US20100144783A1 - Kinase inhibitors with improved cyp safety profile - Google Patents

Kinase inhibitors with improved cyp safety profile Download PDF

Info

Publication number
US20100144783A1
US20100144783A1 US12/632,183 US63218309A US2010144783A1 US 20100144783 A1 US20100144783 A1 US 20100144783A1 US 63218309 A US63218309 A US 63218309A US 2010144783 A1 US2010144783 A1 US 2010144783A1
Authority
US
United States
Prior art keywords
cancer
pediatric
leukemia
carcinoma
aurora
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US12/632,183
Inventor
Michael R. Michaelides
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.)
AbbVie Inc
Original Assignee
Abbott Laboratories
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Abbott Laboratories filed Critical Abbott Laboratories
Priority to US12/632,183 priority Critical patent/US20100144783A1/en
Assigned to ABBOTT LABORATORIES reassignment ABBOTT LABORATORIES ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: Michaelides, Michael R.
Publication of US20100144783A1 publication Critical patent/US20100144783A1/en
Assigned to ABBOTT LABORATORIES reassignment ABBOTT LABORATORIES CORRECTIVE ASSIGNMENT TO CORRECT THE <ASSIGNOR&gt; PREVIOUSLY RECORDED ON REEL 023885 FRAME 0937. ASSIGNOR(S) HEREBY CONFIRMS THE <MICHAEL MICHAELIDES&gt; TO <MICHAEL MICHAELIDES AND ROBIN FREY AND MICHAEL CURTIN&gt;. Assignors: CURTIN, MICHAEL, FREY, ROBIN, Michaelides, Michael R.
Priority to US13/585,333 priority patent/US8722890B2/en
Assigned to ABBVIE INC. reassignment ABBVIE INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ABBOTT LABORATORIES
Abandoned legal-status Critical Current

Links

Classifications

    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4365Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system having sulfur as a ring hetero atom, e.g. ticlopidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D333/00Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom
    • C07D333/02Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings
    • C07D333/04Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings not substituted on the ring sulphur atom
    • C07D333/26Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings not substituted on the ring sulphur atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D333/30Hetero atoms other than halogen
    • C07D333/36Nitrogen atoms

