WO2011054846A1 - Imidazo [4, 5-c] quinoline derivates as bromodomain inhibitors - Google Patents

Imidazo [4, 5-c] quinoline derivates as bromodomain inhibitors Download PDF

Info

Publication number
WO2011054846A1
WO2011054846A1 PCT/EP2010/066699 EP2010066699W WO2011054846A1 WO 2011054846 A1 WO2011054846 A1 WO 2011054846A1 EP 2010066699 W EP2010066699 W EP 2010066699W WO 2011054846 A1 WO2011054846 A1 WO 2011054846A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
dimethyl
isoxazolyl
salt
formula
Prior art date
Application number
PCT/EP2010/066699
Other languages
French (fr)
Inventor
Anne Marie Jeanne Bouillot
Frederic Donche
Francoise Jeanne Gellibert
Yann Lamotte
Olivier Mirguet
Original Assignee
Glaxosmithkline Llc
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 Glaxosmithkline Llc filed Critical Glaxosmithkline Llc
Priority to ES10771770.4T priority Critical patent/ES2446358T3/en
Priority to US13/503,722 priority patent/US8557984B2/en
Priority to JP2012537382A priority patent/JP5766708B2/en
Priority to EP10771770.4A priority patent/EP2496576B1/en
Publication of WO2011054846A1 publication Critical patent/WO2011054846A1/en

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
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • the present invention relates to novel compounds, pharmaceutical compositions containing such compounds and to their use in therapy.
  • the genomes of eukaryotic organisms are highly organised within the nucleus of the cell.
  • the long strands of duplex DNA are wrapped around an octomer of histone proteins (most usually comprising two copies of histones H2A, H2B H3 and H4) to form a nucleosome.
  • This basic unit is then further compressed by the aggregation and folding of nucleosomes to form a highly condensed chromatin structure.
  • a range of different states of condensation are possible, and the tightness of this structure varies during the cell cycle, being most compact during the process of cell division.
  • Chromatin structure plays a critical role in regulating gene transcription, which cannot occur efficiently from highly condensed chromatin.
  • the chromatin structure is controlled by a series of post translational modifications to histone proteins, notably histones H3 and H4, and most commonly within the histone tails which extend beyond the core nucleosome structure. These modifications include acetylation, methylation, phosphorylation, ubiquitinylation, SUMOylation. These epigenetic marks are written and erased by specific enzymes, which place the tags on specific residues within the histone tail, thereby forming an epigenetic code, which is then interpreted by the cell to allow gene specific regulation of chromatin structure and thereby transcription. Histone acetylation is most usually associated with the activation of gene transcription, as the modification loosens the interaction of the DNA and the histone octomer by changing the electrostatics.
  • Bromodomains are small (-1 10 amino acid) distinct domains within proteins that bind to acetylated lysine resides commonly but not exclusively in the context of histones. There is a family of around 50 proteins known to contain bromodomains, and they have a range of functions within the cell.
  • the BET family of bromodomain containing proteins comprises 4 proteins (BRD2, BRD3, BRD4 and BRD-t) which contain tandem bromodomains capable of binding to two acetylated lysine residues in close proximity, increasing the specificity of the interaction.
  • BRD2 and BRD3 are reported to associate with histones along actively transcribed genes and may be involved in facilitating transcriptional elongation (Leroy et al, Mol. Cell.
  • BRD4 appears to be involved in the recruitment of the pTEF- ⁇ complex to inducible genes, resulting in phosphorylation of RNA polymerase and increased transcriptional output (Hargreaves et al, Cell, 2009 138(1 ): 129-145). It has also been reported that BRD4 or BRD3 may fuse with NUT (nuclear protein in testis) forming novel fusion oncogenes, BRD4-NUT or BRD3-NUT, in a highly malignant form of epithelial neoplasia (French et al. Cancer Research, 2003, 63, 304-307 and French et al.
  • BRD-NUT fusion proteins contribute to carcinogenesis (Oncogene, 2008, 27, 2237-2242).
  • BRD-t is uniquely expressed in the testes and ovary. All family members have been reported to have some function in controlling or executing aspects of the cell cycle, and have been shown to remain in complex with chromosomes during cell division - suggesting a role in the maintenance of epigenetic memory. In addition some viruses make use of these proteins to tether their genomes to the host cell chromatin, as part of the process of viral replication (You et al Cell, 2004 1 17(3):349-60).
  • Japanese patent application JP2008-15631 1 discloses a benzimidazole derivative which is said to be a BRD2 bromodomain binding agent which has utility with respect to virus infection / proliferation.
  • Patent application WO2009084693A1 discloses a series of thienotriazolodiazepiene derivatives that are said to inhibit the binding between an acetylated histone and a bromodomain containing protein which are said to be useful as anti-cancer agents.
  • bromodomain inhibitors A novel class of compounds which inhibit the binding of bromodomains with its cognate acetylated proteins, more particularly a class of compounds that inhibit the binding of BET family bromodomains to acetylated lysine residues. Such compounds will hereafter be referred to as "bromodomain inhibitors".
  • a compound of formula (I) or a salt thereof more particularly a compound of formula (I) or a pharmaceutically acceptable salt thereof
  • a pharmaceutical composition comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof and one or more pharmaceutically acceptable carriers, diluents or excipients.
  • a compound of formula (I), or a pharmaceutically acceptable salt thereof for use in therapy, in particular in the treatment of diseases or conditions for which a bromodomain inhibitor is indicated.
  • a method of treating diseases or conditions for which a bromodomain inhibitor is indicated in a subject in need thereof which comprises administering a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof.
  • a compound of formula (I), or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for the treatment of diseases or conditions for which a bromodomain inhibitor is indicated.
  • the present invention relates to compounds of formula (I) or a salt thereof
  • A is a group selected from the following:
  • ⁇ Y represents CH or N with the proviso that when X is N, Y is CH;
  • R x represents O or S
  • R 1 represents C-
  • R 2 is hydrogen or C-
  • R a represents H, C-
  • R b represents H or C-
  • R a and R b together with the N to which they are attached form a 5- or 6-membered heterocyclyl; • R represents H, C-
  • R 3 represents hydrogen
  • R 4 represents hydrogen, cyano or C-
  • ⁇ Z represents O; or when R 4 represents hydrogen and A is a group selected from (i) or (ii) and wherein R x represents O, Z may additionally represent NH;
  • R 5 represents hydrogen or C-
  • R 6 represents hydrogen or C-
  • ⁇ n 0, 1 or 2;
  • aromatic group refers to a 5- to 7-membered monocyclic aromatic group such as phenyl, or a 8- to 1 1 -membered bicyclic aromatic group such as naphthyl, indenyl or azulenyl.
  • heterocyclyl refers to a 5- or 6-membered non-aromatic, saturated or unsaturated ring comprising 1 , 2, or 3 heteroatoms selected from O, N and S.
  • heterocyclyls include morpholinyl, piperidinyl, tetrahydropyranyl, and piperazinyl.
  • heteromatic group refers to a 5- or 6-membered monocyclic aromatic group wherein 1 , 2, 3, 4 of the carbon atoms are replaced by a heteroatom independently selected from O, S and N; or to a 8- to 1 1 -membered bicyclic aromatic group wherein 1 , 2, 3, 4 or 5 of the carbon atoms are replaced by a heteroatom independently selected from O, S and N.
  • Examples of 5- or 6-membered monocyclic heteroaromatic groups include pyrrolinyl, furanyl, thienyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, oxadiazolyl, thiadiazolyl, pyridinyl, triazolyl, triazinyl, pyridazyl, pyrimidinyl, isothiazolyl, isoxazolyl, pyrazinyl, pyrazolyl and pyrimidinyl.
  • Examples of 8- to 1 1 - membered bicyclic heteroaromatic groups include 6H- thieno[2,3-b]pyrrolyl, imidazo[2,1 -b][1 ,3]thiazolyl, imidazo[5,1 -b][1 ,3]thiazolyl, indolyl, isoindolyl, indazolyl, benzimidazolyl, [1 ,3]thiazolo[3,2-fc>][1 ,2,4]triazolyl, benzoxazolyl e.g.
  • alkyl refers to straight or branched hydrocarbon chains containing the specified number of carbon atoms.
  • .galkyl means a straight or branched alkyl containing at least 1 , and at most 6, carbon atoms.
  • alkyl as used herein include, but are not limited to, methyl, ethyl, n-propyl, n-butyl, n- pentyl, n-hexyl, isobutyl, isopropyl, t-butyl and 1 ,1 -dimethylpropyl.
  • C3_gcycloalkyls examples include cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl.
  • alkoxy refers to a straight or branched alkoxy group containing the specified number of carbon atoms.
  • .galkoxy means a straight or branched alkoxy group containing at least 1 , and at most 6, carbon atoms.
  • alkoxy as used herein include, but are not limited to, methoxy, ethoxy, propoxy, prop-2- oxy, butoxy, but-2-oxy, 2-methylprop-1 -oxy, 2-methylprop-2-oxy, pentoxy or hexyloxy.
  • halogen refers to the elements fluorine, chlorine, bromine and iodine, for example, fluorine, chlorine and bromine.
  • substituted refers to substitution with the named substituent or substituents, multiple degrees of substitution being allowed unless otherwise stated.
  • substituent When the substituent is on a ring comprising a heteroatom the substituent may be located on a carbon or a heteroatom, if the latter is appropriate.
  • X is CH. In one embodiment, Y is CH.
  • Z is O.
  • Z is NH (and in consequence R 4 represents hydrogen, A is a group selected from (i) or (ii) as defined above, and R x represents O).
  • R x is O.
  • n is 0 or 1 , such as 1 .
  • A is a group of formula (i) or (ii) as described above.
  • R 1 include, pyridinyl such as pyridin-2-yl, phenyl optionally substituted by 1 or 2 groups such as 1 substitutent, wherein said substituents are independently selected from methyl, t-butyl, fluoro, chloro, and -OCF 3 .
  • Optional substituents on the phenyl may, for example, be in the ortho or para position.
  • R 1 is a heteroaromatic group optionally substituted by one or two groups selected from hydroxy, C-
  • the heteroaromatic group is pyridyl.
  • the heteroaromatic group is selected from furanyl, thienyl, isoxazolyl, thiazolyl, pyrazolyl, pyrazinyl and pyrimidinyl.
  • n is 0 and R 1 is phenyl substituted by Ci -6 alkyl (such as phenyl substituted by t-butyl), or phenyl substituted by haloCi -6 alkoxy (such as OCF 3 ).
  • n 1 and R 1 is unsubstituted phenyl.
  • n is 1 or 2 such as 1 and R 1 is phenyl optionally substituted by, for example 1 or 2 halogens such as chlorine, and/or fluoro.
  • R 2 is hydrogen or methyl.
  • R 2a is H, C-i ⁇ alkyl, (CH2) m OH, (CH2) m C ⁇
  • R a represents H, C-
  • R b represents H or C-
  • R a and R b together with the N to which they are attached form a 5 or 6 membered heterocyclyl
  • R 6 represents H or C ⁇
  • n 1 , 2 or 3;
  • R 2a is hydrogen, C-
  • R 2a is tetrahydropyranyl. In one embodiment R 2b is hydrogen.
  • R 2b is (CH2)2C-
  • R a and R b together with the N to which they are attached form a 5 or 6 membered heterocyclyl such as morpholine, piperidine or pyrrolidine.
  • R 4 is hydrogen
  • R 5 is hydrogen or -OCI-13 such as -OCH3.
  • X is CH, Y is CH and R 5 is -OCH3, for example in the 8 position.
  • X is N, Y is CH and R 5 is -OCH3, for example in the 8 position.
  • the compounds of the present invention have a formula (IA):
  • the compounds of the present invention have the formula (IB):
  • n may, for example, represent 1.
  • R 1 may represent, for example pyridinyl such as pyridin-2-yl or phenyl.
  • R 5 may represent, for example, -OCH3.
  • compounds of the invention have the formula (IC)
  • R 1 , R 2 , R 3 , R 2a and n are defined above for compounds of formula (I).
  • Specific compounds of formula (I) include Examples 1 - 226 as described herein or a salt thereof, in particular a pharmaceutically acceptable salt thereof.
  • the present invention covers compounds of formula (I) as the free base and as salts thereof, for example as a pharmaceutically acceptable salt thereof.
  • the invention relates to compounds of formula (I) or a pharmaceutically acceptable salt thereof.
  • salts of the compounds of formula (I) are desirably pharmaceutically acceptable.
  • Suitable pharmaceutically acceptable salts can include acid or base addition salts.
  • the term 'pharmaceutically acceptable salt' means any pharmaceutically acceptable salt or solvate of a compound of formula (I), which upon administration to the recipient is capable of providing (directly or indirectly).
  • the term 'pharmaceutically acceptable salt' means any pharmaceutically acceptable salt of a compound of formula (I), which upon administration to the recipient is capable of providing (directly or indirectly).
  • suitable salts see Berge et al., J. Pharm. Sci., 66:1 -19, (1977).
  • a pharmaceutically acceptable salt may be readily prepared by using a desired acid or base as appropriate. The resultant salt may precipitate from solution and be collected by filtration or may be recovered by evaporation of the solvent.
  • a pharmaceutically acceptable base addition salt can be formed by reaction of a compound of formula (I) with a suitable inorganic or organic base, (e.g. triethylamine, ethanolamine, triethanolamine, choline, arginine, lysine or histidine), optionally in a suitable solvent, to give the base addition salt which is usually isolated, for example, by crystallisation and filtration.
  • a suitable inorganic or organic base e.g. triethylamine, ethanolamine, triethanolamine, choline, arginine, lysine or histidine
  • Pharmaceutically acceptable base salts include ammonium salts, alkali metal salts such as those of sodium and potassium, alkaline earth metal salts such as those of calcium and magnesium and salts with organic bases, including salts of primary, secondary and tertiary amines, such as isopropylamine, diethylamine, ethanolamine, trimethylamine, dicyclohexyl amine and N-methyl-D-glucamine.
  • a pharmaceutically acceptable acid addition salt can be formed by reaction of a compound of formula (I) with a suitable inorganic or organic acid (such as hydrobromic, hydrochloric, sulphuric, nitric, phosphoric, succinc, maleic, acetic, propionic, fumaric, citric, tartaric, lactic, benzoic, salicylic, glutamaic, aspartic, p-toluenesulfonic, benzenesulfonic, methanesulfonic, ethanesulfonic, naphthalenesulfonic such as 2- naphthalenesulfonic, or hexanoic acid), optionally in a suitable solvent such as an organic solvent, to give the salt which is usually isolated for example by crystallisation and filtration.
  • a suitable inorganic or organic acid such as hydrobromic, hydrochloric, sulphuric, nitric, phosphoric, succinc, maleic, acetic
  • a pharmaceutically acceptable acid addition salt of a compound of formula (I) can comprise or be for example a hydrobromide, hydrochloride, sulfate, nitrate, phosphate, succinate, maleate, acetate, propionate, fumarate, citrate, tartrate, lactate, benzoate, salicylate, glutamate, aspartate, p-toluenesulfonate, benzenesulfonate, methanesulfonate, ethanesulfonate, naphthalenesulfonate (e.g. 2-naphthalenesulfonate) or hexanoate salt.
  • Other non-pharmaceutically acceptable salts e.g. formates, oxalates or trifluoroacetates, may be used, for example in the isolation of the compounds of formula (I), and are included within the scope of this invention.
  • the invention includes within its scope all possible stoichiometric and non-stoichiometric forms of the salts of the compounds of formula (I).
  • the invention includes within its scope all possible stoichiometric and non-stoichiometric forms of the solvates of the compounds of formula (I).
  • the invention encompasses all prodrugs, of the compound of formula (I) or a pharmaceutically acceptable salt thereof, which upon administration to the recipient is capable of providing (directly or indirectly) the compound of formula (I) or a pharmaceutically acceptable salt thereof, or an active metabolite or residue thereof.
  • prodrugs of the compound of formula (I) or a pharmaceutically acceptable salt thereof, which upon administration to the recipient is capable of providing (directly or indirectly) the compound of formula (I) or a pharmaceutically acceptable salt thereof, or an active metabolite or residue thereof.
  • Such derivatives are recognizable to those skilled in the art, without undue experimentation. Nevertheless, reference is made to the teaching of Burger's Medicinal Chemistry and Drug Discovery, 5 th Edition, Vol 1 : Principles and Practice, which is incorporated herein by reference to the extent of teaching such derivatives.
  • the compounds of formula (I) may be in crystalline or amorphous form. Furthermore, some of the crystalline forms of the compounds of formula (I) may exist as polymorphs, which are included within the scope of the present invention. Polymorphic forms of compounds of formula (I) may be characterized and differentiated using a number of conventional analytical techniques, including, but not limited to, X-ray powder diffraction (XRPD) patterns, infrared (IR) spectra, Raman spectra, differential scanning calorimetry (DSC), thermogravimetric analysis (TGA) and solid state nuclear magnetic resonance (SSNMR).
  • XRPD X-ray powder diffraction
  • IR infrared
  • Raman spectra Raman spectra
  • DSC differential scanning calorimetry
  • TGA thermogravimetric analysis
  • SSNMR solid state nuclear magnetic resonance
  • Certain compounds described herein may contain one or more chiral atoms so that optical isomers, e.g. enantiomers or diastereoisomers may be formed. Accordingly, the present invention encompasses all isomers of the compounds of formula (I) whether as individual isomers isolated such as to be substantially free of the other isomer (i.e. pure) or as mixtures (i.e. racemates and racemic mixtures).
  • An individual isomer isolated such as to be substantially free of the other isomer (i.e. pure) may be isolated such that less than 10%, particularly less than about 1 %, for example less than about 0.1 % of the other isomer is present. Separation of isomers may be achieved by conventional techniques known to those skilled in the art, e.g. by fractional crystallisation, chromatography or HPLC.
  • Certain compounds of formula (I) may exist in one of several tautomeric forms. It will be understood that the present invention encompasses all tautomers of the compounds of formula (I) whether as individual tautomers or as mixtures thereof.
  • the present invention provides a process for the preparation of a compound of formula (I) wherein Z represents O, the process comprising the step of reacting a compound of formula (II):
  • R 2a COOH, R 2a CHO or R 2a COCI wherein R 2a is as defined above for compounds of formula (I), or
  • Step a) may be effected by refluxing in a suitable solvent such as acetic acid.
  • R 2a COOH is the reagent
  • activation of the starting material with a reagent such as HOBT may be required.
  • the coupling reaction may be performed in the presence of a coupling agent such as EDCI and an organic base such as triethylamine, in a suitable solvent, for example a polar aprotic solvent such as dichloromethane.
  • an acid chloride R 2a COCI When an acid chloride R 2a COCI is employed, prior to refluxing in acetic acid, the acid chloride is allowed to react with the compound of formula (II) in a suitable solvent, for example a polar aprotic solvent such as dichloromethane in the presence of an organic base such as triethylamine.
  • a suitable solvent for example a polar aprotic solvent such as dichloromethane
  • Step b) may be performed at 80°C for a period of about 2h, followed by treatment at an elevated temperature, for example, 180°C for a period of about 3h in a solvent such as diphenylether.
  • Step c) may be effected at elevated temperatures, for example above 50°C such as 80°C, under basic conditions, for example in the presence of triethylamine in a suitable solvent, for example an alcohol such as ethanol.
  • a suitable solvent for example an alcohol such as ethanol.
  • Step d) may be effected at low temperature, for example sodium nitrite is added at 0°C, after which the reaction mixture is allowed to warm to room temperature and continue reacting for about 18h.
  • the imidazole derivatives of general formula (IE) may be prepared (scheme 1 ) by reacting the amino derivatives (IIA) with aldehydes (R 2 CHO) in acetic acid at reflux, or by coupling the derivatives (IIA) with carboxylic acid of general formula R 2 COOH with HOBT, EDCI in dichloromethane in presence of triethylamine, followed by cyclisation in acetic acid at reflux, or by coupling with acid chloride of general formula R 2 COCI in presence of triethylamine followed by cyclisation in acetic acid at reflux.
  • Compounds of formula (I) wherein R 4 represents H or alkyl, and A represents an imidazolone can be prepared as shown in Scheme 2 below.
  • the imidazolone compounds of general formula (IF) can be prepared, according to scheme 2, by reacting the compounds (IIA) with Boc anhydride followed by a cyclisation in diphenyl ether at 180°C.
  • the imidazolone compounds (IF) may be prepared using Hoffman-type rearrangement conditions (Scheme 3). In this case the compounds (MB) are reacted with bis(trifluoroacetoxy)iodo]benzene, to give after internal cyclisation the imidazolone compounds (IF).
  • the imidazolone compounds (IF) may also be prepared using Curtius rearrangement conditions (Scheme 4). In this case the acid compounds (IIC) are reacted with diphenyl phosphoryl azide in the presence of triethylamine for a period of approximately 2 to 18h to give the imidazolone compounds (IF).
  • the imidazol-2-thione derivatives of general formula (IG) can be prepared by reacting the compounds of general formula (IIA) with carbon disulfide in the presence of triethylamine in a solvent such as ethanol at 60°-80°C for approximately 18h.
  • the starting material (IE) in Scheme 7 can be prepared by an analogous method to that described above in Scheme 1.
  • Compounds of general formula (I I A) wherein R 4 is hydrogen and alkyl may be prepared according to Scheme 8.
  • Compounds (IIIA) are reacted with sodium hydroxide in ethanol under reflux for 5h. After treatment with HCI N, the resulting compounds of general formula (VA) are de-carboxylated in diphenylether under reflux for 2h.
  • Nitration of compounds (VA) is preformed with nitric acid in propanoic acid at room temperature, followed by heating the reaction mixture to a range of temperature 100-125°C for 1 to 2h.
  • Compounds (VIA) are reacted with POCI 3 in toluene under reflux for approximately 18h.
  • the chloro compounds (VI IA) are then reacted with amines of general formula R 1 (R 2 R 3 C) n NH 2 in a solvent such as acetonitrile at 60°C for 2h.
  • the resulting compounds may then be reduced with SnCl 2 ,2H 2 0 in ethanol/HCI or ethanol/THF at a temperature in the range 40°C to reflux for approximately 1 to 3h, to give compounds of general formula (I I A).
  • the 3,5-dimethylisoxazole boronic acid may be reacted with 3-iodo or 3-bromo derivatives using Suzuki coupling conditions to give the corresponding amino compounds.
  • the 3-(3,5-dimethyl-4-isoxazolyl)aniline may be reacted with diethyl ethoxymethylenemalonate at 130°C of a period in the range 20 minutes to 1 h.
  • the resulting diethyl ( ⁇ [3-(3,5-dimethyl-4-isoxazolyl)phenyl] amino ⁇ methylidene)-propanedioate may be cyclised in diphenylether by refluxing for approximately 30 minutes to 1 h (according to the standard procedure described in J.Med.Chem., 1980, 23, 1358).
  • the same procedure may be applied to aminopyridine compounds to prepare the 1 ,5- or 1 ,6- naphthyridine compounds.
  • Suitable amine protecting groups include acyl (e.g. acetyl, carbamate (e.g. 2',2',2'-trichloroethoxycarbonyl, benzyloxycarbonyl or t-butoxycarbonyl) and arylalkyl (e.g. benzyl), which may be removed by hydrolysis (e.g.
  • an acid such as hydrochloric acid in dioxane or trifluoroacetic acid in dichloromethane
  • reductively e.g. hydrogenolysis of a benzyl or benzyloxycarbonyl group or reductive removal of a 2',2',2'-trichloroethoxycarbonyl group using zinc in acetic acid
  • Other suitable amine protecting groups include trifluoroacetyl (-COCF 3 ) which may be removed by base catalysed hydrolysis.
  • the present invention thus provides a compound of formula (I) or a pharmaceutically acceptable salt thereof for use in therapy.
  • a compound of formula (I) or a pharmaceutically acceptable salt thereof for use in therapy.
  • the compound of formula (I) or a pharmaceutically salt thereof can be used in the treatment of diseases or conditions for which a bromodomain inhibitor is indicated.
  • the present invention thus provides a compound of formula (I) or a pharmaceutically acceptable salt thereof for use in the treatment of any diseases or conditions for which a bromodomain inhibitor is indicated.
  • a compound or a pharmaceutically acceptable salt thereof for use in the treatment of chronic auto- immune and/or inflammatory conditions.
  • a compound or a pharmaceutically acceptable salt thereof for use in the treatment of cancer there is provided.
  • a compound of formula (I) or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for the treatment of diseases or conditions for which a bromodomain inhibitor is indicated.
  • a method of treating diseases or conditions for which a bromodomain inhibitor is indicated in a subject in need thereof which comprises administering a therapeutically effective amount of compound of formula (I) or a pharmaceutically acceptable salt thereof.
  • a method of treating diseases or conditions for which a bromodomain inhibitor is indicated in a subject in need thereof which comprises administering a therapeutically effective amount of 7-(3,5- dimethyl-4-isoxazolyl)-8-(methyloxy)-1 -[(1 R)-1 -(2-pyridinyl)ethyl]-1 ,3-dihydro-2H- imidazo[4,5-c]quinolin-2-one or a pharmaceutically acceptable salt thereof.
  • the subject in need thereof is a mammal, particularly a human.
  • the term "effective amount” means that amount of a drug or pharmaceutical agent that will elicit the biological or medical response of a tissue, system, animal or human that is being sought, for instance, by a researcher or clinician.
  • therapeutically effective amount means any amount which, as compared to a corresponding subject who has not received such amount, results in improved treatment, healing, prevention, or amelioration of a disease, disorder, or side effect, or a decrease in the rate of advancement of a disease or disorder.
  • the term also includes within its scope amounts effective to enhance normal physiological function.
  • Bromodomain inhibitors are believed to be useful in the treatment of a variety of diseases or conditions related to systemic or tissue inflammation, inflammatory responses to infection or hypoxia, cellular activation and proliferation, lipid metabolism, fibrosis and in the prevention and treatment of viral infections.
  • Bromodomain inhibitors may be useful in the treatment of a wide variety of chronic autoimmune and inflammatory conditions such as rheumatoid arthritis, osteoarthritis, acute gout, psoriasis, systemic lupus erythematosus, multiple sclerosis, inflammatory bowel disease (Crohn's disease and Ulcerative colitis), asthma, chronic obstructive airways disease, pneumonitis, myocarditis, pericarditis, myositis, eczema, dermatitis, alopecia, vitiligo, bullous skin diseases, nephritis, vasculitis, atherosclerosis, Alzheimer's disease, depression, retinitis, uveitis, scleritis, hepatitis, pancreatitis, primary biliary cirrhosis, sclerosing cholangitis, Addison's disease, hypophysitis, thyroiditis, type I diabetes and
  • Bromodomain inhibitors may be useful in the treatment of a wide variety of acute inflammatory conditions such as acute gout, giant cell arteritis, nephritis including lupus nephritis, vasculitis with organ involvement such as glomerulonephritis, vasculitis including giant cell arteritis, Wegener's granulomatosis, Polyarteritis nodosa, Behcet's disease, Kawasaki disease, Takayasu's Arteritis, vasculitis with organ involvement and acute rejection of transplanted organs.
  • Bromodomain inhibitors may be useful in the prevention or treatment of diseases or conditions which involve inflammatory responses to infections with bacteria, viruses, fungi, parasites or their toxins, such as sepsis, sepsis syndrome, septic shock, endotoxaemia, systemic inflammatory response syndrome (SIRS), multi-organ dysfunction syndrome, toxic shock syndrome, acute lung injury, ARDS (adult respiratory distress syndrome), acute renal failure, fulminant hepatitis, burns, acute pancreatitis, post-surgical syndromes, sarcoidosis, Herxheimer reactions, encephalitis, myelitis, meningitis, malaria and SIRS associated with viral infections such as influenza, herpes zoster, herpes simplex and coronavirus.
  • diseases or conditions which involve inflammatory responses to infections with bacteria, viruses, fungi, parasites or their toxins, such as sepsis, sepsis syndrome, septic shock, endotoxaemia, systemic inflammatory response syndrome (SIRS),
  • Bromodomain inhibitors may be useful in the prevention or treatment of conditions associated with ischaemia-reperfusion injury such as myocardial infarction, cerebro- vascular ischaemia (stroke), acute coronary syndromes, renal reperfusion injury, organ transplantation, coronary artery bypass grafting, cardio-pulmonary bypass procedures, pulmonary, renal, hepatic, gastro-intestinal or peripheral limb embolism.
  • ischaemia-reperfusion injury such as myocardial infarction, cerebro- vascular ischaemia (stroke), acute coronary syndromes, renal reperfusion injury, organ transplantation, coronary artery bypass grafting, cardio-pulmonary bypass procedures, pulmonary, renal, hepatic, gastro-intestinal or peripheral limb embolism.
  • Bromodomain inhibitors may be useful in the treatment of disorders of lipid metabolism via the regulation of APO-A1 such as hypercholesterolemia, atherosclerosis and Alzheimer's disease.
  • Bromodomain inhibitors may be useful in the treatment of fibrotic conditions such as idiopathic pulmonary fibrosis, renal fibrosis, post-operative stricture, keloid formation, scleroderma and cardiac fibrosis.
  • Bromodomain inhibitors may be useful in the prevention and treatment of viral infections such as herpes virus, human papilloma virus, adenovirus and poxvirus and other DNA viruses. Bromodomain inhibitors may be useful in the treatment of cancer, including hematological, epithelial including lung, breast and colon carcinomas, midline carcinomas, mesenchymal, hepatic, renal and neurological tumours.
  • the disease or condition for which a bromodomain inhibitor is indicated is selected from diseases associated with systemic inflammatory response syndrome, such as sepsis, burns, pancreatitis, major trauma, haemorrhage and ischaemia.
  • the bromodomain inhibitor would be administered at the point of diagnosis to reduce the incidence of: SIRS, the onset of shock, multi-organ dysfunction syndrome, which includes the onset of acute lung injury, ARDS, acute renal, hepatic, cardiac and gastro-intestinal injury and mortality.
  • the bromodomain inhibitor would be administered prior to surgical or other procedures associated with a high risk of sepsis, haemorrhage, extensive tissue damage, SIRS or MODS (multiple organ dysfunction syndrome).
  • the disease or condition for which a bromodomain inhibitor is indicated is sepsis, sepsis syndrome, septic shock and endotoxaemia.
  • the bromodomain inhibitor is indicated for the treatment of acute or chronic pancreatitis.
  • the bromodomain is indicated for the treatment of burns.
  • the disease or condition for which a bromodomain inhibitor is indicated is selected from herpes simplex infections and reactivations, cold sores, herpes zoster infections and reactivations, chickenpox, shingles, human papilloma virus, cervical neoplasia, adenovirus infections, including acute respiratory disease, poxvirus infections such as cowpox and smallpox and African swine fever virus.
  • a bromodomain inhibitor is indicated for the treatment of Human papilloma virus infections of skin or cervical epithelia.
  • a compound of formula (I) as well as pharmaceutically acceptable salts thereof may be administered as the raw chemical, it is common to present the active ingredient as a pharmaceutical composition.
  • the present invention therefore provides in a further aspect a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of formula (I) or a pharmaceutically acceptable salt and one or more pharmaceutically acceptable carriers, diluents and/or excipients.
  • the compounds of formula (I) and pharmaceutically acceptable salts are as described above.
  • the carrier(s), diluent(s) or excipient(s) must be acceptable in the sense of being compatible with the other ingredients of the composition and not deleterious to the recipient thereof.
  • a process for the preparation of a pharmaceutical composition including admixing a compound of formula (I), or a pharmaceutically acceptable salt thereof, with one or more pharmaceutically acceptable carriers, diluents or excipients.
  • the pharmaceutical composition can be used in the treatment of any of the conditions described herein.
  • a pharmaceutical composition comprising 7-(3,5- dimethyl-4-isoxazolyl)-8-(methyloxy)-1 -[(1 R)-1 -(2-pyridinyl)ethyl]-1 ,3-dihydro-2H- imidazo[4,5-c]quinolin-2-one or a pharmaceutically acceptable salt and one or more or pharmaceutically acceptable carriers, diluents or excipients.
  • the compounds of formula (I) are intended for use in pharmaceutical compositions it will be readily understood that they are each preferably provided in substantially pure form, for example, at least 60% pure, more suitably at least 75% pure and preferably at least 85% pure, especially at least 98% pure (% in a weight for weight basis).
  • compositions may be presented in unit dose forms containing a predetermined amount of active ingredient per unit dose.
  • Preferred unit dosage compositions are those containing a daily dose or sub-dose, or an appropriate fraction thereof, of an active ingredient. Such unit doses may therefore be administered more than once a day.
  • Preferred unit dosage compositions are those containing a daily dose or sub- dose (for administration more than once a day), as herein above recited, or an appropriate fraction thereof, of an active ingredient.
  • compositions may be adapted for administration by any appropriate route, for example by the oral (including buccal or sublingual), rectal, inhaled, intranasal, topical (including buccal, sublingual or transdermal), vaginal or parenteral (including subcutaneous, intramuscular, intravenous or intradermal) route.
  • Such compositions may be prepared by any method known in the art of pharmacy, for example by bringing into association the active ingredient with the carrier(s) or excipient(s).
  • the pharmaceutical composition is adapted for parenteral administration, particularly intravenous administration.
  • the pharmaceutical composition is adapted for oral administration.
  • compositions adapted for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the composition isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents.
  • the compositions may be presented 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 injections, immediately prior to use.
  • Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets.
  • compositions adapted for oral administration may be presented as discrete units such as capsules or tablets; powders or granules; solutions or suspensions in aqueous or non-aqueous liquids; edible foams or whips; or oil-in-water liquid emulsions or water-in-oil liquid emulsions.
  • the active drug component can be combined with an oral, non-toxic pharmaceutically acceptable inert carrier such as ethanol, glycerol, water and the like.
  • an oral, non-toxic pharmaceutically acceptable inert carrier such as ethanol, glycerol, water and the like.
  • Powders suitable for incorporating into tablets or capsules may be prepared by reducing the compound to a suitable fine size (e.g. by micronisation) and mixing with a similarly prepared pharmaceutical carrier such as an edible carbohydrate, for example, starch or mannitol. Flavoring, preservative, dispersing and coloring agent can also be present.
  • Capsules may be made by preparing a powder mixture, as described above, and filling formed gelatin sheaths.
  • Glidants and lubricants such as colloidal silica, talc, magnesium stearate, calcium stearate or solid polyethylene glycol can be added to the powder mixture before the filling operation.
  • a disintegrating or solubilizing agent such as agar- agar, calcium carbonate or sodium carbonate can also be added to improve the availability of the medicament when the capsule is ingested.
  • suitable binders, glidants, lubricants, sweetening agents, flavours, disintegrating agents and coloring agents can also be incorporated into the mixture.
  • Suitable binders include starch, gelatin, natural sugars such as glucose or beta-lactose, corn sweeteners, natural and synthetic gums such as acacia, tragacanth or sodium alginate, carboxymethylcellulose, polyethylene glycol, waxes and the like.
  • Lubricants used in these dosage forms include sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride and the like.
  • Disintegrators include, without limitation, starch, methyl cellulose, agar, bentonite, xanthan gum and the like. Tablets are formulated, for example, by preparing a powder mixture, granulating or slugging, adding a lubricant and disintegrant and pressing into tablets.
  • a powder mixture is prepared by mixing the compound, suitably comminuted, with a diluent or base as described above, and optionally, with a binder such as carboxymethylcellulose, an aliginate, gelatin, or polyvinyl pyrrolidone, a solution retardant such as paraffin, a resorption accelerator such as a quaternary salt and/or an absorption agent such as bentonite, kaolin or dicalcium phosphate.
  • the powder mixture can be granulated by wetting with a binder such as syrup, starch paste, acadia mucilage or solutions of cellulosic or polymeric materials and forcing through a screen.
  • the powder mixture can be run through the tablet machine and the result is imperfectly formed slugs broken into granules.
  • the granules can be lubricated to prevent sticking to the tablet forming dies by means of the addition of stearic acid, a stearate salt, talc or mineral oil.
  • the lubricated mixture is then compressed into tablets.
  • the compounds of formula (I) and pharmaceutically acceptable salts thereof can also be combined with a free flowing inert carrier and compressed into tablets directly without going through the granulating or slugging steps.
  • a clear or opaque protective coating consisting of a sealing coat of shellac, a coating of sugar or polymeric material and a polish coating of wax can be provided. Dyestuffs can be added to these coatings to distinguish different unit dosages.
  • Oral fluids such as solution, syrups and elixirs can be prepared in dosage unit form so that a given quantity contains a predetermined amount of the compound.
  • Syrups can be prepared by dissolving the compound in a suitably flavored aqueous solution, while elixirs are prepared through the use of a non-toxic alcoholic vehicle.
  • Suspensions can be formulated by dispersing the compound in a non-toxic vehicle.
  • Solubilizers and emulsifiers such as ethoxylated isostearyl alcohols and polyoxy ethylene sorbitol ethers, preservatives, flavor additive such as peppermint oil or natural sweeteners or saccharin or other artificial sweeteners, and the like can also be added.
  • dosage unit compositions for oral administration can be microencapsulated.
  • the formulation can also be prepared to prolong or sustain the release as for example by coating or embedding particulate material in polymers, wax or the like.
  • the compounds of formula (I) and pharmaceutically acceptable salts thereof can also be administered in the form of liposome delivery systems, such as small unilamellar vesicles, large unilamellar vesicles and multilamellar vesicles.
  • liposomes can be formed from a variety of phospholipids, such as cholesterol, stearylamine or phosphatidylcholines.
  • compositions adapted for topical administration may be formulated as ointments, creams, suspensions, lotions, powders, solutions, pastes, gels, sprays, aerosols or oils.
  • compositions are preferably applied as a topical ointment or cream.
  • the active ingredient may be employed with either a paraffinic or a water- miscible ointment base.
  • the active ingredient may be formulated in a cream with an oil-in-water cream base or a water-in-oil base.
  • compositions adapted for topical administrations to the eye include eye drops wherein the active ingredient is dissolved or suspended in a suitable carrier, especially an aqueous solvent.
  • Dosage forms for nasal or inhaled administration may conveniently be formulated as aerosols, solutions, suspensions, gels or dry powders.
  • compositions suitable and/or adapted for inhaled administration it is preferred that the compound of formula (I) or a pharmaceutically acceptable salt thereof, is in a particle-size- reduced form e.g. obtained by micronisation.
  • the preferable particle size of the size- reduced (e.g. micronised) compound or salt is defined by a D50 value of about 0.5 to about 10 microns (for example as measured using laser diffraction).
  • Aerosol formulations, e.g. for inhaled administration can comprise a solution or fine suspension of the active substance in a pharmaceutically acceptable aqueous or non- aqueous solvent.
  • Aerosol formulations can be presented in single or multidose quantities in sterile form in a sealed container, which can take the form of a cartridge or refill for use with an atomising device or inhaler.
  • the sealed container may be a unitary dispensing device such as a single dose nasal inhaler or an aerosol dispenser fitted with a metering valve (metered dose inhaler) which is intended for disposal once the contents of the container have been exhausted.
  • the dosage form comprises an aerosol dispenser
  • it preferably contains a suitable propellant under pressure such as compressed air, carbon dioxide or an organic propellant such as a hydrofluorocarbon (HFC).
  • suitable HFC propellants include 1 ,1 ,1 ,2,3,3,3-heptafluoropropane and 1 ,1 ,1 ,2-tetrafluoroethane.
  • the aerosol dosage forms can also take the form of a pump-atomiser.
  • the pressurised aerosol may contain a solution or a suspension of the active compound. This may require the incorporation of additional excipients e.g. co-solvents and/or surfactants to improve the dispersion characteristics and homogeneity of suspension formulations. Solution formulations may also require the addition of co-solvents such as ethanol.
  • the pharmaceutical composition may be a dry powder inhalable composition.
  • a dry powder inhalable composition can comprise a powder base such as lactose, glucose, trehalose, mannitol or starch, the compound of formula (I) or a pharmaceutically acceptable salt thereof (preferably in particle-size-reduced form, e.g. in micronised form), and optionally a performance modifier such as L-leucine or another amino acid and/or metals salts of stearic acid such as magnesium or calcium stearate.
  • the dry powder inhalable composition comprises a dry powder blend of lactose e.g. lactose monohydrate and the compound of formula (I) or salt thereof.
  • compositions can be administered to the patient using a suitable device such as the DISKUS® device, marketed by GlaxoSmithKline which is for example described in GB 2242134 A.
  • a suitable device such as the DISKUS® device, marketed by GlaxoSmithKline which is for example described in GB 2242134 A.
  • the compounds of formula (I) and pharmaceutically acceptable salts thereof may be formulated as a fluid formulation for delivery from a fluid dispenser, for example a fluid dispenser having a dispensing nozzle or dispensing orifice through which a metered dose of the fluid formulation is dispensed upon the application of a user-applied force to a pump mechanism of the fluid dispenser.
  • Such fluid dispensers are generally provided with a reservoir of multiple metered doses of the fluid formulation, the doses being dispensable upon sequential pump actuations.
  • the dispensing nozzle or orifice may be configured for insertion into the nostrils of the user for spray dispensing of the fluid formulation into the nasal cavity.
  • a fluid dispenser of the aforementioned type is described and illustrated in WO-A-2005/044354.
  • a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof will depend upon a number of factors including, for example, the age and weight of the animal, the precise condition requiring treatment and its severity, the nature of the formulation, and the route of administration, and will ultimately be at the discretion of the attendant physician or veterinarian.
  • each dosage unit for oral or parenteral administration preferably contains from 0.01 to 3000 mg, more preferably 0.5 to 1000 mg, of a compound of formula (I) or a pharmaceutically acceptable salt thereof, calculated as the free base.
  • Each dosage unit for nasal or inhaled administration preferably contains from 0.001 to 50 mg, more preferably 0.01 to 5 mg, of a compound of the formula (I) or a pharmaceutically acceptable salt thereof, calculated as the free base.
  • the pharmaceutically acceptable compounds of formula (I) and pharmaceutically acceptable salts thereof can be administered in a daily dose (for an adult patient) of, for example, an oral or parenteral dose of 0.01 mg to 3000 mg per day or 0.5 to 1000 mg per day, or a nasal or inhaled dose of 0.001 to 50 mg per day or 0.01 to 5 mg per day, of the compound of the formula (I) or a pharmaceutically acceptable salt thereof, calculated as the free base.
  • This amount may be given in a single dose per day or more usually in a number (such as two, three, four, five or six) of sub-doses per day such that the total daily dose is the same.
  • An effective amount of a salt thereof may be determined as a proportion of the effective amount of the compound of formula (I) per se.
  • Combination therapies according to the present invention thus comprise the administration of at least one compound of formula (I) or a pharmaceutically acceptable salt thereof, and the use of at least one other pharmaceutically active agent.
  • combination therapies according to the present invention comprise the administration of at least one compound of formula (I) or a pharmaceutically acceptable salt thereof, and at least one other pharmaceutically active agent.
  • the compound(s) of formula (I) and pharmaceutically acceptable salts thereof, and the other pharmaceutically active agent(s) may be administered together in a single pharmaceutical composition or separately and, when administered separately this may occur simultaneously or sequentially in any order.
  • a combination comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof, and at least one other pharmaceutically active agent.
  • the compound of formula (I) or a pharmaceutically acceptable salt thereof, and pharmaceutical compositions comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof, according to the invention may be used in combination with or include one or more other therapeutic agents, for example selected from antibiotics, anti-virals, glucocorticosteroids, muscarinic antagonists and beta-2 agonists.
  • the compound of formula (I) or a pharmaceutically acceptable salt thereof when administered in combination with other therapeutic agents normally administered by the inhaled, intravenous, oral or intranasal route, that the resultant pharmaceutical composition may be administered by the same routes. Alternatively the individual components of the composition may be administered by different routes.
  • One embodiment of the invention encompasses combinations comprising one or two other therapeutic agents.
  • the other therapeutic ingredient(s) may be used in the form of salts, for example as alkali metal or amine salts or as acid addition salts, or prodrugs, or as esters, for example lower alkyl esters, or as solvates, for example hydrates, to optimise the activity and/or stability and/or physical characteristics, such as solubility, of the therapeutic ingredient. It will be clear also that, where appropriate, the therapeutic ingredients may be used in optically pure form.
  • compositions comprising a combination as defined above together with a pharmaceutically acceptable diluent or carrier represent a further aspect of the invention.
  • the compounds of formula (I) and pharmaceutically acceptable salts thereof may be prepared by the methods described below or by similar methods.
  • the following Intermediates and Examples serve to illustrate the preparation of the compounds of formula (I) and pharmaceutically acceptable salts thereof, and are not to be considered as limiting the scope of the invention in any way.
  • MS mass spectra
  • electrospray positive ionisation (ES+ve to give [M+H] + and [M+NH4] + molecular ions] or electrospray negative ionisation [(ES-ve to give [M-H]- molecular ion] modes.
  • Analytical data from this apparatus are given with the following format : [M+H] + or [M-H] " .
  • LC/HRMS Analytical HPLC was conducted on a Uptisphere-hsc column (3 ⁇ 33 x 3 mm id) eluting with 0.01 M ammonium acetate in water (solvent A) and 100% acetonitrile (solvent B), using the following elution gradient 0-0.5 minutes 5% B, 0.5-3.75 minutes 5 ⁇ 100% B, 3.75-4.5 100% B, 4.5-5 100 ⁇ 5% B, 5-5.5 5% B at a flow rate of 1 .3ml/minute.
  • MS mass spectra
  • Mass directed auto-prep HPLC refers to the method where the material was purified by high performance liquid chromatography on a HPLCABZ+ 5 ⁇ column (5cm x 10mm i.d.) with 0.1 % HCO2H in water and 95% MeCN, 5% water (0.5% HCO2H) utilising the following gradient elution conditions: 0-1 .0 minutes 5%B, 1.0-8.0 minutes 5 ⁇ 30%B, 8.0- 8.9 minutes 30%B, 8.9-9.0 minutes 30 ⁇ 95%B, 9.0-9.9 minutes 95%B, 9.9-10 minutes 95 ⁇ 0%B at a flow rate of 8ml/minute.
  • the Gilson 202-fraction collector was triggered by a VG Platform Mass Spectrometer on detecting the mass of interest.
  • TLC thin layer chromatography
  • the UPLC analysis was conducted on an Acquity UPLC BEH C18 column (50mm x 2.1 mm, i.d. 1 .7 ⁇ packing diameter) at 40°C.
  • the UV detection was a summed signal from wavelength of 210nm to 350nm.
  • the UPLC analysis was conducted on an Acquity UPLC BEH C18 column (50mm x 2.1 mm, i.d. 1 .7 ⁇ packing diameter) at 40°C.
  • A 10mM ammonium hydrogen carbonate in water adjusted to pH10 with ammonia solution
  • the UV detection was a summed signal from wavelength of 210nm to 350n
  • the HPLC analysis was conducted on either a Sunfire C18 column (100mm x 19mm, i.d 5 ⁇ packing diameter) or a Sunfire C18 column (150mm x 30mm, i.d. 5 ⁇ packing diameter) at ambient temperature.
  • the UV detection was a summed signal from wavelength of 210nm to 350nm.
  • Nitric acid 13ml was added dropwise (exothermic reaction) to a mixture of 3,5- dimethylisoxazole (31.3g, 320mmol) and iodine (37.3g, 150 mmol) and the mixture was stirred at room temperature for 1 h.
  • the reaction mixture was hydrolysed with a mixture of ice and water and extracted with DCM.
  • the organic phase was washed with a solution of Na2S 2 0 3 , dried over Na 2 S0 4 and concentrated under reduced pressure to afford the title compound as a yellow solid (60g, 83%).
  • the aqueous layer was acidified to pH 7 with concentrated HCI and extracted with ethyl acetate.
  • the aqueous layer was basified to pH 10 with sodium hydroxide 5N and extracted with ethyl acetate.
  • the organic layers were combined, washed with a saturated aqueous NaCI and dried.
  • the crude brown oil was then purified by flash chromatography on silica gel eluting with DCM/MeOH (9:1 ) to give the title compound as a brown solid (31.4g, 43.9%)
  • Tin (II) chloride dihydrate (0.89 g) was then added in four portions and the reaction mixture was stirred for 1 h at 40°C.
  • the reaction mixture was hydrolysed using a solution of sodium hydroxide (1 N).
  • the mixture was extracted with DCM, the organic dried over Na 2 S0 4 , filtered over Celite and concentrated to dryness.
  • Example 1 1 -[2-(tert-butyl)phenyl]-7-(3,5-dimethyl-4-isoxazolyl)-1 ,3-dihydro-2H- imidazo[4,5-c]quinolin-2-one
  • Example 12 7-(3,5-dimethyl-4-isoxazolyl)-8-(methoxy)-1 - ⁇ 2- [(trifluoromethyl)oxy]phenyl ⁇ -1 ,3-dihydro-2H-imidazo[4,5-c]quinolin-2-one
  • Example 36 7-(3,5-dimethyl-4-isoxazolyl)-8-(methyloxy)-1 -[(1 R)-1 -(2-pyridinyl)ethyl]- 1 ,3-dihydro-2H-imidazo[4,5-c]q inolin-2-one
  • Example 37 7-(3,5-dimethyl-4-isoxazolyl)-8-(methyloxy)-3-[2-(methyloxy)ethyl]-1 - [(1 R)-1 -(2-pyridinyl)ethyl]-1 ,3-dih dro-2H-imidazo[4,5-c]quinolin-2-one
  • the reaction was stirred at ambient temperature, air atm for ⁇ 3h and then left at ambient temperature overnight.
  • the reaction was filtered and the resin washed with methanol.
  • the solvents were evaporated under a stream of nitrogen to leave a yellow gum which was dissolved in DMSO (1 ml) and purified by MDAP (Method formate).
  • Example 38 7-(3,5-dimethyl-4-isoxazolyl)-8-(methyloxy)-1 -[1 -(2-pyridinyl)ethyl]-1 ,3- dihydro-2H-imidazo[4,5-c]quinol -2-one
  • Example 39 7-(3,5-dimethyl-4-isoxazolyl)-8-(methyloxy)-1 -[(1 S)-1 -(2-pyridinyl)ethyl]- 1 ,3-dihydro-2H-imidazo[4,5-c]quinolin-2-one
  • the solutions were purified by chiral HPLC using 6x injection (3ml) of the above solution onto a Chiralpak IA column (20um, 5cm x 25cm) and elution with ethanol / heptanes (30%, flow rate 50ml/min, wavelength 215nm).
  • the mixed fractions were reduced to dryness in vacuo and reprocessed as described in the method above.
  • Example 40 7-(3,5-dimethyl-4-isoxazolyl)-8-(methyloxy)-1 -[1 -(1 -methyl-1 H-pyrazol- 4-yl)ethyl]-1 ,3-dihydro-2H-imidazo 4,5-c]quinolin-2-one (
  • reaction mixture was then taken-up in a 1 :1 mixture of sodium hydroxide (1 N) and water (total 100ml), extracted with DCM and the resulting crude product was purified by chromatography on silica gel eluting with DCM / methanol (95:5).
  • the resulting product was taken-up in diethyl ether and dried under vacuum to give racemic 7-(3,5-dimethyl-4-isoxazolyl)-8-(methyloxy)-1 -[1 -(2-pyridinyl)ethyl]-2-(tetrahydro- 2H-pyran-4-yl)-1 H-imidazo[4,5-c]quinoline (400 mg) as a grey powder.
  • Example 48 7-(3,5-dimethyl-4-isoxazolyl)-8-(methyloxy)-1 -(2-pyrazinylmethyl)-1 ,3- dihydro-2H-imidazo[4,5-c]quinolin-2-one
  • reaction mixture was then concentrated to dryness and hydrolysed with water.
  • the mixture was extracted with DCM and the organic dried over Na 2 S0 4 , filtered and concentrated to dryness.
  • the residue was purified by chromatography on silica gel, eluting with DCM / methanol (95 / 5). The purified residue was taken-up in diethyl ether to give 7-(3,5-dimethyl-4-isoxazolyl)-8-(methyloxy)-1 -(3-pyridinylmethyl)-1 ,3-dihydro-2/-/- imidazo[4,5-c]quinolin-2-one (70mg) as a brown powder.
  • Example 50 7-(3,5-dimethyl-4-isoxazolyl)-1 -[(2,4-dimethyl-1 ,3-thiazol-5-yl)methyl]-8- (methyloxy)-l ,3-dihydro-2H-imidazo[4,5-c]quinolin-2-one
  • the resulting product was dissolved in ethanol (60ml) and a solution of sodium hydroxide (1 N, 15ml) was added. The reaction mixture was heated to 90°C for 16h and was then allowed to cool to room temperature. DCM (150ml) and water (100ml) were added. The phases were separated and the organic phase was dried over Na 2 S0 4 , filtered and evaporated to dryness. The residue was purified by silica gel chromatography eluting with DCM / methanol (95:5).
  • Example 54 7-(3,5-dimethyl-4-isoxazolyl)-1 -[(2,4-dimethyl-1 ,3-thiazol-5-yl)methyl]-8- (methyloxy)-2-(tetrahydro-2H-pyran-4-yl)-1 H-imidazo[4,5-c]quinoline
  • LC/HRMS Analytical HPLC was conducted on a Uptisphere-hsc column (3 ⁇ 33 x 3 mm id) eluting with 0.01 M ammonium acetate in water (solvent A) and 100% acetonitrile (solvent B), using the following elution gradient 0-0.5 minutes 5% B, 0.5-3.75 minutes 5 ⁇ 100% B, 3.75-4.5 100% B, 4.5-5 100 ⁇ 5% B, 5-5.5 5% B at a flow rate of 1.3 mL/minute.
  • MS mass spectra
  • TLC thin layer chromatography refers to the use of TLC plates sold by Merck coated with silica gel 60 F254.
  • Reference compound A 2-methyl-6-(methyloxy)-4H-3,1 -benzoxazin-4-one
  • Reference compound K Mixture of 5- and 6- isomers of Alexa Fluor 488-N-(5- aminopentyl)-2-[(4S)-6-(4-chlorophenyl)-1 -methyl -8 -(methyl oxy)-4H- [1 ,2,4]triazolo[4,3-a][1 ,4]benzodiazepin-4-yl]acetamide
  • N-(5-aminopentyl)-2-[(4S)-6-(4-chlorophenyl)-1 -methyl-8-(methyloxy)-4H- [1 ,2,4]triazolo[4,3-a][1 ,4]benzodiazepin-4-yl]acetamide trifluoroacetate (for a preparation see Reference compound J) (7.65 mg, 0.013 mmol) was dissolved in N,N- dimethylformamide (DMF) (300 ⁇ ) and added to Alexa Fluor 488 carboxylic acid succinimidyl ester (5 mg, 7.77 ⁇ , mixture of 5 and 6 isomers, available from Invitrogen, product number A-20100) in an Eppendorf centrifuge tube.
  • DMF N,N- dimethylformamide
  • the Bromodomain protein, fluorescent ligand (Reference compound K see above) and a variable concentration of test compound are incubated together to reach thermodynamic equilibrium under conditions such that in the absence of test compound the fluorescent ligand is significantly (>50%) bound and in the presence of a sufficient concentration of a potent inhibitor the anisotropy of the unbound fluorescent ligand is measurably different from the bound value.
  • 'a' is the minimum
  • 'b' is the Hill slope
  • 'c' is the plC 50
  • 'd' is the maximum.
  • Bromodomain BRD2 (1 -473), Bromodomain BRD3 (1 -435) and Bromodomain BRD4 (1 -477)
  • E.coli cells in pET15b vector
  • the His-tagged Bromodomain was extracted from E.coli cells using 0.1 mg/ml lysozyme and sonication.
  • the Bromodomain was then purified by affinity chromatography on a HisTRAP HP column, eluting with a linear 10- 500mM Imidazole gradient, over 20 Cv. Further purification was completed by Superdex 200 prep grade size exclusion column.
  • Protocol for Bromodomain BRD2 All components were dissolved in buffer composition of 50 mM HEPES pH7.4, 150mm NaCI and 0.5mM CHAPS with final concentrations of Bromodomain 2, 75nM, fluorescent ligand 5nM.10 ⁇ of this reaction mixture was added using a micro multidrop to wells containing 10Onl of various concentrations of test compound or DMSO vehicle (1 % final) in Greiner 384 well Black low volume microtitre plate and equilibrated in dark 60 mins at room temperature.
  • Activation of monocytic cells by agonists of toll-like receptors such as bacterial lipopolysaccharide (LPS) results in production of key inflammatory mediators including TNFa.
  • LPS bacterial lipopolysaccharide
  • TNFa levels assayed by immunoassay (typically by MesoScale Discovery technology) either immediately or following storage at -20 degrees.
  • Dose response curves for each compound was generated from the data and an IC 50 value was calculated.
  • Examples 1 1 , 12, 15, 27, 31 , 33, 36, 108 and 126 were tested in the above assay were found to have a plC 5 o ⁇ 5.5.
  • Activation of monocytic cells by agonists of toll-like receptors such as bacterial lipopolysaccharide (LPS) results in production of key inflammatory mediators including IL- 6.
  • LPS bacterial lipopolysaccharide
  • Endotoxin bacterial lipopolysaccharide
  • Endotoxin bacterial lipopolysaccharide
  • This pattern of response is very similar to human sepsis and septic shock, where the body's response to a significant bacterial infection can be similarly life threatening.
  • mice were given a lethal dose of 15 mg/kg LPS by intraperitoneal injection.
  • animals were dosed intravenously with vehicle (20% cyclodextrin 1 % ethanol in apyrogen water) or compound (10 mg/kg). The survival of animals was monitored at 4 days.
  • Cells were incubated for 72 hours or the indicated time and each plate was developed with CellTiter-Glo reagent using a volume equivalent to the cell culture volume in the wells. Plates were shaken for approximately 2 minutes and chemiluminescent signal was read on the Analyst GT (Molecular Devices) or EnVision Plate Reader (Perkin Elmer).
  • Results were expressed as a percent of the TO and plotted against the compound concentration.
  • the TO value was normalized to 100% and represents the number of cells at time of compound addition and the concentration response data were fit with a 4 parameter curve fit using XLfit software (model 205).
  • the concentration that inhibited cell growth by 50% (glC 50 ) is the midpoint of the 'growth window' (between the TO and DMSO control).
  • the Ymin - TO value is determined by subtracting the TO value (100%) from the Ymin value (%) determined from the fit of the concentration response curve. Values from the wells with no cells were subtracted from all samples for background correction.
  • the compound of Examples 1 1 and 36 was tested in accordance with the above assay and found to have a glC 50 in the range 1 18-1 1 100 nM across all cell lines, more specifically in the range 1 18 - 1036 nM for heme cell lines and 432 - 1 1 100 nM for breast cell lines.

