WO2010059788A1 - Composés inhibiteurs de pi3k de type pyrazolopyridine et leurs procédés d'utilisation - Google Patents

Composés inhibiteurs de pi3k de type pyrazolopyridine et leurs procédés d'utilisation Download PDF

Info

Publication number
WO2010059788A1
WO2010059788A1 PCT/US2009/065085 US2009065085W WO2010059788A1 WO 2010059788 A1 WO2010059788 A1 WO 2010059788A1 US 2009065085 W US2009065085 W US 2009065085W WO 2010059788 A1 WO2010059788 A1 WO 2010059788A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
alkyl
heteroaryl
heterocyclyl
alkylene
Prior art date
Application number
PCT/US2009/065085
Other languages
English (en)
Inventor
Jennafer Dotson
Tim Heffron
Alan G. Olivero
Daniel P. Sutherlin
Steven Staben
Shumei Wang
Bing-Yan Zhu
Irina S. Chuckowree
Adrian J. Folkes
Nan Chi Wan
Original Assignee
Genentech, Inc.
F. Hoffmann-La Roche Ag
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 Genentech, Inc., F. Hoffmann-La Roche Ag filed Critical Genentech, Inc.
Priority to CN200980146318.8A priority Critical patent/CN102224152B/zh
Priority to SG2011036381A priority patent/SG171765A1/en
Priority to JP2011537609A priority patent/JP5689069B2/ja
Priority to ES09761108T priority patent/ES2392488T3/es
Priority to CA2744154A priority patent/CA2744154A1/fr
Priority to US13/129,091 priority patent/US8653098B2/en
Priority to EP09761108A priority patent/EP2356117B1/fr
Publication of WO2010059788A1 publication Critical patent/WO2010059788A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • 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
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system