Definitions

  • This invention pertains to compounds that inhibit protein kinases such as Aurora-kinases and the VEGFR and PDGFR families of kinases, and which have an improved safety profile due to low CYP3A4 inhibition, compositions containing the compounds, and methods of treating diseases using the compounds.
  • Mitosis is a process by which a complete copy of a duplicated genome is segregated by the microtuble spindle apparatus into two daughter cells.
  • Aurora-kinases key mitotic regulators required for genome stability, have been found to be overexpressed in human tumors. There is therefore an existing need in the therapeutic arts for compounds which inhibit Aurora-kinases, compositions comprising the inhibitors and methods of treating diseases during which Aurora-kinases are unregulated or overexpressed.
  • the reversible phosphorylation of proteins is one of the primary biochemical mechanisms mediating eukaryotic cell signaling. This reaction is catalyzed by protein kinases that transfer the g-phosphate group of ATP to hydroxyl groups on target proteins. 518 such enzymes exist in the human genome of which ⁇ 90 selectively catalyze the phosphorylation of tyrosine hydroxyl groups Cytosolic tyrosine kinases reside intracellularly whereas receptor tyrosine kinases (RTKs) possess both extracellular and intracellular domains and function as membrane spanning cell surface receptors. As such, RTKs mediate the cellular responses to environmental signals and facilitate a broad range of cellular processes including proliferation, migration and survival.
  • RTKs receptor tyrosine kinases
  • RTK signaling pathways are normally highly regulated, yet their over-activation has been shown to promote the growth, survival and metastasis of cancer cells.
  • Dysregulated RTK signaling occurs through gene over-expression or mutation and has been correlated with the progression of various human cancers.
  • the VEGF receptor (VEGFR) family consists of three RTKs, KDR (kinase insert domain-containing receptor; VEGFR2), FLT1 (Fms-like tyrosine kinase; VEGFR1), and FLT4 (VEGFR3). These receptors mediate the biological function of the vascular endothelial growth factors (VEGF-A, -B, -C, -D, -E and placenta growth factor (P1GF)), a family of homodimeric glycoproteins that bind the VEGF receptors with varying affinities.
  • KDR kinase insert domain-containing receptor
  • FLT1 Flms-like tyrosine kinase
  • VEGFR3 FLT4
  • KDR is the major mediator of the mitogenic, angiogenic and permeability-enhancing effects of VEGF-A, hereafter referred to as VEGF.
  • VEGF vascular endothelial growth factor-A
  • Many different cell types are able to produce VEGF, yet its biological activity is limited predominately to the vasculature by way of the endothelial cell-selective expression of KDR.
  • the VEGF/KDR axis is a primary mediator of angiogenesis, the means by which new blood vessels are formed from preexisting vessels.
  • FLT1 binds VEGF, VEGF-B and placental growth factor. FLT1 is expressed on the surface of smooth muscle cells, monocytes and hematopoietic stems cells in addition to endothelial cells. Activation of FLT1 signaling results in the mobilization of marrow-derived endothelial progenitor cells that are recruited to tumors where they contribute to new blood vessel formation.
  • Lymphatic vessels are one of the routes by which cancer cells disseminate from solid tumors during metastasis.
  • the PDGF receptor (PDGFR) family consists of five RTK's, PDGFR-a and b, CSF1R, KIT, and FLT3.
  • the a and b isoforms of the platelet-derived growth factor (PDGF) receptors occur as homodimers or a/b heterodimers and are found most commonly on the surface of fibroblasts and smooth muscle cells.
  • PDGFR-b contributes to tumor angiogenesis through the proliferation and migration of pericytes, the peri-endothelial cells that associate with and stabilize immature blood vessels.
  • autocrine PDGFR stimulation brought about by the co-expression of PDGF and PDGF receptors, mediates tumor cell proliferation and survival.
  • CSF-1R is encoded by the cellular homolog of the retroviral oncogene v-fms and is a major regulator of macrophage development. Macrophages are frequent components of tumor stroma and have been shown to modify the extracellular matrix in a manner beneficial to tumor growth and metastasis.
  • KIT is expressed by hematopoietic progenitor cells, mast cells, germ cells and by pacemaker cells in the gut (interstitial cells of Cajal). It contributes to tumor progression by two general mechanisms namely autocrine stimulation by its ligand, stem cell factor (SCF), and through mutations that result in ligand-independent kinase activity.
  • SCF stem cell factor
  • FLT3 is normally expressed on hematopoietic stem cells where its interaction with FLT3 ligand (FL) stimulates stem cell survival, proliferation and differentiation. In addition to being over-expressed in various leukemia cells, FLT3 is frequently mutated in hematological malignancies with approximately one-third of patients with acute myeloid leukemia (AML) harboring activating mutations.
  • AML acute myeloid leukemia
  • the identification of effective small compounds which specifically inhibit signal transduction and cellular proliferation by modulating the activity of tyrosine 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.
  • cytochrome P450 also known as CYP or P450
  • CYP3A subfamily which includes CYP3A4, 3A5, 3A7 and 3A43 is responsible for the metabolism of about 60% of known therapeutic drugs.
  • CYP3A4 in particular is the most abundant CYP isoenzyme in both liver and intestine and is involved in the metabolism of more than 50% of the clinically used drugs (Mechanism-Based Inhibition of Cytochrome P455 3A4 by Therapeutic Drugs. Clin. Pharmacokinet, 2005, 44, 279-304). Like all other CYP enzymes, CYP3A4 is susceptible to both reversible and pseudo-irreversible or irreversible (mechanism based) inhibition (Time-dependent CYP Inhibition. Expert Opin. Drug Metab. Toxicol. 2007, 3, 51-66). Their low substrate specificity makes the CYP enzymes susceptible to inhibition by a wide variety of structurally distinct drugs.
  • thienopyridine compounds disclosed in WO2005/010009 display potent inhibition of Aurora and PDGFR/VEGFR kinases, they may also be inhibitors of CYP3A4.
  • This invention pertains to novel thienopyridines of formula I, which maintain potent inhibition of both Aurora kinases and the family of kinases encompassing PDGFR and VEGFR and also demonstrate at least a 10-30 fold reduction in CYP3A4 inhibition. Because the compounds of the present invention have significantly reduced CYP3A4 inhibition, they are expected to display low drug-drug interaction liability.
  • the compounds of the invention have demonstrated their utility in additional assays utilized to assess the suitability of the compounds as drug candidates.
  • the compounds of the invention demonstrate potency in whole cell assays (e.g., in the Human Umbilical Vein Endothelial Cell (HUVEC) assay and the assay measuring histone D3 phosphorylation and induction of polyploidy) and suitable pharmokinetic properties (e.g., oral clearance and oral bioavailability), in vivo efficacy (e.g., Uterine Edema model, rodent flank and orthotopic tumor models), cardiovascular safety, CNS assessments and gastroinstestinal assays.
  • HUVEC Human Umbilical Vein Endothelial Cell
  • the present invention provides compounds of formula (I)
  • R 1 is hydroxyalkyl
  • R 2 is selected from the group consisting of alkoxy, alkyl, halo, and haloalkoxy;
  • R 3 is hydrogen or alkyl.
  • Still another embodiment pertains to methods of treating diseases involving mediation, overexpression or disregulation of kinases in a mammal, the methods comprising administering thereto a therapeutically effective amount of a compound having Formula I,
  • R 1 is hydroxyalkyl
  • R 2 is selected from the group consisting of alkoxy, alkyl, halo, and haloalkoxy;
  • R 3 is hydrogen or alkyl.
  • Another embodiment pertains to methods of treating acoustic neuroma, acute leukemia, acute lymphocytic leukemia, acute myelocytic leukemia (monocytic, myeloblastic, adenocarcinoma, angiosarcoma, astrocytoma, myelomonocytic and promyelocytic), acute t-cell leukemia, basal cell carcinoma, bile duct carcinoma, bladder cancer, brain cancer, breast cancer, bronchogenic carcinoma, cervical cancer, chondrosarcoma, chordoma, choriocarcinoma, chronic leukemia, chronic lymphocytic leukemia, chronic myelocytic (granulocytic) leukemia, chronic myleogeneous leukemia, colon cancer, colorectal cancer, craniopharyngioma, cystadenocarcinoma, diffuse large B-cell lymphoma, dysproliferative changes (dysplasias and metaplasias),
  • Another embodiment pertains to methods of treating acoustic neuroma, acute leukemia, acute lymphocytic leukemia, acute myelocytic leukemia (monocytic, myeloblastic, adenocarcinoma, angiosarcoma, astrocytoma, myelomonocytic and promyelocytic), acute t-cell leukemia, basal cell carcinoma, bile duct carcinoma, bladder cancer, brain cancer, breast cancer, bronchogenic carcinoma, cervical cancer, chondrosarcoma, chordoma, choriocarcinoma, chronic leukemia, chronic lymphocytic leukemia, chronic myelocytic (granulocytic) leukemia, chronic myleogeneous leukemia, colon cancer, colorectal cancer, craniopharyngioma, cystadenocarcinoma, diffuse large B-cell lymphoma, dysproliferative changes (dysplasias and metaplasias),
  • Another embodiment pertains to methods of treating acoustic neuroma, acute leukemia, acute lymphocytic leukemia, acute myelocytic leukemia (monocytic, myeloblastic, adenocarcinoma, angiosarcoma, astrocytoma, myelomonocytic and promyelocytic), acute t-cell leukemia, basal cell carcinoma, bile duct carcinoma, bladder cancer, brain cancer, breast cancer, bronchogenic carcinoma, cervical cancer, chondrosarcoma, chordoma, choriocarcinoma, chronic leukemia, chronic lymphocytic leukemia, chronic myelocytic (granulocytic) leukemia, chronic myleogeneous leukemia, colon cancer, colorectal cancer, craniopharyngioma, cystadenocarcinoma, diffuse large B-cell lymphoma, dysproliferative changes (dysplasias and metaplasias),
  • Another embodiment pertains to methods of treating acoustic neuroma, acute leukemia, acute lymphocytic leukemia, acute myelocytic leukemia (monocytic, myeloblastic, adenocarcinoma, angiosarcoma, astrocytoma, myelomonocytic and promyelocytic), acute t-cell leukemia, basal cell carcinoma, bile duct carcinoma, bladder cancer, brain cancer, breast cancer, bronchogenic carcinoma, cervical cancer, chondrosarcoma, chordoma, choriocarcinoma, chronic leukemia, chronic lymphocytic leukemia, chronic myelocytic (granulocytic) leukemia, chronic myleogeneous leukemia, colon cancer, colorectal cancer, craniopharyngioma, cystadenocarcinoma, diffuse large B-cell lymphoma, dysproliferative changes (dysplasias and metaplasias),
  • Another embodiment pertains to methods of treating acoustic neuroma, acute leukemia, acute lymphocytic leukemia, acute myelocytic leukemia (monocytic, myeloblastic, adenocarcinoma, angiosarcoma, astrocytoma, myelomonocytic and promyelocytic), acute t-cell leukemia, basal cell carcinoma, bile duct carcinoma, bladder cancer, brain cancer, breast cancer, bronchogenic carcinoma, cervical cancer, chondrosarcoma, chordoma, choriocarcinoma, chronic leukemia, chronic lymphocytic leukemia, chronic myelocytic (granulocytic) leukemia, chronic myleogeneous leukemia, colon cancer, colorectal cancer, craniopharyngioma, cystadenocarcinoma, diffuse large B-cell lymphoma, dysproliferative changes (dysplasias and metaplasias),
  • Another embodiment pertains to methods of treating acoustic neuroma, acute leukemia, acute lymphocytic leukemia, acute myelocytic leukemia (monocytic, myeloblastic, adenocarcinoma, angiosarcoma, astrocytoma, myelomonocytic and promyelocytic), acute t-cell leukemia, basal cell carcinoma, bile duct carcinoma, bladder cancer, brain cancer, breast cancer, bronchogenic carcinoma, cervical cancer, chondrosarcoma, chordoma, choriocarcinoma, chronic leukemia, chronic lymphocytic leukemia, chronic myelocytic (granulocytic) leukemia, chronic myleogeneous leukemia, colon cancer, colorectal cancer, craniopharyngioma, cystadenocarcinoma, diffuse large B-cell lymphoma, dysproliferative changes (dysplasias and metaplasias),
  • compositions comprising an excipient and a therapeutically effective amount of a compound of formula I, and a therapeutically effective amount of one additional therapeutic agent or more than one additional therapeutic agent.
  • compositions comprising an excipient and a therapeutically effective amount of N-(4- ⁇ 4-amino-7-[1-(2-hydroxyethyl)-1H-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl ⁇ phenyl)-N′-(3-fluorophenyl)urea, and a therapeutically effective amount of one additional therapeutic agent or more than one additional therapeutic agent.
  • compositions comprising an excipient and a therapeutically effective amount of N-(4- ⁇ 4-amino-7-[1-(2-hydroxyethyl)-1H-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl ⁇ phenyl)-N′-[4-(difluoromethoxy)phenyl]urea, and a therapeutically effective amount of one additional therapeutic agent or more than one additional therapeutic agent.
  • compositions comprising an excipient and a therapeutically effective amount of N-[4-(4-amino-7- ⁇ 1-[(2S)-2-hydroxypropyl]-1H-pyrazol-4-yl ⁇ thieno[3,2-c]pyridin-3-yl)phenyl]-N′-(3-methylphenyl)urea, and a therapeutically effective amount of one additional therapeutic agent or more than one additional therapeutic agent.
  • compositions comprising an excipient and a therapeutically effective amount of N-(4- ⁇ 4-amino-7-[1-(2-hydroxy-2-methylpropyl)-1H-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl ⁇ phenyl)-N′-(4-methoxyphenyl)urea, and a therapeutically effective amount of one additional therapeutic agent or more than one additional therapeutic agent.
  • compositions comprising an excipient and a therapeutically effective amount of a N-[4-(4-amino-7- ⁇ 1-[(2S)-2,3-dihydroxypropyl]-1H-pyrazol-4-yl ⁇ thieno[3,2-c]pyridin-3-yl)phenyl]-N′-(4-methoxyphenyl)urea, and a therapeutically effective amount of one additional therapeutic agent or more than one additional therapeutic agent.
  • Still another embodiment pertains to methods of treating diseases involving mediation, overexpression or disregulation of kinases in a mammal, the methods comprising administering thereto a therapeutically effective amount of a compound of formula I, and a therapeutically effective amount of one additional therapeutic agent or more than one additional therapeutic agent alone or in combination with radiotherapy.
  • Still another embodiment pertains to methods of treating diseases involving mediation, overexpression or disregulation of kinases in a mammal, the methods comprising administering thereto a therapeutically effective amount of N-(4- ⁇ 4-amino-7-[1-(2-hydroxyethyl)-1H-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl ⁇ phenyl)-N′-(3-fluorophenyl)urea, and a therapeutically effective amount of one additional therapeutic agent or more than one additional therapeutic agent alone or in combination with radiotherapy.
  • Still another embodiment pertains to methods of treating diseases involving mediation, overexpression or disregulation of kinases in a mammal, the methods comprising administering thereto a therapeutically effective amount of N-(4- ⁇ 4-amino-7-[1-(2-hydroxyethyl)-1H-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl ⁇ phenyl)-N′-[4-(difluoromethoxy)phenyl]urea, and a therapeutically effective amount of one additional therapeutic agent or more than one additional therapeutic agent alone or in combination with radiotherapy.
  • Still another embodiment pertains to methods of treating diseases involving mediation, overexpression or disregulation of kinases in a mammal, the methods comprising administering thereto a therapeutically effective amount of N-[4-(4-amino-7- ⁇ 1-[(2S)-2-hydroxypropyl]-1H-pyrazol-4-yl ⁇ thieno[3,2-c]pyridin-3-yl)phenyl]-N′-(3-methylphenyl)urea, and a therapeutically effective amount of one additional therapeutic agent or more than one additional therapeutic agent alone or in combination with radiotherapy.
  • Still another embodiment pertains to methods of treating diseases involving mediation, overexpression or disregulation of kinases in a mammal, the methods comprising administering thereto a therapeutically effective amount of N-(4- ⁇ 4-amino-7-[1-(2-hydroxy-2-methylpropyl)-1H-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl ⁇ phenyl)-N′-(4-methoxyphenyl)urea, and a therapeutically effective amount of one additional therapeutic agent or more than one additional therapeutic agent alone or in combination with radiotherapy.
  • Still another embodiment pertains to methods of treating diseases involving mediation, overexpression or disregulation of kinases in a mammal, the methods comprising administering thereto a therapeutically effective amount of N-[4-(4-amino-7- ⁇ 1-[(2S)-2,3-dihydroxypropyl]-1H-pyrazol-4-yl ⁇ thieno[3,2-c]pyridin-3-yl)phenyl]-N′-(4-methoxyphenyl)urea, and a therapeutically effective amount of one additional therapeutic agent or more than one additional therapeutic agent alone or in combination with radiotherapy.
  • Another embodiment pertains to methods of treating pediatric cancer or neoplasm such as embryonal rhabdomyosarcoma, pediatric acute lymphoblastic leukemia, pediatric acute myelogenous leukemia, pediatric alveolar rhabdomyosarcoma, pediatric anaplastic ependymoma, pediatric anaplastic large cell lymphoma, pediatric anaplastic medulloblastoma, pediatric atypical teratoid/rhabdoid tumor of the central nervous system, pediatric biphenotypic acute leukemia, pediatric Burkitts lymphoma, pediatric cancers of Ewing's family of tumors such as primitive neuroectodermal rumors, pediatric diffuse anaplastic Wilm's tumor, pediatric favorable histology Wilm's tumor, pediatric glioblastoma, pediatric medulloblastoma, pediatric neuroblastoma, pediatric neuroblastoma-derived myelocytomatosis, pediatric pre-B-cell cancers (such as leukemia), pediatric
  • Another embodiment pertains to methods of treating pediatric cancer or neoplasm such as embryonal rhabdomyosarcoma, pediatric acute lymphoblastic leukemia, pediatric acute myelogenous leukemia, pediatric alveolar rhabdomyosarcoma, pediatric anaplastic ependymoma, pediatric anaplastic large cell lymphoma, pediatric anaplastic medulloblastoma, pediatric atypical teratoid/rhabdoid tumor of the central nervous system, pediatric biphenotypic acute leukemia, pediatric Burkitts lymphoma, pediatric cancers of Ewing's family of tumors such as primitive neuroectodermal rumors, pediatric diffuse anaplastic Wilm's tumor, pediatric favorable histology Wilm's tumor, pediatric glioblastoma, pediatric medulloblastoma, pediatric neuroblastoma, pediatric neuroblastoma-derived myelocytomatosis, pediatric pre-B-cell cancers (such as leukemia), pediatric
  • Another embodiment pertains to methods of treating pediatric cancer or neoplasm such as embryonal rhabdomyosarcoma, pediatric acute lymphoblastic leukemia, pediatric acute myelogenous leukemia, pediatric alveolar rhabdomyosarcoma, pediatric anaplastic ependymoma, pediatric anaplastic large cell lymphoma, pediatric anaplastic medulloblastoma, pediatric atypical teratoid/rhabdoid tumor of the central nervous system, pediatric biphenotypic acute leukemia, pediatric Burkitts lymphoma, pediatric cancers of Ewing's family of tumors such as primitive neuroectodermal rumors, pediatric diffuse anaplastic Wilm's tumor, pediatric favorable histology Wilm's tumor, pediatric glioblastoma, pediatric medulloblastoma, pediatric neuroblastoma, pediatric neuroblastoma-derived myelocytomatosis, pediatric pre-B-cell cancers (such as leukemia), pediatric
  • Another embodiment pertains to methods of treating pediatric cancer or neoplasm such as embryonal rhabdomyosarcoma, pediatric acute lymphoblastic leukemia, pediatric acute myelogenous leukemia, pediatric alveolar rhabdomyosarcoma, pediatric anaplastic ependymoma, pediatric anaplastic large cell lymphoma, pediatric anaplastic medulloblastoma, pediatric atypical teratoid/rhabdoid tumor of the central nervous system, pediatric biphenotypic acute leukemia, pediatric Burkitts lymphoma, pediatric cancers of Ewing's family of tumors such as primitive neuroectodermal rumors, pediatric diffuse anaplastic Wilm's tumor, pediatric favorable histology Wilm's tumor, pediatric glioblastoma, pediatric medulloblastoma, pediatric neuroblastoma, pediatric neuroblastoma-derived myelocytomatosis, pediatric pre-B-cell cancers (such as leukemia), pediatric
  • Another embodiment pertains to methods of treating pediatric cancer or neoplasm such as embryonal rhabdomyosarcoma, pediatric acute lymphoblastic leukemia, pediatric acute myelogenous leukemia, pediatric alveolar rhabdomyosarcoma, pediatric anaplastic ependymoma, pediatric anaplastic large cell lymphoma, pediatric anaplastic medulloblastoma, pediatric atypical teratoid/rhabdoid tumor of the central nervous system, pediatric biphenotypic acute leukemia, pediatric Burkitts lymphoma, pediatric cancers of Ewing's family of tumors such as primitive neuroectodermal rumors, pediatric diffuse anaplastic Wilm's tumor, pediatric favorable histology Wilm's tumor, pediatric glioblastoma, pediatric medulloblastoma, pediatric neuroblastoma, pediatric neuroblastoma-derived myelocytomatosis, pediatric pre-B-cell cancers (such as leukemia), pediatric
  • Another embodiment pertains to methods of treating pediatric cancer or neoplasm such as embryonal rhabdomyosarcoma, pediatric acute lymphoblastic leukemia, pediatric acute myelogenous leukemia, pediatric alveolar rhabdomyosarcoma, pediatric anaplastic ependymoma, pediatric anaplastic large cell lymphoma, pediatric anaplastic medulloblastoma, pediatric atypical teratoid/rhabdoid tumor of the central nervous system, pediatric biphenotypic acute leukemia, pediatric Burkitts lymphoma, pediatric cancers of Ewing's family of tumors such as primitive neuroectodermal rumors, pediatric diffuse anaplastic Wilm's tumor, pediatric favorable histology Wilm's tumor, pediatric glioblastoma, pediatric medulloblastoma, pediatric neuroblastoma, pediatric neuroblastoma-derived myelocytomatosis, pediatric pre-B-cell cancers (such as leukemia), pediatric
  • Still another embodiment pertains to N-(4- ⁇ 4-amino-7-[1-(2-hydroxyethyl)-1H-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl ⁇ phenyl)-N′-(3-fluorophenyl)urea.
  • Still another embodiment pertains to N-(4- ⁇ 4-amino-7-[1-(2-hydroxyethyl)-1H-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl ⁇ phenyl)-N′-[4-(difluoromethoxy)phenyl]urea.
  • Still another embodiment pertains to N-[4-(4-amino-7- ⁇ 1-[(2S)-2-hydroxypropyl]-1H-pyrazol-4-yl ⁇ thieno[3,2-c]pyridin-3-yl)phenyl]-N′-(3-methylphenyl)urea.
  • Still another embodiment pertains to N-(4- ⁇ 4-amino-7-[1-(2-hydroxy-2-methylpropyl)-1H-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl ⁇ phenyl)-N′-(4-methoxyphenyl)urea.
  • Still another embodiment pertains to N-[4-(4-amino-7- ⁇ 1-[(2S)-2,3-dihydroxypropyl]-1H-pyrazol-4-yl ⁇ thieno[3,2-c]pyridin-3-yl)phenyl]-N′-(4-methoxyphenyl)urea.
  • Still another embodiment pertains to a compound of formula I, and therapeutically acceptable salts, prodrugs, esters, amides, salts of prodrugs, salts of esters, and salts of amides thereof.
  • Still another embodiment pertains to N-(4- ⁇ 4-amino-7-[1-(2-hydroxyethyl)-1H-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl ⁇ phenyl)-N′-(3-fluorophenyl)urea, and therapeutically acceptable salts, prodrugs, esters, amides, salts of prodrugs, salts of esters, and salts of amides thereof.
  • Still another embodiment pertains to N-(4- ⁇ 4-amino-7-[1-(2-hydroxyethyl)-1H-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl ⁇ phenyl)-N′-[4-(difluoromethoxy)phenyl]urea, and therapeutically acceptable salts, prodrugs, esters, amides, salts of prodrugs, salts of esters, and salts of amides thereof.
  • Still another embodiment pertains to N-[4-(4-amino-7- ⁇ 1-[(2S)-2-hydroxypropyl]-1H-pyrazol-4-yl ⁇ thieno[3,2-c]pyridin-3-yl)phenyl]-N′-(3-methylphenyl)urea, and therapeutically acceptable salts, prodrugs, esters, amides, salts of prodrugs, salts of esters, and salts of amides thereof.
  • Still another embodiment pertains to N-(4- ⁇ 4-amino-7-[1-(2-hydroxy-2-methylpropyl)-1H-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl ⁇ phenyl)-N′-(4-methoxyphenyl)urea, and therapeutically acceptable salts, prodrugs, esters, amides, salts of prodrugs, salts of esters, and salts of amides thereof.
  • Still another embodiment pertains to N-[4-(4-amino-7- ⁇ 1-[(2S)-2,3-dihydroxypropyl]-1H-pyrazol-4-yl ⁇ thieno[3,2-c]pyridin-3-yl)phenyl]-N-(4-methoxyphenyl)urea, and therapeutically acceptable salts, prodrugs, esters, amides, salts of prodrugs, salts of esters, and salts of amides thereof.
  • Variable moieties of compounds herein are represented by identifiers (capital letters with numerical and/or alphabetical superscripts) and may be specifically embodied.
  • variable moiety may be the same or different as another specific embodiment having the same identifier.
  • alkoxy refers to an alkyl group attached to the parent molecular moiety through an oxygen atom.
  • alkyl refers to a group derived from a straight or branched chain saturated hydrocarbon containing from one to ten carbon atoms.
  • halo and “halogen,” as used herein, refer to F, Cl, Br, or I.
  • haloalkoxy refers to a haloalkyl group attached to the parent molecular moiety through an oxygen atom.
  • haloalkyl refers to an alkyl group, as defined herein, substituted with at least one halogen, as defined herein.
  • hydroxy refers to a —OH group.
  • hydroxyalkyl refers to an alkyl group substituted with at least one hydroxy group.
  • KDR means kinase insert domain receptor (a type III receptor tyrosine kinase) and is also known as FLK1, VEGFR, VEGFR2, and CD309.
  • VEGFR vascular endothelial growth factor receptor
  • PDGFR platelet-derived growth factor receptor
  • Compounds of this invention may contain asymmetrically substituted carbon atoms in the R or S configuration, wherein the terms “R” and “S” are as defined in Pure Appl. Chem. (1976) 45, 13-10.
  • Compounds having asymmetrically substituted carbon atoms with equal amounts of R and S configurations are racemic at those atoms. Atoms having excess of one configuration over the other are assigned the configuration in excess, preferably an excess of about 85%-90%, more preferably an excess of about 95%-99%, and still more preferably an excess greater than about 99%. Accordingly, this invention is meant to embrace racemic mixtures and relative and absolute diastereoisomers of the compounds thereof.
  • Compounds of this invention may also contain carbon-carbon double bonds or carbon-nitrogen double bonds in the E or Z configuration, wherein the term “E” represents higher order substituents on opposite sides of the carbon-carbon or carbon-nitrogen double bond and the term “Z” represents higher order substituents on the same side of the carbon-carbon or carbon-nitrogen double bond as determined by the Cahn-Ingold-Prelog Priority Rules.
  • the compounds of this invention may also exist as a mixture of “E” and “Z” isomers.
  • Compounds of this invention may also exist as tautomers or equilibrium mixtures thereof wherein a proton of a compound shifts from one atom to another.
  • tautomers include, but are not limited to, keto-enol, phenol-keto, oxime-nitroso, nitro-aci, imine-enamine and the like.
  • prodrug-forming moieties may have attached thereto prodrug-forming moieties.
  • the prodrug-forming moieties are removed by metabolic processes and release the compounds having the freed NH, C(O)OH, OH or SH in vivo.
  • Prodrugs are useful for adjusting such pharmacokinetic properties of the compounds as solubility and/or hydrophobicity, absorption in the gastrointestinal tract, bioavailability, tissue penetration, and rate of clearance.
  • Metabolites of compounds of this invention produced by in vitro or in vivo metabolic processes may also have utility for treating diseases associated with overexpression or disregulation of protein kinases.
  • Certain precursor compounds which may be metabolized in vitro or in vivo to form compounds of this invention may also have utility for treating diseases associated with overexpression or disregulation of protein kinases.
  • Compounds of this invention may exist as acid addition salts, basic addition salts or zwitterions. Salts of compounds having Formula I are prepared during their isolation or following their purification. Acid addition salts are those derived from the reaction of a compound of this invention with acid.
  • salts including the acetate, adipate, alginate, bicarbonate, citrate, aspartate, benzoate, benzenesulfonate (besylate), bisulfate, butyrate, camphorate, camphorsulfonate, digluconate, formate, fumarate, glycerophosphate, glutamate, hemisulfate, heptanoate, hexanoate, hydrochloride, hydrobromide, hydroiodide, lactobionate, lactate, maleate, mesitylenesulfonate, methanesulfonate, naphthylenesulfonate, nicotinate, oxalate, pamoate, pectinate, persulfate, phosphate, picrate, propionate, succinate, tartrate, thiocyanate, trichloroacetic, trifluoroacetic, para-toluenesulfonate and undecan
  • Compounds having Formula I may be administered, for example, bucally, ophthalmically, orally, osmotically, parenterally (intramuscularly, intraperintoneally intrasternally, intravenously, subcutaneously), rectally, topically, transdermally, vaginally and intraarterially as well as by intraarticular injection, infusion, and placement in the body, such as, for example, the vasculature.
  • Therapeutically effective amounts of a compound having Formula I depend on recipient of treatment, disease treated and severity thereof, composition comprising it, time of administration, route of administration, duration of treatment, potency, rate of clearance and whether or not another drug is co-administered.
  • the amount of a compound having Formula I used to make a composition to be administered daily to a patient in a single dose or in divided doses is from about 0.03 to about 200 mg/kg body weight.
  • Single dose compositions contain these amounts or a combination of submultiples thereof.
  • Excipients include, but are not limited to, encapsulators and additives such as absorption accelerators, antioxidants, binders, buffers, coating agents, coloring agents, diluents, disintegrating agents, emulsifiers, extenders, fillers, flavoring agents, humectants, lubricants, perfumes, preservatives, propellants, releasing agents, sterilizing agents, sweeteners, solubilizers, wetting agents, mixtures thereof and the like.
  • encapsulators and additives such as absorption accelerators, antioxidants, binders, buffers, coating agents, coloring agents, diluents, disintegrating agents, emulsifiers, extenders, fillers, flavoring agents, humectants, lubricants, perfumes, preservatives, propellants, releasing agents, sterilizing agents, sweeteners, solubilizers, wetting agents, mixtures thereof and the like.
  • Excipients for preparation of compositions comprising a compound having Formula Ito be administered orally include, but are not limited to, agar, alginic acid, aluminum hydroxide, benzyl alcohol, benzyl benzoate, 1,3-butylene glycol, carbomers, castor oil, cellulose, cellulose acetate, cocoa butter, corn starch, corn oil, cottonseed oil, cross-povidone, diglycerides, ethanol, ethyl cellulose, ethyl laureate, ethyl oleate, fatty acid esters, gelatin, germ oil, glucose, glycerol, groundnut oil, hydroxypropylmethyl celluose, isopropanol, isotonic saline, lactose, magnesium hydroxide, magnesium stearate, malt, mannitol, monoglycerides, olive oil, peanut oil, potassium phosphate salts, potato starch, povidone, propylene glycol, Ring
  • Excipients for preparation of compositions comprising a compound having Formula Ito be administered ophthalmically or orally include, but are not limited to, 1,3-butylene glycol, castor oil, corn oil, cottonseed oil, ethanol, fatty acid esters of sorbitan, germ oil, groundnut oil, glycerol, isopropanol, olive oil, polyethylene glycols, propylene glycol, sesame oil, water, mixtures thereof and the like.
  • Excipients for preparation of compositions comprising a compound having Formula I to be administered osmotically include, but are not limited to, chlorofluorohydrocarbons, ethanol, water, mixtures thereof and the like.
  • Excipients for preparation of compositions comprising a compound having Formula Ito be administered parenterally include, but are not limited to, 1,3-butanediol, castor oil, corn oil, cottonseed oil, dextrose, germ oil, groundnut oil, liposomes, oleic acid, olive oil, peanut oil, Ringer's solution, safflower oil, sesame oil, soybean oil, U.S.P. or isotonic sodium chloride solution, water, mixtures thereof and the like.
  • Excipients for preparation of compositions comprising a compound having Formula Ito be administered rectally or vaginally include, but are not limited to, cocoa butter, polyethylene glycol, wax, mixtures thereof and the like.
  • Compounds having Formula I are expected to be useful when used with alkylating agents, angiogenesis inhibitors, antibodies, antimetabolites, antimitotics, antiproliferatives, aurora kinase inhibitors, other apoptosis promoters (for example, Bcl-xL, Bcl-w and Bfl-1) inhibitors, Bcr-Abl kinase inhibitors, BiTE (Bi-Specific T cell Engager) antibodies, biologic response modifiers, cyclin-dependent kinase inhibitors, cell cycle inhibitors, cyclooxygenase-2 inhibitors, DVD's, leukemia viral oncogene homolog (ErbB2) receptor inhibitors, growth factor inhibitors, heat shock protein (HSP)-90 inhibitors, histone deacetylase (HDAC) inhibitors, hormonal therapies, immunologicals, inhibitors of apoptosis proteins (IAP's) intercalating antibiotics, kinase inhibitors, mammalian target of rapamycin
  • a BiTE antibody is a bi-specific antibody that directs T-cells to attach cancer cells by simultaneously binding the two cells. The T-cell then attacks the target cancer cell.
  • Exemplary BiTE antibodies include adecatumumab (Micromet MT201), blinatumomab (Micromet MT103) and the like.
  • SiRNA's are molecules having endogenous RNA bases or chemically modified nucleotides. The modifications shall not abolish cellular activity, but rather impart increased stability and/or increased cellular potency. Examples of chemical modifications include phosphorothioate groups, 2′-deoxynucleotide, 2′-OCH 3 -containing ribonucleotides, 2′-F-ribonucleotides, 2′-methoxyethyl ribonucleotides or a combination thereof.
  • the siRNA can have varying lengths (10-200 bps) and structures (hairpins, single/double strands, bulges, nicks/gaps, mismatches) and processed in the cell to provide active gene silencing.
  • a double-stranded siRNA can have the same number of nucleotides on each strand (blunt ends) or asymmetric ends (overhangs).
  • the overhang of 1-2 nucleotides can be present on the sense and/or the antisense strand, as well as present on the 5′- and/or the 3′- ends of a given strand.
  • Multivalent binding proteins are binding proteins comprising two or more antigen binding sites.
  • the multivalent binding protein is preferably engineered to have the three or more antigen binding sites and is generally not a naturally occurring antibody.
  • the term “multispecific binding protein” means a binding protein capable of binding two or more related or unrelated targets.
  • Dual variable domain (DVD) binding proteins are tetravalent or multivalent binding proteins binding proteins comprising two or more antigen binding sites.
  • DVDs may be monospecific, i.e., capable of binding one antigen or multispecific, i.e., capable of binding two or more antigens.
  • DVD binding proteins comprising two heavy chain DVD polypeptides and two light chain DVD polypeptides are referred to as DVD Ig.
  • Each half of a DVD Ig comprises a heavy chain DVD polypeptide, a light chain DVD polypeptide, and two antigen binding sites.
  • Each binding site comprises a heavy chain variable domain and a light chain variable domain with a total of 6 CDRs involved in antigen binding per antigen binding site.
  • Alkylating agents include altretamine, AMD-473, AP-5280, apaziquone, bendamustine, brostallicin, busulfan, carboquone, carmustine (BCNU), chlorambucil, CLORETAZINE® (laromustine, VNP 40101M), cyclophosphamide, decarbazine, estramustine, fotemustine, glufosfamide, ifosfamide, KW-2170, lomustine (CCNU), mafosfamide, melphalan, mitobronitol, mitolactol, nimustine, nitrogen mustard N-oxide, ranimustine, temozolomide, thiotepa, TREANDA® (bendamustine), treosulfan, rofosfamide and the like.
  • Angiogenesis inhibitors include endothelial-specific receptor tyrosine kinase (Tie-2) inhibitors, epidermal growth factor receptor (EGFR) inhibitors, insulin growth factor-2 receptor (IGFR-2) inhibitors, matrix metalloproteinase-2 (MMP-2) inhibitors, matrix metalloproteinase-9 (MMP-9) inhibitors, platelet-derived growth factor receptor (PDGFR) inhibitors, thrombospondin analogs, vascular endothelial growth factor receptor tyrosine kinase (VEGFR) inhibitors and the like.
  • Tie-2 endothelial-specific receptor tyrosine kinase
  • EGFR epidermal growth factor receptor
  • IGFR-2 insulin growth factor-2 receptor
  • MMP-2 matrix metalloproteinase-2
  • MMP-9 matrix metalloproteinase-9
  • PDGFR platelet-derived growth factor receptor
  • VEGFR vascular endothelial growth factor receptor tyrosine
  • Antimetabolites include ALIMTA® (metrexed disodium, LY231514, MTA), 5-azacitidine, XELODA® (capecitabine), carmofur, LEUSTAT® (cladribine), clofarabine, cytarabine, cytarabine ocfosfate, cytosine arabinoside, decitabine, deferoxamine, doxifluridine, eflornithine, EICAR (5-ethynyl-1- ⁇ -D-ribofuranosylimidazole-4-carboxamide), enocitabine, ethnylcytidine, fludarabine, 5-fluorouracil alone or in combination with leucovorin, GEMZAR® (gemcitabine), hydroxyurea, ALKERAN® (melphalan), mercaptopurine, 6-mercaptopurine riboside, methotrexate, mycophenolic acid, nelara
  • Aurora kinase inhibitors include AZD-1152, MLN-8054, VX-680 and the like.
  • Bcl-2 proteins inhibitors include AT-101 (( ⁇ )gossypol), GENASENSE® (G3139 or oblimersen (Bcl-2-targeting antisense oligonucleotide)), IPI-194, IPI-565, N-(4-(4-((4′-chloro(1,1′-biphenyl)-2-yl)methyl)piperazin-1-yl)benzoyl)-4-(((1R)-3-(dimethylamino)-1-((phenylsulfanyl)methyl)propyl)amino)-3-nitrobenzenesulfonamide) (ABT-737), N-(4-(4-((2-(4-chlorophenyl)-5,5-dimethyl-1-cyclohex-1-en-1-yl)methyl)piperazin-1-yl)benzoyl)-4-(((1R)-3-(morpholin-4-yl)-1-((pheny
  • Bcr-Abl kinase inhibitors include DASATINIB® (BMS-354825), GLEEVEC® (imatinib) and the like.
  • CDK inhibitors include AZD-5438, BMI-1040, BMS-032, BMS-387, CVT-2584, flavopyridol, GPC-286199, MCS-5A, PD0332991, PHA-690509, seliciclib (CYC-202, R-roscovitine), ZK-304709 and the like.
  • COX-2 inhibitors include ABT-963, ARCOXIA® (etoricoxib), BEXTRA® (valdecoxib), BMS347070, CELEBREX® (celecoxib), COX-189 (lumiracoxib), CT-3, DERAMAXX® (deracoxib), JTE-522, 4-methyl-2-(3,4-dimethylphenyl)-1-(4-sulfamoylphenyl-1H-pyrrole), MK-663 (etoricoxib), NS-398, parecoxib, RS-57067, SC-58125, SD-8381, SVT-2016, S-2474, T-614, VIOXX® (rofecoxib) and the like.
  • EGFR inhibitors include ABX-EGF, anti-EGFR immunoliposomes, EGF-vaccine, EMD-7200, ERBITUX® (cetuximab), HR3, IgA antibodies, IRESSA® (gefitinib), TARCEVA® (erlotinib or OSI-774), TP-38, EGFR fusion protein, TYKERB® (lapatinib) and the like.
  • ErbB2 receptor inhibitors include CP-724-714, CI-1033 (canertinib), HERCEPTIN® (trastuzumab), TYKERB® (lapatinib), OMNITARG® (2C4, petuzumab), TAK-165, GW-572016 (ionafarnib), GW-282974, EKB-569, PI-166, dHER2 (HER2 vaccine), APC-8024 (HER-2 vaccine), anti-HER/2neu bispecific antibody, B7.her2IgG3, AS HER2 trifunctional bispecfic antibodies, mAB AR-209, mAB 2B-1 and the like.
  • Histone deacetylase inhibitors include depsipeptide, LAQ-824, MS-275, trapoxin, suberoylanilide hydroxamic acid (SAHA), TSA, valproic acid and the like.
  • HSP-90 inhibitors include 17-AAG-nab, 17-AAG, CNF-101, CNF-1010, CNF-2024, 17-DMAG, geldanamycin, IPI-504, KOS-953, MYCOGRAB® (human recombinant antibody to HSP-90), NCS-683664, PU24FC1, PU-3, radicicol, SNX-2112, STA-9090 VER49009 and the like.
  • Inhibitors of apoptosis proteins include ApoMab (a fully human affinity-matured IgG1 monoclonal antibody), antibodies that target TRAIL or death receptors (e.g., pro-apoptotic receptor agonists DR4 and DR5), conatumumab, ETR2-ST01, GDC0145, (lexatumumab), HGS-1029, LBY-135, PRO-1762 and tratuzumab.
  • ApoMab a fully human affinity-matured IgG1 monoclonal antibody
  • antibodies that target TRAIL or death receptors e.g., pro-apoptotic receptor agonists DR4 and DR5
  • conatumumab e.g., ETR2-ST01, GDC0145, (lexatumumab), HGS-1029, LBY-135, PRO-1762 and tratuzumab.
  • MEK inhibitors include ARRY-142886, ARRY-438162 PD-325901, PD-98059 and the like.
  • mTOR inhibitors include AP-23573, CCI-779, everolimus, RAD-001, rapamycin, temsirolimus and the like.
  • Non-steroidal anti-inflammatory drugs include AMIGESIC® (salsalate), DOLOBID® (diflunisal), MOTRIN® (ibuprofen), ORUDIS® (ketoprofen), RELAFEN® (nabumetone), FELDENE® (piroxicam), ibuprofen cream, ALEVE® (naproxen) and NAPROSYN® (naproxen), VOLTAREN® (diclofenac), INDOCIN® (indomethacin), CLINORIL® (sulindac), TOLECTIN® (tolmetin), LODINE® (etodolac), TORADOL® (ketorolac), DAYPRO® (oxaprozin) and the like.
  • PDGFR inhibitors include C-451, CP-673, CP-868596 and the like.
  • Platinum chemotherapeutics include cisplatin, ELOXATIN® (oxaliplatin) eptaplatin, lobaplatin, nedaplatin, PARAPLATIN® (carboplatin), satraplatin and the like.
  • Polo-like kinase inhibitors include BI-2536 and the like.
  • Thrombospondin analogs include ABT-510, ABT-567, TSP-1 and the like.
  • VEGFR inhibitors include AVASTIN® (bevacizumab), ABT-869, AEE-788, ANGIOZYMETM (a ribozyme that inhibits angiogenesis (Ribozyme Pharmaceuticals (Boulder, Colo.) and Chiron, (Emeryville, Calif.)), axitinib (AG-13736), AZD-2171, CP-547,632, IM-862, MACUGEN (pegaptamib), NEXAVAR® (sorafenib, BAY43-9006), pazopanib (GW-786034), vatalanib (PTK-787, ZK-222584), SUTENT® (sunitinib, SU-11248), VEGF trap, ZACTIMATM (vandetanib, ZD-6474) and the like.
  • Antibiotics include intercalating antibiotics aclarubicin, actinomycin D, amrubicin, annamycin, adriamycin, BLENOXANE® (bleomycin), daunorubicin, CAELYX® or MYOCET® (liposomal doxorubicin), elsamitrucin, epirbucin, glarbuicin, ZAVEDOS® (idarubicin), mitomycin C, nemorubicin, neocarzinostatin, peplomycin, pirarubicin, rebeccamycin, stimalamer, streptozocin, VALSTAR® (valrubicin), zinostatin and the like.
  • Topoisomerase inhibitors include aclarubicin, 9-aminocamptothecin, amonafide, amsacrine, becatecarin, belotecan, BN-80915, CAMPTOSAR® (irinotecan hydrochloride), camptothecin, CARDIOXANE® (dexrazoxine), diflomotecan, edotecarin, ELLENCE® or PHARMORUBICIN® (epirubicin), etoposide, exatecan, 10-hydroxycamptothecin, gimatecan, lurtotecan, mitoxantrone, orathecin, pirarbucin, pixantrone, rubitecan, sobuzoxane, SN-38, tafluposide, topotecan and the like.
  • Antibodies include AVASTIN® (bevacizumab), CD40-specific antibodies, chTNT-1/B, denosumab, ERBITUX® (cetuximab), HUMAX-CD4® (zanolimumab), IGF1R-specific antibodies, lintuzumab, PANOREX® (edrecolomab), RENCAREX® (WX G250), RITUXAN® (rituximab), ticilimumab, trastuzimab and the like.
  • Hormonal therapies include ARIMIDEX® (anastrozole), AROMASIN® (exemestane), arzoxifene, CASODEX® (bicalutamide), CETROTIDE® (cetrorelix), degarelix, deslorelin, DESOPAN® (trilostane), dexamethasone, DROGENIL®, (flutamide), EVISTA® (raloxifene), AFEMATM (fadrozole), FARESTON® (toremifene), FASLODEX® (fulvestrant), FEMARA® (letrozole), formestane, glucocorticoids, HECTOROL® (doxercalciferol), RENAGEL® (sevelamer carbonate), lasofoxifene, leuprolide acetate, MEGACE® (megesterol), MIFEPREX® (mifepristone), NILANDRONTM (nilutamide), NOLVADEX® (tamoxifen
  • Deltoids and retinoids include seocalcitol (EB1089, CB1093), lexacalcitrol (KH1060), fenretinide, PANRETIN® (aliretinoin), ATRAGEN® (liposomal tretinoin), TARGRETIN® (bexarotene), LGD-1550 and the like.
  • PARP inhibitors include ABT-888, olaparib, KU-59436, AZD-2281, AG-014699, BSI-201, BGP-15, INO-1001, ONO-2231 and the like.
  • Plant alkaloids include, but are not limited to, vincristine, vinblastine, vindesine, vinorelbine and the like.
  • Proteasome inhibitors include VELCADE® (bortezomib), MG132, NPI-0052, PR-171 and the like.
  • immunologicals include interferons and other immune-enhancing agents.
  • Interferons include interferon alpha, interferon alpha-2a, interferon alpha-2b, interferon beta, interferon gamma-1a, ACTIMMUNE® (interferon gamma-1b), or interferon gamma-n1, combinations thereof and the like.
  • agents include ALFAFERONE®, (IFN- ⁇ ), BAM-002 (oxidized glutathione), BEROMUN® (tasonermin), BEXXAR® (tositumomab), CAMPATH® (alemtuzumab), CTLA4 (cytotoxic lymphocyte antigen 4), decarbazine, denileukin, epratuzumab, GRANOCYTE® (lenograstim), lentinan, leukocyte alpha interferon, imiquimod, MDX-010 (anti-CTLA-4), melanoma vaccine, mitumomab, molgramostim, MYLOTARGTM (gemtuzumab ozogamicin), NEUPOGEN® (filgrastim), OncoVAC-CL, OVAREX® (oregovomab), pemtumomab (Y-muHMFG1), PROVENGE® (sipuleucel-T), sargaramostim, sizo
  • Biological response modifiers are agents that modify defense mechanisms of living organisms or biological responses, such as survival, growth, or differentiation of tissue cells to direct them to have anti-tumor activity and include include krestin, lentinan, sizofuran, picibanil PF-3512676 (CpG-8954), ubenimex and the like.
  • Pyrimidine analogs include cytarabine (ara C or Arabinoside C), cytosine arabinoside, doxifluridine, FLUDARA® (fludarabine), 5-FU (5-fluorouracil), floxuridine, GEMZAR® (gemcitabine), TOMUDEX® (ratitrexed), TROXATYLTM (triacetyluridine troxacitabine) and the like.
  • Purine analogs include LANVIS® (thioguanine) and PURI-NETHOL® (mercaptopurine).
  • Antimitotic agents include batabulin, epothilone D (KOS-862), N-(2-((4-hydroxyphenyl)amino)pyridin-3-yl)-4-methoxybenzenesulfonamide, ixabepilone (BMS 247550), paclitaxel, TAXOTERE® (docetaxel), PNU100940 (109881), patupilone, XRP-9881 (larotaxel), vinflunine, ZK-EPO (synthetic epothilone) and the like.
  • Radiotherapy examples include external beam radiotherapy, teletherapy, brachtherapy and sealed, unsealed source radiotherapy and the like.
  • compounds having Formula I may be combined with other chemptherapeutic agents such as ABRAXANETM (ABI-007), ABT-100 (farnesyl transferase inhibitor), ADVEXIN® (Ad5CMV-p53 vaccine), ALTOCOR® or MEVACOR® (lovastatin), AMPLIGEN® (poly I:poly C12U, a synthetic RNA), APTOSYN® (exisulind), AREDIA® (pamidronic acid), arglabin, L-asparaginase, atamestane (1-methyl-3,17-dione-androsta-1,4-diene), AVAGE® (tazarotene), AVE-8062 (combreastatin derivative) BEC2 (mitumomab), cachectin or cachexin (tumor necrosis factor), canvaxin (vaccine), CEAVAC® (cancer vaccine), CELEUK® (celmoleukin), CEPLENE® (histamine dihydroch
  • Active Aurora B enzyme (recombinant residues 1-344) and INCENP (recombinant GST fusion protein (Upstate)) were incubated in wells of a 384 well plate with biotinylted histone H3 peptide residues 1-21 (Upstate), 1 mM ATP, and various concentrations of inhibitors in a HEPES buffer, pH 7.4 containing MgCl 2 , sodium othrovanadate, and Triton X-100. After 1 hour, the reaction was stopped with EDTA and anti-phospho-histone H3 Europium Cryptate (Cis-Bio) and SA-APC (Phycolink, Prozyme) were added to detect the phosphopeptide.
  • Cis-Bio anti-phospho-histone H3 Europium Cryptate
  • SA-APC Physicallink, Prozyme
  • the amount of phosphorylation was determined by the time-resolved fluorescence ratio of signals at 665 nm and 615 nm.
  • the IC 50 's were calculated by an exponential fit of the inhibition values with the inhibitor concentrations using Assay Explorer software.
  • HTRF time-resolved fluorescence
  • KDR For example for KDR, cKIT, FLT1, CSF1R and FTL3, purified enzyme was mixed with 0.5 ⁇ M N-biotinylated substrate (Biotin-Ahx-AEEEYFFLA-amide (SEQ. ID. 1)), various concentrations of inhibitor in reaction buffer (50 mM HEPES, pH 7.1, 10 mM MgCl 2 , 2 mM MnCl 2 , 0.1% BSA and 1 mM DTT, 40 ⁇ L final volume), ATP (1 mM final conc.) in a black 384-well plate.
  • reaction buffer 50 mM HEPES, pH 7.1, 10 mM MgCl 2 , 2 mM MnCl 2 , 0.1% BSA and 1 mM DTT, 40 ⁇ L final volume
  • ATP 1 mM final conc.
  • reaction was quenched by addition of a buffered EDTA solution (final approximate concentrations: 30 mM EDTA, 0.1% BSA, 0.1% Triton X-100 and 0.24M KF) and a solution of revelation agents (to give 0.084 ng/well streptavidin-XL-665 (Cis-Bio) and 6.5 ng/well antiphsophotyrosine mAb PT66-K Europium kryptate) was added to the reaction mixture.
  • a buffered EDTA solution final approximate concentrations: 30 mM EDTA, 0.1% BSA, 0.1% Triton X-100 and 0.24M KF
  • revelation agents to give 0.084 ng/well streptavidin-XL-665 (Cis-Bio) and 6.5 ng/well antiphsophotyrosine mAb PT66-K Europium kryptate
  • the quenched reaction was allowed to stand at room temperature for 3 hours and was then read in a time-resolved fluorescence detector (InVision, Perkin-Elmer) at 620 nm and 665 nm sequentially with excitation. The ratio between the signal of 620 nm and 665 nm was used in the calculation of the IC 50 .
  • Table 2 and Table 3 demonstrate the utility of Examples 1-6 as inhibitors of multiple kinases.
  • Assays (200 ⁇ L final volume) were carried out in NUNC polypropylene deep well plates in 50 mM potassium phosphate buffer, pH 7.4, using a microtiter plate shaker in a 37° C. incubator. Pooled human liver microsomes (BD Gentest, 50 ⁇ g/mL) were incubated with 5 concentrations of test compound (from 0.1 ⁇ M to 10 ⁇ M), 1 mM NADPH (Sigma), and 2 ⁇ M midazolam (Sigma). A constant amount of dimethylsulfoxide (1%) was added to the incubations with the test compounds, and each analysis was performed in duplicate.
  • BD Gentest 50 ⁇ g/mL
  • Pre preincubation experiments
  • Co coincubation experiments
  • the compounds, microsomes, and midazolam were mixed and the reaction initiated by addition of NADPH to the wells.
  • the complete reaction was terminated after 10 minutes of shaking by addition of 100 ⁇ L of a 1/1 mixture of acetonitrile and methanol containing 0.33 ⁇ M 1-hydroxytriazolam.
  • the ratio of 1-hydroxymidazolam (CYP 3A4 product) and the internal standard at each concentration of compound was used to calculate the % inhibition of activity relative to the ratio calculated for control incubations without inhibitor. In the absence of NADPH, no hydroxylation of midazolam was observed.
  • Ketoconazole was used as a standard inhibitor, which at 0.1 ⁇ M produces 70-80% inhibition of CYP 3A4.
  • the IC 50 (the concentration at which 50% of the enzyme is inhibited) was calculated for Examples 1-5 and Example 6 (described as EXAMPLE 703 in WO 2005/10009) and is shown in Table 4 below.
  • Compounds described as having low CYP inhibition or as not inhibiting CYP are those compounds with an IC 50 of >10 ⁇ M in the above assay.
  • the compound of this invention inhibits the activity of Aurora-kinase B, it could also have utility as an inhibitor of protein kinases having close structural homology thereto, such as, for example, Aurora-kinase A and Aurora-kinase C.
  • Example 1 This data demonstrates the utility of Example 1 as an inhibitor of Aurora-kinase A and Aurora-kinase B and Aurora-kinase C.
  • compounds of this invention are expected to have utility in treatment of diseases during which protein kinases such as any or all Aurora-kinase family members are expressed.
  • Aurora-A/STK-15 is a Predictive Factor for Recurrent Behaviour in Non-Invasive Bladder Carcinoma: A Study Of 128 Cases of Non-Invasive Neoplasms. Virchows Arch 2007; 450:419-24; Fraizer G. C., Diaz M. F., Lee I. L., Grossman H. B., Sen S. Aurora-A/STK15/BTAK Enhances Chromosomal Instability in Bladder Cancer Cells. Int. J. Oncol.
  • the Putative Serine/Threonine Kinase Gene STK15 on Chromosome 20q13.2 is Amplified In Human Gliomas. Oncol. Rep. 2003; 10:1275-9; Klein A., Reichardt W., Jung V., Zang K. D., Meese E., Urbschat S. Overexpression and Amplification of STK15 Inhuman Gliomas. Int. J. Oncol. 2004; 25:1789-94; and Mau K., Korshunov A., Benner A., et al. Microarray Based Screening for Molecular Markers Nmedulloblastoma Revealed STK15 as Independent Predictor for Survival. Cancer Res 2004; 64:3103-11.
  • Zhao X. Li F. C., Li Y. H., et al. [Mutation of p53 and Overexpression Of STK15 in Laryngeal Squamous-Cell Carcinoma].
  • Protecting groups for C(O)OH moieties include, but are not limited to, acetoxymethyl, allyl, benzoylmethyl, benzyl, benzyloxymethyl, tert-butyl, tert-butyldiphenylsilyl, diphenylmethyl, cyclobutyl, cyclohexyl, cyclopentyl, cyclopropyl, diphenylmethylsilyl, ethyl, para-methoxybenzyl, methoxymethyl, methoxyethoxymethyl, methyl, methylthiomethyl, naphthyl, para-nitrobenzyl, phenyl, n-propyl, 2,2,2-trichloroethyl, triethylsilyl, 2-(trimethylsilyl)ethyl, 2-(trimethylsilyl)ethoxymethyl, triphenylmethyl and the like.
  • Protecting groups for C(O) and C(O)H moieties include, but are not limited to, 1,3-dioxylketal, diethylketal, dimethylketal, 1,3-dithianylketal, O-methyloxime, O-phenyloxime and the like.
  • Protecting groups for NH moieties include, but are not limited to, acetyl, alanyl, benzoyl, benzyl (phenylmethyl), benzylidene, benzyloxycarbonyl (Cbz), tert-butoxycarbonyl (Boc), 3,4-dimethoxybenzyloxycarbonyl, diphenylmethyl, diphenylphosphoryl, formyl, methanesulfonyl, para-methoxybenzyloxycarbonyl, phenylacetyl, phthaloyl, succinyl, trichloroethoxycarbonyl, triethylsilyl, trifluoroacetyl, trimethylsilyl, triphenylmethyl, triphenylsilyl, para-toluenesulfonyl and the like.
  • Protecting groups for OH and SH moieties include, but are not limited to, acetyl, allyl, allyloxycarbonyl, benzyloxycarbonyl (Cbz), benzoyl, benzyl, tert-butyl, tert-butyldimethylsilyl, tert-butyldiphenylsilyl, 3,4-dimethoxybenzyl, 3,4-dimethoxybenzyloxycarbonyl, 1,1-dimethyl-2-propenyl, diphenylmethyl, formyl, methanesulfonyl, methoxyacetyl, 4-methoxybenzyloxycarbonyl, para-methoxybenzyl, methoxycarbonyl, methyl, para-toluenesulfonyl, 2,2,2-trichloroethoxycarbonyl, 2,2,2-trichloroethyl, triethylsilyl, trifluoroacetyl, 2-
  • ADDP means 1,1′-(azodicarbonyl)dipiperidine; AD-mix- ⁇ means a mixture of (DHQD) 2 PHAL, K 3 Fe(CN) 6 , K 2 CO 3 and K 2 SO 4 ); AIBN means 2,2′-azobis(2-methylpropionitrile); 9-BBN means 9-borabicyclo(3.3.1)nonane; Cp means cyclopentadiene; (DHQD) 2 PHAL means hydroquinidine 1,4-phthalazinediyl diethyl ether; DBU means 1,8-diazabicyclo(5.4.0)undec-7-ene; DIBAL means diisobutylaluminum hydride; DIEA means diisopropylethylamine; DMAP means N,N-dimethylaminopyridine; DME means 1,2-dimethoxyethane; DMF means N,N-dimethylformamide; dmpe means 1,2-bis(dimethylphosphino
  • reaction mixture was cooled to room temperature, then partitioned between ethyl acetate and water.
  • the aqueous layer was extracted with additional ethyl acetate and the combined organics were dried (using MgSO 4 ), filtered and the filtrate was concentrated.
  • the residue was purified via silica gel chromatography eluting with 50 to 70% ethyl acetate-hexanes to give crude tert-butyl 4-(4-aminothieno[3,2-c]pyridin-3-yl)phenylcarbamate.
  • the aqueous layer was extracted with additional ethyl acetate (2 ⁇ 75 mL) and the combined organics were washed with sodium thiosulfate (3 ⁇ 20 mL) and brine (50 mL), and then dried (using MgSO 4 ), filtered, and concentrated.
  • the crude material was treated with TFA (20 mL) and CH 2 Cl 2 (5 mL), stirred at room temperature for 2 hours, concentrated under a stream of nitrogen, then concentrated in vacuo.
  • the solid was dissolved in water (100 mL), carefully treated with solid Na 2 CO 3 until gas evolution ceased and filtered, washing with additional water.
  • the solid collected was dried to provide the title compound as a solid (contaminated by ca. 10% mole PPh 3 ).
  • the filtrate was concentrated, then purified by chromatography on an Analogix® IntelliflashTM purification system using a SF60-200 g column at a flow rate of 80 mL/min, eluting as follows: 5 minutes at 20% ethyl acetate/hexanes, then ramped from 40% to 90% ethyl acetate/hexanes over 35 minutes, then 100% ethyl acetate for another 20 minutes, to provide the title compound.
  • EXAMPLE 1A (6 g, 16.34 mmol), EXAMPLE 1B (4.8 g, 20.16 mmol), PdCl 2 (dppf) (1.2 g, 1.640 mmol) and sodium carbonate (4.6 g, 43.4 mmol) were combined in tetrahydrofuran (400 mL), methanol (80 mL) and water (80 mL), and the reaction was degassed by bubbling N 2 through the mixture for 1 hour. The reaction was then heated to 80° C. for 2 hours, then allowed to cool and diluted with 300 mL ethyl acetate.
  • EXAMPLE 1C (2 g, 5.69 mmol) was dissolved in N,N-dimethylformamide (80 ml) and the flask was chilled in a ⁇ 20° C. bath. 1-fluoro-3-isocyanatobenzene (0.715 ml, 6.26 mmol) was added dropwise and the reaction was allowed to warm slowly to room temperature and stirred overnight. The reaction was diluted with water (500 mL) and ethyl acetate (75 mL) and stirred to digest for 1 hour. The mixture was then placed in a separatory funnel. After the layers were allowed to separate, the lower aqueous layer was drained off. Near the interface of the two layers there was a significant amount of precipitate.
  • Example 6 is described as EXAMPLE 703 in WO 2005/10009.

Abstract

Compounds that inhibit protein kinases such as Aurora-kinases and the VEGFR and PDGFR families of kinases, with an improved safety profile due to low CYP3A4 inhibition, compositions containing the compounds and methods of treating diseases using the compounds are disclosed.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application claims priority to provisional application Ser. No. 61/120,281, filed Dec. 5, 2008, and provisional application Ser. No. 61/223,760, filed Jul. 8, 2009.
  • FIELD OF THE INVENTION
  • This invention pertains to compounds that inhibit protein kinases such as Aurora-kinases and the VEGFR and PDGFR families of kinases, and which have an improved safety profile due to low CYP3A4 inhibition, compositions containing the compounds, and methods of treating diseases using the compounds.
  • BACKGROUND OF THE INVENTION
  • Mitosis is a process by which a complete copy of a duplicated genome is segregated by the microtuble spindle apparatus into two daughter cells. Aurora-kinases, key mitotic regulators required for genome stability, have been found to be overexpressed in human tumors. There is therefore an existing need in the therapeutic arts for compounds which inhibit Aurora-kinases, compositions comprising the inhibitors and methods of treating diseases during which Aurora-kinases are unregulated or overexpressed.
  • The reversible phosphorylation of proteins is one of the primary biochemical mechanisms mediating eukaryotic cell signaling. This reaction is catalyzed by protein kinases that transfer the g-phosphate group of ATP to hydroxyl groups on target proteins. 518 such enzymes exist in the human genome of which ˜90 selectively catalyze the phosphorylation of tyrosine hydroxyl groups Cytosolic tyrosine kinases reside intracellularly whereas receptor tyrosine kinases (RTKs) possess both extracellular and intracellular domains and function as membrane spanning cell surface receptors. As such, RTKs mediate the cellular responses to environmental signals and facilitate a broad range of cellular processes including proliferation, migration and survival.
  • RTK signaling pathways are normally highly regulated, yet their over-activation has been shown to promote the growth, survival and metastasis of cancer cells. Dysregulated RTK signaling occurs through gene over-expression or mutation and has been correlated with the progression of various human cancers.
  • The VEGF receptor (VEGFR) family consists of three RTKs, KDR (kinase insert domain-containing receptor; VEGFR2), FLT1 (Fms-like tyrosine kinase; VEGFR1), and FLT4 (VEGFR3). These receptors mediate the biological function of the vascular endothelial growth factors (VEGF-A, -B, -C, -D, -E and placenta growth factor (P1GF)), a family of homodimeric glycoproteins that bind the VEGF receptors with varying affinities.
  • KDR is the major mediator of the mitogenic, angiogenic and permeability-enhancing effects of VEGF-A, hereafter referred to as VEGF. Many different cell types are able to produce VEGF, yet its biological activity is limited predominately to the vasculature by way of the endothelial cell-selective expression of KDR. Not surprisingly, the VEGF/KDR axis is a primary mediator of angiogenesis, the means by which new blood vessels are formed from preexisting vessels.
  • FLT1 binds VEGF, VEGF-B and placental growth factor. FLT1 is expressed on the surface of smooth muscle cells, monocytes and hematopoietic stems cells in addition to endothelial cells. Activation of FLT1 signaling results in the mobilization of marrow-derived endothelial progenitor cells that are recruited to tumors where they contribute to new blood vessel formation.
  • FLT4 mediates the signaling of VEGF-C and VEGF-D, which mediate formation of tumor-associated lymphatic vessels (lymphangiogenesis). Lymphatic vessels are one of the routes by which cancer cells disseminate from solid tumors during metastasis.
  • The PDGF receptor (PDGFR) family consists of five RTK's, PDGFR-a and b, CSF1R, KIT, and FLT3.
  • The a and b isoforms of the platelet-derived growth factor (PDGF) receptors occur as homodimers or a/b heterodimers and are found most commonly on the surface of fibroblasts and smooth muscle cells. PDGFR-b contributes to tumor angiogenesis through the proliferation and migration of pericytes, the peri-endothelial cells that associate with and stabilize immature blood vessels. In gliomas, autocrine PDGFR stimulation, brought about by the co-expression of PDGF and PDGF receptors, mediates tumor cell proliferation and survival.
  • CSF-1R is encoded by the cellular homolog of the retroviral oncogene v-fms and is a major regulator of macrophage development. Macrophages are frequent components of tumor stroma and have been shown to modify the extracellular matrix in a manner beneficial to tumor growth and metastasis.
  • KIT is expressed by hematopoietic progenitor cells, mast cells, germ cells and by pacemaker cells in the gut (interstitial cells of Cajal). It contributes to tumor progression by two general mechanisms namely autocrine stimulation by its ligand, stem cell factor (SCF), and through mutations that result in ligand-independent kinase activity.
  • FLT3 is normally expressed on hematopoietic stem cells where its interaction with FLT3 ligand (FL) stimulates stem cell survival, proliferation and differentiation. In addition to being over-expressed in various leukemia cells, FLT3 is frequently mutated in hematological malignancies with approximately one-third of patients with acute myeloid leukemia (AML) harboring activating mutations.
  • The identification of effective small compounds which specifically inhibit signal transduction and cellular proliferation by modulating the activity of tyrosine kinases to regulate and modulate abnormal or inappropriate cell proliferation, differentiation, or metabolism is therefore desirable. In particular, 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.
  • Administered drugs are eliminated from the body either by excretion or by metabolism to one or more metabolites. One class of metabolizing enzymes that is particularly important in the metabolism of drugs is the cytochrome P450 (also known as CYP or P450) family of enzymes. This is a large family of isoenzymes which has been divided into over 15 subfamilies. The CYP3A subfamily, which includes CYP3A4, 3A5, 3A7 and 3A43 is responsible for the metabolism of about 60% of known therapeutic drugs. CYP3A4 in particular is the most abundant CYP isoenzyme in both liver and intestine and is involved in the metabolism of more than 50% of the clinically used drugs (Mechanism-Based Inhibition of Cytochrome P455 3A4 by Therapeutic Drugs. Clin. Pharmacokinet, 2005, 44, 279-304). Like all other CYP enzymes, CYP3A4 is susceptible to both reversible and pseudo-irreversible or irreversible (mechanism based) inhibition (Time-dependent CYP Inhibition. Expert Opin. Drug Metab. Toxicol. 2007, 3, 51-66). Their low substrate specificity makes the CYP enzymes susceptible to inhibition by a wide variety of structurally distinct drugs.
  • As a result of CYP inhibition, abrupt changes can occur with a co-administered agent in a single individual leading to a substantial increase or decrease in the blood and tissue concentrations of a drug or metabolite. These types of changes can alter a drug's safety and efficacy profile in profound ways, especially drugs with narrow therapeutic windows. As outlined in the FDA guidance to industry, a detailed evaluation of the CYP inhibition potential is required of all new drug candidates (Guidance for Industry. Drug Metabolism/Drug Interaction Studies in the Drug Develeopment Process: Studies in Vitro. US FDA April 1997).
  • This issue of drug-drug interaction is very important in oncology treatment as patients are typically treated with multiple drugs. Thus, reducing the risk of such interaction is an important consideration in the development of novel cancer therapeutics.
  • While thienopyridine compounds disclosed in WO2005/010009 display potent inhibition of Aurora and PDGFR/VEGFR kinases, they may also be inhibitors of CYP3A4. This invention pertains to novel thienopyridines of formula I, which maintain potent inhibition of both Aurora kinases and the family of kinases encompassing PDGFR and VEGFR and also demonstrate at least a 10-30 fold reduction in CYP3A4 inhibition. Because the compounds of the present invention have significantly reduced CYP3A4 inhibition, they are expected to display low drug-drug interaction liability.
  • In addition to the reduction in CYP inhibition, the compounds of the invention have demonstrated their utility in additional assays utilized to assess the suitability of the compounds as drug candidates. For instance, the compounds of the invention demonstrate potency in whole cell assays (e.g., in the Human Umbilical Vein Endothelial Cell (HUVEC) assay and the assay measuring histone D3 phosphorylation and induction of polyploidy) and suitable pharmokinetic properties (e.g., oral clearance and oral bioavailability), in vivo efficacy (e.g., Uterine Edema model, rodent flank and orthotopic tumor models), cardiovascular safety, CNS assessments and gastroinstestinal assays.
  • SUMMARY OF THE INVENTION
  • In one embodiment, the present invention provides compounds of formula (I)
  • Figure US20100144783A1-20100610-C00001
  • wherein
  • R1 is hydroxyalkyl;
  • R2 is selected from the group consisting of alkoxy, alkyl, halo, and haloalkoxy; and
  • R3 is hydrogen or alkyl.
  • Still another embodiment pertains to methods of treating diseases involving mediation, overexpression or disregulation of kinases in a mammal, the methods comprising administering thereto a therapeutically effective amount of a compound having Formula I,
  • Figure US20100144783A1-20100610-C00002
  • wherein
  • R1 is hydroxyalkyl;
  • R2 is selected from the group consisting of alkoxy, alkyl, halo, and haloalkoxy; and
  • R3 is hydrogen or alkyl.
  • Another embodiment pertains to methods of treating acoustic neuroma, acute leukemia, acute lymphocytic leukemia, acute myelocytic leukemia (monocytic, myeloblastic, adenocarcinoma, angiosarcoma, astrocytoma, myelomonocytic and promyelocytic), acute t-cell leukemia, basal cell carcinoma, bile duct carcinoma, bladder cancer, brain cancer, breast cancer, bronchogenic carcinoma, cervical cancer, chondrosarcoma, chordoma, choriocarcinoma, chronic leukemia, chronic lymphocytic leukemia, chronic myelocytic (granulocytic) leukemia, chronic myleogeneous leukemia, colon cancer, colorectal cancer, craniopharyngioma, cystadenocarcinoma, diffuse large B-cell lymphoma, dysproliferative changes (dysplasias and metaplasias), embryonal carcinoma, endometrial cancer, endotheliosarcoma, ependymoma, epithelial carcinoma, erythroleukemia, esophageal cancer, estrogen-receptor positive breast cancer, essential thrombocythemia, Ewing's tumor, fibrosarcoma, follicular lymphoma, germ cell testicular cancer, glioma, heavy chain disease, hemangioblastoma, hepatoma, hepatocellular cancer, hormone insensitive prostate cancer, leiomyosarcoma, liposarcoma, lung cancer, lymphagioendotheliosarcoma, lymphangiosarcoma, lymphoblastic leukemia, lymphoma (Hodgkin's and non-Hodgkin's), malignancies and hyperproliferative disorders of the bladder, breast, colon, lung, ovaries, pancreas, prostate, skin and uterus, lymphoid malignancies of T-cell or B-cell origin, leukemia, lymphoma, medullary carcinoma, medulloblastoma, melanoma, meningioma, mesothelioma, multiple myeloma, myelogenous leukemia, myeloma, myxosarcoma, neuroblastoma, non-small cell lung cancer, oligodendroglioma, oral cancer, osteogenic sarcoma, ovarian cancer, pancreatic cancer, papillary adenocarcinomas, papillary carcinoma, pinealoma, polycythemia vera, prostate cancer, rectal cancer, renal cell carcinoma, retinoblastoma, rhabdomyosarcoma, sarcoma, sebaceous gland carcinoma, seminoma, skin cancer, small cell lung carcinoma, solid tumors (carcinomas and sarcomas), small cell lung cancer, stomach cancer, squamous cell carcinoma, synovioma, sweat gland carcinoma, thyroid cancer, Waldenström's macroglobulinemia, testicular tumors, uterine cancer and Wilms' tumor in a mammal, the methods comprising administering thereto a therapeutically effective amount of a compound of formula I, alone or in combination with radiotherapy.
  • Another embodiment pertains to methods of treating acoustic neuroma, acute leukemia, acute lymphocytic leukemia, acute myelocytic leukemia (monocytic, myeloblastic, adenocarcinoma, angiosarcoma, astrocytoma, myelomonocytic and promyelocytic), acute t-cell leukemia, basal cell carcinoma, bile duct carcinoma, bladder cancer, brain cancer, breast cancer, bronchogenic carcinoma, cervical cancer, chondrosarcoma, chordoma, choriocarcinoma, chronic leukemia, chronic lymphocytic leukemia, chronic myelocytic (granulocytic) leukemia, chronic myleogeneous leukemia, colon cancer, colorectal cancer, craniopharyngioma, cystadenocarcinoma, diffuse large B-cell lymphoma, dysproliferative changes (dysplasias and metaplasias), embryonal carcinoma, endometrial cancer, endotheliosarcoma, ependymoma, epithelial carcinoma, erythroleukemia, esophageal cancer, estrogen-receptor positive breast cancer, essential thrombocythemia, Ewing's tumor, fibrosarcoma, follicular lymphoma, germ cell testicular cancer, glioma, heavy chain disease, hemangioblastoma, hepatoma, hepatocellular cancer, hormone insensitive prostate cancer, leiomyosarcoma, liposarcoma, lung cancer, lymphagioendotheliosarcoma, lymphangiosarcoma, lymphoblastic leukemia, lymphoma (Hodgkin's and non-Hodgkin's), malignancies and hyperproliferative disorders of the bladder, breast, colon, lung, ovaries, pancreas, prostate, skin and uterus, lymphoid malignancies of T-cell or B-cell origin, leukemia, lymphoma, medullary carcinoma, medulloblastoma, melanoma, meningioma, mesothelioma, multiple myeloma, myelogenous leukemia, myeloma, myxosarcoma, neuroblastoma, non-small cell lung cancer, oligodendroglioma, oral cancer, osteogenic sarcoma, ovarian cancer, pancreatic cancer, papillary adenocarcinomas, papillary carcinoma, pinealoma, polycythemia vera, prostate cancer, rectal cancer, renal cell carcinoma, retinoblastoma, rhabdomyosarcoma, sarcoma, sebaceous gland carcinoma, seminoma, skin cancer, small cell lung carcinoma, solid tumors (carcinomas and sarcomas), small cell lung cancer, stomach cancer, squamous cell carcinoma, synovioma, sweat gland carcinoma, thyroid cancer, Waldenström's macroglobulinemia, testicular tumors, uterine cancer and Wilms' tumor in a mammal, the methods comprising administering thereto a therapeutically effective amount of N-(4-{4-amino-7-[1-(2-hydroxyethyl)-1H-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N′-(3-fluorophenyl)urea, alone or in combination with radiotherapy.
  • Another embodiment pertains to methods of treating acoustic neuroma, acute leukemia, acute lymphocytic leukemia, acute myelocytic leukemia (monocytic, myeloblastic, adenocarcinoma, angiosarcoma, astrocytoma, myelomonocytic and promyelocytic), acute t-cell leukemia, basal cell carcinoma, bile duct carcinoma, bladder cancer, brain cancer, breast cancer, bronchogenic carcinoma, cervical cancer, chondrosarcoma, chordoma, choriocarcinoma, chronic leukemia, chronic lymphocytic leukemia, chronic myelocytic (granulocytic) leukemia, chronic myleogeneous leukemia, colon cancer, colorectal cancer, craniopharyngioma, cystadenocarcinoma, diffuse large B-cell lymphoma, dysproliferative changes (dysplasias and metaplasias), embryonal carcinoma, endometrial cancer, endotheliosarcoma, ependymoma, epithelial carcinoma, erythroleukemia, esophageal cancer, estrogen-receptor positive breast cancer, essential thrombocythemia, Ewing's tumor, fibrosarcoma, follicular lymphoma, germ cell testicular cancer, glioma, heavy chain disease, hemangioblastoma, hepatoma, hepatocellular cancer, hormone insensitive prostate cancer, leiomyosarcoma, liposarcoma, lung cancer, lymphagioendotheliosarcoma, lymphangiosarcoma, lymphoblastic leukemia, lymphoma (Hodgkin's and non-Hodgkin's), malignancies and hyperproliferative disorders of the bladder, breast, colon, lung, ovaries, pancreas, prostate, skin and uterus, lymphoid malignancies of T-cell or B-cell origin, leukemia, lymphoma, medullary carcinoma, medulloblastoma, melanoma, meningioma, mesothelioma, multiple myeloma, myelogenous leukemia, myeloma, myxosarcoma, neuroblastoma, non-small cell lung cancer, oligodendroglioma, oral cancer, osteogenic sarcoma, ovarian cancer, pancreatic cancer, papillary adenocarcinomas, papillary carcinoma, pinealoma, polycythemia vera, prostate cancer, rectal cancer, renal cell carcinoma, retinoblastoma, rhabdomyosarcoma, sarcoma, sebaceous gland carcinoma, seminoma, skin cancer, small cell lung carcinoma, solid tumors (carcinomas and sarcomas), small cell lung cancer, stomach cancer, squamous cell carcinoma, synovioma, sweat gland carcinoma, thyroid cancer, Waldenström's macroglobulinemia, testicular tumors, uterine cancer and Wilms' tumor in a mammal, the methods comprising administering thereto a therapeutically effective amount of N-(4-{4-amino-7-[1-(2-hydroxyethyl)-1H-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N′-[4-(difluoromethoxy)phenyl]urea, alone or in combination with radiotherapy.
  • Another embodiment pertains to methods of treating acoustic neuroma, acute leukemia, acute lymphocytic leukemia, acute myelocytic leukemia (monocytic, myeloblastic, adenocarcinoma, angiosarcoma, astrocytoma, myelomonocytic and promyelocytic), acute t-cell leukemia, basal cell carcinoma, bile duct carcinoma, bladder cancer, brain cancer, breast cancer, bronchogenic carcinoma, cervical cancer, chondrosarcoma, chordoma, choriocarcinoma, chronic leukemia, chronic lymphocytic leukemia, chronic myelocytic (granulocytic) leukemia, chronic myleogeneous leukemia, colon cancer, colorectal cancer, craniopharyngioma, cystadenocarcinoma, diffuse large B-cell lymphoma, dysproliferative changes (dysplasias and metaplasias), embryonal carcinoma, endometrial cancer, endotheliosarcoma, ependymoma, epithelial carcinoma, erythroleukemia, esophageal cancer, estrogen-receptor positive breast cancer, essential thrombocythemia, Ewing's tumor, fibrosarcoma, follicular lymphoma, germ cell testicular cancer, glioma, heavy chain disease, hemangioblastoma, hepatoma, hepatocellular cancer, hormone insensitive prostate cancer, leiomyosarcoma, liposarcoma, lung cancer, lymphagioendotheliosarcoma, lymphangiosarcoma, lymphoblastic leukemia, lymphoma (Hodgkin's and non-Hodgkin's), malignancies and hyperproliferative disorders of the bladder, breast, colon, lung, ovaries, pancreas, prostate, skin and uterus, lymphoid malignancies of T-cell or B-cell origin, leukemia, lymphoma, medullary carcinoma, medulloblastoma, melanoma, meningioma, mesothelioma, multiple myeloma, myelogenous leukemia, myeloma, myxosarcoma, neuroblastoma, non-small cell lung cancer, oligodendroglioma, oral cancer, osteogenic sarcoma, ovarian cancer, pancreatic cancer, papillary adenocarcinomas, papillary carcinoma, pinealoma, polycythemia vera, prostate cancer, rectal cancer, renal cell carcinoma, retinoblastoma, rhabdomyosarcoma, sarcoma, sebaceous gland carcinoma, seminoma, skin cancer, small cell lung carcinoma, solid tumors (carcinomas and sarcomas), small cell lung cancer, stomach cancer, squamous cell carcinoma, synovioma, sweat gland carcinoma, thyroid cancer, Waldenström's macroglobulinemia, testicular tumors, uterine cancer and Wilms' tumor in a mammal, the methods comprising administering thereto a therapeutically effective amount of N-[4-(4-amino-7-{1-[(2S)-2-hydroxypropyl]-1H-pyrazol-4-yl}thieno[3,2-c]pyridin-3-yl)phenyl]-N′-(3-methylphenyl)urea, alone or in combination with radiotherapy.
  • Another embodiment pertains to methods of treating acoustic neuroma, acute leukemia, acute lymphocytic leukemia, acute myelocytic leukemia (monocytic, myeloblastic, adenocarcinoma, angiosarcoma, astrocytoma, myelomonocytic and promyelocytic), acute t-cell leukemia, basal cell carcinoma, bile duct carcinoma, bladder cancer, brain cancer, breast cancer, bronchogenic carcinoma, cervical cancer, chondrosarcoma, chordoma, choriocarcinoma, chronic leukemia, chronic lymphocytic leukemia, chronic myelocytic (granulocytic) leukemia, chronic myleogeneous leukemia, colon cancer, colorectal cancer, craniopharyngioma, cystadenocarcinoma, diffuse large B-cell lymphoma, dysproliferative changes (dysplasias and metaplasias), embryonal carcinoma, endometrial cancer, endotheliosarcoma, ependymoma, epithelial carcinoma, erythroleukemia, esophageal cancer, estrogen-receptor positive breast cancer, essential thrombocythemia, Ewing's tumor, fibrosarcoma, follicular lymphoma, germ cell testicular cancer, glioma, heavy chain disease, hemangioblastoma, hepatoma, hepatocellular cancer, hormone insensitive prostate cancer, leiomyosarcoma, liposarcoma, lung cancer, lymphagioendotheliosarcoma, lymphangiosarcoma, lymphoblastic leukemia, lymphoma (Hodgkin's and non-Hodgkin's), malignancies and hyperproliferative disorders of the bladder, breast, colon, lung, ovaries, pancreas, prostate, skin and uterus, lymphoid malignancies of T-cell or B-cell origin, leukemia, lymphoma, medullary carcinoma, medulloblastoma, melanoma, meningioma, mesothelioma, multiple myeloma, myelogenous leukemia, myeloma, myxosarcoma, neuroblastoma, non-small cell lung cancer, oligodendroglioma, oral cancer, osteogenic sarcoma, ovarian cancer, pancreatic cancer, papillary adenocarcinomas, papillary carcinoma, pinealoma, polycythemia vera, prostate cancer, rectal cancer, renal cell carcinoma, retinoblastoma, rhabdomyosarcoma, sarcoma, sebaceous gland carcinoma, seminoma, skin cancer, small cell lung carcinoma, solid tumors (carcinomas and sarcomas), small cell lung cancer, stomach cancer, squamous cell carcinoma, synovioma, sweat gland carcinoma, thyroid cancer, Waldenström's macroglobulinemia, testicular tumors, uterine cancer and Wilms' tumor in a mammal, the methods comprising administering thereto a therapeutically effective amount of N-(4-{4-amino-7-[1-(2-hydroxy-2-methylpropyl)-1H-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N′-(4-methoxyphenyl)urea, alone or in combination with radiotherapy.
  • Another embodiment pertains to methods of treating acoustic neuroma, acute leukemia, acute lymphocytic leukemia, acute myelocytic leukemia (monocytic, myeloblastic, adenocarcinoma, angiosarcoma, astrocytoma, myelomonocytic and promyelocytic), acute t-cell leukemia, basal cell carcinoma, bile duct carcinoma, bladder cancer, brain cancer, breast cancer, bronchogenic carcinoma, cervical cancer, chondrosarcoma, chordoma, choriocarcinoma, chronic leukemia, chronic lymphocytic leukemia, chronic myelocytic (granulocytic) leukemia, chronic myleogeneous leukemia, colon cancer, colorectal cancer, craniopharyngioma, cystadenocarcinoma, diffuse large B-cell lymphoma, dysproliferative changes (dysplasias and metaplasias), embryonal carcinoma, endometrial cancer, endotheliosarcoma, ependymoma, epithelial carcinoma, erythroleukemia, esophageal cancer, estrogen-receptor positive breast cancer, essential thrombocythemia, Ewing's tumor, fibrosarcoma, follicular lymphoma, germ cell testicular cancer, glioma, heavy chain disease, hemangioblastoma, hepatoma, hepatocellular cancer, hormone insensitive prostate cancer, leiomyosarcoma, liposarcoma, lung cancer, lymphagioendotheliosarcoma, lymphangiosarcoma, lymphoblastic leukemia, lymphoma (Hodgkin's and non-Hodgkin's), malignancies and hyperproliferative disorders of the bladder, breast, colon, lung, ovaries, pancreas, prostate, skin and uterus, lymphoid malignancies of T-cell or B-cell origin, leukemia, lymphoma, medullary carcinoma, medulloblastoma, melanoma, meningioma, mesothelioma, multiple myeloma, myelogenous leukemia, myeloma, myxosarcoma, neuroblastoma, non-small cell lung cancer, oligodendroglioma, oral cancer, osteogenic sarcoma, ovarian cancer, pancreatic cancer, papillary adenocarcinomas, papillary carcinoma, pinealoma, polycythemia vera, prostate cancer, rectal cancer, renal cell carcinoma, retinoblastoma, rhabdomyosarcoma, sarcoma, sebaceous gland carcinoma, seminoma, skin cancer, small cell lung carcinoma, solid tumors (carcinomas and sarcomas), small cell lung cancer, stomach cancer, squamous cell carcinoma, synovioma, sweat gland carcinoma, thyroid cancer, Waldenström's macroglobulinemia, testicular tumors, uterine cancer and Wilms' tumor in a mammal, the methods comprising administering thereto a therapeutically effective amount of N-[4-(4-amino-7-{1-[(2S)-2,3-dihydroxypropyl]-1H-pyrazol-4-yl}thieno[3,2-c]pyridin-3-yl)phenyl]-N′-(4-methoxyphenyl)urea, alone or in combination with radiotherapy.
  • Another embodiment pertains to compositions comprising an excipient and a therapeutically effective amount of a compound of formula I, and a therapeutically effective amount of one additional therapeutic agent or more than one additional therapeutic agent.
  • Another embodiment pertains to compositions comprising an excipient and a therapeutically effective amount of N-(4-{4-amino-7-[1-(2-hydroxyethyl)-1H-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N′-(3-fluorophenyl)urea, and a therapeutically effective amount of one additional therapeutic agent or more than one additional therapeutic agent.
  • Another embodiment pertains to compositions comprising an excipient and a therapeutically effective amount of N-(4-{4-amino-7-[1-(2-hydroxyethyl)-1H-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N′-[4-(difluoromethoxy)phenyl]urea, and a therapeutically effective amount of one additional therapeutic agent or more than one additional therapeutic agent.
  • Another embodiment pertains to compositions comprising an excipient and a therapeutically effective amount of N-[4-(4-amino-7-{1-[(2S)-2-hydroxypropyl]-1H-pyrazol-4-yl}thieno[3,2-c]pyridin-3-yl)phenyl]-N′-(3-methylphenyl)urea, and a therapeutically effective amount of one additional therapeutic agent or more than one additional therapeutic agent.
  • Another embodiment pertains to compositions comprising an excipient and a therapeutically effective amount of N-(4-{4-amino-7-[1-(2-hydroxy-2-methylpropyl)-1H-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N′-(4-methoxyphenyl)urea, and a therapeutically effective amount of one additional therapeutic agent or more than one additional therapeutic agent.
  • Another embodiment pertains to compositions comprising an excipient and a therapeutically effective amount of a N-[4-(4-amino-7-{1-[(2S)-2,3-dihydroxypropyl]-1H-pyrazol-4-yl}thieno[3,2-c]pyridin-3-yl)phenyl]-N′-(4-methoxyphenyl)urea, and a therapeutically effective amount of one additional therapeutic agent or more than one additional therapeutic agent.
  • Still another embodiment pertains to methods of treating diseases involving mediation, overexpression or disregulation of kinases in a mammal, the methods comprising administering thereto a therapeutically effective amount of a compound of formula I, and a therapeutically effective amount of one additional therapeutic agent or more than one additional therapeutic agent alone or in combination with radiotherapy.
  • Still another embodiment pertains to methods of treating diseases involving mediation, overexpression or disregulation of kinases in a mammal, the methods comprising administering thereto a therapeutically effective amount of N-(4-{4-amino-7-[1-(2-hydroxyethyl)-1H-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N′-(3-fluorophenyl)urea, and a therapeutically effective amount of one additional therapeutic agent or more than one additional therapeutic agent alone or in combination with radiotherapy.
  • Still another embodiment pertains to methods of treating diseases involving mediation, overexpression or disregulation of kinases in a mammal, the methods comprising administering thereto a therapeutically effective amount of N-(4-{4-amino-7-[1-(2-hydroxyethyl)-1H-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N′-[4-(difluoromethoxy)phenyl]urea, and a therapeutically effective amount of one additional therapeutic agent or more than one additional therapeutic agent alone or in combination with radiotherapy.
  • Still another embodiment pertains to methods of treating diseases involving mediation, overexpression or disregulation of kinases in a mammal, the methods comprising administering thereto a therapeutically effective amount of N-[4-(4-amino-7-{1-[(2S)-2-hydroxypropyl]-1H-pyrazol-4-yl}thieno[3,2-c]pyridin-3-yl)phenyl]-N′-(3-methylphenyl)urea, and a therapeutically effective amount of one additional therapeutic agent or more than one additional therapeutic agent alone or in combination with radiotherapy.
  • Still another embodiment pertains to methods of treating diseases involving mediation, overexpression or disregulation of kinases in a mammal, the methods comprising administering thereto a therapeutically effective amount of N-(4-{4-amino-7-[1-(2-hydroxy-2-methylpropyl)-1H-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N′-(4-methoxyphenyl)urea, and a therapeutically effective amount of one additional therapeutic agent or more than one additional therapeutic agent alone or in combination with radiotherapy.
  • Still another embodiment pertains to methods of treating diseases involving mediation, overexpression or disregulation of kinases in a mammal, the methods comprising administering thereto a therapeutically effective amount of N-[4-(4-amino-7-{1-[(2S)-2,3-dihydroxypropyl]-1H-pyrazol-4-yl}thieno[3,2-c]pyridin-3-yl)phenyl]-N′-(4-methoxyphenyl)urea, and a therapeutically effective amount of one additional therapeutic agent or more than one additional therapeutic agent alone or in combination with radiotherapy.
  • Another embodiment pertains to methods of treating pediatric cancer or neoplasm such as embryonal rhabdomyosarcoma, pediatric acute lymphoblastic leukemia, pediatric acute myelogenous leukemia, pediatric alveolar rhabdomyosarcoma, pediatric anaplastic ependymoma, pediatric anaplastic large cell lymphoma, pediatric anaplastic medulloblastoma, pediatric atypical teratoid/rhabdoid tumor of the central nervous system, pediatric biphenotypic acute leukemia, pediatric Burkitts lymphoma, pediatric cancers of Ewing's family of tumors such as primitive neuroectodermal rumors, pediatric diffuse anaplastic Wilm's tumor, pediatric favorable histology Wilm's tumor, pediatric glioblastoma, pediatric medulloblastoma, pediatric neuroblastoma, pediatric neuroblastoma-derived myelocytomatosis, pediatric pre-B-cell cancers (such as leukemia), pediatric psteosarcoma, pediatric rhabdoid kidney tumor, pediatric rhabdomyosarcoma, and pediatric T-cell cancers such as lymphoma and skin cancer, the methods comprising administering thereto a therapeutically effective amount of a compound of formula I, alone or in combination with radiotherapy.
  • Another embodiment pertains to methods of treating pediatric cancer or neoplasm such as embryonal rhabdomyosarcoma, pediatric acute lymphoblastic leukemia, pediatric acute myelogenous leukemia, pediatric alveolar rhabdomyosarcoma, pediatric anaplastic ependymoma, pediatric anaplastic large cell lymphoma, pediatric anaplastic medulloblastoma, pediatric atypical teratoid/rhabdoid tumor of the central nervous system, pediatric biphenotypic acute leukemia, pediatric Burkitts lymphoma, pediatric cancers of Ewing's family of tumors such as primitive neuroectodermal rumors, pediatric diffuse anaplastic Wilm's tumor, pediatric favorable histology Wilm's tumor, pediatric glioblastoma, pediatric medulloblastoma, pediatric neuroblastoma, pediatric neuroblastoma-derived myelocytomatosis, pediatric pre-B-cell cancers (such as leukemia), pediatric psteosarcoma, pediatric rhabdoid kidney tumor, pediatric rhabdomyosarcoma, and pediatric T-cell cancers such as lymphoma and skin cancer, the methods comprising administering thereto a therapeutically effective amount of N-(4-{4-amino-7-[1-(2-hydroxyethyl)-1H-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N′-(3-fluorophenyl)ure, alone or in combination with radiotherapy.
  • Another embodiment pertains to methods of treating pediatric cancer or neoplasm such as embryonal rhabdomyosarcoma, pediatric acute lymphoblastic leukemia, pediatric acute myelogenous leukemia, pediatric alveolar rhabdomyosarcoma, pediatric anaplastic ependymoma, pediatric anaplastic large cell lymphoma, pediatric anaplastic medulloblastoma, pediatric atypical teratoid/rhabdoid tumor of the central nervous system, pediatric biphenotypic acute leukemia, pediatric Burkitts lymphoma, pediatric cancers of Ewing's family of tumors such as primitive neuroectodermal rumors, pediatric diffuse anaplastic Wilm's tumor, pediatric favorable histology Wilm's tumor, pediatric glioblastoma, pediatric medulloblastoma, pediatric neuroblastoma, pediatric neuroblastoma-derived myelocytomatosis, pediatric pre-B-cell cancers (such as leukemia), pediatric psteosarcoma, pediatric rhabdoid kidney tumor, pediatric rhabdomyosarcoma, and pediatric T-cell cancers such as lymphoma and skin cancer, the methods comprising administering thereto a therapeutically effective amount of N-(4-{4-amino-7-[1-(2-hydroxyethyl)-1H-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N′-[4-(difluoromethoxy)phenyl]urea, alone or in combination with radiotherapy.
  • Another embodiment pertains to methods of treating pediatric cancer or neoplasm such as embryonal rhabdomyosarcoma, pediatric acute lymphoblastic leukemia, pediatric acute myelogenous leukemia, pediatric alveolar rhabdomyosarcoma, pediatric anaplastic ependymoma, pediatric anaplastic large cell lymphoma, pediatric anaplastic medulloblastoma, pediatric atypical teratoid/rhabdoid tumor of the central nervous system, pediatric biphenotypic acute leukemia, pediatric Burkitts lymphoma, pediatric cancers of Ewing's family of tumors such as primitive neuroectodermal rumors, pediatric diffuse anaplastic Wilm's tumor, pediatric favorable histology Wilm's tumor, pediatric glioblastoma, pediatric medulloblastoma, pediatric neuroblastoma, pediatric neuroblastoma-derived myelocytomatosis, pediatric pre-B-cell cancers (such as leukemia), pediatric psteosarcoma, pediatric rhabdoid kidney tumor, pediatric rhabdomyosarcoma, and pediatric T-cell cancers such as lymphoma and skin cancer, the methods comprising administering thereto a therapeutically effective amount of N-[4-(4-amino-7-{1-[(2S)-2-hydroxypropyl]-1H-pyrazol-4-yl}thieno[3,2-c]pyridin-3-yl)phenyl]-N′-(3-methylphenyl)urea, alone or in combination with radiotherapy.
  • Another embodiment pertains to methods of treating pediatric cancer or neoplasm such as embryonal rhabdomyosarcoma, pediatric acute lymphoblastic leukemia, pediatric acute myelogenous leukemia, pediatric alveolar rhabdomyosarcoma, pediatric anaplastic ependymoma, pediatric anaplastic large cell lymphoma, pediatric anaplastic medulloblastoma, pediatric atypical teratoid/rhabdoid tumor of the central nervous system, pediatric biphenotypic acute leukemia, pediatric Burkitts lymphoma, pediatric cancers of Ewing's family of tumors such as primitive neuroectodermal rumors, pediatric diffuse anaplastic Wilm's tumor, pediatric favorable histology Wilm's tumor, pediatric glioblastoma, pediatric medulloblastoma, pediatric neuroblastoma, pediatric neuroblastoma-derived myelocytomatosis, pediatric pre-B-cell cancers (such as leukemia), pediatric psteosarcoma, pediatric rhabdoid kidney tumor, pediatric rhabdomyosarcoma, and pediatric T-cell cancers such as lymphoma and skin cancer, the methods comprising administering thereto a therapeutically effective amount of N-(4-{4-amino-7-[1-(2-hydroxy-2-methylpropyl)-1H-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N′-(4-methoxyphenyl)urea, alone or in combination with radiotherapy.
  • Another embodiment pertains to methods of treating pediatric cancer or neoplasm such as embryonal rhabdomyosarcoma, pediatric acute lymphoblastic leukemia, pediatric acute myelogenous leukemia, pediatric alveolar rhabdomyosarcoma, pediatric anaplastic ependymoma, pediatric anaplastic large cell lymphoma, pediatric anaplastic medulloblastoma, pediatric atypical teratoid/rhabdoid tumor of the central nervous system, pediatric biphenotypic acute leukemia, pediatric Burkitts lymphoma, pediatric cancers of Ewing's family of tumors such as primitive neuroectodermal rumors, pediatric diffuse anaplastic Wilm's tumor, pediatric favorable histology Wilm's tumor, pediatric glioblastoma, pediatric medulloblastoma, pediatric neuroblastoma, pediatric neuroblastoma-derived myelocytomatosis, pediatric pre-B-cell cancers (such as leukemia), pediatric psteosarcoma, pediatric rhabdoid kidney tumor, pediatric rhabdomyosarcoma, and pediatric T-cell cancers such as lymphoma and skin cancer, the methods comprising administering thereto a therapeutically effective amount of N-[4-(4-amino-7-{1-[(2S)-2,3-dihydroxypropyl]-1H-pyrazol-4-yl}thieno[3,2-c]pyridin-3-yl)phenyl]-N′-(4-methoxyphenyl)urea, alone or in combination with radiotherapy.
  • Still another embodiment pertains to compounds
    • N-(4-{4-amino-7-[1-(2-hydroxyethyl)-1H-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N′-(3-fluorophenyl)urea;
    • N-(4-{4-amino-7-[1-(2-hydroxyethyl)-1H-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N′-[4-(difluoromethoxy)phenyl]urea;
    • N-[4-(4-amino-7-{1-[(2S)-2-hydroxypropyl]-1H-pyrazol-4-yl}thieno[3,2-c]pyridin-3-yl)phenyl]-N′-(3-methylphenyl)urea;
    • N-(4-{4-amino-7-[1-(2-hydroxy-2-methylpropyl)-1H-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N′-(4-methoxyphenyl)urea; and
    • N-[4-(4-amino-7-{1-[(2S)-2,3-dihydroxypropyl]-1H-pyrazol-4-yl}thieno[3,2-c]pyridin-3-yl)phenyl]-N′-(4-methoxyphenyl)urea and therapeutically acceptable salts thereof.
  • Still another embodiment pertains to N-(4-{4-amino-7-[1-(2-hydroxyethyl)-1H-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N′-(3-fluorophenyl)urea.
  • Still another embodiment pertains to N-(4-{4-amino-7-[1-(2-hydroxyethyl)-1H-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N′-[4-(difluoromethoxy)phenyl]urea.
  • Still another embodiment pertains to N-[4-(4-amino-7-{1-[(2S)-2-hydroxypropyl]-1H-pyrazol-4-yl}thieno[3,2-c]pyridin-3-yl)phenyl]-N′-(3-methylphenyl)urea.
  • Still another embodiment pertains to N-(4-{4-amino-7-[1-(2-hydroxy-2-methylpropyl)-1H-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N′-(4-methoxyphenyl)urea.
  • Still another embodiment pertains to N-[4-(4-amino-7-{1-[(2S)-2,3-dihydroxypropyl]-1H-pyrazol-4-yl}thieno[3,2-c]pyridin-3-yl)phenyl]-N′-(4-methoxyphenyl)urea.
  • Still another embodiment pertains to a compound of formula I, and therapeutically acceptable salts, prodrugs, esters, amides, salts of prodrugs, salts of esters, and salts of amides thereof.
  • Still another embodiment pertains to N-(4-{4-amino-7-[1-(2-hydroxyethyl)-1H-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N′-(3-fluorophenyl)urea, and therapeutically acceptable salts, prodrugs, esters, amides, salts of prodrugs, salts of esters, and salts of amides thereof.
  • Still another embodiment pertains to N-(4-{4-amino-7-[1-(2-hydroxyethyl)-1H-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N′-[4-(difluoromethoxy)phenyl]urea, and therapeutically acceptable salts, prodrugs, esters, amides, salts of prodrugs, salts of esters, and salts of amides thereof.
  • Still another embodiment pertains to N-[4-(4-amino-7-{1-[(2S)-2-hydroxypropyl]-1H-pyrazol-4-yl}thieno[3,2-c]pyridin-3-yl)phenyl]-N′-(3-methylphenyl)urea, and therapeutically acceptable salts, prodrugs, esters, amides, salts of prodrugs, salts of esters, and salts of amides thereof.
  • Still another embodiment pertains to N-(4-{4-amino-7-[1-(2-hydroxy-2-methylpropyl)-1H-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N′-(4-methoxyphenyl)urea, and therapeutically acceptable salts, prodrugs, esters, amides, salts of prodrugs, salts of esters, and salts of amides thereof.
  • Still another embodiment pertains to N-[4-(4-amino-7-{1-[(2S)-2,3-dihydroxypropyl]-1H-pyrazol-4-yl}thieno[3,2-c]pyridin-3-yl)phenyl]-N-(4-methoxyphenyl)urea, and therapeutically acceptable salts, prodrugs, esters, amides, salts of prodrugs, salts of esters, and salts of amides thereof.
  • DETAILED DESCRIPTION OF THE INVENTION
  • Variable moieties of compounds herein are represented by identifiers (capital letters with numerical and/or alphabetical superscripts) and may be specifically embodied.
  • It is meant to be understood that proper valences are maintained for all moieties and combinations thereof, and that monovalent moieties having more than one atom are attached through their left ends.
  • It is also meant to be understood that a specific embodiment of a variable moiety may be the same or different as another specific embodiment having the same identifier.
  • The term “alkoxy,” as used herein, refers to an alkyl group attached to the parent molecular moiety through an oxygen atom.
  • The term “alkyl,” as used herein, refers to a group derived from a straight or branched chain saturated hydrocarbon containing from one to ten carbon atoms.
  • The terms “halo” and “halogen,” as used herein, refer to F, Cl, Br, or I.
  • The term “haloalkoxy,” as used herein, refers to a haloalkyl group attached to the parent molecular moiety through an oxygen atom.
  • The term “haloalkyl,” as used herein, refers to an alkyl group, as defined herein, substituted with at least one halogen, as defined herein.
  • The term “hydroxy,” as used herein, refers to a —OH group.
  • The term “hydroxyalkyl,” as used herein, refers to an alkyl group substituted with at least one hydroxy group.
  • The term “KDR” means kinase insert domain receptor (a type III receptor tyrosine kinase) and is also known as FLK1, VEGFR, VEGFR2, and CD309.
  • The term “VEGFR” means vascular endothelial growth factor receptor.
  • The term “PDGFR” means platelet-derived growth factor receptor.
  • Compounds of this invention may contain asymmetrically substituted carbon atoms in the R or S configuration, wherein the terms “R” and “S” are as defined in Pure Appl. Chem. (1976) 45, 13-10. Compounds having asymmetrically substituted carbon atoms with equal amounts of R and S configurations are racemic at those atoms. Atoms having excess of one configuration over the other are assigned the configuration in excess, preferably an excess of about 85%-90%, more preferably an excess of about 95%-99%, and still more preferably an excess greater than about 99%. Accordingly, this invention is meant to embrace racemic mixtures and relative and absolute diastereoisomers of the compounds thereof.
  • Compounds of this invention may also contain carbon-carbon double bonds or carbon-nitrogen double bonds in the E or Z configuration, wherein the term “E” represents higher order substituents on opposite sides of the carbon-carbon or carbon-nitrogen double bond and the term “Z” represents higher order substituents on the same side of the carbon-carbon or carbon-nitrogen double bond as determined by the Cahn-Ingold-Prelog Priority Rules. The compounds of this invention may also exist as a mixture of “E” and “Z” isomers.
  • Compounds of this invention may also exist as tautomers or equilibrium mixtures thereof wherein a proton of a compound shifts from one atom to another. Examples of tautomers include, but are not limited to, keto-enol, phenol-keto, oxime-nitroso, nitro-aci, imine-enamine and the like.
  • Compounds of this invention containing NH, C(O)OH, OH or SH moieties may have attached thereto prodrug-forming moieties. The prodrug-forming moieties are removed by metabolic processes and release the compounds having the freed NH, C(O)OH, OH or SH in vivo. Prodrugs are useful for adjusting such pharmacokinetic properties of the compounds as solubility and/or hydrophobicity, absorption in the gastrointestinal tract, bioavailability, tissue penetration, and rate of clearance.
  • Metabolites of compounds of this invention produced by in vitro or in vivo metabolic processes, may also have utility for treating diseases associated with overexpression or disregulation of protein kinases.
  • Certain precursor compounds which may be metabolized in vitro or in vivo to form compounds of this invention may also have utility for treating diseases associated with overexpression or disregulation of protein kinases.
  • Compounds of this invention may exist as acid addition salts, basic addition salts or zwitterions. Salts of compounds having Formula I are prepared during their isolation or following their purification. Acid addition salts are those derived from the reaction of a compound of this invention with acid. Accordingly, salts including the acetate, adipate, alginate, bicarbonate, citrate, aspartate, benzoate, benzenesulfonate (besylate), bisulfate, butyrate, camphorate, camphorsulfonate, digluconate, formate, fumarate, glycerophosphate, glutamate, hemisulfate, heptanoate, hexanoate, hydrochloride, hydrobromide, hydroiodide, lactobionate, lactate, maleate, mesitylenesulfonate, methanesulfonate, naphthylenesulfonate, nicotinate, oxalate, pamoate, pectinate, persulfate, phosphate, picrate, propionate, succinate, tartrate, thiocyanate, trichloroacetic, trifluoroacetic, para-toluenesulfonate and undecanoate salts of the compounds of this invention are meant to be embraced by this invention. Basic addition salts of compounds are those derived from the reaction of the compounds of this invention with the bicarbonate, carbonate, hydroxide or phosphate of cations such as lithium, sodium, potassium, calcium and magnesium.
  • Compounds having Formula I may be administered, for example, bucally, ophthalmically, orally, osmotically, parenterally (intramuscularly, intraperintoneally intrasternally, intravenously, subcutaneously), rectally, topically, transdermally, vaginally and intraarterially as well as by intraarticular injection, infusion, and placement in the body, such as, for example, the vasculature.
  • Therapeutically effective amounts of a compound having Formula I depend on recipient of treatment, disease treated and severity thereof, composition comprising it, time of administration, route of administration, duration of treatment, potency, rate of clearance and whether or not another drug is co-administered. The amount of a compound having Formula I used to make a composition to be administered daily to a patient in a single dose or in divided doses is from about 0.03 to about 200 mg/kg body weight. Single dose compositions contain these amounts or a combination of submultiples thereof.
  • Compounds having Formula I may be administered with or without an excipient. Excipients include, but are not limited to, encapsulators and additives such as absorption accelerators, antioxidants, binders, buffers, coating agents, coloring agents, diluents, disintegrating agents, emulsifiers, extenders, fillers, flavoring agents, humectants, lubricants, perfumes, preservatives, propellants, releasing agents, sterilizing agents, sweeteners, solubilizers, wetting agents, mixtures thereof and the like.
  • Excipients for preparation of compositions comprising a compound having Formula Ito be administered orally include, but are not limited to, agar, alginic acid, aluminum hydroxide, benzyl alcohol, benzyl benzoate, 1,3-butylene glycol, carbomers, castor oil, cellulose, cellulose acetate, cocoa butter, corn starch, corn oil, cottonseed oil, cross-povidone, diglycerides, ethanol, ethyl cellulose, ethyl laureate, ethyl oleate, fatty acid esters, gelatin, germ oil, glucose, glycerol, groundnut oil, hydroxypropylmethyl celluose, isopropanol, isotonic saline, lactose, magnesium hydroxide, magnesium stearate, malt, mannitol, monoglycerides, olive oil, peanut oil, potassium phosphate salts, potato starch, povidone, propylene glycol, Ringer's solution, safflower oil, sesame oil, sodium carboxymethyl cellulose, sodium phosphate salts, sodium lauryl sulfate, sodium sorbitol, soybean oil, stearic acids, stearyl fumarate, sucrose, surfactants, talc, tragacanth, tetrahydrofurfuryl alcohol, triglycerides, water, mixtures thereof and the like. Excipients for preparation of compositions comprising a compound having Formula Ito be administered ophthalmically or orally include, but are not limited to, 1,3-butylene glycol, castor oil, corn oil, cottonseed oil, ethanol, fatty acid esters of sorbitan, germ oil, groundnut oil, glycerol, isopropanol, olive oil, polyethylene glycols, propylene glycol, sesame oil, water, mixtures thereof and the like. Excipients for preparation of compositions comprising a compound having Formula I to be administered osmotically include, but are not limited to, chlorofluorohydrocarbons, ethanol, water, mixtures thereof and the like. Excipients for preparation of compositions comprising a compound having Formula Ito be administered parenterally include, but are not limited to, 1,3-butanediol, castor oil, corn oil, cottonseed oil, dextrose, germ oil, groundnut oil, liposomes, oleic acid, olive oil, peanut oil, Ringer's solution, safflower oil, sesame oil, soybean oil, U.S.P. or isotonic sodium chloride solution, water, mixtures thereof and the like. Excipients for preparation of compositions comprising a compound having Formula Ito be administered rectally or vaginally include, but are not limited to, cocoa butter, polyethylene glycol, wax, mixtures thereof and the like.
  • Compounds having Formula I are expected to be useful when used with alkylating agents, angiogenesis inhibitors, antibodies, antimetabolites, antimitotics, antiproliferatives, aurora kinase inhibitors, other apoptosis promoters (for example, Bcl-xL, Bcl-w and Bfl-1) inhibitors, Bcr-Abl kinase inhibitors, BiTE (Bi-Specific T cell Engager) antibodies, biologic response modifiers, cyclin-dependent kinase inhibitors, cell cycle inhibitors, cyclooxygenase-2 inhibitors, DVD's, leukemia viral oncogene homolog (ErbB2) receptor inhibitors, growth factor inhibitors, heat shock protein (HSP)-90 inhibitors, histone deacetylase (HDAC) inhibitors, hormonal therapies, immunologicals, inhibitors of apoptosis proteins (IAP's) intercalating antibiotics, kinase inhibitors, mammalian target of rapamycin inhibitors, microRNA's mitogen-activated extracellular signal-regulated kinase inhibitors, multivalent binding proteins, non-steroidal anti-inflammatory drugs (NSAIDs), poly ADP (adenosine diphosphate)-ribose polymerase (PARP) inhibitors, platinum chemotherapeutics, polo-like kinase (Plk) inhibitors, proteosome inhibitors, purine analogs, pyrimidine analogs, receptor tyrosine kinase inhibitors, retinoids/deltoids plant alkaloids, small inhibitory ribonucleic acids (siRNA's), topoisomerase inhibitors, combinations thereof and the like.
  • A BiTE antibody is a bi-specific antibody that directs T-cells to attach cancer cells by simultaneously binding the two cells. The T-cell then attacks the target cancer cell. Exemplary BiTE antibodies include adecatumumab (Micromet MT201), blinatumomab (Micromet MT103) and the like.
  • SiRNA's are molecules having endogenous RNA bases or chemically modified nucleotides. The modifications shall not abolish cellular activity, but rather impart increased stability and/or increased cellular potency. Examples of chemical modifications include phosphorothioate groups, 2′-deoxynucleotide, 2′-OCH3-containing ribonucleotides, 2′-F-ribonucleotides, 2′-methoxyethyl ribonucleotides or a combination thereof. The siRNA can have varying lengths (10-200 bps) and structures (hairpins, single/double strands, bulges, nicks/gaps, mismatches) and processed in the cell to provide active gene silencing. In certain embodiments, a double-stranded siRNA (dsRNA) can have the same number of nucleotides on each strand (blunt ends) or asymmetric ends (overhangs). The overhang of 1-2 nucleotides can be present on the sense and/or the antisense strand, as well as present on the 5′- and/or the 3′- ends of a given strand.
  • Multivalent binding proteins are binding proteins comprising two or more antigen binding sites. The multivalent binding protein is preferably engineered to have the three or more antigen binding sites and is generally not a naturally occurring antibody. The term “multispecific binding protein” means a binding protein capable of binding two or more related or unrelated targets. Dual variable domain (DVD) binding proteins are tetravalent or multivalent binding proteins binding proteins comprising two or more antigen binding sites. Such DVDs may be monospecific, i.e., capable of binding one antigen or multispecific, i.e., capable of binding two or more antigens. DVD binding proteins comprising two heavy chain DVD polypeptides and two light chain DVD polypeptides are referred to as DVD Ig. Each half of a DVD Ig comprises a heavy chain DVD polypeptide, a light chain DVD polypeptide, and two antigen binding sites. Each binding site comprises a heavy chain variable domain and a light chain variable domain with a total of 6 CDRs involved in antigen binding per antigen binding site.
  • Alkylating agents include altretamine, AMD-473, AP-5280, apaziquone, bendamustine, brostallicin, busulfan, carboquone, carmustine (BCNU), chlorambucil, CLORETAZINE® (laromustine, VNP 40101M), cyclophosphamide, decarbazine, estramustine, fotemustine, glufosfamide, ifosfamide, KW-2170, lomustine (CCNU), mafosfamide, melphalan, mitobronitol, mitolactol, nimustine, nitrogen mustard N-oxide, ranimustine, temozolomide, thiotepa, TREANDA® (bendamustine), treosulfan, rofosfamide and the like.
  • Angiogenesis inhibitors include endothelial-specific receptor tyrosine kinase (Tie-2) inhibitors, epidermal growth factor receptor (EGFR) inhibitors, insulin growth factor-2 receptor (IGFR-2) inhibitors, matrix metalloproteinase-2 (MMP-2) inhibitors, matrix metalloproteinase-9 (MMP-9) inhibitors, platelet-derived growth factor receptor (PDGFR) inhibitors, thrombospondin analogs, vascular endothelial growth factor receptor tyrosine kinase (VEGFR) inhibitors and the like.
  • Antimetabolites include ALIMTA® (metrexed disodium, LY231514, MTA), 5-azacitidine, XELODA® (capecitabine), carmofur, LEUSTAT® (cladribine), clofarabine, cytarabine, cytarabine ocfosfate, cytosine arabinoside, decitabine, deferoxamine, doxifluridine, eflornithine, EICAR (5-ethynyl-1-β-D-ribofuranosylimidazole-4-carboxamide), enocitabine, ethnylcytidine, fludarabine, 5-fluorouracil alone or in combination with leucovorin, GEMZAR® (gemcitabine), hydroxyurea, ALKERAN® (melphalan), mercaptopurine, 6-mercaptopurine riboside, methotrexate, mycophenolic acid, nelarabine, nolatrexed, ocfosfate, pelitrexol, pentostatin, raltitrexed, Ribavirin, triapine, trimetrexate, S-1, tiazofurin, tegafur, TS-1, vidarabine, UFT and the like.
  • Aurora kinase inhibitors include AZD-1152, MLN-8054, VX-680 and the like.
  • Bcl-2 proteins inhibitors include AT-101 ((−)gossypol), GENASENSE® (G3139 or oblimersen (Bcl-2-targeting antisense oligonucleotide)), IPI-194, IPI-565, N-(4-(4-((4′-chloro(1,1′-biphenyl)-2-yl)methyl)piperazin-1-yl)benzoyl)-4-(((1R)-3-(dimethylamino)-1-((phenylsulfanyl)methyl)propyl)amino)-3-nitrobenzenesulfonamide) (ABT-737), N-(4-(4-((2-(4-chlorophenyl)-5,5-dimethyl-1-cyclohex-1-en-1-yl)methyl)piperazin-1-yl)benzoyl)-4-(((1R)-3-(morpholin-4-yl)-1-((phenylsulfanyl)methyl)propyl)amino)-3-((trifluoromethyl)sulfonyl)benzenesulfonamide (ABT-263), GX-070 (obatoclax) and the like.
  • Bcr-Abl kinase inhibitors include DASATINIB® (BMS-354825), GLEEVEC® (imatinib) and the like.
  • CDK inhibitors include AZD-5438, BMI-1040, BMS-032, BMS-387, CVT-2584, flavopyridol, GPC-286199, MCS-5A, PD0332991, PHA-690509, seliciclib (CYC-202, R-roscovitine), ZK-304709 and the like.
  • COX-2 inhibitors include ABT-963, ARCOXIA® (etoricoxib), BEXTRA® (valdecoxib), BMS347070, CELEBREX® (celecoxib), COX-189 (lumiracoxib), CT-3, DERAMAXX® (deracoxib), JTE-522, 4-methyl-2-(3,4-dimethylphenyl)-1-(4-sulfamoylphenyl-1H-pyrrole), MK-663 (etoricoxib), NS-398, parecoxib, RS-57067, SC-58125, SD-8381, SVT-2016, S-2474, T-614, VIOXX® (rofecoxib) and the like.
  • EGFR inhibitors include ABX-EGF, anti-EGFR immunoliposomes, EGF-vaccine, EMD-7200, ERBITUX® (cetuximab), HR3, IgA antibodies, IRESSA® (gefitinib), TARCEVA® (erlotinib or OSI-774), TP-38, EGFR fusion protein, TYKERB® (lapatinib) and the like.
  • ErbB2 receptor inhibitors include CP-724-714, CI-1033 (canertinib), HERCEPTIN® (trastuzumab), TYKERB® (lapatinib), OMNITARG® (2C4, petuzumab), TAK-165, GW-572016 (ionafarnib), GW-282974, EKB-569, PI-166, dHER2 (HER2 vaccine), APC-8024 (HER-2 vaccine), anti-HER/2neu bispecific antibody, B7.her2IgG3, AS HER2 trifunctional bispecfic antibodies, mAB AR-209, mAB 2B-1 and the like.
  • Histone deacetylase inhibitors include depsipeptide, LAQ-824, MS-275, trapoxin, suberoylanilide hydroxamic acid (SAHA), TSA, valproic acid and the like.
  • HSP-90 inhibitors include 17-AAG-nab, 17-AAG, CNF-101, CNF-1010, CNF-2024, 17-DMAG, geldanamycin, IPI-504, KOS-953, MYCOGRAB® (human recombinant antibody to HSP-90), NCS-683664, PU24FC1, PU-3, radicicol, SNX-2112, STA-9090 VER49009 and the like.
  • Inhibitors of apoptosis proteins include ApoMab (a fully human affinity-matured IgG1 monoclonal antibody), antibodies that target TRAIL or death receptors (e.g., pro-apoptotic receptor agonists DR4 and DR5), conatumumab, ETR2-ST01, GDC0145, (lexatumumab), HGS-1029, LBY-135, PRO-1762 and tratuzumab.
  • MEK inhibitors include ARRY-142886, ARRY-438162 PD-325901, PD-98059 and the like.
  • mTOR inhibitors include AP-23573, CCI-779, everolimus, RAD-001, rapamycin, temsirolimus and the like.
  • Non-steroidal anti-inflammatory drugs include AMIGESIC® (salsalate), DOLOBID® (diflunisal), MOTRIN® (ibuprofen), ORUDIS® (ketoprofen), RELAFEN® (nabumetone), FELDENE® (piroxicam), ibuprofen cream, ALEVE® (naproxen) and NAPROSYN® (naproxen), VOLTAREN® (diclofenac), INDOCIN® (indomethacin), CLINORIL® (sulindac), TOLECTIN® (tolmetin), LODINE® (etodolac), TORADOL® (ketorolac), DAYPRO® (oxaprozin) and the like.
  • PDGFR inhibitors include C-451, CP-673, CP-868596 and the like.
  • Platinum chemotherapeutics include cisplatin, ELOXATIN® (oxaliplatin) eptaplatin, lobaplatin, nedaplatin, PARAPLATIN® (carboplatin), satraplatin and the like.
  • Polo-like kinase inhibitors include BI-2536 and the like.
  • Thrombospondin analogs include ABT-510, ABT-567, TSP-1 and the like.
  • VEGFR inhibitors include AVASTIN® (bevacizumab), ABT-869, AEE-788, ANGIOZYME™ (a ribozyme that inhibits angiogenesis (Ribozyme Pharmaceuticals (Boulder, Colo.) and Chiron, (Emeryville, Calif.)), axitinib (AG-13736), AZD-2171, CP-547,632, IM-862, MACUGEN (pegaptamib), NEXAVAR® (sorafenib, BAY43-9006), pazopanib (GW-786034), vatalanib (PTK-787, ZK-222584), SUTENT® (sunitinib, SU-11248), VEGF trap, ZACTIMA™ (vandetanib, ZD-6474) and the like.
  • Antibiotics include intercalating antibiotics aclarubicin, actinomycin D, amrubicin, annamycin, adriamycin, BLENOXANE® (bleomycin), daunorubicin, CAELYX® or MYOCET® (liposomal doxorubicin), elsamitrucin, epirbucin, glarbuicin, ZAVEDOS® (idarubicin), mitomycin C, nemorubicin, neocarzinostatin, peplomycin, pirarubicin, rebeccamycin, stimalamer, streptozocin, VALSTAR® (valrubicin), zinostatin and the like.
  • Topoisomerase inhibitors include aclarubicin, 9-aminocamptothecin, amonafide, amsacrine, becatecarin, belotecan, BN-80915, CAMPTOSAR® (irinotecan hydrochloride), camptothecin, CARDIOXANE® (dexrazoxine), diflomotecan, edotecarin, ELLENCE® or PHARMORUBICIN® (epirubicin), etoposide, exatecan, 10-hydroxycamptothecin, gimatecan, lurtotecan, mitoxantrone, orathecin, pirarbucin, pixantrone, rubitecan, sobuzoxane, SN-38, tafluposide, topotecan and the like.
  • Antibodies include AVASTIN® (bevacizumab), CD40-specific antibodies, chTNT-1/B, denosumab, ERBITUX® (cetuximab), HUMAX-CD4® (zanolimumab), IGF1R-specific antibodies, lintuzumab, PANOREX® (edrecolomab), RENCAREX® (WX G250), RITUXAN® (rituximab), ticilimumab, trastuzimab and the like.
  • Hormonal therapies include ARIMIDEX® (anastrozole), AROMASIN® (exemestane), arzoxifene, CASODEX® (bicalutamide), CETROTIDE® (cetrorelix), degarelix, deslorelin, DESOPAN® (trilostane), dexamethasone, DROGENIL®, (flutamide), EVISTA® (raloxifene), AFEMA™ (fadrozole), FARESTON® (toremifene), FASLODEX® (fulvestrant), FEMARA® (letrozole), formestane, glucocorticoids, HECTOROL® (doxercalciferol), RENAGEL® (sevelamer carbonate), lasofoxifene, leuprolide acetate, MEGACE® (megesterol), MIFEPREX® (mifepristone), NILANDRON™ (nilutamide), NOLVADEX® (tamoxifen citrate), PLENAXIS™ (abarelix), prednisone, PROPECIA® (finasteride), rilostane, SUPREFACT® (buserelin), TRELSTAR® (luteinizing hormone releasing hormone (LHRH)), VANTAS® (Histrelin implant), VETORYL® (trilostane or modrastane), ZOLADEX® (fosrelin, goserelin) and the like.
  • Deltoids and retinoids include seocalcitol (EB1089, CB1093), lexacalcitrol (KH1060), fenretinide, PANRETIN® (aliretinoin), ATRAGEN® (liposomal tretinoin), TARGRETIN® (bexarotene), LGD-1550 and the like.
  • PARP inhibitors include ABT-888, olaparib, KU-59436, AZD-2281, AG-014699, BSI-201, BGP-15, INO-1001, ONO-2231 and the like.
  • Plant alkaloids include, but are not limited to, vincristine, vinblastine, vindesine, vinorelbine and the like.
  • Proteasome inhibitors include VELCADE® (bortezomib), MG132, NPI-0052, PR-171 and the like.
  • Examples of immunologicals include interferons and other immune-enhancing agents. Interferons include interferon alpha, interferon alpha-2a, interferon alpha-2b, interferon beta, interferon gamma-1a, ACTIMMUNE® (interferon gamma-1b), or interferon gamma-n1, combinations thereof and the like. Other agents include ALFAFERONE®, (IFN-α), BAM-002 (oxidized glutathione), BEROMUN® (tasonermin), BEXXAR® (tositumomab), CAMPATH® (alemtuzumab), CTLA4 (cytotoxic lymphocyte antigen 4), decarbazine, denileukin, epratuzumab, GRANOCYTE® (lenograstim), lentinan, leukocyte alpha interferon, imiquimod, MDX-010 (anti-CTLA-4), melanoma vaccine, mitumomab, molgramostim, MYLOTARG™ (gemtuzumab ozogamicin), NEUPOGEN® (filgrastim), OncoVAC-CL, OVAREX® (oregovomab), pemtumomab (Y-muHMFG1), PROVENGE® (sipuleucel-T), sargaramostim, sizofilan, teceleukin, THERACYS® (Bacillus Calmette-Guerin), ubenimex, VIRULIZIN® (immunotherapeutic, Lorus Pharmaceuticals), Z-100 (Specific Substance of Maruyama (SSM)), WF-10 (Tetrachlorodecaoxide (TCDO)), PROLEUKIN® (aldesleukin), ZADAXIN® (thymalfasin), ZENAPAX® (daclizumab), ZEVALIN® (90Y-Ibritumomab tiuxetan) and the like.
  • Biological response modifiers are agents that modify defense mechanisms of living organisms or biological responses, such as survival, growth, or differentiation of tissue cells to direct them to have anti-tumor activity and include include krestin, lentinan, sizofuran, picibanil PF-3512676 (CpG-8954), ubenimex and the like.
  • Pyrimidine analogs include cytarabine (ara C or Arabinoside C), cytosine arabinoside, doxifluridine, FLUDARA® (fludarabine), 5-FU (5-fluorouracil), floxuridine, GEMZAR® (gemcitabine), TOMUDEX® (ratitrexed), TROXATYL™ (triacetyluridine troxacitabine) and the like.
  • Purine analogs include LANVIS® (thioguanine) and PURI-NETHOL® (mercaptopurine).
  • Antimitotic agents include batabulin, epothilone D (KOS-862), N-(2-((4-hydroxyphenyl)amino)pyridin-3-yl)-4-methoxybenzenesulfonamide, ixabepilone (BMS 247550), paclitaxel, TAXOTERE® (docetaxel), PNU100940 (109881), patupilone, XRP-9881 (larotaxel), vinflunine, ZK-EPO (synthetic epothilone) and the like.
  • Compounds of this invention can also be used as radiosensitizesr that enhance the efficacy of radiotherapy. Examples of radiotherapy include external beam radiotherapy, teletherapy, brachtherapy and sealed, unsealed source radiotherapy and the like.
  • Additionally, compounds having Formula I may be combined with other chemptherapeutic agents such as ABRAXANE™ (ABI-007), ABT-100 (farnesyl transferase inhibitor), ADVEXIN® (Ad5CMV-p53 vaccine), ALTOCOR® or MEVACOR® (lovastatin), AMPLIGEN® (poly I:poly C12U, a synthetic RNA), APTOSYN® (exisulind), AREDIA® (pamidronic acid), arglabin, L-asparaginase, atamestane (1-methyl-3,17-dione-androsta-1,4-diene), AVAGE® (tazarotene), AVE-8062 (combreastatin derivative) BEC2 (mitumomab), cachectin or cachexin (tumor necrosis factor), canvaxin (vaccine), CEAVAC® (cancer vaccine), CELEUK® (celmoleukin), CEPLENE® (histamine dihydrochloride), CERVARIX® (human papillomavirus vaccine), CHOP® (C: CYTOXAN® (cyclophosphamide); H: ADRIAMYCIN® (hydroxydoxorubicin); O: Vincristine (ONCOVIN®); P: prednisone), CYPAT™ (cyproterone acetate), combrestatin A4P, DAB(389)EGF (catalytic and translocation domains of diphtheria toxin fused via a His-Ala linker to human epidermal growth factor) or TransMID-107R™ (diphtheria toxins), dacarbazine, dactinomycin, 5,6-dimethylxanthenone-4-acetic acid (DMXAA), eniluracil, EVIZON™ (squalamine lactate), DIMERICINE® (T4N5 liposome lotion), discodermolide, DX-8951f (exatecan mesylate), enzastaurin, EPO906 (epithilone B), GARDASIL® (quadrivalent human papillomavirus (Types 6, 11, 16, 18) recombinant vaccine), GASTRIMMUNE®, GENASENSE®, GMK (ganglioside conjugate vaccine), GVAX® (prostate cancer vaccine), halofuginone, histerelin, hydroxycarbamide, ibandronic acid, IGN-101, IL-13-PE38, IL-13-PE38QQR (cintredekin besudotox), IL-13-pseudomonas exotoxin, interferon-α, interferon-γ, JUNOVAN™ or MEPACT™ (mifamurtide), lonafarnib, 5,10-methylenetetrahydrofolate, miltefosine (hexadecylphosphocholine), NEOVASTAT® (AE-941), NEUTREXIN® (trimetrexate glucuronate), NIPENT® (pentostatin), ONCONASE® (a ribonuclease enzyme), ONCOPHAGE® (melanoma vaccine treatment), ONCOVAX® (IL-2 Vaccine), ORATHECIN™ (rubitecan), OSIDEM® (antibody-based cell drug), OVAREX® MAb (murine monoclonal antibody), paditaxel, PANDIMEX™ (aglycone saponins from ginseng comprising 20(S)protopanaxadiol (aPPD) and 20(S)protopanaxatriol (aPPT)), panitumumab, PANVAC®-VF (investigational cancer vaccine), pegaspargase, PEG Interferon A, phenoxodiol, procarbazine, rebimastat, REMOVAB® (catumaxomab), REVLIMID® (lenalidomide), RSR13 (efaproxiral), SOMATULINE® LA (lanreotide), SORIATANE® (acitretin), staurosporine (Streptomyces staurospores), talabostat (PT100), TARGRETIN® (bexarotene), TAXOPREXIN® (DHA-paclitaxel), TELCYTA® (canfosfamide, TLK286), temilifene, TEMODAR® (temozolomide), tesmilifene, thalidomide, THERATOPE® (STn-KLH), thymitaq (2-amino-3,4-dihydro-6-methyl-4-oxo-5-(4-pyridylthio)quinazoline dihydrochloride), TNFERADE™ (adenovector: DNA carrier containing the gene for tumor necrosis factor-α), TRACLEER® or ZAVESCA® (bosentan), tretinoin (Retin-A), tetrandrine, TRISENOX® (arsenic trioxide), VIRULIZIN®, ukrain (derivative of alkaloids from the greater celandine plant), vitaxin (anti-alphavbeta3 antibody), XCYTRIN® (motexafin gadolinium), XINLAY™ (atrasentan), XYOTAX™ (paclitaxel poliglumex), YONDELIS® (trabectedin), ZD-6126, ZINECARD® (dexrazoxane), ZOMETA® (zolendronic acid), zorubicin and the like.
  • To determine Aurora B activity of representative compounds of the invention, Active Aurora B enzyme (recombinant residues 1-344) and INCENP (recombinant GST fusion protein (Upstate)) were incubated in wells of a 384 well plate with biotinylted histone H3 peptide residues 1-21 (Upstate), 1 mM ATP, and various concentrations of inhibitors in a HEPES buffer, pH 7.4 containing MgCl2, sodium othrovanadate, and Triton X-100. After 1 hour, the reaction was stopped with EDTA and anti-phospho-histone H3 Europium Cryptate (Cis-Bio) and SA-APC (Phycolink, Prozyme) were added to detect the phosphopeptide. The amount of phosphorylation was determined by the time-resolved fluorescence ratio of signals at 665 nm and 615 nm. The IC50's were calculated by an exponential fit of the inhibition values with the inhibitor concentrations using Assay Explorer software.
  • To determine Aurora A and C activity of representative compounds of the invention, Active Aurora A or C enzyme was incubated in wells of a 384 well plate with biotinylated STK substrate-2 (Upstate), 1 mM ATP, and various concentrations of inhibitors in a Hepes buffer, pH 7.4 containing MgCl2, sodium othrovanadate, and Triton X-100. After 1 hour, the reaction was stopped with EDTA and anti-phospho-STK antibody Europium Cryptate (Upstate) and SA-XL665 (Upstate) were added to detect the phosphopeptide. The amount of phosphorylation was determined by the time-resolved fluorescence ratio of signals at 665 nm and 615 nm. The IC50s were calculated by an exponential fit of the inhibition values with the inhibitor concentrations using Assay Explorer software.
  • To determine the activity of the various kinases, a homogenous time-resolved fluorescence (HTRF) in vitro kinase assay was used. (Mathis, G., HTRF(R) Technology. J Biomol Screen, 1999. 4 (6): p. 309-314; Alfred J. Kolb, Paul V. Kaplita, David J. Hayes, Young-Whan Park, Christine Pernell, John S. Major and Gerard Mathis, Drug Discovery Today, 1998, 3, 333-342.)
  • For example for KDR, cKIT, FLT1, CSF1R and FTL3, purified enzyme was mixed with 0.5 μM N-biotinylated substrate (Biotin-Ahx-AEEEYFFLA-amide (SEQ. ID. 1)), various concentrations of inhibitor in reaction buffer (50 mM HEPES, pH 7.1, 10 mM MgCl2, 2 mM MnCl2, 0.1% BSA and 1 mM DTT, 40 μL final volume), ATP (1 mM final conc.) in a black 384-well plate. After 60 minutes incubation at room temperature, the reaction was quenched by addition of a buffered EDTA solution (final approximate concentrations: 30 mM EDTA, 0.1% BSA, 0.1% Triton X-100 and 0.24M KF) and a solution of revelation agents (to give 0.084 ng/well streptavidin-XL-665 (Cis-Bio) and 6.5 ng/well antiphsophotyrosine mAb PT66-K Europium kryptate) was added to the reaction mixture. The quenched reaction was allowed to stand at room temperature for 3 hours and was then read in a time-resolved fluorescence detector (InVision, Perkin-Elmer) at 620 nm and 665 nm sequentially with excitation. The ratio between the signal of 620 nm and 665 nm was used in the calculation of the IC50.
  • Details for the various kinases are shown in Table I.
  • TABLE I
    HTRF
    ASSAYS
    Enz.
    Reaction
    Conc.
    Enzyme Construct MW (kD) (ng/well)
    KDR His6-KDR 63 7
    789-1354
    cKIT GST- 70 4
    Fusion
    FLT1 His6-Tag 65
    CSF-1r M-His(6)- 50 10
    CSF-1R
    Q547-
    C972
    FLT3 M-His(6)- 50 0.6
    FLT3
    Q569-
    S993
    PDGFR- GST- 100 20
    beta Fusion
  • Table 2 and Table 3 demonstrate the utility of Examples 1-6 as inhibitors of multiple kinases.
  • TABLE 2
    VEGRF family PDGFR Family
    KDR FLT1 CSF1R FLT3 cKIT
    Example IC50 (μM) IC50 (μM) IC50 (μM) IC50 (μM) IC50 (μM)
    1 0.00159 0.00125 0.00342 0.0013 0.01953
    2 0.00265 0.0021 0.00222 0.00113 0.02727
    3 0.0016 0.00204 0.00562 0.00218 0.02711
    4 0.00501 0.00429 0.00787 0.00193 0.05305
    5 0.00183 0.00145 0.00134 0.00122 0.01258
    6 0.00218 0.0019 0.00274 0.00181 0.00428
  • TABLE 3
    Aurora B Aurora A
    Example IC50 (μM) IC50 (μM)
    1 0.00673 0.67542
    2 0.00787 4.7431
    3 0.01121 0.24831
    4 0.01484 3.61985
    5 0.02893 0.57014
    6 0.00844 0.3572
  • CYP 3A4 Assay
  • Assays (200 μL final volume) were carried out in NUNC polypropylene deep well plates in 50 mM potassium phosphate buffer, pH 7.4, using a microtiter plate shaker in a 37° C. incubator. Pooled human liver microsomes (BD Gentest, 50 μg/mL) were incubated with 5 concentrations of test compound (from 0.1 μM to 10 μM), 1 mM NADPH (Sigma), and 2 μM midazolam (Sigma). A constant amount of dimethylsulfoxide (1%) was added to the incubations with the test compounds, and each analysis was performed in duplicate. For preincubation experiments (Pre), the microsomes, test compounds, and NADPH were mixed and incubated 30 minutes before addition of midazolam. For coincubation experiments (Co), the compounds, microsomes, and midazolam were mixed and the reaction initiated by addition of NADPH to the wells. In both protocols, the complete reaction was terminated after 10 minutes of shaking by addition of 100 μL of a 1/1 mixture of acetonitrile and methanol containing 0.33 μM 1-hydroxytriazolam.
  • For analysis, an aliquot of each well were transferred to an autosampler vial and 10 μL injected onto a Shimadzu LC-10A HPLC system equipped with a YMC-AQ (2.0×50 mm) column. The components were separated using a gradient of 10% acetonitrile-0.1% acetic acid to 70% acetonitrile-0.1% acetic acid at a flow rate of at 0.4 mL/minute over 5 minutes. Midazolam, 1-hydroxytriazolam (internal standard), 1-hydroxymidazolam and 4-hydroxymidazolam were quantified by LC-MS/MS using an LCQ Duo (ThermoFinnigan). The ratio of 1-hydroxymidazolam (CYP 3A4 product) and the internal standard at each concentration of compound was used to calculate the % inhibition of activity relative to the ratio calculated for control incubations without inhibitor. In the absence of NADPH, no hydroxylation of midazolam was observed. Ketoconazole was used as a standard inhibitor, which at 0.1 μM produces 70-80% inhibition of CYP 3A4. The IC50 (the concentration at which 50% of the enzyme is inhibited) was calculated for Examples 1-5 and Example 6 (described as EXAMPLE 703 in WO 2005/10009) and is shown in Table 4 below.
  • TABLE 4
    Incubation IC50
    Conditions (μM)
    EXAMPLE 1 Co >10
    Pre >10
    EXAMPLE 2 Co >10
    Pre >10
    EXAMPLE 3 Co >10
    Pre >10
    EXAMPLE 4 Co >10
    Pre >10
    EXAMPLE 5 Co >10
    Pre >10
    EXAMPLE 6 Co .30
    Pre .76
  • The data in tables 2, 3 and 4 illustrate the utility of the compounds of this invention as inhibitors of multiple kinases with the added benefit of reduced CYP inhibition.
  • Compounds described as having low CYP inhibition or as not inhibiting CYP are those compounds with an IC50 of >10 μM in the above assay.
  • The structural homology between Aurora Protein Kinases A, B and C is reported in Nature Reviews/Cancer, Vol. 4 December, 2004.
  • It is expected that, because the compound of this invention inhibits the activity of Aurora-kinase B, it could also have utility as an inhibitor of protein kinases having close structural homology thereto, such as, for example, Aurora-kinase A and Aurora-kinase C.
  • Data for Example 1 is shown in Table 5.
  • TABLE 5
    Aurora B Aurora A Aurora C
    HTRF HTRF HTRF
    Example IC50 (μM) IC50 (μM) IC50 (μM)
    1 0.007 0.120 0.001
  • This data demonstrates the utility of Example 1 as an inhibitor of Aurora-kinase A and Aurora-kinase B and Aurora-kinase C.
  • Accordingly, compounds of this invention are expected to have utility in treatment of diseases during which protein kinases such as any or all Aurora-kinase family members are expressed.
  • Involvement of Aurora Kinase in pancreatic carcinoma cells is reported in Zhu, J., et al., AURKA Amplification, Chromosome Instability, And Centrosome Abnormality in Human Pancreatic Carcinoma Cells. Cancer Genet. Cytogenet., 2005. 159 (1): p. 10-17; and Li D., Zhu J., Firozi P. F., et al. Overexpression of Oncogenic STK15/BTAK/Aurora A Kinase in Human Pancreatic Cancer. Clin. Cancer Res. 2003; 9:991-7.
  • Involvement of Aurora Kinase in non-small cell lung carcinoma is reported in Smith, S. L., et al., Overexpression of Aurora B Kinase (AURKB) in Primary Non-Small Cell Lung Carcinoma is Frequent, Generally Driven from One Allele, and Correlates with the Level of Genetic Instability. Br. J. Cancer, 2005. 93 (6): p. 719-729.
  • Involvement of Aurora Kinase in prostate cancer is reported in Chieffi, P., et al., Aurora B Expression Directly Correlates with Prostate Cancer Malignancy. Prostate, 2006. 66 (3): p. 326-33; and Chieffi P., Cozzolino L., Kisslinger A., et al. Aurora B Expression Directly Correlates with Prostate Cancer Malignancy and Influences Prostate Cell Proliferation. Prostate 2006; 66:326-33.
  • Involvement of Aurora Kinase in head and neck squamous cell carcinoma is reported in Reiter, R., et al., Aurora Kinase A Messenger RNA Overexpression is Correlated with Tumor Progression and Shortened Survival in Head and Neck Squamous Cell Carcinoma. Clin Cancer Res, 2006. 12 (17): p. 5136-41.
  • Involvement of Aurora Kinase in acute myeloid leukemia is reported in Walsby E., Walsh V., Pepper C., Burnett A., and Mills K. Haematologica. 2008 May; 93(5):662-9.
  • Involvement of Aurora Kinase in breast cancer is reported in Tanaka T., Kimura M., Matsunaga K., Fukada D., Mori H., Okano Y. Centrosomal Kinase AIK1 is Overexpressed in Invasive Ductal Carcinoma of The Breast. Cancer Res. 1999; 59:2041-4; Miyoshi Y., Iwao K., Egawa C., Noguchi S. Association of Centrosomal Kinase STK15/BTAK Mrna Expression with Chromosomal Instability in Human Breast Cancers. Int. J. Cancer 2001; 92:370-3; Hogue A., Carter J., Xia W., et al. Loss Of Aurora A/STK15/BTAK Overexpression Correlates with Transition of in Situ to Invasive Ductal Carcinoma of the Breast. Cancer Epidemiol. Biomarkers Prev. 2003; 12:1518-22; Royce M. E., Xia W., Sahin A. A., et al. STK15/Aurora-A Expression in Primary Breast Tumors is Correlated with Nuclear Grade But Not With Prognosis. Cancer 2004; 100:12-9; Bodvarsdottir S. K., Hilmarsdottir H., Birgisdottir V., Steinarsdottir M., Jonasson J. G., Eyfjord J. E., Aurora-A Amplification Associated with BRCA2 Mutation in Breast Tumours. Cancer Lett 2007; 248:96-102; Sen S., Zhou H., White R. A., A Putative Serine/Threonine Kinase Encoding Gene BTAK on Chromosome 20q13 is Amplified and Overexpressed in Human Breast Cancer Cell Lines. Oncogene 1997; 14:2195-200; Lo Y. L., Yu J. C., Chen S. T., et al. Breast Cancer Risk Associated with Genotypic Polymorphism of the Mitosisregulating Gene Aurora-A/STK15/BTAK. In. J. Cancer 2005; 115:276-83; Vidarsdottir L., Bodvarsdottir S. K., Hilmarsdottir H., Tryggvadottir L., Eyfjord J. E., Breast Cancer Risk Associated with AURKA 91T a Polymorphism in Relation to BRCA Mutations. Cancer Lett 2007; 250:206-12; Cox D. G., Hankinson S. E., Hunter D. J., Polymorphisms of the Aurka (STK15/Aurora Kinase) Gene and Breast Cancer Risk (United States). Cancer Causes Control 2006; 17:81-3; and Tchatchou S., Wirtenberger M., Hemminki K., et al. Aurora Kinases A and B and Familial Breast Cancer Risk. Cancer Lett 2007; 247:266-72.
  • Involvement of Aurora Kinase in lung cancer is reported in Smith S. L., Bowers N. L., Betticher D. C., et al. Overexpression Of Aurora B Kinase (AURKB) in Primary Non small Cell Lung Carcinoma is Frequent, Generally Driven Fromone Allele, and Correlates with the Level Of Genetic Instability. Br. J. Cancer 2005; 93:719-29; Xu H. T., Ma L., Qi F. J., et al. Expression of Serine Threonine Kinase15 is Associated with Poor Differentiation in Lung Squamous Cell Carcinoma and Adenocarcinoma. Pathol. Int. 2006; 56:375-80; Vischioni B., Oudejans J. J., Vos W., Rodriguez J. A., Giaccone G. Frequent Overexpression of Aurora B Kinase, a Novel Drug Target, in Non-Small Cell Lung Carcinoma Patients. Mol. Cancer. Ther. 2006; 5:2905-13; and Gu J., Gong Y., Huang M., Lu C., Spitz M. R., Wu X. Polymorphisms Of STK15 (Aurora-A) Gene and Lung Cancer Risk in Caucasians. Carcinogenesis 2007; 28:350-5.
  • Involvement of Aurora Kinase in bladder cancer is reported in Comperat E., Camparo P., Haus R., et al. Aurora-A/STK-15 is a Predictive Factor for Recurrent Behaviour in Non-Invasive Bladder Carcinoma: A Study Of 128 Cases of Non-Invasive Neoplasms. Virchows Arch 2007; 450:419-24; Fraizer G. C., Diaz M. F., Lee I. L., Grossman H. B., Sen S. Aurora-A/STK15/BTAK Enhances Chromosomal Instability in Bladder Cancer Cells. Int. J. Oncol. 2004; 25:1631-9; and Sen S., Zhou H., Zhang R. D., et al. Amplification/Overexpression of A Mitotic Kinase Gene in Human Bladder cancer. J. Natl. Cancer Inst. 2002; 94:1320-9.
  • Involvement of Aurora Kinase in esophageal cancer is reported in Tong T., Zhong Y., Kong J., et al. Overexpression of Aurora-A Contributes to Malignant Development of Human Esophageal Squamous Cell Carcinoma. Clin. Cancer Res. 2004; 10:7304-10; Yang S. B., Zhou X. B., Zhu H. X., et al Amplification and Overexpression of Aurora-A in Esophageal Squamous Cell Carcinoma. Oncol. Rep. 2007; 17:1083-8; and Kimura M. T., Mori T., Conroy J., et al. Two Functional Coding Single Nucleotide Polymorphisms in STK15 (Aurora-A) Coordinately Increase Esophageal Cancer Risk. Cancer Res 2005; 65:3548-54.
  • Involvement of Aurora Kinase in brain cancer is reported in Araki K., Nozaki K., Ueba T., Tatsuka M., Hashimoto N. High Expression of Aurora-B/Aurora and Ip11-Like Midbody-Associated Protein (AIM-1) in Astrocytomas. J. Neurooncol. 2004; 67:53-64; Zeng W. F., Navaratne K., Prayson R. A., Weil R. J. Aurora B Expression Correlates with Aggressive Behaviour in Glioblastoma Multiforme. J. Clin. Pathol. 2007; 60:218-21; Reichardt W., Jung V., Brunner C., et al. The Putative Serine/Threonine Kinase Gene STK15 on Chromosome 20q13.2 is Amplified In Human Gliomas. Oncol. Rep. 2003; 10:1275-9; Klein A., Reichardt W., Jung V., Zang K. D., Meese E., Urbschat S. Overexpression and Amplification of STK15 Inhuman Gliomas. Int. J. Oncol. 2004; 25:1789-94; and Neben K., Korshunov A., Benner A., et al. Microarray Based Screening for Molecular Markers Nmedulloblastoma Revealed STK15 as Independent Predictor for Survival. Cancer Res 2004; 64:3103-11.
  • Involvement of Aurora Kinase in liver cancer is reported in Jeng Y. M., Peng S. Y., Lin C. Y., Hsu H. C. Overexpression and Amplification of Aurora-A in Hepatocellular Carcinoma. Clin. Cancer Res. 2004; 10:2065-71.
  • Involvement of Aurora Kinase in head and neck cancer is reported in Zhao X., Li F. C., Li Y. H., et al. [Mutation of p53 and Overexpression Of STK15 in Laryngeal Squamous-Cell Carcinoma]. Zhonghua Zhong Liu Za Zhi 2005; 27:134-7; Li F. C., Li Y. H., Zhao X., et al. [Deletion of p15 and p16 Genes and Overexpression of STK15 Gene in Human Laryngeal Squamous Cell Carcinoma]. Zhonghua Yi Xue Za Zhi 2003; 83:316-9; Reiter R., Gais P., Jutting U., et al. Aurora Kinase A Messenger RNA Overexpression is Correlated with Tumor Progression and Shortened Survival in Head and Neck Squamous Cell Carcinoma. Clin. Cancer Res. 2006; 12:5136-41; Qi G., Ogawa I., Kudo Y., et al. Aurora-B Expression and Its Correlation with Cell Proliferation and Metastasis in Oral Cancer. Virchows Arch 2007; 450:297-302; and Tatsuka M., Sato S., Kitajima S., et al. Overexpression of Aurora-A Potentiates HRAS-mediated Oncogenic Transformation and is Implicated in Oral Carcinogenesis. Oncogene 2005; 4:1122-7.
  • Involvement of Aurora Kinase in thyroid cancer is reported in Sorrentino R., Libertini S., Pallante P. L., et al. Aurora B Overexpression Associates with the Thyroid Carcinoma Undifferentiated Phenotype and is Required for Thyroid Carcinoma Cell Proliferation. J. Clin. Endocrinol. Metab. 2005; 90:928-35.
  • Involvement of Aurora Kinase in ovarian cancer is reported in Lassmann S., Shen Y., Jutting U., et al. Predictive Value of Aurora-A/STK15 Expression for Late Stage Epithelial Ovarian Cancer Patients Treated By Adjuvant Chemotherapy. Clin Cancer Res 2007; 13:4083-91; and Landen C. N., Jr., Lin Y. G., Immaneni A., et al. Overexpression of the Centrosomal Protein Aurora-A Kinase is Associated with Poor Prognosis in Epithelial Ovarian Cancer Patients. Clin. Cancer Res. 2007; 13:4098-104.
  • Involvement of Aurora Kinase in renal cancer is reported in Kurahashi T., Miyake H., Hara I., Fujisawa M. Significance of Aurora-A Expression in Renal Cell Carcinoma. Urol. Oncol. 2007; 25:128-33.
  • Involvement of Aurora Kinase in endometrium cancer is reported in Moreno-Bueno G., Sanchez-Estevez C., Cassia R., et al. Differential Gene Expression Profile in Endometrioid and Nonendometrioid Endometrial Carcinoma: STK15 is Frequently Overexpressed and Amplified in Nonendometrioid Carcinomas. Cancer Res. 2003; 63:5697-702.
  • Involvement of Aurora Kinase in gastric cancer is reported in Ju H., Cho H., Kim Y. S., et al. Functional Polymorphism 57Val>Ile of Aurora Kinase A Associated with Increased Risk of Gastric Cancer Progression. Cancer Lett. 2006; 242:273-9.
  • Involvement of Aurora Kinase in colon cancer is reported in Nishida N., Nagasaka T., Kashiwagi K., Boland C. R., Goel A. High Copy Amplification of the Aurora-A Gene is Associated with Chromosomal Instability Phenotype in Human Colorectal Cancers. Cancer Biol. Ther. 2007; 6:525-33; Bischoff J. R., Anderson L., Zhu Y., et al. A Homologue of Drosophila Aurora Kinase is Oncogenic and Amplified In Human Colorectal Cancers. EMBO J. 1998; 17:3052-65; Chen J., Sen S., Amos C. I., et al. Association Between Aurora-A Kinase Polymorphisms and Age of Onset of Hereditary Nonpolyposis Colorectal Cancer in a Caucasian Population. Mol. Carcinog. 2007; 46:249-56; Hienonen T., Salovaara R., Mecklin J. P., Jarvinen H., Karhu A., Aaltonen L. A. Preferential Amplification of AURKA 91A (Ile31) in Familial Colorectal Cancers. Int. J. Cancer 2006; 118:505-8; and Ewart-Toland A., Briassouli P., de Koning J. P., et al. Identification of Stk6/STK15 as a Candidate Low-Penetrance Tumor-Susceptibility Gene in Mouse and Human. Nat. Genet. 2003; 34:403-12.
  • Involvement of Aurora Kinase in cancer is reported in Lin, Y. S., et al., Gene Expression Profiles of the Aurora Family Kinases. Gene Expr., 2006. 13 (1): p. 15-26; and Ewart-Toland A., Dai Q., Gao Y. T., et al. Aurora-A/STK15 T+91A is a General Low Penetrance Cancer Susceptibility Gene: A Meta-Analysis of Multiple Cancer Types. Carcinogenesis 2005; 26:1368-73.
  • Involvement of KDR (VEGFR2) in cancer and studies using VEGF-targeted therapy is reported in Ellis, Lee M., Hicklin, Daniel J. VEGF-Targeted Therapy:Mechanisms Of Anti-Tumor Activity. Nature Reviews Cancer 2008; 8:579-591.
  • Involvement of Aurora-kinases in bladder cancer, breast cancer, cervical cancer, colon cancer, endometrial cancer, esophageal cancer, lung cancer, ovarian cancer, pancreatic cancer, prostate cancer, rectal cancer, skin cancer, stomach cancer and thyroid cancer is reported in Nature Reviews/Cancer, Vol. 4 December, 2004.
  • Compounds of this invention may be made by synthetic chemical processes, examples of which are shown herein. It is meant to be understood that the order of the steps in the processes may be varied, that reagents, solvents and reaction conditions may be substituted for those specifically mentioned, and that vulnerable moieties may be protected and deprotected, as necessary. For example, the coupling reaction between 3-bromothieno[3,2-c]pyridin-4-amine and the substituted 4,4,5,5-tetramethyl-2-phenyl-1,3,2-dioxaborolane can be performed prior to or after the formation of the urea linkage.
  • Protecting groups for C(O)OH moieties include, but are not limited to, acetoxymethyl, allyl, benzoylmethyl, benzyl, benzyloxymethyl, tert-butyl, tert-butyldiphenylsilyl, diphenylmethyl, cyclobutyl, cyclohexyl, cyclopentyl, cyclopropyl, diphenylmethylsilyl, ethyl, para-methoxybenzyl, methoxymethyl, methoxyethoxymethyl, methyl, methylthiomethyl, naphthyl, para-nitrobenzyl, phenyl, n-propyl, 2,2,2-trichloroethyl, triethylsilyl, 2-(trimethylsilyl)ethyl, 2-(trimethylsilyl)ethoxymethyl, triphenylmethyl and the like.
  • Protecting groups for C(O) and C(O)H moieties include, but are not limited to, 1,3-dioxylketal, diethylketal, dimethylketal, 1,3-dithianylketal, O-methyloxime, O-phenyloxime and the like.
  • Protecting groups for NH moieties include, but are not limited to, acetyl, alanyl, benzoyl, benzyl (phenylmethyl), benzylidene, benzyloxycarbonyl (Cbz), tert-butoxycarbonyl (Boc), 3,4-dimethoxybenzyloxycarbonyl, diphenylmethyl, diphenylphosphoryl, formyl, methanesulfonyl, para-methoxybenzyloxycarbonyl, phenylacetyl, phthaloyl, succinyl, trichloroethoxycarbonyl, triethylsilyl, trifluoroacetyl, trimethylsilyl, triphenylmethyl, triphenylsilyl, para-toluenesulfonyl and the like.
  • Protecting groups for OH and SH moieties include, but are not limited to, acetyl, allyl, allyloxycarbonyl, benzyloxycarbonyl (Cbz), benzoyl, benzyl, tert-butyl, tert-butyldimethylsilyl, tert-butyldiphenylsilyl, 3,4-dimethoxybenzyl, 3,4-dimethoxybenzyloxycarbonyl, 1,1-dimethyl-2-propenyl, diphenylmethyl, formyl, methanesulfonyl, methoxyacetyl, 4-methoxybenzyloxycarbonyl, para-methoxybenzyl, methoxycarbonyl, methyl, para-toluenesulfonyl, 2,2,2-trichloroethoxycarbonyl, 2,2,2-trichloroethyl, triethylsilyl, trifluoroacetyl, 2-(trimethylsilyl)ethoxycarbonyl, 2-trimethylsilylethyl, triphenylmethyl, 2-(triphenylphosphonio)ethoxycarbonyl and the like.
  • The following abbreviations have the meanings indicated.
  • ADDP means 1,1′-(azodicarbonyl)dipiperidine; AD-mix-β means a mixture of (DHQD)2PHAL, K3Fe(CN)6, K2CO3 and K2SO4); AIBN means 2,2′-azobis(2-methylpropionitrile); 9-BBN means 9-borabicyclo(3.3.1)nonane; Cp means cyclopentadiene; (DHQD)2PHAL means hydroquinidine 1,4-phthalazinediyl diethyl ether; DBU means 1,8-diazabicyclo(5.4.0)undec-7-ene; DIBAL means diisobutylaluminum hydride; DIEA means diisopropylethylamine; DMAP means N,N-dimethylaminopyridine; DME means 1,2-dimethoxyethane; DMF means N,N-dimethylformamide; dmpe means 1,2-bis(dimethylphosphino)ethane; DMSO means dimethylsulfoxide; dppa means diphenylphosphoryl azide; dppb means 1,4-bis(diphenylphosphino)butane; dppe means 1,2-bis(diphenylphosphino)ethane; dppf means 1,1′-bis(diphenylphosphino)ferrocene; dppm means 1,1-bis(diphenylphosphino)methane; EDAC means 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide; Fmoc means fluorenylmethoxycarbonyl; HATU means O-(7-azabenzotriazol-1-yl)-N,N′N′N′-tetramethyluronium hexafluorophosphate; HMPA means hexamethylphosphoramide; HOAT means 1-hydroxy-7-azabenzotriazole; IPA means isopropyl alcohol; LDA means lithium diisopropylamide; LHMDS means lithium bis(hexamethyldisilylamide); MP-BH3 means macroporus triethylammonium methylpolystyrene cyanoborohydride; LAH means lithium aluminum hydride; NCS means N-chlorosuccinimide; PyBOP means benzotriazol-1-yloxytripyrrolidinophosphonium hexafluorophosphate; TBTU means O-benzotriazol-1-yl-N,N,N′, N′-tetramethyluronium tetrafluoroborate; TDA-1 means tris(2-(2-methoxyethoxy)ethyl)amine; TEA means triethylamine; TFA means trifluoroacetic acid; THF means tetrahydrofuran; NCS means N-chlorosuccinimide; NMM means N-methylmorpholine; NMP means N-methylpyrrolidine; PPh3 means triphenylphosphine.
  • Example 1 N-(4-{4-amino-7-[1-(2-hydroxyethyl)-1H-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N′-(3-fluorophenyl)urea Example 1A 3-(4-aminophenyl)-7-iodothieno[3,2-c]pyridin-4-amine
  • A suspension of 3-bromothieno[3,2-c]pyridin-4-amine (13.7 g, 59.7 mmol), tert-butyl 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenylcarbamate (20 g, 62.7 mmol), tetrakis(triphenylphosphine)palladium(0) (2.5 g, 2.1 mmol) and Na2CO3 (13.3 g, 125 mmol) in tetrahydrofuran (150 mL), methanol (40 mL) and water (80 mL) was degassed, then stirred at reflux overnight. The reaction mixture was cooled to room temperature, then partitioned between ethyl acetate and water. The aqueous layer was extracted with additional ethyl acetate and the combined organics were dried (using MgSO4), filtered and the filtrate was concentrated. The residue was purified via silica gel chromatography eluting with 50 to 70% ethyl acetate-hexanes to give crude tert-butyl 4-(4-aminothieno[3,2-c]pyridin-3-yl)phenylcarbamate. A solution of the crude product (59.7 mmol based on 100% yield) in N,N-dimethylformamide (80 mL) was treated with N-iodosuccinimide (13.5 g, 59.7 mol—added in portions) and the resulting dark solution was stirred at room temperature for 2 hours, then partitioned between water (500 mL) and ethyl acetate (100 mL) with NaCl added to facilitate layer separation. The aqueous layer was extracted with additional ethyl acetate (2×75 mL) and the combined organics were washed with sodium thiosulfate (3×20 mL) and brine (50 mL), and then dried (using MgSO4), filtered, and concentrated. The crude material was treated with TFA (20 mL) and CH2Cl2 (5 mL), stirred at room temperature for 2 hours, concentrated under a stream of nitrogen, then concentrated in vacuo. The solid was dissolved in water (100 mL), carefully treated with solid Na2CO3 until gas evolution ceased and filtered, washing with additional water. The solid collected was dried to provide the title compound as a solid (contaminated by ca. 10% mole PPh3).
  • Example 1B 2-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazol-1-yl)ethanol
  • 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (9.66 g, 49.8 mmol), 1,3-dioxolan-2-one (21 g, 238 mmol) and cesium carbonate (16 g, 49.1 mmol) were combined in a 100 mL round bottom flask. The reaction was warmed from room temperature to 100° C. in an oil bath, by which time the carbonate had melted and served as the solvent for the reaction, which remained a slurry. After heating for 3.5 hours, the reaction was cooled to room temperature and diluted with ethyl acetate, then filtered through Celite washing repeatedly with ethyl acetate. The filtrate was concentrated, then purified by chromatography on an Analogix® Intelliflash™ purification system using a SF60-200 g column at a flow rate of 80 mL/min, eluting as follows: 5 minutes at 20% ethyl acetate/hexanes, then ramped from 40% to 90% ethyl acetate/hexanes over 35 minutes, then 100% ethyl acetate for another 20 minutes, to provide the title compound.
  • Example 1C 2-(4-(4-amino-3-(4-aminophenyl)thieno[3,2-c]pyridin-7-yl)-1H-pyrazol-1-yl)ethanol
  • EXAMPLE 1A (6 g, 16.34 mmol), EXAMPLE 1B (4.8 g, 20.16 mmol), PdCl2(dppf) (1.2 g, 1.640 mmol) and sodium carbonate (4.6 g, 43.4 mmol) were combined in tetrahydrofuran (400 mL), methanol (80 mL) and water (80 mL), and the reaction was degassed by bubbling N2 through the mixture for 1 hour. The reaction was then heated to 80° C. for 2 hours, then allowed to cool and diluted with 300 mL ethyl acetate. The mixture was partitioned with H2O (500 mL) and the aqueous layer was extracted with ethyl acetate (2×300 mL). The combined organic extracts were washed with brine, dried (Na2SO4), filtered through a pad of Celite, concentrated to a total volume of about 200 mL, then left standing overnight. The solid that formed was collected by filtration, providing the title compound.
  • Example 1D N-(4-{4-amino-7-[1-(2-hydroxyethyl)-1H-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N′-(3-fluorophenyl)urea
  • EXAMPLE 1C (2 g, 5.69 mmol) was dissolved in N,N-dimethylformamide (80 ml) and the flask was chilled in a −20° C. bath. 1-fluoro-3-isocyanatobenzene (0.715 ml, 6.26 mmol) was added dropwise and the reaction was allowed to warm slowly to room temperature and stirred overnight. The reaction was diluted with water (500 mL) and ethyl acetate (75 mL) and stirred to digest for 1 hour. The mixture was then placed in a separatory funnel. After the layers were allowed to separate, the lower aqueous layer was drained off. Near the interface of the two layers there was a significant amount of precipitate. That material and the organic layer were filtered, giving a solid. The solid was rinsed with ethyl acetate and dried to give the title compound. 1H NMR (300 MHz, DMSO-d6) δ ppm 3.80 (q, J=5.65 Hz, 2H) 4.23 (t, J=5.59 Hz, 2H) 4.96 (t, J=5.09 Hz, 1H) 5.42 (s, 2H) 6.80 (td, J=8.31, 2.37 Hz, 1H) 7.15 (dd, J=8.14, 1.02 Hz, 1H) 7.32 (td, J=8.22, 6.95 Hz, 1H) 7.41 (d, J=8.81 Hz, 2H) 7.51 (ddd, J=11.70, 2.37, 2.20 Hz, 1H) 7.50 (s, 1H) 7.61 (d, J=8.81 Hz, 2H) 7.91 (d, J=1.02 Hz, 1H) 8.05 (s, 1H) 8.15 (d, J=0.68 Hz, 1H) 8.96 (s, 1H) 8.99 (s, 1H); MS (ESI(+)) m/z 489.1 (M+H)+.
  • Example 2 N-(4-{4-amino-7-[1-(2-hydroxyethyl)-1H-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N′-[4-(difluoromethoxy)phenyl]urea
  • The title compound was prepared by substituting 1-(difluoromethoxy)-4-isocyanatobenzene for 1-fluoro-3-isocyanatobenzene in EXAMPLE 1D. 1H NMR (300 MHz, DMSO-d6) δ ppm 3.80 (q, J=5.4 Hz, 2H) 4.23 (t, J=5.6 Hz, 2H) 4.95 (t, J=5.4 Hz, 1H) 5.41 (br s, 2H) 7.13 (t, J=74.4 Hz, 1H) 7.13 (d, J=8.8 Hz, 2H) 7.39 (d, J=8.5 Hz, 2H) 7.48-7.55 (m, 3 H) 7.61 (d, J=8.5 Hz, 2H) 7.90 (s, 1H) 8.05 (s, 1H) 8.15 (s, 1H) 8.85 (s, 1H) 8.91 (s, 1H); MS (ESI(+)) m/e 537 (M+H)+.
  • Example 3 N-[4-(4-amino-7-{1-[(2S)-2-hydroxypropyl]-1H-pyrazol-4-yl}thieno[3,2-c]pyridin-3-yl)phenyl]-N′-(3-methylphenyl)urea
  • The title compound was prepared following the procedures of EXAMPLES 1B-1D except substituting (S)-4-methyl-1,3-dioxolan-2-one for 1,3-dioxolan-2-one for and 1-isocyanato-3-methylbenzene for 1-fluoro-3-isocyanatobenzene in EXAMPLES 1B and 1D, respectively. 1H NMR (300 MHz, DMSO-d6) δ ppm 1.09 (d, J=6.10 Hz, 3H) 2.29 (s, 3H) 4.01-4.12 (m, 3H) 4.96 (d, J=4.75 Hz, 1H) 5.43 (s, 2H) 6.81 (d, J=7.46 Hz, 1H) 7.17 (t, J=7.80 Hz, 1H) 7.26 (app d, J=9.15 Hz, 1H) 7.32 (s, 1H) 7.39 (d, J=8.48 Hz, 2H) 7.50 (s, 1H) 7.61 (d, J=8.81 Hz, 2H) 7.90 (d, J=0.68 Hz, 1H) 8.05 (s, 1H) 8.13 (d, J=1.02 Hz, 1H) 8.66 (s, 1H) 8.86 (s, 1H); MS (ESI(+)) m/z 499.2 (M+H)+.
  • Example 4 N-(4-{4-amino-7-[1-(2-hydroxy-2-methylpropyl)-1H-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N′-(4-methoxyphenyl)urea Example 4A 2-methyl-1-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazol-1-yl)propan-2-ol
  • A mixture of 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (500 mg, 2.57 mmol), Cs2CO3 (840 mg, 2.57 mmol) and 2,2-dimethyloxirane (2 mL) was heated in a sealed vial at 120° C. for 3 minutes with stirring in a Smith Synthesizer microwave oven (at 300 W), then allowed to cool and diluted with CH2Cl2. The resulting suspension was filtered, and the filtrate was concentrated to give the title compound.
  • Example 4B N-(4-{4-amino-7-[1-(2-hydroxy-2-methylpropyl)-1H-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N′-(4-methoxyphenyl)urea
  • The title compound was prepared following the procedures of EXAMPLES 1C-1D except substituting EXAMPLE 4A for EXAMPLE 1B and 1-isocyanato-4-methoxybenzene for 1-fluoro-3-isocyanatobenzene in EXAMPLES 1C and EXAMPLE 1D, respectively. 1H NMR (300 MHz, DMSO-d6) δ ppm 1.12 (s, 6H) 3.72 (s, 3H) 4.11 (s, 2H) 4.76 (s, 1H) 5.43 (s, 2H) 6.88 (d, J=9.15 Hz, 2H) 7.38 (d, J=8.81 Hz, 4H) 7.49 (s, 1H) 7.60 (d, J=8.81 Hz, 2H) 7.89 (d, J=0.68 Hz, 1H) 8.06 (s, 1H) 8.11 (d, J=0.68 Hz, 1H) 8.55 (s, 1H) 8.80 (s, 1H); MS (ESI(+)) m/z 529.3 (M+H)+.
  • Example 5 N-[4-(4-amino-7-{1-[(2S)-2,3-dihydroxypropyl]-1H-pyrazol-4-yl}thieno[3,2-c]pyridin-3-yl)phenyl]-N′-(4-methoxyphenyl)urea Example 5A (S)-1-((2,2-dimethyl-1,3-dioxolan-4-yl)methyl)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole
  • A mixture of (R)-(2,2-dimethyl-1,3-dioxolan-4-yl)methyl methanesulfonate (1.08, 5.15 mmol), 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (1 g, 5.15 mmol) and NaH (262 mg, 10.9 mmol) in N,N-dimethylformamide (25 mL) was heated at 90° C. for 3 hours, then allowed to cool to room temperature., quenched with water and extracted with ethyl acetate (3×). The combined organics were washed with brine, concentrated and the residue was purified by chromatography on an Analogix® Intelliflash™ purification system using a SF25-25 g column at a flow rate of 80 mL/minute, eluting with 0% to 30% ethyl acetate:hexanes over 30 minutes to give the title compound.
  • Example 5B (S)-1-(4-(4-amino-7-(1-((2,2-dimethyl-1,3-dioxolan-4-yl)methyl)-1H-pyrazol-4-yl)thieno[3,2-c]pyridin-3-yl)phenyl)-3-(4-methoxyphenyl)urea
  • The title compound was prepared following the procedures of EXAMPLES 1C-1D except substituting EXAMPLE 5A for 1B and 1-isocyanato-4-methoxybenzene for 1-fluoro-3-isocyanatobenzene in EXAMPLES 1B and 1D, respectively.
  • Example 5C N-[4-(4-amino-7-{1-[(2S)-2,3-dihydroxypropyl]-1H-pyrazol-4-yl}thieno[3,2-c]pyridin-3-yl)phenyl]-N′-(4-methoxyphenyl)urea
  • A solution of EXAMPLE 5B (44 mg, 0.077 mL) in tetrahydrofuran (2 mL) was treated with 2N HCl (1 mL), then stirred at room temperature for 18 hours. The resulting suspension was filtered and the solid was collected and dried to give the title compound. 1H NMR (300 MHz, DMSO-d6) δ ppm 3.72 (s, 3H) 3.32-3.45 (m, 4H) 3.84-3.91 (m, 1H) 4.12 (dd, J=13.90, 7.80 Hz, 1H) 4.34 (dd, J=13.90, 3.73 Hz, 1H) 6.89 (d, J=9.15 Hz, 2H) 6.89-6.93 (m, 2H) 7.38 (d, J=9.15 Hz, 2H) 7.45 (d, J=8.81 Hz, 2H) 7.65 (d, J=8.81 Hz, 2H) 7.94 (s, 1H) 7.99 (d, J=0.68 Hz, 1H) 8.06 (s, 1H) 8.28 (d, J=0.68 Hz, 1H) 8.72 (s, 1H) 9.05 (s, 1H); MS (ESI(+)) m/z 531.2 (M+H)+.
  • Example 6 N-{-4-[4-amino-7-(1H-pyrazol-4-yl)thieno[3,2-c]pyridin-3-yl]phenyl}-N′-(3-methylphenyl)urea
  • Example 6 is described as EXAMPLE 703 in WO 2005/10009.
  • The foregoing is meant to illustrate the invention but not to limit it. Variations and changes obvious to one skilled in the art are intended to be within the scope of the invention as defined in the claims.

Claims (5)

1. A compound having Formula I
Figure US20100144783A1-20100610-C00003
or a therapeutically acceptable salt thereof,
wherein
R1 is hydroxyalkyl;
R2 is selected from the group consisting of alkoxy, alkyl, halo, and haloalkoxy; and
R3 is hydrogen or alkyl.
2. The compound of claim 1 which is
N-(4-{4-amino-7-[1-(2-hydroxyethyl)-1H-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N′-(3-fluorophenyl)urea;
N-(4-{4-amino-7-[1-(2-hydroxyethyl)-1H-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N′-[4-(difluoromethoxy)phenyl]urea;
N-[4-(4-amino-7-{1-[(2S)-2-hydroxypropyl]-1H-pyrazol-4-yl}thieno[3,2-c]pyridin-3-yl)phenyl]-N′-(3-methylphenyl)urea;
N-(4-{4-amino-7-[1-(2-hydroxy-2-methylpropyl)-1H-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N′-(4-methoxyphenyl)urea; or
N-[4-(4-amino-7-{1-[(2S)-2,3-dihydroxypropyl]-1H-pyrazol-4-yl}thieno[3,2-c]pyridin-3-yl)phenyl]-N′-(4-methoxyphenyl)urea.
3. The compound of claim 1 or a pharmaceutically acceptable salt thereof, wherein said compound does not inhibit CYP3A4.
4. A composition comprising an excipient and a therapeutically effective amount of a compound of claim 1.
5. A method of treating cancer in a mammal comprising administering thereto a therapeutically acceptable amount of a compound of claim 1.
US12/632,183 2008-12-05 2009-12-07 Kinase inhibitors with improved cyp safety profile Abandoned US20100144783A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US12/632,183 US20100144783A1 (en) 2008-12-05 2009-12-07 Kinase inhibitors with improved cyp safety profile
US13/585,333 US8722890B2 (en) 2008-12-05 2012-08-14 Thieno[3,2-C]pyridine kinase inhibitors with improved CYP safety profile

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US12028108P 2008-12-05 2008-12-05
US22376009P 2009-07-08 2009-07-08
US12/632,183 US20100144783A1 (en) 2008-12-05 2009-12-07 Kinase inhibitors with improved cyp safety profile

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/585,333 Continuation US8722890B2 (en) 2008-12-05 2012-08-14 Thieno[3,2-C]pyridine kinase inhibitors with improved CYP safety profile

Publications (1)

Publication Number Publication Date
US20100144783A1 true US20100144783A1 (en) 2010-06-10

Family

ID=41559043

Family Applications (2)

Application Number Title Priority Date Filing Date
US12/632,183 Abandoned US20100144783A1 (en) 2008-12-05 2009-12-07 Kinase inhibitors with improved cyp safety profile
US13/585,333 Active US8722890B2 (en) 2008-12-05 2012-08-14 Thieno[3,2-C]pyridine kinase inhibitors with improved CYP safety profile

Family Applications After (1)

Application Number Title Priority Date Filing Date
US13/585,333 Active US8722890B2 (en) 2008-12-05 2012-08-14 Thieno[3,2-C]pyridine kinase inhibitors with improved CYP safety profile

Country Status (26)

Country Link
US (2) US20100144783A1 (en)
EP (1) EP2373662B1 (en)
JP (1) JP5588458B2 (en)
KR (1) KR101639642B1 (en)
CN (1) CN102239171B (en)
AR (1) AR074481A1 (en)
AU (1) AU2009322270B2 (en)
BR (1) BRPI0921392A2 (en)
CA (1) CA2743592A1 (en)
CL (1) CL2011001312A1 (en)
CO (1) CO6382131A2 (en)
DO (1) DOP2011000153A (en)
EC (1) ECSP11011172A (en)
ES (1) ES2524966T3 (en)
HK (1) HK1159628A1 (en)
IL (1) IL212716A (en)
MX (1) MX2011005943A (en)
MY (1) MY179042A (en)
NZ (1) NZ592714A (en)
PA (1) PA8852401A1 (en)
PE (1) PE20110830A1 (en)
RU (2) RU2480472C2 (en)
TW (2) TWI441827B (en)
UA (1) UA103351C2 (en)
UY (1) UY32291A (en)
WO (1) WO2010065825A2 (en)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100069371A1 (en) * 2003-07-24 2010-03-18 Abbott Laboratories Thienopyridine and furopyridine kinase inhibitors
US20110306632A1 (en) * 2010-06-09 2011-12-15 Abbott Laboratories Solid Dispersions Containing Kinase Inhibitors
US8357804B2 (en) 2010-06-09 2013-01-22 Abbvie Inc. Crystal forms of kinase inhibitors
US8436179B2 (en) 2011-07-20 2013-05-07 Abbvie Inc. Kinase inhibitor with improved solubility profile
WO2013090666A1 (en) 2011-12-14 2013-06-20 Abbvie Inc. Compositions containing kinase inhibitors
US8633317B2 (en) 2010-06-09 2014-01-21 Abbvie Inc. Crystalline salts of thieno[3,2-c]pyridine kinase inhibitors with improved cpy safety profile
US8633217B2 (en) 2010-06-09 2014-01-21 Abbvie Inc. Crystal forms of kinase inhibitors
US8911725B2 (en) 2010-06-23 2014-12-16 University Of Central Florida Research Foundation, Inc. Co-targeting of aurora A kinase and LIM kinase 1 for cancer therapy
WO2015157360A1 (en) 2014-04-08 2015-10-15 Abbvie Inc. Processes to make protein kinase inhibitors
WO2015200635A1 (en) 2014-06-25 2015-12-30 Abbvie Inc. N-(4-{4-AMINO-7-[1-(2-HYDROXYETHYL)-1H-PYRAZOL-4-yl]THIENO[3,2-c]PYRIDIN-3-YL}PHENYL)-N'-(3-FLUOROPHENYL)UREA DOCUSATE

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MX2011005943A (en) * 2008-12-05 2011-06-27 Abbott Lab Thieno [3, 2-c] pyridine derivatives as kinase inhibitors for use in the treatment of cancer.
EP2885292A4 (en) * 2012-08-17 2015-07-01 Cancer Therapeutics Crc Pty Ltd Vegfr3 inhibitors
CN104736533B (en) * 2012-08-17 2016-12-07 癌症治疗合作研究中心有限公司 Vegfr3 inhibitor
CN103012428A (en) * 2013-01-08 2013-04-03 中国药科大学 4-(five-membered heterocycle pyrimidin/substituted pyridine) amino-1H-3-pyrazolecarboxamide CDK (cyclin dependent kinase)/Aurora dual inhibitor and application thereof
TW202038957A (en) 2018-12-21 2020-11-01 日商第一三共股份有限公司 Combination of antibody-drug conjugate and kinase inhibitor
CA3141405A1 (en) * 2019-06-12 2020-12-17 H. Charles Manning Amino acid transport inhibitors and the uses thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6500855B1 (en) * 1998-12-23 2002-12-31 Bristol-Myers Squibb Pharma Company Thrombin or factor Xa inhibitors
US20050043347A1 (en) * 2003-07-24 2005-02-24 Patrick Betschmann Thienopyridine and furopyridine kinase inhibitors
US7592352B2 (en) * 2003-05-06 2009-09-22 Smithkline Beecham Corporation Substituted thieno and furo-pyridines

Family Cites Families (38)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB1188529A (en) 1966-06-09 1970-04-15 Wellcome Found Pyramidine Derivatives
US4357535A (en) 1980-04-30 1982-11-02 North American Philips Corporation Apparatus for inspecting hand-held articles and persons carrying same
GB8307154D0 (en) 1983-03-15 1983-04-20 Mk Electric Ltd Terminals
US4767766A (en) 1987-01-30 1988-08-30 Merck & Co., Inc. Derivatives of 3-hydroxyazabenzo(B)thiophene useful as 5-lipoxygenase inhibitors
EP0300688A1 (en) 1987-07-21 1989-01-25 FISONS plc Pyrrole derivatives, process for their preparation and pharmaceutical compositions containing them
JPH0436810Y2 (en) 1987-12-29 1992-08-31
US4843949A (en) 1988-08-29 1989-07-04 Pneumo Abex Corporation Fluid control valve with variable pressure gain
GB8912498D0 (en) 1989-05-31 1989-07-19 De Beers Ind Diamond Diamond growth
EP0438261A3 (en) 1990-01-16 1992-02-26 Takeda Chemical Industries, Ltd. Condensed heterocyclic glutamic acid derivatives, their production and use
DE4129603A1 (en) 1991-09-06 1993-03-11 Thomae Gmbh Dr K CONDENSED 5-LOW HETEROCYCLES, METHOD FOR THE PRODUCTION THEREOF, AND MEDICAMENTS CONTAINING THESE COMPOUNDS
EP0630031B1 (en) 1993-06-16 2002-08-14 E.I. Du Pont De Nemours And Company Water cleanable thick film paste composition
IL112759A0 (en) 1994-02-25 1995-05-26 Khepri Pharmaceuticals Inc Novel cysteine protease inhibitors
DE29511756U1 (en) 1995-07-20 1995-09-28 Ferco Int Usine Ferrures Wing-side corner bearing fitting for tilt and turn windows
AR004010A1 (en) 1995-10-11 1998-09-30 Glaxo Group Ltd HETERO CYCLIC COMPOUNDS
DE19620508A1 (en) 1996-05-22 1997-11-27 Hoechst Ag Sulfur-containing heterocyclic bradykinin antagonists, process for their preparation and their use
CZ295822B6 (en) 1997-04-22 2005-11-16 Neurosearch A/S Substituted phenyl derivatives, process of their preparation and use as well as pharmaceutical composition containing such derivatives
ATE267200T1 (en) 1997-10-20 2004-06-15 Hoffmann La Roche BICYCLIC KINASE INHIBITORS
AU7491400A (en) 1999-09-17 2001-04-17 Abbott Gmbh & Co. Kg Kinase inhibitors as therapeutic agents
US6608053B2 (en) 2000-04-27 2003-08-19 Yamanouchi Pharmaceutical Co., Ltd. Fused heteroaryl derivatives
US20020004511A1 (en) 2000-06-28 2002-01-10 Luzzio Michael Joseph Thiophene derivatives useful as anticancer agents
EP2071827A3 (en) 2000-12-15 2010-08-25 BRITISH TELECOMMUNICATIONS public limited company Transmission and reception of audio and/or video material
WO2002071827A2 (en) 2001-03-14 2002-09-19 Eli Lilly And Company Retinoid x receptor modulators
GB0115109D0 (en) 2001-06-21 2001-08-15 Aventis Pharma Ltd Chemical compounds
EP1425284A2 (en) 2001-09-11 2004-06-09 Smithkline Beecham Corporation Furo- and thienopyrimidine derivatives as angiogenesis inhibitors
US20050019424A1 (en) 2001-12-21 2005-01-27 Adams Paul E. Anti-angiogenesis combination therapies comprising pyridazine or pyridine derivatives
US7265128B2 (en) 2003-01-17 2007-09-04 Merck & Co., Inc. 3-amino-4-phenylbutanoic acid derivatives as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
DE10303311A1 (en) 2003-01-28 2004-07-29 Basf Ag Production of di-, oligo- and polymers of dicarbonyl compounds as aqueous formulation or powder, used for tanning and as preservative, involves acid-catalyzed condensation of 2-substituted heterocyclic compound in water
US20050026944A1 (en) * 2003-07-24 2005-02-03 Patrick Betschmann Thienopyridine and furopyridine kinase inhibitors
US7419978B2 (en) 2003-10-22 2008-09-02 Bristol-Myers Squibb Company Phenyl-aniline substituted bicyclic compounds useful as kinase inhibitors
GB0420719D0 (en) 2004-09-17 2004-10-20 Addex Pharmaceuticals Sa Novel allosteric modulators
US7183300B2 (en) 2004-11-11 2007-02-27 Summers Michael F Inhibitors of HIV-1 capsid formation: substituted aryl aminomethyl thiazole ureas and analogues thereof
KR101078968B1 (en) 2005-05-20 2011-11-01 메틸진 인코포레이티드 Inhibitors of vegf receptor and hgf receptor signaling
US7566721B2 (en) 2005-08-08 2009-07-28 Osi Pharmaceuticals, Inc. Substituted thienol[2,3-d]pyrimidines as kinase inhibitors
MX2008002207A (en) 2005-08-16 2008-03-27 Memory Pharm Corp Phosphodiesterase 10 inhibitors.
EP1951728A4 (en) 2005-11-04 2011-04-20 Glaxosmithkline Llc Thienopyridine b-raf kinase inhibitors
CN101336244B (en) * 2005-12-08 2011-11-30 雅培制药有限公司 9-membered heterobicyclic compounds as inhibitors of protein kinases
JP2009544625A (en) 2006-07-20 2009-12-17 メーメット・カーラマン Benzothiophene inhibitors of RHO kinase
MX2011005943A (en) * 2008-12-05 2011-06-27 Abbott Lab Thieno [3, 2-c] pyridine derivatives as kinase inhibitors for use in the treatment of cancer.

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6500855B1 (en) * 1998-12-23 2002-12-31 Bristol-Myers Squibb Pharma Company Thrombin or factor Xa inhibitors
US7592352B2 (en) * 2003-05-06 2009-09-22 Smithkline Beecham Corporation Substituted thieno and furo-pyridines
US20050043347A1 (en) * 2003-07-24 2005-02-24 Patrick Betschmann Thienopyridine and furopyridine kinase inhibitors
US20100069371A1 (en) * 2003-07-24 2010-03-18 Abbott Laboratories Thienopyridine and furopyridine kinase inhibitors

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8273736B2 (en) * 2003-07-24 2012-09-25 Abbott Laboratories Thienopyridine and furopyridine kinase inhibitors
US20100069371A1 (en) * 2003-07-24 2010-03-18 Abbott Laboratories Thienopyridine and furopyridine kinase inhibitors
US8557995B2 (en) * 2010-06-09 2013-10-15 Abbvie Inc. Solid dispersions containing kinase inhibitors
US8357804B2 (en) 2010-06-09 2013-01-22 Abbvie Inc. Crystal forms of kinase inhibitors
US20110306632A1 (en) * 2010-06-09 2011-12-15 Abbott Laboratories Solid Dispersions Containing Kinase Inhibitors
US8592589B2 (en) 2010-06-09 2013-11-26 Abbvie Inc. Methods of treating cancer with a thieno[3,2-C]pyridine
US8633317B2 (en) 2010-06-09 2014-01-21 Abbvie Inc. Crystalline salts of thieno[3,2-c]pyridine kinase inhibitors with improved cpy safety profile
US8633217B2 (en) 2010-06-09 2014-01-21 Abbvie Inc. Crystal forms of kinase inhibitors
US8911725B2 (en) 2010-06-23 2014-12-16 University Of Central Florida Research Foundation, Inc. Co-targeting of aurora A kinase and LIM kinase 1 for cancer therapy
US8436179B2 (en) 2011-07-20 2013-05-07 Abbvie Inc. Kinase inhibitor with improved solubility profile
WO2013090666A1 (en) 2011-12-14 2013-06-20 Abbvie Inc. Compositions containing kinase inhibitors
CN103987406A (en) * 2011-12-14 2014-08-13 艾伯维公司 Compositions containing kinase inhibitors
WO2015157360A1 (en) 2014-04-08 2015-10-15 Abbvie Inc. Processes to make protein kinase inhibitors
WO2015200635A1 (en) 2014-06-25 2015-12-30 Abbvie Inc. N-(4-{4-AMINO-7-[1-(2-HYDROXYETHYL)-1H-PYRAZOL-4-yl]THIENO[3,2-c]PYRIDIN-3-YL}PHENYL)-N'-(3-FLUOROPHENYL)UREA DOCUSATE
US20170197980A1 (en) * 2014-06-25 2017-07-13 Abbvie Inc. N-(4-phenyl)-n'-(3-fluorophenyl)urea docusate

Also Published As

Publication number Publication date
CN102239171A (en) 2011-11-09
NZ592714A (en) 2013-03-28
WO2010065825A3 (en) 2010-08-26
AU2009322270A1 (en) 2011-07-14
KR101639642B1 (en) 2016-07-14
PE20110830A1 (en) 2011-12-14
TW201026706A (en) 2010-07-16
JP5588458B2 (en) 2014-09-10
IL212716A0 (en) 2011-07-31
WO2010065825A2 (en) 2010-06-10
ES2524966T3 (en) 2014-12-16
IL212716A (en) 2013-10-31
TWI441827B (en) 2014-06-21
UA103351C2 (en) 2013-10-10
HK1159628A1 (en) 2012-08-03
EP2373662B1 (en) 2014-09-24
EP2373662A2 (en) 2011-10-12
DOP2011000153A (en) 2011-06-30
UY32291A (en) 2010-07-30
US8722890B2 (en) 2014-05-13
RU2480472C2 (en) 2013-04-27
ECSP11011172A (en) 2011-08-31
CO6382131A2 (en) 2012-02-15
JP2012511016A (en) 2012-05-17
CA2743592A1 (en) 2010-06-10
US20120309783A1 (en) 2012-12-06
CL2011001312A1 (en) 2011-10-07
TW201431862A (en) 2014-08-16
KR20110091577A (en) 2011-08-11
TWI500621B (en) 2015-09-21
RU2011127451A (en) 2013-01-10
MX2011005943A (en) 2011-06-27
RU2012156958A (en) 2014-06-27
MY179042A (en) 2020-10-26
AU2009322270B2 (en) 2014-12-18
PA8852401A1 (en) 2010-07-27
BRPI0921392A2 (en) 2016-04-26
AR074481A1 (en) 2011-01-19
CN102239171B (en) 2014-06-11

Similar Documents

Publication Publication Date Title
US8722890B2 (en) Thieno[3,2-C]pyridine kinase inhibitors with improved CYP safety profile
US8293738B2 (en) Indazole inhibitors of kinase
US8232273B2 (en) Heterocyclic compounds and methods of use
US8110572B2 (en) Inhibitors of protein kinases
US8486933B2 (en) Pyrimidine inhibitors of kinase activity
EP2435435B1 (en) Pyrimidine inhibitors of kinase activity
US8354422B2 (en) Pyrimidine inhibitors of kinase activity
US9051315B2 (en) Imidazopyridines as a novel scaffold for multi-targeted kinase inhibition
US8436179B2 (en) Kinase inhibitor with improved solubility profile

Legal Events

Date Code Title Description
AS Assignment

Owner name: ABBOTT LABORATORIES,ILLINOIS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:MICHAELIDES, MICHAEL R.;REEL/FRAME:023885/0937

Effective date: 20100202

AS Assignment

Owner name: ABBOTT LABORATORIES, ILLINOIS

Free format text: CORRECTIVE ASSIGNMENT TO CORRECT THE ASSIGNOR PREVIOUSLY RECORDED ON REEL 023885 FRAME 0937. ASSIGNOR(S) HEREBY CONFIRMS THE MICHAEL MICHAELIDES TO MICHAEL MICHAELIDES AND ROBIN FREY AND MICHAEL CURTIN ;ASSIGNORS:MICHAELIDES, MICHAEL R.;FREY, ROBIN;CURTIN, MICHAEL;REEL/FRAME:024753/0850

Effective date: 20100727

STCB Information on status: application discontinuation

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

AS Assignment

Owner name: ABBVIE INC., ILLINOIS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:ABBOTT LABORATORIES;REEL/FRAME:031941/0663

Effective date: 20120801