WO2007013896A2 - Pyrrol (2,3-b) pyridine derivatives protein kinase inhibitors - Google Patents

Pyrrol (2,3-b) pyridine derivatives protein kinase inhibitors Download PDF

Info

Publication number
WO2007013896A2
WO2007013896A2 PCT/US2006/018726 US2006018726W WO2007013896A2 WO 2007013896 A2 WO2007013896 A2 WO 2007013896A2 US 2006018726 W US2006018726 W US 2006018726W WO 2007013896 A2 WO2007013896 A2 WO 2007013896A2
Authority
WO
WIPO (PCT)
Prior art keywords
optionally substituted
nhr
group
lower alkyl
fluoro
Prior art date
Application number
PCT/US2006/018726
Other languages
English (en)
French (fr)
Other versions
WO2007013896A3 (en
Inventor
Chao Zhang
Jiazhong Zhang
Prabha N. Ibrahim
Clarence R. Hurt
Rebecca Zuchkerman
Dean R. Artis
Ryan Bremer
Wayne Spevak
Guoxian Wu
Hongyao Zhu
Original Assignee
Plexxikon, 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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=37683773&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2007013896(A2) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to CA002608733A priority Critical patent/CA2608733A1/en
Priority to AU2006272951A priority patent/AU2006272951A1/en
Priority to EP06813186A priority patent/EP1885723A2/en
Priority to BRPI0610066-0A priority patent/BRPI0610066A2/pt
Priority to JP2008512402A priority patent/JP2008545652A/ja
Application filed by Plexxikon, Inc. filed Critical Plexxikon, Inc.
Priority to NZ563444A priority patent/NZ563444A/en
Priority to MX2007014377A priority patent/MX2007014377A/es
Publication of WO2007013896A2 publication Critical patent/WO2007013896A2/en
Publication of WO2007013896A3 publication Critical patent/WO2007013896A3/en
Priority to IL187344A priority patent/IL187344A0/en
Priority to NO20075992A priority patent/NO20075992L/no

Links

Classifications

    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/437Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • 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
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/12Drugs for disorders of the metabolism for electrolyte homeostasis
    • A61P3/14Drugs for disorders of the metabolism for electrolyte homeostasis for calcium homeostasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • 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
    • A61P41/00Drugs used in surgical methods, e.g. surgery adjuvants for preventing adhesion or for vitreum substitution
    • 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
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis

Definitions

  • This invention relates to ligands for b-kit and c-fins, and to methods for use thereof.
  • the information provided is intended solely to assist the understanding of the reader. None of the information provided nor references cited is admitted to be prior art to the present invention. Each of the references cited is incorporated herein in its entirety and for any purpose.
  • C-kit and c-fms are both type III transmembrane receptor protein tyrosine kinases (RPTKs) that regulate key signal transduction cascades that control cellular growth and proliferation. Both receptors have similar structural features comprising five extracellular immunoglobulin (IG) domains, a single transmembrane domain, and a split cytoplasmic kinase domain separated by a kinase insert segment.
  • IG immunoglobulin
  • SCF Stem Cell Factor
  • KL kit ligand
  • MEF mast cell growth factor
  • SCF refers to the physiological ligand for c-kit.
  • SCF is synthesized as a transmembrane protein with a molecular weight of 220 or 248 Dalton, depending on alternative splicing of the mRNA to encode exon 6.
  • the larger protein can be proteolytically cleaved to form a soluble, glycosylated protein which noncovalently dimerizes.
  • Both the soluble and membrane-bound forms of SCF can bind to and activate c-kit.
  • SCF is predominantly expressed by fibroblasts, keratinocytes, and endothelial cells, which modulate the activity of melanocytes and mast cells expressing c-kit.
  • marrow stromal cells express SCF and regulate hematopoiesis of c-kit expressing stem cells.
  • intestinal epithelial cells express SCF and affect the interstitial cells of Cajal and intraepithelial lymphocytes.
  • Sertoli cells and granulosa cells express SCF which regulates spermatogenesis by interaction with c-kit on germ cells.
  • C-fms is a member of the family of genes originally isolated from the Susan McDonough strain of feline sarcoma viruses.
  • the cellular proto-oncogene FMS c-fins, cellular feline McDonough sarcoma
  • M-CSF macrophage colony- stimulating factor
  • M-CSF 5 first described by Robinson and co-workers (Blood. 1969, 33:396-9), is a cytokine that controls the production, differentiation, and function of macrophages.
  • M- CSF stimulates differentiation of progenitor cells to mature monocytes, and prolongs the survival of monocytes. Furthermore, M-CSF enhances cytotoxicity, superoxide production, phagocytosis, chemotaxis, and secondary cytokine production of additional factors in monocytes and macrophages. Examples of such additional factors include granulocyte colony stimulating factor (G-CSF), interleukin-6 (IL-6), and interleukin-8 (IL- 8).
  • G-CSF granulocyte colony stimulating factor
  • IL-6 interleukin-6
  • IL-8 interleukin-8
  • M-CSF stimulates hematopoiesis, promotes differentiation and proliferation of osteoclast progenitor cells, and has profound effects on lipid metabolism. Furthermore, M-CSF is important in pregnancy. Physiologically, large amounts of M-CSF are produced in the placenta, and M-CSF is believed to play an essential role in trophoblast differentiation (Motoyoshi, hit J Hematol. 1998, 67:109-22). The elevated serum levels of M-CSF in early pregnancy may participate in the immunologic mechanisms responsible for the maintenance of the pregnancy (Flanagan & Lader, Curr Opin Hematol. 1998, 5:181-5).
  • c-fms and c-kit are two platelet-derived growth factor receptors, alpha (i.e., pdgfra) and beta (pdgfrb) (PDGF).
  • alpha i.e., pdgfra
  • beta pdgfrb
  • PDGF platelet-derived growth factor receptors
  • the gene coding for pdgfra is located on chromosome 4ql l-ql2 in the same region of chromosome 4 as the oncogene coding for c- kit.
  • the genes coding for pdgfra and c-fms appear to have evolved from a common ancestral gene by gene duplication, inasmuch as these two genes are tandemly linked on chromosome 5.
  • GIST gastrointestinal stromal tumors
  • M-CSF the major macrophage growth factor
  • diseases such as for example inflammatory diseases.
  • modulation of the activity of c-fms can ameliorate disease associated with increased levels of M-CSF.
  • the present invention relates to compounds active on c-kit, c-fms, or both c-kit and c-fms.
  • the efficacy of treatment can be enhanced if said compounds are dual inhibitors of both c-kit and c-fms.
  • the invention provides methods of using compounds of Formula I as described below.
  • the invention provides methods of using compounds that can be used therapeutically and/or prophylactically involving modulation of c-kit, c-fms, or both c-kit and c-fms.
  • X 1 is N or CR 2
  • X 2 is N or CR 6
  • Y 1 is N or CR 4
  • Y 2 is N or CR 5 , provided, however, that not more than one of X 2 , Y 1 and Y 2 is N
  • L 1 is selected from the group consisting of optionally substituted lower alkylene, -S-,
  • L 2 is selected from the group consisting of a bond, optionally substituted lower alkylene, -(alk) a -S-(alk) b -, -(alk) a -O-(alk) b -, -(alk) a -OC(O)-(alk) b -, -(alk) a -C(O)O-(alk) b -, -(alk) a -OC(S)-(alk) b -, -(alk) a -C(S)O-(alk) b -, -(alk) a -C(O)-(alk) b -, ⁇ alk) a -C(S)-(alk) b -, -(alk) a -C(O)NR 9 -(alk) b -, -(alk) a -OC(O)NR 9
  • alk is optionally substituted C 1-3 alkylene and a and b are independently 0 or 1;
  • R 1 is selected from the group consisting of optionally substituted lower alkyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl;
  • R 2 , R 4 , R 5 and R 6 are independently selected from the group consisting of hydrogen, halogen, optionally substituted lower alkyl, optionally substituted lower alkenyl, optionally substituted lower alkynyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, -OH, -NH 2 , -NO 2 , -CN, -C(O)OH, -C(S)OH, -C(O)NH 2 , -C(S)NH 2 , -S(O) 2 NH 2 , -NHC(O)NH 2 , -NHC(O)NH 2 , -NHC(S)NH 2 , -NHS(O) 2 NH 2 , -NR 10 R 11 , -NHR 3 , -OR 3 , -SR 3 , -C(O)R 3 , -C(S)R
  • Ar 1 is a 5 or 6 membered optionally substituted heteroarylene having the structure
  • F and J are both C or one of F and J is C and the other of F and J is N;
  • P and Q are independently selected from CR, N, NR, O or S; T is selected from CR or N; wherein when n is 1, F and J are C, and P, T and Q are CR, or any one of P, T and Q is
  • N and the other two of P, T and Q are CR, when n is 0 and F and J are both C, then one of P and Q are CR, N or NR and the other of P and Q is C, N 5 NR, O or S, provided both P and Q are not CR, when n is 0, one of F and J is N and the other of F and J is C, then one of P and
  • R is independently selected from the group consisting of optionally substituted lower alkyl, optionally substituted lower alkenyl, provided, however, that no alkene carbon thereof is bound to any -C(O)-, -C(S)-, -S(O)-, -S(O) 2 -, -O-, -S-, or -N- of any of -OR 3 , -SR 3 , -C(O)R 3 , -C(S)R 3 , -S(O)R 3 , -S(O) 2 R 3 , -C(O)OR 3 , -C(S)OR 3 , -C(O)OR 3 , -C(O)NHR 3 , -C(O)NR 3 R 3 , -C(S)NHR 3
  • R 7 is selected from the group consisting of hydrogen, optionally substituted lower alkyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, -C(O)R 8 , and -S(O) 2 R 8 ;
  • R 8 is selected from the group consisting of optionally substituted lower alkyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl and optionally substituted heteroaryl;
  • R 9 at each occurrence is independently selected from the group consisting of hydrogen, lower alkyl, and lower alkyl substituted with one or more substituents selected from the group consisting of fluoro, -OH, -NH 2 , lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkyltbio, mono- alkylamino, fluoro substituted mono-alkylamino, di-alkylamino, fluoro substituted di-alkylamino, and -NR 12 R 13 , provided, however, that when R 9 is substituted lower alkyl, any substitution on the alkyl carbon bound to the -N- of -NR 9 - is fluoro;
  • R 10 and R 11 at each occurrence are independently selected from the group consisting of optionally substituted lower alkyl, optionally substituted lower alkenyl, provided, however, that no alkene carbon thereof is bound to the nitrogen of -NR 10 R 11 , optionally substituted lower alkynyl, provided, however, that no alkyne carbon thereof is bound to the nitrogen Of-NR 10 R 11 , optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl; or
  • R 10 and R 11 together with the nitrogen to which they are attached form a monocyclic 5-7 membered optionally substituted heterocycloalkyl or a monocyclic 5 or 7 membered optionally substituted nitrogen containing heteroaryl;
  • R 12 and R 13 combine with the nitrogen to which they are attached to form a 5-7 membered heterocycloalkyl or 5-7 membered heterocycloalkyl substituted with one or more substituents selected from the group consisting of fluoro, -OH, -NH 2 , lower alkyl, fluoro substituted lower alkyl, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, and fluoro substituted lower alkylthio; provided, however that when compounds have the structure
  • R la is not phenyl, 4-trifh ⁇ oromethyl- phenyl, 4-methoxy-phenyl, 4-chloro-phenyl, 4-fluoro-phenyl, 4-methyl-phenyl, 3- fluoro-phenyl or thiophen-2-yl and compounds do not have the structure
  • azaindole core the core structure shown above with X 1 , X 2 , Y 1 and Y 2 as CH and with L'-Ari-l ⁇ R 1 replaced with H is referred to as the "azaindole core.”
  • azaindole core reference to ring atoms or ring positions is as shown in the following structure:
  • compounds of Formula I have a structure selected from the following: wherein L 1 , Ar 1 , L 2 , R 1 , R 2 , R 4 , R 5 and R 6 are as defined for Formula I.
  • X 1 and X 2 are N or CH.
  • X 1 , X 2 and Y 1 are N or CH, where in a further embodiment, Y 2 is CR 5 and R 5 is other than hydrogen.
  • X 1 , X 2 and Y 2 are N or CH, where in a further embodiment Y 1 is CR 4 and R 4 is other than hydrogen.
  • X 1 , X 2 and Y 1 are CH, where in a further embodiment, Y 2 is CR 5 and R 5 is other than hydrogen.
  • X 1 , X 2 and Y 2 are CH, where in a further embodiment Y 1 is CR 4 and R 4 is other than hydrogen.
  • R 4 or R 5 is other than hydrogen, preferably where R 2 and R 6 are hydrogen.
  • R 2 , R 5 and R 6 are hydrogen and R 4 is other than hydrogen.
  • R 2 , R 4 and R 6 are hydrogen and R 5 is other than hydrogen.
  • X 1 and X 2 are N or CH, preferably wherein both X 1 and X 2 are CH.
  • L is selected from the group consisting of -S-, -O- lower alkylene, -C(O)-, -C(S)-, -S(O)-, -S(O) 2 -, and -NR 7 -, wherein lower alkylene is optionally substituted with fluoro, and wherein when L 2 is optionally substituted lower alkylene or comprises optionally substituted C 1-3 alkylene, the alkylene is optionally substituted with fluoro or lower alkyl.
  • L 1 is selected from the group consisting of -S-, -O-, -CH 2 -, -CF 2 -, -C(O)-, -C(S)-, -S(O)-, -S(O) 2 -, and -NH-.
  • L 2 is selected from the group consisting of a bond, optionally substituted lower alkylene, -O-(alk) b -, -OC(O)-(aIk) b -, -C(O)O-(alk) b -, -OC(S)-(alk) b -, -C(S)O-(alk) b -, -C(O)-(alk) b -, -C(S)-(alk) b -, -C(O)NR 9 -(alk) b -, -OC(O)NR 9 -(alk) b -, -OC(S)NR 9 -(alk) b -, -C(S)NR 9 -(alk) b -, -S(O) 2 -(alk) b
  • the alkylene is substituted with one or more, preferably 1, 2, or 3 substituents selected from the group consisting of fluoro, -OH, -NH 2 , lower alkoxy, lower alkylthio, mono-alkylamino, di-alkylamino, and -NR 12 R 13 , wherein the alkyl chain(s) of lower alkoxy, lower alkylthio, mono-alkylamino or di-alkylamino are optionally substituted with one or more, preferably 1, 2, or 3 substituents selected from the group consisting of fluoro, -OH, -NH 2 , lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, di-alkylamino,
  • variables P, J, Q, T, F, and n are selected to provide structures of Ar 1 selected from the group consisting of
  • each R is independently hydrogen or an optional substituent as defined herein for optionally substituted heteroaryl.
  • a compound of Formula I has a structure according to the following sub-generic structure, Formula Ia,
  • Formula I a all salts, prodrugs, tautomers, and isomers thereof, wherein L 1 , Ar 1 , R 1 , R 2 , R 4 , R 5 and R 6 are as defined for Formula I;
  • L 3 is selected from the group consisting of a bond, optionally substituted lower alkylene,-O-(alk) b -, -S-(alk) b -, -NR 14 -(alk) b -, -C(O)-(alk) b -, -C(S)-(alk) b -, -S(O)-(alk) b -, -S(O) 2 -(alk) b -, -NR 14 C(O)-(alk) b -, -C(O)NR 14 -(alk) b -, -S(O) 2 NR 14 -(alk) b -, -NR 14 S(O) 2 -(alk) b -, -NR 14 C(O)NR 14 -(alk) b -, -NR 14 C(O)NR 14 -(alk) b -, -NR 14 C
  • R 2 , R 5 and R 6 are hydrogen, further wherein R 4 is other than hydrogen.
  • R 2 , R 4 and R 6 are hydrogen, further wherein R 5 is other than hydrogen.
  • the compounds of Formula Ia may be used to treat a subject suffering from or at risk of a Kit and/or Fms protein kinase mediated disease or condition, such as those disclosed in this application.
  • the compound of Formula I has a structure according to the following sub-generic structure, Formula Ib,
  • V and W are independently selected from the group consisting of N and CH;
  • U and Z are independently selected from the group consisting of N and CR 18 , provided, however, that not more than one of W 5 U and Z is N;
  • A is selected from the group consisting of -CR 19 R 20 -, -C(O)-, -C(S)-, -S-, -S(O)-,
  • n O or 1 ;
  • F and J are both C or one of F and J is C and the other of F and J is N; E and K are selected from C, N, O or S; G is selected from C or N; wherein when n is 1, F and J are C, and E, G and K are C, or any one of E, G and K is N and the other two of E, G and K are C, provided that when E, G or K is N, R 15 , R 17 and R 16 , respectively, are absent, when n is O and F and J are both C, then one of E and K is C or N and the other of E and K is C, N, O or S, provided both E and K are not C, and provided that when both E and K are N, one of R 15 and R 16 is absent, and provided that when one of E and K are N and the other is O or S, R 15 and R 16 are absent, when n is O, one of F and J is N and the other of F and J is C, then one of E and K is N and the other of E and K is
  • R 17 is selected from the group consisting of hydrogen, optionally substituted lower alkyl, -OR 22 , -SR 22 and halogen when G is C, or is absent when G is N;
  • R 18 is selected from the group consisting of hydrogen, halogen, optionally substituted lower alkyl, optionally substituted aryl, optionally substituted heteroaryl, -OH, -NH 2 , -NO 2 , -CN, -NHC(O)NH 2 , -NHC(S)NH 2 , -NHS(O) 2 NH 2 , -NR 24 R 25 , -NHR 23 , -OR 23 , -SR 23 , -NHC(O)R 23 , -NR 23 C(O)R 23 , -NHC(S)R 23 , -NR 23 C(S)R 23 , -NHS(O) 2 R 23 , -NR 23 S(O) 2 R 23 , -NHC(O)NHR 23 , -NR 23 C(O)NH 2 , -NR 23 C(O)NHR 23 , -NHC(O)NR 23 R 23 , -NR 23 C(O)NR 23
  • M is selected from the group consisting of a bond, -(CR 19 R 20 ) u -, -(CR 19 R 2 VC(O)-(CR 19 R 2 V, -(CR 19 R 20 VC(S)-(CR 19 R 2 V 5 -(CR 19 R 20 VC(O)O-(CR 19 R 2 V, -(CR 19 R 20 VC(S)O-(CR 19 R 2 V, -(CR 19 R 20 ) t -C(O)NR 26 -(CR 19 R 20 ) s -, -(CR 19 R 20 ) t -C(S)NR 26 -(CR 19 R 20 ) s -, -(CR 19 R 20 VS(O)-(CR 19 R 2 V, -(CR 19 R 20 VS(O) 2 -(CR 19 R 2 V 5 -(CR 19 R 20 VS(O) 2 NR 26 -(CR 19 R 20 ) s -, -(CR 19 R 20
  • R 21 and R 22 at each occurrence are independently hydrogen or optionally substituted lower alkyl
  • R 23 at each occurrence is independently selected from the group consisting of optionally substituted lower alkyl, optionally substituted lower alkenyl, provided, however, that no alkene carbon thereof is bound to any -C(O)-, -C(S)-, -S(O) 2 -, -O-, -S-, or -N- of any of -NHR 23 , -OR 23 , -SR 23 , -NHC(O)R 23 , -NR 23 C(O)R 23 , - NHC(S)R 23 , -NR 23 C(S)R 23 , -NHS(O) 2 R 23 , -NR 23 S(O) 2 R 23 , -NHC(O)NHR 23 , -NR 23 C(O)NH 2 , -NR 23 C(O)NHR 23 , -NHC(O)NR 23 R 23 , -NR 23 C(O)NR 23 R 23 , -NHC(S)
  • R 24 and R 25 at each occurrence are independently selected from the group consisting of optionally substituted lower alkyl, optionally substituted lower alkenyl, provided, however, that no alkene carbon thereof is bound to the nitrogen of -NR 24 R 25 , optionally substituted lower alkynyl, provided, however, that no alkyne carbon thereof is bound to the nitrogen Of-NR 24 R 25 , optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl; or
  • R 24 and R 25 together with the nitrogen to which they are attached form a monocyclic 5-7 membered optionally substituted heterocycloalkyl or a monocyclic 5 or 7 membered optionally substituted nitrogen containing heteroaryl;
  • R 26 at each occurrence is independently selected from the group consisting of hydrogen, lower alkyl, and lower alkyl substituted with one or more substituents selected from the group consisting of fluoro, -OH, -NH 2 , lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono- alkylamino, fluoro substituted mono-alkylamino, di-alkylamino, fluoro substituted di-alkylamino, and -NR 27 R 28 , provided, however, that when R 26 is substituted lower alkyl, any substitution on the lower alkyl carbon bound to the -N- of -NR 26 - is fluoro;
  • E, J, K, G, F, n, R 15 , R 16 and R 17 are selected to provide structures selected from the group consisting of
  • M is selected from the group consisting of -O-(CR 19 R 20 ) s -, -S-(CR 19 R 2 V, -OC(O)-(CR 19 R 20 ) s -, -OC(S)-(CR 19 R 2 V, -OC(O)NR 26 -(CR 19 R 20 ) s - 5 -OC(S)NR 26 -(CR 19 R 20 ) s -, -C(O)NR 26 -(CR 19 R 20 ) s -, -C(S)NR 26 -(CR 19 R 20 ) s - 5 -S(O) 2 NR 26 -(CR 19 R 20 ) s -, -NR 26 -(CR 19 R 20 ) s -, -NR 26 C(O)-(CR 19 R 2 V, -NR 26 C(S)-(CR 19 R 20 ) s -, -NR 26 C(O)-(CR 19 R 2 V, -
  • R 26 at each occurrence is independently selected from the group consisting of hydrogen, lower alkyl, or lower alkyl substituted with 1, 2, or 3 substituents selected from the group consisting of fluoro, -OH, -NH 2 , alkoxy, lower alkylthio, mono-alkylamino, di-alkylamino and cycloalkylamino, provided that any substitution on the carbon that is bound to the nitrogen of -NR 26 is fluoro.
  • R 1 is selected from the group consisting of optionally substituted aryl and optionally substituted heteroaryl.
  • Z is N or CH, n is 1, E-R 15 is N or CH, K-R 16 is N or CH, and G-R 17 is N or CH, provided no more than one of E-R 15 , K-R 16 and G-R 17 is N.
  • Z is N or CH, n is 1, and E-R 15 , K-R 16 and G-R 17 are CH.
  • V, W and Z are CH, U is CR 18 , n is 1, E-R 15 is N or CH, K-R 16 is N or CH, and G-R 17 is N or CH, provided no more than one of E-R 15 , K-R 16 and G-R 17 is N.
  • V, W and Z are CH, U is CR 18 , n is 1, and E-R 15 , K-R 16 and G-R 17 are CH.
  • Z is N or CH
  • n is 1, E-R 15 , K-R 16 and G-R 17 are CH
  • A is -CH 2 -
  • M is -NHCH 2 -, further wherein R 1 is optionally substituted phenyl.
  • V, Z, U and W are CH
  • n is 1, E-R 15 is N or CH
  • K-R 16 is N or CH
  • G-R 17 is N or CH, provided no more than one of E-R 15 , K-R 16 and G-R 17 is N.
  • Z is N or CH
  • n is 1
  • E-R 15 is N or CH
  • K-R 16 is N or CH
  • G-R 17 is N or CH, provided no more than one of E-R 15 , K-R 16 and G-R 17 is N
  • R 1 is phenyl optionally substituted with one or more substituents selected from the group consisting of halogen, -OH, -NH 2 , -NO 2 , -CN, optionally substituted lower alkyl and -OR 29 , where R 29 is selected from the group consisting of optionally substituted lower alkyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl and optionally substituted heteroaryl.
  • V, Z, U and W are CH, n is 1, E-R 15 , K-R 16 and G-R 17 are CH, A is -CH 2 -, M is -NHCH 2 , and R 1 is optionally substituted phenyl, further wherein R 1 is phenyl optionally substituted with one or more substituents selected from the group consisting of halogen, -OH, -NH 2 , -NO 2 , -CN, optionally substituted lower alkyl and -OR 29 , where R 29 is selected from the group consisting of optionally substituted lower alkyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl and optionally substituted heteroaryl.
  • V, W and Z are CH
  • U is CR 18
  • n is 1
  • E-R 15 , K-R 16 and G-R 17 are CH
  • A is -CH 2 -
  • M is -NHCH 2
  • R 1 is optionally substituted phenyl, further wherein R 1 is phenyl optionally substituted with one or more substituents selected from the group consisting of halogen, -OH, -NH 2 , -NO 2 , -CN, optionally substituted lower alkyl and -OR 29 , where R 29 is selected from the group consisting of optionally substituted lower alkyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl and optionally substituted heteroaryl.
  • n when n is 1, and E, K and G are C, at least one of R 15 , R 16 and R 17 is other than hydrogen.
  • n is 1, one of E, K, and G are N and the other two of E, K, and G are C and at least one of R 15 , R 16 and R 17 is other than hydrogen.
  • n is 1, E, K and G are C, and at least one of R 15 , R 16 and R 17 is other than hydrogen.
  • n is 1, V and W are CH, U and Z are independently CR 18 , one of E, K, and G are N and the other two of E, K, and G are C and at least one of R 15 , R 16 and R 17 is other than hydrogen.
  • n is 1, V and W are CH, U and Z are independently CR 18 , E, K and G are C, and at least one of R 15 , R 16 and R 17 is other than hydrogen.
  • n is 1, one of E, K, and G are N and the other two of E, K, and G are C, at least one of R 15 , R 16 and R 17 is other than hydrogen, A is -CH 2 -, M is -NHCH 2 -, further wherein R 1 is optionally substituted phenyl.
  • n is 1, E, K, and G are C, at least one of R 15 , R 16 and R 17 is other than hydrogen, A is -CH 2 -, M is -NHCH 2 -, further wherein R 1 is optionally substituted phenyl.
  • n is 1, V, Z, U and W are CH, one of E, K, and G are N and the other two of E, K, and G are C and at least one of R 15 , R 16 and R 17 is other than hydrogen.
  • V, Z, U and W are CH, E, K and G are C, and at least one of R 15 , R 16 and R 17 is other than hydrogen.
  • Z is CR 18 , wherein R 18 is other than hydrogen, n is 1, E-R 15 is N or CH, K-R 16 is N or CH and G-R 17 is N or CH.
  • Z is CR 18 , wherein R 18 is other than hydrogen, n is 1, and E-R 15 , K-R 16 and G-R 17 are CH.
  • Z is CR 18 , wherein R 18 is other than hydrogen, U is CR 18 , V and W are CH, n is 1, and E-R 15 , K-R 16 and G-R 17 are CH, further wherein U is CH.
  • Z is CR 18 , wherein R 18 is other than hydrogen, n is 1, E-R 15 , K-R 16 and G-R 17 are CH, A is -CH 2 -, M is -NHCH 2 -, further wherein R 1 is optionally substituted phenyl.
  • Z is CR 18 , wherein R 18 is other than hydrogen, U is CR 18 , V and W are CH, n is 1, E-R 15 , K-R 16 and G-R 17 are CH, A is -CH 2 -, M is -NHCH 2 -, further wherein R 1 is optionally substituted phenyl.
  • Z is CR 18 , wherein R 18 is other than hydrogen, V, U and W are CH, n is 1, E-R 15 , K-R 16 and G-R 17 are CH, A is -CH 2 -, M is -NHCH 2 -, further wherein R 1 is optionally substituted phenyl.
  • U is CR 18 , wherein R 18 is other than hydrogen, n is 1, E-R 15 is N or CH, K-R 16 is N or CH and G-R 17 is N or CH.
  • U is CR 18 , wherein R 18 is other than hydrogen, n is 1, and E-R 15 , K-R 16 and G-R 17 are CH.
  • U is CR 18 , wherein R 18 is other than hydrogen, Z is CR 18 , V and W are CH, n is 1, and E-R 15 , K-R 16 and G-R 17 are CH, further wherein Z is CH.
  • U is CR 18 , wherein R 18 is other than hydrogen, n is 1, E-R 15 , K-R 16 and G-R 17 are CH, A is -CH 2 -, M is -NHCH 2 -, further wherein R 1 is optionally substituted phenyl.
  • U is CR 18 , Wherein R 18 is other than hydrogen, Z is CR 18 , V and W are CH, n is 1, E-R 15 , K-R 16 and G-R 17 are CH, A is -CH 2 -, M is -NHCH 2 -, further wherein R 1 is optionally substituted phenyl.
  • U is CR 18 , wherein R 18 is other than hydrogen, V, Z and W are CH, n is 1, E-R 15 , K-R 16 and G-R 17 are CH, A is -CH 2 -, M is -NHCH 2 -, further wherein R 1 is optionally substituted phenyl.
  • R 15 , R 16 and R 17 are independently selected from the group consisting of halogen, -OH, lower alkyl, fluoro substituted lower alkyl, lower alkoxy, and fluoro substituted lower alkoxy.
  • R 1 is phenyl optionally substituted with one or more substituents selected from the group consisting of halogen, -OH, -NH 2 , -NO 2 , -CN, optionally substituted lower alkyl and -OR 29 , where R 29 is selected from the group consisting of optionally substituted lower alkyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl and optionally substituted heteroaryl.
  • R 18 is selected from the group consisting of halogen, -OH, optionally substituted lower alkyl and -OR 29 , where R 29 is selected from the group consisting of optionally substituted lower alkyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl and optionally substituted heteroaryl.
  • R 1 is phenyl optionally substituted with one or more substituents selected from the group consisting of halogen, -OH, -NH 2 , -NO 2 , -CN, optionally substituted lower alkyl and -OR 29 , where R 29 is selected from the group consisting of optionally substituted lower alkyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl and optionally substituted heteroaryl.
  • M is a bond and R 1 is other than thiophenyl.
  • Z is N or CR 18 wherein R 18 is not hydrogen.
  • E is NR 15 or CR 15
  • K is NR 16 or CR 16
  • G is CR 17 , or combinations thereof, wherein at least one of R 15 , R 16 and R 17 is not hydrogen.
  • the compounds of Formula Ib may be used to treat a subject suffering from or at risk of a Kit and/or Fms protein kinase mediated disease or condition, such as those disclosed in this application.
  • a compound of Formula I has a structure according to the following sub-generic structure, Formula Ig,
  • Z 1 is selected from the group consisting of N and CR 34 ;
  • U 1 is selected from the group consisting of N and CR ;
  • a 1 is selected from the group consisting of -CH 2 - and -C(O)-;
  • M 3 is selected from the group consisting of a bond, -NR 39 -, -S-, -O-, -NR 39 CH 2 -, -NR 39 CH(R 40 )-, -SCH 2 -, -OCH 2 -, -C(O)NR 39 -, -S(O) 2 NR 39 -, -CH 2 NR 39 -, -CH(R 40 )NR 39 -, -NR 39 C(O)-, and -NR 39 S(O) 2 -;
  • n is O or 1 ;
  • v is 0, 1, 2 or 3;
  • F 1 and J 1 are both C or one OfF 1 and J 1 is C and the other OfF 1 and J 1 is N; E 1 and K 1 are selected from C, N, O or S; G 1 is selected from C or N; wherein when n is 1, F 1 and J 1 are C, and E 1 , G 1 and K 1 are C, or any one OfE 1 , G 1 and K 1 is N and the other two OfE 1 , G 1 and K 1 are C, provided that when E 1 , G 1 or K 1 is N, R 36 , R 37 and R 38 , respectively, are absent.
  • R 34 and R 35 are independently selected from the group consisting of hydrogen, -OR 41 , -SR 41 , -NHR 41 , -NR 41 R 41 , -NR 39 C(O)R 41 , -NR 39 S(O) 2 R 41 , halogen, lower alkyl, cycloalkyl, heterocycloalkyl, aryl and heteroaryl, wherein lower alkyl is optionally substituted with one or more substituents selected from the group consisting of fluoro, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, di-alkylamino, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl, wherein cycloalkyl, heterocycloalkyl, aryl, and heteroaryl as R 34 or R 35 , or as substituents of lower alkyl are optionally substituted with one or more substituent
  • R 45 at each occurrence is independently selected from the group consisting of -OR 41 , -SR 41 , -NHR 41 , -NR 41 R 41 , -NR 39 C(O)R 41 , -NR 39 S(O) 2 R 41 , halogen, lower alkyl, cycloalkyl, heterocycloalkyl, aryl and heteroaryl, wherein lower alkyl is optionally substituted with one or more substituents selected from the group consisting of fluoro, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, di-alkylamino, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl, wherein cycloalkyl, heterocycloalkyl, aryl, and heteroaryl as R 45 , or as substituents of lower alkyl are optionally substituted with one or more substituents selected from the
  • R 38 is selected from the group consisting of hydrogen, halogen, lower alkyl, fluoro substituted lower alkyl, lower alkoxy, and fluoro substituted lower alkoxy when G 1 is C, or is absent when G 1 is N;
  • R 39 at each occurrence is independently selected from the group consisting of hydrogen and lower alkyl
  • R 40 is selected from the group consisting of lower alkyl, and fluoro substituted lower alkyl
  • R 41 is selected from the group consisting of lower alkyl, cycloalkyl, heterocycloalkyl, aryl and heteroaryl, wherein lower alkyl is optionally substituted with one or more substituents selected from the group consisting of fluoro, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono- alkylamino, di-alkylamino, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl, wherein cycloalkyl, heterocycloalkyl, aryl, and heteroaryl as R 41 or as substituents of lower alkyl are are optionally substituted with one or more substituents selected from the group consisting of -OH, -NH 2 , -CN, -NO 2 , -S(O) 2 NH 2 , -C(O)NH 2 , -OR 42 , -SR 42 , -NHR
  • R 42 at each occurrence is independently selected from the group consisting of lower alkyl, heterocycloalkyl and heteroaryl, wherein lower alkyl is optionally substituted with one or more substituents selected from the group consisting of fluoro, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, di-alkylamino, and cycloalkylamino.
  • n is 1 , G 1 and K 1 are C, and E is N or C, preferably wherein E is C.
  • M 3 is selected from the group consisting of -NR 39 -, -O 5 -NR 39 CH 2 -, -NR 39 CH(R 40 )-, -SCH 2 -, -OCH 2 -, -CH 2 NR 39 -, -NR 39 C(O)-, and -NR 39 S(O) 2 -, preferably wherein M 3 is -NR 39 CH 2 -, -NR 39 CH(R 40 )-, , -SCH 2 -, -OCH 2 -, or -CH 2 NR 39 -.
  • n is 1, G 1 and K 1 are C, and E is N or C, preferably wherein E is C, and M 3 is selected from the group consisting of -NR 39 -, -O-, -NR 39 CH 2 -, -NR 39 CH(R 40 )-, -SCH 2 -, -OCH 2 -, -CH 2 NR 39 -, -NR 39 C(O)-, and -NR 39 S(O) 2 -, preferably wherein M 3 is -NR 39 CH 2 -, -NR 39 CH(R 40 )-, -SCH 2 -, -OCH 2 -, or -CH 2 NR 39 -.
  • each R 45 is selected from the group consisting of -OH, -NH 2 , -CN, -NO 2 , halogen, lower alkyl, fluoro substituted lower alkyl, lower alkoxy, fluoro substituted lower alkoxy, lower thioalkyl, fluoro substituted lower thioalkyl, mono-alkylamino, di-alkylamino and cycloalkylamino, preferably wherein v is 0, 1, or 2, also O or 1.
  • n is 1, G 1 and K 1 are C, and E is N or C, preferably wherein E is C, M 3 is selected from the group consisting of -NR 39 -, -O-, -NR 39 CH 2 -, -NR 39 CH(R 40 )-, -SCH 2 -, -OCH 2 -, -CH 2 NR 39 -, -NR 39 C(O)-, and -NR 39 S(O) 2 -, preferably wherein M 3 is -NR 39 CH 2 -, -NR 39 CH(R 40 )-, -SCH 2 -, -OCH 2 -, or -CH 2 NR 39 -, and each R 45 is selected from the group consisting of -OH, -NH 2 , -CN, -NO 2 , halogen, lower alkyl, fluoro substituted lower alkyl, lower alkoxy, fluoro substituted lower alkoxy, lower thi
  • Z 1 is CR 34 , U 1 is CR 35 , and R 34 and R 35 are both hydrogen.
  • Z 1 is CR 34
  • U 1 is CR 35
  • R 34 and R 35 are independently selected from the group consisting of hydrogen, -OR 41 , halogen, lower alkyl, cycloalkyl, heterocycloalkyl, aryl and heteroaryl, wherein cycloalkyl, heterocycloalkyl, aryl and heteroaryl are optionally substituted with one or more substituents selected from the group consisting of -OH, -NH 2 , -CN, -NO 2 , -S(O) 2 NH 2 , -C(O)NH 2 , -OR 42 , -SR 42 , -NHR 42 , -NR 42 R 42 , -NR 39 C(O)R 42 , -NR 39 S(O) 2 R 42 , -S(O
  • one of R 34 and R 35 is hydrogen, and the other of R 34 and R 35 is selected from the group consisting of hydrogen, halogen, lower alkyl, lower alkoxy, aryl and heteroaryl, wherein aryl and heteroaryl are optionally substituted with one or more substituents selected from the group consisting of -OH, -NH 2 , -CN, -NO 2 , -S(O) 2 NH 2 , -C(O)NH 2 , -OR 42 , -SR 42 , -NHR 42 , -NR 42 R 42 , -NR 39 C(O)R 42 , -NR 39 S(O) 2 R 42 , -S(O) 2 R 42 , halogen, lower alkyl, fluoro substituted lower alkyl, and cycloalkylamino, and wherein lower alkyl and lower alkoxy are optionally substituted with one or more substituents selected from the group consisting of fluoro, lower
  • each R 45 is independently selected from the group consisting of -OH, -NH 2 , -CN, -NO 2 , halogen, lower alkyl, fluoro substituted lower alkyl, lower alkoxy, fluoro substituted lower alkoxy, lower thioalkyl, fluoro substituted lower thioalkyl, mono-alkylamino, di-alkylamino and cycloalkylamino, preferably wherein v is 0, 1, or 2, also O or 1, Z 1 is CR 34 , U 1 is CR 35 , and R 34 and R 35 are independently selected from the group consisting of hydrogen, -OR 41 , halogen, lower alkyl, cycloalkyl, heterocycloalkyl, aryl and heteroaryl, wherein cycloalkyl, heterocycloalkyl, aryl and heteroaryl are optionally substituted with one or more substituents selected from the group consisting of -
  • each R 45 is selected from the group consisting of -OH, -NH 2 , -CN, -NO 2 , halogen, lower alkyl, fluoro substituted lower alkyl, lower alkoxy, fluoro substituted lower alkoxy, lower thioalkyl, fluoro substituted lower thioalkyl, mono-alkylamino, di-alkylamino and cycloalkylamino, preferably wherein v is 0, 1, or 2, also 0 or 1, Z 1 is CR 34 , U 1 is CR 35 , one of R 34 and R 35 is hydrogen, and the other of R 34 and R 35 is selected from the group consisting of hydrogen, halogen, lower alkyl, lower alkoxy, aryl and heteroaryl, wherein aryl and heteroaryl are optionally substituted with one or more substituents selected from the group consisting of -OH, -NH 2 , -CN, -NO 2 , halogen, lower alkyl, fluoro
  • n is 1, G 1 and K 1 are C, and E is N or C, preferably wherein E is C, M 3 is selected from the group consisting of -NR 39 -, -0-, -NR 39 CH 2 -, -NR 39 CH(R 40 )-, -SCH 2 -, -OCH 2 -, -CH 2 NR 39 -, -NR 39 C(O)-, and -NR 39 S(O) 2 -, preferably wherein M 3 is -NR 39 CH 2 -, -NR 39 CH(R 40 )-, -SCH 2 -, -OCH 2 -, or -CH 2 NR 39 -, each R 45 is selected from the group consisting of -OH, -NH 2 , -CN, -NO 2 , halogen, lower alkyl, fluoro substituted lower alkyl, lower alkoxy, fluoro substituted lower alkoxy, lower thioal
  • n is 1, G 1 and K 1 are C, and E is N or C, preferably wherein E is C, M 3 is selected from the group consisting of -NR 39 -, -O-, -NR 39 CH 2 -, -NR 39 CH(R 40 )-, -SCH 2 -, -OCH 2 -, -CH 2 NR 39 -, -NR 39 C(O)-, and -NR 39 S(O) 2 -, preferably wherein M 3 is -NR 39 CH 2 -, -NR 39 CH(R 40 )-, -SCH 2 -, -OCH 2 -, or -CH 2 NR 39 -, each R 45 is selected from the group consisting of -OH 5 -NH 2 , -CN, -NO 2 , halogen, lower alkyl, fluoro substituted lower alkyl, lower alkoxy, fluoro substituted lower alkoxy, lower thio
  • one of R 34 and R 35 is hydrogen, and the other of R 34 and R 35 is selected from the group consisting of halogen, lower alkyl, lower alkoxy, aryl and heteroaryl, wherein aryl and heteroaryl are optionally substituted with one or more substituents selected from the group consisting of -OH, -NH 2 , -CN, -NO 2 , -S(O) 2 NH 2 , -C(O)NH 2 , -OR 42 , -SR 42 , -NHR 42 , -NR 42 R 42 , -NR 39 C(O)R 42 , -NR 39 S(O) 2 R 42 , -S(O) 2 R 42 , halogen, lower alkyl, fluoro substituted lower alkyl, and cycloalkylamino, and wherein lower alkyl and lower alkoxy are optionally substituted with one or more substituents selected from the group consisting of fluoro, lower alk
  • the compounds of Formula Ig may be used to treat a subject suffering from or at risk of a Kit and/or Fms protein kinase mediated disease or condition, such as those disclosed in this application.
  • compounds are excluded where N (except where N is a heteroaryl ring atom), O, or S is bound to a carbon that is also bound to N (except where N is a heteroaryl ring atom), O, or S; or where N (except where N is a heteroaryl ring atom), O, C(S), C(O), or S(O) n (n is 0-2) is bound to an alkene carbon of an alkenyl group or bound to an alkyne carbon of an alkynyl group; accordingly, in certain embodiments compounds which include linkages such as the following are excluded from the present invention: -NR-CH 2 -NR-, -0-CH 2 -NR-, -S-CH 2 -NR-, -NR-CH 2 -O-, -0-CH 2 -O-, -S-CH 2 -O- -NR-CH 2 -S-, -0-CH 2 -S-, -S-CH 2
  • the invention provides methods for treating a c-kit-mediated disease or condition in an animal subject (e.g. a mammal such as a human, other primates, sports animals, animals of commercial interest such as cattle, farm animals such as horses, or pets such as dogs and cats), e.g., a disease or condition characterized by abnormal c-kit activity (e.g. kinase activity).
  • an animal subject e.g. a mammal such as a human, other primates, sports animals, animals of commercial interest such as cattle, farm animals such as horses, or pets such as dogs and cats
  • a disease or condition characterized by abnormal c-kit activity e.g. kinase activity
  • the c-kit mediated disease is selected from the group consisting of malignancies, including mast cell tumors, small cell lung cancer, testicular cancer, gastrointestinal stromal tumors (GISTs), glioblastoma, astrocytoma, neuroblastoma, carcinomas of the female genital tract, sarcomas of neuroectodermal origin, colorectal carcinoma, carcinoma in situ, Schwann cell neoplasia associated with neurofibromatosis, acute myelocytic leukemia, acute lymphocytic leukemia, chronic myelogenous leukemia, mastocytosis, melanoma, and canine mast cell tumors, and inflammatory diseases, including asthma, rheumatoid
  • compounds of Formula I, Formula Ia, Formula Ib, or Formula Ig, and all sub-embodiments thereof can be used in the preparation of a medicament for the treatment of a c-kit-mediated disease or condition selected from the group consisting of malignancies, including mast cell tumors, small cell lung cancer, testicular cancer, gastrointestinal stromal tumors (GISTs), glioblastoma, astrocytoma, neuroblastoma, carcinomas of the female genital tract, sarcomas of neuroectodermal origin, colorectal carcinoma, carcinoma in situ, Schwann cell neoplasia associated with neurofibromatosis, acute myelocytic leukemia, acute lymphocytic leukemia, chronic myelogenous leukemia, mastocytosis, melanoma, and canine mast cell tumors, and inflammatory diseases, including asthma, rheumatoid arthritis, allergic rhinitis, multiple
  • the invention provides methods for treating a c-fms-mediated disease or condition in an animal subject (e.g. a mammal such as a human, other primates, sports animals, animals of commercial interest such as cattle, farm animals such as horses, or pets such as dogs and cats), e.g., a disease or condition characterized by abnormal c-fms activity (e.g. kinase activity).
  • animal subject e.g. a mammal such as a human, other primates, sports animals, animals of commercial interest such as cattle, farm animals such as horses, or pets such as dogs and cats
  • Invention methods involve administering to the subject suffering from or at risk of a c-fms-mediated disease or condition an effective amount of compound of Formula I, Formula Ia, Formula Ib, or Formula Ig, and all sub-embodiments thereof.
  • the c-fms mediated disease is selected from the group consisting of immune disorders, including rheumatoid arthritis, systemic lupus erythematosis (SLE), Wegener's granulomatosis, and transplant rejection, inflammatory diseases including Chronic Obstructive Pulmonary Disease (COPD), emphysema, and atherosclerosis, metabolic disorders, including insulin resistance, hyperglycemia, and lipolysis, disorders of bone structure or mineralization, including osteoporosis, increased risk of fracture, hypercalcemia, and bone metastases, kidney diseases, including nephritis (e.g.
  • glomerulonephritis including interstitial nephritis, Lupus nephritis), tubular necrosis, diabetes- associated renal complications, and hypertrophy and cancers, including multiple myeloma, acute myeloid leukemia, chronic myeloid leukemia (CML), breast cancer, and ovarian cancer.
  • CML chronic myeloid leukemia
  • compounds of Formula I, Formula Ia, Formula Ib, or Formula Ig, and all sub-embodiments thereof can be used in the preparation of a medicament for the treatment of a c-fms-mediated disease or condition selected from the group consisting of immune disorders, including rheumatoid arthritis, systemic lupus erythematosis (SLE), Wegener's granulomatosis, and transplant rejection, inflammatory diseases including Chronic Obstructive Pulmonary Disease (COPD), emphysema, and atherosclerosis, metabolic disorders, including insulin resistance, hyperglycemia, and lipolysis, disorders of bone structure or mineralization, including osteoporosis, increased risk of fracture, hypercalcemia, and bone metastases, kidney diseases, including nephritis (e.g.
  • a c-fms-mediated disease or condition selected from the group consisting of immune disorders, including rheumatoid arthritis, systemic lupus erythematosis (S
  • glomerulonephritis including interstitial nephritis, Lupus nephritis), tubular necrosis, diabetes- associated renal complications, and hypertrophy and cancers, including multiple myeloma, acute myeloid leukemia, chronic myeloid leukemia (CML), breast cancer, and ovarian cancer.
  • CML chronic myeloid leukemia
  • the invention provides methods for treating a c-fms-mediated and/or c-kit-mediated disease or condition in an animal subject (e.g. a mammal such as a human, other primates, sports animals, animals of commercial interest such as cattle, farm animals such as horses, or pets such as dogs and cats), e.g., a disease or condition characterized by abnormal c-fms activity and/or c-kit activity (e.g. kinase activity).
  • an animal subject e.g. a mammal such as a human, other primates, sports animals, animals of commercial interest such as cattle, farm animals such as horses, or pets such as dogs and cats
  • a disease or condition characterized by abnormal c-fms activity and/or c-kit activity e.g. kinase activity
  • Invention methods involve administering to the subject suffering from or at risk of a c- fms-mediated and/or c-kit mediated disease or condition an effective amount of compound of Formula I, Formula Ia, Formula Ib, or Formula Ig, and all sub-embodiments thereof.
  • the c-fms and/or c-kit mediated disease is selected from the group consisting of mast cell tumors, small cell lung cancer, testicular cancer, gastrointestinal stromal tumors, glioblastoma, astrocytoma, neuroblastoma, carcinomas of the female genital tract, sarcomas of neuroectodermal origin, colorectal carcinoma, carcinoma in situ, Schwann cell neoplasia associated with neurofibromatosis, acute myeloid leukemia, acute lymphocytic leukemia, chronic myelogenous leukemia, multiple myeloma, mastocytosis, melanoma, breast cancer, ovarian cancer, canine mast cell tumors, hypertrophy, asthma, rheumatoid arthritis, allergic rhinitis, multiple sclerosis, inflammatory bowel syndrome, transplant rejection, systemic lupus erythematosis, Wegener's granulomatosis, Chronic Ob
  • compounds of Formula I 5 Formula Ia, Formula Ib, or Formula Ig, and all sub-embodiments thereof can be used in the preparation of a medicament for the treatment of a c-fms-mediated and/or c-kit mediated disease or condition selected from the group consisting of mast cell tumors, small cell lung cancer, testicular cancer, gastrointestinal stromal tumors, glioblastoma, astrocytoma, neuroblastoma, carcinomas of the female genital tract, sarcomas of neuroectodermal origin, colorectal carcinoma, carcinoma in situ, Schwann cell neoplasia associated with neurofibromatosis, acute myeloid leukemia, acute lymphocytic leukemia, chronic myelogenous leukemia, multiple myeloma, mastocytosis, melanoma, breast cancer, ovarian cancer, canine mast cell tumors, hypertrophy, asthma, rheuma
  • the invention provides methods of using compounds of Formula I 5 Formula Ia, Formula Ib, or Formula Ig, and all sub-embodiments thereof, as described herein (e.g. compounds that have advantageous levels of activity and/or selectivity on c-kit, c-fms or both c-kit and c-fms).
  • the compounds are substituted at the 3-position of the core bicyclic ring structure (azaindole core) with a substituent group that in order includes a first linker bound to a first aryl or heteroaryl group, which is bound to a linker of 1 to 3 atoms bound to a second aryl or heteroaryl group.
  • the first linker is methylene, ethylene, -C(O)-, -C(S)-, -O-, -S-, or -S(O) 2 -;
  • the first aryl or heteroaryl group is pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, pyrrolyl, imidazolyl, triazolyl, thiazolyl, or oxazolyl;
  • the second linker is methyl amino (NHCH 2 ), ethyl amino (NHCH 2 CH 2 ), amide (NHC(O)), or sulfonamide (NHSO 2 );
  • the second aryl or heteroaryl group is phenyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, pyrrolyl, imidazolyl, triazolyl, thiazolyl
  • alkoxy group e.g. amethoxy group, an ethoxy group, a propoxy group, or a butoxy group
  • a halo substituted lower alkyl e.g. -CH 2 F, -CHF 2 , or -CF 3
  • halo e.g. F or Cl
  • the second aryl or heteroaryl group is a 6-membered ring; the 6-membered ring is substituted at the para position; the 6-membered ring is substituted at the meta position; the 6-membered ring is substituted at the ortho position; or the 6-membered ring is substituted at the meta and para positions, hi particular embodiments, the second aryl or heteroaryl group is a 5-membered ring; the 5-membered ring is substituted at a position adjacent to the atom bound to the second linker; or the 5-membered ring is substituted at a position not adjacent to the atom bound to the second linker.
  • the 3-position substitutent group is the only non-hydrogen substitutent on the azaindole core.
  • the compound has an ICs 0 of less than 100 nM, less than 50 nM, less than 20 nM, less than 10 nM, or less than 5 nM as determined in a generally accepted kinase activity assay, hi certain embodiments, the selectivity of the compound is such that the compound is at least 2-fold, 5 -fold, 10-fold, or 100-fold more active on c-kit than on Ret, PDGF, or both Ret and PDGF. In certain embodiments, the selectivity of the compound is such that the compound is at least 2-fold, 5 -fold, 10-fold, or 100-fold more active on c-kit than on c-fms.
  • the selectivity of the compound is such that the compound is at least 2-fold, 5-fold, 10-fold, or 100-fold more active on c-fms than on c-kit.
  • the compound has in combination each pairing of activity (e.g. IC 50 ) and/or selectivity as specified in this paragraph.
  • the compound has an IC 50 of less than 100 nM, less than 50 nM, less than 20 nM, less than 10 nM, or less than 5 nM as determined in a generally accepted kinase activity assay for c-kit, c-fms, or both c-kit and c-fms kinase activity, hi certain embodiments, the selectivity of the compound is such that the compound is at least 2-fold, 5-fold, 10-fold, or 100-fold more active on c-kit, c-fms, or both c-kit and c-fms than on Ret, PDGF, or both Ret and PDGF.
  • compositions that include a therapeutically effective amount of a compound of Formula I (including Formula Ia, Ib, Ig and all sub-embodiments thereof) and at least one pharmaceutically acceptable carrier, excipient, and/or diluent.
  • the composition can include a plurality of different pharmacologically active compounds, which can include a plurality of compounds of Formula I (including Formula Ia, Ib, Ig and all sub-embodiments thereof).
  • kits that include a composition as described herein.
  • the composition is packaged, e.g., in a vial, bottle, flask, which may be further packaged, e.g., within a box, envelope, or bag; the composition is approved by the U.S.
  • the composition is approved for administration to a mammal, e.g., a human; the composition is approved for administration to a mammal, e.g., a human, for a c-kit- and/or c-fms-mediated disease or condition; the kit of the invention includes written instructions on use and/or other indication that the composition is suitable or approved for administration to a mammal, e.g., a human, for a c-kit- and/or c-fms-mediated disease or condition; the composition is packaged in unit dose or single dose form, e.g., single dose pills, capsules, or the like.
  • the invention also provides a method for identifying or developing additional compounds active on c-kit and c-fms, e.g., improved modulators, by determining whether any of a plurality of test compounds of Formula I, Formula Ia, Formula Ib, or Formula Ig, and all sub-embodiments thereof, active on c-kit and c-fms provides an improvement in one or more desired pharmacologic properties relative to a reference compound active on c-kit and c-fms, and selecting a compound if any, that has an improvement in the desired pharmacologic property, thereby providing an improved modulator.
  • additional compounds active on c-kit and c-fms e.g., improved modulators
  • the desired pharmacologic property is serum half-life longer than 2 hr or longer than 4 hr or longer than 8 hr, aqueous solubility, oral bioavailability more than 10%, or oral bioavailability more than 20%.
  • the process can be repeated multiple times, i.e., multiple rounds of preparation of derivatives and/or selection of additional related compounds and evaluation of such further derivatives of related compounds can be carried out, e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more additional rounds.
  • the present invention also provides a method for modulating c-kit or c-fms activity by contacting c-kit or c-fms with an effective amount of a compound of Formula I (including Formula Ia, Ib, Ig and all sub-embodiments thereof) active on c-kit and/or c-fms (such as compounds developed using methods described herein).
  • the compound is preferably provided at a level sufficient to modulate the activity of the c-kit or c-fms by at least 10%, more preferably at least 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or greater than 90%.
  • the compound will be at a concentration of about 1 ⁇ M, 100 ⁇ M, or 1 mM, or in a range of 1-100 nM, 100-500 nM, 500-1000 nM, 1-100 ⁇ M, 100-500 ⁇ M, or 500-1000 ⁇ M.
  • the contacting is carried out in vitro.
  • Halo and halogen refer to all halogens, that is, chloro (Cl), fluoro (F), bromo (Br), or iodo (I).
