyrazolinyl, isoxazolidinyl, isoxazolinyl, thiazolidinyl, thiazolinyl, isothiazolinyl, dioxanyl, piperazin
  • a heterocyclyl group may be substituted or unsubstituted; when not otherwise specified, the substituent groups are preferably one to three, independently selected from the group consisting of halogen, cyano, nitro, SO n R9, COR10, NR11 R12, OR13, R11 R12N-(Ci-C 6 )-alkyl, R130-(Ci-C 6 )-alkyl and an optionally further substituted straight or branched Ci-C 6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, heterocyclyl, aryl and heteroaryl, wherein R9, R10, R11 , R12, R13 and n are as defined above.
  • heteroaryl refers to aromatic heterocyclic rings, typically 5- to 7-membered heterocycles with from 1 to 3 heteroatoms selected among N, 0 and S; the heteroaryl ring can be optionally further fused or linked to aromatic and non-aromatic carbocyclic and heterocyclic rings.
  • heteroaryl groups are, for instance, pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, indolyl, imidazolyl, thiazolyl, isothiazolyl, pyrrolyl, phenyl-pyrrolyl, furyl, phenyl-furyl, oxazolyl, isoxazolyl, pyrazolyl, thienyl, benzothienyl, isoindolinyl, benzoimidazolyl, quinolinyl, isoquinolinyl, 1 ,2,3-triazolyl, 1 -phenyl-1 ,2,3-triazolyl, 2,3-dihydroindolyl, 2,3-dihydrobenzofuranyl, 2,3- dihydrobenzothiophenyl; benzopyranyl, 2,3-dihydrobenzoxazinyl, 2,3-di
  • the aryl and heteroaryl groups may be substituted or unsubstituted; when not otherwise specified, the substituent groups are preferably one to three, independently selected from the group consisting of halogen, cyano, nitro, S0 n R9, COR10, NR11R12, 0R13, R11 R12N-(Ci-C 6 )-alkyl, R130-(Ci-C 6 )-alkyl and an optionally further substituted straight or branched C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, heterocyclyl, aryl and heteroaryl, wherein R9, R10, R11 , R12, R13 and n are as defined above.
  • halogen indicates fluorine, chlorine, bromine or iodine.
  • C2-C6 alkenyl indicates an aliphatic C2-C6 hydrocarbon chain containing at least one carbon-carbon double bond and which can be straight or branched. Representative examples include, but are not limited to, ethenyl, 1-propenyl, 2-propenyl, 1- or 2-butenyl, and the like.
  • C2-C6 alkynyl indicates an aliphatic C2-C6 hydrocarbon chain containing at least one carbon-carbon triple bond and which can be straight or branched. Representative examples include, but are not limited to, ethynyl, 1- propynyl, 2-propynyl, 1- or 2-butynyl, and the like.
  • the alkenyl and alkynyl groups may be substituted or unsubstituted; when not otherwise specified, the substituent groups are preferably one to three, independently selected from the group consisting of halogen, cyano, nitro, SO n R9, COR10, NR11R12, OR13, R11 R12N-(Ci-C 6 )-alkyl, R130-(Ci-C 6 )-alkyl and an optionally further substituted straight or branched C1-C6 alkyl, C3-C6 cycloalkyl, heterocyclyl, aryl and heteroaryl, wherein R9, R10, R11 , R12, R13 and n are as defined above.
  • nitro indicates a -NO2 group.
  • salts of compounds of formula (I) refers to those salts that retain the biological effectiveness and properties of the parent compound.
  • Such salts include acid addition salts with inorganic acids such as hydrochloric, hydrobromic, nitric, phosphoric, sulfuric, perchloric acid and the like, or with organic acids such as acetic, trifluoroacetic, propionic, glycolic, lactic, (D) or (L) malic, maleic, fumaric, methanesulfonic, ethanesulfonic, benzoic, p-toluenesulfonic, salicylic, cinnamic, mandelic, tartaric, citric, succinic, malonic acid and the like; salts formed when an acidic proton present in a compound of formula (I) is either replaced by a metal ion, - e.g.
  • an alkali metal ion such as sodium or potassium - or an alkaline earth ion, such as calcium or magnesium, or coordinates with an organic base such as ethanolamine, diethanolamine, triethanolamine, tromethamine, N-methylglucamine, and the like.
  • a preferred class of compounds of formula (I) are the compounds wherein:
  • R1 is A, NR6R7, OR8 or an optionally substituted heterocyclyl, wherein A, R6, R7 and R8 are as defined above.
  • a more preferred class of compounds of formula (I) are the compounds wherein:
  • Ar is AM or Ar2; and R2, R3, R4, R5 are each independently hydrogen, halogen, NR11 R12 or OR13, wherein R11 , R12 and R13 are as defined above.
  • the present invention also provides a process for the preparation of a compound of formula (I) as defined above, by using the reaction routes and synthetic schemes described below, employing the techniques available in the art and starting materials readily available.
  • the preparation of certain embodiments of the present invention is described in the examples that follow, but those of ordinary skill in the art will recognize that the preparations described may be readily adapted to prepare other embodiments of the present invention.
  • the synthesis of non- exemplified compounds according to the invention may be performed by modifications apparent to those skilled in the art, for instance by appropriately protecting interfering groups, by changing to other suitable reagents known in the art, or by making routine modifications of reaction conditions.
  • other reactions referred to herein or known in the art will be recognized as having adaptability for preparing other compounds of the invention.
  • Ar is as defined in formula (I); W is hydroxy, halogen or a suitable leaving group; X, Y, Z and Ar' are as defined in formula (I); and L is a suitable leaving group, such as halogen, methanesulfonyloxy, trifluoromethanesulfonyloxy or p-toluenesulfonyloxy.
  • Ar is as defined in formula (I); W is hydroxy, halogen or a suitable leaving group; X, Y, Z and Ar' are as defined in formula (I); and L is a suitable leaving group, such as halogen, methanesulfonyloxy, trifl uoromethanesulfonyloxy or p-toluenesulfonyloxy.
  • a process of the present invention comprises the following steps:
  • Ar', Z and Y are as defined in formula (I) and X is an optionally substituted group selected from straight or branched ⁇ -0 ⁇ alkyl and heterocyclyl;
  • Ar' is as defined in formula (I) and X, Y and Z are a bond;
  • X is an optionally substituted group selected from straight or branched C1-C6 alkyl or heterocyclyl and L is a suitable leaving group, such as halogen, methanesulfonyloxy, trifl uoromethanesulfonyloxy or p-toluenesulfonyloxy;
  • the intermediate compound of formula (XI), wherein Ar, Ar', Y and Z are as defined in formula (I) and X is an optionally substituted group selected from straight or branched ⁇ - ⁇ alkyl and heterocyclyl, can be obtained in a process comprising the following steps:
  • R6 is as defined in formula (I) and R7 is hydrogen, a substituted straight or branched C1-C6 alkyl or an optionally substituted group selected from straight or branched C2-C6 alkenyl, straight or branched C2-C6 alkynyl, C3- C6 cycloalkyi, heterocyclyl, aryl and heteroaryl, so as to obtain a compound of formula (I), wherein R1 is NR6R7, wherein R6 is as defined in formula (I) and R7 is hydrogen, a substituted straight or branched C1-C6 alkyl or an optionally substituted group selected from straight or branched C2-C6 alkenyl, straight or branched C2-C6 alkynyl, C3- C6 cycloalkyi, heterocyclyl, aryl and heteroaryl, and Ar, Ar', X, Y and Z are as defined in formula (I); optionally separating the resultant compound of
  • R9, R10 and W are as defined above, for obtaining the corresponding compound of formula (I) wherein such substituent is a NHSO2R9 or NHCOR10 residue, wherein R9 and R10 are as defined above;
  • NR6R7 group wherein R7 is hydrogen and R6 is as defined in formula (I) except hydrogen;
  • R11 and R12 are as defined in formula (I) except SO n R9 or COR10, for obtaining the corresponding compound of formula (I) wherein such substituent is a CONR11 R12 residue, wherein R11 and R12 are as defined in formula (I) except SO n R9 or COR10;
  • step A) the transformation of the compound of formula (II) into the compound of formula (III) can be accomplished in a variety of ways and experimental conditions, which are widely known in the art for the introduction of the tert-butoxy-carbonyl group, for example using di-tert-butyl dicarbonate.
  • this reaction is carried out in a suitable solvent such as, for instance, tetrahydrofuran, dichloromethane, toluene, 1 ,4-dioxane, and in the presence of a proton scavenger such as, for example, pyridine, triethylamine, ⁇ , ⁇ -diisopropylethylamine, at a temperature ranging from room temperature to reflux, for a time ranging from about 30 min. to about 96 hours.
  • a suitable solvent such as, for instance, tetrahydrofuran, dichloromethane, toluene, 1 ,4-dioxane
  • a proton scavenger such as, for example, pyridine, triethylamine, ⁇ , ⁇ -diisopropylethylamine
  • step B) the cleavage of the phthalimido group of the compound of formula (III) to give the compound of formula (IV) can be accomplished in a variety of ways and experimental conditions, which are widely known in the art, for example using hydrazine.
  • this reaction is carried out in a suitable solvent such as, for instance, tetrahydrofuran, dichloromethane, toluene, 1 ,4-dioxane, at a temperature ranging from room temperature to reflux, for a time ranging from about 30 min. to about 96 hours.
  • a suitable solvent such as, for instance, tetrahydrofuran, dichloromethane, toluene, 1 ,4-dioxane
  • a compound of formula (VI) can be obtained by reacting a compound of formula (IV) with a compound of formula (V) in a variety of ways and experimental conditions, which are widely known in the art for acylation reactions.
  • a compound of formula (V) wherein W is hydroxy is converted into its corresponding acyl chloride wherein W is chlorine in the presence of thionyl chloride or oxalyl chloride, in a suitable solvent, such as toluene, dichloromethane, chloroform, diethyl ether, tetrahydrofuran, 1 ,4-dioxane, or a mixture thereof, at a temperature ranging from about -10°C to reflux and for a period of time varying from about 1 hour to about 96 hours.
  • the acyl chloride is isolated by evaporation of the solvent and further reacted with (IV) in the presence of a base such as pyridine, triethylamine or ⁇ , ⁇ -diisopropylethylamine, at a temperature ranging from about -40°C to reflux and for a period of time varying from about 1 hour to about 96 hours.
  • a suitable solvent may also be added, such as toluene, dichloromethane, chloroform, diethyl ether, tetrahydrofuran, 1 ,4-dioxane.
  • a compound of formula (IV) is reacted with a compound of formula (V) wherein W is hydroxy in the presence of an activating agent such as hydroxybenzotriazole, dicyclohexyl carbodiimide, diisopropyl carbodiimide, 1-ethyl-3-(3'- dimethylamino)carbodiimide hydrochloric acid salt, 0-(benzotriazol-1-yl)-N,N,N',N'-tetramethyluronium tetrafluoroborate.
  • an activating agent such as hydroxybenzotriazole, dicyclohexyl carbodiimide, diisopropyl carbodiimide, 1-ethyl-3-(3'- dimethylamino)carbodiimide hydrochloric acid salt, 0-(benzotriazol-1-yl)-N,N,N',N'-tetramethyluronium tetrafluoroborate.
  • this reaction is carried out in a suitable solvent such as, for instance, tetrahydrofuran, dichloromethane, toluene, 1 ,4-dioxane, N,N-dimethylformamide, ⁇ , ⁇ -dimethylacetamide and in the presence of a proton scavenger such as, for example, pyridine, triethylamine, ⁇ , ⁇ -diisopropylethylamine, at a temperature ranging from room temperature to reflux, for a time ranging from about 30 min. to about 96 hours.
  • a suitable solvent such as, for instance, tetrahydrofuran, dichloromethane, toluene, 1 ,4-dioxane, N,N-dimethylformamide, ⁇ , ⁇ -dimethylacetamide
  • a proton scavenger such as, for example, pyridine, triethylamine, ⁇ , ⁇ -diisopropyleth
  • step D) the selective cleavage of the tert-butyldimethylsilyl ether of the compound of formula (VI) to give the compound of formula (VII) can be carried out in a variety of ways, according to conventional methods well known in the literature.
  • this conversion is carried out in the presence of tetrabutylammonium fluoride in a suitable solvent, such as, for instance, tetrahydrofuran, dichloromethane, toluene, 1 ,4-dioxane, at a temperature ranging from -10°C to reflux, for a time ranging from about 30 min. to about 96 hours.
  • a suitable solvent such as, for instance, tetrahydrofuran, dichloromethane, toluene, 1 ,4-dioxane
  • step Ea) the coupling of a compound of formula (VII) with an alcohol of formula (VIII) to give a compound of formula (XI) can be accomplished in a variety of ways and experimental conditions which are widely known in the art for the synthesis of aryl ethers under Mitsunobu-like conditions.
  • this conversion is carried out in the presence of an azodicarboxylate, such as, for example, diethyl azodicarboxylate, diisopropyl azodicarboxylate or di-tert-butyl azodicarboxylate and a phosphine, such as, for example, triphenylphosphine or polymer-bound triphenylphosphine, in a suitable solvent, such as, for instance, tetrahydrofuran, dichloromethane, 1 ,4-dioxane, toluene, acetonitrile at a temperature ranging from -20°C to reflux, for a time ranging from about 30 min. to about 96 hours.
  • an azodicarboxylate such as, for example, diethyl azodicarboxylate, diisopropyl azodicarboxylate or di-tert-butyl azodicarboxylate and a phosphine, such as, for
  • step Eb) the coupling of a compound of formula (VII) with a boronic acid of formula (IX) to give a compound of formula (XI) can be accomplished in a variety of ways and experimental conditions which are widely known in the art for the synthesis of di-aryl ethers.
  • this conversion is carried out in the presence of copper diacetate and 4A molecular sieve or silica gel, in a suitable solvent, such as, for instance, tetrahydrofuran, dichloromethane, 1 ,4-dioxane and in the presence of a proton scavenger such as, for example, pyridine, triethylamine, ⁇ , ⁇ -diisopropylethylamine, at a temperature ranging from -10°C to reflux, for a time ranging from about 30 min. to about 96 hours.
  • a suitable solvent such as, for instance, tetrahydrofuran, dichloromethane, 1 ,4-dioxane
  • a proton scavenger such as, for example, pyridine, triethylamine, ⁇ , ⁇ -diisopropylethylamine, at a temperature ranging from -10°C to reflux, for a time ranging from about 30 min. to about
  • step Ec) the coupling of a compound of formula (VII) with a compound of formula (X) to give a compound of formula (XI) can be accomplished in a variety of ways and experimental conditions which are widely known in the art for the alkylation of phenols.
  • a compound of formula (VII) is treated with a chloride, bromide, iodide, mesylate or triflate of formula (X), in which case L represents chlorine, bromine, iodine, methanesulfonyloxy or trifluoromethanesulfonyloxy, respectively, in the presence of a proton scavenger such as, for example, triethylamine, ⁇ , ⁇ -diisopropylethylamine, sodium, potassium or cesium carbonate, in a suitable solvent such as, for instance, tetrahydrofuran, 1 ,4-dioxane, ⁇ , ⁇ -dimethylformamide, N,N-dimethylacetamide, dimethoxyethane, at a temperature ranging from -10°C to reflux, for a time ranging from about 30 min. to about 96 hours.
  • a proton scavenger such as, for example, triethylamine, ⁇
  • step F) the transformation of a compound of formula (XI) into a compound of formula (I) can be carried out in a variety of ways, according to conventional methods well known in the literature for the cleavage of a tert- butoxy-carbonyl group.
  • this reaction may be run under acidic conditions, for example in the presence of an inorganic or organic acid such as hydrochloric, trifluoroacetic or methanesulfonic acid, in a suitable solvent such as dichloromethane, 1 ,4-dioxane, a lower alcohol, such as methanol or ethanol, water, or a mixture thereof, at a temperature ranging from room temperature to reflux and for a period of time ranging from about 30 min. to about 96 hours.
  • an inorganic or organic acid such as hydrochloric, trifluoroacetic or methanesulfonic acid
  • a suitable solvent such as dichloromethane, 1 ,4-dioxane
  • a lower alcohol such
  • step G) the transformation of the compound of formula (IV) into the compound of formula (XII) can be carried out in a way analogous to that specified above under D).
  • step Ha) the coupling between the compound of formula (XII) and an alcohol of formula (VIII) can be carried out in a way analogous to that specified above under Ea).
  • step Hb) the coupling between the compound of formula (XII) and a compound of formula (X) can be carried out in a way analogous to that specified above under Ec).
  • step I) the acylation of the compound of formula (XIII) with a compound of formula (V) can be carried out in a way analogous to that specified above under C).
  • a compound of formula (XV) can be transformed into a compound of formula (XVI) in a variety of ways and experimental conditions.
  • this reaction is carried out in the presence of hydrazine or hydrazine monohydrate in a suitable solvent such as, for instance, toluene, tetrahydrofuran, 1 ,4-dioxane, dimethyl sulfoxide, acetonitrile, methanol, ethanol or n-butanol, at a temperature ranging from 0 °C to reflux and for a period of time varying from about 30 min to about 96 hours.
  • a suitable solvent such as, for instance, toluene, tetrahydrofuran, 1 ,4-dioxane, dimethyl sulfoxide, acetonitrile, methanol, ethanol or n-butanol
  • step N) the coupling of a compound of formula (XVIII) with an amine of formula (XIX) can be carried out in a variety of ways, according to conventional methods well known in the literature for Buchwald-Hartwig aminations.
  • a compound of formula (XVIII) wherein L is chlorine, bromine, iodine or trifluoromethanesulfonyloxy is reacted with a compound of formula (XIX) in a suitable solvent such as, for example, tetrahydrofuran, 1 ,4-dioxane, N,N-dimethylformamide, ⁇ , ⁇ -dimethylacetamide, dimethoxyethane, acetonitrile, toluene, in the presence of catalytic amounts of a palladium derivative, such as, for example, tris(dibenzylideneacetone)dipalladium(0), palladium diacetate, and a phopsphine ligand, such
  • the reduction of a compound of formula (I) wherein one of the substituents R1 , R2, R3, R4 or R5 is nitro, for obtaining a compound of formula (I) wherein such substituent is amino can be carried out in a variety of ways, according to conventional methods well known in the literature.
  • this conversion is carried out in a suitable solvent such as, for instance, methanol, ethanol, water, tetrahydrofuran, 1 ,4-dioxane, N,N- dimethylformamide, acetic acid, or a mixture thereof, in the presence of a suitable reducing agent, such as, for instance, hydrogen and a hydrogenation catalyst, or by treatment with cyclohexene or cyclohexadiene, or formic acid or ammonium formate and a hydrogenation catalyst, or a metal such as iron or zinc in the presence of an inorganic acid, such as hydrochloric acid, or by treatment with tin (II) chloride or sodium hydrosulfite in the presence of tetrabutylammonium chloride, at a temperature ranging from 0°C to reflux and for a time varying from about 1 hour to about 96 hours.
  • the hydrogenation catalyst is usually a metal, most often palladium, which can be used as such or supported on carbon.
  • the reaction of a compound of formula (I), wherein the substituent R1 is NH2, with a suitable aldehyde or ketone for obtaining a compound of formula (I) wherein such substituent is a NR6R7 group can be conducted in a variety of ways, according to conventional methods for carrying out reductive alkylations.
  • this reaction is carried out in a suitable solvent such as, for instance, methanol, N,N-dimethylformamide, dichloromethane, tetrahydrofuran, or a mixture thereof, in the presence of a suitable reducing agent such as, for instance, sodium borohydride, tetra-alkylammonium borohydride, sodium cyano borohydride, sodium triacetoxyborohydride, tetramethylammonium triacetoxy borohydride and in the presence of an acid catalyst, such as, for instance, acetic acid or trifluoroacetic acid, at a temperature ranging from about 0°C to reflux and for a time varying from about 1 hour to about 96 hours.
  • a suitable solvent such as, for instance, methanol, N,N-dimethylformamide, dichloromethane, tetrahydrofuran, or a mixture thereof
  • a suitable reducing agent such as, for instance, sodium borohydride, tetra-al
  • the reaction of a compound of formula (I), wherein one of the substituents R2, R3, R4 or R5 is NH2, with a suitable aldehyde or ketone in the presence of a reducing agent, for obtaining a compound of formula (I) wherein such substituent is a NR11 R12 group can be conducted in a variety of ways, according to conventional methods for carrying out reductive alkylations. Preferably this conversion is carried out in a way analogous to that specified above under 3).
  • the hydrolysis of a compound of formula (I) wherein one of the substituents R1 , R2, R3, R4 or R5 is a COOR13 residue, wherein R13 is a straight or branched ⁇ - ⁇ alkyl, to give the corresponding carboxylic acid can be conducted in a variety of ways, according to methods widely known in the art for the hydrolysis of ester groups.
  • such hydrolysis is carried out in the presence of an inorganic base, such as, for example, lithium, sodium or potassium hydroxide, or an inorganic or organic acid, such as, for example, hydrochloric acid, trifluoroacetic acid, in a suitable solvent such as, for instance, methanol, ethanol, tetrahydrofuran, 1 ,4-dioxane, water or a mixture thereof, at a temperature ranging from about 0°C to reflux and for a time varying from about 1 hour to about 96 hours.
  • an inorganic base such as, for example, lithium, sodium or potassium hydroxide
  • an inorganic or organic acid such as, for example, hydrochloric acid, trifluoroacetic acid
  • a suitable solvent such as, for instance, methanol, ethanol, tetrahydrofuran, 1 ,4-dioxane, water or a mixture thereof, at a temperature ranging from about 0°C to reflux and for a time varying
  • the amidation of a compound of formula (I) wherein one of the substituents R1 , R2, R3, R4 or R5 is a COOH residue, with an amine of formula (XXII), can be can be conducted in a variety of ways, according to conventional methods for the synthesis of carboxamides.
  • this conversion is carried out in the presence of an activating agent such as hydroxybenzotriazole, dicyclohexyl carbodiimide, diisopropyl carbodiimide, 1-ethyl-3-(3'-dimethylamino)carbodiimide hydrochloric acid salt, 0-(benzotriazol-1-yl)-N,N,N',N'-tetramethyluronium tetrafluoroborate, in a suitable solvent such as, for instance, tetrahydrofuran, dichloromethane, toluene, 1 ,4-dioxane, N,N-dimethylformamide, ⁇ , ⁇ -dimethylacetamide and in the presence of a proton scavenger such as, for example, pyridine, triethylamine, ⁇ , ⁇ -diisopropylethylamine, at a temperature ranging from room temperature to reflux, for a time ranging from about
  • the oxidation of a compound of formula (I) wherein R1 is 4-methyl-piperazin-1-yl for obtaining a compound of formula (I) wherein such substituent is 4-methyl-4-oxy-piperazin-1-yl can be conducted in a variety of ways, according to conventional methods for the N-oxidation of tertiary amines.
  • this conversion is carried out in the presence of an oxidizing agent such as, for example, 3-chloroperbenzoic acid, hydrogen peroxide, dimethyldioxirane, in a suitable solvent such as, for instance, dichloromethane, methanol, ethanol, water, acetone, or a mixture thereof, at a temperature ranging from -10°C to reflux, for a time ranging from about 30 min. to about 96 hours.
  • an oxidizing agent such as, for example, 3-chloroperbenzoic acid, hydrogen peroxide, dimethyldioxirane
  • a suitable solvent such as, for instance, dichloromethane, methanol, ethanol, water, acetone, or a mixture thereof
  • such reaction can be carried out by treatment with an inorganic acid, such as hydrochloric, sulphuric or perchloric acid, or an organic acid, such as trifluoroacetic or methanesulfonic acid, in a suitable solvent, such as water, methanol, ethanol, 1 ,4-dioxane, tetrahydrofuran, diethyl ether, diisopropyl ether, acetonitrile, ⁇ , ⁇ -dimethylformamide, dichloromethane or mixtures thereof, at a temperature ranging from -20°C to 80°C, and for a period of time ranging from 30 minutes to 48 hours.
  • an inorganic acid such as hydrochloric, sulphuric or perchloric acid
  • an organic acid such as trifluoroacetic or methanesulfonic acid
  • a suitable solvent such as water, methanol, ethanol, 1 ,4-dioxane, tetrahydrofur
  • a compound of formula (I) contains one or more asymmetric centers
  • said compound can be separated into the single isomers by procedures known to those skilled in the art. Such procedures comprise standard chromatographic techniques, including chromatography using a chiral stationary phase, or crystallization. General methods for separation of compounds containing one or more asymmetric centers are reported, for instance, in Jacques, Jean; Collet, Andre; Wilen, Samuel H., - Enantiomers, Racemates, and Resolutions, John Wiley & Sons Inc., New York (NY), 1981.
  • a compound of formula (I) can also be transformed into a pharmaceutically acceptable salt according to standard procedures that are known to those skilled in the art.
  • a compound of formula (I) that is obtained as a salt can be transformed into the free base or the free acid according to standard procedures that are known to the skilled person.
  • any variant of the process for preparing the compounds of formula (I), the starting materials and any other reactant i.e. compounds of formula (II), (V), (VIII), (IX), (X), (XIV), (XVII), (XIX), (XX), (XXI) and (XXII) are either commercially available, known, or easily prepared according to well-known methods described, for instance, in: B.M.Trost and I. Fleming, Comprehensive Organic Synthesis, 1991 , Pergamon Press; A.R. Katritzky, 0. Meth-Cohn and C.W. Rees, Comprehensive Organic Functional Group Transformations, 1995, Elsevier Pergamon; A.R. Katritzky and R.J.K. Taylor, Comprehensive Organic Functional Group Transformations II, 2005, Elsevier Pergamon.
  • the compound of formula (II) can be prepared as described in WO2003028720 and the compound of formula (XIV) is commercially available.
  • the compounds of the present invention can be administered either as single agents or, alternatively, in combination with known anticancer treatments such as radiation therapy or chemotherapy regimen in combination with cytostatic or cytotoxic agents, antibiotic-type agents, alkylating agents, antimetabolite agents, hormonal agents, immunological agents, interferon-type agents, cyclooxygenase inhibitors (e.g. COX-2 inhibitors), matrixmetalloprotease inhibitors, telomerase inhibitors, tyrosine kinase inhibitors, anti-growth factor receptor agents, anti-HER agents, anti-EGFR agents, anti-angiogenesis agents (e.g.
  • cytostatic or cytotoxic agents antibiotic-type agents, alkylating agents, antimetabolite agents, hormonal agents, immunological agents, interferon-type agents, cyclooxygenase inhibitors (e.g. COX-2 inhibitors), matrixmetalloprotease inhibitors, telomerase inhibitors, tyrosine kinase inhibitors, anti-growth factor receptor agents, anti
  • such combination products employ the compounds of this invention within the dosage range described below and the other pharmaceutically active agent within the approved dosage range.
  • the solid oral forms may contain, together with the active compound, diluents, e.g., lactose, dextrose saccharose, sucrose, cellulose, corn starch or potato starch; lubricants, e.g., silica, talc, stearic acid, magnesium or calcium stearate, and/or polyethylene glycols; binding agents, e.g., starches, arabic gum, gelatine methylcellulose, carboxymethylcellulose or polyvinyl pyrrolidone; disintegrating agents, e.g., starch, alginic acid, alginates or sodium starch glycolate; effervescing mixtures; dyestuffs; sweeteners; wetting agents such as lecithin, polysorbates, laurylsulphates; and, in general, nontoxic and pharmacologically inactive substances used in pharmaceutical formulations.
  • diluents e.g., lactose, dextrose saccharose, sucrose
  • the liquid dispersions for oral administration may be, e.g., syrups, emulsions and suspensions.
  • the syrups may contain, as carrier, saccharose or saccharose with glycerine and/or mannitol and sorbitol.
  • the suspensions and the emulsions may contain, as examples of carriers, natural gum, agar, sodium alginate, pectin, methylcellulose, carboxymethylcellulose, or polyvinyl alcohol.
  • the suspension or solutions for intramuscular injections may contain, together with the active compound, a pharmaceutically acceptable carrier, e.g., sterile water, olive oil, ethyl oleate, glycols, e.g., propylene glycol and, if desired, a suitable amount of lidocaine hydrochloride.
  • the suppositories may contain, together with the active compound, a pharmaceutically acceptable carrier, e.g., cocoa butter, polyethylene glycol, a polyoxyethylene sorbitan fatty acid ester surfactant or lecithin.
  • a pharmaceutically acceptable carrier e.g., cocoa butter, polyethylene glycol, a polyoxyethylene sorbitan fatty acid ester surfactant or lecithin.
  • TLC Thin-layer chromatography
  • Electrospray (ESI) mass spectra were obtained on a Finnigan LCQ ion trap. Unless otherwise specified, all final compounds were homogeneous (purity of not less than 95%), as determined by high-performance liquid chromatography (HPLC). HPLC-UV-MS analyses, used to assess compound purity, were carried out combining the ion trap MS instrument with HPLC system SSP4000 (Thermo Separation Products) equipped with an autosampler LC Pal (CTC Analytics) and UV6000LP diode array detector (UV detection 215-400 nm). Instrument control, data acquisition and processing were performed with the Xcalibur 1.2 software (Finnigan).
  • HPLC chromatography was run at r.t., and 1 mL/min flow rate, using a Waters X Terra RP 18 column (4.6x 50 mm; 3.5 ⁇ ).
  • Mobile phase A was ammonium acetate 5 mM buffer (pH 5.5 with acetic acid): acetonitrile 90:10
  • mobile phase B was ammonium acetate 5 mM buffer (pH 5.5 with acetic acid): acetonitrile 10:90; the gradient was from 0 to 100% B in 7 minutes then hold 100% B for 2 minutes before requilibration.
  • Methanesulfonic acid 2-(4-trifluoromethyl-benzyloxy)-ethyl ester To a solution of 2-(4-trifluoromethyl-benzyloxy)-ethanol (1.0 g, 4.5 mmol) in dry DCM (20 ml) and DIPEA (2.36 ml, 13.5 mmol), at 0°C, under argon atmosphere, was added methanesulfonyl chloride (421 ⁇ , 5.4 mmol). The reaction mixture was stirred at 0°C for 10 minutes, then the ice-bath removed and the stirring continued for 2 hours at r.t..
  • the reaction mixture was heated to reflux and stirred for 15 min then cooled to r.t., poured into 200 ml of water and extracted with 200 ml of EtOAc. The organic layer was separated, dried over sodium sulfate and evaporated to dryness.
  • the crude residue was purified by chromatography (Biotage SP1 Flash Purification system) on a silica gel cartridge (Varian SF40-120g) using DCM as eluant A and DCM / 7N NH 3 in MeOH 10:1 as eluant B.
  • the precipitated solid was filtered, washed with water, dried and purified by chromatography (Biotage SP1 Flash Purification system) on a silica gel cartridge (Biotage SNAP 25g) using DCM as eluant A and DCM / 7N NH 3 in MeOH 10:1 as eluant B. Elution with a gradient from A / B 100:0 to 0:100 over 15 CV, followed by an isocratic elution with eluant B (5 CV), gave a yellow solid that was triturated with diethyl ether (15 ml) affording 111 mg (yield: 75%) of the title compound as a white solid.
  • FLT3 cytoplasmic domain (aa 564-993end of the 993 aminoacid long full length sequence, accession number P36888 of UniProtKB/Swiss-Prot. database) was amplified by PCR starting from a testis cDNA library and then cloned into pVL vector for expression in insect cells through the baculovirus system.
  • the GST-FLT3 cytoplasmic domain has been expressed in Sf21 cells infected for 72 hours at 27°C.
  • the recombinant protein has been purified by affinity on GSH-sepharose and eluted with glutathione. A further purification step has been performed on heparine sepharose.
  • KIT cytoplasmic domain (aa 544-976end of the 976 aminoacid long full length sequence, accession number P10721 of UniProtKB/Swiss-Prot database) was cloned into pVL vector for expression in insect cells through the baculovirus system.
  • the GST-KIT cytoplasmic domain has been expressed in Sf21 cells infected for 66 hrs at 27°C.
  • the recombinant protein has been purified by affinity on GSH-sepharose and eluted with glutathione.
  • the final yield was of 9 mg/billion cells and the protein resulted >80% pure by coomassie staining.
  • Enzyme - The assay was performed using FLT3 cytoplasmic domain product and purified in house as GST fused protein.
  • the FLT3 protein (1 microM) was pre activated with 800 microM ATP for 1 hour at 28 * C in order to obtain a linear kinetic.
  • FLT3 Kinase Buffer (KB) - Kinase buffer was composed of 50 mM HEPES pH 7.9 containing 4 mM MgC , 1 mM DTT, 10 microM Na 3 V0 4 , and 0.2 mg/mL BSA
  • Enzyme - The assay has been performed using KIT cytoplasmic domain product and purified in house as GST fused protein.
  • the KIT protein (4.5 microM ) was pre activated with 300 microM ATP for 1 hour at 28 * C in order to obtain a linear kinetic.
  • KIT kinase Buffer (KB) - Kinase buffer was composed of 50 mM HEPES pH 7.9 containing 5 mM MgC , 1 mM MnCI 2 , 10 mM DTT, 3 microM Na 3 V0 4 , and 0.2 mg/mL BSA
  • test compounds were received as a 1 mM solution in 100% DMSO, distributed into 96 well plates: compounds were then plated into the first column of a microtiter plate (A1 to G1), 100 microL/well.
  • An automated station for serial dilutions Biomek FX, Beckman was used for producing 1 :3 dilutions in 100 % DMSO, from line A1 to A10, and for all the compounds in the column.
  • Each 384 well-plate contained at least one curve of the standard inhibitor staurosporine and reference wells (total enzyme activity vs. no enzymatic activity) for the Z' and signal to background evaluation.
  • the cells were incubated at 37°C and 5 % CO2 and after 72 h the plates were processed using CellTiter-Glo assay (Promega) following the manufacturer's instruction.
  • CellTiter-Glo is a homogenous method based on the quantification of the ATP present, an indicator of metabolitically active cells. ATP was quantified using a system based on luciferase and D-luciferin resultant into light generation. The luminescent signal was proportional to the number of cells present in culture.
  • the IC50 values are tipically lower than 2 microM on FLT3 and lower than 3 microM on KIT.
  • the IC50 values are tipically lower than 3 microM, with 26 compounds having IC50 values lower than 0.1 microM on both cell lines.