Abstract

Novel compounds of Formula (I) and salts thereof, pharmaceutical compositions containing such compounds and their use in therapy.

Description

IMIDAZO [4,5-C] QUINOLINE DERIVATIVES AS BROMODOMAIN INHIBITORS
Field of the Invention
The present invention relates to novel compounds, pharmaceutical compositions containing such compounds and to their use in therapy.
Background of the Invention
The genomes of eukaryotic organisms are highly organised within the nucleus of the cell. The long strands of duplex DNA are wrapped around an octomer of histone proteins (most usually comprising two copies of histones H2A, H2B H3 and H4) to form a nucleosome. This basic unit is then further compressed by the aggregation and folding of nucleosomes to form a highly condensed chromatin structure. A range of different states of condensation are possible, and the tightness of this structure varies during the cell cycle, being most compact during the process of cell division. Chromatin structure plays a critical role in regulating gene transcription, which cannot occur efficiently from highly condensed chromatin. The chromatin structure is controlled by a series of post translational modifications to histone proteins, notably histones H3 and H4, and most commonly within the histone tails which extend beyond the core nucleosome structure. These modifications include acetylation, methylation, phosphorylation, ubiquitinylation, SUMOylation. These epigenetic marks are written and erased by specific enzymes, which place the tags on specific residues within the histone tail, thereby forming an epigenetic code, which is then interpreted by the cell to allow gene specific regulation of chromatin structure and thereby transcription. Histone acetylation is most usually associated with the activation of gene transcription, as the modification loosens the interaction of the DNA and the histone octomer by changing the electrostatics. In addition to this physical change, specific proteins bind to acetylated lysine residues within histones to read the epigenetic code. Bromodomains are small (-1 10 amino acid) distinct domains within proteins that bind to acetylated lysine resides commonly but not exclusively in the context of histones. There is a family of around 50 proteins known to contain bromodomains, and they have a range of functions within the cell.
The BET family of bromodomain containing proteins comprises 4 proteins (BRD2, BRD3, BRD4 and BRD-t) which contain tandem bromodomains capable of binding to two acetylated lysine residues in close proximity, increasing the specificity of the interaction. BRD2 and BRD3 are reported to associate with histones along actively transcribed genes and may be involved in facilitating transcriptional elongation (Leroy et al, Mol. Cell. 2008 30(1 ):51 -60), while BRD4 appears to be involved in the recruitment of the pTEF-β complex to inducible genes, resulting in phosphorylation of RNA polymerase and increased transcriptional output (Hargreaves et al, Cell, 2009 138(1 ): 129-145). It has also been reported that BRD4 or BRD3 may fuse with NUT (nuclear protein in testis) forming novel fusion oncogenes, BRD4-NUT or BRD3-NUT, in a highly malignant form of epithelial neoplasia (French et al. Cancer Research, 2003, 63, 304-307 and French et al. Journal of Clinical Oncology, 2004, 22 (20), 4135-4139). Data suggests that BRD-NUT fusion proteins contribute to carcinogenesis (Oncogene, 2008, 27, 2237-2242). BRD-t is uniquely expressed in the testes and ovary. All family members have been reported to have some function in controlling or executing aspects of the cell cycle, and have been shown to remain in complex with chromosomes during cell division - suggesting a role in the maintenance of epigenetic memory. In addition some viruses make use of these proteins to tether their genomes to the host cell chromatin, as part of the process of viral replication (You et al Cell, 2004 1 17(3):349-60).
Japanese patent application JP2008-15631 1 discloses a benzimidazole derivative which is said to be a BRD2 bromodomain binding agent which has utility with respect to virus infection / proliferation.
Patent application WO2009084693A1 discloses a series of thienotriazolodiazepiene derivatives that are said to inhibit the binding between an acetylated histone and a bromodomain containing protein which are said to be useful as anti-cancer agents.
A novel class of compounds have been found which inhibit the binding of bromodomains with its cognate acetylated proteins, more particularly a class of compounds that inhibit the binding of BET family bromodomains to acetylated lysine residues. Such compounds will hereafter be referred to as "bromodomain inhibitors".
Summary of the Invention
In a first aspect of the present invention, there is provided a compound of formula (I) or a salt thereof, more particularly a compound of formula (I) or a pharmaceutically acceptable salt thereof
Figure imgf000005_0001
In a second aspect of the present invention, there is provided a pharmaceutical composition comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof and one or more pharmaceutically acceptable carriers, diluents or excipients.
In a third aspect of the present invention, there is provided a compound of formula (I), or a pharmaceutically acceptable salt thereof for use in therapy, in particular in the treatment of diseases or conditions for which a bromodomain inhibitor is indicated. In a fourth aspect of the present invention, there is provided a method of treating diseases or conditions for which a bromodomain inhibitor is indicated in a subject in need thereof which comprises administering a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof. In a fifth aspect of the present invention, there is provided the use of a compound of formula (I), or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for the treatment of diseases or conditions for which a bromodomain inhibitor is indicated. Detailed Description of the Invention
The present invention relates to compounds of formula (I) or a salt thereof
Figure imgf000005_0002
wherein:
• A is a group selected from the following:
Figure imgf000006_0001
• X represents CH or N;
· Y represents CH or N with the proviso that when X is N, Y is CH;
• Rx represents O or S;
• R1 represents C-| .galkyl, C3_gcycloalkyl, a 5- or 6-membered heterocyclyl, an aromatic group or a heteroaromatic group, wherein the aromatic group or the heteroaromatic group is optionally substituted by one to three groups selected from:
halogen, hydroxy, cyano, nitro, C-| .galkyl, C-|_4alkoxy, haloC-| _4alkyl, haloC-| _4alkoxy, hydroxy Chalky I, C-| _4alkoxyC-| _4alkyl, C-μ 4alkoxycarbonyl, C-| _4alkylsulfonyl, C-| _4alkylsulfonyloxy, C-μ 4alkylsulfonylC-| _4alkyl and C-| .4alkylsulfonamido;
* R2 is hydrogen or C-| .galkyl,
• R2a represents:
o H, C-| .galkyl, C-| .ghaloalkyl, (CH2)mcyano, (CH2)mOH, (CH2)mC-| . galkoxy, (CH2)mC-| .ghaloalkoxy, (CH2)mC-| .ghaloalkyl
(CH2)mC(0)NRaRb, (CH2)mNRaRb, (CH2)mC(0)CH3,
o (CHR6)pphenyl optionally substituted by C-| .galkyl, C-| .galkoxy, cyano haloC-| _4alkoxy, haloC-| .4alkyl,
o (CHR6)pheteroaromatic or (CHR6)pheterocyclyl,
wherein
Ra represents H, C-| .galkyl, or heterocyclyl;
Rb represents H or C-| .galkyl, or
Ra and Rb together with the N to which they are attached form a 5- or 6-membered heterocyclyl; • R represents H, C-| .galkyl, (CH2)2C-| .galkoxy, (CH2)2cyano or (CH2)mphenyl, (CH2)2heterocyclyl;
• R3 represents hydrogen;
• R4 represents hydrogen, cyano or C-| .galkyl;
· Z represents O; or when R4 represents hydrogen and A is a group selected from (i) or (ii) and wherein Rx represents O, Z may additionally represent NH;
• R5 represents hydrogen or C-| .galkoxy;
• R6 represents hydrogen or C-| .galkyl;
• m represents 1 , 2 or 3;
· n represents 0, 1 or 2; and
• p represents 0, 1 or 2.
As used herein, the term "aromatic group" refers to a 5- to 7-membered monocyclic aromatic group such as phenyl, or a 8- to 1 1 -membered bicyclic aromatic group such as naphthyl, indenyl or azulenyl.
As used herein heterocyclyl refers to a 5- or 6-membered non-aromatic, saturated or unsaturated ring comprising 1 , 2, or 3 heteroatoms selected from O, N and S. Examples of heterocyclyls include morpholinyl, piperidinyl, tetrahydropyranyl, and piperazinyl.
As used herein, the term "heteroaromatic group" refers to a 5- or 6-membered monocyclic aromatic group wherein 1 , 2, 3, 4 of the carbon atoms are replaced by a heteroatom independently selected from O, S and N; or to a 8- to 1 1 -membered bicyclic aromatic group wherein 1 , 2, 3, 4 or 5 of the carbon atoms are replaced by a heteroatom independently selected from O, S and N.
Examples of 5- or 6-membered monocyclic heteroaromatic groups include pyrrolinyl, furanyl, thienyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, oxadiazolyl, thiadiazolyl, pyridinyl, triazolyl, triazinyl, pyridazyl, pyrimidinyl, isothiazolyl, isoxazolyl, pyrazinyl, pyrazolyl and pyrimidinyl. Examples of 8- to 1 1 - membered bicyclic heteroaromatic groups include 6H- thieno[2,3-b]pyrrolyl, imidazo[2,1 -b][1 ,3]thiazolyl, imidazo[5,1 -b][1 ,3]thiazolyl, indolyl, isoindolyl, indazolyl, benzimidazolyl, [1 ,3]thiazolo[3,2-fc>][1 ,2,4]triazolyl, benzoxazolyl e.g. benzoxazol-2-yl, benzisoxazolyl, benzothiazolyl, benzisothiazolyl, benzothienyl, benzofuranyl, naphthridinyl, quinolyl, quinoxalinyl, quinazolinyl, cinnolinyl and isoquinolyl. As used herein, the term "alkyl" refers to straight or branched hydrocarbon chains containing the specified number of carbon atoms. For example, C-| .galkyl means a straight or branched alkyl containing at least 1 , and at most 6, carbon atoms. Examples of "alkyl" as used herein include, but are not limited to, methyl, ethyl, n-propyl, n-butyl, n- pentyl, n-hexyl, isobutyl, isopropyl, t-butyl and 1 ,1 -dimethylpropyl.
Examples of C3_gcycloalkyls include cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl.
As used herein, the term "alkoxy" refers to a straight or branched alkoxy group containing the specified number of carbon atoms. For example, C-| .galkoxy means a straight or branched alkoxy group containing at least 1 , and at most 6, carbon atoms. Examples of "alkoxy" as used herein include, but are not limited to, methoxy, ethoxy, propoxy, prop-2- oxy, butoxy, but-2-oxy, 2-methylprop-1 -oxy, 2-methylprop-2-oxy, pentoxy or hexyloxy. As used herein, the term "halogen" refers to the elements fluorine, chlorine, bromine and iodine, for example, fluorine, chlorine and bromine.
As used herein, the term "substituted" refers to substitution with the named substituent or substituents, multiple degrees of substitution being allowed unless otherwise stated. When the substituent is on a ring comprising a heteroatom the substituent may be located on a carbon or a heteroatom, if the latter is appropriate.
In one embodiment, X is CH. In one embodiment Y is CH.
In one embodiment Z is O.
In another embodiment Z is NH (and in consequence R4 represents hydrogen, A is a group selected from (i) or (ii) as defined above, and Rx represents O).
In one embodiment Rx is O.
In one embodiment n is 0 or 1 , such as 1 . In one embodiment, A is a group of formula (i) or (ii) as described above.
Representative examples of R1 include, pyridinyl such as pyridin-2-yl, phenyl optionally substituted by 1 or 2 groups such as 1 substitutent, wherein said substituents are independently selected from methyl, t-butyl, fluoro, chloro, and -OCF3. Optional substituents on the phenyl may, for example, be in the ortho or para position.
In one embodiment R1 is a heteroaromatic group optionally substituted by one or two groups selected from hydroxy, C-| _3alkyl, C-| _3alkoxy, hydroxyC-| _4alkyl and C-μ 4alkoxyC-| _4alkyl. In one embodiment the heteroaromatic group is pyridyl. In a further embodiment the heteroaromatic group is selected from furanyl, thienyl, isoxazolyl, thiazolyl, pyrazolyl, pyrazinyl and pyrimidinyl.
In one embodiment, n is 0 and R1 is phenyl substituted by Ci-6alkyl (such as phenyl substituted by t-butyl), or phenyl substituted by haloCi-6alkoxy (such as OCF3).
In one embodiment n is 1 and R1 is unsubstituted phenyl.
In one embodiment n is 1 or 2 such as 1 and R1 is phenyl optionally substituted by, for example 1 or 2 halogens such as chlorine, and/or fluoro.
In one embodiment R2 is hydrogen or methyl.
In one embodiment R2a is H, C-i ^alkyl, (CH2)mOH, (CH2)mC<| _3alkoxy, (CH2)mNRaRb or (CHR6)pheterocyclyl,
wherein
Ra represents H, C-| _3alkyl, or heterocyclyl;
Rb represents H or C-| _3alkyl, or
Ra and Rb together with the N to which they are attached form a 5 or 6 membered heterocyclyl;
R6 represents H or C<| _3alkyl;
m represents 1 , 2 or 3; and
p represents 0, 1 , 2. In one embodiment, R2a is hydrogen, C-| .galkyl such as methyl, isopropyl or t-butyl or C-μ galkoxyC-| .galkyl such as -(CH2)20CH3.
In one embodiment R2a is tetrahydropyranyl. In one embodiment R2b is hydrogen.
In one embodiment R2b is (CH2)2C-| .galkoxy such as (CH2)20Me.
In one embodiment Ra and Rb together with the N to which they are attached form a 5 or 6 membered heterocyclyl such as morpholine, piperidine or pyrrolidine.
In one embodiment R4 is hydrogen.
In one embodiment, R5 is hydrogen or -OCI-13 such as -OCH3. In one embodiment X is CH, Y is CH and R5 is -OCH3, for example in the 8 position. In one embodiment X is N, Y is CH and R5 is -OCH3, for example in the 8 position In one embodiment, the compounds of the present invention have a formula (IA):
Figure imgf000010_0001
wherein A and R4 are as defined for formula (I).
one embodiment, the compounds of the present invention have the formula (IB):
Figure imgf000011_0001
wherein R1, R2, R3, R5 and n are as defined for compounds of formula (I). In this aspect of the invention n may, for example, represent 1.
In this aspect of the invention R1 may represent, for example pyridinyl such as pyridin-2-yl or phenyl.
In this aspect of the invention R5 may represent, for example, -OCH3.
In one embodiment, compounds of the invention have the formula (IC)
Figure imgf000011_0002
wherein R1, R2, R3, R2a and n are defined above for compounds of formula (I).
It is to be understood that the present invention covers all combinations of suitable, convenient and preferred groups described herein above. Thus all features and embodiments of formula (I) may apply to formula (IA), (IB) and (IC).
Specific compounds of formula (I) include Examples 1 - 226 as described herein or a salt thereof, in particular a pharmaceutically acceptable salt thereof. In one embodiment there is provided a compound which is 7-(3,5-dimethyl-4-isoxazolyl)- 8-(methyloxy)-1 -[(1 R)-1 -(2-pyridinyl)ethyl]-1 ,3-dihydro-2H-imidazo[4,5-c]quinolin-2-one or a salt thereof. In another embodiment there is provided a compound which is 7-(3,5- dimethyl-4-isoxazolyl)-8-(methyloxy)-1 -[(1 R)-1 -(2-pyridinyl)ethyl]-1 ,3-dihydro-2H- imidazo[4,5-c]quinolin-2-one or a pharmaceutically acceptable salt thereof. In another embodiment there is provided a compound which is 7-(3,5-dimethyl-4-isoxazolyl)-8- (methyloxy)-l -[(1 R)-1 -(2-pyridinyl)ethyl]-1 ,3-dihydro-2H-imidazo[4,5-c]quinolin-2-one.
It will be appreciated that the present invention covers compounds of formula (I) as the free base and as salts thereof, for example as a pharmaceutically acceptable salt thereof. In one embodiment the invention relates to compounds of formula (I) or a pharmaceutically acceptable salt thereof.
Because of their potential use in medicine, salts of the compounds of formula (I) are desirably pharmaceutically acceptable. Suitable pharmaceutically acceptable salts can include acid or base addition salts. As used herein, the term 'pharmaceutically acceptable salt' means any pharmaceutically acceptable salt or solvate of a compound of formula (I), which upon administration to the recipient is capable of providing (directly or indirectly). In one embodiment, the term 'pharmaceutically acceptable salt' means any pharmaceutically acceptable salt of a compound of formula (I), which upon administration to the recipient is capable of providing (directly or indirectly). For a review on suitable salts see Berge et al., J. Pharm. Sci., 66:1 -19, (1977). Typically, a pharmaceutically acceptable salt may be readily prepared by using a desired acid or base as appropriate. The resultant salt may precipitate from solution and be collected by filtration or may be recovered by evaporation of the solvent.
A pharmaceutically acceptable base addition salt can be formed by reaction of a compound of formula (I) with a suitable inorganic or organic base, (e.g. triethylamine, ethanolamine, triethanolamine, choline, arginine, lysine or histidine), optionally in a suitable solvent, to give the base addition salt which is usually isolated, for example, by crystallisation and filtration. Pharmaceutically acceptable base salts include ammonium salts, alkali metal salts such as those of sodium and potassium, alkaline earth metal salts such as those of calcium and magnesium and salts with organic bases, including salts of primary, secondary and tertiary amines, such as isopropylamine, diethylamine, ethanolamine, trimethylamine, dicyclohexyl amine and N-methyl-D-glucamine. A pharmaceutically acceptable acid addition salt can be formed by reaction of a compound of formula (I) with a suitable inorganic or organic acid (such as hydrobromic, hydrochloric, sulphuric, nitric, phosphoric, succinc, maleic, acetic, propionic, fumaric, citric, tartaric, lactic, benzoic, salicylic, glutamaic, aspartic, p-toluenesulfonic, benzenesulfonic, methanesulfonic, ethanesulfonic, naphthalenesulfonic such as 2- naphthalenesulfonic, or hexanoic acid), optionally in a suitable solvent such as an organic solvent, to give the salt which is usually isolated for example by crystallisation and filtration. A pharmaceutically acceptable acid addition salt of a compound of formula (I) can comprise or be for example a hydrobromide, hydrochloride, sulfate, nitrate, phosphate, succinate, maleate, acetate, propionate, fumarate, citrate, tartrate, lactate, benzoate, salicylate, glutamate, aspartate, p-toluenesulfonate, benzenesulfonate, methanesulfonate, ethanesulfonate, naphthalenesulfonate (e.g. 2-naphthalenesulfonate) or hexanoate salt. Other non-pharmaceutically acceptable salts, e.g. formates, oxalates or trifluoroacetates, may be used, for example in the isolation of the compounds of formula (I), and are included within the scope of this invention.
The invention includes within its scope all possible stoichiometric and non-stoichiometric forms of the salts of the compounds of formula (I).
It will be appreciated that many organic compounds can form complexes with solvents in which they are reacted or from which they are precipitated or crystallized. These complexes are known as "solvates". For example, a complex with water is known as a "hydrate". Solvents with high boiling points and/or capable of forming hydrogen bonds such as water, xylene, /V-methyl pyrrolidinone, methanol and ethanol may be used to form solvates. Methods for identification of solvates include, but are not limited to, NMR and microanalysis. Solvates of the compounds of formula (I) are within the scope of the invention.
The invention includes within its scope all possible stoichiometric and non-stoichiometric forms of the solvates of the compounds of formula (I).
The invention encompasses all prodrugs, of the compound of formula (I) or a pharmaceutically acceptable salt thereof, which upon administration to the recipient is capable of providing (directly or indirectly) the compound of formula (I) or a pharmaceutically acceptable salt thereof, or an active metabolite or residue thereof. Such derivatives are recognizable to those skilled in the art, without undue experimentation. Nevertheless, reference is made to the teaching of Burger's Medicinal Chemistry and Drug Discovery, 5th Edition, Vol 1 : Principles and Practice, which is incorporated herein by reference to the extent of teaching such derivatives.
The compounds of formula (I) may be in crystalline or amorphous form. Furthermore, some of the crystalline forms of the compounds of formula (I) may exist as polymorphs, which are included within the scope of the present invention. Polymorphic forms of compounds of formula (I) may be characterized and differentiated using a number of conventional analytical techniques, including, but not limited to, X-ray powder diffraction (XRPD) patterns, infrared (IR) spectra, Raman spectra, differential scanning calorimetry (DSC), thermogravimetric analysis (TGA) and solid state nuclear magnetic resonance (SSNMR).
Certain compounds described herein may contain one or more chiral atoms so that optical isomers, e.g. enantiomers or diastereoisomers may be formed. Accordingly, the present invention encompasses all isomers of the compounds of formula (I) whether as individual isomers isolated such as to be substantially free of the other isomer (i.e. pure) or as mixtures (i.e. racemates and racemic mixtures). An individual isomer isolated such as to be substantially free of the other isomer (i.e. pure) may be isolated such that less than 10%, particularly less than about 1 %, for example less than about 0.1 % of the other isomer is present. Separation of isomers may be achieved by conventional techniques known to those skilled in the art, e.g. by fractional crystallisation, chromatography or HPLC.
Certain compounds of formula (I) may exist in one of several tautomeric forms. It will be understood that the present invention encompasses all tautomers of the compounds of formula (I) whether as individual tautomers or as mixtures thereof.
It will be appreciated from the foregoing that included within the scope of the invention are solvates, isomers and polymorphic forms of the compounds of formula (I) and salts thereof. The compounds of formula (I) or salts thereof may be made by a variety of methods, including standard chemistry. Any previously defined variable will continue to have the previously defined meaning unless otherwise indicated. Illustrative general synthetic methods are set out below and then specific compounds of formula (I) and pharmaceutically acceptable salts thereof, are prepared in the working Examples. These processes form further aspects of the present invention.
Throughout the specification, general formulae are designated by Roman numerals (I), (II), (III), (IV) etc.
In another aspect, the present invention provides a process for the preparation of a compound of formula (I) wherein Z represents O, the process comprising the step of reacting a compound of formula (II):
Figure imgf000015_0001
wherein X, Y, R1, R2 and R3 are as defined for compounds of formula (I) with:
a) R2aCOOH, R2aCHO or R2aCOCI, wherein R2a is as defined above for compounds of formula (I), or
b) (t-butylOCO)20, or
c) CS2, or
d) NaN02,
under appropriate conditions.
Step a) may be effected by refluxing in a suitable solvent such as acetic acid.
When R2aCOOH is the reagent, prior to refluxing in acetic acid, activation of the starting material with a reagent such as HOBT may be required. The coupling reaction may be performed in the presence of a coupling agent such as EDCI and an organic base such as triethylamine, in a suitable solvent, for example a polar aprotic solvent such as dichloromethane.
When an acid chloride R2aCOCI is employed, prior to refluxing in acetic acid, the acid chloride is allowed to react with the compound of formula (II) in a suitable solvent, for example a polar aprotic solvent such as dichloromethane in the presence of an organic base such as triethylamine.
Step b) may be performed at 80°C for a period of about 2h, followed by treatment at an elevated temperature, for example, 180°C for a period of about 3h in a solvent such as diphenylether.
Step c) may be effected at elevated temperatures, for example above 50°C such as 80°C, under basic conditions, for example in the presence of triethylamine in a suitable solvent, for example an alcohol such as ethanol.
Step d) may be effected at low temperature, for example sodium nitrite is added at 0°C, after which the reaction mixture is allowed to warm to room temperature and continue reacting for about 18h.
In the schemes below X, Y, R1, R2, R3, n etc have the definitions as given above for compounds of formula (I).
Compounds of formula (I) wherein R4 represents H or alkyl and A is an imidazole derivative can be prepared as shown in Scheme 1 below.
Scheme 1
Figure imgf000017_0001
The imidazole derivatives of general formula (IE) may be prepared (scheme 1 ) by reacting the amino derivatives (IIA) with aldehydes (R2CHO) in acetic acid at reflux, or by coupling the derivatives (IIA) with carboxylic acid of general formula R2COOH with HOBT, EDCI in dichloromethane in presence of triethylamine, followed by cyclisation in acetic acid at reflux, or by coupling with acid chloride of general formula R2COCI in presence of triethylamine followed by cyclisation in acetic acid at reflux. Compounds of formula (I) wherein R4 represents H or alkyl, and A represents an imidazolone can be prepared as shown in Scheme 2 below.
Scheme 2
Figure imgf000017_0002
The imidazolone compounds of general formula (IF) can be prepared, according to scheme 2, by reacting the compounds (IIA) with Boc anhydride followed by a cyclisation in diphenyl ether at 180°C. The imidazolone compounds (IF) may be prepared using Hoffman-type rearrangement conditions (Scheme 3). In this case the compounds (MB) are reacted with bis(trifluoroacetoxy)iodo]benzene, to give after internal cyclisation the imidazolone compounds (IF).
Scheme 3
Figure imgf000018_0001
The imidazolone compounds (IF) may also be prepared using Curtius rearrangement conditions (Scheme 4). In this case the acid compounds (IIC) are reacted with diphenyl phosphoryl azide in the presence of triethylamine for a period of approximately 2 to 18h to give the imidazolone compounds (IF).
Scheme 4
Figure imgf000018_0002
Compounds of formula (I) wherein R4 is H, and A is an imidazol-2-thione derivative can be prepared as shown in Scheme 5 below. Scheme 5
Figure imgf000019_0001
The imidazol-2-thione derivatives of general formula (IG) can be prepared by reacting the compounds of general formula (IIA) with carbon disulfide in the presence of triethylamine in a solvent such as ethanol at 60°-80°C for approximately 18h.
Compounds of formula (I) wherein R4 is H or alkyl, and A is a triazole derivative can be prepared as shown in Scheme 6 below. The amino derivative (IIA) is reacted at at 0°C with sodium nitrite in water and acetic acid in a solvent as DMF. The reaction mixture is then stirred at room temperature for approximately 18h.
Scheme 6
Figure imgf000019_0002
Compounds of formula (I) wherein R4 is cyano and A is an imidazole derivative can be prepared as shown in Scheme 7 below, using modified a Reissert-Henze reaction (Harusawa, S et al., Heterocycles, 1981, 15, 981-984). Compound (IE) is reacted with m- chloroperbenzoic acid in dichloromethane at room temperature for approximately 2h. The resulting N-oxide intermediate is then reacted with diethyl phosphorocyanidate in the presence of triethylamine in an amprotic solvent such as acetonitrile, the reaction mixture is then heated under reflux for 4h.
Scheme 7
Figure imgf000020_0001
The starting material (IE) in Scheme 7 can be prepared by an analogous method to that described above in Scheme 1. Compounds of general formula (I I A) wherein R4 is hydrogen and alkyl may be prepared according to Scheme 8. Compounds (IIIA) are reacted with sodium hydroxide in ethanol under reflux for 5h. After treatment with HCI N, the resulting compounds of general formula (VA) are de-carboxylated in diphenylether under reflux for 2h. Nitration of compounds (VA) is preformed with nitric acid in propanoic acid at room temperature, followed by heating the reaction mixture to a range of temperature 100-125°C for 1 to 2h. Compounds (VIA) are reacted with POCI3 in toluene under reflux for approximately 18h. The chloro compounds (VI IA) are then reacted with amines of general formula R1 (R2R3C)nNH2 in a solvent such as acetonitrile at 60°C for 2h. The resulting compounds may then be reduced with SnCl2,2H20 in ethanol/HCI or ethanol/THF at a temperature in the range 40°C to reflux for approximately 1 to 3h, to give compounds of general formula (I I A).
Scheme 8
Figure imgf000021_0001
Compounds of general formula (VA) wherein X and Y are CH and R4 is hydrogen may be also prepared according to Scheme 9.
Scheme 9
Figure imgf000021_0002
The 3-iodo aniline derivatives are reacted meldrum's acid in presence of methyl orthoformate under reflux for approximately 1 h, followed by cyclisation in boiling diphenylether for approximately 10 minutes. Compounds of general formula (VA) wherein R4 is alkyl, for example methyl, may be prepared following the same synthetic route by replacing meldrum's acid by ethyl acetoacetate. Compounds of formula VA wherein R5 is situated at a different position on the ring can be prepared by a method analogous to that set out in Scheme 8, starting from appropriate starting materials.
Compounds of general formula (MB) wherein R4 is hydrogen or alkyl may be prepared according to Scheme 10. Compounds (IVA) are treated with POCI3 with DMF under reflux. The corresponding 4-chloro-3-chloroacyl derivatives are then reacted with ammonia^) at 0°-5°C in a solvent such as dioxane to afford the carboxamide compounds (VINA). Coupling with the amine was performed as previously described for Scheme 8.
Scheme 10
Figure imgf000022_0001
Compounds of general formula (IIIA), wherein R4 is hydrogen may be prepared according to Scheme 1 1 .
Scheme 1 1
Figure imgf000022_0002
The 3,5-dimethylisoxazole boronic acid may be reacted with 3-iodo or 3-bromo derivatives using Suzuki coupling conditions to give the corresponding amino compounds.
When X and Y are CH, the 3-(3,5-dimethyl-4-isoxazolyl)aniline may be reacted with diethyl ethoxymethylenemalonate at 130°C of a period in the range 20 minutes to 1 h. The resulting diethyl ({[3-(3,5-dimethyl-4-isoxazolyl)phenyl] amino}methylidene)-propanedioate may be cyclised in diphenylether by refluxing for approximately 30 minutes to 1 h (according to the standard procedure described in J.Med.Chem., 1980, 23, 1358). The same procedure may be applied to aminopyridine compounds to prepare the 1 ,5- or 1 ,6- naphthyridine compounds.
Compounds of formula (I) wherein R4 is alkyl, for example methyl, can be prepared by analogous methods to those described Scheme 1 1 replacing diethyl ethoxymethylenemalonate by diethyl acetylmalonate.
Compounds of general formula (IIC), wherein R4 is hydrogen or alkyl may be prepared according to Scheme 12.
Scheme 12
Figure imgf000023_0001
Compounds (IIIA) are refluxed in POCI3 for approximately 18h, and compounds (XA) are then coupled with amines of general formula R1(R2R3C)nNH2 in a solvent such as acetonitrile or dioxane at a temperature in the range 60° to 1 10°C for approximately 1 to 4h. Saponification of compounds (IID) with NaOH (N) in Ethanol heated to reflux, for approximately 6 to 24h, affords compounds of formula (IIC).
Compounds of formula (I) wherein Z represents NH (and in consequence R4 represents hydrogen, A is a group selected from (i) or (ii) as defined above, and Rx represents O) can be prepared according to processes as shown in Schemes 13 to 15 below.
Figure imgf000024_0001
Scheme 15
Figure imgf000025_0001
It will be appreciated by those skilled in the art that it may be advantageous to protect one or more functional groups of the compounds described above. Examples of protecting groups and the means for their removal can be found in T. W. Greene 'Protective Groups in Organic Synthesis' (4th edition, J. Wiley and Sons, 2006). Suitable amine protecting groups include acyl (e.g. acetyl, carbamate (e.g. 2',2',2'-trichloroethoxycarbonyl, benzyloxycarbonyl or t-butoxycarbonyl) and arylalkyl (e.g. benzyl), which may be removed by hydrolysis (e.g. using an acid such as hydrochloric acid in dioxane or trifluoroacetic acid in dichloromethane) or reductively (e.g. hydrogenolysis of a benzyl or benzyloxycarbonyl group or reductive removal of a 2',2',2'-trichloroethoxycarbonyl group using zinc in acetic acid) as appropriate. Other suitable amine protecting groups include trifluoroacetyl (-COCF3) which may be removed by base catalysed hydrolysis.
It will be appreciated that in any of the routes described above, the precise order of the synthetic steps by which the various groups and moieties are introduced into the molecule may be varied. It will be within the skill of the practitioner in the art to ensure that groups or moieties introduced at one stage of the process will not be affected by subsequent transformations and reactions, and to select the order of synthetic steps accordingly. Certain intermediate compounds described above are believed to be novel and therefore form a yet further aspect of the invention. The compounds of formula (I) and salts thereof are bromodomain inhibitors, and thus are believed to have potential utility in the treatment of diseases or conditions for which a bromodomain inhibitor is indicated.
The present invention thus provides a compound of formula (I) or a pharmaceutically acceptable salt thereof for use in therapy. In one embodiment there is provided 7-(3,5- dimethyl-4-isoxazolyl)-8-(methyloxy)-1 -[(1 R)-1 -(2-pyridinyl)ethyl]-1 ,3-dihydro-2H- imidazo[4,5-c]quinolin-2-one or a pharmaceutically acceptable salt thereof for use in therapy. The compound of formula (I) or a pharmaceutically salt thereof can be used in the treatment of diseases or conditions for which a bromodomain inhibitor is indicated.