Definitions

  • the invention relates generally to compounds with anti-cancer activity or anti- inflammation effects, and more specifically to compounds which inhibit PB kinase activity.
  • the invention also relates to methods of using the compounds for in vitro, in situ, and in vivo diagnosis or treatment of mammalian cells, or associated pathological conditions.
  • Phosphatidylinositol (hereinafter abbreviated as "PI") is one of a number of phospholipids found in cell membranes.
  • PI Phosphatidylinositol
  • 3'-phosphorylated phosphoinositides has been implicated in a variety of cellular processes, e.g., malignant transformation, growth factor signaling, inflammation, and immunity (Rameh et al (1999) J. Biol Chem, 274:8347-8350).
  • phosphatidylinositol 3-kinase also referred to as PI 3-kinase or PI3K
  • PI 3-kinase PI 3-kinase
  • Phosphoinositide 3-kinases are lipid kinases that phosphorylate lipids at the 3-hydroxyl residue of an inositol ring (Whitman et al (1988) Nature, 332:664).
  • the 3- phosphorylated phospholipids (PIP3s) generated by P 13 -kinases act as second messengers recruiting kinases with lipid binding domains (including plekstrin homology (PH) regions), such as Akt and phosphoinositide-dependent kinase-1 (PDKl).
  • Akt and PDKl are important in the regulation of many cellular processes including cell cycle regulation, proliferation, survival, apoptosis and motility and are significant components of the molecular mechanisms of diseases such as cancer, diabetes and immune inflammation (Vivanco et al (2002) Nature Rev. Cancer 2:489; Phillips et al (1998) Cancer 83:41).
  • the main PI3-kinase isoform in cancer is the Class I PI3-kinase, pi 10 ⁇
  • PI3 kinase/ Akt/PTEN pathway is an attractive target for cancer drug development since such agents would be expected to inhibit proliferation, reverse the repression of apoptosis and surmount resistance to cytotoxic agents in cancer cells (Garcia- Echeverria et al (2008) Oncogene 27:5511-5526). PI3 kinase inhibitors have been reported (Folkes et al (2008) Jour. Med. Chem.
  • the invention relates generally to pyrazolo[3,4-b]pyridine compounds of
  • Formula I with anti-cancer activity, and more specifically with PI3 kinase inhibitory activity.
  • Certain hyperproliferative disorders are characterized by the modulation of PI3 kinase function, for example by mutations or overexpression of the proteins.
  • the compounds of the invention may be useful in the treatment of hyperproliferative disorders such as cancer or inflammation related disorders such as rheumatoid arthritis.
  • the compounds may inhibit tumor growth in mammals and may be useful for treating human cancer patients.
  • the invention also relates to methods of using the pyrazolo[3,4-b]pyridines compounds of Formula I for in vitro, in situ, and in vivo diagnosis or treatment of mammalian cells, organisms, or associated pathological conditions.
  • Formula I compounds include:
  • Another aspect of the invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a 1,3,4,6-substituted pyrazolo[3,4- ⁇ ]pyridine compound of Formula I and a pharmaceutically acceptable carrier.
  • the pharmaceutical composition may further comprise one or more additional therapeutic agent.
  • Another aspect of the invention provides methods of inhibiting PI3 kinase activity, comprising contacting a PI3 kinase with an effective inhibitory amount of a compound of Formula I.
  • Another aspect of the invention provides methods of preventing or treating a hyperproliferative disease or disorder modulated by PI3 kinases, comprising administering to a mammal in need of such treatment an effective amount of a compound of Formula I.
  • hyperproliferative disease or disorder include, but are not limited to, cancer.
  • Another aspect of the invention provides methods of preventing or treating a hyperproliferative disorder, comprising administering to a mammal in need of such treatment an effective amount of a compound of Formula I, alone or in combination with one or more additional compounds having anti-hyperproliferative properties.
  • Another aspect of the invention provides methods of preventing or treating a inflammation disorder, comprising administering to a mammal in need of such treatment an effective amount of a compound of Formula I, alone or in combination with one or more additional compounds having anti-hyperproliferative properties.
  • the present invention provides a method of using a compound of this invention to treat a hyperproliferative disease or condition modulated by
  • An additional aspect of the invention is the use of a compound of this invention in the preparation of a medicament for the treatment or prevention of a disease or condition modulated by PB kinase in a mammal.
  • kits comprising a compound of
  • Formula I a container, and optionally a package insert or label indicating a treatment.
  • Another aspect of the invention includes methods of preparing, methods of separating, and methods of purifying compounds of Formula I.
  • Another aspect of the invention includes novel intermediates useful for preparing Formula I compounds.
  • alkyl refers to a saturated linear or branched-chain monovalent hydrocarbon radical of one to twelve carbon atoms (C 1 -C 12 ), wherein the alkyl radical may be optionally substituted independently with one or more substituents described below.
  • an alkyl radical is one to eight carbon atoms (C 1 -Cs), or one to six carbon atoms (C 1 -C 6 ).
  • alkyl groups include, but are not limited to, methyl (Me, -CH 3 ), ethyl (Et, -CH 2 CH 3 ), 1 -propyl (n-Pr, n-propyl, -CH 2 CH 2 CH 3 ), 2-propyl (i-Pr, i-propyl, -CH(CH 3 ) 2 ), 1 -butyl (n-Bu, n-butyl, -CH 2 CH 2 CH 2 CH 3 ), 2-methyl-l -propyl (i- Bu, i-butyl, -CH 2 CH(CH 3 ) 2 ), 2-butyl (s-Bu, s-butyl, -CH(CH 3 )CH 2 CH 3 ), 2-methyl-2-propyl (t-Bu, t-butyl, -C(CHs) 3 ), 1-pentyl (n-pentyl, -CH 2 CH 2 CH 2 CH 3 ), 2-pentyl
  • alkylene refers to a saturated linear or branched- chain divalent hydrocarbon radical of one to twelve carbon atoms (C 1 -C 12 ), wherein the alkylene radical may be optionally substituted independently with one or more substituents described below.
  • an alkylene radical is one to eight carbon atoms (C 1 -Cs), or one to six carbon atoms (C 1 -C 6 ).
  • alkylene groups include, but are not limited to, methylene (-CH 2 -), ethylene (-CH 2 CH 2 -), propylene (-CH 2 CH 2 CH 2 -), and the like.
  • alkynyl refers to a linear or branched monovalent hydrocarbon radical of two to eight carbon atoms (C 2 -Cs) with at least one site of unsaturation, i.e., a carbon-carbon, sp triple bond, wherein the alkynyl radical may be optionally substituted. Examples include, but are not limited to, ethynyl (-C ⁇ CH), propynyl (propargyl, -CH 2 C ⁇ CH), and the like.
  • alkynylene refers to a linear or branched divalent hydrocarbon radical of two to eight carbon atoms (C 2 -Cs) with at least one site of unsaturation, i.e., a carbon-carbon, sp triple bond, wherein the alkynyl radical may be optionally. Examples include, but are not limited to, ethynylene (-C ⁇ C-), propynylene (propargylene, -CH 2 C ⁇ C-), and the like.
  • carrier refers to a monovalent non-aromatic, saturated or partially unsaturated ring having 3 to 12 carbon atoms (Cs-C 12 ) as a monocyclic ring or 7 to 12 carbon atoms as a bicyclic ring.
  • Bicyclic carbocycles having 7 to 12 atoms can be arranged, for example, as a bicyclo [4,5], [5,5], [5,6] or [6,6] system, and bicyclic carbocycles having 9 or 10 ring atoms can be arranged as a bicyclo [5,6] or [6,6] system, or as bridged systems such as bicyclo[2.2.1]heptane, bicyclo[2.2.2]octane and bicyclo[3.2.2]nonane.
  • monocyclic carbocycles include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, 1-cyclopent-l-enyl, l-cyclopent-2-enyl, l-cyclopent-3-enyl, cyclohexyl, 1-cyclohex-l-enyl, l-cyclohex-2-enyl, l-cyclohex-3-enyl, cyclohexadienyl, cycloheptyl, cyclooctyl, cyclononyl, cyclodecyl, cycloundecyl, cyclododecyl, and the like.
  • Aryl means a monovalent aromatic hydrocarbon radical of 6-20 carbon atoms (C 6 -C 2O ) derived by the removal of one hydrogen atom from a single carbon atom of a parent aromatic ring system. Some aryl groups are represented in the exemplary structures as "Ar”. Aryl includes bicyclic radicals comprising an aromatic ring fused to a saturated, partially unsaturated ring, or aromatic carbocyclic ring.
  • Typical aryl groups include, but are not limited to, radicals derived from benzene (phenyl), substituted benzenes, naphthalene, anthracene, biphenyl, indenyl, indanyl, 1 ,2-dihydronaphthalene, 1,2,3,4-tetrahydronaphthyl, and the like.
  • Aryl groups are optionally substituted.
  • “Arylene” means a divalent aromatic hydrocarbon radical of 6-20 carbon atoms (C 6 -C 2 O) derived by the removal of two hydrogen atom from a two carbon atoms of a parent aromatic ring system.
  • Arylene includes bicyclic radicals comprising an aromatic ring fused to a saturated, partially unsaturated ring, or aromatic carbocyclic ring.
  • Typical arylene groups include, but are not limited to, radicals derived from benzene (phenylene), substituted benzenes, naphthalene, anthracene, biphenylene, indenylene, indanylene, 1 ,2- dihydronaphthalene, 1,2,3,4-tetrahydronaphthyl, and the like.
  • Arylene groups are optionally substituted
  • a heterocycle may be a monocycle having 3 to 7 ring members (2 to 6 carbon atoms and 1 to 4 heteroatoms selected from N, O, P, and S) or a bicycle having 7 to 10 ring members (4 to 9 carbon atoms and 1 to 6 heteroatoms selected from N, O, P, and S), for example: a bicyclo [4,5], [5,5], [5,6], or [6,6] system.
  • Heterocycles are described in Paquette, Leo A.; "Principles of Modern Heterocyclic Chemistry" (W. A.
  • Heterocyclyl also includes radicals where heterocycle radicals are fused with a saturated, partially unsaturated ring, or aromatic carbocyclic or heterocyclic ring.
  • heterocyclic rings include, but are not limited to, pyrrolidinyl, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, tetrahydropyranyl, dihydropyranyl, tetrahydrothiopyranyl, piperidino, morpholino, thiomorpholino, thioxanyl, piperazinyl, homopiperazinyl, azetidinyl, oxetanyl, thietanyl, homopiperidinyl, oxepanyl, thiepanyl, oxazepinyl, diazepinyl, thiazepinyl, 2-pyrrolinyl, 3-pyrrolinyl, indolinyl, 2H-pyranyl, 4H- pyranyl, dioxanyl, 1,3-dioxolanyl, pyrazolinyl, dithianyl, dithiola
  • Spiro moieties are also included within the scope of this definition.
  • heteroaryl refers to a monovalent aromatic radical of 5-, 6-, or 7- membered rings, and includes fused ring systems (at least one of which is aromatic) of 5 to about 20 ring atoms, containing one or more heteroatoms independently selected from nitrogen, oxygen, and sulfur.
  • heteroaryl groups are pyridinyl (including, for example, 2-hydroxypyridinyl), imidazolyl, imidazopyridinyl, pyrimidinyl (including, for example, 4-hydroxypyrimidinyl), pyrazolyl, triazolyl, pyrazinyl, tetrazolyl, furyl, thienyl, isoxazolyl, thiazolyl, oxadiazolyl, oxazolyl, isothiazolyl, pyrrolyl, quinolinyl, isoquinolinyl, tetrahydroisoquinolinyl, indolyl, benzimidazolyl, benzofuranyl, cinnolinyl, indazolyl, indolizinyl, phthalazinyl, pyridazinyl, triazinyl, isoindolyl, pteridinyl, purinyl, oxadiazol
  • the heterocycle or heteroaryl groups may be carbon (carbon-linked), or nitrogen (nitrogen-linked) bonded to the pyrazolo[3,4-b]pyridine where such is possible.
  • carbon bonded heterocycles or heteroaryls are bonded at position 2, 3, 4, 5, or 6 of a pyridine, position 3, 4, 5, or 6 of a pyridazine, position 2, 4, 5, or 6 of a pyrimidine, position 2, 3, 5, or 6 of a pyrazine, position 2, 3, 4, or 5 of a furan, tetrahydrofuran, thiofuran, thiophene, pyrrole or tetrahydropyrrole, position 2, 4, or 5 of an oxazole, imidazole or thiazole, position 3, 4, or 5 of an isoxazole, pyrazole, or isothiazole, position 2 or 3 of an aziridine, position 2, 3, or 4 of an azetidine, position 2, 3,
  • nitrogen bonded heterocycles or heteroaryls are bonded at position 1 of an aziridine, azetidine, pyrrole, pyrrolidine, 2- pyrroline, 3-pyrroline, imidazole, imidazolidine, 2-imidazoline, 3 -imidazoline, pyrazole, pyrazoline, 2-pyrazoline, 3-pyrazoline, piperidine, piperazine, indole, indoline, lH-indazole, position 2 of a isoindole, or isoindoline, position 4 of a morpholine, and position 9 of a carbazole, or ⁇ -carboline.
  • monocyclic heteroaryl refers to a five- or six-membered, unsubstituted or substituted, monocyclic heteroaryl radical which contains 1, 2, 3 or 4 ring heteroatoms independently selected from N, O and S. Any carbon (carbon- linked) atom of the monocyclic heteroaryl may be attached to the C-4 position as R 4 or at the C-6 position as R of the pyridine ring according to Formula I.
  • Monocyclic heteroaryl radicals include, but are not limited to: 2-pyridyl, 3-pyridyl, 4-pyridyl, 3-isoxazolyl, 4-isoxazolyl, 5-isoxazolyl, 2- imidazolyl, 4-imidazolyl, 3-pyrazolyl, 4-pyrazolyl, 2-pyrrolyl, 3-pyrrolyl, 2-thiazolyl, 4- thiazolyl, 5-thiazolyl, 3-pyridazinyl, 4-pyridazinyl, 5-pyridazinyl, 2-pyrimidinyl, 5- pyrimidinyl, 6-pyrimidinyl, 2-pyrazinyl, 2-oxazolyl, 4-oxazolyl, 5-oxazolyl, 2-furanyl, 3- furanyl, 2-thienyl, 3-thienyl, 3-triazolyl, 1-triazolyl, 5-tetrazolyl, 1-tetrazolyl, and 2- tetrazolyl
  • Fused bicyclic heterocyclyl and heteroaryl radicals include, but are not limited to: lH-indazole, lH-indole, indolin-2-one, l-(indolin-l-yl)ethanone, IH- benzo[d][l,2,3]triazole, lH-pyrazolo[3,4-b]pyridine, lH-pyrazolo[3,4-d]pyrimidine, IH- benzo[d]imidazole, lH-benzo[d]imidazol-2(3H)-one, lH-pyrazolo[3,4-c]pyridine, IH- pyrrolo[2,3-c]pyridine, 3H-imidazo[4,5-c]pyridine, 7H-pyrrolo[2,3-d]pyrimidine, 7H-purine, lH-pyrazolo[4,3-d]pyrimidine, 5H-pyrrolo[3,2-d]pyrimidine,
  • beneficial or desired clinical results include, but are not limited to, alleviation of symptoms, diminishment of extent of disease, stabilized (i.e., not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, and remission (whether partial or total), whether detectable or undetectable.
  • Treatment can also mean prolonging survival as compared to expected survival if not receiving treatment.
  • Those in need of treatment include those already with the condition or disorder as well as those prone to have the condition or disorder or those in which the condition or disorder is to be prevented.
  • the phrase "therapeutically effective amount” means an amount of a compound of the present invention that (i) treats or prevents the particular disease, condition, or disorder, (ii) attenuates, ameliorates, or eliminates one or more symptoms of the particular disease, condition, or disorder, or (iii) prevents or delays the onset of one or more symptoms of the particular disease, condition, or disorder described herein.
  • the therapeutically effective amount of the drug may reduce the number of cancer cells; reduce the tumor size; inhibit (i.e., slow to some extent and preferably stop) cancer cell infiltration into peripheral organs; inhibit (i.e., slow to some extent and preferably stop) tumor metastasis; inhibit, to some extent, tumor growth; and/or relieve to some extent one or more of the symptoms associated with the cancer.
  • the drug may prevent growth and/or kill existing cancer cells, it may be cytostatic and/or cytotoxic.
  • efficacy can be measured, for example, by assessing the time to disease progression (TTP) and/or determining the response rate (RR).
  • cancer and “cancerous” refer to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth.
  • a “tumor” comprises one or more cancerous cells. Examples of cancer include, but are not limited to, carcinoma, lymphoma, blastoma, sarcoma, and leukemia or lymphoid malignancies.
  • squamous cell cancer e.g., epithelial squamous cell cancer
  • lung cancer including small- cell lung cancer, non-small cell lung cancer ("NSCLC"), adenocarcinoma of the lung and squamous carcinoma of the lung, cancer of the peritoneum, hepatocellular cancer, gastric or stomach cancer including gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatoma, breast cancer, colon cancer, rectal cancer, colorectal cancer, endometrial or uterine carcinoma, salivary gland carcinoma, kidney or renal cancer, prostate cancer, vulval cancer, thyroid cancer, hepatic carcinoma, anal carcinoma, penile carcinoma, as well as head and neck cancer.
  • NSCLC non-small cell lung cancer
  • adenocarcinoma of the lung and squamous carcinoma of the lung cancer of the peritoneum, hepatocellular cancer, gastric or stomach cancer including gastrointestinal cancer
  • a "chemotherapeutic agent” is a chemical compound useful in the treatment of cancer, regardless of mechanism of action.
  • Classes of chemotherapeutic agents include, but are not limited to: alkyating agents, antimetabolites, spindle poison plant alkaloids, cytoxic/antitumor antibiotics, topoisomerase inhibitors, antibodies, photosensitizers, and kinase inhibitors.
  • Chemotherapeutic agents include compounds used in "targeted therapy” and conventional chemotherapy.
