WO2010036380A1 - Heterocyclic kinase inhibitors - Google Patents

Heterocyclic kinase inhibitors Download PDF

Info

Publication number
WO2010036380A1
WO2010036380A1 PCT/US2009/005380 US2009005380W WO2010036380A1 WO 2010036380 A1 WO2010036380 A1 WO 2010036380A1 US 2009005380 W US2009005380 W US 2009005380W WO 2010036380 A1 WO2010036380 A1 WO 2010036380A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
aryl
cycloalkyl
heteroaryl
heteroalkyl
Prior art date
Application number
PCT/US2009/005380
Other languages
French (fr)
Inventor
Pingda Ren
Yi Liu
Liansheng Li
Katrina Chan
Troy Edward Wilson
Original Assignee
Intellikine, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Intellikine, Inc. filed Critical Intellikine, Inc.
Priority to JP2011529032A priority Critical patent/JP5731978B2/en
Priority to US13/121,157 priority patent/US8703778B2/en
Priority to CA2738429A priority patent/CA2738429C/en
Priority to EP09816603.6A priority patent/EP2346508B1/en
Publication of WO2010036380A1 publication Critical patent/WO2010036380A1/en
Priority to US14/222,500 priority patent/US9296742B2/en
Priority to US15/050,029 priority patent/US9790228B2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • A61K31/52Purines, e.g. adenine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • 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
    • 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
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D473/00Heterocyclic compounds containing purine ring systems
    • C07D473/26Heterocyclic compounds containing purine ring systems with an oxygen, sulphur, or nitrogen atom directly attached in position 2 or 6, but not in both
    • C07D473/32Nitrogen atom
    • C07D473/34Nitrogen atom attached in position 6, e.g. adenine
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the activity of cells can be regulated by external signals that stimulate or inhibit intracellular events.
  • the process by which stimulatory or inhibitory signals are transmitted into and within a cell to elicit an intracellular response is referred to as signal transduction.
  • cascades of signal transduction events have been elucidated and found to play a central role in a variety of biological responses. Defects in various components of signal transduction pathways have been found to account for a vast number of diseases, including numerous forms of cancer, inflammatory disorders, metabolic disorders, vascular and neuronal diseases (Gaestel et al. Current Medicinal Chemistry (2007) 14:2214-2234).
  • Kinases represent a class of important signaling molecules. Kinases can generally be classified into protein kinases and lipid kinases, and certain kinases exhibit dual specificities. Protein kinases are enzymes that phosphorylate other proteins and/or themselves (i.e., autophosphorylation).
  • Protein kinases can be generally classified into three major groups based upon their substrate utilization: tyrosine kinases which predominantly phosphorylate substrates on tyrosine residues (e.g., erb2, PDGF receptor, EGF receptor, VEGF receptor, src, abl), serine/threonine kinases which predominantly phosphorylate substrates on serine and/or threonine residues (e.g., mTorCl, mTorC2, ATM, ATR, DNA-PK, Akt), and dual-specificity kinases which phosphorylate substrates on tyrosine, serine and/or threonine residues.
  • tyrosine kinases which predominantly phosphorylate substrates on tyrosine residues (e.g., erb2, PDGF receptor, EGF receptor, VEGF receptor, src, abl), serine/threonine kinases which predominantly phosphorylate substrates
  • Lipid kinases are enzymes that catalyze the phosphorylation of lipids. These enzymes, and the resulting phosphorylated lipids and lipid-derived biologically active organic molecules, play a role in many different physiological processes, including cell proliferation, migration, adhesion, and differentiation. Certain lipid kinases are membrane associated and they catalyze the phosphorylation of lipids contained in or associated with cell membranes. Examples of such enzymes include phosphoinositide(s) kinases (such as PI3-kinases, PI4- Kinases), diacylglycerol kinases, and sphingosine kinases.
  • PDKs phosphoinositide 3-kinases
  • PI3K signaling pathway is one of the most highly mutated systems in human cancers.
  • PI3K signaling is also a key factor in many other diseases in humans.
  • PI3K signaling is involved in many disease states including allergic contact dermatitis, rheumatoid arthritis, osteoarthritis, inflammatory bowel diseases, chronic obstructive pulmonary disorder, psoriasis, multiple sclerosis, asthma, disorders related to diabetic complications, and inflammatory complications of the cardiovascular system such as acute coronary syndrome.
  • PBKs are members of a unique and conserved family of intracellular lipid kinases that phosphorylate the 3 '-OH group on phosphatidylinositols or phosphoinositides.
  • the PI3K family comprises 15 kinases with distinct substrate specificities, expression patterns, and modes of regulation (Katso et al., 2001).
  • the class I PDKs (pi 10a, pi lO ⁇ , pi lO ⁇ , and pi 10 ⁇ ) are typically activated by tyrosine kinases or G-protein coupled receptors to generate PIP3, which engages downstream effectors such as those in the Akt/PDKl pathway, mTOR, the Tec family kinases, and the Rho family GTPases.
  • the class II and III PD-Ks play a key role in intracellular trafficking through the synthesis of PI(3)P and PI(3,4)P2.
  • the PIKKs are protein kinases that control cell growth (mTORCl) or monitor genomic integrity (ATM, ATR, DNA-PK, and hSmg-1).
  • the delta ( ⁇ ) isoform of class I PI3K has been implicated, in particular, in a number of diseases and biological processes.
  • PDK ⁇ is expressed primarily in hematopoietic cells including leukocytes such as T-cells, dendritic cells, neutrophils, mast cells, B-cells, and macrophages.
  • PDK ⁇ is integrally involved in mammalian immune system functions such as T-cell function, B-cell activation, mast cell activation, dendritic cell function, and neutrophil activity.
  • PDK ⁇ Due to its integral role in immune system function, PDK ⁇ is also involved in a number of diseases related to undesirable immune response such as allergic reactions, inflammatory diseases, inflammation mediated angiogenesis, rheumatoid arthritis, auto-immune diseases such as lupus, asthma, emphysema and other respiratory diseases.
  • Other class I PDK involved in immune system function includes PDK ⁇ , which plays a role in leukocyte signaling and has been implicated in inflammation, rheumatoid arthritis, and autoimmune diseases such as lupus.
  • Downstream mediators of the PDK signal transduction pathway include Akt and mammalian target of rapamycin (mTOR).
  • Akt possesses a plckstrin homology (PH) domain that binds PIP3, leading to Akt kinase activation.
  • Akt phosphorylates many substrates and is a central downstream effector of PDK for diverse cellular responses.
  • One important function of Akt is to augment the activity of mTOR, through phosphorylation of TSC2 and other mechanisms.
  • mTOR is a serine-threonine kinase related to the lipid kinases of the PDK family.
  • mTOR has been implicated in a wide range of biological processes including cell growth, cell proliferation, cell motility and survival. Disregulation of the mTOR pathway has been reported in various types of cancer.
  • mTOR is a multifunctional kinase that integrates growth factor and nutrient signals to regulate protein translation, nutrient uptake, autophagy, and mitochondria
  • kinases particularly PDKs are prime targets for drug development.
  • PDK inhibitors suitable for drug development.
  • the present invention addresses this need and provides related advantages as well by providing new classes of kinase inhibitors.
  • the present invention provides a compound having a structure of one of the following formulae:
  • X 5 and X 6 are C-R 6 , N, C- L 1 -R 1 , or N- L'-R 1 wherein one of X 5 and X 6 is C- L'-R 1 or N- L'-R 1 ; X, is C or N; and X 2 and X 8 are independently N or C-R 6 ; X 3 and X 7 are C or N, and at least one of X 3 and X 7 is C; [0013] X 4 in Formula I-A or I-C is C or N; and X 4 in Formula I-B or I-D is C-R 6 , NH, or N; [0014] and wherein no more than two adjacent ring atoms are N or NH; [0015] R 1 is hydrogen, halogen, -CN, -OR 10 , -S(O) n R 11 , -NR 12 R 13 , -C(O)R 14 , alkyl, alkenyl, alkyn
  • each R 11 is independently -NR 16 R 17 , alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
  • each R 12 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
  • each R 13 is independently hydrogen, -S(O) n R 18 , -C(O)R 19 , alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; [0029] or R 12 and R 13 are taken together to form a 5, 6, 7, or 8 membered ring, wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom Of -NR 12 R 13 ; and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted; [
  • each R 18 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
  • each R 19 is independently -NR 24 R 25 , hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
  • each of R 20 and R 21 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; [0036] or R 20 and R 21 are taken together to form a 5, 6, 7, or 8 membered ring, wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom Of-NR 20 R 21 ; and further wherein the 5, 6, 7, or 8
  • each R 24 and R 25 are independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, or R 24 and R 25 are taken together to form a 5, 6, 7, or 8 membered ring, wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom
  • W is CR 6 or N.
  • the compound of Formula I- A or I-C is a compound having a structure of one of the following formulae:
  • the compound of Formula I-E is a compound of Formula I-J or Formula I- J- 1 :
  • the compound of Formula I-A is a compound having a structure of one of the following formulae:
  • the compound of Formula I-B or I-D having a structure of one of the following formulae:
  • the invention provides for a compound having is a compound having a structure of the formula
  • X 5 and X 6 are C-R 6 , N, C- L 1 -R 1 , or N- L 1 -R 1 wherein one and no more than one of X 5 and X 6 is C- L 1 -R 1 or N-
  • L'-R 1 ; XI is C or N; and X 2 and X 8 are independently N, or C-R 6 ; X 3 and X 7 are C or N, at least one of X 3 and
  • X 7 is C
  • X 4 in Formula V-A and Formula V-C is C or N; and X 4 in Formula V-B and Formula V-D is C-R 6 , NH, or N; [0048] and wherein no more than two adjacent ring atoms are N or NH; [0049] R 1 is hydrogen, halogen, -CN, -OR 10 , -S(O) n R 11 , -NR 12 R 13 , -C(O)R 14 , alkyl, alkenyl, alkynyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, and n is an integer from 0 to 2; [0050] L 1 is a bond, alkylene, heteroalkylene, alkenylene, alkynylene, cycloalkylene, heterocycloalkylene, arylene, or heteroarylene; [0051] each of R 2 and R 3 is independently hydrogen, halogen, -
  • each of R 7 and R 8 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocyclyalkyl, or R 7 and R 8 taken together form a cycloalkyl, heterocycloalkyl or aryl ring;
  • R 9 is hydrogen, -C(O)R 14 , alkyl, alkenyl, alkynyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
  • each R 10 is independently hydrogen, -C(O)R 15 , alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
  • each R 11 is independently -NR 16 R 17 , alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
  • each R 12 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
  • each R 13 is independently hydrogen, -S(O) n R 18 , -C(O)R 19 , alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
  • R 12 and R 13 are taken together to form a 5, 6, 7, or 8 membered ring, wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom Of-NR 12 R 13 , and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted;
  • each R 14 is independently -NR 20 R 21 , hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
  • each R 15 is independently -NR 22 R 23 , hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
  • each of R 16 and R 17 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; or R 16 and R 17 are taken together to form a 5, 6, 7 or 8 membered ring, wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom Of-NR 16 R 17 , and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted;
  • each R 18 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
  • each R 19 is independently -NR 24 R 25 , hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
  • each of R 20 and R 21 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, or R 20 and R 21 are taken together to form a 5, 6, 7, or 8 membered ring, wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom Of-NR 20 R 21 , and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted;
  • each of R 22 and R 23 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, or R 22 and R 23 are taken together to form a 5, 6, 7, or 8 membered ring, wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom Of-NR 22 R 23 , and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted; and
  • each of R 24 and R 25 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, or R 24 and R 25 are taken together to form a 5, 6, 7, or 8 membered ring wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom Of-NR 24 R 25 , and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted.
  • the compound of Formula V-A is a compound having a structure of Formula VI-Al : [0073] In another aspect, the invention provides for a compound having a structure of one of the following formulae
  • X 5 and X 6 are C-R 6 , N, C- L 1 -R 1 , or N- L 1 -R 1 wherein one and no more than one of X 5 and X 6 is C- L 1 -R 1 or N-
  • Xi is C or N; and X 2 and X 8 are independently N, or C-R 6 ; [0077] X 3 and X 7 are C or N, at least one of X 3 and X 7 is C; [0078] X 4 in Formula VII-A and Formula VII-C is C or N; and X 4 in Formula VII-B and Formula VII-D is C-R 6 , NH, or N; and wherein no more than two adjacent ring atoms are N or NH; [0079] R 1 is hydrogen, halogen, -CN, -OR 10 , -S(O) n R 11 , -NR 12 R 13 , -C(O)R 14 , alkyl, alkenyl, alkynyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, and n is an integer from 0 to 2; [0080] L 1 is a bond, alkylene, heteroalky
  • R 9 is hydrogen, -C(O)R 14 , alkyl, alkenyl, alkynyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; [0089] each R 10 is independently hydrogen, -C(O)R 15 , alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
  • each R 11 is independently -NR 16 R 17 , alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
  • each R 12 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
  • each R 13 is independently hydrogen, -S(O) n R 18 , -C(O)R 19 , alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
  • R 12 and R 13 are taken together to form a 5, 6, 7, or 8 membered ring, wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom Of -NR 12 R 13 , and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted;
  • each R 14 is independently -NR 20 R 21 , hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
  • each R 15 is independently -NR 22 R 23 , hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
  • each of R 16 and R 17 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; or R 16 and R 17 are taken together to form a 5, 6, 7, or 8 membered ring, wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 additional ring heteroatoms selected from N, S, or O in addition to the nitrogen atom Of-NR 16 R 17 , and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted;
  • each R 18 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
  • each R 19 is independently -NR 24 R 25 , hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
  • each of R 20 and R 21 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, or R 20 and R 21 are taken together to form a 5, 6, 7, or 8 membered ring, wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 additional ring heteroatoms selected from N, S, or O in addition to the nitrogen atom Of-NR 20 R 21 , and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted;
  • each of R 22 and R 23 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, or R 22 and R 23 are taken together to form a 5, 6, 7, or 8 membered ring, wherein the 5, 6, 7, or 8 membered ring contains 0, 1 , 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom Of-NR 22 R 23 , and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted; and
  • each of R 24 and R 25 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, or R 24 and R 25 are taken together to form a 5, 6, 7, or 8 membered ring wherein the 5, 6, 7, or 8 membered ring contains 0, 1 , 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom Of-NR 24 R 25 , wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 additional ring heteroatoms selected from N, S, or O, and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted.
  • compound of Formula VII-A is a compound having a structure of Formula VII-Al :
  • R 9 is -OR 10 , -NR 12 R 13 , -C(O)R 14 , alkyl, alkenyl, alkynyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl.
  • R 9 when R 9 is aryl, then R 9 is substituted with halogen, alkyl or heteroalkyl. In other embodiments, wherein when R 9 is -NR 12 R 13 , then R 12 and R 13 are taken together to form a 5, 6, 7 or 8 membered ring.
  • R 1 is bicyclic heteroaryl.
  • R 2 is -NH 2 .
  • a compound of Formula I-A, I-B, I-C, I-D, I-E, I-F, I-G, I-H, I-J, I-J-l, I-K, II-A, II- Al, II- A2, III-A, IV-A, V-A, V-Al, V-B, V-C, V-D, VI-A, VI-B, VI-C, VI-D, VII-A, VII-B, VII-C, or VII-D, R 7 and R 8 are hydrogen.
  • a compound of Formula I-A, I-B, I-C, I-D, I-E, I-F, I-G, I-H, I-J, I-J-l, I-K, II-A, II- Al, II- A2, III-A, IV-A, V-A, V-Al, V-B, V-C, V-D, VI-A, VI-Al, VI- A2, VI-B, VI-C, or VI-D, W is CH.
  • a compound of Formula I-A, I-B, I-C, I-D, I-E, I-F, I-G, I-H, I-J, I-J-l, I- K, II-A, II-A1, II-A2, III-A, IV-A, V-A, V-Al, V-B, V-C, V-D, VI-A, VI-Al, VI- A2, VI-B, VI-C, VI-D, VII-A, VII-Al, VII- A2, VII-B, VII-C, or VII-D is provided, wherein R 9 is halogen, -CN, -
  • OR 10 -S(O) n R", -NR 12 R 13 , -C(O)R 14 , alkyl, alkenyl, alkynyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, and wherein n is independently an integer from 0 to 2.
  • R 9 is aryl substituted with halogen, aryl substituted with alkyl, or aryl substituted with heteroalkyl,.
  • a compound of Formula I-A, I-B, I-C, I-D, I-E, I-F, I-G, I-H, I-J, I-J-l, I-K, H-A, II-A1, II-A2, III-A, IV-A, V-A, V-Al, V-B, V-C, V-D, VI- A, VI-Al, VI- A2, VI-B, VI-C, VI-D, VII-A, VII-Al, VII- A2, VII-B, VII-C, or VII-D is provided, wherein R 12 is -H and R 13 is unsubstituted alkyl or cycloalkyl.
  • R 12 and R 13 are unsubstituted alkyl or cycloalkyl.
  • a compound of Formula I-A, I-B, I-C, I-D, I-E, I-F, I-G, I-H, I-J, I-J-l, I- K, H-A, II-A1, II- A2, III-A, IV-A, V-A, V-Al, V-B, V-C, V-D, VI-A, VI-Al, VI-A2, VI-B, VI-C, VI-D, VII-A, VII-Al, VII- A2, VII-B, VII-C, or VII-D is provided, wherein R 1 is monocyclic aryl, bicyclic aryl, monocyclic heteroaryl, or bicyclic heteroaryl.
  • a compound of Formula I-A, I-B, I-C, I-D, I-E, I-F, I-G, I-H, I-J, I-J-l, I- K, H-A, II-A1, II- A2, III-A, IV-A, V-A, V-Al, V-B, V-C, V-D, VI-A, VI-Al, VI- A2, VI-B, VI-C, VI-D, VII-A, VII-Al, VII- A2, VII-B, VII-C, or VII-D is provided wherein R 4 and R 5 taken together form a 6-membered ring.
  • the 6-membered ring can be substituted with R 6 .
  • a compound of Formula I-A, I-B, I-C, I-D, I-E, I-F, I-G, I-H, I-J, I-J-l, I-K, H-A, II-A1, II- A2, IH-A, IV-A, V-A, V-Al, V-B, V-C, V-D, VI-A, VI-Al, VI- A2, VI-B, VI-C, VI-D, VII-A, VII-Al, VII-A2, VII-B, VII-C, or VII-D is provided wherein R 2 is -NR 12 R 13 .
  • a compound of Formula I-A, I-B, I-C, I-D, I-E, I-F, I-G, I-H, I-J, I-J-l, I-K, II-A, II-A1, II-A2, HI-A, IV-A, V-A, V-Al, V-B, V-C, V-D, VI-A, VI-Al, VI- A2, VI-B, VI-C, VI-D, VH-A, VII-Al, VII- A2, VII-B, VII-C, or VII-D is provided wherein W is CH.
  • the compound selectively inhibits one or more members of type I phosphatidylinositol 3-kinases (PI3-kinase) relative to other members of type I PI3-kinase, ascertained by an in vitro kinase assay.
  • PI3-kinase phosphatidylinositol 3-kinases
  • the compound selectively inhibits PI3 -kinase ⁇ and PI3-kinase ⁇ as compared to PI3-kinase ⁇ and PI3-kinase ⁇ . In other embodiments, the compound selectively inhibits PI3- kinase ⁇ and PI3-kinase ⁇ as compared to PI3-kinase ⁇ and PI3-kinase ⁇ .
  • the invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of Formula I-A, I-B, I-C, I-D, I-E, I-F, I-G, I-H, I-J, I-J-l, I-K, ⁇ -A, II- Al, II-A2, III-A, IV-A, V-A, V-Al, V-B, V-C, V-D, VI-
  • the invention also provides a method of treating a medical condition mediated by a type I-PI3 kinase (e.g. mediated by pi lO ⁇ , pi lO ⁇ , pi 10a, or pi lO ⁇ kinase), comprising administering to a subject in need thereof a therapeutically effective amount of a compound of Formula I-A, I-B, I-C, I-D, I-E, I-F, I-G, I-H, I-J, I-J-l, I-K, II-A, II-A1, II- A2, III- A, IV-A, V-A, V-Al, V-B, V-C, V-D, VI-A, VI-Al, VI- A2, VI-B, VI-C, VI-D, VII-A, VII-Al, VII-A2, VII-B, VII-C, or VII-D, or a pharmaceutically acceptable salt of a compound of Formula I-A, I-B, I-C, I
  • the medical condition is selected from the group consisting of hematologic malignancy, inflammation, autoimmune disease, rheumatoid arthritis, systemic lupus erythematosus, asthma, and cardiovascular disease.
  • the medical condition is selected from the group consisting of acute myelogenous leukemia, chronic myelogenous leukemia, mastocytosis, chronic lymphocytic leukemia, multiple myeloma, and myelodysplastic syndrome.
  • the method of treating a medical condition further comprises administering an anti-cancer agent.
  • the present invention also provides a method of inhibiting activity of a protein kinase and/or a lipid kinase present in a cell, comprising contacting said cell with an effective amount of a compound of Formula I-A, I-B, I-
  • structures depicted herein are also meant to include compounds which differ only in the presence of one or more isotopically enriched atoms.
  • compounds having the present structures wherein hydrogen is replaced by deuterium or tritium, or wherein carbon atom is replaced by 13 C- or 14 C- enriched carbon are within the scope of this invention.
  • the compounds of the present invention may also contain unnatural proportions of atomic isotopes at one or more of atoms that constitute such compounds.
  • the compounds may be radiolabeled with radioactive isotopes, such as for example tritium ( 3 H), iodine-125 ( 125 I) or carbon-14 ( 14 C). All isotopic variations of the compounds of the present invention, whether radioactive or not, are encompassed within the scope of the present invention.
  • agent refers to a biological, pharmaceutical, or chemical compound or other moiety.
  • Non-limiting examples include simple or complex organic or inorganic molecule, a peptide, a protein, an oligonucleotide, an antibody, an antibody derivative, antibody fragment, a vitamin derivative, a carbohydrate, a toxin, or a chemotherapeutic compound.
  • Various compounds can be synthesized, for example, small molecules and oligomers (e.g., oligopeptides and oligonucleotides), and synthetic organic compounds based on various core structures.
  • various natural sources can provide compounds for screening, such as plant or animal extracts, and the like. A skilled artisan can readily recognize that there is no limit as to the structural nature of the agents of the present invention.
  • agonist refers to a compound having the ability to initiate or enhance a biological function of a target protein, whether by inhibiting the activity or expression of the target protein. Accordingly, the term “agonist” is defined in the context of the biological role of the target polypeptide. While preferred agonists herein specifically interact with (e.g. bind to) the target, compounds that initiate or enhance a biological activity of the target polypeptide by interacting with other members of the signal transduction pathway of which the target polypeptide is a member are also specifically included within this definition.
  • antagonists are used interchangeably, and they refer to a compound having the ability to inhibit a biological function of a target protein, whether by inhibiting the activity or expression of the target protein. Accordingly, the terms “antagonist” and “inhibitors” are defined in the context of the biological role of the target protein. While preferred antagonists herein specifically interact with (e.g. bind to) the target, compounds that inhibit a biological activity of the target protein by interacting with other members of the signal transduction pathway of which the target protein is a member are also specifically included within this definition.
  • a preferred biological activity inhibited by an antagonist is associated with the development, growth, or spread of a tumor, or an undesired immune response as manifested in autoimmune disease.
  • an "anti-cancer agent”, “anti-tumor agent” or “chemotherapeutic agent” refers to any agent useful in the treatment of a neoplastic condition.
  • One class of anti-cancer agents comprises chemotherapeutic agents.
  • “Chemotherapy” means the administration of one or more chemotherapeutic drugs and/or other agents to a cancer patient by various methods, including intravenous, oral, intramuscular, intraperitoneal, intravesical, subcutaneous, transdermal, buccal, or inhalation or in the form of a suppository.
  • cell proliferation refers to a phenomenon by which the cell number has changed as a result of division. This term also encompasses cell growth by which the cell morphology has changed (e.g., increased in size) consistent with a proliferative signal.
  • the term "effective amount” or “therapeutically effective amount” refers to that amount of a compound described herein that is sufficient to effect the intended application including but not limited to disease treatment, as defined below.
  • the therapeutically effective amount may vary depending upon the intended application (in vitro or in vivo), or the subject and disease condition being treated, e.g., the weight and age of the subject, the severity of the disease condition, the manner of administration and the like, which can readily be determined by one of ordinary skill in the art.
  • the term also applies to a dose that will induce a particular response in target cells, e.g. reduction of platelet adhesion and/or cell migration.
  • the specific dose will vary depending on the particular compounds chosen, the dosing regimen to be followed, whether it is administered in combination with other compounds, timing of administration, the tissue to which it is administered, and the physical delivery system in which it is carried.
  • treatment or “treating,” or “palliating” or “ameliorating” is used interchangeably herein. These terms refer to an approach for obtaining beneficial or desired results including but not limited to therapeutic benefit and/or a prophylactic benefit.
  • therapeutic benefit is meant eradication or amelioration of the underlying disorder being treated.
  • a therapeutic benefit is achieved with the eradication or amelioration of one or more of the physiological symptoms associated with the underlying disorder such that an improvement is observed in the patient, notwithstanding that the patient may still be afflicted with the underlying disorder.
  • the compositions may be administered to a patient at risk of developing a particular disease, or to a patient reporting one or more of the physiological symptoms of a disease, even though a diagnosis of this disease may not have been made.
  • a prophylactic effect includes delaying or eliminating the appearance of a disease or condition, delaying or eliminating the onset of symptoms of a disease or condition, slowing, halting, or reversing the progression of a disease or condition, or any combination thereof.
  • salts refers to salts derived from a variety of organic and inorganic counter ions well known in the art.
  • Pharmaceutically acceptable acid addition salts can be formed with inorganic acids and organic acids.
  • Inorganic acids from which salts can be derived include, for example, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like.
  • Organic acids from which salts can be derived include, for example, acetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid, and the like.
  • Pharmaceutically acceptable base addition salts can be formed with inorganic and organic bases.
  • Inorganic bases from which salts can be derived include, for example, sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum, and the like.
  • Organic bases from which salts can be derived include, for example, primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, basic ion exchange resins, and the like, specifically such as isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, and ethanolamine.
  • the pharmaceutically acceptable base addition salt is chosen from ammonium, potassium, sodium, calcium, and magnesium salts.
  • “Pharmaceutically acceptable carrier” or “pharmaceutically acceptable excipient” includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions of the invention is contemplated. Supplementary active ingredients can also be incorporated into the compositions.
  • Signal transduction is a process during which stimulatory or inhibitory signals are transmitted into and within a cell to elicit an intracellular response.
  • a modulator of a signal transduction pathway refers to a compound which modulates the activity of one or more cellular proteins mapped to the same specific signal transduction pathway.
  • a modulator may augment (agonist) or suppress (antagonist) the activity of a signaling molecule.
  • Radionucleotides e.g., actinium and thorium radionuclides
  • LET low linear energy transfer
  • beta emitters conversion electron emitters
  • high-energy radiation including without limitation x- rays, gamma rays, and neutrons.
  • Prodrug is meant to indicate a compound that may be converted under physiological conditions or by solvolysis to a biologically active compound described herein.
  • prodrug refers to a precursor of a biologically active compound that is pharmaceutically acceptable.
  • a prodrug may be inactive when administered to a subject, but is converted in vivo to an active compound, for example, by hydrolysis.
  • the prodrug compound often offers advantages of solubility, tissue compatibility or delayed release in a mammalian organism (see, e.g., Bundgard, H., Design of Prodrugs (1985), pp. 7-9, 21-24 (Elsevier, Amsterdam).
  • prodrugs are also meant to include any covalently bonded carriers, which release the active compound in vivo when such prodrug is administered to a mammalian subject.
  • Prodrugs of an active compound, as described herein may be prepared by modifying functional groups present in the active compound in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent active compound.
  • Prodrugs include compounds wherein a hydroxy, amino or mercapto group is bonded to any group that, when the prodrug of the active compound is administered to a mammalian subject, cleaves to form a free hydroxy, free amino or free mercapto group, respectively.
  • Examples of prodrugs include, but are not limited to, acetate, formate and benzoate derivatives of an alcohol or acetamide, formamide and benzamide derivatives of an amine functional group in the active compound and the like.
  • R of an acyloxy group is optionally substituted by one or more substituents which independently are: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, trifluoromethyl, trifluoromethoxy, nitro, trimethylsilanyl, -OR a , -
  • R of an acyloxy group is optionally substituted by one or more of the following substituents: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, nitro, oxo, thioxo, trimethylsilanyl, -OR", - SR a , -OC(O)-R 8 , -N(R a ) 2) -C(O)R a , -C(O)OR 3 , -C(O)N(R 3 ) 2 , -N(R a )C(0)0R a , -N(R a )C(0)R a , -N(R a )S(0),R 3 (where t is 1 or 2), -S(O)
  • alkyl refers to an (aryl)alkyl — radical where aryl and alkyl are as disclosed herein and which are optionally substituted by one or more of the subsituents described as suitable substituents for aryl and alkyl respectively.
  • Alkoxy refers to a (alkyl)O-radical, where alkyl is as described herein and contains 1 to 10 carbons (e.g., Q- Cio alkyl). Whenever it appears herein, a numerical range such as “1 to 10" refers to each integer in the given range; e.g., "1 to 10 carbon atoms” means that the alkyl group may consist of 1 carbon atom, 2 carbon atoms, 3 carbon atoms, etc., up to and including 10 carbon atoms. In some embodiments, it is a C ⁇ -C 4 alkoxy group. A alkoxy moiety may be substituted by one or more of the substituents described as suitable substituents for an alkyl radical.
  • Alkyl refers to a straight or branched hydrocarbon chain radical consisting solely of carbon and hydrogen atoms, containing no unsaturation, having from one to ten carbon atoms (e.g., Ci-Ci 0 alkyl).
  • a numerical range such as “1 to 10” refers to each integer in the given range; e.g., "1 to 10 carbon atoms” means that the alkyl group may consist of 1 carbon atom, 2 carbon atoms, 3 carbon atoms, etc., up to and including 10 carbon atoms, although the present definition also covers the occurrence of the term "alkyl" where no numerical range is designated.
  • Typical alkyl groups include, but are in no way limited to, methyl, ethyl, propyl, isopropyl, n-butyl, iso-butyl, sec-butyl isobutyl, tertiary butyl, pentyl, isopentyl, neopentyl, hexyl, septyl, octyl, nonyl, decyl, and the like.
  • the alkyl is attached to the rest of the molecule by a single bond, for example, methyl (Me), ethyl (Et), w-propyl, 1-methylethyl (iso-propyl), w-butyl, /i-pentyl, 1,1-dimethylethyl (f-butyl), 3-methylhexyl, 2-methylhexyl, and the like.
  • an alkyl group is optionally substituted by one or more of the following substituents: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, nitro, oxo, thioxo, trimethylsilanyl, -OR a , - SR 8 , -OC(O)-R 3 , -N(R a ) 2 , -C(O)R 3 , -C(O)OR 3 , -C(O)N(R 3 ) 2 , -N(R a )C(O)OR a , -N(R a )C(O)R a , -N(R a )S(O) t R a (where t is 1 or 2), -
  • alkene refers to a group consisting of at least two carbon atoms and at least one carbon-carbon double bond
  • an "alkyne” moiety refers to a group consisting of at least two carbon atoms and at least one carbon-carbon triple bond.
  • the alkyl moiety, whether saturated or unsaturated, may be branched, straight chain, or cyclic.
  • alkenyl refers to a straight or branched hydrocarbon chain radical group consisting solely of carbon and hydrogen atoms, containing at least one double bond, and having from two to ten carbon atoms (ie. C 2 -Ci 0 alkenyl).
  • a numerical range such as “2 to 10” refers to each integer in the given range; e.g., "2 to 10 carbon atoms” means that the alkenyl group may consist of 2 carbon atoms, 3 carbon atoms, etc., up to and including 10 carbon atoms.
  • an alkenyl comprises two to eight carbon atoms.
  • an alkenyl comprises two to five carbon atoms (e.g., C 2 -C 5 alkenyl).
  • the alkenyl is attached to the rest of the molecule by a single bond, for example, ethenyl (i.e., vinyl), prop-1-enyl (i.e., allyl), but-1-enyl, pent-1-enyl, penta-l,4-dienyl, and the like.
  • an alkenyl group is optionally substituted by one or more of the following substituents alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, nitro, oxo, thioxo, trimethylsilanyl, -OR", -
  • Alkynyl refers to a straight or branched hydrocarbon chain radical group consisting solely of carbon and hydrogen atoms, containing at least one triple bond, having from two to ten carbon atoms (ie. C 2 -Ci 0 alkynyl). Whenever it appears herein, a numerical range such as “2 to 10" refers to each integer in the given range; e.g., "2 to 10 carbon atoms” means that the alkynyl group may consist of 2 carbon atoms, 3 carbon atoms, etc., up to and including 10 carbon atoms. In certain embodiments, an alkynyl comprises two to eight carbon atoms.
  • an alkynyl has two to five carbon atoms (e.g., C 2 -C 5 alkynyl).
  • the alkynyl is attached to the rest of the molecule by a single bond, for example, ethynyl, propynyl, butynyl, pentynyl, hexynyl, and the like.
  • an alkynyl group is optionally substituted by one or more of the following substituents alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, nitro, oxo, thioxo, trimethylsilanyl, -OR a , - SR a , -OC(O)-R 8 , -N(R a ) 2 , -C(O)R 3 , -C(O)OR 8 , -C(0)N(R a ) 2 , -N(R 8 )C(O)OR a , -N(R 8 )C(O)R 8 , -N(R 8 )S(O) t R 8 (where t is 1 or 2), -S(O)
  • Amino or “amine” refers to a -N(R 8 ) 2 radical group, where each R a is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl or heteroarylalkyl, unless stated otherwise specifically in the specification.
  • an amino group is optionally substituted by one or more substituents which independently are: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, nitro, oxo, thioxo, trimethylsilanyl, -OR a , -
  • Amide refers to a chemical moiety with formula -C(O)NHR or -NHC(O)R, where R is selected from the group consisting of alkyl, cycloalkyl, aryl, heteroaryl (bonded through a ring carbon) and heteroalicyclic (bonded through a ring carbon). In some embodiments it is a CpC 4 amido or amide radical, which includes the amide carbonyl in the total number of carbons in the radical. Unless stated otherwise specifically in the specification, an amino group is optionally substituted independently by one or more of the substituents as described herein for alkyl, cycloalkyl, aryl, heteroaryl, or heterocyclyl.
  • An amide may be an amino acid or a peptide molecule attached to a compound of Formula (I), thereby forming a prodrug. Any amine, hydroxy, or carboxyl side chain on the compounds described herein can be amidified. The procedures and specific groups to make such amides are known to those of skill in the art and can readily be found in reference sources such as Greene and Wuts, Protective Groups in Organic Synthesis, 3.sup.rd Ed., John Wiley & Sons, New York, N.Y., 1999, which is incorporated herein by reference in its entirety.
  • Aromatic or “aryl” refers to an aromatic radical with six to ten ring atoms (e.g., C 6 -Ci 0 aromatic or C 6 -C 10 aryl) which has at least one ring having a conjugated pi electron system which is carbocyclic (e.g., phenyl, fluorenyl, and naphthyl).
  • a numerical range such as “6 to 10” refers to each integer in the given range; e.g., "6 to 10 ring atoms” means that the aryl group may consist of 6 ring atoms, 7 ring atoms, etc., up to and including 10 ring atoms.
  • an aryl moiety is optionally substituted by one or more substituents which are independently: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, nitro, oxo, thioxo, trimethylsilanyl, -OR a , -
  • Cyano refers to a -CN radical.
  • Cycloalkyl refers to a monocyclic or polycyclic radical that contains only carbon and hydrogen, and may be saturated, or partially unsaturated. Cycloalkyl groups include groups having from 3 to 10 ring atoms (ie. C 2 -Ci 0 cycloalkyl). Whenever it appears herein, a numerical range such as “3 to 10" refers to each integer in the given range; e.g., "3 to 10 carbon atoms” means that the cycloalkyl group may consist of 3 carbon atoms, etc., up to and including 10 carbon atoms. In some embodiments, it is a C 3 -C 5 cycloalkyl radical.
  • cycloalkyl groups include, but are not limited to the following moieties: cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloseptyl, cyclooctyl, cyclononyl, cyclodecyl, norbornyl, and the like.
  • a cycloalkyl group is optionally substituted by one or more substituents which independently are: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, nitro, oxo, thioxo, trimethylsilanyl, -OR a , -
  • Ester refers to a chemical radical of formula -COOR, where R is selected from the group consisting of alkyl, cycloalkyl, aryl, heteroaryl (bonded through a ring carbon) and heteroalicyclic (bonded through a ring carbon). Any amine, hydroxy, or carboxyl side chain on the compounds described herein can be esterified. The procedures and specific groups to make such esters are known to those of skill in the art and can readily be found in reference sources such as Greene and Wuts, Protective Groups in Organic Synthesis, 3.sup.rd Ed., John Wiley & Sons, New York, N.Y., 1999, which is incorporated herein by reference in its entirety.
  • an ester group is optionally substituted by one or more substituents which independently are: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, nitro, oxo, thioxo, trimethylsilanyl, -OR 8 , - SR a , -OC(O)-R 8 , -N(R a ) 2 , -C(O)R 8 , -C(O)OR 8 , -C(0)N(R a ) 2 , -N(R 8 )C(O)OR a , -N(R a )C(0)R a , -N(R a )S(O) t R 8 (where t is 1 or 2), -S(O) 1 OR
  • Fluoroalkyl refers to an alkyl radical, as defined above, that is substituted by one or more fluoro radicals, as defined above, for example, trifluoromethyl, difluoromethyl, 2,2,2-trifluoroethyl, l-fluoromethyl-2-fluoroethyl, and the like.
  • the alkyl part of the fluoroalkyl radical may be optionally substituted as defined above for an alkyl group.
  • Halo means fluoro, chloro, bromo or iodo.
  • haloalkyl means fluoro, chloro, bromo or iodo.
  • haloalkenyl means fluoro, chloro, bromo or iodo.
  • haloalkynyl means alkyl, alkenyl, alkynyl and alkoxy structures that are substituted with one or more halo groups or with combinations thereof.
  • fluoroalkyl and fluoroalkoxy include haloalkyl and haloalkoxy groups, respectively, in which the halo is fluorine.
  • Heteroalkyl “heteroalkenyl” and “heteroalkynyl” include optionally substituted alkyl, alkenyl and alkynyl radicals and which have one or more skeletal chain atoms selected from an atom other than carbon, e.g., oxygen, nitrogen, sulfur, phosphorus or combinations thereof.
  • a numerical range may be given, e.g. C 1 -C 4 heteroalkyl which refers to the chain length in total, which in this example is 4 atoms long.
  • a — CH 2 OCH 2 CH 3 radical is referred to as a "C 4 " heteroalkyl, which includes the heteroatom center in the atom chain length description.
  • a heteroalkyl group may be substituted with one or more substituents which independently are: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, nitro, oxo, thioxo, trimethylsilanyl, -OR a , -
  • Heteroaryl or, alternatively, “heteroaromatic” refers to a 5- to 18-membered aromatic radical (e.g., C 5 -C 13 heteroaryl) that includes one or more ring heteroatoms selected from nitrogen, oxygen and sulfur, and which may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system.
  • a numerical range such as “5 to 18” refers to each integer in the given range; e.g., "5 to 18 ring atoms” means that the heteroaryl group may consist of 5 ring atoms, 6 ring atoms, etc., up to and including 18 ring atoms.
  • heteroaryl refers to an aromatic group in which at least one of the skeletal atoms of the ring is a nitrogen atom.
  • the polycyclic heteroaryl group may be fused or non-fused.
  • the heteroatom(s) in the heteroaryl radical is optionally oxidized.
  • One or more nitrogen atoms, if present, are optionally quaternized.
  • the heteroaryl is attached to the rest of the molecule through any atom of the ring(s).
  • heteroaryls include, but are not limited to, azepinyl, acridinyl, benzimidazolyl, benzindolyl, 1,3-benzodioxolyl, benzofuranyl, benzooxazolyl, benzofd]thiazolyl, benzothiadiazolyl, benzo[6][l,4]dioxepinyl, benzo[b][l,4]oxazinyl, 1,4-benzodioxanyl, benzonaphthofuranyl, benzoxazolyl, benzodioxolyl, benzodioxinyl, benzoxazolyl, benzopyranyl, benzopyranonyl, benzofuranyl, benzofuranonyl, benzofurazanyl, benzothiazolyl, benzothienyl (benzothiophenyl), benzothieno[3,2-d]pyrimidinyl
  • a heteraryl moiety is optionally substituted by one or more substituents which are independently: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, nitro, oxo, thioxo, trimethylsilanyl, -OR a , -
  • Heteroarylalkyl or “hetarylalkyl” refers to an (heteroaryl)alkyl — radical where heteroaryl and alkyl are as disclosed herein and which are optionally substituted by one or more of the subsituents described as suitable substituents for aryl and alkyl respectively.
  • ' ⁇ eterocyclyl or “heterocycloalkyl” refers to a stable 3- to 18-membered non-aromatic ring (e.g., C 3 -Ci 8 heterocyclyl) radical that comprises two to twelve carbon atoms and from one to six heteroatoms selected from nitrogen, oxygen and sulfur.
  • a numerical range such as “3 to 18” refers to each integer in the given range; e.g., "3 to 18 ring atoms” means that the heteroaryl group may consist of 3 ring atoms, 4 ring atoms, etc., up to and including 18 ring atoms.
  • the heterocyclyl radical is a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems.
  • the heteroatoms in the heterocyclyl radical may be optionally oxidized.
  • One or more nitrogen atoms, if present, are optionally quaternized.
  • the heterocyclyl radical is partially or fully saturated.
  • heterocyclyl may be attached to the rest of the molecule through any atom of the ring(s).
  • heterocyclyl radicals include, but are not limited to 6,7- dihydro-5H-cyclopenta[b]pyridine, dioxolanyl, thienyl[l,3]dithianyl, decahydroisoquinolyl, imidazolinyl, imidazolidinyl, isothiazolidinyl, isoxazolidinyl, morpholinyl, octahydroindolyl, octahydroisoindolyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolidinyl, oxazolidinyl, piperidinyl, piperazinyl, 4-piperidonyl, pyrrolidinyl, pyrazolidinyl, quinuclidinyl, thiazolidinyl, t
  • a heterocylyl moiety is optionally substituted by one or more substituents which independently are: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, nitro, oxo, thioxo, trimethylsilanyl, -OR", - SR a , -OC(O)-R 3 , -N(R 3 ) 2 , -C(O)R 3 , -C(O)OR 3 , -C(0)N(R a ) 2 , -N(R 3 )C(O)OR a , -N(R a )C(O)R a , -N(R 3 )S(O) t R 3 (where t is 1 or 2), -S(O)
  • Heteroalicyclic refers to a cycloalkyl radical that includes at least one heteroatom selected from nitrogen, oxygen and sulfur.
  • the radicals may be fused with an aryl or heteroaryl.
  • heteroalicyclic also includes all ring forms of the carbohydrates, including but not limited to the monosaccharides, the disaccharides and the oligosaccharides.
  • a heteroalicyclic group is optionally substituted by one or more of substituents which are independently: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, nitro, oxo, thioxo, trimethylsilanyl, -OR a , -
  • Moiety refers to a specific segment or functional group of a molecule. Chemical moieties are often recognized chemical entities embedded in or appended to a molecule.
  • Niro refers to the -NO 2 radical.
  • Oxa refers to the -O- radical.
  • a sulfonamido group is optionally substituted by one or more of the subsituents described for alkyl, cycloalkyl, aryl, heteroaryl respectively
  • R is selected from the group consisting of alkyl, cycloalkyl, aryl, heteroaryl (bonded through a ring carbon) and heteroalicyclic (bonded through a ring carbon).
  • a sulfonategroup is optionally substituted on R by one or more of the subsituents described for alkyl, cycloalkyl, aryl, heteroaryl respectively.
  • Thiocyanato refers to a -CNS radical.
  • substituents themselves may be substituted, for example, a cycloakyl substituent may have a halide substituted at one or more ring carbons, and the like.
  • the protecting groups that may form the protective derivatives of the above substituents are known to those of skill in the art and may be found in references such as Greene and Wuts, above.
  • the compounds presented herein may possess one or more chiral centers and each center may exist in the R or S configuration.
  • the compounds presented herein include all diastereomeric, enantiomeric, and epimeric forms as well as the appropriate mixtures thereof.
  • Stereoisomers may be obtained, if desired, by methods known in the art as, for example, the separation of stereoisomers by chiral chromatographic columns.
  • the methods and formulations described herein include the use of N-oxides, crystalline forms (also known as polymorphs), or pharmaceutically acceptable salts of compounds having the structure of Formula (I), as well as active metabolites of these compounds having the same type of activity.
  • the compounds described herein can exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like.
  • the solvated forms of the compounds presented herein are also considered to be disclosed herein.
  • selective inhibition or “selectively inhibit” as referred to a biologically active agent refers to the agent's ability to preferentially reduce the target signaling activity as compared to off-target signaling activity, via direct or interact interaction with the target.
  • Compounds described can contain one or more asymmetric centers and may thus give rise to diastereomers and optical isomers.
  • the present invention includes all such possible diastereomers as well as their racemic mixtures, their substantially pure resolved enantiomers, all possible geometric isomers, and pharmaceutically acceptable salts thereof. All formulae disclosed here are shown without a definitive stereochemistry at certain positions.
  • the present invention includes all stereoisomers of the shown formulae and pharmaceutically acceptable salts thereof. Further, mixtures of stereoisomers as well as isolated specific stereoisomers are also included.
  • the products of such procedures can be a mixture of stereoisomers.
  • “Isomers” are different compounds that have the same molecular formula.
  • “Stereoisomers” are isomers that differ only in the way the atoms are arranged in space, i.e. having a different stereochemical configuration.
  • “Enantiomers” are a pair of stereoisomers that are non-superimposable mirror images of each other.
  • a 1 : 1 mixture of a pair of enantiomers is a "racemic” mixture.
  • the term “(. ⁇ .)” is used to designate a racemic mixture where appropriate.
  • “Diastereoisomers” are stereoisomers that have at least two asymmetric atoms, but which are not mirror-images of each other.
  • the absolute stereochemistry is specified according to the Cahn-Ingold-Prelog R-S system.
  • the stereochemistry at each chiral carbon can be specified by either R or S.
  • Resolved compounds whose absolute configuration is unknown can be designated (+) or (-) depending on the direction (dextro- or levorotatory) which they rotate plane polarized light at the wavelength of the sodium D line.
  • Certain of the compounds described herein contain one or more asymmetric centers and can thus give rise to enantiomers, diastereomers, and other stereoisomeric forms that can be defined, in terms of absolute stereochemistry, as (R)- or (S)-.
  • Optically active (R)- and (S)- isomers can be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques.
  • R optically active
  • S chiral reagents
  • the compounds described herein contain olef ⁇ nic double bonds or other centers of geometric asymmetry, and unless specified otherwise, it is intended that the compounds include both E and Z geometric isomers.
  • Moiety refers to a specific segment or functional group of a molecule. Chemical moieties are often recognized chemical entities embedded in or appended to a molecule.
  • a "leaving group or atom” is any group or atom that will, under the reaction conditions, cleave from the starting material, thus promoting reaction at a specified site. Suitable examples of such groups unless otherwise specified are halogen atoms, mesyloxy, p-nitrobenzensulphonyloxy and tosyloxy groups.
  • Protecting group has the meaning conventionally associated with it in organic synthesis, i.e. a group that selectively blocks one or more reactive sites in a multifunctional compound such that a chemical reaction can be carried out selectively on another unprotected reactive site and such that the group can readily be removed after the selective reaction is complete.
  • a variety of protecting groups are disclosed, for example, in T.H. Greene and P. G. M. Wuts, Protective Groups in Organic Synthesis, Third Edition, John Wiley & Sons, New York (1999).
  • a hydroxy protected form is where at least one of the hydroxy groups present in a compound is protected with a hydroxy protecting group.
  • amines and other reactive groups may similarly be protected
  • a prophylactic effect includes delaying or eliminating the appearance of a disease or condition, delaying or eliminating the onset of symptoms of a disease or condition, slowing, halting, or reversing the progression of a disease or condition, or any combination thereof.
  • co-administration encompass administration of two or more agents to an animal so that both agents and/or their metabolites are present in the animal at the same time.
  • Co-administration includes simultaneous administration in separate compositions, administration at different times in separate compositions, or administration in a composition in which both agents are present.
  • in vivo refers to an event that takes place in a subject's body.
  • in vitro refers to an event that takes places outside of a subject's body.
  • an in vitro assay encompasses any assay run outside of a subject assay.
  • in vitro assays encompass cell-based assays in which cells alive or dead are employed.
  • In vitro assays also encompass a cell-free assay in which no intact cells are employed.
  • a "subject,” “individual” or “patient” is used interchangeably herein, which refers to a vertebrate, preferably a mammal, more preferably a human. Mammals include, but are not limited to, murines, simians, humans, farm animals, sport animals, and pets. Tissues, cells and their progeny of a biological entity obtained in vitro or cultured in vitro are also encompassed. ⁇ . COMPOSITIONS 189] In one aspect, the present invention provides a compound of the formula:
  • X 5 and X 6 are C-R 6 , N, C- I ⁇ R 1 , or N- i ⁇ R 1 wherein one of X 5 and X 6 is C- L'-R 1 or N- L'-R 1 ;
  • X 1 is C or N; and
  • X 2 and X 8 are independently N or C-R 6 ;
  • X 3 and X 7 are C or N, and at least one of X 3 and X 7 is C;
  • X 4 in Formula I-A or I-C is C or N; and X 4 in Formula I-B or I-D is C-R 6 , NH, or N; and no more than two adjacent ring atoms are N or NH;
  • R 1 is hydrogen, halogen, -CN, -OR 10 , -S(O) n R 11 , -NR 12 R 13 , -C(O)R 14 , alkyl, alkenyl, alkyn
  • each R 11 is independently -NR 16 R 17 , alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
  • each R 12 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
  • each R 13 is independently hydrogen, -S(O) n R 18 , -C(O)R 19 , alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; [00209] or R 12 and R 13 are taken together to form a 5, 6 or 7-membered ring, wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom Of-NR 12 R 13 ; and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted; [
  • each R 18 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
  • each R 19 is independently -NR 24 R 25 , hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
  • each of R 20 and R 21 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, or R 20 and R 21 are taken together to form a 5, 6 or 7-membered ring, wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom
  • each of R 22 and R 23 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, or R 22 and R 23 are taken together to form a 5, 6 or 7-membered ring, wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom
  • each of R 24 and R 25 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, or R 24 and R 25 are taken together to form a 5, 6 or 7-membered ring, wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom
  • a compound of one of Formula I- A, I-B, I-C, or I-D has a structure of one of the following formulae:
  • the compound of Formula I-E has a structure of Formula I-J or Formula
  • the compound of Formula!- J is a compound of Formula I-J- 1 :
  • the compound of Formula I-E is a compound of Formula H-A, Formula H-A 1 or Formula II-A2:
  • a compound of Formula I-E has a structure of Formula IH-A: [00225] In other embodiments, a compound of Formula I-E has a structure of Formula IV-A:
  • W is C-R 6 . In other embodiments, W is CH. In yet other embodiments W is N. [00227] In another aspect, a compound of the invention has a structure of one of the following formulae:
  • Xi is C or N; and X 2 and X 8 are independently N, or C-R 6 ; [00232] X 3 and X 7 are C or N, at least one of X 3 and X 7 is C; [00233] X 4 in Formula V-A and Formula V-C is C or N; and X 4 in Formula V-B and Formula V-D is C-R 6 , NH, or N; and wherein no more than two adjacent ring atoms are N or NH; [00234] R 1 is hydrogen, halogen, -CN, -OR 10 , -S(O) n R 11 , -NR 12 R 13 , -C(O)R 14 , alkyl, alkenyl, alkynyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, and n is an integer from 0 to 2; [00235] L 1 is a bond, alkylene, hetero
  • q is an integer from 0 to 5;
  • each R 6 is independently hydrogen, halogen, -CN, -OR 10 , -S(O) n R 11 , -NR 12 R 13 , -C(O)R 14 , alkyl, alkenyl, alkynyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, wherein n is independently an integer from 0 to 2;
  • each of R 7 and R 8 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocyclyalkyl, or R 7 and R 8 taken together form a cycloalkyl, heterocycloalkyl or aryl ring;
  • R 9 is hydrogen, -C(O)R 14 , alkyl, alkenyl, alkynyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
  • each R 10 is independently hydrogen, -C(O)R 15 , alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
  • each R 11 is independently -NR 16 R 17 , alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
  • each R 12 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
  • each R 13 is independently hydrogen, -S(O) n R 18 , -C(O)R 19 , alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
  • R 12 and R 13 are taken together to form a 5, 6, 7, or 8 membered ring, wherein the 5, 6, 7, or 8 membered ring contains 0, 1 , 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom of -NR 12 R 13 , and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted;
  • each R 14 is independently -NR 20 R 21 , hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
  • each R 15 is independently -NR 22 R 23 , hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
  • each of R 16 and R 17 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; or R 16 and R 17 are taken together to form a 5, 6, 7 or 8 membered ring, wherein the 5, 6, 7, or 8 membered ring contains 0, 1 , 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom Of -NR 16 R 17 , and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted;
  • each R 18 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
  • each R 19 is independently -NR 24 R 25 , hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
  • each of R 20 and R 21 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, or R 20 and R 21 are taken together to form a 5, 6, 7, or 8 membered ring, wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom Of-NR 20 R 21 , and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted;
  • each of R 22 and R 23 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, or R 22 and R 23 are taken together to form a 5, 6, 7, or 8 membered ring, wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom Of-NR 22 R 23 , and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted; and
  • each of R 24 and R 25 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, or R 24 and R 25 are taken together to form a 5, 6, 7, or 8 membered ring wherein the 5, 6, 7, or 8 membered ring contains 0, 1 , 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom Of-NR 24 R 25 , and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted.
  • the compound of Formula V-A is a compound having the structure of Formula V-Al:
  • the compound of Formula V-A, Formula V-B, Formula V-C or Formula V-D is a compond having a structure of one of the following formulae:
  • a compound of Formula VI-A is a compound having a structure of Formula VI-Al or Formula VI- A2:
  • the compound of Formula V-A, Formula V-B, Formula V-C or Formula V-D is a compond having a structure of one of the following formulae:
  • a compound of Formula VI-A is a compound having a structure of Formula VI-Al or Formula VI- A2:
  • z is 1, 2 or 3.
  • z may be 1.
  • Each R 7 and R 8 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocyclyalkyl, or R 7 and R 8 taken together form a cycloalkyl, heterocycloalkyl or aryl ring.
  • L is methylene or ethylene. In other embodiments, L is methylene or ethylene substituted with additional alkyl groups such as methyl, ethyl or isopropyl.
  • R 1 is halogen, -CN, -OR 10 , -S(O) n R 11 , -NR 12 R 13 , -C(O)R 14 , alkyl, alkenyl, alkynyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, wherein n is independently an integer from 0
  • R 1 may be an monocyclic aryl group, that can be substituted or is unsubstituted; a bicyclic aryl group, that can be substituted or unsubstituted; a monocyclic heteroaryl group, that can be substituted or is unsubstituted; or a bicyclic heteroaryl group, that can be substituted or is unsubstituted.
  • R 1 may be a monocyclic heterocycloalkyl group, that can be substituted or is unsubstituted; a bicyclic heterocycloalkyl group, that can be substituted or is unsubstituted; a heteroalkyl group, that can be substituted or is unsubstituted; or an alkyl group, that can be substituted or is unsubstituted.
  • R 1 is substituted with halogen, -CN, -OR 10 , -S(O) n R 11 , -NR 12 R 13 , -C(O)R 14 , alkyl, alkenyl, alkynyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl.
  • R 1 is an aryl group substituted with -C(O)R 14 .
  • R 1 is a heteroaryl group substituted with halogen or -NR 12 R 13 .
  • R 1 is a monocyclic aromatic or heteroaryl group
  • substitutions on R 1 may be effected, for example, at the ortho, meta and/or para positions.
  • R 1 is monocyclic aryl. In other embodiments, R 1 is bicyclic aryl.
  • R 1 groups are illustrated below :
  • L 1 may be a bond or a linker connecting R 1 to a heterocyclic moiety of the compounds of the invention.
  • L 1 is a bond.
  • L 1 is an alkylene group.
  • R 4 and R 5 taken together form a 5, 6 or 7-membered ring, unsubstituted or substituted with (R 6 ) q .
  • R 4 and R 5 taken together form a 5 or 6-membered unsubstituted or substituted with (R 6 ) q .
  • the ring may be saturated, unsaturated, or partially unsaturated, and may be aromatic or nonaromatic.
  • the ring is aromatic and substituted by R 6 .
  • R 4 and R 5 taken together form a group having a structure of one of the folowing formulae:
  • R 6 may be any substituent such as halogen, -CN, -OR 10 , -S(O) n R 11 , -NR 12 R 13 , -C(O)R 14 , alkyl, alkenyl, alkynyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, wherein n is independently an integer from
  • R 9 may be any cyclic, linear or branched substituent.
  • R 9 is halogen, -CN, -OR 10 , -S(O) n R 11 , -NR 12 R 13 , -C(O)R 14 , alkyl, alkenyl, alkynyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, wherein n is independently an integer from 0 to 2.
  • R 9 may be an monocyclic aryl group, substituted or unsubstituted; a bicyclic aryl group, substituted or unsubstituted; a monocyclic heteroaryl group, substituted or unsubstituted; a bicyclic heteroaryl group, substituted or unsubstituted; a monocyclic heterocycloalkyl group, substituted or unsubstituted; a bicyclic heterocycloalkyl group, substituted or unsubstituted; a heteroalkyl group, substituted or unsubstituted; an alkyl group, substituted or unsubstituted.
  • R 9 is substituted with halogen, -CN, -
  • OR 10 -S(O) n R 11 , -NR 12 R 13 , -C(O)R 14 , alkyl, alkenyl, alkynyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl.
  • R 9 is any of the groups shown below:
  • R 12 and R 13 in -NR 12 R 13 , R 16 and R 17 in -NR 16 R 17 , R 20 and R 21 in -NR 20 R 21 , R 22 and R 23 in -NR 22 R 23 , or R 24 and R 25 i -NR 24 R 25 are taken together with the nitrogen atom to which they are attached to form a moiety of one of the following formulae:
  • the compound of the invention comprises a heterocyclic subunit of the formulas Za or Zb shown below.
  • the symbol ⁇ represents the point of attachment to the linker L.
  • the point of attachment to the linker L is X 4 or the exocyclic amine moiety. Additional substituents may be present at any or several of the X 1 , X 2 , X 4 , X 5 , and X 6 positions.
  • an R 1 group as defined above may be connected to X 6 .
  • N , -N ⁇ or -NH- as required for proper valency.
  • Any illustrated subunit Z may be combined with the embodiments disclosed for R 1 through R 9 , L and W to design a compound of the invention
  • X represents a halo group (Cl, Br, I or F).
  • X represents a halo group (Cl, Br, I or F).
  • Varioius reagents may be used to perform the coupling reactions necessary to introduce the R 9 , L and Z moieties.
  • X and Y are reactive groups which can react with precursors of R 9 and L or Z moieties.