Abstract

The present invention relates to substituted indazole compounds which modulate the activity of protein kinases and are therefore useful in treating diseases caused by degulated protein kinase activity, like cancer. The present invention also provides methods for preparing these compounds, pharmaceutical compositions comprising these compounds, and methods of treating diseases utilizing such these compounds or the pharmaceutical compositions containing them.

Description

SUBSTITUTED INDAZOLE DERIVATIVES ACTIVE AS KINASE INHIBITORS
The present invention relates to certain substituted indazole compounds, which modulate the activity of protein kinases. The compounds of this invention are therefore useful in treating diseases caused by deregulated protein kinase activity. The present invention also provides methods for preparing these compounds, pharmaceutical compositions comprising these compounds, and methods of treating diseases utilizing pharmaceutical compositions comprising these compounds.
The malfunctioning of protein kinases (PKs) is the hallmark of numerous diseases. A large share of the oncogenes and proto-oncogenes involved in human cancers encode for PKs. The enhanced activities of PKs are also implicated in many non-malignant diseases, such as benign prostate hyperplasia, familial adenomatosis, polyposis, neurofibromatosis, psoriasis, vascular smooth cell proliferation associated with atherosclerosis, pulmonary fibrosis, arthritis glomerulonephritis and post-surgical stenosis and restenosis.
PKs are also implicated in inflammatory conditions and in the multiplication of viruses and parasites. PKs may also play a major role in the pathogenesis and development of neurodegenerative disorders.
For a general reference to PKs malfunctioning or deregulation see, for instance, Current Opinion in Chemical Biology (1999) 3, 459 - 465; Cell (2000) 100, 113-127; Nature Rev. Drug Discov. (2002) 1 , 309-315; and Carcinogenesis (2008) 29, 1087-191.
A subset of PK is a group of membrane receptors with intrinsic protein-tyrosine kinase activity (RPTK). Upon binding of growth factors, RPTKs become activated and phosphorylate themselves and a series of substrates in the cytoplasm. Through this mechanism, they can transduce intracellular signallings for proliferation, differentiation or other biological changes. Structural abnormalities, over-expression and activation of RTPKs are frequently observed in human tumors, suggesting that constitutive ignition of the signal transduction leading to cell proliferation can result in malignant transformation.
FMS-like tyrosine kinase 3 (FLT3) and KIT are both members of the PDGFR family class III receptor tyrosine kinases characterized by an extracellular domain with 5 immunoglobulin-like loops, a transmembrane region and a cytoplasmic domain containing not only the kinase domain (divided in two regions) but also an autoinhibitory juxtamembrane (JM) domain that docks with the kinase domain to stabilize a catalytically inactive conformation. Normally, FLT3 has a crucial role in normal haematopoiesis and its expression is restricted to CD34+ hematopoietic stem/progenitor cells, brain, placenta, and gonads. Activation of FLT3 by FLT3-ligand promotes the normal growth of early progenitor cells.
In acute leukemia, mutations of the FLT3 gene have been found to be one of the most common acquired genetic lesions. FLT3 mutations can be detected in 30% of acute myeloid leukemia (AML) patients (Nakao M, et al. Leukemia. 1996 Dec; 10(12): 1911-8), and also in 5-10% of patients with myelodisplastic syndrome (Horiike S, et al. Leukemia. 1997 Sep; 11(9): 1442-6). There are two frequent types of somatic FLT3 genetic mutations: internal tandem duplications (ITDs) in the JM domain and point mutations in the activation loop of the tyrosine kinase domain (TKD). ITD mutations are any elongation or shortening of the JM domain of FLT3 due to additions or deletions of amino acids that result in the constitutive activation of FLT3. The presence of FLT3/ITD mutations is associated with a poor clinical outcome in both pediatric and adult patients with AML. Point mutations in the activation loop of the kinase domain (FLT3/TKD) involve the aspartic acid, D835 residue, which leads to an activated configuration and transformation of myeloid cells. D835 mutations are missense mutations that result in substitution of tyrosine, histidine, valine, glutamic acid or asparagine for aspartatic acid at amino acid 835 of FLT3. These mutations have been reported in 7% of patients with AML. TKD mutations, unlike ITD mutations, have not been shown to have any prognostic significance in AML patients. Both types of FLT3 mutation cause ligand-independent activation of the receptor and activation of downstream signalling pathways. Mutant FLT3 provides survival advantage to leukemic cells because it causes activation of three major intracellular signalling pathways: PI3K/AKT; RAS/RAF/MAPK and JAK/STAT (Masson K, Ronnstrand L. Cell Signal. 2009 Dec; 21(12): 1717-26).
In conclusion, interfering with the FLT3 signalling likely represents a specific and effective way to block tumor cell proliferation in AML and possibly other indications.
KIT is normally activated by stem cell factor. Signalling by KIT plays an important role in erythropoiesis, lymphopoiesis, mast cell development and function, megakaryopoiesis, gametogenesis and melanogenesis. Hematopoietic stem cells, multipotent progenitors and common myeloid progenitors, but also early T lineage progenitors and thymocytes express high levels of KIT. In addition, mast cells, melanocytes in the skin, and interstitial cells of Cajal in the digestive tract express KIT (Pittoni P. et al. Oncogene 2011 Feb 17; 30(7): 757-69). KIT overexpression or mutations can lead to cancer. Mutations in this gene are frequently associated with gastrointestinal stromal tumors (GIST) (Antonescu CR. J Pathol. 2011 ; 223(2): 251-6). About 65-85% of GISTs have KIT mutations, divided into two categories: mutations of the receptor regulatory domains (extracellular and juxtamembrane) and mutation in the enzymatic domain. At diagnosis, the most frequent mutations, deletions and point mutations, affect JM domain. Extracellular domain mutations are the second most common mutations followed by tyrosine kinase domain mutations. Mutation of KIT have been identified also in melanoma (Curtin JA, JCO, 2006, 24 (26): 4340-4346), acute myeloid leukemia (Malaise M, Steinbach D, Corbacioglu S, Curr Hematol Malig Rep. 2009, 4(2): 77-82), and primary adenoid cystic carcinoma of the salivary gland (Vila L, Liu H, Al-Quran SZ, Coco DP, Dong HJ, Liu C, Mod Pathol. 2009; 22(10): 1296-302). Overexpression is reported also in thymic carcinoma (Strobel P, Hohenberger P, Marx A, J Thorac Oncol. 2010; 5 (10 Suppl 4): S286-90), glioma (Morris PG, Abrey LE.Target Oncol. 2010; 5(3): 193-200), testicular seminoma (Nikolaou M. et al. Anticancer Res. 2007; 27(3B): 1685-8), and small cell lung cancers (SCLC) (Micke P, et al. Clin Cancer Res. 2003; 9(1): 188-94). Additional disorders are linked to KIT activation such as mast cell disease (Lim KH, Pardanani A, Tefferi A. Acta Haematol. 2008;119(4):194-8) or piebaldism (Murakami T, et al. J Dermatol Sci. 2004 Jun;35(1):29-33).
Based on the collection of data, KIT kinase activation appears to be the triggering factor for an important group of malignancies, both hematological and solid cancer diseases, thereby suggesting that it could represent a good therapeutic target for the treatment of these pathologies.
Several indazole derivatives useful for the therapy of a variety of diseases such as cancers, neurodegeneration and atherosclerosis have been disclosed in WO2003028720, WO2005085206, WO2008003396 and WO201069966 in the name respectively of Pharmacia Italia spa, Hoffmann La Roche AG, Merck GMBH and Nerviano Medical Sciences. Despite these developments, there is still a need for more effective agents.
We have now discovered that a series of indazoles are potent protein kinase inhibitors and are thus useful in anticancer therapy.
Accordingly, a first object of the present invention is to provide a substituted indazole compound represented by formula (I),
Figure imgf000004_0001
AM Ar2 Ar3
wherein:
R1 is A, NR6R7, OR8, SOnR9, COR10, nitro, cyano or an optionally substituted group selected from C3-C6 cycloalkyl, heterocyclyl and heteroaryl;
R2, R3, R4 and R5 are independently hydrogen, halogen, nitro, cyano, SOnR9, COR10, NR11 R12, OR13 or an optionally substituted group selected from straight or branched C1-C6 alkyl, straight or branched C2-C6 alkenyl, straight or branched C2-C6 alkynyl, C3-C6 cycloalkyl and heterocyclyl wherein:
A is a straight or branched C1-C6 alkyl substituted with a group selected from an optionally substituted heterocyclyl, an optionally substituted heteroaryl, SOnR9, COR10, NR11 R12 and OR13;
R6 is hydrogen or an optionally substituted group selected from straight or branched Ο-Οβ alkyl, straight or branched C2-C6 alkenyl, straight or branched C2-C6 alkynyl, C3-C6 cycloalkyl, heterocyclyl, aryl and heteroaryl;
R7 is hydrogen, SOnR9, COR10, a substituted straight or branched C1-C6 alkyl or an optionally substituted group selected from straight or branched C2-C6 alkenyl, straight or branched C2-C6 alkynyl, C3-C6 cycloalkyl, heterocyclyl, aryl and heteroaryl or
R6 and R7, taken together with the nitrogen atom to which they are bound, may form an optionally substituted heterocyclyl group;
R8 is hydrogen, A, COR10 or an optionally substituted group selected from straight or branched C2-C6 alkenyl, straight or branched C2-C6 alkynyl, C3-C6 cycloalkyl, heterocyclyl, aryl and heteroaryl, wherein A is as defined above; R9 is NR11 R12 or an optionally substituted group selected from straight or branched Ο-Οβ alkyl, straight or branched C2-C6 alkenyl, straight or branched C2-C6 alkynyl, C3-C6 cycloalkyl, heterocyclyl, aryl and heteroaryl;
R10 is hydrogen, NR11 R12, OR13 or an optionally substituted group selected from straight or branched O- C6 alkyl, straight or branched C2-C6 alkenyl, straight or branched C2-C6 alkynyl, C3-C6 cycloalkyl, heterocyclyl, aryl and heteroaryl;
R11 and R12 are independently hydrogen, SOnR9, COR10 or an optionally substituted group selected from straight or branched Ο-Οβ alkyl, straight or branched C2-C6 alkenyl, straight or branched C2-C6 alkynyl, C3- C6 cycloalkyl, heterocyclyl, aryl and heteroaryl, wherein R9 and R10 are as defined above, or
R11 and R12, taken together with the nitrogen atom to which they are bound, may form an optionally substituted heterocyclyl group;
R13 is hydrogen, COR10 or an optionally substituted group selected from straight or branched Ο-Οβ alkyl, straight or branched C2-C6 alkenyl, straight or branched C2-C6 alkynyl, C3-C6 cycloalkyl, heterocyclyl, aryl and heteroaryl, wherein R10 is as defined above;
n is 0,1 or 2;
X is a bond or an optionally substituted group selected from straight or branched Ο-Οβ alkyl, heterocycly and aryl; Y is a bond, oxygen, or an optionally substituted group selected from straight or branched Ο-Οβ alkyl, straight or branched C2-C6 alkenyl, straight or branched C2-C6 alkynyl, heterocyclyl and aryl;
Z is a bond, oxygen or an optionally substituted straight or branched C1-C6 alkyl;
Ar' is an optionally substituted aryl or an optionally substituted heteroaryl;
or a pharmaceutically acceptable salt thereof.
The present invention also provides methods of synthesizing the substituted indazole derivatives of formula (I) prepared through a process consisting of standard synthetic transformations and isomers, tautomers, hydrates, solvates, complexes, metabolites, prodrugs, carriers, N-oxides.
The present invention also provides a method of treating diseases caused by and/or associated with deregulated protein kinase activity, particularly ABL, ACK1 , AKT1 , ALK, AUR1 , AUR2, BRK, BUB1 , CDC7/DBF4, CDK2/CYCA, CHK1 , CK2, EEF2K, EGFR1 , EphA2, EphB4, ERK2, FAK, FGFR1 , FLT3, GSK3beta, Haspin, IGFR1 , IKK2, IR, JAK1 , JAK2, JAK3, KIT, LCK, LYN, MAPKAPK2, MELK, MET, MNK2, MPS1 , MST4, NEK6, NIM1 , P38alpha, PAK4, PDGFR, PDK1 , PERK, PIM1 , PIM2, PKAalpha, PKCbeta, PLK1 , RET, ROS1 , SULU1 , Syk, TLK2, TRKA, TYK, VEGFR2, VEGFR3 or ZAP70 activity, more particularly FLT3, PDGFR, VEGFR3, TRKA or KIT activity, and further more particularly FLT3 or KIT activity, which comprises administering to a mammal in need thereof an effective amount of a substituted indazole compound represented by formula (I) as defined above.
A preferred method of the present invention is to treat a disease caused by and/or associated with deregulated protein kinase activity selected from the group consisting of cancer, cell proliferation disorders and immune cell- associated diseases and disorders.
Another preferred method of the present invention is to treat specific types of cancer selected from the group consisting of, but not limited to, carcinoma such as bladder, breast, colon, kidney, liver, lung (including small cell lung cancer), salivary gland, esophagus, gall-bladder, ovary, pancreas, stomach, cervix, thyroid, thymus, prostate, and skin, including squamous cell carcinoma; hematopoietic tumors of lymphoid lineage, including leukemia, acute lymphocitic leukemia, acute lymphoblastic leukemia, B-cell lymphoma, T-cell-lymphoma, Hodgkin's lymphoma, non- Hodgkin's lymphoma, hairy cell lymphoma and Burkitt's lymphoma; hematopoietic tumors of myeloid lineage, including acute and chronic myelogenous leukemias, myelodysplastic syndrome and promyelocytic leukemia; tumors of mesenchymal origin, including gastrointestinal stromal tumor, fibrosarcoma and rhabdomyosarcoma; tumors of the central and peripheral nervous system, including astrocytoma neuroblastoma, glioma and schwannomas; other tumors, including mast cell disease, melanoma, seminoma, teratocarcinoma, osteosarcoma, xeroderma pigmentosum, keratoxanthoma, thyroid follicular cancer, Kaposi's sarcoma and mesothelioma and others.
Another preferred method of the present invention is to treat specific cellular proliferation disorders such as, for example, benign prostate hyperplasia, familial adenomatosis polyposis, neurofibromatosis, psoriasis, vascular smooth cell proliferation associated with atherosclerosis, pulmonary fibrosis, arthritis, glomerulonephritis and postsurgical stenosis and restenosis.
Another preferred method of the present invention is to treat immune cell-associated diseases and disorders, such as inflammatory and autoimmune diseases, for examples multiple sclerosis, systemic lupus erythematosis, inflammatory bowel diseases (IBD), Crohn's disease, irritable bowel syndrome, pancreatitis, ulcerative colitis, diverticulosis, myasthenia gravis, vasculitis, psoriasis, scleroderma, asthma, allergy, systemic sclerosis, vitiligo, arthritis such as osteoarthritis, juvenile rheumatoid arthritis, ankylosing spondylitis.
Another preferred method of the present invention is to treat FLT3 mutated cancers, such as acute myeloid leukemia or myelodisplastic syndrome.
Another preferred method of the present invention is to treat KIT mutated cancers, such as gastrointestinal stromal tumors, melanoma, acute myeloid leukemia, primary adenoid cystic carcinoma of the salivary gland, thymic carcinoma, glioma, testicular seminoma, small cell lung cancers, mast cell disease or piebaldism.
In addition, the method of the present invention also provides tumor angiogenesis and metastasis inhibition.
In a further preferred embodiment, the method of the present invention further comprises subjecting the mammal in need thereof to a radiation therapy or chemotherapy regimen in combination with at least one cytostatic or cytotoxic agent.
Moreover the invention provides an in vitro method for inhibiting FLT3 or KIT protein kinase activity which comprises contacting the said protein with an effective amount of a compound of formula (I).
The present invention also provides a pharmaceutical composition comprising a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof and at least one pharmaceutically acceptable excipient, carrier and/or diluent.
The present invention further provides a pharmaceutical composition comprising a compound of formula (I) in combination with one or more chemotherapeutic - e.g. cytostatic or cytotoxic - agents, antibiotic-type agents, alkylating agents, antimetabolite agents, hormonal agents, immunological agents, interferon-type agents, cyclooxygenase inhibitors (e.g. COX-2 inhibitors), matrixmetalloprotease inhibitors, telomerase inhibitors, tyrosine kinase inhibitors, anti-growth factor receptor agents, anti-HER agents, anti-EGFR agents, anti-angiogenesis agents (e.g. angiogenesis inhibitors), farnesyl transferase inhibitors, ras-raf signal transduction pathway inhibitors, cell cycle inhibitors, other cdks inhibitors, tubulin binding agents, topoisomerase I inhibitors, topoisomerase II inhibitors, and the like.
Additionally, the invention provides a product comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof, as defined above, and one or more chemotherapeutic agents, as a combined preparation for simultaneous, separate or sequential use in anticancer therapy.
In yet another aspect the invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof, as defined above, for use as a medicament.
Moreover, the invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof, as defined above, for use in a method of treating cancer.
Finally, the invention provides the use of a compound of formula (I) or a pharmaceutically acceptable salt thereof, as defined above, in the manufacture of a medicament with antitumor activity.
The compounds of formula (I) may have one or more asymmetric centres, and may therefore exist as individual optical isomers or racemic mixtures. Accordingly, all the possible isomers, and their mixtures, of the compounds of formula (I) are within the scope of the present invention.
In cases in which the compounds of formula (I) have unsaturated carbon-carbon double bonds, both the cis (Z) and trans (E) isomers are within the scope of this invention.
Derivatives of compounds of formula (I) originating from metabolism in a mammal, and the pharmaceutically acceptable bio-precursors (otherwise referred to as pro-drugs) of the compounds of formula (I) are also within the scope of the present invention.
In addition to the above, as known to those skilled in the art, the unsubstituted nitrogen on the pyrazole ring of the compounds of formula (I) rapidly equilibrates in solution to form a mixture of tautomers, as depicted below:
Figure imgf000007_0001
(I) (la)
wherein Ar, Ar', X, Y and Z are as defined above.
Accordingly, in the present invention, where only one tautomer is indicated for the compounds of formula (I), the other tautomer (la) is also within the scope of the present invention, unless specifically noted otherwise.
Moreover, if easily obtainable from the compounds of formula (I) as defined above, also their hydrates, solvates, complexes and N-oxides are within the scope of the present invention.
N-oxides are compounds of formula (I) wherein nitrogen and oxygen are tethered through a dative bond.
The general terms as used herein, unless otherwise specified, have the meaning reported below.
The term "straight or branched Ο-Οβ alkyl" refers to a saturated aliphatic hydrocarbon radical, including straight chain and branched chain groups of from 1 to 6 carbon atoms, e.g. methyl, ethyl, propyl, 2-propyl, n-butyl, iso-butyl, tert- butyl, pentyl and the like. The alkyl group may be substituted or unsubstituted; when not otherwise specified, the substituent groups are preferably one to three, independently selected from the group consisting of halogen, cyano, nitro, SOnR9, COR10, NR11 R12, OR13, R11 R12N-(Ci-C6)-alkyl, R130-(Ci-C6)-alkyl and an optionally further substituted group selected from C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, heterocyclyl, aryl and heteroaryl, wherein R9, R10, R11 , R12, R13 and n are as defined above.
The term "C3-C6 cycloalkyl" refers to a 3- to 6-membered all-carbon monocyclic ring, which may contain one or more double bonds but does not have a completely conjugated ττ-electron system. Examples of cycloalkyl groups, without limitation, are cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl and cyclohexadienyl. A cycloalkyl group may be substituted or unsubstituted; when not otherwise specified, the substituent groups are preferably one to three, independently selected from the group consisting of halogen, cyano, nitro, SOnR9, COR10, NR11 R12, OR13, R11 R12N-(Ci-C6)-alkyl, R130-(Ci-C6)-alkyl and an optionally further substituted straight or branched C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, heterocyclyl, aryl and heteroaryl, wherein R9, R10, R11 , R12, R13 and n are as defined above.
The term "heterocyclyl" refers to a 3- to 7-membered, saturated or partially unsaturated carbocyclic ring where one or more carbon atoms are replaced by heteroatoms such as nitrogen, oxygen and sulfur. Not limiting examples of heterocyclyl groups are, for instance, oxiranyl, aziridinyl, oxetanyl, azetidinyl, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothiophenyl, dihydrothiophenyl, pyrrolidinyl, dihydropyrrolyl, pyranyl, dihydropyranyl, tetrahydropyranyl, tetrahydrothiopyranyl, piperidinyl, pyrazolinyl, isoxazolidinyl, isoxazolinyl, thiazolidinyl, thiazolinyl, isothiazolinyl, dioxanyl, piperazinyl, morpholinyl, thiomorpholinyl, examethyleneiminyl, homopiperazinyl and the like. A heterocyclyl group may be substituted or unsubstituted; when not otherwise specified, the substituent groups are preferably one to three, independently selected from the group consisting of halogen, cyano, nitro, SOnR9, COR10, NR11 R12, OR13, R11 R12N-(Ci-C6)-alkyl, R130-(Ci-C6)-alkyl and an optionally further substituted straight or branched Ci-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, heterocyclyl, aryl and heteroaryl, wherein R9, R10, R11 , R12, R13 and n are as defined above.
The term "aryl" refers to a mono-, bi- or poly-carbocyclic hydrocarbon with from 1 to 4 ring systems, optionally further fused or linked to each other by single bonds, wherein at least one of the carbocyclic rings is "aromatic", wherein the term "aromatic" refers to completely conjugated ττ-electron bond system. Non limiting examples of such aryl groups are phenyl, a- or β-naphthyl or biphenyl groups.
The term "heteroaryl" refers to aromatic heterocyclic rings, typically 5- to 7-membered heterocycles with from 1 to 3 heteroatoms selected among N, 0 and S; the heteroaryl ring can be optionally further fused or linked to aromatic and non-aromatic carbocyclic and heterocyclic rings. Not limiting examples of such heteroaryl groups are, for instance, pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, indolyl, imidazolyl, thiazolyl, isothiazolyl, pyrrolyl, phenyl-pyrrolyl, furyl, phenyl-furyl, oxazolyl, isoxazolyl, pyrazolyl, thienyl, benzothienyl, isoindolinyl, benzoimidazolyl, quinolinyl, isoquinolinyl, 1 ,2,3-triazolyl, 1 -phenyl-1 ,2,3-triazolyl, 2,3-dihydroindolyl, 2,3-dihydrobenzofuranyl, 2,3- dihydrobenzothiophenyl; benzopyranyl, 2,3-dihydrobenzoxazinyl, 2,3-dihydroquinoxalinyl and the like.
The aryl and heteroaryl groups may be substituted or unsubstituted; when not otherwise specified, the substituent groups are preferably one to three, independently selected from the group consisting of halogen, cyano, nitro, S0nR9, COR10, NR11R12, 0R13, R11 R12N-(Ci-C6)-alkyl, R130-(Ci-C6)-alkyl and an optionally further substituted straight or branched C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, heterocyclyl, aryl and heteroaryl, wherein R9, R10, R11 , R12, R13 and n are as defined above.
The term "halogen" indicates fluorine, chlorine, bromine or iodine.
The term "C2-C6 alkenyl" indicates an aliphatic C2-C6 hydrocarbon chain containing at least one carbon-carbon double bond and which can be straight or branched. Representative examples include, but are not limited to, ethenyl, 1-propenyl, 2-propenyl, 1- or 2-butenyl, and the like.
The term " C2-C6 alkynyl" indicates an aliphatic C2-C6 hydrocarbon chain containing at least one carbon-carbon triple bond and which can be straight or branched. Representative examples include, but are not limited to, ethynyl, 1- propynyl, 2-propynyl, 1- or 2-butynyl, and the like.
The alkenyl and alkynyl groups may be substituted or unsubstituted; when not otherwise specified, the substituent groups are preferably one to three, independently selected from the group consisting of halogen, cyano, nitro, SOnR9, COR10, NR11R12, OR13, R11 R12N-(Ci-C6)-alkyl, R130-(Ci-C6)-alkyl and an optionally further substituted straight or branched C1-C6 alkyl, C3-C6 cycloalkyl, heterocyclyl, aryl and heteroaryl, wherein R9, R10, R11 , R12, R13 and n are as defined above.
The term "nitro" indicates a -NO2 group.
The term "cyano" indicates a -CN residue.
The term "pharmaceutically acceptable salt" of compounds of formula (I) refers to those salts that retain the biological effectiveness and properties of the parent compound. Such salts include acid addition salts with inorganic acids such as hydrochloric, hydrobromic, nitric, phosphoric, sulfuric, perchloric acid and the like, or with organic acids such as acetic, trifluoroacetic, propionic, glycolic, lactic, (D) or (L) malic, maleic, fumaric, methanesulfonic, ethanesulfonic, benzoic, p-toluenesulfonic, salicylic, cinnamic, mandelic, tartaric, citric, succinic, malonic acid and the like; salts formed when an acidic proton present in a compound of formula (I) is either replaced by a metal ion, - e.g. an alkali metal ion such as sodium or potassium - or an alkaline earth ion, such as calcium or magnesium, or coordinates with an organic base such as ethanolamine, diethanolamine, triethanolamine, tromethamine, N-methylglucamine, and the like.
A preferred class of compounds of formula (I) are the compounds wherein:
R1 is A, NR6R7, OR8 or an optionally substituted heterocyclyl, wherein A, R6, R7 and R8 are as defined above. A more preferred class of compounds of formula (I) are the compounds wherein:
Ar is AM or Ar2; and R2, R3, R4, R5 are each independently hydrogen, halogen, NR11 R12 or OR13, wherein R11 , R12 and R13 are as defined above.
Specific compounds (Cpd.) of the invention or a salt thereof are listed below:
N-(6-Benzyloxy-1 H-indazol-3-yl)-4-(4-methyl-piperazin-1-yl)-benzamide,
2 4-(4-Methyl-piperazin-1-yl)-N-(6-phenoxy-1 H-indazol-3-yl)-benzamide,
3 N-[6-(3-Fluoro-phenoxy)-1 H-indazol-3-yl]-4-(4-methyl-piperazin-1-yl)-benzamide,
4, N-[6-(4-Benzyloxy-phenoxy)-1 H-indazol-3-yl]-4-(4-methyl-piperazin-1-yl)-benzamide,
5. 4-(4-Methyl-piperazin-1-yl)-N-[6-(3-phenoxy-benzyloxy)-1 H-indazol-3-yl]-benzamide, N-[6-(1-Benzyl-piperidin-4-yloxy)-1 H-indazol-3-yl]-4-(4-methyl-piperazin-1-yl)-benzamide,
4-(4-Methyl-piperazin-1-yl)-N-[6-(3-phenyl-prop-2-ynyloxy)-1 H-indazol-3-yl]-benzamide,
4-(4-Methyl-piperazin-1-yl)-N-[6-(4-phenoxy-phenoxy)-1 H-indazol-3-yl]-benzamide,
N-[6-(3-Benzyloxy-phenoxy)-1 H-indazol-3-yl]-4-(4-methyl-piperazin-1-yl)-benzamide,
4-(4-Methyl-piperazin-1-yl)-N-[6-(2-phenoxy-ethoxy)-1 H-indazol-3-yl]-benzamide,
N-[6-(2-Benzyloxy-ethoxy)-1 H-indazol-3-yl] -(4-methyl-piperazin-1-yl)-benzamide,
N-[6-(1-Benzyl-piperidin-3-yloxy)-1 H-indazol-3-yl]-4-(4-methyl-piperazin-1-yl)-benzamide,
N-[6-(1-Benzyl-pyrrolidin-2-ylmethoxy)-1 H-indazol-3-yl]-4-(4-methyl-piperazin-1-yl)-benzamide, N-[6-(2-Benzyloxy-ethoxy)-1 H-indazol-3-yl] -(4-methyl-4-oxy-piperazin-1-yl)-benzamide,
N-[6-(2-Benzyloxy-ethoxy)-1 H-indazol-3-yl]-4-(4-dimethylamino-piperidin-1-yl)-benzamide,
N-[6-(2-Benzyloxy-ethoxy)-1 H-indazol-3-yl]-4-[(2-dimethylamino-ethyl)-methyl-amino]-ben
N-[6-(2-Benzyloxy-ethoxy)-1 H-indazol-3-yl]-4-[(3-dimethylamino-propyl)-methyl-amino]-benzam 4-{4-[6-(2-Benzyloxy-ethoxy)-1 H-indazol-3-ylcarbamoyl]-phenyl}-piperazine-1-carboxylic acid tert-butyl ester,
N-[6-(2-Benzyloxy-ethoxy)-1 H-indazol-3-yl]-4-(1-methyl-piperidin-4-ylamino)-benzamide,
N-[6-(2-Benzyloxy-ethoxy)-1 H-indazol-3-yl]-4-piperazin-1-yl-benzamide,
N-[6-(2-Benzyloxy-ethoxy)-1 H-indazol-3-yl]-4-dimethylaminomethyl-benzamide,
N-[6-(2-Benzyloxy-ethoxy)-1 H-indazol-3-yl]-4-(1-methyl-piperidin-4-yloxy)-benzamide,
N-[6-(2-Benzyloxy-ethoxy)-1H-indazol-3-yl] -[methyl-(1-methyl-piperidin -yl)-amino]-benza
N-[6-(2-Benzyloxy-ethoxy)-1 H-indazol-3-yl]4-morpholin4-yl-benzamide,
N-[6-(2-Benzyloxy-ethoxy)-1 H-indazol-3-yl]4-(2-morpholin4-yl-ethylamino)-benzamide,
N-[6-(2-Benzyloxy-ethoxy)-1 H-indazol-3-yl]4-(tetrahydro-pyran4-ylamino)-benzamide,
N-[6-(2-Benzyloxy-ethoxy)-1 H-indazol-3-yl]4-[(1-methyl-piperidin4-ylmethyl)-amino]-b^
N-[6-(2-Benzyloxy-ethoxy)-1 H-indazol-3-yl]4-(3-pyrrolidin-1-yl-azetidin-1-yl)-benzamide,
N-[6-(2-Benzyloxy-ethoxy)-1 H-indazol-3-yl]-3-(4-methyl-piperazin-1-yl)-benzamide,
N-{6-[2-(2-Fluoro-benzyloxy)-ethoxy]-1 H-indazol-3-yl}-4-(4-methyl-piperazin-1-yl)-benzamide, N-{6-[2-(3-Fluoro-benzyloxy)-ethoxy]-1 H-indazol-3-yl}-4-(4-methyl-piperazin-1-yl)-benzamide, N-{6-[2-(4-Fluoro-benzyloxy)-ethoxy]-1 H-indazol-3-yl}-4-(4-methyl-piperazin-1-yl)-benzamide, 4-(4-Methyl-piperazin-1-yl)-N-{6-[2-(4-trifluoromethyl-benzyloxy)-ethoxy]-1 H-indazol-3-yl}-benzamide, 4-(4-Methyl-piperazin-1-yl)-N-{6-[2-(3-trifluoromethyl-benzyloxy)-ethoxy]-1 H-indazol-3-yl}-benzamide, 4-(4-Methyl-piperazin-1-yl)-N-{6-[2-(pyridin-4-ylmethoxy)-ethoxy]-1 H-indazol-3-yl}-benzam
4-(4- ethyl-piperazin-1-yl)-N-{6-[2-(pyridin-3-ylmethoxy)-ethoxy]-1H-indazol-3-yl}-benzam
4-(4-Methyl-piperazin-1-yl)-N-{6-[2-(pyridin-2-ylmethoxy)-ethoxy]-1 H-indazol-3-yl}-benzami
N-[6-(2-Benzyloxy-ethoxy)-1 H-indazol-3-yl]-2-fluoro4-(4-methyl-piperazin-1-yl)-benzamide,
4-(4-Methyl-piperazin-1-yl)-N-[6-((E)-3-phenyl-allyloxy)-1 H-indazol-3-yl]-benzamide,
N-{6-[2-(4-Methoxy-benzyloxy)-ethoxy]-1 H-indazol-3-yl}-4-(4-methyl-piperazin-1-yl)-benzamide, N-[6-(2-Benzyloxy-ethoxy)-1 H-indazol-3-yl]4-[methyl-(1-methyl-1-oxy-piperidin4-yl)-am 42. 4-(4-Methyl-4-oxy-piperazin-1-yl)-N-{6-[2-(4-trifluoromethyl-benzyloxy)-ethoxy]-1 H-indazol-3-yl}-benzamide and
43. N-[6-(2-Benzyloxy-ethoxy)-1 H-indazol-3-yl]-2,4-bis-(4-methyl-piperazin-1-yl)-benzamide.
The present invention also provides a process for the preparation of a compound of formula (I) as defined above, by using the reaction routes and synthetic schemes described below, employing the techniques available in the art and starting materials readily available. The preparation of certain embodiments of the present invention is described in the examples that follow, but those of ordinary skill in the art will recognize that the preparations described may be readily adapted to prepare other embodiments of the present invention. For example, the synthesis of non- exemplified compounds according to the invention may be performed by modifications apparent to those skilled in the art, for instance by appropriately protecting interfering groups, by changing to other suitable reagents known in the art, or by making routine modifications of reaction conditions. Alternatively other reactions referred to herein or known in the art will be recognized as having adaptability for preparing other compounds of the invention.
The reported Scheme 1 and Scheme 2 show the preparations of a compound of formula (I).
Figure imgf000011_0001
Scheme 1
Figure imgf000012_0001
wherein Ar is as defined in formula (I); W is hydroxy, halogen or a suitable leaving group; X, Y, Z and Ar' are as defined in formula (I); and L is a suitable leaving group, such as halogen, methanesulfonyloxy, trifluoromethanesulfonyloxy or p-toluenesulfonyloxy. Scheme 2
Figure imgf000013_0001
(XVIII) (I)
wherein Ar is as defined in formula (I); W is hydroxy, halogen or a suitable leaving group; X, Y, Z and Ar' are as defined in formula (I); and L is a suitable leaving group, such as halogen, methanesulfonyloxy, trifl uoromethanesulfonyloxy or p-toluenesulfonyloxy.
All those with ordinary skills in the art will appreciate that any transformation performed according to said methods may require standard modifications such as, for instance, protection of interfering groups, change to other suitable reagents known in the art, or make routine modifications of reaction conditions.
Accordingly, a process of the present invention comprises the following steps:
A) introducing the tert-butoxy-carbonyl roup into the compound of formula (II)
Figure imgf000013_0002
B) cleaving the phthalimido group of the resultant compound of formula
Figure imgf000014_0001
C) acylating the resultant compound of formula (IV)
Figure imgf000014_0002
by reaction with a compound of formula (V)
Figure imgf000014_0003
wherein Ar is as defined in formula (I) and W is hydroxy, halogen or a suitable leaving group; D) selectively cleaving the tert-butyldim resultant compound of formula (VI)
Figure imgf000014_0004
wherein Ar is as defined in formula (I);
E) coupling the resultant compound of formula (VII)
Figure imgf000014_0005
wherein Ar is as defined in formula (I), alternatively with:
Ea) a compound of formula (VIII)
Figure imgf000015_0001
wherein Ar', Z and Y are as defined in formula (I) and X is an optionally substituted group selected from straight or branched Ο-0β alkyl and heterocyclyl;
or
Eb) a compound of formula (IX)
Ar'/Z "X^B(OH)2 (IX)
wherein Ar', Z and Y are as defined in formula (I) and X is an optionally substituted aryl or
wherein Ar' is as defined in formula (I) and X, Y and Z are a bond;
or
Ec) a compound of formula (X)
Figure imgf000015_0002
wherein Ar', Z and Y are as defined in formula (I), X is an optionally substituted group selected from straight or branched C1-C6 alkyl or heterocyclyl and L is a suitable leaving group, such as halogen, methanesulfonyloxy, trifl uoromethanesulfonyloxy or p-toluenesulfonyloxy;
F) cleaving the tert-butoxy-carbonyl group of the resultant compound of formula (XI) obtained in step Ea), Eb) or Ec)
Figure imgf000015_0003
wherein Ar, Ar', X, Y and Z are as defined in formula (I), so as to obtain a compound of formula (I), as defined above; optionally separating the resultant compound of formula (I) into the single isomers; optionally converting the resultant compound of formula (I) into a different compound of formula (I), and/or into a pharmaceutically acceptable salt if desired.
Alternatively, the intermediate compound of formula (XI), wherein Ar, Ar', Y and Z are as defined in formula (I) and X is an optionally substituted group selected from straight or branched Ο-Οβ alkyl and heterocyclyl, can be obtained in a process comprising the following steps:
G) selectively cleaving the tert-butyldimethylsilyl ether of the compound of formula (IV), as defined above;
H) coupling the resultant compound of formula (XII)
Figure imgf000016_0001
alternatively with:
Ha) a compound of formula (VIII), as defined above;
or
Hb) a compound of formula (X), as defined above;
I) acylating the resultant compound of formula (XIII)
Figure imgf000016_0002
wherein Ar, Ar', Y and Z are as defined in formula (I) and X is an optionally substituted group selected from straight or branched C1-C6 alkyl and heterocyclyl, with a compound of formula (V), as defined above, so as to obtain a compound of formula (XI), wherein Ar, Ar', Y and Z are as defined in formula (I) and X is an optionally substituted group selected from straight or branched C1-C6 alkyl and heterocyclyl.
It is a further object of the present invention a process for preparing the compound of formula (I), as defined above, comprising the following steps:
J) coupling the compound of formula (XIV)
Figure imgf000016_0003
alternatively with:
Ja) a compound of formula (VIII), as defined above;
or
Jb) a compound of formula (IX), as defined above;
or
Jc) a compound of formula (X), as defined above;
K) converting the resultant compound of formula (XV)
Figure imgf000016_0004
wherein Ar', X, Y and Z are as defined in formula (I); L) acylating the resultant compound of formula (XVI)
Figure imgf000017_0001
wherein Ar', X, Y and Z are as defined in formula (I), with a compound of formula (V), as defined above, so as to obtain a compound of formula (I), as defined above; optionally separating the resultant compound of formula (I) into the single isomers; optionally converting the resultant compound of formula (I) into a different compound of formula (I), and/or into a pharmaceutically acceptable salt if desired.
It is a further object of the present invention a process for preparing a compound of formula (I), wherein R1 is NR6R7, wherein R6 is as defined above and R7 is hydrogen, a substituted straight or branched C1-C6 alkyl or an optionally substituted group selected from straight or branched C2-C6 alkenyl, straight or branched C2-C6 alkynyl, C3- C6 cycloalkyi, heterocyclyl, aryl and heteroaryl, and wherein Ar, Ar', X, Y and Z are as defined above, comprising the following steps:
M) acylating the compound of formula (XVI), as defined above, with a compound of formula (XVII)
Figure imgf000017_0002
wherein R2, R3, R4 and R5 are as defined in formula (I) and W and L are as defined above;
N) coupling the resultant compound of formula (XVIII)
Figure imgf000017_0003
wherein Ar', X, Y, Z, R2, R3, R4 and R5 are as defined in formula (I) and L is as defined above, with a compound of formula (XIX)
N (XIX) wherein R6 is as defined in formula (I) and R7 is hydrogen, a substituted straight or branched C1-C6 alkyl or an optionally substituted group selected from straight or branched C2-C6 alkenyl, straight or branched C2-C6 alkynyl, C3- C6 cycloalkyi, heterocyclyl, aryl and heteroaryl, so as to obtain a compound of formula (I), wherein R1 is NR6R7, wherein R6 is as defined in formula (I) and R7 is hydrogen, a substituted straight or branched C1-C6 alkyl or an optionally substituted group selected from straight or branched C2-C6 alkenyl, straight or branched C2-C6 alkynyl, C3- C6 cycloalkyi, heterocyclyl, aryl and heteroaryl, and Ar, Ar', X, Y and Z are as defined in formula (I); optionally separating the resultant compound of formula (I) into the single isomers; optionally converting the resultant compound of formula (I) into a different compound of formula (I), and/or into a pharmaceutically acceptable salt if desired.
As said above, the compounds of formula (I) which are prepared according to the process object of the invention, can be conveniently converted into other compounds of formula (I) by operating according to well-known synthetic conditions, the following being examples of possible conversions:
1) reducing a compound of formula (I) wherein one of the substituents R1 , R2, R3, R4 or R5 is NO2, for obtaining the corresponding compound of formula (I) wherein such substituent is NH2;
2) acylating a compound of formula (I) wherein one of the substituents R1 , R2, R3, R4 or R5 is NH2, by reaction with an acylating agent of formula (XX) or (XXI)
11
R9 W R10 W
(XX) (XXI)
wherein R9, R10 and W are as defined above, for obtaining the corresponding compound of formula (I) wherein such substituent is a NHSO2R9 or NHCOR10 residue, wherein R9 and R10 are as defined above;
3) reacting a compound of formula (I) wherein the substituent R1 is NH2, with a suitable aldehyde or ketone in the presence of a reducing agent, for obtaining the corresponding compound of formula (I) wherein such substituent is a
NR6R7 group, wherein R7 is hydrogen and R6 is as defined in formula (I) except hydrogen;
4) reacting a compound of formula (I) wherein one of the substituents R2, R3, R4 or R5 is NH2, with a suitable aldehyde or ketone in the presence of a reducing agent, for obtaining the corresponding compound of formula (I) wherein such substituent is a NR11 R12 group, wherein one of the R11 or R12 is hydrogen and the other is as defined in formula (I) except hydrogen, SOnR9 or C0R10;
5) hydrolysing a compound of formula (I) wherein one of the substituents R1 , R2, R3, R4 or R5 is a COOR13 residue, wherein R13 is a straight or branched Ο-Οβ alkyl, under acid or basic catalysis, so as to obtain the corresponding compound of formula (I) wherein such substituent is a COOH group, in which case R13 represents hydrogen;
6) amidating a compound of formula (I) wherein one of the substituents R1 , R2, R3, R4 or R5 is a COOH residue, with an amine of formula (XXII)
H
R11 R12
(XXII)
wherein R11 and R12 are as defined in formula (I) except SOnR9 or COR10, for obtaining the corresponding compound of formula (I) wherein such substituent is a CONR11 R12 residue, wherein R11 and R12 are as defined in formula (I) except SOnR9 or COR10;
7) oxidazing a compound of formula (I) wherein R1 is 4-methyl-piperazin-1-yl for obtaining the corresponding compound of formula (I) wherein such substituent is 4-methyl-4-oxy-piperazin-1-yl;
8) cleaving the tert-butoxy-carbonyl group of a compound of formula (I) wherein R1 is 4-tert-butoxycarbonyl- piperazin-1-yl for obtaining the corresponding compound of formula (I) wherein such substituent is piperazin-1-yl. According to step A), the transformation of the compound of formula (II) into the compound of formula (III) can be accomplished in a variety of ways and experimental conditions, which are widely known in the art for the introduction of the tert-butoxy-carbonyl group, for example using di-tert-butyl dicarbonate. Preferably, this reaction is carried out in a suitable solvent such as, for instance, tetrahydrofuran, dichloromethane, toluene, 1 ,4-dioxane, and in the presence of a proton scavenger such as, for example, pyridine, triethylamine, Ν,Ν-diisopropylethylamine, at a temperature ranging from room temperature to reflux, for a time ranging from about 30 min. to about 96 hours.
According to step B), the cleavage of the phthalimido group of the compound of formula (III) to give the compound of formula (IV) can be accomplished in a variety of ways and experimental conditions, which are widely known in the art, for example using hydrazine. Preferably, this reaction is carried out in a suitable solvent such as, for instance, tetrahydrofuran, dichloromethane, toluene, 1 ,4-dioxane, at a temperature ranging from room temperature to reflux, for a time ranging from about 30 min. to about 96 hours.
According to step C), a compound of formula (VI) can be obtained by reacting a compound of formula (IV) with a compound of formula (V) in a variety of ways and experimental conditions, which are widely known in the art for acylation reactions. Preferably a compound of formula (V) wherein W is hydroxy is converted into its corresponding acyl chloride wherein W is chlorine in the presence of thionyl chloride or oxalyl chloride, in a suitable solvent, such as toluene, dichloromethane, chloroform, diethyl ether, tetrahydrofuran, 1 ,4-dioxane, or a mixture thereof, at a temperature ranging from about -10°C to reflux and for a period of time varying from about 1 hour to about 96 hours. The acyl chloride is isolated by evaporation of the solvent and further reacted with (IV) in the presence of a base such as pyridine, triethylamine or Ν,Ν-diisopropylethylamine, at a temperature ranging from about -40°C to reflux and for a period of time varying from about 1 hour to about 96 hours. A suitable solvent may also be added, such as toluene, dichloromethane, chloroform, diethyl ether, tetrahydrofuran, 1 ,4-dioxane. Alternatively, a compound of formula (IV) is reacted with a compound of formula (V) wherein W is hydroxy in the presence of an activating agent such as hydroxybenzotriazole, dicyclohexyl carbodiimide, diisopropyl carbodiimide, 1-ethyl-3-(3'- dimethylamino)carbodiimide hydrochloric acid salt, 0-(benzotriazol-1-yl)-N,N,N',N'-tetramethyluronium tetrafluoroborate. Preferably, this reaction is carried out in a suitable solvent such as, for instance, tetrahydrofuran, dichloromethane, toluene, 1 ,4-dioxane, N,N-dimethylformamide, Ν,Ν-dimethylacetamide and in the presence of a proton scavenger such as, for example, pyridine, triethylamine, Ν,Ν-diisopropylethylamine, at a temperature ranging from room temperature to reflux, for a time ranging from about 30 min. to about 96 hours.
According to step D), the selective cleavage of the tert-butyldimethylsilyl ether of the compound of formula (VI) to give the compound of formula (VII) can be carried out in a variety of ways, according to conventional methods well known in the literature. Preferably this conversion is carried out in the presence of tetrabutylammonium fluoride in a suitable solvent, such as, for instance, tetrahydrofuran, dichloromethane, toluene, 1 ,4-dioxane, at a temperature ranging from -10°C to reflux, for a time ranging from about 30 min. to about 96 hours.
According to step Ea), the coupling of a compound of formula (VII) with an alcohol of formula (VIII) to give a compound of formula (XI) can be accomplished in a variety of ways and experimental conditions which are widely known in the art for the synthesis of aryl ethers under Mitsunobu-like conditions. Preferably this conversion is carried out in the presence of an azodicarboxylate, such as, for example, diethyl azodicarboxylate, diisopropyl azodicarboxylate or di-tert-butyl azodicarboxylate and a phosphine, such as, for example, triphenylphosphine or polymer-bound triphenylphosphine, in a suitable solvent, such as, for instance, tetrahydrofuran, dichloromethane, 1 ,4-dioxane, toluene, acetonitrile at a temperature ranging from -20°C to reflux, for a time ranging from about 30 min. to about 96 hours.
According to step Eb), the coupling of a compound of formula (VII) with a boronic acid of formula (IX) to give a compound of formula (XI) can be accomplished in a variety of ways and experimental conditions which are widely known in the art for the synthesis of di-aryl ethers. Preferably this conversion is carried out in the presence of copper diacetate and 4A molecular sieve or silica gel, in a suitable solvent, such as, for instance, tetrahydrofuran, dichloromethane, 1 ,4-dioxane and in the presence of a proton scavenger such as, for example, pyridine, triethylamine, Ν,Ν-diisopropylethylamine, at a temperature ranging from -10°C to reflux, for a time ranging from about 30 min. to about 96 hours.
According to step Ec), the coupling of a compound of formula (VII) with a compound of formula (X) to give a compound of formula (XI) can be accomplished in a variety of ways and experimental conditions which are widely known in the art for the alkylation of phenols. Preferably a compound of formula (VII) is treated with a chloride, bromide, iodide, mesylate or triflate of formula (X), in which case L represents chlorine, bromine, iodine, methanesulfonyloxy or trifluoromethanesulfonyloxy, respectively, in the presence of a proton scavenger such as, for example, triethylamine, Ν,Ν-diisopropylethylamine, sodium, potassium or cesium carbonate, in a suitable solvent such as, for instance, tetrahydrofuran, 1 ,4-dioxane, Ν,Ν-dimethylformamide, N,N-dimethylacetamide, dimethoxyethane, at a temperature ranging from -10°C to reflux, for a time ranging from about 30 min. to about 96 hours.
According to step F), the transformation of a compound of formula (XI) into a compound of formula (I) can be carried out in a variety of ways, according to conventional methods well known in the literature for the cleavage of a tert- butoxy-carbonyl group. As an example, this reaction may be run under acidic conditions, for example in the presence of an inorganic or organic acid such as hydrochloric, trifluoroacetic or methanesulfonic acid, in a suitable solvent such as dichloromethane, 1 ,4-dioxane, a lower alcohol, such as methanol or ethanol, water, or a mixture thereof, at a temperature ranging from room temperature to reflux and for a period of time ranging from about 30 min. to about 96 hours.
According to step G), the transformation of the compound of formula (IV) into the compound of formula (XII) can be carried out in a way analogous to that specified above under D).
According to step Ha), the coupling between the compound of formula (XII) and an alcohol of formula (VIII) can be carried out in a way analogous to that specified above under Ea).
According to step Hb), the coupling between the compound of formula (XII) and a compound of formula (X) can be carried out in a way analogous to that specified above under Ec).
According to step I), the acylation of the compound of formula (XIII) with a compound of formula (V) can be carried out in a way analogous to that specified above under C).
According to step Ja), the coupling between the compound of formula (XIV) and an alcohol of formula (VIII) can be carried out in a way analogous to that specified above under Ea). According to step Jb), the coupling between the compound of formula (XIV) and a boronic acid of formula (IX) can be carried out in a way analogous to that specified above under Eb).
According to step Jc), the coupling between the compound of formula (XIV) and a compound of formula (X) can be carried out in a way analogous to that specified above under Ec).
According to step K), a compound of formula (XV) can be transformed into a compound of formula (XVI) in a variety of ways and experimental conditions. Preferably, this reaction is carried out in the presence of hydrazine or hydrazine monohydrate in a suitable solvent such as, for instance, toluene, tetrahydrofuran, 1 ,4-dioxane, dimethyl sulfoxide, acetonitrile, methanol, ethanol or n-butanol, at a temperature ranging from 0 °C to reflux and for a period of time varying from about 30 min to about 96 hours.
According to step L), the acylation of the compound of formula (XVI) with a compound of formula (V) can be carried out in a way analogous to that specified above under C).
According to step M), the acylation of the compound of formula (XVI) with a compound of formula (XVII) can be carried out in a way analogous to that specified above under C).
According to step N), the coupling of a compound of formula (XVIII) with an amine of formula (XIX) can be carried out in a variety of ways, according to conventional methods well known in the literature for Buchwald-Hartwig aminations. Preferably a compound of formula (XVIII) wherein L is chlorine, bromine, iodine or trifluoromethanesulfonyloxy is reacted with a compound of formula (XIX) in a suitable solvent such as, for example, tetrahydrofuran, 1 ,4-dioxane, N,N-dimethylformamide, Ν,Ν-dimethylacetamide, dimethoxyethane, acetonitrile, toluene, in the presence of catalytic amounts of a palladium derivative, such as, for example, tris(dibenzylideneacetone)dipalladium(0), palladium diacetate, and a phopsphine ligand, such as, for example, 2,2'-bis(diphenylphosphino)-1 ,1'-binaphthyl, 4,5- bis(diphenylphosphino)-9,9-dimethylxanthene, 2-dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl, 2- dicyclohexylphosphino-2'-(N,N-dimethylamino)biphenyl, in the presence of a base, such as, for instance, sodium or lithium bis(trimethylsilyl)amide, sodium or potassium tert-butoxide, sodium, potassium or cesium carbonate, at a temperature ranging from 0 °C to reflux and for a period of time varying from about 15 min to about 96 hours.
According to the conversion 1), the reduction of a compound of formula (I) wherein one of the substituents R1 , R2, R3, R4 or R5 is nitro, for obtaining a compound of formula (I) wherein such substituent is amino, can be carried out in a variety of ways, according to conventional methods well known in the literature. Preferably this conversion is carried out in a suitable solvent such as, for instance, methanol, ethanol, water, tetrahydrofuran, 1 ,4-dioxane, N,N- dimethylformamide, acetic acid, or a mixture thereof, in the presence of a suitable reducing agent, such as, for instance, hydrogen and a hydrogenation catalyst, or by treatment with cyclohexene or cyclohexadiene, or formic acid or ammonium formate and a hydrogenation catalyst, or a metal such as iron or zinc in the presence of an inorganic acid, such as hydrochloric acid, or by treatment with tin (II) chloride or sodium hydrosulfite in the presence of tetrabutylammonium chloride, at a temperature ranging from 0°C to reflux and for a time varying from about 1 hour to about 96 hours. The hydrogenation catalyst is usually a metal, most often palladium, which can be used as such or supported on carbon.
According to the conversion 2), the acylation of a compound of formula (I) wherein one of the substituents R1 , R2, R3, R4 or R5 is NH2, by reaction with an acylating agent of formula (XX) or (XXI) to give a compound of formula (I) wherein such substituent is a NHSO2R9 or NHCOR10 residue, can be carried out in a variety of ways, according to conventional methods well known in the literature. Preferably this conversion is carried out in a way analogous to that specified above under C).
According to the conversion 3), the reaction of a compound of formula (I), wherein the substituent R1 is NH2, with a suitable aldehyde or ketone for obtaining a compound of formula (I) wherein such substituent is a NR6R7 group, can be conducted in a variety of ways, according to conventional methods for carrying out reductive alkylations. Preferably, this reaction is carried out in a suitable solvent such as, for instance, methanol, N,N-dimethylformamide, dichloromethane, tetrahydrofuran, or a mixture thereof, in the presence of a suitable reducing agent such as, for instance, sodium borohydride, tetra-alkylammonium borohydride, sodium cyano borohydride, sodium triacetoxyborohydride, tetramethylammonium triacetoxy borohydride and in the presence of an acid catalyst, such as, for instance, acetic acid or trifluoroacetic acid, at a temperature ranging from about 0°C to reflux and for a time varying from about 1 hour to about 96 hours.