The present invention thus provides a compound of formula (I) or a pharmaceutically acceptable salt thereof for use in the treatment of any diseases or conditions for which a bromodomain inhibitor is indicated. In another embodiment there is provided a compound or a pharmaceutically acceptable salt thereof for use in the treatment of chronic auto- immune and/or inflammatory conditions. In a further embodiment there is provided a compound or a pharmaceutically acceptable salt thereof for use in the treatment of cancer.
In one embodiment there is provided 7-(3,5-dimethyl-4-isoxazolyl)-8-(methyloxy)-1 -[(1 R)- 1 -(2-pyridinyl)ethyl]-1 ,3-dihydro-2H-imidazo[4,5-c]quinolin-2-one or a pharmaceutically acceptable salt thereof for use in the treatment of diseases or conditions for which a bromodomain inhibitor indicated.
Also provided is the use of a compound of formula (I) or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for the treatment of diseases or conditions for which a bromodomain inhibitor is indicated. In one embodiment there is provided 7-(3,5-dimethyl-4-isoxazolyl)-8-(methyloxy)-1 -[(1 R)-1 -(2-pyridinyl)ethyl]-1 ,3- dihydro-2H-imidazo[4,5-c]quinolin-2-one or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for the treatment of diseases or conditions for which a bromodomain inhibitor is indicated. Also provided is a method of treating diseases or conditions for which a bromodomain inhibitor is indicated in a subject in need thereof which comprises administering a therapeutically effective amount of compound of formula (I) or a pharmaceutically acceptable salt thereof. In one embodiment there is provided a method of treating diseases or conditions for which a bromodomain inhibitor is indicated in a subject in need thereof which comprises administering a therapeutically effective amount of 7-(3,5- dimethyl-4-isoxazolyl)-8-(methyloxy)-1 -[(1 R)-1 -(2-pyridinyl)ethyl]-1 ,3-dihydro-2H- imidazo[4,5-c]quinolin-2-one or a pharmaceutically acceptable salt thereof. Suitably the subject in need thereof is a mammal, particularly a human.
As used herein, the term "effective amount" means that amount of a drug or pharmaceutical agent that will elicit the biological or medical response of a tissue, system, animal or human that is being sought, for instance, by a researcher or clinician. Furthermore, the term "therapeutically effective amount" means any amount which, as compared to a corresponding subject who has not received such amount, results in improved treatment, healing, prevention, or amelioration of a disease, disorder, or side effect, or a decrease in the rate of advancement of a disease or disorder. The term also includes within its scope amounts effective to enhance normal physiological function.
Bromodomain inhibitors are believed to be useful in the treatment of a variety of diseases or conditions related to systemic or tissue inflammation, inflammatory responses to infection or hypoxia, cellular activation and proliferation, lipid metabolism, fibrosis and in the prevention and treatment of viral infections.
Bromodomain inhibitors may be useful in the treatment of a wide variety of chronic autoimmune and inflammatory conditions such as rheumatoid arthritis, osteoarthritis, acute gout, psoriasis, systemic lupus erythematosus, multiple sclerosis, inflammatory bowel disease (Crohn's disease and Ulcerative colitis), asthma, chronic obstructive airways disease, pneumonitis, myocarditis, pericarditis, myositis, eczema, dermatitis, alopecia, vitiligo, bullous skin diseases, nephritis, vasculitis, atherosclerosis, Alzheimer's disease, depression, retinitis, uveitis, scleritis, hepatitis, pancreatitis, primary biliary cirrhosis, sclerosing cholangitis, Addison's disease, hypophysitis, thyroiditis, type I diabetes and acute rejection of transplanted organs. Bromodomain inhibitors may be useful in the treatment of a wide variety of acute inflammatory conditions such as acute gout, giant cell arteritis, nephritis including lupus nephritis, vasculitis with organ involvement such as glomerulonephritis, vasculitis including giant cell arteritis, Wegener's granulomatosis, Polyarteritis nodosa, Behcet's disease, Kawasaki disease, Takayasu's Arteritis, vasculitis with organ involvement and acute rejection of transplanted organs.
Bromodomain inhibitors may be useful in the prevention or treatment of diseases or conditions which involve inflammatory responses to infections with bacteria, viruses, fungi, parasites or their toxins, such as sepsis, sepsis syndrome, septic shock, endotoxaemia, systemic inflammatory response syndrome (SIRS), multi-organ dysfunction syndrome, toxic shock syndrome, acute lung injury, ARDS (adult respiratory distress syndrome), acute renal failure, fulminant hepatitis, burns, acute pancreatitis, post-surgical syndromes, sarcoidosis, Herxheimer reactions, encephalitis, myelitis, meningitis, malaria and SIRS associated with viral infections such as influenza, herpes zoster, herpes simplex and coronavirus.
Bromodomain inhibitors may be useful in the prevention or treatment of conditions associated with ischaemia-reperfusion injury such as myocardial infarction, cerebro- vascular ischaemia (stroke), acute coronary syndromes, renal reperfusion injury, organ transplantation, coronary artery bypass grafting, cardio-pulmonary bypass procedures, pulmonary, renal, hepatic, gastro-intestinal or peripheral limb embolism.
Bromodomain inhibitors may be useful in the treatment of disorders of lipid metabolism via the regulation of APO-A1 such as hypercholesterolemia, atherosclerosis and Alzheimer's disease.
Bromodomain inhibitors may be useful in the treatment of fibrotic conditions such as idiopathic pulmonary fibrosis, renal fibrosis, post-operative stricture, keloid formation, scleroderma and cardiac fibrosis.
Bromodomain inhibitors may be useful in the prevention and treatment of viral infections such as herpes virus, human papilloma virus, adenovirus and poxvirus and other DNA viruses. Bromodomain inhibitors may be useful in the treatment of cancer, including hematological, epithelial including lung, breast and colon carcinomas, midline carcinomas, mesenchymal, hepatic, renal and neurological tumours. In one embodiment the disease or condition for which a bromodomain inhibitor is indicated is selected from diseases associated with systemic inflammatory response syndrome, such as sepsis, burns, pancreatitis, major trauma, haemorrhage and ischaemia. In this embodiment the bromodomain inhibitor would be administered at the point of diagnosis to reduce the incidence of: SIRS, the onset of shock, multi-organ dysfunction syndrome, which includes the onset of acute lung injury, ARDS, acute renal, hepatic, cardiac and gastro-intestinal injury and mortality. In another embodiment the bromodomain inhibitor would be administered prior to surgical or other procedures associated with a high risk of sepsis, haemorrhage, extensive tissue damage, SIRS or MODS (multiple organ dysfunction syndrome). In a particular embodiment the disease or condition for which a bromodomain inhibitor is indicated is sepsis, sepsis syndrome, septic shock and endotoxaemia. In another embodiment, the bromodomain inhibitor is indicated for the treatment of acute or chronic pancreatitis. In another embodiment the bromodomain is indicated for the treatment of burns. In one embodiment the disease or condition for which a bromodomain inhibitor is indicated is selected from herpes simplex infections and reactivations, cold sores, herpes zoster infections and reactivations, chickenpox, shingles, human papilloma virus, cervical neoplasia, adenovirus infections, including acute respiratory disease, poxvirus infections such as cowpox and smallpox and African swine fever virus. In one particular embodiment a bromodomain inhibitor is indicated for the treatment of Human papilloma virus infections of skin or cervical epithelia.
The term "diseases or conditions for which a bromodomain inhibitor is indicated", is intended to include each of or all of the above disease states.
While it is possible that for use in therapy, a compound of formula (I) as well as pharmaceutically acceptable salts thereof may be administered as the raw chemical, it is common to present the active ingredient as a pharmaceutical composition.
The present invention therefore provides in a further aspect a pharmaceutical composition comprising a compound of formula (I) or a pharmaceutically acceptable salt and one or more pharmaceutically acceptable carriers, diluents and/or excipients. The compounds of formula (I) and pharmaceutically acceptable salts, are as described above. The carrier(s), diluent(s) or excipient(s) must be acceptable in the sense of being compatible with the other ingredients of the composition and not deleterious to the recipient thereof. In accordance with another aspect of the invention there is also provided a process for the preparation of a pharmaceutical composition including admixing a compound of formula (I), or a pharmaceutically acceptable salt thereof, with one or more pharmaceutically acceptable carriers, diluents or excipients. The pharmaceutical composition can be used in the treatment of any of the conditions described herein.
In one embodiment there is provided a pharmaceutical composition comprising 7-(3,5- dimethyl-4-isoxazolyl)-8-(methyloxy)-1 -[(1 R)-1 -(2-pyridinyl)ethyl]-1 ,3-dihydro-2H- imidazo[4,5-c]quinolin-2-one or a pharmaceutically acceptable salt and one or more or pharmaceutically acceptable carriers, diluents or excipients.
Since the compounds of formula (I) are intended for use in pharmaceutical compositions it will be readily understood that they are each preferably provided in substantially pure form, for example, at least 60% pure, more suitably at least 75% pure and preferably at least 85% pure, especially at least 98% pure (% in a weight for weight basis).
Pharmaceutical compositions may be presented in unit dose forms containing a predetermined amount of active ingredient per unit dose. Preferred unit dosage compositions are those containing a daily dose or sub-dose, or an appropriate fraction thereof, of an active ingredient. Such unit doses may therefore be administered more than once a day. Preferred unit dosage compositions are those containing a daily dose or sub- dose (for administration more than once a day), as herein above recited, or an appropriate fraction thereof, of an active ingredient.
Pharmaceutical compositions may be adapted for administration by any appropriate route, for example by the oral (including buccal or sublingual), rectal, inhaled, intranasal, topical (including buccal, sublingual or transdermal), vaginal or parenteral (including subcutaneous, intramuscular, intravenous or intradermal) route. Such compositions may be prepared by any method known in the art of pharmacy, for example by bringing into association the active ingredient with the carrier(s) or excipient(s). In one embodiment the pharmaceutical composition is adapted for parenteral administration, particularly intravenous administration.
In one embodiment the pharmaceutical composition is adapted for oral administration.
Pharmaceutical compositions adapted for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the composition isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents. The compositions may be presented 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 injections, immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets.
Pharmaceutical compositions adapted for oral administration may be presented as discrete units such as capsules or tablets; powders or granules; solutions or suspensions in aqueous or non-aqueous liquids; edible foams or whips; or oil-in-water liquid emulsions or water-in-oil liquid emulsions.
For instance, for oral administration in the form of a tablet or capsule, the active drug component can be combined with an oral, non-toxic pharmaceutically acceptable inert carrier such as ethanol, glycerol, water and the like. Powders suitable for incorporating into tablets or capsules may be prepared by reducing the compound to a suitable fine size (e.g. by micronisation) and mixing with a similarly prepared pharmaceutical carrier such as an edible carbohydrate, for example, starch or mannitol. Flavoring, preservative, dispersing and coloring agent can also be present. Capsules may be made by preparing a powder mixture, as described above, and filling formed gelatin sheaths. Glidants and lubricants such as colloidal silica, talc, magnesium stearate, calcium stearate or solid polyethylene glycol can be added to the powder mixture before the filling operation. A disintegrating or solubilizing agent such as agar- agar, calcium carbonate or sodium carbonate can also be added to improve the availability of the medicament when the capsule is ingested. Moreover, when desired or necessary, suitable binders, glidants, lubricants, sweetening agents, flavours, disintegrating agents and coloring agents can also be incorporated into the mixture. Suitable binders include starch, gelatin, natural sugars such as glucose or beta-lactose, corn sweeteners, natural and synthetic gums such as acacia, tragacanth or sodium alginate, carboxymethylcellulose, polyethylene glycol, waxes and the like. Lubricants used in these dosage forms include sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride and the like. Disintegrators include, without limitation, starch, methyl cellulose, agar, bentonite, xanthan gum and the like. Tablets are formulated, for example, by preparing a powder mixture, granulating or slugging, adding a lubricant and disintegrant and pressing into tablets. A powder mixture is prepared by mixing the compound, suitably comminuted, with a diluent or base as described above, and optionally, with a binder such as carboxymethylcellulose, an aliginate, gelatin, or polyvinyl pyrrolidone, a solution retardant such as paraffin, a resorption accelerator such as a quaternary salt and/or an absorption agent such as bentonite, kaolin or dicalcium phosphate. The powder mixture can be granulated by wetting with a binder such as syrup, starch paste, acadia mucilage or solutions of cellulosic or polymeric materials and forcing through a screen. As an alternative to granulating, the powder mixture can be run through the tablet machine and the result is imperfectly formed slugs broken into granules. The granules can be lubricated to prevent sticking to the tablet forming dies by means of the addition of stearic acid, a stearate salt, talc or mineral oil. The lubricated mixture is then compressed into tablets. The compounds of formula (I) and pharmaceutically acceptable salts thereof, can also be combined with a free flowing inert carrier and compressed into tablets directly without going through the granulating or slugging steps. A clear or opaque protective coating consisting of a sealing coat of shellac, a coating of sugar or polymeric material and a polish coating of wax can be provided. Dyestuffs can be added to these coatings to distinguish different unit dosages.
Oral fluids such as solution, syrups and elixirs can be prepared in dosage unit form so that a given quantity contains a predetermined amount of the compound. Syrups can be prepared by dissolving the compound in a suitably flavored aqueous solution, while elixirs are prepared through the use of a non-toxic alcoholic vehicle. Suspensions can be formulated by dispersing the compound in a non-toxic vehicle. Solubilizers and emulsifiers such as ethoxylated isostearyl alcohols and polyoxy ethylene sorbitol ethers, preservatives, flavor additive such as peppermint oil or natural sweeteners or saccharin or other artificial sweeteners, and the like can also be added. Where appropriate, dosage unit compositions for oral administration can be microencapsulated. The formulation can also be prepared to prolong or sustain the release as for example by coating or embedding particulate material in polymers, wax or the like.
The compounds of formula (I) and pharmaceutically acceptable salts thereof, can also be administered in the form of liposome delivery systems, such as small unilamellar vesicles, large unilamellar vesicles and multilamellar vesicles. Liposomes can be formed from a variety of phospholipids, such as cholesterol, stearylamine or phosphatidylcholines.
Pharmaceutical compositions adapted for topical administration may be formulated as ointments, creams, suspensions, lotions, powders, solutions, pastes, gels, sprays, aerosols or oils.
For treatments of the eye or other external tissues, for example mouth and skin, the compositions are preferably applied as a topical ointment or cream. When formulated in an ointment, the active ingredient may be employed with either a paraffinic or a water- miscible ointment base. Alternatively, the active ingredient may be formulated in a cream with an oil-in-water cream base or a water-in-oil base.
Pharmaceutical compositions adapted for topical administrations to the eye include eye drops wherein the active ingredient is dissolved or suspended in a suitable carrier, especially an aqueous solvent.
Dosage forms for nasal or inhaled administration may conveniently be formulated as aerosols, solutions, suspensions, gels or dry powders.
For compositions suitable and/or adapted for inhaled administration, it is preferred that the compound of formula (I) or a pharmaceutically acceptable salt thereof, is in a particle-size- reduced form e.g. obtained by micronisation. The preferable particle size of the size- reduced (e.g. micronised) compound or salt is defined by a D50 value of about 0.5 to about 10 microns (for example as measured using laser diffraction). Aerosol formulations, e.g. for inhaled administration, can comprise a solution or fine suspension of the active substance in a pharmaceutically acceptable aqueous or non- aqueous solvent. Aerosol formulations can be presented in single or multidose quantities in sterile form in a sealed container, which can take the form of a cartridge or refill for use with an atomising device or inhaler. Alternatively the sealed container may be a unitary dispensing device such as a single dose nasal inhaler or an aerosol dispenser fitted with a metering valve (metered dose inhaler) which is intended for disposal once the contents of the container have been exhausted.
Where the dosage form comprises an aerosol dispenser, it preferably contains a suitable propellant under pressure such as compressed air, carbon dioxide or an organic propellant such as a hydrofluorocarbon (HFC). Suitable HFC propellants include 1 ,1 ,1 ,2,3,3,3-heptafluoropropane and 1 ,1 ,1 ,2-tetrafluoroethane. The aerosol dosage forms can also take the form of a pump-atomiser. The pressurised aerosol may contain a solution or a suspension of the active compound. This may require the incorporation of additional excipients e.g. co-solvents and/or surfactants to improve the dispersion characteristics and homogeneity of suspension formulations. Solution formulations may also require the addition of co-solvents such as ethanol.
For pharmaceutical compositions suitable and/or adapted for inhaled administration, the pharmaceutical composition may be a dry powder inhalable composition. Such a composition can comprise a powder base such as lactose, glucose, trehalose, mannitol or starch, the compound of formula (I) or a pharmaceutically acceptable salt thereof (preferably in particle-size-reduced form, e.g. in micronised form), and optionally a performance modifier such as L-leucine or another amino acid and/or metals salts of stearic acid such as magnesium or calcium stearate. Preferably, the dry powder inhalable composition comprises a dry powder blend of lactose e.g. lactose monohydrate and the compound of formula (I) or salt thereof. Such compositions can be administered to the patient using a suitable device such as the DISKUS® device, marketed by GlaxoSmithKline which is for example described in GB 2242134 A. The compounds of formula (I) and pharmaceutically acceptable salts thereof may be formulated as a fluid formulation for delivery from a fluid dispenser, for example a fluid dispenser having a dispensing nozzle or dispensing orifice through which a metered dose of the fluid formulation is dispensed upon the application of a user-applied force to a pump mechanism of the fluid dispenser. Such fluid dispensers are generally provided with a reservoir of multiple metered doses of the fluid formulation, the doses being dispensable upon sequential pump actuations. The dispensing nozzle or orifice may be configured for insertion into the nostrils of the user for spray dispensing of the fluid formulation into the nasal cavity. A fluid dispenser of the aforementioned type is described and illustrated in WO-A-2005/044354. A therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof, will depend upon a number of factors including, for example, the age and weight of the animal, the precise condition requiring treatment and its severity, the nature of the formulation, and the route of administration, and will ultimately be at the discretion of the attendant physician or veterinarian. In the pharmaceutical composition, each dosage unit for oral or parenteral administration preferably contains from 0.01 to 3000 mg, more preferably 0.5 to 1000 mg, of a compound of formula (I) or a pharmaceutically acceptable salt thereof, calculated as the free base. Each dosage unit for nasal or inhaled administration preferably contains from 0.001 to 50 mg, more preferably 0.01 to 5 mg, of a compound of the formula (I) or a pharmaceutically acceptable salt thereof, calculated as the free base.
The pharmaceutically acceptable compounds of formula (I) and pharmaceutically acceptable salts thereof, can be administered in a daily dose (for an adult patient) of, for example, an oral or parenteral dose of 0.01 mg to 3000 mg per day or 0.5 to 1000 mg per day, or a nasal or inhaled dose of 0.001 to 50 mg per day or 0.01 to 5 mg per day, of the compound of the formula (I) or a pharmaceutically acceptable salt thereof, calculated as the free base. This amount may be given in a single dose per day or more usually in a number (such as two, three, four, five or six) of sub-doses per day such that the total daily dose is the same. An effective amount of a salt thereof, may be determined as a proportion of the effective amount of the compound of formula (I) per se.
The compounds of formula (I) and pharmaceutically acceptable salts thereof, and may be employed alone or in combination with other therapeutic agents. Combination therapies according to the present invention thus comprise the administration of at least one compound of formula (I) or a pharmaceutically acceptable salt thereof, and the use of at least one other pharmaceutically active agent. Preferably, combination therapies according to the present invention comprise the administration of at least one compound of formula (I) or a pharmaceutically acceptable salt thereof, and at least one other pharmaceutically active agent. The compound(s) of formula (I) and pharmaceutically acceptable salts thereof, and the other pharmaceutically active agent(s) may be administered together in a single pharmaceutical composition or separately and, when administered separately this may occur simultaneously or sequentially in any order. The amounts of the compound(s) of formula (I) and pharmaceutically acceptable salts thereof, and the other pharmaceutically active agent(s) and the relative timings of administration will be selected in order to achieve the desired combined therapeutic effect. Thus in a further aspect, there is provided a combination comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof, and at least one other pharmaceutically active agent.
Thus in one aspect, the compound of formula (I) or a pharmaceutically acceptable salt thereof, and pharmaceutical compositions comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof, according to the invention may be used in combination with or include one or more other therapeutic agents, for example selected from antibiotics, anti-virals, glucocorticosteroids, muscarinic antagonists and beta-2 agonists.
It will be appreciated that when the compound of formula (I) or a pharmaceutically acceptable salt thereof, is administered in combination with other therapeutic agents normally administered by the inhaled, intravenous, oral or intranasal route, that the resultant pharmaceutical composition may be administered by the same routes. Alternatively the individual components of the composition may be administered by different routes.
One embodiment of the invention encompasses combinations comprising one or two other therapeutic agents.
It will be clear to a person skilled in the art that, where appropriate, the other therapeutic ingredient(s) may be used in the form of salts, for example as alkali metal or amine salts or as acid addition salts, or prodrugs, or as esters, for example lower alkyl esters, or as solvates, for example hydrates, to optimise the activity and/or stability and/or physical characteristics, such as solubility, of the therapeutic ingredient. It will be clear also that, where appropriate, the therapeutic ingredients may be used in optically pure form.
The combinations referred to above may conveniently be presented for use in the form of a pharmaceutical composition and thus pharmaceutical compositions comprising a combination as defined above together with a pharmaceutically acceptable diluent or carrier represent a further aspect of the invention.
The compounds of formula (I) and pharmaceutically acceptable salts thereof, may be prepared by the methods described below or by similar methods. Thus the following Intermediates and Examples serve to illustrate the preparation of the compounds of formula (I) and pharmaceutically acceptable salts thereof, and are not to be considered as limiting the scope of the invention in any way. Intermediates and Examples
The following non-limiting Examples illustrate the present invention. Abbreviations
TLC - thin layer chromatography
AcOH - acetic acid
DCM - dichloromethane
DME - 1 ,2-dimethoxyethane
DMF - N,N-dimethylformamide
HOBT - 1 -(hydroxy)benzotriazole
EDCI - 1 -ethyl-3-(3-dimethylaminopropyl)carbodimide
diethyl ether - diethyl ether
EtOAc - ethyl acetate
/'-Pr20 - di-isopropyl ether
Config. - absolute configuration
CH3CN - acetonitrile
MeOH - methanol
THF - tetrahydrofuran
RT - room temperature
Rt - retention time
DIEA - Λ/,/V-diisopropylethylamine
APCI MS - Atmospheric Pressure Chemical Ionization quadrupole
Mass Spectrometer
CS2 - carbon disulfide Na2C03 - sodium carbonate
NaHC03 - sodium hydrogen carbonate
NaN02 - sodium nitrite
NaOH - sodium hydroxide
Na2S04 - sodium sulfate
POCI3 - Phosphorus (III) oxychloride
Ba(OH)2.8H20 - Barium hydroxide octahydrate
SnCI2,2H20 - Tin (II) chloride dihydrate
Pd/C - Palladium on carbon
mCPBA - m-chloroperbenzoic acid
CDCI3 - deuterated chloroform
DMSO d6 - deuterated dimethylsulfoxide
BOC " tert-butyloxycarbonyl
N - 1 Normal (concentration)
PEPPSI™ - Pyridine-Enhanced Precatalyst Preparation Stabilization
and Initiation
Analytical HPLC was conducted on two kinds of apparatus: a) On a Supelcosil LCABZ+PLUS column (3μηΊ, 3.3cm x 4.6mm ID) eluting with 0.1 % HCO2H and 0.01 M ammonium acetate in water (solvent A), and 95% acetonitrile and 0.05% HCO2H in water (solvent B), using the following elution gradient 0-0.7 minutes 0%B, 0.7-4.2 minutes 0→100%B, 4.2-5.3 minutes 100%B, 5.3-5.5 minutes 100→0%B at a flow rate of 3ml/minute. The mass spectra (MS) were recorded on a Fisons VG Platform mass spectrometer using electrospray positive ionisation [(ES+ve to give [M+H]+ and [M+NH4]+ molecular ions] or electrospray negative ionisation [(ES-ve to give [M-H]- molecular ion] modes. Analytical data from this apparatus are given with the following format : [M+H]+ or [M-H]". b) On a Chromolith Performance RP 18 column (100 x 4.6 mm id) eluting with 0.01 M ammonium acetate in water (solvent A) and 100% acetonitrile (solvent B), using the following elution gradient 0-4 minutes 0→100% B, 4-5 minutes 100% B at a flow rate of 5ml/minute. The mass spectra (MS) were recorded on a micromass Platform-LC mass spectrometer using atmospheric pressure chemical positive ionisation [AP+ve to give MH+ molecular ions] or atmospheric pressure chemical negative ionisation [AP-ve to give (M-H)" molecular ions] modes. Analytical data from this apparatus are given with the following format: [M+H]+ or [M-H]- preceded by the acronym APCI to specify between both mass spectrometry analyses sources.
LC/HRMS: Analytical HPLC was conducted on a Uptisphere-hsc column (3μηι 33 x 3 mm id) eluting with 0.01 M ammonium acetate in water (solvent A) and 100% acetonitrile (solvent B), using the following elution gradient 0-0.5 minutes 5% B, 0.5-3.75 minutes 5→100% B, 3.75-4.5 100% B, 4.5-5 100→5% B, 5-5.5 5% B at a flow rate of 1 .3ml/minute. The mass spectra (MS) were recorded on a micromass LCT mass spectrometer using electrospray positive ionisation [ES+ve to give MH+ molecular ions] or electrospray negative ionisation [ES-ve to give (M-H)- molecular ions] modes.
Mass directed auto-prep HPLC refers to the method where the material was purified by high performance liquid chromatography on a HPLCABZ+ 5μηι column (5cm x 10mm i.d.) with 0.1 % HCO2H in water and 95% MeCN, 5% water (0.5% HCO2H) utilising the following gradient elution conditions: 0-1 .0 minutes 5%B, 1.0-8.0 minutes 5→30%B, 8.0- 8.9 minutes 30%B, 8.9-9.0 minutes 30→95%B, 9.0-9.9 minutes 95%B, 9.9-10 minutes 95→0%B at a flow rate of 8ml/minute. The Gilson 202-fraction collector was triggered by a VG Platform Mass Spectrometer on detecting the mass of interest.
TLC (thin layer chromatography) refers to the use of TLC plates sold by Merck coated with silica gel 60 F254.
LC/MS Method
Analytical HPLC was conducted on a X-Terra MS C18 column (2.5μηι 30 x 3 mm id) eluting with 0.01 M ammonium acetate in water (solvent A) and 100% acetonitrile (solvent B), using the following elution gradient 0-4 minutes 0 to 100% B, 4-5 minutes 100% B at a flow rate of 1.1 ml/minute. The mass spectra (MS) were recorded on a micromass Platform-LC mass spectrometer using atmospheric pressure chemical positive ionisation [AP+ve to give MH+ molecular ions] or atmospheric pressure chemical negative ionisation [AP-ve to give (M-H)- molecular ions] modes.
LC/HRMS
Analytical HPLC was conducted on a Uptisphere-hsc column (3μηι 33 x 3 mm id) eluting with 0.01 M ammonium acetate in water (solvent A) and 100% acetonitrile (solvent B), using the following elution gradient 0-0.5 minutes 5% B, 0.5-3.75 minutes 5 to100% B, 3.75-4.5 100% B, 4.5-5 100 to 5% B, 5-5.5 5% B at a flow rate of 1 .3ml/minute. The mass spectra (MS) were recorded on a micromass LCT mass spectrometer using electrospray positive ionisation [ES+ve to give MH+ molecular ions] or electrospray negative ionisation [ES-ve to give (M-H)- molecular ions] modes. LCMS a) Method Formate
LC conditions
The UPLC analysis was conducted on an Acquity UPLC BEH C18 column (50mm x 2.1 mm, i.d. 1 .7μηι packing diameter) at 40°C.
The solvents employed were:
A = 0.1 % v/v solution of formic acid in water
B = 0.1 % v/v solution of formic acid in acetonitrile
The gradient employed was:
Figure imgf000040_0001
The UV detection was a summed signal from wavelength of 210nm to 350nm. MS conditions
MS Waters ZQ
lonisation mode Alternate-scan positive and negative electrospray
Scan range 100 to 1000 AMU
Scan time 0.27sec
Inter scan delay 0.10sec b) Method HpH
LC conditions
The UPLC analysis was conducted on an Acquity UPLC BEH C18 column (50mm x 2.1 mm, i.d. 1 .7μηι packing diameter) at 40°C.
The solvents employed were:
A = 10mM ammonium hydrogen carbonate in water adjusted to pH10 with ammonia solution
B = acetonitrile
The gradient employed was:
Figure imgf000041_0001
The UV detection was a summed signal from wavelength of 210nm to 350n
MS conditions
MS Waters ZQ
lonisation mode Alternate-scan positive and negative electrospray
Scan range 100 to 1000 AMU
Scan time 0.27sec
Inter scan delay 0.10sec
MDAP methodology Method Formate LC conditions
The HPLC analysis was conducted on either a Sunfire C18 column (100mm x 19mm, i.d 5μηι packing diameter) or a Sunfire C18 column (150mm x 30mm, i.d. 5μηι packing diameter) at ambient temperature.
The solvents employed were:
A = 0.1 % v/v solution of formic acid in water
B = 0.1 % v/v solution of formic acid in acetonitrile
Run as a gradient over either 15 or 25min (extended run) with a flow rate of 20ml/min (100mm x 19mm, i.d 5μηι packing diameter) or 40ml/min (150mm x 30mm, i.d. 5μηι packing diameter).
The UV detection was a summed signal from wavelength of 210nm to 350nm.
MS conditions
MS Waters ZQ
lonisation mode Alternate-scan positive and negative electrospray
Scan range 100 to 1000 AMU
Scan time 0.50sec
Inter scan delay 0.20sec
In the procedures that follow, after each starting material, reference to an Intermediate by number is typically provided. This is provided merely for assistance to the skilled chemist. The starting material may not necessarily have been prepared from the batch referred to.