  • X and Y are latent or protected reaction groups which are converted to reactive groups during or prior to the coupling reactions.
  • Various coupling reactions may be suitable for this purpose.
  • Some known types of coupling reactions which can involve aromatic ring compounds include the Heck reactions and Suzuki reactions. Such reactions are described, for example, in N. Miyaura and A. Suzuki, Chem. Rev. 1995, 95, 2457-2483.
  • X may be a halogen atom.
  • IfR 9 comprises a primary or secondary amino group, it may be attached to the aryl group via an amination reaction. Such a reaction may be performed in the presence of a base and/or a catalyst such as a palladium catalyst.
  • X is a halogen atom and R 9 is an aryl group.
  • a coupling may be effected between X and a precursor compound of R 9 , such as a boronic acid (Suzuki coupling) or stannane precursor (Stille coupling).
  • Scheme 3 shows the preparation of a compound of the invention by reduction of a starting aldehyde 3a to an alcohol in step 1. Such a reaction may be performed, for example, by sodium borohydride reduction in methanol.
  • step 2 compound 3b is coupled to pyrrolidine in the presence of a solvent such as 1 ,4-dioxane.
  • the alcohol 3c is converted to a reactive halide in step 3 by reaction with a reagent such as CBrVPPh 3 in a solvent such as dichloromethane.
  • a heterocyclic moiety such as a pyrazolopyrimide (e.g. 3-iodo-lH-pyrazolo[3,4-d]pyrimidin-4-amine) may be coupled to the halide 3d (e.g. in a base such as potassium carbonate and a solvent such as dimethylformamide) to form the compound 3e in step 4.
  • the pyrazolopyrimidine subunit is further derivatized via a reaction such as a Suzuki coupling (e.g. using Pd(PPh 3 ) 4 as a catalyst) to yield compound 3f.
  • Scheme 3 Synthesis of 3-(4-amino-l-((2-(pyrrolidin-l-yl)quinoIin-3-yl)methyl)-lH-pyrazolo[3,4-d]pyrimidin-3- yl)-5-fluorophenol
  • Scheme 4 shows an alternative synthesis of the compound 3f.
  • a starting aldehyde 3a is reduced to an alcohol.
  • Such a reaction may be performed, for example, by sodium borohydride reduction in methanol.
  • compound 3b is converted to a reactive halide 4a by reaction with a reagent such as CBr,
  • a heterocyclic moiety such as a pyrazolopyrimide (e.g. 3-iodo-lH- pyrazolo[3,4-d]pyrimidin-4-amine) is performed in step 3, for example in a base such as potassium carbonate and a solvent such as dimethylformamide) to yield compound 4b.
  • Compound 4b is then coupled to pyrrolidine in Step 4 in the presence of a solvent such as 1,4-dioxane to produce compound 3e.
  • compound 3e is further derivatized via a reaction such as a Suzuki coupling (e.g. using Pd(PPh 3 ) 4 as a catalyst) to yield compound 3f.
  • Scheme 5 shows an additional alternative synthesis of the compound 3f.
  • a starting aldehyde 3a is reduced to an alcohol. Such a reaction may be performed, for example, by sodium borohydride reduction in methanol.
  • compound 3b is converted to a reactive halide 4b by reaction with a reagent such as CBr 4 ZPPh 3 in a solvent such as dichloromethane. Coupling to a heterocyclic moiety such as a derivatized pyrazolopyrimide (e.g.
  • 3-(3-fluoro-5-methoxyphenyl)-lH-pyrazolo[3,4-d]pyrimidin-4-amine) is performed in step 3, for example in a base such as potassium t-butoxide and a solvent such as dimethylformamide) to yield compound 5a.
  • a base such as potassium t-butoxide and a solvent such as dimethylformamide
  • the resulting compound 5a is deprotected, for example by removal of a methoxy group by a reagent such as BBr 3 in dichloromethane.
  • the resulting compound 5b is then coupled to pyrrolidine in the presence of a solvent such as 1 ,4-dioxane to yield compound 3f .
  • Scheme 6 shows the preparation of a compound of the invention by reduction of a starting aldehyde 6a to an alcohol in step 1. Such a reaction may be performed, for example, by sodium borohydride reduction in methanol.
  • step 2 compound 6b is coupled to an aryl group via, for example, a Suzuki coupling.
  • o-tolylboronic acid is reacted with 6b in the presence of Pd(PPh 3 ) 4 , a base such as Na 2 CO 3 and a solvent such as DMF.
  • the resulting alcohol 6c is converted to a reactive halide in step 3 by reaction with a reagent such as CBr 4 ZPPh 3 in a solvent such as dichloromethane.
  • a heterocyclic moiety such as a pyrazolopyrimide e.g. 3-iodo-lH-pyrazolo[3,4-d]pyrimidin-4-amine
  • the halide 6d e.g. in a base such as potassium carbonate and a solvent such as dimethylformamide
  • the pyrazolopyrimidine subunit is further derivatized via a reaction such as a Suzuki coupling (e.g. using Pd(PPh 3 ) 4 as a catalyst) to yield compound 6f.
  • Scheme 7 shows an alternative synthesis of the compound 6f.
  • a starting aldehyde 6a is reduced to an alcohol. Such a reaction may be performed, for example, by sodium borohydride reduction in methanol.
  • compound 6b is converted to a reactive halide 7a by reaction with a reagent such as CBr 4 ZPPh 3 in a solvent such as dichloromethane. Coupling to a heterocyclic moiety such as a derivatized pyrazolopyrimide (e.g.
  • 3-(3- fluoro-5-methoxyphenyl)-lH-pyrazolo[3,4-d]pyrimidin-4-amine) is performed in step 3, for example in a base such as potassium t-butoxide and a solvent such as dimethylformamide).
  • the resulting compound 7b is then coupled to an aryl group via, for example, a Suzuki coupling.
  • o-tolylboronic acid is reacted with 7b in the presence of Pd(PPh 3 ) 4 , a base such as Na 2 CO 3 and a solvent such as DMF.
  • the resulting compound 7c is deprotected, for example by removal of a methoxy group by a reagent such as BBr 3 in dichloromethane to yield compound 6f.
  • Scheme 8 shows the preparation of a compound of the invention by reduction of a starting acid 8a to an alcohol in step 1. Such a reaction may be performed, for example, by LiAlH 4 reduction in methanol.
  • step 2 compound 8b is coupled to an aryl group via, for example, a Suzuki coupling.
  • 2- fluorophenylboronic acid is reacted with 8b in the presence of Pd(PPh 3 ) 4 , a base such as Na 2 CO 3 and a solvent such as DME-water.
  • the resulting alcohol 8c is converted to a reactive halide in step 3 by reaction with a reagent such as CBr 4 ZPPh 3 in a solvent such as dichloromethane.
  • a heterocyclic moiety such as a pyrazolopyrimide e.g. 3-iodo-lH-pyrazolo[3,4-d]pyrimidin-4-amine
  • the halide 8d e.g. in a base such as potassium carbonate and a solvent such as dimethylformamide
  • the pyrazolopyrimidine subunit is further derivatized via a reaction such as a Suzuki coupling (e.g. using Pd(PPh 3 ) 4 as a catalyst) to yield compound 8f .
  • Scheme 8 Synthesis of 3-(4-amino-l-((2-(2-fluorophenyl)-6,7-dihydro-5H-cyclopenta[b]pyridin-3-yl)methyl)-lH- pyrazolo [3,4-d] pyrimidin-3-yl)-5-fluorophenol (80
  • Scheme 9 shows the preparation of a compound of the invention by reduction of a starting aldehyde 9a to an alcohol in step 1. Such a reaction may be performed, for example, by sodium borohydride reduction in methanol.
  • step 2 compound 9b is coupled to pyrrolidine in the presence of a solvent such as 1,4-dioxane.
  • the resulting alcohol 9c is converted to a reactive halide in step 3 by reaction with a reagent such as CBr + ZPPh 3 in a solvent such as acetonitrile.
  • a heterocyclic moiety such as a pyrazolopyrimide (e.g. 3-iodo-lH-pyrazolo[3,4- d]pyrimidin-4-amine) may be coupled to the halide 9d (e.g. in a base such as potassium carbonate and a solvent such as dimethylformamide) to form the compound 9e in step 4.
  • the pyrazolopyrimidine subunit is further derivatized via a reaction such as a Suzuki coupling (e.g. using Pd(PPh 3 ) 4 as a catalyst) with 3-fluoro-5- hydroxyphenylboronic acid to yield compound 9f.
  • Scheme 10 illustrates a synthesis of the compound 1Of.
  • a starting aldehyde 10a is converted to an alcohol, for example, by Grignard addition of an alkylmagnesium halide reagent.
  • compound 10b is converted to a reactive halide by reaction with a reagent such as CBr 4 /PPh 3 in a solvent such as acetonitrile. Coupling to a heterocyclic moiety such as a pyrazolopyrimide (e.g.
  • 3-iodo-lH-pyrazolo[3,4-d]pyrimidin-4- amine is performed in step 3, for example in a base such as potassium t-butoxide and a solvent such as dimethylformamide).
  • the resulting compound 1Od is then coupled to pyrrolidine in the presence of a solvent such as 1,4-dioxane.
  • the pyrazolopyrimidine subunit is further derivatized via a reaction such as a Suzuki coupling (e.g. using Pd(PPh 3 ) 4 as a catalyst) to yield compound 1Of.
  • Scheme 11 exemplifies a synthesis of the compound Hf.
  • a starting aldehyde Ha is converted to an alcohol, for example, by Grignard addition of an alkylmagnesium halide reagent.
  • compound Hb is coupled to to an aryl group via, for example, a Suzuki coupling using phenylboronic acid in the presence of Pd(OAc) 2 , a base such as Na 2 CO 3 and a solvent such as ethanol/DMF/water.
  • the resulting compounds Hc is converted to a reactive halide by reaction with a reagent such as CBr 4 ZPPh 3 in a solvent such as acetonitrile.
  • a heterocyclic moiety such as a pyrazolopyrimide (e.g. 3-iodo-lH-pyrazolo[3,4-d]pyrimidin-4- amine) is performed in step 3, for example in a base such as potassium t-butoxide and a solvent such as dimethylformamide.
  • the resulting compound Hd is further derivatized via a reaction such as a Suzuki coupling (e.g. using Pd(PPh 3 ) 4 as a catalyst) to yield compound Hf.
  • Scheme 11 Synthesis of 3-(4-amino-l-(l-(2-phenylquinolin-3-yl)ethyl)-lH-pyrazolo[3,4-d]pyrimidin-3-yl)phenol
  • Scheme 12 shows a synthesis of the compound 12e.
  • a starting aldehyde 12a is converted to an alcohol, for example, by Grignard addition of an alkylmagnesium halide reagent.
  • compound 12b is converted to a reactive halide by reaction with a reagent such as CBr 4 ZPPh 3 in a solvent such as acetonitrile.
  • a heterocyclic compound such as 9H-purin-6-amine leads to formation of the intermediate 12d.
  • This intermediae is then coupled to an aryl group via, for example, a Suzuki coupling using phenylboronic acid in the presence of Pd(O Ac) 2 , a base such as Na 2 CO 3 and a solvent such as ethanol/DMF/water.
  • compound 12d is reacted with pyrrolidine in the presence of a solvent such as 1,4-dioxane, leading to formation of product 12f.
  • Scheme 13 describes the synthesis of additional compounds comprising heteroaromatic residues.
  • the starting material is the intermediate 12c prepared as described above. Coupling with a derivatized bicyclic heteroaromatic residue (in the example shown, 9-(tetrahydro-2H-pyran-2-yl)-9H-purin-6-amine) results in compound 13b, which is further reacted with a group such as pyrrolidine (shown) to yield compounds 13c. Deprotection of the purine moiety in HCl/ethanol results in compound 13d.
  • Scheme 13 Synthesis of N-Cl-Cl-Cpyrrolidin-l-ylJquinolin-S-yOethyO-PH-purin-o-ainine (13d)
  • Scheme 15 describes the synthesis of compounds such as 15g.
  • Intermediate 15d is prepared as described for compound 4b.
  • Treatment with NaOH in a solvent such as 1,4-dioxane at 8OC yields 15e, which is further alkylated using a reagent such as methyl iodide in potassium t-butoxide/DMF to yield 15f.
  • Compound 15g is obtained following Suzuki coupling as described above.
  • one or more subject compounds bind specifically to a PI3 kinase or a protein kinase selected from the group consisting of mTor, DNA-dependent protein kinase DNA-dependent protein kinase (Pubmed protein accession number (PPAN) AAA79184), AbI tyrosine kinase (CAA52387), Bcr-Abl, hemopoietic cell kinase (PPAN CAI19695), Src (PPAN CAA24495), vascular endothelial growth factor receptor 2 (PPAN ABB82619), vascular endothelial growth factor receptor-2 (PPAN ABB82619), epidermal growth factor receptor (PPAN AG43241), EPH receptor B4 (PPAN EAL23820), stem cell factor receptor (PPAN AAF22141), Tyrosine-protein kinase receptor TIE-2 (PPAN Q02858), fins-related tyrosine kinase 3 (
  • the IC50 of a subject compound for pi 10a, pi lO ⁇ , pi lO ⁇ , or pi 105 is less than about 1 uM, less than about 100 nM, less than about 50 nM, less than about 10 nM, less than 1 nM or even less than about 0.5nM. In some embodiments, the IC50 of a subject compound for mTor is less than about 1 uM, less than about 100 nM, less than about 50 nM, less than about 10 nM, less than 1 nM or even less than about 0.5nM.
  • one or more subject compounds exhibit dual binding specificity and are capable of inhibiting a PI3 kinase (e.g., a class I PI3 kinease) as well as a protein kinase (e.g., mTor) with an IC50 value less than about 1 uM, less than about 100 nM, less than about 50 nM, less than about 10 nM, less than 1 nM or even less than about 0.5 nM.
  • a PI3 kinase e.g., a class I PI3 kinease
  • protein kinase e.g., mTor
  • One or more subject compounds are capable of inhibiting tyrosine kinases including, for example, DNA-dependent protein kinase DNA-dependent protein kinase (Pubmed protein accession number (PPAN) AAA79184), AbI tyrosine kinase (CAA52387), Bcr-Abl, hemopoietic cell kinase (PPAN CAI19695), Src (PPAN CAA24495), vascular endothelial growth factor receptor 2 (PPAN ABB82619), vascular endothelial growth factor receptor-2 (PPAN ABB82619), epidermal growth factor receptor (PPAN AG43241), EPH receptor B4 (PPAN EAL23820), stem cell factor receptor (PPAN AAF22141), Tyrosine- protein kinase receptor TIE-2 (PPAN Q02858), fins-related tyrosine kinase 3 (PPAN NP 004110), platelet- derived growth factor receptor alpha (
  • the compounds of the present invention exhibits one or more functional characteristics disclosed herein.
  • one or more subject compounds bind specifically to a PI3 kinase.
  • the IC50 of a subject compound for pi 10a, pi lO ⁇ , pi lO ⁇ , or pl lO ⁇ is less than about 1 uM, less than about 100 nM, less than about 50 nM, less than about 10 nM, less than about 1 nM, less than about 0.5nM, less than about 10OpM, or less than about 50 pM.
  • one or more of the subject compounds may selectively inhibit one or more members of type I or class I phosphatidylinositol 3-kinases (PI3-kinase) with an IC50 value of about 100 nM, 50 nM, 10 nM, 5 nM, 100 pM, 10 pM or 1 pM, or less as measured in an in vitro kinase assay.
  • PI3-kinase phosphatidylinositol 3-kinases
  • one or more of the subject compound may selectively inhibit one or two members of type I or class I phosphatidylinositol 3-kinases (PI3-kinase) consisting of PI3-kinase ⁇ , PI3-kinase ⁇ , PI3-kinase ⁇ , and PI3-kinase ⁇ .
  • PI3-kinase phosphatidylinositol 3-kinases
  • some of the subject compounds selectively inhibit PI3-kinase ⁇ as compared to all other type I PI3-kinases.
  • some of the subject compounds selectively inhibit PI3-kinase ⁇ and PI3-kinase ⁇ as compared to the rest of the type I PI3-kinases.
  • some of the subject compounds selectively inhibit PI3-kinase ⁇ and PI3-kinase ⁇ as compared to the rest of the type I PI3-kinases. In still yet some other aspects, some of the subject compounds selectively inhibit PI3-kinase ⁇ and PI3-kinase ⁇ as compared to the rest of the type I PI3-kinases.
  • some of the subject compounds selectively inhibit PI3-kinase ⁇ and PI3-kinase ⁇ as compared to the rest of the type I PI3-kinases, or selectively inhibit PI3-kinase ⁇ and PI3-kinase ⁇ as compared to the rest of the type I PI3-kinases, or selectively inhibit PI3-kinase ⁇ and PI3-kinase ⁇ as compared to the rest of the type I PI3-kinases, or selectively inhibit PI3-kinase ⁇ and PI3-kinase ⁇ as compared to the rest of the type I PI3-kinases.
  • an inhibitor that selectively inhibits one or more members of type I PI3-kinases or an inhibitor that selectively inhibits one or more type I PI3-kinase mediated signaling pathways, alternatively can be understood to refer to a compound that exhibits a 50% inhibitory concentration (IC50) with respect to a given type I PI3-kinase, that is at least at least 10-fold, at least 20-fold, at least 50-fold, at least 100-fold, at least 1000- fold, at least 10,100-fold, or lower, than the inhibitor's IC50 with respect to the rest of the other type I PI3- kinases.
  • IC50 50% inhibitory concentration
  • the invention provides pharmaceutical compositions comprising one or more compounds of the present invention.
  • the invention provides pharmaceutical compositions for treating diseases or conditions related to an undesirable, over-active, harmful or deleterious immune response in a mammal.
  • Such undesirable immune response can be associated with or result in, e.g., asthma, emphysema, bronchitis, psoriasis, allergy, anaphylaxsis, auto-immune diseases, rhuematoid arthritis, graft versus host disease, and lupus erythematosus.
  • compositions of the present invention can be used to treat other respiratory diseases including but not limited to diseases affecting the lobes of lung, pleural cavity, bronchial tubes, trachea, upper respiratory tract, or the nerves and muscle for breathing.
  • disorders such as hyperproliferative disorder including but not limited to cancer such as acute myeloid leukemia, thymus, brain, lung, squamous cell, skin, eye, retinoblastoma, intraocular melanoma, oral cavity and oropharyngeal, bladder, gastric, stomach, pancreatic, bladder, breast, cervical, head, neck, renal, kidney, liver, ovarian, prostate, colorectal, esophageal, testicular, gynecological, thyroid, CNS, PNS, AIDS related AIDS-Related (e.g.
  • said pharmaceutical composition is for the treatment of a non-cancerous hyperproliferative disorder such as benign hyperplasia of the skin (e. g., psoriasis), restenosis, or prostate (e. g., benign prostatic hypertrophy (BPH)).
  • a non-cancerous hyperproliferative disorder such as benign hyperplasia of the skin (e. g., psoriasis), restenosis, or prostate (e. g., benign prostatic hypertrophy (BPH)).
  • the invention also provides compositions for the treatment of liver diseases (including diabetes), pancreatitis or kidney disease (including proliferative glomerulonephritis and diabetes- induced renal disease) or pain in a mammal.
  • liver diseases including diabetes
  • pancreatitis or kidney disease including proliferative glomerulonephritis and diabetes- induced renal disease
  • pain in a mammal.
  • the invention further provides a composition for the prevention of blastocyte implantation in a mammal.
  • the invention also relates to a composition for treating a disease related to vasculogenesis or angiogenesis in a mammal which can manifest as tumor angiogenesis, chronic inflammatory disease such as rheumatoid arthritis, inflammatory bowel disease, atherosclerosis, skin diseases such as psoriasis, eczema, and scleroderma, diabetes, diabetic retinopathy, retinopathy of prematurity, age-related macular degeneration, hemangioma, glioma, melanoma, Kaposi's sarcoma and ovarian, breast, lung, pancreatic, prostate, colon and epidermoid cancer.
  • chronic inflammatory disease such as rheumatoid arthritis, inflammatory bowel disease, atherosclerosis, skin diseases such as psoriasis, eczema, and scleroderma
  • diabetes diabetic retinopathy, retinopathy of prematurity
  • the subject pharmaceutical compositions are typically formulated to provide a therapeutically effective amount of a compound of the present invention as the active ingredient, or a pharmaceutically acceptable salt, ester, prodrug, solvate, hydrate or derivative thereof.
  • the pharmaceutical compositions contain pharmaceutically acceptable salt and/or coordination complex thereof, and one or more pharmaceutically acceptable excipients, carriers, including inert solid diluents and fillers, diluents, including sterile aqueous solution and various organic solvents, permeation enhancers, solubilizers and adjuvants.
  • the subject pharmaceutical compositions can be administered alone or in combination with one or more other agents, which are also typically administered in the form of pharmaceutical compositions.
  • the subject compounds and other agent(s) may be mixed into a preparation or both components may be formulated into separate preparations to use them in combination separately or at the same time.
  • the concentration of one or more of the compounds provided in the pharmaceutical compositions of the present invention is less than 100%, 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 19%, 18%, 17%, 16%, 15%,14%, 13%, 12%, 11%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.5%, 0.4%, 0.3%, 0.2%, 0.1%, 0.09%, 0.08%, 0.07%, 0.06%, 0.05%, 0.04%, 0.03%, 0.02%, 0.01%, 0.009%, 0.008%, 0.007%, 0.006%, 0.005%, 0.004%, 0.003%, 0.002%, 0.001%, 0.0009%, 0.0008%, 0.0007%, 0.0006%, 0.0005%, 0.0004%, 0.0003%, 0.0002%, or 0.0001% w/w, w/v or v/v.
  • the concentration of one or more of the compounds of the present invention is greater than 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 19.75%, 19.50%, 19.25% 19%, 18.75%, 18.50%, 18.25% 18%, 17.75%, 17.50%, 17.25% 17%, 16.75%, 16.50%, 16.25% 16%, 15.75%, 15.50%, 15.25% 15%, 14.75%, 14.50%, 14.25% 14%, 13.75%, 13.50%, 13.25% 13%, 12.75%, 12.50%, 12.25% 12%, 11.75%, 11.50%, 11.25% 11%, 10.75%, 10.50%, 10.25% 10%, 9.75%, 9.50%, 9.25% 9%, 8.75%, 8.50%, 8.25% 8%, 7.75%, 7.50%, 7.25% 7%, 6.75%, 6.50%, 6.25% 6%, 5.75%, 5.50%, 5.25% 5%, 4.75%,
  • the concentration of one or more of the compounds of the present invention is in the range from approximately 0.0001% to approximately 50%, approximately 0.001% to approximately 40 %, approximately 0.01% to approximately 30%, approximately 0.02% to approximately 29%, approximately 0.03% to approximately 28%, approximately 0.04% to approximately 27%, approximately 0.05% to approximately 26%, approximately 0.06% to approximately 25%, approximately 0.07% to approximately 24%, approximately 0.08% to approximately 23%, approximately 0.09% to approximately 22%, approximately 0.1% to approximately 21%, approximately 0.2% to approximately 20%, approximately 0.3% to approximately 19%, approximately 0.4% to approximately 18%, approximately 0.5% to approximately 17%, approximately 0.6% to approximately 16%, approximately 0.7% to approximately 15%, approximately 0.8% to approximately 14%, approximately 0.9% to approximately 12%, approximately 1% to approximately 10% w/w, w/v or v/v. v/v.
  • the concentration of one or more of the compounds of the present invention is in the range from approximately 0.001% to approximately 10%, approximately 0.01% to approximately 5%, approximately 0.02% to approximately 4.5%, approximately 0.03% to approximately 4%, approximately 0.04% to approximately 3.5%, approximately 0.05% to approximately 3%, approximately 0.06% to approximately 2.5%, approximately 0.07% to approximately 2%, approximately 0.08% to approximately 1.5%, approximately 0.09% to approximately 1%, approximately 0.1% to approximately 0.9% w/w, w/v or v/v.
  • the amount of one or more of the compounds of the present invention is equal to or less than 10 g, 9.5 g, 9.0 g, 8.5 g, 8.0 g, 7.5 g, 7.0 g, 6.5 g, 6.0 g, 5.5 g, 5.0 g, 4.5 g, 4.0 g, 3.5 g, 3.0 g, 2.5 g, 2.0 g, 1.5 g, 1.0 g, 0.95 g, 0.9 g, 0.85 g, 0.8 g, 0.75 g, 0.7 g, 0.65 g, 0.6 g, 0.55 g, 0.5 g, 0.45 g, 0.4 g, 0.35 g, 0.3 g, 0.25 g, 0.2 g, 0.15 g, 0.1 g, 0.09 g, 0.08 g, 0.07 g, 0.06 g, 0.05 g, 0.04 g, 0.03 g, 0.02 g, 0.01 g,
  • the amount of one or more of the compounds of the present invention is more than 0.0001 g, 0.0002 g, 0.0003 g, 0.0004 g, 0.0005 g, 0.0006 g, 0.0007 g, 0.0008 g, 0.0009 g, 0.001 g, 0.0015 g, 0.002 g, 0.0025 g, 0.003 g, 0.0035 g, 0.004 g, 0.0045 g, 0.005 g, 0.0055 g, 0.006 g, 0.0065 g, 0.007 g, 0.0075 g, 0.008 g, 0.0085 g, 0.009 g, 0.0095 g, 0.01 g, 0.015 g, 0.02 g, 0.025 g, 0.03 g, 0.035 g, 0.04 g, 0.045 g, 0.05 g, 0.055 g, 0.06 g, 0.065 g, 0.07
  • the amount of one or more of the compounds of the present invention is in the range of 0.0001-10 g, 0.0005-9 g, 0.001-8 g, 0.005-7 g, 0.01-6 g, 0.05-5 g, 0.1-4 g, 0.5-4 g, or 1-3 g.
  • the compounds according to the invention are effective over a wide dosage range.
  • dosages from 0.01 to 1000 mg, from 0.5 to 100 mg, from 1 to 50 mg per day, and from 5 to 40 mg per day are examples of dosages that may be used.
  • An exemplary dosage is 10 to 30 mg per day. The exact dosage will depend upon the route of administration, the form in which the compound is administered, the subject to be treated, the body weight of the subject to be treated, and the preference and experience of the attending physician.
  • compositions for oral administration In some embodiments, the invention provides a pharmaceutical composition for oral administration containing a compound of the present invention, and a pharmaceutical excipient suitable for oral administration.
  • the invention provides a solid pharmaceutical composition for oral administration containing: (i) an effective amount of a compound of the present invention; optionally (ii) an effective amount of a second agent; and (iii) a pharmaceutical excipient suitable for oral administration.
  • the composition further contains: (iv) an effective amount of a third agent.
  • the pharmaceutical composition may be a liquid pharmaceutical composition suitable for oral consumption.
  • Pharmaceutical compositions of the invention suitable for oral administration can be presented as discrete dosage forms, such as capsules, cachets, or tablets, or liquids or aerosol sprays each containing a predetermined amount of an active ingredient as a powder or in granules, a solution, or a suspension in an aqueous or non-aqueous liquid, an oil-in-water emulsion, or a water-in-oil liquid emulsion.
  • Such dosage forms can be prepared by any of the methods of pharmacy, but all methods include the step of bringing the active ingredient into association with the carrier, which constitutes one or more necessary ingredients.
  • compositions are prepared by uniformly and intimately admixing the active ingredient with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product into the desired presentation.
  • a tablet can be prepared by compression or molding, optionally with one or more accessory ingredients.
  • Compressed tablets can be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as powder or granules, optionally mixed with an excipient such as, but not limited to, a binder, a lubricant, an inert diluent, and/or a surface active or dispersing agent.
  • Molded tablets can be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
  • This invention further encompasses anhydrous pharmaceutical compositions and dosage forms comprising an active ingredient, since water can facilitate the degradation of some compounds.
  • water may be added (e.g., 5%) in the pharmaceutical arts as a means of simulating long-term storage in order to determine characteristics such as shelf-life or the stability of formulations over time.
  • Anhydrous pharmaceutical compositions and dosage forms of the invention can be prepared using anhydrous or low moisture containing ingredients and low moisture or low humidity conditions.
  • Pharmaceutical compositions and dosage forms of the invention which contain lactose can be made anhydrous if substantial contact with moisture and/or humidity during manufacturing, packaging, and/or storage is expected.
  • An anhydrous pharmaceutical composition may be prepared and stored such that its anhydrous nature is maintained.
  • anhydrous compositions may be packaged using materials known to prevent exposure to water such that they can be included in suitable formulary kits.
  • suitable packaging include, but are not limited to, hermetically sealed foils, plastic or the like, unit dose containers, blister packs, and strip packs.
  • An active ingredient can be combined in an intimate admixture with a pharmaceutical carrier according to conventional pharmaceutical compounding techniques.
  • the carrier can take a wide variety of forms depending on the form of preparation desired for administration.
  • any of the usual pharmaceutical media can be employed as carriers, such as, for example, water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents, and the like in the case of oral liquid preparations (such as suspensions, solutions, and elixirs) or aerosols; or carriers such as starches, sugars, micro-crystalline cellulose, diluents, granulating agents, lubricants, binders, and disintegrating agents can be used in the case of oral solid preparations, in some embodiments without employing the use of lactose.
  • suitable carriers include powders, capsules, and tablets, with the solid oral preparations. If desired, tablets can be coated by standard aqueous or nonaqueous techniques.
  • Binders suitable for use in pharmaceutical compositions and dosage forms include, but are not limited to, corn starch, potato starch, or other starches, gelatin, natural and synthetic gums such as acacia, sodium alginate, alginic acid, other alginates, powdered tragacanth, guar gum, cellulose and its derivatives (e.g., ethyl cellulose, cellulose acetate, carboxymethyl cellulose calcium, sodium carboxymethyl cellulose), polyvinyl pyrrolidone, methyl cellulose, pre-gelatinized starch, hydroxypropyl methyl cellulose, microcrystalline cellulose, and mixtures thereof.
  • natural and synthetic gums such as acacia, sodium alginate, alginic acid, other alginates, powdered tragacanth, guar gum, cellulose and its derivatives (e.g., ethyl cellulose, cellulose acetate, carboxymethyl cellulose calcium, sodium carboxymethyl cellulose), polyvinyl pyrrol
  • suitable fillers for use in the pharmaceutical compositions and dosage forms disclosed herein include, but are not limited to, talc, calcium carbonate (e.g., granules or powder), microcrystalline cellulose, powdered cellulose, dextrates, kaolin, mannitol, silicic acid, sorbitol, starch, pre-gelatinized starch, and mixtures thereof.
  • Disintegrants may be used in the compositions of the invention to provide tablets that disintegrate when exposed to an aqueous environment. Too much of a disintegrant may produce tablets which may disintegrate in the bottle. Too little may be insufficient for disintegration to occur and may thus alter the rate and extent of release of the active ingredient(s) from the dosage form. Thus, a sufficient amount of disintegrant that is neither too little nor too much to detrimentally alter the release of the active ingredient(s) may be used to form the dosage forms of the compounds disclosed herein. The amount of disintegrant used may vary based upon the type of formulation and mode of administration, and may be readily discernible to those of ordinary skill in the art.
  • Disintegrants that can be used to form pharmaceutical compositions and dosage forms of the invention include, but are not limited to, agar-agar, alginic acid, calcium carbonate, microcrystalline cellulose, croscarmellose sodium, crospovidone, polacrilin potassium, sodium starch glycolate, potato or tapioca starch, other starches, pre-gelatinized starch, other starches, clays, other algins, other celluloses, gums or mixtures thereof.
  • Lubricants which can be used to form pharmaceutical compositions and dosage forms of the invention include, but are not limited to, calcium stearate, magnesium stearate, mineral oil, light mineral oil, glycerin, sorbitol, mannitol, polyethylene glycol, other glycols, stearic acid, sodium lauryl sulfate, talc, hydrogenated vegetable oil (e.g., peanut oil, cottonseed oil, sunflower oil, sesame oil, olive oil, corn oil, and soybean oil), zinc stearate, ethyl oleate, ethylaureate, agar, or mixtures thereof.
  • Additional lubricants include, for example, a syloid silica gel, a coagulated aerosol of synthetic silica, or mixtures thereof.
  • a lubricant can optionally be added, in an amount of less than about 1 weight percent of the pharmaceutical composition.
  • the essential active ingredient therein may be combined with various sweetening or flavoring agents, coloring matter or dyes and, if so desired, emulsifying and/or suspending agents, together with such diluents as water, ethanol, propylene glycol, glycerin and various combinations thereof.
  • the tablets can be uncoated or coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period.
  • a time delay material such as glyceryl monostearate or glyceryl distearate can be employed.
  • Formulations for oral use can also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example, peanut oil, liquid paraffin or olive oil.
  • Surfactant which can be used to form pharmaceutical compositions and dosage forms of the invention include, but are not limited to, hydrophilic surfactants, lipophilic surfactants, and mixtures thereof. That is, a mixture of hydrophilic surfactants may be employed, a mixture of lipophilic surfactants may be employed, or a mixture of at least one hydrophilic surfactant and at least one lipophilic surfactant may be employed.
  • a suitable hydrophilic surfactant may generally have an HLB value of at least 10, while suitable lipophilic surfactants may generally have an HLB value of or less than about 10.
  • An empirical parameter used to characterize the relative hydrophilicity and hydrophobicity of non-ionic amphiphilic compounds is the hydrophilic-lipophilic balance ("HLB" value).
  • HLB hydrophilic-lipophilic balance
  • Surfactants with lower HLB values are more lipophilic or hydrophobic, and have greater solubility in oils, while surfactants with higher HLB values are more hydrophilic, and have greater solubility in aqueous solutions.
  • Hydrophilic surfactants are generally considered to be those compounds having an HLB value greater than about 10, as well as anionic, cationic, or zwitterionic compounds for which the HLB scale is not generally applicable.
  • lipophilic (i.e., hydrophobic) surfactants are compounds having an HLB value equal to or less than about 10.
  • HLB value of a surfactant is merely a rough guide generally used to enable formulation of industrial, pharmaceutical and cosmetic emulsions.
  • Hydrophilic surfactants may be either ionic or non-ionic. Suitable ionic surfactants include, but are not limited to, alkylammonium salts; fusidic acid salts; fatty acid derivatives of amino acids, oligopeptides, and polypeptides; glyceride derivatives of amino acids, oligopeptides, and polypeptides; lecithins and hydrogenated lecithins; lysolecithins and hydrogenated lysolecithins; phospholipids and derivatives thereof; lysophospholipids and derivatives thereof; carnitine fatty acid ester salts; salts of alkylsulfates; fatty acid salts; sodium docusate; acylactylates; mono- and di-acetylated tartaric acid esters of mono- and di-glycerides; succinylated mono- and di-glycerides; citric acid esters of mono- and di-glycerides
  • ionic surfactants include, by way of example: lecithins, lysolecithin, phospholipids, lysophospholipids and derivatives thereof; carnitine fatty acid ester salts; salts of alkylsulfates; fatty acid salts; sodium docusate; acylactylates; mono- and di-acetylated tartaric acid esters of mono- and di- glycerides; succinylated mono- and di-glycerides; citric acid esters of mono- and di-glycerides; and mixtures thereof.
  • Ionic surfactants may be the ionized forms of lecithin, lysolecithin, phosphatidylcholine, phosphatidylethanolamine, phosphatidylglycerol, phosphatide acid, phosphatidylserine, lysophosphatidylcholine, lysophosphatidylethanolamine, lysophosphatidylglycerol, lysophosphatidic acid, lysophosphatidylserine, PEG-phosphatidylethanolamine, PVP-phosphatidylethanolamine, lactylic esters of fatty acids, stearoyl-2-lactylate, stearoyl lactylate, succinylated monoglycerides, mono/diacetylated tartaric acid esters of mono/diglycerides, citric acid esters of mono/diglycerides, cholylsarcosine, caproate, caprylate,
  • Hydrophilic non-ionic surfactants may include, but not limited to, alkylglucosides; alkylmaltosides; alkylthioglucosides; lauryl macrogolglycerides; polyoxyalkylene alkyl ethers such as polyethylene glycol alkyl ethers; polyoxyalkylene alkylphenols such as polyethylene glycol alkyl phenols; polyoxyalkylene alkyl phenol fatty acid esters such as polyethylene glycol fatty acids monoesters and polyethylene glycol fatty acids diesters; polyethylene glycol glycerol fatty acid esters; polyglycerol fatty acid esters; polyoxyalkylene sorbitan fatty acid esters such as polyethylene glycol sorbitan fatty acid esters; hydrophilic transesterification products of a polyol with at least one member of the group consisting of glycerides, vegetable oils, hydrogenated vegetable oils, fatty acids, and sterols; polyoxyethylene sterol
  • hydrophilic-non-ionic surfactants include, without limitation, PEG-IO laurate, PEG-12 laurate, PEG-20 laurate, PEG-32 laurate, PEG-32 dilaurate, PEG-12 oleate, PEG- 15 oleate, PEG-20 oleate, PEG-20 dioleate, PEG-32 oleate, PEG-200 oleate, PEG-400 oleate, PEG- 15 stearate, PEG-32 distearate, PEG-40 stearate, PEG- 100 stearate, PEG-20 dilaurate, PEG-25 glyceryl trioleate, PEG-32 dioleate, PEG-20 glyceryl laurate, PEG-30 glyceryl laurate, PEG-20 glyceryl stearate, PEG-20 glyceryl oleate, PEG-30 glyceryl oleate, PEG-30 glyceryl
  • Suitable lipophilic surfactants include, by way of example only: fatty alcohols; glycerol fatty acid esters; acetylated glycerol fatty acid esters; lower alcohol fatty acids esters; propylene glycol fatty acid esters; sorbitan fatty acid esters; polyethylene glycol sorbitan fatty acid esters; sterols and sterol derivatives; polyoxyethylated sterols and sterol derivatives; polyethylene glycol alkyl ethers; sugar esters; sugar ethers; lactic acid derivatives of mono- and di-glycerides; hydrophobic transesterification products of a polyol with at least one member of the group consisting of glycerides, vegetable oils, hydrogenated vegetable oils, fatty acids and sterols; oil-soluble vitamins/vitamin derivatives; and mixtures thereof.
  • preferred lipophilic surfactants include glycerol fatty acid esters, propylene glycol fatty acid esters, and mixtures thereof, or are hydrophobic transesterification products of a polyol with at least one member of the group consisting of vegetable oils, hydrogenated vegetable oils, and triglycerides.
  • the composition may include a solubilizer to ensure good solubilization and/or dissolution of the compound of the present invention and to minimize precipitation of the compound of the present invention. This can be especially important for compositions for non-oral use, e.g., compositions for injection.
  • a solubilizer may also be added to increase the solubility of the hydrophilic drug and/or other components, such as surfactants, or to maintain the composition as a stable or homogeneous solution or dispersion.
  • solubilizers include, but are not limited to, the following: alcohols and polyols, such as ethanol, isopropanol, butanol, benzyl alcohol, ethylene glycol, propylene glycol, butanediols and isomers thereof, glycerol, pentaerythritol, sorbitol, mannitol, transcutol, dimethyl isosorbide, polyethylene glycol, polypropylene glycol, polyvinylalcohol, hydroxypropyl methylcellulose and other cellulose derivatives, cyclodextrins and cyclodextrin derivatives; ethers of polyethylene glycols having an average molecular weight of about 200 to about 6000, such as tetrahydrofurfuryl alcohol PEG ether (glycofurol) or methoxy PEG ; amides and other nitrogen-containing compounds such as 2-pyrrolidone, 2-piperidone,
  • solubilizers may also be used. Examples include, but not limited to, triacetin, triethylcitrate, ethyl oleate, ethyl caprylate, dimethylacetamide, N-methylpyrrolidone, N-hydroxyethylpyrrolidone, polyvinylpyrrolidone, hydroxypropyl methylcellulose, hydroxypropyl cyclodextrins, ethanol, polyethylene glycol 200-100, glycofurol, transcutol, propylene glycol, and dimethyl isosorbide. Particularly preferred solubilizers include sorbitol, glycerol, triacetin, ethyl alcohol, PEG-400, glycofurol and propylene glycol.
  • the amount of solubilizer that can be included is not particularly limited.
  • the amount of a given solubilizer may be limited to a bioacceptable amount, which may be readily determined by one of skill in the art.
  • the solubilizer can be in a weight ratio of 10%, 25%, 50%, 100%, or up to about 200% by weight, based on the combined weight of the drug, and other excipients.
  • very small amounts of solubilizer may also be used, such as 5%, 2%, 1% or even less.
  • the solubilizer may be present in an amount of about 1% to about 100%, more typically about 5% to about 25% by weight.
  • the composition can further include one or more pharmaceutically acceptable additives and excipients.
  • additives and excipients include, without limitation, detackifiers, anti-foaming agents, buffering agents, polymers, antioxidants, preservatives, chelating agents, viscomodulators, tonicifiers, flavorants, colorants, odorants, opacifiers, suspending agents, binders, fillers, plasticizers, lubricants, and mixtures thereof.
  • an acid or a base may be incorporated into the composition to facilitate processing, to enhance stability, or for other reasons.
  • pharmaceutically acceptable bases include amino acids, amino acid esters, ammonium hydroxide, potassium hydroxide, sodium hydroxide, sodium hydrogen carbonate, aluminum hydroxide, calcium carbonate, magnesium hydroxide, magnesium aluminum silicate, synthetic aluminum silicate, synthetic hydrocalcite, magnesium aluminum hydroxide, diisopropylethylamine, ethanolamine, ethylenediamine, triethanolamine, triethylamine, triisopropanolamine, trimethylamine, tris(hydroxymethyl)aminomethane (TRIS) and the like.
  • bases that are salts of a pharmaceutically acceptable acid, such as acetic acid, acrylic acid, adipic acid, alginic acid, alkanesulfonic acid, amino acids, ascorbic acid, benzoic acid, boric acid, butyric acid, carbonic acid, citric acid, fatty acids, formic acid, fumaric acid, gluconic acid, hydroquinosulfonic acid, isoascorbic acid, lactic acid, maleic acid, oxalic acid, para-bromophenylsulfonic acid, propionic acid, p-toluenesulfonic acid, salicylic acid, stearic acid, succinic acid, tannic acid, tartaric acid, thioglycolic acid, toluenesulfonic acid, uric acid, and the like.
  • a pharmaceutically acceptable acid such as acetic acid, acrylic acid, adipic acid, alginic acid, alkanesulfonic acid, amino acids
  • Salts of polyprotic acids such as sodium phosphate, disodium hydrogen phosphate, and sodium dihydrogen phosphate can also be used.
  • the cation can be any convenient and pharmaceutically acceptable cation, such as ammonium, alkali metals, alkaline earth metals, and the like.
  • Example may include, but not limited to, sodium, potassium, lithium, magnesium, calcium and ammonium.
  • Suitable acids are pharmaceutically acceptable organic or inorganic acids. Examples of suitable inorganic acids include hydrochloric acid, hydrobromic acid, hydriodic acid, sulfuric acid, nitric acid, boric acid, phosphoric acid, and the like.
  • suitable organic acids include acetic acid, acrylic acid, adipic acid, alginic acid, alkanesulfonic acids, amino acids, ascorbic acid, benzoic acid, boric acid, butyric acid, carbonic acid, citric acid, fatty acids, formic acid, fumaric acid, gluconic acid, hydroquinosulfonic acid, isoascorbic acid, lactic acid, maleic acid, methanesulfonic acid, oxalic acid, para-bromophenylsulfonic acid, propionic acid, p- toluenesulfonic acid, salicylic acid, stearic acid, succinic acid, tannic acid, tartaric acid, thioglycolic acid, toluenesulfonic acid, uric acid and the like.
  • compositions for injection containing a compound of the present invention and a pharmaceutical excipient suitable for injection.
  • Components and amounts of agents in the compositions are as described herein.
  • the forms in which the novel compositions of the present invention may be incorporated for administration by injection include aqueous or oil suspensions, or emulsions, with sesame oil, corn oil, cottonseed oil, or peanut oil, as well as elixirs, mannitol, dextrose, or a sterile aqueous solution, and similar pharmaceutical vehicles.
  • Aqueous solutions in saline are also conventionally used for injection.
  • Ethanol, glycerol, propylene glycol, liquid polyethylene glycol, and the like (and suitable mixtures thereof), cyclodextrin derivatives, and vegetable oils may also be employed.
  • the proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, for the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • the prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like.
  • Sterile injectable solutions are prepared by incorporating the compound of the present invention in the required amount in the appropriate solvent with various other ingredients as enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above.
  • certain desirable methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • compositions for topical e.g.. transdermal
  • the invention provides a pharmaceutical composition for transdermal delivery containing a compound of the present invention and a pharmaceutical excipient suitable for transdermal delivery.
  • compositions of the present invention can be formulated into preparations in solid, semi-solid, or liquid forms suitable for local or topical administration, such as gels, water soluble jellies, creams, lotions, suspensions, foams, powders, slurries, ointments, solutions, oils, pastes, suppositories, sprays, emulsions, saline solutions, dimethylsulfoxide (DMSO)-based solutions.
  • DMSO dimethylsulfoxide
  • carriers with higher densities are capable of providing an area with a prolonged exposure to the active ingredients.
  • a solution formulation may provide more immediate exposure of the active ingredient to the chosen area.
  • compositions also may comprise suitable solid or gel phase carriers or excipients, which are compounds that allow increased penetration of, or assist in the delivery of, therapeutic molecules across the stratum corneum permeability barrier of the skin.
  • suitable solid or gel phase carriers or excipients which are compounds that allow increased penetration of, or assist in the delivery of, therapeutic molecules across the stratum corneum permeability barrier of the skin.
  • penetration-enhancing molecules known to those trained in the art of topical formulation.
  • humectants e.g., urea
  • glycols e.g., propylene glycol
  • alcohols e.g., ethanol
  • fatty acids e.g., oleic acid
  • surfactants e.g., isopropyl myristate and sodium lauryl sulfate
  • pyrrolidones e.g., isopropyl myristate and sodium lauryl sulfate
  • pyrrolidones e.glycerol monolaurate, sulfoxides, terpenes (e.g., menthol)
  • amines amides, alkanes, alkanols, water, calcium carbonate, calcium phosphate, various sugars, starches, cellulose derivatives, gelatin, and polymers such as polyethylene glycols.
  • transdermal delivery devices patches
  • Such transdermal patches may be used to provide continuous or discontinuous infusion of a compound of the present invention in controlled amounts, either with or without another agent.
  • transdermal patches for the delivery of pharmaceutical agents is well known in the art. See, e.g., U.S. Pat. Nos. 5,023,252, 4,992,445 and 5,001,139. Such patches may be constructed for continuous, pulsatile, or on demand delivery of pharmaceutical agents.
  • compositions for inhalation include solutions and suspensions in pharmaceutically acceptable, aqueous or organic solvents, or mixtures thereof, and powders.
  • the liquid or solid compositions may contain suitable pharmaceutically acceptable excipients as described supra.
  • the compositions are administered by the oral or nasal respiratory route for local or systemic effect.
  • Compositions in preferably pharmaceutically acceptable solvents may be nebulized by use of inert gases. Nebulized solutions may be inhaled directly from the nebulizing device or the nebulizing device may be attached to a face mask tent, or intermittent positive pressure breathing machine. Solution, suspension, or powder compositions may be administered, preferably orally or nasally, from devices that deliver the formulation in an appropriate manner.
  • compositions may also be prepared from compositions described herein and one or more pharmaceutically acceptable excipients suitable for sublingual, buccal, rectal, intraosseous, intraocular, intranasal, epidural, or intraspinal administration. Preparations for such pharmaceutical compositions are well-known in the art.
  • Administration of the compounds or pharmaceutical composition of the present invention can be effected by any method that enables delivery of the compounds to the site of action. These methods include oral routes, intraduodenal routes, parenteral injection (including intravenous, intraarterial, subcutaneous, intramuscular, intravascular, intraperitoneal or infusion), topical (e.g. transdermal application), rectal administration, via local delivery by catheter or stent or through inhalation. Compounds can also abe administered intraadiposally or intrathecally.
  • the amount of the compound administered will be dependent on the mammal being treated, the severity of the disorder or condition, the rate of administration, the disposition of the compound and the discretion of the prescribing physician.
  • an effective dosage is in the range of about 0.001 to about 100 mg per kg body weight per day, preferably about 1 to about 35 mg/kg/day, in single or divided doses. For a 70 kg human, this would amount to about 0.05 to 7 g/day, preferably about 0.05 to about 2.5 g/day.
  • dosage levels below the lower limit of the aforesaid range may be more than adequate, while in other cases still larger doses may be employed without causing any harmful side effect, e.g. bydividing such larger doses into several small doses for administration throughout the day.
  • a compound of the invention is administered in a single dose.
  • administration will be by injection, e.g., intravenous injection, in order to introduce the agent quickly.
  • other routes may be used as appropriate.
  • a single dose of a compound of the invention may also be used for treatment of an acute condition.
  • a compound of the invention is administered in multiple doses. Dosing may be about once, twice, three times, four times, five times, six times, or more than six times per day. Dosing may be about once a month, once every two weeks, once a week, or once every other day. In another embodiment a compound of the invention and another agent are administered together about once per day to about 6 times per day. In another embodiment the administration of a compound of the invention and an agent continues for less than about 7 days. In yet another embodiment the administration continues for more than about 6, 10, 14, 28 days, two months, six months, or one year. In some cases, continuous dosing is achieved and maintained as long as necessary.
  • an agent of the invention is administered for more than 1, 2, 3, 4, 5, 6, 7, 14, or 28 days. In some embodiments, an agent of the invention is administered for less than 28, 14, 7, 6, 5, 4, 3, 2, or 1 day. In some embodiments, an agent of the invention is administered chronically on an ongoing basis, e.g., for the treatment of chronic effects.
  • An effective amount of a compound of the invention may be administered in either single or multiple doses by any of the accepted modes of administration of agents having similar utilities, including rectal, buccal, intranasal and transdermal routes, by intra-arterial injection, intravenously, intraperitoneally, parenterally, intramuscularly, subcutaneously, orally, topically, or as an inhalant.
  • compositions of the invention may also be delivered via an impregnated or coated device such as a stent, for example, or an artery-inserted cylindrical polymer.
  • a method of administration may, for example, aid in the prevention or amelioration of restenosis following procedures such as balloon angioplasty.
  • compounds of the invention may slow or inhibit the migration and proliferation of smooth muscle cells in the arterial wall which contribute to restenosis.
  • a compound of the invention may be administered, for example, by local delivery from the struts of a stent, from a stent graft, from grafts, or from the cover or sheath of a stent.
  • a compound of the invention is admixed with a matrix.
  • Such a matrix may be a polymeric matrix, and may serve to bond the compound to the stent.
  • Polymeric matrices suitable for such use include, for eample, lactone-based polyesters or copolyesters such as polylactide, polycaprolactonglycolide, polyorthoesters, polyanhydrides, polyaminoacids, polysaccharides, polyphosphazenes, poly (ether-ester) copolymers (e.g. PEO-PLLA); polydimethylsiloxane, poly(ethylene- vinylacetate), acrylate-based polymers or copolymers (e.g.
  • Compounds of the invention may be applied to the surface of the stent by various methods such as dip/spin coating, spray coating, dip- coating, and/or brush-coating.
  • the compounds may be applied in a solvent and the solvent may be allowed to evaporate, thus forming a layer of compound onto the stent.
  • the compound may be located in the body of the stent or graft, for example in microchannels or micropores.
  • stents When implanted, the compound diffuses out of the body of the stent to contact the arterial wall.
  • stents may be prepared by dipping a stent manufactured to contain such micropores or microchannels into a solution of the compound of the invention in a suitable solvent, followed by evaporation of the solvent. Excess drug on the surface of the stent may be removed via an additional brief solvent wash.
  • compounds of the invention may be covalently linked to a stent or graft.
  • a covalent linker may be used which degrades in vivo, leading to the release of the compound of the invention. Any bio- labile linkage may be used for such a purpose, such as ester, amide or anhydride linkages.
  • Compounds of the invention may additionally be administered intravascularly from a balloon used during angioplasty. Extravascular administration of the compounds via the pericard or via advential application of formulations of the invention may also be performed to decrease restenosis.
  • the compounds of the invention may be administered in dosages. It is known in the art that due to intersubject variability in compound pharmacokinetics, individualization of dosing regimen is necessary for optimal therapy. Dosing for a compound of the invention may be found by routine experimentation in light of the instant disclosure.
  • the subject pharmaceutical composition may, for example, be in a form suitable for oral administration as a tablet, capsule, pill, powder, sustained release formulations, solution, suspension, for parenteral injection as a sterile solution, suspension or emulsion, for topical administration as an ointment or cream or for rectal administration as a suppository.
  • the pharmaceutical composition may be in unit dosage forms suitable for single administration of precise dosages.
  • the pharmaceutical composition will include a conventional pharmaceutical carrier or excipient and a compound according to the invention as an active ingredient. In addition, it may include other medicinal or pharmaceutical agents, carriers, adjuvants, etc.
  • Exemplary parenteral administration forms include solutions or suspensions of active compound in sterile aqueous solutions, for example, aqueous propylene glycol or dextrose solutions. Such dosage forms can be suitably buffered, if desired.
  • the activity of the compounds of the present invention may be determined by the following procedure, as well as the procedure described in the examples below.
  • the activity of the kinase is assessed by measuring the incorporation of ⁇ - 33 P- ⁇ hosphate from ⁇ - 33 P-ATP onto N-terminal His tagged substrate, which is expressed in E. coli and is purified by conventional methods, in the presence of the kinase.
  • the assay is carried out in 96- well polypropylene plate.
  • the incubation mixture (100, ⁇ L) comprises of 25 mM Hepes, pH 7.4, 10 mM MgCl 2 , 5 mM ⁇ -glycerolphosphate, 100 ⁇ M Na-orthovanadate, 5 mM DTT, 5 nM kinase, and 1 ⁇ M substrate.
  • Inhibitors are suspended in DMSO, and all reactions, including controls are performed at a final concentration of 1% DMSO. Reactions are initiated by the addition of 10 ⁇ M ATP (with 0.5 ⁇ Ci ⁇ - 33 P- ATP/well) and incubated at ambient temperature for 45 minutes. Equal volume of 25% TCA is added to stop the reaction and precipitate the proteins.
  • kits include a compound or compounds of the present invention as described herein, in suitable packaging, and written material that can include instructions for use, discussion of clinical studies, listing of side effects, and the like.
  • kits may also include information, such as scientific literature references, package insert materials, clinical trial results, and/or summaries of these and the like, which indicate or establish the activities and/or advantages of the composition, and/or which describe dosing, administration, side effects, drug interactions, or other information useful to the health care provider. Such information may be based on the results of various studies, for example, studies using experimental animals involving in vivo models and studies based on human clinical trials.
  • the kit may further contain another agent.
  • the compound of the present invention and the agent are provided as separate compositions in separate containers within the kit.
  • the compound of the present invention and the agent are provided as a single composition within a container in the kit.
  • Kits described herein can be provided, marketed and/or promoted to health providers, including physicians, nurses, pharmacists, formulary officials, and the like. Kits may also, in some embodiments, be marketed directly to the consumer.
  • the invention also provides methods of using the compounds or pharmaceutical compositions of the present invention to treat disease conditions, including but not limited to diseases associated with malfunctioning of one or more types of PD kinase.
  • diseases associated with malfunctioning of one or more types of PD kinase include but not limited to diseases associated with malfunctioning of one or more types of PD kinase.
  • a detailed description of conditions and disorders mediated by pi lO ⁇ kinase activity is set forth in Sadu et al., WO 01/81346, which is incorporated herein by reference in its entirety for all purposes.
  • the treatment methods provided herein comprise administering to the subject a therapeutically effective amount of a compound of the invention.
  • the present invention provides a method of treating an inflammation disorder, including autoimmune diseases in a mammal. The method comprises administering to said mammal a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt, ester, prodrug, solvate, hydrate or derivative thereof.
  • autoimmune diseases includes but is not limited to acute disseminated encephalomyelitis (ADEM), Addison's disease, antiphospholipid antibody syndrome (APS), aplastic anemia, autoimmune hepatitis, coeliac disease, Crohn's disease, Diabetes mellitus (type 1), Goodpasture's syndrome, Graves' disease, Guillain-Barr ⁇ syndrome (GBS), Hashimoto's disease, lupus erythematosus, multiple sclerosis, myasthenia gravis, opsoclonus myoclonus syndrome (OMS), optic neuritis, Ord's thyroiditis, oemphigus, polyarthritis, primary biliary cirrhosis, psoriasis, rheumatoid arthritis, Reiter's syndrome, Takayasu's arteritis, temporal arteritis (also known as "giant cell arteritis”), warm autoimmune hemolytic anemia, Wegener
  • the method of treating inflammatory or autoimmune diseases comprises administering to a subject (e.g. a mammal) a therapeutically effective amount of one or more compounds of the present invention that selectively inhibit PDK- ⁇ and/or PI3K- ⁇ as compared to all other type I PI3 kinases.
  • a subject e.g. a mammal
  • Such selective inhibition of PI3K- ⁇ and/or PI3K- ⁇ may be advantageous for treating any of the diseases or conditions described herein.
  • selective inhibition of PI3K.-5 may inhibit inflammatory responses associated with inflammatory diseases, autoimmune disease, or diseases related to an undesirable immune response including but not limited to asthma, emphysema, allergy, dermatitis, rhuematoid arthritis, psoriasis, lupus erythematosus, or graft versus host disease.
  • Selective inhibition of PI3K-6 may further provide for a reduction in the inflammatory or undesirable immune response without a concomittant reduction in the ability to reduce a bacterial, viral, and/or fungal infection.
  • one or more of the subject methods are effective in reducing antigen specific antibody production in vivo by about 2-fold, 3-fold, 4-fold, 5-fold, 7.5- fold, 10-fold, 25-fold, 50-fold, 100-fold, 250-fold, 500-fold, 750-fold, or about 1000-fold or more.
  • one or more of the subject methods are effective in reducing antigen specific IgG3 and/or IgGM production in vivo by about 2-fold, 3-fold, 4-fold, 5-fold, 7.5-fold, 10-fold, 25-fold, 50-fold, 100-fold, 250-fold, 500-fold, 750-fold, or about 1000-fold or more.
  • one of more of the subject methods are effective in ameliorating symptoms assoicated with rhuematoid arthritis including but not limited to a reduction in the swelling of joints, a reduction in serum anti- collagen levels, and/or a reduction in joint pathology such as bone resorption, cartilage damage, pannus, and/or inflammation.
  • the subject methods are effective in reducing ankle inflammation by at least about 2%, 5%, 10%, 15%, 20%, 25%, 30%, 50%, 60%, or about 75% to 90%.
  • the subject methods are effective in reducing knee inflammation by at least about 2%, 5%, 10%, 15%, 20%, 25%, 30%, 50%, 60%, or about 75% to 90% or more.
  • the subject methods are effective in reducing serum anti-type II collagen levels by at least about 10%, 12%, 15%, 20%, 24%, 25%, 30%, 35%, 50%, 60%, 75%, 80%, 86%, 87%, or about 90% or more.
  • the subject methods are effective in reducing ankle histopathology scores by about 5%, 10%, 15%, 20%, 25%, 30%, 40%, 50%, 60%, 75%, 80%, 90% or more.
  • the subject methods are effective in reducing knee histopathology scores by about 5%, 10%, 15%, 20%, 25%, 30%, 40%, 50%, 60%, 75%, 80%, 90% or more.
  • the present invention provides methods of using the compounds or pharmaceutical compositions to treat respiratory diseases including but not limited to diseases affecting the lobes of lung, pleural cavity, bronchial tubes, trachea, upper respiratory tract, or the nerves and muscle for breathing.
  • respiratory diseases including but not limited to diseases affecting the lobes of lung, pleural cavity, bronchial tubes, trachea, upper respiratory tract, or the nerves and muscle for breathing.
  • methods are provided to treat obstructive pulmonary disease.
  • COPD chronic obstructive pulmonary disease
  • COPD chronic obstructive pulmonary disease
  • the compounds described herein are used for the treatment of asthma.
  • the compounds or pharmaceutical compositions described herein may be used for the treatment of endotoxemia and sepsis.
  • the compounds or pharmaceutical compositions described herein are used to for the treatment of rheumatoid arthritis (RA).
  • the compounds or pharmaceutical compositions described herein is used for the treatment of contact or atopic dermatitis.
  • Contact dermatitis includes irritant dermatitis, phototoxic dermatitis, allergic dermatitis, photoallergic dermatitis, contact urticaria, systemic contact-type dermatitis and the like.
  • Irritant dermatitis can occur when too much of a substance is used on the skin of when the skin is sensitive to certain substance.
  • Atopic dermatitis sometimes called eczema, is a kind of dermatitis, an atopic skin disease.
  • the invention also relates to a method of treating a hyperproliferative disorder in a mammal that comprises administering to said mammal a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt, ester, prodrug, solvate, hydrate or derivative thereof.
  • said method relates to the treatment of cancer such as acute myeloid leukemia, thymus, brain, lung, squamous cell, skin, eye, retinoblastoma, intraocular melanoma, oral cavity and oropharyngeal, bladder, gastric, stomach, pancreatic, bladder, breast, cervical, head, neck, renal, kidney, liver, ovarian, prostate, colorectal, esophageal, testicular, gynecological, thyroid, CNS, PNS, AIDS-related (e.g. Lymphoma and Kaposi's Sarcoma) or viral- induced cancer.
  • cancer such as acute myeloid leukemia, thymus, brain, lung, squamous cell, skin, eye, retinoblastoma, intraocular melanoma, oral cavity and oropharyngeal, bladder, gastric, stomach, pancreatic, bladder, breast, cervical, head, neck, renal, kidney, liver, ovarian, prostate
  • said method relates to the treatment of a non-cancerous hyperproliferative disorder such as benign hyperplasia of the skin (e. g., psoriasis), restenosis, or prostate (e. g., benign prostatic hypertrophy (BPH)).
  • a non-cancerous hyperproliferative disorder such as benign hyperplasia of the skin (e. g., psoriasis), restenosis, or prostate (e. g., benign prostatic hypertrophy (BPH)).
