EP1127051A2 - Derives de pyrazole tricyclique - Google Patents

Derives de pyrazole tricyclique

Info

Publication number
EP1127051A2
EP1127051A2 EP99962700A EP99962700A EP1127051A2 EP 1127051 A2 EP1127051 A2 EP 1127051A2 EP 99962700 A EP99962700 A EP 99962700A EP 99962700 A EP99962700 A EP 99962700A EP 1127051 A2 EP1127051 A2 EP 1127051A2
Authority
EP
European Patent Office
Prior art keywords
phenyl
group
dihydroindeno
alkyl
optionally substituted
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP99962700A
Other languages
German (de)
English (en)
Inventor
Kevin J. Doyle
Paul Rafferty
Robert W. Steele
David J. Wilkins
Michael Hockley
Lee D. Arnold
Anna M. Ericsson
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Abbott GmbH and Co KG
Original Assignee
BASF SE
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by BASF SE filed Critical BASF SE
Publication of EP1127051A2 publication Critical patent/EP1127051A2/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • 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
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/08Drugs for genital or sexual disorders; Contraceptives for gonadal disorders or for enhancing fertility, e.g. inducers of ovulation or of spermatogenesis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/06Antiglaucoma agents or miotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/14Drugs for disorders of the endocrine system of the thyroid hormones, e.g. T3, T4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/10Antioedematous agents; Diuretics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • 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
    • 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
    • C07D401/04Heterocyclic 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 directly linked by a ring-member-to-ring-member bond
    • 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
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/02Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D409/04Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/02Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D495/04Ortho-condensed systems

Definitions

  • This invention relates to certain 3-aryl or 3-heteroaryl pyrazoles with 4,5(3,4)-bicyclic ring fusion which are inhibitors of protein kinases, particularly tyrosine kinases and serine/threonine kinases, of which some are novel compounds, to pharmaceutical compositions containing these pyrazoles and to processes for preparing these pyrazoles.
  • tyrosine kinases There are at least 400 enzymes identified as protein kinases. These enzymes catalyze the phosphorylation of target protein substrates. The phosphorylation is usually a transfer reaction of a phosphate group from ATP to the protein substrate. The specific structure in the target substrate to which the phosphate is transferred is a tyrosine, serine or threonine residue. Since these amino acid residues are the target structures for the phosphoryl transfer, these protein kinase enzymes are commonly referred to as tyrosine kinases or serine/threonine kinases.
  • the phosphorylation reactions, and counteracting phosphatase reactions, at the tyrosine, serine and threonine residues are involved in countless cellular processes that underlie responses to diverse intracellular signals (typically mediated through cellular receptors), regulation of cellular functions, and activation or deactivation of cellular processes.
  • a cascade of protein kinases often participate in intracellular signal transduction and are necessary for the realization of these cellular processes. Because of their ubiquity in these processes, the protein kinases can be found as an integral part of the plasma membrane or as cytoplasmic enzymes or localized in the nucleus, often as components of enzyme complexes. In many instances, these protein kinases are an essential element of enzyme and structural protein complexes that determine where and when a cellular process occurs within a cell.
  • Protein Tyrosine Kinases Protein tyrosine kinases.
  • PTKs Protein tyrosine kinases
  • This post-translational modification of these substrate proteins often enzymes themselves, acts as a molecular switch regulating cell proliferation, activation or differentiation (for review, see Schlessinger and Ulrich, 1992, Neuron 9:383-391).
  • Aberrant or excessive PTK activity has been observed in many disease states including benign and malignant proliferative disorders as well as diseases resulting from inappropriate activation of the immune system (e.g., autoimmune disorders), allograft rejection, and graft vs. host disease.
  • endothelial-cell specific receptor PTKs such as KDR and Tie-2 mediate the angiogenic process, and are thus involved in supporting the progression of cancers and other diseases involving inappropriate vascularization (e.g., diabetic retinopathy, choroidal neovascularization due to age-related macular degeneration, psoriasis, arthritis, retinopathy of prematurity, infantile hemangiomas).
  • inappropriate vascularization e.g., diabetic retinopathy, choroidal neovascularization due to age-related macular degeneration, psoriasis, arthritis, retinopathy of prematurity, infantile hemangiomas.
  • Tyrosine kinases can be of the receptor-type (having extracellular, transmembrane and intracellular domains) or the non-receptor type (being wholly intracellular).
  • RTKs Receptor Tyrosine Kinases
  • the RTKs comprise a large family of transmembrane receptors with diverse biological activities. At present, at least nineteen (19) distinct RTK subfamilies have been identified.
  • the receptor tyrosine kinase (RTK) family includes receptors that are crucial for the growth and differentiation of a variety of cell types (Yarden and Ullrich, Ann. Rev. Biochem. 57:433-478, 1988; Ullrich and Schlessinger, Cell 61:243-254, 1990).
  • RTKs The intrinsic function of RTKs is activated upon ligand binding, which results in phosphorylation of the receptor and multiple cellular substrates, and subsequently in a variety of cellular responses (Ullrich & Schlessinger, 1990, Cell 61 :203-212).
  • receptor tyrosine kinase mediated signal transduction is initiated by extracellular interaction with a specific growth factor (ligand), typically followed by receptor dimerization, stimulation of the intrinsic protein tyrosine kinase activity and receptor trans- phosphorylation.
  • Binding sites are thereby created for intracellular signal transduction molecules and lead to the formation of complexes with a spectrum of cytoplasmic signaling molecules that facilitate the appropriate cellular response (e.g., cell division, differentiation, metabolic effects, changes in the extracellular microenvironment); see Schlessinger and Ullrich, 1992, Neuron 9: 1-20.
  • Proteins with SH2 (src homology -2) or phosphotyrosine binding (PTB) domains bind activated tyrosine kinase receptors and their substrates with high affinity to propagate signals into cells. Both of the domains recognize phosphotyrosine (Fantl et al., 1992, Cell 69:413-423; Songyang et al., 1994, Mol. Cell. Biol 14:2777-2785; Songyang et al., 1993, Cell 72:767-778; and Koch et al, 1991, Science 252:668-678; Shoelson, Curr. Opin. Chem. Biol. (1997), 1(2), 227- 234; Cowburn, Curr. Opin.
  • RTKs receptor tyrosine kinases
  • FLK-1 fetal liver kinase 1
  • KDR kinase insert domain-containing receptor
  • FLK-1/KDR vascular endothelial cell growth factor receptor 2
  • VEGFR-2 vascular endothelial cell growth factor receptor 2
  • DNAs encoding mouse, rat and human FLK-1 have been isolated, and the nucleotide and encoded amino acid sequences reported (Matthews et al, Proc. Natl. Acad. Sci. USA, 88:9026-30, 1991; Terman et al, 1991, supra; Terman et al, Biochem. Biophys. Res. Comm.
  • VEGF and FLK-1 /KDR/VEGFR-2 are a ligand-receptor pair that play an important role in the proliferation of vascular endothelial cells, and formation and sprouting of blood vessels, termed vasculogenesis and angiogenesis, respectively.
  • Flt-1 Flt-1-like tyrosine kinase- 1
  • FLK-1/KDR FLK-1/KDR
  • Flt-1 vascular endothelial cell growth factor receptor 1
  • VEGF vascular endothelial cell growth factor
  • VEGF vascular endothelial cell growth factor
  • VEGF vascular endothelial cell growth factor
  • Flt-1 expression is associated with early vascular development in mouse embryos, and with neovascularization during wound healing (Mustonen and Alitalo, supra). Expression of Flt-1 in adult organs such as kidney glomeruli suggests an additional function for this receptor that is not related to cell growth (Mustonen and Alitalo, supra).
  • VEGF plays a role in the stimulation of both normal and pathological angiogenesis (Jakeman et al, Endocrinology 133: 848-859, 1993; Kolch et al, Breast Cancer Research and Treatment 36: 139-155, 1995; Ferrara et al, Endocrine Reviews 18(1); 4-25, 1997; Ferrara et al., Regulation of Angiogenesis (ed. L. D. Goldberg and E.M. Rosen), 209-232, 1997).
  • VEGF has been implicated in the control and enhancement of vascular permeability (Connolly, et al, J. Biol. Chem. 264: 20017- 20024, 1989; Brown et al, Regulation of Angiogenesis (ed. L.D. Goldberg and E.M. Rosen), 233-269, 1997).
  • VEGF vascular endothelial growth factor
  • VEGF vascular endothelial growth factor
  • Placenta growth factor has an amino acid sequence that exhibits significant homology to the VEGF sequence (Park et al, J. Biol. Chem. 269:25646- 54, 1994; Maglione et al. Oncogene 8:925-31, 1993).
  • PIGF Placenta growth factor
  • different species of PIGF arise from alternative splicing of mRNA, and the protein exists in dimeric form (Park et al, supra).
  • P1GF-1 and P1GF-2 bind to Flt-1 with high affinity, and P1GF-2 also avidly binds to neuropilin-1 (Migdal et al, J. Biol. Chem.
  • PIGF has been reported to potentiate both the vascular permeability and mitogenic effect of VEGF on endothelial cells when VEGF is present at low concentrations (purportedly due to heterodimer formation) (Park et al, supra).
  • VEGF-B is produced as two isofoims (167 and 185 residues) that also appear to bind Flt-l/VEGFR-1. It may play a role in the regulation of extracellular matrix degradation, cell adhesion, and migration through modulation of the expression and activity of urokinase type plasminogen activator and plasminogen activator inhibitor 1 (Pepper et al, Proc. Natl Acad. Sci. U. S. A. (1998), 95(20): 11709-11714).
  • VEGF-C was originally cloned as a ligand for VEGFR-3/Flt-4 which is primarily expressed by lymphatic endothelial cells.
  • VEGF-C can also bind KDRNEGFR-2 and stimulate proliferation and migration of endothelial cells in vitro and angiogenesis in in vivo models ( Lymboussaki et al, Am. J. Pathol. (1998), 153(2): 395-403; Witzenbichler et al, Am. J. Pathol. (1998), 153(2), 381-394).
  • the transgenic overexpression of VEGF-C causes proliferation and enlargement of only lymphatic vessels, while blood vessels are unaffected.
  • the expression of VEGF-C is not induced by hypoxia (Ristimaki et al, J. Biol. Chem. (1998), 273(14),8413-8418).
  • VEGF-D is structurally very similar to VEGF- C.
  • VEGF-D is reported to bind and activate at least two VEGFRs, VEGFR-3/Flt-4 and KDR/VEGFR-2. It was originally cloned as a c-fos inducible mitogen for fibroblasts and is most prominently expressed in the mesenchymal cells of the lung and skin (Achen et al, Proc. Natl. Acad. Sci. U. S. A. (1998), 95(2), 548-553 and references therein).
  • VEGF-C and VEGF-D have been claimed to induce increases in vascular permeability in vivo in a Miles assay when injected into cutaneous tissue
  • VEGF homologs may involve formation of VEGF ligand heterodimers, and/or heterodimerization of receptors, or binding to a yet undiscovered VEGFR (Witzenbichler et al, supra).
  • VEGFR VEGFR-3/FU-4
  • receptors other than KDR/VEGFR-2 may be responsible for the induction of vascular permeability
  • the Non-Receptor Tyrosine Kinases represent a collection of cellular enzymes which lack extracellular and transmembrane sequences. At present, over twenty-four individual non-receptor tyrosine kinases, comprising eleven (11) subfamilies (Src, Frk, Btk, Csk, Abl, Zap70, Fes/Fps, Fak, Jak, Ack and LIMK) have been identified.
  • the Src subfamily of non-receptor tyrosine kinases is comprised of the largest number of PTKs and include Src, Yes, Fyn, Lyn, Lck, Blk, Hck, Fgr and Yrk.
  • the Src subfamily of enzymes has been linked to oncogenesis.
  • a more detailed discussion of non-receptor tyrosine kinases is provided in Bolen, 1993, Oncogene 8:2025-2031, which is incorporated herein by reference.
  • tyrosine kinases whether an RTK or non-receptor tyrosine kinase, have been found to be involved in cellular signaling pathways involved in numerous pathogenic conditions, including cancer, psoriasis, and other hyperproliferative disorders or hyper-immune responses.
  • RNA ligands linek, et al, Biochemistry 33:10450- 56; Takano, et al, 1993, Mol. Bio. Cell 4:358A; Kinsella, et al. 1992, Exp. Cell Res. 199:56-62; Wright, et al, 1992, J. Cellular Phys. 152:448-57
  • tyrosine kinase inhibitors WO 94/03427; WO 92/21660; WO 91/15495; WO 94/14808; U.S. Patent No. 5,330,992; Mariani, et al, 1994, Proc.
  • PCT WO 94/03427 selenoindoles and selenides
  • PCT WO 92/21660 tricyclic polyhydroxylic compounds
  • PCT WO 91/15495 benzylphosphonic acid compounds
  • Anilinocinno lines PCT WO97/34876
  • quinazoline derivative compounds PCT WO97/22596; PCT WO97/42187
  • the identification of effective small compounds which specifically inhibit signal transduction by modulating the activity of receptor and non-receptor tyrosine and serine/threonine kinases to regulate and modulate abnormal or inappropriate cell proliferation, differentiation, or metabolism is therefore desirable.
  • the identification of methods and compounds that specifically inhibit the function of a tyrosine kinase which is essential for angiogenic processes or the formation of vascular hyperpermeability leading to edema, ascites, effusions, exudates, and macromolecular extravasation and matrix deposition as well as associated disorders would be beneficial.
  • the present invention provides a method of inhibiting the kinase activity of tyrosine kinases and serine/threonine kinases comprising the administration of a compound represented by formula I :
  • R represents H ;
  • R 2 represents aryl, pyridyl, thienyl, furyl or pyrrolyl each of which is optionally substituted;
  • R3 , R4 , R5 and R6 independently represent a) H, b) halo, c) a Ci-6 alkyl group optionally substituted by one or more of the following: hydroxy, halo or an amino group of formula NRhRj wherein R and Rj are as defined below, d) a C,.
  • R2 when X represents carbonyl or a substituted methylene, Ri is hydrogen and R3, R4, R5 and R6 each represent hydrogen, then R2 is not pyridyl, phenyl or phenyl substituted by Ci-2 alkyl, a halogen atom, a lower alkoxy group, a hydroxyl group or an amino group;
  • R2 when X is a group of formula (CH 2 ) n in which n is 1 , 2 or 3, Ri is hydrogen and two of R3, R4, R5 and R6 independently represent hydrogen, halogen having an atomic weight of about 19 to 36, Ci .4 alkyl, Ci-4 alkoxy or trifluoromethyl and the other two represent hydrogen, then R2 is not thienyl, furyl, pyrrolyl, pyridyl, each of which is unsubstituted, or phenyl having two or less substituents wherein the substituents are halogen having an atomic weight of about 19 to 36, Ci-4 alkyl, C1.4 alkoxy or trifluoromethyl;
  • R3 , R4 , R5 and R independently represent a) H, b) halo, c) a Cl-6 alkyl group optionally substituted by one or more of the following: hydroxy, halo or an amino group of formula NRhRj wherein Rh and Rj are as defined below, d) a C 6 alkoxy group optionally substituted by one or more of the following: hydroxy, a C ] .
  • X represents S and R , R3, R4, R5 and R6 each represent hydrogen and R2 represents 2,4-dichlorophenyl is disclosed in Monatsh Chem. 1974, 105. 869.
  • WO97/15308 discloses that 3-(4-methylphenyl)indeno[l,2-c]pyrazol-4(l ⁇ )- one oxime may be used to treat bone deficit conditions. 3-(3,4-Dimethoxyphenyl)- indeno[l,2-c]pyrazol-4(lH)-one oxime is commercially available.
  • the present invention provides a first group of novel compounds of formula I and pharmaceutically acceptable salts thereof in which:
  • X represents a group of formula S(O) p in which p represents 0,1 or 2 ;
  • Ri represents H ;
  • R 2 represents aryl, pyridyl, thienyl, furyl or pyrrolyl each of which is optionally substituted; and R 3 , R4, R5 and R6 are as previously defined; with the provisos that 1) when Ri, R3, R4, R5 and R6 each represent hydrogen and X represents SO 2 , then R 2 does not represent phenyl and
  • the present invention provides a second group of novel compounds of formula I and pharmaceutically acceptable salts thereof in which:
  • X represents oxygen
  • R ! represents H ;
  • R represents aryl, pyridyl, thienyl, furyl or pyrrolyl each of which is optionally substituted;
  • R3, R4, R5 and R6 are as previously defined; with the proviso that when Ri, R3, R4, R5 and R6 each represent hydrogen, then R 2 does not represent phenyl, 2,4-dimethylphenyl or 2,4-dichlorophenyl.
  • the present invention provides a third group of novel compounds and pharmaceutically acceptable salts thereof in which:
  • X represents a group of formula NR 8 ;
  • R x represents H ;
  • R 2 represents aryl, pyridyl, thienyl, furyl or pyrrolyl each of which is optionally substituted;
  • R3, R4, R5 and R6 are as previously defined.
  • the present invention provides a fourth group of novel compounds of formula I and pharmaceutically acceptable salts thereof in which:
  • R j represents H ;
  • R 2 represents aryl, pyridyl, thienyl, furyl or pyrrolyl each of which is optionally substituted;
  • the present invention provides a fifth group of novel compounds of formula I and pharmaceutically acceptable salts thereof in which: X represents substituted methylene or a carbonyl group; R, represents H ;
  • R 2 represents aryl, pyridyl, thienyl, furyl or pyrrolyl each of which is optionally substituted; and R3 , R4, R5 and R6 are as previously defined; with the provisos that
  • R2 when X represents carbonyl or a substituted methylene, Ri is hydrogen and R3, R4, R5 and R6 each represent hydrogen, then R2 is not pyridyl, 2- thienyl, 3-thienyl, phenyl or phenyl substituted by C 1-2 alkyl, a halogen atom, a lower alkoxy group, a hydroxyl group or an amino group;
  • Ri is hydrogen and two of R3, R4, R5 and R independently represent hydrogen, halogen having an atomic weight of about 19 to 36, C 1-4 alkyl, Ci-4 alkoxy or trifluoromethyl and the other two represent hydrogen, then R2 is not thienyl, furyl, pyrrolyl, pyridyl, each of which is unsubstituted, or phenyl having two or less substituents wherein the substituents are halogen having an atomic weight of about 19 to 36, C 1-4 alkyl, Ci-4 alkoxy or trifluoromethyl; and
  • R3 when X represents a carbonyl group, R2 represents phenyl, 4-chlorophenyl or 4-methoxyphenyl, then R3, R4, R5 and R6 are not trifluoromethyl;
  • the present invention provides a sixth group of novel compounds of formula I and pharmaceutically acceptable salts thereof in which X represents a group of formula (CH2)n in which n is 1 , 2 or 3
  • R[ represents H ;
  • R 2 represents aryl, pyridyl, thienyl, furyl or pyrrolyl each of which is optionally substituted;
  • R 3 , R 4 , R 5 and R 6 are as previously defined with the proviso that when X is a group of formula (CH 2 ) classroom in which n is 1, 2 or 3,
  • R is hydrogen and two of R3, R4, R5 and R6 independently represent hydrogen, halogen having an atomic weight of about 19 to 36, Ci-4 alkyl, C1-4 alkoxy or trifluoromethyl and the other two represent hydrogen, then R2 is not thienyl, furyl, pyrrolyl, pyridyl, each of which is unsubstituted, or phenyl having two or less substituents wherein the substituents are halogen having an atomic weight of about
  • R2 does not represent 3-carboxypyrid-2-yl, 3-methoxycarbonylpyrid-2-yl or 2-carboxy- phenyl
  • the present invention provides a seventh group of novel compounds of formula I and pharmaceutically acceptable salts thereof in which :
  • X represents a) substituted methylene b) carbonyl , c) oxygen , d) a group of formula
  • -C NOR7 in which R7 represents H or a Ci-4 alkyl group, e) a group of formula R.8 in which R ⁇ represents H, an optionally substituted Ci-4 alkyl group or optionally substituted phenyl, f) a group of formula (CH 2 ) n in which n is 1, 2 or 3, or g) a group of formula S(O)p in which p is 0, 1 or 2;
  • R represents H ;
  • R 2 represents aryl, pyridyl, thienyl, furyl or pyrrolyl each of which is optionally substituted;
  • R3 , R4 , R5 and R6 independently represent a) H, b) halo, c) a Cl-6 alkyl group optionally substituted by one or more of the following: hydroxy, halo or an amino group of formula NRhRj wherein Rh and Rj are as defined below, d) a C,_ 6 alkoxy group optionally substituted by one or more of the following: hydroxy, or an amino group of formula NRhRj wherein Rh and Rj are as defined below provided that these groups are not attached to the carbon which is attached to the oxygen of the alkoxy group; or halo e) optionally substituted phenoxy, f) hydroxy, g) a group formula COR a in which R a represents hydroxy, a Ci-6 alkoxy group or R a represents a group of formula NRb Re in which
  • R3 , R4 , R5 and R 6 or a substituent on R 2 represents one of the following: a) a group of formula O(CH2)m Rg in which m is 2, 3, 4 or 5 and Rg represents hydroxy or a group of formula NRdRe in which R and R e are as defined above; or Rg represents a group of formula COR a wherein R a is as defined above and m is 1, 2, 3, 4 or 5, b) a Ci-6 alkyl group substituted by one or more of the following: hydroxy or an amino group of formula NRhRj wherein Rh and Rj are as defined above, c) a C,.
  • aryl as used herein means phenyl or naphthyl.
  • optionally substituted as used herein means substituted by one or more of the following a) halo, b) a Ci- lkyl group optionally substituted by one or more of the following: hydroxy, halo or an amino group of formula NRhRj wherein Rh and Rj are as defined below, c) a C,_ 6 alkoxy group optionally substituted by one or more of the following: hydroxy, halo or an amino group of formula NRhRj wherein Rh and Rj are as defined below provided that these groups are not attached to the carbon which is attached to the oxygen of the alkoxy group, d) optionally substituted phenoxy, e) hydroxy, f) a group formula COR in which R represents hydroxy, a Ci-6 alkoxy group or R a represents a group of formula NRb Rc in which Rb and R c independently represent hydrogen, a Ci-12 alkyl group , a
  • the term "optionally substituted” as used herein means substituted by one or more of the following: a) halo, b) a Ci- alkyl group optionally substituted by one or more of the following: hydroxy, halo or an amino group of formula NRhRj wherein Rh and Rj are as defined below, c) a C [ .
  • C 6 )alkyl mono- or di-(C,-C 6 )alkylamino(C,-C 6 )alkyl, mo holinyl-(C,- C 6 )alkyl, pyrrolidinyl-(C 1 -C 6 )alkyl, pyridinyl, phenyl(C 0 -C 6 )alkyl where the phenyl portion is optionally substituted by one or more moieties selected from the group consisting of halo, hydroxy, nitro, amino, mono- or di-(C,- C 6 )alkylamino, (C,-C 6 )alkyl and (C r C 6 )alkoxy; or Rh and Rj together with the nitrogen atom to which they are attached represent a four, five, six or seven membered heterocyclic ring which optionally contains one or more additional hetero atoms selected from O, S and N and is optionally substituted by a Ci-6 alkyl group or a hetero
  • R2 represents optionally substituted pyridyl
  • the pyridine ring may be in the form of its N-oxide.
  • NR Re represents a saturated heterocyclic ring
  • the ring is preferably morpholino, perhydrothiazin-4-yl, piperidino, pyrrolidin-1-yl, piperazin-1-yl or 4- methylpiperazin-4-yl.
  • substituted methylene means for example methylene substituted by one or more of the following: hydroxy or a C l .4 alkyl group wherein the alkyl group is optionally further substituted by a group of formula NR r Rs wherein R r and R s independently represent H or a Cl-6 alkyl group.
  • any group mentioned herein which contains a chain of three or more atoms signifies a group in which the chain may be straight or branched.
  • an alkyl group may comprise propyl, which includes n-propyl and isopropyl, and butyl, which includes n-butyl, sec-butyl, isobutyl and tert-butyl.
  • the term C3-12 cycloalkyl group includes bridged groups for example adamantyl.
  • the term 'halo' as used herein signifies fluoro, chloro, bromo and iodo.
  • Compounds of formula I may exist as salts with pharmaceutically acceptable acids.
  • the present invention includes such salts.
  • Examples of such salts include hydrochlorides, hydrobromides, sulphates, methanesulphonates, nitrates, maleates, acetates, citrates, fumarates, tartrates [eg (+)-tartrates, (-)-tartrates or mixtures thereof including racemic mixtures], succinates, benzoates and salts with amino acids such as glutamic acid.
  • These salts may be prepared by methods known to those skilled in the art.
  • Certain compounds of formula I which have acidic substituents may exist as salts with pharmaceutically acceptable bases.
  • the present invention includes such salts.
  • Example of such salts include sodium salts, potassium salts, lysine salts and arginine salts. These salts may be prepared by methods known to those skilled in the art.
  • Certain compounds of formula I and their salts may exist in more than one crystal form and the present invention includes each crystal form and mixtures thereof.
  • Certain compounds of formula I and their salts may also exist in the form of solvates, for example hydrates, and the present invention includes each solvate and mixtures thereof.
  • Certain compounds of formula I may contain one or more chiral centres, and exist in different optically active forms. When compounds of formula I contain one chiral centre, the compounds exist in two enantiomeric forms and the present invention includes both enantiomers and mixtures of enantiomers.
  • the enantiomers may be resolved by methods known to those skilled in the art, for example by formation of diastereoisomeric salts which may be separated, for example, by crystallization; formation of diastereoisomeric derivatives or complexes which may be separated, for example, by crystallization, gas-liquid or liquid chromatography; selective reaction of one enantiomer with an enantiomer-specific reagent, for example enzymatic esterification; or gas-liquid or liquid chromatography in a chiral environment, for example on a chiral support for example silica with a bound chiral ligand or in the presence of a chiral solvent.
  • enantiomers may be synthesized by asymmetric synthesis using optically active reagents, substrates, catalysts or solvents, or by converting one enantiomer into the other by asymmetric transformation.
  • a compound of formula I When a compound of formula I contains more than one chiral centre it may exist in diastereoisomeric forms.
  • the diastereoisomeric pairs may be separated by methods known to those skilled in the art, for example chromatography or crystallization and the individual enantiomers within each pair may be separated as described above.
  • the present invention includes each diastereoisomer of compounds of formula I and mixtures thereof.
  • Certain compounds of formula I may exist in different tautomeric forms or as different geometric isomers, and the present invention includes each tautomer and or geometric isomer of compounds of formula I and mixtures thereof.
  • Certain compounds of formula I may exist in different stable conformational forms which may be separable. Torsional asymmetry due to restricted rotation about an asymmetric single bond, for example because of steric hindrance or ring strain, may permit separation of different conformers.
  • the present invention includes each conformational isomer of compounds of formula I and mixtures thereof.
  • Certain compounds of formula I may exist in zwitterionic form and the present invention includes each zwitterionic form of compounds of formula I and mixtures thereof.
  • the compounds of this invention are useful as inhibitors of serine/ threonine and tyrosine kinases.
  • the compounds of this invention are useful as inhibitors of tyrosine kinases that are important in hyperproliferative diseases, especially in the process of angiogenesis. Since these compounds are anti- angiogenic, they are important substances for inhibiting the progression of disease states where angiogenesis is an important component. Preferred definitions of the substituents are now given.
  • R 2 represents optionally substituted phenyl, naphthyl, optionally substituted thienyl, optionally substituted pyridyl, optionally substituted furyl, or optionally substituted pyrrolyl.
  • R 2 represents 2-thienyl, 3-thienyl, 2-furyl, 3-furyl, 2-pyridyl, 3 -pyridyl or 4-pyridyl each of which is optionally substituted, naphthyl and optionally mono-, di- and tri-substituted phenyl, wherein the substituents are selected from optionally substituted alkoxy (particularly methoxy, 3- morpholinopropoxy, 2-morpholinoethoxy, 3-carboxypropoxy, carboxymethoxy, 2- carboxyethoxy, 2-carbamoylethoxy, carbamoylmethoxy, 3-carbamoylpropoxy, 2- piperidinoethoxy, 2-(piperazin-l-yl)ethoxy, 2-(pyrrolidin-l-yl)ethoxy, 2- dimethylaminoethoxy, 2-(perhydro-thiazin-4-yl)ethoxy, 3-piperidinopropoxy, 3- (piperazin-l, 3-
  • R 2 represents 4-pyridyl, 2-formamidomethyl-4-pyridyl, 2- aminomethyl-4-pyridyl, 2-(hydroxyamidino)-4-pyridyl, 2-carbamoyl-4-pyridyl, 4-pyridyl- ⁇ -oxide, 2-chloro-4-pyridyl, 2-cyano-4-pyridyl, 5-methyl-2-furyl, 5-(2- nitro-4-trifluoromethylphenyl)fur-2-yl, phenyl, 4-methoxyphenyl, 3-methoxyphenyl, 2-methoxyphenyl, 3,4-dimethoxyphenyl, 3,4,5-trimethoxyphenyl, 4-(3-morpholino- propoxy)phenyl, 4-(2-morpholinoethoxy)phenyl, 4-(3-carboxypropoxy)phenyl, 4- carboxymethoxyphenyl, 4-(3-carbamoylpropoxy)phenyl, 4-, 4-
  • R 3 , R 4 , R 5 and R 6 independently represent hydrogen , halo
  • optionally substituted lower alkoxy particularly methoxy, 3- mo ⁇ holinopropoxy, 2-mo ⁇ holinoethoxy, 3-carboxypropoxy, carboxymethoxy, 2- carboxyethoxy, 2-carbamoylethoxy, 3-carbamoylpropoxy, 2-piperidinoethoxy, 2-
  • R 3 , R 4 , R 5 and R 6 represent the following 6,7-dimethoxy, 6,7,8-trimethoxy, 6-fluoro, 6-acetamido, 7-methoxy, 6-carbamoyl, 6-(7V-methyl- carbamoyl), 6-( ⁇ -phenylcarbamoyl), (3-mo ⁇ holino)propoxy and 2-mo ⁇ holino)- ethoxy.
  • the present invention further includes the use of these compounds in pharmaceutical compositions with a pharmaceutically effective amount of the above- described compounds and a pharmaceutically acceptable carrier or excipient.
  • These pharmaceutical compositions can be administered to individuals to slow or halt the process of angiogenesis in angiogenesis-aided diseases, or to treat edema, effusions, exudates, or ascites and other conditions associated with vascular hype ⁇ ermeability.
  • the compounds of this invention have antiangiogenic properties. These antiangiogenic properties are due at least in part to the inhibition of protein tyrosine kinases essential for angiogenic processes. For this reason, these compounds can be used as active agents against such disease states as arthritis, atherosclerosis, psoriasis, hemangiomas, myocardial angiogenesis, coronary and cerebral collaterals, ischemic limb angiogenesis, wound healing, peptic ulcer Helicobacter related diseases, fractures, cat scratch fever, rubeosis, neovascular glaucoma and retinopathies such as those associated with diabetic retinopathy, retinopathy of prematurity, or age-related macular degeneration.
  • some of these compounds can be used as active agents against solid tumors, malignant ascites, hematopoietic cancers and hype ⁇ roliferative disorders such as thyroid hype ⁇ lasia (especially Grave's disease), and cysts (such as hypervascularity of ovarian stroma characteristic of polycystic ovarian syndrome (Stein-Leventhal syndrome)) since such diseases require a proliferation of blood vessel cells for growth and/or metastasis.
  • thyroid hype ⁇ lasia especially Grave's disease
  • cysts such as hypervascularity of ovarian stroma characteristic of polycystic ovarian syndrome (Stein-Leventhal syndrome)
  • some of these compounds can be used as active agents against burns, chronic lung disease, stroke, polyps, anaphylaxis, chronic and allergic inflammation, ovarian hyperstimulation syndrome, brain tumor-associated cerebral edema, high- altitude, trauma or hypoxia induced cerebral or pulmonary edema, ocular and macular edema, ascites, and other diseases where vascular hype ⁇ ermeability, effusions, exudates, protein extravasation, or edema is a manifestation of the disease.
  • the compounds will also be useful in treating disorders in which protein extravasation leads to the deposition of fibrin and extracellular matrix, promoting stromal proliferation (e.g. fibrosis, cirrhosis and ca ⁇ al tunnel syndrome).
  • VEGF's are unique in that they are the only angiogenic growth factors known to contribute to vascular hype ⁇ ermeability and the formation of edema. Indeed, vascular hype ⁇ ermeability and edema that is associated with the expression or administration of many other growth factors appears to be mediated via VEGF production. Inflammatory cytokines stimulate VEGF production. Hypoxia results in a marked upregulation of VEGF in numerous tissues, hence situations involving infarct, occlusion, ischemia, anemia, or circulatory impairment typically invoke NEGF/VPF mediated responses.
  • VEGF-mediated hype ⁇ ermeability can significantly contribute to disorders with these etiologic features. It is envisaged that the disorders listed above are mediated to a significant extent by protein tyrosine kinase activity involving the KDR/VEGFR-2 and/or the Flt-lNEGFR-1 tyrosine kinases.
  • the compounds of this invention have inhibitory activity against protein kinases. That is, these compounds modulate signal transduction by protein kinases.
  • Compounds of this invention inhibit protein kinases from serine/threonine and tyrosine kinase classes. In particular, these compounds selectively inhibit the activity of the KDR/FLK-1 /VEGFR-2 tyrosine kinases.
  • Certain compounds of this invention also inhibit the activity of additional tyrosine kinases such as Flt- 1NEGFR-1, Src-subfamily kinases such as Lck, Src, fyn, yes, etc.
  • some compounds of this invention significantly inhibit serine/threonine kinases such as CDKs which play an essential role in cell-cycle progression.
  • the potency and specificity of the generic compounds of this invention towards a particular protein kinase can often be altered and optimized by variations in the nature, number and arrangement of the substituents (i.e., Ri, R2, R3, R4, R5 and R6) and conformational restrictions.
  • the metabolites of certain compounds may also possess significant protein kinase inhibitory activity.
  • the compounds of this invention when administered to individuals in need of such compounds, inhibit vascular hype ⁇ ermeability and the formation of edema in these individuals. These compounds act, it is believed, by inhibiting the activity of KDR tyrosine kinase which is involved in the process of vascular hype ⁇ ermeability and edema formation.
  • the KDR tyrosine kinase may also be referred to as FLK-1 tyrosine kinase, NYK tyrosine kinase or VEGFR-2 tyrosine kinase.
  • KDR tyrosine kinase is activated when vascular endothelial cell growth factor (VEGF) or another activating ligand (such as VEGF-C, VEGF-D or HIV Tat protein) binds to a KDR tyrosine kinase receptor which lies on the surface of vascular endothelial cells.
  • VEGF vascular endothelial cell growth factor
  • another activating ligand such as VEGF-C, VEGF-D or HIV Tat protein
  • KDR tyrosine kinase stimulation results in the proliferation and chemotaxis of vascular endothelial cells and formation of new vessels.
  • KDR tyrosine kinase activity By inhibiting KDR tyrosine kinase activity, either by blocking the production of the activating ligand, by blocking the activating ligand binding to the KDR tyrosine kinase receptor, by preventing receptor dimerization and transphosphorylation, by inhibiting the enzyme activity of the KDR tyrosine kinase (inhibiting the phosphorylation function of the enzyme) or by some other mechanism that interrupts its downstream signaling (D. Mukhopedhyay et al, Cancer Res. 55:1278-1284 (1998) and references therein), hype ⁇ ermeability, as well as associated extravasation, subsequent edema formation and matrix deposition, and angiogenic responses, may be inhibited and minimized.
  • One group of preferred compounds of this invention have the property of inhibiting KDR tyrosine kinase activity without significantly inhibiting Flt-1 tyrosine kinase activity (Flt-1 tyrosine kinase is also referred to as VEGFR-1 tyrosine kinase). Both KDR tyrosine kinase and Flt-1 tyrosine kinase are activated by VEGF binding to KDR tyrosine kinase receptors and to Flt-1 tyrosine kinase receptors, respectively.
  • Certain preferred compounds of this invention are unique because they inhibit the activity of one VEGF-receptor tyrosine kinase (KDR) that is activated by activating ligands but do not inhibit other receptor tyrosine kinases, such as Flt-1, that are also activated by certain activating ligands.
  • KDR VEGF-receptor tyrosine kinase
  • the preferred compounds of this invention are, therefore, selective in their tyrosine kinase inhibitory activity.
  • the compounds of the present invention are also useful in the treatment of ulcers - bacterial, fungal, Mooren ulcers and ulcerative colitis.
  • the compounds of the present invention are also useful in the treatment of conditions wherein undesired angiogenesis, edema, or stromal deposition occurs in viral infections such as He ⁇ es simplex, He ⁇ es Zoster, AIDS, Kaposi's sarcoma, protozoan infections and toxoplasmosis, endometriosis, ovarian hyperstimulation syndrome, systemic lupus, sarcoidosis, synovitis, Crohn's disease, sickle cell anaemia, Lyme's disease, pemphigoid, Paget's disease, hyperviscosity syndrome, Osier- Weber-Rendu disease, chronic inflammation, chronic occlusive pulmonary disease, asthma, rheumatoid arthritis and osteoarthritis, and edema following trauma, radiation, or stroke.
  • the compounds of the present invention are also useful in the treatment of ocular conditions such as ocular and macular edema, ocular neovascular disease, scleritis, radial keratotomy, uveitis, vitritis, myopia, optic pits, chronic retinal detachment, post-laser complications, conjunctivitis, Stargardt's disease and Eales disease in addition to retinopathy and macular degeneration.
  • ocular conditions such as ocular and macular edema, ocular neovascular disease, scleritis, radial keratotomy, uveitis, vitritis, myopia, optic pits, chronic retinal detachment, post-laser complications, conjunctivitis, Stargardt's disease and Eales disease in addition to retinopathy and macular degeneration.
  • the compounds of the present invention are also useful in the treatment of cardiovascular conditions such as atherosclerosis, restenosis, vascular occlusion and carotid obstructive disease.
  • the compounds of the present invention are also useful in the treatment of cancer related indications such as solid tumors, sarcomas (especially Ewing's sarcoma and osteosarcoma), retinoblastoma, rhabdomyosarcomas, neuroblastoma, hematopoietic malignancies, including leukaemia and lymphoma, tumor-induced pleural or pericardial effusions, and malignant ascites.
  • cancer related indications such as solid tumors, sarcomas (especially Ewing's sarcoma and osteosarcoma), retinoblastoma, rhabdomyosarcomas, neuroblastoma, hematopoietic malignancies, including leukaemia and lymphoma, tumor-induced pleural or pericardial effusions, and malignant ascites.
  • the compounds of the present invention are also useful in the treatment of diabetic conditions such as glaucoma, diabetic retinopathy and microangiopathy.
  • the disorders listed above are mediated to a significant extent by protein tyrosine kinase activity involving the VEGF receptors (e.g. KDR and Flt-1).
  • VEGF receptors e.g. KDR and Flt-1
  • KDR and Flt-1 protein tyrosine kinase activity involving the VEGF receptors
  • the progression of the listed disorders is inhibited because the angiogenic component of the disease state is severely curtailed.
  • the action of the compounds of this invention by their selectivity for specific tyrosine kinases, result in a minimization of side effects that would occur if less selective tyrosine kinase inhibitors were used.
  • the present invention provides compounds of formula I as defined initially above (including the provisos) for use as medicaments, particularly as inhibitors of protein kinase activity for example tyrosine kinase activity, serine kinase activity and threonine kinase activity.
  • the present invention provides the use of compounds of formula I as defined initially above (including the provisos) in the manufacture of a medicament for use in the inhibition of protein kinase activity.
  • “Pharmaceutically acceptable salts” refers to those salts which retain the biological effectiveness and properties of the free bases and which are obtained by reaction with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid or organic acids such as sulfonic acid, carboxylic acid, organic phosphoric acid, methanesulfonic acid, ethanesulfonic acid, p- toluenesulfonic acid, salicylic acid, lactic acid, tartaric acid and the like.
  • inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid or organic acids such as sulfonic acid, carboxylic acid, organic phosphoric acid, methanesulfonic acid, ethanesulfonic acid, p- toluenesulfonic acid, salicylic acid, lactic acid, tartaric acid and the like.
  • Alkyl refers to a saturated aliphatic hydrocarbon, including straight-chain and branched-chain groups having 1 to 4 carbons.
  • Alkoxy refers to an "O-alkyl” group, where “alkyl” is defined as described above.
  • the compounds of this invention can be administered to a human patient by themselves or in pharmaceutical compositions where they are mixed with suitable carriers or excipient(s) at doses to treat or ameliorate vascular hype ⁇ ermeability, edema and associated disorders. Mixtures of these compounds can also be administered to the patient as a simple mixture or in suitable formulated pharmaceutical compositions.
  • a therapeutically effective dose further refers to that amount of the compound or compounds sufficient to result in the prevention or attenuation of inappropriate neovascularization, progression of hype ⁇ roliferative disorders, edema, VEGF-associated hype ⁇ ermeability and/or VEGF-related hypotension.
  • Suitable routes of administration may, for example, include oral, eyedrop, rectal, transmucosal, topical, or intestinal administration; parenteral delivery, including intramuscular, subcutaneous, intramedullary injections, as well as intrathecal, direct intraventricular, intravenous, intraperitoneal, intranasal, or intraocular inj ections.
  • compositions of the present invention may be manufactured in a manner that is itself known, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or lyophilizing processes.
  • compositions for use in accordance with the present invention thus may be formulated in conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen.
  • the agents of the invention may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hanks's solution, Ringer's solution, or physiological saline buffer.
  • physiologically compatible buffers such as Hanks's solution, Ringer's solution, or physiological saline buffer.
  • penetrants appropriate to the barrier to be permeated are used in the formulation.
  • the compounds can be formulated readily by combining the active compounds with pharmaceutically acceptable carriers well known in the art.
  • Such carriers enable the compounds of the invention to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a patient to be treated.
  • compositions for oral use can be obtained by combining the active compound with a solid excipient, optionally grinding a resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores.
  • suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, and/or polyvinylpyrrolidone (PVP).
  • disintegrating agents may be added, such as the cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
  • Dragee cores are provided with suitable coatings.
  • suitable coatings For this pu ⁇ ose, concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures.
  • Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
  • compositions which can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol.
  • the push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers.
  • the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols.
  • stabilizers may be added. All formulations for oral administration should be in dosages suitable for such administration.
  • compositions may take the form of tablets or lozenges formulated in conventional manner.
  • the compounds for use according to the present invention are conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebulizer, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • the dosage unit may be determined by providing a valve to deliver a metered amount.
  • gelatin for use in an inhaler or insufflator may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.
  • the compounds may be formulated for parenteral administration by injection, e.g. bolus injection or continuous infusion.
  • Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative.
  • the compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • compositions for parenteral administration include aqueous solutions of the active compounds in water-soluble form. Additionally, suspensions of the active compounds may be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes.
  • Aqueous injection suspensions may contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran.
  • the suspension may also contain suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.
  • the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
  • a suitable vehicle e.g., sterile pyrogen-free water
  • the compounds may also be formulated in rectal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter or other glycerides.
  • the compounds may also be formulated as a depot preparation. Such long acting formulations may be administered by implantation (for example subcutaneously or intramuscularly or by intramuscular injection).
  • the compounds may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
  • An example of a pharmaceutical carrier for the hydrophobic compounds of the invention is a cosolvent system comprising benzyl alcohol, a nonpolar surfactant, a water-miscible organic polymer, and an aqueous phase.
  • the cosolvent system may be the VPD co-solvent system.
  • VPD is a solution of 3% w/v benzyl alcohol, 8% w/v of the nonpolar surfactant polysorbate 80, and 65% w/v polyethylene glycol 300, made up to volume in absolute ethanol.
  • the VPD co-solvent system (VPD:5W) consists of VPD diluted 1 :1 with a 5% dextrose in water solution.
  • This co-solvent system dissolves hydrophobic compounds well, and itself produces low toxicity upon systemic administration.
  • the proportions of a co-solvent system may be varied considerably without destroying its solubility and toxicity characteristics.
  • identity of the co-solvent components may be varied: for example, other low-toxicity nonpolar surfactants may be used instead of polysorbate 80; the fraction size of polyethylene glycol may be varied; other biocompatible polymers may replace polyethylene glycol, e.g. polyvinyl pyrrolidone; and other sugars or polysaccharides may substitute for dextrose.
  • other delivery systems for hydrophobic pharmaceutical compounds may be employed. Liposomes and emulsions are well known examples of delivery vehicles or carriers for hydrophobic drugs.
  • Certain organic solvents such as dimethysulfoxide also may be employed, although usually at the cost of greater toxicity.
  • the compounds may be delivered using a sustained-release system, such as semipermeable matrices of solid hydrophobic polymers containing the therapeutic agent.
  • sustained-release materials have been established and are well known by those skilled in the art.
  • Sustained-release capsules may, depending on their chemical nature, release the compounds for a few weeks up to over 100 days.
  • additional strategies for protein stabilization may be employed.
  • the pharmaceutical compositions also may comprise suitable solid or gel phase carriers or excipients. Examples of such carriers or excipients include but are not limited to calcium carbonate, calcium phosphate, various sugars, starches, cellulose derivatives, gelatin, and polymers such as polyethylene glycols.
  • organic molecule compounds of the invention may be provided as salts with pharmaceutically compatible counterions.
  • Pharmaceutically compatible salts may be formed with many acids, including but not limited to hydrochloric, sulfuric, acetic, lactic, tartaric, malic, succinic, etc. Salts tend to be more soluble in aqueous or other protonic solvents than are the corresponding free base forms.
  • compositions suitable for use in the present invention include compositions wherein the active ingredients are contained in an effective amount to achieve its intended pu ⁇ ose. More specifically, a therapeutically effective amount means an amount effective to prevent development of or to alleviate the existing symptoms of the subject being treated. Determination of the effective amounts is well within the capability of those skilled in the art.
  • the therapeutically effective dose can be estimated initially from cellular assays.
  • a dose can be formulated in cellular and animal models to achieve a circulating concentration range that includes the IC 50 as determined in cellular assays (i.e., the concentration of the test compound which achieves a half-maximal inhibition of a given protein kinase activity).
  • the IC 50 as determined in cellular assays (i.e., the concentration of the test compound which achieves a half-maximal inhibition of a given protein kinase activity).
  • Such information can be used to more accurately determine useful doses in humans.
  • the most preferred compounds for systemic administration effectively inhibit protein kinase signaling in intact cells at levels that are safely achievable in plasma.
  • a therapeutically effective dose refers to that amount of the compound that results in amelioration of symptoms in a patient.
  • Toxicity and therapeutic efficacy of such compounds can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the maximum tolerated dose (MTD) and the ED 50 (effective dose for 50% maximal response).
  • the dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio between MTD and ED 50 .
  • Compounds which exhibit high therapeutic indices are preferred.
  • the data obtained from these cell culture assays and animal studies can be used in formulating a range of dosage for use in humans.
  • the dosage of such compounds lies preferably within a range of circulating concentrations that include the ED 50 with little or no toxicity.
  • the dosage may vary within this range depending upon the dosage form employed and the route of administration utilized.
  • the exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient's condition. (See e.g. Fingl et al, 1975, in "The Pharmacological Basis of Therapeutics", Ch. 1 pi).
  • the administration of an acute bolus or an infusion approaching the MTD may be required to obtain a rapid response.
  • Dosage amount and interval may be adjusted individually to provide plasma levels of the active moiety which are sufficient to maintain the kinase modulating effects, or minimal effective concentration (MEC).
  • MEC minimal effective concentration
  • the MEC will vary for each compound but can be estimated from in vitro data; e.g. the concentration necessary to achieve 50-90%) inhibition of protein kinase using the assays described herein. Dosages necessary to achieve the MEC will depend on individual characteristics and route of administration. However, HPLC assays or bioassays can be used to determine plasma concentrations.
  • Dosage intervals can also be determined using the MEC value.
  • Compounds should be administered using a regimen which maintains plasma levels above the MEC for 10-90%) of the time, preferably between 30-90%> and most preferably between 50-90%) until the desired amelioration of symptoms is achieved.
  • the effective local concentration of the drug may not be related to plasma concentration.
  • compositions administered will, of course, be dependent on the subject being treated, on the subject's weight, the severity of the affliction, the manner of administration and the judgment of the prescribing physician.
  • compositions may, if desired, be presented in a pack or dispenser device which may contain one or more unit dosage forms containing the active ingredient.
  • the pack may for example comprise metal or plastic foil, such as a blister pack.
  • the pack or dispenser device may be accompanied by instructions for administration.
  • Compositions comprising a compound of the invention formulated in a compatible pharmaceutical carrier may also be prepared, placed in an appropriate container, and labeled for treatment of an indicated condition.
  • the compounds of the present invention in the form of particles of very small size, for example as obtained by fluid energy milling.
  • the active compound may, if desired, be associated with other compatible pharmacologically active ingredients.
  • the compounds of this invention can be administered in combination with one or more additional pharmaceutical agents that inhibit or prevent the production of VEGF, attenuate intracellular responses to VEGF, block intracellular signal transduction, inhibit vascular hype ⁇ ermeability, reduce inflammation, or inhibit or prevent the formation of edema or neovascularization.
  • the compounds of the invention can be administered prior to, subsequent to or simultaneously with the additional pharmaceutical agent, whichever course of administration is appropriate.
  • the additional pharmaceutical agents include but are not limited to anti-edemic steroids, NSAIDS, ras inhibitors, anti-TNF agents, anti-ILl agents, antihistamines, PAF-antagonists, COX-1 inhibitors, COX-2 inhibitors, NO synthase inhibitors, PKC inhibitors and PI3 kinase inhibitors.
  • the compounds of the invention and the additional pharmaceutical agents act either additively or synergistically.
  • the administration of such a combination of substances that inhibit angiogenesis, vascular hype ⁇ ermeability and/or inhibit the formation of edema can provide greater relief from the deleterious effects of a hype ⁇ roliferative disorder, angiogenesis, vascular hype ⁇ ermeability or edema than the administration of either substance alone.
  • combinations with antiproliferative or cytotoxic chemotherapies or radiation are anticipated.
  • the present invention also comprises the use of a compound of formula I as a medicament.
  • the Src family currently includes Fyn, Lck, Fgr, Fes, Lyn, Src, Yes, Hck, and Blk.
  • the Syk family is currently understood to include only Zap and Syk.
  • the Janus family of kinases is involved in the transduction of growth factor and pro inflammatory cytokine signals through a number of receptors.
  • BTK and ITK members of the Tec family of kinases, play a less well understood role in immunobiology, their modulation by an inhibitor may prove therapeutically beneficial.
  • the kinases RIP, IRAK- 1 , IRAK-2, NIK, IKK- 1 and IKK-2 are involved in the signal transduction pathways for the key pro-inflammatory cytokines TNF and IL-1.
  • compounds of formula I may function as immunomodulatory agents useful for the maintenance of allografts and the treatment of autoimmune disorders. Through their ability to regulate T cell activation or the potentiation of an inflammatory process, these compounds could be used to treat such autoimmune diseases.
  • Transplants due to rejection phenomena are limited by the toxicity of currently available immunosuppressive agents and would benefit from an efficacious drug with improved therapeutic index.
  • Gene targeting experiments have demonstrated the essential role of Src in the biology of osteoclasts, the cells responsible for bone reso ⁇ tion.
  • Compounds of formula I may also be useful in the treatment of osteoporosis, osteopetrosis, Paget's disease, tumor-induced hypercalcemia and in the treatment of bone metastases.
  • a number of protein kinases have been demonstrated to be protooncogenes.
  • Chromosome breakage at the ltk kinase break point on chromosome 5
  • translocation as in the case of the Abl gene with BCR (Philadelphia chromosome)
  • truncation in instances such as c-Kit or EGFR or mutation (e.g., Met) result in the creation of dysregulated proteins converting them from protooncogene to oncogene products.
  • oncogenesis is driven by an autocrine or paracrine ligand/growth factor receptor interactions.
  • Members of the src-family kinases are typically involved in downstream signal transduction thereby potentiating the oncogenesis and they may become oncogenic by over-expression or mutation.
  • Vascular restenosis may involve process of FGF and/or PDGF - promoted smooth muscle and endothelial cell proliferation. Inhibition of FGFr or PDGFr kinase activity may be an efficacious strategy for inhibiting this phenomenon.
  • compounds of formula I which inhibit the kinase activity of normal or aberrant c-kit, c-met, c-fms, src-family members, EGFr, erbB2, erbB4, BCR-Abl, PDGFr, FGFr, and other receptor or cytosolic tyrosine kinases may be of value in the treatment of benign and neoplastic proliferative diseases.
  • pathological conditions for example, solid primary tumors and metastases, Kaposi's sarcoma, rheumatoid arthritis, blindness due to inappropriate ocular neovascularization, psoriasis and atherosclerosis
  • Polypeptide growth factors often produced by the disease tissue or associated inflammatory cells, and their corresponding endothelial cell specific receptor tyrosine kinases (e.g., KDR/NEGFR-2, Flt-
  • VEGF- stimulation of a VEGFR kinase is also believed to play an important role in the formation of tumor ascites, cerebral and pulmonary edema, pleural and pericardial effusions, delayed-type hypersensitivity reactions, tissue edema and organ dysfunction following trauma, burns, ischemia, diabetic complications, endometriosis, adult respiratory distress syndrome (ARDS), post-cardiopulmonary bypass-related hypotension and hype ⁇ ermeability, and ocular edema leading to glaucoma or blindness due to inappropriate neovascularization.
  • ARDS adult respiratory distress syndrome
  • Tie-2 is expressed also in a select population of hematopoietic stem cells in which it may play a role in their recruitment, adhesion, regulation and differentiation (Blood 89, 4317-4326 (1997)); this Tie-2 expressing population may serve as circulating angiogenic endothelial progenitors. Certain agents according to formula I capable of blocking the kinase activity of endothelial cell specific kinases could therefore inhibit disease progression involving these situations.
  • the compounds of formula I or a salt thereof or pharmaceutical compositions containing a therapeutically effective amount thereof may be used in the treatment of benign and neoplastic proliferative diseases and disorders of the immune system.
  • diseases include autoimmune diseases, such as rheumatoid arthritis, thyroiditis, type 1 diabetes, multiple sclerosis, sarcoidosis, inflammatory bowel disease, myasthenia gravis and systemic lupus erythematosus; psoriasis, organ transplant rejection (eg.
  • kidney rejection graft versus host disease
  • benign and neoplastic proliferative diseases human cancers such as lung, breast, stomach, bladder, colon, pancreas, ovarian, prostate and rectal cancer and hematopoietic malignancies (leukemia and lymphoma), and diseases involving inappropriate vascularization for example diabetic retinopathy, retinopathy of prematurity, choroidal neovascularization due to age-related macular degeneration, and infantile hemangiomas in human beings.
  • inhibitors may be useful in the treatment of disorders involving VEGF mediated edema, ascites, effusions, and exudates, including for example macular edema, cerebral edema, and adult respiratory distress syndrome (ARDS).
  • VEGF mediated edema ascites, effusions, and exudates
  • ARDS adult respiratory distress syndrome
  • the compounds of the present invention may also be useful in the prophylaxis of the above diseases.
  • a further aspect of the present invention provides the use of a compound of formula I or a salt thereof in the manufacture of a medicament for treating vascular hype ⁇ ermeability, angiogenesis-dependent disorders, proliferative diseases and/or disorders of the immune system in mammals, particularly human beings.
  • the present invention also provides a method of treating vascular hype ⁇ ermeability, inappropriate neovascularization, proliferative diseases and/or disorders of the immune system which comprises the administration of a therapeutically effective amount of a compound of formula I to a mammal, particularly a human being, in need thereof.
  • the in vitro potency of compounds in inhibiting these protein kinases may be determined by the procedures detailed below.
  • the potency of compounds can be determined by the amount of inhibition of the phosphorylation of an exogenous substrate (e.g., synthetic peptide (Z. Songyang et al, Nature. 373:536-539) by a test compound relative to control.
  • an exogenous substrate e.g., synthetic peptide (Z. Songyang et al, Nature. 373:536-539)
  • the coding sequence for the human KDR intra-cellular domain was generated through PCR using cDNAs isolated from HUVEC cells. A poly-His6 sequence was introduced at the N-terminus of this protein as well. This fragment was cloned into transfection vector pVL1393 at the Xba 1 and Not 1 site. Recombinant baculovirus (BV) was generated through co-transfection using the BaculoGold Transfection reagent (PharMingen). Recombinant BV was plaque purified and verified through Western analysis. For protein production, SF-9 cells were grown in SF-900-II medium at 2 x 106/ml, and were infected at 0.5 plaque forming units per cell (MOI). Cells were harvested at 48 hours post infection.
  • MOI plaque forming units per cell
  • SF-9 cells expressing (His) 6 KDR(aa789-1354) were lysed by adding 50 ml of Triton X-100 lysis buffer (20 M Tris, pH 8.0, 137 mM NaCl, 10% glycerol, 1% Triton X-100, lmM PMSF, lO ⁇ g/ml aprotinin, 1 ⁇ g/ml leupeptin) to the cell pellet from IL of cell culture.
  • the lysate was centrifuged at 19,000 ⁇ m in a Sorval SS-34 rotor for 30 min at 4°C.
  • the cell lysate was applied to a 5 ml NiCl 2 chelating sepharose column, equilibrated with 50 mM HEPES, pH7.5, 0.3 M NaCl.
  • KDR was eluted using the same buffer containing 0.25 M imidazole.
  • Column fractions were analyzed using SDS-PAGE and an ELISA assay (below) which measures kinase activity.
  • the purified KDR was exchanged into 25mM HEPES, pH7.5, 25mM NaCl, 5 mM DTT buffer and stored at -80°C.
  • the coding sequence for the human Tie-2 intra-cellular domain was generated through PCR using cDNAs isolated from human placenta as a template. A poly-His 6 sequence was introduced at the N-terminus and this construct was cloned into transfection vector pVL 1939 at the Xba 1 and Not 1 site. Recombinant BV was generated through co-transfection using the BaculoGold Transfection reagent (PharMingen). Recombinant BV was plaque purified and verified through Western analysis. For protein production, SF-9 insect cells were grown in SF-900-II medium at 2 x lOVml, and were infected at MOI of 0.5. Purification of the His-tagged kinase used in screening was analogous to that described for KDR.
  • the baculoviral expression vector pVL1393 (Phar Mingen, Los Angeles, CA) was used. A nucleotide sequence encoding poly-His6 was placed 5' to the nucleotide region encoding the entire intracellular kinase domain of human Flt-1 (amino acids 786-1338). The nucleotide sequence encoding the kinase domain was generated through PCR using cDNA libraries isolated from HUVEC cells. The histidine residues enabled affinity purification of the protein in a manner analogous to that for KDR and ZAP70. SF-9 insect cells were infected at a 0.5 multiplicity and harvested 48 hours post infection.
  • EGFR was purchased from Sigma (Cat # E-3641 ; 500 units/50 ⁇ l) and the EGF ligand was acquired from Oncogene Research Products/Calbiochem (Cat # PF011-100).
  • the baculoviral expression vector used was pVL1393 (Pharmingen, Los Angeles, Ca.).
  • the nucleotide sequence encoding amino acids M(H)6 LVPRjS was placed 5' to the region encoding the entirety of ZAP70 (amino acids 1-619).
  • the nucleotide sequence encoding the ZAP70 coding region was generated through PCR using cDNA libraries isolated from Jurkat immortalized T-cells. The histidine residues enabled affinity purification of the protein (vide infra).
  • the LVPRgS bridge constitutes a recognition sequence for proteolytic cleavage by thrombin, enabling removal of the affinity tag from the enzyme.
  • SF-9 insect cells were infected at a multiplicity of infection of 0.5 and harvested 48 hours post infection. Extraction and purification of ZAP70
  • SF-9 cells were lysed in a buffer consisting of 20 mM Tris, pH 8.0, 137 mM NaCl, 10% glycerol, 1% Triton X-100, 1 mM PMSF, 1 ⁇ g/ml leupeptin, 10 ⁇ g/ml aprotinin and 1 mM sodium orthovanadate.
  • the soluble lysate was applied to a chelating sepharose HiTrap column (Pharmacia) equilibrated in 50 mM HEPES, pH 7.5, 0.3 M NaCl. Fusion protein was eluted with 250 mM imidazole.
  • the enzyme was stored in buffer containing 50 mM HEPES, pH 7.5, 50 mM NaCl and 5 mM DTT.
  • Lck or truncated forms of Lck may be commercially obtained (e.g. from Upstate Biotechnology Inc. (Saranac Lake, N.Y) and Santa Cruz Biotechnology Inc. (Santa Cruz, Ca.)) or purified from known natural or recombinant sources using conventional methods.
  • Enzyme linked immunosorbent assays were used to detect and measure the presence of tyrosine kinase activity.