  • chemotherapeutic agents include: erlotinib (TARCEVA®, Genentech/OSI Pharm.), docetaxel (TAXOTERE®, Sanofi-Aventis), 5-FU (fluorouracil, 5-fluorouracil, CAS No. 51-21-8), gemcitabine (GEMZAR®, Lilly), PD- 0325901 (CAS No. 391210-10-9, Pfizer), cisplatin (cis-diamine,dichloroplatinum(II), CAS No. 15663-27-1), carboplatin (CAS No.
  • paclitaxel TAXOL®, Bristol-Myers Squibb Oncology, Princeton, N.J.
  • trastuzumab HERCEPTIN®, Genentech
  • temozolomide 4-methyl-5-oxo- 2,3,4,6,8-pentazabicyclo [4.3.0] nona-2,7,9-triene- 9-carboxamide, CAS No.
  • chemotherapeutic agents include: oxaliplatin
  • dynemicin dynemicin A
  • bisphosphonates such as clodronate
  • an esperamicin as well as neocarzinostatin chromophore and related chromoprotein enediyne antibiotic chromophores
  • aclacinomysins actinomycin, authramycin, azaserine, bleomycins, cactinomycin, carabicin, carminomycin, carzinophilin, chromomycinis, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, morpholino-doxorubicin, cyanomorpholino-doxorubicin, 2-pyrrolino-doxorubicin and deoxy doxorubicin), epirubicin, esorubicin, idarubicin,
  • chemotherapeutic agent also included in the definition of "chemotherapeutic agent” are: (i) anti- hormonal agents that act to regulate or inhibit hormone action on tumors such as anti- estrogens and selective estrogen receptor modulators (SERMs), including, for example, tamoxifen (including NOLVADEX®; tamoxifen citrate), raloxifene, droloxifene, A- hydroxytamoxifen, trioxifene, keoxifene, LYl 17018, onapristone, and FARESTON® (toremifme citrate); (ii) aromatase inhibitors that inhibit the enzyme aromatase, which regulates estrogen production in the adrenal glands, such as, for example, 4(5)-imidazoles, aminoglutethimide, MEGASE® (megestrol acetate), AROMASIN® (exemestane; Pfizer), formestanie, fadrozole, RIVISOR® (vorozole), FEMARA
  • chemotherapeutic agent therapeutic antibodies such as alemtuzumab (Campath), bevacizumab (AVASTIN®, Genentech); cetuximab (ERBITUX®, Imclone); panitumumab (VECTIB IX®, Amgen), rituximab (RITUXAN®, Genentech/Biogen pie), pertuzumab (OMNITARGTM, 2C4, Genentech), trastuzumab (HERCEPTIN®, Genentech), tositumomab (Bexxar, Corixia), and the antibody drug conjugate, gemtuzumab ozogamicin (MYLOTARG®, Wyeth).
  • therapeutic antibodies such as alemtuzumab (Campath), bevacizumab (AVASTIN®, Genentech); cetuximab (ERBITUX®, Imclone); panitumumab (VECTIB IX®, Amgen), rituximab
  • Humanized monoclonal antibodies with therapeutic potential as chemotherapeutic agents in combination with the PDK inhibitors of the invention include: alemtuzumab, apolizumab, aselizumab, atlizumab, bapineuzumab, bevacizumab, bivatuzumab mertansine, cantuzumab mertansine, cedelizumab, certolizumab pegol, cidfusituzumab, cidtuzumab, daclizumab, eculizumab, efalizumab, epratuzumab, erlizumab, felvizumab, fontolizumab, gemtuzumab ozogamicin, inotuzumab ozogamicin, ipilimumab, labetuzumab, lintuzumab, matuzumab, mepolizumab, motavizumab,
  • a "metabolite” is a product produced through metabolism in the body of a specified compound or salt thereof. Metabolites of a compound may be identified using routine techniques known in the art and their activities determined using tests such as those described herein. Such products may result for example from the oxidation, reduction, hydrolysis, amidation, deamidation, esterif ⁇ cation, deesterif ⁇ cation, enzymatic cleavage, and the like, of the administered compound. Accordingly, the invention includes metabolites of compounds of the invention, including compounds produced by a process comprising contacting a compound of this invention with a mammal for a period of time sufficient to yield a metabolic product thereof.
  • package insert is used to refer to instructions customarily included in commercial packages of therapeutic products, that contain information about the indications, usage, dosage, administration, contraindications and/or warnings concerning the use of such therapeutic products.
  • stereoisomers refers to compounds which have identical chemical constitution, but differ with regard to the arrangement of the atoms or groups in space.
  • Diastereomer refers to a stereoisomer with two or more centers of chirality and whose molecules are not mirror images of one another. Diastereomers have different physical properties, e.g. melting points, boiling points, spectral properties, and reactivities.
  • Mixtures of diastereomers may separate under high resolution analytical procedures such as electrophoresis and chromatography.
  • Enantiomers refer to two stereoisomers of a compound which are non- superimposable mirror images of one another.
  • the compounds of the invention may contain asymmetric or chiral centers, and therefore exist in different stereoisomeric forms. It is intended that all stereoisomeric forms of the compounds of the invention, including but not limited to, diastereomers, enantiomers and atropisomers, as well as mixtures thereof such as racemic mixtures, form part of the present invention.
  • optically active compounds i.e., they have the ability to rotate the plane of plane -polarized light.
  • the prefixes D and L, or R and S are used to denote the absolute configuration of the molecule about its chiral center(s).
  • the prefixes d and 1 or (+) and (-) are employed to designate the sign of rotation of plane-polarized light by the compound, with (-) or 1 meaning that the compound is levorotatory.
  • a compound prefixed with (+) or d is dextrorotatory.
  • these stereoisomers are identical except that they are mirror images of one another.
  • a specific stereoisomer may also be referred to as an enantiomer, and a mixture of such isomers is often called an enantiomeric mixture.
  • a 50:50 mixture of enantiomers is referred to as a racemic mixture or a racemate, which may occur where there has been no stereoselection or stereospecificity in a chemical reaction or process.
  • racemic mixture and “racemate” refer to an equimolar mixture of two enantiomeric species, devoid of optical activity.
  • tautomer or “tautomeric form” refers to structural isomers of different energies which are interconvertible via a low energy barrier.
  • proton tautomers also known as prototropic tautomers
  • proton tautomers include interconversions via migration of a proton, such as keto-enol and imine-enamine isomerizations.
  • Valence tautomers include interconversions by reorganization of some of the bonding electrons.
  • pharmaceutically acceptable salt refers to pharmaceutically acceptable organic or inorganic salts of a compound of the invention.
  • Exemplary salts include, but are not limited, to sulfate, citrate, acetate, oxalate, chloride, bromide, iodide, nitrate, bisulfate, phosphate, acid phosphate, isonicotinate, lactate, salicylate, acid citrate, tartrate, oleate, tannate, pantothenate, bitartrate, ascorbate, succinate, maleate, gentisinate, fumarate, gluconate, glucuronate, saccharate, formate, benzoate, glutamate, methanesulfonate "mesylate", ethanesulfonate, benzenesulfonate, /?-toluenesulfonate, and pamoate (i.e., l,l'-methylene-bis(2-hydroxy-3-naphthoate)) salts.
  • sulfate citrate, acetate, oxa
  • a pharmaceutically acceptable salt may involve the inclusion of another molecule such as an acetate ion, a succinate ion or other counter ion.
  • the counter ion may be any organic or inorganic moiety that stabilizes the charge on the parent compound.
  • a pharmaceutically acceptable salt may have more than one charged atom in its structure. Instances where multiple charged atoms are part of the pharmaceutically acceptable salt can have multiple counter ions. Hence, a pharmaceutically acceptable salt can have one or more charged atoms and/or one or more counter ion.
  • the desired pharmaceutically acceptable salt may be prepared by any suitable method available in the art, for example, treatment of the free base with an inorganic acid, such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, methanesulfonic acid, phosphoric acid and the like, or with an organic acid, such as acetic acid, trifluoroacetic acid, maleic acid, succinic acid, mandelic acid, fumaric acid, malonic acid, pyruvic acid, oxalic acid, glycolic acid, salicylic acid, a pyranosidyl acid, such as glucuronic acid or galacturonic acid, an alpha hydroxy acid, such as citric acid or tartaric acid, an amino acid, such as aspartic acid or glutamic acid, an aromatic acid, such as benzoic acid or cinnamic acid, a sulfonic acid, such as p-toluenesulf
  • an inorganic acid such as hydrochloric acid
  • the desired pharmaceutically acceptable salt may be prepared by any suitable method, for example, treatment of the free acid with an inorganic or organic base, such as an amine (primary, secondary or tertiary), an alkali metal hydroxide or alkaline earth metal hydroxide, or the like.
  • an inorganic or organic base such as an amine (primary, secondary or tertiary), an alkali metal hydroxide or alkaline earth metal hydroxide, or the like.
  • Suitable salts include, but are not limited to, organic salts derived from amino acids, such as glycine and arginine, ammonia, primary, secondary, and tertiary amines, and cyclic amines, such as piperidine, morpholine and piperazine, and inorganic salts derived from sodium, calcium, potassium, magnesium, manganese, iron, copper, zinc, aluminum and lithium.
  • organic salts derived from amino acids such as glycine and arginine, ammonia, primary, secondary, and tertiary amines, and cyclic amines, such as piperidine, morpholine and piperazine
  • inorganic salts derived from sodium, calcium, potassium, magnesium, manganese, iron, copper, zinc, aluminum and lithium.
  • a “solvate” refers to an association or complex of one or more solvent molecules and a compound of the invention.
  • solvents that form solvates include, but are not limited to, water, isopropanol, ethanol, methanol, DMSO, ethyl acetate, acetic acid, and ethanolamine.
  • hydrate refers to the complex where the solvent molecule is water.
  • protecting group refers to a substituent that is commonly employed to block or protect a particular functionality while reacting other functional groups on the compound.
  • an “amino-protecting group” is a substituent attached to an amino group that blocks or protects the amino functionality in the compound.
  • Suitable amino- protecting groups include acetyl, trifluoroacetyl, t-butoxycarbonyl (BOC), benzyloxycarbonyl (CBZ) and 9-fluorenylmethylenoxycarbonyl (Fmoc).
  • a "hydroxy-protecting group” refers to a substituent of a hydroxy group that blocks or protects the hydroxy functionality. Suitable protecting groups include acetyl and silyl.
  • a “carboxy-protecting group” refers to a substituent of the carboxy group that blocks or protects the carboxy functionality.
  • Common carboxy-protecting groups include phenylsulfonylethyl, cyanoethyl, 2-(trimethylsilyl)ethyl, 2-(trimethylsilyl)ethoxymethyl, 2-(p-toluenesulfonyl)ethyl, 2-(p- nitrophenylsulfenyl)ethyl, 2-(diphenylphosphino)-ethyl, nitroethyl and the like.
  • protecting groups and their use see T. W. Greene, Protective Groups in Organic Synthesis, John Wiley & Sons, New York, 1991.
  • the present invention provides pyrazolo[3,4-b]pyridine compounds, and pharmaceutical formulations thereof, which are potentially useful in the treatment of diseases, conditions and/or disorders modulated by PB kinases. More specifically, the present invention provides compounds of Formula I
  • R > 2 z is Ci-Ci 2 alkyl
  • R 10 , R 11 and R 12 are independently selected from H, Ci-Ci 2 alkyl, C 2 -C 8 alkenyl, C 2 - C 8 alkynyl, C 3 -Ci 2 carbocyclyl, C 2 -C 20 heterocyclyl, C 6 -C 20 aryl, and Ci-C 20 heteroaryl, where alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl, and heteroaryl are optionally substituted with one or more groups independently selected from F, Cl, Br, I, -CH 2 OH, - CH 2 C 6 H 5 , -CN, -CF 3 , -CO 2 H, -CONH 2 , -CONHCH 3 , -NO 2 , -N(CH 3 ) 2 , -NHCOCH 3 , - NHS(O) 2 CH 3 , -OH, -OCH 3 , -OCH 2 CH 3 , -S(
  • R 13 is selected from Ci-Ci 2 alkyl, C 2 -C 8 alkenyl, C 2 -C 8 alkynyl, C 3 -Ci 2 carbocyclyl, C2-C20 heterocyclyl, C 6 -C 2 O aryl, and C 1 -C 2 O heteroaryl, where alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl, and heteroaryl are optionally substituted with one or more groups independently selected from F, Cl, Br, I, -CH 2 OH, -CH 2 C 6 H 5 , -CN, -CF 3 , -CO 2 H, -
  • R 10 and R 13 together with the nitrogen atom to which they are attached form a C 2 -C 2 O heterocyclyl ring.
  • Exemplary embodiments include wherein R 1 is H or CH 3 .
  • Exemplary embodiments include wherein R is CH 3 .
  • R 3 is a monocyclic heteroaryl selected from pyridyl, isoxazolyl, imidazolyl, pyrazolyl, pyrrolyl, thiazolyl, pyridazinyl, pyrimidinyl, pyrazinyl, oxazolyl, oxadiazolyl, furanyl, thienyl, triazolyl, and tetrazolyl.
  • Exemplary embodiments include wherein R 3 is a fused bicyclic C 4 -C 2 O heterocyclyl or C 1 -C 2 O heteroaryl selected from
  • R is selected from:
  • R 3 is a fused bicyclic C 4 -C 2 2I0 heterocyclyl or C 1 -C 2 Q heteroaryl selected from:
  • Exemplary embodiments include wherein R 3 is lH-indazol-4-yl or lH-indol- 4-yl.
  • R 3 is a monocyclic heteroaryl selected from the structures:
  • R 3 is a monocyclic heteroaryl selected from the structures:
  • R 3 is a monocyclic heteroaryl selected from the structures:
  • Exemplary embodiments include wherein R 4 is optionally substituted pyrimidin-5-yl, optionally substituted C 6 -C 2 O aryl, or optionally substituted phenyl. [0088] Exemplary embodiments include wherein R 4 is -OR 10 where R 10 is optionally substituted phenyl.
  • R 4 is -OR 10 where R 10 is optionally substituted pyridyl or optionally substituted C 1 -C 12 alkyl.
  • Exemplary embodiments include wherein R 4 is -0(C 1 -C 12 alkylene)-(C 2 -C 2 o heterocyclyl), -0(C 1 -C 12 alkylene)-(C 6 -C 20 aryl), or -0(C 1 -C 12 alkylene)-(Ci-C 20 heteroaryl).
  • Exemplary embodiments include wherein R 4 is -NR 10 R 13 where R 10 is H, C 1 -
  • R 13 is optionally substituted phenyl, indazol-
  • Exemplary embodiments include wherein R 4 is -NR ⁇ (C 1 -C 12 alkyl)NR 10 R 13 ,
  • Exemplary embodiments include wherein R 4 is -NR 10 R 13 where R 10 and R 13 together with the nitrogen atom to which they are attached form morpholinyl, 4- methylpiperazin-1-yl, 4-methylsulfonylpiperazin-l-yl, or 4-(2-pyridyl)piperazin-l-yl.
  • the Formula I compounds of the invention may contain asymmetric or chiral centers, and therefore exist in different stereoisomeric forms. It is intended that all stereoisomeric forms of the compounds of the invention, including but not limited to, diastereomers, enantiomers and atropisomers, as well as mixtures thereof such as racemic mixtures, form part of the present invention.
  • the present invention embraces all geometric and positional isomers.
  • a Formula I compound incorporates a double bond or a fused ring
  • the cis- and trans-forms, as well as mixtures thereof are embraced within the scope of the invention.
  • Both the single positional isomers and mixture of positional isomers are also within the scope of the present invention.
  • the compounds of the present invention may exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like, and it is intended that the invention embrace both solvated and unsolvated forms.
  • tautomer or “tautomeric form” refers to structural isomers of different energies which are interconvertible via a low energy barrier.
  • proton tautomers also known as prototropic tautomers
  • Valence tautomers include interconversions by reorganization of some of the bonding electrons.
  • the present invention also embraces isotopically-labeled compounds of the present invention which are identical to those recited herein, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature. All isotopes of any particular atom or element as specified are contemplated within the scope of the compounds of the invention, and their uses.
  • Exemplary isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, chlorine and A i-odAi-ne, suc Uh as 2x H ⁇ , 3x H ⁇ , 11 ⁇ C, M n C, U n C, 13- Nv ⁇ , I 5 - NV ⁇ , 15 ⁇ O, 17 ⁇ O, 18 ⁇ O, 32 n P, 33 n P, 35 C S, 18 C F, 36 CTMl, 123 T 1 and 125 I.
  • Certain isotopically-labeled compounds of the present invention e.g., those labeled with 3 H and 14 C are useful in compound and/or substrate tissue distribution assays.
  • Tritiated ( 3 H) and carbon- 14 ( 14 C) isotopes are useful for their ease of preparation and detectability. Further, substitution with heavier isotopes such as deuterium (i.e., 2 H) may afford certain therapeutic advantages resulting from greater metabolic stability (e.g., increased in vivo half- life or reduced dosage requirements) and hence may be preferred in some circumstances.
  • Positron emitting isotopes such as 15 O, 13 N, 11 C and 18 F are useful for positron emission tomography (PET) studies to examine substrate receptor occupancy.