  • the invention also relates to a method of treating diseases related to vasculogenesis or angiogenesis in a mammal that comprises administering to said mammal a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt, ester, prodrug, solvate, hydrate or derivative thereof.
  • said method is for treating a disease selected from the group consisting of tumor angiogenesis, chronic inflammatory disease such as rheumatoid arthritis, atherosclerosis, inflammatory bowel disease, skin diseases such as psoriasis, eczema, and scleroderma, diabetes, diabetic retinopathy, retinopathy of prematurity, age-related macular degeneration, hemangioma, glioma, melanoma, Kaposi's sarcoma and ovarian, breast, lung, pancreatic, prostate, colon and epidermoid cancer.
  • a disease selected from the group consisting of tumor angiogenesis, chronic inflammatory disease such as rheumatoid arthritis, atherosclerosis, inflammatory bowel disease, skin diseases such as psoriasis, eczema, and scleroderma
  • diabetes diabetic retinopathy, retinopathy of prematurity
  • age-related macular degeneration hemangio
  • Patients that can be treated with compounds of the present invention, or pharmaceutically acceptable salt, ester, prodrug, solvate, hydrate or derivative of said compounds, according to the methods of this invention include, for example, patients that have been diagnosed as having psoriasis; restenosis; atherosclerosis; BPH; breast cancer such as a ductal carcinoma in duct tissue in a mammary gland, medullary carcinomas, colloid carcinomas, tubular carcinomas, and inflammatory breast cancer; ovarian cancer, including epithelial ovarian tumors such as adenocarcinoma in the ovary and an adenocarcinoma that has migrated from the ovary into the abdominal cavity; uterine cancer; cervical cancer such as adenocarcinoma in the cervix epithelial including squamous cell carcinoma and adenocarcinomas; prostate cancer, such as a prostate cancer selected from the following: an adenocarcinoma or an adenocarinom
  • the invention also relates to a method of treating diabetes in a mammal that comprises administering to said mammal a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt, ester, prodrug, solvate, hydrate or derivative thereof.
  • the compounds described herein may be used to treat acne.
  • arteriosclerosis is a general term describing any hardening of medium or large arteries.
  • Atherosclerosis is a hardening of an artery specifically due to an atheromatous plaque.
  • Glomerulonephritis is a primary or secondary autoimmune renal disease characterized by inflammation of the glomeruli. It may be asymptomatic, or present with hematuria and/or proteinuria. There are many recognized types, divided in acute, subacute or chronic glomerulonephritis. Causes are infectious (bacterial, viral or parasitic pathogens), autoimmune or paraneoplastic.
  • the compounds described herein may be used for the treatment of bursitis, lupus, acute disseminated encephalomyelitis (ADEM), addison's disease, antiphospholipid antibody syndrome (APS), aplastic anemia, autoimmune hepatitis, coeliac disease, Crohn's disease, diabetes mellitus (type 1), goodpasture's syndrome, graves' disease, guillain-barr ⁇ syndrome (GBS), hashimoto's disease, inflammatory bowel disease, lupus erythematosus, myasthenia gravis, opsoclonus myoclonus syndrome (OMS), optic neuritis, ord's thyroiditis,ostheoarthritis, uveoretinitis, pemphigus, polyarthritis, primary biliary cirrhosis, reiter's syndrome, takayasu's arteritis, temporal arteritis, warm autoimmune hemo
  • the invention also relates to a method of treating a cardiovascular disease in a mammal that comprises administering to said mammal a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt, ester, prodrug, solvate, hydrate or derivative thereof.
  • cardiovascular conditions include, but are not limited to, atherosclerosis, restenosis, vascular occlusion and carotid obstructive disease.
  • the present invention provides methods of disrupting the function of a leukocyte or disrupting a function of an osteoclast.
  • the method includes contacting the leukocyte or the osteoclast with a function disrupting amount of a compound of the invention.
  • methods are provided for treating ophthalmic disease by administering one or more of the subject compounds or pharmaceutical compositions to the eye of a subject.
  • Methods are further provided for administering the compounds of the present invention via eye drop, intraocular injection, intravitreal injection, topically, or through the use of a drug eluting device, microcapsule, implant, or microfluidic device.
  • the compounds of the present invention are administered with a carrier or excipient that increases the intraocular penetrance of the compound such as an oil and water emulsion with colloid particles having an oily core surrounded by an interfacial film.
  • the colloid particles include at least one cationic agent and at least one non-ionic sufactant such as a poloxamer, tyloxapol, a polysorbate, a polyoxyethylene castor oil derivative, a sorbitan ester, or a polyoxyl stearate.
  • the cationic agent is an alkylamine, a tertiary alkyl amine, a quarternary ammonium compound, a cationic lipid, an amino alcohol, a biguanidine salt, a cationic compound or a mixture thereof.
  • the cationic agent is a biguanidine salt such as chlorhexidine, polyaminopropyl biguanidine, phenformin, alkylbiguanidine, or a mixture thereof.
  • the quaternary ammonium compound is a benzalkonium halide, lauralkonium halide, cetrimide, hexadecyltrimethylammonium halide, tetradecyltrimethylammonium halide, dodecyltrimethylammonium halide, cetrimonium halide, benzethonium halide, behenalkonium halide, cetalkonium halide, cetethyldimonium halide, cetylpyridinium halide, benzododecinium halide, chlorallyl methenamine halide, rnyristylalkonium halide, stearalkonium halide or a mixture of two or more thereof.
  • cationic agent is a benzalkonium chloride, lauralkonium chloride, benzododecinium bromide, benzethenium chloride, hexadecyltrimethylammonium bromide, tetradecyltrimethylammonium bromide, dodecyltrimethylammonium bromide or a mixture of two or more thereof.
  • the oil phase is mineral oil and light mineral oil, medium chain triglycerides (MCT), coconut oil; hydrogenated oils comprising hydrogenated cottonseed oil, hydrogenated palm oil, hydrogenate castor oil or hydrogenated soybean oil; polyoxyethylene hydrogenated castor oil derivatives comprising poluoxyl-40 hydrogenated castor oil, polyoxyl-60 hydrogenated castor oil or polyoxyl- 100 hydrogenated castor oil.
  • MCT medium chain triglycerides
  • coconut oil hydrogenated oils comprising hydrogenated cottonseed oil, hydrogenated palm oil, hydrogenate castor oil or hydrogenated soybean oil
  • polyoxyethylene hydrogenated castor oil derivatives comprising poluoxyl-40 hydrogenated castor oil, polyoxyl-60 hydrogenated castor oil or polyoxyl- 100 hydrogenated castor oil.
  • the invention further provides methods of modulating kinase activity by contacting a kinase with an amount of a compound of the invention sufficient to modulate the activity of the kinase. Modulate can be inhibiting or activating kinase activity. In some embodiments, the invention provides methods of inhibiting kinase activity by contacting a kinase with an amount of a compound of the invention sufficient to inhibit the activity of the kinase. In some embodiments, the invention provides methods of inhibiting kinase activity in a solution by contacting said solution with an amount of a compound of the invention sufficient to inhibit the activity of the kinase in said solution.
  • the invention provides methods of inhibiting kinase activity in a cell by contacting said cell with an amount of a compound of the invention sufficient to inhibit the activity of the kinase in said cell. In some embodiments, the invention provides methods of inhibiting kinase activity in a tissue by contacting said tissue with an amount of a compound of the invention sufficient to inhibit the activity of the kinase in said tissue. In some embodiments, the invention provides methods of inhibiting kinase activity in an organism by contacting said organism with an amount of a compound of the invention sufficient to inhibit the activity of the kinase in said organism.
  • the invention provides methods of inhibiting kinase activity in an animal by contacting said animal with an amount of a compound of the invention sufficient to inhibit the activity of the kinase in said animal. In some embodiments, the invention provides methods of inhibiting kinase activity in a mammal by contacting said mammal with an amount of a compound of the invention sufficient to inhibit the activity of the kinase in said mammal. In some embodiments, the invention provides methods of inhibiting kinase activity in a human by contacting said human with an amount of a compound of the invention sufficient to inhibit the activity of the kinase in said human.
  • the % of kinase activity after contacting a kinase with a compound of the invention is less than 1, 5, 10, 20, 30, 40, 50, 60, 70, 80 90, 95, or 99% of the kinase activity in the absence of said contacting step.
  • the kinase is a lipid kinase or a protein kinase.
  • the kinase is selected from the group consisting of PI3 kinase including different isorforms such as PI3 kinase ⁇ , PI3 kinase ⁇ , PI3 kinase ⁇ , PI3 kinase ⁇ ; DNA-PK; mTor; AbI, VEGFR, Ephrin receptor B4 (EphB4); TEK receptor tyrosine kinase (TIE2); FMS-related tyrosine kinase 3 (FLT-3); Platelet derived growth factor receptor (PDGFR); RET; ATM; ATR; hSmg-1; Hck; Src; Epidermal growth factor receptor (EGFR); KIT; Inulsin Receptor (IR) and IGFR.
  • PI3 kinase including different isorforms such as PI3
  • the invention further provides methods of modulating PI3 kinase activity by contacting a PI3 kinase with an amount of a compound of the invention sufficient to modulate the activity of the PI3 kinase. Modulate can be inhibiting or activating PI3 kinase activity. In some embodiments, the invention provides methods of inhibiting PI3 kinase activity by contacting a PI3 kinase with an amount of a compound of the invention sufficient to inhibit the activity of the PI3 kinase. In some embodiments, the invention provides methods of inhibiting PI3 kinase activity.
  • Such inhibition can take place in solution, in a cell expressing one or more PI3 kinases, in a tissue comprising a cell expressing one or more PI3 kinases, or in an organism expressing one or more PI3 kinases.
  • the invention provides methods of inhibiting PI3 kinase activity in an animal (including mammal such as humans) by contacting said animal with an amount of a compound of the invention sufficient to inhibit the activity of the PI3 kinase in said animal.
  • the present invention also provides methods for combination therapies in which an agent known to modulate other pathways, or other components of the same pathway, or even overlapping sets of target enzymes are used in combination with a compound of the present invention, or a pharmaceutically acceptable salt, ester, prodrug, solvate, hydrate or derivative thereof.
  • such therapy includes but is not limited to the combination of the subject compound with chemotherapeutic agents, therapeutic antibodies, and radiation treatment, to provide a synergistic or additive therapeutic effect.
  • the compounds or pharmaceutical compositions of the present invention may present synergistic or additive efficacy when administered in combination with agents that inhibit IgE production or activity. Such combination can reduce the undesired effect of high level of IgE associated with the use of one or more PI3K ⁇ inhibitors, if such effect occurs. This may be particularly useful in treatment of autoimmune and inflammatory disorders (AIID) such as rheumatoid arthritis. Additionally, the administration of PDK ⁇ or PI3K ⁇ / ⁇ inhibitors of the present invention in combination with inhibitors of mTOR may also exhibit synergy through enhanced inhibition of the PDK pathway.
  • AIID autoimmune and inflammatory disorders
  • the present invention provides a combination treatment of a disease associated with PDK ⁇ comprising administering to a PDK ⁇ inhibitor and an agent that inhibits IgE production or activity.
  • a PDK ⁇ inhibitor is applicable and they are described, e.g., US Patent No. 6,800,620.
  • Such combination treatment is particularly useful for treating autoimmune and inflammatory diseases (AIID) including but not limited to rheumatoid arthritis.
  • AIID autoimmune and inflammatory diseases
  • Agents that inhibit IgE production include but are not limited to one or more of TEI-9874, 2-(4-(6-cyclohexyloxy-2-naphtyloxy)phenylacetamide)benzoic acid, rapamycin, rapamycin analogs (i.e. rapalogs), TORCl inhibitors, TORC2 inhibitors, and any other compounds that inhibit mTORCl and mT0RC2.
  • Agents that inhibit IgE activity include, for example, anti-IgE antibodies such as for example Omalizumab and TNX-901.
  • the subject compounds or pharmaceutical compositions can be used in combination with commonly prescribed drugs including but not limited to Enbrel ® , Remicade ® , Humira ® , Avonex ® , and Rebif ® .
  • the subject compounds or pharmaceutical compositions can be administered in combination with commonly prescribed drugs including but not limited to Xolair ® , Advair ® , Singulair ® , and Spiriva ® .
  • the compounds of the invention may be formulated or administered in conjunction with other agents that act to relieve the symptoms of inflammatory conditions such as encephalomyelitis, asthma, and the other diseases described herein.
  • agents include non-steroidal anti-inflammatory drugs (NSAIDs), e.g. acetylsalicylic acid; ibuprofen; naproxen; indomethacin; nabumetone; tolmetin; etc.
  • NSAIDs non-steroidal anti-inflammatory drugs
  • Corticosteroids are used to reduce inflammation and suppress activity of the immune system.
  • the most commonly prescribed drug of this type is Prednisone.
  • Chloroquine (Aralen) or hydroxychloroquine (Plaquenil) may also be very useful in some individuals with lupus.
  • Azathioprine Imuran
  • Cytoxan cyclophosphamide
  • Other agents e.g. methotrexate and cyclosporin are used to control the symptoms of lupus.
  • Anticoagulants are employed to prevent blood from clotting rapidly. They range from aspirin at very low dose which prevents platelets from sticking, to heparin/coumadin.
  • this invention also relates to a pharmaceutical composition for inhibiting abnormal cell growth in a mammal which comprises an amount of a compound of the present invention, or a pharmaceutically acceptable salt, ester, prodrug, solvate, hydrate or derivative thereof, in combination with an amount of an anticancer agent (e.g. a chemotherapeutic agent).
  • an anticancer agent e.g. a chemotherapeutic agent.
  • Many chemotherapeutics are presently known in the art and can be used in combination with the compounds of the invention.
  • the chemotherapeutic is selected from the group consisting of mitotic inhibitors, alkylating agents, anti-metabolites, intercalating antibiotics, growth factor inhibitors, cell cycle inhibitors, enzymes, topoisomerase inhibitors, biological response modifiers, anti-hormones, angiogenesis inhibitors, and anti-androgens.
  • chemotherapeutic agents include cytotoxic agents, and non-peptide small molecules such as Gleevec (Imatinib Mesylate), Velcade (bortezomib), Casodex (bicalutamide), Iressa (gefitinib), and Adriamycin as well as a host of chemotherapeutic agents.
  • Non-limiting examples of chemotherapeutic agents include alkylating agents such as thiotepa and cyclosphosphamide (CYTOXANTM); alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, trietylenephosphoramide, triethylenethiophosphaoramide and trimethylolomelamine; nitrogen mustards such as chlorambucil, chlornaphazine, cholophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard; nitrosureas such as car
  • paclitaxel TAXOLTM, Bristol-Myers Squibb Oncology, Princeton, NJ.
  • docetaxel TAXOTERETM, Rhone-Poulenc Rorer, Antony, France
  • retinoic acid esperamicins
  • capecitabine ecitabine
  • pharmaceutically acceptable salts, acids or derivatives of any of the above TAXOLTM, Bristol-Myers Squibb Oncology, Princeton, NJ.
  • chemotherapeutic cell conditioners are anti-hormonal agents that act to regulate or inhibit hormone action on tumors such as anti-estrogens including for example tamoxifen (NolvadexTM), raloxifene, aromatase inhibiting 4(5)-imidazoles, 4-hydroxytamoxifen, trioxifene, keoxifene, LY 117018, onapristone, and toremifene (Fareston); and anti-androgens such as flutamide, nilutamide, bicalutamide, leuprolide, and goserelin; chlorambucil; gemcitabine; 6-thioguanine; mercaptopurine; methotrexate; platinum analogs such as cisplatin and carboplatin; vinblastine; platinum; etoposide (VP- 16); ifosfamide; mitomycin C; mitoxantrone; vincristine; vinorelbine; navelbine;
  • the compounds or pharmaceutical composition of the present invention can be used in combination with commonly prescribed anti-cancer drugs such as Herceptin ® , Avastin ® , Erbitux ® , Rituxan ® , Taxol , Arimidex ® , Taxotere ® , and Velcade .
  • This invention further relates to a method for using the compounds or pharmaceutical composition in combination with radiation therapy in inhibiting abnormal cell growth or treating the hyperproliferative disorder in the mammal. Techniques for administering radiation therapy are known in the art, and these techniques can be used in the combination therapy described herein. The administration of the compound of the invention in this combination therapy can be determined as described herein.
  • Radioactive isotopes e.g. At-211, 1-131, 1-125, Y-90, Re-186, Re-188, Sm-153, Bi-212, P-32, and radioactive isotopes of Lu.
  • Suitable radiation sources for use as a cell conditioner of the present invention include both solids and liquids.
  • the radiation source can be a radionuclide, such as 1-125, 1-131, Yb-169, Ir-192 as a solid source, 1-125 as a solid source, or other radionuclides that emit photons, beta particles, gamma radiation, or other therapeutic rays.
  • the radioactive material can also be a fluid made from any solution of radionuclide(s), e.g., a solution of 1-125 or 1-131, or a radioactive fluid can be produced using a slurry of a suitable fluid containing small particles of solid radionuclides, such as Au- 198, Y-90.
  • the radionuclide(s) can be embodied in a gel or radioactive micro spheres.
  • the compounds of the present invention can render abnormal cells more sensitive to treatment with radiation for purposes of killing and/or inhibiting the growth of such cells. Accordingly, this invention further relates to a method for sensitizing abnormal cells in a mammal to treatment with radiation which comprises administering to the mammal an amount of a compound of the present invention or pharmaceutically acceptable salt, ester, prodrug, solvate, hydrate or derivative thereof, which amount is effective is sensitizing abnormal cells to treatment with radiation.
  • the amount of the compound, salt, or solvate in this method can be determined according to the means for ascertaining effective amounts of such compounds described herein.
  • the compounds or pharmaceutical compositions of the present invention can be used in combination with an amount of one or more substances selected from anti-angiogenesis agents, signal transduction inhibitors, and antiproliferative agents.
  • Anti-angiogenesis agents such as MMP-2 (matrix-metalloprotienase 2) inhibitors, MMP-9 (matrix- metalloprotienase 9) inhibitors, and COX-11 (cyclooxygenase 11) inhibitors, can be used in conjunction with a compound of the present invention and pharmaceutical compositions described herein.
  • MMP-2 matrix-metalloprotienase 2
  • MMP-9 matrix- metalloprotienase 9
  • COX-11 cyclooxygenase 11
  • useful COX-II inhibitors include CELEBREXTM (alecoxib), valdecoxib, and rofecoxib.
  • Examples of useful matrix metalloproteinase inhibitors are described in WO 96/33172 (published October 24,1996), WO 96/27583 (published March 7,1996), European Patent Application No.
  • MMP-2 and MMP-9 inhibitors are those that have little or no activity inhibiting MMP-I.
  • MMP-2 and/or AMP-9 are those that selectively inhibit MMP-2 and/or AMP-9 relative to the other matrix-metalloproteinases (i. e., MAP-I, MMP-3, MMP-4, MMP-5, MMP-6, MMP- 7, MMP-8, MMP-IO, MMP-U, MMP-12, andMMP-13).
  • MMP inhibitors useful in the present invention are AG-3340, RO 32-3555, and RS 13-0830.
  • the invention also relates to a method of and to a pharmaceutical composition of treating a cardiovascular disease in a mammal which comprises an amount of a compound of the present invention, or a pharmaceutically acceptable salt, ester, prodrug, solvate, hydrate or derivative thereof, or an isotopically-labeled derivative thereof, and an amount of one or more therapeutic agents use for the treatment of cardiovascular diseases.
  • anti-thrombotic agents e.g., prostacyclin and salicylates
  • thrombolytic agents e.g., streptokinase, urokinase, tissue plasminogen activator (TPA) and anisoylated plasminogen-streptokinase activator complex (APSAC)
  • anti-platelets agents e.g., acetyl-salicylic acid (ASA) and clopidrogel
  • vasodilating agents e.g., nitrates
  • calcium channel blocking drugs antiproliferative agents, e.g., colchicine and alkylating agents, intercalating agents, growth modulating factors such as interleukins, transformation growth factor-beta and congeners of platelet derived growth factor, monoclonal antibodies directed against growth factors, anti-inflammatory agents, both steroidal and non-steroidal, and other agents that can modulate vessel tone, function,
  • Antibiotics can also be included in combinations or coatings comprised by the invention. Moreover, a coating can be used to effect therapeutic delivery focally within the vessel wall. By incorporation of the active agent in a swellable polymer, the active agent will be released upon swelling of the polymer.
  • tissue barriers also known as lubricants.
  • tissue barriers include, but are not limited to, polysaccharides, polyglycans, seprafilm, interceed and hyaluronic acid.
  • Medicaments which may be administered in conjunction with the compounds described herein include any suitable drugs usefully delivered by inhalation for example, analgesics, e.g. codeine, dihydromorphine, ergotamine, fentanyl or morphine; anginal preparations, e.g. diltiazem; antiallergics, e.g. cromoglycate, ketotifen or nedocromil; anti-infectives, e.g. cephalosporins, penicillins, streptomycin, sulphonamides, tetracyclines or pentamidine; antihistamines, e.g.
  • analgesics e.g. codeine, dihydromorphine, ergotamine, fentanyl or morphine
  • anginal preparations e.g. diltiazem
  • antiallergics e.g. cromoglycate, ketotifen or nedocromil
  • anti-inflammatories e.g. beclomethasone, flunisolide, budesonide, tipredane, triamcinolone acetonide or fluticasone
  • antitussives e.g. noscapine
  • bronchodilators e.g.
  • ephedrine adrenaline, fenoterol, formoterol, isoprenaline, metaproterenol, phenylephrine, phenylpropanolamine, pirbuterol, reproterol, rimiterol, salbutamol, salmeterol, terbutalin, isoetharine, tulobuterol, orciprenaline or (-)- 4-amino-3,5-dichloro- ⁇ -[[[6-[2-(2-pyridinyl)ethoxy]hexyl]-amino]methyl]benzenemethanol; diuretics, e.g. amiloride; anticholinergics e.g.
  • the medicaments may be used in the form of salts (e.g. as alkali metal or amine salts or as acid addition salts) or as esters (e.g. lower alkyl esters) or as solvates (e.g.
  • exemplary therapeutic agents useful for a combination therapy include but are not limited to agents as described above, radiation therapy, hormone antagonists, hormones and their releasing factors, thyroid and antithyroid drugs, estrogens and progestins, androgens, adrenocorticotropic hormone; adrenocortical steroids and their synthetic analogs; inhibitors of the synthesis and actions of adrenocortical hormones, insulin, oral hypoglycemic agents, and the pharmacology of the endocrine pancreas, agents affecting calcification and bone turnover: calcium, phosphate, parathyroid hormone, vitamin D, calcitonin, vitamins such as water-soluble vitamins, vitamin B complex, ascorbic acid, fat-soluble vitamins, vitamins A, K, and E, growth factors, cytokines, chemokines, muscarinic receptor agonists and antagonists; anticholinesterase agents; agents acting at the neuromuscular junction and/or
  • Therapeutic agents can also include agents for pain and inflammation such as histamine and histamine antagonists, bradykinin and bradykinin antagonists, 5-hydroxytryptamine (serotonin), lipid substances that are generated by biotransformation of the products of the selective hydrolysis of membrane phospholipids, eicosanoids, prostaglandins, thromboxanes, leukotrienes, aspirin, nonsteroidal anti-inflammatory agents, analgesic-antipyretic agents, agents that inhibit the synthesis of prostaglandins and thromboxanes, selective inhibitors of the inducible cyclooxygenase, selective inhibitors of the inducible cyclooxygenase-2, autacoids, paracrine hormones, somatostatin, gastrin, cytokines that mediate interactions involved in humoral and cellular immune responses, lipid-derived autacoids, eicosanoids, ⁇ -adrenergic agonists, ipra
  • Additional therapeutic agents contemplated herein include diuretics, vasopressin, agents affecting the renal conservation of water, rennin, angiotensin, agents useful in the treatment of myocardial ischemia, antihypertensive agents, angiotensin converting enzyme inhibitors, ⁇ -adrenergic receptor antagonists, agents for the treatment of hypercholesterolemia, and agents for the treatment of dyslipidemia.
  • Other therapeutic agents contemplated include drugs used for control of gastric acidity, agents for the treatment of peptic ulcers, agents for the treatment of gastroesophageal reflux disease, prokinetic agents, antiemetics, agents used in irritable bowel syndrome, agents used for diarrhea, agents used for constipation, agents used for inflammatory bowel disease, agents used for biliary disease, agents used for pancreatic disease.
  • Therapeutic agents used to treat protozoan infections drugs used to treat Malaria, Amebiasis, Giardiasis, Trichomoniasis, Trypanosomiasis, and/or Leishmaniasis, and/or drugs used in the chemotherapy of helminthiasis.
  • therapeutic agents include antimicrobial agents, sulfonamides, trimethoprim-sulfamethoxazole quinolones, and agents for urinary tract infections, penicillins, cephalosporins, and other, ⁇ -Lactam antibiotics, an agent comprising an aminoglycoside, protein synthesis inhibitors, drugs used in the chemotherapy of tuberculosis, mycobacterium avium complex disease, and leprosy, antifungal agents, antiviral agents including nonretroviral agents and antiretroviral agents.
  • anti-receptor tyrosine kinase antibodies cetuximab, panitumumab, trastuzumab
  • anti CD20 antibodies rituximab, tositumomab
  • other antibodies such as alemtuzumab, bevacizumab, and gemtuzumab.
  • therapeutic agents used for immunomodulation such as immunomodulators, immunosuppressive agents, tolerogens, and immunostimulants are contemplated by the methods herein.
  • therapeutic agents acting on the blood and the blood-forming organs such as hematopoietic agents, growth factors, minerals, and vitamins, anticoagulant, thrombolytic, and antiplatelet drugs.
  • the compounds described herein can be used in combination with the agents disclosed herein or other suitable agents, depending on the condition being treated. Hence, in some embodiments the compounds of the invention will be co-administer with other agents as described above.
  • the compounds described herein may be administered with the second agent simultaneously or separately.
  • This administration in combination can include simultaneous administration of the two agents in the same dosage form, simultaneous administration in separate dosage forms, and separate administration. That is, a compound described herein and any of the agents described above can be formulated together in the same dosage form and administered simultaneously. Alternatively, a compound of the present invention and any of the agents described above can be simultaneously administered, wherein both the agents are present in separate formulations.
  • a compound of the present invention can be administered just followed by and any of the agents described above, or vice versa.
  • a compound of the present invention and any of the agents described above may be administered a few minutes apart, or a few hours apart, or a few days apart.
  • Administration of the compounds of the present invention can be effected by any method that enables delivery of the compounds to the site of action. These methods include oral routes, intraduodenal routes, parenteral injection (including intravenous, intraarterial, subcutaneous, intramuscular, intravascular, intraperitoneal or infusion), topical (e.g. transdermal application), rectal administration, via local delivery by catheter or stent. Compounds can also abe administered intraadiposally or intrathecally.
  • the amount of the compound administered will be dependent on the mammal being treated, the severity of the disorder or condition, the rate of administration, the disposition of the compound and the discretion of the prescribing physician.
  • an effective dosage is in the range of about 0.001 to about 100 mg per kg body weight per day, preferably about 1 to about 35 mg/kg/day, in single or divided doses. For a 70 kg human, this would amount to about 0.05 to 7 g/day, preferably about 0.05 to about 2.5 g/day.
  • dosage levels below the lower limit of the aforesaid range may be more than adequate, while in other cases still larger doses may be employed without causing any harmful side effect, e.g. bydividing such larger doses into several small doses for administration throughout the day.
  • the compound may be applied as a sole therapy or may involve one or more other anti-tumor substances, for example those selected from, mitotic inhibitors, for example vinblastine; alkylating agents, for example cis- platin, carboplatin and cyclophosphamide; anti-metabolites, for example 5-fluorouracil, cytosine arabinside and hydroxyurea, or, for example, one of the preferred anti-metabolites disclosed in European Patent Application No.
  • mitotic inhibitors for example vinblastine
  • alkylating agents for example cis- platin, carboplatin and cyclophosphamide
  • anti-metabolites for example 5-fluorouracil, cytosine arabinside and hydroxyurea, or, for example, one of the preferred anti-metabolites disclosed in European Patent Application No.
  • Such conjoint treatment may be achieved by way of the simultaneous, sequential or separate dosing of the individual components of treatment.
  • a compound of the invention is administered in a single dose.
  • administration will be by injection, e.g., intravenous injection, in order to introduce the agent quickly.
  • other routes may be used as appropriate.
  • a single dose of a compound of the invention may also be used for treatment of an acute condition.
  • a compound of the invention is administered in multiple doses. Dosing may be about once, twice, three times, four times, five times, six times, or more than six times per day. Dosing may be about once a month, once every two weeks, once a week, or once every other day. In another embodiment a compound of the invention and another agent are administered together about once per day to about 6 times per day. In another embodiment the administration of a compound of the invention and an agent continues for less than about 7 days. In yet another embodiment the administration continues for more than about 6, 10, 14, 28 days, two months, six months, or one year. In some cases, continuous dosing is achieved and maintained as long as necessary.
  • Administration of the agents of the invention may continue as long as necessary.
  • an agent of the invention is administered for more than 1, 2, 3, 4, 5, 6, 7, 14, or 28 days.
  • an agent of the invention is administered for less than 28, 14, 7, 6, 5, 4, 3, 2, or 1 day.
  • an agent of the invention is administered chronically on an ongoing basis, e.g., for the treatment of chronic effects.
  • An effective amount of a compound of the invention may be administered in either single or multiple doses by any of the accepted modes of administration of agents having similar utilities, including rectal, buccal, intranasal and transdermal routes, by intra-arterial injection, intravenously, intraperitoneally, parenterally, intramuscularly, subcutaneously, orally, topically, or as an inhalant.
  • compositions of the invention may also be delivered via an impregnated or coated device such as a stent, for example, or an artery-inserted cylindrical polymer.
  • a method of administration may, for example, aid in the prevention or amelioration of restenosis following procedures such as balloon angioplasty.
  • compounds of the invention may slow or inhibit the migration and proliferation of smooth muscle cells in the arterial wall which contribute to restenosis.
  • a compound of the invention may be administered, for example, by local delivery from the struts of a stent, from a stent graft, from grafts, or from the cover or sheath of a stent.
  • a compound of the invention is admixed with a matrix.
  • Such a matrix may be a polymeric matrix, and may serve to bond the compound to the stent.
  • Polymeric matrices suitable for such use include, for eample, lactone-based polyesters or copolyesters such as polylactide, polycaprolactonglycolide, polyorthoesters, polyanhydrides, polyaminoacids, polysaccharides, polyphosphazenes, poly (ether-ester) copolymers (e.g. PEO-PLLA); polydimethylsiloxane, poly(ethylene- vinylacetate), acrylate-based polymers or copolymers (e.g.
  • Suitable matrices may be nondegrading or may degrade with time, releasing the compound or compounds.
  • Compounds of the invention may be applied to the surface of the stent by various methods such as dip/spin coating, spray coating, dip- coating, and/or brush-coating. The compounds may be applied in a solvent and the solvent may be allowed to evaporate, thus forming a layer of compound onto the stent. Alternatively, the compound may be located in the body of the stent or graft, for example in microchannels or micropores.
  • stents When implanted, the compound diffuses out of the body of the stent to contact the arterial wall.
  • stents may be prepared by dipping a stent manufactured to contain such micropores or microchannels into a solution of the compound of the invention in a suitable solvent, followed by evaporation of the solvent. Excess drug on the surface of the stent may be removed via an additional brief solvent wash.
  • compounds of the invention may be covalently linked to a stent or graft.
  • a covalent linker may be used which degrades in vivo, leading to the release of the compound of the invention. Any bio-labile linkage may be used for such a purpose, such as ester, amide or anhydride linkages.
  • Compounds of the invention may additionally be administered intravascularly from a balloon used during angioplasty. Extravascular administration of the compounds via the pericard or via advential application of formulations of the invention may also be performed to decrease restenosis.
  • a variety of stent devices which may be used as described are disclosed, for example, in the following references, all of which are hereby incorporated by reference: U.S. Pat. No. 5451233; U.S. Pat. No. 5040548; U.S. Pat. No. 5061273; U.S. Pat. No. 5496346; U.S. Pat. No. 5292331; U.S. Pat. No. 5674278; U.S. Pat. No. 3657744; U.S. Pat. No.
  • the compounds of the invention may be administered in dosages as described herein (see, e.g., Compositions).
  • the subject pharmaceutical composition may, for example, be in a form suitable for oral administration as a tablet, capsule, pill, powder, sustained release formulations, solution, suspension, for parenteral injection as a sterile solution, suspension or emulsion, for topical administration as an ointment or cream or for rectal administration as a suppository.
  • the pharmaceutical composition may be in unit dosage forms suitable for single administration of precise dosages.
  • the pharmaceutical composition will include a conventional pharmaceutical carrier or excipient and a compound according to the invention as an active ingredient. In addition, it may include other medicinal or pharmaceutical agents, carriers, adjuvants, etc.
  • Exemplary parenteral administration forms include solutions or suspensions of active compound in sterile aqueous solutions, for example, aqueous propylene glycol or dextrose solutions. Such dosage forms can be suitably buffered, if desired.
  • the activity of the compounds of the present invention may be determined by the following procedure, as well as the procedure described in the examples below. N-terminal 6 His-tagged, constitutively active kinase is expressed in E. coli and protein is purified by conventional methods (Ann et al. Science 1994, 265, 966-970).
  • the activity of the kinase is assessed by measuring the incorporation of ⁇ - 33 P-phosphate from ⁇ - 33 P-ATP onto
  • N-terminal His tagged substrate which is expressed in E. coli and is purified by conventional methods, in the presence of the kinase.
  • the assay is carried out in 96-well polypropylene plate.
  • the incubation mixture (100, ⁇ L) comprises of 25 mM Hepes, pH 7.4, 10 mM MgCl 2 , 5 mM ⁇ -glycerolphosphate, 100 ⁇ M Na-orthovanadate,
  • Inhibitors are suspended in DMSO, and all reactions, including controls are performed at a final concentration of 1% DMSO. Reactions are initiated by the addition of 10 ⁇ M ATP (with 0.5 ⁇ Ci ⁇ - 33 P- ATP/well) and incubated at ambient temperature for 45 minutes. Equal volume of 25% TCA is added to stop the reaction and precipitate the proteins. Precipitated proteins are trapped onto glass fiber B filterplates, and excess labeled ATP washed off using a Tomtec MACH III harvestor. Plates are allowed to air-dry prior to adding 30 ⁇ L/well of Packard Microscint 20, and plates are counted using a Packard TopCount.
  • Example 1 Expression and Inhibition Assays of pll ⁇ /p85 ⁇ , pll ⁇ /p85 ⁇ , pll ⁇ /p85 ⁇ , and pllO ⁇ :
  • Class I PB-Ks can be either purchased (pi 10 ⁇ /p85 ⁇ , pi 10 ⁇ /p85 ⁇ , pi 10 ⁇ /p85 ⁇ from Upstate, and pi lO ⁇ from Sigma) or expressed as previously described (Knight et al., 2004).
  • IC50 values are measured using either a standard TLC assay for lipid kinase activity (described below) or a high-throughput membrane capture assay.
  • Kinase reactions are performed by preparing a reaction mixture containing kinase, inhibitor (2% DMSO final concentration), buffer (25 mM HEPES, pH 7.4, 10 mM MgC12), and freshly sonicated phosphatidylinositol (100 ⁇ g/ml).
  • Reactions are initiated by the addition of ATP containing 10 ⁇ Ci of ⁇ -32P- ATP to a final concentration 10 or 100 ⁇ M and allowed to proceed for 5 minutes at room temperature.
  • ATP containing 10 ⁇ Ci of ⁇ -32P- ATP
  • reactions are then terminated by the addition of 105 ⁇ l IN HCl followed by 160 ⁇ l CHC13:MeOH (1:1).
  • the biphasic mixture is vortexed, briefly centrifuged, and the organic phase is transferred to a new tube using a gel loading pipette tip precoated with CHCl 3 .
  • This extract is spotted on TLC plates and developed for 3 - 4 hours in a 65:35 solution of n- ⁇ ropanol:lM acetic acid.
  • TLC plates are then dried, exposed to a phosphorimager screen (Storm, Amersham), and quantitated.
  • kinase activity is measured at 10 — 12 inhibitor concentrations representing two-fold dilutions from the highest concentration tested (typically, 200 ⁇ M).
  • IC50 determinations are repeated two to four times, and the reported value is the average of these independent measurements.
  • kits or systems for assaying PI3-K activities are avaiable.
  • the commercially available kits or systems can be used to screen for inhibitors and/or agonists of PD-Ks including but not limited to PI 3-Kinase ⁇ , ⁇ , ⁇ , and ⁇ .
  • Anr exemplary system is PI 3-Kinase (human) HTRFTM Assay from Upstate.
  • the assay can be carried out according to the procedures suggested by the manufacturer. Briefly, the assay is a time resolved FRET assay that indirectly measures PIP3 product formed by the activity of a PI3- K.
  • the kinase reaction is performed in a microtitre plate (e.g., a 384 well microtitre plate).
  • the total reaction volume is approximately 20ul per well.
  • each well receives 2ul of test compound in 20% dimethylsulphoxide resulting in a 2% DMSO final concentration.
  • approximately 14.5ul of a kinase/PIP2 mixture (diluted in IX reaction buffer) is added per well for a final concentration of 0.25-
  • kinase and lOuM PIP2 The plate is sealed and incubated for 15 minutes at room temperature. To start the reaction, 3.5ul of ATP (diluted in IX reaction buffer) is added per well for a final concentration of lOuM ATP. The plate is sealed and incubated for 1 hour at room temperature. The reaction is stopped by adding 5ul of Stop Solution per well and then 5ul of Detection Mix is added per well. The plate is sealed, incubated for 1 hour at room temperature, and then read on an appropriate plate reader. Data is analyzed and IC50s are generated using GraphPad Prism 5.
  • AbI T315I (Upstate) in an assay containing 25 mM HEPES, pH 7.4, 10 mM MgC12, 200 ⁇ M ATP (2.5 ⁇ Ci of ⁇ -32P-ATP), and 0.5 mg/mL BSA.
  • the optimized AbI peptide substrate EAIY AAPF AKKK is used as phosphoacceptor (200 ⁇ M). Reactions are terminated by spotting onto phosphocellulose sheets, which are washed with 0.5% phosphoric acid (approximately 6 times, 5-10 minutes each). Sheets are dried and the transferred radioactivity quantitated by phosphorimaging.
  • the compounds described herein can be assayed in triplicate against recombinant full-length Hck in an assay containing 25 mM HEPES, pH 7.4, 10 mM MgC12, 200 ⁇ M ATP (2.5 ⁇ Ci of ⁇ -32P-ATP), and 0.5 mg/mL BSA.
  • the optimized Src family kinase peptide substrate EIYGEFKKK is used as phosphoacceptor (200 ⁇ M). Reactions are terminated by spotting onto phosphocellulose sheets, which are washed with 0.5% phosphoric acid (approximately 6 times, 5-10 minutes each). Sheets are dried and the transferred radioactivity quantitated by phosphorimaging.
  • the compounds described herein can be assayed in triplicate against recombinant insulin receptor kinase domain (Upstate) in an assay containing 25 mM HEPES, pH 7.4, 10 mM MgC12, 10 mM MnC12, 200 ⁇ M ATP (2.5 ⁇ Ci of ⁇ -32P-ATP), and 0.5 mg/mL BSA.
  • Poly E-Y (Sigma; 2 mg/mL) is used as a substrate. Reactions are terminated by spotting onto nitrocellulose, which is washed with IM NaCl/ 1% phosphoric acid (approximately 6 times, 5-10 minutes each). Sheets are dried and the transferred radioactivity quantitated by phosphorimaging.
  • DNA-PK can be purchased from Promega and assayed using the DNA-PK Assay System (Promega) according to the manufacturer's instructions.
  • Example 7 Expression and Inhibition Assays mTOR
  • the compounds described herein can be tested against recombinant mTOR (Invitrogen) in an assay containing 50 mM HEPES, pH 7.5, ImM EGTA, 10 mM MgC12, 2.5 mM, 0.01% Tween, 10 ⁇ M ATP (2.5 ⁇ Ci of ⁇ -32P-ATP), and 3 ⁇ g/mL BSA.
  • Rat recombinant PHAS- 1/4EBP1 (Calbiochem; 2 mg/mL) is used as a substrate. Reactions are terminated by spotting onto nitrocellulose, which is washed with IM NaCl/ 1% phosphoric acid (approximately 6 times, 5-10 minutes each). Sheets are dried and the transferred radioactivity quantitated by phosphorimaging.
  • kits or systems for assaying mTOR activity are commercially avaiable. For instance, one can use Invitrogen's LanthaScreenTM Kinase assay to test the inhibitors of mTOR disclosed herein.
  • This assay is a time resolved FRET platform that measures the phosphorylation of GFP labeled 4EBP 1 by mTOR kinase.
  • the kinase reaction is performed in a white 384 well microtitre plate. The total reaction volume is 20ul per well and the reaction buffer composition is 5OmM HEPES pH7.5, 0.01% Polysorbate 20, ImM EGTA, 1OmM MnC12, and 2mM DTT.
  • each well receives 2ul of test compound in 20% dimethylsulphoxide resulting in a 2% DMSO final concentration.
  • 8ul of mTOR diluted in reaction buffer is added per well for a 60ng/ml final concentration.
  • lOul of an ATP/GFP-4EBP1 mixture (diluted in reaction buffer) is added per well for a final concentration of lOuM ATP and 0.5uM GFP-4EBP1. The plate is sealed and incubated for 1 hour at room temperature.
  • the reaction is stopped by adding lOul per well of a Tb-anti-pT46 4EBP 1 antibody/EDTA mixture (diluted in TR-FRET buffer) for a final concentration of 1.3nM antibody and 6.7mM EDTA.
  • the plate is sealed, incubated for 1 hour at room temperature, and then read on a plate reader set up for LanthaScreenTM TR-FRET. Data is analyzed and IC50s are generated using GraphPad Prism 5.
  • Example 8 Expression and Inhibition Assays of Vascular endothelial growth receptor
  • the compounds described herein can be tested against recombinant Ephrin receptor B4 kinase domain (Invitrogen) in an assay containing 25 mM HEPES, pH 7.4, 10 mM MgC12, 0.1% BME, 10 ⁇ M ATP (2.5 ⁇ Ci of ⁇ -32P-ATP), and 3 ⁇ g/mL BSA.
  • Poly E-Y (Sigma; 2 mg/mL) is used as a substrate. Reactions are terminated by spotting onto nitrocellulose, which is washed with IM NaCl/ 1% phosphoric acid (approximately 6 times, 5-10 minutes each). Sheets are dried and the transferred radioactivity quantitated by phosphorimaging.
  • the compounds described herein can be tested against recombinant KIT kinase domain (Invitrogen) in an assay containing 25 mM HEPES, pH 7.4, 10 mM MgC12, ImM DTT, 1OmM MnC12, 10 ⁇ M ATP (2.5 ⁇ Ci of ⁇ -32P-ATP), and 3 ⁇ g/mL BSA.
  • Poly E-Y (Sigma; 2 mg/mL) is used as a substrate. Reactions are terminated by spotting onto nitrocellulose, which is washed with IM NaCl/1% phosphoric acid (approximately 6 times, 5-10 minutes each). Sheets are dried and the transferred radioactivity quantitated by phosphorimaging.
  • Example 12 Expression and Inhibition Assays of RET
  • the compounds described herein can be tested against recombinant RET kinase domain (Invitrogen) in an assay containing 25 mM HEPES, pH 7.4, 10 mM MgC12, 2.5mM DTT, 10 ⁇ M ATP (2.5 ⁇ Ci of ⁇ -32P- ATP), and 3 ⁇ g/mL BSA.
  • the optimized AbI peptide substrate EAIY AAPF AKKK is used as phosphoacceptor (200 ⁇ M). Reactions are terminated by spotting onto phosphocellulose sheets, which are washed with 0.5% phosphoric acid (approximately 6 times, 5-10 minutes each). Sheets are dried and the transferred radioactivity quantitated by phosphorimaging.
  • Example 13 Expression and Inhibition Assays of Platelet derived growth factor receptor
  • Example 14 Expression and Inhibition Assays of FMS-related tyrosine kinase 3 (FLT-3)
  • TIE2 TEK receptor tyrosine kinase
  • the compounds described herein can be tested against recombinant TIE2 kinase domain (Invitrogen) in an assay containing 25 mM HEPES, pH 7.4, 10 mM MgC12, 2mM DTT, 1OmM MnC12, 10 ⁇ M ATP (2.5 ⁇ Ci of ⁇ -32P-ATP), and 3 ⁇ g/mL BSA.
  • Poly E-Y (Sigma; 2 mg/mL) is used as a substrate. Reactions are terminated by spotting onto nitrocellulose, which is washed with IM NaCl/1% phosphoric acid (approximately 6 times, 5-10 minutes each). Sheets are dried and the transferred radioactivity quantitated by phosphorimaging.
  • Example 16 B Cell Activation and Proliferation Assay
  • the ability of one or more subject compounds to inhibit B cell activon and proliferation is determined according to standard procedures known in the art. For example, an in vitro cellular proliferation assay is established that measures the metabolic activity of live cells. The assay is performed in a 96 well microtiter plate using Alamar Blue reduction. Balb/c splenic B cells are purified over a Ficoll-PaqueTM PLUS gradient followed by magnetic cell separation using a MACS B cell Isolation Kit (Miletenyi). Cells are plated in 90ul at 50,000 cells/well in B Cell Media (RPMI + 10%FBS + Penn/Strep + 5OuM bME + 5mM HEPES). A compound disclosed herein is diluted in B Cell Media and added in a lOul volume. Plates are incubated for
  • a 50ul B cell stimulation cocktail is then added containing either lOug/ml LPS or 5ug/ml F(ab')2 Donkey anti-mouse IgM plus 2ng/ml recombinant mouse IL4 in B Cell Media. Plates are incubated for 72 hours at 37 0 C and 5% CO 2 . A volume of 15uL of Alamar Blue reagent is added to each well and plates are incubated for 5 hours at 37C and 5% CO 2 . Alamar Blue fluoresce is read at 560Ex/590Em, and IC50 or EC50 values are calculated using GraphPad Prism 5.
  • an in vitro cellular proliferation assay can be performed to measure the metabolic activity of live cells.
  • the assay is performed in a 96 well microtiter plate using Alamar Blue reduction.
  • Human tumor cell lines are obtained from ATCC (e.g., MCF7, U-87 MG, MDA-MB-468, PC-3), grown to confluency in T75 flasks, trypsinized with 0.25% trypsin, washed one time with Tumor Cell Media (DMEM + 10%FBS), and plated in 90ul at 5,000 cells/well in Tumor Cell Media.
  • a compound disclosed herein is diluted in Tumor Cell Media and added in a lOul volume. Plates are incubated for 72 hours at 37C and 5% CO 2 . A volume of lOuL of Alamar Blue reagent is added to each well and plates are incubated for 3 hours at 37C and 5% CO 2 . Alamar Blue fluoresce is read at 560Ex/590Em, and IC50 values are calculated using GraphPad Prism 5.
  • This tumor model is established from a tumor biopsy of an ovarian cancer patient. Tumor biopsy is taken from the patient.
  • A2780Tax is a paclitaxel-resistant human ovarian carcinoma model. It is derived from the sensitive parent A2780 line by co-incubation of cells with paclitaxel and verapamil, an MDR-reversal agent. Its resistance mechanism has been shown to be non-MDR related and is attributed to a mutation in the gene encoding the beta-tubulin protein.
  • the compounds described herein can be administered to mice bearing staged tumors on an every 2 days x 5 schedule.
  • HCTl 16/VM46 Human Colon Carcinoma Xenograft (Multi-Drug Resistant).
  • HCTl 16/VM46 is an MDR-resistant colon carcinoma developed from the sensitive HCTl 16 parent line. In vivo, grown in nude mice, HCTl 16/VM46 has consistently demonstrated high resistance to paclitaxel.
  • the compounds described herein can be administered to mice bearing staged tumors on an every 2 days x 5 schedule.
  • M5076 is a mouse fibrosarcoma that is inherently refractory to paclitaxel in vivo.
  • the compounds described herein can be administered to mice bearing staged tumors on an every 2 days x 5 schedule.
  • One or more compounds of the invention can be used in combination other therapeutic agents in vivo in the multidrug resistant human colon carcinoma xenografts HCT/VM46 or any other model known in the art including those described herein.
  • the stability of one or more subject compounds is determined according to standard procedures known in the art. For example, stability of one or more subject compounds is established by an in vitro assay. In particular, an in vitro microsome stability assay is established that measures stability of one or more subject compounds when reacting with mouse, rat or human microsomes from liver. The microsome reaction with compounds is performed in 1.5 mL Eppendorf tube. Each tube contains 0.1 ⁇ L of 10.0 mg/ml NADPH; 75 ⁇ L of 20.0 mg/ml mouse, rat or human liver microsome; 0.4 ⁇ L of 0.2 M phosphate buffer, and 425 ⁇ L of ddH 2 O.
  • Negative control (without NADPH) tube contains 75 ⁇ L of 20.0 mg/ml mouse, rat or human liver microsome; 0.4 ⁇ L of 0.2 M phosphate buffer, and 525 ⁇ L of ddH 2 O.
  • the reaction is started by adding 1.0 ⁇ L of 10.0 mM tested compound.
  • the reaction tubes are incubated at 37°C. 100 ⁇ L sample is collected into new Eppendorf tube containing 300 ⁇ L cold Methanol at 0, 5, 10, 15, 30 and 60 minutes of reaction. Samples are centrifuged at 15,000 rpm to remove protein. Supernatant of centrifuged sample is transferred to new tube. Concentration of stable compound after reaction with microsome in the supernatant is measured by Liquid Chromatography/Mass Spectrometry (LC-MS).
  • LC-MS Liquid Chromatography/Mass Spectrometry
  • Example 20 Plasma stability assay
  • the stability of one or more subject compounds in plasma is determined according to standard procedures known in the art. See, e.g., Rapid Commun. Mass Spectrom., 10: 1019-1026. The following procedure is an HPLC-MS/MS assay using human plasma; other species including monkey, dog, rat, and mouse are also available. Frozen, heparinized human plasma is thawed in a cold water bath and spun for 10 minutes at 2000 rpm at 4 0 C prior to use.
  • a subject compound is added from a 400 ⁇ M stock solution to an aliquot of pre- warmed plasma to give a final assay volume of 400 ⁇ L (or 800 ⁇ L for half-life determination), containing 5 ⁇ M test compound and 0.5 % DMSO.
  • Reactions are incubated, with shaking, for 0 minutes and 60 minutes at 37 0 C, or for 0, 15, 30, 45 and 60 minutes at 37 C for half life determination. Reactions are stopped by transferring 50 ⁇ L of the incubation mixture to 200 ⁇ L of ice-cold acetonitrile and mixed by shaking for 5 minutes.
  • the samples are centrifuged at 6000 x g for 15 minutes at 4 0 C and 120 ⁇ L of supernatant removed into clean tubes. The samples are then evaporated to dryness and submitted for analysis by HPLC-MS/MS.
  • one or more control or reference compounds (5 ⁇ M) are tested simultaneously with the test compounds: one compound, propoxycaine, with low plasma stability and another compound, propantheline, with intermediate plasma stability.
  • MS/MS using selected reaction monitoring SRM.
  • the HPLC conditions consist of a binary LC pump with autosampler, a mixed-mode, C 12, 2 x 20 mm column, and a gradient program. Peak areas corresponding to the analytes are recorded by HPLC-MS/MS. The ratio of the parent compound remaining after 60 minutes relative to the amount remaining at time zero, expressed as percent, is reported as plasma stability. In case of half-life determination, the half-life is estimated from the slope of the initial linear range of the logarithmic curve of compound remaining (%) vs. time, assuming first order kinetics.
  • Example 21 Chemical Stability
  • the chemical stability of one or more subject compounds is determined according to standard procedures known in the art. The following details an exemplary procedure for ascertaining chemical stability of a subject compound.
  • the default buffer used for the chemical stability assay is phosphate-buffered saline (PBS) at pH 7.4; other suitable buffers can be used.
  • PBS phosphate-buffered saline
  • a subject compound is added from a 100 ⁇ M stock solution to an aliquot of PBS (in duplicate) to give a final assay volume of 400 ⁇ L, containing 5 ⁇ M test compound and 1% DMSO (for half-life determination a total sample volume of 700 ⁇ L is prepared).
  • Reactions are incubated, with shaking, for 0 minutes and 24 hours at 37°C; for half-life determination samples are incubated for 0, 2, 4, 6, and 24 hours. Reactions are stopped by adding immediately 100 ⁇ L of the incubation mixture to 100 ⁇ L of acetonitrile and vortexing for 5 minutes. The samples are then stored at -2O 0 C until analysis by HPLC-MS/MS. Where desired, a control compound or a reference compound such as chlorambucil (5 ⁇ M) is tested simultaneously with a subject compound of interest, as this compound is largely hydrolyzed over the course of 24 hours. Samples are analyzed via (RP)HPLC-MS/MS using selected reaction monitoring (SRM).
  • SRM selected reaction monitoring
  • the HPLC conditions consist of a binary LC pump with autosampler, a mixed-mode, C12, 2 x 20 mm column, and a gradient program. Peak areas corresponding to the analytes are recorded by HPLC-MS/MS. The ratio of the parent compound remaining after 24 hours relative to the amount remaining at time zero, expressed as percent, is reported as chemical stability. In case of half-life determination, the half-life is estimated from the slope of the initial linear range of the logarithmic curve of compound remaining (%) vs. time, assuming first order kinetics.
  • Cells comprising components of the Akt/mTOR pathway, including but not limited to L6 myoblasts,
  • B-ALL cells B-cells, T-cells, leukemia cells, bone marrow cells, pi 90 transduced cells, philladelphia chromosome positive cells (Ph+), and mouse embryonic fibroblasts, are typically grown in cell growth media such as DMEM supplemented with fetal bovine serum and/or antibiotics, and grown to confluency.
  • cell growth media such as DMEM supplemented with fetal bovine serum and/or antibiotics
  • said cells are serum starved overnight and incubated with one or more compounds disclosed herein or about 0.1% DMSO for approximately 1 minute to about 1 hour prior to stimulation with insulin (e.g. 100 nM) for about 1 minutes to about 1 hour.
  • insulin e.g. 100 nM
  • Cells are lysed by scraping into ice cold lysis buffer containing detergents such as sodium dodecyl sulfate and protease inhibitors (e.g., PMSF).
  • the solution After contacting cells with lysis buffer, the solution is briefly sonicated, cleared by centrifugation, resolved by SDS-PAGE, transferred to nitrocellulose or PVDF and immunoblotted using antibodies to phospho- Akt S473, phospho- Akt T308, Akt, and ⁇ -actin (Cell Signaling Technologies).
  • results demonstrate that one or more compounds of the present disclosure inhibit insulin stimulated phosphorylation of Akt at S473.
  • some compounds disclosed herein additionally inhibit insulin stimulated phosphorylation of Akt at T308.
  • Such class of compounds can inhibit Akt more effectively than rapamycin and may be indicative of mTORC2 inhibitors or inhibitors of upstream kinases such as PI3K or Akt.
  • PI3K/ Akt /mTor signaling is measured in blood cells using the phosflow method (Methods
  • T cell receptor anti-CD3 with secondary antibody
  • BCR B cell receptor
  • Fab '2 fragments anti-kappa light chain antibody
  • the level of phosphrylation of kinase substrates such as Akt and S6 are then measured by incubating the fixed cells with labeled antibodies specific to the phosphorylated isoforms of these proteins. The population of cells are then analyzed by flow cytometry.
  • Murine bone marrow cells freshly transformed with a pi 90 BCR-AbI retrovirus (herein referred to as p 190 transduced cells) are plated in the presence of various drug combinations in M3630 methylcellulose media for about 7 days with recombinant human IL-7 in about 30% serum, and the number of colonies formed is counted by visual examination under a microscope.
  • human peripheral blood mononuclear cells are obtained from Philadelphia chromosome positive (Ph+) and negative (Ph-) patients upon initial diagnosis or relapse. Live cells are isolated and enriched for CD 19+ CD34+ B cell progenitors. After overnight liquid culture, cells are plated in methocult GF+ H4435, Stem Cell Tehcnologies) suplemented with cytokines (IL-3, IL-6, IL-7, G-CSF, GM- CSF, CF, Flt3 ligand, and erythropoietin) and various concentrations of known chemotherapeutic agents in combination with either compounds of the present disclosure. Colonies are counted by microscopy 12-14 days later. This method can be used to test for evidence of additive or synergistic activity.
  • cytokines IL-3, IL-6, IL-7, G-CSF, GM- CSF, CF, Flt3 ligand, and erythropoietin
  • Example 25 In Vivo Effect of Kinase Inhibitors on Leukemic Cells
  • mice Female recipient mice are lethally irradiated from a ⁇ source in two doses about 4 hr apart, with approximately 5Gy each. About lhr after the second radiation dose, mice are injected i.v. with about IxIO 6 leukemic cells (e.g. Ph+ human or murine cells, or pi 90 transduced bone marrow cells). These cells are administered together with a radioprotective dose of about 5x10* normal bone marrow cells from 3-5 week old donor mice. Recipients are given antibiotics in the water and monitored daily. Mice who become sick after about 14 days are euthanized and lymphoid organs are harvested for analysis. Kinase inhibitor treatment begins about 10 days after leukemic cell injection and continues daily until the mice become sick or a maximum of approximately 35 days post-transplant. Inhibitors are given by oral lavage.
  • IxIO 6 leukemic cells e.g. Ph+ human or murine cells, or pi 90 transduced bone marrow cells. These cells are
  • Peripheral blood cells are collected approximately on day 10 (pre-treatment) and upon euthanization
  • Example 26 Treatment of Lupus Disease Model Mice
  • Mice lacking the inhibitory receptor Fc ⁇ RIIb that opposes PDK signaling in B cells develop lupus with high penetrance.
  • Fc ⁇ RIIb knockout mice R2KO, Jackson Labs
  • R2KO Jackson Labs
  • Fc ⁇ RIIb knockout mice are considered a valid model of the human disease as some lupus patients show decreased expression or function of Fc ⁇ RIIb (S. Bolland and J. V. Ravtech 2000. Immunity 12:277-285).
  • the R2KO mice develop lupus-like disease with anti-nuclear antibodies, glomerulonephritis and proteinurea within about 4-6 months of age.
  • the rapamycin analogue RADOOl available from LC Laboratories
  • This compound has been shown to ameliorate lupus symptoms in the B6.Slelz.Sle3z model (T. Wu et al. J. Clin Invest. 117:2186-2196).
  • mice such as R2KO, BXSB or MLR/lpr are treated at about 2 months old, approximately for about two months.
  • Mice are given doses of: vehicle, RADOOl at about 10mg/kg, or compounds disclosed herein at approximately 1 mg/kg to about 500 mg/kg.
  • Blood and urine samples are obtained at approximately throughout the testing period, and tested for antinuclear antibodies (in dilutions of serum) or protein concentration (in urine). Serum is also tested for anti-ssDNA and anti-dsDNA antibodies by ELISA. Animals are euthanized at day 60 and tissues harvested for measuring spleen weight and kidney disease. Glomerulonephritis is assessed in kidney sections stained with H&E. Other animals are studied for about two months after cessation of treatment, using the same endpoints.
  • Example 27 Murine Bone Marrow Transplant Assay
  • mice Female recipient mice are lethally irradiated from a ⁇ ray source. About lhr after the radiation dose, mice are injected with about 1x106 leukemic cells from early passage pl90 transduced cultures (e.g. as described in Cancer Genet Cytogenet. 2005 Aug;161(l):51-6) . These cells are administered together with a radioprotective dose of approximately 5x106 normal bone marrow cells from 3-5 wk old donor mice. Recipients are given antibiotics in the water and monitored daily. Mice who become sick after about 14 days are euthanized and lymphoid organs harvested for flow cytometry and/or magnetic enrichment.
  • Treatment begins on approximately day 10 and continues daily until mice become sick, or after a maximum of about 35 days post-transplant. Drugs are given by oral gavage (p.o.).
  • chemotherapeutic that is not curative but delays leukemia onset by about one week or less is identified; controls are vehicle- treated or treated with chemotherapeutic agent, previously shown to delay but not cure leukemogenesis in this model (e.g. imatinib at about 70mg/kg twice daily).
  • pi 90 cells that express eGFP are used, and postmortem analysis is limited to enumeration of the percentage of leukemic cells in bone marrow, spleen and lymph node (LN) by flow cytometry.
  • pi 90 cells that express a tailless form of human CD4 are used and the postmortem analysis includes magnetic sorting of hCD4+ cells from spleen followed by immunoblot analysis of key signaling endpoints: p Akt -T3O8 and S473; pS6 and p4EBP-l.
  • sorted cells are incubated in the presence or absence of kinase inhibitors of the present disclosure inhibitors before lysis.
  • "phosfiow" is used to detect p Akt -S473 and pS6-S235/236 in hCD4-gated cells without prior sorting.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Epidemiology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Rheumatology (AREA)
  • Pulmonology (AREA)
  • Pain & Pain Management (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Transplantation (AREA)
  • Cardiology (AREA)
  • Hematology (AREA)
  • Oncology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Dermatology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Nitrogen And Oxygen Or Sulfur-Condensed Heterocyclic Ring Systems (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The present invention provides heterocyclic compounds for use as kinase inhibitors and in other applications. Also provided are pharmaceutical compositions and methods of treatments of diseases and conditions associated with P13 kinase activity.

Description

HETEROCYCLIC KINASE INHIBITORS
[0001] This application claims the benefit of U.S. Provisional Application Serial No. 61/194,310 filed on September 26, 2008, which is hereby incorporated by reference in its entirety for all purposes.
BACKGROUND OF THE INVENTION
[0002] The activity of cells can be regulated by external signals that stimulate or inhibit intracellular events. The process by which stimulatory or inhibitory signals are transmitted into and within a cell to elicit an intracellular response is referred to as signal transduction. Over the past decades, cascades of signal transduction events have been elucidated and found to play a central role in a variety of biological responses. Defects in various components of signal transduction pathways have been found to account for a vast number of diseases, including numerous forms of cancer, inflammatory disorders, metabolic disorders, vascular and neuronal diseases (Gaestel et al. Current Medicinal Chemistry (2007) 14:2214-2234).
[0003] Kinases represent a class of important signaling molecules. Kinases can generally be classified into protein kinases and lipid kinases, and certain kinases exhibit dual specificities. Protein kinases are enzymes that phosphorylate other proteins and/or themselves (i.e., autophosphorylation). Protein kinases can be generally classified into three major groups based upon their substrate utilization: tyrosine kinases which predominantly phosphorylate substrates on tyrosine residues (e.g., erb2, PDGF receptor, EGF receptor, VEGF receptor, src, abl), serine/threonine kinases which predominantly phosphorylate substrates on serine and/or threonine residues (e.g., mTorCl, mTorC2, ATM, ATR, DNA-PK, Akt), and dual-specificity kinases which phosphorylate substrates on tyrosine, serine and/or threonine residues.
[0004] Lipid kinases are enzymes that catalyze the phosphorylation of lipids. These enzymes, and the resulting phosphorylated lipids and lipid-derived biologically active organic molecules, play a role in many different physiological processes, including cell proliferation, migration, adhesion, and differentiation. Certain lipid kinases are membrane associated and they catalyze the phosphorylation of lipids contained in or associated with cell membranes. Examples of such enzymes include phosphoinositide(s) kinases (such as PI3-kinases, PI4- Kinases), diacylglycerol kinases, and sphingosine kinases.
[0005] The phosphoinositide 3-kinases (PDKs) signaling pathway is one of the most highly mutated systems in human cancers. PI3K signaling is also a key factor in many other diseases in humans. PI3K signaling is involved in many disease states including allergic contact dermatitis, rheumatoid arthritis, osteoarthritis, inflammatory bowel diseases, chronic obstructive pulmonary disorder, psoriasis, multiple sclerosis, asthma, disorders related to diabetic complications, and inflammatory complications of the cardiovascular system such as acute coronary syndrome.
[0006] PBKs are members of a unique and conserved family of intracellular lipid kinases that phosphorylate the 3 '-OH group on phosphatidylinositols or phosphoinositides. The PI3K family comprises 15 kinases with distinct substrate specificities, expression patterns, and modes of regulation (Katso et al., 2001). The class I PDKs (pi 10a, pi lOβ, pi lOδ, and pi 10γ) are typically activated by tyrosine kinases or G-protein coupled receptors to generate PIP3, which engages downstream effectors such as those in the Akt/PDKl pathway, mTOR, the Tec family kinases, and the Rho family GTPases. The class II and III PD-Ks play a key role in intracellular trafficking through the synthesis of PI(3)P and PI(3,4)P2. The PIKKs are protein kinases that control cell growth (mTORCl) or monitor genomic integrity (ATM, ATR, DNA-PK, and hSmg-1). [0007] The delta (δ) isoform of class I PI3K has been implicated, in particular, in a number of diseases and biological processes. PDK δ is expressed primarily in hematopoietic cells including leukocytes such as T-cells, dendritic cells, neutrophils, mast cells, B-cells, and macrophages. PDK δ is integrally involved in mammalian immune system functions such as T-cell function, B-cell activation, mast cell activation, dendritic cell function, and neutrophil activity. Due to its integral role in immune system function, PDK δ is also involved in a number of diseases related to undesirable immune response such as allergic reactions, inflammatory diseases, inflammation mediated angiogenesis, rheumatoid arthritis, auto-immune diseases such as lupus, asthma, emphysema and other respiratory diseases. Other class I PDK involved in immune system function includes PDK γ, which plays a role in leukocyte signaling and has been implicated in inflammation, rheumatoid arthritis, and autoimmune diseases such as lupus.
[0008] Downstream mediators of the PDK signal transduction pathway include Akt and mammalian target of rapamycin (mTOR). Akt possesses a plckstrin homology (PH) domain that binds PIP3, leading to Akt kinase activation. Akt phosphorylates many substrates and is a central downstream effector of PDK for diverse cellular responses. One important function of Akt is to augment the activity of mTOR, through phosphorylation of TSC2 and other mechanisms. mTOR is a serine-threonine kinase related to the lipid kinases of the PDK family. mTOR has been implicated in a wide range of biological processes including cell growth, cell proliferation, cell motility and survival. Disregulation of the mTOR pathway has been reported in various types of cancer. mTOR is a multifunctional kinase that integrates growth factor and nutrient signals to regulate protein translation, nutrient uptake, autophagy, and mitochondrial function.
[0009] As such, kinases, particularly PDKs are prime targets for drug development. There remains a need for PDK inhibitors suitable for drug development. The present invention addresses this need and provides related advantages as well by providing new classes of kinase inhibitors.
SUMMARY OF THE EVVENTION
[0010] The present invention provides a compound having a structure of one of the following formulae:
Figure imgf000003_0001
[0011] or a pharmaceutically acceptable salt thereof, wherein:
[0012] X5 and X6 are C-R6, N, C- L1 -R1, or N- L'-R1 wherein one of X5 and X6 is C- L'-R1 or N- L'-R1 ; X, is C or N; and X2 and X8 are independently N or C-R6 ; X3 and X7 are C or N, and at least one of X3 and X7 is C; [0013] X4 in Formula I-A or I-C is C or N; and X4 in Formula I-B or I-D is C-R6, NH, or N; [0014] and wherein no more than two adjacent ring atoms are N or NH; [0015] R1 is hydrogen, halogen, -CN, -OR10, -S(O)nR11, -NR12R13, -C(O)R14, alkyl, alkenyl, alkynyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, and n is an integer from 0 to 2; [0016] L1 is a bond, alkylene, heteroalkylene, alkenylene, alkynylene, cycloalkylene, heterocycloalkylene, arylene, or heteroarylene; [0017] R2 and R3 are independently hydrogen, halogen, -CN, -OR10, -S(O)nR1 ', -NR12R13, -C(O)R14, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; [0018] R4 and R5 are independently hydrogen or R6, or R4 and R5 are taken together to form a 5, 6 or 7 membered ring, wherein the 5, 6, or 7 membered ring is optionally substituted with (R6)q; [0019] L is -NH-CR7R8-, - (CR7R8),-, -C=O-, or -CR7R8(C=O) -, -O-, -SO-, or -SO2-; [0020] z is an integer from 0 to 10; [0021] q is an integer from 0 to 5; [0022] each of R6 is independently hydrogen, halogen, -CN, -OR10, -S(O)nR11, -NR12R13, -C(O)R14, alkyl, alkenyl, alkynyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; [0023] each of R7 and R8 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocyclyalkyl, or R7 and R8 taken together form a cycloalkyl, heterocycloalkyl or aryl ring; [0024] R9 is hydrogen, halogen, -CN, -OR10, -S(O)nR1 \ -NR12R13, -C(O)R14, alkyl, alkenyl, alkynyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; [0025] each R10 is independently hydrogen, -C(O)R15, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
[0026] each R11 is independently -NR16R17, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; [0027] each R12 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; [0028] each R13 is independently hydrogen, -S(O)nR18, -C(O)R19, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; [0029] or R12 and R13 are taken together to form a 5, 6, 7, or 8 membered ring, wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom Of -NR12R13; and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted; [0030] each R14 is independently -NR20R21, hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; [0031] each R15 is independently -NR22R23, hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; [0032] each of R16 and R17 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; or R16 and R17 are taken together to form a 5, 6, 7, or 8 membered ring, wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom
Of-NR16R17; and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted; [0033] each R18 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; [0034] each R19 is independently -NR24R25, hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; [0035] each of R20 and R21 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; [0036] or R20 and R21 are taken together to form a 5, 6, 7, or 8 membered ring, wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom Of-NR20R21; and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted; [0037] each R22 and R23 are independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, or R22 and R23 are taken together to form a 5, 6, 7, or 8 membered ring, wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom
Of-NR22R23; and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted;
[0038] each R24 and R25 are independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, or R24 and R25 are taken together to form a 5, 6, 7, or 8 membered ring, wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom
Of-NR24R25; and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted; and
[0039] W is CR6 or N. [0040] In some embodiments, the compound of Formula I- A or I-C is a compound having a structure of one of the following formulae:
Figure imgf000005_0001
[0041] In some embodiments, the compound of Formula I-E is a compound of Formula I-J or Formula I- J- 1 :
Figure imgf000005_0002
[0042] In some embodiments, the compound of Formula I-A is a compound having a structure of one of the following formulae:
Figure imgf000005_0003
[0043] In some embodiments, the compound of Formula I-B or I-D, having a structure of one of the following formulae:
Figure imgf000006_0001
[0044] In another aspect, the invention provides for a compound having is a compound having a structure of the formula
Figure imgf000006_0002
[0045] or a pharmaceutically acceptable salt thereof, wherein:
[0046] X5 and X6 are C-R6, N, C- L1 -R1, or N- L1 -R1 wherein one and no more than one of X5 and X6 is C- L1 -R1 or N-
L'-R1; XI is C or N; and X2 and X8 are independently N, or C-R6; X3 and X7 are C or N, at least one of X3 and
X7 is C;
[0047] X4 in Formula V-A and Formula V-C is C or N; and X4 in Formula V-B and Formula V-D is C-R6, NH, or N; [0048] and wherein no more than two adjacent ring atoms are N or NH; [0049] R1 is hydrogen, halogen, -CN, -OR10, -S(O)nR11, -NR12R13, -C(O)R14, alkyl, alkenyl, alkynyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, and n is an integer from 0 to 2; [0050] L1 is a bond, alkylene, heteroalkylene, alkenylene, alkynylene, cycloalkylene, heterocycloalkylene, arylene, or heteroarylene; [0051] each of R2 and R3 is independently hydrogen, halogen, -CN, -OR10, -S(O)nR11, -NR12R13, -C(O)R14, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; [0052] each of R4 and R5 is independently hydrogen or R6, or R4 and R5 are taken together to form a 5, 6 or 7 membered ring, wherein the 5, 6, or 7 membered ring is optionally substituted with (R6)q; [0053] L is -NH-CR7R8-, - (CR7R8)Z-, -C=O-, or -CR7R8(C=O) -, -O-, -SO-, or -SO2-; [0054] z is an integer from 0 to 10; [0055] q is an integer from 0 to 5; [0056] each R6 is independently hydrogen, halogen, -CN, -OR10, -S(O)nR11, -NR12R13, -C(O)R14, alkyl, alkenyl, alkynyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, wherein n is independently an integer from 0 to 2;
(0057] each of R7 and R8 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocyclyalkyl, or R7 and R8 taken together form a cycloalkyl, heterocycloalkyl or aryl ring;
[0058] R9 is hydrogen, -C(O)R14, alkyl, alkenyl, alkynyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
[0059] each R10 is independently hydrogen, -C(O)R15, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
[0060] each R11 is independently -NR16R17, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
[0061] each R12 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
[0062] each R13 is independently hydrogen, -S(O)nR18, -C(O)R19, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
[0063] or R12 and R13 are taken together to form a 5, 6, 7, or 8 membered ring, wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom Of-NR12R13, and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted;
[0064] each R14 is independently -NR20R21, hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
[0065] each R15 is independently -NR22R23, hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
[0066] each of R16 and R17 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; or R16 and R17 are taken together to form a 5, 6, 7 or 8 membered ring, wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom Of-NR16R17, and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted;
[0067] each R18 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
[0068] each R19 is independently -NR24R25, hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
[0069] each of R20 and R21 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, or R20 and R21 are taken together to form a 5, 6, 7, or 8 membered ring, wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom Of-NR20R21, and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted;
[0070] each of R22 and R23 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, or R22 and R23 are taken together to form a 5, 6, 7, or 8 membered ring, wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom Of-NR22R23, and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted; and
[0071] each of R24 and R25 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, or R24 and R25 are taken together to form a 5, 6, 7, or 8 membered ring wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom Of-NR24R25, and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted.
[0072] In some embodiments, the compound of Formula V-A is a compound having a structure of Formula VI-Al :
Figure imgf000008_0001
[0073] In another aspect, the invention provides for a compound having a structure of one of the following formulae
Figure imgf000008_0002
[0074] or a pharmaceutically acceptable salt thereof, wherein:
[0075] X5 and X6 are C-R6, N, C- L1 -R1 , or N- L1 -R1 wherein one and no more than one of X5 and X6 is C- L1 -R1 or N-
L'-R1;
[0076] Xi is C or N; and X2 and X8 are independently N, or C-R6; [0077] X3 and X7 are C or N, at least one of X3 and X7 is C; [0078] X4 in Formula VII-A and Formula VII-C is C or N; and X4 in Formula VII-B and Formula VII-D is C-R6, NH, or N; and wherein no more than two adjacent ring atoms are N or NH; [0079] R1 is hydrogen, halogen, -CN, -OR10, -S(O)nR11, -NR12R13, -C(O)R14, alkyl, alkenyl, alkynyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, and n is an integer from 0 to 2; [0080] L1 is a bond, alkylene, heteroalkylene, alkenylene, alkynylene, cycloalkylene, heterocycloalkylene, arylene, or heteroarylene; [0081] each of R2 and R3 is independently hydrogen, halogen, -CN, -OR10, -S(O)nR11, -NR12R13, -C(O)R14, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; [0082] each of R4 and R5 is independently hydrogen or R6, or R4 and R5 are taken together to form a 5, 6 or 7 membered ring, wherein the 5, 6, 7, or 8 membered ring is optionally substituted with (R6)q; [0083] L is -NH-CR7R8-, - (CR7R8)Z-, -C=O-, or -CR7R8(C=O) -, -O-, -SO-, or -SO2-; [0084] z is an integer from 0 to 10; [0085] q is an integer from 0 to 5; [0086] each R6 is independently hydrogen, halogen, -CN, -OR10, -S(O)nR11, -NR12R13, -C(O)R14, alkyl, alkenyl, alkynyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, wherein n is independently an integer from 0 to 2; [0087] each of R7 and R8 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocyclyalkyl, or R7 and R8 taken together form a cycloalkyl, heterocycloalkyl or aryl ring;
[0088] R9 is hydrogen, -C(O)R14, alkyl, alkenyl, alkynyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; [0089] each R10 is independently hydrogen, -C(O)R15, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
[0090] each R11 is independently -NR16R17, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
[0091] each R12 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
[0092] each R13 is independently hydrogen, -S(O)nR18, -C(O)R19, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
[0093] or R12 and R13 are taken together to form a 5, 6, 7, or 8 membered ring, wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom Of -NR12R13, and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted;
[0094] each R14 is independently -NR20R21, hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
[0095] each R15 is independently -NR22R23, hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
[0096] each of R16 and R17 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; or R16 and R17 are taken together to form a 5, 6, 7, or 8 membered ring, wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 additional ring heteroatoms selected from N, S, or O in addition to the nitrogen atom Of-NR16R17, and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted;
[0097] each R18 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
[0098] each R19 is independently -NR24R25, hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
[0099] each of R20 and R21 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, or R20 and R21 are taken together to form a 5, 6, 7, or 8 membered ring, wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 additional ring heteroatoms selected from N, S, or O in addition to the nitrogen atom Of-NR20R21, and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted;
[00100] each of R22 and R23 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, or R22 and R23 are taken together to form a 5, 6, 7, or 8 membered ring, wherein the 5, 6, 7, or 8 membered ring contains 0, 1 , 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom Of-NR22R23, and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted; and
[00101] each of R24 and R25 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, or R24 and R25 are taken together to form a 5, 6, 7, or 8 membered ring wherein the 5, 6, 7, or 8 membered ring contains 0, 1 , 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom Of-NR24R25, wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 additional ring heteroatoms selected from N, S, or O, and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted.
[00102] In some embodiments, compound of Formula VII-A is a compound having a structure of Formula VII-Al :
Figure imgf000009_0001
Formula VU-Al
[00103] In some embodiments of a compound of Formula I- A, I-B, I-C, I-D, I-E, I-F, I-G, I-H, I- J, I-J-l, I-K, H-A, II- Al, II- A2, III- A, IV-A, V-A, V-Al, V-B, V-C, V-D, VI-A, VI-Al, VI- A2, VI-B, VI-C, VI-D, VII-A, VII-Al, VII- A2, Vπ-B, Vπ-C, or VII-D , when L is - (CR7R8)Z- and z is 1, then R1 is alkynyl, aryl, or heteroaryl. In some other embodiments, when L is - (CR7R8) z - and z is 1 , then L1 is a bond, and R1 is alkynyl, aryl, or heteroaryl.
[00104] In some embodiments of a compound of Formula I- A, I-B, I-C, I-D, I-E, I-F, I-G, I-H, I-J, I-J-l, I-K, H-A, II- Al, II- A2, III- A, IV-A, V-A, V-Al, V-B, V-C, V-D, VI-A, VI-Al, VI- A2, VI-B, VI-C, VI-D, VII-A, VII-Al, VII-A2, VII-B, VII-C, or VII-D, R9 is -OR10, -NR12R13, -C(O)R14, alkyl, alkenyl, alkynyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl. In some embodiments, when R9 is aryl, then R9 is substituted with halogen, alkyl or heteroalkyl. In other embodiments, wherein when R9 is -NR12R13, then R12 and R13 are taken together to form a 5, 6, 7 or 8 membered ring.
[00105] In some embodiments of a compound of Formula I-A, I-B, I-C, I-D, I-E, I-F, I-G, I-H, I-J, I-K, H-A, II- Al, II- A2, III- A, IV-A, V-A, V-Al, V-B, V-C, V-D, VI-A, VI-Al, VI- A2, VI-B, VI-C, VI-D, VII-A, VII-Al, VII- A2, VII-B, VII-C, or VU-D, R1 is bicyclic heteroaryl.
[00106] In some embodiments of a compound of Formula I-A, I-B, I-C, I-D, I-E, I-F, I-G, I-H, I-J, I-J-l, I-K, H-A, II- Al, II-A2, V-A, V-Al, V-B, V-C, V-D, VI-A, VI-Al, VI- A2, VI-B, VI-C, VI-D, VII-A, VII-Al, VII- A2, VII- B, VII-C, or VII-D, R4 and R5 are taken together to form a 6-membered ring..
[00107] In some embodiments of a compound of Formula I-A, I-C, I-E, I-G, I-J, I-J-l, I-K, H-A, II-A1, II-A2, III-A, IV- A, V-A, V-Al, V-C, VI-A, VI-Al, VI- A2, VI-C, VII-A, VII-Al, VII- A2, or VII-C, R2 is -NH2.
[00108] In some embodiments of a compound of Formula I-A, I-B, I-C, I-D, I-E, I-F, I-G, I-H, II-A, II-A1, II-A2, HI-A, rV-A, V-A, V-B, V-C, V-D, VI-A, VI-Al, VI- A2, VI-B, VI-C, VI-D, VII-A, VII-Al, VII- A2, VII-B, VII-C, or VII-D, R7 is alkyl.
[00109] In some embodiments of a compound of Formula I-A, I-B, I-C, I-D, I-E, I-F, I-G, I-H, I-J, I-J-l, I-K, II-A, II- Al, II- A2, III-A, IV-A, V-A, V-Al, V-B, V-C, V-D, VI-A, VI-B, VI-C, VI-D, VII-A, VII-B, VII-C, or VII-D, R7 and R8 are hydrogen.
[00110] In some embodiments of a compound of Formula I-A, I-B, I-C, I-D, I-E, I-F, I-G, I-H, I-J, I-J-l, I-K, II-A, II- Al, II- A2, III-A, IV-A, V-A, V-Al, V-B, V-C, V-D, VI-A, VI-Al, VI- A2, VI-B, VI-C, or VI-D, W is CH.
[00111] In some embodiments of a compound of Formula I-A, I-B, I-C, I-D, I-E, I-F, I-G, I-H, III-A, IV-A, V-A, V-Al, V-B, V-C, V-D, VII-A, VII-B, VII-C, or VII-D, W is N.
[00112] In some embodiments of a compound of Formula I-A, I-B, I-C, I-D, I-E, I-F, I-G, I-H, I-J, I-J-l, I-K, II-A, II- Al, II- A2, III-A, IV-A, V-A, V-Al, V-B, V-C, V-D, VI-A, VI-Al, VI- A2, VI-B, VI-C, VI-D, VII-A, VII-Al, VII-A2, VII-B, VII-C, or VII-D, R3 is hydrogen.
[00113] In some embodiments of the invention, a compound of Formula I-A, I-B, I-C, I-D, I-E, I-F, I-G, I-H, I-J, I-J-l, I- K, II-A, II-A1, II-A2, III-A, IV-A, V-A, V-Al, V-B, V-C, V-D, VI-A, VI-Al, VI- A2, VI-B, VI-C, VI-D, VII-A, VII-Al, VII- A2, VII-B, VII-C, or VII-D is provided, wherein R9 is halogen, -CN, -
OR10, -S(O)nR", -NR12R13, -C(O)R14, alkyl, alkenyl, alkynyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, and wherein n is independently an integer from 0 to 2. In some embodiments of the invention a compound of Formula Formula I-A, I-B, I-C, I-D, I-E, I-F, I-G, I-H, I-J, I-J-l, I-K, II-A, II-A1, II-A2, IU-A, IV- A, V-A, V-Al, V-B, V-C, V-D, VI-A, VI-Al, VI- A2, VI-B, VI-C, VI-D, VII-A, VU-Al, VU- A2, VII-B, VII-C, or VII-D is provided, wherein R9 is unsubstituted heterocycloalkyl. In some embodiments of the invention a compound of Formula I-A, I-B, I-C, I-D, I-E, I-F, I-G, I-H, I-J, I-J-l, I-K, H-A, II-A1, II-A2, III-A, IV-A, V-A, V-Al, V-B, V-C, V-D, VI-A, VI-Al, VI- A2, VI-B, VI-C, VI-D, VII-A, VII-Al, VII- A2, VII-B, VII-C, or VII- D is provided, wherein R9 is substituted heterocycloalkyl. In some embodiments of the invention a compound of Formula I-A, I-B, I-C, I-D, I-E, I-F, I-G, I-H, I-J, I-J-l, I-K, H.-A, II-A1, II-A2, III-A, IV-A, V-A, V-Al, V-B, V-C, V-D, VI-A, VI-Al, VI- A2, VI-B, VI-C, VI-D, VII-A, VII-Al, VII-A2, VII-B, VII-C, or VII-D is provided, wherein R9 is unsubstituted aryl or substituted aryl. In some embodiments, R9 is aryl substituted with halogen, aryl substituted with alkyl, or aryl substituted with heteroalkyl,. In some embodiments of the invention a compound of Formula I-A, I-B, I-C, I-D, I-E, I-F, I-G, I-H, I-J, I-J-l, I-K, H-A, II-A1, II-A2, III-A, IV-A, V- A, V-Al, V-B, V-C, V-D, VI-A, VI-Al, VI- A2, VI-B, VI-C, VI-D, VII-A, VII-Al, VII- A2, VII-B, VII-C, or VII-D is provided, wherein R9 is -NR12R13. In some embodiments of the invention a compound of Formula I-A, I-B, I-C, I-D, I-E, I-F, I-G, I-H, I-J, I-J-l, I-K, H-A, II-A1, II-A2, III-A, IV-A, V-A, V-Al, V-B, V-C, V-D, VI- A, VI-Al, VI- A2, VI-B, VI-C, VI-D, VII-A, VII-Al, VII- A2, VII-B, VII-C, or VII-D is provided, wherein R12 is -H and R13 is unsubstituted alkyl or cycloalkyl. Alternatively, R12 and R13 are unsubstituted alkyl or cycloalkyl. In some embodiments of the invention a compound of Formula I-A, I-B, I-C, I-D, I-E, I-F, I-G, I-H, I-J, I-J-l, I-K, H-A, II-A1, II-A2, III-A, IV-A, V-A, V-Al, V-B, V-C, V-D, VI-A, VI-Al, VI-A2, VI-B, VI-C, VI-D, VII-A, VII-Al, VII-A2, VII-B, VII-C, or VII-DD is provided, wherein L1 is a bond.
[00114] In some embodiments of the invention a compound of Formula I-A, I-B, I-C, I-D, I-E, I-F, I-G, I-H, I-J, I-J-l, I- K, H-A, II-A1, II- A2, III-A, IV-A, V-A, V-Al, V-B, V-C, V-D, VI-A, VI-Al, VI-A2, VI-B, VI-C, VI-D, VII-A, VII-Al, VII- A2, VII-B, VII-C, or VII-D is provided, wherein R1 is monocyclic aryl, bicyclic aryl, monocyclic heteroaryl, or bicyclic heteroaryl. In some embodiments, R1 is unsubstituted aryl, or substituted aryl. In some embodiments, R1 is aryl substituted with halogen or hydroxy. In other embodiments, R1 is aryl substituted with halogen and hydroxy. In yet other embodiments, R1 is unsubstituted heteroaryl or substituted heteroaryl. Alternatively, R1 is unsubstituted alkynyl or substituted alkynyl. In some embodiments, R1 is alkynyl substituted with hydroxy. In further embodiments, R1 is halogen, -CN, -C(=O)R14, alkyl, or alkenyl.
[00115] In some embodiments of the invention a compound of Formula I-A, I-B, I-C, I-D, I-E, I-F, I-G, I-H, I-J, I-J-l, I- K, H-A, II-A1, II- A2, III-A, IV-A, V-A, V-Al, V-B, V-C, V-D, VI-A, VI-Al, VI- A2, VI-B, VI-C, VI-D, VII-A, VII-Al, VII- A2, VII-B, VII-C, or VII-D is provided wherein R4 and R5 taken together form a 6-membered ring. The 6-membered ring can be substituted with R6. In some embodiments, R6 in a compound of Formula I-A, I-B, I-C, I-D, I-E, I-F, I-G, I-H, I-J, I-J-l, I-K, H-A, II-A1, H.-A2, III-A, IV-A, V-A, V-Al, V-B, V-C, V-D, VI-A, VI-Al, VI- A2, VI-B, VI-C, VI-D, VU-A, VII-Al, VII-A2, VII-B, VII-C, or VII-D, is alkyl. In some embodiments of the invention a compound of Formula I-A, I-B, I-C, I-D, I-E, I-F, I-G, I-H, I-J, I-J-l, I-K, H-A, II-A1, II- A2, IH-A, IV-A, V-A, V-Al, V-B, V-C, V-D, VI-A, VI-Al, VI- A2, VI-B, VI-C, VI-D, VII-A, VII-Al, VII-A2, VII-B, VII-C, or VII-D is provided wherein R2 is -NR12R13. In some embodiments, R2 in a compound of Formula I-A, I-B, I-C, I-D, I-E, I-F, I-G, I-H, I-J, I-J-l, I-K, H-A, U-Al, II- A2, III-A, IV-A, V-A, V-Al, V-B, V-C, V-D, VI-A, VI-Al, VI- A2, VI-B, VI-C, VI-D, VU-A, VII-Al, VII- A2, VII-B, VII-C, or VII-D, is -NH2. In some embodiments of the invention a compound of Formula I-A, I-B, I-C, I-D, I-E, I-F, I-G, I-H, I-J, I-J-l, I- K, H-A, II-A1, II- A2, III-A, IV-A, V-A, V-Al, V-B, V-C, V-D, VI-A, VI-Al, VI- A2, VI-B, VI-C, VI-D, VII-A, VII-Al, VII- A2, VII-B, VII-C, or VII-D is provided wherein R7 is alkyl. In some embodiments of the invention a compound of Formula I-A, I-B, I-C, I-D, I-E, I-F, I-G, I-H, I-J, I-J-l, I-K, II-A, II- Al, II-A2, III-A, IV-A, V- A, V-Al, V-B, V-C, V-D, VI-A, VI-Al, VI- A2, VI-B, VI-C, VI-D, VII-A, VU-Al, VII- A2, VII-B, VII-C, or VII-D is provided wherein R7 and R8 are hydrogen. In some embodiments of the invention a compound of Formula I-A, I-B, I-C, I-D, I-E, I-F, I-G, I-H, I-J, I-J-l, I-K, II-A, II-A1, II-A2, HI-A, IV-A, V-A, V-Al, V-B, V-C, V-D, VI-A, VI-Al, VI- A2, VI-B, VI-C, VI-D, VH-A, VII-Al, VII- A2, VII-B, VII-C, or VII-D is provided wherein W is CH. In some embodiments of the invention a compound of Formula I-A, I-B, I-C, I-D, I-E, I-F, I- G, I-H, I-J, I-J-l, I-K, π-A, II-A1, II-A2, III-A, IV-A, V-A, V-Al, V-B, V-C, V-D, VI-A, VI-Al, VI-A2, VI-B, VI-C, VI-D, VII-A, VII-Al, VII- A2, VII-B, VII-C, or VII-D is provided wherein z is 1. In some embodiments of the invention a compound of Formula I-A, I-B, I-C, I-D, I-E, I-F, I-G, I-H, I-J, I-J-l, I-K, II-A, II-A1, II-A2, III-A, IV-A, V-A, V-Al, V-B, V-C, V-D, VI-A, VI-Al, VI- A2, VI-B, VI-C, VI-D, VII-A, VII-Al, VII-A2, VII- B, VII-C, or VII-D is provided wherein R3 is hydrogen.
[00116] In some embodiments of a compound of Formula I-A, I-B, I-C, I-D, I-E, I-F, I-G, I-H, I-J, I-J-l, I-K, II-A, II- Al, π-A2, III-A, IV-A, V-A, V-Al, V-B, V-C, V-D, VI-A, VI-Al, VI- A2, VI-B, VI-C, VI-D, VII-A, VII-Al, VII- A2, VII-B, VII-C, or VII-D, the compound selectively inhibits one or more members of type I phosphatidylinositol 3-kinases (PI3-kinase) relative to other members of type I PI3-kinase, ascertained by an in vitro kinase assay. In some embodiments, the compound selectively inhibits PI3 -kinase γ and PI3-kinase δ as compared to PI3-kinase β and PI3-kinase α.. In other embodiments, the compound selectively inhibits PI3- kinase δ and PI3-kinase β as compared to PI3-kinase α and PI3-kinase γ.
[00117] In another aspect, the invention provides a pharmaceutical composition comprising a compound of Formula I-A, I-B, I-C, I-D, I-E, I-F, I-G, I-H, I-J, I-J-l, I-K, π-A, II- Al, II-A2, III-A, IV-A, V-A, V-Al, V-B, V-C, V-D, VI-
A, VI-Al, VI- A2, VI-B, VI-C, VI-D, VII-A, VII-Al, VII- A2, VII-B, VII-C, VII-D or a pharmaceutically acceptable salt of a compound of Formula I-A, I-B, I-C, I-D, I-E, I-F, I-G, I-H, I-J, I-J-l, I-K, II-A, U-Al, II- A2, III-A, IV-A, V-A, V-Al, V-B, V-C, V-D, VI-A, VI-Al, VI-A2, VI-B, VI-C, VI-D, VII-A, VII-Al, VII-A2, VII-
B, VII-C, or VII-D, and a pharmaceutically acceptable excipient.
[00118] The invention also provides a method of treating a medical condition mediated by a type I-PI3 kinase (e.g. mediated by pi lOδ, pi lOγ, pi 10a, or pi lOβ kinase), comprising administering to a subject in need thereof a therapeutically effective amount of a compound of Formula I-A, I-B, I-C, I-D, I-E, I-F, I-G, I-H, I-J, I-J-l, I-K, II-A, II-A1, II- A2, III- A, IV-A, V-A, V-Al, V-B, V-C, V-D, VI-A, VI-Al, VI- A2, VI-B, VI-C, VI-D, VII-A, VII-Al, VII-A2, VII-B, VII-C, or VII-D, or a pharmaceutically acceptable salt of a compound of Formula I-A, I-B, I-C, I-D, I-E, I-F, I-G, I-H, I-J, I-J-l, I-K, II-A, II-A1, II-A2, III-A, IV-A, V-A, V-Al, V-B, V-C, V-D, VI- A, VI-Al, VI- A2, VI-B, VI-C, VI-D, VII-A, VII-Al, VII- A2, VII-B, VII-C, or VII-D. In some embodiments, the medical condition is selected from the group consisting of hematologic malignancy, inflammation, autoimmune disease, rheumatoid arthritis, systemic lupus erythematosus, asthma, and cardiovascular disease. In other embodiments, the medical condition is selected from the group consisting of acute myelogenous leukemia, chronic myelogenous leukemia, mastocytosis, chronic lymphocytic leukemia, multiple myeloma, and myelodysplastic syndrome. In other embodiments, the method of treating a medical condition further comprises administering an anti-cancer agent.
[00119] The present invention also provides a method of inhibiting activity of a protein kinase and/or a lipid kinase present in a cell, comprising contacting said cell with an effective amount of a compound of Formula I-A, I-B, I-
C, I-D, I-E, I-F, I-G, I-H, I-J, I-J-l, I-K, II-A, II-A1, II-A2, HJ-A, IV-A, V-A, V-Al, V-B, V-C, V-D, VI-A, VI- Al, VI- A2, VI-B, VI-C, VI-D, VII-A, VII-Al, VII- A2, VU-B, VII-C, or VII-D or a pharmaceutically acceptable salt of a compound of Formula I-A, I-B, I-C, I-D, I-E, I-F, I-G, I-H, I-J, I-J-l, I-K, II-A, II-A1, II-A2, III-A, IV- A, V-A, V-Al, V-B, V-C, V-D, VI-A, VI-Al, VI- A2, VI-B, VI-C, VI-D, VII-A, VII-Al, VII- A2, VII-B, VII-C, or VII-D. INCORPORATION BY REFERENCE
[00120] All publications, patents, and patent applications mentioned in this specification are herein incorporated by reference to the same extent as if each individual publication, patent, or patent application was specifically and individually indicated to be incorporated by reference.
DETAILED DESCRIPTION OF THE INVENTION I. DEFINITIONS
[00121] Unless otherwise stated, structures depicted herein are also meant to include compounds which differ only in the presence of one or more isotopically enriched atoms. For example, compounds having the present structures wherein hydrogen is replaced by deuterium or tritium, or wherein carbon atom is replaced by 13C- or 14C- enriched carbon, are within the scope of this invention.
[00122] The compounds of the present invention may also contain unnatural proportions of atomic isotopes at one or more of atoms that constitute such compounds. For example, the compounds may be radiolabeled with radioactive isotopes, such as for example tritium (3H), iodine-125 (125I) or carbon-14 (14C). All isotopic variations of the compounds of the present invention, whether radioactive or not, are encompassed within the scope of the present invention.
[00123] When ranges are used herein for physical properties, such as molecular weight, or chemical properties, such as chemical formulae, all combinations and subcombinations of ranges and specific embodiments therein are intended to be included. The term "about" when referring to a number or a numerical range means that the number or numerical range referred to is an approximation within experimental variability (or within statistical experimental error), and thus the number or numerical range may vary from, for example, between 1% and 15% of the stated number or numerical range. The term "comprising" (and related terms such as "comprise" or "comprises" or "having" or "including") includes those embodiments, for example, an embodiment of any composition of matter, composition, method, or process, or the like, that "consist of or "consist essentially of the described features.
[00124] As used in the specification and claims, the singular form "a", "an" and "the" includes plural references unless the context clearly dictates otherwise.
[00125] As used herein, "agent" or "biologically active agent" refers to a biological, pharmaceutical, or chemical compound or other moiety. Non-limiting examples include simple or complex organic or inorganic molecule, a peptide, a protein, an oligonucleotide, an antibody, an antibody derivative, antibody fragment, a vitamin derivative, a carbohydrate, a toxin, or a chemotherapeutic compound. Various compounds can be synthesized, for example, small molecules and oligomers (e.g., oligopeptides and oligonucleotides), and synthetic organic compounds based on various core structures. In addition, various natural sources can provide compounds for screening, such as plant or animal extracts, and the like. A skilled artisan can readily recognize that there is no limit as to the structural nature of the agents of the present invention.
[00126] The term "agonist" as used herein refers to a compound having the ability to initiate or enhance a biological function of a target protein, whether by inhibiting the activity or expression of the target protein. Accordingly, the term "agonist" is defined in the context of the biological role of the target polypeptide. While preferred agonists herein specifically interact with (e.g. bind to) the target, compounds that initiate or enhance a biological activity of the target polypeptide by interacting with other members of the signal transduction pathway of which the target polypeptide is a member are also specifically included within this definition. [00127] The terms "antagonist" and "inhibitor" are used interchangeably, and they refer to a compound having the ability to inhibit a biological function of a target protein, whether by inhibiting the activity or expression of the target protein. Accordingly, the terms "antagonist" and "inhibitors" are defined in the context of the biological role of the target protein. While preferred antagonists herein specifically interact with (e.g. bind to) the target, compounds that inhibit a biological activity of the target protein by interacting with other members of the signal transduction pathway of which the target protein is a member are also specifically included within this definition. A preferred biological activity inhibited by an antagonist is associated with the development, growth, or spread of a tumor, or an undesired immune response as manifested in autoimmune disease.
[00128] An "anti-cancer agent", "anti-tumor agent" or "chemotherapeutic agent" refers to any agent useful in the treatment of a neoplastic condition. One class of anti-cancer agents comprises chemotherapeutic agents. "Chemotherapy" means the administration of one or more chemotherapeutic drugs and/or other agents to a cancer patient by various methods, including intravenous, oral, intramuscular, intraperitoneal, intravesical, subcutaneous, transdermal, buccal, or inhalation or in the form of a suppository.
[00129] The term "cell proliferation" refers to a phenomenon by which the cell number has changed as a result of division. This term also encompasses cell growth by which the cell morphology has changed (e.g., increased in size) consistent with a proliferative signal.
[00130] The term "effective amount" or "therapeutically effective amount" refers to that amount of a compound described herein that is sufficient to effect the intended application including but not limited to disease treatment, as defined below. The therapeutically effective amount may vary depending upon the intended application (in vitro or in vivo), or the subject and disease condition being treated, e.g., the weight and age of the subject, the severity of the disease condition, the manner of administration and the like, which can readily be determined by one of ordinary skill in the art. The term also applies to a dose that will induce a particular response in target cells, e.g. reduction of platelet adhesion and/or cell migration. The specific dose will vary depending on the particular compounds chosen, the dosing regimen to be followed, whether it is administered in combination with other compounds, timing of administration, the tissue to which it is administered, and the physical delivery system in which it is carried.
[00131] As used herein, "treatment" or "treating," or "palliating" or "ameliorating" is used interchangeably herein. These terms refer to an approach for obtaining beneficial or desired results including but not limited to therapeutic benefit and/or a prophylactic benefit. By therapeutic benefit is meant eradication or amelioration of the underlying disorder being treated. Also, a therapeutic benefit is achieved with the eradication or amelioration of one or more of the physiological symptoms associated with the underlying disorder such that an improvement is observed in the patient, notwithstanding that the patient may still be afflicted with the underlying disorder. For prophylactic benefit, the compositions may be administered to a patient at risk of developing a particular disease, or to a patient reporting one or more of the physiological symptoms of a disease, even though a diagnosis of this disease may not have been made.
[00132] A "therapeutic effect," as that term is used herein, encompasses a therapeutic benefit and/or a prophylactic benefit as described above. A prophylactic effect includes delaying or eliminating the appearance of a disease or condition, delaying or eliminating the onset of symptoms of a disease or condition, slowing, halting, or reversing the progression of a disease or condition, or any combination thereof.
[00133] The term "pharmaceutically acceptable salt" refers to salts derived from a variety of organic and inorganic counter ions well known in the art. Pharmaceutically acceptable acid addition salts can be formed with inorganic acids and organic acids. Inorganic acids from which salts can be derived include, for example, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like. Organic acids from which salts can be derived include, for example, acetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid, and the like. Pharmaceutically acceptable base addition salts can be formed with inorganic and organic bases. Inorganic bases from which salts can be derived include, for example, sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum, and the like. Organic bases from which salts can be derived include, for example, primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, basic ion exchange resins, and the like, specifically such as isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, and ethanolamine. In some embodiments, the pharmaceutically acceptable base addition salt is chosen from ammonium, potassium, sodium, calcium, and magnesium salts.
[00134] "Pharmaceutically acceptable carrier" or "pharmaceutically acceptable excipient" includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions of the invention is contemplated. Supplementary active ingredients can also be incorporated into the compositions.
[00135] "Signal transduction" is a process during which stimulatory or inhibitory signals are transmitted into and within a cell to elicit an intracellular response. A modulator of a signal transduction pathway refers to a compound which modulates the activity of one or more cellular proteins mapped to the same specific signal transduction pathway. A modulator may augment (agonist) or suppress (antagonist) the activity of a signaling molecule.
[00136] "Radiation therapy" means exposing a patient, using routine methods and compositions known to the practitioner, to radiation emitters such as alpha-particle emitting radionucleotides (e.g., actinium and thorium radionuclides), low linear energy transfer (LET) radiation emitters (i.e. beta emitters), conversion electron emitters (e.g. strontium-89 and samarium- 153-EDTMP, or high-energy radiation, including without limitation x- rays, gamma rays, and neutrons.
[00137] "Prodrug" is meant to indicate a compound that may be converted under physiological conditions or by solvolysis to a biologically active compound described herein. Thus, the term "prodrug" refers to a precursor of a biologically active compound that is pharmaceutically acceptable. A prodrug may be inactive when administered to a subject, but is converted in vivo to an active compound, for example, by hydrolysis. The prodrug compound often offers advantages of solubility, tissue compatibility or delayed release in a mammalian organism (see, e.g., Bundgard, H., Design of Prodrugs (1985), pp. 7-9, 21-24 (Elsevier, Amsterdam). A discussion of prodrugs is provided in Higuchi, T., et al., "Pro-drugs as Novel Delivery Systems," A.C.S. Symposium Series, Vol. 14, and in Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987, both of which are incorporated in full by reference herein. The term "prodrug" is also meant to include any covalently bonded carriers, which release the active compound in vivo when such prodrug is administered to a mammalian subject. Prodrugs of an active compound, as described herein, may be prepared by modifying functional groups present in the active compound in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent active compound. Prodrugs include compounds wherein a hydroxy, amino or mercapto group is bonded to any group that, when the prodrug of the active compound is administered to a mammalian subject, cleaves to form a free hydroxy, free amino or free mercapto group, respectively. Examples of prodrugs include, but are not limited to, acetate, formate and benzoate derivatives of an alcohol or acetamide, formamide and benzamide derivatives of an amine functional group in the active compound and the like.
[00138] "Acyl" refers to a -(C=O)R radical wherein "R" is alkyl, aryl, heteroaryl, heteroalkyl, or heterocyclyl, which are as described herein. In some embodiments, it is a Ci-C10 acyl radical which refers to the total number of chain or ring atoms of the alkyl, aryl, heteroaryl or heterocyclyl portion of the acyloxy group plus the carbonyl carbon of acyl, i.e three other ring or chain atoms plus carbonyl. If the R radical is heteroaryl or heterocyclyl, the hetero ring or chain atoms contribute to the total number of chain or ring atoms. Unless stated otherwise specifically in the specification, the "R" of an acyloxy group is optionally substituted by one or more substituents which independently are: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, trifluoromethyl, trifluoromethoxy, nitro, trimethylsilanyl, -ORa, -
SRa, -OC(O)-R", -N(Ra)2, -C(O)R3, -C(O)OR3, -OC(O)N(Ra)2, -C(O)N(R3)2, -N(Ra)C(O)ORa, -N(Ra)C(O)Ra, - N(R3)C(O)N(R3)2, N(Ra)C(NRa)N(Ra)2, -N(Ra)S(O)tRa (where t is 1 or 2), -S(O)4OR3 (where t is 1 or 2), -S(O)tN(Ra)2 (where t is 1 or 2), or PO3(Ra)2, where each Ra is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl or heteroarylalkyl. "Acyloxy" refers to a R(C=O)O- radical wherein "R" is alkyl, aryl, heteroaryl or heterocyclyl, which are as described herein. In some embodiments, it is a CpC4 acyloxy radical which refers to the total number of chain or ring atoms of the alkyl, aryl, heteroaryl or heterocyclyl portion of the acyloxy group plus the carbonyl carbon of acyl, i.e three other ring or chain atoms plus carbonyl. If the R radical is heteroaryl or heterocyclyl, the hetero ring or chain atoms contribute to the total number of chain or ring atoms. Unless stated otherwise specifically in the specification, the "R" of an acyloxy group is optionally substituted by one or more of the following substituents: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, nitro, oxo, thioxo, trimethylsilanyl, -OR", - SRa, -OC(O)-R8, -N(Ra)2) -C(O)Ra, -C(O)OR3, -C(O)N(R3)2, -N(Ra)C(0)0Ra, -N(Ra)C(0)Ra, -N(Ra)S(0),R3 (where t is 1 or 2), -S(O)1OR3 (where t is 1 or 2) -S(O),N(R3)2 (where t is 1 or 2) , -OPO3WY ( where W and Y are hydrogen, methyl, ethyl, alkyl, lithium, sodium or potassiun) or- OPO3Z ( where Z is calcium, magnesium or iron) where each R" is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl or heteroarylalkyl.
[00139] "Aralkyl" or "arylalkyl" refers to an (aryl)alkyl — radical where aryl and alkyl are as disclosed herein and which are optionally substituted by one or more of the subsituents described as suitable substituents for aryl and alkyl respectively.
[00140] "Alkoxy" refers to a (alkyl)O-radical, where alkyl is as described herein and contains 1 to 10 carbons (e.g., Q- Cio alkyl). Whenever it appears herein, a numerical range such as "1 to 10" refers to each integer in the given range; e.g., "1 to 10 carbon atoms" means that the alkyl group may consist of 1 carbon atom, 2 carbon atoms, 3 carbon atoms, etc., up to and including 10 carbon atoms. In some embodiments, it is a C\-C4 alkoxy group. A alkoxy moiety may be substituted by one or more of the substituents described as suitable substituents for an alkyl radical. [00141] "Alkyl" refers to a straight or branched hydrocarbon chain radical consisting solely of carbon and hydrogen atoms, containing no unsaturation, having from one to ten carbon atoms (e.g., Ci-Ci0 alkyl). Whenever it appears herein, a numerical range such as "1 to 10" refers to each integer in the given range; e.g., "1 to 10 carbon atoms" means that the alkyl group may consist of 1 carbon atom, 2 carbon atoms, 3 carbon atoms, etc., up to and including 10 carbon atoms, although the present definition also covers the occurrence of the term "alkyl" where no numerical range is designated. In some embodiments, it is a Ci-C4 alkyl group. Typical alkyl groups include, but are in no way limited to, methyl, ethyl, propyl, isopropyl, n-butyl, iso-butyl, sec-butyl isobutyl, tertiary butyl, pentyl, isopentyl, neopentyl, hexyl, septyl, octyl, nonyl, decyl, and the like. The alkyl is attached to the rest of the molecule by a single bond, for example, methyl (Me), ethyl (Et), w-propyl, 1-methylethyl (iso-propyl), w-butyl, /i-pentyl, 1,1-dimethylethyl (f-butyl), 3-methylhexyl, 2-methylhexyl, and the like. Unless stated otherwise specifically in the specification, an alkyl group is optionally substituted by one or more of the following substituents: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, nitro, oxo, thioxo, trimethylsilanyl, -ORa, - SR8, -OC(O)-R3, -N(Ra)2, -C(O)R3, -C(O)OR3, -C(O)N(R3)2, -N(Ra)C(O)ORa, -N(Ra)C(O)Ra, -N(Ra)S(O)tRa (where t is 1 or 2), -S(O)1OR3 (where t is 1 or 2) -S(O)tN(R3)2 (where t is 1 or 2) , -OPO3WY ( where W and Y are hydrogen, methyl, ethyl, alkyl, lithium, sodium or potassiun) or- OPO3Z ( where Z is calcium, magnesium or iron) where each R3 is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl or heteroarylalkyl.
[00142] An "alkene" moiety refers to a group consisting of at least two carbon atoms and at least one carbon-carbon double bond, and an "alkyne" moiety refers to a group consisting of at least two carbon atoms and at least one carbon-carbon triple bond. The alkyl moiety, whether saturated or unsaturated, may be branched, straight chain, or cyclic.
[00143] "Alkenyl" refers to a straight or branched hydrocarbon chain radical group consisting solely of carbon and hydrogen atoms, containing at least one double bond, and having from two to ten carbon atoms (ie. C2-Ci0 alkenyl). Whenever it appears herein, a numerical range such as "2 to 10" refers to each integer in the given range; e.g., "2 to 10 carbon atoms" means that the alkenyl group may consist of 2 carbon atoms, 3 carbon atoms, etc., up to and including 10 carbon atoms.In certain embodiments, an alkenyl comprises two to eight carbon atoms. In other embodiments, an alkenyl comprises two to five carbon atoms (e.g., C2-C5 alkenyl). The alkenyl is attached to the rest of the molecule by a single bond, for example, ethenyl (i.e., vinyl), prop-1-enyl (i.e., allyl), but-1-enyl, pent-1-enyl, penta-l,4-dienyl, and the like. Unless stated otherwise specifically in the specification, an alkenyl group is optionally substituted by one or more of the following substituents alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, nitro, oxo, thioxo, trimethylsilanyl, -OR", -
SR", -OC(O)-R8, -N(R")2, -C(O)R8, -C(O)OR8, -C(O)N(R8)2, -N(R8)C(O)ORa, -N(Ra)C(O)Ra, -N(R3)S(O)tR3 (where t is 1 or 2), -S(O)1OR8 (where t is 1 or 2) -S(0),N(Ra)2 (where t is 1 or 2) , -OPO3WY ( where W and Y are hydrogen, methyl, ethyl, alkyl, lithium, sodium or potassiun) or- OPO3Z ( where Z is calcium, magnesium or iron) where each Ra is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl or heteroarylalkyl.
[00144] "Alkynyl" refers to a straight or branched hydrocarbon chain radical group consisting solely of carbon and hydrogen atoms, containing at least one triple bond, having from two to ten carbon atoms (ie. C2-Ci0 alkynyl). Whenever it appears herein, a numerical range such as "2 to 10" refers to each integer in the given range; e.g., "2 to 10 carbon atoms" means that the alkynyl group may consist of 2 carbon atoms, 3 carbon atoms, etc., up to and including 10 carbon atoms. In certain embodiments, an alkynyl comprises two to eight carbon atoms. In other embodiments, an alkynyl has two to five carbon atoms (e.g., C2-C5 alkynyl). The alkynyl is attached to the rest of the molecule by a single bond, for example, ethynyl, propynyl, butynyl, pentynyl, hexynyl, and the like. Unless stated otherwise specifically in the specification, an alkynyl group is optionally substituted by one or more of the following substituents alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, nitro, oxo, thioxo, trimethylsilanyl, -ORa, - SRa, -OC(O)-R8, -N(Ra)2, -C(O)R3, -C(O)OR8, -C(0)N(Ra)2, -N(R8)C(O)ORa, -N(R8)C(O)R8, -N(R8)S(O)tR8 (where t is 1 or 2), -S(O)4OR8 (where t is 1 or 2) -S(O),N(Ra)2 (where t is 1 or 2) , -OPO3WY ( where W and Y are hydrogen, methyl, ethyl, alkyl, lithium, sodium or potassiun) or- OPO3Z ( where Z is calcium, magnesium or iron) where each R8 is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl or heteroarylalkyl.
[00145] "Amino" or "amine" refers to a -N(R8)2 radical group, where each Ra is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl or heteroarylalkyl, unless stated otherwise specifically in the specification. Unless stated otherwise specifically in the specification, an amino group is optionally substituted by one or more substituents which independently are: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, nitro, oxo, thioxo, trimethylsilanyl, -ORa, -
SRa, -OC(O)-R8, -N(Ra)2, -C(O)R8, -C(O)OR8, -C(O)N(R8)2, -N(R8)C(O)ORa, -N(Ra)C(O)R8, -N(Ra)S(O),R8 (where t is 1 or 2), -S(O)1OR8 (where t is 1 or 2) -S(O),N(Ra)2 (where t is 1 or 2) , -OPO3WY (where W and Y are hydrogen, methyl, ethyl, alkyl, lithium, sodium or potassiun) or- OPO3Z ( where Z is calcium, magnesium or iron) where each Ra is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl or heteroarylalkyl.
[00146] "Amide" or "amido" refers to a chemical moiety with formula -C(O)NHR or -NHC(O)R, where R is selected from the group consisting of alkyl, cycloalkyl, aryl, heteroaryl (bonded through a ring carbon) and heteroalicyclic (bonded through a ring carbon). In some embodiments it is a CpC4 amido or amide radical, which includes the amide carbonyl in the total number of carbons in the radical. Unless stated otherwise specifically in the specification, an amino group is optionally substituted independently by one or more of the substituents as described herein for alkyl, cycloalkyl, aryl, heteroaryl, or heterocyclyl. An amide may be an amino acid or a peptide molecule attached to a compound of Formula (I), thereby forming a prodrug. Any amine, hydroxy, or carboxyl side chain on the compounds described herein can be amidified. The procedures and specific groups to make such amides are known to those of skill in the art and can readily be found in reference sources such as Greene and Wuts, Protective Groups in Organic Synthesis, 3.sup.rd Ed., John Wiley & Sons, New York, N.Y., 1999, which is incorporated herein by reference in its entirety.
[00147] "Aromatic" or "aryl" refers to an aromatic radical with six to ten ring atoms (e.g., C6-Ci0 aromatic or C6-C10 aryl) which has at least one ring having a conjugated pi electron system which is carbocyclic (e.g., phenyl, fluorenyl, and naphthyl). Whenever it appears herein, a numerical range such as "6 to 10" refers to each integer in the given range; e.g., "6 to 10 ring atoms" means that the aryl group may consist of 6 ring atoms, 7 ring atoms, etc., up to and including 10 ring atoms. The term includes monocyclic or fused-ring polycyclic (i.e., rings which share adjacent pairs of ring atoms) groups. Unless stated otherwise specifically in the specification, an aryl moiety is optionally substituted by one or more substituents which are independently: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, nitro, oxo, thioxo, trimethylsilanyl, -ORa, -
SRa, -OC(O)-R8, -N(Ra)2, -C(O)R8, -C(O)OR8, -C(O)N(R8)2, -N(Ra)C(0)0Ra, -N(Ra)C(O)Ra, -N(R8)S(0)tRa (where t is 1 or 2), -S(O)tORa (where t is 1 or 2) -S(O),N(Ra)2 (where t is 1 or 2) , -OPO3WY ( where W and Y are hydrogen, methyl, ethyl, alkyl, lithium, sodium or potassiun) or- OPO3Z ( where Z is calcium, magnesium or iron) where each Ra is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl or heteroarylalkyl.
[00148] "Carboxaldehyde" refers to a -(C=O)H radical.
[00149] "Carboxyl" refers to a -(C=O)OH radical.
[00150] "Cyano" refers to a -CN radical.
[00151] "Cycloalkyl" refers to a monocyclic or polycyclic radical that contains only carbon and hydrogen, and may be saturated, or partially unsaturated. Cycloalkyl groups include groups having from 3 to 10 ring atoms (ie. C2-Ci0 cycloalkyl). Whenever it appears herein, a numerical range such as "3 to 10" refers to each integer in the given range; e.g., "3 to 10 carbon atoms" means that the cycloalkyl group may consist of 3 carbon atoms, etc., up to and including 10 carbon atoms. In some embodiments, it is a C3-C5 cycloalkyl radical. Illustrative examples of cycloalkyl groups include, but are not limited to the following moieties: cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloseptyl, cyclooctyl, cyclononyl, cyclodecyl, norbornyl, and the like. Unless stated otherwise specifically in the specification, a cycloalkyl group is optionally substituted by one or more substituents which independently are: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, nitro, oxo, thioxo, trimethylsilanyl, -ORa, -
SR8, -OC(O)-R8, -N(Ra)2, -C(O)R8, -C(O)OR8, -C(O)N(R8)2, -N(Ra)C(O)ORa, -N(Ra)C(O)Ra, -N(Ra)S(O)tRa (where t is 1 or 2), -S(O)1OR8 (where t is 1 or 2) -S(O),N(Ra)2 (where t is 1 or 2) , -OPO3WY ( where W and Y are hydrogen, methyl, ethyl, alkyl, lithium, sodium or potassiun) or- OPO3Z ( where Z is calcium, magnesium or iron) where each Ra is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl or heteroarylalkyl.
[00152] "Ester" refers to a chemical radical of formula -COOR, where R is selected from the group consisting of alkyl, cycloalkyl, aryl, heteroaryl (bonded through a ring carbon) and heteroalicyclic (bonded through a ring carbon). Any amine, hydroxy, or carboxyl side chain on the compounds described herein can be esterified. The procedures and specific groups to make such esters are known to those of skill in the art and can readily be found in reference sources such as Greene and Wuts, Protective Groups in Organic Synthesis, 3.sup.rd Ed., John Wiley & Sons, New York, N.Y., 1999, which is incorporated herein by reference in its entirety. Unless stated otherwise specifically in the specification, an ester group is optionally substituted by one or more substituents which independently are: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, nitro, oxo, thioxo, trimethylsilanyl, -OR8, - SRa, -OC(O)-R8, -N(Ra)2, -C(O)R8, -C(O)OR8, -C(0)N(Ra)2, -N(R8)C(O)ORa, -N(Ra)C(0)Ra, -N(Ra)S(O)tR8 (where t is 1 or 2), -S(O)1OR8 (where t is 1 or 2) -S(O),N(R8)2 (where t is 1 or 2) , -OPO3WY ( where W and Y are hydrogen, methyl, ethyl, alkyl, lithium, sodium or potassiun) or- OPO3Z ( where Z is calcium, magnesium or iron) where each Ra is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl or heteroarylalkyl.
[00153] "Fluoroalkyl" refers to an alkyl radical, as defined above, that is substituted by one or more fluoro radicals, as defined above, for example, trifluoromethyl, difluoromethyl, 2,2,2-trifluoroethyl, l-fluoromethyl-2-fluoroethyl, and the like. The alkyl part of the fluoroalkyl radical may be optionally substituted as defined above for an alkyl group.
[00154) "Halo", "halide", or, alternatively, "halogen" means fluoro, chloro, bromo or iodo. The terms "haloalkyl," "haloalkenyl," "haloalkynyl" and "haloalkoxy" include alkyl, alkenyl, alkynyl and alkoxy structures that are substituted with one or more halo groups or with combinations thereof. For example, the terms "fluoroalkyl" and "fluoroalkoxy" include haloalkyl and haloalkoxy groups, respectively, in which the halo is fluorine.
100155] "Heteroalkyl" "heteroalkenyl" and "heteroalkynyl" include optionally substituted alkyl, alkenyl and alkynyl radicals and which have one or more skeletal chain atoms selected from an atom other than carbon, e.g., oxygen, nitrogen, sulfur, phosphorus or combinations thereof. A numerical range may be given, e.g. C1-C4 heteroalkyl which refers to the chain length in total, which in this example is 4 atoms long. For example, a — CH2OCH2CH3 radical is referred to as a "C4" heteroalkyl, which includes the heteroatom center in the atom chain length description. Connection to the rest of the molecule may be through either a heteroatom or a carbon in the heteroalkyl chain. A heteroalkyl group may be substituted with one or more substituents which independently are: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, nitro, oxo, thioxo, trimethylsilanyl, -ORa, -
SRa, -OC(O)-R8, -N(Ra)2, -C(O)R8, -C(O)OR8, -C(O)N(R8)2, -N(R8)C(O)ORa, -N(Ra)C(O)Ra, -N(R8)S(O)tR8 (where t is 1 or 2), -S(O)4OR8 (where t is 1 or 2) -S(O)tN(R8)2 (where t is 1 or 2) , -OPO3WY ( where W and Y are hydrogen, methyl, ethyl, alkyl, lithium, sodium or potassiun) or- OPO3Z ( where Z is calcium, magnesium or iron) where each Ra is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl or heteroarylalkyl.
[00156] "Heteroaryl" or, alternatively, "heteroaromatic" refers to a 5- to 18-membered aromatic radical (e.g., C5-C13 heteroaryl) that includes one or more ring heteroatoms selected from nitrogen, oxygen and sulfur, and which may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system. Whenever it appears herein, a numerical range such as "5 to 18" refers to each integer in the given range; e.g., "5 to 18 ring atoms" means that the heteroaryl group may consist of 5 ring atoms, 6 ring atoms, etc., up to and including 18 ring atoms. An N- containing "heteroaromatic" or "heteroaryl" moiety refers to an aromatic group in which at least one of the skeletal atoms of the ring is a nitrogen atom. The polycyclic heteroaryl group may be fused or non-fused. The heteroatom(s) in the heteroaryl radical is optionally oxidized. One or more nitrogen atoms, if present, are optionally quaternized. The heteroaryl is attached to the rest of the molecule through any atom of the ring(s). Examples of heteroaryls include, but are not limited to, azepinyl, acridinyl, benzimidazolyl, benzindolyl, 1,3-benzodioxolyl, benzofuranyl, benzooxazolyl, benzofd]thiazolyl, benzothiadiazolyl, benzo[6][l,4]dioxepinyl, benzo[b][l,4]oxazinyl, 1,4-benzodioxanyl, benzonaphthofuranyl, benzoxazolyl, benzodioxolyl, benzodioxinyl, benzoxazolyl, benzopyranyl, benzopyranonyl, benzofuranyl, benzofuranonyl, benzofurazanyl, benzothiazolyl, benzothienyl (benzothiophenyl), benzothieno[3,2-d]pyrimidinyl, benzotriazolyl, benzo[4,6]imidazo[l,2-a]pyridinyl, carbazolyl, cinnolinyl, cyclopenta[d]pyrimidinyl, 6,7-dihydro-5H-cyclopenta[4,5]thieno[2,3-d]pyrimidinyl, 5,6-dihydrobenzo[h]quinazolinyl, 5,6-dihydrobenzo[h]cinnolinyl, 6,7-dihydro-5H-benzo[6,7]cyclohepta[l ,2-c]pyridazinyl, dibenzofuranyl, dibenzothiophenyl, furanyl, furazanyl, furanonyl, furo[3,2-c]pyridinyl,
5,6,7,8,9, 10-hexahydrocycloocta[d]pyrimidinyl, 5,6,7,8,9, 10-hexahydrocycloocta[d]pyridazinyl, 5,6,7,8,9,10-hexahydrocycloocta[d]pyridinyl,isothiazolyl, imidazolyl, indazolyl, indolyl, indazolyl, isoindolyl, indolinyl, isoindolinyl, isoquinolyl, indolizinyl, isoxazolyl, 5,8-methano-5,6,7,8-tetrahydroquinazolinyl, naphthyridinyl, 1,6-naphthyridinonyl, oxadiazolyl, 2-oxoazepinyl, oxazolyl, oxiranyl, 5,6,6a,7,8,9,10,10a-octahydrobenzo[h]quinazolinyl, 1 -phenyl- 1/f-pyrrolyl, phenazinyl, phenothiazinyl, phenoxazinyl, phthalazinyl, pteridinyl, purinyl, pyranyl, pyrrolyl, pyrazolyl, pyrazolo[3,4-d]pyrimidinyl, pyridinyl, pyrido[3,2-d]pyrimidinyl, pyrido[3,4-d]pyrimidinyl, pyrazinyl, pyrimidinyl, pyridazinyl, pyrrolyl, quinazolinyl, quinoxalinyl, quinolinyl, isoquinolinyl, tetrahydroquinolinyl, 5,6,7,8-tetrahydroquinazolinyl, 5,6,7,8-tetrahydrobenzo[4,5]thieno[2,3-d]pyrimidinyl,
6,7,8,9-tetrahydro-5H-cyclohepta[4,5]thieno[2,3-d]ρyrimidinyl, 5,6,7,8-tetrahydroρyrido[4,5-c]pyrida2inyl, thiazolyl, thiadiazolyl, thiapyranyl, triazolyl, tetrazolyl, triazinyl, thieno[2,3-d]pyrimidinyl, thieno[3,2-d]pyrimidinyl, thieno[2,3-c]pridinyl, and thiophenyl {i.e. thienyl). Unless stated otherwise specifically in the specification, a heteraryl moiety is optionally substituted by one or more substituents which are independently: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, nitro, oxo, thioxo, trimethylsilanyl, -ORa, -
SR3, -OC(O)-Ra, -N(Ra)2, -C(O)R8, -C(O)OR3, -C(O)N(Ra)2, -N(Ra)C(O)ORa, -N(Ra)C(O)Ra, -N(Ra)S(O)tRa (where t is 1 or 2), -S(O)1OR" (where t is 1 or 2) -S(O),N(Ra)2 (where t is 1 or 2) , -OPO3WY ( where W and Y are hydrogen, methyl, ethyl, alkyl, lithium, sodium or potassiun) or- OPO3Z ( where Z is calcium, magnesium or iron) where each Ra is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl or heteroarylalkyl.
[00157] "Heteroarylalkyl" or "hetarylalkyl" refers to an (heteroaryl)alkyl — radical where heteroaryl and alkyl are as disclosed herein and which are optionally substituted by one or more of the subsituents described as suitable substituents for aryl and alkyl respectively.
[00158] 'Ηeterocyclyl" or "heterocycloalkyl" refers to a stable 3- to 18-membered non-aromatic ring (e.g., C3-Ci8 heterocyclyl) radical that comprises two to twelve carbon atoms and from one to six heteroatoms selected from nitrogen, oxygen and sulfur. Whenever it appears herein, a numerical range such as "3 to 18" refers to each integer in the given range; e.g., "3 to 18 ring atoms" means that the heteroaryl group may consist of 3 ring atoms, 4 ring atoms, etc., up to and including 18 ring atoms. In some embodiments, it is a C5-Ci0 heterocyclyl. In some embodiments, it is a C4-C]0 heterocyclyl. In some embodiments, it is a C3-Ci0 heterocyclyl. Unless stated otherwise specifically in the specification, the heterocyclyl radical is a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems. The heteroatoms in the heterocyclyl radical may be optionally oxidized. One or more nitrogen atoms, if present, are optionally quaternized. The heterocyclyl radical is partially or fully saturated. The heterocyclyl may be attached to the rest of the molecule through any atom of the ring(s). Examples of such heterocyclyl radicals include, but are not limited to 6,7- dihydro-5H-cyclopenta[b]pyridine, dioxolanyl, thienyl[l,3]dithianyl, decahydroisoquinolyl, imidazolinyl, imidazolidinyl, isothiazolidinyl, isoxazolidinyl, morpholinyl, octahydroindolyl, octahydroisoindolyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolidinyl, oxazolidinyl, piperidinyl, piperazinyl, 4-piperidonyl, pyrrolidinyl, pyrazolidinyl, quinuclidinyl, thiazolidinyl, tetrahydrofuryl, trithianyl, tetrahydropyranyl, thiomorpholinyl, thiamorpholinyl, 1-oxo-thiomorpholinyl, and 1,1-dioxo-thiomorpholinyl. Unless stated otherwise specifically in the specification, a heterocylyl moiety is optionally substituted by one or more substituents which independently are: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, nitro, oxo, thioxo, trimethylsilanyl, -OR", - SRa, -OC(O)-R3, -N(R3)2, -C(O)R3, -C(O)OR3, -C(0)N(Ra)2, -N(R3)C(O)ORa, -N(Ra)C(O)Ra, -N(R3)S(O)tR3 (where t is 1 or 2), -S(O)1OR3 (where t is 1 or 2) -S(O),N(R3)2 (where t is 1 or 2) , -OPO3WY ( where W and Y are hydrogen, methyl, ethyl, alkyl, lithium, sodium or potassiun) or- OPO3Z ( where Z is calcium, magnesium or iron) where each Ra is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl or heteroarylalkyl.
[00159] "Heteroalicyclic" refers to a cycloalkyl radical that includes at least one heteroatom selected from nitrogen, oxygen and sulfur. The radicals may be fused with an aryl or heteroaryl. The term heteroalicyclic also includes all ring forms of the carbohydrates, including but not limited to the monosaccharides, the disaccharides and the oligosaccharides. Unless stated otherwise specifically in the specification, a heteroalicyclic group is optionally substituted by one or more of substituents which are independently: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, nitro, oxo, thioxo, trimethylsilanyl, -ORa, -
SR", -OC(O)-R3, -N(Ra)2, -C(O)R", -C(O)OR8, -C(0)N(Ra)2, -N(Ra)C(O)ORa, -N(Ra)C(O)Ra, -N(Ra)S(O)tRa (where t is 1 or 2), -S(O)1OR8 (where t is 1 or 2) -S(O),N(R8)2 (where t is 1 or 2) , -OPO3WY ( where W and Y are hydrogen, methyl, ethyl, alkyl, lithium, sodium or potassiun) or- OPO3Z ( where Z is calcium, magnesium or iron) where each Ra is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl or heteroarylalkyl.
[00160] "Imino" refers to the =N-H radical.
[00161] "Isocyanato" refers to a -NCO radical.
[00162] "Isothiocyanato" refers to a -NCS radical.
[00163] "Mercaptyl" refers to a (alkyl)S- or (H)S- radical.
[00164] "Moiety" refers to a specific segment or functional group of a molecule. Chemical moieties are often recognized chemical entities embedded in or appended to a molecule.
[00165] "Nitro" refers to the -NO2 radical.
[00166] "Oxa" refers to the -O- radical.
[00167] "Oxo" refers to the =O radical.
[00168] "Sulfinyl" refers to a -S(=O) — R radical, where R is selected from the group consisting of alkyl, cycloalkyl, aryl, heteroaryl (bonded through a ring carbon) and heteroalicyclic (bonded through a ring carbon).
[00169] "Sulfonyl" refers to a -S(=O)2-R radical, where R is selected from the group consisting of alkyl, cycloalkyl, aryl, heteroaryl (bonded through a ring carbon) and heteroalicyclic (bonded through a ring carbon).
[00170] "Sulfonamidyl" or "sulfonamido" refers to a -S(=O)2-NRR radical, where each R is selected independently from the group consisting of hydrogen, alkyl, cycloalkyl, aryl, heteroaryl (bonded through a ring carbon) and heteroalicyclic (bonded through a ring carbon). In some embodiments, it is a CpC4 sulfonamido, wherein each R in sulfonamido contains 1 carbon, 2 carbons, 3 carbons, or 4 carbons total. A sulfonamido group is optionally substituted by one or more of the subsituents described for alkyl, cycloalkyl, aryl, heteroaryl respectively
[00171] "Sulfoxyl" refers to a -S(=O)2OH radical.
[00172] "Sulfonate" refers to a -S(=O)2-OR radical, where R is selected from the group consisting of alkyl, cycloalkyl, aryl, heteroaryl (bonded through a ring carbon) and heteroalicyclic (bonded through a ring carbon). A sulfonategroup is optionally substituted on R by one or more of the subsituents described for alkyl, cycloalkyl, aryl, heteroaryl respectively.
[00173] "Thiocyanato" refers to a -CNS radical.
[00174] "Thioxo" refers to the =S radical. 0175] "Substituted" means that the referenced group may be substituted with one or more additional group(s) individually and independently selected from acyl, alkyl, alkylaryl, cycloalkyl, aralkyl, aryl, carbohydrate, heteroaryl, heterocyclic, hydroxy, alkoxy, aryloxy, mercapto, alkylthio, arylthio, cyano, halo, carbonyl, ester, thiocarbonyl, isocyanato, thiocyanato, isothiocyanato, nitro, perhaloalkyl, perfluoroalkyl, phosphate, silyl, sulfinyl, sulfonyl, sulfonamidyl, sulfoxyl, sulfonate, and amino, including mono- and di-substituted amino groups, and the protected derivatives thereof. The substituents themselves may be substituted, for example, a cycloakyl substituent may have a halide substituted at one or more ring carbons, and the like.The protecting groups that may form the protective derivatives of the above substituents are known to those of skill in the art and may be found in references such as Greene and Wuts, above.
[00176] The compounds presented herein may possess one or more chiral centers and each center may exist in the R or S configuration. The compounds presented herein include all diastereomeric, enantiomeric, and epimeric forms as well as the appropriate mixtures thereof. Stereoisomers may be obtained, if desired, by methods known in the art as, for example, the separation of stereoisomers by chiral chromatographic columns. [00177] The methods and formulations described herein include the use of N-oxides, crystalline forms (also known as polymorphs), or pharmaceutically acceptable salts of compounds having the structure of Formula (I), as well as active metabolites of these compounds having the same type of activity. In addition, the compounds described herein can exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like. The solvated forms of the compounds presented herein are also considered to be disclosed herein. [00178] The term "selective inhibition" or "selectively inhibit" as referred to a biologically active agent refers to the agent's ability to preferentially reduce the target signaling activity as compared to off-target signaling activity, via direct or interact interaction with the target.
[00179] Compounds described can contain one or more asymmetric centers and may thus give rise to diastereomers and optical isomers. The present invention includes all such possible diastereomers as well as their racemic mixtures, their substantially pure resolved enantiomers, all possible geometric isomers, and pharmaceutically acceptable salts thereof. All formulae disclosed here are shown without a definitive stereochemistry at certain positions. The present invention includes all stereoisomers of the shown formulae and pharmaceutically acceptable salts thereof. Further, mixtures of stereoisomers as well as isolated specific stereoisomers are also included. During the course of the synthetic procedures used to prepare such compounds, or in using racemization or epimerization procedures known to those skilled in the art, the products of such procedures can be a mixture of stereoisomers.
[00180] "Isomers" are different compounds that have the same molecular formula. "Stereoisomers" are isomers that differ only in the way the atoms are arranged in space, i.e. having a different stereochemical configuration. "Enantiomers" are a pair of stereoisomers that are non-superimposable mirror images of each other. A 1 : 1 mixture of a pair of enantiomers is a "racemic" mixture. The term "(.±.)" is used to designate a racemic mixture where appropriate. "Diastereoisomers" are stereoisomers that have at least two asymmetric atoms, but which are not mirror-images of each other. The absolute stereochemistry is specified according to the Cahn-Ingold-Prelog R-S system. When a compound is a pure enantiomer the stereochemistry at each chiral carbon can be specified by either R or S. Resolved compounds whose absolute configuration is unknown can be designated (+) or (-) depending on the direction (dextro- or levorotatory) which they rotate plane polarized light at the wavelength of the sodium D line. Certain of the compounds described herein contain one or more asymmetric centers and can thus give rise to enantiomers, diastereomers, and other stereoisomeric forms that can be defined, in terms of absolute stereochemistry, as (R)- or (S)-. The present chemical entities, pharmaceutical compositions and methods are meant to include all such possible isomers, including racemic mixtures, optically pure forms and intermediate mixtures. Optically active (R)- and (S)- isomers can be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques. When the compounds described herein contain olefϊnic double bonds or other centers of geometric asymmetry, and unless specified otherwise, it is intended that the compounds include both E and Z geometric isomers.
[00181] "Moiety" refers to a specific segment or functional group of a molecule. Chemical moieties are often recognized chemical entities embedded in or appended to a molecule.
[00182] A "leaving group or atom" is any group or atom that will, under the reaction conditions, cleave from the starting material, thus promoting reaction at a specified site. Suitable examples of such groups unless otherwise specified are halogen atoms, mesyloxy, p-nitrobenzensulphonyloxy and tosyloxy groups.
[00183] "Protecting group" has the meaning conventionally associated with it in organic synthesis, i.e. a group that selectively blocks one or more reactive sites in a multifunctional compound such that a chemical reaction can be carried out selectively on another unprotected reactive site and such that the group can readily be removed after the selective reaction is complete. A variety of protecting groups are disclosed, for example, in T.H. Greene and P. G. M. Wuts, Protective Groups in Organic Synthesis, Third Edition, John Wiley & Sons, New York (1999). For example, a hydroxy protected form is where at least one of the hydroxy groups present in a compound is protected with a hydroxy protecting group. Likewise, amines and other reactive groups may similarly be protected
[00184] A "therapeutic effect," as that term is used herein, encompasses a therapeutic benefit and/or a prophylactic benefit as described above. A prophylactic effect includes delaying or eliminating the appearance of a disease or condition, delaying or eliminating the onset of symptoms of a disease or condition, slowing, halting, or reversing the progression of a disease or condition, or any combination thereof.
[00185] The term "co-administration," "administered in combination with," and their grammatical equivalents, as used herein, encompass administration of two or more agents to an animal so that both agents and/or their metabolites are present in the animal at the same time. Co-administration includes simultaneous administration in separate compositions, administration at different times in separate compositions, or administration in a composition in which both agents are present.
[00186] The term "in vivo" refers to an event that takes place in a subject's body.
[00187] The term "in vitro" refers to an event that takes places outside of a subject's body. For example, an in vitro assay encompasses any assay run outside of a subject assay. In vitro assays encompass cell-based assays in which cells alive or dead are employed. In vitro assays also encompass a cell-free assay in which no intact cells are employed.
[00188] A "subject," "individual" or "patient" is used interchangeably herein, which refers to a vertebrate, preferably a mammal, more preferably a human. Mammals include, but are not limited to, murines, simians, humans, farm animals, sport animals, and pets. Tissues, cells and their progeny of a biological entity obtained in vitro or cultured in vitro are also encompassed. π. COMPOSITIONS 189] In one aspect, the present invention provides a compound of the formula:
Figure imgf000025_0001
[00190] or a pharmaceutically acceptable salt thereof, wherein:
[00191] X5 and X6 are C-R6, N, C- IΛR1, or N- iΛR1 wherein one of X5 and X6 is C- L'-R1 or N- L'-R1; [00192] X1 is C or N; and X2 and X8 are independently N or C-R6 ; [00193] X3 and X7 are C or N, and at least one of X3 and X7 is C; [00194] X4 in Formula I-A or I-C is C or N; and X4 in Formula I-B or I-D is C-R6, NH, or N; and no more than two adjacent ring atoms are N or NH; [00195] R1 is hydrogen, halogen, -CN, -OR10, -S(O)nR11, -NR12R13, -C(O)R14, alkyl, alkenyl, alkynyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, and n is an integer from 0 to 2; [00196] L1 is a bond, alkylene, heteroalkylene, alkenylene, alkynylene, cycloalkylene, heterocycloalkylene, arylene, or heteroarylene; [00197] R2 and R3 are independently hydrogen, halogen, -CN, -OR10, -S(O)nR11, -NR12R13, -C(O)R14, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; [00198] R4 and R5 are independently hydrogen or R6, or R4 and R5 are taken together to form a 5, 6 or 7-membered ring, wherein the 5, 6, or 7 membered ring is optionally substituted with (R6)q; [00199] L is -NH-CR7R8-, - (CR7R8),-, -C=O-, or -CR7R8(C=O) -, -O-, -SO-, or -SO2-; [00200] z is an integer from 0 to 10; [00201] q is an integer from 0 to 5; [00202] each of R6 is independently hydrogen, halogen, -CN, -OR10, -S(O)nR11, -NR12R13, -C(O)R14, alkyl, alkenyl, alkynyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; [00203] each of R7 and R8 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocyclyalkyl, or R7 and R8 taken together form a cycloalkyl, heterocycloalkyl or aryl ring; [00204] R9 is hydrogen, halogen, -CN, -OR10, -S(O)nR11, -NR12R13, -C(O)R14, alkyl> alkenyl, alkynyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; [00205] each R10 is independently hydrogen, -C(O)R15, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
[00206] each R11 is independently -NR16R17, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; [00207] each R12 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; [00208] each R13 is independently hydrogen, -S(O)nR18, -C(O)R19, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; [00209] or R12 and R13 are taken together to form a 5, 6 or 7-membered ring, wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom Of-NR12R13; and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted; [00210] each R14 is independently -NR20R21, hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; [00211] each R15 is independently -NR22R23, hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; [00212] each of R16 and R17 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; [00213] or R16 and R17 are taken together to form a 5, 6 or 7-membered ring, wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom Of -NR16R17; and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted;
[00214] each R18 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; [00215] each R19 is independently -NR24R25, hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; [00216] each of R20 and R21 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, or R20 and R21 are taken together to form a 5, 6 or 7-membered ring, wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom
Of-NR20R21; and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted; [00217] each of R22 and R23 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, or R22 and R23 are taken together to form a 5, 6 or 7-membered ring, wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom
Of-NR22R23; and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted; [00218] each of R24 and R25 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, or R24 and R25 are taken together to form a 5, 6 or 7-membered ring, wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom
Of-NR24R25; and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted; and [00219] W is C-R6 or N. [00220] In some embodiments, a compound of one of Formula I- A, I-B, I-C, or I-D has a structure of one of the following formulae:
Figure imgf000027_0001
[00221] In some embodiments of the invention, the compound of Formula I-E has a structure of Formula I-J or Formula
I-K:
Figure imgf000027_0002
[00222] In some embodiments, the compound of Formula!- J is a compound of Formula I-J- 1 :
Figure imgf000027_0003
[00223] In some embodiments, the compound of Formula I-E is a compound of Formula H-A, Formula H-A 1 or Formula II-A2:
Figure imgf000027_0004
[00224] In other embodiments, a compound of Formula I-E has a structure of Formula IH-A:
Figure imgf000028_0001
[00225] In other embodiments, a compound of Formula I-E has a structure of Formula IV-A:
Figure imgf000028_0002
[00226] In some embodiments, W is C-R6. In other embodiments, W is CH. In yet other embodiments W is N. [00227] In another aspect, a compound of the invention has a structure of one of the following formulae:
Figure imgf000028_0003
[00228] or a pharmaceutically acceptable salt thereof, wherein: [00229] W is C-R6 or N; [00230] X5 and X6 are C-R6, N, C- L1 -R1, or N- L1 -R1 wherein one and no more than one of X5 and X6 is C- L1 -R1 or N-
L'-R1;
[00231] Xi is C or N; and X2 and X8 are independently N, or C-R6; [00232] X3 and X7 are C or N, at least one of X3 and X7 is C; [00233] X4 in Formula V-A and Formula V-C is C or N; and X4 in Formula V-B and Formula V-D is C-R6, NH, or N; and wherein no more than two adjacent ring atoms are N or NH; [00234] R1 is hydrogen, halogen, -CN, -OR10, -S(O)nR11, -NR12R13, -C(O)R14, alkyl, alkenyl, alkynyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, and n is an integer from 0 to 2; [00235] L1 is a bond, alkylene, heteroalkylene, alkenylene, alkynylene, cycloalkylene, heterocycloalkylene, arylene, or heteroarylene; [00236] each of R2 and R3 is independently hydrogen, halogen, -CN, -OR10, -S(O)nR11, -NR12R13, -C(O)R14, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; [00237] each of R4 and R5 is independently hydrogen or R6, or R4 and R5 are taken together to form a 5, 6 or 7 membered ring, wherein the 5, 6, or 7 membered ring is optionally substituted with (R6)q; [00238] L is -NH-CR7R8-, - (CR7R8),-, -C=O-, or -CR7R8(C=O) -, -O-, -SO-, or -SO2-; [00239] z is an integer from O to 10;
[00240] q is an integer from 0 to 5;
[00241] each R6 is independently hydrogen, halogen, -CN, -OR10, -S(O)nR11, -NR12R13, -C(O)R14, alkyl, alkenyl, alkynyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, wherein n is independently an integer from 0 to 2;
[00242] each of R7 and R8 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocyclyalkyl, or R7 and R8 taken together form a cycloalkyl, heterocycloalkyl or aryl ring;
[00243] R9 is hydrogen, -C(O)R14, alkyl, alkenyl, alkynyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
[00244] each R10 is independently hydrogen, -C(O)R15, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
[00245] each R11 is independently -NR16R17, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
[00246] each R12 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
[00247] each R13 is independently hydrogen, -S(O)nR18, -C(O)R19, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
[00248] or R12 and R13 are taken together to form a 5, 6, 7, or 8 membered ring, wherein the 5, 6, 7, or 8 membered ring contains 0, 1 , 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom of -NR12R13, and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted;
[00249] each R14 is independently -NR20R21, hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
[00250] each R15 is independently -NR22R23, hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
[00251] each of R16 and R17 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; or R16 and R17 are taken together to form a 5, 6, 7 or 8 membered ring, wherein the 5, 6, 7, or 8 membered ring contains 0, 1 , 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom Of -NR16R17, and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted;
[00252] each R18 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
[00253] each R19 is independently -NR24R25, hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
[00254] each of R20 and R21 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, or R20 and R21 are taken together to form a 5, 6, 7, or 8 membered ring, wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom Of-NR20R21, and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted;
[00255] each of R22 and R23 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, or R22 and R23 are taken together to form a 5, 6, 7, or 8 membered ring, wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom Of-NR22R23, and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted; and
[00256] each of R24 and R25 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, or R24 and R25 are taken together to form a 5, 6, 7, or 8 membered ring wherein the 5, 6, 7, or 8 membered ring contains 0, 1 , 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom Of-NR24R25, and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted.
[00257] In some embodiments, the compound of Formula V-A is a compound having the structure of Formula V-Al:
Figure imgf000030_0001
[00258] In some embodiments, the compound of Formula V-A, Formula V-B, Formula V-C or Formula V-D is a compond having a structure of one of the following formulae:
Figure imgf000030_0002
[00259] In some embodiments, a compound of Formula VI-A is a compound having a structure of Formula VI-Al or Formula VI- A2:
Figure imgf000030_0003
[00260] In some other embodiments, the compound of Formula V-A, Formula V-B, Formula V-C or Formula V-D is a compond having a structure of one of the following formulae:
Figure imgf000030_0004
[00261] In some embodiments, a compound of Formula VI-A is a compound having a structure of Formula VI-Al or Formula VI- A2:
Figure imgf000031_0001
[00262] In some embodiments of the compound of Formula I- A, I-B, I-C, I-D, I-E, I-F, I-G, I-H, I-J, I-J-l, I-K, H-A, II- Al, II- A2, IH-A, IV-A, V-A, V-Al, V-B, V-C, V-D, VI-A, VI-Al, VI- A2, VI-B, VI-C, VI-D, VII-A, VII-Al, VII-A2, VII-B, VII-C, or VII-D, L is a group of the formula - (CR7R8)Z-, where z is an integer from 0 to 10. In some embodiments, z is 1, 2 or 3. For example, z may be 1. Each R7 and R8 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocyclyalkyl, or R7 and R8 taken together form a cycloalkyl, heterocycloalkyl or aryl ring. In some embodiments, L is methylene or ethylene. In other embodiments, L is methylene or ethylene substituted with additional alkyl groups such as methyl, ethyl or isopropyl.
[00263] In some embodiments of the compound of Formula I-A, I-B, I-C, I-D, I-E, I-F, I-G, I-H, I-J, I-J-l, I-K, H-A, II- Al, II- A2, HI-A, IV-A, V-A, V-Al, V-B, V-C, V-D, VI-A, VI-Al, VI- A2, VI-B, VI-C, VI-D, VII-A, VII-Al, VII-A2, VII-B, VII-C, or VU-D, R1 is halogen, -CN, -OR10, -S(O)nR11, -NR12R13, -C(O)R14, alkyl, alkenyl, alkynyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, wherein n is independently an integer from 0 to 2. For example, R1 may be an monocyclic aryl group, that can be substituted or is unsubstituted; a bicyclic aryl group, that can be substituted or unsubstituted; a monocyclic heteroaryl group, that can be substituted or is unsubstituted; or a bicyclic heteroaryl group, that can be substituted or is unsubstituted. In other embodiments, R1 may be a monocyclic heterocycloalkyl group, that can be substituted or is unsubstituted; a bicyclic heterocycloalkyl group, that can be substituted or is unsubstituted; a heteroalkyl group, that can be substituted or is unsubstituted; or an alkyl group, that can be substituted or is unsubstituted. In some embodiments, R1 is substituted with halogen, -CN, -OR10, -S(O)nR11, -NR12R13, -C(O)R14, alkyl, alkenyl, alkynyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl. In some embodiments, R1 is an aryl group substituted with -C(O)R14. In other embodiments, R1 is a heteroaryl group substituted with halogen or -NR12R13. When R1 is a monocyclic aromatic or heteroaryl group, substitutions on R1 may be effected, for example, at the ortho, meta and/or para positions. In some embodiments, R1 is monocyclic aryl. In other embodiments, R1 is bicyclic aryl.
[00264] Additional examples of R1 groups are illustrated below :
Figure imgf000031_0002
Figure imgf000032_0001
[0026SJ In some embodiments of the compound of Formula I-A, I-B, I-C, I-D, I-E, I-F, I-G, I-H, I-J, I- J- 1 , 1-K, H-A, II- Al, II- A2, III-A, IV-A, V-A, V-Al, V-B, V-C, V-D, VI-A, VI-Al, VI- A2, VI-B, VI-C, VI-D, VII-A, VII-Al, VII- A2, VII-B, VII-C, or VII-D, L1 may be a bond or a linker connecting R1 to a heterocyclic moiety of the compounds of the invention. In some embodiments, L1 is a bond. In other embodiments, L1 is an alkylene group.
[00266] In some embodiments of the compound of Formula I-A, I-B, I-C, I-D, I-E, I-F, I-G, I-H, I-J, I-J-l, I-K, H-A, II- Al, II-A2, III-A, IV-A, V-A, V-Al, V-B, V-C, V-D, VI-A, VI-Al, VI- A2, VI-B, VI-C, VI-D, VII-A, VII-Al, VII-A2, VII-B, VII-C, or VII-D, R4 and R5 are independently hydrogen. Alternatively, R4 and R5 taken together form a 5, 6 or 7-membered ring, unsubstituted or substituted with (R6)q. In some embodiments, R4 and R5 taken together form a 5 or 6-membered unsubstituted or substituted with (R6)q. The ring may be saturated, unsaturated, or partially unsaturated, and may be aromatic or nonaromatic. In some embodiments, the ring is aromatic and substituted by R6. In some embodiments, R4 and R5 taken together form a group having a structure of one of the folowing formulae:
Figure imgf000033_0001
[00267] In some embodiments of the compound of Formula I-A, I-B, I-C, I-D, I-E, I-F, I-G, I-H, I-J, I-K, H-A, H-Al, II- A2, IH-A, IV-A, V-A, V-Al, V-B, V-C, V-D, VI-A, VI-Al, VI- A2, VI-B, VI-C, VI-D, VII-A, VU-Al , VII- A2, VII-B, VII-C, or VII-D, R6 may be any substituent such as halogen, -CN, -OR10, -S(O)nR11, -NR12R13, -C(O)R14, alkyl, alkenyl, alkynyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, wherein n is independently an integer from 0 to 2. In some embodiments, R6 is halogen. Alternatively, R6 is an alkyl group such as methyl. In further embodiments, R6 is a substituted alkyl group such as CF3.
[00268] In some embodiments of the compound of Formula I-A, I-B, I-C, I-D, I-E, I-F, I-G, I-H, I-J, I-K, H-A, II- Al, II- A2, III- A, IV-A, V-A, V-Al, V-B, V-C, V-D, VI-A, VI-Al, VI- A2, VI-B, VI-C, VI-D, VII-A, VII-Al, VII- A2, VII-B, VII-C, or VII-D, R9 may be any cyclic, linear or branched substituent. In some embodiments, R9 is halogen, -CN, -OR10, -S(O)nR11, -NR12R13, -C(O)R14, alkyl, alkenyl, alkynyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, wherein n is independently an integer from 0 to 2. For example, R9 may be an monocyclic aryl group, substituted or unsubstituted; a bicyclic aryl group, substituted or unsubstituted; a monocyclic heteroaryl group, substituted or unsubstituted; a bicyclic heteroaryl group, substituted or unsubstituted; a monocyclic heterocycloalkyl group, substituted or unsubstituted; a bicyclic heterocycloalkyl group, substituted or unsubstituted; a heteroalkyl group, substituted or unsubstituted; an alkyl group, substituted or unsubstituted. In some embodiments, R9 is substituted with halogen, -CN, -
OR10, -S(O)nR11, -NR12R13, -C(O)R14, alkyl, alkenyl, alkynyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl.
[00269] In some embodiments, R9 is any of the groups shown below:
Figure imgf000033_0002
Figure imgf000034_0001
[00270] In some embodiments of the compound of Formula I-A, I-B, I-C, I-D, I-E, I-F, I-G, I-H, I- J, I- J- 1 , 1-K, H-A, II- Al, II- A2, IH-A, IV-A, V-A, V-Al, V-B, V-C, V-D, VI-A, VI-Al, VI- A2, VI-B, VI-C, VI-D, VII-A, VII-Al, VII- A2, VII-B, VII-C, or VII-D, R12 and R13 in -NR12R13, R16 and R17 in -NR16R17, R20 and R21 in -NR20R21, R22 and R23 in - NR22R23, or R24 and R25 i -NR24R25, are taken together with the nitrogen atom to which they are attached to form a 5-8 membered saturated or unsaturated ring; wherein said ring is independently unsubstituted or is substituted by one or more — NR12R13, hydroxyl, halogen, oxo, aryl, hetaryl, Ci^alkyl, -C(=O)OCi^alkyl , -OPO3H2 or -O-aryl, and wherein said 5-8 membered saturated or unsaturated ring independently contains 0, 1 , or 2 more heteroatoms selected from N, O, or S in addition to the nitrogen.
[00271] In some embodiments, R12 and R13 in -NR12R13, R16 and R17 in -NR16R17, R20 and R21 in -NR20R21, R22 and R23 in -NR22R23, or R24 and R25 i -NR24R25, are taken together with the nitrogen atom to which they are attached to form a moiety of one of the following formulae:
Figure imgf000035_0001
[00272] In some embodiments, the compound of the invention comprises a heterocyclic subunit of the formulas Za or Zb shown below. The symbol ^ represents the point of attachment to the linker L. In specific embodiments, the point of attachment to the linker L is X4 or the exocyclic amine moiety. Additional substituents may be present at any or several of the X1, X2, X4, X5, and X6 positions. For example, an R1 group as defined above may be connected to X6.
Figure imgf000035_0002
Figure imgf000035_0003
Figure imgf000036_0002
Table 1. Heterocyclic subunits of the compounds of the invention. "C" represents -CH= or >C=, and "N" represents -
N=, -N< or -NH- as required for proper valency. Any illustrated subunit Z may be combined with the embodiments disclosed for R1 through R9, L and W to design a compound of the invention
[00273] Non-limiting examples of compounds of the invention are described in more detail in Tables 2-5 below.
Figure imgf000036_0001
Table 2. Exemplary compounds of Formula H-A, wherein X represents Cl, Br, or F.
Figure imgf000037_0001
Figure imgf000038_0001
Figure imgf000039_0001
Figure imgf000040_0001
Figure imgf000041_0001
Figure imgf000042_0002
Figure imgf000042_0001
Figure imgf000043_0001
Figure imgf000044_0001
Figure imgf000045_0001
Figure imgf000046_0001
Figure imgf000047_0001
Figure imgf000048_0002
Table 3. Exemplary compounds of the invention of Formula III-A, whereinX represents a halo group (Cl, Br, I or F).
Figure imgf000048_0001
Formula III-A
Figure imgf000049_0001
Figure imgf000050_0001
Figure imgf000051_0001
Figure imgf000052_0001
Figure imgf000053_0001
Figure imgf000054_0001
Figure imgf000055_0001
Figure imgf000056_0001
Figure imgf000057_0001
Figure imgf000058_0001
Figure imgf000059_0001
Figure imgf000060_0002
Table 4. Exemplary compounds of the invention of Formula IV-A. X represents a halo group (Cl, Br, I or F).
Figure imgf000060_0001
Figure imgf000061_0001
Figure imgf000062_0001
Figure imgf000063_0001
Figure imgf000064_0001
Figure imgf000065_0001
Figure imgf000066_0001
Figure imgf000067_0001
Figure imgf000068_0001
Figure imgf000069_0001
Figure imgf000070_0001
Figure imgf000071_0001
Figure imgf000072_0001
Table 5. Exemplary compounds of the invention of Formula V-Al. X represents a halo group (Cl, Br, I or F).
Figure imgf000073_0001
Figure imgf000073_0002
Figure imgf000074_0001
Figure imgf000075_0001
Figure imgf000076_0001
Figure imgf000077_0001
Figure imgf000078_0001
Figure imgf000079_0001
Figure imgf000080_0001
Figure imgf000081_0001
Figure imgf000082_0001
Figure imgf000083_0001
Figure imgf000084_0001
[00274] Additional non-limiting examples of compounds of the invention are described in Table 6 along with several biological properties.
Figure imgf000084_0002
Figure imgf000085_0001
Figure imgf000086_0001
Figure imgf000087_0001
Figure imgf000088_0001
Figure imgf000089_0001
Figure imgf000090_0001
Figure imgf000091_0001
Figure imgf000092_0001
Figure imgf000093_0001
Figure imgf000094_0001
Figure imgf000095_0001
Figure imgf000096_0001
Figure imgf000097_0001
Figure imgf000098_0001
Figure imgf000099_0001
Figure imgf000100_0001
Figure imgf000101_0001
Figure imgf000102_0001
Figure imgf000103_0001
Figure imgf000104_0001
Figure imgf000105_0001
Figure imgf000106_0001
Figure imgf000107_0001
Figure imgf000108_0001
Figure imgf000109_0001
Figure imgf000110_0001
Figure imgf000111_0001
Figure imgf000112_0001
Figure imgf000113_0002
Table 6. Biological properties of exemplary compounds of the invention. The measured activity of the compounds of the invention (EC50 or IC50) is denoted as "+++" if less or equal to 100 nM, "++" if greater than 100 nM but less than or equal to 1 μM, and "+" if greater than 1 μM. [00275] Additional compounds of the invention are described below:
Figure imgf000113_0001
Preparation of the compounds of the invention
[00276] Compounds of the invention may be synthesized by the general synthetic Schemes 1 and 2 illustrated below. In general synthetic scheme 1 , an aromatic group comprising two reactive groups X and Y is first coupled to the R9 group via the reactive group X. The introduction of the linker L and the heteroaromatic moiety Z is performed in a subsequent step. In general scheme 2, the linker L and heteroaromatic moiety Z are first coupled to the aromatic group comprising two reactive groups X and Y. The coupling step to introduce the R9 moiety is performed subsequently.
[00277] Varioius reagents may be used to perform the coupling reactions necessary to introduce the R9, L and Z moieties. X and Y are reactive groups which can react with precursors of R9 and L or Z moieties. Alternatively, X and Y are latent or protected reaction groups which are converted to reactive groups during or prior to the coupling reactions. Various coupling reactions may be suitable for this purpose. Some known types of coupling reactions which can involve aromatic ring compounds include the Heck reactions and Suzuki reactions. Such reactions are described, for example, in N. Miyaura and A. Suzuki, Chem. Rev. 1995, 95, 2457-2483. [00278] For example, X may be a halogen atom. IfR9 comprises a primary or secondary amino group, it may be attached to the aryl group via an amination reaction. Such a reaction may be performed in the presence of a base and/or a catalyst such as a palladium catalyst. In other embodiments, X is a halogen atom and R9 is an aryl group. A coupling may be effected between X and a precursor compound of R9, such as a boronic acid (Suzuki coupling) or stannane precursor (Stille coupling).
General Scheme 1
Figure imgf000114_0001
General Scheme 2
Figure imgf000114_0002
[00279] The synthesis of several compounds of the invention is illustrated in Synthetic Schemes 3-15. The general synthetic strategy employed in these schemes is believed to be widely applicable to synthesize the compounds of the invention, and the specific compounds and reagents describe serve merely to exemplify the general synthetic approaches disclosed herein. Scheme 3 shows the preparation of a compound of the invention by reduction of a starting aldehyde 3a to an alcohol in step 1. Such a reaction may be performed, for example, by sodium borohydride reduction in methanol. In step 2, compound 3b is coupled to pyrrolidine in the presence of a solvent such as 1 ,4-dioxane. The alcohol 3c is converted to a reactive halide in step 3 by reaction with a reagent such as CBrVPPh3 in a solvent such as dichloromethane. A heterocyclic moiety such as a pyrazolopyrimide (e.g. 3-iodo-lH-pyrazolo[3,4-d]pyrimidin-4-amine) may be coupled to the halide 3d (e.g. in a base such as potassium carbonate and a solvent such as dimethylformamide) to form the compound 3e in step 4. In step 5, the pyrazolopyrimidine subunit is further derivatized via a reaction such as a Suzuki coupling (e.g. using Pd(PPh3)4 as a catalyst) to yield compound 3f.
Scheme 3: Synthesis of 3-(4-amino-l-((2-(pyrrolidin-l-yl)quinoIin-3-yl)methyl)-lH-pyrazolo[3,4-d]pyrimidin-3- yl)-5-fluorophenol (30
Figure imgf000115_0001
Scheme 4 shows an alternative synthesis of the compound 3f. In Step 1, a starting aldehyde 3a is reduced to an alcohol. Such a reaction may be performed, for example, by sodium borohydride reduction in methanol. In Step 2, compound 3b is converted to a reactive halide 4a by reaction with a reagent such as CBr,|/PPh3 in a solvent such as dichloromethane. Coupling to a heterocyclic moiety such as a pyrazolopyrimide (e.g. 3-iodo-lH- pyrazolo[3,4-d]pyrimidin-4-amine) is performed in step 3, for example in a base such as potassium carbonate and a solvent such as dimethylformamide) to yield compound 4b. Compound 4b is then coupled to pyrrolidine in Step 4 in the presence of a solvent such as 1,4-dioxane to produce compound 3e. In step 5, compound 3e is further derivatized via a reaction such as a Suzuki coupling (e.g. using Pd(PPh3)4 as a catalyst) to yield compound 3f.
Scheme 4: Synthesis of 3-(4-amino-l-((2-(pyrrolidin-l-yl)quinolin-3-yl)methyl)-lH-pyrazolo[3,4-d]pyrimidin-3- yl)-5-fluorophenol (3f)
Figure imgf000116_0001
0281] Scheme 5 shows an additional alternative synthesis of the compound 3f. In step 1, a starting aldehyde 3a is reduced to an alcohol. Such a reaction may be performed, for example, by sodium borohydride reduction in methanol. In step 2, compound 3b is converted to a reactive halide 4b by reaction with a reagent such as CBr4ZPPh3 in a solvent such as dichloromethane. Coupling to a heterocyclic moiety such as a derivatized pyrazolopyrimide (e.g. 3-(3-fluoro-5-methoxyphenyl)-lH-pyrazolo[3,4-d]pyrimidin-4-amine) is performed in step 3, for example in a base such as potassium t-butoxide and a solvent such as dimethylformamide) to yield compound 5a. If necessary, the resulting compound 5a is deprotected, for example by removal of a methoxy group by a reagent such as BBr3 in dichloromethane. The resulting compound 5b is then coupled to pyrrolidine in the presence of a solvent such as 1 ,4-dioxane to yield compound 3f .
Scheme 5: Synthesis of 3-(4-amino-l-((2-(pyrrolidin-l-yl)quinolin-3-yl)methyl)-lH-pyrazolo[3,4-d]pyrimidiii-3- yl)-5-fluorophenol (30
Figure imgf000117_0001
0282] Scheme 6 shows the preparation of a compound of the invention by reduction of a starting aldehyde 6a to an alcohol in step 1. Such a reaction may be performed, for example, by sodium borohydride reduction in methanol. In step 2, compound 6b is coupled to an aryl group via, for example, a Suzuki coupling. In the example shown, o-tolylboronic acid is reacted with 6b in the presence of Pd(PPh3)4, a base such as Na2CO3 and a solvent such as DMF. The resulting alcohol 6c is converted to a reactive halide in step 3 by reaction with a reagent such as CBr4ZPPh3 in a solvent such as dichloromethane. A heterocyclic moiety such as a pyrazolopyrimide (e.g. 3-iodo-lH-pyrazolo[3,4-d]pyrimidin-4-amine) may be coupled to the halide 6d (e.g. in a base such as potassium carbonate and a solvent such as dimethylformamide) to form the compound 6e in step 4. In step 5, the pyrazolopyrimidine subunit is further derivatized via a reaction such as a Suzuki coupling (e.g. using Pd(PPh3)4 as a catalyst) to yield compound 6f.
Scheme 6: Synthesis of S-C^amino-l-Wl-o-tolylquinolin-S-yOmethyO-lH-pyrazoloIS^-dlpyrimidin-S-yO-S- fluorophenol (6f)
Figure imgf000118_0001
83] Scheme 7 shows an alternative synthesis of the compound 6f. In step 1, a starting aldehyde 6a is reduced to an alcohol. Such a reaction may be performed, for example, by sodium borohydride reduction in methanol. In step 2, compound 6b is converted to a reactive halide 7a by reaction with a reagent such as CBr4ZPPh3 in a solvent such as dichloromethane. Coupling to a heterocyclic moiety such as a derivatized pyrazolopyrimide (e.g. 3-(3- fluoro-5-methoxyphenyl)-lH-pyrazolo[3,4-d]pyrimidin-4-amine) is performed in step 3, for example in a base such as potassium t-butoxide and a solvent such as dimethylformamide). The resulting compound 7b is then coupled to an aryl group via, for example, a Suzuki coupling. In the example shown, o-tolylboronic acid is reacted with 7b in the presence of Pd(PPh3)4, a base such as Na2CO3 and a solvent such as DMF. If necessary, the resulting compound 7c is deprotected, for example by removal of a methoxy group by a reagent such as BBr3 in dichloromethane to yield compound 6f.
Scheme 7: Synthesis of 3-(4-amino-l-((2-o-tolylquinolin-3-yl)methyl)-lH-pyrazolo[3,4-d]pyrimidin-3-yl)-5- fluorophenol (6f)
Figure imgf000119_0001
[00284] Scheme 8 shows the preparation of a compound of the invention by reduction of a starting acid 8a to an alcohol in step 1. Such a reaction may be performed, for example, by LiAlH4 reduction in methanol. In step 2, compound 8b is coupled to an aryl group via, for example, a Suzuki coupling. In the example shown, 2- fluorophenylboronic acid is reacted with 8b in the presence of Pd(PPh3)4, a base such as Na2CO3 and a solvent such as DME-water. The resulting alcohol 8c is converted to a reactive halide in step 3 by reaction with a reagent such as CBr4ZPPh3 in a solvent such as dichloromethane. A heterocyclic moiety such as a pyrazolopyrimide (e.g. 3-iodo-lH-pyrazolo[3,4-d]pyrimidin-4-amine) may be coupled to the halide 8d (e.g. in a base such as potassium carbonate and a solvent such as dimethylformamide) to form the compound 8e in step 4. In step 5, the pyrazolopyrimidine subunit is further derivatized via a reaction such as a Suzuki coupling (e.g. using Pd(PPh3)4 as a catalyst) to yield compound 8f .
Scheme 8: Synthesis of 3-(4-amino-l-((2-(2-fluorophenyl)-6,7-dihydro-5H-cyclopenta[b]pyridin-3-yl)methyl)-lH- pyrazolo [3,4-d] pyrimidin-3-yl)-5-fluorophenol (80
Figure imgf000120_0001
Scheme 9 shows the preparation of a compound of the invention by reduction of a starting aldehyde 9a to an alcohol in step 1. Such a reaction may be performed, for example, by sodium borohydride reduction in methanol. In step 2, compound 9b is coupled to pyrrolidine in the presence of a solvent such as 1,4-dioxane. The resulting alcohol 9c is converted to a reactive halide in step 3 by reaction with a reagent such as CBr+ZPPh3 in a solvent such as acetonitrile. A heterocyclic moiety such as a pyrazolopyrimide (e.g. 3-iodo-lH-pyrazolo[3,4- d]pyrimidin-4-amine) may be coupled to the halide 9d (e.g. in a base such as potassium carbonate and a solvent such as dimethylformamide) to form the compound 9e in step 4. In step 5, the pyrazolopyrimidine subunit is further derivatized via a reaction such as a Suzuki coupling (e.g. using Pd(PPh3)4 as a catalyst) with 3-fluoro-5- hydroxyphenylboronic acid to yield compound 9f.
Scheme 9: Synthesis of 3-(4-amino-l-((2-(pyrrolidin-l-yl)pyridin-3-yl)methyl)-lH-pyrazolo[3,4-d]pyrimidin-3-yl)-
5-fluorophenol (90
Figure imgf000121_0001
[00286] Scheme 10 illustrates a synthesis of the compound 1Of. In step 1, a starting aldehyde 10a is converted to an alcohol, for example, by Grignard addition of an alkylmagnesium halide reagent. In step 2, compound 10b is converted to a reactive halide by reaction with a reagent such as CBr4/PPh3 in a solvent such as acetonitrile. Coupling to a heterocyclic moiety such as a pyrazolopyrimide (e.g. 3-iodo-lH-pyrazolo[3,4-d]pyrimidin-4- amine) is performed in step 3, for example in a base such as potassium t-butoxide and a solvent such as dimethylformamide). The resulting compound 1Od is then coupled to pyrrolidine in the presence of a solvent such as 1,4-dioxane. In step 5, the pyrazolopyrimidine subunit is further derivatized via a reaction such as a Suzuki coupling (e.g. using Pd(PPh3)4 as a catalyst) to yield compound 1Of.
Scheme 10: Synthesis of 3-(4-amino-l-(l-(2-(pyrrolidin-l-yl)quinolin-3-yl)ethyI)-lH-pyrazolo[3,4-d]pyrimidin-3- yl)-5-fluorophenol (100
Figure imgf000122_0001
[00287] Scheme 11 exemplifies a synthesis of the compound Hf. In step 1, a starting aldehyde Ha is converted to an alcohol, for example, by Grignard addition of an alkylmagnesium halide reagent. In step 2, compound Hb is coupled to to an aryl group via, for example, a Suzuki coupling using phenylboronic acid in the presence of Pd(OAc)2, a base such as Na2CO3 and a solvent such as ethanol/DMF/water. The resulting compounds Hc is converted to a reactive halide by reaction with a reagent such as CBr4ZPPh3 in a solvent such as acetonitrile. Coupling to a heterocyclic moiety such as a pyrazolopyrimide (e.g. 3-iodo-lH-pyrazolo[3,4-d]pyrimidin-4- amine) is performed in step 3, for example in a base such as potassium t-butoxide and a solvent such as dimethylformamide. In step 5, the resulting compound Hd is further derivatized via a reaction such as a Suzuki coupling (e.g. using Pd(PPh3)4 as a catalyst) to yield compound Hf. Scheme 11: Synthesis of 3-(4-amino-l-(l-(2-phenylquinolin-3-yl)ethyl)-lH-pyrazolo[3,4-d]pyrimidin-3-yl)phenol
(Hf)
Figure imgf000123_0001
[00288] Scheme 12 shows a synthesis of the compound 12e. In step 1, a starting aldehyde 12a is converted to an alcohol, for example, by Grignard addition of an alkylmagnesium halide reagent. In step 2, compound 12b is converted to a reactive halide by reaction with a reagent such as CBr4ZPPh3 in a solvent such as acetonitrile. Subsequent reaction with a heterocyclic compound such as 9H-purin-6-amine leads to formation of the intermediate 12d. This intermediae is then coupled to an aryl group via, for example, a Suzuki coupling using phenylboronic acid in the presence of Pd(O Ac)2, a base such as Na2CO3 and a solvent such as ethanol/DMF/water. Alternatively, compound 12d is reacted with pyrrolidine in the presence of a solvent such as 1,4-dioxane, leading to formation of product 12f.
Scheme 12: Synthesis of 9-(l-(2-phenylquinolin-3-yl)ethyl)-9H-purin-6-amine (12e) and 9-(l-(2-(pyrrolidln-l- yl)quinolin-3-yl)ethyl)-9H-purin-6-amine (12f)
Figure imgf000124_0001
289] Scheme 13 describes the synthesis of additional compounds comprising heteroaromatic residues. The starting material is the intermediate 12c prepared as described above. Coupling with a derivatized bicyclic heteroaromatic residue (in the example shown, 9-(tetrahydro-2H-pyran-2-yl)-9H-purin-6-amine) results in compound 13b, which is further reacted with a group such as pyrrolidine (shown) to yield compounds 13c. Deprotection of the purine moiety in HCl/ethanol results in compound 13d. Scheme 13: Synthesis of N-Cl-Cl-Cpyrrolidin-l-ylJquinolin-S-yOethyO-PH-purin-o-ainine (13d)
Figure imgf000125_0001
A synthetic route to quinoxaline compounds of the invention is illustrated in Scheme 14. Quinoxaline 14b is prepared by cyclization of compound 14a with a reagent such as ethyl 3-bromo-2-oxopropanoate. Conversion to the halide 14c is performed using a reagent such as phosphorus oxychloride or similar reagents. Steps 3, 4 and 5 follow the corresponding steps in Scheme 10, yielding compound 14f.
Scheme 14: Synthesis of 3-(4-amino-l-((3-(pyrrolidin-l-yl)quinoxalin-2-yl)methyl)-lH- pyrazolo[3,4-d]pyrimidin-3-yl)-5-fluorophenol (14f)
Figure imgf000126_0001
[00291] Scheme 15 describes the synthesis of compounds such as 15g. Intermediate 15d is prepared as described for compound 4b. Treatment with NaOH in a solvent such as 1,4-dioxane at 8OC yields 15e, which is further alkylated using a reagent such as methyl iodide in potassium t-butoxide/DMF to yield 15f. Compound 15g is obtained following Suzuki coupling as described above.
Scheme 15: Synthesis of 3-((4-amino-3-(3-fluoro-5-hydroxyphenyl)-lH-pyrazolo[3,4-d]pyrimidin-l-yl)methyl)-l- methylquinolin-2(lH)-one (ISg)
Figure imgf000126_0002
Figure imgf000127_0001
A synthetic route to additional quinoxaline compounds of the invention is illustrated in Scheme 16. Quinoxaline 16b is prepared by cyclization of compound 16a with a reagent such as ethyl 3-bromo-2-oxopropanoate in ethanol. Reaction with Boc anhydride in DMAP/CH2C12 leads to the protected intermediate 16c. Coupling to a heterocyclic moiety such as a pyrazolopyrimide is performed in step 3. Deprotection and alkylation, e.g. using methyl iodide, results in compound 16g.
Scheme 16: Synthesis of 3-((4-amino-3-(3-fluoro-5-hydroxyphenyl)-lH-pyrazolo[3,4-d]pyrimidin-l- yl)methyI)-l-methylquinoxalin-2(lH)-one
Figure imgf000128_0001
In some embodiments, one or more subject compounds bind specifically to a PI3 kinase or a protein kinase selected from the group consisting of mTor, DNA-dependent protein kinase DNA-dependent protein kinase (Pubmed protein accession number (PPAN) AAA79184), AbI tyrosine kinase (CAA52387), Bcr-Abl, hemopoietic cell kinase (PPAN CAI19695), Src (PPAN CAA24495), vascular endothelial growth factor receptor 2 (PPAN ABB82619), vascular endothelial growth factor receptor-2 (PPAN ABB82619), epidermal growth factor receptor (PPAN AG43241), EPH receptor B4 (PPAN EAL23820), stem cell factor receptor (PPAN AAF22141), Tyrosine-protein kinase receptor TIE-2 (PPAN Q02858), fins-related tyrosine kinase 3 (PPAN NP 004110), platelet-derived growth factor receptor alpha (PPAN NP_990080), RET (PPAN CAA73131), and any other protein kinases listed in the appended tables and figures, as well as any functional mutants thereof. In some embodiments, the IC50 of a subject compound for pi 10a, pi lOβ, pi lOγ, or pi 105 is less than about 1 uM, less than about 100 nM, less than about 50 nM, less than about 10 nM, less than 1 nM or even less than about 0.5nM. In some embodiments, the IC50 of a subject compound for mTor is less than about 1 uM, less than about 100 nM, less than about 50 nM, less than about 10 nM, less than 1 nM or even less than about 0.5nM. In some other embodiments, one or more subject compounds exhibit dual binding specificity and are capable of inhibiting a PI3 kinase (e.g., a class I PI3 kinease) as well as a protein kinase (e.g., mTor) with an IC50 value less than about 1 uM, less than about 100 nM, less than about 50 nM, less than about 10 nM, less than 1 nM or even less than about 0.5 nM. One or more subject compounds are capable of inhibiting tyrosine kinases including, for example, DNA-dependent protein kinase DNA-dependent protein kinase (Pubmed protein accession number (PPAN) AAA79184), AbI tyrosine kinase (CAA52387), Bcr-Abl, hemopoietic cell kinase (PPAN CAI19695), Src (PPAN CAA24495), vascular endothelial growth factor receptor 2 (PPAN ABB82619), vascular endothelial growth factor receptor-2 (PPAN ABB82619), epidermal growth factor receptor (PPAN AG43241), EPH receptor B4 (PPAN EAL23820), stem cell factor receptor (PPAN AAF22141), Tyrosine- protein kinase receptor TIE-2 (PPAN Q02858), fins-related tyrosine kinase 3 (PPAN NP 004110), platelet- derived growth factor receptor alpha (PPAN NP 990080), RET (PPAN CAA73131), and functional mutants thereof. In some embodiments, the tyrosine kinase is AbI, Bcr-Abl, EGFR, or Flt-3, and any other kinases listed in the Tables herein.
[00294] In some embodiments, the compounds of the present invention exhibits one or more functional characteristics disclosed herein. For example, one or more subject compounds bind specifically to a PI3 kinase. In some embodiments, the IC50 of a subject compound for pi 10a, pi lOβ, pi lOγ, or pl lOδ is less than about 1 uM, less than about 100 nM, less than about 50 nM, less than about 10 nM, less than about 1 nM, less than about 0.5nM, less than about 10OpM, or less than about 50 pM.
[00295] In some embodiments, one or more of the subject compounds may selectively inhibit one or more members of type I or class I phosphatidylinositol 3-kinases (PI3-kinase) with an IC50 value of about 100 nM, 50 nM, 10 nM, 5 nM, 100 pM, 10 pM or 1 pM, or less as measured in an in vitro kinase assay.
[00296] In some embodiments, one or more of the subject compound may selectively inhibit one or two members of type I or class I phosphatidylinositol 3-kinases (PI3-kinase) consisting of PI3-kinase α, PI3-kinase β, PI3-kinase γ, and PI3-kinase δ. In some aspects, some of the subject compounds selectively inhibit PI3-kinase δ as compared to all other type I PI3-kinases. In other aspects, some of the subject compounds selectively inhibit PI3-kinase δ and PI3-kinase γ as compared to the rest of the type I PI3-kinases. In yet other aspects, some of the subject compounds selectively inhibit PI3-kinase α and PI3-kinase β as compared to the rest of the type I PI3-kinases. In still yet some other aspects, some of the subject compounds selectively inhibit PI3-kinase δ and PI3-kinase α as compared to the rest of the type I PI3-kinases. In still yet some other aspects, some of the subject compounds selectively inhibit PI3-kinase δ and PI3-kinase β as compared to the rest of the type I PI3-kinases, or selectively inhibit PI3-kinase δ and PI3-kinase α as compared to the rest of the type I PI3-kinases, or selectively inhibit PI3-kinase α and PI3-kinase γ as compared to the rest of the type I PI3-kinases, or selectively inhibit PI3-kinase γand PI3-kinase β as compared to the rest of the type I PI3-kinases.
[00297] In yet another aspect, an inhibitor that selectively inhibits one or more members of type I PI3-kinases, or an inhibitor that selectively inhibits one or more type I PI3-kinase mediated signaling pathways, alternatively can be understood to refer to a compound that exhibits a 50% inhibitory concentration (IC50) with respect to a given type I PI3-kinase, that is at least at least 10-fold, at least 20-fold, at least 50-fold, at least 100-fold, at least 1000- fold, at least 10,100-fold, or lower, than the inhibitor's IC50 with respect to the rest of the other type I PI3- kinases.
[00298] The invention provides pharmaceutical compositions comprising one or more compounds of the present invention. In some embodiments, the invention provides pharmaceutical compositions for treating diseases or conditions related to an undesirable, over-active, harmful or deleterious immune response in a mammal. Such undesirable immune response can be associated with or result in, e.g., asthma, emphysema, bronchitis, psoriasis, allergy, anaphylaxsis, auto-immune diseases, rhuematoid arthritis, graft versus host disease, and lupus erythematosus. The pharmaceutical compositions of the present invention can be used to treat other respiratory diseases including but not limited to diseases affecting the lobes of lung, pleural cavity, bronchial tubes, trachea, upper respiratory tract, or the nerves and muscle for breathing. [00299] In some embodiments, the invention provides pharmaceutical compositions for the treatment of disorders such as hyperproliferative disorder including but not limited to cancer such as acute myeloid leukemia, thymus, brain, lung, squamous cell, skin, eye, retinoblastoma, intraocular melanoma, oral cavity and oropharyngeal, bladder, gastric, stomach, pancreatic, bladder, breast, cervical, head, neck, renal, kidney, liver, ovarian, prostate, colorectal, esophageal, testicular, gynecological, thyroid, CNS, PNS, AIDS related AIDS-Related (e.g. Lymphoma and Kaposi's Sarcoma) or Viral-Induced cancer. In some embodiments, said pharmaceutical composition is for the treatment of a non-cancerous hyperproliferative disorder such as benign hyperplasia of the skin (e. g., psoriasis), restenosis, or prostate (e. g., benign prostatic hypertrophy (BPH)).
[00300] The invention also provides compositions for the treatment of liver diseases (including diabetes), pancreatitis or kidney disease (including proliferative glomerulonephritis and diabetes- induced renal disease) or pain in a mammal.
[00301] The invention further provides a composition for the prevention of blastocyte implantation in a mammal.
[00302] The invention also relates to a composition for treating a disease related to vasculogenesis or angiogenesis in a mammal which can manifest as tumor angiogenesis, chronic inflammatory disease such as rheumatoid arthritis, inflammatory bowel disease, atherosclerosis, skin diseases such as psoriasis, eczema, and scleroderma, diabetes, diabetic retinopathy, retinopathy of prematurity, age-related macular degeneration, hemangioma, glioma, melanoma, Kaposi's sarcoma and ovarian, breast, lung, pancreatic, prostate, colon and epidermoid cancer.
[00303] The subject pharmaceutical compositions are typically formulated to provide a therapeutically effective amount of a compound of the present invention as the active ingredient, or a pharmaceutically acceptable salt, ester, prodrug, solvate, hydrate or derivative thereof. Where desired, the pharmaceutical compositions contain pharmaceutically acceptable salt and/or coordination complex thereof, and one or more pharmaceutically acceptable excipients, carriers, including inert solid diluents and fillers, diluents, including sterile aqueous solution and various organic solvents, permeation enhancers, solubilizers and adjuvants.
[00304] The subject pharmaceutical compositions can be administered alone or in combination with one or more other agents, which are also typically administered in the form of pharmaceutical compositions. Where desired, the subject compounds and other agent(s) may be mixed into a preparation or both components may be formulated into separate preparations to use them in combination separately or at the same time.
[00305] In some embodiments, the concentration of one or more of the compounds provided in the pharmaceutical compositions of the present invention is less than 100%, 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 19%, 18%, 17%, 16%, 15%,14%, 13%, 12%, 11%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.5%, 0.4%, 0.3%, 0.2%, 0.1%, 0.09%, 0.08%, 0.07%, 0.06%, 0.05%, 0.04%, 0.03%, 0.02%, 0.01%, 0.009%, 0.008%, 0.007%, 0.006%, 0.005%, 0.004%, 0.003%, 0.002%, 0.001%, 0.0009%, 0.0008%, 0.0007%, 0.0006%, 0.0005%, 0.0004%, 0.0003%, 0.0002%, or 0.0001% w/w, w/v or v/v.
[00306] In some embodiments, the concentration of one or more of the compounds of the present invention is greater than 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 19.75%, 19.50%, 19.25% 19%, 18.75%, 18.50%, 18.25% 18%, 17.75%, 17.50%, 17.25% 17%, 16.75%, 16.50%, 16.25% 16%, 15.75%, 15.50%, 15.25% 15%, 14.75%, 14.50%, 14.25% 14%, 13.75%, 13.50%, 13.25% 13%, 12.75%, 12.50%, 12.25% 12%, 11.75%, 11.50%, 11.25% 11%, 10.75%, 10.50%, 10.25% 10%, 9.75%, 9.50%, 9.25% 9%, 8.75%, 8.50%, 8.25% 8%, 7.75%, 7.50%, 7.25% 7%, 6.75%, 6.50%, 6.25% 6%, 5.75%, 5.50%, 5.25% 5%, 4.75%, 4.50%, 4.25%, 4%, 3.75%, 3.50%, 3.25%, 3%, 2.75%, 2.50%, 2.25%, 2%, 1.75%, 1.50%, 125%, 1%, 0.5%, 0.4%, 0.3%, 0.2%, 0.1%, 0.09%, 0.08%, 0.07%, 0.06%, 0.05%, 0.04%, 0.03%, 0.02%, 0.01%, 0.009%, 0.008%, 0.007%, 0.006%, 0.005%, 0.004%, 0.003%, 0.002%, 0.001%, 0.0009%, 0.0008%, 0.0007%, 0.0006%, 0.0005%, 0.0004%, 0.0003%, 0.0002%, or 0.0001% w/w, w/v, or v/v.
[003071 In some embodiments, the concentration of one or more of the compounds of the present invention is in the range from approximately 0.0001% to approximately 50%, approximately 0.001% to approximately 40 %, approximately 0.01% to approximately 30%, approximately 0.02% to approximately 29%, approximately 0.03% to approximately 28%, approximately 0.04% to approximately 27%, approximately 0.05% to approximately 26%, approximately 0.06% to approximately 25%, approximately 0.07% to approximately 24%, approximately 0.08% to approximately 23%, approximately 0.09% to approximately 22%, approximately 0.1% to approximately 21%, approximately 0.2% to approximately 20%, approximately 0.3% to approximately 19%, approximately 0.4% to approximately 18%, approximately 0.5% to approximately 17%, approximately 0.6% to approximately 16%, approximately 0.7% to approximately 15%, approximately 0.8% to approximately 14%, approximately 0.9% to approximately 12%, approximately 1% to approximately 10% w/w, w/v or v/v. v/v.
[00308] In some embodiments, the concentration of one or more of the compounds of the present invention is in the range from approximately 0.001% to approximately 10%, approximately 0.01% to approximately 5%, approximately 0.02% to approximately 4.5%, approximately 0.03% to approximately 4%, approximately 0.04% to approximately 3.5%, approximately 0.05% to approximately 3%, approximately 0.06% to approximately 2.5%, approximately 0.07% to approximately 2%, approximately 0.08% to approximately 1.5%, approximately 0.09% to approximately 1%, approximately 0.1% to approximately 0.9% w/w, w/v or v/v.
[00309] In some embodiments, the amount of one or more of the compounds of the present invention is equal to or less than 10 g, 9.5 g, 9.0 g, 8.5 g, 8.0 g, 7.5 g, 7.0 g, 6.5 g, 6.0 g, 5.5 g, 5.0 g, 4.5 g, 4.0 g, 3.5 g, 3.0 g, 2.5 g, 2.0 g, 1.5 g, 1.0 g, 0.95 g, 0.9 g, 0.85 g, 0.8 g, 0.75 g, 0.7 g, 0.65 g, 0.6 g, 0.55 g, 0.5 g, 0.45 g, 0.4 g, 0.35 g, 0.3 g, 0.25 g, 0.2 g, 0.15 g, 0.1 g, 0.09 g, 0.08 g, 0.07 g, 0.06 g, 0.05 g, 0.04 g, 0.03 g, 0.02 g, 0.01 g, 0.009 g, 0.008 g, 0.007 g, 0.006 g, 0.005 g, 0.004 g, 0.003 g, 0.002 g, 0.001 g, 0.0009 g, 0.0008 g, 0.0007 g, 0.0006 g, 0.0005 g, 0.0004 g, 0.0003 g, 0.0002 g, or 0.0001 g.
[00310] In some embodiments, the amount of one or more of the compounds of the present invention is more than 0.0001 g, 0.0002 g, 0.0003 g, 0.0004 g, 0.0005 g, 0.0006 g, 0.0007 g, 0.0008 g, 0.0009 g, 0.001 g, 0.0015 g, 0.002 g, 0.0025 g, 0.003 g, 0.0035 g, 0.004 g, 0.0045 g, 0.005 g, 0.0055 g, 0.006 g, 0.0065 g, 0.007 g, 0.0075 g, 0.008 g, 0.0085 g, 0.009 g, 0.0095 g, 0.01 g, 0.015 g, 0.02 g, 0.025 g, 0.03 g, 0.035 g, 0.04 g, 0.045 g, 0.05 g, 0.055 g, 0.06 g, 0.065 g, 0.07 g, 0.075 g, 0.08 g, 0.085 g, 0.09 g, 0.095 g, 0.1 g, 0.15 g, 0.2 g, 0.25 g, 0.3 g, 0.35 g, 0.4 g, 0.45 g, 0.5 g, 0.55 g, 0.6 g, 0.65 g, 0.7 g, 0.75 g, 0.8 g, 0.85 g, 0.9 g, 0.95 g, 1 g, 1.5 g, 2 g, 2.5, 3 g, 3.5, 4 g, 4.5 g, 5 g, 5.5 g, 6 g, 6.5g, 7 g, 7.5g, 8 g, 8.5 g, 9 g, 9.5 g, or 10 g.
[00311] In some embodiments, the amount of one or more of the compounds of the present invention is in the range of 0.0001-10 g, 0.0005-9 g, 0.001-8 g, 0.005-7 g, 0.01-6 g, 0.05-5 g, 0.1-4 g, 0.5-4 g, or 1-3 g.
[00312] The compounds according to the invention are effective over a wide dosage range. For example, in the treatment of adult humans, dosages from 0.01 to 1000 mg, from 0.5 to 100 mg, from 1 to 50 mg per day, and from 5 to 40 mg per day are examples of dosages that may be used. An exemplary dosage is 10 to 30 mg per day. The exact dosage will depend upon the route of administration, the form in which the compound is administered, the subject to be treated, the body weight of the subject to be treated, and the preference and experience of the attending physician.
[00313] Described below are non-limiting exemplary pharmaceutical compositions and methods for preparing the same. [00314] Pharmaceutical compositions for oral administration In some embodiments, the invention provides a pharmaceutical composition for oral administration containing a compound of the present invention, and a pharmaceutical excipient suitable for oral administration.
[00315] In some embodiments, the invention provides a solid pharmaceutical composition for oral administration containing: (i) an effective amount of a compound of the present invention; optionally (ii) an effective amount of a second agent; and (iii) a pharmaceutical excipient suitable for oral administration. In some embodiments, the composition further contains: (iv) an effective amount of a third agent.
[00316] In some embodiments, the pharmaceutical composition may be a liquid pharmaceutical composition suitable for oral consumption. Pharmaceutical compositions of the invention suitable for oral administration can be presented as discrete dosage forms, such as capsules, cachets, or tablets, or liquids or aerosol sprays each containing a predetermined amount of an active ingredient as a powder or in granules, a solution, or a suspension in an aqueous or non-aqueous liquid, an oil-in-water emulsion, or a water-in-oil liquid emulsion. Such dosage forms can be prepared by any of the methods of pharmacy, but all methods include the step of bringing the active ingredient into association with the carrier, which constitutes one or more necessary ingredients. In general, the compositions are prepared by uniformly and intimately admixing the active ingredient with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product into the desired presentation. For example, a tablet can be prepared by compression or molding, optionally with one or more accessory ingredients. Compressed tablets can be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as powder or granules, optionally mixed with an excipient such as, but not limited to, a binder, a lubricant, an inert diluent, and/or a surface active or dispersing agent. Molded tablets can be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
[00317] This invention further encompasses anhydrous pharmaceutical compositions and dosage forms comprising an active ingredient, since water can facilitate the degradation of some compounds. For example, water may be added (e.g., 5%) in the pharmaceutical arts as a means of simulating long-term storage in order to determine characteristics such as shelf-life or the stability of formulations over time. Anhydrous pharmaceutical compositions and dosage forms of the invention can be prepared using anhydrous or low moisture containing ingredients and low moisture or low humidity conditions. Pharmaceutical compositions and dosage forms of the invention which contain lactose can be made anhydrous if substantial contact with moisture and/or humidity during manufacturing, packaging, and/or storage is expected. An anhydrous pharmaceutical composition may be prepared and stored such that its anhydrous nature is maintained. Accordingly, anhydrous compositions may be packaged using materials known to prevent exposure to water such that they can be included in suitable formulary kits. Examples of suitable packaging include, but are not limited to, hermetically sealed foils, plastic or the like, unit dose containers, blister packs, and strip packs.
[00318] An active ingredient can be combined in an intimate admixture with a pharmaceutical carrier according to conventional pharmaceutical compounding techniques. The carrier can take a wide variety of forms depending on the form of preparation desired for administration. In preparing the compositions for an oral dosage form, any of the usual pharmaceutical media can be employed as carriers, such as, for example, water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents, and the like in the case of oral liquid preparations (such as suspensions, solutions, and elixirs) or aerosols; or carriers such as starches, sugars, micro-crystalline cellulose, diluents, granulating agents, lubricants, binders, and disintegrating agents can be used in the case of oral solid preparations, in some embodiments without employing the use of lactose. For example, suitable carriers include powders, capsules, and tablets, with the solid oral preparations. If desired, tablets can be coated by standard aqueous or nonaqueous techniques.
[00319] Binders suitable for use in pharmaceutical compositions and dosage forms include, but are not limited to, corn starch, potato starch, or other starches, gelatin, natural and synthetic gums such as acacia, sodium alginate, alginic acid, other alginates, powdered tragacanth, guar gum, cellulose and its derivatives (e.g., ethyl cellulose, cellulose acetate, carboxymethyl cellulose calcium, sodium carboxymethyl cellulose), polyvinyl pyrrolidone, methyl cellulose, pre-gelatinized starch, hydroxypropyl methyl cellulose, microcrystalline cellulose, and mixtures thereof.
[00320] Examples of suitable fillers for use in the pharmaceutical compositions and dosage forms disclosed herein include, but are not limited to, talc, calcium carbonate (e.g., granules or powder), microcrystalline cellulose, powdered cellulose, dextrates, kaolin, mannitol, silicic acid, sorbitol, starch, pre-gelatinized starch, and mixtures thereof.
[00321] Disintegrants may be used in the compositions of the invention to provide tablets that disintegrate when exposed to an aqueous environment. Too much of a disintegrant may produce tablets which may disintegrate in the bottle. Too little may be insufficient for disintegration to occur and may thus alter the rate and extent of release of the active ingredient(s) from the dosage form. Thus, a sufficient amount of disintegrant that is neither too little nor too much to detrimentally alter the release of the active ingredient(s) may be used to form the dosage forms of the compounds disclosed herein. The amount of disintegrant used may vary based upon the type of formulation and mode of administration, and may be readily discernible to those of ordinary skill in the art. About 0.5 to about 15 weight percent of disintegrant, or about 1 to about 5 weight percent of disintegrant, may be used in the pharmaceutical composition. Disintegrants that can be used to form pharmaceutical compositions and dosage forms of the invention include, but are not limited to, agar-agar, alginic acid, calcium carbonate, microcrystalline cellulose, croscarmellose sodium, crospovidone, polacrilin potassium, sodium starch glycolate, potato or tapioca starch, other starches, pre-gelatinized starch, other starches, clays, other algins, other celluloses, gums or mixtures thereof.
[00322] Lubricants which can be used to form pharmaceutical compositions and dosage forms of the invention include, but are not limited to, calcium stearate, magnesium stearate, mineral oil, light mineral oil, glycerin, sorbitol, mannitol, polyethylene glycol, other glycols, stearic acid, sodium lauryl sulfate, talc, hydrogenated vegetable oil (e.g., peanut oil, cottonseed oil, sunflower oil, sesame oil, olive oil, corn oil, and soybean oil), zinc stearate, ethyl oleate, ethylaureate, agar, or mixtures thereof. Additional lubricants include, for example, a syloid silica gel, a coagulated aerosol of synthetic silica, or mixtures thereof. A lubricant can optionally be added, in an amount of less than about 1 weight percent of the pharmaceutical composition.
[00323] When aqueous suspensions and/or elixirs are desired for oral administration, the essential active ingredient therein may be combined with various sweetening or flavoring agents, coloring matter or dyes and, if so desired, emulsifying and/or suspending agents, together with such diluents as water, ethanol, propylene glycol, glycerin and various combinations thereof.
[00324] The tablets can be uncoated or coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a time delay material such as glyceryl monostearate or glyceryl distearate can be employed. Formulations for oral use can also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example, peanut oil, liquid paraffin or olive oil.
[003251 Surfactant which can be used to form pharmaceutical compositions and dosage forms of the invention include, but are not limited to, hydrophilic surfactants, lipophilic surfactants, and mixtures thereof. That is, a mixture of hydrophilic surfactants may be employed, a mixture of lipophilic surfactants may be employed, or a mixture of at least one hydrophilic surfactant and at least one lipophilic surfactant may be employed.
[00326] A suitable hydrophilic surfactant may generally have an HLB value of at least 10, while suitable lipophilic surfactants may generally have an HLB value of or less than about 10. An empirical parameter used to characterize the relative hydrophilicity and hydrophobicity of non-ionic amphiphilic compounds is the hydrophilic-lipophilic balance (" HLB" value). Surfactants with lower HLB values are more lipophilic or hydrophobic, and have greater solubility in oils, while surfactants with higher HLB values are more hydrophilic, and have greater solubility in aqueous solutions. Hydrophilic surfactants are generally considered to be those compounds having an HLB value greater than about 10, as well as anionic, cationic, or zwitterionic compounds for which the HLB scale is not generally applicable. Similarly, lipophilic (i.e., hydrophobic) surfactants are compounds having an HLB value equal to or less than about 10. However, HLB value of a surfactant is merely a rough guide generally used to enable formulation of industrial, pharmaceutical and cosmetic emulsions.
[00327] Hydrophilic surfactants may be either ionic or non-ionic. Suitable ionic surfactants include, but are not limited to, alkylammonium salts; fusidic acid salts; fatty acid derivatives of amino acids, oligopeptides, and polypeptides; glyceride derivatives of amino acids, oligopeptides, and polypeptides; lecithins and hydrogenated lecithins; lysolecithins and hydrogenated lysolecithins; phospholipids and derivatives thereof; lysophospholipids and derivatives thereof; carnitine fatty acid ester salts; salts of alkylsulfates; fatty acid salts; sodium docusate; acylactylates; mono- and di-acetylated tartaric acid esters of mono- and di-glycerides; succinylated mono- and di-glycerides; citric acid esters of mono- and di-glycerides; and mixtures thereof.
[00328] Within the aforementioned group, ionic surfactants include, by way of example: lecithins, lysolecithin, phospholipids, lysophospholipids and derivatives thereof; carnitine fatty acid ester salts; salts of alkylsulfates; fatty acid salts; sodium docusate; acylactylates; mono- and di-acetylated tartaric acid esters of mono- and di- glycerides; succinylated mono- and di-glycerides; citric acid esters of mono- and di-glycerides; and mixtures thereof.
[00329] Ionic surfactants may be the ionized forms of lecithin, lysolecithin, phosphatidylcholine, phosphatidylethanolamine, phosphatidylglycerol, phosphatide acid, phosphatidylserine, lysophosphatidylcholine, lysophosphatidylethanolamine, lysophosphatidylglycerol, lysophosphatidic acid, lysophosphatidylserine, PEG-phosphatidylethanolamine, PVP-phosphatidylethanolamine, lactylic esters of fatty acids, stearoyl-2-lactylate, stearoyl lactylate, succinylated monoglycerides, mono/diacetylated tartaric acid esters of mono/diglycerides, citric acid esters of mono/diglycerides, cholylsarcosine, caproate, caprylate, caprate, laurate, myristate, palmitate, oleate, ricinoleate, linoleate, linolenate, stearate, lauryl sulfate, teracecyl sulfate, docusate, lauroyl carnitines, palmitoyl carnitines, myristoyl carnitines, and salts and mixtures thereof.
[00330] Hydrophilic non-ionic surfactants may include, but not limited to, alkylglucosides; alkylmaltosides; alkylthioglucosides; lauryl macrogolglycerides; polyoxyalkylene alkyl ethers such as polyethylene glycol alkyl ethers; polyoxyalkylene alkylphenols such as polyethylene glycol alkyl phenols; polyoxyalkylene alkyl phenol fatty acid esters such as polyethylene glycol fatty acids monoesters and polyethylene glycol fatty acids diesters; polyethylene glycol glycerol fatty acid esters; polyglycerol fatty acid esters; polyoxyalkylene sorbitan fatty acid esters such as polyethylene glycol sorbitan fatty acid esters; hydrophilic transesterification products of a polyol with at least one member of the group consisting of glycerides, vegetable oils, hydrogenated vegetable oils, fatty acids, and sterols; polyoxyethylene sterols, derivatives, and analogues thereof; polyoxyethylated vitamins and derivatives thereof; polyoxyethylene-polyoxypropylene block copolymers; and mixtures thereof; polyethylene glycol sorbitan fatty acid esters and hydrophilic transesterification products of a polyol with at least one member of the group consisting of triglycerides, vegetable oils, and hydrogenated vegetable oils. The polyol may be glycerol, ethylene glycol, polyethylene glycol, sorbitol, propylene glycol, pentaerythritol, or a saccharide.
[00331] Other hydrophilic-non-ionic surfactants include, without limitation, PEG-IO laurate, PEG-12 laurate, PEG-20 laurate, PEG-32 laurate, PEG-32 dilaurate, PEG-12 oleate, PEG- 15 oleate, PEG-20 oleate, PEG-20 dioleate, PEG-32 oleate, PEG-200 oleate, PEG-400 oleate, PEG- 15 stearate, PEG-32 distearate, PEG-40 stearate, PEG- 100 stearate, PEG-20 dilaurate, PEG-25 glyceryl trioleate, PEG-32 dioleate, PEG-20 glyceryl laurate, PEG-30 glyceryl laurate, PEG-20 glyceryl stearate, PEG-20 glyceryl oleate, PEG-30 glyceryl oleate, PEG-30 glyceryl laurate, PEG-40 glyceryl laurate, PEG-40 palm kernel oil, PEG-50 hydrogenated castor oil, PEG-40 castor oil, PEG-35 castor oil, PEG-60 castor oil, PEG-40 hydrogenated castor oil, PEG-60 hydrogenated castor oil, PEG- 60 corn oil, PEG-6 caprate/caprylate glycerides, PEG-8 caprate/caprylate glycerides, polyglyceryl-10 laurate, PEG-30 cholesterol, PEG-25 phyto sterol, PEG-30 soya sterol, PEG-20 trioleate, PEG-40 sorbitan oleate, PEG- 80 sorbitan laurate, polysorbate 20, polysorbate 80, POE-9 lauryl ether, POE-23 lauryl ether, POE-10 oleyl ether, POE-20 oleyl ether, POE-20 stearyl ether, tocopheryl PEG-100 succinate, PEG-24 cholesterol, polyglyceryl-lOoleate, Tween 40, Tween 60, sucrose monostearate, sucrose monolaurate, sucrose monopalmitate, PEG 10-100 nonyl phenol series, PEG 15-100 octyl phenol series, and poloxamers.
[00332] Suitable lipophilic surfactants include, by way of example only: fatty alcohols; glycerol fatty acid esters; acetylated glycerol fatty acid esters; lower alcohol fatty acids esters; propylene glycol fatty acid esters; sorbitan fatty acid esters; polyethylene glycol sorbitan fatty acid esters; sterols and sterol derivatives; polyoxyethylated sterols and sterol derivatives; polyethylene glycol alkyl ethers; sugar esters; sugar ethers; lactic acid derivatives of mono- and di-glycerides; hydrophobic transesterification products of a polyol with at least one member of the group consisting of glycerides, vegetable oils, hydrogenated vegetable oils, fatty acids and sterols; oil-soluble vitamins/vitamin derivatives; and mixtures thereof. Within this group, preferred lipophilic surfactants include glycerol fatty acid esters, propylene glycol fatty acid esters, and mixtures thereof, or are hydrophobic transesterification products of a polyol with at least one member of the group consisting of vegetable oils, hydrogenated vegetable oils, and triglycerides.
[00333] In one embodiment, the composition may include a solubilizer to ensure good solubilization and/or dissolution of the compound of the present invention and to minimize precipitation of the compound of the present invention. This can be especially important for compositions for non-oral use, e.g., compositions for injection. A solubilizer may also be added to increase the solubility of the hydrophilic drug and/or other components, such as surfactants, or to maintain the composition as a stable or homogeneous solution or dispersion.
[00334] Examples of suitable solubilizers include, but are not limited to, the following: alcohols and polyols, such as ethanol, isopropanol, butanol, benzyl alcohol, ethylene glycol, propylene glycol, butanediols and isomers thereof, glycerol, pentaerythritol, sorbitol, mannitol, transcutol, dimethyl isosorbide, polyethylene glycol, polypropylene glycol, polyvinylalcohol, hydroxypropyl methylcellulose and other cellulose derivatives, cyclodextrins and cyclodextrin derivatives; ethers of polyethylene glycols having an average molecular weight of about 200 to about 6000, such as tetrahydrofurfuryl alcohol PEG ether (glycofurol) or methoxy PEG ; amides and other nitrogen-containing compounds such as 2-pyrrolidone, 2-piperidone, .epsilon.-caprolactam, N- alkylpyrrolidone, N-hydroxyalkylpyrrolidone, N-alkylpiperidone, N-alkylcaprolactam, dimethylacetamide and polyvinylpyrrolidone; esters such as ethyl propionate, tributylcitrate, acetyl triethylcitrate, acetyl tributyl citrate, triethylcitrate, ethyl oleate, ethyl caprylate, ethyl butyrate, triacetin, propylene glycol monoacetate, propylene glycol diacetate, ε-caprolactone and isomers thereof, δ-valerolactone and isomers thereof, β-butyrolactone and isomers thereof; and other solubilizers known in the art, such as dimethyl acetamide, dimethyl isosorbide, N- methyl pyrrolidones, monooctanoin, diethylene glycol monoethyl ether, and water.
[00335] Mixtures of solubilizers may also be used. Examples include, but not limited to, triacetin, triethylcitrate, ethyl oleate, ethyl caprylate, dimethylacetamide, N-methylpyrrolidone, N-hydroxyethylpyrrolidone, polyvinylpyrrolidone, hydroxypropyl methylcellulose, hydroxypropyl cyclodextrins, ethanol, polyethylene glycol 200-100, glycofurol, transcutol, propylene glycol, and dimethyl isosorbide. Particularly preferred solubilizers include sorbitol, glycerol, triacetin, ethyl alcohol, PEG-400, glycofurol and propylene glycol.
[00336] The amount of solubilizer that can be included is not particularly limited. The amount of a given solubilizer may be limited to a bioacceptable amount, which may be readily determined by one of skill in the art. In some circumstances, it may be advantageous to include amounts of solubilizers far in excess of bioacceptable amounts, for example to maximize the concentration of the drug, with excess solubilizer removed prior to providing the composition to a patient using conventional techniques, such as distillation or evaporation. Thus, if present, the solubilizer can be in a weight ratio of 10%, 25%, 50%, 100%, or up to about 200% by weight, based on the combined weight of the drug, and other excipients. If desired, very small amounts of solubilizer may also be used, such as 5%, 2%, 1% or even less. Typically, the solubilizer may be present in an amount of about 1% to about 100%, more typically about 5% to about 25% by weight.
[00337] The composition can further include one or more pharmaceutically acceptable additives and excipients. Such additives and excipients include, without limitation, detackifiers, anti-foaming agents, buffering agents, polymers, antioxidants, preservatives, chelating agents, viscomodulators, tonicifiers, flavorants, colorants, odorants, opacifiers, suspending agents, binders, fillers, plasticizers, lubricants, and mixtures thereof.
[00338] In addition, an acid or a base may be incorporated into the composition to facilitate processing, to enhance stability, or for other reasons. Examples of pharmaceutically acceptable bases include amino acids, amino acid esters, ammonium hydroxide, potassium hydroxide, sodium hydroxide, sodium hydrogen carbonate, aluminum hydroxide, calcium carbonate, magnesium hydroxide, magnesium aluminum silicate, synthetic aluminum silicate, synthetic hydrocalcite, magnesium aluminum hydroxide, diisopropylethylamine, ethanolamine, ethylenediamine, triethanolamine, triethylamine, triisopropanolamine, trimethylamine, tris(hydroxymethyl)aminomethane (TRIS) and the like. Also suitable are bases that are salts of a pharmaceutically acceptable acid, such as acetic acid, acrylic acid, adipic acid, alginic acid, alkanesulfonic acid, amino acids, ascorbic acid, benzoic acid, boric acid, butyric acid, carbonic acid, citric acid, fatty acids, formic acid, fumaric acid, gluconic acid, hydroquinosulfonic acid, isoascorbic acid, lactic acid, maleic acid, oxalic acid, para-bromophenylsulfonic acid, propionic acid, p-toluenesulfonic acid, salicylic acid, stearic acid, succinic acid, tannic acid, tartaric acid, thioglycolic acid, toluenesulfonic acid, uric acid, and the like. Salts of polyprotic acids, such as sodium phosphate, disodium hydrogen phosphate, and sodium dihydrogen phosphate can also be used. When the base is a salt, the cation can be any convenient and pharmaceutically acceptable cation, such as ammonium, alkali metals, alkaline earth metals, and the like. Example may include, but not limited to, sodium, potassium, lithium, magnesium, calcium and ammonium. [00339] Suitable acids are pharmaceutically acceptable organic or inorganic acids. Examples of suitable inorganic acids include hydrochloric acid, hydrobromic acid, hydriodic acid, sulfuric acid, nitric acid, boric acid, phosphoric acid, and the like. Examples of suitable organic acids include acetic acid, acrylic acid, adipic acid, alginic acid, alkanesulfonic acids, amino acids, ascorbic acid, benzoic acid, boric acid, butyric acid, carbonic acid, citric acid, fatty acids, formic acid, fumaric acid, gluconic acid, hydroquinosulfonic acid, isoascorbic acid, lactic acid, maleic acid, methanesulfonic acid, oxalic acid, para-bromophenylsulfonic acid, propionic acid, p- toluenesulfonic acid, salicylic acid, stearic acid, succinic acid, tannic acid, tartaric acid, thioglycolic acid, toluenesulfonic acid, uric acid and the like.
[00340] Pharmaceutical compositions for injection. In some embodiments, the invention provides a pharmaceutical composition for injection containing a compound of the present invention and a pharmaceutical excipient suitable for injection. Components and amounts of agents in the compositions are as described herein. The forms in which the novel compositions of the present invention may be incorporated for administration by injection include aqueous or oil suspensions, or emulsions, with sesame oil, corn oil, cottonseed oil, or peanut oil, as well as elixirs, mannitol, dextrose, or a sterile aqueous solution, and similar pharmaceutical vehicles.
[00341] Aqueous solutions in saline are also conventionally used for injection. Ethanol, glycerol, propylene glycol, liquid polyethylene glycol, and the like (and suitable mixtures thereof), cyclodextrin derivatives, and vegetable oils may also be employed. The proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, for the maintenance of the required particle size in the case of dispersion and by the use of surfactants. The prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like.
[00342] Sterile injectable solutions are prepared by incorporating the compound of the present invention in the required amount in the appropriate solvent with various other ingredients as enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, certain desirable methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
[00343] Pharmaceutical compositions for topical (e.g.. transdermal) delivery. In some embodiments, the invention provides a pharmaceutical composition for transdermal delivery containing a compound of the present invention and a pharmaceutical excipient suitable for transdermal delivery.
[00344] Compositions of the present invention can be formulated into preparations in solid, semi-solid, or liquid forms suitable for local or topical administration, such as gels, water soluble jellies, creams, lotions, suspensions, foams, powders, slurries, ointments, solutions, oils, pastes, suppositories, sprays, emulsions, saline solutions, dimethylsulfoxide (DMSO)-based solutions. In general, carriers with higher densities are capable of providing an area with a prolonged exposure to the active ingredients. In contrast, a solution formulation may provide more immediate exposure of the active ingredient to the chosen area.
[00345] The pharmaceutical compositions also may comprise suitable solid or gel phase carriers or excipients, which are compounds that allow increased penetration of, or assist in the delivery of, therapeutic molecules across the stratum corneum permeability barrier of the skin. There are many of these penetration-enhancing molecules known to those trained in the art of topical formulation. Examples of such carriers and excipients include, but are not limited to, humectants (e.g., urea), glycols (e.g., propylene glycol), alcohols (e.g., ethanol), fatty acids (e.g., oleic acid), surfactants (e.g., isopropyl myristate and sodium lauryl sulfate), pyrrolidones, glycerol monolaurate, sulfoxides, terpenes (e.g., menthol), amines, amides, alkanes, alkanols, water, calcium carbonate, calcium phosphate, various sugars, starches, cellulose derivatives, gelatin, and polymers such as polyethylene glycols.
[00346] Another exemplary formulation for use in the methods of the present invention employs transdermal delivery devices ("patches"). Such transdermal patches may be used to provide continuous or discontinuous infusion of a compound of the present invention in controlled amounts, either with or without another agent.
[00347] The construction and use of transdermal patches for the delivery of pharmaceutical agents is well known in the art. See, e.g., U.S. Pat. Nos. 5,023,252, 4,992,445 and 5,001,139. Such patches may be constructed for continuous, pulsatile, or on demand delivery of pharmaceutical agents.
[00348] Pharmaceutical compositions for inhalation. Compositions for inhalation or insufflation include solutions and suspensions in pharmaceutically acceptable, aqueous or organic solvents, or mixtures thereof, and powders. The liquid or solid compositions may contain suitable pharmaceutically acceptable excipients as described supra. Preferably the compositions are administered by the oral or nasal respiratory route for local or systemic effect. Compositions in preferably pharmaceutically acceptable solvents may be nebulized by use of inert gases. Nebulized solutions may be inhaled directly from the nebulizing device or the nebulizing device may be attached to a face mask tent, or intermittent positive pressure breathing machine. Solution, suspension, or powder compositions may be administered, preferably orally or nasally, from devices that deliver the formulation in an appropriate manner.
[00349] Other pharmaceutical compositions. Pharmaceutical compositions may also be prepared from compositions described herein and one or more pharmaceutically acceptable excipients suitable for sublingual, buccal, rectal, intraosseous, intraocular, intranasal, epidural, or intraspinal administration. Preparations for such pharmaceutical compositions are well-known in the art. See, e.g., See, e.g., Anderson, Philip O.; Knoben, James E.; Troutman, William G, eds., Handbook of Clinical Drug Data, Tenth Edition, McGraw-Hill, 2002; Pratt and Taylor, eds., Principles of Drug Action, Third Edition, Churchill Livingston, New York, 1990; Katzung, ed., Basic and Clinical Pharmacology, Ninth Edition, McGraw Hill, 20037ybg; Goodman and Gilman, eds., The Pharmacological Basis of Therapeutics, Tenth Edition, McGraw Hill, 2001; Remingtons Pharmaceutical Sciences, 20th Ed., Lippincott Williams & Wilkins., 2000; Martindale, The Extra Pharmacopoeia, Thirty-Second Edition (The Pharmaceutical Press, London, 1999); all of which are incorporated by reference herein in their entirety.
[00350] Administration of the compounds or pharmaceutical composition of the present invention can be effected by any method that enables delivery of the compounds to the site of action. These methods include oral routes, intraduodenal routes, parenteral injection (including intravenous, intraarterial, subcutaneous, intramuscular, intravascular, intraperitoneal or infusion), topical (e.g. transdermal application), rectal administration, via local delivery by catheter or stent or through inhalation. Compounds can also abe administered intraadiposally or intrathecally.
[00351] The amount of the compound administered will be dependent on the mammal being treated, the severity of the disorder or condition, the rate of administration, the disposition of the compound and the discretion of the prescribing physician. However, an effective dosage is in the range of about 0.001 to about 100 mg per kg body weight per day, preferably about 1 to about 35 mg/kg/day, in single or divided doses. For a 70 kg human, this would amount to about 0.05 to 7 g/day, preferably about 0.05 to about 2.5 g/day. In some instances, dosage levels below the lower limit of the aforesaid range may be more than adequate, while in other cases still larger doses may be employed without causing any harmful side effect, e.g. bydividing such larger doses into several small doses for administration throughout the day.
[00352] In some embodiments, a compound of the invention is administered in a single dose. Typically, such administration will be by injection, e.g., intravenous injection, in order to introduce the agent quickly. However, other routes may be used as appropriate. A single dose of a compound of the invention may also be used for treatment of an acute condition.
[00353] In some embodiments, a compound of the invention is administered in multiple doses. Dosing may be about once, twice, three times, four times, five times, six times, or more than six times per day. Dosing may be about once a month, once every two weeks, once a week, or once every other day. In another embodiment a compound of the invention and another agent are administered together about once per day to about 6 times per day. In another embodiment the administration of a compound of the invention and an agent continues for less than about 7 days. In yet another embodiment the administration continues for more than about 6, 10, 14, 28 days, two months, six months, or one year. In some cases, continuous dosing is achieved and maintained as long as necessary.
[00354] Administration of the agents of the invention may continue as long as necessary. In some embodiments, an agent of the invention is administered for more than 1, 2, 3, 4, 5, 6, 7, 14, or 28 days. In some embodiments, an agent of the invention is administered for less than 28, 14, 7, 6, 5, 4, 3, 2, or 1 day. In some embodiments, an agent of the invention is administered chronically on an ongoing basis, e.g., for the treatment of chronic effects.
[00355] An effective amount of a compound of the invention may be administered in either single or multiple doses by any of the accepted modes of administration of agents having similar utilities, including rectal, buccal, intranasal and transdermal routes, by intra-arterial injection, intravenously, intraperitoneally, parenterally, intramuscularly, subcutaneously, orally, topically, or as an inhalant.
[00356] The compositions of the invention may also be delivered via an impregnated or coated device such as a stent, for example, or an artery-inserted cylindrical polymer. Such a method of administration may, for example, aid in the prevention or amelioration of restenosis following procedures such as balloon angioplasty. Without being bound by theory, compounds of the invention may slow or inhibit the migration and proliferation of smooth muscle cells in the arterial wall which contribute to restenosis. A compound of the invention may be administered, for example, by local delivery from the struts of a stent, from a stent graft, from grafts, or from the cover or sheath of a stent. In some embodiments, a compound of the invention is admixed with a matrix. Such a matrix may be a polymeric matrix, and may serve to bond the compound to the stent. Polymeric matrices suitable for such use, include, for eample, lactone-based polyesters or copolyesters such as polylactide, polycaprolactonglycolide, polyorthoesters, polyanhydrides, polyaminoacids, polysaccharides, polyphosphazenes, poly (ether-ester) copolymers (e.g. PEO-PLLA); polydimethylsiloxane, poly(ethylene- vinylacetate), acrylate-based polymers or copolymers (e.g. polyhydroxyethyl methylmethacrylate, polyvinyl pyrrolidinone), fluorinated polymers such as polytetrafluoroethylene and cellulose esters. Suitable matrices may be nondegrading or may degrade with time, releasing the compound or compounds. Compounds of the invention may be applied to the surface of the stent by various methods such as dip/spin coating, spray coating, dip- coating, and/or brush-coating. The compounds may be applied in a solvent and the solvent may be allowed to evaporate, thus forming a layer of compound onto the stent. Alternatively, the compound may be located in the body of the stent or graft, for example in microchannels or micropores. When implanted, the compound diffuses out of the body of the stent to contact the arterial wall. Such stents may be prepared by dipping a stent manufactured to contain such micropores or microchannels into a solution of the compound of the invention in a suitable solvent, followed by evaporation of the solvent. Excess drug on the surface of the stent may be removed via an additional brief solvent wash. In yet other embodiments, compounds of the invention may be covalently linked to a stent or graft. A covalent linker may be used which degrades in vivo, leading to the release of the compound of the invention. Any bio- labile linkage may be used for such a purpose, such as ester, amide or anhydride linkages. Compounds of the invention may additionally be administered intravascularly from a balloon used during angioplasty. Extravascular administration of the compounds via the pericard or via advential application of formulations of the invention may also be performed to decrease restenosis.
[00357] A variety of stent devices which may be used as described are disclosed, for example, in the following references, all of which are hereby incorporated by reference: U.S. Pat. No. 5451233; U.S. Pat. No. 5040548; U.S. Pat. No. 5061273; U.S. Pat. No. 5496346; U.S. Pat. No. 5292331; U.S. Pat. No. 5674278; U.S. Pat. No. 3657744; U.S. Pat. No. 4739762; U.S. Pat. No. 5195984; U.S. Pat. No. 5292331; U.S. Pat. No. 5674278; U.S. Pat. No. 5879382; U.S. Pat. No. 6344053.
[00358] The compounds of the invention may be administered in dosages. It is known in the art that due to intersubject variability in compound pharmacokinetics, individualization of dosing regimen is necessary for optimal therapy. Dosing for a compound of the invention may be found by routine experimentation in light of the instant disclosure.
[00359] When a compound of the invention, is administered in a composition that comprises one or more agents, and the agent has a shorter half-life than the compound of the invention unit dose forms of the agent and the compound of the invention may be adjusted accordingly.
[00360] The subject pharmaceutical composition may, for example, be in a form suitable for oral administration as a tablet, capsule, pill, powder, sustained release formulations, solution, suspension, for parenteral injection as a sterile solution, suspension or emulsion, for topical administration as an ointment or cream or for rectal administration as a suppository. The pharmaceutical composition may be in unit dosage forms suitable for single administration of precise dosages. The pharmaceutical composition will include a conventional pharmaceutical carrier or excipient and a compound according to the invention as an active ingredient. In addition, it may include other medicinal or pharmaceutical agents, carriers, adjuvants, etc.
[00361] Exemplary parenteral administration forms include solutions or suspensions of active compound in sterile aqueous solutions, for example, aqueous propylene glycol or dextrose solutions. Such dosage forms can be suitably buffered, if desired.
[00362] The activity of the compounds of the present invention may be determined by the following procedure, as well as the procedure described in the examples below. The activity of the kinase is assessed by measuring the incorporation of γ-33P-ρhosphate from γ -33P-ATP onto N-terminal His tagged substrate, which is expressed in E. coli and is purified by conventional methods, in the presence of the kinase. The assay is carried out in 96- well polypropylene plate. The incubation mixture (100, μL) comprises of 25 mM Hepes, pH 7.4, 10 mM MgCl2, 5 mM β-glycerolphosphate, 100 μM Na-orthovanadate, 5 mM DTT, 5 nM kinase, and 1 μM substrate. Inhibitors are suspended in DMSO, and all reactions, including controls are performed at a final concentration of 1% DMSO. Reactions are initiated by the addition of 10 μM ATP (with 0.5 μCi γ-33P- ATP/well) and incubated at ambient temperature for 45 minutes. Equal volume of 25% TCA is added to stop the reaction and precipitate the proteins. Precipitated proteins are trapped onto glass fiber B filterplates, and excess labeled ATP washed off using a Tomtec MACH III harvestor. Plates are allowed to air-dry prior to adding 30 μL/well of Packard Microscint 20, and plates are counted using a Packard TopCount. [00363] The invention also provides kits. The kits include a compound or compounds of the present invention as described herein, in suitable packaging, and written material that can include instructions for use, discussion of clinical studies, listing of side effects, and the like. Such kits may also include information, such as scientific literature references, package insert materials, clinical trial results, and/or summaries of these and the like, which indicate or establish the activities and/or advantages of the composition, and/or which describe dosing, administration, side effects, drug interactions, or other information useful to the health care provider. Such information may be based on the results of various studies, for example, studies using experimental animals involving in vivo models and studies based on human clinical trials. The kit may further contain another agent. In some embodiments, the compound of the present invention and the agent are provided as separate compositions in separate containers within the kit. In some embodiments, the compound of the present invention and the agent are provided as a single composition within a container in the kit. Suitable packaging and additional articles for use (e.g., measuring cup for liquid preparations, foil wrapping to minimize exposure to air, and the like) are known in the art and may be included in the kit. Kits described herein can be provided, marketed and/or promoted to health providers, including physicians, nurses, pharmacists, formulary officials, and the like. Kits may also, in some embodiments, be marketed directly to the consumer.
[00364] The invention also provides methods of using the compounds or pharmaceutical compositions of the present invention to treat disease conditions, including but not limited to diseases associated with malfunctioning of one or more types of PD kinase. A detailed description of conditions and disorders mediated by pi lOδ kinase activity is set forth in Sadu et al., WO 01/81346, which is incorporated herein by reference in its entirety for all purposes.
[00365] The treatment methods provided herein comprise administering to the subject a therapeutically effective amount of a compound of the invention. In one embodiment, the present invention provides a method of treating an inflammation disorder, including autoimmune diseases in a mammal. The method comprises administering to said mammal a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt, ester, prodrug, solvate, hydrate or derivative thereof. Examples of autoimmune diseases includes but is not limited to acute disseminated encephalomyelitis (ADEM), Addison's disease, antiphospholipid antibody syndrome (APS), aplastic anemia, autoimmune hepatitis, coeliac disease, Crohn's disease, Diabetes mellitus (type 1), Goodpasture's syndrome, Graves' disease, Guillain-Barrέ syndrome (GBS), Hashimoto's disease, lupus erythematosus, multiple sclerosis, myasthenia gravis, opsoclonus myoclonus syndrome (OMS), optic neuritis, Ord's thyroiditis, oemphigus, polyarthritis, primary biliary cirrhosis, psoriasis, rheumatoid arthritis, Reiter's syndrome, Takayasu's arteritis, temporal arteritis (also known as "giant cell arteritis"), warm autoimmune hemolytic anemia, Wegener's granulomatosis, alopecia universalis, Chagas' disease, chronic fatigue syndrome, dysautonomia, endometriosis, hidradenitis suppurativa, interstitial cystitis, neuromyotonia, sarcoidosis, scleroderma, ulcerative colitis, vitiligo, and vulvodynia. Other disorders include bone-resorption disorders and thromobsis.
[00366] In some embodiments, the method of treating inflammatory or autoimmune diseases comprises administering to a subject (e.g. a mammal) a therapeutically effective amount of one or more compounds of the present invention that selectively inhibit PDK-δ and/or PI3K-γ as compared to all other type I PI3 kinases. Such selective inhibition of PI3K-δ and/or PI3K-γ may be advantageous for treating any of the diseases or conditions described herein. For example, selective inhibition of PI3K.-5 may inhibit inflammatory responses associated with inflammatory diseases, autoimmune disease, or diseases related to an undesirable immune response including but not limited to asthma, emphysema, allergy, dermatitis, rhuematoid arthritis, psoriasis, lupus erythematosus, or graft versus host disease. Selective inhibition of PI3K-6 may further provide for a reduction in the inflammatory or undesirable immune response without a concomittant reduction in the ability to reduce a bacterial, viral, and/or fungal infection. Selective inhibition of both POK-δ and PI3K-γ may be advantageous for inhibiting the inflammatory response in the subject to a greater degree than that would be provided for by inhibitors that selectively inhibit PI3K -δ or PI3K-γ alone. In one aspect, one or more of the subject methods are effective in reducing antigen specific antibody production in vivo by about 2-fold, 3-fold, 4-fold, 5-fold, 7.5- fold, 10-fold, 25-fold, 50-fold, 100-fold, 250-fold, 500-fold, 750-fold, or about 1000-fold or more. In another aspect, one or more of the subject methods are effective in reducing antigen specific IgG3 and/or IgGM production in vivo by about 2-fold, 3-fold, 4-fold, 5-fold, 7.5-fold, 10-fold, 25-fold, 50-fold, 100-fold, 250-fold, 500-fold, 750-fold, or about 1000-fold or more.
[00367] In one aspect, one of more of the subject methods are effective in ameliorating symptoms assoicated with rhuematoid arthritis including but not limited to a reduction in the swelling of joints, a reduction in serum anti- collagen levels, and/or a reduction in joint pathology such as bone resorption, cartilage damage, pannus, and/or inflammation. In another aspect, the subject methods are effective in reducing ankle inflammation by at least about 2%, 5%, 10%, 15%, 20%, 25%, 30%, 50%, 60%, or about 75% to 90%. In another aspect, the subject methods are effective in reducing knee inflammation by at least about 2%, 5%, 10%, 15%, 20%, 25%, 30%, 50%, 60%, or about 75% to 90% or more. In still another aspect, the subject methods are effective in reducing serum anti-type II collagen levels by at least about 10%, 12%, 15%, 20%, 24%, 25%, 30%, 35%, 50%, 60%, 75%, 80%, 86%, 87%, or about 90% or more. In another aspect, the subject methods are effective in reducing ankle histopathology scores by about 5%, 10%, 15%, 20%, 25%, 30%, 40%, 50%, 60%, 75%, 80%, 90% or more. In still another aspect, the subject methods are effective in reducing knee histopathology scores by about 5%, 10%, 15%, 20%, 25%, 30%, 40%, 50%, 60%, 75%, 80%, 90% or more.
[00368] In other embodiments, the present invention provides methods of using the compounds or pharmaceutical compositions to treat respiratory diseases including but not limited to diseases affecting the lobes of lung, pleural cavity, bronchial tubes, trachea, upper respiratory tract, or the nerves and muscle for breathing. For example, methods are provided to treat obstructive pulmonary disease. Chronic obstructive pulmonary disease (COPD) is an umbrella term for a group of respiratory tract diseases that are characterized by airflow obstruction or limitation. Conditions included in this umbrella term are: chronic bronchitis, emphysema, and bronchiectasis.
[00369] In another embodiment, the compounds described herein are used for the treatment of asthma. Also, the compounds or pharmaceutical compositions described herein may be used for the treatment of endotoxemia and sepsis. In one embodiment, the compounds or pharmaceutical compositions described herein are used to for the treatment of rheumatoid arthritis (RA). In yet another embodiment, the compounds or pharmaceutical compositions described herein is used for the treatment of contact or atopic dermatitis. Contact dermatitis includes irritant dermatitis, phototoxic dermatitis, allergic dermatitis, photoallergic dermatitis, contact urticaria, systemic contact-type dermatitis and the like. Irritant dermatitis can occur when too much of a substance is used on the skin of when the skin is sensitive to certain substance. Atopic dermatitis, sometimes called eczema, is a kind of dermatitis, an atopic skin disease.
[00370] The invention also relates to a method of treating a hyperproliferative disorder in a mammal that comprises administering to said mammal a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt, ester, prodrug, solvate, hydrate or derivative thereof. In some embodiments, said method relates to the treatment of cancer such as acute myeloid leukemia, thymus, brain, lung, squamous cell, skin, eye, retinoblastoma, intraocular melanoma, oral cavity and oropharyngeal, bladder, gastric, stomach, pancreatic, bladder, breast, cervical, head, neck, renal, kidney, liver, ovarian, prostate, colorectal, esophageal, testicular, gynecological, thyroid, CNS, PNS, AIDS-related (e.g. Lymphoma and Kaposi's Sarcoma) or viral- induced cancer. In some embodiments, said method relates to the treatment of a non-cancerous hyperproliferative disorder such as benign hyperplasia of the skin (e. g., psoriasis), restenosis, or prostate (e. g., benign prostatic hypertrophy (BPH)).
[00371] The invention also relates to a method of treating diseases related to vasculogenesis or angiogenesis in a mammal that comprises administering to said mammal a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt, ester, prodrug, solvate, hydrate or derivative thereof. In some embodiments, said method is for treating a disease selected from the group consisting of tumor angiogenesis, chronic inflammatory disease such as rheumatoid arthritis, atherosclerosis, inflammatory bowel disease, skin diseases such as psoriasis, eczema, and scleroderma, diabetes, diabetic retinopathy, retinopathy of prematurity, age-related macular degeneration, hemangioma, glioma, melanoma, Kaposi's sarcoma and ovarian, breast, lung, pancreatic, prostate, colon and epidermoid cancer.
[00372] Patients that can be treated with compounds of the present invention, or pharmaceutically acceptable salt, ester, prodrug, solvate, hydrate or derivative of said compounds, according to the methods of this invention include, for example, patients that have been diagnosed as having psoriasis; restenosis; atherosclerosis; BPH; breast cancer such as a ductal carcinoma in duct tissue in a mammary gland, medullary carcinomas, colloid carcinomas, tubular carcinomas, and inflammatory breast cancer; ovarian cancer, including epithelial ovarian tumors such as adenocarcinoma in the ovary and an adenocarcinoma that has migrated from the ovary into the abdominal cavity; uterine cancer; cervical cancer such as adenocarcinoma in the cervix epithelial including squamous cell carcinoma and adenocarcinomas; prostate cancer, such as a prostate cancer selected from the following: an adenocarcinoma or an adenocarinoma that has migrated to the bone; pancreatic cancer such as epitheliod carcinoma in the pancreatic duct tissue and an adenocarcinoma in a pancreatic duct; bladder cancer such as a transitional cell carcinoma in urinary bladder, urothelial carcinomas (transitional cell carcinomas), tumors in the urothelial cells that line the bladder, squamous cell carcinomas, adenocarcinomas, and small cell cancers; leukemia such as acute myeloid leukemia (AML), acute lymphocytic leukemia, chronic lymphocytic leukemia, chrome myeloid leukemia, hairy cell leukemia, myelodysplasia, myeloproliferative disorders, acute myelogenous leukemia (AML), chronic myelogenous leukemia (CML), mastocytosis, chronic lymphocytic leukemia (CLL), multiple myeloma (MM), and myelodysplastic syndrome (MDS); bone cancer; lung cancer such as non-small cell lung cancer (NSCLC), which is divided into squamous cell carcinomas, adenocarcinomas, and large cell undifferentiated carcinomas, and small cell lung cancer; skin cancer such as basal cell carcinoma, melanoma, squamous cell carcinoma and actinic keratosis, which is a skin condition that sometimes develops into squamous cell carcinoma; eye retinoblastoma; cutaneous or intraocular (eye) melanoma; primary liver cancer (cancer that begins in the liver); kidney cancer; thyroid cancer such as papillary, follicular, medullary and anaplastic; AIDS-related lymphoma such as diffuse large B-cell lymphoma, B-cell immunoblastic lymphoma and small non-cleaved cell lymphoma; Kaposi's Sarcoma; viral-induced cancers including hepatitis B virus (HBV), hepatitis C virus (HCV), and hepatocellular carcinoma; human lymphotropic virus- type 1 (HTLV-I) and adult T-cell leukemia/lymphoma; and human papilloma virus (HPV) and cervical cancer; central nervous system cancers (CNS) such as primary brain tumor, which includes gliomas (astrocytoma, anaplastic astrocytoma, or glioblastoma multiforme), Oligodendroglioma, Ependymoma, Meningioma, Lymphoma, Schwannoma, and Medulloblastoma; peripheral nervous system (PNS) cancers such as acoustic neuromas and malignant peripheral nerve sheath tumor (MPNST) including neurofibromas and schwannomas, malignant fibrous cytoma, malignant fibrous histiocytoma, malignant meningioma, malignant mesothelioma, and malignant mixed Mϋllerian tumor; oral cavity and oropharyngeal cancer such as, hypopharyngeal cancer, laryngeal cancer, nasopharyngeal cancer, and oropharyngeal cancer; stomach cancer such as lymphomas, gastric stromal tumors, and carcinoid tumors; testicular cancer such as germ cell tumors (GCTs), which include seminomas and nonseminomas, and gonadal stromal tumors, which include Leydig cell tumors and Sertoli cell tumors; thymus cancer such as to thymomas, thymic carcinomas, Hodgkin disease, non- Hodgkin lymphomas carcinoids or carcinoid tumors; rectal cancer; and colon cancer.
[00373] The invention also relates to a method of treating diabetes in a mammal that comprises administering to said mammal a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt, ester, prodrug, solvate, hydrate or derivative thereof.
[00374] In addition, the compounds described herein may be used to treat acne.
[00375] In addition, the compounds described herein may be used for the treatment of arteriosclerosis, including atherosclerosis. Arteriosclerosis is a general term describing any hardening of medium or large arteries. Atherosclerosis is a hardening of an artery specifically due to an atheromatous plaque.
[00376] Further the compounds described herein may be used for the treatment of glomerulonephritis.
Glomerulonephritis is a primary or secondary autoimmune renal disease characterized by inflammation of the glomeruli. It may be asymptomatic, or present with hematuria and/or proteinuria. There are many recognized types, divided in acute, subacute or chronic glomerulonephritis. Causes are infectious (bacterial, viral or parasitic pathogens), autoimmune or paraneoplastic.
[00377] Additionally, the compounds described herein may be used for the treatment of bursitis, lupus, acute disseminated encephalomyelitis (ADEM), addison's disease, antiphospholipid antibody syndrome (APS), aplastic anemia, autoimmune hepatitis, coeliac disease, Crohn's disease, diabetes mellitus (type 1), goodpasture's syndrome, graves' disease, guillain-barrέ syndrome (GBS), hashimoto's disease, inflammatory bowel disease, lupus erythematosus, myasthenia gravis, opsoclonus myoclonus syndrome (OMS), optic neuritis, ord's thyroiditis,ostheoarthritis, uveoretinitis, pemphigus, polyarthritis, primary biliary cirrhosis, reiter's syndrome, takayasu's arteritis, temporal arteritis, warm autoimmune hemolytic anemia, Wegener's granulomatosis, alopecia universalis, chagas' disease, chronic fatigue syndrome, dysautonomia, endometriosis, hidradenitis suppurativa, interstitial cystitis, neuromyotonia, sarcoidosis, scleroderma, ulcerative colitis, vitiligo, vulvodynia, appendicitis, arteritis, arthritis, blepharitis, bronchiolitis, bronchitis, cervicitis, cholangitis, cholecystitis, chorioamnionitis, colitis, conjunctivitis, cystitis, dacryoadenitis, deπnatomyositis, endocarditis, endometritis, enteritis, enterocolitis, epicondylitis, epididymitis, fasciitis, fibrositis, gastritis, gastroenteritis, gingivitis, hepatitis, hidradenitis, ileitis, iritis, laryngitis, mastitis, meningitis, myelitis, myocarditis, myositis, nephritis, omphalitis, oophoritis, orchitis, osteitis, otitis, pancreatitis, parotitis, pericarditis, peritonitis, pharyngitis, pleuritis, phlebitis, pneumonitis, proctitis, prostatitis, pyelonephritis, rhinitis, salpingitis, sinusitis, stomatitis, synovitis, tendonitis, tonsillitis, uveitis, vaginitis, vasculitis, or vulvitis.
[00378] The invention also relates to a method of treating a cardiovascular disease in a mammal that comprises administering to said mammal a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt, ester, prodrug, solvate, hydrate or derivative thereof. Examples of cardiovascular conditions include, but are not limited to, atherosclerosis, restenosis, vascular occlusion and carotid obstructive disease.
[00379] In another aspect, the present invention provides methods of disrupting the function of a leukocyte or disrupting a function of an osteoclast. The method includes contacting the leukocyte or the osteoclast with a function disrupting amount of a compound of the invention.
[00380] In another aspect of the present invention, methods are provided for treating ophthalmic disease by administering one or more of the subject compounds or pharmaceutical compositions to the eye of a subject.
[00381] Methods are further provided for administering the compounds of the present invention via eye drop, intraocular injection, intravitreal injection, topically, or through the use of a drug eluting device, microcapsule, implant, or microfluidic device. In some cases, the compounds of the present invention are administered with a carrier or excipient that increases the intraocular penetrance of the compound such as an oil and water emulsion with colloid particles having an oily core surrounded by an interfacial film.
[00382] In some cases, the colloid particles include at least one cationic agent and at least one non-ionic sufactant such as a poloxamer, tyloxapol, a polysorbate, a polyoxyethylene castor oil derivative, a sorbitan ester, or a polyoxyl stearate. In some cases, the cationic agent is an alkylamine, a tertiary alkyl amine, a quarternary ammonium compound, a cationic lipid, an amino alcohol, a biguanidine salt, a cationic compound or a mixture thereof. In some cases the cationic agent is a biguanidine salt such as chlorhexidine, polyaminopropyl biguanidine, phenformin, alkylbiguanidine, or a mixture thereof. In some cases, the quaternary ammonium compound is a benzalkonium halide, lauralkonium halide, cetrimide, hexadecyltrimethylammonium halide, tetradecyltrimethylammonium halide, dodecyltrimethylammonium halide, cetrimonium halide, benzethonium halide, behenalkonium halide, cetalkonium halide, cetethyldimonium halide, cetylpyridinium halide, benzododecinium halide, chlorallyl methenamine halide, rnyristylalkonium halide, stearalkonium halide or a mixture of two or more thereof. In some cases, cationic agent is a benzalkonium chloride, lauralkonium chloride, benzododecinium bromide, benzethenium chloride, hexadecyltrimethylammonium bromide, tetradecyltrimethylammonium bromide, dodecyltrimethylammonium bromide or a mixture of two or more thereof. In some cases, the oil phase is mineral oil and light mineral oil, medium chain triglycerides (MCT), coconut oil; hydrogenated oils comprising hydrogenated cottonseed oil, hydrogenated palm oil, hydrogenate castor oil or hydrogenated soybean oil; polyoxyethylene hydrogenated castor oil derivatives comprising poluoxyl-40 hydrogenated castor oil, polyoxyl-60 hydrogenated castor oil or polyoxyl- 100 hydrogenated castor oil.
[00383] The invention further provides methods of modulating kinase activity by contacting a kinase with an amount of a compound of the invention sufficient to modulate the activity of the kinase. Modulate can be inhibiting or activating kinase activity. In some embodiments, the invention provides methods of inhibiting kinase activity by contacting a kinase with an amount of a compound of the invention sufficient to inhibit the activity of the kinase. In some embodiments, the invention provides methods of inhibiting kinase activity in a solution by contacting said solution with an amount of a compound of the invention sufficient to inhibit the activity of the kinase in said solution. In some embodiments, the invention provides methods of inhibiting kinase activity in a cell by contacting said cell with an amount of a compound of the invention sufficient to inhibit the activity of the kinase in said cell. In some embodiments, the invention provides methods of inhibiting kinase activity in a tissue by contacting said tissue with an amount of a compound of the invention sufficient to inhibit the activity of the kinase in said tissue. In some embodiments, the invention provides methods of inhibiting kinase activity in an organism by contacting said organism with an amount of a compound of the invention sufficient to inhibit the activity of the kinase in said organism. In some embodiments, the invention provides methods of inhibiting kinase activity in an animal by contacting said animal with an amount of a compound of the invention sufficient to inhibit the activity of the kinase in said animal. In some embodiments, the invention provides methods of inhibiting kinase activity in a mammal by contacting said mammal with an amount of a compound of the invention sufficient to inhibit the activity of the kinase in said mammal. In some embodiments, the invention provides methods of inhibiting kinase activity in a human by contacting said human with an amount of a compound of the invention sufficient to inhibit the activity of the kinase in said human. In some embodiments, the % of kinase activity after contacting a kinase with a compound of the invention is less than 1, 5, 10, 20, 30, 40, 50, 60, 70, 80 90, 95, or 99% of the kinase activity in the absence of said contacting step.
[00384] In some embodiments, the kinase is a lipid kinase or a protein kinase. In some embodiments, the kinase is selected from the group consisting of PI3 kinase including different isorforms such as PI3 kinase α, PI3 kinase β, PI3 kinase γ, PI3 kinase δ; DNA-PK; mTor; AbI, VEGFR, Ephrin receptor B4 (EphB4); TEK receptor tyrosine kinase (TIE2); FMS-related tyrosine kinase 3 (FLT-3); Platelet derived growth factor receptor (PDGFR); RET; ATM; ATR; hSmg-1; Hck; Src; Epidermal growth factor receptor (EGFR); KIT; Inulsin Receptor (IR) and IGFR.
[00385] The invention further provides methods of modulating PI3 kinase activity by contacting a PI3 kinase with an amount of a compound of the invention sufficient to modulate the activity of the PI3 kinase. Modulate can be inhibiting or activating PI3 kinase activity. In some embodiments, the invention provides methods of inhibiting PI3 kinase activity by contacting a PI3 kinase with an amount of a compound of the invention sufficient to inhibit the activity of the PI3 kinase. In some embodiments, the invention provides methods of inhibiting PI3 kinase activity. Such inhibition can take place in solution, in a cell expressing one or more PI3 kinases, in a tissue comprising a cell expressing one or more PI3 kinases, or in an organism expressing one or more PI3 kinases. In some embodiments, the invention provides methods of inhibiting PI3 kinase activity in an animal (including mammal such as humans) by contacting said animal with an amount of a compound of the invention sufficient to inhibit the activity of the PI3 kinase in said animal.
[00386] The present invention also provides methods for combination therapies in which an agent known to modulate other pathways, or other components of the same pathway, or even overlapping sets of target enzymes are used in combination with a compound of the present invention, or a pharmaceutically acceptable salt, ester, prodrug, solvate, hydrate or derivative thereof. In one aspect, such therapy includes but is not limited to the combination of the subject compound with chemotherapeutic agents, therapeutic antibodies, and radiation treatment, to provide a synergistic or additive therapeutic effect.
[00387] In one aspect, the compounds or pharmaceutical compositions of the present invention may present synergistic or additive efficacy when administered in combination with agents that inhibit IgE production or activity. Such combination can reduce the undesired effect of high level of IgE associated with the use of one or more PI3Kδ inhibitors, if such effect occurs. This may be particularly useful in treatment of autoimmune and inflammatory disorders (AIID) such as rheumatoid arthritis. Additionally, the administration of PDKδ or PI3Kδ/γ inhibitors of the present invention in combination with inhibitors of mTOR may also exhibit synergy through enhanced inhibition of the PDK pathway.
[00388] In a separate but related aspect, the present invention provides a combination treatment of a disease associated with PDKδ comprising administering to a PDKδ inhibitor and an agent that inhibits IgE production or activity. Other exemplary PDKδ inhibitors are applicable and they are described, e.g., US Patent No. 6,800,620. Such combination treatment is particularly useful for treating autoimmune and inflammatory diseases (AIID) including but not limited to rheumatoid arthritis.
[00389] Agents that inhibit IgE production are known in the art and they include but are not limited to one or more of TEI-9874, 2-(4-(6-cyclohexyloxy-2-naphtyloxy)phenylacetamide)benzoic acid, rapamycin, rapamycin analogs (i.e. rapalogs), TORCl inhibitors, TORC2 inhibitors, and any other compounds that inhibit mTORCl and mT0RC2. Agents that inhibit IgE activity include, for example, anti-IgE antibodies such as for example Omalizumab and TNX-901.
[00390] For treatment of autoimmune diseases, the subject compounds or pharmaceutical compositions can be used in combination with commonly prescribed drugs including but not limited to Enbrel®, Remicade®, Humira®, Avonex®, and Rebif®. For treatment of respiratory diseaseses, the subject compounds or pharmaceutical compositions can be administered in combination with commonly prescribed drugs including but not limited to Xolair®, Advair®, Singulair®, and Spiriva®.
[00391] The compounds of the invention may be formulated or administered in conjunction with other agents that act to relieve the symptoms of inflammatory conditions such as encephalomyelitis, asthma, and the other diseases described herein. These agents include non-steroidal anti-inflammatory drugs (NSAIDs), e.g. acetylsalicylic acid; ibuprofen; naproxen; indomethacin; nabumetone; tolmetin; etc. Corticosteroids are used to reduce inflammation and suppress activity of the immune system. The most commonly prescribed drug of this type is Prednisone. Chloroquine (Aralen) or hydroxychloroquine (Plaquenil) may also be very useful in some individuals with lupus. They are most often prescribed for skin and joint symptoms of lupus. Azathioprine (Imuran) and cyclophosphamide (Cytoxan) suppress inflammation and tend to suppress the immune system. Other agents, e.g. methotrexate and cyclosporin are used to control the symptoms of lupus. Anticoagulants are employed to prevent blood from clotting rapidly. They range from aspirin at very low dose which prevents platelets from sticking, to heparin/coumadin.
[00392] In another one aspect, this invention also relates to a pharmaceutical composition for inhibiting abnormal cell growth in a mammal which comprises an amount of a compound of the present invention, or a pharmaceutically acceptable salt, ester, prodrug, solvate, hydrate or derivative thereof, in combination with an amount of an anticancer agent (e.g. a chemotherapeutic agent). Many chemotherapeutics are presently known in the art and can be used in combination with the compounds of the invention.
[00393] In some embodiments, the chemotherapeutic is selected from the group consisting of mitotic inhibitors, alkylating agents, anti-metabolites, intercalating antibiotics, growth factor inhibitors, cell cycle inhibitors, enzymes, topoisomerase inhibitors, biological response modifiers, anti-hormones, angiogenesis inhibitors, and anti-androgens. Non-limiting examples are chemotherapeutic agents, cytotoxic agents, and non-peptide small molecules such as Gleevec (Imatinib Mesylate), Velcade (bortezomib), Casodex (bicalutamide), Iressa (gefitinib), and Adriamycin as well as a host of chemotherapeutic agents. Non-limiting examples of chemotherapeutic agents include alkylating agents such as thiotepa and cyclosphosphamide (CYTOXAN™); alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, trietylenephosphoramide, triethylenethiophosphaoramide and trimethylolomelamine; nitrogen mustards such as chlorambucil, chlornaphazine, cholophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard; nitrosureas such as carmustine, chlorozotocin, fotemustine, lomustine, nimustine, ranimustine; antibiotics such as aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, calicheamicin, carabicin, carminomycin, carzinophilin, Casodex™ , chromomycins, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, doxorubicin, epirubicin, esorubicin, idarubicin, marcellomycin, mitomycins, mycophenolic acid, nogalamycin, olivomycins, peplomycin, potfiromycin, puromycin, quelamycin, rodorubicin, streptonigrin, streptozocin, tubercidin, ubenimex, zinostatin, zorubicin; anti-metabolites such as methotrexate and 5-fluorouracil (5-FU); folic acid analogues such as denopterin, methotrexate, pteropterin, trimetrexate; purine analogs such as fludarabine, 6-mercaptopurine, thiamiprine, thioguanine; pyrimidine analogs such as ancitabine, azacitidine, 6-azauridine, carmofur, cytarabine, dideoxyuridine, doxifluridine, enocitabine, floxuridine, androgens such as calusterone, dromostanolone propionate, epitiostanol, mepitiostane, testolactone; anti-adrenals such as aminoglutethimide, mitotane, trilostane; folic acid replenisher such as frolmic acid; aceglatone; aldophosphamide glycoside; aminolevulinic acid; amsacrine; bestrabucil; bisantrene; edatraxate; defofamine; demecolcine; diaziquone; elfomithine; elliptinium acetate; etoglucid; gallium nitrate; hydroxyurea; lentinan; lonidamine; mitoguazone; mitoxantrone; mopidamol; nitracrine; pentostatin; phenamet; pirarubicin; podophyllinic acid; 2-ethylhydrazide; procarbazine; PSK.R™ ; razoxane; sizofiran; spirogermanium; tenuazonic acid; triaziquone; 2,2',2"-trichlorotriethyla- mine; urethan; vindesine; dacarbazine; mannomustine; mitobronitol; mitolactol; pipobroman; gacytosine; arabinoside ("Ara-C"); cyclophosphamide; thiotepa; taxanes, e.g. paclitaxel (TAXOL™, Bristol-Myers Squibb Oncology, Princeton, NJ.) and docetaxel (TAXOTERE™, Rhone-Poulenc Rorer, Antony, France); retinoic acid; esperamicins; capecitabine; and pharmaceutically acceptable salts, acids or derivatives of any of the above. Also included as suitable chemotherapeutic cell conditioners are anti-hormonal agents that act to regulate or inhibit hormone action on tumors such as anti-estrogens including for example tamoxifen (Nolvadex™), raloxifene, aromatase inhibiting 4(5)-imidazoles, 4-hydroxytamoxifen, trioxifene, keoxifene, LY 117018, onapristone, and toremifene (Fareston); and anti-androgens such as flutamide, nilutamide, bicalutamide, leuprolide, and goserelin; chlorambucil; gemcitabine; 6-thioguanine; mercaptopurine; methotrexate; platinum analogs such as cisplatin and carboplatin; vinblastine; platinum; etoposide (VP- 16); ifosfamide; mitomycin C; mitoxantrone; vincristine; vinorelbine; navelbine; novantrone; teniposide; daunomycin; aminopterin; xeloda; ibandronate; camptothecin-11 (CPT-11); topoisomerase inhibitor RFS 2000; difluoromethylornithine (DMFO). Where desired, the compounds or pharmaceutical composition of the present invention can be used in combination with commonly prescribed anti-cancer drugs such as Herceptin®, Avastin®, Erbitux®, Rituxan®, Taxol , Arimidex®, Taxotere®, and Velcade . This invention further relates to a method for using the compounds or pharmaceutical composition in combination with radiation therapy in inhibiting abnormal cell growth or treating the hyperproliferative disorder in the mammal. Techniques for administering radiation therapy are known in the art, and these techniques can be used in the combination therapy described herein. The administration of the compound of the invention in this combination therapy can be determined as described herein.
[003951 Radiation therapy can be administered through one of several methods, or a combination of methods, including without limitation external-beam therapy, internal radiation therapy, implant radiation, stereotactic radiosurgery, systemic radiation therapy, radiotherapy and permanent or temporary interstitial brachytherapy. The term "brachytherapy," as used herein, refers to radiation therapy delivered by a spatially confined radioactive material inserted into the body at or near a tumor or other proliferative tissue disease site. The term is intended without limitation to include exposure to radioactive isotopes (e.g. At-211, 1-131, 1-125, Y-90, Re-186, Re-188, Sm-153, Bi-212, P-32, and radioactive isotopes of Lu). Suitable radiation sources for use as a cell conditioner of the present invention include both solids and liquids. By way of non-limiting example, the radiation source can be a radionuclide, such as 1-125, 1-131, Yb-169, Ir-192 as a solid source, 1-125 as a solid source, or other radionuclides that emit photons, beta particles, gamma radiation, or other therapeutic rays. The radioactive material can also be a fluid made from any solution of radionuclide(s), e.g., a solution of 1-125 or 1-131, or a radioactive fluid can be produced using a slurry of a suitable fluid containing small particles of solid radionuclides, such as Au- 198, Y-90. Moreover, the radionuclide(s) can be embodied in a gel or radioactive micro spheres.
[00396] Without being limited by any theory, the compounds of the present invention can render abnormal cells more sensitive to treatment with radiation for purposes of killing and/or inhibiting the growth of such cells. Accordingly, this invention further relates to a method for sensitizing abnormal cells in a mammal to treatment with radiation which comprises administering to the mammal an amount of a compound of the present invention or pharmaceutically acceptable salt, ester, prodrug, solvate, hydrate or derivative thereof, which amount is effective is sensitizing abnormal cells to treatment with radiation. The amount of the compound, salt, or solvate in this method can be determined according to the means for ascertaining effective amounts of such compounds described herein.
[00397] The compounds or pharmaceutical compositions of the present invention can be used in combination with an amount of one or more substances selected from anti-angiogenesis agents, signal transduction inhibitors, and antiproliferative agents.
[00398] Anti-angiogenesis agents, such as MMP-2 (matrix-metalloprotienase 2) inhibitors, MMP-9 (matrix- metalloprotienase 9) inhibitors, and COX-11 (cyclooxygenase 11) inhibitors, can be used in conjunction with a compound of the present invention and pharmaceutical compositions described herein. Examples of useful COX-II inhibitors include CELEBREX™ (alecoxib), valdecoxib, and rofecoxib. Examples of useful matrix metalloproteinase inhibitors are described in WO 96/33172 (published October 24,1996), WO 96/27583 (published March 7,1996), European Patent Application No. 97304971.1 (filed July 8,1997), European Patent Application No. 99308617.2 (filed October 29, 1999), WO 98/07697 (published February 26,1998), WO 98/03516 (published January 29,1998), WO 98/34918 (published August 13,1998), WO 98/34915 (published August 13,1998), WO 98/33768 (published August 6,1998), WO 98/30566 (published July 16, 1998), European Patent Publication 606,046 (published July 13,1994), European Patent Publication 931, 788 (published July 28,1999), WO 90/05719 (published May 31,1990), WO 99/52910 (published October 21,1999), WO 99/52889 (published October 21, 1999), WO 99/29667 (published June 17,1999), PCT International Application No. PCT/TB98/01113 (filed July 21,1998), European Patent Application No. 99302232.1 (filed March 25,1999), Great Britain Patent Application No. 9912961.1 (filed June 3, 1999), United States Provisional Application No. 60/148,464 (filed August 12,1999), United States Patent 5,863, 949 (issued January 26,1999), United States Patent 5,861, 510 (issued January 19,1999), and European Patent Publication 780,386 (published June 25, 1997), all of which are incorporated herein in their entireties by reference. Preferred MMP-2 and MMP-9 inhibitors are those that have little or no activity inhibiting MMP-I. More preferred, are those that selectively inhibit MMP-2 and/or AMP-9 relative to the other matrix-metalloproteinases (i. e., MAP-I, MMP-3, MMP-4, MMP-5, MMP-6, MMP- 7, MMP-8, MMP-IO, MMP-U, MMP-12, andMMP-13). Some specific examples of MMP inhibitors useful in the present invention are AG-3340, RO 32-3555, and RS 13-0830.
[00399] The invention also relates to a method of and to a pharmaceutical composition of treating a cardiovascular disease in a mammal which comprises an amount of a compound of the present invention, or a pharmaceutically acceptable salt, ester, prodrug, solvate, hydrate or derivative thereof, or an isotopically-labeled derivative thereof, and an amount of one or more therapeutic agents use for the treatment of cardiovascular diseases.
[00400] Examples for use in cardiovascular disease applications are anti-thrombotic agents, e.g., prostacyclin and salicylates, thrombolytic agents, e.g., streptokinase, urokinase, tissue plasminogen activator (TPA) and anisoylated plasminogen-streptokinase activator complex (APSAC), anti-platelets agents, e.g., acetyl-salicylic acid (ASA) and clopidrogel, vasodilating agents, e.g., nitrates, calcium channel blocking drugs, antiproliferative agents, e.g., colchicine and alkylating agents, intercalating agents, growth modulating factors such as interleukins, transformation growth factor-beta and congeners of platelet derived growth factor, monoclonal antibodies directed against growth factors, anti-inflammatory agents, both steroidal and non-steroidal, and other agents that can modulate vessel tone, function, arteriosclerosis, and the healing response to vessel or organ injury post intervention. Antibiotics can also be included in combinations or coatings comprised by the invention. Moreover, a coating can be used to effect therapeutic delivery focally within the vessel wall. By incorporation of the active agent in a swellable polymer, the active agent will be released upon swelling of the polymer.
[00401] The compounds describe herein may be formulated or administered in conjunction with liquid or solid tissue barriers also known as lubricants. Examples of tissue barriers include, but are not limited to, polysaccharides, polyglycans, seprafilm, interceed and hyaluronic acid.
[00402] Medicaments which may be administered in conjunction with the compounds described herein include any suitable drugs usefully delivered by inhalation for example, analgesics, e.g. codeine, dihydromorphine, ergotamine, fentanyl or morphine; anginal preparations, e.g. diltiazem; antiallergics, e.g. cromoglycate, ketotifen or nedocromil; anti-infectives, e.g. cephalosporins, penicillins, streptomycin, sulphonamides, tetracyclines or pentamidine; antihistamines, e.g. methapyrilene; anti-inflammatories, e.g. beclomethasone, flunisolide, budesonide, tipredane, triamcinolone acetonide or fluticasone; antitussives, e.g. noscapine; bronchodilators, e.g. ephedrine, adrenaline, fenoterol, formoterol, isoprenaline, metaproterenol, phenylephrine, phenylpropanolamine, pirbuterol, reproterol, rimiterol, salbutamol, salmeterol, terbutalin, isoetharine, tulobuterol, orciprenaline or (-)- 4-amino-3,5-dichloro-α-[[[6-[2-(2-pyridinyl)ethoxy]hexyl]-amino]methyl]benzenemethanol; diuretics, e.g. amiloride; anticholinergics e.g. ipratropium, atropine or oxitropium; hormones, e.g. cortisone, hydrocortisone or prednisolone; xanthines e.g. aminophylline, choline theophyllinate, lysine theophyllinate or theophylline; and therapeutic proteins and peptides, e.g. insulin or glucagon. It will be clear to a person skilled in the art that, where appropriate, the medicaments may be used in the form of salts (e.g. as alkali metal or amine salts or as acid addition salts) or as esters (e.g. lower alkyl esters) or as solvates (e.g. hydrates) to optimize the activity and/or stability of the medicament. [004031 Other exemplary therapeutic agents useful for a combination therapy include but are not limited to agents as described above, radiation therapy, hormone antagonists, hormones and their releasing factors, thyroid and antithyroid drugs, estrogens and progestins, androgens, adrenocorticotropic hormone; adrenocortical steroids and their synthetic analogs; inhibitors of the synthesis and actions of adrenocortical hormones, insulin, oral hypoglycemic agents, and the pharmacology of the endocrine pancreas, agents affecting calcification and bone turnover: calcium, phosphate, parathyroid hormone, vitamin D, calcitonin, vitamins such as water-soluble vitamins, vitamin B complex, ascorbic acid, fat-soluble vitamins, vitamins A, K, and E, growth factors, cytokines, chemokines, muscarinic receptor agonists and antagonists; anticholinesterase agents; agents acting at the neuromuscular junction and/or autonomic ganglia; catecholamines, sympathomimetic drugs, and adrenergic receptor agonists or antagonists; and 5-hydroxytryptamine (5-HT, serotonin) receptor agonists and antagonists.
[00404] Therapeutic agents can also include agents for pain and inflammation such as histamine and histamine antagonists, bradykinin and bradykinin antagonists, 5-hydroxytryptamine (serotonin), lipid substances that are generated by biotransformation of the products of the selective hydrolysis of membrane phospholipids, eicosanoids, prostaglandins, thromboxanes, leukotrienes, aspirin, nonsteroidal anti-inflammatory agents, analgesic-antipyretic agents, agents that inhibit the synthesis of prostaglandins and thromboxanes, selective inhibitors of the inducible cyclooxygenase, selective inhibitors of the inducible cyclooxygenase-2, autacoids, paracrine hormones, somatostatin, gastrin, cytokines that mediate interactions involved in humoral and cellular immune responses, lipid-derived autacoids, eicosanoids, β-adrenergic agonists, ipratropium, glucocorticoids, methylxanthines, sodium channel blockers, opioid receptor agonists, calcium channel blockers, membrane stabilizers and leukotriene inhibitors.
[00405] Additional therapeutic agents contemplated herein include diuretics, vasopressin, agents affecting the renal conservation of water, rennin, angiotensin, agents useful in the treatment of myocardial ischemia, antihypertensive agents, angiotensin converting enzyme inhibitors, β-adrenergic receptor antagonists, agents for the treatment of hypercholesterolemia, and agents for the treatment of dyslipidemia.
[00406] Other therapeutic agents contemplated include drugs used for control of gastric acidity, agents for the treatment of peptic ulcers, agents for the treatment of gastroesophageal reflux disease, prokinetic agents, antiemetics, agents used in irritable bowel syndrome, agents used for diarrhea, agents used for constipation, agents used for inflammatory bowel disease, agents used for biliary disease, agents used for pancreatic disease. Therapeutic agents used to treat protozoan infections, drugs used to treat Malaria, Amebiasis, Giardiasis, Trichomoniasis, Trypanosomiasis, and/or Leishmaniasis, and/or drugs used in the chemotherapy of helminthiasis. Other therapeutic agents include antimicrobial agents, sulfonamides, trimethoprim-sulfamethoxazole quinolones, and agents for urinary tract infections, penicillins, cephalosporins, and other, β-Lactam antibiotics, an agent comprising an aminoglycoside, protein synthesis inhibitors, drugs used in the chemotherapy of tuberculosis, mycobacterium avium complex disease, and leprosy, antifungal agents, antiviral agents including nonretroviral agents and antiretroviral agents.
[00407] Examples of therapeutic antibodies that can be combined with a subject compound include but are not limited to anti-receptor tyrosine kinase antibodies (cetuximab, panitumumab, trastuzumab), anti CD20 antibodies (rituximab, tositumomab), and other antibodies such as alemtuzumab, bevacizumab, and gemtuzumab.
[00408] Moreover, therapeutic agents used for immunomodulation, such as immunomodulators, immunosuppressive agents, tolerogens, and immunostimulants are contemplated by the methods herein. In addition, therapeutic agents acting on the blood and the blood-forming organs, hematopoietic agents, growth factors, minerals, and vitamins, anticoagulant, thrombolytic, and antiplatelet drugs.
J00409] Further therapeutic agents that can be combined with a subject compound may be found in Goodman and
Gilman's "The Pharmacological Basis of Therapeutics" Tenth Edition edited by Hardman, Limbird and Gilman or the Physician's Desk Reference, both of which are incorporated herein by reference in their entirety.
[00410] The compounds described herein can be used in combination with the agents disclosed herein or other suitable agents, depending on the condition being treated. Hence, in some embodiments the compounds of the invention will be co-administer with other agents as described above. When used in combination therapy, the compounds described herein may be administered with the second agent simultaneously or separately. This administration in combination can include simultaneous administration of the two agents in the same dosage form, simultaneous administration in separate dosage forms, and separate administration. That is, a compound described herein and any of the agents described above can be formulated together in the same dosage form and administered simultaneously. Alternatively, a compound of the present invention and any of the agents described above can be simultaneously administered, wherein both the agents are present in separate formulations. In another alternative, a compound of the present invention can be administered just followed by and any of the agents described above, or vice versa. In the separate administration protocol, a compound of the present invention and any of the agents described above may be administered a few minutes apart, or a few hours apart, or a few days apart.
[00411] The examples and preparations provided below further illustrate and exemplify the compounds of the present invention and methods of preparing such compounds. It is to be understood that the scope of the present invention is not limited in any way by the scope of the following examples and preparations. In the following examples molecules with a single chiral center, unless otherwise noted, exist as a racemic mixture. Those molecules with two or more chiral centers, unless otherwise noted, exist as a racemic mixture of diastereomers. Single enantiomers/diastereomers may be obtained by methods known to those skilled in the art.
[00412] Administration of the compounds of the present invention can be effected by any method that enables delivery of the compounds to the site of action. These methods include oral routes, intraduodenal routes, parenteral injection (including intravenous, intraarterial, subcutaneous, intramuscular, intravascular, intraperitoneal or infusion), topical (e.g. transdermal application), rectal administration, via local delivery by catheter or stent. Compounds can also abe administered intraadiposally or intrathecally.
[00413] The amount of the compound administered will be dependent on the mammal being treated, the severity of the disorder or condition, the rate of administration, the disposition of the compound and the discretion of the prescribing physician. However, an effective dosage is in the range of about 0.001 to about 100 mg per kg body weight per day, preferably about 1 to about 35 mg/kg/day, in single or divided doses. For a 70 kg human, this would amount to about 0.05 to 7 g/day, preferably about 0.05 to about 2.5 g/day. In some instances, dosage levels below the lower limit of the aforesaid range may be more than adequate, while in other cases still larger doses may be employed without causing any harmful side effect, e.g. bydividing such larger doses into several small doses for administration throughout the day.
[00414] The compound may be applied as a sole therapy or may involve one or more other anti-tumor substances, for example those selected from, mitotic inhibitors, for example vinblastine; alkylating agents, for example cis- platin, carboplatin and cyclophosphamide; anti-metabolites, for example 5-fluorouracil, cytosine arabinside and hydroxyurea, or, for example, one of the preferred anti-metabolites disclosed in European Patent Application No. 239362 such as N- (5- [N- (3, 4-dihydro-2-methyl-4- oxoquinazolin-6-yhnethyl)-N-methylamino]-2- thenoyl)-L-glutamic acid; growth factor inhibitors; cell cycle inhibitors; intercalating antibiotics, for example adriamycin and bleomycin; enzymes, for example, interferon; and anti-hormones, for example anti- estrogens such as Nolvadex™ (tamoxifen) or, for example anti-androgens such as Casodex™ (4'-cyano-3- (4- fluorophenylsulphonyl)-2-hydroxy-2-methyl-3'- (trifluoromethyl) propionanilide). Such conjoint treatment may be achieved by way of the simultaneous, sequential or separate dosing of the individual components of treatment.
[00415] In some embodiments, a compound of the invention is administered in a single dose. Typically, such administration will be by injection, e.g., intravenous injection, in order to introduce the agent quickly. However, other routes may be used as appropriate. A single dose of a compound of the invention may also be used for treatment of an acute condition.
[00416] In some embodiments, a compound of the invention is administered in multiple doses. Dosing may be about once, twice, three times, four times, five times, six times, or more than six times per day. Dosing may be about once a month, once every two weeks, once a week, or once every other day. In another embodiment a compound of the invention and another agent are administered together about once per day to about 6 times per day. In another embodiment the administration of a compound of the invention and an agent continues for less than about 7 days. In yet another embodiment the administration continues for more than about 6, 10, 14, 28 days, two months, six months, or one year. In some cases, continuous dosing is achieved and maintained as long as necessary.
[00417] Administration of the agents of the invention may continue as long as necessary. In some embodiments, an agent of the invention is administered for more than 1, 2, 3, 4, 5, 6, 7, 14, or 28 days. In some embodiments, an agent of the invention is administered for less than 28, 14, 7, 6, 5, 4, 3, 2, or 1 day. In some embodiments, an agent of the invention is administered chronically on an ongoing basis, e.g., for the treatment of chronic effects.
[00418] An effective amount of a compound of the invention may be administered in either single or multiple doses by any of the accepted modes of administration of agents having similar utilities, including rectal, buccal, intranasal and transdermal routes, by intra-arterial injection, intravenously, intraperitoneally, parenterally, intramuscularly, subcutaneously, orally, topically, or as an inhalant.
[00419] The compositions of the invention may also be delivered via an impregnated or coated device such as a stent, for example, or an artery-inserted cylindrical polymer. Such a method of administration may, for example, aid in the prevention or amelioration of restenosis following procedures such as balloon angioplasty. Without being bound by theory, compounds of the invention may slow or inhibit the migration and proliferation of smooth muscle cells in the arterial wall which contribute to restenosis. A compound of the invention may be administered, for example, by local delivery from the struts of a stent, from a stent graft, from grafts, or from the cover or sheath of a stent. In some embodiments, a compound of the invention is admixed with a matrix. Such a matrix may be a polymeric matrix, and may serve to bond the compound to the stent. Polymeric matrices suitable for such use, include, for eample, lactone-based polyesters or copolyesters such as polylactide, polycaprolactonglycolide, polyorthoesters, polyanhydrides, polyaminoacids, polysaccharides, polyphosphazenes, poly (ether-ester) copolymers (e.g. PEO-PLLA); polydimethylsiloxane, poly(ethylene- vinylacetate), acrylate-based polymers or copolymers (e.g. polyhydroxyethyl methylmethacrylate, polyvinyl pyrrolidinone), fiuorinated polymers such as polytetrafluoroethylene and cellulose esters. Suitable matrices may be nondegrading or may degrade with time, releasing the compound or compounds. Compounds of the invention may be applied to the surface of the stent by various methods such as dip/spin coating, spray coating, dip- coating, and/or brush-coating. The compounds may be applied in a solvent and the solvent may be allowed to evaporate, thus forming a layer of compound onto the stent. Alternatively, the compound may be located in the body of the stent or graft, for example in microchannels or micropores. When implanted, the compound diffuses out of the body of the stent to contact the arterial wall. Such stents may be prepared by dipping a stent manufactured to contain such micropores or microchannels into a solution of the compound of the invention in a suitable solvent, followed by evaporation of the solvent. Excess drug on the surface of the stent may be removed via an additional brief solvent wash. In yet other embodiments, compounds of the invention may be covalently linked to a stent or graft. A covalent linker may be used which degrades in vivo, leading to the release of the compound of the invention. Any bio-labile linkage may be used for such a purpose, such as ester, amide or anhydride linkages. Compounds of the invention may additionally be administered intravascularly from a balloon used during angioplasty. Extravascular administration of the compounds via the pericard or via advential application of formulations of the invention may also be performed to decrease restenosis. A variety of stent devices which may be used as described are disclosed, for example, in the following references, all of which are hereby incorporated by reference: U.S. Pat. No. 5451233; U.S. Pat. No. 5040548; U.S. Pat. No. 5061273; U.S. Pat. No. 5496346; U.S. Pat. No. 5292331; U.S. Pat. No. 5674278; U.S. Pat. No. 3657744; U.S. Pat. No. 4739762; U.S. Pat. No. 5195984; U.S. Pat. No. 5292331; U.S. Pat. No. 5674278; U.S. Pat. No. 5879382; U.S. Pat. No. 6344053; U.S. Pat. No. 4762129; U.S. Pat. No. 6152946; U.S. Pat. No. 6663652; U.S. Pat. No. 6027520 6676682; U.S. Pat. No. 6663652; U.S. Pat. No. 6872216; U.S. Pat. No. 6027520; U.S. Pat. No. 6114653; U.S. Pat. No. 5852277; U.S. Pat. No. 5843120; U.S. Pat. No. 5643312; U.S. Pat. No. 5733303 ; U.S. Pat. No. 5597378; U.S. Pat. No. 5653727; U.S. Pat. No. 4762129; U.S. Pat. No. 5922021; U.S. Pat. No. 3657744; U.S. Pat. No. 4739762; U.S. Pat. No. 5195984; U.S. Pat. No. 5451233; U.S. Pat. No. 3657744; U.S. Pat. No. 4739762; U.S. Pat. No. 5195984; U.S. Pat. No. 4739762; U.S. Pat. No. 3657744; U.S. Pat. No. 4739762; U.S. Pat. No. 5195984; U.S. Pat. No. 5292331 ; U.S. Pat. No. 5674278; U.S. Pat. No. 5643312; U.S. Pat. No. 5879370; U.S. Pat. No. 5421955; U.S. Pat. No. 5514154; U.S. Pat. No. 5603721; U.S. Pat. No. 5421955; U.S. Pat. No. 5514154; U.S. Pat. No. 5603721; U.S. Pat. No. 5292331; U.S. Pat. No. 5674278; U.S. Pat. No. 5879382; U.S. Pat. No. 6344053; U.S. Pat. No. 5728067; U.S. Pat. No. 5980486; U.S. Pat. No. 6129708; U.S. Pat. No. 5733303; U.S. Pat. No. 5843120; U.S. Pat. No. 5972018; U.S. Pat. No. 5972018; U.S. Pat. No. 5733303; U.S. Pat. No. 5843120; U.S. Pat. No. 4739762; U.S. Pat. No. 5195984; U.S. Pat. No. 5902332; U.S. Pat. No. 5156594; U.S. Pat. No. 5395334; U.S. Pat. No. 6090083; U.S. Pat. No. 5639278; U.S. Pat. No. 6051020; U.S. Pat. No. 6117167; U.S. Pat. No. 5632772; U.S. Pat. No. 6165213; U.S. Pat. No. 4762129; U.S. Pat. No. 5156594; U.S. Pat. No. 5217482; U.S. Pat. No. 5395334; U.S. Pat. No. 4641653; U.S. Pat. No. 4739762; U.S. Pat. No. 5922021; U.S. Pat. No. 5895406; U.S. Pat. No. 6251920; U.S. Pat. No. 6120536; U.S. Pat. No. 5292331; U.S. Pat. No. 5674278; U.S. Pat. No. 5879382; U.S. Pat. No. 6344053; U.S. Pat. No. 5609627; U.S. Pat. No. 6251920; U.S. Pat. No. 5733303; U.S. Pat. No. 5843120; U.S. Pat. No. 5972018; U.S. Pat. No. 6344053; U.S. Pat. No. 5292331; U.S. Pat. No. 5674278; U.S. Pat. No. 5879382; U.S. Pat. No. 5653760; U.S. Pat. No. 6190358; U.S. Pat. No. 6210364; U.S. Pat. No. 6283939; U.S. Pat. No. 6605057; U.S. Pat. No. 5292331; U.S. Pat. No. 5674278; U.S. Pat. No. 5879382; U.S. Pat. No. 6344053; U.S. Pat. No. 5423851; U.S. Pat. No. 6007575; U.S. Pat. No. 5501759; U.S. Pat. No. 5674208; U.S. Pat. No. 5843032; U.S. Pat. No. 5961765; U.S. Pat. No. 6027477; U.S. Pat. No. 6319228; U.S. Pat. No. 6471673; U.S. Pat. No. 6190358; U.S. Pat. No. 6605057; U.S. Pat. No. 6858037; U.S. Pat. No.
7001358; U.S. Pat. No. 5156594; U.S. Pat. No. 5217482; U.S. Pat. No. 5395334; U.S. Pat. No. 5702439; U.S.
Pat. No. 5501759; U.S. Pat. No. 5674208; U.S. Pat. No. 5843032; U.S. Pat. No. 5961765; U.S. Pat. No.
6027477; U.S. Pat. No. 6319228; U.S. Pat. No. 6471673; U.S. Pat. No. 5759192; U.S. Pat. No. 6527789; U.S.
Pat. No. 5147302; U.S. Pat. No. 5342307; U.S. Pat. No. 6290485; U.S. Pat. No. 6352551; U.S. Pat. No.
6402778; U.S. Pat. No. 6488694; U.S. Pat. No. 6511505; U.S. Pat. No. 6613073; U.S. Pat. No. 6582458; U.S.
Pat. No. 5820594; U.S. Pat. No. 5824173; U.S. Pat. No. 5538510; U.S. Pat. No. 4323071; U.S. Pat. No.
4762129; U.S. Pat. No. 4846186; U.S. Pat. No. 5156594; U.S. Pat. No. 5217482; U.S. Pat. No. 5395334; U.S.
Pat. No. 5156594; U.S. Pat. No. 4323071; U.S. Pat. No. 5040548; U.S. Pat. No. 5061273; U.S. Pat. No.
5451233; U.S. Pat. No. 5496346; U.S. Pat. No. 5496275; U.S. Pat. No. 5496346; U.S. Pat. No. 5040548; U.S.
Pat. No. 5061273; U.S. Pat. No. 5451233; U.S. Pat. No. 5496346; U.S. Pat. No. 4596563; U.S. Pat. No.
5040548; U.S. Pat. No. 5061273; U.S. Pat. No. 5350395; U.S. Pat. No. 5451233; U.S. Pat. No. 5445625; U.S.
Pat. No. 6083213 ; U.S. Pat. No. 6475195; U.S. Pat. No. 5421955; U.S. Pat. No. 5514154; U.S. Pat. No.
5603721; U.S. Pat. No. 5292331; U.S. Pat. No. 5674278; U.S. Pat. No. 5879382; U.S. Pat. No. 6344053; U.S.
Pat. No. 6238415; U.S. Pat. No. 5421955; U.S. Pat. No. 5514154; and U.S. Pat. No. 5603721. [00421] The compounds of the invention may be administered in dosages as described herein (see, e.g., Compositions).
It is known in the art that due to intersubject variability in compound pharmacokinetics, individualization of dosing regimen is necessary for optimal therapy. Dosing for a compound of the invention may be found by routine experimentation. [00422] When a compound of the invention, is administered in a composition that comprises one or more agents, and the agent has a shorter half-life than the compound of the invention unit dose forms of the agent and the compound of the invention may be adjusted accordingly. See e.g., Compositions. [00423] The subject pharmaceutical composition may, for example, be in a form suitable for oral administration as a tablet, capsule, pill, powder, sustained release formulations, solution, suspension, for parenteral injection as a sterile solution, suspension or emulsion, for topical administration as an ointment or cream or for rectal administration as a suppository. The pharmaceutical composition may be in unit dosage forms suitable for single administration of precise dosages. The pharmaceutical composition will include a conventional pharmaceutical carrier or excipient and a compound according to the invention as an active ingredient. In addition, it may include other medicinal or pharmaceutical agents, carriers, adjuvants, etc. [00424] Exemplary parenteral administration forms include solutions or suspensions of active compound in sterile aqueous solutions, for example, aqueous propylene glycol or dextrose solutions. Such dosage forms can be suitably buffered, if desired. [00425] The activity of the compounds of the present invention may be determined by the following procedure, as well as the procedure described in the examples below. N-terminal 6 His-tagged, constitutively active kinase is expressed in E. coli and protein is purified by conventional methods (Ann et al. Science 1994, 265, 966-970).
The activity of the kinase is assessed by measuring the incorporation of γ-33P-phosphate from γ -33P-ATP onto
N-terminal His tagged substrate, which is expressed in E. coli and is purified by conventional methods, in the presence of the kinase. The assay is carried out in 96-well polypropylene plate. The incubation mixture (100, μL) comprises of 25 mM Hepes, pH 7.4, 10 mM MgCl2, 5 mM β-glycerolphosphate, 100 μM Na-orthovanadate,
5 mM DTT, 5 nM kinase, and 1 μM substrate. Inhibitors are suspended in DMSO, and all reactions, including controls are performed at a final concentration of 1% DMSO. Reactions are initiated by the addition of 10 μM ATP (with 0.5 μCi γ-33P- ATP/well) and incubated at ambient temperature for 45 minutes. Equal volume of 25% TCA is added to stop the reaction and precipitate the proteins. Precipitated proteins are trapped onto glass fiber B filterplates, and excess labeled ATP washed off using a Tomtec MACH III harvestor. Plates are allowed to air-dry prior to adding 30 μL/well of Packard Microscint 20, and plates are counted using a Packard TopCount.
[00426] The examples and preparations provided below further illustrate and exemplify the compounds of the present invention and methods of preparing such compounds. It is to be understood that the scope of the present invention is not limited in any way by the scope of the following examples and preparations. In the following examples molecules with a single chiral center, unless otherwise noted, exist as a racemic mixture. Those molecules with two or more chiral centers, unless otherwise noted, exist as a racemic mixture of diastereomers. Single enantiomers/diastereomers may be obtained by methods known to those skilled in the art.
EXAMPLES
[00427] Example 1: Expression and Inhibition Assays of pllθα/p85α, pllθβ/p85α, pllθδ/p85α, and pllOγ:
[00428] Class I PB-Ks can be either purchased (pi 10α/p85α, pi 10β/p85α, pi 10δ/p85α from Upstate, and pi lOγ from Sigma) or expressed as previously described (Knight et al., 2004). IC50 values are measured using either a standard TLC assay for lipid kinase activity (described below) or a high-throughput membrane capture assay. Kinase reactions are performed by preparing a reaction mixture containing kinase, inhibitor (2% DMSO final concentration), buffer (25 mM HEPES, pH 7.4, 10 mM MgC12), and freshly sonicated phosphatidylinositol (100 μg/ml). Reactions are initiated by the addition of ATP containing 10 μCi of γ-32P- ATP to a final concentration 10 or 100 μM and allowed to proceed for 5 minutes at room temperature. For TLC analysis, reactions are then terminated by the addition of 105 μl IN HCl followed by 160 μl CHC13:MeOH (1:1). The biphasic mixture is vortexed, briefly centrifuged, and the organic phase is transferred to a new tube using a gel loading pipette tip precoated with CHCl3. This extract is spotted on TLC plates and developed for 3 - 4 hours in a 65:35 solution of n-ρropanol:lM acetic acid. The TLC plates are then dried, exposed to a phosphorimager screen (Storm, Amersham), and quantitated. For each compound, kinase activity is measured at 10 — 12 inhibitor concentrations representing two-fold dilutions from the highest concentration tested (typically, 200 μM). For compounds showing significant activity, IC50 determinations are repeated two to four times, and the reported value is the average of these independent measurements.
[00429] Other commercial kits or systems for assaying PI3-K activities are avaiable. The commercially available kits or systems can be used to screen for inhibitors and/or agonists of PD-Ks including but not limited to PI 3-Kinase α, β, δ, and γ. Anr exemplary system is PI 3-Kinase (human) HTRF™ Assay from Upstate. The assay can be carried out according to the procedures suggested by the manufacturer. Briefly, the assay is a time resolved FRET assay that indirectly measures PIP3 product formed by the activity of a PI3- K. The kinase reaction is performed in a microtitre plate (e.g., a 384 well microtitre plate). The total reaction volume is approximately 20ul per well. In the first step, each well receives 2ul of test compound in 20% dimethylsulphoxide resulting in a 2% DMSO final concentration. Next, approximately 14.5ul of a kinase/PIP2 mixture (diluted in IX reaction buffer) is added per well for a final concentration of 0.25-
0.3ug/ml kinase and lOuM PIP2. The plate is sealed and incubated for 15 minutes at room temperature. To start the reaction, 3.5ul of ATP (diluted in IX reaction buffer) is added per well for a final concentration of lOuM ATP. The plate is sealed and incubated for 1 hour at room temperature. The reaction is stopped by adding 5ul of Stop Solution per well and then 5ul of Detection Mix is added per well. The plate is sealed, incubated for 1 hour at room temperature, and then read on an appropriate plate reader. Data is analyzed and IC50s are generated using GraphPad Prism 5.
[00430] Example 2: Expression and Inhibition Assays of AbI
[00431] The compounds described herein can be assayed in triplicate against recombinant full-length AbI or
AbI (T315I) (Upstate) in an assay containing 25 mM HEPES, pH 7.4, 10 mM MgC12, 200 μM ATP (2.5 μCi of γ-32P-ATP), and 0.5 mg/mL BSA. The optimized AbI peptide substrate EAIY AAPF AKKK is used as phosphoacceptor (200 μM). Reactions are terminated by spotting onto phosphocellulose sheets, which are washed with 0.5% phosphoric acid (approximately 6 times, 5-10 minutes each). Sheets are dried and the transferred radioactivity quantitated by phosphorimaging.
[00432] Example 3: Expression and Inhibition Assays of Hck
[00433] The compounds described herein can be assayed in triplicate against recombinant full-length Hck in an assay containing 25 mM HEPES, pH 7.4, 10 mM MgC12, 200 μM ATP (2.5 μCi of γ-32P-ATP), and 0.5 mg/mL BSA. The optimized Src family kinase peptide substrate EIYGEFKKK is used as phosphoacceptor (200 μM). Reactions are terminated by spotting onto phosphocellulose sheets, which are washed with 0.5% phosphoric acid (approximately 6 times, 5-10 minutes each). Sheets are dried and the transferred radioactivity quantitated by phosphorimaging.
[00434] Example 4: Expression and Inhibition Assays of Inulsin Receptor (IR)
[00435] The compounds described herein can be assayed in triplicate against recombinant insulin receptor kinase domain (Upstate) in an assay containing 25 mM HEPES, pH 7.4, 10 mM MgC12, 10 mM MnC12, 200 μM ATP (2.5 μCi of γ-32P-ATP), and 0.5 mg/mL BSA. Poly E-Y (Sigma; 2 mg/mL) is used as a substrate. Reactions are terminated by spotting onto nitrocellulose, which is washed with IM NaCl/ 1% phosphoric acid (approximately 6 times, 5-10 minutes each). Sheets are dried and the transferred radioactivity quantitated by phosphorimaging.
[00436] Example 5: Expression and Inhibition Assays of Src
[00437] The compounds described herein can be assayed in triplicate against recombinant full-length Src or
Src (T338I) in an assay containing 25 mM HEPES, pH 7.4, 10 mM MgC12, 200 μM ATP (2.5 μCi of γ-32P- ATP), and 0.5 mg/mL BSA. The optimized Src family kinase peptide substrate EIYGEFKKK is used as phosphoacceptor (200 μM). Reactions are terminated by spotting onto phosphocellulose sheets, which are washed with 0.5% phosphoric acid (approximately 6 times, 5-10 minutes each). Sheets were dried and the transferred radioactivity quantitated by phosphorimaging.
[00438] Example 6: Expression and Inhibition Assays of DNA-PK (DNAK)
[00439] DNA-PK can be purchased from Promega and assayed using the DNA-PK Assay System (Promega) according to the manufacturer's instructions.
[00440] Example 7: Expression and Inhibition Assays mTOR
[00441] The compounds described herein can be tested against recombinant mTOR (Invitrogen) in an assay containing 50 mM HEPES, pH 7.5, ImM EGTA, 10 mM MgC12, 2.5 mM, 0.01% Tween, 10 μM ATP (2.5 μCi of μ-32P-ATP), and 3 μg/mL BSA. Rat recombinant PHAS- 1/4EBP1 (Calbiochem; 2 mg/mL) is used as a substrate. Reactions are terminated by spotting onto nitrocellulose, which is washed with IM NaCl/ 1% phosphoric acid (approximately 6 times, 5-10 minutes each). Sheets are dried and the transferred radioactivity quantitated by phosphorimaging.
[00442] Other kits or systems for assaying mTOR activity are commercially avaiable. For instance, one can use Invitrogen's LanthaScreen™ Kinase assay to test the inhibitors of mTOR disclosed herein. This assay is a time resolved FRET platform that measures the phosphorylation of GFP labeled 4EBP 1 by mTOR kinase. The kinase reaction is performed in a white 384 well microtitre plate. The total reaction volume is 20ul per well and the reaction buffer composition is 5OmM HEPES pH7.5, 0.01% Polysorbate 20, ImM EGTA, 1OmM MnC12, and 2mM DTT. In the first step, each well receives 2ul of test compound in 20% dimethylsulphoxide resulting in a 2% DMSO final concentration. Next, 8ul of mTOR diluted in reaction buffer is added per well for a 60ng/ml final concentration. To start the reaction, lOul of an ATP/GFP-4EBP1 mixture (diluted in reaction buffer) is added per well for a final concentration of lOuM ATP and 0.5uM GFP-4EBP1. The plate is sealed and incubated for 1 hour at room temperature. The reaction is stopped by adding lOul per well of a Tb-anti-pT46 4EBP 1 antibody/EDTA mixture (diluted in TR-FRET buffer) for a final concentration of 1.3nM antibody and 6.7mM EDTA. The plate is sealed, incubated for 1 hour at room temperature, and then read on a plate reader set up for LanthaScreen™ TR-FRET. Data is analyzed and IC50s are generated using GraphPad Prism 5.
[00443] Example 8: Expression and Inhibition Assays of Vascular endothelial growth receptor
[00444] The compounds described herein can be tested against recombinant KDR receptor kinase domain
(Invitrogen) in an assay containing 25 mM HEPES, pH 7.4, 10 mM MgC12, 0.1% BME, 10 μM ATP (2.5 μCi of μ-32P-ATP), and 3 μg/mL BSA. Poly E-Y (Sigma; 2 mg/mL) is used as a substrate. Reactions are terminated by spotting onto nitrocellulose, which is washed with IM NaCl/1% phosphoric acid (approximately 6 times, 5-10 minutes each). Sheets are dried and the transferred radioactivity quantitated by phosphorimaging.
[00445] Example 9: Expression and Inhibition Assays of Ephrin receptor B4 (EphB4)
[00446] The compounds described herein can be tested against recombinant Ephrin receptor B4 kinase domain (Invitrogen) in an assay containing 25 mM HEPES, pH 7.4, 10 mM MgC12, 0.1% BME, 10 μM ATP (2.5 μCi of μ-32P-ATP), and 3 μg/mL BSA. Poly E-Y (Sigma; 2 mg/mL) is used as a substrate. Reactions are terminated by spotting onto nitrocellulose, which is washed with IM NaCl/ 1% phosphoric acid (approximately 6 times, 5-10 minutes each). Sheets are dried and the transferred radioactivity quantitated by phosphorimaging.
[00447] Example 10: Expression and Inhibition Assays of Epidermal growth factor receptor (EGFR)
[00448] The compounds described herein can be tested against recombinant EGF receptor kinase domain
(Invitrogen) in an assay containing 25 mM HEPES, pH 7.4, 10 mM MgC12, 0.1% BME, 10 μM ATP (2.5 μCi of μ-32P-ATP), and 3 μg/mL BSA. Poly E-Y (Sigma; 2 mg/mL) is used as a substrate. Reactions are terminated by spotting onto nitrocellulose, which is washed with IM NaCl/ 1% phosphoric acid (approximately 6 times, 5-10 minutes each). Sheets are dried and the transferred radioactivity quantitated by phosphorimaging.
[00449] Example 11: Expression and Inhibition Assays of KIT Assay
[00450] The compounds described herein can be tested against recombinant KIT kinase domain (Invitrogen) in an assay containing 25 mM HEPES, pH 7.4, 10 mM MgC12, ImM DTT, 1OmM MnC12, 10 μM ATP (2.5 μCi of μ-32P-ATP), and 3 μg/mL BSA. Poly E-Y (Sigma; 2 mg/mL) is used as a substrate. Reactions are terminated by spotting onto nitrocellulose, which is washed with IM NaCl/1% phosphoric acid (approximately 6 times, 5-10 minutes each). Sheets are dried and the transferred radioactivity quantitated by phosphorimaging.
[00451] Example 12: Expression and Inhibition Assays of RET
[00452] The compounds described herein can be tested against recombinant RET kinase domain (Invitrogen) in an assay containing 25 mM HEPES, pH 7.4, 10 mM MgC12, 2.5mM DTT, 10 μM ATP (2.5 μCi of μ-32P- ATP), and 3 μg/mL BSA. The optimized AbI peptide substrate EAIY AAPF AKKK is used as phosphoacceptor (200 μM). Reactions are terminated by spotting onto phosphocellulose sheets, which are washed with 0.5% phosphoric acid (approximately 6 times, 5-10 minutes each). Sheets are dried and the transferred radioactivity quantitated by phosphorimaging.
[00453] Example 13: Expression and Inhibition Assays of Platelet derived growth factor receptor
(PDGFR)
[00454] The compounds described herein can be tested against recombinant PDG receptor kinase domain
(Invitrogen) in an assay containing 25 mM HEPES, pH 7.4, 10 mM MgC12, 2.5mM DTT,10 μM ATP (2.5 μCi of μ-32P-ATP), and 3 μg/mL BSA. The optimized AbI peptide substrate EAIY AAPF AKKK is used as phosphoacceptor (200 μM). Reactions are terminated by spotting onto phosphocellulose sheets, which are washed with 0.5% phosphoric acid (approximately 6 times, 5-10 minutes each). Sheets are dried and the transferred radioactivity quantitated by phosphorimaging.
[00455] Example 14: Expression and Inhibition Assays of FMS-related tyrosine kinase 3 (FLT-3)
[00456] The compounds described herein can be tested against recombinant FLT-3 kinase domain
(Invitrogen) in an assay containing 25 mM HEPES, pH 7.4, 10 mM MgC12, 2.5mM DTT1IO μM ATP (2.5 μCi of μ-32P-ATP), and 3 μg/mL BSA. The optimized AbI peptide substrate EAIY AAPF AKKK is used as phosphoacceptor (200 μM). Reactions are terminated by spotting onto phosphocellulose sheets, which are washed with 0.5% phosphoric acid (approximately 6 times, 5-10 minutes each). Sheets are dried and the transferred radioactivity quantitated by phosphorimaging.
[00457] Example 15: Expression and Inhibition Assays of TEK receptor tyrosine kinase (TIE2)
[00458] The compounds described herein can be tested against recombinant TIE2 kinase domain (Invitrogen) in an assay containing 25 mM HEPES, pH 7.4, 10 mM MgC12, 2mM DTT, 1OmM MnC12, 10 μM ATP (2.5 μCi of μ-32P-ATP), and 3 μg/mL BSA. Poly E-Y (Sigma; 2 mg/mL) is used as a substrate. Reactions are terminated by spotting onto nitrocellulose, which is washed with IM NaCl/1% phosphoric acid (approximately 6 times, 5-10 minutes each). Sheets are dried and the transferred radioactivity quantitated by phosphorimaging.
[00459] Example 16: B Cell Activation and Proliferation Assay
[00460] The ability of one or more subject compounds to inhibit B cell activitation and proliferation is determined according to standard procedures known in the art. For example, an in vitro cellular proliferation assay is established that measures the metabolic activity of live cells. The assay is performed in a 96 well microtiter plate using Alamar Blue reduction. Balb/c splenic B cells are purified over a Ficoll-Paque™ PLUS gradient followed by magnetic cell separation using a MACS B cell Isolation Kit (Miletenyi). Cells are plated in 90ul at 50,000 cells/well in B Cell Media (RPMI + 10%FBS + Penn/Strep + 5OuM bME + 5mM HEPES). A compound disclosed herein is diluted in B Cell Media and added in a lOul volume. Plates are incubated for
30min at 37C and 5% CO2 (0.2% DMSO final concentration). A 50ul B cell stimulation cocktail is then added containing either lOug/ml LPS or 5ug/ml F(ab')2 Donkey anti-mouse IgM plus 2ng/ml recombinant mouse IL4 in B Cell Media. Plates are incubated for 72 hours at 370C and 5% CO2. A volume of 15uL of Alamar Blue reagent is added to each well and plates are incubated for 5 hours at 37C and 5% CO2. Alamar Blue fluoresce is read at 560Ex/590Em, and IC50 or EC50 values are calculated using GraphPad Prism 5.
[00461] Example 17: Tumor Cell Line Proliferation Assay
[00462] The ability of one or more subject compounds to inhibit tumor cell line proliferation is determined according to standard procedures known in the art. For instance, an in vitro cellular proliferation assay can be performed to measure the metabolic activity of live cells. The assay is performed in a 96 well microtiter plate using Alamar Blue reduction. Human tumor cell lines are obtained from ATCC (e.g., MCF7, U-87 MG, MDA-MB-468, PC-3), grown to confluency in T75 flasks, trypsinized with 0.25% trypsin, washed one time with Tumor Cell Media (DMEM + 10%FBS), and plated in 90ul at 5,000 cells/well in Tumor Cell Media. A compound disclosed herein is diluted in Tumor Cell Media and added in a lOul volume. Plates are incubated for 72 hours at 37C and 5% CO2. A volume of lOuL of Alamar Blue reagent is added to each well and plates are incubated for 3 hours at 37C and 5% CO2. Alamar Blue fluoresce is read at 560Ex/590Em, and IC50 values are calculated using GraphPad Prism 5.
[00463] Example 18: Antitumor Activity in Vivo
[00464] The compounds described herein can be evaluated in a panel of human and murine tumor models.
[00465] Paclitaxel-refractorv Tumor Models
[00466] 1. Clinically-derived Ovarian Carcinoma Model.
[00467] This tumor model is established from a tumor biopsy of an ovarian cancer patient. Tumor biopsy is taken from the patient.
[00468] The compounds described herein are administered to nude mice bearing staged tumors using an every 2 days x 5 schedule.
[00469] 2. A2780Tax Human Ovarian Carcinoma Xenograft (Mutated Tubulin).
[00470] A2780Tax is a paclitaxel-resistant human ovarian carcinoma model. It is derived from the sensitive parent A2780 line by co-incubation of cells with paclitaxel and verapamil, an MDR-reversal agent. Its resistance mechanism has been shown to be non-MDR related and is attributed to a mutation in the gene encoding the beta-tubulin protein.
[00471] The compounds described herein can be administered to mice bearing staged tumors on an every 2 days x 5 schedule.
[00472] 3. HCTl 16/VM46 Human Colon Carcinoma Xenograft (Multi-Drug Resistant).
[00473] HCTl 16/VM46 is an MDR-resistant colon carcinoma developed from the sensitive HCTl 16 parent line. In vivo, grown in nude mice, HCTl 16/VM46 has consistently demonstrated high resistance to paclitaxel.
[00474] The compounds described herein can be administered to mice bearing staged tumors on an every 2 days x 5 schedule.
[00475] 5. M5076 Murine Sarcoma Model
[00476] M5076 is a mouse fibrosarcoma that is inherently refractory to paclitaxel in vivo.
[00477] The compounds described herein can be administered to mice bearing staged tumors on an every 2 days x 5 schedule. [00478] One or more compounds of the invention can be used in combination other therapeutic agents in vivo in the multidrug resistant human colon carcinoma xenografts HCT/VM46 or any other model known in the art including those described herein.
[00479] Example 19: Microsome stability assay
[00480] The stability of one or more subject compounds is determined according to standard procedures known in the art. For example, stability of one or more subject compounds is established by an in vitro assay. In particular, an in vitro microsome stability assay is established that measures stability of one or more subject compounds when reacting with mouse, rat or human microsomes from liver. The microsome reaction with compounds is performed in 1.5 mL Eppendorf tube. Each tube contains 0.1 μL of 10.0 mg/ml NADPH; 75 μL of 20.0 mg/ml mouse, rat or human liver microsome; 0.4 μL of 0.2 M phosphate buffer, and 425 μL of ddH2O. Negative control (without NADPH) tube contains 75 μL of 20.0 mg/ml mouse, rat or human liver microsome; 0.4 μL of 0.2 M phosphate buffer, and 525 μL of ddH2O. The reaction is started by adding 1.0 μL of 10.0 mM tested compound. The reaction tubes are incubated at 37°C. 100 μL sample is collected into new Eppendorf tube containing 300 μL cold Methanol at 0, 5, 10, 15, 30 and 60 minutes of reaction. Samples are centrifuged at 15,000 rpm to remove protein. Supernatant of centrifuged sample is transferred to new tube. Concentration of stable compound after reaction with microsome in the supernatant is measured by Liquid Chromatography/Mass Spectrometry (LC-MS).
[00481] Example 20: Plasma stability assay
[00482] The stability of one or more subject compounds in plasma is determined according to standard procedures known in the art. See, e.g., Rapid Commun. Mass Spectrom., 10: 1019-1026. The following procedure is an HPLC-MS/MS assay using human plasma; other species including monkey, dog, rat, and mouse are also available. Frozen, heparinized human plasma is thawed in a cold water bath and spun for 10 minutes at 2000 rpm at 4 0C prior to use. A subject compound is added from a 400 μM stock solution to an aliquot of pre- warmed plasma to give a final assay volume of 400 μL (or 800 μL for half-life determination), containing 5 μM test compound and 0.5 % DMSO. Reactions are incubated, with shaking, for 0 minutes and 60 minutes at 37 0C, or for 0, 15, 30, 45 and 60 minutes at 37 C for half life determination. Reactions are stopped by transferring 50 μL of the incubation mixture to 200 μL of ice-cold acetonitrile and mixed by shaking for 5 minutes. The samples are centrifuged at 6000 x g for 15 minutes at 40C and 120 μL of supernatant removed into clean tubes. The samples are then evaporated to dryness and submitted for analysis by HPLC-MS/MS.
[00483] Where desired, one or more control or reference compounds (5 μM) are tested simultaneously with the test compounds: one compound, propoxycaine, with low plasma stability and another compound, propantheline, with intermediate plasma stability.
[00484] Samples are reconstituted in acetonitrile/methanol/water (1/1/2, v/v/v) and analyzed via (RP)HPLC-
MS/MS using selected reaction monitoring (SRM). The HPLC conditions consist of a binary LC pump with autosampler, a mixed-mode, C 12, 2 x 20 mm column, and a gradient program. Peak areas corresponding to the analytes are recorded by HPLC-MS/MS. The ratio of the parent compound remaining after 60 minutes relative to the amount remaining at time zero, expressed as percent, is reported as plasma stability. In case of half-life determination, the half-life is estimated from the slope of the initial linear range of the logarithmic curve of compound remaining (%) vs. time, assuming first order kinetics.
[00485] Example 21: Chemical Stability [00486] The chemical stability of one or more subject compounds is determined according to standard procedures known in the art. The following details an exemplary procedure for ascertaining chemical stability of a subject compound. The default buffer used for the chemical stability assay is phosphate-buffered saline (PBS) at pH 7.4; other suitable buffers can be used. A subject compound is added from a 100 μM stock solution to an aliquot of PBS (in duplicate) to give a final assay volume of 400 μL, containing 5 μM test compound and 1% DMSO (for half-life determination a total sample volume of 700 μL is prepared). Reactions are incubated, with shaking, for 0 minutes and 24 hours at 37°C; for half-life determination samples are incubated for 0, 2, 4, 6, and 24 hours. Reactions are stopped by adding immediately 100 μL of the incubation mixture to 100 μL of acetonitrile and vortexing for 5 minutes. The samples are then stored at -2O0C until analysis by HPLC-MS/MS. Where desired, a control compound or a reference compound such as chlorambucil (5 μM) is tested simultaneously with a subject compound of interest, as this compound is largely hydrolyzed over the course of 24 hours. Samples are analyzed via (RP)HPLC-MS/MS using selected reaction monitoring (SRM). The HPLC conditions consist of a binary LC pump with autosampler, a mixed-mode, C12, 2 x 20 mm column, and a gradient program. Peak areas corresponding to the analytes are recorded by HPLC-MS/MS. The ratio of the parent compound remaining after 24 hours relative to the amount remaining at time zero, expressed as percent, is reported as chemical stability. In case of half-life determination, the half-life is estimated from the slope of the initial linear range of the logarithmic curve of compound remaining (%) vs. time, assuming first order kinetics.
[00487] Example 22: Akt Kinase Assay
[00488] Cells comprising components of the Akt/mTOR pathway, including but not limited to L6 myoblasts,
B-ALL cells, B-cells, T-cells, leukemia cells, bone marrow cells, pi 90 transduced cells, philladelphia chromosome positive cells (Ph+), and mouse embryonic fibroblasts, are typically grown in cell growth media such as DMEM supplemented with fetal bovine serum and/or antibiotics, and grown to confluency.
[00489] In order to compare the effect of one or more compounds disclosed herein on Akt activation, said cells are serum starved overnight and incubated with one or more compounds disclosed herein or about 0.1% DMSO for approximately 1 minute to about 1 hour prior to stimulation with insulin (e.g. 100 nM) for about 1 minutes to about 1 hour. Cells are lysed by scraping into ice cold lysis buffer containing detergents such as sodium dodecyl sulfate and protease inhibitors (e.g., PMSF). After contacting cells with lysis buffer, the solution is briefly sonicated, cleared by centrifugation, resolved by SDS-PAGE, transferred to nitrocellulose or PVDF and immunoblotted using antibodies to phospho- Akt S473, phospho- Akt T308, Akt, and β-actin (Cell Signaling Technologies).
[00490] The results demonstrate that one or more compounds of the present disclosure inhibit insulin stimulated phosphorylation of Akt at S473. Alternatively, some compounds disclosed herein additionally inhibit insulin stimulated phosphorylation of Akt at T308. Such class of compounds can inhibit Akt more effectively than rapamycin and may be indicative of mTORC2 inhibitors or inhibitors of upstream kinases such as PI3K or Akt.
[00491] Example 23: Kinase Signaling in Blood
[00492] PI3K/ Akt /mTor signaling is measured in blood cells using the phosflow method (Methods
Enzymol. 2007;434: 131-54). The advantage of this method is that it is by nature a single cell assay so that cellular heterogeneity can be detected rather than population averages. This allows concurrent dinstinction of signaling states in different populations defined by other markers. Phosflow is also highly quantitative. To test the effects of one or more compounds disclosed herein, unfractionated splenocytes, or peripheral blood mononuclear cells are stimulated with anti-CD3 to initiate T-cell receptor signaling. The cells are then fixed and stained for surface markers and intracellular phosphoproteins. It is expected that inhibitors disclosed herein inhibit anti-CD3 mediated phosphorylation of Akt -S473 and S6, whereas rapamycin inhibits S6 phosphorylation and enhances Akt phosphorylation under the conditions tested.
[00493] Similarly, aliquots of whole blood are incubated for 15 minutes with vehicle (e.g. 0.1%DMSO) or kinase inhibitors at various concentrations, before addition of stimuli to crosslink the T cell receptor (TCR) (anti-CD3 with secondary antibody) or the B cell receptor (BCR) using anti-kappa light chain antibody (Fab '2 fragments). After approximately 5 and 15 minutes, samples are fixed (e.g. with cold 4% paraformaldehyde) and used for phosflow. Surface staining is used to distinguish T and B cells using antibodies directed to cell surface markers that are known to the art. The level of phosphrylation of kinase substrates such as Akt and S6 are then measured by incubating the fixed cells with labeled antibodies specific to the phosphorylated isoforms of these proteins. The population of cells are then analyzed by flow cytometry.
[00494] Example 24: Colony Formation Assay
[00495] Murine bone marrow cells freshly transformed with a pi 90 BCR-AbI retrovirus (herein referred to as p 190 transduced cells) are plated in the presence of various drug combinations in M3630 methylcellulose media for about 7 days with recombinant human IL-7 in about 30% serum, and the number of colonies formed is counted by visual examination under a microscope.
[00496] Alternatively, human peripheral blood mononuclear cells are obtained from Philadelphia chromosome positive (Ph+) and negative (Ph-) patients upon initial diagnosis or relapse. Live cells are isolated and enriched for CD 19+ CD34+ B cell progenitors. After overnight liquid culture, cells are plated in methocult GF+ H4435, Stem Cell Tehcnologies) suplemented with cytokines (IL-3, IL-6, IL-7, G-CSF, GM- CSF, CF, Flt3 ligand, and erythropoietin) and various concentrations of known chemotherapeutic agents in combination with either compounds of the present disclosure. Colonies are counted by microscopy 12-14 days later. This method can be used to test for evidence of additive or synergistic activity.
[00497] Example 25: In Vivo Effect of Kinase Inhibitors on Leukemic Cells
[00498] Female recipient mice are lethally irradiated from a γ source in two doses about 4 hr apart, with approximately 5Gy each. About lhr after the second radiation dose, mice are injected i.v. with about IxIO6 leukemic cells (e.g. Ph+ human or murine cells, or pi 90 transduced bone marrow cells). These cells are administered together with a radioprotective dose of about 5x10* normal bone marrow cells from 3-5 week old donor mice. Recipients are given antibiotics in the water and monitored daily. Mice who become sick after about 14 days are euthanized and lymphoid organs are harvested for analysis. Kinase inhibitor treatment begins about 10 days after leukemic cell injection and continues daily until the mice become sick or a maximum of approximately 35 days post-transplant. Inhibitors are given by oral lavage.
[00499] Peripheral blood cells are collected approximately on day 10 (pre-treatment) and upon euthanization
(post treatment), contacted with labled anti-hCD4 antibodies and counted by flow cytometry. This method can be used to demonstrate that the synergistic effect of one or more compouns disclosed herein in combination with known chemotherapeutic agents significantly reduce leukemic blood cell counts as compared to treatment with known chemotherapeutic agents (e.g. Gleevac) alone under the conditions tested.
[00500] Example 26: Treatment of Lupus Disease Model Mice [00501] Mice lacking the inhibitory receptor FcγRIIb that opposes PDK signaling in B cells develop lupus with high penetrance. FcγRIIb knockout mice (R2KO, Jackson Labs) are considered a valid model of the human disease as some lupus patients show decreased expression or function of FcγRIIb (S. Bolland and J. V. Ravtech 2000. Immunity 12:277-285).
[00502] The R2KO mice develop lupus-like disease with anti-nuclear antibodies, glomerulonephritis and proteinurea within about 4-6 months of age. For these experiments, the rapamycin analogue RADOOl (available from LC Laboratories) is used as a benchmark compound, and administered orally. This compound has been shown to ameliorate lupus symptoms in the B6.Slelz.Sle3z model (T. Wu et al. J. Clin Invest. 117:2186-2196).
[00503] Lupus disease model mice such as R2KO, BXSB or MLR/lpr are treated at about 2 months old, approximately for about two months. Mice are given doses of: vehicle, RADOOl at about 10mg/kg, or compounds disclosed herein at approximately 1 mg/kg to about 500 mg/kg. Blood and urine samples are obtained at approximately throughout the testing period, and tested for antinuclear antibodies (in dilutions of serum) or protein concentration (in urine). Serum is also tested for anti-ssDNA and anti-dsDNA antibodies by ELISA. Animals are euthanized at day 60 and tissues harvested for measuring spleen weight and kidney disease. Glomerulonephritis is assessed in kidney sections stained with H&E. Other animals are studied for about two months after cessation of treatment, using the same endpoints.
[00504] This model established in the art can be employed to demonstrate that the kinase inhibitors disclosed herein can suppress or delay the onset of lupus symptoms in lupus disease model mice.
[00505] Example 27: Murine Bone Marrow Transplant Assay
[00506] Female recipient mice are lethally irradiated from a γ ray source. About lhr after the radiation dose, mice are injected with about 1x106 leukemic cells from early passage pl90 transduced cultures (e.g. as described in Cancer Genet Cytogenet. 2005 Aug;161(l):51-6) . These cells are administered together with a radioprotective dose of approximately 5x106 normal bone marrow cells from 3-5 wk old donor mice. Recipients are given antibiotics in the water and monitored daily. Mice who become sick after about 14 days are euthanized and lymphoid organs harvested for flow cytometry and/or magnetic enrichment. Treatment begins on approximately day 10 and continues daily until mice become sick, or after a maximum of about 35 days post-transplant. Drugs are given by oral gavage (p.o.). In a pilot experiment a dose of chemotherapeutic that is not curative but delays leukemia onset by about one week or less is identified; controls are vehicle- treated or treated with chemotherapeutic agent, previously shown to delay but not cure leukemogenesis in this model (e.g. imatinib at about 70mg/kg twice daily). For the first phase pi 90 cells that express eGFP are used, and postmortem analysis is limited to enumeration of the percentage of leukemic cells in bone marrow, spleen and lymph node (LN) by flow cytometry. In the second phase, pi 90 cells that express a tailless form of human CD4 are used and the postmortem analysis includes magnetic sorting of hCD4+ cells from spleen followed by immunoblot analysis of key signaling endpoints: p Akt -T3O8 and S473; pS6 and p4EBP-l. As controls for immunoblot detection, sorted cells are incubated in the presence or absence of kinase inhibitors of the present disclosure inhibitors before lysis. Optionally, "phosfiow" is used to detect p Akt -S473 and pS6-S235/236 in hCD4-gated cells without prior sorting. These signaling studies are particularly useful if, for example, drug- treated mice have not developed clinical leukemia at the 35 day time point. Kaplan-Meier plots of survival are generated and statistical analysis done according to methods known in the art. Results from pi 90 cells are analyzed separated as well as cumulatively. [00507] Samples of peripheral blood (100-200μl) are obtained weekly from all mice, starting on day 10 immediately prior to commencing treatment. Plasma is used for measuring drug concentrations, and cells are analyzed for leukemia markers (eGFP or hCD4) and signaling biomarkers as described herein.
[00508] This general assay known in the art may be used to demonstrate that effective therapeutic doses of the compounds disclosed herein can be used for inhibiting the proliferation of leukemic cells.

Claims

CLAIMSWHAT IS CLAIMED IS:
1. A compound having a structure of one of the following formulae:
Figure imgf000166_0001
or a pharmaceutically acceptable salt thereof, wherein:
X5 and X6 are C-R6, N, C- L1 -R1, or N- L'-R1 wherein one of X5 and X6 is C- L'-R1 or N- L'-R1; XI is C or N; and X2 and X8 are independently N or C-R6 ; X3 and X7 are C or N, and at least one of X3 and X7 is C;
X4 in Formula I-A or I-C is C or N; and X4 in Formula I-B or I-D is C-R6, NH, or N; no more than two adjacent ring atoms are N or NH;
R1 is hydrogen, halogen, -CN, -OR10, -S(O)nR11, -NR12R13, -C(O)R14, alkyl, alkenyl, alkynyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, and n is an integer from 0 to 2; L1 is a bond, alkylene, heteroalkylene, alkenylene, alkynylene, cycloalkylene, heterocycloalkylene, arylene, or heteroarylene;
R2 and R3 are independently hydrogen, halogen, -CN, -OR10, -S(O)nR11, -NR12R13, -C(O)R14, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;
R4 and R5 are independently hydrogen or R6, or R4 and R5 are taken together to form a 5, 6 or 7 membered ring, wherein the 5, 6, or 7 membered ring is optionally substituted with (R6)q;
L is -NH-CR7R8-, - (CR7R8)Z- -C=O-, or -CR7R8(C=O) -, -O-, -SO-, or -SO2-; z is an integer from O to 10; q is an integer from O to 5; each ofR6 is independently hydrogen, halogen, -CN, -OR10, -S(O)nR11, -NR12R13, -C(O)R14, alkyl, alkenyl, alkynyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; each of R7 and R8 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocyclyalkyl, or R7 and R8 taken together form a cycloalkyl, heterocycloalkyl or aryl ring;
R9 is hydrogen, halogen, -CN, -OR10, -S(O)nR11, -NR12R13, -C(O)R14, alkyl, alkenyl, alkynyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; each R10 is independently hydrogen, -C(O)R15, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; each R11 is independently -NR16R17, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; each R12 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; each R13 is independently hydrogen, -S(O)nR18, -C(O)R19, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; or R12 and R13 are taken together to form a 5, 6, 7, or 8 membered ring, wherein the 5, 6, 7, or 8 membered ring contains O, 1, 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom Of-NR12R13; and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted; each R14 is independently -NR20R21, hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; each R15 is independently -NR22R23, hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; each of R16 and R17 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; or R16 and R17 are taken together to form a 5, 6, 7, or 8 membered ring, wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom Of -NR16R17; and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted; each R18 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; each R19 is independently -NR24R25, hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; each of R20 and R21 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, or R20 and R21 are taken together to form a 5, 6, 7, or 8 membered ring, wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom of -NR 0R21; and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted; each R22 and R23 are independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, or R22 and R23 are taken together to form a 5, 6, 7, or 8 membered ring, wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom of -NR22R23; and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted; each R24 and R25 are independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, or R24 and R25 are taken together to form a 5, 6, 7, or 8 membered ring, wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom Of-NR24R25; and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted; and W is CR6 or N.
2. The compound of claim 1, wherein when L is - (CR7R8)Z- and z is 1, then R1 is alkynyl, aryl, or heteroaryl.
3. The compound of claim 1, wherein when L is - (CR7R8) z - and z is 1, then L1 is a bond, and R1 is alkynyl, aryl, or heteroaryl.
4. The compound of claim 2 or 3 having a structure of the formula:
Figure imgf000168_0001
5. The compound of claim 2 or 3 having a structure of the formula:
Figure imgf000168_0002
6. The compound of claim 2 or 3, wherein R9 is -OR10, -NR12R13, -C(O)R14, alkyl, alkenyl, alkynyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl.
7. The compound of claim 6, wherein when R9 is aryl, then R9 is substituted with halogen, alkyl or heteroalkyl.
8. The compound of claim 2 or 3, wherein when R9 is -NR12R13, then R12 and R13 are taken together to form a 5, 6, 7 or 8 membered ring.
9. The compound of claim 2 or 3, wherein R1 is bicyclic heteroaryl.
10. The compound of claim 2 or 3, wherein R4 and R5 are taken together to form a 6-membered ring..
11. The compound of claim 2 or 3, wherein R2 is -NH2.
12. The compound of claim 2 or 3, wherein R7 is alkyl.
13. The compound of claim 2 or 3, wherein R7 and R8 are hydrogen.
14. The compound of claim 2 or 3, wherein W is CH.
15. The compound of claim 2 or 3, wherein R3 is hydrogen.
16. The compound of claim 2 or 3, wherein the compound has a structure of one of the following formulae:
Figure imgf000169_0001
17. A compound having a structure of one of the following formulae:
Figure imgf000169_0002
or a pharmaceutically acceptable salt thereof, wherein:
X5 and X6 are C-R6, N, C- L1 -R1, or N- L1 -R1 wherein one and no more than one of X5 and X6 is C- L1- R' or N- L'-R1;
XI is C or N; and X2 and X8 are independently N, or C-R6; X3 and X7 are C or N, at least one of X3 and X7 is C;
X4 in Formula V-A and Formula V-C is C or N; and X4 in Formula V-B and Formula V-D is C-R6, NH, or N; and wherein no more than two adjacent ring atoms are N or NH;
R1 is hydrogen, halogen, -CN, -OR10, -S(O)nR11, -NR12R13, -C(O)R14, alkyl, alkenyl, alkynyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, and n is an integer from 0 to 2;
L1 is a bond, alkylene, heteroalkylene, alkenylene, alkynylene, cycloalkylene, heterocycloalkylene, arylene, or heteroarylene; each of R2 and R3 is independently hydrogen, halogen, -CN, -OR10, -S(O)nR11, -NR12R13, - C(O)R14, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; each of R4 and R5 is independently hydrogen or R6, or R4 and R5 are taken together to form a 5, 6 or 7 membered ring, wherein the 5, 6, or 7 membered ring is optionally substituted with (R6)q;
L is -NH-CR7R8-, - (CR7R8)Z-, -C=O-, or -CR7R8(C=O) -, -O-, -SO-, or -SO2-; z is an integer from O to 10; q is an integer from O to 5; each R6 is independently hydrogen, halogen, -CN, -OR10, -S(O)nR11, -NR12R13, -C(O)R14, alkyl, alkenyl, alkynyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, wherein n is independently an integer from O to 2; each of R7 and R8 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocyclyalkyl, or R7 and R8 taken together form a cycloalkyl, heterocycloalkyl or aryl ring;
R9 is hydrogen, -C(O)R14, alkyl, alkenyl, alkynyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; each R10 is independently hydrogen, -C(O)R15, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; each R11 is independently -NR16R17, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; each R12 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; each R13 is independently hydrogen, -S(O)nR18, -C(O)R19, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; or R12 and R13 are taken together to form a 5, 6, 7, or 8 membered ring, wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom Of-NR12R13, and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted; each R14 is independently -NR20R21, hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; each R15 is independently -NR22R23, hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; each of R16 and R17 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; or R16 and R17 are taken together to form a 5, 6, 7 or 8 membered ring, wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom Of-NR16R17, and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted; each R18 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; each R19 is independently -NR24R25, hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; each of R20 and R21 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, or R20 and R21 are taken together to form a 5, 6, 7, or 8 membered ring, wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom Of-NR20R21, and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted; each of R22 and R23 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, or R22 and R23 are taken together to form a 5, 6, 7, or 8 membered ring, wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom Of-NR22R23, and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted; and each of R24 and R25 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, or R24 and R25 are taken together to form a 5, 6, 7, or 8 membered ring wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom Of-NR24R25, and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted.
18. The compound of claim 17, having a structure of the formula:
Figure imgf000171_0001
19. A compound having a structure of one of the following formulae:
Figure imgf000171_0002
or a pharmaceutically acceptable salt thereof, wherein:
X5 and X6 are C-R6, N, C- iΛR1, or N- iΛR1 wherein one and no more than one of X5 and X6 is C- L1 -R1 or N- L1 -R1; Xi is C or N; and X2 and X8 are independently N, or C-R6; X3 and X7 are C or N, at least one of X3 and X7 is C;
X4 in Formula VII-A and Formula VII-C is C or N; and X4 in Formula VII-B and Formula VII-D is C- R6, NH, or N; and wherein no more than two adjacent ring atoms are N or NH; R1 is hydrogen, halogen, -CN, -OR10, -S(O)nR11, -NR12R13, -C(O)R14, alkyl, alkenyl, alkynyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, and n is an integer from O to 2;
L1 is a bond, alkylene, heteroalkylene, alkenylene, alkynylene, cycloalkylene, heterocycloalkylene, arylene, or heteroarylene; each ofR2 and R3 is independently hydrogen, halogen, -CN, -OR10, -S(O)nR11, -NR12R13, - C(O)R14, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; each of R4 and R5 is independently hydrogen or R6, or R4 and R5 are taken together to form a 5, 6 or 7 membered ring, wherein the 5, 6, 7, or 8 membered ring is optionally substituted with (R6)q;
L is -NH-CR7R8-, - (CR7RV, -C=O-, or -CR7R8(C=O) -, -0-, -SO-, or -SO2-; z is an integer from O to 10; q is an integer from O to 5; each R6 is independently hydrogen, halogen, -CN, -OR10, -S(O)nR11, -NR12R13, -C(O)R14, alkyl, alkenyl, alkynyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, wherein n is independently an integer from 0 to 2; each of R7 and R8 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocyclyalkyl, or R7 and R8 taken together form a cycloalkyl, heterocycloalkyl or aryl ring;
R9 is hydrogen, -C(O)R14, alkyl, alkenyl, alkynyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; each R10 is independently hydrogen, -C(O)R15, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; each R11 is independently -NR16R17, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; each R12 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; each R13 is independently hydrogen, -S(O)nR18, -C(O)R19, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; or R12 and R13 are taken together to form a 5, 6, 7, or 8 membered ring, wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom Of-NR12R13, and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted; each R14 is independently -NR20R21, hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; each R15 is independently -NR22R23, hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; each of R16 and R17 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; or R16 and R17 are taken together to form a 5, 6, 7, or 8 membered ring, wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 additional ring heteroatoms selected from N, S, or O in addition to the nitrogen atom Of-NR16R17, and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted; each R18 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; each R19 is independently -NR24R25, hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; each of R20 and R21 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, or R20 and R21 are taken together to form a 5, 6, 7, or 8 membered ring, wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 additional ring heteroatoms selected from N, S, or O in addition to the nitrogen atom Of-NR20R21, and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted; each of R22 and R23 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, or R22 and R23 are taken together to form a 5, 6, 7, or 8 membered ring, wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom Of-NR22R23, and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted; and each of R24 and R25 is independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, or R24 and R25 are taken together to form a 5, 6, 7, or 8 membered ring wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 ring heteroatoms selected from N, S, or O in addition to the nitrogen atom Of-NR24R25, wherein the 5, 6, 7, or 8 membered ring contains 0, 1, 2 or 3 additional ring heteroatoms selected from N, S, or O, and further wherein the 5, 6, 7, or 8 membered ring is optionally substituted.
20. The compound of claim 19 having a structure of the formula:
Figure imgf000173_0001
21. The compound of any one of the preceding claims wherein the compound selectively inhibits one or more members of type I phosphatidylinositol 3-kinases (PI3-kinase) relative to other members of type I PI3-kinase, ascertained by an in vitro kinase assay.
22. The compound of claim 21 wherein the compound selectively inhibits PI3 -kinase γ and PI3-kinase δ as compared to PI3-kinase β and PI3-kinase α.
23. The compound of claim 21 wherein the compound selectively inhibits PI3-kinase δ and PI3-kinase β as compared to PI3-kinase α and PI3-kinase γ.
24. A pharmaceutical composition comprising a compound of any one of claims 1 to 20 or a pharmaceutically acceptable salt of a compound of any one of claims 1 to 20 and a pharmaceutically acceptable excipient.
25. A method of treating a medical condition mediated by a type I-PI3 kinase, comprising administering to a subject in need thereof a therapeutically effective amount of a compound of any one of claims lto 20 or apharmaceutically acceptable salt of a compound of any one of claims lto 20.
26. The method of claim 25, wherein the medical condition is selected from the group consisting of hematologic malignancy, inflammation, autoimmune disease, rheumatoid arthritis, systemic lupus erythematosus, asthma, and cardiovascular disease.
27. The method of claim 25, wherein the medical condition is selected from the group consisting of acute myelogenous leukemia, chronic myelogenous leukemia, mastocytosis, chronic lymphocytic leukemia, multiple myeloma, and myelodysplastic syndrome.
28. The method of claim 25, further comprising administering an anti-cancer agent.
29. A method of inhibiting activity of a protein kinase and/or a lipid kinase present in a cell, comprising contacting said cell with an effective amount of a compound of any one of claims 1 to 20 or a pharmaceutically acceptable salt of a compound of any one of claims 1 to 20.
PCT/US2009/005380 2008-09-26 2009-09-28 Heterocyclic kinase inhibitors WO2010036380A1 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
JP2011529032A JP5731978B2 (en) 2008-09-26 2009-09-28 Heterocyclic kinase inhibitor
US13/121,157 US8703778B2 (en) 2008-09-26 2009-09-28 Heterocyclic kinase inhibitors
CA2738429A CA2738429C (en) 2008-09-26 2009-09-28 Heterocyclic kinase inhibitors
EP09816603.6A EP2346508B1 (en) 2008-09-26 2009-09-28 Heterocyclic kinase inhibitors
US14/222,500 US9296742B2 (en) 2008-09-26 2014-03-21 Heterocyclic kinase inhibitors
US15/050,029 US9790228B2 (en) 2008-09-26 2016-02-22 Heterocyclic kinase inhibitors

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US19431008P 2008-09-26 2008-09-26
US61/194,310 2008-09-26

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US13/121,157 A-371-Of-International US8703778B2 (en) 2008-09-26 2009-09-28 Heterocyclic kinase inhibitors
US14/222,500 Continuation US9296742B2 (en) 2008-09-26 2014-03-21 Heterocyclic kinase inhibitors

Publications (1)

Publication Number Publication Date
WO2010036380A1 true WO2010036380A1 (en) 2010-04-01

Family

ID=42060027

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2009/005380 WO2010036380A1 (en) 2008-09-26 2009-09-28 Heterocyclic kinase inhibitors

Country Status (5)

Country Link
US (3) US8703778B2 (en)
EP (1) EP2346508B1 (en)
JP (1) JP5731978B2 (en)
CA (1) CA2738429C (en)
WO (1) WO2010036380A1 (en)

Cited By (144)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011058109A1 (en) 2009-11-12 2011-05-19 Ucb Pharma S.A. Fused bicyclic pyrrole and imidazole derivatives as kinase inhibitors
WO2011058111A1 (en) 2009-11-12 2011-05-19 Ucb Pharma S.A. Aminopurine derivatives as kinase inhibitors
WO2011058112A1 (en) 2009-11-12 2011-05-19 Ucb Pharma S.A. Fused bicyclic pyrazole derivatives as kinase inhibitors
WO2011058113A1 (en) 2009-11-12 2011-05-19 Ucb Pharma S.A. Fused bicyclic pyridine and pyrazine derivatives as kinase inhibitors
EP2365750A1 (en) * 2008-11-03 2011-09-21 Intellikine, Inc. Benzoxazole kinase inhibitors and methods of use
WO2011146882A1 (en) 2010-05-21 2011-11-24 Intellikine, Inc. Chemical compounds, compositions and methods for kinase modulation
WO2011149937A1 (en) * 2010-05-24 2011-12-01 Intellikine, Inc. Heterocyclic compounds and uses thereof
WO2012003278A1 (en) * 2010-06-30 2012-01-05 Amgen Inc. Quinolines as p13k inhibitors
WO2012003262A1 (en) * 2010-07-01 2012-01-05 Amgen Inc. Heterocyclic compounds and their use as inhibitors of pi3k activity
WO2012003283A1 (en) * 2010-06-30 2012-01-05 Amgen Inc. Heterocyclic compounds and their use as inhibitors of pi3k activity
WO2012003271A1 (en) * 2010-07-02 2012-01-05 Amgen Inc. Heterocyclic compounds and their use as inhibitors of pi3k activity
WO2012037204A1 (en) * 2010-09-14 2012-03-22 Exelixis, Inc. Inhibitors of pi3k-delta and methods of their use and manufacture
US20120077815A1 (en) * 2009-02-13 2012-03-29 Ucb Pharma S.A. Fused Pyridine And Pyrazine Derivatives As Kinase Inhibitors
WO2012064973A2 (en) 2010-11-10 2012-05-18 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US8183258B2 (en) 2007-03-23 2012-05-22 Amgen Inc. Heterocyclic compounds and their uses
US8193182B2 (en) 2008-01-04 2012-06-05 Intellikine, Inc. Substituted isoquinolin-1(2H)-ones, and methods of use thereof
US8193199B2 (en) 2007-03-23 2012-06-05 Amgen Inc. Heterocyclic compounds and their uses
WO2012087881A1 (en) * 2010-12-20 2012-06-28 Incyte Corporation N-(1-(substituted-phenyl)ethyl)-9h-purin-6-amines as pi3k inhibitors
WO2012097000A1 (en) 2011-01-10 2012-07-19 Pingda Ren Processes for preparing isoquinolinones and solid forms of isoquinolinones
US20120208799A1 (en) * 2009-10-19 2012-08-16 John King-Underwood Compounds
WO2012116237A2 (en) 2011-02-23 2012-08-30 Intellikine, Llc Heterocyclic compounds and uses thereof
WO2012148540A1 (en) * 2011-02-23 2012-11-01 Intellikine, Llc Combination of kanase inhibitors and uses threof
WO2012151562A1 (en) 2011-05-04 2012-11-08 Intellikine, Llc Combination pharmaceutical compositions and uses thereof
WO2013012918A1 (en) 2011-07-19 2013-01-24 Infinity Pharmaceuticals Inc. Heterocyclic compounds and uses thereof
WO2013012915A1 (en) 2011-07-19 2013-01-24 Infinity Pharmaceuticals Inc. Heterocyclic compounds and uses thereof
WO2013032591A1 (en) 2011-08-29 2013-03-07 Infinity Pharmaceuticals Inc. Heterocyclic compounds and uses thereof
WO2013033569A1 (en) * 2011-09-02 2013-03-07 Incyte Corporation Heterocyclylamines as pi3k inhibitors
WO2013049332A1 (en) 2011-09-29 2013-04-04 Infinity Pharmaceuticals, Inc. Inhibitors of monoacylglycerol lipase and methods of their use
WO2013045653A1 (en) 2011-09-30 2013-04-04 Oncodesign S.A. Macrocyclic flt3 kinase inhibitors
WO2013088404A1 (en) 2011-12-15 2013-06-20 Novartis Ag Use of inhibitors of the activity or function of PI3K
WO2013154878A1 (en) 2012-04-10 2013-10-17 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
JP2013539780A (en) * 2010-10-18 2013-10-28 レスピバート・リミテツド Quinazolin-4 (3H) -one derivatives for use as PI3 kinase inhibitors
WO2014011568A1 (en) 2012-07-10 2014-01-16 Takeda Pharmaceutical Company Limited Azaindole derivatives which act as pi3k inhibitors
US8633313B2 (en) 2009-12-18 2014-01-21 Amgen Inc. Heterocyclic compounds and their uses
US8637542B2 (en) 2008-03-14 2014-01-28 Intellikine, Inc. Kinase inhibitors and methods of use
US8642604B2 (en) 2006-04-04 2014-02-04 The Regents Of The University Of California Substituted pyrazolo[3,2-d]pyrimidines as anti-cancer agents
US8697709B2 (en) 2008-10-16 2014-04-15 The Regents Of The University Of California Fused ring heteroaryl kinase inhibitors
US8703777B2 (en) 2008-01-04 2014-04-22 Intellikine Llc Certain chemical entities, compositions and methods
US8703778B2 (en) 2008-09-26 2014-04-22 Intellikine Llc Heterocyclic kinase inhibitors
WO2014071109A1 (en) 2012-11-01 2014-05-08 Infinity Pharmaceuticals, Inc. Treatment of cancers using pi3 kinase isoform modulators
US8729074B2 (en) 2009-03-20 2014-05-20 Amgen Inc. Inhibitors of PI3 kinase
US8785454B2 (en) 2009-05-07 2014-07-22 Intellikine Llc Heterocyclic compounds and uses thereof
WO2014134426A1 (en) * 2013-03-01 2014-09-04 Incyte Corporation USE OF PYRAZOLOPYRIMIDINE DERIVATIVES FOR THE TREATMENT OF PI3Kδ RELATED DISORDERS
US8828998B2 (en) 2012-06-25 2014-09-09 Infinity Pharmaceuticals, Inc. Treatment of lupus, fibrotic conditions, and inflammatory myopathies and other disorders using PI3 kinase inhibitors
WO2014144850A1 (en) 2013-03-15 2014-09-18 Genentech, Inc. Methods of treating cancer and preventing cancer drug resistance
WO2014151386A1 (en) 2013-03-15 2014-09-25 Infinity Pharmaceuticals, Inc. Salts and solid forms of isoquinolinones and composition comprising and methods of using the same
JP2014528451A (en) * 2011-10-04 2014-10-27 ギリアード カリストガ エルエルシー Novel quinoxaline inhibitor of PI3K
WO2014194254A1 (en) 2013-05-30 2014-12-04 Infinity Pharmaceuticals, Inc. Treatment of cancers using pi3 kinase isoform modulators
US8940752B2 (en) 2009-06-29 2015-01-27 Incyte Corporation Pyrimidinones as PI3K inhibitors
US8980899B2 (en) 2009-10-16 2015-03-17 The Regents Of The University Of California Methods of inhibiting Ire1
US8993580B2 (en) 2008-03-14 2015-03-31 Intellikine Llc Benzothiazole kinase inhibitors and methods of use
WO2015051241A1 (en) 2013-10-04 2015-04-09 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
WO2015051244A1 (en) 2013-10-04 2015-04-09 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
WO2015061204A1 (en) 2013-10-21 2015-04-30 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9062055B2 (en) 2010-06-21 2015-06-23 Incyte Corporation Fused pyrrole derivatives as PI3K inhibitors
WO2015106012A1 (en) 2014-01-09 2015-07-16 Takeda Pharmaceutical Company Limited Azaindole derivatives
WO2015106014A1 (en) 2014-01-09 2015-07-16 Takeda Pharmaceutical Company Limited Azaindole derivatives
US9085560B2 (en) 2009-08-17 2015-07-21 Intellikine, Inc. Heterocyclic compounds and uses thereof
US9096611B2 (en) 2008-07-08 2015-08-04 Intellikine Llc Kinase inhibitors and methods of use
US9108984B2 (en) 2011-03-14 2015-08-18 Incyte Corporation Substituted diamino-pyrimidine and diamino-pyridine derivatives as PI3K inhibitors
US9126948B2 (en) 2011-03-25 2015-09-08 Incyte Holdings Corporation Pyrimidine-4,6-diamine derivatives as PI3K inhibitors
WO2015138920A1 (en) 2014-03-14 2015-09-17 Novartis Ag Antibody molecules to lag-3 and uses thereof
WO2015143012A1 (en) 2014-03-19 2015-09-24 Infinity Pharmaceuticals, Inc. Heterocyclic compounds for use in the treatment of pi3k-gamma mediated disorders
US9156845B2 (en) 2012-06-29 2015-10-13 Pfizer Inc. 4-(substituted amino)-7H-pyrrolo[2,3-d] pyrimidines as LRRK2 inhibitors
WO2015160986A2 (en) 2014-04-16 2015-10-22 Infinity Pharmaceuticals, Inc. Combination therapies
WO2015160975A2 (en) 2014-04-16 2015-10-22 Infinity Pharmaceuticals, Inc. Combination therapies
WO2015168079A1 (en) 2014-04-29 2015-11-05 Infinity Pharmaceuticals, Inc. Pyrimidine or pyridine derivatives useful as pi3k inhibitors
US9193721B2 (en) 2010-04-14 2015-11-24 Incyte Holdings Corporation Fused derivatives as PI3Kδ inhibitors
WO2015191677A1 (en) * 2014-06-11 2015-12-17 Incyte Corporation Bicyclic heteroarylaminoalkyl phenyl derivatives as pi3k inhibitors
US9227977B2 (en) 2013-03-15 2016-01-05 Respivert Ltd. Phosphoinositide 3-kinase inhibitors
US9233111B2 (en) 2011-07-08 2016-01-12 Novartis Ag Pyrrolo pyrimidine derivatives
US9273058B2 (en) 2013-11-14 2016-03-01 Bristol-Myers Squibb Company Substituted pyrazolo-piperazines as casein kinase 1 δ/ε inhibitors
WO2016040880A1 (en) 2014-09-13 2016-03-17 Novartis Ag Combination therapies of alk inhibitors
WO2016054555A2 (en) 2014-10-03 2016-04-07 Novartis Ag Combination therapies
WO2016054491A1 (en) 2014-10-03 2016-04-07 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9309251B2 (en) 2012-04-02 2016-04-12 Incyte Holdings Corporation Bicyclic azaheterocyclobenzylamines as PI3K inhibitors
WO2016057841A1 (en) 2014-10-08 2016-04-14 Novartis Ag Compositions and methods of use for augmented immune response and cancer therapy
WO2016061142A1 (en) 2014-10-14 2016-04-21 Novartis Ag Antibody molecules to pd-l1 and uses thereof
US9321772B2 (en) 2011-09-02 2016-04-26 The Regents Of The University Of California Substituted pyrazolo[3,4-D]pyrimidines and uses thereof
US9359349B2 (en) 2007-10-04 2016-06-07 Intellikine Llc Substituted quinazolines as kinase inhibitors
WO2016100882A1 (en) 2014-12-19 2016-06-23 Novartis Ag Combination therapies
US9403847B2 (en) 2009-12-18 2016-08-02 Incyte Holdings Corporation Substituted heteroaryl fused derivatives as P13K inhibitors
US9481667B2 (en) 2013-03-15 2016-11-01 Infinity Pharmaceuticals, Inc. Salts and solid forms of isoquinolinones and composition comprising and methods of using the same
US9493450B2 (en) 2014-02-13 2016-11-15 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US9493442B2 (en) 2014-02-13 2016-11-15 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US9512125B2 (en) 2004-11-19 2016-12-06 The Regents Of The University Of California Substituted pyrazolo[3.4-D] pyrimidines as anti-inflammatory agents
US9527835B2 (en) 2014-02-13 2016-12-27 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
WO2017019894A1 (en) 2015-07-29 2017-02-02 Novartis Ag Combination therapies comprising antibody molecules to lag-3
WO2017019897A1 (en) 2015-07-29 2017-02-02 Novartis Ag Combination therapies comprising antibody molecules to tim-3
US9629843B2 (en) 2008-07-08 2017-04-25 The Regents Of The University Of California MTOR modulators and uses thereof
US9642799B2 (en) 2012-03-13 2017-05-09 Respivert, Ltd. Crystalline 6-(2-((4-amino-3-(3-hydroxyphenyl)-1H-pyrazolo[3,4-D]pyrimidin-1-yl)methyl)-3-(2-chlorobenzyl)-4-0X0-3,4-dihydroquinazolin-5-yl)-N,N-bis(2-methoxyethyl)hex-5-ynamide
WO2017079116A2 (en) 2015-11-03 2017-05-11 Janssen Biotech, Inc. Antibodies specifically binding pd-1 and tim-3 and their uses
US9670210B2 (en) 2014-02-13 2017-06-06 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
WO2017106656A1 (en) 2015-12-17 2017-06-22 Novartis Ag Antibody molecules to pd-1 and uses thereof
US9695171B2 (en) 2013-12-17 2017-07-04 Pfizer Inc. 3,4-disubstituted-1 H-pyrrolo[2,3-b]pyridines and 4,5-disubstituted-7H-pyrrolo[2,3-c]pyridazines as LRRK2 inhibitors
US9695167B2 (en) 2014-07-10 2017-07-04 Incyte Corporation Substituted triazolo[1,5-a]pyridines and triazolo[1,5-a]pyrazines as LSD1 inhibitors
US9695168B2 (en) 2014-07-10 2017-07-04 Incyte Corporation Substituted imidazo[1,5-α]pyridines and imidazo[1,5-α]pyrazines as LSD1 inhibitors
US9695180B2 (en) 2014-07-10 2017-07-04 Incyte Corporation Substituted imidazo[1,2-a]pyrazines as LSD1 inhibitors
US9724354B2 (en) 2013-03-22 2017-08-08 Millennium Pharmaceuticals, Inc. Combination of catalytic mTORC1/2 inhibitors and selective inhibitors of Aurora A kinase
US9732097B2 (en) 2015-05-11 2017-08-15 Incyte Corporation Process for the synthesis of a phosphoinositide 3-kinase inhibitor
US9745306B2 (en) 2013-03-15 2017-08-29 Respivert Limited 2-((4-amino-3-(3-fluoro-5-hydroxyphenyl)-1H-pyrazolo[3,4-D]pyrimidin-1-yl)methyl)-3-(2-(trifluoromethyl)benzyl) quinazolin-4(3H)-one derivatives and their use as phosphoinositide 3-kinase inhibitors
US9758523B2 (en) 2014-07-10 2017-09-12 Incyte Corporation Triazolopyridines and triazolopyrazines as LSD1 inhibitors
US9775841B2 (en) 2011-05-04 2017-10-03 Rhizen Pharmaceuticals Sa Compounds as modulators of protein kinases
WO2017214269A1 (en) 2016-06-08 2017-12-14 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
WO2017223422A1 (en) 2016-06-24 2017-12-28 Infinity Pharmaceuticals, Inc. Combination therapies
US9873704B2 (en) 2009-06-25 2018-01-23 Amgen Inc. Heterocyclic compounds and their uses
US9944639B2 (en) 2014-07-04 2018-04-17 Lupin Limited Quinolizinone derivatives as PI3K inhibitors
US9944647B2 (en) 2015-04-03 2018-04-17 Incyte Corporation Heterocyclic compounds as LSD1 inhibitors
US9988401B2 (en) 2015-05-11 2018-06-05 Incyte Corporation Crystalline forms of a PI3K inhibitor
US10039753B2 (en) 2015-09-14 2018-08-07 Pfizer Inc. Imidazo[4,5-c]quinoline and imidazo[4,5-c][1,5]naphthyridine derivatives as LRRK2 inhibitors
US10117945B2 (en) 2014-09-11 2018-11-06 The Regents Of The University Of California mTORC1 inhibitors
US10131668B2 (en) 2012-09-26 2018-11-20 The Regents Of The University Of California Substituted imidazo[1,5-a]pYRAZINES for modulation of IRE1
EP3409669A1 (en) 2014-06-19 2018-12-05 ARIAD Pharmaceuticals, Inc. Heteroaryl compounds for kinase inhibition
US10160761B2 (en) 2015-09-14 2018-12-25 Infinity Pharmaceuticals, Inc. Solid forms of isoquinolinones, and process of making, composition comprising, and methods of using the same
WO2018237157A1 (en) 2017-06-22 2018-12-27 Novartis Ag Antibody molecules to cd73 and uses thereof
WO2018237173A1 (en) 2017-06-22 2018-12-27 Novartis Ag Antibody molecules to cd73 and uses thereof
US10166221B2 (en) 2016-04-22 2019-01-01 Incyte Corporation Formulations of an LSD1 inhibitor
US10214519B2 (en) 2016-09-23 2019-02-26 Gilead Sciences, Inc. Phosphatidylinositol 3-kinase inhibitors
US10227350B2 (en) 2016-09-23 2019-03-12 Gilead Sciences, Inc. Phosphatidylinositol 3-kinase inhibitors
US10329255B2 (en) 2015-08-12 2019-06-25 Incyte Corporation Salts of an LSD1 inhibitor
US10336759B2 (en) 2015-02-27 2019-07-02 Incyte Corporation Salts and processes of preparing a PI3K inhibitor
EP3514179A1 (en) 2014-01-24 2019-07-24 Dana-Farber Cancer Institute, Inc. Antibody molecules to pd-1 and uses thereof
US10479770B2 (en) 2016-09-23 2019-11-19 Gilead Sciences, Inc. Phosphatidylinositol 3-kinase inhibitors
WO2019224718A2 (en) 2018-05-24 2019-11-28 Janssen Biotech, Inc. Psma binding agents and uses thereof
WO2019232244A2 (en) 2018-05-31 2019-12-05 Novartis Ag Antibody molecules to cd73 and uses thereof
WO2019229658A1 (en) 2018-05-30 2019-12-05 Novartis Ag Entpd2 antibodies, combination therapies, and methods of using the antibodies and combination therapies
US10759806B2 (en) 2016-03-17 2020-09-01 Infinity Pharmaceuticals, Inc. Isotopologues of isoquinolinone and quinazolinone compounds and uses thereof as PI3K kinase inhibitors
US10858359B2 (en) 2016-06-07 2020-12-08 Jacobio Pharmaceuticals Co., Ltd. Heterocyclic ring derivatives useful as SHP2 inhibitors
WO2021053559A1 (en) 2019-09-18 2021-03-25 Novartis Ag Entpd2 antibodies, combination therapies, and methods of using the antibodies and combination therapies
US10968200B2 (en) 2018-08-31 2021-04-06 Incyte Corporation Salts of an LSD1 inhibitor and processes for preparing the same
US10980889B1 (en) 2018-05-01 2021-04-20 Revolution Medicines, Inc. C40-, C28-, and C-32-linked rapamycin analogs as mTOR inhibitors
US10988466B2 (en) 2017-03-23 2021-04-27 Jacobio Pharmaceuticals Co., Ltd. Heterocyclic derivatives useful as SHP2 inhibitors
US11111247B2 (en) 2018-09-25 2021-09-07 Incyte Corporation Pyrazolopyrimidine compounds and uses thereof
RU2760669C2 (en) * 2017-04-13 2021-11-29 Кэнсер Рисерч Текнолоджи Лимитед Inhibitory compounds
WO2022162518A2 (en) 2021-01-28 2022-08-04 Janssen Biotech, Inc. Psma binding proteins and uses thereof
US11548896B2 (en) 2016-04-15 2023-01-10 Cancer Research Technology Limited Heterocyclic compounds as RET kinase inhibitors
WO2023016477A1 (en) * 2021-08-11 2023-02-16 Taizhou Eoc Pharma Co., Ltd. A cyclin-dependent kinase inhibitor
US11591395B2 (en) 2019-04-19 2023-02-28 Janssen Biotech, Inc. Methods of treating prostate cancer with an anti-PSMA/CD3 antibody
US11661423B2 (en) 2016-04-15 2023-05-30 Cancer Research Technology Limited Heterocyclic compounds as RET kinase inhibitors
US11685749B2 (en) 2018-05-01 2023-06-27 Revolution Medicines, Inc. C26-linked rapamycin analogs as mTOR inhibitors
WO2023186065A1 (en) * 2022-04-02 2023-10-05 Taizhou Eoc Pharma Co., Ltd. A cyclin-dependent kinase inhibitor
EP4324518A2 (en) 2014-01-31 2024-02-21 Novartis AG Antibody molecules to tim-3 and uses thereof
TWI841376B (en) 2013-03-01 2024-05-01 美商英塞特控股公司 USE OF PYRAZOLOPYRIMIDINE DERIVATIVES FOR THE TREATMENT OF PI3Kδ RELATED DISORDERS
EP4378957A2 (en) 2015-07-29 2024-06-05 Novartis AG Combination therapies comprising antibody molecules to pd-1

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105062961A (en) 2012-05-23 2015-11-18 弗·哈夫曼-拉罗切有限公司 Compositions and methods of obtaining and using endoderm and hepatocyte cells
WO2014026329A1 (en) * 2012-08-15 2014-02-20 Merck Sharp & Dohme Corp. N-alkylated indole and indazole compounds as rorgammat inhibitors and uses thereof
EP2920181B1 (en) 2012-11-16 2019-01-09 University Health Network Pyrazolopyrimidine compounds
ES2685568T3 (en) 2012-12-21 2018-10-10 Gilead Calistoga Llc Inhibitors of isoquinolinone or quinazolinone phosphatidylinositol 3-kinase
PT2941426T (en) 2012-12-21 2018-07-18 Gilead Calistoga Llc Substituted pyrimidine aminoalkyl-quinazolones as phosphatidylinositol 3-kinase inhibitors
SI3008053T1 (en) 2013-06-14 2018-06-29 Gilead Calistoga Llc Phosphatidylinositol 3-kinase inhibitors
US10980755B2 (en) 2015-09-10 2021-04-20 The Regents Of The University Of California LRH-1 modulators
MX2021001186A (en) 2015-11-20 2022-10-11 Forma Therapeutics Inc Purinones as ubiquitin-specific protease 1 inhibitors.

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060235031A1 (en) * 2004-04-02 2006-10-19 Arnold Lee D 6,6-Bicyclic ring substituted heterobicyclic protein kinase inhibitors
US20070054915A1 (en) * 2005-08-25 2007-03-08 Roche Palo Alto Llc p38 MAP kinase inhibitors and methods for using the same

Family Cites Families (364)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB812366A (en) 1955-08-18 1959-04-22 Wellcome Found Improvements in and relating to derivatives of pyrimidine and the preparation thereof
GB937725A (en) 1960-05-11 1963-09-25 Ciba Ltd Pyrazolo[3:4-d]pyrimidines
DE2139107A1 (en) 1971-08-04 1973-02-15 Merck Patent Gmbh N(6)-subst adenosine derivs - with coronary circulatory etc activity
IT1153216B (en) 1981-10-16 1987-01-14 Schering Ag PROCEDURE FOR THE PREPARATION OF CYANETHEROCYCLIC COMPOUNDS
DE3244594A1 (en) 1982-12-02 1984-06-07 Hoechst Ag, 6230 Frankfurt 1-PHENYLISOCHINOLINE DERIVATIVES AND METHOD FOR THE PRODUCTION THEREOF, PHARMACEUTICAL PREPARATIONS CONTAINING THIS COMPOUND AND THE USE THEREOF
DE3406533A1 (en) 1984-02-23 1985-08-29 Boehringer Mannheim Gmbh, 6800 Mannheim USE OF ADENOSINE DERIVATIVES AS ANTIALLERGICA AND MEDICINAL PRODUCTS CONTAINING THEM
US5310731A (en) 1984-06-28 1994-05-10 Whitby Research, Inc. N-6 substituted-5'-(N-substitutedcarboxamido)adenosines as cardiac vasodilators and antihypertensive agents
US4795627A (en) 1984-10-18 1989-01-03 University Of Pittsburgh Tritium labelled N-mustard type compounds and a process for their production
US4656159A (en) 1984-10-31 1987-04-07 Georgetown University Galactose-C-6 nitrogen mustard compounds and their uses
JPS61109797A (en) 1984-11-01 1986-05-28 Yuki Gosei Yakuhin Kogyo Kk Labeled nucleotide and labeled polynucleotide
WO1990003370A1 (en) 1988-09-28 1990-04-05 Microprobe Corporation DERIVATIVES OF PYRAZOLO[3,4-d]PYRIMIDINE
US5442039A (en) 1989-07-17 1995-08-15 The Dow Chemical Company Mesogenic polycyanates and thermosets thereof
US5428125A (en) 1989-07-17 1995-06-27 The Dow Chemical Company Mesogenic polycyanates and thermosets thereof
US5646128A (en) 1989-09-15 1997-07-08 Gensia, Inc. Methods for treating adenosine kinase related conditions
US5763596A (en) 1989-09-15 1998-06-09 Metabasis Therapeutics, Inc. C-4' modified adenosine kinase inhibitors
US5674998A (en) 1989-09-15 1997-10-07 Gensia Inc. C-4' modified adenosine kinase inhibitors
US5763597A (en) 1989-09-15 1998-06-09 Metabasis Therapeutics, Inc. Orally active adenosine kinase inhibitors
US5795977A (en) 1989-09-15 1998-08-18 Metabasis Therapeutics, Inc. Water soluble adenosine kinase inhibitors
US5721356A (en) 1989-09-15 1998-02-24 Gensia, Inc. Orally active adenosine kinase inhibitors
GB9009542D0 (en) 1990-04-27 1990-06-20 Beecham Group Plc Novel compounds
GB9113137D0 (en) 1990-07-13 1991-08-07 Ici Plc Thioxo heterocycles
DE4026265A1 (en) 1990-08-20 1992-02-27 Boehringer Mannheim Gmbh NEW PHOSPHOLIPID DERIVATIVES OF NUCLEOSIDES, THEIR PRODUCTION AND THEIR USE AS ANTIVIRAL MEDICINAL PRODUCTS
US5563257A (en) 1990-08-20 1996-10-08 Boehringer Mannheim Gmbh Phospholipid derivatives of nucleosides
US5652366A (en) 1990-09-25 1997-07-29 Rhone-Poulenc Rorer Pharmaceuticals Inc. DI (1R)-(-)camphosulfonic acid) salt, preparation thereof and use thereof
AU654507B2 (en) 1990-09-25 1994-11-10 Rhone-Poulenc Rorer International (Holdings) Inc. Compounds having antihypertensive and anti-ischemic properties
US5561134A (en) 1990-09-25 1996-10-01 Rhone-Poulenc Rorer Pharmaceuticals Inc. Compounds having antihypertensive, cardioprotective, anti-ischemic and antilipolytic properties
GB9103839D0 (en) 1991-02-23 1991-04-10 Smithkline Beecham Plc Pharmaceuticals
US5916891A (en) 1992-01-13 1999-06-29 Smithkline Beecham Corporation Pyrimidinyl imidazoles
IL104369A0 (en) 1992-01-13 1993-05-13 Smithkline Beecham Corp Novel compounds and compositions
DE4204032A1 (en) 1992-02-12 1993-08-19 Boehringer Mannheim Gmbh NEW LIPONUCLEOTIDES, THEIR PRODUCTION AND THEIR USE AS ANTIVIRAL MEDICINAL PRODUCTS
DE4204031A1 (en) 1992-02-12 1993-08-19 Boehringer Mannheim Gmbh NEW LIPID PHOSPHONIC ACID-NUCLEOSIDE CONJUGATES AND THEIR USE AS ANTIVIRAL MEDICINAL PRODUCTS
WO1993018035A1 (en) 1992-03-04 1993-09-16 Abbott Laboratories Angiotensin ii receptor antagonists
JP2737518B2 (en) 1992-03-16 1998-04-08 富士通株式会社 Cooling structure of infrared detector
US5294612A (en) 1992-03-30 1994-03-15 Sterling Winthrop Inc. 6-heterocyclyl pyrazolo [3,4-d]pyrimidin-4-ones and compositions and method of use thereof
AU684461B2 (en) 1992-04-07 1997-12-18 Regents Of The University Of Michigan, The CD28 pathway immunoregulation
GB9208135D0 (en) 1992-04-13 1992-05-27 Ludwig Inst Cancer Res Polypeptides having kinase activity,their preparation and use
EP0672159B1 (en) 1992-04-24 2005-12-28 Sri International Homologous sequence targeting in eukaryotic cells
CA2117476C (en) 1992-06-19 2000-02-22 R. Andrew Wood Infrared camera with thermoelectric temperature stabilization
US6057305A (en) 1992-08-05 2000-05-02 Institute Of Organic Chemistry And Biochemistry Of The Academy Of Sciences Of The Czech Republic Antiretroviral enantiomeric nucleotide analogs
TW444018B (en) 1992-12-17 2001-07-01 Pfizer Pyrazolopyrimidines
IL108523A0 (en) 1993-02-03 1994-05-30 Gensia Inc Pharmaceutical compositions containing adenosine kinase inhibitors for preventing or treating conditions involving inflammatory responses and pain
JPH08506343A (en) 1993-02-03 1996-07-09 ジェンシア・インコーポレイテッド Adenosine kinase inhibitors containing lysofuranosyl derivatives
GB9308957D0 (en) 1993-04-30 1993-06-16 Cancer Res Campaign Tech Novel produgs
EP0700430B1 (en) 1993-06-04 2005-04-20 The United States of America as Represented by the Secretary of the Navy Methods for selectively stimulating proliferation of t cells
US5504103A (en) 1993-08-25 1996-04-02 Eli Lilly And Company Inhibition of phosphatidylinositol 3-kinase with 17 β-hydroxywortmannin and analogs thereof
US5525503A (en) 1993-09-28 1996-06-11 Dana-Farber Cancer Institute, Inc. Signal transduction via CD28
WO1995012588A1 (en) 1993-11-05 1995-05-11 Biochem Pharma Inc. Antineoplastic heteronaphthoquinones
IL112249A (en) 1994-01-25 2001-11-25 Warner Lambert Co Pharmaceutical compositions containing di and tricyclic pyrimidine derivatives for inhibiting tyrosine kinases of the epidermal growth factor receptor family and some new such compounds
US5654307A (en) 1994-01-25 1997-08-05 Warner-Lambert Company Bicyclic compounds capable of inhibiting tyrosine kinases of the epidermal growth factor receptor family
US6632789B1 (en) 1994-04-29 2003-10-14 The United States Of America As Represented By The Secretary Of The Navy Methods for modulating T cell responses by manipulating intracellular signal transduction
DE4418690A1 (en) 1994-05-28 1996-01-11 Boehringer Mannheim Gmbh Nucleoside 5'-mono-phosphate ester(s) of glyceryl di:ether derivs.
US6323201B1 (en) 1994-12-29 2001-11-27 The Regents Of The University Of California Compounds for inhibition of ceramide-mediated signal transduction
AU711592B2 (en) 1995-04-03 1999-10-14 Novartis Ag Pyrazole derivatives and processes for the preparation thereof
US6312894B1 (en) 1995-04-03 2001-11-06 Epoch Pharmaceuticals, Inc. Hybridization and mismatch discrimination using oligonucleotides conjugated to minor groove binders
US5977061A (en) 1995-04-21 1999-11-02 Institute Of Organic Chemistry And Biochemistry Of The Academy Of Sciences Of The Czech Republic N6 - substituted nucleotide analagues and their use
JPH08295667A (en) 1995-04-27 1996-11-12 Takeda Chem Ind Ltd Heterocyclic compound, its production and pharmaceutical preparation
WO1996037777A1 (en) 1995-05-23 1996-11-28 Nelson Randall W Mass spectrometric immunoassay
US5593997A (en) 1995-05-23 1997-01-14 Pfizer Inc. 4-aminopyrazolo(3-,4-D)pyrimidine and 4-aminopyrazolo-(3,4-D)pyridine tyrosine kinase inhibitors
US6403599B1 (en) 1995-11-08 2002-06-11 Pfizer Inc Corticotropin releasing factor antagonists
AU5982296A (en) 1995-06-07 1996-12-30 G.D. Searle & Co. Method to treat cardiofibrosis with a combination of an angi otensin ii antagonist and spironolactone
EP0831911B1 (en) 1995-06-07 2002-04-17 G.D. Searle & Co. Spironolactone and angiotensin ii antagonist combination therapy for treatment of congestive heart failure
PL185150B1 (en) 1995-06-07 2003-03-31 Searle & Co Combination therapy by means of epoxy-steroidifc aldosterone antagonist and antagonist of angiotensin ii while treating hypostatic hearth failures
US5665721A (en) 1995-06-07 1997-09-09 Abbott Laboratories Heterocyclic substituted cyclopentane compounds
US5763885A (en) 1995-12-19 1998-06-09 Loral Infrared & Imaging Systems, Inc. Method and apparatus for thermal gradient stabilization of microbolometer focal plane arrays
JPH09143163A (en) 1995-11-29 1997-06-03 Fuji Photo Film Co Ltd Production of nitrogen-containing heteroaromatic amides
US5747235A (en) 1996-01-26 1998-05-05 Eastman Kodak Company Silver halide light sensitive emulsion layer having enhanced photographic sensitivity
CH690773A5 (en) 1996-02-01 2001-01-15 Novartis Ag Pyrrolo (2,3-d) pyrimides and their use.
DE19603576A1 (en) 1996-02-01 1997-08-07 Bayer Ag Acylated 4-amino and 4-hydrazinopyrimidines
GB2310952B (en) 1996-03-05 1998-08-19 Mitsubishi Electric Corp Infrared detector
GB9611460D0 (en) 1996-06-01 1996-08-07 Ludwig Inst Cancer Res Novel lipid kinase
CA2258822A1 (en) 1996-06-20 1997-12-24 Sean Kerwin Compounds and methods for providing pharmacologically active preparations and uses thereof
US6153631A (en) 1996-10-23 2000-11-28 Zymogenetics, Inc. Compositions and methods for treating bone deficit conditions
US6342514B1 (en) 1996-10-23 2002-01-29 Zymogenetics, Inc. Compositions and methods for treating bone deficit conditions
US5965573A (en) 1996-10-23 1999-10-12 Zymogenetics, Inc. Compositions and methods for treating bone deficit conditions
US5990169A (en) 1996-10-23 1999-11-23 Zymogenetics, Inc. Compositions and methods for treating bone deficit conditions
US5922753A (en) 1996-10-23 1999-07-13 Zymogenetics, Inc. Methods for treating bone deficit conditions with benzothiazole
US5994358A (en) 1996-10-23 1999-11-30 Zymogenetics, Inc. Compositions and methods for treating bone deficit conditions
US5948776A (en) 1996-10-23 1999-09-07 Zymogenetic, Inc. Compositions and methods for treating bone deficit conditions
US5919808A (en) 1996-10-23 1999-07-06 Zymogenetics, Inc. Compositions and methods for treating bone deficit conditions
US6251901B1 (en) 1996-10-23 2001-06-26 Zymogenetics, Inc. Compositions and methods for treating bone deficit conditions
US5858753A (en) 1996-11-25 1999-01-12 Icos Corporation Lipid kinase
WO1998024804A2 (en) 1996-12-06 1998-06-11 Vertex Pharmaceuticals Incorporated INHIBITORS OF INTERLEUKIN-1β CONVERTING ENZYME
US6093737A (en) 1996-12-30 2000-07-25 Merck & Co., Inc. Inhibitors of farnesyl-protein transferase
JPH10206995A (en) 1997-01-21 1998-08-07 Konica Corp Silver halide photographic sensitive material
WO1998035048A2 (en) 1997-02-07 1998-08-13 Princeton University Engineered protein kinases which can utilize modified nucleotide triphosphate substrates
US7863444B2 (en) 1997-03-19 2011-01-04 Abbott Laboratories 4-aminopyrrolopyrimidines as kinase inhibitors
ATE242245T1 (en) 1997-03-19 2003-06-15 Basf Ag PYRROLO(2,3-D)PYRIMIDINES AND THEIR USE AS TYROSINE KINASE INHIBITORS
AU7449598A (en) 1997-05-23 1998-12-11 Nippon Shinyaku Co. Ltd. Medicinal composition for prevention or treatment of hepatopathy
US6207679B1 (en) 1997-06-19 2001-03-27 Sepracor, Inc. Antimicrobial agents uses and compositions related thereto
IL133800A0 (en) * 1997-07-03 2001-04-30 Du Pont Pharm Co Imidazopyrimidines and imidazopyridines for the treatment of neurological disorders
EP1020445B1 (en) 1997-10-02 2008-08-13 Eisai R&D Management Co., Ltd. Fused pyridine derivatives
US6649631B1 (en) 1997-10-23 2003-11-18 The Board Of Regents Of The University Of Texas System Compositions and methods for treating bone deficit conditions
CN1130363C (en) 1997-11-12 2003-12-10 三菱化学株式会社 Purine derivatives and medicine containing the same as the active ingredient
US6191170B1 (en) 1998-01-13 2001-02-20 Tularik Inc. Benzenesulfonamides and benzamides as therapeutic agents
US7715989B2 (en) 1998-04-03 2010-05-11 Elitech Holding B.V. Systems and methods for predicting oligonucleotide melting temperature (TmS)
US6127121A (en) 1998-04-03 2000-10-03 Epoch Pharmaceuticals, Inc. Oligonucleotides containing pyrazolo[3,4-D]pyrimidines for hybridization and mismatch discrimination
JP2000072773A (en) 1998-08-28 2000-03-07 Zeria Pharmaceut Co Ltd Purine derivative
CZ2001959A3 (en) 1998-09-18 2001-12-12 Basf Aktiengesellschaft 4-Aminopyrrolopyrimidines functioning as kinase inhibitors
EP1140938B1 (en) 1999-01-11 2003-08-27 Princeton University High affinity inhibitors for target validation and uses thereof
CZ27399A3 (en) 1999-01-26 2000-08-16 Ústav Experimentální Botaniky Av Čr Substituted nitrogen heterocyclic derivatives process of their preparation, the derivatives employed as medicaments, pharmaceutical composition and a compound pharmaceutical preparation in which these derivatives are comprised as well as use of these derivatives for preparing medicaments
EP1040831A3 (en) 1999-04-02 2003-05-02 Pfizer Products Inc. Use of corticotropin releasing factor (CRF) antagonists to prevent sudden death
SE515856C2 (en) 1999-05-19 2001-10-22 Ericsson Telefon Ab L M Carrier for electronic components
KR20020062808A (en) 1999-06-03 2002-07-31 크놀 게엠베하 Benzothiazinone and benzoxazinone compounds
US6387894B1 (en) 1999-06-11 2002-05-14 Pfizer Inc. Use of CRF antagonists and renin-angiotensin system inhibitors
TWI262914B (en) 1999-07-02 2006-10-01 Agouron Pharma Compounds and pharmaceutical compositions for inhibiting protein kinases
PE20010306A1 (en) 1999-07-02 2001-03-29 Agouron Pharma INDAZOLE COMPOUNDS AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM USEFUL FOR THE INHIBITION OF PROTEIN KINASE
DK1221444T3 (en) * 1999-07-02 2005-11-14 Eisai Co Ltd Condensed imidazole compounds and anti-diabetes mellitus drugs
GB9919588D0 (en) 1999-08-18 1999-10-20 Hoechst Schering Agrevo Gmbh Fungicidal compounds
JP4831906B2 (en) 1999-08-27 2011-12-07 ケモセントリックス, インコーポレイテッド Heterocyclic compounds and methods for modulating CXCR3 function
ATE286033T1 (en) 1999-09-16 2005-01-15 Curis Inc BROKERS OF IGEL SIGNAL PATHS, THEIR COMPOSITIONS AND USES
US6921763B2 (en) 1999-09-17 2005-07-26 Abbott Laboratories Pyrazolopyrimidines as therapeutic agents
CA2385747A1 (en) 1999-09-17 2001-03-22 Gavin C. Hirst Pyrazolopyrimidines as therapeutic agents
JP5036112B2 (en) 1999-10-06 2012-09-26 ベーリンガー インゲルハイム ファーマシューティカルズ インコーポレイテッド Heterocyclic compounds useful as inhibitors of tyrosine kinases
US6506769B2 (en) 1999-10-06 2003-01-14 Boehringer Ingelheim Pharmaceuticals, Inc. Heterocyclic compounds useful as inhibitors of tyrosine kinases
US6472153B1 (en) 1999-10-26 2002-10-29 Epoch Biosciences, Inc. Hybridization-triggered fluorescent detection of nucleic acids
US6660845B1 (en) 1999-11-23 2003-12-09 Epoch Biosciences, Inc. Non-aggregating, non-quenching oligomers comprising nucleotide analogues; methods of synthesis and use thereof
GB0002032D0 (en) 2000-01-28 2000-03-22 Zeneca Ltd Chemical compounds
US7217722B2 (en) 2000-02-01 2007-05-15 Kirin Beer Kabushiki Kaisha Nitrogen-containing compounds having kinase inhibitory activity and drugs containing the same
US6613798B1 (en) 2000-03-30 2003-09-02 Curis, Inc. Small organic molecule regulators of cell proliferation
US7115653B2 (en) 2000-03-30 2006-10-03 Curis, Inc. Small organic molecule regulators of cell proliferation
US20020127625A1 (en) 2000-03-31 2002-09-12 Forskarpatent Is Syd Ab Methods of diagnosing immune related diseases
KR100785363B1 (en) 2000-04-25 2007-12-18 이코스 코포레이션 Inhibitors of human phosphatidyl-inositol 3-kinase delta
US6667300B2 (en) 2000-04-25 2003-12-23 Icos Corporation Inhibitors of human phosphatidylinositol 3-kinase delta
US6777439B2 (en) 2000-05-30 2004-08-17 Advanced Research & Technology Institute, Inc. Compositions and methods for identifying agents which modulate PTEN function and PI-3 kinase pathways
WO2002000650A2 (en) 2000-06-27 2002-01-03 Genelabs Technologies, Inc. Novel compounds possessing antibacterial, antifungal or antitumor activity
AU2001267852B2 (en) 2000-06-30 2006-01-19 Msd K.K. Novel pyrazinone derivatives
US6534691B2 (en) 2000-07-18 2003-03-18 E. I. Du Pont De Nemours And Company Manufacturing process for α-olefins
AU2002213467A8 (en) 2000-10-11 2009-07-30 Chemocentryx Inc Modulation of ccr4 function
EP1317464B1 (en) 2000-10-11 2009-05-13 Applera Corporation Fluorescent nucleobase conjugates having anionic linkers
JP2002131859A (en) 2000-10-19 2002-05-09 Konica Corp Infrared sensitive silver halide photographic sensitive material for photographing and infrared sensitive silver halide emulsion
US6890747B2 (en) 2000-10-23 2005-05-10 Warner-Lambert Company Phosphoinositide 3-kinases
IL156304A0 (en) 2000-12-11 2004-01-04 Tularik Inc Cxcr3 antagonists
RU2290403C2 (en) 2000-12-28 2006-12-27 Дайити Фармасьютикал Ко., Лтд. Vla-4 inhibitors
MY134070A (en) 2001-01-22 2007-11-30 Isis Pharmaceuticals Inc Nucleoside derivatives as inhibitors of rna-dependent rna viral polymerase
US7105499B2 (en) 2001-01-22 2006-09-12 Merck & Co., Inc. Nucleoside derivatives as inhibitors of RNA-dependent RNA viral polymerase
GB0102239D0 (en) 2001-01-29 2001-03-14 Cancer Res Ventures Ltd Methods of chemical synthisis
US20030207910A1 (en) 2001-02-02 2003-11-06 Tao Wang Composition and antiviral activity of substituted azaindoleoxoacetic piperazine derivatives
MXPA03008560A (en) 2001-03-22 2004-06-30 Abbot Gmbh & Co Kg Single-stage pfc + ballast control circuit/general purpose power converter.
JP4331944B2 (en) 2001-04-17 2009-09-16 大日本住友製薬株式会社 New adenine derivatives
US7250569B2 (en) 2001-04-26 2007-07-31 New York University School Of Medicine Method for dissolving nanostructural materials
JP2004528376A (en) 2001-05-08 2004-09-16 クドス ファーマシューティカルズ リミテッド Isoquinolinone derivatives as PARP inhibitors
WO2002094264A1 (en) 2001-05-23 2002-11-28 Tularik Inc. Ccr4 antagonists
US6777425B2 (en) 2001-06-13 2004-08-17 Genesoft Pharmaceuticals, Inc. Isoquinoline compounds having antiinfective activity
US6825228B2 (en) 2001-06-13 2004-11-30 Genesoft Pharmaceuticals, Inc. Benzothiophene compounds having antiinfective activity
WO2002101007A2 (en) 2001-06-13 2002-12-19 Genesoft Pharmaceuticals, Inc Antipathogenic benzamide compounds
PE20030008A1 (en) 2001-06-19 2003-01-22 Bristol Myers Squibb Co DUAL INHIBITORS OF PDE 7 AND PDE 4
WO2003000187A2 (en) 2001-06-21 2003-01-03 Ariad Pharmaceuticals, Inc. Novel pyrazolo-and pyrrolo-pyrimidines and uses thereof
WO2003011285A1 (en) 2001-08-01 2003-02-13 Merck & Co., Inc. BENZIMIDAZO[4,5-f]ISOQUINOLINONE DERIVATIVES
AU2002327422A1 (en) 2001-08-03 2003-03-18 Abbott Laboratories Method of identifying inhibitors of lck
ATE530520T1 (en) 2001-08-10 2011-11-15 Shionogi & Co ANTIVIRAL AGENT
JP2003073357A (en) 2001-09-03 2003-03-12 Mitsubishi Pharma Corp Rho KINASE INHIBITOR COMPRISING AMIDE COMPOUND
EP1572072A4 (en) 2001-09-13 2009-04-01 Genesoft Inc Methods of treating infection by drug resistant bacteria
AUPR769501A0 (en) 2001-09-14 2001-10-11 Biomolecular Research Institute Limited Cytokine receptor 1
US7101884B2 (en) 2001-09-14 2006-09-05 Merck & Co., Inc. Tyrosine kinase inhibitors
US7269663B2 (en) 2001-09-28 2007-09-11 Intel Corporation Tagging packets with a lookup key to facilitate usage of a unified packet forwarding cache
TWI330183B (en) 2001-10-22 2010-09-11 Eisai R&D Man Co Ltd
AU2002363176B2 (en) 2001-11-01 2008-09-25 Janssen Pharmaceutica N.V. Heteroaryl amines as glycogen synthase kinase 3Beta inhibitors (GSK3 inhibitors)
AU2002350217A1 (en) 2001-12-04 2003-06-17 Bristol-Myers Squibb Company Glycinamides as factor xa inhibitors
JP4085237B2 (en) 2001-12-21 2008-05-14 日本電気株式会社 Mobile phone usage contract system and communication method
EP1465869B1 (en) 2001-12-21 2013-05-15 Exelixis Patent Company LLC Modulators of lxr
US7064218B2 (en) 2001-12-26 2006-06-20 Genelabs Technologies, Inc. Aromatic compounds and poly(oxyalkylene) containing aromatic compounds possessing antibacterial, antifungal or antitumor activity
US7414036B2 (en) 2002-01-25 2008-08-19 Muscagen Limited Compounds useful as A3 adenosine receptor agonists
US20040043959A1 (en) 2002-03-04 2004-03-04 Bloom Laura A. Combination therapies for treating methylthioadenosine phosphorylase deficient cells
AU2003225933A1 (en) 2002-03-22 2003-10-13 Cellular Genomics, Inc. AN IMPROVED FORMULATION OF CERTAIN PYRAZOLO(3,4-d) PYRIMIDINES AS KINASE MODULATORS
JP4663986B2 (en) 2002-03-26 2011-04-06 ツェントファーム・ゲーエムベーハー Fredericamycin-derivative
US7166293B2 (en) 2002-03-29 2007-01-23 Carlsbad Technology, Inc. Angiogenesis inhibitors
DE10217046A1 (en) 2002-04-17 2003-11-06 Bioleads Gmbh Fredericamycin derivatives
AU2003223012A1 (en) 2002-04-26 2003-11-10 Pfizer Products Inc. Pyrimidine-2, 4, 6-trione metallo-proteinase inhibitors
US6794562B2 (en) 2002-05-01 2004-09-21 Stine Seed Farm, Inc. Soybean cultivar 0332143
CA2485343A1 (en) 2002-05-23 2004-05-13 Merck & Co., Inc. Mitotic kinesin inhibitors
WO2003106426A1 (en) 2002-06-14 2003-12-24 Cytokinetics, Inc. Compounds, compositions, and methods
RU2340605C2 (en) 2002-06-27 2008-12-10 Ново Нордиск А/С Arylcarbonyl derivatives as therapeutic agents
EP1531815B1 (en) 2002-06-27 2014-09-24 Novo Nordisk A/S Glucokinase activators
US7265111B2 (en) 2002-06-27 2007-09-04 Sanofi-Aventis Deutschland Gmbh Adenosine analogues and their use as pharmaceutical agents
DE10230917A1 (en) 2002-07-09 2004-02-05 Bioleads Gmbh Fredericamycin derivatives
AU2003249244A1 (en) 2002-07-15 2004-02-02 Combinatorx, Incorporated Methods for the treatment of neoplasms
WO2004014377A1 (en) 2002-08-13 2004-02-19 Warner-Lambert Company Llc 4-hydroxyquinoline derivatives as matrix metalloproteinase inhibitors
GB0219054D0 (en) * 2002-08-15 2002-09-25 Cyclacel Ltd New purine derivatives
NZ538420A (en) 2002-08-16 2007-06-29 Kinacia Pty Ltd Inhibition of phosphoinositide 3-kinase beta
US20040048853A1 (en) 2002-08-21 2004-03-11 Gustave Bergnes Compounds, compositions, and methods
US20030139427A1 (en) 2002-08-23 2003-07-24 Osi Pharmaceuticals Inc. Bicyclic pyrimidinyl derivatives and methods of use thereof
GB0220319D0 (en) 2002-09-02 2002-10-09 Cancer Res Campaign Tech Enzyme activated self-immolative nitrogen mustard drugs
JP2004115450A (en) * 2002-09-27 2004-04-15 Tanabe Seiyaku Co Ltd Medicinal composition
AU2003266668A1 (en) 2002-09-30 2004-04-23 Banyu Pharmaceutical Co., Ltd. 2-aminobenzimidazole derivative
JP2004161716A (en) 2002-11-15 2004-06-10 Takeda Chem Ind Ltd Jnk inhibitor
ATE496893T1 (en) 2002-12-20 2011-02-15 X Ceptor Therapeutics Inc ISOQUINOLINONE DERIVATIVES AND THEIR USE AS MEDICATIONS
US7247736B2 (en) 2002-12-23 2007-07-24 4Sc Ag Method of identifying inhibitors of DHODH
US7365094B2 (en) 2002-12-23 2008-04-29 4Sc Ag Compounds as anti-inflammatory, immunomodulatory and anti-proliferatory agents
WO2004056746A1 (en) 2002-12-23 2004-07-08 4Sc Ag Cycloalkene dicarboxylic acid compounds as anti-inflammatory, immunomodulatory and anti-proliferatory agents
RU2328499C2 (en) * 2003-01-09 2008-07-10 Астеллас Фарма Инк. Pyrrolopyrazine derivatives, pharmaceutical composition, method for prevention and treatment of diseases, application as phosphodiesterase iv and/or tumor necrosis factor production inhibitor
WO2004078702A1 (en) 2003-03-06 2004-09-16 Dsm Ip Assets B.V. PROCESS FOR THE PREPARATION OF AN α-AMINO CARBONYL COMPOUND
US7687625B2 (en) 2003-03-25 2010-03-30 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
GB0306907D0 (en) 2003-03-26 2003-04-30 Angiogene Pharm Ltd Boireductively-activated prodrugs
US7217794B2 (en) 2003-04-02 2007-05-15 Daiamed, Inc. Compounds and methods for treatment of thrombosis
WO2004111014A1 (en) 2003-06-06 2004-12-23 Vertex Pharmaceuticals Incorporated Pyrimidine derivatives as modulators of atp-binding cassette transporters
US7429596B2 (en) 2003-06-20 2008-09-30 The Regents Of The University Of California 1H-pyrrolo [2,3-D] pyrimidine derivatives and methods of use thereof
WO2005002585A1 (en) 2003-07-02 2005-01-13 Warner-Lambert Company Llc Combination of an allosteric inhibitor of matrix metalloproteinase-13 and a ligand to an alpha-2-delta receptor
GB0317951D0 (en) 2003-07-31 2003-09-03 Trigen Ltd Compounds
US7459472B2 (en) 2003-08-08 2008-12-02 Transtech Pharma, Inc. Aryl and heteroaryl compounds, compositions, and methods of use
US7208601B2 (en) 2003-08-08 2007-04-24 Mjalli Adnan M M Aryl and heteroaryl compounds, compositions, and methods of use
WO2005016348A1 (en) 2003-08-14 2005-02-24 Icos Corporation Method of inhibiting immune responses stimulated by an endogenous factor
US20050054614A1 (en) 2003-08-14 2005-03-10 Diacovo Thomas G. Methods of inhibiting leukocyte accumulation
MXPA06001758A (en) 2003-08-15 2006-08-11 Irm Llc 6-substituted anilino purines as rtk inhibitors.
US7390820B2 (en) 2003-08-25 2008-06-24 Amgen Inc. Substituted quinolinone derivatives and methods of use
WO2005044181A2 (en) 2003-09-09 2005-05-19 Temple University-Of The Commonwealth System Of Higher Education Protection of tissues and cells from cytotoxic effects of ionizing radiation by abl inhibitors
GB0322409D0 (en) 2003-09-25 2003-10-29 Astrazeneca Ab Quinazoline derivatives
AU2004289303A1 (en) 2003-11-10 2005-05-26 Synta Pharmaceuticals, Corp. Fused heterocyclic compounds
JP2007511596A (en) 2003-11-17 2007-05-10 ファイザー・プロダクツ・インク Pyrrolopyrimidine compounds useful in the treatment of cancer
US7449477B2 (en) 2003-11-25 2008-11-11 Eli Lilly And Company 7-phenyl-isoquinoline-5-sulfonylamino derivatives as inhibitors of akt (protein kinase B)
EP1692112A4 (en) 2003-12-08 2008-09-24 Cytokinetics Inc Compounds, compositions, and methods
AU2004308974A1 (en) 2003-12-22 2005-07-14 Gilead Sciences, Inc. Kinase inhibitor phosphonate conjugates
AU2004309420B2 (en) 2003-12-23 2008-10-30 Novartis Ag Bicyclic heterocyclic p-38 kinase inhibitors
KR20060105872A (en) 2003-12-29 2006-10-11 반유 세이야꾸 가부시끼가이샤 Novel 2-heteroaryl-substituted benzimidazole derivative
CA2552664A1 (en) 2004-01-08 2005-07-28 Michigan State University Methods for treating and preventing hypertension and hypertension-related disorders
US20050214310A1 (en) 2004-01-23 2005-09-29 Seattle Genetics, Inc. Melphalan prodrugs
CA2553724A1 (en) 2004-02-03 2005-08-18 Abbott Laboratories Aminobenzoxazoles as therapeutic agents
UA83416C2 (en) 2004-02-13 2008-07-10 Баниу Фармасьютикал Ко., Лтд. Fused ring 4-oxopyrimidine derivative
US20050187418A1 (en) 2004-02-19 2005-08-25 Small Brooke L. Olefin oligomerization
CA2556589A1 (en) 2004-02-24 2005-09-01 Bioaxone Therapeutique Inc. 4-substituted piperidine derivatives
BRPI0508036A (en) 2004-02-27 2007-07-17 Hoffmann La Roche fused pyrazole derivatives
MXPA06012333A (en) 2004-04-30 2007-01-17 Takeda Pharmaceutical Heterocyclic amide compound and use thereof as an mmp-13 inhibitor.
DE102004022897A1 (en) 2004-05-10 2005-12-08 Bayer Cropscience Ag Azinyl-imidazoazines
CA2730540A1 (en) 2004-05-13 2005-12-01 Vanderbilt University Phosphoinositide 3-kinase delta selective inhibitors for inhibiting angiogenesis
CA2566609C (en) 2004-05-13 2012-06-26 Icos Corporation Quinazolinones as inhibitors of human phosphatidylinositol 3-kinase delta
CA2567883A1 (en) 2004-05-25 2005-12-15 Icos Corporation Methods for treating and/or preventing aberrant proliferation of hematopoietic cells
EP1750715A1 (en) 2004-06-04 2007-02-14 Icos Corporation Methods for treating mast cell disorders
GB0420722D0 (en) 2004-09-17 2004-10-20 Addex Pharmaceuticals Sa Novel allosteric modulators
WO2006038865A1 (en) 2004-10-01 2006-04-13 Betagenon Ab Nucleotide derivatives for the treatment of type 2 diabetes and other disorders
US8212012B2 (en) 2004-11-03 2012-07-03 University Of Kansas Novobiocin analogues having modified sugar moieties
US8212011B2 (en) 2004-11-03 2012-07-03 University Of Kansas Novobiocin analogues
US7622451B2 (en) 2004-11-03 2009-11-24 University Of Kansas Novobiocin analogues as neuroprotective agents and in the treatment of autoimmune disorders
CA2585091C (en) 2004-11-03 2016-07-19 University Of Kansas Novobiocin analogues as anticancer agents
GB0425035D0 (en) 2004-11-12 2004-12-15 Novartis Ag Organic compounds
US9512125B2 (en) 2004-11-19 2016-12-06 The Regents Of The University Of California Substituted pyrazolo[3.4-D] pyrimidines as anti-inflammatory agents
MX2007006178A (en) 2004-11-23 2007-06-20 Ptc Therapeutics Inc Substituted phenols as active agents inhibiting vegf production.
US20060156485A1 (en) 2005-01-14 2006-07-20 The Procter & Gamble Company Keratin dyeing compounds, keratin dyeing compositions containing them, and use thereof
GB0501999D0 (en) 2005-02-01 2005-03-09 Sentinel Oncology Ltd Pharmaceutical compounds
EP1885356A2 (en) 2005-02-17 2008-02-13 Icos Corporation Phosphoinositide 3-kinase inhibitors for inhibiting leukocyte accumulation
US7579348B2 (en) 2005-02-25 2009-08-25 Pgxhealth, Llc Derivatives of 8-substituted xanthines
US20090124654A1 (en) 2005-03-01 2009-05-14 Mjalli Adnan M M Aryl and Heteroaryl Compounds, Compositions, Methods of Use
US7872050B2 (en) 2005-03-14 2011-01-18 Yaupon Therapeutics Inc. Stabilized compositions of volatile alkylating agents and methods of using thereof
US7544672B2 (en) 2005-03-30 2009-06-09 Conforma Therapeutics Corporation Alkynyl pyrrolo[2,3-d]pyrimidines and related analogs as HSP90-inhibitors
KR20080013886A (en) * 2005-04-05 2008-02-13 파마코페이아, 인코포레이티드 Purine and imidazopyridine derivatives for immunosuppression
MX2007012449A (en) 2005-04-06 2007-12-05 Irm Llc Diarylamine-containing compounds and compositions, and their use as modulators of steroid hormone nuclear receptors.
KR100781704B1 (en) 2005-04-20 2007-12-03 에스케이케미칼주식회사 Novel pyridine derivatives, process for preparing thereof and pharmaceutical compositions containing them
WO2006114065A2 (en) 2005-04-25 2006-11-02 Institute Of Organic Chemistry And Biochemistry A Cademy Of Sciences Of The Czech Republic Use of compounds to inhibit neoplasia
CN100526315C (en) 2005-06-16 2009-08-12 浙江医药股份有限公司新昌制药厂 N2-quinoline or isoquinoline substituted purine derivative and its preparation method and uses
SI1906965T1 (en) 2005-06-22 2015-09-30 Chemocentryx, Inc. Azaindazole compounds and methods of use
ATE474829T1 (en) 2005-06-27 2010-08-15 Amgen Inc ANTI-INFLAMMATORY ARYLNITRIL COMPOUNDS
WO2007006547A1 (en) 2005-07-11 2007-01-18 Devgen N.V. Amide derivatives as kinase inhibitors
JP2009505948A (en) 2005-07-11 2009-02-12 デブジェン エヌブイ Amide derivatives as kinase inhibitors
US20070017915A1 (en) 2005-07-22 2007-01-25 Weder Donald E Collapsible and/or erectable substantially egg-shaped container
GB0516723D0 (en) 2005-08-15 2005-09-21 Novartis Ag Organic compounds
US20070049591A1 (en) 2005-08-25 2007-03-01 Kalypsys, Inc. Inhibitors of MAPK/Erk Kinase
EP1919873A1 (en) 2005-09-01 2008-05-14 BioAgency AG Fredericamycin derivatives
US20070072897A1 (en) 2005-09-29 2007-03-29 Wyeth Phenylaminopropanol derivatives and methods of their use
GB0522880D0 (en) * 2005-11-09 2005-12-21 Glaxo Group Ltd Novel compounds
CA2628534A1 (en) * 2005-11-10 2007-05-18 Schering Corporation Methods for inhibiting protein kinases
BRPI0618622A2 (en) 2005-11-17 2011-09-06 Osi Pharm Inc compound, composition, and use of a compound
CA2629192A1 (en) 2005-11-22 2007-05-31 Merck & Co., Inc. Indole orexin receptor antagonists
US7572809B2 (en) 2005-12-19 2009-08-11 Hoffmann-La Roche Inc. Isoquinoline aminopyrazole derivatives
WO2007075554A2 (en) 2005-12-19 2007-07-05 Osi Pharmaceuticals, Inc. Combination of igfr inhibitor and anti-cancer agent
CN101389630A (en) 2005-12-29 2009-03-18 艾博特公司 Protein kinase inhibitors
CN101374844B (en) 2006-01-16 2012-10-24 宇部兴产株式会社 Pyrrolopyridazinone compound
WO2007089669A2 (en) 2006-01-26 2007-08-09 Wyeth Processes for the preparation of compounds which modulate cell proliferation
US7723330B2 (en) 2006-03-07 2010-05-25 Array Biopharma Inc. Heterobicyclic pyrazole compounds and methods of use
DK2004683T3 (en) 2006-03-24 2016-08-22 Biogen Hemophilia Inc PC5 AS A FACTOR IX PROPEPTID PROCESSING ENZYM
EP2007373A4 (en) 2006-03-29 2012-12-19 Foldrx Pharmaceuticals Inc Inhibition of alpha-synuclein toxicity
MX2008012482A (en) * 2006-03-31 2008-10-10 Abbott Lab Indazole compounds.
US20080032960A1 (en) 2006-04-04 2008-02-07 Regents Of The University Of California PI3 kinase antagonists
WO2007121453A2 (en) 2006-04-17 2007-10-25 The Regents Of The University Of California 2-hydroxy-1-oxo 1,2 dihydro isoquinoline chelating agents
GB0607950D0 (en) 2006-04-21 2006-05-31 Novartis Ag Organic compounds
GB0607948D0 (en) 2006-04-21 2006-05-31 Novartis Ag Organic compounds
ATE549337T1 (en) 2006-04-21 2012-03-15 Novartis Ag PURINE DERIVATIVES FOR USE AS ADENOSINE A2A RECEPTOR AGONISTS
WO2007125315A2 (en) 2006-04-25 2007-11-08 Astex Therapeutics Limited Pharmaceutical compounds
WO2007125310A2 (en) 2006-04-25 2007-11-08 Astex Therapeutics Limited Pharmaceutical combinations of pk inhibitors and other active agents
DE102006020327A1 (en) 2006-04-27 2007-12-27 Bayer Healthcare Ag Heterocyclic substituted, fused pyrazole derivatives and their use
EP2029593A1 (en) 2006-05-22 2009-03-04 AstraZeneca AB Indole derivatives
GB0610242D0 (en) 2006-05-23 2006-07-05 Novartis Ag Organic compounds
EP1859772B1 (en) 2006-05-24 2010-12-15 Guardant S.r.l. Alkalized local anesthetic in bag
GB0610317D0 (en) 2006-05-24 2006-07-05 Medical Res Council Antiparasitic compounds and compositions
DE602007006835D1 (en) 2006-07-20 2010-07-08 Amgen Inc SUBSTITUTED PYRIDONE COMPOUNDS AND APPLICATION METHODS
MX2009000864A (en) 2006-07-28 2009-02-03 Novartis Ag 2,4-substituted quinazolines as lipid kinase inhibitors.
US8541428B2 (en) 2006-08-22 2013-09-24 Technion Research And Development Foundation Ltd. Heterocyclic derivatives, pharmaceutical compositions and methods of use thereof
CA2661436A1 (en) 2006-08-24 2008-02-28 Serenex, Inc. Isoquinoline, quinazoline and phthalazine derivatives
MX2009002298A (en) 2006-09-01 2009-06-04 Cylene Pharmaceuticals Inc Serine-threonine protein kinase and parp modulators.
WO2008025755A1 (en) 2006-09-01 2008-03-06 Basf Se Use of n-containing heterocycles in dermocosmetics
EP1903044A1 (en) 2006-09-14 2008-03-26 Novartis AG Adenosine Derivatives as A2A Receptor Agonists
US8492405B2 (en) 2006-10-18 2013-07-23 Takeda Pharmaceutical Company Limited Glucokinase-activating fused heterocyclic compounds and methods of treating diabetes and obesity
US7772180B2 (en) 2006-11-09 2010-08-10 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
CN101605797A (en) 2006-11-13 2009-12-16 伊莱利利公司 The Thienopyrimidinones of treatment inflammatory disease and cancer
WO2008063625A2 (en) 2006-11-20 2008-05-29 Adolor Corporation Pyridine compounds and methods of their use
US8436177B2 (en) 2006-11-20 2013-05-07 Novartis Ag Salts and crystall forms of 2-methyl-2-[4-(3-methyl-2-oxo-8-quinolin-3-yl-2,3-dihydro-imidazo[4,5-c]quinolin-1-yl)-phenyl]-propionitrile
JP2010512331A (en) 2006-12-06 2010-04-22 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Glucocorticoid mimetics, methods for their production, pharmaceutical compositions, and uses thereof
EP2132178B1 (en) 2006-12-20 2015-08-19 Merck Sharp & Dohme Corp. Jnk inhibitors
DK2114925T3 (en) 2006-12-22 2012-05-29 Ind Res Ltd Azetidine analogs of nucleosidase and phosphorylase inhibitors
BRPI0807182A2 (en) 2007-01-26 2014-05-27 Irm Llc COMPOUNDS AND COMPOSITIONS AS KINASE INHIBITORS
WO2008112715A2 (en) 2007-03-12 2008-09-18 Vm Discovery Inc. Novel agents of calcium ion channel modulators
WO2008118454A2 (en) * 2007-03-23 2008-10-02 Amgen Inc. Derivatives of quinoline or benzopyrazine and their uses for the treatment of (inter alia) inflammatory diseases, autoimmune diseases or various kinds of cancer
ES2563458T3 (en) 2007-03-23 2016-03-15 Amgen Inc. Heterocyclic compounds and their uses
PT2139882E (en) 2007-03-23 2014-01-30 Amgen Inc 3- substituted quinoline or quinoxaline derivatives and their use as phosphatidylinositol 3-kinase (pi3k) inhibitors
US7867983B2 (en) 2007-03-29 2011-01-11 The University Of Connecticut Methods to protect skeletal muscle against injury
EP2134682B1 (en) 2007-04-13 2011-12-14 Sanofi A transition metal catalyzed synthesis of n-aminoindoles
CN101636397B (en) 2007-04-13 2012-06-13 中国人民解放军军事医学科学院毒物药物研究所 Urea compounds, preparation methods and pharmaceutical uses thereof
JP2010163361A (en) 2007-04-27 2010-07-29 Dainippon Sumitomo Pharma Co Ltd Quinoline derivative
US7960353B2 (en) 2007-05-10 2011-06-14 University Of Kansas Novobiocin analogues as neuroprotective agents and in the treatment of autoimmune disorders
US20090214529A9 (en) 2007-05-22 2009-08-27 Taigen Biotechnology Co., Ltd. Kinesin inhibitors
US9603848B2 (en) 2007-06-08 2017-03-28 Senomyx, Inc. Modulation of chemosensory receptors and ligands associated therewith
US7928111B2 (en) 2007-06-08 2011-04-19 Senomyx, Inc. Compounds including substituted thienopyrimidinone derivatives as ligands for modulating chemosensory receptors
EP2173747B1 (en) 2007-06-26 2011-12-21 Sanofi A regioselective metal catalyzed synthesis of annelated benzimidazoles and azabenzimidazoles
EP2170274A1 (en) 2007-07-02 2010-04-07 Technion Research and Development Foundation, Ltd. Compositions, articles and methods comprising tspo ligands for preventing or reducing tobacco-associated damage
RU2345996C1 (en) 2007-07-17 2009-02-10 Андрей Александрович Иващенко Annelated azaheterocyclic amides, which include pyrimidine fragment, method of obtaining and their application
CA2696113A1 (en) 2007-08-10 2009-04-02 Burnham Institute For Medical Research Tissue-nonspecific alkaline phosphatase (tnap) activators and uses thereof
CN106279283A (en) 2007-08-13 2017-01-04 症变治疗公司 Novel activators of glucokinase
WO2009029617A1 (en) 2007-08-27 2009-03-05 Kalypsys, Inc. Diarylamine-substituted quinolones useful as inducible nitric oxide synthase inhibitors
JP5227965B2 (en) 2007-10-03 2013-07-03 独立行政法人理化学研究所 Nitrotriazole derivative and method for producing compound using the same
KR20100089082A (en) 2007-10-15 2010-08-11 아스트라제네카 아베 Combination 059
JP5256693B2 (en) 2007-10-31 2013-08-07 信越化学工業株式会社 Production method of titanium oxide photocatalyst thin film
AU2008321099A1 (en) 2007-11-13 2009-05-22 Icos Corporation Inhibitors of human phosphatidyl-inositol 3-kinase delta
US20090163481A1 (en) 2007-12-13 2009-06-25 Murphy Brian J Ppar-delta ligands and methods of their use
US8399483B2 (en) 2007-12-21 2013-03-19 Ucb Pharma S.A. Quinoxaline and quinoline derivatives as kinase inhibitors
US7960397B2 (en) 2007-12-28 2011-06-14 Institute Of Experimental Botany, Academy Of Sciences Of The Czech Republic 6,9-disubstituted purine derivatives and their use as cosmetics and cosmetic compositions
US8193182B2 (en) 2008-01-04 2012-06-05 Intellikine, Inc. Substituted isoquinolin-1(2H)-ones, and methods of use thereof
NZ613219A (en) 2008-01-04 2014-11-28 Intellikine Llc Heterocyclic containing entities, compositions and methods
EA201001135A1 (en) 2008-01-09 2011-02-28 ПиДжиИксХЭЛС ЭлЭлСи INTRATECAL TREATMENT OF NEUROPATHIC PAIN BY AR AGONISTS
EP2252616B1 (en) * 2008-01-30 2014-07-23 Genentech, Inc. Pyrazolopyrimidine pi3k inhibitor compounds and methods of use
WO2009100406A2 (en) 2008-02-07 2009-08-13 Synta Pharmaceuticals Corp. Topical formulations for the treatment of psoriasis
JP2011511802A (en) 2008-02-07 2011-04-14 ギリアード・パロ・アルト・インコーポレイテッド Compounds that increase ABCA-1 and methods of using such compounds
TWI444384B (en) 2008-02-20 2014-07-11 Gilead Sciences Inc Nucleotide analogues and their use in the treatment of malignancies
WO2009114874A2 (en) 2008-03-14 2009-09-17 Intellikine, Inc. Benzothiazole kinase inhibitors and methods of use
MX2010010151A (en) 2008-03-20 2010-10-25 Amgen Inc Aurora kinase modulators and method of use.
EP2279188B1 (en) 2008-05-30 2015-01-28 Genentech, Inc. Purine pi3k inhibitor compounds and methods of use
US20090312406A1 (en) 2008-06-12 2009-12-17 Hsing-Pang Hsieh Coumarin compounds and their use for treating viral infection
US20110224223A1 (en) 2008-07-08 2011-09-15 The Regents Of The University Of California, A California Corporation MTOR Modulators and Uses Thereof
CN102124009B (en) 2008-07-08 2014-07-23 因特利凯公司 Kinase inhibitors and methods of use
CA2730610A1 (en) 2008-07-16 2010-01-21 Schering Corporation Bicyclic heterocycle derivatives and their use as gpcr modulators
US8450344B2 (en) 2008-07-25 2013-05-28 Aerie Pharmaceuticals, Inc. Beta- and gamma-amino-isoquinoline amide compounds and substituted benzamide compounds
CN102177152A (en) 2008-08-11 2011-09-07 哈佛大学校长及研究员协会 Halofuginone analogs for inhibition of tRNA synthetases and uses thereof
EP2344481B9 (en) 2008-09-23 2014-12-31 Georgetown University Viral and fungal inhibitors
WO2010036380A1 (en) 2008-09-26 2010-04-01 Intellikine, Inc. Heterocyclic kinase inhibitors
US8476431B2 (en) 2008-11-03 2013-07-02 Itellikine LLC Benzoxazole kinase inhibitors and methods of use
US20110269779A1 (en) 2008-11-18 2011-11-03 Intellikine, Inc. Methods and compositions for treatment of ophthalmic conditions
CA2745280A1 (en) 2008-12-04 2010-06-10 The United States Of America, As Represented By The Secretary, Departmen T Of Health And Human Services Phosphatidylinositol-3-kinase p110 delta-targeted drugs in the treatment of cns disorders
EA020548B1 (en) 2008-12-19 2014-12-30 Бёрингер Ингельхайм Интернациональ Гмбх Cyclic pyrimidin-4-carboxamides as ccr2 receptor antagonists for treatment of inflammation, asthma and copd
CA2749585A1 (en) 2009-01-13 2010-07-22 Van Andel Research Institute Methods of using substituted isoxazolo pyridinones as dissociated glucocorticoids
WO2010092340A1 (en) 2009-02-13 2010-08-19 Ucb Pharma S.A. Fused pyridine and pyrazine derivatives as kinase inhibitors
EP2427195B1 (en) 2009-05-07 2019-05-01 Intellikine, LLC Heterocyclic compounds and uses thereof
GB0908957D0 (en) 2009-05-22 2009-07-01 Ucb Pharma Sa Therapeutic agents
CN101602768B (en) 2009-07-17 2012-05-30 河南省农科院农副产品加工研究所 Method for purifying sesamin and sesamolin
US8106146B2 (en) 2009-10-06 2012-01-31 Medtronic, Inc. Therapeutic polymers and methods of generation
EP2499129B1 (en) 2009-11-12 2014-07-30 UCB Pharma, S.A. Quinoline and quinoxaline derivatives as kinase inhibitors
WO2011058109A1 (en) 2009-11-12 2011-05-19 Ucb Pharma S.A. Fused bicyclic pyrrole and imidazole derivatives as kinase inhibitors
US8637543B2 (en) 2009-11-12 2014-01-28 Ucb Pharma S.A. Quinoline derivatives as kinase inhibitors
WO2011075628A1 (en) 2009-12-18 2011-06-23 Amgen Inc. Heterocyclic compounds and their uses
ES2842399T3 (en) 2009-12-22 2021-07-14 Vertex Pharma Isoindolinone phosphatidylinositol 3-kinase inhibitors
ES2593256T3 (en) 2010-05-21 2016-12-07 Infinity Pharmaceuticals, Inc. Chemical compounds, compositions and methods for kinase modulations
AR082799A1 (en) 2010-09-08 2013-01-09 Ucb Pharma Sa DERIVATIVES OF QUINOLINE AND QUINOXALINE AS QUINASE INHIBITORS
WO2012061696A1 (en) 2010-11-04 2012-05-10 Amgen Inc. 5 -cyano-4, 6 -diaminopyrimidine or 6 -aminopurine derivatives as pi3k- delta inhibitors
CA2817577A1 (en) 2010-11-10 2012-05-18 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
UA115767C2 (en) 2011-01-10 2017-12-26 Інфініті Фармасьютікалз, Інк. Processes for preparing isoquinolinones and solid forms of isoquinolinones
US8791107B2 (en) 2011-02-25 2014-07-29 Takeda Pharmaceutical Company Limited N-substituted oxazinopteridines and oxazinopteridinones
US8969363B2 (en) 2011-07-19 2015-03-03 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9056877B2 (en) 2011-07-19 2015-06-16 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
PE20141371A1 (en) 2011-08-29 2014-10-13 Infinity Pharmaceuticals Inc HETEROCYCLIC COMPOUNDS AND USES OF THEM

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060235031A1 (en) * 2004-04-02 2006-10-19 Arnold Lee D 6,6-Bicyclic ring substituted heterobicyclic protein kinase inhibitors
US20070054915A1 (en) * 2005-08-25 2007-03-08 Roche Palo Alto Llc p38 MAP kinase inhibitors and methods for using the same

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP2346508A4 *

Cited By (367)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9512125B2 (en) 2004-11-19 2016-12-06 The Regents Of The University Of California Substituted pyrazolo[3.4-D] pyrimidines as anti-inflammatory agents
US8642604B2 (en) 2006-04-04 2014-02-04 The Regents Of The University Of California Substituted pyrazolo[3,2-d]pyrimidines as anti-cancer agents
US9493467B2 (en) 2006-04-04 2016-11-15 The Regents Of The University Of California PI3 kinase antagonists
US8183258B2 (en) 2007-03-23 2012-05-22 Amgen Inc. Heterocyclic compounds and their uses
US8586739B2 (en) 2007-03-23 2013-11-19 Amgen Inc. Heterocyclic compounds and their uses
US9873701B2 (en) 2007-03-23 2018-01-23 Amgen Inc. Heterocyclic compounds and their uses
US8901135B2 (en) 2007-03-23 2014-12-02 Amgen Inc. Heterocyclic compounds and their uses
US8329910B2 (en) 2007-03-23 2012-12-11 Amgen Inc. Quinolines for the treatment of PI3K-δ mediated diseases
US20120220586A1 (en) * 2007-03-23 2012-08-30 Amgen Inc. Heterocyclic compounds and their uses
US8193199B2 (en) 2007-03-23 2012-06-05 Amgen Inc. Heterocyclic compounds and their uses
US8183259B2 (en) 2007-03-23 2012-05-22 Amgen Inc. Heterocyclic compounds and their uses
US9359349B2 (en) 2007-10-04 2016-06-07 Intellikine Llc Substituted quinazolines as kinase inhibitors
US11433065B2 (en) 2008-01-04 2022-09-06 Intellikine Llc Certain chemical entities, compositions and methods
US8703777B2 (en) 2008-01-04 2014-04-22 Intellikine Llc Certain chemical entities, compositions and methods
US9655892B2 (en) 2008-01-04 2017-05-23 Intellikine Llc Certain chemical entities, compositions and methods
US8785456B2 (en) 2008-01-04 2014-07-22 Intellikine Llc Substituted isoquinolin-1(2H)-ones, and methods of use thereof
US8193182B2 (en) 2008-01-04 2012-06-05 Intellikine, Inc. Substituted isoquinolin-1(2H)-ones, and methods of use thereof
US9216982B2 (en) 2008-01-04 2015-12-22 Intellikine Llc Certain chemical entities, compositions and methods
US9822131B2 (en) 2008-01-04 2017-11-21 Intellikine Llc Certain chemical entities, compositions and methods
US9637492B2 (en) 2008-03-14 2017-05-02 Intellikine Llc Benzothiazole kinase inhibitors and methods of use
US8993580B2 (en) 2008-03-14 2015-03-31 Intellikine Llc Benzothiazole kinase inhibitors and methods of use
US8637542B2 (en) 2008-03-14 2014-01-28 Intellikine, Inc. Kinase inhibitors and methods of use
US9096611B2 (en) 2008-07-08 2015-08-04 Intellikine Llc Kinase inhibitors and methods of use
US9828378B2 (en) 2008-07-08 2017-11-28 Intellikine Llc Kinase inhibitors and methods of use
US9629843B2 (en) 2008-07-08 2017-04-25 The Regents Of The University Of California MTOR modulators and uses thereof
US8703778B2 (en) 2008-09-26 2014-04-22 Intellikine Llc Heterocyclic kinase inhibitors
US9296742B2 (en) 2008-09-26 2016-03-29 Intellikine Llc Heterocyclic kinase inhibitors
US9790228B2 (en) 2008-09-26 2017-10-17 Intellikine Llc Heterocyclic kinase inhibitors
US8697709B2 (en) 2008-10-16 2014-04-15 The Regents Of The University Of California Fused ring heteroaryl kinase inhibitors
EP2365750A4 (en) * 2008-11-03 2012-08-01 Intellikine Inc Benzoxazole kinase inhibitors and methods of use
EP2365750A1 (en) * 2008-11-03 2011-09-21 Intellikine, Inc. Benzoxazole kinase inhibitors and methods of use
US9345706B2 (en) 2008-11-03 2016-05-24 Intellikine, Llc Benzoxazole kinase inhibitors and methods of use
US8476431B2 (en) 2008-11-03 2013-07-02 Itellikine LLC Benzoxazole kinase inhibitors and methods of use
US8476282B2 (en) 2008-11-03 2013-07-02 Intellikine Llc Benzoxazole kinase inhibitors and methods of use
AU2009310364B2 (en) * 2008-11-03 2016-08-18 Takeda Pharmaceutical Company Limited Benzoxazole kinase inhibitors and methods of use
EP3153023A1 (en) * 2008-11-03 2017-04-12 Intellikine, LLC Benzoxazole kinase inhibitors and methods of use
US8513284B2 (en) * 2009-02-13 2013-08-20 Ucb Pharma, S.A. Fused pyridine and pyrazine derivatives as kinase inhibitors
US20120077815A1 (en) * 2009-02-13 2012-03-29 Ucb Pharma S.A. Fused Pyridine And Pyrazine Derivatives As Kinase Inhibitors
US8729074B2 (en) 2009-03-20 2014-05-20 Amgen Inc. Inhibitors of PI3 kinase
US8785454B2 (en) 2009-05-07 2014-07-22 Intellikine Llc Heterocyclic compounds and uses thereof
US9315505B2 (en) 2009-05-07 2016-04-19 Intellikine Llc Heterocyclic compounds and uses thereof
US9873704B2 (en) 2009-06-25 2018-01-23 Amgen Inc. Heterocyclic compounds and their uses
US11401280B2 (en) 2009-06-29 2022-08-02 Incyte Holdings Corporation Pyrimidinones as PI3K inhibitors
US8940752B2 (en) 2009-06-29 2015-01-27 Incyte Corporation Pyrimidinones as PI3K inhibitors
US10428087B2 (en) 2009-06-29 2019-10-01 Incyte Corporation Pyrimidinones as PI3K inhibitors
US9975907B2 (en) 2009-06-29 2018-05-22 Incyte Holdings Corporation Pyrimidinones as PI3K inhibitors
US10829502B2 (en) 2009-06-29 2020-11-10 Incyte Corporation Pyrimidinones as PI3K inhibitors
US9434746B2 (en) 2009-06-29 2016-09-06 Incyte Corporation Pyrimidinones as PI3K inhibitors
US9206182B2 (en) 2009-07-15 2015-12-08 Intellikine Llc Substituted isoquinolin-1(2H)-one compounds, compositions, and methods thereof
US9522146B2 (en) 2009-07-15 2016-12-20 Intellikine Llc Substituted Isoquinolin-1(2H)-one compounds, compositions, and methods thereof
CN109912599B (en) * 2009-07-15 2021-11-02 英特利凯恩有限责任公司 Chemical entities, compositions, and methods
US8569323B2 (en) 2009-07-15 2013-10-29 Intellikine, Llc Substituted isoquinolin-1(2H)-one compounds, compositions, and methods thereof
CN109912599A (en) * 2009-07-15 2019-06-21 英特利凯恩有限责任公司 Chemical entities, composition and method
US11547697B2 (en) 2009-08-17 2023-01-10 Millennium Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9085560B2 (en) 2009-08-17 2015-07-21 Intellikine, Inc. Heterocyclic compounds and uses thereof
US8980899B2 (en) 2009-10-16 2015-03-17 The Regents Of The University Of California Methods of inhibiting Ire1
US20120208799A1 (en) * 2009-10-19 2012-08-16 John King-Underwood Compounds
US9834560B2 (en) 2009-10-19 2017-12-05 Respivert Ltd. Compounds
US9321773B2 (en) 2009-10-19 2016-04-26 Respivert, Ltd. Compounds
US8741909B2 (en) 2009-10-19 2014-06-03 Respivert Ltd. PI3 kinase inhibitors
WO2011058109A1 (en) 2009-11-12 2011-05-19 Ucb Pharma S.A. Fused bicyclic pyrrole and imidazole derivatives as kinase inhibitors
WO2011058111A1 (en) 2009-11-12 2011-05-19 Ucb Pharma S.A. Aminopurine derivatives as kinase inhibitors
WO2011058113A1 (en) 2009-11-12 2011-05-19 Ucb Pharma S.A. Fused bicyclic pyridine and pyrazine derivatives as kinase inhibitors
WO2011058112A1 (en) 2009-11-12 2011-05-19 Ucb Pharma S.A. Fused bicyclic pyrazole derivatives as kinase inhibitors
US9403847B2 (en) 2009-12-18 2016-08-02 Incyte Holdings Corporation Substituted heteroaryl fused derivatives as P13K inhibitors
US8633313B2 (en) 2009-12-18 2014-01-21 Amgen Inc. Heterocyclic compounds and their uses
US9193721B2 (en) 2010-04-14 2015-11-24 Incyte Holdings Corporation Fused derivatives as PI3Kδ inhibitors
US9181221B2 (en) 2010-05-21 2015-11-10 Infinity Pharmaceuticals, Inc. Chemical compounds, compositions and methods for kinase modulation
US8604032B2 (en) 2010-05-21 2013-12-10 Infinity Pharmaceuticals, Inc. Chemical compounds, compositions and methods for kinase modulation
US9738644B2 (en) 2010-05-21 2017-08-22 Infinity Pharmaceuticals, Inc. Chemical compounds, compositions and methods for kinase modulation
WO2011146882A1 (en) 2010-05-21 2011-11-24 Intellikine, Inc. Chemical compounds, compositions and methods for kinase modulation
CN103153062A (en) * 2010-05-24 2013-06-12 因特利凯有限责任公司 Heterocyclic compounds and uses thereof
WO2011149937A1 (en) * 2010-05-24 2011-12-01 Intellikine, Inc. Heterocyclic compounds and uses thereof
CN103153062B (en) * 2010-05-24 2015-07-15 因特利凯有限责任公司 Heterocyclic compounds and uses thereof
US9359352B2 (en) 2010-05-24 2016-06-07 Intellikine Llc Substituted benzimidazoles as PI3 kinase inhibitors
US9096590B2 (en) 2010-05-24 2015-08-04 Intellikine Llc Substituted benzoxazoles as PI3 kinase inhibitors
US9062055B2 (en) 2010-06-21 2015-06-23 Incyte Corporation Fused pyrrole derivatives as PI3K inhibitors
WO2012003283A1 (en) * 2010-06-30 2012-01-05 Amgen Inc. Heterocyclic compounds and their use as inhibitors of pi3k activity
WO2012003278A1 (en) * 2010-06-30 2012-01-05 Amgen Inc. Quinolines as p13k inhibitors
AU2011271460B2 (en) * 2010-07-01 2014-02-06 Amgen Inc. Heterocyclic compounds and their use as inhibitors of P13K activity
WO2012003262A1 (en) * 2010-07-01 2012-01-05 Amgen Inc. Heterocyclic compounds and their use as inhibitors of pi3k activity
WO2012003271A1 (en) * 2010-07-02 2012-01-05 Amgen Inc. Heterocyclic compounds and their use as inhibitors of pi3k activity
US10053470B2 (en) 2010-09-14 2018-08-21 Exelixis, Inc. Inhibitors of PI3K-delta and methods of their use and manufacture
WO2012037204A1 (en) * 2010-09-14 2012-03-22 Exelixis, Inc. Inhibitors of pi3k-delta and methods of their use and manufacture
US9670212B2 (en) 2010-09-14 2017-06-06 Exelixis, Inc. Inhibitors of PI3K-delta and methods of their use and manufacture
JP2013540746A (en) * 2010-09-14 2013-11-07 エクセリクシス, インク. Inhibitors of PI3K-delta and methods for their use and production
AU2011302196B2 (en) * 2010-09-14 2016-04-28 Exelixis, Inc. Inhibitors of PI3K-delta and methods of their use and manufacture
US10028959B2 (en) 2010-10-18 2018-07-24 Respivert Ltd. Quinazolin-4 (3H)-one derivatives used as P13 kinase inhibitors
US9637494B2 (en) 2010-10-18 2017-05-02 Respivert, Ltd. Quinazolin-4 (3H)-one derivatives used as P13 kinase inhibitors
US9340545B2 (en) 2010-10-18 2016-05-17 Respivert Ltd. Quinazolin-4 (3H)—one derivatives used as P13 kinase inhibitors
JP2013539780A (en) * 2010-10-18 2013-10-28 レスピバート・リミテツド Quinazolin-4 (3H) -one derivatives for use as PI3 kinase inhibitors
US9388183B2 (en) 2010-11-10 2016-07-12 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US8901133B2 (en) 2010-11-10 2014-12-02 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
WO2012064973A2 (en) 2010-11-10 2012-05-18 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
EP3660016A1 (en) * 2010-12-20 2020-06-03 Incyte Holdings Corporation N-(1-(substituted-phenyl)ethyl)-9h-purin-6-amines as pi3k inhibitors
WO2012087881A1 (en) * 2010-12-20 2012-06-28 Incyte Corporation N-(1-(substituted-phenyl)ethyl)-9h-purin-6-amines as pi3k inhibitors
US9527848B2 (en) 2010-12-20 2016-12-27 Incyte Holdings Corporation N-(1-(substituted-phenyl)ethyl)-9H-purin-6-amines as PI3K inhibitors
JP2014500324A (en) * 2010-12-20 2014-01-09 インサイト・コーポレイション N- (1- (substituted phenyl) ethyl) -9H-purin-6-amine as a PI3K inhibitor
US9815839B2 (en) 2010-12-20 2017-11-14 Incyte Corporation N-(1-(substituted-phenyl)ethyl)-9H-purin-6-amines as PI3K inhibitors
US9096600B2 (en) 2010-12-20 2015-08-04 Incyte Corporation N-(1-(substituted-phenyl)ethyl)-9H-purin-6-amines as PI3K inhibitors
US8809349B2 (en) 2011-01-10 2014-08-19 Infinity Pharmaceuticals, Inc. Processes for preparing isoquinolinones and solid forms of isoquinolinones
US9290497B2 (en) 2011-01-10 2016-03-22 Infinity Pharmaceuticals, Inc. Processes for preparing isoquinolinones and solid forms of isoquinolinones
WO2012097000A1 (en) 2011-01-10 2012-07-19 Pingda Ren Processes for preparing isoquinolinones and solid forms of isoquinolinones
US11312718B2 (en) 2011-01-10 2022-04-26 Infinity Pharmaceuticals, Inc. Formulations of (S)-3-(1-(9H-purin-6-ylamino)ethyl)-8-chloro-2-phenylisoquinolin-1(2H)-one
US10550122B2 (en) 2011-01-10 2020-02-04 Infinity Pharmaceuticals, Inc. Solid forms of (S)-3-(1-(9H-purin-6-ylamino)ethyl)-8-chloro-2-phenylisoquinolin-1(2H)-one and methods of use thereof
EP3581574A1 (en) 2011-01-10 2019-12-18 Infinity Pharmaceuticals, Inc. A composition for oral administration for use in the treatment of cancer, an inflammatory disease or an auto-immune disease
USRE46621E1 (en) 2011-01-10 2017-12-05 Infinity Pharmaceuticals, Inc. Processes for preparing isoquinolinones and solid forms of isoquinolinones
EP3238722A1 (en) 2011-01-10 2017-11-01 Infinity Pharmaceuticals, Inc. Solid forms of isoquinolinones
US9840505B2 (en) 2011-01-10 2017-12-12 Infinity Pharmaceuticals, Inc. Solid forms of (S)-3-(1-(9H-purin-6-ylamino)ethyl)-8-chloro-2-phenylisoquinolin-1 (2H)-one and methods of use thereof
WO2012116237A2 (en) 2011-02-23 2012-08-30 Intellikine, Llc Heterocyclic compounds and uses thereof
WO2012116237A3 (en) * 2011-02-23 2012-12-27 Intellikine, Llc Heterocyclic compounds and uses thereof
EP2678016A4 (en) * 2011-02-23 2014-11-19 Intellikine Llc Heterocyclic compounds and uses thereof
JP2014506601A (en) * 2011-02-23 2014-03-17 インテリカイン, エルエルシー Heterocyclic compounds and uses thereof
US9295673B2 (en) 2011-02-23 2016-03-29 Intellikine Llc Combination of mTOR inhibitors and P13-kinase inhibitors, and uses thereof
WO2012148540A1 (en) * 2011-02-23 2012-11-01 Intellikine, Llc Combination of kanase inhibitors and uses threof
EP2678016A2 (en) * 2011-02-23 2014-01-01 Intellikine, LLC Heterocyclic compounds and uses thereof
US9127000B2 (en) 2011-02-23 2015-09-08 Intellikine, LLC. Heterocyclic compounds and uses thereof
US9108984B2 (en) 2011-03-14 2015-08-18 Incyte Corporation Substituted diamino-pyrimidine and diamino-pyridine derivatives as PI3K inhibitors
US9126948B2 (en) 2011-03-25 2015-09-08 Incyte Holdings Corporation Pyrimidine-4,6-diamine derivatives as PI3K inhibitors
US9775841B2 (en) 2011-05-04 2017-10-03 Rhizen Pharmaceuticals Sa Compounds as modulators of protein kinases
US10172858B2 (en) 2011-05-04 2019-01-08 Intellikine Llc Combination pharmaceutical compositions and uses thereof
US10220035B2 (en) 2011-05-04 2019-03-05 Rhizen Pharmaceuticals Sa Compounds as modulators of protein kinases
US11020399B2 (en) 2011-05-04 2021-06-01 Rhizen Pharmaceuticals Sa Intermediates useful in the synthesis of compounds as modulators of protein kinases
EP2705181A1 (en) * 2011-05-04 2014-03-12 Intellikine, LLC Combination pharmaceutical compositions and uses thereof
WO2012151562A1 (en) 2011-05-04 2012-11-08 Intellikine, Llc Combination pharmaceutical compositions and uses thereof
EP2705181A4 (en) * 2011-05-04 2014-10-08 Intellikine Llc Combination pharmaceutical compositions and uses thereof
US10322130B2 (en) 2011-05-04 2019-06-18 Rhizen Pharmaceuticals Sa Substituted chromenones as modulators of protein kinases
US9233111B2 (en) 2011-07-08 2016-01-12 Novartis Ag Pyrrolo pyrimidine derivatives
US9056877B2 (en) 2011-07-19 2015-06-16 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US8969363B2 (en) 2011-07-19 2015-03-03 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
WO2013012918A1 (en) 2011-07-19 2013-01-24 Infinity Pharmaceuticals Inc. Heterocyclic compounds and uses thereof
US9605003B2 (en) 2011-07-19 2017-03-28 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
WO2013012915A1 (en) 2011-07-19 2013-01-24 Infinity Pharmaceuticals Inc. Heterocyclic compounds and uses thereof
US9718815B2 (en) 2011-07-19 2017-08-01 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9546180B2 (en) 2011-08-29 2017-01-17 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9115141B2 (en) 2011-08-29 2015-08-25 Infinity Pharmaceuticals, Inc. Substituted isoquinolinones and methods of treatment thereof
WO2013032591A1 (en) 2011-08-29 2013-03-07 Infinity Pharmaceuticals Inc. Heterocyclic compounds and uses thereof
US8785470B2 (en) 2011-08-29 2014-07-22 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
JP7384891B2 (en) 2011-09-02 2023-11-21 インサイト・ホールディングス・コーポレイション Heterocyclylamines as PI3K inhibitors
EA033646B1 (en) * 2011-09-02 2019-11-13 Incyte Holdings Corp Heterocyclylamines as pi3k inhibitors
KR20200084905A (en) * 2011-09-02 2020-07-13 인사이트 홀딩스 코포레이션 Heterocyclylamines as pi3k inhibitors
AU2019201423B2 (en) * 2011-09-02 2020-10-22 Incyte Holdings Corporation Heterocyclylamines as pi3k inhibitors
US10646492B2 (en) 2011-09-02 2020-05-12 Incyte Corporation Heterocyclylamines as PI3K inhibitors
KR102507287B1 (en) 2011-09-02 2023-03-07 인사이트 홀딩스 코포레이션 Heterocyclylamines as pi3k inhibitors
WO2013033569A1 (en) * 2011-09-02 2013-03-07 Incyte Corporation Heterocyclylamines as pi3k inhibitors
KR102249236B1 (en) 2011-09-02 2021-05-10 인사이트 홀딩스 코포레이션 Heterocyclylamines as pi3k inhibitors
KR20210054022A (en) * 2011-09-02 2021-05-12 인사이트 홀딩스 코포레이션 Heterocyclylamines as pi3k inhibitors
EP3888657A1 (en) * 2011-09-02 2021-10-06 Incyte Holdings Corporation Heterocyclylamines as pi3k inhibitors
KR101982475B1 (en) 2011-09-02 2019-05-27 인사이트 홀딩스 코포레이션 Heterocyclylamines as pi3k inhibitors
KR20190122801A (en) * 2011-09-02 2019-10-30 인사이트 홀딩스 코포레이션 Heterocyclylamines as pi3k inhibitors
US9199982B2 (en) 2011-09-02 2015-12-01 Incyte Holdings Corporation Heterocyclylamines as PI3K inhibitors
KR102371532B1 (en) 2011-09-02 2022-03-07 인사이트 홀딩스 코포레이션 Heterocyclylamines as pi3k inhibitors
US9895373B2 (en) 2011-09-02 2018-02-20 The Regents Of The University Of California Substituted pyrazolo[3,4-D]pyrimidines and uses thereof
KR20220035265A (en) * 2011-09-02 2022-03-21 인사이트 홀딩스 코포레이션 Heterocyclylamines as pi3k inhibitors
KR102030609B1 (en) 2011-09-02 2019-10-11 인사이트 홀딩스 코포레이션 Heterocyclylamines as pi3k inhibitors
EA028890B1 (en) * 2011-09-02 2018-01-31 Инсайт Холдингс Корпорейшн Heterocyclylamines as pi3k inhibitors
JP2017019851A (en) * 2011-09-02 2017-01-26 インサイト・ホールディングス・コーポレイションIncyte Holdings Corporation Heterocyclylamines as pi3k inhibitors
JP2018044008A (en) * 2011-09-02 2018-03-22 インサイト・ホールディングス・コーポレイションIncyte Holdings Corporation Heterocyclylamines as pi3k inhibitors
US10376513B2 (en) 2011-09-02 2019-08-13 Incyte Holdings Corporation Heterocyclylamines as PI3K inhibitors
EP3513793A1 (en) * 2011-09-02 2019-07-24 Incyte Holdings Corporation Heterocyclylamines as pi3k inhibitors
JP2017052805A (en) * 2011-09-02 2017-03-16 インサイト・ホールディングス・コーポレイションIncyte Holdings Corporation Heterocyclylamines as pi3k inhibitors
TWI765515B (en) * 2011-09-02 2022-05-21 美商英塞特控股公司 Heterocyclylamines as pi3k inhibitors
KR20140082680A (en) * 2011-09-02 2014-07-02 인사이트 코포레이션 Heterocyclylamines as pi3k inhibitors
KR102131612B1 (en) 2011-09-02 2020-07-08 인사이트 홀딩스 코포레이션 Heterocyclylamines as pi3k inhibitors
CN104024253A (en) * 2011-09-02 2014-09-03 因塞特公司 Heterocyclylamines as pi3k inhibitors
US11819505B2 (en) 2011-09-02 2023-11-21 Incyte Corporation Heterocyclylamines as PI3K inhibitors
US11433071B2 (en) 2011-09-02 2022-09-06 Incyte Corporation Heterocyclylamines as PI3K inhibitors
US10092570B2 (en) 2011-09-02 2018-10-09 Incyte Holdings Corporation Heterocyclylamines as PI3K inhibitors
US9321772B2 (en) 2011-09-02 2016-04-26 The Regents Of The University Of California Substituted pyrazolo[3,4-D]pyrimidines and uses thereof
CN106986867B (en) * 2011-09-02 2019-06-28 因塞特控股公司 Heterocycle amine as PI3K inhibitor
US9730939B2 (en) 2011-09-02 2017-08-15 Incyte Holdings Corporation Heterocyclylamines as PI3K inhibitors
AU2021200266B2 (en) * 2011-09-02 2022-09-29 Incyte Holdings Corporation Heterocyclylamines as pi3k inhibitors
AU2012301721B2 (en) * 2011-09-02 2017-08-10 Incyte Holdings Corporation Heterocyclylamines as PI3K inhibitors
TWI717002B (en) * 2011-09-02 2021-01-21 美商英塞特控股公司 Heterocyclylamines as pi3k inhibitors
JP2014527959A (en) * 2011-09-02 2014-10-23 インサイト・コーポレイションIncyte Corporation Heterocyclylamines as PI3K inhibitors
KR20190045381A (en) * 2011-09-02 2019-05-02 인사이트 홀딩스 코포레이션 Heterocyclylamines as pi3k inhibitors
CN106986867A (en) * 2011-09-02 2017-07-28 因塞特控股公司 It is used as the heterocyclic radical amine of PI3K inhibitor
EP3196202A1 (en) * 2011-09-02 2017-07-26 Incyte Holdings Corporation Heterocyclylamines as pi3k inhibitors
AU2017206260B2 (en) * 2011-09-02 2019-02-14 Incyte Holdings Corporation Heterocyclylamines as pi3k inhibitors
JP2019011375A (en) * 2011-09-02 2019-01-24 インサイト・ホールディングス・コーポレイションIncyte Holdings Corporation Heterocyclylamines as pi3k inhibitors
US9707233B2 (en) 2011-09-02 2017-07-18 Incyte Holdings Corporation Heterocyclylamines as PI3K inhibitors
TWI648277B (en) * 2011-09-02 2019-01-21 美商英塞特控股公司 Heterocyclic amine as PI3K inhibitor
US9630979B2 (en) 2011-09-29 2017-04-25 Infinity Pharmaceuticals, Inc. Inhibitors of monoacylglycerol lipase and methods of their use
WO2013049332A1 (en) 2011-09-29 2013-04-04 Infinity Pharmaceuticals, Inc. Inhibitors of monoacylglycerol lipase and methods of their use
WO2013045653A1 (en) 2011-09-30 2013-04-04 Oncodesign S.A. Macrocyclic flt3 kinase inhibitors
US9090630B2 (en) 2011-09-30 2015-07-28 Oncodesign S.A. Macrocyclic FLT3 kinase inhibitors
US9370519B2 (en) 2011-09-30 2016-06-21 Oncodesign S.A. Macrocyclic FLT3 kinase inhibitors
JP2014528451A (en) * 2011-10-04 2014-10-27 ギリアード カリストガ エルエルシー Novel quinoxaline inhibitor of PI3K
EP2763994A4 (en) * 2011-10-04 2015-08-26 Gilead Calistoga Llc Novel quinoxaline inhibitors of pi3k
WO2013088404A1 (en) 2011-12-15 2013-06-20 Novartis Ag Use of inhibitors of the activity or function of PI3K
US9642799B2 (en) 2012-03-13 2017-05-09 Respivert, Ltd. Crystalline 6-(2-((4-amino-3-(3-hydroxyphenyl)-1H-pyrazolo[3,4-D]pyrimidin-1-yl)methyl)-3-(2-chlorobenzyl)-4-0X0-3,4-dihydroquinazolin-5-yl)-N,N-bis(2-methoxyethyl)hex-5-ynamide
US9944646B2 (en) 2012-04-02 2018-04-17 Incyte Holdings Corporation Bicyclic azaheterocyclobenzylamines as PI3K inhibitors
US10259818B2 (en) 2012-04-02 2019-04-16 Incyte Corporation Bicyclic azaheterocyclobenzylamines as PI3K inhibitors
US9309251B2 (en) 2012-04-02 2016-04-12 Incyte Holdings Corporation Bicyclic azaheterocyclobenzylamines as PI3K inhibitors
WO2013154878A1 (en) 2012-04-10 2013-10-17 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US8940742B2 (en) 2012-04-10 2015-01-27 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9255108B2 (en) 2012-04-10 2016-02-09 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US8828998B2 (en) 2012-06-25 2014-09-09 Infinity Pharmaceuticals, Inc. Treatment of lupus, fibrotic conditions, and inflammatory myopathies and other disorders using PI3 kinase inhibitors
US9527847B2 (en) 2012-06-25 2016-12-27 Infinity Pharmaceuticals, Inc. Treatment of lupus, fibrotic conditions, and inflammatory myopathies and other disorders using PI3 kinase inhibitors
US9642855B2 (en) 2012-06-29 2017-05-09 Pfizer Inc. Substituted pyrrolo[2,3-d]pyrimidines as LRRK2 inhibitors
US9156845B2 (en) 2012-06-29 2015-10-13 Pfizer Inc. 4-(substituted amino)-7H-pyrrolo[2,3-d] pyrimidines as LRRK2 inhibitors
CN104619708A (en) * 2012-07-10 2015-05-13 武田药品工业株式会社 Azaindole derivatives which act as pi3k inhibitors
WO2014011568A1 (en) 2012-07-10 2014-01-16 Takeda Pharmaceutical Company Limited Azaindole derivatives which act as pi3k inhibitors
US8835420B2 (en) 2012-07-10 2014-09-16 Takeda Pharmaceutical Company Limited Azaindole derivatives
US10822340B2 (en) 2012-09-26 2020-11-03 The Regents Of The University Of California Substituted imidazolopyrazine compounds and methods of using same
US10131668B2 (en) 2012-09-26 2018-11-20 The Regents Of The University Of California Substituted imidazo[1,5-a]pYRAZINES for modulation of IRE1
US11613544B2 (en) 2012-09-26 2023-03-28 The Regents Of The University Of California Substituted imidazo[1,5-a]pyrazines for modulation of IRE1
WO2014071109A1 (en) 2012-11-01 2014-05-08 Infinity Pharmaceuticals, Inc. Treatment of cancers using pi3 kinase isoform modulators
EP3632442A1 (en) * 2013-03-01 2020-04-08 Incyte Holdings Corporation Use of pyrazolopyrimidine derivatives for the treatment of pi3k related disorders
KR20210110409A (en) * 2013-03-01 2021-09-07 인사이트 홀딩스 코포레이션 USE OF PYRAZOLOPYRIMIDINE DERIVATIVES FOR THE TREATMENT OF PI3Kδ RELATED DISORDERS
TWI841376B (en) 2013-03-01 2024-05-01 美商英塞特控股公司 USE OF PYRAZOLOPYRIMIDINE DERIVATIVES FOR THE TREATMENT OF PI3Kδ RELATED DISORDERS
US9932341B2 (en) 2013-03-01 2018-04-03 Incyte Corporation Use of pyrazolopyrimidine derivatives for the treatment of PI3K-delta related disorders
TWI657090B (en) * 2013-03-01 2019-04-21 英塞特控股公司 USE OF PYRAZOLOPYRIMIDINE DERIVATIVES FOR THE TREATMENT OF PI3Kδ RELATED DISORDERS
KR20150135327A (en) * 2013-03-01 2015-12-02 인사이트 코포레이션 Use of pyrazolopyrimidine derivatives for the treatment of pi3kδ related disorders
WO2014134426A1 (en) * 2013-03-01 2014-09-04 Incyte Corporation USE OF PYRAZOLOPYRIMIDINE DERIVATIVES FOR THE TREATMENT OF PI3Kδ RELATED DISORDERS
TWI736135B (en) * 2013-03-01 2021-08-11 美商英塞特控股公司 USE OF PYRAZOLOPYRIMIDINE DERIVATIVES FOR THE TREATMENT OF PI3Kδ RELATED DISORDERS
EP4233869A3 (en) * 2013-03-01 2023-11-01 Incyte Holdings Corporation Use of pyrazolopyrimidine derivatives for the treatment of pi3k related disorders
KR102298150B1 (en) * 2013-03-01 2021-09-07 인사이트 홀딩스 코포레이션 USE OF PYRAZOLOPYRIMIDINE DERIVATIVES FOR THE TREATMENT OF PI3Kδ RELATED DISORDERS
KR102586858B1 (en) 2013-03-01 2023-10-11 인사이트 홀딩스 코포레이션 USE OF PYRAZOLOPYRIMIDINE DERIVATIVES FOR THE TREATMENT OF PI3Kδ RELATED DISORDERS
IL283943B1 (en) * 2013-03-01 2023-04-01 Incyte Holdings Corp Use of pyrazolopyrimidine derivatives for the treatment of pi3k related disorders
CN105120871A (en) * 2013-03-01 2015-12-02 因赛特公司 Use of pyrazolopyrimidine derivatives for the treatment of pi3kDelta related disorders
TWI687220B (en) * 2013-03-01 2020-03-11 美商英塞特控股公司 Use of pyrazolopyrimidine derivatives for the treatment of pi3kδ related disorders
CN109010343A (en) * 2013-03-01 2018-12-18 因赛特公司 The purposes of Pyrazolopyrimidine derivative treatment PI3K δ associated disease
KR102454308B1 (en) 2013-03-01 2022-10-14 인사이트 홀딩스 코포레이션 USE OF PYRAZOLOPYRIMIDINE DERIVATIVES FOR THE TREATMENT OF PI3Kδ RELATED DISORDERS
KR20220143146A (en) * 2013-03-01 2022-10-24 인사이트 홀딩스 코포레이션 USE OF PYRAZOLOPYRIMIDINE DERIVATIVES FOR THE TREATMENT OF PI3Kδ RELATED DISORDERS
US9481667B2 (en) 2013-03-15 2016-11-01 Infinity Pharmaceuticals, Inc. Salts and solid forms of isoquinolinones and composition comprising and methods of using the same
US9745306B2 (en) 2013-03-15 2017-08-29 Respivert Limited 2-((4-amino-3-(3-fluoro-5-hydroxyphenyl)-1H-pyrazolo[3,4-D]pyrimidin-1-yl)methyl)-3-(2-(trifluoromethyl)benzyl) quinazolin-4(3H)-one derivatives and their use as phosphoinositide 3-kinase inhibitors
US9227977B2 (en) 2013-03-15 2016-01-05 Respivert Ltd. Phosphoinositide 3-kinase inhibitors
WO2014144850A1 (en) 2013-03-15 2014-09-18 Genentech, Inc. Methods of treating cancer and preventing cancer drug resistance
US9556184B2 (en) 2013-03-15 2017-01-31 Respivert, Ltd. Phosphoinositide 3-kinase inhibitors
WO2014151386A1 (en) 2013-03-15 2014-09-25 Infinity Pharmaceuticals, Inc. Salts and solid forms of isoquinolinones and composition comprising and methods of using the same
US9724354B2 (en) 2013-03-22 2017-08-08 Millennium Pharmaceuticals, Inc. Combination of catalytic mTORC1/2 inhibitors and selective inhibitors of Aurora A kinase
EP3811974A1 (en) 2013-05-30 2021-04-28 Infinity Pharmaceuticals, Inc. Treatment of cancers using pi3 kinase isoform modulators
WO2014194254A1 (en) 2013-05-30 2014-12-04 Infinity Pharmaceuticals, Inc. Treatment of cancers using pi3 kinase isoform modulators
EP3964507A1 (en) 2013-10-04 2022-03-09 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9359365B2 (en) 2013-10-04 2016-06-07 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9751888B2 (en) 2013-10-04 2017-09-05 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
WO2015051241A1 (en) 2013-10-04 2015-04-09 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US10329299B2 (en) 2013-10-04 2019-06-25 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9828377B2 (en) 2013-10-04 2017-11-28 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
WO2015051244A1 (en) 2013-10-04 2015-04-09 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
WO2015061204A1 (en) 2013-10-21 2015-04-30 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9273058B2 (en) 2013-11-14 2016-03-01 Bristol-Myers Squibb Company Substituted pyrazolo-piperazines as casein kinase 1 δ/ε inhibitors
US9695171B2 (en) 2013-12-17 2017-07-04 Pfizer Inc. 3,4-disubstituted-1 H-pyrrolo[2,3-b]pyridines and 4,5-disubstituted-7H-pyrrolo[2,3-c]pyridazines as LRRK2 inhibitors
US9371321B2 (en) 2014-01-09 2016-06-21 Astrazeneca Ab Azaindole derivatives
WO2015106012A1 (en) 2014-01-09 2015-07-16 Takeda Pharmaceutical Company Limited Azaindole derivatives
WO2015106014A1 (en) 2014-01-09 2015-07-16 Takeda Pharmaceutical Company Limited Azaindole derivatives
EP3514179A1 (en) 2014-01-24 2019-07-24 Dana-Farber Cancer Institute, Inc. Antibody molecules to pd-1 and uses thereof
EP4324518A2 (en) 2014-01-31 2024-02-21 Novartis AG Antibody molecules to tim-3 and uses thereof
US11155532B2 (en) 2014-02-13 2021-10-26 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US9994546B2 (en) 2014-02-13 2018-06-12 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US10676457B2 (en) 2014-02-13 2020-06-09 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US10300051B2 (en) 2014-02-13 2019-05-28 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US9493450B2 (en) 2014-02-13 2016-11-15 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US9670210B2 (en) 2014-02-13 2017-06-06 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US10174030B2 (en) 2014-02-13 2019-01-08 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US11247992B2 (en) 2014-02-13 2022-02-15 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US9493442B2 (en) 2014-02-13 2016-11-15 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US10513493B2 (en) 2014-02-13 2019-12-24 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US10717737B2 (en) 2014-02-13 2020-07-21 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US9527835B2 (en) 2014-02-13 2016-12-27 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
WO2015138920A1 (en) 2014-03-14 2015-09-17 Novartis Ag Antibody molecules to lag-3 and uses thereof
EP3660050A1 (en) 2014-03-14 2020-06-03 Novartis AG Antibody molecules to lag-3 and uses thereof
WO2015143012A1 (en) 2014-03-19 2015-09-24 Infinity Pharmaceuticals, Inc. Heterocyclic compounds for use in the treatment of pi3k-gamma mediated disorders
US11541059B2 (en) 2014-03-19 2023-01-03 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9775844B2 (en) 2014-03-19 2017-10-03 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US10675286B2 (en) 2014-03-19 2020-06-09 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
EP4066834A1 (en) 2014-03-19 2022-10-05 Infinity Pharmaceuticals, Inc. Heterocyclic compounds for use in the treatment of pi3k-gamma mediated disorders
US11944631B2 (en) 2014-04-16 2024-04-02 Infinity Pharmaceuticals, Inc. Combination therapies
US11110096B2 (en) 2014-04-16 2021-09-07 Infinity Pharmaceuticals, Inc. Combination therapies
WO2015160975A2 (en) 2014-04-16 2015-10-22 Infinity Pharmaceuticals, Inc. Combination therapies
WO2015160986A2 (en) 2014-04-16 2015-10-22 Infinity Pharmaceuticals, Inc. Combination therapies
WO2015168079A1 (en) 2014-04-29 2015-11-05 Infinity Pharmaceuticals, Inc. Pyrimidine or pyridine derivatives useful as pi3k inhibitors
WO2015191677A1 (en) * 2014-06-11 2015-12-17 Incyte Corporation Bicyclic heteroarylaminoalkyl phenyl derivatives as pi3k inhibitors
US10077277B2 (en) 2014-06-11 2018-09-18 Incyte Corporation Bicyclic heteroarylaminoalkyl phenyl derivatives as PI3K inhibitors
US11130767B2 (en) 2014-06-11 2021-09-28 Incyte Corporation Bicyclic heteroarylaminoalkyl phenyl derivatives as PI3K inhibitors
US10479803B2 (en) 2014-06-11 2019-11-19 Incyte Corporation Bicyclic heteroarylaminoalkyl phenyl derivatives as PI3K inhibitors
EP3409669A1 (en) 2014-06-19 2018-12-05 ARIAD Pharmaceuticals, Inc. Heteroaryl compounds for kinase inhibition
EP3778584A1 (en) 2014-06-19 2021-02-17 ARIAD Pharmaceuticals, Inc. Production process of 2-chloro-4-heteroaryl-pyrimidine derivatives
US9944639B2 (en) 2014-07-04 2018-04-17 Lupin Limited Quinolizinone derivatives as PI3K inhibitors
US9758523B2 (en) 2014-07-10 2017-09-12 Incyte Corporation Triazolopyridines and triazolopyrazines as LSD1 inhibitors
US9695180B2 (en) 2014-07-10 2017-07-04 Incyte Corporation Substituted imidazo[1,2-a]pyrazines as LSD1 inhibitors
US10640503B2 (en) 2014-07-10 2020-05-05 Incyte Corporation Imidazopyridines and imidazopyrazines as LSD1 inhibitors
US10138249B2 (en) 2014-07-10 2018-11-27 Incyte Corporation Triazolopyridines and triazolopyrazines as LSD1 inhibitors
US10047086B2 (en) 2014-07-10 2018-08-14 Incyte Corporation Imidazopyridines and imidazopyrazines as LSD1 inhibitors
US10112950B2 (en) 2014-07-10 2018-10-30 Incyte Corporation Substituted imidazo[1,2-a]pyrazines as LSD1 inhibitors
US10556908B2 (en) 2014-07-10 2020-02-11 Incyte Corporation Substituted imidazo[1,2-a]pyrazines as LSD1 inhibitors
US9695167B2 (en) 2014-07-10 2017-07-04 Incyte Corporation Substituted triazolo[1,5-a]pyridines and triazolo[1,5-a]pyrazines as LSD1 inhibitors
US10968221B2 (en) 2014-07-10 2021-04-06 Incyte Corporation Substituted [1,2,4]triazolo[1,5-a]pyrazines as LSD1 inhibitors
US9695168B2 (en) 2014-07-10 2017-07-04 Incyte Corporation Substituted imidazo[1,5-α]pyridines and imidazo[1,5-α]pyrazines as LSD1 inhibitors
US10125133B2 (en) 2014-07-10 2018-11-13 Incyte Corporation Substituted [1,2,4]triazolo[1,5-a]pyridines and substituted [1,2,4]triazolo[1,5-a]pyrazines as LSD1 inhibitors
US10117945B2 (en) 2014-09-11 2018-11-06 The Regents Of The University Of California mTORC1 inhibitors
US10646577B2 (en) 2014-09-11 2020-05-12 The Regents Of The University Of California mTORC1 inhibitors
US11452780B2 (en) 2014-09-11 2022-09-27 The Regents Of The University Of California Mtorc1 inhibitors
WO2016040880A1 (en) 2014-09-13 2016-03-17 Novartis Ag Combination therapies of alk inhibitors
EP3659621A1 (en) 2014-09-13 2020-06-03 Novartis AG Combination therapies for cancer
WO2016040892A1 (en) 2014-09-13 2016-03-17 Novartis Ag Combination therapies
EP3925622A1 (en) 2014-09-13 2021-12-22 Novartis AG Combination therapies
WO2016054555A2 (en) 2014-10-03 2016-04-07 Novartis Ag Combination therapies
EP3662903A2 (en) 2014-10-03 2020-06-10 Novartis AG Combination therapies
US9708348B2 (en) 2014-10-03 2017-07-18 Infinity Pharmaceuticals, Inc. Trisubstituted bicyclic heterocyclic compounds with kinase activities and uses thereof
WO2016054491A1 (en) 2014-10-03 2016-04-07 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US10941162B2 (en) 2014-10-03 2021-03-09 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US10253047B2 (en) 2014-10-03 2019-04-09 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
WO2016057841A1 (en) 2014-10-08 2016-04-14 Novartis Ag Compositions and methods of use for augmented immune response and cancer therapy
EP4245376A2 (en) 2014-10-14 2023-09-20 Novartis AG Antibody molecules to pd-l1 and uses thereof
WO2016061142A1 (en) 2014-10-14 2016-04-21 Novartis Ag Antibody molecules to pd-l1 and uses thereof
WO2016100882A1 (en) 2014-12-19 2016-06-23 Novartis Ag Combination therapies
US11084822B2 (en) 2015-02-27 2021-08-10 Incyte Corporation Salts and processes of preparing a PI3K inhibitor
US10336759B2 (en) 2015-02-27 2019-07-02 Incyte Corporation Salts and processes of preparing a PI3K inhibitor
US9944647B2 (en) 2015-04-03 2018-04-17 Incyte Corporation Heterocyclic compounds as LSD1 inhibitors
US10800779B2 (en) 2015-04-03 2020-10-13 Incyte Corporation Heterocyclic compounds as LSD1 inhibitors
US11401272B2 (en) 2015-04-03 2022-08-02 Incyte Corporation Heterocyclic compounds as LSD1 inhibitors
US9988401B2 (en) 2015-05-11 2018-06-05 Incyte Corporation Crystalline forms of a PI3K inhibitor
US9732097B2 (en) 2015-05-11 2017-08-15 Incyte Corporation Process for the synthesis of a phosphoinositide 3-kinase inhibitor
US10125150B2 (en) 2015-05-11 2018-11-13 Incyte Corporation Crystalline forms of a PI3K inhibitor
WO2017019894A1 (en) 2015-07-29 2017-02-02 Novartis Ag Combination therapies comprising antibody molecules to lag-3
EP3878465A1 (en) 2015-07-29 2021-09-15 Novartis AG Combination therapies comprising antibody molecules to tim-3
WO2017019897A1 (en) 2015-07-29 2017-02-02 Novartis Ag Combination therapies comprising antibody molecules to tim-3
EP3964528A1 (en) 2015-07-29 2022-03-09 Novartis AG Combination therapies comprising antibody molecules to lag-3
EP4378957A2 (en) 2015-07-29 2024-06-05 Novartis AG Combination therapies comprising antibody molecules to pd-1
US11498900B2 (en) 2015-08-12 2022-11-15 Incyte Corporation Salts of an LSD1 inhibitor
US10329255B2 (en) 2015-08-12 2019-06-25 Incyte Corporation Salts of an LSD1 inhibitor
US10723700B2 (en) 2015-08-12 2020-07-28 Incyte Corporation Salts of an LSD1 inhibitor
US10039753B2 (en) 2015-09-14 2018-08-07 Pfizer Inc. Imidazo[4,5-c]quinoline and imidazo[4,5-c][1,5]naphthyridine derivatives as LRRK2 inhibitors
US11247995B2 (en) 2015-09-14 2022-02-15 Infinity Pharmaceuticals, Inc. Solid forms of isoquinolinones, and process of making, composition comprising, and methods of using the same
US10160761B2 (en) 2015-09-14 2018-12-25 Infinity Pharmaceuticals, Inc. Solid forms of isoquinolinones, and process of making, composition comprising, and methods of using the same
US11939333B2 (en) 2015-09-14 2024-03-26 Infinity Pharmaceuticals, Inc. Solid forms of isoquinolinones, and process of making, composition comprising, and methods of using the same
EP4046655A1 (en) 2015-11-03 2022-08-24 Janssen Biotech, Inc. Antibodies specifically binding pd-1 and their uses
WO2017079115A1 (en) 2015-11-03 2017-05-11 Janssen Biotech, Inc. Antibodies specifically binding tim-3 and their uses
US10894830B2 (en) 2015-11-03 2021-01-19 Janssen Biotech, Inc. Antibodies specifically binding PD-1, TIM-3 or PD-1 and TIM-3 and their uses
WO2017079116A2 (en) 2015-11-03 2017-05-11 Janssen Biotech, Inc. Antibodies specifically binding pd-1 and tim-3 and their uses
WO2017079112A1 (en) 2015-11-03 2017-05-11 Janssen Biotech, Inc. Antibodies specifically binding pd-1 and their uses
WO2017106656A1 (en) 2015-12-17 2017-06-22 Novartis Ag Antibody molecules to pd-1 and uses thereof
US10759806B2 (en) 2016-03-17 2020-09-01 Infinity Pharmaceuticals, Inc. Isotopologues of isoquinolinone and quinazolinone compounds and uses thereof as PI3K kinase inhibitors
US11661423B2 (en) 2016-04-15 2023-05-30 Cancer Research Technology Limited Heterocyclic compounds as RET kinase inhibitors
US11548896B2 (en) 2016-04-15 2023-01-10 Cancer Research Technology Limited Heterocyclic compounds as RET kinase inhibitors
US10166221B2 (en) 2016-04-22 2019-01-01 Incyte Corporation Formulations of an LSD1 inhibitor
US10858359B2 (en) 2016-06-07 2020-12-08 Jacobio Pharmaceuticals Co., Ltd. Heterocyclic ring derivatives useful as SHP2 inhibitors
WO2017214269A1 (en) 2016-06-08 2017-12-14 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US10919914B2 (en) 2016-06-08 2021-02-16 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US11147818B2 (en) 2016-06-24 2021-10-19 Infinity Pharmaceuticals, Inc. Combination therapies
WO2017223422A1 (en) 2016-06-24 2017-12-28 Infinity Pharmaceuticals, Inc. Combination therapies
US10479770B2 (en) 2016-09-23 2019-11-19 Gilead Sciences, Inc. Phosphatidylinositol 3-kinase inhibitors
US10227350B2 (en) 2016-09-23 2019-03-12 Gilead Sciences, Inc. Phosphatidylinositol 3-kinase inhibitors
US10214519B2 (en) 2016-09-23 2019-02-26 Gilead Sciences, Inc. Phosphatidylinositol 3-kinase inhibitors
US10988466B2 (en) 2017-03-23 2021-04-27 Jacobio Pharmaceuticals Co., Ltd. Heterocyclic derivatives useful as SHP2 inhibitors
RU2760669C2 (en) * 2017-04-13 2021-11-29 Кэнсер Рисерч Текнолоджи Лимитед Inhibitory compounds
US11680068B2 (en) 2017-04-13 2023-06-20 Cancer Research Technology Limited Compounds useful as RET inhibitors
WO2018237157A1 (en) 2017-06-22 2018-12-27 Novartis Ag Antibody molecules to cd73 and uses thereof
WO2018237173A1 (en) 2017-06-22 2018-12-27 Novartis Ag Antibody molecules to cd73 and uses thereof
US11364300B2 (en) 2018-05-01 2022-06-21 Revolution Medicines, Inc. C40-, C28-, and C-32-linked rapamycin analogs as mTOR inhibitors
US11685749B2 (en) 2018-05-01 2023-06-27 Revolution Medicines, Inc. C26-linked rapamycin analogs as mTOR inhibitors
US10980889B1 (en) 2018-05-01 2021-04-20 Revolution Medicines, Inc. C40-, C28-, and C-32-linked rapamycin analogs as mTOR inhibitors
US11746157B2 (en) 2018-05-24 2023-09-05 Janssen Biotech, Inc. PSMA binding agents and uses thereof
WO2019224718A2 (en) 2018-05-24 2019-11-28 Janssen Biotech, Inc. Psma binding agents and uses thereof
WO2019229658A1 (en) 2018-05-30 2019-12-05 Novartis Ag Entpd2 antibodies, combination therapies, and methods of using the antibodies and combination therapies
WO2019232244A2 (en) 2018-05-31 2019-12-05 Novartis Ag Antibody molecules to cd73 and uses thereof
US10968200B2 (en) 2018-08-31 2021-04-06 Incyte Corporation Salts of an LSD1 inhibitor and processes for preparing the same
US11512064B2 (en) 2018-08-31 2022-11-29 Incyte Corporation Salts of an LSD1 inhibitor and processes for preparing the same
US11111247B2 (en) 2018-09-25 2021-09-07 Incyte Corporation Pyrazolopyrimidine compounds and uses thereof
US11591395B2 (en) 2019-04-19 2023-02-28 Janssen Biotech, Inc. Methods of treating prostate cancer with an anti-PSMA/CD3 antibody
WO2021053559A1 (en) 2019-09-18 2021-03-25 Novartis Ag Entpd2 antibodies, combination therapies, and methods of using the antibodies and combination therapies
WO2022162518A2 (en) 2021-01-28 2022-08-04 Janssen Biotech, Inc. Psma binding proteins and uses thereof
WO2023016477A1 (en) * 2021-08-11 2023-02-16 Taizhou Eoc Pharma Co., Ltd. A cyclin-dependent kinase inhibitor
US11999751B2 (en) 2021-08-24 2024-06-04 Incyte Corporation Bicyclic heteroarylaminoalkyl phenyl derivatives as PI3K inhibitors
WO2023186065A1 (en) * 2022-04-02 2023-10-05 Taizhou Eoc Pharma Co., Ltd. A cyclin-dependent kinase inhibitor

Also Published As

Publication number Publication date
US20140206685A1 (en) 2014-07-24
EP2346508B1 (en) 2016-08-24
US20110281866A1 (en) 2011-11-17
US9790228B2 (en) 2017-10-17
EP2346508A1 (en) 2011-07-27
CA2738429A1 (en) 2010-04-01
JP5731978B2 (en) 2015-06-10
US9296742B2 (en) 2016-03-29
CA2738429C (en) 2016-10-25
US20160168157A1 (en) 2016-06-16
EP2346508A4 (en) 2012-04-18
US8703778B2 (en) 2014-04-22
JP2012503655A (en) 2012-02-09

Similar Documents

Publication Publication Date Title
US11433065B2 (en) Certain chemical entities, compositions and methods
EP3009436B1 (en) Kinase inhibitors and methods of use
CA2738429C (en) Heterocyclic kinase inhibitors
EP2427195B1 (en) Heterocyclic compounds and uses thereof
US9637492B2 (en) Benzothiazole kinase inhibitors and methods of use
EP2240451B1 (en) Isoquinolinone derivatives substituted with a purine useful as PI3K inhibitors
EP2252293B1 (en) Kinase inhibitors and methods of use
WO2009046448A1 (en) Chemical entities and therapeutic uses thereof
AU2015201582B2 (en) Certain chemical entities, compositions and methods

Legal Events

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

Ref document number: 09816603

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2738429

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2011529032

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

REEP Request for entry into the european phase

Ref document number: 2009816603

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2009816603

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 13121157

Country of ref document: US