Where reference is made to the use of a "similar" procedure, as will be appreciated by those skilled in the art, such a procedure may involve minor variation, for example reaction temperature, reagent/solvent amount, reaction time, work-up conditions or chromatographic purification conditions.
Intermediate 1 : 4-iodo-3,5-Dimethylisoxazole
Figure imgf000042_0001
Nitric acid (13ml) was added dropwise (exothermic reaction) to a mixture of 3,5- dimethylisoxazole (31.3g, 320mmol) and iodine (37.3g, 150 mmol) and the mixture was stirred at room temperature for 1 h. The reaction mixture was hydrolysed with a mixture of ice and water and extracted with DCM. The organic phase was washed with a solution of Na2S203, dried over Na2S04 and concentrated under reduced pressure to afford the title compound as a yellow solid (60g, 83%). [APCI MS] m/z: 224 MH+, Rt 2.17min.
Intermediate 2 : 3-(3,5-Dimethyl-4-isoxazolyl)aniline
Figure imgf000043_0001
To a solution of 4-iodo-3,5-dimethylisoxazole (for a preparation see intermediate 1 , 142g, 640mmol, 1 eq.) and (3-aminophenyl)boronic acid (100g, 640mmol, 1 eq.) in DME (600ml) were added tetrakis(triphenylphosphine) palladium(O) (18.5g , 16mol) and a solution of Na2C03 (203.5g,192mmol, 3eq.) in water (750ml). The mixture was heated under reflux for 24h. To complete the reaction intermediate 1 (0.2eq.) and tetrakis(triphenylphosphine) palladium(O) (5g) were added and the mixture was refluxed overnight. The cooled mixture was poured into water and extracted with DCM. The organic phase was washed with water, dried over Na2S04 and filtered. Evaporation of the solvent in vacuo gave a crude oil which was precipitated with iPr20 to afford the title compound as a beige solid (102g, 85%). [ES- MS] m/z: 189 MH+, Rt 2.20min. Intermediate 3 : 3,5-Dimethyl-4-[2-(methoxy)-5-nitrophenyl]isoxazole
Figure imgf000043_0002
To a solution of 2-iodo-1 -(methoxy)-4-nitrobenzene (2g, 7.17mmol, 1 eq.) and (3,5- dimethylisoxazole)boronic acid (3.03g, 21 .5 mmol, 3eq.) in DME (44ml) and water (7ml) were added tetrakis(triphenylphosphine) palladium(O) (0.415g, 0.05eq.) and Ba(OH)2.8H20 (4.52g, 14.33 mmol, 2eq.). The mixture was heated at 80°C for 16h. To complete the reaction (3,5-dimethylisoxazole)boronic acid (1 eq.) was added and the mixture was heated for 4h. The cooled mixture was filtered and extracted with DCM. The organic phase was washed with saturated aqueous Sodium hydrogen carbonate and water, dried over Na2S04 and filtered. Evaporation of the solvent in vacuo gave a crude oil which was precipitated with iPr20 to afford the title compound as a rust solid (1.735g, 97%). GC/MS m/z: 248
Intermediate 4: 3-(3,5-Dimethyl-4-isoxazolyl)-4-(methoxy)aniline
Figure imgf000044_0001
To a solution of 3,5-dimethyl-4-[2-(methoxy)-5-nitrophenyl]isoxazole (for a preparation see intermediate 3, 1 .7g, 6.85 mmol, 1 eq.) in ethanol (170ml), was added Pd/C (10% on carbon, 85mg) and the reaction was stirred under hydrogen for 4h. AcOH (1.7ml) was added and the reaction was hydrogenated for 20h. After filtration, the solvent was evaporated in vacuo. The crude compound was dissolved into DCM and washed with saturated aqueous Sodium hydrogen carbonate, dried over Na2S04, filtered and evaporated. The title compound was obtained as a red oil (1 .38g, 88%). GC/MS m/z: 218. Intermediate 5: Diethyl ({[3-(3,5-dimethyl-4-isoxazolyl)phenyl]amino}methylidene)- propanedioate
Figure imgf000044_0002
3-(3,5-Dimethyl-4-isoxazolyl)aniline (for a preparation see Intermediate 2, 80g, 420 mmol) and diethyl ethoxymethylenemalonate (92g, 425mmol) were mixed together and heated to 130°C for 20 minutes, liberating ethanol, which was evaporated under reduced pressure. The reaction mixture was poured into iPr20 (1 L) and the resulted precipitate filtered off and washed with iPr20. The resulting solid was recrystallised from acetonitrile to give the title compound as a brown solid (78g, 52%).
1H NMR (300 MHz, DMSO, ppm) δ : 10.8 (d, J=13.9Hz, 1 H), 8.50 (d, J=13.9Hz, 1 H), 7.62- 7.41 (m, 3H), 7.26 (d, J=7.7Hz, 1 H), 4.3 (q, J=7.2Hz,2H), 4.21 (q, J=7.2Hz,2H),2.51 (s, 3H), 2.34 (s,3H), 1.35 (t, J=7.2Hz, 3H), 1 .33 (s, J=7.2Hz,3H).
The following intermediate was prepared in an analogous manner to intermediate 5:
Figure imgf000045_0001
Figure imgf000045_0005
Intermediate 7: Ethyl 7-(3,5-dimethyl-4-isoxazolyl)-4-hydroxy-3-quinolinecarboxylate
Figure imgf000045_0002
Diethyl ({[3-(3,5-dimethyl-4-isoxazolyl)phenyl]amino}methylidene)-propanedioate (for a preparation see Intermediate 5, 75g, 210mmol) was suspended in boiling diphenylether (1 L) and heated to reflux for 30 minutes. The reaction mixture was cooled and precipitated with iPr20 to give the title compound as a brown powder (50g, 76%). 1H NMR (300 MHz, DMSO-d6, ppm) δ: 12.45 (brs, 1 H), 8.76 (s, 1 H), 8.34 (d, J=8.6Hz, 1 H), 7.76 (s, 1 H), 7.59 (d, J=8.6Hz, 1 H), 4.36 (q, J=7.1 Hz, 2H), 2.62 (s, 3H), 2.43 (s,3H), 1 .43 (s, J=7.1 Hz,3H).
The following intermediate was prepared in an analogous manner to intermediate 7:
Figure imgf000045_0003
Figure imgf000045_0004
Intermediate 9: 7-(3,5-dimethyl- -isoxazolyl)-4-hydroxy-3-quinolinecarboxylate
Figure imgf000046_0001
A suspension of ethyl 7-(3,5-dimethyl-4-isoxazolyl)-4-hydroxy-3-quinolinecarboxylate (for a preparation see Intermediate 7, 5.5g, 17.6mmol) in a aqueous solution of sodium hydroxide 1 N (80ml) was heated to reflux for 5h. The reaction mixture was treated with HCI 1 N and the resulting white precipitate was filtered and taken up with MeOH, concentratred to dryness to give the title compound as a yellow powder (4.27g, 85.3%). 1H NMR (300 MHz, DMSO, ppm) δ : 8.81 (brs, 1 H), 8.22 (d, J=8.5Hz, 1 H), 7.72 (brs, 1 H), 7.52 (dd, J=8.5, 1.7Hz, 1 H), 2.36 (s, 3H), 2.16 (s,3H).
The following intermediate was prepared in an analogous manner to intermediate 9:
Figure imgf000046_0002
Figure imgf000046_0003
Intermediate 11 : 7-(3,5-dimethyl-4-isoxazolyl)-4-quinolinol
Figure imgf000047_0001
7-(3,5-dimethyl-4-isoxazolyl)-4-hydroxy-3-quinolinecarboxylate (for a preparation see Intermediate 9, 10g, 35.21 mmol) was suspended by small portions to boiling diphenyl ether (200ml) and allowed to reflux for 2h. The reaction mixture was poured into hexane (500ml) at 0°C, the precipitate was filtered and washed several times with hexane to give the title compound as a white solid (6.8g, 86%). [APCI-MS] m/z: 240 MH+, Rt 1 .89min.1H NMR (300 MHz, CDCI3, ppm) δ : 12.44 (brs, 1 H), 8.67 (d, J=8.5Hz,1 H), 8.10 (d, J=7.2Hz, 1 H), 7.80 (s, 1 H), 7.62-7.55 (m, 1 H), 7.25 (d, J=8.5Hz, 1 H), 6.61 (d, J=7.4Hz, 1 H), 2.64 (s, 3H), 2.49 (s,3H).
Intermediate 12: 5-({[3-iodo-4-(methoxy)phenyl]amino}methylidene)-2,2-dimethyl- 1 ,3-dioxane-4,6-dione
Figure imgf000047_0002
A mixture of 2,2-dimethyl-1 ,3-dioxane-4,6-dione (203g, 1.4mol) and trimethoxymethane (1 -51) was heated to reflux for 1 h, then the 3-iodo-4-methoxy-aniline (349.2g, 1 .402mol) was added portionwise. The reaction mixture was stirred at reflux for 1 h, then cooled to room temperature. The resulting precipitate was filtered off, washed with diisopropyl ether and dried to give the title compound as a beige powder (485g, 85.9%)
1H NMR (300 MHz, DMSO-d6, ppm) δ : 1 1.2 (d, J=14.6Hz, 1 H), 8.50-8.39 (m,1 H), 8.05 (d, J=2.7Hz, 1 H), 7.60 (dd, J=8.9, 2.7Hz, 1 H), 7.05 (d, J=8.9Hz, 1 H), 3.84 (s, 3H), 1.67 (s,6H).
Intermediate 13: 6-iodo-7-(methoxy)-1 -quinolinol
Figure imgf000047_0003
5-({[3-iodo-4-(methoxy)phenyl]amino}methylidene)-2,2-dimethyl-1 ,3-dioxane-4,6-dione (for a preparation see Intermediate 12, 200g, 0.496mol) was added to diphenylether (2I) at 260°C. The reaction mixture was stirred at 260°C for 10 minutes. The black solution was then cooled at 100°C and poured into diisopropyl ether (8I) previously cooled at 0°C. The precipitate was filtered off, poured into cyclohexane (11) and then heated to reflux for 1 h. The solid was filtered off, poured into methanol (250ml) and heated at 45°C for 15 minutes. The solid was then filtered off and dried over pallets pump to give the title compound (105g, 70%).
1H NMR (300 MHz, DMSO-d6, ppm) δ 1H NMR (300 MHz, D6DMSO, ppm) δ : 8.07 (s, 1 H), 7.88 (d, J= 7.3Hz, 1 H), 7.45 (s, 1 H), 6.06 (d, J=7.3Hz, 1 H), 3.90 (s, 3H).
Intermediate 14 : 7-(3,5-dimethyl-4-isoxazolyl)-4-quinolinol
Figure imgf000048_0001
3,5-dimethylisoxazole-4-boronic acid (49.2g, 0.349mol) and Ba(OH)2.8H20 (91.8g, 0.291 mol, Acros) were added to a solution of intermediate 13 (35g, 0.1 16mol) in a mixture of water (180ml)) and 1 ,2-dimethoxyethane (600ml). The reaction was put under nitrogen for 15 minutes and tetrakis(triphenylphosphine)palladium (0) was added (4.1 g, 3.55mmol, Aldrich). The reaction mixture was stirred at 105°C overnight. After cooling at room temperature, the mixture was poured into water and extracted with ethyl acetate. The aqueous layer was acidified to pH 7 with concentrated HCI and extracted with ethyl acetate. The aqueous layer was basified to pH 10 with sodium hydroxide 5N and extracted with ethyl acetate. The organic layers were combined, washed with a saturated aqueous NaCI and dried. The crude brown oil was then purified by flash chromatography on silica gel eluting with DCM/MeOH (9:1 ) to give the title compound as a brown solid (31.4g, 43.9%)
1H NMR (300 MHz, DMSO-d6, ppm) δ : 1 1.72 (bs, 1 H), 7.88 (d, J=7.7Hz, 1 H), 7.61 (s, 1 H), 7.42 (s, 1 H), 6.03 (d, J=7.3Hz, 1 H), 3.86 (s, 3H), 2.31 (s, 1 H), 2.1 1 (s, 1 H).
Intermediate 15 : 7-(3,5-dimethyl-4-isoxazolyl)-3-nitro-4-quinolinol
Figure imgf000048_0002
To a solution of 7-(3,5-dimethyl-4-isoxazolyl)-4-quinolinol (for a preparation see Intermediate 1 1 , 8.5g, 35mmol) in propanoic acid (442ml) was added nitric acid (7ml) at room temperature, followed by heating the reaction mixture to 125°C for 2h. After cooling, the mixture was filtered, washed with isopropyl ether to give the title compound as a yellow solid (7.5g, 75%). [APCI-MS] m/z: 283 [M-H]", Rt 2.41 min.
1H NMR (300 MHz, DMSO-d6, ppm) δ : 13.01 (brs, 1 H), 9.28 (s, 1 H), 8.33 (d, J=8.3Hz 1 H), 7.73 (s, 1 H), 7.57 (d, J=8.3Hz, 1 H), 2.50 (s, 3H), 2.31 (s, 3H).
Intermediate 16 : 4-chloro-7-(3,5-dimethyl-4-isoxazolyl)-3-nitroquinoline
Figure imgf000049_0001
A suspension of 7-(3,5-dimethyl-4-isoxazolyl)-3-nitro-4-quinolinol (for a preparation see intermediate 15, 5g, 17mmol) in POCI3 (50ml) was refluxed overnight. After cooling the solvent was evaporated in vacuo. The resulting residue was poured over saturated aqueous Sodium hydrogen carbonate, extracted with DCM, washed with water, dried over Na2S04. The solvent was evaporated under reduce pressure to give the title compound as a light brown solid (4g, 75%). 1H NMR (300 MHz, CDCI3, ppm) δ : 9.23 (s, 1 H), 8.45 (d, J=8.9Hz, 1 H), 8.04 (d, J=1.5Hz, 1 H), 7.67 (dd, J=8.9, 1.5Hz, 1 H), 2.47 (s, 3H), 2.32 (s,3H).
Intermediate 17: 7-(3,5-dimethyl-4-isoxazolyl)-6-(methoxy)-3-nitro-4-quinolinol
Figure imgf000049_0002
Nitric acid (10ml) was added slowly to a solution of 7-(3,5-dimethyl-4-isoxazolyl)-4- quinolinol (for a preparation see Intermediate 14 (28g, 104mmol) in propanoic acid (450ml) at room temperature, followed by heating the reaction mixture to 100°C for 1 h. After cooling with an ice bath, the precipitate was filtered off, washed with pentane to give the title compound as a yellow powder (27g, 82%). [APCI-MS] m/z: 314 [M-H]", Rt 2.12min .1H NMR (300 MHz, DMSO-d6, ppm) δ : 13.06 (s, 1 H), 9.26 (s, 1 H), 7.84 (s, 1 H), 7.67 (s, 1 H), 3.98 (s, 3H), 2.39 (s, 3H), 2.19 (s,3H).
Intermediate 18: 4-chloro-7-(3,5-dimethyl-4-isoxazolyl)-6-(methoxy)-3-nitroquinoline
Figure imgf000050_0001
A suspension of 7-(3,5-dimethyl-4-isoxazolyl)-6-(methoxy)-3-nitro-4-quinolinol (for a preparation see Intermediate 17, 5g, 16mmol) in POCI3 (20ml) was refluxed overnight. After cooling, the mixture was evaporated to dryness. The resulting residue was poured over saturated aqueous Sodium hydrogen carbonate and extracted with DCM. The organic layer was washed with water, dried over Na2S04. The solvent was evaporated under reduce pressure to give the title compound as a light brown powder (5g, 94%). 1H
NMR (300 MHz, DMSO-d6, ppm) δ: 9.27 (s, 1 H), 8.15 (s, 1 H), 7.73 (s, 1 H), 4.05 (s, 3H), 2.36 (s, 3H), 2.16 (s,3H). yV-[2-(tert-butyl)phenyl]-7-(3,5-dimethyl-4-isoxazolyl)-3-nitro-4-
Figure imgf000050_0002
A mixture of 4-chloro-7-(3,5-dimethyl-4-isoxazolyl)-3-nitroquinoline (for a preparation see Intermediate 16) (2.2g, 7.2mmol) and 2-tert-butylaniline (1 .2g, 8mmol) in CH3CN (20ml) was refluxed for 1 h. The solvent was evaporated in vacuo and the residue treated with an aqueous solution of sodium hydroxide N, extracted with DCM, washed with water and dried over Na2S04. The residue was purified by chromatography on silica gel eluting with DCM/ MeOH (99:1 ) to give the title compound as a yellow solid (2.4g, 80%). [APCI-MS] m/z: 417 MH+, Rt 3.73min. NMR (300 MHz, DMSO-d6, ppm) δ : 9.87 (brs, 1 H), 8.92 (s, 1 H), 8.34 (d, J=8.7Hz, 1 H), 7.82(s, 1 H), 7.50( d, J=8.9Hz, 1 H), 7.25-6.91 (m, 4H), 2.32 (s, 3H), 2.15 (s, 3H). The following intermediates were prepared in an analogous manner to intermediate 19:
Figure imgf000051_0001
Figure imgf000051_0002
Intermediate 28 7-(3,5-dimethyl-4-isoxazolyl)-N4-(phenylmethyl)-3,4- quinolinediamine
Figure imgf000052_0001
To a solution of /V-[2-(tert-butyl)phenyl]-7-(3,5-dimethyl-4-isoxazolyl)-3-nitro-4- quinolinamine (see Intermediate 19, 2.4g, 5.77mmol) in a mixture of ethanol (60ml) and THF (20ml), was added SnCl2,2H20 (7.81 g, 34.6mmol). The reaction mixture was refluxed for 3h and then concentrated to dryness. The resulting residue was poured into aqueous sodium hydroxide. The organic phase was extracted with EtOAc, washed with water, dried and concentrated to give the title compound as a yellow solid (2g, 71 %). [APCI-MS] m/z: 387 MH+, Rt 3.37min. The following intermediates were prepared in an analogous manner to intermediate 28 from the corresponding nitro derivatives:
Figure imgf000052_0002
Figure imgf000053_0002
Intermediate 37: 7-^S^dimethyl^-isoxazoly -6-(methoxy) V4-^ -(2- pyridinyl)ethyl]-3,4-quinolinediamin
Figure imgf000053_0001
A mixture of 4-chloro-7-(3,5-dimethyl-4-isoxazolyl)-6-(methoxy)-3-nitroquinoline (for a preparation see Intermediate 18, 0.4g, 1.2mmol) and (1 /?)-1 -(2-pyridinyl)ethanamine (2eq, 0.293g) in CH3CN (20ml) was heated at 60°C for 2h. The mixture was extracted with DCM. The organic phase washed with saturated aqueous Sodium hydrogen carbonate and dried over Na2S04. The solvent was evaporated under reduced pressure and the residue taken up in diethyl ether. The precipitate was filtered off and dried in vacuo to give 7-(3,5-dimethyl-4-isoxazolyl)-6-(methoxy)-3-nitro-/V-[(1 R)-1 -(2-pyridinyl)ethyl]-4- quinolinamine (0.4g) which was used without purification in the next step.
To a solution of this intermediate (0.4g, 0.95mmol) in a mixture of ethanol (20ml) and HCI (3.8ml), was added SnCI2.2H20 (0.89g, 3.96mmol). The reaction mixture was heated to 40°C for 1 h, then hydrolysed with sodium hydroxide N and extracted with DCM. The organic phase was washed with water, dried and concentrated. The residue was purified by flash chromatography on silica gel (DCM/ MeOH, 95:5), the resulting compound was triturated in diisopropyl ether to give the title compound as a brown powder (0.25g, 53.5%). [APCI-MS] m/z: 390 MH+, Rt 2.62min. 1H NMR (300 MHz, DMSO-d6, ppm) δ : 8.54 (d, J=4.7Hz, 1 H), 8.30 (s, 1 H), 7.73 (dd, J= 7.8, 7.6 Hz, 1 H), 7.52 (s, 1 H), 7.52-7.48 (m, 1 H), 7.33 (s, 1 H), 7.27-7.21 (m, 1 H), 5.29-5.10 (m, 3H), 3.81 (s, 3H), 2.28 (s, 3H), 2.08 (s,3H), 1 .56 (d, J=6Hz, 3H) Intermediate 38: -iS.S-dimethyl^-isoxazolylJ-G-imethoxyJ-yS^^-pyridinylmethyl)- 3,4-quinolinediamine
Figure imgf000054_0001
A mixture of 4-chloro-7-(3,5-dimethyl-4-isoxazolyl)-6-(methoxy)-3-nitroquinoline (for a preparation see Intermediate 18, 2.5g, 7.5mmol) and 2-aminomethylpyridine (2eq, 1.41 g) in CH3CN (30ml) was heated at 60°C for 2h. The mixture was extracted with DCM. The organic phase washed with saturated aqueous sodium hydrogen carbonate and dried over Na2S04. The solvent was evaporated under reduced pressure and the residue taken up in diethyl ether. The precipitate was filtered off and dried in vacuo to give 7-(3,5- dimethyl-4-isoxazolyl)-6-(methoxy)-3-nitro-/V-(2-pyridinylmethyl)-4-quinolinamine (2.5g) which was used without purification in the next step.
To a solution of this nitro intermediate (2.5g, 24.82mmol) in a mixture of ethanol (20ml) and HCI (3.8ml), was added portionwise SnCI2.2H20 (5.6g, 24.82mmol). The reaction mixture was heated to 40°C for 1 h, then hydrolysed with sodium hydroxide N and extracted with DCM. The organic phase was washed with water, dried and concentrated to give the title compound as a brown powder (0.5g, 17.8%). (APCI-MS) m/z: 376 MH+, Rt 2.46min. 1H NMR (300 MHz, DMSO-d6, ppm) δ: 8.56 (d, 1 H), 8.29 (s, 1 H), 7.77 (dd, J= 7.8, 7.6 Hz, 1 H), 7.54 (s, 1 H), 7.53-7.51 (m, 1 H), 7.31 -7.25 (m, 1 H), 5.31 (s, 2H), 4.46 ( d, J=7.2Hz, 2H), 3.78 (s, 3H), 2.28 (s, 3H), 2.09 (s,3H).
Intermediate 39: -iS.S-dimethyl^-isoxazolylJ-G-imethoxyJ-yS^-^l -phenylethyl]- 3,4-quinolinediamine
Figure imgf000055_0001
A mixture of 4-chloro-7-(3,5-dimethyl-4-isoxazolyl)-6-(methoxy)-3-nitroquinoline (for a preparation see Intermediate 18, 2.5g, 7.5mmol) and (R)-(+)-alpha-methylbenzylamine (2eq, 1 .82g, Aldrich) in CH3CN (30ml) was heated at 60°C for 2h. The mixture was extracted with DCM. The organic phase washed with saturated aqueous sodium hydrogen carbonate and dried over Na2S04. The solvent was evaporated under reduced pressure and the residue taken up in diethyl ether. The precipitate was filtered off and dried under vacuum to give 7-(3,5-dimethyl-4-isoxazolyl)-6-(methoxy)-3-nitro-/V-[(1 /?)-1 -phenylethyl]-4- quinolinamine (2.5g) which was used without purification in the next step.
To a solution of this nitro intermediate (2.5g, 24.82mmol) in a mixture of ethanol (20ml) and HCI (3.8ml), was added portionwise SnCI2.2H20 (5.6g, 24.82mmol). The reaction mixture was heated to 40°C for 1 h, then hydrolysed with sodium hydroxide N and extracted with DCM. The organic phase was washed with water, dried and concentrated to give the title compound as a brown powder (0.5g, 17%). (APCI-MS) m/z: 389 MH+, Rt 2.95min.
Intermediate 40: 4-chloro-7-(3,5-dimethyl-4-isoxazolyl)-6-(methoxy)-3- quinolinecarboxamide
Figure imgf000055_0002
A mixture of intermediate 10 (27.7 g, 0.079 mol) and 10 drops of anhydrous DMF in POCI3 (250ml) was refluxed for 5h. Then, the mixture was concentrated to dryness under vacuum. The residue was treated twice with 100ml of toluene and evaporated to dryness to remove the last traces of POCI3. The dry foam obtained is added portionwise to an aqueous solution of ammonia (25%, 300ml) cooled to 0/5°C with an ice bath. After the end of the addition, the vigourous stirring was maintained for 1 h at this temperature. Then, the brown solid material was filtered off and washed respectively with water (3 x 200ml), diisopropyl ether (2 x 200ml) and pentane (100ml) to give after drying the crude product. This material was purified by a flash chromatography on silica gel (eluant = CH2CI2/MeOH, 95/5) to give the title compound (16.8 g, 64.7%).
1H NMR (300 MHz, CDCI3, ppm) δ : 8.96 (s, 1 H), 7.86 (s, 1 H), 7.54 (s, 1 H), 3.95(s, 3H), 2.30 (s, 3H), 2.15 (s,3H).
Intermediate 41 : Ethyl 7-(3,5-dimethyl-4-isoxazolyl)-4-chloro-3-quinolinecarboxylate
Figure imgf000056_0001
Ethyl 7-(3,5-dimethyl-4-isoxazolyl)-4-hydroxy-3-quinolinecarboxylate (for a preparation see Intermediate 7, 20g, 64.04mmol) was reacted with thionyl chloride (326ml) and heated to reflux overnight. Excess thionyl chloride was co-evaporated with toluene. The crude product was triturated with iPr20 and filtered to give the title compound as a brown powder (20.15g, 95%).
1H NMR (300 MHz, DMSO, ppm) δ: 9 (s, 1 H), 8.2 (d, 1 H), 8.0 (s, 1 H), 7.8 (d, 1 H), 4.25 (q, 2H), 2.2 (s, 3H), 2.1 (s, 3H), 1.2 (t,3H).
Intermediate 42: Ethyl 4-chloro-7-(3,5-dimethyl-4-isoxazolyl)-6-(methoxy)-3- quinolinecarboxylate
Figure imgf000056_0002
Intermediate 8 (5g, 14.62mmol) was reacted with phosphorus oxychloride (50ml) and heated to reflux overnight. Excess phosphorus oxychloride was evaporated with toluene. The crude product was washed with 1 N sodium hydroxide solution and extracted with DCM. The organic phase was dried over Na2S04 and evaporated under reduced pressure to give the title compound as a brown solid (5.1 g, 96.9%). APCI-MS: m/z 361 MH+, Rt =3.31 min
Intermediate 43: Ethyl 4-{[2-(1 -dimethylethyl)phenyl]amino}-7-(3,5-dimethyl-4- isoxazolyl)-6-(methoxy)-3-quinolinecarbox late
Figure imgf000057_0001
A mixture of ethyl 4-chloro-7-(3,5-dimethyl-4-isoxazolyl)-6-(methoxy)-3- quinolinecarboxylate (for a preparation see Intermediate 42 (3.3g, 9.2mmol) and 2-tert- butylaniline (2.7ml, 18.4mmol) in dioxane (50ml) was refluxed for 2 days. After cooling, the reaction mixture was evaporated under reduced pressure and the precipitate was purified by chromatography on silica gel eluting with DCM/MeOH (95/5) to give the title compound as a solid (1.2g, 27.7%).
HRMS Target Mass: 474.2393 MH+. Found: 474.2379; Rt 3.82min
The following intermediates were prepared in an analogous manner to intermediate 43
Figure imgf000057_0002
Figure imgf000057_0003
Figure imgf000058_0004
Intermediate 49 4-{[2-(1 ,1 -dimethylethyl)phenyl]amino}-7-(3,5-dimethyl-4- isoxazolyl)-6-(methoxy)-3-quin linecarboxylic acid hydrochloride
Figure imgf000058_0001
A mixture of ethyl 4-{[2-(1 ,1 -dimethylethyl)phenyl]amino}-7-(3,5-dimethyl-4-isoxazolyl)-6- (methoxy)-3-quinolinecarboxylate (for a preparation see Intermediate 43, 1 .2g, 2.54mmol) and 1 N sodium hydroxide (3ml) in Ethanol was heated to reflux for 1 day. The reaction mixture was concentrated, taken up in water and neutralised with 1 N HCI. The precipitate was filtered, and dried to give the title compound as a yellow powder (0.96g, 85%). LC/HRMS Target Mass calculated for C26H27N3O4: 446.2080 MH+. Found : 446.2038; Rt 2.39min
The following intermediates were prepared in an analogous manner to intermediate 49 from the corresponding ester derivatives:
Figure imgf000058_0002
Figure imgf000058_0003
Figure imgf000059_0002
Intermediate 55: 7-(3,5-dimethyl-4-isoxazolyl)-6-(methyloxy)-4-{[(1 R)-1 -(2- pyridinyl)ethyl]amino}-3-quinolinecarboxamide
Figure imgf000059_0001
4-Chloro-7-(3,5-dimethyl-4-isoxazolyl)-6-(methyloxy)-3-quinolinecarboxamide (for a preparation see Intermediate 56, 50g, 151 mmol,) and [(1 R)-1 -(2-pyridinyl)ethyl]amine (35.3g, 181 mmol, NetChem) were dissolved in N-methyl-2-pyrrolidone (NMP, 250ml). DIPEA (79ml, 452 mmol) was added, the solution was heated at 120°C overnight, cooled and diluted with ethyl acetate (11). The solution was washed with water (2 x 11), brine (500ml), dried (sodium sulphate) and the solvent evaporated to give a dark brown gum. The aqueous washings were extracted with DCM (2 x 600ml). The combined extracts were washed with a mixture of saturated brine (300ml) and water (11) giving a dense emulsion which took around 2h to separate. The organic layer was dried (sodium sulphate) and evaporated to leave a brown liquid. This liquid was dissolved in ethyl acetate (200ml), washed with water (2 x 200ml), dried (sodium sulphate) and evaporated to give a brown gum. This material was combined with the previous brown gum, dissolved in DCM (150ml) and loaded onto a silica column (750g), which was eluted with a 2M ammonia in methanol / DCM gradient (0-12%) to give, after evaporation of the solvents in vacuo 7-(3,5-dimethyl-4-isoxazolyl)-6-(methyloxy)-4-{[(1 R)-1 -(2- pyridinyl)ethyl]amino}-3-quinolinecarboxamide as beige foam (45.1 g). Used in the subsequent step (Example 36) without further purification.
1 H NMR CDCI3: δΗ 9.44(11-1, d), 8.71 (1 H, s), 8.62(1 H, d), 7.74(1 H, m), 7.68(1 H, s), 7.65(1 H, d), 7.34(11-1, s), 7.23(1 H, m), 6.06(2H, b), 5.36(1 H, m), 3.51 (3H, s), 2.32(3H, s), 2.17(3H, s), 1.73(31-1, d, partially obscured by water).
The mixed fractions from the column were collected and evaporated to give a brown gum. This was dissolved in DCM (20ml) loaded onto a silica column (330g) and eluted with an 2M ammonia in methanol / DCM gradient (0-10%) to give after evaporation of solvents 7- (3,5-dimethyl-4-isoxazolyl)-6-(methyloxy)-4-{[(1 R)-1 -(2-pyridinyl)ethyl]amino}-3- quinolinecarboxamide as beige gum (3.4g).
1 H NMR CDCI3: δΗ 9.44(11-1, d), 8.71 (11-1, s), 8.62(11-1, d), 7.74(11-1, m), 7.68(1 H, s), 7.65(1 H, d), 7.34(11-1, s), 7.23(1 H, m), 6.07(2H, b), 5.36(1 H, m), 3.51 (3H, s), 2.32(3H, s), 2.17(31-1, s), 1.73(31-1, d, partially obscured by water).
LCMS (Method HpH): MH+ 418, Rt 0.87min.
Intermediate 56: 4-Chloro-7-(3,5-dimethyl-4-isoxazolyl)-6-(methyloxy)-3- quinolinecarboxamide
Figure imgf000060_0001
7-(3,5-Dimethyl-4-isoxazolyl)-6-(methyloxy)-4-oxo-1 ,4-dihydro-3-quinolinecarboxylic acid (for a preparation see intermediate 57, 54g, 172 mmol) was heated in phosphorous oxychloride (80ml, 859 mmol) for 4h, then allowed to cool to room temperature and to stand overnight. The mixture was reduced to dryness in vacuo and the brown residue azeotroped with toluene (2 x 300ml). The resulting dark brown gum was dissolved in THF (300ml) with heating and sonication and the resulting solution added dropwise to ammonium hydroxide (33% w/w, 500ml) with ice bath cooling. The mixture was stirred for 30min, then concentrated to half volume, diluted with water (100ml) and the resulting dark brown solid collected by filtration. The solid was washed with water (100ml) and dried under vacuum at 50°C for 3 days to give 4-chloro-7-(3,5-dimethyl-4-isoxazolyl)-6- (methyloxy)-3-quinolinecarboxamide as brown solid (50.8g).
1 H NMR D6-DMSO: δΗ 8.75(1 H, s), 8.19(1 H, bs), 8.01 (1 H, s), 7.98(1 H, bs), 7.62(11-1, s), 4.00(31-1, s), 2.34(31-1, s), 2.14(3H, s).
LCMS (Method Formate): MH+ 332 / 334, Rt 0.76min. Intermediate 57: 7-(3,5-Dimethyl-4-isoxazolyl)-6-(methyloxy)-4-oxo-1 ,4-dihydro-3- quinolinecarboxylic acid
Figure imgf000061_0001
Ethyl 7-(3,5-dimethyl-4-isoxazolyl)-4-hydroxy-6-(methyloxy)-3-quinolinecarboxylate (for a preparation see Intermediate 58, 70.2g, 205 mmol,) was suspended in a mixture of ethanol (200ml) and sodium hydroxide (2M, 308ml, 615 mmol) and the mixture was heated at reflux overnight. The reaction was concentrated to ca 200ml in vacuo, diluted with water (500ml) and the resulting solution washed with ethyl acetate (200ml). The aqueous phase was acidified to pH4 with hydrochloric acid (1 M), the resulting suspension stirred vigorously for 10min. The solid was collected by filtration, washed with water (200ml) and dried overnight at 40°C in vacuo to give 7-(3,5-dimethyl-4-isoxazolyl)-4- hydroxy-6-(methyloxy)-3-quinolinecarboxylic acid as a beige solid (54.7g). Used without further purification.
1 H NMR D6-DMSO: δΗ 15.51 (1 H, s), 13.36(1 H, b), 8.88(1 H, s), 7.77(11-1, s), 7.71 (11-1, s), 3.94(31-1, s), 2.33(31-1, s), 2.13(3H, s).
LCMS (Method Formate): MH+ 315, Rt 0.80min.
Intermediate 58: Ethyl 7-(3,5-dimethyl-4-isoxazolyl)-4-hydroxy-6-(methyloxy)-3- quinolinecarboxylate
Figure imgf000061_0002
Diethyl({[3-(3,5-dimethyl-4-isoxazolyl)-4-(methyloxy)phenyl]amino}methylidene) propanedioate (for a preparation see Intermediate 59, 50g, 129 mmol) was added in small portions over 15min to boiling diphenyl ether (300ml) (temperature ca 255°C). The mixture was heated for a further 30min and the mixture was allowed to cool to 50°C. The reaction was diluted with cyclohexane (300ml) and cooled further, giving a thick tarry residue. The supernatant solvent was decanted and the residue was heated at reflux in ethyl acetate (300ml) for 20min, then diluted with cyclohexane (200ml) and cooled to room temperature. The solid residue was collected by filtration, washed with diethyl ether (200ml) and dried in vacuo at 40°C to give ethyl 7-(3,5-dimethyl-4-isoxazolyl)-4-hydroxy-6- (methyloxy)-3-quinolinecarboxylate as brown powder (28.1 g). Used without further purification in the subsequent reaction (Intermediate 57).
1 H NMR D6-DMSO: δΗ 12.28(1 H, s), 8.55(1 H, s), 7.69(1 H, s), 7.51 (1 H, s), 4.22(2H, q), 3.88(3H, s), 2.31 (3H, s), 2.1 1 (3H, s), 1 .29(3H, t).
Intermediate 59: Diethyl ({[3-(3,5-dimethyl-4-isoxazolyl)-4-
(methyloxy)phenyl]amino}meth lidene)propanedioate
Figure imgf000062_0001
[3-(3,5-Dimethyl-4-isoxazolyl)-4-(methyloxy)phenyl]amine (for a preparation see Intermediate 60, 27.4g, 126 mmol) was dissolved in diethyl [(ethyloxy)methylidene]propanedioate (27.1 g, 126 mmol) and heated to 130°C. The solution was heated for 1 h, then cooled to room temperature and reduced to dryness in vacuo to give diethyl ({[3-(3,5-dimethyl-4-isoxazolyl)-4- (methyloxy)phenyl]amino}methylidene)propanedioate (50.1 g), which after standing overnight became as a brown crystalline solid.
1 H NMR CDCI3: δΗ 1 1 .02(1 H, d), 8.44(11-1, d), 7.16(1 H, dd), 6.98(1 H, d), 6.91 (11-1, d), 4.34-4.22(41-1, m), 3.81 (3H, s), 2.32(3H, s), 2.17(3H, s), 1 .40-1 .31 (6H, m).
LCMS (Method Formate): MH+ 389, Rt 1 .17min.
Intermediate 60: [3-(3,5-Dimethyl-4-isoxazol l)-4-(methyloxy)phenyl]amine
Figure imgf000062_0002
3,5-Dimethyl-4-[2-(methyloxy)-5-nitrophenyl]isoxazole (for a preparation see Intermediate 61 , 68g, 274 mmol) was dissolved in ethyl acetate (11) and the solution washed with sodium sulphite solution (5%, 500ml). The organic layer was dried (sodium sulphate) and filtered through a 3cm pad of silica gel. The filtrate was diluted with ethanol (11) and added to Pd/C (E101 type NO/W, 10g) in a 5I nitrogen / vacuum purged hydrogenation flask under vacuum, The mixture was stirred for 24h. The mixture was purged with vacuum / nitrogen cycles (x 3), filtered through Celite under nitrogen and the filtrate evaporated in vacuo to give [3-(3,5-dimethyl-4-isoxazolyl)-4-(methyloxy)phenyl]amine 3-(3,5-dimethyl-4- isoxazolyl)-4-(methyloxy)aniline (62.3g).
1 H NMR CDCI3: δΗ 6.82(11-1, d), 6.71 (1 H, dd), 6.50(1 H, d), 3.71 (3H, s), 3.46(2H, b), 2.31 (3H, s), 2.18(3H, s).
LCMS (Method Formate): MH+ 219, Rt 0.48min.
Intermediate 61 : 3,5-Dimethyl-4-[2- methyloxy)-5-nitrophenyl]isoxazole
Figure imgf000063_0001
3-lodo-4-(methoxy) nitrobenzene (65g, 233 mmol, Matrix Scientific), (3,5-dimethyl-4- isoxazolyl)boronic acid (36.1 g, 256 mmol) and cesium carbonate (152g, 466 mmol) were combined with DME (80ml) and water (40ml) and the mixture was degassed with nitrogen for 10min, then PEPPSI™ catalyst (3.96g, 5.82 mmol) was added and the mixture heated at 90°C for 4h, then cooled and diluted with ethyl acetate (800ml). The resulting suspension was filtered through Celite and the filtrate washed with water (2 x 500ml). The solvent was dried (sodium sulphate), filtered and evaporated to give a brown solid which was triturated with ether (30ml). The solid product was washed with ether (100ml) to give 3,5-dimethyl-4-[2-(methyloxy)-5-nitrophenyl]isoxazole as beige solid (48.2g) .
1 H NMR CDCI3: δΗ 8.30(1 H, dd), 8.06(1 H, d), 7.07(1 H, d), 3.94(3H, s), 2.33(3H, s), 2.17(31-1, s). LCMS (Method Formate): MH+ 249, Rt 1.01 min.
Intermediate 62: 7-(3,5-Dimethyl-4-isoxazolyl)-6-(methyloxy)-4-{[1 -(1 -methyl-1 H- pyrazol-4-yl)ethyl]amino}-3- uinolinecarboxamide
Figure imgf000063_0002
4-Chloro-7-(3,5-dimethyl-4-isoxazolyl)-6-(methyloxy)-3-quinolinecarboxamide (for a preparation see Intermediate 56, 200mg, 0.603 mmol) and [1 -(1 -methyl-1 /-/-pyrazol-4- yl)ethyl]amine (1 13mg) were heated in a mixture of DIPEA (0.326ml, 1 .869 mmol) and N- Methyl-2-pyrrolidone (NMP) (15ml) at 120°C for 4h. The reaction mixture was cooled to room temperature, diluted with water (300ml) and extracted with ethyl acetate (2 x 200ml). The combined organics were washed with water (300ml) and brine (200ml), dried (sodium sulphate) and evaporated. The resulting gum was dissolved in DCM (3ml) loaded onto a loaded onto a silica cartridge (100g), which was then eluted with a 2M methanolic ammonia / DCM gradient (0-12%) to give, after evaporation of the product containing fractions in vacuo 7-(3,5-dimethyl-4-isoxazolyl)-6-(methyloxy)-4-{[1 -(1 -methyl-1 H-pyrazol- 4-yl)ethyl]amino}-3-quinolinecarboxamide (1 10mg).
LCMS (Method Formate): MH+ 421 , Rt 0.62min
Intermediate 63: 7-(3,5-dimethylisoxazol-4-yl)-6-methoxy-N4-(pyridin-2- ylmethyl)quinoline-3,4-diamine
A solution of 4-chloro-7-(3,5-dimethyl-4-isoxazolyl)-6-(methoxy)-3-nitroquinoline (for a preparation see intermediate 18, 2.5 g,) and (2-pyridinylmethyl)amine (1.41 g) in acetonitrile (30ml) were heated at 80°C for 2h. The mixture was extracted with DCM and the organic washed with a saturated solution of sodium hydrogen carbonate, dried over Na2S04, filtered and concentrated to dryness. The residue was taken-up in diethyl ether, filtered and dried under vacuum. The residue was dissolved in ethanol (20ml) and HCI (3.8ml, cone). Tin (II) chloride dihydrate (5.6 g) was then added in four portions and the reaction mixture was stirred for 1 h at 40°C. The mixture was hydrolysed using sodium hydroxide (1 N), extracted with DCM and the organic were dried over Na2S04. The mixture was filtered through Celite and concentrated to dryness. The crude product was purified by chromatography on silica gel eluting with DCM / methanol (56.5/3.5) to give 7- (3,5-dimethylisoxazol-4-yl)-6-methoxy-N4-(pyridin-2-ylmethyl)quinoline-3,4-diamine (500mg) as a clear brown foam. Intermediate 64: -iS.S-dimethyl^-isoxazoly -e-imethyloxyJ-yS^-KI -(2- pyridinyl)ethyl]-3,4-quinolinediamine
Figure imgf000065_0001
A solution of 4-chloro-7-(3,5-dimethyl-4-isoxazolyl)-6-(methoxy)-3-nitroquinoline (for a preparation see Intermediate 18, 400 mg,) and (R)-1 -(pyridin-2-yl)ethanamine (293 mg) in acetonitrile (30ml) was heated at 60°C for 2h. The reaction mixture was extracted with DCM and washed with a saturated solution of sodium hydrogen carbonate. The organic was dried over Na2S04, filtered and concentrated to dryness. The resulting compound was taken-up in ether, filtered and dried in vacuo. The residue was dissolved in ethanol (20ml) and HCI (3.8ml). Tin (II) chloride dihydrate (0.89 g) was then added in four portions and the reaction mixture was stirred for 1 h at 40°C. The reaction mixture was hydrolysed using a solution of sodium hydroxide (1 N). The mixture was extracted with DCM, the organic dried over Na2S04, filtered over Celite and concentrated to dryness. The crude product was purified via flash column chromatography on silica gel eluting with DCM / MeOH (95 / 5) and the obtained product was taken-up in diisopropylether to give 7-(3,5- dimethyl-4-isoxazolyl)-6-(methyloxy)-/V4-[(1 R)-1 -(2-pyridinyl)ethyl]-3,4-quinolinediamine (250mg) as a brown powder. LC/MS: Rt 2.62 min.
Intermediate 65: 7-(3,5-dimethyl-4-isoxazolyl)-4-{[(2,4-dimethyl-1 ,3-thiazol-5- yl)methyl]amino}-6-(methylox -3-quinolinecarboxamide
Figure imgf000065_0002
To a solution of 4-chloro-7-(3,5-dimethyl-4-isoxazolyl)-6-(methyloxy)-3- quinolinecarboxamide (for a preparation see Intermediate 56, 0.5g,) in acetonitrile (50ml) was added a solution of [(2,4-dimethyl-1 ,3-thiazol-5-yl)methyl]amine (0.64g, dihydrochloride available from Matrix Scientific) in acetonitrile (10ml). The reaction mixture was heated at reflux for 3h, whereupon another portion of amine (0.5g) was added. The reaction mixture was refluxed overnight and was then poured into water. The mixture phase was extracted with DCM and the organic dried over Na2S04 and concentrated to dryness to give a yellow sticky solid. The latter was recrystallised in acetonitrile to give 7- (3,5-dimethyl-4-isoxazolyl)-4-{[(2,4-dimethyl-1 ,3-thiazol-5-yl)methyl]amino}-6-(methyloxy)- 3-quinolinecarboxamide (0.33g) as white crystals.
LC/MS: MH+ 438.06, [M-H]" 436.13 Intermediate 66: 7-(3,5-dimethyl-4-isoxazolyl)-6-(methyloxy)-yS/ -[1 -(2-pyridinyl)ethyl]- 3,4-quinolinediamine
Figure imgf000066_0001
A solution of 4-chloro-7-(3,5-dimethyl-4-isoxazolyl)-6-(methoxy)-3-nitroquinoline (for a preparation see Intermediate 18, 1 g,) and (2-pyridinylmethyl)amine (810mg) in acetonitrile (30ml) was heated at 80°C for 2h. The mixture was extracted with DCM, washed with a saturated solution of sodium hydrogen carbonate, dried over Na2S04, filtered and concentrated to dryness. The crude compound was taken-up in diethyl ether, filtered and the fitrate dried under vacuum.
The solid was dissolved in ethanol (20ml) and hydrochloric acid (3.8ml, cone). Tin (II) chloride dehydrate (5.6g) was added in 4 portions and the reaction heated at 40°C for 1 h. The reaction was hydrolysed with sodium hydroxide (1 N) and extracted with DCM. The organic was dried over Na2S04, filtered through Celite and evaporated to dryness. The residue which was purified by chromatography on silica gel, eluting with DCM / methanol (95:5) to yield 7-(3,5-dimethyl-4-isoxazolyl)-6-(methyloxy)-/V4-[1 -(2-pyridinyl)ethyl]-3,4- quinolinediamine (600mg) as a orange oil.
Intermediate 67: 7-(3,5-dimethyl-4-isoxazolyl)-4-{[(5-methyl-3- isoxazolyl)methyl]amino}-6-(methyloxy)-3-quinolinecarboxamide
Figure imgf000067_0001
A solution of 4-chloro-7-(3,5-dimethyl-4-isoxazolyl)-6-(methyloxy)-3-quinolinecarboxamide (for a preparation see Intermediate 56, 331 mg) and [(5-methyl-3-isoxazolyl)methyl]amine (2.5 eq) in butanol (20ml) was heated to 1 10°C for 4h. The reaction mixture was evaporated to dryness and hydrolysed with water. The mixture was extracted with DCM and the organic dried over Na2S04, filtered and concentrated to dryness (280mg). Used without purification in the subsequent reaction (Example 52).
Intermediate 68: -iS.S-dimethyl^-isoxazolylJ-G-imethyloxyJ-yS^^-thienylmethyl)- 3,4-quinolinediamine
Figure imgf000067_0002
To a magnetically stirred solution 4-chloro-7-(3,5-dimethyl-4-isoxazolyl)-6-(methyloxy)-3- quinolinecarboxamide (for a preparation see Intermediate 56, 1.5g) in acetonitrile (10ml) in a Shlenk tube was added (2-thienylmethyl)amine (1 .53g). The Shlenk tube was sealed and the reaction mixture heated at 80°C for 24h. The water was added to the reaction followed by an aqueous saturated solution of sodium hydrogen carbonate. The mixture was extracted with DCM and the organic phase dried over Na2S04, filtered and concentrated to dryness. The resulting crude compound was purified by chromatography on silica gel (25g) eluting with DCM / methanol (95:5) to give 7-(3,5-dimethyl-4- isoxazolyl)-6-(methyloxy)-/V4-(2-thienylmethyl)-3,4-quinolinediamine (1 .493g) as a cream coloured powder. LC/MS: MH+ 409.12, [M-H]" 407.17, Rt 2.70min
Intermediate 69: 7-(3,5-dimethyl-4-isoxazolyl)-yV-[(2,4-dimethyl-1 ,3-thiazol-5- yl)methyl]-6-(methyloxy)-3-nitro-4-quinolinamine
Figure imgf000068_0001
To a solution of 4-chloro-7-(3,5-dimethyl-4-isoxazolyl)-6-(methyloxy)-3-nitroquinoline (for a preparation see Intermediate 18, 1 .5 g) in acetonitrile was added [(2,4-dimethyl-1 ,3- thiazol-5-yl)methyl]amine (1 .6 g) and the resulting mixture was stirred overnight at 80°C. Another portion of amine (0.64 g) was added and the mixture was stirred at 80°C for 6h. The reaction mixture was diluted with water, extracted with DCM and the organic phase were dried over Na2S04, and concentrated to dryness to give a dark red oil. The residue was purified by flash chromatography on silica gel, eluting with DCM / methanol (99:1 then 95:5) to give 7-(3,5-dimethyl-4-isoxazolyl)-/V-[(2,4-dimethyl-1 ,3-thiazol-5-yl)methyl]-6- (methyloxy)-3-nitro-4-quinolinamine (1 .13g) as an orange oil.
LC/MS: MH+ 440.05, [M-H]" 438.1 1
Intermediate 70: 7-(3,5-dimethyl-4-isoxazolyl)-yS/ -[(2,4-dimethyl-1 ,3-thiazol-5- l)methyl]-6-(methyloxy)-3,4-quinolinediamine
Figure imgf000068_0002
To a solution of 7-(3,5-dimethyl-4-isoxazolyl)-/V-[(2,4-dimethyl-1 ,3-thiazol-5-yl)methyl]-6- (methyloxy)-3-nitro-4-quinolinamine (for a preparation see intermediate 69, 1.13g) in ethanol was added hydrochloric acid (2ml, cone). Tin (II) chloride dihydrate (2.32 g) was added and the mixture was stirred at 40°C for 1 h. The reaction mixture was cooled, diluted with water and basified using sodium hydroxide solution (1 N). The formed precipitate was filtered through Celite and rinsed with DCM. The filtrate was washed with water and the organic was dried over Na2S04 and evaporated to dryness. The residue was purified by flash chromatography on silica gel eluting with DCM / methanol (98:2 then 93:7) to give 7-(3,5-dimethyl-4-isoxazolyl)-/V4-[(2,4-dimethyl-1 ,3-thiazol-5-yl)methyl]-6- (methyloxy)-3,4-quinolinediamine (0.6 g) as a beige foam.
LC/MS: MH+ 410.10, [M-H]" 408.18.
Intermediate 71 : yV-[7-(3,5-dimethyl-4-isoxazolyl)-4-{[(2,4-dimethyl-1 ,3-thiazol-5- yl)methyl]amino}-6-(methylox -3-quinolinyl]tetrahydro-2H^yran-4-carboxamide
Figure imgf000069_0001
To a solution of tetrahydro-2/-/-pyran-4-carboxylic acid (0.381 g, intermediate 69) in DCM (60ml) were successively added triethylamine (0.41 ml) then HATU (1 .1 15 g) and the resulting mixture was stirred for 20min room temperature. 7-(3,5-dimethyl-4-isoxazolyl)- /V4-[(2,4-dimethyl-1 ,3-thiazol-5-yl)methyl]-6-(methyloxy)-3,4-quinolinediamine (0.6g, intermediate 69) was added and the mixture stirred at ambient temperature overnight. The reaction mixture was poured into water and extracted with DCM. The organic phase was washed with a dilute sodium hydroxide solution and then with water. The organic phase was dried over Na2S04 and concentrated to dryness. The crude residue was purified by flash silica gel chromatography eluting DCM / methanol (99:1 then 95:5) to give the desired product as a beige sticky solid (0.71 g). The solid was triturated with hot diisopropylether to give /V-[7-(3,5-dimethyl-4-isoxazolyl)-4-{[(2,4-dimethyl-1 ,3-thiazol-5- yl)methyl]amino}-6-(methyloxy)-3-quinolinyl]tetrahydro-2/-/-pyran-4-carboxamide (0.6 g) as cream coloured crystals.
LC-HRMS: ES+ exact mass calculated for CZ7H32N5O4 S1 522.2175 MH+, found: 522.2225
Intermediate 72: 7-(3,5-dimethyl-4-isoxazolyl)-4-{[(5-methyl-2-furanyl)methyl]amino}- 6-(methyloxy)-3-quinolinecarboxamide
Figure imgf000070_0001
A mixture of 4-chloro-7-(3,5-dimethyl-4-isoxazolyl)-6-(methyloxy)-3-quinolinecarboxamide (for a preparation see Intermediate 56, 200mg,) and [(5-methyl-2-furanyl)methyl]amine (200mg) in 1 ,4-dioxane was heated to reflux overnight. The reaction mixture was evaporated to dryness and the residue was taken-up in DCM and washed with a saturated solution of sodium hydrogen carbonate. The organic was dried over Na2S04, filtered and concentrated. The residue was purified by chromatography to give 7-(3,5- dimethyl-4-isoxazolyl)-4-{[(5-methyl-2-furanyl)methyl]amino}-6-(methyloxy)-3- quinolinecarboxamide (177 mg) as a beige powder.
LC/MS: Rt 2.75, MH+ 406.99, [M-H]" 405.05
Example 1 : 1 -[2-(tert-butyl)phenyl]-7-(3,5-dimethyl-4-isoxazolyl)-1 ,3-dihydro-2H- imidazo[4,5-c]quinolin-2-one
Figure imgf000070_0002
A mixture of 7-(3,5-dimethyl-4-isoxazolyl)-/V4-(phenylmethyl)-3,4-quinolinediamine (for a preparation see Intermediate 28, 0.2g, 0.52mmol), Boc anhydride (2g, 9.17mmol) and triethyl amine (0.1 g) was heated at 80°C for 2h. Diphenylether (3g) was then added and the reaction mixture was heated to 180°C for 3 h. After cooling, the mixture was treated with aqueous sodium hydroxide and washed with DCM. The aqueous phase was acidified with acetic acid and extracted with DCM, washed with water, dried over Na2S04 and concentrated. The resulting residue was precipitated with a mixture of DCM/hexane to give the title compound (0.044g, 20%). 1H NMR (300 MHz, CDCI3, ppm) δ : 10.47 (brs, 1 H), 8.85 (s, 1 H), 7.93 (s, 1 H), 7.78 (d, J=8.3Hz, 1 H), 7.58 (dd, J=8.3 7.4Hz, 1 H), 7.39 (dd, J=7.7, 7.4Hz, 1 H), 7.17 (d, J=8.9Hz, 1 H), 7.05 (d, J=8.9Hz,1 H), 6.82 (d, J=8.7Hz,1 H), 2.37 (s, 3H), 2.24 (s,3H), 1.24 (s, 9H). (APCI-MS) m/z 413 MH+, Rt 2.92min. LC-HRMS ES+ exact mass calculated for C25H24N4O2413.1978 MH+, found: 413.2018, Rt 2.67min. The following examples were prepared in an analogous manner to Example 1 :
Figure imgf000071_0001
Figure imgf000072_0002
Figure imgf000072_0001
Figure imgf000073_0001
Figure imgf000074_0002
Example 10: 7-(3,5-dimethyl-4-isoxazolyl)-8-(methoxy)-1 -(2-pyridinylmethyl)-1 ,3- dihydro-2H imidazo[4,5-c]quinoli -2-one
Figure imgf000074_0001
The mixture of 4-chloro-7-(3,5-dimethyl-4-isoxazolyl)-6-(methoxy)-3-quinolinecarboxamide (for a preparation see Intermediate 40, 6g, 18mmol) was reacted with 2- aminomethylpyridine (2.5eq, 4.23g, 45mmol) in CH3CN (100ml) was stirred at 1 10°C for 4h. The reaction mixture was concentrated in vacuo. The residue was partitioned between water and DCM. The organic layer was dried over Na2S04 and concentrated to dryness to give 7-(3,5-dimethyl-4-isoxazolyl)-6-(methoxy)-4-[(2-pyridinylmethyl)amino]-3-quinoline carboxamide (5.84g) which was used in the next step without purification.
An excess of [bis(trifluoroacetoxy)iodo]benzene (19.35g, 45mmol) was added to a solution of the previous carboxamide intermediate (5.84g, 15mmol). The mixture was stirred at 50°C for 12h and then concentrated. The residue was partitioned between DCM and water, the organic layer was dried over Na2S04, filtered and concentrated in vacuo. The crude product was purified by flash chromatography on silica gel (DCM/ MeOH, 95:5), then triturated with diethylether to give the title compound as a beige powder (3.9g, 53.7%).
LC-HRMS ES+ exact mass calculated for C22H19N503: 402.1566 MH+. Found 402.1574, Rt = 2.14min. (APCI -MS) m/z 402.10 MH+, Rt 2.39min. Example 11 : 7-(3,5-dimethyl-4-isoxazolyl)-8-(methoxy)-1 -[(1 -(2-pyridinyl)ethyl]- 1 ,3-dihydro-2H-imidazo[4,5-c]quinolin-2-one
Figure imgf000075_0002
The mixture of 4-chloro-7-(3,5-dimethyl-4-isoxazolyl)-6-(methoxy)-3-quinolinecarboxamide (for a preparation see intermediate 40, 6g, 18mmol) was reacted with (1 /?)-1 -(2- pyridinyl)ethanamine (2eq., 4.43g, 36mmol) in CH3CN (100ml) and was stirred at 1 10°C for 4h. The reaction mixture was concentrated in vacuo. The residue was partitioned between water and DCM. The organic layer was dried over Na2S04 and concentrated to dryness to give 7-(3,5-dimethyl-4-isoxazolyl)-6-(methoxy)-4-{[(1 /?)-1 -(2-pyridinyl)ethyl]- amino}-3-quinolinecarboxamide which was used in the next step without purification. The carboxamide intermediate (6g) was treated with an excess of [bis(trifluoroacetoxy)iodo]benzene (19.35g, 45mmol) in CH3CN (100ml), the mixture was stirred at room temperature overnignt. The reaction mixture was concentrated in vacuo, the resulting residue was dissolved in DCM and washed with water. The crude product was purified by flash chromatography on silica gel (DCM/ MeOH, 95:5), the resulting compound was dissolved in DCM and precipitated from diethylether to give the title compound as an off-white powder (2.5g, 33%)
LC-HRMS ES+ exact mass calculated for C23H2iN503: 416.1722 MH+. Found 416.1736, Rt = 2.22min. (APCI-MS) m/z 415.97MH +, Rt 2.5min.
For an alternative preparation of this compound see Example 36.
Example 12: 7-(3,5-dimethyl-4-isoxazolyl)-8-(methoxy)-1 -{2- [(trifluoromethyl)oxy]phenyl} -1 ,3-dihydro-2H-imidazo[4,5-c]quinolin-2-one
Figure imgf000075_0001
A mixture of intermediate 50 (0.1 g, 0.2mmol) and diphenyl phosphoryl azide (0.065g, 0.24mmol) in DMF (10ml) was heated at 1 10°C for 2h. The reaction mixture was concentrated to dryness. The crude product was purified by flash chromatography on silica gel eluting with DCM/MeOH (98:2), the resulting compound was re-crystallised from diisopropyl ether to give the title compound as a yellow solid (0.02, %).
1H NMR (300 MHz, DMSO-d6, ppm) δ: 8.74 (s, 1 H), 7.99 (dd, J=1.5 ,7.7Hz ,1 H), 7.92 (s, 1 H), 7.89 (m, 2H), 7.84-7.77 (m, 1 H), 6.33 (s, 1 H), 3.37(s, 3H), 2.33 (s, 3H), 2.12 (s,3H). LC-HRMS Target Mass calculated for C23^3N404: 471.1280 MH+, Found : 471 .1250; Rt 2.47min.
The following examples were prepared in an analogous manner to example 12:
Figure imgf000076_0001
Figure imgf000076_0002
Figure imgf000077_0002
Example 18: 1 -[2-(tert-butyl)phenyl]-7-(3,5-dimethyl-4-isoxazolyl)-1 H-imidazo[4,5- c]quinoline
Figure imgf000077_0001
To a solution of 7-(3,5-dimethyl-4-isoxazolyl)-/V4-(phenylmethyl)-3,4-quinolinediamine_(for aq preparation see Intermediate 28, 0.2g, 0.52mmol) in DCM (10ml) were added HOBT (0.081 g, 0.62mmol), EDCI (0.1 18g, 0.62mmol), Et3N (0.062g, 0.62mmol) and formic acid (0.025g, 0.52mmol). The reaction mixture was stirred at room temperature overnight, then poured into water and extracted with DCM. The organic phase was washed with water, dried over Na2S04 and concentrated. The crude solid was re-crystallized from EtOAc to give the title compound as off-white crystals (0.1 g, 48%). M.p: 216°C. 1H NMR (300 MHz, CDCI3, ppm) δ : 9.36(s, 1 H), 8.06 (s, 1 H), 8.01 (s, 1 H), 7.73 (d, J=7.2Hz, 1 H), 7.56 (dd, J=8.1, 8.1Hz, 1H), 7.34 (dd, J=7.7, 7.7Hz, 1H), 7.15-7.06 (m,2H), 2.36 (s, 3H), 2.23 (s,3H), 1.07 (s, 9H). LC-HRMS: ES+ exact mass calculated for C25H24N40: 397.2028 MH+, found: 397.2021, Rt3.03min. The following examples were prepared in an analogous manner to example 18 from the corresponding intermediates:
Figure imgf000078_0001
Figure imgf000079_0001
Figure imgf000081_0002
Example 27: 7-(3,5-dimethyl-4-isoxazolyl)-8-(methoxy)-1 -(2-pyridinylmethyl)-2- (tetrahydro-2H-pyran-4-yl)-1 H-imi zo[4,5-c]quinoline
Figure imgf000081_0001
To a solution of 7-(3,5-dimethyl-4-isoxazolyl)-6-(methoxy)-/V4-(2-pyridinylmethyl)-3,4- quinolinediamine (for a preparation see Intermediate 38, 0.5g, 1.33mmol) in DCM (20ml) was added tetrahydro-2H-pyran-4-carbonyl chloride (1 .1 eq., 1.46mmol, 0.15g, Apollo Scientific) at 0°C, the mixture was stirred at 0°C for 30 minutes. The reaction mixture was hydrolysed with saturated aqueous Sodium hydrogen carbonate and extracted with DCM. The organic phase was dried over Na2S04 and concentrated to give the crude product which was used in the next step without purification. AcOH (5ml) was added to the crude product and the mixture was stirred at 100°C overnight. The reaction mixture was concentrated under reduce pressure, hydrolised with sodium hydroxide 1 N and extracted with DCM. The organic phase was dried over over Na2S04, evaporated under reduce pressure. The residue was purified by flash chromatography on silica gel (DCM/ MeOH, 95:5) and the compound was re-crystallised from acetonitrile to give the title compound as an off- white powder (0.13g, 21 % ) LC-HRMS: ES+ exact mass calculated for C27H27N5O3: 470.2192 (MH+). Found: 470.2153, Rt = 2.18min. (APCI-MS) m/z: 471 .01 MH+, Rt = 2.51 min
The following examples were prepared in an analogous manner to example
Figure imgf000082_0001
Figure imgf000082_0002
Figure imgf000083_0002
Example 31 : 7-(3,5-dimethyl-4-isoxazolyl)-8-(methoxy)-1 -[(1 -phenylethyl]-2- (tetrahydro-2H-pyran-4-yl)-1 H-imidazo 4,5-c]quinoline
Figure imgf000083_0001
To a solution of intermediate 39 (0.5g, 1.28mmol) in DCM (25ml) at 0°C was added tetrahydro-2H-pyran-4-carbonyl chloride (1 .05eq., 1.4mmol, 0.15g, Apollo Scientific), the mixture was then stirred at 0°C for 15 minutes. The reaction mixture was hydrolysed with saturated aqueous Sodium hydrogen carbonate and extracted with DCM. The organic phase was dried over Na2S04 and concentrated to give the crude product which was used in the next step without purification. AcOH was added to the crude product and the mixture was stirred at 100°C overnight. The reaction mixture was concentrated under reduce pressure, hydrolised with sodium hydroxide 1 N and extracted with DCM. The organic phase was dried over over Na2S04, evaporated under reduce pressure. The residue was purified by flash chromatography on silica gel (DCM/ MeOH, 95:5) to give the title compound as a white powder (0.16g, 26%).
(APCI-MS) m/z 483 MH+, Rt 2.93min. [a]D 20= -38.7° (c=0.8005g/100ml, CHCI3).
1H NMR (300 MHz, DMSO-d6, ppm) δ : 9.13 (s, 1 H), 7.90 (s, 1 H), 7.51 -7.21 (m, 5H), 6.73 (bs, 1 H), 6.51 (s, 1 H), 4.09-3.85 (m, 2H), 3.69-3.45 (m, 2H), 3.31 (s, 3H), 2.27 (s, 3H), 2.07 (d, J= 7Hz, 3H), 2.06 (s, 3H), 2.04-1.92 (m, 2H), 1 .91 -1.80 (m, 2H).
Example 32: 7-(3,5-dimethyl-4-isoxazolyl)-8-(methoxy)-1 -[(1 -phenylethyl]-1 H- [1 ,2,3]triazolo[4,5-c]quinoline
Figure imgf000084_0001
To a solution of /-(S.S-dimethyl^-isoxazoly -B-imethoxy^A^-til ^-l -phenylethy -S^- quinolinediamine (for a preparation see Intermediate 39, 0.2g, 0.515mmol) in DCM (10ml) cooled at 0°C, were added AcOH (0.5ml) and a solution of sodium nitrite ( 0.07g) in water (1 ml). The mixture was stirred at room temperature overnight. The reaction mixture was treated with saturated aqueous Sodium hydrogen carbonate and extracted with DCM. The organic phase was dried over Na2S04, evaporated under reduced pressure. The residue was purified by flash chromatography on silica gel (DCM/MeOH, 95:5). The solid was triturated in diisopropyl ether to give the title compound as a green powder (0.18g, 87.5%).
LC-HRMS: ES+ exact mass calculated for C23H21 N5O2: 400.1773 MH+. Found: 400.1785, Rt = 3.04min. (APCI-MS) m/z: 400 MH+, Rt 3.15min.
The following examples were prepared in an analogous manner to example 32:
Figure imgf000084_0002
Figure imgf000084_0003
Example 34: 7-(3,5-dimethyl-4-isoxazolyl)-1 -(benzyl)-1 ,3-dihydro-2H-imidazo[4,5- c]quinoline-2-thione
Figure imgf000085_0001
A mixture of intermediate 30 (0.5g, 1 .45 mmol), CS2 (1 .6ml) and triethylamine (0.3ml) in Ethanol (10ml) was stirred at 60°C overnight. The reaction mixture was hydrolysed with water and extracted with CH2CI2. The organic layer was dried over Na2S04 and concentrated. The crude solid was purified by chromatography on silica gel eluting with CH2CI2/MeOH (95/5) to give the title compound as a brown powder (0.1 g, 17.9%). 1H NMR (300 MHz, DMSO-d6, ppm) δ : 13.93 (s, 1 H), 9.01 (s, 1 H), 8.22 (d, J=8.7Hz, 1 H), 8.15 (d, J=1 .7Hz, 1 H), 7.66 (dd, J= 8.7, 1 .7Hz, 1 H), 7.48-7.27 (m, 5H), 6.17 (s, 2H), 2.52 (s, 3H), 2.34 (s,3H).
TOF MS ES+ exact Mass calculated for C22H18N4OS: 387.1280 MH+, Found: 387.1308; Rt 2.59min
The following example was prepared in an analogous manner to example 34:
Figure imgf000085_0002
Figure imgf000085_0003
The following examples were prepared from the appropriate intermediate compounds by a method analogous to those described earlier:
Example 36: 7-(3,5-dimethyl-4-isoxazolyl)-8-(methyloxy)-1 -[(1 R)-1 -(2-pyridinyl)ethyl]- 1 ,3-dihydro-2H-imidazo[4,5-c]q inolin-2-one
Figure imgf000086_0001
7-(3,5-Dimethyl-4-isoxazolyl)-6-(methyloxy)-4-{[(1 R)-1 -(2-pyridinyl)ethyl]amino}-3- quinolinecarboxamide (for a preparation see Intermediate 55, 45g, 102 mmol) was dissolved in methanol (500ml) and potassium hydroxide (7.47g, 133 mmol) was added. The mixture was stirred in an ice bath, then iodobenzene diacetate (39.6g, 123 mmol) was added in small portions over 20min and the mixture stirred for a further 1 h. The solvent was evaporated in vacuo, the residue diluted with water (11), and the resulting gummy suspension extracted with DCM (2 x 300ml). The solvent was dried (sodium sulphate) and loaded directly onto a silica column (750g), which was then eluted with a 2M ammonia in methanol / DCM gradient (0-10%). After evaporation of solvents in vacuo 7-(3,5-dimethyl-4-isoxazolyl)-8-(methyloxy)-1 -[(1 R)-1 -(2-pyridinyl)ethyl]-1 ,3-dihydro-2H- imidazo[4,5-c]quinolin-2-one (32.7g) was obtained as a beige solid.
1 H NMR CDCI3: δΗ 1 1.02(1 H, bs), 8.80(1 H, s), 8.70(1 H, d), 7.85(1 H, s), 7.67(1 H, m), 7.36(1 H, b), 7.29-7.26(1 H, m, partially obscured by chloroform), 6.84(1 H, b), 6.52(1 H, m), 3.55(3H, bs), 2.32(3H, s), 2.22(3H, d), 2.16(3H, s).
LCMS (Method HpH): MH+ 416, Rt 0.85min.
Example 37: 7-(3,5-dimethyl-4-isoxazolyl)-8-(methyloxy)-3-[2-(methyloxy)ethyl]-1 - [(1 R)-1 -(2-pyridinyl)ethyl]-1 ,3-dih dro-2H-imidazo[4,5-c]quinolin-2-one
Figure imgf000086_0002
7-(3,5-Dimethyl-4-isoxazolyl)-8-(methyloxy)-1 -[(1 R)-1 -(2-pyridinyl)ethyl]-1 ,3-dihydro-2H- imidazo[4,5-c]quinolin-2-one (for a preparation see Example 36, 100mg) in DMF (5ml) was treated with 2-tert-butylimino-2-diethylamino-1 ,3-dimethylperhydro-1 ,3,2- diazaphosphorine, polymer bound (300mg) and a solution of 2-bromoethyl methyl ether (40mg) in DMF (0.5ml). The reaction was stirred at ambient temperature, air atm for ~3h and then left at ambient temperature overnight. The reaction was filtered and the resin washed with methanol. The solvents were evaporated under a stream of nitrogen to leave a yellow gum which was dissolved in DMSO (1 ml) and purified by MDAP (Method formate). The product fractions were combined and reduced to dryness in vacuo to give 7-(3,5-dimethyl-4-isoxazolyl)-8-(methyloxy)-3-[2-(methyloxy)ethyl]-1 -[(1 R)-1 -(2- pyridinyl)ethyl]-1 ,3-dihydro-2H-imidazo[4,5-c]quinolin-2-one as a pale yellow gum (56mg) LCMS (Method Formate): MH+ 474, Rt 0.80min. Example 38: 7-(3,5-dimethyl-4-isoxazolyl)-8-(methyloxy)-1 -[1 -(2-pyridinyl)ethyl]-1 ,3- dihydro-2H-imidazo[4,5-c]quinol -2-one
Figure imgf000087_0001
7- (3,5-Dimethyl-4-isoxazolyl)-6-(methyloxy)-4-{[1 -(2-pyridinyl)ethyl]amino}-3- quinolinecarboxamide (0.39g, 0.794 mmol) was dissolved in methanol (10ml) and potassium hydroxide (0.089g, 1 .588 mmol) was added, the mixture was stirred in an ice- bath, bis(acetyloxy)(phenyl)-l3-iodane (0.307g, 0.953 mmol) was added in small portions over 20min and the mixture stirred for a further 1 h. The mixture was acidified with acetic acid (0.2ml), evaporated in vacuo and the residue partitioned between ethyl acetate (20ml) and water (20ml). The solvent was dried (sodium sulphate) and evaporated and the residue dissolved in DCM (5ml), applied to a silica cartridge (50g), which was eluted with a methanol / ethyl acetate gradient (0-20%). Appropriate fractions were reduced to dryness in vacuo to give a beige solid. This solid was triturated with hot ethyl acetate (5ml) and cooled and the solid collected by filtration to give 7-(3,5-dimethyl-4-isoxazolyl)-
8- (methyloxy)-1 -[1 -(2-pyridinyl)ethyl]-1 ,3-dihydro-2H-imidazo[4,5-c]quinolin-2-one as pale cream powder (0.20g). 1 H NMR CDCI3: δΗ 8.82(1 H, s), 8.70(1 H, d), 7.87(1 H, s), 7.67(1 H, m), 7.36(1 H, b), 7.29- 7.26(1 H, m, partially obscured by chloroform), 6.87(1 H, b), 6.51 (1 H, m), 3.56(3H, s), 2.32(3H, s), 2.22(3H, d), 2.16(3H, s).
LCMS (Method Formate): MH+ 416, Rt 0.69min.
Example 39: 7-(3,5-dimethyl-4-isoxazolyl)-8-(methyloxy)-1 -[(1 S)-1 -(2-pyridinyl)ethyl]- 1 ,3-dihydro-2H-imidazo[4,5-c]quinolin-2-one
Figure imgf000088_0001
7-(3,5-Dimethyl-4-isoxazolyl)-8-(methyloxy)-1 -[1 -(2-py rid iny I )ethyl]-1 ,3-dihydro-2H- imidazo[4,5-c]quinolin-2-one (for a preparation see Example 38 , ~ 50mg) was dissolved in 3ml of ethanol (3ml) and heptanes (3ml) with warming (x3). The solutions were purified by chiral HPLC using 6x injection (3ml) of the above solution onto a Chiralpak IA column (20um, 5cm x 25cm) and elution with ethanol / heptanes (30%, flow rate 50ml/min, wavelength 215nm). The mixed fractions were reduced to dryness in vacuo and reprocessed as described in the method above. The bulked fractions of the first eluting enantiomer were reduced to dryness in vacuo to give 7-(3,5-dimethyl-4-isoxazolyl)-8- (methyloxy)-l -[(1 S)-1 -(2-pyridinyl)ethyl]-1 ,3-dihydro-2H-imidazo[4,5-c]quinolin-2-one
(61 mg).
1 H NMR CDCI3: δΗ 8.86(1 H, s), 8.68(1 H, d), 7.91 (1 H, s), 7.67(1 H, m), 7.37(1 H, b), 7.29- 7.26(1 H, m, partially obscured by chloroform), 6.90(1 H, b), 6.49(1 H, m), 3.58(3H, s), 2.32(3H, s), 2.20(3H, d), 2.16(3H, s).
LCMS (Method Formate): MH+ 416, Rt 0.71 min
Example 40: 7-(3,5-dimethyl-4-isoxazolyl)-8-(methyloxy)-1 -[1 -(1 -methyl-1 H-pyrazol- 4-yl)ethyl]-1 ,3-dihydro-2H-imidazo 4,5-c]quinolin-2-one (
Figure imgf000088_0002
7-(3,5-Dimethyl-4-isoxazolyl)-6-(methyloxy)-4-{[1 -(1 -methyl-1 H-pyrazol-4-yl)ethyl]amino}- 3-quinolinecarboxamide (1 10mg, 0.262 mmol, intermediate 61 ) was dissolved in methanol (10ml) and cooled to 0°C in an ice bath, potassium hydroxide (19mg, 0.340 mmol) was added and the solution stirred for 10min. lodobenzene diacetate (1 10mg, 0.340 mmol) was added and the mixture stirred for a further 2h. The solution was diluted with ethyl acetate (50ml), washed with water, dried (sodium sulphate) and evaporated. The residue was and the residue was dissolved in DCM (4ml), applied to a silica cartridge (25g) and the cartridge eluted with a 2M ammonia in methanol / DCM gradient (0-10%) to give, after evaporation in vacuo of the product-containing fractions, 7-(3,5-dimethyl-4- isoxazolyl)-8-(methyloxy)-1 -[1 -(1 -methyl-1 H-pyrazol-4-yl)ethyl]-1 ,3-dihydro-2H- imidazo[4,5-c]quinolin-2-one as beige solid (46mg).
LCMS (Method Formate): MH+ 419, Rt 0.62min
Example 41 : 7-(3,5-dimethyl-4-isoxazolyl)-8-(methyloxy)-1 -(2-pyridinylmethyl)-2- (tetrahydro-2H-pyran-4-yl)-1 H-imidazo[4,5-c]quinoline
Figure imgf000089_0001
To a solution of 4-chloro-7-(3,5-dimethyl-4-isoxazolyl)-6-(methoxy)-3-nitroquinoline (for a preparation see intermediate 18, 7 g) in acetonitrile (100ml) was added (2- pyridinylmethyl)amine (6.8 g) and the resulting mixture was heated at 60°C for 1 h. The reaction mixture was then hydrolysed, extracted with DCM and the combined organic phases were dried (Na2S04), filtered and evaporated to dryness. A portion of this material (2 g) was dissolved in Ethanol (80ml) and HCI (1 ml, cone). Tin (II) chloride dihydrate (1 g) was then added in two portions and the reaction mixture was stirred for 2h at room temperature. The reaction mixture was hydrolysed using a saturated solution of sodium hydrogen carbonate. The m was extracted with DCM and the combined organic layers were filtered over Celite. The filtrate was dried (Na2S04), filtered and concentrated to dryness to give, after chromatography, 3 g of material. The latter was dissolved in DCM (100ml) and to this mixture were added HATU (3.65 g) and tetrahydro-2H-pyran-4- carboxylic acid (1 .25 g). The resulting reaction mixture was stirred overnight at room temperature and hydrolysed by adding a solution of sodium hydrogen carbonate. The reaction mixture was extracted with DCM, dried (Na2S04), filtered and concentrated to dryness to give the crude intermediate (2.9g). This was dissolved on acetic acid (30ml) and the resulting mixture was heated to
90°C for 6h. The reaction mixture was concentrated to dryness. A saturated aqueous solution of Sodium hydrogen carbonate was added and the mixture was extracted with DCM, dried (Na2S04), filtered and concentrated to dryness. The crude compound was purified by chromatography on silica gel, eluting with DCM / methanol (95:5), then recrystallised from ethanol and was dried under vacuum to give 7-(3,5-dimethyl-4- isoxazolyl)-8-(methyloxy)-1 -(2-pyridinylmethyl)-2-(tetrahydro-2/-/-pyran-4-yl)-1 /-/- imidazo[4,5-c]quinoline as a beige powder (1.88 g).
LC-HRMS: ES+ exact mass calculated for CZTHZSNSOS 470.2192 MH+ , found: 470.21 15, Rt 2.25min.
Example 43: 4-(8-methoxy-2-(methoxymethyl)-1 -(pyridin-2-ylmethyl)-1 H
c]quinolin-7-yl)-3,5-dimethylisoxazole
Figure imgf000090_0001
7-(3,5-dimethylisoxazol-4-yl)-6-methoxy-N4-(pyridin-2-ylmethyl)quinoline-3,4-diamine (for a preparation see Intermediate 63, 500mg) was dissolved in DCM (20ml) and the reaction mixture was cooled to 0°C. 2-Methoxyacetyl chloride (1.1 eq) was added dropwise and the reaction mixture was stirred at room temperature for 30 min. The mixture was hydrolysed using a solution of sodium hydrogen carbonate and extracted with DCM. The organic was dried over Na2S04, filtered and concentrated to dryness. The obtained residue was dissolved in acetic acid (5ml) and the reaction mixture was heated to 130°C for 16h. The reaction mixture was treated with sodium hydroxide (1 M) and extracted with DCM. The organic was dried over Na2S04, filtered and concentrated to dryness. The residue was purified by chromatography on silica gel, eluting with DCM / methanol (95:5). The resulting compound was recrystallised from acetonitrile and dried under vacuum to give 4-(8-methoxy-2-(methoxymethyl)-1 -(pyridin-2-ylmethyl)-1 H-imidazo[4,5-c]quinolin-7- yl)-3,5-dimethylisoxazole (50mg) as a cream-coloured powder.
LC-HRMS: ES+ exact mass calculated for C22H19N503 430.1879 MH+ , found: 430.1833, Rt 2.23 min. Example 44: 7-(3,5-dimethyl-4-isoxazolyl)-8-(methyloxy)-1 -[1 -(2-pyridinyl)ethyl]-2- (tetrahydro-2H-pyran-4-yl)-1 H-imidazo[4,5-c]quinoline
Figure imgf000091_0001
To a solution of 7-(3,5-dimethyl-4-isoxazolyl)-6-(methyloxy)-/V4-^
3,4-quinolinediamine (for a preparation see Intermediate 64, 380 mg) in DCM (20ml) was added tetrahydro-2H-pyran-4-carboxylic acid (240 mg) followed by HATU (1 .2 equiv) and triethylamine (1 .5 equiv). The reaction mixture was stirred at room temperature for 1 h, and then hydrolysed using a saturated solution of sodium hydrogen carbonate. The mixture was extracted with DCM and the organic was dried over Na2S04, filtered and concentrated to dryness. The resulting crude product was diluted in acetic acid (5ml) and heated at 100°C overnight. The reaction mixture was then taken-up in a 1 :1 mixture of sodium hydroxide (1 N) and water (total 100ml), extracted with DCM and the resulting crude product was purified by chromatography on silica gel eluting with DCM / methanol (95:5). The resulting product was taken-up in diethyl ether and dried under vacuum to give racemic 7-(3,5-dimethyl-4-isoxazolyl)-8-(methyloxy)-1 -[1 -(2-pyridinyl)ethyl]-2-(tetrahydro- 2H-pyran-4-yl)-1 H-imidazo[4,5-c]quinoline (400 mg) as a grey powder.
LC-HRMS: ES+ exact mass calculated for C28H30N5O3 484.2349 MH+ , found: 484.2382, Rt 2.30 min. Example 45: 7-(3,5-dimethyl-4-isoxazolyl)-8-(methyloxy)-1 -[(1 ?)-1 -(2-pyridinyl)ethyl]- 2-(tetrahydro-2H-pyran-4-yl)-1 H-imidazo[4,5-c]quinoline
and
Example 46: 7-(3,5-dimethyl-4-isoxazolyl)-8-(methyloxy)-1 -[(1 S)-1 -(2-pyridinyl)ethyl]- 2-(tetrahydro-2H-pyran-4-yl)-1 H-imidazo[4,5-c]quinoline
Figure imgf000091_0002
The racemic mixture of 7-(3,5-dimethyl-4-isoxazolyl)-8-(methyloxy)-1 -[1 -(2-pyridinyl)ethyl]- 2-(tetrahydro-2/-/-pyran-4-yl)-1 /-/-imidazo[4,5-c]quinoline (400 mg) was separated using a semi-preparative column: Chiracel OD, 250 x 20 nm . 10 μηη, eluting with hexane / ethanol (85:15) [Flow rate: 18ml/min; rt; UV detection: 254 nm; Injection volume 900 μΙ_] to give:
-Isomer 1 : 7-(3,5-dimethyl-4-isoxazolyl)-8-(methyloxy)-1 -[(1 ?*)-1 -(2-pyridinyl)ethyl]- 2-(tetrahydro-2H-pyran-4-yl)-1 H-imidazo[4,5-c]quinoline (55 mg) as a white powder. LC-HRMS: ES+ exact mass calculated for C28H3oN503 484.2349 MH+, found: 484.2393, Rt 2.46 min. Analytical HPLC (Chiracel OD column, 250 x 4.6 nm . 10 μηι, eluting with hexane / ethanol (60/40): Rt 7.868
-Isomer 2: 7-(3,5-dimethyl-4-isoxazolyl)-8-(methyloxy)-1 -[(1 S*)-1 -(2-pyridinyl)ethyl]- 2-(tetrahydro-2H-pyran-4-yl)-1 H-imidazo[4,5-c]quinoline (55 mg) as a white powder. LC-HRMS ES+ exact mass calculated for C28H30N5O3 484.2349 MH+, found: 484.2394, Rt 2.43 min. Analytical HPLC (Chiracel OD column, 250 x 4.6 nm . 10 μηι, eluting with hexane / ethanol (60:40): Rt 9.377
Example 47: 7-(3,5-dimethyl-4-isoxazolyl)-8-(methyloxy)-1 -(2-pyrimidinylmethyl)-1 ,3- dihydro-2H-imidazo[4,5-c]quinolin-2-one
Figure imgf000092_0001
In a 100ml flask a mixture of 4-chloro-7-(3,5-dimethyl-4-isoxazolyl)-6-(methyloxy)-3- quinolinecarboxamide (for a preparation see Intermediate 56, 331 mg) and (2- pyrimidinylmethyl)amine (273mg) in acetonitrile was heated at 100°C for 2h. The reaction mixture was hydrolyzed with a solution of sodium hydrogen carbonate, extracted with DCM and the organic dried over Na2S04, filtered and concentrated to dryness to give 350mg of crude intermediate. The residue was dissolved in acetonitrile (15ml), bis(trifluoroacetoxy)iodo]benzene (0.5g) was added and the reaction mixture was stirred for 3h at room temperature. The reaction mixture was washed with water, extracted with DCM and the organic dried over Na2S04, filtered and concentrated to dryness. The resulting crude compound was purified by chromatography on silica gel eluting with DCM / methanol (95:5) and the resulting residue was recrystallised from acetonitrile and dried in vacuo to give 7-(3,5-dimethyl-4-isoxazolyl)-8-(methyloxy)-1 -(2-pyrimidinylmethyl)-1 ,3- dihydro-2H-imidazo[4,5-c]quinolin-2-one (70mg) as a beige powder.
LC-HRMS: ES+ exact mass calculated for C2iH19N603 403.1519 MH+, found: 403.1516, Rt 2.04min.
Example 48: 7-(3,5-dimethyl-4-isoxazolyl)-8-(methyloxy)-1 -(2-pyrazinylmethyl)-1 ,3- dihydro-2H-imidazo[4,5-c]quinolin-2-one
Figure imgf000093_0001
In a 100ml flask a mixture of 4-chloro-7-(3,5-dimethyl-4-isoxazolyl)-6-(methyloxy)-3- quinolinecarboxamide (for a preparation see Intermediate 56, 331 mg,) and (2- pyrazinylmethyl)amine (2.5 eq) in acetonitrile was heated at 100°C for 2h. The reaction mixture was hydrolyzed with a solution of sodium hydrogen carbonate, extracted with DCM and the organic dried over Na2S04, filtered and concentrated to dryness to give the crude intermediate. The residue was dissolved in acetonitrile (15ml), bis(trifluoroacetoxy)iodo]benzene (0.5g) was added and the reaction mixture was stirred for 3h at room temperature. The reaction mixture was washed with water, extracted with DCM and the organic were dried over Na2S04, filtered and concentrated to dryness. The resulting crude compound was purified by chromatography on silica gel eluting with DCM / methanol (95:5) and the resulting residue was recrystallised from acetonitrile and dried in vacuo to give 7-(3,5-dimethyl-4-isoxazolyl)-8-(methyloxy)-1 -(2-pyrimidinylmethyl)-1 ,3- dihydro-2H-imidazo[4,5-c]quinolin-2-one (100mg) as a beige powder.
LC-HRMS: ES+ exact mass calculated for C2i H19N603 403.1519 MH+, found: 403.1550, Rt 2.06min.
Example 49: 7-(3,5-dimethyl-4-isoxazolyl)-8-(methyloxy)-1 -(3-pyridinylmethyl)-1 ,3- dihydro-2H-imidazo[4,5-c]quinolin-2-one
Figure imgf000094_0001
A solution of 4-chloro-7-(3,5-dimethyl-4-isoxazolyl)-6-(methyloxy)-3-quinolinecarboxamide (for a preparation see intermediate 56, 332mg,) and (3-pyridinylmethyl)amine (2.5 equiv in acetonitrile was heated for 4h. The reaction mixture was then concentrated to dryness and hydrolyzed with water. The mixture extracted with DCM and the organic was dried over Na2S04, filtered and concentrated to dryness to give the crude intermediate. The residue was partially dissolved in acetonitrile (30ml) and bis(trifluoroacetoxy)iodo]benzene (800 mg) was added. The resulting mixture was stirred at room temperature for 24h. The reaction mixture was then concentrated to dryness and hydrolysed with water. The mixture was extracted with DCM and the organic dried over Na2S04, filtered and concentrated to dryness. The residue was purified by chromatography on silica gel, eluting with DCM / methanol (95 / 5). The purified residue was taken-up in diethyl ether to give 7-(3,5-dimethyl-4-isoxazolyl)-8-(methyloxy)-1 -(3-pyridinylmethyl)-1 ,3-dihydro-2/-/- imidazo[4,5-c]quinolin-2-one (70mg) as a brown powder.
LC-HRMS: ES+ exact mass calculated for C22H20N5O3 402.1566 MH+, found: 402.1576, Rt 1.96min.
Example 50: 7-(3,5-dimethyl-4-isoxazolyl)-1 -[(2,4-dimethyl-1 ,3-thiazol-5-yl)methyl]-8- (methyloxy)-l ,3-dihydro-2H-imidazo[4,5-c]quinolin-2-one
Figure imgf000094_0002
To a solution of 7-(3,5-dimethyl-4-isoxazolyl)-4-{[(2,4-dimethyl-1 ,3-thiazol-5- yl)methyl]amino}-6-(methyloxy)-3-quinolinecarboxamide (for a preparation see intermediate 65, 0.3g) in acetonitrile (50ml) was added bis(trifluoroacetoxy)iodo]benzene (0.443g) and the reaction mixture was stirred at room temperature overnight. The reaction mixture was then poured into water and extracted with DCM. The organic phase was dried over Na2S04 and concentrated to dryness to give an orange oil. The residue was purified by flash chromatography on silica gel eluting with DCM / methanol (98: 2 then 97:3) give a sticky pale yellow solid, which was triturated in hot diisopropylether to give 7- (3,5-dimethyl-4-isoxazolyl)-1 -[(2,4-dimethyl-1 ,3-thiazol-5-yl)methyl]-8-(methyloxy)-1 ,3- dihydro-2H-imidazo[4,5-c]quinolin-2-one (0.21 g) as a cream coloured solid.
LC/MS: MH+ 436.06, [M-H]" 434.12
Example 51 : 7-(3,5-dimethyl-4-isoxazolyl)-8-(methyloxy)-2-[(methyloxy)methyl]-1 -[1 - (2-pyridinyl)ethyl]-1 H-imidaz 4,5-c]quinoline
Figure imgf000095_0001
To a solution of 7-(3,5-dimethyl-4-isoxazolyl)-6-(methyloxy)-/V4-[1 -(2-pyridinyl)ethyl]-3,4- quinolinediamine (for a preparation see Intermediate 65, 250mg) in DCM (20ml) was added (methyloxy)acetic acid (1 16 mg) followed by HATU (1 .2 equiv) and triethylamine (1 .5 equiv). The reaction mixture was stirred at room temperature for 1 h, and was then hydrolysed using a saturated solution of sodium hydrogen carbonate. The mixture was extracted with DCM and the organic dried over Na2S04, filtered and concentrated to dryness. The resulting product was dissolved in ethanol (60ml) and a solution of sodium hydroxide (1 N, 15ml) was added. The reaction mixture was heated to 90°C for 16h and was then allowed to cool to room temperature. DCM (150ml) and water (100ml) were added. The phases were separated and the organic phase was dried over Na2S04, filtered and evaporated to dryness. The residue was purified by silica gel chromatography eluting with DCM / methanol (95:5). The resulting residue was taken-up in acetonitrile, washed with diethyl ether and dried in vacuo to give 7-(3,5-dimethyl-4- isoxazolyl)-8-(methyloxy)-2-[(methyloxy)methyl]-1 -[1 -(2-pyridinyl)ethyl]-1 /-/-imidazo[4,5- c]quinoline (90mg) as a beige powder.
LC-HRMS: ES+ exact mass calculated for C28H3oN503 444.2036 MH+, found: 444.2072, Rt 2.39 min. Example 52: 7-(3,5-dimethyl-4-isoxazolyl)-1 -[(5-methyl-3-isoxazolyl)methyl]-8- (methyloxy)-1 ,3-dihydro-2H-imid -c]quinolin-2-one
Figure imgf000096_0001
7-(3,5-Dimethyl-4-isoxazolyl)-4-{[(5-methyl-3-isoxazolyl)methyl]amino}-6-(meth
quinolinecarboxamide (for a preparation see Intermediate 67, 280mg) was partially dissolved in acetonitrile (30ml) and bis(trifluoroacetoxy)iodo]benzene (800mg) was added. The reaction mixture was stirred at 50°C for 12h and was then concentrated to dryness. The crude was taken-up in water and extracted with DCM. The organic was dried over Na2S04, filtered and evaporated to dryness. The residue was purified by silica gel chromatography eluting with DCM / methanol (95:5), and the obtained product was subsequently taken-up in diethyl ether to give 7-(3,5-dimethyl-4-isoxazolyl)-1 -[(5-methyl-3- isoxazolyl)methyl]-8-(methyloxy)-1 ,3-dihydro-2/-/-imidazo[4,5-c]quinolin-2-one (200mg). LC/MS: Rt 2.52min. Example 53: 7-(3,5-dimethyl-4-isoxazolyl)-8-(methyloxy)-1 -(2-thienylmethyl)-1 ,3- dihydro-2H-imidazo[4,5-c]quinolin-2-one
Figure imgf000096_0002
To a magnetically stirred solution of 7-(3,5-dimethyl-4-isoxazolyl)-6-(methyloxy)-/V4-(2- thienylmethyl)-3,4-quinolinediamine (for a preparation see intermediate 68, 1 .5g,) in acetonitrile (50ml) in a 250ml flask equipped with a reflux condenser was added bis(trifluoroacetoxy)iodo]benzene (2.37g). The reaction mixture was stirred at 20°C for 2h then heated at 50°C for 1 h. Water was added to the reaction and the mixture extracted with DCM. The organic phase was dried over Na2S04, filtered and concentrated to dryness. The residue was purified by chromatography on silica gel (25 g, BP-SUP silica) eluting with DCM and then DCM / methanol (95:5). The resulting residue was triturated in hot isopropyl ether, filtered and dried to give 7-(3,5-dimethyl-4-isoxazolyl)-8-(methyloxy)- 1 -(2-thienylmethyl)-1 ,3-dihydro-2/-/-imidazo[4,5-c]quinolin-2-one in 2 batches (1 .1 14g total) as a brown powder.
LC-HRMS: ES+ exact mass calculated for ΰ2ιΗ18Ν4038407.1 178 MH+, found: 407.1215; Rt 2.38min.
Example 54: 7-(3,5-dimethyl-4-isoxazolyl)-1 -[(2,4-dimethyl-1 ,3-thiazol-5-yl)methyl]-8- (methyloxy)-2-(tetrahydro-2H-pyran-4-yl)-1 H-imidazo[4,5-c]quinoline
Figure imgf000097_0001
A solution of /V-[7-(3,5-dimethyl-4-isoxazolyl)-4-{[(2,4-dimethyl-1 ,3-thiazol-5- yl)methyl]amino}-6-(methyloxy)-3-quinolinyl]tetrahydro-2/-/-pyran-4-carboxamide (for a preparation see Intermediate 71 , 0.37g) in acetic acid was stirred for 2h at 100°C. The reaction mixture was then evaporated to dryness and the residue was then taken-up in a saturated solution of sodium hydrogen carbonate. The mixture was extracted with DCM and the organic phase dried over Na2S04 and evaporated to dryness. The residue was purified by flash chromatography on silica gel, eluting with DCM / methanol ((8:2 then 94:6) to give a sticky beige solid (0.3 g). The latter was triturated with hot diisopropylether to give 7-(3,5-dimethyl-4-isoxazolyl)-1 -[(2,4-dimethyl-1 ,3-thiazol-5-yl)methyl]-8- (methyloxy)-2-(tetrahydro-2/-/-pyran-4-yl)-1 /-/-imidazo[4,5-c]quinoline (0.22g) as a white solid.
LC-HRMS: ES+ exact mass calculated for C27H30N5O3S1 504.2069 MH+, found: 504.2021 . Example 55: 7-(3,5-dimethyl-4-isoxazolyl)-1 -[(5-methyl-2-furanyl)methyl]-8- (methyloxy)-1 ,3-dihydro-2H-imidazo[4,5-c]quinolin-2-one
Figure imgf000098_0001
A mixture of 7-(3,5-dimethyl-4-isoxazolyl)-4-{[(5-methyl-2-furanyl)methyl]amino}-6- (methyloxy)-3-quinolinecarboxamide (for a preparation see Intermediate 72, 177mg) and bis(trifluoroacetoxy)iodo]benzene (375mg) in acetonitrile (10ml) was stirred overnight at room temperature. The reaction mixture was evaporated to dryness, the residue was taken-up in water and extracted with DCM. The organic was dried over Na2S04, filtered and evaporated to give 7-(3,5-dimethyl-4-isoxazolyl)-1 -[(5-methyl-2-furanyl)methyl]-8- (methyloxy)-1 ,3-dihydro-2H-imidazo[4,5-c]quinolin-2-one (10mg) as a beige powder.
LC-HRMS: ES+ exact mass calculated for C22H2iN404 405.1563 MH+, found: 405.1626; Rt 2.39
Figure imgf000098_0002
Figure imgf000099_0001
Figure imgf000100_0001
Figure imgf000101_0001
Figure imgf000102_0001
Figure imgf000103_0001
Figure imgf000104_0001
Figure imgf000105_0001
Figure imgf000106_0001
Figure imgf000107_0001
Figure imgf000108_0001
Figure imgf000109_0001
Figure imgf000110_0001
Figure imgf000111_0001
Figure imgf000112_0001
Figure imgf000113_0001
Figure imgf000114_0001
Figure imgf000115_0001
Figure imgf000116_0001
Figure imgf000117_0001
Figure imgf000118_0001
Figure imgf000119_0001
Figure imgf000120_0001
Figure imgf000121_0001
Figure imgf000122_0001
Figure imgf000123_0001
Figure imgf000124_0001
Figure imgf000125_0001
Figure imgf000126_0001
Figure imgf000127_0001
Figure imgf000128_0001
Figure imgf000129_0001
Figure imgf000130_0001
Figure imgf000131_0001
Reference Compounds
Experimental details of LC-MS methods D and F as referred to in the Reference compounds below are as follows: LC/MS (Method D) was conducted on a Supelcosil LCABZ+PLUS column (3μηΊ, 3.3cm x 4.6mm ID) eluting with 0.1 % HCO2H and 0.01 M ammonium acetate in water (solvent A), and 95% acetonitrile and 0.05% HCO2H in water (solvent B), using the following elution gradient 0-0.7 minutes 0%B, 0.7-4.2 minutes 0→100%B, 4.2-5.3 minutes 100%B, 5.3-5.5 minutes 100→0%B at a flow rate of 3 mL/minute. The mass spectra (MS) were recorded on a Fisons VG Platform mass spectrometer using electrospray positive ionisation
[(ES+ve to give [M+H]+ and [M+NH4]+ molecular ions] or electrospray negative ionisation
[(ES-ve to give [M-H]- molecular ion] modes. Analytical data from this apparatus are given with the following format : [M+H]+ or [M-H]". LC/MS (Method F) was conducted on an Sunfire C18 column (30mm x 4.6mm i.d. 3.5μηι packing diameter) at 30 degrees centigrade, eluting with 0.1 % v/v solution of trifluoroacetic acid in water (Solvent A) and 0.1 % v/v solution of trifluoroacetic acid in acetonitrile (Solvent B) using the following elution gradient 0-0.1 min 3%B, 0.1 - 4.2min 3 - 100% B, 4.2-4.8min 100% B, 4.8-4.9min 100-3%B, 4.9 - 5.0min 3% B at a flow rate of 3ml/min. The UV detection was an averaged signal from wavelength of 210nm to 350nm and mass spectra were recorded on a mass spectrometer using positive electrospray ionization, lonisation data was rounded to the nearest integer.
LC/HRMS: Analytical HPLC was conducted on a Uptisphere-hsc column (3μηι 33 x 3 mm id) eluting with 0.01 M ammonium acetate in water (solvent A) and 100% acetonitrile (solvent B), using the following elution gradient 0-0.5 minutes 5% B, 0.5-3.75 minutes 5→100% B, 3.75-4.5 100% B, 4.5-5 100→5% B, 5-5.5 5% B at a flow rate of 1.3 mL/minute. The mass spectra (MS) were recorded on a micromass LCT mass spectrometer using electrospray positive ionisation [ES+ve to give MH+ molecular ions] or electrospray negative ionisation [ES-ve to give (M-H)- molecular ions] modes.
TLC (thin layer chromatography) refers to the use of TLC plates sold by Merck coated with silica gel 60 F254. Reference compound A: 2-methyl-6-(methyloxy)-4H-3,1 -benzoxazin-4-one
Figure imgf000133_0001
A solution of 5-methoxyanthranilic acid (Lancaster) (41 .8 g, 0.25 mol) was refluxed in acetic anhydride (230ml) for 3.5 h before being concentrated under reduced pressure. The crude compound was then concentrated twice in the presence of toluene before being filtered and washed twice with ether to yield to the title compound (33.7 g, 71 % yield) as a brown solid; LC/MS (Method D): m/z 192 [M+H]+, Rt 1 .69 min.
Reference compound B: [2-amino-5-(methyloxy)phenyl](4-chlorophenyl)methanone
Figure imgf000133_0002
To a solution of 2-methyl-6-(methyloxy)-4H-3,1 -benzoxazin-4-one (for a preparation see Reference compound A) (40.0 g, 0.21 mol) in a toluene/ether (2/1 ) mixture (760ml) at 0°C was added dropwise a solution of 4-chlorophenylmagnesium bromide (170ml, 1 M in diethyl ether, 0.17 mol). The reaction mixture was allowed to warm to room temperature and stirred for 1 h before being quenched with 1 N HCI (200ml). The aqueous layer was extracted with EtOAc (3 x 150ml) and the combined organics were washed with brine (100ml), dried over Na2S04, filtered and concentrated under reduced pressure. The crude compound was then dissolved in ethanol (400ml) and 6N HCI (160ml) was added. The reaction mixture was refluxed for 2 h before being concentrated to one-third in volume. The resulting solid was filtered and washed twice with ether before being suspended in EtOAc and neutralised with 1 N sodium hydroxide. The aqueous layer was extracted with EtOAc (3 x 150ml) and the combined organics were washed with brine (150ml), dried over Na2S04, filtered and concentrated under reduced pressure. The title compound was obtained as a yellow solid (39 g, 88 % yield); LC/MS (Method D): m/z 262 [M+H]+, Rt 2.57 min.
Reference Compound C: Methyl A^-^-^-chlorophenylJcarbonyl]^- (methyloxy)phenyl]-yV2-{[(9H-fluoren-9-ylmethyl)oxy]carbonyl}-L-D-asparaginate
Figure imgf000134_0001
Methyl /V-{[(9/-/-fluoren-9-ylmethyl)oxy]carbonyl}-L-a-aspartyl chloride {Int. J. Peptide Protein Res. 1992, 40, 13-18) (93 g, 0.24 mol) was dissolved in CHCI3 (270ml) and [2- amino-5-(methyloxy)phenyl](4-chlorophenyl)methanone (for a preparation see Reference compound B) (53 g, 0.2 mol) was added. The resulting mixture was stirred at 60°C for 1 h before being cooled and concentrated at 60% in volume. Ether was added at 0°C and the resulting precipitate was filtered and discarded. The filtrate was concentrated under reduced pressure and used without further purification. Reference compound D: Methyl [(3S)-5-(4-chlorophenyl)-7-(methyloxy)-2-oxo-2,3- dihydro-1 H-1 ,4-benzodiazepin- -yl]acetate
Figure imgf000134_0002
To a solution of methyl N1 -[2-[(4-chlorophenyl)carbonyl]-4-(methyloxy)phenyl]-N2-{[(9H- fluoren-9-ylmethyl)oxy]carbonyl}-L-a-asparaginate (for a preparation see Reference compound C) (assumed 0.2 mol) in DCM (500ml) was added Et3N (500ml, 3.65 mol) and the resulting mixture was refluxed for 24h before being concentrated. The resulting crude amine was dissolved in 1 ,2-DCE (1.5 L) and AcOH (104ml, 1 .8 mol) was added carefully. The reaction mixture was then stirred at 60°C for 2h before being concentrated in vacuo and dissolved in DCM. The organic layer was washed with 1 N HCI and the aqueous layer was extracted with DCM (x3). The combined organic layers were washed twice with water, and brine, dried over Na2S04, filtered and concentrated under reduced pressure. The crude solid was recrystallised in MeCN leading to the title compound (51 g) as a pale yellow solid. The filtrate could be concentrated and recrystallised in MeCN to give to another 10 g of the desired product Rf = 0.34 (DCM/MeOH : 95/5).
HRMS MH+calculated for Ci9H18 35CIN204 373.0955; found 373.0957. Reference compound E: Methyl [(3S)-5-(4-chlorophenyl)-7-(methyloxy)-2-thioxo-2,3- dihydro-1 H-1 ,4-benzodiazepin- -yl]acetate
Figure imgf000135_0001
A suspension of P4S10 (36.1 g, 81.1 mmol) and Na2C03 (8.6 g, 81 .1 mmol) in 1 ,2-DCE (700ml) at room temperature was stirred for 2 h before methyl [(3S)-5-(4-chlorophenyl)-7- (methyloxy)-2-oxo-2,3-dihydro-1 /-/-1 ,4-benzodiazepin-3-yl]acetate (for a preparation see Reference compound D) (16.8 g, 45.1 mmol) was added. The resulting mixture was stirred at 70°C for 2 h before being cooled and filtered. The solid was washed twice with DCM and the filtrate washed with sat. Sodium hydrogen carbonate and brine. The organic layer was dried over Na2S04, filtered and concentrated under reduced pressure. The crude product was purified by flash-chromatography on silica gel (DCM/MeOH : 99/1 ) to afford the title compound (17.2 g, 98% yield) as a yellowish solid. LC/MS (Method D): m/z 389 [M(35CI)+H]+, Rt 2.64 min
HRMS MH+calculated for Ci9H18 35CIN203S 389.0727; found 389.0714.
Reference compound F: Methyl [(3S)-2-[(1Z)-2-acetylhydrazino]-5-(4-chlorophenyl)- 7-(methyloxy)-3H-1 ,4-benzodiazepin-3-yl]acetate
Figure imgf000135_0002
To a suspension of methyl [(3S)-5-(4-chlorophenyl)-7-(methyloxy)-2-thioxo-2,3-dihydro- 1 H-1 ,4-benzodiazepin-3-yl]acetate (for a preparation see Reference compound E (9.0 g, 23.2 mmol) in THF (300ml) at 0°C was added hydrazine monohydrate (3.4ml, 69.6 mmol) dropwise. The reaction mixture was stirred for 5h between 5°C and 15°C before being cooled at 0°C. Et3N (9.7ml, 69.6 mmol) was then added slowly and acetyl chloride (7.95ml, 69.6 mmol) was added dropwise. The mixture was then allowed to warm to room temperature for 16h before being concentrated under reduced pressure. The crude product was dissolved in DCM and washed with water. The organic layer was dried over Na2S04, filtered and concentrated in vacuo to give the crude title compound (9.7 g, 98% yield) which was used without further purification. Rf = 0.49 (DCM/MeOH : 90/10).
Reference compound G: Methyl [(4S)-6-(4-chlorophenyl)-1 -methyl-8-(methyloxy)-4H- [1 ,2,4]triazolo[4,3-a][1 ,4]benzodiazepin-4-yl]acetate
Figure imgf000136_0001
The crude methyl [(3S)-2-[(1Z)-2-acetylhydrazino]-5-(4-chlorophenyl)-7-(methyloxy)-3H- 1 ,4-benzodiazepin-3-yl]acetate (for a preparation see Reference compound F) (assumed 9.7 g) was suspended in THF (100ml) and AcOH (60ml) was added at room temperature. The reaction mixture was stirred at this temperature for 2 days before being concentrated under reduced pressure. The crude solid was triturated in /'-Pr20 and filtered to give the title compound (8.7 g, 91 % over 3 steps) as an off-white solid.
HRMS MH+calculated for C2iH2oCIN403 41 1 .1229; found 41 1.1245.
Reference compound H: [(4S)-6-(4-Chlorophenyl)-1 -methyl-8-(methyloxy)-4H- [1 ,2,4]triazolo[4,3-a][1 ,4]benzodiazepin-4-yl]acetic acid
Figure imgf000136_0002
To a solution of methyl [(4S)-6-(4-chlorophenyl)-1 -methyl-8-(methyloxy)-4/-/- [1 ,2,4]triazolo[4,3-a][1 ,4]benzodiazepin-4-yl]acetate (for a preparation see Reference compound G) (7.4 g, 18.1 mmol) in THF (130ml) at room temperature was added 1 N sodium hydroxide (36.2ml, 36.2 mmol). The reaction mixture was stirred at this temperature for 5h before being quenched with 1 N HCI (36.2ml) and concentrated in vacuo. Water is then added and the aqueous layer was extracted with DCM (x3) and the combined organic layers were dried over Na2S04, filtered and concentrated under reduced pressure to give the title compound (7 g, 98% yield) as a pale yellow solid. Reference compound H: 1 ,1 -dimethylethyl [5-({[(4S)-6-(4-chlorophenyl)-1 -methyl-8- (methyloxy)-4H-[1 ,2,4]triazolo[4,3-a][1 ,4]benzodiazepin-4- yl]acetyl}amino)pentyl]carbamate
Figure imgf000137_0001
A mixture of [(4S)-6-(4-chlorophenyl)-1 -methyl-8-(methyloxy)-4H-[1 !2,4]triazolo[4,3- a][1 ,4]benzodiazepin-4-yl]acetic acid (for a preparation see Reference compound G) (1 .0g, 2.5mmol), HATU (1 .9g, 5mmol) and DIPEA (0.88ml, 5mmol) was stirred for 80 minutes at room temperature, to this was added 1 ,1 -dimethylethyl (4- aminobutyl)carbamate (1 .05ml, 5.0mmol, available from Aldrich). The reaction mixture was stirred at room temperature for 2h before it was concentrated. The residue was taken up in dichloromethane and washed with 1 N HCI. The aqueous layer was extracted with dichloromethane twice. Organic layer was washed with 1 N sodium hydroxide, followed by a saturated solution of sodium chloride, dried over sodium sulphate and concentrated. The residue was purified by flash-chromatography on silica using dichloromethane/ methanol 95/5 to give the title compound as a yellow solid (1 .2g). LC/MS (Method D): rt = 3.04 min.
Reference compound J: N-(5-aminopentyl)-2-[(4S)-6-(4-chlorophenyl)-1 -methyl-8-
(methyloxy)-4H-[1 ,2,4]triazolo[4,3-a][1 ,4]benzodiazepin-4-yl]acetamide
trifluoroacetate
Figure imgf000137_0002
To a solution of 1 ,1 -dimethylethyl [5-({[(4S)-6-(4-chlorophenyl)-1 -methyl-8-(methyloxy)-
4H-[1 ,2,4]triazolo[4,3-a][1 ,4]benzodiazepin-4-yl]acetyl}amino)pentyl]carbamate (for a preparation see Reference compound H) (0.2 g, 0.34 mmol) in dichloromethane (3ml) was added trifluoroacetic acid (0.053ml, 0.68 mmol) dropwise at 0°C. The reaction mixture was stirred for 3h from 0°C to room temperature. The reaction mixture was concentrated to dryness to afford the title compound as a hygroscopic yellow oil (200mg)
LC/MS (Method D): rt = 2.33min.
HRMS MH+calculated for CaH CINeCk 481 .21 19; found 481.2162.
Reference compound K: Mixture of 5- and 6- isomers of Alexa Fluor 488-N-(5- aminopentyl)-2-[(4S)-6-(4-chlorophenyl)-1 -methyl -8 -(methyl oxy)-4H- [1 ,2,4]triazolo[4,3-a][1 ,4]benzodiazepin-4-yl]acetamide
Figure imgf000138_0001
N-(5-aminopentyl)-2-[(4S)-6-(4-chlorophenyl)-1 -methyl-8-(methyloxy)-4H- [1 ,2,4]triazolo[4,3-a][1 ,4]benzodiazepin-4-yl]acetamide trifluoroacetate (for a preparation see Reference compound J) (7.65 mg, 0.013 mmol) was dissolved in N,N- dimethylformamide (DMF) (300 μΙ) and added to Alexa Fluor 488 carboxylic acid succinimidyl ester (5 mg, 7.77 μηηοΙ, mixture of 5 and 6 isomers, available from Invitrogen, product number A-20100) in an Eppendorf centrifuge tube. Hunig's base (7.0 μΙ, 0.040 mmol) was added and the mixture vortex mixed overnight. After 18h the reaction mixture was evaporated to dryness and the residue redissolved in DMSO/water (50%, <1 ml total), applied to a preparative Phenomenex Jupiter C18 column and eluted with a gradient of 95% A: 5% B to 100% B (A = 0.1 % trifluoroacetic acid in water, B= 0.1 % TFA/90% acetonitrile/10% water) at a flow rate of 10ml/min over 150 minutes. Impure fractions were combined and re-purified using the same system. Fractions were combined and evaporated to yield the title product (2.8mg) as a mixture of the 2 regioisomers shown. LC/MS (Method F):, MH+ = 999, rt = 1 .88min. Biological Test Methods
Fluorescence anisotropy binding assay
The binding of the compounds of formula (I) to Bromodomain BRD2, BRD3 and BRD4 was assessed using a Fluorescence Anisotropy Binding Assay.
The Bromodomain protein, fluorescent ligand (Reference compound K see above) and a variable concentration of test compound are incubated together to reach thermodynamic equilibrium under conditions such that in the absence of test compound the fluorescent ligand is significantly (>50%) bound and in the presence of a sufficient concentration of a potent inhibitor the anisotropy of the unbound fluorescent ligand is measurably different from the bound value.
All data was normalized to the mean of 16 high and 16 low control wells on each plate. A four parameter curve fit of the following form was then applied: y = a + ((b - a) / ( 1 + ( 10 Λ x / 10 Λ c ) Λ d )
Where 'a' is the minimum, 'b' is the Hill slope, 'c' is the plC50 and 'd' is the maximum.
Recombinant Human Bromodomains (Bromodomain BRD2 (1 -473), Bromodomain BRD3 (1 -435) and Bromodomain BRD4 (1 -477)) were expressed in E.coli cells (in pET15b vector) with a six-His tag at the N-terminal. The His-tagged Bromodomain was extracted from E.coli cells using 0.1 mg/ml lysozyme and sonication. The Bromodomain was then purified by affinity chromatography on a HisTRAP HP column, eluting with a linear 10- 500mM Imidazole gradient, over 20 Cv. Further purification was completed by Superdex 200 prep grade size exclusion column. Purified protein was stored at -80°C in 20mM HEPES pH 7.5 and 100mM NaCI. Protocol for Bromodomain BRD2: All components were dissolved in buffer composition of 50 mM HEPES pH7.4, 150mm NaCI and 0.5mM CHAPS with final concentrations of Bromodomain 2, 75nM, fluorescent ligand 5nM.10 μΙ of this reaction mixture was added using a micro multidrop to wells containing 10Onl of various concentrations of test compound or DMSO vehicle (1 % final) in Greiner 384 well Black low volume microtitre plate and equilibrated in dark 60 mins at room temperature. Fluorescence anisotropy was read in Envision (Aex= 485nm, AEM = 530nm; Dichroic -505nM). Protocol for Bromodomain BRD3 : All components were dissolved in buffer of composition 50 mM HEPES pH7.4, 150mm NaCI and 0.5mM CHAPS with final concentrations of Bromodomains 3 75nM, fluorescent ligand 5nM. 10 μΙ of this reaction mixture was added using a micro multidrop to wells containing 10Onl of various concentrations of test compound or DMSO vehicle (1 % final) in Greiner 384 well Black low volume microtitre plate and equilibrated in dark 60 mins at room temperature. Fluorescence anisotropy was read in Envision (Aex= 485nm, AEM = 530nm; Dichroic -505nM). Protocol for Bromodomain BRD4: All components were dissolved in buffer of composition 50 mM HEPES pH7.4, 150mm NaCI and 0.5mM CHAPS with final concentrations of Bromodomain 4 75nM, fluorescent ligand 5nM. 10 μΙ of this reaction mixture was added using a micro multidrop to wells containing 10Onl of various concentrations of test compound or DMSO vehicle (1 % final) in Greiner 384 well Black low volume microtitre plate and equilibrated in dark 60 mins at room temperature. Fluorescence anisotropy was read in Envision (Aex= 485nm, AEM = 530nm; Dichroic - 505nM).
All examples (with the exception of examples 22, 38 and 210 -225) were tested in the assays described above. All tested compounds had a plC5o≥ 5.0 in one or more of the BRD2, BRD3 and BRD4 assays described above. Examples 1 , 3-8, 10-21 , 23-31 , 33-37, 39-43, 45-61 , 63-158, 160, 162-170, 172, 173, 176-181 , 183-185 and 187 had a plC50≥ 6.0 in one or more of the BRD2, BRD3 and BRD4 assays described above. LPS stimulated Whole Blood measuring TNF levels assay
Activation of monocytic cells by agonists of toll-like receptors such as bacterial lipopolysaccharide (LPS) results in production of key inflammatory mediators including TNFa. Such pathways are widely considered to be central to the pathophysiology of a range of auto-immune and inflammatory disorders.
Compounds to be tested are diluted to give a range of appropriate concentrations and 1 ul of the dilution stocks is added to wells of a 96 plate. Following addition of whole blood (130ul) the plates are incubated at 37 degrees (5% C02) for 30 min before the addition of 10ul of 2.8ug/ml LPS, diluted in complete RPMI 1640 (final concentration =200ng/ml), to give a total volume of 140ul per well. After further incubation for 24h at 37 degrees, 140ul of PBS are added to each well. The plates are sealed, shaken for 10 minutes and then centrifuged (2500rpm x 10 min). 10Oul of the supernatant are removed and TNFa levels assayed by immunoassay (typically by MesoScale Discovery technology) either immediately or following storage at -20 degrees. Dose response curves for each compound was generated from the data and an IC50 value was calculated.
Examples 1 1 , 12, 15, 27, 31 , 33, 36, 108 and 126 were tested in the above assay were found to have a plC5o≥ 5.5.
Measurement of LPS induced IL-6 secretion from whole blood
Activation of monocytic cells by agonists of toll-like receptors such as bacterial lipopolysaccharide (LPS) results in production of key inflammatory mediators including IL- 6. Such pathways are widely considered to be central to the pathophysiology of a range of auto-immune and inflammatory disorders.
Compounds to be tested are diluted to give a range of appropriate concentrations of which 1 ul of the diluted stocks is added to a 96 well plate. Following addition of whole blood (130ul) the plates are incubated at 37 degrees (5% C02) for 30 min before the addition of 10ul of 2.8ug/ml LPS, diluted in complete RPMI 1640 (final concentration =200ng/ml), to give a total volume of 140ul per well. After further incubation for 24 hours at 37 degrees, 140ul of PBS are added to each well. The plates are sealed, shaken for 10 minutes and then centrifuged (2500rpm x 10 min). 10Oul of the supernatant are removed and IL-6 levels assayed by immunoassay (typically by MesoScale Discovery technology) either immediately or following storage at -20 degrees. Concentration response curves for each compound was generated from the data and an IC50 value was calculated Examples 1 1 , 36-37, 39-40, 45 and 51 were tested in the above assay were found to have a plC5o≥ 5.5.
These data demonstrate that bromodomain inhibitors tested in the above two whole blood assays inhibited the production of key inflammatory mediators TNFa and/or IL-6.
In Vivo Mouse Endotoxemia Model
High doses of Endotoxin (bacterial lipopolysaccharide) administered to animals produce a profound shock syndrome including a strong inflammatory response, dysregulation of cardiovascular function, organ failure and ultimately mortality. This pattern of response is very similar to human sepsis and septic shock, where the body's response to a significant bacterial infection can be similarly life threatening.
To test the compounds for use in the invention groups of eight Balb/c male mice were given a lethal dose of 15 mg/kg LPS by intraperitoneal injection. Ninety minutes later, animals were dosed intravenously with vehicle (20% cyclodextrin 1 % ethanol in apyrogen water) or compound (10 mg/kg). The survival of animals was monitored at 4 days.
Numbers of animals surviving at 4 days (summed across multiple repeat experiments) Vehicle 4/66 (6%)
Compound of Example 1 1 55/66 (83%)
Compound of Example 31 9/24 (38%)
These data demonstrate that the bromodomain inhibitors teated in the above model gave rise to a significant animal survival effect following intravenous administration.
Oncology Cell Growth Assay
Human cell lines (n = 33 comprising 15 heme cell lines, 14 breast cell lines and 4 other cell lines) were cultured in RPMI-1640 containing 10% fetal bovine serum, 1000 viable cells per well were plated in 384-well black flat bottom polystyrene plates (Greiner #781086) in 48 μΙ of culture media. All plates were placed at 5% C02, 37°C overnight. The following day one plate was harvested with CellTiter-Glo (CTG, Promega #G7573) for a time equal to 0 (TO) measurement and compound (20 point titration from 14.7 uM to 7 pM) was added to the remaining plates. The final concentration of DMSO in all wells was 0.15%. Cells were incubated for 72 hours or the indicated time and each plate was developed with CellTiter-Glo reagent using a volume equivalent to the cell culture volume in the wells. Plates were shaken for approximately 2 minutes and chemiluminescent signal was read on the Analyst GT (Molecular Devices) or EnVision Plate Reader (Perkin Elmer).
Results were expressed as a percent of the TO and plotted against the compound concentration. The TO value was normalized to 100% and represents the number of cells at time of compound addition and the concentration response data were fit with a 4 parameter curve fit using XLfit software (model 205). The concentration that inhibited cell growth by 50% (glC50) is the midpoint of the 'growth window' (between the TO and DMSO control). The Ymin - TO value is determined by subtracting the TO value (100%) from the Ymin value (%) determined from the fit of the concentration response curve. Values from the wells with no cells were subtracted from all samples for background correction.
The compound of Examples 1 1 and 36 was tested in accordance with the above assay and found to have a glC50 in the range 1 18-1 1 100 nM across all cell lines, more specifically in the range 1 18 - 1036 nM for heme cell lines and 432 - 1 1 100 nM for breast cell lines.
These data demonstrate that the bromodomain inhibitor tested in the above assay inhibited cell growth in a panel of oncology cell lines.
All publications, including but not limited to patents and patent applications, cited in this specification are herein incorporated by reference as if each individual publication were specifically and individually indicated to be incorporated by reference herein as though fully set forth.

Claims

CLAIMS 1. A compound of formula (I) or a salt thereof
Figure imgf000144_0001
wherein:
• A is a group selected from the following:
Figure imgf000144_0002
• X represents CH or N;
• Y represents CH or N with the proviso that when X is N, Y is CH;
· Rx represents O or S;
• R1 represents C-| .galkyl, C3_gcycloalkyl, a 5 or 6 membered heterocyclyl, an aromatic group or a heteroaromatic group, wherein the aromatic group or the heteroaromatic group is optionally substituted by one to three groups selected from:
halogen, hydroxy, cyano, nitro, C-| .galkyl, C-|_4alkoxy, haloC-| _4alkyl, haloC-| _4alkoxy, hydroxy Chalky I, C-| _4alkoxyC-| _4alkyl, C-μ 4alkoxycarbonyl, C-| _4alkylsulfonyl, C-| _4alkylsulfonyloxy, C-μ 4alkylsulfonylC-| _4alkyl, C-| ^alkylsulfonamido;
• R2 is hydrogen or C-| .galkyl,
· R2a represents: o H, C-| .6alkyl, C-| _6haloallkyl, (CH2)mcyano, (CH2)mOH! (CH2)mC1. galkoxy, (CH2)mC-| .ghaloalkoxy, (CH2)mC-| .ghaloalkyl
(CH2)mC(0)NRaRb, (CH2)mNRaRb and (CH2)mC(0)CH3,
o (CHR6)pphenyl optionally substituted by C-| .galkyl, C-| .galkoxy, cyano haloC-| galkoxy, haloC-| .4alkyl,
o (CHR6)pheteroaromatic, (CHR6)pheterocyclyl,
wherein
Ra represents H, C-|_galkyl, or heterocyclyl;
Rb represents H or C-| .galkyl, or
Ra and Rb together with the N to which they are attached form a 5 or 6 membered heterocyclyl;
• R2b represents H, C-| .galkyl, (CH2)2C-| .galkoxy, (CH2)2cyano, (CH2)mphenyl,
(CH2)2heterocyclyl;
• R3 represents hydrogen;
· R4 represents hydrogen, cyano or C-| .galkyl;
• Z represents O; or when R4 represents hydrogen and A is a group selected from (i) or (ii) and wherein Rx represents O, Z may additionally represent NH;
• R5 represents hydrogen or C-| .galkoxy;
• R6 represents hydrogen or C-| .galkyl;
· m represents 1 , 2 or 3; and
• n represents 0, 1 or 2
• p represents 0, 1 or 2.
2. A compound or a salt thereof according to claim 1 which is a compound of formula (1A)
Figure imgf000145_0001
wherein A and R4 are as defined for formula (I).
3. A compound or a salt thereof according to claim 2 in which R4 is hydrogen and A is a group of formula (i) or (ii) wherein n is 1 .
4. A compound or a salt thereof according to any one of claims 1 - 3 in which A is a group of formula (ii) wherein R2b represents (CH2)2C-| .galkoxy.
A compound or a salt thereof according to claim 1 which is a compound of formula
Figure imgf000146_0001
wherein R1, R2, R3, R5 and n are as defined for compounds of formula (I).
6. A compound or a salt thereof according to claim 5 in which R5 is -OCH3.
7. A compound or a salt thereof according to claim 1 which is a compound of formula (1 C)
Figure imgf000146_0002
wherein R1, R2, R3, R2a and n are defined above for compounds of formula (I).
8. A compound or a salt thereof according to claim 7 in which R2a represents H, C-μ 3alkyl, (CH2)mOH! (CH2)mC1.3alkoxy! (CH2)mNRaRb or (CHR6)pheterocyclyl, wherein
Ra represents H, C-| _3alkyl, or heterocyclyl;
Rb represents H or C-| _3alkyl, or
Ra and Rb together with the N to which they are attached form a 5 or 6 membered heterocyclyl;
R6 represents H or C<| _3alkyl;
m represents 1 , 2 or 3; and
p represents 0, 1 , 2.
9. A compound or a salt thereof according to claim 7 in which R2a is tetrahydropyranyl.
10. A compound or a salt thereof according to any one of claims 1 - 9 in which n is 1.
1 1 . A compound or a salt thereof according to any one of claims 1 - 10 in which R2 is hydrogen or methyl.
12. A compound or a salt thereof according to any one of claims 1 - 1 1 in which R1 represents a heteroaromatic group optionally substituted by one or two groups selected from hydroxy, C-| _3alkyl, C-| _3alkoxy, hydroxyC-| _4alkyl and C<| _4alkoxyC<| _4alkyl.
13. A compound or a salt thereof according to claim 12 in which the heteroaromatic group is pyridyl.
14. A compound or a salt thereof according to claim 12 in which the heteroaromatic group is selected from furanyl, thienyl, isoxazolyl, thiazolyl, pyrazolyl, pyrazinyl and pyrimidinyl.
15. A compound which is any of one of Examples 1 - 226 or a salt thereof.
16 A compound which is 7-(3,5-dimethyl-4-isoxazolyl)-8-(methyloxy)-1 -[(1 R)-1 -(2- pyridinyl)ethyl]-1 ,3-dihydro-2/-/-imidazo[4,5-c]quinolin-2-one
Figure imgf000148_0001
or a salt thereof.
17. A compound according to any one of claims 1 - 16 or a pharmaceutically acceptable salt thereof.
18. A pharmaceutical composition which comprises a compound or a pharmaceutically acceptable salt thereof as defined in claim 17 and one or more pharmaceutically acceptable carriers, diluents or excipients.
19. A combination pharmaceutical product comprising a compound or a pharmaceutically acceptable salt thereof as defined in claim 17 together with one or more other therapeutically active agents.
20. A compound or a pharmaceutically acceptable salt thereof as defined in claim 17 for use in therapy.
21 . A compound or a pharmaceutically acceptable salt thereof as defined in claim 17 for use in the treatment of diseases or conditions for which a bromodomain inhibitor is indicated.
22. A compound according to claim 21 , wherein the disease or condition is a chronic autoimmune and/or inflammatory condition.
23. A compound according to claim 21 , wherein the disease or condition is cancer.
24. The use of a compound or a pharmaceutically acceptable salt thereof as defined in claim 17 in the manufacture of a medicament for the treatment of diseases or conditions for which a bromodomain inhibitor is indicated.
25. A method of treating diseases or conditions for which a bromodomain inhibitor is indicated in a subject in need thereof which comprises administering a therapeutically effective amount of compound or a pharmaceutically acceptable salt thereof as defined in claim 17.
26. A method of treatment according to claim 25, wherein the disease or condition is a chronic autoimmune and/or inflammatory condition.
27. A method of treatment according to claim 25, wherein the disease or condition is cancer.
28. A method for inhibiting a bromodomain which comprises contacting the bromodomain with a compound of formula (I) or a pharmaceutically acceptable salt thereof as defined in claim 17.
PCT/EP2010/066699 2009-11-05 2010-11-03 Imidazo [4, 5-c] quinoline derivates as bromodomain inhibitors WO2011054846A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
ES10771770.4T ES2446358T3 (en) 2009-11-05 2010-11-03 Imidazo [4,5-c] quinoline derivatives as bromodomain inhibitors
US13/503,722 US8557984B2 (en) 2009-11-05 2010-11-03 Imidazo [4, 5-C] quinoline derivatives as bromodomain inhibitors
JP2012537382A JP5766708B2 (en) 2009-11-05 2010-11-03 Imidazo [4,5-c] quinoline derivatives as bromodomain inhibitors
EP10771770.4A EP2496576B1 (en) 2009-11-05 2010-11-03 Imidazo[4,5-c]quinoline derivatives as bromodomain inhibitors

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GBGB0919423.4A GB0919423D0 (en) 2009-11-05 2009-11-05 Novel compounds
GB0919423.4 2009-11-05

Publications (1)

Publication Number Publication Date
WO2011054846A1 true WO2011054846A1 (en) 2011-05-12

Family

ID=41501966

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2010/066699 WO2011054846A1 (en) 2009-11-05 2010-11-03 Imidazo [4, 5-c] quinoline derivates as bromodomain inhibitors

Country Status (6)

Country Link
US (1) US8557984B2 (en)
EP (1) EP2496576B1 (en)
JP (1) JP5766708B2 (en)
ES (1) ES2446358T3 (en)
GB (1) GB0919423D0 (en)
WO (1) WO2011054846A1 (en)

Cited By (117)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013024104A1 (en) 2011-08-17 2013-02-21 Glaxosmithkline Llc 4-(8-methoxy-1-((1-methoxypropan-2-yl)-2-(tetrahydro-2h-pyran-4-yl)-1 h-imidazo[4,5-c]quinolin-7-yl)-3,5-dimethylisoxazole and its use as bromodomain inhibitor
WO2013027168A1 (en) 2011-08-22 2013-02-28 Pfizer Inc. Novel heterocyclic compounds as bromodomain inhibitors
WO2013033269A1 (en) * 2011-08-29 2013-03-07 Coferon, Inc. Bioorthogonal monomers capable of dimerizing and targeting bromodomains and methods of using same
EP2635279A2 (en) * 2010-11-05 2013-09-11 Glaxosmithkline Intellectual Property (No. 2) Limited Chemical compounds
WO2014076146A1 (en) 2012-11-13 2014-05-22 Boehringer Ingelheim International Gmbh Triazolopyridazine
WO2014076237A1 (en) 2012-11-16 2014-05-22 Boehringer Ingelheim International Gmbh Triazolopyrazine
US8796261B2 (en) 2010-12-02 2014-08-05 Constellation Pharmaceuticals, Inc. Bromodomain inhibitors and uses thereof
WO2014128655A1 (en) * 2013-02-25 2014-08-28 Aurigene Discovery Technologies Limited Substituted imidazo[4,5-c]quinoline derivatives as bromodomain inhibitors
WO2014140077A1 (en) 2013-03-14 2014-09-18 Glaxosmithkline Intellectual Property (No.2) Limited Furopyridines as bromodomain inhibitors
WO2014154760A1 (en) * 2013-03-27 2014-10-02 Boehringer Ingelheim International Gmbh Indolinone analogues as brd4 inhibitors
WO2014173241A1 (en) 2013-04-26 2014-10-30 Beigene, Ltd. Substituted5-(3,5-dimethylisoxazol-4-yl)indoline-2-ones
JP2014531458A (en) * 2011-09-30 2014-11-27 シー・アンド・シー・リサーチ・ラボラトリーズC&C Research Laboratories Novel heterocyclic derivatives and uses thereof
WO2015013635A2 (en) 2013-07-25 2015-01-29 Dana-Farber Cancer Institute, Inc. Inhibitors of transcription factors and uses thereof
WO2015049629A1 (en) * 2013-10-01 2015-04-09 Piramal Enterprises Limited Imidazoquinoline compounds as bromodomain inhibitors
WO2015067770A1 (en) 2013-11-07 2015-05-14 Boehringer Ingelheim International Gmbh Triazolopyrazine derivatives as brd4 inhibitors
WO2015080707A1 (en) * 2013-11-26 2015-06-04 Gilead Sciences, Inc. Quinoline derivatives as bromodomain inhibitors
WO2015092118A1 (en) 2013-12-17 2015-06-25 Orion Corporation Spiro[cyclobutane-1,3'-indolin]-2'-one derivatives as bromodomain inhibitors
WO2015104653A1 (en) 2014-01-09 2015-07-16 Aurigene Discovery Technologies Limited Bicyclic heterocyclic derivatives as bromodomain inhibitors
WO2015117087A1 (en) 2014-01-31 2015-08-06 Dana-Farber Cancer Institute, Inc. Uses of diazepane derivatives
WO2015131005A1 (en) 2014-02-28 2015-09-03 The Regents Of The University Of Michigan 9h-pyrimido[4,5-b]indoles and related analogs as bet bromodomain inhibitors
WO2015169962A1 (en) * 2014-05-09 2015-11-12 Boehringer Ingelheim International Gmbh Benzimidazole derivatives
US9227985B2 (en) 2013-03-15 2016-01-05 Incyte Corporation Tricyclic heterocycles as bet protein inhibitors
US9249161B2 (en) 2010-12-02 2016-02-02 Constellation Pharmaceuticals, Inc. Bromodomain inhibitors and uses thereof
US9271978B2 (en) 2012-12-21 2016-03-01 Zenith Epigenetics Corp. Heterocyclic compounds as bromodomain inhibitors
US9278940B2 (en) 2012-11-21 2016-03-08 Zenith Epigenetics Corp. Cyclic amines as bromodomain inhibitors
CN105407894A (en) * 2013-03-14 2016-03-16 康威基内有限公司 Methods and compositions for inhibition of bromodomain-containing proteins
US9290514B2 (en) 2013-07-08 2016-03-22 Incyte Holdings Corporation Tricyclic heterocycles as BET protein inhibitors
US9296741B2 (en) 2011-12-30 2016-03-29 Abbvie Inc. Bromodomain inhibitors
US9309246B2 (en) 2013-12-19 2016-04-12 Incyte Corporation Tricyclic heterocycles as BET protein inhibitors
US9315501B2 (en) 2013-11-26 2016-04-19 Incyte Corporation Bicyclic heterocycles as BET protein inhibitors
US9328117B2 (en) 2011-06-17 2016-05-03 Constellation Pharmaceuticals, Inc. Bromodomain inhibitors and uses thereof
US9399640B2 (en) 2013-11-26 2016-07-26 Incyte Corporation Substituted pyrrolo[2,3-c]pyridines and pyrazolo[3,4-c]pyridines as BET protein inhibitors
JP2016522246A (en) * 2013-06-21 2016-07-28 ゼニス・エピジェネティクス・コーポレイションZenith Epigenetics Corp. Novel substituted bicyclic compounds as bromodomain inhibitors
JP2016523259A (en) * 2013-06-21 2016-08-08 ゼニス・エピジェネティクス・コーポレイションZenith Epigenetics Corp. Novel bicyclic bromodomain inhibitors
WO2016123709A1 (en) 2015-02-03 2016-08-11 Trillium Therapeutics Inc. Fluorinated imidazo[4,5-c]quinoline derivatives as inhibitors of bromodomain containing proteins
US9422292B2 (en) 2011-05-04 2016-08-23 Constellation Pharmaceuticals, Inc. Bromodomain inhibitors and uses thereof
WO2016138332A1 (en) 2015-02-27 2016-09-01 The Regents Of The University Of Michigan 9h-pyrimido [4,5-b] indoles as bet bromodomain inhibitors
WO2016146755A1 (en) 2015-03-19 2016-09-22 Glaxosmithkline Intellectual Property Development Limited Covalent conjugates of bet inhibitors and alpha amino acid esters
WO2016155884A1 (en) * 2015-04-02 2016-10-06 Merck Patent Gmbh Imidazolonyl quinolines and use thereof as atm kinase inhibitors
US9493483B2 (en) 2012-06-06 2016-11-15 Constellation Pharmaceuticals, Inc. Benzo [C] isoxazoloazepine bromodomain inhibitors and uses thereof
WO2016186453A1 (en) * 2015-05-20 2016-11-24 Kainos Medicine, Inc. Quinoline derivatives as bromodomain inhibitors
WO2016196065A1 (en) 2015-05-29 2016-12-08 Genentech, Inc. Methods and compositions for assessing responsiveness of cancers to bet inhibitors
WO2016199943A1 (en) * 2015-06-11 2016-12-15 Takeda Pharmaceutical Company Limited Heterocyclic compounds
WO2016203112A1 (en) 2015-06-16 2016-12-22 Orion Corporation Spiro[cyclobutane-1,3'-indolin]-2'-one derivatives as bromodomain inhibitors
WO2016203335A1 (en) 2015-06-18 2016-12-22 Pfizer Inc. Novel pyrido[2,3-b]pyrazinones as bet-family bromodomain inhibitors
US9527864B2 (en) 2014-09-15 2016-12-27 Incyte Corporation Tricyclic heterocycles as BET protein inhibitors
WO2016210275A1 (en) 2015-06-26 2016-12-29 Tensha Therapeutics, Inc. Treatment of nut midline carcinoma
WO2017001733A1 (en) 2015-07-02 2017-01-05 Orion Corporation Bicyclic heterocycle derivatives as bromodomain inhibitors
US9540368B2 (en) 2014-04-23 2017-01-10 Incyte Corporation 1H-pyrrolo[2,3-c]pyridin-7(6H)-ones and pyrazolo[3,4-c]pyridin-7(6H)-ones as inhibitors of BET proteins
US9561231B2 (en) 2012-06-12 2017-02-07 Abbvie Inc. Pyridinone and pyridazinone derivatives
WO2017050714A1 (en) 2015-09-22 2017-03-30 Glaxosmithkline Intellectual Property (No.2) Limited Pyridinone dicarboxamide for use as bromodomain inhibitors
US9624244B2 (en) 2012-06-06 2017-04-18 Constellation Pharmaceuticals, Inc. Benzo [B] isoxazoloazepine bromodomain inhibitors and uses thereof
WO2017091661A1 (en) * 2015-11-25 2017-06-01 Strovel Jeffrey William Bicyclic bet bromodomain inhibitors and uses thereof
US9695172B2 (en) 2014-01-31 2017-07-04 Dana-Farber Cancer Institute, Inc. Diazepane derivatives and uses thereof
US9714946B2 (en) 2013-03-14 2017-07-25 Dana-Farber Cancer Institute, Inc. Bromodomain binding reagents and uses thereof
WO2017142881A1 (en) 2016-02-15 2017-08-24 The Regents Of The University Of Michigan Fused 1,4-oxazepines and related analogs as bet bromodomain inhibitors
US9763956B2 (en) 2012-06-19 2017-09-19 The Broad Institute, Inc. Diagnostic and treatment methods in subjects having or at risk of developing resistance to cancer therapy
US9765039B2 (en) 2012-11-21 2017-09-19 Zenith Epigenetics Ltd. Biaryl derivatives as bromodomain inhibitors
US9776990B2 (en) 2012-04-20 2017-10-03 Abbvie Inc. Isoindolone derivatives
WO2017174620A1 (en) 2016-04-07 2017-10-12 Glaxosmithkline Intellectual Property (No.2) Limited Benzo[b]furans as bromodomain inhibitors
WO2017176958A1 (en) 2016-04-06 2017-10-12 The Regents Of The University Of Michigan Monofunctional intermediates for ligand-dependent target protein degradation
WO2017174621A1 (en) 2016-04-07 2017-10-12 Glaxosmithkline Intellectual Property (No.2) Limited Pyridyl derivatives as bromodomain inhibitors
US9789120B2 (en) 2010-05-14 2017-10-17 Dana-Farber Cancer Institute, Inc. Male contraceptive compositions and methods of use
WO2017180417A1 (en) 2016-04-12 2017-10-19 The Regents Of The University Of Michigan Bet protein degraders
WO2017202742A1 (en) 2016-05-24 2017-11-30 Glaxosmithkline Intellectual Property (No.2) Limited Pyridine dicarboxamide derivatives as bromodomain inhibitors
US9856255B2 (en) 2015-09-17 2018-01-02 Astrazeneca Ab Imidazo[4,5-c]quinolin-2-one compounds and their use in treating cancer
US9855271B2 (en) 2013-07-31 2018-01-02 Zenith Epigenetics Ltd. Quinazolinones as bromodomain inhibitors
WO2018041964A1 (en) 2016-09-02 2018-03-08 Glaxosmithkline Intellectual Property (No.2) Limited Imidazole derivatives and their use in the treatment of autoimmune or inflammatory diseases or cancers
WO2018041947A1 (en) 2016-09-02 2018-03-08 Glaxosmithkline Intellectual Property (No.2) Limited Imidazole derivatives and their use in the treatment of autoimmune or inflammatory diseases or cancers
WO2018041946A1 (en) 2016-09-02 2018-03-08 Glaxosmithkline Intellectual Property Development Limited Crystalline hydrate of the compound (2s,3r)-isopropyl 2-(((2-(1,5-dimethyl-6-oxo-1,6-dihydropyridin-3-yl)-1-((tetrahydro-2h-pyran-4-yl)methyl)-1h-benzo[d]imidazol-5-yl)methyl)amino)-3-hydroxybutanoate edisylate
WO2018052945A1 (en) 2016-09-13 2018-03-22 The Regents Of The University Of Michigan Fused 1,4-oxazepines as bet protein degraders
WO2018052949A1 (en) 2016-09-13 2018-03-22 The Regents Of The University Of Michigan Fused 1,4-diazepines as bet protein degraders
US9951074B2 (en) 2014-08-08 2018-04-24 Dana-Farber Cancer Institute, Inc. Dihydropteridinone derivatives and uses thereof
US9969747B2 (en) 2014-06-20 2018-05-15 Constellation Pharmaceuticals, Inc. Crystalline forms of 2-((4S)-6-(4-chlorophenyl)-1-methyl-4H-benzo[C]isoxazolo[4,5-e]azepin-4-yl)acetamide
WO2018109271A1 (en) 2016-12-13 2018-06-21 Orion Corporation New bromodomain inhibitors
WO2018144789A1 (en) 2017-02-03 2018-08-09 The Regents Of The University Of Michigan Fused 1,4-diazepines as bet bromodomain inhibitors
WO2018158210A1 (en) 2017-03-01 2018-09-07 Glaxosmithkline Intellectual Property (N°.2) Limited Pyridyl derivatives as bromodomain inhibitors
US10124009B2 (en) 2014-10-27 2018-11-13 Tensha Therapeutics, Inc. Bromodomain inhibitors
US10150756B2 (en) 2014-01-31 2018-12-11 Dana-Farber Cancer Institute, Inc. Diaminopyrimidine benzenesulfone derivatives and uses thereof
US10179125B2 (en) 2014-12-01 2019-01-15 Zenith Epigenetics Ltd. Substituted pyridines as bromodomain inhibitors
US10189834B2 (en) 2014-05-08 2019-01-29 Astrazeneca Ab Imidazo[4,5-c]quinolin-2-one compounds and their use in treating cancer
US10189832B2 (en) 2016-06-20 2019-01-29 Incyte Corporation Crystalline solid forms of a BET inhibitor
CN109384784A (en) * 2017-08-10 2019-02-26 浙江海正药业股份有限公司 Sulfonic acid amide derivatives, preparation method and its purposes in medicine
US10231953B2 (en) 2014-12-17 2019-03-19 Zenith Epigenetics Ltd. Inhibitors of bromodomains
WO2019055444A1 (en) 2017-09-13 2019-03-21 The Regents Of The University Of Michigan Bet bromodomain protein degraders with cleavable linkers
WO2019068782A1 (en) 2017-10-06 2019-04-11 Glaxosmithkline Intellectual Property (No.2) Limited 2,3-dihydrobenzofurans as boromodomain inhibitors
WO2019068783A1 (en) 2017-10-06 2019-04-11 Glaxosmithkline Intellectual Property (No.2) Limited Benzofuran derivatives and their use as bromodomain inhibitors
US10292968B2 (en) 2014-12-11 2019-05-21 Zenith Epigenetics Ltd. Substituted heterocycles as bromodomain inhibitors
US10308653B2 (en) 2014-08-08 2019-06-04 Dana-Farber Cancer Institute, Inc. Diazepane derivatives and uses thereof
US10329305B2 (en) 2015-10-29 2019-06-25 Incyte Corporation Amorphous solid form of a BET protein inhibitor
US10407441B2 (en) 2010-05-14 2019-09-10 Dana-Farber Cancer Institute, Inc. Compositions and methods for treating neoplasia, inflammatory disease and other disorders
EP3549939A1 (en) 2015-03-19 2019-10-09 GlaxoSmithKline Intellectual Property Development Limited Benzimidazole derivatives as bromodomain inhibitors
US10501438B2 (en) 2015-08-11 2019-12-10 Neomed Institute Aryl-substituted dihydroquinolinones, their preparation and their use as pharmaceuticals
US10501459B2 (en) 2015-10-21 2019-12-10 Neomed Institute Substituted imidazo[1,2-a]pyridines as bromodomain inhibitors
US10519151B2 (en) 2016-01-28 2019-12-31 Neomed Institute Substituted [1,2,4]triazolo[4,3-A]pyridines, their preparation and their use as pharmaceuticals
WO2020043821A1 (en) 2018-08-31 2020-03-05 Glaxosmithkline Intellectual Property (No.2) Limited Furan derivatives as bromodomain inhibitors
US10633379B2 (en) 2016-04-15 2020-04-28 Abbvie Inc. Bromodomain inhibitors
US10676484B2 (en) 2010-05-14 2020-06-09 Dana-Farber Cancer Institute, Inc. Compositions and methods for treating leukemia
US10702527B2 (en) 2015-06-12 2020-07-07 Dana-Farber Cancer Institute, Inc. Combination therapy of transcription inhibitors and kinase inhibitors
US10703740B2 (en) 2015-08-12 2020-07-07 Neomed Institute Substituted benzimidazoles, their preparation and their use as pharmaceuticals
US10710992B2 (en) 2014-12-01 2020-07-14 Zenith Epigenetics Ltd. Substituted pyridinones as bromodomain inhibitors
US10836742B2 (en) 2015-08-11 2020-11-17 Neomed Institute N-substituted bicyclic lactams, their preparation and their use as pharmaceuticals
US10881668B2 (en) 2015-09-11 2021-01-05 Dana-Farber Cancer Institute, Inc. Acetamide thienotriazolodiazepines and uses thereof
US10913752B2 (en) 2015-11-25 2021-02-09 Dana-Farber Cancer Institute, Inc. Bivalent bromodomain inhibitors and uses thereof
US10925881B2 (en) 2014-02-28 2021-02-23 Tensha Therapeutics, Inc. Treatment of conditions associated with hyperinsulinaemia
US10973922B2 (en) 2013-05-02 2021-04-13 Glykos Finland Oy Glycoprotein-toxic payload conjugates
WO2021175432A1 (en) 2020-03-04 2021-09-10 Boehringer Ingelheim International Gmbh Method for administration of an anti cancer agent
US11192898B2 (en) 2016-04-06 2021-12-07 The Regents Of The University Of Michigan MDM2 protein degraders
US11306105B2 (en) 2015-09-11 2022-04-19 Dana-Farber Cancer Institute, Inc. Cyano thienotriazolodiazepines and uses thereof
US11446309B2 (en) 2013-11-08 2022-09-20 Dana-Farber Cancer Institute, Inc. Combination therapy for cancer using bromodomain and extra-terminal (BET) protein inhibitors
WO2022198905A1 (en) * 2021-03-26 2022-09-29 浙江海正药业股份有限公司 Tricyclic derivative and preparation method therefor and use thereof
WO2023034290A1 (en) * 2021-08-31 2023-03-09 Incyte Corporation Naphthyridine compounds as inhibitors of kras
WO2023041744A1 (en) 2021-09-17 2023-03-23 Institut Curie Bet inhibitors for treating pab1 deficient cancer
US11666580B2 (en) 2015-08-10 2023-06-06 Dana-Farber Cancer Institute, Inc. Mechanism of resistance to bet bromodomain inhibitors
US11673889B2 (en) 2019-09-23 2023-06-13 Nanjing Zhengxiang Pharmaceuticals Co., Ltd. Substituted imidazo[4,5-c][1,8]naphthyridines as phosphodiesterase inhibitors
WO2023205251A1 (en) 2022-04-19 2023-10-26 Nuevolution A/S Compounds active towards bromodomains
US11833155B2 (en) 2020-06-03 2023-12-05 Incyte Corporation Combination therapy for treatment of myeloproliferative neoplasms

Families Citing this family (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2011340167A1 (en) * 2010-12-06 2013-07-18 Piramal Enterprises Limited Substituted imidazoquinoline derivatives
US9675697B2 (en) 2013-03-11 2017-06-13 The Regents Of The University Of Michigan BET bromodomain inhibitors and therapeutic methods using the same
EP2968311A4 (en) * 2013-03-15 2016-07-20 Epigenetix Inc Oxazolo[5,4-c]quinolin-2-one compounds as bromodomain inhibitors
US9199988B2 (en) * 2013-03-27 2015-12-01 Boehringer Ingelheim International Gmbh Dihydroquinazolinone analogues
TWI530499B (en) 2013-03-28 2016-04-21 吉李德科學股份有限公司 Benzimidazolone derivatives as bromodomain inhibitors
TWI527811B (en) 2013-05-09 2016-04-01 吉李德科學股份有限公司 Benzimidazole derivatives as bromodomain inhibitors
US8975417B2 (en) * 2013-05-27 2015-03-10 Novartis Ag Pyrazolopyrrolidine derivatives and their use in the treatment of disease
EA028175B1 (en) * 2013-05-28 2017-10-31 Новартис Аг Pyrazolo-pyrrolidin-4-one derivatives as bet inhibitors and their use in the treatment of disease
US9108953B2 (en) 2013-11-26 2015-08-18 Gilead Sciences, Inc. Quinoline derivatives as bromodomain inhibitors
AU2015282627B2 (en) 2014-06-30 2020-04-02 Glykos Finland Oy Saccharide derivative of a toxic payload and antibody conjugates thereof
WO2018022802A1 (en) 2016-07-26 2018-02-01 University Of Southern California Selective bromodomain inhibition of fungal bdf1
WO2018026877A1 (en) 2016-08-05 2018-02-08 Calitor Sciences, Llc Process for preparing substituted quinolin-4-ol compounds
KR20220125287A (en) * 2020-01-10 2022-09-14 인사이트 코포레이션 Tricyclic compounds as KRAS inhibitors
US11530218B2 (en) 2020-01-20 2022-12-20 Incyte Corporation Spiro compounds as inhibitors of KRAS
IL297165A (en) * 2020-04-16 2022-12-01 Incyte Corp Fused tricyclic kras inhibitors
US11739102B2 (en) 2020-05-13 2023-08-29 Incyte Corporation Fused pyrimidine compounds as KRAS inhibitors
US11767320B2 (en) 2020-10-02 2023-09-26 Incyte Corporation Bicyclic dione compounds as inhibitors of KRAS
EP4236942A1 (en) * 2020-10-27 2023-09-06 Arena Pharmaceuticals, Inc. Isoxazole derivatives as modulators of the 5-ht2a serotonin receptor useful for the treatment of disorders related thereto
CA3235146A1 (en) 2021-10-14 2023-04-20 Incyte Corporation Quinoline compounds as inhibitors of kras

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB2242134A (en) 1990-03-02 1991-09-25 Glaxo Group Ltd Inhalation device
US20040147543A1 (en) * 2002-12-20 2004-07-29 3M Innovative Properties Company Aryl substituted imidazoquinolines
WO2005044354A1 (en) 2003-11-03 2005-05-19 Glaxo Group Limited A fluid dispensing device
JP2008156311A (en) 2006-12-26 2008-07-10 Institute Of Physical & Chemical Research Brd2 bromodomain binder
WO2009084693A1 (en) 2007-12-28 2009-07-09 Mitsubishi Tanabe Pharma Corporation Antitumor agent

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB0510390D0 (en) * 2005-05-20 2005-06-29 Novartis Ag Organic compounds
MX2008007864A (en) * 2005-12-16 2009-03-04 Coley Pharm Group Inc Substituted imidazoquinolines, imidazonaphthyridines, and imidazopyridines, compositions, and methods.

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB2242134A (en) 1990-03-02 1991-09-25 Glaxo Group Ltd Inhalation device
US20040147543A1 (en) * 2002-12-20 2004-07-29 3M Innovative Properties Company Aryl substituted imidazoquinolines
WO2005044354A1 (en) 2003-11-03 2005-05-19 Glaxo Group Limited A fluid dispensing device
JP2008156311A (en) 2006-12-26 2008-07-10 Institute Of Physical & Chemical Research Brd2 bromodomain binder
WO2009084693A1 (en) 2007-12-28 2009-07-09 Mitsubishi Tanabe Pharma Corporation Antitumor agent

Non-Patent Citations (12)

* Cited by examiner, † Cited by third party
Title
"Burger's Medicinal Chemistry and Drug Discovery", vol. 1
BERGE ET AL., J. PHARM. SCI., vol. 66, 1977, pages 1 - 19
FRENCH ET AL., CANCER RESEARCH, vol. 63, 2003, pages 304 - 307
FRENCH ET AL., JOURNAL OF CLINICAL ONCOLOGY, vol. 22, no. 20, 2004, pages 4135 - 4139
HARGREAVES ET AL., CELL, vol. 138, no. 1, 2009, pages 129 - 145
HARUSAWA, S ET AL., HETEROCYCLES, vol. 15, 1981, pages 981 - 984
INT. J. PEPTIDE PROTEIN RES., vol. 40, 1992, pages 13 - 18
J.MED.CHEM., vol. 23, 1980, pages 1358
LEROY ET AL., MOL. CELL, vol. 30, no. 1, 2008, pages 51 - 60
ONCOGENE, vol. 27, 2008, pages 2237 - 2242
T. W. GREENE: "Protective Groups in Organic Synthesis", 2006, J. WILEY AND SONS
YOU ET AL., CELL, vol. 117, no. 3, 2004, pages 349 - 60

Cited By (221)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10676484B2 (en) 2010-05-14 2020-06-09 Dana-Farber Cancer Institute, Inc. Compositions and methods for treating leukemia
US10407441B2 (en) 2010-05-14 2019-09-10 Dana-Farber Cancer Institute, Inc. Compositions and methods for treating neoplasia, inflammatory disease and other disorders
US9789120B2 (en) 2010-05-14 2017-10-17 Dana-Farber Cancer Institute, Inc. Male contraceptive compositions and methods of use
EP2635279A4 (en) * 2010-11-05 2014-10-29 Glaxosmithkline Ip No 2 Ltd Chemical compounds
EP2635279A2 (en) * 2010-11-05 2013-09-11 Glaxosmithkline Intellectual Property (No. 2) Limited Chemical compounds
US9522920B2 (en) 2010-12-02 2016-12-20 Constellation Pharmaceuticals, Inc. Bromodomain inhibitors and uses thereof
US9249161B2 (en) 2010-12-02 2016-02-02 Constellation Pharmaceuticals, Inc. Bromodomain inhibitors and uses thereof
US8796261B2 (en) 2010-12-02 2014-08-05 Constellation Pharmaceuticals, Inc. Bromodomain inhibitors and uses thereof
US9422292B2 (en) 2011-05-04 2016-08-23 Constellation Pharmaceuticals, Inc. Bromodomain inhibitors and uses thereof
US9328117B2 (en) 2011-06-17 2016-05-03 Constellation Pharmaceuticals, Inc. Bromodomain inhibitors and uses thereof
EP3150601A1 (en) * 2011-08-17 2017-04-05 Glaxosmithkline LLC 4-(8-methoxy-1-((1-methoxypropan-2-yl)-2-(tetrahydro-2h-pyran-4-yl)-1 h-imidazo[4,5-c]quinolin-7-yl)-3,5-dimethylisoxazole and its use as bromodomain inhibitor
JP2014521735A (en) * 2011-08-17 2014-08-28 グラクソスミスクライン・リミテッド・ライアビリティ・カンパニー 4- (8-Methoxy-1-((1-methoxypropan-2-yl) -2- (tetrahydro-2H-pyran-4-yl) -1H-imidazo [4,5-c] quinolin-7-yl ) -3,5-dimethylisoxazole and its use as a bromodomain inhibitor
CN103889984A (en) * 2011-08-17 2014-06-25 葛兰素史克有限责任公司 4-(8-methoxy-1-((1-methoxypropan-2-yl)-2-(tetrahydro-2h-pyran-4-yl)-1 h-imidazo[4,5-c]quinolin-7-yl)-3,5-dimethylisoxazole and its use as bromodomain inhibitor
US9861627B2 (en) 2011-08-17 2018-01-09 Glaxosmithkline Llc 4-(8-methoxy-1-((1-methoxypropan-2-yl)-2-(tetrahydro-2H-pyran-4-yl)-1 H-imidazo[4,5-C]quinolin-7-yl)-3,5-dimethylisoxazole and its use as bromodomain inhibitor
US9416130B2 (en) 2011-08-17 2016-08-16 Glaxosmithkline Llc 4-(8-methoxy-1-((1-methoxypropan-2-yl)-2-(tetrahydro-2H-pyran-4-yl)-1 H-imidazo[4,5-C]quinolin-7-yl)-3,5-dimethylisoxazole and its use as bromodomain inhibitor
AU2012296914B2 (en) * 2011-08-17 2016-03-03 Glaxosmithkline Llc 4-(8-methoxy-1-((1-methoxypropan-2-yl)-2-(tetrahydro-2H-pyran-4-yl)-1 H-imidazo(4,5-c)quinolin-7-yl)-3,5-dimethylisoxazole and its use as bromodomain inhibitor
EA023374B1 (en) * 2011-08-17 2016-05-31 ГЛАКСОСМИТКЛАЙН ЭлЭлСи 4-(8-METHOXY-1-((1-METHOXYPROPAN-2-YL)-2-(TETRAHYDRO-2H-PYRAN-4-YL)-1H-IMIDAZO[4,5-c]QUINOLIN-7-YL)-3,5-DIMETHYLISOXAZOLE AND ITS USE AS BROMODOMAIN INHIBITOR
US9610284B2 (en) 2011-08-17 2017-04-04 Glaxosmithkline Llc 4-(8-methoxy-1-((1-methoxypropan-2-yl)-2-(tetrahydro-2H-pyran-4-yl)-1H-imidazo[4,5-c]quinolin-7-yl)-3,5-dimethylisoxazole and its use as bromodomain inhibitor
US9067936B2 (en) 2011-08-17 2015-06-30 Glaxosmithkline Llc 4-(8-methoxy-1-((1-methoxypropan-2-yl)-2-(tetrahydro-2H-pyran-4-yl)-1 H-imidazo[4,5-C]quinolin-7-yl)-3,5-dimethylisoxazole and its use as bromodomain inhibitor
WO2013024104A1 (en) 2011-08-17 2013-02-21 Glaxosmithkline Llc 4-(8-methoxy-1-((1-methoxypropan-2-yl)-2-(tetrahydro-2h-pyran-4-yl)-1 h-imidazo[4,5-c]quinolin-7-yl)-3,5-dimethylisoxazole and its use as bromodomain inhibitor
TWI558704B (en) * 2011-08-17 2016-11-21 葛蘭素史密斯克藍有限責任公司 4-(8-methoxy-1-(1-methoxypropan-2-yl)-2-(tetrahydro-2h-pyran-4-yl)-1h-imidazo[4,5-c]quinolin-7-yl)-3,5-dimethylisoxazole and use thereof
WO2013027168A1 (en) 2011-08-22 2013-02-28 Pfizer Inc. Novel heterocyclic compounds as bromodomain inhibitors
WO2013033269A1 (en) * 2011-08-29 2013-03-07 Coferon, Inc. Bioorthogonal monomers capable of dimerizing and targeting bromodomains and methods of using same
JP2014531458A (en) * 2011-09-30 2014-11-27 シー・アンド・シー・リサーチ・ラボラトリーズC&C Research Laboratories Novel heterocyclic derivatives and uses thereof
US9586959B2 (en) 2011-09-30 2017-03-07 C&C Research Laboratories Substituted tricyclic heterocycles as histamine 4 receptor inhibitors
US9296741B2 (en) 2011-12-30 2016-03-29 Abbvie Inc. Bromodomain inhibitors
US9776990B2 (en) 2012-04-20 2017-10-03 Abbvie Inc. Isoindolone derivatives
US9493483B2 (en) 2012-06-06 2016-11-15 Constellation Pharmaceuticals, Inc. Benzo [C] isoxazoloazepine bromodomain inhibitors and uses thereof
US9925197B2 (en) 2012-06-06 2018-03-27 Constellation Pharmaceuticals, Inc. Benzo [C] isoxazoloazepine bromodomain inhibitors and uses thereof
US9624244B2 (en) 2012-06-06 2017-04-18 Constellation Pharmaceuticals, Inc. Benzo [B] isoxazoloazepine bromodomain inhibitors and uses thereof
US9561231B2 (en) 2012-06-12 2017-02-07 Abbvie Inc. Pyridinone and pyridazinone derivatives
US9763956B2 (en) 2012-06-19 2017-09-19 The Broad Institute, Inc. Diagnostic and treatment methods in subjects having or at risk of developing resistance to cancer therapy
US9422290B2 (en) 2012-11-13 2016-08-23 Boehringer Ingelheim International Gmbh Triazolopyridazine
WO2014076146A1 (en) 2012-11-13 2014-05-22 Boehringer Ingelheim International Gmbh Triazolopyridazine
WO2014076237A1 (en) 2012-11-16 2014-05-22 Boehringer Ingelheim International Gmbh Triazolopyrazine
US10328074B2 (en) 2012-11-16 2019-06-25 Boehringer Ingelheim International Gmbh Substituted [1,2,4]triazolo[4,3-a]pyrazines as BRD4 inhibitors
US11077107B2 (en) 2012-11-16 2021-08-03 Boehringer Ingelheim International Gmbh Triazolopyrazine
US9278940B2 (en) 2012-11-21 2016-03-08 Zenith Epigenetics Corp. Cyclic amines as bromodomain inhibitors
US9765039B2 (en) 2012-11-21 2017-09-19 Zenith Epigenetics Ltd. Biaryl derivatives as bromodomain inhibitors
US9271978B2 (en) 2012-12-21 2016-03-01 Zenith Epigenetics Corp. Heterocyclic compounds as bromodomain inhibitors
US9598367B2 (en) 2012-12-21 2017-03-21 Zenith Epigenetics Ltd. Heterocyclic compounds as bromodomain inhibitors
US9861637B2 (en) 2012-12-21 2018-01-09 Zenith Epigenetics Ltd. Heterocyclic compounds as bromodomain inhibitors
WO2014128655A1 (en) * 2013-02-25 2014-08-28 Aurigene Discovery Technologies Limited Substituted imidazo[4,5-c]quinoline derivatives as bromodomain inhibitors
AU2014244426B2 (en) * 2013-03-14 2019-02-28 Convergene Llc Methods and compositions for inhibition of bromodomain-containing proteins
US9695179B2 (en) 2013-03-14 2017-07-04 Convergene Llc Methods and compositions for inhibition of bromodomain-containing proteins
US9714946B2 (en) 2013-03-14 2017-07-25 Dana-Farber Cancer Institute, Inc. Bromodomain binding reagents and uses thereof
EP2968360A4 (en) * 2013-03-14 2017-03-08 ConverGene LLC Methods and compositions for inhibition of bromodomain-containing proteins
US10717739B2 (en) 2013-03-14 2020-07-21 Convergene Llc Methods and compositions for inhibition of bromodomain-containing proteins
US10266536B2 (en) 2013-03-14 2019-04-23 Convergene Llc Methods and compositions for inhibition of bromodomain-containing proteins
WO2014140077A1 (en) 2013-03-14 2014-09-18 Glaxosmithkline Intellectual Property (No.2) Limited Furopyridines as bromodomain inhibitors
CN105407894A (en) * 2013-03-14 2016-03-16 康威基内有限公司 Methods and compositions for inhibition of bromodomain-containing proteins
US11498926B2 (en) 2013-03-15 2022-11-15 Incyte Corporation Tricyclic heterocycles as BET protein inhibitors
US10464947B2 (en) 2013-03-15 2019-11-05 Incyte Holdings Corporation Tricyclic heterocycles as BET protein inhibitors
US9938294B2 (en) 2013-03-15 2018-04-10 Incyte Corporation Tricyclic heterocycles as BET protein inhibitors
US10919912B2 (en) 2013-03-15 2021-02-16 Incyte Holdings Corporation Tricyclic heterocycles as BET protein inhibitors
US9227985B2 (en) 2013-03-15 2016-01-05 Incyte Corporation Tricyclic heterocycles as bet protein inhibitors
US9624241B2 (en) 2013-03-15 2017-04-18 Incyte Corporation Tricyclic heterocycles as BET protein inhibitors
WO2014154760A1 (en) * 2013-03-27 2014-10-02 Boehringer Ingelheim International Gmbh Indolinone analogues as brd4 inhibitors
US9102684B2 (en) 2013-03-27 2015-08-11 Boehringer Ingelheim International Gmbh Indolinone analogues
JP2016515560A (en) * 2013-03-27 2016-05-30 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Indolinone analogs as BRD4 inhibitors
WO2014173241A1 (en) 2013-04-26 2014-10-30 Beigene, Ltd. Substituted5-(3,5-dimethylisoxazol-4-yl)indoline-2-ones
US10973922B2 (en) 2013-05-02 2021-04-13 Glykos Finland Oy Glycoprotein-toxic payload conjugates
US10166215B2 (en) 2013-06-21 2019-01-01 Zenith Epigenetics Ltd. Substituted bicyclic compounds as bromodomain inhibitors
US9663520B2 (en) 2013-06-21 2017-05-30 Zenith Epigenetics Ltd. Bicyclic bromodomain inhibitors
JP2016523259A (en) * 2013-06-21 2016-08-08 ゼニス・エピジェネティクス・コーポレイションZenith Epigenetics Corp. Novel bicyclic bromodomain inhibitors
JP2016522246A (en) * 2013-06-21 2016-07-28 ゼニス・エピジェネティクス・コーポレイションZenith Epigenetics Corp. Novel substituted bicyclic compounds as bromodomain inhibitors
US10363257B2 (en) 2013-06-21 2019-07-30 Zenith Epigenetics Ltd. Bicyclic bromodomain inhibitors
US10772892B2 (en) 2013-06-21 2020-09-15 Zenith Epigenetics Ltd. Bicyclic bromodomain inhibitors
US11446306B2 (en) 2013-06-21 2022-09-20 Zenith Epigenetics Ltd. Bicyclic bromodomain inhibitors
US11026926B2 (en) 2013-06-21 2021-06-08 Zenith Epigenetics Ltd. Substituted bicyclic compounds as bromodomain inhibitors
US10010556B2 (en) 2013-06-21 2018-07-03 Zenith Epigenetics Ltd. Bicyclic bromodomain inhibitors
US10226451B2 (en) 2013-06-21 2019-03-12 Zenith Epigenetics Ltd. Substituted bicyclic compounds as bromodomain inhibitors
US9636328B2 (en) 2013-06-21 2017-05-02 Zenith Epigenetics Ltd. Substituted bicyclic compounds as bromodomain inhibitors
US9662311B2 (en) 2013-06-21 2017-05-30 Zenith Epigenetics Ltd. Substituted bicyclic compounds as bromodomain inhibitors
US9850257B2 (en) 2013-07-08 2017-12-26 Incyte Corporation Tricyclic heterocycles as BET protein inhibitors
US9290514B2 (en) 2013-07-08 2016-03-22 Incyte Holdings Corporation Tricyclic heterocycles as BET protein inhibitors
US9533997B2 (en) 2013-07-08 2017-01-03 Incyte Holdings Corporation Tricyclic heterocycles as BET protein inhibitors
WO2015013635A2 (en) 2013-07-25 2015-01-29 Dana-Farber Cancer Institute, Inc. Inhibitors of transcription factors and uses thereof
US9975896B2 (en) 2013-07-25 2018-05-22 Dana-Farber Cancer Institute, Inc. Inhibitors of transcription factors and uses thereof
US10500209B2 (en) 2013-07-31 2019-12-10 Zenith Epigenetics Ltd. Quinazolinones as bromodomain inhibitors
US9855271B2 (en) 2013-07-31 2018-01-02 Zenith Epigenetics Ltd. Quinazolinones as bromodomain inhibitors
WO2015049629A1 (en) * 2013-10-01 2015-04-09 Piramal Enterprises Limited Imidazoquinoline compounds as bromodomain inhibitors
WO2015067770A1 (en) 2013-11-07 2015-05-14 Boehringer Ingelheim International Gmbh Triazolopyrazine derivatives as brd4 inhibitors
US11446309B2 (en) 2013-11-08 2022-09-20 Dana-Farber Cancer Institute, Inc. Combination therapy for cancer using bromodomain and extra-terminal (BET) protein inhibitors
AU2013406206B2 (en) * 2013-11-26 2017-07-20 Gilead Sciences, Inc. Quinoline derivatives as bromodomain inhibitors
US9315501B2 (en) 2013-11-26 2016-04-19 Incyte Corporation Bicyclic heterocycles as BET protein inhibitors
US9399640B2 (en) 2013-11-26 2016-07-26 Incyte Corporation Substituted pyrrolo[2,3-c]pyridines and pyrazolo[3,4-c]pyridines as BET protein inhibitors
US9737516B2 (en) 2013-11-26 2017-08-22 Incyte Corporation Bicyclic heterocycles as bet protein inhibitors
WO2015080707A1 (en) * 2013-11-26 2015-06-04 Gilead Sciences, Inc. Quinoline derivatives as bromodomain inhibitors
US9918990B2 (en) 2013-11-26 2018-03-20 Incyte Corporation Substituted pyrrolo[2,3-c]pyridines and pyrazolo[3,4-c]pyridines as BET protein inhibitors
WO2015092118A1 (en) 2013-12-17 2015-06-25 Orion Corporation Spiro[cyclobutane-1,3'-indolin]-2'-one derivatives as bromodomain inhibitors
US9777003B2 (en) 2013-12-19 2017-10-03 Incyte Corporation Tricyclic heterocycles as bet protein inhibitors
US11091484B2 (en) 2013-12-19 2021-08-17 Incyte Corporation Tricyclic heterocycles as BET protein inhibitors
US10442803B2 (en) 2013-12-19 2019-10-15 Incyte Corporation Tricyclic heterocycles as bet protein inhibitors
US9309246B2 (en) 2013-12-19 2016-04-12 Incyte Corporation Tricyclic heterocycles as BET protein inhibitors
WO2015104653A1 (en) 2014-01-09 2015-07-16 Aurigene Discovery Technologies Limited Bicyclic heterocyclic derivatives as bromodomain inhibitors
US10077259B2 (en) 2014-01-09 2018-09-18 Orion Corporation Bicyclic heterocyclic derivatives as bromodomain inhibitors
WO2015117087A1 (en) 2014-01-31 2015-08-06 Dana-Farber Cancer Institute, Inc. Uses of diazepane derivatives
US10793571B2 (en) 2014-01-31 2020-10-06 Dana-Farber Cancer Institute, Inc. Uses of diazepane derivatives
US10150756B2 (en) 2014-01-31 2018-12-11 Dana-Farber Cancer Institute, Inc. Diaminopyrimidine benzenesulfone derivatives and uses thereof
US10730860B2 (en) 2014-01-31 2020-08-04 Dana-Farber Cancer Institute, Inc. Diaminopyrimidine benzenesulfone derivatives and uses thereof
US9695172B2 (en) 2014-01-31 2017-07-04 Dana-Farber Cancer Institute, Inc. Diazepane derivatives and uses thereof
WO2015131005A1 (en) 2014-02-28 2015-09-03 The Regents Of The University Of Michigan 9h-pyrimido[4,5-b]indoles and related analogs as bet bromodomain inhibitors
US10925881B2 (en) 2014-02-28 2021-02-23 Tensha Therapeutics, Inc. Treatment of conditions associated with hyperinsulinaemia
US9957268B2 (en) 2014-04-23 2018-05-01 Incyte Corporation 1H-pyrrolo[2,3,c]pyridin-7(6H)-ones and pyrazolo[3,4-c]pyridin-7(6H)-ones as inhibitors of BET proteins
US11702416B2 (en) 2014-04-23 2023-07-18 Incyte Corporation 1H-pyrrolo[2,3-c]pyridin-7(6H)-ones and pyrazolo[3,4-c]pyridin-7(6H)-ones as inhibitors of BET proteins
US10472358B2 (en) 2014-04-23 2019-11-12 Incyte Corporation 1H-pyrrolo[2,3-c]pyridin-7(6H)-ones and pyrazolo[3,4-c]pyridin-7(6H)-ones as inhibitors of BET proteins
US9540368B2 (en) 2014-04-23 2017-01-10 Incyte Corporation 1H-pyrrolo[2,3-c]pyridin-7(6H)-ones and pyrazolo[3,4-c]pyridin-7(6H)-ones as inhibitors of BET proteins
US11059821B2 (en) 2014-04-23 2021-07-13 Incyte Corporation 1H-pyrrolo[2,3-c]pyridin-7(6H)-ones and pyrazolo[3,4-c]pyridin-7(6H)-ones as inhibitors of BET proteins
US10781209B2 (en) 2014-04-23 2020-09-22 Incyte Corporation 1H-pyrrolo[2,3-c]pyridin-7(6H)-ones and pyrazolo[3,4-c]pyridin-7(6H)-ones as inhibitors of BET proteins
US10189834B2 (en) 2014-05-08 2019-01-29 Astrazeneca Ab Imidazo[4,5-c]quinolin-2-one compounds and their use in treating cancer
WO2015169962A1 (en) * 2014-05-09 2015-11-12 Boehringer Ingelheim International Gmbh Benzimidazole derivatives
US9428515B2 (en) 2014-05-09 2016-08-30 Boehringer Ingelheim International Gmbh Benzimidazole derivatives
US9969747B2 (en) 2014-06-20 2018-05-15 Constellation Pharmaceuticals, Inc. Crystalline forms of 2-((4S)-6-(4-chlorophenyl)-1-methyl-4H-benzo[C]isoxazolo[4,5-e]azepin-4-yl)acetamide
US9951074B2 (en) 2014-08-08 2018-04-24 Dana-Farber Cancer Institute, Inc. Dihydropteridinone derivatives and uses thereof
US10308653B2 (en) 2014-08-08 2019-06-04 Dana-Farber Cancer Institute, Inc. Diazepane derivatives and uses thereof
US10618910B2 (en) 2014-09-15 2020-04-14 Incyte Corporation Tricyclic heterocycles as BET protein inhibitors
US10227359B2 (en) 2014-09-15 2019-03-12 Incyte Corporation Tricyclic heterocycles as bet protein inhibitors
US9834565B2 (en) 2014-09-15 2017-12-05 Incyte Corporation Tricyclic heterocycles as bet protein inhibitors
US9527864B2 (en) 2014-09-15 2016-12-27 Incyte Corporation Tricyclic heterocycles as BET protein inhibitors
US10124009B2 (en) 2014-10-27 2018-11-13 Tensha Therapeutics, Inc. Bromodomain inhibitors
US10710992B2 (en) 2014-12-01 2020-07-14 Zenith Epigenetics Ltd. Substituted pyridinones as bromodomain inhibitors
US10179125B2 (en) 2014-12-01 2019-01-15 Zenith Epigenetics Ltd. Substituted pyridines as bromodomain inhibitors
US10292968B2 (en) 2014-12-11 2019-05-21 Zenith Epigenetics Ltd. Substituted heterocycles as bromodomain inhibitors
US10231953B2 (en) 2014-12-17 2019-03-19 Zenith Epigenetics Ltd. Inhibitors of bromodomains
US10259809B2 (en) 2015-02-03 2019-04-16 Trillium Therapeutics Inc. Fluorinated imidazo[4,5-C]quinoline derivatives as inhibitors of bromodomain containing proteins
WO2016123709A1 (en) 2015-02-03 2016-08-11 Trillium Therapeutics Inc. Fluorinated imidazo[4,5-c]quinoline derivatives as inhibitors of bromodomain containing proteins
US10696670B2 (en) 2015-02-03 2020-06-30 Trillium Therapeutics Inc. Fluorinated imidazo[4,5-C]quinoline derivatives as inhibitors of bromodomain containing proteins
WO2016138332A1 (en) 2015-02-27 2016-09-01 The Regents Of The University Of Michigan 9h-pyrimido [4,5-b] indoles as bet bromodomain inhibitors
US10307407B2 (en) 2015-02-27 2019-06-04 The Regents Of The University Of Michigan 9H-pyrimido [4,5-B] indoles as BET bromodomain inhibitors
WO2016146755A1 (en) 2015-03-19 2016-09-22 Glaxosmithkline Intellectual Property Development Limited Covalent conjugates of bet inhibitors and alpha amino acid esters
EP3549939A1 (en) 2015-03-19 2019-10-09 GlaxoSmithKline Intellectual Property Development Limited Benzimidazole derivatives as bromodomain inhibitors
EP3868761A1 (en) * 2015-04-02 2021-08-25 Merck Patent GmbH Imidazolonylchinolines and their use as atm kinase inhibitors
EP3560924A1 (en) * 2015-04-02 2019-10-30 Merck Patent GmbH Imidazolonyl quinolines and their use as atm kinase inhibitors
KR102652052B1 (en) 2015-04-02 2024-03-27 메르크 파텐트 게엠베하 Imidazolonyl quinolines and use thereof as atm kinase inhibitors
CN107889488A (en) * 2015-04-02 2018-04-06 默克专利股份公司 Imidazoles ketone group quinoline and its purposes as ATM kinase inhibitors
KR20170132323A (en) * 2015-04-02 2017-12-01 메르크 파텐트 게엠베하 Imidazolonyl quinolines and use thereof as atm kinase inhibitors
RU2743343C2 (en) * 2015-04-02 2021-02-17 Мерк Патент Гмбх Imidazolonyl quinolines and use thereof as kinase atm inhibitors
AU2016239270B2 (en) * 2015-04-02 2020-03-26 Merck Patent Gmbh Imidazolonyl quinolines and use thereof as ATM kinase inhibitors
US11608338B2 (en) 2015-04-02 2023-03-21 Merck Patent Gmbh Imidazolonylquinolines and the use thereof as ATM kinase inhibitors
AU2020204241B2 (en) * 2015-04-02 2022-11-17 Merck Patent Gmbh Imidazolonyl quinolines and use thereof as ATM kinase inhibitors
CN111689963A (en) * 2015-04-02 2020-09-22 默克专利股份公司 Imidazoquinolinones and their use as ATM kinase inhibitors
CN111747952A (en) * 2015-04-02 2020-10-09 默克专利股份公司 Imidazoquinolinones and their use as ATM kinase inhibitors
US10457677B2 (en) 2015-04-02 2019-10-29 Merck Patent Gmbh Imidazolonylquinolines and the use thereof as ATM kinase inhibitors
US10975075B2 (en) 2015-04-02 2021-04-13 Merck Patent Gmbh Imidazolonylquinolines and the use thereof as ATM kinase inhibitors
WO2016155884A1 (en) * 2015-04-02 2016-10-06 Merck Patent Gmbh Imidazolonyl quinolines and use thereof as atm kinase inhibitors
US10745399B2 (en) 2015-04-02 2020-08-18 Merck Patent Gmbh Imidazolonylquinolines and the use thereof as ATM kinase inhibitors
CN107889488B (en) * 2015-04-02 2020-08-11 默克专利股份公司 Imidazoquinolinones and their use as ATM kinase inhibitors
WO2016186453A1 (en) * 2015-05-20 2016-11-24 Kainos Medicine, Inc. Quinoline derivatives as bromodomain inhibitors
WO2016196065A1 (en) 2015-05-29 2016-12-08 Genentech, Inc. Methods and compositions for assessing responsiveness of cancers to bet inhibitors
WO2016199943A1 (en) * 2015-06-11 2016-12-15 Takeda Pharmaceutical Company Limited Heterocyclic compounds
US10702527B2 (en) 2015-06-12 2020-07-07 Dana-Farber Cancer Institute, Inc. Combination therapy of transcription inhibitors and kinase inhibitors
WO2016203112A1 (en) 2015-06-16 2016-12-22 Orion Corporation Spiro[cyclobutane-1,3'-indolin]-2'-one derivatives as bromodomain inhibitors
WO2016203335A1 (en) 2015-06-18 2016-12-22 Pfizer Inc. Novel pyrido[2,3-b]pyrazinones as bet-family bromodomain inhibitors
WO2016210275A1 (en) 2015-06-26 2016-12-29 Tensha Therapeutics, Inc. Treatment of nut midline carcinoma
WO2017001733A1 (en) 2015-07-02 2017-01-05 Orion Corporation Bicyclic heterocycle derivatives as bromodomain inhibitors
US11666580B2 (en) 2015-08-10 2023-06-06 Dana-Farber Cancer Institute, Inc. Mechanism of resistance to bet bromodomain inhibitors
US10501438B2 (en) 2015-08-11 2019-12-10 Neomed Institute Aryl-substituted dihydroquinolinones, their preparation and their use as pharmaceuticals
US10836742B2 (en) 2015-08-11 2020-11-17 Neomed Institute N-substituted bicyclic lactams, their preparation and their use as pharmaceuticals
US11365186B2 (en) 2015-08-12 2022-06-21 Epigenetix, Inc. Substituted benzimidazoles, their preparation and their use as pharmaceuticals
US10703740B2 (en) 2015-08-12 2020-07-07 Neomed Institute Substituted benzimidazoles, their preparation and their use as pharmaceuticals
US10881668B2 (en) 2015-09-11 2021-01-05 Dana-Farber Cancer Institute, Inc. Acetamide thienotriazolodiazepines and uses thereof
US11306105B2 (en) 2015-09-11 2022-04-19 Dana-Farber Cancer Institute, Inc. Cyano thienotriazolodiazepines and uses thereof
US11406645B2 (en) 2015-09-11 2022-08-09 Dana-Farber Cancer Institute, Inc. Acetamide thienotriazolodiazepines and uses thereof
US10882858B2 (en) 2015-09-17 2021-01-05 Astrazeneca Ab Imidazo[4,5-c]quinolin-2-one compounds and their use in treating cancer
US9856255B2 (en) 2015-09-17 2018-01-02 Astrazeneca Ab Imidazo[4,5-c]quinolin-2-one compounds and their use in treating cancer
US10457679B2 (en) 2015-09-17 2019-10-29 Astrazeneca Ab Imidazo[4,5-c]quinolin-2-one compounds and their use in treating cancer
US11613539B2 (en) 2015-09-17 2023-03-28 Astrazeneca Ab Imidazo[4,5-c]quinolin-2-one compounds and their use in treating cancer
WO2017050714A1 (en) 2015-09-22 2017-03-30 Glaxosmithkline Intellectual Property (No.2) Limited Pyridinone dicarboxamide for use as bromodomain inhibitors
US10501459B2 (en) 2015-10-21 2019-12-10 Neomed Institute Substituted imidazo[1,2-a]pyridines as bromodomain inhibitors
US10858372B2 (en) 2015-10-29 2020-12-08 Incyte Corporation Amorphous solid form of a BET protein inhibitor
US10329305B2 (en) 2015-10-29 2019-06-25 Incyte Corporation Amorphous solid form of a BET protein inhibitor
US10913752B2 (en) 2015-11-25 2021-02-09 Dana-Farber Cancer Institute, Inc. Bivalent bromodomain inhibitors and uses thereof
EP4050005A1 (en) * 2015-11-25 2022-08-31 Convergene Llc Bicyclic bet bromodomain inhibitors and uses thereof
CN109641886A (en) * 2015-11-25 2019-04-16 康威基内有限公司 Bicyclic BET Bu Luomo structural domain inhibitor and application thereof
WO2017091661A1 (en) * 2015-11-25 2017-06-01 Strovel Jeffrey William Bicyclic bet bromodomain inhibitors and uses thereof
US11028079B2 (en) 2015-11-25 2021-06-08 Convergene, Llc Small molecule BET bromodomain inhibitors and uses thereof
US10508106B2 (en) 2015-11-25 2019-12-17 Convergene Llc Bicyclic BET bromodomain inhibitors and uses thereof
CN109641886B (en) * 2015-11-25 2022-11-18 康威基内有限公司 Bicyclic BET bromodomain inhibitors and uses thereof
US10519151B2 (en) 2016-01-28 2019-12-31 Neomed Institute Substituted [1,2,4]triazolo[4,3-A]pyridines, their preparation and their use as pharmaceuticals
US11548899B2 (en) 2016-02-15 2023-01-10 The Regents Of The University Of Michigan Fused 1,4-oxazepines and related analogs as BET bromodomain inhibitors
WO2017142881A1 (en) 2016-02-15 2017-08-24 The Regents Of The University Of Michigan Fused 1,4-oxazepines and related analogs as bet bromodomain inhibitors
WO2017176958A1 (en) 2016-04-06 2017-10-12 The Regents Of The University Of Michigan Monofunctional intermediates for ligand-dependent target protein degradation
US11192898B2 (en) 2016-04-06 2021-12-07 The Regents Of The University Of Michigan MDM2 protein degraders
WO2017174620A1 (en) 2016-04-07 2017-10-12 Glaxosmithkline Intellectual Property (No.2) Limited Benzo[b]furans as bromodomain inhibitors
WO2017174621A1 (en) 2016-04-07 2017-10-12 Glaxosmithkline Intellectual Property (No.2) Limited Pyridyl derivatives as bromodomain inhibitors
US10633386B2 (en) 2016-04-12 2020-04-28 The Regents Of The University Of Michigan BET protein degraders
WO2017180417A1 (en) 2016-04-12 2017-10-19 The Regents Of The University Of Michigan Bet protein degraders
US10633379B2 (en) 2016-04-15 2020-04-28 Abbvie Inc. Bromodomain inhibitors
WO2017202742A1 (en) 2016-05-24 2017-11-30 Glaxosmithkline Intellectual Property (No.2) Limited Pyridine dicarboxamide derivatives as bromodomain inhibitors
US11377446B2 (en) 2016-06-20 2022-07-05 Incyte Corporation Crystalline solid forms of a BET inhibitor
US11091480B2 (en) 2016-06-20 2021-08-17 Incyte Corporation Crystalline solid forms of a BET inhibitor
US10189832B2 (en) 2016-06-20 2019-01-29 Incyte Corporation Crystalline solid forms of a BET inhibitor
US10626114B2 (en) 2016-06-20 2020-04-21 Incyte Corporation Crystalline solid forms of a BET inhibitor
WO2018041946A1 (en) 2016-09-02 2018-03-08 Glaxosmithkline Intellectual Property Development Limited Crystalline hydrate of the compound (2s,3r)-isopropyl 2-(((2-(1,5-dimethyl-6-oxo-1,6-dihydropyridin-3-yl)-1-((tetrahydro-2h-pyran-4-yl)methyl)-1h-benzo[d]imidazol-5-yl)methyl)amino)-3-hydroxybutanoate edisylate
WO2018041947A1 (en) 2016-09-02 2018-03-08 Glaxosmithkline Intellectual Property (No.2) Limited Imidazole derivatives and their use in the treatment of autoimmune or inflammatory diseases or cancers
WO2018041964A1 (en) 2016-09-02 2018-03-08 Glaxosmithkline Intellectual Property (No.2) Limited Imidazole derivatives and their use in the treatment of autoimmune or inflammatory diseases or cancers
WO2018052949A1 (en) 2016-09-13 2018-03-22 The Regents Of The University Of Michigan Fused 1,4-diazepines as bet protein degraders
US10975093B2 (en) 2016-09-13 2021-04-13 The Regents Of The University Of Michigan Fused 1,4-diazepines as BET protein degraders
US11466028B2 (en) 2016-09-13 2022-10-11 The Regents Of The University Of Michigan Fused 1,4-oxazepines as BET protein degraders
EP3858837A1 (en) 2016-09-13 2021-08-04 The Regents of The University of Michigan Fused 1,4-diazepines as bet protein degraders
WO2018052945A1 (en) 2016-09-13 2018-03-22 The Regents Of The University Of Michigan Fused 1,4-oxazepines as bet protein degraders
WO2018109271A1 (en) 2016-12-13 2018-06-21 Orion Corporation New bromodomain inhibitors
WO2018144789A1 (en) 2017-02-03 2018-08-09 The Regents Of The University Of Michigan Fused 1,4-diazepines as bet bromodomain inhibitors
US11046709B2 (en) 2017-02-03 2021-06-29 The Regents Of The University Of Michigan Fused 1,4-diazepines as BET bromodomain inhibitors
WO2018158210A1 (en) 2017-03-01 2018-09-07 Glaxosmithkline Intellectual Property (N°.2) Limited Pyridyl derivatives as bromodomain inhibitors
CN109384784A (en) * 2017-08-10 2019-02-26 浙江海正药业股份有限公司 Sulfonic acid amide derivatives, preparation method and its purposes in medicine
CN109384784B (en) * 2017-08-10 2021-01-12 浙江海正药业股份有限公司 Sulfonamide derivative, preparation method thereof and application thereof in medicine
US11267822B2 (en) 2017-09-13 2022-03-08 The Regents Of The University Of Michigan BET bromodomain protein degraders with cleavable linkers
WO2019055444A1 (en) 2017-09-13 2019-03-21 The Regents Of The University Of Michigan Bet bromodomain protein degraders with cleavable linkers
WO2019068782A1 (en) 2017-10-06 2019-04-11 Glaxosmithkline Intellectual Property (No.2) Limited 2,3-dihydrobenzofurans as boromodomain inhibitors
WO2019068783A1 (en) 2017-10-06 2019-04-11 Glaxosmithkline Intellectual Property (No.2) Limited Benzofuran derivatives and their use as bromodomain inhibitors
WO2020043821A1 (en) 2018-08-31 2020-03-05 Glaxosmithkline Intellectual Property (No.2) Limited Furan derivatives as bromodomain inhibitors
US11673889B2 (en) 2019-09-23 2023-06-13 Nanjing Zhengxiang Pharmaceuticals Co., Ltd. Substituted imidazo[4,5-c][1,8]naphthyridines as phosphodiesterase inhibitors
WO2021175824A1 (en) 2020-03-04 2021-09-10 Boehringer Ingelheim International Gmbh Method for administration of an anti cancer agent
WO2021175432A1 (en) 2020-03-04 2021-09-10 Boehringer Ingelheim International Gmbh Method for administration of an anti cancer agent
US11833155B2 (en) 2020-06-03 2023-12-05 Incyte Corporation Combination therapy for treatment of myeloproliferative neoplasms
WO2022198905A1 (en) * 2021-03-26 2022-09-29 浙江海正药业股份有限公司 Tricyclic derivative and preparation method therefor and use thereof
WO2023034290A1 (en) * 2021-08-31 2023-03-09 Incyte Corporation Naphthyridine compounds as inhibitors of kras
WO2023041744A1 (en) 2021-09-17 2023-03-23 Institut Curie Bet inhibitors for treating pab1 deficient cancer
WO2023205251A1 (en) 2022-04-19 2023-10-26 Nuevolution A/S Compounds active towards bromodomains

Also Published As

Publication number Publication date
JP2013510124A (en) 2013-03-21
EP2496576B1 (en) 2013-12-18
GB0919423D0 (en) 2009-12-23
JP5766708B2 (en) 2015-08-19
ES2446358T3 (en) 2014-03-07
EP2496576A1 (en) 2012-09-12
US20120232074A1 (en) 2012-09-13
US8557984B2 (en) 2013-10-15

Similar Documents

Publication Publication Date Title
EP2496576B1 (en) Imidazo[4,5-c]quinoline derivatives as bromodomain inhibitors
EP2585465B1 (en) Benzotriazolodiazepine compounds inhibitors of bromodomains
US8846709B2 (en) 7-(3,5-dimethyl-4-isoxazolyl)-8-(methyloxy)-1H-imidazo[4,5-C]quinoline derivatives
US9315487B2 (en) Tetrahydroquinoline derivatives useful as bromodomain inhibitors
EP2699551B1 (en) Tetrahydroquinoline derivatives useful as bromodomain inhibitors
US9029395B2 (en) Tetrahydroquinoline derivatives useful as bromodomain inhibitors
EP2496580B1 (en) Benzodiazepine bromodomain inhibitor
EP2496557B1 (en) Tetrahydroquinoline derivatives and their pharmaceutical use
US20150133436A1 (en) Bromodomain Inhibitors For Treating Autoimmune And Inflammatory Diseases
WO2011054848A1 (en) Thetrahydroquinolines derivatives as bromodomain inhibitors

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: 10771770

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2010771770

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 13503722

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 2012537382

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE