WO2016026423A1 - Macrocycles substitués utilisés comme inhibiteurs des kinases et leurs méthodes d'utilisation - Google Patents

Macrocycles substitués utilisés comme inhibiteurs des kinases et leurs méthodes d'utilisation Download PDF

Info

Publication number
WO2016026423A1
WO2016026423A1 PCT/CN2015/087343 CN2015087343W WO2016026423A1 WO 2016026423 A1 WO2016026423 A1 WO 2016026423A1 CN 2015087343 W CN2015087343 W CN 2015087343W WO 2016026423 A1 WO2016026423 A1 WO 2016026423A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
compounds
cancer
pharmaceutically acceptable
kinase
Prior art date
Application number
PCT/CN2015/087343
Other languages
English (en)
Inventor
Xiaoyang Xia
Dawei Zhang
Original Assignee
Teligene Ltd
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 Teligene Ltd filed Critical Teligene Ltd
Priority to JP2017510559A priority Critical patent/JP6609308B2/ja
Priority to EP15833622.2A priority patent/EP3183256A4/fr
Priority to US15/500,927 priority patent/US10159663B2/en
Priority to CN201580043435.7A priority patent/CN107207528B/zh
Publication of WO2016026423A1 publication Critical patent/WO2016026423A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/439Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom the ring forming part of a bridged ring system, e.g. quinuclidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/12Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains three hetero rings
    • C07D498/18Bridged systems

Definitions

  • the present invention is directed to inhibitors of kinase and pharmaceutically acceptable salts, solvates, hydrates, prodrugs and metabolites thereof, the preparation method thereof, and the use of such compounds to treat kinase mediated diseases and conditions such as cancer.
  • Protein kinases represent a large family of enzymes, which catalyze the phosphorylation of target protein substrates.
  • the phosphorylation is usually a transfer reaction of a phosphate group from ATP to the protein substrate.
  • Common points of attachment for the phosphate group to the protein substrate include, for example, a tyrosine, serine or threonine residue.
  • kinases in the protein kinase family include, without limitation, Abl1 (v-Abl Abelson murine leukemia viral oncogene homolog 1) , Akt, Alk, Bcr-Abl1, Blk, Brk, Btk, c-Kit, c-Met, c-Src, c-Fms, CDK1-10, b-Raf, c-Raf1, CSF1R, CSK, EGFR, ErbB2, ErbB3, ErbB4, Erk, FGFR1, FGFR2, FGFR3, FGFR4, FGFR5, Flt-1, Fps, Frk, Jak, KDR, MEK, PDGFR, PIK, PKC, PYK2, Ros, Tie, Tie2, and Zap70. Due to their activity in numerous cellular processes, protein kinases have emerged as important therapeutic targets.
  • Abl1 v-Abl Abelson murine leukemia viral oncogene homolog
  • ALK is a 1620 amino acid transmembrane protein, consisting of extracellular domain with amino-terminal signal peptide, intracellular domain with a juxtamembranous segment harboring a binding site for insulin receptor substrate-1, and a carboxy-terminal kinase domain.
  • ALK is a member of the insulin receptor tyrosine kinases.
  • Echinoderm microtubule-associated protein-like 4 (EML4) is a 120 KDa cytoplasmic protein, which involves in the formation of microtubules and microtubule binding protein .
  • EML4-ALK is a novel fusion gene arising from an inversion on the short arm of chromosome 2 that joined exons 1-13 of EML4 to exons 20-29 of ALK .
  • the presence of EML4-ALK fusion is identified in approximately 3-13%of NSCLC (non-small cell lung cancer) patients.
  • Epidermal growth factor is a widely distributed growth factor that in cancer, can stimulate cancer-cell proliferation, block apoptosis, activate invasion and metastasis, and stimulate angiogenesis (Citri, et al., Nat. Rev. Mol. Cell. Biol. 7: 505, 2006; Hynes, et al., Nat. Rev. Cancer 5: 341, 2005) .
  • the EGF receptor (EGFR or ErbB) is a transmembrane, tyrosine kinase receptor that belongs to a family of four related receptors. The majority of human epithelial cancers are marked by functional activation of growth factors and receptors of this family (Ciardiello, et al., New Eng. J. Med.
  • the human epidermal growth factor receptor (HER) tyrosine kinase family consists of four structurally related cellular receptors: the epidermal growth factor receptor (EGFR; HER1) , HER2 (ErbB2) , HER3 (ErbB3) , and HER4.
  • the compounds that can inhibit protein kinases such as ALK, ROS1, or EGFR kinases activity together can be used to treat human diseases such as cancers.
  • the present invention provides compounds of Formula I:
  • R 1 is C 1-6 alkyl or NH 2 (R 2 R 3 ) C-;
  • R 2 and R 3 are independently H or C 1-6 alkyl.
  • the present invention further provides pharmaceutical compositions comprising a compound of Formula I described above and a pharmaceutically acceptable carrier.
  • the present invention further provides methods for treating or preventing a kinase mediated disorder comprising administrating to a mammalian subject a therapeutically effective amount of any of the compounds of Formula I described above.
  • R 1 is C 1-6 alkyl, or NH 2 (R 2 R 3 ) C-;
  • R 2 and R 3 are independently H or C 1-6 alkyl.
  • the compound of this invention is in the form of a metabolite. In other embodiments, the compound of this invention is in the form of a prodrug. In some embodiments, the compound of this invention is an enantiomer. In other embodiments, the compound of this invention is a diastereomer. In another embodiment, the deuterium enrichment in compounds of this invention is at least about 1%.
  • compositions comprising a compound of the invention and a pharmaceutically acceptable carrier.
  • the compositions are for the treatment of a disease regulated by a protein kinase.
  • the compositions are for the prevention or the treatment of a hyper-proliferative disorder and/or angiogenesis disorder.
  • the pharmaceutical compositions are suitable for oral, parenteral, or intravenous administration.
  • the present invention provides methods for regulating the kinase signaling transduction, said method comprises. administrating to a mammalian subject a therapeutically effective amount of any of the inventive compounds described herein.
  • inventions provide herein methods for treating or preventing an ALK, ROS1, and/or EGFR (including all fusion and/or mutant kinases) mediated disorder, said method comprises administrating to a mammalian subject a therapeutically effective amount of any of the inventive compounds described herein.
  • the present invention provides methods for treating neoplasia.
  • the present invention provides methods for treating cancer diseases, including but not limited to, lung cancer, breast cancer, brain cancer, chronic lymphocytic leukemia, mantle cell lymphoma, and diffuse large B-cell lymphoma.
  • cancer diseases including but not limited to, lung cancer, breast cancer, brain cancer, chronic lymphocytic leukemia, mantle cell lymphoma, and diffuse large B-cell lymphoma.
  • compositions comprising a compound of the present invention in combination with one or more anti-cancer agents for treating neoplasia.
  • the present invention provides methods for treating or preventing of a hyper-proliferative disorder.
  • alkyl is intended to include straight, branched, and cyclic hydrocarbon groups, which contain only single carbon-carbon bonds and which may be unsubstituted or optionally substituted with one or more functional groups.
  • the preferred chain length of an alkyl group is from 1 to 6 carbon atoms.
  • C 1 -C 6 alkyl is intended to include C 1 (methyl) , C 2 (ethyl) , C 3 (n-propyl, isopropyl) , C 4 (e.g. n-butyl, isobutyl, sec-butyl, tert-butyl) , C 5 (e.g. n-pentyl) and C 6 alkyl groups.
  • Alkyl may be substituted or unsubstituted.
  • Illustrative substituted alkyl groups include, but are not limited to, fluoromethyl, difluoromethyl, trifluoromethyl, aminomethyl, aminoethyl, hydoxymethyl, methoxymethyl, 2-fluoroethyl, and 2-methoxyethyl, etc.
  • a pharmaceutically acceptable when used with reference to a compound of the invention is intended to refer to a form of the compound that is safe for administration to a subject.
  • a free base, a salt form, a solvate, a hydrate, a prodrug or derivative form of a compound of this invention which has been approved for mammalian use, via oral ingestion or any other route of administration, by a governing authority or regulatory agency, such as the Food and Drug Administration (FDA) of the United States, is pharmaceutically acceptable.
  • FDA Food and Drug Administration
  • the phrase "effective amount" is intended to quantify the amount of each agent, which will achieve the goal of improvement in disorder severity and the frequency of incidence over treatment of each agent by itself, while avoiding adverse side effects typically associated with alternative therapies.
  • the effective amount in one embodiment, is administered in a single dosage form or in multiple dosage forms.
  • Starting materials of the invention are either known, commercially available, or can be synthesized in analogy to or according to methods that are known in the art. Many starting materials may be prepared according to known processes and, in particular, can be prepared using processes described in the examples. While synthesizing starting materials, functional groups in some cases are protected with suitable protecting groups when necessary. Protecting groups, their introduction and removal are described hereinafter.
  • the steps in some embodiment are performed in an order suitable to prepare the compound, including a procedure described herein or by an alternate order of steps described herein, and in one embodiment, be preceded, or followed, by additional protection/deprotection steps as necessary.
  • the intermediates in some embodiments are isolated or carried on in situ, with or without purification.
  • Synthetic chemistry transformations and protecting group methodologies (protection and deprotection) useful in synthesizing the inhibitor compounds described herein are known in the art and include, for example, those such as described in R. Larock, Comprehensive Organic Transformations, VCH Publishers (1989) ; T.W. Greene and P.G.M.
  • the compounds of this invention in some embodiments also are represented in multiple tautomeric forms.
  • the invention expressly includes all tautomeric forms of the compounds described herein.
  • the compounds in one embodiment also occur in cis-or trans-or E-or Z-double bond isomeric forms. All such isomeric forms of such compounds are expressly included in the present invention.