  • Isotopically labeled compounds of the present invention can generally be prepared by following procedures analogous to those disclosed in the Schemes and/or in the Examples herein below, by substituting an isotopically labeled reagent for a non-isotopically labeled reagent.
  • Pyrazolo[3,4-b]pyridines compounds of Formula I may be synthesized by synthetic routes that include processes analogous to those well-known in the chemical arts, particularly in light of the description contained herein.
  • the starting materials are generally available from commercial sources such as Aldrich Chemicals (Milwaukee, WI) or are readily prepared using methods well known to those skilled in the art (e.g., prepared by methods generally described in Louis F. Fieser and Mary Fieser, Reagents for Organic
  • compounds of Formula I may be readily prepared using well-known procedures to prepare pyrazolo[3,4-b]pyridines (Southwick et al (1975) J. of Heterocyclic Chem. 12(6): 1199-1205; Van Haverbeke et al (1979) Jour, of Heterocyclic Chem.
  • Compounds of Formula I may be prepared singly or as compound libraries comprising at least 2, for example 5 to 1,000 compounds, or 10 to 100 compounds.
  • Libraries of compounds of Formula I may be prepared by a combinatorial 'split and mix' approach or by multiple parallel syntheses using either solution phase or solid phase chemistry, by procedures known to those skilled in the art.
  • a compound library comprising at least 2 compounds, or pharmaceutically acceptable salts thereof.
  • Suitable amino-protecting groups include acetyl, trifluoroacetyl, t- butoxycarbonyl (BOC), benzyloxycarbonyl (CBz) and 9-fluorenylmethyleneoxycarbonyl (Fmoc).
  • BOC t- butoxycarbonyl
  • CBz benzyloxycarbonyl
  • Fmoc 9-fluorenylmethyleneoxycarbonyl
  • the Suzuki-type coupling reaction is useful to attach a monocyclic heteroaryl, a fused bicyclic heterocycle, or a fused bicyclic heteroaryl, at the 6-position of the pyridine ring of a 6-chloro-lH-pyrazolo[3,4-b]pyridine 21 compound.
  • 21 may be combined with about 1.5 equivalents of 4-(4 ,4,5,5 -tetramethyl- 1,3, 2-dioxaborolan-2-yl) IH- indazole 24 (US 2008/0039459, US 2008/0076758) or 5-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)pyrimidin-2-amine 25 (US 2008/0269210, US 2008/0242665), and dissolved in about 3 equivalents of sodium carbonate as a 1 molar solution in water and an equal volume of acetonitrile.
  • 4-(4 ,4,5,5 -tetramethyl- 1,3, 2-dioxaborolan-2-yl) IH- indazole 24 (US 2008/0039459, US 2008/0076758) or 5-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)pyrimidin-2-amine 25 (US 2008/0269210, US 2008/024266
  • a variety of boronic acids or boronic esters can be used in place of the indazole boronic ester indicated.
  • the nitrogen of the indazole may be protected, for example N-THP.
  • potassium acetate is used in place of sodium carbonate to adjust the pH of the aqueous layer.
  • the reaction is then heated to about 140-150 0 C under pressure in a microwave reactor such as the Biotage Optimizer (Biotage, Inc.) for 10 to 30 minutes.
  • the contents are extracted with ethyl acetate, or another organic solvent.
  • 1,3,4-substituted 6-(lH-indazol-4-yl)- lH-pyrazolo[3,4- b]pyridine 22, or 1,3,4-substituted 5-(lH-pyrazolo[3,4-b]pyridin-6-yl)pyrimidin-2-amine 23, may be purified on silica or by reverse phase HPLC.
  • Substituents R 1 , R 2 , R 4 may be R 1 , R 2 , R 4 as defined, or protected forms or precursors thereof.
  • Suzuki coupling is a palladium mediated cross coupling reaction of an arylhalide, such as 21, with a boronic acid such as 24 or 25.
  • Pd(II) and Pd(O) catalysts may be used in the Suzuki coupling reaction, including PdC12(PPh 3 ) 2 , Pd(t-Bu) 3 , PdCl 2 dppf CH 2 Cl 2 , Pd(PPh 3 ) 4 , Pd(OAc)/PPh 3 , Cl 2 Pd[(Pet 3 )] 2 , Pd(DIPHOS) 2 , Cl 2 Pd(Bipy), [PdCl(Ph 2 PCH 2 PPh 2 )] 2 , Cl 2 Pd[P(o-tol) 3 ] 2 , Pd 2 (dba) 3 /P(o-tol) 3 , Pd 2 (dba)/P(furyl) 3 , Cl 2 Pd[P(furyl) 3 ] 2 , Cl 2 Pd(PMePh 2 ) 2 , Cl 2 Pd[P(4-F-Ph) 3
  • a 4,6-dichloro pyrazolo[3,4-b]pyridine intermediate 27 in a solvent such as ethanol is added a primary or secondary amine (R 10 R 13 NH, 1.1 equiv.) and optionally a non- nucleophilic base such as triethylamine (NEt 3 , 1.5 eq, 63 ⁇ l).
  • a primary or secondary amine R 10 R 13 NH, 1.1 equiv.
  • a non- nucleophilic base such as triethylamine (NEt 3 , 1.5 eq, 63 ⁇ l).
  • acetonitrile may be used as the solvent and potassium carbonate may be used as the base.
  • the reaction mixture is stirred at room temperature for about 1 hour or overnight, volatiles removed in vacuo and residue partitioned between DCM and brine. If the mixture is insoluble it may be sonicated and the solid product was collected by filtration.
  • R 4' OR 10 , HNR 10 R 13
  • a general synthetic route to 6-heterocycle pyrazolo[3,4- ⁇ ]pyridines 35 includes chlorination of a 4-hydroxy-lH-pyrazolo[3,4- ⁇ ]pyridine-6(7H)-one compound 31 at the 2- and 4-positions with a chlorinating reagent such as a phosphorylchloride like phenylphosphoryldi chloride or phosphoryltrichloride to give the 4,6-dichloro-lH- pyrazolo[3,4- ⁇ ]pyridine intermediate 32.
  • a chlorinating reagent such as a phosphorylchloride like phenylphosphoryldi chloride or phosphoryltrichloride
  • a Suzuki-type coupling reaction according to General Procedure A can attach a monocyclic heteroaryl, a fused bicyclic heterocycle, or a fused bicyclic heteroaryl, at the 6-position of the pyridine ring of 32 with a (4,4,5,5- tetramethyl-l,3,2-dioxaborolan-2-yl)heterocycle reagent, such as 24 or 25, and a palladium catalyst, such as bis(triphenylphosphine)palladium(II) chloride (0.05 eq) in acetonitrile and sodium carbonate aqueous solution.
  • the reaction mixture is heated to about 150 0 C in a microwave for about 15 min or more.
  • LC/MS indicates when the reaction is complete.
  • reaction products from one another and/or from starting materials.
  • the desired products of each step or series of steps is separated and/or purified (hereinafter separated) to the desired degree of homogeneity by the techniques common in the art.
  • separations involve multiphase extraction, crystallization from a solvent or solvent mixture, distillation, sublimation, or chromatography.
  • Chromatography can involve any number of methods including, for example: reverse-phase and normal phase; size exclusion; ion exchange; high, medium and low pressure liquid chromatography methods and apparatus; small scale analytical; simulated moving bed (SMB) and preparative thin or thick layer chromatography, as well as techniques of small scale thin layer and flash chromatography.
  • SMB simulated moving bed
  • Another class of separation methods involves treatment of a mixture with a reagent selected to bind to or render otherwise separable a desired product, unreacted starting material, reaction by product, or the like.
  • reagents include adsorbents or absorbents such as activated carbon, molecular sieves, ion exchange media, or the like.
  • the reagents can be acids in the case of a basic material, bases in the case of an acidic material, binding reagents such as antibodies, binding proteins, selective chelators such as crown ethers, liquid/liquid ion extraction reagents (LIX), or the like.
  • Selection of appropriate methods of separation depends on the nature of the materials involved.
  • Diastereomeric mixtures can be separated into their individual diastereomers on the basis of their physical chemical differences by methods well known to those skilled in the art, such as by chromatography and/or fractional crystallization.
  • Enantiomers can be separated by converting the enantiomeric mixture into a diastereomeric mixture by reaction with an appropriate optically active compound (e.g., chiral auxiliary such as a chiral alcohol or Mosher's acid chloride), separating the diastereomers and converting (e.g., hydrolyzing) the individual diastereoisomers to the corresponding pure enantiomers.
  • an appropriate optically active compound e.g., chiral auxiliary such as a chiral alcohol or Mosher's acid chloride
  • converting e.g., hydrolyzing
  • some of the compounds of the present invention may be atropisomers (e.g., substituted biaryls) and are considered as part of this invention.
  • Enantiomers can also be separated by use of a chiral HPLC column.
  • a single stereoisomer, e.g., an enantiomer, substantially free of its stereoisomer may be obtained by resolution of the racemic mixture using a method such as formation of diastereomers using optically active resolving agents (Eliel, E. and Wilen, S. "Stereochemistry of Organic Compounds,” John Wiley & Sons, Inc., New York, 1994; Lochmuller, C. H., (1975) J. Chromatogr., 113(3):283-302).
  • Racemic mixtures of chiral compounds of the invention can be separated and isolated by any suitable method, including: (1) formation of ionic, diastereomeric salts with chiral compounds and separation by fractional crystallization or other methods, (2) formation of diastereomeric compounds with chiral derivatizing reagents, separation of the diastereomers, and conversion to the pure stereoisomers, and (3) separation of the substantially pure or enriched stereoisomers directly under chiral conditions. See: “Drug Stereochemistry, Analytical Methods and Pharmacology,” Irving W. Wainer, Ed., Marcel Dekker, Inc., New York (1993).
  • diastereomeric salts can be formed by reaction of enantiomerically pure chiral bases such as brucine, quinine, ephedrine, strychnine, ⁇ -methyl- ⁇ -phenylethylamine (amphetamine), and the like with asymmetric compounds bearing acidic functionality, such as carboxylic acid and sulfonic acid.
  • the diastereomeric salts may be induced to separate by fractional crystallization or ionic chromatography.
  • the substrate to be resolved is reacted with one enantiomer of a chiral compound to form a diastereomeric pair (Eliel, E. and Wilen, S. "Stereochemistry of Organic Compounds", John Wiley & Sons, Inc., 1994, p. 322).
  • Diastereomeric compounds can be formed by reacting asymmetric compounds with enantiomerically pure chiral derivatizing reagents, such as menthyl derivatives, followed by separation of the diastereomers and hydrolysis to yield the pure or enriched enantiomer.
  • a method of determining optical purity involves making chiral esters, such as a menthyl ester, e.g., (-) menthyl chloroformate in the presence of base, or Mosher ester, ⁇ -methoxy- ⁇ - (trifluoromethyl)phenyl acetate (Jacob III. J. Org. Chem. (1982) 47:4165), of the racemic mixture, and analyzing the 1 H NMR spectrum for the presence of the two atropisomeric enantiomers or diastereomers.
  • chiral esters such as a menthyl ester, e.g., (-) menthyl chloroformate in the presence of base, or Mosher ester, ⁇ -methoxy- ⁇ - (trifluoromethyl)phenyl acetate (Jacob III. J. Org. Chem. (1982) 47:4165), of the racemic mixture, and analyzing the 1 H NMR spectrum for the presence of the two atropis
  • Stable diastereomers of atropisomeric compounds can be separated and isolated by normal- and reverse-phase chromatography following methods for separation of atropisomeric naphthyl-isoquinolines (WO 1996/15111).
  • a racemic mixture of two enantiomers can be separated by chromatography using a chiral stationary phase ("Chiral Liquid Chromatography” (1989) W. J. Lough, Ed., Chapman and Hall, New York; Okamoto, J. Chromatogr., (1990) 513:375-378).
  • Enriched or purif ⁇ ed enantiomers can be distinguished by methods used to distinguish other chiral molecules with asymmetric carbon atoms, such as optical rotation and circular dichroism.
  • I is possible by a number of direct and indirect detection methods.
  • Certain exemplary compounds described herein were assayed for their PI3K binding activity (Example 32) and in vitro activity against tumor cells (Example 33). The range of PI3K binding activities was less than 1 nM (nanomolar) to about 10 ⁇ M (micromolar).
  • Certain exemplary compounds of the invention had PI3K binding activity IC 50 values less than 10 nM.
  • Certain compounds of the invention had tumor cell-based activity IC 50 values less than 100 nM.
  • the cytotoxic or cytostatic activity of Formula I exemplary compounds was measured by: establishing a proliferating mammalian tumor cell line in a cell culture medium, adding a Formula I compound, culturing the cells for a period from about 6 hours to about 5 days; and measuring cell viability (Example 33).
  • Cell-based in vitro assays were used to measure viability, i.e. proliferation (IC 50 ), cytotoxicity (EC 50 ), and induction of apoptosis (caspase activation).
  • the CellTiter-Glo ® Assay was conducted in 96 or 384 well format, making it amenable to automated high-throughput screening (HTS) (Cree et al (1995) Anticancer Drugs 6:398-404).
  • the homogeneous assay procedure involves adding the single reagent (CellTiter-Glo ® Reagent) directly to cells cultured in serum-supplemented medium. Cell washing, removal of medium and multiple pipetting steps are not required.
  • the system detects as few as 15 cells/well in a 384-well format in 10 minutes after adding reagent and mixing.
  • the homogeneous "add-mix -measure" format results in cell lysis and generation of a luminescent signal proportional to the amount of ATP present.
  • the amount of ATP is directly proportional to the number of cells present in culture.
  • the CellTiter-Glo ® Assay generates a "glow-type" luminescent signal, produced by the luciferase reaction, which has a half-life generally greater than five hours, depending on cell type and medium used. Viable cells are reflected in relative luminescence units (RLU).
  • the substrate, Beetle Luciferin is oxidatively decarboxylated by recombinant firefly luciferase with concomitant conversion of ATP to AMP and generation of photons.
  • This cell proliferation assay an be used with various multiwell formats, e.g. 96 or 384 well format. Data can be recorded by luminometer or CCD camera imaging device. The luminescence output is presented as relative light units (RLU), measured over time.
  • RLU relative light units
  • Exemplary Formula I compounds No. 101-121 in Table 1 were made, characterized, and tested for PI3K activity according to the methods of this invention, and have the following structures and corresponding names (ChemDraw Ultra, Version 9.0.1, CambridgeSoft Corp., Cambridge MA).
  • compound 101 had an IC50 of 0.0064 micromole; compound 102 had an IC50 of 0.224 micromole; compound 103 had an IC50 of 0.0828 micromole; compound 104 had an IC 50 of 0.00223 micromole; compound 105 had an IC 50 of 0.0137 micromole; compound 106 had an IC 50 of 0.399 micromole; compound 107 had an IC 50 of 0.053 micromole; compound 108 had an IC 50 of 0.016 micromole; compound 117 had an IC 50 of 0.0475 micromole; compound 120 had an IC 50 of 0.575 micromole; and compound 121 had an IC50 of 0.153 micromole
  • the compounds of the invention may be administered by any route appropriate to the condition to be treated. Suitable routes include oral, parenteral (including subcutaneous, intramuscular, intravenous, intraarterial, intradermal, intrathecal and epidural), transdermal, rectal, nasal, topical (including buccal and sublingual), vaginal, intraperitoneal, intrapulmonary and intranasal.
  • routes include oral, parenteral (including subcutaneous, intramuscular, intravenous, intraarterial, intradermal, intrathecal and epidural), transdermal, rectal, nasal, topical (including buccal and sublingual), vaginal, intraperitoneal, intrapulmonary and intranasal.
  • the compounds may be administered by intralesional administration, including perfusing or otherwise contacting the graft with the inhibitor before transplantation.
  • the preferred route may vary with for example the condition of the recipient.
  • the compound may be formulated as a pill, capsule, tablet, etc. with a pharmaceutically acceptable carrier or excipient.
  • the compound
  • a dose to treat human patients may range from about 10 mg to about 1000 mg of Formula I compound.
  • a typical dose may be about 100 mg to about 300 mg of the compound.
  • a dose may be administered once a day (QID), twice per day (BID), or more frequently, depending on the pharmacokinetic and pharmacodynamic properties, including absorption, distribution, metabolism, and excretion of the particular compound.
  • QID once a day
  • BID twice per day
  • toxicity factors may influence the dosage and administration regimen.
  • the pill, capsule, or tablet may be ingested daily or less frequently for a specified period of time. The regimen may be repeated for a number of cycles of therapy.
  • Compounds of the present invention are useful for treating hyperproliferative diseases, conditions and/or disorders including, but not limited to, those characterized by over expression of lipid kinases, e.g. PI3 kinase, also known as PI3K.
  • another aspect of this invention includes methods of treating or preventing diseases or conditions that can be treated or prevented by inhibiting lipid kinases, including PB.