According to the conversion 4), the reaction of a compound of formula (I), wherein one of the substituents R2, R3, R4 or R5 is NH2, with a suitable aldehyde or ketone in the presence of a reducing agent, for obtaining a compound of formula (I) wherein such substituent is a NR11 R12 group, can be conducted in a variety of ways, according to conventional methods for carrying out reductive alkylations. Preferably this conversion is carried out in a way analogous to that specified above under 3).
According to the conversion 5), the hydrolysis of a compound of formula (I) wherein one of the substituents R1 , R2, R3, R4 or R5 is a COOR13 residue, wherein R13 is a straight or branched Ο-Οβ alkyl, to give the corresponding carboxylic acid can be conducted in a variety of ways, according to methods widely known in the art for the hydrolysis of ester groups. Preferably such hydrolysis is carried out in the presence of an inorganic base, such as, for example, lithium, sodium or potassium hydroxide, or an inorganic or organic acid, such as, for example, hydrochloric acid, trifluoroacetic acid, in a suitable solvent such as, for instance, methanol, ethanol, tetrahydrofuran, 1 ,4-dioxane, water or a mixture thereof, at a temperature ranging from about 0°C to reflux and for a time varying from about 1 hour to about 96 hours.
According to the conversion 6), the amidation of a compound of formula (I) wherein one of the substituents R1 , R2, R3, R4 or R5 is a COOH residue, with an amine of formula (XXII), can be can be conducted in a variety of ways, according to conventional methods for the synthesis of carboxamides. Preferably, this conversion is carried out in the presence of an activating agent such as hydroxybenzotriazole, dicyclohexyl carbodiimide, diisopropyl carbodiimide, 1-ethyl-3-(3'-dimethylamino)carbodiimide hydrochloric acid salt, 0-(benzotriazol-1-yl)-N,N,N',N'-tetramethyluronium tetrafluoroborate, in a suitable solvent such as, for instance, tetrahydrofuran, dichloromethane, toluene, 1 ,4-dioxane, N,N-dimethylformamide, Ν,Ν-dimethylacetamide and in the presence of a proton scavenger such as, for example, pyridine, triethylamine, Ν,Ν-diisopropylethylamine, at a temperature ranging from room temperature to reflux, for a time ranging from about 30 min. to about 96 hours.
According to the conversion 7), the oxidation of a compound of formula (I) wherein R1 is 4-methyl-piperazin-1-yl for obtaining a compound of formula (I) wherein such substituent is 4-methyl-4-oxy-piperazin-1-yl can be conducted in a variety of ways, according to conventional methods for the N-oxidation of tertiary amines. Preferably, this conversion is carried out in the presence of an oxidizing agent such as, for example, 3-chloroperbenzoic acid, hydrogen peroxide, dimethyldioxirane, in a suitable solvent such as, for instance, dichloromethane, methanol, ethanol, water, acetone, or a mixture thereof, at a temperature ranging from -10°C to reflux, for a time ranging from about 30 min. to about 96 hours.
According to the conversion 8) the cleavage of the tert-butoxy-carbonyl group of a compound of formula (I) wherein R1 is 4-te rt- b u toxyca rbonyl-piperazin-1-yl for obtaining a compound of formula (I) wherein such substituent is piperazin-1-yl can be carried out in a way analogous to that specified above under F).
It is known to the skilled person that when a compound of formula (V), formula (XVII), formula (XX) or formula (XXI) carries functional groups that may interfere in acylation reactions, such groups have to be protected before carrying out the reaction. In particular, when a compound of formula (V), formula (XVII), formula (XX) or formula (XXI) is substituted by residues of general formula NR6R7, NR11 R12, OR8, or OR13 wherein R8 or R13 or at least one of R6 and R7 or at least one of R11 and R12 represent hydrogen, such groups may be protected as known in the art. It is also known to the skilled person that such protecting group may be removed just after the reaction or at a later stage in the synthetic process.
The deprotection of a compound of formula (I) wherein one of the substituents is a protected amino group can be made in a variety of ways according to conventional methods for deprotecting amino groups. Depending on the amino protecting group, this reaction can be conducted in different ways. In one aspect, such reaction can be carried out by treatment with an inorganic acid, such as hydrochloric, sulphuric or perchloric acid, or an organic acid, such as trifluoroacetic or methanesulfonic acid, in a suitable solvent, such as water, methanol, ethanol, 1 ,4-dioxane, tetrahydrofuran, diethyl ether, diisopropyl ether, acetonitrile, Ν,Ν-dimethylformamide, dichloromethane or mixtures thereof, at a temperature ranging from -20°C to 80°C, and for a period of time ranging from 30 minutes to 48 hours. In another aspect, such reaction can be carried out by treatment with an inorganic base, such as lithium or sodium or potassium hydroxide, or sodium or potassium or cesium carbonate, or with an organic base, such as triethylamine or Ν,Ν-diisopropylethylamine, or with anhydrous hydrazine or hydrazine hydrate in a suitable solvent such as water, methanol, ethanol, 1 ,4-dioxane, tetrahydrofuran, diethyl ether, diisopropyl ether, acetonitrile, N,N-dimethylformamide, dichlorometane or mixtures thereof, at a temperature ranging from -20°C to 80°C, and for a period of time ranging from 30 minutes to 72 hours.
It is known to the skilled person that transformation of a chemical function into another may require that one or more reactive centers in the compound containing this function be protected in order to avoid undesired side reactions. Protection of such reactive centers, and subsequent deprotection at the end of the synthetic transformations, can be accomplished following standard procedures described, for instance, in: Green, Theodora W. and Wuts, Peter G.M. - Protective Groups in Organic Synthesis, Third Edition, John Wiley & Sons Inc., New York (NY), 1999.
In cases where a compound of formula (I) contains one or more asymmetric centers, said compound can be separated into the single isomers by procedures known to those skilled in the art. Such procedures comprise standard chromatographic techniques, including chromatography using a chiral stationary phase, or crystallization. General methods for separation of compounds containing one or more asymmetric centers are reported, for instance, in Jacques, Jean; Collet, Andre; Wilen, Samuel H., - Enantiomers, Racemates, and Resolutions, John Wiley & Sons Inc., New York (NY), 1981.
A compound of formula (I) can also be transformed into a pharmaceutically acceptable salt according to standard procedures that are known to those skilled in the art. Alternatively, a compound of formula (I) that is obtained as a salt can be transformed into the free base or the free acid according to standard procedures that are known to the skilled person.
According to any variant of the process for preparing the compounds of formula (I), the starting materials and any other reactant, i.e. compounds of formula (II), (V), (VIII), (IX), (X), (XIV), (XVII), (XIX), (XX), (XXI) and (XXII) are either commercially available, known, or easily prepared according to well-known methods described, for instance, in: B.M.Trost and I. Fleming, Comprehensive Organic Synthesis, 1991 , Pergamon Press; A.R. Katritzky, 0. Meth-Cohn and C.W. Rees, Comprehensive Organic Functional Group Transformations, 1995, Elsevier Pergamon; A.R. Katritzky and R.J.K. Taylor, Comprehensive Organic Functional Group Transformations II, 2005, Elsevier Pergamon. In particular, the compound of formula (II) can be prepared as described in WO2003028720 and the compound of formula (XIV) is commercially available.
The compounds of the present invention can be administered either as single agents or, alternatively, in combination with known anticancer treatments such as radiation therapy or chemotherapy regimen in combination with cytostatic or cytotoxic agents, antibiotic-type agents, alkylating agents, antimetabolite agents, hormonal agents, immunological agents, interferon-type agents, cyclooxygenase inhibitors (e.g. COX-2 inhibitors), matrixmetalloprotease inhibitors, telomerase inhibitors, tyrosine kinase inhibitors, anti-growth factor receptor agents, anti-HER agents, anti-EGFR agents, anti-angiogenesis agents (e.g. angiogenesis inhibitors), farnesyl transferase inhibitors, ras-raf signal transduction pathway inhibitors, cell cycle inhibitors, other cdks inhibitors, tubulin binding agents, topoisomerase I inhibitors, topoisomerase II inhibitors, and the like.
If formulated as a fixed dose, such combination products employ the compounds of this invention within the dosage range described below and the other pharmaceutically active agent within the approved dosage range.
Compounds of formula (I) may be used sequentially with known anticancer agents when a combination formulation is inappropriate.
The compounds of formula (I) of the present invention, suitable for administration to a mammal, e.g., to humans, can be administered by the usual routes and the dosage level depends upon the age, weight, conditions of the patient and administration route.
For example, a suitable dosage adopted for oral administration of a compound of formula (I) may range from about 10 to about 1000 mg per dose, from 1 to 5 times daily. The compounds of the invention can be administered in a variety of dosage forms, e.g., orally, in the form tablets, capsules, sugar or film coated tablets, liquid solutions or suspensions; rectally in the form suppositories; parenterally, e.g., intramuscularly, or through intravenous and/or intrathecal and/or intraspinal injection or infusion.
The present invention also includes pharmaceutical compositions comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof in association with a pharmaceutically acceptable excipient, which may be a carrier or a diluent. The pharmaceutical compositions containing the compounds of the invention are usually prepared following conventional methods and are administered in a suitable pharmaceutical form. For example, the solid oral forms may contain, together with the active compound, diluents, e.g., lactose, dextrose saccharose, sucrose, cellulose, corn starch or potato starch; lubricants, e.g., silica, talc, stearic acid, magnesium or calcium stearate, and/or polyethylene glycols; binding agents, e.g., starches, arabic gum, gelatine methylcellulose, carboxymethylcellulose or polyvinyl pyrrolidone; disintegrating agents, e.g., starch, alginic acid, alginates or sodium starch glycolate; effervescing mixtures; dyestuffs; sweeteners; wetting agents such as lecithin, polysorbates, laurylsulphates; and, in general, nontoxic and pharmacologically inactive substances used in pharmaceutical formulations. These pharmaceutical preparations may be manufactured in known manner, for example, by means of mixing, granulating, tabletting, sugar-coating, or film-coating processes.
The liquid dispersions for oral administration may be, e.g., syrups, emulsions and suspensions. As an example, the syrups may contain, as carrier, saccharose or saccharose with glycerine and/or mannitol and sorbitol.
The suspensions and the emulsions may contain, as examples of carriers, natural gum, agar, sodium alginate, pectin, methylcellulose, carboxymethylcellulose, or polyvinyl alcohol. The suspension or solutions for intramuscular injections may contain, together with the active compound, a pharmaceutically acceptable carrier, e.g., sterile water, olive oil, ethyl oleate, glycols, e.g., propylene glycol and, if desired, a suitable amount of lidocaine hydrochloride. The solutions for intravenous injections or infusions may contain, as a carrier, sterile water or preferably they may be in the form of sterile, aqueous, isotonic, saline solutions or they may contain propylene glycol as a carrier.
The suppositories may contain, together with the active compound, a pharmaceutically acceptable carrier, e.g., cocoa butter, polyethylene glycol, a polyoxyethylene sorbitan fatty acid ester surfactant or lecithin.
With the aim of better illustrating the present invention, without posing any limitation to it, the following examples are now given.
EXPERIMENTAL SECTION
For a reference to any specific compound of formula (I) of the invention, optionally in the form of a pharmaceutically acceptable salt, see the experimental section and claims. Referring to the examples that follow, compounds of the present invention were synthesized using the methods described herein, or other methods, which are well known in the art.
In the examples below, the short forms and abbreviations used herein have the following meaning.
ABBREVIATIONS
EtOAc Ethyl acetate
DCM Dichloromethane
DIPEA A,A -Diisopropylethylamine
DMF N,N-dimethylformamide
DMSO Dimethyl sulfoxide
Et20 Diethyl ether
EtOH Ethanol
HCI Hydrochloric acid
K2CO3 Potassium carbonate
□HMDS Lithium bis(trimethylsilyl)amide
MeOH Methanol
Figure imgf000026_0001
With the aim at better illustrating the present invention, without posing any limitation to it, the following examples are now given.
As used herein the symbols and conventions used in the processes, schemes and examples are consistent with those used in the contemporary scientific literature, for example, the Journal of the American Chemical Society or the Journal of Biological Chemistry.
Unless otherwise noted, all materials were obtained from commercial suppliers, of the best grade and used without further purification. Anhydrous solvent such as DMF, THF, DCM and toluene were obtained from the Aldrich Chemical Company. All reactions involving air- or moisture-sensitive compounds were performed under nitrogen or argon atmosphere.
General purification and analytical methods
Thin-layer chromatography (TLC) was performed on Merck silica gel 60 F254 pre-coated plates. Column chromatography was conducted either under medium pressure on silica (Merck silica gel 40-63 μιτι) or performed by using a Biotage SP1 Flash Purification system with prepacked silica gel cartridges (Biotage or Varian).
1H-NMR spectra were recorded in DMSO-de or CDCI3 at a constant temperature of 28°C on a Varian INOVA 400 spectrometer operating at 400.50 MHz and equipped with a 5 mm z-axis PFG Indirect Detection Probe (1H{15N-31P}) and on a Varian Inova 500 spectrometer operating at 499.75 MHz. Residual solvent signal was used as reference (δ = 2.50 or 7.27 ppm). Chemical shifts (δ) are reported in parts per million (ppm) and coupling constants (J) in Hz. The following abbreviations are used for multiplicities: s = singlet; bs = broad signal; d = doublet; t = triplet; m = multiplet; dd = doublet of doublets.
Electrospray (ESI) mass spectra were obtained on a Finnigan LCQ ion trap. Unless otherwise specified, all final compounds were homogeneous (purity of not less than 95%), as determined by high-performance liquid chromatography (HPLC). HPLC-UV-MS analyses, used to assess compound purity, were carried out combining the ion trap MS instrument with HPLC system SSP4000 (Thermo Separation Products) equipped with an autosampler LC Pal (CTC Analytics) and UV6000LP diode array detector (UV detection 215-400 nm). Instrument control, data acquisition and processing were performed with the Xcalibur 1.2 software (Finnigan). HPLC chromatography was run at r.t., and 1 mL/min flow rate, using a Waters X Terra RP 18 column (4.6x 50 mm; 3.5 μιτι). Mobile phase A was ammonium acetate 5 mM buffer (pH 5.5 with acetic acid): acetonitrile 90:10, and mobile phase B was ammonium acetate 5 mM buffer (pH 5.5 with acetic acid): acetonitrile 10:90; the gradient was from 0 to 100% B in 7 minutes then hold 100% B for 2 minutes before requilibration.
ESI(+) high resolution mass spectra (HRMS) were obtained on a Waters Q-Tof Ultima directly connected with micro HPLC 1100 Agilent as previously described (Colombo, M.; Riccardi-Sirtori, F.; Rizzo, V.; A fully automated method for accurate mass determination using high-performance liquid chromatography with a quadrupole/orthogonal acceleration time-of-flight mass spectrometer. Rapid Commun. Mass Spectrom. 2004, 18, 511-517).
Preparation 1
6-(tert-butyl-dimethyl-silanyloxy)-3-(1,3-dioxo-1,3-dihydro-isoindol-2-yl)-ind acid tert-butyl ester (III)
Scheme 1, Step A)
A solution of 2-[6-(tert-butyl-dimethyl-silanyloxy)-1 H-indazol-3-yl]-isoindole-1 ,3-dione (786 mg, 2 mmol) in dry THF (10 ml) and DIPEA (1.4 ml, 8 mmol), under argon atmosphere, was treated with di-tert-butyl dicarbonate (495 mg, 2.2 mmol) and stirred at r.t. overnight. The solvent was evaporated under reduced pressure and the crude residue purified by flash chromatography over silica gel eluting with hexane / EtOAc 7:3 affording 884 mg (yield: 89%) of the title compound as a white solid.
1H-NMR (400 MHz), δ (ppm, DMSO-cie): 8.09 - 8.04 (m, 2 H), 8.02 - 7.96 (m, 2 H), 7.77 (d, J=8.29 Hz, 1 H), 7.55 (d, J=1.95 Hz, 1 H), 7.00 (dd, J=2.07, 8.78 Hz, 1 H), 1.69 (s, 9H), 1.01 (s, 9H), 0.28 (s, 6H)
ESI (+) MS m/z 494 (MH+)
Preparation 2
3-Amino-6-(tert-butyl-dimethyl-silanyloxy)-indazole-1-carboxylic acid tert-butyl ester (IV)
Scheme 1, Step B)
A solution of 6-(tert-butyl-dimethyl-silanyloxy)-3-(1 ,3-dioxo-1 ,3-dihydro-isoindol-2-yl)-indazole-1-carboxylic acid tert- butyl ester (1.17 g, 2.37 mmol) in dry THF (20 ml), under argon atmosphere, was treated with 3.56 ml of 1 M hydrazine in THF (3.56 mmol). The mixture was stirred at reflux for 1 h then more 1 M hydrazine in THF was added (8 ml). After stirring for additional 2h at reflux, the reaction mixture was cooled to r.t. and the precipitated solid filtered and washed with THF. The filtrate was evaporated to dryness and the residue purified by flash chromatography over silica gel eluting with DCM / acetone 7:3 affording 785 mg (yield: 91%) of the title compound as a yellowish solid. 1H-NMR (400 MHz), δ (ppm, DMSO-cie): 7.71 (d, J=8.90 Hz, 1 H), 7.37 (bs, 1 H), 6.81 (dd, J=2.07, 8.54 Hz, 1 H), 6.21 (bs, 2H), 1.59 (s, 9H), 0.99 (s, 9H), 0.23 (s, 6H)
ESI (+) MS mil 364 (MH+)
Preparation 3
6-(tert-Butyl-dimethyl-silanyloxy)-3-[4-(4-methyl-piperazin-1-yl)-benzoylamino]-i acid tert-butyl ester
Scheme 1, Step C)
To a suspension of 4-(4-methyl-piperazin-1-yl)-benzoic acid (1.64 g, 7.48 mmol) in dry THF (50 ml), under argon atmosphere, at r.t., was added thionyl chloride (1.37 ml, 18.9 mmol) and 3 drops of dry DMF. The reaction mixture was heated to 50°C and stirred for 5h then the volatiles were removed under reduced pressure. The residue was taken up in dry toluene (50 ml), re-evaporated and the solid residue dried under high vacuum. The resultant crude 4- (4-methyl-piperazin-1-yl)-benzoyl chloride hydrochloride was suspended in 20 ml of dry THF and treated dropwise at r.t., under argon atmosphere, with a solution of 3-amino-6-(tert-butyl-dimethyl-silanyloxy)-indazole-1-carboxylic acid tert-butyl ester (1.81 g, 4.98 mmol) in 20 ml of dry THF and 2.56 ml of DIPEA (14.96 mmol). The reaction mixture was heated to 50°C and stirred for 22h. The volatiles were removed under reduced pressure, the residue diluted with DCM (100 ml) and washed with a saturated solution of NaHC03 (75 ml). The organic layer was separated, dried over sodium sulfate and evaporated to dryness. The crude residue was purified by flash chromatography over silica gel eluting with DCM / MeOH / 30% NH3 95:5:0.5 affording 1.88 g (yield: 67%) of the title compound.
1H-NMR (400 MHz), δ (ppm, DMSO-cie): 10.95 (s, 1 H), 8.03 - 7.96 (m, 2H), 7.73 (d, J=9.15 Hz, 1 H), 7.51 (d, J=2.07 Hz, 1 H), 7.06 - 7.01 (m, 2H), 6.90 (dd, J=2.19, 8.78 Hz, 1 H), 3.34 - 3.32 (m, 4H), 2.53 - 2.50 (m, 4H), 2.27 (s, 3H), 1.65 (s, 9H), 1.00 (s, 9H), 0.27 (s, 6H)
ESI (+) MS mil 566 (MH+)
Preparation 4
6-Hydroxy-3-[4-(4-methyl-piperazin-1-yl)-benzoylamino]-indazole-1-carboxylic acid tert-butyl ester
Scheme 1, Step D)
A solution of 6-(tert-butyl-dimethyl-silanyloxy)-3-[4-(4-methyl-piperazin-1-yl)-benzoylamino]-indazole-1-carboxylic acid tert-butyl ester 1.88 g (3.33 mmol) in dry THF (20 ml) was treated with 1 M TBAF in THF (4 ml, 4 mmol) and stirred at r.t. for 30 min. Water (20 ml) was then added and the mixture extracted with EtOAc (100ml). The separated organic layer was dried over sodium sulfate and evaporated to dryness. The residue was purified by flash chromatography over silica gel eluting with DCM / MeOH / 30% NH3 92:8:0.8 affording, after trituration with Et20, 1.5 g (yield: 100%) of the title compound.
1H-NMR (400 MHz), δ (ppm, DMSO-cie): 10.86 (s, 1 H), 10.16 (s, 1 H), 8.02 - 7.94 (m, 2H), 7.65 (d, J=8.78 Hz, 1 H), 7.48 (d, J=1.95 Hz, 1 H), 7.05 - 7.00 (m, 2H), 6.82 (dd, J=2.07, 8.78 Hz, 1 H), 3.34 - 3.32 (m, 4H), 2.55 - 2.45 (m, 4H), 2.27 (s, 3H), 1.65 (s, 9H) ESI (+) MS m/z 452 (MH+)
Preparation 5
3-[4-(4-Methyl-piperazin-1-yl)-benzoylamino]-6-(3-phenoxy-benzyloxy)-indazole-1-carboxyN tert-butyl ester
Scheme 1, Step Ea)
A solution of triphenylphosphine (209 mg, 0.798 mmol) and diisopropyl azodicarboxylate (0.152 ml, 0.732 mmol) in dry DCM (2 ml) was stirred under argon at 4°C for 15 min. The resultant mixture was then added to a stirred solution of 6-hydroxy-3-[4-(4-methyl-piperazin-1-yl)-benzoylamino]-indazole-1-carboxylic acid tert-butyl ester (100 mg, 0.222 mmol) and (3-phenoxy-phenyl)-methanol (149 mg, 0.732 mmol) in 2 ml of dry DCM, at r.t., under argon. After stirring at r.t. overnight the reaction mixture was adsorbed onto silica gel, dried, loaded into a silica gel chromatographic column and eluted with DCM / MeOH / 30% NH3 95:5:0.5 affording 87 mg (yield: 62%) of the title compound.
ESI (+) MS m/z 634 (MH+)
Operating in an analogous way, the following compounds were obtained:
6-Benzyloxy-3-[4-(4-methyl-piperazin-1-yl)-benzoylamino]-indazole-1-carboxylic acid tert-butyl ester ESI (+) MS mil 542 (MH+)
3-[4-(4-Methyl-piperazin-1-yl)-benzoylamino]-6-(3-phenyl-prop-2-ynyloxy)-indazole-1-carboxylic acid tert-butyl ester
ESI (+) MS mil 566 (MH+)
6-(1-Benzyl-piperidin-4-yloxy)-3-[4-(4-methyl-piperazin-1-yl)-benzoylamino]-indazole-1-carboxylic acid tert- butyl ester
ESI (+) MS m/z 625 (MH+)
3-[4-(4-Methyl-piperazin-1-yl)-benzoylamino]-6-(2-phenoxy-ethoxy)-indazole-1-carboxylic acid tert-butyl ester
ESI (+) MS mil 572 (MH+)
6-(1-Benzyl-piperidin-3-yloxy)-3-[4-(4-methyl-piperazin-1-yl)-benzoylamino]-indazole-1-carboxylic acid tert- butyl ester
ESI (+) MS m/z 625 (MH+)
6-(1-Benzyl-pyrrolidin-2-ylmethoxy)-3-[4-(4-methyl-piperazin-1-yl)-benzoylamino]-indazole-1-carbo acid tert-butyl ester
ESI (+) MS m/z 625 (MH+)
Preparation 6
3-[4-(4-Methyl-piperazin-1-yl)-benzoylamino]-6-phenoxy-indazole-1-carboxylic acid tert-butyl ester
Scheme 1, Step Eb)
A mixture of 6-hydroxy-3-[4-(4-methyl-piperazin-1-yl)-benzoylamino]-indazole-1-carboxylic acid tert-butyl ester (110 mg, 0.244 mmol), phenylboronic acid (92 mg, 0.732 mmol), copper diacetate (45 mg, 0.244 mmol) and 4A molecular sieves (200 mg) in 5 ml of DCM was treated with TEA (0.339 ml, 2.44 mmol). After stirring at r.t. for 2 days the solvent was removed under reduced pressure, the residue treated with DCM / MeOH / 30% NH3 95:5:0.5, filtered and the filtrate loaded into a silica gel chromatographic column and eluted with DCM / MeOH / 30% NH3 95:5:0.5 affording 88 mg (yield: 68%) of the title compound.
ESI (+) MS mil 528 (MH+)
Operating in an analogous way, the following compounds were obtained:
6-(3-Fluoro^henoxy)-3-[4-(4-methyl-piperazin-1-yl)-benzoylamino]-indazole-1-carboxylic acid tert-butyl ester
ESI (+) MS mil 546 (MH+)
6-(4-Benzyloxy-phenoxy)-3-[4-(4-methyl-piperazin-1-yl)-benzoylamino]-indazole-1-carboxylic acid tert-butyl ester
ESI (+) MS m/z 634 (MH+)
3-[4-(4-Methyl-piperazin-1-yl)-benzoylamino]-6-(4-phenoxy-phenoxy)-indazole-1-carboxylic acid tert-butyl ester
ESI (+) MS m/z 620 (MH+)
6-(3-Benzyloxy-phenoxy)-3-[4-(4-methyl-piperazin-1-yl)-benzoylamino]-indazole-1-carboxylic acid tert-butyl ester
ESI (+) MS m/z 634 (MH+)
Preparation 7
3-Amino-6-hydroxy-indazole-1-carboxylic acid tert-butyl ester (XII)
Scheme 1, Step G)
A solution of 3-amino-6-(tert-butyl-dimethyl-silanyloxy)-indazole-1-carboxylic acid tert-butyl ester 672 mg (1.85 mmol) in dry THF (10 ml) was treated with 1 M TBAF in THF (1.85 ml, 1.85 mmol) and stirred at r.t. for 30 min. Water (20 ml) was then added and the mixture extracted with EtOAc (3X50 ml). The separated organic layers were combined, dried over sodium sulfate and evaporated to dryness. The residue was purified by flash chromatography over silica gel eluting with DCM / EtOAc 6:4 affording 429 mg (yield: 93%) of the title compound.
1H-NMR (400 MHz), δ (ppm, DMSO-cie): 9.93 (s, 1 H), 7.60 (d, J=8.54 Hz, 1 H), 7.35 (bs, 1 H), 6.71 (dd, J=2.19, 8.54 Hz, 1 H), 6.10 (bs, 2H), 1.58 (s, 9H)
ESI (+) MS m/z 250 (MH+)
Preparation 8
3-Amino-6-(2-benzyloxy-ethoxy)-indazole-1-carboxylic acid tert-butyl ester
Scheme 1, Step Hb)
A mixture of 3-amino-6-hydroxy-indazole-1-carboxylic acid tert-butyl ester (249 mg, 1 mmol), K2CO3 (152 mg, 1.1 mmol) and benzyl 2-bromoethyl ether (0.179 ml, 1.1 mmol) in dry DMF (5 ml) was stirred at 50°C for 12h. More benzyl 2-bromoethyl ether (30 μΙ) was added and the mixture stirred at 50°C for additional 4h. The reaction mixture was poured into water (50 ml) and extracted with EtOAc (50 ml). The organic layer wad dried over sodium sulfate and evaporated to dryness. The crude residue was purified by flash chromatography over silica gel eluting with DCM / EtOAc 7:3 affording 274 mg (yield: 72%) of the title compound.
1H-NMR (400 MHz), δ (ppm, DMSO-cie): 7.71 (d, J=8.66 Hz, 1 H), 7.48 (bs, 1 H), 7.38 - 7.27 (m, 5H), 6.91 (dd, J=2.19, 8.66 Hz, 1 H), 6.19 (bs, 2H), 4.59 (s, 2H), 4.25 - 4.20 (m, 2H), 3.85 - 3.80 (m, 2H), 1.58 (s, 9H) ESI (+) MS m/z 384 (MH+)
Preparation 9
6-(2-Benzyloxy-ethoxy)-3-[4-(4-methyl^iperazin-1-yl)-benzoylamino]-indazole-1-carboxylic acid tert-butyl ester
Scheme 1, Step I)
A mixture of 4-(4-methyl-piperazin-1-yl)-benzoyl chloride hydrochloride, prepared from 248 mg (1.13 mmol) of 4-(4- methyl-piperazin-1 -yl)-benzoic acid as described above in preparation 3, and DIPEA (0.386 ml, 2.25 mmol) in dry THF (10 ml) was treated dropwise at r.t., under argon atmosphere, with a solution of 3-amino-6-(2-benzyloxy- ethoxy)-indazole-1-carboxylic acid tert-butyl ester (143 mg, 0.374 mmol) in 10 ml of dry THF. The reaction mixture was heated up to 50°C and stirred for 12h. The volatiles were removed under reduced pressure, the residue taken up in DCM (100 ml) and washed with a saturated solution of NaHC03 (75 ml). The organic layer was separated, dried over sodium sulfate and evaporated to dryness and the crude residue purified by flash chromatography over silica gel eluting with DCM / MeOH 98:2 then 90:10 affording 124 mg (yield: 57%) of the title compound as a white solid.
1H-NMR (400 MHz), δ (ppm, DMSO-cie): 10.94 (s, 1 H), 8.02 - 7.96 (m, 2H), 7.75 (d, J=8.90 Hz, 1 H), 7.61 (d, J=2.19 Hz, 1 H), 7.39 - 7.27 (m, 5H), 7.06 - 7.01 (m, 2H), 7.00 (dd, J=2.19, 8.90 Hz, 1 H), 4.60 (s, 2H), 4.31 - 4.25 (m, 2H), 3.88 - 3.83 (m, 2H), 3.34 - 3.32 (m, 4H), 2.53 - 2.46 (m, 4H), 2.27 (s, 3H), 1.66 (s, 9H)
ESI (+) MS m/z 586 (MH+)
Example 1
4-(4-Methyl^iperazin-1-yl)-N-[6-(3-phenoxy-benzyloxy)-1 H-indazol-3-yl]-benzamide (Cpd. 5)
Scheme 1, Step F)
Figure imgf000031_0001
A solution of 3-[4-(4-methyl-piperazin-1-yl)-benzoylamino]-6-(3-phenoxy-benzyloxy)-indazole-1-carboxylic acid tert- butyl ester (87 mg, 0.137 mmol) in DCM / TFA 8:2 (5 ml) was stirred at r.t. for 2h. The volatiles were removed under reduced pressure and the residue purified by flash chromatography over silica gel eluting with DCM / MeOH / 30% NH3 90:10:1 affording 71 mg (yield: 97%) of the title compound as a white solid.
1H-NMR (400 MHz), δ (ppm, DMSO-cie): 12.46 (s, 1 H) 10.38 (s, 1 H) 7.94 (d, J=8.97 Hz, 2 H) 7.59 (d, J=8.79 Hz, 1 H) 7.30 - 7.43 (m, 3 H) 7.25 (d, J=7.69 Hz, 1 H) 7.07 - 7.17 (m, 2 H) 6.97 - 7.04 (m, 4 H) 6.95 (dd, J=8.06, 1.83 Hz, 1 H) 6.90 (d, J=2.01 Hz, 1 H) 6.75 (dd, J=8.88, 2.11 Hz, 1 H) 5.18 (s, 2 H) 3.25 - 3.32 (m, 4 H) 2.42 - 2.48 (m, 4 H) 2.23 (s, 3 H)
ESI (+) MS m/z 534 (MH+) ESI (+) HRMS calcd for C32H3iN503 + H+: 534.2500; found 534.2488
Operating in an analogous way, the following compounds were obtained:
N-(6-Benzyloxy-1 H-indazol-3-yl)-4-(4-methyl-piperazin-1-yl)-benzamide (Cpd. 1)
Figure imgf000032_0001
1H-NMR (400 MHz), δ (ppm, DMSO-cie): 12.46 (s, 1 H), 10.40 (s, 1 H), 8.00-7.94 (m, 2H), 7.62 (d, J=9.02 Hz, 1 H), 7.53 - 7.32 (m, 5H), 7.05 - 7.00 (m, 2H), 6.93 (d, J=1.83 Hz, 1 H), 6.78 (dd, J=2.19, 8.90 Hz, 1 H), 5.20 (s, 2H), 3.34 - 3.32 (m, 4H), 2.53 - 2.46 (m, 4H), 2.28 (s, 3H)
ESI (+) MS m/z 442 (MH+)
ESI (+) HRMS calcd for C26H27N502 + H+: 442.2238; found 442.2237
4-(4-Methyl-piperazin-1-yl)-N-[6-(3-phenyl-prop-2- n lox -1 H-indazol-3-yl]-benzamide (Cpd. 7)
Figure imgf000032_0002
1H-NMR (400 MHz), δ (ppm, DMSO-cie): 12.53 (s, 1 H), 10.42 (s, 1 H), 8.00 - 7.95 (m, 2H), 7.64 (d, J=8.90 Hz, 1 H), 7.50 - 7.37 (m, 5H), 7.06 - 7.00 (m, 3H), 6.78 (dd, J=2.19, 9.02 Hz, 1 H), 5.13 (s, 2H), 3.34 - 3.32 (m, 4H), 2.53 - 2.46 (m, 4H), 2.28 (s, 3H)
ESI (+) MS m/z 466 (MH+)
ESI (+) HRMS calcd for C28H27N502 + H+: 466.2237; found 466.2255
N-[6-(1-Benzyl-piperidin -yloxy)-1 H-indazol-3-yl]-4-(4-methyl-piperazin-1-yl)-benzamide (Cpd. 6)
Figure imgf000032_0003
1H-NMR (400 MHz), δ (ppm, DMSO-cie): 12.38 (s, 1 H), 10.39 (s, 1 H), 7.99 - 7.94 (m, 2H), 7.59 (d, J=8.90 Hz, 1 H), 7.38 - 7.24 (m, 5H), 7.04 - 6.99 (m, 2H), 6.88 (d, J=2.07 Hz, 1 H), 6.71 (dd, J=2.07, 8.90 Hz, 1 H), 4.52 - 4.44 (m, 1 H), 3.53 (s, 2H), 3.34 - 3.32 (m, 4H), 2.78 - 2.65 (m, 2H), 2.53 - 2.46 (m, 4H), 2.37 - 2.23 (m, 5H), 2.05 - 1.94 (m, 2H), 1.77 - 1.64 (m, 2H)
ESI (+) MS mil 525 (MH+)
ESI (+) HRMS calcd for CsiHseNeCfc + H+: 525.2972; found 525.2988
4-(4-Methyl-piperazin-1-yl)-N-[6-(2-phenox -ethoxy)-1 H-indazol-3-yl]-benzamide (Cpd. 10)
Figure imgf000033_0001
1H-NMR (400 MHz), δ (ppm, DMSO-cie): 12.49 (s, 1 H), 10.40 (s, 1 H), 7.99 - 7.95 (m, 2H), 7.62 (d, J=8.90 Hz, 1 H), 7.37 - 7.29 (m, 2H), 7.05 - 7.00 (m, 4H), 7.00 - 6.94 (m, 1 H), 6.92 (d, J=2.07 Hz, 1 H), 6.74 (dd, J=2.07, 8.90 Hz, 1 H), 4.41 - 4.36 (m, 4H), 3.35 - 3.30 (m, 4H), 2.52 - 2.46 (m, 4H), 2.27 (s, 3H)
ESI (+) MS m/z 472 (MH+)
ESI (+) HRMS calcd for C27N503H29 + H+: 472.2343; found 472.2357
N-[6-(1-Benzyl-piperidin-3-yloxy)-1 H-indazol-3-yl]-4-(4-methyl-piperazin-1-yl)-benzami (Cpd. 12)
Figure imgf000033_0002
1 H-NMR (400 MHz), δ (ppm, DMSO-cie): 12.37 (s, 1 H), 10.38 (s, 1 H), 7.99 - 7.93 (m, 2H), 7.58 (d, J=8.90 Hz, 1 H), 7.36 - 7.22 (m, 5H), 7.05 - 6.98 (m, 2H), 6.86 (d, J=1.83 Hz, 1 H), 6.68 (dd, J=2.20, 8.90 Hz, 1 H), 4.51 - 4.42 (m, 1 H), 3.55 (s, 2H), 3.35 - 3.30 (m, 4H), 3.02 - 2.95 (m, 1 H), 2.70 - 2.62 (m, 1 H), 2.49 - 2.43 (m, 4H), 2.27 (s, 3H), 2.20 - 2.02 (m, 3H), 1.81 - 1.71 (m, 1 H), 1.65 - 1.53 (m, 1 H), 1.48 - 1.36 (m, 1 H)
ESI (+) MS mil 525 (MH+)
ESI (+) HRMS calcd for C31 N6 02 H36 + H+: 525.2972; found 525.2975
N-[6-(1-Benzyl-pyrrolidin-2-ylmethoxy)-1 H-indazol-3-yl]-4-(4-methyl-piperazin-1-yl)-ben (Cpd. 13)
Figure imgf000034_0001
1H-NMR (400 MHz), δ (ppm, DMSO-cie): 12.43 (s, 1H), 10.39 (s, 1H), 7.99 - 7.93 (m, 2H), 7.59 (d, J=8.90 Hz, 1 H), 7.38 - 7.21 (m, 5H), 7.04 - 6.99 (m, 2H), 6.83 (d, J=1.95 Hz, 1H), 6.69 (dd, J=2.07, 8.90 Hz, 1H), 4.17 (d, J=13.17 Hz, 1H), 4.04 (dd, J=5.49, 9.63 Hz, 1H), 3.90 (dd, J=6.58, 9.63 Hz, 1H), 3.49 (d, J=13.17 Hz, 1H), 3.35 - 3.26 (m, 5H), 3.06 - 2.98 (m, 1H), 2.88 - 2.83 (m, 1H), 2.49 - 2.44 (m, 4H), 2.32 - 2.24 (m, 1H), 2.24 (s, 3H), 2.07 - 1.96 (m, 1H), 1.76- 1.66 (m,2H)
ESI (+) MS mil 525 (MH+)
ESI (+) HRMS calcd for C31N6O2H36 + H+: 525.2972; found 525.2969
4-(4-Methyl-piperazin-1-yl)-N-(6-phenoxy-1H-in zol-3-yl)-benzamide (Cpd.2)
Figure imgf000034_0002
1H-NMR (400 MHz), δ (ppm, DMSO-cie): 12.57 (s, 1H), 10.54 (s, 1H), 8.06 - 7.99 (m, 2H), 7.74 (d, J=8.78 Hz, 1 H), 7.48 - 7.40 (m, 2H), 7.23 - 7.17 (m, 1H), 7.13 - 7.06 (m, 4H), 6.90 (d, J=2.07 Hz, 1H), 6.84 (dd, J=2.07, 8.90 Hz, 1H), 3.35- 3.00 (m,8H), 2.74 (s, 3H)
ESI (+) MSm/z428(MH+)
ESI (+) HRMS calcd for C25N502H25 + H+: 428.2081; found 428.2092
N-[6-(3-Fluoro-phenoxy)-1H-indazol-3-yl]-4- -methyl-piperazin-1-yl)-benzamide (Cpd.3)
Figure imgf000034_0003
1H-NMR (400 MHz), δ (ppm, DMSO-cie): 12.64 (s, 1H), 10.53 (s, 1H), 8.03 - 7.98 (m, 2H), 7.77 (d, J=8.78 Hz, 1 H), 7.49 - 7.41 (m, 1H), 7.10 - 6.88 (m, 6H), 6.86 (dd, J=2.07, 8.78 Hz, 1H), 3.35 - 3.24 (m, 4H), 2.90 - 2.65 (m, 4H), 2.47 (bs, 3H) ESI (+) MS m/z 446 (MH+)
ESI (+) HRMS calcd for C25N502FH24 + H+: 446.1987; found 446.1981
N-[6-(4-Benzyloxy^henoxy)-1 H-indazol-3-ylH-(4-methyl^iperazin-1-yl)-benzamide (Cpd. 4)
Figure imgf000035_0001
1H-NMR (400 MHz), δ (ppm, DMSO-cie): 12.46 (s, 1 H), 10.47 (s, 1 H), 8.01 - 7.96 (m, 2H), 7.70 (d, J=8.90 Hz, 1 H), 7.52 - 7.32 (m, 5H), 7.15 - 7.00 (m, 6H), 6.81 (dd, J=2.19, 8.90 Hz, 1 H), 6.74 (d, J=2.19 Hz, 1 H), 5.13 (s, 2H), 3.35 - 3.30 (m, 4H), 2.70 - 2.55 (m, 4H), 2.38 (bs, 3H)
ESI (+) MS mil 534 (MH+)
ESI (+) HRMS calcd for C32N503H3i + H+: 534.2500; found 534.2498
4-(4-Methyl-piperazin-1-yl)-N-[6-(4-phenox -phenoxy)-1 H-indazol-3-yl]-benzamide (Cpd. 8)
Figure imgf000035_0002
1H-NMR (400 MHz), δ (ppm, DMSO-cie): 12.53 (s, 1 H), 10.48 (s, 1 H), 8.01 - 7.96 (m, 2H), 7.74 (d, J=8.78 Hz, 1 H), 7.45 - 7.39 (m, 2H), 7.19 - 6.99 (m, 9H), 6.89 (d, J=2.07 Hz, 1 H), 6.85 (dd, J=2.07, 8.78 Hz, 1 H), 3.35 - 3.30 (m, 4H), 2.52 - 2.47 (m, 4H), 2.28 (bs, 3H)
ESI (+) MS mil 520 (MH+)
ESI (+) HRMS calcd for C3iN503H29 + H+: 520.2343; found 520.2346
N-[6-(3-Benzyloxy^henoxy)-1 H-indazol-3-yl]-4-(4-methyl-piperazin-1-yl)-benzamide (Cpd. 9)
Figure imgf000035_0003
1H-NMR (400 MHz), δ (ppm, DMSO-cie): 12.58 (s, 1 H), 10.54 (s, 1 H), 8.05 - 7.99 (m, 2H), 7.74 (d, J=8.90 Hz, 1 H), 7.47 - 7.29 (m, 6H), 7.12 - 7.06 (m, 2H), 6.93 (d, J=1.95 Hz, 1 H), 6.87 - 6.81 (m, 2H), 6.76 - 6.73 (m, 1 H), 6.65 (dd, J=2.19, 8.17 Hz, 1 H), 5.11 (s, 2H), 3.35 - 2.80 (m, 8H), 2.65 (bs, 3H)
ESI (+) MS mil 534 (MH+)
ESI (+) HRMS calcd for C32N503H3i + H+: 534.2500; found 534.2501
N-[6-(2-Benzyloxy-ethoxy)-1H-indazol-3- l]-4-(4-methyl-piperazin-1-yl)-benzamide (Cpd. 11)
Figure imgf000036_0001
1H-NMR (400 MHz), δ (ppm, DMSO-cie): 12.44 (s, 1 H) 10.38 (s, 1 H) 7.95 (d, J=9.03 Hz, 2 H) 7.59 (d, J=8.91 Hz, 1 H) 7.32 - 7.42 (m, 4 H) 7.23 - 7.32 (m, 1 H) 7.00 (d, J=9.03 Hz, 2 H) 6.85 (d, J=1.95 Hz, 1 H) 6.71 (dd, J=8.91 , 2.07 Hz, 1 H) 4.58 (s, 2 H) 4.08 - 4.31 (m, 2 H) 3.72 - 3.92 (m, 2 H) 3.20 - 3.30 (m, 4H) 2.40 - 2.48 (m, 4 H) 2.23 (s, 3 H) ESI (+) MS m/z 486 (MH+)
ESI (+) HRMS calcd for C28N503H3i + H+: 486.2500; found 486.2502
Preparation 10
4-(2-Benzyloxy-ethoxy)-2-fluoro-benzonitrile
Scheme 2, Step Jc)
A mixture of 2-fluoro-4-hydroxy-benzonitrile (4.57 g, 33.3 mmol), K2C03 (13.8 g, 99.9 mmol) and benzyl 2-bromoethyl ether (5.79 ml, 36.6 mmol) in dry DMF (15 ml) was stirred at 70°C for 6h. The reaction mixture was cooled to r.t., poured into 300 ml of water and extracted with EtOAc (2X100 ml). The organic layers were combined, dried over sodium sulfate and evaporated to dryness. The crude residue was purified by chromatography (Biotage SP1 Flash Purification system) on a silica gel cartridge (Biotage SNAP 100 g) eluting with a gradient from hexane / EtOAc 100:0 to 60:40 over 20 CV, affording 8.79 g (yield: 97%) of the title compound as a colourless oil.
1H-NMR (400 MHz), δ (ppm, DMSO-de): 7.75 - 7.90 (m, 1 H) 7.26 - 7.38 (m, 5 H) 7.19 (dd, J=11.96, 2.44 Hz, 1 H) 6.99 (dd, J=8.79, 2.32 Hz, 1 H) 4.55 (s, 2 H) 4.22 - 4.34 (m, 2 H) 3.65 - 3.87 (m, 2 H)
ESI (+) MS m/z 272 (MH+)
Operating in an analogous way, the following compounds were obtained:
2-Fluoro-4-[2-(4-trifluoromethyl-benzyloxy)-ethoxy]-benzonitrile
1H-NMR (400 MHz), δ (ppm, DMSO-cie): 7.82 (t, J=8.33 Hz, 1 H) 7.71 (d, J=8.06 Hz, 2 H) 7.55 (d, J=7.88 Hz, 2 H) 7.19 (dd, J=12.00, 2.29 Hz, 1 H) 7.00 (dd, J=8.70, 2.29 Hz, 1 H) 4.66 (s, 2 H) 4.28 - 4.31 (m, 2 H) 3.79 - 3.84 (m, 2 H)
ESI (+) MS mil 340 (MH+)
ESI (+) HRMS calcd for C17H13F4N02+ Na+: 362.0774; found 362.0771 2-Fluoro-4-[2-(4-fluoro-benzyloxy)-ethoxy]-benzonitrile
1H-NMR (400 MHz), δ (ppm, DMSO-de): 7.82 (t, J=8.43 Hz, 1 H) 7.31 - 7.40 (m, 2 H) 7.13 - 7.23 (m, 3 H) 6.99 (dd, J=8.79, 2.20 Hz, 1 H) 4.53 (s, 2 H) 4.25 - 4.29 (m, 2 H) 3.75 - 3.79 (m, 2 H)
ESI (+) MS m/z 290 (MH+)
ESI (+) HRMS calcd for C16H13F2N02+ Na+: 312.0806; found 312.0812
2-Fluoro-4-[2-(3-trifluoromethyl-benzyloxy)-ethoxy]-benzonitrile
1H-NMR (400 MHz), δ (ppm, DMSO-de): 7.79 - 7.85 (m, 1 H) 7.55 - 7.67 (m, 4 H) 7.18 (dd, J=11.96, 2.32 Hz, 1 H) 6.99 (dd, J=8.79, 2.44 Hz, 1 H) 4.65 (s, 2 H) 4.29 - 4.34 (m, 2 H) 3.80 - 3.85 (m, 2 H)
ESI (+) MS mil 340 (MH+)
ESI (+) HRMS calcd for C17H13F4N02+ H+: 340.0955; found 340.0948
2-Fluoro-4-[2-(2-fluoro-benzyloxy)-ethoxy]-benzonitrile
1H-NMR (400 MHz), δ (ppm, DMSO-de): 7.82 (t, J=8.33 Hz, 1 H) 7.44 (td, J=7.65, 1.74 Hz, 1 H) 7.33 - 7.39 (m, 1 H) 7.15 - 7.23 (m, 3 H) 6.98 (dd, J=8.79, 2.20 Hz, 1 H) 4.60 (s, 2 H) 4.26 - 4.30 (m, 2 H) 3.79 - 3.83 (m, 2 H)
ESI (+) MS m/z 290 (MH+)
ESI (+) HRMS calcd for C16H13F2N02+ H+: 290.0987; found 290.0995
2-Fluoro-4-[2-(4-methoxy-benzyloxy)-ethoxy]-benzonitrile
1H-NMR (400 MHz), δ (ppm, DMSO-cie): 7.81 (dd, J=8.61 , 8.06 Hz, 1 H) 7.21 - 7.31 (m, 2 H) 7.18 (dd, J=11.90, 2.38 Hz, 1 H) 6.98 (dd, J=8.79, 2.38 Hz, 1 H) 6.85 - 6.91 (m, 2 H) 4.46 (s, 2 H) 4.21 - 4.31 (m, 2 H) 3.63 - 3.81 (m, 5 H) ESI (+) MS mil 302 (MH+)
ESI (+) HRMS calcd for C17H16FN03+ Na+: 324.1006; found 324.1008
2-Fluoro-4-[2-(pyridin-4-ylmethoxy)-ethoxy]-benzonitrile
1H-NMR (400 MHz), δ (ppm, DMSO-de): 8.50 - 8.54 (m, 2 H) 7.83 (t, J=8.33 Hz, 1 H) 7.32 (d, J=5.49 Hz, 2 H) 7.20 (dd, J=11.91 , 2.20 Hz, 1 H) 7.01 (dd, J=8.79, 2.38 Hz, 1 H) 4.61 (s, 2 H) 4.30 - 4.33 (m, 2 H) 3.81 - 3.84 (m, 2 H) ESI (+) MS m/z 273 (MH+)
ESI (+) HRMS calcd for C15H13FN202+ H+: 273.1034; found 273.1031
2-Fluoro-4-((E)-3-phenyl-allyloxy)-benzonitrile
1H-NMR (400 MHz), δ (ppm, DMSO-de): 7.84 (t, J=8.33 Hz, 1 H) 7.44 - 7.53 (m, 2 H) 7.36 (t, J=7.60 Hz, 2 H) 7.26 - 7.31 (m, 1 H) 7.23 (dd, J=11.90, 2.38 Hz, 1 H) 7.04 (dd, J=8.79, 2.38 Hz, 1 H) 6.80 (d, J=15.93 Hz, 1 H) 6.50 (dt, J=15.93, 6.04 Hz, 1 H) 4.86 (dd, J=6.04, 1.10 Hz, 2 H)
ESI (+) MS m/z 254 (MH+)
ESI (+) HRMS calcd for C16H12FNO+ Na+: 276.0795; found 276.0795
Preparation 11
6-(2-Benzyloxy-ethoxy)-1 H-indazol-3-ylamine
Scheme 2, Step K)
A mixture of 4-(2-benzyloxy-ethoxy)-2-fluoro-benzonitrile (8.79 g, 32.4 mmol) and hydrazine monohydrate (4.72 ml, 97.2 mmol) in n-butanol (15 ml) was stirred at 120°C for 8h. The reaction mixture was cooled to r.t., treated with 250 ml of water and stirred for 30 min. The precipitated solid was filtered, washed with water and dried in oven at 50°C under high vacuum, affording 9.0 g of the title compound as white crystals (yield: 98%).
1H-NMR (400 MHz), δ (ppm, DMSO-de): 11.13 (s, 1 H) 7.52 (d, J=8.67 Hz, 1 H) 7.33 - 7.38 (m, 4 H) 7.25 - 7.32 (m, 1 H) 6.64 (d, J=1.83 Hz, 1 H) 6.54 (dd, J=8.67, 2.07 Hz, 1 H) 5.26 (bs, 2 H) 4.57 (s, 2 H) 4.12 - 4.16 (m, 2 H) 3.77 - 3.80 (m, 2 H)
ESI (+) MS m/z 284 (MH+)
Operating in an analogous way, the following compounds were obtained:
6-[2-(4-Trifluoromethyl-benzyloxy)-ethoxy]-1 H-indazol-3-ylamine
1H-NMR (400 MHz), δ (ppm, DMSO-cie): 11.12 (s, 1 H) 7.72 (d, J=8.06 Hz, 2 H) 7.58 (d, J=8.06 Hz, 2 H) 7.52 (d, J=8.79 Hz, 1 H) 6.65 (d, J=1.83 Hz, 1 H) 6.55 (dd, J=8.70, 2.11 Hz, 1 H) 5.19 (s, 2 H) 4.68 (s, 2 H) 4.14 - 4.19 (m, 2 H) 3.81 - 3.85 (m, 2 H)
ESI (+) MS mil 352 (MH+)
ESI (+) HRMS calcd for G7H16F3N3O2+ H+: 352.1268; found 352.1278
6-[2-(4-Fluoro-benzyloxy)-ethoxy]-1 H-indazol-3-ylamine
1H-NMR (400 MHz), δ (ppm, DMSO-de): 11.12 (s, 1 H) 7.52 (d, J=8.79 Hz, 1 H) 7.39 (dd, J=8.52, 5.77 Hz, 2 H) 7.14 - 7.20 (m, 2 H) 6.64 (d, J=2.01 Hz, 1 H) 6.53 (dd, J=8.70, 2.11 Hz, 1 H) 5.19 (s, 2 H) 4.55 (s, 2 H) 4.10 - 4.15 (m, 2 H) 3.76 - 3.80 (m, 2 H)
ESI (+) MS mil 302 (MH+)
ESI (+) HRMS calcd for C16H16FN302+ H+: 302.1300; found 302.1306
6-[2-(3-Trifluoromethyl-benzyloxy)-ethoxy]-1 H-indazol-3-ylamine
1H-NMR (400 MHz), δ (ppm, DMSO-de): 11.13 (s, 1 H) 7.71 (s, 1 H) 7.57 - 7.69 (m, 3 H) 7.48 - 7.54 (m, 1 H) 6.65 (d, J=1.95 Hz, 1 H) 6.54 (dd, J=8.79, 2.07 Hz, 1 H) 5.21 (br. s., 2 H) 4.68 (s, 2 H) 4.13 - 4.19 (m, 2 H) 3.80 - 3.87 (m, 2 H)
ESI (+) MS mil 352 (MH+)
ESI (+) HRMS calcd for G7H16F3N3O2+ H+: 352.1268; found 352.1274
6-[2-(2-Fluoro-benzyloxy)-ethoxy]-1 H-indazol-3-ylamine
1H-NMR (400 MHz), δ (ppm, DMSO-de): 11.12 (s, 1 H) 7.52 (d, J=8.79 Hz, 1 H) 7.45 - 7.50 (m, 1 H) 7.33 - 7.40 (m, 1 H) 7.16 - 7.24 (m, 2 H) 6.64 (d, J=1.83 Hz, 1 H) 6.53 (dd, J=8.70, 2.11 Hz, 1 H) 5.19 (s, 2 H) 4.63 (s, 2 H) 4.13 (dd, J=5.40, 3.75 Hz, 2 H) 3.82 (dd, J=5.31 , 3.85 Hz, 2 H)
ESI (+) MS mil 302 (MH+)
ESI (+) HRMS calcd for C16H16FN302+ H+: 302.1300; found 302.1302
6-[2-(4-Methoxy-benzyloxy)-ethoxy]-1 H-indazol-3-ylamine
1H-NMR (400 MHz), δ (ppm, DMSO-de): 11.11 (s, 1 H) 7.51 (d, J=8.79 Hz, 1 H) 7.27 (d, J=8.61 Hz, 2 H) 6.91 (d, J=8.61 Hz, 2 H) 6.63 (d, J=1.65 Hz, 1 H) 6.53 (dd, J=8.79, 1.83 Hz, 1 H) 5.19 (s, 2 H) 4.48 (s, 2 H) 4.09 - 4.13 (m, 2 H) 3.68 - 3.78 (m, 5 H)
ESI (+) MS m/z 314 (MH+)
ESI (+) HRMS calcd for C17H19N303+ H+: 314.1499; found 314.1502 6-[2-(Pyridin-4-ylmethoxy)-ethoxy]-1 H-indazol-3-ylamine
1H-NMR (400 MHz), δ (ppm, DMSO-cie): 11.13 (s, 1 H) 8.51 - 8.55 (m, 2 H) 7.52 (d, J=8.79 Hz, 1 H) 7.35 (d, J=5.86
Hz, 2 H) 6.65 (d, J=1.83 Hz, 1 H) 6.55 (dd, J=8.61 , 2.01 Hz, 1 H) 5.20 (s, 2 H) 4.63 (s, 2 H) 4.15 - 4.19 (m, 2 H) 3.81
- 3.88 (m, 2 H)
ESI (+) MS m/z 285 (MH+)
ESI (+) HRMS calcd for C15H16N402+ H+: 285.1346; found 285.1339
6-((E)-3-Phenyl-allyloxy)-1 H-indazol-3-ylamine
1H-NMR (400 MHz), δ (ppm, DMSO-de): 11.13 (s, 1 H) 7.54 (d, J=8.61 Hz, 1 H) 7.49 (d, J=7.14 Hz, 2 H) 7.35 (t, J=7.51 Hz, 2 H) 7.24 - 7.30 (m, 1 H) 6.77 (d, J=15.93 Hz, 1 H) 6.70 (s, 1 H) 6.57 - 6.60 (m, 1 H) 6.53 (dt, J=15.93, 5.68 Hz, 1 H) 5.20 (s, 2 H) 4.73 (d, J=5.31 Hz, 2 H)
ESI (+) MS m/z 266 (MH+)
ESI (+) HRMS calcd for C16H15N30+ H+: 266.1288; found 266.1286
Example 2
N-[6-(2-Benzyloxy-ethoxy)-1H-indazol-3-yl] -(4-methyl-piperazin-1-yl)-benzamide (Cpd. 11)
Scheme 2, Step L)
Figure imgf000039_0001
A stirred suspension of 4-(4-methyl-piperazin-1-yl)-benzoyl chloride hydrochloride, prepared from 4.55 g (20.7 mmol) of 4-(4-methyl-piperazin-1-yl)-benzoic acid as described above in preparation 3, in dry pyridine (50 ml) was treated dropwise, at 0°C, under argon atmosphere, with a solution of 6-(2-benzyloxy-ethoxy)-1 H-indazol-3-ylamine (5.31 g, 18.8 mmol) in 80 ml of dry pyridine. The reaction mixture was allowed to warm to r.t. under stirring overnight, then concentrated to 30 ml by rotary evaporation, poured into 500 ml of saturated solution of NaHC03 and extracted with 300 + 100 ml of DCM. The organic layers were combined, dried over sodium sulfate and evaporated to dryness. The crude residue was treated with 200 ml of EtOAc and stirred at reflux for 2h. After cooling to r.t. the solid was filtered, washed with EtOAc and dried in oven at 50°C under high vacuum to afford 5.23 g (yield: 57%) of the title compound as a white solid.
1H-NMR (400 MHz), δ (ppm, DMSO-cie): 12.44 (s, 1 H) 10.38 (s, 1 H) 7.95 (d, J=9.03 Hz, 2 H) 7.59 (d, J=8.91 Hz, 1 H) 7.33 - 7.41 (m, 4 H) 7.25 - 7.32 (m, 1 H) 7.00 (d, J=9.15 Hz, 2 H) 6.85 (d, J=2.08 Hz, 1 H) 6.71 (dd, J=8.91 , 2.20 Hz, 1 H) 4.58 (s, 2 H) 4.15 - 4.25 (m, 2 H) 3.70 - 3.88 (m, 2 H) 3.25 - 3.35 (m, 4 H) 2.42 - 2.48 (m, 4 H) 2.23 (s, 3 H) ESI (+) MS m/z 486 (MH+)
ESI (+) HRMS calcd for C28N503H3i + H+: 486.2500; found 486.2501
Operating in an analogous way, the following compounds were obtained: N-[6-(2-Benzyloxy-ethoxy)-1H-indazol-3- -4-dimethylaminomethyl-benzamide (Cpd. 21)
Figure imgf000040_0001
1H-NMR (400 MHz), δ (ppm, DMSO-cie): 12.51 (s, 1 H) 10.65 (s, 1 H) 8.02 (d, J=8.30 Hz, 2 H) 7.61 (d, J=8.91 Hz, 1 H) 7.43 (d, J=8.42 Hz, 2 H) 7.33 - 7.38 (m, 4 H) 7.26 - 7.32 (m, 1 H) 6.87 (d, J=1.95 Hz, 1 H) 6.73 (dd, J=8.91 , 2.08 Hz, 1 H) 4.59 (s, 2 H) 4.19 - 4.23 (m, 2 H) 3.79 - 3.84 (m, 2 H) 3.47 (s, 2 H) 2.17 (s, 6 H)
ESI (+) MS m/z 445 (MH+)
ESI (+) HRMS calcd for C26N4O3H28 + H+: 445.2234; found 445.2224
N-[6-(2-Benzyloxy-ethoxy)-1H-indazol- -yl]-4-(1-methyl-piperidin-4-yloxy)-benzamide (Cpd. 22)
Figure imgf000040_0002
1H-NMR (400 MHz), δ (ppm, DMSO-cie): 12.48 (s, 1 H) 10.52 (s, 1 H) 7.99 - 8.04 (m, 2 H) 7.59 (d, J=8.91 Hz, 1 H) 7.33 - 7.40 (m, 4 H) 7.26 - 7.31 (m, 1 H) 7.04 - 7.09 (m, 2 H) 6.86 (d, J=1.95 Hz, 1 H) 6.72 (dd, J=8.91 , 2.20 Hz, 1 H) 4.58 (s, 2 H) 4.44 - 4.54 (m, 1 H) 4.17 - 4.23 (m, 2 H) 3.79 - 3.86 (m, 2 H) 2.57 - 2.67 (m, 2 H) 2.14 - 2.25 (m, 5 H) 1.91 - 2.02 (m, 2 H) 1.59 - 1.75 (m, 2 H)
ESI (+) MS mil 501 (MH+)
ESI (+) HRMS calcd for C29N4O4H32 + H+: 501.2497; found 501.2482
N-[6-(2-Benzyloxy-ethoxy)-1H-indazol-3-yl]-3-(4-methyl-piperazin-1-yl)-benzamide hydrochloride (Cpd. 29)
Figure imgf000040_0003
1H-NMR (400 MHz), δ (ppm, DMSO-cie): 12.56 (br. s., 1 H) 10.69 (s, 1 H) 10.17 (br. s., 1 H) 7.66 (br. s, 1 H) 7.61 (d, J=8.79 Hz, 1 H) 7.55 (d, J=7.69 Hz, 1 H) 7.41 (dd, J=8.43, 7.69 Hz, 1 H) 7.32 - 7.39 (m, 4 H) 7.27 - 7.31 (m, 1 H) 7.24 (dd, J=8.43, 1.83 Hz, 1 H) 6.88 (d, J=1.83 Hz, 1 H) 6.73 (dd, J=8.79 Hz, 2.01 Hz, 1 H) 4.8 (s, 2 H) 4.16 - 4.24 (m, 2 H) 3.91 - 4.02 (m, 2 H) 3.78 - 3.84 (m, 2 H) 3.50 - 3.56 (m, 2 H) 3.01 - 3.23 (m, 4 H) 2.85 (d, J=4.21 Hz, 3 H) ESI (+) MS m/z 486 (MH+) ESI (+) HRMS calcd for C28H3iN503 + H+: 486.2500; found 486.2514
N-[6-(2-Benzyloxy-ethoxy)-1H-indazol-3- -2-fluoro -(4-methyl-piperazin-1-yl)-benzamide (Cpd. 38)
Figure imgf000041_0001
1H-NMR (400 MHz), δ (ppm, DMSO-de): 12.45 (s, 1 H) 10.06 (d, J=3.66 Hz, 1 H) 7.61 - 7.69 (m, 2 H) 7.34 - 7.39 (m, 4 H) 7.27 - 7.31 (m, 1 H) 6.78 - 6.86 (m, 3 H) 6.72 (dd, J=8.88, 2.11 Hz, 1 H) 4.58 (s, 2 H) 4.18 - 4.21 (m, 2 H) 3.79 - 3.84 (m, 2 H) 3.29 - 3.33 (m, 4 H) 2.40 - 2.45 (m, 4 H) 2.22 (s, 3 H)
ESI (+) MS mil 504 (MH+)
ESI (+) HRMS calcd for C28H3oFN503+ H+: 504.2406; found 504.2383
4-(4-Methyl^iperazin-1-yl)-N-{6-[2-(4-trifl romethyl-benzyloxy)-ethoxy]-1 H-indazol-3-yl}-benzamide (Cpd. 33)
Figure imgf000041_0002
1H-NMR (400 MHz), δ (ppm, DMSO-cie): 12.45 (s, 1 H) 10.38 (s, 1 H) 7.95 (d, J=8.97 Hz, 2 H) 7.72 (d, J=8.06 Hz, 2 H) 7.57 - 7.61 (m, 3 H) 7.00 (d, J=8.97 Hz, 2 H) 6.86 (d, J=1.83 Hz, 1 H) 6.72 (dd, J=8.88, 2.11 Hz, 1 H) 4.70 (s, 2 H) 4.20 - 4.25 (m, 2 H) 3.82 - 3.89 (m, 2 H) 3.27 - 3.35 (m, 4 H) 2.40 - 2.47 (m, 4 H) 2.23 (s, 3 H)
ESI (+) MS mil 554 (MH+)
ESI (+) HRMS calcd for C29H30F3N5O3+ H+: 554.2374; found 554.2389
N-{6-[2-(4-Fluoro-benzyloxy)-ethoxy]-1 -indazol-3-yl} -(4-methyl-piperazin-1-yl)-benzamide (Cpd. 32)
Figure imgf000041_0003
1H-NMR (400 MHz), δ (ppm, DMSO-cie): 12.44 (s, 1 H) 10.38 (s, 1 H) 7.95 (d, J=8.97 Hz, 2 H) 7.59 (d, J=8.79 Hz, 1 H) 7.37 - 7.44 (m, 2 H) 7.15 - 7.21 (m, 2 H) 7.00 (d, J=8.97 Hz, 2 H) 6.85 (d, J=2.01 Hz, 1 H) 6.71 (dd, J=8.88, 2.11 Hz, 1 H) 4.56 (s, 2 H) 4.18 - 4.22 (m, 2 H) 3.79 - 3.82 (m, 2 H) 3.27 - 3.37 (m, 4 H) 2.43 - 2.47 (m, 4 H) 2.23 (s, 3 H) ESI (+) MS m/z 504 (MH+)
ESI (+) HRMS calcd for C28H3oFN503+ H+: 504.2406; found 504.2414
4-(4-Methyl^iperazin-1-yl)-N-{6-[2-(3-triflu^ (Cpd. 34)
Figure imgf000042_0001
1H-NMR (400 MHz), δ (ppm, DMSO-cie): 12.45 (s, 1 H) 10.38 (s, 1 H) 7.95 (d, J=9.03 Hz, 2 H) 7.72 (s, 1 H) 7.63 - 7.70 (m, 2 H) 7.56 - 7.63 (m, 2 H) 7.00 (d, J=9.15 Hz, 2 H) 6.86 (d, J=2.08 Hz, 1 H) 6.71 (dd, J=8.91 , 2.07 Hz, 1 H) 4.70 (s, 2 H) 4.20 - 4.24 (m, 2 H) 3.83 - 3.89 (m, 2 H) 3.24 - 3.40 (m, 4 H) 2.42 - 2.48 (m, 4 H) 2.23 (s, 3 H)
ESI (+) MS m/z 554 (MH+)
ESI (+) HRMS calcd for C29H3oF3N503+ H+: 554.2374; found 554.2371
N-{6-[2-(2-Fluoro-benzyloxy)-ethoxy]-1 H-indazol-3-yl} -(4-methyl-piperazin-1-yl)-benzamide (Cpd. 30)
Figure imgf000042_0002
1H-NMR (400 MHz), δ (ppm, DMSO-cie): 12.44 (s, 1 H) 10.38 (s, 1 H) 7.95 (d, J=8.97 Hz, 2 H) 7.58 (d, J=8.79 Hz, 1 H) 7.46 - 7.51 (m, 1 H) 7.35 - 7.40 (m, 1 H) 7.16 - 7.25 (m, 2 H) 7.00 (d, J=9.16 Hz, 2 H) 6.85 (d, J=1.83 Hz, 1 H) 6.70 (dd, J=8.88, 2.11 Hz, 1 H) 4.64 (s, 2 H) 4.18 - 4.21 (m, 2 H) 3.82 - 3.87 (m, 2 H) 3.22 - 3.31 (m, 4 H) 2.40 - 2.48 (m, 4 H) 2.23 (s, 3 H)
ESI (+) MS m/z 504 (MH+)
ESI (+) HRMS calcd for C28H3oFN503+ H+: 504.2406; found 504.2409
N-{6-[2-(4-Methoxy-benzyloxy)-ethoxy]-1 H-indazol-3-yl} -(4-methyl-piperazin-1-yl)-benzamide (Cpd. 40)
Figure imgf000042_0003
1H-NMR (400 MHz), δ (ppm, DMSO-cie): 12.44 (s, 1 H) 10.38 (s, 1 H) 7.95 (d, J=8.97 Hz, 2 H) 7.59 (d, J=8.97 Hz, 1 H) 7.28 (d, J=8.61 Hz, 2 H) 7.00 (d, J=8.97 Hz, 2 H) 6.85 - 6.93 (m, 2 H) 6.84 (d, J=2.01 Hz, 1 H) 6.70 (dd, J=8.88, 2.11 Hz, 1 H) 4.50 (s, 2 H) 4.12 - 4.24 (m, 2 H) 3.76 - 3.80 (m, 2 H) 3.74 (s, 3 H) 3.27 - 3.34 (m, 4 H) 2.42 - 2.47 (m, 4 H) 2.23 (s, 3 H)
ESI (+) MS m/z 516 (MH+)
ESI (+) HRMS calcd for C29H33N504+ H+: 516.2606; found 516.2617
N-[6-(2-Benzyloxy-ethoxy)-1H-indazol- -yl]-2,4-bis-(4-methyl-piperazin-1-yl)-benzamide (Cpd. 43)
Figure imgf000043_0001
1H-NMR (400 MHz), δ (ppm, DMSO-de): 12.65 (s, 1 H) 12.41 (s, 1 H) 8.01 - 8.05 (m, 1 H) 7.96 - 8.00 (m, 1 H) 7.34 - 7.39 (m, 4 H) 7.26 - 7.32 (m, 1 H) 6.84 - 6.89 (m, 2 H) 6.81 - 6.83 (m, 1 H) 6.68 - 6.73 (m, 1 H) 4.58 (s, 2 H) 4.17 - 4.21 (m, 2 H) 3.79 - 3.83 (m, 2 H) 3.27 - 3.32 (m, 4 H) 3.01 - 3.06 (m, 4 H) 2.56 - 2.71 (m, 4 H) 2.41 - 2.48 (m, 4 H) 2.25 (s, 3 H) 2.23 (s, 3H)
ESI (+) MS mil 584 (MH+)
ESI (+) HRMS calcd for C33H41 N7O3+ H+: 584.3344; found 584.3340
4-(4-Methyl^iperazin-1-yl)-N-{6-[2-(pyrid -ylmethoxy)-ethoxy]-1 H-indazol-3-yl}-benzamide (Cpd. 35)
Figure imgf000043_0002
1H-NMR (400 MHz), δ (ppm, DMSO-cie): 12.45 (s, 1 H) 10.38 (s, 1 H) 8.53 (d, J=4.58 Hz, 2 H) 7.95 (d, J=8.06 Hz, 2 H) 7.60 (d, J=8.97 Hz, 1 H) 7.35 (d, J=4.58 Hz, 2 H) 7.00 (d, J=8.24 Hz, 2 H) 6.87 (s, 1 H) 6.72 (d, J=8.97 Hz, 1 H) 4.65 (s, 2 H) 4.21 - 4.25 (m, 2 H) 3.85 - 3.88 (m, 2 H) 3.27 - 3.37 (m, 4 H) 2.43 - 2.48 (m, 4 H) 2.23 (s, 3 H)
ESI (+) MS m/z 487 (MH+)
ESI (+) HRMS calcd for C27H30N6O3+ H+: 487.2452; found 487.2450
4-(4-Methyl-piperazin-1-yl)-N-[6-((E)-3-phenyl-allyloxy)-1 H-indazol-3-yl]-benzamide (Cpd. 39)
Figure imgf000044_0001
1H-NMR (400 MHz), δ (ppm, DMSO-cie): 12.45 (s, 1 H) 10.39 (s, 1 H) 7.95 (d, J=8.97 Hz, 2 H) 7.61 (d, J=8.97 Hz, 1 H) 7.48 - 7.52 (m, 2 H) 7.36 (t, J=7.69 Hz, 2 H) 7.26 - 7.31 (m, 1 H) 7.00 (d, J=8.97 Hz, 2 H) 6.91 (d, J=1.83 Hz, 1 H) 6.81 (d, J=16.12 Hz, 1 H) 6.75 (dd, J=8.97, 2.20 Hz, 1 H) 6.56 (dt, J=15.98, 5.75 Hz, 1 H) 4.80 (d, J=5.13 Hz, 2 H) 3.28 - 3.32 (m, 4 H) 2.43 - 2.47 (m, 4 H) 2.23 (s, 3 H)
ESI (+) MS m/z 468 (MH+)
ESI (+) HRMS calcd for C28H29N502+ H+: 468.2394; found 468.2394
Preparation 12
N-[6-(2-Benzyloxy-ethoxy)-1H-indazol-3-yl]-4-bromo-benzamide
Scheme 2, Step M)
To a stirred solution of 6-(2-benzyloxy-ethoxy)-1 H-indazol-3-ylamine (3.0 g, 10.6 mmol) in 30 ml of dry pyridine, at 0°C, under argon atmosphere, was added portionwise 4-bromo-benzoyl chloride (2.32 g, 10.6 mmol). The reaction mixture was allowed to warm to r.t. under stirring overnight then evaporated to dryness. The residue was treated with 50 ml of MeOH and 25 ml of 2N NaOH and stirred at r.t. for 1 h. The mixture was concentrated to ca. 10 ml by rotary evaporation, diluted with 200 ml of water, stirred for 15 min at r.t. and the suspended solid filtered and washed with water. After drying under vacuum, the crude solid was purified by chromatography (Biotage SP1 Flash Purification system) on a silica gel cartridge (Biotage SNAP 100 g) using DCM as eluant A and DCM / MeOH 9:1 as eluant B. Elution with a gradient from A / B 100:0 to 70:30 over 25 CV gave a pink solid that was triturated with EtOAc (50 ml), filtered, washed with EtOAc and dried affording 3.01 g (yield: 61 %) of the title compound as a whitish solid.
1H-NMR (400 MHz), δ (ppm, DMSO-cie): 12.54 (s, 1 H) 10.81 (s, 1 H) 7.99 (d, J=8.54 Hz, 2 H) 7.75 (d, J=8.67 Hz, 2 H) 7.62 (d, J=8.91 Hz, 1 H) 7.21 - 7.42 (m, 5 H) 6.87 (d, J=1.95 Hz, 1 H) 6.73 (dd, J=8.97, 2.14 Hz, 1 H) 4.58 (s, 2 H) 4.11 - 4.29 (m, 2 H) 3.71 - 3.98 (m, 2 H)
ESI (+) MS m/z 467 (MH+)
Preparation 13
2-(4-trifluoromethyl-benzyloxy)-ethanol
Under argon atmosphere, at r.t., sodium hydride (60% dispersion in mineral oil, 1.92 g, 48 mmol) was stirred with n- hexane (ca. 10 ml). The hexane/mineral oil solution was drawn off and discarded and the left sodium hydride was treated with 20 ml of dry THF. Ethylene glycol (17.8 ml, 320 mmol) was then slowly dropped at r.t. (caution: hydrogen evolution) and the mixture stirred at r.t. for 1.5 hours. After heating up to reflux (80°C oil bath) a solution of 1- bromomethyl-4-trifluoromethyl-benzene (7.6 g, 32 mmol) in 20 ml of dry THF was added and the reaction mixture stirred at reflux for 2.5 hours. After cooling to r.t., 100 ml of ammonium chloride saturated solution was added. The organic layer was separated, washed with 50 ml of water, dried over sodium sulfate and evaporated to dryness. The crude residue was purified by flash chromatography (Biotage SP1 Flash Purification system) on a silica gel cartridge (Varian SF40-120g) using n-hexane as eluant A and EtOAc as eluant B. Elution with a gradient from A/B 75:25 to 70:30 over 2 CV then from 70:30 to 0:100 over 2 CV then 100% of B afforded 5.9 g (yield: 84%) of the title compound as a yellow oil (yield: 93%).
1H-NMR (400 MHz), δ (ppm, DMSO-cie): 7.71 (d, J=8.06 Hz, 2 H) 7.56 (d, J=7.88 Hz, 2 H) 4.66 (t, J=5.49 Hz, 1 H) 4.60 (s, 2 H) 3.56 (q, J=5.31 Hz, 2 H) 3.48 - 3.51 (m, 2 H)
ESI (+) MS m/z 221 (MH+)
ESI (+) HRMS calcd for C1oH11F302 + Na+: 243.0603; found 243.0594
Operating in an analogous way, the following compounds were obtained:
2-(4-Fluoro-benzyloxy)-ethanol
1H-NMR (400 MHz), δ (ppm, DMSO-cie): 7.37 (dd, J=8.43, 5.86 Hz, 2 H) 7.16 (t, J=8.88 Hz, 2 H) 4.62 (t, J=5.49 Hz, 1 H) 4.46 (s, 2 H) 3.53 (q, J=5.19 Hz, 2 H) 3.41 - 3.47 (m, 2 H)
ESI (+) MS m/z 171 (MH+)
ESI (+) HRMS calcd for C9H11 FO2+ Na+: 193.0635; found 193.0635
2-(3-Trifluoromethyl-benzyloxy)-ethanol
1H-NMR (400 MHz), δ (ppm, DMSO-de): 7.55 - 7.72 (m, 4 H) 4.66 (t, J=5.49 Hz, 1 H) 4.59 (s, 2 H) 3.53 - 3.59 (m, 2 H) 3.47 - 3.52 (m, 2 H)
ESI (+) MS m/z 221 (MH+)
2-(2-Fluoro-benzyloxy)-ethanol
1H-NMR (400 MHz), δ (ppm, DMSO-de): 7.47 (td, J=7.51 , 1.65 Hz, 1 H) 7.36 (tdd, J=7.76, 7.76, 5.63, 1.74 Hz, 1 H) 7.14 - 7.21 (m, 2 H) 4.63 (t, J=5.49 Hz, 1 H) 4.54 (s, 2 H) 3.51 - 3.55 (m, 2 H) 3.46 - 3.50 (m, 2 H)
ESI (+) MS m/z 171 (MH+)
ESI (+) HRMS calcd for C9H11 FO2+ Na+: 193.0635; found 193.0635
2-(4-Methoxy-benzyloxy)-ethanol
1H-NMR (400 MHz), δ (ppm, DMSO-cie): 7.25 (d, J=8.79 Hz, 2 H) 6.90 (d, J=8.61 Hz, 2 H) 4.58 (t, J=5.59 Hz, 1 H) 4.40 (s, 2 H) 3.74 (s, 3 H) 3.51 (q, J=5.31 Hz, 2 H) 3.39 - 3.43 (m, 2 H)
ESI (+) MS m/z 183 (MH+)
ESI (+) HRMS calcd for C10H14O3+ Na+: 205.0835; found 205.0835
2-(Pyridin-4-ylmethoxy)-ethanol
1H-NMR (400 MHz), δ (ppm, DMSO-de): 8.51 - 8.54 (m, 2 H) 7.32 - 7.36 (m, 2 H) 4.68 (t, J=5.49 Hz, 1 H) 4.55 (s, 2 H) 3.57 (q, J=5.25 Hz, 2 H) 3.46 - 3.52 (m, 2 H)
ESI (+) MS m/z 154 (MH+)
ESI (+) HRMS calcd for CeHiiNO_+ H+: 154.0863; found 154.0857
Preparation 14
Methanesulfonic acid 2-(4-trifluoromethyl-benzyloxy)-ethyl ester To a solution of 2-(4-trifluoromethyl-benzyloxy)-ethanol (1.0 g, 4.5 mmol) in dry DCM (20 ml) and DIPEA (2.36 ml, 13.5 mmol), at 0°C, under argon atmosphere, was added methanesulfonyl chloride (421 μΙ, 5.4 mmol). The reaction mixture was stirred at 0°C for 10 minutes, then the ice-bath removed and the stirring continued for 2 hours at r.t.. The mixture was then diluted with 70 ml of DCM and washed with 50 ml of NaHC03 saturated solution, 100 ml of water, 100 ml of 2N HCI and 100 ml of water, dried over sodium sulfate and evaporated to dryness affording 1.35 g (yield: quantitative) of methanesulfonic acid 2-(4-trifluoromethyl-benzyloxy)-ethyl ester as a brown oil that was used as such for the next step without further purification.
1H-NMR (400 MHz), δ (ppm, DMSO-de): 7.73 (d, J=8.06 Hz, 2 H) 7.57 (d, J=7.88 Hz, 2 H) 4.64 (s, 2 H) 4.35 - 4.42 (m, 2 H) 3.71 - 3.78 (m, 2 H) 3.18 (s, 3 H)
ESI (+) MS m/z 299 (MH+)
ESI (+) HRMS calcd for C11H13F304S+ Na+: 321.0379; found 321.0380
Operating in an analogous way, the following compounds were obtained:
Methanesulfonic acid 2-(4-fluoro-benzyloxy)-ethyl ester
1H-NMR (400 MHz), δ (ppm, DMSO-de): 7.30 - 7.44 (m, 2 H) 7.12 - 7.23 (m, 2 H) 4.51 (s, 2 H) 4.29 - 4.38 (m, 2 H) 3.63 - 3.75 (m, 2 H) 3.17 (s, 3 H)
ESI (+) MS m/z 249 (MH+)
ESI (+) HRMS calcd for C11H13F304 S+ Na+: 271.0411 ; found 271.0412
Methanesulfonic acid 2-(3-trifluoromethyl-benzyloxy)-ethyl ester
1H-NMR (400 MHz), δ (ppm, DMSO-de): 7.55 - 7.72 (m, 4 H) 4.64 (s, 2 H) 4.34 - 4.42 (m, 2 H) 3.71 - 3.78 (m, 2 H) 3.18 (s, 3 H)
ESI (+) MS m/z 299 (MH+)
Methanesulfonic acid 2-(2-fluoro-benzyloxy)-ethyl ester
1H-NMR (400 MHz), δ (ppm, DMSO-de): 7.44 - 7.48 (m, 1 H) 7.34 - 7.41 (m, 1 H) 7.16 - 7.25 (m, 2 H) 4.59 (s, 2 H) 4.34 - 4.36 (m, 2 H) 3.71 - 3.75 (m, 2 H) 3.16 (s, 3 H)
ESI (+) MS m/z 249 (MH+)
ESI (+) HRMS calcd for CnH13F304S+ Na+: 271.0411 ; found 271.0411
Methanesulfonic acid 2-(4-methoxy-benzyloxy)-ethyl ester
1H-NMR (400 MHz), δ (ppm, DMSO-de): 7.26 (d, J=8.79 Hz, 2 H) 6.76 - 7.00 (m, 2 H) 4.45 (s, 2 H) 4.28 - 4.35 (m, 2 H) 3.74 (s, 3 H) 3.60 - 3.68 (m, 2 H) 3.16 (s, 3 H)
ESI (+) MS m/z 261 (MH+)
ESI (+) HRMS calcd for CnH1605S+ Na+: 283.0610; found 283.0614
Methanesulfonic acid 2-(pyridin-4-ylmethoxy)-ethyl ester
1H-NMR (400 MHz), δ (ppm, DMSO-de): 8.51 - 8.57 (m, 2 H) 7.34 (d, J=5.86 Hz, 2 H) 4.60 (s, 2 H) 4.36 - 4.41 (m, 2 H) 3.72 - 3.78 (m, 2 H) 3.19 (s, 3 H)
ESI (+) MS m/z 232 (MH+)
ESI (+) HRMS calcd for C9H13N04S+ H+: 232.0638; found 232.0636
Example 3 N-[6-(2-Benzyloxy-ethoxy)-1H-indazol-3-yl] -(4-dimethylamino^iperidin-1-yl)-benzam (Cpd. 15)
Scheme 2, Step N)
Figure imgf000047_0001
A solution of N-[6-(2-benzyloxy-ethoxy)-1 H-indazol-3-yl]-4-bromo-benzamide (1.3 g, 2.79 mmol) and 4- dimethylamino-piperidine (1.18 ml, 8.37 mmol) in dry THF (20 ml) was degassed by three vacuum-argon atmosphere cycles and treated at r.t., under argon atmosphere, with Pd2(dba)3 (50 mg), 2-dicyclohexylphosphino-2'-(N,N- dimethylamino)biphenyl (50 mg) and 1 M LiHMDS in THF (22.3 ml, 22.3 mmol). The reaction mixture was heated to reflux and stirred for 15 min then cooled to r.t., poured into 200 ml of water and extracted with 200 ml of EtOAc. The organic layer was separated, dried over sodium sulfate and evaporated to dryness. The crude residue was purified by chromatography (Biotage SP1 Flash Purification system) on a silica gel cartridge (Varian SF40-120g) using DCM as eluant A and DCM / 7N NH3 in MeOH 10:1 as eluant B. Elution with a gradient from A / B 100:0 to 0:100 over 10 CV, followed by an isocratic elution with eluant B (5 CV), gave a yellow solid that was triturated with EtOAc (15 ml), filtered, washed with EtOAc and dried affording 1.04 g (yield: 73%) of the title compound as a white solid.
1H-NMR (400 MHz), δ (ppm, DMSO-cie): 12.44 (s, 1 H) 10.35 (s, 1 H) 7.85 - 8.01 (m, 2 H) 7.59 (d, J=8.91 Hz, 1 H) 7.33 - 7.40 (m, 4 H) 7.25 - 7.32 (m, 1 H) 6.93 - 7.06 (m, 2 H) 6.85 (d, J=2.07 Hz, 1 H) 6.71 (dd, J=8.91 , 2.08 Hz, 1 H) 4.58 (s, 2 H) 4.14 - 4.26 (m, 2 H) 3.86 - 3.98 (m, 2 H) 3.73 - 3.85 (m, 2 H) 2.74 - 2.90 (m, 2 H) 2.23 - 2.35 (m, 1 H) 2.19 (s, 6 H) 1.77 - 1.87 (m, 2 H) 1.35 - 1.50 (m, 2 H)
ESI (+) MS m/z 514 (MH+)
ESI (+) HRMS calcd for C3oN503H35 + H+: 514.2813; found 514.2817
Operating in an analogous way, the following compounds were obtained:
N-[6-(2-Benzyloxy-ethoxy)-1H-indazol- -yl] -[(2-dimethylamino-ethyl)-methyl-amin (Cpd. 16)
Figure imgf000047_0002
1H-NMR (400 MHz), δ (ppm, DMSO-d6): 12.41 (s, 1 H) 10.27 (s, 1 H) 7.89 - 7.96 (m, 2 H) 7.59 (d, J=8.91 Hz, 1 H) 7.33 - 7.38 (m, 4 H) 7.25 - 7.32 (m, 1 H) 6.85 (d, J=2.07 Hz, 1 H) 6.68 - 6.76 (m, 3 H) 4.58 (s, 2 H) 4.17 - 4.22 (m, 2 H) 3.78 - 3.85 (m, 2 H) 3.50 (t, J=7.08 Hz, 2 H) 2.99 (s, 3 H) 2.40 (t, J=7.02 Hz, 2 H) 2.19 (s, 6 H) ESI (+) MS m/z 488 (MH+)
ESI (+) HRMS calcd for C28N503H33 + H+: 488.2656; found 488.2654
N-[6-(2-Benzyloxy-ethoxy)-1H-indazol-3-ylH- (Cpd. 17)
Figure imgf000048_0001
1H-NMR (400 MHz), δ (ppm, DMSO-de): 12.41 (s, 1 H) 10.26 (s, 1 H) 7.90 - 7.95 (m, 2 H) 7.59 (d, J=8.91 Hz, 1 H) 7.34 - 7.40 (m, 4 H) 7.27 - 7.32 (m, 1 H) 6.85 (d, J=2.07 Hz, 1 H) 6.72 - 6.77 (m, 2 H) 6.70 (dd, J=8.91 , 2.20 Hz, 1 H) 4.58 (s, 2 H) 4.17 - 4.23 (m, 2 H) 3.79 - 3.83 (m, 2 H) 3.43 (t, J=7.14 Hz, 2 H) 2.98 (s, 3 H) 2.23 (t, J=6.84 Hz, 2 H) 2.14 (s, 6 H) 1.61 - 1.71 (m, 2 H)
ESI (+) MS mil 502 (MH+)
ESI (+) HRMS calcd for C29N503H35 + H+: 502.2813; found 502.2794
4-{4-[6-(2-Benzyloxy-ethoxy)-1 H-indazol-3-ylcarbamoyl]-phenyl}-piperazine-1-carboxylic acid tert-butyl ester (Cpd. 18)
Figure imgf000048_0002
1H-NMR (400 MHz), δ (ppm, DMSO-cie): 12.45 (s, 1 H) 10.40 (s, 1 H) 7.95 - 7.99 (m, 2 H) 7.59 (d, J=9.03 Hz, 1 H) 7.34 - 7.38 (m, 4 H) 7.26 - 7.33 (m, 1 H) 6.98 - 7.05 (d, J=9.15 Hz, 2 H) 6.85 (d, J=2.20 Hz, 1 H) 6.71 (dd, J=8.91 , 2.07 Hz, 1 H) 4.58 (s, 2 H) 4.16 - 4.23 (m, 2 H) 3.79 - 3.86 (m, 2 H) 3.44 - 3.51 (m, 4 H) 3.27 - 3.32 (m, 4 H) 1.43 (s, 9 H)
ESI (+) MS mil 572 (MH+)
ESI (+) HRMS calcd for C32N505H37 + H+: 572.2868; found 572.2862
N-[6-(2-Benzyloxy-ethoxy)-1H-indazol-3-yl] -(1-meto (Cpd. 19)
Figure imgf000049_0001
1H-NMR (400 MHz), δ (ppm, DMSO-cie): 12.40 (s, 1 H) 10.18 (s, 1 H) 7.83 (d, J=8.91 Hz, 2 H) 7.58 (d, J=8.91 Hz, 1 H) 7.34 - 7.39 (m, 4 H) 7.25 - 7.30 (m, 1 H) 6.84 (d, J=2.07 Hz, 1 H) 6.69 (dd, J=8.91 , 2.20 Hz, 1 H) 6.62 (d, J=8.91 Hz, 2 H) 6.15 (d, J=7.81 Hz, 1 H) 4.58 (s, 2 H) 4.17 - 4.23 (m, 2 H) 3.78 - 3.84 (m, 2 H) 2.70 - 2.78 (m, 2 H) 2.17 (s, 3 H) 1.96 - 2.09 (m, 2 H) 1.83 - 1.94 (m, 2 H) 1.33 - 1.51 (m, 2 H)
ESI (+) MS mil 500 (MH+)
ESI (+) HRMS calcd for C29N503H33 + H+: 500.2656; found 500.2648
N-[6-(2-Benzyloxy-ethoxy)-1H-indazol-3-ylH-[m^ (Cpd. 23)
Figure imgf000049_0002
1H-NMR (400 MHz), δ (ppm, DMSO-cie): 12.42 (s, 1 H) 10.28 (s, 1 H) 7.93 (d, J=8.91 Hz, 2 H) 7.59 (d, J=8.91 Hz, 1 H) 7.33 - 7.39 (m, 4 H) 7.25 - 7.34 (m, 1 H) 6.80 - 6.87 (m, 3 H) 6.70 (dd, J=8.91 , 2.07 Hz, 1 H) 4.58 (s, 2 H) 4.17 - 4.23 (m, 2 H) 3.79 - 3.85 (m, 2 H) 3.66 - 3.79 (m, 1 H) 2.82 - 2.88 (m, 2 H) 2.82 (s, 3 H) 2.19 (s, 3 H) 2.00 - 2.11 (m, 2 H) 1.71 - 1.85 (m, 2 H) 1.55 - 1.64 (m, 2 H)
ESI (+) MS m/z 514 (MH+)
ESI (+) HRMS calcd for C3oN503H35 + H+: 514.2813; found 514.2792
N-[6-(2-Benzyloxy-ethoxy)-1H-indazol-3- -4-morpholin-4-y -benzamide (Cpd. 24)
Figure imgf000049_0003
1H-NMR (400 MHz), δ (ppm, DMSO-cie): 12.45 (s, 1 H) 10.40 (s, 1 H) 7.97 (d, J=9.03 Hz, 2 H) 7.59 (d, J=8.91 Hz, 1 H) 7.34 - 7.39 (m, 4 H) 7.25 - 7.33 (m, 1 H) 7.02 (d, J=9.03 Hz, 2 H) 6.85 (d, J=1.83 Hz, 1 H) 6.71 (dd, J=8.91 , 2.20 Hz, 1 H) 4.58 (s, 2 H) 4.16 - 4.23 (m, 2 H) 3.78 - 3.84 (m, 2 H) 3.71 - 3.77 (m, 4 H) 3.24 - 3.28 (m, 4 H)
ESI (+) MS m/z 473 (MH+)
ESI (+) HRMS calcd for C27N4O4H28 + H+: 473.2184; found 473.2169 N-[6-(2-Benzyloxy-ethoxy)-1H-indazol-3-yl] -(2-morpholin-4-yl-ethylamino)-benzamide hydrochloride (Cpd. 25)
Figure imgf000050_0001
1H-NMR (400 MHz), δ (ppm, DMSO-cie): 12.44 (bs, 1 H) 10.66 (bs, 1 H) 10.30 (s, 1 H) 7.91 (d, J=8.79 Hz, 2 H) 7.59 (d, J=8.91 Hz, 1 H) 7.33 - 7.39 (m, 4 H) 7.25 - 7.32 (m, 1 H) 6.85 (d, J=1.83 Hz, 1 H) 6.67 - 6.75 (m, 3 H) 4.58 (s, 2 H) 4.16 - 4.23 (m, 2 H) 3.90 - 4.04 (m, 2 H) 3.70 - 3.87 (m, 4 H) 3.54 - 3.62 (m, 2 H) 3.44 - 3.54 (m, 2 H) 3.24 - 3.33 (m, 2 H) 3.03 - 3.23 (m, 2 H)
ESI (+) MS m/z 516 (MH+)
ESI (+) HRMS calcd for C29N504H33 + H+: 516.2606; found 516.2584
N-[6-(2-Benzyloxy-ethoxy)-1H-indazol- -yl] -(tetrahydro^yran-4-ylamino)-benzamide (Cpd. 26)
Figure imgf000050_0002
1H-NMR (400 MHz), δ (ppm, DMSO-cie): 12.40 (s, 1 H) 10.20 (s, 1 H) 7.84 (d, J=8.67 Hz, 2 H) 7.58 (d, J=8.91 Hz, 1 H) 7.34 - 7.38 (m, 4 H) 7.26 - 7.31 (m, 1 H) 6.84 (d, J=1.71 Hz, 1 H) 6.70 (dd, J=8.91 , 1.83 Hz, 1 H) 6.65 (d, J=8.67 Hz, 2 H) 6.22 (d, J=7.81 Hz, 1 H) 4.58 (s, 2 H) 4.16 - 4.22 (m, 2 H) 3.84 - 3.92 (m, 2 H) 3.79 - 3.84 (m, 2 H) 3.50 - 3.63 (m, 1 H) 3.40 - 3.48 (m, 2 H) 1.85 - 1.94 (m, 2 H) 1.32 - 1.47 (m, 2 H)
ESI (+) MS m/z 487 (MH+)
ESI (+) HRMS calcd for C28N4O4H30 + H+: 487.2340; found 487.2340
N-[6-(2-Benzyloxy-ethoxy)-1H-indazol- -ylH-[(1-m^ (Cpd. 27)
Figure imgf000050_0003
1H-NMR (400 MHz), δ (ppm, DMSO-cie): 12.39 (s, 1 H) 10.18 (s, 1 H) 7.83 (d, J=8.67 Hz, 2 H) 7.58 (d, J=8.91 Hz, 1 H) 7.33 - 7.40 (m, 4 H) 7.25 - 7.30 (m, 1 H) 6.84 (d, J=1.83 Hz, 1 H) 6.70 (dd, J=8.91 , 2.07 Hz, 1 H) 6.61 (d, J=8.79 Hz, 2 H) 6.33 (t, J=5.68 Hz, 1 H) 4.58 (s, 2 H) 4.17 - 4.23 (m, 2 H) 3.78 - 3.85 (m, 2 H) 2.97 (t, J=6.16 Hz, 2 H) 2.72 - 2.80 (m, 2 H) 2.14 (s, 3 H) 1.76 - 1.86 (m, 2 H) 1.67 - 1.76 (m, 2 H) 1.42 - 1.59 (m, 1 H) 1.14 - 1.30 (m, 2 H) ESI (+) MS m/z 514 (MH+)
ESI (+) HRMS calcd for C3oN503H35 + H+: 514.2813; found 514.2797
N-[6-(2-Benzyloxy-ethoxy)-1H-indazol-3- l]-4-(3-pyrrolidin-1-yl-azetidin-1-yl)-benzamide (Cpd. 28)
Figure imgf000051_0001
1H-NMR (400 MHz), δ (ppm, DMSO-cie): 12.42 (s, 1 H) 10.30 (s, 1 H) 7.93 (d, J=8.67 Hz, 2 H) 7.59 (d, J=8.91 Hz, 1 H) 7.32 - 7.40 (m, 4 H) 7.26 - 7.30 (m, 1 H) 6.85 (d, J=1.95 Hz, 1 H) 6.70 (dd, J=8.91 , 2.07 Hz, 1 H) 6.45 (d, J=8.79 Hz, 2 H) 4.58 (s, 2 H) 4.10 - 4.27 (m, 2 H) 4.00 (t, J=7.38 Hz, 2 H) 3.80 - 3.87 (m, 2 H) 3.75 (dd, J=7.87, 4.94 Hz, 2 H) 3.41 - 3.49 (m, 1 H) 2.45 - 2.49 (m, 4 H) 1.66 - 1.82 (m, 4 H)
ESI (+) MS m/z 512 (MH+)
ESI (+) HRMS calcd for C3oN503H33 + H+: 512.2656; found 512.2650
Example 4
N-[6-(2-Benzyloxy-ethoxy)-1H-indazol-3-yl]-4-(4-methyl-4-oxy-piperazin-1-yl)-benzamide (Cpd. 14)
Conversion 7
Figure imgf000051_0002
A solution of N-[6-(2-benzyloxy-ethoxy)-1 H-indazol-3-yl]-4-(4-methyl-piperazin-1-yl)-benzamide (300 mg, 0.62 mmol) in DCM (5 ml) and MeOH (5 ml) was treated at r.t. with 3-chloroperbenzoic acid (107 mg, 0.62 mmol). After stirring for 2h at r.t. the precipitated solid was filtered, washed with few ml of DCM / MeOH 1 :1 and with MeOH and dried in vacuo affording 125 mg of the title compound (yield: 40%) as a white solid.
1H-NMR (400 MHz), δ (ppm, DMSO-d6): 12.47 (s, 1 H) 10.43 (s, 1 H) 7.95 - 8.02 (m, 2 H) 7.60 (d, J=9.08 Hz, 1 H) 7.25 - 7.40 (m, 5 H) 7.03 - 7.12 (m, 2 H) 6.86 (d, J=2.05 Hz, 1 H) 6.71 (dd, J=9.08, 2.05 Hz, 1 H) 4.59 (s, 2 H) 4.16 - 4.24 (m, 2 H) 3.79 - 3.86 (m, 2 H) 3.67 - 3.77 (m, 2 H) 3.43 - 3.63 (m, 4H) 3.12 (s, 3 H) 3.96 - 3.07 (m, 2 H) ESI (+) MS mil 502 (MH+) ESI (+) HRMS calcd for C28N504H3i + H+: 502.2449; found 502.2443
The title compound (120 mg) was then suspended in 10 ml of ethanol, treated with 2N HCI (0.5 ml) and stirred until a clear solution was obtained. Evaporation of the solvent, trituration with diethyl ether and drying in vacuo afforded 127 mg of the hydrochloride salt of the title compound as a white solid.
1H-NMR (400 MHz), δ (ppm, DMSO-cie): 12.55 (s, 1 H) 12.51 (bs, 1 H) 10.48 (s, 1 H) 8.02 (d, J=8.79 Hz, 2 H) 7.59 (d, J=8.91 Hz, 1 H) 7.33 - 7.41 (m, 4 H) 7.25 - 7.32 (m, 1 H) 7.13 (d, J=8.91 Hz, 2 H) 6.86 (d, J=1.59 Hz, 1 H) 6.72 (dd, J=8.85, 1.89 Hz, 1 H) 4.58 (s, 2 H) 4.14 - 4.26 (m, 2 H) 3.94 - 4.03 (m, 2 H) 3.74 - 3.89 (m, 6 H) 3.59 (s, 3 H) 3.40 - 3.50 (m, 2 H)
ESI (+) MS mil 502 (MH+)
ESI (+) HRMS calcd for C28N504H3i + H+: 502.2449; found 502.2438
Operating in an analogous way, the following compounds were obtained:
N-[6-(2-Benzyloxy-ethoxy)-1H-indazol-3-ylH^ (Cpd. 41)
Figure imgf000052_0001
1H-NMR (400 MHz), δ (ppm, DMSO-cie): 12.49 (s, 1 H) 10.32 (s, 1 H) 7.95 (d, J=9.03 Hz, 2 H) 7.59 (d, J=8.91 Hz, 1 H) 7.33 - 7.41 (m, 4 H) 7.24 - 7.33 (m, 1 H) 6.87 (d, J=9.15 Hz, 2 H) 6.85 (d, J=2.20 Hz, 1 H) 6.70 (dd, J=8.97, 2.14 Hz, 1 H) 4.58 (s, 2 H) 4.15 - 4.24 (m, 2 H) 3.93 - 4.06 (m, 1 H) 3.77 - 3.85 (m, 2 H) 3.45 - 3.56 (m, 2 H) 3.07 (s, 3 H) 3.03 (br. s., 1 H) 2.86 (s, 3 H) 2.43 - 2.55 (m, 3H) 1.48 (m, J=12.08 Hz, 2 H)
ESI (+) MS mil 530 (MH+)
ESI (+) HRMS calcd for C3oH35N504+ H+: 530.2762; found 530.2769
4-(4-Methyl -oxy^iperazin-1-yl)-N-{6-[2-(4-trifluoromethyl-benzyloxy)-ethoxy]-1 H-indazol-3-yl}-benz^ hydrochloride (Cpd. 42)
Figure imgf000052_0002
1H-NMR (400 MHz), δ (ppm, DMSO-cie): 12.55 (s, 1 H) 12.50 (br. s., 1 H) 10.49 (s, 1 H) 8.03 (d, J=8.97 Hz, 2 H) 7.72 (d, J=8.06 Hz, 2 H) 7.57 - 7.61 (m, 3 H) 7.10 - 7.15 (m, 2 H) 6.87 (d, J=1.83 Hz, 1 H) 6.73 (dd, J=8.88, 2.11 Hz, 1 H) 4.70 (s, 2 H) 4.21 - 4.25 (m, 2 H) 3.99 (d, J=14.29 Hz, 2 H) 3.75 - 3.89 (m, 6 H) 3.59 (s, 3 H) 3.40 - 3.48 (m, 2H) ESI (+) MS m/z 570 (MH+)
ESI (+) HRMS calcd for C29H3oF3N504+ H+: 570.2323; found 570.2330
Example 5
N-[6-(2-Benzyloxy-ethoxy)-1H-indazol-3-yl]-4-piperazin-1-yl-benzamide (Cpd. 20)
Conversion 8
Figure imgf000053_0001
A solution of 4-{4-[6-(2-benzyloxy-ethoxy)-1 H-indazol-3-ylcarbamoyl]-phenyl}-piperazine-1-carboxylic acid tert-butyl ester (180 mg, 0.31 mmol) in 1 ,4-dioxane (10 ml) and MeOH (5 ml) was treated with 4M hydrochloric acid in 1 ,4- dioxane (6 ml, 24 mmol). After stirring for 2h at r.t. the reaction mixture was evaporated to dryness, diluted with water (50 ml) and brought to basic pH by addition of a saturated solution of NaHC03. The precipitated solid was filtered, washed with water, dried and purified by chromatography (Biotage SP1 Flash Purification system) on a silica gel cartridge (Biotage SNAP 25g) using DCM as eluant A and DCM / 7N NH3 in MeOH 10:1 as eluant B. Elution with a gradient from A / B 100:0 to 0:100 over 15 CV, followed by an isocratic elution with eluant B (5 CV), gave a yellow solid that was triturated with diethyl ether (15 ml) affording 111 mg (yield: 75%) of the title compound as a white solid. 1H-NMR (400 MHz), δ (ppm, DMSO-cie): 12.44 (s, 1 H) 10.36 (s, 1 H) 7.95 (d, J=9.03 Hz, 2 H) 7.59 (d, J=9.03 Hz, 1 H) 7.33 - 7.40 (m, 4 H) 7.25 - 7.32 (m, 1 H) 6.98 (d, J=9.15 Hz, 2 H) 6.85 (d, J=1.95 Hz, 1 H) 6.71 (dd, J=2.07, 8.91 Hz, 1 H) 4.58 (s, 2 H) 4.15 - 4.25 (m, 2 H) 3.77 - 3.86 (m, 2 H) 3.17 - 3.23 (m, 4 H) 2.78 - 2.86 (m, 4 H)
ESI (+) MS m/z 472 (MH+)
ESI (+) HRMS calcd for C27N503H29 + H+: 472.2343; found 472.2327
Figure imgf000053_0002
PHARMACOLOGY
The short forms and abbreviations used herein have the following meaning:
DMSO dimethylsulfoxide
g gram
1C50 concentration inhibiting by 50%
mg milligram
microg microgram
microL microliter
mL milliliter
mM millimolar
microM micromolar
nM nanomolar
Assays
Compounds of the present invention were tested in biochemical assays, as described below.
Preparation of FLT3 and KIT kinase cytoplasmic domains for use in biochemical assay
Cloning, expression and purification
FLT3 cytoplasmic domain (aa 564-993end of the 993 aminoacid long full length sequence, accession number P36888 of UniProtKB/Swiss-Prot. database) was amplified by PCR starting from a testis cDNA library and then cloned into pVL vector for expression in insect cells through the baculovirus system. The GST-FLT3 cytoplasmic domain has been expressed in Sf21 cells infected for 72 hours at 27°C. The recombinant protein has been purified by affinity on GSH-sepharose and eluted with glutathione. A further purification step has been performed on heparine sepharose. The final yield was of 0.5 mg/billion cells and the protein resulted >90% pure by coomassie staining. KIT cytoplasmic domain (aa 544-976end of the 976 aminoacid long full length sequence, accession number P10721 of UniProtKB/Swiss-Prot database) was cloned into pVL vector for expression in insect cells through the baculovirus system. The GST-KIT cytoplasmic domain has been expressed in Sf21 cells infected for 66 hrs at 27°C. The recombinant protein has been purified by affinity on GSH-sepharose and eluted with glutathione. The final yield was of 9 mg/billion cells and the protein resulted >80% pure by coomassie staining.
Purified proteins were stored at -80°C prior its use in biochemical assay.
Biochemical assays
i. General Principle - A specific peptidic substrate was trans-phosphorylated by the kinase in the presence of ATP traced with 33Ργ-ΑΤΡ. At the end of the phosphorylation reaction, the not reacted ATP, cold and radioactive, was captured by an excess of dowex ion exchange resin that eventually settled by gravity to the bottom of the reaction plate. The supernatant was subsequently withdrawn and transferred into a counting plate that was then evaluated by β-counting.
ii. Dowex resin preparation - 500 g of wet resin (SIGMA, custom prepared resin DOWEX 1x8 200-400 mesh, 2.5 Kg) were weighed out and diluted to 2 L in 150 mM sodium formate, pH 3.00. The resin was allowed to settle down overnight and then the supernatant was discarded. After three washes as above over a couple of days, the resin was allowed to settle and two volumes (with respect to the resin volume) were added of 150 mM sodium formate buffer. Biochemical assay for inhibitors of FLT3 kinase activity
i. Enzyme - The assay was performed using FLT3 cytoplasmic domain product and purified in house as GST fused protein. The FLT3 protein (1 microM) was pre activated with 800 microM ATP for 1 hour at 28*C in order to obtain a linear kinetic.
ii. FLT3 Kinase Buffer (KB) - Kinase buffer was composed of 50 mM HEPES pH 7.9 containing 4 mM MgC , 1 mM DTT, 10 microM Na3V04, and 0.2 mg/mL BSA
iii. Assay conditions - The FLT3 kinase assay was run with a final pre activated enzyme concentration of 2 nM, in the presence of 254 microM ATP (residual ATP from KIT pre activation step is negligible), 8 nM 33P-y-ATP and 55 microM of substrate BioDB n*24 (Aminoacidic sequence: GGKKKVSRSGLYRSPSMPENLNRPR- SEQ ID NO: 1). The peptide was purchased from American Peptide Company (Sunnyvale, CA).
Biochemical assay for inhibitors of KIT kinase activity
i. Enzyme - The assay has been performed using KIT cytoplasmic domain product and purified in house as GST fused protein. The KIT protein (4.5 microM ) was pre activated with 300 microM ATP for 1 hour at 28*C in order to obtain a linear kinetic.
ii. KIT kinase Buffer (KB) - Kinase buffer was composed of 50 mM HEPES pH 7.9 containing 5 mM MgC , 1 mM MnCI2, 10 mM DTT, 3 microM Na3V04, and 0.2 mg/mL BSA
iii. Assay conditions - The KIT kinase assay was run with a final pre activated enzyme concentration of 4 nM, in the presence of 4.4 microM ATP (residual ATP from KIT pre activation step is negligible), 3.9 nM 33P-y-ATP and 2.5 microM of substrate BioDB n*138 (Aminoacidic sequence: KWEEINGNNYVYIDPTQLPYDHKWEFPRNR- SEQ ID NO: 2). The peptide was purchased from American Peptide Company (Sunnyvale, CA).
Compound testing
i. Compound Dilution - For IC50 determination, test compounds were received as a 1 mM solution in 100% DMSO, distributed into 96 well plates: compounds were then plated into the first column of a microtiter plate (A1 to G1), 100 microL/well. An automated station for serial dilutions (Biomek FX, Beckman) was used for producing 1 :3 dilutions in 100 % DMSO, from line A1 to A10, and for all the compounds in the column. Moreover, 4-5 copies of daughter plates were prepared by reformatting 5 microL of this first set of 100% DMSO dilution plates into 384 deep well-plates: one of these plates with the serial dilutions of test compounds was thawed the day of the experiments, reconstituted at a 3X concentration with water and used in the IC50 determination assays. In a standard experiment, the highest concentration (3X) of all compounds was 30 microM, while the lowest one was 1.5 nM.
Each 384 well-plate contained at least one curve of the standard inhibitor staurosporine and reference wells (total enzyme activity vs. no enzymatic activity) for the Z' and signal to background evaluation.
ii. Assay Scheme - 384-well plates, V bottom (test plates) were prepared with 5 microL of the compound dilution (3X) and then placed onto a PlateTrak 12 robotized station (Perkin Elmer; the robot had one 384-tip pipetting head for starting the assay plus one 96-tip head for dispensing the resin) together with one reservoir for the Enzyme mix (3X) and one for the ATP mix (3X). At the start of the run, the robot aspirated 5 μΙ of ATP mix, made an air gap inside the tips (3 microL) and aspirated 5 microL of Enzyme mix. The following dispensation into the plates plus 3 cycles of mixing, done by the robot itself, started the kinase reaction. At this point, the correct concentrations were restored for all the reagents. The robot incubated the plates for 60 minutes at r.t., and then stoped the reaction by pipetting 60 microL of dowex resin suspension into the reaction mix. In order to avoid tip clogging, wide bore tips were used to dispense the resin suspension. Three cycles of mixing were done immediately after the addition of the resin. Another mixing cycle was performed after all the plates were stopped, this time using normal tips: the plates were then allowed to rest for about one hour in order to allow resin sedimentation. At this point, 27 microL of the supernatant were transferred into 384-Optiplates (Perkin-Elmer), with 50 microL of Microscint 40 (Perkin-Elmer); after 5 min of orbital shaking the plates were read on a Perkin-Elmer Top Count radioactivity counter.
iii. Data Fitting - Data were analyzed by an internally customized version of the SW package "Assay Explorer" that provided sigmoidal fitting of the ten-dilutions curves for IC50 determination in the secondary assays/hit confirmation routines.
Cell-based assays for inhibitors of FLT3 kinase activity
In vitro cell proliferation assay for inhibitors of FLT3 kinase activity
The human acute leukemia MOLM-13 AND MV-4-11 cells, bearing a FLT3-ITD mutation, were seeded (5000 cells/well) in white 384 well-plates in complete medium (RPMI 1640 plus 10% Fetal bovine serum) and treated with compounds dissolved in 0.1% DMSO, 24h after seeding. The cells were incubated at 37°C and 5 % CO2 and after 72 h the plates were processed using CellTiter-Glo assay (Promega) following the manufacturer's instruction.
CellTiter-Glo is a homogenous method based on the quantification of the ATP present, an indicator of metabolitically active cells. ATP was quantified using a system based on luciferase and D-luciferin resultant into light generation. The luminescent signal was proportional to the number of cells present in culture.
Briefly, 25 microL/well reagent solution were added to each well and, after 5 minutes shaking, microplates were read by Envision (PerkinElmer) luminometer. Inhibitory activity was evaluated comparing treated versus control data using Symyx Assay Explorer (Symyx Technologies Inc.) program. IC50 was calculated using sigmoidal interpolation curve. The compounds of formula (I) tested as described above, resulted to possess a remarkable FLT3 and KIT inhibitory activity, together with very good potency in inhibiting MOLM-13 AND MV-4-11 cells proliferation.
See, as an example, the following Table I, reporting the experimental data of some representative compounds of the invention being tested in biochemical assays as FLT3 and KIT kinase inhibitors (IC50 microM), and Table II, reporting the experimental data of some representative compounds of the invention being tested in cell proliferation assays as MOLM-13 and MV-4-11 inhibitors (IC50 microM), in comparison with the closest compound of the prior art (Ref. compound), described in WO03/028720, page 77, Table XI, entry 226, compound A02-M2-B05, having the following structure: Ref. Compound
Figure imgf000057_0001
In biochemical assays, the IC50 values are tipically lower than 2 microM on FLT3 and lower than 3 microM on KIT. In cell proliferation assays, the IC50 values are tipically lower than 3 microM, with 26 compounds having IC50 values lower than 0.1 microM on both cell lines.
Table I
Cpd No. FLT3 IC50 (microM) KIT IC50 (microM)
Biochemical assay Biochemical assay
1 0.428 2.805
2 1.134 0.172
3 0.639 0.564
4 0.974 >10
5 1.171 2.619
6 0.248 6.888
7 0.249 1.505
9 1.504 0.713
10 <0.050 0.956
11 <0.050 0.113
14 <0.050 0.118
15 <0.050 0.076
16 <0.050 0.106
17 0.062 0.146
18 0.414 1.392
19 <0.050 0.061
20 <0.050 0.052
21 0.104 0.134
22 0.056 0.055
23 <0.050 0.109
24 0.162 0.260
25 0.074 0.197
26 0.090 0.124 Cpd No. FLT3 ICso (microM) KIT ICso (microM)
Biochemical assay Biochemical assay
27 <0.050 0.091
28 0.136 0.306
29 0.635 1.078
30 0.166 1.405
32 <0.050 <0.050
33 <0.050 <0.050
34 0.056 0.058
35 1.176 2.370
38 0.539 0.613
39 0.062 0.528
40 <0.050 0.088
41 0.052 0.180
42 <0.050 0.055
Ref. Compound 3.175 >10
Table II
Cpd No. MOLM-13 IC50 (microM) MV-4-1 1 ICso (microM)
Cell proliferation assay Cell proliferation assay
1 0.789 0.683
2 1.158 1.475
3 0.990 0.987
4 0.092 <0.050
5 0.557 0.513
6 0.673 0.163
7 0.240 0.1 10
8 1.978 2.753
9 0.245 0.101
10 0.139 0.1 18
1 1 <0.050 <0.050
14 <0.050 <0.050
15 <0.050 <0.050
16 <0.050 <0.050
17 0.087 <0.050
19 <0.050 <0.050
20 <0.050 <0.050 Cpd No. MOLM-13 IC5o (microM) MV-4-1 1 ICso (microM) Cell proliferation assay Cell proliferation assay
21 0.068 <0.050
22 0.065 <0.050
23 <0.050 <0.050
24 <0.050 <0.050
25 <0.050 <0.050
26 <0.050 <0.050
27 <0.050 <0.050
28 <0.050 <0.050
29 <0.050 <0.050
30 <0.050 <0.050
32 <0.050 <0.050
33 <0.050 <0.050
34 <0.050 <0.050
35 <0.050 <0.050
38 <0.050 <0.050
39 0.081 0.050
40 <0.050 <0.050
41 <0.050 <0.050
43 0.240 0.1 18
Ref. Compound 3.91 1 5.537

Claims

1. A compound of formula (I):
Figure imgf000060_0001
AM Ar2 Ar3
wherein:
R1 is A, NR6R7, OR8, SOnR9, COR10, nitro, cyano or an optionally substituted group selected from C3-C6 cycloalkyi, heterocyclyl and heteroaryl;
R2, R3, R4 and R5 are independently hydrogen, halogen, nitro, cyano, SOnR9, COR10, NR11 R12, OR13 or an optionally substituted group selected from straight or branched C1-C6 alkyl, straight or branched C2-C6 alkenyl, straight or branched C2-C6 alkynyl, C3-C6 cycloalkyi and heterocyclyl wherein:
A is a straight or branched C1-C6 alkyl substituted with a group selected from an optionally substituted heterocyclyl, an optionally substituted heteroaryl, SOnR9, COR10, NR11 R12 and OR13;
R6 is hydrogen or an optionally substituted group selected from straight or branched Ο-Οβ alkyl, straight or branched C2-C6 alkenyl, straight or branched C2-C6 alkynyl, C3-C6 cycloalkyi, heterocyclyl, aryl and heteroaryl;
R7 is hydrogen, SOnR9, COR10, a substituted straight or branched C1-C6 alkyl or an optionally substituted group selected from straight or branched C2-C6 alkenyl, straight or branched C2-C6 alkynyl, C3-C6 cycloalkyi, heterocyclyl, aryl and heteroaryl or
R6 and R7, taken together with the nitrogen atom to which they are bound, may form an optionally substituted heterocyclyl group;
R8 is hydrogen, A, COR10 or an optionally substituted group selected from straight or branched C2-C6 alkenyl, straight or branched C2-C6 alkynyl, C3-C6 cycloalkyi, heterocyclyl, aryl and heteroaryl, wherein A is as defined above;
R9 is NR11 R12 or an optionally substituted group selected from straight or branched Ο-Οβ alkyl, straight or branched C2-C6 alkenyl, straight or branched C2-C6 alkynyl, C3-C6 cycloalkyi, heterocyclyl, aryl and heteroaryl; R10 is hydrogen, NR11 R12, 0R13 or an optionally substituted group selected from straight or branched O- C6 alkyl, straight or branched C2-C6 alkenyl, straight or branched C2-C6 alkynyl, C3-C6 cycloalkyl, heterocyclyl, aryl and heteroaryl;
R11 and R12 are independently hydrogen, SOnR9, COR10 or an optionally substituted group selected from straight or branched C1-C6 alkyl, straight or branched C2-C6 alkenyl, straight or branched C2-C6 alkynyl, C3-
C6 cycloalkyl, heterocyclyl, aryl and heteroaryl, wherein R9 and R10 are as defined above, or
R11 and R12, taken together with the nitrogen atom to which they are bound, may form an optionally substituted heterocyclyl group;
R13 is hydrogen, COR10 or an optionally substituted group selected from straight or branched C1-C6 alkyl, straight or branched C2-C6 alkenyl, straight or branched C2-C6 alkynyl, C3-C6 cycloalkyl, heterocyclyl, aryl and heteroaryl, wherein R10 is as defined above;
n is 0,1 or 2;
X is a bond or an optionally substituted group selected from straight or branched C1-C6 alkyl, heterocycly and aryl; Y is a bond, oxygen, or an optionally substituted group selected from straight or branched C1-C6 alkyl, straight or branched C2-C6 alkenyl, straight or branched C2-C6 alkynyl, heterocyclyl and aryl;
Z is a bond, oxygen or an optionally substituted straight or branched C1-C6 alkyl;
Ar' is an optionally substituted aryl or an optionally substituted heteroaryl;
or a pharmaceutically acceptable salt thereof.
2. A compound of formula (I) as defined in claim 1 wherein:
R1 is A, NR6R7, OR8 or an optionally substituted heterocyclyl, wherein A, R6, R7 and R8 are as defined in claim 1.
3. A compound of formula (I) as defined in claims 1 or 2 wherein:
Ar is Ar1 or Ar2; and R2, R3, R4, R5 are each independently hydrogen, halogen, NR11 R12 or OR13, wherein R11 , R12 and R13 are as defined in claim 1.
4. A compound of formula (I) or a pharmaceutically acceptable salt thereof as defined in claim 1 which is selected from the group consisting of:
N-(6-Benzyloxy-1 H-indazol-3-yl)-4-(4-methyl-piperazin-1-yl)-benzamide,
4-(4-Methyl-piperazin-1-yl)-N-(6-phenoxy-1 H-indazol-3-yl)-benzamide,
N-[6-(3-Fluoro-phenoxy)-1 H-indazol-3-yl]-4-(4-methyl-piperazin-1-yl)-benzamide,
N-[6-(4-Benzyloxy-phenoxy)-1 H-indazol-3-yl]-4-(4-methyl-piperazin-1-yl)-benzamide,
4-(4-Methyl-piperazin-1-yl)-N-[6-(3-phenoxy-benzyloxy)-1 H-indazol-3-yl]-benzamide,
N-[6-(1 -Benzyl-piperidin-4-yloxy)-1 H-indazol-3-yl]-4-(4-methyl-piperazin-1 -yl)-benzamide,
4-(4-Methyl-piperazin-1-yl)-N-[6-(3-phenyl-prop-2-ynyloxy)-1 H-indazol-3-yl]-benzamide,
4-(4-Methyl-piperazin-1-yl)-N-[6-(4-phenoxy-phenoxy)-1 H-indazol-3-yl]-benzamide,
N-[6-(3-Benzyloxy-phenoxy)-1 H-indazol-3-yl]-4-(4-methyl-piperazin-1-yl)-benzamide,
4-(4-Methyl-piperazin-1-yl)-N-[6-(2-phenoxy-ethoxy)-1 H-indazol-3-yl]-benzamide,
N-[6-(2-Benzyloxy-ethoxy)-1 H-indazol-3-yl]-4-(4-methyl-piperazin-1-yl)-benzamide,
N-[6-(1 -Benzyl-piperidin-3-yloxy)-1 H-indazol-3-yl]-4-(4-methyl-piperazin-1 -yl)-benzamide, N-[6-(1-Benzyl-pyrrolidin-2-ylmethoxy)-1 H-indazol-3-yl]-4-(4-methyl-piperazin-1-yl)-benzamide, N-[6-(2-Benzyloxy-ethoxy)-1 H-indazol-3-yl]-4-(4-methyl-4-oxy-piperazin-1-yl)-benzamide,
N-[6-(2-Benzyloxy-ethoxy)-1 H-indazol-3-yl]-4-(4-dimethylamino-piperidin-1-yl)-benzamide,
N-[6-(2-Benzyloxy-ethoxy)-1 H-indazol-3-yl]-4-[(2-dimethylamino-ethyl)-methyl-amino]-benzamide,
N-[6-(2-Benzyloxy-ethoxy)-1 H-indazol-3-yl] -[(3-dimethylamino-propyl)-m
4-{4-[6-(2-Benzyloxy-ethoxy)-1 H-indazol-3-ylcarbamoyl]-phenyl}-piperazine-1-carboxylic acid tert-butyl ester, N-[6-(2-Benzyloxy-ethoxy)-1 H-indazol-3-yl]-4-(1-methyl-piperidin-4-ylamino)-benzarriide,
N-[6-(2-Benzyloxy-ethoxy)-1 H-indazol-3-yl]-4-piperazin-1-yl-benzamide,
N-[6-(2-Benzyloxy-ethoxy)-1 H-indazol-3-yl] -dimethylaminomethyl-benzarnide,
N-[6-(2-Benzyloxy-ethoxy)-1 H-indazol-3-yl]-4-(1-methyl-piperidin-4-yloxy)-benzamide,
N-[6-(2-Benzyloxy-ethoxy)-1 H-indazol-3-yl]-4-[methyl-(1-methyl-piperidin-4-yl)-amino]-benzamide,
N-[6-(2-Benzyloxy-ethoxy)-1 H-indazol-3-yl]-4-morpholin-4-yl-benzamide,
N-[6-(2-Benzyloxy-ethoxy)-1 H-indazol-3-yl]-4-(2-morpholin-4-yl-ethylarnino)-benzarnide,
N-[6-(2-Benzyloxy-ethoxy)-1 H-indazol-3-yl] -(tetrahydro-pyran -ylamino)-benzarnide,
N-[6-(2-Benzyloxy-ethoxy)-1 H-indazol-3-yl]-4-[(1-methyl-piperidin-4-ylmethyl)-amino]-benzamide,
N-[6-(2-Benzyloxy-ethoxy)-1 H-indazol-3-yl]-4-(3-pyrrolidin-1-yl-azetidin-1-yl)-benzamide,
N-[6-(2-Benzyloxy-ethoxy)-1 H-indazol-3-yl]-3-(4-methyl-piperazin-1-yl)-benzamide,
N-{6-[2-(2-Fluoro-benzyloxy)-ethoxy]-1 H-indazol-3-yl}-4-(4-methyl-piperazin-1-yl)-benzamide,
N-{6-[2-(3-Fluoro-benzyloxy)-ethoxy]-1 H-indazol-3-yl}-4-(4-methyl-piperazin-1-yl)-benzamide,
N-{6-[2-(4-Fluoro-benzyloxy)-ethoxy]-1 H-indazol-3-yl}-4-(4-methyl-piperazin-1-yl)-benzamide,
4-(4-Methyl-piperazin-1-yl)-N-{6-[2-(4-tnfluoromethyl-benzyloxy)-ethoxy]-1 H-indazol-3-yl}-benzamide,
4-(4-Methyl-piperazin-1-yl)-N-{6-[2-(3-tnfluoromethyl-benzyloxy)-ethoxy]-1 H-indazol-3-yl}-benzamide,
4-(4-Methyl-piperazin-1-yl)-N-{6-[2-(pyridin-4-ylmethoxy)-ethoxy]-1 H-indazol-3-yl}-benzamide,
4-(4-Methyl-piperazin-1-yl)-N-{6-[2-(pyridin-3-ylmethoxy)-ethoxy]-1 H-indazol-3-yl}-benzamide,
4-(4-Methyl-piperazin-1-yl)-N-{6-[2-(pyridin-2-ylmethoxy)-ethoxy]-1 H-indazol-3-yl}-benzamide,
N-[6-(2-Benzyloxy-ethoxy)-1 H-indazol-3-yl]-2-fluoro-4-(4-methyl-piperazin-1-yl)-benzamide,
4-(4-Methyl-piperazin-1-yl)-N-[6-((E)-3-phenyl-allyloxy)-1 H-indazol-3-yl]-benzamide,
N-{6-[2-(4-Methoxy-benzyloxy)-ethoxy]-1 H-indazol-3-yl} -(4-methyl-piperazin-1-yl)-benzam
N-[6-(2-Benzyloxy-ethoxy)-1 H-indazol-3-yl]-4-[methyl-(1-methyl-1-oxy-piperidin-4-yl)-amino]-benzamide, 4-(4-Methyl-4-oxy-piperazin-1-yl)-N-{6-[2-(4-trifluoromethyl-benzyloxy)-ethoxy]-1 H-indazol-3-yl}-benzamide and N-[6-(2-Benzyloxy-ethoxy)-1 H-indazol-3-yl]-2,4-bis-(4-methyl-piperazin-1-yl)-benzamide.
5. A process for preparing a compound of formula (I) as defined in claim 1 or a pharmaceutically acceptable salt thereof, characterized in that the process comprises the following steps:
F) cleaving the tert-butoxy-carbonyl group of the compound of formula (XI)
Figure imgf000063_0001
wherein Ar, Ar', X, Y and Z are as defined in claim 1 , so as to obtain a compound of formula (I), as defined above; or in alternative
L) acylating the compound of formula (XVI)
Figure imgf000063_0002
wherein Ar', X, Y and Z are as defined in claim 1 , with a compound of formula (V)
Figure imgf000063_0003
wherein Ar is as defined in claim 1 and W is hydroxy, halogen or a suitable leaving group; so as to obtain a compound of formula (I), as defined above;
or in alternative
M) acylating the compound of formula (XVI), as defined above, with a compound of formula (XVII)
Figure imgf000063_0004
wherein R2, R3, R4 and R5 are as defined in claim 1 , W is as defined above and L is a suitable leaving group, such as halogen, methanesulfonyloxy, trifl uoromethanesulfonyloxy or p-toluenesulfonyloxy;
N) coupling the resultant compound of formula (XVIII)
Figure imgf000063_0005
wherein Ar', X, Y, Z, R2, R3, R4 and R5 are as defined in claim 1 and L is as defined above, with a compound of formula (XIX) (XIX)
H
wherein R6 is as defined in claim 1 and R7 is hydrogen, a substituted straight or branched Ο-0β alkyl or an optionally substituted group selected from straight or branched C2-C6 alkenyl, straight or branched C2-C6 alkynyl, C3- C6 cycloalkyl, heterocyclyl, aryl and heteroaryl, so as to obtain a compound of formula (I), wherein R1 is NR6R7, wherein R6 is as defined in formula (I) and R7 is hydrogen, a substituted straight or branched C1-C6 alkyl or an optionally substituted group selected from straight or branched C2-C6 alkenyl, straight or branched C2-C6 alkynyl, C3- C6 cycloalkyl, heterocyclyl, aryl and heteroaryl, and Ar, Ar', X, Y, Z are as defined in claim 1 ;
optionally separating the resultant compound of formula (I) into the single isomers; optionally converting the resultant compound of formula (I) into a different compound of formula (I), and/or into a pharmaceutically acceptable salt.
6. A process according to claim 5, characterized in that the compound of formula (XI) as defined in claim 5 is prepared according to the following steps:
A) introducing the tert-butoxy-carbonyl roup into the compound of formula (II)
Figure imgf000064_0001
B) cleaving the phthalimido group of the resultant com ound of formula (III)
Figure imgf000064_0002
C) acylating the resultant compound of formula (IV)
Figure imgf000064_0003
by reaction with a compound of formula (V) (V)
Ar W
wherein Ar is as defined in claim 1 and W is hydroxy, halogen or a suitable leaving group;
D) selectively cleaving the tert-butyldim resultant compound of formula (VI)
Figure imgf000065_0001
wherein Ar is as defined above;
E) coupling the resultant compound of formula (VII)
Figure imgf000065_0002
wherein Ar is as defined in claim 1 , alternatively with:
Ea) a compound of formula (VIII)
Ar'/Z 'X^OH (VI I I) wherein Ar', Z and Y are as defined in claim 1 and X is an optionally substituted group selected from straight or branched Ο-0β alkyl and heterocyclyl;
or
Eb) a compound of formula (IX)
Ar'/Z -X^B(OH)2 (IX)
wherein Ar', Z and Y are as defined in claim 1 and X is an optionally substituted aryl or
wherein Ar' is as defined in claim 1 and X, Y and Z are a bond;
or
Ec) a compound of formula (X)
Figure imgf000065_0003
wherein Ar', Z and Y are as defined in claim 1 , X is an optionally substituted group selected from straight or branched 0-C6 alkyl or heterocyclyl and L is a suitable leaving group such as halogen, methanesulfonyloxy, trifluoromethanesulfonyloxy or p-toluenesulfonyloxy; so as to obtain a compound of formula (XI), as defined in claim 5.
7. A process according to claim 5, characterized in that the compound of formula (XI) as defined in claim 5 is prepared according to the following steps:
G) selectively cleaving the tert-butyldimethylsilyl ether of the compound of formula (IV), as defined in claim 6;
H) coupling the resultant compound of formula (XII)
Figure imgf000066_0001
alternatively with:
Ha) a compound of formula (VIII), as defined in claim 6;
or
Hb) a compound of formula (X), as defined in claim 6;
I) acylating the resultant compound of formula (XIII)
Figure imgf000066_0002
wherein Ar, Ar', Y and Z are as defined in claim 1 and X is an optionally substituted group selected from straight or branched Ο-0β alkyl and heterocyclyl, with a compound of formula (V), as defined in claim 6, so as to obtain a compound of formula (XI), wherein Ar, Ar', Y and Z are as defined in claim 1 and X is an optionally substituted group selected from straight or branched C1-C6 alkyl and heterocyclyl.
8. A process according to claim 5, characterized in that the compound of formula (XVI) as defined in claim 5 is prepared according to the following steps:
J) coupling the compound of formula (XIV)
Figure imgf000066_0003
alternatively with:
Ja) a compound of formula (VIII), as defined in claim 6;
or
Jb) a compound of formula (IX), as defined in claim 6;
or Jc) a compound of formula (X), as defined in claim 6;
K) converting the resultant compound of formula (XV)
Figure imgf000067_0001
wherein Ar', X, Y and Z are as defined in claim 1 ; so as to obtain a compound of formula (XVI), as defined in claim 5.
9. A method of treating a disease caused by and/or associated with a degulated protein kinase activity which comprises administering to a mammal in need thereof an effective amount of a compound of formula (I) as defined in claim 1.
10. The method according to claim 9 wherein the mammal in need thereof is a human.
11. The method according to claim 9 of treating a disease caused by and/or associated with a degulated FLT3 or KIT kinase activity.
12. The method according to claim 9 wherein the disease is selected from the group consisting of cancer, cell proliferation disorders and immune cell-associated diseases and disorders.
13. The method according to claim 12 wherein the cancer is selected from the group consisting of carcinoma such as bladder, breast, colon, kidney, liver, lung, including small cell lung cancer, salivary gland, esophagus, gall-bladder, ovary, pancreas, stomach, cervix, thyroid, thymus, prostate, and skin, including squamous cell carcinoma; hematopoietic tumors of lymphoid lineage including leukemia, acute lymphocitic leukemia, acute lymphoblastic leukemia, B-cell lymphoma, T-cell-lymphoma, Hodgkin's lymphoma, non-Hodgkin's lymphoma, hairy cell lymphoma and Burkitt's lymphoma; hematopoietic tumors of myeloid lineage, including acute and chronic myelogenous leukemias, myelodysplasia syndrome and promyelocytic leukemia; tumors of mesenchymal origin, including gastrointestinal stromal tumor, fibrosarcoma and rhabdomyosarcoma; tumors of the central and peripheral nervous system, including astrocytoma neuroblastoma, glioma and schwannomas; other tumors, including mast cell disease, melanoma, seminoma, teratocarcinoma, osteosarcoma, xeroderma pigmentosum, keratoxanthoma, thyroid follicular cancer, Kaposi's sarcoma and mesothelioma and others.
14. The method according to claim 12 wherein the cellular proliferation disorder is selected from the group consisting of benign prostate hyperplasia, familial adenomatosis polyposis, neurofibromatosis, psoriasis, vascular smooth cell proliferation associated with atherosclerosis, pulmonary fibrosis, arthritis, glomerulonephritis and post-surgical stenosis and restenosis.
15. The method according to claim 12 wherein the immune cell-associated disease or disorder is selected from the group consisting of inflammatory and autoimmune diseases, multiple sclerosis, systemic lupus erythematosis, inflammatory bowel diseases(IBD), Crohn's disease, irritable bowel syndrome, pancreatitis, ulcerative colitis, diverticulosis, myasthenia gravis, vasculitis, psoriasis, scleroderma, asthma, allergy, systemic sclerosis, vitiligo, arthritis, osteoarthritis, juvenile rheumatoid arthritis and ankylosing spondylitis.
16. The method according to claim 13 wherein the cancer is a FLT3 mutated cancer, such as acute myeloid leukemia or myelodisplastic syndrome.
17. The method according to claim 13 wherein the cancer is a KIT mutated cancer, such as gastrointestinal stromal tumors, melanoma, acute myeloid leukemia, primary adenoid cystic carcinoma of the salivary gland, thymic carcinoma, glioma, testicular seminoma, small cell lung cancers, mast cell disease or piebaldism.
18. The method according to claim 9 which provides tumor angiogenesis and metastasis inhibition.
19. The method according to claim 13 further comprising subjecting the mammal in need thereof to a radiation therapy or chemotherapy regimen in combination with at least one cytostatic or cytotoxic agent.
20. An in vitro method for inhibiting FLT3 or KIT protein kinase activity which comprises contacting the said protein with an effective amount of a compound of formula (I) as defined in claim 1.
21. A pharmaceutical composition comprising a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof, as defined in claim 1 , and at least one pharmaceutically acceptable excipient, carrier and/or diluent.
22. A pharmaceutical composition according to claim 21 further comprising one or more chemotherapeutic agents.
23. A product comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof, as defined in claim 1 , and one or more chemotherapeutic agents, as a combined preparation for simultaneous, separate or sequential use in anticancer therapy.
24. A compound of formula (I) or a pharmaceutically acceptable salt thereof, as defined in claim 1 , for use as a medicament.
25. A compound of formula (I) or a pharmaceutically acceptable salt thereof, as defined in claim 1 , for use in a method of treating cancer.
26. Use of a compound of formula (I) or a pharmaceutically acceptable salt thereof, as defined in claim 1 , in the manufacture of a medicament with anticancer activity.
PCT/EP2012/058389 2011-05-12 2012-05-07 Substituted indazole derivatives active as kinase inhibitors WO2012152763A1 (en)

Priority Applications (20)

Application Number Priority Date Filing Date Title
SG2013083696A SG194911A1 (en) 2011-05-12 2012-05-07 Substituted indazole derivatives active as kinase inhibitors
SI201230838A SI2707359T1 (en) 2011-05-12 2012-05-07 Substituted indazole derivatives active as kinase inhibitors
KR1020137033115A KR101953272B1 (en) 2011-05-12 2012-05-07 Substituted indazole derivatives active as kinase inhibitors
AU2012252468A AU2012252468B2 (en) 2011-05-12 2012-05-07 Substituted indazole derivatives active as kinase inhibitors
ES12718275.6T ES2611779T3 (en) 2011-05-12 2012-05-07 Active substituted indazole derivatives as kinase inhibitors
CA2835478A CA2835478C (en) 2011-05-12 2012-05-07 Substituted indazole derivatives active as kinase inhibitors
CN201280023142.9A CN103534239B (en) 2011-05-12 2012-05-07 There is the indazole derivatives of the replacement of kinase inhibitory activity
BR112013028733-0A BR112013028733B1 (en) 2011-05-12 2012-05-07 Substituted indazole derivatives active as kinase inhibitors
EP12718275.6A EP2707359B1 (en) 2011-05-12 2012-05-07 Substituted indazole derivatives active as kinase inhibitors
US14/116,512 US9408850B2 (en) 2011-05-12 2012-05-07 Substituted indazole derivatives active as kinase inhibitors
JP2014509702A JP6396210B2 (en) 2011-05-12 2012-05-07 Substituted indazole derivatives active as kinase inhibitors
EA201391682A EA023579B1 (en) 2011-05-12 2012-05-07 Substituted indazole derivatives active as kinase inhibitors
DK12718275.6T DK2707359T3 (en) 2011-05-12 2012-05-07 SUBSTITUTED INDAZOLD DERIVATIVES ACTIVE AS KINase INHIBITORS
MX2013012981A MX342509B (en) 2011-05-12 2012-05-07 Substituted indazole derivatives active as kinase inhibitors.
IL229252A IL229252A (en) 2011-05-12 2013-11-05 Substituted indazole derivatives active as kinase inhibitors, process for their preparation, pharmaceutical compositions comprising them and their use in the manufacture of medicaments
ZA2013/09345A ZA201309345B (en) 2011-05-12 2013-12-11 Substituted indazole derivatives active as kinase inhibitors
HK14107159.6A HK1193812A1 (en) 2011-05-12 2014-07-14 Substituted indazole derivatives active as kinase inhibitors
US15/203,087 US9597317B2 (en) 2011-05-12 2016-07-06 Substituted indazole derivatives active as kinase inhibitiors
US15/203,092 US10028934B2 (en) 2011-05-12 2016-07-06 Substituted indazole derivatives active as kinase inhibitiors
US16/013,019 US10478423B2 (en) 2011-05-12 2018-06-20 Substituted indazole derivatives active as kinase inhibitiors

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP11165882.9 2011-05-12
EP11165882 2011-05-12

Related Child Applications (3)

Application Number Title Priority Date Filing Date
US14/116,512 A-371-Of-International US9408850B2 (en) 2011-05-12 2012-05-07 Substituted indazole derivatives active as kinase inhibitors
US15/203,092 Division US10028934B2 (en) 2011-05-12 2016-07-06 Substituted indazole derivatives active as kinase inhibitiors
US15/203,087 Division US9597317B2 (en) 2011-05-12 2016-07-06 Substituted indazole derivatives active as kinase inhibitiors

Publications (1)

Publication Number Publication Date
WO2012152763A1 true WO2012152763A1 (en) 2012-11-15

Family

ID=46025764

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2012/058389 WO2012152763A1 (en) 2011-05-12 2012-05-07 Substituted indazole derivatives active as kinase inhibitors

Country Status (24)

Country Link
US (4) US9408850B2 (en)
EP (1) EP2707359B1 (en)
JP (1) JP6396210B2 (en)
KR (1) KR101953272B1 (en)
CN (1) CN103534239B (en)
AR (1) AR086357A1 (en)
AU (1) AU2012252468B2 (en)
BR (1) BR112013028733B1 (en)
CA (1) CA2835478C (en)
CL (1) CL2013003227A1 (en)
DK (1) DK2707359T3 (en)
EA (1) EA023579B1 (en)
ES (1) ES2611779T3 (en)
HK (1) HK1193812A1 (en)
HU (1) HUE031166T2 (en)
IL (1) IL229252A (en)
MX (1) MX342509B (en)
PL (1) PL2707359T3 (en)
PT (1) PT2707359T (en)
SG (1) SG194911A1 (en)
SI (1) SI2707359T1 (en)
TW (1) TWI573790B (en)
WO (1) WO2012152763A1 (en)
ZA (1) ZA201309345B (en)

Cited By (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103739550A (en) * 2014-01-02 2014-04-23 中国药科大学 2, 3-dimethyl-6-urea-2H-indazole compound and preparation method and application thereof
WO2017011776A1 (en) 2015-07-16 2017-01-19 Array Biopharma, Inc. Substituted pyrazolo[1,5-a]pyridine compounds as ret kinase inhibitors
DE102015012050A1 (en) * 2015-09-15 2017-03-16 Merck Patent Gmbh Compounds as ASIC inhibitors and their uses
WO2017176744A1 (en) 2016-04-04 2017-10-12 Loxo Oncology, Inc. Methods of treating pediatric cancers
WO2017176751A1 (en) 2016-04-04 2017-10-12 Loxo Oncology, Inc. Liquid formulations of (s)-n-(5-((r)-2-(2,5-difluorophenyl)-pyrrolidin-1-yl)-pyrazolo[1,5-a]pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide
WO2018071454A1 (en) 2016-10-10 2018-04-19 Andrews Steven W Substituted pyrazolo[1,5-a]pyridine compounds as ret kinase inhibitors
WO2018071447A1 (en) 2016-10-10 2018-04-19 Andrews Steven W Substituted pyrazolo[1,5-a]pyridine compounds as ret kinase inhibitors
WO2018136661A1 (en) 2017-01-18 2018-07-26 Andrews Steven W SUBSTITUTED PYRAZOLO[1,5-a]PYRAZINE COMPOUNDS AS RET KINASE INHIBITORS
WO2018136663A1 (en) 2017-01-18 2018-07-26 Array Biopharma, Inc. Ret inhibitors
US10045991B2 (en) 2016-04-04 2018-08-14 Loxo Oncology, Inc. Methods of treating pediatric cancers
WO2018170381A1 (en) 2017-03-16 2018-09-20 Andrews Steven W Macrocyclic compounds as ros1 kinase inhibitors
US10251889B2 (en) 2009-07-09 2019-04-09 Array BioPharm Inc. Substituted pyrazolo[1,5-a]pyrimidine compounds as Trk kinase inhibitors
WO2019075114A1 (en) 2017-10-10 2019-04-18 Mark Reynolds Formulations comprising 6-(2-hydroxy-2-methylpropoxy)-4-(6-(6-((6-methoxypyridin-3-yl)methyl)-3,6-diazab icyclo[3.1.1]heptan-3-yl)pyridin-3-yl)pyrazolo[1,5-a]pyridine-3-carbonitrile
WO2019075108A1 (en) 2017-10-10 2019-04-18 Metcalf Andrew T Crystalline forms
WO2019143994A1 (en) 2018-01-18 2019-07-25 Array Biopharma Inc. Substituted pyrazolyl[4,3-c]pyridinecompounds as ret kinase inhibitors
WO2019143977A1 (en) 2018-01-18 2019-07-25 Array Biopharma Inc. Substituted pyrrolo[2,3-d]pyrimidines compounds as ret kinase inhibitors
WO2020028258A1 (en) 2018-07-31 2020-02-06 Loxo Oncology, Inc. Spray-dried dispersions and formulations of (s)-5-amino-3-(4-((5-fluoro-2-methoxybenzamido)methyl)phenyl)-1-(1,1,1-trifluoro propan-2-yl)-1h-pyrazole-4-carboxamide
WO2020055672A1 (en) 2018-09-10 2020-03-19 Array Biopharma Inc. Fused heterocyclic compounds as ret kinase inhibitors
WO2020131674A1 (en) 2018-12-19 2020-06-25 Array Biopharma Inc. 7-((3,5-dimethoxyphenyl)amino)quinoxaline derivatives as fgfr inhibitors for treating cancer
WO2020131627A1 (en) 2018-12-19 2020-06-25 Array Biopharma Inc. Substituted pyrazolo[1,5-a]pyridine compounds as inhibitors of fgfr tyrosine kinases
US10724102B2 (en) 2015-10-26 2020-07-28 Loxo Oncology, Inc. Point mutations in TRK inhibitor-resistant cancer and methods relating to the same
EP3725777A1 (en) * 2019-04-17 2020-10-21 Rottapharm Biotech S.r.l. Benzo- and pyrido-pyrazoles as protein kinase inhibitors
WO2021134004A1 (en) 2019-12-27 2021-07-01 Schrodinger, Inc. Cyclic compounds and methods of using same
US11091486B2 (en) 2016-10-26 2021-08-17 Array Biopharma, Inc Process for the preparation of pyrazolo[1,5-a]pyrimidines and salts thereof
US11214571B2 (en) 2016-05-18 2022-01-04 Array Biopharma Inc. Process for the preparation of (S)-N-(5-((R)-2-(2,5-difluorophenyl)pyrrolidin-1-yl)-pyrazolo[1,5-a]pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide and salts thereof
WO2022055963A1 (en) 2020-09-10 2022-03-17 Schrödinger, Inc. Heterocyclic pericondensed cdc7 kinase inhibitors for the treatment of cancer
WO2022164789A1 (en) 2021-01-26 2022-08-04 Schrödinger, Inc. Tricyclic compounds useful in the treatment of cancer, autoimmune and inflammatory disoders
WO2022197898A1 (en) 2021-03-18 2022-09-22 Schrödinger, Inc. Cyclic compounds and methods of using same
US11524963B2 (en) 2018-01-18 2022-12-13 Array Biopharma Inc. Substituted pyrazolo[3,4-d]pyrimidines as RET kinase inhibitors
WO2024088829A2 (en) 2022-10-25 2024-05-02 Nerviano Medical Sciences S.R.L. Salts and polymorphs of 4-(4-methyl-piperazin-1-yl)-n-{6-[2-(4-trifluoromethyl-benzyloxy)-ethoxy]-1h-indazol-3-yl}-benzamide

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9408850B2 (en) 2011-05-12 2016-08-09 Nerviano Medical Sciences S.R.L. Substituted indazole derivatives active as kinase inhibitors
CN112794820B (en) * 2019-11-14 2022-06-07 四川大学 Indazole derivative and preparation method and application thereof
CN111116734A (en) * 2019-12-23 2020-05-08 维塔恩(广州)医药有限公司 Tumor-associated gene c-kit mutation-associated antigen short peptide and application thereof
WO2022033472A1 (en) * 2020-08-11 2022-02-17 河南迈英诺医药科技有限公司 Fgfr inhibitor compound and use thereof

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003028720A1 (en) 2001-09-26 2003-04-10 Pharmacia Italia S.P.A. Aminoindazole derivatives active as kinase inhibitors, process for their preparation and pharmaceutical compositions containing them
WO2005073224A2 (en) * 2004-01-23 2005-08-11 Amgen Inc Quinoline quinazoline pyridine and pyrimidine counds and their use in the treatment of inflammation angiogenesis and cancer
WO2005085206A1 (en) 2004-02-27 2005-09-15 F. Hoffmann-La Roche Ag Indazole derivatives and pharmaceutical compositions containing them
WO2008003396A1 (en) 2006-07-01 2008-01-10 Merck Patent Gmbh Indazole derivatives for treating hsp90-induced diseases
WO2010069966A1 (en) 2008-12-18 2010-06-24 Nerviano Medical Sciences S.R.L. Substituted indazole derivatives active as kinase inhibitors

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DK2120932T3 (en) * 2006-12-20 2014-10-13 Nerviano Medical Sciences Srl INDAZOL DERIVATIVES AS KINase INHIBITORS FOR TREATMENT OF CANCER
EP2176231B1 (en) * 2007-07-20 2016-10-19 Nerviano Medical Sciences S.r.l. Substituted indazole derivatives active as kinase inhibitors
AR071780A1 (en) * 2008-05-15 2010-07-14 Nerviano Medical Sciences Srl BICYCLE CARBONILAMINO-PIRAZOLES CARBAMILILE DERIVATIVES AS PROFARMACOS
US9408850B2 (en) 2011-05-12 2016-08-09 Nerviano Medical Sciences S.R.L. Substituted indazole derivatives active as kinase inhibitors

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003028720A1 (en) 2001-09-26 2003-04-10 Pharmacia Italia S.P.A. Aminoindazole derivatives active as kinase inhibitors, process for their preparation and pharmaceutical compositions containing them
WO2005073224A2 (en) * 2004-01-23 2005-08-11 Amgen Inc Quinoline quinazoline pyridine and pyrimidine counds and their use in the treatment of inflammation angiogenesis and cancer
WO2005085206A1 (en) 2004-02-27 2005-09-15 F. Hoffmann-La Roche Ag Indazole derivatives and pharmaceutical compositions containing them
WO2008003396A1 (en) 2006-07-01 2008-01-10 Merck Patent Gmbh Indazole derivatives for treating hsp90-induced diseases
WO2010069966A1 (en) 2008-12-18 2010-06-24 Nerviano Medical Sciences S.R.L. Substituted indazole derivatives active as kinase inhibitors

Non-Patent Citations (24)

* Cited by examiner, † Cited by third party
Title
A.R. KATRITZKY; 0. METH-COHN; C.W. REES: "Comprehensive Organic Functional Group Transformations", 1995, ELSEVIER PERGAMON
A.R. KATRITZKY; R.J.K. TAYLOR: "Comprehensive Organic Functional Group Transformations II", 2005, ELSEVIER PERGAMON
ANTONESCU CR., J PATHOL., vol. 223, no. 2, 2011, pages 251 - 6
B.M.TROST; I. FLEMING: "Comprehensive Organic Synthesis", 1991, PERGAMON PRESS
CARCINOGENESIS, vol. 29, 2008, pages 1087 - 191
CELL, vol. 100, 2000, pages 113 - 127
COLOMBO, M.; RICCARDI-SIRTORI, F.; RIZZO, V.: "A fully automated method for accurate mass determination using high-performance liquid chromatography with a quadrupole/orthogonal acceleration time-of-flight mass spectrometer", RAPID COMMUN. MASS SPECTROM., vol. 18, 2004, pages 511 - 517
CURRENT OPINION IN CHEMICAL BIOLOGY, vol. 3, 1999, pages 459 - 465
CURTIN JA, JCO, vol. 24, no. 26, 2006, pages 4340 - 4346
GREEN, THEODORA W.; WUTS, PETER G.M.: "Protective Groups in Organic Synthesis, Third Edition,", 1999, JOHN WILEY & SONS INC.
HORIIKE S ET AL., LEUKEMIA, vol. 11, no. 9, September 1997 (1997-09-01), pages 1442 - 6
JACQUES, JEAN; COLLET, ANDRE; WILEN, SAMUEL H.: "Enantiomers, Racemates, and Resolutions", 1981, JOHN WILEY & SONS INC.
LIM KH; PARDANANI A; TEFFERI A, ACTA HAEMATOL., vol. 119, no. 4, 2008, pages 194 - 8
MALAISE M; STEINBACH D; CORBACIOGLU S, CURR HEMATOL MALIG REP., vol. 4, no. 2, 2009, pages 77 - 82
MASSON K; RONNSTRAND L., CELL SIGNAL., vol. 21, no. 12, December 2009 (2009-12-01), pages 1717 - 26
MICKE P ET AL., CLIN CANCER RES., vol. 9, no. 1, 2003, pages 188 - 94
MORRIS PG; ABREY LE., TARGET ONCOL., vol. 5, no. 3, 2010, pages 193 - 200
MURAKAMI T ET AL., J DERMATOL SCI., vol. 35, no. 1, June 2004 (2004-06-01), pages 29 - 33
NAKAO M ET AL., LEUKEMIA, vol. 10, no. 12, December 1996 (1996-12-01), pages 1911 - 8
NATURE REV. DRUG DISCOV., vol. 1, 2002, pages 309 - 315
NIKOLAOU M. ET AL., ANTICANCER RES., vol. 27, no. 3B, 2007, pages 1685 - 8
PITTONI P. ET AL., ONCOGENE, vol. 30, no. 7, 17 February 2011 (2011-02-17), pages 757 - 69
STROBEL P; HOHENBERGER P; MARX A, J THORAC ONCOL., vol. 5, no. 10, 2010, pages 286 - 90
VILA L; LIU H; AI-QURAN SZ; COCO DP; DONG HJ; LIU C, MOD PATHOL., vol. 22, no. 10, 2009, pages 1296 - 302

Cited By (62)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10758542B2 (en) 2009-07-09 2020-09-01 Array Biopharma Inc. Substituted pyrazolo[l,5-a]pyrimidine compounds as Trk kinase inhibitors
US10251889B2 (en) 2009-07-09 2019-04-09 Array BioPharm Inc. Substituted pyrazolo[1,5-a]pyrimidine compounds as Trk kinase inhibitors
CN103739550A (en) * 2014-01-02 2014-04-23 中国药科大学 2, 3-dimethyl-6-urea-2H-indazole compound and preparation method and application thereof
WO2017011776A1 (en) 2015-07-16 2017-01-19 Array Biopharma, Inc. Substituted pyrazolo[1,5-a]pyridine compounds as ret kinase inhibitors
US10174028B2 (en) 2015-07-16 2019-01-08 Array Biopharma Inc. Substituted pyrazolo[1,5-A]pyridine compounds as RET kinase inhibitors
US10174027B2 (en) 2015-07-16 2019-01-08 Array Biopharma Inc. Substituted pyrazolo[1,5-a]pyridine compounds as RET kinase inhibitors
US10023570B2 (en) 2015-07-16 2018-07-17 Array Biopharma Inc. Substituted pyrazolo[1,5-A]pyridine compounds as RET kinase inhibitors
US10138243B2 (en) 2015-07-16 2018-11-27 Array Biopharma Inc. Substituted pyrazolo[1,5-a]pyridine compounds as RET kinase inhibitors
DE102015012050A1 (en) * 2015-09-15 2017-03-16 Merck Patent Gmbh Compounds as ASIC inhibitors and their uses
WO2017045751A1 (en) * 2015-09-15 2017-03-23 Merck Patent Gmbh Compounds as asic inhibitors and uses thereof
US10907215B2 (en) 2015-10-26 2021-02-02 Loxo Oncology, Inc. Point mutations in TRK inhibitor-resistant cancer and methods relating to the same
US10724102B2 (en) 2015-10-26 2020-07-28 Loxo Oncology, Inc. Point mutations in TRK inhibitor-resistant cancer and methods relating to the same
WO2017176751A1 (en) 2016-04-04 2017-10-12 Loxo Oncology, Inc. Liquid formulations of (s)-n-(5-((r)-2-(2,5-difluorophenyl)-pyrrolidin-1-yl)-pyrazolo[1,5-a]pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide
US10045991B2 (en) 2016-04-04 2018-08-14 Loxo Oncology, Inc. Methods of treating pediatric cancers
US11191766B2 (en) 2016-04-04 2021-12-07 Loxo Oncology, Inc. Methods of treating pediatric cancers
US10668072B2 (en) 2016-04-04 2020-06-02 Loxo Oncology, Inc. Liquid formulations of (S)-N-(5-((R)-2-(2,5-difluorophenyl)-pyrrolidin-1-yl)-pyrazolo[1,5-a]pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide
US10137127B2 (en) 2016-04-04 2018-11-27 Loxo Oncology, Inc. Liquid formulations of (S)-N-(5-((R)-2-(2,5-difluorophenyl)-pyrrolidin-1-yl)-pyrazolo[1,5-A]pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide
US10588908B2 (en) 2016-04-04 2020-03-17 Loxo Oncology, Inc. Methods of treating pediatric cancers
US11484535B2 (en) 2016-04-04 2022-11-01 Loxo Oncology, Inc. Liquid formulations of (S)-N-(5-((R)-2-(2,5-difluorophenyl)-pyrrolidin-1-yl)-pyrazolo[1,5-a] pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide
WO2017176744A1 (en) 2016-04-04 2017-10-12 Loxo Oncology, Inc. Methods of treating pediatric cancers
US11214571B2 (en) 2016-05-18 2022-01-04 Array Biopharma Inc. Process for the preparation of (S)-N-(5-((R)-2-(2,5-difluorophenyl)pyrrolidin-1-yl)-pyrazolo[1,5-a]pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide and salts thereof
US10555944B2 (en) 2016-10-10 2020-02-11 Eli Lilly And Company Substituted pyrazolo[1,5-A]pyridine compounds as RET kinase inhibitors
US10953005B1 (en) 2016-10-10 2021-03-23 Array Biopharma Inc. Substituted pyrazolo[1,5-a]pyridine compounds as RET kinase inhibitors
US11648243B2 (en) 2016-10-10 2023-05-16 Array Biopharma Inc. Substituted pyrazolo[1,5-A]pyridine compounds as RET kinase inhibitors
EP4144735A1 (en) 2016-10-10 2023-03-08 Array Biopharma, Inc. Substituted pyrazolo[1,5-a]pyridine compounds as ret kinase inhibitors
WO2018071454A1 (en) 2016-10-10 2018-04-19 Andrews Steven W Substituted pyrazolo[1,5-a]pyridine compounds as ret kinase inhibitors
WO2018071447A1 (en) 2016-10-10 2018-04-19 Andrews Steven W Substituted pyrazolo[1,5-a]pyridine compounds as ret kinase inhibitors
US10441581B2 (en) 2016-10-10 2019-10-15 Array Biopharma Inc. Substituted pyrazolo[1,5-A]pyridine compounds as RET kinase inhibitors
EP3753939A1 (en) 2016-10-10 2020-12-23 Array Biopharma Inc. Substituted pyrazolo[1,5-a]pyridine compounds as ret kinase inhibitors
US10172845B2 (en) 2016-10-10 2019-01-08 Array Biopharma Inc. Substituted pyrazolo[1,5-A]pyridine compounds as RET kinase inhibitors
US10144734B2 (en) 2016-10-10 2018-12-04 Array Biopharma Inc. Substituted pyrazolo[1,5-A]pyridine compounds as RET kinase inhibitors
US10112942B2 (en) 2016-10-10 2018-10-30 Array Biopharma Inc. Substituted pyrazolo[1,5-A]pyridine compounds as RET kinase inhibitors
US10137124B2 (en) 2016-10-10 2018-11-27 Array Biopharma Inc. Substituted pyrazolo[1,5-a]pyridine compounds as RET kinase inhibitors
US10172851B2 (en) 2016-10-10 2019-01-08 Array Biopharma Inc. Substituted pyrazolo[1,5-A]pyridine compounds as RET kinase inhibitors
US10881652B2 (en) 2016-10-10 2021-01-05 Array Biopharma Inc. Substituted pyrazolo[1,5-A]pyridine compounds as RET kinase inhibitors
US11091486B2 (en) 2016-10-26 2021-08-17 Array Biopharma, Inc Process for the preparation of pyrazolo[1,5-a]pyrimidines and salts thereof
WO2018136663A1 (en) 2017-01-18 2018-07-26 Array Biopharma, Inc. Ret inhibitors
US11168090B2 (en) 2017-01-18 2021-11-09 Array Biopharma Inc. Substituted pyrazolo[1,5-a]pyrazines as RET kinase inhibitors
US11851434B2 (en) 2017-01-18 2023-12-26 Array Biopharma Inc. Substituted pyrazolo[1,5-A]pyrazine compounds as ret kinase inhibitors
WO2018136661A1 (en) 2017-01-18 2018-07-26 Andrews Steven W SUBSTITUTED PYRAZOLO[1,5-a]PYRAZINE COMPOUNDS AS RET KINASE INHIBITORS
WO2018170381A1 (en) 2017-03-16 2018-09-20 Andrews Steven W Macrocyclic compounds as ros1 kinase inhibitors
US10688100B2 (en) 2017-03-16 2020-06-23 Array Biopharma Inc. Macrocylic compounds as ROS1 kinase inhibitors
US10966985B2 (en) 2017-03-16 2021-04-06 Array Biopharma Inc. Macrocyclic compounds as ROS1 kinase inhibitors
WO2019075114A1 (en) 2017-10-10 2019-04-18 Mark Reynolds Formulations comprising 6-(2-hydroxy-2-methylpropoxy)-4-(6-(6-((6-methoxypyridin-3-yl)methyl)-3,6-diazab icyclo[3.1.1]heptan-3-yl)pyridin-3-yl)pyrazolo[1,5-a]pyridine-3-carbonitrile
WO2019075108A1 (en) 2017-10-10 2019-04-18 Metcalf Andrew T Crystalline forms
US11524963B2 (en) 2018-01-18 2022-12-13 Array Biopharma Inc. Substituted pyrazolo[3,4-d]pyrimidines as RET kinase inhibitors
WO2019143977A1 (en) 2018-01-18 2019-07-25 Array Biopharma Inc. Substituted pyrrolo[2,3-d]pyrimidines compounds as ret kinase inhibitors
US11603374B2 (en) 2018-01-18 2023-03-14 Array Biopharma Inc. Substituted pyrrolo[2,3-d]pyrimidines compounds as ret kinase inhibitors
US11472802B2 (en) 2018-01-18 2022-10-18 Array Biopharma Inc. Substituted pyrazolyl[4,3-c]pyridine compounds as RET kinase inhibitors
WO2019143994A1 (en) 2018-01-18 2019-07-25 Array Biopharma Inc. Substituted pyrazolyl[4,3-c]pyridinecompounds as ret kinase inhibitors
WO2020028258A1 (en) 2018-07-31 2020-02-06 Loxo Oncology, Inc. Spray-dried dispersions and formulations of (s)-5-amino-3-(4-((5-fluoro-2-methoxybenzamido)methyl)phenyl)-1-(1,1,1-trifluoro propan-2-yl)-1h-pyrazole-4-carboxamide
US11964988B2 (en) 2018-09-10 2024-04-23 Array Biopharma Inc. Fused heterocyclic compounds as RET kinase inhibitors
WO2020055672A1 (en) 2018-09-10 2020-03-19 Array Biopharma Inc. Fused heterocyclic compounds as ret kinase inhibitors
WO2020131674A1 (en) 2018-12-19 2020-06-25 Array Biopharma Inc. 7-((3,5-dimethoxyphenyl)amino)quinoxaline derivatives as fgfr inhibitors for treating cancer
WO2020131627A1 (en) 2018-12-19 2020-06-25 Array Biopharma Inc. Substituted pyrazolo[1,5-a]pyridine compounds as inhibitors of fgfr tyrosine kinases
WO2020212530A1 (en) * 2019-04-17 2020-10-22 Rottapharm Biotech S.R.L. Benzo- and pyrido-pyrazoles as protein kinase inhibitors
EP3725777A1 (en) * 2019-04-17 2020-10-21 Rottapharm Biotech S.r.l. Benzo- and pyrido-pyrazoles as protein kinase inhibitors
WO2021134004A1 (en) 2019-12-27 2021-07-01 Schrodinger, Inc. Cyclic compounds and methods of using same
WO2022055963A1 (en) 2020-09-10 2022-03-17 Schrödinger, Inc. Heterocyclic pericondensed cdc7 kinase inhibitors for the treatment of cancer
WO2022164789A1 (en) 2021-01-26 2022-08-04 Schrödinger, Inc. Tricyclic compounds useful in the treatment of cancer, autoimmune and inflammatory disoders
WO2022197898A1 (en) 2021-03-18 2022-09-22 Schrödinger, Inc. Cyclic compounds and methods of using same
WO2024088829A2 (en) 2022-10-25 2024-05-02 Nerviano Medical Sciences S.R.L. Salts and polymorphs of 4-(4-methyl-piperazin-1-yl)-n-{6-[2-(4-trifluoromethyl-benzyloxy)-ethoxy]-1h-indazol-3-yl}-benzamide

Also Published As

Publication number Publication date
EA023579B1 (en) 2016-06-30
PT2707359T (en) 2017-01-09
AR086357A1 (en) 2013-12-04
US10478423B2 (en) 2019-11-19
DK2707359T3 (en) 2017-01-23
AU2012252468B2 (en) 2017-03-16
CN103534239B (en) 2016-04-20
IL229252A (en) 2017-05-29
US20140080807A1 (en) 2014-03-20
EP2707359B1 (en) 2016-11-30
US20180289672A1 (en) 2018-10-11
BR112013028733B1 (en) 2022-05-10
US10028934B2 (en) 2018-07-24
US9597317B2 (en) 2017-03-21
US9408850B2 (en) 2016-08-09
CN103534239A (en) 2014-01-22
CA2835478C (en) 2019-02-05
ZA201309345B (en) 2015-03-25
SG194911A1 (en) 2013-12-30
ES2611779T3 (en) 2017-05-10
TW201249806A (en) 2012-12-16
CL2013003227A1 (en) 2014-06-27
US20160310465A1 (en) 2016-10-27
MX2013012981A (en) 2013-12-12
HK1193812A1 (en) 2014-10-03
IL229252A0 (en) 2014-01-30
JP2014514353A (en) 2014-06-19
TWI573790B (en) 2017-03-11
KR20140037871A (en) 2014-03-27
PL2707359T3 (en) 2017-05-31
EA201391682A1 (en) 2014-04-30
MX342509B (en) 2016-10-03
CA2835478A1 (en) 2012-11-15
HUE031166T2 (en) 2017-07-28
BR112013028733A2 (en) 2017-08-29
EP2707359A1 (en) 2014-03-19
KR101953272B1 (en) 2019-02-28
AU2012252468A1 (en) 2014-01-09
SI2707359T1 (en) 2017-02-28
JP6396210B2 (en) 2018-09-26
US20160311776A1 (en) 2016-10-27

Similar Documents

Publication Publication Date Title
US10478423B2 (en) Substituted indazole derivatives active as kinase inhibitiors
US10081622B2 (en) Substituted indazole derivatives active as kinase inhibitors
KR20170031241A (en) 2-h-indazole derivatives as cyclin-dependent kinase (cdk) inhibitors and therapeutic uses thereof
KR20140027902A (en) Substituted pyrazolo-quinazoline derivatives as kinase inhibitors
TW201922724A (en) Substituted 3-phenylquinazolin-4(3H)-ones and uses thereof

Legal Events

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

Ref document number: 201280023142.9

Country of ref document: CN

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

Ref document number: 12718275

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: MX/A/2013/012981

Country of ref document: MX

ENP Entry into the national phase

Ref document number: 2835478

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 14116512

Country of ref document: US

ENP Entry into the national phase

Ref document number: 2014509702

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

REEP Request for entry into the european phase

Ref document number: 2012718275

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2012718275

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 201391682

Country of ref document: EA

ENP Entry into the national phase

Ref document number: 20137033115

Country of ref document: KR

Kind code of ref document: A

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112013028733

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 2012252468

Country of ref document: AU

Date of ref document: 20120507

Kind code of ref document: A

REG Reference to national code

Ref country code: BR

Ref legal event code: B01E

Ref document number: 112013028733

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 112013028733

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20131107