  • the ELISA were conducted according to known protocols which are described in, for example, Voller, et al, 1980, "Enzyme-Linked Immunosorbent Assay," In: Manual of Clinical Immunology, 2d ed., edited by Rose and Friedman, pp 359-371 Am. Soc. of Microbiology, Washington, D.C.
  • the disclosed protocol was adapted for determining activity with respect to a specific PTK.
  • preferred protocols for conducting the ELISA experiments is provided below. Adaptation of these protocols for determining a compound's activity for other members of the receptor PTK family, as well as non- receptor tyrosine kinases, are well within the abilities of those in the art.
  • a universal PTK substrate e.g., random copolymer of poly(Glu 4 Tyr), 20,000-50,000 MW
  • ATP typically 5 ⁇ M
  • PBS phosphate buffered saline
  • Reaction Buffer 1 OOmM Hepes, 20mM MgCl 2 , 4mM MnCl 2 , 5mM DTT,
  • ATP Store aliquots of lOOmM at -20°C. Dilute to 20 ⁇ M in water
  • Washing Buffer PBS with 0.1% Tween 20
  • Buffer 0.1 % bovine serum albumin (BSA) in PBS TMB Substrate: mix TMB substrate and peroxide solutions 9: 1 just before use or use
  • -Prepare inhibitor solutions at a 4x concentration in 20%) DMSO in water DMSO in water.
  • -Prepare reaction buffer -Prepare enzyme solution so that desired units are in 50 ⁇ l, e.g. for KDR make to 1 ng/ ⁇ l for a total of 50ng per well in the reactions.
  • Store on ice. -Make 4x ATP solution to 20 ⁇ M from lOOmM stock in water.
  • the Reaction Buffer utilized was 100 mM MOPSO, pH 6.5, 4 mM MnCl 2 , 20 mM MgCl 2 , 5 mM DTT, 0.2% BSA, 200 mM NaVO 4 under the analogous assay conditions.
  • Compounds of formula I may have therapeutic utility in the treatment of diseases involving both identified, including those not mentioned herein, and as yet unidentified protein tyrosine kinases which are inhibited by compounds of formula I. All compounds exemplified herein significantly inhibit KDR kinase at concentrations of 50 micromolar or below. Some compounds of this invention also significantly inhibit other PTKs such as lck at concentrations of 50 micromolar or below.
  • the human recombinant enzyme and assay buffer may be obtained commercially (New England Biolabs, Beverly, MA. USA) or purified from known natural or recombinant sources using conventional methods.
  • Cdc2 Assay The protocol used was that provided with the purchased reagents with minor modifications.
  • the reaction was carried out in a buffer consisting of 50mM Tris pH 7.5, lOOmM NaCl, ImM EGTA, 2mM DTT, 0.01% Brij, 5% DMSO and lOmM MgCl 2 (commercial buffer) supplemented with fresh 300 ⁇ M ATP (31 ⁇ Ci/ml) and 30 ⁇ g/ml histone type IIIss final concentrations.
  • a reaction volume of 80 ⁇ L, containing units of enzyme, was run for 20 minutes at 25 degrees C in the presence or absence of inhibitor. The reaction was terminated by the addition of 120 ⁇ L of 10%) acetic acid.
  • the substrate was separated from uninco ⁇ orated label by spotting the mixture on phosphocellulose paper, followed by 3 washes of 5 minutes each with 75mM phosphoric acid. Counts were measured by a betacounter in the presence of liquid scintillant.
  • Certain compounds of this invention significantly inhibit cdc2 at concentrations below 50 uM.
  • the catalytic subunit of PKC may be obtained commercially (Calbiochem).
  • a radioactive kinase assay was employed following a published procedure (Yasuda, I., Kirshimoto, A., Tanaka, S., Tominaga, M., Sakurai, A., Nishizuka, Y. Biochemical and Biophysical Research Communication 3:166, 1220-1227 (1990)). Briefly, all reactions were performed in a kinase buffer consisting of 50 mM Tris- HC1 pH7.5, lOmM MgCl 2 , 2mM DTT, ImM EGTA, 100 ⁇ M ATP, 8 ⁇ M peptide, 5% DMSO and 33 P ATP (8Ci/mM).
  • the recombinant murine enzyme and assay buffer may be obtained commercially (New England Biolabs, Beverly MA. USA) or purified from known natural or recombinant sources using conventional methods.
  • reaction was carried out in a buffer consisting of 50 mM Tris pH 7.5, ImM EGTA, 2mM DTT, 0.01% Brij, 5% DMSO and 10 mM MgCl 2 (commercial buffer) supplemented with fresh 100 ⁇ M ATP (31 ⁇ Ci/ml) and 30 ⁇ M myelin basic protein under conditions recommended by the supplier. Reaction volumes and method of assaying inco ⁇ orated radioactivity were as described for the PKC assay (vide supra).
  • T-cells Upon activation by mitogen or antigen, T-cells are induced to secrete IL-2, a growth factor that supports their subsequent proliferative phase. Therefore, one may measure either production of IL-2 from or cell proliferation of, primary T-cells or appropriate T-cell lines as a surrogate for T-cell activation. Both of these assays are well described in the literature and their parameters well documented (in Current Protocols in Immunology, Vol 2, 7.10.1-7.11.2). In brief, T-cells may be activated by co-culture with allogenic stimulator cells, a process termed the one-way mixed lymphophocyte reaction. Responder and stimulator peripheral blood mononuclear cells are purified by Ficoll-Hypaque gradient (Pharmacia) per directions of the manufacturer.
  • Stimulator cells are mitotically inactivated by treatment with mitomycin C (Sigma) or gamma irradiation. Responder and stimulator cells are co-cultured at a ratio of two to one in the presence or absence of the test compound. Typically 10 5 responders are mixed with 5 x 10" stimulators and plated (200 ⁇ l volume) in a U bottom microtiter plate (Costar Scientific).
  • the cells are cultured in RPMI 1640 supplemented with either heat inactivated fetal bovine serum (Hyclone Laboratories) or pooled human AB serum from male donors, 5 x 10 "5 M 2mercaptoethanol and 0.5% DMSO,
  • the cultures are pulsed with 0.5 ⁇ Ci of 3 H thymidine (Amersham) one day prior to harvest (typically day three).
  • the cultures are harvested (Betaplate harvester, Wallac) and isotope uptake assessed by liquid scintillation (Betaplate, Wallac).
  • the same culture system may be used for assessing T-cell activation by measurement of IL-2 production. Eighteen to twenty- four hours after culture initiation, the supernatants are removed and the IL-2 concentration is measured by ELISA (R and D Systems) following the directions of the manufacturer.
  • T-cells can be activated in vivo by ligation of the constant portion of the T-cell receptor with a monoclonal anti-CD3 antibody (Ab).
  • Ab monoclonal anti-CD3 antibody
  • BALB/c mice are given lO ⁇ g of anti-CD 3 Ab intraperitoneally two hours prior to exsanguination. Animals to receive a test drug are pre-treated with a single dose of the compound one hour prior to anti-CD3 Ab administration.
  • Serum levels of the proinflammatory cytokines interferon- ⁇ (IFN- ⁇ ) and tumor necrosis factor- ⁇ (TNF- ⁇ ), indicators of T-cell activation are measured by ELISA.
  • a similar model employs in vivo T-cell priming with a specific antigen such as keyhole limpet hemocyanin (KLH) followed by a secondary in vitro challenge of draining lymph node cells with the same antigen.
  • KLH keyhole limpet hemocyanin
  • measurement of cytokine production is used to assess the activation state of the cultured cells. Briefly, C57BL/6 mice are immunized subcutaneously with 100 ⁇ g KLH emulsified in complete Freund's adjuvant (CFA) on day zero.
  • CFA complete Freund's adjuvant
  • Interleukin-2 IL-2
  • IFN- ⁇ Interleukin-2
  • ELISA Interleukin-2
  • Lead compounds can also be tested in animal models of human disease.
  • EAE auto-immune encephalomyelitis
  • CIA collagen-induced arthritis
  • mice or rats are immunized with an emulsion of myelin basic protein (MBP), or neurogenic peptide derivatives thereof, and CFA.
  • MBP myelin basic protein
  • Acute disease can be induced with the addition of bacterial toxins such as bordetella pertussis. Relapsing/remitting disease is induced by adoptive transfer of T-cells from MBP/ peptide immunized animals.
  • CIA may be induced in DBA 1 mice by immunization with type II collagen (J.
  • mice will develop signs of arthritis as early as ten days following antigen challenge and may be scored for as long as ninety days after immunization.
  • a compound may be administered either prophylactically or at the time of disease onset. Efficacious drugs should reduce severity and/or incidence.
  • Certain compounds of this invention which inhibit one or more angiogenic receptor PTK, and or a protein kinase such as lck involved in mediating inflammatory responses can reduce the severity and incidence of arthritis in these models.
  • HUVEC cells from pooled donors were purchased from Clonetics (San Diego, CA) and cultured according to the manufacturer directions. Only early passages (3-8) were used for this assay. Cells were cultured in 100 mm dishes (Falcon for tissue culture; Becton Dickinson; Madison, England) using complete EBM media (Clonetics).
  • Equal amounts of proteins were then precipitated by addition of cold (-20°C) ethanol (2 volumes) for a minimum of 1 hour or a maximum of overnight.
  • Pellets were reconstituted in Laemli sample buffer containing 5%> ⁇ -mercaptoethanol (BioRad; Hercules, CA) and boiled for 5min.
  • the proteins were resolved by polyacrylamide gel electrophoresis (6%>, 1.5mm Novex, San Deigo, CA) and transferred onto a nitrocellulose membrane using the Novex system.
  • This assay measures the capacity of compounds to inhibit the acute increase in uterine weight in mice which occurs in the first few hours following estrogen stimulation.
  • This early onset of uterine weight increase is known to be due to edema caused by increased permeability of uterine vasculature.
  • Cullinan-Bove and Koss demonstrated a close temporal relationship of estrogen-stimulated uterine edema with increased expression of VEGF mRNA in the uterus.
  • DMSO dimethyl methacrylate
  • Cremaphor EL Vehicle components
  • mice (Balb/c, 8-12 weeks old) were purchased from Taconic (Germantown, NY) and housed in a pathogen-free animal facility in accordance with institutional Animal Care and Use Committee Guidelines.
  • mice were given an intraperitoneal (i.p.) injection of 12.5 units of pregnant mare's serum gonadotropin (PMSG).
  • PMSG pregnant mare's serum gonadotropin
  • mice were randomized and divided into groups of 5-10. Test compounds were administered by i.p., i.v. or p.o. routes depending on solubility and vehicle at doses ranging from 1-100 mg/kg. Vehicle control group received vehicle only and two groups were left untreated.
  • results demonstrate that certain compounds of the present invention inhibit the formation of edema when administered systemically by various routes.
  • Certain compounds of this invention which are inhibitors of angiogenic receptor tyrosine kinases can also be shown active in a Matrigel implant model of neovascularization.
  • the Matrigel neovascularization model involves the formation of new blood vessels within a clear "marble" of extracellular matrix implanted subcutaneously which is induced by the presence of proangiogenic factor producing tumor cells (for examples see: Passaniti, A., et al, Lab. Investig. (1992), 67(4), 519- 528; Anat. Rec. (1997), 249(1), 63-73; Int. J.
  • the model preferably runs over 3-4days and endpoints include macroscopic visual/image scoring of neovascularization, microscopic microvessel density determinations, and hemoglobin quantitation (Drabkin method) following removal of the implant versus controls from animals untreated with inhibitors.
  • the model may alternatively employ bFGF or HGF as the stimulus.
  • Certain compounds of this invention which inhibit one or more oncogenic, protooncogenic, or proliferation-dependent protein kinases, or angiogenic receptor PTK also inhibit the growth of primary murine, rat or human xenograft tumors in mice, or inhibit metastasis in murine models.
  • a second method of preparing the ring systems of the compounds of formula I in which R ! is hydrogen is set forth by U.S. Patent 3,843,666 where compounds with the general formula VI are heated to 75° to 175°C with a catalytic amount of an organic carboxylic acid or an organic sulfonic acid in an inert solvent such as an aromatic hydrocarbon for a period of 6 to 24 hours,
  • R 2 , R 3 , R 4 , R 5 , R 6 and X are as previously defined.
  • Compounds of formula VI are prepared by treating compounds of the general formula VII with hydrazine in an inert solvent. The reaction is carried out at 15° to 20°C for a period of up to 24 hours.
  • compounds of formula I may be prepared directly by reacting a compound of formula VII with hydrazine without isolating the compound of formula VI, for example by heating a compound of formula VII with hydrazine in an inert solvent, e.g. methanol, in the presence of an acid catalyst, e.g. acetic acid, at a temperature in the range from 60°C to the boiling point of the inert solvent employed.
  • an inert solvent e.g. methanol
  • an acid catalyst e.g. acetic acid
  • Rl, R 2 , R 3 , R 4 , R 5 , R 6 and X are as previously defined with hydrazine in an inert solvent e.g. methanol, at a temperature in the range of from 15°C to the boiling point of the inert solvent employed.
  • an inert solvent e.g. methanol
  • Y is any conventional leaving group, such as chlorine, bromine, iodine, tosylate or mesylate and R 3 , R 4 , R 5 , R 6 and X are as previously defined .
  • Compounds of formula VII may also be prepared by reacting a compound of formula II with an epoxidizing agent, for example hydrogen peroxide, in an inert solvent, for example methanol, dichloromethane, water or mixtures thereof, at a temperature in the range of 0° to 100°C optionally in the presence of a base, for example sodium hydroxide.
  • an epoxidizing agent for example hydrogen peroxide
  • an inert solvent for example methanol, dichloromethane, water or mixtures thereof
  • Cyclization of VI can also be effected by treatment with a mineral acid such as hydrochloric acid, sulfuric acid or phosphoric acid.
  • the reaction is carried out in a lower alkanol at a temperature between 15°C to 20°C for a period of 12 to 48 hours.
  • the product of the reaction, IX can then be aromatized to I by heating to a temperature of 50°C to 150°C with an organic carboxylic acid or an organic sulfonic acid in a straight chain ether or cyclic ether for the period of 8 to 30 hours.
  • Compound IX can be diacetylated by treatment with an acid anhydride of formula (R x CO)2 ⁇ (X) in which R x is a Ci-4 alkyl group in an inert solvent such as an aromatic hydrocarbon at a temperature between 35°C to 200°C for a period of 5 to 8 hours to give a compound of formula XI.
  • Compound XI can then be aromatized to compound XII by heating to a temperature of 35°C to 200°C with a mineral acid or an organic acid in an inert solvent for the period of 4 to 8 hours. Finally, compound XII can be converted to I by heating to a temperature of 50°C to 150°C in an inert solvent such as water or a lower alcohol in the presence of an alkali metal or an alkali metal hydroxide for the period of 8 to 30 hours.
  • an inert solvent such as water or a lower alcohol
  • Compounds of formula I may be prepared by reacting a compound of formula XIII with a dehydrogenating agent, for example sulphur, oxygen, palladium, manganese dioxide or lead dioxide optionally in the presence of an inert solvent, for example a hydrocarbon, at a temperature in the range of 15 to 250°C.
  • a dehydrogenating agent for example sulphur, oxygen, palladium, manganese dioxide or lead dioxide
  • an inert solvent for example a hydrocarbon
  • the bridging carbonyl can be transformed to a methylene group via a Wolf- Kishner reduction of the corresponding hydrazone (Mosher, W.A., Tawfik, E.-Z., Lipp, D. W. J. Org. Chem. 1971, 36, 3890).
  • alkoxy substituents may be reacted with a suitable ether cleaving reagent for example hydrobromic acid, boron tribromide or pyridine hydrochloride to give a compound of formula I with a hydroxy substituent.
  • a suitable ether cleaving reagent for example hydrobromic acid, boron tribromide or pyridine hydrochloride
  • compounds of formula I with an alkoxy substituent may be prepared by alkylating compounds of formula I which have a hydroxy substituent.
  • Carboxylic ester substituents may be converted into carboxy or amide substituents and carboxylic acid substituents may be converted into carboxylic ester or amide substituents.
  • Nitro substituents may be reduced to amines and amines may be acylated by methods known to those skilled in the art.
  • Example 1 a A mixture of indan-1-one (10.0 g), ethanol (35 ml), hydrazine hydrate (10.0 ml) and glacial acetic acid (2.0 ml) was boiled under reflux under nitrogen for 1 hour. The mixture was cooled to 20°C and the mixture concentrated under reduced pressure to give a solid which was collected by filtration to give indan-1-one hydrazone, m.p. 84-86°C.
  • the aqueous layer was neutralized with sodium bicarbonate and extracted with ether to give a brown oil.
  • the oil was purified by flash column chromatography on silica using ethyl acetate/petroleum ether (1:4) as the mobile phase to give 3-(3,4,5- trimethoxyphenyl)-l,4-dihydroindeno[l,2-c]-pyrazole, m.p.185-187°C.
  • indan-1-one hydrazone (3.55g) was dissolved in tetrahydrofuran (80ml) at 0°C under nitrogen with stirring, n- Butyllithium (28.8ml of a 2.5M solution in hexane) was added to the solution and the mixture was stirred at 0°C for 0.5h.
  • Ethyl 3-methoxybenzoate (2.16g) was added, followed by 3M hydrochloric acid (80ml) and the mixture was worked up as described in Example 1 to give 3-(3- methoxyphenyl)-l,4-dihydroindeno[l,2-cjpyrazole, m.p. 172-174°C.
  • Example 4 A solution of 3-chloroperoxybenzoic acid (450 mg, 60%> pure) in dichloromethane (30 ml) was added dropwise with stirring over 15 minutes to a solution of 3-phenyl-lH-[l]benzothieno[3,2-c] ⁇ yrazole (400 mg) in dichloromethane (50 ml) at 0-5°C. The mixture was stirred at 5-15°C for 4 hours, then washed with water, dried and evaporated to give a solid which was purified by chromatography on silica using petroleum ether/ethyl acetate (1:3) as the mobile phase.
  • Example 7 a A mixture of indan-1 -one (3.3 g), methyl 4-formylbenzoate (5.0 g), piperidine (0.6 ml) and glacial acetic acid (0.5 ml) was heated on a steam bath for 3 hours. The solid mass obtained was boiled up in industrial methylated spirits (200 ml) and then hot filtered. The solid residue obtained was washed with industrial methylated spirits and dried to give methyl 4-(l- oxoindan-2-ylidenemethyl)benzoate, m.p. 194-198°C.
  • the aqueous phase was acidified with 5M hydrochloric acid and extracted with dichloromethane to give 4-(l-oxospiro[indan-2,2'- oxiran]-3'-yl)benzoic acid, m.p. 220°C with decomposition
  • Example 9 A mixture of 4-(l,4-dihydroindeno[l,2-c]pyrazol-3-yl)benzoic acid (1.05 g) and dry tetrahydrofuran (30 ml) was stirred at ambient temperature whilst triethylamine (1.1 ml) was added. The mixture was stirred at ambient temperature for 0.5 h then cooled to 3°C and methyl chloroformate (1.2 ml) was added dropwise over 5 to 10 minutes at 3-9°C. The mixture was stirred for 1 hour at 3-9°C and then added to concentrated aqueous ammonia solution (70 ml, SG 0.880) with rapid stirring. The mixture was stirred for 2 hours at 5°C and allowed to warm up to ambient temperature.
  • the mixture was concentrated under reduced pressure and then treated with 1M sodium hydroxide solution (200 ml). The mixture was stirred for 0.5 hour and then filtered. The solid obtained was stirred with 2M sodium hydroxide solution (50 ml) for 6 hours and then allowed to stand at ambient temperature for 18 hours. The mixture was filtered to give a solid which was washed with water and dried. This solid was purified by flash column chromatography on silica using dichloromethane/industrial methylated spirit/triethylamine (25:2.5:1.5) as the mobile phase.
  • Example 12 a A mixture of indan-1-one (20.0 g), 4-nitrobenzaldehyde (27.0 g), glacial acetic acid (3.0 g) and piperidine (3.06 g) was heated at 95°C under nitrogen for 3.5 h. The mixture was cooled to 20°C and filtered to give a solid which was recrystallized from industrial methylated spirit to give 2-(4- nitrobenzylidene)indan-l -one.
  • Example 12 The product from Example 12 (3.0 g) was suspended in industrial methylated spirit (200 ml) and 5% palladium on charcoal (250 mg) was added followed by ammonium formate (2.05 g). The mixture was stirred and heated at 70°C for 3 hours and then cooled to ambient temperature and then filtered. The filtrate was concentrated under reduced pressure and triturated with dichloromethane to give 4- (l,4-dihydroindeno[l,2-c]pyrazol-3-yl)aniline, m.p. 253-254°C.
  • Example 14 3-(4-Pyridyl)-4,5-dihydro-lH-benzo[g]indazole (100 mg, compound commercially available from Aldrich) was dissolved in tetrahydrofuran (5-10 ml) with warming and then 3-chloroperoxybenzoic acid (1.25 molar equivalents of 70- 75% pure material) was added. The mixture was heated at 55-60°C for 4 hours. A precipitate was collected by filtration, washed with tetrahydrofuran and then ether to give 4-(4,5-dihydro-lH-benzo[g]indazol-3-yl)pyridine 1 -oxide.
  • N-oxide product from Example 14 (25 mg) was suspended in dimethyl- formamide (200 ⁇ l) and to this suspension was added trimethylsilyl cyanide (5 molar equivalents) and triethylamine (3 molar equivalents). The mixture was heated at 110°C for 12 hours and was then diluted with dichloromethane and washed with saturated sodium bicarbonate solution. The organic layer was separated, dried, filtered and evaporated to give, after flash column chromatography using 20-40%> of acetonitrile in dichloromethane as the mobile phase, 4-(4,5-dihydro-lH- benzo[g]indazol-3-yl)-2-pyridinecarbonitrile.
  • Example 17 The product from Example 16 (50 mg) was suspended in a mixture of ethanol (1 ml) and water (0.5 ml) containing sodium carbonate (1 molar equivalent) and hydroxylamine hydrochloride (2 molar equivalents). The mixture was boiled under reflux for 4 hours, then cooled. A precipitate formed which was collected by filtration to give 4-(4,5-dihydro-lH-benzo[g]indazol-3-yl)-2-pyridinecarboxamide oxime.
  • Potassium carbonate (1 molar equivalent) was added to a mixture of the product from Example 16 (50 mg), DMSO (600 ⁇ l), followed by the addition of hydrogen peroxide (250 ⁇ l of a 30% solution) which was added dropwise. Within a few minutes of the completion of the addition a precipitate had formed. The mixture was stirred for 1 hour and then water was added. The precipitate was collected by filtration to give 4-(4,5-dihydro-lH-benzo[g]indazol-3-yl)-2-pyridine carboxamide.
  • Example 19 The product from Example 19 (100 mg) was suspended in propyl formate (10 ml) and ethanol (1.5 ml). This mixture was boiled under reflux and after 2 minutes triethylamine (0.5 ml) was added. The mixture was boiled under reflux for 4 hours and then cooled and diluted with dichloromethane (50 ml). The mixture was washed with water, then dried, filtered and evaporated to give a residue which was triturated with ether and dichloromethane to give /V- ⁇ [4-(4,5-dihydro-lH-benzo[g]- indazol-3-yl)-2-pyridyl]methyl ⁇ formamide.
  • Lithium diisopropylamide (27.2 ml of a 2.0M solution in heptane/THF/ ethylbenzene) was added dropwise to a stirred solution of 5-methoxyindan-l- one tert-butyloxycarbonylhydrazone (5.0 g) in tetrahydrofuran (150 ml) at -78°C under nitrogen with stirring. After the addition was complete the mixture was stirred for 1.5 hours at -78°C and then a solution of ethyl thiophen-2-carboxylate (3.39 g) in tetrahydrofuran (25 ml) was added dropwise.
  • Triethylamine (0.48 ml) was added to a solution of 4-(l,4- dihydroindeno[l,2-cjpyrazol-3-yl)aniline (400 mg) in dichloromethane (20 ml) with stirring under nitrogen at ambient temperature followed by benzoyl chloride (0.4 ml). The mixture was stirred for 3 hours and then allowed to stand for 18 hours. The mixture was filtered to give 4'-(l - benzoyl-l,4-dihydroindeno[l,2-c]pyrazol-3-yl)benzanilide, m.p. 241°C.
  • Example 35 In a similar manner to example 33, a mixture of 3-(l ,4-dihydroindeno-
  • 60-80°C (50ml) was added and then decanted off to leave a gum which was further treated with petroleum ether as above and the residual gum dissolved in dichloromethane and purified by flash column chromatography on silica using dichloromethane/iMS/triethylamine (25:2:1) as the mobile phase to give a solid which was dissolved in ethanol (70ml) and then dry hydrogen chloride gas was passed through the solution.
  • step 1 products were taken on on the basis of being the corresponding 2,3-epoxyketones.
  • step 1 To the products from step 1 in a septum sealed tube was added, via a Gilson 215 liquid handler, n-butanol (2ml) followed by hydrazine hydrate (2 molar equivalents based on amount of starting enone used in step 1) as a 10% solution by volume in n-butanol (see Table B). To each tube was finally added glacial acetic acid (2 drops) manually by syringe. The reactions were heated at 100°C with shaking for the times indicated in Table B, based on monitoring by t.l.c. analysis.
  • Example 46 3-(4-Bromo-2-thienyl)-4,5-dihydroindeno[l ,2-c]pyrazole
  • Example 47 3-(4-Benzyloxyphenyl)-4,5-dihydro-2H-benz[g]indazole
  • Example 51 The following examples were prepared in a similar manner to Example 51 by reacting indan-1-one with the appropriate aldehyde;
  • Example 56 A mixture of 4-(l ,4-dihydroindeno [1 ,2-c]pyrazol-3-yl)phenol (0.28 g,
  • Example 30 potassium carbonate (0.16 g) and 2-bromoacetamide (0.16 g) was stirred together at ambient temperature in dry dimethyl formamide (5 ml) for 4 days and then allowed to stand at ambient temperature for 9 days. The mixture was diluted with dichloromethane (50 ml) and washed with 1M aqueous sodium hydroxide solution and then with water. Some insoluble material which remained throughout the extractions was collected by filtration. The solid was purified by flash column chromatography on silica using ethyl acetate as the mobile phase. Appropriate fractions were collected, combined and evaporated to give 2-[4-(l,4- dihydroindeno[l,2-c]pyrazol-3-yl)phenoxy]acetamide, m.p. 247-248°C.
  • Example 58 a A mixture of 4-cyanophenacyl bromide (3.0 g), methyl thiosalicylate (2.25 g), sodium methoxide (1.52 g) and ethanol (100 ml) was boiled under reflux for 3.5 hours. The mixture was cooled and diluted with 2M hydrochloric acid (100 ml). A precipitate was collected by filtration and recrystallized from ethanol to give 2-(4-cyanobenzoyl)-3-hydroxybenzo[b]thiophene.
  • Example 60 In a similar manner to Example 58, 4-methoxyphenacyl bromide (0.81 g) was reacted with methylthiosahcylate (5.0 g) in sodium methoxide and methanol to give 2- (4-methoxybenzoyl)-3-hydroxy[b]thiophene which was reacted with hydrazine hydrate to give 3-(4-methoxyphenyl)-lH-benzothieno[3,2-cjpyrazole, m.p. 185-187°C.
  • Example 64 The compounds shown in Tables 1, 2 and 3 were prepared in a similar manner to Example 7b and 7c. Alternatively these compounds are prepared in a similar manner to Example 30.
  • Example 64 The compounds shown in Tables 1, 2 and 3 were prepared in a similar manner to Example 7b and 7c. Alternatively these compounds are prepared in a similar manner to Example 30.
  • Example 64
  • Example 7a In a similar manner to Example 7a, indan-1-one was reacted with 4- cyanobenzaldehyde and the intermediate product obtained was reacted with hydrogen peroxide in a similar manner to Example 7b to give an intermediate spiro compound which was reacted with hydrazine hydrate in a similar manner to Example 7c to give 4-(l,4-dihydroindeno[l,2-c]pyrazol-3-yl)benzonitrile, m.p. 245°C.
  • 6-Methoxy-2-(4-thiomethoxybenzylidene)-l-tetralone was reacted with hydrogen peroxide and then with hydrazine hydrate following the general procedure described for examples 41-50 to give 7-methoxy-3-(4-methylsulphonylphenyl)-4,5- dihydro-2H-benz[g]indazole (C 19 ⁇ 18 ⁇ 2 O 3 S MWt 354, M + found, percentage purity by HPLC 66%).
  • 3-Phenylindeno[l,2-c]pyrazol-4-(lH)-one (2.4g) was suspended in toluene (40 ml) under nitrogen and trimethyl aluminium (2.2 ml of a 2M solution in heptane) was added at 20°C with stirring. The mixture was heated at 90°C for 16 hours, then cooled to ambient temperature and poured on to crushed ice (approx. 600 ml) containing concentrated hydrochloric acid (30 ml).
  • Example 70 4- ⁇ H- ⁇ iBenzothieno 3.2-clpyrazol-3-yl)-N-r3-Cimidazol-l- vDpropyljbenzylamine trihydrochloride.
  • the reaction mixture was poured onto a solution of saturated sodium bicarbonate (20ml) and stirred for about 30 mins, the layers separated and the aqueous layer extracted with dichloromethane (2x50ml). The combined extracts and organic layer were washed with water (1x25ml), dried (MgSO 4 ), filtered and concentrated in vacuo to give a waxy solid which was taken up in ethanol.
  • the solid was purified by flash column chromatography on silica using ethyl acetate as eluant to give a solid which was washed with diethyl ether and dried to give 4-(l,4-dihydroindeno[l,2-cjpyrazol- 3-yl)benzamide oxime, m.p.> 300°C.
  • the oil was purified by flash column chromatography on silica using ethyl acetate/ ethanol/ triethylamine (7 : 2 : 1) as eluant to give a pale yellow gum.
  • the gum was dissolved in warm ethanol (5ml), hydrochloric acid (cone, 0.6ml) was added and the solvent then removed under reduced pressure.
  • the gummy solid residue was boiled up with ethanol (10ml) and cooled in ice.
  • N-(3-Phenyl-l,4-dihydroindeno[l,2-c]pyrazol-6-yl)-2-mo ⁇ holinoacetamide (0.6 g) was dissolved in tetrahydrofuran (40 ml) at ambient temperature under nitrogen and lithium aluminium hydride (0.24 g) added portionwise over about 10 minutes. The mixture was stirred at ambient temperature for about 24 hours. A further addition of lithium aluminium hydride (0.24 g) was added and the mixture was stirred at ambient temperature for about another 24 hours. It was then poured onto a saturated aqueous solution of sodium sulphate (30 ml) and extracted with diethyl ether (2 x 30 ml).
  • N-[2-(Diethylamino)ethyl]-N-[4-(l,4-dihydroindeno[l,2-c]pyrazol-3- yl)phenyljurea A suspension of of 3-(4-isocyanatophenyl)-l,4-dihydroindeno[l,2- cjpyrazole, 2 (75mg, 0.274 mmol), Et 3 ⁇ (0.191 mL, 1.37 mmol), N,N- diethylethylenediamine (0.112 mL, 1.37 mmol) in toluene (3 mL) was heated to about 95 °C for about 3 hours. The white solid was filtered off and washed with EtOAc and dried under vacuum to afford 61 mg (57%>) of pure product. LC/MS 390 (M+l); LC retention time 2.76 minutes.
  • Example 143 -[4-(l ,4-Dihydroindeno [ 1 ,2-c]pyrazol-3-yl)phenyl]-4-(4-fluorophenyl)-l - piperazinecarboxamide: 63 mg (63%) of pure product was isolated. LC/MS 454 (M+l); LC retention time 3.53 minutes.
  • condition A in EtOh, r.t. (for allphatoc primary amines) condition B: p-TS A (cat.) in toluene, reflux (for secondary or aromatic amines)
  • active compound denotes any compound of the invention but particularly any compound which is the final product of one of the preceding Examples.
  • Capsules In the preparation of capsules, 10 parts by weight of active compound and 240 parts by weight of lactose are de-aggregated and blended. The mixture is filled into hard gelatin capsules, each capsule containing a unit dose or part of a unit dose of active compound.
  • Tablets are prepared from the following ingredients.
  • the active compound, the lactose and some of the starch are de-aggregated, blended and the resulting mixture is granulated with a solution of the polyvinylpyrrolidone in ethanol.
  • the dry granulate is blended with the magnesium stearate and the rest of the starch.
  • the mixture is then compressed in a tabletting machine to give tablets each containing a unit dose or apart of a unit dose of active compound.
  • Tablets are prepared by the method described in (b) above.
  • the tablets are enteric coated in a conventional manner using a solution of 20%> cellulose acetate phthalate and 3% diethyl phthalate in ethanol: dichloromethane (1:1).
  • suppositories 100 parts by weight of active compound is inco ⁇ orated in 1300 parts by weight of triglyceride suppository base and the mixture formed into suppositories each containing a therapeutically effective amount of active ingredient.

Abstract

L'invention concerne certains 3-aryle 3-hétéroaryle pyrazoles avec un noyau de fusion 4,5(3,4)-bicyclique qui sont des inhibiteurs de l'activité de protéine kinase et dont certains sont des composés nouveaux; l'invention concerne aussi des compositions pharmaceutiques à base de ces pyrazoles et des procédés de fabrication correspondants.
EP99962700A 1998-11-06 1999-11-04 Derives de pyrazole tricyclique Withdrawn EP1127051A2 (fr)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US10746798P 1998-11-06 1998-11-06
US107467P 1998-11-06
PCT/US1999/026105 WO2000027822A2 (fr) 1998-11-06 1999-11-04 Derives de pyrazole tricyclique

Publications (1)

Publication Number Publication Date
EP1127051A2 true EP1127051A2 (fr) 2001-08-29

Family

ID=22316761

Family Applications (1)

Application Number Title Priority Date Filing Date
EP99962700A Withdrawn EP1127051A2 (fr) 1998-11-06 1999-11-04 Derives de pyrazole tricyclique

Country Status (19)

Country Link
EP (1) EP1127051A2 (fr)
JP (1) JP2003517447A (fr)
KR (1) KR20010086005A (fr)
CN (1) CN1335836A (fr)
AU (1) AU762992B2 (fr)
BG (1) BG105481A (fr)
BR (1) BR9915132A (fr)
CA (1) CA2350235A1 (fr)
CZ (1) CZ20011563A3 (fr)
HK (1) HK1042895A1 (fr)
HU (1) HUP0200310A3 (fr)
ID (1) ID30132A (fr)
IL (1) IL142584A0 (fr)
NO (1) NO20012219L (fr)
PL (1) PL348210A1 (fr)
SK (1) SK5282001A3 (fr)
TR (1) TR200102277T2 (fr)
WO (1) WO2000027822A2 (fr)
ZA (1) ZA200103610B (fr)

Families Citing this family (47)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6462036B1 (en) * 1998-11-06 2002-10-08 Basf Aktiengesellschaft Tricyclic pyrazole derivatives
BR0009322A (pt) * 1999-03-26 2002-04-30 Euro Celtique Sa Pirazóis, imidazóis, oxazóis, tiazóis e pirróis substituìdos com arila e o uso dos mesmos
US6297238B1 (en) 1999-04-06 2001-10-02 Basf Aktiengesellschaft Therapeutic agents
DK1165544T3 (da) * 1999-04-06 2003-12-01 Abbott Gmbh & Co Kg Substituerede 1,4-dihydroindeno[1,2-c]pyrazoler som inhibitorer af tyrosinkinase
US20040048844A1 (en) 1999-10-20 2004-03-11 Bristol-Myers Squibb Pharma Company Acylsemicarbazides as cyclin dependent kinase inhibitors useful as anti-cancer and anti-proliferative agents
US6291504B1 (en) 1999-10-20 2001-09-18 Dupont Pharmaceuticals Company Acylsemicarbazides and their uses
DE60125543T2 (de) 2000-04-05 2007-10-04 Zymogenetics, Inc., Seattle Löslicher zytokinrezeptor zalpha11
US6723730B2 (en) 2000-07-20 2004-04-20 Neurogen Corporation Capsaicin receptor ligands
ATE295354T1 (de) 2000-08-18 2005-05-15 Agouron Pharma Heterozyklische-hydroximino-fluorene und ihre verwendung zur inhibierung von proteinkinasen
US6849631B2 (en) 2000-12-08 2005-02-01 Bristol Myers Squibb Pharma Company Semicarbazides and their uses
WO2003024935A2 (fr) 2001-09-19 2003-03-27 Pharmacia Corporation Composes de pyrazolyle substitues destines au traitement de l'inflammation
US6849653B2 (en) 2001-09-19 2005-02-01 Pharmacia Corporation Substituted pyrazolyl benzenesulfamide compounds for the treatment of inflammation
MXPA04008680A (es) * 2002-02-19 2004-12-06 Pharmacia Italia Spa Derivados de pirazol triciclicos, procedimientos para su preparacion y su uso como agentes anti-tumorales.
CA2516254A1 (fr) * 2003-02-17 2004-08-26 Pharmacia Italia S.P.A. Derives de pyrazole tetracyclique utilises comme inhibiteurs de kinase, processus de preparation correspondant et compositions pharmaceutiques les comprenant
US7456169B2 (en) 2003-02-27 2008-11-25 Abbott Laboratories Inc. Heterocyclic kinase inhibitors
JP2006520400A (ja) * 2003-03-07 2006-09-07 アボット・ラボラトリーズ 縮合三および四環式ピラゾールキナーゼ阻害剤
US7320986B2 (en) 2003-03-07 2008-01-22 Abbott Labortories Fused tri and tetra-cyclic pyrazole kinase inhibitors
WO2004092139A2 (fr) * 2003-04-07 2004-10-28 Gpc Biotech, Inc. Inhibiteurs de kinases dependant des cyclines, compositions et utilisations associees
AR045037A1 (es) 2003-07-10 2005-10-12 Aventis Pharma Sa Tetrahidro-1h-pirazolo [3,4-c] piridinas sustituidas, composiciones que las contienen y su utilizacion.
GB0326601D0 (en) * 2003-11-14 2003-12-17 Novartis Ag Organic compounds
EP1740579B1 (fr) 2004-03-24 2015-08-19 AbbVie Inc. Inhibiteurs de kinases a base de tyrazoles tricycliques
WO2005118543A1 (fr) * 2004-06-03 2005-12-15 Ono Pharmaceutical Co., Ltd. Inhibiteur des kinases et utilisation de cet inhibiteur
PT3424932T (pt) 2005-02-16 2021-05-19 Anacor Pharmaceuticals Inc Boronoftalidas para utilização terapêutica
JP2009519974A (ja) * 2005-12-16 2009-05-21 ジェネンテック・インコーポレーテッド 四環系キナーゼ阻害剤
RU2008131324A (ru) 2005-12-30 2010-02-10 Анакор Фармасьютикалз, Инк. (Us) Борсодержащие малые молекулы
ES2728455T3 (es) 2006-02-16 2019-10-24 Anacor Pharmaceuticals Inc Moléculas pequeñas que contienen boro como agentes antiinflamatorios
WO2007132307A1 (fr) 2006-05-09 2007-11-22 Pfizer Products Inc. Dérivés d'acide cycloalkylaminé et compositions pharmaceutiques à base de ceux-ci
KR101672511B1 (ko) 2008-03-06 2016-11-03 아나코르 파마슈티칼스 인코포레이티드 소염제로써 붕소가 함유된 소분자
WO2010028005A1 (fr) 2008-09-04 2010-03-11 Anacor Pharmaceuticals, Inc. Petites molécules contenant du bore
EP2348863A4 (fr) 2008-09-04 2012-03-07 Anacor Pharmaceuticals Inc Petites molécules contenant du bore
WO2011049971A1 (fr) 2009-10-20 2011-04-28 Anacor Pharmaceuticals, Inc. Petites molécules contenant du bore comme agents antiprotozoaires
EP2597089A1 (fr) * 2009-10-29 2013-05-29 Bristol-Myers Squibb Company Composés hétérocycliques tricycliques
WO2011060199A1 (fr) 2009-11-11 2011-05-19 Anacor Pharmaceuticals, Inc. Petites molecules contenant du bore
WO2011116348A1 (fr) 2010-03-19 2011-09-22 Anacor Pharmaceuticals, Inc. Petites molécules borées en tant qu'agent anti-protozoaire
EP2613788B1 (fr) 2010-09-07 2017-06-21 Anacor Pharmaceuticals, Inc. Petites molécules contenant du bore
US8853207B2 (en) 2012-04-12 2014-10-07 Development Center For Biotechnology Heterocyclic pyrazole compounds, method for preparing the same and use thereof
CN102675326B (zh) * 2012-04-26 2014-08-20 华东理工大学 3,4-二氢苯并吡喃[3,4-c]吡唑类三环化合物的制备方法
EP2909212B1 (fr) 2012-09-07 2017-02-22 Takeda Pharmaceutical Company Limited 1,4-dihydropyrazolo[4,3-b]indoles substituées
JP2016529234A (ja) * 2013-07-15 2016-09-23 ビーエーエスエフ ソシエタス・ヨーロピアBasf Se 殺有害生物化合物
WO2016113261A1 (fr) * 2015-01-13 2016-07-21 Basf Se Composés tricycliques fusionnés, compositions comprenant ces composés et leur utilisation aux fins de lutte contre les invertébrés nuisibles
CN105884828A (zh) 2015-02-16 2016-08-24 上海迪诺医药科技有限公司 多环化合物、其药物组合物及应用
CN110234633A (zh) 2016-09-02 2019-09-13 百时美施贵宝公司 取代的三环杂环化合物
US11225470B2 (en) 2016-11-28 2022-01-18 Sumitomo Chemical Company, Limited Tetrazolinone compounds and its use as pest control agents
WO2019032631A1 (fr) 2017-08-09 2019-02-14 Bristol-Myers Squibb Company Composés d'éther d'oxime
WO2019032632A1 (fr) 2017-08-09 2019-02-14 Bristol-Myers Squibb Company Composés d'alkylphényle
WO2021115560A1 (fr) * 2019-12-09 2021-06-17 Rottapharm Biotech S.R.L. Nouveaux inhibiteurs de kinases fyn et vegfr2
CN113773258B (zh) * 2020-06-09 2023-08-25 兰州大学 人源lrrk2蛋白小分子抑制剂及其应用

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3843665A (en) * 1973-04-11 1974-10-22 Sandoz Ag Process for preparing substituted indeno,naphtho and cyclohepta pyrazoles
JPS60130521A (ja) * 1983-12-19 1985-07-12 Morishita Seiyaku Kk 抗癌剤
AU706262B2 (en) * 1995-10-23 1999-06-10 Osteoscreen, Inc. Compositions and methods for treating bone deficit conditions
EP1021182A1 (fr) * 1997-10-06 2000-07-26 Basf Aktiengesellschaft Derives d'indeno 1,2-c]pyrazole utiles pour inhiber l'activite de tyrosine kinase
CN1278725A (zh) * 1997-10-06 2001-01-03 巴斯福股份公司 茚并[1,2-c]-、萘并[1,2-c]-和苯并[6,7]芳庚并[1,2-c]吡唑类衍生物

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO0027822A2 *

Also Published As

Publication number Publication date
KR20010086005A (ko) 2001-09-07
HUP0200310A3 (en) 2002-12-28
NO20012219D0 (no) 2001-05-04
ZA200103610B (en) 2002-09-23
WO2000027822A2 (fr) 2000-05-18
CA2350235A1 (fr) 2000-05-18
TR200102277T2 (tr) 2002-01-21
SK5282001A3 (en) 2002-01-07
JP2003517447A (ja) 2003-05-27
AU762992B2 (en) 2003-07-10
AU1909100A (en) 2000-05-29
ID30132A (id) 2001-11-08
WO2000027822A3 (fr) 2000-08-10
IL142584A0 (en) 2002-03-10
CN1335836A (zh) 2002-02-13
HUP0200310A2 (hu) 2002-11-28
HK1042895A1 (zh) 2002-08-30
BR9915132A (pt) 2001-08-07
BG105481A (en) 2001-12-29
NO20012219L (no) 2001-06-13
PL348210A1 (en) 2002-05-06
CZ20011563A3 (cs) 2003-02-12

Similar Documents

Publication Publication Date Title
AU762992B2 (en) Tricyclic pyrazole derivatives
US6462036B1 (en) Tricyclic pyrazole derivatives
US6297238B1 (en) Therapeutic agents
US7829570B2 (en) Substituted 4-amino isoxazolo[5,4-d]pyrimidines as kinase inhibitors
AU752474B2 (en) 4-aminopyrrolopyrimidines as kinase inhibitors
US20030153568A1 (en) Benzothiazole derivatives
EP1218373A2 (fr) 2-pyrazolin-5-ones utiles comme inhibiteurs de tyrosine kinase
EP1268481A2 (fr) Inhibiteurs de kinase utilises comme agents therapeutiques
EP1496910A1 (fr) Inhibiteurs de kinases
EP1165544B1 (fr) 1,4-dihydroindeno[1,2-c]pyrazoles substitues utilises comme inhibiteurs de la tyrosine kinase
EP1073435B1 (fr) Derives substitues de pyrazole tricyclique exer ant une activite de proteine kinase
US7060822B1 (en) 2-pyrazolin-5-ones

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20010606

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LI LU MC NL PT SE

AX Request for extension of the european patent

Free format text: AL PAYMENT 20010606;LT PAYMENT 20010606;LV PAYMENT 20010606;MK PAYMENT 20010606;RO PAYMENT 20010606;SI PAYMENT 20010606

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: ABBOTT GMBH & CO. KG

17Q First examination report despatched

Effective date: 20030918

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20050611

REG Reference to a national code

Ref country code: HK

Ref legal event code: WD

Ref document number: 1038351

Country of ref document: HK