  • Thiol refers to the group -SH.
  • “Lower alkyl” alone or in combination means an alkane-derived radical containing from 1 to 6 carbon atoms (unless specifically defined) that includes a straight chain alkyl or branched alkyl.
  • the straight chain or branched alkyl group is attached at any available point to produce a stable compound.
  • a lower alkyl is a straight or branched alkyl group containing from 1-6, 1-4, or 1-2, carbon atoms, such as methyl, ethyl, propyl, isopropyl, butyl, t-butyl, and the like.
  • Optionally substituted lower alkyl denotes lower alkyl that is independently substituted, unless indicated otherwise, with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2, or 3 substituents, attached at any available atom to produce a stable compound, wherein the substituents are selected from the group consisting of -F, -OH, -NH 2 , -NO 2 , -CN, -C(O)OH, -C(S)OH, -C(O)NH 2 , -C(S)NH 2 , -S(O) 2 NH 2 , -NHC(O)NH 2 , -NHC(O)NH 2 , -NHC(S)NH 2 , -NHS(O) 2 NH 2 , -C(NH)NH 2 , -OR a , -SR a , -OC(O)R a , -OC(S)R 8 , -C(O)R a , -C
  • substitutions include subsets of these substitutions, such as are indicated herein, for example, in the description of compounds of Formula I (including Formulae Ia, Ib, Ig and all sub-embodiments thereof), attached at any available atom to produce a stable compound.
  • fluoro substituted lower alkyl denotes a lower alkyl group substituted with one or more fluoro atoms, such as perfluoroalkyl, where preferably the lower alkyl is substituted with 1, 2, 3, 4 or 5 fluoro atoms, also 1, 2, or 3 fluoro atoms.
  • substitutions are attached at any available atom to produce a stable compound
  • substitution of the alkyl R group is such that substitution of the alkyl carbon bound to any -O-, -S-, or -N- of the moiety (except where -N- is a heteroaryl ring atom) excludes substituents that would result in any -O-, -S-, or -N- of the substituent (except where -N- is a heteroaryl ring atom) being bound to the alkyl carbon bound to any -O-, -S-, or -N- of the moiety.
  • Lower alkylene refers to a divalent alkane-derived radical containing 1-6 carbon atoms, straight chain or branched, from which two hydrogen atoms are taken from the same carbon atom or from different carbon atoms.
  • Examples of lower alkylene include, but are not limited to, methylene -CH 2 -, ethylene - CH 2 CH 2 -, propylene -CH 2 CH 2 CH 2 -, isopropylene -CH(CH 3 )CH-, and the like.
  • Optionally substituted lower alkylene denotes lower alkylene that is independently substituted, unless indicated otherwise, with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2, or 3 substituents, attached at any available atom to produce a stable compound, wherein the substituents are selected from the group consisting of -F, -OH, -NH 2 , -NO 2 , -CN, -C(O)OH, -C(S)OH, -C(O)NH 2 , -C(S)NH 2 , -S(O) 2 NH 2 , -NHC(O)NH 2 , -NHC(O)NH 2 , -NHC(S)NH 2 , -NHS(O) 2 NH 2 , -C(NH)NH 2 , -0R a , -SR a , -OC(O)R 8 , -OC(S)R a , -C(0)R a , -C(S)
  • “Lower alkenyl” alone or in combination means a straight or branched hydrocarbon containing 2-6 carbon atoms (unless specifically defined) and at least one, preferably 1-3, more preferably 1-2, most preferably one, carbon to carbon double bond. Carbon to carbon double bonds may be either contained within a straight chain or branched portion. Examples of lower alkenyl groups include ethenyl, propenyl, isopropenyl, butenyl, and the like.
  • Substituted lower alkenyl denotes lower alkenyl that is independently substituted, unless indicated otherwise, with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2, or 3 substituents, attached at any available atom to produce a stable compound, wherein the substituents are selected from the group consisting of -F, -OH, -NH 2 , -NO 2 , -CN, -C(O)OH, -C(S)OH, -C(O)NH 2 , -C(S)NH 2 , -S(O) 2 NH 2 , -NHC(O)NH 2 , -NHC(O)NH 2 , -NHC(S)NH 2 , -NHS(O) 2 NH 2 , -C(NH)NH 2 , -OR 3 , -SR ⁇ -OC(O)R 3 , -OC(S)R a , -C(O)R 3 , -C(S)R
  • substitutions include subsets of these substitutions, such as are indicated herein, for example, in the description of compounds of Formula I (including Formulae Ia, Ib, Ig and all sub-embodiments thereof), attached at any available atom to produce a stable compound.
  • fluoro substituted lower alkenyl denotes a lower alkenyl group substituted with one or more fluoro atoms, where preferably the lower alkenyl is substituted with 1, 2, 3, 4 or 5 fluoro atoms, also 1, 2, or 3 fluoro atoms.
  • substitutions are attached at any available atom to produce a stable compound
  • substitution of alkenyl groups are such that -F, -C(O)-, -C(S)-, -C(NH)-, -S(O)-, -S(O) 2 -, -0-, -S-, or -N- (except where -N- is a heteroaryl ring atom), are not bound to an alkene carbon thereof.
  • alkenyl is a substituent of another moiety or an R group of a moiety such as -OR, -NHR, -C(O)R, and the like
  • substitution of the moiety is such that any -C(O)-, -C(S)-, -S(O)-, -S(O) 2 -, -O-, -S-, or -N- thereof (except where -N- is a heteroaryl ring atom) are not bound to an alkene carbon of the alkenyl substituent or R group.
  • alkenyl is a substituent of another moiety or an R group of a moiety such as -OR, -NHR, -C(O)NHR, and the like
  • substitution of the alkenyl R group is such that substitution of the alkenyl carbon bound to any -0-, -S-, or -N- of the moiety (except where -N- is a heteroaryl ring atom) excludes substituents that would result in any -0-, -S-, or -N- of the substituent (except where -N- is a heteroaryl ring atom) being bound to the alkenyl carbon bound to any -0-, -S-, or -N- of the moiety.
  • alkenyl carbon refers to any carbon within an alkenyl group, whether saturated or part of the carbon to carbon double bond.
  • alkene carbon refers to a carbon within an alkenyl group that is part of a carbon to carbon double bond.
  • “Lower alkynyl” alone or in combination means a straight or branched hydrocarbon containing 2-6 carbon atoms (unless specifically defined) containing at least one, preferably one, carbon to carbon triple bond.
  • alkynyl groups include ethynyl, propynyl, butynyl, and the like.
  • Substituted lower alkynyl denotes lower alkynyl that is independently substituted, unless indicated otherwise, with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2, or 3 substituents, attached at any available atom to produce a stable compound, wherein the substituents are selected from the group consisting of -F, -OH, -NH 2 , -NO 2 , -CN, -C(O)OH, -C(S)OH, -C(O)NH 2 , -C(S)NH 2 , -S(O) 2 NH 2 , -NHC(O)NH 2 , -NHC(S)NH 2 , -NHS(O) 2 NH 2 , -C(NH)NH 2 , -0R a , -SR a , -OC(O)R a , -OC(S)R 3 , -C(O)R 3 , -C(S)R 3 ,
  • substitutions include subsets of these substitutions, such as are indicated herein, for example, in the description of compounds of Formula I (including Formulae Ia, Ib, Ig and all sub-embodiments thereof), attached at any available atom to produce a stable compound.
  • fluoro substituted lower alkynyl denotes a lower alkynyl group substituted with one or more fluoro atoms, where preferably the lower alkynyl is substituted with 1, 2, 3, 4 or 5 fluoro atoms, also 1, 2, or 3 fluoro atoms.
  • substitutions are attached at any available atom to produce a stable compound
  • substitution of alkynyl groups are such that -F, -C(O)-, -C(S)-, -C(NH)-, -S(O)-, -S(O) 2 -, -0-, -S-, or -N- (except where -N- is a heteroaryl ring atom), are not bound to an alkyne carbon thereof.
  • alkynyl is a substituent of another moiety or an R group of a moiety such as -OR, -NHR, -C(O)R, and the like
  • substitution of the moiety is such that any -C(O)-, -C(S)-,-S(O)-, -S(O) 2 -, -O-, -S-, or -N- thereof (except where -N- is a heteroaryl ring atom) are not bound to an alkyne carbon of the alkynyl substituent or R group.
  • alkynyl is a substituent of another moiety or an R group of a moiety such as -OR, -NHR, -C(O)NHR, and the like
  • substitution of the alkynyl R group is such that substitution of the alkynyl carbon bound to any -0-, -S-, or -N- of the moiety (except where -N- is a heteroaryl ring atom) excludes substituents that would result in any -0-, -S-, or -N- of the substituent (except where -N- is a heteroaryl ring atom) being bound to the alkynyl carbon bound to any -0-, -S-, or -N- of the moiety.
  • alkynyl carbon refers to any carbon within an alkynyl group, whether saturated or part of the carbon to carbon triple bond.
  • alkyne carbon refers to a carbon within an alkynyl group that is part of a carbon to carbon triple bond.
  • Cycloalkyl refers to saturated or unsaturated, non-aromatic monocyclic, bicyclic or tricyclic carbon ring systems of 3-10, also 3-8, more preferably 3-6, ring members per ring, such as cyclopropyl, cyclopentyl, cyclohexyl, adamantyl, and the like.
  • Cycloalkylene is a divalent cycloalkyl.
  • a “substituted cycloalkyl” is a cycloalkyl that is independently substituted, unless indicated otherwise, with one or more, preferably 1, 2, 3, 4 or 5, also 1 , 2, or 3 substituents, attached at any available atom to produce a stable compound, wherein the substituents are selected from the group consisting of halogen, -OH, -NH 2 , -NO 2 , -CN, -C(O)OH, -C(S)OH, -C(O)NH 2 , -C(S)NH 2 , -S(O) 2 NH 2 , -NHC(O)NH 2 , -NHC(O)NH 2 , -NHC(S)NH 2 , -NHS(O) 2 NH 2 , -C(NH)NH 2 , -0R a , -SR a , -0C(0)R a , -OC(S)R a , -C(O
  • Heterocycloalkyl refers to a saturated or unsaturated non-aromatic cycloalkyl group having from 5 to 10 atoms in which from 1 to 3 carbon atoms in the ring are replaced by heteroatoms of O, S or N, and are optionally fused with benzo or heteroaryl of 5-6 ring members. Heterocycloalkyl is also intended to include oxidized S or N, such as sulfmyl, sulfonyl and N-oxide of a tertiary ring nitrogen. Heterocycloalkyl is also intended to include compounds in which one of the ring carbons is oxo substituted, i.e.
  • the ring carbon is a carbonyl group, such as lactones and lactams.
  • the point of attachment of the heterocycloalkyl ring is at a carbon or nitrogen atom such that a stable ring is retained.
  • heterocycloalkyl groups include, but are not limited to, morpholino, tetrahydrofuranyl, dihydropyridinyl, piperidinyl, pyrrolidinyl, pyrrolidonyl, piperazinyl, dihydrobenzofuryl, and dihydroindolyl.
  • Heterocycloalkylene is a divalent heterocycloalkyl.
  • a “substituted heterocycloalkyl” is a heterocycloalkyl that is independently substituted, unless indicated otherwise, with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2, or 3 substituents, attached at any available atom to produce a stable compound, wherein the substituents are selected from the group consisting of halogen, -OH, -NH 2 , -NO 2 , -CN, -C(O)OH, -C(S)OH, -C(O)NH 2 , -C(S)NH 2 , -S(O) 2 NH 2 , -NHC(O)NH 2 , -NHC(O)NH 2 , -NHC(S)NH 2 , -NHS(O) 2 NH 2 , -C(NH)NH 2 , -OR 3 , -SR 3 , -OC(O)R 3 , -OC(S)R 3 , -C(O)R 3 ,
  • Substituted heterocycloalkylene is a divalent substituted heterocycloalkyl.
  • Aryl alone or in combination refers to a monocyclic or bicyclic ring system containing aromatic hydrocarbons such as phenyl or naphthyl, which may be optionally fused with a cycloalkyl of preferably 5-7, more preferably 5-6, ring members.
  • Arylene is a divalent aryl.
  • a "substituted aryl” is an aryl that is independently substituted, unless indicated otherwise, with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2, or 3 substituents, attached at any available atom to produce a stable compound, wherein the substituents are selected from the group consisting of halogen, -OH, -NH 2 , -NO 2 , -CN, -C(O)OH, -C(S)OH, -C(O)NH 2 , -C(S)NH 2 , -S(O) 2 NH 2 , -NHC(O)NH 2 , -NHC(O)NH 2 , -NHC(S)NH 2 , -NHS(O) 2 NH 2 , -C(NH)NH 2 , -OR a , -SR a , -OC(O)R 3 , -OC(S)R 3 , -C(O)R 3 , -C(S)
  • Heteroaryl alone or in combination refers to a monocyclic aromatic ring structure containing 5 or 6 ring atoms, or a bicyclic aromatic group having 8 to 10 atoms, containing one or more, preferably 1-4, more preferably 1-3, even more preferably 1-2, heteroatoms independently selected from the group consisting of O, S, and N. Heteroaryl is also intended to include oxidized S or N, such as sulfinyl, sulfonyl and N-oxide of a tertiary ring nitrogen. A carbon or nitrogen atom is the point of attachment of the heteroaryl ring structure such that a stable compound is produced.
  • heteroaryl groups include, but are not limited to, pyridinyl, pyridazinyl, pyrazinyl, quinaoxalyl, indolizinyl, benzo[b]thienyl, quinazolinyl, purinyl, indolyl, quinolinyl, pyrimidinyl, pyrrolyl, oxazolyl, thiazolyl, thienyl, isoxazolyl, oxathiadiazolyl, isothiazolyl, tetrazolyl, imidazolyl, triazinyl, furanyl, benzofuryl, and indolyl.
  • “Nitrogen containing heteroaryl” refers to heteroaryl wherein any heteroatoms are N.
  • Heteroarylene is a divalent heteroaryl.
  • a “substituted heteroaryl” is a heteroaryl that is independently substituted, unless indicated otherwise, with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2, or 3 substituents, attached at any available atom to produce a stable compound, wherein the substituents are selected from the group consisting of halogen, -OH, -NH 2 , -NO 2 , -CN, -C(O)OH, -C(S)OH, -C(O)NH 2 , -C(S)NH 2 , -S(O) 2 NH 2 , -NHC(O)NH 2 , -NHC(S)NH 2 , -NHS(O) 2 NH 2 , -C(NH)NH 2 , -0R a , -SR a ,
  • R 3 , R b , R c , -R d , -R e , -R f and -R g as used in the description of optional substituents for alkyl, alkylene, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl and heteroaryl are defined as follows:
  • each R a , R , and R c are independently selected from the group consisting of -R d , -R e , -R f , and -R g , or R b and R c combine with the nitrogen to which they are attached to form a 5-7 membered heterocycloalkyl or a 5 or 7 membered nitrogen containing heteroaryl, wherein the 5-7 membered heterocycloalkyl or 5 or 7 membered nitrogen containing heteroaryl are optionally substituted with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2, or 3 substituents selected from the group consisting of halogen, -NO 2 , -CN, -OH, -NH 2 , -OR U , -SR U , -NHR U , -NR U R U , -R x , and -R y ;
  • each -R d is independently lower alkyl, wherein lower alkyl is optionally substituted with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2 or 3 substituents selected from the group consisting of fiuoro, -OH, -NH 2 , -NO 2 , -CN, -C(O)OH, -C(S)OH, -C(O)NH 2 , -C(S)NH 2 , -S(O) 2 NH 2 , -NHC(O)NH 2 , -NHC(S)NH 2 , -NHS(O) 2 NH 2 , -C(NH)NH 2 , -0R k , -SR k , -OC(O)R k , -OC(S)R k , -C(O)R k , -C(S)R k , -C(O)OR k , -C(S)OR k , -S
  • each -R f is independently lower alkynyl, wherein lower alkynyl is optionally substituted with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2 or 3 substituents selected from the group consisting of fluoro, -OH, -NH 2 , -NO 2 , -CN, -C(O)OH, -C(S)OH, -C(O)NH 2 , -C(S)NH 2 , -S(O) 2 NH 2 , -NHC(O)NH 2 , -NHC(S)NH 2 , -NHS(O) 2 NH 2 , -C(NH)NH 2 , -OR k , -SR k , -OC(O)R k , -OC(S)R k , -C(O)R k , -C(S)R k , -C(O)OR k , -C(S)OR k ,
  • each -R g is independently selected from the group consisting of cycloalkyl, heterocycloalkyl, aryl, and heteroaryl, wherein cycloalkyl, heterocycloalkyl, aryl, and heteroaryl are optionally substituted with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2 or 3 substituents selected from the group consisting of halogen, -OH, -NH 2 , -NO 2 , -CN, -C(O)OH, -C(S)OH, -C(O)NH 2 , -C(S)NH 2 , -S(O) 2 NH 2 , -NHC(O)NH 2 , -NHC(S)NH 2 , -NHS(O) 2 NH 2 , -C(NH)NH 2 , -0R k , -SR k , -OC(O)R k , -OC(S)R k , -C(O
  • R k , R m , and R n at each occurrence are independently selected from the group consisting of -R h , -R 1 , and -R j , or R m and R n combine with the nitrogen to which they are attached form a 5-7 membered heterocycloalkyl or a 5 or 7 membered nitrogen containing heteroaryl, wherein the 5-7 membered heterocycloalkyl or 5 or 7 membered nitrogen containing heteroaryl are optionally substituted with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2, or 3 substituents selected from the group consisting of halogen, -NO 2 , -CN, -OH, -NH 2 , OR U , -SR U , -NHR U , -NR U R U , -R x , and -R y ;
  • each -R h is independently lower alkyl optionally substituted with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2, or 3 substituents selected from the group consisting of fluoro, -OH, -NH 2 , -NO 2 , -CN, -C(O)OH, -C(S)OH, -C(O)NH 2 , -C(S)NH 2 , -S(O) 2 NH 2 , -NHC(O)NH 2 , -NHC(S)NH 2 , -NHS(O) 2 NH 2 , -C(NH)NH 2 , -OR r , -SR r , -OC(O)R r , -OC(S)R r , -C(O)R r , -C(S)R r , -C(O)OR r , -C(S)OR r , -S(O)R r , -
  • each -R 1 is independently selected from the group consisting of lower alkenyl and lower alkynyl, wherein lower alkenyl or lower alkynyl are optionally substituted with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2 or 3 substituents selected from the group consisting of fluoro, -OH, -NH 2 , -NO 2 , -CN, -C(O)OH, -C(S)OH, -C(O)NH 2 , -C(S)NH 2 , -S(O) 2 NH 2 , -NHC(O)NH 2 , -NHC(O)NH 2 , -NHC(S)NH 2 , -NHS(O) 2 NH 2 , -C(NH)NH 2 , -OR r , -SR r , -OC(O)R r , -OC(S)R r , -C(O)R r , -C(S
  • each-R J is independently selected from the group consisting of cycloalkyl, heterocycloalkyl, aryl, and heteroaryl, wherein cycloalkyl, heterocycloalkyl, aryl, and heteroaryl are optionally substituted with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2 or 3 substituents selected from the group consisting of halogen, -OH, -NH 2 , -NO 2 , -CN, -C(O)OH, -C(S)OH, -C(O)NH 2 , -C(S)NH 2 , -S(O) 2 NH 2 , -NHC(O)NH 2 , -NHC(O)NH 2 , -NHC(S)NH 2 , -NHS(O) 2 NH 2 , -C(NH)NH 2 , -0R r , -SR r , -OC(O)R r , -OC(S)R
  • each R r , R s , and R* at each occurrence are independently selected from the group consisting of lower alkyl, C 3-6 alkenyl, C 3-6 alkynyl, cycloalkyl, heterocycloalkyl, aryl and heteroaryl, wherein lower alkyl is optionally substituted with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2, or 3 substituents selected from the group consisting of-R y , fluoro, -OH, -NH 2 , lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono- alkylamino, di-alkylamino, and cycloalkylamino, provided that any substitution of the lower alkyl carbon bound to any -0-, -S-, or -N-, of -0R r , -SR r , -C(O)OR r , - C(S)OR
  • each R u is independently selected from the group consisting of lower alkyl, C 3-6 alkenyl, C 3-6 alkynyl, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl, wherein lower alkyl is optionally substituted with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2, or 3 substituents selected from the group consisting of-R y , fluoro, - OH, -NH 2 , lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, di-alkylamino, and cycloalkylamino, provided that any substitution of the lower alkyl carbon bound to the -O- of -OR U , -S- of -SR U , or -N- of-NHR u is fluoro or -R y , and wherein C 3-6 alkenyl or C 3-6 alkyl
  • each -R x is selected from the group consisting of lower alkyl, lower alkenyl and lower alkynyl, wherein lower alkyl is optionally substituted with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2, or 3 substituents selected from the group consisting of-R y , fluoro, -OH, -NH 2 , lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkyl amino, di- alkyl amino, and cycloalkylamino, and wherein lower alkenyl or lower alkynyl are optionally substituted with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2, or 3 substituents selected from the group consisting of-R y , fluoro, -OH, -NH 2 , lower alkyl, fluoro substituted lower alkyl, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio
  • each — R y is selected from the group consisting of cycloalkyl, heterocycloalkyl, aryl, and heteroaryl, wherein cycloalkyl, heterocycloalkyl, aryl, and heteroaryl are optionally substituted with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2, or 3 substituents selected from the group consisting of halogen, -OH, -NH 2 , -NO 2 , -CN, lower alkyl, fluoro substituted lower alkyl, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono- alkyl amino, di-alkyl amino, and cycloalkylamino.
  • “Lower alkoxy” denotes the group -OR Z , where R z is lower alkyl.
  • “Substituted lower alkoxy” denotes lower alkoxy in which R z is lower alkyl substituted with one or more substituents as indicated herein, for example, in the description of compounds of Formula I (including Formulae Ia, Ib, Ig and all sub-embodiments thereof), including descriptions of substituted cycloalkyl, cycloheteroalkyl, aryl and heteroaryl, attached at any available atom to produce a stable compound.
  • substitution of lower alkoxy is with 1, 2, 3, 4, or 5 substituents, also 1, 2, or 3 substituents.
  • fluoro substituted lower alkoxy denotes lower alkoxy in which the lower alkyl is substituted with one or more fluoro atoms, where preferably the lower alkoxy is substituted with 1, 2, 3, 4 or 5 fluoro atoms, also 1, 2, or 3 fluoro atoms. While it is understood that substitutions on alkoxy are attached at any available atom to produce a stable compound, substitution of alkoxy is such that -O-, -S-, or -N- (except where N is a heteroaryl ring atom), are not bound to the alkyl carbon bound to the alkoxy -O-.
  • alkoxy is described as a substituent of another moiety
  • the alkoxy oxygen is not bound to a carbon atom that is bound to an -O-, -S-, or -N- of the other moiety (except where N is a heteroaryl ring atom), or to an alkene or alkyne carbon of the other moiety.
  • “Lower alkylthio” denotes the group -SR aa , where R aa is lower alkyl.
  • “Substituted lower alkylthio” denotes lower alkylthio in which R aa is lower alkyl substituted with one or more substituents as indicated herein, for example, in the description of compounds of Formula I (including Formulae Ia, Ib, Ig and all sub- embodiments thereof), including descriptions of substituted cycloalkyl, cycloheteroalkyl, aryl and heteroaryl, attached at any available atom to produce a stable compound.
  • substitution of lower alkylthio is with 1, 2, 3, 4, or 5 substituents, also 1, 2, or 3 substituents.
  • fluoro substituted lower alkylthio denotes lower alkylthio in which the lower alkyl is substituted with one or more fluoro atoms, where preferably the lower alkylthio is substituted with 1, 2, 3, 4 or 5 fluoro atoms, also 1, 2, or 3 fluoro atoms. While it is understood that substitutions on alkylthio are attached at any available atom to produce a stable compound, substitution of alkylthio is such that -O-, -S-, or -N- (except where N is a heteroaryl ring atom), are not bound to the alkyl carbon bound to the alkylthio -S-.
  • alkylthio is described as a substituent of another moiety
  • the alkylthio sulfur is not bound to a carbon atom that is bound to an -O-, -S-, or -N- of the other moiety (except where N is a heteroaryl ring atom), or to an alkene or alkyne carbon of the other moiety.
  • Amino or "amine” denotes the group -NH 2 .
  • Mono-alkylamino denotes the group -NHR bb where R bb is lower alkyl.
  • Di-alkylamino denotes the group -NR bb R cc , where R bb and R cc are independently lower alkyl.
  • Cycloalkylamino denotes the group -NR dd R ee , where R dd and R ee combine with the nitrogen to form a 5-7 membered heterocycloalkyl, where the heterocycloalkyl may contain an additional heteroatom within the ring, such as -O-, -N-, or -S-, and may also be further substituted with lower alkyl.
  • 5-7 membered heterocycloalkyl include, but are not limited to, piperidine, piperazine, 4-methylpiperazine, morpholine, and thiomorpholine.
  • c-kit-mediated disease or condition refers to a disease or condition in which the biological function of c-kit affects the development and/or course of the disease or condition, and/or in which modulation of c-kit alters the development, course, and/or symptoms.
  • mutations in the c-kit gene such as the W42, Wv, and W41 mutations reported by Herbst et al al (J. Biol. Chem., 1992, 267: 13210-13216) confer severe, intermediate, and mild phenotypic characteristics, respectively. These mutations attenuate the intrinsic tyrosine kinase activity of the receptor to different degrees and are models for the effect of modulation of c-kit activity.
  • a c-kit mediated disease or condition includes a disease or condition for which c-kit inhibition provides a therapeutic benefit, e.g. wherein treatment with c-kit inhibitors, including compounds described herein, provides a therapeutic benefit to the subject suffering from or at risk of the disease or condition.
  • c-fms-mediated disease or condition refers to a disease or condition in which the biological function of c-fms affects the development and/or course of the disease or condition, and/or in which modulation of c-fms alters the development, course, and/or symptoms.
  • the Csflr ' /Csflf mutant mouse of Dai et al (Blood, 2002, 99: 111-120) which lacks c-fms is an animal model for diseases or conditions wherein c-fins activity has been abolished.
  • a c-fins mediated disease or condition includes a disease or condition for which c-fins inhibition provides a therapeutic benefit, e.g. wherein treatment with c-fins inhibitors, including compounds described herein, provides a therapeutic benefit to the subject suffering from or at risk of the disease or condition.
  • composition refers to a formulation suitable for administration to an intended animal subject for therapeutic purposes that contains at least one pharmaceutically active compound and at least one pharmaceutically acceptable carrier or excipient.
  • pharmaceutically acceptable indicates that the indicated material does not have properties that would cause a reasonably prudent medical practitioner to avoid administration of the material to a patient, taking into consideration the disease or conditions to be treated and the respective route of administration. For example, it is commonly required that such a material be essentially sterile, e.g., for injectibles.
  • the terms "therapeutically effective” and “effective amount” indicate that the materials or amount of material is effective to prevent, alleviate, or ameliorate one or more symptoms of a disease or medical condition, and/or to prolong the survival of the subject being treated.
  • Reference to particular amino acid residues in human c-kit polypeptide is defined by the numbering corresponding to the Kit sequence in GenBank NP_000213 (SEQ ID NO: 1). Reference to particular nucleotide positions in a nucleotide sequence encoding all or a portion of c-kit is defined by the numbering corresponding to the sequence provided in GenBank NM_000222 (SEQ ID NO:2). Reference to particular amino acid residues in human c-fms polypeptide is defined by the numbering corresponding to the FMS precursor sequence in GenBank NP 005202 (SEQ ID NO:3). Reference to particular nucleotide positions in a nucleotide sequence encoding all or a portion of c-fms is defined by the numbering corresponding to the sequence provided in GenBank NM 005211 (SEQ ID NO:4).
  • kit means an enzymatically active kinase that contains a portion with greater than 90% amino acid sequence identity to amino acid residues including the ATP binding site of full-length c-kit (e.g., human c-kit, e.g., the sequence NP_000213, SEQ ID NO:1), for a maximal alignment over an equal length segment; or that contains a portion with greater than 90% amino acid sequence identity to at least 200 contiguous amino acids of native c-kit and retains kinase activity.
  • sequence identity is at least 95, 97, 98, 99, or even 100%.
  • the specified level of sequence identity is over a sequence at least 100-500, at least 200-400, or at least 300 contiguous amino acid residues in length.
  • the term includes reference to wild-type c-kit, allelic variants, and mutated forms (e.g., having activating mutations).
  • c-fms mean an enzymatically active kinase that contains a portion with greater than 90% amino acid sequence identity to amino acid residues including the ATP binding site of full-length c-fms (e.g. human c-fms, e.g. residues 20-972 of GenBank sequence NP 005202, SEQ ID NO:3), for a maximal alignment over an equal length segment; or that contains a portion with greater than 90% amino acid sequence identity to at least 200 contiguous amino acids of native c-fms and retains kinase activity.
  • sequence identity is at least 95, 97, 98, 99, or 100%.
  • the specified level of sequence identity is over a sequence at least 100-150, at least 200-400, or at least 300 contiguous amino acid residues in length.
  • the term includes wild-type c-fms, allelic variants, and mutated forms (e.g. having activating mutations).
  • the term “pFMS” refers to phosphorylated c-fms.
  • the term “c-fms activity” refers to a biological activity of c-fms, particularly including kinase activity.
  • M-CSF refers to the ligand for the c-fms RPTK
  • SCF refers to the ligand for the c-Kit RPTK.
  • c-kit kinase domain refers to a reduced length c-kit (i. e. , shorter than a full-length c-kit by at least 100 amino acids) that includes the kinase catalytic region in c-kit.
  • c-fms kinase domain refers to a c-fms of reduced length (i.e., shorter than a full-length c-fms by at least 100 amino acids) that includes the kinase catalytic region of c-fms.
  • the kinase domain retains kinase activity, preferably at least 60, 70, 80, 90, or 100% of the native c-fms kinase activity.
  • the term "the kinase” or terms of similar import relate to either c-kit or c-fms.
  • the terms "ligand” and “modulator” are used equivalently to refer to a compound that changes (i.e., increases or decreases) the activity of a target biomolecule, e.g., an enzyme such as a kinase or kinase.
  • a ligand or modulator will be a small molecule, where "small molecule” refers to a compound with a molecular weight of 1500 daltons or less, or preferably 1000 daltons or less, 800 daltons or less, or 600 daltons or less.
  • binding compound in connection with the interaction between a target and a potential binding compound indicates that the potential binding compound associates with the target to a statistically significant degree as compared to association with proteins generally (i.e., non-specific binding).
  • binding compound refers to a compound that has a statistically significant association with a target molecule.
  • a binding compound interacts with a specified target with a dissociation constant (K D ) of 1 mM or less.
  • K D dissociation constant
  • a binding compound can bind with "low affinity”, “very low affinity”, “extremely low affinity”, “moderate affinity”, “moderately high affinity”, or “high affinity” as described herein.
  • the term “greater affinity” indicates that the compound binds more tightly than a reference compound, or than the same compound in a reference condition, i.e., with a lower dissociation constant.
  • the greater affinity is at least 2, 3, 4, 5, 8, 10, 50, 100, 200, 400, 500, 1000, or 10,000-fold greater affinity.
  • the term "greater specificity" indicates that a compound binds to a specified target to a greater extent than to another biomolecule or biomolecules that may be present under relevant binding conditions, where binding to such other biomolecules produces a different biological activity than binding to the specified target.
  • the specificity is with reference to a limited set of other biomolecules, e.g., in the case of c-kit or c-frns, other tyrosine kinases or even other type of enzymes.
  • the greater specificity is at least 2, 3, 4, 5, 8, 10, 50, 100, 200, 400, 500, or 1000-fold greater specificity.
  • the term "specific for c-kit kinase”, “specific for c-kit”, and terms of like import mean that a particular compound binds to c-kit to a statistically greater extent than to other kinases that may be present in a particular sample. Also, where biological activity other than binding is indicated, the term “specific for c-kit” indicates that a particular compound has greater biological effect associated with binding c-kit than to other tyrosine kinases, e.g., kinase activity inhibition. Preferably, the specificity is also with respect to other biomolecules (not limited to tyrosine kinases) that may be present in a particular sample.
  • c-fms kinase specifically binds to c-fms to a statistically greater extent than to other kinases that may be present in a particular sample.
  • specific for c-fms indicates that a particular compound has greater biological effect associated with binding c-fms than to other tyrosine kinases, e.g., kinase activity inhibition.
  • the specificity is also with respect to other biomolecules (not limited to tyrosine kinases) that may be present in a particular sample.
  • the term “synthesizing” and like terms means chemical synthesis from one or more precursor materials.
  • enzymes can be assayed based on their ability to act upon a detectable substrate.
  • a compound or ligand can be assayed based on its ability to bind to a particular target molecule or molecules.
  • the term “modulating” or “modulate” refers to an effect of altering a biological activity, especially a biological activity associated with a particular biomolecule such as c-kit or c-fms.
  • a biological activity associated with a particular biomolecule such as c-kit or c-fms.
  • an agonist or antagonist of a particular biomolecule modulates the activity of that biomolecule, e.g., an enzyme.
  • c-kit activity refers to a biological activity of c-kit, particularly including kinase activity.
  • c-fms activity refers to a biological activity of c-fms, particularly including kinase activity.
  • the term "contacting" means that the compound(s) are caused to be in sufficient proximity to a particular molecule, complex, cell, tissue, organism, or other specified material that potential binding interactions and/or chemical reaction between the compound and other specified material can occur.
  • isolated indicates that the sequence is separated from at least a portion of the amino acid and/or nucleic acid sequences with which it would normally be associated.
  • the term "purified" indicates that the particular molecule constitutes a significantly greater proportion of the biomolecules in a composition than in a prior composition, e.g., in a cell culture.
  • the . greater proportion can be 2-fold, 5-fold, 10-fold or more greater.
  • the present invention concerns compounds of Formula I, Formula Ia, Formula Ib, or Formula Ig and all sub-embodiments thereof, that are inhibitors of c-kit, c-fms, or both c-kit and c-fms, and the use of the compounds in treating diseases that are mediated by c-kit, c-fms, or both c-kit and c-fms.
  • Exemplary compounds of Formula I, Formula Ia, Formula Ib or Formula Ig prepared following methods described in the Examples herein are as follows: Benzyl-[5-(lH- ⁇ yrrolo[2,3-b] ⁇ yridin-3-ylmethyl)- ⁇ yridin-2-yl]-amine (P-OOOl), (6-Benzylamino-pyridin-3-yl)-(lH- ⁇ yrrolo[2,3-b]pyridin-3-yl)-methanone (P-0002), [5-(lH-Pyrrolo[2,3-b]pyridin-3-ylmethyl)-pyridin-2-yl]-(4trifluoromethyl-benzyl)-ai-nine
  • the compounds described herein are useful for treating disorders related to c-kit e.g., diseases related to unregulated kinase signal transduction, including cell proliferative disorders, fibrotic disorders and metabolic disorders, among others.
  • disorders related to c-kit e.g., diseases related to unregulated kinase signal transduction, including cell proliferative disorders, fibrotic disorders and metabolic disorders, among others.
  • cell proliferative disorders which can be treated by the present invention include cancers, and mast cell proliferative disorders.
  • c-kit has also been associated with a number of different types of cancers, as described below.
  • association between abnormalities in c-kit and disease are not restricted to cancer.
  • c-kit has been associated with malignancies, including mast cell tumors, small cell lung cancer, testicular cancer, gastrointestinal stromal tumors (GISTs), glioblastoma, astrocytoma, neuroblastoma, carcinomas of the female genital tract, sarcomas of neuroectodermal origin, colorectal carcinoma, carcinoma in situ, Schwann cell neoplasia associated with neurofibromatosis, acute myelocytic leukemia, acute lymphocytic leukemia, chronic myelogenous leukemia, mastocytosis, melanoma, and canine mast cell tumors, and inflammatory diseases, including asthma, rheumatoid arthritis, allergic rhinitis, multiple sclerosis, inflammatory bowel syndrome
  • c-kit Aberrant expression and/or activation of c-kit has been implicated in a variety of cancers.
  • Evidence for a contribution of c-kit to neoplastic pathology includes its association with leukemias and mast cell tumors, small cell lung cancer, testicular cancer, and some cancers of the gastrointestinal tract and central nervous system.
  • c-kit has been implicated in playing a role in carcinogenesis of the female genital tract (Inoue, et al., 1994, Cancer Res.
  • c-kit is a useful target in treating neurofibromatosis as well as malignant tumors.
  • SCLC small cell lung carcinoma
  • c-kit kinase receptor has been found to be aberrantly expressed in many cases of small cell lung carcinoma (SCLC) cells (Hibi, et al., 1991, Oncogene 6:2291-2296).
  • SCLC small cell lung carcinoma
  • inhibition of c-kit kinase can be beneficial in treatment of SCLC, e.g., to improve the long term survival of patients with SCLC.
  • Leukemias SCF binding to the c-kit protects hematopoietic stem and progenitor cells from apoptosis (Lee, et al., 1997, J. Immunol. 159:3211-3219), thereby contributing to colony formation and hematopoiesis.
  • Expression of c-kit is frequently observed in acute myelocytic leukemia (AML), and in some cases of acute lymphocytic leukemia (ALL) (for reviews, seesperling, et al., 1997, Haemat 82:617-621; Escribano, et al., 1998, Leuk. Lymph. 30:459-466).
  • c-kit is expressed in the majority of AML cells, its expression does not appear to be prognostic of disease progression (Sperling, et al, 1997, Haemat 82:617-621). However, SCF protected AML cells from apoptosis induced by chemotherapeutic agents (Hassan, et al., 1996, Acta. Hem. 95:257-262). Inhibition of c-kit by the present invention will enhance the efficacy of these agents and can induce apoptosis of AML cells.
  • CML chronic myelogenous leukemia
  • p210 BCR"ABL Philadelphia chromosome
  • p210 BCR'ABL and c-kit both inhibit apoptosis and p62 dok has been suggested as a substrate (Carpino, et al., Cell 1997, 88:197-204)
  • clonal expansion mediated by these kinases may occur through a common signaling pathway.
  • c-kit has also been reported to interact directly with p210 BCR"ABL (Hallek, et al., Brit. J Haem. 1996, 94:5-16), which suggests that c-kit has a more causative role in CML pathology. Therefore, inhibition of c-kit will be useful in the treatment of the above disorders.
  • Gastrointestinal cancers Normal colorectal mucosa does not express c-kit (Bellone, et al., 1997, J. Cell Physiol. 172:1-11). However, c-kit is frequently expressed in colorectal carcinoma (Bellone, et al., 1997, J. Cell Physiol. 172: 1-11), and autocrine loops of SCF and c-kit have been observed in several colon carcinoma cell lines (Toyota, et al., 1993, Turn Biol 14:295-302; Lahm, et al., 1995, Cell Growth &Differ 6:1111-1118; Bellone, et al., 1997, J. Cell Physiol. 172:1-11).
  • GISTs are the most common mesenchymal tumor of the digestive system. More than 90% of GISTs express c-kit, which is consistent with the putative origin of these tumor cells from interstitial cells of Cajal (ICCs) (Hirota, et al., 1998, Science 279:577-580).
  • ICCs are thought to regulate contraction of the gastrointestinal tract, and patients lacking c-kit in their ICCs exhibited a myopathic form of chronic idiopathic intestinal pseudo-obstruction (Isozaki, et al., 1997, Amer. J. of Gast. 9 332- 334).
  • the c-kit expressed in GISTs from several different patients was observed to have mutations in the intracellular juxtamembrane domain leading to constitutive activation of c-kit (Hirota, et al., 1998, Science 279:577-580). Hence, inhibition of c-kit kinase will be an efficacious means for the treatment of these cancers.
  • Testicular cancers Male germ cell tumors have been histologically categorized into seminomas, which retain germ cell characteristics, and nonseminomas which can display characteristics of embryonal differentiation. Both seminomas and nonseminomas are thought to initiate from a preinvasive stage designated carcinoma in situ (CIS) (Murty, et al., 1998, Sem. Oncol. 25:133-144). Both c-kit and SCF have been reported to be essential for normal gonadal development during embryogenesis (Loveland, et al., 1997, J. Endocrinol 153 :337-344). Loss of either the receptor or the ligand resulted in animals devoid of germ cells.
  • CIS carcinoma in situ
  • c-kit has been found to be expressed in Leydig cells and spermatogonia, while SCF was expressed in Sertoli cells (Loveland, et al., 1997, J. Endocrinol 153:337-344).
  • Testicular tumors develop from Leydig cells with high frequency in transgenic mice expressing human papilloma virus 16 (HPVl 6) E6 and E7 oncogenes (Kondoh, et al., 1991, J. Virol. 65:3335-3339; Kondoh, et al., 1994, J. Urol. 152:2151-2154).
  • tumors express both c-kit and SCF, and an autocrine loop may contribute to the tumorigenesis (Kondoh, et al., 1995, Oncogene 10:341-347) associated with cellular loss of functional p53 and the retinoblastoma gene product by association with E6 and E7 (Dyson, et al., 1989, Science 243:934-937; Werness, et al., 1990, Science 248:76-79; Scheffher, et al., 1990, Cell 63:1129-1136).
  • Defective signaling mutants of SCF (Kondoh, et al., 1995, Oncogene 10:341-347) or c-kit (Li, et al., 1996, Cane. Res. 56:4343-4346) inhibited formation of testicular tumors in mice expressing HPV16 E6 and E7.
  • the c-kit kinase activation is pivotal to tumorigenesis in these animals and thus modulation of the c-kit kinase pathway by the present invention will prevent or treat such disorders.
  • CNS cancers SCF and c-kit are expressed throughout the CNS of developing rodents, and the pattern of expression indicates a role in growth, migration and differentiation of neuroectodermal cells. Expression of both receptor and ligand have also been reported in the adult brain (Hamel, et al., 1997, J. Neuro-Onc. 35:327-333). Expression of c-kit has also been observed in normal human brain tissue (Tada, et al. 1994, J. Neuro 80:1063-1073).
  • Glioblastoma and astrocytoma which define the majority of intracranial tumors, arise from neoplastic transformation of astrocytes (Levin, et al., 1997, Principles & Practice of Oncology:2022-2082). Expression of c-kit has been observed in glioblastoma cell lines and tissues (Berdel, et al., 1992, Cane. Res. 52:3498- 3502; Tada, et al. 1994, J. Neuro 80:1063-1073; Stanulla, et al., 1995, Act Neuropath 89:158-165).
  • mast cells and eosinophils represent key cells involved in allergy, inflammation and asthma (Thomas, et al., 1996, Gen.
  • SCF SCF
  • c-kit directly and indirectly regulates activation of both mast cells and eosinophils, thereby influencing the primary cells involved in allergy and asthma through multiple mechanisms. Because of this mutual regulation of mast cell and eosinophil function, and the role that SCF can play in this regulation, inhibition of c-kit can be used to treat allergy-associated chronic rhinitis, inflammation and asthma.
  • mast cell growth factor also known as mast cell growth factor
  • Mastocytosis is limited to the skin in the majority of patients, but can involve other organs in 15-20% of patients (Valent, 1996, Wein/Klin Klischr 108:385-397; Golkar, et al., 1997, Lancet 349:1379-1385). Even among patients with systemic mastocytosis, the disease can range from having a relatively benign prognosis to aggressive mastocytosis and mast cell leukemia. (Valent, 1996, Wein/Klin Klischr 108:385-397; Golkar, et al., 1997, Lancet 349:1379-1385). c-kit has been observed on malignant mast cells from canine mast cell tumors (London, et al., 1996, J.
  • SCF has been shown to be expressed on stromal cells as a membrane-bound protein, and its expression can be induced by fibrogenic growth factors such as PDGF. It has also been shown to be expressed on keratinocytes as a membrane-bound protein in normal skin. However, in the skin of patients with mastocytosis, an increased amount of soluble SCF has been observed (Longley, et al., 1993, New Engl. J. Med. 328:1302-1307). [0134] Mast cell chymase has been reported to cleave membrane-associated SCF to a soluble and biologically active form.
  • This mast cell-mediated process can generate a feedback loop to enhance mast cell proliferation and function (Longley, et al., 1997, Proc. Natl. Acad. Sci. 94:9017-9021), and maybe important for the etiology of mastocytosis.
  • Transgenic mice overexpressing a form of SCF that could not be proteolytically released from keratinocytes did not develop mastocytosis, while similar animals expressing normal SCF in keratinocytes exhibited a phenotype resembling human cutaneous mastocytosis (Kunisada, et al., 1998, J. Exp. Med. 187:1565-1573).
  • Formation of large amounts of soluble SCF can contribute to the pathology associated with mastocytosis in some patients and the present invention can treat or prevent such disorders by modulating the interaction between SCF and c-kit kinase.
  • c-kit Several different mutations of c-kit that resulted in constitutive kinase activity have been found in human and rodent mast cell tumor cell lines (Furitsu, et al., 1993, J. Clin. Invest. 92:1736-1744; Tsujimura, et al., 1994, Blood 9:2619- 2626; Tsujimura, et al., 1995, Int. Arch. Aller.
  • activating mutations of c-kit may be responsible for the pathogenesis of the disease and these patients can be treated, or their diseases prevented, by modulation of the SCF interaction with c-kit kinase.
  • SCF activation of c-kit as been shown to prevent mast cell apoptosis which may be critical for maintaining cutaneous mast cell homeostasis (Iemura, et al., 1994, Amer. J. Pathol 144:321-328; Yee, et al., 1994, J. Exp. Med. 179:1777-1787; Mekori, et al., 1994, J. Immunol 153:2194-2203; Mekori, et al., 1995, Int. Arch.
  • c-kit inhibitors can be used against both wild-type c-kit as well as c-kit having mutations, e.g., activating mutations in the regulatory region and/or catalytic region.
  • Asthma & Allergy Mast cells and eosinophils represent key cells in parasitic infection, allergy, inflammation, and asthma (Thomas, et al., 1996, Gen. Pharmacol 27:593-597; Metcalfe, et al., 1997, Physiol Rev 77:1033-1079; Holgate, 1997, CIBA Found. Symp.; Naclerio, et al, 1997, JAMA 278:1842-1848; Costa, et al., 1997, JAMA 778:1815-1822). SCF has been shown to be essential for mast cell development, survival and growth (Kitamura, et al., 1995, Int. Arch. Aller. Immunol.
  • SCF cooperates with the eosinophil-specific regulator, IL-5, to increase the development of eosinophil progenitors (Metcalf, et al., 1998, Proc. Natl. Acad. Sci., USA 95:6408-6412). SCF has also been reported to induce mast cells to secrete factors (Okayama, et al., 1997, Lit. Arch. Aller. Immunol. 114:75-77; Okayama, et al., 1998, Eur. J. Immunol.
  • SCF induces mediator release from mast cells, as well as priming these cells for IgE-induced degranulation (Columbo, et al., 1992, J. Immunol 149:599-602) and sensitizing their responsiveness to eosinophil-derived granule major basic protein (Furuta, et al., 1998, Blood 92:1055-1061).
  • factors released by activated mast cells are IL-5, GM-CSF and TNF- ⁇ , which influence eosinophil protein secretion (Okayama, et al., 1997, Int. Arch. Aller. Immunol. 114:75-77; Okayama, et al., 1998, Eur. J. Immunol.
  • SCF promotes the mast cell production of the eosinophil chemotactic factor, eotaxin (Hogaboam, et al., 1998, J. Immunol. 160:6166-6171), and eosinophil infiltration (Luckacs, et al., 1996, J. Immunol. 156:3945-3951).
  • SCF also directly influences the adhesion of both mast cells (Dastych, et al., 1994, J. Immunol. 152:213-219; Kinashi, et al., 1994, Blood 83:1033-1038) and eosinophils (Yuan, et al., 1997, J. Exp. Med. 186:313-323), which in turn, regulates tissue infiltration.
  • SCF can influence the primary cells involved in allergy and asthma through multiple mechanisms.
  • corticosteroids are the most effective treatment for chronic rhinitis and inflammation associated with allergy (Naclerio, et al., 1997, JAMA 278:1842-1848; Meltzer, 1997, Aller. 52:33-40).
  • Inflammatory arthritis e.g. rheumatoid arthritis: Due to the association of mast cells with the arthritic process (Lee et al., 2002, Science 297:1689-1692), c-kit provides a useful target for prevention, delay, and/or treatment of inflammatory arthritis, such as rheumatoid arthritis.
  • Mast cells have been shown to play an extensive role in autoimmune diseases, as demonstrated in the mouse model of multiple sclerosis (MS), experimental allergic encephalomyelitis (EAE). Mast cells were indicated to be required for full manifestation of the disease. Secor et al., 2000, J Exp Med 191:813-821. Thus, c-kit also provides a useful target for the prevention, delay, and/or treatment of multiple sclerosis.
  • c-fms has been associated with a number of different types of diseases.
  • c-fms has been associated with immune disorders, including rheumatoid arthritis, systemic lupus erythematosis (SLE), Wegener's granulomatosis, and transplant rejection, inflammatory diseases including Chronic Obstructive Pulmonary Disease (COPD), emphysema, and atherosclerosis, metabolic disorders, including insulin resistance, hyperglycemia, and lipolysis, disorders of bone structure or mineralization, including osteoporosis, increased risk of fracture, hypercalcemia, and bone metastases, kidney diseases, including nephritis (e.g.
  • glomerulonephritis including interstitial nephritis, Lupus nephritis), tubular necrosis, diabetes-associated renal complications, and hypertrophy and cancers, including multiple myeloma, acute myeloid leukemia, chronic myeloid leukemia (CML), breast cancer, and ovarian cancer.
  • CML chronic myeloid leukemia
  • a condition related to AML is chronic myeloid leukemia (CML).
  • CML chronic myeloid leukemia
  • BC myeloid blast crisis
  • non-random additional chromosome abnormalities occur in over 80% of patients.
  • these cytogenetic changes have been reported to precede the clinical signs of CML-BC by several months to years suggesting that other biological events may participate in the multistep process of acute transformation of CML.
  • the autocrine production of growth factors has been shown to occur in several hematological malignancies and particularly in AML. Specchia et al [Br J Haematol.
  • COPD is characterized by airflow limitation that is not fully reversible.
  • the airflow limitation is usually progressive and associated with an abnormal inflammatory response of the lungs to noxious particles or gases.
  • the chronic inflammation of COPD is observed through the airways, parenchyma, and pulmonary vasculature.
  • the inflammatory cell population consists of neutrophils, macrophages, and T lymphocytes, along with eosinophils in some patients.
  • Macrophages are postulated to play an orchestrating role in COPD inflammation by releasing mediators such as TNF- ⁇ , IL-8 and LTB4, which are capable of damaging lung structures and/or sustaining neutrophilic inflammation.
  • M-CSF/Fms signaling is critical to osteoclast formation and survival of osteoclast precursors.
  • estrogen loss in menopause results in increased M-CSF and thus increased osteoclast number and bone resorption which leads to increased risk of fracture and osteoporosis.
  • blockage of this signal is a target for the inhibition of bone resorption (Teitelbaum, Science. 2000;289:1504; Rohan, Science. 2000;289:1508.)
  • Atherosclerosis an inflammatory disease of the vessel walls, is associated with significant morbidity and mortality.
  • a effect for c-fms inhibition in the treatment and prevention of atherosclerosis depends on several observations (Libby, Nature. 2002;420: 868-874.)
  • monocytes resident in the arterial intima increase expression of scavenger receptors and internalize modified lipoproteins.
  • the resulting lipid-laden macrophages develop into foam cells characteristic of the atherosclerotic lesion.
  • Macrophages in atheroma secrete cytokines and growth factors involved in lesion progression. Additionally, macrophages replicate within the intima.
  • M-CSF activates the transition from monocyte to lipid-laden macrophage and augments expression of scavenger receptor A.
  • atherosclerotic plaques over-express M-CSF which is critical for atherosclerotic progression.
  • Mice deficient in M-CSF have been found to experience less severe atherosclerosis than mice with normal M-CSF (Rajavashisth, et. al., J. Clin. Invest. 1998;101 -.2702-2710; Qiao, et. al., Am. J. Path. 1997;150:1687-1699).
  • inhibitors of c-fins disrupt M-CSF signaling, compromising monocyte to macrophage foam cell progression, macrophage survival and r eplication, and cytokine signaling that participates in lesion progression.
  • Wegener's granulomatosis also known as vasculitis, is characterized by granulomatous inflammation of the blood vessels with necrosis. This inflammation limits blood flow to organs with consequent damage. Although the disease can involve any organ system, Wegener's granulomatosis mainly affects the respiratory tract (i.e., sinuses, nose, trachea, and lungs) and the kidneys.
  • the endothelium plays a central role in the immunopathology of several vascular disorders in many inflammatory conditions such as Wegener's granulomatosis in which use of intravenous immunoglobulin (IV Ig) has been shown to be beneficial (see e.g., Basta et al, J Clin Invest 1994, 94:1729-1735). It has been reported (Xu et al, Am. J. Path.
  • IV Ig inhibits endothelial cell proliferation in a dose- and time-dependent manner and down-regulates the expression of adhesion molecule mRNA (ICAM-I and VCAM-I), chemokine mRNA (MCP-I, M-CSF, and GM-CSF), and proinflammatory cytokine mRNA (TNF-w, IL-IB, and IL-6) induced by TNF- ⁇ : or IL-IB.
  • IAM-I and VCAM-I adhesion molecule mRNA
  • MCP-I chemokine mRNA
  • MCP-I chemokine mRNA
  • GM-CSF chemokine mRNA
  • TNF-w proinflammatory cytokine mRNA
  • IL-IB proinflammatory cytokine mRNA
  • M-CSF alveolar macrophages
  • c-fins c-fins in emphysema
  • M-CSF has a role in the modulation of the accumulation and function of alveolar macrophages (AMs) in vivo (Shibata et al, Blood 2001, 98: pp. 2845-2852).
  • AMs alveolar macrophages
  • Osteopetrotic (Op/Op) mice have no detectable M-CSF and show variable tissue-specific reductions in macrophage numbers. Accordingly, it was hypothesized that AMs would be decreased in number and have altered function in Op/Op mice because of the absence of M-CSF.
  • Metastatic cancer cells cause bone destruction, with associated fracture, pain, deformation, and hypercalcaemia, due to production of osteoclasticogenic factors including M-CSF by tumor cells (Clohisy et al, Clin. Orthop. 2000, 373: 104-14). Binding of M-CSF to the c-fms product stimulates formation of osteoclasts and osteolytic activity (Kodama et al, J. Exp,. Med. 1991, 173: 269-72; Feng et al, Endocrinology 2002, 143: 4868-74). Accordingly, inhibition of osteoclast activity at the level of c-fms offers a compelling target for amelioration of bone metastasis.
  • Macrophage accumulation is a prominent feature in many forms of glomerulonephritis.
  • Local proliferation of macrophages within the kidney has been described in human and experimental glomerulonephritis and may have an important role in augmenting the inflammatory response.
  • Isbel et al (Nephrol Dial Transplant 2001, 16: 1638-1647) examined the relationship between local macrophage proliferation and renal expression of M-CSF. Glomerular and tubulointerstitial M-CSF expression was found to be up-regulated in human glomerulonephritis, being most prominent in proliferative forms of disease.
  • Insulin resistance and obesity are hallmark of type II diabetes and there is a strong correlation between insulin resistance and abdominal visceral fat accumulation (Bjorntrop, Diabetes Metab. Res. Rev., 1999, 15: 427-441).
  • Current evidence indicates that macrophages accumulating in adipose tissue release TNF-a and other factors that cause adipocyte changes (hypertrophy, lipolysis, reduced insulin sensitivity) and also promote insulin resistance in surrounding tissues. Therefore, macrophage accumulation in type 2 diabetes is important for disease progression. Accordingly, inhibition of c-fms has potential in preventing the development of insulin resistance and hyperglycemia.
  • imatinib also specifically targets the macrophage colony stimulating factor receptor, c-fms, at therapeutic concentrations. Although this finding has important implications with regard to potential side effects in patients currently receiving imatinib therapy, these results suggest that imatinib may also be useful in the treatment of diseases where c-fms is implicated. This includes breast and ovarian cancer and inflammatory conditions such as rheumatoid arthritis. Dewar et al. also speculate that imatinib may be used in diseases where bone destruction occurs due to excessive osteoclast activity, such as in the haematologic malignancy, multiple myeloma (Dewar et al., Cell Cycle 2005, 4(7):851-3).
  • modulators of both c-fms and c-kit function can be used against diseases such as those indicated above, where in some instances, the dual activity of the modulator for both c-fms and c-kit provides distinct advantages in treating such diseases.
  • the complementary activities provided by a single compound would provide added benefits over compounds targeting one or the other activity, or separate compounds targeting these activities. For example, by attenuating release of macrophage chemo-attractants by mast cells or mast cell chemoattractants by macrophages, these anti-inflammatory effects would synergize with the concomitant inhibition of intrinsic cellular function. Limitations in coadministration are absent in a dual inhibitor. Further, the dual activity may result in much lower effective doses for treatment. II. Production of c-kit and c-fms related Polypeptides
  • the native and mutated kinase polypeptides described herein may be chemically synthesized in whole or part using techniques that are well-known in the art (see, e.g., Creighton (1983) Biopolymers 22(l):49-58).
  • a variety of host-expression vector systems may be utilized to express the kinase coding sequence. These include but are not limited to microorganisms such as bacteria transformed with recombinant bacteriophage DNA, plasmid DNA or cosmid DNA expression vectors containing the kinase domain coding sequence; yeast transformed with recombinant yeast expression vectors containing the kinase domain coding sequence; insect cell systems infected with recombinant virus expression vectors (e.g. baculovirus) containing the kinase domain coding sequence; plant cell systems infected with recombinant virus expression vectors (e.g.
  • plasmid expression vectors e.g. Ti plasmid
  • the expression elements of these systems vary in their strength and specificities.
  • any of a number of suitable transcription and translation elements may be used in the expression vector.
  • inducible promoters such as pL of bacteriophage ⁇ plac, ptrp, ptac (ptrp-lac hybrid promoter) and the like may be used;
  • promoters such as the baculovirus polyhedrin promoter may be used;
  • promoters derived from the genome of plant cells e.g.
  • heat shock promoters the promoter for the small subunit of RUBISCO; the promoter for the chlorophyll a/b binding protein) or from plant viruses (e.g. the 35S RNA promoter of CaMV; the coat protein promoter of TMV) may be used; when cloning in mammalian cell systems, promoters derived from the genome of mammalian cells (e.g. metallothionein promoter) or from mammalian viruses (e.g.
  • the adenovirus late promoter may be used; when generating cell lines that contain multiple copies of the kinase domain DNA, SV4O-, BPV- and EBV-based vectors may be used with an appropriate selectable marker.
  • the methods of the present invention can involve assays that are able to detect the binding of compounds to a target molecule. Such binding is at a statistically significant level, preferably with a confidence level of at least 90%, more preferably at least 95, 97, 98, 99% or greater confidence level that the assay signal represents binding to the target molecule, i.e., is distinguished from background. Preferably controls are used to distinguish target binding from non-specific binding. A large variety of assays indicative of binding are known for different target types and can be used for this invention.
  • Binding compounds can be characterized by their effect on the activity of the target molecule.
  • a “low activity” compound has an inhibitory concentration (IC50) or effective concentration (EC 50 ) of greater than 1 ⁇ M under standard conditions.
  • very low activity is meant an IC 50 or EC 50 of above 100 ⁇ M under standard conditions.
  • extreme low activity is meant an IC 50 or EC 50 of above 1 mM under standard conditions.
  • moderate activity is meant an IC 50 or EC 50 of 200 nM to 1 ⁇ M under standard conditions.
  • Moderately high activity is meant an IC 50 or EC 50 of 1 nM to 200 nM.
  • high activity is meant an IC 50 or EC 50 of below 1 nM under standard conditions.
  • the IC 50 or EC 5O is defined as the concentration of compound at which 50% of the activity of the target molecule (e.g. enzyme or other protein) activity being measured is lost or gained relative to the range of activity observed when no compound is present.
  • Activity can be measured using methods known to those of ordinary skill in the art, e.g., by measuring any detectable product or signal produced by occurrence of an enzymatic reaction, or other activity by a protein being measured.
  • background signal in reference to a binding assay is meant the signal that is recorded under standard conditions for the particular assay in the absence of a test compound, molecular scaffold, or ligand that binds to the target molecule.
  • background signal in reference to a binding assay is meant the signal that is recorded under standard conditions for the particular assay in the absence of a test compound, molecular scaffold, or ligand that binds to the target molecule.
  • standard deviation is meant the square root of the variance.
  • Binding parameters can be measured using surface plasmon resonance, for example, with a BIAcore ® chip (Biacore, Japan) coated with immobilized binding components.
  • Surface plasmon resonance is used to characterize the microscopic association and dissociation constants of reaction between an sFv or other ligand directed against target molecules.
  • Such methods are generally described in the following references which are incorporated herein by reference. VeIy F. et al., (2000) BIAcore ® analysis to test phosphopeptide-SH2 domain interactions, Methods in Molecular Biology. 121:313-21; Liparoto et al., (1999) Biosensor analysis of the interleukin-2 receptor complex, Journal of Molecular Recognition.
  • BIAcore ® uses the optical properties of surface plasmon resonance (SPR) to detect alterations in protein concentration bound to a dextran matrix lying on the surface of a gold/glass sensor chip interface, a dextran biosensor matrix.
  • SPR surface plasmon resonance
  • proteins are covalently bound to the dextran matrix at a known concentration and a ligand for the protein is injected through the dextran matrix.
  • Near infrared light, directed onto the opposite side of the sensor chip surface is reflected and also induces an evanescent wave in the gold film, which in turn, causes an intensity dip in the reflected light at a particular angle known as the resonance angle. If the refractive index of the sensor chip surface is altered (e.g.
  • This angle shift can be measured and is expressed as resonance units (RUs) such that 1000 RUs is equivalent to a change in surface protein concentration of 1 ng/mm . These changes are displayed with respect to time along the y-axis of a sensorgram, which depicts the association and dissociation of any biological reaction.
  • HTS typically uses automated assays to search through large numbers of compounds for a desired activity.
  • HTS assays are used to find new drugs by screening for chemicals that act on a particular enzyme or molecule. For example, if a chemical inactivates an enzyme it might prove to be effective in preventing a process in a cell which causes a disease.
  • High throughput methods enable researchers to assay thousands of different chemicals against each target molecule very quickly using robotic handling systems and automated analysis of results.
  • “high throughput screening” or “HTS” refers to the rapid in vitro screening of large numbers of compounds (libraries); generally tens to hundreds of thousands of compounds, using robotic screening assays.
  • Ultra high-throughput Screening (uHTS) generally refers to the high-throughput screening accelerated to greater than 100,000 tests per day.
  • a multicontainer carrier facilitates measuring reactions of a plurality of candidate compounds simultaneously.
  • Multi-well microplates may be used as the carrier. Such multi-well microplates, and methods for their use in numerous assays, are both known in the art and commercially available.
  • Screening assays may include controls for purposes of calibration and confirmation of proper manipulation of the components of the assay. Blank wells that contain all of the reactants but no member of the chemical library are usually included.
  • a known inhibitor (or activator) of an enzyme for which modulators are sought can be incubated with one sample of the assay, and the resulting decrease (or increase) in the enzyme activity used as a comparator or control.
  • modulators can also be combined with the enzyme activators or inhibitors to find modulators which inhibit the enzyme activation or repression that is otherwise caused by the presence of the known the enzyme modulator.
  • Spectrophotometric and spectrofluorometric assays are well known in the art. Examples of such assays include the use of colorimetric assays for the detection of peroxides, as described in Gordon, A. J. and Ford, R. A., (1972) The Chemist's Companion: A Handbook Of Practical Data, Techniques, And References, John Wiley and Sons, N.Y., Page 437.
  • Fluorescence spectrometry may be used to monitor the generation of reaction products. Fluorescence methodology is generally more sensitive than the absorption methodology. The use of fluorescent probes is well known to those skilled in the art. For reviews, see Bashford et al., (1987) Spectrophotometry and Spectrofluorometrv: A Practical Approach, pp. 91-114, IRL Press Ltd.; and Bell, (1981) Spectroscopy In Biochemistry. Vol. I, pp. 155-194, CRC Press.
  • SMase activity can be detected using the Amplex ® Red reagent (Molecular Probes, Eugene, OR). In order to measure sphingomyelinase activity using Amplex ® Red, the following reactions occur. First, SMase hydrolyzes sphingomyelin to yield ceramide and phosphorylcholine. Second, alkaline phosphatase hydrolyzes phosphorylcholine to yield choline.
  • choline is oxidized by choline oxidase to betaine.
  • H 2 O 2 in the presence of horseradish peroxidase, reacts with Amplex ® Red to produce the fluorescent product, Resorufm, and the signal therefrom is detected using spectrofluorometry.
  • Fluorescence polarization is based on a decrease in the speed of molecular rotation of a fluorophore that occurs upon binding to a larger molecule, such as a receptor protein, allowing for polarized fluorescent emission by the bound ligand.
  • FP is empirically determined by measuring the vertical and horizontal components of fluorophore emission following excitation with plane polarized light. Polarized emission is increased when the molecular rotation of a fluorophore is reduced.
  • a fluorophore produces a larger polarized signal when it is bound to a larger molecule (i.e. a receptor), slowing molecular rotation of the fluorophore.
  • the magnitude of the polarized signal relates quantitatively to the extent of fluorescent ligand binding. Accordingly, polarization of the "bound" signal depends on maintenance of high affinity binding.
  • FP is a homogeneous technology and reactions are very rapid, taking seconds to minutes to reach equilibrium. The reagents are stable, and large batches may be prepared, resulting in high reproducibility. Because of these properties, FP has proven to be highly automatable, often performed with a single incubation with a single, premixed, tracer- receptor reagent. For a review, see Owickiet al., (1997), Application of Fluorescence Polarization Assays in High-Throughput Screening, Genetic Engineering News, 17:27. [0177] FP is particularly desirable since its readout is independent of the emission intensity (Checovich, W. J., et al., (1995) Nature 375:254-256; Dandliker, W.
  • FP and FRET are well-suited for identifying compounds that block interactions between sphingolipid receptors and their ligands. See, for example, Parker et al., (2000) Development of high throughput screening assays using fluorescence polarization: nuclear receptor-ligand-binding and kinase/phosphatase assays, J Biomol Screen 5:77-88.
  • Fluorophores derived from sphingolipids that may be used in FP assays are commercially available.
  • Molecular Probes (Eugene, OR) currently sells sphingomyelin and one ceramide flurophores.
  • N-(4,4-difluoro- 5,7-dimethyl-4-bora-3 a,4a-diaza-s-indacene- 3-pentanoyl)sphingosyl phosphocholine BODIPY® FL C5-sphingomyelin
  • N-(4,4-difluoro-5,7-dimethyl-4-bora-3a,4a-diaza-s- indacene- 3-dodecanoyl)sphingosyl phosphocholine BODIPY® FL C12-sphingomyelin
  • N-(4,4-difluoro-5,7-dimethyl-4-bora-3 a,4a-diaza-s-indacene- 3 -pentanoyl)sphingosine BODIPY ® FL C5-ceramide
  • U.S. Patent No. 4,150,949 discloses fluorescein-labelled gentamicins, including fluoresceinthiocarbanyl gentamicin. Additional fluorophores may be prepared using methods well known to the skilled artisan.
  • Exemplary normal-and-polarized fluorescence readers include the POLARION ® fluorescence polarization system (Tecan AG, Hombrechtikon, Switzerland).
  • General multiwell plate readers for other assays are available, such as the VERSAMAX ® reader and the SPECTRAMAX ® multiwell plate spectrophotometer (both from Molecular Devices).
  • Fluorescence resonance energy transfer is another useful assay for detecting interaction and has been described. See, e.g., Heim et al., (1996) Curr. Biol. 6:178-182; Mitra et al., (1996) Gene 173:13-17; and Selvin et al., (1995) Meth. Enzymol. 246:300-345.
  • FRET detects the transfer of energy between two fluorescent substances in close proximity, having known excitation and emission wavelengths.
  • a protein can be expressed as a fusion protein with green fluorescent protein (GFP).
  • GFP green fluorescent protein
  • the resonance energy can be transferred from one excited molecule to the other.
  • the emission spectrum of the sample shifts, which can be measured by a fluorometer, such as a fMAX multiwell fluorometer (Molecular Devices, Sunnyvale Calif.).
  • SPA Scintillation proximity assay
  • SPA is a particularly useful assay for detecting an interaction with the target molecule.
  • SPA is widely used in the pharmaceutical industry and has been described (Hanselman et al., (1997) J. Lipid Res. 38:2365-2373; Kahl et al., (1996) Anal. Biochem. 243:282-283; Undenfriend et al., (1987) Anal. Biochem. 161:494- 500). See also U.S. Patent Nos. 4,626,513 and 4,568,649, and European Patent No. 0,154,734.
  • FLASHPLATE ® scintillant-coated plates NN Life Science Products, Boston, MA).
  • the target molecule can be bound to the scintillator plates by a variety of well known means. Scintillant plates are available that are derivatized to bind to fusion proteins such as GST, His6 or Flag fusion proteins. Where the target molecule is a protein complex or a multimer, one protein or subunit can be attached to the plate first, then the other components of the complex added later under binding conditions, resulting in a bound complex.
  • the gene products in the expression pool will have been radiolabeled and added to the wells, and allowed to interact with the solid phase, which is the immobilized target molecule and scintillant coating in the wells.
  • the assay can be measured immediately or allowed to reach equilibrium. Either way, when a radiolabel becomes sufficiently close to the scintillant coating, it produces a signal detectable by a device such as a TOPCOUNT NXT ® microplate scintillation counter (Packard BioScience Co., Meriden Conn.). If a radiolabeled expression product binds to the target molecule, the radiolabel remains in proximity to the scintillant long enough to produce a detectable signal.
  • a number of different assays for kinase activity can be utilized for assaying for active modulators and/or determining specificity of a modulator for a particular kinase or group or kinases.
  • assays for kinase activity can be utilized for assaying for active modulators and/or determining specificity of a modulator for a particular kinase or group or kinases.
  • one of ordinary skill in the art will know of other assays that can be utilized and can modify an assay for a particular application. For example, numerous papers concerning kinases described assays that can be used.
  • Additional alternative assays can employ binding determinations.
  • this sort of assay can be formatted either in a fluorescence resonance energy transfer (FRET) format, or using an AlphaScreen (amplified /uminescent proximity homogeneous assay) format by varying the donor and acceptor reagents that are attached to streptavidin or the phospho-specific antibody.
  • FRET fluorescence resonance energy transfer
  • AlphaScreen amplified /uminescent proximity homogeneous assay
  • solvents include polar and non-polar solvents known to those of skill in the art, including polar aprotic and polar protic solvents.
  • Polar solvents include, without limitation, protic solvents such as methanol, ethanol, isopropyl alcohol, t-butanol, n-butanol, acetic acid, formic acid or water, or aprotic solvents such as tetrahydrofuran (THF), acetonitrile, dioxane, methylene chloride, dimethylsulfoxide (DMSO), acetone, N,N-dimethylformamide (DMF), N 5 N- dimethylacetamide (DMA), ethyl acetate, 1,2-dimethoxyethane, 1,2-dichloroethane, chloroform, 1 ,2-dichloroethane, or pyridine.
  • protic solvents such as methanol, ethanol, isopropyl alcohol, t-butanol, n
  • Polar solvents include a mixture of water with any of the above, or a mixture of any two or more of the above.
  • Apolar solvents include, without limitation, toluene, benzene, chlorobenzene, xylenes and hexanes.
  • reducing agent includes, without limitation, a reducing agent such as catalytic reducing agents using hydrogen and transition metal catalysts such as palladium, platinum, rhodium, etc.(e.g. Pt/acetic acid/H 2 ); a mixture of trifluoroacetic acid and triethylsilane, borane tetrahydrofuran complex, diborane, borane dimethylsulfide complex, and a combination of sodium borohydride and boron trifiuoride; metals such as reduced iron, zinc powder, magnesium etc.; metal hydrogen complex compounds such as alkali metal borohydrides (for example, potassium borohydride, sodium borohydride, lithium borohydride, zinc borohydride, sodium triacetoxyborohydride, etc.), aluminum lithium hydride, etc.; metal hydrides such as sodium hydride, etc.; organic tin compounds (triphenyltin hydride, etc.); and
  • oxidizing agent includes, without limitation, an oxidizing agent such as Dess-Martin reagent, TEMPO (2,2,6,6- tetramethylpiperidine-N-oxide), DDQ (2,3-Dichloro-5,6-dicyano-l,4-benzoquinone), PDC (pyridinium dichromate), PCC (pyridinium chlorochromate), Pyridine.SC ⁇ , Chromium trioxide, p-nitroperbenzoic acid, magnesium monoperoxyphthalate, sodium periodate, potassium periodate, hydrogen peroxide, urea peroxide, alkali metal bromates, cumene hydroperoxide, tert-butyl peroxide, peracids such as performic acid, peracetic acid, pertrifluoroacetic acid, perbenzoic acid, m-chloroperbenzoic acid, o-carboxyperbenzoic acid and the like; sodium metaperiodate,
  • some of the compounds according to the present invention may exist as stereoisomers, i.e. they have the same sequence of covalently bonded atoms and differ in the spatial orientation of the atoms.
  • compounds may be optical stereoisomers, which contain one or more chiral centers, and therefore, may exist in two or more stereoisomeric forms (e.g. enantiomers or diastereomers).
  • stereoisomers i.e., essentially free of other stereoisomers
  • racemates i.e., essentially free of other stereoisomers
  • mixtures of enantiomers and/or diastereomers are examples of compounds according to the present invention.
  • stereoisomers include geometric isomers, such as cis- or trans- orientation of substituents on adjacent carbons of a double bond. All such single stereoisomers, racemates and mixtures thereof are intended to be within the scope of the present invention. Unless specified to the contrary, all such steroisomeric forms are included within the formulae provided herein.
  • a chiral compound of the present invention is in a form that contains at least 80% of a single isomer (60% enantiomeric excess ("e.e.") or diastereomeric excess (“d.e.”)), or at least 85% (70% e.e. or d.e.), 90% (80% e.e. or d.e.),
  • an optically pure compound having one chiral center is one that consists essentially of one of the two possible enantiomers (i.e., is enantiomerically pure), and an optically pure compound having more than one chiral center is one that is both diastereomerically pure and enantiomerically pure.
  • the compound is present in optically pure form.
  • the addition may occur at either of the double bond-linked atoms.
  • the present invention includes both such regioisomers.
  • the formulae are intended to cover solvated as well as unsolvated forms of the identified structures.
  • the indicated structures include both hydrated and non-hydrated forms.
  • Other examples of solvates include the structures in combination with isopropanol, ethanol, methanol, DMSO, ethyl acetate, acetic acid, or ethanolamine.
  • the invention also includes prodrugs (generally pharmaceutically acceptable prodrugs), active metabolic derivatives (active metabolites), and their pharmaceutically acceptable salts.
  • Prodrugs are compounds or pharmaceutically acceptable salts thereof which, when metabolized under physiological conditions or when converted by solvolysis, yield the desired active compound.
  • the prodrug is inactive, or less active than the active compound, but may provide advantageous handling, administration, or metabolic properties.
  • some prodrugs are esters of the active compound; during metabolysis, the ester group is cleaved to yield the active drug.
  • some prodrugs are activated enzymatically to yield the active compound, or a compound which, upon further chemical reaction, yields the active compound.
  • bioprecursor prodrugs can be conceptually divided into two non-exclusive categories, bioprecursor prodrugs and carrier prodrugs.
  • bioprecursor prodrugs are compounds that are inactive or have low activity compared to the corresponding active drug compound, that contain one or more protective groups and are converted to an active form by metabolism or solvolysis. Both the active drug form and any released metabolic products should have acceptably low toxicity.
  • active drag compound involves a metabolic process or reaction that is one of the follow types:
  • Oxidative reactions are exemplified without limitation to reactions such as oxidation of alcohol, carbonyl, and acid functions, hydroxylation of aliphatic carbons, hydroxylation of alicyclic carbon atoms, oxidation of aromatic carbon atoms, oxidation of carbon-carbon double bonds, oxidation of nitrogen-containing functional groups, oxidation of silicon, phosphorus, arsenic, and sulfur, oxidative N- dealkylation, oxidative O- and S-dealkylation, oxidative deamination, as well as other oxidative reactions.
  • Reductive reactions are exemplified without limitation to reactions such as reduction of carbonyl groups, reduction of hydroxyl groups and carbon- carbon double bonds, reduction of nitrogen-containing functions groups, and other reduction reactions.
  • Reactions without change in the oxidation state are exemplified without limitation to reactions such as hydrolysis of esters and ethers, hydrolytic cleavage of carbon-nitrogen single bonds, hydrolytic cleavage of non-aromatic heterocycles, hydration and dehydration at multiple bonds, new atomic linkages resulting from dehydration reactions, hydrolytic dehalogenation, removal of hydrogen halide molecule, and other such reactions.
  • Carrier prodrugs are drug compounds that contain a transport moiety, e.g., that improves uptake and/or localized delivery to a site(s) of action.
  • a transport moiety e.g., that improves uptake and/or localized delivery to a site(s) of action.
  • the linkage between the drug moiety and the transport moiety is a covalent bond
  • the prodrug is inactive or less active than the drag compound
  • the prodrug and any release transport moiety are acceptably non-toxic.
  • the transport moiety is intended to enhance uptake
  • the release of the transport moiety should be rapid.
  • it is desirable to utilize a moiety that provides slow release e.g., certain polymers or other moieties, such as cyclodextrins.
  • Carrier prodrugs are often advantageous for orally administered drugs.
  • Carrier prodrugs can, for example, be used to improve one or more of the following properties: increased lipophilicity, increased duration of pharmacological effects, increased site- specificity, decreased toxicity and adverse reactions, and/or improvement in drug formulation (e.g. stability, water solubility, suppression of an undesirable organoleptic or physiochemical property).
  • lipophilicity can be increased by esterification of hydroxyl groups with lipophilic carboxylic acids, or of carboxylic acid groups with alcohols, e.g., aliphatic alcohols.
  • alcohols e.g., aliphatic alcohols. Wermuth, The Practice of Medicinal Chemistry, Ch. 31- 32, Ed. Wermuth, Academic Press, San Diego, CA, 2001.
  • Prodrugs may proceed from prodrug form to active form in a single step or may have one or more intermediate forms which may themselves have activity or may be inactive.
  • Metabolites e.g., active metabolites
  • prodrugs as described above, e.g., bioprecursor prodrugs.
  • metabolites are pharmacologically active compounds or compounds that further metabolize to pharmacologically active compounds that are derivatives resulting from metabolic process in the body of a subject or patient.
  • active metabolites are such pharmacologically active derivative compounds.
  • prodrugs the prodrug compounds is generally inactive or of lower activity than the metabolic product.
  • the parent compound may be either an active compound or may be an inactive prodrug.
  • Prodrugs and active metabolites may be identified using routine techniques known in the art. See, e.g., Bertolini et al., 1997, J. Med. Chem., 40:2011-2016; Shan et al., 1997, JPharm Sd 86(7):756-757; Bagshawe, 1995, DrugDev. Res., 34:220-230; Wermuth, The Practice of Medicinal Chemistry, Ch. 31-32, Academic Press, San Diego, CA, 2001.
  • Compounds can be formulated as or be in the form of pharmaceutically acceptable salts.
  • Pharmaceutically acceptable salts are non-toxic salts in the amounts and concentrations at which they are administered. The preparation of such salts can facilitate the pharmacological use by altering the physical characteristics of a compound without preventing it from exerting its physiological effect. Useful alterations in physical properties include lowering the melting point to facilitate transmucosal administration and increasing the solubility to facilitate administering higher concentrations of the drug.
  • Pharmaceutically acceptable salts include acid addition salts such as those containing sulfate, chloride, hydrochloride, fumarate, maleate, phosphate, sulfamate, acetate, citrate, lactate, tartrate, methanesulfonate, ethanesulfonate, benzenesulfonate, p- toluenesulfonate, cyclohexylsulfamate and quinate.
  • acid addition salts such as those containing sulfate, chloride, hydrochloride, fumarate, maleate, phosphate, sulfamate, acetate, citrate, lactate, tartrate, methanesulfonate, ethanesulfonate, benzenesulfonate, p- toluenesulfonate, cyclohexylsulfamate and quinate.
  • Pharmaceutically acceptable salts can be obtained from acids such as hydrochloric acid, maleic acid, sulfuric acid, phosphoric acid, sulfamic acid, acetic acid, citric acid, lactic acid, tartaric acid, malonic acid, methanesulfonic acid, ethanesulfonic acid, benzenesulfonic acid, /?-toluenesulfonic acid, cyclohexylsulfamic acid, fumaric acid, and quinic acid.
  • acids such as hydrochloric acid, maleic acid, sulfuric acid, phosphoric acid, sulfamic acid, acetic acid, citric acid, lactic acid, tartaric acid, malonic acid, methanesulfonic acid, ethanesulfonic acid, benzenesulfonic acid, /?-toluenesulfonic acid, cyclohexylsulfamic acid, fumaric acid, and quinic acid.
  • Pharmaceutically acceptable salts also include basic addition salts such as those containing benzathine, chloroprocaine, choline, diethanolamine, ethylenediamine, meglumine, procaine, aluminum, calcium, lithium, magnesium, potassium, sodium, ammonium, alkylamine, and zinc, when acidic functional groups, such as carboxylic acid or phenol are present.
  • basic addition salts such as those containing benzathine, chloroprocaine, choline, diethanolamine, ethylenediamine, meglumine, procaine, aluminum, calcium, lithium, magnesium, potassium, sodium, ammonium, alkylamine, and zinc.
  • acidic functional groups such as carboxylic acid or phenol are present.
  • Such salts can be prepared using the appropriate corresponding bases.
  • salts can be prepared by standard techniques.
  • the free-base form of a compound can be dissolved in a suitable solvent, such as an aqueous or aqueous-alcohol solution containing the appropriate acid and then isolated by evaporating the solution,
  • a salt can be prepared by reacting the free base and acid in an organic solvent.
  • the desired pharmaceutically acceptable salt may be prepared by any suitable method available in the art, for example, treatment of the free base with an inorganic acid, such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like, or with an organic acid, such as acetic acid, maleic acid, succinic acid, mandelic acid, fumaric acid, malonic acid, pyruvic acid, oxalic acid, glycolic acid, salicylic acid, a pyranosidyl acid, such as glucuronic acid or galacturonic acid, an alpha-hydroxy acid, such as citric acid or tartaric acid, an amino acid, such as aspartic acid or glutamic acid, an aromatic acid, such as benzoic acid or cinnamic acid, a sulfonic acid, such as p-toluenesulfonic acid or ethanesulfonic acid
  • an inorganic acid such as hydrochloric acid, hydrobromic
  • the desired pharmaceutically acceptable salt may be prepared by any suitable method, for example, treatment of the free acid with an inorganic or organic base, such as an amine (primary, secondary or tertiary), an alkali metal hydroxide or alkaline earth metal hydroxide, or the like.
  • suitable salts include organic salts derived from amino acids, such as glycine and arginine, ammonia, primary, secondary, and tertiary amines, and cyclic amines, such as piperidine, morpholine and piperazine, and inorganic salts derived from sodium, calcium, potassium, magnesium, manganese, iron, copper, zinc, aluminum and lithium.
  • the pharmaceutically acceptable salt of the different compounds may be present as a complex.
  • complexes include 8-chlorotheophylline complex (analogous to, e.g., dimenhydrinate: diphenhydramine 8-chlorotheophylline (1:1) complex; Dramamme) and various cyclodextrin inclusion complexes.
  • the methods and compounds will typically be used in therapy for human patients. However, they may also be used to treat similar or identical diseases in other vertebrates, e.g., mammals such as other primates, animals of commercial significance, e.g., sports animals, farm animals, e.g., bovines, equines, porcines, and ovines, and pets such as dogs and cats.
  • vertebrates e.g., mammals such as other primates, animals of commercial significance, e.g., sports animals, farm animals, e.g., bovines, equines, porcines, and ovines, and pets such as dogs and cats.
  • Suitable dosage forms depend upon the use or the route of administration, for example, oral, transdermal, transmucosal, inhalant, or by injection (parenteral). Such dosage forms should allow the compound to reach target cells. Other factors are well known in the art, and include considerations such as toxicity and dosage forms that retard the compound or composition from exerting its effects. Techniques and formulations generally may be found in Remington: The Science and Practice of Pharmacy, 21 st edition, Lippincott, Williams and Wilkins, Philadelphia, PA, 2005 (hereby incorporated by reference herein).
  • Carriers or excipients can be used to produce compositions.
  • the carriers or excipients can be chosen to facilitate administration of the compound.
  • Examples of carriers include calcium carbonate, calcium phosphate, various sugars such as lactose, glucose, or sucrose, or types of starch, cellulose derivatives, gelatin, vegetable oils, polyethylene glycols and physiologically compatible solvents.
  • Examples of physiologically compatible solvents include sterile solutions of water for injection (WFI), saline solution, and dextrose.
  • the compounds can be administered by different routes including intravenous, intraperitoneal, subcutaneous, intramuscular, oral, transmucosal, rectal, transdermal, or inhalant.
  • oral administration is preferred.
  • the compounds can be formulated into conventional oral dosage forms such as capsules, tablets, and liquid preparations such as syrups, elixirs, and concentrated drops.
  • compositions for oral use can be obtained, for example, by combining the active compounds with solid excipients, optionally grinding a resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores.
  • suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose (CMC), and/or polyvinylpyrrolidone (PVP: povidone).
  • disintegrating agents may be added, such as the cross-linked polyvinylpyrrolidone, agar, or alginic acid, or a salt thereof such as sodium alginate.
  • Dragee cores are provided with suitable coatings.
  • suitable coatings may optionally contain, for example, gum arabic, talc, polyvinylpyrrolidone, carbopol gel, polyethylene glycol (PEG), and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures.
  • Dye-stuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
  • compositions that can be used orally include push-fit capsules made of gelatin (“gelcaps”), as well as soft, sealed capsules made of gelatin, and a plasticizer, such as glycerol or sorbitol.
  • the push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers.
  • the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols (PEGs).
  • PEGs liquid polyethylene glycols
  • stabilizers may be added.
  • injection parenteral administration
  • the compounds of the invention are formulated in sterile liquid solutions, preferably in physiologically compatible buffers or solutions, such as saline solution, Hank's solution, or Ringer's solution.
  • physiologically compatible buffers or solutions such as saline solution, Hank's solution, or Ringer's solution.
  • the compounds may be formulated in solid form and redissolved or suspended immediately prior to use. Lyophilized forms can also be produced.
  • Administration can also be by transmucosal, topical, transdermal, or inhalant means.
  • penetrants appropriate to the barrier to be permeated are used in the formulation.
  • penetrants are generally known in the art, and include, for example, for transmucosal administration, bile salts and fusidic acid derivatives.
  • detergents may be used to facilitate permeation.
  • Transmucosal administration for example, may be through nasal sprays or suppositories (rectal or vaginal).
  • the topical compositions of this invention are formulated preferably as oils, creams, lotions, ointments, and the like by choice of appropriate carriers known in the art.
  • suitable carriers include vegetable or mineral oils, white petrolatum (white soft paraffin), branched chain fats or oils, animal fats and high molecular weight alcohol (greater than C 12 ).
  • the preferred carriers are those in which the active ingredient is soluble.
  • Emulsifiers, stabilizers, humectants and antioxidants may also be included as well as agents imparting color or fragrance, if desired.
  • Creams for topical application are preferably formulated from a mixture of mineral oil, self-emulsifying beeswax and water in which mixture the active ingredient, dissolved in a small amount solvent (e.g. an oil), is admixed.
  • administration by transdermal means may comprise a transdermal patch or dressing such as a bandage impregnated with an active ingredient and optionally one or more carriers or diluents known in the art.
  • the dosage administration will, of course, be continuous rather than intermittent throughout the dosage regimen.
  • compounds of the invention may be formulated as dry powder or a suitable solution, suspension, or aerosol.
  • Powders and solutions may be formulated with suitable additives known in the art.
  • powders may include a suitable powder base such as lacatose or starch, and solutions may comprise propylene glycol, sterile water, ethanol, sodium chloride and other additives, such as acid, alkali and buffer salts.
  • Such solutions or suspensions may be administered by inhaling via spray, pump, atomizer, or nebulizer, and the like.
  • the compounds of the invention may also be used in combination with other inhaled therapies, for example corticosteroids such as fluticasone proprionate, beclomethasone dipropionate, triamcinolone acetonide, budesonide, and mometasone furoate; beta agonists such as albuterol, salmeterol, and formoterol; anticholinergic agents such as ipratroprium bromide or tiotropium; vasodilators such as treprostinal and iloprost; enzymes such as DNAase; therapeutic proteins; immunoglobulin antibodies; an oligonucleotide, such as single or double stranded DNA or RNA, siRNA; antibiotics such as tobramycin; muscarinic receptor antagonists; leukotriene antagonists; cytokine antagonists; protease inhibitors; cromolyn sodium; nedocril sodium; and sodium cromoglycate.
  • corticosteroids such as
  • the compounds of the invention may also be used in combination with other therapies for treating the same disease.
  • Such combination use includes administration of the compounds and one or more other therapeutics at different times, or co-administration of the compound and one or more other therapies.
  • dosage may be modified for one or more of the compounds of the invention or other therapeutics used in combination, e.g., reduction in the amount dosed relative to a compound or therapy used alone, by methods well known to those of ordinary skill in the art.
  • use in combination includes use with other therapies, drugs, medical procedures etc., where the other therapy or procedure may be administered at different times (e.g. within a short time, such as within hours (e.g. 1, 2, 3, 4-24 hours), or within a longer time (e.g. 1-2 days, 2-4 days, 4-7 days, 1-4 weeks)) than a compound of the present invention, or at the same time as a compound of the invention.
  • Use in combination also includes use with a therapy or medical procedure that is administered once or infrequently, such as surgery, along with a compound of the invention administered within a short time or longer time before or after the other therapy or procedure.
  • the present invention provides for delivery of compounds of the invention and one or more other drug therapeutics delivered by a different route of administration or by the same route of administration.
  • the use in combination for any route of administration includes delivery of compounds of the invention and one or more other drug therapeutics delivered by the same route of administration together in any formulation, including formulations where the two compounds are chemically linked in such a way that they maintain their therapeutic activity when administered.
  • the other drug therapy may be co-administered with one or more compounds of the invention.
  • Use in combination by co-administration includes administration of co-formulations or formulations of chemically joined compounds, or administration of two or more compounds in separate formulations within a short time of each other (e.g.
  • Co-administration of separate formulations includes co-administration by delivery via one device, for example the same inhalant device, the same syringe, etc., or administration from separate devices within a short time of each other.
  • Co-formulations of compounds of the invention and one or more additional drug therapies delivered by the same route includes preparation of the materials together such that they can be administered by one device, including the separate compounds combined in one formulation, or compounds that are modified such that they are chemically joined, yet still maintain their biological activity.
  • Such chemically joined compounds may have a linkage that is substantially maintained in vivo, or the linkage may break down in vivo, separating the two active components.
  • a dose will be between about 0.01 and 50 mg/kg, preferably 0.1 and 20 mg/kg of the subject being treated. Multiple doses may be used.
  • nucleic acids such as, e.g., subcloning, labeling probes (e.g. random-primer labeling using Klenow polymerase, nick translation, amplification), sequencing, hybridization and the like are well disclosed in the scientific and patent literature, see, e.g., Sambrook, ed., Molecular Cloning: a Laboratory Manual (2nd ed.), VoIs. 1-3, Cold Spring Harbor Laboratory, (1989); Current Protocols in Molecular Biology, Ausubel, ed. John Wiley & Sons, Inc., New York (1997); Laboratory Techniques in Biochemistry and Molecular Biology: Hybridization With Nucleic Acid Probes, Part I. Theory and Nucleic Acid Preparation, Tijssen, ed. Elsevier, N.Y. (1993).
  • labeling probes e.g. random-primer labeling using Klenow polymerase, nick translation, amplification
  • sequencing hybridization and the like
  • Nucleic acid sequences can be amplified as necessary for further use using amplification methods, such as PCR, isothermal methods, rolling circle methods, etc., are well known to the skilled artisan. See, e.g., Saiki, "Amplification of Genomic DNA” in PCR Protocols, Innis et al., Eds., Academic Press, San Diego, CA 1990, pp 13-20; Wharam et al., Nucleic Acids Res.
