WO2010051781A1 - Kinase inhibitors and their use as pharmaceutical agents - Google Patents

Kinase inhibitors and their use as pharmaceutical agents Download PDF

Info

Publication number
WO2010051781A1
WO2010051781A1 PCT/CN2009/075214 CN2009075214W WO2010051781A1 WO 2010051781 A1 WO2010051781 A1 WO 2010051781A1 CN 2009075214 W CN2009075214 W CN 2009075214W WO 2010051781 A1 WO2010051781 A1 WO 2010051781A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
pyrimidin
pyrrolo
pyridin
ylamino
Prior art date
Application number
PCT/CN2009/075214
Other languages
French (fr)
Inventor
Ying Luo
Fang SHU
Original Assignee
Shanghai Genomics, Inc.
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 Shanghai Genomics, Inc. filed Critical Shanghai Genomics, Inc.
Publication of WO2010051781A1 publication Critical patent/WO2010051781A1/en
Priority to US13/095,526 priority Critical patent/US8309550B2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems

Definitions

  • Described herein are compounds, methods of making such compounds, pharmaceutical compositions and medicaments comprising such compounds, and methods of using such compounds to treat, prevent or diagnose diseases, disorders or conditions associated with one or more protein kinases.
  • Many diseases are associated with abnormal cellular responses triggered by protein kinase-mediated events. These diseases include, but are not limited to allergies, asthma, Alzheimer's disease, autoimmune diseases, bone diseases, cancer, cardiovascular diseases, inflammatory diseases, hormone-related diseases, metabolic diseases, neurological and neurodegenerative diseases. Described herein are protein kinase inhibitors that are effective as therapeutic agents.
  • compositions, medicaments, and methods for (a) diagnosing preventing, or treating conditions, disorders, or disorders associated with one or more protein kinases, (b) mitigating adverse signs and symptoms that are associated with one or more protein kinases, and/or (c) controlling conditions, disorders, or disorders associated with one or more protein kinases.
  • These conditions, disorders, or disorders may arise from one or more of a genetic, iatrogenic, immunological, infectious, metabolic, oncological, toxic, surgical, and/or traumatic etiology.
  • the methods, compounds, pharmaceutical compositions, and medicaments described herein comprise inhibitors of one or more protein kinases.
  • R 3 is H or alkyl;
  • R 1 , R 2 , R 3 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 and R 11 are each independently selected from the group consisting of H, halogen, - NH 2 , -NO 2 , -CN, -OH, -CO 2 H, -CONH 2 , -SO 3 H, -SO 2 NH 2 , -SO 2 CH 3 , -OCH 3 , -CF 3, -CH 3 , and -L A -L B -R 32 ;
  • L A is a covalent bond or an alkyl group;
  • Z is -NR 3 -.
  • R 3 is H.
  • R 5 and R 6 are each H.
  • R 2 is H.
  • each R 3 is independently selected from the group consisting of H, halogen, -NH 2 , -NO 2 ,
  • R 7 , R 8 , R 9 , R 10 and R 11 are each independently selected from the group consisting of H, halogen, -NH 2 , -NO 2 , -CN, -OH, -CO 2 H, -CONH 2 , -SO 3 H, -SO 2 NH 2 , -SO 2 CH 3 , -OCH 3 , -CF 3, -CH 3 , and -L A -L B -R 32 ;
  • L A is a covalent bond or an alkyl group;
  • R 32 is H, substituted or unsubstituted alkyl, haloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl; wherein any R 32 group, when substituted, is substituted with one or more groups selected from halogen, -
  • X is -C(R 3 )- or -N-;
  • R 1 is unsubstituted Ci-C 6 alkyl, unsubstituted or substituted aryl, or unsubstituted or substituted heteroaryl;
  • R 2 , R 3 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 and R 11 are each independently selected from the group consisting of H, halogen, -
  • L A is a covalent bond or an alkyl group
  • R 3 is H or alkyl.
  • X is N. In another aspect, X is -C(R 3 )-. [0012] In some embodiments, Z is -NR 3 -.
  • R 1 is unsubstituted Ci-C 6 alkyl or unsubstituted or substituted phenyl. In some other embodiments, R 1 is unsubstituted Ci-C 6 alkyl.
  • each R 3 is independently selected from the group consisting of H, halogen, -NH 2 , -NO 2 , -CN, -OH, -CO 2 H, -CONH 2 , -SO 3 H, -SO 2 NH 2 , -SO 2 CH 3 , -OCH 3 , -CF 3, and -CH 3 .
  • R 2 , R 5 and R 6 are each H.
  • R 7 , R 8 , R 9 , R 10 and R 11 are each independently selected from the group consisting of H, halogen, -NH 2 , -NO 2 , -CN, -OH, -CO 2 H, -CONH 2 , -SO 3 H, -SO 2 NH 2 , -SO 2 CH 3 , -OCH 3 , -CF 3, -CH 3, and -L A -L B -R 32 ;
  • L A is a covalent bond or an alkyl group;
  • R 32 is H, substituted or unsubstituted alkyl, haloalkyl, substituted or unsubstituted ary
  • R 1 , R 2 , R 3 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 and R 11 are each independently selected from the group consisting of H, halogen,
  • L A is a covalent bond or an alkyl group
  • R 3 is H or alkyl.
  • Z is -NR 3 -.
  • R 1 is unsubstituted Ci-C 6 alkyl.
  • each R 3 is independently selected from the group consisting of H, halogen, -NH 2 , -NO 2 , -CN, -OH, -CO 2 H, -CONH 2 , -SO 3 H, -SO 2 NH 2 , -SO 2 CH 3 , -OCH 3 , -CF 3, and -CH 3 .
  • R 2 , R 5 and R 6 are each H.
  • R 1 , R 2 , R 3 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 and R 11 are each independently selected from the group consisting of H, halogen, -NH 2 , -NO 2 , -CN, -OH, -CO 2 H, -CONH 2 , -SO 3 H, -SO 2 NH 2 , -SO 2 CH 3 , -OCH 3 , -CF 3, -CH 3 , and -L A -L B -R 32 ;
  • L A is a covalent bond or an alkyl group
  • R 32 is H, substituted or unsubstituted alkyl, haloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted non-aromatic heterocycle, or substituted or unsubstituted cycloalkyl; wherein any R 32 group, when substituted, is substituted with one or more groups selected from halogen, -NH 2 , -NO 2 , -CN, -OH, -CO 2 H, -CONH 2 , -SO 3 H, -SO 2 NH 2 , -SO 2 CH 3 , -OCH 3 , and -CF 3 ;
  • R 3 is H or alkyl.
  • Z is -NR 3 -.
  • R 1 is unsubstituted Ci-C 6 alkyl.
  • each R 3 is independently selected from the group consisting of H, halogen, -NH 2 , -NO 2 , -CN, -OH, -CO 2 H, -CONH 2 , -SO 3 H, -SO 2 NH 2 , -SO 2 CH 3 , -OCH 3 , -CF 3, and -CH 3 .
  • R 2 , R 5 and R 6 are each H.
  • R 32 is H, substituted or unsubstituted alkyl, or haloalkyl; wherein any R 32 group, when substituted, is substituted with one or more groups selected from halogen, -NH 2 , -NO 2 , -CN, -OH, -CO 2 H, -CONH 2 , - SO 3 H, -SO 2 NH 2 , -SO 2 CH 3 , -OCH 3 , and -CF 3 .
  • R 32 is H, substituted or unsubstituted alkyl, or haloalkyl.
  • R 1 , R 3 , R 7 , R 8 , R 9 , R 10 , R 11 are as defined in Table 1, Table 2, Table 3 and Table 4.
  • R 2 , R 5 , and R 6 are each H and R 1 , R 3 , R 7 , R 8 , R 9 , R 10 , R 11 are as defined in Table 1, Table 2, Table 3 and Table 4.
  • the compound of Formula (I) inhibits at least one kinase selected from among a Cyclin- dependent kinase (CDK), a Glycogen synthase kinase (GSK), an aurora kinase, a polo-like kinase (PLK), BCR-ABL,
  • CDK Cyclin- dependent kinase
  • GSK Glycogen synthase kinase
  • aurora kinase an aurora kinase
  • PLK polo-like kinase
  • BCR-ABL BCR-ABL
  • IKK IkappaB kinase complex
  • FLT3 Fms-like tyrosine kinase 3
  • JAK Janus kinase
  • LCK lymphocyte-specific protein tyrosine kinase
  • PDGFR Platelet-derived growth factor receptor kinase
  • Src Src kinase
  • VEGF vascular endothelial growth factor
  • a pharmaceutical composition comprising a compound of Formula (I) and a pharmaceutically excipient.
  • the pharmaceutical composition is in the form of an aqueous dispersion, liquid, gel, syrup, elixir, slurry, suspension, aerosol, controlled release formulation, fast melt formulation, effervescent formulation, lyophilized formulation, tablet, powder, pill, dragee, capsule, delayed release formulation, extended release formulation, pulsatile release formulation, multiparticulate formulation, or immediate release formulation.
  • a method for treating or preventing a kinase mediated or kinase dependent disease, disorder or condition in a patient comprising administrating to the patient an effective amount of a compound of
  • the kinase is selected from among a Cyclin-dependent kinase (CDK), a Glycogen synthase kinase (GSK), an aurora kinase, a polo-like kinase (PLK), BCR-ABL, IkappaB kinase complex (IKK), Fms-like tyrosine kinase 3 (FLT3), Janus kinase (JAK), lymphocyte-specific protein tyrosine kinase (LCK), Platelet-derived growth factor receptor kinase (PDGFR), a Src kinase, and vascular endothelial growth factor (VEGF) receptor.
  • CDK Cyclin-dependent kinase
  • GSK Glycogen synthase kinase
  • aurora kinase a polo-like kinase
  • PLK PCR-ABL
  • IKK IkappaB
  • the kinase is a Cyclin-dependent kinase (CDK).
  • CDK Cyclin-dependent kinase
  • the kinase mediated or kinase dependent disease, disorder or condition is a proliferative disorder.
  • the proliferative disorder is cancer or leukemia.
  • the proliferative disorder is cancer or leukemia selected from cervical cancer, osteosarcoma, esophageal cancer, liver cancer, myelogenous leukaemia, melanoma, acute lymphoblastic leukemia, lung cancer, breast cancer, or renal cancer.
  • a pharmaceutical composition comprising an amount of a compound of Formula (I) effective for inhibiting cyclin-dependent kinase 2 activity in a cell, and a pharmaceutically acceptable excipient.
  • a method for inhibiting undesired cyclin-dependent kinase 2 activity in a cell comprising contacting the cell with a compound of Formula (I).
  • a method for treating cancer in a patient in which the cancer is characterized by undesired cyclin-dependent kinase 2 activity, comprising administering to the patient an amount of a compound of
  • Formula (I) effective for inhibiting the undesired cyclin-dependent kinase 2 activity.
  • the cancer is selected from breast cancer and cervical cancer.
  • a pharmaceutical composition comprising a pharmaceutically acceptable diluent, excipient or binder, and a compound of Formula (I), or pharmaceutically acceptable salt, pharmaceutically acceptable prodrug, or pharmaceutically acceptable solvate thereof.
  • compositions which includes an effective amount of a compound provided herein, and a pharmaceutically acceptable excipient.
  • compositions further including a second pharmaceutically active ingredient in addition to the compound of Formula (I).
  • a pharmaceutical composition comprising: i) a physiologically acceptable carrier, diluent, and/or excipient; and ii) one or more compounds described herein.
  • any of the aforementioned aspects are further embodiments that include single administrations of the effective amount of the compound of Formula (I), including further embodiments in which: (i) the compound of
  • Formula (I) is administered once; (ii) the compound of Formula (I) is administered to the mammal multiple times over the span of one day; (iii) the compound of Formula (I) is administered continually; or (iv) the compound of Formula (I) is administered continuously.
  • any of the aforementioned aspects are further embodiments that include multiple administrations of the effective amount of the compound of Formula (I), including further embodiments in which (i) the compound of Formula (I) is administered in a single dose; (ii) the time between multiple administrations is every 6 hours; (iii) the compound of Formula (I) is administered to the mammal every 8 hours.
  • the method comprises a drug holiday, wherein the administration of the compound of Formula (I) is temporarily suspended or the dose of the compound of Formula (I) being administered is temporarily reduced; at the end of the drug holiday, dosing of the compound of Formula (I) is resumed.
  • the length of the drug holiday can vary from 2 days to 1 year.
  • compounds of Formula (I) described herein are administered to a human. In some embodiments, compounds of Formula (I) described herein are orally administered.
  • a further aspect relates to the use of a compound of Formula (I) in the manufacture of a medicament for treating a condition mediated by an enzyme selected from one or more CDK, aurora kinase, GSK, PLK, BCR-ABL, FLT, IKK, JAK, PDGF or VEGF and Src family enzymes.
  • an enzyme selected from one or more CDK, aurora kinase, GSK, PLK, BCR-ABL, FLT, IKK, JAK, PDGF or VEGF and Src family enzymes.
  • a compound of Formula (I) in the manufacture of a medicament for treating a condition mediated by at least one enzyme selected from CDKl, CDK2, CDK4/6, CDK5, CDK7, CDK8, CDK9, CDKl 1, GSK-3, aurora kinase, PLK and a tyrosine kinase.
  • one or more enzymes selected from CDK, aurora kinase, GSK, PLK, BCR-ABL, FLT, IKK, JAK, PDGF or VEGF and Src family enzymes, particularly from one or more CDKl, CDK2, CDK4/6, CDK5, CDK7, CDK8, CDK9, CDKl
  • a compound of Formula (I) in a method for treating a condition mediated by an enzyme selected from one or more CDK, aurora kinase, GSK, PLK, BCR-ABL, FLT, IKK, JAK, PDGF or VEGF and Src family enzymes, particularly from one or more CDKl, CDK2, CDK4/6, CDK5, CDK7, CDK8, CDK9, CDKl 1, GSK-3, aurora kinase, PLK or tyrosine kinase.
  • an enzyme selected from one or more CDK, aurora kinase, GSK, PLK, BCR-ABL, FLT, IKK, JAK, PDGF or VEGF and Src family enzymes, particularly from one or more CDKl, CDK2, CDK4/6, CDK5, CDK7, CDK8, CDK9, CDKl 1, GSK-3, aurora kinase, PLK or tyrosine kinase.
  • a further aspect relates to the use of a compound of Formula (I) or a pharmaceutically acceptable salt or solvate or physiologically hydrolysable, solubilising or immobilising derivative thereof, in an assay for identifying candidate compounds capable of treating a condition mediated by an enzyme selected from one or more CDK, aurora kinase, GSK, PLK, BCR-ABL, FLT, IKK, JAK, PDGF or VEGF and Src family enzymes, particularly from one or more CDKl, CDK2, CDK4/6, CDK5, CDK7, CDK8, CDK9, CDKIl, GSK-3, aurora kinase, PLK or tyrosine kinase.
  • an enzyme selected from one or more CDK, aurora kinase, GSK, PLK, BCR-ABL, FLT, IKK, JAK, PDGF or VEGF and Src family enzymes, particularly from one or more CDKl, CDK2, CDK4/6, CD
  • the protein kinase family is one of the largest in the human genome, comprising 500 genes.
  • the majority of kinases contain a 250-300 amino acid residue catalytic domain with a conserved core structure. This domain comprises a binding pocket for ATP, whose terminal phosphate group transfers covalently to its macromolecular substrates.
  • the protein kinases may be categorized by the substrates they phosphorylate, e.g. protein-serine/threonine, protein-tyrosine.
  • Protein kinases mediate intracellular signalling by affecting a phosphoryl transfer from a nucleoside triphosphate to a protein acceptor that is involved in a signalling pathway.
  • phosphorylation events are triggered in response to a variety of extracellular and other stimuli and act as molecular on/off switches that can modulate or regulate the target protein biological function.
  • An extracellular stimulus may affect one or more cellular responses related to cell growth, migration, differentiation, secretion of hormones, activation of transcription factors, muscle contraction, glucose metabolism, control of protein synthesis, and regulation of the cell cycle.
  • These diseases include, but are not limited to allergies, asthma, Alzheimer's disease, autoimmune diseases, bone diseases, cancer, cardiovascular diseases, inflammatory diseases, hormone-related diseases, metabolic diseases, neurological and neurodegenerative diseases.
  • protein kinase inhibitors that are effective as therapeutic agents.
  • CDKs Cyclin-dependent kinases
  • CDK 1-9 and 11 are involved in a variety of important regulatory pathways in eukaryotic cells, including cell-cycle control, apoptosis, neuronal physiology, differentiation and transcription.
  • CDKs are classified into two major groups, reflecting their functions.
  • CDKs composed primarily of CDKl, CDK2, CDK3, CDK4 and CDK6 function with their cyclin partners including cyclin A, B, Dl, D2, D3, E, and F to regulate promotion of the cell cycle.
  • the transcription regulator CDKs which include CDK7, CDK8, CDK9 and CDKl 1 work together with cyclin C, H, K, Ll, L2, Tl and T2, tend to play roles in transcriptional regulation.
  • CDKs are implicated in cell proliferation disorders, particularly in cancer.
  • Cell proliferation is a result of the direct or indirect deregulation of the cell division cycle and the CDKs play a critical role in the regulation of the various phases of this cycle. Therefore, inhibitors of CDKs and their associated cyclins are useful targets for cancer therapy.
  • CDKs also play a role in apoptosis and T-cell development, which is predominantly due to the CDK functions in regulation of transcription.
  • CDK inhibitors have been reported to restrain viral replication including human immunodeficiency virus, human cytomegalovirus, herpes virus, and varicella-zoster virus.
  • Inhibition of CDK is also useful for the treatment of neurodegenerative disorders such as Alzheimer's disease.
  • Paired Helical Filaments associated with Alzheimer's disease, is caused by the hyperphosphorylation of Tau protein by CDK5/p25.
  • Inhibition of one or more other serine/threonine kinases including the Aurora kinases, Glycogen synthesis kinases (GSKs), polo-like kinases (PLKs) and tyrosine kinases including Ableson tyrosine kinase (BCR-ABL), FMS- related tyrosine kinases (FLT), IkB kinases (IKK), Janus kinases (JAK), platelet-derived growth factor (PDGF) receptor tyrosine kinases, vascular endothelial growth factor (VEGF) receptor tyrosine kinases, and Src family are also useful for the treatment of numerous diseases, conditions or disorders mediated by these kinases.
  • GSKs Glycogen synthesis kinases
  • PLKs polo-like kinases
  • tyrosine kinases including Ableson tyrosine kinase (B
  • GSK3 is known to phosphorylate many substrates and is thus involved in the regulation of multiple biochemical pathways. For example, GSK3 is over-expressed in muscle cells of type II diabetics and that an inverse correlation exists between skeletal muscle GSK3 activity and insulin action. GSK3 inhibition is therefore of therapeutic significance in the treatment of diabetes, particularly type II, and diabetic neuropathy.
  • GSK is highly expressed in the central and peripheral nervous systems. GSK3 inhibition is therefore of therapeutic significance in the treatment of CNS disorders such as Parkinsons and Alzheimers diseases.
  • Aurora kinases and PLK are also important therapeutic targets for treatment of proliferative disorders. Based on their known functions inhibition of Aurora kinases and PLKs activity should disrupt mitosis leading to cell cycle arrest and therefore slowing tumor growth and induce apoptosis.
  • Described herein are compounds that inhibit one or more protein kinases.
  • the compounds described herein are heteroaryl-pyrimidine derivatives. More specifically, described herein are substituted 4-(indol-3- yl)-7V-phenyrpyrimidin-2 -amine compounds, substituted 4-(pyrrolo[2,3-i]pyridin-3-yl)-7V-phenylpyrimidin-2 -amine compounds, 4-(pyrrolo[2,3-c]pyridin-3-yl)-N-phenylpyrimidin-2-amine compounds, substituted 4-(pyrrolo[3,2-
  • the compounds described herein have broad therapeutic use as protein kinase inhibitors.
  • Het-Ar is an unsubstituted or substituted indolyl ring, or an unsubstituted or substituted azaindolyl ring;
  • R 3 is H or alkyl
  • R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , and R 11 are each independently H, alkyl, alkyl-R 19 , aryl, aryl-R 19 , aralkyl, aralkyl-R 19 , halogen,
  • alkyl (aryl), -NH-R 19 , -N-(R 19 )(R 20 ), -N-(alkyl)(R 19 ), -N-CaTyI)(R 19 ), -COOH, -CONH 2 , -CONH-alkyl, -CONH-aryl, -
  • alkyl, aryl, aralkyl groups may be further substituted with one or more groups selected from halogen, -
  • R 19 , R 20 , and R 21 are physiologically hydrolysable group, solubilising group or immobilisable group.
  • R 19 and R 20 are each independently solubilising groups selected from:
  • a mono-, di- or polyhydroxylated alicyclic group a di- or polyhydroxylated aliphatic or aromatic group; a carbohydrate derivative; an O- and/or S-containing heterocyclic group optionally substituted by one or more hydroxyl groups; an aliphatic or aromatic group containing a carboxamide, sulfoxide, sulfone, or sulfonamide function; or a halogenated alkylcarbonyl group;
  • COOH, SO 3 H, OSO 3 H, PO 3 H 2 , or OPO 3 H 2 a mono-, di- or polyhydroxylated alicyclic group; a di- or polyhydroxylated aliphatic or aromatic group; a carbohydrate derivative; an O- and/or S-containing heterocyclic group optionally substituted by one or more hydroxyl groups; an aliphatic or aromatic group containing a carboxamide, sulfoxide, sulfone, or sulfonamide function; or a
  • each R 21 is a solubilising group as defined for R 19 and R 20 in (i) or (iv) above, or is selected from
  • R 14 and R 15 are each independently H or alkyl, and n and n' are each independently O, 1, 2, or 3,
  • R 22 and R 23 are each independently H, alkyl or aryl, with the proviso that at least one of R 22 and R 23 is other than H, or R 22 and R 23 are linked to form a cyclic group optionally containing one or more heteroatoms selected from N, O and S, and wherein said alkyl, aryl or cyclic group is optionally substituted by one or more substituents selected from halogen, NO 2 , OH, alkoxy, NH 2 , COOH, CONH 2 and CF 3
  • At least one of R 7 , R 8 , R 9 , R 10 , and R 11 is selected from (CH 2 ) n NR 17 COR 18 ,
  • Het-Ar is a substituted or unsubstituted group selected from indol-3-yl, pyrrolo[2,3-£]pyridin-3- yl, pyrrolo[2,3-c]pyndin-3-yl, pyrrolo[3,2-£]pyridin-3-yl, and pyrrolo[3,2-c]pyridin-3-yl
  • Het-Ar is a substituted or unsubstituted group selected from l-(alkyl)-indol-3-yl, l-(alkyl)- pyrrolo[2,3-£]pyridin-3-yl, l-(alkyl)-pyrrolo[2,3-c]pyndin-3-yl, l-(alkyl)-pyrrolo[3,2-£]pyridin-3-yl, and l-(alkyl)- pyrrolo[3 ,2-c]pyndin-3 -yl
  • the compound of Formula (I) has the structure
  • alkyl, aryl, aralkyl groups may be further substituted with one or more groups selected from halogen, -NO 2 , -OH, -0-methyl, -NH 2 , -COOH, -CONH 2 and -CF 3 ; where R 19 , R 20 , and R 21 are physiologically hydrolysable group, solubilising group or immobilisable group.
  • R 1 , R 2 , R 3 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , and R 11 are each independently H, alkyl, aryl, aralkyl, halogen, -NO 2 , -CN, -OH, -O-alkyl, -O-aryl, -NH 2 , -NH-alkyl, -NH-aryl, -N-(alkyl) 2 , -N-(aryl) 2 , -N-(alkyl)(aryl), -
  • alkyl, aryl, aralkyl groups may be further substituted with one or more groups selected from halogen, -NO 2 , -
  • R 5 and R 6 are each H.
  • R 2 is H or alkyl. In another aspect, R 2 is H.
  • At least one R 3 is H. In another aspect, at least two R 3 are H.
  • At least one of R 7 , R 8 , R 9 , R 10 , and R 11 is H. In another aspect, at least two of R 7 , R 8 , R 9 , R 10 , and R 11 are H.
  • the compound of Formula (I) has the structure:
  • R 3 is H or alkyl; and wherein R 1 , R 2 , R 3 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , and R 11 , are as described herein.
  • the compound of Formula (I) has the structure of Formula (Ia):
  • R 3 is H or alkyl; and wherein R 1 , R 2 , R 3 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , and R 11 , are as described herein.
  • the compound of Formula (I) has the structure of Formula (Ib): Formula (Ib) wherein
  • R 3 is H or alkyl; and wherein R 1 , R 2 , R 3 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , and R 11 , are as described herein.
  • the compound of Formula (I) has the structure of Formula (Ic)
  • R 3 is H or alkyl; and wherein R 1 , R 2 , R 3 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , and R 11 , are as described herein.
  • the compound of Formula (I) has the structure of Formula (Id)
  • R 3 is H or alkyl; and wherein R 1 , R 2 , R 3 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , and R 11 , are as described herein.
  • R 3 is H or alkyl.
  • R 3 is H.
  • Z is -NR 3 -. In one aspect, Z is -NH-.
  • R 19 and R 20 are each independently solubilising groups selected from a mono-, di- or polyhydroxylated alicyclic group; a di- or polyhydroxylated aliphatic or aromatic group; a carbohydrate derivative; an O- and/or S-containing heterocyclic group optionally substituted by one or more hydroxyl groups, an aliphatic or aromatic group containing a carboxamide, sulfoxide, sulfone, or sulfonamide function, or a halogenated alkylcarbonyl group, COOH, SO 3 H, OSO 3 H, PO 3 H 2 , or OPO 3 H 2
  • At least one of R 19 and R 20 is a natural or unnatural amino acid residues and peptides, or their derivatives
  • R 19 or R 20 is selected from OSO 3 H, PO 3 H 2 , or OPO 3 H 2 ,
  • each R 21 is a solubilising group as defined for R 19 and R 20 hereinbefore defined, or is selected from OSO 3 H, PO 3 H 2 , or OPO 3 H 2 , Y as defined above, but excluding guanidine and quarternary amine salts, NHCO(CH 2 ) m [NHCO(CH 2 ) m ] p [NHCO(CH 2 ) m ] q Y or NHCO(CH 2 ) t NH(CH 2 ) t Y where p and q are each O or 1, and m, m',m", t and t' are each independently an integer from 1 to 10, and (CHz) n NR 14 COR 12 , (CH 2 ) n NR 15 SO 2 R 13 , or SO 2 R 16 , where R 12 , R 13 and R 16 are each alkyl groups optionally comprising one or more heteroatoms, and which are optionally substituted by one or more substituents selected
  • R 7 , R 8 , R 9 , R 10 and R 11 are each independently H, alkyl, alkyl-R 19 , aryl, aryl-R 19 , aralkyl, aralkyl-R 19 , halogen, -NO 2 , -CN, -OH, -O-alkyl, -COR 19 , -COOR 19 , -O-aryl, -0-R 19 , -NH 2 , NH-alkyl, NH-aryl, N- (alkyl) 2 , N-(aryl) 2 , -N-(alkyl)(aryl), NH-R 19 , N-(R 19 )(R 20 ), N-(alkyl)(R 19 ), -N-tarylXR 19 ), -COOH, -CONH 2 , -CONH- alkyl, -CONH-aryl, -CON-(alkyl) , halogen,
  • R 1 is selected from H, Ci-Cs-alkyl, aryl, heteroaryl, cycloalkyl, alkyl-aryl, alkyl-heteroaryl and alkyl-cycloalkyl
  • R 2 is selected from H, alkyl, C 1 -C 5 alkyl, CONH 2 , CONH-alkyl, CN, CF 3 , O-alkyl, halogen, NH 2 , NH-alkyl, NHCO-alkyl, alkyl-heteroaryl, and alkyl-cycloalkyl.
  • R 2 is selected from H, C 1 -C 5 alkyl, CONH 2 , CONH-alkyl, CN, CF 3 , O-alkyl, halogen, NH 2 , NH-alkyl, NHCO-alkyl, alkyl-heteroaryl, and alkyl-cycloalkyl.
  • each R 3 is independently selected from H, alkyl, CONH 2 , CONH-alkyl, CN, OH, CF 3 , O-alkyl, halogen, NH 2 , NH-alkyl, NHCO-alkyl, alkyl-heteroaryl, alkyl-cycloalkyl.
  • R 7 is H, O-alkyl, CF 3 , alkyl or halogen.
  • R 7 is selected from H, OCH 3 , CF 3 , alkyl or halogen.
  • R 8 and R 10 are independently selected from H, CONH 2 , CONH-alkyl, CN, OH, CF 3 , sulphonyl, carbonyl, amide or sulphonamide, or thioether link to an unsubstituted or substituted 6 membered cyclic or heterocyclic, or aromatic or heteroaromatic ring, wherein substituents are as hereinbefore defined.
  • a cycloheteroalkyl is a 7V-alkyl-morpholino, 7V-alkyl-piperazine or 7V-alkyl-piperadine.
  • R 9 is selected from H, alkyl, for example C 1-5 alkyl, CONH 2 , CONH-alkyl, CN, OH, CF 3 , O- alkyl, halogen, NH 2 , NH-alkyl, NHCO-alkyl, sulphonyl, carbonyl, amide or sulphonamide, or thioether link to an unsubstituted or substituted 6 membered cyclic or heterocyclic, or aromatic or heteroaromatic ring, wherein substituents are as hereinbefore defined.
  • R 11 is selected from H, halogen, O-alkyl. In one aspect, R 11 is selected from H, halogen and O- C 1 -C 5 alkyl.
  • R 1 , R 2 , R 3 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , and R 11 shall correspond to or contain one or more of the group R 19 or R 20 .
  • R 8 , R 9 or R 10 comprise or contain one or more of the group R 20 or R 21 .
  • Two or more groups R 19 and/or R 20 may be the same or different.
  • R 1 , R 2 , R 3 , R 8 or R 9 comprises or contains a solubilising moiety R 19 or R 20 .
  • R 1 , R 2 , R 3 , R 8 or R 9 comprises or contains a solubilising moiety R 19 or R 20 .
  • R 1 , R 2 , R 3 , R 8 or R 9 comprises or contains a solubilising moiety R 19 or R 20 .
  • R 1 X is CR 3 or N
  • Z is NH
  • R 1 and R 2 are selected from H, alkyl, such as Q.s-alkyl or aryl, such as C 6 aryl; halogen, or heteroaryl
  • R 3 is one, two or three groups independently selected from H, amino, Ci -4 alkyl, CN, CF 3 , halogen, NO 2 O-alkyl, NH 2 , NH-alkyl, N(alkyl) 2, CO 2 alkyl, CO-alkyl, CONH 2 ,
  • R 1 and R 2 are selected from H, alkyl, such as Ci_ 5 -alkyl or aryl, such as C 6 aryl; halogen, or heteroaryl;
  • R 3 is one, two or three groups independently selected from H, amino, Ci -4 alkyl, CN, CF 3 , halogen, NO 2 O-alkyl, NH 2 ,
  • R 7 is H, OMe, alkyl, or halogen
  • each R 8 , R 9 or R 10 is independently alkyl, OH, CF 3 , O-alkyl, halogen, O-heteroaryl, alkyl-heteroaryl, alkyl-cycloalkyl which may be part unsaturated; NH 2 SO 2 -NH 2 , SO 2 -NR 20 , SO 2 -cycloheteroalkyl, SO-cycloheteroalkyl, SO 2 -heteroaryl,
  • Z is NH
  • R 1 and R 2 are selected from H, alkyl, such as Ci_ 5 -alkyl or aryl, such as C 6 aryl, halogen, or heteroaryl;
  • R 3 is one or more groups selected from H, amino, Ci _ 4 alkyl, CN, CF 3 , halogen, NO 2 O-alkyl, NH 2 , NH-alkyl, N(alkyl) 2
  • R 7 is H, OMe, alkyl, or halogen
  • each R 8 , R 9 or R 10 is independently alkyl, OH, CF 3 , O-alkyl, halogen, O-heteroaryl, alkyl-heteroaryl, alkyl-cycloalkyl which may be part unsaturated; NH 2 SO 2 -NH 2 , SO 2 -NR 20 , SO 2 -cycloheteroalkyl, SO-cycloheteroalkyl, SO 2 -heteroaryl,
  • Such devices may be chemical functions that can be used for covalent attachment to solid phases such as functionalized polymers (e.g. agarose, polyacrylamide, polystyrene etc.) as commonly found in matrices (microtitre plate wells, microbeads, membranes, etc.) used for biochemical assays and affinity chromatography.
  • the devices may be small molecules (e.g. biotin) or polypeptides (e.g. antigens), which can be used for non-covalent immobilization through binding to an immobilized receptor (e.g. avidin or streptavidin in the case of biotin, or a specific antibody in the case of antigens).
  • an immobilized receptor e.g. avidin or streptavidin in the case of biotin, or a specific antibody in the case of antigens.
  • R 1 , R 2 , R 3 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 and R 11 are each independently selected from the group consisting of H, halogen, -NH 2 , -NO 2 , -CN, -OH, -CO 2 H, -CONH 2 , -SO 3 H, -SO 2 NH 2 , -SO 2 CH 3 , -OCH 3 , -CF 3 -CH 3 , and -L A -L B -R 32 ;
  • L A is a covalent bond or an alkyl group;
  • R 32 is H, substituted or unsubstituted alkyl, haloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted non-aromatic heterocycle, or substituted or unsubstituted cycloalkyl; wherein any R 32 group, when substituted, is substituted with one or more groups selected from halogen, -
  • X is -C(R 3 )- or -N-;
  • R 1 is unsubstituted Ci-C 6 alkyl, unsubstituted or substituted aryl, or unsubstituted or substituted heteroaryl;
  • R 2 , R 3 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 and R 11 are each independently selected from the group consisting of H, halogen, - NH 2 , -NO 2 , -CN, -OH, -CO 2 H, -CONH 2 , -SO 3 H, -SO 2 NH 2 , -SO 2 CH 3 , -OCH 3 , -CF 3, -CH 3 , and -L A -L B -R 32 ;
  • L A is a covalent bond or an alkyl group
  • R 32 is H, substituted or unsubstituted alkyl, haloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted non-aromatic heterocycle, or substituted or unsubstituted cycloalkyl; wherein any R 32 group, when substituted, is substituted with one or more groups selected from halogen, -NH 2 , -NO 2 , -CN, -OH, -CO 2 H, -CONH 2 , -SO 3 H, -SO 2 NH 2 , -SO 2 CH 3 , -OCH 3 , and -CF 3 ;
  • R 3 is H or alkyl.
  • X is N. In another aspect, X is -C(R 3 )-. [00114] In some embodiments, Z is -NR 3 -.
  • R 1 is unsubstituted Ci-C 6 alkyl or unsubstituted or substituted phenyl.
  • each R 3 is independently selected from the group consisting of H, halogen, -NH 2 , -NO 2 , -CN, -OH, -CO 2 H, -CONH 2 , -SO 3 H, -SO 2 NH 2 , -SO 2 CH 3 , -OCH 3 , -CF 3, and -CH 3 .
  • R 2 , R 5 and R 6 are each H.
  • R 7 , R 8 , R 9 , R 10 and R 11 are each independently selected from the group consisting of H, halogen, -NH 2 , -NO 2 , -CN, -OH, -CO 2 H, -CONH 2 , -SO 3 H, -SO 2 NH 2 , -SO 2 CH 3 , -OCH 3 , -CF 3, -CH 3, and -L A -L B -R 32 ;
  • L A is a covalent bond or an alkyl group;
  • R 32 is H, substituted or unsubstituted alkyl, haloalkyl, substituted or unsubstituted aryl
  • R 1 , R 2 , R 3 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 and R 11 are each independently selected from the group consisting of H, halogen,
  • L A is a covalent bond or an alkyl group
  • R 3 is H or alkyl.
  • Z is -NR 3 -.
  • R 1 is unsubstituted Ci-C 6 alkyl.
  • each R 3 is independently selected from the group consisting of H, halogen, -NH 2 , -NO 2 , -CN, -OH, -CO 2 H, -CONH 2 , -SO 3 H, -SO 2 NH 2 , -SO 2 CH 3 , -OCH 3 , -CF 3, and -CH 3 .
  • R 2 , R 5 and R 6 are each H.
  • R 1 , R 2 , R 3 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 and R 11 are each independently selected from the group consisting of H, halogen, -NH 2 , -NO 2 , -CN, -OH, -CO 2 H, -CONH 2 , -SO 3 H, -SO 2 NH 2 , -SO 2 CH 3 , -OCH 3 , -CF 3, -CH 3 , and -L A -L B -R 32 ;
  • L A is a covalent bond or an alkyl group
  • R 32 is H, substituted or unsubstituted alkyl, haloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted non-aromatic heterocycle, or substituted or unsubstituted cycloalkyl; wherein any R 32 group, when substituted, is substituted with one or more groups selected from halogen, -NH 2 , -NO 2 , -CN, -OH, -CO 2 H, -CONH 2 , -SO 3 H, -SO 2 NH 2 , -SO 2 CH 3 , -OCH 3 , and -CF 3 ;
  • R 3 is H or alkyl.
  • Z is -NR 3 -.
  • R 1 is unsubstituted Ci-C 6 alkyl.
  • each R 3 is independently selected from the group consisting of H, halogen, -NH 2 , -NO 2 , -CN, -OH, -CO 2 H, -CONH 2 , -SO 3 H, -SO 2 NH 2 , -SO 2 CH 3 , -OCH 3 , -CF 3, and -CH 3 .
  • R 2 , R 5 and R 6 are each H.
  • R 32 is H, substituted or unsubstituted alkyl, or haloalkyl; wherein any R 32 group, when substituted, is substituted with one or more groups selected from halogen, -NH 2 , -NO 2 , -CN, -OH, -CO 2 H, -CONH 2 , - SO 3 H, -SO 2 NH 2 , -SO 2 CH 3 , -OCH 3 , and -CF 3 .
  • R 32 is H, substituted or unsubstituted alkyl, or haloalkyl.
  • R 1 , R 3 , R 7 , R 8 , R 9 , R 10 , R 11 are as defined in Table 1, Table 2, Table 3 and Table 4.
  • R 2 , R 5 , and R 6 are each H and R 1 , R 3 , R 7 , R 8 , R 9 , R 10 , R 11 are as defined in Table 1, Table 2, Table 3 and Table 4.
  • compounds of Formula (I) include, but are not limited to, those described in Table 1: Table 1.
  • compounds of Formula (I) include, but are not limited to, those described in Table 2:
  • compounds of Formula (I) include, but are not limited to, those described in Table 3:
  • the starting material used for the synthesis of the compounds of Formula (I) described in the prior section are either synthesized or obtained from commercial sources, such as, but not limited to, Aldrich Chemical Co. (Milwaukee,
  • acylheteroaryl compounds of structure l-II are converted to an acrylate compound of structure l-III.
  • Het-Ar is a bicylic heteroaryl that includes at least one nitrogen in the bicyclic ring.
  • Het-Ar is selected from indole, pyrrolo[2,3-£]pyridine, pyrrolo[2,3-c]pyridine, pyrrolo[3,2-£]pyridine, and pyrrolo[3,2- cjpyridine, where Het-Ar is substituted or unsubstituted.
  • Het-Ar is selected from indol-3-yl, pyrrolo[2,3- £]pyridine-2-yl, pyrrolo[2,3-c]pyridine-3-yl, pyrrolo[3,2-£]pyridine-3-yl and pyrrolo[3,2-c]pyridine-3-yl, where Het-Ar is substituted or unsusbtituted.
  • acrylate compounds of structure l-III are reacted with phenylguanidine compounds of structure
  • L 1 is any leaving group. In some embodiments, L 1 is
  • L 1 is -NMe 2 .
  • a compound of structure 1-VI where W is a halide, triflate or boronic acid, boronate ester, or other suitable group, is reacted with a pyrimidine compound of structure 1-V, where L 2 and L 3 are leaving groups, such as a halide.
  • Suitable reaction conditions include, but are not limited to, metal-mediated reactions such as Suzuki reactions, Stille cross couplings, Negishi couplings, Kumada couplings, Ullmann reactions, Hiyama Coupling, and variants thereof (Metal-Catalyzed Cross-Coupling Reactions, Armin de Meijere (Editor), Francois Diederich (Editor),
  • Z is NH.
  • Suitable reaction conditions include, but are not limited to, metal-catalyzed cross couplings and SN AR reaction conditions.
  • Het-Ar is a 1 -substituted indol-3-yl, 1-substituted pyrrolo[2,3-£]pyridine-3-yl, 1-substituted pyrrolo[2,3-c]pyridine-3-yl, or 1-substituted pyrrolo[3,2-£]pyridine-3-yl.
  • the 1-substitutuent of the 1- substituted indol-3-yl, 1-substituted pyrrolo[2,3-£]pyridine-3-yl, 1-substituted pyrrolo[2,3-c]pyridine-3-yl, or 1- substituted pyrrolo[3,2-£]pyridine-3-yl is selected from -Q-C ⁇ alkyl, and -Q-C ⁇ alkyltsubstituted or unsubstituted aryl).
  • the 1-substitutuent of the 1-substituted indol-3-yl, 1-substituted pyrrolo[2,3-£]pyridine-3-yl, 1- substituted pyrrolo[2,3-c]pyridine-3-yl, or 1-substituted pyrrolo[3,2-£]pyridine-3-yl is selected from -CH 3 , -CH 2 CH 3 , -
  • C-R 3 are reacted with vinylmagnesium bromide to provide compounds of structure 2-XI.
  • at least two X are C-R 3 .
  • at least three X are C-R 3 .
  • the process of reacting 2-IX with a Grignard agent is as described in Zhang, Z, et al. J. Org. Chem., 2002, 67, 2345.
  • N-Alkylation of compounds of structure 2-XI with compounds of 2-XII provides compounds of structure 2-XIII.
  • compounds of structure 2-XI are reacted with compounds of 2-XII in the presence of a base, such as, but not limited to NaH or KOH.
  • Compounds of structure 2-XIII are acylated with compounds of structure 2-XIV to provide compounds of structure 2-XV.
  • Suitable reaction conditions include, but are not limited to, Friedel-Crafts acylation conditions.
  • L 1 is any leaving group.
  • L 1 is -
  • phenylguanidines of structure 1-IV are obtained by reaction of cyanamide or cyanamide derivatives using the method of Katritzky, A. R.; et al. Synthetic Communications 1995, 25, 1173.
  • Groups such as trityl, dimethoxytrityl, acetal and t-butyldimethylsilyl are acid labile and are used in certain embodiments to protect carboxy and hydroxy reactive moieties in the presence of amino groups protected with Cbz groups, which are removable by hydrogenolysis, and/or Fmoc groups, which are base labile.
  • carboxylic acid and hydroxy reactive moieties are blocked with base labile groups such as, but not limited to, methyl, ethyl, and acetyl in the presence of amines blocked with acid labile groups such as t-butyl carbamate or with carbamates that are both acid and base stable but hydrolytically removable.
  • carboxylic acid and hydroxy reactive moieties are blocked with hydrolytically removable protective groups such as the benzyl group, while amine groups capable of hydrogen bonding with acids are blocked with base labile groups such as Fmoc.
  • carboxylic acid reactive moieties are protected by conversion to simple ester compounds as exemplified herein, or they are, in yet another embodiment, blocked with oxidatively-removable protective groups such as 2,4-dimethoxybenzyl, while co-existing amino groups are blocked with fluoride labile silyl carbamates.
  • Allyl blocking groups are useful in the presence of acid- and base- protecting groups since the former are stable and can be subsequently removed by metal or pi-acid catalysts.
  • an allyl-blocked carboxylic acid can be deprotected with a Pd(O)-catalyzed reaction in the presence of acid labile t-butyl carbamate or base-labile acetate amine protecting groups.
  • Yet another form of protecting group is a resin to which a compound or intermediate is attached. As long as the residue is attached to the resin, that functional group is blocked and cannot react. Once released from the resin, the functional group is available to react.
  • blocking/protecting groups are, by way of example only:
  • compounds of Formula (I) are prepared as a pharmaceutically acceptable acid addition salt (which is a type of a pharmaceutically acceptable salt) by reacting the free base form of the compound with a pharmaceutically acceptable inorganic or organic acid, including, but not limited to, inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid metaphosphoric acid, and the like; and organic acids such as acetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, p-toluenesulfonic acid, tartaric acid, trifluoroacetic acid, citric acid, benzoic acid, 3-(4-hydroxybenzoyl)benzoic acid, cinnamic acid, mandelic acid, arylsulfonic acid, methan
  • pharmaceutically acceptable refers a material, such as a carrier or diluent, which does not abrogate the biological activity or properties of the compound, and is relatively nontoxic, i.e., the material may be administered to an individual without causing undesirable biological effects or interacting in a deleterious manner with any of the components of the composition in which it is contained.
  • pharmaceutically acceptable salt refers to a formulation of a compound that does not cause significant irritation to an organism to which it is administered and does not abrogate the biological activity and properties of the compound.
  • pharmaceutically acceptable salts are obtained by reacting a compound of Formula (I) with acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesurfonic acid, salicylic acid and the like.
  • Pharmaceutically acceptable salts are also obtained by reacting a compound of Formula (I) with a base to form a salt such as an ammonium salt, an alkali metal salt, such as a sodium or a potassium salt, an alkaline earth metal salt, such as a calcium or a magnesium salt, a salt of organic bases such as dicyclohexylamine, TV-methyl-D-glucamine, tris(hydroxymethyl)methylamine, and salts with amino acids such as arginine, lysine, and the like.
  • a salt such as an ammonium salt, an alkali metal salt, such as a sodium or a potassium salt, an alkaline earth metal salt, such as a calcium or a magnesium salt, a salt of organic bases such as dicyclohexylamine, TV-methyl-D-glucamine, tris(hydroxymethyl)methylamine, and salts with amino acids such as arginine, lysine, and the like.
  • compounds of Formula (I) are prepared as a pharmaceutically acceptable salts by reacting the free acid form of the compound with a pharmaceutically acceptable inorganic or organic base, including, but not limited to organic bases such as ethanolamine, diethanolamine, triethanolamine, tromethamine, N- methylglucamine, and the like, or with an inorganic base such as aluminum hydroxide, calcium hydroxide, potassium hydroxide, sodium carbonate, sodium hydroxide, and the like.
  • organic bases such as ethanolamine, diethanolamine, triethanolamine, tromethamine, N- methylglucamine, and the like
  • an inorganic base such as aluminum hydroxide, calcium hydroxide, potassium hydroxide, sodium carbonate, sodium hydroxide, and the like.
  • a reference to a pharmaceutically acceptable salt includes the solvent addition forms or crystal forms thereof, particularly solvates or polymorphs.
  • Solvates contain either stoichiometric or non- stoichiometric amounts of a solvent, and are optionally formed during the process of crystallization with pharmaceutically acceptable solvents such as water, ethanol, and the like. Hydrates are formed when the solvent is water, or alcoholates are formed when the solvent is alcohol. Solvates of compounds of Formula (I) are conveniently prepared or formed during the processes described herein.
  • hydrates of compounds of Formula (I) are conveniently prepared by recrystallization from an aqueous/organic solvent mixture, using organic solvents including, but not limited to, dioxane, tetrahydrofuran, ethanol, or methanol.
  • organic solvents including, but not limited to, dioxane, tetrahydrofuran, ethanol, or methanol.
  • the compounds provided herein can exist in unsolvated as well as solvated forms. In general, the solvated forms are considered equivalent to the unsolvated forms for the purposes of the compounds and methods provided herein.
  • the compounds of Formula (I) are prepared in various forms, including but not limited to, amorphous forms, milled forms and nano-particulate forms.
  • compounds of Formula (I) include crystalline forms, also known as polymorphs.
  • Polymorphs include the different crystal packing arrangements of the same elemental composition of a compound. Polymorphs usually have different X-ray diffraction patterns, infrared spectra, melting points, density, hardness, crystal shape, optical and electrical properties, stability, and solubility. Various factors such as the recrystallization solvent, rate of crystallization, and storage temperature may cause a single crystal form to dominate.
  • compounds of Formula (I) are prepared as prodrugs.
  • a "prodrug” refers to an agent that is converted into the parent drug in vivo. Prodrugs are often useful because, in some situations, they may be easier to administer than the parent drug. They may, for instance, be bioavailable by oral administration whereas the parent is not. The prodrug may also have improved solubility in pharmaceutical compositions over the parent drug.
  • prodrug a compound of Formula (I) which is administered as an ester (the "prodrug") to facilitate transmittal across a cell membrane where water solubility is detrimental to mobility but which then is metabolically hydrolyzed to the carboxylic acid, the active entity, once inside the cell where water-solubility is beneficial.
  • prodrug a short peptide (polyaminoacid) bonded to an acid group where the peptide is metabolized to reveal the active moiety.
  • Prodrugs are generally drug precursors that, following administration to a subject and subsequent absorption, are converted to an active, or a more active species via some process, such as conversion by a metabolic pathway. Some prodrugs have a chemical group present on the prodrug that renders it less active and/or confers solubility or some other property to the drug. Once the chemical group has been cleaved and/or modified from the prodrug the active drug is generated. Prodrugs are often useful because, in some situations, they are easier to administer than the parent drug. In certain embodiments, the prodrug of a compound described herein is bioavailable by oral administration whereas the parent is not. Furthermore, in some embodiments, the prodrug of a compound described herein has improved solubility in pharmaceutical compositions over the parent drug.
  • prodrugs are designed as reversible drug derivatives, for use as modifiers to enhance drug transport to site-specific tissues.
  • the design of prodrugs to date is to increase the effective water solubility of the therapeutic compound for targeting to regions where water is the principal solvent.
  • the compounds described herein are labeled isotopically (e.g. with a radioisotope) or by another other means, including, but not limited to, the use of chromophores or fluorescent moieties, bioluminescent labels, or chemiluminescent labels.
  • the compounds of Formula (I) possess one or more stereocenters and each center exists independently in either the R or S configuration.
  • the compounds presented herein include all diastereomeric, enantiomeric, and epimeric forms as well as the appropriate mixtures thereof.
  • compounds of Formula (I) are prepared as their individual stereoisomers by reacting a racemic mixture of the compound with an optically active resolving agent to form a pair of diastereoisomeric compounds, separating the diastereomers and recovering the optically pure enantiomers.
  • resolution of enantiomers is carried out using covalent diastereomeric derivatives of the compounds described herein.
  • dissociable complexes are utilized (e.g., crystalline diastereomeric salts).
  • Diastereomers have distinct physical properties (e.g., melting points, boiling points, solubilities, reactivity, etc.) and are, in specific embodiments, separated by taking advantage of these dissimilarities.
  • the diastereomers are separated by chiral chromatography or by separation/resolution techniques based upon differences in solubility. The optically pure enantiomer is then recovered, along with the resolving agent, by any practical means that does not result in racemization. Jean Jacques, Andre Collet, Samuel H. Wilen, “Enantiomers, Racemates and Resolutions", John Wiley And Sons, Inc., 1981.
  • the compounds provided herein exist as geometric isomers.
  • the compounds and methods provided herein include all cis, trans, syn, anti,
  • E
  • Z
  • the compounds described herein exist as tautomers. All tautomers are intended to be within the scope of the molecular formulas described herein.
  • mixtures of enantiomers and/or diastereoisomers, resulting from a single preparative step, combination, or interconversion are envisioned.
  • Compound of Formula (I) refers to compound of Formula (I), compound of Formula (Ia), compound of
  • alkyl refers to an aliphatic hydrocarbon group.
  • the alkyl moiety may be a saturated alkyl group
  • the alkyl moiety may be an unsaturated alkyl group (which means that it contains at least one unit of unsaturation).
  • the alkyl moiety, whether saturated or unsaturated, may be branched, or straight chain.
  • the "alkyl” moiety may have 1 to 8 carbon atoms (whenever it appears herein, a numerical range such as “1 to 8" refers to each integer in the given range; e.g. , " 1 to 8 carbon atoms” means that the alkyl group may consist of 1 carbon atom, 2 carbon atoms, 3 carbon atoms, etc., up to and including 8 carbon atoms, although the present definition also covers the occurrence of the term "alkyl” where no numerical range is designated).
  • the alkyl group of the compounds described herein may be designated as "Ci-C 6 alkyl" or similar designations. By way of example only, "Ci-C 6 alkyl"
  • C 6 alkyl indicates that there are one, two , three, four, five, or six carbon atoms in the alkyl chain.
  • Typical alkyl groups include, but are in no way limited to, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tertiary butyl, pentyl, hexyl, and the like.
  • the term "lower alkyl” is similarly used for groups having from 1 to 4 carbon atoms.
  • aralkyl is refers to -alkyl-aryl, where alkyl and aryl are as defined herein.
  • alicyclic refers to a cyclic aliphatic group.
  • aliphatic takes its normal meaning in the art and includes non-aromatic groups such as alkanes, alkenes and alkynes and substituted derivatives thereof.
  • alkoxy refers to a (alkyl)O- group, where alkyl is as defined herein.
  • An amide may be an amino acid or a peptide molecule attached to a compound of Formula (I), thereby forming a prodrug. Any amine, or carboxyl side chain on the compounds described herein is optionally amidified, as desired. See, e.g., Greene and Wuts, Protective Groups in Organic Synthesis, 3 rd Ed., John Wiley & Sons,
  • aromatic refers to a planar ring having a delocalized ⁇ -electron system containing 4n+2 ⁇ electrons, where n is an integer. Aromatic rings can be formed from five, six, seven, eight, nine, ten, or more than ten atoms. Aromatics are optionally substituted.
  • aromatic includes both carbocyclic aryl ("aryl”, e.g., phenyl) and heterocyclic aryl (or “heteroaryl” or “heteroaromatic”) groups (e.g., pyridine).
  • aryl e.g., phenyl
  • heterocyclic aryl or “heteroaryl” or “heteroaromatic” groups (e.g., pyridine).
  • the term includes monocyclic or fused-ring polycyclic (i.e., rings which share adjacent pairs of carbon atoms) groups.
  • Carbocyclic refers to a ring or ring system where the atoms forming the backbone of the ring are all carbon atoms. The term thus distinguishes carbocyclic from heterocyclic rings in which the ring backbone contains at least one atom which is different from carbon.
  • aryl refers to an aromatic ring wherein each of the atoms forming the ring is a carbon atom.
  • Aryl rings are formed by five, six, seven, eight, nine, or more than nine carbon atoms.
  • Aryl groups are optionally substituted.
  • an aryl is a phenyl or a naphthalenyl.
  • an aryl group can be a monoradical or a diradical (i.e., an arylene group).
  • an aryl is a C 6 -Q o aryl.
  • cycloalkyl refers to a monocyclic or polycyclic aliphatic, non-aromatic radical, wherein each of the atoms forming the ring (i.e. skeletal atoms) is a carbon atom. Cycloalkyls may be saturated, or partially unsaturated.
  • Cycloalkyls may be fused with an aromatic ring, and the point of attachment is at a carbon that is not an aromatic ring carbon atom.
  • Cycloalkyl groups include groups having from 3 to 10 ring atoms. In some embodiments, cycloalkyl groups are selected from among cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl.
  • Cycloalkyl groups may be substituted or unsubstituted.
  • carbohydrate derivative refers to a compound of general formula C x (H 2 O) y or a derivative thereof.
  • the carbohydrate is a mono-, di- or tri-saccharide.
  • Monosaccharides can exist as either straight chain or ring-shaped molecules and are classified according to the number of carbon atoms they possess; tnoses have three carbons, tetroses four, pentoses five and hexoses six. Each of these subgroups may be further divided into aldoses and ketoses, depending on whether the molecule contains an aldehyde group (-CHO) or a ketone group
  • Typical examples of monosaccharides include glucose, fructose, and galactose.
  • Disaccharides consist of two linked monosaccharide molecules, and include for example, maltose and lactose.
  • Trisaccharides consist of three linked monosaccharide molecules.
  • esters refers to a chemical moiety with formula -COOR, where R is selected from the group consisting of alkyl, cycloalkyl, aryl, heteroaryl (bonded through a ring carbon) and heteroalicyclic (bonded through a ring carbon). Any hydroxy, or carboxyl side chain on the compounds described herein is esterified, if desired.
  • halo or, alternatively, "halogen” or “halide” means fluoro, chloro, bromo or iodo.
  • haloalkyl refers to an alkyl group in which one or more hydrogen atoms are replaced by one or more halide atoms.
  • a haloalkyl is a Ci-C 6 haloalkyl.
  • fluoroalkyl refers to a alkyl in which one or more hydrogen atoms are replaced by a fluorine atom.
  • a fluoralkyl is a Ci-C 6 fluoroalkyl.
  • heteroalkyl refers to an alkyl group in which one or more skeletal atoms of the alkyl are selected from an atom other than carbon, e.g. , oxygen, nitrogen, sulfur, phosphorus or combinations thereof.
  • a heteroalkyl is a Ci-C 6 heteroalkyl.
  • heterocycle refers to heteroaromatic rings (also known as heteroaryls) and heterocycloalkyl rings (also known as heteroalicyclic groups) containing one to four heteroatoms in the ring(s), where each heteroatom in the ring(s) is selected from O, S and N, wherein each heterocyclic group has from 4 to 10 atoms in its ring system, and with the proviso that the any ring does not contain two adjacent O or S atoms.
  • Non-aromatic heterocyclic groups also known as heterocycloalkyls
  • the heterocyclic groups include benzo- fused ring systems.
  • An example of a 3-membered heterocyclic group is aziridinyl.
  • An example of a 4-membered heterocyclic group is azetidinyl.
  • An example of a 5-membered heterocyclic group is thiazolyl.
  • An example of a 6- membered heterocyclic group is pyridyl, and an example of a 10-membered heterocyclic group is quinolinyl.
  • non-aromatic heterocyclic groups are pyrrolidinyl, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, oxazolidinonyl, tetrahydropyranyl, dihydropyranyl, tetrahydrothiopyranyl, piperidinyl, morpholinyl, thiomorpholinyl, thioxanyl, piperazinyl, aziridinyl, azetidinyl, oxetanyl, thietanyl, homopiperidinyl, oxepanyl, thiepanyl, oxazepinyl, diazepinyl, thiazepinyl, 1,2,3,6-tetrahydropyridinyl, pyrrolin-2-yl, pyrrolin-3-yl, indolinyl, 2H-pyranyl, 4H-pyranyl, dioxanyl,
  • aromatic heterocyclic groups are pyridinyl, imidazolyl, pyrimidinyl, pyrazolyl, triazolyl, pyrazinyl, tetrazolyl, furyl, thienyl, isoxazolyl, thiazolyl, oxazolyl, isothiazolyl, pyrrolyl, quinolinyl, isoquinolinyl, indolyl, benzimidazolyl, benzofuranyl, cinnolinyl, indazolyl, indolizinyl, phthalazinyl, pyridazinyl, triazinyl, isoindolyl, pteridinyl, purinyl, oxadiazolyl, thiadiazolyl, furazanyl, benzofurazanyl, benzothiophenyl, benzothiazolyl, benzoxazolyl, quinazolinyl, quinox
  • the foregoing groups may be C-attached or TV-attached where such is possible.
  • a group derived from pyrrole may be pyrrol- 1-yl (TV-attached) or pyrrol-3-yl (C-attached).
  • a group derived from imidazole may be imidazol-1-yl or imidazol-3-yl (both TV- attached) or imidazol-2-yl, imidazol-4-yl or imidazol-5-yl (all C-attached).
  • heteroaryl or, alternatively, “heteroaromatic” refers to an aryl group that includes one or more ring heteroatoms selected from nitrogen, oxygen and sulfur.
  • Preferred heteroaryl groups include indole, azaindole, pyrrole, pyrazole, pyrimidine, pyrazine, pyridine, quinoline, thiophene and furan.
  • a heteroaryl contains 0-3 N atoms.
  • a heteroaryl contains 0-3 Natoms, 0-1 O atoms, and 0-1 S atoms.
  • a heteroaryl is a monocyclic or bicyclic heteroaryl.
  • a “heterocycloalkyl”, “heteroalicyclic” or “non-aromatic heterocycle” refers to a cycloalkyl group that includes at least one heteroatom selected from nitrogen, oxygen and sulfur.
  • the radicals may be fused with an aryl or heteroaryl.
  • the heterocycloalkyl is selected from oxazolidinonyl, pyrrolidinyl, tetrahydrofuranyl, tetrahydrothienyl, tetrahydropyranyl, tetrahydrothiopyranyl, piperidinyl, morpholinyl, thiomorpholinyl, piperazinyl, and indolinyl.
  • heteroalicyclic also includes all ring forms of the carbohydrates, including but not limited to the monosaccharides, the disaccharides and the oligosaccharides.
  • a heterocycloalkyl is a C 2 - Cioheterocycloalkyl.
  • a heterocycloalkyl is a C 4 -Ci oheterocycloalkyl.
  • a heterocycloalkyl contains 0-2 N atoms.
  • a heterocycloalkyl contains 0-2 N atoms, 0-2 O atoms or 0-1 S atoms.
  • bond refers to a chemical bond between two atoms, or two moieties when the atoms joined by the bond are considered to be part of larger substructure. In one aspect, when a group described herein is a bond, the referenced group is absent thereby allowing a bond to be formed between the remaining identified groups.
  • membered ring includes any cyclic structure. The term “membered” is meant to denote the number of skeletal atoms that constitute the ring.
  • cyclohexyl, pyridinyl, pyranyl and thiopyranyl are 6-membered rings and cyclopentyl, pyrrolyl, furanyl, and thienyl are 5-membered rings.
  • moiety refers to a specific segment or functional group of a molecule. Chemical moieties are often recognized chemical entities embedded in or appended to a molecule.
  • optionally substituted or “substituted” means that the referenced group may be substituted with one or more additional group(s) individually and independently selected from alkyl, cycloalkyl, aryl, heteroaryl, heteroalicyclic, hydroxy, alkoxy, aryloxy, alkylthio, arylthio, alkylsulfoxide, arylsulfoxide, alkylsulfone, arylsulfone, cyano, halo, carbonyl, thiocarbonyl, nitro, haloalkyl, fluoroalkyl, and amino, including mono- and di-substituted amino groups, and the protected derivatives thereof.
  • an optional substituents may be halide, -CN, -NO 2 , or
  • each R 8 is selected from H, alkyl, fluoroalkyl, heteroalkyl, cycloalkyl, aryl, heteroaryl, or heterocycloalkyl.
  • the protecting groups that may form the protective derivatives of the above substituents may be found in sources such as Greene and Wuts, above.
  • optional substituents are selected from halogen, CF 3 , OH, CN, NO 2 , SO 3 H, SO 2 NH 2 , SO 2 Me,
  • the compounds presented herein possess one or more stereocenters and each center independently exists in either the R or S configuration.
  • the compounds presented herein include all diastereomeric, enantiomeric, and epimeric forms as well as the appropriate mixtures thereof.
  • Stereoisomers are obtained, if desired, by methods such as, the separation of stereoisomers by chiral chromatographic columns.
  • compounds having the structure of Formula (I) may exist as tautomers. All tautomers are included within the scope of the compounds presented herein.
  • the compounds described herein exist in solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like. In other embodiments, the compounds described herein exist in unsolvated form.
  • kinase-dependent refers to conditions or disorders that would not occur, or would not occur to the same extent, in the absence of a kinase enzyme.
  • kinase-mediated refers to refers to conditions or disorders that might occur in the absence of kinase enzyme but can occur in the presence of kinase enzyme.
  • asthma refers to any disorder of the lungs characterized by variations in pulmonary gas flow associated with airway constriction of whatever cause (intrinsic, extrinsic, or both; allergic or non-allergic).
  • asthma may be used with one or more adjectives to indicate cause.
  • bone disease refers to a disease or condition of the bone, including, but not limited to, inappropriate bone remodeling, loss or gain, osteopenia, osteomalacia, osteofibrosis, and Paget' s disease.
  • cardiac disease refers to diseases affecting the heart or blood vessels or both, including but not limited to: arrhythmia (atrial or ventricular or both); atherosclerosis and its sequelae; angina; cardiac rhythm disturbances; myocardial ischemia; myocardial infarction; cardiac or vascular aneurysm; vasculitis, stroke; peripheral obstructive arteriopathy of a limb, an organ, or a tissue; reperfusion injury following ischemia of the brain, heart or other organ or tissue; endotoxic, surgical, or traumaticshock; hypertension, valvular heart disease, heart failure, abnormal blood pressure; shock; vasoconstriction (including that associated with migraines); vascular abnormality, inflammation, insufficiency limited to a single organ or tissue.
  • cancer refers to an abnormal growth of cells which tend to proliferate in an uncontrolled way and, in some cases, to metastasize (spread).
  • types of cancer include, but are not limited to, solid tumors (such as those of the bladder, bowel, brain, breast, endometrium, heart, kidney, lung, lymhatic tissue (lymphoma), ovary, pancreas or other endocrine organ (thyroid), prostate, skin (melanoma) or hematological tumors (such as the leukemias).
  • the terms "co-administration” or the like, as used herein, are meant to encompass administration of the selected therapeutic agents to a single patient, and are intended to include treatment regimens in which the agents are administered by the same or different route of administration or at the same or different time.
  • the term "dermatological disorder,” as used herein refers to a skin disorder. Such dermatological disorders include, but are not limited to, proliferative or inflammatory disorders of the skin such as, atopic dermatitis, bullous disorders, collagenoses, contact dermatitis eczema, Kawasaki Disease, rosacea, Sjogren-Larsso Syndrome, urticaria.
  • the terms “enhance” or “enhancing,” as used herein, means to increase or prolong either in potency or duration a desired effect.
  • the term “enhancing” refers to the ability to increase or prolong, either in potency or duration, the effect of other therapeutic agents on a system.
  • An “enhancing-effective amount,” as used herein, refers to an amount adequate to enhance the effect of another therapeutic agent in a desired system.
  • the term "iatrogenic" means a condition, disorder, or disease created or worsened by medical or surgical therapy.
  • inflammatory disorders refers to those diseases or conditions that are characterized by one or more of the signs of pain, heat, redness, swelling, and loss of function (temporary or permanent). Inflammation takes many forms and includes, but is not limited to, inflammation that is one or more of the following: acute, adhesive, atrophic, catarrhal, chronic, cirrhotic, diffuse, disseminated, exudative, fibrinous, fibrosing, focal, granulomatous, hyperplastic, hypertrophic, interstitial, metastatic, necrotic, obliterative, parenchymatous, plastic, productive, proliferous, pseudomembranous, purulent, sclerosing, seroplastic, serous, simple, specific, subacute, suppurative, toxic, traumatic, and/or ulcerative.
  • Inflammatory disorders further include, without being limited to those affecting the blood vessels (polyarteritis, temporal arteritis); joints (arthritis: crystalline, osteo-, psoriatic, reactive, rheumatoid, Reiter's); gastrointestinal tract (colitis); skin (dermatitis); or multiple organs and tissues (systemic lupus erythematosus).
  • the term "immunological disorders” refers to those diseases or conditions that are characterized by inappropriate or deleterious response to an endogenous or exogenous antigen that may result in cellular dysfunction or destruction and consequently dysfunction or destruction of an organ or tissue and which may or may not be accompanied by signs or symptoms of inflammation.
  • kit and "article of manufacture” are used as synonyms.
  • a "metabolite” of a compound disclosed herein is a derivative of that compound that is formed when the compound is metabolized.
  • active metabolite refers to a biologically active derivative of a compound that is formed when the compound is metabolized.
  • metabolism refers to the sum of the processes (including, but not limited to, hydrolysis reactions and reactions catalyzed by enzymes) by which a particular substance is changed by an organism. Thus, enzymes may produce specific structural alterations to a compound.
  • cytochrome P450 catalyzes a variety of oxidative and reductive reactions while uridine diphosphate glucuronyltransferases catalyze the transfer of an activated glucuronic-acid molecule to aromatic alcohols, aliphatic alcohols, carboxylic acids, amines and free sulphydryl groups.
  • Metabolites of the compounds disclosed herein are optionally identified either by administration of compounds to a host and analysis of tissue samples from the host, or by incubation of compounds with hepatic cells in vitro and analysis of the resulting compounds.
  • neurogenerative disease or "nervous system disorder,” as used herein, refers to conditions that alter the structure or function of the brain, spinal cord or peripheral nervous system, including but not limited to Alzheimer's Disease, cerebral edema, cerebral ischemia, multiple sclerosis, neuropathies, Parkinson's Disease, those found after blunt or surgical trauma (including post-surgical cognitive dysfunction and spinal cord or brain stem injury), as well as the neurological aspects of disorders such as degenerative disk disease and sciatica.
  • CNS refers to disorders of the central nervous system, i.e., brain and spinal cord.
  • subject or “patient” encompasses mammals and non-mammals.
  • mammals include, but are not limited to, any member of the Mammalian class: humans, non-human primates such as chimpanzees, and other apes and monkey species; farm animals such as cattle, horses, sheep, goats, swine; domestic animals such as rabbits, dogs, and cats; laboratory animals including rodents, such as rats, mice and guinea pigs, and the like.
  • non-mammals include, but are not limited to, birds, fish and the like.
  • the mammal is a human.
  • treat include alleviating, abating or ameliorating a disease or condition symptoms, preventing additional symptoms, ameliorating or preventing the underlying metabolic causes of symptoms, inhibiting the disease or condition, e.g., arresting the development of the disease or condition, relieving the disease or condition, causing regression of the disease or condition, relieving a condition caused by the disease or condition, or stopping the symptoms of the disease or condition either prophylactically and/or therapeutically.
  • amelioration of the symptoms of a particular disease, disorder or condition by administration of a particular compound or pharmaceutical composition refers to any lessening of severity, delay in onset, slowing of progression, or shortening of duration, whether permanent or temporary, lasting or transient that can be attributed to or associated with administration of the compound or composition.
  • the compounds of Formula (I) inhibit one or more protein kinase(s). In one aspect, the compounds of Formula (I) demonstrate anti-proliferative activity and are useful in the treatment of proliferative disorders. In one aspect, the compounds of Formula (I) demonstrate anti-proliferative activity and are useful in the treatment of proliferative disorders such as cancers, leukaemias and other disorders associated with uncontrolled cellular proliferation such as psoriasis and restenosis.
  • an anti-proliferative effect within the scope of the present disclosure is the ability to inhibit cell proliferation in an in vitro whole cell assay.
  • the in vitro whole cell assay uses any one of the cell lines selected from A549, A2780, HepG2, HCT-116, HeLa, HT29, MCF-7, NCI-H460, and Saos-2.
  • an antiproliferative effect within the scope of the present disclosure is the ability to inhibit at least one CDK enzyme, such as CDKl, CDK2, CDK4, CDK5, CDK6, CDK7, CDK8, CDK9, CDKIl, or other protein kinases; or inhibition of the interaction between HDM2 and p53 in an appropriate assay.
  • the compounds of Formula (I) are used in the treatment of proliferative disorders.
  • the proliferative disorder is a cancer or leukaemia.
  • the term proliferative disorder is used herein in a broad sense to include any disorder that requires control of the cell cycle, for example cardiovascular disorders, auto-immune disorders, dermatological disorders, inflammatory, fungal, parasitic disorders, emphysema and alopecia.
  • the compounds of Formula (I) induce apoptosis or maintain stasis in these disorders within the appropriate cells as required.
  • cardiovascular disorders include, but are not limited to, restenosis and cardiomyopathy.
  • auto-immune disorders include, but are not limited to, glomerulonephritis and rheumatoid arthritis.
  • dermatological disorders include, but are not limited to, psoriasis.
  • parasitic disorders include, but are not limited to, malaria.
  • the compounds of Formula (I) are used in the treatment of a disease, disorder, or condition mediated by one or more protein kinase selected from a CDK, aurora kinase, GSK, PLK and one of Tyrosine kinases. In one aspect, the compounds of Formula (I) inhibit such protein kinases.
  • the compounds of Formula (I) inhibit any of the steps or stages in the cell cycle, for example, formation of the nuclear envelope, exit from the quiescent phase of the cell cycle (GO), Gl progression, chromosome decondensation, nuclear envelope breakdown, START, initiation of DNA replication, progression of DNA replication, termination of DNA replication, centrosome duplication, G2 progression, activation of mitotic or meiotic functions, chromosome condensation, centrosome separation, microtubule nucleation, spindle formation and function, interactions with microtubule motor proteins, chromatid separation and segregation, inactivation of mitotic functions, formation of contractile ring, and cytokinesis functions.
  • compounds of Formula (I) influence certain gene functions such as chromatin binding, formation of replication complexes, replication licensing, phosphorylation or other secondary modification activity, proteolytic degradation, microtubule binding, actin binding, septin binding, microtubule organising centre nucleation activity and binding to components of cell cycle signalling pathways.
  • compounds of Formula (I) are useful in the treatment of a proliferative disorder, disease or condition mediated by at least one of a CDK, Auora kinase and/or PLK.
  • the proliferative disorder, disease or condition mediated by at least one of a CDK, Auora kinase and/or PLK include, but are not limited to cancers, leukaemias and other disorders associated with uncontrolled cellular proliferation such as psoriasis and restenosis, a viral disorder, a cardiovascular disease, a CNS disorder, an autoimmune disease, a bone disease, a hormone-related disease, a metabolic disorder, stroke, alopecia, an inflammatory disease or an infectious disease.
  • the compounds of Formula (I) inhibit one or more of the host cell kinases involved in cell proliferation, viral replication, a cardiovascular disorder, neurodegeneration, autoimmunity, a metabolic disorder, stroke, alopecia, an inflammatory disease or an infectious disease.
  • a proliferative disorder requires treatment of a susceptible neoplasm and may be selected from the group consisting of chronic lymphocytic leukaemia, lymphoma, leukaemia, breast cancer, lung cancer, prostate cancer, colon cancer, melanoma, pancreatic cancer, ovarian cancer, squamous carcinoma, carcinoma of head and neck, endometrial cancer, and aesophageal carcinoma.
  • the compounds, compositions and methods provided herein are useful for the treatment and prevention of cancer including solid tumors such as skin, breast, brain, cervical carcinomas, testicular carcinomas, etc.
  • solid tumors such as skin, breast, brain, cervical carcinomas, testicular carcinomas, etc.
  • compounds described herein are useful for treating cancer.
  • cancers include, but are not limited to:
  • Cardiac cancers such as, but not limited to, sarcoma (angiosarcoma, fibrosarcoma, rhabdomyosarcoma, liposarcoma), myxoma, rhabdomyoma, fibroma, lipoma and teratoma;
  • Lung cancers such as, but not limited to, bronchogenic carcinoma (squamous cell, undifferentiated small cell, undifferentiated large cell, non-small cell, adenocarcinoma), alveolar (bronchiolar) carcinoma, bronchial adenoma, sarcoma, lymphoma, chondromatous hamartoma, mesothelioma;
  • bronchogenic carcinoma squamous cell, undifferentiated small cell, undifferentiated large cell, non-small cell, adenocarcinoma
  • alveolar (bronchiolar) carcinoma bronchial adenoma
  • sarcoma sarcoma
  • lymphoma chondromatous hamartoma
  • mesothelioma mesothelioma
  • Gastrointestinal cancers such as, but not limited to: esophagus (squamous cell carcinoma, adenocarcinoma, leiomyosarcoma, lymphoma), stomach (carcinoma, lymphoma, leiomyosarcoma), pancreas (ductal adenocarcinoma, insulinoma, glucagonoma, gastrinoma, carcinoid tumors, vipoma), small bowel (adenocarcinoma, lymphoma, carcinoid tumors, Karposi's sarcoma, leiomyoma, hemangioma, lipoma, neurofibroma, fibroma), large bowel (adenocarcinoma, tubular adenoma, villous adenoma, hamartoma, leiomyoma), rectal, colorectal and colon; [00239] Genitourinary tract cancers, such as,
  • Liver cancers such as, but not limited to: hepatoma (hepatocellular carcinoma), cholangiocarcinoma, hepatoblastoma, angiosarcoma, hepatocellular adenoma, hemangioma;
  • Bone cancers such as, but not limited to: osteogenic sarcoma (osteosarcoma), fibrosarcoma, malignant fibrous histiocytoma, chondrosarcoma, Ewing's sarcoma, malignant lymphoma (reticulum cell sarcoma), multiple myeloma, malignant giant cell tumor chordoma, osteochronfroma (osteocartilaginous exostoses), benign chondroma, chondroblastoma, chondromyxofibroma, osteoid osteoma and giant cell tumors;
  • osteogenic sarcoma osteosarcoma
  • fibrosarcoma malignant fibrous histiocytoma
  • chondrosarcoma chondrosarcoma
  • Ewing's sarcoma malignant lymphoma
  • multiple myeloma malignant giant cell tumor chordoma
  • osteochronfroma osteo
  • Nervous system cancers such as, but not limited to: skull (osteoma, hemangioma, granuloma, xanthoma, osteitis deformans), meninges (meningioma, meningiosarcoma, gliomatosis), brain (astrocytoma, medulloblastoma, glioma, ependymoma, germinoma [pinealoma], glioblastoma multiform, oligodendroglioma, schwannoma, retinoblastoma, congenital tumors), spinal cord neurofibroma, meningioma, glioma, sarcoma);
  • skull osteoma, hemangioma, granuloma, xanthoma, osteitis deformans
  • meninges meningioma, meningiosarcoma, gliomatosis
  • Gynecological cancers such as, but not limited to: uterus (endometrial carcinoma), cervix (cervical carcinoma, pre-tumor cervical dysplasia), ovaries (ovarian carcinoma [serous cystadenocarcinoma, mucinous cystadenocarcinoma, unclassified carcinoma], granulosa-thecal cell tumors, Sertoli-Leydig cell tumors, dysgerminoma, malignant teratoma), vulva (squamous cell carcinoma, intraepithelial carcinoma, adenocarcinoma, fibrosarcoma, melanoma), vagina (clear cell carcinoma, squamous cell carcinoma, botryoid sarcoma (embryonal rhabdomyosarcoma), fallopian tubes (carcinoma);
  • Hematologic cancers such as, but not limited to: blood (myeloid leukemia [acute and chronic], acute lymphoblastic leukemia, chronic lymphocytic leukemia, myeloproliferative diseases, multiple myeloma, myelodysplastic syndrome), Hodgkin's disease, non-Hodgkin's lymphoma [malignant lymphoma];
  • Skin cancers such as, but not limited to: malignant melanoma, basal cell carcinoma, squamous cell carcinoma, head and neck squamous cell carcinomas, Karposi's sarcoma, moles dysplastic nevi, lipoma, angioma, dermatofibroma, keloids, psoriasis; and Adrenal glands: neuroblastoma.
  • cancerous cell includes a cell afflicted by any one of the above-identified conditions.
  • compound described herein are useful for treating or preventing cancer selected from: head and neck squamous cell carcinomas, histiocytic lymphoma, lung adenocarcinoma, small cell lung cancer, non- small cell lung cancer, pancreatic cancer, papillary renal carcinoma, liver cancer, gastric cancer, colon cancer, multiple myeloma, glioblastomas and breast carcinoma.
  • compounds described herein are useful for the prevention or modulation of the metastases of cancer cells and cancer.
  • compounds described herein are useful to prevent or modulate the metastases of ovarian cancer, childhood hepatocellular carcinoma, metastatic head and neck squamous cell carcinomas, gastric cancers, breast cancer, colorectal cancer, cervical cancer, lung cancer, nasopharyngeal cancer, pancreatic cancer, glioblastoma and sarcomas.
  • a further embodiment relates to the use of compounds of Formula (I) described herein, or pharmaceutically acceptable salts thereof, in the manufacture of a medicament capable of treating a viral disorder mediated by one or more of the host cell CDKs involved in viral replication.
  • the one or more CDKs are selected from among CDKl, CDK2, CDK4, CDK7, CDK8, CDK9 and CDKI l.
  • the viral disorder is selected from among human cytomegalovirus (HCMV), herpes simplex virus type 1 (HSV-I), human immunodeficiency virus type 1 (HIV-I), and varicella zoster virus (VZV).
  • HCMV human cytomegalovirus
  • HSV-I herpes simplex virus type 1
  • HV-I human immunodeficiency virus type 1
  • VZV varicella zoster virus
  • CDK dependent disorders are associated with an above normal level of activity of one or more CDK enzymes.
  • such disorders are typically associated with an abnormal level of activity of CDKl, CDK2, CDK4, CDK6, CDK7, CDK8, CDK9 and/or
  • a CDK sensitive disorder is a disorder in which an aberration in the CDK level is not the primary cause, but is downstream of the primary metabolic aberration.
  • CDK9 and/or CDKl 1 can be said to be part of the sensitive metabolic pathway and inhibitors of one or more of these
  • CDKs are therefore useful in treating such disorders.
  • the compound of Formula (I) is capable of inhibiting one or more CDKs selected from among CDK2, CDK7, and CDK9.
  • compounds of Formula (I) are used in the treatment of cardiovascular diseases mediated by one or more CDKs.
  • a cardiovascular disease may be selected from the group consisting of ischaemic heart disease
  • Cardiac hypertrophy is characterized by global increases in mRNA and protein synthesis. CDK9 activity has been demonstrated to be necessary for hypertrophy in cardiomyocytes. Heart-specific activation of CDK9 by cyclin Tl was found to provoke hypertrophy.
  • compounds of Formula (I) inhibit one or more CDKs and are therefore useful in the prevention and treatment of cardiac hypertrophy.
  • Yet another embodiment relates to the use of a compound of Formula (I) in the treatment of neurodegenerative disorders mediated by one or more GSKs or CDKs.
  • the neurodegenerative disorder is
  • Tau is a GSK-3 substrate which has been implicated in the etiology of Alzheimer's disease. In healthy nerve cells, Tau co-assembles with tubulin into microtubules. However, in Alzheimer's disease, tau forms large tangles of filaments, which disrupt the microtubule structures in the nerve cell, thereby impairing the transport of nutrients as well as the transmission of neuronal messages.
  • GSK3 inhibitors prevent and/or reverse the abnormal hyperphosphorylation of the microtubule-associated protein tau that is an invariant feature of Alzheimer's disease and a number of other neurodegenerative diseases, such as progressive supranuclear palsy, corticobasal degeneration and
  • compounds of Formula (I) are used in the treatment a metabolic disorder mediated by one or more GSKs.
  • Metabolic disorders include Type II diabetes (non insulin dependent diabetes mellitus) and diabetic neuropathy.
  • compounds of Formula (I) inhibit GSK-3, which is implicated in Type II diabetes.
  • GSK3 is one of several protein kinases that phosphorylate glycogen synthase (GS) and is involved in the stimulation of glycogen synthesis by insulin in skeletal muscle. GSK3's action on GS thus results in the latter' s deactivation and thus suppression of the conversion of glucose into glycogen in muscles.
  • Type II diabetes non-insulin dependent diabetes mellitus
  • Type II diabetes is a multi-factorial disease.
  • Hyperglycaemia is due to insulin resistance in the liver, muscles, and other tissues, coupled with impaired secretion of insulin.
  • Skeletal muscle is the main site for insulin- stimulated glucose uptake, there it is either removed from circulation or converted to glycogen.
  • Muscle glycogen deposition is the main determinant in glucose homeostasis and type II diabetics have defective muscle glycogen storage. There is evidence that an increase in GSK3 activity is important in type II diabetes.
  • compounds of Formula (I) are used in the treatment of bipolar disorder mediated by one or more kinases.
  • compounds of Formula (I) are used for treating a stroke mediated by one or more GSKs.
  • Reducing neuronal apoptosis is an important therapeutic goal in the context of head trauma, stroke, epilepsy, and motor neuron disease.
  • GSK3 as a pro-apoptotic factor in neuronal cells makes this protein kinase an attractive therapeutic target for the design of inhibitory drugs to treat these diseases.
  • compounds of Formula (I) are used in the treatment of alopecia mediated by one or more GSKs.
  • the ectopic application of GSK3 inhibitors is therapeutically useful in the treatment of baldness and in restoring hair growth following chemotherapy-induced alopecia.
  • a condition is a GSK3 -dependent disorder, said method comprising administering to a subject in need thereof, a compound of Formula (I) in an amount sufficient to inhibit GSK3.
  • the compound of Formula (I) is administered in an amount sufficient to inhibit GSK3 ⁇ .
  • a method of treating a PLK-dependent disease, disorder and/or condition comprising administering to a subject in need thereof, a compound of the Formula (I) in an amount sufficient to inhibit a PLK.
  • human PLKs regulate fundamental aspects of mitosis. Both PLKl and PLK2 may have additional post-mitotic functions. Deregulated PLK expressions result in cell cycle arrest and apoptosis.
  • Compounds of Formula (I) are used in treating PLK-mediated diseases, disorders and/or conditions.
  • the PLK-mediated diseases, disorders and/or conditions include proliferative disorders.
  • the compounds of Formula (I) inhibit one or more PLK enzymes selected from PLKl, PLK2, PLK3 and PLK4
  • a method of treating an aurora kinase-dependent disease, disorder and/or condition comprising administering to a subject in need thereof, a compound of Formula (I).
  • the compound of Formula (I) inhibits one or more aurora kinases selected from among aurora kinase A, aurora kinase B or aurora kinase C.
  • a method of treating a tyrosine kinase-dependent disorder comprising administering to a subject in need thereof, a compound of Formula (I) in an amount sufficient to inhibit a tyrosine kinase.
  • compounds of Formula (I) are administered to a subject in an amount sufficient to inhibit at least one protein kinase selected from BCR-ABL, IKK, FLT3, JAK, LCK, PDGF, Src, and VEGF.
  • described herein is a method of selectively treating a protein kinase-dependent disorder, said method comprising administering to a subject in need thereof, a compound of the invention or a pharmaceutically acceptable salt thereof, as defined above in an amount sufficient to inhibit a selected protein kinase.
  • said method comprises contacting said protein kinase with a compound described herein.
  • the compound of Formula (I) is administered in an amount sufficient to inhibit at least one of a CDK, GSK, aurora kinase, or PLK, or a tyrosine kinase including, but not limiting to BCR-ABL, IKK, FLT3,
  • JAK JAK, LCK, PDGF, Src, or VEGF.
  • the protein kinase is a CDK. In one aspect, the protein kinase is CDKl, CDK2, CDK3,
  • CDK4 CDK5, CDK6, CDK7, CDK8, CDK9 or CDKl 1.
  • the protein kinase is CDKl, CDK2, CDK7 or CDK9.
  • the compound of Formula (I) inhibits at least one CDK enzyme, preferably at least one of
  • CDKl CDK2, CDK7 and CDK9.
  • the compound of Formula (I) inhibits a CDK.
  • the compound of Formula (I) inhibits a CDK selected from CDKl, CDK2, CDK7 and CDK9 at sub-micromolar IC 50 values, more preferably at IC 50 of less than 0.5 micromolar, more preferably less than 0.25 micromolar.
  • the compound of Formula (I) inhibits CDK9 at IC 50 of less than 0.1 micromolar or less than 0.05 micromolar.
  • the compound of Formula (I) demonstrates an antiproliferative effect in human cell lines, as measured by a standard 72h MTT cytotoxicity assay. In some embodiments, the compound of Formula (I) exhibits an IC 50 value of less than 1 micromolar.
  • a method of treating a proliferative disease or disorder, a viral disorder, a cardiovascular disease, a CNS disorder, an autoimmune disease, a metabolic disorder, stroke, alopecia, an inflammatory disease or an infectious disease comprising administering to a subject in need thereof, a compound of Formula (I) as hereinbefore defined in an effective amount.
  • a compound of the invention in the manufacture of a medicament as hereinbefore defined includes the use of the compound directly, or in any stage of the manufacture of such a medicament, or in vitro in a screening programme to identify further agents for the prevention or treatment of the hereinbefore defined diseases or conditions.
  • a further aspect relates to the use of a compound of Formula (I) or a pharmaceutically acceptable salt or solvate or physiologically hydrolysable, solubilising or immobilising derivative thereof, in an assay for identifying candidate compounds capable of treating one or more disorders or diseases as hereinbefore defined.
  • a compound is of use in identifying candidate compounds capable of inhibiting a protein kinase, more preferably one or more of a CDK, aurora kinase, GSK, PLK or tyrosine kinase enzyme.
  • Suitable routes of administration include, but are not limited to, oral, intravenous, rectal, aerosol, parenteral, ophthalmic, pulmonary, transmucosal, transdermal, vaginal, otic, nasal, and topical administration.
  • parenteral delivery includes intramuscular, subcutaneous, intravenous, intramedullary injections, as well as intrathecal, direct intraventricular, intraperitoneal, intralymphatic, and intranasal injections.
  • a compound as described herein is administered in a local rather than systemic manner, for example, via injection of the compound directly into an organ, often in a depot preparation or sustained release formulation.
  • long acting formulations are administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection.
  • the drug is delivered in a targeted drug delivery system, for example, in a liposome coated with organ-specific antibody.
  • the liposomes are targeted to and taken up selectively by the organ.
  • the compound as described herein is provided in the form of a rapid release formulation, in the form of an extended release formulation, or in the form of an intermediate release formulation.
  • the compound described herein is administered topically.
  • kits and articles of manufacture are also described herein.
  • Such kits can comprise a carrier, package, or container that is compartmentalized to receive one or more containers such as vials, tubes, and the like, each of the container(s) comprising one of the separate elements to be used in a method described herein.
  • Suitable containers include, for example, bottles, vials, syringes, and test tubes.
  • the containers are formed from any acceptable material including, e.g., glass or plastic.
  • the container(s) can comprise one or more compounds described herein, optionally in a composition or in combination with another agent as disclosed herein.
  • the container(s) optionally have a sterile access port (for example the container can be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
  • kits optionally comprising a compound with an identifying description or label or instructions relating to its use in the methods described herein.
  • a kit will typically comprise one or more additional containers, each with one or more of various materials
  • Non-limiting examples of such materials include, but not limited to, buffers, diluents, filters, needles, syringes; carrier, package, container, vial and/or tube labels listing contents and/or instructions for use, and package inserts with instructions for use. A set of instructions will also typically be included.
  • a label can be on or associated with the container.
  • a label can be on a container when letters, numbers or other characters forming the label are attached, molded or etched into the container itself; a label can be associated with a container when it is present within a receptacle or carrier that also holds the container, e.g., as a package insert.
  • a label can be used to indicate that the contents are to be used for a specific therapeutic application. The label can also indicate directions for use of the contents, such as in the methods described herein.
  • TLC thin-layer chromatography
  • silica gel G60 alumina plates coated with silica gel G60. Developed plates were air dried and analyzed under a UV lamp (254 / 365 run). Silica gel (EM Kieselgel 60, 0.040-0.063 mm, Merck) or ISOLUTE pre-packed columns was used for flash chromatography.
  • EXAMPLE 14 4-(4-(l-Propyl-l//-pyrrolo[2,3- ⁇ ]pyridin-3-yl)pyrimidin-2-ylamino)benzenesulfonamide (Compound 23)
  • EXAMPLE 16 (4-Methylpiperazin-l-yl)(3-(4-(l-propyl-l ⁇ -pyrrolo[2,3-b]pyridin-3-yl)pyrimidin-2- ylamino)phenyl)methanone (Compound 34)
  • reaction mixture was purified by column chromatography using EtOAc/PE (1: 1, v/v) to elute 3-(4-(5-chloro-7-methyl-l-propyl-l/f-pyrrolo[3,2-£]pyridin-3- yl)pyrimidin-2-ylamino)benzenesulfonamide which was re-crystallised from EtOAc to give white solid.
  • EXAMPLE 24 4-(7-Chloro-l-propyl-l#-pyrrolo[2,3-c]pyridin-3-yl)-7V-(4-methyl-3- (morpholinosulfonyl)phenyl)pyrimidin-2-amine (Compound 101)
  • Kinase dosage and Z'-LYTETM substrate for each kinase assay is as follows: CDK2/cyclinA-8ng/reaction, Ser/Thr 12 Peptide; CDKl/cyclinB-0.8ng/reaction, Ser/Thr 12 Peptide, CDK5/p35-0.8ng/reaction, Ser/Thr 12 Peptide, and Aurora A- long/reaction, Ser/Thr 1 Peptide. All enzymatic assays were performed using Z'-LYTETM Kinase Assay Kits and were carried out following protocols provided by Invitrogen. To prepare stock solutions, compounds were dissolved at 1OmM in 100% DMSO and stored at 4°C in amber glass vials.
  • Enzyme reactions were initiated by adding recombinant enzyme and Z'-LYTETM Ser/Thr peptide substrate and ATP mixture to 384-well plates containing diluted test compounds.
  • Kinase and corresponding peptide substrate were incubated for lhr at room temperature in the presence of varying amounts of test compound with reaction buffer (20 ⁇ M ATP, 25 mM HEPES [pH 7.5], 5 niM MgCl 2 , 0.5 niM EGTA, 0.005% Brij-35). Development buffer was added and incubated for another one hour at room temperature.
  • HeLa cells Human cervix adenocarcinoma cell line, ATCC NO, CCL-2
  • A549 cells Human lung carcinoma cells line, ATCC NO, CCL- 185
  • U2OS cells Human osteosarcoma cell line, ATCC NO, HTB-96
  • Eca- 109 cells Human esophageal carcinoma cell line
  • HepG2 Human hepatocellular liver carcinoma cell line, ATCC NO, HB-8065
  • K562 cells Human chronic myelogenous leukemia cell line, ATCC NO, CCL-243
  • SK- MEL-28 Human malignant melanoma cell line, ATCC NO, HTB-72
  • Molt-4 Human acute lymphoblastic leukemia cell line, ATCC NO, CRL-1582
  • H460 Human large cell lung cancer cell line, ATCC NO
  • HepG2, MDA-MB-231, WI-38 and MRC-5 cells were obtained from Shanghai Institute of Cell Biology, Chinese Academy of Sciences (Shanghai 200031, China).
  • A549, HeLa, K562, MCF7, ACHN, H460, U2OS, Eca-109, MCF7, HL60, THP-I, Jurkat, HCT-116, HT-29, PC-3M, SK-MEL-28, Molt-4, H460 and ACHN cells were purchased from ATCC (American Type Culture Collection, USA).
  • Standard 72h MTT thiazolye blue; 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide
  • 72h MTT thiazolye blue; 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide
  • MTT solution (5 mg/ml) was added at 10 ⁇ l per well and incubated in the dark at 37°C for 4 hr. Culture medium and MTT solution was removed. MTT dye was solubilised with 100 ⁇ l per well of DMSO with agitation. Absorbance was read at 562 nm and data analyzed using Sigmoid curve-fitting software Origin 6.0 (MicroCal, LLC, USA) to determine IC 50 value. Antiproliferative activity of representative compounds disclosed herein is presented in Table II.
  • mice Female BALB/c nude mice from SHANGHAI SLAC LABORATORY ANIMAL CO with age of 5 to 6 weeks and body weight of 19 to 20 g were used in the study. Autoclaved water and irradiated food were provided ad libitum, and animals were maintained on a 12-hour light and 12-hour dark cycle. Cages, bedding, and water bottles were autoclaved before use and changed weekly. The health of all animals was determined by daily gross observation of experimental animals, and analyses of blood samples of sentinel animals housed on shared shelf racks. All animals were allowed to acclimate and recover from any shipping-related stress for a minimum of 72 hours before experimental use. All animal experiments were conducted according to the guidelines of Chinese.
  • Xenograft tumor models were established with human tumor cell lines.
  • HT-29 Human colon cancer cell
  • H460 Human large cell lung cancer cell line
  • Molt-4 Human acute lymphoblastic leukemia cell line
  • PC-3M Human prostate adenocarcinoma cell line
  • HL60 Human acute promyelocytic leukemia cell line
  • Tumor growth was monitored twice per week until the tumor reached to a volume of 100 mm 3 .
  • the day before the first administration of a compound tumor bearing animals were stratified into several groups according to tumor volume.. Only animals with appropriate tumor volumes were selected and randomly distributed to treatment and control groups. Each group consisted of 8 mice.
  • the synergistic action or enhancement of conventional chemotherapeutic agent by a test compound can also be determined with this model by comparing animals treated with the standard therapy alone to animals treated with test compound plus the same standard therapy. An additive effect on the delay of tumor growth will be observed if synergistic action due to test compound is occurring.
  • the concentration of the test compound in the dosing solution was reconfirmed by HPLC method. Then the test compound (10, 100 mg/kg) was given orally in a volume of 0.1 mL/10 g body weight to SD rats. Plasma samples were collected from retro-orbital puncture at pre-dose and 0.083, 0.25, 0.5, 1, 2, 4, 6, 8, and 24 hours post-dose. All samples and the dose formulation were stored at -20 0 C until bioanalysis. The concentrations of the test compound in plasma were determined using a high performance liquid chromatography/mass spectrometry (HPLC/MS/MS) method.
  • HPLC/MS/MS high performance liquid chromatography/mass spectrometry
  • ORR objective response rate
  • Formula (I) when administered every 2 weeks to patients with adenocarcinoma of the pancreas is to measure time-to-event variables including: time to objective tumor response for responding patients (TtOR), duration of response for responding patients, time to treatment failure (TtTF), time to progressive disease (TtPD), progression-free survival (PFS), overall survival (OS); the toxicities of therapy.
  • TtOR time to objective tumor response for responding patients
  • TtTF time to treatment failure
  • TtPD time to progressive disease
  • PFS progression-free survival
  • OS overall survival
  • the study is a multi-center, double-blind, randomized, placebo-controlled Phase 2 study. Tumor assessments will be repeated every 4 cycles (approximately 8 weeks). Patients will receive study therapy for 12 treatments, or until tumor progression is documented, unacceptable toxicity is experienced, the patient withdrew consent, or the patient is unable to fulfill the responsibilities of study participation as determined by the treating physician or the qualified investigator. After study discontinuation, patients who have not progressed will have tumor assessments performed approximately every 8 weeks until disease progression. Once patients have disease progression, patients will enter a post-study follow-up period, and will be followed every 12 weeks for 24 months for overall survival. Patients will also be followed for ongoing or any new toxicities.
  • Main inclusion criteria will include: histologically proven adenocarcinoma; performance Status of 0 or 1 on the Eastern Cooperative Oncology Group (ECOG) scale; a complete history and physical, chest x-ray, CT scan of abdomen and pelvis; barium enema, or colonoscopy.
  • Patients with pain will be requires to have had their pain stabilized for 1 week prior to commencing therapy.
  • Patients requiring opioids for pain control will be required to have been on a fixed analgesic regimen aimed to provide adequate pain control with no more than three breakthrough
  • supplemental doses of analgesics per day to control pain Patients will be requires to demonstrate adequate bone marrow reserve (i.e. Neutrophil count > 1.5 x 10 9 cells/L; Platelets ⁇ 100 x 10 9 cells/L). Patients will be required to have negative tumor markers for alpha-fetoprotein (AFP) and monoclonal antichorionic gonadotropin ( ⁇ -subunit)
  • AFP alpha-fetoprotein
  • ⁇ -subunit monoclonal antichorionic gonadotropin
  • Main exclusion criteria will include: prior chemotherapy; pregnancy or breastfeeding; inability or unwillingness to take folic acid, vitamin B 12 supplementation, or dexamethasone.
  • Compound of Formula (I) dosage will be 500 mg/m 2 and will be given as a 10-minute infusion on Day 1 of each 14-day cycle. Folic acid and vitamin B 12 supplementation, and dexamethasone (or equivalent corticosteroid) prophylaxis will also be administered.
  • Tumor response rate will be defined as the number of patients with documented partial response (PR) or complete response (CR) divided by the number of patients qualified for tumor response analysis.
  • Time-to-event analyses will be performed on the observed distributions of time to objective progressive disease, progression-free survival (PFS), time to treatment failure (TtTF), and overall survival (OS) using the Kaplan-Meier (K-M) method. All patients with best overall response of CR or PR will be analyzed for response duration by using the K-M method.
  • Safety analyses will include adverse event (AE) rates, serious AEs, vital signs, laboratory data, blood transfusions required, and deaths. Toxicities using laboratory and non-laboratory adverse events will be evaluated using the common terminology criteria for adverse events (CTCAE, version 3.0).
  • Statistical The primary analysis will be to estimate the objective best overall response rate and its 95% confidence interval (CI). Medians for each of the time-to-event endpoints, and time-to-event variables will be estimated using the K-M method. All estimates of treatment effects will be conducted at a two-sided alpha level of 0.05, and CI for all parameters will be estimated were to be constructed using a 95% level.
  • Laboratory requirements include the following: absolute granulocyte count > 1.5 x 10 /L; platelet count > 100> ⁇ 10 9 /L; AST ⁇ 2.5 ⁇ upper normal limit (UNL); serum creatinine ⁇ UNL; bilirubin ⁇ UNL. All patients must give informed consent according to the requirements of their local Institutional and/or University Human Experimentation Committee. Treatment
  • Compound of Formula (I) is prepared as a sterile 10 mg/ml solution. The individually calculated dose is diluted prior to infusion with 0.9% sodium chloride injection USP or 5% dextrose injection USP to final concentrations ranging from 0.09 to 0.5 mg/ml of compound of Formula (I).
  • Compound of Formula (I) is administered in the outpatient setting as a 1 hour infusion at a dose of 50 mg/m 2 daily ⁇ 3 days every 21 days. Vital signs are monitored every 30 minutes ⁇ 5 beginning at the start of infusion until 1 hour post infusion.
  • Antidiarrheal prophylaxis and antiemetic prophylaxis are prescribed as follows: 4 chewable Pepto Bismol tablets 1 hour before first dose of compound of Formula (I), then 2 tablets every 6 hours until 12 hours after the last dose of compound of Formula (I) (day 3), ondansetron 8 mg orally every 12 hours beginning 12 hours before treatment and continuing until 12 hours after the last dose of compound of Formula (I) (day 3).
  • Treatment continues until progression, or for 2 cycles after complete or stable partial response.
  • Nonresponding stable patients continue on treatment until progression, or alternatively are removed from therapy after 6 cycles at the investigator's discretion.
  • After termination from protocol treatment all patients are seen at 4 weeks, then every 3 months until progression or death.
  • Patients are considered evaluable for response if they have received at least 1 cycle of therapy and have their disease reevaluated. Patients are evaluated for response every 6 weeks (every 2 cycles) using the same investigations that demonstrated measurable disease at baseline. Response and progression are evaluated using the RECIST criteria (response evaluation criteria in solid tumors) (Therasse P, Arbuck SG, Eisenhauer EA, et al. New guidelines to evaluate the response to treatment in solid tumors (RECIST guidelines). Journal of the National Cancer Institute. 2000;92(3):205-216).
  • Patients are considered evaluable for toxicity after their first infusion of compound of Formula (I).
  • a history and physical examination is performed on day 1 of each cycle with an assessment of toxicity during the previous treatment interval.
  • CBC, platelets, differential, bilirubin and AST are performed on days 1, 8, and 15, and BUN, creatinine, electrolytes, LDH, fasting glucose, and alkaline phosphatase are performed on day 1 of each cycle.
  • Toxicities are graded according to the NCI Common Toxicity Criteria Version 2.0.
  • the primary endpoint of this phase II study is objective response rate, with secondary endpoints of toxicity, time to progression, early progression rate, and response duration.
  • a two-stage design is used for patient accrual. This design tests the null hypothesis (H 0 ) that the true response rate is ⁇ 5% versus the alternative hypothesis (H A ) that the true response rate is > 20%.
  • the significance level i.e., the probability of rejecting H 0 when it is true
  • the power is 0.865 when the true response probability is 20%. If there were no responses after the first 15 patients, the response rate is concluded to be ⁇ 5% and accrual was to be stopped. If there were one or more responses, accrual is continued to 30 evaluable patients.
  • PSA prostate-specific antigen
  • compound of Formula (I) is initially administered via a battery-powered ambulatory infusion pump connected to a central venous catheter as a 72 -h continuous infusion at the starting dose of 40 mg/m2/day.
  • the compound of Formula (I) cassette is changed daily with courses repeated every 14 days until disease progression or overt toxicity.
  • patients completing two courses at 40 mg/m2/day without ⁇ grade 3 toxicity (except alopecia) their dose could be escalated to 50 mg/m2/day at the discretion of the investigator. If the subsequent two courses are again tolerated, another dose escalation to 60 mg/m2/day could be performed.
  • the primary end points of this study are response rate and toxicity.
  • the complete blood counts and serum chemistries are repeated weekly for the first month, then at the beginning of each new course (every 2 weeks) of therapy.
  • Vitals signs, performance status, and toxicity are also assessed weekly for the first month and then at the beginning of each additional course of therapy as well.
  • PSA is obtained every 4 weeks, and computed tomography scans and/or bone scans were repeated every 12 weeks while on study. All patients receiving compound of Formula (I) are considered evaluable for toxicity (using National Cancer Institute Common Toxicity Criteria, version 1.0), but only those patients completing at least four courses of therapy (8 weeks) are considered evaluable for response.
  • a CR is defined as disappearance of all measurable disease and normalization of the bone scan for 4 weeks.
  • a PR is defined as ⁇ 50% decrease from baseline in the sum of the products of the maximum perpendicular diameters for indicator lesions with no progressive disease observed for at least 4 weeks. For patients with bone scan only abnormalities, a PR requires ⁇ _.50% decrease in the number of lesions.
  • Stable disease (SD) is defined as the absence of a CR, PR, or disease progression.
  • Progressive disease (PD) is defined as unequivocal increase of at least 25% in the size of any measurable lesion or the appearance of any new lesion. Serum PSA levels are not used in the response assessment.
  • PFS Progression-free survival
  • Example 35 Pharmaceutical Compositions
  • Example 35a Parenteral Composition
  • a parenteral pharmaceutical composition suitable for administration by injection 100 mg of a water-soluble salt of a compound of Formula (I) is dissolved in DMSO and then mixed with 10 mL of 0.9% sterile saline. The mixture is incorporated into a dosage unit form suitable for administration by injection.
  • a pharmaceutical composition for oral delivery 100 mg of a compound of Formula (I) is mixed with 750 mg of starch. The mixture is incorporated into an oral dosage unit for, such as a hard gelatin capsule, which is suitable for oral administration.
  • Example 35c Sublingual (Hard Lozenge) Composition
  • a pharmaceutical composition for buccal delivery such as a hard lozenge
  • a pharmaceutical composition for buccal delivery such as a hard lozenge
  • the mixture is gently blended and poured into a mold to form a lozenge suitable for buccal administration.
  • a fast-disintegrating sublingual tablet is prepared by mixing 48.5% by weigh of a compound of Formula (I), 44.5% by weight of microcrystalline cellulose (KG-802), 5% by weight of low-substituted hydroxypropyl cellulose (50 ⁇ m), and 2% by weight of magnesium stearate. Tablets are prepared by direct compression (AAPS PharmSciTech. 2006;7(2):E41). The total weight of the compressed tablets is maintained at 150 mg.
  • the formulation is prepared by mixing the amount of compound of Formula (I) with the total quantity of microcrystalline cellulose (MCC) and two- thirds of the quantity of low-substituted hydroxypropyl cellulose (L-HPC) by using a three dimensional manual mixer (lnversina ®, Bioengineering AG, Switzerland) for 4.5 minutes. All of the magnesium stearate (MS) and the remaining one-third of the quantity of L-HPC are added 30 seconds before the end of mixing.
  • MCC microcrystalline cellulose
  • L-HPC low-substituted hydroxypropyl cellulose
  • a pharmaceutical composition for inhalation delivery 20 mg of a compound of Formula (I) is mixed with 50 mg of anhydrous citric acid and 100 mL of 0.9% sodium chloride solution. The mixture is incorporated into an inhalation delivery unit, such as a nebulizer, which is suitable for inhalation administration.
  • an inhalation delivery unit such as a nebulizer
  • a pharmaceutical composition for rectal delivery 100 mg of a compound of Formula (I) is mixed with 2.5 g of methylcelluose (1500 mPa), 100 mg of methylparapen, 5 g of glycerin and 100 mL of purified water. The resulting gel mixture is then incorporated into rectal delivery units, such as syringes, which are suitable for rectal administration.
  • Example 35g Topical Gel Composition
  • 100 mg of a compound of Formula (I) is mixed with 1.75 g of hydroxypropyl cellulose, 10 mL of propylene glycol, 10 niL of isopropyl myristate and 100 niL of purified alcohol USP.
  • the resulting gel mixture is then incorporated into containers, such as tubes, which are suitable for topical administration.
  • a pharmaceutical opthalmic solution composition 100 mg of a compound of Formula (I) is mixed with 0.9 g of NaCl in 100 mL of purified water and filtered using a 0.2 micron filter. The resulting isotonic solution is then incorporated into ophthalmic delivery units, such as eye drop containers, which are suitable for ophthalmic administration.
  • ophthalmic delivery units such as eye drop containers
  • a pharmaceutical nasal spray solution 10 g of a compound of Formula (I) is mixed with 30 mL of a 0.05M phosphate buffer solution (pH 4.4). The solution is placed in a nasal administrator designed to deliver 100 ⁇ l of spray for each application.
  • a 0.05M phosphate buffer solution pH 4.4

Landscapes

  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Immunology (AREA)
  • Pulmonology (AREA)
  • Neurology (AREA)
  • Rheumatology (AREA)
  • Hematology (AREA)
  • Biomedical Technology (AREA)
  • Diabetes (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Neurosurgery (AREA)
  • Hospice & Palliative Care (AREA)
  • Obesity (AREA)
  • Pain & Pain Management (AREA)
  • Psychiatry (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Transplantation (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

Described herein are compounds that are inhibitors of one or more protein kinases. Also described are pharmaceutical compositions and medicaments that include the compounds described herein. Also described herein are methods of using such protein kinase inhibitors, alone and in combination with other compounds, for conditions or diseases mediated or dependent upon protein kinases.

Description

KINASE INHIBITORS AND THEIR USE AS PHARMACEUTICAL AGENTS
FIELD OF THE INVENTION
[0001] Described herein are compounds, methods of making such compounds, pharmaceutical compositions and medicaments comprising such compounds, and methods of using such compounds to treat, prevent or diagnose diseases, disorders or conditions associated with one or more protein kinases.
BACKGROUND OF THE INVENTION
[0002] Many diseases are associated with abnormal cellular responses triggered by protein kinase-mediated events. These diseases include, but are not limited to allergies, asthma, Alzheimer's disease, autoimmune diseases, bone diseases, cancer, cardiovascular diseases, inflammatory diseases, hormone-related diseases, metabolic diseases, neurological and neurodegenerative diseases. Described herein are protein kinase inhibitors that are effective as therapeutic agents.
SUMMARY OF THE INVENTION
[0003] Presented herein are compounds, pharmaceutical compositions, medicaments, and methods, for (a) diagnosing preventing, or treating conditions, disorders, or disorders associated with one or more protein kinases, (b) mitigating adverse signs and symptoms that are associated with one or more protein kinases, and/or (c) controlling conditions, disorders, or disorders associated with one or more protein kinases. These conditions, disorders, or disorders may arise from one or more of a genetic, iatrogenic, immunological, infectious, metabolic, oncological, toxic, surgical, and/or traumatic etiology. In one aspect, the methods, compounds, pharmaceutical compositions, and medicaments described herein comprise inhibitors of one or more protein kinases.
[0004] In one aspect is a compound having the structure of Formula (I), pharmaceutically acceptable salts, pharmaceutically acceptable solvates, or pharmaceutically acceptable prodrugs thereof:
Figure imgf000002_0001
Formula (I) wherein each X is -C(R3)- or -N-, provided that at least two X are -C(R3)-;
Z is -NRaC(=O)-, -C(=O)NRa-, -NR3SO2-, -SO2NR3-, -NR3-, -CH2NR3-, - NR3CH2-, -CH2-, -CH2CH2-, -CH=CH-, - CH2C(=O)NR3-, -NR3C(=O)CH2-, -SO2-, or -SO-; R3 is H or alkyl;
R1, R2, R3, R5, R6, R7, R8, R9, R10 and R11 are each independently selected from the group consisting of H, halogen, - NH2, -NO2, -CN, -OH, -CO2H, -CONH2, -SO3H, -SO2NH2, -SO2CH3, -OCH3, -CF3, -CH3, and -LA-LB-R32; LA is a covalent bond or an alkyl group; LB is a covalent bond, -0-, -NR32-, -NH-, -C(=0)-, -C(=0)0-, -0C(=0)-, - C(=O)NR32-, -C(=O)NH-, -NR32C(=O)-, -NHC(=O)-, -SO2-, -SO2NR32-, - NR32SO2-, -SO2NH-, or -NHSO2-; R32 is H, substituted or unsubstituted alkyl, haloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted non-aromatic heterocycle, or substituted or unsubstituted cycloalkyl; wherein any R32 group, when substituted, is substituted with one or more groups selected from halogen, -NH2, -NO2, -CN, -OH, -CO2H, -CONH2, -SO3H, -SO2NH2, -SO2CH3, -OCH3, and -CF3.
[0005] In some embodiments, Z is -NR3-. In one aspect, R3 is H. [0006] In one aspect, R5 and R6 are each H.
[0007] In some embodiments, R2 is H.
[0008] In some embodiments, each R3 is independently selected from the group consisting of H, halogen, -NH2, -NO2,
-CN, -OH, -CO2H, -CONH2, -SO3H, -SO2NH2, -SO2CH3, -OCH3, -CF3, and -CH3.
[0009] In some embodiments, R7, R8, R9, R10 and R11 are each independently selected from the group consisting of H, halogen, -NH2, -NO2, -CN, -OH, -CO2H, -CONH2, -SO3H, -SO2NH2, -SO2CH3, -OCH3, -CF3, -CH3, and -LA-LB-R32; LA is a covalent bond or an alkyl group; LB is a covalent bond, -0-, -NR32-, -NH-, -C(=0)0-, -C(=O)NH-, or -SO2NH-;
R32 is H, substituted or unsubstituted alkyl, haloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl; wherein any R32 group, when substituted, is substituted with one or more groups selected from halogen, -
NH2, -NO2, -CN, -OH, -CO2H, -CONH2, -SO3H, -SO2NH2, -SO2CH3, -OCH3, and -CF3.
[0010] In one aspect, described herein is a compound, or pharmaceutically acceptable salt, solvate, or prodrug thereof, having the structure of Formula (Ia):
Figure imgf000003_0001
wherein:
X is -C(R3)- or -N-;
R1 is unsubstituted Ci-C6 alkyl, unsubstituted or substituted aryl, or unsubstituted or substituted heteroaryl;
R2, R3, R5, R6, R7, R8, R9, R10 and R11 are each independently selected from the group consisting of H, halogen, -
NH2, -NO2, -CN, -OH, -CO2H, -CONH2, -SO3H, -SO2NH2, -SO2CH3, -OCH3, -CF3, -CH3, and -LA-LB-R32; LA is a covalent bond or an alkyl group; LB is a covalent bond, -0-, -NR32-, -NH-, -C(=0)-, -C(=0)0-, -0C(=0)-, -C(=O)NR32-, -C(=O)NH-, -NR32C(=O)-,
-NHC(=O)-, -SO2-, -SO2NR32-, - NR32SO2-, -SO2NH-, or -NHSO2-;
R32 is H, substituted or unsubstituted alkyl, haloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted non-aromatic heterocycle, or substituted or unsubstituted cycloalkyl; wherein any R32 group, when substituted, is substituted with one or more groups selected from halogen, -NH2, -NO2, -CN, -OH, -CO2H, -CONH2, -SO3H, -SO2NH2, -SO2CH3, -OCH3, and -CF3; Z is -NRaC(=0)-, -C(=0)NRa-, -NR3SO2-, -SO2NR3-, -NR3-, -CH2NR3-, - NR3CH2-, -CH2-, -CH2CH2-, -CH=CH-,
-CH2C(=O)NR3-, -NR3C(=O)CH2-, -SO2-, or -SO-; R3 is H or alkyl.
[0011] In one aspect, X is N. In another aspect, X is -C(R3)-. [0012] In some embodiments, Z is -NR3-.
[0013] In some embodiments, R1 is unsubstituted Ci-C6 alkyl or unsubstituted or substituted phenyl. In some other embodiments, R1 is unsubstituted Ci-C6 alkyl.
[0014] In some embodiments, each R3 is independently selected from the group consisting of H, halogen, -NH2, -NO2, -CN, -OH, -CO2H, -CONH2, -SO3H, -SO2NH2, -SO2CH3, -OCH3, -CF3, and -CH3. [0015] In some embodiments, R2, R5 and R6 are each H.
[0016] In other embodiments, R7, R8, R9, R10 and R11 are each independently selected from the group consisting of H, halogen, -NH2, -NO2, -CN, -OH, -CO2H, -CONH2, -SO3H, -SO2NH2, -SO2CH3, -OCH3, -CF3, -CH3, and -LA-LB-R32; LA is a covalent bond or an alkyl group; LB is a covalent bond, -O-, -NR32-, -NH-, -C(=0)O, -C(=O)NR32-, -C(=O)NH-, - SO2NR32-, or -SO2NH-; R32 is H, substituted or unsubstituted alkyl, haloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted non-aromatic heterocycle, or substituted or unsubstituted cycloalkyl; wherein any R32 group, when substituted, is substituted with one or more groups selected from halogen, -NH2, -NO2, -CN, -OH, -CO2H, -CONH2, -SO3H, -SO2NH2, -SO2CH3, -OCH3, and -CF3. [0017] In one aspect, provided herein is a compound, or pharmaceutically acceptable salt, solvate, or prodrug thereof, having the structure of Formula (Ib):
Figure imgf000004_0001
Formula (Ib), wherein:
R1, R2, R3, R5, R6, R7, R8, R9, R10 and R11 are each independently selected from the group consisting of H, halogen,
-NH2, -NO2, -CN, -OH, -CO2H, -CONH2, -SO3H, -SO2NH2, -SO2CH3, -OCH3, -CF3, -CH3, and -LA-LB-R32; LA is a covalent bond or an alkyl group; LB is a covalent bond, -0-, -NR32-, -NH-, -C(=0)-, -C(=0)0-, -0C(=0)-, -C(=O)NR32-, -C(=0)NH-, -NR32C(=O)-,
-NHC(=0)-, -SO2-, -SO2NR32-, - NR32SO2-, -SO2NH-, or -NHSO2-;
R32 is H, substituted or unsubstituted alkyl, haloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted non-aromatic heterocycle, or substituted or unsubstituted cycloalkyl; wherein any R32 group, when substituted, is substituted with one or more groups selected from halogen, -NH2, -NO2, -CN, -OH, -CO2H, -CONH2, -SO3H, -SO2NH2, -SO2CH3, -OCH3, and -CF3; Z is -NRaC(=0)-, -C(=0)NRa-, -NR3SO2-, -SO2NR3-, -NR3-, -CH2NR3-, - NR3CH2-, -CH2-, -CH2CH2-, -CH=CH-,
-CH2C(=0)NR3-, -NR3C(=0)CH2-, -SO2-, or -SO-; R3 is H or alkyl.
[0018] In some embodiments, Z is -NR3-. [0019] In some embodiments, R1 is unsubstituted Ci-C6 alkyl.
[0020] In some embodiments, each R3 is independently selected from the group consisting of H, halogen, -NH2, -NO2, -CN, -OH, -CO2H, -CONH2, -SO3H, -SO2NH2, -SO2CH3, -OCH3, -CF3, and -CH3. [0021] In other embodiments, R2, R5 and R6 are each H.
[0022] In some embodiments, R7, R8, R9, R10 and R11 are each independently selected from the group consisting of H, halogen, -NH2, -NO2, -CN, -OH, -CO2H, -CONH2, -SO3H, -SO2NH2, -SO2CH3, -OCH3, -CF3 -CH3, and -LA-LB-R32, LA is a covalent bond or an alkyl group; LB is a covalent bond, -0-, -NR32-, -NH-, -C(=0)0-, -C(=O)NR32-, -C(=0)NH-, - SO2NR32-, or -SO2NH-; R32 is H, substituted or unsubstituted alkyl, haloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted non-aromatic heterocycle, or substituted or unsubstituted cycloalkyl; wherein any R32 group, when substituted, is substituted with one or more groups selected from halogen, -NH2, -NO2, -CN, -OH, -CO2H, -CONH2, -SO3H, -SO2NH2, -SO2CH3, -OCH3, and -CF3. [0023] In one aspect, provided herein is a compound, or pharmaceutically acceptable salt, solvate, or prodrug thereof, having the structure of Formula (Ic):
Figure imgf000005_0001
wherein:
R1, R2, R3, R5, R6, R7, R8, R9, R10 and R11 are each independently selected from the group consisting of H, halogen, -NH2, -NO2, -CN, -OH, -CO2H, -CONH2, -SO3H, -SO2NH2, -SO2CH3, -OCH3, -CF3, -CH3, and -LA-LB-R32;
LA is a covalent bond or an alkyl group;
LB is a covalent bond, -0-, -NR32-, -NH-, -C(=0)-, -C(=0)0-, -0C(=0)-, -C(=O)NR32-, -C(=0)NH-, -NR32C(=O)-, -NHC(=0)-, -SO2-, -SO2NR32-, - NR32SO2-, -SO2NH-, or -NHSO2-;
R32 is H, substituted or unsubstituted alkyl, haloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted non-aromatic heterocycle, or substituted or unsubstituted cycloalkyl; wherein any R32 group, when substituted, is substituted with one or more groups selected from halogen, -NH2, -NO2, -CN, -OH, -CO2H, -CONH2, -SO3H, -SO2NH2, -SO2CH3, -OCH3, and -CF3;
Z is -NRaC(=0)-, -C(=0)NRa-, -NR3SO2-, -SO2NR3-, -NR3-, -CH2NR3-, - NR3CH2-, -CH2-, -CH2CH2-, -CH=CH-, -CH2C(=0)NR3-, -NR3CC=O)CH2-, -SO2-, or -SO-;
R3 is H or alkyl.
[0024] In some embodiments, Z is -NR3-. [0025] In some embodiments, R1 is unsubstituted Ci-C6 alkyl.
[0026] In some embodiments, each R3 is independently selected from the group consisting of H, halogen, -NH2, -NO2, -CN, -OH, -CO2H, -CONH2, -SO3H, -SO2NH2, -SO2CH3, -OCH3, -CF3, and -CH3. [0027] In some embodiments, R2, R5 and R6 are each H.
[0028] In some other embodiments, R7, R8, R9, R10 and R11 are each independently selected from the group consisting ofH, halogen, -NH2, -NO2, -CN, -OH, -CO2H, -CONH2, -SO3H, -SO2NH2, -SO2CH3, -OCH3, -CF3, -CH3, and -LA-LB- R32, LA is a covalent bond or an alkyl group; LB is a covalent bond, -0-, -NR32-, -NH-, -C(=0)0-, -C(=O)NR32-, - C(=0)NH-, -SO2NR32-, or -SO2NH-; R32 is H, substituted or unsubstituted alkyl, haloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted non-aromatic heterocycle, or substituted or unsubstituted cycloalkyl; wherein any R32 group, when substituted, is substituted with one or more groups selected from halogen, -NH2, -NO2, -CN, -OH, -CO2H, -CONH2, -SO3H, -SO2NH2, -SO2CH3, -OCH3, and -CF3. [0029] In some embodiments, LA is a covalent bond.
[0030] In some embodiments, LB is a covalent bond, -0-, -NH-, -C(=0)0-, -C(=0)NH-, or -SO2NH-. [0031] In some embodiments, R32 is H, substituted or unsubstituted alkyl, or haloalkyl; wherein any R32 group, when substituted, is substituted with one or more groups selected from halogen, -NH2, -NO2, -CN, -OH, -CO2H, -CONH2, - SO3H, -SO2NH2, -SO2CH3, -OCH3, and -CF3. In some embodiments, R32 is H, substituted or unsubstituted alkyl, or haloalkyl.
[0032] In one aspect, R1, R3, R7, R8, R9, R10, R11 are as defined in Table 1, Table 2, Table 3 and Table 4. In one aspect, R2, R5, and R6 are each H and R1, R3, R7, R8, R9, R10, R11 are as defined in Table 1, Table 2, Table 3 and Table 4. [0033] Any combination of the groups described above for the various variables is contemplated herein. [0034] In one aspect, the compound of Formula (I) inhibits at least one kinase selected from among a Cyclin- dependent kinase (CDK), a Glycogen synthase kinase (GSK), an aurora kinase, a polo-like kinase (PLK), BCR-ABL,
IkappaB kinase complex (IKK), Fms-like tyrosine kinase 3 (FLT3), Janus kinase (JAK), lymphocyte-specific protein tyrosine kinase (LCK), Platelet-derived growth factor receptor kinase (PDGFR), a Src kinase, and vascular endothelial growth factor (VEGF) receptor.
[0035] In one aspect, provided is a pharmaceutical composition comprising a compound of Formula (I) and a pharmaceutically excipient.
[0036] In one aspect, the pharmaceutical composition is in the form of an aqueous dispersion, liquid, gel, syrup, elixir, slurry, suspension, aerosol, controlled release formulation, fast melt formulation, effervescent formulation, lyophilized formulation, tablet, powder, pill, dragee, capsule, delayed release formulation, extended release formulation, pulsatile release formulation, multiparticulate formulation, or immediate release formulation.
[0037] In one aspect, provided is a method for treating or preventing a kinase mediated or kinase dependent disease, disorder or condition in a patient, comprising administrating to the patient an effective amount of a compound of
Formula (I). In one aspect, the kinase is selected from among a Cyclin-dependent kinase (CDK), a Glycogen synthase kinase (GSK), an aurora kinase, a polo-like kinase (PLK), BCR-ABL, IkappaB kinase complex (IKK), Fms-like tyrosine kinase 3 (FLT3), Janus kinase (JAK), lymphocyte-specific protein tyrosine kinase (LCK), Platelet-derived growth factor receptor kinase (PDGFR), a Src kinase, and vascular endothelial growth factor (VEGF) receptor. In some embodiments, the kinase is a Cyclin-dependent kinase (CDK). In some embodiments, the kinase mediated or kinase dependent disease, disorder or condition is a proliferative disorder. In some embodiments, the proliferative disorder is cancer or leukemia. In some embodiments, the proliferative disorder is cancer or leukemia selected from cervical cancer, osteosarcoma, esophageal cancer, liver cancer, myelogenous leukaemia, melanoma, acute lymphoblastic leukemia, lung cancer, breast cancer, or renal cancer.
[0038] In one aspect, provided is a pharmaceutical composition comprising an amount of a compound of Formula (I) effective for inhibiting cyclin-dependent kinase 2 activity in a cell, and a pharmaceutically acceptable excipient.
[0039] In one aspect, provided is a method for inhibiting undesired cyclin-dependent kinase 2 activity in a cell comprising contacting the cell with a compound of Formula (I).
[0040] In one aspect, provided is a method for treating cancer in a patient, in which the cancer is characterized by undesired cyclin-dependent kinase 2 activity, comprising administering to the patient an amount of a compound of
Formula (I) effective for inhibiting the undesired cyclin-dependent kinase 2 activity. In some embodiments, the cancer is selected from breast cancer and cervical cancer.
[0041] In one aspect, described herein is a pharmaceutical composition comprising a pharmaceutically acceptable diluent, excipient or binder, and a compound of Formula (I), or pharmaceutically acceptable salt, pharmaceutically acceptable prodrug, or pharmaceutically acceptable solvate thereof.
[0042] In one aspect, described herein is a pharmaceutical composition, which includes an effective amount of a compound provided herein, and a pharmaceutically acceptable excipient. In a further aspect, provided are compositions further including a second pharmaceutically active ingredient in addition to the compound of Formula (I).
[0043] In certain embodiments, provided herein is a pharmaceutical composition comprising: i) a physiologically acceptable carrier, diluent, and/or excipient; and ii) one or more compounds described herein.
[0044] In any of the aforementioned aspects are further embodiments that include single administrations of the effective amount of the compound of Formula (I), including further embodiments in which: (i) the compound of
Formula (I) is administered once; (ii) the compound of Formula (I) is administered to the mammal multiple times over the span of one day; (iii) the compound of Formula (I) is administered continually; or (iv) the compound of Formula (I) is administered continuously.
[0045] In any of the aforementioned aspects are further embodiments that include multiple administrations of the effective amount of the compound of Formula (I), including further embodiments in which (i) the compound of Formula (I) is administered in a single dose; (ii) the time between multiple administrations is every 6 hours; (iii) the compound of Formula (I) is administered to the mammal every 8 hours. In further or alternative embodiments, the method comprises a drug holiday, wherein the administration of the compound of Formula (I) is temporarily suspended or the dose of the compound of Formula (I) being administered is temporarily reduced; at the end of the drug holiday, dosing of the compound of Formula (I) is resumed. The length of the drug holiday can vary from 2 days to 1 year. [0046] In one aspect, compounds of Formula (I) described herein are administered to a human. In some embodiments, compounds of Formula (I) described herein are orally administered.
[0047] A further aspect relates to the use of a compound of Formula (I) in the manufacture of a medicament for treating a condition mediated by an enzyme selected from one or more CDK, aurora kinase, GSK, PLK, BCR-ABL, FLT, IKK, JAK, PDGF or VEGF and Src family enzymes. In another aspect is the use of a compound of Formula (I) in the manufacture of a medicament for treating a condition mediated by at least one enzyme selected from CDKl, CDK2, CDK4/6, CDK5, CDK7, CDK8, CDK9, CDKl 1, GSK-3, aurora kinase, PLK and a tyrosine kinase. [0048] In another aspect, there is provided a method for treating a condition mediated by one or more enzymes selected from CDK, aurora kinase, GSK, PLK, BCR-ABL, FLT, IKK, JAK, PDGF or VEGF and Src family enzymes, particularly from one or more CDKl, CDK2, CDK4/6, CDK5, CDK7, CDK8, CDK9, CDKl 1, GSK-3, aurora kinase, PLK or tyrosine kinase enzyme, in a human or animal subject, the method comprising administering to a human or animal in need thereof a therapeutically effective amount of a compound of Formula (I).
[0049] In a further aspect, there is provided the use of a compound of Formula (I) in a method for treating a condition mediated by an enzyme selected from one or more CDK, aurora kinase, GSK, PLK, BCR-ABL, FLT, IKK, JAK, PDGF or VEGF and Src family enzymes, particularly from one or more CDKl, CDK2, CDK4/6, CDK5, CDK7, CDK8, CDK9, CDKl 1, GSK-3, aurora kinase, PLK or tyrosine kinase.
[0050] A further aspect relates to the use of a compound of Formula (I) or a pharmaceutically acceptable salt or solvate or physiologically hydrolysable, solubilising or immobilising derivative thereof, in an assay for identifying candidate compounds capable of treating a condition mediated by an enzyme selected from one or more CDK, aurora kinase, GSK, PLK, BCR-ABL, FLT, IKK, JAK, PDGF or VEGF and Src family enzymes, particularly from one or more CDKl, CDK2, CDK4/6, CDK5, CDK7, CDK8, CDK9, CDKIl, GSK-3, aurora kinase, PLK or tyrosine kinase. [0051] Other objects, features and advantages of the compounds, methods and compositions described herein will become apparent from the following detailed description. It should be understood, however, that the detailed description and the specific examples, while indicating specific embodiments, are given by way of illustration only, since various changes and modifications within the spirit and scope of the instant disclosure will become apparent to those skilled in the art from this detailed description.
DETAILED DESCRIPTION OF THE INVENTION
[0052] The protein kinase family is one of the largest in the human genome, comprising 500 genes. The majority of kinases contain a 250-300 amino acid residue catalytic domain with a conserved core structure. This domain comprises a binding pocket for ATP, whose terminal phosphate group transfers covalently to its macromolecular substrates. The protein kinases may be categorized by the substrates they phosphorylate, e.g. protein-serine/threonine, protein-tyrosine. [0053] Protein kinases mediate intracellular signalling by affecting a phosphoryl transfer from a nucleoside triphosphate to a protein acceptor that is involved in a signalling pathway. These phosphorylation events are triggered in response to a variety of extracellular and other stimuli and act as molecular on/off switches that can modulate or regulate the target protein biological function. An extracellular stimulus may affect one or more cellular responses related to cell growth, migration, differentiation, secretion of hormones, activation of transcription factors, muscle contraction, glucose metabolism, control of protein synthesis, and regulation of the cell cycle.
[0054] Many diseases are associated with abnormal cellular responses triggered by protein kinase-mediated events.
These diseases include, but are not limited to allergies, asthma, Alzheimer's disease, autoimmune diseases, bone diseases, cancer, cardiovascular diseases, inflammatory diseases, hormone-related diseases, metabolic diseases, neurological and neurodegenerative diseases. There is a need for protein kinase inhibitors that are effective as therapeutic agents.
[0055] Cyclin-dependent kinases (CDKs) are serine/threonine protein kinases that associate with various cyclin subunits, playing pivotal roles in the regulation of cell cycle progression and transcriptional cycle. Ten distinct CDKs
(CDK 1-9 and 11) are involved in a variety of important regulatory pathways in eukaryotic cells, including cell-cycle control, apoptosis, neuronal physiology, differentiation and transcription.
[0056] In one aspect, CDKs are classified into two major groups, reflecting their functions. The cell cycle regulator
CDKs composed primarily of CDKl, CDK2, CDK3, CDK4 and CDK6 function with their cyclin partners including cyclin A, B, Dl, D2, D3, E, and F to regulate promotion of the cell cycle. The transcription regulator CDKs, which include CDK7, CDK8, CDK9 and CDKl 1 work together with cyclin C, H, K, Ll, L2, Tl and T2, tend to play roles in transcriptional regulation.
[0057] The CDKs are implicated in cell proliferation disorders, particularly in cancer. Cell proliferation is a result of the direct or indirect deregulation of the cell division cycle and the CDKs play a critical role in the regulation of the various phases of this cycle. Therefore, inhibitors of CDKs and their associated cyclins are useful targets for cancer therapy.
[0058] CDKs also play a role in apoptosis and T-cell development, which is predominantly due to the CDK functions in regulation of transcription.
[0059] Furthermore, numerous viruses require CDKs, particular CDK2, CDK7, and CDK9, for their replication process. CDK inhibitors have been reported to restrain viral replication including human immunodeficiency virus, human cytomegalovirus, herpes virus, and varicella-zoster virus.
[0060] Inhibition of CDK is also useful for the treatment of neurodegenerative disorders such as Alzheimer's disease.
The appearance of Paired Helical Filaments, associated with Alzheimer's disease, is caused by the hyperphosphorylation of Tau protein by CDK5/p25.
[0061] Inhibition of one or more other serine/threonine kinases including the Aurora kinases, Glycogen synthesis kinases (GSKs), polo-like kinases (PLKs) and tyrosine kinases including Ableson tyrosine kinase (BCR-ABL), FMS- related tyrosine kinases (FLT), IkB kinases (IKK), Janus kinases (JAK), platelet-derived growth factor (PDGF) receptor tyrosine kinases, vascular endothelial growth factor (VEGF) receptor tyrosine kinases, and Src family are also useful for the treatment of numerous diseases, conditions or disorders mediated by these kinases.
[0062] GSK3 is known to phosphorylate many substrates and is thus involved in the regulation of multiple biochemical pathways. For example, GSK3 is over-expressed in muscle cells of type II diabetics and that an inverse correlation exists between skeletal muscle GSK3 activity and insulin action. GSK3 inhibition is therefore of therapeutic significance in the treatment of diabetes, particularly type II, and diabetic neuropathy.
[0063] Furthermore, GSK is highly expressed in the central and peripheral nervous systems. GSK3 inhibition is therefore of therapeutic significance in the treatment of CNS disorders such as Parkinsons and Alzheimers diseases. [0064] Aurora kinases and PLK are also important therapeutic targets for treatment of proliferative disorders. Based on their known functions inhibition of Aurora kinases and PLKs activity should disrupt mitosis leading to cell cycle arrest and therefore slowing tumor growth and induce apoptosis.
[0065] Described herein are compounds that inhibit one or more protein kinases. In one aspect, the compounds described herein are heteroaryl-pyrimidine derivatives. More specifically, described herein are substituted 4-(indol-3- yl)-7V-phenyrpyrimidin-2 -amine compounds, substituted 4-(pyrrolo[2,3-i]pyridin-3-yl)-7V-phenylpyrimidin-2 -amine compounds, 4-(pyrrolo[2,3-c]pyridin-3-yl)-N-phenylpyrimidin-2-amine compounds, substituted 4-(pyrrolo[3,2-
£]pyridin-3-yl)-7V-phenyrpyrimidin-2 -amine compounds, and substituted 4-(pyrrolo[3,2-c]pyridin-3-yl)-7V- phenylpyrimidin-2 -amine compounds. In one aspect, the compounds described herein have broad therapeutic use as protein kinase inhibitors.
Compounds
[0066] In one aspect, described herein is a compound of Formula (I), or pharmaceutically acceptable salt, solvate, or prodrug thereof:
Figure imgf000009_0001
I wherein,
Het-Ar is an unsubstituted or substituted indolyl ring, or an unsubstituted or substituted azaindolyl ring;
Z is -NRaC(=O)-, -C(=O)NRa-, -NR3SO2-, -SO2NR3-, -NR3-, -CH2NR3-, - NR3CH2-, -CH2-, -CH2CH2-, -CH=CH-, -
CH2C(=O)NR3-, -NR3CC=O)CH2-, -SO2-, or -SO-;
R3 is H or alkyl;
R5, R6, R7, R8, R9, R10, and R11, are each independently H, alkyl, alkyl-R19, aryl, aryl-R19, aralkyl, aralkyl-R19, halogen,
-NO2, -CN, -OH, -O-alkyl, -COR19, -COOR19, -O-aryl, -0-R19, -NH2, -NH-alkyl, -NH-aryl, -N-(alkyl)2, -N-(aryl)2, -N-
(alkyl)(aryl), -NH-R19, -N-(R19)(R20), -N-(alkyl)(R19), -N-CaTyI)(R19), -COOH, -CONH2, -CONH-alkyl, -CONH-aryl, -
CON-(alkyl)(R19), -CONCaTyI)(R19), -CONH-R19, -CON-(R19)(R20), -SO3H, -SO2-alkyl, -SO2-alkyl-R19, -SO2-aryl, -
SO2-aryl-R19, -SO2NH2, -SO2NH-R19, -SO2N-(R19)(R20), -CF3, -CO-alkyl, -CO-alkyl-R19, -CO-aryl, -CO-aryl-R19 or -
R21, wherein alkyl, aryl, aralkyl groups may be further substituted with one or more groups selected from halogen, -
NO2, -OH, -0-methyl, -NH2, -COOH, -CONH2 and -CF3; where R19, R20, and R21 are physiologically hydrolysable group, solubilising group or immobilisable group.
[0067] In one aspect, R19 and R20 are each independently solubilising groups selected from:
(i) a mono-, di- or polyhydroxylated alicyclic group; a di- or polyhydroxylated aliphatic or aromatic group; a carbohydrate derivative; an O- and/or S-containing heterocyclic group optionally substituted by one or more hydroxyl groups; an aliphatic or aromatic group containing a carboxamide, sulfoxide, sulfone, or sulfonamide function; or a halogenated alkylcarbonyl group; (ii) COOH, SO3H, OSO3H, PO3H2, or OPO3H2;
(iii) Y, where Y is selected from an alicyclic, aromatic, or heterocyclic group comprising one or more of the functions =N-, -0-, -NH2, -NH-, a quarternary amine salt, guanidine, and amidine, where Y is optionally substituted by one or more substituents selected from SO2-alkyl, alkyl optionally substituted by one or more OH groups, CO-alkyl, aralkyl, COO-alkyl, and an ether group optionally substituted by one or more OH groups, and
(iv) a natural or unnatural amino acid, a peptide or a peptide derivative, each R21 is a solubilising group as defined for R19 and R20 in (i) or (iv) above, or is selected from
(v) OSO3H, PO3H2, or OPO3H2,
(vi) Y as defined above, but excluding guanidine and quarternary amine salts,
(vii) NHCO(CH2)m[NHCO(CH2)m ]p[NHCO(CH2)m ]qY or NHCO(CH2)tNH(CH2)t Y where p and q are each O or 1, and m, m',m", t and t' are each independently an integer from 1 to 10, and
(viii) (CH2)nNR14COR12, (CH2)n NR15SO2R13, or SO2R16, where R12, R13 and R16 are each alkyl groups optionally comprising one or more heteroatoms, and which are optionally substituted by one or more substituents selected from
OH, NH2, halogen and NO2, R14 and R15 are each independently H or alkyl, and n and n' are each independently O, 1, 2, or 3,
(ix) an ether or polyether optionally substituted by one or more hydroxyl groups or one or more Y groups,
(x) (CH2XNH2, where r is O, 1, 2, or 3 ,
(xi) (CH2)r OH, where r' is O, 1, 2, or 3,
(xii) (CH2)n NR17COR18 where R17 is H or alkyl, n" is O, 1, 2 or 3 and R18 is an aryl group optionally substituted by one or more substituents selected from halogen, NO2, OH, alkoxy, NH2, COOH, CONH2 and CF3,
(xiu) SO2NR22R23 where R22 and R23 are each independently H, alkyl or aryl, with the proviso that at least one of R22 and R23 is other than H, or R22 and R23 are linked to form a cyclic group optionally containing one or more heteroatoms selected from N, O and S, and wherein said alkyl, aryl or cyclic group is optionally substituted by one or more substituents selected from halogen, NO2, OH, alkoxy, NH2, COOH, CONH2 and CF3
[0068] In one aspect, at least one of R7, R8, R9, R10, and R11 is selected from (CH2)n NR17COR18 ,
(CH2)n Q=O)NR17R18 and SO2NR22R23
[0069] In one aspect, Het-Ar is a substituted or unsubstituted group selected from indol-3-yl, pyrrolo[2,3-£]pyridin-3- yl, pyrrolo[2,3-c]pyndin-3-yl, pyrrolo[3,2-£]pyridin-3-yl, and pyrrolo[3,2-c]pyridin-3-yl
[0070] In one aspect, Het-Ar is a substituted or unsubstituted group selected from l-(alkyl)-indol-3-yl, l-(alkyl)- pyrrolo[2,3-£]pyridin-3-yl, l-(alkyl)-pyrrolo[2,3-c]pyndin-3-yl, l-(alkyl)-pyrrolo[3,2-£]pyridin-3-yl, and l-(alkyl)- pyrrolo[3 ,2-c]pyndin-3 -yl
[0071] In one aspect, the compound of Formula (I) has the structure
Figure imgf000010_0001
Formula (I) wherein each X is -C(R3)- or -N-, provided that at least two X are -C(R3)-,
Z is -NRaC(=O)-, -C(=O)NRa-, -NR3SO2-, -SO2NR3-, -NR3-, -CH2NR3-, - NR3CH2-, -CH2-, -CH2CH2-, -CH=CH-, - CH2C(=O)NR3-, -NR3C(=O)CH2-, -SO2-, or -SO-, R3 is H or alkyl, and wherein R1, R2, R3, R5, R6, R7, R8, R9, R10, and R11, are each independently H, alkyl, alkyl-R19, aryl, aryl-R19, aralkyl, aralkyl-R19, halogen, -NO2, -CN, -OH, -O-alkyl, -COR19, -COOR19, -O-aryl, -0-R19, -NH2, -NH-alkyl, -NH- aryl, -N-(alkyl)2, -N-(aryl)2, -N-(alkyl)(aryl), -NH-R19, -N-(R19)(R20), -N-(alkyl)(R19), -N-tarylXR19), -COOH, -CONH2, -CONH-alkyl, -CONH-aryl, -CON-(alkyl)(R19), -CONCaTyI)(R19), -CONH-R19, -CON-(R19)(R20), -SO3H, -SO2-alkyl, -
SO2-alkyl-R19, -SO2-aryl, -SO2-aryl-R19, -SO2NH2, -SO2NH-R19, -SO2N-(R19)(R20), -CF3, -CO-alkyl, -CO-alkyl-R19, -
CO-aryl, -CO-aryl-R19 or -R21, wherein alkyl, aryl, aralkyl groups may be further substituted with one or more groups selected from halogen, -NO2, -OH, -0-methyl, -NH2, -COOH, -CONH2 and -CF3; where R19, R20, and R21 are physiologically hydrolysable group, solubilising group or immobilisable group.
[0072] In one aspect, R1, R2, R3, R5, R6, R7, R8, R9, R10, and R11, are each independently H, alkyl, aryl, aralkyl, halogen, -NO2, -CN, -OH, -O-alkyl, -O-aryl, -NH2, -NH-alkyl, -NH-aryl, -N-(alkyl)2, -N-(aryl)2, -N-(alkyl)(aryl), -
COOH, -CONH2, -CONH-alkyl, -CONH-aryl, -SO3H, -SO2-alkyl, -SO2-aryl, -SO2NH2, -CF3, -CO-alkyl, -CO-aryl, wherein alkyl, aryl, aralkyl groups may be further substituted with one or more groups selected from halogen, -NO2, -
OH, -0-methyl, -NH2, -COOH, -CONH2 and -CF3.
[0073] In one aspect, R5 and R6 are each H.
[0074] In one aspect, R2 is H or alkyl. In another aspect, R2 is H.
[0075] In one aspect, at least one R3 is H. In another aspect, at least two R3 are H.
[0076] In one aspect, at least one of R7, R8, R9, R10, and R11 is H. In another aspect, at least two of R7, R8, R9, R10, and R11 are H.
[0077] In one aspect, the compound of Formula (I) has the structure:
Figure imgf000011_0001
Formula (I) wherein each X is -C(R3)- or -N-, provided that at least two X are -C(R3)-;
Z is -NRaC(=O)-, -C(=O)NRa-, -NR3SO2-, -SO2NR3-, -NR3-, -CH2NR3-, - NR3CH2-, -CH2-, -CH2CH2-, -CH=CH-, - CH2C(=O)NR3-, -NR3C(=O)CH2-, -SO2-, or -SO-;
R3 is H or alkyl; and wherein R1, R2, R3, R5, R6, R7, R8, R9, R10, and R11, are as described herein. [0078] In one aspect, the compound of Formula (I) has the structure of Formula (Ia):
Figure imgf000011_0002
Formula (Ia) wherein X is -C(R3)- or -N-;
Z is -NR3C(=0)-, -C(=0)NR3-, -NR3SO2-, -SO2NR3-, -NR3-, -CH2NR3-, - NR3CH2-, -CH2-, -CH2CH2-, -CH=CH-, - CH2C(=O)NR3-, -NR3C(=O)CH2-, -SO2-, or -SO-;
R3 is H or alkyl; and wherein R1, R2, R3, R5, R6, R7, R8, R9, R10, and R11, are as described herein. [0079] In another aspect, the compound of Formula (I) has the structure of Formula (Ib):
Figure imgf000012_0001
Formula (Ib) wherein
Z is -NRaC(=O)-, -C(=O)NRa-, -NR3SO2-, -SO2NR3-, -NR3-, -CH2NR3-, - NR3CH2-, -CH2-, -CH2CH2-, -CH=CH-, - CH2C(=O)NR3-, -NR3CC=O)CH2-, -SO2-, or -SO-;
R3 is H or alkyl; and wherein R1, R2, R3, R5, R6, R7, R8, R9, R10, and R11, are as described herein. [0080] In one aspect, the compound of Formula (I) has the structure of Formula (Ic)
Figure imgf000012_0002
Formula (Ic) wherein
Z is -NR3C(=0)-, -C(=0)NR3-, -NR3SO2-, -SO2NR3-, -NR3-, -CH2NR3-, - NR3CH2-, -CH2-, -CH2CH2-, -CH=CH-, CH2C(=O)NR3-, -NR3C(=O)CH2-, -SO2-, or -SO-;
R3 is H or alkyl; and wherein R1, R2, R3, R5, R6, R7, R8, R9, R10, and R11, are as described herein. [0081] In one aspect, the compound of Formula (I) has the structure of Formula (Id)
Figure imgf000012_0003
wherein
Z is -NR3C(=0)-, -C(=0)NR3-, -NR3SO2-, -SO2NR3-, -NR3-, -CH2NR3-, - NR3CH2-, -CH2-, -CH2CH2-, -CH=CH-, - CH2C(=O)NR3-, -NR3C(=O)CH2-, -SO2-, or -SO-;
R3 is H or alkyl; and wherein R1, R2, R3, R5, R6, R7, R8, R9, R10, and R11, are as described herein. [0082] In one aspect, Z is -NR3C(=0)-, -C(=0)NR3-, -NR3SO2-, -SO2NR3-, -NR3-, -CH2NR3-, - NR3CH2-, -CH2-, - CH2CH2-, -CH=CH-, -CH2C(=O)NR3-, -NR3C(=O)CH2-, -SO2-, or -SO-; R3 is H or alkyl.
[0083] In one aspect, -Z- is -NH-, -NHC(=O)-, -NHSO2-, -NHCH2., -CH2-, -CH2CH2-, -CH=CH-, -CH2C(=O)NH-, - SO2-, or -SO-.
[0084] In one aspect, R3 is H.
[0085] In one aspect, Z is -NR3-. In one aspect, Z is -NH-.
[0086] In one aspect, R19 and R20 are each independently solubilising groups selected from a mono-, di- or polyhydroxylated alicyclic group; a di- or polyhydroxylated aliphatic or aromatic group; a carbohydrate derivative; an O- and/or S-containing heterocyclic group optionally substituted by one or more hydroxyl groups, an aliphatic or aromatic group containing a carboxamide, sulfoxide, sulfone, or sulfonamide function, or a halogenated alkylcarbonyl group, COOH, SO3H, OSO3H, PO3H2, or OPO3H2
[0087] In one aspect, at least one of R19 or R20 comprises an ionisable basic group selected from an alicyclic, aromatic, or heterocyclic group comprising one or more of the functions =N-, -0-, -NH2, -NH-, a quarternary amine salt, guanidine, amidine, optionally substituted by one or more substituents selected from SO2-alkyl, alkyl optionally substituted by one or more OH groups, CO-alkyl, aralkyl, COO-alkyl, and an ether group optionally substituted by one or more OH groups
[0088] In one aspect, at least one of R19 and R20 is a natural or unnatural amino acid residues and peptides, or their derivatives
[0089] In one aspect, R19 or R20 is selected from OSO3H, PO3H2, or OPO3H2,
Y, where Y is selected from an alicyclic, aromatic, or heterocyclic group comprising one or more of the functions =N-, -0-, -NH2, -NH-, a quarternary amine salt, guanidine, and amidine, where Y is optionally substituted by one or more substituents selected from SO2-alkyl, alkyl optionally substituted by one or more OH groups, CO-alkyl, aralkyl, COO- alkyl, and an ether group optionally substituted by one or more OH groups
[0090] In one aspect, each R21 is a solubilising group as defined for R19 and R20 hereinbefore defined, or is selected from OSO3H, PO3H2, or OPO3H2, Y as defined above, but excluding guanidine and quarternary amine salts, NHCO(CH2)m[NHCO(CH2)m ]p[NHCO(CH2)m ]qY or NHCO(CH2)tNH(CH2)t Y where p and q are each O or 1, and m, m',m", t and t' are each independently an integer from 1 to 10, and (CHz)nNR14COR12, (CH2)n NR15SO2R13, or SO2R16, where R12, R13 and R16 are each alkyl groups optionally comprising one or more heteroatoms, and which are optionally substituted by one or more substituents selected from OH, NH2, halogen and NO2, R14 and R15 are each independently H or alkyl, and n and n' are each independently O, 1, 2, or 3, an ether or polyether optionally substituted by one or more hydroxyl groups or one or more Y groups, (CH2)rNH2, where r is O, 1, 2, or 3, (CH2)r OH, where r' is O, 1, 2, or 3, (CH2)n NR17COR18 where R17 is H or alkyl, n" is O, 1, 2 or 3 and R18 is an aryl group optionally substituted by one or more substituents selected from halogen, NO2, OH, alkoxy, NH2, COOH, CONH2 and CF3, SO2NR22R23 where R22 and R23 are each independently H, alkyl or aryl, with the proviso that at least one of R22 and R23 is other than H, or R22 and R23 are linked to form a cyclic group optionally containing one or more heteroatoms selected from N, O and S, and wherein said alkyl, aryl or cyclic group is optionally substituted by one or more substituents selected from halogen, NO2, OH, alkoxy, NH2, COOH, CONH2 and CF3
[0091] In one aspect, R7, R8, R9, R10 and R11 are each independently H, alkyl, alkyl-R19, aryl, aryl-R19, aralkyl, aralkyl-R19, halogen, -NO2, -CN, -OH, -O-alkyl, -COR19, -COOR19, -O-aryl, -0-R19, -NH2, NH-alkyl, NH-aryl, N- (alkyl)2, N-(aryl)2, -N-(alkyl)(aryl), NH-R19, N-(R19)(R20), N-(alkyl)(R19), -N-tarylXR19), -COOH, -CONH2, -CONH- alkyl, -CONH-aryl, -CON-(alkyl)(R19), -CONCaTyI)(R19), -CONH-R19, -CON-(R19)(R20), -SO3H, -SO2-alkyl, -SO2- alkyl-R19, -SO2-aryl, -SO2-aryl-R19, -SO2NH2, -SO2NH-R19, -SO2N-(R19)(R20), -CF3, -CO-alkyl, -CO-alkyl-R19, -CO- aryl, -CO-aryl-R19 or R21, wherein alkyl, aryl, aralkyl groups may be further substituted with one or more groups selected from halogen, -NO2, -OH, -0-methyl, -NH2, -COOH, -CONH2 and -CF3 [0092] In another embodiment, at least one of R1, R2, R3, R5, R6, R7, R8, R9, R10 and R11 is R21 [0093] In one aspect, R1 is selected from H, alkyl, aryl, heteroaryl, cycloalkyl, alkyl-aryl, alkyl-heteroaryl and alkyl- cycloalkyl
[0094] In one aspect, R1 is selected from H, Ci-Cs-alkyl, aryl, heteroaryl, cycloalkyl, alkyl-aryl, alkyl-heteroaryl and alkyl-cycloalkyl [0095] In one aspect, R2 is selected from H, alkyl, C1-C5 alkyl, CONH2, CONH-alkyl, CN, CF3, O-alkyl, halogen, NH2, NH-alkyl, NHCO-alkyl, alkyl-heteroaryl, and alkyl-cycloalkyl.
[0096] In one aspect, R2 is selected from H, C1-C5 alkyl, CONH2, CONH-alkyl, CN, CF3, O-alkyl, halogen, NH2, NH-alkyl, NHCO-alkyl, alkyl-heteroaryl, and alkyl-cycloalkyl.
[0097] In one aspect, each R3 is independently selected from H, alkyl, CONH2, CONH-alkyl, CN, OH, CF3, O-alkyl, halogen, NH2, NH-alkyl, NHCO-alkyl, alkyl-heteroaryl, alkyl-cycloalkyl. [0098] In one embodiment, R7 is H, O-alkyl, CF3, alkyl or halogen. [0099] In one embodiment, R7 is selected from H, OCH3, CF3, alkyl or halogen.
[00100] In one embodiment, R8 and R10 are independently selected from H, CONH2, CONH-alkyl, CN, OH, CF3, sulphonyl, carbonyl, amide or sulphonamide, or thioether link to an unsubstituted or substituted 6 membered cyclic or heterocyclic, or aromatic or heteroaromatic ring, wherein substituents are as hereinbefore defined. [00101] In one aspect, R8 and R10 are independently selected from SO2-NR20, SO2-cycloheteroalkyl, SO2-cycloalkyl, SO2-heteroaryl, SO-cycloheteroalkyl, SO-cycloalkyl, SO-heteroaryl, C(=O)-cycloheteroalkyl, C(=O)-cycloalkyl, C(=O)-heteroaryl, N-(alkyl)(cycloalkyl), N-(alkyl)(cycloheteroalkyl), or N-(alkyl)(heteroaryl) more preferably wherein the cycloheteroalkyl is heteroatom linked and may be unsubstituted or substituted comprising one, two or three heteroatoms selected from N, O, S. More preferably a cycloheteroalkyl is a 7V-alkyl-morpholino, 7V-alkyl-piperazine or 7V-alkyl-piperadine. Most preferably R5 and R7 are independently selected from TV-linked 7V-(alkyl)(cycloheteroalkyl), SO2-cycloheteroalkyl and CO-cycloheteroalkyl most preferably such as N-(alkyl)(morpholino), N-(alkyl)(piperazine), N-(alkyl)(piperadine), SO2-piperazines, SO2-morpholines, -C(=O)-piperazines, -C(=O)-morphorines, -C(=O)- piperadine or the like.
[00102] In one aspect, R9 is selected from H, alkyl, for example C1-5 alkyl, CONH2, CONH-alkyl, CN, OH, CF3, O- alkyl, halogen, NH2, NH-alkyl, NHCO-alkyl, sulphonyl, carbonyl, amide or sulphonamide, or thioether link to an unsubstituted or substituted 6 membered cyclic or heterocyclic, or aromatic or heteroaromatic ring, wherein substituents are as hereinbefore defined.
[00103] In one aspect, R11 is selected from H, halogen, O-alkyl. In one aspect, R11 is selected from H, halogen and O- C1-C5 alkyl.
[00104] In one aspect, up to six of R1, R2, R3, R5, R6, R7, R8, R9, R10, and R11, for example one, two, three or four thereof, shall correspond to or contain one or more of the group R19 or R20. Preferably either or all of R8, R9 or R10 comprise or contain one or more of the group R20 or R21. Two or more groups R19 and/or R20 may be the same or different.
[00105] Preferably R1, R2, R3, R8 or R9 comprises or contains a solubilising moiety R19 or R20. [00106] In one aspect, described herein are compounds of Formula (Ia) wherein: X is CR3 or N; Z is NH; R1 and R2 are selected from H, alkyl, such as Q.s-alkyl or aryl, such as C6 aryl; halogen, or heteroaryl; R3 is one, two or three groups independently selected from H, amino, Ci-4 alkyl, CN, CF3, halogen, NO2 O-alkyl, NH2, NH-alkyl, N(alkyl)2, CO2alkyl, CO-alkyl, CONH2, CONH-alkyl or heteroaryl or is contained with R20 or R21; R7 is H, OMe, alkyl, or halogen, each R8, R9 or R10 is independently H, alkyl, OH, CF3, O-alkyl, halogen, 0-heteroaryl, alkyl-heteroaryl, alkyl- cycloalkyl which may be part unsaturated; NH2 SO2-NH2, SO2-NR20, SO2-cycloheteroalkyl, SO-cycloheteroalkyl, SO2- heteroaryl, SO-heteroaryl, CO-cycloheteroalkyl or CO-heteroarylalkyl, and the solubilising moiety corresponds to or is contained within R1, R2, R3, or R7; and wherein R11 is H and the solubilising moiety corresponds to or is contained within R1, R2, R3 or R7. [00107] In another embodiment, described herein are compounds of Formula (Ib) wherein: Z is NH; R1 and R2 are selected from H, alkyl, such as Ci_5-alkyl or aryl, such as C6 aryl; halogen, or heteroaryl;
R3 is one, two or three groups independently selected from H, amino, Ci-4 alkyl, CN, CF3, halogen, NO2 O-alkyl, NH2,
NH-alkyl, N(alkyl)2, C02alkyl, CO-alkyl, CONH2, CONH-alkyl or heteroaryl or is contained with R20 or R21;
R7 is H, OMe, alkyl, or halogen, each R8, R9 or R10 is independently alkyl, OH, CF3, O-alkyl, halogen, O-heteroaryl, alkyl-heteroaryl, alkyl-cycloalkyl which may be part unsaturated; NH2 SO2-NH2, SO2-NR20, SO2-cycloheteroalkyl, SO-cycloheteroalkyl, SO2-heteroaryl,
SO-heteroaryl, CO-cycloheteroalkyl or CO-heteroarylalkyl, and the solubilising moiety corresponds to or is contained within R1, R2, R3 or R7; and wherein R11 is H and the solubilising moiety corresponds to or is contained within R1, R2,
R3 Or R7.
[00108] In another embodiment, described herein are compounds of Formula (Ic) wherein:
Z is NH;
R1 and R2 are selected from H, alkyl, such as Ci_5-alkyl or aryl, such as C6 aryl, halogen, or heteroaryl;
R3 is one or more groups selected from H, amino, Ci _4 alkyl, CN, CF3, halogen, NO2 O-alkyl, NH2, NH-alkyl, N(alkyl)2
C02alkyl, CO-alkyl, CONH2, CONH-alkyl or heteroaryl or is contained with R10 or R11;
R7 is H, OMe, alkyl, or halogen, each R8, R9 or R10 is independently alkyl, OH, CF3, O-alkyl, halogen, O-heteroaryl, alkyl-heteroaryl, alkyl-cycloalkyl which may be part unsaturated; NH2 SO2-NH2, SO2-NR20, SO2-cycloheteroalkyl, SO-cycloheteroalkyl, SO2-heteroaryl,
SO-heteroaryl, CO-cycloheteroalkyl or CO-heteroarylalkyl, and the solubilising moiety corresponds to or is contained within R1, R2, R3 or R7; and wherein R11 is H and the solubilising moiety corresponds to or is contained within R1, R2,
R3 Or R7.
[00109] In a further aspect of the invention there is provided a compound of formula I as hereinbefore defined wherein one or more R19 or R20 alternatively or additionally comprise devices for immobilization thereof. Such devices may be chemical functions that can be used for covalent attachment to solid phases such as functionalized polymers (e.g. agarose, polyacrylamide, polystyrene etc.) as commonly found in matrices (microtitre plate wells, microbeads, membranes, etc.) used for biochemical assays and affinity chromatography. Alternatively, the devices may be small molecules (e.g. biotin) or polypeptides (e.g. antigens), which can be used for non-covalent immobilization through binding to an immobilized receptor (e.g. avidin or streptavidin in the case of biotin, or a specific antibody in the case of antigens).
[00110] In a further aspect of the invention there is provided a precursor to a compound of formula I as hereinbefore defined wherein one or more R19 or R20 is a solubilising moiety comprising a natural or unnatural amino acid residue, peptide or derivative as hereinbefore defined.
[00111] In one aspect, R1, R2, R3, R5, R6, R7, R8, R9, R10 and R11 are each independently selected from the group consisting of H, halogen, -NH2, -NO2, -CN, -OH, -CO2H, -CONH2, -SO3H, -SO2NH2, -SO2CH3, -OCH3, -CF3 -CH3, and -LA-LB-R32; LA is a covalent bond or an alkyl group; LB is a covalent bond, -0-, -NR32-, -NH-, -C(=0)-, -C(=0)0-,
-0C(=0)-, -C(=O)NR32-, -CC=O)NH-, -NR32C(=O)-, -NHC(=0)-, -SO2-, -SO2NR32-, - NR32SO2-, -SO2NH-, or -
NHSO2-; R32 is H, substituted or unsubstituted alkyl, haloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted non-aromatic heterocycle, or substituted or unsubstituted cycloalkyl; wherein any R32 group, when substituted, is substituted with one or more groups selected from halogen, -
NH2, -NO2, -CN, -OH, -CO2H, -CONH2, -SO3H, -SO2NH2, -SO2CH3, -OCH3, and -CF3.
[00112] In one aspect, described herein is a compound, or pharmaceutically acceptable salt, solvate, or prodrug thereof, having the structure of Formula (Ia):
Figure imgf000016_0001
Formula (Ia), wherein:
X is -C(R3)- or -N-;
R1 is unsubstituted Ci-C6 alkyl, unsubstituted or substituted aryl, or unsubstituted or substituted heteroaryl;
R2, R3, R5, R6, R7, R8, R9, R10 and R11 are each independently selected from the group consisting of H, halogen, - NH2, -NO2, -CN, -OH, -CO2H, -CONH2, -SO3H, -SO2NH2, -SO2CH3, -OCH3, -CF3, -CH3, and -LA-LB-R32;
LA is a covalent bond or an alkyl group;
LB is a covalent bond, -0-, -NR32-, -NH-, -C(=0)-, -C(=0)0-, -0C(=0)-, -C(=O)NR32-, -C(=0)NH-, -NR32C(=O)-, -NHC(=0)-, -SO2-, -SO2NR32-, - NR32SO2-, -SO2NH-, or -NHSO2-;
R32 is H, substituted or unsubstituted alkyl, haloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted non-aromatic heterocycle, or substituted or unsubstituted cycloalkyl; wherein any R32 group, when substituted, is substituted with one or more groups selected from halogen, -NH2, -NO2, -CN, -OH, -CO2H, -CONH2, -SO3H, -SO2NH2, -SO2CH3, -OCH3, and -CF3;
Z is -NRaC(=0)-, -C(=0)NRa-, -NR3SO2-, -SO2NR3-, -NR3-, -CH2NR3-, - NR3CH2-, -CH2-, -CH2CH2-, -CH=CH-, -CH2C(=0)NR3-, -NR3C(=0)CH2-, -SO2-, or -SO-;
R3 is H or alkyl.
[00113] In one aspect, X is N. In another aspect, X is -C(R3)-. [00114] In some embodiments, Z is -NR3-.
[00115] In some embodiments, R1 is unsubstituted Ci-C6 alkyl or unsubstituted or substituted phenyl. [00116] In some embodiments, each R3 is independently selected from the group consisting of H, halogen, -NH2, -NO2, -CN, -OH, -CO2H, -CONH2, -SO3H, -SO2NH2, -SO2CH3, -OCH3, -CF3, and -CH3. [00117] In some embodiments, R2, R5 and R6 are each H.
[00118] In other embodiments, R7, R8, R9, R10 and R11 are each independently selected from the group consisting of H, halogen, -NH2, -NO2, -CN, -OH, -CO2H, -CONH2, -SO3H, -SO2NH2, -SO2CH3, -OCH3, -CF3, -CH3, and -LA-LB-R32; LA is a covalent bond or an alkyl group; LB is a covalent bond, -0-, -NR32-, -NH-, -C(=0)0-, -C(=O)NR32-, -C(=0)NH-, - SO2NR32-, or -SO2NH-; R32 is H, substituted or unsubstituted alkyl, haloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted non-aromatic heterocycle, or substituted or unsubstituted cycloalkyl; wherein any R32 group, when substituted, is substituted with one or more groups selected from halogen, -NH2, -NO2, -CN, -OH, -CO2H, -CONH2, -SO3H, -SO2NH2, -SO2CH3, -OCH3, and -CF3. [00119] In one aspect, provided herein is a compound, or pharmaceutically acceptable salt, solvate, or prodrug thereof, having the structure of Formula (Ib):
Figure imgf000017_0001
Formula (Ib), wherein:
R1, R2, R3, R5, R6, R7, R8, R9, R10 and R11 are each independently selected from the group consisting of H, halogen,
-NH2, -NO2, -CN, -OH, -CO2H, -CONH2, -SO3H, -SO2NH2, -SO2CH3, -OCH3, -CF3, -CH3, and -LA-LB-R32; LA is a covalent bond or an alkyl group; LB is a covalent bond, -0-, -NR32-, -NH-, -C(=0)-, -C(=0)0-, -0C(=0)-, -C(=O)NR32-, -C(=0)NH-, -NR32C(=O)-,
-NHC(=0)-, -SO2-, -SO2NR32-, - NR32SO2-, -SO2NH-, or -NHSO2-;
R32 is H, substituted or unsubstituted alkyl, haloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted non-aromatic heterocycle, or substituted or unsubstituted cycloalkyl; wherein any R32 group, when substituted, is substituted with one or more groups selected from halogen, -NH2, -NO2, -CN, -OH, -CO2H, -CONH2, -SO3H, -SO2NH2, -SO2CH3, -OCH3, and -CF3; Z is -NRaC(=0)-, -C(=0)NRa-, -NR3SO2-, -SO2NR3-, -NR3-, -CH2NR3-, - NR3CH2-, -CH2-, -CH2CH2-, -CH=CH-,
-CH2C(=0)NR3-, -NR3CC=O)CH2-, -SO2-, or -SO-; R3 is H or alkyl.
[00120] In some embodiments, Z is -NR3-. [00121] In some embodiments, R1 is unsubstituted Ci-C6 alkyl.
[00122] In some embodiments, each R3 is independently selected from the group consisting of H, halogen, -NH2, -NO2, -CN, -OH, -CO2H, -CONH2, -SO3H, -SO2NH2, -SO2CH3, -OCH3, -CF3, and -CH3. [00123] In other embodiments, R2, R5 and R6 are each H.
[00124] In some embodiments, R7, R8, R9, R10 and R11 are each independently selected from the group consisting of H, halogen, -NH2, -NO2, -CN, -OH, -CO2H, -CONH2, -SO3H, -SO2NH2, -SO2CH3, -OCH3, -CF3 -CH3, and -LA-LB-R32, LA is a covalent bond or an alkyl group; LB is a covalent bond, -0-, -NR32-, -NH-, -C(=0)0-, -C(=O)NR32-, -C(=0)NH-, - SO2NR32-, or -SO2NH-; R32 is H, substituted or unsubstituted alkyl, haloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted non-aromatic heterocycle, or substituted or unsubstituted cycloalkyl; wherein any R32 group, when substituted, is substituted with one or more groups selected from halogen, -NH2, -NO2, -CN, -OH, -CO2H, -CONH2, -SO3H, -SO2NH2, -SO2CH3, -OCH3, and -CF3. [00125] In one aspect, provided herein is a compound, or pharmaceutically acceptable salt, solvate, or prodrug thereof, having the structure of Formula (Ic):
Figure imgf000017_0002
wherein: R1, R2, R3, R5, R6, R7, R8, R9, R10 and R11 are each independently selected from the group consisting of H, halogen, -NH2, -NO2, -CN, -OH, -CO2H, -CONH2, -SO3H, -SO2NH2, -SO2CH3, -OCH3, -CF3, -CH3, and -LA-LB-R32;
LA is a covalent bond or an alkyl group;
LB is a covalent bond, -0-, -NR32-, -NH-, -C(=0)-, -C(=0)0-, -0C(=0)-, -C(=O)NR32-, -C(=0)NH-, -NR32C(=O)-, -NHC(=0)-, -SO2-, -SO2NR32-, - NR32SO2-, -SO2NH-, or -NHSO2-;
R32 is H, substituted or unsubstituted alkyl, haloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted non-aromatic heterocycle, or substituted or unsubstituted cycloalkyl; wherein any R32 group, when substituted, is substituted with one or more groups selected from halogen, -NH2, -NO2, -CN, -OH, -CO2H, -CONH2, -SO3H, -SO2NH2, -SO2CH3, -OCH3, and -CF3;
Z is -NRaC(=0)-, -C(=0)NRa-, -NR3SO2-, -SO2NR3-, -NR3-, -CH2NR3-, - NR3CH2-, -CH2-, -CH2CH2-, -CH=CH-, -CH2C(=0)NR3-, -NR3CC=O)CH2-, -SO2-, or -SO-;
R3 is H or alkyl.
[00126] In some embodiments, Z is -NR3-. [00127] In some embodiments, R1 is unsubstituted Ci-C6 alkyl.
[00128] In some embodiments, each R3 is independently selected from the group consisting of H, halogen, -NH2, -NO2, -CN, -OH, -CO2H, -CONH2, -SO3H, -SO2NH2, -SO2CH3, -OCH3, -CF3, and -CH3. [00129] In some embodiments, R2, R5 and R6 are each H.
[00130] In some other embodiments, R7, R8, R9, R10 and R11 are each independently selected from the group consisting ofH, halogen, -NH2, -NO2, -CN, -OH, -CO2H, -CONH2, -SO3H, -SO2NH2, -SO2CH3, -OCH3, -CF3, -CH3, and -LA-LB- R32, LA is a covalent bond or an alkyl group; LB is a covalent bond, -0-, -NR32-, -NH-, -C(=0)0-, -C(=O)NR32-, - C(=0)NH-, -SO2NR32-, or -SO2NH-; R32 is H, substituted or unsubstituted alkyl, haloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted non-aromatic heterocycle, or substituted or unsubstituted cycloalkyl; wherein any R32 group, when substituted, is substituted with one or more groups selected from halogen, -NH2, -NO2, -CN, -OH, -CO2H, -CONH2, -SO3H, -SO2NH2, -SO2CH3, -OCH3, and -CF3. [00131] In some embodiments, LA is a covalent bond.
[00132] In some embodiments, LB is a covalent bond, -0-, -NH-, -C(=0)0-, -C(=0)NH-, or -SO2NH-. [00133] In some embodiments, R32 is H, substituted or unsubstituted alkyl, or haloalkyl; wherein any R32 group, when substituted, is substituted with one or more groups selected from halogen, -NH2, -NO2, -CN, -OH, -CO2H, -CONH2, - SO3H, -SO2NH2, -SO2CH3, -OCH3, and -CF3. In some embodiments, R32 is H, substituted or unsubstituted alkyl, or haloalkyl.
[00134] In one aspect, R1, R3, R7, R8, R9, R10, R11 are as defined in Table 1, Table 2, Table 3 and Table 4. In one aspect, R2, R5, and R6 are each H and R1, R3, R7, R8, R9, R10, R11 are as defined in Table 1, Table 2, Table 3 and Table 4. [00135] Any combination of the groups described above for the various variables is contemplated herein. [00136] In one aspect, compounds of Formula (I) include, but are not limited to, those described in Table 1: Table 1.
Figure imgf000018_0001
Figure imgf000018_0002
Figure imgf000019_0002
[00137] Compound in Table 1 are named:
3 -(4-( 1 -Methyl- l/f-indol-3 -yl)pyrimidin-2-ylamino)phenol (Compound 1)
4-(4-( 1 -Methyl- l/f-indol-3 -yl)pyrimidin-2-ylamino)phenol (Compound 2)
3 -Chloro-4-(4-( 1 -methyl- l/f-indol-3 -yl)pyrimidin-2-ylamino)phenol (Compound 3)
3 -Methyl-4-[4-( 1 -methyl- l/f-indol-3 -yl)-pyrimidin-2-ylamino] -phenol (Compound 4)
3 ,5-Dimethyl-4-[4-( 1 -methyl- l/f-indol-3 -yl)-pyrimidin-2-ylamino] -phenol (Compound 5)
4-[4-( 1 -Ethyl- l/f-indol-3 -yl)-pyrimidin-2-ylamino] -phenol (Compound 6)
3 -[4-( 1 -Ethyl- l/f-indol-3 -yl)-pyrimidin-2-ylamino] -phenol (Compound 7)
4-[4-( 1 -Ethyl- l/f-indol-3 -yl)-pyrimidin-2-ylamino] -3 -methyl -phenol (Compound 8)
4-[4-( 1 -Ethyl- l/f-indol-3 -yl)-pyrimidin-2-ylamino] -3 ,5-dimethyl-phenol (Compound 9)
4-[4-( 1 -Benzyl- l/f-indol-3 -yl)-pyrimidin-2-ylamino] -phenol (Compound 10)
3-[4-(l-Benzyl-l/f-indol-3-yl)-pyrimidin-2-ylamino]-phenol (Compound 11)
[00138] In one aspect, compounds of Formula (I) include, but are not limited to, those described in Table 2:
Table 2.
Figure imgf000019_0001
Figure imgf000019_0003
Figure imgf000020_0001
[00139] Compound in Table 2 are named:
4-[4-(l-Ethyl-lH-pyrrolo[2,3-b]pyridin-3-yl)-pyrimidin-2-ylamino]-phenol (Compound 12)
3 -[4-( 1 -Ethyl- lH-pyrrolo[2,3 -b]pyridin-3 -yl)-pyrimidin-2-ylamino] -phenol (Compound 13)
[4-( 1 -Ethyl- lH-pyrrolo[2,3 -£]pyridin-3 -yl)-pyrimidin-2-yl] -(3 -nitro-phenyl)-amine (Compound 14)
4-[4-( 1 -Propyl- lH-pyrrolo[2,3 -£]pyridin-3 -yl)-pyrimidin-2-ylamino] -phenol (Compound 15)
3-[4-(l-Propyl-lH-pyrrolo[2,3-£]pyridin-3-yl)-pyrimidin-2-ylamino]-phenol (Compound 16)
(3 -Nitro-phenyl)-[4-( 1 -propyl- lH-pyrrolo[2,3 -£]pyridin-3 -yl)-pyrimidin-2-yl] -amine (Compound 17)
3 -(4-( 1 -Ethyl- lH-pyrrolo[2,3 -£]pyridin-3 -yl)pyrimidin-2-ylamino)benzenesulfonamide (Compound 18)
3-(4-(l-Ethyl-l/f-pyrrolo[2,3-i]pyridin-3-yl)pyrimidin-2-ylamino)benzamide (Compound 19).
3 -(4-( 1 -ethyl- l/f-pyrrolo[2,3 -£]pyridin-3 -yl)pyrimidin-2-ylamino)benzonitrile (Compound 20).
4-(4-(l-ethyl-l/f-pyrrolo[2,3-i]pyridin-3-yl)pyrimidin-2-ylamino)benzenesulfonamide (Compound 21).
3 -(4-( 1 -propyl- l/f-pyrrolo[2,3 -£]pyridin-3 -yl)pyrimidin-2-ylamino)benzenesulfonamide (Compound 22).
4-(4-(l-Propyl-l/f-pyrrolo[2,3-i]pyridin-3-yl)pyrimidin-2-ylamino)benzenesulfonamide (Compound 23).
7V-(4-Methyl-3-(moφholinosulfonyl)phenyl)-4-(l-propyl-lH-pyrrolo[2,3-i]pyridin-3-yl)pyrimidin-2 -amine
(Compound 24).
N-(3 -(Piperazin- 1 -ylsulfonyl)phenyl)-4-( 1 -propyl- l/f-pyrrolo[2,3 -£]pyridin-3 -yl)pyrimidin-2-amine (Compound 25).
7V-(4-(Piperazin- 1 -ylsulfonyl)phenyl)-4-( 1 -propyl- l/f-pyrrolo[2,3 -£]pyridin-3 -yl)pyrimidin-2-amine (Compound 26).
7V-(4-(Moφholinosulfonyl)phenyl)-4-(l-propyl-l/f-pyrrolo[2,3-i]pyridin-3-yl)pyrimidin-2 -amine (Compound 27).
N-(3 -(4-Methylpiperazin- 1 -ylsulfonyl)phenyl)-4-( 1 -propyl- l/f-pyrrolo[2,3 -£]pyridin-3 -yl)pyrimidin-2 -amine
(Compound 28).
7V-(4-(4-Methylpiperazin- 1 -ylsulfonyl)phenyl)-4-( 1 -propyl- l/f-pyrrolo[2,3 -£]pyridin-3 -yl)pyrimidin-2 -amine
(Compound 29).
7V-(3-Moφholinophenyl)-4-(l-propyl-l/f-pyrrolo[2,3-b]pyridin-3-yl)pyrimidin-2 -amine (Compound 30).
7V-(4-Moφholinophenyl)-4-(l-propyl-l/f-pyrrolo[2,3-b]pyridin-3-yl)pyrimidin-2 -amine (Compound 31).
N-(3 -(Piperazin- 1 -yl)phenyl)-4-( 1 -propyl- l/f-pyrrolo[2,3 -b]pyridin-3 -yl)pyrimidin-2 -amine (Compound 32).
7V-(4-(Piperazin-l-yl)phenyl)-4-(l-propyl-l/f-pyrrolo[2,3-i]pyridin-3-yl)pyrimidin-2 -amine (Compound 33).
(4-Methylpiperazin- l-yl)(3 -(4-( 1 -propyl- l/f-pyrrolo[2,3 -b]pyridin-3 -yl)pyrimidin-2-ylamino)phenyl)methanone
(Compound 34).
7V-(4-(4-Methylpiperazin- 1 -yl)phenyl)-4-( 1 -propyl- l/f-pyrrolo[2,3 -i]pyridin-3 -yl)pyrimidin-2-amine (Compound 35).
1 -(4-(3 -(4-( 1 -Propyl- l/f-pyrrolo[2,3 -£]pyridin-3 -yl)pyrimidin-2-ylamino)phenyl)piperazin- 1 -yl)ethanone (Compound
36).
1 -(4-(4-(4-( 1 -Propyl- l/f-pyrrolo[2,3 -i]pyridin-3 -yl)pyrimidin-2-ylamino)phenyl)piperazin- 1 -yl)ethanone (Compound
37).
[00140] In one aspect, compounds of Formula (I) include, but are not limited to, those described in Table 3:
Table 3.
Figure imgf000021_0001
Figure imgf000021_0002
[00141] Compound in Table 3 are named:
4-(4-(5-Chloro-7 -methyl- 1 -propyl- l/f-pyrrolo[3 ,2-£]pyridin-3 -yl)pyrimidin-2-ylamino)phenol (Compound 38).
4-(4-(5-Chloro-7-methyl-l-propyl-lH-pyrrolo[3,2-i]pyridin-3-yl)pyrimidin-2-ylamino)benzenesulfonamide
(Compound 39)
3 -(4-(5-Chloro-7 -methyl- 1 -propyl- l/f-pyrrolo[3 ,2-£]pyridin-3 -yl)pyrimidin-2-ylamino)phenol (Compound 40).
4-(4-(5-Chloro-7 -methyl- 1 -propyl- l/f-pyrrolo[3 ,2-£]pyridin-3 -yl)pyrimidin-2-ylamino)-3 -methylphenol (Compound
41).
3 -(4-(5-Chloro-7 -methyl- 1 -propyl- l/f-pyrrolo[3 ,2-£]pyridin-3 -yl)pyrimidin-2-ylamino)benzenesulfonamide
(Compound 42)
4-(5-Chloro-7-methyl-l-propyl-l/f-pyrrolo[3,2-i]pyridin-3-yl)-7V-(4-methyl-3-(moφholinosulfonyl)phenyl)pyrimidin-
2-amine (Compound 43) 4-(5-Chloro-7 -methyl- 1 -propyl- l/f-pyrrolo[3 ,2-£]pyridin-3-yl)-7V-(3 -(piperazin- 1 -ylsulfonyl)phenyl)pyrimidin-2- amine (Compound 44)
4-(5-Chloro-7-methyl-l-propyl-l/f-pyrrolo[3,2-i]pyridin-3-yl)-7V-(4-(piperazin-l-ylsulfonyl)phenyl)pyrimidin-2- amine (Compound 45)
4-(5-Chloro-7 -methyl- 1 -propyl- l/f-pyrrolo[3 ,2-£]pyridin-3-yl)-7V-(3 -(piperazin- 1 -yl)phenyl)pyrimidin-2 -amine
(Compound 46).
4-(5-Chloro-7-methyl-l-propyl-l/f-pyrrolo[3,2-i]pyridin-3-yl)-7V-(4-(piperazin-l-yl)phenyl)pyrimidin-2 -amine
(Compound 47)
4-(5-Chloro-7-methyl-l-propyl-l/f-pyrrolo[3,2-i]pyridin-3-yl)-7V-(3-moφholinophenyl)pyrimidin-2 -amine
(Compound 48).
4-(5-Chloro-7-methyl-l-propyl-l/f-pyrrolo[3,2-i]pyridin-3-yl)-7V-(4-moφholinophenyl)pyrimidin-2 -amine
(Compound 49).
1 -(4-(3 -(4-(5-Chloro-7 -methyl- 1 -propyl- l/f-pyrrolo[3 ,2-£]pyridin-3 -yl)pyrimidin-2-ylamino)phenyl)piperazin- 1- yl)ethanone (Compound 50).
1 -(4-(4-(4-(5-Chloro-7 -methyl- 1 -propyl- l/f-pyrrolo[3 ,2-£]pyridin-3 -yl)pyrimidin-2-ylamino)phenyl)piperazin- 1- yl)ethanone (Compound 51).
(3 -(4-(5-Chloro-7 -methyl- 1 -propyl- l/f-pyrrolo[3 ,2-£]pyridin-3 -yl)pyrimidin-2- ylamino)phenyl)(morpholino)methanone (Compound 52).
(4-(4-(5-Chloro-7 -methyl- 1 -propyl- l/f-pyrrolo[3 ,2-£]pyridin-3 -yl)pyrimidin-2- ylamino)phenyl)(moφholino)methanone (Compound 53).
3 -(4-(7 -Methyl- 1 -propyl- l/f-pyrrolo[3 ,2-£]pyridin-3 -yl)pyrimidin-2-ylamino)benzenesulfonamide (Compound 54).
4-(4-(7-Methyl-l-propyl-l/f-pyrrolo[3,2-i]pyridin-3-yl)pyrimidin-2-ylamino)benzenesulfonamide (Compound 55).
4-(7 -Methyl- 1 -propyl- l/f-pyrrolo[3 ,2-£]pyridin-3 -yl)-7V-(3 -(piperazin- 1 -ylsulfonyl)phenyl)pyrimidin-2 -amine
(Compound 56).
4-(7 -Methyl- 1 -propyl- l/f-pyrrolo[3 ,2-b]pyridin-3 -yl)-7V-(4-(piperazin- 1 -ylsulfonyl)phenyl)pyrimidin-2 -amine
(Compound 57).
4-(7 -Methyl- 1 -propyl- l/f-pyrrolo[3 ,2-£]pyridin-3 -yl)-7V-(3 -(piperazin- 1 -yl)phenyl)pyrimidin-2 -amine (Compound 58).
4-(7 -Methyl- 1 -propyl- l/f-pyrrolo[3 ,2-£]pyridin-3 -yl)-7V-(4-(piperazin- 1 -yl)phenyl)pyrimidin-2 -amine (Compound 59).
4-(7 -Methyl- 1 -propyl- lH-pyrrolo[3 ,2-b]pyridin-3 -yl)-7V-(3 -moφholinophenyl)pyrimidin-2 -amine (Compound 60).
4-(7-Methyl-l-propyl-lH-pyrrolo[3,2-b]pyridin-3-yl)-7V-(4-moφholinophenyl)pyrimidin-2 -amine (Compound 61).
1 -(4-(3 -(4-(7 -Methyl- 1 -propyl- l/f-pyrrolo[3 ,2-£]pyridin-3-yl)pyrimidin-2-ylamino)phenyl)piperazin- 1 -yl)ethanone
(Compound 62).
1 -(4-(4-(4-(7 -Methyl- 1 -propyl- l/f-pyrrolo[3 ,2-£]pyridin-3-yl)pyrimidin-2-ylamino)phenyl)piperazin- 1 -yl)ethanone
(Compound 63).
3 -(4-( 1 -Ethyl-7 -methyl- l/f-pyrrolo[3 ,2-i]pyridin-3 -yl)pyrimidin-2-ylamino)benzenesulfonamide (Compound 64).
4-(4-( 1 -Ethyl-7 -methyl- l/f-pyrrolo[3 ,2-i]pyridin-3 -yl)pyrimidin-2-ylamino)benzenesulfonamide (Compound 65).
(3 -(4-( 1 -Ethyl-7 -methyl- l/f-pyrrolo[3 ,2-i]pyridin-3 -yl)pyrimidin-2-ylamino)phenyl)(moφholino)methanone
(Compound 66).
(4-(4-( 1 -Ethyl-7 -methyl- l/f-pyrrolo[3 ,2-i]pyridin-3 -yl)pyrimidin-2-ylamino)phenyl)(moφholino)methanone
(Compound 67).
4-( 1 -Ethyl-7 -methyl- l/f-pyrrolo[3 ,2-£]pyridin-3 -yl)-7V-(3 -(piperazin- 1 -yl)phenyl)pyrimidin-2 -amine (Compound 68).
4-( 1 -Ethyl-7 -methyl- l/f-pyrrolo[3 ,2-£]pyridin-3 -yl)-7V-(4-(piperazin- 1 -yl)phenyl)pyrimidin-2 -amine (Compound 69). Table 4.
Figure imgf000023_0001
Figure imgf000023_0002
[00142] Compound in Table 4 are named:
4-(4-(7-Chloro- 1 -ethyl- l/f-pyrrolo[2,3 -c]pyridin-3 -yl)pyrimidin-2-ylamino)phenol (Compound 70) 4-(4-(7-Chloro-l-ethyl-l/f-pyrrolo[2,3-c]pyridin-3-yl)pyrimidin-2-ylamino)benzenesulfonamide (Compound 71) 3 -(4-(7-Chloro- 1 -ethyl- l/f-pyrrolo[2,3 -c]pyridin-3 -yl)pyrimidin-2-ylamino)phenol (Compound 72) 3-(4-(7-Chloro-l-ethyl-l/f-pyrrolo[2,3-c]pyridin-3-yl)pyrimidin-2-ylamino)benzenesulfonamide (Compound 73)
4-(7-Chloro- 1 -ethyl- l/f-pyrrolo[2,3 -c]pyridin-3 -yl)-7V-(4-methyl-3 -(moφholinosulfonyl)phenyl)pyrimidin-2 -amine
(Compound 74)
(3 -(4-(7-Chloro- 1 -ethyl- l/f-pyrrolo[2,3 -c]pyridin-3 -yl)pyrimidin-2-ylamino)phenyl)(morpholino)methanone
(Compound 75)
(4-(4-(7-Chloro-l-ethyl-l/f-pyrrolo[2,3-c]pyridin-3-yl)pyrimidin-2-ylamino)phenyl)(moφholino)methanone
(Compound 76)
4-(7-Chloro- 1 -ethyl- l/f-pyrrolo[2,3 -c]pyridin-3 -yl)-7V-(3 -(piperazin- 1 -ylsulfonyl)phenyl)pyrimidin-2 -amine
(Compound 77)
4-(7-Chloro- 1 -ethyl- l/f-pyrrolo[2,3 -c]pyridin-3 -yl)-7V-(4-(piperazin- 1 -ylsulfonyl)phenyl)pyrimidin-2 -amine
(Compound 78).
4-(7-Chloro- 1 -ethyl- l/f-pyrrolo[2,3 -c]pyridin-3 -yl)-7V-(3 -(piperazin- 1 -yl)phenyl)pyrimidin-2 -amine (Compound 79).
4-(7-Chloro- 1 -ethyl- l/f-pyrrolo[2,3 -c]pyridin-3 -yl)-7V-(4-(piperazin- 1 -yl)phenyl)pyrimidin-2 -amine (Compound 80).
4-(7-Chloro-l-ethyl-l/f-pyrrolo[2,3-c]pyridin-3-yl)-7V-(3-moφholinophenyl)pyrimidin-2 -amine (Compound 81).
4-(7-Chloro-l-ethyl-l/f-pyrrolo[2,3-c]pyridin-3-yl)-7V-(4-moφholinophenyl)pyrimidin-2 -amine (Compound 82).
3 -(4-( 1 -Ethyl- 1/f-pyrrolo [2,3 -c]pyridin-3 -yl)pyrimidin-2-ylamino)phenol (Compound 83 ).
3-(4-(l-Ethyl-l/f-pyrrolo[2,3-c]pyridin-3-yl)pyrimidin-2-ylamino)benzenesulfonamide (Compound 84).
4-(4-(l-Ethyl-l/f-pyrrolo[2,3-c]pyridin-3-yl)pyrimidin-2-ylamino)phenol (Compound 85).
4-(4-(l-Ethyl-l/f-pyrrolo[2,3-c]pyridin-3-yl)pyrimidin-2-ylamino)benzenesulfonamide (Compound 86).
4-( 1 -Ethyl- l/f-pyrrolo[2,3 -c]pyridin-3 -yl)-7V-(4-methyl-3 -(moφholinosulfonyl)phenyl)pyrimidin-2 -amine (Compound
87).
(3-(4-(l-Ethyl-l/f-pyrrolo[2,3-c]pyridin-3-yl)pyrimidin-2-ylamino)phenyl)(moφholino)methanone (Compound 88).
(4-(4-(l-Ethyl-l/f-pyrrolo[2,3-c]pyridin-3-yl)pyrimidin-2-ylamino)phenyl)(moφholino)methanone (Compound 89).
1 -(4-(3 -(4-( 1 -Ethyl- l/f-pyrrolo[2,3 -c]pyridin-3 -yl)pyrimidin-2-ylamino)benzoyl)piperazin-l -yl)ethanone (Compound
90).
1 -(4-(4-(4-( 1 -Ethyl- l/f-pyrrolo[2,3 -c]pyridin-3 -yl)pyrimidin-2-ylamino)benzoyl)piperazin-l -yl)ethanone (Compound
91).
4-( 1 -Ethyl- l/f-pyrrolo[2,3 -c]pyridin-3 -yl)-7V-(3 -(piperazin- l-yl)phenyl)pyrimidin-2 -amine (Compound 92).
4-(l-Ethyl-l/f-pyrrolo[2,3-c]pyridin-3-yl)-7V-(4-(piperazin-l-yl)phenyl)pyrimidin-2 -amine (Compound 93).
4-( 1 -Ethyl- l/f-pyrrolo[2,3 -c]pyridin-3 -yl)-7V-(3 -moφholinophenyl)pyrimidin-2 -amine (Compound 94).
4-( 1 -Ethyl- l/f-pyrrolo[2,3 -c]pyridin-3 -yl)-7V-(4-moφholinophenyl)pyrimidin-2 -amine (Compound 95).
4-(4-(7-Chloro-l-propyl-l/f-pyrrolo[2,3-c]pyridin-3-yl)pyrimidin-2-ylamino)benzenesulfonamide (Compound 96).
3 -(4-(7-Chloro- 1 -propyl- l/f-pyrrolo[2,3 -c]pyridin-3 -yl)pyrimidin-2-ylamino)benzenesulfonamide (Compound 97). l-(4-(3-(4-(7-Chloro-l-propyl-l/f-pyrrolo[2,3-c]pyridin-3-yl)pyrimidin-2-ylamino)benzoyl)piperazin-l-yl)ethanone
(Compound 98). l-(4-(4-(4-(7-Chloro-l-propyl-l/f-pyrrolo[2,3-c]pyridin-3-yl)pyrimidin-2-ylamino)benzoyl)piperazin-l-yl)ethanone
(Compound 99).
4-(7-Chloro-l-propyl-lH-pyrrolo[2,3-c]pyridin-3-yl)-N-(4-moφholinophenyl)pyrimidin-2 -amine (Compound 100).
4-(7-Chloro- 1 -propyl- l/f-pyrrolo[2,3 -c]pyridin-3 -yl)-7V-(4-methyl-3 -(moφholinosulfonyl)phenyl)pyrimidin-2 -amine
(Compound 101).
7V-(4-Methyl-3 -(moφholinosulfonyl)phenyl)-4-( 1 -propyl- l/f-pyrrolo[2,3 -c]pyridin-3 -yl)pyrimidin-2 -amine
(Compound 102). 7V-(4-Moφholinophenyl)-4-(l-propyl-l/f-pyrrolo[2,3-c]pyridin-3-yl)pyrimidin-2 -amine (Compound 103).
N-(3 -(Piperazin- 1 -yl)phenyl)-4-( 1 -propyl- l/f-pyrrolo[2,3 -c]pyridin-3 -yl)pyrimidin-2 -amine (Compound 104).
7V-(4-(Piperazin- 1 -yl)phenyl)-4-( 1 -propyl- l/f-pyrrolo[2,3 -c]pyridin-3 -yl)pyrimidin-2 -amine (Compound 105).
1 -(4-(4-(4-( 1 -Propyl- l/f-pyrrolo[2,3 -c]pyridin-3 -yl)pyrimidin-2-ylamino)benzoyl)piperazin- 1 -yl)ethanone (Compound
106).
Piperazin- 1 -yl(3 -(4-( 1 -propyl- 1/f-pyrrolo [2,3 -c]pyridin-3 -yl)pyrimidin-2-ylamino)phenyl)methanone (Compound 107).
Moφholino(3-(4-(l-propyl-l/f-pyrrolo[2,3-c]pyridin-3-yl)pyrimidin-2-ylamino)phenyl)methanone (Compound 108).
Piperazin-l-yl(4-(4-(l-propyl-l/f-pyrrolo[2,3-c]pyridin-3-yl)pyrimidin-2-ylamino)phenyl)methanone (Compound 109).
Moφholino(4-(4-(l-propyl-l/f-pyrrolo[2,3-c]pyridin-3-yl)pyrimidin-2-ylamino)phenyl)methanone (Compound 110).
3-(4-(l-Propyl-l/f-pyrrolo[2,3-c]pyridin-3-yl)pyrimidin-2-ylamino)benzenesulfonamide (Compound 111).
4-(4-(l-Propyl-l/f-pyrrolo[2,3-c]pyridin-3-yl)pyrimidin-2-ylamino)benzenesulfonamide (Compound 112).
Synthesis of Compounds
[00143] Compounds of Formula (I) described in the prior section are synthesized using standard synthetic techniques or using methods known in the art in combination with methods described herein. In additions, solvents, temperatures and other reaction conditions presented herein may vary.
[00144] The starting material used for the synthesis of the compounds of Formula (I) described in the prior section are either synthesized or obtained from commercial sources, such as, but not limited to, Aldrich Chemical Co. (Milwaukee,
Wis.), or Sigma Chemical Co. (St. Louis, Mo.). The compounds described herein, and other related compounds having different substituents are synthesized using known techniques and materials, including those found in March,
ADVANCED ORGANIC CHEMISTRY 4th Ed., (Wiley 1992); Carey and Sundberg, ADVANCED ORGANIC CHEMISTRY 4th
Ed., VoIs. A and B (Plenum 2000, 2001), and Green and Wuts, PROTECTIVE GROUPS IN ORGANIC SYNTHESIS 3rd Ed.,
(Wiley 1999). General methods for the preparation of compounds can be modified by the use of appropriate reagents and conditions for the introduction of the various moieties found in the formulae as provided herein.
[00145] In one aspect, compounds described herein are prepared according to methods known in the art. In one aspect, compounds disclosed herein are prepared as outlined in Scheme 1 and Scheme 2.
Scheme 1
Figure imgf000025_0001
[00146] In one aspect, acylheteroaryl compounds of structure l-II are converted to an acrylate compound of structure l-III. In one aspect, Het-Ar is a bicylic heteroaryl that includes at least one nitrogen in the bicyclic ring. In one aspect, Het-Ar is selected from indole, pyrrolo[2,3-£]pyridine, pyrrolo[2,3-c]pyridine, pyrrolo[3,2-£]pyridine, and pyrrolo[3,2- cjpyridine, where Het-Ar is substituted or unsubstituted. In one aspect, Het-Ar is selected from indol-3-yl, pyrrolo[2,3- £]pyridine-2-yl, pyrrolo[2,3-c]pyridine-3-yl, pyrrolo[3,2-£]pyridine-3-yl and pyrrolo[3,2-c]pyridine-3-yl, where Het-Ar is substituted or unsusbtituted.
[00147] In one aspect, acrylate compounds of structure l-III are reacted with phenylguanidine compounds of structure
1-IV to provide compounds of Formula (I). In one aspect, L1 is any leaving group. In some embodiments, L1 is
N(alkyl)2, halogen, ester (-OC(=O)R), or thioester. In one aspect, L1 is -NMe2.
[00148] In another aspect, a compound of structure 1-VI, where W is a halide, triflate or boronic acid, boronate ester, or other suitable group, is reacted with a pyrimidine compound of structure 1-V, where L2 and L3 are leaving groups, such as a halide. Suitable reaction conditions include, but are not limited to, metal-mediated reactions such as Suzuki reactions, Stille cross couplings, Negishi couplings, Kumada couplings, Ullmann reactions, Hiyama Coupling, and variants thereof (Metal-Catalyzed Cross-Coupling Reactions, Armin de Meijere (Editor), Francois Diederich (Editor),
John Wiley & Sons; 2nd edition, 2004; Ozdemir, et al., Tetrahedron, 2005, 61, 9791-9798; Ackermann, et al, Org.
Lett., 2006, 8, 3457-3460; Blakey, et al., J. Am. Chem. Soc, 2003, 125, 6046-6047; Dai, et al., Org. Lett., 2004, 6, 221-
224; Yoshikai, et al, J. Am. Chem. Soc, 2005, 127, 17978-17979; Tang, et al, J. Org. Chem., 2006, 71, 2167-2169;
Murata, et al, Synthesis, 2001, 2231-2233).
[00149] In one aspect, compounds of structure 1-VII are reacted with compounds of 1-VIII to provide compounds of
Formula (I). In one aspect, Z is NH. Suitable reaction conditions include, but are not limited to, metal-catalyzed cross couplings and SNAR reaction conditions.
[00150] In one aspect, treatment of 1-V with 1-VI, and a Grignard reagent such as alkyl magnesium bromide, forms 1-
VII which reacts as hereinbefore defined with 1-VIII to provide the compounds of formula I.
[00151] In one aspect, Het-Ar is a 1 -substituted indol-3-yl, 1-substituted pyrrolo[2,3-£]pyridine-3-yl, 1-substituted pyrrolo[2,3-c]pyridine-3-yl, or 1-substituted pyrrolo[3,2-£]pyridine-3-yl. In one aspect, the 1-substitutuent of the 1- substituted indol-3-yl, 1-substituted pyrrolo[2,3-£]pyridine-3-yl, 1-substituted pyrrolo[2,3-c]pyridine-3-yl, or 1- substituted pyrrolo[3,2-£]pyridine-3-yl is selected from -Q-Cβalkyl, and -Q-Cβalkyltsubstituted or unsubstituted aryl).
In one aspect, the 1-substitutuent of the 1-substituted indol-3-yl, 1-substituted pyrrolo[2,3-£]pyridine-3-yl, 1- substituted pyrrolo[2,3-c]pyridine-3-yl, or 1-substituted pyrrolo[3,2-£]pyridine-3-yl is selected from -CH3, -CH2CH3, -
CH2CH2CH3, and benzyl.
[00152] In one aspect, methods and reaction conditions are as described in Wang, S, et al. J. Med. Chem. 2004, 47,
1662.
[00153] In another aspect, compounds described herein are prepared as outlined in Scheme 2.
Scheme 2
Figure imgf000027_0001
[00154] In one aspect, nitrobenzene compounds or nitro-pyridine compounds of structure 2-IX, where X is either N or
C-R3, are reacted with vinylmagnesium bromide to provide compounds of structure 2-XI. In one aspect, at least two X are C-R3. In one aspect, at least three X are C-R3. In one aspect, the process of reacting 2-IX with a Grignard agent is as described in Zhang, Z, et al. J. Org. Chem., 2002, 67, 2345.
[00155] N-Alkylation of compounds of structure 2-XI with compounds of 2-XII (where Hal is a halide) provides compounds of structure 2-XIII. In one aspect, compounds of structure 2-XI are reacted with compounds of 2-XII in the presence of a base, such as, but not limited to NaH or KOH.
[00156] Compounds of structure 2-XIII are acylated with compounds of structure 2-XIV to provide compounds of structure 2-XV. Suitable reaction conditions include, but are not limited to, Friedel-Crafts acylation conditions.
[00157] Compounds of structure 2-XV are converted to acrylate compounds of structure 2-XVI, where L1 is any leaving group. In some embodiments, L1 is N(alkyl)2, halogen, ester (-OC(=O)R), or thioester. In one aspect, L1 is -
NMe2. In one aspect, compounds of structure 2-XV are reacted with 7V,7V'-dimethylformamide dimethylacetal (where
R6 = H, L1 = NMe2) or ferZ-butoxy-bis(dimethylamino)methane (Bredereck, H.;.et al. Chemische Benchte 1964, 97,
(12), 3397) to provide acrylate compounds of structure 2-XVI.
[00158] Compounds of structure 2-XVI are reacted with phenylguanidine of structure 1-IV to provide compounds of
Formula (I).
[00159] In one aspect, phenylguanidines of structure 1-IV are obtained by reaction of cyanamide or cyanamide derivatives using the method of Katritzky, A. R.; et al. Synthetic Communications 1995, 25, 1173.
[00160] In one aspect, compounds of Formula (I) are synthesized as outlined in the Examples.
Use of Protecting Groups
[00161] In the reactions described, it is necessary in certain embodiments to protect reactive functional groups, for example hydroxy, amino, thiol or carboxy groups, where these are desired in the final product, to avoid their unwanted participation in the reactions. Protecting groups are used to block some or all reactive moieties and prevent such groups from participating in chemical reactions until the protective group is removed. In one embodiment, each protective group is removable by a different means. Protective groups that are cleaved under totally disparate reaction conditions fulfill the requirement of differential removal. In some embodiments, protective groups are removed by acid, base, and/or hydrogenolysis. Groups such as trityl, dimethoxytrityl, acetal and t-butyldimethylsilyl are acid labile and are used in certain embodiments to protect carboxy and hydroxy reactive moieties in the presence of amino groups protected with Cbz groups, which are removable by hydrogenolysis, and/or Fmoc groups, which are base labile. In other embodiments, carboxylic acid and hydroxy reactive moieties are blocked with base labile groups such as, but not limited to, methyl, ethyl, and acetyl in the presence of amines blocked with acid labile groups such as t-butyl carbamate or with carbamates that are both acid and base stable but hydrolytically removable.
[00162] In another embodiment, carboxylic acid and hydroxy reactive moieties are blocked with hydrolytically removable protective groups such as the benzyl group, while amine groups capable of hydrogen bonding with acids are blocked with base labile groups such as Fmoc. In another embodiment, carboxylic acid reactive moieties are protected by conversion to simple ester compounds as exemplified herein, or they are, in yet another embodiment, blocked with oxidatively-removable protective groups such as 2,4-dimethoxybenzyl, while co-existing amino groups are blocked with fluoride labile silyl carbamates.
[00163] Allyl blocking groups are useful in the presence of acid- and base- protecting groups since the former are stable and can be subsequently removed by metal or pi-acid catalysts. For example, an allyl-blocked carboxylic acid can be deprotected with a Pd(O)-catalyzed reaction in the presence of acid labile t-butyl carbamate or base-labile acetate amine protecting groups. Yet another form of protecting group is a resin to which a compound or intermediate is attached. As long as the residue is attached to the resin, that functional group is blocked and cannot react. Once released from the resin, the functional group is available to react. [00164] Typically blocking/protecting groups are, by way of example only:
Figure imgf000028_0001
*j H3C^ ^H3 O
(H3C)3C^ (H3C)3C-Si\j- Me3SK ^i
Et TBDMS Teoc
Figure imgf000028_0003
Figure imgf000028_0002
BOC BMBn trit^l acetyl Fmoc
[00165] Other protecting groups, plus a detailed description of techniques applicable to the creation of protecting groups and their removal are described in Greene and Wuts, Protective Groups in Organic Synthesis, 3rd Ed., John Wiley & Sons, New York, NY, 1999, and Kocienski, Protective Groups, Thieme Verlag, New York, NY, 1994, which are incorporated herein by reference for such disclosure. Further Forms of Compounds
[00166] In certain embodiments, compounds of Formula (I) are prepared as a pharmaceutically acceptable acid addition salt (which is a type of a pharmaceutically acceptable salt) by reacting the free base form of the compound with a pharmaceutically acceptable inorganic or organic acid, including, but not limited to, inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid metaphosphoric acid, and the like; and organic acids such as acetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, p-toluenesulfonic acid, tartaric acid, trifluoroacetic acid, citric acid, benzoic acid, 3-(4-hydroxybenzoyl)benzoic acid, cinnamic acid, mandelic acid, arylsulfonic acid, methanesulfonic acid, ethanesulfonic acid, 1,2-ethanedisulfonic acid, 2-hydroxyethanesulfonic acid, benzenesulfonic acid, 2-naphthalenesulfonic acid, 4-methylbicyclo-[2.2.2]oct-2-ene-l-carboxylic acid, glucoheptonic acid, 4,4'-methylenebis-(3-hydroxy-2-ene-l -carboxylic acid), 3-phenylpropionic acid, trimethylacetic acid, tertiary butylacetic acid, lauryl sulfuric acid, gluconic acid, glutamic acid, hydroxynaphthoic acid, salicylic acid, stearic acid, and muconic acid.
[00167] By "pharmaceutically acceptable," as used herein, refers a material, such as a carrier or diluent, which does not abrogate the biological activity or properties of the compound, and is relatively nontoxic, i.e., the material may be administered to an individual without causing undesirable biological effects or interacting in a deleterious manner with any of the components of the composition in which it is contained.
[00168] The term "pharmaceutically acceptable salt" refers to a formulation of a compound that does not cause significant irritation to an organism to which it is administered and does not abrogate the biological activity and properties of the compound. In some embodiments, pharmaceutically acceptable salts are obtained by reacting a compound of Formula (I) with acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesurfonic acid, salicylic acid and the like. Pharmaceutically acceptable salts are also obtained by reacting a compound of Formula (I) with a base to form a salt such as an ammonium salt, an alkali metal salt, such as a sodium or a potassium salt, an alkaline earth metal salt, such as a calcium or a magnesium salt, a salt of organic bases such as dicyclohexylamine, TV-methyl-D-glucamine, tris(hydroxymethyl)methylamine, and salts with amino acids such as arginine, lysine, and the like. [00169] In other embodiments, compounds of Formula (I) are prepared as a pharmaceutically acceptable salts by reacting the free acid form of the compound with a pharmaceutically acceptable inorganic or organic base, including, but not limited to organic bases such as ethanolamine, diethanolamine, triethanolamine, tromethamine, N- methylglucamine, and the like, or with an inorganic base such as aluminum hydroxide, calcium hydroxide, potassium hydroxide, sodium carbonate, sodium hydroxide, and the like.
[00170] It should be understood that a reference to a pharmaceutically acceptable salt includes the solvent addition forms or crystal forms thereof, particularly solvates or polymorphs. Solvates contain either stoichiometric or non- stoichiometric amounts of a solvent, and are optionally formed during the process of crystallization with pharmaceutically acceptable solvents such as water, ethanol, and the like. Hydrates are formed when the solvent is water, or alcoholates are formed when the solvent is alcohol. Solvates of compounds of Formula (I) are conveniently prepared or formed during the processes described herein. By way of example only, hydrates of compounds of Formula (I) are conveniently prepared by recrystallization from an aqueous/organic solvent mixture, using organic solvents including, but not limited to, dioxane, tetrahydrofuran, ethanol, or methanol. In addition, the compounds provided herein can exist in unsolvated as well as solvated forms. In general, the solvated forms are considered equivalent to the unsolvated forms for the purposes of the compounds and methods provided herein.
[00171] In yet other embodiments, the compounds of Formula (I) are prepared in various forms, including but not limited to, amorphous forms, milled forms and nano-particulate forms. In addition, compounds of Formula (I) include crystalline forms, also known as polymorphs. Polymorphs include the different crystal packing arrangements of the same elemental composition of a compound. Polymorphs usually have different X-ray diffraction patterns, infrared spectra, melting points, density, hardness, crystal shape, optical and electrical properties, stability, and solubility. Various factors such as the recrystallization solvent, rate of crystallization, and storage temperature may cause a single crystal form to dominate.
[00172] In some embodiments, compounds of Formula (I) are prepared as prodrugs. A "prodrug" refers to an agent that is converted into the parent drug in vivo. Prodrugs are often useful because, in some situations, they may be easier to administer than the parent drug. They may, for instance, be bioavailable by oral administration whereas the parent is not. The prodrug may also have improved solubility in pharmaceutical compositions over the parent drug. An example, without limitation, of a prodrug would be a compound of Formula (I) which is administered as an ester (the "prodrug") to facilitate transmittal across a cell membrane where water solubility is detrimental to mobility but which then is metabolically hydrolyzed to the carboxylic acid, the active entity, once inside the cell where water-solubility is beneficial. A further example of a prodrug might be a short peptide (polyaminoacid) bonded to an acid group where the peptide is metabolized to reveal the active moiety.
[00173] Prodrugs are generally drug precursors that, following administration to a subject and subsequent absorption, are converted to an active, or a more active species via some process, such as conversion by a metabolic pathway. Some prodrugs have a chemical group present on the prodrug that renders it less active and/or confers solubility or some other property to the drug. Once the chemical group has been cleaved and/or modified from the prodrug the active drug is generated. Prodrugs are often useful because, in some situations, they are easier to administer than the parent drug. In certain embodiments, the prodrug of a compound described herein is bioavailable by oral administration whereas the parent is not. Furthermore, in some embodiments, the prodrug of a compound described herein has improved solubility in pharmaceutical compositions over the parent drug.
[00174] In other embodiments, prodrugs are designed as reversible drug derivatives, for use as modifiers to enhance drug transport to site-specific tissues. In specific embodiments, the design of prodrugs to date is to increase the effective water solubility of the therapeutic compound for targeting to regions where water is the principal solvent. Fedorak et al., Am. J. Physiol., 269:G210-218 (1995); McLoed et al., Gastroenterol, 106:405-413 (1994); Hochhaus et al, Biomed. Chrom., 6:283-286 (1992); J. Larsen and H. Bundgaard, Int. J. Pharmaceutics, 37, 87 (1987); J. Larsen et al, Int. J. Pharmaceutics, 47, 103 (1988); Sinkula e/ α/., J. Pharm. Sa., 64: 181-210 (1975); T. Higuchi and V. Stella, Prodrugs as Novel Delivery Systems, Vol. 14 of the A. CS. Symposium Series; and Edward B. Roche, Bioreversible Carriers in Drug Design, American Pharmaceutical Association and Pergamon Press, 1987. [00175] In some embodiments, sites on the aromatic ring portion of compounds of Formula (I) are susceptible to various metabolic reactions Therefore incorporation of appropriate substituents on the aromatic ring structures will reduce, minimize or eliminate this metabolic pathway. In specific embodiments, the appropriate substituent to decrease or eliminate the susceptibility of the aromatic ring to metabolic reactions is, by way of example only, a halogen, or an alkyl group.
[00176] In another embodiment, the compounds described herein are labeled isotopically (e.g. with a radioisotope) or by another other means, including, but not limited to, the use of chromophores or fluorescent moieties, bioluminescent labels, or chemiluminescent labels.
[00177] In yet another embodiment, the compounds of Formula (I) possess one or more stereocenters and each center exists independently in either the R or S configuration. The compounds presented herein include all diastereomeric, enantiomeric, and epimeric forms as well as the appropriate mixtures thereof. In certain embodiments, compounds of Formula (I) are prepared as their individual stereoisomers by reacting a racemic mixture of the compound with an optically active resolving agent to form a pair of diastereoisomeric compounds, separating the diastereomers and recovering the optically pure enantiomers. In some embodiments, resolution of enantiomers is carried out using covalent diastereomeric derivatives of the compounds described herein. In other embodiments, dissociable complexes are utilized (e.g., crystalline diastereomeric salts). Diastereomers have distinct physical properties (e.g., melting points, boiling points, solubilities, reactivity, etc.) and are, in specific embodiments, separated by taking advantage of these dissimilarities. In these embodiments, the diastereomers are separated by chiral chromatography or by separation/resolution techniques based upon differences in solubility. The optically pure enantiomer is then recovered, along with the resolving agent, by any practical means that does not result in racemization. Jean Jacques, Andre Collet, Samuel H. Wilen, "Enantiomers, Racemates and Resolutions", John Wiley And Sons, Inc., 1981. [00178] Additionally, in certain embodiments, the compounds provided herein exist as geometric isomers. The compounds and methods provided herein include all cis, trans, syn, anti, entgegen (E), and zusammen (Z) isomers as well as the appropriate mixtures thereof. In some embodiments, the compounds described herein exist as tautomers. All tautomers are intended to be within the scope of the molecular formulas described herein. In additional embodiments of the compounds and methods provided herein, mixtures of enantiomers and/or diastereoisomers, resulting from a single preparative step, combination, or interconversion are envisioned.
Certain Terminology
[00179] Unless otherwise stated, the following terms used in this application, including the specification and claims, have the definitions given below. It must be noted that, as used in the specification and the appended claims, the singular forms "a," "an" and "the" include plural referents unless the context clearly dictates otherwise. Unless otherwise indicated, conventional methods of mass spectroscopy, NMR, HPLC, protein chemistry, biochemistry, recombinant DNA techniques and pharmacology are employed. In this application, the use of "or" or "and" means
"and/or" unless stated otherwise. Furthermore, use of the term "including" as well as other forms, such as "include",
"includes," and "included," is not limiting.
[00180] "Compound of Formula (I)" refers to compound of Formula (I), compound of Formula (Ia), compound of
Formula (Ib), compound of Formula (Ic), compound of Formula (Id).
[00181] An "alkyl" group refers to an aliphatic hydrocarbon group. The alkyl moiety may be a saturated alkyl group
(which means that it does not contain any units of unsaturation, e.g. carbon-carbon double bonds or carbon-carbon triple bonds) or the alkyl moiety may be an unsaturated alkyl group (which means that it contains at least one unit of unsaturation). The alkyl moiety, whether saturated or unsaturated, may be branched, or straight chain.
[00182] The "alkyl" moiety may have 1 to 8 carbon atoms (whenever it appears herein, a numerical range such as "1 to 8" refers to each integer in the given range; e.g. , " 1 to 8 carbon atoms" means that the alkyl group may consist of 1 carbon atom, 2 carbon atoms, 3 carbon atoms, etc., up to and including 8 carbon atoms, although the present definition also covers the occurrence of the term "alkyl" where no numerical range is designated). The alkyl group of the compounds described herein may be designated as "Ci-C6 alkyl" or similar designations. By way of example only, "Ci-
C6 alkyl" indicates that there are one, two , three, four, five, or six carbon atoms in the alkyl chain. Typical alkyl groups include, but are in no way limited to, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tertiary butyl, pentyl, hexyl, and the like. The term "lower alkyl" is similarly used for groups having from 1 to 4 carbon atoms.
[00183] The term "aralkyl" is refers to -alkyl-aryl, where alkyl and aryl are as defined herein.
[00184] The term "alicyclic" refers to a cyclic aliphatic group.
[00185] The term "aliphatic" takes its normal meaning in the art and includes non-aromatic groups such as alkanes, alkenes and alkynes and substituted derivatives thereof.
[00186] An "alkoxy" group refers to a (alkyl)O- group, where alkyl is as defined herein.
[00187] The term "alkylamine" refers to the -N(alkyl)xHy group, where x and y are selected from the group x=l, y=l and x=2, y=0. In some embodiments, when x=2 and y=0, the alkyl groups taken together with the nitrogen atom to which they are attached form a cyclic ring system.
[00188] An "amide" is a chemical moiety with formula -C(=O)NHR or -NHC(=O)R, where R is selected from the group consisting of alkyl, cycloalkyl, aryl, heteroaryl (bonded through a ring carbon) and heteroalicyclic (bonded through a ring carbon). An amide may be an amino acid or a peptide molecule attached to a compound of Formula (I), thereby forming a prodrug. Any amine, or carboxyl side chain on the compounds described herein is optionally amidified, as desired. See, e.g., Greene and Wuts, Protective Groups in Organic Synthesis, 3rd Ed., John Wiley & Sons,
New York, NY, 1999, is incorporated herein by reference for such disclosure. [00189] The term "aromatic" refers to a planar ring having a delocalized π-electron system containing 4n+2 π electrons, where n is an integer. Aromatic rings can be formed from five, six, seven, eight, nine, ten, or more than ten atoms. Aromatics are optionally substituted. The term "aromatic" includes both carbocyclic aryl ("aryl", e.g., phenyl) and heterocyclic aryl (or "heteroaryl" or "heteroaromatic") groups (e.g., pyridine). The term includes monocyclic or fused-ring polycyclic (i.e., rings which share adjacent pairs of carbon atoms) groups.
[00190] The term "carbocyclic" refers to a ring or ring system where the atoms forming the backbone of the ring are all carbon atoms. The term thus distinguishes carbocyclic from heterocyclic rings in which the ring backbone contains at least one atom which is different from carbon.
[00191] As used herein, the term "aryl" refers to an aromatic ring wherein each of the atoms forming the ring is a carbon atom. Aryl rings are formed by five, six, seven, eight, nine, or more than nine carbon atoms. Aryl groups are optionally substituted. In one aspect, an aryl is a phenyl or a naphthalenyl. Depending on the structure, an aryl group can be a monoradical or a diradical (i.e., an arylene group). In one aspect, an aryl is a C6-Q oaryl.
The term "cycloalkyl" refers to a monocyclic or polycyclic aliphatic, non-aromatic radical, wherein each of the atoms forming the ring (i.e. skeletal atoms) is a carbon atom. Cycloalkyls may be saturated, or partially unsaturated.
Cycloalkyls may be fused with an aromatic ring, and the point of attachment is at a carbon that is not an aromatic ring carbon atom. Cycloalkyl groups include groups having from 3 to 10 ring atoms. In some embodiments, cycloalkyl groups are selected from among cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl.
Cycloalkyl groups may be substituted or unsubstituted.
[00192] As used herein, the term "carbohydrate derivative" refers to a compound of general formula Cx(H2O)y or a derivative thereof. Preferably, the carbohydrate is a mono-, di- or tri-saccharide. Monosaccharides can exist as either straight chain or ring-shaped molecules and are classified according to the number of carbon atoms they possess; tnoses have three carbons, tetroses four, pentoses five and hexoses six. Each of these subgroups may be further divided into aldoses and ketoses, depending on whether the molecule contains an aldehyde group (-CHO) or a ketone group
(C=O). Typical examples of monosaccharides include glucose, fructose, and galactose. Disaccharides consist of two linked monosaccharide molecules, and include for example, maltose and lactose. Trisaccharides consist of three linked monosaccharide molecules.
[00193] The term "ester" refers to a chemical moiety with formula -COOR, where R is selected from the group consisting of alkyl, cycloalkyl, aryl, heteroaryl (bonded through a ring carbon) and heteroalicyclic (bonded through a ring carbon). Any hydroxy, or carboxyl side chain on the compounds described herein is esterified, if desired.
Examples of procedures and specific groups to make such esters are found in sources such as Greene and Wuts,
Protective Groups in Organic Synthesis, 3rd Ed., John Wiley & Sons, New York, NY, 1999.
[00194] The term "halo" or, alternatively, "halogen" or "halide" means fluoro, chloro, bromo or iodo.
[00195] The term "haloalkyl" refers to an alkyl group in which one or more hydrogen atoms are replaced by one or more halide atoms. In one aspect, a haloalkyl is a Ci-C6haloalkyl.
[00196] The term "fluoroalkyl" refers to a alkyl in which one or more hydrogen atoms are replaced by a fluorine atom.
In one aspect, a fluoralkyl is a Ci-C6fluoroalkyl.
[00197] The term "heteroalkyl" refers to an alkyl group in which one or more skeletal atoms of the alkyl are selected from an atom other than carbon, e.g. , oxygen, nitrogen, sulfur, phosphorus or combinations thereof. In one aspect, a heteroalkyl is a Ci-C6 heteroalkyl.
[00198] The term "heterocycle" or "heterocyclic" refers to heteroaromatic rings (also known as heteroaryls) and heterocycloalkyl rings (also known as heteroalicyclic groups) containing one to four heteroatoms in the ring(s), where each heteroatom in the ring(s) is selected from O, S and N, wherein each heterocyclic group has from 4 to 10 atoms in its ring system, and with the proviso that the any ring does not contain two adjacent O or S atoms. Non-aromatic heterocyclic groups (also known as heterocycloalkyls) include groups having only 3 atoms in their ring system, but aromatic heterocyclic groups must have at least 5 atoms in their ring system. The heterocyclic groups include benzo- fused ring systems. An example of a 3-membered heterocyclic group is aziridinyl. An example of a 4-membered heterocyclic group is azetidinyl. An example of a 5-membered heterocyclic group is thiazolyl. An example of a 6- membered heterocyclic group is pyridyl, and an example of a 10-membered heterocyclic group is quinolinyl. Examples of non-aromatic heterocyclic groups are pyrrolidinyl, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, oxazolidinonyl, tetrahydropyranyl, dihydropyranyl, tetrahydrothiopyranyl, piperidinyl, morpholinyl, thiomorpholinyl, thioxanyl, piperazinyl, aziridinyl, azetidinyl, oxetanyl, thietanyl, homopiperidinyl, oxepanyl, thiepanyl, oxazepinyl, diazepinyl, thiazepinyl, 1,2,3,6-tetrahydropyridinyl, pyrrolin-2-yl, pyrrolin-3-yl, indolinyl, 2H-pyranyl, 4H-pyranyl, dioxanyl, 1,3-dioxolanyl, pyrazolinyl, dithianyl, dithiolanyl, dihydropyranyl, dihydrothienyl, dihydrofuranyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, 3-azabicyclo[3.1.0]hexanyl, 3-azabicyclo[4.1.0]heptanyl, 3H-indolyl and quinolizinyl. Examples of aromatic heterocyclic groups are pyridinyl, imidazolyl, pyrimidinyl, pyrazolyl, triazolyl, pyrazinyl, tetrazolyl, furyl, thienyl, isoxazolyl, thiazolyl, oxazolyl, isothiazolyl, pyrrolyl, quinolinyl, isoquinolinyl, indolyl, benzimidazolyl, benzofuranyl, cinnolinyl, indazolyl, indolizinyl, phthalazinyl, pyridazinyl, triazinyl, isoindolyl, pteridinyl, purinyl, oxadiazolyl, thiadiazolyl, furazanyl, benzofurazanyl, benzothiophenyl, benzothiazolyl, benzoxazolyl, quinazolinyl, quinoxalinyl, naphthyridinyl, and furopyridinyl. The foregoing groups may be C-attached or TV-attached where such is possible. For instance, a group derived from pyrrole may be pyrrol- 1-yl (TV-attached) or pyrrol-3-yl (C-attached). Further, a group derived from imidazole may be imidazol-1-yl or imidazol-3-yl (both TV- attached) or imidazol-2-yl, imidazol-4-yl or imidazol-5-yl (all C-attached). The heterocyclic groups include benzo- fused ring systems. Non-aromatic heterocycles may be substituted with one or two oxo (=0) moieties, such as pyrrolidin-2-one.
[00199] The terms "heteroaryl" or, alternatively, "heteroaromatic" refers to an aryl group that includes one or more ring heteroatoms selected from nitrogen, oxygen and sulfur. Preferred heteroaryl groups include indole, azaindole, pyrrole, pyrazole, pyrimidine, pyrazine, pyridine, quinoline, thiophene and furan. In one aspect, a heteroaryl contains 0-3 N atoms. In another aspect, a heteroaryl contains 0-3 Natoms, 0-1 O atoms, and 0-1 S atoms. In another aspect, a heteroaryl is a monocyclic or bicyclic heteroaryl.
[00200] A "heterocycloalkyl", "heteroalicyclic" or "non-aromatic heterocycle" refers to a cycloalkyl group that includes at least one heteroatom selected from nitrogen, oxygen and sulfur. The radicals may be fused with an aryl or heteroaryl. In some embodiments, the heterocycloalkyl is selected from oxazolidinonyl, pyrrolidinyl, tetrahydrofuranyl, tetrahydrothienyl, tetrahydropyranyl, tetrahydrothiopyranyl, piperidinyl, morpholinyl, thiomorpholinyl, piperazinyl, and indolinyl. The term heteroalicyclic also includes all ring forms of the carbohydrates, including but not limited to the monosaccharides, the disaccharides and the oligosaccharides. In one aspect, a heterocycloalkyl is a C2- Cioheterocycloalkyl. In another aspect, a heterocycloalkyl is a C4-Ci oheterocycloalkyl. In one aspect, a heterocycloalkyl contains 0-2 N atoms. In another aspect, a heterocycloalkyl contains 0-2 N atoms, 0-2 O atoms or 0-1 S atoms.
[00201] The term "bond" or "single bond" refers to a chemical bond between two atoms, or two moieties when the atoms joined by the bond are considered to be part of larger substructure. In one aspect, when a group described herein is a bond, the referenced group is absent thereby allowing a bond to be formed between the remaining identified groups. [00202] The term "membered ring" includes any cyclic structure. The term "membered" is meant to denote the number of skeletal atoms that constitute the ring. Thus, for example, cyclohexyl, pyridinyl, pyranyl and thiopyranyl are 6-membered rings and cyclopentyl, pyrrolyl, furanyl, and thienyl are 5-membered rings. [00203] The term "moiety" refers to a specific segment or functional group of a molecule. Chemical moieties are often recognized chemical entities embedded in or appended to a molecule.
[00204] The term "optionally substituted" or "substituted" means that the referenced group may be substituted with one or more additional group(s) individually and independently selected from alkyl, cycloalkyl, aryl, heteroaryl, heteroalicyclic, hydroxy, alkoxy, aryloxy, alkylthio, arylthio, alkylsulfoxide, arylsulfoxide, alkylsulfone, arylsulfone, cyano, halo, carbonyl, thiocarbonyl, nitro, haloalkyl, fluoroalkyl, and amino, including mono- and di-substituted amino groups, and the protected derivatives thereof. By way of example an optional substituents may be halide, -CN, -NO2, or
L8R8, wherein each L8 is independently selected from a bond, -O-, -C(=O)-, -C(=0)O, -S-, -S(=O)-, -S(=O)2-, -NH-, -
NHC(=O)-, -CC=O)NH-, SC=O)2NH-, -NHS(=O)2, -OC(=O)NH-, -NHC(=O)O-, or -(C1-C6 alkyl)-; and each R8 is selected from H, alkyl, fluoroalkyl, heteroalkyl, cycloalkyl, aryl, heteroaryl, or heterocycloalkyl. The protecting groups that may form the protective derivatives of the above substituents may be found in sources such as Greene and Wuts, above. In one aspect, optional substituents are selected from halogen, CF3, OH, CN, NO2, SO3H, SO2NH2, SO2Me,
NH2, COOH, CONH2, alkoxy, -N(CH3)2, and alkyl.
[00205] In certain embodiments, the compounds presented herein possess one or more stereocenters and each center independently exists in either the R or S configuration. The compounds presented herein include all diastereomeric, enantiomeric, and epimeric forms as well as the appropriate mixtures thereof. Stereoisomers are obtained, if desired, by methods such as, the separation of stereoisomers by chiral chromatographic columns.
[00206] The methods and formulations described herein include the use of TV-oxides (if appropriate), crystalline forms
(also known as polymorphs), or pharmaceutically acceptable salts of compounds having the structure of Formula (I), as well as active metabolites of these compounds having the same type of activity. In some situations, compounds may exist as tautomers. All tautomers are included within the scope of the compounds presented herein. In specific embodiments, the compounds described herein exist in solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like. In other embodiments, the compounds described herein exist in unsolvated form.
Certain Pharmaceutical and Medical Terminology
[00207] The term "acceptable" with respect to a formulation, composition or ingredient, as used herein, means having no persistent detrimental effect on the general health of the subject being treated.
[00208] The term "kinase-dependent", as used herein, refers to conditions or disorders that would not occur, or would not occur to the same extent, in the absence of a kinase enzyme.
[00209] The term "kinase-mediated", as used herein, refers to refers to conditions or disorders that might occur in the absence of kinase enzyme but can occur in the presence of kinase enzyme.
[00210] The term "asthma" as used herein refers to any disorder of the lungs characterized by variations in pulmonary gas flow associated with airway constriction of whatever cause (intrinsic, extrinsic, or both; allergic or non-allergic).
The term asthma may be used with one or more adjectives to indicate cause.
[00211] The term "bone disease,' as used herein, refers to a disease or condition of the bone, including, but not limited to, inappropriate bone remodeling, loss or gain, osteopenia, osteomalacia, osteofibrosis, and Paget' s disease.
[00212] The term "cardiovascular disease," as used herein refers to diseases affecting the heart or blood vessels or both, including but not limited to: arrhythmia (atrial or ventricular or both); atherosclerosis and its sequelae; angina; cardiac rhythm disturbances; myocardial ischemia; myocardial infarction; cardiac or vascular aneurysm; vasculitis, stroke; peripheral obstructive arteriopathy of a limb, an organ, or a tissue; reperfusion injury following ischemia of the brain, heart or other organ or tissue; endotoxic, surgical, or traumaticshock; hypertension, valvular heart disease, heart failure, abnormal blood pressure; shock; vasoconstriction (including that associated with migraines); vascular abnormality, inflammation, insufficiency limited to a single organ or tissue.. [00213] The term "cancer," as used herein refers to an abnormal growth of cells which tend to proliferate in an uncontrolled way and, in some cases, to metastasize (spread). The types of cancer include, but are not limited to, solid tumors (such as those of the bladder, bowel, brain, breast, endometrium, heart, kidney, lung, lymhatic tissue (lymphoma), ovary, pancreas or other endocrine organ (thyroid), prostate, skin (melanoma) or hematological tumors (such as the leukemias).
[00214] The terms "co-administration" or the like, as used herein, are meant to encompass administration of the selected therapeutic agents to a single patient, and are intended to include treatment regimens in which the agents are administered by the same or different route of administration or at the same or different time. [00215] The term "dermatological disorder," as used herein refers to a skin disorder. Such dermatological disorders include, but are not limited to, proliferative or inflammatory disorders of the skin such as, atopic dermatitis, bullous disorders, collagenoses, contact dermatitis eczema, Kawasaki Disease, rosacea, Sjogren-Larsso Syndrome, urticaria. [00216] The terms "enhance" or "enhancing," as used herein, means to increase or prolong either in potency or duration a desired effect. Thus, in regard to enhancing the effect of therapeutic agents, the term "enhancing" refers to the ability to increase or prolong, either in potency or duration, the effect of other therapeutic agents on a system. An "enhancing-effective amount," as used herein, refers to an amount adequate to enhance the effect of another therapeutic agent in a desired system.
[00217] The term "iatrogenic" means a condition, disorder, or disease created or worsened by medical or surgical therapy.
[00218] The term "inflammatory disorders" refers to those diseases or conditions that are characterized by one or more of the signs of pain, heat, redness, swelling, and loss of function (temporary or permanent). Inflammation takes many forms and includes, but is not limited to, inflammation that is one or more of the following: acute, adhesive, atrophic, catarrhal, chronic, cirrhotic, diffuse, disseminated, exudative, fibrinous, fibrosing, focal, granulomatous, hyperplastic, hypertrophic, interstitial, metastatic, necrotic, obliterative, parenchymatous, plastic, productive, proliferous, pseudomembranous, purulent, sclerosing, seroplastic, serous, simple, specific, subacute, suppurative, toxic, traumatic, and/or ulcerative. Inflammatory disorders further include, without being limited to those affecting the blood vessels (polyarteritis, temporal arteritis); joints (arthritis: crystalline, osteo-, psoriatic, reactive, rheumatoid, Reiter's); gastrointestinal tract (colitis); skin (dermatitis); or multiple organs and tissues (systemic lupus erythematosus). [00219] The term "immunological disorders" refers to those diseases or conditions that are characterized by inappropriate or deleterious response to an endogenous or exogenous antigen that may result in cellular dysfunction or destruction and consequently dysfunction or destruction of an organ or tissue and which may or may not be accompanied by signs or symptoms of inflammation. [00220] The terms "kit" and "article of manufacture" are used as synonyms.
[00221] A "metabolite" of a compound disclosed herein is a derivative of that compound that is formed when the compound is metabolized. The term "active metabolite" refers to a biologically active derivative of a compound that is formed when the compound is metabolized. The term "metabolized," as used herein, refers to the sum of the processes (including, but not limited to, hydrolysis reactions and reactions catalyzed by enzymes) by which a particular substance is changed by an organism. Thus, enzymes may produce specific structural alterations to a compound. For example, cytochrome P450 catalyzes a variety of oxidative and reductive reactions while uridine diphosphate glucuronyltransferases catalyze the transfer of an activated glucuronic-acid molecule to aromatic alcohols, aliphatic alcohols, carboxylic acids, amines and free sulphydryl groups. Metabolites of the compounds disclosed herein are optionally identified either by administration of compounds to a host and analysis of tissue samples from the host, or by incubation of compounds with hepatic cells in vitro and analysis of the resulting compounds. [00222] The terms "neurogenerative disease" or "nervous system disorder," as used herein, refers to conditions that alter the structure or function of the brain, spinal cord or peripheral nervous system, including but not limited to Alzheimer's Disease, cerebral edema, cerebral ischemia, multiple sclerosis, neuropathies, Parkinson's Disease, those found after blunt or surgical trauma (including post-surgical cognitive dysfunction and spinal cord or brain stem injury), as well as the neurological aspects of disorders such as degenerative disk disease and sciatica. The acronym "CNS" refers to disorders of the central nervous system, i.e., brain and spinal cord.
[00223] The term "subject" or "patient" encompasses mammals and non-mammals. Examples of mammals include, but are not limited to, any member of the Mammalian class: humans, non-human primates such as chimpanzees, and other apes and monkey species; farm animals such as cattle, horses, sheep, goats, swine; domestic animals such as rabbits, dogs, and cats; laboratory animals including rodents, such as rats, mice and guinea pigs, and the like. Examples of non-mammals include, but are not limited to, birds, fish and the like. In one embodiment, the mammal is a human. [00224] The terms "treat," "treating" or "treatment," as used herein, include alleviating, abating or ameliorating a disease or condition symptoms, preventing additional symptoms, ameliorating or preventing the underlying metabolic causes of symptoms, inhibiting the disease or condition, e.g., arresting the development of the disease or condition, relieving the disease or condition, causing regression of the disease or condition, relieving a condition caused by the disease or condition, or stopping the symptoms of the disease or condition either prophylactically and/or therapeutically.
[00225] As used herein, amelioration of the symptoms of a particular disease, disorder or condition by administration of a particular compound or pharmaceutical composition refers to any lessening of severity, delay in onset, slowing of progression, or shortening of duration, whether permanent or temporary, lasting or transient that can be attributed to or associated with administration of the compound or composition. THERAPEUTIC USE
[00226] In one aspect, the compounds of Formula (I) inhibit one or more protein kinase(s). In one aspect, the compounds of Formula (I) demonstrate anti-proliferative activity and are useful in the treatment of proliferative disorders. In one aspect, the compounds of Formula (I) demonstrate anti-proliferative activity and are useful in the treatment of proliferative disorders such as cancers, leukaemias and other disorders associated with uncontrolled cellular proliferation such as psoriasis and restenosis.
[00227] In one aspect, an anti-proliferative effect within the scope of the present disclosure is the ability to inhibit cell proliferation in an in vitro whole cell assay. In one aspect, the in vitro whole cell assay uses any one of the cell lines selected from A549, A2780, HepG2, HCT-116, HeLa, HT29, MCF-7, NCI-H460, and Saos-2. In one aspect, an antiproliferative effect within the scope of the present disclosure is the ability to inhibit at least one CDK enzyme, such as CDKl, CDK2, CDK4, CDK5, CDK6, CDK7, CDK8, CDK9, CDKIl, or other protein kinases; or inhibition of the interaction between HDM2 and p53 in an appropriate assay. These assays, including methods for their performance, are known in the art and/or are described in more detail in the accompanying Examples.
[00228] In one aspect, the compounds of Formula (I) are used in the treatment of proliferative disorders. In a further aspect, there is provided the use of one or more compounds of Formula (I) in the manufacture of a medicament for treating proliferative disorders. In one aspect, the proliferative disorder is a cancer or leukaemia. The term proliferative disorder is used herein in a broad sense to include any disorder that requires control of the cell cycle, for example cardiovascular disorders, auto-immune disorders, dermatological disorders, inflammatory, fungal, parasitic disorders, emphysema and alopecia. In one aspect, the compounds of Formula (I) induce apoptosis or maintain stasis in these disorders within the appropriate cells as required. [00229] In one aspect, cardiovascular disorders include, but are not limited to, restenosis and cardiomyopathy. In one aspect, auto-immune disorders include, but are not limited to, glomerulonephritis and rheumatoid arthritis. In one aspect, dermatological disorders include, but are not limited to, psoriasis. In one aspect, parasitic disorders include, but are not limited to, malaria.
[00230] In one aspect, the compounds of Formula (I) are used in the treatment of a disease, disorder, or condition mediated by one or more protein kinase selected from a CDK, aurora kinase, GSK, PLK and one of Tyrosine kinases. In one aspect, the compounds of Formula (I) inhibit such protein kinases.
[00231] In one aspect, the compounds of Formula (I) inhibit any of the steps or stages in the cell cycle, for example, formation of the nuclear envelope, exit from the quiescent phase of the cell cycle (GO), Gl progression, chromosome decondensation, nuclear envelope breakdown, START, initiation of DNA replication, progression of DNA replication, termination of DNA replication, centrosome duplication, G2 progression, activation of mitotic or meiotic functions, chromosome condensation, centrosome separation, microtubule nucleation, spindle formation and function, interactions with microtubule motor proteins, chromatid separation and segregation, inactivation of mitotic functions, formation of contractile ring, and cytokinesis functions. In particular, the compounds of Formula (I) influence certain gene functions such as chromatin binding, formation of replication complexes, replication licensing, phosphorylation or other secondary modification activity, proteolytic degradation, microtubule binding, actin binding, septin binding, microtubule organising centre nucleation activity and binding to components of cell cycle signalling pathways. [00232] In one embodiment, compounds of Formula (I) are useful in the treatment of a proliferative disorder, disease or condition mediated by at least one of a CDK, Auora kinase and/or PLK. In one aspect, the proliferative disorder, disease or condition mediated by at least one of a CDK, Auora kinase and/or PLK include, but are not limited to cancers, leukaemias and other disorders associated with uncontrolled cellular proliferation such as psoriasis and restenosis, a viral disorder, a cardiovascular disease, a CNS disorder, an autoimmune disease, a bone disease, a hormone-related disease, a metabolic disorder, stroke, alopecia, an inflammatory disease or an infectious disease. [00233] In one aspect, the compounds of Formula (I) inhibit one or more of the host cell kinases involved in cell proliferation, viral replication, a cardiovascular disorder, neurodegeneration, autoimmunity, a metabolic disorder, stroke, alopecia, an inflammatory disease or an infectious disease.
[00234] A proliferative disorder requires treatment of a susceptible neoplasm and may be selected from the group consisting of chronic lymphocytic leukaemia, lymphoma, leukaemia, breast cancer, lung cancer, prostate cancer, colon cancer, melanoma, pancreatic cancer, ovarian cancer, squamous carcinoma, carcinoma of head and neck, endometrial cancer, and aesophageal carcinoma.
[00235] In one aspect, the compounds, compositions and methods provided herein are useful for the treatment and prevention of cancer including solid tumors such as skin, breast, brain, cervical carcinomas, testicular carcinomas, etc. In one aspect, compounds described herein are useful for treating cancer. In one aspect, cancers include, but are not limited to:
[00236] Cardiac cancers, such as, but not limited to, sarcoma (angiosarcoma, fibrosarcoma, rhabdomyosarcoma, liposarcoma), myxoma, rhabdomyoma, fibroma, lipoma and teratoma;
[00237] Lung cancers, such as, but not limited to, bronchogenic carcinoma (squamous cell, undifferentiated small cell, undifferentiated large cell, non-small cell, adenocarcinoma), alveolar (bronchiolar) carcinoma, bronchial adenoma, sarcoma, lymphoma, chondromatous hamartoma, mesothelioma;
[00238] Gastrointestinal cancers, such as, but not limited to: esophagus (squamous cell carcinoma, adenocarcinoma, leiomyosarcoma, lymphoma), stomach (carcinoma, lymphoma, leiomyosarcoma), pancreas (ductal adenocarcinoma, insulinoma, glucagonoma, gastrinoma, carcinoid tumors, vipoma), small bowel (adenocarcinoma, lymphoma, carcinoid tumors, Karposi's sarcoma, leiomyoma, hemangioma, lipoma, neurofibroma, fibroma), large bowel (adenocarcinoma, tubular adenoma, villous adenoma, hamartoma, leiomyoma), rectal, colorectal and colon; [00239] Genitourinary tract cancers, such as, but not limited to: kidney (adenocarcinoma, Wilm's tumor [nephroblastoma], lymphoma, leukemia, papillary renal carcinoma), bladder and urethra (squamous cell carcinoma, transitional cell carcinoma, adenocarcinoma), prostate (adenocarcinoma, sarcoma), testis (seminoma, teratoma, embryonal carcinoma, teratocarcinoma, choriocarcinoma, sarcoma, interstitial cell carcinoma, fibroma, fibroadenoma, adenomatoid tumors, lipoma);
[00240] Liver cancers, such as, but not limited to: hepatoma (hepatocellular carcinoma), cholangiocarcinoma, hepatoblastoma, angiosarcoma, hepatocellular adenoma, hemangioma;
[00241] Bone cancers, such as, but not limited to: osteogenic sarcoma (osteosarcoma), fibrosarcoma, malignant fibrous histiocytoma, chondrosarcoma, Ewing's sarcoma, malignant lymphoma (reticulum cell sarcoma), multiple myeloma, malignant giant cell tumor chordoma, osteochronfroma (osteocartilaginous exostoses), benign chondroma, chondroblastoma, chondromyxofibroma, osteoid osteoma and giant cell tumors;
[00242] Nervous system cancers, such as, but not limited to: skull (osteoma, hemangioma, granuloma, xanthoma, osteitis deformans), meninges (meningioma, meningiosarcoma, gliomatosis), brain (astrocytoma, medulloblastoma, glioma, ependymoma, germinoma [pinealoma], glioblastoma multiform, oligodendroglioma, schwannoma, retinoblastoma, congenital tumors), spinal cord neurofibroma, meningioma, glioma, sarcoma);
[00243] Gynecological cancers, such as, but not limited to: uterus (endometrial carcinoma), cervix (cervical carcinoma, pre-tumor cervical dysplasia), ovaries (ovarian carcinoma [serous cystadenocarcinoma, mucinous cystadenocarcinoma, unclassified carcinoma], granulosa-thecal cell tumors, Sertoli-Leydig cell tumors, dysgerminoma, malignant teratoma), vulva (squamous cell carcinoma, intraepithelial carcinoma, adenocarcinoma, fibrosarcoma, melanoma), vagina (clear cell carcinoma, squamous cell carcinoma, botryoid sarcoma (embryonal rhabdomyosarcoma), fallopian tubes (carcinoma);
[00244] Hematologic cancers, such as, but not limited to: blood (myeloid leukemia [acute and chronic], acute lymphoblastic leukemia, chronic lymphocytic leukemia, myeloproliferative diseases, multiple myeloma, myelodysplastic syndrome), Hodgkin's disease, non-Hodgkin's lymphoma [malignant lymphoma]; [00245] Skin cancers, such as, but not limited to: malignant melanoma, basal cell carcinoma, squamous cell carcinoma, head and neck squamous cell carcinomas, Karposi's sarcoma, moles dysplastic nevi, lipoma, angioma, dermatofibroma, keloids, psoriasis; and Adrenal glands: neuroblastoma.
[00246] The term "cancerous cell" as provided herein, includes a cell afflicted by any one of the above-identified conditions.
[00247] In another embodiment, compound described herein are useful for treating or preventing cancer selected from: head and neck squamous cell carcinomas, histiocytic lymphoma, lung adenocarcinoma, small cell lung cancer, non- small cell lung cancer, pancreatic cancer, papillary renal carcinoma, liver cancer, gastric cancer, colon cancer, multiple myeloma, glioblastomas and breast carcinoma.
[00248] In another embodiment, compounds described herein are useful for the prevention or modulation of the metastases of cancer cells and cancer. In yet another embodiment, compounds described herein are useful to prevent or modulate the metastases of ovarian cancer, childhood hepatocellular carcinoma, metastatic head and neck squamous cell carcinomas, gastric cancers, breast cancer, colorectal cancer, cervical cancer, lung cancer, nasopharyngeal cancer, pancreatic cancer, glioblastoma and sarcomas.
[00249] A further embodiment relates to the use of compounds of Formula (I) described herein, or pharmaceutically acceptable salts thereof, in the manufacture of a medicament capable of treating a viral disorder mediated by one or more of the host cell CDKs involved in viral replication. In one aspect, the one or more CDKs are selected from among CDKl, CDK2, CDK4, CDK7, CDK8, CDK9 and CDKI l.
[00250] Assays for determining CDK activity are described in more detail in the accompanying examples. Using such enzymes assays it may be determined whether a compound is anti-viral in the context of the present invention.
[00251] In one aspect, compounds of Formula (I),or pharmaceutical compositions comprising compounds of Formula
(I) are used in the treatment of viral disorders. In one aspect, the viral disorder is selected from among human cytomegalovirus (HCMV), herpes simplex virus type 1 (HSV-I), human immunodeficiency virus type 1 (HIV-I), and varicella zoster virus (VZV). Typically such disorder is CDK dependent or sensitive. CDK dependent disorders are associated with an above normal level of activity of one or more CDK enzymes. In one aspect, such disorders are typically associated with an abnormal level of activity of CDKl, CDK2, CDK4, CDK6, CDK7, CDK8, CDK9 and/or
CDKI l.
[00252] A CDK sensitive disorder is a disorder in which an aberration in the CDK level is not the primary cause, but is downstream of the primary metabolic aberration. In such scenarios, CDKl, CDK2, CDK4, CDK6, CDK7, CDK8
CDK9 and/or CDKl 1 can be said to be part of the sensitive metabolic pathway and inhibitors of one or more of these
CDKs are therefore useful in treating such disorders.
[00253] In one aspect, for use in the treatment of viral disorders, the compound of Formula (I) is capable of inhibiting one or more CDKs selected from among CDK2, CDK7, and CDK9.
[00254] In another aspect, compounds of Formula (I) are used in the treatment of cardiovascular diseases mediated by one or more CDKs. A cardiovascular disease may be selected from the group consisting of ischaemic heart disease
(also known as myocardial infarction or angina), hypertension, heart failure, restenosis and cardiomyopathy.
[00255] Cardiac hypertrophy is characterized by global increases in mRNA and protein synthesis. CDK9 activity has been demonstrated to be necessary for hypertrophy in cardiomyocytes. Heart-specific activation of CDK9 by cyclin Tl was found to provoke hypertrophy. In one aspect, compounds of Formula (I) inhibit one or more CDKs and are therefore useful in the prevention and treatment of cardiac hypertrophy.
[00256] Yet another embodiment relates to the use of a compound of Formula (I) in the treatment of neurodegenerative disorders mediated by one or more GSKs or CDKs. In one aspect, the neurodegenerative disorder is
Alzheimer's disease.
[00257] The appearances of Paired Helical Filaments, associated with Alzeimer's disease, are caused by the hyperphosphorylation of Tau protein by CDK5-p25. In one aspect, compounds of Formula (I) inhibit CDK5 and are useful in the prevention and treatment of neurodegenerative disorders.
[00258] Tau is a GSK-3 substrate which has been implicated in the etiology of Alzheimer's disease. In healthy nerve cells, Tau co-assembles with tubulin into microtubules. However, in Alzheimer's disease, tau forms large tangles of filaments, which disrupt the microtubule structures in the nerve cell, thereby impairing the transport of nutrients as well as the transmission of neuronal messages. In one aspect, GSK3 inhibitors prevent and/or reverse the abnormal hyperphosphorylation of the microtubule-associated protein tau that is an invariant feature of Alzheimer's disease and a number of other neurodegenerative diseases, such as progressive supranuclear palsy, corticobasal degeneration and
Pick's disease. Mutations in the tau gene cause inherited forms of fronto-temporal dementia, further underscoring the relevance of tau protein dysfunction for the neurodegenerative process.
[00259] In one aspect, compounds of Formula (I) are used in the treatment a metabolic disorder mediated by one or more GSKs. Metabolic disorders include Type II diabetes (non insulin dependent diabetes mellitus) and diabetic neuropathy. In one aspect, compounds of Formula (I) inhibit GSK-3, which is implicated in Type II diabetes. [00260] GSK3 is one of several protein kinases that phosphorylate glycogen synthase (GS) and is involved in the stimulation of glycogen synthesis by insulin in skeletal muscle. GSK3's action on GS thus results in the latter' s deactivation and thus suppression of the conversion of glucose into glycogen in muscles. Type II diabetes (non-insulin dependent diabetes mellitus) is a multi-factorial disease. Hyperglycaemia is due to insulin resistance in the liver, muscles, and other tissues, coupled with impaired secretion of insulin. Skeletal muscle is the main site for insulin- stimulated glucose uptake, there it is either removed from circulation or converted to glycogen. Muscle glycogen deposition is the main determinant in glucose homeostasis and type II diabetics have defective muscle glycogen storage. There is evidence that an increase in GSK3 activity is important in type II diabetes.
[00261] In one aspect, compounds of Formula (I) are used in the treatment of bipolar disorder mediated by one or more kinases.
[00262] In another aspect, compounds of Formula (I) are used for treating a stroke mediated by one or more GSKs. [00263] Reducing neuronal apoptosis is an important therapeutic goal in the context of head trauma, stroke, epilepsy, and motor neuron disease. GSK3 as a pro-apoptotic factor in neuronal cells makes this protein kinase an attractive therapeutic target for the design of inhibitory drugs to treat these diseases.
[00264] In one aspect, compounds of Formula (I) are used in the treatment of alopecia mediated by one or more GSKs. [00265] In one aspect, the ectopic application of GSK3 inhibitors is therapeutically useful in the treatment of baldness and in restoring hair growth following chemotherapy-induced alopecia.
[00266] In one aspect, described herein is a method of treating a disease, disorder and/or condition mediated by one or more enzymes selected from a CDK, aurora kinase, GSK, PLK or tyrosine kinase enzyme as hereinbefore described. [00267] In one aspect, such condition is a GSK3 -dependent disorder, said method comprising administering to a subject in need thereof, a compound of Formula (I) in an amount sufficient to inhibit GSK3. In one aspect, the compound of Formula (I) is administered in an amount sufficient to inhibit GSK3β.
[00268] In one aspect, described herein is a method of treating a PLK-dependent disease, disorder and/or condition, said method comprising administering to a subject in need thereof, a compound of the Formula (I) in an amount sufficient to inhibit a PLK. In one aspect, human PLKs regulate fundamental aspects of mitosis. Both PLKl and PLK2 may have additional post-mitotic functions. Deregulated PLK expressions result in cell cycle arrest and apoptosis. Compounds of Formula (I) are used in treating PLK-mediated diseases, disorders and/or conditions. In one aspect, the PLK-mediated diseases, disorders and/or conditions include proliferative disorders. [00269] In one aspect, the compounds of Formula (I) inhibit one or more PLK enzymes selected from PLKl, PLK2, PLK3 and PLK4
[00270] In another embodiment, described herein is a method of treating an aurora kinase-dependent disease, disorder and/or condition, said method comprising administering to a subject in need thereof, a compound of Formula (I). [00271] In one aspect, the compound of Formula (I) inhibits one or more aurora kinases selected from among aurora kinase A, aurora kinase B or aurora kinase C.
[00272] In another embodiment, described herein is a method of treating a tyrosine kinase-dependent disorder, said method comprising administering to a subject in need thereof, a compound of Formula (I) in an amount sufficient to inhibit a tyrosine kinase.
[00273] In one aspect, compounds of Formula (I) are administered to a subject in an amount sufficient to inhibit at least one protein kinase selected from BCR-ABL, IKK, FLT3, JAK, LCK, PDGF, Src, and VEGF. [00274] In some embodiments, described herein is a method of selectively treating a protein kinase-dependent disorder, said method comprising administering to a subject in need thereof, a compound of the invention or a pharmaceutically acceptable salt thereof, as defined above in an amount sufficient to inhibit a selected protein kinase. In one aspect, said method comprises contacting said protein kinase with a compound described herein.
[00275] In some embodiments, the compound of Formula (I) is administered in an amount sufficient to inhibit at least one of a CDK, GSK, aurora kinase, or PLK, or a tyrosine kinase including, but not limiting to BCR-ABL, IKK, FLT3,
JAK, LCK, PDGF, Src, or VEGF.
[00276] In some embodiments, the protein kinase is a CDK. In one aspect, the protein kinase is CDKl, CDK2, CDK3,
CDK4, CDK5, CDK6, CDK7, CDK8, CDK9 or CDKl 1. In another aspect, the protein kinase is CDKl, CDK2, CDK7 or CDK9.
[00277] In one aspect, the compound of Formula (I) inhibits at least one CDK enzyme, preferably at least one of
CDKl, CDK2, CDK7 and CDK9.
[00278] In one aspect, the compound of Formula (I) inhibits a CDK. In one aspect, the compound of Formula (I) inhibits a CDK selected from CDKl, CDK2, CDK7 and CDK9 at sub-micromolar IC50 values, more preferably at IC50 of less than 0.5 micromolar, more preferably less than 0.25 micromolar. In one aspect, the compound of Formula (I) inhibits CDK9 at IC50 of less than 0.1 micromolar or less than 0.05 micromolar.
[00279] In some embodiments, the compound of Formula (I) demonstrates an antiproliferative effect in human cell lines, as measured by a standard 72h MTT cytotoxicity assay. In some embodiments, the compound of Formula (I) exhibits an IC50 value of less than 1 micromolar.
[00280] In a further aspect, there is provided a method of treating a proliferative disease or disorder, a viral disorder, a cardiovascular disease, a CNS disorder, an autoimmune disease, a metabolic disorder, stroke, alopecia, an inflammatory disease or an infectious disease, said method comprising administering to a subject in need thereof, a compound of Formula (I) as hereinbefore defined in an effective amount.
[00281] The use of a compound of the invention in the manufacture of a medicament as hereinbefore defined includes the use of the compound directly, or in any stage of the manufacture of such a medicament, or in vitro in a screening programme to identify further agents for the prevention or treatment of the hereinbefore defined diseases or conditions.
[00282] A further aspect relates to the use of a compound of Formula (I) or a pharmaceutically acceptable salt or solvate or physiologically hydrolysable, solubilising or immobilising derivative thereof, in an assay for identifying candidate compounds capable of treating one or more disorders or diseases as hereinbefore defined. Preferably a compound is of use in identifying candidate compounds capable of inhibiting a protein kinase, more preferably one or more of a CDK, aurora kinase, GSK, PLK or tyrosine kinase enzyme.
Routes of Administration
[00283] Suitable routes of administration include, but are not limited to, oral, intravenous, rectal, aerosol, parenteral, ophthalmic, pulmonary, transmucosal, transdermal, vaginal, otic, nasal, and topical administration. In addition, by way of example only, parenteral delivery includes intramuscular, subcutaneous, intravenous, intramedullary injections, as well as intrathecal, direct intraventricular, intraperitoneal, intralymphatic, and intranasal injections.
[00284] In certain embodiments, a compound as described herein is administered in a local rather than systemic manner, for example, via injection of the compound directly into an organ, often in a depot preparation or sustained release formulation. In specific embodiments, long acting formulations are administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection. Furthermore, in other embodiments, the drug is delivered in a targeted drug delivery system, for example, in a liposome coated with organ-specific antibody. In such embodiments, the liposomes are targeted to and taken up selectively by the organ. In yet other embodiments, the compound as described herein is provided in the form of a rapid release formulation, in the form of an extended release formulation, or in the form of an intermediate release formulation. In yet other embodiments, the compound described herein is administered topically.
Kits/Articles of Manufacture
[00285] For use in the therapeutic applications described herein, kits and articles of manufacture are also described herein. Such kits can comprise a carrier, package, or container that is compartmentalized to receive one or more containers such as vials, tubes, and the like, each of the container(s) comprising one of the separate elements to be used in a method described herein. Suitable containers include, for example, bottles, vials, syringes, and test tubes. The containers are formed from any acceptable material including, e.g., glass or plastic.
[00286] For example, the container(s) can comprise one or more compounds described herein, optionally in a composition or in combination with another agent as disclosed herein. The container(s) optionally have a sterile access port (for example the container can be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle). Such kits optionally comprising a compound with an identifying description or label or instructions relating to its use in the methods described herein.
[00287] A kit will typically comprise one or more additional containers, each with one or more of various materials
(such as reagents, optionally in concentrated form, and/or devices) desirable from a commercial and user standpoint for use of a compound described herein. Non-limiting examples of such materials include, but not limited to, buffers, diluents, filters, needles, syringes; carrier, package, container, vial and/or tube labels listing contents and/or instructions for use, and package inserts with instructions for use. A set of instructions will also typically be included.
[00288] A label can be on or associated with the container. A label can be on a container when letters, numbers or other characters forming the label are attached, molded or etched into the container itself; a label can be associated with a container when it is present within a receptacle or carrier that also holds the container, e.g., as a package insert. A label can be used to indicate that the contents are to be used for a specific therapeutic application. The label can also indicate directions for use of the contents, such as in the methods described herein.
[00289] All of the features disclosed in this specification (including any accompanying claims, abstract and drawings), and/or all of the steps of any method or process so disclosed, may be combined in any combination, except combinations where at least some of such features and/or steps are mutually exclusive.
EXAMPLES
[00290] These examples are provided for illustrative purposes only and not to limit the scope of the claims provided herein.
[00291] General. 1H-NMR spectra were obtained using a Bruker-400 spectrometer. Chemical shifts are reported in parts per million relative to internal tetramethylsilane standard. Coupling constants (J) are quoted to the nearest 0.1 Hz. The following abbreviations are used: s, singlet; d, doublet; t, triplet; q, quartet; qu, quintuplet; m, multiplet and br, broad. Mass spectra were obtained using a Waters 2795 single quadrupole mass spectrometer with electrospray ionization (ESI). Microwave assisted chemistry was carried out using CEM Discovery model (Biotage Ltd. UK). TLC (thin-layer chromatography) was performed using alumina plates coated with silica gel G60. Developed plates were air dried and analyzed under a UV lamp (254 / 365 run). Silica gel (EM Kieselgel 60, 0.040-0.063 mm, Merck) or ISOLUTE pre-packed columns was used for flash chromatography.
EXAMPLE 1 : 3-(4-(l-Methyl-lff-indol-3-yl)pyrimidin-2-ylamino)phenol (Compound 1)
Figure imgf000042_0001
[00292] A mixture of 3-acetylindole (20 mmol) and 7V,7V,-dimethylformamide demethylacetal (65 mmol) was heated to reflux for 48 hours. The excess amount of 7V,7V,-dimethyrformamide demethylacetal was removed in vacuo. The oil residue was precipitated by the addition of a minima amount of EtOAc. The solid was filtered and washed with EtOAc/PE to give (E)-3-(dimethylamino)-l -(I -methyl- lH-indol-3-yl)prop-2-en-l -one as orange powder. 1H-NMR (DMSOd6) δ: 2.97 (s, 6H, CH3x2), 3.83 (s, 3H, CH3), 2.85 (s, 3H, CH3), 5.71 (d, IH, J = 12.8 Hz, CH), 7.14 (m, IH, Ph-H), 7.21 (m, IH, Ar-H), 7.46 (d, IH, J = 8.4 Hz, Ar-H), 7.53 (d, IH, J= 12.4Hz, CH), 8.15 (s, IH, Ar-H), 8.28 (d, IH, J= 8.0Hz, Ar-H). MS (ESI+) m/z 229.3037 (M+H)+.
[00293] To a mixture of (E)-3-(dimethylamino)-l-(l-methyl-l/f-indol-3-yl)prop-2-en-l-one (l.Ommol, 0.2Ig) in MeCN (3.5 mL) was added 1.0 mmol l-(3-hydroxyphenyl)guanidine hydrogen chloride salt in the presence of NaOH (l.Ommol). The mixture was irradiated at 1500C for 20 minutes in microwave. The mixture was purified by chromatography using EtOAc/PE (1: 1, v/v) to elute the titled product as a off-white solid. 1H-NMR (DMSO-J6) δ: 3.89 (s, 3H, CH3), 6.38 (m IH, Ph-H), 7.07 (m, IH, Ph-H), 7.19 (d, IH, J= 5.6Hz, Pyrimidin-H), 7.25 (m, IH, Ar-H), 7.38 (s, IH, Ph-H), 7.53 (d, IH, J= 8.0Hz, Ph-H), 8.32 (m, 2H, Ar-H and Pyrimidin-H), 8.64 (d, IH, J= 8.0 Hz, OH), 9.23 (s, IH, Ar-H), 9.27 (s, IH, Ar-H). MS (ESI+) m/z 317.1311 (M+H)+ [00294] The following compounds were prepared in an analogous manner: EXAMPLE 2 : 4-(4-(l-Methyl-lff-indol-3-yl)pyrimidin-2-ylamino)phenol (Compound 2)
Figure imgf000043_0001
[00295] The compound was obtained by treatment of (E)-3-(dimethylamino)-l -(I -methyl- l/f-indol-3-yl)prop-2 -en- 1- one with l-(4-hydroxyphenyl)guanidine hydrochloride as white solid; 1H-NMR (DMSO-J6) δ: 3.88 (s, 3H, CH3), 6.74 (d, IH, J= 8.8 Hz, Ph-H), 7.10 (d, 2H, J= 5.6Hz, Pyrimidin-H), 7.17 (t, IH, J= 7.2 Hz, Ar-H), 7.28 (t, IH, J = 7.2 Hz, Ar-H), 7.53 (m, 3H, Ar-H), 8.26 (m, 2H, Ar-H and Pyrimidin-H), 8.58 (d, IH, J= 7.6 Hz, OH), 9.03 (s, 2H, Ph-H). MS (ESI+) m/z 317.1351 (M+H)+ EXAMPLE 3 : 3-Chloro-4-(4-(l-methyl-l//-indol-3-yl)pyrimidin-2-ylamino)phenol (Compound 3)
Figure imgf000043_0002
[00296] Obtained by treatment of (E)-3 -(dimethylamino)- 1 -( 1 -methyl- l/f-indol-3 -yl)prop-2-en- 1 -one with 1 -(2- chloro-4-hydroxyphenyl)guanidine hydrochloride as white solid; 1H-NMR (DMSO-J6) δ: 3.85 (s, 3H, CH3), 6.80 (dd, IH, J= 2.8, 8.4 Hz, Ph-H), 6.93 (d, 2H, J= 2.8 Hz, Ar-H), 7.04 (t, IH, J= 7.2 Hz, Ar-H), 7.09 (d, IH J= 5.2 Hz, Pyrimidin-H), 7.22 (t, IH, J= 7.2 Hz, Ar-H), 7.45 (d, IH, J= 8.8 Hz, Ph-H), 7.48 (d, IH, J= 8.4 Hz, Ar-H), 8.20 (d, IH, J= 5.2 Hz, Pyrimidin-H), 8.23 (s, IH, Ph-H), 8.26 (d, IH, J= 8.0 Hz, OH), 8.48 (s, IH, Ar-H), 9.74 (s, IH, NH). MS (ESI+) m/z 351.1048 (M+H)+ EXAMPLE 4 : 4-(4-(l-Ethyl-l#-pyrrolo[2,3-Λ]pyridin-3-yl)pyrimidin-2-ylamino)phenol (Compound 12)
Figure imgf000043_0003
[00297] A solution of l/f-pyrrolo[2,3-£]pyridine (10 mmol, 1.18g) in dry DMF (1OmL) was cooled on a an ice bath. NaH (130 mmol, 3.12g) was added portion wise under N2 atmosphere. After completion of the addition the reaction mixture was allowed to stir at room temperature for 45 minutes. Upon cooling at 00C iodoethane (30 mmol) was added and the reaction mixture was stirred at room temperature for 3 hours. Ice water was added. The mixture was diluted with diethyl ether and extracted with diethyl ether (4x30mL). The organic layers were combined and washed with brine, dried on MgSO4 and filtered. The solvent was evaporated to give brown oil which was purified by chromatography using EtOAc/PE to elute 1 -ethyl- l/f-pyrrolo[2,3-b]pyridine as light yellow oil. 1H-NMR (CD3Cl) δ: 1.50 (t, 3H, J = 7.6 Hz, CH3), 4.37 (q, 2H, J = 7.6 Hz, CH2), 6.47 (d, IH, J = 3.6 Hz, Ar-H), 7.07 (m, IH, Ar-H), 7.26 (d, IH, J= 3.6 Hz, Ar-H), 7.92 (d, IH, J= 8.0 Hz, Ar-H), 8.34 (d, IH, J= 4.8 Hz, Ar-H). MS (ESI+) m/z 147.1001 (M+H)+. [00298] Above compound (8.4 mmol, 1.22g) was added to stirred suspension Of AlCl3 (42 mmol) in dry CH2Cl2. After stirring at room temperature for 1 hour, acylchloride (42 mmol) was added dropwise. The reaction mixture was allowed to sire for further 1 hour at room temperature. Cooling on an ice bath methanol (40 niL) was added cautiously to quench the reaction. The solvent was evaporated in vacuo and the residue was purified by chromatography using EtOAc/MeOH (10: 1, v/v) to give l-(l-ethyl-l/f-pyrrolo[2,3-ft]pyridin-3-yl)ethanone as light yellow solid (0.69g, 44% yield). MS (ESI+) m/z 189.0967 (M+H)+.
[00299] A mixture of l-(l-ethyl-l/f-pyrrolo[2,3-£]pyridin-3-yl)ethanone (3 mmol) and 7V,7V-dimethyformamide dimethyal acetal (25 mmol) was microwaved at 1500C for 15 minutes. The precipitate was filtered and washed with EtOAc. The filtrate was concentrated in vacuo. The resulting solid was collected by filtration and washed with EtOAc followed by diethyl ether to result in (E)-3-(dimethylamino)-l-(l-ethyl-l/f-pyrrolo[2,3-i]pyridin-3-yl)prop-2-en-l-one. 1H-NMR (DMSO-d6) δ: 1.43 (t, 3H, J= 7.2 Hz, CH3), 4.33 (q, 2H, J= 7.6 Hz, CH2), 5.77 (d, IH, J= 12.4 Hz, CH), 7.20 (q, IH, J= 4.8 Hz, Ar-H), 7.58 (d, IH, J= 12.4 Hz, CH), 8.29 (m, IH, Ar-H), 8.41 (s, IH, Ar-H), 8.56 (m, IH, Ar-H). MS (ESI+) m/z 244.1222 (M+H)+.
[00300] A mixture of (E)-3-(dimethylamino)-l-(l-ethyl-l/f-pyrrolo[2,3-i]pyridin-3-yl)prop-2-en-l-one (5 mmol) in 3 mL EtOH/MeOH (1:2, v/v) was treated with l-(4-hydroxyphenyl)guanidine hydrochloride (5 mmol) and NaOH (10 mmol). The reaction mixture was microwaved at 1500C for 25 minutes. The residue was purified by chromatography using EtOAc to elute the titled compound. Recrystallisation from EtOAc yielded white solid. 1H-NMR (DMSO-d6) δ: 1.45 (t, 3H, J= 7.2 Hz, CH3), 4.37 (q, 2H, J= 7.6 Hz, CH2), 6.74 (d, 2H, J= 8.8 Hz, Ph-H), 7.15 (d, IH, J= 5.2 Hz, Pyrimidin-H), 7.20 (q, IH, J= 4.8 Hz, Ar-H), 7.51 (d, IR, J= 8.8 Hz, Ph-H), 8.29 (d, IH, J= 5.2 Hz, Pyrimidin-H), 8.35 (m, IH, Ar-H), 8.54 (s, IH, Ar-H), 8.88 (d, IH, J = 6.8 Hz, OH), 9.04 (s, IH, Ar-H), 9.10 (s, IH, NH). MS (ESI+) m/z 322.1469 (M+H)+.
[00301] The following compounds were synthesized by an analogous route. EXAMPLE 5 : 3-(4-(l-Ethyl-l//-pyrrolo[2,3-Λ]pyridin-3-yl)pyrimidin-2-ylamino)phenol (Compound 13)
Figure imgf000044_0001
[00302] Prepared by treatment of (E)-3-(dimethylamino)-l-(l-ethyl-l/f-pyrrolo[2,3-i]pyridin-3-yl)prop-2-en-l-one and l-(3-hydroxyphenyl)guanidine hydrochloride. 1H-NMR (DMSO-J6) δ: 1.18 (t, 3H, J= 7.2 Hz, CH3), 4.37 (q, 2H, J
= 7.2 Hz, CH2), 6.40 (m, IH, Ph-H), 7.08 (t, IH, J= 7.6 Hz, Ph-H), 7.18 (m, IH, Ar-H), 7.24 (d, IH, J= 5.6 Hz,
Pyrimidin-H), 7.37 (s, IH, Ph-H), 8.36 (m, 2H, Pyrimidin-H and Ar-H), 8.59 (s, IH, Ar-H), 9.00 (d, IH, J= 8.0 Hz,
Ph-H), 9.24 (s, IH, Ar-H), 9.33 (s, IH, NH). MS (ESI+) m/z 332.1355 (M+H)+.
[00303]
EXAMPLE 6: 4-(l-Ethyl-l^-pyrrolo[2,3-A]pyridin-3-yl)-7V-(3-nitrophenyl)pyrimidin-2-amine (Compound 14)
Figure imgf000045_0001
[00304] Prepared by treatment of (E)-3-(dimethylamino)-l-(l-ethyl-l/f-pyrrolo[2,3-i]pyridin-3-yl)prop-2-en-l-one and l-(3-nitrophenyl)guanidine hydrochloride 1H-NMR (DMSO-J6) δ 1 49 (t, 3H, J = 7 2 Hz, CH3), 4 37 (q, IH, J =
7 6 Hz, CH2), 7 26 (m, IH, Ar-H), 7 39 (d, IH, J= 5 2 Hz, Pyπmidin-H), 7 61 (t, IH, J= 8 0 Hz, Ph-H), 7 83 (m, IH, Ar-H), 8 12 (d, IH, J= 8 0 Hz, Ph-H), 8 38 (m, IH, Ar-H), 8 49 (d, IH, J= 5 2 Hz, Pyπmidin-H), 8 62 (s, IH, Ph-H),
8 92 (d, IH, J= 8 0 Hz, Ph-H), 9 01 (s, IH, Ar-H) MS (ESI+) m/z 361 1308 (M+H)+
EXAMPLE 7 : 4-(4-(l-Propyl-l//-pyrrolo[2,3-Z)]pyridin-3-yl)pyrimidin-2-ylamino)phenol (Compound 15)
Figure imgf000045_0002
[00305] Prepared by treatment of (E)-3-(dimethylamino)-l-(l-propyl-l/f-pyrrolo[2,3-£]pyridin-3-yl)prop-2-en-l-one with l-(4-hydroxyphenyl)guanidine hydrochloride 1H-NMR (DMSO-J6) δ 0 88 (t, 3H, J= 7 2 Hz, CH3), 1 90 (t, 2H, J= 7 6 Hz, CH2), 4 30 (t, 2H, J= 6 8 Hz, CH2), 6 74 (m, 2H, Ph-H), 7 16 (d, IH, J= 5 6 Hz, Pyπmidin-H), 7 20 (t, IH, J= 4 8 Hz, Ar-H), 7 51 (m, 2H, Ph-H), 8 30 (d, IH, J= 5 6 Hz, Pyπmidin-H), 8 35 (m, IH, Ar-H), 8 52 (s, IH, Ar-H), 8 88 (d, IH, J = 7 6 Hz, Ar-H), 9 05 (s, IH, NH), 9 11 (s, IH, OH) MS (ESI+) m/z 346 1659 (M+H)+ EXAMPLE 8: 3-(4-(l-Propyl-l//-pyrrolo[2,3-Λ]pyridin-3-yl)pyrimidin-2-ylamino)phenol Compound 16)
Figure imgf000045_0003
[00306] Prepared by treatment of (E)-3-(dimethylamino)-l-(l-propyl-l/f-pyrrolo[2,3-£]pyridin-3-yl)prop-2-en-l-one with l-(3-hydroxyphenyl)guanidine hydrochloride 1H-NMR (DMSO-J6) δ 0 88 (t, 3H, J= 7 2 Hz, CH3), 1 91 (t, 2H, J = 1 6 Hz, CH2), 4 31 (t, 2H, J= 6 8 Hz, CH2), 6 40 (m, IH, Ph-H), 7 08 (t, IH, J= 1 6 Hz, Ph-H), 7 18 (m, IH, Ar-H),
7 24 (d, IH, J= 5 6 Hz, Pyπmidin-H), 7 37 (s, IH, Ph-H), 8 36 (m, 2H, Pyπmidin-H and Ar-H), 8 57 (s, IH, Ar-H),
8 30 (d, IH, J= 5 6 Hz, Pyπmidin-H), 8 35 (m, IH, Ph-H), 8 57 (s, IH, Ar-H), 9 00 (d, IH, J= 8 0 Hz, Ar-H), 9 24 (s, IH, NH), 9 33 (s, IH, OH) MS (ESI+) m/z 346 1654 (M+H)+
EXAMPLE 9: N-(3-Nitrophenyl)-4-(l-propyl-l#-pyrrolo[2,3-Λ]pyridin-3-yl)pyrimidin-2-amine (Compound 17)
Figure imgf000045_0004
[00307] Prepared by treatment of (E)-3-(dimethylamino)-l-(l-propyl-l/f-pyrrolo[2,3-£]pyridin-3-yl)prop-2-en-l-one with l-(3-nitrophenyl)guanidine hydrochloride 1H-NMR (DMSO-J6) δ 0 90 (t, 3H, J= 7 2 Hz, CH3), 1 93 (t, IH, J = 7 6 Hz, CH2), 4 33 (t, 2H, J= 6 8 Hz, CH2), 7 26 (q, IH, J= 4 4 Hz, Ar-H), 7 40 (d, IH, J= 5 6 Hz, Pyπmidin-H), 7 61 (t, IH, J= 8 4 Hz, Ph-H), 7 81 (m, IH, Ph-H), 8 12 (m, IH, Ph-H), 8 37 (m, IH, Ar-H), 8 48 (d, IH, J= 5 6 Hz, Pyπmidin-H), 8 64 (s, IH, Ar-H), 8 92 (d, IH, J= 8 0 Hz, Ar-H), 9 00 (s, IH, NH) MS (ESI+) m/z 375 1519 (M+H)+ EXAMPLE 10 : 3-(4-(l-Ethyl-l//-pyrrolo[2,3-Λ]pyridin-3-yl)pyrimidin-2-ylamino)benzenesulfonamide (Compound 18)
Figure imgf000046_0001
[00308] Prepared by treatment of (E)-3-(dimethylamino)-l-(l-ethyl-l/f-pyrrolo[2,3-i]pyridin-3-yl)prop-2-en-l-one and 3-guanidinobenzenesulfonamide hydrochloride 1H-NMR (DMSO-J6) δ 1 48 (t, 3H, J = 7 2 Hz, CH3), 4 39 (q, 2H,
J = 7 2 Hz, CH2), 7 27 (m, IH, Ar-H), 7 33 (m, 3H, Pyπmidin-H and NH2), 7 42 (m, IH, Ph-H), 7 50 (t, IH, J = 7 6 Hz,
Ph-H), 7 92 (d, IH, J = 8 0 Hz, Ph-H), 8 36 (m, IH, Ar-H), 8 45 (d, IH, J= 5 6 Hz, Pyπmidin-H), 8 57 (s, IH, Ar-H),
8 65 ( s, IH, Ar-H), 8 90 (d, IH, J= 8 0 Hz, Ph-H), 9 82 (s, IH, Ar-H), 9 33 (s, IH, NH) MS (ESI+) m/z 332 1355
(M+H)+
EXAMPLE 11 : 3-(4-(l-Ethyl-l//-pyrrolo[2,3-Λ]pyridin-3-yl)pyrimidin-2-ylamino)benzamide (Compound 19)
Figure imgf000046_0002
[00309] Prepared by treatment of (E)-3-(dimethylamino)-l-(l-ethyl-l/f-pyrrolo[2,3-i]pyridin-3-yl)prop-2-en-l-one and 3-guanidinobenzamide hydrochloride 1H-NMR (DMSO-J6) δ 1 48 (t, 3H, J= 7 2 Hz, CH3), 4 38 (q, IH, J= 1 2 Hz, CH2), 7 23 (m, IH, Ar-H), 7 25 (m, IH, Pyπmidin-H), 7 30 (m, 2H, Ph-H and Ar-H), 7 46 (m, 2H, Ph-H and Ar-H), 8 35 (d, IH, J= 5 6 Hz, Pyπmidin-H), 8 42 (m, 3H, Ph-H and NH2), 8 65 ( s, IH, Ar-H), 8 91 (d, IH, J= 8 0 Hz, Ph- H), 9 59 (s, IH, NH) MS (ESI+) m/z 359 1619 (M+H)+ EXAMPLE 12: 3-(4-(l-Ethyl-l#-pyrrolo[2,3-Λ]pyridin-3-yl)pyrimidin-2-ylamino)benzonitrile (Compound 20)
Figure imgf000046_0003
[00310] Prepared by treatment of (E)-3-(dimethylamino)-l-(l-ethyl-l/f-pyrrolo[2,3-i]pyridin-3-yl)prop-2-en-l-one and l-(3-cyanophenyl)guamdine hydrochloride 1H-NMR (DMSO-J6) δ 1 48 (t, 3H, J= 7 2 Hz, CH3), 4 39 (q, 2H, J = 1 2 Hz, CH2), 7 27 (m, IH, Ar-H), 7 36 (d, IH, J= 4 4 Hz, Pyπmidin-H), 7 42 (m, IH, Ph-H or Ar-H), 7 51 (m, IH, Ph-H or Ar-H), 7 98 (m, IH, Ar-H), 8 38 (m, IH, Ph-H or Ar-H), 8 46 (m, 2H, Pyπmidin-H or Ar-H), 8 63 ( s, IH, Ar- H), 8 94 (m, IH, Ph-H or Ar-H), 9 85 (s, IH, NH) MS (ESI+) m/z 341 1489 (M+H)+
[00311] EXAMPLE 13: 3-(4-(l-Propyl-l//-pyrrolo[2,3-b]pyridin-3-yl)pyrimidin-2-ylamino)benzenesulfonamide (Compound 22)
Figure imgf000046_0004
[00312] Prepared by treatment of (E)-3-(dimethylamino)-l-(l-ethyl-l/f-pyrrolo[2,3-i]pyridin-3-yl)prop-2-en-l-one and 3-guanidinobenzenesulfonamide hydrochloride as described above 1H-NMR (DMSO-J6) δ 0 88 (t, 3H, J= 1 2 Hz, CH3), 1 91 (m, 2H, CH2), 4 32 (t, 2H, J = 7 2 Hz, CH2), 7 27 (m, IH, Ar-H), 7 32 (s, 2H, NH2), 7 36 (m, IH, J= 5 2 Hz, Pyrimidin-H), 7.43 (m, IH, Ph-H), 7.51 (m, IH, Ph-H), 7.92 (d, IH, J = 8.0 Hz, Ph-H), 8.36 (m, IH, Ar-H), 8.44 (d, IH, J = 5.2 Hz, Pyrimidin-H), 8.56 (s, IH, Ar-H), 8.63 (s, IH, Ph-H), 8.91 (d, IH, , J = 6.8 Hz, Ar-H), 9.82 (s, IH, NH). MS (ESI+) m/z 409.8665 (M+H)+.
[00313] EXAMPLE 14: 4-(4-(l-Propyl-l//-pyrrolo[2,3-Λ]pyridin-3-yl)pyrimidin-2-ylamino)benzenesulfonamide (Compound 23)
Figure imgf000047_0001
[00314] Prepared by treatment of (E)-3-(dimethylamino)-l-(l-ethyl-l/f-pyrrolo[2,3-i]pyridin-3-yl)prop-2-en-l-one and 4-guanidinobenzenesulfonamide hydrochloride as described above. 1H-NMR (DMSO-J6) δ: 0.90 (t, 3H, J= 7.2 Hz, CH3), 1.91 (m, 2H, CH2), 4.33 (t, 2H, J = 7.2 Hz, CH2), 7.19 (s, 2H, NH2), 7.29 (m, IH, Ar-H), 7.36 (m, IH, J= 5.6 Hz, Pyrimidin-H), 7.77 (d, 2H, J= 7.2 Hz, Ph-H), 8.00 (d, 2H, J= 7.2 Hz, Ph-H), 8.38 (m, IH, Ar-H), 8.45 (d, IH, J= 5.6 Hz, Pyrimidin-H), 8.62 (s, IH, Ar-H), 8.97 (m, IH, Ar-H), 9.88 (s, IH, NH). MS (ESI+) m/z 409.8708 (M+H)+. [00315] EXAMPLE 15: 7V-(4-Methyl-3-(morpholinosulfonyl)phenyl)-4-(l-propyl-l^-pyrrolo[2,3-A]pyridin-3- yl)pyrimidin-2-amine (Compound 24)
Figure imgf000047_0002
[00316] Prepared by treatment of (E)-3-(dimethylamino)-l-(l-ethyl-l/f-pyrrolo[2,3-i]pyridin-3-yl)prop-2-en-l-one and l-(4-methyl-3-(moφholinosulfonyl)phenyl)guanidine hydrochloride as described above. 1H-NMR (DMSO-J6) δ: 0.89 (t, 3H, J= 7.2 Hz, CH3), 1.92 (m, 2H, CH2), 1.99 (s, 3H, CH3), 3.07 (m, 4H, CH2x2), 3.64 (m, 4H, CH2x2), 4.32 (t, 2H, J= 7.2 Hz, CH2), 7.27 (m, IH, Ar-H), 7.33 (m, IH, J= 5.6 Hz, Pyrimidin-H), 7.40 (d, IH, J= 8.4 Hz, Ph-H), 7.98 (m, IH, Ph-H), 8.36 (m, IH, Ar-H), 8.43 (d, IH, J= 5.6 Hz, Pyrimidin-H), 8.49 (m, IH, Ph-H), 8.64 (s, IH, Ar-H), 8.92 (m, IH, Ar-H), 9.79 (s, IH, NH). MS (ESI+) m/z 493.7652(M+H)+.
[00317] EXAMPLE 16: (4-Methylpiperazin-l-yl)(3-(4-(l-propyl-l^-pyrrolo[2,3-b]pyridin-3-yl)pyrimidin-2- ylamino)phenyl)methanone (Compound 34)
Figure imgf000047_0003
[00318] Prepared by treatment of (E)-3-(dimethylamino)-l-(l-ethyl-l/f-pyrrolo[2,3-i]pyridin-3-yl)prop-2-en-l-one and l-(3-(4-methylpiperazine-l-carbonyl)phenyl)guanidine hydrochloride as described above. 1H-NMR (DMSO-J6) δ: 0.90 (t, 3H, J= 7.2 Hz, CH3), 1.90 (m, 2H, CH2), 2.19 (s, 3H, CH3), 3.07 (m, 4H, CH2x2), 3.61 (m, 4H, CH2x2), 4.31 (t, 2H, J= 7.2 Hz, CH2), 7.26 (m, IH, Ar-H), 7.34 (m, IH, J= 5.6 Hz, Pyrimidin-H), 7.44 (m, IH, Ph-H), 7.98 (m, IH, Ph-H), 8.34 (m, IH, Ar-H), 8.45 (d, IH, J= 5.6 Hz, Pyrimidin-H), 8.49 (m, IH, Ph-H), 8.64 (s, IH, Ar-H), 8.92 (m, IH, Ar-H), 9.79 (s, IH, NH). MS (ESI+) m/z 456.8986 (M+H)+. [00319] EXAMPLE 17: 3-(4-(5-Chloro-7-methyl-l-propyl-l^-pyrrolo[3,2-A]pyridin-3-yl)pyrimidin-2- ylamino)benzenesulfonamide (Compound 42)
Figure imgf000048_0001
[00320] A solution of 2-chloro-4-methyl-5-nitro-pyridine (31.5 mmol) in dry THF cooling at -78°C was added 10OmL vinylmagnisium bromide (1.0 M in THF). After stirring at -78°C for 1 hr, the mixture was warmed to -200C and stirred for further 8 hrs. NH4Cl (20% in H2O, 15OmL) was added cautiously and the mixture was extracted with EtOAc (3xl50mL). The organic layers were combined, washed with brine, dried on MgSO4 and filtered. The filtrate was evaporated in vacuo. The residue was purified by column chromatography using EtOAcv/PE (1:4, v/v) to elute 2.4g 5- chloro-7-methyl-l/f-pyrrolo[3,2-£]pyridine as white solid. 1H-NMR (DMSO-J6) δ: 6.51 (m, IH, Ar-H), 6.99 (s, IH, Ar-H), 7.68 (t, IH, J = 2.8 Hz, Ar-H). MS (ESI+) m/z 167.0419 (M+H)+.
[00321] A solution of 5-chloro-7 -methyl- l/f-pyrrolo[3,2-£]pyridine (5 mmol) in dry DMF (1OmL) cooling on an ice bath was treated with NaH (60 mmol) under N2. After stirring at room temperature for 45 minutes iodopropane (10 mmol) was added. The mixture was allowed to react for further 3 hrs. Upon cooling on an ice bath it was treated with H2O (3mL). The mixture was then extracted with diethyl ether (4x50mL). The ether layers were combined, washed with brine, dried on MgSO4 and filtered. The solvent was evaporated to give 5-chloro-7-methyl-l-propyl-l/f- pyrrolo[3,2-6]pyridine as orange oil. 1H-NMR (DMSO-J6) δ: 0.85 (t, 3H, J = 7.6 Hz, CH3), 1.75 (m, 2H, CH2), 4.28 (t, 2H, J= 7.6 Hz, CH2), 6.48 (d, IH, J= 3.2 Hz, Ar-H), 6.97 (s, IH, Ar-H), 7.66 (d, IH, J= 3.6 Hz, Ar-H). MS (ESI+) m/z 209.0648 (M+H)+.
[00322] A mixture of 5-chloro-7-methyl-l-propyl-l/f-pyrrolo[3,2-i]pyridine (4 mmol) and AlCl3 (120 mmol) in dry CH2Cl2 was stirred at room temperature for lhr under N2. Upon cooling on an ice bath the mixture was added acetyl chloride (30 mmol) and stirred at room temperature for further 2 hrs. MeOH (50 mL) was added dropwise. The mixture was concentrated in vacuo and purified by column chromatography using EtOAc/MeOH (10: 1, v/v) to elute l-(5- chloro-7-methyl-l-propyl-l/f-pyrrolo[3,2-ft]pyridin-3-yl)ethanone as light yellow solid. 1H-NMR (DMSO-J6) δ: 0.85 (t, 3H, J= 7.6 Hz, CH3), 1.79 (m, 2H, CH2), 2.51 (s, 3H, CH3), 4.37 (t, 2H, J= 7.3 Hz, CH2), 7.17 (s, IH, Ar-H), 8.38 (s, IH, Ar-H). MS (ESI+) m/z 251.0894 (M+H)+.
[00323] l-(5-Chloro-7-methyl-l-propyl-l/f-pyrrolo[3,2-ft]pyridin-3-yl)ethanone (0.8 mmol) was treated with NJV- dimethyformamide dimethyal acetal (3mL). The mixture was microwaved at 1600C for 60 minutes. The precipitates were collected by filtration followed by recrystallisation from EtOAc to give (E)-l-(5-chloro-7-methyl-l-propyl-l/f- pyrrolo[3,2-ft]pyridin-3-yl)-3-(dimethylamino)prop-2-en-l-one as yellow crystals. 1H-NMR (DMSO-J6) δ: 0.86 (t, 3H, J= 7.2 Hz, CH3), 1.77 (m, 2H, CH2), 4.34 (t, 2H, J= 12 Hz, CH2), 6.65 (d, IH, J= 12.4 Hz, CH), 7.09 (s, IH, Ar-H), 7.67 (d, IH, J= 12.4 Hz, CH), 8.17 (s, IH, Ar-H). MS (ESI+) m/z 306.1259 (M+H)+.
[00324] A mixture of (E)-l-(5-chloro-7-methyl-l-propyl-l/f-pyrrolo[3,2-i]pyridin-3-yl)-3-(dimethylamino)prop-2- en-l-one (0.2 mmol), 3-guanidinobenzenesulfonamide hydrochloride (0.5mmol) and NaOH (0.8mmol) in 3 mL of 2- methoxyethanol was heated in a Microwave at 1500C for 30 minutes. The reaction mixture was purified by column chromatography using EtOAc/PE (1: 1, v/v) to elute 3-(4-(5-chloro-7-methyl-l-propyl-l/f-pyrrolo[3,2-£]pyridin-3- yl)pyrimidin-2-ylamino)benzenesulfonamide which was re-crystallised from EtOAc to give white solid. 1H-NMR (DMSO-J6) δ: 0.92 (t, 3H, J= 7.2 Hz, CH3), 1.87 (m, 2H, CH2), 4.42 (t, 2H, J= 7.2 Hz, CH2), 7.19 (s, IH, Ar-H or Ph- H), 7.34 (s, 2H, NH2), 7.43 (m, IH, Ar-H or Ph-H), 7.49 (t, IH, J= 7.6 Hz, Ph-H), 7.62 (m, IH, Ph-H), 8.02 (d, IH, J = 4.8 Hz, Pyrimidin-H), 8.55 (d, IH J= 5.2 Hz, Pyrimidin-H), 8.63 (s, IH, Ar-H), 9.92 (s, IH, NH). MS (ESI+) m/z 457.1245 (M+H)+.
[00325] EXAMPLE 18: 4-(4-(5-Chloro-7-methyl-l-propyl-lH-pyrrolo[3,2-b]pyridin-3-yl)pyrimidin-2- ylamino)benzenesulfonamide (Compound 39)
Figure imgf000049_0001
[00326] Prepared by treatment of (E)-l-(5-chloro-7-methyl-l-propyl-l/f-pyrrolo[3,2-£]pyridin-3-yl)-3-
(dimethylamino)prop-2-en-l-one and 3-guanidinobenzenesulfonamide hydrochloride. 1H-NMR (DMSO-J6) δ: 0.93 (t,
3H, J = 7.2 Hz, CH3), 1.84 (m, 2H, CH2), 4.47 (t, 2H, J = 7.2 Hz, CH2), 7.18 (m, 3H, Ar-H and NH2), 7.79 (d, 2H, J =
8.8 Hz, Ph-H), 8.03 (d, IH, J= 5.2 Hz, Pyrimidin-H), 8.07 (d, 2H, J= 8.8 Hz, Ph-H), 8.48 (s, IH, Ar-H), 8.56 (d, IH,
J= 5.2 Hz, Pyrimidin-H), 9.90 (s, IH, NH). MS (ESI+) m/z 456.7704.
EXAMPLE 19: 4-(4-(7-Chloro-l-ethyl-l//-pyrrolo[2,3-c]pyridin-3-yl)pyrimidin-2-ylamino)phenol (Compound
70)
Figure imgf000049_0002
[00327] A solution of 2-chloro-3-nitropyridine (31.5 mmol) in dry THF (200 mL) was cooled to -78°C. Vinylmagnisium bromide (1.0 M in THF, 10OmL) was added dropwise under N2. The mixture was stirred at -78°C for lhr, followed by -200C for further 8 hrs. The mixture was quenched with 150 mL NH4Cl (20% in H2O) and extracted with EtOAc (3xl50mL). The organic layers were combined, washed with brine, dried on MgSO4 and filtered. The solvent was evaporated and the residue was purified by column chromatography using EtOAc/PE (1:3, v/v) to elute 7- chloro-l/f-pyrrolo[2,3-c]pyridine as white solid (1.77 g, yield 37%). 1H-NMR (DMSO-J6) δ: 6.63 (d, IH, J= 3.2 Hz, Ar-H), 7.58 (d, IH, J= 5.6 Hz, Ar-H), 7.66 (d, IH, J= 2.8 Hz, Ar-H), 7.90 (d, IH, J= 5.2 Hz, Ar-H). MS (ESI+) m/z 153.0260 (M+H)+.
[00328] A mixture of 7-chloro-l/f-pyrrolo[2,3-c]pyridine (11 mmol) in dry DMF (20 mL) was cooled on an ice bath, NaH (143 mmol) was added portion wise under N2. The mixture was allowed to stir at room temperature for 1 hr. Upon cooling to 00C, iodoethane (26 mmol) was added and the mixture was reacted for further 3 hrs. After quenching with H2O (2OmL), the mixture was diluted with diethyl ether and extracted with ether (4x50mL). The organic layers were combined, washed with brine, dried with MgSO4 and filtered. The solvent was evaporated to give 7-chloro-l -ethyl- IH- pyrrolo[2,3-c]pyridine as orange oil. MS (ESI+) m/z 181.0533 (M+H)+.
[00329] To a suspension Of AlCl3 (55 mmol) in dry CH2Cl2 (300 mL) 7-chloro-l-ethyl-l/f-pyrrolo[2,3-c]pyridine (11 mmol) was added. After stirred at room temperature for 1 hr, acetyl chloride (55 mmol) was added. The reaction mixture was allowed to react for further 2 hrs. Upon cooling on an ice bath MeOH (80 mL) was added. The mixture was concentrated followed by column chromatography using EtOAc/MeOH (10: 1, v/v) to elute 1 -(7-chloro-l -ethyl- l/f-pyrrolo[2,3-c]pyridin-3-yl)ethanone as off-white solid. 1H-NMR (DMSO-J6) δ: 1.47 (t, 3H, J= 7.2 Hz, CH3), 4.62 (t, 2H, J= 7.2 Hz, CH2), 8.11 (m, 2H, Ar-H), 8.68 (s, IH, Ar-H). MS (ESI+) m/z 223.0384 (M+H)+ [00330] A mixture of l-(7-chloro-l-ethyl-l/f-pyrrolo[2,3-c]pyridin-3-yl)ethanone (3 mmol) treated with 3.5 mL NJV- dimethylformamide dimethyl acetal was microwaved at 1500C for 35 minutes. The mixture was evaporated to dryness and purified by column chromatography using EtOAc/MeOH (10: 1) to elute (E)-l-(7-chloro-l-ethyl-l/f-pyrrolo[2,3- c]pyridin-3-yl)-3-(dimethylamino)prop-2-en-l-one as yellow solid. 1H-NMR (DMSO-J6) δ: 1.43 (t, 3H, J = 7.2 Hz,
CH3), 4.43 (t, 2H, J = 7.2 Hz, CH2), 5.72 (d, IH, J= 12.4 Hz, CH), 7.58 (d, IH, J= 12.4 Hz, CH), 7.71 (d, IH, J = 5.6
Hz, Ar-H), 7.79 (d, IH, J= 5.6 Hz, Ar-H), 8.25(s, IH, Ar-H). MS (ESI+) m/z 278.1045 (M+H)+.
[00331] A mixture of (E)-l-(7-chloro-l-ethyl-l/f-pyrrolo[2,3-c]pyridin-3-yl)-3-(dimethylamino)prop-2-en-l-one (0.5 mmol), l-(4-hydroxyphenyl)guanidine hydrochloride (0.5 mmol) and NaOH (2 mmol) in a mixture of EtOH-MeCN
(1:2, v/v, 3 mL) was heated in a microwave at 1500C for 25 minutes. The residue was purified by column chromatography using EtOAc/PE (1:3, v/v) to yield 4-(4-(7-chloro-l-ethyl-l/f-pyrrolo[2,3-c]pyridin-3-yl)pyrimidin-2- ylamino)phenol as white solid. 1H-NMR (DMSO-J6) δ: 1.43(m, 3H, CH3), 4.49 (m, 2H, CH2), 6.74 (m, 2H, Ph-H),
7.11 (d, IH, J= 5.6 Hz, Pyrimidin-H), 7.52 (m, 2H, Ph-H), 7.74 (d, IH, J= 5.6 Hz, Ar-H), 8.10 (d, IH, J= 5.2 Hz, Ar-
H), 8.29 (d, IH J= 5.6 Hz, Pyrimidin-H), 8.37 (s, IH, Ar-H), 9.04 (s, IH, OH), 9.09 (s, IH, NH). MS (ESI+) m/z
362.1609 (M+H)+.
[00332] The following compounds were synthesised by an analogous route.
EXAMPLE 20: 3-(4-(7-Chloro-l-ethyl-l/f-pyrrolo[2,3-c]pyridin-3-yl)pyrimidin-2-ylamino)phenol (Compound
72)
Figure imgf000050_0001
[00333] Prepared by treatment of (E)-l-(7-chloro-l-ethyl-l/f-pyrrolo[2,3-c]pyridin-3-yl)-3-(dimethylamino)prop-2- en-l-one with l-(3-hydroxyphenyl)guanidine hydrochloride. 1H-NMR (DMSO-J6) δ: 1.46 (m, 3H, CH3), 4.49 (m, 2H, CH2), 6.39 (m, IH, Ph-H), 7.07 (m, IH, Ph-H), 7.20 (m, 2H, Ph-H and Ar-H), 7.77 (d, IH, J= 5.6 Hz, Pyrimidin-H), 8.18 (d, IH, J= 5.6 Hz, Ar-H), 8.37 (d, IH, J= 5.6 Hz, Pyrimidin-H), 8.41 (s, IH, Ar-H), 9.23 (s, IH, OH), 9.32 (s, IH, NH). MS (ESI+) m/z 362.1670 (M+H)+
EXAMPLE 21: 3-(4-(7-Chloro-l-ethyl-l/f-pyrrolo[2,3-c]pyridin-3-yl)pyrimidin-2-ylamino)benzenesulfonamide (Compound 73)
Figure imgf000050_0002
[00334] Prepared by treatment of (E)-l-(7-chloro-l-ethyl-l/f-pyrrolo[2,3-c]pyridin-3-yl)-3-(dimethylamino)prop-2- en-l-one and 3-guanidinobenzenesulfonamide hydrochloride. 1H-NMR (DMSO-J6) δ: 1.45 (t, 3H, J= 7.2 Hz, CH3), 4.48 (t, 2H, J= 7.2 Hz, CH2), 7.30 (m, 3H, Pyrimidin-H and NH2), 7.43 (m, IH, Ar-H or Ph-H), 7.50 (t, IH, J= 7.6 Hz, Ph-H), 7.82 (d, IH, J= 5.6 Hz, Ar-H), 7.89 (m, IH, Ar-H or Ph-H), 8.07 (m, IH, Ar-H or Ph-H), 8.45 (d, IH, J = 5.6 Hz, Pyrimidin-H), 8.48 (s, IH, Ar-H), 8.61 (s, IH, NH). MS (ESI+) m/z 426.1490 (M+H)+. [00335] EXAMPLE 22: 3-(4-(7-chloro-l-propyl-lH-pyrrolo[2,3-c]pyridin-3-yl)pyrimidin-2- ylamino)benzenesulfonamide (Compound 96)
Figure imgf000050_0003
[00336] Prepared by treatment of (E)-l-(7-chloro-l-ethyl-l/f-pyrrolo[2,3-c]pyridin-3-yl)-3-(dimethylamino)prop-2- en-l-one and 3-guanidinobenzenesulfonamide hydrochloride. 1H-NMR (DMSO-J6) δ: 0.79 (t, 3H, J = 7.2 Hz, CH3), 1.18 (m, 2H, J = 7.2 Hz, CH2), 4.46 (m, 2H, CH2), 7.34 (m, 3H, Ar-H and NH2), 7.46 (m, IH, Ar-H or Ph-H), 7.54 (m, IH, Ph-H or Ar-H), 7.91 (m, IH, Ar-H), 8.11 (d, IH, J= 5.6 Hz, Pyrimidin-H), 8.49 (d, IH, J= 5.6 Hz, Pyrimidin-H), 8.54 (s, IH, Ar-H or Ph-H), 8.56 (m, IH, Ar-H), 8.71 (s, IH, Ar-H), 9.90 (s, IH, NH). MS (ESI+) m/z 444.7773 (M+H)+.
[00337] EXAMPLE 23: 4-(4-(7-Chloro-l-propyl-l#-pyrrolo[2,3-c]pyridin-3-yl)pyrimidin-2- ylamino)benzenesulfonamide (Compound 97)
Figure imgf000051_0001
[00338] Prepared by treatment of (E)-l-(7-chloro-l-ethyl-l/f-pyrrolo[2,3-c]pyridin-3-yl)-3-(dimethylamino)prop-2- en-l-one and 4-guanidinobenzenesulfonamide hydrochloride. 1H-NMR (DMSO-J6) δ: 0.79 (t, 3H, J= 7.2 Hz, CH3), 1.18 (m, 2H, J= 7.2 Hz, CH2), 4.46 (m, 2H, CH2), 5.79 (s, 2H, NH2), 6.58 (m, 3H, Ph-H and Ar-H), 6.88 (m, 2H, Ar- H), 7.21 (m, 2H, Ph-H), 7.61 (d, IH, J= 6.0 Hz, Pyrimidin-H), 7.73 (d, IH, J= 6.0 Hz, Pyrimidin-H), 10.14 (s, IH, NH). MS (ESI+) m/z 442.0773.
[00339] EXAMPLE 24: 4-(7-Chloro-l-propyl-l#-pyrrolo[2,3-c]pyridin-3-yl)-7V-(4-methyl-3- (morpholinosulfonyl)phenyl)pyrimidin-2-amine (Compound 101)
Figure imgf000051_0002
[00340] Prepared by treatment of (E)-l-(7-chloro-l-ethyl-l/f-pyrrolo[2,3-c]pyridin-3-yl)-3-(dimethylamino)prop-2- en-l-one and l-(3-(moφholinosulfonyl)phenyl)guanidine hydrochloride. 1H-NMR (DMSO-J6) δ: 0.86 (t, 3H, J= 7.2 Hz, CH3), 1.86 (m, 2H, CH2), 2.53(m, 4H, CH2x2), 3.25 (m, 4H, CH2x2), 4.06 (s, 3H, CH3), 4.35 (t, 2H, J= 7.2 Hz, CH2), 7.28 (d, IH, J= 5.6 Hz, Pyrimidin-H), 7.40 (d, IH, J= 8.8 Hz, Ar-H or Ph-H), 7.81 (d, IH, J= 5.6 Hz, Ar-H), 7.95 (m, IH Ar-H or Ph-H), 8.18 (m, IH, Ar-H or Ph-H), 8.43 (d, IH, J = 5.6 Hz, Pyrimidin -H), 8.48 (s, IH, Ar-H or Ph-H), 8.54 (m, IH, Ar-H), 9.79 (s, IH, NH). MS (ESI+) m/z 526.6872.
EXAMPLE 25: 7V-(4-Methyl-3-(morpholinosulfonyl)phenyl)-4-(l-propyl-l^-pyrrolo[2,3-c]pyridin-3- yl)pyrimidin-2-amine (Compound 102)
Figure imgf000051_0003
[00341] Hydrogenation of 7-chloro-l/f-pyrrolo[2,3-c]pyridine (10 mmol) was carried out in the presence of Pd-C (10%) in EtOH (5OmL). After stirred at room temperature for 20 hrs, the mixture was filtered through a pad of celite 521. The residue was washed with EtOH several times. The filtrate was evaporated to yield l/f-pyrrolo[2,3-c]pyridine as brown oil. MS (ESI+) m/z 119.0657 (M+H)+.
[00342] A solution of l/f-pyrrolo[2,3-c]pyridine (10 mmol) in DMF cooling on an ice bath was treated with NaH (143 mmol). After stirred at room temperature for lhr iodopropane (26 mmol) was added dropwise. The mixture was allowed to react at room temperature for further 3 hrs. Upon cooling on an ice bath the mixture was quenched with 20 niL H2O, followed by extraction with diethyl ether (5x50mL). The organic layers were combined, washed with brine, dried on Mg SO4 and filtered. The solvent was evaporated and dried on a high vacuum to give 1 -propyl- IH- pyrrolo[2,3-c]pyridine as orange oil. MS (ESI+) m/z 161.1063 (M+H)+.
[00343] A mixture of 1 -propyl- l/f-pyrrolo[2,3-c]pyridine (10 mmol) and AlCl3 (50 mmol) in dry CH2Cl2 (200 mL) was stirred at room temperature for 1 hr before being treated with acetyl chloride (50 mL). After stirred for further 2 hrs, the mixture was quenched with MeOH (55 mL) cautiously. The solvent was evaporated and the residue was purified by column chromatography using EtOAc/MeOH (10: 1, v/v) to elute l-(l-propyl-l/f-pyrrolo[2,3-c]pyridin-3- yl)ethanone as off-white solid. 1H-NMR (DMSO-J6) δ: 0.89 (t, 3H, J = 7.2 Hz, CH3), 1.89 (m, 2H, CH2), 2.47 (s, 3H, CH3), 4.32 (t, 2H, J = 7.2 Hz, CH2), 8.05 (d, IH, J= 5.2 Hz, Ar-H), 8.31 (d, IH, J= 5.2 Hz, Ar-H), 8.56 (s, IH, Ar-H), 9.00 (s, IH, Ar-H). MS (ESI+) m/z 223.0384 (M+H)+
[00344] A mixture of l-(l-propyl-l/f-pyrrolo[2,3-c]pyridin-3-yl)ethanone (4 mmol) and 7V,7V-dimethyrformamide dimethylacetal (25 mmol) was heated in microwave at 1500C for 15 minutes. The mixture was evaporated to dryness and purified by column chromatography using EtOAc/MeOH (10: 1, v/v) to elute (E)-3-(dimethylamino)-l-(l-propyl- l/f-pyrrolo[2,3-c]pyridin-3-yl)prop-2-en-l-one as orange solid. 1H-NMR (DMSO-J6) δ: 0.86 (t, 3H, J= 7.2 Hz, CH3), 1.87 4.29 (t, 2H, J= 7.2 Hz, CH2), 5.74 (d, IH, J= 12.4 Hz, CH), 7.58 (d, IH, J= 12.4 Hz, CH), 8.12 (d, IH, J= 5.2 Hz, Ar-H), 8.21 (d, IH, J= 5.2 Hz, Ar-H), 8.40 (s, IH, Ar-H), 8.92 (s, IH, Ar-H). MS (ESI+) m/z 258.1546 (M+H)+ [00345] A mixture of (E)-3-(dimethylamino)-l-(l-propyl-l/f-pyrrolo[2,3-c]pyridin-3-yl)prop-2-en-l-one (0.5 mmol), l-(4-methyl-3-(morpholinosurfonyl)phenyl)guanidine hydrochloride (0.5 mmol) and NaOH (1.0 mmol) was microwaved at 1500C for 15 minutes. The residue was purified by column chromatography using EtOAc to yield 7V-(4- methyl-3 -(morpholinosurfonyl)phenyl)-4-( 1 -propyl- l/f-pyrrolo[2,3 -c]pyridin-3 -yl)pyrimidin-2 -amine as light yellow solid. 1H-NMR (DMSO-J6) δ: 0.89 (t, 3H, J= 7.2 Hz, CH3), 1.91 (m, 2H, CH2), 3.07(m, 4H, CH2x2), 3.42 (s, 3H, CH3), 3.64 (m, 4H, CH2x2), 4.35 (t, IR, J= 7.2 Hz, CH2), 7.33 (d, IH, J= 5.6 Hz, Pyrimidin-H), 7.41 (m, IH, Ar-H or Ph-H), 7.98 (m, IH, Ar-H or Ph-H), 8.33 (d, IH J= 5.2 Hz, Ar-H), 8.45 (d, IH, J= 5.6 Hz, Pyrimidin-H), 8.52 (s, IH, Ar-H or Ph-H), 8.65 (s, IH, Ph-H or Ar-H), 9.02 (s, IH, Ar-H or Ph-H), 9.92 (s, IH, Ar-H or Ph-H). MS (ESI+) m/z 493.2182 (M+H)+. EXAMPLE 26: 3-(4-(l-Propyl-lff-pyrrolo[2,3-c]pyridin-3-yl)pyrimidin-2- ylamino)benzenesulfonamide (Compound 111)
Figure imgf000052_0001
Prepared by treatment of (E)-3-(dimethylamino)-l-(l-propyl-l/f-pyrrolo[2,3-c]pyridin-3-yl)prop-2-en-l-one and 3- guanidinobenzenesulfonamide hydrochloride as described above. 1H-NMR (DMSO-J6) δ: 0.89 (t, 3 H, J= 7.2 Hz, CH3), 1.91 (m, 2H, CH2), 4.36 (t, IR, J= 7.2 Hz, CH2), 6.56 (s, 2H, NH2), 7.34 (m, 2H, Pyrimidin-H and Ph-H), 7.44 (m, IH, Ar-H or Ph-H), 7.91 (m, IH, Ar-H or Ph-H), 8.33 (d, IH J= 5.2 Hz, Ar-H), 8.45 (d, IH, J= 5.6 Hz, Pyrimidin-H), 8.52 (m, 2H, Ar-H or Ph-H), 9.02 (s, IH, Ar-H or Ph-H), 9.84 (s, IH, Ar-H or Ph-H). MS (ESI+) m/z 409.1514 (M+H)+. EXAMPLE 27: 4-(4-(l-Propyl-l//-pyrrolo[2,3-c]pyridin-3-yl)pyrimidin-2-ylamino)benzenesulfonamide (Compound 112)
Figure imgf000053_0001
Prepared by treatment of (E)-l-(7-chloro-l-ethyl-l/f-pyrrolo[2,3-c]pyridin-3-yl)-3-(dimethylamino)prop-2-en-l-one and 4-guanidinobenzenesulfonamide hydrochloride as described above. 1H-NMR (DMSO-Ii6) δ: 0.92 (t, 3H, J= 7.2 Hz, CH3), 1.91 (m, 2H, CH2), 4.36 (t, IR, J= 7.2 Hz, CH2), 7.21 (m, 2H, NH2), 7.36 (d, IH, J= 5.6 Hz, Pyrimidin-H), 7.76 (m, 2H, Ph-H), 8.01 (m, 2H, Ph-H), 8.32 (d, IH J= 5.2 Hz, Ar-H), 8.46 (d, IH, J= 5.6 Hz, Pyrimidin-H), 8.52 (m, IH, Ar-H), 8.61 (s, IH, Ar-H), 9.02 (s, IH, Ar-H), 9.90 (s, IH, NH). MS (ESI+) m/z 409.8821 (M+H)+. EXAMPLE 28 - KINASE INHIBITORY ACTIVITY
[00346] Kinase assays. The compounds from the example above were tested for their ability to inhibit kinases. Recombinant human CDK2/cyclinA (PV3267), CDKl/cyclinB (PV3292), CDK5/p35 (PV3000) and Aurora A (PV3612) were purchased from Invitrogen (Carlsbad, California, USA). Kinase dosage and Z'-LYTE™ substrate for each kinase assay is as follows: CDK2/cyclinA-8ng/reaction, Ser/Thr 12 Peptide; CDKl/cyclinB-0.8ng/reaction, Ser/Thr 12 Peptide, CDK5/p35-0.8ng/reaction, Ser/Thr 12 Peptide, and Aurora A- long/reaction, Ser/Thr 1 Peptide. All enzymatic assays were performed using Z'-LYTE™ Kinase Assay Kits and were carried out following protocols provided by Invitrogen. To prepare stock solutions, compounds were dissolved at 1OmM in 100% DMSO and stored at 4°C in amber glass vials. Enzyme reactions were initiated by adding recombinant enzyme and Z'-LYTE™ Ser/Thr peptide substrate and ATP mixture to 384-well plates containing diluted test compounds. Kinase and corresponding peptide substrate were incubated for lhr at room temperature in the presence of varying amounts of test compound with reaction buffer (20 μM ATP, 25 mM HEPES [pH 7.5], 5 niM MgCl2, 0.5 niM EGTA, 0.005% Brij-35). Development buffer was added and incubated for another one hour at room temperature. Reactions were terminated by addition of stop buffer, and fluorescence emission intensity ratio 445 nm/520 nm (excited at 400 ran) was quantified by VICTOR-3 Multilabel Reader (Perkin-Elmer, Waltham, Massachusetts, USA). Phosphorylation was calculated from the emission ratio. IC50 was determined using Sigmoid curve-fitting software Origin 6.0 (MicroCal, LLC, Northampton, Massachusetts, USA). Kinase activity of representative compounds is presented in Table I. Table I. Kinase inhibitory activity
Figure imgf000053_0002
Figure imgf000054_0001
ND-Not Determined
EXAMPLE 29 - CYTOTOXICITY
[00347] MTT cytotoxicity assay. The compounds of the invention were subjected to a standard cellular proliferation assay using human cell lines. HeLa cells (Human cervix adenocarcinoma cell line, ATCC NO, CCL-2); A549 cells (Human lung carcinoma cells line, ATCC NO, CCL- 185 ); U2OS cells (Human osteosarcoma cell line, ATCC NO, HTB-96); Eca- 109 cells (Human esophageal carcinoma cell line); HepG2 (Human hepatocellular liver carcinoma cell line, ATCC NO, HB-8065); K562 cells ( Human chronic myelogenous leukemia cell line, ATCC NO, CCL-243); SK- MEL-28 (Human malignant melanoma cell line, ATCC NO, HTB-72); Molt-4 (Human acute lymphoblastic leukemia cell line, ATCC NO, CRL-1582); H460 (Human large cell lung cancer cell line, ATCC NO, HTB-177); MDA-MB-231 (Human breast adenocarcinoma cell line, ATCC NO, HTB-26); MCF7 (Human breast adenocarcinoma cell line, ATCC NO, HTB-22); ACHN (Human renal cell adenocarcinoma cell line, ATCC NO, CRL- 1611); PC-3M (Human prostate adenocarcinoma cell line, ATCC NO CRL-1435); HL60(Human acute promyelocytic leukemia cell line, ATCC NO, CCL-240); THP-I (Human acute monocytic leukemia, ATCC NO, TIB-202); Jurkat (Human acute T cell leukemia cell line, ATCC NO, CRL-2063); HCT-116 (Human colorectal carcinoma cell line, ATCC NO, CCL-247); HT-29 (Human colorectal adenocarcinoma cell line, ATCC NO, HTB-38™); WI-38 (Human normal lung cell line, ATCC NO,CCL- 75); MRC-5 (Human normal lung cell line, ATCC NO, CCL-171).
[00348] HepG2, MDA-MB-231, WI-38 and MRC-5 cells were obtained from Shanghai Institute of Cell Biology, Chinese Academy of Sciences (Shanghai 200031, China). A549, HeLa, K562, MCF7, ACHN, H460, U2OS, Eca-109, MCF7, HL60, THP-I, Jurkat, HCT-116, HT-29, PC-3M, SK-MEL-28, Molt-4, H460 and ACHN cells were purchased from ATCC (American Type Culture Collection, USA).
[00349] Standard 72h MTT (thiazolye blue; 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide) assays were performed. Briefly, the cell lines used in this study were recovered and cultured according to the conditions recommended. 4000 cells per well were seeded into 96-well plates according to doubling time and incubated overnight at 37°C. Compounds were prepared in DMSO and a 1/2 dilution series in 100 μl cell culture media, added to cells (in duplicates) with the final DMSO concentration of 0.025%, and incubated for 72 hrs at 37°C. MTT solution (5 mg/ml) was added at 10 μl per well and incubated in the dark at 37°C for 4 hr. Culture medium and MTT solution was removed. MTT dye was solubilised with 100 μl per well of DMSO with agitation. Absorbance was read at 562 nm and data analyzed using Sigmoid curve-fitting software Origin 6.0 (MicroCal, LLC, USA) to determine IC50 value. Antiproliferative activity of representative compounds disclosed herein is presented in Table II.
Table II, Antiproliferative activity
Figure imgf000055_0001
Figure imgf000055_0002
Figure imgf000056_0001
ND-Not Determined
Example 30 - Xenograft Studies
[00350] Female BALB/c nude mice from SHANGHAI SLAC LABORATORY ANIMAL CO with age of 5 to 6 weeks and body weight of 19 to 20 g were used in the study. Autoclaved water and irradiated food were provided ad libitum, and animals were maintained on a 12-hour light and 12-hour dark cycle. Cages, bedding, and water bottles were autoclaved before use and changed weekly. The health of all animals was determined by daily gross observation of experimental animals, and analyses of blood samples of sentinel animals housed on shared shelf racks. All animals were allowed to acclimate and recover from any shipping-related stress for a minimum of 72 hours before experimental use. All animal experiments were conducted according to the guidelines of Chinese.
[00351] Xenograft tumor models were established with human tumor cell lines. HT-29 (Human colon cancer cell),H460 (Human large cell lung cancer cell line), Molt-4 (Human acute lymphoblastic leukemia cell line), PC-3M (Human prostate adenocarcinoma cell line) and HL60 (Human acute promyelocytic leukemia cell line),purchased from ATCC were used in this study. Briefly, mice were anesthetized by gas anesthesia (3% isoflurane), then tumor cells diluted in 100 μl DPBS mixed with Matrigel 1 : 1 were injected subcutaneously into the dorsal region near the thigh of the mouse. Tumor growth was monitored twice per week until the tumor reached to a volume of 100 mm3. The day before the first administration of a compound, tumor bearing animals were stratified into several groups according to tumor volume.. Only animals with appropriate tumor volumes were selected and randomly distributed to treatment and control groups. Each group consisted of 8 mice.
[00352] The test compounds were dissolved in a buffer containing 5%NMP, 15%PEG400 and 80% 0. IM tartrate buffer (pH =4.0). Solutions were prepared fresh daily. The test compounds were given by i.p, qd. The test compounds were administrated at dosage of 12.5mg/kg, 25mg/kg/day, 50mg/kg/day and lOOmg/kg/day over 21 days. Control mice received vehicle using the same administration schedule as that of the test compound.
[00353] Mortality checks were conducted daily. Body weight of the mice was determined twice a week .All animals were individually followed throughout the experiment. Weight loss was calculated as percentage change in mean group body weight using the following formula: [(W - WO) / WO] x 100, where 'W' represents mean body weight of the treated group at a particular day and 'WO' represents mean body weight of the same treated group at initiation of treatment. Tumor volumes (in mm3) were calculated after 21 days drug administration. The calculation was used as the following ellipsoid formula: [D x (d2)] / 2, where 'D' represents the large diameter of the tumor and 'd' represents the small diameter. Statistical analysis is done using the rank sum test and one-way ANOVA and a post hoc Bonferroni t test (SigmaStat version 2.0, Jandel Scientific, San Francisco, CA). Differences between groups are considered significant when P < 0.05. [00354] Administrations of compound 18 and compound 102 in xenograft mice showed anti-tumor activities at the dosage from 12.5mg/kg tolOOmg/kg. The effects were significant with p<0.05 (T-test).
[00355] The synergistic action or enhancement of conventional chemotherapeutic agent by a test compound can also be determined with this model by comparing animals treated with the standard therapy alone to animals treated with test compound plus the same standard therapy. An additive effect on the delay of tumor growth will be observed if synergistic action due to test compound is occurring.
Example 31- Pharmacokinetic Determination
[00356] Male Sprague-Dawley rats were maintained on food and water ad libitum. They were housed in an individual ventilated caging system. The test compound at a dose of 5 mg/kg and 20mg/kg was given i.v. and orally to SD rats respectively (3 animals per group). The appropriate quantity compound with free base (Wt.: 346.1) was accurately weighted into a tube and dissolved in NMP, vortexed to get a clear solution. 0.015N HCl in Saline was added and the solution was vortexed to yield a clear and colorless solution at 1 mg/mL for both intravenous and oral administration.
The concentration of the test compound in the dosing solution was reconfirmed by HPLC method. Then the test compound (10, 100 mg/kg) was given orally in a volume of 0.1 mL/10 g body weight to SD rats. Plasma samples were collected from retro-orbital puncture at pre-dose and 0.083, 0.25, 0.5, 1, 2, 4, 6, 8, and 24 hours post-dose. All samples and the dose formulation were stored at -200C until bioanalysis. The concentrations of the test compound in plasma were determined using a high performance liquid chromatography/mass spectrometry (HPLC/MS/MS) method.
Example 32 - Pancreatic Cancer Clinical Trial
Length of Study
[00357] 8 months [length of time from FPV to LPV]
Objectives
[00358] The primary objective of this study is to determine the objective response rate (ORR) for compound of
Formula (I) when administered every 2 weeks to patients with adenocarcinoma of the pancreas. The secondary objectives of this study is to measure time-to-event variables including: time to objective tumor response for responding patients (TtOR), duration of response for responding patients, time to treatment failure (TtTF), time to progressive disease (TtPD), progression-free survival (PFS), overall survival (OS); the toxicities of therapy.
Study Design
[00359] The study is a multi-center, double-blind, randomized, placebo-controlled Phase 2 study. Tumor assessments will be repeated every 4 cycles (approximately 8 weeks). Patients will receive study therapy for 12 treatments, or until tumor progression is documented, unacceptable toxicity is experienced, the patient withdrew consent, or the patient is unable to fulfill the responsibilities of study participation as determined by the treating physician or the qualified investigator. After study discontinuation, patients who have not progressed will have tumor assessments performed approximately every 8 weeks until disease progression. Once patients have disease progression, patients will enter a post-study follow-up period, and will be followed every 12 weeks for 24 months for overall survival. Patients will also be followed for ongoing or any new toxicities.
Diagnosis and Main Criteria for Inclusion:
[00360] Male and females > 18 years of age will be eligible for this study if they are diagnosed with adenocarcinoma of the pancrease. Patients must have had as their initial presentation pancreatic metastasis without evidence of pulmonary metastasis.
[00361] Main inclusion criteria will include: histologically proven adenocarcinoma; performance Status of 0 or 1 on the Eastern Cooperative Oncology Group (ECOG) scale; a complete history and physical, chest x-ray, CT scan of abdomen and pelvis; barium enema, or colonoscopy. Patients with pain will be requires to have had their pain stabilized for 1 week prior to commencing therapy. Patients requiring opioids for pain control will be required to have been on a fixed analgesic regimen aimed to provide adequate pain control with no more than three breakthrough
(supplemental) doses of analgesics per day to control pain. Patients will be requires to demonstrate adequate bone marrow reserve (i.e. Neutrophil count > 1.5 x 109 cells/L; Platelets ≥ 100 x 109 cells/L). Patients will be required to have negative tumor markers for alpha-fetoprotein (AFP) and monoclonal antichorionic gonadotropin (β-subunit)
(βHCG). Patients will be required to demonstrate at least one unidimensionally measurable lesion, meeting Response
Evaluation Criteria in Solid Tumors (RECIST). Patient will also be required to have an estimated life expectancy of at least 12 weeks.
[00362] Main exclusion criteria will include: prior chemotherapy; pregnancy or breastfeeding; inability or unwillingness to take folic acid, vitamin B 12 supplementation, or dexamethasone.
Study Drug, Dose, and Mode of Administration
[00363] Compound of Formula (I) dosage will be 500 mg/m2 and will be given as a 10-minute infusion on Day 1 of each 14-day cycle. Folic acid and vitamin B 12 supplementation, and dexamethasone (or equivalent corticosteroid) prophylaxis will also be administered.
Variables
[00364] Efficacy: Tumor response rate will be defined as the number of patients with documented partial response (PR) or complete response (CR) divided by the number of patients qualified for tumor response analysis. Time-to-event analyses will be performed on the observed distributions of time to objective progressive disease, progression-free survival (PFS), time to treatment failure (TtTF), and overall survival (OS) using the Kaplan-Meier (K-M) method. All patients with best overall response of CR or PR will be analyzed for response duration by using the K-M method.
[00365] Safety: Safety analyses will include adverse event (AE) rates, serious AEs, vital signs, laboratory data, blood transfusions required, and deaths. Toxicities using laboratory and non-laboratory adverse events will be evaluated using the common terminology criteria for adverse events (CTCAE, version 3.0).
Evaluation Methods
[00366] Statistical: The primary analysis will be to estimate the objective best overall response rate and its 95% confidence interval (CI). Medians for each of the time-to-event endpoints, and time-to-event variables will be estimated using the K-M method. All estimates of treatment effects will be conducted at a two-sided alpha level of 0.05, and CI for all parameters will be estimated were to be constructed using a 95% level.
Example 33 - Clinical Trial Protocol of Compound of Formula (I) in Patients With Previously Untreated
Advanced Soft Tissue Sarcoma
Patient eligibility
[00367] Patients with documented metastatic or locally advanced soft tissue sarcoma, not curable by other means, are eligible for this trial. Eligibility criteria were as follows: histologically documented soft tissue sarcoma; presence of at least one site of unidimensionally measurable clinically and/or radiologically documented disease; age > 18 years, life expectancy of at least 12 weeks; Eastern Cooperative Oncology Group performance status < 2; and no prior systemic therapy for metastatic disease. Prior adjuvant chemotherapy is permitted as long as treatment had been completed > 6 months since the last dose of therapy. Patients could not have received radiotherapy to the sole site of measurable disease unless it was a current site of progressive disease, and no more than 25% of functioning bone marrow could have been irradiated. Previous surgery is allowed if > 4 weeks prior to initiating treatment. Laboratory requirements include the following: absolute granulocyte count > 1.5 x 10 /L; platelet count > 100>< 109/L; AST < 2.5χ upper normal limit (UNL); serum creatinine < UNL; bilirubin < UNL. All patients must give informed consent according to the requirements of their local Institutional and/or University Human Experimentation Committee. Treatment
[00368] Compound of Formula (I) is prepared as a sterile 10 mg/ml solution. The individually calculated dose is diluted prior to infusion with 0.9% sodium chloride injection USP or 5% dextrose injection USP to final concentrations ranging from 0.09 to 0.5 mg/ml of compound of Formula (I).
[00369] Compound of Formula (I) is administered in the outpatient setting as a 1 hour infusion at a dose of 50 mg/m2 daily χ3 days every 21 days. Vital signs are monitored every 30 minutes χ5 beginning at the start of infusion until 1 hour post infusion. Antidiarrheal prophylaxis and antiemetic prophylaxis are prescribed as follows: 4 chewable Pepto Bismol tablets 1 hour before first dose of compound of Formula (I), then 2 tablets every 6 hours until 12 hours after the last dose of compound of Formula (I) (day 3), ondansetron 8 mg orally every 12 hours beginning 12 hours before treatment and continuing until 12 hours after the last dose of compound of Formula (I) (day 3). Loperamide is started if diarrhea occurred, using a dose regimen of 2 mg orally q.2 h while awake and q.4 h during sleep until 12 h diarrhea- free. Dose reductions are allowed (dose level: 1 = 37.5 mg/m2/d, dose level: 2 = 28 mg/m2/d), if the following toxicities are seen: diarrhea > grade 3 associated with mucus or dehydration; nausea and vomiting > grade 3 despite antiemetics; granulocyte nadir < 0.5 x 109/L and/or neutropenia with fever or infection; platelet nadir < 25 x 109/L and/or thrombocytopenic bleeding; or any nonhematologic toxicity (except alopecia) > grade 3. Patients requiring more than two dose reductions are removed from protocol treatment.
[00370] Treatment continues until progression, or for 2 cycles after complete or stable partial response. Nonresponding stable patients continue on treatment until progression, or alternatively are removed from therapy after 6 cycles at the investigator's discretion. After termination from protocol treatment, all patients are seen at 4 weeks, then every 3 months until progression or death. Response and toxicity assessment
[00371] Patients are considered evaluable for response if they have received at least 1 cycle of therapy and have their disease reevaluated. Patients are evaluated for response every 6 weeks (every 2 cycles) using the same investigations that demonstrated measurable disease at baseline. Response and progression are evaluated using the RECIST criteria (response evaluation criteria in solid tumors) (Therasse P, Arbuck SG, Eisenhauer EA, et al. New guidelines to evaluate the response to treatment in solid tumors (RECIST guidelines). Journal of the National Cancer Institute. 2000;92(3):205-216).
[00372] Patients are considered evaluable for toxicity after their first infusion of compound of Formula (I). A history and physical examination is performed on day 1 of each cycle with an assessment of toxicity during the previous treatment interval. CBC, platelets, differential, bilirubin and AST are performed on days 1, 8, and 15, and BUN, creatinine, electrolytes, LDH, fasting glucose, and alkaline phosphatase are performed on day 1 of each cycle. Toxicities are graded according to the NCI Common Toxicity Criteria Version 2.0. Statistical analysis
[00373] The primary endpoint of this phase II study is objective response rate, with secondary endpoints of toxicity, time to progression, early progression rate, and response duration. In order to minimize the expected number of patients treated in the event that the regimen proves to be very disappointing or very successful, a two-stage design is used for patient accrual. This design tests the null hypothesis (H0) that the true response rate is < 5% versus the alternative hypothesis (HA) that the true response rate is > 20%. The significance level (i.e., the probability of rejecting H0 when it is true) is 0.058. The power is 0.865 when the true response probability is 20%. If there were no responses after the first 15 patients, the response rate is concluded to be < 5% and accrual was to be stopped. If there were one or more responses, accrual is continued to 30 evaluable patients. Example 34- Metastatic Adenocarcinoma of the Prostate Clinical Trial Eligibility
[00374] All patients are required to have histologically confirmed metastatic adenocarcinoma of the prostate that was progressive despite an adequate trial of hormonal therapy. Patients are ambulatory with an Eastern Cooperative Group performance status of 0-2 and have an estimated life expectancy of at least 3 months. One prior secondary hormonal agent is allowed, but it must have been discontinued at least 4 weeks before to exclude an antiandrogen withdrawal response. No prior chemotherapy, experimental therapy, strontium, or suramin therapy is allowed. Prior radiation therapy is permitted provided that at least 4 weeks had elapsed from the day of the last treatment. Patients who did not have an orchiectomy are continued on their luteinizing hormone-releasing hormone injections, but no other hormonal agent is allowed while on study. Other requirements include: adequate bone marrow function with absolute neutrophil count ^ l 200/μl, hemoglobin iδ g/day, and platelets i l00,000/μl; stable renal function with creatinine ≤ 1.5 upper limits of normal; and adequate hepatic reserve with bilirubin £1.5 and asparatase aminotransferase:^.5 times the upper limit of normal. A prostate-specific antigen (PSA) ^: 10 ng/ml is required if the patient has bone only metastases. No serious concurrent illnesses, previous or concurrent malignancy (except nonmelanoma skin cancer treated with curative intent; other cancers allowed only if the patient has been disease free for 5 years), or history of central nervous system metastasis is allowed. A written informed consent is obtained from all patients before registration. Treatment.
[00375] On the basis of Phase I trial, compound of Formula (I) is initially administered via a battery-powered ambulatory infusion pump connected to a central venous catheter as a 72 -h continuous infusion at the starting dose of 40 mg/m2/day. The compound of Formula (I) cassette is changed daily with courses repeated every 14 days until disease progression or overt toxicity. For patients completing two courses at 40 mg/m2/day without ϋ grade 3 toxicity (except alopecia), their dose could be escalated to 50 mg/m2/day at the discretion of the investigator. If the subsequent two courses are again tolerated, another dose escalation to 60 mg/m2/day could be performed. All patients who are undergoing dose escalation are required to use antidiarrheal prophylaxis with loperamide with/without questran. For all patients experiencing a grade 12 increase in creatinine, grade ≥4 hematological toxicity, or other grade ≥3 toxicity, the infusion is held until resolution of the toxicity to grade 1.1, at which time, the drug is resumed at the next lowest dose level (the lowest level allowed was 30 mg/m2/day). If the toxicity does not resolve by day 14, the patient is removed from the study. For nausea/vomiting and diarrhea, no dose reduction is performed unless the toxicities persisted despite maximal antiemetic and antidiarrheal prophylaxis. No dose re-escalation is allowed. End Points
[00376] The primary end points of this study are response rate and toxicity. After the baseline assessments, the complete blood counts and serum chemistries are repeated weekly for the first month, then at the beginning of each new course (every 2 weeks) of therapy. Vitals signs, performance status, and toxicity are also assessed weekly for the first month and then at the beginning of each additional course of therapy as well. PSA is obtained every 4 weeks, and computed tomography scans and/or bone scans were repeated every 12 weeks while on study. All patients receiving compound of Formula (I) are considered evaluable for toxicity (using National Cancer Institute Common Toxicity Criteria, version 1.0), but only those patients completing at least four courses of therapy (8 weeks) are considered evaluable for response. Any patients receiving less than four cycles of therapy (unevaluable for response) are replaced because it is felt that <8 weeks of drug is inadequate to evaluate for any potential objective response using a cytostatic agent. A CR is defined as disappearance of all measurable disease and normalization of the bone scan for 4 weeks. A PR is defined as ^50% decrease from baseline in the sum of the products of the maximum perpendicular diameters for indicator lesions with no progressive disease observed for at least 4 weeks. For patients with bone scan only abnormalities, a PR requires _.50% decrease in the number of lesions. Stable disease (SD) is defined as the absence of a CR, PR, or disease progression. Progressive disease (PD) is defined as unequivocal increase of at least 25% in the size of any measurable lesion or the appearance of any new lesion. Serum PSA levels are not used in the response assessment.
Statistical Methods.
[00377] Progression-free survival (PFS) is used as the primary end point. Survival probabilities were calculated by the method of Kaplan and Meier (Kaplan E., Meier R. Nonparametric estimation from incomplete observations. J. Am.
Stat. Assoc, 53: 457-481, 1958).
Example 35: Pharmaceutical Compositions Example 35a: Parenteral Composition
[00378] To prepare a parenteral pharmaceutical composition suitable for administration by injection, 100 mg of a water-soluble salt of a compound of Formula (I) is dissolved in DMSO and then mixed with 10 mL of 0.9% sterile saline. The mixture is incorporated into a dosage unit form suitable for administration by injection.
Example 35b: Oral Composition
[00379] To prepare a pharmaceutical composition for oral delivery, 100 mg of a compound of Formula (I) is mixed with 750 mg of starch. The mixture is incorporated into an oral dosage unit for, such as a hard gelatin capsule, which is suitable for oral administration.
Example 35c: Sublingual (Hard Lozenge) Composition
[00380] To prepare a pharmaceutical composition for buccal delivery, such as a hard lozenge, mix 100 mg of a compound of Formula (I) with 420 mg of powdered sugar mixed, with 1.6 mL of light corn syrup, 2.4 mL distilled water, and 0.42 mL mint extract. The mixture is gently blended and poured into a mold to form a lozenge suitable for buccal administration.
Example 35d: Fast-Disintegrating Sublingual Tablet
[00381] A fast-disintegrating sublingual tablet is prepared by mixing 48.5% by weigh of a compound of Formula (I), 44.5% by weight of microcrystalline cellulose (KG-802), 5% by weight of low-substituted hydroxypropyl cellulose (50 μm), and 2% by weight of magnesium stearate. Tablets are prepared by direct compression (AAPS PharmSciTech. 2006;7(2):E41). The total weight of the compressed tablets is maintained at 150 mg. The formulation is prepared by mixing the amount of compound of Formula (I) with the total quantity of microcrystalline cellulose (MCC) and two- thirds of the quantity of low-substituted hydroxypropyl cellulose (L-HPC) by using a three dimensional manual mixer (lnversina ®, Bioengineering AG, Switzerland) for 4.5 minutes. All of the magnesium stearate (MS) and the remaining one-third of the quantity of L-HPC are added 30 seconds before the end of mixing.
Example 35e: Inhalation Composition
[00382] To prepare a pharmaceutical composition for inhalation delivery, 20 mg of a compound of Formula (I) is mixed with 50 mg of anhydrous citric acid and 100 mL of 0.9% sodium chloride solution. The mixture is incorporated into an inhalation delivery unit, such as a nebulizer, which is suitable for inhalation administration.
Example 35f: Rectal Gel Composition
[00383] To prepare a pharmaceutical composition for rectal delivery, 100 mg of a compound of Formula (I) is mixed with 2.5 g of methylcelluose (1500 mPa), 100 mg of methylparapen, 5 g of glycerin and 100 mL of purified water. The resulting gel mixture is then incorporated into rectal delivery units, such as syringes, which are suitable for rectal administration.
Example 35g: Topical Gel Composition [00384] To prepare a pharmaceutical topical gel composition, 100 mg of a compound of Formula (I) is mixed with 1.75 g of hydroxypropyl cellulose, 10 mL of propylene glycol, 10 niL of isopropyl myristate and 100 niL of purified alcohol USP. The resulting gel mixture is then incorporated into containers, such as tubes, which are suitable for topical administration.
Example 35h: Ophthalmic Solution Composition
[00385] To prepare a pharmaceutical opthalmic solution composition, 100 mg of a compound of Formula (I) is mixed with 0.9 g of NaCl in 100 mL of purified water and filtered using a 0.2 micron filter. The resulting isotonic solution is then incorporated into ophthalmic delivery units, such as eye drop containers, which are suitable for ophthalmic administration.
Example 35i: Nasal spray solution
[00386] To prepare a pharmaceutical nasal spray solution, 10 g of a compound of Formula (I) is mixed with 30 mL of a 0.05M phosphate buffer solution (pH 4.4). The solution is placed in a nasal administrator designed to deliver 100 μl of spray for each application.
[00387] The examples and embodiments described herein are for illustrative purposes only and various modifications or changes suggested to persons skilled in the art are to be included within the spirit and purview of this application and scope of the appended claims.

Claims

WHAT IS CLAIMED IS:
1. A compound, or pharmaceutically acceptable salt, solvate, or prodrug thereof, having the structure of Formula
(Ia):
Figure imgf000063_0001
Formula (Ia), wherein:
X is -C(R3)- or -N-;
R1 is unsubstituted Ci-C6 alkyl, unsubstituted or substituted aryl, or unsubstituted or substituted heteroaryl;
R2, R3, R5, R6, R7, R8, R9, R10 and R11 are each independently selected from the group consisting of H, halogen, -
NH2, -NO2, -CN, -OH, -CO2H, -CONH2, -SO3H, -SO2NH2, -SO2CH3, -OCH3, -CF3, -CH3, and -LA-LB-R32; LA is a covalent bond or an alkyl group; LB is a covalent bond, -0-, -NR32-, -NH-, -C(=0)-, -C(=0)0-, -0C(=0)-, -C(=O)NR32-, -C(=0)NH-, -NR32C(=O)-,
-NHC(=0)-, -SO2-, -SO2NR32-, - NR32SO2-, -SO2NH-, or -NHSO2-; R32 is H, substituted or unsubstituted alkyl, haloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted non-aromatic heterocycle, or substituted or unsubstituted cycloalkyl; wherein any R32 group, when substituted, is substituted with one or more groups selected from halogen, -NH2,
-NO2, -CN, -OH, -CO2H, -CONH2, -SO3H, -SO2NH2, -SO2CH3, -OCH3, and -CF3; Z is -NRaC(=0)-, -C(=0)NRa-, -NR3SO2-, -SO2NR3-, -NR3-, -CH2NR3-, - NR3CH2-, -CH2-, -CH2CH2-, -CH=CH-,
-CH2C(=0)NR3-, -NR3C(=0)CH2-, -SO2-, or -SO-; R3 is H or alkyl.
2. The compound of Claim 1, wherein: X is N
3. The compound of Claim 1, wherein: X is -C(R3)-.
4. The compound of Claim 1, wherein: Z is -NR3-.
5. The compound of Claim 1, wherein:
R1 is unsubstituted Ci-C6 alkyl or unsubstituted or substituted phenyl; each R3 is independently selected from the group consisting of H, halogen, -NH2, -NO2, -CN, -OH, -CO2H, -
CONH2, -SO3H, -SO2NH2, -SO2CH3, -OCH3, -CF3, and -CH3; R2, R5 and R6 are each H; R7, R8, R9, R10 and R11 are each independently selected from the group consisting of H, halogen, -NH2, -NO2, -
CN, -OH, -CO2H, -CONH2, -SO3H, -SO2NH2, -SO2CH3, -OCH3, -CF3, -CH3, or -LA-LB-R32, LA is a covalent bond or an alkyl group;
LB is a covalent bond, -0-, -NR32-, -NH-, -C(=0)0-, -C(=O)NR32-, -C(=0)NH-, -SO2NR32-, or -SO2NH-; R32 is H, substituted or unsubstituted alkyl, haloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted non-aromatic heterocycle, or substituted or unsubstituted cycloalkyl; wherein any R32 group, when substituted, is substituted with one or more groups selected from halogen, -NH2, -NO2, -CN, -OH, -CO2H, -CONH2, -SO3H, -SO2NH2, -SO2CH3, -OCH3, and -CF3.
6. A compound, or pharmaceutically acceptable salt, solvate, or prodrug thereof, having the structure of Formula
(Ib):
Figure imgf000064_0001
Formula (Ib), wherein:
R1, R2, R3, R5, R6, R7, R8, R9, R10 and R11 are each independently selected from the group consisting of H, halogen,
-NH2, -NO2, -CN, -OH, -CO2H, -CONH2, -SO3H, -SO2NH2, -SO2CH3, -OCH3, -CF3, -CH3, and -LA-LB-R32; LA is a covalent bond or an alkyl group; LB is a covalent bond, -0-, -NR32-, -NH-, -C(=0)-, -C(=0)0-, -0C(=0)-, -C(=O)NR32-, -C(=0)NH-, -NR32C(=O)-,
-NHC(=0)-, -SO2-, -SO2NR32-, - NR32SO2-, -SO2NH-, or -NHSO2-; R32 is H, substituted or unsubstituted alkyl, haloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted non-aromatic heterocycle, or substituted or unsubstituted cycloalkyl; wherein any R32 group, when substituted, is substituted with one or more groups selected from halogen, -NH2,
-NO2, -CN, -OH, -CO2H, -CONH2, -SO3H, -SO2NH2, -SO2CH3, -OCH3, and -CF3; Z is -NRaC(=0)-, -C(=0)NRa-, -NR3SO2-, -SO2NR3-, -NR3-, -CH2NR3-, - NR3CH2-, -CH2-, -CH2CH2-, -CH=CH-,
-CH2C(=O)NR3-, -NR3C(=O)CH2-, -SO2-, or -SO-; R3 is H or alkyl.
7. The compound of Claim 6, wherein: Z is -NR3-.
8. The compound of Claim 7, wherein: R1 is unsubstituted Ci-C6 alkyl; each R3 is independently selected from the group consisting of H, halogen, -NH2, -NO2, -CN, -OH, -CO2H, - CONH2, -SO3H, -SO2NH2, -SO2CH3, -OCH3, -CF3, and -CH3;
R2, R5 and R6 are each H;
R7, R8, R9, R10 and R11 are each independently selected from the group consisting of H, halogen, -NH2, -NO2, - CN, -OH, -CO2H, -CONH2, -SO3H, -SO2NH2, -SO2CH3, -OCH3, -CF3, -CH3, and -LA-LB-R32,
LA is a covalent bond or an alkyl group;
LB is a covalent bond, -0-, -NR32-, -NH-, -C(=0)0-, -C(=O)NR32-, -C(=0)NH-, -SO2NR32-, or -SO2NH-;
R32 is H, substituted or unsubstituted alkyl, haloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted non-aromatic heterocycle, or substituted or unsubstituted cycloalkyl; wherein any R32 group, when substituted, is substituted with one or more groups selected from halogen, -NH2, -NO2, -CN, -OH, -CO2H, -CONH2, -SO3H, -SO2NH2, -SO2CH3, -OCH3, and -CF3.
9. A compound, or pharmaceutically acceptable salt, solvate, or prodrug thereof, having the structure of Formula (Ic):
Figure imgf000065_0001
Formula (Ic), wherein:
R1, R2, R3, R5, R6, R7, R8, R9, R10 and R11 are each independently selected from the group consisting of H, halogen,
-NH2, -NO2, -CN, -OH, -CO2H, -CONH2, -SO3H, -SO2NH2, -SO2CH3, -OCH3, -CF3, -CH3, and -LA-LB-R32; LA is a covalent bond or an alkyl group; LB is a covalent bond, -0-, -NR32-, -NH-, -C(=0)-, -C(=0)0-, -0C(=0)-, -C(=O)NR32-, -C(=0)NH-, -NR32C(=O)-,
-NHC(=0)-, -SO2-, -SO2NR32-, - NR32SO2-, -SO2NH-, or -NHSO2-; R32 is H, substituted or unsubstituted alkyl, haloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted non-aromatic heterocycle, or substituted or unsubstituted cycloalkyl; wherein any R32 group, when substituted, is substituted with one or more groups selected from halogen, -NH2,
-NO2, -CN, -OH, -CO2H, -CONH2, -SO3H, -SO2NH2, -SO2CH3, -OCH3, and -CF3; Z is -NRaC(=0)-, -C(=0)NRa-, -NR3SO2-, -SO2NR3-, -NR3-, -CH2NR3-, - NR3CH2-, -CH2-, -CH2CH2-, -CH=CH-,
-CH2C(=0)NR3-, -NR3CC=O)CH2-, -SO2-, or -SO-; R3 is H or alkyl.
10. The compound of Claim 9, wherein: Z is -NR3-.
11. The compound of Claim 10, wherein: R1 is unsubstituted Ci-C6 alkyl; each R3 is independently selected from the group consisting of H, halogen, -NH2, -NO2, -CN, -OH, -CO2H, - CONH2, -SO3H, -SO2NH2, -SO2CH3, -OCH3, -CF3, and -CH3;
R2, R5 and R6 are each H;
R7, R8, R9, R10 and R11 are each independently selected from the group consisting of H, halogen, -NH2, -NO2, - CN, -OH, -CO2H, -CONH2, -SO3H, -SO2NH2, -SO2CH3, -OCH3, -CF3, -CH3, and -LA-LB-R32,
LA is a covalent bond or an alkyl group;
LB is a covalent bond, -0-, -NR32-, -NH-, -C(=0)0-, -C(=O)NR32-, -C(=0)NH-, -SO2NR32-, or -SO2NH-;
R32 is H, substituted or unsubstituted alkyl, haloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted non-aromatic heterocycle, or substituted or unsubstituted cycloalkyl; wherein any R32 group, when substituted, is substituted with one or more groups selected from halogen, -NH2, -NO2, -CN, -OH, -CO2H, -CONH2, -SO3H, -SO2NH2, -SO2CH3, -OCH3, and -CF3.
12. A compound selected from:
3 -(4-( 1 -Methyl- l/f-indol-3 -yl)pyrimidin-2-ylamino)phenol (Compound 1)
4-(4-( 1 -Methyl- l/f-indol-3 -yl)pyrimidin-2-ylamino)phenol (Compound 2)
3 -Chloro-4-(4-( 1 -methyl- l/f-indol-3 -yl)pyrimidin-2-ylamino)phenol (Compound 3)
3 -Methyl-4-[4-( 1 -methyl- l/f-indol-3 -yl)-pyrimidin-2-ylamino] -phenol (Compound 4)
3 ,5-Dimethyl-4-[4-( 1 -methyl- l/f-indol-3 -yl)-pyrimidin-2-ylamino] -phenol (Compound 5)
4-[4-( 1 -Ethyl- l/f-indol-3 -yl)-pyrimidin-2-ylamino] -phenol (Compound 6) 3 -[4-( 1 -Ethyl- l/f-indol-3 -yl)-pyrimidin-2-ylamino] -phenol (Compound 7)
4-[4-( 1 -Ethyl- l/f-indol-3 -yl)-pyrimidin-2-ylamino] -3 -methyl -phenol (Compound 8)
4-[4-( 1 -Ethyl- l/f-indol-3 -yl)-pyrimidin-2-ylamino] -3 ,5-dimethyl-phenol (Compound 9)
4-[4-( 1 -Benzyl- l/f-indol-3 -yl)-pyrimidin-2-ylamino] -phenol (Compound 10)
3-[4-(l-Benzyl-l/f-indol-3-yl)-pyrimidin-2-ylamino]-phenol (Compound 11)
4-[4-(l-Ethyl-l/f-pyrrolo[2,3-i]pyridin-3-yl)-pyrimidin-2-ylamino]-phenol (Compound 12)
3 -[4-( 1 -Ethyl- l/f-pyrrolo[2,3 -£]pyridin-3 -yl)-pyrimidin-2-ylamino] -phenol (Compound 13)
[4-( 1 -Ethyl- l/f-pyrrolo[2,3 -£]pyridin-3 -yl)-pyrimidin-2-yl] -(3 -nitro-phenyl)-amine (Compound 14)
4-[4-(l-Propyl-l/f-pyrrolo[2,3-£]pyridin-3-yl)-pyrimidin-2-ylamino]-phenol (Compound 15)
3 -[4-( 1 -Propyl- l/f-pyrrolo[2,3 -£]pyridin-3 -yl)-pyrimidin-2-ylamino] -phenol (Compound 16)
(3 -Nitro-phenyl)-[4-( 1 -propyl- l/f-pyrrolo[2,3 -£]pyridin-3 -yl)-pyrimidin-2-yl] -amine (Compound 17)
3 -(4-( 1 -Ethyl- l/f-pyrrolo[2,3 -£]pyridin-3 -yl)pyrimidin-2-ylamino)benzenesulfonamide (Compound 18)
3 -(4-( 1 -Ethyl- l/f-pyrrolo[2,3 -£]pyridin-3 -yl)pyrimidin-2-ylamino)benzamide (Compound 19)
3 -(4-( 1 -ethyl- l/f-pyrrolo[2,3 -£]pyridin-3 -yl)pyrimidin-2-ylamino)benzonitrile (Compound 20).
4-(4-(l-ethyl-l/f-pyrrolo[2,3-i]pyridin-3-yl)pyrimidin-2-ylamino)benzenesulfonamide (Compound 21).
3 -(4-( 1 -propyl- l/f-pyrrolo[2,3 -£]pyridin-3 -yl)pyrimidin-2-ylamino)benzenesulfonamide (Compound 22).
4-(4-(l-Propyl-l/f-pyrrolo[2,3-i]pyridin-3-yl)pyrimidin-2-ylamino)benzenesulfonamide (Compound 23).
7V-(4-Methyl-3-(moφholinosulfonyl)phenyl)-4-(l-propyl-lH-pyrrolo[2,3-i]pyridin-3-yl)pyrimidin-2 -amine
(Compound 24).
N-(3 -(Piperazin- 1 -ylsulfonyl)phenyl)-4-( 1 -propyl- l/f-pyrrolo[2,3 -£]pyridin-3 -yl)pyrimidin-2-amine
(Compound 25).
7V-(4-(Piperazin- 1 -ylsulfonyl)phenyl)-4-( 1 -propyl- l/f-pyrrolo[2,3 -£]pyridin-3 -yl)pyrimidin-2-amine
(Compound 26).
7V-(4-(Moφholinosulfonyl)phenyl)-4-(l-propyl-l/f-pyrrolo[2,3-i]pyridin-3-yl)pyrimidin-2 -amine (Compound
27).
N-(3 -(4-Methylpiperazin- 1 -ylsulfonyl)phenyl)-4-( 1 -propyl- l/f-pyrrolo[2,3 -i]pyridin-3 -yl)pyrimidin-2 -amine
(Compound 28).
7V-(4-(4-Methylpiperazin- 1 -ylsulfonyl)phenyl)-4-( 1 -propyl- l/f-pyrrolo[2,3 -i]pyridin-3 -yl)pyrimidin-2 -amine
(Compound 29).
7V-(3-Moφholinophenyl)-4-(l-propyl-l/f-pyrrolo[2,3-b]pyridin-3-yl)pyrimidin-2 -amine (Compound 30).
7V-(4-Moφholinophenyl)-4-(l-propyl-l/f-pyrrolo[2,3-b]pyridin-3-yl)pyrimidin-2 -amine (Compound 31).
N-(3 -(Piperazin- 1 -yl)phenyl)-4-( 1 -propyl- l/f-pyrrolo[2,3 -b]pyridin-3 -yl)pyrimidin-2 -amine (Compound 32).
7V-(4-(Piperazin-l-yl)phenyl)-4-(l-propyl-l/f-pyrrolo[2,3-i]pyridin-3-yl)pyrimidin-2 -amine (Compound 33).
(4-Methylpiperazin- l-yl)(3 -(4-( 1 -propyl- l/f-pyrrolo[2,3 -b]pyridin-3 -yl)pyrimidin-2- ylamino)phenyl)methanone (Compound 34).
7V-(4-(4-Methylpiperazin- 1 -yl)phenyl)-4-( 1 -propyl- l/f-pyrrolo[2,3 -i]pyridin-3 -yl)pyrimidin-2-amine
(Compound 35).
1 -(4-(3 -(4-( 1 -Propyl- l/f-pyrrolo[2,3 -i]pyridin-3 -yl)pyrimidin-2-ylamino)phenyl)piperazin- 1 -yl)ethanone
(Compound 36).
1 -(4-(4-(4-( 1 -Propyl- l/f-pyrrolo[2,3 -i]pyridin-3 -yl)pyrimidin-2-ylamino)phenyl)piperazin- 1 -yl)ethanone
(Compound 37). 4-(4-(5-Chloro-7-methyl-l-propyl-l/f-pyrrolo[3,2-i]pyridin-3-yl)pyrimidin-2-ylamino)phenol (Compound
38).
4-(4-(5-Chloro-7-methyl-l-propyl-lH-pyrrolo[3,2-i]pyridin-3-yl)pyrimidin-2-ylamino)benzenesulfonamide
(Compound 39)
3 -(4-(5-Chloro-7 -methyl- 1 -propyl- l/f-pyrrolo[3 ,2-£]pyridin-3 -yl)pyrimidin-2-ylamino)phenol (Compound
40).
4-(4-(5-Chloro-7 -methyl- 1 -propyl- l/f-pyrrolo[3 ,2-i]pyridin-3 -yl)pyrimidin-2-ylamino)-3 -methylphenol
(Compound 41).
3 -(4-(5-Chloro-7 -methyl- 1 -propyl- l/f-pyrrolo[3 ,2-£]pyridin-3 -yl)pyrimidin-2-ylamino)benzenesulfonamide
(Compound 42)
4-(5-Chloro-7-methyl-l-propyl-l/f-pyrrolo[3,2-ft]pyridin-3-yl)-Λ^-(4-methyl-3-
(moφholinosulfonyl)phenyl)pyrimidin-2 -amine (Compound 43)
4-(5-Chloro-7 -methyl- 1 -propyl- l/f-pyrrolo[3 ,2-£]pyridin-3-yl)-7V-(3 -(piperazin- 1 - ylsulfonyl)phenyl)pyrimidin-2 -amine (Compound 44)
4-(5-Chloro-7 -methyl- 1 -propyl- l/f-pyrrolo[3 ,2-£]pyridin-3-yl)-7V-(4-(piperazin- 1 - ylsulfonyl)phenyl)pyrimidin-2 -amine (Compound 45)
4-(5-Chloro-7 -methyl- 1 -propyl- l/f-pyrrolo[3 ,2-£]pyridin-3-yl)-7V-(3 -(piperazin- 1 -yl)phenyl)pyrimidin-2- amine (Compound 46).
4-(5-Chloro-7-methyl-l-propyl-l/f-pyrrolo[3,2-i]pyridin-3-yl)-7V-(4-(piperazin-l-yl)phenyl)pyrimidin-2- amine (Compound 47)
4-(5-Chloro-7-methyl-l-propyl-l/f-pyrrolo[3,2-i]pyridin-3-yl)-7V-(3-moφholinophenyl)pyrimidin-2 -amine
(Compound 48).
4-(5-Chloro-7-methyl-l-propyl-l/f-pyrrolo[3,2-i]pyridin-3-yl)-7V-(4-moφholinophenyl)pyrimidin-2 -amine
(Compound 49).
1 -(4-(3 -(4-(5-Chloro-7 -methyl- 1 -propyl- l/f-pyrrolo[3 ,2-£]pyridin-3 -yl)pyrimidin-2- ylamino)phenyl)piperazin-l-yl)ethanone (Compound 50).
1 -(4-(4-(4-(5-Chloro-7 -methyl- 1 -propyl- l/f-pyrrolo[3 ,2-£]pyridin-3 -yl)pyrimidin-2- ylamino)phenyl)piperazin-l-yl)ethanone (Compound 51).
(3 -(4-(5-Chloro-7 -methyl- 1 -propyl- l/f-pyrrolo[3 ,2-£]pyridin-3 -yl)pyrimidin-2- ylamino)phenyl)(moφholino)methanone (Compound 52).
(4-(4-(5-Chloro-7 -methyl- 1 -propyl- l/f-pyrrolo[3 ,2-£]pyridin-3 -yl)pyrimidin-2- ylamino)phenyl)(moφholino)methanone (Compound 53).
3 -(4-(7 -Methyl- 1 -propyl- l/f-pyrrolo[3 ,2-£]pyridin-3 -yl)pyrimidin-2-ylamino)benzenesulfonamide
(Compound 54).
4-(4-(7 -Methyl- 1 -propyl- l/f-pyrrolo[3 ,2-i]pyridin-3 -yl)pyrimidin-2-ylamino)benzenesulfonamide
(Compound 55).
4-(7 -Methyl- 1 -propyl- l/f-pyrrolo[3 ,2-£]pyridin-3 -yl)-7V-(3 -(piperazin- 1 -ylsulfonyl)phenyl)pyrimidin-2 -amine
(Compound 56).
4-(7 -Methyl- 1 -propyl- l/f-pyrrolo[3 ,2-b]pyridin-3 -yl)-7V-(4-(piperazin- 1 -ylsulfonyl)phenyl)pyrimidin-2 -amine
(Compound 57).
4-(7 -Methyl- 1 -propyl- l/f-pyrrolo[3 ,2-£]pyridin-3 -yl)-7V-(3 -(piperazin- 1 -yl)phenyl)pyrimidin-2 -amine
(Compound 58). 4-(7 -Methyl- 1 -propyl- l/f-pyrrolo[3 ,2-£]pyridin-3 -yl)-7V-(4-(piperazin- 1 -yl)phenyl)pyrimidin-2 -amine
(Compound 59).
4-(7 -Methyl- 1 -propyl- lH-pyrrolo[3 ,2-b]pyridin-3 -yl)-7V-(3 -moφholinophenyl)pyrimidin-2 -amine (Compound
60).
4-(7-Methyl-l-propyl-lH-pyrrolo[3,2-b]pyridin-3-yl)-7V-(4-moφholinophenyl)pyrimidin-2 -amine (Compound
61).
1 -(4-(3 -(4-(7 -Methyl- 1 -propyl- l/f-pyrrolo[3 ,2-£]pyridin-3-yl)pyrimidin-2-ylamino)phenyl)piperazin- 1 - yl)ethanone (Compound 62).
1 -(4-(4-(4-(7 -Methyl- 1 -propyl- l/f-pyrrolo[3 ,2-£]pyridin-3-yl)pyrimidin-2-ylamino)phenyl)piperazin- 1 - yl)ethanone (Compound 63).
3 -(4-( 1 -Ethyl-7 -methyl- l/f-pyrrolo[3 ,2-£]pyridin-3 -yl)pyrimidin-2-ylamino)benzenesulfonamide (Compound
64).
4-(4-( 1 -Ethyl-7 -methyl- l/f-pyrrolo[3 ,2-£]pyridin-3 -yl)pyrimidin-2-ylamino)benzenesulfonamide (Compound
65).
(3 -(4-( 1 -Ethyl-7 -methyl- l/f-pyrrolo[3 ,2-£]pyridin-3 -yl)pyrimidin-2-ylamino)phenyl)(moφholino)methanone
(Compound 66).
(4-(4-( 1 -Ethyl-7 -methyl- l/f-pyrrolo[3 ,2-£]pyridin-3 -yl)pyrimidin-2-ylamino)phenyl)(moφholino)methanone
(Compound 67).
4-( 1 -Ethyl-7 -methyl- l/f-pyrrolo[3 ,2-£]pyridin-3 -yl)-7V-(3 -(piperazin- 1 -yl)phenyl)pyrimidin-2 -amine
(Compound 68).
4-( 1 -Ethyl-7 -methyl- l/f-pyrrolo[3 ,2-£]pyridin-3 -yl)-7V-(4-(piperazin- 1 -yl)phenyl)pyrimidin-2 -amine
(Compound 69).
4-(4-(7-Chloro- 1 -ethyl- l/f-pyrrolo[2,3 -c]pyridin-3 -yl)pyrimidin-2-ylamino)phenol (Compound 70)
4-(4-(7-Chloro-l-ethyl-l/f-pyrrolo[2,3-c]pyridin-3-yl)pyrimidin-2-ylamino)benzenesulfonamide (Compound
71)
3 -(4-(7-Chloro- 1 -ethyl- l/f-pyrrolo[2,3 -c]pyridin-3 -yl)pyrimidin-2-ylamino)phenol (Compound 72)
3 -(4-(7-Chloro- 1 -ethyl- l/f-pyrrolo[2,3 -c]pyridin-3 -yl)pyrimidin-2-ylamino)benzenesulfonamide (Compound
73)
4-(7-Chloro- 1 -ethyl- l/f-pyrrolo[2,3 -c]pyridin-3 -yl)-7V-(4-methyl-3 -(moφholinosulfonyl)phenyl)pyrimidin-2- amine (Compound 74)
(3 -(4-(7-Chloro- 1 -ethyl- l/f-pyrrolo[2,3 -c]pyridin-3 -yl)pyrimidin-2-ylamino)phenyl)(moφholino)methanone
(Compound 75)
(4-(4-(7-Chloro-l-ethyl-l/f-pyrrolo[2,3-c]pyridin-3-yl)pyrimidin-2-ylamino)phenyl)(moφholino)methanone
(Compound 76)
4-(7-Chloro- 1 -ethyl- l/f-pyrrolo[2,3 -c]pyridin-3 -yl)-7V-(3 -(piperazin- 1 -ylsulfonyl)phenyl)pyrimidin-2 -amine
(Compound 77)
4-(7-Chloro- 1 -ethyl- l/f-pyrrolo[2,3 -c]pyridin-3 -yl)-7V-(4-(piperazin- 1 -ylsulfonyl)phenyl)pyrimidin-2 -amine
(Compound 78).
4-(7-Chloro- 1 -ethyl- l/f-pyrrolo[2,3 -c]pyridin-3 -yl)-7V-(3 -(piperazin- 1 -yl)phenyl)pyrimidin-2 -amine
(Compound 79).
4-(7-Chloro- 1 -ethyl- l/f-pyrrolo[2,3 -c]pyridin-3 -yl)-7V-(4-(piperazin- 1 -yl)phenyl)pyrimidin-2 -amine
(Compound 80). 4-(7-Chloro- 1 -ethyl- l/f-pyrrolo[2,3 -c]pyridin-3 -yl)-7V-(3 -morpholinophenyl)pyrimidin-2 -amine (Compound
81).
4-(7-Chloro-l-ethyl-l/f-pyrrolo[2,3-c]pyridin-3-yl)-7V-(4-moφholinophenyl)pyrimidin-2 -amine (Compound
82).
3 -(4-( 1 -Ethyl- 1/f-pyrrolo [2,3 -c]pyridin-3 -yl)pyrimidin-2-ylamino)phenol (Compound 83 ).
3-(4-(l-Ethyl-l/f-pyrrolo[2,3-c]pyridin-3-yl)pyrimidin-2-ylamino)benzenesulfonamide (Compound 84).
4-(4-(l-Ethyl-l/f-pyrrolo[2,3-c]pyridin-3-yl)pyrimidin-2-ylamino)phenol (Compound 85).
4-(4-(l-Ethyl-l/f-pyrrolo[2,3-c]pyridin-3-yl)pyrimidin-2-ylamino)benzenesulfonamide (Compound 86).
4-( 1 -Ethyl- l/f-pyrrolo[2,3 -c]pyridin-3 -yl)-7V-(4-methyl-3 -(moφholinosulfonyl)phenyl)pyrimidin-2 -amine
(Compound 87).
(3-(4-(l-Ethyl-l/f-pyrrolo[2,3-c]pyridin-3-yl)pyrimidin-2-ylamino)phenyl)(moφholino)methanone
(Compound 88).
(4-(4-(l-Ethyl-l/f-pyrrolo[2,3-c]pyridin-3-yl)pyrimidin-2-ylamino)phenyl)(moφholino)methanone
(Compound 89).
1 -(4-(3 -(4-( 1 -Ethyl- l/f-pyrrolo[2,3 -c]pyridin-3 -yl)pyrimidin-2-ylamino)benzoyl)piperazin-l -yl)ethanone
(Compound 90).
1 -(4-(4-(4-( 1 -Ethyl- l/f-pyrrolo[2,3 -c]pyridin-3 -yl)pyrimidin-2-ylamino)benzoyl)piperazin-l -yl)ethanone
(Compound 91).
4-( 1 -Ethyl- l/f-pyrrolo[2,3 -c]pyridin-3 -yl)-7V-(3 -(piperazin- l-yl)phenyl)pyrimidin-2 -amine (Compound 92).
4-(l-Ethyl-l/f-pyrrolo[2,3-c]pyridin-3-yl)-7V-(4-(piperazin-l-yl)phenyl)pyrimidin-2 -amine (Compound 93).
4-( 1 -Ethyl- l/f-pyrrolo[2,3 -c]pyridin-3 -yl)-7V-(3 -moφholinophenyl)pyrimidin-2 -amine (Compound 94).
4-( 1 -Ethyl- l/f-pyrrolo[2,3 -c]pyridin-3 -yl)-7V-(4-moφholinophenyl)pyrimidin-2 -amine (Compound 95).
4-(4-(7-Chloro- 1 -propyl- l/f-pyrrolo[2,3 -c]pyridin-3 -yl)pyrimidin-2-ylamino)benzenesulfonamide
(Compound 96).
3 -(4-(7-Chloro- 1 -propyl- l/f-pyrrolo[2,3 -c]pyridin-3 -yl)pyrimidin-2-ylamino)benzenesulfonamide
(Compound 97). l-(4-(3-(4-(7-Chloro-l-propyl-l/f-pyrrolo[2,3-c]pyridin-3-yl)pyrimidin-2-ylamino)benzoyl)piperazin-l- yl)ethanone (Compound 98). l-(4-(4-(4-(7-Chloro-l-propyl-l/f-pyrrolo[2,3-c]pyridin-3-yl)pyrimidin-2-ylamino)benzoyl)piperazin-l- yl)ethanone (Compound 99).
4-(7-Chloro-l-propyl-lH-pyrrolo[2,3-c]pyridin-3-yl)-N-(4-moφholinophenyl)pyrimidin-2 -amine (Compound
100).
4-(7-Chloro- 1 -propyl- l/f-pyrrolo[2,3 -c]pyridin-3 -yl)-7V-(4-methyl-3 -(moφholinosulfonyl)phenyl)pyrimidin-
2-amine (Compound 101).
7V-(4-Methyl-3 -(moφholinosulfonyl)phenyl)-4-( 1 -propyl- l/f-pyrrolo[2,3 -c]pyridin-3 -yl)pyrimidin-2 -amine
(Compound 102).
7V-(4-Moφholinophenyl)-4-(l-propyl-l/f-pyrrolo[2,3-c]pyridin-3-yl)pyrimidin-2 -amine (Compound 103).
N-(3 -(Piperazin- 1 -yl)phenyl)-4-( 1 -propyl- l/f-pyrrolo[2,3 -c]pyridin-3 -yl)pyrimidin-2 -amine (Compound 104).
7V-(4-(Piperazin- 1 -yl)phenyl)-4-( 1 -propyl- l/f-pyrrolo[2,3 -c]pyridin-3 -yl)pyrimidin-2 -amine (Compound 105).
1 -(4-(4-(4-( 1 -Propyl- l/f-pyrrolo[2,3 -c]pyridin-3 -yl)pyrimidin-2-ylamino)benzoyl)piperazin- 1 -yl)ethanone
(Compound 106). Piperazin- 1 -yl(3 -(4-( 1 -propyl- l/f-pyrrolo[2,3 -c]pyridin-3 -yl)pyrimidin-2-ylamino)phenyl)methanone
(Compound 107).
Morpholino(3 -(4-( 1 -propyl- l/f-pyrrolo[2,3 -c]pyridin-3 -yl)pyrimidin-2-ylamino)phenyl)methanone
(Compound 108).
Piperazin- 1 -yl(4-(4-( 1 -propyl- l/f-pyrrolo[2,3 -c]pyridin-3 -yl)pyrimidin-2-ylamino)phenyl)methanone
(Compound 109).
Morpholino(4-(4-( 1 -propyl- l/f-pyrrolo[2,3 -c]pyridin-3 -yl)pyrimidin-2-ylamino)phenyl)methanone
(Compound 110).
3-(4-(l-Propyl-l/f-pyrrolo[2,3-c]pyridin-3-yl)pyrimidin-2-ylamino)benzenesulfonamide (Compound 111).
4-(4-(l-Propyl-l/f-pyrrolo[2,3-c]pyridin-3-yl)pyrimidin-2-ylamino)benzenesulfonamide (Compound 112).
13. The compound of claim 1, 6, 9, or 12, wherein the compound inhibits the activity of at least one kinase selected from among a Cyclin-dependent kinase (CDK), a Glycogen synthase kinase (GSK), an Aurora kinase, a Polo-like kinase (PLK), BCR-ABL, IkappaB kinase complex (IKK), Fms-like tyrosine kinase 3 (FLT3), Janus kinase (JAK), Lymphocyte-specific protein tyrosine kinase (LCK), Platelet-derived growth factor receptor kinase (PDGFR), a Src kinase, and Vascular endothelial growth factor (VEGF) receptor.
14. A pharmaceutical composition comprising a compound of Claim 13 and a pharmaceutically excipient.
15. The pharmaceutical composition of claim 13, wherein the pharmaceutical composition is in the form of an aqueous dispersion, liquid, gel, syrup, elixir, slurry, suspension, aerosol, controlled release formulation, fast melt formulation, effervescent formulation, lyophilized formulation, tablet, powder, pill, dragee, capsule, delayed release formulation, extended release formulation, pulsatile release formulation, multiparticulate formulation, or immediate release formulation.
16. A method for treating or preventing a kinase mediated or kinase dependent disease, disorder or condition in a patient, comprising administrating to the patient an effective amount of a compound of claim 13.
17. The method of claim 16, wherein the kinase is selected from among a Cyclin-dependent kinase (CDK), a Glycogen synthase kinase (GSK), an Aurora kinase, a Polo-like kinase (PLK), BCR-ABL, IkappaB kinase complex (IKK), Fms-like tyrosine kinase 3 (FLT3), Janus kinase (JAK), Lymphocyte-specific protein tyrosine kinase (LCK), Platelet-derived growth factor receptor kinase (PDGFR), a Src kinase, and Vascular endothelial growth factor (VEGF) receptor.
18. The method of claim 17, wherein the kinase mediated or kinase dependent disease, disorder or condition is a proliferative disorder.
19. The method of claim 18, wherein the proliferative disorder is cancer or leukemia.
20. The method of claim 19, wherein the proliferative disorder is cancer or leukemia selected from cervical cancer, colon cancer, prostate cancer, endometrial cancer, osteosarcoma, esophageal cancer, liver cancer, myelogenous leukaemia, melanoma, acute lymphoblastic leukemia, chronic lymphocytic leukaemia, lung cancer, breast cancer, renal cancer, carcinoma of the head and neck.
21. The novel compounds, processes, uses and intermediates substantially as described herein before.
PCT/CN2009/075214 2008-10-27 2009-11-30 Kinase inhibitors and their use as pharmaceutical agents WO2010051781A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US13/095,526 US8309550B2 (en) 2008-10-27 2011-04-27 Kinase inhibitors and their use as pharmaceutical agents

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN200810201791.9 2008-10-27
CN200810201791.9A CN101723936B (en) 2008-10-27 2008-10-27 Kinase suppressor and pharmaceutical application thereof

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/095,526 Continuation US8309550B2 (en) 2008-10-27 2011-04-27 Kinase inhibitors and their use as pharmaceutical agents

Publications (1)

Publication Number Publication Date
WO2010051781A1 true WO2010051781A1 (en) 2010-05-14

Family

ID=42152520

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2009/075214 WO2010051781A1 (en) 2008-10-27 2009-11-30 Kinase inhibitors and their use as pharmaceutical agents

Country Status (3)

Country Link
US (1) US8309550B2 (en)
CN (1) CN101723936B (en)
WO (1) WO2010051781A1 (en)

Cited By (42)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012018909A1 (en) 2010-08-04 2012-02-09 Takeda Pharmaceutical Company Limited Fused heterocyclic compounds
WO2012126181A1 (en) * 2011-03-24 2012-09-27 Ying Luo Use of kinase inhibitors in preventing and treating inflammatory disorder
WO2012127506A1 (en) 2011-03-22 2012-09-27 Advinus Therapeutics Limited Substituted fused tricyclic compounds, compositions and medicinal applications thereof
WO2012104007A3 (en) * 2011-02-01 2013-10-17 Merck Patent Gmbh 7-azaindole derivatives
JP2013544273A (en) * 2011-07-27 2013-12-12 アストラゼネカ アクチボラグ 2- (2,4,5-substituted-anilino) pyrimidine compounds
WO2014063068A1 (en) * 2012-10-18 2014-04-24 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinase 7 (cdk7)
US9012462B2 (en) 2008-05-21 2015-04-21 Ariad Pharmaceuticals, Inc. Phosphorous derivatives as kinase inhibitors
US9180127B2 (en) 2009-12-29 2015-11-10 Dana-Farber Cancer Institute, Inc. Type II Raf kinase inhibitors
US9273077B2 (en) 2008-05-21 2016-03-01 Ariad Pharmaceuticals, Inc. Phosphorus derivatives as kinase inhibitors
WO2016062792A1 (en) * 2014-10-23 2016-04-28 Janssen Pharmaceutica Nv New compounds as nik inhibitors
US9382239B2 (en) 2011-11-17 2016-07-05 Dana-Farber Cancer Institute, Inc. Inhibitors of c-Jun-N-terminal kinase (JNK)
US9505784B2 (en) 2009-06-12 2016-11-29 Dana-Farber Cancer Institute, Inc. Fused 2-aminothiazole compounds
US9593115B2 (en) 2012-09-21 2017-03-14 Advinus Therapeutics Ltd. Substituted fused tricyclic compounds, compositions, and medicinal applications thereof
US9611283B1 (en) 2013-04-10 2017-04-04 Ariad Pharmaceuticals, Inc. Methods for inhibiting cell proliferation in ALK-driven cancers
US9643964B2 (en) 2013-04-24 2017-05-09 Janssen Pharmaceutica Nv 3-(2-aminopyrimidin-4-yl)-5-(3-hydroxypropynyl)-1H-pyrrolo[2,3-C]pyridine derivatives as NIK inhibitors for the treatment of cancer
US9758522B2 (en) 2012-10-19 2017-09-12 Dana-Farber Cancer Institute, Inc. Hydrophobically tagged small molecules as inducers of protein degradation
EP3241830A1 (en) 2016-05-04 2017-11-08 Bayer CropScience Aktiengesellschaft Condensed bicyclic heterocyclic derivatives as pesticides
JP2017533266A (en) * 2014-11-05 2017-11-09 インベンティスバイオ シャンハイ リミテッド Pyrimidine or pyridine-based compounds, production methods thereof and pharmaceutical use
US9834518B2 (en) 2011-05-04 2017-12-05 Ariad Pharmaceuticals, Inc. Compounds for inhibiting cell proliferation in EGFR-driven cancers
US9834571B2 (en) 2012-05-05 2017-12-05 Ariad Pharmaceuticals, Inc. Compounds for inhibiting cell proliferation in EGFR-driven cancers
US9862688B2 (en) 2014-04-23 2018-01-09 Dana-Farber Cancer Institute, Inc. Hydrophobically tagged janus kinase inhibitors and uses thereof
WO2018033455A1 (en) 2016-08-15 2018-02-22 Bayer Cropscience Aktiengesellschaft Condensed bicyclic heterocycle derivatives as pest control agents
US9981963B2 (en) 2013-09-26 2018-05-29 Janssen Pharmaceutica Nv 3-(1H-pyrazol-4-yl)-1H-pyrrolo[2,3-c]pyridine derivatives as NIK inhibitors
US9981962B2 (en) 2014-10-23 2018-05-29 Janssen Pharmaceutica Nv Pyrazole derivatives as NIK inhibitors
US10000483B2 (en) 2012-10-19 2018-06-19 Dana-Farber Cancer Institute, Inc. Bone marrow on X chromosome kinase (BMX) inhibitors and uses thereof
US10005773B2 (en) 2013-09-26 2018-06-26 Janssen Pharmaceutica Nv 1-(4-pyrimidinyl)-1H-pyrrolo[3,2-c]pyridine derivatives as NIK inhibitors
US10017477B2 (en) 2014-04-23 2018-07-10 Dana-Farber Cancer Institute, Inc. Janus kinase inhibitors and uses thereof
US10059690B2 (en) 2014-04-04 2018-08-28 Syros Pharmaceuticals, Inc. Inhibitors of cyclin-dependent kinase 7 (CDK7)
JP2018525431A (en) * 2015-08-31 2018-09-06 ウーシー ショアンリャン バイオテクノロジー カンパニー,リミティド 2-Arylaminopyridine, pyrimidine or triazine derivatives and their production and use
US10221180B2 (en) 2014-10-23 2019-03-05 Janssen Pharmaceutica Nv Pyrazolopyrimidine derivatives as NIK inhibitors
US10323045B2 (en) 2014-10-23 2019-06-18 Janssen Pharmaceutica Nv Thienopyrimidine derivatives as NIK inhibitors
US10508113B2 (en) 2018-03-12 2019-12-17 Abbvie Inc. Inhibitors of tyrosine kinase 2 mediated signaling
US10513509B2 (en) 2016-05-26 2019-12-24 Recurium Ip Holdings, Llc EGFR inhibitor compounds
US10550121B2 (en) 2015-03-27 2020-02-04 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinases
JP2020023554A (en) * 2017-01-26 2020-02-13 ハンミ ファーマシューティカルズ カンパニー リミテッド Pyrimidine compound and medical use thereof
US10702527B2 (en) 2015-06-12 2020-07-07 Dana-Farber Cancer Institute, Inc. Combination therapy of transcription inhibitors and kinase inhibitors
US10870651B2 (en) 2014-12-23 2020-12-22 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinase 7 (CDK7)
US10906889B2 (en) 2013-10-18 2021-02-02 Dana-Farber Cancer Institute, Inc. Polycyclic inhibitors of cyclin-dependent kinase 7 (CDK7)
US11040957B2 (en) 2013-10-18 2021-06-22 Dana-Farber Cancer Institute, Inc. Heteroaromatic compounds useful for the treatment of proliferative diseases
KR20210095621A (en) * 2018-09-21 2021-08-02 상하이 엔노바바이오 파마슈티컬스 씨오 엘티디. Aromatic heterocyclic compounds with kinase inhibitory activity
US11142507B2 (en) 2015-09-09 2021-10-12 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinases
US11471455B2 (en) 2018-10-05 2022-10-18 Annapurna Bio, Inc. Compounds and compositions for treating conditions associated with APJ receptor activity

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104876914B (en) * 2014-02-28 2017-04-19 山东轩竹医药科技有限公司 Pyrimidine derivative type anaplastic lymphoma kinase inhibitor
EP3143015B1 (en) * 2014-05-13 2019-02-20 ARIAD Pharmaceuticals, Inc. Heteroaryl compounds for kinase inhibition
EA034691B1 (en) 2014-06-19 2020-03-06 Ариад Фармасьютикалз, Инк. Heteroaryl compounds for kinase inhibition
ES2901114T3 (en) 2014-08-29 2022-03-21 Tes Pharma S R L Alpha-amino-beta-carboxymuconic acid semialdehyde decarboxylase inhibitors
CN106810553B (en) * 2015-11-30 2020-03-17 江苏正大丰海制药有限公司 3- (4, 5-substituted aminopyrimidine) phenyl derivative and application thereof
CN113166110B (en) * 2018-12-12 2023-08-11 暨南大学 2-aminopyrimidine compound and application thereof
CN114174269B (en) * 2019-07-26 2023-07-18 华润医药控股有限公司 Pyrimidine compounds acting on EGFR and ERBB2
CN112661745A (en) * 2020-07-24 2021-04-16 浙江同源康医药股份有限公司 Compounds useful as CDK7 kinase inhibitors and uses thereof
CN113248500B (en) * 2021-06-10 2021-10-19 中国药科大学 Azaindolopyrimidineamine heterocyclic compounds, and preparation method and application thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1995009847A1 (en) * 1993-10-01 1995-04-13 Ciba-Geigy Ag Pyrimidineamine derivatives and processes for the preparation thereof
CN101014600A (en) * 2004-09-21 2007-08-08 霍夫曼-拉罗奇有限公司 6-(2-alkyl-phenyl) - pyrido[2,3-d] pyrimidines useful as proteine kinase inhibitors
WO2007107221A1 (en) * 2006-03-20 2007-09-27 Merck Patent Gmbh 4-(pyrrolopyridinyl)pyrimidinyl-2-amine derivatives

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB0500492D0 (en) * 2005-01-11 2005-02-16 Cyclacel Ltd Compound
ES2345629T3 (en) * 2005-05-16 2010-09-28 Irm Llc PIRROLOPIRIDINE DERIVATIVES AS PROTEIN KINASE INHIBITORS.
DE102008005493A1 (en) * 2008-01-22 2009-07-23 Merck Patent Gmbh 4- (Pyrrolo [2,3-c] pyridines-3-yl) -pyrimidin-2-yl-amine derivatives
US20110183975A1 (en) * 2008-10-07 2011-07-28 Yasuhiro Goto Novel 6-azaindole aminopyrimidine derivatives having nik inhibitory activity

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1995009847A1 (en) * 1993-10-01 1995-04-13 Ciba-Geigy Ag Pyrimidineamine derivatives and processes for the preparation thereof
CN101014600A (en) * 2004-09-21 2007-08-08 霍夫曼-拉罗奇有限公司 6-(2-alkyl-phenyl) - pyrido[2,3-d] pyrimidines useful as proteine kinase inhibitors
WO2007107221A1 (en) * 2006-03-20 2007-09-27 Merck Patent Gmbh 4-(pyrrolopyridinyl)pyrimidinyl-2-amine derivatives

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
HYUNG-HO HA ET AL.: "Novel heterocycle-substituted pyrimidines as inhibitors ofNF-KB transcription regulation related to TNF-a cytokine release", BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, vol. 18, no. 2, 15 January 2008 (2008-01-15), pages 653 - 656 *

Cited By (82)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9012462B2 (en) 2008-05-21 2015-04-21 Ariad Pharmaceuticals, Inc. Phosphorous derivatives as kinase inhibitors
US9273077B2 (en) 2008-05-21 2016-03-01 Ariad Pharmaceuticals, Inc. Phosphorus derivatives as kinase inhibitors
US9505784B2 (en) 2009-06-12 2016-11-29 Dana-Farber Cancer Institute, Inc. Fused 2-aminothiazole compounds
US11826365B2 (en) 2009-12-29 2023-11-28 Dana-Farber Cancer Institute, Inc. Type II raf kinase inhibitors
US9180127B2 (en) 2009-12-29 2015-11-10 Dana-Farber Cancer Institute, Inc. Type II Raf kinase inhibitors
WO2012018909A1 (en) 2010-08-04 2012-02-09 Takeda Pharmaceutical Company Limited Fused heterocyclic compounds
US9029536B2 (en) 2010-08-04 2015-05-12 Takeda Pharmaceutical Company Limited Fused heterocyclic compounds
EP2746281A3 (en) * 2011-02-01 2014-10-08 Merck Patent GmbH 7-azaindole derivatives
WO2012104007A3 (en) * 2011-02-01 2013-10-17 Merck Patent Gmbh 7-azaindole derivatives
US9725446B2 (en) 2011-02-01 2017-08-08 Merck Patent Gmbh 7-azaindole derivatives
US8981101B2 (en) 2011-02-01 2015-03-17 Merck Patent Gmbh 7-azaindole derivatives
CN103502247A (en) * 2011-02-01 2014-01-08 默克专利股份公司 7-azaindole derivatives
EP3133074A3 (en) * 2011-02-01 2017-05-17 Merck Patent GmbH 7-azaindole derivatives
US9266887B2 (en) 2011-02-01 2016-02-23 Merck Patent Gmbh 7-azaindole derivatives
CN103502247B (en) * 2011-02-01 2016-02-24 默克专利股份公司 7-7-azaindole derivatives
WO2012127506A1 (en) 2011-03-22 2012-09-27 Advinus Therapeutics Limited Substituted fused tricyclic compounds, compositions and medicinal applications thereof
WO2012126181A1 (en) * 2011-03-24 2012-09-27 Ying Luo Use of kinase inhibitors in preventing and treating inflammatory disorder
CN103608014A (en) * 2011-03-24 2014-02-26 罗楹 Use of kinase inhibitors in preventing and treating inflammatory disorder
US9834518B2 (en) 2011-05-04 2017-12-05 Ariad Pharmaceuticals, Inc. Compounds for inhibiting cell proliferation in EGFR-driven cancers
US10858336B2 (en) 2011-07-27 2020-12-08 Astazeneca Ab 2-(2,4,5-substituted-anilino)pyrimidine compounds
JP2013544273A (en) * 2011-07-27 2013-12-12 アストラゼネカ アクチボラグ 2- (2,4,5-substituted-anilino) pyrimidine compounds
JP2014094930A (en) * 2011-07-27 2014-05-22 Astrazeneca Ab 2-(2,4,5-substituted-anilino)pyrimidine compound
US11524951B2 (en) 2011-07-27 2022-12-13 Astrazeneca Ab 2-(2,4,5-substituted-anilino)pyrimidine compounds
EP2736895B1 (en) 2011-07-27 2016-01-06 Astrazeneca AB 2-(2,4,5-substituted-anilino) pyrimidine derivatives as egfr modulators useful for treating cancer
US8946235B2 (en) 2011-07-27 2015-02-03 Astrazeneca Ab 2-(2,4,5-substituted-anilino) pyrimidine compounds
US9732058B2 (en) 2011-07-27 2017-08-15 Astrazeneca Ab 2-(2,4,5-substituted-anilino)pyrimidine compounds
US10017493B2 (en) 2011-07-27 2018-07-10 Astrazeneca Ab 2-(2,4,5-substituted-anilino)pyrimidine compounds
EP3009431B1 (en) 2011-07-27 2017-10-25 Astrazeneca AB 2-(2,4,5-substituted-anilino)pyrimidine derivatives as egfr modulators useful for treating cancer
US9382239B2 (en) 2011-11-17 2016-07-05 Dana-Farber Cancer Institute, Inc. Inhibitors of c-Jun-N-terminal kinase (JNK)
US10981903B2 (en) 2011-11-17 2021-04-20 Dana-Farber Cancer Institute, Inc. Inhibitors of c-Jun-N-terminal kinase (JNK)
US10144730B2 (en) 2011-11-17 2018-12-04 Dana-Farber Cancer Institute, Inc. Inhibitors of c-Jun-N-terminal kinase (JNK)
US9834571B2 (en) 2012-05-05 2017-12-05 Ariad Pharmaceuticals, Inc. Compounds for inhibiting cell proliferation in EGFR-driven cancers
US9593115B2 (en) 2012-09-21 2017-03-14 Advinus Therapeutics Ltd. Substituted fused tricyclic compounds, compositions, and medicinal applications thereof
US10112927B2 (en) 2012-10-18 2018-10-30 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinase 7 (CDK7)
US10787436B2 (en) 2012-10-18 2020-09-29 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinase 7 (CDK7)
WO2014063068A1 (en) * 2012-10-18 2014-04-24 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinase 7 (cdk7)
USRE48175E1 (en) 2012-10-19 2020-08-25 Dana-Farber Cancer Institute, Inc. Hydrophobically tagged small molecules as inducers of protein degradation
US9758522B2 (en) 2012-10-19 2017-09-12 Dana-Farber Cancer Institute, Inc. Hydrophobically tagged small molecules as inducers of protein degradation
US10000483B2 (en) 2012-10-19 2018-06-19 Dana-Farber Cancer Institute, Inc. Bone marrow on X chromosome kinase (BMX) inhibitors and uses thereof
US9611283B1 (en) 2013-04-10 2017-04-04 Ariad Pharmaceuticals, Inc. Methods for inhibiting cell proliferation in ALK-driven cancers
US9643964B2 (en) 2013-04-24 2017-05-09 Janssen Pharmaceutica Nv 3-(2-aminopyrimidin-4-yl)-5-(3-hydroxypropynyl)-1H-pyrrolo[2,3-C]pyridine derivatives as NIK inhibitors for the treatment of cancer
US9981963B2 (en) 2013-09-26 2018-05-29 Janssen Pharmaceutica Nv 3-(1H-pyrazol-4-yl)-1H-pyrrolo[2,3-c]pyridine derivatives as NIK inhibitors
US10005773B2 (en) 2013-09-26 2018-06-26 Janssen Pharmaceutica Nv 1-(4-pyrimidinyl)-1H-pyrrolo[3,2-c]pyridine derivatives as NIK inhibitors
US11040957B2 (en) 2013-10-18 2021-06-22 Dana-Farber Cancer Institute, Inc. Heteroaromatic compounds useful for the treatment of proliferative diseases
US10906889B2 (en) 2013-10-18 2021-02-02 Dana-Farber Cancer Institute, Inc. Polycyclic inhibitors of cyclin-dependent kinase 7 (CDK7)
US10106526B2 (en) 2014-04-04 2018-10-23 Syros Pharmaceuticals, Inc. Inhibitors of cyclin-dependent kinase 7 (CDK7)
US10059690B2 (en) 2014-04-04 2018-08-28 Syros Pharmaceuticals, Inc. Inhibitors of cyclin-dependent kinase 7 (CDK7)
US10017477B2 (en) 2014-04-23 2018-07-10 Dana-Farber Cancer Institute, Inc. Janus kinase inhibitors and uses thereof
US9862688B2 (en) 2014-04-23 2018-01-09 Dana-Farber Cancer Institute, Inc. Hydrophobically tagged janus kinase inhibitors and uses thereof
US10221180B2 (en) 2014-10-23 2019-03-05 Janssen Pharmaceutica Nv Pyrazolopyrimidine derivatives as NIK inhibitors
US10005776B2 (en) 2014-10-23 2018-06-26 Janssen Pharmaceutica Nv Compounds as NIK inhibitors
JP2017531681A (en) * 2014-10-23 2017-10-26 ヤンセン ファーマシューティカ エヌ.ベー. Novel compounds as NIK inhibitors
US9981962B2 (en) 2014-10-23 2018-05-29 Janssen Pharmaceutica Nv Pyrazole derivatives as NIK inhibitors
US10323045B2 (en) 2014-10-23 2019-06-18 Janssen Pharmaceutica Nv Thienopyrimidine derivatives as NIK inhibitors
WO2016062792A1 (en) * 2014-10-23 2016-04-28 Janssen Pharmaceutica Nv New compounds as nik inhibitors
EA030908B1 (en) * 2014-10-23 2018-10-31 Янссен Фармацевтика Нв Compounds as nik inhibitors
KR20170066473A (en) * 2014-10-23 2017-06-14 얀센 파마슈티카 엔.브이. New compounds as nik inhibitors
KR102500071B1 (en) 2014-10-23 2023-02-14 얀센 파마슈티카 엔.브이. New compounds as nik inhibitors
US10822342B2 (en) 2014-10-23 2020-11-03 Janssen Pharmaceutica Nv Pyrazolopyrimidine derivatives as NIK inhibitors
CN107074855A (en) * 2014-10-23 2017-08-18 詹森药业有限公司 It is used as the new compound of NIK inhibitor
JP2017533266A (en) * 2014-11-05 2017-11-09 インベンティスバイオ シャンハイ リミテッド Pyrimidine or pyridine-based compounds, production methods thereof and pharmaceutical use
US11203589B2 (en) 2014-11-05 2021-12-21 InventisBio Co., Ltd. Pyrimidine or pyridine compounds, preparation method therefor and pharmaceutical uses thereof
US11498921B1 (en) 2014-11-05 2022-11-15 InventisBio Co., Ltd. Pyrimidine or pyridine compounds, preparation method therefor and pharmaceutical uses thereof
US10870651B2 (en) 2014-12-23 2020-12-22 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinase 7 (CDK7)
US10550121B2 (en) 2015-03-27 2020-02-04 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinases
US11325910B2 (en) 2015-03-27 2022-05-10 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinases
US10702527B2 (en) 2015-06-12 2020-07-07 Dana-Farber Cancer Institute, Inc. Combination therapy of transcription inhibitors and kinase inhibitors
JP2018525431A (en) * 2015-08-31 2018-09-06 ウーシー ショアンリャン バイオテクノロジー カンパニー,リミティド 2-Arylaminopyridine, pyrimidine or triazine derivatives and their production and use
US11142507B2 (en) 2015-09-09 2021-10-12 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinases
EP3241830A1 (en) 2016-05-04 2017-11-08 Bayer CropScience Aktiengesellschaft Condensed bicyclic heterocyclic derivatives as pesticides
US10513509B2 (en) 2016-05-26 2019-12-24 Recurium Ip Holdings, Llc EGFR inhibitor compounds
US11098030B2 (en) 2016-05-26 2021-08-24 Recurium Ip Holdings, Llc EGFR inhibitor compounds
WO2018033455A1 (en) 2016-08-15 2018-02-22 Bayer Cropscience Aktiengesellschaft Condensed bicyclic heterocycle derivatives as pest control agents
JP7191799B2 (en) 2017-01-26 2022-12-19 ハンミ ファーマシューティカルズ カンパニー リミテッド Pyrimidine compounds and pharmaceutical uses thereof
JP2020023554A (en) * 2017-01-26 2020-02-13 ハンミ ファーマシューティカルズ カンパニー リミテッド Pyrimidine compound and medical use thereof
US10508113B2 (en) 2018-03-12 2019-12-17 Abbvie Inc. Inhibitors of tyrosine kinase 2 mediated signaling
JP2022502484A (en) * 2018-09-21 2022-01-11 シャンハイ エンノババイオ ファーマシューティカルズ カンパニー リミテッドShanghai Ennovabio Pharmaceuticals Co., Ltd. Aromatic heterocyclic compound with kinase inhibitory activity
KR20210095621A (en) * 2018-09-21 2021-08-02 상하이 엔노바바이오 파마슈티컬스 씨오 엘티디. Aromatic heterocyclic compounds with kinase inhibitory activity
JP7349750B2 (en) 2018-09-21 2023-09-25 シャンハイ エンノババイオ ファーマシューティカルズ カンパニー リミテッド Aromatic heterocyclic compounds with kinase inhibitory activity
KR102669660B1 (en) * 2018-09-21 2024-05-27 상하이 엔노바바이오 파마슈티컬스 씨오 엘티디. Aromatic heterocyclic compound with kinase inhibitory activity
US11471455B2 (en) 2018-10-05 2022-10-18 Annapurna Bio, Inc. Compounds and compositions for treating conditions associated with APJ receptor activity
US11944622B2 (en) 2018-10-05 2024-04-02 Annapurna Bio, Inc. Compounds and compositions for treating conditions associated with APJ receptor activity

Also Published As

Publication number Publication date
CN101723936B (en) 2014-01-15
US8309550B2 (en) 2012-11-13
CN101723936A (en) 2010-06-09
US20110263541A1 (en) 2011-10-27

Similar Documents

Publication Publication Date Title
US8309550B2 (en) Kinase inhibitors and their use as pharmaceutical agents
US11591338B2 (en) Pyrrolotriazine compounds as TAM inhibitors
US11406624B2 (en) Pyrazolopyridine compounds and uses thereof
US11866432B2 (en) Dihydropyrido[2,3-d]pyrimidinone compounds as CDK2 inhibitors
RU2675105C9 (en) Substituted aminopyrimidine compounds and methods of use
BR112021007679A2 (en) tyk2 inhibitors and their uses
CA2892608C (en) Compounds useful as inhibitors of atr kinase
EP4011882A1 (en) 4-substituted aminoisoquinoline derivatives
JP5752232B2 (en) Substituted pyrrolotriazine compounds as protein kinase inhibitors
WO2016026445A1 (en) Indazole compounds as fgfr kinase inhibitor, preparation and use thereof
EP3209665B1 (en) Substituted pyrrolotriazine amine compounds as pi3k inhibitors
JP2008540556A5 (en)
KR20090086219A (en) Imidazo[1,2-b]pyridazine and pyrazolo[1,5-a]pyrimidine derivatives and their use as protein kinase inhibitors
TW200829582A (en) Anilinopiperazine derivatives and methods of use thereof
BR112012011287A2 (en) kinase inhibitor compounds, their pharmaceutical formulation and their uses
US8895571B2 (en) Isoindolinone and pyrrolopyridinone derivatives as Akt inhibitors
CN111406054A (en) 1, 2, 4-oxadiazole derivatives as inhibitors of histone deacetylase 6
JP2022517085A (en) Halogenated allylamine compounds and their applications
CN111153891B (en) Substituted benzimidazole PI3K alpha/mTOR double-target inhibitor and pharmaceutical composition and application thereof
US10807983B2 (en) Imidazo-fused heterocycles and uses thereof
US20210221820A1 (en) Compounds for the modulation of myc activity
CA3192158A1 (en) 2-amino-3-carbonyl imidazopyridine and pyrazolopyridine compounds
US20230183251A1 (en) Bicyclic amines as cdk12 inhibitors
EP4310081A1 (en) Ctla-4 small molecule degradation agent and application thereof
BR112018069612B1 (en) PYROLOTRIAZINE COMPOUNDS, USE THEREOF, PHARMACEUTICAL COMPOSITION COMPRISING THEM AND METHODS FOR INHIBITING A TAM KINASE, AND AXL AND MER KINASE IN VITRO

Legal Events

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

Ref document number: 09824436

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 09824436

Country of ref document: EP

Kind code of ref document: A1