  • the method comprises administering to a mammal in need thereof a therapeutically effective amount of a compound of Formula I.
  • a human patient is treated with a compound of Formula I and a pharmaceutically acceptable carrier, adjuvant, or vehicle, wherein said compound of Formula I is present in an amount to detectably inhibit PD kinase activity.
  • Cancers which can be treated according to the methods of this invention include, but are not limited to, breast, ovary, cervix, prostate, testis, genitourinary tract, esophagus, larynx, glioblastoma, neuroblastoma, stomach, skin, keratoacanthoma, lung, epidermoid carcinoma, large cell carcinoma, non-small cell lung carcinoma (NSCLC), small cell carcinoma, lung adenocarcinoma, bone, colon, adenoma, pancreas, adenocarcinoma, thyroid, follicular carcinoma, undifferentiated carcinoma, papillary carcinoma, seminoma, melanoma, sarcoma, bladder carcinoma, liver carcinoma and biliary passages, kidney carcinoma, myeloid disorders, lymphoid disorders, hairy cells, buccal cavity and pharynx (oral), lip, tongue, mouth, pharynx, small intestine, colon-rectum, large intestine,
  • Another aspect of this invention provides a compound of this invention for use in the treatment of the diseases or conditions described herein in a mammal, for example, a human, suffering from such disease or condition. Also provided is the use of a compound of this invention in the preparation of a medicament for the treatment of the diseases and conditions described herein in a warm-blooded animal, such as a mammal, for example a human, suffering from such disorder.
  • composition comprising a compound of this invention in association with a pharmaceutically acceptable diluent or carrier.
  • a typical formulation is prepared by mixing a compound of the present invention and a carrier, diluent or excipient.
  • Suitable carriers, diluents and excipients are well known to those skilled in the art and include materials such as carbohydrates, waxes, water soluble and/or swellable polymers, hydrophilic or hydrophobic materials, gelatin, oils, solvents, water and the like.
  • the particular carrier, diluent or excipient used will depend upon the means and purpose for which the compound of the present invention is being applied. Solvents are generally selected based on solvents recognized by persons skilled in the art as safe (GRAS) to be administered to a mammal.
  • GRAS solvents recognized by persons skilled in the art as safe
  • safe solvents are nontoxic aqueous solvents such as water and other non-toxic solvents that are soluble or miscible in water.
  • Suitable aqueous solvents include water, ethanol, propylene glycol, polyethylene glycols (e.g., PEG 400, PEG 300), etc. and mixtures thereof.
  • the formulations may also include one or more buffers, stabilizing agents, surfactants, wetting agents, lubricating agents, emulsif ⁇ ers, suspending agents, preservatives, antioxidants, opaquing agents, glidants, processing aids, colorants, sweeteners, perfuming agents, flavoring agents and other known additives to provide an elegant presentation of the drug (i.e., a compound of the present invention or pharmaceutical composition thereof) or aid in the manufacturing of the pharmaceutical product (i.e., medicament).
  • buffers stabilizing agents, surfactants, wetting agents, lubricating agents, emulsif ⁇ ers, suspending agents, preservatives, antioxidants, opaquing agents, glidants, processing aids, colorants, sweeteners, perfuming agents, flavoring agents and other known additives to provide an elegant presentation of the drug (i.e., a compound of the present invention or pharmaceutical composition thereof) or aid in the manufacturing of the pharmaceutical product (i.e., medicament).
  • the formulations may be prepared using conventional dissolution and mixing procedures.
  • the bulk drug substance i.e., compound of the present invention or stabilized form of the compound (e.g., complex with a cyclodextrin derivative or other known complex ation agent) is dissolved in a suitable solvent in the presence of one or more of the excipients described above.
  • the compound of the present invention is typically formulated into pharmaceutical dosage forms to provide an easily controllable dosage of the drug and to enable patient compliance with the prescribed regimen.
  • the pharmaceutical composition (or formulation) for application may be packaged in a variety of ways depending upon the method used for administering the drug.
  • an article for distribution includes a container having deposited therein the pharmaceutical formulation in an appropriate form.
  • Suitable containers are well known to those skilled in the art and include materials such as bottles (plastic and glass), sachets, ampoules, plastic bags, metal cylinders, and the like.
  • the container may also include a tamper-proof assemblage to prevent indiscreet access to the contents of the package.
  • the container has deposited thereon a label that describes the contents of the container. The label may also include appropriate warnings.
  • compositions of the compounds of the present invention may be prepared for various routes and types of administration.
  • a compound of Formula I having the desired degree of purity may optionally be mixed with pharmaceutically acceptable diluents, carriers, excipients or stabilizers (Remington's Pharmaceutical Sciences (1980) 16th edition, Osol, A. Ed.), in the form of a lyophilized formulation, milled powder, or an aqueous solution.
  • Formulation may be conducted by mixing at ambient temperature at the appropriate pH, and at the desired degree of purity, with physiologically acceptable carriers, i.e., carriers that are non-toxic to recipients at the dosages and concentrations employed.
  • the pH of the formulation depends mainly on the particular use and the concentration of compound, but may range from about 3 to about 8.
  • Formulation in an acetate buffer at pH 5 is a suitable embodiment.
  • the compound of this invention for use herein is preferably sterile.
  • formulations to be used for in vivo administration must be sterile. Such sterilization is readily accomplished by filtration through sterile filtration membranes.
  • the compound ordinarily can be stored as a solid composition, a lyophilized formulation or as an aqueous solution.
  • compositions of the invention will be formulated, dosed and administered in a fashion, i.e., amounts, concentrations, schedules, course, vehicles and route of administration, consistent with good medical practice.
  • Factors for consideration in this context include the particular disorder being treated, the particular mammal being treated, the clinical condition of the individual patient, the cause of the disorder, the site of delivery of the agent, the method of administration, the scheduling of administration, and other factors known to medical practitioners.
  • the "therapeutically effective amount" of the compound to be administered will be governed by such considerations, and is the minimum amount necessary to prevent, ameliorate, or treat the coagulation factor mediated disorder. Such amount is preferably below the amount that is toxic to the host or renders the host significantly more susceptible to bleeding.
  • the initial pharmaceutically effective amount of the inhibitor administered parenterally per dose will be in the range of about 0.01-100 mg/kg, namely about 0.1 to 20 mg/kg of patient body weight per day, with the typical initial range of compound used being 0.3 to 15 mg/kg/day.
  • Acceptable diluents, carriers, excipients and stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, aspara
  • the active pharmaceutical ingredients may also be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methylmethacylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions.
  • colloidal drug delivery systems for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules
  • Sustained-release preparations of compounds of Formula I may be prepared.
  • sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing a compound of Formula I, which matrices are in the form of shaped articles, e.g., films, or microcapsules.
  • sustained-release matrices include polyesters, hydrogels (for example, poly(2-hydroxyethyl-methacrylate), or poly( vinyl alcohol)), polylactides (U.S. Patent No.
  • copolymers of L-glutamic acid and gamma-ethyl-L-glutamate non-degradable ethylene-vinyl acetate
  • degradable lactic acid- glycolic acid copolymers such as the LUPRON DEPOTTM (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate) and poly-D-(-)-3- hydroxybutyric acid.
  • the formulations include those suitable for the administration routes detailed herein.
  • the formulations may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. Techniques and formulations generally are found in Remington 's Pharmaceutical Sciences (Mack Publishing Co., Easton, PA). Such methods include the step of bringing into association the active ingredient with the carrier which constitutes one or more accessory ingredients. In general the formulations are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product.
  • Formulations of a compound of Formula I suitable for oral administration may be prepared as discrete units such as pills, capsules, cachets or tablets each containing a predetermined amount of a compound of Formula I.
  • Compressed tablets may be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as a powder or granules, optionally mixed with a binder, lubricant, inert diluent, preservative, surface active or dispersing agent. Molded tablets may be made by molding in a suitable machine a mixture of the powdered active ingredient moistened with an inert liquid diluent. The tablets may optionally be coated or scored and optionally are formulated so as to provide slow or controlled release of the active ingredient therefrom.
  • a free-flowing form such as a powder or granules, optionally mixed with a binder, lubricant, inert diluent, preservative, surface active or dispersing agent.
  • Molded tablets may be made by molding in a suitable machine a mixture of the powdered active ingredient moistened with an inert liquid diluent.
  • the tablets may optionally be coated or scored and optionally are
  • Tablets, troches, lozenges, aqueous or oil suspensions, dispersible powders or granules, emulsions, hard or soft capsules, e.g., gelatin capsules, syrups or elixirs may be prepared for oral use.
  • Formulations of compounds of Formula I intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents including sweetening agents, flavoring agents, coloring agents and preserving agents, in order to provide a palatable preparation. Tablets containing the active ingredient in admixture with non-toxic pharmaceutically acceptable excipient which are suitable for manufacture of tablets are acceptable.
  • excipients may be, for example, inert diluents, such as calcium or sodium carbonate, lactose, calcium or sodium phosphate; granulating and disintegrating agents, such as maize starch, or alginic acid; binding agents, such as starch, gelatin or acacia; and lubricating agents, such as magnesium stearate, stearic acid or talc. Tablets may be uncoated or may be coated by known techniques including microencapsulation to delay disintegration and adsorption 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 alone or with a wax may be employed.
  • inert diluents such as calcium or sodium carbonate, lactose, calcium or sodium phosphate
  • granulating and disintegrating agents such as maize starch, or alginic acid
  • binding agents such as starch, ge
  • the formulations are preferably applied as a topical ointment or cream containing the active ingredient(s) in an amount of, for example, 0.075 to 20% w/w.
  • the active ingredients may be employed with either a paraffinic or a water-miscible ointment base.
  • the active ingredients may be formulated in a cream with an oil-in-water cream base.
  • the aqueous phase of the cream base may include a polyhydric alcohol, i.e. an alcohol having two or more hydroxyl groups, such as propylene glycol, butane 1,3-diol, mannitol, sorbitol, glycerol and polyethylene glycol (including PEG 400) and mixtures thereof.
  • the topical formulations may include a compound which enhances absorption or penetration of the active ingredient through the skin or other affected areas. Examples of such dermal penetration enhancers include dimethyl sulfoxide and related analogs.
  • the oily phase of the emulsions of this invention may be constituted from known ingredients in a known manner. While the phase may comprise merely an emulsifier, it desirably comprises a mixture of at least one emulsifier with a fat or an oil, or with both a fat and an oil. A hydrophilic emulsifier may be included together with a lipophilic emulsifier which acts as a stabilizer. It is also preferred to include an oil and a fat.
  • Emulsifier(s) with or without stabilizer(s) make up the so-called emulsifying wax
  • the wax together with the oil and fat make up the so-called emulsifying ointment base which forms the oily dispersed phase of the cream formulations.
  • Emulsif ⁇ ers and emulsion stabilizers suitable for use in the formulation of the invention include Tween® 60, Span® 80, cetostearyl alcohol, benzyl alcohol, myristyl alcohol, glyceryl mono-stearate and sodium lauryl sulfate.
  • Aqueous suspensions of Formula I compounds contain the active materials in admixture with excipients suitable for the manufacture of aqueous suspensions.
  • excipients include a suspending agent, such as sodium carboxymethylcellulose, croscarmellose, povidone, methylcellulose, hydroxypropyl methylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia, and dispersing or wetting agents such as a naturally occurring phosphatide (e.g., lecithin), a condensation product of an alkylene oxide with a fatty acid (e.g., polyoxyethylene stearate), a condensation product of ethylene oxide with a long chain aliphatic alcohol (e.g., heptadecaethyleneoxycetanol), a condensation product of ethylene oxide with a partial ester derived from a fatty acid and a hexitol anhydride (e.g., polyoxyethylene sorbitan monoo
  • the aqueous suspension may also contain one or more preservatives such as ethyl or n-propyl p-hydroxybenzoate, one or more coloring agents, one or more flavoring agents and one or more sweetening agents, such as sucrose or saccharin.
  • preservatives such as ethyl or n-propyl p-hydroxybenzoate
  • coloring agents such as a coloring agent
  • flavoring agents such as sucrose or saccharin.
  • sweetening agents such as sucrose or saccharin.
  • compositions of compounds of Formula I may be in the form of a sterile injectable preparation, such as a sterile injectable aqueous or oleaginous suspension.
  • a sterile injectable preparation such as a sterile injectable aqueous or oleaginous suspension.
  • This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, such as a solution in 1,3- butanediol or prepared as a lyophilized powder.
  • a non-toxic parenterally acceptable diluent or solvent such as a solution in 1,3- butanediol or prepared as a lyophilized powder.
  • acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution.
  • sterile fixed oils may conventionally be employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid may likewise be used in the preparation of injectables.
  • a time-release formulation intended for oral administration to humans may contain approximately 1 to 1000 mg of active material compounded with an appropriate and convenient amount of carrier material which may vary from about 5 to about 95% of the total compositions (weightweight).
  • the pharmaceutical composition can be prepared to provide easily measurable amounts for administration.
  • an aqueous solution intended for intravenous infusion may contain from about 3 to 500 ⁇ g of the active ingredient per milliliter of solution in order that infusion of a suitable volume at a rate of about 30 mL/hr can occur.
  • Formulations suitable for parenteral administration include aqueous and nonaqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents.
  • Formulations suitable for topical administration to the eye also include eye drops wherein the active ingredient is dissolved or suspended in a suitable carrier, especially an aqueous solvent for the active ingredient.
  • the active ingredient is preferably present in such formulations in a concentration of about 0.5 to 20% w/w, for example about 0.5 to 10% w/w, for example about 1.5% w/w.
  • Formulations suitable for topical administration in the mouth include lozenges comprising the active ingredient in a flavored basis, usually sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert basis such as gelatin and glycerin, or sucrose and acacia; and mouthwashes comprising the active ingredient in a suitable liquid carrier.
  • Formulations for rectal administration may be presented as a suppository with a suitable base comprising for example cocoa butter or a salicylate.
  • Formulations suitable for intrapulmonary or nasal administration have a particle size for example in the range of 0.1 to 500 microns (including particle sizes in a range between 0.1 and 500 microns in increments microns such as 0.5, 1, 30 microns, 35 microns, etc.), which is administered by rapid inhalation through the nasal passage or by inhalation through the mouth so as to reach the alveolar sacs.
  • Suitable formulations include aqueous or oily solutions of the active ingredient.
  • Formulations suitable for aerosol or dry powder administration may be prepared according to conventional methods and may be delivered with other therapeutic agents such as compounds heretofore used in the treatment or prophylaxis disorders as described below.
  • Formulations suitable for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams or spray formulations containing in addition to the active ingredient such carriers as are known in the art to be appropriate.
  • the formulations may be packaged in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water, for injection immediately prior to use. Extemporaneous injection solutions and suspensions are prepared from sterile powders, granules and tablets of the kind previously described.
  • Preferred unit dosage formulations are those containing a daily dose or unit daily sub-dose, as herein above recited, or an appropriate fraction thereof, of the active ingredient.
  • the invention further provides veterinary compositions comprising at least one active ingredient as above defined together with a veterinary carrier therefore.
  • Veterinary carriers are materials useful for the purpose of administering the composition and may be solid, liquid or gaseous materials which are otherwise inert or acceptable in the veterinary art and are compatible with the active ingredient. These veterinary compositions may be administered parenterally, orally or by any other desired route.
  • the compounds of Formula I may be employed alone or in combination with other therapeutic agents for the treatment of a disease or disorder described herein, such as a hyperproliferative disorder (e.g., cancer).
  • a compound of Formula I is combined in a pharmaceutical combination formulation, or dosing regimen as combination therapy, with a second compound that has anti-hyperproliferative properties or that is useful for treating a hyperproliferative disorder (e.g., cancer).
  • the second compound of the pharmaceutical combination formulation or dosing regimen preferably has complementary activities to the compound of Formula I such that they do not adversely affect each other.
  • Such compounds are suitably present in combination in amounts that are effective for the purpose intended.
  • a composition of this invention comprises a compound of Formula I, or a stereoisomer, geometric isomer, tautomer, solvate, metabolite, or pharmaceutically acceptable salt or prodrug thereof, in combination with a chemotherapeutic agent such as described herein.
  • the combination therapy may be administered as a simultaneous or sequential regimen.
  • the combination may be administered in two or more administrations.
  • the combined administration includes coadministration, using separate formulations or a single pharmaceutical formulation, and consecutive administration in either order, wherein preferably there is a time period while both (or all) active agents simultaneously exert their biological activities.
  • Suitable dosages for any of the above coadministered agents are those presently used and may be lowered due to the combined action (synergy) of the newly identified agent and other chemotherapeutic agents or treatments.
  • the combination therapy may provide "synergy” and prove “synergistic", i.e., the effect achieved when the active ingredients used together is greater than the sum of the effects that results from using the compounds separately.
  • a synergistic effect may be attained when the active ingredients are: (1) co-formulated and administered or delivered simultaneously in a combined, unit dosage formulation; (2) delivered by alternation or in parallel as separate formulations; or (3) by some other regimen.
  • a synergistic effect may be attained when the compounds are administered or delivered sequentially, e.g., by different injections in separate syringes, separate pills or capsules, or separate infusions.
  • an effective dosage of each active ingredient is administered sequentially, i.e., serially
  • effective dosages of two or more active ingredients are administered together.
  • a compound of Formula I, or a stereoisomer, geometric isomer, tautomer, solvate, metabolite, or pharmaceutically acceptable salt or prodrug thereof may be combined with other chemotherapeutic, hormonal or antibody agents such as those described herein, as well as combined with surgical therapy and radiotherapy.
  • Combination therapies according to the present invention thus comprise the administration of at least one compound of Formula I, or a stereoisomer, geometric isomer, tautomer, solvate, metabolite, or pharmaceutically acceptable salt or prodrug thereof, and the use of at least one other cancer treatment method.
  • the amounts of the compound(s) of Formula I and the other pharmaceutically active chemotherapeutic agent(s) and the relative timings of administration will be selected in order to achieve the desired combined therapeutic effect.
  • the in vivo metabolic products of Formula I described herein include metabolites of compounds of Formula I, including compounds produced by a process comprising contacting a compound of this invention with a mammal for a period of time sufficient to yield a metabolic product thereof.
  • Metabolite products typically are identified by preparing a radiolabeled (e.g.,
  • metabolites In general, analysis of metabolites is done in the same way as conventional drug metabolism studies well known to those skilled in the art.
  • the metabolite products so long as they are not otherwise found in vivo, are useful in diagnostic assays for therapeutic dosing of the compounds of the invention.
  • kits containing materials useful for the treatment of the diseases and disorders described above.
  • the kit comprises a container comprising a compound of Formula I, or a stereoisomer, geometric isomer, tautomer, solvate, metabolite, or pharmaceutically acceptable salt or prodrug thereof.
  • the kit may further comprise a label or package insert on or associated with the container.
  • package insert is used to refer to instructions customarily included in commercial packages of therapeutic products, that contain information about the indications, usage, dosage, administration, contraindications and/or warnings concerning the use of such therapeutic products.
  • Suitable containers include, for example, bottles, vials, syringes, blister pack, etc.
  • the container may be formed from a variety of materials such as glass or plastic.
  • the container may hold a compound of Formula I or a formulation thereof which is effective for treating the condition and may have a sterile access port (for example, the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
  • At least one active agent in the composition is a compound of Formula I.
  • the label or package insert indicates that the composition is used for treating the condition of choice, such as cancer.
  • the label or package insert may indicate that the patient to be treated is one having a disorder such as a hyperproliferative disorder, neurodegeneration, cardiac hypertrophy, pain, migraine or a neurotraumatic disease or event.
  • the label or package inserts indicates that the composition comprising a compound of Formula I can be used to treat a disorder resulting from abnormal cell growth.
  • the label or package insert may also indicate that the composition can be used to treat other disorders.
  • the article of manufacture may further comprise a second container comprising a pharmaceutically acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer's solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes.
  • BWFI bacteriostatic water for injection
  • phosphate-buffered saline such as bacteriostatic water for injection (BWFI), phosphate-buffered
  • the kit may further comprise directions for the administration of the compound of Formula I and, if present, the second pharmaceutical formulation.
  • the kit may further comprise directions for the simultaneous, sequential or separate administration of the first and second pharmaceutical compositions to a patient in need thereof.
  • kits are suitable for the delivery of solid oral forms of a compound of Formula I, such as tablets or capsules.
  • a kit preferably includes a number of unit dosages.
  • Such kits can include a card having the dosages oriented in the order of their intended use.
  • An example of such a kit is a "blister pack".
  • Blister packs are well known in the packaging industry and are widely used for packaging pharmaceutical unit dosage forms.
  • a memory aid can be provided, for example in the form of numbers, letters, or other markings or with a calendar insert, designating the days in the treatment schedule in which the dosages can be administered.
  • a kit may comprise (a) a first container with a compound of Formula I contained therein; and optionally (b) a second container with a second pharmaceutical formulation contained therein, wherein the second pharmaceutical formulation comprises a second compound with anti-hyperproliferative activity.
  • the kit may further comprise a third container comprising a pharmaceutically-acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate -buffered saline, Ringer's solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes.
  • BWFI bacteriostatic water for injection
  • kit comprises a composition of
  • the kit may comprise a container for containing the separate compositions such as a divided bottle or a divided foil packet, however, the separate compositions may also be contained within a single, undivided container.
  • the kit comprises directions for the administration of the separate components.
  • the kit form is particularly advantageous when the separate components are preferably administered in different dosage forms (e.g., oral and parenteral), are administered at different dosage intervals, or when titration of the individual components of the combination is desired by the prescribing physician.
  • Reagents were purchased from commercial suppliers such as Sigma Aldrich Chemical Company, Lancaster, TCI or Maybridge, and were used without further purification unless otherwise indicated.
  • the reactions set forth below were done generally under a positive pressure of nitrogen or argon or with a drying tube (unless otherwise stated) in anhydrous solvents, and the reaction flasks were typically fitted with rubber septa for the introduction of substrates and reagents via syringe. Glassware was oven dried and/or heat dried. Column chromatography was conducted on a Biotage system (Manufacturer: Dyax Corporation) having a silica gel column or on a silica SEP PAK® cartridge (Waters).
  • Example 15 7-( 1 -methyl-4-(4-(methylsulfonyl)phenoxy)- 1 H-pyrazolo [3 ,A- b]pyridin-6-yl)-lH-indazol-3 -amine 105
  • Example 16 4-(2,6-dimethylphenoxy)-6-( 1 H-indazol-4-yl)- 1 -methyl- 1 H- pyrazolo[3,4-b]pyridine 106
  • Nitrogen was bubbled through the resultant reaction mixture and heated in a microwave at
  • Example 18 4-(3 ,4-dimethoxyphenoxy)-6-( 1 H-indazol-4-yl)- 1 -methyl- 1 H- pyrazolo[3,4-b]pyridine 108
  • Example 21 3 -(6-( 1 H-indazol-4-yl)- 1 -methyl- 1 H-pyrazolo [3 ,4-b]pyridin-4- yl)-N-methylbenzamide 111
  • Example 24 3 -(6-( 1 H-indazol-4-yl)- 1 -methyl- 1 H-pyrazolo [3 ,4-b]pyridin-4- ylamino)-2,2-dimethylpropan- 1 -ol 114
  • Example 26 6-( 1 H-indazol-4-yl)- 1 -methyl-N-(3 -(methylsulfonyl)propyl)- lH-pyrazolo[3,4-b]pyridin-4-amine 116
  • reaction mixture was concentrated in vacuo and purified by flash chromatography (30% EtOAc in hexane) to provide 4-chloro-6-(6-methoxypyridin-3-yl)-l-methyl-l ⁇ -pyrazolo[3,4-b]pyridine (0.32 g).
  • Example 30 4-( 1 -methyl-4-(4-(methylsulfonyl)phenoxy)- 1 H-pyrazolo [3 ,4- b]pyridin-6-yl)pyridin-2-ol 120
  • Example 31 7-( 1 -methyl-4-(4-(methylsulfonyl)phenoxy)- 1 H-pyrazolo [3 ,4- b]pyridin-6-yl)benzo[d]isoxazol-3-amine 121
  • Binding Assays Initial polarization experiments were performed on an
  • Samples for fluorescence polarization affinity measurements were prepared by addition of 1 :3 serial dilutions of pi 10 alpha PI3K (Upstate Cell Signaling Solutions, Charlottesville, VA) starting at a final concentration of 20ug/mL in polarization buffer (10 mM Tris pH 7.5, 50 mM NaCl, 4mM MgCl 2 , 0.05%Chaps, and 1 mM DTT) to 1OmM PIP 2 (Echelon-Inc, Salt Lake City, UT.) final concentration.
  • polarization buffer 10 mM Tris pH 7.5, 50 mM NaCl, 4mM MgCl 2 , 0.05%Chaps, and 1 mM DTT
  • Inhibitor IC50 values were determined by addition of the 0.04 mg/mL pi 10 alpha PI3K (final concentration) combined with PIP 2 (1OmM final concentration) to wells containing 1 :3 serial dilutions of the antagonists in a final concentration of 25mM ATP (Cell Signaling Technology, Inc., Danvers, MA) in the polarization buffer. After an incubation time of 30 minutes at room temperature, the reactions were stopped by the addition of GRP-I and PIP3-TAMRA probe (Echelon-Inc, Salt Lake City, UT.) 100 nM and 5 nM final concentrations respectively.
  • GRP-I and PIP3-TAMRA probe Echelon-Inc, Salt Lake City, UT.
  • Efficacy of Formula I compounds were measured by a cell proliferation assay employing the following protocol (Promega Corp. Technical Bulletin TB288; Mendoza et al (2002) Cancer Res. 62:5485-5488): [00262] 1. An aliquot of 100 ⁇ l of cell culture containing about 10 cells (PC3 ,
  • Control wells were prepared containing medium and without cells.
  • cells were seeded at optimal density in a 96 well plate and incubated for 4 days in the presence of test compound.
  • Alamar BlueTM was subsequently added to the assay medium, and cells were incubated for 6 h before reading at 544nm excitation, 590nm emission.
  • EC 50 values were calculated using a sigmoidal dose response curve fit.
  • Example 34 Caco-2 Permeability
  • Suspensions of cryopreserved human hepatocytes are used. Incubations are performed at compound concentration of ImM or 3 ⁇ M at a cell density of 0.5 x 10 6 viable cells/mL. The final DMSO concentration in the incubation is about 0.25%. Control incubations are also performed in the absence of cells to reveal any non-enzymatic degradation. Duplicate samples (50 ⁇ L) are removed from the incubation mixture at 0, 5, 10, 20, 40 and 60 minutes (control sample at 60 minutes only) and added to MeOH - containing internal standard (lOO ⁇ L) - to terminate the reaction. Tolbutamide, 7-hydroxycoumarin, and testosterone may be used as control compounds.
  • Formula I compounds may be screened against CYP450 targets (1 A2, 2C9,
  • 2Cl 9, 2D6, 3A4 at about 10 concentrations in duplicate, with a top concentration of about 10OuM.
  • Standard inhibitors furafylline, sulfaphenazole, tranylcypromine, quinidine, ketoconazole
  • Plates may be read using a BMG LabTechnologies PolarStar in fluorescence mode.
  • Freshly isolated human hepatocytes from a single donor may be cultured for about 48 h prior to addition of Formula I compound at three concentrations and incubated for 72 h.
  • Probe substrates for CYP3A4 and CYP 1A2 are added for 30 minutes and 1 h before the end of the incubation.
  • cells and media are removed and the extent of metabolism of each probe substrate quantified by LC-MS/MS. The experiment is controlled by using inducers of the individual P450s incubated at one concentration in triplicate.
  • Formula I compounds are evaluated for ability to modulate rubidium efflux from HEK-294 cells stably expressing hERG potassium channels using established flux methodology.
  • Cells are prepared in medium containing RbCl, plated into 96-well plates and grown overnight to form monolayers.
  • the efflux experiment is initiated by aspirating the media and washing each well with 3 x lOO ⁇ L of pre-incubation buffer (containing low [K + ]) at room temperature. Following the final aspiration, 50 ⁇ L of working stock (2x) compound is added to each well and incubated at room temperature for 10 minutes. 50 ⁇ L of stimulation buffer (containing high [K+]) is then added to each well giving the final test compound concentrations.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Diabetes (AREA)
  • Hematology (AREA)
  • Immunology (AREA)
  • Oncology (AREA)
  • Obesity (AREA)
  • Pain & Pain Management (AREA)
  • Communicable Diseases (AREA)
  • Emergency Medicine (AREA)
  • Endocrinology (AREA)
  • Virology (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Rheumatology (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

L'invention porte sur des composés de la formule I qui comprennent des stéréo-isomères, des isomères géométriques, des tautomères, des solvates, des métabolites et des sels de qualité pharmaceutique de ceux-ci et qui sont utiles pour inhiber des lipides kinases comprenant la p110 alpha et autres isoformes de PI3K, et pour traiter des troubles tels qu'un cancer ou une inflammation à médiation par des lipides kinases. L'invention porte sur des procédés d'utilisation de composés de la formule I pour le diagnostic in vitro, in situ et in vivo, la prévention ou le traitement de tels troubles dans des cellules mammifères, ou des états pathologiques associés.
PCT/US2009/065085 2008-11-20 2009-11-19 Composés inhibiteurs de pi3k de type pyrazolopyridine et leurs procédés d'utilisation WO2010059788A1 (fr)

Priority Applications (7)

Application Number Priority Date Filing Date Title
CN200980146318.8A CN102224152B (zh) 2008-11-20 2009-11-19 吡唑并吡啶pi3k抑制剂化合物及使用方法
SG2011036381A SG171765A1 (en) 2008-11-20 2009-11-19 Pyrazolopyridine pi3k inhibitor compounds and methods of use
JP2011537609A JP5689069B2 (ja) 2008-11-20 2009-11-19 ピラゾロピリジンpi3k阻害剤化合物及び使用方法
ES09761108T ES2392488T3 (es) 2008-11-20 2009-11-19 Compuestos inhibidores de PI3K de tipo pirazolopiridina y sus procedimientos de uso
CA2744154A CA2744154A1 (fr) 2008-11-20 2009-11-19 Composes inhibiteurs de pi3k de type pyrazolopyridine et leurs procedes d'utilisation
US13/129,091 US8653098B2 (en) 2008-11-20 2009-11-19 Pyrazolopyridine PI3K inhibitor compounds and methods of use
EP09761108A EP2356117B1 (fr) 2008-11-20 2009-11-19 Composés inhibiteurs de pi3k de type pyrazolopyridine et leurs procédés d'utilisation

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US11642708P 2008-11-20 2008-11-20
US61/116,427 2008-11-20

Publications (1)

Publication Number Publication Date
WO2010059788A1 true WO2010059788A1 (fr) 2010-05-27

Family

ID=41694921

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2009/065085 WO2010059788A1 (fr) 2008-11-20 2009-11-19 Composés inhibiteurs de pi3k de type pyrazolopyridine et leurs procédés d'utilisation

Country Status (8)

Country Link
US (1) US8653098B2 (fr)
EP (1) EP2356117B1 (fr)
JP (1) JP5689069B2 (fr)
CN (1) CN102224152B (fr)
CA (1) CA2744154A1 (fr)
ES (1) ES2392488T3 (fr)
SG (1) SG171765A1 (fr)
WO (1) WO2010059788A1 (fr)

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012156379A1 (fr) 2011-05-13 2012-11-22 Universität Zürich Inhibiteurs de pik3/p110α pour la prévention de la resténose par application sous la forme d'un revêtement pour implant
WO2013174794A1 (fr) 2012-05-23 2013-11-28 F. Hoffmann-La Roche Ag Compositions et procédés d'obtention et d'utilisation de cellules endodermiques et d'hépatocytes
US8673922B2 (en) 2010-07-15 2014-03-18 Bristol-Myers Squibb Company Azaindazole compounds
US20140235647A1 (en) * 2011-09-27 2014-08-21 Bristol-Myers Squibb Company Substituted bicyclic heteroaryl compounds
WO2014144850A1 (fr) 2013-03-15 2014-09-18 Genentech, Inc. Procédés de traitement d'un cancer et de prévention de la résistance médicamenteuse d'un cancer
US8883799B2 (en) 2010-12-16 2014-11-11 Genentech, Inc. Tricyclic PI3K inhibitor compounds and methods of use
WO2014198942A1 (fr) * 2013-06-14 2014-12-18 Sanofi Dérivés de pyrazolopyridine à utiliser dans le traitement du cancer de la vessie
US9266880B2 (en) 2010-11-12 2016-02-23 Bristol-Myers Squibb Company Substituted azaindazole compounds
WO2017053604A1 (fr) * 2015-09-23 2017-03-30 The Regents Of The University Of California Composés pyrazolopyridines antiviraux puissants
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
US10479770B2 (en) 2016-09-23 2019-11-19 Gilead Sciences, Inc. Phosphatidylinositol 3-kinase inhibitors
CN116239594A (zh) * 2023-03-05 2023-06-09 贵州省中国科学院天然产物化学重点实验室(贵州医科大学天然产物化学重点实验室) 6-(咪唑并[1,2-a]吡啶-6-基)喹唑啉衍生物及用途

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR101735975B1 (ko) * 2012-11-29 2017-05-15 에프. 호프만-라 로슈 아게 이미다조피리딘 유도체
WO2023239846A1 (fr) * 2022-06-10 2023-12-14 Synnovation Therapeutics, Inc. Composés hétérocycliques utilisés comme inhibiteurs de pi3kα

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003008405A1 (fr) * 2001-07-18 2003-01-30 L'oreal Composes derives de diaminopyrazole substitues par un radical heteroaromatique et leur utilisation en teinture d'oxydation des fibres keratiniques
WO2007114926A2 (fr) * 2006-04-04 2007-10-11 The Regents Of The University Of California Antagonistes de la kinase
WO2007127183A1 (fr) * 2006-04-26 2007-11-08 Genentech, Inc. Composes inhibiteurs de phosphoinositide 3-kinase et compositions pharmaceutiques les contenant
WO2007135398A1 (fr) * 2006-05-22 2007-11-29 Astrazeneca Ab Dérivés de l'indole
WO2009059030A1 (fr) * 2007-10-31 2009-05-07 Burnham Institute For Medical Research Dérivés de pyrazole en tant qu'inhibiteurs de kinase
WO2009097446A1 (fr) * 2008-01-30 2009-08-06 Genentech, Inc. Composés de pyrazolopyrimidine inhibant pi3k et procédés d'utilisation
WO2009145357A1 (fr) * 2008-05-30 2009-12-03 Otsuka Pharmaceutical Co., Ltd. Benzo[1,4]diazépines substituées en position 1 par un cycle pour utilisation en tant qu'antidépresseurs

Family Cites Families (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3403158A (en) 1966-03-11 1968-09-24 Upjohn Co 1h-pyrazolo[3, 4-b]pyridines
US4115394A (en) * 1974-05-06 1978-09-19 E. R. Squibb & Sons, Inc. Amino derivatives of 6-phenylpyrazolo[3,4-b]pyridines
US3894005A (en) 1974-05-24 1975-07-08 Squibb & Sons Inc Hydrazones of pyrazolo{8 3,4-b{9 pyridines
DE2643753A1 (de) 1976-09-29 1978-04-06 Thomae Gmbh Dr K Neue 1h-pyrazolo eckige klammer auf 3,4-b eckige klammer zu pyridine
IL134748A0 (en) 1997-09-02 2001-04-30 Du Pont Pharm Co Heterocyclyl-substituted ring-fused pyridines and pyrimidines as corticotropin releasing hormone (crh) antagonists, useful for treating cns and stress-related disorders
ATE355279T1 (de) 1998-08-07 2006-03-15 Novartis Vaccines & Diagnostic Pyrazole als modulatoren des östrogenrezeptors
JP2004526789A (ja) 2001-04-25 2004-09-02 アルタナ ファルマ アクチエンゲゼルシャフト 新規フタラジノン
ES2309206T3 (es) 2001-10-02 2008-12-16 Smithkline Beecham Corporation Compuestos quimicos.
TW200400034A (en) 2002-05-20 2004-01-01 Bristol Myers Squibb Co Pyrazolo-pyrimidine aniline compounds useful as kinase inhibitors
EP1608631A4 (fr) 2003-03-28 2008-08-20 Scios Inc Inhibiteurs bi-cycliques a base de pyrimidine de tgf beta
CN102786482A (zh) 2003-11-21 2012-11-21 阿雷生物药品公司 Akt蛋白激酶抑制剂
AU2005288858B2 (en) 2004-09-30 2011-04-21 Janssen R&D Ireland HCV inhibiting bi-cyclic pyrimidines
EP1814885A4 (fr) 2004-11-23 2009-12-16 Us Therapeutics Inc Reddy Nouveaux composes bicycliques heterocycliques, leur procede de preparation et compositions les contenant
WO2006071819A1 (fr) 2004-12-28 2006-07-06 Exelixis, Inc. Composes piperazine ou [1h-pyrazolo[3,4-d]pyrimidin-4-yl]-piperidine utilises en tant que modulateurs des serine-theoronine kinases (p70s6k, atk1 et atk2) pour le traitement de maladies immunologiques, inflammatoires et proliferantes
US20100160255A1 (en) 2005-07-29 2010-06-24 Takeda Pharmaceutical Company Limited Spiro-cyclic compound
JO2660B1 (en) * 2006-01-20 2012-06-17 نوفارتيس ايه جي Pi-3 inhibitors and methods of use
MX2008013584A (es) * 2006-04-26 2009-03-23 Genentech Inc Compuestos farmaceuticos.
TW200815438A (en) 2006-06-13 2008-04-01 Bayer Schering Pharma Ag Substituted pyrazolopyridines and salts thereof, pharmaceutical compositions comprising same, methods of preparing same and uses of same
TW200815437A (en) 2006-06-13 2008-04-01 Bayer Schering Pharma Ag Substituted aminopyrazolopyridines and salts thereof, pharmaceutical compositions comprising same, methods of preparing same and uses of same
BRPI0717564A2 (pt) * 2006-09-29 2013-10-22 Novartis Ag Pirazolopirimidinas como inibidores de pi3k lipídeo cinase
WO2008088692A2 (fr) 2007-01-12 2008-07-24 Merck & Co., Inc. Dérivés spirochromanone
PE20081559A1 (es) 2007-01-12 2008-11-20 Merck & Co Inc DERIVADOS DE ESPIROCROMANONA SUSTITUIDOS COMO INHIBIDORES DE ACETIL CoA CARBOXILASA

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003008405A1 (fr) * 2001-07-18 2003-01-30 L'oreal Composes derives de diaminopyrazole substitues par un radical heteroaromatique et leur utilisation en teinture d'oxydation des fibres keratiniques
WO2007114926A2 (fr) * 2006-04-04 2007-10-11 The Regents Of The University Of California Antagonistes de la kinase
WO2007127183A1 (fr) * 2006-04-26 2007-11-08 Genentech, Inc. Composes inhibiteurs de phosphoinositide 3-kinase et compositions pharmaceutiques les contenant
WO2007135398A1 (fr) * 2006-05-22 2007-11-29 Astrazeneca Ab Dérivés de l'indole
WO2009059030A1 (fr) * 2007-10-31 2009-05-07 Burnham Institute For Medical Research Dérivés de pyrazole en tant qu'inhibiteurs de kinase
WO2009097446A1 (fr) * 2008-01-30 2009-08-06 Genentech, Inc. Composés de pyrazolopyrimidine inhibant pi3k et procédés d'utilisation
WO2009145357A1 (fr) * 2008-05-30 2009-12-03 Otsuka Pharmaceutical Co., Ltd. Benzo[1,4]diazépines substituées en position 1 par un cycle pour utilisation en tant qu'antidépresseurs

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
GONZALEZ E ET AL: "Acylation d'aminopyrazoles: formation de pyrazolo[3,4-b]pyridines, pyrazolo[3,4-d]pyrimidines et dipyrazolo[3,4-b:4',3'-e]pyridines", TETRAHEDRON, ELSEVIER SCIENCE PUBLISHERS, AMSTERDAM, NL, vol. 34, no. 8, 1 January 1978 (1978-01-01), pages 1175 - 1178, XP026648639, ISSN: 0040-4020, [retrieved on 19780101] *
VOLOCHNYUK D M ET AL: "Electron-rich amino heterocycles for regiospecific synthesis of trifluoromethyl containing fused pyridines", SYNTHESIS, vol. 10, 2003, pages 1531 - 1540, XP002570587 *

Cited By (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8673922B2 (en) 2010-07-15 2014-03-18 Bristol-Myers Squibb Company Azaindazole compounds
US9266880B2 (en) 2010-11-12 2016-02-23 Bristol-Myers Squibb Company Substituted azaindazole compounds
US8883799B2 (en) 2010-12-16 2014-11-11 Genentech, Inc. Tricyclic PI3K inhibitor compounds and methods of use
US9546182B2 (en) 2010-12-16 2017-01-17 Genentech, Inc. Tricyclic PI3K inhibitor compounds and methods of use
WO2012156379A1 (fr) 2011-05-13 2012-11-22 Universität Zürich Inhibiteurs de pik3/p110α pour la prévention de la resténose par application sous la forme d'un revêtement pour implant
US8969586B2 (en) 2011-09-27 2015-03-03 Bristol-Myers Squibb Company Substituted bicyclic heteroaryl compounds
US20140235647A1 (en) * 2011-09-27 2014-08-21 Bristol-Myers Squibb Company Substituted bicyclic heteroaryl compounds
US9242967B2 (en) 2011-09-27 2016-01-26 Bristol-Myers Squibb Company Substituted bicyclic heteroaryl compounds
US9598436B2 (en) 2011-09-27 2017-03-21 Bristol-Myers Squibb Company Substituted bicyclic heteroaryl compounds
WO2013174794A1 (fr) 2012-05-23 2013-11-28 F. Hoffmann-La Roche Ag Compositions et procédés d'obtention et d'utilisation de cellules endodermiques et d'hépatocytes
WO2014144850A1 (fr) 2013-03-15 2014-09-18 Genentech, Inc. Procédés de traitement d'un cancer et de prévention de la résistance médicamenteuse d'un cancer
US9572799B2 (en) 2013-06-14 2017-02-21 Sanofi Pyrazolopyridine derivatives for use in the treatment of bladder cancer
WO2014198942A1 (fr) * 2013-06-14 2014-12-18 Sanofi Dérivés de pyrazolopyridine à utiliser dans le traitement du cancer de la vessie
CN105283184A (zh) * 2013-06-14 2016-01-27 赛诺菲 用于治疗膀胱癌的吡唑并吡啶衍生物
EA030558B1 (ru) * 2013-06-14 2018-08-31 Санофи Применение производных пиразолопиридина для лечения рака мочевого пузыря
WO2017053604A1 (fr) * 2015-09-23 2017-03-30 The Regents Of The University Of California Composés pyrazolopyridines antiviraux puissants
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
US10479770B2 (en) 2016-09-23 2019-11-19 Gilead Sciences, Inc. Phosphatidylinositol 3-kinase inhibitors
CN116239594A (zh) * 2023-03-05 2023-06-09 贵州省中国科学院天然产物化学重点实验室(贵州医科大学天然产物化学重点实验室) 6-(咪唑并[1,2-a]吡啶-6-基)喹唑啉衍生物及用途
CN116239594B (zh) * 2023-03-05 2023-09-22 贵州省中国科学院天然产物化学重点实验室(贵州医科大学天然产物化学重点实验室) 6-(咪唑并[1,2-a]吡啶-6-基)喹唑啉衍生物及用途

Also Published As

Publication number Publication date
ES2392488T3 (es) 2012-12-11
EP2356117A1 (fr) 2011-08-17
JP2012509345A (ja) 2012-04-19
EP2356117B1 (fr) 2012-09-26
CA2744154A1 (fr) 2010-05-27
US8653098B2 (en) 2014-02-18
CN102224152B (zh) 2014-05-21
SG171765A1 (en) 2011-07-28
CN102224152A (zh) 2011-10-19
JP5689069B2 (ja) 2015-03-25
US20120035208A1 (en) 2012-02-09

Similar Documents

Publication Publication Date Title
EP2252616B1 (fr) Composés de pyrazolopyrimidine inhibant pi3k et procédés d'utilisation
EP2356117B1 (fr) Composés inhibiteurs de pi3k de type pyrazolopyridine et leurs procédés d'utilisation
US8445487B2 (en) Purine PI3K inhibitor compounds and methods of use
US9546182B2 (en) Tricyclic PI3K inhibitor compounds and methods of use
CA2704711C (fr) Composes inhibiteurs de la pi3k a base de thiazolopyrimidine
EP2890698B1 (fr) Composés dioxino- et oxazine-[2,3-d]pyrimidine, inhibiteurs de pi3k, et leurs procédés d'utilisation
AU2015268776B2 (en) Tricyclic PI3k inhibitor compounds and methods of use

Legal Events

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

Ref document number: 200980146318.8

Country of ref document: CN

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

Ref document number: 09761108

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2843/DELNP/2011

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2009761108

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2011537609

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 2744154

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 13129091

Country of ref document: US