  • Nucleic acids, vectors, capsids, polypeptides, and the like can be analyzed and quantified by any of a number of general means well known to those of skill in the art. These include, e.g., analytical biochemical methods such as NMR 5 spectrophotometry, radiography, electrophoresis, capillary electrophoresis, high performance liquid chromatography (HPLC), thin layer chromatography (TLC), and hyperdiffusion chromatography, various immunological methods, e.g.
  • Obtaining and manipulating nucleic acids used to practice the methods of the invention can be performed by cloning from genomic samples, and, if desired, screening and re-cloning inserts isolated or amplified from, e.g., genomic clones or cDNA clones.
  • Sources of nucleic acid used in the methods of the invention include genomic or cDNA libraries contained in, e.g., mammalian artificial chromosomes (MACs), see, e.g., U.S. Patent Nos. 5,721,118; 6,025,155; human artificial chromosomes, see, e.g., Rosenfeld (1997) Nat. Genet.
  • MACs mammalian artificial chromosomes
  • yeast artificial chromosomes YAC
  • bacterial artificial chromosomes BAC
  • Pl artificial chromosomes see, e.g., Woon (1998) Genomics 50:306-316
  • Pl-derived vectors see, e.g., Kern (1997) Biotechniques 23:120-124; cosmids, recombinant viruses, phages or plasmids.
  • the nucleic acids of the invention can be operatively linked to a promoter.
  • a promoter can be one motif or an array of nucleic acid control sequences which direct transcription of a nucleic acid.
  • a promoter can include necessary nucleic acid sequences near the start site of transcription, such as, in the case of a polymerase II type promoter, a TATA element.
  • a promoter also optionally includes distal enhancer or repressor elements which can be located as much as several thousand base pairs from the start site of transcription.
  • a "constitutive" promoter is a promoter which is active under most environmental and developmental conditions.
  • An “inducible” promoter is a promoter which is under environmental or developmental regulation.
  • tissue specific promoter is active in certain tissue types of an organism, but not in other tissue types from the same organism.
  • operably linked refers to a functional linkage between a nucleic acid expression control sequence (such as a promoter, or array of transcription factor binding sites) and a second nucleic acid sequence, wherein the expression control sequence directs transcription of the nucleic acid corresponding to the second sequence.
  • the nucleic acids of the invention can also be provided in expression vectors and cloning vehicles, e.g., sequences encoding the polypeptides of the invention.
  • Expression vectors and cloning vehicles of the invention can comprise viral particles, baculovirus, phage, plasmids, phagemids, cosmids, fosmids, bacterial artificial chromosomes, viral DNA (e.g. vaccinia, adenovirus, foul pox virus, pseudorabies and derivatives of SV40), Pl -based artificial chromosomes, yeast plasmids, yeast artificial chromosomes, and any other vectors specific for specific hosts of interest (such as bacillus, Aspergillus and yeast).
  • Vectors of the invention can include chromosomal, non-chromosomal and synthetic DNA sequences. Large numbers of suitable vectors are known to those of skill in the art, and are commercially available.
  • nucleic acids of the invention can be cloned, if desired, into any of a variety of vectors using routine molecular biological methods; methods for cloning in vitro amplified nucleic acids are disclosed, e.g., U.S. Pat. No. 5,426,039.
  • restriction enzyme sites can be "built into” a PCR primer pair.
  • Vectors may be introduced into a genome or into the cytoplasm or a nucleus of a cell and expressed by a variety of conventional techniques, well described in the scientific and patent literature. See, e.g., Roberts (1987) Nature 328:731; Schneider (1995) Protein Expr. Purif.
  • the vectors can be isolated from natural sources, obtained from such sources as ATCC or GenBank libraries, or prepared by synthetic or recombinant methods.
  • the nucleic acids of the invention can be expressed in expression cassettes, vectors or viruses which are stably or transiently expressed in cells (e.g. episomal expression systems).
  • Selection markers can be incorporated into expression cassettes and vectors to confer a selectable phenotype on transformed cells and sequences. For example, selection markers can code for episomal maintenance and replication such that integration into the host genome is not required.
  • the nucleic acids of the invention are administered in vivo for in situ expression of the peptides or polypeptides of the invention.
  • the nucleic acids can be administered as "naked DNA” (see, e.g., U.S. Patent No. 5,580,859) or in the form of an expression vector, e.g., a recombinant virus.
  • the nucleic acids can be administered by any route, including peri- or intra-tumorally, as described below.
  • Vectors administered in vivo can be derived from viral genomes, including recombinant ⁇ modified enveloped or non-enveloped DNA and RNA viruses, preferably selected from baculoviridiae, parvoviridiae, picornoviridiae, herpesveridiae, poxviridae, adenoviridiae, or picornnaviridiae. Chimeric vectors may also be employed which exploit advantageous merits of each of the parent vector properties (See e.g., Feng (1997) Nature Biotechnology 15:866-870). Such viral genomes may be modified by recombinant DNA techniques to include the nucleic acids of the invention; and may be further engineered to be replication deficient, conditionally replicating or replication competent.
  • vectors are derived from the adenoviral (e.g. replication incompetent vectors derived from the human adenovirus genome, see, e.g., U.S. Patent Nos. 6,096,718; 6,110,458; 6,113,913; 5,631,236); adeno-associated viral and retroviral genomes.
  • Retroviral vectors can include those based upon murine leukemia virus (MuLV), gibbon ape leukemia virus (GaLV), Simian mimuno deficiency virus (SIV), human immuno deficiency virus (HIV), and combinations thereof; see, e.g., U.S. Patent Nos.
  • Adeno-associated virus (AAV)-based vectors can be used to transduce cells with target nucleic acids, e.g., in the in vitro production of nucleic acids and peptides, and in in vivo and ex vivo gene therapy procedures; see, e.g., U.S. Patent Nos. 6,110,456; 5,474,935; Okada (1996) Gene Ther. 3:957-964.
  • the present invention also relates to fusion proteins, and nucleic acids encoding them.
  • a polypeptide of the invention can be fused to a heterologous peptide or polypeptide, such as N-terminal identification peptides which impart desired characteristics, such as increased stability or simplified purification.
  • Peptides and polypeptides of the invention can also be synthesized and expressed as fusion proteins with one or more additional domains linked thereto for, e.g., producing a more immunogenic peptide, to more readily isolate a recombinantly synthesized peptide, to identify and isolate antibodies and antibody-expressing B cells, and the like.
  • Detection and purification facilitating domains include, e.g., metal chelating peptides such as polyhistidine tracts and histidine-tryptophan modules that allow purification on immobilized metals, protein A domains that allow purification on immobilized immunoglobulin, and the domain utilized in the FLAGS extension/affinity purification system (Immunex Corp, Seattle WA).
  • metal chelating peptides such as polyhistidine tracts and histidine-tryptophan modules that allow purification on immobilized metals
  • protein A domains that allow purification on immobilized immunoglobulin
  • the domain utilized in the FLAGS extension/affinity purification system Immunex Corp, Seattle WA.
  • the inclusion of a cleavable linker sequences such as Factor Xa or enterokinase (Invitrogen, San Diego CA) between a purification domain and the motif-comprising peptide or polypeptide to facilitate purification.
  • an expression vector can include an epitope-encoding nucleic acid sequence linked to six histidine residues followed by a thioredoxin and an enterokinase cleavage site (see e.g., Williams (1995) Biochemistry 34:1787-1797; Dobeli (1998) Protein Expr. Purif. 12:404- 414).
  • the histidine residues facilitate detection and purification while the enterokinase cleavage site provides a means for purifying the epitope from the remainder of the fusion protein.
  • a nucleic acid encoding a polypeptide of the invention is assembled in appropriate phase with a leader sequence capable of directing secretion of the translated polypeptide or fragment thereof.
  • the nucleic acids and polypeptides of the invention can be bound to a solid support, e.g., for use in screening and diagnostic methods.
  • Solid supports can include, e.g., membranes (e.g. nitrocellulose or nylon), a microtiter dish (e.g. PVC, polypropylene, or polystyrene), a test tube (glass or plastic), a dip stick (e.g. glass, PVC, polypropylene, polystyrene, latex and the like), a microfuge tube, or a glass, silica, plastic, metallic or polymer bead or other substrate such as paper.
  • a metal e.g. cobalt or nickel
  • Adhesion of molecules to a solid support can be direct (i.e., the molecule contacts the solid support) or indirect (a "linker” is bound to the support and the molecule of interest binds to this linker).
  • Molecules can be immobilized either covalently (e.g. utilizing single reactive thiol groups of cysteine residues (see, e.g., Colliuod (1993) Bioconjugate Chem. 4:528-536) or non-covalently but specifically (e.g. via immobilized antibodies (see, e.g., Schuhmann (1991) Adv. Mater. 3:388-391; Lu (1995) Anal. Chem.
  • the reactive ends can be any of a variety of functionalities including, but not limited to: amino reacting ends such as N-hydroxysuccinimide (NHS) active esters, imidoesters, aldehydes, epoxides, sulfonyl halides, isocyanate, isothiocyanate, and nitroaryl halides; and thiol reacting ends such as pyridyl disulfides, maleimides, thiophthalimides, and active halogens.
  • amino reacting ends such as N-hydroxysuccinimide (NHS) active esters, imidoesters, aldehydes, epoxides, sulfonyl halides, isocyanate, isothiocyanate, and nitroaryl halides
  • thiol reacting ends such as pyridyl disulfides, maleimides, thiophthalimides, and active halogens.
  • heterobifunctional crosslinking reagents have two different reactive ends, e.g., an amino-reactive end and a thiol-reactive end, while homobifunctional reagents have two similar reactive ends, e.g., bismaleimidohexane (BMH) which permits the cross-linking of sulfhydryl-containing compounds.
  • BMH bismaleimidohexane
  • the spacer can be of varying length and be aliphatic or aromatic.
  • Examples of commercially available homobifunctional cross-linking reagents include, but are not limited to, the imidoesters such as dimethyl adipimidate dihydrochloride (DMA); dimethyl pimelimidate dihydrochloride (DMP); and dimethyl suberimidate dihydrochloride (DMS).
  • DMA dimethyl adipimidate dihydrochloride
  • DMP dimethyl pimelimidate dihydrochloride
  • DMS dimethyl suberimidate dihydrochloride
  • Heterobifunctional reagents include commercially available active halogen-NHS active esters coupling agents such as N-succinimidyl bromoacetate and N-succinimidyl (4-iodoacetyl)aminobenzoate (SIAB) and the sulfosuccinimidyl derivatives such as sulfosuccinimidyl(4-iodoacetyl)aminobenzoate (sulfo-SIAB) (Pierce).
  • active halogen-NHS active esters coupling agents such as N-succinimidyl bromoacetate and N-succinimidyl (4-iodoacetyl)aminobenzoate (SIAB) and the sulfosuccinimidyl derivatives such as sulfosuccinimidyl(4-iodoacetyl)aminobenzoate (sulfo-SIAB) (
  • Another group of coupling agents is the heterobifunctional and thiol cleavable agents such as N-succinimidyl 3-(2-pyridyidithio)propionate (SPDP) (Pierce Chemicals, Rockford, IL).
  • SPDP N-succinimidyl 3-(2-pyridyidithio)propionate
  • Antibodies can also be used for binding paolypeptides and peptides of the invention to a solid support. This can be done directly by binding peptide-specif ⁇ c antibodies to the column or it can be done by creating fusion protein chimeras comprising motif-containing peptides linked to, e.g., a known epitope (e.g. a tag (e.g. FLAG, myc) or an appropriate immunoglobulin constant domain sequence (an "immunoadhesin,” see, e.g., Capon (1989) Nature 377:525-531 (1989).
  • a known epitope e.g. a tag (e.g. FLAG, myc)
  • an appropriate immunoglobulin constant domain sequence an "immunoadhesin," see, e.g., Capon (1989) Nature 377:525-531 (1989).
  • Nucleic acids or polypeptides of the invention can be immobilized to or applied to an array.
  • Arrays can be used to screen for or monitor libraries of compositions (e.g. small molecules, antibodies, nucleic acids, etc.) for their ability to bind to or modulate the activity of a nucleic acid or a polypeptide of the invention.
  • a monitored parameter is transcript expression of a gene comprising a nucleic acid of the invention.
  • One or more, or, all the transcripts of a cell can be measured by hybridization of a sample comprising transcripts of the cell, or, nucleic acids representative of or complementary to transcripts of a cell, by hybridization to immobilized nucleic acids on an array, or "biochip.”
  • arrays comprising genomic nucleic acid can also be used to determine the genotype of a newly engineered strain made by the methods of the invention.
  • Polypeptide arrays can also be used to simultaneously quantify a plurality of proteins.
  • array or “microarray” or “biochip” or “chip” as used herein is a plurality of target elements, each target element comprising a defined amount of one or more polypeptides (including antibodies) or nucleic acids immobilized onto a defined area of a substrate surface.
  • any known array and/or method of making and using arrays can be incorporated in whole or in part, or variations thereof, as disclosed, for example, in U.S. Patent Nos.
  • the invention also provides a transformed cell comprising a nucleic acid sequence of the invention, e.g., a sequence encoding a polypeptide of the invention, or a vector of the invention.
  • the host cell maybe any of the host cells familiar to those skilled in the art, including prokaryotic cells, eukaryotic cells, such as bacterial cells, fungal cells, yeast cells, mammalian cells, insect cells, or plant cells.
  • Exemplary bacterial cells include E. coli, Streptomyces, Bacillus subtilis, Salmonella typhimurium and various species within the genera Pseudomonas, Streptomyces, and Staphylococcus.
  • Exemplary insect cells include Drosophila S2 and Spodoptera Sf9.
  • Exemplary animal cells include CHO, COS or Bowes melanoma or any mouse or human cell line. The selection of an appropriate host is within the abilities of those skilled in the art.
  • Vectors may be introduced into the host cells using any of a variety of techniques, including transformation, transfection, transduction, viral infection, gene guns, or Ti-mediated gene transfer. Particular methods include calcium phosphate transfection, DEAE-Dextran mediated transfection, lipofection, or electroporation.
  • Engineered host cells can be cultured in conventional nutrient media modified as appropriate for activating promoters, selecting transformants or amplifying the genes of the invention. Following transformation of a suitable host strain and growth of the host strain to an appropriate cell density, the selected promoter may be induced by appropriate means (e.g. temperature shift or chemical induction) and the cells may be cultured for an additional period to allow them to produce the desired polypeptide or fragment thereof.
  • appropriate means e.g. temperature shift or chemical induction
  • Cells can be harvested by centrifugation, disrupted by physical or chemical means, and the resulting crude extract is retained for further purification.
  • Microbial cells employed for expression of proteins can be disrupted by any convenient method, including freeze-thaw cycling, sonication, mechanical disruption, or use of cell lysing agents. Such methods are well known to those skilled in the art.
  • the expressed polypeptide or fragment can be recovered and purified from recombinant cell cultures by methods including ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxylapatite chromatography and lectin chromatography. Protein refolding steps can be used, as necessary, in completing configuration of the polypeptide. If desired, high performance liquid chromatography (HPLC) can be employed for final purification steps.
  • HPLC high performance liquid chromatography
  • mammalian cell culture systems can also be employed to express recombinant protein.
  • mammalian expression systems include the COS-7 lines of monkey kidney fibroblasts and other cell lines capable of expressing proteins from a compatible vector, such as the C127, 3T3, CHO, HeLa and BHK cell lines.
  • the constructs in host cells can be used in a conventional manner to produce the gene product encoded by the recombinant sequence.
  • the polypeptides produced by host cells containing the vector may be glycosylated or may be non-glycosylated.
  • Polypeptides of the invention may or may not also include an initial methionine amino acid residue.
  • Cell-free translation systems can also be employed to produce a polypeptide of the invention.
  • Cell-free translation systems can use mRNAs transcribed from a DNA construct comprising a promoter operably linked to a nucleic acid encoding the polypeptide or fragment thereof.
  • the DNA construct may be linearized prior to conducting an in vitro transcription reaction.
  • the transcribed mRNA is then incubated with an appropriate cell-free translation extract, such as a rabbit reticulocyte extract, to produce the desired polypeptide or fragment thereof.
  • the expression vectors can contain one or more selectable marker genes to provide a phenotypic trait for selection of transformed host cells such as dihydrofolate reductase or neomycin resistance for eukaryotic cell culture, or such as tetracycline or ampicillin resistance in E. coli.
  • cDNA encoding a polypeptide of interest may be incorporated into a mammalian expression vector, e.g. pcDNAl, which is available commercially from Invitrogen Corporation (San Diego, Calif., U.S.A.; catalogue number V490-20).
  • pcDNAl a mammalian expression vector
  • a polylinker is located appropriately downstream of the CMV promoter (and 3' of the T7 promoter).
  • the cDNA insert may be first released from the above phagemid incorporated at appropriate restriction sites in the pcDNAl polylinker. Sequencing across the junctions may be performed to confirm proper insert orientation in pcDNAl. The resulting plasmid may then be introduced for transient expression into a selected mammalian cell host, for example, the monkey-derived, fibroblast like cells of the COS-I lineage (available from the American Type Culture Collection, Rockville, Md. as ATCC CRL 1650).
  • COS-I cells may be transfected with approximately 8 ⁇ g DNA per 10 6 COS cells, by DEAE-mediated DNA transfection and treated with chloroquine according to the procedures described by Sambrook et al, Molecular Cloning: A Laboratory Manual, 1989, Cold Spring Harbor Laboratory Press, Cold Spring Harbor N.Y, pp. 16.30-16.37.
  • An exemplary method is as follows. Briefly, COS-I cells are plated at a density of 5 x 10 6 cells/dish and then grown for 24 hours in FBS-supplemented DMEM/F12 medium. Medium is then removed and cells are washed in PBS and then in medium.
  • a transfection solution containing DEAE dextran (0.4 mg/ml), 100 ⁇ M chloroquine, 10% NuSerum, DNA (0.4 mg/ml) in DMEM/F12 medium is then applied on the cells 10 ml volume. After incubation for 3 hours at 37 0 C, cells are washed in PBS and medium as just described and then shocked for 1 minute with 10% DMSO in DMEM/F12 medium. Cells are allowed to grow for 2-3 days in 10% FBS-supplemented medium, and at the end of incubation dishes are placed on ice, washed with ice cold PBS and then removed by scraping.
  • Cells are then harvested by centrifugation at 1000 rpm for 10 minutes and the cellular pellet is frozen in liquid nitrogen, for subsequent use in protein expression.
  • Northern blot analysis of a thawed aliquot of frozen cells may be used to confirm expression of receptor-encoding cDNA in cells under storage.
  • stably transfected cell lines can also prepared, for example, using two different cell types as host: CHO Kl and CHO Pro5.
  • cDNA coding for the relevant protein may be incorporated into the mammalian expression vector pRC/CMV (Invitrogen), which enables stable expression. Insertion at this site places the cDNA under the expression control of the cytomegalovirus promoter and upstream of the polyadenylation site and terminator of the bovine growth hormone gene, and into a vector background comprising the neomycin resistance gene (driven by the S V40 early promoter) as selectable marker.
  • An exemplary protocol to introduce plasmids constructed as described above is as follows.
  • the host CHO cells are first seeded at a density of 5x10 in 10% FBS- supplemented MEM medium. After growth for 24 hours, fresh medium is added to the plates and three hours later, the cells are transfected using the calcium phosphate-DNA co- precipitation procedure (Sambrook et al, supra). Briefly, 3 ⁇ g of DNA is mixed and incubated with buffered calcium solution for 10 minutes at room temperature. An equal volume of buffered phosphate solution is added and the suspension is incubated for 15 minutes at room temperature. Next, the incubated suspension is applied to the cells for 4 hours, removed and cells were shocked with medium containing 15% glycerol.
  • Example 1 Synthesis of compound of Formula I, where Xi, X 2 , Yi and Y 2 are CH and L 1 is -CH 2 -:
  • Step - 1- Synthesis of compound 2.
  • Compound 2 is synthesized from commercially available 7-azaindole following the literature procedure (Robinson, J Am. Chem. Soc, 1955, 77, p. 457).
  • Compound of Formula II is synthesized by deprotonation using base (e.g. BuLi, NaH) in aprotic solvent like tetrahydrofuran or ether and reacting the anion with a silyl chloride (e.g. TIPS) or an anhydride (e.g. Boc anhydride).
  • base e.g. BuLi, NaH
  • aprotic solvent like tetrahydrofuran or ether
  • an anhydride e.g. Boc anhydride
  • the reaction is worked up in the usual manner and purified by flash silica gel chromatography to give the nitrogen-protected compound.
  • the final compound can be realized through the deprotection of the protecting group (Boc, TIPS) using standard conditions (TFA or NH 4 F) at room temperature.
  • Compound of Formula III, where P is a protecting group is synthesized by reacting compound 3 with an appropriate reagent to introduce a protecting group (e.g. tert- butyloxycarbonyl di anhydride) and a base (e.g. sodium hydride) in an appropriate solvent (e.g. tetrahydrofuran) typically at room temperature for 12-18 hours.
  • a protecting group e.g. tert- butyloxycarbonyl di anhydride
  • a base e.g. sodium hydride
  • an appropriate solvent e.g. tetrahydrofuran
  • Compound of Formula IV wherein R 24 is Ar 1 , is synthesized by reacting compound of Formula III in an appropriate solvent (e.g. 1,2-dimethoxyethane) with a Grignard reagent of the formula R 24 MgCl or R 24 MgBr (e.g. pyridinyl magnesium bromide) or an equivalent nucleophile in an appropriate solvent (e.g. tetrahydrofuran) under inert atmosphere cooled typically to —10 0 C. The reaction is typically allowed to warm to room temperature and stirred for 12-18 hours. The desired compound is purified by reverse phase high pressure liquid chromatography.
  • an appropriate solvent e.g. 1,2-dimethoxyethane
  • a Grignard reagent of the formula R 24 MgCl or R 24 MgBr e.g. pyridinyl magnesium bromide
  • an appropriate solvent e.g. tetrahydrofuran
  • Steps - 4 and 5 - Synthesis of an intermediate of compound of Formula I An intermediate of compound of Formula I is synthesized by reacting compound of Formula IV with a reducing agent (e.g. sodium borohydride) in a polar solvent (e.g. ethanol) typically with heating to 80 0 C for 1-4 hours. The reaction is quenched with the addition of methanol and concentrated and purified by reverse phase high performance liquid chromatography. Compound of Formula I where R 24 is Ar 1 is synthesized by reacting this intermediate with an appropriate reagent to remove the protecting group, P, (e.g. hydrochloric acid) in an apolar solvent (e.g. dioxane). The final compound is isolated by standard procedures (e.g. reverse phase preparative high pressure liquid chromatography).
  • a reducing agent e.g. sodium borohydride
  • a polar solvent e.g. ethanol
  • P e.g. hydrochloric acid
  • apolar solvent e.g. dioxan
  • Compound of Formula I' where R 24 is Ar 1 is synthesized by reacting compound 1 with an activating agent (e.g. methyl magnesium bromide and zinc dichloride or anhydrous aluminum chloride) and a heteroaryl acid chloride (e.g. nicotinic acid chloride) in a non-reactive solvent (e.g. dichloromethane), under inert atmosphere (e.g. argon), at room temperature or with heating up to reflux for 18-24 hours.
  • an activating agent e.g. methyl magnesium bromide and zinc dichloride or anhydrous aluminum chloride
  • a heteroaryl acid chloride e.g. nicotinic acid chloride
  • a non-reactive solvent e.g. dichloromethane
  • inert atmosphere e.g. argon
  • Example 2 Synthesis of intermediate 3-(6 ⁇ Chloro-pyridin-3-ylmethyl)-l- triisopropylsilanyl-lH-pyrrolo[2,3-b]pyridine (6) and (3-(6-Bromo-pyridin-3- ylmethyl)-l-tr ⁇ sopropylsilanyl-lH-pyrrolo[2,3-b]pyridine) (6a)
  • Compound 6 an intermediate to compounds of Formula I where X 1 , X 2 , Y 1 and Y 2 are CH, n is 1, P, Q and T are CH and L 1 is -CH 2 -, may be synthesized in four steps from 7-azaindole according to the following Scheme 4.
  • Step -1 Synthesis ofdimethyl-(lH-pyrrolo[2,3-b]pyridin-3-ylmethyl)-amine (2)
  • Isopropyl alcohol 320.0 mL
  • lH-pyrrolo[2,3-b]pyridine 1 7.10 g, 60.1 rnmol
  • dimethylamine hydrochloride 5.4 g, 0.066 mol
  • formaldehyde 2.0 g, 0.066 mol
  • Step -2 Synthesis ofdimethyl-(l-triisopropylsilanyl-lH-pyrrolo[2,3-b]pyridin-3- ylmethyl)-amine (4)
  • Step - 3- Synthesis of 3-chloromethyl- 1-triisopropylsilanyl-lH-pyrrolo [2, 3-b] pyridine (5)
  • compound 4 500.0 mg, 1.51 mmol
  • toluene 5.0 mL, 0.047 mol
  • Into the reaction mixture 1.0 M isopropyl chloroformate in toluene (1.6 mL) was added slowly at room temperature. The reaction mixture was stirred for another 2 hours to give desired compound 5 used for next step without purification.
  • Compound 7, an intermediate to compounds of Formula I where X 1 , X 2 , Y 1 and Y 2 are CH, n is 1, P, Q and T are CH and L 1 is -CO-, may be synthesized in one step from 7-azaindole according to the following Scheme 5.
  • Step -1 Synthesis ofbenzyl-[5-(l-triisopropylsilanyl-lH-pyrrolo[2,3-b]pyridin-3- ylmethyl)-pyridin-2-yl] -amine (10):
  • Step -2 Synthesis of benzyl-[5-(lH-pyrrolo[2,3-b]pyridin-3-ylmethyl)-pyridin-2-yl] -amine (P-OOOl):
  • Step -1 Synthesis ofIsobutyl-[5-(lH ⁇ pyrrolo[2, 3-b]pyridin-3-ylmethyl)-pyridin-2-yl]- amine (P-0028).
  • Step -1 Synthesis of(lH-Pyrrolo[2, 3-b]pyridin-3-yl)-[6-(4-trifluoromethyl-benzylamino)- pyridin-3-yl] -methanone (P-0017)
  • Example 9 Synthesis of compounds of Formula I where n is 1, P, Q and T are CH X 1 , X 2 and Y 2 are CH, Yi is CR 4 , L 1 is -CH2-, L 2 is -NHCH 2 -, and R 1 is 4 substituted phenyl (Formula Ic). [0295] Compounds of Formula Ic, where R 4 is as defined for Formula I and Z is a substituent as defined for optionally substituted aryl, can be synthesized in five Steps from 2-amino-5-bromopyridines as shown in the following general Scheme 11.
  • Compound of Formula V is dissolved in a non-reactive solvent (e.g. tetrahydrofuran) and typically cooled at -78 0 C under an inert atmosphere.
  • a non-reactive solvent e.g. tetrahydrofuran
  • an organo lithium reagent e.g. methyl lithium
  • the reaction mixture is typically stirred at -78 0 C for several hours.
  • an organo lithium reagent e.g. tert-buiyl lithium
  • the reaction mixture is maintained at -78 0 C, and an appropriate formylating reagent (e.g. 1-piperidine carboxaldehyde) is added.
  • an appropriate formylating reagent e.g. 1-piperidine carboxaldehyde
  • the reaction is allowed to stir at -78 0 C for an additional several hours and slowly warmed to room temperature. Isolation by conventional means (e.g. extraction) affords compounds of Formula VI.
  • compounds of Formula VIII and IX is combined and dissolved in an appropriate polar aprotic solvent (e.g. acetonitrile).
  • an appropriate polar aprotic solvent e.g. acetonitrile
  • Reagents appropriate to effect the reduction e.g. triethylsilane and trifluoroacetic acid
  • the reactions are stirred at room temperature for several days. Isolation by conventional means (e.g. extraction, silica gel chromatography) affords compounds of Formula Ic.
  • Step 1 Preparation of(5-Bromo-pyridin-2-yl)-(4-trifluoromethyl-benzyl)-amine (17) [0302] Into a round bottom flask fitted with stirrer and reflux condenser was added 2- amino-5-brornopyridine (15, 1.73 mol, 300 g) and/?-trifluoromethylbenzaldehyde (16, 1.723 mol, 300 g) to a solution of trifluoroacetic acid (400 mL), triethylsilane (825 mL) and acetonitrile (7500 mL). The reaction was heated to reflux overnight (24 hours).
  • Step 2 Preparation of6-(4-Trifluoromethyl-benzylamino)-pyridine-3-carbaldehyde (18) [0303] Into a 5 L round bottom flask was added compound 17 (0.6 mol, 198.6 g,) and tetrahydrofuran (2.5 L) under an atmosphere of argon at -78 0 C. Into the reaction mixture was added 1.7 M tert-butyllithium in pentane (800 mL) over 60 mins. Two hours after the addition of tert-butyllithium, N,N-dimethylformamide (100 mL) was added.
  • reaction mixture was stirred at -78 0 C for 2 hours, then allowed to stand at room temperature for another 1 hour.
  • the reaction mixture was poured into saturated ammonium chloride solution and extracted with ethyl acetate. The organic layer was washed with brine, dried over sodium sulfate, concentrated and triturated with hexane/isopropyl ether (1:1) to give aldehyde compound 18.
  • Step 3 Preparation of(5-Formyl-pyridin-2-yl)-(4-trifluoromethyl-benzyl)-carbamic acid tert-butyl ester (19)
  • Steps 4 and 5 Preparation of[5-(4-Methoxy-lH-pyrrolo[2,3-b]pyridin-3-ylmethyl)- pyridin-2-yl]-(4-trifluoromethyl-benzyl)-amine (P-OOIl)
  • Step 4 Into a solution of methanol (20 mL, 0.5 mol) was added sodium hydroxide (0.62 g, 0.016 mol), followed by 4-methoxy-7-azaindole (20, 600 mg, 4 mmol, prepared as described in Example 12). Once the mixture was homogeneous, compound 19 (1.7 g, 4.46 mmol) was added and the mixture was stirred at room temperature for 48 hours. The solvent was evaporated and dilute HCl was added to the residue. The residue was extracted with ethyl acetate and washed with 10% sodium bicarbonate, followed by brine. The organic layer was dried over MgSO 4 , filtered and evaporated to give a mixture of crude compounds 21 and 22, which was used in the next step.
  • lH-Pyrrolo[2,3-b]pyridine 7-oxide 23 was synthesized by reacting commercially available 7-azaindole 1 with an oxidizing agent (e.g. m-CPBA) in a non-reactive solvent (e.g. dimethoxyethane) as described by Schneller, S. W.; Luo, Jiann-Kuan. J. Org. Chem. 1980, 45:4045-4048. The compound was isolated by filtration of the resulting solid that forms upon standing at 5 0 C for typically 1-3 h.
  • an oxidizing agent e.g. m-CPBA
  • a non-reactive solvent e.g. dimethoxyethane
  • 4-chloro-7-azaindole 24 was synthesized by reacting lH-Pyrrolo[2,3-b]pyridine 7-oxide 23 with a chlorinating agent (e.g. POCl 3 ) neat as described by Schneller, S. W.; Luo, Jiann-Kuan. J. Org. Chem. 1980, 45:4045-4048. The resulting solution after heating for 3-5 h at elevated temperatures (100-150 0 C) was neutralized with abase (e.g. NH 4 OH) until a solid precipitated. The solid was isolated by filtration.
  • a chlorinating agent e.g. POCl 3
  • abase e.g. NH 4 OH
  • 4-methoxy-7-azaindole 20 was prepared by reacting 4-chloro-7-azaindole 24 (prepared as described in Example 9) with sodium hydroxide in methanol as described by Girgis, N. etal., J. Heterocyclic. Chem. 1989, 26:317-325.
  • Example 13 Synthesis of compounds of Formula I where n is 1, P is CR 30 , Q, T, Xi, X 2 , Yi and Y 2 are CH, L 1 is -CH2-, L 2 is -NHCH 2 -, and R 1 is substituted phenyl (Formula Id).
  • 2-halopyridine X e.g. 2-chloro-6- methoxypyridine
  • Y is a halogen, preferably chlorine or bromine
  • a non-reactive solvent e.g. tetrahydrofuran
  • organolithium reagent e.g. tert-butyllithium
  • an appropriately substituted benzylamine XV e.g. 4-(trifluoromethyl)benzylamine
  • a base e.g. sodium tert-butoxide
  • a catalyst e.g. tris(dibenzylideneacetone)dipalladium(0)
  • ligand e.g. 2,2'-Bis(diphenylphosphino)- l,l'-binaphthyl
  • a non-reactive solvent e.g. toluene
  • the reaction is heated (e.g. 80 °C) for several hours. Isolation by conventional means (e.g. extraction and silica gel chromatography) affords compounds of Formula XVI.
  • Compound of Formula XIX is dissolved in a non-reactive solvent (e.g. tetrahydrofuran) and typically cooled at -78 0 C under an inert atmosphere.
  • a non-reactive solvent e.g. tetrahydrofuran
  • an organolithium reagent e.g. methyllithium
  • the reaction mixture is typically stirred at -78 0 C for several hours.
  • an organolithium reagent e.g. tert-butyllithium
  • an appropriate formylating reagent e.g. 1- ⁇ i ⁇ eridine carboxaldehyde
  • the reaction is allowed to stir at -78 0 C for an additional several hours and slowly warmed to room temperature. Isolation by conventional means (e.g. extraction) affords compounds of Formula XX.
  • Compound of Formula XX is dissolved in a non-reactive solvent (e.g. tetrahydrofuran) and stirred under an inert atmosphere.
  • abase e.g. triethylamine
  • a catalyst e.g. 4-dimethylaminopyridine
  • the mixture is stirred for a few minutes, and then a reagent appropriate for the introduction of a protecting group (e.g. di-tert-butyldicarbonate) is added.
  • a protecting group e.g. di-tert-butyldicarbonate
  • the reaction is stirred overnight. Isolation by conventional means (e.g. extraction) affords compounds of Formula XXI.
  • lH-Pyrrolo[2,3-b]pyridine 1 is added to a stirred solution of base (e.g. potassium hydroxide) in an appropriate polar solvent (e.g. methanol).
  • base e.g. potassium hydroxide
  • an appropriate polar solvent e.g. methanol
  • Compound of Formula XXI is added, and the mixture is typically stirred at room temperature for several days. The solvent is evaporated and 1 M HCl is added to the residue. Isolation by conventional means (e.g. extraction, silica gel chromatography) affords compounds of Formula XXII and XXIII.
  • Step - 5 - Preparation of compounds of Formula XIV of Scheme 14 [0326] Typically, compounds of Formula XII and XIII are combined and dissolved in an appropriate polar aprotic solvent (e.g. acetonitrile). Reagents appropriate to effect the reduction (e.g. triethylsilane and trifluoro acetic acid) are added. Typically, the reaction is stirred at room temperature for several days. Isolation by conventional means (e.g. extraction, silica gel chromatography) affords compounds of Formula Ie.
  • an appropriate polar aprotic solvent e.g. acetonitrile
  • Reagents appropriate to effect the reduction e.g. triethylsilane and trifluoro acetic acid
  • the reaction is stirred at room temperature for several days. Isolation by conventional means (e.g. extraction, silica gel chromatography) affords compounds of Formula Ie.
  • Step 1 Preparation of ⁇ -chloro-I-methoxypyridineS-carbaldehyde (26)
  • 2-Chloro-6-methoxypyridine 25, 0.511 g, 3.56 mmol
  • tetrahydrofuran 10 mL
  • tert-butyllithium 1.7 M in pentane, 5.0 mL, 7.66 mmol
  • the reaction was allowed to stir for 1 hour.
  • Dimethylformamide (0.673 mL, 17.4 mmol) was added and the reaction was allowed to continue for an additional 30 minutes at -78 0 C, then stirred for 30 minutes outside of the dry-ice bath.
  • Step 4 Preparation 3-(6-chloro-2-methoxypyridin-3-ylmethyl)-l-(triisopropylsilyl)-lH- pyrrolo [2, 3 -b] pyridine (29)
  • Step 6 Preparation of[6-Methoxy-5-(lH-pyrrolo[2,3-b]pyridin-3-ylmethyl)-pyridin-2- yl]-(4-trifluoromethyl-benzyl)-amine (P-0012)
  • Step - 1 Preparation of(5-Bromo-6 ⁇ methyl-pyridin-2-yl)-(4-trtfluoromethyl-benzyl)- amine (34)
  • Step -3 Preparation of(5-Formyl-6-methyl-pyridin-2 ⁇ yl)-(4 ⁇ trifluoromethyl-benzyl)- carbamic acid tert-butyl ester (36)
  • Step - 4 Preparation of ⁇ 5-[Hydroxy-(lH-pyrrolo[2,3-b]pyridin-3-yl)-methyl]-6-methyl- pyridin-2-yl ⁇ -(4-trifluoromethyl-benzyl) ⁇ carbamic acid tert-butyl ester (37) and ⁇ 5- [Methoxy-(lH-pyrrolo[2,3-b]pyridin-3-yl)-methyl]-6-methyl-pyridin-2-yl ⁇ -(4- tri ⁇ uoromethyl-benzyl)-carbamic acid tert-butyl ester (38)
  • Step - 5 Preparation of[6-Methyl-5-(lH-pyrrolo[2,3-b]bipyridin-3-ylmethyl)-pyridin-2- yl]-(4-trifluoromethyl-benzyl)-amine (P-0013)
  • Step 1- Synthesis of(5-Bromo-pyridin-2-yl)-(4-chloro-benzyl)-amine (41) [0342] To 2-Amino-5-bromopyridine (15, 6.1O g, 0.0352 mol) in toluene (90.0 mL) were added 4-chlorobenzaldehyde (40, 5.00 g, 0.0356 mol), trifluoroacetic acid (8.0 mL, 0.10 mol) and triethylsilane (16.5 mL, 0.103 mol). The reaction was heated to reflux for 48 hours. The reaction was concentrated, poured into aqueous potassium carbonate and extracted with ethyl acetate. The organic layer was washed with brine, dried over sodium sulfate and concentrated. The crude residue was crystallized with ethyl acetate to give compound (41, 6.8 g, 65.4%).
  • Step 2 Synthesis of6-(4-Chloro-benzylamino)-pyridine-3-carbaldehyde (42) [0343] To (5-Bromo-pyridin-2-yl)-(4-chloro-benzyl)-amine (41, 10.00 g, 0.03360 mol) in tetrahydrofuran (400.0 mL) under an atmosphere of nitrogen at -78 0 C was added n- butyllithium (17.5 mL, 2.00 M in cyclohexane). After 90 minutes, tert-butyllithium (42.00 mL, 1.70 M in hexane) was added to the reaction.
  • N 5 N- dimethylformamide (6.9 mL, 0.089 mol) was added to the reaction.
  • the reaction mixture was stirred at -78 0 C for 2 hours, then allowed to warm to room temperature for 1 hour.
  • the reaction mixture was poured into water and extracted with ethyl acetate. The organic layer was washed with brine, dried over sodium sulfate and concentrated to give the crude compound, which was crystallized from tert-butoxyl methyl ether to provide compound (42, 7.66 g, 92.2%).
  • Step 3 Synthesis of(4-Chloro-benzyl) ⁇ (5-formyl-pyridin-2-yl)-carbamic acid tert-butyl ester (43)
  • Step 4 Synthesis of ⁇ 5-[(5-Bromo-lH-pyrrolo[2,3-b] pyridin-3-yl)-hydroxy-methyl] - pyridin-2-yl ⁇ -(4-chloro-benzyl)-carbamic acid tert-butyl ester (45) [0345] To 5-bromo-7-azaindole (44, 198.0 mg, 1.01 mmol) in methanol (30.0 mL, 0.741 mol) were added (4-Chloro-benzyl)-(5-formyl-pyridin-2-yl)-carbamic acid tert-butyl ester (43, 355.0 mg, 1.02 mmol) and potassium hydroxide (80.0 mg, 1.42 mmol).
  • Example 18 Synthesis of l-triisopropylsilanyl-lH-pyrrolo[2,3-b]pyridine-3- carbaldehyde 47.
  • Step 2 Synthesis ofN-5-[Hydroxy-(l-triisopropylsilanyl-lH-pyrrolo[2,3-b]pyridin-3-yl)- methyl]-pyridin-2-yl-4-trifluoromethyl-benzenesulfonamide (50):
  • N-[5-(lH-Pyrrolo[2,3-b] ⁇ yridin-3-ylmethyl)- ⁇ yridin-2-yl]-4-trifluoromethyl- benzamide P-0072 was synthesized in one step from (3-(6-Bromo-pyridin-3-ylmethyl)-l- triisopropylsilanyl-lH-pyrrolo[2,3-b]pyridine 6a as shown in Scheme 22.
  • Step 1 Synthesis ofN-[5-(lH-Pyrrolo[2,3-b]pyridin-3-ylmethyl)-pyridin-2-yl]-4- trifluoromethyl-benzamide (P-0072) :
  • Step 1 Synthesis of(5-Bromo-pyridin-2-ylmethyl)-(4-chloro-phenyl)-amine (54): [0361] To 5-Bromo-pyridine-2-carbaldehyde (52, 1.00 g, 5.38 mmol) in acetonitrile (50.0 mL) were added p-chloroaniline (53, 0.686 g, 5.38 mmol), triethylsilane (6.00 niL, 0.0376 mol) and trifluoroacetic acid (3.00 mL, 0.0389 mol). The reaction was heated to reflux for 3 hours. The reaction was concentrated, poured into water and then extracted with ethyl acetate.
  • the reaction was cooled to -78 °C, followed by addition of 1.70 M tert-butyllithium in hexane (1.58 mL). After 30 minutes, l-benzenesulfonyl-lH-pyrrolo[2,3-b]pyridine-3- carbaldehyde (55, 0.380 g, 1.33 mmol, prepared as described in Example 18) in tetrahydrofuran (10.0 mL) was added to the reaction. After 20 minutes, the reaction was allowed to warm to room temperature. The reaction was poured into water and extracted with ethyl acetate. The organic layer was dried over anhydrous sodium sulfate and filtered.
  • Step 2 Synthesis of 2,6-Difluoro-nicotinic acid methyl ester (60): [0367] To 2,6-difluoro-nicotinic acid (59, 5.60 g, 0.0352 mol) in methanol (60.0 mL) was added concentrated sulfuric acid (1.0 mL, 0.019 mol). The reaction was heated to reflux overnight, then poured into water, basified with IM potassium carbonate to pH around 9, and extracted with ethyl acetate. The organic layer was dried over anhydrous sodium sulfate and concentrated to give a yellow oil (60, 3.5 g, 57.0%).
  • Step 3 Synthesis of6-(4-Chloro-benzylamino)-2-fluoro-nicotinic acid methyl ester (62): [0368] To 2,6-difluoro-nicotinic acid methyl ester (60, 2.00 g, 0.0116 mol) in N 5 N- dimethylformamide (20.0 mL), under an atmosphere of nitrogen at -40 °C, was added p- chlorobenzylamine (61, 2.60 mL, 0.0214 mol). The reaction was stirred at -40 °C to -20 0 C for 2 hours, then poured into water and extracted with ethyl acetate. The organic layer was dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated and purified by silica gel column chromatography eluting with 25% ethyl acetate in hexane to give compound (62, 2.0 g, 58.7%).
  • Step 4 Synthesis of [6-(4-Chloro-benzylamino)-2-fluoro-pyridin-3-yl] -methanol (63): [0369] To 6-(4-Chloro-benzylamino)-2-fluoro-nicotinic acid methyl ester (62, 2.00 g, 6.79 mmol) in tetrahydrofuran (100.0 mL) was added lithium tetrahydroaluminate (13.6 mL, 1.00 M in Tetrahydrofuran) under an atmosphere of nitrogen. The reaction was stirred at room temperature overnight. To the reaction was added an excessive amount of NaSO 4 " 10H 2 O, and then stirred for 1 hour. Filtration, concentration and purification with silica gel column chromatography eluting with 30% ethyl acetate in hexane provided compound 63 (1.0 g, 55.0%).
  • Step 5 Synthesis of6-(4-Chloro-benzylamino)-2-fluoro-pyridine-3-carbaldehyde (64): [0370] To [6-(4-Chloro-benzylamino)-2-fluoro-pyridin-3-yl]-methanol (63, 1.0 g, 3.7 mmol) in tetrahydrofuran (50.0 mL) was added Dess-Martin periodinane (1.75 g, 4.12 mmol). The reaction was stirred at room temperature for 10 minutes, then poured into water and extracted with ethyl acetate. The organic layer was dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated and purified by silica gel column chromatography eluting with 20% ethyl acetate in hexane to give a white solid (64, 0.67 g, 68.0%).
  • Step 6 Synthesis of(4-Chloro-benzyl)-(6-fluoro-5-formyl-pyridin-2-yl)-carbamic acid tert-butyl ester (65):
  • Step 7 Synthesis of(5-[l-(tert-Butyl-dimethyl-silanyl)-lH-pyrrolo[2,3-b]pyridin-3-yl]- hydroxy-methyl-6-fluoro-pyridin-2 ⁇ yl)-(4-chloro-benzyl)-carbamic acid tert-butyl ester (67):
  • Step 8 Synthesis of(4-Chloro-benzyl)-[6-fluoro-5-(lH-pyrrolo[2,3-b]pyridin-3- ylmethyl)-pyridin-2-yl] -amine (P-0082):
  • Step 1 Synthesis of(4-Chloro-benzyl)-5-[hydroxy-(lH-pyrrolo[2,3-b]pyridin-3-yl)- methyl]-6-methoxy-pyridin-2-yl-carbamic acid tert-butyl ester (68): [0375] To lH-Pyrrolo[2,3-b]pyridine (1, 90.0 mg, 0.76 mmol) in methanol (30.0 niL) were added (4-chloro-benzyl)-(6-fluoro-5-formyl-pyridin-2-yl)-carbamic acid tert-butyl ester (65, 300.0 mg, 0.82 mmol) and potassium hydroxide (720.0 mg, 12.83 mmol) under an atmosphere of nitrogen.
  • Step 2 Synthesis of(4-Chloro-benzyl)-[6-methoxy-5-(lH-pyrrolo[2,3-b]pyridin-3- ylmethyl)-pyridin-2-yl] -amine (P-0081).
  • Step 1 Synthesis of S-Bromo-py ⁇ dine ⁇ -carboxylic acid (4-chloro-phenyl) -amide (70): [0378] To 5-Bromo-pyridine-2-carbonyl chloride (69, 0.76 g, 3.4 mmol) in acetonitrile (29.0 mL) were added p-chloroaniline (53, 0.702 g, 5.50 mmol), 4-dimethylamino- pyridine (0.12 g, 0.96 mmol) and pyridine (2.9 mL, 0.036 mol).
  • Step 2 Synthesis of5-[Hydroxy-(l-triisopropylsilanyl-lH-pyrrolo[2,3-b]pyridin-3-yl)- methyl] -pyridine-2-carboxylic acid (4-chloro-phenyl) -amide (71): [0379] To 5-Bromo-pyridine-2-carboxylic acid (4-chloro-phenyl)-amide (70, 0.50 g, 1.60 mmol) in tetrahydrofuran (20.0 mL), under an atmosphere of nitrogen at -78 °C, tert- butyllithium (3.02 mL, 1.70 M in Hexane) was added.
  • Step 1 Preparation of(6-Chloro-pyridin-3-yl)-(lH-pyrrolo[2,3-b]pyridin-3-yl)- methanone (7):
  • Step 2 Preparation of(lH ⁇ pyrrolo[2,3-b]pyridin-3-yl)-[6-(3-trifluoromethyl-benzyloxy)- pyridin-3-yl] -methanone (73):
  • Step 3 Preparation (lH-Pyrrolo[2,3-b]pyridin-3-yl)-[6-(3-trifluoromethyl-benzyloxy)- pyridin-3-yl] -methanol (74): [0386] To (lH-pyrrolo[2,3-b]pyridin-3-yl)-[6-(3-trifluoromethyl-benzyloxy)-pyridin-3- yl]-methanone 73 in ethanol was added sodium borohydride. After one hour, the reaction was quenched with water and extracted with ethyl acetate.
  • Step 1 Preparation of2,6-dichloropyridine-3-carbonyl chloride (76): [0390] To 2,6-dichloro ⁇ yridine-3-carboxylic acid (75, 1.00 g, 0.00521 mol) in dichloromethane (75 mL) was added 2 M Oxalyl chloride (2.61 mL, 0.727 g, 0.00573 mol). The solution began to show vigorous gas evolution, which slowed but continued for about 2 hours. The reaction was allowed to continue at room temperature for an additional 3 hours. The reaction was concentrated to give the compound as a brown oil that crystallized on standing (76, 1.09 g, 99%).
  • Step 1 Preparation of(6-(4-chlorobenzylamino)-2-chloropyridin-3-yl)(lH-pyrrolo[2,3- b]pyridin-3-yl)methanol (P-0050) :
  • Step 1 5-(2-Morpholin-4-yl-ethoxy)-lH-pyrrolo[2,3-b]pyridine (79): [0398] To 4-morpholineethanol (30 mL, 0.2 mol) in N, N-dimethylformamide (30 mL) was slowly added sodium hydride (7 g, 60% dispersion in mineral oil, 0.2 mol). After the solution turned clear, a solution of 5-bromo-7-azaindole (44, 1.0 g, 0.0051 mol) in N,N ⁇ dimethylformamide (5 mL) and copper(I) bromide (1.4 g, 0.0098 mol) were added. The reaction mixture was stirred at 120 0 C under nitrogen for 2 hours.
  • Step 1 Preparation of6-(4-Trifluoromethyl-benzylamino)-pyridine-3-carbaldehyde (18): [0401] To a solution of (5-bromo-pyridin-2-yl)-(4-trifluoromethyl-benzyl)-amine (17, 3.55 g, 0.0107 mol, commercially available, or prepared as described in Example 10) in tetrahydrofuran (150 mL) was added tert-butyllithium (13.2 niL, 1.70 M in pentane, 0.0224 mol) slowly under an atmosphere of nitrogen at -78 °C over 10 minutes. The reaction mixture was stirred at -78 °C for 90 minutes.
  • Step 4 Preparation of(5- ⁇ Hydroxy-[5-(2-morpholin-4-yl-ethoxy)-lH-pyrrolo[2,3- b]pyridin-3-yl]-methyl ⁇ -pyridin-2-yl)-(4 -trifluoromethyl-benzyl)-carbamic acid tert-butyl ester (80):
  • Step 5 Preparation of ⁇ 5-[5-(2-Morpholin-4-yl-ethoxy)-lH-pyrrolo[2,3-b]pyridin-3- ylmethyl]-pyridin-2-yl ⁇ -(4-trifluorom ethyl-benzyl)-amine (P-0065) : [0404] A mixture of (5- ⁇ Hydroxy-[5-(2-morpholin-4-yl-ethoxy)-lH-pyrrolo[2,3- b] ⁇ yridin-3-yl]-methyl ⁇ -pyridin-2-yl)-(4 -Mfluoromethyl-benzyl)-carbamic acid tert-butyl ester (80, 0.2 g, 0.3 mmol), triethylsilane (4 mL, 0.02 mol), and trifluoroacetic acid (2 mL, 0.02 mol) in acetonitrile (30 mL) was refluxed for 2 hours
  • Step 3 Preparation of 5-Triisopropylsilanyloxy-lH-pyrrolo [2,3 -b] pyridine (83): [0409] To a solution of lH-Pyrrolo[2,3-b]pyridin-5-ol (0.5 g, 0.004 mol) and IH- imidazole (0.98 g, 0.014 mol) in N,N-dimethylformamide (5 mL) was added triisopropylsilyl chloride (1 mL, 0.005 mol). The reaction mixture was stirred at room temperature overnight. Dichloromethane (10 mL) was added and the solution was washed with brine and dried over sodium sulfate. After removal of solvent, the residue was purified by silica gel column chromatography eluting with ethyl acetate in hexane to provide the compound as a viscous liquid (83, 0.4 g, 40%).
  • Step 4 Preparation of ⁇ 5-[Hydroxy-(5-triisopropylsilanyloxy-lH-pyrrolo[2,3-b]pyridin- 3-yl)-methyl]-pyridin-2-yl ⁇ -(4-trifluoromethyl-benzyl)-carbamic acid tert-butyl ester (84): [0410] A mixture of (5-Formyl-pyridin-2-yl)-(4-trifluoromethyl-benzyl)-carbamic acid tert-butyl ester (19, 41 mg, 0.11 mmol, prepared as described in Example 30), 5- triisopropylsilanyloxy-lH-pyrrolo[2,3-b]pyridme (83, 34 mg, 0.12 mmol) and potassium hydroxide (9.8 mg, 0.17 mmol) in methanol (10 mL) was stirred at room temperature overnight.
  • Step 5 Preparation of(4-Trifluoromethyl-benzyl)-[5-(5-triisopropylsilanyloxy-lH- pyrrolo[2, 3-b]pyridin-3-ylmethyl)-pyridin-2-yl] -amine (85) :
  • Step 6 Preparation of3-[6-(4-Trifluoromethyl-benzylamino)-pyridin-3-ylmethyl]-lH- pyrrolo[2, 3-b]pyridin-5-ol (P-0061) :
  • Step 1 Preparation of(6-Bromo-pyridin-3-yl)-(lH-pyrrolo[2,3-b]pyridin-3-yl)- methanone (87):
  • Step 2 Preparation ofN-[5-(lH-Pyrrolo[2,3-b]pyridine-3-carbonyl)-pyridin-2-yl]-4- trifluoromethyl-benzamide (P-0067):
  • Step 2 Preparation of 3-(6-Bromo-pyridin-3-ylmethyl)-lH-pyrrolo [2, 3-b] pyridine (91): [0420] A mixture of (6-bromo-pyridin-3-yl)-(lH-pyrrolo[ 2,3-b]pyridin-3-yl)-methanol (89, 1 g, 0.003 mol) and 3-[(6-bromo-pyridin-3-yl)-methoxy-methyl]-lH- ⁇ yrrolo[2,3- bjpyridine (90, 2 g, 0.006 mol), triethylsilane (1 mL, 0.006 mol), and trifluoroacetic acid (0.5 mL, 0.006 mol) in acetonitrile (25 mL) was refluxed for 2 hours.
PCT/US2006/018726 2005-05-17 2006-05-16 Pyrrol (2,3-b) pyridine derivatives protein kinase inhibitors WO2007013896A2 (en)

Priority Applications (9)

Application Number Priority Date Filing Date Title
MX2007014377A MX2007014377A (es) 2005-05-17 2006-05-16 Inhibidores de proteina cinasa de derivados de pirrol (2,3-b) piridina.
AU2006272951A AU2006272951A1 (en) 2005-05-17 2006-05-16 Pyrrol (2,3-b) pyridine derivatives protein kinase inhibitors
EP06813186A EP1885723A2 (en) 2005-05-17 2006-05-16 Pyrrolo[2,3-b]pyridine derivatives as protein kinase inhibitors
BRPI0610066-0A BRPI0610066A2 (pt) 2005-05-17 2006-05-16 compostos que modulam atividade de c-kit e c-fms e usos para estes
JP2008512402A JP2008545652A (ja) 2005-05-17 2006-05-16 c−kitおよびc−fms活性を調節する化合物およびその用途
CA002608733A CA2608733A1 (en) 2005-05-17 2006-05-16 Pyrrol (2,3-b) pyridine derivatives protein kinase inhibitors
NZ563444A NZ563444A (en) 2005-05-17 2006-05-16 Pyrrolo(2,3-b)pyridine derivatives as protein kinase inhibitors
IL187344A IL187344A0 (en) 2005-05-17 2007-11-13 Compounds modulating c-kit and c-fms activity and uses therefor
NO20075992A NO20075992L (no) 2005-05-17 2007-11-23 Forbindelser som modulerer C-Kit og C-FMS aktivitet og anvendelser derav

Applications Claiming Priority (10)

Application Number Priority Date Filing Date Title
US68206305P 2005-05-17 2005-05-17
US68205105P 2005-05-17 2005-05-17
US68204205P 2005-05-17 2005-05-17
US60/682,042 2005-05-17
US60/682,051 2005-05-17
US60/682,063 2005-05-17
US69296005P 2005-06-22 2005-06-22
US69275005P 2005-06-22 2005-06-22
US60/692,960 2005-06-22
US60/692,750 2005-06-22

Publications (2)

Publication Number Publication Date
WO2007013896A2 true WO2007013896A2 (en) 2007-02-01
WO2007013896A3 WO2007013896A3 (en) 2007-05-31

Family

ID=37683773

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2006/018726 WO2007013896A2 (en) 2005-05-17 2006-05-16 Pyrrol (2,3-b) pyridine derivatives protein kinase inhibitors

Country Status (14)

Country Link
US (2) US7846941B2 (US07846941-20101207-C00154.png)
EP (1) EP1885723A2 (US07846941-20101207-C00154.png)
JP (1) JP2008545652A (US07846941-20101207-C00154.png)
KR (1) KR20080027775A (US07846941-20101207-C00154.png)
AU (1) AU2006272951A1 (US07846941-20101207-C00154.png)
BR (1) BRPI0610066A2 (US07846941-20101207-C00154.png)
CA (1) CA2608733A1 (US07846941-20101207-C00154.png)
CR (1) CR9591A (US07846941-20101207-C00154.png)
EC (1) ECSP077985A (US07846941-20101207-C00154.png)
IL (1) IL187344A0 (US07846941-20101207-C00154.png)
MX (1) MX2007014377A (US07846941-20101207-C00154.png)
NO (1) NO20075992L (US07846941-20101207-C00154.png)
NZ (1) NZ563444A (US07846941-20101207-C00154.png)
WO (1) WO2007013896A2 (US07846941-20101207-C00154.png)

Cited By (60)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008064265A2 (en) * 2006-11-22 2008-05-29 Plexxikon, Inc. Compounds modulating c-fms and/or c-kit activity and uses therefor
WO2008079909A1 (en) * 2006-12-21 2008-07-03 Plexxikon, Inc. Pyrrolo [2,3-b] pyridines as kinase modulators
WO2008080001A2 (en) * 2006-12-21 2008-07-03 Plexxikon, Inc. Compounds and methods for kinase modulation, and indications therefor
JP2009523812A (ja) * 2006-01-19 2009-06-25 オーエスアイ・ファーマスーティカルズ・インコーポレーテッド 融合へテロ二環式キナーゼ阻害剤
WO2009111280A1 (en) 2008-02-29 2009-09-11 Array Biopharma Inc. N- (6-aminopyridin-3-yl) -3- (sulfonamido) benzamide derivatives as b-raf inhibitors for the treatment of cancer
WO2009143024A2 (en) * 2008-05-19 2009-11-26 Plexxikon, Inc. Compounds and methods for kinase modulation, and indications therefor
WO2009143018A2 (en) * 2008-05-19 2009-11-26 Plexxikon, Inc. Compounds and methods for kinase modulation, and indications therefor
WO2009152087A1 (en) * 2008-06-10 2009-12-17 Plexxikon, Inc. Bicyclic heteroaryl compounds and methods for kinase modulation, and indications therefor
WO2010111527A1 (en) * 2009-03-26 2010-09-30 Plexxikon, Inc. Pyrazolo [ 3, 4 -b] pyridines as kinase inhibitors and their medical use
WO2011025968A1 (en) 2009-08-28 2011-03-03 Array Biopharma Inc. 1h-pyrazolo [ 3, 4-b] pyridine compounds for inhibiting raf kinase
EP2313411A1 (en) * 2008-06-10 2011-04-27 Plexxikon, Inc. 5h-pyrr0l0 [2,3-b]pyrazine derivatives for kinase modulation, and indications therefor
US8173635B2 (en) 2007-11-02 2012-05-08 Vertex Pharmaceuticals Incorporated Kinase inhibitors
US8338452B2 (en) 2008-02-29 2012-12-25 Array Biopharma Inc. Raf inhibitor compounds and methods of use thereof
EP2560972A2 (en) * 2010-04-21 2013-02-27 Plexxikon, Inc. Compounds and methods for kinase modulation, and indications therefor
US8394795B2 (en) 2008-02-29 2013-03-12 Array Biopharma Inc. Pyrazole [3, 4-B] pyridine Raf inhibitors
US8580803B2 (en) 2009-12-30 2013-11-12 Arqule, Inc. Substituted pyrrolo-aminopyrimidine compounds
US8648086B2 (en) 2009-08-24 2014-02-11 Ascepion Pharmaceuticals, Inc. 5,6-bicyclic heteroaryl-containing urea compounds as kinase inhibitors
US8741920B2 (en) 2009-08-03 2014-06-03 Hoffmann-La Roche, Inc. Process for the manufacture of pharmaceutically active compounds
WO2014206344A1 (en) 2013-06-28 2014-12-31 Beigene, Ltd. Fused tricyclic amide compounds as multiple kinase inhibitors
US9163021B2 (en) 2012-10-04 2015-10-20 Pfizer Limited Pyrrolo[3,2-c]pyridine tropomyosin-related kinase inhibitors
US9358235B2 (en) 2012-03-19 2016-06-07 Plexxikon Inc. Kinase modulation, and indications therefor
US9440969B2 (en) 2009-12-23 2016-09-13 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9550768B2 (en) 2011-05-17 2017-01-24 Plexxikon Inc. Kinase modulation and indications therefor
EP3124472A1 (en) 2011-12-31 2017-02-01 BeiGene, Ltd. Fused tricyclic compounds as raf kinase inhibitors
US9617267B2 (en) 2009-11-18 2017-04-11 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9624213B2 (en) 2011-02-07 2017-04-18 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9630968B1 (en) 2015-12-23 2017-04-25 Arqule, Inc. Tetrahydropyranyl amino-pyrrolopyrimidinone and methods of use thereof
US9663517B2 (en) 2009-04-03 2017-05-30 Plexxikon Inc. Compositions and uses thereof
US9676748B2 (en) 2012-12-21 2017-06-13 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9695169B2 (en) 2012-05-31 2017-07-04 Plexxikon Inc. Synthesis of heterocyclic compounds
US9718847B2 (en) 2013-03-15 2017-08-01 Plexxikon Inc. Heterocyclic compounds and uses thereof
US9771363B2 (en) 2015-09-21 2017-09-26 Plexxikon Inc. Heterocyclic compounds and uses thereof
US9771369B2 (en) 2014-03-04 2017-09-26 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9822109B2 (en) 2013-03-15 2017-11-21 Plexxikon Inc. Heterocyclic compounds and uses thereof
US9844539B2 (en) 2007-07-17 2017-12-19 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9856259B2 (en) 2014-09-15 2018-01-02 Plexxikon Inc. Heterocyclic compounds and uses thereof
US9873700B2 (en) 2013-05-30 2018-01-23 Plexxikon Inc. Compounds for kinase modulation, and indications therefor
US9938273B2 (en) 2015-12-07 2018-04-10 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US10160747B2 (en) 2016-03-16 2018-12-25 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US10160755B2 (en) 2015-04-08 2018-12-25 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US10227357B2 (en) 2012-09-06 2019-03-12 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US10428067B2 (en) 2017-06-07 2019-10-01 Plexxikon Inc. Compounds and methods for kinase modulation
US10435404B2 (en) 2017-07-25 2019-10-08 Plexxikon Inc. Formulations of a compound modulating kinases
EP3550031A1 (en) 2012-07-24 2019-10-09 Pharmacyclics, LLC Mutations associated with resistance to inhibitors of bruton's tyrosine kinase (btk)
US10508085B2 (en) 2016-09-22 2019-12-17 Plexxikon Inc. Compounds and methods for IDO and TDO modulation, and indications therefor
US10577366B2 (en) 2017-03-20 2020-03-03 Plexxikon Inc. Crystalline forms of a compound that inhibits bromodomain
WO2020079581A1 (en) 2018-10-16 2020-04-23 Novartis Ag Tumor mutation burden alone or in combination with immune markers as biomarkers for predicting response to targeted therapy
US10703757B2 (en) 2016-12-23 2020-07-07 Plexxikon Inc. Compounds and methods for CDK8 modulation and indications therefor
US10717735B2 (en) 2017-10-13 2020-07-21 Plexxikon Inc. Solid forms of a compound for modulating kinases
US10829484B2 (en) 2015-07-28 2020-11-10 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US11020398B2 (en) 2016-08-24 2021-06-01 Arqule, Inc. Amino-pyrrolopyrimidinone compounds and methods of use thereof
WO2021144360A1 (en) 2020-01-17 2021-07-22 F. Hoffmann-La Roche Ag Small molecule csf-1r inhibitors in therapeutic and cosmetic uses
US11103505B2 (en) 2017-10-27 2021-08-31 Plexxikon Inc. Formulations of a compound modulating kinases
US11149011B2 (en) 2018-03-20 2021-10-19 Plexxikon Inc. Compounds and methods for IDO and TDO modulation, and indications therefor
US11274108B2 (en) 2015-07-20 2022-03-15 Genzyme Corporation Colony stimulating factor-1 receptor (CSF-1R) inhibitors
WO2022195551A1 (en) 2021-03-18 2022-09-22 Novartis Ag Biomarkers for cancer and methods of use thereof
US11530216B2 (en) 2020-12-23 2022-12-20 Genzyme Corporation Deuterated colony stimulating factor-1 receptor (CSF-1R) inhibitors
US11731968B2 (en) 2018-06-21 2023-08-22 Heparegenix Gmbh Tricyclic protein kinase inhibitors for promoting liver regeneration or reducing or preventing hepatocyte death
US11858927B2 (en) 2018-01-31 2024-01-02 Heparegenix Gmbh Protein kinase MKK4 inhibitors for promoting liver regeneration or reducing or preventing hepatocyte death
US11912701B2 (en) 2018-07-16 2024-02-27 Heparegenix Gmbh Protein kinase inhibitors for promoting liver regeneration or reducing or preventing hepatocyte death

Families Citing this family (37)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SI1696920T1 (sl) 2003-12-19 2015-02-27 Plexxikon Inc. Spojine in postopki za razvoj modulatorjev ret
US7498342B2 (en) * 2004-06-17 2009-03-03 Plexxikon, Inc. Compounds modulating c-kit activity
KR20070053205A (ko) 2004-09-17 2007-05-23 에자이 알앤드디 매니지먼트 가부시키가이샤 의약 조성물
US7846941B2 (en) 2005-05-17 2010-12-07 Plexxikon, Inc. Compounds modulating c-kit and c-fms activity and uses therefor
PE20070100A1 (es) * 2005-06-22 2007-03-10 Plexxikon Inc DERIVADOS DE PIRROLO[2,3-b]PIRIDINA COMO MODULADORES DE QUINASAS
WO2007015578A1 (ja) 2005-08-02 2007-02-08 Eisai R & D Management Co., Ltd. 血管新生阻害物質の効果を検定する方法
WO2007026864A1 (ja) * 2005-09-01 2007-03-08 Eisai R & D Management Co., Ltd. 崩壊性の改善された医薬組成物の製造方法
EP2027087A2 (en) * 2006-05-18 2009-02-25 MannKind Corporation Intracellular kinase inhibitors
RU2448708C3 (ru) 2006-05-18 2017-09-28 Эйсай Ар Энд Ди Менеджмент Ко., Лтд. Противоопухолевое средство против рака щитовидной железы
PE20121126A1 (es) * 2006-12-21 2012-08-24 Plexxikon Inc Compuestos pirrolo [2,3-b] piridinas como moduladores de quinasa
ES2392482T3 (es) * 2008-02-29 2012-12-11 Array Biopharma, Inc. Derivados de imidazo[4,5-b] piridina usados como inhibidores de RAF
WO2010059771A1 (en) 2008-11-20 2010-05-27 Osi Pharmaceuticals, Inc. Substituted pyrrolo[2,3-b]-pyridines and-pyrazines
MX2011005597A (es) * 2008-12-05 2011-06-16 Hoffmann La Roche Pirrolopirazinil-ureas como inhibidores de cinasas.
AU2010315126B2 (en) * 2009-11-06 2015-06-25 Plexxikon, Inc. Compounds and methods for kinase modulation, and indications therefor
RS53500B1 (en) * 2009-12-21 2015-02-27 Array Biopharma, Inc. N- (1H-INDAZOL-4-IL) IMIDASO [1,2-A] PIRIDINE-3-CARBOXAMIDE SUBSTITUTED COMPOUNDS AS CFMS INHIBITORS
AR081039A1 (es) 2010-05-14 2012-05-30 Osi Pharmaceuticals Llc Inhibidores biciclicos fusionados de quinasa
EP2569315A1 (en) 2010-05-14 2013-03-20 OSI Pharmaceuticals, LLC Fused bicyclic kinase inhibitors
TWI558702B (zh) 2011-02-21 2016-11-21 普雷辛肯公司 醫藥活性物質的固態形式
MX2013011612A (es) * 2011-04-05 2013-10-17 Pfizer Ltd Derivados de pirrolo[2,3-d]pirimidina como inhibidores de quinasa relacionados con tropomiosina.
CA2828946C (en) 2011-04-18 2016-06-21 Eisai R&D Management Co., Ltd. Therapeutic agent for tumor
EP2710003A1 (en) 2011-05-16 2014-03-26 OSI Pharmaceuticals, LLC Fused bicyclic kinase inhibitors
WO2012166899A2 (en) 2011-06-03 2012-12-06 Eisai R&D Management Co., Ltd. Biomarkers for predicting and assessing responsiveness of thyroid and kidney cancer subjects to lenvatinib compounds
BR112015009004A8 (pt) 2012-12-21 2021-07-20 Eisai R&D Man Co Ltd forma amorfa de derivado de quinolina e método de produção da mesma
CA2912219C (en) 2013-05-14 2021-11-16 Eisai R&D Management Co., Ltd. Biomarkers for predicting and assessing responsiveness of endometrial cancer subjects to lenvatinib compounds
TW201613919A (en) 2014-07-02 2016-04-16 Pharmacyclics Llc Inhibitors of Bruton's tyrosine kinase
KR20230043234A (ko) 2014-08-28 2023-03-30 에자이 알앤드디 매니지먼트 가부시키가이샤 고순도의 퀴놀린 유도체 및 이를 제조하는 방법
AU2016224583B2 (en) 2015-02-25 2021-06-03 Eisai R&D Management Co., Ltd. Method for suppressing bitterness of quinoline derivative
KR20170122809A (ko) 2015-03-04 2017-11-06 머크 샤프 앤드 돔 코포레이션 암을 치료하기 위한 pd-1 길항제 및 vegfr/fgfr/ret 티로신 키나제 억제제의 조합
MX2017013975A (es) 2015-05-06 2018-01-16 Plexxikon Inc Formas solidas de quinasas que modulan un compuesto.
US9745298B2 (en) 2015-05-06 2017-08-29 Plexxikon Inc. Synthesis of a compound that modulates kinases
CA2988707C (en) 2015-06-16 2023-10-10 Eisai R&D Management Co., Ltd. Combination of cbp/catenin inhibitor and immune checkpoint inhibitor for treating cancer
US10669278B2 (en) * 2016-09-16 2020-06-02 Helmholtz Zentrum Muenchen—Deutsches Forschungszentrum Für Gesundheit Und Umwelt (Gmbh) TRAF 6 inhibitors
JP7131837B2 (ja) * 2017-06-19 2022-09-06 アビスコ セラピューティクス カンパニー リミテッド Csf-1r阻害活性を有するアザアリール誘導体、その製造方法および応用
EP3953351A1 (en) 2019-04-09 2022-02-16 Plexxikon Inc. Condensed azines for ep300 or cbp modulation and indications therefor
US11807626B2 (en) 2020-04-23 2023-11-07 Opna Bio SA Compounds and methods for CD73 modulation and indications therefor
TW202227068A (zh) 2020-08-21 2022-07-16 美商普雷辛肯公司 組合藥物療法
CN115557985B (zh) * 2022-12-06 2023-03-10 成都泰蓉生物科技有限公司 一种选择性裂解硫醚中c-s键合成硅烷的方法

Citations (48)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5143854A (en) 1989-06-07 1992-09-01 Affymax Technologies N.V. Large scale photolithographic solid phase synthesis of polypeptides and receptor binding screening thereof
US5426039A (en) 1993-09-08 1995-06-20 Bio-Rad Laboratories, Inc. Direct molecular cloning of primer extended DNA containing an alkane diol
US5434049A (en) 1992-02-28 1995-07-18 Hitachi, Ltd. Separation of polynucleotides using supports having a plurality of electrode-containing cells
US5449614A (en) 1988-09-01 1995-09-12 Whitehead Institue For Biomedical Research Recombinant retroviruses with amphotropic and ecotropic host ranges
US5474935A (en) 1990-05-23 1995-12-12 The United States Of America As Represented By The Department Of Health And Human Services Adeno-associated virus (AAV)-based eucaryotic vectors
WO1996017958A1 (en) 1994-12-09 1996-06-13 The Regents Of The University Of California Comparative fluorescence hybridization to nucleic acid arrays
WO1996018738A2 (en) 1994-12-15 1996-06-20 Sugen, Inc. PROBIN TYROSINE KINASE (PYK2) ITS cDNA CLONING AND ITS USES
US5556752A (en) 1994-10-24 1996-09-17 Affymetrix, Inc. Surface-bound, unimolecular, double-stranded DNA
US5580859A (en) 1989-03-21 1996-12-03 Vical Incorporated Delivery of exogenous DNA sequences in a mammal
US5631236A (en) 1993-08-26 1997-05-20 Baylor College Of Medicine Gene therapy for solid tumors, using a DNA sequence encoding HSV-Tk or VZV-Tk
US5632957A (en) 1993-11-01 1997-05-27 Nanogen Molecular biological diagnostic systems including electrodes
US5658775A (en) 1988-05-17 1997-08-19 Sloan-Kettering Institute For Cancer Research Double copy retroviral vector
WO1997046313A1 (en) 1996-06-07 1997-12-11 Eos Biotechnology, Inc. Immobilised linear oligonucleotide arrays
US5700637A (en) 1988-05-03 1997-12-23 Isis Innovation Limited Apparatus and method for analyzing polynucleotide sequences and method of generating oligonucleotide arrays
US5721118A (en) 1995-10-31 1998-02-24 The Regents Of The University Of California, San Diego Mammalian artificial chromosomes and methods of using same
US5744305A (en) 1989-06-07 1998-04-28 Affymetrix, Inc. Arrays of materials attached to a substrate
US5770456A (en) 1989-06-07 1998-06-23 Affymetrix, Inc. Cyclic nucleic acid and polypeptide arrays
US5800992A (en) 1989-06-07 1998-09-01 Fodor; Stephen P.A. Method of detecting nucleic acids
US5807522A (en) 1994-06-17 1998-09-15 The Board Of Trustees Of The Leland Stanford Junior University Methods for fabricating microarrays of biological samples
US5856174A (en) 1995-06-29 1999-01-05 Affymetrix, Inc. Integrated nucleic acid diagnostic device
WO1999009217A1 (en) 1997-08-15 1999-02-25 Hyseq, Inc. Methods and compositions for detection or quantification of nucleic acid species
US5959098A (en) 1996-04-17 1999-09-28 Affymetrix, Inc. Substrate preparation process
US5965452A (en) 1996-07-09 1999-10-12 Nanogen, Inc. Multiplexed active biologic array
WO1999051773A1 (en) 1998-04-03 1999-10-14 Phylos, Inc. Addressable protein arrays
US6013440A (en) 1996-03-11 2000-01-11 Affymetrix, Inc. Nucleic acid affinity columns
US6022963A (en) 1995-12-15 2000-02-08 Affymetrix, Inc. Synthesis of oligonucleotide arrays using photocleavable protecting groups
US6025155A (en) 1996-04-10 2000-02-15 Chromos Molecular Systems, Inc. Artificial chromosomes, uses thereof and methods for preparing artificial chromosomes
US6045996A (en) 1993-10-26 2000-04-04 Affymetrix, Inc. Hybridization assays on oligonucleotide arrays
US6048695A (en) 1998-05-04 2000-04-11 Baylor College Of Medicine Chemically modified nucleic acids and methods for coupling nucleic acids to solid support
US6054270A (en) 1988-05-03 2000-04-25 Oxford Gene Technology Limited Analying polynucleotide sequences
US6096718A (en) 1997-06-05 2000-08-01 Gene Targeting Corp. Tissue specific adenovirus vectors for breast cancer treatment
US6107478A (en) 1995-09-08 2000-08-22 Pedersen; Finn Skou Retroviral vector, a replication system for said vector and avian or mammalian cells transfected with said vector
US6110456A (en) 1995-06-07 2000-08-29 Yale University Oral delivery or adeno-associated viral vectors
US6110458A (en) 1991-07-26 2000-08-29 University Of Rochester Cancer therapy utilizing ex vivo gene transduction
US6113913A (en) 1998-06-26 2000-09-05 Genvec, Inc. Recombinant adenovirus
US6117681A (en) 1995-03-29 2000-09-12 Bavarian Nordic Research Inst. A/S Pseudotyped retroviral particles
US20010008765A1 (en) 1999-12-06 2001-07-19 Fuji Photo Film Co., Ltd. DNA chip and reactive solid carrier
US20010012537A1 (en) 1999-07-30 2001-08-09 Anderson Norman G. Dry deposition of materials for microarrays using matrix displacement
US20010014449A1 (en) 1993-11-01 2001-08-16 Michael I. Nerenberg Methods for determination of single nucleic acid polymorphisms using bioelectronic microchip
US20010014448A1 (en) 1997-09-30 2001-08-16 Surmodics, Inc. Target molecule attachment to surfaces
US6277628B1 (en) 1998-10-02 2001-08-21 Incyte Genomics, Inc. Linear microarrays
US6277489B1 (en) 1998-12-04 2001-08-21 The Regents Of The University Of California Support for high performance affinity chromatography and other uses
US20010016322A1 (en) 1999-04-27 2001-08-23 Caren Michael P. Method of performing array-based hybridization assays using thermal inkjet deposition of sample fluids
US20010018642A1 (en) 1997-07-25 2001-08-30 David Balaban Method and system for providing a probe array chip design database
US20010019827A1 (en) 1997-09-11 2001-09-06 Dawson Elliott P. Method of making high density arrays
US20040002534A1 (en) 1999-12-22 2004-01-01 Sugen, Inc. Methods of modulating c-kit tyrosine protein kinase function with indolinone compounds
WO2004016610A1 (en) 2002-08-14 2004-02-26 Astrazeneca Ab Substituted pyrrolopyridines
US20040077595A1 (en) 2002-09-06 2004-04-22 Insert Therapeutics, Inc. Cyclodextrin-based polymers for therapeutics delivery

Family Cites Families (86)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US617687A (en) * 1899-01-10 Vapor-burning apparatus
US2234705A (en) * 1940-04-12 1941-03-11 Eastman Kodak Co Cellulose organic derivative composition containing esters of monoalkoxy benzoic acids
US2413258A (en) * 1942-07-07 1946-12-24 United Gas Improvement Co Polystyrene-type resins plasticized with high boiling fatty acid alkyl esters
DE2413258A1 (de) 1974-03-20 1975-10-02 Bayer Ag Alkoxycarbonylphenylharnstoffe, verfahren zu ihrer herstellung und ihre verwendung als herbizide
GB1573212A (en) * 1976-04-15 1980-08-20 Technicon Instr Immunoassay for gentamicin
US4664504A (en) * 1983-01-20 1987-05-12 Tokyo Shibaura Denki Kabushiki Kaisha Image forming apparatus
US4568649A (en) * 1983-02-22 1986-02-04 Immunex Corporation Immediate ligand detection assay
US4626513A (en) * 1983-11-10 1986-12-02 Massachusetts General Hospital Method and apparatus for ligand detection
DE3483099D1 (de) 1984-03-15 1990-10-04 Immunex Corp Test zur sofortigen feststellung von liganden, testsatz und seine herstellung.
US5688655A (en) * 1988-02-10 1997-11-18 Ict Pharmaceuticals, Inc. Method of screening for protein inhibitors and activators
DE4022414A1 (de) 1990-07-13 1992-01-16 Bayer Ag Substituierte pyrrolo-pyridine
FR2687402B1 (fr) * 1992-02-14 1995-06-30 Lipha Nouveaux azaindoles, procedes de preparation et medicaments les contenant.
GB9226855D0 (en) * 1992-12-23 1993-02-17 Erba Carlo Spa Vinylene-azaindole derivatives and process for their preparation
DE69420394T2 (de) * 1993-03-01 2000-04-13 Merck Sharp & Dohme Pyrrolopyridinderivate als dopaminrezeptor liganden
CN1118598A (zh) * 1993-03-01 1996-03-13 默克·夏普-道姆公司 吡咯并吡啶衍生物
US5576319A (en) * 1993-03-01 1996-11-19 Merck, Sharp & Dohme Ltd. Pyrrolo-pyridine derivatives
US5840485A (en) * 1993-05-27 1998-11-24 Selectide Corporation Topologically segregated, encoded solid phase libraries
ES2289194T3 (es) * 1993-05-27 2008-02-01 Aventis Pharmaceuticals Inc. Bibliotecas en fase solida codificadas, segregadas topologicamente.
US5763198A (en) * 1994-07-22 1998-06-09 Sugen, Inc. Screening assays for compounds
GB9416189D0 (en) 1994-08-10 1994-09-28 Merck Sharp & Dohme Therapeutic agents
GB9416162D0 (en) 1994-08-10 1994-09-28 Merck Sharp & Dohme Therapeutic agents
GB2298199A (en) * 1995-02-21 1996-08-28 Merck Sharp & Dohme Synthesis of azaindoles
GB9503400D0 (en) 1995-02-21 1995-04-12 Merck Sharp & Dohme Therpeutic agents
GB2299581A (en) 1995-04-07 1996-10-09 Merck Sharp & Dohme 3-(Tetrahydropyridin-1-yl-methyl)pyrrolo[2,3-b]pyridine derivatives as ligands for dopamine receptor subtypes
GB9507291D0 (en) * 1995-04-07 1995-05-31 Merck Sharp & Dohme Therapeutic agents
US5747276A (en) * 1995-09-15 1998-05-05 The Scripps Research Institute Screening methods for the identification of novel antibiotics
US5804585A (en) * 1996-04-15 1998-09-08 Texas Biotechnology Corporation Thieno-pyridine sulfonamides derivatives thereof and related compounds that modulate the activity of endothelin
US6294330B1 (en) * 1997-01-31 2001-09-25 Odyssey Pharmaceuticals Inc. Protein fragment complementation assays for the detection of biological or drug interactions
EP0972071A4 (en) * 1997-03-07 2004-04-21 Tropix Inc TEST ARRANGEMENT FOR PROTEAS INHIBITORS
US6235769B1 (en) * 1997-07-03 2001-05-22 Sugen, Inc. Methods of preventing and treating neurological disorders with compounds that modulate the function of the C-RET receptor protein tyrosine kinase
US6161776A (en) * 1997-08-12 2000-12-19 Nibco Inc. Multi-layered, porous mat turf irrigation apparatus and method
US6178384B1 (en) * 1997-09-29 2001-01-23 The Trustees Of Columbia University In The City Of New York Method and apparatus for selecting a molecule based on conformational free energy
CA2319495A1 (en) * 1998-06-08 1999-12-16 Advanced Medicine, Inc. Multibinding inhibitors of microsomal triglyceride transferase protein
FR2793793B1 (fr) 1999-05-19 2004-02-27 Adir Nouveaux derives dimeriques substitues, leur procede de preparation et les compositions pharmaceutiques qui les contiennent
CN101310771B (zh) 2001-04-11 2012-08-08 千寿制药株式会社 视觉功能障碍改善剂
WO2002085896A1 (en) 2001-04-24 2002-10-31 Wyeth Antidepressant azaheterocyclylmethyl derivatives of 2,3-dihydro-1,4-benzodioxan
GB0114417D0 (en) * 2001-06-13 2001-08-08 Boc Group Plc Lubricating systems for regenerative vacuum pumps
EP1267111A1 (en) 2001-06-15 2002-12-18 Dsm N.V. Pressurized fluid conduit
WO2002102783A1 (en) 2001-06-19 2002-12-27 Merck & Co., Inc. Tyrosine kinase inhibitors
US7291639B2 (en) 2001-06-20 2007-11-06 Wyeth Aryloxy-acetic acid compounds useful as inhibitors of plasminogen activator inhibitor-1 (PAI-1)
UA75425C2 (en) 2001-07-09 2006-04-17 Piperazine oxime derivatives with antagonistic activity to nk-1 receptor, use thereof, a pharmaceutical composition based thereon, a method for producing and a method for producing intermediary compounds
GB0117583D0 (en) 2001-07-19 2001-09-12 Astrazeneca Ab Novel compounds
US6858860B2 (en) * 2001-07-24 2005-02-22 Seiko Epson Corporation Apparatus and method for measuring natural period of liquid
GB0118479D0 (en) 2001-07-28 2001-09-19 Astrazeneca Ab Novel compounds
WO2003028724A1 (en) 2001-10-04 2003-04-10 Smithkline Beecham Corporation Chk1 kinase inhibitors
US20050014942A1 (en) 2001-10-30 2005-01-20 Yasufumi Maruyama Amide derivatives and drugs
US20030236277A1 (en) 2002-02-14 2003-12-25 Kadow John F. Indole, azaindole and related heterocyclic pyrrolidine derivatives
US6884889B2 (en) 2002-03-25 2005-04-26 Bristol-Myers Squibb Co. Processes for the preparation of antiviral 7-azaindole derivatives
AU2003214414B2 (en) 2002-03-28 2008-10-09 Eisai R & D Management Co., Ltd. Azaindoles as inhibitors of c-Jun N-terminal kinases
EP1490364B1 (en) 2002-03-28 2007-09-26 Eisai R&D Management Co., Ltd. 7-azaindoles as inhibitors of c-jun n-terminal kinases for the treatment of neurodegenerative disorders
UA78999C2 (en) 2002-06-04 2007-05-10 Wyeth Corp 1-(aminoalkyl)-3-sulfonylazaindoles as ligands of 5-hydroxytryptamine-6
TW200403243A (en) 2002-07-18 2004-03-01 Wyeth Corp 1-Heterocyclylalkyl-3-sulfonylazaindole or-azaindazole derivatives as 5-hydroxytryptamine-6 ligands
TWI329112B (en) 2002-07-19 2010-08-21 Bristol Myers Squibb Co Novel inhibitors of kinases
US6878887B2 (en) * 2002-08-07 2005-04-12 Matsushita Electric Industrial Co., Ltd. Anti-malfunction mechanism for variable output device
EP1558751A4 (en) * 2002-09-16 2007-08-22 Plexxikon Inc CRYSTALLINE STRUCTURE OF PROTEIN KINASE PIM-1
US7696225B2 (en) * 2003-01-06 2010-04-13 Osi Pharmaceuticals, Inc. (2-carboxamido)(3-Amino) thiophene compounds
SE0300120D0 (sv) 2003-01-17 2003-01-17 Astrazeneca Ab Novel compounds
SE0300119D0 (sv) 2003-01-17 2003-01-17 Astrazeneca Ab Novel compounds
US20050085463A1 (en) * 2003-01-23 2005-04-21 Weiner David M. Use of N-desmethylclozapine to treat human neuropsychiatric disease
BRPI0407493A (pt) * 2003-02-14 2006-02-14 Wyeth Corp derivados heterociclil-3-sulfinilazaindol ou -azaindazol como ligantes de 5-hidroxitriptamina-6
WO2004078923A2 (en) * 2003-02-28 2004-09-16 Plexxikon, Inc. Pyk2 crystal structure and uses
US20050164300A1 (en) * 2003-09-15 2005-07-28 Plexxikon, Inc. Molecular scaffolds for kinase ligand development
DE10357510A1 (de) 2003-12-09 2005-07-07 Bayer Healthcare Ag Heteroarylsubstituierte Benzole
SI1696920T1 (sl) * 2003-12-19 2015-02-27 Plexxikon Inc. Spojine in postopki za razvoj modulatorjev ret
GB0330043D0 (en) 2003-12-24 2004-01-28 Pharmacia Italia Spa Pyrrolo [2,3-b] pyridine derivatives active as kinase inhibitors process for their preparation and pharmaceutical compositions comprising them
GB0330042D0 (en) 2003-12-24 2004-01-28 Pharmacia Italia Spa Pyrrolo [2,3-b] pyridine derivatives active as kinase inhibitors process for their preparation and pharmaceutical compositions them
GB0403635D0 (en) 2004-02-18 2004-03-24 Devgen Nv Pyridinocarboxamides with improved activity as kinase inhibitors
GB0405055D0 (en) 2004-03-05 2004-04-07 Eisai London Res Lab Ltd JNK inhibitors
MXPA06011327A (es) 2004-03-30 2006-12-15 Vertex Pharma Azaindoles utiles como inhibidores de jak y otras proteinas cinasas.
MXPA06013805A (es) * 2004-05-27 2007-02-02 Pfizer Prod Inc Derivados de pirrolopirimidina de utilidad en el tratamiento contra el cancer.
US7498342B2 (en) * 2004-06-17 2009-03-03 Plexxikon, Inc. Compounds modulating c-kit activity
EP1755597A2 (en) * 2004-06-17 2007-02-28 Plexxikon, Inc. Azaindoles modulating c-kit activity and uses therefor
EP1765819B1 (en) 2004-06-30 2014-03-12 Vertex Pharmaceuticals Inc. Azaindoles useful as inhibitors of protein kinases
BRPI0513899A (pt) * 2004-07-27 2008-05-20 Sgx Pharmaceuticals Inc moduladores de cinase heterocìclica de anel fundido
US7626021B2 (en) * 2004-07-27 2009-12-01 Sgx Pharmaceuticals, Inc. Fused ring heterocycle kinase modulators
US7709645B2 (en) * 2004-07-27 2010-05-04 Sgx Pharmaceuticals, Inc. Pyrrolo-pyridine kinase modulators
US7361764B2 (en) * 2004-07-27 2008-04-22 Sgx Pharmaceuticals, Inc. Pyrrolo-pyridine kinase modulators
CA2573362A1 (en) * 2004-07-27 2006-02-09 Sgx Pharmaceuticals, Inc. Pyrrolo-pyridine kinase modulators
ATE524467T1 (de) 2005-04-25 2011-09-15 Merck Patent Gmbh Neuartige aza-heterozyklen als kinase-inhibitoren
US7846941B2 (en) 2005-05-17 2010-12-07 Plexxikon, Inc. Compounds modulating c-kit and c-fms activity and uses therefor
CA2609126A1 (en) 2005-05-20 2006-11-30 Vertex Pharmaceuticals Incorporated Pyrrolopyridines useful as inhibitors of protein kinase
PE20070100A1 (es) 2005-06-22 2007-03-10 Plexxikon Inc DERIVADOS DE PIRROLO[2,3-b]PIRIDINA COMO MODULADORES DE QUINASAS
GB0516156D0 (en) 2005-08-05 2005-09-14 Eisai London Res Lab Ltd JNK inhibitors
WO2008079909A1 (en) * 2006-12-21 2008-07-03 Plexxikon, Inc. Pyrrolo [2,3-b] pyridines as kinase modulators
PE20121126A1 (es) * 2006-12-21 2012-08-24 Plexxikon Inc Compuestos pirrolo [2,3-b] piridinas como moduladores de quinasa
TW200908968A (en) * 2007-05-29 2009-03-01 Sgx Pharmaceuticals Inc Substituted pyrrolopyridines and pyrazolopyridines as kinase modulators

Patent Citations (51)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6054270A (en) 1988-05-03 2000-04-25 Oxford Gene Technology Limited Analying polynucleotide sequences
US5700637A (en) 1988-05-03 1997-12-23 Isis Innovation Limited Apparatus and method for analyzing polynucleotide sequences and method of generating oligonucleotide arrays
US5658775A (en) 1988-05-17 1997-08-19 Sloan-Kettering Institute For Cancer Research Double copy retroviral vector
US5449614A (en) 1988-09-01 1995-09-12 Whitehead Institue For Biomedical Research Recombinant retroviruses with amphotropic and ecotropic host ranges
US5580859A (en) 1989-03-21 1996-12-03 Vical Incorporated Delivery of exogenous DNA sequences in a mammal
US5770456A (en) 1989-06-07 1998-06-23 Affymetrix, Inc. Cyclic nucleic acid and polypeptide arrays
US6261776B1 (en) 1989-06-07 2001-07-17 Affymetrix, Inc. Nucleic acid arrays
US5744305A (en) 1989-06-07 1998-04-28 Affymetrix, Inc. Arrays of materials attached to a substrate
US5143854A (en) 1989-06-07 1992-09-01 Affymax Technologies N.V. Large scale photolithographic solid phase synthesis of polypeptides and receptor binding screening thereof
US5800992A (en) 1989-06-07 1998-09-01 Fodor; Stephen P.A. Method of detecting nucleic acids
US5474935A (en) 1990-05-23 1995-12-12 The United States Of America As Represented By The Department Of Health And Human Services Adeno-associated virus (AAV)-based eucaryotic vectors
US6110458A (en) 1991-07-26 2000-08-29 University Of Rochester Cancer therapy utilizing ex vivo gene transduction
US5434049A (en) 1992-02-28 1995-07-18 Hitachi, Ltd. Separation of polynucleotides using supports having a plurality of electrode-containing cells
US5631236A (en) 1993-08-26 1997-05-20 Baylor College Of Medicine Gene therapy for solid tumors, using a DNA sequence encoding HSV-Tk or VZV-Tk
US5426039A (en) 1993-09-08 1995-06-20 Bio-Rad Laboratories, Inc. Direct molecular cloning of primer extended DNA containing an alkane diol
US6045996A (en) 1993-10-26 2000-04-04 Affymetrix, Inc. Hybridization assays on oligonucleotide arrays
US20010014449A1 (en) 1993-11-01 2001-08-16 Michael I. Nerenberg Methods for determination of single nucleic acid polymorphisms using bioelectronic microchip
US5632957A (en) 1993-11-01 1997-05-27 Nanogen Molecular biological diagnostic systems including electrodes
US5807522A (en) 1994-06-17 1998-09-15 The Board Of Trustees Of The Leland Stanford Junior University Methods for fabricating microarrays of biological samples
US5556752A (en) 1994-10-24 1996-09-17 Affymetrix, Inc. Surface-bound, unimolecular, double-stranded DNA
US5830645A (en) 1994-12-09 1998-11-03 The Regents Of The University Of California Comparative fluorescence hybridization to nucleic acid arrays
WO1996017958A1 (en) 1994-12-09 1996-06-13 The Regents Of The University Of California Comparative fluorescence hybridization to nucleic acid arrays
WO1996018738A2 (en) 1994-12-15 1996-06-20 Sugen, Inc. PROBIN TYROSINE KINASE (PYK2) ITS cDNA CLONING AND ITS USES
US6117681A (en) 1995-03-29 2000-09-12 Bavarian Nordic Research Inst. A/S Pseudotyped retroviral particles
US6110456A (en) 1995-06-07 2000-08-29 Yale University Oral delivery or adeno-associated viral vectors
US5856174A (en) 1995-06-29 1999-01-05 Affymetrix, Inc. Integrated nucleic acid diagnostic device
US6107478A (en) 1995-09-08 2000-08-22 Pedersen; Finn Skou Retroviral vector, a replication system for said vector and avian or mammalian cells transfected with said vector
US5721118A (en) 1995-10-31 1998-02-24 The Regents Of The University Of California, San Diego Mammalian artificial chromosomes and methods of using same
US6022963A (en) 1995-12-15 2000-02-08 Affymetrix, Inc. Synthesis of oligonucleotide arrays using photocleavable protecting groups
US6013440A (en) 1996-03-11 2000-01-11 Affymetrix, Inc. Nucleic acid affinity columns
US6025155A (en) 1996-04-10 2000-02-15 Chromos Molecular Systems, Inc. Artificial chromosomes, uses thereof and methods for preparing artificial chromosomes
US5959098A (en) 1996-04-17 1999-09-28 Affymetrix, Inc. Substrate preparation process
WO1997046313A1 (en) 1996-06-07 1997-12-11 Eos Biotechnology, Inc. Immobilised linear oligonucleotide arrays
US5965452A (en) 1996-07-09 1999-10-12 Nanogen, Inc. Multiplexed active biologic array
US6258606B1 (en) 1996-07-09 2001-07-10 Nanogen, Inc. Multiplexed active biologic array
US6096718A (en) 1997-06-05 2000-08-01 Gene Targeting Corp. Tissue specific adenovirus vectors for breast cancer treatment
US20010018642A1 (en) 1997-07-25 2001-08-30 David Balaban Method and system for providing a probe array chip design database
WO1999009217A1 (en) 1997-08-15 1999-02-25 Hyseq, Inc. Methods and compositions for detection or quantification of nucleic acid species
US20010019827A1 (en) 1997-09-11 2001-09-06 Dawson Elliott P. Method of making high density arrays
US20010014448A1 (en) 1997-09-30 2001-08-16 Surmodics, Inc. Target molecule attachment to surfaces
WO1999051773A1 (en) 1998-04-03 1999-10-14 Phylos, Inc. Addressable protein arrays
US6048695A (en) 1998-05-04 2000-04-11 Baylor College Of Medicine Chemically modified nucleic acids and methods for coupling nucleic acids to solid support
US6113913A (en) 1998-06-26 2000-09-05 Genvec, Inc. Recombinant adenovirus
US6277628B1 (en) 1998-10-02 2001-08-21 Incyte Genomics, Inc. Linear microarrays
US6277489B1 (en) 1998-12-04 2001-08-21 The Regents Of The University Of California Support for high performance affinity chromatography and other uses
US20010016322A1 (en) 1999-04-27 2001-08-23 Caren Michael P. Method of performing array-based hybridization assays using thermal inkjet deposition of sample fluids
US20010012537A1 (en) 1999-07-30 2001-08-09 Anderson Norman G. Dry deposition of materials for microarrays using matrix displacement
US20010008765A1 (en) 1999-12-06 2001-07-19 Fuji Photo Film Co., Ltd. DNA chip and reactive solid carrier
US20040002534A1 (en) 1999-12-22 2004-01-01 Sugen, Inc. Methods of modulating c-kit tyrosine protein kinase function with indolinone compounds
WO2004016610A1 (en) 2002-08-14 2004-02-26 Astrazeneca Ab Substituted pyrrolopyridines
US20040077595A1 (en) 2002-09-06 2004-04-22 Insert Therapeutics, Inc. Cyclodextrin-based polymers for therapeutics delivery

Non-Patent Citations (21)

* Cited by examiner, † Cited by third party
Title
BOWTELL, NATURE GENETICS SUPP., vol. 21, 1999, pages 25 - 32
BUCHSCHER, J. VIROL., vol. 66, 1992, pages 2731 - 2739
CAPON, NATURE, vol. 377, 1989, pages 525 - 531
COLLIUOD, BIOCONJUGATE CHEM., vol. 4, 1993, pages 528 - 536
DOBELI, PROTEIN EXPR. PURIF., vol. 12, 1998, pages 404 - 414
ECKSTEIN ET AL., NUCL. ACIDS RES., vol. 13, 1985, pages 8764 - 8785
FENG, NATURE BIOTECHNOLOGY, vol. 15, 1997, pages 866 - 870
IWANE, BIOPHYS. BIOCHEM. RES. COMM., vol. 230, 1997, pages 76 - 80
JOHANN, J. VIROL., vol. 66, 1992, pages 1635 - 1640
JOHNSTON, CURR. BIOL., vol. 8, 1998, pages R171 - R174
KERN, BIOTECHNIQUES, vol. 23, 1997, pages 120 - 124
KROLL, DNA CELL. BIOL., vol. 12, 1993, pages 441 - 53
KUNKEL, PROC. NATL. ACAD. SCI., vol. 82, 1985, pages 488 - 492
LU, ANAL. CHEM., vol. 67, 1995, pages 83 - 87
NG, LANGMUIR, vol. 11, 1995, pages 4048 - 55
OKADA, GENE THER., vol. 3, 1996, pages 957 - 964
SCHUHMANN, ADV. MATER., vol. 3, 1991, pages 388 - 391
SCHUMMER, BIOTECHNIQUES, vol. 23, 1997, pages 1087 - 1092
SIGAL, ANAL. CHEM., vol. 68, 1996, pages 490 - 497
SOLINAS-TOLDO, GENES, CHROMOSOMES & CANCER, vol. 20, 1997, pages 399 - 407
WILLIAMS, BIOCHEMISTRY, vol. 34, 1995, pages 1787 - 1797

Cited By (96)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2009523812A (ja) * 2006-01-19 2009-06-25 オーエスアイ・ファーマスーティカルズ・インコーポレーテッド 融合へテロ二環式キナーゼ阻害剤
WO2008064255A3 (en) * 2006-11-22 2008-08-14 Plexxikon Inc Compounds modulating c-fms and/or c-kit activity and uses therefor
WO2008064265A3 (en) * 2006-11-22 2008-11-13 Plexxikon Inc Compounds modulating c-fms and/or c-kit activity and uses therefor
NO342175B1 (no) * 2006-11-22 2018-04-09 Plexxikon Inc Kjemiske forbindelser, preparater omfattende slike forbindelser, slike forbindelser og preparater for behandling av sykdom samt sett omfattende slike forbindelser
WO2008064265A2 (en) * 2006-11-22 2008-05-29 Plexxikon, Inc. Compounds modulating c-fms and/or c-kit activity and uses therefor
JP2018021055A (ja) * 2006-11-22 2018-02-08 プレキシコン インコーポレーテッドPlexxikon Inc. C−fmsおよび/またはc−kit活性を調節する化合物およびその用途
WO2008080015A3 (en) * 2006-12-21 2008-10-02 Plexxikon Inc Compounds and methods for kinase modulation, and indications therefor
WO2008080001A3 (en) * 2006-12-21 2009-01-08 Plexxikon Inc Compounds and methods for kinase modulation, and indications therefor
WO2008080015A2 (en) * 2006-12-21 2008-07-03 Plexxikon, Inc. Compounds and methods for kinase modulation, and indications therefor
WO2008080001A2 (en) * 2006-12-21 2008-07-03 Plexxikon, Inc. Compounds and methods for kinase modulation, and indications therefor
WO2008079909A1 (en) * 2006-12-21 2008-07-03 Plexxikon, Inc. Pyrrolo [2,3-b] pyridines as kinase modulators
US9844539B2 (en) 2007-07-17 2017-12-19 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US10426760B2 (en) 2007-07-17 2019-10-01 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US8173635B2 (en) 2007-11-02 2012-05-08 Vertex Pharmaceuticals Incorporated Kinase inhibitors
US8367697B2 (en) 2007-11-02 2013-02-05 Vertex Pharmaceuticals Incorporated Kinase inhibitors
WO2009111280A1 (en) 2008-02-29 2009-09-11 Array Biopharma Inc. N- (6-aminopyridin-3-yl) -3- (sulfonamido) benzamide derivatives as b-raf inhibitors for the treatment of cancer
US8394795B2 (en) 2008-02-29 2013-03-12 Array Biopharma Inc. Pyrazole [3, 4-B] pyridine Raf inhibitors
US8338452B2 (en) 2008-02-29 2012-12-25 Array Biopharma Inc. Raf inhibitor compounds and methods of use thereof
WO2009143024A3 (en) * 2008-05-19 2010-01-07 Plexxikon, Inc. Compounds and methods for kinase modulation, and indications therefor
WO2009143018A2 (en) * 2008-05-19 2009-11-26 Plexxikon, Inc. Compounds and methods for kinase modulation, and indications therefor
US8153641B2 (en) 2008-05-19 2012-04-10 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US8158636B2 (en) 2008-05-19 2012-04-17 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9260437B2 (en) 2008-05-19 2016-02-16 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
WO2009143018A3 (en) * 2008-05-19 2010-04-15 Plexxikon, Inc. Pyrrolo [2, 3-d] pyrimidines as fm and kit kinase modulators
US8912204B2 (en) 2008-05-19 2014-12-16 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
WO2009143024A2 (en) * 2008-05-19 2009-11-26 Plexxikon, Inc. Compounds and methods for kinase modulation, and indications therefor
US8110576B2 (en) 2008-06-10 2012-02-07 Plexxikon Inc. Substituted pyrrolo[2,3b]pyrazines and methods for treatment of raf protein kinase-mediated indications
WO2009152087A1 (en) * 2008-06-10 2009-12-17 Plexxikon, Inc. Bicyclic heteroaryl compounds and methods for kinase modulation, and indications therefor
EP2313411A1 (en) * 2008-06-10 2011-04-27 Plexxikon, Inc. 5h-pyrr0l0 [2,3-b]pyrazine derivatives for kinase modulation, and indications therefor
JP2011522894A (ja) * 2008-06-10 2011-08-04 プレキシコン,インコーポレーテッド キナーゼを調節する5h−ピロロ[2,3−b]ピラジン誘導体、およびその適応症
WO2010111527A1 (en) * 2009-03-26 2010-09-30 Plexxikon, Inc. Pyrazolo [ 3, 4 -b] pyridines as kinase inhibitors and their medical use
US9663517B2 (en) 2009-04-03 2017-05-30 Plexxikon Inc. Compositions and uses thereof
US8741920B2 (en) 2009-08-03 2014-06-03 Hoffmann-La Roche, Inc. Process for the manufacture of pharmaceutically active compounds
US8648086B2 (en) 2009-08-24 2014-02-11 Ascepion Pharmaceuticals, Inc. 5,6-bicyclic heteroaryl-containing urea compounds as kinase inhibitors
WO2011025968A1 (en) 2009-08-28 2011-03-03 Array Biopharma Inc. 1h-pyrazolo [ 3, 4-b] pyridine compounds for inhibiting raf kinase
US9617267B2 (en) 2009-11-18 2017-04-11 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9440969B2 (en) 2009-12-23 2016-09-13 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US8580803B2 (en) 2009-12-30 2013-11-12 Arqule, Inc. Substituted pyrrolo-aminopyrimidine compounds
US8901118B2 (en) 2010-04-21 2014-12-02 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
AU2011242778B2 (en) * 2010-04-21 2015-09-03 Opna Immuno Oncology, SA Compounds and methods for kinase modulation, and indications therefor
KR101799126B1 (ko) 2010-04-21 2017-11-17 플렉시콘 인코퍼레이티드 키나아제 조절을 위한 화합물 및 방법, 및 그에 대한 적응증
US9682981B2 (en) 2010-04-21 2017-06-20 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
EP2560972A4 (en) * 2010-04-21 2013-11-20 Plexxikon Inc COMPOSITIONS AND METHODS FOR MODULATING KINASES AND THEIR INDICATIONS
EP2560972A2 (en) * 2010-04-21 2013-02-27 Plexxikon, Inc. Compounds and methods for kinase modulation, and indications therefor
US9624213B2 (en) 2011-02-07 2017-04-18 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US11337976B2 (en) 2011-02-07 2022-05-24 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9550768B2 (en) 2011-05-17 2017-01-24 Plexxikon Inc. Kinase modulation and indications therefor
EP3124472A1 (en) 2011-12-31 2017-02-01 BeiGene, Ltd. Fused tricyclic compounds as raf kinase inhibitors
US10123998B2 (en) 2012-03-19 2018-11-13 Plexxikon Inc. Kinase modulation, and indications therefor
US9358235B2 (en) 2012-03-19 2016-06-07 Plexxikon Inc. Kinase modulation, and indications therefor
US9730918B2 (en) 2012-03-19 2017-08-15 Plexxikon Inc. Kinase modulation, and indications therefor
US9695169B2 (en) 2012-05-31 2017-07-04 Plexxikon Inc. Synthesis of heterocyclic compounds
EP3550031A1 (en) 2012-07-24 2019-10-09 Pharmacyclics, LLC Mutations associated with resistance to inhibitors of bruton's tyrosine kinase (btk)
US10227357B2 (en) 2012-09-06 2019-03-12 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9163021B2 (en) 2012-10-04 2015-10-20 Pfizer Limited Pyrrolo[3,2-c]pyridine tropomyosin-related kinase inhibitors
US10301280B2 (en) 2012-12-21 2019-05-28 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9676748B2 (en) 2012-12-21 2017-06-13 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9718847B2 (en) 2013-03-15 2017-08-01 Plexxikon Inc. Heterocyclic compounds and uses thereof
US9822109B2 (en) 2013-03-15 2017-11-21 Plexxikon Inc. Heterocyclic compounds and uses thereof
US9873700B2 (en) 2013-05-30 2018-01-23 Plexxikon Inc. Compounds for kinase modulation, and indications therefor
US10421761B2 (en) 2013-05-30 2019-09-24 Plexxikon Inc. Compounds for kinase modulation, and indications therefor
WO2014206344A1 (en) 2013-06-28 2014-12-31 Beigene, Ltd. Fused tricyclic amide compounds as multiple kinase inhibitors
US9771369B2 (en) 2014-03-04 2017-09-26 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9856259B2 (en) 2014-09-15 2018-01-02 Plexxikon Inc. Heterocyclic compounds and uses thereof
US10160755B2 (en) 2015-04-08 2018-12-25 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US11274108B2 (en) 2015-07-20 2022-03-15 Genzyme Corporation Colony stimulating factor-1 receptor (CSF-1R) inhibitors
US10829484B2 (en) 2015-07-28 2020-11-10 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9975894B2 (en) 2015-09-21 2018-05-22 Plexxikon Inc. Heterocyclic compounds and uses thereof
US10370374B2 (en) 2015-09-21 2019-08-06 Plexxikon Inc. Heterocyclic compounds and uses thereof
US10899761B2 (en) 2015-09-21 2021-01-26 Plexxikon Inc. Heterocyclic compounds and uses thereof
US10647716B2 (en) 2015-09-21 2020-05-12 Plexxikon Inc. Heterocyclic compounds and uses thereof
US9771363B2 (en) 2015-09-21 2017-09-26 Plexxikon Inc. Heterocyclic compounds and uses thereof
US9938273B2 (en) 2015-12-07 2018-04-10 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US10933065B2 (en) 2015-12-23 2021-03-02 Arqule Inc. Tetrahydropyranyl amino-pyrrolopyrimidinone and methods of use thereof
US9630968B1 (en) 2015-12-23 2017-04-25 Arqule, Inc. Tetrahydropyranyl amino-pyrrolopyrimidinone and methods of use thereof
US11020400B2 (en) 2015-12-23 2021-06-01 Arqule, Inc. Tetrahydropyranyl amino-pyrrolopyrimidinone and methods of use thereof
US10245263B2 (en) 2015-12-23 2019-04-02 Arqule, Inc. Tetrahydropyranyl amino-pyrrolopyrimidinone and methods of use thereof
US10160747B2 (en) 2016-03-16 2018-12-25 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US11020398B2 (en) 2016-08-24 2021-06-01 Arqule, Inc. Amino-pyrrolopyrimidinone compounds and methods of use thereof
US10508085B2 (en) 2016-09-22 2019-12-17 Plexxikon Inc. Compounds and methods for IDO and TDO modulation, and indications therefor
US10703757B2 (en) 2016-12-23 2020-07-07 Plexxikon Inc. Compounds and methods for CDK8 modulation and indications therefor
US10577366B2 (en) 2017-03-20 2020-03-03 Plexxikon Inc. Crystalline forms of a compound that inhibits bromodomain
US10428067B2 (en) 2017-06-07 2019-10-01 Plexxikon Inc. Compounds and methods for kinase modulation
US10941142B2 (en) 2017-07-25 2021-03-09 Plexxikon Inc. Formulations of a compound modulating kinases
US10961240B2 (en) 2017-07-25 2021-03-30 Plexxikon Inc. Formulations of a compound modulating kinases
US10435404B2 (en) 2017-07-25 2019-10-08 Plexxikon Inc. Formulations of a compound modulating kinases
US10717735B2 (en) 2017-10-13 2020-07-21 Plexxikon Inc. Solid forms of a compound for modulating kinases
US11103505B2 (en) 2017-10-27 2021-08-31 Plexxikon Inc. Formulations of a compound modulating kinases
US11858927B2 (en) 2018-01-31 2024-01-02 Heparegenix Gmbh Protein kinase MKK4 inhibitors for promoting liver regeneration or reducing or preventing hepatocyte death
US11149011B2 (en) 2018-03-20 2021-10-19 Plexxikon Inc. Compounds and methods for IDO and TDO modulation, and indications therefor
US11731968B2 (en) 2018-06-21 2023-08-22 Heparegenix Gmbh Tricyclic protein kinase inhibitors for promoting liver regeneration or reducing or preventing hepatocyte death
US11912701B2 (en) 2018-07-16 2024-02-27 Heparegenix Gmbh Protein kinase inhibitors for promoting liver regeneration or reducing or preventing hepatocyte death
WO2020079581A1 (en) 2018-10-16 2020-04-23 Novartis Ag Tumor mutation burden alone or in combination with immune markers as biomarkers for predicting response to targeted therapy
WO2021144360A1 (en) 2020-01-17 2021-07-22 F. Hoffmann-La Roche Ag Small molecule csf-1r inhibitors in therapeutic and cosmetic uses
US11530216B2 (en) 2020-12-23 2022-12-20 Genzyme Corporation Deuterated colony stimulating factor-1 receptor (CSF-1R) inhibitors
WO2022195551A1 (en) 2021-03-18 2022-09-22 Novartis Ag Biomarkers for cancer and methods of use thereof

Also Published As

Publication number Publication date
BRPI0610066A2 (pt) 2010-05-25
JP2008545652A (ja) 2008-12-18
US20110166174A1 (en) 2011-07-07
AU2006272951A1 (en) 2007-02-01
CR9591A (es) 2008-03-26
KR20080027775A (ko) 2008-03-28
US20070032519A1 (en) 2007-02-08
WO2007013896A3 (en) 2007-05-31
ECSP077985A (es) 2008-01-23
MX2007014377A (es) 2008-02-06
US7846941B2 (en) 2010-12-07
NZ563444A (en) 2011-04-29
IL187344A0 (en) 2008-04-13
NO20075992L (no) 2008-02-13
EP1885723A2 (en) 2008-02-13
CA2608733A1 (en) 2007-02-01

Similar Documents

Publication Publication Date Title
US7846941B2 (en) Compounds modulating c-kit and c-fms activity and uses therefor
US9487515B2 (en) Compounds modulating c-fms and/or c-kit activity and uses therefor
US10426760B2 (en) Compounds and methods for kinase modulation, and indications therefor
US10160747B2 (en) Compounds and methods for kinase modulation, and indications therefor
US7872018B2 (en) Compounds and methods for kinase modulation, and indications therefor
JP6113151B2 (ja) キナーゼ調節およびその適応症

Legal Events

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

Ref document number: 200680026005.5

Country of ref document: CN

WWE Wipo information: entry into national phase

Ref document number: 187344

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 4363/KOLNP/2007

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 12007502546

Country of ref document: PH

Ref document number: 563444

Country of ref document: NZ

ENP Entry into the national phase

Ref document number: 2608733

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: MX/a/2007/014377

Country of ref document: MX

Ref document number: 07121351

Country of ref document: CO

ENP Entry into the national phase

Ref document number: 2008512402

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2006272951

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2006813186

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: CR2007-009591

Country of ref document: CR

WWE Wipo information: entry into national phase

Ref document number: 2007142326

Country of ref document: RU

Ref document number: 1020077029428

Country of ref document: KR

ENP Entry into the national phase

Ref document number: 2006272951

Country of ref document: AU

Date of ref document: 20060516

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: PI0610066

Country of ref document: BR

Kind code of ref document: A2