WO2011161031A1 - Benzotriazolodiazepine compounds inhibitors of bromodomains - Google Patents

Benzotriazolodiazepine compounds inhibitors of bromodomains Download PDF

Info

Publication number
WO2011161031A1
WO2011161031A1 PCT/EP2011/060179 EP2011060179W WO2011161031A1 WO 2011161031 A1 WO2011161031 A1 WO 2011161031A1 EP 2011060179 W EP2011060179 W EP 2011060179W WO 2011161031 A1 WO2011161031 A1 WO 2011161031A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
methyl
salt
chlorophenyl
triazolo
Prior art date
Application number
PCT/EP2011/060179
Other languages
French (fr)
Inventor
James Matthew Bailey
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
Priority claimed from GBGB1010514.6A external-priority patent/GB201010514D0/en
Priority claimed from GBGB1106801.2A external-priority patent/GB201106801D0/en
Application filed by Glaxosmithkline Llc filed Critical Glaxosmithkline Llc
Priority to US13/702,177 priority Critical patent/US9085582B2/en
Priority to JP2013515827A priority patent/JP5844358B2/en
Priority to EP11725769.1A priority patent/EP2585465B1/en
Priority to ES11725769.1T priority patent/ES2526671T3/en
Publication of WO2011161031A1 publication Critical patent/WO2011161031A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/551Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having two nitrogen atoms, e.g. dilazep
    • A61K31/55131,4-Benzodiazepines, e.g. diazepam or clozapine
    • A61K31/55171,4-Benzodiazepines, e.g. diazepam or clozapine condensed with five-membered rings having nitrogen as a ring hetero atom, e.g. imidazobenzodiazepines, triazolam
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention relates to benzodiazepine 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 nuclesome.
  • 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.
  • 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 resides 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- ⁇ complex to inducible genes, resulting in phosphorylation of RNA polymerase and increased transcriptional output (Hargreaves et al, Cell, 2009 138(1 ): 129-145).
  • 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).
  • BRD-NUT fusion proteins contribute to carcinogensesis (Oncogene, 2008, 27, 2237-2242).
  • BRD-t is uniquely expressed in the testes and ovary.
  • 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 containg protein which are said to be useful as anti-cancer agents.
  • PCT patent application PCT/EP2010/066699 discloses a series of benzodiazapine derivatives that inhibit the binding of BET family bromodomains with acetylated lysine residues.
  • 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 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
  • R 1 is C-
  • R 2 is -NR 2 aR 2 a' or -OR 2b ;
  • R 2a or R2'a is hydrogen
  • R2° or the other of R 2a or R2'a are selected from C-
  • any of the carbocyclyl or heterocyclyl groups are optionally substituted by one or more groups selected from halogen, C-
  • -CO-carbocyclyl group may be further optionally substituted by one or more groups selected from halogen, C-
  • R2'a together with the N-atom to which they are attached form a 5-, 6- or 7- membered ring which may optionally contain 1 or 2 heteroatoms independently selected from O, S and N; wherein the 5-, 6- or 7-membered ring may be further optionally substituted by C-
  • R2C and R2'c are independently hydrogen or C-
  • each R3 is independently selected from hydrogen, hydroxy, halo, C-
  • R4 is an aromatic carbocyclic or aromatic heterocyclyl group; wherein the aromatic carbocyclyl or heterocyclyl group is optionally substituted by one or more groups selected from halo, C-
  • R 5 is C ⁇
  • R ⁇ and R 7 are independently hydrogen, C-
  • R6 and R 7 together with the N-atom to which they are attached form a 5-, 6- or 7-membered ring which optionally contains 1 or 2 further heteroatoms independently selected from O, S and N; wherein the 5-, 6- or 7-membered ring may be further optionally substituted by one or more C-
  • R 1 is C-
  • R 2 is -NR 2 aR 2 a' or -OR 2b ;
  • R 2a or R 2 a j s hydrogen
  • R ⁇ b or the other of R 2a or R 2 a are selected from C-
  • any of the carbocyclyl or heterocyclyl groups are optionally substituted by one or more groups selected from halogen, C-
  • R 2a and R 2 a together with the N-atom to which they are attached form a 5-, 6- or 7- membered ring which may optionally contain 1 or 2 heteroatoms independently selected from O, S and N; wherein the 5-, 6- or 7-membered ring may be further optionally substituted by C-
  • R 2 c and R 2 ' c are independently hydrogen or C-
  • each R3 is independently selected from hydrogen, hydroxy, halo, C-
  • R4 is an aromatic carbocyclic or aromatic heterocyclyl group
  • aromatic carbocyclyl or heterocyclyl group is optionally substituted by one or more groups selected from halo, C-
  • R 5 is C ⁇
  • R ⁇ and R 7 are independently hydrogen, C-
  • R6 and R 7 together with the N-atom to which they are attached form a 5-, 6- or 7-membered ring which optionally contains 1 or 2 further heteroatoms independently selected from O, S and N; wherein the 5-, 6- or 7-membered ring may be further optionally substituted by one or more C-
  • the S-enantiomers are preferred.
  • alkyl refers to straight or branched hydrocarbon chains containing the specified number of carbon atoms.
  • Ci -6 alkyl 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.
  • alkoxy refers to a straight or branched alkoxy group containing the specified number of carbon atoms.
  • Ci -6 alkoxy 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.
  • suitable halogens are fluorine, chlorine and bromine.
  • any carbocyclyl group contains 3 to 8 ring-atoms, and may be saturated, unsaturated or aromatic.
  • saturated carbocyclyl groups include cyclopropyl, cyclopentyl or cyclohexyl.
  • unsaturated carbocyclyl groups include those which contain up to 3 double bonds.
  • An example of a suitable aromatic carbocyclyl group is phenyl.
  • carbocylic should be similarly construed.
  • carbocyclyl includes any fused combination of carbocyclyl groups, for example naphthyl, anthryl, phenanthryl, indanyl, indenyl, azulenyl, azulanyl, and fluorenyl.
  • any heterocyclyl group contains 5 to 7 ring-atoms up to 4 of which may be hetero-atoms such as nitrogen, oxygen and sulfur, and may be saturated, unsaturated or aromatic.
  • heterocyclyl groups are furyl, thienyl, pyrrolyl, pyrrolinyl, pyrrolidinyl, imidazolyl, dioxolanyl, oxazolyl, thiazolyl, imidazolyl, imidazolinyl, imidazolidinyl, pyrazolyl, pyrazolinyl, pyrazolidinyl, isoxazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl, pyranyl, pyridyl, piperidinyl, homopiperazinyl, dioxanyl, morpholino, dithianyl, thiomorpholino, thiomorpholino-1 ,1-dioxid
  • heterocyclyl includes fused heterocyclyl groups, for example benzimidazolyl, benzoxazolyl, imidazopyridinyl, benzoxazinyl, benzothiazinyl, oxazolopyridinyl, benzofuranyl, quinolinyl, quinazolinyl, quinoxalinyl, dihydroquinazolinyl, benzothiazolyl, phthalimido, benzofuranyl, benzodiazepinyl, indolyl and isoindolyl.
  • heterocyclic should be similarly construed.
  • substituted refers to substitution with the named substituent or substituents. With multiple degrees of substitution (e.g. 1 or 2) the groups can be the same or different.
  • R 2 is -OR 2b .
  • I n a further embodiment R 2 is -OR 2b in which R 2b is
  • R 2b is selected from methyl, ethyl, isopropyl and butyl.
  • R 2 is -NR 2a R 2a .
  • R 2 is -NR 2a R 2a ' in which R 2a is hydrogen and R 2 a is C-
  • R 2 is -NR 2a R 2a ' in which R 2a is hydrogen and R 2 a is R 2c R 2 c N- C-i .galkyl.
  • R 2 ' a is selected from R 2c R 2 ' c N-CH2CH2-, R 2c R 2 c N- CH 2 CH 2 CH 2 - and R 2 CR2'C N _CH2CH2CH 2 CH2-.
  • R 2 c and R 2 ' C are selected from hydrogen and methyl.
  • R 2 is -NR 2A R 2A ' in which R 2A is hydrogen and R 2 a is carbocyclyl.
  • R 2'a is cyclopentyl or cyclohexyl, wherein each group is optionally substituted once by amino or hydroxyl.
  • R 2 is -NR 2A R 2A ' in which R 2A is hydrogen and R 2 a is heterocyclyl.
  • R 2 a is selected from pyrrolidinyl, piperidinyl, tetrahydrofuranyl and tetrahydropyran, wherein each group is optionally substituted by C-
  • R 2 a is selected from:
  • R 2 is -NR 2A R 2A ' in which R 2A is hydrogen and R 2 A is heterocyclylC-
  • R 2'a is selected from heterocyclyl-CH2-, heterocyclyl-CH2CH2-, heterocyclyl-Ch ⁇ CI- ⁇ CI- ⁇ - and heterocyclyl-Ch ⁇ CI- ⁇ CI- ⁇ CI- ⁇ - ⁇ -, wherein the heterocyclyl is optionally substituted once by amino, hydroxyl or methyl.
  • heterocyclyl is selected from pyrrolidinyl, piperidinyl, tetrahydropyranyl, piperazinyl, morpholino and thiomorpholinodioxide, wherein each group is optionally substituted once by amino, hydroxyl or methyl.
  • heterocyclyl is selected from:
  • n may for example represent an integer in the range 1 to 3 such as 1.
  • R3 is halo, C-
  • R3 is chloro, fluoro, methoxy or CF3.
  • R3 is 3-fluoro, 4-chloro, 4-fluoro, 4- methoxy, or 4-CF3.
  • R3 is 4-chloro.
  • R4 is phenyl, pyridyl, imadolzolyl or pyrazolyl, such groups being optionally substituted by one or more groups selected from halo, C-
  • R4 is selected from
  • R ⁇ is hydrogen, methyl or C(0)Me.
  • R4 is phenyl substituted by a group C- ⁇ alkylNR ⁇ R? in which R6 and
  • R 4 is phenyl substituted by a group CH2NR6R7 in which R6 and R ⁇ together with the N-atom to which they are attached form a pyrolidinyl ring or a piperazinyl ring substituted by a methyl group.
  • the R ⁇ group is at the 8 position of the benzodiazepine ring.
  • 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 relates to compounds of formula (I) as the free base.
  • the invention 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.
  • 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, succinic, 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, succinic, maleic, ace
  • 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.
  • the invention includes within its scope all possible stoichiometric and non-stoichiometric forms of the salts of the compounds of formula (I).
  • the invention encompasses all prodrugs, of the compound of formula (I) and pharmaceutically acceptable salts thereof, which upon administration to the recipient are capable of providing (directly or indirectly) a compound of formula (I) or a pharmaceutically acceptable salts 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
  • the compounds described herein 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).
  • the invention also extends to conformational isomers of compounds of formula (I) and any geometric (c/ ' s and/or trans) isomers of said compounds.
  • An individual isomer isolated such as to be substantially free of the other isomer 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.
  • the compound of formula (I) and pharmaceutically acceptable 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.
  • the compounds of formula (I) may be prepared by the methods decribed in Scheme 1.
  • 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.
  • the compound of formula (I) or pharmaceutically salt thereof can be for use in the treatment of diseases or conditions for which a bromodomain inhibitor indicated.
  • a compound of formula (I) or a pharmaceutically acceptable salt thereof for use in the treatment of diseases or conditions for which a bromodomain is indicated In another embodiment, there is provided a compound or a pharmaceutically acceptable salt thereof for use in the treatment of a chronic autoimmune and/or inflammatory condition. In a further embodiment, there is provided a compound or a pharmaceutically acceptable salt thereof for use in the treatment of cancer, such as midline carcinoma.
  • 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 compound of formula (I) or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for the treatment of a chronic autoimmune and/or inflammatory condition.
  • a compound of formula (I) or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for the treatment of cancer, such as midline carcinoma.
  • a method for treatment of a disease or condition, 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 for treatment of a chronic autoimmune and/or inflammatory condition 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 for treatment of cancer such as midline carcinoma, in a subject in need thereof which comprises administering a therapeutically effective amount of compound of formula (I) 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, dry eye, uveitis, scleritis, hepatitis, pancreatitis, primary biliary cirrhosis, sclerosing cholangitis, Addison's disease, hypophysitis, thyroiditis, type
  • Bromodomain inhibitors may be useful in the treatment of a wide variety of acute and chronic obstructive pulmonary disease.
  • 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 Arteritism, pyoderma gangrenosum 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, SIRS associated with viral infections such as influenza, herpes zoster, herpes simplex and coronavirus.
  • SIRS systemic inflammatory response syndrome
  • multi-organ dysfunction syndrome toxic shock syndrome
  • acute lung injury ARDS (adult respiratory distress syndrome)
  • ARDS adult respiratory distress syndrome
  • fulminant hepatitis burns
  • acute pancreatitis
  • 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
  • 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, 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.
  • 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 acute on 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, human
  • HIV immunodeficiency virus
  • cervical neoplasia adenovirus infections, including acute respiratory disease, and poxvirus infections such as cowpox and smallpox and African swine fever virus.
  • 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 or excipients.
  • the compounds of the 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 the formula (I), or a pharmaceutically acceptable salt thereof, with one or more pharmaceutically acceptable carriers, diluents or excipients.
  • the pharmaceutical composition can be for use in the treatment of any of the conditions described herein.
  • 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.
  • the pharmaceutical composition is adapted for topical administration (e.g. topical administration to the skin or the eye).
  • 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, as, 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 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 the present invention 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 compound 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, foams, sprays, aerosols or oils.
  • Such pharmaceutical compositions may include conventional additives which include, but are not limited to, preservatives, solvents to assist drug penetration, co- solvents, emollients, propellants, viscosity modifying agents (gelling agents), surfactants and carriers.
  • a pharmaceutical composition adapted for topical administration which comprises between 0.01 - 10%, or between 0.01 - 1 % of the compound of formula (I), or a pharmaceutically acceptable salt thereof, by weight of the composition.
  • compositions are preferably applied as a topical ointment, cream, gel or spray foam.
  • 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 compounds of formula (I) or pharmaceutically acceptable salts thereof are 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 can comprise a solution or fine suspension of the active substance in a pharmaceutically acceptable aqueous or nonaqueous 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.
  • a metering valve metered dose inhaler
  • 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 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.
  • Such compositions can be administered to the patient using a suitable device such as the DISKUS
  • GlaxoSmithKline which is for example described in GB 2242134 A.
  • the compound 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.
  • 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-2005/04
  • 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 pharmaceutically acceptable salts 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 formula (I) or a pharmaceutically acceptable salt thereof, calculated as the free base.
  • the pharmaceutically acceptable compound 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 tformula (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 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 pharmaceutically acceptable salts thereof and at least one other pharmaceutically active agent.
  • a combination pharmaceutical product comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof together with one or more other therapeutically active agents.
  • the compound and pharmaceutical compositions 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, beta-2 agonists and Vitamin D3 analogues.
  • one or more other therapeutic agents for example selected from antibiotics, anti-virals, glucocorticosteroids, muscarinic antagonists, beta-2 agonists and Vitamin D3 analogues.
  • the compound of the present invention 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.
  • BINAP refers to 2,2'-bis(diphenylphosphino)-1 ,1 '-binaphthyl
  • BOC refers to tert-butoxycarbonyl
  • DCM refers to dichloromethane
  • 1 ,2-DCE refers to 1 ,2-dichloroethane
  • DIC refers to Diisoproylcarbodiimide
  • DIPEA refers to diisopropylethylamine
  • DMAP refers to 4-dimethylaminopyridine
  • DMSO dimethylsulfoxide
  • DME refers to 1 ,2-dimethoxyethanol
  • DMF refers to ⁇ /,/V-dimethylformamide
  • Et 2 0 refers to diethyl ether
  • EtOAc refers to ethyl acetate
  • FMOC refers to 9-fluorenylmethoxycarbonyl
  • HATU refers to 0-(7-azabenzotriazol-1-yl)-N,N,N',N'-tetramethyluronium hexafluorophosphate
  • HBTU refers to 0-(Benzotriazol-1 -yl)-N,n,N',N'tetramethyluronium hexafluorophosphate
  • HPLC refers to high performance liquid chromatography
  • I PA refers to propan-2-ol
  • i-Pr 2 0 refers to di-isopropyl ether
  • UAIH4 refers to lithium aluminium hydride
  • LCMS Liquid chromatography / mass spectrometry
  • MDAP mass-directed autoprep / mass-directed HPLC
  • MeCN refers to acetonitrile
  • MeOH refers to methanol
  • MgS0 4 refers to magnesium sulfate
  • Mp refers to melting point
  • r.t. refers to room temperature
  • Na 2 S0 4 refers to sodium sulfate
  • TFA refers to trifluoroacetic acid
  • THF refers to tetrahydrofuran
  • 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 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.
  • 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 5 ⁇ packing diameter) or 40ml/min (150mm x 30mm, i.d. 5 ⁇ packing diameter).
  • the UV detection was a summed signal from wavelength of 210nm to 350nm.
  • the de-gassed DMF (4 ml) was added and the reaction heated at 140°C for 2 h (microwave).
  • the reaction mixture was filtered through CeliteTM and the filter bed was washed with ethyl acetate.
  • the filtrate was partitioned between ethyl acetate and water.
  • the organic phase was washed with water (2 x 10 ml) and brine (10 ml). The organics were dried and the solvent was evaporated.
  • Example 1 2-[(4S)-6-(4-Chlorophenyl)-1 -methyl-8-phenyl-4H-[1 ,2,4]triazolo[4,3- a][1 ,4]benzodiazepin-4-yl -N-ethylacetamide
  • the reaction mixture was heated at 120°C for 90 min (microwave). Ethyl acetate (2 ml) was added and the water layer was separated and acidified with glacial acetic acid. The aqueous was extracted with ethyl acetate (3 x 5 ml), the organic fractions dried and the solvent evaporated.
  • reaction mixture was heated at 120°C for 30 min (microwave).
  • Ethyl acetate (2 ml) was added and the water layer was separated.
  • the organic phase was dried and the solvent evaporated.
  • the residue was chromatographed (0-5% methanokDCM) and the product further purified by MDAP (formate modifier).
  • o-Tolylboronic acid (30.1 mg), sodium carbonate (43.0 mg) and bis(triphenylphosphine)palladium(ll) chloride (12.95 mg) were added to a 2-5 ml microwave vial.
  • a solution of (4S)-6-(4-chlorophenyl)-4-[2-(ethylamino)-2-oxoethyl]-1-methyl-4H- [1 ,2,4]triazolo[4,3-a][1 ,4]benzodiazepin-8-yl trifluoromethanesulfonate (for a preparation see intermediate 3) (100 mg) in DME (3 ml) and water (1 ml) was added and the reaction mixture was heated at 120°C for 20 min (microwave) and then at 120°C for a further 30 min (microwave).
  • Example 7 2-[(4S)-6-(4-Chlorophenyl)-1 -methyl-8-(3-pyridinyl)-4H-[1 ,2,4]triazolo[4,3- a][1 ,4]benzodiazepin-4-yl]-N-ethylacetamide
  • 3-Pyridinylboronic acid (27.2 mg), sodium carbonate (43.0 mg) and bis(triphenylphosphine)palladium(ll) chloride (12.95 mg) were added to a 2-5 ml microwave vial.
  • a solution of (4S)-6-(4-chlorophenyl)-4-[2-(ethylamino)-2-oxoethyl]-1-methyl-4H- [1 ,2,4]triazolo[4,3-a][1 ,4]benzodiazepin-8-yl trifluoromethanesulfonate (for a preparation see Intermediate 3) (100 mg) in DME (3 ml) and water (1 ml) was added and the reaction mixture was heated at 120°C for 20 min (microwave).
  • the organic phase was washed with water (2 x 10 ml) and brine (10 ml).
  • the aqueous layer was passed through a pre equilibrated Biotage 103 column.
  • the organic content was eluted with methanol, the methanolic solution was evaporated and the residue was chromatographed (10-20% methanokDCM).
  • LC/MS (Method A) was conducted on a Supelcosil LCABZ+PLUS column (3 ⁇ , 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.
  • 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/MS (Method B) was conducted on an Sunfire C18 column (30mm x 4.6mm i.d. 3.5 ⁇ 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 (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
  • Reference compound K Mixture of 5- and 6- isomers of Alexa Fluor 488-N-(5-aminopentyl)-
  • 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 ⁇ , ⁇ -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 ⁇ , ⁇ -Dimethylformamide
  • the compounds of formula (I) may be tested in the following assays. Fluorescence anisotropy binding assay
  • the binding of the compounds of formula (I) to Bromodomain 2, 3 and 4 can be assessed using an 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.
  • 'a' is the minimum
  • 'b' is the Hill slope
  • 'c' is the plC50
  • 'd' is the maximum.
  • Recombinant Human Bromodomains (Bromodomain 2 (1 -473), Bromodomain 3 (1 -435) and Bromodomain 4 (1 -477)) are 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 are then purified by affinity chromatography on a HisTRAP HP column, eluting with a linear 10-500mM Imidazole gradient, over 20 Cv. Further purification are completed by Superdex 200 prep grade size exclusion column.
  • Protocol for Bromodoamin 3 All components are dissolved in buffer of composition 50 mM HEPES pH7.4, 150mm NaCI and 0.5mM CHAPS with final concentrations of Bromodomains
  • Protocol for Bromodomain 4 All components are dissolved in buffer of composition 50 mM HEPES pH7.4, 150mm NaCI and 0.5mM CHAPS with final concentrations of Bromodomain
  • TR-FRET Fluorescence Resonance Energy Transfer
  • the binding of the compounds of formula (I) to Bromodomains BRD2, BRD3 and BRD4 was assessed using a time resolved fluorescent resonance energy transfer binding assay, that measures the binding of an acetylated histone peptide to the bromodomain protein.
  • the bromodomain protein, histone peptide and a variable concentration of test compound are incubated together to reach thermodynamic equilibrium.
  • the assay is configured such that in the absence of test compound the bromodomain and peptide are significantly bound (-30%) and in the presence of a sufficient concentration of a potent inhibitor this interaction is disrupted leading to a measurable drop in fluorescent resonance energy transfer.
  • the protected peptide was assembled on a solid-phase synthesiser using preloaded Wang resin and utilising standard Fmoc synthesis protocols.
  • the C-terminal lysine was protected by a hyper acid-labile group allowing for its selective removal at the end of the assembly and attachment of the biotin.
  • the crude peptide was obtained after cleavage from the resin with a mixture of trifluoroacetic acid (TFA), triisopropylsilane and water (95:2.5:2.5) for 3h at room temperature and was then purified using a C18 reverse-phase column utilising a 0.1 %TFA- buffered water/acetonitrile gradient.
  • the resulting fractions were analysed and fractions which were >95% pure by analytical HPLC and giving the correct mw (by MALDiTOF mass spectroscopy) were pooled and freeze dried. The final material was analysed by HPLC to confirm purity.
  • Recombinant Human Bromodomains (BRD2 (1-473), BRD3 (1-435) and 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 sonication and purified using a nickel sepharose 6FF column, the proteins were washed and then eluted with 50mM Tris-Hcl pH8.0. 300mM NaCI, 1 mM ⁇ -mercaptoethanol and 20mM Imidazole.
  • Protocol for Bromodomain BRD2, 3 and 4 assays All assay components were dissolved in buffer composition of 50 mM HEPES pH7.4, 50mM NaCI and 0.5mM CHAPS. The final concentration of bromodomain proteins were 100nM and the histone peptide was 300nM, these components are premixed and allowed to equilibrate for 1 hour in the dark. 8 ⁇ of this reaction mixture was added to all wells containing 50nl of various concentrations of test compound or DMSO vehicle (0.5% final) in Greiner 384 well black low volume microtitre plates and incubated in dark for 60 mins at room temperature.
  • 'a' is the minimum
  • 'b' is the Hill slope
  • 'c' is the plC50
  • 'd' is the maximum.
  • Examples 1 and 3-17 were tested in one or more of the above assays and were found to have a plC50 ⁇ 5.5.
  • Examples 1 , 3-5 and 7-17 were tested in one or more of the above assays and were found to have a plC50 ⁇ 6.5.
  • 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 is generated from the data and an IC50 value is calculated.
  • LPS bacterial lipopolysaccharide
  • Examples 1 , 3, 4, 5 and 13 were tested in the above assay were found to have a plC 50 ⁇ 5.5.
  • 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 are given a lethal dose of 15 mg/kg LPS by intraperitoneal injection.
  • animals are dosed intravenously with vehicle (20% cyclodextrin 1 % ethanol in apyrogen water) or compound (10 mg/kg).
  • vehicle 20% cyclodextrin 1 % ethanol in apyrogen water
  • compound 10 mg/kg. The survival of animals is 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 are 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.