  • the present invention provides compounds which are capable of modulating one or more signal transduction pathways comprising, but not limited to ALK, ROS1, and EGFR.
  • modulating it is meant that the functional activity of the pathway (or a component thereof) is changed in comparison to its normal activity in the absence of the compound. This effect includes any quality or degree of modulation, including, increasing, agonizing, augmenting, enhancing, facilitating, stimulating, decreasing, blocking, inhibiting, reducing, diminishing, antagonizing, etc.
  • the compounds of the present invention can also modulate one or more of the following processes, including, but not limited to, e.g., cell growth (including, e.g., differentiation, cell survival, and/or proliferation) , tumor cell growth (including, e.g., differentiation, cell survival, and/or proliferation) , tumor regression, endothelial cell growth (including, e.g., differentiation, cell survival, and/or proliferation) , angiogenesis (blood vessel growth) , lymphangiogenesis (lymphatic vessel growth) , and/or hematopoiesis (e.g., T-and B-cell development, dendritic cell development, etc. ) .
  • cell growth including, e.g., differentiation, cell survival, and/or proliferation
  • tumor cell growth including, e.g., differentiation, cell survival, and/or proliferation
  • tumor regression including, e.g., endothelial cell growth (including, e.g., differentiation, cell survival, and/or proliferation)
  • kinase activity it is meant a catalytic activity in which a gamma-phosphate from adenosine triphosphate (ATP) is transferred to an amino acid residue (e.g., serine, threonine, or tyrosine) in a protein substrate.
  • ATP adenosine triphosphate
  • a compound can modulate kinase activity, e.g., inhibiting it by directly competing with ATP for the ATP-binding pocket of the kinase, by producing a conformational change in the enzyme's structure that affects its activity (e.g., by disrupting the biologically-active three-dimensional structure) , by binding to and locking the kinase in an inactive conformation, etc.
  • the amount of compound (s) which is/are administered and the dosage regimen for treating cancer with the compounds and/or compositions of this invention depend on a variety of factors, including the age, weight, sex and medical condition of the subject, the type of disease, the severity of the disease, the route and frequency of administration, and the particular compound employed. Thus, the dosage regimen may vary widely, but can be determined routinely using standard methods.
  • a daily dose of about 0.01 to 500 mg/kg, advantageously between about 0.01 and about 50 mg/kg, more advantageously about 0.01 and about 30 mg/kg, even more advantageously between about 0.1 and about 10 mg/kg may be appropriate, and should be useful for all methods of use disclosed herein.
  • the daily dose can be administered in one to four doses per day.
  • a pharmaceutical composition comprising a compound of this invention in combination with a pharmaceutically acceptable carrier, which includes diluents, excipients, adjuvants and the like (collectively referred to herein as "carrier" materials) as described herein, and, if desired, other active ingredients.
  • carrier includes diluents, excipients, adjuvants and the like (collectively referred to herein as "carrier” materials) as described herein, and, if desired, other active ingredients.
  • carrier includes diluents, excipients, adjuvants and the like
  • An effective dosage amount of a compound of the invention includes an amount less than, equal to or greater than an effective amount of the compound; for example, a pharmaceutical composition in which two or more unit dosages, such as in tablets, capsules and the like, are required to administer an effective amount of the compound, or alternatively, a multi-dose pharmaceutical composition, such as powders, liquids and the like, in which an effective amount of the compound is administered by administering a portion of the composition.
  • Suitable routes of administration include, but are not limited to, oral, intravenous, rectal, aerosol, parenteral, ophthalmic, pulmonary, transmucosal, transdermal, vaginal, otic, nasal, and topical administration.
  • parenteral delivery includes intramuscular, subcutaneous, intravenous, intramedullary injections, as well as intrathecal, direct intraventricular, intraperitoneal, intralymphatic, and intranasal injections.
  • the compounds of the invention may be administered orally.
  • Oral administration may involve swallowing, so that the compound enters the gastrointestinal tract, or buccal or sublingual administration may be employed by which the compound enters the blood stream directly from the mouth.
  • Formulations suitable for oral administration include solid formulations such as tablets, capsules containing particulates, liquids, or powders, lozenges (including liquid-filled) , chews, multi-and nanoparticulates, gels, solid solution, liposome, films (including muco-adhesive) , ovules, sprays and liquid formulations.
  • the compounds of the invention may also be used in fast-dissolving, fast-disintegrating dosage forms such as those described in Expert Opinion in Therapeutic Patents, 11 (6) , 981-986 by Liang and Chen (2001) , the disclosure of which is incorporated herein by reference in its entirety.
  • Formulations for parenteral administration may be formulated to be immediate and/or modified release.
  • Modified release formulations include delayed-, sustained-, pulsed-, controlled-, targeted and programmed release.
  • compounds of the invention may be formulated as a solid, semi-solid, or thixotropic liquid for administration as an implanted depot providing modified release of the active compound. Examples of such formulations include drug-coated stents and PGLA microspheres.
  • the compounds of the invention can be dosed or administered as the sole active pharmaceutical agent, they can also be used in combination with one or more compounds of the invention or in conjunction with other agents.
  • the therapeutic agents can be formulated as separate compositions that are administered simultaneously or sequentially at different times, or the therapeutic agents can be given as a single composition.
  • Compound 6 is synthesized using the procedure described in Scheme 2.
  • the acylation of Compound 1 affords Compound 2, which couples with Compound 3 with n-propylphosphonoic anhydride (T3P) to generate Compound 4.
  • T3P n-propylphosphonoic anhydride
  • Deprotection of Fmoc group leads to the synthesis of Compound 6 (Scheme 2) .
  • DMF means N, N-dimethylformamide.
  • DCM dichloromethane
  • DIPEA means diisopropyl ethylamine.
  • DCC means Dicyclohexylcarbodiimide.
  • THF means tetrahydrofuran
  • EA means ethyl acetate.
  • m-CPBA meta-Chloroperoxybenzoic acid.
  • Boc-Gly-OH means N- (tert-Butoxycarbonyl) glycine.
  • BOP means (Benzotriazol-1-yloxy) tris (dimethylamino) phosphonium hexafluorophosphate.
  • Pd (dppf) Cl 2 means [1, 1′-Bis (diphenylphosphino) ferrocene] dichloropalladium.
  • T3P means n-propylphosphonoic anhydride
  • Et 3 N means triethyl amine
  • Fmoc Fluorenylmethyloxycarbonyl
  • cataCXium means Di (1-adamantyl) -n-butylphosphine
  • Pd (OAc) 2 means palladium acetate
  • CYP Cytochrome P450
  • EGFR epidermal growth factor receptor
  • ERBB4 means receptor tyrosine-protein kinase erbB-4
  • Example 1 The synthesis of (10R) -7-acetamido-12-fluoro-2, 10, 16-trimethyl-15-oxo-10, 15, 16, 17-tetrahydro-2H-8, 4- (metheno) pyrazolo [4, 3-h] [2, 5, 11] -benzoxadiazacyclotetradecine-3-carbonitrile (Compound 10) .
  • Example 2 The synthesis of (10R) -7- (2-aminoacetyl) amino-12-fluoro-2, 10, 16-trimethyl-15-oxo-10, 15, 16, 17-tetra hydro-2H-8, 4- (metheno) pyrazolo [4, 3-h] [2, 5, 11] -benzoxadiazacyclotetradecine-3-carb onitrile (Compound 6) .
  • the Fmoc-glycine (763.4 mg, 2.6 mmol, 20.0 eq) was added into SOCl 2 (16.0 mL) and the mixture was heated to reflux for 2 h. Evaporation in vacuum to give a white solid.
  • the solid was dissolved in DCM (8.0 mL) , and a solution of (10R) -7-amino-12-fluoro-2, 10, 16-trimethyl-15-oxo-10, 15, 16, 17-tetrahydro-2H-8, 4- (metheno) pyrazolo [4, 3-h] [2, 5, 11] -benzoxadiazacyclotetradecine-3-carbonitrile (Compound A, 50.0 mg, 0.13 mmol, 1.0 eq) in pyridine (16.0 mL) was added in two portions.
  • the compounds defined in the present invention possess anti-proliferation activity. These properties may be assesses, for example, using one or more of the procedures set out below:
  • kinase-tagged T7 phage strains were prepared in an E. coli host derived from the BL21 strain. E. coli were grown to log-phase and infected with T7 phage and incubated with shaking at 32°C until lysis. The lysates were centrifuged and filtered to remove cell debris. The remaining kinases were produced in HEK-293 cells and subsequently tagged with DNA for qPCR detection. Streptavidin-coated magnetic beads were treated with biotinylated small molecule ligands for 30 minutes at room temperature to generate affinity resins for kinase assays.
  • the liganded beads were blocked with excess biotin and washed with blocking buffer (SeaBlock (Pierce) , 1%BSA, 0.05%Tween 20, 1 mM DTT) to remove unbound ligand and to reduce non-specific binding. Binding reactions were assembled by combining kinases, liganded affinity beads, and test compounds in 1x binding buffer (20%SeaBlock, 0.17x PBS, 0.05%Tween 20, 6 mM DTT) . All reactions were performed in polystyrene 96-well plates in a final volume of 0.135 ml.
  • blocking buffer SeaBlock (Pierce) , 1%BSA, 0.05%Tween 20, 1 mM DTT
  • the assay plates were incubated at room temperature with shaking for 1 hour and the affinity beads were washed with wash buffer (1x PBS, 0.05%Tween 20) .
  • the beads were then re-suspended in elution buffer (1x PBS, 0.05%Tween 20, 0.5 ⁇ M non-biotinylated affinity ligand) and incubated at room temperature with shaking for 30 minutes.
  • the kinase concentration in the eluates was measured by qPCR.
  • Table A lists a compound representative of the invention and its inhibition activity in kinases assays.
  • Compound 6 is a potent inhibitor (420 nM) for a mutant EGFR (L747-E749del, A750P) .
  • the assay plate was centrifuged at 800 g for 10 min.
  • the medium was aspirated, 150 ⁇ l DMSO was added into each well.
  • the plate was gently shaken for 10 min.
  • Table B lists compounds representative of the invention and their activity in cell assays.
  • HLM Human liver microsomes
  • Incubation mixtures in duplicate contain pooled human liver microsome (0.1 mg/mL) , 3.3 mM MgCl 2 , CYP 3A4 probe substrate testosterone (50 ⁇ M) , specific inhibitor or test compounds (30, 10, 3, 0.1, 0.03, 0.01, 0.003, 0.01 ⁇ M) in 0.1 M potassium phosphate buffer (total volume 0.1 mL) .
  • Negative control contains 0.1 M phosphate buffer instead of specific inhibitor or test compound.
  • the final concentrations of DMSO and acetonitrile were equal or less than 0.1%.
  • the mixtures are pre-incubated for 10 min at 37 °C. Then, 1 mM NADPH is added to initiate reaction. Following a 10-min incubation at 37 °C, the reactions are terminated by the addition of 300 ⁇ L acetonitrile containing an internal standard. The formation of the corresponding products is detected by LC/MS/MS.
  • LCMS method A Waters ACQUITY UPLC system coupled API 4000 Qtrap system was used. The mass spectrometer is equipped with Turbo Ion Spray (ESI) Interface (Applied Biosystems, Concord, Ontario, Canada) . Analyst 1.5 software packages (Applied Biosystems) were used to control the LC-MS/MS system, as well as for data acquisition and processing. Chromatographic separation was achieved on Waters ACQUITY UPLC BEH C18 column (50x2.1 mm ID, 1.7 ⁇ m) . The column temperature was maintained at ambient temperature (25 °C) . The mobile phase A is pure water supplemented with 0.1%formic acid (v/v) . The mobile phase B is acetonitrile supplemented with 0.1%formic acid (v/v) . The flow rate was maintained at 0.6 mL/min.
  • Compound PF-06463922 showed moderate Cytochrome P450 3A4 inhibition.
  • Compound 6 and 10 showed low Cytochrome P450 3A4 inhibition, which indicates they have better safety profiles.
  • Incubation 1.0 ⁇ M of test compound in duplicate was incubated in blood at 37 °C. Aliquots of 50 ⁇ L sample were collected at 0 h, 0.25 h, 0.5 h, 1 h, and 2 h.
  • Sample preparation Reactions were terminated at various time points (0, 0.25, 0.5, 1, 2 h) by adding 150 ⁇ L of ice-cold acetonitrile containing an internal standard. Centrifuge the plate (4000 rpm, 15 min) . 100 ⁇ L of supernatants were transferred into a daughter plate containing 200 ⁇ L of H 2 O and 0.1%formic acid (v/v) in each well. The samples were analyzed with UPLC-MS/MS.
  • the peak area ratio of test compound to internal standard is plotted as a percentage of the relevant zero time point control (%Remained) for each reaction.
  • the rate of metabolism (k) is the slope of the linear regression from log percentage remaining versus incubation time.
  • the T1/2 is calculated as -0.693/k.
  • Table D lists compounds representative of the invention and their rate of metabolism (k) in in Rat whole blood.
  • Compound 6 was found to have much larger K value than PF-06463922 and Compound 10. Faster metabolism may result in much less toxicity for chemical compounds in a test subject.
  • sample solution 2 mg of Compound A or Compound 6 was added individually to 2 mL eppendorf tube (EP) , followed by addition of 1 mL of pH 7.0 or 10.0 buffer solution (20 mM) . The solution was shook for 2 minutes and left for 30 minutes at 25°C. After standing for 30 minutes, precipitate was formed in the bottom of the EP. The solutions was filtered through 0.2 um membrane filter, and then diluted by 50 times with water.

Abstract

L'invention concerne de nouveaux composés macrocycles substitués, des sels pharmaceutiquement acceptables, des solvates et des hydrates de ceux-ci. Les composés et compositions de l'invention présentent une activité inhibitrice des protéines kinases, et devraient donc s'avérer utiles pour le traitement de maladies et affections induites par les protéines kinases.
PCT/CN2015/087343 2014-08-20 2015-08-18 Macrocycles substitués utilisés comme inhibiteurs des kinases et leurs méthodes d'utilisation WO2016026423A1 (fr)

Priority Applications (4)

Application Number Priority Date Filing Date Title
JP2017510559A JP6609308B2 (ja) 2014-08-20 2015-08-18 キナーゼ阻害剤としての置換マクロサイクル
EP15833622.2A EP3183256A4 (fr) 2014-08-20 2015-08-18 Macrocycles substitués utilisés comme inhibiteurs des kinases et leurs méthodes d'utilisation
US15/500,927 US10159663B2 (en) 2014-08-20 2015-08-18 Substituted macrocycles useful as kinases inhibitors and methods of use thereof
CN201580043435.7A CN107207528B (zh) 2014-08-20 2015-08-18 作为激酶抑制剂的经取代大环及其使用方法

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201461070250P 2014-08-20 2014-08-20
US61/070,250 2014-08-20

Publications (1)

Publication Number Publication Date
WO2016026423A1 true WO2016026423A1 (fr) 2016-02-25

Family

ID=55350197

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2015/087343 WO2016026423A1 (fr) 2014-08-20 2015-08-18 Macrocycles substitués utilisés comme inhibiteurs des kinases et leurs méthodes d'utilisation

Country Status (1)

Country Link
WO (1) WO2016026423A1 (fr)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018137679A1 (fr) * 2017-01-25 2018-08-02 Teligene Ltd Procédé de préparation de (10r) -7- (2-aminoacétyl) amino-12-fluoro-2, 10, 16-triméthyl-15-oxo-10, 15, 16, 17-tétrahydro-2h-8, 4- (metheno) pyrazolo [4, 3-h] [2, 5, 11] -benzoxadiazacyclotétradécine-3-carbonitrile
CN110950890A (zh) * 2018-09-27 2020-04-03 北京赛林泰医药技术有限公司 取代的咪唑并[4,5-c]喹啉大环化合物作为多靶点激酶抑制剂
CN111201235A (zh) * 2017-10-10 2020-05-26 辉瑞公司 劳拉替尼游离碱水合物的结晶形式
WO2020228747A1 (fr) 2019-05-14 2020-11-19 Teligene Ltd Macrocycles substitués utiles en tant qu'inhibiteurs de kinase
CN115746023A (zh) * 2022-10-27 2023-03-07 复旦大学 一种作为蛋白激酶抑制剂的含吲唑结构的杂环大环化合物及其制备方法

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011138751A2 (fr) * 2010-05-04 2011-11-10 Pfizer Inc. Dérivés hétérocycliques destinés au traitement de maladies
WO2013132376A1 (fr) * 2012-03-06 2013-09-12 Pfizer Inc. Dérivés macrocycliques pour le traitement de maladies prolifératives
WO2014207606A1 (fr) * 2013-06-28 2014-12-31 Pfizer Inc. Formes solides d'un inhibiteur de kinases macrocycliques
WO2015050989A2 (fr) * 2013-10-01 2015-04-09 Cs Therapeutics Inc. Composés macrocycliques destinés au traitement de maladies prolifératives

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011138751A2 (fr) * 2010-05-04 2011-11-10 Pfizer Inc. Dérivés hétérocycliques destinés au traitement de maladies
WO2013132376A1 (fr) * 2012-03-06 2013-09-12 Pfizer Inc. Dérivés macrocycliques pour le traitement de maladies prolifératives
WO2014207606A1 (fr) * 2013-06-28 2014-12-31 Pfizer Inc. Formes solides d'un inhibiteur de kinases macrocycliques
WO2015050989A2 (fr) * 2013-10-01 2015-04-09 Cs Therapeutics Inc. Composés macrocycliques destinés au traitement de maladies prolifératives

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP3183256A4 *

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018137679A1 (fr) * 2017-01-25 2018-08-02 Teligene Ltd Procédé de préparation de (10r) -7- (2-aminoacétyl) amino-12-fluoro-2, 10, 16-triméthyl-15-oxo-10, 15, 16, 17-tétrahydro-2h-8, 4- (metheno) pyrazolo [4, 3-h] [2, 5, 11] -benzoxadiazacyclotétradécine-3-carbonitrile
CN111201235A (zh) * 2017-10-10 2020-05-26 辉瑞公司 劳拉替尼游离碱水合物的结晶形式
CN111201235B (zh) * 2017-10-10 2023-02-10 辉瑞公司 劳拉替尼游离碱水合物的结晶形式
CN110950890A (zh) * 2018-09-27 2020-04-03 北京赛林泰医药技术有限公司 取代的咪唑并[4,5-c]喹啉大环化合物作为多靶点激酶抑制剂
WO2020228747A1 (fr) 2019-05-14 2020-11-19 Teligene Ltd Macrocycles substitués utiles en tant qu'inhibiteurs de kinase
CN112533927A (zh) * 2019-05-14 2021-03-19 苏州韬略生物科技有限公司 可用作激酶抑制剂的被取代的大环
CN115746023A (zh) * 2022-10-27 2023-03-07 复旦大学 一种作为蛋白激酶抑制剂的含吲唑结构的杂环大环化合物及其制备方法

Similar Documents

Publication Publication Date Title
US10159663B2 (en) Substituted macrocycles useful as kinases inhibitors and methods of use thereof
US9694011B2 (en) Substituted pyrazolopyrimidines as kinases inhibitors
EP2833886B1 (fr) Quinoléines substituées en tant qu'inhibiteurs de tyrosine kinase de bruton
WO2016026423A1 (fr) Macrocycles substitués utilisés comme inhibiteurs des kinases et leurs méthodes d'utilisation
EP3102571B1 (fr) Pyrimidines substituées utiles en tant qu'inhibiteurs de kinase d'egfr-t790m
WO2013173254A1 (fr) Composés bicycliques utilisés en tant qu'inhibiteurs des kinases
MX2011004683A (es) Antagonistas de la via hedgehog de ftalazina disustituida.
CN111542522B (zh) 可用作激酶抑制剂的被取代的吡唑并嘧啶
EP3844166B1 (fr) Macrocycles substitués utiles en tant qu'inhibiteurs de kinase
US20140350050A1 (en) Pyridine compounds as inhibitors of kinase
US11427559B2 (en) Substituted quinolines useful as kinase inhibitors
JP2014532063A (ja) キナーゼ阻害剤としてのキナゾリン誘導体およびその使用方法
WO2014110466A1 (fr) Composés pyridine utilisés en tant qu'inhibiteurs de pi3 kinase
WO2014139391A1 (fr) Utilisation de composés de pyridine substituée en tant qu'inhibiteurs de kinases

Legal Events

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

Ref document number: 15833622

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 15500927

Country of ref document: US

ENP Entry into the national phase

Ref document number: 2017510559

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

REEP Request for entry into the european phase

Ref document number: 2015833622

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2015833622

Country of ref document: EP