Abstract

Benzodiazepine compounds, pharmaceutical compositions containing such compounds and to their use in therapy.

Description

BENZOTRIAZOLODIAZEPINE COMPOUNDS INHIBITORS OF BROMODOMAINS
Field of the Invention
The present invention relates to benzodiazepine 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 nuclesome. 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 resides 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 carcinogensesis (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 containg protein which are said to be useful as anti-cancer agents.
PCT patent application PCT/EP2010/066699 discloses a series of benzodiazapine derivatives that inhibit the binding of BET family bromodomains with acetylated lysine residues.
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 imgf000004_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 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_0001
where
R1 is C-|_3alkyl;
R2 is -NR2aR2a' or -OR2b;
wherein one of R2a or R2'a is hydrogen, and R2° or the other of R2a or R2'a are selected from C-|.galkyl, haloC-|.galkyl, R2c 2'cN_c-|_galkyl, carbocyclyl, carbocyclylC-|.4alkyl, heterocyclyl and heterocyclylC-|.4alkyl,
wherein any of the carbocyclyl or heterocyclyl groups are optionally substituted by one or more groups selected from halogen, C-|.galkyl, haloC-|.galkyl, C-|.galkoxy, haloC-|.galkoxy, carbonyl, -CO-carbocyclyl, azido, amino, hydroxyl, nitro and cyano,
wherein the -CO-carbocyclyl group may be further optionally substituted by one or more groups selected from halogen, C-|.galkyl, haloC-|.galkyl, C-|.galkoxy, haloC-|.galkoxy, azido, nitro and cyano;
or two adjacent groups on any of the carbocyclyl or heterocyclyl groups together with the interconnecting atoms form a 5- or 6-membered ring which ring may contain 1 or 2 heteroatoms independently selected from O, S and N;
or R2a anc| R2'a together with the N-atom to which they are attached form a 5-, 6- or 7- membered ring which may optionally contain 1 or 2 heteroatoms independently selected from O, S and N; wherein the 5-, 6- or 7-membered ring may be further optionally substituted by C-|.galkyl;
R2C and R2'c are independently hydrogen or C-|.galkyl;
each R3 is independently selected from hydrogen, hydroxy, halo, C-|.galkyl, halo C-|.galkyl,
C-|.galkoxy, halo C-|.galkoxy, nitro, cyano, CF3, -OCF3, -COOR5, -C-|_4alkylNR6R7 and - C-|_4alkylOH;
R4 is an aromatic carbocyclic or aromatic heterocyclyl group; wherein the aromatic carbocyclyl or heterocyclyl group is optionally substituted by one or more groups selected from halo, C-|.galkyl, C-|.galkoxy and a group C"|_4alkylNR6R7;
R5 is C<|_3alkyl;
at each occurrence R^ and R7 are independently hydrogen, C-|.galkyl or C(0)C-|.galkyl, or
R6 and R7 together with the N-atom to which they are attached form a 5-, 6- or 7-membered ring which optionally contains 1 or 2 further heteroatoms independently selected from O, S and N; wherein the 5-, 6- or 7-membered ring may be further optionally substituted by one or more C-|.galkyl, -C(0)C-|.galkyl, amino or hydroxyl groups; and n is an integer 1 to 5.
In one embodiment there is provided a compound of formula (I)
where
R1 is C-|_3alkyl;
R2 is -NR2aR2a' or -OR2b;
wherein one of R2a or R2 a js hydrogen, and R^b or the other of R2a or R2 a are selected from C-|.galkyl, haloC-|.galkyl, R2cR2'cN-C2-galkyl R2cO-C2-galkyl, carbocyclyl, carbocyclylC-|_4alkyl, heterocyclyl and heterocyclylC-^alkyl,
wherein any of the carbocyclyl or heterocyclyl groups are optionally substituted by one or more groups selected from halogen, C-|.galkyl, haloC-|.galkyl, C-| .galkoxy, haloC-| .galkoxy, carbonyl, -CO-carbocyclyl, amino, hydroxyl and cyano,
or two adjacent groups on any of the carbocyclyl or heterocyclyl groups together with the interconnecting atoms form a 5- or 6-membered ring which ring may contain 1 or 2 heteroatoms independently selected from O, S and N;
or R2a and R2 a together with the N-atom to which they are attached form a 5-, 6- or 7- membered ring which may optionally contain 1 or 2 heteroatoms independently selected from O, S and N; wherein the 5-, 6- or 7-membered ring may be further optionally substituted by C-|.galkyl;
R2c and R2'c are independently hydrogen or C-|.galkyl;
each R3 is independently selected from hydrogen, hydroxy, halo, C-|.galkyl, halo C-|.galkyl,
C-|.galkoxy, halo C-|.galkoxy, cyano, CF3, -OCF3, -COOR5, -C-|_4alkylNR6R7 and -C<|_ 4alkylOH; R4 is an aromatic carbocyclic or aromatic heterocyclyl group;
wherein the aromatic carbocyclyl or heterocyclyl group is optionally substituted by one or more groups selected from halo, C-|.galkyl, C-|.galkoxy and a group C-^alkylNR^R7
R5 is C<|_3alkyl;
at each occurrence R^ and R7 are independently hydrogen, C-|.galkyl or C(0)C-|.galkyl, or
R6 and R7 together with the N-atom to which they are attached form a 5-, 6- or 7-membered ring which optionally contains 1 or 2 further heteroatoms independently selected from O, S and N; wherein the 5-, 6- or 7-membered ring may be further optionally substituted by one or more C-|.galkyl, -C(0)C-|.galkyl, amino or hydroxyl groups; and n is an integer 1 to 5.
In one embodiment of the invention the S-enantiomers are preferred.
As used herein, the term "alkyl" refers to straight or branched hydrocarbon chains containing the specified number of carbon atoms. For example, Ci-6alkyl 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.
As used herein, the term "alkoxy" refers to a straight or branched alkoxy group containing the specified number of carbon atoms. For example, Ci-6alkoxy 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" or "halo" refers to the elements fluorine, chlorine, bromine and iodine. Examples of suitable halogens are fluorine, chlorine and bromine.
Unless otherwise indicated, any carbocyclyl group contains 3 to 8 ring-atoms, and may be saturated, unsaturated or aromatic. Examples of saturated carbocyclyl groups include cyclopropyl, cyclopentyl or cyclohexyl. Examples of unsaturated carbocyclyl groups include those which contain up to 3 double bonds. An example of a suitable aromatic carbocyclyl group is phenyl. The term carbocylic should be similarly construed. In addition, the term carbocyclyl includes any fused combination of carbocyclyl groups, for example naphthyl, anthryl, phenanthryl, indanyl, indenyl, azulenyl, azulanyl, and fluorenyl.
Unless otherwise indicated, any heterocyclyl group contains 5 to 7 ring-atoms up to 4 of which may be hetero-atoms such as nitrogen, oxygen and sulfur, and may be saturated, unsaturated or aromatic. Examples of heterocyclyl groups are furyl, thienyl, pyrrolyl, pyrrolinyl, pyrrolidinyl, imidazolyl, dioxolanyl, oxazolyl, thiazolyl, imidazolyl, imidazolinyl, imidazolidinyl, pyrazolyl, pyrazolinyl, pyrazolidinyl, isoxazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl, pyranyl, pyridyl, piperidinyl, homopiperazinyl, dioxanyl, morpholino, dithianyl, thiomorpholino, thiomorpholino-1 ,1-dioxide, pyridazinyl, pyrimidinyl, pyrazinyl, piperazinyl, sulfolanyl, tetrazolyl, triazinyl, azepinyl, oxazepinyl, thiazepinyl, diazepinyl and thiazolinyl. In addition, the term heterocyclyl includes fused heterocyclyl groups, for example benzimidazolyl, benzoxazolyl, imidazopyridinyl, benzoxazinyl, benzothiazinyl, oxazolopyridinyl, benzofuranyl, quinolinyl, quinazolinyl, quinoxalinyl, dihydroquinazolinyl, benzothiazolyl, phthalimido, benzofuranyl, benzodiazepinyl, indolyl and isoindolyl. The term heterocyclic should be similarly construed.
As used herein, the term "substituted" refers to substitution with the named substituent or substituents. With multiple degrees of substitution (e.g. 1 or 2) the groups can be the same or different.
In one embodiment, is methyl.
In one embodiment R2 is -OR2b. I n a further embodiment R2 is -OR2b in which R2b is
C-| .galkyl. In a yet further embodiment R2b is selected from methyl, ethyl, isopropyl and butyl.
In one embodiment R2 is -NR2aR2a .
In another embodiment R2 is -NR2aR2a' in which R2a is hydrogen and R2 a is C-| .galkyl. In a further embodiment R2 a is ethyl.
In another embodiment R2 is -NR2aR2a' in which R2a is hydrogen and R2 a is R2cR2 cN- C-i .galkyl. In another embodiment R2'a is selected from R2cR2'cN-CH2CH2-, R2cR2 cN- CH2CH2CH2- and R2CR2'CN_CH2CH2CH2CH2-. In a further embodiment R2c and R2'C are selected from hydrogen and methyl.
In another embodiment R2 is -NR2AR2A' in which R2A is hydrogen and R2 a is carbocyclyl. In a further embodiment R2'a is cyclopentyl or cyclohexyl, wherein each group is optionally substituted once by amino or hydroxyl.
In another embodiment R2 is -NR2AR2A' in which R2A is hydrogen and R2 a is heterocyclyl.
In another embodiment R2 a is selected from pyrrolidinyl, piperidinyl, tetrahydrofuranyl and tetrahydropyran, wherein each group is optionally substituted by C-|.galkyl. In a further embodiment R2 a is selected from:
Figure imgf000009_0001
In another embodiment R2 is -NR2AR2A' in which R2A is hydrogen and R2 A is heterocyclylC-|_4alkyl, wherein the heterocyclyl is optionally substituted once by amino, hydroxyl or methyl. In another embodiment R2'a is selected from heterocyclyl-CH2-, heterocyclyl-CH2CH2-, heterocyclyl-Ch^CI-^CI-^- and heterocyclyl-Ch^CI-^CI-^CI-^-, wherein the heterocyclyl is optionally substituted once by amino, hydroxyl or methyl. In another embodiment heterocyclyl is selected from pyrrolidinyl, piperidinyl, tetrahydropyranyl, piperazinyl, morpholino and thiomorpholinodioxide, wherein each group is optionally substituted once by amino, hydroxyl or methyl. In a further embodiment heterocyclyl is selected from:
Figure imgf000010_0001
n may for example represent an integer in the range 1 to 3 such as 1.
In one embodiment, R3 is halo, C-|.galkyl, C-|.galkoxy or CF3. In another embodiment, R3 is chloro, fluoro, methoxy or CF3. In a further embodiment, R3 is 3-fluoro, 4-chloro, 4-fluoro, 4- methoxy, or 4-CF3. In a further embodiment R3 is 4-chloro.
In one embodiment R4 is phenyl, pyridyl, imadolzolyl or pyrazolyl, such groups being optionally substituted by one or more groups selected from halo, C-|.galkyl, C-|.galkoxy or a group C-^alkylNR^R? jn which R^ is hydrogen and R^ is hydrogen, C-|.galkyl or C(0)C-|. galkyl. In a further embodiment R4 is selected from
Figure imgf000010_0002
in which R^ is hydrogen, methyl or C(0)Me.
In one embodiment R4 is phenyl substituted by a group C-^alkylNR^R? in which R6 and
R^ together with the N-atom to which they are attached form a pyrolidinyl, piperazinyl, piperidinyl or morpholinyl ring said rings being optionally substituted by one or more (e.g 2) C-|.galkyl (such as methyl), -C(0)C-| .galkyl (such as COMe), amino or hydroxyl groups. In a further embodiment R4 is phenyl substituted by a group CH2NR6R7 in which R6 and R^ together with the N-atom to which they are attached form a pyrolidinyl ring or a piperazinyl ring substituted by a methyl group.
In one embodiment the R^ group is at the 8 position of the benzodiazepine ring.
While the embodiments for each variable have generally been listed above separately for each variable this invention includes those compounds in which several or each embodiment in formula (I) is selected from each of the embodiments listed above. Therefore, this invention is intended to include all combinations of embodiments for each variable described hereinabove including salts thereof.
In one embodiment, there is a compound which is selected from
2-[(4S)-6-(4-Chlorophenyl)-1-methyl-8-phenyl-4H-[1 ,2,4]triazolo[4,3-a][1 ,4]benzodiazepin-4- yl]-N-ethylacetamide;
Ethyl [(4S)-6-(4-Chlorophenyl)-1-methyl-8-(4-pyridinyl)-4H-[1 ,2,4]triazolo[4,3- a][1 ,4]benzodiazepin-4-yl]acetate;
2-{(4S)-6-(4-Chlorophenyl)-1-methyl-8-[4-(methyloxy)phenyl]-4H-[1 ,2,4]triazolo[4,3- a][1 ,4]benzodiazepin-4-yl}-N-ethylacetamide;
2-[(4S)-6-(4-Chlorophenyl)-1-methyl-8-(1 H-pyrazol-5-yl)-4H-[1 ,2,4]triazolo[4,3- a][1 ,4]benzodiazepin-4-yl]-N-ethylacetamide;
2-[(4S)-6-(4-Chlorophenyl)-1-methyl-8-(1 H-pyrazol-4-yl)-4H-[1 ,2,4]triazolo[4,3- a][1 ,4]benzodiazepin-4-yl]-N-ethylacetamide;
2-[(4S)-6-(4-Chlorophenyl)-1-methyl-8-(2-methylphenyl)-4H-[1 ,2,4]triazolo[4,3- a][1 ,4]benzodiazepin-4-yl]-N-ethylacetamide;
2-[(4S)-6-(4-Chlorophenyl)-1-methyl-8-(3-pyridinyl)-4H-[1 ,2,4]triazolo[4,3- a][1 ,4]benzodiazepin-4-yl]-N-ethylacetamide;
(S)-2-(6-(4-Chlorophenyl)-8-(3-methoxyphenyl)-1-methyl-4H-benzo[f][1 ,2,4]triazolo[4,3- a][1 ,4]diazepin-4-yl)-N-ethylacetamide;
(S)-2-(6-(4-Chlorophenyl)-1-methyl-8-(pyridin-4-yl)-4H-benzo[f][1 ,2,4]triazolo[4,3- a][1 ,4]diazepin-4-yl)-N-ethylacetamide;
(S)-2-(6-(4-Chlorophenyl)-1-methyl-8-(3-((4-methylpiperazin-1-yl)methyl)phenyl)-4H- benzo[f][1 ,2,4]triazolo[4,3-a][1 ,4]diazepin-4-yl)-N-ethylacetamide; (S)-2-(6-(4-Chlorophenyl)-8-(3-((ethylamino)methyl)phenyl)-1 -methyl-4H- benzo[f][1 ,2,4]triazolo[4,3-a][1 ,4]diazepin-4-yl)-N-ethylacetamide ;
(S)-2-(6-(4-Chlorophenyl)-1-methyl-8-(3-(pyrrolidin-1-ylmethyl)phenyl)-4H- benzo[f][1 ,2,4]triazolo[4,3-a][1 ,4]diazepin-4-yl)-N-ethylacetamide;
(S)-2-(6-(4-Chlorophenyl)-1-methyl-8-(4-(pyrrolidin-1-ylmethyl)phenyl)-4H- benzo[f][1 ,2,4]triazolo[4,3-a][1 ,4]diazepin-4-yl)-N-ethylacetamide;
(S)-2-(6-(4-Chlorophenyl)-1-methyl-8-(4-((4-methylpiperazin-1 -yl)methyl)phen
benzo[f][1 ,2,4]triazolo[4,3-a][1 ,4]diazepin-4-yl)-N-ethylacetamide;
(S)-2-(6-(4-Chlorophenyl)-8-(4-((ethylamino)methyl)phenyl)-1 -methyl-4H- benzo[f][1 ,2,4]triazolo[4,3-a][1 ,4]diazepin-4-yl)-N-ethylacetamide;
(S)-2-(6-(4-Chlorophenyl)-8-(4-((dimethylamino)methyl)phenyl)-1 -meth
benzo[f][1 ,2,4]triazolo[4,3-a][1 ,4]diazepin-4-yl)-N-ethylacetamide; and
(S)-2-(6-(4-Chlorophenyl)-1-methyl-8-(2H-tetrazol-5-yl)-4H-benzo[f][1 ,2,4]triazolo[4,3- a][1 ,4]diazepin-4-yl)-N-ethylacetamide
or a salt thereof.
In a particular embodiment there is provided (S)-2-(6-(4-chlorophenyl)-1-methyl-8-(4- (pyrrolidin-1-ylmethyl)phenyl)-4H-benzo[f][1 ,2,4]triazolo[4,3-a][1 ,4]diazepin-4-yl)-N- ethylacetamide or a salt thereof.
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 relates to compounds of formula (I) as the free base. In another embodiment the invention 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. 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, succinic, 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) and pharmaceutically acceptable salts thereof, which upon administration to the recipient are capable of providing (directly or indirectly) a compound of formula (I) or a pharmaceutically acceptable salts 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).
The compounds described herein 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).
Similarly the invention also extends to conformational isomers of compounds of formula (I) and any geometric (c/'s and/or trans) isomers of said compounds.
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 compound of formula (I) and pharmaceutically acceptable 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.
The compounds of formula (I) may be prepared by the methods decribed in Scheme 1.
Scheme 1
Figure imgf000016_0001
Figure imgf000016_0002
Figure imgf000016_0003
Compounds of formula (II) may be prepared in accordance with the methods described in Scheme 2.
Scheme 2
Figure imgf000017_0001
 Compounds of formula (V) may also be prepared according to the methods described in Scheme 3.
Scheme 3
Figure imgf000018_0001
Figure imgf000019_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 in the above processes. 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 to belived to have potential utility in the treatment of diseases or conditions for which a bromodomain is indicated.
The present invention thus provides a compound of formula (I) or a pharmaceutically acceptable salt thereof for use in therapy. The compound of formula (I) or pharmaceutically salt thereof can be for use in the treatment of diseases or conditions for which a bromodomain inhibitor indicated.
In one embodiment there is provided a compound of formula (I) or a pharmaceutically acceptable salt thereof for use in the treatment of diseases or conditions for which a bromodomain is indicated. In another embodiment, there is provided a compound or a pharmaceutically acceptable salt thereof for use in the treatment of a chronic autoimmune and/or inflammatory condition. In a further embodiment, there is provided a compound or a pharmaceutically acceptable salt thereof for use in the treatment of cancer, such as midline carcinoma.
In one embodiment 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. In another embodiment, 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 a chronic autoimmune and/or inflammatory condition. In a further embodiment, 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 cancer, such as midline carcinoma.
In one embodiment there is provided a method for treatment of a disease or condition, 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 another embodiment there is provided a method for treatment of a chronic autoimmune and/or inflammatory condition, 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 a further embodiment there is provided a method for treatment of cancer, such as midline carcinoma, 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 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, dry eye, 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 Arteritism, pyoderma gangrenosum 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, 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, and 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, 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. 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 acute on 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, human
immunodeficiency virus (HIV), cervical neoplasia, adenovirus infections, including acute respiratory disease, and 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 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 or excipients. The compounds of the 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 the formula (I), or a pharmaceutically acceptable salt thereof, with one or more pharmaceutically acceptable carriers, diluents or excipients. The pharmaceutical composition can be for use in the treatment of any of the conditions described herein.
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.
In one embodiment the pharmaceutical composition is adapted for topical administration (e.g. topical administration to the skin or the eye).
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, as, 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 the present invention 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 compound 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, foams, sprays, aerosols or oils. Such pharmaceutical compositions may include conventional additives which include, but are not limited to, preservatives, solvents to assist drug penetration, co- solvents, emollients, propellants, viscosity modifying agents (gelling agents), surfactants and carriers. In one embodiment there is provided a pharmaceutical composition adapted for topical administration which comprises between 0.01 - 10%, or between 0.01 - 1 % of the compound of formula (I), or a pharmaceutically acceptable salt thereof, by weight of the composition.
For treatments of the eye or other external tissues, for example mouth and skin, the compositions are preferably applied as a topical ointment, cream, gel or spray foam. 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 compounds of formula (I) or pharmaceutically acceptable salts thereof are 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 nonaqueous 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 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 compound 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 pharmaceutically acceptable salts 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 formula (I) or a pharmaceutically acceptable salt thereof, calculated as the free base.
The pharmaceutically acceptable compound 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 tformula (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 compound of formula (I) and pharmaceutically acceptable salts thereof 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 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 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 pharmaceutically acceptable salts thereof and at least one other pharmaceutically active agent. In one embodiment there is provided a combination pharmaceutical product comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof together with one or more other therapeutically active agents.
Thus in one aspect, the compound and pharmaceutical compositions 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, beta-2 agonists and Vitamin D3 analogues.
It will be appreciated that when the compound of the present invention 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.
General Experimental Details
All temperatures referred to are in °C.
The names of the following compounds have been obtained using the compound naming programme "ACD Name Pro 6.02" or Chem Draw Ultra 12.0.
Abbreviations
AcOH refers to acetic acid
BINAP refers to 2,2'-bis(diphenylphosphino)-1 ,1 '-binaphthyl BOC refers to tert-butoxycarbonyl
CV refers to column volumes
DCM refers to dichloromethane
1 ,2-DCE refers to 1 ,2-dichloroethane
DIC refers to Diisoproylcarbodiimide
DIPEA refers to diisopropylethylamine
DMAP refers to 4-dimethylaminopyridine
DMSO refers to dimethylsulfoxide.
DME refers to 1 ,2-dimethoxyethanol
DMF refers to Λ/,/V-dimethylformamide
Et20 refers to diethyl ether
EtOAc refers to ethyl acetate
FMOC refers to 9-fluorenylmethoxycarbonyl
HATU refers to 0-(7-azabenzotriazol-1-yl)-N,N,N',N'-tetramethyluronium hexafluorophosphate
HBTU refers to 0-(Benzotriazol-1 -yl)-N,n,N',N'tetramethyluronium hexafluorophosphate
HPLC refers to high performance liquid chromatography
I PA refers to propan-2-ol
i-Pr20 refers to di-isopropyl ether
UAIH4 refers to lithium aluminium hydride
LCMS = Liquid chromatography / mass spectrometry
MDAP = mass-directed autoprep / mass-directed HPLC
MeCN refers to acetonitrile
MeOH refers to methanol
MgS04 refers to magnesium sulfate
Mp refers to melting point
r.t. refers to room temperature
RT = retention time
Na2S04 refers to sodium sulfate
TFA refers to trifluoroacetic acid
THF refers to tetrahydrofuran
LCMS methodology
Method Formate LC conditions
The UPLC analysis was conducted on an Acquity UPLC BEH C18 column (50mm x 2.1 mm, i.d. 1.7μηη 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 imgf000032_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
MDAP methodology
Method Formate modifier
LC conditions
The HPLC analysis was conducted on either a Sunfire C18 column (100mm x 19mm, i.d 5μηη packing diameter) or a Sunfire C18 column (150mm x 30mm, i.d. 5μηη 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 5μηη packing diameter) or 40ml/min (150mm x 30mm, i.d. 5μηη 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.
Intermediate 1 : [(4S)-6-(4-Chlorophenyl)-1 -methyl-8-phenyl-4H-[1 ,2,4]triazolo[4,3- a][1 ,4]benzodiazepin-4-yl]acetic acid
Figure imgf000033_0001
Sodium carbonate (35.8 mg), phenylboronic acid (22.5 mg) and bis(triphenylphosphine)palladium(ll) chloride (10.8 mg) were added to a 2-5 ml microwave vial. A solution of ethyl ((4S)-6-(4-chlorophenyl)-1 -methyl-8-{[(trifluoromethyl)sulfonyl]oxy}- 4H-[1 ,2,4]triazolo[4,3-a][1 ,4]benzodiazepin-4-yl)acetate (for a preparation see Intermediate 2) (83.4 mg) in DME (3 ml) and water (1 ml) was added to the microwave vial. The reaction mixture was heated at 120°C for 90 min (microwave). Ethyl acetate (2 ml) was added and the water layer was separated and acidified with glacial acetic acid. The water layer was extracted with ethyl acetate (3 x 5 ml) and the organic layer was dried and the solvent evaporated. The residue was chromatographed (0-10% methanohDCM) to give [(4S)-6-(4- chlorophenyl)-1-methyl-8-phenyl-4H-[1 ,2,4]triazolo[4,3-a][1 ,4]benzodiazepin-4-yl]acetic acid as a white oil (30.1 mg), which was used without further purification in the subsequent reaction. LCMS (formate): MH+ 443 / 445, RT 1.09 min
Intermediate 2: Ethyl ((4S)-6-(4-chlorophenyl)-1 -methyl-8-
{[(trifluoromethyl)sulfon nzodiazepin-4-yl)acetate
Figure imgf000034_0001
Ethyl [(4S)-6-(4-chlorophenyl)-8-hydroxy-1 -methyl-4H-[1 ,2,4]triazolo[4,3- a][1 ,4]benzodiazepin-4-yl]acetate (508 mg, for a preparation see intermediate 8) was dissolved in DCM (30 ml). Pyridine (500 μΙ) was added and the reaction mixture was stirred and cooled an ice bath. Triflic anhydride (251 μΙ) was added slowly. After addition the mixture was stirred in the ice bath for 2.5 h. The solvent was evaporated and the residue was chromatographed (0-5% methanohDCM) to give ethyl ((4S)-6-(4-chlorophenyl)-1 -methyl-8- {[(trifluoromethyl)sulfonyl]oxy}-4H-[1 ,2,4]triazolo[4,3-a][1 ,4]benzodiazepin-4-yl)acetate (295mg) as an orange oil and further impure ethyl ((4S)-6-(4-chlorophenyl)-1-methyl-8- {[(trifluoromethyl)sulfonyl]oxy}-4H-[1 ,2,4]triazolo[4,3-a][1 ,4]benzodiazepin-4-yl)acetate. The impure material was chromatographed (0-5% methanohDCM) to give the pure ethyl ((4S)-6- (4-chlorophenyl)-1-methyl-8-{[(trifluoromethyl)sulfonyl]oxy}-4H-[1 ,2,4]triazolo[4,3- a][1 ,4]benzodiazepin-4-yl)acetate (88mg) as a white solid.
LCMS (formate): MH+ 543 / 545, RT 1.22 min
Intermediate 3: (4S)-6-(4-Chlorophenyl)-4-[2-(ethylamino)-2-oxoethyl]-1 -methyl-4H- [1 ,2,4]triazolo[4,3-a][1 ,4]benzodiazepin-8-yl trifluoromethanesulfonate 2-[(4S)-6-(4-Chlorophenyl)-8-hydroxy-1 -methyl-4H-[1 ,2,4]triazolo[4,3-a][1 ,4]benzodiazepin-4- yl]-N-ethylacetamide (for a preparation see Intermediate 4) (104.2 mg) was dissolved in DCM (10 ml). DIPEA (53 μΙ) and N-phenyl-bis(trifluoromethanesulfonamide) (100 mg) were added and the reaction mixture was stirred at room temperature for 3.5 h. The solvent was evaporated and the residue was chromatographed (0-5% methanokDCM) to give (4S)-6-(4- chlorophenyl)-4-[2-(ethylamino)-2-oxoethyl]-1 -methyl-4H-[1 ,2,4]triazolo[4,3- a][1 ,4]benzodiazepin-8-yl trifluoromethanesulfonate as a white glassy solid (130mg).
LCMS (formate): MH+ 542 / 544, RT 1.10 min
Intermediate 4: 2-[(4S)-6-(4-Chlorophenyl)-8-hydroxy-1 -methyl-4H-[1 ,2,4]triazolo[4,3- a][1 ,4]benzodiazepin-4-yl]- -ethylacetamide
Figure imgf000035_0001
2-[(4S)-6-(4-Chlorophenyl)-1-methyl-8-(methyloxy)-4H-[1 ,2,4]triazolo[4,3- a][1 ,4]benzodiazepin-4-yl]-/V-ethylacetamide (which may be prepared, for example, by reaction of Reference compound H with ethylamine)(0.424 g) was dissolved in DCM (8 ml) and cooled to -78°C. A solution of boron tribromide (0.945 ml) in DCM (2ml) was added slowly to the reaction mixture. The reaction was stirred at -78°C for 35 min under nitrogen and then at room temperature for 2.5 h. The reaction was quenched by adding a solution of ethanol (1 ml) in DCM (5 ml) dropwise. The solvent was evaporated and the residue was chromatographed (0-10% methanokDCM) to give 2-[(4S)-6-(4-chlorophenyl)-8-hydroxy-1 - methyl-4H-[1 ,2,4]triazolo[4,3-a][1 ,4]benzodiazepin-4-yl]-N-ethylacetamide as a yellow solid. The aqueous layer was passed through a pre equilibrated Biotage 103 column, the organic content was eluted with methanol and the methanolic solution was evaporated to give a further quantity of 2-[(4S)-6-(4-chlorophenyl)-8-hydroxy-1-methyl-4H-[1 ,2,4]triazolo[4,3- a][1 ,4]benzodiazepin-4-yl]-N-ethylacetamide as a yellow solid (total yield 271.3mg).
LCMS (formate): MH+ 410 / 412, RT 0.76 min
Intermediate 5: (S)-2-(6-(4-Chlorophenyl)-8-(3-formylphenyl)-1 -methyl-4H- benzo[f][1,2,4]triazolo[4,3- ide
Figure imgf000036_0001
(S)-6-(4-Chlorophenyl)-4-(2-(ethylamino)-2-oxoethyl)-1 -methyl-4H-benzo[f][1 ,2,4]triazolo[4,3- a][1 ,4]diazepin-8-yl trifluoromethanesulfonate (for a preparation see Intermediate 3) (500 mg) potassium carbonate (638 mg), 3-formylphenylboronic acid (166 mg) and bis(triphenylphosphine)palladium(ll) chloride (64.8 mg) were added to a 20 ml microwave vial. Ethanol (9 ml) and toluene (9ml) were added to the vial and the reaction mixture was heated at 120°C for 20 min (microwave). The reaction mixture was filtered through Celite™ and the filter bed was washed with ethanol. The solvent was evaporated and the residue was chromatographed (0-10% methanokDCM) to give (S)-2-(6-(4-chlorophenyl)-8-(3- formylphenyl)-1-methyl-4H-benzo[f][1 ,2,4]triazolo[4,3-a][1 ,4]diazepin-4-yl)-N-ethylacetamide as a light brown solid (287 mg). LCMS (formate): MH+ 498 / 500, RT 0.99 min
Intermediate 6: (S)-2-(6-(4-Chlorophenyl)-8-(4-formylphenyl)-1 -methyl-4H- benzo[f][1,2,4]triazolo[4, -a][1 ,4]diazepin-4-yl)-N-ethylacetamide
Figure imgf000036_0002
(S)-6-(4-Chlorophenyl)-4-(2-(ethylamino)-2-oxoethyl)-1 -methyl-4H-benzo[f][1 ,2,4]triazolo[4,3- a][1 ,4]diazepin-8-yl trifluoromethanesulfonate (for a preparation see intermediate 3) (500 mg), 4-formylphenylboronic acid (166 mg), potassium carbonate (638 mg) and bis(triphenylphosphine)palladium(ll) chloride (64.8 mg) were added to a 20 ml microwave vial. Ethanol (9 ml) and toluene (9 ml) were added to the vial. The reaction mixture was heated in a microwave at 120°C for 20 min. The reaction mixture was filtered through Celite™ and and the filter bed was washed with ethanol. The solvent was evaporated, and the residue was chromatographed (0-5% methanohDCM) to give (S)-2-(6-(4-chlorophenyl)-8- (4-formylphenyl)-1-methyl-4H-benzo[f][1 ,2,4]triazolo[4,3-a][1 ,4]diazepin-4-yl)-N- ethylacetamide (327 mg) as a brown solid. LCMS (formate): MH+ 498 / 500, RT 0.98 min
Intermediate 7: (S)-2-(6-(4-Chlorophenyl)-8-cyano-1 -methyl-4H- benzo[f][1,2,4]triazolo[4,3-a][1 ,4]diazepin-4-yl)-N-ethylacetamide
Figure imgf000037_0001
DMF (4 ml) was de-gassed for 20 min. (S)-6-(4-Chlorophenyl)-4-(2-(ethylamino)-2-oxoethyl)- 1-methyl-4H-benzo[f][1 ,2,4]triazolo[4,3-a][1 ,4]diazepin-8-yl trifluoromethanesulfonate for a preparation see Intermediate 3) (250 mg) zinc cyanide (54.2 mg) and tetrakis(triphenylphosphine)palladium(0) (53.3 mg) were added to a 2-5 ml microwave vial. The de-gassed DMF (4 ml) was added and the reaction heated at 140°C for 2 h (microwave). The reaction mixture was filtered through Celite™ and the filter bed was washed with ethyl acetate. The filtrate was partitioned between ethyl acetate and water. The organic phase was washed with water (2 x 10 ml) and brine (10 ml). The organics were dried and the solvent was evaporated. The residue was chromatographed (0-5% methanohDCM) and triturated with diethyl ether to give (S)-2-(6-(4-chlorophenyl)-8-cyano-1 -methyl-4H- benzo[f][1 ,2,4]triazolo[4,3-a][1 ,4]diazepin-4-yl)-N-ethylacetamide (73.5 mg) as a white solid. LCMS (formate): MH+ 419 / 421 , RT 0.86 min
Intermediate 8: Ethyl [(4S)-6-(4-chlorophenyl)-8-hydroxy-1 -methyl-4H- [1 ,2,4]triazolo[4,3-a][1 ,4]benzodiazepin-4-yl]acetate CI
[(4S)-6-(4-Chlorophenyl)-1 -methyl-8-(methyloxy)-4H-[1 ,2,4]triazolo[4,3-a][1 ,4]benzodiazepin- 4-yl]acetic acid (1 .2 g) was dissolved in DCM (30 ml) and cooled to -78°C. A solution of boron tribromide (2.86 ml) in DCM (6 ml) was added to the mixture slowly. The reaction was stirred at -78°C for 25 min and then at room temperature overnight. The reaction was quenched by dropwise addition of a solution of ethanol (6 ml) in DCM (30 ml). The solvent was evaporated and the residue dissolved in ethanol (20 ml). Concentrated sulphuric acid (1 ml) was added and the mixture was refluxed for 3.5 h. The solvent was evaporated and the residue was dissolved in ethyl acetate (45 ml). The solution was washed with water (2 x 30 ml) then brine (10 ml), the organic phase dried and the solvent evaporated to give Ethyl [(4S)-6-(4-chlorophenyl)-8-hydroxy-1-methyl-4H-[1 ,2,4]triazolo[4,3-a][1 ,4]benzodiazepin-4- yl]acetate (519 mg) as a yellow solid, which was used without further purification in the subsequent reaction. LCMS (formate): MH+ 41 1 / 413, RT 0.90 min
Example 1 : 2-[(4S)-6-(4-Chlorophenyl)-1 -methyl-8-phenyl-4H-[1 ,2,4]triazolo[4,3- a][1 ,4]benzodiazepin-4-yl -N-ethylacetamide
Figure imgf000038_0001
[(4S)-6-(4-Chlorophenyl)-1 -methyl-8-phenyl-4H-[1 ,2,4]triazolo[4,3-a][1 ,4]benzodiazepin-4- yl]acetic acid (for a preparation see Intermediate 1 ) (29 mg) was dissolved in DMF (2 ml). DIPEA (23 μΙ), HATU (29.9 mg) and ethylamine hydrochloride (10.68 mg) were added to the reaction mixture. The reaction was stirred at room temperature overnight. The mixture was dissolved in ethyl acetate (10 ml) and washed with saturated hydrogen carbonate solution (5 ml). The organic phase was washed with water (5 ml) and brine (5 ml). The organic layer was dried and the solvent evaporated. The residue was chromatographed (0-5% methanohDCM) and then further purified by MDAP (formate modifier) to give 2-[(4S)-6-(4- chlorophenyl)-1-methyl-8-phenyl-4H-[1 ,2,4]triazolo[4,3-a][1 ,4]benzodiazepin-4-yl]-N- ethylacetamide (7.5 mg) as a white oil. LCMS (formate): MH+ 470 / 472, RT 1.10 min
Example 2: ethyl [(4S)-6-(4-Chlorophenyl)-1 -methyl-8-(4-pyridinyl)-4H- [1 ,2,4]triazolo[4,3-a][1 ,4]benzodiazepin-4-yl]acetate
Figure imgf000039_0001
Sodium carbonate (66.3 mg), 4-pyridinylboronic acid (22.5 mg) and bis(triphenylphosphine)palladium(ll) chloride (19.1 mg) were added to a 2-5 ml microwave vial. A solution of ethyl ((4S)-6-(4-chlorophenyl)-1-methyl-8-{[(trifluoromethyl)sulfonyl]oxy}- 4H-[1 ,2,4]triazolo[4,3-a][1 ,4]benzodiazepin-4-yl)acetate (for a preparation see intermediate 2) (147.5 mg) in DME (3 ml) and water (1 ml) was added to the microwave vial. The reaction mixture was heated at 120°C for 90 min (microwave). Ethyl acetate (2 ml) was added and the water layer was separated and acidified with glacial acetic acid. The aqueous was extracted with ethyl acetate (3 x 5 ml), the organic fractions dried and the solvent evaporated. The residue was chromatographed (0-10% methanohDCM) and then further purified by MDAP (formate modifier) to give ethyl [(4S)-6-(4-chlorophenyl)-1-methyl-8-(4- pyridinyl)-4H-[1 ,2,4]triazolo[4,3-a][1 ,4]benzodiazepin-4-yl]acetate as a white solid (5.5 mg). LCMS (formate): MH+ 472 / 474, RT 0.79 min
Example 3: 2-{(4S)-6-(4-Chlorophenyl)-1 -methyl-8-[4-(methyloxy)phenyl]-4H- [1 ,2,4]triazolo[4,3-a][1 ,
Figure imgf000039_0002
4-Methoxybenzeneboronic acid (50.5 mg), sodium carbonate (64.5 mg) and bis(triphenylphosphine)palladium(ll) chloride (19.4 mg) were added to a 2-5 ml microwave vial. A solution of (4S)-6-(4-chlorophenyl)-4-[2-(ethylamino)-2-oxoethyl]-1-methyl-4H- [1 ,2,4]triazolo[4,3-a][1 ,4]benzodiazepin-8-yl trifluoromethanesulfonate (for a preparation see Intermediate 3) (150 mg) in DME (3 ml) and water (1 ml) was added and the reaction mixture was heated at 120°C for 20 min (microwave). The reaction mixture was partitioned between ethyl acetate (10 ml) and water (10 ml). The organic phase was separated, washed with water (10 ml) and brine (10 ml). The organic phase was dried and the solvent was evaporated. The residue was chromatographed (0-5% methanokDCM) to give 2-{(4S)-6-(4- chlorophenyl)-1-methyl-8-[4-(methyloxy)phenyl]-4H-[1 ,2,4]triazolo[4,3-a][1 ,4]benzodiazepin- 4-yl}-N-ethylacetamide as a light brown solid (84 mg). LCMS (formate): MH+ 500 / 502, RT 1 .08 min
Example 4: 2-[(4S)-6-(4-Chlorophenyl)-1 -methyl-8-(1 H-pyrazol-5-yl)-4H-
[1 ,2,4]triazolo[4,3-a][1 ,4]b
Figure imgf000040_0001
Sodium carbonate (43.0 mg), 1 H-pyrazole-5-boronic acid (24.8 mg) and bis(triphenylphosphine)palladium(ll) chloride (13.0 mg) were added to a 2-5 ml microwave vial. A solution of (4S)-6-(4-chlorophenyl)-4-[2-(ethylamino)-2-oxoethyl]-1-methyl-4H- [1 ,2,4]triazolo[4,3-a][1 ,4]benzodiazepin-8-yl trifluoromethanesulfonate (for a preparation see intermediate 3) (100 mg) in DME (3 ml) and water (1 ml) was added. The reaction mixture was heated at 120°C for 30 min (microwave). Ethyl acetate (2 ml) was added and the water layer was separated. The organic phase was dried and the solvent evaporated. The residue was chromatographed (0-5% methanokDCM) and the product further purified by MDAP (formate modifier). The resulting gum was loaded on a 1 g SCX-2 column and washed with ammonia in methanol (2M) to give 2-[(4S)-6-(4-chlorophenyl)-1 -methyl-8-(1 H-pyrazol-5-yl)- 4H-[1 ,2,4]triazolo[4,3-a][1 ,4]benzodiazepin-4-yl]-N-ethylacetamide as a brown oil (15.6 mg). LCMS (formate): MH+ 460 / 462, RT 0.80 min Example 5: 2-[(4S)-6-(4-Chlorophenyl)-1 -methyl-8-(1 H-pyrazol-4-yl)-4H-
[1 ,2,4]triazolo[4,3-a][1 ,4]b
Figure imgf000041_0001
Sodium carbonate (43.0 mg), 1 H-pyrazole-4-boronic acid (24.8 mg) and bis(triphenylphosphine)palladium(ll) chloride (13.0 mg) were added to a 2-5 ml microwave vial. A solution of (4S)-6-(4-chlorophenyl)-4-[2-(ethylamino)-2-oxoethyl]-1-methyl-4H- [1 ,2,4]triazolo[4,3-a][1 ,4]benzodiazepin-8-yl trifluoromethanesulfonate (100 mg, intermediate 3) in DME (3 ml) and water (1 ml) was added. The reaction mixture was heated at 120°C for 30 min (microwave). Ethyl acetate (2 ml) was added and the water layer was separated. The organic layer was dried and the solvent evaporated. The residue was purified by MDAP (formate modifier). The product was loaded on a 1 g SCX-2 column in methanol and eluted with ammonia in methanol (2M) to give 2-[(4S)-6-(4-chlorophenyl)-1-methyl-8-(1 H-pyrazol-4- yl)-4H-[1 ,2,4]triazolo[4,3-a][1 ,4]benzodiazepin-4-yl]-N-ethylacetamide as a white oil (6.1 mg). LCMS (formate): MH+ 460 / 462, RT 0.79 min
Example 6: 2-[(4S)-6-(4-Chlorophenyl)-1 -methyl-8-(2-methylphenyl)-4H-
[1 ,2,4]triazolo[4,3-a][1 ,4]
Figure imgf000041_0002
o-Tolylboronic acid (30.1 mg), sodium carbonate (43.0 mg) and bis(triphenylphosphine)palladium(ll) chloride (12.95 mg) were added to a 2-5 ml microwave vial. A solution of (4S)-6-(4-chlorophenyl)-4-[2-(ethylamino)-2-oxoethyl]-1-methyl-4H- [1 ,2,4]triazolo[4,3-a][1 ,4]benzodiazepin-8-yl trifluoromethanesulfonate (for a preparation see intermediate 3) (100 mg) in DME (3 ml) and water (1 ml) was added and the reaction mixture was heated at 120°C for 20 min (microwave) and then at 120°C for a further 30 min (microwave). Bis(triphenylphosphine)palladium(ll) chloride (12.7 mg) was added and the reaction mixture was heated at 120°C for 30 min (microwave). The reaction mixture was partitioned between ethyl acetate (10 ml) and water (10 ml). The organic phase was separated and washed with water (10 ml) and brine (10 ml), dried and the solvent evaporated. The residue was chromatographed (0-5% methanohDCM). The resulting mixture was repurified by MDAP (formate modifier) to give 2-[(4S)-6-(4-chlorophenyl)-1-methyl-8-(2- methylphenyl)-4H-[1 ,2,4]triazolo[4,3-a][1 ,4]benzodiazepin-4-yl]-N-ethylacetamide as a white oil (16.8 mg). LCMS (formate): MH+ 484 / 486, RT 1 .14 min
Example 7: 2-[(4S)-6-(4-Chlorophenyl)-1 -methyl-8-(3-pyridinyl)-4H-[1 ,2,4]triazolo[4,3- a][1 ,4]benzodiazepin-4-yl]-N-ethylacetamide
Figure imgf000042_0001
3-Pyridinylboronic acid (27.2 mg), sodium carbonate (43.0 mg) and bis(triphenylphosphine)palladium(ll) chloride (12.95 mg) were added to a 2-5 ml microwave vial. A solution of (4S)-6-(4-chlorophenyl)-4-[2-(ethylamino)-2-oxoethyl]-1-methyl-4H- [1 ,2,4]triazolo[4,3-a][1 ,4]benzodiazepin-8-yl trifluoromethanesulfonate (for a preparation see Intermediate 3) (100 mg) in DME (3 ml) and water (1 ml) was added and the reaction mixture was heated at 120°C for 20 min (microwave). The reaction mixture was partitioned between ethyl acetate (10 ml) and water (10 ml). The organic phase was separated, washed with water (10 ml) and brine (10 ml). The organic phase was dried and the solvent evaporated. The residue was chromatographed (0-10% methanohDCM) product was further purified by MDAP (formate modifier) to give 2-[(4S)-6-(4-chlorophenyl)-1 -methyl-8-(3-pyridinyl)-4H- [1 ,2,4]triazolo[4,3-a][1 ,4]benzodiazepin-4-yl]-N-ethylacetamide as a white oil (1 1.9 mg).
LCMS (formate): MH+ 471 / 473, RT 0.73 min
Example 8: (S)-2-(6-(4-Chlorophenyl)-8-(3-methoxyphenyl)-1 -methyl-4H- benzo[f][1,2,4]triazolo[4,3-a][1 ,4]diazepin-4-yl)-N-ethylacetamide
Figure imgf000043_0001
(S)-6-(4-ChlorophenylH-(2-(ethylamino)-2-oxoethyl)-1 -methyl-4H-benzo[fl[1 ,2,4]triazolo[4,3- a][1 ,4]diazepin-8-yl trifluoromethanesulfonate (for a preparation see Intermediate 3) (100 mg), bis(triphenylphosphine)palladium(ll) chloride (12.95 mg), potassium carbonate (128 mg) and 3-methoxyphenylboronic acid (33.6 mg) were added to a 2-5 ml microwave vial. Toluene (2 ml) and ethanol (2 ml) were added to the vial and the reaction mixture was heated at 120°C for 20 min (microwave). The reaction mixture was filtered through Celite™ and the filter bed was washed with ethanol. The solvent was evaporated and the residue was chromatographed (0-5% methanokDCM), triturated with diethyl ether and dried in a vacuum dessicator at 40°C overnight to give (S)-2-(6-(4-chlorophenyl)-8-(3-methoxyphenyl)-1 -methyl- 4H-benzo[f][1 ,2,4]triazolo[4,3-a][1 ,4]diazepin-4-yl)-N-ethylacetamide as a cream solid (59.6 mg). LCMS (formate): MH+ 500 / 502, RT 1.09 min
Example 9: (S)-2-(6-(4-Chlorophenyl)-1 -methyl-8-(pyridin-4-yl)-4H- benzo[f][1,2,4]triazolo[4, -a][1 ,4]diazepin-4-yl)-N-ethylacetamide
Figure imgf000043_0002
(S)-6-(4-Chlorophenyl)-4-(2-(ethylamino)-2-oxoethyl)-1 -methyl-4H-benzo[f][1 ,2,4]triazolo[4,3- a][1 ,4]diazepin-8-yl trifluoromethanesulfonate (for a preparation see Intermediate 3) (100 mg), bis(triphenylphosphine)palladium(ll) chloride (12.95 mg), potassium carbonate (128 mg) and 4-pyridineboronic acid (27.2 mg) were added to a 2-5 ml microwave vial. Toluene (2 ml) and ethanol (2 ml) were added to the vial and the reaction mixture was heated at 120°C for 20 min (microwave). The reaction mixture was filtered through Celite™ and the filter bed was washed with ethanol. The solvent was evaporated and the residue was chromatographed (0- 5% methanokDCM), triturated with diethyl ether and dried in a vacuum dessicator at 40°C overnight to give the product as a cream solid. (S)-2-(6-(4-chlorophenyl)-1 -methyl-8-(pyridin- 4-yl)-4H-benzo[f][1 ,2,4]triazolo[4,3-a][1 ,4]diazepin-4-yl)-N-ethylacetamide (51.7 mg).
LCMS (formate): MH+ 471 / 473, RT 0.67 min
Example 10: (S)-2-(6-(4-Chlorophenyl)-1 -methyl-8-(3-((4-methylpiperazin-1 - yl)methyl)phenyl)-4H-b -a][1,4]diazepin-4-yl)-N-ethylacetamide
Figure imgf000044_0001
(S)-2-(6-(4-Chlorophenyl)-8-(3-formylphenyl)-1 -methyl-4H-benzo[f][1 ,2,4]triazolo[4,3- a][1 ,4]diazepin-4-yl)-N-ethylacetamide (for a preparation see Intermediate 5) (75 mg) was dissolved in DCM (5 ml). 1 -methylpiperazine (25 μΙ) and acetic acid (8.62 μΙ) were added and the reaction mixture was stirred for 5 min. Sodium triacetoxyborohydride (160 mg) was added and the reaction mixture was stirred at room temperature, under nitrogen, overnight. Saturated sodium hydrogen carbonate solution (5 ml) was added and the reaction mixture was stirred for 10 min. The organic phase was separated and the saturated sodium hydrogen carbonate solution was extracted with DCM (10 ml). The DCM extracts were combined, dried and the solvent was evaporated. The residue was chromatographed (0-15 % methanohDCM), triturated with diethyl ether and dried in a vacuum dessicator at 40°C over the weekend to give (S)-2-(6-(4-chlorophenyl)-1-methyl-8-(3-((4-methylpiperazin-1- yl)methyl)phenyl)-4H-benzo[f][1 ,2,4]triazolo[4,3-a][1 ,4]diazepin-4-yl)-N-ethylacetamide (49.2 mg) as a white solid. LCMS (formate): MH+ 582 / 584, RT 0.77 min
Example 11 : (S)-2-(6-(4-Chlorophenyl)-8-(3-((ethylamino)methyl)phenyl)-1 -methyl-4H- benzo[f][1,2,4]triazolo[4,3 de
Figure imgf000044_0002
(S)-2-(6-(4-Chlorophenyl)-8-(3-formylphenyl)-1 -methyl-4H-benzo[f][1 ,2,4]triazolo[4,3- a][1 ,4]diazepin-4-yl)-N-ethylacetamide (for a preparation see Intermediate 5) (75 mg) was dissolved in DCM (5 ml). Ethylamine hydrochloride (18.42 mg) and acetic acid (8.62 μΙ) were added and the reaction mixture was stirred for 5 min. Sodium triacetoxyborohydride (160 mg) was added and the reaction mixture was stirred at room temperature, under nitrogen, overnight. Saturated sodium hydrogen carbonate solution (5 ml) was added and the reaction mixture was stirred for 10 min. The organic phase was separated and the saturated sodium hydrogen carbonate solution was extracted with DCM (10 ml). The DCM extracts were combined, dried and the solvent was evaporated. The residue was chromatographed (0-15 % methanokDCM) and triturated with diethyl ether. The solid was dried in a vacuum dessicator at 40°C over the weekend to give (S)-2-(6-(4-chlorophenyl)-8-(3- ((ethylamino)methyl)phenyl)-1 -methyl-4H-benzo[f][1 ,2,4]triazolo[4,3-a][1 ,4]diazepin-4-yl)-N- ethylacetamide (52.4 mg) as a white solid. LCMS (formate): MH+ 527, RT 0.75 min
Example 12: (S)-2-(6-(4-Chlorophenyl)-1 -methyl-8-(3-(pyrrolidin-1 -ylmethyl)phenyl)-4H- benzo[f][1,2,4]triazolo[4, -a][1 ,4]diazepin-4-yl)-N-ethylacetamide
Figure imgf000045_0001
(S)-2-(6-(4-Chlorophenyl)-8-(3-formylphenyl)-1 -methyl-4H-benzo[f][1 ,2,4]triazolo[4,3- a][1 ,4]diazepin-4-yl)-N-ethylacetamide (for a preparation see Intermediate 5) (95 mg) was dissolved in DCM (5 ml). Pyrrolidine (24 μΙ) and acetic acid (8.62 μΙ) were added and the reaction mixture was stirred for 5 min. Sodium triacetoxyborohydride (202 mg) was added and the reaction mixture was stirred at room temperature, under nitrogen, overnight. Saturated sodium hydrogen carbonate solution (5 ml) was added and the reaction mixture was stirred for 10 min. The organic phase was separated and the saturated sodium hydrogen carbonate solution was extracted with DCM (10 ml). The DCM extracts were combined, dried and the solvent was evaporated. The residue was chromatographed (0-15 % methanokDCM), triturated with diethyl ether and dried in a vacuum dessicator at 40°C over the weekend to give (S)-2-(6-(4-chlorophenyl)-1-methyl-8-(3-(pyrrolidin-1 - ylmethyl)phenyl)-4H-benzo[f][1 ,2,4]triazolo[4,3-a][1 ,4]diazepin-4-yl)-N-ethylacetamide (71.7 mg) as a white solid. LCMS (formate): MH+ 553 / 555, RT 0.76 min Example 13: (S)-2-(6-(4-Chlorophenyl)-1 -methyl-8-(4-(pyrrolidin-1 -ylmethyl)phenyl)-4H- benzo[f][1,2,4]triazolo[ -a][1 ,4]diazepin-4-yl)-N-ethylacetamide
Figure imgf000046_0001
(S)-2-(6-(4-Chlorophenyl)-8-(4-formylphenyl)-1 -methyMH-benzo[fl[1 ,2,4]triazolo[4,3- a][1 ,4]diazepin-4-yl)-N-ethylacetamide (for a preparation see Intermediate 6) (75 mg) was dissolved in DCM (5 mL). Pyrrolidine (19 μΙ) and acetic acid (8.62 μΙ) were added and the reaction mixture was stirred for 5 min. Sodium triacetoxyborohydride (160 mg) was added and the reaction mixture was stirred at room temperature, under nitrogen, overnight. Saturated sodium hydrogen carbonate solution (5 ml) was added and the reaction mixture was stirred for 10 min. The organic phase was separated and the saturated sodium hydrogen carbonate solution was extracted with DCM (10 ml). The DCM extracts were combined, dried and the solvent was evaporated. The residue was chromatographed (0-15 % methanokDCM), triturated with diethyl ether and dried in a vacuum dessicator at 40°C overnight to give (S)-2-(6-(4-chlorophenyl)-1-methyl-8-(4-(pyrrolidin-1-ylmethyl)phenyl)-4H- benzo[f][1 ,2,4]triazolo[4,3-a][1 ,4]diazepin-4-yl)-N-ethylacetamide (59.2 mg) as a white solid. LCMS (formate): MH+ 553, RT 0.69 min
NMR (D6-DMSO): δΗ 8.25(1 H, t), 8.10(1 H, dd), 7.94(1 H, d), 7.65(2H, d), 7.62(1 H, d), 7.55(2H, d), 7.49(2H, d), 7.40(2H, d), 4.55(1 H, m), 3.62(2H, bs), 3.29-3.07(4H, m), 2.61 (3H, s), 2.45(4H, bs), 1.70(41-1, bs), 1.07(31-1, t).
Example 14: (S)-2-(6-(4-Chlorophenyl)-1 -methyl-8-(4-((4-methylpiperazin-1 - yl)methyl)phenyl)-4H- -a][1,4]diazepin-4-yl)-N-ethylacetamide
Figure imgf000046_0002
(S)-2-(6-(4-Chlorophenyl)-8-(4-formylphenyl)-1 -methyl-4H-benzo[f][1 ,2,4]triazolo[4,3- a][1 ,4]diazepin-4-yl)-N-ethylacetamide (for a preparation see Intermediate 6) (90 mg) was dissolved in DCM (5 ml). 1 -Methylpiperazine (30 μΙ) and acetic acid (10.35 μΙ) were added and the reaction mixture was stirred for 5 min. Sodium triacetoxyborohydride (192 mg) was added and the reaction mixture was stirred at room temperature, under nitrogen, over the weekend. Saturated sodium hydrogen carbonate solution (5 ml) was added and the reaction mixture was stirred for 10 min. The organic phase was separated and the saturated sodium hydrogen carbonate solution was extracted with DCM (10 ml). The DCM extracts were combined, dried and the solvent was evaporated. The residue was chromatographed (0-15 % methanokDCM), triturated with diethyl ether and dried in a vacuum dessicator at 40°C overnight to give (S)-2-(6-(4-chlorophenyl)-1 -methyl-8-(4-((4-methylpiperazin-1- yl)methyl)phenyl)-4H-benzo[f][1 ,2,4]triazolo[4,3-a][1 ,4]diazepin-4-yl)-N-ethylacetamide (66.5 mg) as a white solid. LCMS (formate): MH+ 582, RT 0.65 min
Example 15: (S)-2-(6-(4-Chlorophenyl)-8-(4-((ethylamino)methyl)phenyl)-1 -methyl-4H- benzo[f][1,2,4]triazolo[ -a][1 ,4]diazepin-4-yl)-N-ethylacetamide
Figure imgf000047_0001
(S)-2-(6-(4-Chlorophenyl)-8-(4-formylphenyl)-1 -methyl-4H-benzo[f][1 ,2,4]triazolo[4,3- a][1 ,4]diazepin-4-yl)-N-ethylacetamide (for a preparation see Intermediate 6) (75 mg) was dissolved in DCM (5 ml). Ethylamine hydrochloride (18.42 mg) and acetic acid (8.62 μΙ) were added and the reaction mixture was stirred for 5 min. Sodium triacetoxyborohydride (160 mg) was added and the reaction mixture was stirred at room temperature, under nitrogen, over the weekend. Saturated sodium hydrogen carbonate solution (5 ml) was added and the reaction mixture was stirred for 10 min. The organic phase was separated and the saturated sodium hydrogen carbonate solution was extracted with DCM (10 ml). The DCM extracts were combined, dried and the solvent was evaporated. The residue was chromatographed (0-15 % methanokDCM), triturated with diethyl ether and dried in a vacuum dessicator at 40°C overnight to give (S)-2-(6-(4-chlorophenyl)-8-(4-((ethylamino)methyl)phenyl)-1-methyl- 4H-benzo[f][1 ,2,4]triazolo[4,3-a][1 ,4]diazepin-4-yl)-N-ethylacetamide (28.7 mg) as a white solid. LCMS (formate): MH+ 527 / 529, RT 0.67 min Example 16: (S)-2-(6-(4-Chlorophenyl)-8-(4-((dimethylamino)methyl)phenyl)-1 -methyl- 4H-benzo[f][1 ,2,4]triazo -a][1 ,4]diazepin-4-yl)-N-ethylacetamide
Figure imgf000048_0001
(S)-2-(6-(4-Chlorophenyl)-8-(4-formylphenyl)-1 -methyMH-benzo[fl[1 ,2,4]triazolo[4,3- a][1 ,4]diazepin-4-yl)-N-ethylacetamide (for a preparation see Intermediate 6) (75 mg) was dissolved in DCM (5 ml). Dimethylamine hydrochloride (18.42 mg) and acetic acid (8.62 μΙ) were added and the reaction mixture was stirred for 5 min. Sodium triacetoxyborohydride (160 mg) was added and the reaction mixture was stirred at room temperature, under nitrogen, for 5 h. Saturated sodium hydrogen carbonate solution (5 ml) was added and the reaction mixture was stirred for 10 min. The organic phase was separated and the saturated sodium hydrogen carbonate solution was extracted with DCM (10 ml). The DCM extracts were combined, dried and the solvent was evaporated. The residue was chromatographed (0-15 % methanohDCM), triturated with diethyl ether and dried in a vacuum dessicator overnight to give (S)-2-(6-(4-chlorophenyl)-8-(4-((dimethylamino)methyl)phenyl)-1-methyl- 4H-benzo[f][1 ,2,4]triazolo[4,3-a][1 ,4]diazepin-4-yl)-N-ethylacetamide (48.2 mg) as a white solid. LCMS (formate): MH+ 527 / 529, RT 0.66 min
Example 17: (S)-2-(6-(4-Chlorophenyl)-1 -methyl-8-(2H-tetrazol-5-yl)-4H- benzo[f][1,2,4]triazolo[4,3-a][1 ,4]diazepin-4-yl)-N-ethylacetamide
Figure imgf000048_0002
(S)-2-(6-(4-Chlorophenyl)-8-cyano-1-methyl-4H-benzo[f][1 ,2,4]triazolo[4,3-a][1 ,4]diazepin-4- yl)-N-ethylacetamide (for a preparation see Intermediate 7) (68.4 mg) was dissolved in DMF (5 ml). Triethylamine hydrochloride (45.0 mg) and sodium azide (21.23 mg) were added and the reaction mixture was heated at 100°C, under nitrogen, overnight. The temperature was raised to 120°C and the reaction mixture was heated overnight. The reaction mixture was allowed to cool to room temperature, partitioned between ethyl acetate and water. The organic phase was washed with water (2 x 10 ml) and brine (10 ml). The aqueous layer was passed through a pre equilibrated Biotage 103 column. The organic content was eluted with methanol, the methanolic solution was evaporated and the residue was chromatographed (10-20% methanokDCM). Trituration with diethyl ether gave (S)-2-(6-(4-chlorophenyl)-1 - methyl-8-(2H-tetrazol-5-yl)-4H-benzo[f][1 ,2,4]triazolo[4,3-a][1 ,4]diazepin-4-yl)-N- ethylacetamide (25.6 mg) as a yellow solid. LCMS (formate): MH+ 462 / 464, RT 0.75 min
Reference Compounds
Experimental details of LC/MS methods A and B as referred to in the Reference compounds described below are as follows:
LC/MS (Method A) was conducted on a Supelcosil LCABZ+PLUS column (3μηι, 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 B) was conducted on an Sunfire C18 column (30mm x 4.6mm i.d. 3.5μηη 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 (3μηη 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.
Reference compound A : 2-methyl-6-(methyloxy)-4/-/-3,1 -benzoxazin-4-one
Figure imgf000050_0001
A solution of 5-methoxyanthranilic acid (Lancaster) (41 .8 g, 0.25 mol) was refluxed in acetic anhydride (230 mL) 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 A): m/z 192 [M+H]+, Rt 1.69 min.
Reference compound B: r2-amino-5-(methyloxy)phenyl1(4-chlorophenyl)methanone
Figure imgf000050_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 (760 mL) at 0°C was added dropwise a solution of 4-chlorophenylmagnesium bromide (170 mL, 1 M in Et20, 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 (200 mL). The aqueous layer was extracted with EtOAc (3 x 150 mL) and the combined organics were washed with brine (100 mL), dried over Na2S04, filtered and concentrated under reduced pressure. The crude compound was then dissolved in EtOH (400 mL) and 6N HCI (160 mL) 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 NaOH. The aqueous layer was extracted with EtOAc (3 x 150 mL) and the combined organics were washed with brine (150 mL), 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 A): m/z 262 [M+H]+, Rt 2.57 min.
Reference Compound C: Methyl /V1-r2-r(4-chlorophenyl)carbonyll-4-(methyloxy)phenyll-/V2- {[(9/-/-fluoren-9-ylmethyl)oxylcarbonyl}-L-a-asparaginate
Figure imgf000051_0001
Methyl /V-{[(9H-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 (270 ml.) 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 r(3S)-5-(4-chlorophenyl)-7-(methyloxy)-2-oxo-2,3-dihvdro-
1 H-1 ,4-benzodiazepin-3-yllaceta
Figure imgf000051_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 (500 ml.) was added Et3N (500 ml_, 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 (104 ml_, 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 (M+H)+ calculated for Ci9H18 35CIN204 373.0955; found 373.0957.
Reference compound E: Methyl [(3S)-5-(4-chlorophenyl)-7-(methyloxy)-2-thioxo-2,3-dihvdro-
1 H-1 ,4-benzodiazepin-3-yllacetat
Figure imgf000052_0001
A suspension of P4Si0 (36.1 g, 81.1 mmol) and Na2C03 (8.6 g, 81.1 mmol) in 1 ,2-DCE (700 ml.) at room temperature was stirred for 2 h before Methyl [(3S)-5-(4-chlorophenyl)-7- (methyloxy)-2-oxo-2,3-dihydro-1 H-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. NaHC03 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 A): m/z 389 [M(35CI)+H]+, Rt 2.64 min
HRMS (M+H)+ calculated for Ci9H18 35CIN203S 389.0727; found 389.0714.
Reference compound F: Methyl r(3S)-2-r(1 Z)-2-acetylhvdrazinol-5-(4-chlorophenyl)-7- (methyloxy)-3H-1 ,4-benzodiazepin-3-yllacetate
Figure imgf000052_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 (300 mL) at 0°C was added hydrazine monohydrate (3.4 mL, 69.6 mmol) dropwise. The reaction mixture was stirred for 5h between 5°C and 15°C before being cooled at 0°C. Et3N (9.7 mL, 69.6 mmol) was then added slowly and acetyl chloride (7.95 mL, 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 ,2,41triazolor4,3-ain ,41benzodiazepin-4-yllacetate
Figure imgf000053_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 (100 ml) and AcOH (60 mL) 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 (M+H)+ calculated for C2i H2oCIN403 41 1.1229; found 41 1.1245.
Reference compound Hj [(4S)-6-(4-Chlorophenyl)-1-methyl-8-(methyloxy)-4H-
[1 ,2,4ltriazolo[4,3-al[1 ,41benzodiazepin-4-yllacetic acid
Figure imgf000053_0002
To a solution of Methyl [(4S)-6-(4-chlorophenyl)-1 -methyl-8-(methyloxy)-4H- [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 (130 ml.) at room temperature was added 1 N NaOH (36.2 ml_, 36.2 mmol). The reaction mixture was stirred at this temperature for 5h before being quenched with 1 N HCI (36.2 ml.) 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 I: 1 ,1-dimethylethyl [5-({[(4S)-6-(4-chlorophenyl)-1-methyl-8-
(methyloxy)-4H-[1 ,2,4ltriazolo[4,3-al[1 ,4lbenzodiazepin-4-yllacetyl}amino)pentyllcarbamate
Figure imgf000054_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 H) (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 A): rt = 3.04 min.
Reference compound J: N-(5-aminopentyl)-2-[(4S)-6-(4-chlorophenyl)-1-methyl-8- (methyloxy)-4H-[1 ,2,4ltriazolo[4,3-al[1 ,41benzodiazepin-4-yllacetamide trifluoroacetate
Figure imgf000055_0001
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 I) (0.2 g, 0.34 mmol) in dichloromethane (3 ml) was added trifluoroacetic acid (0.053 ml, 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 A): rt = 2.33min.
HRMS (M+H)+ calculated for CzsHzgCINeOz 481.21 19; found 481.2162.
Reference compound K: Mixture of 5- and 6- isomers of Alexa Fluor 488-N-(5-aminopentyl)-
2-r(4S)-6-(4-chloroDhenvn-1 -methyl-8-(methyloxy)-4H-n.2.41triazolor4.3- ain.41benzodiazepin-4-yllacetamide
Figure imgf000055_0002
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 Ν,Ν-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 B):, MH+ = 999, rt = 1 .88min.
Biological Test Methods
The compounds of formula (I) may be tested in the following assays. Fluorescence anisotropy binding assay
The binding of the compounds of formula (I) to Bromodomain 2, 3 and 4 can be assessed using an 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 are normalized to the mean of 16 high and 16 low control wells on each plate. A four parameter curve fit of the following form are then applied: y = a + (( b - a) / ( 1 + ( 10 A x / 10 A c ) A d )
Where 'a' is the minimum, 'b' is the Hill slope, 'c' is the plC50 and 'd' is the maximum.
Recombinant Human Bromodomains (Bromodomain 2 (1 -473), Bromodomain 3 (1 -435) and Bromodomain 4 (1 -477)) are 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 are then purified by affinity chromatography on a HisTRAP HP column, eluting with a linear 10-500mM Imidazole gradient, over 20 Cv. Further purification are completed by Superdex 200 prep grade size exclusion column. Purified protein are stored at -80C in 20mM HEPES pH 7.5 and 100mM NaCI. Protocol for Bromodomain 2: All components are 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 are 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 are read in Envision
Figure imgf000057_0001
485nm, λΕΜ = 530nm; Dichroic -505nM).
Protocol for Bromodoamin 3: All components are 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 are 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 are read in Envision
Figure imgf000057_0002
485nm, λΕΜ = 530nm; Dichroic -505nM).
Protocol for Bromodomain 4: All components are 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 are read in Envision
Figure imgf000057_0003
485nm, λΕΜ = 530nm; Dichroic -505nM).
Time Resolved Fluorescence Resonance Energy Transfer (TR-FRET) assay
The binding of the compounds of formula (I) to Bromodomains BRD2, BRD3 and BRD4 was assessed using a time resolved fluorescent resonance energy transfer binding assay, that measures the binding of an acetylated histone peptide to the bromodomain protein.
The bromodomain protein, histone peptide and a variable concentration of test compound are incubated together to reach thermodynamic equilibrium. The assay is configured such that in the absence of test compound the bromodomain and peptide are significantly bound (-30%) and in the presence of a sufficient concentration of a potent inhibitor this interaction is disrupted leading to a measurable drop in fluorescent resonance energy transfer. Histone Peptide:
H-Ser-Gly-Arg-Gly-Lys(Ac)-Gly-Gly-Lys(Ac)-Gly-Leu-Gly-Lys(Ac)-Gly-Gly-Ala-Lys(Ac)-Arg- His-Gly-Ser-Gly-Ser-Lys(Biotin)-OH. 3TFA
The protected peptide was assembled on a solid-phase synthesiser using preloaded Wang resin and utilising standard Fmoc synthesis protocols. The C-terminal lysine was protected by a hyper acid-labile group allowing for its selective removal at the end of the assembly and attachment of the biotin. The crude peptide was obtained after cleavage from the resin with a mixture of trifluoroacetic acid (TFA), triisopropylsilane and water (95:2.5:2.5) for 3h at room temperature and was then purified using a C18 reverse-phase column utilising a 0.1 %TFA- buffered water/acetonitrile gradient. The resulting fractions were analysed and fractions which were >95% pure by analytical HPLC and giving the correct mw (by MALDiTOF mass spectroscopy) were pooled and freeze dried. The final material was analysed by HPLC to confirm purity.
Protein production: Recombinant Human Bromodomains (BRD2 (1-473), BRD3 (1-435) and 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 sonication and purified using a nickel sepharose 6FF column, the proteins were washed and then eluted with 50mM Tris-Hcl pH8.0. 300mM NaCI, 1 mM β-mercaptoethanol and 20mM Imidazole. Further purification was performed by affinity chromatography on a HisTRAP HP column, eluting with a linear 0-500mM sodium chloride gradient, over 20 column volumes. Final purification was completed by Superdex 200 prep grade size exclusion column. Purified protein was stored at -80C in 20mM HEPES pH 7.5 and 100mM NaCI. Protein identity was confirmed by peptide mass fingerprinting and predicted molecular weight confirmed by mass spectrometry.
Protocol for Bromodomain BRD2, 3 and 4 assays: All assay components were dissolved in buffer composition of 50 mM HEPES pH7.4, 50mM NaCI and 0.5mM CHAPS. The final concentration of bromodomain proteins were 100nM and the histone peptide was 300nM, these components are premixed and allowed to equilibrate for 1 hour in the dark. 8 μΙ of this reaction mixture was added to all wells containing 50nl of various concentrations of test compound or DMSO vehicle (0.5% final) in Greiner 384 well black low volume microtitre plates and incubated in dark for 60 mins at room temperature. 2μΙ of detection mixture containing anti-6his XL665 labeled antibody and streptavidin labeled with europium cryptate was added to all wells and a further dark incubation of at least 30mins was performed. Plates were then read on the Envision platereader, 317nm, donor λΕΜ = 615nm; acceptor λΕΜ = 665nm; Dichroic LANCE dual). Time resolved fluorescent intensity measurements were made at both emission wavelengths and the ratio of acceptor/donor was calculated and used for data analysis. 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 A x / 10 A c ) A d )
Where 'a' is the minimum, 'b' is the Hill slope, 'c' is the plC50 and 'd' is the maximum.
Examples 1 and 3-17 were tested in one or more of the above assays and were found to have a plC50≥ 5.5. Examples 1 , 3-5 and 7-17 were tested in one or more of the above assays and were found to have a plC50≥ 6.5.
LPS stimulated Whole Blood measuring TNFa 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 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 TNFa levels assayed by immunoassay (typically by MesoScale Discovery technology) either immediately or following storage at -20 degrees. Dose response curves for each compound is generated from the data and an IC50 value is calculated.
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 , 3, 4, 5 and 13 were tested in the above assay were found to have a plC50≥ 5.5.
These data demonstrate that bromodomain inhibitors tested in the above whole blood assay inhibited the production of key inflammatory mediator 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 of formula (I) groups of eight Balb/c male mice are given a lethal dose of 15 mg/kg LPS by intraperitoneal injection. Ninety minutes later, animals are dosed intravenously with vehicle (20% cyclodextrin 1 % ethanol in apyrogen water) or compound (10 mg/kg). The survival of animals is monitored at 4 days.
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 are 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.
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

1. A compound of formula (I) or a salt thereof
Figure imgf000062_0001
where
R1 is C<|_3alkyl;
R2 is -NR2aR2a' or -OR2b;
wherein one of R2a or R2 a is hydrogen, and R^b or the other of R2a or R2 a are selected from C-i.galkyl, haloC-|_galkyl, R2cR2'cN-C2-6alkyl, R2cO-C2-6alkyl carbocyclyl, carbocyclylC-|_4alkyl, heterocyclyl and heterocyclylC-|.4alkyl,
wherein any of the carbocyclyl or heterocyclyl groups are optionally substituted by one or more groups selected from halogen, C-|.galkyl, haloC-|.galkyl, C-|.galkoxy, haloC-|.galkoxy, carbonyl, -CO-carbocyclyl, amino, hydroxyl and cyano,
or two adjacent groups on any of the carbocyclyl or heterocyclyl groups together with the interconnecting atoms form a 5- or 6-membered ring which ring may contain 1 or 2 heteroatoms independently selected from O, S and N;
or R2a and R2'a together with the N-atom to which they are attached form a 5-, 6- or 7- membered ring which may optionally contain 1 or 2 heteroatoms independently selected from O, S and N; wherein the 5-, 6- or 7-membered ring may be further optionally substituted by C-|.galkyl;
R2c and R2'c are independently hydrogen or C-|.galkyl;
each R3 is independently selected from hydrogen, hydroxy, halo, C-|.galkyl, halo C-|.galkyl,
C-|.galkoxy, halo C-|.galkoxy, cyano, CF3, -OCF3, -COOR5, -C-|_4alkylNR6R7 and -C<|_ 4alkylOH; R4 is an aromatic carbocyclic or aromatic heterocyclyl group;
wherein the aromatic carbocyclyl or heterocyclyl group is optionally substituted by one or more groups selected from halo, C-|.galkyl, C-|.galkoxy and a group C-^alkylNR^R7;
R5 is C<|_3alkyl;
At each occurrence R^ and R7 are independently hydrogen, C-|.galkyl or C(0)C-|.galkyl, or
R6 and R7 together with the N-atom to which they are attached form a 5-, 6- or 7-membered ring which optionally contains 1 or 2 further heteroatoms independently selected from O, S and N; wherein the 5-, 6- or 7-membered ring may be further optionally substituted by one or more C-|.galkyl, -C(0)C-|.galkyl, amino or hydroxyl groups; and n is an integer 1 to 5.
2. A compound or a salt thereof according to claim 1 , wherein R^ is methyl.
3. A compound or a salt thereof according to claim 1 or claim 2, wherein R2 is OR^b.
4. A compound or a salt thereof according to claim 3, wherein R2D js C-|.galkyl.
5. A compound or a salt thereof according to claim 4, wherein R2D js selected from methyl, ethyl, isopropyl and butyl.
6. A compound or a salt thereof according to claim 1 or claim 2, wherein R2 IS - NR2aR2a'.
7. A compound or a salt thereof according to claim 6, wherein R2a js hydrogen and R2'a is C-|.galkyl.
8. A compound or a salt thereof according to claim 7, wherein R2'a is ethyl.
9. A compound or a salt thereof according to claim 6, wherein R2a js hydrogen and R2'a js R2cR2'cN.Cl 6a|ky|.
10. A compound or a salt thereof according to claim 9 wherein R2 a js selected from R2cR2'cN-CH2CH2-, R2cR2'cN-CH2CH2CH2- and R2cR2'cN-CH2CH2CH2CH2-.
1 1. A compound or a salt thereof according to claim 10, wherein R2c and R2'c are selected from hydrogen and methyl.
12. A compound or a salt thereof according to claim 6, wherein R2a is hydrogen and R2 a is carbocyclyl.
13. A compound or a salt thereof according to claim 12, wherein R2'a is cyclopentyl or cyclohexyl, wherein each group is optionally substituted once by amino or hydroxyl.
14. A compound or a salt thereof according to claim 6, wherein R2a is hydrogen and R2 a is heterocyclyl.
15. A compound or a salt thereof according to claim 14, wherein R2 a is selected from pyrrolidinyl, piperidinyl, tetrahydrofuranyl and tetrahydropyran, wherein each group is optionally substituted by C-| .galkyl.
A compound or a salt thereof according to claim 15, wherein R2 a is selected from
Figure imgf000064_0001
17. A compound or a salt thereof according to claim 6, wherein R2a is hydrogen and R2'a is heterocyclylC-|_4alkyl, wherein the heterocyclyl is optionally substituted once by amino, hydroxyl or methyl.
18. A compound or a salt thereof according to claim 17 wherein R2'a is selected from heterocyclyl-CH2-, heterocyclyl-CH2CH2-, heterocyclyl-CH2CH2CH2- and heterocyclyl- CH2CH2CH2CH2-, wherein the heterocyclyl is optionally substituted once by amino, hydroxyl or methyl.
19. A compound or a salt thereof according to claim 18, wherein heterocyclyl is selected from tetrahydrofuranyl, pyrrolidinyl, piperidinyl, tetrahydropyranyl, piperazinyl, morpholino and thiomorpholinodioxide, wherein each group is optionally substituted once by amino, hydroxyl or methyl.
A compound or a salt thereof according to claim 18, wherein heterocyclyl is selected
Figure imgf000065_0001
21. A compound or a salt thereof according to any one of claims 1 - 20, wherein n is 1.
22. A compound or a salt thereof according to any one of claims 1 - 21 , wherein R3 is 3-fluoro, 4-chloro, 4-fluoro, 4-methoxy or 4-CF3.
23. A compound or a salt thereof according to claim 22, wherein R3 is 4-chloro.
24. A compound or a salt thereof according to any one of claims 1 - 23, wherein is phenyl, pyridyl, imadolzolyl or pyrazolyl, such groups being optionally substituted by one or more groups selected from halo, C-|.galkyl, C-|.galkoxy or a group C-^alkylNR^R? in which R6 is hydrogen and R7 is hydrogen, C-|.galkyl or C(0)C-|.galkyl.
Figure imgf000066_0001
in which R^ is hydrogen, methyl or C(0)Me.
26. A compound or a salt thereof according to any one of claims 1 - 23 wherein R^ is phenyl substituted by a group C-^alkylNR^R? in which R6 and R^ together with the N-atom to which they are attached form a pyrolidinyl, piperazinyl, piperidinyl or morpholinyl ring said rings being optionally substituted by one or more C-|.galkyl, -C(0)C-|.galkyl, amino or hydroxyl groups.
27. A compound or a salt thereof according to any one of claims 1 - 26 in which the R4 group is at the 8 position of the benzodiazepine ring.
28. A compound which is selected from
2-[(4S)-6-(4-Chlorophenyl)-1-methyl-8-phenyl-4H-[1 ,2,4]triazolo[4,3-a][1 ,4]benzodiazepin-4- yl]-N-ethylacetamide;
Ethyl [(4S)-6-(4-Chlorophenyl)-1-methyl-8-(4-pyridinyl)-4H-[1 ,2,4]triazolo[4,3- a][1 ,4]benzodiazepin-4-yl]acetate;
2-{(4S)-6-(4-Chlorophenyl)-1 -methyl-8-[4-(methyloxy)phenyl]-4H-[1 ,2,4]triazolo[4,3- a][1 ,4]benzodiazepin-4-yl}-N-ethylacetamide;
2-[(4S)-6-(4-Chlorophenyl)-1-methyl-8-(1 H-pyrazol-5-yl)-4H-[1 ,2,4]triazolo[4,3- a][1 ,4]benzodiazepin-4-yl]-N-ethylacetamide;
2-[(4S)-6-(4-Chlorophenyl)-1-methyl-8-(1 H-pyrazol-4-yl)-4H-[1 ,2,4]triazolo[4,3- a][1 ,4]benzodiazepin-4-yl]-N-ethylacetamide;
2-[(4S)-6-(4-Chlorophenyl)-1-methyl-8-(2-methylphenyl)-4H-[1 ,2,4]triazolo[4,3- a][1 ,4]benzodiazepin-4-yl]-N-ethylacetamide;
2-[(4S)-6-(4-Chlorophenyl)-1-methyl-8-(3-pyridinyl)-4H-[1 ,2,4]triazolo[4,3- a][1 ,4]benzodiazepin-4-yl]-N-ethylacetamide;
(S)-2-(6-(4-Chlorophenyl)-8-(3-methoxyphenyl)-1-methyl-4H-benzo[f][1 ,2,4]triazolo[4,3- a][1 ,4]diazepin-4-yl)-N-ethylacetamide;
(S)-2-(6-(4-Chlorophenyl)-1-methyl-8-(pyridin-4-yl)-4H-benzo[f][1 ,2,4]triazolo[4,3- a][1 ,4]diazepin-4-yl)-N-ethylacetamide; (S)-2-(6-(4-Chlorophenyl)-1-methyl-8-(3-((4-methylpiperazin-1 -yl)methyl)ph
benzo[f][1 ,2,4]triazolo[4,3-a][1 ,4]diazepin-4-yl)-N-ethylacetamide;
(S)-2-(6-(4-Chlorophenyl)-8-(3-((ethylamino)methyl)phenyl)-1 -methyl-4H- benzo[f][1 ,2,4]triazolo[4,3-a][1 ,4]diazepin-4-yl)-N-ethylacetamide ;
(S)-2-(6-(4-Chlorophenyl)-1-methyl-8-(3-(pyrrolidin-1-ylmethyl)phenyl)-4H- benzo[f][1 ,2,4]triazolo[4,3-a][1 ,4]diazepin-4-yl)-N-ethylacetamide;
(S)-2-(6-(4-Chlorophenyl)-1-methyl-8-(4-(pyrrolidin-1-ylmethyl)phenyl)-4H- benzo[f][1 ,2,4]triazolo[4,3-a][1 ,4]diazepin-4-yl)-N-ethylacetamide;
(S)-2-(6-(4-Chlorophenyl)-1-methyl-8-(4-((4-methylpiperazin-1 -yl)methyl)phen
benzo[f][1 ,2,4]triazolo[4,3-a][1 ,4]diazepin-4-yl)-N-ethylacetamide;
(S)-2-(6-(4-Chlorophenyl)-8-(4-((ethylamino)methyl)phenyl)-1 -methyl-4H- benzo[f][1 ,2,4]triazolo[4,3-a][1 ,4]diazepin-4-yl)-N-ethylacetamide;
(S)-2-(6-(4-Chlorophenyl)-8-(4-((dimethylamino)methyl)phenyl)-1 -meth
benzo[f][1 ,2,4]triazolo[4,3-a][1 ,4]diazepin-4-yl)-N-ethylacetamide; and
(S)-2-(6-(4-Chlorophenyl)-1-methyl-8-(2H-tetrazol-5-yl)-4H-benzo[f][1 ,2,4]triazolo[4,3- a][1 ,4]diazepin-4-yl)-N-ethylacetamide
or a salt thereof.
29 A compound according to any one of claims 1 - 28, or a pharmaceutically acceptable salt thereof.
30. A pharmaceutical composition which comprises a compound of formula (I) or a pharmaceutically acceptable salt thereof as defined in claim 29 and one or more pharmaceutically acceptable carriers, diluents or excipients.
31. A combination pharmaceutical product comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof as defined in claim 29 together with one or more other therapeutically active agents.
32. A compound of formula (I) or a pharmaceutically acceptable salt thereof as defined in claim 29, for use in therapy.
33. A compound of formula (I) or a pharmaceutically acceptable salt thereof as defined in claim 29, for use in the treatment of diseases or conditions for which a bromodomain inhibitor is indicated.
34. A compound or a pharmaceutically acceptable salt thereof for use according to claim 33, wherein the disease or condition is a chronic autoimmune and/or inflammatory condition.
35. A compound or a pharmaceutically acceptable salt thereof for use according to claim 33, wherein the disease or condition is cancer.
36. The use of a compound of formula (I) or a pharmaceutically acceptable salt thereof as defined in claim 29, in the manufacture of a medicament for the treatment of diseases or conditions for which a bromodomain inhibitor is indicated.
37. A method for treatment of a disease or condition, 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, as defined in claim 29.
38. A method for treatment according to claim 37, wherein the disease or condition is a chronic autoimmune and/or inflammatory condition.
39 A method for treatment according to claim 37, wherein the disease or condition is cancer.
40. A method for treatment according to claim 37 - 39 wherein the subject is a human.
41. A method for inhibiting a bromodomain which comprises contacting the bromodomain with a compound or a pharmaceutically acceptable salt thereof, as defined in claim 29.
PCT/EP2011/060179 2010-06-22 2011-06-20 Benzotriazolodiazepine compounds inhibitors of bromodomains WO2011161031A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
US13/702,177 US9085582B2 (en) 2010-06-22 2011-06-20 Benzotriazolodiazepine compounds inhibitors of bromodomains
JP2013515827A JP5844358B2 (en) 2010-06-22 2011-06-20 Bromodomain inhibitors containing benzotriazolodiazepine compounds
EP11725769.1A EP2585465B1 (en) 2010-06-22 2011-06-20 Benzotriazolodiazepine compounds inhibitors of bromodomains
ES11725769.1T ES2526671T3 (en) 2010-06-22 2011-06-20 Compounds of benzotriazoldiazepine bromodomain inhibitors

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
GBGB1010514.6A GB201010514D0 (en) 2010-06-22 2010-06-22 Novel compounds
GB1010514.6 2010-06-22
GB1106801.2 2011-04-21
GBGB1106801.2A GB201106801D0 (en) 2011-04-21 2011-04-21 Novel compounds

Publications (1)

Publication Number Publication Date
WO2011161031A1 true WO2011161031A1 (en) 2011-12-29

Family

ID=44310320

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2011/060179 WO2011161031A1 (en) 2010-06-22 2011-06-20 Benzotriazolodiazepine compounds inhibitors of bromodomains

Country Status (5)

Country Link
US (1) US9085582B2 (en)
EP (1) EP2585465B1 (en)
JP (1) JP5844358B2 (en)
ES (1) ES2526671T3 (en)
WO (1) WO2011161031A1 (en)

Cited By (77)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013027168A1 (en) 2011-08-22 2013-02-28 Pfizer Inc. Novel heterocyclic compounds as bromodomain inhibitors
WO2013184876A1 (en) * 2012-06-06 2013-12-12 Constellation Pharmaceuticals, Inc. Benzo [c] isoxazoloazepine bromodomain inhibitors and uses thereof
WO2013184878A1 (en) * 2012-06-06 2013-12-12 Constellation Pharmaceuticals, Inc. Benzo [b] isoxazoloazepine bromodomain inhibitors and uses thereof
WO2014076146A1 (en) * 2012-11-13 2014-05-22 Boehringer Ingelheim International Gmbh Triazolopyridazine
US8796261B2 (en) 2010-12-02 2014-08-05 Constellation Pharmaceuticals, Inc. Bromodomain inhibitors and uses thereof
US20140243321A1 (en) * 2011-08-29 2014-08-28 Coferon, Inc. Bioorthogonal monomers capable of dimerizing and targeting bromodomains, and methods of using same
US20140243286A1 (en) * 2011-08-29 2014-08-28 Coferon, Inc. Bromodomain ligands capable of dimerizing in an aqueous solution, and methods of using same
WO2014128070A1 (en) * 2013-02-22 2014-08-28 Bayer Pharma Aktiengesellschaft Pyrrolo- and pyrazolo-triazolodiazepines as bet-protein inhibitors for treating hyperproliferative diseases
WO2014164771A1 (en) * 2013-03-11 2014-10-09 Abbvie Inc. Bromodomain inhibitors
WO2014173241A1 (en) 2013-04-26 2014-10-30 Beigene, Ltd. Substituted5-(3,5-dimethylisoxazol-4-yl)indoline-2-ones
CN104136435A (en) * 2011-12-30 2014-11-05 艾伯维公司 Bromodomain inhibitors
WO2015013635A2 (en) 2013-07-25 2015-01-29 Dana-Farber Cancer Institute, Inc. Inhibitors of transcription factors and uses thereof
US8969341B2 (en) 2011-11-29 2015-03-03 Novartis Ag Pyrazolopyrrolidine compounds
US8975417B2 (en) 2013-05-27 2015-03-10 Novartis Ag Pyrazolopyrrolidine derivatives and their use in the treatment of disease
US8980879B2 (en) 2013-03-11 2015-03-17 Abbvie Inc. Bromodomain inhibitors
US9051279B2 (en) 2009-12-22 2015-06-09 Novartis Ag Substituted isoquinolinones and quinazolinones
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
JP2015531362A (en) * 2012-09-28 2015-11-02 バイエル ファーマ アクチエンゲゼルシャフト BET protein inhibitory 5-aryltriazoloazepines
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
US9290514B2 (en) 2013-07-08 2016-03-22 Incyte Holdings Corporation Tricyclic heterocycles as BET protein inhibitors
JP2016510772A (en) * 2013-03-12 2016-04-11 アッヴィ・インコーポレイテッド Tetracyclic 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
US9365576B2 (en) 2012-05-24 2016-06-14 Novartis Ag Pyrrolopyrrolidinone compounds
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
US9403827B2 (en) 2013-01-22 2016-08-02 Novartis Ag Substituted purinone compounds
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
WO2016196065A1 (en) 2015-05-29 2016-12-08 Genentech, Inc. Methods and compositions for assessing responsiveness of cancers to bet 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
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
US9550796B2 (en) 2013-11-21 2017-01-24 Novartis Ag Pyrrolopyrrolone derivatives and their use as BET inhibitors
US9556180B2 (en) 2013-01-22 2017-01-31 Novartis Ag Pyrazolo[3,4-d]pyrimidinone compounds as inhibitors of the P53/MDM2 interaction
US9624247B2 (en) 2013-05-28 2017-04-18 Novartis Ag Pyrazolo-pyrrolidin-4-one derivatives as bet inhibitors and their use in the treatment of disease
US9695172B2 (en) 2014-01-31 2017-07-04 Dana-Farber Cancer Institute, Inc. Diazepane derivatives and uses thereof
US9714249B2 (en) 2013-05-28 2017-07-25 Novartis Ag Pyrazolo-pyrrolidin-4-one derivatives and their use in the treatment of disease
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
WO2017176958A1 (en) 2016-04-06 2017-10-12 The Regents Of The University Of Michigan Monofunctional intermediates for ligand-dependent target protein degradation
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
US9890166B2 (en) 2013-05-27 2018-02-13 Novartis Ag Imidazopyrrolidine derivatives and their use in the treatment of disease
CN107759607A (en) * 2017-11-29 2018-03-06 河南龙湖生物技术有限公司 Triazole with antitumor activity and phenodiazine Zhuo compound and preparation method thereof
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
WO2018144789A1 (en) 2017-02-03 2018-08-09 The Regents Of The University Of Michigan Fused 1,4-diazepines as bet 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
US10189832B2 (en) 2016-06-20 2019-01-29 Incyte Corporation Crystalline solid forms of a BET inhibitor
WO2019055444A1 (en) 2017-09-13 2019-03-21 The Regents Of The University Of Michigan Bet bromodomain protein degraders with cleavable linkers
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
US10328074B2 (en) 2012-11-16 2019-06-25 Boehringer Ingelheim International Gmbh Substituted [1,2,4]triazolo[4,3-a]pyrazines as BRD4 inhibitors
US10407441B2 (en) 2010-05-14 2019-09-10 Dana-Farber Cancer Institute, Inc. Compositions and methods for treating neoplasia, inflammatory disease and other disorders
WO2020020288A1 (en) 2018-07-25 2020-01-30 正大天晴药业集团股份有限公司 Sulfoximine compound as bromodomain protein inhibitor and pharmaceutical composition and medical use thereof
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
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
WO2021175824A1 (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
WO2021262731A2 (en) 2020-06-23 2021-12-30 Genentech, Inc. Macrocyclic compounds and methods of use thereof
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
US11759527B2 (en) 2021-01-20 2023-09-19 Abbvie Inc. Anti-EGFR antibody-drug conjugates
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 (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014164596A1 (en) 2013-03-11 2014-10-09 The Regents Of The University Of Michigan Bet bromodomain inhibitors and therapeutic methods using the same
GB201320994D0 (en) * 2013-11-28 2014-01-15 Univ Dundee Enzyme function probes
EP3277683B1 (en) 2015-03-30 2022-03-09 Jubilant Biosys Limited Tricyclic fused derivatives of 1-(cyclo)alkyl pyridin-2-one useful for the treatment of cancer
EP3490552B1 (en) 2016-07-26 2022-11-23 University of Southern California Selective bromodomain inhibition of fungal bdf1

Citations (4)

* 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
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
CA2710740A1 (en) * 2007-12-28 2009-07-09 Shinji Miyoshi Thienotriazolodiazepine compound as antitumor agent

Family Cites Families (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE3610848A1 (en) 1986-04-01 1987-10-15 Boehringer Ingelheim Kg NEW 1,4-DIAZEPINE
MC1999A1 (en) * 1987-12-18 1990-01-26 Hoffmann La Roche TRICYCLIC COMPOUNDS
KR100255493B1 (en) 1991-10-11 2000-08-01 가마쿠라 아키오 Remedy for osteoporosis and diazepine compounds
JPH06128257A (en) 1992-10-13 1994-05-10 Yoshitomi Pharmaceut Ind Ltd Pyridodiazepine compound
EP0730596A4 (en) 1993-11-22 1997-07-30 Merck & Co Inc Benzodiazepines
WO1996016062A1 (en) 1994-11-18 1996-05-30 Japan Tobacco Inc. Osteoporosis remedy and triazepine compound
CA2258053A1 (en) 1996-06-12 1997-12-18 Yoshihisa Yamamoto Cytokine production inhibitors, triazepine compounds, and intermediates thereof
EP0989131B1 (en) 1996-09-13 2002-11-13 Mitsubishi Pharma Corporation Thienotriazolodiazepine compounds and medicinal uses thereof
SG115845A1 (en) 2004-03-31 2005-10-28 Univ Singapore Modulation of trip-br function and method of treating proliferative disorders
GB0420970D0 (en) 2004-09-21 2004-10-20 Smithkline Beecham Corp Novel triazoloquinoline compounds
EP1888594A2 (en) * 2005-05-16 2008-02-20 Wisys Technology Foundation, Inc. Gabaergic agents to treat memory deficits
EP1887008B1 (en) 2005-05-30 2021-04-21 Mitsubishi Tanabe Pharma Corporation Thienotriazolodiazepine compound and a medicinal use thereof
CL2007003244A1 (en) * 2006-11-16 2008-04-04 Millennium Pharm Inc COMPOUNDS DERIVED FROM PIRIMIDO [5,4-D] [2] BENZAZEPINA; PHARMACEUTICAL COMPOSITION THAT INCLUDES SUCH COMPOUND; AND USE OF THE COMPOUND FOR THE TREATMENT OF CANCER.
GB0919431D0 (en) 2009-11-05 2009-12-23 Glaxosmithkline Llc Novel compounds

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
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
CA2710740A1 (en) * 2007-12-28 2009-07-09 Shinji Miyoshi Thienotriazolodiazepine compound as antitumor agent
WO2009084693A1 (en) 2007-12-28 2009-07-09 Mitsubishi Tanabe Pharma Corporation Antitumor agent

Non-Patent Citations (10)

* 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
INT. J. PEPTIDE PROTEIN RES., vol. 40, 1992, pages 13 - 18
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 (146)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9051279B2 (en) 2009-12-22 2015-06-09 Novartis Ag Substituted isoquinolinones and quinazolinones
US10676484B2 (en) 2010-05-14 2020-06-09 Dana-Farber Cancer Institute, Inc. Compositions and methods for treating leukemia
US9789120B2 (en) 2010-05-14 2017-10-17 Dana-Farber Cancer Institute, Inc. Male contraceptive compositions and methods of use
US10407441B2 (en) 2010-05-14 2019-09-10 Dana-Farber Cancer Institute, Inc. Compositions and methods for treating neoplasia, inflammatory disease and other disorders
US9522920B2 (en) 2010-12-02 2016-12-20 Constellation Pharmaceuticals, Inc. Bromodomain inhibitors and uses thereof
US8796261B2 (en) 2010-12-02 2014-08-05 Constellation Pharmaceuticals, Inc. Bromodomain inhibitors and uses thereof
US9249161B2 (en) 2010-12-02 2016-02-02 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
WO2013027168A1 (en) 2011-08-22 2013-02-28 Pfizer Inc. Novel heterocyclic compounds as bromodomain inhibitors
US20140243321A1 (en) * 2011-08-29 2014-08-28 Coferon, Inc. Bioorthogonal monomers capable of dimerizing and targeting bromodomains, and methods of using same
WO2013033270A3 (en) * 2011-08-29 2016-04-28 Coferon, Inc. Bromodomain ligands capable of dimerizing in an aqueous solution, and methods of using same
US20140243286A1 (en) * 2011-08-29 2014-08-28 Coferon, Inc. Bromodomain ligands capable of dimerizing in an aqueous solution, and methods of using same
US8969341B2 (en) 2011-11-29 2015-03-03 Novartis Ag Pyrazolopyrrolidine compounds
RU2671571C1 (en) * 2011-12-30 2018-11-02 Эббви Инк. Bromdomain inhibitors
EP3360874A1 (en) * 2011-12-30 2018-08-15 AbbVie Inc. Bromodomain inhibitors
JP2017128594A (en) * 2011-12-30 2017-07-27 アッヴィ・インコーポレイテッド Bromodomain inhibitors
US9296741B2 (en) 2011-12-30 2016-03-29 Abbvie Inc. Bromodomain inhibitors
JP2015503529A (en) * 2011-12-30 2015-02-02 アッヴィ・インコーポレイテッド Bromodomain inhibitor
EP2797918A4 (en) * 2011-12-30 2015-07-01 Abbvie Inc Bromodomain inhibitors
TWI602815B (en) * 2011-12-30 2017-10-21 艾伯維有限公司 Bromodomain inhibitors
JP2018172428A (en) * 2011-12-30 2018-11-08 アッヴィ・インコーポレイテッド Bromodomain inhibitors
CN104136435B (en) * 2011-12-30 2017-05-10 艾伯维公司 Bromine structural domain inhibitor
CN106986872A (en) * 2011-12-30 2017-07-28 艾伯维公司 Bromine domain inhibitor
RU2647592C2 (en) * 2011-12-30 2018-03-16 Эббви Инк. Bromdomain inhibitors
CN106986872B (en) * 2011-12-30 2020-04-21 艾伯维公司 Bromodomain inhibitors
CN104136435A (en) * 2011-12-30 2014-11-05 艾伯维公司 Bromodomain inhibitors
US9365576B2 (en) 2012-05-24 2016-06-14 Novartis Ag Pyrrolopyrrolidinone compounds
TWI602820B (en) * 2012-06-06 2017-10-21 星宿藥物公司 Bromodomain inhibitors and uses thereof
CN104428304A (en) * 2012-06-06 2015-03-18 星座制药公司 Benzo [c] isoxazoloazepine bromodomain inhibitors and uses thereof
AU2013271569B2 (en) * 2012-06-06 2017-09-14 Constellation Pharmaceuticals, Inc. Benzo [C] isoxazoloazepine bromodomain inhibitors and uses thereof
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
WO2013184878A1 (en) * 2012-06-06 2013-12-12 Constellation Pharmaceuticals, Inc. Benzo [b] isoxazoloazepine bromodomain inhibitors and uses thereof
JP2015518899A (en) * 2012-06-06 2015-07-06 コンステレーション・ファーマシューティカルズ・インコーポレイテッドConstellation Pharmaceuticals,Inc. Benzo [C] isoxazoloazepine as bromodomain inhibitor and use thereof
US9624244B2 (en) 2012-06-06 2017-04-18 Constellation Pharmaceuticals, Inc. Benzo [B] isoxazoloazepine bromodomain inhibitors and uses thereof
CN104428304B (en) * 2012-06-06 2016-09-21 星座制药公司 Benzo [c] isoxazole azepine * bromodomain inhibitor and application thereof
EA026894B1 (en) * 2012-06-06 2017-05-31 Констеллейшен Фармасьютикалс, Инк. BENZO[c]ISOXAZOLOAZEPINE BROMODOMAIN INHIBITORS AND USE THEREOF
WO2013184876A1 (en) * 2012-06-06 2013-12-12 Constellation Pharmaceuticals, Inc. Benzo [c] isoxazoloazepine bromodomain inhibitors and uses thereof
JP2015531362A (en) * 2012-09-28 2015-11-02 バイエル ファーマ アクチエンゲゼルシャフト BET protein inhibitory 5-aryltriazoloazepines
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
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
US9403827B2 (en) 2013-01-22 2016-08-02 Novartis Ag Substituted purinone compounds
US9556180B2 (en) 2013-01-22 2017-01-31 Novartis Ag Pyrazolo[3,4-d]pyrimidinone compounds as inhibitors of the P53/MDM2 interaction
WO2014128070A1 (en) * 2013-02-22 2014-08-28 Bayer Pharma Aktiengesellschaft Pyrrolo- and pyrazolo-triazolodiazepines as bet-protein inhibitors for treating hyperproliferative diseases
CN105189514A (en) * 2013-02-22 2015-12-23 拜耳医药股份有限公司 Pyrrolo- and pyrazolo-triazolodiazepines as bet-protein inhibitors for treating hyperproliferative diseases
US9050346B2 (en) 2013-03-11 2015-06-09 Abbvie Inc. Bromodomain inhibitors
US8980879B2 (en) 2013-03-11 2015-03-17 Abbvie Inc. Bromodomain inhibitors
WO2014164771A1 (en) * 2013-03-11 2014-10-09 Abbvie Inc. Bromodomain inhibitors
US10035800B2 (en) 2013-03-12 2018-07-31 Abbvie Inc. Substituted 1,4,10-triazadibenzo[cd,f]azulenes, substituted 1,4,5,10-tetraazadibenzo[cd,f]azulenes and substituted 1,4,5,7,10-pentaazadibenzo[cd,f]azulenes as bromodomain inhibitors
JP2016510772A (en) * 2013-03-12 2016-04-11 アッヴィ・インコーポレイテッド Tetracyclic bromodomain inhibitors
TWI635087B (en) * 2013-03-12 2018-09-11 艾伯維有限公司 Tetracyclic bromodomain inhibitors
EP3539962A1 (en) * 2013-03-12 2019-09-18 AbbVie Inc. Tetracyclic bromodomain inhibitors
EP2970330A4 (en) * 2013-03-12 2016-10-19 Abbvie Inc Tetracyclic bromodomain inhibitors
US9714946B2 (en) 2013-03-14 2017-07-25 Dana-Farber Cancer Institute, Inc. Bromodomain binding reagents and uses thereof
US10464947B2 (en) 2013-03-15 2019-11-05 Incyte Holdings Corporation Tricyclic heterocycles as BET protein inhibitors
US9624241B2 (en) 2013-03-15 2017-04-18 Incyte Corporation Tricyclic heterocycles as BET protein inhibitors
US9938294B2 (en) 2013-03-15 2018-04-10 Incyte Corporation Tricyclic heterocycles as BET protein inhibitors
US11498926B2 (en) 2013-03-15 2022-11-15 Incyte Corporation Tricyclic heterocycles as BET protein inhibitors
US9227985B2 (en) 2013-03-15 2016-01-05 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
WO2014173241A1 (en) 2013-04-26 2014-10-30 Beigene, Ltd. Substituted5-(3,5-dimethylisoxazol-4-yl)indoline-2-ones
US9890166B2 (en) 2013-05-27 2018-02-13 Novartis Ag Imidazopyrrolidine derivatives and their use in the treatment of disease
US8975417B2 (en) 2013-05-27 2015-03-10 Novartis Ag Pyrazolopyrrolidine derivatives and their use in the treatment of disease
US9624247B2 (en) 2013-05-28 2017-04-18 Novartis Ag Pyrazolo-pyrrolidin-4-one derivatives as bet inhibitors and their use in the treatment of disease
US9714249B2 (en) 2013-05-28 2017-07-25 Novartis Ag Pyrazolo-pyrrolidin-4-one derivatives and their use in the treatment of disease
US9533997B2 (en) 2013-07-08 2017-01-03 Incyte Holdings Corporation Tricyclic heterocycles as BET protein inhibitors
US9290514B2 (en) 2013-07-08 2016-03-22 Incyte Holdings Corporation Tricyclic heterocycles as BET protein inhibitors
US9850257B2 (en) 2013-07-08 2017-12-26 Incyte Corporation Tricyclic heterocycles as BET protein inhibitors
EP3024327A4 (en) * 2013-07-25 2017-03-01 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
WO2015013635A2 (en) 2013-07-25 2015-01-29 Dana-Farber Cancer Institute, Inc. Inhibitors of transcription factors 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
US9550796B2 (en) 2013-11-21 2017-01-24 Novartis Ag Pyrrolopyrrolone derivatives and their use as BET inhibitors
US9737516B2 (en) 2013-11-26 2017-08-22 Incyte Corporation Bicyclic heterocycles as bet protein inhibitors
US9315501B2 (en) 2013-11-26 2016-04-19 Incyte Corporation Bicyclic heterocycles as BET protein 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
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
US11091484B2 (en) 2013-12-19 2021-08-17 Incyte Corporation Tricyclic heterocycles as BET protein inhibitors
US9309246B2 (en) 2013-12-19 2016-04-12 Incyte Corporation Tricyclic heterocycles as BET protein inhibitors
US10442803B2 (en) 2013-12-19 2019-10-15 Incyte Corporation Tricyclic heterocycles as bet protein inhibitors
US9777003B2 (en) 2013-12-19 2017-10-03 Incyte Corporation Tricyclic heterocycles as bet protein inhibitors
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
US10150756B2 (en) 2014-01-31 2018-12-11 Dana-Farber Cancer Institute, Inc. Diaminopyrimidine benzenesulfone derivatives and uses thereof
US10793571B2 (en) 2014-01-31 2020-10-06 Dana-Farber Cancer Institute, Inc. Uses of diazepane derivatives
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
US10925881B2 (en) 2014-02-28 2021-02-23 Tensha Therapeutics, Inc. Treatment of conditions associated with hyperinsulinaemia
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
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
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
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
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
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
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
US10618910B2 (en) 2014-09-15 2020-04-14 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
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
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
WO2016196065A1 (en) 2015-05-29 2016-12-08 Genentech, Inc. Methods and compositions for assessing responsiveness of cancers to bet inhibitors
US10702527B2 (en) 2015-06-12 2020-07-07 Dana-Farber Cancer Institute, Inc. Combination therapy of transcription inhibitors and kinase 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
US11406645B2 (en) 2015-09-11 2022-08-09 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
US10881668B2 (en) 2015-09-11 2021-01-05 Dana-Farber Cancer Institute, Inc. Acetamide thienotriazolodiazepines and uses thereof
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
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
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
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
WO2017180417A1 (en) 2016-04-12 2017-10-19 The Regents Of The University Of Michigan Bet protein degraders
US10633386B2 (en) 2016-04-12 2020-04-28 The Regents Of The University Of Michigan BET protein degraders
US10633379B2 (en) 2016-04-15 2020-04-28 Abbvie Inc. Bromodomain inhibitors
US11377446B2 (en) 2016-06-20 2022-07-05 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
US10189832B2 (en) 2016-06-20 2019-01-29 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
US10975093B2 (en) 2016-09-13 2021-04-13 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
EP3858837A1 (en) 2016-09-13 2021-08-04 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
WO2018052949A1 (en) 2016-09-13 2018-03-22 The Regents Of The University Of Michigan Fused 1,4-diazepines as bet protein degraders
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
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
CN107759607A (en) * 2017-11-29 2018-03-06 河南龙湖生物技术有限公司 Triazole with antitumor activity and phenodiazine Zhuo compound and preparation method thereof
WO2020020288A1 (en) 2018-07-25 2020-01-30 正大天晴药业集团股份有限公司 Sulfoximine compound as bromodomain protein inhibitor and pharmaceutical composition and medical use thereof
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
WO2021262731A2 (en) 2020-06-23 2021-12-30 Genentech, Inc. Macrocyclic compounds and methods of use thereof
US11759527B2 (en) 2021-01-20 2023-09-19 Abbvie Inc. Anti-EGFR antibody-drug conjugates
WO2023205251A1 (en) 2022-04-19 2023-10-26 Nuevolution A/S Compounds active towards bromodomains

Also Published As

Publication number Publication date
ES2526671T3 (en) 2015-01-14
EP2585465A1 (en) 2013-05-01
US9085582B2 (en) 2015-07-21
JP5844358B2 (en) 2016-01-13
JP2013529611A (en) 2013-07-22
EP2585465B1 (en) 2014-11-12
US20130079335A1 (en) 2013-03-28

Similar Documents

Publication Publication Date Title
EP2585465B1 (en) Benzotriazolodiazepine compounds inhibitors of bromodomains
EP2496580B1 (en) Benzodiazepine bromodomain inhibitor
EP3050885B1 (en) Benzodiazepine bromodomain inhibitor
AU2012244759B2 (en) Tetrahydroquinoline derivatives useful as bromodomain inhibitors
EP2705032B1 (en) Dihydroquinoline derivatives as bromodomain inhibitors
WO2011054848A1 (en) Thetrahydroquinolines derivatives as bromodomain inhibitors
EP2699573A2 (en) 7-(3,5-dimethyl-4-isoxazolyl)-8-(methyloxy)-1h-imidazo[4,5-c]quinoline derivatives

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: 11725769

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2011725769

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 13702177

Country of ref document: US

ENP Entry into the national phase

Ref document number: 2013515827

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE