WO2012052390A1 - N-2-(2-pyridinyl)-4-pyrimidinyl-beta-alanine derivatives as inhibitors of histone demethylase jmjd3 - Google Patents

N-2-(2-pyridinyl)-4-pyrimidinyl-beta-alanine derivatives as inhibitors of histone demethylase jmjd3 Download PDF

Info

Publication number
WO2012052390A1
WO2012052390A1 PCT/EP2011/068085 EP2011068085W WO2012052390A1 WO 2012052390 A1 WO2012052390 A1 WO 2012052390A1 EP 2011068085 W EP2011068085 W EP 2011068085W WO 2012052390 A1 WO2012052390 A1 WO 2012052390A1
Authority
WO
WIPO (PCT)
Prior art keywords
alanine
pyrimidinyl
pyridinyl
alkyl
phenyl
Prior art date
Application number
PCT/EP2011/068085
Other languages
French (fr)
Inventor
Stephen John Atkinson
Michael David Barker
Matthew Campbell
Philip Humphreys
John Liddle
Robert John Sheppard
David Wilson
Original Assignee
Glaxo Group Limited
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Glaxo Group Limited filed Critical Glaxo Group Limited
Publication of WO2012052390A1 publication Critical patent/WO2012052390A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • 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
    • A61P37/02Immunomodulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/14Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings

Definitions

  • the present invention relates to compounds, compositions, combinations and medicaments containing said compounds and processes for their preparation.
  • the invention also relates to the use of said compounds, combinations, compositions and medicaments, for example as inhibitors of the activity of the histone demethylase JMJD3, to modify the epigenetic status of cells and/or the treatment of diseases and conditions mediated by JMJD3, in particular cancer, inflammation and autoimmune diseases.
  • Chromatin is the complex combination of DNA and protein that makes up chromosomes. It is found inside the nuclei of eukaryotic cells and is divided between heterochromatin (condensed) and euchromatin (extended) forms.
  • the major components of chromatin are DNA and proteins, including histones.
  • the basic building blocks of chromatin are nucleosomes, each of which is composed of 146 base pairs of DNA wrapped around a histone octamer consisting of 2 copies of each H2A, H2B, H3 and H4.
  • chromatin The functions of chromatin are to package DNA into a smaller volume to fit in the cell, to strengthen the DNA to allow mitosis and meiosis, and to serve as a mechanism to control expression and DNA replication.
  • Chromatin contains genetic material serving as instructions to direct cell functions. Changes in chromatin structure are regulated by modifications on histone and DNA methylation. Epigenetic mechanisms do not change the DNA sequence but allow the genes to be expressed differently. Epigenetics modification include numerous mechanisms including DNA methylation and post-translational modification of N-terminal tails of histone proteins such as methylation, acetylation, phosphorylation and ubiquitination.
  • Histone methylation is an abundant epigenetic modification of core histones found in eukaryotic organisms that has been linked to a number of cellular processes including DNA repair, cell cycle progression, cell differentiation and regulation of gene expression. This modification is catalysed by the specific histone methyltransferases (HMTs), lysine methyl transferase and arginine methyl transferase, which introduce methyl groups at lysine (K) or arginine (R) residues respectively.
  • HMTs histone methyltransferases
  • K lysine
  • R arginine
  • histone methylation has been found to occur at six major sites, including histone H3 lysine 4 (H3K4), H3K9, H3K27, H3K36, H3K79 and H4K20.
  • H3K4 histone H3 lysine 4
  • H3K9 histone H3K27
  • H3K36 histone H3K9
  • H3K27 histone K3 lysine 4
  • H4K20 histone H3 lysine 4
  • H3K9 promoter methylation is considered a repressive mark for euchromatic genes (Nielsen et al., Nature 2001 ,412, 561 -565; Shi et al., Nature 2003, 422, 735-738) and is also one of the landmark modifications associated with heterochromatin (Nakayama et al., Science 2001 , 292, 1 10-1 13) but some studies have also identified association of H3K9 trimethylation (H3K9me3) with actively transcribed genes (Vakoc C. et al, Mol. Cell 2005, 19. 381 -391).
  • Histone methylation appears thus to be regulated by a complex network that involves a large number of site-specific methylases, demethylases and methyl recognition proteins, which play an important role in controlling the expression of genetic information through transcriptional changes and chromatin structure alterations. Since levels of lysine methylation are known to change during processes such as transcriptional regulation, it was proposed that specific enzymatic activity might remove the methyl groups (Bannister et al., 2002 Cell 109, 801 -806).
  • the Jumonji protein is the founding member of a group of proteins characterised by a novel structural motif, the JmjC domain.
  • This is an extensive group of demethylase enzymes which can be defined into several families according to sequence similarity within the JmjC domain and the presence of other domains in the full length protein.
  • the JmjC domain of several members of this family has been shown to possess lysine demethylation activity, which is dependent on iron (Fe (II)) and a- ketoglutarate as co-factors (Klose RJ et al, Nat Rev Genet. 2006 Sep; 7(9); 715-27).
  • the JmjC-domain-containing histone demethylases JHDMs
  • JHDMs can remove all three histone lysine-methylation states.
  • JmjC domain-containing proteins including the histone demethylase JMJD3, have been implicated in tumorogenesis and thus have identified histone demethylases as targets of research for anti-cancer therapies (see for example WO2009/1 1401 1 and WO2010/043566).
  • JmjD3 (KDM6B) is one of the approximately 30 JmjC family members found in humans, and functions as a specific demethylase of lysine 27 of histone H3 (H3K27). JmjD3 can demethylate both the tri- and dimethylated H3K27-repressive histone marks, thereby facilitating gene transcription. This was first demonstrated in C.
  • JmjD3 was shown to regulate gonadal development through modulation of HOX gene expression (Agger K et al, Nature 2007 Oct; 449(7163); 731 -734). Further studies have placed JmjD3 at key cell fate decision checkpoints in T lymphocytes (Miller SA et al, Genes Dev. 2008 Oct; 22; 2280-2993) and macrophages (Ishii M et al, Blood 2009 Oct; 1 14(15); 3244-3254). In addition, JmjD3 has been demonstrated to regulate the differentiation state of the epidermis (Sen GL et al, Genes Dev.
  • JmjD3 also appears to be involved in more acute, externally-driven, inflammatory processes. In macrophages, for example, JmjD3 is rapidly induced through an NF-kB- dependent mechanism in response to bacterial products and inflammatory stimuli (De Santa F et al, Cell 2007 Sept; 130; 1083-1094).
  • JmjD3 participates directly in the inflammatory transcriptional response, although it remains unclear whether this is achieved through demethylation of H3K27me3 at target gene promoters (De Santa F et al, EMBO J. 2009 Sept; 28; 3341 -3352).
  • composition comprising a compound of the formula
  • R' is:
  • each R a and R b is independently selected from:
  • C ⁇ alkyl which is optionally substituted by one or more substituents independently selected from phenyl (which phenyl is optionally substituted by one or more substituents independently selected from C 1-3 alkyl, 0-C 1-3 alkyl, C(0)NR c 2 , halo and cyano), C(0)NR c 2 , a 4, 5, 6 or 7-membered heterocyclic or heteroaryl group (containing one or more heteroatoms independently selected from N, O and, S), a 3, 4, 5, 6 or 7-membered cycloalkyl group (which is optionally fused to phenyl), halo, OC 1-3 alkyl, OH, -NHCOC ⁇ alkylNR ⁇ and C(0)NHCH 2 C(0)NR c 2 ;
  • R a and R b together form a 5, 6 or 7-membered heterocyclic group optionally containing one or more further heteroatoms independently selected from N, O, S or S(0) 2 said heterocyclic group being optionally fused to a 5, 6 or 7-membered aryl or heteroaryl ring containing one or more heteroatoms independently selected from N, O and S; the heterocylic ring and/or the aryl or heteroaryl to which it is optionally fused being optionally substituted by one or more substituents independently selected from halo, OH, C 1-3 alkyl, 0-C 1-3 alkyl, C(0)C 1-3 alkyl, S(0) 2 C 1-3 alkyl, NHC(0)C 1-3 alkyl, NHS(0) 2 C 1-3 alkyl, C(0)NR c 2 , C(0)NR d 2 (wherein R d and R d together form a 5 or 6-membered heterocylic ring), NR C 2 C(0)phenyl, S(0) 2 NR c
  • R 2 and R 3 are each independently selected from:
  • each R c is independently selected from hydrogen and C 1-3 alkyl or a pharmaceutically acceptable salt thereof and one or more of pharmaceutically acceptable carriers, diluents and excipients.
  • a compound of the formula (I) or a pharmaceutically acceptable salt thereof as defined above with the proviso R is not -C(CH 3 ) 3 .
  • composition comprising a compound of formula (la):
  • R is C 1-6 alkyl, C 3 . 7 cycloalkyl, C 1-6 haloalkyl, NR a R b ; each R a and R b is independently selected from H, C ⁇ alkyl (which alkyl is optionally substituted by one or more substituents independently selected from phenyl, halo, OH) or R a and R b together form a 4, 5 or 6 membered heterocyclic group optionally containing one or more further heteroatoms selected from N or O, said heterocyclic group being optionally fused to a phenyl ring. or a pharmaceutically acceptable salt thereof and one or more of pharmaceutically acceptable carriers, diluents and excipients
  • R is C 1-6 alkyl, C 3 . 7 cycloalkyl, C ⁇ haloalkyl, NR a R b each R a and R b is independently selected from H, C ⁇ alkyl (which alkyl is optionally substituted by one or more substituents independently selected from phenyl, halo, OH) or R a and R b together form a ,a 4,5 or 6 membered heterocyclic group optionally containing one or more further heteroatoms selected from N or O, said heterocyclic group being optionally fused to a phenyl ring; with the proviso R is not -C(CH 3 ) 3 . or a pharmaceutically acceptable salt thereof.
  • a method of changing the epigenetic status of cells, treating cancer, inflammation or autoimmune diseases in a subject comprising administering a therapeutically effective amount of a compound of formula (I) or of formula (la) or of formula (lb) or a pharmaceutically acceptable salt thereof.
  • a combination comprising a compound of formula (I) or of formula (la) or of formula (lb), or a pharmaceutically acceptable salt thereof and at least one further therapeutic agent.
  • a combination comprising a compound of formula (I) or of formula (la) or of formula (lb), or a pharmaceutically acceptable salt thereof and at least one further therapeutic agent for use in therapy, particularly for changing the epigenetic status of cells and treating cancer, inflammation or autoimmune diseases.
  • a compound of formula (I) or of formula (la) or of formula (lb), or a pharmaceutically acceptable salt thereof for use in the treatment of cancer in a further aspect there is provided a method of treating cancer comprising administering to a human in need thereof a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof.
  • a method of treating cancer comprising administering to a human in need thereof a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof.
  • a combination comprising a compound of formula (I) or of formula (la) or of formula (lb), or a pharmaceutically acceptable salt thereof and at least one anti-neoplastic agent.
  • a combination comprising a compound of formula (I) or of formula (la) or of formula (lb), or a pharmaceutically acceptable salt thereof and at least one anti-neoplastic agent, for use in therapy.
  • a combination comprising a compound of formula (I) or of formula (la) or of formula (lb), or pharmaceutically acceptable salt thereof and at least one anti-neoplastic agent, for use in treating cancer.
  • a combination comprising a compound of formula (I) or of formula (la) or of formula (lb), or a pharmaceutically acceptable salt thereof and at least one antineoplastic agent, in the manufacture of a medicament for the treatment of cancer.
  • a method of treating cancer comprising administering to a human in need thereof a therapeutically effective amount of a combination comprising a compound of formula (I) or of formula (la) or of formula (lb), or a pharmaceutically acceptable salt thereof and at least one anti-neoplastic agent.
  • a pharmaceutical composition comprising a combination comprising a compound of formula (I) or of formula (la) or of formula (lb), or a pharmaceutically acceptable salt thereof and at least one further therapeutic agent, particularly at least one antineoplastic agent and one or more of pharmaceutically acceptable carriers, diluents and excipients.
  • a method of treating inflammation or autoimmune diseases comprising administering to human in need thereof, a therapeutically effect amount of a compound of formula (I) or of formula (la) or of formula (lb), or a pharmaceutically acceptable salt thereof.
  • a compound of formula (I) or of formula (la) or of formula (lb), or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for the treatment of inflammation or autoimmune diseases.
  • JmjC domain-containing proteins as histone demethylases that mediate the reversal of methylation at histone H3K4, H3K9 and H3K36.
  • the JmjD3 family of proteins is a subfamily of JmjC domain-containing proteins that have been shown to demethyate H3K27.
  • Such inhibitors are therefore useful in changing the epigenetic status of cells resulting in inhibiting or activating chromatin remodelling by modifying histone methylation and thus in treating disorders associated with such modified histone methylation including cancer and other conditions associated with undesirable cell proliferation, autoimmune and inflammatory diseases or conditions and psychiatric disorders including depression.
  • a compound of formula (I) or “a compound or formula (la)” or “a compound or formula (lb)” includes all solvates, complexes, polymorphs, radiolabeled derivatives (including deuterated derivatives where one or more H are replaced by D), stereoisomers and optical isomers of the compounds of formula (I) or (la) or (lb) and salts thereof.
  • references herein to "compounds of the formula (I)” typically also apply to compounds of the formula (la) and (lb) and are to be understood as such unless it is indicated otherwise.
  • 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.
  • the compounds of formula (I) may exist in solid or liquid form.
  • compound of the invention may exist in a continuum of solid states ranging from fully amorphous to fully crystalline.
  • the term 'amorphous' refers to a state in which the material lacks long range order at the molecular level and, depending upon the temperature, may exhibit the physical properties of a solid or a liquid. Typically such materials do not give distinctive X-ray diffraction patterns and, while exhibiting the properties of a solid, are more formally described as a liquid.
  • a change from solid to liquid properties occurs which is characterized by a change of state, typically second order ('glass transition').
  • 'crystalline' refers to a solid phase in which the material has a regular ordered internal structure at the molecular level and gives a distinctive X-ray diffraction pattern with defined peaks. Such materials when heated sufficiently will also exhibit the properties of a liquid, but the change from solid to liquid is characterized by a phase change, typically first order ('melting point').
  • the compound of formula (I) may exist in solvated and unsolvated forms.
  • solvate refers to a complex of variable stoichiometry formed by a solute (in this invention, a compound of formula (I) or a salt) and a solvent. Such solvents for the purpose of the invention may not interfere with the biological activity of the solute.
  • pharmaceutically acceptable solvates may be formed for crystalline compounds wherein solvent molecules are incorporated into the crystalline lattice during crystallization.
  • the incorporated solvent molecules may be water molecules or non-aqueous such as ethanol, isopropanol, DMSO, acetic acid, ethanolamine, and ethyl acetate molecules.
  • Crystalline lattice incorporated with water molecules are typically referred to as "hydrates". Hydrates include stoichiometric hydrates as well as compositions containing variable amounts of water. The present invention includes all such solvates.
  • the compounds of formula (I) may have the ability to crystallize in more than one form, a characteristic, which is known as polymorphism, and it is understood that such polymorphic forms (“polymorphs”) are within the scope of the invention. Polymorphism generally can occur as a response to changes in temperature or pressure or both and can also result from variations in the crystallization process. Polymorphs can be distinguished by various physical characteristics known in the art such as x-ray diffraction patterns, solubility and melting point.
  • the compounds of formula (I) may form tautomers. It is understood that all tautomers and mixtures of tautomers of the compounds of the present invention are included within the scope of the compounds of the present invention.
  • histone demethylase inhibitor refers to any compound or treatment capable of inhibiting or reducing the expression or activity of a histone demethylase.
  • the inhibitor is preferably selective against one or more histone demethylase enzymes with no direct activity as any other histone modifying enzymes.
  • alkyl and alkylene refers to a saturated hydrocarbon chain having the specified number of member atoms.
  • C ⁇ alkyl refers to an alkyl group having from 1 to 6 member atoms, for example 1 to 4 member atoms.
  • Alkyl groups may be straight or branched. Representative branched alkyl groups have one, two, or three branches.
  • Alkyl includes methyl, ethyl, propyl (n-propyl and isopropyl), butyl (n-butyl, isobutyl, and t-butyl), pentyl (n-pentyl, isopentyl, and neopentyl), and hexyl.
  • halo refers to the halogen radical fluoro, chloro, bromo, or iodo.
  • C ⁇ Ce haloalkyl refers to a straight or branched chain alkyl group as defined above containing at least 1 , and at most 6 carbon atoms respectively substituted with at least one halo group, halo being as defined herein.
  • branched or straight chained haloalkyl groups useful in the present invention include, but are not limited to, methyl, ethyl, propyl, isopropyl, isobutyl and n-butyl substituted independently with one or more halos, e.g., fluoro, chloro, bromo and iodo.
  • heterocyclic group refers to a non-aromatic ring having the specified number of member atoms being saturated or having one or more degrees of unsaturation and, unless otherwise specified, containing one or more heteroatoms selected from N, O or S.
  • aryl refers to an aromatic ring having the specified number of member atoms. Bicyclic and other polycyclic ring systems containing an aryl are described as fused systems.
  • heteroaryl refers to an aromatic ring having the specified number of member atoms and, unless otherwise specified, containing one or more heteroatoms selected from N, O or S.
  • Bicyclic and other polycyclic ring systems comprising a heteroaryl ring are described as fused systems.
  • member atoms refers to the atom or atoms that form a chain or ring. Where more than one member atom is present in a chain and within a ring, each member atom is covalently bound to an adjacent member atom in the chain or ring. Atoms that make up a substituent group on a chain or ring are not member atoms in the chain or ring.
  • substituted in reference to a group indicates that a hydrogen atom attached to a member atom within a group is replaced. It should be understood that the term “substituted” includes the implicit provision that such substitution be in accordance with the permitted valence of the substituted atom and the substituent and that the substitution results in a stable compound (i.e. one that does not spontaneously undergo transformation such as by rearrangement, cyclization, or elimination). In certain embodiments, a single atom may be substituted with more than one substituent as long as such substitution is in accordance with the permitted valence of the atom. Suitable substituents are defined herein for each substituted or optionally substituted group.
  • the term "pharmaceutically acceptable” refers to those compounds, materials, compositions, and dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • R is:
  • a 5-membered heteroaryl which contains one or two heteroatoms selected from N, O and S and which is optionally fused to phenyl), said 5-membered heteroaryl being optionally substituted with one or more substituents independently selected from C 1-3 alkyl;
  • O-C ⁇ alkyl which is optionally substituted by phenyl (optionally substituted by one of more chloro) or napthyl);
  • R a is H or C 1-4 alkyl, especially H or methyl, and R b is C ⁇ alkyl which is optionally substituted by one or more substituents independently selected from:
  • phenyl (which is optionally substituted by one or more substiuents independently selected from methyl, O-methyl, C(0)NR c 2 , chloro, bromo and cyano),
  • R a is H and R b is a 3, 4 or 5-membered cycloalkyl group (which is optionally fused to phenyl), or
  • R a and R b together form a 5, 6 or 7-membered heterocyclic group optionally containing a further heteroatom selected from N, O, S or S(0) 2 said heterocyclic group being optionally fused to a phenyl ring or to a 5 or 6-membered heteroaryl ring (containing one or more heteroatoms independently selected from N, O and S); the heterocylic ring and/or the phenyl or heteroaryl to which it is optionally fused being optionally substituted by one or more substituents independently selected from:
  • each R c is independently selected from hydrogen and methyl.
  • R is:
  • a 5-membered heteroaryl which contains one or two heteroatoms selected from N, O and S and which is optionally fused to phenyl), said 5-membered heteroaryl being optionally substituted with one or two methyl substituents;
  • R a is H or methyl
  • R b is C ⁇ alkyl which is optionally substituted by one or more substituents independently selected from:
  • phenyl (which is optionally substituted by one or more substiuents independently selected from methyl, O-methyl, C(0)NR c 2 , chloro and cyano),
  • a 3 or 5-membered cycloalkyl group especially cyclopropyl
  • R a is H and R b is a 5-membered cycloalkyl group (which is optionally fused to phenyl), or
  • R a and R b together form a 5, 6 or 7-membered heterocyclic group optionally containing a further heteroatom selected from N, O, S or S(0) 2 said heterocyclic group being optionally fused to a phenyl ring or to a 5 or 6-membered heteroaryl ring (containing one or more heteroatoms independently selected from N, O and S); the heterocylic ring and/or the phenyl or heteroaryl to which it is optionally fused being optionally substituted by one or more substituents independently selected from:
  • o 0-C 1-3 alkyl especially 0-CH 3 ,
  • NHC(0)C 1-3 alkyl especially NHC(0)CH 3 ,
  • o 5 or 6-membered heteroaryl (containing one or more heteroatoms independently selected from N, O and S);
  • NR C 2 is selected from NHCH 3 and NH 2 .
  • R is:
  • R a and R together form a 5, 6 or 7-membered heterocyclic group optionally containing a further heteroatom selected from N, O, S or S(0) 2 said heterocyclic group being optionally fused to a phenyl ring or to a 5 or 6-membered heteroaryl ring (containing one or more heteroatoms independently selected from N, O and S); the heterocylic ring and/or the phenyl or heteroaryl to which it is optionally fused being optionally substituted by one or more substituents independently selected from:
  • o 5 or 6-membered heteroaryl (containing one or more heteroatoms independently selected from N, O and S),
  • each R c 2 is independently selected from H and CH 3 .
  • R is:
  • R a and R b together form a 5, 6 or 7-membered heterocyclic group said heterocyclic group being optionally fused to a phenyl ring; the heterocylic ring and/or the phenyl to which it is optionally fused being optionally substituted by one or more substituents independently selected from: chloro, bromo, OH, C 1-3 alkyl, 0-C 1-3 alkyl, C(0)C 1-3 alkyl, and S(0) 2 C 1-3 alkyl.
  • R is:
  • R a and R b together form a 5, 6 or 7-membered heterocyclic group said heterocyclic group being fused to a phenyl ring; the heterocylic ring and/or the phenyl to which it is fused being unsubstiuted or substituted by one or more substituents independently selected from: chloro, OH, methyl, O-methyl and S(0) 2 methyl, for example, unsubstituted.
  • R is selected from:
  • R is selected from:
  • R is selected from:
  • R 2 and R 3 are selected from H, (CH ⁇ ! ⁇ NR ⁇ CH ⁇ ! ⁇ NR ⁇ and (CH 2 ) 0 . 6 NR C 2 .
  • one of R 2 and R 3 is H and the other is selected from (CH ⁇ NR' CI-y ⁇ NR 0 ;, and (CH 2 ) 0 . 6 NR C 2 .
  • R 2 and R 3 are both H.
  • R 2 and R 3 may be as defined in this paragraph in all of the embodiments of the invention described above where different options for R are disclosed.
  • each R c is independently selected from hydrogen and methyl.
  • NR C 2 is selected from NHCH 3 and NH 2 . While aspects for each variable have generally been listed above separately for each variable this invention includes those compounds in which several or each aspect in formula (I) is selected from each of the aspects listed above. Therefore, this invention is intended to include all combinations of aspects for each variable. Specific examples of compounds of formula (I) and salts thereof include the compounds and salts of Examples 1 -125 disclosed herein.
  • the compounds of formula (I) may be in the form of a pro-drug, for example an ester which upon administration to the patient is capable of providing a compound of the present invention.
  • the ester is a C ⁇ alkyl ester.
  • Esters have a higher cell penetrance than the corresponding acids so may offer a pro-drug approach to achieving cell activity. Esters have also been found to be weakly active inhibitors of JmjD3 in themselves.
  • the compounds of Formula (I) may be in the form of a salt.
  • the salts of the present invention are pharmaceutically acceptable salts.
  • Salts encompassed within the term “pharmaceutically acceptable salts” refer to non-toxic salts of the compounds of this invention.
  • suitable salts see Berge et al, J. Pharm. Sci. 1977, 66, 1 -19.
  • Suitable pharmaceutically acceptable salts can include acid addition salts.
  • 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, sulfuric, nitric, phosphoric, p-toluenesulfonic, benzenesulfonic, methanesulfonic, ethanesulfonic, naphthalenesulfonic such as 2-naphthalenesulfonic), 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, sulfuric, nitric, phosphoric, p-toluenesulfonic, benzenesulfonic, methanesulfonic, ethanesulfonic, naphthalenesulfonic such as 2-naphthalenesulfonic
  • a suitable solvent such as an organic solvent
  • a pharmaceutically acceptable acid addition salt of a compound of formula (I) can comprise or be for example a hydrobromide, hydrochloride, sulfate, nitrate, phosphate, p-toluenesulfonate, benzenesulfonate, methanesulfonate, ethanesulfonate, naphthalenesulfonate (e.g. 2-naphthalenesulfonate) salt.
  • Other non-pharmaceutically acceptable salts e.g. trifluoroacetates, may be used, for example in the isolation of 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 compounds of formula (I).
  • the compound of formula (I) or a pharmaceutically acceptable salt thereof may be administered as the raw chemical, it is possible to present the active ingredient as a pharmaceutical composition.
  • the invention further provides pharmaceutical compositions comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof and one or more pharmaceutically acceptable carriers, diluents, or excipients.
  • the carrier(s), diluents(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 a compound of formula (I), or pharmaceutically acceptable salts thereof, with one or more pharmaceutically acceptable carriers, diluents or excipients.
  • the pharmaceutical composition can be for use in the treatment and/or prophylaxis of any of the conditions described herein.
  • 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 once or more than once a day.
  • Such pharmaceutical compositions may be prepared by any of the methods well known in the pharmacy art.
  • 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).
  • 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.
  • Powders are prepared by reducing the compound to a suitable fine size and mixing with a similarly prepared pharmaceutical carrier such as an edible carbohydrate, as, for example, starch or mannitol.
  • Flavouring, preservative, dispersing and colouring agent can also be present.
  • Capsules are made by preparing a powder mixture, as described above, and filling formed gelatin sheaths.
  • Glidants and lubricants such as colloidal silica, talc, magnesium stearate, calcium stearate or solid polyethylene glycol can be added to the powder mixture before the filling operation.
  • a disintegrating or solubilizing agent such as agar-agar, calcium carbonate or sodium carbonate can also be added to improve the availability of the medicament when the capsule is ingested.
  • suitable binders, glidants, lubricants, sweetening agents, flavours, disintegrating agents and colouring agents can also be incorporated into the mixture.
  • Suitable binders include starch, gelatin, natural sugars such as glucose or beta-lactose, corn sweeteners, natural and synthetic gums such as acacia, tragacanth or sodium alginate, carboxymethylcellulose, polyethylene glycol, waxes and the like.
  • Lubricants used in these dosage forms include sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride and the like.
  • Disintegrators include, without limitation, starch, methyl cellulose, agar, bentonite, xanthan gum and the like. Tablets are formulated, for example, by preparing a powder mixture, granulating or slugging, adding a lubricant and disintegrant and pressing into tablets.
  • a powder mixture is prepared by mixing the compound, suitably comminuted, with a diluent or base as described above, and optionally, with a binder such as carboxymethylcellulose, an aliginate, gelatin, or polyvinyl pyrrolidone, a solution retardant such as paraffin, a resorption accelerator such as a quaternary salt and/or an absorption agent such as bentonite, kaolin or dicalcium phosphate.
  • the powder mixture can be granulated by wetting with a binder such as syrup, starch paste, acadia mucilage or solutions of cellulosic or polymeric materials and forcing through a screen.
  • the powder mixture can be run through the tablet machine and the result is imperfectly formed slugs broken into granules.
  • the granules can be lubricated to prevent sticking to the tablet forming dies by means of the addition of stearic acid, a stearate salt, talc or mineral oil.
  • the lubricated mixture is then compressed into tablets.
  • the compounds of 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 flavoured 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 composition can also be prepared to prolong or sustain the release as for example by coating or embedding particulate material in polymers, wax or the like.
  • the compounds of formula (I) or a pharmaceutically acceptable salt thereof may 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 transdermal administration may be presented as discrete patches intended to remain in intimate contact with the epidermis of the recipient for a prolonged period of time.
  • compositions adapted for topical administration may be formulated as ointments, creams, suspensions, lotions, powders, solutions, pastes, gels, sprays, aerosols or oils.
  • the compositions are preferably applied as a topical ointment or cream.
  • the active ingredient may be employed with either a paraffinic or a water-miscible ointment base.
  • the active ingredient may be formulated in a cream with an oil-in-water cream base or a water-in-oil base.
  • compositions adapted for topical administrations to the eye include eye drops wherein the active ingredient is dissolved or suspended in a suitable carrier, especially an aqueous solvent.
  • compositions adapted for topical administration in the mouth include lozenges, pastilles and mouth washes.
  • compositions adapted for rectal administration may be presented as suppositories or as enemas.
  • Dosage forms for nasal or inhaled administration may conveniently be formulated as aerosols, solutions, suspensions drops, gels or dry powders.
  • compositions suitable and/or adapted for inhaled administration it is preferred that the agent is in a particle-size-reduced form, and more preferably the size-reduced form is obtained or obtainable by micronisation.
  • the preferable particle size of the size-reduced (e.g. micronised) compound or salt or solvate is defined by a D50 value of about 0.5 to about 10 microns (for example as measured using laser diffraction).
  • compositions adapted for administration by inhalation include the particle dusts or mists.
  • compositions wherein the carrier is a liquid for administration as a nasal spray or drops include aqueous or oil solutions/suspensions of the active ingredient which may be generated by means of various types of metered dose pressurised aerosols, nebulizers or insufflators.
  • Aerosol formulations can comprise a solution or fine suspension of the agent in a pharmaceutically acceptable aqueous or non-aqueous solvent. Aerosol formulations can be presented in single or multidose quantities in sterile form in a sealed container, which can take the form of a cartridge or refill for use with an atomising device or inhaler. Alternatively the sealed container may be a unitary dispensing device such as a single dose nasal inhaler or an aerosol dispenser fitted with a metering valve (metered dose inhaler) which is intended for disposal once the contents of the container have been exhausted.
  • 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.
  • Other excipient modifiers may also be incorporated to improve, for example, the stability and/or taste and/or fine particle mass characteristics (amount and/or profile) of the formulation.
  • 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 agent, (preferably in particle-size-reduced form, e.g. in micronised form), and optionally a performance modifier such as L-leucine or another amino acid, cellobiose octaacetate and/or metals salts of stearic acid such as magnesium or calcium stearate.
  • Aerosol formulations are preferably arranged so that each metered dose or "puff of aerosol contains a particular amount of a compound of the invention.
  • Administration may be once daily or several times daily, for example 2, 3 4 or 8 times, giving for example 1 , 2 or 3 doses each time.
  • the overall daily dose and the metered dose delivered by capsules and cartridges in an inhaler or insufflator will generally be double those with aerosol formulations.
  • compositions adapted for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams or spray formulations.
  • Pharmaceutical compositions adapted for parental 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 nonaqueous sterile suspensions which may include suspending agents and thickening agents.
  • 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.
  • 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 may include other agents conventional in the art having regard to the type of formulation in question, for example those suitable for oral administration may include flavouring agents.
  • Antisense or RNA interference molecules may be administered to the mammal in need thereof.
  • constructs including the same may be administered.
  • Such molecules and constructs can be used to interfere with the expression of the protein of interest, e.g., histone demethylase and as such, modify histone demethylation.
  • delivery is by means known in the art.
  • Antisense or RNA interference molecules can be delivered in vitro to cells or in vivo, e.g., to tumors of a mammal. Nodes of delivery can be used without limitations, including: intravenous, intramuscular, intraperitoneal, intra-arterial, local delivery during surgery, endoscopic, subcutaneous, and per os.
  • Vectors can be selected for desirable properties for any particular application. Vectors can be viral or plasmid. Adenoviral vectors are useful in this regard. Tissue-specific, cell-type specific, or otherwise regulatable promoters can be used to control the transcription of the inhibitory polynucleotide molecules. Non-viral carriers such as liposomes or nanospheres can also be used.
  • a therapeutically effective amount of the 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 subject, 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.
  • the subject to be treated is a mammal, particularly a human.
  • the compound of formula (I) or a pharmaceutically acceptable salt thereof may be administered in a daily dose.
  • 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.
  • the amount of the compound of compound of formula (I) or a pharmaceutically acceptable salt thereof administered according to the present invention will be an amount selected from 0.01 mg to 1000 mg per day (calculated as the free or unsalted compound).
  • the compounds of the formula (I) or a pharmaceutically acceptable salt thereof may be used in combination with or include one or more other therapeutic agents and may be administered either sequentially or simultaneously by any convenient route in separate or combined pharmaceutical compositions.
  • the compounds of formula (I) or a pharmaceutically acceptable salt thereof and further therapeutic agent(s) may be employed in combination by administration simultaneously in a unitary pharmaceutical composition including both compounds.
  • the combination may be administered separately in separate pharmaceutical compositions, each including one of the compounds in a sequential manner wherein, for example, the compound of formula or a pharmaceutically acceptable salt thereof is administered first and the other second and visa versa.
  • Such sequential administration may be close in time (e.g. simultaneously) or remote in time.
  • the compounds are administered in the same dosage form, e.g. one compound may be administered topically and the other compound may be administered orally.
  • both compounds are administered orally.
  • kits or kit of parts
  • the combination kit can contain the agents in a single pharmaceutical composition, such as a tablet, or in separate pharmaceutical compositions.
  • the combination kit will contain each agent in separate pharmaceutical compositions either in a single package or in separate pharmaceutical compositions in separate packages.
  • the combination kit can also be provided by instruction, such as dosage and administration instructions.
  • dosage and administration instructions can be of the kind that are provided to a doctor, for example by a drug product label, or they can be of the kind that are provided by a doctor, such as instructions to a patient.
  • each agent of the combination may be repeated one or more times.
  • such sequential administration may be close in time or remote in time.
  • administration of the other agent several minutes to several dozen minutes after the administration of the first agent, and administration of the other agent several hours to several days after the administration of the first agent are included, wherein the lapse of time is not limited, For example, one agent may be administered once a day, and the other agent may be administered 2 or 3 times a day, or one agent may be administered once a week, and the other agent may be administered once a day and the like.
  • the other therapeutic ingredients(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 two compounds When combined in the same composition it will be appreciated that the two compounds must be stable and compatible with each other and the other components of the composition and may be formulated for administration. When formulated separately they may be provided in any convenient composition, conveniently, in such a manner as known for such compounds in the art.
  • the mammal is a human.
  • methods of changing the epigenetic status of cells and the treatment or prevention of cancer, inflammation and autoimmune diseases and conditions which may be improved by modulating the methylation status of histones, particularly H3K27 and thereby, e.g., modulate the level of expression of methylation repressed target genes.
  • a method may comprise administering to a subject, e.g. a subject in need thereof, a therapeutically effective amount of an agent described herein.
  • a compound of formula (I) or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for treating cancer, autoimmune and inflammatory diseases or conditions.
  • the disease or condition is cancer.
  • cancer diseases and conditions in which compounds of formula (I), or a pharmaceutically acceptable salt or solvates thereof may have potentially beneficial antitumour effects include, but are not limited to, cancers of the lung, bone, pancreas, skin, head, neck, uterus, ovaries, stomach, colon, breast, esophagus, small intestine, bowel, endocrine system, thyroid glad, parathyroid gland, adrenal gland, urethra, prostate, penis, testes, ureter, bladder, kidney or liver; rectal cancer; cancer of the anal region; carcinomas of the fallopian tubes, endometrium, cervix, vagina, vulva, renal pelvis, renal cell; sarcoma of soft tissue; myxoma; rhabdomyoma; fibroma; lipoma; teratoma; cholangiocarcinoma; hepatoblastoma; angiosarcoma; hemagioma; hepatoma;
  • the compounds of the compound of formula (I) or a pharmaceutically acceptable salt thereof may also be useful in the treatment of one or more diseases afflicting mammals which are characterized by cellular proliferation in the area of disorders associated with neo-vascularization and/or vascular permeability including blood vessel proliferative disorders including arthritis (rheumatoid arthritis) and restenosis; fibrotic disorders including hepatic cirrhosis and atherosclerosis; mesangial cell proliferative disorders include glomerulonephritis, diabetic nephropathy, malignant nephrosclerosis, thrombotic microangiopathy syndromes, proliferative retinopathies, organ transplant rejection and glomerulopathies; and metabolic disorders include psoriasis, diabetes mellitus, chronic wound healing, inflammation and neurodegenerative diseases.
  • diseases afflicting mammals which are characterized by cellular proliferation in the area of disorders associated with neo-vascularization and/or vascular permeability including blood vessel proliferative disorders
  • the compound of compound of formula (I) or a pharmaceutically acceptable salt thereof may be employed with other therapeutic methods of cancer treatment.
  • combination therapy with other chemotherapeutic, hormonal, antibody agents as well as surgical and/or radiation treatments other than those mentioned above are envisaged.
  • therapeutically effective amounts of the compound of compound of formula (I) or a pharmaceutically acceptable salt thereof are discussed above.
  • the therapeutically effective amount of the further therapeutic agents of the present invention will depend upon a number of factors including, for example, the age and weight of the mammal, the precise condition requiring treatment, the severity of the condition, the nature of the formulation, and the route of administration. Ultimately, the therapeutically effective amount will be at the discretion of the attendant physician or veterinarian. The relative timings of administration will be selected in order to achieve the desired combined therapeutic effect.
  • the further anti-cancer therapy is surgical and/or radiotherapy.
  • the further anti-cancer therapy is at least one additional anti-neoplastic agent.
  • anti-neoplastic agent that has activity versus a susceptible tumor being treated may be utilized in the combination.
  • Typical anti-neoplastic agents useful include, but are not limited to, anti-microtubule agents such as diterpenoids and vinca alkaloids; platinum coordination complexes; alkylating agents such as nitrogen mustards, oxazaphosphorines, alkylsulfonates, nitrosoureas, and triazenes; antibiotic agents such as anthracyclins, actinomycins and bleomycins; topoisomerase II inhibitors such as epipodophyllotoxins; antimetabolites such as purine and pyrimidine analogues and anti-folate compounds; topoisomerase I inhibitors such as camptothecins; hormones and hormonal analogues; signal transduction pathway inhibitors; non-receptor tyrosine angiogenesis inhibitors; immunotherapeutic agents; proapoptotic agents; and cell cycle signaling inhibitors.
  • Anti-microtubule or anti-mitotic agents are examples of anti-microtubule or anti-mitotic agents.
  • Anti-microtubule or anti-mitotic agents are phase specific agents active against the microtubules of tumor cells during M or the mitosis phase of the cell cycle.
  • anti-microtubule agents include, but are not limited to, diterpenoids and vinca alkaloids.
  • Diterpenoids which are derived from natural sources, are phase specific anti -cancer agents that operate at the G 2 /M phases of the cell cycle. It is believed that the diterpenoids stabilize the ⁇ -tubulin subunit of the microtubules, by binding with this protein. Disassembly of the protein appears then to be inhibited with mitosis being arrested and cell death following. Examples of diterpenoids include, but are not limited to, paclitaxel and its analog docetaxel.
  • Paclitaxel 5p,20-epoxy-1 ,2 ,4,7p,10p,13 -hexa-hydroxytax-1 1 -en-9-one 4,10-diacetate 2-benzoate 13-ester with (2R,3S)-N-benzoyl-3-phenylisoserine; is a natural diterpene product isolated from the Pacific yew tree Taxus brevifolia and is commercially available as an injectable solution TAXOL®. It is a member of the taxane family of terpenes.
  • Paclitaxel has been approved for clinical use in the treatment of refractory ovarian cancer in the United States (Markman et al., Yale Journal of Biology and Medicine, 64:583, 1991 ; McGuire et al., Ann. Intern, Med., 1 1 1 :273,1989) and for the treatment of breast cancer (Holmes et al., J. Nat. Cancer Inst., 83:1797,1991 .) It is a potential candidate for treatment of neoplasms in the skin (Einzig et. al., Proc. Am. Soc. Clin. Oncol., 20:46) and head and neck carcinomas (Forastire et. al., Sem. Oncol., 20:56, 1990).
  • the compound also shows potential for the treatment of polycystic kidney disease (Woo et. al., Nature, 368:750. 1994), lung cancer and malaria.
  • Treatment of patients with paclitaxel results in bone marrow suppression (multiple cell lineages, Ignoff, R.J. et. al, Cancer Chemotherapy Pocket Guide i 1998) related to the duration of dosing above a threshold concentration (50nM) (Kearns, CM. et. al., Seminars in Oncology, 3(6) p.16-23, 1995).
  • Docetaxel (2R.3S)- N-carboxy-3-phenylisoserine,N-fe/ -butyl ester, 13-ester with ⁇ -20-epoxy- 1 ,2 ,4,7p,10p,13 -hexahydroxytax-1 1 -en-9-one 4-acetate 2-benzoate, trihydrate; is commercially available as an injectable solution as TAXOTERE®.
  • Docetaxel is indicated for the treatment of breast cancer.
  • Docetaxel is a semisynthetic derivative of paclitaxel q.v. , prepared using a natural precursor, 10-deacetyl-baccatin III, extracted from the needle of the European Yew tree.
  • Vinca alkaloids are phase specific anti-neoplastic agents derived from the periwinkle plant. Vinca alkaloids act at the M phase (mitosis) of the cell cycle by binding specifically to tubulin. Consequently, the bound tubulin molecule is unable to polymerize into microtubules. Mitosis is believed to be arrested in metaphase with cell death following. Examples of vinca alkaloids include, but are not limited to, vinblastine, vincristine, and vinorelbine.
  • Vinblastine vincaleukoblastine sulfate
  • VELBAN® an injectable solution.
  • Myelosuppression is the dose limiting side effect of vinblastine.
  • Vincristine vincaleukoblastine, 22-oxo-, sulfate
  • ONCOVIN® an injectable solution.
  • Vincristine is indicated for the treatment of acute leukemias and has also found use in treatment regimens for Hodgkin's and non-Hodgkin's malignant lymphomas.
  • Alopecia and neurologic effects are the most common side effect of vincristine and to a lesser extent myelosupression and gastrointestinal mucositis effects occur.
  • Vinorelbine 3',4'-didehydro -4'-deoxy-C'-norvincaleukoblastine [R-(R*,R*)-2,3-dihydroxybutanedioate (1 :2)(salt)], commercially available as an injectable solution of vinorelbine tartrate (NAVELBINE®), is a semisynthetic vinca alkaloid. Vinorelbine is indicated as a single agent or in combination with other chemotherapeutic agents, such as cisplatin, in the treatment of various solid tumors, particularly non- small cell lung, advanced breast, and hormone refractory prostate cancers. Myelosuppression is the most common dose limiting side effect of vinorelbine. Platinum coordination complexes:
  • Platinum coordination complexes are non-phase specific anti-cancer agents, which are interactive with DNA.
  • the platinum complexes enter tumor cells, undergo, aquation and form intra- and interstrand crosslinks with DNA causing adverse biological effects to the tumor.
  • Examples of platinum coordination complexes include, but are not limited to, oxaliplatin, cisplatin and carboplatin.
  • Cisplatin, cis-diamminedichloroplatinum is commercially available as PLATINOL® as an injectable solution. Cisplatin is primarily indicated in the treatment of metastatic testicular and ovarian cancer and advanced bladder cancer.
  • Carboplatin platinum, diammine [1 ,1 -cyclobutane-dicarboxylate(2-)-0,0'], is commercially available as PARAPLATIN® as an injectable solution. Carboplatin is primarily indicated in the first and second line treatment of advanced ovarian carcinoma.
  • Alkylating agents are non-phase anti-cancer specific agents and strong electrophiles. Typically, alkylating agents form covalent linkages, by alkylation, to DNA through nucleophilic moieties of the DNA molecule such as phosphate, amino, sulfhydryl, hydroxyl, carboxyl, and imidazole groups. Such alkylation disrupts nucleic acid function leading to cell death.
  • alkylating agents include, but are not limited to, nitrogen mustards such as cyclophosphamide, melphalan, and chlorambucil; alkyl sulfonates such as busulfan; nitrosoureas such as carmustine; and triazenes such as dacarbazine.
  • Cyclophosphamide 2-[bis(2-chloroethyl)amino]tetrahydro-2H-1 ,3,2-oxazaphosphorine 2-oxide monohydrate, is commercially available as an injectable solution or tablets as CYTOXAN®. Cyclophosphamide is indicated as a single agent or in combination with other chemotherapeutic agents, in the treatment of malignant lymphomas, multiple myeloma, and leukemias.
  • Melphalan 4-[bis(2-chloroethyl)amino]-L-phenylalanine, is commercially available as an injectable solution or tablets as ALKERAN®. Melphalan is indicated for the palliative treatment of multiple myeloma and non-resectable epithelial carcinoma of the ovary. Bone marrow suppression is the most common dose limiting side effect of melphalan.
  • Chlorambucil 4-[bis(2-chloroethyl)amino]benzenebutanoic acid, is commercially available as LEUKERAN® tablets. Chlorambucil is indicated for the palliative treatment of chronic lymphatic leukemia, and malignant lymphomas such as lymphosarcoma, giant follicular lymphoma, and Hodgkin's disease.
  • Busulfan 1 ,4-butanediol dimethanesulfonate, is commercially available as MYLERAN® TABLETS. Busulfan is indicated for the palliative treatment of chronic myelogenous leukemia.
  • Carmustine 1 ,3-[bis(2-chloroethyl)-1 -nitrosourea, is commercially available as single vials of lyophilized material as BiCNU®. Carmustine is indicated for the palliative treatment as a single agent or in combination with other agents for brain tumors, multiple myeloma, Hodgkin's disease, and non- Hodgkin's lymphomas.
  • dacarbazine 5-(3,3-dimethyl-1 -triazeno)-imidazole-4-carboxamide, is commercially available as single vials of material as DTIC-Dome®. dacarbazine is indicated for the treatment of metastatic malignant melanoma and in combination with other agents for the second line treatment of Hodgkin's Disease.
  • Antibiotic anti-neoplastics are non-phase specific agents, which bind or intercalate with DNA. Typically, such action results in stable DNA complexes or strand breakage, which disrupts ordinary function of the nucleic acids leading to cell death.
  • antibiotic anti-neoplastic agents include, but are not limited to, actinomycins such as dactinomycin, anthrocyclins such as daunorubicin and doxorubicin; and bleomycins.
  • Dactinomycin also know as Actinomycin D, is commercially available in injectable form as COSMEGEN® . Dactinomycin is indicated for the treatment of Wilm's tumor and rhabdomyosarcoma.
  • Daunorubicin (8S-cis-)-8-acetyl-10-[(3-amino-2,3,6-trideoxy- -L-lyxo-hexopyranosyl)oxy]-7,8,9,10- tetrahydro-6,8,1 1 -trihydroxy-1 -methoxy-5,12 naphthacenedione hydrochloride, is commercially available as a liposomal injectable form as DAUNOXOME® or as an injectable as CERUBIDINE®. Daunorubicin is indicated for remission induction in the treatment of acute nonlymphocytic leukemia and advanced HIV associated Kaposi's sarcoma.
  • Doxorubicin (8S, 10S)-10-[(3-amino-2,3,6-trideoxy- -L-lyxo-hexopyranosyl)oxy]-8-glycoloyl, 7,8,9,10- tetrahydro-6,8,1 1 -trihydroxy-1 -methoxy-5,12 naphthacenedione hydrochloride, is commercially available as an injectable form as RUBEX® or ADRIAMYCIN RDF®.
  • Doxorubicin is primarily indicated for the treatment of acute lymphoblastic leukemia and acute myeloblasts leukemia, but is also a useful component in the treatment of some solid tumors and lymphomas.
  • Bleomycin a mixture of cytotoxic glycopeptide antibiotics isolated from a strain of Streptomyces verticillus, is commercially available as BLENOXANE®. Bleomycin is indicated as a palliative treatment, as a single agent or in combination with other agents, of squamous cell carcinoma, lymphomas, and testicular carcinomas.
  • Topoisomerase II inhibitors include, but are not limited to, epipodophyllotoxins.
  • Epipodophyllotoxins are phase specific anti-neoplastic agents derived from the mandrake plant. Epipodophyllotoxins typically affect cells in the S and G 2 phases of the cell cycle by forming a ternary complex with topoisomerase II and DNA causing DNA strand breaks. The strand breaks accumulate and cell death follows. Examples of epipodophyllotoxins include, but are not limited to, etoposide and teniposide. Etoposide, 4'-demethyl-epipodophyllotoxin 9[4,6-0-(R )-ethylidene-p-D-glucopyranoside], is commercially available as an injectable solution or capsules as VePESID® and is commonly known as VP-16. Etoposide is indicated as a single agent or in combination with other chemotherapy agents in the treatment of testicular and non-small cell lung cancers.
  • Teniposide 4'-demethyl-epipodophyllotoxin 9[4,6-0-(R )-thenylidene-p-D-glucopyranoside], is commercially available as an injectable solution as VUMON® and is commonly known as VM-26. Teniposide is indicated as a single agent or in combination with other chemotherapy agents in the treatment of acute leukemia in children.
  • Antimetabolite neoplastic agents are:
  • Antimetabolite neoplastic agents are phase specific anti-neoplastic agents that act at S phase (DNA synthesis) of the cell cycle by inhibiting DNA synthesis or by inhibiting purine or pyrimidine base synthesis and thereby limiting DNA synthesis. Consequently, S phase does not proceed and cell death follows.
  • Examples of antimetabolite anti-neoplastic agents include, but are not limited to, fluorouracil, methotrexate, cytarabine, mecaptopurine, thioguanine, and gemcitabine.
  • 5-fluorouracil 5-fluoro-2,4- (1 H,3H) pyrimidinedione
  • fluorouracil is commercially available as fluorouracil.
  • Administration of 5-fluorouracil leads to inhibition of thymidylate synthesis and is also incorporated into both RNA and DNA. The result typically is cell death.
  • 5-fluorouracil is indicated as a single agent or in combination with other chemotherapy agents in the treatment of carcinomas of the breast, colon, rectum, stomach and pancreas.
  • Other fluoropyrimidine analogs include 5-fluoro deoxyuridine (floxuridine) and 5-fluorodeoxyuridine monophosphate.
  • Cytarabine 4-amino-1 -p-D-arabinofuranosyl-2 (I H)-pyrimidinone, is commercially available as CYTOSAR-U® and is commonly known as Ara-C. It is believed that cytarabine exhibits cell phase specificity at S-phase by inhibiting DNA chain elongation by terminal incorporation of cytarabine into the growing DNA chain. Cytarabine is indicated as a single agent or in combination with other chemotherapy agents in the treatment of acute leukemia. Other cytidine analogs include 5- azacytidine and 2',2'-difluorodeoxycytidine (gemcitabine).
  • Mercaptopurine 1 ,7-dihydro-6H-purine-6-thione monohydrate, is commercially available as PURINETHOL®.
  • Mercaptopurine exhibits cell phase specificity at S-phase by inhibiting DNA synthesis by an as of yet unspecified mechanism.
  • Mercaptopurine is indicated as a single agent or in combination with other chemotherapy agents in the treatment of acute leukemia.
  • a useful mercaptopurine analog is azathioprine.
  • Thioguanine 2-amino-1 ,7-dihydro-6H-purine-6-thione
  • TABLOID® Thioguanine exhibits cell phase specificity at S-phase by inhibiting DNA synthesis by an as of yet unspecified mechanism.
  • Thioguanine is indicated as a single agent or in combination with other chemotherapy agents in the treatment of acute leukemia.
  • Other purine analogs include pentostatin, erythrohydroxynonyladenine, fludarabine phosphate, and cladribine.
  • Gemcitabine 2'-deoxy-2', 2'-difluorocytidine monohydrochloride ( ⁇ -isomer), is commercially available as GEMZAR®.
  • Gemcitabine exhibits cell phase specificity at S-phase and by blocking progression of cells through the G1/S boundary. Gemcitabine is indicated in combination with cisplatin in the treatment of locally advanced non-small cell lung cancer and alone in the treatment of locally advanced pancreatic cancer.
  • Methotrexate N-[4[[(2,4-diamino-6-pteridinyl) methyljmethylamino] benzoyl]-L-glutamic acid, is commercially available as methotrexate sodium. Methotrexate exhibits cell phase effects specifically at S-phase by inhibiting DNA synthesis, repair and/or replication through the inhibition of dyhydrofolic acid reductase which is required for synthesis of purine nucleotides and thymidylate.
  • Methotrexate is indicated as a single agent or in combination with other chemotherapy agents in the treatment of choriocarcinoma, meningeal leukemia, non-Hodgkin's lymphoma, and carcinomas of the breast, head, neck, ovary and bladder.
  • Topoisomerase I inhibitors are indicated as a single agent or in combination with other chemotherapy agents in the treatment of choriocarcinoma, meningeal leukemia, non-Hodgkin's lymphoma, and carcinomas of the breast, head, neck, ovary and bladder.
  • Camptothecins including, camptothecin and camptothecin derivatives are available or under development as Topoisomerase I inhibitors. Camptothecins cytotoxic activity is believed to be related to its Topoisomerase I inhibitory activity. Examples of camptothecins include, but are not limited to irinotecan, topotecan, and the various optical forms of 7-(4-methylpiperazino-methylene)-10,1 1 - ethylenedioxy-20-camptothecin described below.
  • Irinotecan is a derivative of camptothecin which binds, along with its active metabolite SN-38, to the topoisomerase I - DNA complex.
  • cytotoxicity occurs as a result of irreparable double strand breaks caused by interaction of the topoisomerase I : DNA : irintecan or SN-38 ternary complex with replication enzymes.
  • Irinotecan is indicated for treatment of metastatic cancer of the colon or rectum.
  • Topotecan HCI (S)-10-[(dimethylamino)methyl]-4-ethyl-4,9-dihydroxy-1 H- pyrano[3',4',6,7]indolizino[1 ,2-b]quinoline-3,14-(4H,12H)-dione monohydrochloride, is commercially available as the injectable solution HYCAMTIN®.
  • Topotecan is a derivative of camptothecin which binds to the topoisomerase I - DNA complex and prevents religation of singles strand breaks caused by Topoisomerase I in response to torsional strain of the DNA molecule. Topotecan is indicated for second line treatment of metastatic carcinoma of the ovary and small cell lung cancer.
  • Hormones and hormonal analogues are hormones and hormonal analogues:
  • Hormones and hormonal analogues are useful compounds for treating cancers in which there is a relationship between the hormone(s) and growth and/or lack of growth of the cancer.
  • hormones and hormonal analogues useful in cancer treatment include, but are not limited to, adrenocorticosteroids such as prednisone and prednisolone which are useful in the treatment of malignant lymphoma and acute leukemia in children ; aminoglutethimide and other aromatase inhibitors such as anastrozole, letrazole, vorazole, and exemestane useful in the treatment of adrenocortical carcinoma and hormone dependent breast carcinoma containing estrogen receptors; progestrins such as megestrol acetate useful in the treatment of hormone dependent breast cancer and endometrial carcinoma; estrogens, androgens, and anti-androgens such as flutamide, nilutamide, bicalutamide, cyproterone acetate and 5 -reductases
  • GnRH gonadotropin-releasing hormone
  • LH leutinizing hormone
  • FSH follicle stimulating hormone
  • Signal transduction pathway inhibitors are those inhibitors, which block or inhibit a chemical process which evokes an intracellular change. As used herein this change is cell proliferation or differentiation.
  • Signal tranduction inhibitors useful in the present invention include inhibitors of receptor tyrosine kinases, non-receptor tyrosine kinases, SH2/SH3domain blockers, serine/threonine kinases, phosphotidyl inositol-3 kinases, myo-inositol signaling, and Ras oncogenes.
  • Protein tyrosine kinases catalyse the phosphorylation of specific tyrosyl residues in various proteins involved in the regulation of cell growth.
  • Such protein tyrosine kinases can be broadly classified as receptor or non-receptor kinases.
  • Receptor tyrosine kinases are transmembrane proteins having an extracellular ligand binding domain, a transmembrane domain, and a tyrosine kinase domain.
  • Receptor tyrosine kinases are involved in the regulation of cell growth and are generally termed growth factor receptors. Inappropriate or uncontrolled activation of many of these kinases, i.e.
  • aberrant kinase growth factor receptor activity for example by over-expression or mutation, has been shown to result in uncontrolled cell growth. Accordingly, the aberrant activity of such kinases has been linked to malignant tissue growth. Consequently, inhibitors of such kinases could provide cancer treatment methods.
  • Growth factor receptors include, for example, epidermal growth factor receptor (EGFr), platelet derived growth factor receptor (PDGFr), erbB2, erbB4, ret, vascular endothelial growth factor receptor (VEGFr), tyrosine kinase with immunoglobulin-like and epidermal growth factor homology domains (TIE-2), insulin growth factor -I (IGFI) receptor, macrophage colony stimulating factor (cfms), BTK, ckit, cmet, fibroblast growth factor (FGF) receptors, Trk receptors (TrkA, TrkB, and TrkC), ephrin (eph) receptors, and the RET protooncogene.
  • EGFr epidermal growth factor receptor
  • PDGFr platelet derived growth factor receptor
  • erbB2 erbB2
  • VEGFr vascular endothelial growth factor receptor
  • TIE-2 immunoglobulin-like and epidermal growth factor homo
  • inhibitors of growth receptors include ligand antagonists, antibodies, tyrosine kinase inhibitors and anti-sense oligonucleotides.
  • Growth factor receptors and agents that inhibit growth factor receptor function are described, for instance, in Kath, John C, Exp. Opin. Ther. Patents (2000) 10(6):803-818; Shawver et al DDT Vol 2, No. 2 February 1997; and Lofts, F. J. et al, "Growth factor receptors as targets", New Molecular Targets for Cancer Chemotherapy, ed. Workman, Paul and Kerr, David, CRC press 1994, London.
  • Non-receptor tyrosine kinases which are not growth factor receptor kinases are termed non-receptor tyrosine kinases.
  • Non-receptor tyrosine kinases useful in the present invention include cSrc, Lck, Fyn, Yes, Jak, cAbl, FAK (Focal adhesion kinase), Brutons tyrosine kinase, and Bcr-Abl.
  • Such non-receptor kinases and agents which inhibit nonreceptor tyrosine kinase function are described in Sinh, S.
  • SH2/SH3 domain blockers are agents that disrupt SH2 or SH3 domain binding in a variety of enzymes or adaptor proteins including, PI3-K p85 subunit, Src family kinases, adaptor molecules (She, Crk, Nek, Grb2) and Ras-GAP.
  • SH2/SH3 domains as targets for anti-cancer drugs are discussed in Smithgall, T.E. (1995), Journal of Pharmacological and Toxicological Methods. 34(3) 125-32.
  • Inhibitors of Serine/Threonine Kinases including MAP kinase cascade blockers which include blockers of Raf kinases (rafk), Mitogen or Extracellular Regulated Kinase (MEKs), and Extracellular Regulated Kinases (ERKs); and Protein kinase C family member blockers including blockers of PKCs (alpha, beta, gamma, epsilon, mu, lambda, iota, zeta).
  • IkB kinase family IKKa, IKKb
  • PKB family kinases akt kinase family members
  • TGF beta receptor kinases TGF beta receptor kinases.
  • Serine/Threonine kinases and inhibitors thereof are described in Yamamoto, T., Taya, S., Kaibuchi, K., (1999), Journal of Biochemistry. 126 (5) 799-803; Brodt, P, Samani, A., and Navab, R. (2000), Biochemical Pharmacology, 60. 1 101 -1 107; Massague, J., Weis-Garcia, F. (1996) Cancer Surveys. 27:41 -64; Philip, P.A., and Harris, A.L. (1995), Cancer Treatment and Research. 78: 3-27, Lackey, K. et al Bioorganic and Medicinal Chemistry Letters, (10), 2000, 223-226; U.S. Patent No. 6,268,391 ; and Martinez-lacaci, L., et al, Int. J. Cancer (2000), 88(1), 44-52.
  • Inhibitors of Phosphotidyl inositol-3 Kinase family members including blockers of PI3-kinase, ATM, DNA-PK, and Ku are also useful in the present invention.
  • Such kinases are discussed in Abraham, R.T. (1996), Current Opinion in Immunology. 8 (3) 412-8; Canman, C.E., Lim, D.S. (1998), Oncogene 17 (25) 3301 -3308; Jackson, S.P. (1997), International Journal of Biochemistry and Cell Biology. 29 (7):935-8; and Zhong, H. et al, Cancer res, (2000) 60(6), 1541 -1545.
  • Myo-inositol signaling inhibitors such as phospholipase C blockers and Myoinositol analogues.
  • signal inhibitors are described in Powis, G., and Kozikowski A., (1994) New Molecular Targets for Cancer Chemotherapy ed., Paul Workman and David Kerr, CRC press 1994, London.
  • Ras Oncogene Another group of signal transduction pathway inhibitors are inhibitors of Ras Oncogene.
  • Such inhibitors include inhibitors of farnesyltransferase, geranyl-geranyl transferase, and CAAX proteases as well as anti-sense oligonucleotides, ribozymes and immunotherapy.
  • Such inhibitors have been shown to block ras activation in cells containing wild type mutant ras , thereby acting as antiproliferation agents.
  • Ras oncogene inhibition is discussed in Scharovsky, O.G., Rozados, V.R., Gervasoni, S.I. Matar, P. (2000), Journal of Biomedical Science. 7(4) 292-8; Ashby, M.N. (1998), Current Opinion in Lipidology. 9 (2) 99 - 102; and BioChim. Biophys. Acta, (19899) 1423(3):19-30.
  • antibody antagonists to receptor kinase ligand binding may also serve as signal transduction inhibitors.
  • This group of signal transduction pathway inhibitors includes the use of humanized antibodies to the extracellular ligand binding domain of receptor tyrosine kinases.
  • Imclone C225 EGFR specific antibody see Green, M.C. et al, Monoclonal Antibody Therapy for Solid Tumors, Cancer Treat.
  • Herceptin ® erbB2 antibody see Tyrosine Kinase Signalling in Breast cancererbB Family Receptor Tyrosine Kinases, Breast cancer Res., 2000, 2(3), 176-183
  • 2CB VEGFR2 specific antibody see Brekken, R.A. et al, Selective Inhibition of VEGFR2 Activity by a monoclonal Anti-VEGF antibody blocks tumor growth in mice, Cancer Res. (2000) 60, 51 17-5124).
  • Anti-angiogenic agents including non-receptorMEKngiogenesis inhibitors may alo be useful.
  • Anti-angiogenic agents such as those which inhibit the effects of vascular edothelial growth factor, (for example the anti-vascular endothelial cell growth factor antibody bevacizumab [AvastinTM], and compounds that work by other mechanisms (for example linomide, inhibitors of integrin ⁇ 3 function, endostatin and angiostatin);
  • Immunotherapy approaches including for example ex-vivo and in-vivo approaches to increase the immunogenecity of patient tumour cells, such as transfection with cytokines such as interleukin 2, interleukin 4 or granulocyte-macrophage colony stimulating factor, approaches to decrease T-cell anergy, approaches using transfected immune cells such as cytokine-transfected dendritic cells, approaches using cytokine-transfected tumour cell lines and approaches using anti- idiotypic antibodies
  • cytokines such as interleukin 2, interleukin 4 or granulocyte-macrophage colony stimulating factor
  • approaches to decrease T-cell anergy approaches using transfected immune cells such as cytokine-transfected dendritic cells
  • Proapoptotoc agents including for example ex-vivo and in-vivo approaches to increase the immunogenecity of patient tumour cells, such as transfection with cytok
  • Agents used in proapoptotic regimens may also be used in the combination of the present invention.
  • Cell cycle signalling inhibitors inhibit molecules involved in the control of the cell cycle.
  • a family of protein kinases called cyclin dependent kinases (CDKs) and their interaction with a family of proteins termed cyclins controls progression through the eukaryotic cell cycle. The coordinate activation and inactivation of different cyclin/CDK complexes is necessary for normal progression through the cell cycle.
  • CDKs cyclin dependent kinases
  • Several inhibitors of cell cycle signalling are under development. For instance, examples of cyclin dependent kinases, including CDK2, CDK4, and CDK6 and inhibitors for the same are described in, for instance, Rosania et al, Exp. Opin. Ther. Patents (2000) 10(2):215-230.
  • the combination of the present invention comprises a compound of formula I or a pharmaceutically acceptable salt thereof and at least one anti-neoplastic agent selected from anti- microtubule agents, platinum coordination complexes, alkylating agents, antibiotic agents, topoisomerase II inhibitors, antimetabolites, topoisomerase I inhibitors, hormones and hormonal analogues, signal transduction pathway inhibitors, non-receptor tyrosine MEKngiogenesis inhibitors, immunotherapeutic agents, proapoptotic agents, and cell cycle signaling inhibitors.
  • anti-neoplastic agent selected from anti- microtubule agents, platinum coordination complexes, alkylating agents, antibiotic agents, topoisomerase II inhibitors, antimetabolites, topoisomerase I inhibitors, hormones and hormonal analogues, signal transduction pathway inhibitors, non-receptor tyrosine MEKngiogenesis inhibitors, immunotherapeutic agents, proapoptotic agents, and cell cycle signaling inhibitors.
  • the combination of the present invention comprises a compound of formula I or a pharmaceutically acceptable salt thereof and at least one anti-neoplastic agent which is an anti- microtubule agent selected from diterpenoids and vinca alkaloids.
  • the at least one anti-neoplastic agent agent is a diterpenoid.
  • the at least one anti-neoplastic agent is a vinca alkaloid.
  • the combination of the present invention comprises a compound of formula I or a salt or solvate thereof and at least one anti-neoplastic agent, which is a platinum coordination complex.
  • the at least one anti-neoplastic agent is paclitaxel, carboplatin, or vinorelbine.
  • the at least one anti-neoplastic agent is carboplatin. In a further embodiment, the at least one anti-neoplastic agent is vinorelbine. In a further embodiment, the at least one anti-neoplastic agent is paclitaxel.
  • the combination of the present invention comprises a compound of formula I or a pharmaceutically acceptable salt thereof and at least one anti-neoplastic agent which is a signal transduction pathway inhibitor.
  • the signal transduction pathway inhibitor is an inhibitor of a growth factor receptor kinase VEGFR2, TIE2, PDGFR, BTK, erbB2, EGFr, IGFR-1 , TrkA, TrkB, TrkC, or c-fms.
  • the signal transduction pathway inhibitor is an inhibitor of a serine/threonine kinase rafk, akt, or PKC-zeta.
  • the signal transduction pathway inhibitor is an inhibitor of a non- receptor tyrosine kinase selected from the src family of kinases.
  • the signal transduction pathway inhibitor is an inhibitor of c-src. In a further embodiment the signal transduction pathway inhibitor is an inhibitor of Ras oncogene selected from inhibitors of farnesyl transferase and geranylgeranyl transferase. In a further embodiment the signal transduction pathway inhibitor is an inhibitor of a serine/threonine kinase selected from the group consisting of PI3K.
  • the signal transduction pathway inhibitor is a dual EGFr/erbB2 inhibitor, for example N- ⁇ 3-Chloro-4-[(3-fluorobenzyl) oxy]phenyl ⁇ -6-[5-( ⁇ [2-(methanesulphonyl)
  • the combination of the present invention comprises a compound of formula (I) or a pharmaceutically acceptable salt thereof and at least one anti-neoplastic agent which is a cell cycle signaling inhibitor.
  • cell cycle signaling inhibitor is an inhibitor of CDK2, CDK4 or CDK6.
  • the disease or condition is autoimmune.
  • Autoimmune diseases associated with type 1 interferon include, but are not limited to Systemic lupus erythematosus, Psoriasis, insulin-dependent diabetes mellitus (IDDM), dermatomyositis and Sjogren's syndrome (SS).
  • IDDM insulin-dependent diabetes mellitus
  • SS Sjogren's syndrome
  • the disease or condition is inflammation.
  • Inflammation represents a group of vascular, cellular and neurological responses to trauma. Inflammation can be characterised as the movement of inflammatory cells such as monocytes, neutrophils and granulocytes into the tissues. This is usually associated with reduced endothelial barrier function and oedema into the tissues. Inflammation can be classified as either acute or chronic. Acute inflammation is the initial response of the body to harmful stimuli and is achieved by the increased movement of plasma and leukocytes from the blood into the injured tissues. A cascade of biochemical event propagates and matures the inflammatory response, involving the local vascular system, the immune system, and various cells within the injured tissue. Prolonged inflammation, known as chronic inflammation, leads to a progressive shift in the type of cells which are present at the site of inflammation and is characterised by simultaneous destruction and healing of the tissue from the inflammatory process.
  • Acute inflammation is the initial response of the body to harmful stimuli and is achieved by the increased movement of plasma and leukocytes from the blood into the injured tissues.
  • a cascade of biochemical event propag
  • inflammation When occurring as part of an immune response to infection or as an acute response to trauma, inflammation can be beneficial and is normally self-limiting. However, inflammation can be detrimental under various conditions. This includes the production of excessive inflammation in response to infectious agents, which can lead to significant organ damage and death (for example, in the setting of sepsis). Moreover, chronic inflammation is generally deleterious and is at the root of numerous chronic diseases, causing severe and irreversible damage to tissues. In such settings, the immune response is often directed against self-tissues (autoimmunity), although chronic responses to foreign entities can also lead to bystander damage to self tissues.
  • autoimmunity autoimmunity
  • the aim of anti-inflammatory therapy is therefore to reduce this inflammation, to inhibit autoimmunity when present and to allow for the physiological process or healing and tissue repair to progress.
  • the compound of formula (I) may be used to treat inflammation of any tissue and organs of the body, including musculoskeletal inflammation, vascular inflammation, neural inflammation, digestive system inflammation, ocular inflammation, inflammation of the reproductive system, and other inflammation, as exemplified below.
  • Musculoskeletal inflammation refers to any inflammatory condition of the musculoskeletal system, particularly those conditions affecting skeletal joints, including joints of the hand, wrist, elbow, shoulder, jaw, spine, neck, hip, knew, ankle, and foot, and conditions affecting tissues connecting muscles to bones such as tendons.
  • musculoskeletal inflammation which may be treated with compounds of formula (I) include arthritis (including, for example, osteoarthritis, rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis, acute and chronic infectious arthritis, arthritis associated with gout and pseudogout, and juvenile idiopathic arthritis), tendonitis, synovitis, tenosynovitis, bursitis, fibrositis (fibromyalgia), epicondylitis, myositis, and osteitis (including, for example, Paget's disease, osteitis pubis, and osteitis fibrosa cystic).
  • arthritis including, for example, osteoarthritis, rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis, acute and chronic infectious arthritis, arthritis associated with gout and pseudogout, and juvenile idiopathic arthritis
  • tendonitis synovitis, tenosynovit
  • Ocular inflammation refers to inflammation of any structure of the eye, including the eye lids.
  • ocular inflammation which may be treated with the compounds of formula (I) include blepharitis, blepharochalasis, conjunctivitis, dacryoadenitis, keratitis, keratoconjunctivitis sicca (dry eye), scleritis, trichiasis, and uveitis.
  • inflammation of the nervous system which may be treated with the compounds of formula (I) include encephalitis, Guillain-Barre syndrome, meningitis, neuromyotonia, narcolepsy, multiple sclerosis, myelitis and schizophrenia.
  • inflammation of the vasculature or lymphatic system which may be treated with the compounds of formula (I) include arthrosclerosis, arthritis, phlebitis, vasculitis, and lymphangitis.
  • Examples of inflammatory conditions of the digestive system which may be treated with the compounds of formula (I) include cholangitis, cholecystitis, enteritis, enterocolitis, gastritis, gastroenteritis, inflammatory bowel disease (such as Crohn's disease and ulcerative colitis), ileitis, and proctitis.
  • Examples of inflammatory conditions of the reproductive system which may be treated with the compounds of formula (I) include cervicitis, chorioamnionitis, endometritis, epididymitis, omphalitis, oophoritis, orchitis, salpingitis, tubo-ovarian abscess, urethritis, vaginitis, vulvitis, and vulvodynia.
  • the compound of formula (I) may be used to treat autoimmune conditions having an inflammatory component.
  • Such conditions include acute disseminated alopecia universalise, Behcet's disease, Chagas' disease, chronic fatigue syndrome, dysautonomia, encephalomyelitis, ankylosing spondylitis, aplastic anemia, hidradenitis suppurativa, autoimmune hepatitis, autoimmune oophoritis, celiac disease, Crohn's disease, diabetes mellitus type 1 , giant cell arteritis, goodpasture's syndrome, Grave's disease, Guillain-Barre syndrome, Hashimoto's disease, Henoch-Schonlein purpura, Kawasaki's disease, lupus erythematosus, microscopic colitis, microscopic polyarteritis, mixed connective tissue disease, multiple sclerosis, myasthenia gravis, opsoclonus myoclonus syndrome, optic neuritis,
  • the compound of formula (I) may be used to treat T-cell mediated hypersensitivity diseases having an inflammatory component.
  • T-cell mediated hypersensitivity diseases having an inflammatory component.
  • Such conditions include contact hypersensitivity, contact dermatitis (including that due to poison ivy), uticaria, skin allergies, respiratory allergies (hayfever, allergic rhinitis) and gluten-sensitive enteropathy (Celliac disease).
  • inflammatory conditions which may be treated with the agents include, for example, appendicitis, dermatitis, dermatomyositis, endocarditis, fibrositis, gingivitis, glossitis, hepatitis, hidradenitis suppurativa, ulceris, laryngitis, mastitis, myocarditis, nephritis, otitis, pancreatitis, parotitis, percarditis, peritonoitis, pharyngitis, pleuritis, pneumonitis, prostatistis, pyelonephritis, and stomatisi, transplant rejection (involving organs such as kidney, liver, heart, lung, pancreas (e.g., islet cells), bone marrow, cornea, small bowel, skin allografts, skin homografts, and heart valve xengrafts, sewrum sickness, and graft vs host disease), acute pan
  • Preferred treatments include treatment of transplant rejection, rheumatoid arthritis, psoriatic arthritis, multiple sclerosis, Type 1 diabetes, asthma, inflammatory bowel disease, systemic lupus erythematosis, psoriasis, chronic obstructive pulmonary disease, and inflammation accompanying infectious conditions (e.g., sepsis).
  • the compound of formula (I) and pharmaceutical compositions containing it may be used in combination with or include one or more other therapeutic agents, for example selected from NSAIDS, corticosteroids, COX-2 inhibitors, cytokine inhibitors, anti-TNF agents, inhibitors oncostatin M, antimalarials, immunsuppressive and cytostatics.
  • one or more other therapeutic agents for example selected from NSAIDS, corticosteroids, COX-2 inhibitors, cytokine inhibitors, anti-TNF agents, inhibitors oncostatin M, antimalarials, immunsuppressive and cytostatics.
  • DCM refers to dichloromethane
  • DIPEA refers to /V,/V-diisopropylethylamine
  • DMAP refers to A/,/V-dimethyl-4-pyridinamine
  • DMSO dimethylsulfoxide
  • DMF refers to A/,A/-dimethylformamide
  • DMA refers to dimethyl acetamide
  • Ether refers to diethyl ether
  • EtOAC refers to ethyl acetate
  • HATU refers to A/-[(dimethylamino)(3-oxido-1 /-/-[1 ,2,3]triazolo[4,5-Jb]pyridin-1 -yl)methylidene]-/V- methylmethanaminium hexafluorophosphate
  • HPLC refers to high performance liquid chromatography
  • MDAP refers to mass-directed autopreparative HPLC
  • NEt 3 refers to triethylamine
  • r.t. refers to room temperature
  • Rt refers to retention time
  • THF refers to tetrahydrofuran
  • TFA refers to trifluoroacetic acid
  • Tos refers to para-toluene sulfonyl
  • TosCI refers to para-toluene sulfonyl chloride H NMR spectra were recorded using a Varian NMR 400 MHz, referenced to tetramethylsilane.
  • LC/MS Method A was conducted on an Acquity UPLC BEH C18 column (50mm x 2.1 mm i.d.
  • LC/MS (Method B) was conducted on an Acquity UPLC BEH C18 column (50mm x 2.1 mm i.d. 1 .7 ⁇ packing diameter) at 40 degrees centigrade, eluting with 10 mM Ammonium Bicarbonate in water adjusted to pH 10 with Ammonia solution (Solvent A) and Acetonitrile (Solvent B) using the following elution gradient 0-1 .5min 1 - 97% B, 1 .5-1 .9min 97% B, 1 .9 - 2.1 min 100% B at a flow rate of 1 ml/min.
  • the UV detection was a summed signal from wavelength of 210nm to 350nm.
  • the mass spectra were recorded on a Waters ZQ Mass Spectrometer using Alternate-scan Positive and Negative Electrospray. lonisation data was rounded to the nearest integer.
  • LC/MS (Method C) was conducted on an Acquity UPLC BEH C18 column (50mm x 2.1 mm i.d. 1 .7 ⁇ packing diameter) at 40 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 0-1 .5min 3 - 100% B, 1 .5-1.9min 100% B, 1 .9 - 2.0min 3% B at a flow rate of 1 ml/min.
  • the UV detection was a summed signal from wavelength of 210nm to 350nm.
  • the UV detection was a summed signal from wavelength of 214nm to
  • LCMS (Method H) was conducted on a HALO C18 Column (50mm x 4.6 mm i.d. 2.7 ⁇ packing diameter) at 40 degrees centigrade, eluting with Water (Solvent A) and Acetonitrile (Solvent B) using the following elution gradient 0-1 .Omin 5 - 95% B, 1 .0-2. Omin - 95%, 2.0-2.01 min - 95-5%, 2.01 - 2.5min - 5% at a flow rate of 1 .8ml/min.
  • the UV detection was a summed signal from wavelength of 214nm to 254nm.
  • the UV detection was from wavelength 214nm to 254nm.
  • Silica chromatography techniques include either automated (Flashmaster, Biotage SP4) techniques manual chromatography on pre-packed cartridges (SPE) or manually-packed flash columns.
  • MDAP mass-directed autopreparative chromatography
  • Method E The HPLC analysis was conducted on an XBridge C18 column (100mm x 30mm i.d. 5 ⁇ packing diameter) at ambient temperature, eluting with 10mM Ammonium Bicarbonate in water adjusted to pH 10 with Ammonia solution (Solvent A) and Acetonitrile (Solvent B) using an elution gradient of between 0 and 100% over 15 or 25 minutes.
  • Solvent A Ammonia solution
  • Solvent B Acetonitrile
  • the UV detection was an averaged signal from wavelength of 210nm to 350nm.
  • the mass spectra were recorded on a Waters ZQ Mass Spectrometer using Alternate-scan Positive and Negative Electrospray. lonisation data was rounded to the nearest integer.
  • the UV detection was an averaged signal from wavelength of 210nm to 350nm.
  • the mass spectra were recorded on a Waters ZQ Mass Spectrometer using Alternate-scan Positive and Negative Electrospray. lonisation data was rounded to the nearest integer.
  • the UV detection was an averaged signal from wavelength of 210nm to 350nm.
  • the mass spectra were recorded on a Waters ZQ Mass Spectrometer using Alternate-scan Positive and Negative Electrospray. lonisation data was rounded to the nearest integer.
  • the UV detection was a summed signal from wavelength of 200nm to 310nm.
  • the mass spectra were recorded on a Waters ZQ Mass Spectrometer using Alternate-scan Positive and Negative Electrospray. lonisation data was rounded to the nearest integer.
  • MDAP Method J
  • the HPLC was conducted on a Gemini C18 column (150mm x 21 ,2mm i.d. 5um packing diameter), eluting with Acetonitrile (Solvent A) and a 0.1 % solution of Trifluoroacetic acid in water (Solvent B) using the following elution gradient 0-10min 40-50%B, 10-1 1 min 50-5%B, 1 1 -14min 5%B, 14-15min 5 -40%B at a flow rate of 20ml/min.
  • the UV detection was a summed signal from wavelength of 214nm to 254 nm.
  • the reaction mixture was cooled and quenched with saturated aq. NH 4 CI (50ml) and extracted with dichloromethane (100ml x 3). The combined organic layers were washed with H 2 0, brine, dried over anhydrous Na 2 S0 4 , filtered and evaporated to give the crude product.
  • the crude product was purified with column chromatography (eluted with petroleum ether and ethyl acetate from 4/1 to 1/2) to give the title compound as a light yellow solid (500mg).
  • the reaction mixture was heated 100°C for 1 hr using a microwave then concentrated under reduced pressure then diluted with IPA ( ⁇ 1 ml_) and neutralised using 2M HCI (aq).
  • the neutralised reaction mixture was loaded onto an SCX cartridge, washed five times with IPA then product eluted with a solution of 10% ammonia in IPA. Ammonia washes were combined and concentrated under reduced pressure to afford the crude product as an orange oil.
  • the oil dissolved in DMSO then purified by MDAP (Method E) and the resulting product was dissolved in methanol and passed down a thiol SPE cartridge, eluting the product with methanol. The appropriate fractions were combined and concentrated under reduced pressure to afford the title compound as a white solid, 35mg (29%).
  • 2-(3-Nitrophenyl)ethanamine hydrochloride salt (495 mg, 2.44 mmol) was dissolved in MeOH (25 mL) and then loaded onto an aminopropyl column that had been prewashed with MeOH. The column was then eluted with MeOH. The appropriate fractions were combined and evaporated under reduced pressure to give a colourless oil. The oil was dissolved in DMSO (5 mL) and added in a single portion to a microwave vial containing ethyl A/-[6-chloro-2-(2-pyridinyl)-4-pyrimidinyl]-p-alaninate (500 mg, 1 .63 mmol) at r.t. under N 2 .
  • HATU (53.3 mg, 0.14 mmol) and DIPEA (0.05 mL, 0.26 mmol) were added to a solution of 3- ⁇ [6- ⁇ [3- (ethyloxy)-3-oxopropyl]amino ⁇ -2-(2-pyridinyl)-4-pyrimidinyl]amino ⁇ propanoic acid (42.0 mg, 0.12 mmol) in A/,A/-dimethylformamide (5 mL). After 30 min of stirring N 1 -methylglycinamide hydrochloride (17.5 mg, 0.14 mmol) was added to the reaction mixture which was then stirred for 3 hr. The reaction mixture was concentrated under reduced pressure and the residue purified by column chromatography, eluting with 0-14% methanolic ammonia (2M) - DCM to give the title compound as a colourless oil, 31 .6 mg (63%).
  • Phenylmethanol (0.22 mL, 2.10 mmol) was added to a cold solution of triphenylphosphane (551 mg, 2.10 mmol), bis(l -methylethyl) -1 ,2-diazenedicarboxylate (0.41 mL, 2.10 mmol) and 6-chloro-2-(2- pyridinyl)-4(1 H)-pyrimidinone (218 mg, 1 .05 mmol) in THF (5 mL) stirred under nitrogen at 0 °C. After 2 hr of stirring at r.t., the reaction mixture was concentrated under reduced pressure.
  • the crude material was purified by column chromatography, eluting with 0-50% EtOAc-cyclohexane.
  • a second column chromatography purification step was undertaken on the resulting material, eluting with 0-30% EtOAc-cyclohexane.
  • a third column chromatography step was undertaken on the resulting material, eluting with 5-30% EtOAc-cyclohexane.
  • the impure product was further purified by column chromatography, eluting with 0-50% diethyl ether-cyclohexane to give the title compound as a yellow solid, 270 mg (86%).
  • Tetrakis(triphenylphosphine)palladium (109.0 mg, 0.09 mmol) was added to a suspension of 1 - benzothien-2-ylboronic acid (201 .0 mg, 1 .13 mmol), 4,6-dichloro-2-(2-pyridinyl)pyrimidine (213.0 mg, 0.94 mmol) and Cs 2 C0 3 (614.0 mg, 1 .88 mmol) in 1 ,4-dioxane (3 mL) and water (0.75 mL). The mixture was degassed with nitrogen for 30 min and heated to 100 °C in a Biotage initiator microwave for 0.5 hr.
  • the reaction mixture was concentrated under reduced pressure and the product purified by column chromatography, eluting with 0-40% cyclohexane-EtOAc.
  • the resulting material was dissolved with DCM (10 mL), then thiourea silica (1 .71 g) was added. After 1 hr of stirring at r.t., the silica was filtered off using a hydrophobic frit, under vacuum. The filtrate was concentrated under reduced pressure to give the title compound as a colourless oil, 167.0 mg (55%).
  • DIPEA 0.1 1 ml_, 0.64 mmol
  • 2,3,4, 5-tetrahydro-1 H-3-benzazepine 31 .5 mg, 0.21 mmol
  • DMSO 0.5 ml_
  • the system was heated to 160 °C in a microwave for 2 hr.
  • the reaction mixture was concentrated under reduced pressure.
  • the crude material was purified by column chromatography, eluting with 0-10% DCM-methanol.
  • 1 ,1 -dimethylethyl ⁇ -alaninate hydrochloride (1 .14 g, 6.25 mmol) was dissolved in MeOH (5 ml_) and then loaded onto an aminopropyl SPE column (20 g), previously prewashed with MeOH. The column was eluted with MeOH. The appropriate fractions were combined and evaporated under reduced pressure to give 1 ,1 -dimethylethyl ⁇ -alaninate (916.0 mg) as a white oil.
  • the reaction mixture was cooled and quenched by the addition of NH 4 CI solution (20 mL) and water (20 mL).
  • the organics were extracted with Et 2 0 (30 mL) and the layers separated.
  • the aqueous layer was further extracted with Et 2 0 (2 x 20 mL) and the combined organics dried (Na 2 S0 4 ) and concentrated in vacuo to afford the crude product as a yellow oil.
  • the crude product was further purified by silica chromatography, eluting with 0% to 100% ethyl acetate/cyclohexane to give the title compound (151 mg, 41 % yield).
  • the reaction mixture was then diluted with EtOAc (10 mL) and concentrated in vacuo to give the crude product as a yellow oil.
  • the crude product was purified by column chromatography using a gradient of 0% to 40% of a 20% solution of MeOH in DCM/DCM to give the title product as a yellow oil, 219 mg (60%).
  • the aqueous layer was extracted with further DCM (2 x 20 mL) and the combined organics washed sequentially with NaHC0 3 solution (20 mL) and then brine (20 mL). The organics were dried over Na 2 S0 4 then concentrated in vacuo.
  • the crude product was re-dissolved in DCM (2 mL) and a small aliquot ( ⁇ 0.1 mL) removed and dried under a stream of N 2 to allow an NMR to be taken. The remainder of the solution was concentrated to afford the crude title compound as a yellow-green oil, 43 mg, (92%) which was used directly in the subsequent reactions.
  • the reaction was concentrated in vacuo to afford the crude product as a yellow oil.
  • the crude product was purified by column chromatography using a gradient of 0 to 100% of a 20% solution of 2M NH 3 /MeOH in DCM)/DCM to give the title product as a yellow oil, 58 mg (69%).
  • the reaction mixture was then diluted with EtOAc (10ml_) and concentrated in vacuo to give the crude product as a yellow oil.
  • the crude product was purified by silica chromatography using a gradient of 0 to 40% of a 20% solution of MeOH in DCM/DCM to give the impure product as a yellow oil.
  • the product was further purified in two batches by high pH MDAP (Method E).
  • the reaction mixture was then diluted with EtOAc (10 ml_) and concentrated in vacuo to give the crude product as a yellow oil.
  • the crude product was purified by silica chromatography using a gradient of 0 to 40% of a 20% solution of MeOH in DCM/DCM to give the impure product as a yellow oil. Further purification was attempted using 0 to 100% of a 10% solution of MeOH in EtOAc/EtOAc. The same column was then eluted with 20 to 100% of a 20% solution of MeOH in DCM/DCM and finally the same column was eluted with 100% of a 20% solution of 2M NH 3 in MeOH/DCM. All of the fractions containing UV active material were concentrated together and the resultant crude material ( ⁇ 60mg) further purified by high pH MDAP (Method E).
  • the aqueous layer was extracted with further DCM (2 x 20 ml_) and the combined organics washed sequentially with NaHC0 3 solution (20 ml_) and then brine (20 ml_). The organics were dried over Na 2 S0 4 then concentrated in vacuo.
  • the crude product was re-dissolved in DCM (2 ml_) and a small aliquot ( ⁇ 0.1 ml_) removed and dried under a stream of N 2 to allow an NMR to be taken. The remainder of the solution was concentrated to afford the crude title product as a yellow-green oil, 87 mg (100 %) which was used directly in the next step without further purification.
  • the reaction was quenched by the addition of water (20 ml_) and DCM (20 ml_). The layers were separated and the aqueous layer further extracted with DCM (2 x 20 ml_). The combined organics were concentrated in vacuo to afford the crude product as a yellow oil.
  • the crude product was taken up in Ethanol (1 ml_), HCI (1 .0M in Et 2 0) (0.3 ml_, 0.300 mmol) was added and the reaction mixture allowed to stand for 1 h. The product was concentrated in vacuo to afford the crude product as a yellow oil.
  • the crude product was purified by silica chromatography using a gradient of 0 to 100% of a 20% solution of 2M NH3 in MeOH/DCM to give the product as a yellow oil which still contained a ⁇ 10% impurity.
  • the sample was further purified by high pH MDAP (Method E) to give the title product as a colourless gum (12 mg, 27%)
  • the crude product was purified by silica chromatography using a gradient of 0 to 100% solution of 20% 2M NH3/MeOH in DCM/DCM to give the impure product as a yellow oil.
  • the product was further purified by high pH MDAP (Method E). The appropriate fractions were combined together and concentrated in vacuo to afford the title product as a yellow gum, 19 mg (34%).
  • the crude product was purified by column chromatography on silica, eluting with 0 to 5% of a 20% solution of 2M ammonia in methanol in DCM/DCM.
  • the title compound was afforded as a yellow oil, 77 mg (49 %).
  • the reaction mixture was heated to 150°C in a microwave for 3 hr.
  • the sample was diluted with 1 :1 MeOH:DMSO and purified by MDAP (Method E) in 3 injections to give the title compound as a clear solid (58 mg, 0.130 mmol, 28 %).
  • Example 1 Af-r6-(1 ,1-dimethylethyl)-2-(2-pyridinyl)-4-pyrimidinvn-
  • Methyl N-[2-chloro-6-(trifluoromethyl)-4-pyrimidinyl]-p-alaninate 50 mg, 0.176 mmol was dissolved in toluene (0.5 ml_) and the system degassed for 1 min.
  • Bromo(2-pyridinyl)zinc 0.5M in THF
  • Palladium (II) acetate 5.94 mg, 0.026 mmol
  • S-Phos 21 .71 mg, 0.053 mmol
  • LiOH (2.6g, 0.109mmol) was dissolved in 200ml 1 :1 :1 THF/MeOH/water then a 2ml aliquot of this solution added to a vial containing ethyl A/-[6-chloro-2-(2-pyridinyl)-4-pyrimidinyl]-p-alaninate
  • Example 60 A -r6-(4-acetyl-1 -piperazinyl)-2-(2-pyridinyl)-4-pyrimidinyl1-B-alanine
  • Acetyl chloride (0.02 mL, 0.21 mmol) was added to a suspension of ethyl A/-[6-(1 -piperazinyl)-2-(2- pyridinyl)-4-pyrimidinyl]-p-alaninate (67 mg, 0.19 mmol) in DCM (3 mL).
  • the reaction mixture was stirred at r.t. for 18 hr then washed with water (3 mL) and the layers were separated using a hydrophobic frit.
  • the aqueous layer was loaded on a 5 g SCX cartridge that had been preconditioned with methanol, washed with methanol and product was eluted with methanolic ammonia (2M).
  • Example R1 Yield % LCMS
  • Example R1 Yield % LCMS
  • Methanesulfonyl chloride (0.02 mL, 0.21 mmol) was added to a suspension of ethyl ⁇ /-[6-(1- piperazinyl)-2-(2-pyridinyl)-4-pyrimidinyl]-p-alaninate (67 mg, 0.188 mmol) in DCM (3 mL).
  • the reaction mixture was stirred at r.t. for 18 hr then washed with water (3 mL) and the layers were separated using a hydrophobic frit.
  • the aqueous layer was loaded onto a 5 g SCX cartridge that had been conditioned with methanol, washed with methanol and product was eluted with methanolic ammonia (2M).
  • ⁇ -alanine (66.1 mg, 0.74 mmol) and DIPEA (0.26 mL, 1 .49 mmol) were added to a solution of 4- chloro-6-[(phenylmethyl)oxy]-2-(2-pyridinyl)pyrimidine (221 .0 mg, 0.74 mmol) in DMSO (0.5 mL) in a microwave vial.
  • the reaction mixture was heated to 150 °C in a microwave for 2 hr.
  • the reaction mixture was diluted with MeOH (3 mL) and purified by MDAP (Method E). The appropriate fractions were combined and evaporated under reduced pressure to give the impure product.
  • the reaction mixture was diluted with 1 :1 MeOH:DMSO (2 mL) and purified by MDAP (Method E). The solvent was evaporated under reduced pressure to give the impure product which was dissolved in 1 :1 MeOH:DMSO (1 mL) and purified by MDAP (Method G). The solvent was evaporated under reduced pressure to give a solid which was taken up in ethanol, loaded on a 2 g SCX column, washed with ethanol and eluted with 20% ammonium hydroxide in ethanol. The appropriate fractions were combined and evaporated under reduced pressure to give the title compound as a yellow solid, 28 mg (38 %).
  • Example R1 Yield % LCMS
  • Example R1 Yield % LCMS
  • Example 100 ⁇ - ⁇ 6-(1 -benzot ien-3-yl)-2-(2-pyridinyl)-4-pyrimidinyl1-B-alanine
  • Example 101 Af-(6-(1 ,3-dihvdro-2H-isoindol-2-yl)-2-f4-r(phenylamino)methvn-2-pyridinyl>-4- pyrimidinvD-B-alanine
  • Lithium hydroxide monohydrate (17.3 mg, 0.41 mmol) was added to a solution of 2-( ⁇ [2-( ⁇ 2-[4-[(2- carboxyethyl)amino]-6-(1 ,2,4,5-tetrahydro-3/-/-3-benzazepin-3-yl)-2-pyrimidinyl]-4- pyridinyl ⁇ oxy)ethyl]amino ⁇ carbonyl)benzoic acid (250.0 mg, 0.41 mmol) in 1 :3 water/THF solution (4 ml). After 40 hr of stirring at r.t. (the solvent evaporated during that time), the residue was then taken up in water (5 ml) then acidified with HCI (2M) until pH 2. The solid was filtered off, washed with water and dried in a vacuum oven for 5 hr to give a yellow solid, 50 mg.
  • the present application does not include an Example 27, an Example 42, an Example 48, an Example 58, an Example 59, an Example 69, an Example 70, an Example 71 , an Example 72, an Example 75, an Example 84, an Example 105, an Example 108, or an Example 1 16.
  • JMJD3 MALDI-TOF Demethylase Assay Protocol monitors demethylation of a histone H3 peptide containing tri-methylated K27, by recombinant Jumonji D3 demethylase enzyme.

Landscapes

  • Organic Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

A method of treating autoimmune diseases or conditions in a mammal, such as a human, which comprises the administration of a therapeutically effective amount of histone demethylase enzymes of formula (I).

Description

N-2- (2 - PYRIDINYL) - 4 - PYRIMIDINYL - BETA -ALANINE DERIVATIVES AS INHIBITORS OF HISTONE DEMETHYLASE JMJD3
Field of the Invention
The present invention relates to compounds, compositions, combinations and medicaments containing said compounds and processes for their preparation. The invention also relates to the use of said compounds, combinations, compositions and medicaments, for example as inhibitors of the activity of the histone demethylase JMJD3, to modify the epigenetic status of cells and/or the treatment of diseases and conditions mediated by JMJD3, in particular cancer, inflammation and autoimmune diseases. Background of the Invention
The structure of chromatin is complex and dynamic and has a major effect on gene transcription. Chromatin is the complex combination of DNA and protein that makes up chromosomes. It is found inside the nuclei of eukaryotic cells and is divided between heterochromatin (condensed) and euchromatin (extended) forms. The major components of chromatin are DNA and proteins, including histones. The basic building blocks of chromatin are nucleosomes, each of which is composed of 146 base pairs of DNA wrapped around a histone octamer consisting of 2 copies of each H2A, H2B, H3 and H4. The functions of chromatin are to package DNA into a smaller volume to fit in the cell, to strengthen the DNA to allow mitosis and meiosis, and to serve as a mechanism to control expression and DNA replication. Chromatin contains genetic material serving as instructions to direct cell functions. Changes in chromatin structure are regulated by modifications on histone and DNA methylation. Epigenetic mechanisms do not change the DNA sequence but allow the genes to be expressed differently. Epigenetics modification include numerous mechanisms including DNA methylation and post-translational modification of N-terminal tails of histone proteins such as methylation, acetylation, phosphorylation and ubiquitination.
In recent years many of the target proteins involved in epigenetic control have been identified. These targets can be classified into families that read, write and erase covalent modification (e.g. methylation, acetylation) in histones. Histone methylation is an abundant epigenetic modification of core histones found in eukaryotic organisms that has been linked to a number of cellular processes including DNA repair, cell cycle progression, cell differentiation and regulation of gene expression. This modification is catalysed by the specific histone methyltransferases (HMTs), lysine methyl transferase and arginine methyl transferase, which introduce methyl groups at lysine (K) or arginine (R) residues respectively. Thus far, histone methylation has been found to occur at six major sites, including histone H3 lysine 4 (H3K4), H3K9, H3K27, H3K36, H3K79 and H4K20. Unlike other modifications, the same lysine residue can be methylated to different degrees to include mono-, di- or trimethyl moieties, which may have different functional consequences. In general, lysine methylation at H3K4, H3K36 and H3K39 is associated with regions of transcriptionally active chromatin, whereas methylation at H3K9, H3K27 and H4K20 is associated with transcriptionally silenced regions (Martin C. and Zhang Y., Nature Rev. Mol. Cell Biol. 2005, 6, 838-849). H3K9 promoter methylation is considered a repressive mark for euchromatic genes (Nielsen et al., Nature 2001 ,412, 561 -565; Shi et al., Nature 2003, 422, 735-738) and is also one of the landmark modifications associated with heterochromatin (Nakayama et al., Science 2001 , 292, 1 10-1 13) but some studies have also identified association of H3K9 trimethylation (H3K9me3) with actively transcribed genes (Vakoc C. et al, Mol. Cell 2005, 19. 381 -391).
Unlike other histone modifications such as acetylation, methylation used to be regarded as a permanent/irreversible modification. However, with the identification of histone demethylases, this process has been shown to be reversible and dynamically controlled. Histone methylation appears thus to be regulated by a complex network that involves a large number of site-specific methylases, demethylases and methyl recognition proteins, which play an important role in controlling the expression of genetic information through transcriptional changes and chromatin structure alterations. Since levels of lysine methylation are known to change during processes such as transcriptional regulation, it was proposed that specific enzymatic activity might remove the methyl groups (Bannister et al., 2002 Cell 109, 801 -806). Recent work has confirmed the existence of enzymatic demethylation and two separate mechanisms of lysine demethylation have been demonstrated: amine oxidation by Lysine Specific Demethylase 1 (LSD1) and hydroxylation by JmjC-domain containing proteins, which indicate these proteins as being novel histone modifying enzymes that can remove methyl groups on lysines (Shi et al., 2004; Cell 1 19, 941 -953; Tsukada et al., 2006, Nature 439, 81 1 -816).
The Jumonji protein is the founding member of a group of proteins characterised by a novel structural motif, the JmjC domain. This is an extensive group of demethylase enzymes which can be defined into several families according to sequence similarity within the JmjC domain and the presence of other domains in the full length protein. The JmjC domain of several members of this family has been shown to possess lysine demethylation activity, which is dependent on iron (Fe (II)) and a- ketoglutarate as co-factors (Klose RJ et al, Nat Rev Genet. 2006 Sep; 7(9); 715-27). Unlike LSD1 , which can only remove mono- and dimethyl lysine modifications, the JmjC-domain-containing histone demethylases (JHDMs) can remove all three histone lysine-methylation states.
Some JmjC domain-containing proteins, including the histone demethylase JMJD3, have been implicated in tumorogenesis and thus have identified histone demethylases as targets of research for anti-cancer therapies (see for example WO2009/1 1401 1 and WO2010/043566). JmjD3 (KDM6B) is one of the approximately 30 JmjC family members found in humans, and functions as a specific demethylase of lysine 27 of histone H3 (H3K27). JmjD3 can demethylate both the tri- and dimethylated H3K27-repressive histone marks, thereby facilitating gene transcription. This was first demonstrated in C. Elegans embryogenesis, where JmjD3 was shown to regulate gonadal development through modulation of HOX gene expression (Agger K et al, Nature 2007 Oct; 449(7163); 731 -734). Further studies have placed JmjD3 at key cell fate decision checkpoints in T lymphocytes (Miller SA et al, Genes Dev. 2008 Oct; 22; 2280-2993) and macrophages (Ishii M et al, Blood 2009 Oct; 1 14(15); 3244-3254). In addition, JmjD3 has been demonstrated to regulate the differentiation state of the epidermis (Sen GL et al, Genes Dev. 2008 Jul; 22; 1865-1870) and to activate the tumour suppressor, INK4A-Arf, in response stress induced signals (Agger K et al, Genes Dev. 2009 Apr; 23; 1 171 -1 176). JmjD3 also appears to be involved in more acute, externally-driven, inflammatory processes. In macrophages, for example, JmjD3 is rapidly induced through an NF-kB- dependent mechanism in response to bacterial products and inflammatory stimuli (De Santa F et al, Cell 2007 Sept; 130; 1083-1094). Moreover, depletion experiments in these cells have demonstrated that JmjD3 participates directly in the inflammatory transcriptional response, although it remains unclear whether this is achieved through demethylation of H3K27me3 at target gene promoters (De Santa F et al, EMBO J. 2009 Sept; 28; 3341 -3352).
Summary of the Invention
In one aspect there is provided a pharmaceutical composition comprising a compound of the formula
(I)
Figure imgf000005_0001
(I)
wherein
R' is:
• C1-6 alkyl;
• C3_7 cycloalkyl;
• C1-6 haloalkyl;
• a 5, 6 or 7-membered aryl or heteroaryl (which heteroaryl contains one or more heteroatoms selected from N, O and S and which is optionally fused to phenyl), said 5, 6 or 7- membered aryl or heteroaryl being optionally substituted with one or more substituents independently selected from C1-3alkyl;
• 0-C1-6alkyl (which is optionally substituted by phenyl or naphthyl, each of which may be substituted by one of more substituents independently selected from halo);
• -O-cyclohexyl (which is optionally fused with phenyl);
C(0)NRc 2;
or
NRaRb, each Ra and Rb is independently selected from:
H;
• C^alkyl which is optionally substituted by one or more substituents independently selected from phenyl (which phenyl is optionally substituted by one or more substituents independently selected from C1-3alkyl, 0-C1-3alkyl, C(0)NRc 2, halo and cyano), C(0)NRc 2, a 4, 5, 6 or 7-membered heterocyclic or heteroaryl group (containing one or more heteroatoms independently selected from N, O and, S), a 3, 4, 5, 6 or 7-membered cycloalkyl group (which is optionally fused to phenyl), halo, OC1-3alkyl, OH, -NHCOC^alkylNR^ and C(0)NHCH2C(0)NRc 2;
• a 3, 4, 5, 6 or 7-membered cycloalkyl group (which is optionally fused to phenyl), or
• Ra and Rb together form a 5, 6 or 7-membered heterocyclic group optionally containing one or more further heteroatoms independently selected from N, O, S or S(0)2 said heterocyclic group being optionally fused to a 5, 6 or 7-membered aryl or heteroaryl ring containing one or more heteroatoms independently selected from N, O and S; the heterocylic ring and/or the aryl or heteroaryl to which it is optionally fused being optionally substituted by one or more substituents independently selected from halo, OH, C1-3alkyl, 0-C1-3alkyl, C(0)C1-3alkyl, S(0)2C1-3alkyl, NHC(0)C1-3alkyl, NHS(0)2C1-3alkyl, C(0)NRc 2, C(0)NRd 2 (wherein Rd and Rd together form a 5 or 6-membered heterocylic ring), NRC 2 C(0)phenyl, S(0)2NRc 2, =0 (oxo) and 5, 6 or 7-membered aryl or heteroaryl (containing one or more heteroatoms independently selected from N, O and S);
R2 and R3 are each independently selected from:
H,
(CH^NR^CH^NR^,
(CHz eNR^;
• C1-3 alkyl;
• 0-C1-3alkyl;
• C1-3haloalkyl;
• (CH2)0-3NRaRb (wherein Ra and Rb are as defined above);
(CH2)0-3NHPh;
(CH2)0-3OPh;
(CH2)0.3Ph;
or R2 and R3 together form a fused phenyl ring, and each Rc is independently selected from hydrogen and C1-3alkyl or a pharmaceutically acceptable salt thereof and one or more of pharmaceutically acceptable carriers, diluents and excipients.
In another aspect of the present invention, there is provided a compound of the formula (I) or a pharmaceutically acceptable salt thereof as defined above with the proviso R is not -C(CH3)3.
In a further aspect of the present invention, there is provided a pharmaceutical composition comprising a compound of formula (la):
Figure imgf000007_0001
wherein
R is C1-6 alkyl, C3.7cycloalkyl, C1-6 haloalkyl, NRaRb ; each Ra and Rb is independently selected from H, C^alkyl ( which alkyl is optionally substituted by one or more substituents independently selected from phenyl, halo, OH) or Ra and Rb together form a 4, 5 or 6 membered heterocyclic group optionally containing one or more further heteroatoms selected from N or O, said heterocyclic group being optionally fused to a phenyl ring. or a pharmaceutically acceptable salt thereof and one or more of pharmaceutically acceptable carriers, diluents and excipients
In a further aspect of the present invention, there is provided a compound of formula (lb),
Figure imgf000007_0002
(lb) wherein
R is C1-6 alkyl, C3.7cycloalkyl, C^haloalkyl, NRaRb each Ra and Rb is independently selected from H, C^alkyl (which alkyl is optionally substituted by one or more substituents independently selected from phenyl, halo, OH) or Ra and Rb together form a ,a 4,5 or 6 membered heterocyclic group optionally containing one or more further heteroatoms selected from N or O, said heterocyclic group being optionally fused to a phenyl ring; with the proviso R is not -C(CH3)3. or a pharmaceutically acceptable salt thereof.
In a further aspect of the present invention, there is provided a compound of formula (I) or of formula (la) or of formula (lb), or a pharmaceutically acceptable salt thereof for use in therapy, in particular in changing the epigenetic status of cells, treating cancer, inflammation or autoimmune diseases.
In a further aspect of the present invention, there is provided a method of changing the epigenetic status of cells, treating cancer, inflammation or autoimmune diseases in a subject comprising administering a therapeutically effective amount of a compound of formula (I) or of formula (la) or of formula (lb) or a pharmaceutically acceptable salt thereof.
In a further aspect of the present invention, there is provided the use of a compound of formula (I) or of formula (la) or of formula (lb), or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for use in changing the epigenetic status of cells, treating cancer, inflammation or autoimmune diseases
In a further aspect there is provided a combination comprising a compound of formula (I) or of formula (la) or of formula (lb), or a pharmaceutically acceptable salt thereof and at least one further therapeutic agent.
In a further aspect there is provided a combination comprising a compound of formula (I) or of formula (la) or of formula (lb), or a pharmaceutically acceptable salt thereof and at least one further therapeutic agent for use in therapy, particularly for changing the epigenetic status of cells and treating cancer, inflammation or autoimmune diseases.
In a further aspect of the invention there is provided a compound of formula (I) or of formula (la) or of formula (lb), or a pharmaceutically acceptable salt thereof for use in the treatment of cancer. In a further aspect there is provided a method of treating cancer comprising administering to a human in need thereof a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof. In a further aspect there is provided the use of a compound of formula (I) or of formula (la) or of formula (lb), or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for the treatment of cancer.
In a further aspect there is provided a combination comprising a compound of formula (I) or of formula (la) or of formula (lb), or a pharmaceutically acceptable salt thereof and at least one anti-neoplastic agent.
In a further aspect there is provided a combination comprising a compound of formula (I) or of formula (la) or of formula (lb), or a pharmaceutically acceptable salt thereof and at least one anti-neoplastic agent, for use in therapy.
In a further aspect there is provided a combination comprising a compound of formula (I) or of formula (la) or of formula (lb), or pharmaceutically acceptable salt thereof and at least one anti-neoplastic agent, for use in treating cancer.
In a further aspect there is provided the use of a combination comprising a compound of formula (I) or of formula (la) or of formula (lb), or a pharmaceutically acceptable salt thereof and at least one antineoplastic agent, in the manufacture of a medicament for the treatment of cancer. In a further aspect there is provided a method of treating cancer, comprising administering to a human in need thereof a therapeutically effective amount of a combination comprising a compound of formula (I) or of formula (la) or of formula (lb), or a pharmaceutically acceptable salt thereof and at least one anti-neoplastic agent. In a further aspect there is provided a pharmaceutical composition comprising a combination comprising a compound of formula (I) or of formula (la) or of formula (lb), or a pharmaceutically acceptable salt thereof and at least one further therapeutic agent, particularly at least one antineoplastic agent and one or more of pharmaceutically acceptable carriers, diluents and excipients. In a further aspect there is provided a compound of formula (I) or of formula (la) or of formula (lb), or a pharmaceutically acceptable salt thereof for use in the treatment of inflammation or autoimmune diseases.
In a further aspect there is provided a method of treating inflammation or autoimmune diseases comprising administering to human in need thereof, a therapeutically effect amount of a compound of formula (I) or of formula (la) or of formula (lb), or a pharmaceutically acceptable salt thereof. In a further aspect there is provided the use of a compound of formula (I) or of formula (la) or of formula (lb), or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for the treatment of inflammation or autoimmune diseases.
Detailed Description of the Invention
Studies have identified JmjC domain-containing proteins as histone demethylases that mediate the reversal of methylation at histone H3K4, H3K9 and H3K36. The JmjD3 family of proteins is a subfamily of JmjC domain-containing proteins that have been shown to demethyate H3K27. Such inhibitors are therefore useful in changing the epigenetic status of cells resulting in inhibiting or activating chromatin remodelling by modifying histone methylation and thus in treating disorders associated with such modified histone methylation including cancer and other conditions associated with undesirable cell proliferation, autoimmune and inflammatory diseases or conditions and psychiatric disorders including depression.
As used herein, "a compound of formula (I)" or "a compound or formula (la)" or "a compound or formula (lb)" includes all solvates, complexes, polymorphs, radiolabeled derivatives (including deuterated derivatives where one or more H are replaced by D), stereoisomers and optical isomers of the compounds of formula (I) or (la) or (lb) and salts thereof. As compounds of the formula (la) and compounds of the formula (lb) are subsets of the compounds of formula (I), references herein to "compounds of the formula (I)" typically also apply to compounds of the formula (la) and (lb) and are to be understood as such unless it is indicated otherwise.
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.
The compounds of formula (I) may exist in solid or liquid form. In solid form, compound of the invention may exist in a continuum of solid states ranging from fully amorphous to fully crystalline. The term 'amorphous' refers to a state in which the material lacks long range order at the molecular level and, depending upon the temperature, may exhibit the physical properties of a solid or a liquid. Typically such materials do not give distinctive X-ray diffraction patterns and, while exhibiting the properties of a solid, are more formally described as a liquid. Upon heating, a change from solid to liquid properties occurs which is characterized by a change of state, typically second order ('glass transition'). The term 'crystalline' refers to a solid phase in which the material has a regular ordered internal structure at the molecular level and gives a distinctive X-ray diffraction pattern with defined peaks. Such materials when heated sufficiently will also exhibit the properties of a liquid, but the change from solid to liquid is characterized by a phase change, typically first order ('melting point').
The compound of formula (I) may exist in solvated and unsolvated forms. As used herein, the term "solvate" refers to a complex of variable stoichiometry formed by a solute (in this invention, a compound of formula (I) or a salt) and a solvent. Such solvents for the purpose of the invention may not interfere with the biological activity of the solute. The skilled artisan will appreciate that pharmaceutically acceptable solvates may be formed for crystalline compounds wherein solvent molecules are incorporated into the crystalline lattice during crystallization. The incorporated solvent molecules may be water molecules or non-aqueous such as ethanol, isopropanol, DMSO, acetic acid, ethanolamine, and ethyl acetate molecules. Crystalline lattice incorporated with water molecules are typically referred to as "hydrates". Hydrates include stoichiometric hydrates as well as compositions containing variable amounts of water. The present invention includes all such solvates. The compounds of formula (I) may have the ability to crystallize in more than one form, a characteristic, which is known as polymorphism, and it is understood that such polymorphic forms ("polymorphs") are within the scope of the invention. Polymorphism generally can occur as a response to changes in temperature or pressure or both and can also result from variations in the crystallization process. Polymorphs can be distinguished by various physical characteristics known in the art such as x-ray diffraction patterns, solubility and melting point.
It is also noted that the compounds of formula (I) may form tautomers. It is understood that all tautomers and mixtures of tautomers of the compounds of the present invention are included within the scope of the compounds of the present invention.
As used herein, the term "histone demethylase inhibitor", or "inhibitor" refers to any compound or treatment capable of inhibiting or reducing the expression or activity of a histone demethylase. The inhibitor is preferably selective against one or more histone demethylase enzymes with no direct activity as any other histone modifying enzymes.
As used herein, the term "alkyl" and "alkylene" refers to a saturated hydrocarbon chain having the specified number of member atoms. C^alkyl refers to an alkyl group having from 1 to 6 member atoms, for example 1 to 4 member atoms. Alkyl groups may be straight or branched. Representative branched alkyl groups have one, two, or three branches. Alkyl includes methyl, ethyl, propyl (n-propyl and isopropyl), butyl (n-butyl, isobutyl, and t-butyl), pentyl (n-pentyl, isopentyl, and neopentyl), and hexyl.
As used herein, the term "halo" refers to the halogen radical fluoro, chloro, bromo, or iodo.
As used herein, the terms "C^Ce haloalkyl", "C^haloalkyl" and the like refer to a straight or branched chain alkyl group as defined above containing at least 1 , and at most 6 carbon atoms respectively substituted with at least one halo group, halo being as defined herein. Examples of such branched or straight chained haloalkyl groups useful in the present invention include, but are not limited to, methyl, ethyl, propyl, isopropyl, isobutyl and n-butyl substituted independently with one or more halos, e.g., fluoro, chloro, bromo and iodo. As used herein, the term "heterocyclic group" refers to a non-aromatic ring having the specified number of member atoms being saturated or having one or more degrees of unsaturation and, unless otherwise specified, containing one or more heteroatoms selected from N, O or S.
As used herein, the term "aryl" refers to an aromatic ring having the specified number of member atoms. Bicyclic and other polycyclic ring systems containing an aryl are described as fused systems.
As used herein, the term "heteroaryl" refers to an aromatic ring having the specified number of member atoms and, unless otherwise specified, containing one or more heteroatoms selected from N, O or S. Bicyclic and other polycyclic ring systems comprising a heteroaryl ring are described as fused systems.
As used herein, the term "member atoms" refers to the atom or atoms that form a chain or ring. Where more than one member atom is present in a chain and within a ring, each member atom is covalently bound to an adjacent member atom in the chain or ring. Atoms that make up a substituent group on a chain or ring are not member atoms in the chain or ring.
As used herein, the term "optionally substituted" indicates that a group may be unsubstituted or substituted with one or more substituents as defined herein.
As used herein, the term "substituted" in reference to a group indicates that a hydrogen atom attached to a member atom within a group is replaced. It should be understood that the term "substituted" includes the implicit provision that such substitution be in accordance with the permitted valence of the substituted atom and the substituent and that the substitution results in a stable compound (i.e. one that does not spontaneously undergo transformation such as by rearrangement, cyclization, or elimination). In certain embodiments, a single atom may be substituted with more than one substituent as long as such substitution is in accordance with the permitted valence of the atom. Suitable substituents are defined herein for each substituted or optionally substituted group.
As used herein, the term "pharmaceutically acceptable" refers to those compounds, materials, compositions, and dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
In one embodiment, R is:
• C1- alkyl;
· C3.7 cycloalkyl; • C1- fluoroalkyl;
• phenyl
• a 5-membered heteroaryl (which contains one or two heteroatoms selected from N, O and S and which is optionally fused to phenyl), said 5-membered heteroaryl being optionally substituted with one or more substituents independently selected from C1-3alkyl;
• O-C^alkyl (which is optionally substituted by phenyl (optionally substituted by one of more chloro) or napthyl);
• 0-C3_7 cycloalkyl (which is optionally fused with phenyl);
C(0)NRc 2;
or
NRaRb, either
• Ra is H or C1-4alkyl, especially H or methyl, and Rb is C^alkyl which is optionally substituted by one or more substituents independently selected from:
o phenyl (which is optionally substituted by one or more substiuents independently selected from methyl, O-methyl, C(0)NRc 2, chloro, bromo and cyano),
o a 5 or 6-membered heteroaryl group (containing one or more heteroatoms independently selected from N, O and, S),
o a 3, 4 or 5-membered cycloalkyl group,
o OH
Figure imgf000013_0001
o NHCO(CH2)2NRc 2and
o C(0)NHCH2C(0)NRc 2;
• Ra is H and Rb is a 3, 4 or 5-membered cycloalkyl group (which is optionally fused to phenyl), or
• Ra and Rb together form a 5, 6 or 7-membered heterocyclic group optionally containing a further heteroatom selected from N, O, S or S(0)2 said heterocyclic group being optionally fused to a phenyl ring or to a 5 or 6-membered heteroaryl ring (containing one or more heteroatoms independently selected from N, O and S); the heterocylic ring and/or the phenyl or heteroaryl to which it is optionally fused being optionally substituted by one or more substituents independently selected from:
o chloro or bromo,
o OH,
o C1-3alkyl,
o 0-C1-3alkyl,
o C(0)C1-3alkyl,
o S(0)2C1-3alkyl,
o NHC(0)C1-3alkyl, NHS(0)2C1-3alkyl, (wherein Rd and Rd together form a 5 or 6-membered heterocylic ring),
Figure imgf000014_0001
C(0)phenyl,
Figure imgf000014_0002
=0 (oxo),
phenyl and
5 or 6-membered heteroaryl (containing one or more heteroatoms independently selected from N, O and S);
each Rc is independently selected from hydrogen and methyl.
In another embodiment, R is:
• C1- alkyl;
• C3.7 cycloalkyl, especially cyclopropyl;
• trifluoromethyl;
• phenyl
• a 5-membered heteroaryl (which contains one or two heteroatoms selected from N, O and S and which is optionally fused to phenyl), said 5-membered heteroaryl being optionally substituted with one or two methyl substituents;
• O-C^alkyl (which is optionally substituted by phenyl);
• O-cyclohexyl (which is fused with phenyl);
C(0)NRc 2;
or
NRaRb, either
• Ra is H or methyl, and Rb is C^alkyl which is optionally substituted by one or more substituents independently selected from:
o phenyl (which is optionally substituted by one or more substiuents independently selected from methyl, O-methyl, C(0)NRc 2, chloro and cyano),
o a 5 or 6-membered heteroaryl group (containing one or two heteroatoms independently selected from N, O and, S),
o a 3 or 5-membered cycloalkyl group, especially cyclopropyl,
o OH,
o C(0)NRc 2,and o C(0)NHCH2C(0)NRc 2;
• Ra is H and Rb is a 5-membered cycloalkyl group (which is optionally fused to phenyl), or
• Ra and Rb together form a 5, 6 or 7-membered heterocyclic group optionally containing a further heteroatom selected from N, O, S or S(0)2 said heterocyclic group being optionally fused to a phenyl ring or to a 5 or 6-membered heteroaryl ring (containing one or more heteroatoms independently selected from N, O and S); the heterocylic ring and/or the phenyl or heteroaryl to which it is optionally fused being optionally substituted by one or more substituents independently selected from:
o chloro,
o OH,
o C1-3alkyl, especially methyl,
o 0-C1-3alkyl, especially 0-CH3,
o C(0)C1-3alkyl, especially C(0)CH3,
o S(0)2C1-3alkyl, especially S(0)2CH3,
o NHC(0)C1-3alkyl, especially NHC(0)CH3,
o NHS(0)2C1-3alkyl, especially NHS(0)2CH3i
Figure imgf000015_0001
o C(0)NRd 2 (wherein Rd and Rd together form a 5 or 6-membered heterocylic ring), o NRc 2i
o C(0)phenyl,
Figure imgf000015_0002
o =0 (oxo),
o phenyl and
o 5 or 6-membered heteroaryl (containing one or more heteroatoms independently selected from N, O and S);
and
NRC2 is selected from NHCH3 and NH2.
In one embodiment, R is:
• C1- alkyl;
• C3_7 cycloalkyl;
• C1- fluoroalkyl; or
NRaRb,
wherein
• Ra and R together form a 5, 6 or 7-membered heterocyclic group optionally containing a further heteroatom selected from N, O, S or S(0)2 said heterocyclic group being optionally fused to a phenyl ring or to a 5 or 6-membered heteroaryl ring (containing one or more heteroatoms independently selected from N, O and S); the heterocylic ring and/or the phenyl or heteroaryl to which it is optionally fused being optionally substituted by one or more substituents independently selected from:
o chloro or bromo,
o OH,
o C1-3alkyl,
o 0-C1-3alkyl,
o C(0)C1-3alkyl,
o S(0)2C1-3alkyl,
o NHC(0)C1-3alkyl,
o NHS(0)2C1-3alkyl,
Figure imgf000016_0001
o C(0)NRd 2 (wherein Rd and Rd together form a 5 or 6-membered heterocylic ring), o NRc 2i
o C(0)phenyl,
Figure imgf000016_0002
o =0 (oxo),
o phenyl and
o 5 or 6-membered heteroaryl (containing one or more heteroatoms independently selected from N, O and S),
and each Rc 2 is independently selected from H and CH3.
In a further embodiment, R is:
• C1- alkyl;
· C3_7 cycloalkyl;
• C1- fluoroalkyl; or
NRaRb,
wherein
Ra and Rb together form a 5, 6 or 7-membered heterocyclic group said heterocyclic group being optionally fused to a phenyl ring; the heterocylic ring and/or the phenyl to which it is optionally fused being optionally substituted by one or more substituents independently selected from: chloro, bromo, OH, C1-3alkyl, 0-C1-3alkyl, C(0)C1-3alkyl, and S(0)2C1-3alkyl.
In a yet further embodiment, R is:
· C1- alkyl;
NRaRb,
wherein
Ra and Rb together form a 5, 6 or 7-membered heterocyclic group said heterocyclic group being fused to a phenyl ring; the heterocylic ring and/or the phenyl to which it is fused being unsubstiuted or substituted by one or more substituents independently selected from: chloro, OH, methyl, O-methyl and S(0)2methyl, for example, unsubstituted.
In one embodiment in compounds of formula (I), R is selected from:
-C(CH)3, CF3, -NHCH3, -N(CH3)2, -NHCH2CH2OH, - N(CH3)CH2CH2OH
Figure imgf000017_0001
In one embodiment in compounds of formula (la) R is selected from:
CF3, -NHCH3, -N(CH3)2, -NHCH2CH2OH, - N(CH3)CH2CH2OH
Figure imgf000017_0002
In one embodiment in compounds of formula (lb) R is selected from:
CF3, -NHCH3, -N(CH3)2, -NHCH2CH2OH, - N(CH3)CH2CH2OH
Figure imgf000017_0003
In one aspect of the invention, in compounds of the formula (I), (la) and (lb), R2 and R3 are selected from H, (CH^!^NR^CH^!^NR^ and (CH2)0.6NRC 2. In a further aspect, one of R2 and R3 is H and the other is selected from (CH^NR' CI-y^NR0;, and (CH2)0.6NRC 2. In a yet further aspect of the invention, R2 and R3 are both H. For the avoidance of doubt, R2 and R3 may be as defined in this paragraph in all of the embodiments of the invention described above where different options for R are disclosed. In one aspect of the invention, in compounds of the formula (I), (la) and (lb) in the aspects and embodiments of the invention described above, each Rc is independently selected from hydrogen and methyl. In a further aspect of the invention NRC 2 is selected from NHCH3 and NH2. While aspects for each variable have generally been listed above separately for each variable this invention includes those compounds in which several or each aspect in formula (I) is selected from each of the aspects listed above. Therefore, this invention is intended to include all combinations of aspects for each variable. Specific examples of compounds of formula (I) and salts thereof include the compounds and salts of Examples 1 -125 disclosed herein.
Specific examples of compounds of formula (I) include the following:
Λ/-[6-(1 ,1 -dimethylethyl)-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine;
A/-[2-(2-pyridinyl)-6-(trifluoromethyl)-4-pyrimidinyl]-p-alanine;
A/-[6-(4-morpholinyl)-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine;
A/-[6-(methylamino)-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine;
A/-[2-(2-pyridinyl)-6-(1 -pyrrolidinyl)-4-pyrimidinyl]-p-alanine;
A/-[6-[(2-hydroxyethyl)amino]-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine;
A/-[6-[(phenylmethyl)amino]-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine;
A/-[6-[(2-hydroxyethyl)(methyl)amino]-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine;
A/-[6-(dimethylamino)-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine;
A/-[2-(2-pyridinyl)-6-(1 ,2,4,5-tetrahydro-3/-/-3-benzazepin-3-yl)-4-pyrimidinyl]-p-alanine. Further specific examples of compounds of formula (I) include the following:
A/-[6-phenyl-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine;
A/-[6-[3-(aminocarbonyl)-1 -piperidinyl]-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine;
A/-[6-[4-(aminocarbonyl)-1 -piperidinyl]-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine;
/V-[6-({[3,4-Jb/s(methyloxy)phenyl]methyl}amino)-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine;
A/-[6-[(3-amino-3-oxopropyl)(methyl)amino]-2-(2-pyridinyl)-4-pyrimidinyl]- β-alanine;
A/-/6-{[(3,4-dichlorophenyl)methyl]amino}-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine;
A/-[6-({[3-(aminocarbonyl)phenyl]methyl}amino)-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine;
A/-[6-{[(4-chlorophenyl)methyl]amino}-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine;
A/-[6-(7/-/-pyrazol-4-yl)-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine;
A/-[6-(3-methyl-1 /-/-pyrazol-4-yl)-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine;
A/-[6-{[(3-chlorophenyl)methyl]amino}-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine;
A/-[6-{[(2-methylphenyl)methyl]amino}-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine;
A/-{2-(2-pyridinyl)-6-[(2-thienylmethyl)amino]-4-pyrimidinyl}-p-alanine; A/-[6-({[3-(methyloxy)phenyl]methyl}amino)-2-(2-pyr^
A/-{2-(2-pyridinyl)-6-[(2-pyridinylmethyl)amino]^^^
A -[6-({[4-(methyloxy)phenyl]methyl}amino)-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanin
A/-[6-[(cyclopropylmethyl)amino]-2-(2^yridinyl)-4-pyrimidinyl]-p-alanine;
Λ/-[6-(1 ,3-dihydro-2H-isoindol-2-yl)-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine;
A/-[6-(1 -methylethyl)-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine;
A/-[6-cyclopropyl-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine;
A/-[6-[3-(diethylamino)-1 -piperidinyl]-2-(2-pyridm^^
Λ/-[6-[4-(1 ,3,4-oxadiazol-2-yl)-1 ^iperidinyl]-2-(2^yridinyl)-4-pyrimidinyl]-p-alanine;
A/-{2-(2-pyridinyl)-6-[4-(1 ,3-thiazol-2-yl)-1 -piperazinyl]-4-pyrimidinyl}-p-alanine;
A/-[6-(4-phenyl-1 -piperazinyl)-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine;
A -[6-{[(4-chlorophenyl)methyl]amino}-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine;
A/-[6-(hexahydro-1 H-azepin-1 -yl)-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine;
A/-[6-(1 -benzothien-2-yl)-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine;
A/-[6-(3,4-dihydro-2(1 /^-isoquinolinyl)-2-(2^yridinyl)-4^yrimidinyl]-p-alanine;
A/-[6-{4-[(methylamino)carbonyl]-1 -piperidinyl^
A/-[6-(7-hydroxy-1 ,2,4,5-tetrahydro-3H-3-benzazepin-3-yl)-2-(2^yridinyl)-4^yrimidinyl]-p-alanine; A/-[6-(7-bromo-1 ,2,4,5-tetrahydro-3H-3-benzazepin-3-yl)-2-(2^yridinyl)-4-pyrimidinyl]-p-alanine; A/-[6-(5-hydroxy-1 ,3-dihydro-2H-isoindol-2-yl)-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine.
Yet further specific examples of compounds of formula (I) include the following:
A/-{2-(2-pyridinyl)-6-[(4-pyridinylmethyl)amino]-4-pyrimidinyl}-p-alanine;
A/-{2-(2-pyridinyl)-6-[(3-pyridinylmethyl)amino]-4-pyrimidinyl}- β -alanine
A/-[6-(2,3-dihydro-1 /-/-inden-2-ylamino)-2-(2-pyridinyl)-4-pyrimidinyl]- β -alanine;
A/-[6-[(2-phenylethyl)amino]-2-(2-pyridinyl)-4-pyrimidinyl]- β -alanine;
A/-[6-[methyl(phenylmethyl)amino]-2-(2-pyridinyl)-4-pyrimidinyl]- β -alanine;
3-{[6-(1 ,1 -dimethylethyl)-2-(2-pyridinyl)-4-pyrimidinyl]amino}-2-methylpropanoic acid;
A/-[6-(methyloxy)-2-(2-pyridinyl)-4-pyrimidinyl]- β-alanine;
Λ/-[6-(1 ,1 -dimethylethyl)-2-(3-isoquinolinyl)-4-pyrimidinyl]- β-alanine;
Λ/-{6-(1 ,1 -dimethylethyl)-2-[5-(trifluoromethyl)-2-pyridinyl]-4-pyrimidinyl}- β-alanine;
Λ/-[6-(1 ,1 -dimethylethyl)-2-(4-methyl-2-pyridinyl)-4-pyrimidinyl]- β -alanine;
Λ/-{6-(1 ,1 -dimethylethyl)-2-[4-(methyloxy)-2-pyridinyl]-4-pyrimidinyl}- β-alanine;
Λ/-{6-(1 ,1 -dimethylethyl)-2-[5-(methyloxy)-2-pyridinyl]-4-pyrimidinyl}- β-alanine;
Λ/-[6-(1 ,1 -dimethylethyl)-2-(5-methyl-2-pyridinyl)-4-pyrimidinyl]- β-alanine;
A/-[2-[4-(dimethylamino)-2-pyridinyl]-6-(1 ,1 -dimethylethyl)-4-pyrimidinyl]- β-alanine;
A/-[6-[7-(methyloxy)-1 ,2,4,5-tetrahydro-3/-/-3-benzazepin-3-yl]-2-(2-pyridinyl)-4-pyrimidinyl]- β-alanine;
A/-[6-(4-acetyl-1 -piperazinyl)-2-(2-pyridinyl)-4-pyrimidinyl]- β-alanine;
A/-[6-[4-(methylsulfonyl)-1 -piperazinyl]-2-(2-pyridinyl)-4-pyrimidinyl]- β-alanine; A/-[6-[3-(acetylamino)-1^yrrolidinyl]-2-(2^yridinyl)-4^yrimidinyl]- β-alanine;
A/-[6-{3-[(methylsulfonyl)amino]-1-pyrrolidm^^
A/-[6-[(phenylmethyl)oxy]-2-(2-pyridinyl)-4-pyrimidinyr]-p-alanine;
A/-[6-[3-(acetylamino)-1^iperidinyl]-2-(2^yridinyl)-4^yrimidinyi]-p-alanine;
A/-[6-{3-[(methylsulfonyl)amino]-1-piperid^
A/-[6-(4^henyl-1^iperidinyl)-2-(2^yridinyl)-4-pyrimidinyr|-p-alanine;
A/-[6-(4-hydroxy-1^iperidinyl)-2-(2^yridinyl)-4^yrimidinyr]-p-alanine;
A/-{2-(2^yridinyl)-6-[(4^yridinylmethyl)amino]-4^yrimidinyl}-p-alanine;
A/-{2-(2^yridinyl)-6-[(3^yridinylmethyl)amino]-4^yrimidinyl}-p-alanine;
A/-[6-(2,3-dihydro-1 H-inden-2-ylamino)-2-(2^yridinyl)-4^yrimidinyi]-p-alanine;
A/-[6-[(2^henylethyl)amino]-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine;
A/-[2-(2^yridinyl)-6-(4-thiomorpholinyl)-4-pyrimidinyl]-p-alanine;
A/-[6-(3-hydroxy-1^yrrolidinyl)-2-(2^yridinyl)-4-pyrimidinyr|-p-alanine;
Λ/-[6-(1 ,3-dihydro-2H-isoindol-2-yl)-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine;
A/-[2-(2-pyridinyl)-6-(1 ,3,4,5-tetrahydro-2H-2-benzazepin-2-yl)-4-pyrimidinyr|-p-alanine;
A/-[6-(1-piperidinyl)-2-(2-pyridinyl)-4-pyrimidinyT]-p-alanine;
A/-[6-(4-methyl-1^iperazinyl)-2-(2^yridinyl)-4^yrimidinyr]-p-alanine;
A/-[6-({[3-(methyloxy)phenyl]methyl}amino)-2-(2^yridinyl)-4^yrimidinyl]-p-alani
A/-[6-{[(3-cyanophenyl)methyl]amino}-2-(2^yridinyl)-4^yrimidinyi]-p-alanine;
A/-[6-{[2-(methyloxy)ethyl]amino}-2-(2^yridinyl)-4^yrimidinyT]-p-alanine;
Λ/-[6-(1 ,1-dioxido-4-thiomorpholinyl)-2-(2^yridinyl)-4^yrimidinyT]-p-alanine;
A/-[6-[bis(2-hydroxyethyl)amino]-2-(2^yridinyl)-4^yrimidinyT]-p-alanine;
A/-[6-[4-(phenylcarbonyl)-1^iperazinyl]-2-(2^yridinyl)-4^yrimidinyi]-p-alanine;
A/-[6-{3-[(methylamino)carbonyl]-1-piperidiny^
A/-{2-(2-pyridinyl)-6-[3-(1-pyrrolidinylcarbonyl)-^^
A/-[6-(4-methyl-1^iperidinyl)-2-(2^yridinyl)-4^yrimidinyr|-p-alanine;
A/-[6-(4,4-dimethyl-1^iperidinyl)-2-(2^yridinyl)-4^yrimidinyi]-p-alanine;
A/-[6-(4^ropanoyl-1^iperazinyl)-2-(2^yridinyl)-4-pyrimidinyr|-p-alanine;
A/-[6-(5-chloro-1 ,3-dihydro-2H-isoindol-2-yl)-2-(2^yridinyl)-4-pyrimidinyr|-p-alanine;
A/-[6-[5-(methyloxy)-1 ,3-dihydro-2H-isoindol-2-yl]-2-(2^yridinyl)-4^yrimidinyr]-p-alanine;
/V-[6-[(2-phenylethyl)oxy]-2-(2-pyridinyl)-4-pyrimidinyr]-p-alanine;
Λ/-[6-(1 -oxo-3, 4-dihydro-2(1 - )-isoquinolinyl)-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine;
A/-[6-(2-methyl-4,5,7,8-tetrahydro-6H-[1 ,3]thiazolo[4,5-olazepin-6-yl)-2-(2-pyridinyl)-4-pyrim^ alanine;
A/-[6-[7-(methylsulfonyl)-1 ,2,4,5-tetrahydro-3/-/-3-benzazepin-3-yl]-2-(2-pyridinyl)-4-pyrimidinyr|-p- alanine;
A/-[6-[7-(aminosulfonyl)-1 ,2,4,5-tetrahydro-3/-/-3-benzazepin-3-yl]-2-(2-pyridinyl)-4-pyrimidinyr]-p- alanine;
A/-[2-(2-pyridinyl)-6-(1 ,2,3,4-tetrahydro-2-naphthalenyloxy)-4-pyrimidinyi]-p-alanine; Λ/-{6-(1 ,2,4,5-tetrahydro-3H-3-benzazepin-3-yl)-2-[4-(1 ,2,4,5-tetrahydro-3H-3-benzazepin-3-yl)-2- pyridinyl]-4-pyrimidinyl}-p-alanine;
A/-[6-(1 -benzothien-3-yl)-2-(2^yridinyl)-4-pyrimidinyl]-p-alanine;
Λ/-(6-(1 ,3-dihydro-2H-isoindol-2-yl)-2-{4-[(phenylamino)methyl]-2^yridinyl}-4^y
Λ/-(6-(1 ,3 lihydro-2H-isoindol-2-yl)-2-{4-[(phenyloxy)methy^^
Λ/-[6-(1 ,3-dihydro-2H-isoindol-2-yl)-2-(4^henyl-2^yridinyl)-4^yrimidinyl]-p-alani
A/-[6-[(methylamino)carbonyl]-2-(2^yridinyl)-4^yrimidinyl]-p-alanine;
Λ/-[6-(1 -dimethylethyl)-2-(4-methyl-2^yridinyl)-4^yrimidinyl]-p-alanine;
A/-[6-[{[3-(aminocarbonyl)phenyl]methyl}(methyh^
A/-[2-{4-[3-(methylamino)propyl]-2^yridinyl}-6-(1 ,2,4,5-tetrahydro-3H-3-benzazepi
pyrimidinyl]-p-alanine;
3-({2-{4-[3-(methylamino)propyl]-2^yridinyl}-6-[(phenylmethyl)amino]-4^ propanoic acid;
3-({2-(4-{3-[(2-aminoethyl)amino]propyl}-2^yridinyl)-6-[(phenylmethyl)amino]-4- pyrimidinyl}amino)propanoic acid;
3-{[2-{4-[3-(methylamino)propyl]-2^yridinyl}-6-(1 ,2,4,5-tetrahydro-3H-3-benzazepin-3-yl)-4- pyrimidinyl]amino}propanic acid;
3-{[6-{[3-(methylamino)-3-oxopropyl]amino}-2-(2-pyridinyl)-4-pyrimidinyl]amin acid;
A/-[6-[(2-carboxyethyl)amino]-2-(2^yridinyl)-4^yrimidinyl]-p-alanyl-A/ -methylglycinam
A/-[6-({2-[3-(b-alanylamino)phenyl]ethyl}amino)-2-(2^yridinyl)-4^yrimidinyl]-p-alanine.
Further specific compounds are:
A/-[6-{[(2,6-dimethylphenyl)methyl]amino}-2-(2^yridinyl)-4^yrimidinyl]-p-alanine
A/-[2-{4-[(2-hydroxyethyl)amino]-2^yridinyl}-6-(1 ,2,4,5-tetrahydro-3H-3-benzazepin-3-yl)-4- pyrimidinyl]-p-alanine;
A/-{2-(2^yridinyl)-6-[4-(1 ^yrrolidinylcarbonyl)-1 ^iperidinyl]-4^yrimidinyl}-p-alanine;
A/-[6-(cyclohexylamino)-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine;
A/-[6-{4-[(dimethylamino)carbonyl]-1 ^iperidinyl^^^
3-{[6-{methyl[3-(methylamino)-3-oxopropyl]amino}-2- ^ acid
A/-[6-{[(3,4-dichlorophenyl)methyl]oxy}-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine
A/-[6-[(1 -phenylethyl)oxy]-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine
A/-[6-{[(3-chlorophenyl)methyl]oxy}-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine
/V-[6-[(2-naphthalenylmethyl)oxy]-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine In one aspect of the invention, there is provided a compound selected from:
N-[2-(2-pyridinyl)-6-(1 ,2,4,5-tetrahydro-3H-3-benzazepin-3-yl)-4-pyrimidinyl]-p-alanine and
N-[6-(1 -methylethyl)-2-(2-pyridinyl)-4-pyrimidinyl]-p- alanine, or a pharmaceutically acceptable salts thereof.
Specific examples of compounds formula (la) include the following:
N-[2-(2-pyridinyl)-6-(trifluoromethyl)-4-pyrimidinyl]-p-alanine;
N-[6-(4-morpholinyl)-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine;
N-[6-(methylamino)-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine;
N-[2-(2-pyridinyl)-6-(1 -pyrrolidinyl)-4-pyrimidinyl]-p-alanine;
N-[6-[(2-hydroxyethyl)amino]-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine;
N-[6-[(phenylmethyl)amino]-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine;
N-[6-[(2-hydroxyethyl)(methyl)amino]-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine;
N-[6-(dimethylamino)-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine;
N-[2-(2-pyridinyl)-6-(1 ,2,4,5-tetrahydro-3H-3-benzazepin-3-yl)-4-pyrimidinyl]-p-alanine.
The compounds of formula (I) may be in the form of a pro-drug, for example an ester which upon administration to the patient is capable of providing a compound of the present invention. In one aspect the ester is a C^alkyl ester. Esters have a higher cell penetrance than the corresponding acids so may offer a pro-drug approach to achieving cell activity. Esters have also been found to be weakly active inhibitors of JmjD3 in themselves.
The compounds of Formula (I) may be in the form of a salt.
Typically, the salts of the present invention are pharmaceutically acceptable salts. Salts encompassed within the term "pharmaceutically acceptable salts" refer to non-toxic salts of the compounds of this invention. For a review on suitable salts see Berge et al, J. Pharm. Sci. 1977, 66, 1 -19.
Suitable pharmaceutically acceptable salts can include acid addition salts.
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, sulfuric, nitric, phosphoric, p-toluenesulfonic, benzenesulfonic, methanesulfonic, ethanesulfonic, naphthalenesulfonic such as 2-naphthalenesulfonic), 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, p-toluenesulfonate, benzenesulfonate, methanesulfonate, ethanesulfonate, naphthalenesulfonate (e.g. 2-naphthalenesulfonate) salt. Other non-pharmaceutically acceptable salts, e.g. trifluoroacetates, may be used, for example in the isolation of 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 compounds of formula (I).
While it is possible that, for use in therapy, the compound of formula (I) or a pharmaceutically acceptable salt thereof may be administered as the raw chemical, it is possible to present the active ingredient as a pharmaceutical composition. Accordingly, the invention further provides pharmaceutical compositions comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof and one or more pharmaceutically acceptable carriers, diluents, or excipients. The carrier(s), diluents(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 a compound of formula (I), or pharmaceutically acceptable salts thereof, with one or more pharmaceutically acceptable carriers, diluents or excipients. The pharmaceutical composition can be for use in the treatment and/or prophylaxis of any of the conditions described herein.
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 once or more than once a day. Such pharmaceutical compositions may be prepared by any of the methods well known in the pharmacy art. 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).
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 are prepared by reducing the compound to a suitable fine size and mixing with a similarly prepared pharmaceutical carrier such as an edible carbohydrate, as, for example, starch or mannitol. Flavouring, preservative, dispersing and colouring agent can also be present.
Capsules are 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 colouring 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 flavoured 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 composition can also be prepared to prolong or sustain the release as for example by coating or embedding particulate material in polymers, wax or the like.
The compounds of formula (I) or a pharmaceutically acceptable salt thereof may 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 transdermal administration may be presented as discrete patches intended to remain in intimate contact with the epidermis of the recipient for a prolonged period of time.
Pharmaceutical compositions adapted for topical administration may be formulated as ointments, creams, suspensions, lotions, powders, solutions, pastes, gels, sprays, aerosols or oils. For treatments of the eye or other external tissues, for example mouth and skin, the compositions are preferably applied as a topical ointment or cream. When formulated in an ointment, the active ingredient may be employed with either a paraffinic or a water-miscible ointment base. Alternatively, the active ingredient may be formulated in a cream with an oil-in-water cream base or a water-in-oil base.
Pharmaceutical compositions adapted for topical administrations to the eye include eye drops wherein the active ingredient is dissolved or suspended in a suitable carrier, especially an aqueous solvent.
Pharmaceutical compositions adapted for topical administration in the mouth include lozenges, pastilles and mouth washes.
Pharmaceutical compositions adapted for rectal administration may be presented as suppositories or as enemas. Dosage forms for nasal or inhaled administration may conveniently be formulated as aerosols, solutions, suspensions drops, gels or dry powders.
For compositions suitable and/or adapted for inhaled administration, it is preferred that the agent is in a particle-size-reduced form, and more preferably the size-reduced form is obtained or obtainable by micronisation. The preferable particle size of the size-reduced (e.g. micronised) compound or salt or solvate is defined by a D50 value of about 0.5 to about 10 microns (for example as measured using laser diffraction). Compositions adapted for administration by inhalation include the particle dusts or mists. Suitable compositions wherein the carrier is a liquid for administration as a nasal spray or drops include aqueous or oil solutions/suspensions of the active ingredient which may be generated by means of various types of metered dose pressurised aerosols, nebulizers or insufflators.
Aerosol formulations, e.g. for inhaled administration, can comprise a solution or fine suspension of the agent in a pharmaceutically acceptable aqueous or non-aqueous solvent. Aerosol formulations can be presented in single or multidose quantities in sterile form in a sealed container, which can take the form of a cartridge or refill for use with an atomising device or inhaler. Alternatively the sealed container may be a unitary dispensing device such as a single dose nasal inhaler or an aerosol dispenser fitted with a metering valve (metered dose inhaler) which is intended for disposal once the contents of the container have been exhausted.
Where the dosage form comprises an aerosol dispenser, it preferably contains a suitable propellant under pressure such as compressed air, carbon dioxide or an organic propellant such as a hydrofluorocarbon (HFC). Suitable HFC propellants include 1 ,1 ,1 ,2,3,3, 3-heptafluoropropane and 1 ,1 ,1 ,2-tetrafluoroethane. The aerosol dosage forms can also take the form of a pump-atomiser. The pressurised aerosol may contain a solution or a suspension of the active compound. This may require the incorporation of additional excipients e.g. co-solvents and/or surfactants to improve the dispersion characteristics and homogeneity of suspension formulations. Solution formulations may also require the addition of co-solvents such as ethanol. Other excipient modifiers may also be incorporated to improve, for example, the stability and/or taste and/or fine particle mass characteristics (amount and/or profile) of the formulation.
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 agent, (preferably in particle-size-reduced form, e.g. in micronised form), and optionally a performance modifier such as L-leucine or another amino acid, cellobiose octaacetate and/or metals salts of stearic acid such as magnesium or calcium stearate. Aerosol formulations are preferably arranged so that each metered dose or "puff of aerosol contains a particular amount of a compound of the invention. Administration may be once daily or several times daily, for example 2, 3 4 or 8 times, giving for example 1 , 2 or 3 doses each time. The overall daily dose and the metered dose delivered by capsules and cartridges in an inhaler or insufflator will generally be double those with aerosol formulations.
Pharmaceutical compositions adapted for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams or spray formulations. Pharmaceutical compositions adapted for parental 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 nonaqueous 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.
It should be understood that in addition to the ingredients particularly mentioned above, the compositions may include other agents conventional in the art having regard to the type of formulation in question, for example those suitable for oral administration may include flavouring agents.
Antisense or RNA interference molecules may be administered to the mammal in need thereof. Alternatively, constructs including the same may be administered. Such molecules and constructs can be used to interfere with the expression of the protein of interest, e.g., histone demethylase and as such, modify histone demethylation. Typically delivery is by means known in the art.
Antisense or RNA interference molecules can be delivered in vitro to cells or in vivo, e.g., to tumors of a mammal. Nodes of delivery can be used without limitations, including: intravenous, intramuscular, intraperitoneal, intra-arterial, local delivery during surgery, endoscopic, subcutaneous, and per os. Vectors can be selected for desirable properties for any particular application. Vectors can be viral or plasmid. Adenoviral vectors are useful in this regard. Tissue-specific, cell-type specific, or otherwise regulatable promoters can be used to control the transcription of the inhibitory polynucleotide molecules. Non-viral carriers such as liposomes or nanospheres can also be used.
A therapeutically effective amount of the 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 subject, 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 particular, the subject to be treated is a mammal, particularly a human.
The compound of formula (I) or a pharmaceutically acceptable salt thereof may be administered in a daily dose. 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.
Suitably, the amount of the compound of compound of formula (I) or a pharmaceutically acceptable salt thereof administered according to the present invention will be an amount selected from 0.01 mg to 1000 mg per day (calculated as the free or unsalted compound).
The compounds of the formula (I) or a pharmaceutically acceptable salt thereof may be used in combination with or include one or more other therapeutic agents and may be administered either sequentially or simultaneously by any convenient route in separate or combined pharmaceutical compositions.
The compounds of formula (I) or a pharmaceutically acceptable salt thereof and further therapeutic agent(s) may be employed in combination by administration simultaneously in a unitary pharmaceutical composition including both compounds. Alternatively, the combination may be administered separately in separate pharmaceutical compositions, each including one of the compounds in a sequential manner wherein, for example, the compound of formula or a pharmaceutically acceptable salt thereof is administered first and the other second and visa versa. Such sequential administration may be close in time (e.g. simultaneously) or remote in time. Furthermore, it does not matter if the compounds are administered in the same dosage form, e.g. one compound may be administered topically and the other compound may be administered orally. Suitably, both compounds are administered orally.
The combinations may be presented as a combination kit. By the term "combination kit" "or kit of parts" as used herein is meant the pharmaceutical composition or compositions that are used to administer the combination according to the invention. When the agents of the combination are administered simultaneously, the combination kit can contain the agents in a single pharmaceutical composition, such as a tablet, or in separate pharmaceutical compositions. When the agents are not administered simultaneously, the combination kit will contain each agent in separate pharmaceutical compositions either in a single package or in separate pharmaceutical compositions in separate packages.
The combination kit can also be provided by instruction, such as dosage and administration instructions. Such dosage and administration instructions can be of the kind that are provided to a doctor, for example by a drug product label, or they can be of the kind that are provided by a doctor, such as instructions to a patient.
During a treatment regime, it will be appreciated that administration of each agent of the combination may be repeated one or more times.
When the combination is administered separately in a sequential manner wherein one is administered first and the other second or vice versa, such sequential administration may be close in time or remote in time. For example, administration of the other agent several minutes to several dozen minutes after the administration of the first agent, and administration of the other agent several hours to several days after the administration of the first agent are included, wherein the lapse of time is not limited, For example, one agent may be administered once a day, and the other agent may be administered 2 or 3 times a day, or one agent may be administered once a week, and the other agent may be administered once a day and the like.
It will be clear to a person skilled in the art that, where appropriate, the other therapeutic ingredients(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.
When combined in the same composition it will be appreciated that the two compounds must be stable and compatible with each other and the other components of the composition and may be formulated for administration. When formulated separately they may be provided in any convenient composition, conveniently, in such a manner as known for such compounds in the art.
When the compound of compound of formula (I) or a pharmaceutically acceptable salt thereof is used in combination with further therapeutic agent or agents active against the same disease, condition or disorder the dose of each agent may differ from that when the compound is used alone. Appropriate doses will be readily appreciated by those skilled in the art.
In one embodiment in the methods and uses of the present invention the mammal is a human. Provided herein are methods of changing the epigenetic status of cells and the treatment or prevention of cancer, inflammation and autoimmune diseases and conditions which may be improved by modulating the methylation status of histones, particularly H3K27 and thereby, e.g., modulate the level of expression of methylation repressed target genes. A method may comprise administering to a subject, e.g. a subject in need thereof, a therapeutically effective amount of an agent described herein. Thus in one aspect there is provided the use of a compound of formula (I) or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for treating cancer, autoimmune and inflammatory diseases or conditions. In one aspect the disease or condition is cancer.
Examples of cancer diseases and conditions in which compounds of formula (I), or a pharmaceutically acceptable salt or solvates thereof may have potentially beneficial antitumour effects include, but are not limited to, cancers of the lung, bone, pancreas, skin, head, neck, uterus, ovaries, stomach, colon, breast, esophagus, small intestine, bowel, endocrine system, thyroid glad, parathyroid gland, adrenal gland, urethra, prostate, penis, testes, ureter, bladder, kidney or liver; rectal cancer; cancer of the anal region; carcinomas of the fallopian tubes, endometrium, cervix, vagina, vulva, renal pelvis, renal cell; sarcoma of soft tissue; myxoma; rhabdomyoma; fibroma; lipoma; teratoma; cholangiocarcinoma; hepatoblastoma; angiosarcoma; hemagioma; hepatoma; fibrosarcoma; chondrosarcoma; myeloma; chronic or acute leukemia; lymphocytic lymphomas; primary CNS lymphoma; neoplasms of the CNS; spinal axis tumours; squamous cell carcinomas; synovial sarcoma; malignant pleural mesotheliomas; brain stem glioma; pituitary adenoma; bronchial adenoma; chondromatous hanlartoma; inesothelioma; Hodgkin's Disease or a combination of one or more of the foregoing cancers. The compounds of the compound of formula (I) or a pharmaceutically acceptable salt thereof may also be useful in the treatment of one or more diseases afflicting mammals which are characterized by cellular proliferation in the area of disorders associated with neo-vascularization and/or vascular permeability including blood vessel proliferative disorders including arthritis (rheumatoid arthritis) and restenosis; fibrotic disorders including hepatic cirrhosis and atherosclerosis; mesangial cell proliferative disorders include glomerulonephritis, diabetic nephropathy, malignant nephrosclerosis, thrombotic microangiopathy syndromes, proliferative retinopathies, organ transplant rejection and glomerulopathies; and metabolic disorders include psoriasis, diabetes mellitus, chronic wound healing, inflammation and neurodegenerative diseases. In the embodiment, the compound of compound of formula (I) or a pharmaceutically acceptable salt thereof may be employed with other therapeutic methods of cancer treatment. In particular, in antineoplastic therapy, combination therapy with other chemotherapeutic, hormonal, antibody agents as well as surgical and/or radiation treatments other than those mentioned above are envisaged. As indicated, therapeutically effective amounts of the compound of compound of formula (I) or a pharmaceutically acceptable salt thereof are discussed above. The therapeutically effective amount of the further therapeutic agents of the present invention will depend upon a number of factors including, for example, the age and weight of the mammal, the precise condition requiring treatment, the severity of the condition, the nature of the formulation, and the route of administration. Ultimately, the therapeutically effective amount will be at the discretion of the attendant physician or veterinarian. The relative timings of administration will be selected in order to achieve the desired combined therapeutic effect. In one embodiment, the further anti-cancer therapy is surgical and/or radiotherapy.
In one embodiment, the further anti-cancer therapy is at least one additional anti-neoplastic agent.
Any anti-neoplastic agent that has activity versus a susceptible tumor being treated may be utilized in the combination. Typical anti-neoplastic agents useful include, but are not limited to, anti-microtubule agents such as diterpenoids and vinca alkaloids; platinum coordination complexes; alkylating agents such as nitrogen mustards, oxazaphosphorines, alkylsulfonates, nitrosoureas, and triazenes; antibiotic agents such as anthracyclins, actinomycins and bleomycins; topoisomerase II inhibitors such as epipodophyllotoxins; antimetabolites such as purine and pyrimidine analogues and anti-folate compounds; topoisomerase I inhibitors such as camptothecins; hormones and hormonal analogues; signal transduction pathway inhibitors; non-receptor tyrosine angiogenesis inhibitors; immunotherapeutic agents; proapoptotic agents; and cell cycle signaling inhibitors.
Anti-microtubule or anti-mitotic agents:
Anti-microtubule or anti-mitotic agents are phase specific agents active against the microtubules of tumor cells during M or the mitosis phase of the cell cycle. Examples of anti-microtubule agents include, but are not limited to, diterpenoids and vinca alkaloids.
Diterpenoids, which are derived from natural sources, are phase specific anti -cancer agents that operate at the G2/M phases of the cell cycle. It is believed that the diterpenoids stabilize the β-tubulin subunit of the microtubules, by binding with this protein. Disassembly of the protein appears then to be inhibited with mitosis being arrested and cell death following. Examples of diterpenoids include, but are not limited to, paclitaxel and its analog docetaxel. Paclitaxel, 5p,20-epoxy-1 ,2 ,4,7p,10p,13 -hexa-hydroxytax-1 1 -en-9-one 4,10-diacetate 2-benzoate 13-ester with (2R,3S)-N-benzoyl-3-phenylisoserine; is a natural diterpene product isolated from the Pacific yew tree Taxus brevifolia and is commercially available as an injectable solution TAXOL®. It is a member of the taxane family of terpenes. Paclitaxel has been approved for clinical use in the treatment of refractory ovarian cancer in the United States (Markman et al., Yale Journal of Biology and Medicine, 64:583, 1991 ; McGuire et al., Ann. Intern, Med., 1 1 1 :273,1989) and for the treatment of breast cancer (Holmes et al., J. Nat. Cancer Inst., 83:1797,1991 .) It is a potential candidate for treatment of neoplasms in the skin (Einzig et. al., Proc. Am. Soc. Clin. Oncol., 20:46) and head and neck carcinomas (Forastire et. al., Sem. Oncol., 20:56, 1990). The compound also shows potential for the treatment of polycystic kidney disease (Woo et. al., Nature, 368:750. 1994), lung cancer and malaria. Treatment of patients with paclitaxel results in bone marrow suppression (multiple cell lineages, Ignoff, R.J. et. al, Cancer Chemotherapy Pocket Guidei 1998) related to the duration of dosing above a threshold concentration (50nM) (Kearns, CM. et. al., Seminars in Oncology, 3(6) p.16-23, 1995).
Docetaxel, (2R.3S)- N-carboxy-3-phenylisoserine,N-fe/ -butyl ester, 13-ester with δβ-20-epoxy- 1 ,2 ,4,7p,10p,13 -hexahydroxytax-1 1 -en-9-one 4-acetate 2-benzoate, trihydrate; is commercially available as an injectable solution as TAXOTERE®. Docetaxel is indicated for the treatment of breast cancer. Docetaxel is a semisynthetic derivative of paclitaxel q.v. , prepared using a natural precursor, 10-deacetyl-baccatin III, extracted from the needle of the European Yew tree. Vinca alkaloids are phase specific anti-neoplastic agents derived from the periwinkle plant. Vinca alkaloids act at the M phase (mitosis) of the cell cycle by binding specifically to tubulin. Consequently, the bound tubulin molecule is unable to polymerize into microtubules. Mitosis is believed to be arrested in metaphase with cell death following. Examples of vinca alkaloids include, but are not limited to, vinblastine, vincristine, and vinorelbine.
Vinblastine, vincaleukoblastine sulfate, is commercially available as VELBAN® as an injectable solution. Although, it has possible indication as a second line therapy of various solid tumors, it is primarily indicated in the treatment of testicular cancer and various lymphomas including Hodgkin's Disease; and lymphocytic and histiocytic lymphomas. Myelosuppression is the dose limiting side effect of vinblastine.
Vincristine, vincaleukoblastine, 22-oxo-, sulfate, is commercially available as ONCOVIN® as an injectable solution. Vincristine is indicated for the treatment of acute leukemias and has also found use in treatment regimens for Hodgkin's and non-Hodgkin's malignant lymphomas. Alopecia and neurologic effects are the most common side effect of vincristine and to a lesser extent myelosupression and gastrointestinal mucositis effects occur.
Vinorelbine, 3',4'-didehydro -4'-deoxy-C'-norvincaleukoblastine [R-(R*,R*)-2,3-dihydroxybutanedioate (1 :2)(salt)], commercially available as an injectable solution of vinorelbine tartrate (NAVELBINE®), is a semisynthetic vinca alkaloid. Vinorelbine is indicated as a single agent or in combination with other chemotherapeutic agents, such as cisplatin, in the treatment of various solid tumors, particularly non- small cell lung, advanced breast, and hormone refractory prostate cancers. Myelosuppression is the most common dose limiting side effect of vinorelbine. Platinum coordination complexes:
Platinum coordination complexes are non-phase specific anti-cancer agents, which are interactive with DNA. The platinum complexes enter tumor cells, undergo, aquation and form intra- and interstrand crosslinks with DNA causing adverse biological effects to the tumor. Examples of platinum coordination complexes include, but are not limited to, oxaliplatin, cisplatin and carboplatin. Cisplatin, cis-diamminedichloroplatinum, is commercially available as PLATINOL® as an injectable solution. Cisplatin is primarily indicated in the treatment of metastatic testicular and ovarian cancer and advanced bladder cancer. Carboplatin, platinum, diammine [1 ,1 -cyclobutane-dicarboxylate(2-)-0,0'], is commercially available as PARAPLATIN® as an injectable solution. Carboplatin is primarily indicated in the first and second line treatment of advanced ovarian carcinoma.
Alkylating agents:
Alkylating agents are non-phase anti-cancer specific agents and strong electrophiles. Typically, alkylating agents form covalent linkages, by alkylation, to DNA through nucleophilic moieties of the DNA molecule such as phosphate, amino, sulfhydryl, hydroxyl, carboxyl, and imidazole groups. Such alkylation disrupts nucleic acid function leading to cell death. Examples of alkylating agents include, but are not limited to, nitrogen mustards such as cyclophosphamide, melphalan, and chlorambucil; alkyl sulfonates such as busulfan; nitrosoureas such as carmustine; and triazenes such as dacarbazine.
Cyclophosphamide, 2-[bis(2-chloroethyl)amino]tetrahydro-2H-1 ,3,2-oxazaphosphorine 2-oxide monohydrate, is commercially available as an injectable solution or tablets as CYTOXAN®. Cyclophosphamide is indicated as a single agent or in combination with other chemotherapeutic agents, in the treatment of malignant lymphomas, multiple myeloma, and leukemias.
Melphalan, 4-[bis(2-chloroethyl)amino]-L-phenylalanine, is commercially available as an injectable solution or tablets as ALKERAN®. Melphalan is indicated for the palliative treatment of multiple myeloma and non-resectable epithelial carcinoma of the ovary. Bone marrow suppression is the most common dose limiting side effect of melphalan.
Chlorambucil, 4-[bis(2-chloroethyl)amino]benzenebutanoic acid, is commercially available as LEUKERAN® tablets. Chlorambucil is indicated for the palliative treatment of chronic lymphatic leukemia, and malignant lymphomas such as lymphosarcoma, giant follicular lymphoma, and Hodgkin's disease.
Busulfan, 1 ,4-butanediol dimethanesulfonate, is commercially available as MYLERAN® TABLETS. Busulfan is indicated for the palliative treatment of chronic myelogenous leukemia.
Carmustine, 1 ,3-[bis(2-chloroethyl)-1 -nitrosourea, is commercially available as single vials of lyophilized material as BiCNU®. Carmustine is indicated for the palliative treatment as a single agent or in combination with other agents for brain tumors, multiple myeloma, Hodgkin's disease, and non- Hodgkin's lymphomas.
Dacarbazine, 5-(3,3-dimethyl-1 -triazeno)-imidazole-4-carboxamide, is commercially available as single vials of material as DTIC-Dome®. Dacarbazine is indicated for the treatment of metastatic malignant melanoma and in combination with other agents for the second line treatment of Hodgkin's Disease.
Antibiotic anti-neoplastics
Antibiotic anti-neoplastics are non-phase specific agents, which bind or intercalate with DNA. Typically, such action results in stable DNA complexes or strand breakage, which disrupts ordinary function of the nucleic acids leading to cell death. Examples of antibiotic anti-neoplastic agents include, but are not limited to, actinomycins such as dactinomycin, anthrocyclins such as daunorubicin and doxorubicin; and bleomycins.
Dactinomycin, also know as Actinomycin D, is commercially available in injectable form as COSMEGEN® . Dactinomycin is indicated for the treatment of Wilm's tumor and rhabdomyosarcoma.
Daunorubicin, (8S-cis-)-8-acetyl-10-[(3-amino-2,3,6-trideoxy- -L-lyxo-hexopyranosyl)oxy]-7,8,9,10- tetrahydro-6,8,1 1 -trihydroxy-1 -methoxy-5,12 naphthacenedione hydrochloride, is commercially available as a liposomal injectable form as DAUNOXOME® or as an injectable as CERUBIDINE®. Daunorubicin is indicated for remission induction in the treatment of acute nonlymphocytic leukemia and advanced HIV associated Kaposi's sarcoma. Doxorubicin, (8S, 10S)-10-[(3-amino-2,3,6-trideoxy- -L-lyxo-hexopyranosyl)oxy]-8-glycoloyl, 7,8,9,10- tetrahydro-6,8,1 1 -trihydroxy-1 -methoxy-5,12 naphthacenedione hydrochloride, is commercially available as an injectable form as RUBEX® or ADRIAMYCIN RDF®. Doxorubicin is primarily indicated for the treatment of acute lymphoblastic leukemia and acute myeloblasts leukemia, but is also a useful component in the treatment of some solid tumors and lymphomas.
Bleomycin, a mixture of cytotoxic glycopeptide antibiotics isolated from a strain of Streptomyces verticillus, is commercially available as BLENOXANE®. Bleomycin is indicated as a palliative treatment, as a single agent or in combination with other agents, of squamous cell carcinoma, lymphomas, and testicular carcinomas.
Topoisomerase II inhibitors:
Topoisomerase II inhibitors include, but are not limited to, epipodophyllotoxins.
Epipodophyllotoxins are phase specific anti-neoplastic agents derived from the mandrake plant. Epipodophyllotoxins typically affect cells in the S and G2 phases of the cell cycle by forming a ternary complex with topoisomerase II and DNA causing DNA strand breaks. The strand breaks accumulate and cell death follows. Examples of epipodophyllotoxins include, but are not limited to, etoposide and teniposide. Etoposide, 4'-demethyl-epipodophyllotoxin 9[4,6-0-(R )-ethylidene-p-D-glucopyranoside], is commercially available as an injectable solution or capsules as VePESID® and is commonly known as VP-16. Etoposide is indicated as a single agent or in combination with other chemotherapy agents in the treatment of testicular and non-small cell lung cancers.
Teniposide, 4'-demethyl-epipodophyllotoxin 9[4,6-0-(R )-thenylidene-p-D-glucopyranoside], is commercially available as an injectable solution as VUMON® and is commonly known as VM-26. Teniposide is indicated as a single agent or in combination with other chemotherapy agents in the treatment of acute leukemia in children.
Antimetabolite neoplastic agents:
Antimetabolite neoplastic agents are phase specific anti-neoplastic agents that act at S phase (DNA synthesis) of the cell cycle by inhibiting DNA synthesis or by inhibiting purine or pyrimidine base synthesis and thereby limiting DNA synthesis. Consequently, S phase does not proceed and cell death follows. Examples of antimetabolite anti-neoplastic agents include, but are not limited to, fluorouracil, methotrexate, cytarabine, mecaptopurine, thioguanine, and gemcitabine.
5-fluorouracil, 5-fluoro-2,4- (1 H,3H) pyrimidinedione, is commercially available as fluorouracil. Administration of 5-fluorouracil leads to inhibition of thymidylate synthesis and is also incorporated into both RNA and DNA. The result typically is cell death. 5-fluorouracil is indicated as a single agent or in combination with other chemotherapy agents in the treatment of carcinomas of the breast, colon, rectum, stomach and pancreas. Other fluoropyrimidine analogs include 5-fluoro deoxyuridine (floxuridine) and 5-fluorodeoxyuridine monophosphate.
Cytarabine, 4-amino-1 -p-D-arabinofuranosyl-2 (I H)-pyrimidinone, is commercially available as CYTOSAR-U® and is commonly known as Ara-C. It is believed that cytarabine exhibits cell phase specificity at S-phase by inhibiting DNA chain elongation by terminal incorporation of cytarabine into the growing DNA chain. Cytarabine is indicated as a single agent or in combination with other chemotherapy agents in the treatment of acute leukemia. Other cytidine analogs include 5- azacytidine and 2',2'-difluorodeoxycytidine (gemcitabine). Mercaptopurine, 1 ,7-dihydro-6H-purine-6-thione monohydrate, is commercially available as PURINETHOL®. Mercaptopurine exhibits cell phase specificity at S-phase by inhibiting DNA synthesis by an as of yet unspecified mechanism. Mercaptopurine is indicated as a single agent or in combination with other chemotherapy agents in the treatment of acute leukemia. A useful mercaptopurine analog is azathioprine.
Thioguanine, 2-amino-1 ,7-dihydro-6H-purine-6-thione, is commercially available as TABLOID®. Thioguanine exhibits cell phase specificity at S-phase by inhibiting DNA synthesis by an as of yet unspecified mechanism. Thioguanine is indicated as a single agent or in combination with other chemotherapy agents in the treatment of acute leukemia. Other purine analogs include pentostatin, erythrohydroxynonyladenine, fludarabine phosphate, and cladribine. Gemcitabine, 2'-deoxy-2', 2'-difluorocytidine monohydrochloride (β-isomer), is commercially available as GEMZAR®. Gemcitabine exhibits cell phase specificity at S-phase and by blocking progression of cells through the G1/S boundary. Gemcitabine is indicated in combination with cisplatin in the treatment of locally advanced non-small cell lung cancer and alone in the treatment of locally advanced pancreatic cancer.
Methotrexate, N-[4[[(2,4-diamino-6-pteridinyl) methyljmethylamino] benzoyl]-L-glutamic acid, is commercially available as methotrexate sodium. Methotrexate exhibits cell phase effects specifically at S-phase by inhibiting DNA synthesis, repair and/or replication through the inhibition of dyhydrofolic acid reductase which is required for synthesis of purine nucleotides and thymidylate. Methotrexate is indicated as a single agent or in combination with other chemotherapy agents in the treatment of choriocarcinoma, meningeal leukemia, non-Hodgkin's lymphoma, and carcinomas of the breast, head, neck, ovary and bladder. Topoisomerase I inhibitors:
Camptothecins, including, camptothecin and camptothecin derivatives are available or under development as Topoisomerase I inhibitors. Camptothecins cytotoxic activity is believed to be related to its Topoisomerase I inhibitory activity. Examples of camptothecins include, but are not limited to irinotecan, topotecan, and the various optical forms of 7-(4-methylpiperazino-methylene)-10,1 1 - ethylenedioxy-20-camptothecin described below.
Irinotecan HCI, (4S)-4,1 1 -diethyl-4-hydroxy-9-[(4-piperidinopiperidino) carbonyloxy]-1 H- pyrano[3',4',6,7]indolizino[1 ,2-b]quinoline-3, 14(41-1, 12H)-dione hydrochloride, is commercially available as the injectable solution CAMPTOSAR®. Irinotecan is a derivative of camptothecin which binds, along with its active metabolite SN-38, to the topoisomerase I - DNA complex. It is believed that cytotoxicity occurs as a result of irreparable double strand breaks caused by interaction of the topoisomerase I : DNA : irintecan or SN-38 ternary complex with replication enzymes. Irinotecan is indicated for treatment of metastatic cancer of the colon or rectum. Topotecan HCI, (S)-10-[(dimethylamino)methyl]-4-ethyl-4,9-dihydroxy-1 H- pyrano[3',4',6,7]indolizino[1 ,2-b]quinoline-3,14-(4H,12H)-dione monohydrochloride, is commercially available as the injectable solution HYCAMTIN®. Topotecan is a derivative of camptothecin which binds to the topoisomerase I - DNA complex and prevents religation of singles strand breaks caused by Topoisomerase I in response to torsional strain of the DNA molecule. Topotecan is indicated for second line treatment of metastatic carcinoma of the ovary and small cell lung cancer.
Hormones and hormonal analogues:
Hormones and hormonal analogues are useful compounds for treating cancers in which there is a relationship between the hormone(s) and growth and/or lack of growth of the cancer. Examples of hormones and hormonal analogues useful in cancer treatment include, but are not limited to, adrenocorticosteroids such as prednisone and prednisolone which are useful in the treatment of malignant lymphoma and acute leukemia in children ; aminoglutethimide and other aromatase inhibitors such as anastrozole, letrazole, vorazole, and exemestane useful in the treatment of adrenocortical carcinoma and hormone dependent breast carcinoma containing estrogen receptors; progestrins such as megestrol acetate useful in the treatment of hormone dependent breast cancer and endometrial carcinoma; estrogens, androgens, and anti-androgens such as flutamide, nilutamide, bicalutamide, cyproterone acetate and 5 -reductases such as finasteride and dutasteride, useful in the treatment of prostatic carcinoma and benign prostatic hypertrophy; anti-estrogens such as tamoxifen, toremifene, raloxifene, droloxifene, iodoxyfene, as well as selective estrogen receptor modulators (SERMS) such those described in U.S. Patent Nos. 5,681 ,835, 5,877,219, and 6,207,716, useful in the treatment of hormone dependent breast carcinoma and other susceptible cancers; and gonadotropin-releasing hormone (GnRH) and analogues thereof which stimulate the release of leutinizing hormone (LH) and/or follicle stimulating hormone (FSH) for the treatment prostatic carcinoma, for instance, LHRH agonists and antagagonists such as goserelin acetate and luprolide.
Signal transduction pathway inhibitors:
Signal transduction pathway inhibitors are those inhibitors, which block or inhibit a chemical process which evokes an intracellular change. As used herein this change is cell proliferation or differentiation. Signal tranduction inhibitors useful in the present invention include inhibitors of receptor tyrosine kinases, non-receptor tyrosine kinases, SH2/SH3domain blockers, serine/threonine kinases, phosphotidyl inositol-3 kinases, myo-inositol signaling, and Ras oncogenes.
Several protein tyrosine kinases catalyse the phosphorylation of specific tyrosyl residues in various proteins involved in the regulation of cell growth. Such protein tyrosine kinases can be broadly classified as receptor or non-receptor kinases. Receptor tyrosine kinases are transmembrane proteins having an extracellular ligand binding domain, a transmembrane domain, and a tyrosine kinase domain. Receptor tyrosine kinases are involved in the regulation of cell growth and are generally termed growth factor receptors. Inappropriate or uncontrolled activation of many of these kinases, i.e. aberrant kinase growth factor receptor activity, for example by over-expression or mutation, has been shown to result in uncontrolled cell growth. Accordingly, the aberrant activity of such kinases has been linked to malignant tissue growth. Consequently, inhibitors of such kinases could provide cancer treatment methods. Growth factor receptors include, for example, epidermal growth factor receptor (EGFr), platelet derived growth factor receptor (PDGFr), erbB2, erbB4, ret, vascular endothelial growth factor receptor (VEGFr), tyrosine kinase with immunoglobulin-like and epidermal growth factor homology domains (TIE-2), insulin growth factor -I (IGFI) receptor, macrophage colony stimulating factor (cfms), BTK, ckit, cmet, fibroblast growth factor (FGF) receptors, Trk receptors (TrkA, TrkB, and TrkC), ephrin (eph) receptors, and the RET protooncogene. Several inhibitors of growth receptors are under development and include ligand antagonists, antibodies, tyrosine kinase inhibitors and anti-sense oligonucleotides. Growth factor receptors and agents that inhibit growth factor receptor function are described, for instance, in Kath, John C, Exp. Opin. Ther. Patents (2000) 10(6):803-818; Shawver et al DDT Vol 2, No. 2 February 1997; and Lofts, F. J. et al, "Growth factor receptors as targets", New Molecular Targets for Cancer Chemotherapy, ed. Workman, Paul and Kerr, David, CRC press 1994, London. Tyrosine kinases, which are not growth factor receptor kinases are termed non-receptor tyrosine kinases. Non-receptor tyrosine kinases useful in the present invention, which are targets or potential targets of anti-cancer drugs, include cSrc, Lck, Fyn, Yes, Jak, cAbl, FAK (Focal adhesion kinase), Brutons tyrosine kinase, and Bcr-Abl. Such non-receptor kinases and agents which inhibit nonreceptor tyrosine kinase function are described in Sinh, S. and Corey, S.J., (1999) Journal of Hematotherapy and Stem Cell Research 8 (5): 465 - 80; and Bolen, J.B., Brugge, J.S., (1997) Annual review of Immunology. 15: 371 -404. SH2/SH3 domain blockers are agents that disrupt SH2 or SH3 domain binding in a variety of enzymes or adaptor proteins including, PI3-K p85 subunit, Src family kinases, adaptor molecules (She, Crk, Nek, Grb2) and Ras-GAP. SH2/SH3 domains as targets for anti-cancer drugs are discussed in Smithgall, T.E. (1995), Journal of Pharmacological and Toxicological Methods. 34(3) 125-32. Inhibitors of Serine/Threonine Kinases including MAP kinase cascade blockers which include blockers of Raf kinases (rafk), Mitogen or Extracellular Regulated Kinase (MEKs), and Extracellular Regulated Kinases (ERKs); and Protein kinase C family member blockers including blockers of PKCs (alpha, beta, gamma, epsilon, mu, lambda, iota, zeta). IkB kinase family (IKKa, IKKb), PKB family kinases, akt kinase family members, and TGF beta receptor kinases. Such Serine/Threonine kinases and inhibitors thereof are described in Yamamoto, T., Taya, S., Kaibuchi, K., (1999), Journal of Biochemistry. 126 (5) 799-803; Brodt, P, Samani, A., and Navab, R. (2000), Biochemical Pharmacology, 60. 1 101 -1 107; Massague, J., Weis-Garcia, F. (1996) Cancer Surveys. 27:41 -64; Philip, P.A., and Harris, A.L. (1995), Cancer Treatment and Research. 78: 3-27, Lackey, K. et al Bioorganic and Medicinal Chemistry Letters, (10), 2000, 223-226; U.S. Patent No. 6,268,391 ; and Martinez-lacaci, L., et al, Int. J. Cancer (2000), 88(1), 44-52.
Inhibitors of Phosphotidyl inositol-3 Kinase family members including blockers of PI3-kinase, ATM, DNA-PK, and Ku are also useful in the present invention. Such kinases are discussed in Abraham, R.T. (1996), Current Opinion in Immunology. 8 (3) 412-8; Canman, C.E., Lim, D.S. (1998), Oncogene 17 (25) 3301 -3308; Jackson, S.P. (1997), International Journal of Biochemistry and Cell Biology. 29 (7):935-8; and Zhong, H. et al, Cancer res, (2000) 60(6), 1541 -1545.
Also useful in the present invention are Myo-inositol signaling inhibitors such as phospholipase C blockers and Myoinositol analogues. Such signal inhibitors are described in Powis, G., and Kozikowski A., (1994) New Molecular Targets for Cancer Chemotherapy ed., Paul Workman and David Kerr, CRC press 1994, London.
Another group of signal transduction pathway inhibitors are inhibitors of Ras Oncogene. Such inhibitors include inhibitors of farnesyltransferase, geranyl-geranyl transferase, and CAAX proteases as well as anti-sense oligonucleotides, ribozymes and immunotherapy. Such inhibitors have been shown to block ras activation in cells containing wild type mutant ras , thereby acting as antiproliferation agents. Ras oncogene inhibition is discussed in Scharovsky, O.G., Rozados, V.R., Gervasoni, S.I. Matar, P. (2000), Journal of Biomedical Science. 7(4) 292-8; Ashby, M.N. (1998), Current Opinion in Lipidology. 9 (2) 99 - 102; and BioChim. Biophys. Acta, (19899) 1423(3):19-30.
As mentioned above, antibody antagonists to receptor kinase ligand binding may also serve as signal transduction inhibitors. This group of signal transduction pathway inhibitors includes the use of humanized antibodies to the extracellular ligand binding domain of receptor tyrosine kinases. For example Imclone C225 EGFR specific antibody (see Green, M.C. et al, Monoclonal Antibody Therapy for Solid Tumors, Cancer Treat. Rev., (2000), 26(4), 269-286); Herceptin ® erbB2 antibody (see Tyrosine Kinase Signalling in Breast cancererbB Family Receptor Tyrosine Kinases, Breast cancer Res., 2000, 2(3), 176-183); and 2CB VEGFR2 specific antibody (see Brekken, R.A. et al, Selective Inhibition of VEGFR2 Activity by a monoclonal Anti-VEGF antibody blocks tumor growth in mice, Cancer Res. (2000) 60, 51 17-5124).
Anti-angiogenic agents:
(i) Anti-angiogenic agents including non-receptorMEKngiogenesis inhibitors may alo be useful. Anti-angiogenic agents such as those which inhibit the effects of vascular edothelial growth factor, (for example the anti-vascular endothelial cell growth factor antibody bevacizumab [Avastin™], and compounds that work by other mechanisms (for example linomide, inhibitors of integrin ανβ3 function, endostatin and angiostatin);
Immunotherapeutic agents:
Agents used in immunotherapeutic regimens may also be useful in combination with the compounds of formula (I). Immunotherapy approaches, including for example ex-vivo and in-vivo approaches to increase the immunogenecity of patient tumour cells, such as transfection with cytokines such as interleukin 2, interleukin 4 or granulocyte-macrophage colony stimulating factor, approaches to decrease T-cell anergy, approaches using transfected immune cells such as cytokine-transfected dendritic cells, approaches using cytokine-transfected tumour cell lines and approaches using anti- idiotypic antibodies Proapoptotoc agents:
Agents used in proapoptotic regimens (e.g., bcl-2 antisense oligonucleotides) may also be used in the combination of the present invention.
Cell cycle signalling inhibitors
Cell cycle signalling inhibitors inhibit molecules involved in the control of the cell cycle. A family of protein kinases called cyclin dependent kinases (CDKs) and their interaction with a family of proteins termed cyclins controls progression through the eukaryotic cell cycle. The coordinate activation and inactivation of different cyclin/CDK complexes is necessary for normal progression through the cell cycle. Several inhibitors of cell cycle signalling are under development. For instance, examples of cyclin dependent kinases, including CDK2, CDK4, and CDK6 and inhibitors for the same are described in, for instance, Rosania et al, Exp. Opin. Ther. Patents (2000) 10(2):215-230. In one embodiment, the combination of the present invention comprises a compound of formula I or a pharmaceutically acceptable salt thereof and at least one anti-neoplastic agent selected from anti- microtubule agents, platinum coordination complexes, alkylating agents, antibiotic agents, topoisomerase II inhibitors, antimetabolites, topoisomerase I inhibitors, hormones and hormonal analogues, signal transduction pathway inhibitors, non-receptor tyrosine MEKngiogenesis inhibitors, immunotherapeutic agents, proapoptotic agents, and cell cycle signaling inhibitors.
In one embodiment, the combination of the present invention comprises a compound of formula I or a pharmaceutically acceptable salt thereof and at least one anti-neoplastic agent which is an anti- microtubule agent selected from diterpenoids and vinca alkaloids.
In a further embodiment, the at least one anti-neoplastic agent agent is a diterpenoid.
In a further embodiment, the at least one anti-neoplastic agent is a vinca alkaloid.
In one embodiment, the combination of the present invention comprises a compound of formula I or a salt or solvate thereof and at least one anti-neoplastic agent, which is a platinum coordination complex. In a further embodiment, the at least one anti-neoplastic agent is paclitaxel, carboplatin, or vinorelbine.
In a further embodiment, the at least one anti-neoplastic agent is carboplatin. In a further embodiment, the at least one anti-neoplastic agent is vinorelbine. In a further embodiment, the at least one anti-neoplastic agent is paclitaxel.
In one embodiment, the combination of the present invention comprises a compound of formula I or a pharmaceutically acceptable salt thereof and at least one anti-neoplastic agent which is a signal transduction pathway inhibitor.
In a further embodiment the signal transduction pathway inhibitor is an inhibitor of a growth factor receptor kinase VEGFR2, TIE2, PDGFR, BTK, erbB2, EGFr, IGFR-1 , TrkA, TrkB, TrkC, or c-fms.
In a further embodiment the signal transduction pathway inhibitor is an inhibitor of a serine/threonine kinase rafk, akt, or PKC-zeta.
In a further embodiment the signal transduction pathway inhibitor is an inhibitor of a non- receptor tyrosine kinase selected from the src family of kinases.
In a further embodiment the signal transduction pathway inhibitor is an inhibitor of c-src. In a further embodiment the signal transduction pathway inhibitor is an inhibitor of Ras oncogene selected from inhibitors of farnesyl transferase and geranylgeranyl transferase. In a further embodiment the signal transduction pathway inhibitor is an inhibitor of a serine/threonine kinase selected from the group consisting of PI3K.
In a further embodiment the signal transduction pathway inhibitor is a dual EGFr/erbB2 inhibitor, for example N-{3-Chloro-4-[(3-fluorobenzyl) oxy]phenyl}-6-[5-({[2-(methanesulphonyl)
ethyl]amino}methyl)-2-furyl]-4-quinazolinamine (structure below):
Figure imgf000041_0001
In one embodiment, the combination of the present invention comprises a compound of formula (I) or a pharmaceutically acceptable salt thereof and at least one anti-neoplastic agent which is a cell cycle signaling inhibitor.
In further embodiment, cell cycle signaling inhibitor is an inhibitor of CDK2, CDK4 or CDK6. In a further aspect, the disease or condition is autoimmune.
Autoimmune diseases associated with type 1 interferon include, but are not limited to Systemic lupus erythematosus, Psoriasis, insulin-dependent diabetes mellitus (IDDM), dermatomyositis and Sjogren's syndrome (SS).
In one aspect the disease or condition is inflammation.
Inflammation represents a group of vascular, cellular and neurological responses to trauma. Inflammation can be characterised as the movement of inflammatory cells such as monocytes, neutrophils and granulocytes into the tissues. This is usually associated with reduced endothelial barrier function and oedema into the tissues. Inflammation can be classified as either acute or chronic. Acute inflammation is the initial response of the body to harmful stimuli and is achieved by the increased movement of plasma and leukocytes from the blood into the injured tissues. A cascade of biochemical event propagates and matures the inflammatory response, involving the local vascular system, the immune system, and various cells within the injured tissue. Prolonged inflammation, known as chronic inflammation, leads to a progressive shift in the type of cells which are present at the site of inflammation and is characterised by simultaneous destruction and healing of the tissue from the inflammatory process.
When occurring as part of an immune response to infection or as an acute response to trauma, inflammation can be beneficial and is normally self-limiting. However, inflammation can be detrimental under various conditions. This includes the production of excessive inflammation in response to infectious agents, which can lead to significant organ damage and death (for example, in the setting of sepsis). Moreover, chronic inflammation is generally deleterious and is at the root of numerous chronic diseases, causing severe and irreversible damage to tissues. In such settings, the immune response is often directed against self-tissues (autoimmunity), although chronic responses to foreign entities can also lead to bystander damage to self tissues.
The aim of anti-inflammatory therapy is therefore to reduce this inflammation, to inhibit autoimmunity when present and to allow for the physiological process or healing and tissue repair to progress.
The compound of formula (I) may be used to treat inflammation of any tissue and organs of the body, including musculoskeletal inflammation, vascular inflammation, neural inflammation, digestive system inflammation, ocular inflammation, inflammation of the reproductive system, and other inflammation, as exemplified below.
Musculoskeletal inflammation refers to any inflammatory condition of the musculoskeletal system, particularly those conditions affecting skeletal joints, including joints of the hand, wrist, elbow, shoulder, jaw, spine, neck, hip, knew, ankle, and foot, and conditions affecting tissues connecting muscles to bones such as tendons. Examples of musculoskeletal inflammation which may be treated with compounds of formula (I) include arthritis (including, for example, osteoarthritis, rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis, acute and chronic infectious arthritis, arthritis associated with gout and pseudogout, and juvenile idiopathic arthritis), tendonitis, synovitis, tenosynovitis, bursitis, fibrositis (fibromyalgia), epicondylitis, myositis, and osteitis (including, for example, Paget's disease, osteitis pubis, and osteitis fibrosa cystic).
Ocular inflammation refers to inflammation of any structure of the eye, including the eye lids. Examples of ocular inflammation which may be treated with the compounds of formula (I) include blepharitis, blepharochalasis, conjunctivitis, dacryoadenitis, keratitis, keratoconjunctivitis sicca (dry eye), scleritis, trichiasis, and uveitis.
Examples of inflammation of the nervous system which may be treated with the compounds of formula (I) include encephalitis, Guillain-Barre syndrome, meningitis, neuromyotonia, narcolepsy, multiple sclerosis, myelitis and schizophrenia. Examples of inflammation of the vasculature or lymphatic system which may be treated with the compounds of formula (I) include arthrosclerosis, arthritis, phlebitis, vasculitis, and lymphangitis.
Examples of inflammatory conditions of the digestive system which may be treated with the compounds of formula (I) include cholangitis, cholecystitis, enteritis, enterocolitis, gastritis, gastroenteritis, inflammatory bowel disease (such as Crohn's disease and ulcerative colitis), ileitis, and proctitis.
Examples of inflammatory conditions of the reproductive system which may be treated with the compounds of formula (I) include cervicitis, chorioamnionitis, endometritis, epididymitis, omphalitis, oophoritis, orchitis, salpingitis, tubo-ovarian abscess, urethritis, vaginitis, vulvitis, and vulvodynia.
The compound of formula (I) may be used to treat autoimmune conditions having an inflammatory component. Such conditions include acute disseminated alopecia universalise, Behcet's disease, Chagas' disease, chronic fatigue syndrome, dysautonomia, encephalomyelitis, ankylosing spondylitis, aplastic anemia, hidradenitis suppurativa, autoimmune hepatitis, autoimmune oophoritis, celiac disease, Crohn's disease, diabetes mellitus type 1 , giant cell arteritis, goodpasture's syndrome, Grave's disease, Guillain-Barre syndrome, Hashimoto's disease, Henoch-Schonlein purpura, Kawasaki's disease, lupus erythematosus, microscopic colitis, microscopic polyarteritis, mixed connective tissue disease, multiple sclerosis, myasthenia gravis, opsoclonus myoclonus syndrome, optic neuritis, ord's thyroiditis, pemphigus, polyarteritis nodosa, polymyalgia, rheumatoid arthritis, Reiter's syndrome, Sjogren's syndrome, temporal arteritis, Wegener's granulomatosis, warm autoimmune haemolytic anemia, interstitial cystitis, lyme disease, morphea, psoriasis, sarcoidosis, scleroderma, ulcerative colitis, and vitiligo.
The compound of formula (I) may be used to treat T-cell mediated hypersensitivity diseases having an inflammatory component. Such conditions include contact hypersensitivity, contact dermatitis (including that due to poison ivy), uticaria, skin allergies, respiratory allergies (hayfever, allergic rhinitis) and gluten-sensitive enteropathy (Celliac disease).
Other inflammatory conditions which may be treated with the agents include, for example, appendicitis, dermatitis, dermatomyositis, endocarditis, fibrositis, gingivitis, glossitis, hepatitis, hidradenitis suppurativa, iritis, laryngitis, mastitis, myocarditis, nephritis, otitis, pancreatitis, parotitis, percarditis, peritonoitis, pharyngitis, pleuritis, pneumonitis, prostatistis, pyelonephritis, and stomatisi, transplant rejection (involving organs such as kidney, liver, heart, lung, pancreas (e.g., islet cells), bone marrow, cornea, small bowel, skin allografts, skin homografts, and heart valve xengrafts, sewrum sickness, and graft vs host disease), acute pancreatitis, chronic pancreatitis, acute respiratory distress syndrome, Sexary's syndrome, congenital adrenal hyperplasis, nonsuppurative thyroiditis, hypercalcemia associated with cancer, pemphigus, bullous dermatitis herpetiformis, severe erythema multiforme, exfoliative dermatitis, seborrheic dermatitis, seasonal or perennial allergic rhinitis, bronchial asthma, contact dermatitis, astopic dermatitis, drug hypersensistivity reactions, allergic conjunctivitis, keratitis, herpes zoster ophthalmicus, iritis and oiridocyclitis, chorioretinitis, optic neuritis, symptomatic sarcoidosis, fulminating or disseminated pulmonary tuberculosis chemotherapy, idiopathic thrombocytopenic purpura in adults, secondary thrombocytopenia in adults, acquired (autroimmine) haemolytic anemia, leukaemia and lymphomas in adults, acute leukaemia of childhood, regional enteritis, autoimmune vasculitis, multiple sclerosis, chronic obstructive pulmonary disease, solid organ transplant rejection, sepsis. Preferred treatments include treatment of transplant rejection, rheumatoid arthritis, psoriatic arthritis, multiple sclerosis, Type 1 diabetes, asthma, inflammatory bowel disease, systemic lupus erythematosis, psoriasis, chronic obstructive pulmonary disease, and inflammation accompanying infectious conditions (e.g., sepsis).
The compound of formula (I) and pharmaceutical compositions containing it may be used in combination with or include one or more other therapeutic agents, for example selected from NSAIDS, corticosteroids, COX-2 inhibitors, cytokine inhibitors, anti-TNF agents, inhibitors oncostatin M, antimalarials, immunsuppressive and cytostatics.
The methods of treatment and uses of the invention can be used in mammals, particularly in humans. General Synthetic Methods
Compounds of general formula (I) may be prepared by methods known in the art of organic synthesis as set forth in the schemes below and/or the specific Examples described below. In all of the methods, it is well understood that protecting groups for sensitive or reactive groups may be employed where necessary in accordance with general principles of chemistry. Protecting groups are manipulated according to standard methods of organic synthesis (T. W. Green and P. G. M. Wuts (1999) Protective Groups in Organic Synthesis, 3rd edition, John Wiley & Sons). These groups are removed at a convenient stage of the compound synthesis using methods that are readily apparent to those skilled in the art. The selection of processes as well as the reaction conditions and order of their execution shall be consistent with the preparation of compounds of Formula (I).
Compounds of formula (I) may be prepared by one or more of the following general schemes, and the routes described in the specific Examples section below.
Scheme 1 - Synthesis of A/-[6-(alkyl- and aryl-)-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanines
Figure imgf000045_0001
t-BuMgCI, Cul, THF
methyl 3-aminopropanoate, Et3N, DMF
2-pyridylzinc bromide, Pd(OAc)2 and S-Phos in toluene
2-pyridylzinc bromide, dichloropalladium - triphenylphosphane (1 :2) in THF, microwave LiOH, THF, H20 with or without MeOH
ArB(OH)2, Pd(OAc)2, Na2C03, THF/water
Figure imgf000045_0002
methyl β-alaninate (HCI salt), K2C03, DMA
2-pyridylzinc bromide, Pd(OAc)2, S-Phos, toluene
Scheme 3 - Synthesis of A/-[6-alkyl-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanines, A/-[6-amide-2-(2- pyridinyl)-4-pyrimidinyl]-p-alanines and A/-[6-alkyl amino-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanines
Figure imgf000046_0001
(i) NaOMe, ammonium chloride, EtOH
(ii) Sodium ethoxide in EtOH, Reflux or Na in EtOH, Reflux
(iii) POCI3
(iv) Ethyl or f-butyl /3-alaninate hydrochloride, DIPEA, 1 ,4-Dioxane, microwave
(v) Where R2 = Me or Et, LiOH, 1 :1 THF/H20 with or without MeOH, or KOH in MeOH,
Reflux
(vi) Where R2 = fBu, 5M HCI, THF
(vii) Where R3 = CI, R4NH2, DIPEA, DMSO, microwave
(viii) Where R3 = CI, R4NH2,NaOfBu, [2'-(dicyclohexylphosphanyl)-2- biphenylyl]dimethylamine, Pd2(dba)3, 1 ,4-dioxane, microwave
Where R3 = -C02Et, the following transformations were undertaken:
Figure imgf000046_0002
(i) LiOH, MeOH, H20 with or without MeOH (ii) MeNH2, HATU, DIPEA, DMF
(iii) 5M HCI, THF
Scheme 4 - Synthesis of /V-[6-(aryl-)-2-(2-pyridinyl)-4-pyrimidinyl]-/3-alanines, A/-[6-(alkyloxy-)-2-(2- pyridinyl)-4-pyrimidinyl]-/3-alanines, and A/-[6-(alkylamino-)-2-(2-pyridinyl)-4-pyrimidinyl]-D-alanines and their methyl and ethyl esters
Figure imgf000047_0001
(i) Methyl /3-alaninate or Ethyl /3-alaninate hydrochloride, DIPEA, 1 ,4-Dioxane
(ii) R2R3NH, DIPEA, DMSO, microwave
(iii) LiOH, 1 :1 THF/H20 with or without MeOH
(iv) Aryl boronic acid or aryl boronic acid pinacol ester, Pd(PPh3)4,1 ,4-dioxane/H20, microwave
(v) 25% by wt NaOMe in MeOH, DMSO, microwave
The following additional transformations were undertaken where, for example, where
R2R3N =
Figure imgf000047_0002
, according to documented chemical procedures:
Figure imgf000048_0001
(i) TFA, DCM
(ii) Methyl sulfonyl chloride, pyridine
(iii) Acetyl chloride, pyridine
(iv) LiOH, 1 :1 THF/H20 with or without MeOH
The followin additional transformations were undertaken where for exam le
Figure imgf000048_0002
(i) TFA, DCM
(ii) Polymer supported cyanoborohydride, AcOH, MeOH
(iii) LiOH, 1 :1 THF/HzO he following additional transformations were undertaken where, for example,
Figure imgf000049_0001
(i) TFA, DCM
(ii) A/ -methylglycinamide. HCI, DIPEA, HATU, DMF
(iii) MeNH2, DIPEA. HATU, DMF
(iv) LiOH, 1 :1 THF/H20
Scheme 5 - Synthesis of A/-[6-[(alkyloxy- or A/-amido-]-2-(2-pyridinyl)-4-pyrimidinyl]-p-alani
(iii)
Figure imgf000050_0001
POCI3
R1 R20H or R1 R2NH, NaH, DMF
^-alanine, DIPEA, 1 ,4-Dioxane, microwave
Ethyl ^-alaninate, DIPEA, 1 ,4-Dioxane, microwave then LiOH, THF/H20 R1 R2CHOH, DIAD, PPh3, THF
Scheme 6 - Synthesis of A/-{2-[4-(3-aminopropyl)-2^yridinyl]-6-[(phenylmethyl)amino]-4^yrimidinyl}-
Figure imgf000051_0001
Figure imgf000051_0002
(i) Pd(PPh3)4,1 10oC, microwave, 1 h
NaOMe, MeOH,NH4CI then diethyl propanedioate, NaOMe, reflux
p-TosCI, NEt3, DMAP, DCM
Ethyl β-alaninate (HCI salt), MeCN, NEt3,100°C, microwave, 20min
R1 R2NH,150°C, microwave, IPA, 1 h
AcOH, H20, 80°C
MeNH2,NaBH4,THF
Figure imgf000051_0003
(x) TFA, DCM The following examples are set forth to illustrate the effectiveness of the approach described in the present invention and to further exemplify particular applications of general processes described above. Accordingly, the following Example section is in no way intended to limit the scope of the invention contemplated herein. Examples
General All temperatures are in °C.
DCM refers to dichloromethane
DIPEA refers to /V,/V-diisopropylethylamine
DMAP refers to A/,/V-dimethyl-4-pyridinamine
DMSO refers to dimethylsulfoxide.
DMF refers to A/,A/-dimethylformamide
DMA refers to dimethyl acetamide
Ether refers to diethyl ether
EtOAC refers to ethyl acetate
HATU refers to A/-[(dimethylamino)(3-oxido-1 /-/-[1 ,2,3]triazolo[4,5-Jb]pyridin-1 -yl)methylidene]-/V- methylmethanaminium hexafluorophosphate
HPLC refers to high performance liquid chromatography
MDAP refers to mass-directed autopreparative HPLC
NEt3 refers to triethylamine
r.t. refers to room temperature
Rt refers to retention time
S-Phos: 2-dicyclohexylphosphino-2',6'-dimethoxybiphenyl
THF refers to tetrahydrofuran
TFA refers to trifluoroacetic acid
Tos refers to para-toluene sulfonyl
TosCI refers to para-toluene sulfonyl chloride H NMR spectra were recorded using a Varian NMR 400 MHz, referenced to tetramethylsilane. LC/MS (Method A) was conducted on an Acquity UPLC BEH C18 column (50mm x 2.1 mm i.d. 1 .7μηι packing diameter) at 40 degrees centigrade, eluting with 0.1 % v/v solution of Formic Acid in Water (Solvent A) and 0.1 % v/v solution of Formic Acid in Acetonitrile (Solvent B) using the following elution gradient 0-1 .5min 3 - 100% B, 1 .5-1 .9min 100% B, 1 .9 - 2.1 min 3% B at a flow rate of 1 ml/min. The UV detection was a summed signal from wavelength of 210nm to 350nm. The mass spectra were recorded on a Waters ZQ Mass Spectrometer using Alternate-scan Positive and Negative Electrospray. lonisation data was rounded to the nearest integer.
LC/MS (Method B) was conducted on an Acquity UPLC BEH C18 column (50mm x 2.1 mm i.d. 1 .7μηι packing diameter) at 40 degrees centigrade, eluting with 10 mM Ammonium Bicarbonate in water adjusted to pH 10 with Ammonia solution (Solvent A) and Acetonitrile (Solvent B) using the following elution gradient 0-1 .5min 1 - 97% B, 1 .5-1 .9min 97% B, 1 .9 - 2.1 min 100% B at a flow rate of 1 ml/min. The UV detection was a summed signal from wavelength of 210nm to 350nm. The mass spectra were recorded on a Waters ZQ Mass Spectrometer using Alternate-scan Positive and Negative Electrospray. lonisation data was rounded to the nearest integer.
LC/MS (Method C) was conducted on an Acquity UPLC BEH C18 column (50mm x 2.1 mm i.d. 1 .7μηι packing diameter) at 40 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 0-1 .5min 3 - 100% B, 1 .5-1.9min 100% B, 1 .9 - 2.0min 3% B at a flow rate of 1 ml/min. The UV detection was a summed signal from wavelength of 210nm to 350nm. The mass spectra were recorded on a Waters ZQ Mass Spectrometer using Alternate-scan Positive and Negative Electrospray. lonisation data was rounded to the nearest integer. LC/MS (Method D) was conducted on an Acquity UPLC BEH C18 column (50mm x
4.6mm i.d. 2.7μηι packing diameter) at 40 degrees centigrade, eluting with 0.05% v/v
solution of formic Acid in Water (Solvent A) and 0.05% v/v solution of formic Acid in
Acetonitrile (Solvent B) using the following elution gradient 0-1 .Omin 5-95% B,
1 .0-2. Omin 95% B, 2.0 - 2.01 min 95-5% B, 2.01 - 2.5min 5% B at a flow rate of
1 .8ml/min. The UV detection was a summed signal from wavelength of 214nm to
254nm. The mass spectra were recorded on a Waters ZQ Mass Spectrometer using
Positive Electrospray. lonisation data was rounded to the nearest integer.
LCMS (Method H) was conducted on a HALO C18 Column (50mm x 4.6 mm i.d. 2.7 μηι packing diameter) at 40 degrees centigrade, eluting with Water (Solvent A) and Acetonitrile (Solvent B) using the following elution gradient 0-1 .Omin 5 - 95% B, 1 .0-2. Omin - 95%, 2.0-2.01 min - 95-5%, 2.01 - 2.5min - 5% at a flow rate of 1 .8ml/min. The UV detection was a summed signal from wavelength of 214nm to 254nm. Preparative HPLC was conducted on a Gemini 5u C18 column (150 x 21 .2mm) eluting with acetonitrile (Solvent A) and 0.1 % TFA in water (Solvent B) using the following elution gradient at a flow rate of 20ml/min and injection volume of 4ml/inj:
Time(min) A% B%
0 3 97
10 60 40
10.5 95 5
12.5 95 5
13 3 97
Figure imgf000054_0001
The UV detection was from wavelength 214nm to 254nm.
Silica chromatography techniques include either automated (Flashmaster, Biotage SP4) techniques manual chromatography on pre-packed cartridges (SPE) or manually-packed flash columns.
Where compounds have been purified by mass-directed autopreparative chromatography (MDAP), one of the following 3 methods may be used: MDAP (Method E). The HPLC analysis was conducted on an XBridge C18 column (100mm x 30mm i.d. 5μηι packing diameter) at ambient temperature, eluting with 10mM Ammonium Bicarbonate in water adjusted to pH 10 with Ammonia solution (Solvent A) and Acetonitrile (Solvent B) using an elution gradient of between 0 and 100% over 15 or 25 minutes. The UV detection was an averaged signal from wavelength of 210nm to 350nm. The mass spectra were recorded on a Waters ZQ Mass Spectrometer using Alternate-scan Positive and Negative Electrospray. lonisation data was rounded to the nearest integer.
MDAP (Method F). The HPLC analysis was conducted on a Sunfire C18 column (150mm x 30mm i.d. 5μηι packing diameter) at ambient temperature, 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 an elution gradient of between 0 and 100% over 15 or 25 minutes.
The UV detection was an averaged signal from wavelength of 210nm to 350nm. The mass spectra were recorded on a Waters ZQ Mass Spectrometer using Alternate-scan Positive and Negative Electrospray. lonisation data was rounded to the nearest integer.
MDAP (Method G). The HPLC analysis was conducted on a Sunfire C18 column (150mm x 30mm i.d. 5μηι packing diameter) at ambient temperature, eluting with 0.1 % formic acid in water (Solvent A) and 0.1 % formic acid in acetonitrile (Solvent B) using an elution gradient of between 0 and 100% over 15 or 25 minutes.
The UV detection was an averaged signal from wavelength of 210nm to 350nm. The mass spectra were recorded on a Waters ZQ Mass Spectrometer using Alternate-scan Positive and Negative Electrospray. lonisation data was rounded to the nearest integer.
MDAP (Method l)._The HPLC was conducted on an XBridge C 18 column (100mm x 19mm i.d. 5μηι packing diameter) at room temperature, eluting with 10 mM Ammonium Bicarbonate in water adjusted to pH 10 with Ammonia solution (Solvent A) and Acetonitrile (Solvent B) using the following elution gradient 0-5min 30 - 30% B, 5-15min 30 - 85% B, 15-15.2min 85 - 99 %B, 15.2-18.9min 99 - 99 %B, 18.9-19.0min 99 - 30 %B, 19-20min 30 - 30 %B at a flow rate of 20ml/min. The UV detection was a summed signal from wavelength of 200nm to 310nm. The mass spectra were recorded on a Waters ZQ Mass Spectrometer using Alternate-scan Positive and Negative Electrospray. lonisation data was rounded to the nearest integer.
MDAP (Method J) The HPLC was conducted on a Gemini C18 column (150mm x 21 ,2mm i.d. 5um packing diameter), eluting with Acetonitrile (Solvent A) and a 0.1 % solution of Trifluoroacetic acid in water (Solvent B) using the following elution gradient 0-10min 40-50%B, 10-1 1 min 50-5%B, 1 1 -14min 5%B, 14-15min 5 -40%B at a flow rate of 20ml/min. The UV detection was a summed signal from wavelength of 214nm to 254 nm.
When the name of a commercial supplier is given after the name of a compound or a reagent, for instance "compound X (Aldrich)" or "compound X / Aldrich", this means that compound X is obtainable from a commercial supplier, such as the commercial supplier named.
Similarly, when a literature or a patent reference is given after the name of a compound, for instance compound Y (EP 0 123 456), this means that the preparation of the compound is described in the named reference. The names of the above mentioned Examples have been obtained using the compound naming programme "ACD Name Pro 6.02".
Figure imgf000055_0001
To a mixture of 2,4,6-trichloropyrimidine (10g, 54.5mmol)(Aldrich) and copper (I) iodide (519mg, 2.725mmol) in dry THF (100ml) was added dropwise t-butylmagnesium chloride (27.2ml, 2.0M in ethyl ether, 54.5mmol) at -10°C under N2. Then the reaction mixture was allowed to stir at -10°C ~ 0°C for 1 h. TLC (petroleum ether /ethyl acetate = 80/1) showed the reaction was complete. The reaction was quenched with saturated aq. NH4CI (50ml) at 0°C and extracted with ethyl acetate (100ml x 2) The combined organic layers were washed with H20, brine, dried over anhydrous Na2S04 and evaporated to give a crude product. The crude product was purified by column chromatography (eluted with petroleum ether and ethyl acetate from 1/0 to 100/1) to give the title compound as a white solid (10g).
LCMS (Method D): Rt = 1 .77min, MH+ 205/207
Intermediate 2: methyl A -r2-chloro-6-(1 ,1 -dimetriyletriyl)-4-pyrimidinyl1-B-alaninate
Figure imgf000056_0001
To a mixture of 2,4-dichloro-6-(1 ,1 -dimethylethyl)pyrimidine (3.0g, 14.6mmol), methyl 3- aminopropanoate hydrochloride (2.44g, 17.52mmol) (Shanghai Hanhong) in DMF (50ml) was added triethylamine (4.4g, 43.8mmol) at room temperature. The reaction mixture was allowed to stir at 40°C for 2 h. TLC (petroleum ether /ethyl acetate = 80/1) showed the reaction was complete. The reaction mixture was cooled and poured into H20 (100ml) and extracted with dichloromethane (100ml x 3). The combined organic layers were washed with H20, brine, dried over anhydrous Na2S04 and evaporated to give a crude product. The crude product was purified by column chromatography (eluted with petroleum ether and ethyl acetate from 80/1 to 6/1) to give the title compound as a white solid (2.6g).
LCMS (Method D): Rt = 1 .60min, MH+ 272
Intermediate 3: Methyl N-r2-chlor -6-(trifluoromethyl)-4-pyrimidinyl1- β -alaninate
Figure imgf000056_0002
To a solution of 2,4-dichloro-6-(trifluoromethyl)pyrimidine (350 mg, 1 .613 mmol) (Tyger Scientific Inc.) in A/,/V-Dimethylacetamide (DMA) (10 mL) were added methyl β-alaninate (HCI salt) (225 mg, 1 .613 mmol) and potassium carbonate (557 mg, 4.03 mmol). The reaction was stirred overnight at room temperature under an atmosphere of 15ml_ of water was added to the reaction mixture, followed by 30ml_ of ethyl acetate. The phases were separated and the aqueous one was extracted twice with 25ml_ of ethyl acetate. The organics were combined and washed with 30ml_ of brine, dried over magnesium suflate and filtered. It was concentrated to dryness to give 1 .6g of a yellow liquid. This product was purified by column chromatography biotage SP4, on a 100g SNAP cartridge, eluted with a gradient form 0 to 50% of ethyl acetate in cyclohexane. Fractions were concentrated to give the title compound as a white solid (232mg).
LCMS (Method A) : Rt = 0.96min, MH+ 284
Similarly prepared was the intermediate in the following table:
Figure imgf000057_0001
Figure imgf000057_0004
Intermediate 4: 4,6-dichloro-2-(2-pyridinyl)pyrimidine
Figure imgf000057_0002
The preparation of 4,6-dichloro-2-(2-pyridinyl)pyrimidine is known in the literature (Ref: WO
2009/087224, CAS No.: 10235-65-1)
Figure imgf000057_0003
To a solution of 4,6-dichloro-2-(2-pyridinyl)pyrimidine (500 mg, 2.212 mmol) in 1 ,4-Dioxane (5 mL), was added DIPEA (1 .159 mL, 6.64 mmol) and ethyl β-alaninate hydrochloride (374 mg, 2.433 mmol) and the reaction heated in a Biotage microwave at 80 °C for 2 h. A further portion of ethyl b-alaninate hydrochloride (34.0 mg, 0.221 mmol) was added and the reaction mixture heated in a Biotage microwave at 80 °C for 2 h in total. The reaction mixture was then partitioned between DCM and water. The aqueous layers were re-extracted, the organics were combined, passed through a hydrophobic frit and the solvent removed by vacuum to give a brown oil (688mg). The product was left under high vacuum overnight to give ethyl A/-[6-chloro-2-(2-pyridinyl)-4-pyrimidinyl]-p-alaninate as a brown solid (533mg, 79%).
LCMS (Method A): rt = 0.69 min, MH+ = 307.06 Similarly prepared were the intermediates in the following table using methyl β-alaninate
hydrochloride in place of ethyl β-alaninate hydrochloride in cases where R1 = Et:
Figure imgf000058_0001
Figure imgf000058_0002
Intermediate 6: methyl A-r6-(1,1-dimethyletriyl)-2-(2-pyridinyl)-4-pyrimidinyl1-B-alaninate
Figure imgf000059_0001
To a solution of methyl A/-[2-chloro-6-(1 ,1 -dimethylethyl)-4-pyrimidinyl]-p-alaninate_(1 .0g, 3.69mmol), palladium acetate (41 .5mg, 0.1845mmol), 2-dicyclohexylphosphino-2',6'-dimethoxybiphenyl (S-Phos) (151 .5mg, 0.369mmol)(Aldrich) in dry toluene (100ml) was added 2-pyridylzinc bromide (15ml, 0.5 M in THF, 7.5mmol) (Aldrich) at room temperature under N2. The reaction mixture was heated to 100°C and stirred for 3h. TLC (petroleum ether /ethyl acetate = 4/1) showed most starting material was consumed. The reaction mixture was cooled and quenched with saturated aq. NH4CI (50ml) and extracted with dichloromethane (100ml x 3). The combined organic layers were washed with H20, brine, dried over anhydrous Na2S04, filtered and evaporated to give the crude product. The crude product was purified with column chromatography (eluted with petroleum ether and ethyl acetate from 4/1 to 1/2) to give the title compound as a light yellow solid (500mg).
LCMS (Method D): Rt = 1 .32min, MH+ 315
Similarly prepared was the intermediate in the following table:
Figure imgf000059_0003
Figure imgf000059_0002
To a suspension of 5-(methyloxy)-2-pyridinecarbonitrile (3 g, 22.4 mmol) in dry EtOH (35 mL) was added NaOMe (0.12 g, 2.24 mmol). The resulting mixture was stirred at room temperature for 17 hr. Ammonium chloride (1 .56 g, 29.1 mmol) was added then the resulting mixture refluxed for 1 hr. The mixture was allowed to cool to room temperature then concentrated under reduced pressure to give the crude product as a brown solid. The crude product was triturated with diethyl ether and the resulting suspension filtered under vacuum then dried at 40°C under vacuum to give the title compound as an orange solid (4.3g). LCMS (Method B): Rt = 0.49min, MH+ 152.0
Similarly prepared were the intermediate table:
Figure imgf000060_0001
Figure imgf000060_0003
Figure imgf000060_0002
To a solution of 2-pyridinecarboximidamide hydrochloride (5 g, 31 .7 mmol) in EtOH (30 ml) under nitrogen and at 45 °C was added sodium ethoxide solution (1 1 .8 ml, 31 .7 mmol). The reaction mixture was heated to reflux for 1 hr then a solution of ethyl 4-methyl-3-oxopentanoate (5.12 ml, 31 .7 mmol) in EtOH (2ml) was added. The reaction mixture was allowed to reflux over the weekend. The reaction was cooled to room temperature then filtered and the filtrate concentrated under reduced pressure to give 6.82g material as a light brown/orange oil.
The crude product was purified with column chromatography (eluted with 0 to 1 % of a solution of 20% of 2M ammonia in MeOH and DCM) to give the title compound, 5.13g.
LCMS (Method A): Rt = 0.82 min, MH+ 216.1 Similarly prepared were the intermediates in the following table:
Figure imgf000061_0001
Figure imgf000061_0003
Intermediate 9: 4-chloro-6-(1 -methylethyl)-2- 2-pyridinyl)pyrimidine
Figure imgf000061_0002
A suspension of 6-(1 -methylethyl)-2-(2-pyridinyl)-4(1 /-/)-pyrimidinone (5.13 g, 23.8 mmol) in phosphorus oxychloride (1 1 .1 ml, 1 19 mmol) was heated under reflux for 4h. The reaction was allowed to cool to room temperature overnight then quenched by portionwise addition to aqueous sodium triacetate (1 13g in 300ml water) maintaining the temperature below 25°C. The mixture was then filtered no solid was collected. Sodium bicarbonate was added then the reaction mixture was partitioned between DCM and water. The aqueous layer was re-extracted then combined organics passed through a hydrophobic frit then concentrated under reduced pressure and dried overnight in a vacuum oven to afford the title compound as a brown oil, 5.05g.
LCMS (Method A): Rt = 0.94 min, MH+ 234.1
Similarly prepared was the intermediate in the following table:
Figure imgf000062_0001
Figure imgf000062_0003
Intermediate 10: ethyl N-r6-(1 ,1 -dimethylethyl)-2-(2-pyridinyl)-4-pyrimidinvn-|3-alaninate
Figure imgf000062_0002
To a solution of 4-chloro-6-(1 -methylethyl)-2-(2-pyridinyl)pyrimidine (400 mg, 1 .71 mmol) in 1 ,4- Dioxane (5 ml) was added DIPEA (0.897 ml_, 5.13 mmol) followed by ethyl β-alaninate hydrochloride (289 mg, 1 .88 mmol) were added and the reaction was heated in a microwave at 120 °C for 1 .5hr, then at 120°C for 1 hr and at 140°C for a further 1 .5hr. The mixture was partitioned between DCM and water then the organic layer isolated and the aqueous layer reextracted. Combined organic layers were passed through a hydrophobic frit then concentrated under reduced pressure to give a yellow oil. The crude product was purified with column chromatography (eluted with 0 to 10% 2M ammonia in MeOH in DCM) to give the title compound, 381 mg.
LCMS (Method A): Rt = 0.67min, MH+ 315.1
Similarly prepared was the intermediate in the following table:
Figure imgf000063_0001
Intermediate R Yield % LCMS
10a 53 Rt = 0.65 min, MH+ = ethyl A/-[6-cyclopropyl-2-(2-pyridinyl)- 313.26 (Method A)
*
4-pyrimidinyl]-p-alaninate
General procedure for the preparation of bis N-linked pyrimidines: Intermediate 11 : Ethyl A -r6-(4-morpholinyl)-2-(2-pyridinyl)-4-pyrimidinyl1-B-alaninate
Figure imgf000063_0002
Ethyl A/-[6-chloro-2-(2-pyridinyl)-4-pyrimidinyl]-p-alaninate (210 mg, 0.685 mmol) was taken up in dimethyl Sulfoxide (DMSO) (0.5 mL) and DIPEA (0.359 mL, 2.054 mmol) was added followed by morpholine (0.060 mL, 0.685 mmol) and the reaction was heated in a Biotage microwave at 120 °C for 2 h and then 140 °C for 4 h. DMSO (0.5mL) and MeOH (1 mL) were then added to the reaction mixture which was purified by Mass Directed AutoPreparative LCMS under high pH elution conditions (Method E). The solvent was evaporated under vacuum to give ethyl A/-[6-(4-morpholinyl)-2-(2- pyridinyl)-4-pyrimidinyl]-p-alaninate (191 mg, 78%) as a brown film. LCMS (Method A): Rt = 0.65 min, MH+ = 358.2
Other intermediates indicated in following table were prepared similarly, in some cases using 1 ,4- dioxane or EtOH in place of DMSO:
Figure imgf000063_0003
Figure imgf000064_0001
Figure imgf000065_0001
Figure imgf000066_0001
Further intermediates indicated in following table were prepared similarly:
Figure imgf000066_0002
Intermediate R1 R2 Ar Yield /% LCMS
Figure imgf000067_0001
Figure imgf000068_0001
Figure imgf000069_0001
To a microwave vial containing ethyl A/-[6-chloro-2-(2-pyridinyl)-4-pyrimidinyl]-p-alaninate (100 mg, 0.33 mmol) was added 3-methyl-4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)-1 /-/-pyrazole (102 mg, 0.49 mmol) then cesium carbonate (212 mg, 0.65 mmol), palladium tetrakis (37.7 mg, 0.033 mmol) and a solution of 1 ,4-Dioxane (1 ml) in water (0.50 ml). The reaction mixture was heated 100°C for 1 hr using a microwave then concentrated under reduced pressure then diluted with IPA (<1 ml_) and neutralised using 2M HCI (aq). The neutralised reaction mixture was loaded onto an SCX cartridge, washed five times with IPA then product eluted with a solution of 10% ammonia in IPA. Ammonia washes were combined and concentrated under reduced pressure to afford the crude product as an orange oil. The oil dissolved in DMSO then purified by MDAP (Method E) and the resulting product was dissolved in methanol and passed down a thiol SPE cartridge, eluting the product with methanol. The appropriate fractions were combined and concentrated under reduced pressure to afford the title compound as a white solid, 35mg (29%).
LCMS (Method B): Rt = 0.79 min, MH+ = 353.2 (Method A) Further intermediates indicated in following table were prepared similarly:
A N O
Figure imgf000070_0001
Figure imgf000070_0004
Figure imgf000070_0002
A mixture of 4-formyl-2-pyridinecarbonitrile (1 .38 g, 10.45 mmol) and aniline (1 .167 g, 12.53 mmol) in MeOH (200 mL) was stirred for 1 hr then NaCNBH4 (3.28 g, 52.2 mmol) was added and the mixture stirred for 1 hr. The solvent was evaporated under reduced pressure to give the crude product which was purified by column chromatography, eluting with a hexane and EtOAc solvent system to give the title compound, 1 .78 g, (81 %).
LCMS Rt 1 .54min, MH+ 210.1 , (Method H) Intermediate 68: 6-hvdroxy-2-f4-r(phenyloxy)methvn-2-pyridinylV4(1 ti)-pyrimidinone
Figure imgf000070_0003
To a solution of sodium (382 mg, 16.6 mmol) in EtOH (150 ml) stirred under nitrogen was added portionwise a solution of 4-[(phenyloxy)methyl]-2-pyridinecarboximidamide (756 mg, 3.3 mmol) in EtOH (50 ml). The reaction mixture was stirred for 1 hr then diethyl propanedioate (1066 mg, 6.65 mmol) was added portionwise. The reaction mixture refluxed for 12 hr then allowed to cool to room temperature then acidified to pH 7 by adding c.HCI dropwise. The mixture was filtered and the filter cake was washed with water (50ml) then dissolved in MeOH (300 ml). The resulting mixture was filtered and the organic layer evaporated to give a crude solid which was recrystallised using ethanol (300 ml) to give the title compound, 960mg (98%)..
LCMS (Method H) Rt 1 .38 min, MH+ = 295.1 The intermediates in the following table were similarly prepared:
Figure imgf000071_0001
Figure imgf000071_0003
Intermediate 72: phenylmethyl fr2-(4-hvdroxy-6-oxo-1 ,6-dihvdro-2-pyrimidinyl)-4- pyridinvUmethyDp enylcarbamate
Figure imgf000071_0002
To a mixture of 6-hydroxy-2-{4-[(phenylamino)methyl]-2-pyridinyl}-4(1 H)-pyrimidinone (100 mg, 0.340 mmol) and sodium hydrogen carbon ate (143 mg, 1 .699 mmol) in a solution of water (10 ml) and THF (10 ml) was added dropwise phenylmethyl chloridocarbonate (0.242 ml_, 1 .699 mmol).The mixture was stirred for 16 hr then concentrated under reduced pressure. The crude product was purified by column chromatography using MeOH in DCM to give the title compound, 93 mg (64 %).
LCMS (Method H) Rt 1 .48 min, MH+ = 429.1
Intermediate 73: methyl A -r6-phen l-2-(2-pyridinyl)-4-pyrimidinyl1-B-alaninate
Figure imgf000072_0001
To a solution of methyl A/-(2-chloro-6-phenyl-4-pyrimidinyl)-p-alaninate (50 mg, 0.171 mmol) and bis(triphenylphosphine)palladium dichloride (18.0 mg, 0.026 mmol) in THF (0.65ml) in a microwave vial was added bromo(2-pyridinyl)zinc (0.686 ml_, 0.343 mmol) under nitrogen. The reaction mixture was heated at 60°C for 1 hr using a microwave then diluted with DCM (20ml), washed with ammonium chloride (15ml) then water (25ml_). Combined aqueous phases were washed with DCM (25ml) then concentrated. The resulting yellow crude product was redissolved in DCM (20ml) then a saturated solution of potassium sodium tartrate tetrahydrate (20ml) was added then the mixture stirred for 1 h. The organic layer was washed with water (15ml) and combined aqueous phases back-extracted with DCM (30ml). The combined organic layers were concentrated and the resulting yellow solid purified by column chromatography, eluting with MeOH in DCM (0 to 5%). The resulting material was further purified using MDAP (Method E) to give the title compound as a white solid, 7mg (12%).
LCMS (Method A): Rt = 0.73 min, MH+ 335.1 Intermediate 74: 4-phenyl-2-pyridinecarboximidamide.hvdrochloride
Figure imgf000072_0002
Available commercially from Acesys Pharmatech and Nanjing Chemlin Chemical Industry Co.
Intermediate 75: 4,6-dichloro-2-(4-phenyl-2-pyridinyl)pyrimidine
Figure imgf000073_0001
A mixture of phosphoric trichloride (20 mL, 218 mmol) and 6-hydroxy-2-(4-phenyl-2-pyridinyl)-4(1 H)- pyrimidinone (1 .3 g, 4.90 mmol) was heated to reflux for 12 hr. The solvent was removed and crude product purified by column chromatography using an EtOAc/petroleum ether (1 :2) solvent system to give the title compound as a white solid, 280 mg (19%).
LCMS (Method H): Rt 1 .73 min, MH+ = 302.0
Similarly prepared were the intermediates in the following table:
Figure imgf000073_0002
Figure imgf000073_0003
Intermediate 80: ethyl Af-r6-fr2-(3-nitrophenyl)ethvnaminoV2-(2-pyridinyl)-4-pyrimidinvn-|3- alaninate
Figure imgf000074_0001
2-(3-Nitrophenyl)ethanamine hydrochloride salt (495 mg, 2.44 mmol) was dissolved in MeOH (25 mL) and then loaded onto an aminopropyl column that had been prewashed with MeOH. The column was then eluted with MeOH. The appropriate fractions were combined and evaporated under reduced pressure to give a colourless oil. The oil was dissolved in DMSO (5 mL) and added in a single portion to a microwave vial containing ethyl A/-[6-chloro-2-(2-pyridinyl)-4-pyrimidinyl]-p-alaninate (500 mg, 1 .63 mmol) at r.t. under N2. DIPEA (0.712 mL, 4.08 mmol) was added in a single portion and the vial was sparged with N2 for 10 min. The vial was then sealed and heated in a microwave to 160 °C for 30 min. Following cooling, the vial was reheated in microwave to 150 °C for 30 min. Following cooling, the solution was diluted with EtOAc (50 mL) and sat. aq. NH4CI (20 mL). The separated aqueous phase was extracted with EtOAc (2 χ 20 mL), the combined organic phase was passed through a hydrophobic frit and evaporated under reduced pressure to give an orange oil. The oil was purified by column chromatography, eluting with 0-6% 2 M NH3 in MeOH / CH2CI2 to give the title compound as an orange oil, 656 mg (92%).
LCMS (Method A): Rt = 0.88min, MH+ 437.1
Intermediate 81 : ethyl A -r6-(r2-(3-aminophenyl)ethyl1amino)-2-(2-pyridinyl)-4-pyrimidinyl1-B- alaninate
Figure imgf000074_0002
A solution of ethyl A/-[6-{[2-(3-nitrophenyl)ethyl]amino}-2-(2-pyridinyl)-4-pyrimidinyl]-p-alaninate (656 mg, 1.503 mmol) in EtOH (20 mL) was added to a flask containing palladium on carbon (160 mg, 10% by weight, 0.15 mmol) at r.t. under N2. The flask was evacuated and back-filled with H2 and then stirred under an atmosphere of H2 for 40 h. The suspension was then filtered through a plug of celite and the filtrate was evaporated under reduced pressure to give the title compound as an orange oil, 534 mg (87%). LCMS (Method A): Rt = 0.66min, MH 407.2 Intermediate 82: ethyl jV-r6-r(2-f3-r(Af-fr(1 ,1 -dimethylethyl)oxy1carbonylVB-
Figure imgf000075_0001
2,4,6-tripropyl-1 ,3,5,2,4,6-trioxatriphosphorinane 2,4,6-trioxide (1 .17 g, 50% by weight in EtOAc, 1 .84 mmol) was added dropwise over 2 min to a stirred solution of ethyl Λ/-[6-{[2-(3- aminophenyl)ethyl]amino}-2-(2-pyridinyl)-4-pyrimidinyl]-p-alaninate (534 mg, 1 .31 mmol), Λ/-{[(1 ,1 - dimethylethyl)oxy]carbonyl}-p-alanine (249 mg, 1 .31 mmol) and DIPEA (0.757 ml_, 4.34 mmol) in CH2CI2 (25 ml_) at r.t. under N2. The resultant solution was stirred at r.t. for 30 min and then sat. aq. NaHC03 (50 ml_) was added. The separated aqueous phase was extracted with CH2CI2 (2 50 ml_), the combined organic phase was passed through a hydrophobic frit and evaporated under reduced pressure to give a brown oil. The oil was purified by column chromatography, eluting with 0-10% 2 M NH3 in MeOH / CH2CI2 to give the title compound as a pale yellow oil, 238 mg (31 %).
LCMS (Method A): Rt = 0.92min, MH+ 578.5
Intermediate 83: ethyl A -r6-((2-r3-(B-alanylamino)phenyl1etriyl)amino)-2-(2-pyridinyl)-4- pyrimidinyll-B-alaninate
Figure imgf000075_0002
TFA (0.317 ml_, 4.12 mmol) was added in a single portion to a stirred solution of ethyl Λ/-[6-[(2-{3-[(Λ/- {[(1 ,1 -dimethylethyl)oxy]carbonyl}-p-alanyl)amino]phenyl}ethyl)amino]-2-(2-pyridinyl)-4-pyrimidinyl]- β - alaninate (238 mg, 0.41 mmol) in CH2CI2 (10 ml_) at r.t.. Following stirring at r.t. for 4 h, sat. aq. NaHC03 (10 ml_) was added portionwise over 2 min. The resultant biphasic solution was stirred rapidly at r.t. for 10 min and then the separated aqueous phase was extracted with CH2CI2 (2 30 ml_). The combined organic phase was passed through a hydrophobic frit and evaporated under reduced pressure to give a yellow oil. The oil was purified by column chromatography, eluting with 0- 20% 2 M NH3 in MeOH / CH2CI2 to give the title compound as a pale yellow oil, 83 mg (42%). LCMS (Method A): Rt = 0.63min, MH+ 478.3
Intermediate 84: ethyl 3-fr6-fmethvir3-(methylamino)-3-oxopropynamino>-2-(2-pyridinyl)-4- pyrimidinyl1amino)propanoate
Figure imgf000076_0001
A flask containing 3-[[6-{[3-(ethyloxy)-3-oxopropyl]amino}-2-(2-pyridinyl)-4- pyrimidinyl](methyl)amino]propanoic acid (60.0 mg, 0.16 mmol) and DCM (5 mL) was evacuated 3 times before oxalyl chloride (0.03 mL, 0.32 mmol) was added dropwise, followed by N,N- dimethylformamide (0.02 mL, 0.13 mmol). After 2 hr of stirring at r.t., the reaction mixture was concentrated under reduced pressure to give a brown solid which was taken up in DCM (5 mL). Methylamine (2M in THF) (0.09 mL, 0.18 mmol) and DIPEA (0.06 mL, 0.32 mmol) were added to this solution. After stirring at r.t. for 36 hr, the reaction mixture was concentrated under reduced pressure then purified by column chromatography, eluting with 0-10% methanolic ammonia (2M) - DCM to give the impure title compound as a brown oil, 36.0 mg (58%).
LCMS (Method A): Rt = 0.64 min, MH+ 387.2. Intermediate 85: ethyl 3-fr6-r(3-fr2-(methylamino)-2-oxoethvnamino>-3-oxopropyl)amino1-2-(2-
Pyridinyl)-4-pyrimidinyl1amino)propanoate
Figure imgf000076_0002
HATU (53.3 mg, 0.14 mmol) and DIPEA (0.05 mL, 0.26 mmol) were added to a solution of 3-{[6-{[3- (ethyloxy)-3-oxopropyl]amino}-2-(2-pyridinyl)-4-pyrimidinyl]amino}propanoic acid (42.0 mg, 0.12 mmol) in A/,A/-dimethylformamide (5 mL). After 30 min of stirring N1-methylglycinamide hydrochloride (17.5 mg, 0.14 mmol) was added to the reaction mixture which was then stirred for 3 hr. The reaction mixture was concentrated under reduced pressure and the residue purified by column chromatography, eluting with 0-14% methanolic ammonia (2M) - DCM to give the title compound as a colourless oil, 31 .6 mg (63%).
LCMS (Method A): Rt = 0.59 min, MH+ 430.2.
The intermediate indicated in following table was prepared similarly, but using methylamine (2M, in THF) in place of N1-methylglycinamide hydrochloride:
Figure imgf000077_0001
Figure imgf000077_0003
Figure imgf000077_0002
TFA (1 .13 mL, 14.60 mmol) was added to a solution of 1 ,1 -dimethylethyl 4-[6-{[3-(ethyloxy)-3- oxopropyl]amino}-2-(2-pyridinyl)-4-pyrimidinyl]-1 -piperazinecarboxylate (476 mg, 1 .04 mmol) in DCM (3 mL). After 1 h of stirring at r.t., the volatiles were removed in vacuo to afford a brown oil which was loaded in MeOH onto a 50 g SCX cartridge. The column was eluted with MeOH, followed by methanolic ammonia (2M). The appropriate fractions were combined and concentrated to give the title compound as a brown oil, 444 mg (Quantitative).
LCMS (Method A): Rt = 0.42 min, MH+ 357.2. Further intermediates indicated in following table were prepared similarly:
Figure imgf000078_0001
Intermediate R1 Yield/% LC MS
88 Quantitative Rt = 0.45 min ethyl A/-[6-(3-amino-1 - pyrrolidinyl)-2-(2-pyridinyl)- * MH+ 357.2
4-pyrimidinyl]-p-alaninate (Method A)
89 96 Rt = 0.48 min ethyl A/-[6-(3-amino-1 - MH+ 371 .3 piperidinyl)-2-(2-pyridinyl)- 4-pyrimidinyl]-p-alaninate (Method A)
*
Intermediate 90: 6-chloro-2-(2-pyridinyl)- -pyrimidinone
Figure imgf000078_0002
A suspension of 6-hydroxy-2-(2-pyridinyl)-4(1 /-/)-pyrimidinone (21 .6 g, 1 14 mmol) in phosphorus oxychloride (54 ml, 579 mmol) was heated under reflux for 4 hr, then stirred at r.t. for 74 hr and under reflux for 5.5 hr. The reaction mixture was concentrated in vacuo. The resulting black tar was carefully treated with an aqueous solution of NaOAc. The mixture was then taken to pH 10 by addition of a saturated solution of NaHC03. After addition of DCM, the mixture was filtered. The filtrate was partitioned, and the aqueous phase was extracted with DCM. The organic layers were combined, dried over IS^SC^, filtered, and concentrated in vacuo to give a dark brown/black oil (16.2 g). The residue was taken up in DCM and purified by column chromatography, eluting with 0-70% EtOAc in cyclohexane to give the title compound as a brown solid, 228.1 mg (0.9%). LCMS (Method A): Rt = 0.70 min, MH 208.1
Intermediate 91 : 4-chloro-6-r(phenylmethyl)oxy1-2-(2-pyridinyl)pyrimidine
Figure imgf000079_0001
Phenylmethanol (0.22 mL, 2.10 mmol) was added to a cold solution of triphenylphosphane (551 mg, 2.10 mmol), bis(l -methylethyl) -1 ,2-diazenedicarboxylate (0.41 mL, 2.10 mmol) and 6-chloro-2-(2- pyridinyl)-4(1 H)-pyrimidinone (218 mg, 1 .05 mmol) in THF (5 mL) stirred under nitrogen at 0 °C. After 2 hr of stirring at r.t., the reaction mixture was concentrated under reduced pressure. The crude material was purified by column chromatography, eluting with 0-50% EtOAc-cyclohexane. A second column chromatography purification step was undertaken on the resulting material, eluting with 0-30% EtOAc-cyclohexane. A third column chromatography step was undertaken on the resulting material, eluting with 5-30% EtOAc-cyclohexane. The impure product was further purified by column chromatography, eluting with 0-50% diethyl ether-cyclohexane to give the title compound as a yellow solid, 270 mg (86%).
LCMS (Method A): Rt = 1 .09 min, MH+ 298.1 .
Figure imgf000079_0002
Tetrakis(triphenylphosphine)palladium (109.0 mg, 0.09 mmol) was added to a suspension of 1 - benzothien-2-ylboronic acid (201 .0 mg, 1 .13 mmol), 4,6-dichloro-2-(2-pyridinyl)pyrimidine (213.0 mg, 0.94 mmol) and Cs2C03 (614.0 mg, 1 .88 mmol) in 1 ,4-dioxane (3 mL) and water (0.75 mL). The mixture was degassed with nitrogen for 30 min and heated to 100 °C in a Biotage initiator microwave for 0.5 hr. The reaction mixture was concentrated under reduced pressure and the product purified by column chromatography, eluting with 0-40% cyclohexane-EtOAc. The resulting material was dissolved with DCM (10 mL), then thiourea silica (1 .71 g) was added. After 1 hr of stirring at r.t., the silica was filtered off using a hydrophobic frit, under vacuum. The filtrate was concentrated under reduced pressure to give the title compound as a colourless oil, 167.0 mg (55%).
LCMS (Method A): Rt = 1 .18 min, MH+ 324.0. Intermediate 93: 4-r(phenyloxy)methyl1-2-pyridinecarbonitrile
Figure imgf000080_0001
To a solution of 4-(hydroxymethyl)-2-pyridinecarbonitrile (1 .03 g, 7.68 mmol), phenol (0.759 g, 8.06 mmol) and triphenylphosphane (2.1 15 g, 8.06 mmol) in Tetrahydrofuran (THF) (37.1 ml) stirred under nitrogen at 0°C was added diethyl-1 ,2-diazenedicarboxylate (1 .277 ml, 8.06 mmol) dropwise over 5 min, The reaction mixture was stirred at r.t. for 1 h, and then Celite (3 g) was added and the solvent was removed under reduced pressure. The resulting residue was purified by column chromatography using a hexane/ethyl acetate: 8/1 , to afford the title compound as yellow solid (1 .1 g, 64.7%).
1 H NMR (300 Hz, d6-DMSO) ppm, 8.74 (1 H, d), 8.08 (1 H, s), 7.80 (1 H, d), 7.34-7.29 (2H, m), 7.05- 6.95 (3H, m), 5.25 (2H, s).
Intermediate 94: 4-chloro-2-pyridinecarboximidamide
Figure imgf000080_0002
LiHMDS (1 M) (43.3 ml_, 43.3 mmol) was added dropwise, over 5 min, to a solution of 4-chloro-2- pyridinecarbonitrile (5.0 g, 36.1 mmol) in THF (300 ml_) stirred under nitrogen at -78 °C. The reaction mixture was stirred at r.t. for 20 min and quenched with aqueous HCI (1 .5M).The aqueous phase was washed with EtOAc and basified with an aqueous solution of NaHC03 (10%). The crude product was added to a silica gel column and was eluted with DCM/MeOH (20/1) to give the title compound as a brown solid, 3.1 g (53%).
LCMS (Method H): Rt = 0.43, MH+ = 156.
Intermediate 95: ethyl ΛΚ 6-chloro-2-r4-(1 ,2,4,5-tetrahydro-3H-3-benzazepin-3-yl)-2-pyridinyl1-4- pyrimidinvD-B-alaninate
Figure imgf000081_0001
DIPEA (0.1 1 ml_, 0.64 mmol) and 2,3,4, 5-tetrahydro-1 H-3-benzazepine (31 .5 mg, 0.21 mmol) were added to a solution of ethyl A/-[6-chloro-2-(4-chloro-2-pyridinyl)-4-pyrimidinyl]-p-alaninate (73.0 mg, 0.21 mmol) in DMSO (0.5 ml_) in a microwave vial. The system was heated to 160 °C in a microwave for 2 hr. The reaction mixture was concentrated under reduced pressure. The crude material was purified by column chromatography, eluting with 0-10% DCM-methanol. A second column chromatography purification step was undertaken, using 0-100% cyclohexane-EtOAc and then 2-12% DCM-methanol. Impure product was loaded onto a 5 g SCX column that had been pre conditioned with methanol, washed with methanol and then product eluted with methanolic ammonia (2M) (3 CV). The appropriate fractions were combined and evaporated in vacuo to give the crude title compound, 34.0 mg (35%).
LCMS (Method A): Rt = 0.99 min, MH+ 452.2.
Figure imgf000081_0002
2-(2-hydroxyethyl)-1 /-/-isoindole-1 ,3(2H)-dione (4.14 g, 21 .65 mmol) was added to a cold solution (0°C) of sodium hydride (0.95 g, 23.82 mmol) in A/,A/-dimethylformamide (80 ml_). After 30 min of stirring at 0 °C, 4-chloro-2-pyridinecarbonitrile (3.0 g, 21 .65 mmol) was added. After 6 hr of stirring at r.t., reaction mixture was concentrated under reduced pressure. EtOAc (100 ml_) and water (100 ml_) were added to the residue, a precipitate formed which was filtered off, washed with EtOAc to give the required product. The aqueous layer from the filtrate was extracted with EtOAc (100 ml_). The combined organic layers was dried through a hydrophobic frit and concentrated under reduced pressure to give a yellow solid. The yellow solid was combined with the solid from the precipitate, washed with methanol (3x20 ml_) to give the title compound as a cream solid, 4.77 g (75%).
LCMS (Method A): Rt = 0.87 min, MH+ 294.1 . Intermediate 97: 1 ,1 -dimethylethyl ethyl 3,3'-fr2-(2-pyridinyl)-4,6- pyrimidinediyl1diimino)dipropanoate
Figure imgf000082_0001
1 ,1 -dimethylethyl β-alaninate hydrochloride (1 .14 g, 6.25 mmol) was dissolved in MeOH (5 ml_) and then loaded onto an aminopropyl SPE column (20 g), previously prewashed with MeOH. The column was eluted with MeOH. The appropriate fractions were combined and evaporated under reduced pressure to give 1 ,1 -dimethylethyl β-alaninate (916.0 mg) as a white oil. 1 ,1 -dimethylethyl β-alaninate (706.0 mg, 4.86 mmol) and DIPEA (0.43 ml_, 2.43 mmol) were added to a solution of ethyl Λ/-[6- chloro-2-(2-pyridinyl)-4-pyrimidinyl]^-alaninate (746.0 mg, 2.43 mmol) in /'sopropanol (8 ml_) in a microwave vial. The reaction mixture was heated to 160 °C in a microwave for 2 hr. 1 ,1 -dimethylethyl β-alaninate (210.0 mg, 1 .45 mmol) and DIPEA (0.26 ml_, 1 .46 mmol) were added and the reaction mixture heated to 160°C in a microwave for 2 hr. The reaction mixture was concentrated under reduced pressure to give a brown oil. The residue was purified by column chromatography, eluting with 0-5% methanolic ammonia (2M). A second column chromatography purification was undertaken on the material, eluting with 0-5% methanolic ammonia (2M). The appropriate fractions were combined and evaporated in vacuo to give the title compound as a brown oil, 555.0 mg.
LCMS (Method A): Rt = 0.88 min, MH+ 416.2
Other intermediates indicated in following table were prepared similarly:
Figure imgf000082_0002
Intermediate R1 Yield /% LCMS Intermediate R1 Yield /% LCMS
98 Me 54 Rt = 0.93 min, ethyl 3-{[6-[{3-[(1 ,1 - MH+ = 430.2 dimethylethyl)oxy]-3- (Method A) oxopropyl}(methyl)amino]-2-(2- pyridinyl)-4- pyrimidinyl]amino}propanoate
Intermediate 99: 3-fr6-fr3-(ethyloxy)-3-oxopropynamino>-2-(2-pyridinyl)-4- pyrimidinyl1amino)propanoic acid
Figure imgf000083_0001
TFA (2.06 mL, 26.7 mmol) was added to a solution of 1 ,1 -dimethylethyl ethyl 3,3'-{[2-(2-pyridinyl)-4,6- pyrimidinediyl]diimino}dipropanoate (555.0 mg, 1 .3 mmol) in DCM (20 mL). The reaction mixture was stirred for 2 hr at r.t. and left without stirring for 16 hr. The reaction was concentrated under reduced pressure then water (20 mL) was added to the residue followed by DCM (30 mL). The layers were separated and the aqueous layer further extracted with DCM (2x30 mL). The combined organic layer was concentrated under reduced pressure to give a brown oil. The residue was dissolved in DMSO (2 mL) and purified by MDAP (Method G). The solvent was evaporated in vacuo to give the title compound as a yellow oil, 126 mg. (23%). LCMS (Method A): Rt = 0.63 min, MH+ 360.1 .
Other intermediates indicated in following table were prepared similarly:
Figure imgf000083_0002
Intermediates R1 Yield /% LCMS Intermediates R1 Yield /% LCMS
100 Me 85 Rt = 0.67 min,
3-[[6-{[3-(ethyloxy)-3- MH+ = 374.1 oxopropyl]amino}-2-(2-pyridinyl)-4- (Method A) pyrimidinyl](methyl)amino]propanoi
c acid
Intermediate 101 : 6-(1 ,1 -dimethylethyl)-2-(3-isoquinolinyl)-4(1 ti)-pyrimidinone
Figure imgf000084_0001
A suspension of 3-isoquinolinecarboximidamide dihydrochloride (1 .01 g, 4.14 mmol) (Tyger Scientific^ in Ethanol (4 ml) was treated with a solution of 21 % Sodium Ethanolate in Ethanol (3.40 ml, 9.10 mmol). To this mixture was added ethyl 4,4-dimethyl-3-oxopentanoate (0.760 ml, 4.14 mmol) and the resulting solution heated to reflux for 17 hours then allowed to cool down. 21 % Sodium Ethanolate in ethanol (3.40 ml, 9.10 mmol) was added and the resulting mixture was heated to reflux for 2 hours, ethyl 4,4-dimethyl-3-oxopentanoate (0.760 ml, 4.14 mmol) was added and the reaction mixture was re-heated to reflux for 1 hour. The reaction mixture was cooled to room temperature and filtered under light vacuum to afford the title compound. LCMS (Method A) Rt=1 .18 min, MH+ 280
The following intermediate in the table was similarl prepared:
Figure imgf000084_0002
Intermediate Ar Yield LCMS Intermediate Ar Yield LCMS
102 Quantitative Rt =
2-[4-(dimethylamino)-2-pyridinyl]-6-(1 ,1 -dimethylethyl)-4- OJOmin, pyrimidinol MH+ =
I 273.0
(Method A)
Intermediate 103: 6-(1 ,1 -dimethylethyl)-2- -(trifluoromethyl)-2-pyridinvn-4(1 ti)-pyrimidinone
Figure imgf000085_0001
Sodium methoxide (0.031 g, 0.581 mmol) was added to a suspension of 5-(trifluoromethyl)-2- pyridinecarbonitrile (1 g, 5.81 mmol) in dry Ethanol (10 mL). The resulting mixture was stirred overnight at r.t.. Ammonium chloride (0.405 g, 7.57 mmol) was added and mixture was stirred at r.t. for 2 hours, then refluxed for 90 min, Ethyl 4,4-dimethyl-3-oxopentanoate (2.197 mL, 1 1 .96 mmol) and sodium ethanolate in ethanol (5.49 mL, 14.70 mmol) were added and resulting mixture was refluxed overnight. The mixture was allowed to cool down, volatiles were removed under reduce pressure, then material was triturated with diethylether to afford after 6 hours in a vacuum oven the title product (1 .8g, 94% ) as a white solid.
LCMS (Method B), Rt = 1 .14min, MH+ = 298.1
The following intermediates in the table were similarl prepared:
Figure imgf000085_0002
Intermediate Ar Yield LCMS
104 * 35 Rt = 1 .08 min,
6-(1 ,1 -dimethylethyl)-2-(4-methyl-2- MH+ = 244.1 pyridinyl)-4(1 H)-pyrimidinone Jt) (Method A)
Figure imgf000086_0001
Intermediate 108: ethyl 6-hvdroxy-2-(2-pyridinyl)-4-pyrimidinecarboxylate
Figure imgf000086_0002
Diethyl 2-oxobutanedioate (4.31 mL, 26.6 mmol)(TCI Europe) was added to a suspension of 2- pyridinecarboximidamide (4.19 g, 26.6 mmol)(Alfa Aesar) in Ethanol (130 mL) at r.t. under nitrogen. Triethylamine (7.41 mL, 53.1 mmol) was added dropwise and the reaction mixture heated to 80 °C and stirred for 24 h in total. The reaction was allowed to cool to r.t. and concentrated in vacuo to a black oil. The crude product was purified by column chromatography using a 0 to 100% solution of 20% MeOH in DCM/DCM to give the title product as a brown oil (2.91 g, 45% yield) which was used in the subsequent reaction without further purification.
LCMS (Method A): Rt = 0.69 min, MH+ = 246.1
Intermediate 109: ethyl 6-chloro-2-(2-pyridinyl)-4-pyrimidinecarboxylate
Figure imgf000086_0003
A suspension of ethyl 6-hydroxy-2-(2-pyridinyl)-4-pyrimidinecarboxylate (2.91 g, 1 1 .87 mmol) in phosphorus oxychloride (6.08 mL, 65.3 mmol) was heated to 90 °C for 2 h. The reaction mixture was cooled to room temperature and then cautiously added to a stirring solution of sodium acetate (89 g, 653 mmol) in water (350 mL). The quenched reaction mixture was diluted with 10% MeOH/DCM (200mL) and the layers separated. The aqueous phase was further extracted with 10% MeOH/DCM (2 x 150mL) and the organic phase was dried (Na2S04), filtered and concentrated in vacuo to give a brown oil. The crude product was purified by column chromatography using a gradient of 0 to 75% EtOAc/cyclohexane to give the title compound as a beige solid (631 mg, 20 % yield)
LCMS (Method A): Rt = 0.85 min, MH+ = 264.0
The following intermediates in the table were similarly prepared:
Figure imgf000087_0001
Figure imgf000088_0001
Intermediate 117: ethyl 6-(f3-r(1 ,1 -dimethylethyl)oxy1-3-oxopropyl>amino)-2-(2-pyridinyl)-4- pyrimidinecarboxylate
Figure imgf000088_0002
To a solution of DIPEA (0.397 ml, 2.275 mmol) and 1 ,1-dimethylethyl β-alaninate hydrochloride (138 mg, 0.758 mmol) in Dimethyl Sulfoxide (DMSO) (4 ml) was added ethyl 6-chloro-2-(2-pyridinyl)-4- pyrimidinecarboxylate (200 mg, 0.758 mmol) and the reaction was stirred at room temperature overnight. The reaction mixture was purified by column chromatography eluting with a gradient of 0% to 5% of a 20% solution of 2M NH3/MeOH in DCM/DCM. The fractions were concentrated to give the title compound (208 mg, 0.475 mmol, 62.6 % yield) as a yellow oil.
LCMS (Method A): Rt = 0.81 min, MH+ = 373.1 Intermediate 118: 6-(f 3-Γ(1 ,1 -dimethylethyl)oxy1-3-oxopropyl>amino)-2-(2-pyridinyl)-4- pyrimidinecarboxylic acid HO
To a solution of ethyl 6-({3-[(1 ,1 -dimethylethyl)oxy]-3-oxopropyl}amino)-2-(2-pyridinyl)-4- pyrimidinecarboxylate (200 mg, 0.430 mmol) in Methanol (4 mL) and Water (1.333 mL) was added lithium hydroxide hydrate (90 mg, 2.148 mmol). The reaction was stirred at 0°C for 1 h. HCI (0.4 mL, 2M solution) was added to the reaction mixture. The mixture was concentrated to give a dark orange oil. The residue was dissolved in DMSO/MeOH 1 :1 and was purified by MDAP (Method E). The fractions were concentrated together to give the title compound, 105 mg (70%) as an orange solid.
LCMS (Method A): Rt = 0.74 min, MH+ = 345.2
Intermediate 119: 1 ,1 -dimethylethyl A -r6-r(methylamino)carbonyl1-2-(2-pyridinyl)-4-pyrimidinyl1- β-alaninate
Figure imgf000089_0001
To a stirring solution of 6-({3-[(1 ,1 -dimethylethyl)oxy]-3-oxopropyl}amino)-2-(2-pyridinyl)-4- pyrimidinecarboxylic acid (30 mg, 0.087 mmol) and methylamine (0.044 mL, 0.087 mmol) in N,N- Dimethylformamide (DMF) (2 mL) were added HATU (33.1 mg, 0.087 mmol) and DIPEA (0.030 mL, 0.174 mmol). The reaction mixture was stirred overnight at room temperature under nitrogen. Further methylamine (0.1 mL, 2eq.) was added. Subsequently, further methylamine (10eq.) was added and the reaction was stirred at 50 °C overnight. Further methylamine (10 eq.) was added along with further HATU (1 eq.). The reaction was stirred at r.t for 3h then concentrated under vacuum then material was dissolved in 0.9 mL of 1 :1 DMSO/Methanol and purified by MDAP (Method E) to give the title compound, 6.1 mg, (20%) as a yellow solid.
LCMS (Method A): Rt = 0.70 min, MH+ = 358.2
Intermediate 120: 4-Γ2-Π ,3-dioxolan-2-yl)ethyl1-2-pyridinecarbonitrile
Figure imgf000089_0002
To a dry microwave vial under nitrogen was added bromo[2-(1 ,3-dioxolan-2-yl)ethyl]zinc (5774 μΙ_, 2.89 mmol, 0.5M solution in THF)/(Aldrich) and 4-chloro-2-pyridinecarbonitrile (250 mg, 1 .804 mmol)/(Aldrich). N2 was bubbled through the mixture for 5 min. Pd(PPh3)4 (104 mg, 0.090 mmol) was added and N2 bubbled through the brown suspension for a further 2 min. The reaction mixture was then heated in a microwave to 1 10 °C and stirred for 1 h. The reaction mixture was cooled and quenched by the addition of NH4CI solution (20 mL) and water (20 mL). The organics were extracted with Et20 (30 mL) and the layers separated. The aqueous layer was further extracted with Et20 (2 x 20 mL) and the combined organics dried (Na2S04) and concentrated in vacuo to afford the crude product as a yellow oil. The crude product was further purified by silica chromatography, eluting with 0% to 100% ethyl acetate/cyclohexane to give the title compound (151 mg, 41 % yield).
LCMS (Method B): Rt = 0.75 min, MH+ = 205.1 .
Intermediate 121 : 2-f4-r2-(1 ,3-dioxol -2-yl)ethvn-2-pyridinylV6-hvdroxy-4(1 tf)-pyrimidinone
Figure imgf000090_0001
To a solution of 4-[2-(1 ,3-dioxolan-2-yl)ethyl]-2-pyridinecarbonitrile (1 .22 g, 5.99 mmol) in Methanol (1 1 .6 mL) was added sodium methoxide (0.259 g, 1 .197 mmol, 25-30% w/w in MeOH) and the reaction stirred at r.t. for ~1 .5 h. Ammonium chloride (0.480 g, 8.98 mmol) was added and the reaction mixture stirred at r.t.. Diethyl propanedioate (4.56 ml, 29.9 mmol) and sodium methoxide (10.35 g, 47.9 mmol) were added and the reaction heated to reflux (80 °C) for 8.5 h. Further sodium methoxide (10.35 g, 47.9 mmol) and diethyl propanedioate (4.56 ml, 29.9 mmol) were added and heating continued for a further 16 h. The reaction was then allowed to cool and concentrated in vacuo. EtOH (50 mL) was added and the reaction mixture sonicated for 5 min. The resultant suspension was filtered and the residue washed with further EtOH (3 x 40 mL). The combined filtrate was concentrated in vacuo to afford the title compound as an orange solid (8.31 g). Material was used without further purification in the subsequent reaction.
LCMS (Method A): Rt = 0.59 min, MH+ = 290.1 . Intermediate 122: 2-(4-r2-(1 ,3-dioxolan-2-yl)etriyl1-2-pyridinyl)-4,6-pyrimidinediyl bis(4- methylbenzenesulfonate)
Figure imgf000091_0001
To a solution of 2-{4-[2-(1 ,3-dioxolan-2-yl)ethyl]-2^yridinyl}-6-hydroxy-4(1 /^^yrimidinone (8.31 g, 5.75 mmol, 20% purity) in DCM (40 mL) was added sequentially triethylamine (5.29 mL, 37.9 mmol), 4-methylbenzenesulfonyl chloride (5.26 g, 27.6 mmol) and DMAP (0.140 g, 1 .149 mmol) and the resultant solution stirred at r.t. for 16 h. Further triethylamine (5.29 mL, 37.9 mmol) and 4- methylbenzenesulfonyl chloride (5.26 g, 27.6 mmol) were added and the reaction stirred for a further 2 h. The reaction mixture was then quenched by the addition of water (50 mL), further DCM (30 mL) was added and the layers separated. The aqueous phase was further extracted with DCM (2 x 40 mL) and the combined organic phase was dried over Na2S04, filtered and concentrated in vacuo to give a brown oil. The crude product was purified by column chromatography using a gradient of 10 to 100% EtOAc/cyclohexane to give the title product as a pale cream foam (2.35 g, 68% yield).
LCMS (Method A): Rt = 1 .33 min, MH+ = 598.4. Intermediate 123: ethyl jV-(2-f4-r2-(1 ,3-dioxolan-2-yl)ethvn-2-pyridinylV6-fr(4- methylphenyl)sulfonyl1oxy)-4-pyrimidinyl)-B-alaninate
Figure imgf000091_0002
2-{4-[2-(1 ,3-dioxolan-2-yl)ethyl]-2-pyridinyl}-4,6-pyrimidinediyl bis(4-methylbenzenesulfonate) (3 x 1 .0 g, 1 .67 mmol and 1 x 857 mg, 1 .43 mmol) were dissolved in MeCN (3 x 15 mL and 1 x 12 mL, respectively) in 4 microwave vials equipped with stirrers. To each was added ethyl β-alaninate hydrochloride salt (3 x 265 mg, 1 .72 mmol and 1 x 227 mg, 1 .48 mmol, respectively) followed by triethylamine (3 x 688 μί, 4.94 mmol and 1 x 590 μί, 4.23 mmol, respectively). The vials were heated at 100 °C for 20 min using a microwave. The mixtures were combined together and concentrated under reduced pressure to give an orange viscous liquid. The liquid was purified by silica chromatography, eluting with 0 to 50% of a 10% solution of MeOH in EtOAc/EtOAc to give the title compound as a colourless waxy oil, 1 .4 g (40%).
LCMS (Method A): Rt = 0.93 min, MH+ = 543.3. Intermediate 124: ethyl Μ-Γ2-Γ4-Γ2-(1 ,3-dioxolan-2-yl)ethyl1-2-pyridinylV6-(1 ,2,4,5-tetrahydro-3H-
Figure imgf000092_0001
To a solution of ethyl Λ/-(2-{4-[2-(1 ,3-dioxolan-2-yl)ethyl]-2-pyridinyl}-6-{[(4- methylphenyl)sulfonyl]oxy}-4-pyrimidinyl)-p-alaninate (385 mg, 0.710 mmol) in isopropanol (12 mL) in a microwave vial was added 2,3,4,5-tetrahydro-1 /-/-3-benzazepine (522 mg, 3.55 mmol). The resultant solution was heated to 150 °C for 1 h. The reaction mixture was then diluted with EtOAc (10 mL) and concentrated in vacuo to give the crude product as a yellow oil. The crude product was purified by column chromatography using a gradient of 0% to 40% of a 20% solution of MeOH in DCM/DCM to give the title product as a yellow oil, 219 mg (60%).
LCMS (Method B): Rt = 1 .23 min, MH+ = 518.3.
Intermediate 125: ethyl jV-r2-r4-(3-oxopropyl)-2-pyridinvn-6-(1 ,2,4,5-tetrahvdro-3tf-3-
Figure imgf000092_0002
A solution of ethyl Λ/-[2-{4-[2-(1 ,3-dioxolan-2-yl)ethyl]-2-pyridinyl}-6-(1 ,2,4,5-tetrahydro-3H-3- benzazepin-3-yl)-4-pyrimidinyl]-p-alaninate (51 mg, 0.099 mmol) in Acetic Acid (1 .5 mL) / Water (0.375 mL) was heated to 80 °C and stirred for 16 h. The reaction mixture was diluted with DCM (20 mL) and then quenched by the addition of NaHC03 solution. The quenched reaction was stirred until the effervescence ceased and the layers then separated. The aqueous layer was extracted with further DCM (2 x 20 mL) and the combined organics washed sequentially with NaHC03 solution (20 mL) and then brine (20 mL). The organics were dried over Na2S04 then concentrated in vacuo. The crude product was re-dissolved in DCM (2 mL) and a small aliquot (~0.1 mL) removed and dried under a stream of N2 to allow an NMR to be taken. The remainder of the solution was concentrated to afford the crude title compound as a yellow-green oil, 43 mg, (92%) which was used directly in the subsequent reactions.
LCMS (Method B): Rt = 1 .18 min, MH+ = 474.2.
Intermediate 126: ethyl jV-r2-f4-r3-(methylamino)propyn-2-pyridinylV6-(1 ,2,4,5-tetrahydro-3tf-3-
Figure imgf000093_0001
To a solution of ethyl A/-[2-[4-(3-oxopropyl)-2-pyridinyl]-6-(1 ,2,4,5-tetrahydro-3/-/-3-benzazepin-3-yl)-4- pyrimidinyl]-p-alaninate (81 mg, 0.171 mmol) in THF (4 mL) was added MgS04 (515 mg, 4.28 mmol) and methanamine (0.428 mL, 0.855 mmol, 2M solution in THF). The mixture was stirred for 16 h and an aliquot removed. Sodium borohydride (6.47 mg, 0.171 mmol) was then added and the reaction mixture stirred at r.t. for 50 min. The reaction was quenched by the addition of water (20 mL) and DCM (20 mL). The layers were separated and the aqueous layer further extracted with DCM (2 x 20 mL). The combined organics were dried over Na2S04 then concentrated in vacuo to afford the crude product as a yellow oil. The crude product was taken up in Ethanol (2 mL), HCI (1 .0M in Et20) (0.2 mL, 0.200 mmol) was added and the reaction mixture allowed to stand for 1 h. A further portion of HCI (1 .0M in Et20) (0.4 mL, 0.400 mmol) was added and the reaction mixture allowed to stand for a further 30 min. The reaction was concentrated in vacuo to afford the crude product as a yellow oil. The crude product was purified by column chromatography using a gradient of 0 to 100% of a 20% solution of 2M NH3/MeOH in DCM)/DCM to give the title product as a yellow oil, 58 mg (69%).
LCMS (Method A): Rt = 0.74 min, MH+ = 489.4.
Intermediate 127: ethyl A 2-{4-r2-(1 ,3-dioxolan-2-yl)ethvn-2-pyridinylV6- r(phenylmethyl)amino1-4-pyrimidinyl)-B-alaninate
Figure imgf000094_0001
To a solution of ethyl Λ/-(2-{4-[2-(1 ,3-dioxolan-2-yl)ethyl]-2-pyridinyl}-6-{[(4-methylphenyl)sulfonyl]oxy}- 4-pyrimidinyl)-p-alaninate (285 mg, 0.525 mmol) in Isopropanol (10 ml_) in a microwave vial was added 1 -phenylmethanamine (0.287 ml_, 2.63 mmol). The resultant solution was heated to 150 °C in a microwave for 1 h. The reaction mixture was heated at 150 °C for a further 1 hr. The reaction mixture was then diluted with EtOAc (10ml_) and concentrated in vacuo to give the crude product as a yellow oil. The crude product was purified by silica chromatography using a gradient of 0 to 40% of a 20% solution of MeOH in DCM/DCM to give the impure product as a yellow oil. The product was further purified in two batches by high pH MDAP (Method E).
In a separate reaction: to a solution of ethyl Λ/-(2-{4-[2-(1 ,3-dioxolan-2-yl)ethyl]-2-pyridinyl}-6-{[(4- methylphenyl)sulfonyl]oxy}-4-pyrimidinyl)-p-alaninate (100 mg, 0.184 mmol) in Isopropanol (3 ml_) in a 2-5ml_ microwave vial was added 1 -phenylmethanamine (0.101 ml_, 0.921 mmol). The resultant solution was heated in a microwave to 150 °C for 1 hr and then at 150 °C for a further 1 h. The reaction mixture was then diluted with EtOAc (10 ml_) and concentrated in vacuo to give the crude product as a yellow oil. The crude product was purified by silica chromatography using a gradient of 0 to 40% of a 20% solution of MeOH in DCM/DCM to give the impure product as a yellow oil. Further purification was attempted using 0 to 100% of a 10% solution of MeOH in EtOAc/EtOAc. The same column was then eluted with 20 to 100% of a 20% solution of MeOH in DCM/DCM and finally the same column was eluted with 100% of a 20% solution of 2M NH3 in MeOH/DCM. All of the fractions containing UV active material were concentrated together and the resultant crude material (~60mg) further purified by high pH MDAP (Method E).
The appropriate fractions from both reactions were combined to afford the title compound as an off- white gum (96 mg, 28%).
LCMS (Method A): Rt = 0.92 min, MH+ = 478.3.
Intermediate 128: ethyl jV-f2-r4-(3-oxopropyl)-2-pyridinvn-6-r(phenylmethyl)amino1-4- pyrimidinylVB-alaninate O
A solution of ethyl Λ/-{2-{4-[2-(1 ,3-dioxolan-2-yl)ethyl]-2-pyridinyl}-6-[(phenylmethyl)amino]-4- pyrimidinyl}-p-alaninate (96 mg, 0.201 mmol) in Acetic Acid (3 ml_)/Water (0.750 ml_) was heated to 80 °C and stirred for 16 hr. The reaction mixture was diluted with dichloromethane (20 ml_) and then quenched by the addition of NaHC03 solution. The quenched reaction was stirred until the effervescense ceased and the layers then separated. The aqueous layer was extracted with further DCM (2 x 20 ml_) and the combined organics washed sequentially with NaHC03 solution (20 ml_) and then brine (20 ml_). The organics were dried over Na2S04 then concentrated in vacuo. The crude product was re-dissolved in DCM (2 ml_) and a small aliquot (~0.1 ml_) removed and dried under a stream of N2 to allow an NMR to be taken. The remainder of the solution was concentrated to afford the crude title product as a yellow-green oil, 87 mg (100 %) which was used directly in the next step without further purification.
LCMS (Method B): Rt = 1 .01 min, MH+ = 434.3.
Intermediate 129: ethyl AM 2-f4-r3-(methylamino)propyn-2-pyridinylV6-r(phenylmethyl)amino1-4- pyrimidinylVB-alaninate
Figure imgf000095_0001
To a solution of ethyl A/-{2-[4-(3-oxopropyl)-2-pyridinyl]-6-[(phenylmethyl)amino]-4-pyrimidinyl}-p- alaninate (42.5 mg, 0.098 mmol) in Tetrahydrofuran (THF) (2.3 ml_) was added magnesium sulfate (295 mg, 2.451 mmol) and methanamine (0.245 ml_, 0.490 mmol). The mixture was stirred for 16 h. Sodium borohydride (3.71 mg, 0.098 mmol) was then added and the reaction mixture stirred at r.t. for 45 min. The reaction was quenched by the addition of water (20 ml_) and DCM (20 ml_). The layers were separated and the aqueous layer further extracted with DCM (2 x 20 ml_). The combined organics were concentrated in vacuo to afford the crude product as a yellow oil. The crude product was taken up in Ethanol (1 ml_), HCI (1 .0M in Et20) (0.3 ml_, 0.300 mmol) was added and the reaction mixture allowed to stand for 1 h. The product was concentrated in vacuo to afford the crude product as a yellow oil. The crude product was purified by silica chromatography using a gradient of 0 to 100% of a 20% solution of 2M NH3 in MeOH/DCM to give the product as a yellow oil which still contained a ~10% impurity. The sample was further purified by high pH MDAP (Method E) to give the title product as a colourless gum (12 mg, 27%)
LCMS (Method A): Rt = 0.67 min, MH+ = 449.3.
Intermediate 130: ethyl jV-f 2-Γ4-ί3-^ r2-ff ff 1.1 - dimethylethyl)oxy1carbonyl)amino)ethyl1am
pyrimidinvD-B-alaninate
Figure imgf000096_0001
To a solution of ethyl A/-{2-[4-(3-oxopropyl)-2-pyridinyl]-6-[(phenylmethyl)amino]-4-pyrimidinyl}-p- alaninate (42.5 mg, 0.098 mmol) in Tetrahydrofuran (THF) (2.3 mL) was added magnesium sulfate (295 mg, 2.451 mmol) and 1 ,1 -dimethylethyl (2-aminoethyl)carbamate (79 mg, 0.490 mmol). The mixture was stirred for 16 h. Sodium borohydride (3.71 mg, 0.098 mmol) was added in 1 portion and the reaction stirred for 1 hr. The reaction was quenched by the addition of water (20 mL) and DCM (20 mL). The layers were separated and the aqueous layer further extracted with DCM (2 x 20 mL). The combined organics were dried (Na2S04) and concentrated in vacuo to afford the crude product as a yellow oil. The crude product was taken up in Ethanol (1 mL) and HCI (1 .0M in Et20) (0.3 mL, 0.300 mmol) added. The resultant solution was allowed to stand for 2 h and then concentrated in vacuo to afford the crude product as a yellow oil. The crude product was purified by silica chromatography using a gradient of 0 to 100% solution of 20% 2M NH3/MeOH in DCM/DCM to give the impure product as a yellow oil. The product was further purified by high pH MDAP (Method E). The appropriate fractions were combined together and concentrated in vacuo to afford the title product as a yellow gum, 19 mg (34%).
LCMS (Method A): Rt = 0.79 min, MH+ = 578.4. Intermediate 131 : ethyl jV-f2-(4-f3-r(2-aminoethyl)aminolpropylV2-pyridinyl)-6- r(phenylmethyl)amino1-4-pyrimidinyl)-B-alaninate
Figure imgf000097_0001
To a solution of ethyl Λ/-{2-[4-(3-{[2-({[(1 ,1 -dimethylethyl)oxy]carbonyl}amino)ethyl]amino}propyl)-2- pyridinyl]-6-[(phenylmethyl)amino]-4-pyrimidinyl}-p-alaninate (19 mg, 0.033 mmol) in Dichloromethane (DCM) (1 mL) at r.t. was added TFA (0.228 ml_, 2.96 mmol) The mixture was stirred for 1 hr. The reaction mixture was concentrated under reduced pressure to afford a yellow oil which was then dissolved in methanol and loaded onto an SCX cartridge (5g) and eluted with methanol. Product was eluted with 2M ammonia in methanol. The filtrate from the ammonia fractions was concentrated under reduced pressure to yield the title compound as a yellow oil (15 mg, 95 %). LCMS (Method B): Rt = 0.83 min, MH+ = 478.3.
Figure imgf000097_0002
Sodium Hydride (31 .8 mg, 0.796 mmol) was added to DMF (1 mL), followed by 3,4-dihydro-1 (2H)- isoquinolinone (107 mg, 0.730 mmol). The resulting suspension was stirred at r.t. for 5 min, then 4,6- dichloro-2-(2-pyridinyl)pyrimidine (150 mg, 0.664 mmol) was added and resulting mixture was stirred at r.t. for 45 min.
The mixture was partitioned between EtOAc and water then the aqueous layer extracted with DCM three times. The organic layers were combined, filtered throught a phase separator and volatiles removed under reduced pressure to afford 212 mg of a white-brown powder.The crude material was purified by column chromatography, eluting with a 0 to 100% EtOAc in cyclohexane, to give the title compound, 121 mg (54%) as an off white powder.
LCMS (Method A) Rt 1 .05 min, MH+=337.0
Intermediate 133: ethyl A/-r6-r3-(diethylamino)-1 -piperidinyl1-2-(2-pyridinyl)-4-pyrimidinyl1-B- alaninate
Figure imgf000098_0001
A stock solution of ethyl A/-[6-(3-amino-1 ^iperidinyl)-2-(2^yridinyl)-4^yrimidinyl]-p-alaninate (500 mg) in Methanol (48 ml_) was created. 12ml_ aliquots were split into 4 flasks equipped with stirrers, then Acetic Acid (0.200 ml_) was added to each vial. To one vial was added acetaldehyde (36mg, 0.810 mmol). Polymer supported cyanoborohydride, (270 mg, 0.540 mmol) was added and the mixtures were then stirred overnight at room temperature under an atmosphere of Nitrogen. All the reactions were filtered and concentrated under reduced pressure. The crude product was purified by column chromatography on silica, eluting with 0 to 5% of a 20% solution of 2M ammonia in methanol in DCM/DCM. The title compound was afforded as a yellow oil, 77 mg (49 %).
LCMS (Method A): Rt = 0.50 min, MH+ = 427.4 Intermediate 134: 4-chloro-2-(2-pyridi vdro-2-naphthalenyloxy)pyrimidine
Figure imgf000098_0002
1 ,2,3,4-tetrahydro-2-naphthalenol (164.0 mg, 1 .1 1 mmol) was added to a cold (0°C) solution of sodium hydride (48.7 mg, 1 .22 mmol) in A/,A/-dimethylformamide (10 ml_). After 30 min of stirring under nitrogen at 0 °C, 4,6-dichloro-2-(2-pyridinyl)pyrimidine (250.0 mg, 1 .1 1 mmol) was added. After 16 hr of stirring at r.t., the reaction mixture was cooled at 0 °C and sodium hydride (24.0 mg, 0.60 mmol) was added. After 2 hr of stirring under nitrogen at room temperature, water (50 ml_) was added to the reaction mixture, followed by EtOAc (50 ml_). The aqueous layer was further extracted with EtOAc (50 ml_). The combined organic layers was dried through a hydrophobic frit and concentrated to dryness to give a brown oil. The crude was purified by column chromatography eluting with a gradient from 0-50% of EtOAc-cyclohexane to give the impure product (278.0 mg). 168 mg of the impure product was dissolved in 1 :1 MeOH:DMSO (2 mL) and purified by MDAP (Method E). The solvent was evaporated in vacuo to give the title compound as a yellow oil, 64.2 mg (17%).
LCMS (Method A): Rt = 1 .25 min, MH+ 338.1 .
The compounds in the following table were similarly prepared:
Figure imgf000099_0001
Figure imgf000099_0002
Intermediate 140: ethyl jV-r6-fr(3,4-dichlorophenyl)methvnoxyV2-(2-pyridinyl)-4-pyrimidinvn-B- alaninate
Figure imgf000100_0001
To a solution of 4-chloro-6-{[(3,4-dichlorophenyl)methyl]oxy}-2-(2^yridinyl)pyrimidine (170 mg, 0.464 mmol) in Dimethyl Sulfoxide (DMSO) (1.5ml) in a microwave vial was added DIPEA (0.405 ml, 2.318 mmol) and ethyl β-alaninate hydrochloride (135 mg, 0.881 mmol).
The reaction mixture was heated to 150°C in a microwave for 3 hr. The sample was diluted with 1 :1 MeOH:DMSO and purified by MDAP (Method E) in 3 injections to give the title compound as a clear solid (58 mg, 0.130 mmol, 28 %).
LCMS (Method A) Rt 1.06 min, MH+=447
The following intermediates in the table were similarly prepared:
Figure imgf000100_0002
Figure imgf000100_0003
Example 1 : Af-r6-(1 ,1-dimethylethyl)-2-(2-pyridinyl)-4-pyrimidinvn-|3-alanine trifluoroacetate
Figure imgf000101_0001
To a solution of methyl Λ/-[6-(1 ,1 -dimethylethyl)-2-(2-pyridinyl)-4-pyrimidinyl]-p-alaninate (150mg, 0.48mmol) in a solvent mixture of tetrahydrofuran (20ml) and H20 (5 ml) was added lithium hydroxide (80.3mg, 1 .91 mmol) in one portion at room temperature. The reaction mixture was stirred at room temperature overnight. TLC (DCM/MeOH = 10/1) showed the reaction was complete. The mixture was evaporated and the residue was diluted with H20 (5ml). The mixture was acidified with 2. ON aq. HCI to pH ~ 4. The mixture was purified by prep HPLC to give the trifluoroacetate salt of the title compound as a white solid (60mg).
A portion of this was further purified by high pH MDAP (Method E) to give the title compound as a white solid, 19mg.
1 H NMR (400 MHz, DMSO-86, 300K) 8.70ppm (1 H, d, CH), 8.28ppm (1 H, d, CH), 7.90ppm (1 H, t, CH), 7.50-7.35ppm (2H, m, CH, NH), 6.46ppm (1 H, s, CH), 3.57ppm (2H, br s, CH2), 2.46ppm (2H, t, CH2), 1 .27ppm (9H, s, 3xCH3)
LCMS: Rt = 0.69 min, MH+ = 301 (Method A)
Example 2 :N-r2-(2-pyridinyl)-6-(trifluoromethyl)-4-pyrimidinyl1-B-alanine
Figure imgf000101_0002
Methyl N-[2-chloro-6-(trifluoromethyl)-4-pyrimidinyl]-p-alaninate (50 mg, 0.176 mmol) was dissolved in toluene (0.5 ml_) and the system degassed for 1 min. Bromo(2-pyridinyl)zinc (0.5M in THF) (2.1 15 ml_, 1 .058 mmol) was added and the resulting mixture degassed for a further 5min. Palladium (II) acetate (5.94 mg, 0.026 mmol) and S-Phos (21 .71 mg, 0.053 mmol) were added and the system degassed for 5min. The reaction mixture was then heated in the microwave at 100 °C for 1 hour. LCMS showed no starting material and a mixture of the methyl ester and hydrolysed carboxylic acid.
The reaction mixture was filtered through celite and washed with MeOH (3 x 10ml_). The combined organics were concentrated in vacuo and submitted to purification by high pH MDAP (Method E) to yield the title compound as a beige solid (12.5 mg). 1 H NMR (400 MHz, DMSO-d6, 300K) 8.73ppm (1 H, br s, CH), 8.30ppm (1 H, d, CH), 8.18ppm (1 H, br s, NH), 7.96ppm (1 H, t, CH), 7.52ppm (1 H, dd, CH), 6.95ppm (1 H, s, CH), 3.68ppm (2H, br s, CH2), 2.46ppm (2H, br s, CH2)
LCMS (Method A) : Rt = 0.60min, MH+ 313
General procedure for the preparation of bis N-linked pyrimidines:
Example 3: A -r6-(4-morpholinyl)-2-(2-pyridinyl)-4-pyrimidinyl1-B-alanine
Figure imgf000102_0001
Ethyl N-[6-(4-morpholinyl)-2-(2^yridinyl)-4-pyrimidinyl]-p-alaninate (191 mg, 0.534 mmol) was dissolved in 1 :1 tetrahydrofuran (THF) (2 ml_)/water (2.000 ml_). To this, lithium hydroxide (38.4 mg, 1 .603 mmol) was added and the reaction stirred at 25 °C for 1 .5 hour. The reaction was left stirring at room temperature overnight. 2M HCI dil. (0.802ml_) was added to the reaction, and volatiles were removed to give the crude product (227mg) as a yellow solid. This was dissolved in 1 :1 MeOH:DMSO (1 ml_) and purified by Mass Directed Auto Preparative LCMS under high pH elution conditions (Method E). The solvent was evaporated under vacuum to give A/-[6-(4-morpholinyl)-2-(2-pyridinyl)-4- pyrimidinyl]-p-alanine (145mg, 82%) as a cream solid.
1H NMR (400 MHz, DMSO-d6) 8.65ppm (1 H, d, CH), 8.24ppm (2H, d, CH), 7.86ppm (3H, dd, CH), 7.42ppm (1 H, dd, CH), 6.83ppm (1 H, br. s., NH), 5.68ppm (1 H, s, CH), 4.08-3.19 (10 H, m, CH2, obscured by water peak), 2.46 (2H, t, CH2).
LCMS: Rt = 0.55 min, MH+ = 330.18 (Method A)
Other examples indicated in following table were prepared similarly:
Figure imgf000102_0002
Figure imgf000103_0001
Figure imgf000104_0001
Figure imgf000105_0001
Figure imgf000106_0001
Figure imgf000107_0001
Figure imgf000108_0001
Figure imgf000109_0001
Figure imgf000110_0001
Figure imgf000111_0001
Example R1 Ar Yield % LCMS
124 * 64 Rt = 0.88 Λ/-[6-{[(3- chlorophenyl)methyl]oxy}- 2-(2-pyridinyl)-4- .- min, MH+
= 385.2 (M pyrimidinyl]-p-alanine ( CI ethod
B)
125 * 88 Rt = 0.88 Λ/-[6-[(2- min, MH+ naphthalenylmethyhoxy]- = 401 .2 2-(2-pyridinyl)-4- (Method pyrimidinyl]-p-alanine A)
Example 42, iv-r2-(2-pyridinyl)-6-(1 ,2,4,5-tetrahvdro-3tf-3-benzazepin-3-yl)-4-pyrimidinvn-B- alanine
Figure imgf000112_0001
To a solution of A/-[6-chloro-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine (100 mg, 0.359 mmol) in Dimethyl Sulfoxide (DMSO) (1 mL) in a microwave vial were added DIPEA (0.188 ml_, 1 .076 mmol) and 2,3,4, 5-tetrahydro-1 H-3-benzazepine (52.8 mg, 0.359 mmol).
The system was heated to 150°C in a Biotage initiator microwave for 1 hour. The reaction had gone to completion. 1 ml of MeOH was added and the reaction purified by formic MDAP (Method G). For product fractions, solvent was removed in the blow down apparatus to give product.
This was dissolved in IPA and loaded onto an SCX cartridge, which was previously conditioned with IPA. This was washed with IPA and then product eluted using 10% ammonia in IPA. The appropriate fractions were combined and the solvent removed in the nitrogen blow down apparatus, to give the title compound as a yellow solid, 120mg (86%). 1H NMR (400 MHz, DMSO-d6) 8.67ppm (1 H, d, CH), 8.25ppm (1 H, d, CH), 7.89ppm (1 H, dd, CH), 7.43ppm (1 H, dd, CH), 7.20 - 7.04ppm (4H, m, CH), 6.82ppm (1 H, br. s., NH), 5.73ppm (1 H, s, CH), 3.93-3.73ppm (4H, m, CH2), 3.62-3.40ppm (4H, m, CH2, obscured by water peak), 2.99-2.86 ppm (4H,m, CH2).
LCMS (Method A): Rt = 0.86 min, MH+ 390.3
Further examples indicated in following table were prepared similarly from their respective ethyl- or methyl- esters, and in some cases using 1 ,4-dioxane in the place of DMSO:
Figure imgf000113_0001
Figure imgf000113_0002
Figure imgf000114_0001
Figure imgf000115_0001
A stock solution of ethyl A/-[6-chloro-2-(2-pyridinyl)-4-pyrimidinyl]-p-alaninate (300mg, 0.978 mmol) in DMSO (3ml) was created with the aid of a heat gun. A 1 ml aliquot (0.326mmol) was put into a microwave vial equipped with a stirrer then DIPEA (1 10ul) added, followed by /V-methyl-1 - phenylmethanamine (0.360 mmol). The mixtures were then heated at 150°C for 1 hr using a microwave. The DMSO solution was then purified directly by MDAP (Method I) and fractions containing desired product combined then concentrated under reduced pressure to give ethyl N-[6- chloro-2-(2-pyridinyl)-4-pyrimidinyl]-p-alaninate.
LiOH (2.6g, 0.109mmol) was dissolved in 200ml 1 :1 :1 THF/MeOH/water then a 2ml aliquot of this solution added to a vial containing ethyl A/-[6-chloro-2-(2-pyridinyl)-4-pyrimidinyl]-p-alaninate
(0.255mmol). The mixture was stirred for 2h at room temperature then concentrated under a stream of nitrogen. The reaction mixture was neutralised using 2N HCI (aq) then diluted with IPA (around 10ml) then loaded onto a SCX-SPE column. The SPE column was washed with IPA, then product eluted with 10% ammonia in MeOH. Fractions containing the desired product were combined then concentrated under reduced pressure to give the title compound as a white solid, 56.6mg (61 %).
LCMS (Method A): Rt = 0.74 min, MH+ 364.3
Further examples indicated in following table were prepared similarly:
Figure imgf000116_0001
Figure imgf000117_0001
Example 49: A -r6-(met yloxy)-2-(2-pyridinyl)-4-pyrimidinyl1-B-alanine
Figure imgf000117_0002
Ethyl /V-[6-chloro-2-(2-pyridinyl)-4-pyrimidinyl]-^-alaninate (100 mg, 0.326 mmol) was dissolved in MeOH (2 ml) then sodium methoxide in MeOH (25%, 0.21 1 ml, 0.977 mmol) was added and the reaction mixture stirred at 50 °C overnight. Methanol (2 ml) was added then the resulting suspension concentrated under reduced pressure. DMSO (3 ml) was added and the resulting solution transferred to a microwave vial and further sodium methoxide in methanol (25%, 0.21 1 ml, 0.977 mmol) was added. The reaction mixture was then heated to 80°C for 1 hour. LCMS showed that the reaction had gone to completion. The reaction mixture was cooled to room temperature, directly pipetted into four vials and purified by MDAP (Method G) to give the title compound as a yellow solid, 37mg (41 %).
LCMS (Method A): Rt = 0.48 min, MH+ 275.1
Example 60: A -r6-(4-acetyl-1 -piperazinyl)-2-(2-pyridinyl)-4-pyrimidinyl1-B-alanine
Figure imgf000118_0001
Acetyl chloride (0.02 mL, 0.21 mmol) was added to a suspension of ethyl A/-[6-(1 -piperazinyl)-2-(2- pyridinyl)-4-pyrimidinyl]-p-alaninate (67 mg, 0.19 mmol) in DCM (3 mL). The reaction mixture was stirred at r.t. for 18 hr then washed with water (3 mL) and the layers were separated using a hydrophobic frit. The aqueous layer was loaded on a 5 g SCX cartridge that had been preconditioned with methanol, washed with methanol and product was eluted with methanolic ammonia (2M). The ammonia containing fractions were combined, mixed with the organic layer from the aqueous wash and evaporated under reduced pressure. The residue was taken up in 1 :1 water/THF solution (4 mL) and treated with lithium hydroxide monohydrate (15.8 mg, 0.38 mmol). After 2 hr of stirring at r.t., the reaction mixture was concentrated using the blow down unit. The residue was dissolved in 1 :1 MeOH:DMSO (1 mL) and purified by MDAP (Method E). The appropriate fractions were combined and evaporated under reduced pressure to give the title compound as a yellow solid, 37 mg (53%.
LCMS (Method A): Rt = 0.49 min, MH 371 .2
Other examples indicated in the following table were prepared similarly:
Figure imgf000118_0002
Example R1 Yield % LCMS Example R1 Yield % LCMS
62 38 Rt = 0.50 min,
A/-[6-[3-(acetylamino)-1 - MH+ = 371.2 pyrrolidinyl]-2-(2-pyridinyl)-4- * (Method A) pyrimidinyl]-p-alanine
65 19 Rt = 0.54 min,
Λ/-[6- [3- (a cety I a m i n o)- 1 - pi pe rid i n y I]- 2-(2-pyridinyl)-4-pyrimidinyl]-p- AH MH+ = 385.1
(Method A) alanine
*
Example 61 , A -r6-r4-(met ylsulfonyl)-1 -piperazinyl1-2-(2-pyridinyl)-4-pyrimidinyl1-B-alanine
Figure imgf000119_0001
Methanesulfonyl chloride (0.02 mL, 0.21 mmol) was added to a suspension of ethyl Λ/-[6-(1- piperazinyl)-2-(2-pyridinyl)-4-pyrimidinyl]-p-alaninate (67 mg, 0.188 mmol) in DCM (3 mL). The reaction mixture was stirred at r.t. for 18 hr then washed with water (3 mL) and the layers were separated using a hydrophobic frit. The aqueous layer was loaded onto a 5 g SCX cartridge that had been conditioned with methanol, washed with methanol and product was eluted with methanolic ammonia (2M). The ammonia containing fractions were combined, mixed with the organic layer from the aqueous wash and evaporated under reduced pressure. The residue was taken up in 1 :1 water/THF solution (4 mL) and treated with lithium hydroxide monohydrate (15.8 mg, 0.38 mmol). After 2 hr of stirring at r.t., the reaction mixture was concentrated under a stream of nitrogen. The residue was dissolved in 1 :1 MeOH:DMSO (1 mL) and purified by MDAP (Method E). The appropriate fractions were combined and evaporated under reduced pressure to give the title compound as a white solid, 25 mg (33%).
LCMS (Method A): Rt = 0.56 min, MH+ 407.2
Other examples indicated in the following table were prepared similarly:
Figure imgf000120_0002
Figure imgf000120_0001
β-alanine (66.1 mg, 0.74 mmol) and DIPEA (0.26 mL, 1 .49 mmol) were added to a solution of 4- chloro-6-[(phenylmethyl)oxy]-2-(2-pyridinyl)pyrimidine (221 .0 mg, 0.74 mmol) in DMSO (0.5 mL) in a microwave vial. The reaction mixture was heated to 150 °C in a microwave for 2 hr. The reaction mixture was diluted with MeOH (3 mL) and purified by MDAP (Method E). The appropriate fractions were combined and evaporated under reduced pressure to give the impure product. The residue was dissolved in 1 :1 MeOH:DMSO (1 mL) and purified by MDAP (Method G). The appropriate fractions were loaded onto a hydrogen carbonate column (previously conditioned with MeOH).The product was eluted with HCI in MeOH (1 M). The solvent was removed under reduced pressure. The residue which contained methyl ester product was dissolved with 1 :1 THF/water (2 mL) and treated with lithium hydroxide monohydrate (62.3 mg, 1 .49 mmol). After 2 hr of stirring at r.t., the solvent was
concentrated under reduced pressure. The residue was dissolved in a mixture of 1 :1 MeOH:DMSO (1 .5 mL) and HCI (2M) (0.5 mL) and purified by MDAP (Method E). The solvent was evaporated under reduced pressure to give the title compound as a white solid, 25 mg (10%). LCMS (Method A): Rt = 0.72 min, MH+ 351 .2.
Example 98, A -r2-(2-pyridinyl)-6-(1 ,2,3,4-tetra vdro-2-nap t alenyloxy)-4-pyrimidinyl1-B-alanine
Figure imgf000121_0001
DIPEA (0.10 ml, 0.57 mmol) and β-alanine (20.3 mg, 0.23 mmol) were added to a solution of 4-chloro- 2-(2-pyridinyl)-6-(1 ,2,3,4-tetrahydro-2-naphthalenyloxy)pyrimidine (64.0 mg, 0.19 mmol) in DMSO (0.5 ml_) in a microwave vial. The reaction mixture was heated to 150 °C in a microwave for 2 hr, then β- alanine (10.0 mg, 0.03 mmol) and DIPEA (66.0 μΙ_, 0.38 mmol) were added to the reaction mixture which was heated to 150 °C in a microwave for a further 1 hr. The reaction mixture was diluted with 1 :1 MeOH:DMSO (2 mL) and purified by MDAP (Method E). The solvent was evaporated under reduced pressure to give the impure product which was dissolved in 1 :1 MeOH:DMSO (1 mL) and purified by MDAP (Method G). The solvent was evaporated under reduced pressure to give a solid which was taken up in ethanol, loaded on a 2 g SCX column, washed with ethanol and eluted with 20% ammonium hydroxide in ethanol. The appropriate fractions were combined and evaporated under reduced pressure to give the title compound as a yellow solid, 28 mg (38 %).
LCMS (Method A): Rt = 0.88, MH+ 391 .2.
The example indicated in the following table was prepared similarly:
Figure imgf000121_0002
Example R1 Yield % LCMS Example R1 Yield % LCMS
93 30 Rt = 0.76 min,
A/-[6-[(2-phenylethyl)oxy]-2-(2- MH+ = 365.1 pyridinyl)-4-pyrimidinyl]-p-alanine (Method B)
Example 100: Λ -Γ6-(1 -benzot ien-3-yl)-2-(2-pyridinyl)-4-pyrimidinyl1-B-alanine
Figure imgf000122_0001
DIPEA (0.07 ml, 0.42 mmol) and β-alanine (14.9 mg, 0.17 mmol) were added to a solution of 4-(1 - benzothien-3-yl)-6-chloro-2-(2-pyridinyl)pyrimidine (45.0 mg, 0.14 mmol) in DMSO (0.5 ml_) in a microwave vial. The reaction mixture was heated to 150 °C for 2 hr using a microwave. The reaction mixture was diluted with 1 :1 MeOH:DMSO (2 mL) and purified by MDAP (Method E). The solvent was evaporated under reduced pressure to give the impure product, which was dissolved in 1 :1
MeOH:DMSO (1 mL) and purified by MDAP (Method F). The solvent was evaporated under reduced pressure to give a yellow solid, which was taken up in ethanol, loaded on a 2 g SCX-SPE column, washed with ethanol and eluted with 20% ammonium hydroxide in ethanol. The appropriate fractions were combined and evaporated under reduced pressure to give the title compound as a yellow solid, 28 mg (54%).
LCMS (Method A): Rt = 0.79 min, MH+ 377.2.
Other examples in the following table were prepared similarly:
Figure imgf000122_0002
Example R1 Ar Yield % LCMS Example R1 Ar Yield % LCMS
50 tBu * 51 Rt = 0.78 min,
A/-[6-(1 ,1 -dimethylethyl)-2- MH+ = 351 (3-isoquinolinyl)-4- (Method A) pyrimidinyl]-p-alanine
51 tBu 33 Rt = 0.79 min,
A/-{6-(1 ,1 -dimethylethyl)-2- MH+ = 369
[5-(trifluoromethyl)-2- (Method C) pyridinyl]-4-pyrimidinyl}-p- F c— — F
F
alanine
52 tBu 3 Rt = 0.73 min,
A/-[6-(1 ,1 -dimethylethyl)-2- MH+ = 315 (4-methyl-2-pyridinyl)-4- (Method C) pyrimidinyl]-p-alanine
53 tBu * 9 Rt = 0.72 min,
MH+ = 331
A/-{6-(1 ,1 -dimethylethyl)-2- (Method B)
[4-(methyloxy)-2-pyridinyl]-4- pyrimidinyl}-p-alanine
54 tBu 43 Rt = 0.70 min,
MH+ = 331 .1
(Method A)
Λ/-{6-(1 ,1 - dimethylethyl)-2-[5-
OMe
(methyloxy)-2- pyridinyl]-4-pyrimidinyl}- β-alanine
55 tBu 26 Rt = 0.70 min,
A/-[6-(1 ,1 -dimethylethyl)-2- (5-methyl-2-pyridinyl)-4- J MH+ = 315.2
J (Method A) pyrimidinyl]-p-alanine
Figure imgf000124_0001
Example 101 : Af-(6-(1 ,3-dihvdro-2H-isoindol-2-yl)-2-f4-r(phenylamino)methvn-2-pyridinyl>-4- pyrimidinvD-B-alanine
Figure imgf000124_0002
A mixture of methyl Λ/-(6-(1 ,3-dihydro-2/-/-isoindol-2-yl)-2-{4-[(phenylamino)methyl]-2-pyridinyl}-4- pyrimidinyl)-p-alaninate (103 mg, 0.17 mmol) and KOH (40% by wt, 20ml) in MeOH (10 ml) was refluxed for 2h then material purified by prep-HPLC (Method J) to give the title compound as a solid, 46 mg. LCMS (Method H) Rt 1 .41 min, MH+ = 467.2
Figure imgf000124_0003
To a solution of 1 ,1 -dimethylethyl A/-[6-[(methylamino)carbonyl]-2-(2-pyridinyl)-4-pyrimidinyl]-p- alaninate (5 mg, 0.014 mmol) in Tetrahydrofuran (THF) (0.5 mL) was added HCI (0.168 mL, 0.839 mmol, 5M). The reaction was stirred at room temperature overnight. Another 0.1 mL of 5M HCI was added to the reaction mixture which was stirred for a further 5 h. The reaction mixture was heated to 50 °C and was stirred overnight. The volatiles were removed in vacuum and the residue was purified by MDAP, (Method E) to give the title compound (3.6 mg, 0.012 mmol, 85 % yield) as a white solid. LCMS (Method B): Rt = 0.47 min, MH+ = 302.7
Example 117, jV-r2-f4-r(2-hvdroxyethyl)amino1-2-pyridinylV6-(1 ,2,4,5-tetrahydro-3tf-3- benzazepin-3-yl)-4-pyrimidinyl1-B-alanine
Figure imgf000125_0001
Lithium hydroxide monohydrate (17.3 mg, 0.41 mmol) was added to a solution of 2-({[2-({2-[4-[(2- carboxyethyl)amino]-6-(1 ,2,4,5-tetrahydro-3/-/-3-benzazepin-3-yl)-2-pyrimidinyl]-4- pyridinyl}oxy)ethyl]amino}carbonyl)benzoic acid (250.0 mg, 0.41 mmol) in 1 :3 water/THF solution (4 ml). After 40 hr of stirring at r.t. (the solvent evaporated during that time), the residue was then taken up in water (5 ml) then acidified with HCI (2M) until pH 2. The solid was filtered off, washed with water and dried in a vacuum oven for 5 hr to give a yellow solid, 50 mg.
Hydrazine hydrate (0.02 ml_, 0.32 mmol) was added to a solution of the solid in a mixture of 1 ,4- dioxane (1 .0 ml) and /V,/V-dimethylform amide (0.40 ml). Methanol (0.40 ml) was added and the reaction mixture was stirred at r.t. for 24 hr. Hydrazine hydrate (0.02 ml, 0.32 mmol) was then added to the reaction mixture which was stirred at 80 °C for 5 hr. The reaction mixture was concentrated under reduced pressure and the residue purified by column chromatography, eluting with 0-80%
DCM-methanolic ammonia (2M). Appropriate fractions were combined and evaporated under reduced pressure to give the impure product which was further purified by MDAP (Method E) to give the title compound as a yellow oil, 5.5 mg (15%). LCMS (Method A): Rt = 0.80 min, MH+ 449.2.
For the avoidance of doubt, the present application does not include an Example 27, an Example 42, an Example 48, an Example 58, an Example 59, an Example 69, an Example 70, an Example 71 , an Example 72, an Example 75, an Example 84, an Example 105, an Example 108, or an Example 1 16.
JMJD3 MALDI-TOF Demethylase Assay Protocol The JMJD3 MALDI-TOF mass spectrometric assay monitors demethylation of a histone H3 peptide containing tri-methylated K27, by recombinant Jumonji D3 demethylase enzyme.
Compound plate preparation:
Compounds were diluted to 10mM in DMSO, and a 1 :3, 1 1 point serial dilution performed across a 384 well hibase plate (Greiner Bio-one, Stonehouse, UK). 10Onl of this dilution series was then transferred into a 384 well V-base plate (Greiner Bio-one) using the Echo™ acoustic dispenser (Labcyte Inc, Sunnyvale, CA, USA) to create the assay plate. 10Onl DMSO was added to column 6 to generate a high control.
Assay method:
10ul of a 1 % trifluoroacetic acid (TFA)solution was added to column 18 of compound plates to generate a 100% inhibition control, before 5ul of an enzyme solution containing 1 .2uM 6H-Tev-Flag- JMJD3 (Biological Reagents department, GSK, UK) and 200uM CHAPS (Sigma-Aldrich, St. Louis, MO) diluted in a buffer of 20mM Tris HCL and 150mM NaCI (both Sigma-Aldrich) was added to each well of the plate using a Multidrop Combi® dispenser (Thermo Fisher Scientific, Waltham, MA, USA). This gives a final assay concentration of 600nM JMJD3 enzyme and 100uM CHAPS.
Plates were incubated for 15 minutes at room temperature, after which time 5ul of a substrate solution containing 4mM ascorbic acid (Sigma -Aldrich), 100uM ammonium iron (II) sulphate (Fisher Scientific Waltham, MA, USA), 70uM alphaketoglutarate (Sigma-Aldrich) and 30uM H3K27Me3 peptide (Molecular Tools group, GSK, UK), all diluted in a buffer of 50mM Hepes, was added to each well of the plate using a Multidrop Combi® dispenser, to initiate the reaction. Final assay concentrations are 2mM ascorbic acid, 50uM ammonium iron (II) sulphate, 35uM alphaketoglutarate and 15uM H3K27Me3 peptide. After precisely 8 minutes, the reaction was quenched by the addition of 10ul of a 1 % TFA solution to all wells except column 18 (to which this was previously added) using a Multidrop Combi ©dispenser.
Samples were diluted 1 :100 in 5% acetonitrile using the Biomek® FX (Beckman Coulter, Brea, CA, USA) and 1 ul of this dilution transferred on to a 384 well PAC target (Bruker Daltonics, Bremen, Germany) again using the Biomek® FX. The target was washed by immersing in a solution of 10mM ammonium phosphate (Sigma-Aldrich) and 0.1 % TFA for 10 seconds before being dried at room temperature and analysed by MALDI-TOF (Ultraflex III, Bruker Daltonics) to generate relative peak areas of H3K27Me3 substrate and H3K27Me2 product. JMJD3 RapidFire™ Mass Spectromety Assay Protocol
The JMJD3 RapidFire™ mass spectromety assay monitors demethylation of a histone H3 peptide containing tri-methylated K27, by recombinant Jumonji D3 family demethylase enzyme.
Materials Ascorbic acid.a-ketoglutarate, dimethyl sulfoxide (DMSO), 4-(2-hydroxyethyl)-1 - piperazineethanesulfonic acid (Hepes), 3-[(3-Cholamidopropyl)dimethylammonio]-1 -propanesulfonate (CHAPS) and bovine serum albumin (BSA) were purchased from Sigma-Aldrich (Gillingham, Dorset, UK). Trifluoroacetic acid (TFA) was from Applied Biosystems (Warrington, UK). Acetonitrile and ammonium iron (II) sulphate were purchased from Fisher Scientific (Loughborough, UK). H3K27-Me3 peptide was synthesized and HPLC-purified to >95% purity by Cambridge Research Biochemicals (Cambridge, UK).
Expression and purification of JMJD3
JMJD3 DNA encoding the catalytic domain (residues 1 141-1682), and lacking residues 1637-1675 (a splice variant deletion; GenBank BC009994) was cloned into the pFB-HTb vector (Invitrogen) using BamHI and Xhol restriction sites, which encodes for a protein with an N-terminal 6His tag followed by a TEV-protease cleavage site. pFB1 -HTb-JMJD3-6H 1 141 -1682 (del 1637-1675) was transposed into the baculovirus genome using the BAC-to-BAC technology (Invitrogen). Bacmid DNA was transfected into Spodoptera frugiperda (Sf9) cells using Cellfectin II (Invitrogen), and expression was performed at a 1 L scale in Excel 420 media (SAFC Biosciences). The culture, at a cell concentration of 3.8xe6 cells/ml, was infected with P1 recombinant Baculovirus at a nominal multiplicity of infection of 3 and incubated for 48 hours. The cells were removed from the media by centrifugation at 2500g for 20 minutes, and the cell pellet was frozen for subsequent purification.
The pellet from the Baculovirus culture was resuspended in buffer A (20mM Tris pH 8.5, 300mM NaCI, 10% glycerol, 1 ul/ml Protease Inhibitor Cocktail Set III (Calbiochem 539134)).
Cells were lysed by Dounce Homogenisation, on ice, and centrifuged at 100,000 x g for 90 minutes at 4°C. The 100,000 x g supernatant was applied to a HisTRAP HP Column (GE Healthcare 17-5248- 02). The column was washed with ten column volumes of buffer A, followed by ten column volumes of buffer A containing 20mM Imidazole. Bound protein was eluted from the column using a linear gradient of 20-250mM Imidazole over twenty column volumes. The JMJD3 protein was eluted between 100mM and 200mM Imidazole. Eluted JMJD3 protein from the HisTRAP column was concentrated fourfold (Amicon Ultrafree-15 30kDa, Millipore UFC903024) and loaded onto a HiLoad 26/20 Superdex 200 prep grade size exclusion column (GE Healthcare 17-1069-01), equilibrated with buffer B (20mM Tris pH 8.0, 150mM NaCI, 5% glycerol, 0.5mM TCEP, 2mM a-Ketoglutaric acid sodium salt (Sigma K1875) and 10uM Fe(NH4)2(S04)2). Fractions containing JMJD3 were pooled and concentrated (Ultrafree-15 30kDa). Protein identity was confirmed by peptide mass fingerprinting and predicted molecular weight confirmed by mass spectrometry.
Peptide Synthesis
Peptide sequence : ATKAARKSAPATGGVKKPHRYRPG (the bold underlined lysine corresponds to K27 in human histone 3 and is tri-methylated): ATKAAR-K(TriMe)-SAPATGGVKKPHRYRPG The peptide was supplied by Cambridge Research Biochemicals. In detail, the protected peptide was assembled on a solid-phase synthesiser using preloaded Wang resin and utilising standard Fmoc synthesis protocols. 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 3 hours 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 molecular weight (mw) (by MALDiTOF mass spectroscopy) were pooled and freeze dried. The final material was analysed by HPLC and MALDiTOF mass spectroscopy.
The trimethyl lysine was incorporated into the sequence during assembly as Fmoc-Lys(TriMe)-OH. This was made by reacting a suitably protected alpha-nitrogen lysine with an excess of methyl chloride or bromide to form the quaternary salt. Preparation of compound screening plates
For dose-response curves, 3-fold serial dilutions were prepared from 10 mM compound solutions in DMSO across 384 well HiBase plates (Greiner Bio-one, Stonehouse, UK). 10Onl of this dilution series was transferred into 384 well V base assay plates (Greiner Bio-one, Stonehouse, UK) giving a concentration range between 100 μΜ and 1 .7 nM. Columns 6 and 18 of the assay plates were reserved for the high and low controls, respectively. The high controls had 100 nL of DMSO, but no compound, while the low controls also had 100 nL of DMSO but the enzyme was acid-inactivated by pretreatment with 0.5% (v/v) TFA. Compounds and DMSO were dispensed using the Echo™ acoustic dispenser (Labcyte Inc, Sunnyvale, CA, USA). RapidFire® mass spectrometry assay.
In this assay, disappearance of the trimethylated peptide substrate and formation of the dimethylated product are both monitored by mass spectrometry. Assays were performed by initially dispensing 5 μΐ of a 2x solution containing 0.3 μΜ JMJD3, 0.5 mg/mL BSA and 200μΜ CHAPS in 50 mM Hepes pH 7.0 into the 384-well plates containing 100 nL compound. Plates were allowed to incubate for 10 minutes at ambient temperature before reactions were initiated by the addition of 5 μί of a 2χ substrate solution containing 100 μΜ ascorbate, 100 μΜ Fe2+, 20 μΜ -ketoglutarate, and 60 μΜ H3K27(me3) peptide also in 50 mM Hepes pH 7.0. Plates were centrifuged at 1000 rpm for 1 minute and the reactions incubated at room temperature for 6 minutes, before being quenched by the addition of 30 μL· of a 0.5% (w/v) TFA solution. For the enzyme-inactivated control wells in column 18, the TFA solution was dispensed prior to addition of the enzyme solution. Plates were centrifuged at 1000 rpm for 5 minutes before analysis. All solutions were dispensed using a Multidrop Combi® dispenser (Thermo Fisher Scientific, Waltham, MA, USA). Assay plates were transferred onto a RapidFire200 integrated autosampler/solid phase extraction (SPE) system (Agilent Technologies Inc., Wakefield, MA, USA) coupled to an API4000 triple quadrupole mass spectrometer (Applied Biosystems, Concord, Ontario, Canada). Solvent A was water containing 0.01 % (v/v) TFA and 0.09% (v/v) formic acid. Solvent B was acetonitrile/water (8:2, v/v) containing 0.01 % (v/v) TFA and 0.09% (v/v) formic acid. Samples were aspirated under vacuum directly from 384-well assay plates for 500 ms. The sample was then loaded onto a C4 solid phase extraction cartridge to remove non-volatile buffer salts, using solvent A at a flow rate of 1 .5 mL/min for 3 s. The retained analytes were eluted to the mass spectrometer by washing the cartridge with solvent B at 1 .25 mL/min for 3 s. The cartridge was re-equilibrated with solvent A for 500 ms at 1 .5 mL/min. The entire sampling cycle was approximately 7 seconds per well, enabling analysis of a 384 well plate in approximately 45 minutes.
The mass spectrometer was operated in positive electrospray MRM mode. MRM transitions (Q1/Q3) for each species were as follows. H3K27Me3: 418.3/515.0; H3K27Me2: 414.8/519.7. A dwell time of 50 ms was used for all of the MRM transitions. The mass spectrometer was operated with a spray voltage of 3500 V and at a source temperature of 650 °C. The peaks detected by mass spectrometry were approximately 1 .2 s wide at half-height and they were integrated and processed using the RapidFire® peak integration software.
Data analysis
Data were analyzed using the ActivityBase Suite (ID Business Solutions Ltd, Surrey, UK). The extent of enzymatic turnover of the tri-methylated substrate to di-methylated product was expressed as percent conversion as shown in Eq. (1):
% Conversion = 100 χ Me2/(Me2 + Me3) (1) where Me2 and Me3 represent the integrated peak areas of the di- and trimethylated peptides. Data were then fitted to Eq. (2) to determine lc50 values
% Inhibition = (a - d)/[1 + ([\]/\C50)h] + d (2) where a is the uninhibited value, d is the fully inhibited value, [I] is the inhibitor concentration, IC50 is [I] that gives ½ χ (a - d), and h is the Hill slope. The compounds of examples 1 -125 have tested on at least one occasion with an IC50 value of < 100 μΜ in either the JmjD3 MALDI-TOF assay or the JmjD3 RapidFire™ assay, or both.
The esters intermediates were also found to be but weakly active inhibitors of JmjD3. Esters have a higher cell penetration than the corresponding acids so may offer a pro-drug approach to achieving cell activity.

Claims

Claims
1 . A pharmaceutical composition comprising a compound of formula (I)
Figure imgf000130_0001
(I)
wherein
R1 is:
• C1-6 alkyl;
• C3.7 cycloalkyl;
• C1-6 haloalkyl;
• a 5, 6 or 7-membered aryl or heteroaryl (which heteroaryl contains one or more heteroatoms selected from N, O and S and which is optionally fused to phenyl), said 5, 6 or 7- membered aryl or heteroaryl being optionally substituted with one or more substituents independently selected from C1-3alkyl;
• 0-C1-6alkyl (which is optionally substituted by phenyl or naphthyl, each of which may be substituted by one of more substituents independently selected from halo);
• O-cyclohexyl (which is optionally fused with phenyl);
C(0)NRc 2;
or
NRaRb, each Ra and Rb is independently selected from:
H;
• C1-3alkyl which is optionally substituted by one or more substituents independently selected from phenyl (which phenyl is optionally substituted by one or more substituents independently selected from C1-3alkyl, 0-C1-3alkyl, C(0)NR°2, halo and cyano), C(0)NR°2, a 4, 5, 6 or 7-membered heterocyclic or heteroaryl group (containing one or more heteroatoms independently selected from N, O and, S), a 3, 4, 5, 6 or 7-membered cycloalkyl group (which is optionally fused to phenyl), halo, OC1-3alkyl, OH, -NHCOC1-3alkylNRc 2 and C(0)NHCH2C(0)NRc 2; • a 3, 4, 5, 6 or 7-membered cycloalkyl group (which is optionally fused to phenyl), or
• Ra and Rb together form a 5, 6 or 7-membered heterocyclic group optionally containing one or more further heteroatoms independently selected from N, O, S or S(0)2 said heterocyclic group being optionally fused to a 5, 6 or 7-membered aryl or heteroaryl ring containing one or more heteroatoms independently selected from N, O and S; the heterocylic ring and/or the aryl or heteroaryl to which it is optionally fused being optionally substituted by one or more substituents independently selected from halo, OH, C1-3alkyl, 0-C1-3alkyl, C(0)C1-3alkyl, S(0)2C1.3alkyl, NHC(0)C1-3alkyl, NHS(0)2C1.3alkyl, C(0)NRc 2, C(0)NRd 2 (wherein Rd and Rd together form a 5 or 6-membered heterocylic ring), NR°2 C(0)phenyl, S(0)2NR°2, =0 (oxo) and 5, 6 or 7-membered aryl or heteroaryl (containing one or more heteroatoms independently selected from N, O and S);
R2 and R3 are each independently selected from:
H,
(CH^NR^CH^NR^,
(CH2)1-eNRc 2;
• C1-3 alkyl;
• 0-C1-3alkyl;
• C1-3haloalkyl;
• (CH2)o-3NRaRb (wherein Ra and Rb are as defined above);
(CH2)0_3NHPh;
(CH2)0.3OPh;
(CH2)0-3Ph;
or R2 and R3 together form a fused phenyl ring, and each R° is independently selected from hydrogen and C1-3alkyl or a pharmaceutically acceptable salt thereof and one or more of pharmaceutically acceptable carriers, diluents and excipients.
A compound of formula (I):
Figure imgf000132_0001
wherein
R1 is:
• C1-6 alkyl;
• C3.7 cycloalkyl;
• C1-6 haloalkyl;
• a 5, 6 or 7-membered aryl or heteroaryl (which heteroaryl contains one or more heteroatoms selected from N, O and S and which is optionally fused to phenyl), said 5, 6 or 7- membered aryl or heteroaryl being optionally substituted with one or more substituents independently selected from C1-3alkyl;
• O-C^alkyl (which is optionally substituted by phenyl or naphthyl, each of which may be substituted by one of more substituents independently selected from halo);
• O-cyclohexyl (which is optionally fused with phenyl);
C(0)NRc 2;
or
NRaRb, each Ra and Rb is independently selected from:
H;
• C^alkyl which is optionally substituted by one or more substituents independently selected from phenyl (which phenyl is optionally substituted by one or more substituents independently selected from C1-3alkyl, 0-C1-3alkyl, C(0)NR°2, halo and cyano), C(0)NR°2, a 4, 5, 6 or 7-membered heterocyclic or heteroaryl group (containing one or more heteroatoms independently selected from N, O and, S), a 3, 4, 5, 6 or 7-membered cycloalkyl group (which is optionally fused to phenyl), halo, OC1-3alkyl, OH, -NHCOC1-3alkylNRc 2 and C(0)NHCH2C(0)NRc 2;
• a 3, 4, 5, 6 or 7-membered cycloalkyl group (which is optionally fused to phenyl), or
• Ra and Rb together form a 5, 6 or 7-membered heterocyclic group optionally containing one or more further heteroatoms independently selected from N, O, S or S(0)2 said heterocyclic group being optionally fused to a 5, 6 or 7-membered aryl or heteroaryl ring containing one or more heteroatoms independently selected from N, O and S; the heterocylic ring and/or the aryl or heteroaryl to which it is optionally fused being optionally substituted by one or more substituents independently selected from halo, OH, C1-3alkyl, 0-C1-3alkyl, C(0)C1-3alkyl, S(0)2C1.3alkyl, NHC(0)C1-3alkyl, NHS(0)2C1.3alkyl, C(0)NRc 2, C(0)NRd 2 (wherein Rd and Rd together form a 5 or 6-membered heterocylic ring), NR°2 C(0)phenyl, S(0)2NR°2, =0 (oxo) and 5, 6 or 7-membered aryl or heteroaryl (containing one or more heteroatoms independently selected from N, O and S);
R2 and R3 are each independently selected from:
H,
(CH^NR^CH^NR^,
(CH2)1-eNRc 2;
• C1-3 alkyl;
• 0-C1-3alkyl;
• C1-3haloalkyl;
• (CH2)0_3NRaRb (wherein Ra and Rb are as defined above);
(CH2)0-3NHPh;
(CH2)0_3OPh;
(CH2)0.3Ph;
or R2 and R3 together form a fused phenyl ring, and each R° is independently selected from hydrogen and C1-3alkyl
or a pharmaceutically acceptable salt thereof with the proviso R1 is not -C(CH3)3.
A compound of formula (I) as defined in claim 1 or a pharmaceutically acceptable salt thereof where R1 is:-
• C1-4 alkyl;
• C3_7 cycloalkyl;
• C1-4 fluoroalkyl;
• phenyl
• a 5-membered heteroaryl (which contains one or two heteroatoms selected from N, O and S and which is optionally fused to phenyl), said 5-membered heteroaryl being optionally substituted with one or more substituents independently selected from C1-3alkyl;
• 0-C1-4alkyl (which is optionally substituted by phenyl (optionally substituted by one of more chloro) or napthyl);
• O-cyclohexyl (which is optionally fused with phenyl);
C(0)NRc 2; or
NRaRb, either
• Ra is H or C1-4alkyl, especially H or methyl, and Rb is C^alkyl which is optionally substituted by one or more substituents independently selected from:
o phenyl (which is optionally substituted by one or more substiuents independently selected from methyl, O-methyl, C(0)NR°2, chloro, bromo and cyano),
o a 5 or 6-membered heteroaryl group (containing one or more heteroatoms independently selected from N, O and, S),
o a 3, 4 or 5-membered cycloalkyl group,
o OH
Figure imgf000134_0001
o NHCO(CH2)2NRc 2and
o C(0)NHCH2C(0)NRc 2;
• Ra is H and Rb is a 3, 4 or 5-membered cycloalkyl group (which is optionally fused to phenyl), or
• Ra and Rb together form a 5, 6 or 7-membered heterocyclic group optionally containing a further heteroatom selected from N, O, S or S(0)2 said heterocyclic group being optionally fused to a phenyl ring or to a 5 or 6-membered heteroaryl ring (containing one or more heteroatoms independently selected from N, O and S); the heterocylic ring and/or the phenyl or heteroaryl to which it is optionally fused being optionally substituted by one or more substituents independently selected from:
a. chloro or bromo,
b. OH,
c. C1-3alkyl,
d. 0-C1-3alkyl,
e. C(0)C1-3alkyl,
f. S(0)2C1.3alkyl,
g. NHC(0)C1-3alkyl,
h. NHS(0)2C1.3alkyl,
i. C(0)NRc 2,
j. C(0)NRd 2 (wherein Rd and Rd together form a 5 or 6-membered heterocylic ring), k. NRc 2i
I. C(0)phenyl,
m. S(0)2NRc 2,
n. =0 (oxo),
o. phenyl and p. 5 or 6-membered heteroaryl (containing one or more heteroatoms independently selected from N, O and S); and each R° is independently selected from hydrogen and methyl.
A compound of formula (I) as defined in claim 1 or a pharmaceutically acceptable salt thereof where R1 is:-
• C1-4 alkyl;
• C3.7 cycloalkyl, especially cyclopropyl;
• trifluoromethyl;
• phenyl
• a 5-membered heteroaryl (which contains one or two heteroatoms selected from N, O and S and which is optionally fused to phenyl), said 5-membered heteroaryl being optionally substituted with one or two methyl substituents;
• O-C^alkyl (which is optionally substituted by phenyl);
• O-cyclohexyl (which is fused with phenyl);
C(0)NRc 2;
or
NRaRb, either
• Ra is H or methyl, and Rb is C^alkyl which is optionally substituted by one or more substituents independently selected from:
o phenyl (which is optionally substituted by one or more substiuents independently selected from methyl, O-methyl, C(0)NR°2, chloro and cyano),
o a 5 or 6-membered heteroaryl group (containing one or two heteroatoms independently selected from N, O and, S),
o a 3 or 5-membered cycloalkyl group, especially cyclopropyl,
o OH,
o C(0)NRc 2,and
o C(0)NHCH2C(0)NRc 2;
• Ra is H and Rb is a 5-membered cycloalkyl group (which is optionally fused to phenyl), or
• Ra and Rb together form a 5, 6 or 7-membered heterocyclic group optionally containing a further heteroatom selected from N, O, S or S(0)2 said heterocyclic group being optionally fused to a phenyl ring or to a 5 or 6-membered heteroaryl ring (containing one or more heteroatoms independently selected from N, O and S); the heterocylic ring and/or the phenyl or heteroaryl to which it is optionally fused being optionally substituted by one or more substituents independently selected from:
a. chloro,
b. OH,
c. C1-3alkyl, especially methyl,
d. 0-C1-3alkyl, especially 0-CH3,
e. C(0)C1-3alkyl, especially C(0)CH3,
f. S(0)2C1.3alkyl, especially S(0)2CH3,
g. NHC(0)C1-3alkyl, especially NHC(0)CH3,
h. NHS(0)2C1.3alkyl, especially NHS(0)2CH3,
i. C(0)NRc 2,
j. C(0)NRd 2 (wherein Rd and Rd together form a 5 or 6-membered heterocylic ring), k. NRc 2i
I. C(0)phenyl,
m. S(0)2NRc 2,
n. =0 (oxo),
o. phenyl and
p. 5 or 6-membered heteroaryl (containing one or more heteroatoms independently selected from N, O and S);
and
NR°2 is selected from NHCH3 and NH2.
A compound of formula (I) as defined in claim 1 or a pharmaceutically acceptable salt thereof where R1 is:-
• C1-4 alkyl;
• C3_7 cycloalkyl;
• C1-4 fluoroalkyl; or
NRaRb,
wherein
• Ra and Rb together form a 5, 6 or 7-membered heterocyclic group optionally containing a further heteroatom selected from N, O, S or S(0)2 said heterocyclic group being optionally fused to a phenyl ring or to a 5 or 6-membered heteroaryl ring (containing one or more heteroatoms independently selected from N, O and S); the heterocylic ring and/or the phenyl or heteroaryl to which it is optionally fused being optionally substituted by one or more substituents independently selected from:
a. chloro or bromo, b. OH,
c. C1-3alkyl,
d. 0-C1-3alkyl,
e. C(0)C1-3alkyl,
f. S(0)2C1.3alkyl,
g. NHC(0)C1-3alkyl,
h. NHS(0)2C1.3alkyl, (wherein Rd and Rd together form a 5 or 6-membered heterocylic ring),
Figure imgf000137_0001
I. C(0)phenyl,
Figure imgf000137_0002
n. =0 (oxo),
o. phenyl and
p. 5 or 6-membered heteroaryl (containing one or more heteroatoms independently selected from N, O and S),
and each R°2 is independently selected from H and CH3.
A compound of formula (I) as defined in claim 1 or a pharmaceutically acceptable salt thereof where R1 is:-
• C1-4 alkyl;
• C3_7 cycloalkyl;
• C1-4 fluoroalkyl; or
NRaRb,
wherein
Ra and Rb together form a 5, 6 or 7-membered heterocyclic group said heterocyclic group being optionally fused to a phenyl ring; the heterocylic ring and/or the phenyl to which it is optionally fused being optionally substituted by one or more substituents independently selected from: chloro, bromo, OH, C1-3alkyl, 0-C1-3alkyl, C(0)C1-3alkyl, and S(0)2C1-3alkyl.
In a yet further embodiment, R1 is:
• C1-4 alkyl;
NRaRb,
wherein
Ra and Rb together form a 5, 6 or 7-membered heterocyclic group said heterocyclic group being fused to a phenyl ring; the heterocylic ring and/or the phenyl to which it is fused being unsubstiuted or substituted by one or more substituents independently selected from: chloro, OH, methyl, O-methyl and S(0)2methyl, for example, unsubstituted. R2 and R3 are selected from H, (CH2)1.3NRC(CH2)1.3NRC 2 and (CH2)0-6NRC 2.
7. A compound of formula (I) as defined in claim 1 or a pharmaceutically acceptable salt thereof wherein one of R2 and R3 is H and the other is selected from (CI-y^NR^CI-y^NR^ and (CH2)o_6NRc 2.
8. A compound of formula (I) as defined in claim 1 or a pharmaceutically acceptable salt thereof R2 and R3 are both H.
9. A compound of formula (I) as defined in claim 1 which is;
Λ/-[6-(1 ,1 -dimethylethyl)-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine trifluoroacetate
N-[2-(2-pyridinyl)-6-(trifluoromethyl)-4-pyrimidinyl]-p-alanine
A/-[6-(4-morpholinyl)-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine
A/-[6-(methylamino)-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine
A/-[2-(2-pyridinyl)-6-(1 -pyrrolidinyl)-4-pyrimidinyl]-p-alanine
A/-[6-[(2-hydroxyethyl)amino]-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine. formate
A/-[6-[(phenylmethyl)amino]-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine
A/-[6-phenyl-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine
A/-[6-[3-(aminocarbonyl)-1 -piperidinyl]-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine
A/-[6-[4-(aminocarbonyl)-1 -piperidinyl]-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine
A/-[6-({[3,4-Jb/s(methyloxy)phenyl]methyl}amino)-2-(2-pyridinyl)-4-pyrimidinyl]-p-anine
A/-[6-[(3-amino-3-oxopropyl)(methyl)amino]-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine
A/-[6-[{[3-(aminocarbonyl)phenyl]methyl}(methyl)amino]-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine
A/-/6-{[(3,4-dichlorophenyl)methyl]amino}-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine
A/-[6-({[3-(aminocarbonyl)phenyl]methyl}amino)-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine
A/-[6-{[(4-chlorophenyl)methyl]amino}-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine
A/-[6-(7/-/-pyrazol-4-yl)-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine
A/-[6-(3-methyl-1 /-/-pyrazol-4-yl)-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine
A/-[6-{[(3-chlorophenyl)methyl]amino}-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine
A/-[6-{[(2-methylphenyl)methyl]amino}-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine
A/-{2-(2-pyridinyl)-6-[(2-thienylmethyl)amino]-4-pyrimidinyl}-p-alanine
A/-[6-({[3-(methyloxy)phenyl]methyl}amino)-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine
A/-{2-(2-pyridinyl)-6-[(2-pyridinylmethyl)amino]-4-pyrimidinyl}-p-alanine
A/-[6-({[4-(methyloxy)phenyl]methyl}amino)-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine
A/-[6-[(cyclopropylmethyl)amino]-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine
Λ/-[6-(1 ,3-dihydro-2/-/-isoindol-2-yl)-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine
A/-[6-(1 -methylethyl)-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine /V-[6-cyclopropyl-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine
A/-[6-[3-(diethylamino)-1 ^iperidinyl]-2^
Λ/-[6-[4-(1 ,3,4-oxadiazol-2-yl)-1 ^iperidinyl]-2-(2^yridinyl)-4^yrimidinyl]-p-alanine
A/-{2-(2-pyridinyl)-6-[4-(1 ,3-thiazol-2-yl)-1 -piperazinyl]-4-pyrimidinyl}-p-alanine
A/-[6-(4^henyl-1 ^iperazinyl)-2-(2^yridinyl)-4-pyrimidinyl]-p-alanine
A -[6-{[(4-chlorophenyl)methyl]amino}-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine
A/-[6-(hexahydro-1 H-azepin-1 -yl)-2-(2^yridinyl)-4^yrimidinyl]-p-alanine
/V-[6-(1 -benzothien-2-yl)-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine
A/-[6-{4-[(methylamino)carbonyl]-1 ^iperid^
A/-[6-(7-hydroxy-1 ,2,4,5-tetrahydro-3H-3-benzazepin-3-yl)-2-(2^yridinyl)-4^yrimidinyl]-p-alanine
A/-[6-(7-bromo-1 ,2,4,5-tetrahydro-3H-3-benzazepin-3-yl)-2-(2^yridinyl)-4^yrimidinyl]-p-alanine
A/-[6-(5-hydroxy-1 ,3-dihydro-2H-isoindol-2-yl)-2-(2^yridinyl)-4-pyrimidinyl]-p-alanine
Λ/-{6-(1 ,2,4,5-tetrahydro-3H-3-benzazepin-3-yl)-2-[4-(1 ,2,4,5-tetrahydro-3H-3-benzazepin-3-yl)-2- pyridinyl]-4-pyrimidinyl}-p-alanine
Λ/-(6-(1 ,3-dihydro-2H-isoindol-2-yl)-2-{4-[(phenyloxy)
Λ/-[6-(1 ,3-dihydro-2H-isoindol-2-yl)-2-(4^henyl-2^yridinyl)-4^yrimidinyl]-p-alanin
A/-[6-[(2-hydroxyethyl)(methyl)amino]-2-(2^yridinyl)-4^yrimidinyl]-p-alanin
A/-[6-(dimethylamino)-2-(2-pyridinyl)-4-pyrimidinyl]- β-alanine
Lithium A/-{2-{4-[3-(methylamino)propyl]-2^yridinyl}-6-[(phenylmethyl)amino]-4^yrimidinyl}-p- alanine
A/-{2-(4-{3-[(2-aminoethyl)amino]propyl}-2^yridinyl)-6-[(phenylmethyl)amino]-4^yrimidi lithium A/-[2-{4-[3-(methylamino)propyl]-2-pyridinyl}-6-(1 ,2,4,5-tetrahydro-3/-/-3-benzazepin-3-yl)-4- pyrimidinyl]-p-alanine
3-{[6-{[3-(methylamino)-3-oxopropyl]amino}-2-(2^yridinyl)-4-pyrimidinyl]amino}propanoic acid
A/-[6-[(2-carboxyethyl)amino]-2-(2^yrid^
A/-[6-({2-[3-(p-alanylamino)phenyl]ethyl}amino)-2-^^^
A/-{2-(2^yridinyl)-6-[4-(1 ^yrrolidinylcarbony^^
A/-[6-(cyclohexylamino)-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine
3-{[6-{methyl[3-(methylamino)-3-oxopropyl]amino}-2-(2^yridinyl)-4^yrimidinyl]amino}propanoic acid
A/-[6-{[(3,4-dichlorophenyl)methyl]oxy}-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine
A/-[6-[(1 -phenylethyl)oxy]-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine
A/-[6-{[(3-chlorophenyl)methyl]oxy}-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine
A/-[6-[(2-naphthalenylmethyl)oxy]-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine
A/-[2-(2-pyridinyl)-6-(1 ,2,4,5-tetrahydro-3/-/-3-benzazepin-3-yl)-4-pyrimidinyl]-p-alanine
A/-[6-(3,4-dihydro-2(1 /^-isoquinolinyl)-2-(2^yridinyl)-4^yrimidinyl]-p-alanine
A/-[6-[7-(methyloxy)-1 ,2,4,5-tetrahydro-3H-3-benzazepin-3-yl]-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine A/-[6-(4^henyl-1 ^iperidinyl)-2-(2^yridinyl)-4^yrimidinyl]-p-alanine
A/-[6-(4-hydroxy-1 ^iperidinyl)-2-(2^yridinyl)-4^yrimidinyl]-p-alanine
A/-[2-(2^yridinyl)-6-(4-thiomorpholinyl)-4-pyrimidinyl]-p-alanine
A/-[6-(3-hydroxy-1 ^yrrolidinyl)-2-(2^yridinyl)-4-pyrimidinyl]-p-alanine
A/-[2-(2-pyridinyl)-6-(1 ,3,4,5-tetrahydro-2/-/-2-benzazepin-2-yl)-4-pyrimidinyl]-p-alanine
A/-[6-(1 -piperidinyl)-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine
A/-[6-(4-methyl-1 ^iperazinyl)-2-(2^yridinyl)-4-pyrimidinyl]-p-alanine
A/-[6-[4-(phenylcarbonyl)-1 ^iperazinyl]-2-(2^yridinyl)-4^yrimidinyl]-p-alanine
A/-[6-{3-[(methylamino)carbonyl]-1 ^iper^
A/-{2-(2^yridinyl)-6-[3-(1 ^yrrolidinylcarbo^^
A/-[6-(4-methyl-1 ^iperidinyl)-2-(2^yridinyl)-4^yrimidinyl]-p-alanine
A/-[6-(4,4-dimethyl-1 ^iperidinyl)-2-(2^yridinyl)-4^yrimidinyl]-p-alanine
A/-[6-(4^ropanoyl-1 ^iperazinyl)-2-(2^yridinyl)-4-pyrimidinyl]-p-alanine
A/-[6-(5-chloro-1 ,3-dihydro-2H-isoindol-2-yl)-2-(2^yridinyl)-4-pyrimidinyl]-p-alanine
A/-[6-[5-(methyloxy)-1 ,3-dihydro-2H-isoindol-2-yl]-2-(2^yridinyl)-4-pyrimidinyl]-p-alanine
A/-[6-(2-methyl-4,5J,8-tetrahydro-6H-[1 ,3]thiazolo[4,5-olazepin-6-yl)-2-(2-pyridinyl)-4- pyrimidinyl]-p-alanine
A/-[6-[7-(methylsulfonyl)-1 ,2,4,5-tetrahydro-3H-3-benzazepin-3-yl]-2-(2-pyridinyl)-4- pyrimidinyl]-p-alanine
A/-[6-[7-(aminosulfonyl)-1 ,2,4,5-tetrahydro-3H-3-benzazepin-3-yl]-2-(2-pyridinyl)-4- pyrimidinyl]-p-alanine
A/-[6-{4-[(dimethylamino)carbonyl]-1 ^iper^
A/-[6-[methyl(phenylmethyl)amino]-2-(2^yridinyl)-4^yrimidinyl]-p-alanine
A/ 2-(2^yridinyl)-6-[(4^yridinylmethyl)amino]-4^yrimidinyl}-p-alanine
A/ 2-(2^yridinyl)-6-[(3^yridinylmethyl)amino]-4^yrimidinyl}-p-alanine
A/-[6-(2,3-dihydro-1 H-inden-2-ylamino)-2-(2^yridinyl)-4^yrimidinyl]-p-alani
A/-[6-[(2^henylethyl)amino]-2-(2^yridinyl)-4-pyrimidinyl]-p-alanine
A/-[6-({[3-(methyloxy)phenyl]methyl}amino)^
A/-[6-{[(3-cyanophenyl)methyl]amino}-2-(2^y^
A/-[6-{[2-(methyloxy)ethyl]amino}-2-(2^yridinyl)-4^yrimidinyl]-p-alanine
Λ/-[6-(1 -dioxido-4-thiomorpholinyl)-2-(2^yridinyl)-4^yrimidinyl]-p-alanine
A/-[6-{[(2,6-dimethylphenyl)methyl]amino}-2-(2^yridinyl)-4^yrimidinyl]-p-a
A/-[6-(methyloxy)-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine
A/-[6-(4-acetyl-1 ^iperazinyl)-2-(2^yridinyl)-4-pyrimidinyl]-p-alanine
A/-[6-[3-(acetylamino)-1 ^yrrolidinyl]-2-(2^yridinyl)-4^yrimidinyl]-p-alanin
A/-[6-[3-(acetylamino)-1 ^iperidinyl]-2-(2^yridinyl)-4^yrimidinyl]-p-alanine A/-[6-[4-(methylsulfonyl)-1 -piperazinyl]-2-(2-pyridinyl)-4-pyrimidinyl]-p-a
N- [6-{3- [(m eth y Is u If o n y I) a m i n o]- 1 - py rro lidinyl}-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine
A/-[6-{3-[(methylsulfonyl)amino]-1 -piperidinyl}-2-(2-pyridinyl)-4-pyrim
A/-[6-[(phenylmethyl)oxy]-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine
A/-[2-(2-pyridinyl)-6-(1 ,2,3,4-tetrahydro-2-naphthalenyloxy)-4-pyrimidinyl]-p-alanine
A/-[6-[(2-phenylethyl)oxy]-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine
A/-[6-(1 -benzothien-3-yl)-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine
Λ/-[6-(1 ,1 -dimethylethyl)-2-(3-isoquinolinyl)-4-pyrimidinyl]-p-alanine
Λ/-{6-(1 -dimethylethyl)-2-[5-(trifluoromethyl)-2-pyridinyl]-4-pyrimidinyl}- -alanine
Λ/-[6-(1 ,1 -dimethylethyl)-2-(4-methyl-2-pyridinyl)-4-pyrimidinyl]-p-alanine
Λ/-{6-(1 ,1 -dimethylethyl)-2-[4-(methyloxy)-2-pyridinyl]-4-pyrimidinyl}-p-alanine
Λ/-{6-(1 ,1 -dimethylethyl)-2-[5-(methyloxy)-2-pyridinyl]-4-pyrimidinyl}-p-alanine
Λ/-[6-(1 ,1 -dimethylethyl)-2-(5-methyl-2-pyridinyl)-4-pyrimidinyl]-p-alanine
A/-[2-[4-(dimethylamino)-2-pyridinyl]-6-(1 ,1 -dimethylethyl)-4-pyrimidinyl]-p-alanine
Λ/-[6-(1 -oxo-3, 4-dihydro-2(1 /^-isoquinolinyl)-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine
Λ/-(6-(1 ,3-dihydro-2H-isoindol-2-yl)-2-{4-[(phenylamino)methyl]-2-pyridinyl}-4-pyrim
A/-[6-[(methylamino)carbonyl]-2-(2-pyridinyl)-4-pyrimidinyl]-p-alanine
A/-[2-{4-[(2-hydroxyethyl)amino]-2-pyridinyl}-6-(1 ,2,4,5-tetrahydro-3H-3-benzazepin-3-yl)-4- pyrimidinyl]-p
10. A compound of formula (I) as defined in claim 1 or a pharmaceutically acceptable salt thereof, or a compound of formula (la) as defined in claim 2 or a pharmaceutically acceptable salt thereof, for use in therapy, in particular in changing the epigenetic status of cells, treating cancer, inflammation or autoimmune diseases.
5
1 1 . A method of changing the epigenetic status of cells, treating cancer, inflammation or autoimmune diseases in a subject comprising administering a therapeutically effective amount of a compound of formula (I) as defined in claim 1 or a pharmaceutically acceptable salt thereof, or a compound of formula (la) as defined in claim 2 or a pharmaceutically
10 acceptable salt thereof.
12. A compound of formula (I) as defined in claim 1 or a pharmaceutically acceptable salt thereof, or a compound of formula (la) as defined in claim 2 or a pharmaceutically acceptable salt
15 thereof, for use in the treatment of cancer.
13. A method of treating cancer comprising administering to a human in need thereof a therapeutically effective amount of a compound of formula (I) as defined in claim 1 or a pharmaceutically acceptable salt thereof, or a compound of formula (la) as defined in claim 2
20 or a pharmaceutically acceptable salt thereof.
14. A compound of formula (I) as defined in claim 1 or a pharmaceutically acceptable salt thereof, for use in the treatment of inflammation or autoimmune diseases.
25 15. A method of treating inflammation or autoimmune diseases comprising administering to human in need thereof, a therapeutically effect amount of a compound of formula (I) as defined in claim 1 or a pharmaceutically acceptable salt thereof.
40
PCT/EP2011/068085 2010-10-19 2011-10-17 N-2-(2-pyridinyl)-4-pyrimidinyl-beta-alanine derivatives as inhibitors of histone demethylase jmjd3 WO2012052390A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US39456310P 2010-10-19 2010-10-19
US61/394,563 2010-10-19
GB1112607.5 2011-07-22
GBGB1112607.5A GB201112607D0 (en) 2011-07-22 2011-07-22 Novel compounds

Publications (1)

Publication Number Publication Date
WO2012052390A1 true WO2012052390A1 (en) 2012-04-26

Family

ID=44652147

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2011/068085 WO2012052390A1 (en) 2010-10-19 2011-10-17 N-2-(2-pyridinyl)-4-pyrimidinyl-beta-alanine derivatives as inhibitors of histone demethylase jmjd3

Country Status (2)

Country Link
GB (1) GB201112607D0 (en)
WO (1) WO2012052390A1 (en)

Cited By (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013030665A1 (en) * 2011-09-02 2013-03-07 Purdue Pharma L.P. Pyrimidines as sodium channel blockers
CN103524535A (en) * 2013-10-16 2014-01-22 苏州云轩医药科技有限公司 Amido thiazole-pyridine heterocycle compound with activity of hedgehog path antagonist
CN103694236A (en) * 2013-01-15 2014-04-02 苏州云轩医药科技有限公司 Novel antineoplastic compound containing pyrimidine skeleton and hedgehog pathway antagonist activity
WO2014100818A1 (en) 2012-12-21 2014-06-26 Quanticel Pharmaceuticals, Inc. Histone demethylase inhibitors
WO2014190035A2 (en) * 2013-05-21 2014-11-27 Dana-Farber Cancer Institute, Inc. Compositions and methods for identification, assessment, prevention, and treatment of cancer using histone h3k27me2 biomarkers and modulators
KR20150123894A (en) * 2013-02-27 2015-11-04 에피테라퓨틱스 에이피에스 Inhibitors of histone demethylases
WO2015200709A1 (en) * 2014-06-25 2015-12-30 Quanticel Pharmaceuticals, Inc. Histone demethylase inhibitors
JP2016512834A (en) * 2013-03-15 2016-05-09 クオンティセル ファーマシューティカルズ,インク. Histone demethylase inhibitor
JP2016514159A (en) * 2013-03-14 2016-05-19 クオンティセル ファーマシューティカルズ,インク. Histone demethylase inhibitor
CN106608869A (en) * 2015-10-27 2017-05-03 中国科学院上海药物研究所 Histone demethylase JMJD3 inhibitor, preparation method and application thereof
CN106810538A (en) * 2017-01-09 2017-06-09 湖南华腾制药有限公司 A kind of preparation method of 2 pyrrole radicals pyrimidine derivatives
CN106854181A (en) * 2015-12-08 2017-06-16 湖南华腾制药有限公司 A kind of preparation method of pyrimidine compound
WO2017157825A1 (en) 2016-03-15 2017-09-21 F. Hoffmann-La Roche Ag Combinations of lsd1 inhibitors for use in the treatment of solid tumors
WO2018106984A1 (en) 2016-12-09 2018-06-14 Constellation Pharmaceuticals, Inc. Markers for personalized cancer treatment with lsd1 inhibitors
US10206930B2 (en) 2015-08-14 2019-02-19 New York University Methods for treating T-cell acute lymphoblastic leukemia
WO2019046378A1 (en) * 2017-09-01 2019-03-07 The Johns Hopkins University H3k27 demethylase inhibitors in pediatric and juvenile osteoporosis
EP3579834A4 (en) * 2017-02-10 2021-04-28 Temple Otorongo LLC Treatment of diabetes and associated metabolic conditions with epigenetic modulators
CN113387938A (en) * 2020-03-11 2021-09-14 上海科技大学 Substituted pyrimidine compound, preparation method, intermediate and application thereof
EP3793550A4 (en) * 2018-05-15 2022-04-13 Arbutus Biopharma Corporation Substituted 2,2'-bipyrimidinyl compounds, analogues thereof, and methods using same
US11400099B2 (en) 2016-09-23 2022-08-02 Thomas Jefferson University Drug-induced epigenetic remodeling to prevent fibrosis
CN115466245A (en) * 2022-06-29 2022-12-13 深圳大学 Hydroxamic acid derivative of pyrimidopyridine and preparation method and application thereof
US12083125B2 (en) 2022-07-07 2024-09-10 Thomas Jefferson University Drug-induced epigenetic remodeling to prevent fibrosis

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0123456A2 (en) 1983-03-28 1984-10-31 Compression Labs, Inc. A combined intraframe and interframe transform coding method
US5681835A (en) 1994-04-25 1997-10-28 Glaxo Wellcome Inc. Non-steroidal ligands for the estrogen receptor
US6268391B1 (en) 1997-08-06 2001-07-31 Glaxo Wellcome Inc. Benzylidene-1,3-dihydro-indol-2-one derivatives a receptor tyrosine kinase inhibitors, particularly of Raf kinases
WO2009087224A1 (en) 2008-01-11 2009-07-16 Novartis Ag Pyrimidines as kinase inhibitors
WO2009114011A1 (en) 2008-03-11 2009-09-17 President And Fellows Of Harvard College Histone demethylation proteins and methods of use thereof
WO2010043866A2 (en) * 2008-10-15 2010-04-22 Isis Innovation Limited Histone lysine demethylase inhibitors
WO2010043566A2 (en) 2008-10-17 2010-04-22 Sanofi-Aventis Deutschland Gmbh Combination of an insulin and a glp-1 agonist

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0123456A2 (en) 1983-03-28 1984-10-31 Compression Labs, Inc. A combined intraframe and interframe transform coding method
US5681835A (en) 1994-04-25 1997-10-28 Glaxo Wellcome Inc. Non-steroidal ligands for the estrogen receptor
US5877219A (en) 1994-04-25 1999-03-02 Glaxo Wellcomeinc. Non-steroidal ligands for the estrogen receptor
US6207716B1 (en) 1994-04-25 2001-03-27 Glaxo Wellcome Inc. Non-steroidal ligands for the estrogen receptor
US6268391B1 (en) 1997-08-06 2001-07-31 Glaxo Wellcome Inc. Benzylidene-1,3-dihydro-indol-2-one derivatives a receptor tyrosine kinase inhibitors, particularly of Raf kinases
WO2009087224A1 (en) 2008-01-11 2009-07-16 Novartis Ag Pyrimidines as kinase inhibitors
WO2009114011A1 (en) 2008-03-11 2009-09-17 President And Fellows Of Harvard College Histone demethylation proteins and methods of use thereof
WO2010043866A2 (en) * 2008-10-15 2010-04-22 Isis Innovation Limited Histone lysine demethylase inhibitors
WO2010043566A2 (en) 2008-10-17 2010-04-22 Sanofi-Aventis Deutschland Gmbh Combination of an insulin and a glp-1 agonist

Non-Patent Citations (50)

* Cited by examiner, † Cited by third party
Title
"Tyrosine Kinase Signalling in Breast cancer:erbB Family Receptor Tyrosine Kinases", BREAST CANCER RES., vol. 2, no. 3, 2000, pages 176 - 183
ABRAHAM, R.T., CURRENT OPINION IN IMMUNOLOGY., vol. 8, no. 3, 1996, pages 412 - 8
AGGER K ET AL., GENES DEV., vol. 23, April 2009 (2009-04-01), pages 1171 - 1176
AGGER K ET AL., NATURE, vol. 449, no. 7163, October 2007 (2007-10-01), pages 731 - 734
ASHBY, M.N., CURRENT OPINION IN LIPIDOLOGY., vol. 9, no. 2, 1998, pages 99 - 102
BANNISTER ET AL., CELL, vol. 109, 2002, pages 801 - 806
BERGE ET AL., J. PHARM. SCI., vol. 66, 1977, pages 1 - 19
BIOCHIM. BIOPHYS. ACTA, vol. 1423, no. 3, 1989, pages 19 - 30
BOLEN, J.B., BRUGGE, J.S., ANNUAL REVIEW OF IMMUNOLOGY., vol. 15, 1997, pages 371 - 404
BREKKEN, R.A. ET AL.: "elective Inhibition of VEGFR2 Activity by a monoclonal Anti-VEGF antibody blocks tumor growth in mice", CANCER RES., vol. 60, 2000, pages 5117 - 5124, XP002340113
BRODT, P, SAMANI, A., NAVAB, R., BIOCHEMICAL PHARMACOLOGY, vol. 60., 2000, pages 1101 - 1107
CANMAN, C.E., LIM, D.S., ONCOGENE, vol. 17, no. 25, 1998, pages 3301 - 3308
DE SANTA F ET AL., CELL, vol. 130, September 2007 (2007-09-01), pages 1083 - 1094
DE SANTA F ET AL., EMBO J., vol. 28, September 2009 (2009-09-01), pages 3341 - 3352
EINZIG, PROC. AM. SOC. CLIN. ONCOL., vol. 20, pages 46
FORASTIRE, SEM. ONCOL., vol. 20, 1990, pages 56
GREEN, M.C. ET AL.: "Monoclonal Antibody Therapy for Solid Tumors", CANCER TREAT. REV., vol. 26, no. 4, 2000, pages 269 - 286, XP009019784, DOI: doi:10.1053/ctrv.2000.0176
HOLMES ET AL., J. NAT. CANCER INST., vol. 83, 1991, pages 1797
IGNOFF, R.J., CANCER CHEMOTHERAPY POCKET GUIDE, 1998
ISHII M ET AL., BLOOD, vol. 114, no. 15, October 2009 (2009-10-01), pages 3244 - 3254
JACKSON, S.P., INTERNATIONAL JOURNAL OF BIOCHEMISTRY AND CELL BIOLOGY., vol. 29, no. 7, 1997, pages 935 - 8
KATH, JOHN C., EXP. OPIN. THER. PATENTS, vol. 10, no. 6, 2000, pages 803 - 818
KEARNS, C.M., SEMINARS IN ONCOLOGY, vol. 3, no. 6, 1995, pages 16 - 23
KLOSE RJ ET AL., NAT REV GENET., vol. 7, no. 9, September 2006 (2006-09-01), pages 715 - 27
LACKEY, K. ET AL., BIOORGANIC AND MEDICINAL CHEMISTRY LETTERS, no. 10, 2000, pages 223 - 226
LOFTS, F. J. ET AL.: "New Molecular Targets for Cancer Chemotherapy", 1994, CRC PRESS, article "Growth factor receptors as targets"
MARKMAN ET AL., YALE JOURNAL OF BIOLOGY AND MEDICINE, vol. 64, 1991, pages 583
MARTIN C., ZHANG Y., NATURE REV. MOL. CELL BIOL., vol. 6, 2005, pages 838 - 849
MARTINEZ-LACACI, L. ET AL., INT. J. CANCER, vol. 88, no. 1, 2000, pages 44 - 52
MASSAGUE, J., WEIS-GARCIA, F., CANCER SURVEYS., vol. 27, 1996, pages 41 - 64
MCGUIRE ET AL., ANN. INTEM, MED., vol. 111, 1989, pages 273
MILLER SA ET AL., GENES DEV., vol. 22, October 2008 (2008-10-01), pages 2280 - 2993
NAKAYAMA ET AL., SCIENCE, vol. 292, 2001, pages 110 - 113
NIELSEN ET AL., NATURE, vol. 412, 2001, pages 561 - 565
PHILIP, P.A., HARRIS, A.L., CANCER TREATMENT AND RESEARCH., vol. 78, 1995, pages 3 - 27
POWIS, G., KOZIKOWSKI A.: "New Molecular Targets for Cancer Chemotherapy", 1994, CRC PRESS
ROSANIA ET AL., EXP. OPIN. THER. PATENTS, vol. 10, no. 2, 2000, pages 215 - 230
SCHAROVSKY, O.G., ROZADOS, V.R., GERVASONI, S.I., MATAR, P., JOURNAL OF BIOMEDICAL SCIENCE, vol. 7, no. 4, 2000, pages 292 - 8
SEN GL ET AL., GENES DEV., vol. 22, July 2008 (2008-07-01), pages 1865 - 1870
SHAWVER ET AL., DDT, vol. 2, no. 2, February 1997 (1997-02-01)
SHI ET AL., CELL, vol. 119, 2004, pages 941 - 953
SHI ET AL., NATURE, vol. 422, 2003, pages 735 - 738
SINH, S., COREY, S.J., JOURNAL OF HEMATOTHERAPY AND STEM CELL RESEARCH, vol. 8, no. 5, 1999, pages 465 - 80
SMITHGALL, T.E., JOURNAL OF PHARMACOLOGICAL AND TOXICOLOGICAL METHODS., vol. 34, no. 3, 1995, pages 125 - 32
T. W. GREEN, P. G. M. WUTS: "Protective Groups in Organic Synthesis", 1999, JOHN WILEY & SONS
TSUKADA ET AL., NATURE, vol. 439, 2006, pages 811 - 816
VAKOC C. ET AL., MOL. CELL, vol. 19, 2005, pages 381 - 391
WOO, NATURE, vol. 368, 1994, pages 750
YAMAMOTO, T., TAYA, S., KAIBUCHI, K., JOURNAL OF BIOCHEMISTRY, vol. 126, no. 5, 1999, pages 799 - 803
ZHONG, H. ET AL., CANCER RES, vol. 60, no. 6, 2000, pages 1541 - 1545

Cited By (69)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9163008B2 (en) 2011-09-02 2015-10-20 Purdue Pharma, L.P. Pyrimidines as sodium channel blockers
EP3255041A1 (en) * 2011-09-02 2017-12-13 Purdue Pharma L.P. Pyrimidines as sodium channel blockers
US10059675B2 (en) 2011-09-02 2018-08-28 Purdue Pharma L.P. Pyrimidines as sodium channel blockers
WO2013030665A1 (en) * 2011-09-02 2013-03-07 Purdue Pharma L.P. Pyrimidines as sodium channel blockers
AU2012300567B2 (en) * 2011-09-02 2016-03-10 Purdue Pharma L.P. Pyrimidines as sodium channel blockers
US10774050B2 (en) 2011-09-02 2020-09-15 Purdue Pharma, L.P. Pyrimidines as sodium channel blockers
US9656968B2 (en) 2011-09-02 2017-05-23 Purdue Pharma L.P Pyrimidines as sodium channel blockers
US8952151B2 (en) 2012-12-21 2015-02-10 Quanticel Pharmaceuticals, Inc. Histone demethylase inhibitors
WO2014100818A1 (en) 2012-12-21 2014-06-26 Quanticel Pharmaceuticals, Inc. Histone demethylase inhibitors
US9725441B2 (en) 2012-12-21 2017-08-08 Celgene Quanticel Research, Inc. Histone demethylase inhibitors
US10807956B2 (en) 2012-12-21 2020-10-20 Celgene Quanticel Research, Inc. Histone demethylase inhibitors
US9611221B2 (en) 2012-12-21 2017-04-04 Celgene Quanticel Research, Inc. Histone demethylase inhibitors
US11214542B2 (en) 2012-12-21 2022-01-04 Celgene Quanticel Research, Inc. Histone demethylase inhibitors
US9828343B2 (en) 2012-12-21 2017-11-28 Celgene Quanticel Research, Inc. Histone demethylase inhibitors
US10597365B2 (en) 2012-12-21 2020-03-24 Celgene Quanticel Research, Inc. Histone demethylase inhibitors
US10179769B2 (en) 2012-12-21 2019-01-15 Celgene Quanticel Research, Inc. Histone demethylase inhibitors
US9085534B2 (en) 2012-12-21 2015-07-21 Quanticel Pharmaceuticals, Inc. Histone demethylase inhibitors
US11639333B2 (en) 2012-12-21 2023-05-02 Celgene Quanticel Research, Inc Histone demethylase inhibitors
US9447045B2 (en) 2012-12-21 2016-09-20 Celgene Quanticel Research, Inc. Histone demethylase inhibitors
EA028774B1 (en) * 2012-12-21 2017-12-29 Квонтисел Фармасьютикалс, Инк. Histone demethylase inhibitors
CN103694236A (en) * 2013-01-15 2014-04-02 苏州云轩医药科技有限公司 Novel antineoplastic compound containing pyrimidine skeleton and hedgehog pathway antagonist activity
JP2016509047A (en) * 2013-02-27 2016-03-24 エピセラピューティクス アーペーエス Inhibitors of histone demethylase
KR102229992B1 (en) 2013-02-27 2021-03-19 에피테라퓨틱스 에이피에스 Inhibitors of histone demethylases
KR20150123894A (en) * 2013-02-27 2015-11-04 에피테라퓨틱스 에이피에스 Inhibitors of histone demethylases
EP2969007A4 (en) * 2013-03-14 2016-07-27 Quanticel Pharmaceuticals Inc Histone demethylase inhibitors
US10273222B2 (en) 2013-03-14 2019-04-30 Celgene Quantscel Research, Inc. Histone demethylase inhibitors
JP2016514159A (en) * 2013-03-14 2016-05-19 クオンティセル ファーマシューティカルズ,インク. Histone demethylase inhibitor
US9963441B2 (en) 2013-03-14 2018-05-08 Celgene Quanticel Research, Inc. Histone demethylase inhibitors
US9701657B2 (en) 2013-03-14 2017-07-11 Celgene Quanticel Research, Inc. Histone demethylase inhibitors
US9527829B2 (en) 2013-03-14 2016-12-27 Celgene Quanticel Research, Inc. Histone demethylase inhibitors
US9994562B2 (en) 2013-03-15 2018-06-12 Celgene Quanticel Research, Inc. Histone demethylase inhibitors
JP2016512834A (en) * 2013-03-15 2016-05-09 クオンティセル ファーマシューティカルズ,インク. Histone demethylase inhibitor
US10526327B2 (en) 2013-03-15 2020-01-07 Celgene Quanticel Research, Inc. Histone demethylase inhibitors
EP2970211A4 (en) * 2013-03-15 2016-08-03 Quanticel Pharmaceuticals Inc Histone demethylase inhibitors
AU2014235280B2 (en) * 2013-03-15 2017-08-31 Celgene Quanticel Research, Inc. Histone demethylase inhibitors
WO2014190035A3 (en) * 2013-05-21 2015-01-22 Dana-Farber Cancer Institute, Inc. Compositions and methods for identification, assessment, prevention, and treatment of cancer using histone h3k27me2 biomarkers and modulators
WO2014190035A2 (en) * 2013-05-21 2014-11-27 Dana-Farber Cancer Institute, Inc. Compositions and methods for identification, assessment, prevention, and treatment of cancer using histone h3k27me2 biomarkers and modulators
CN103524535A (en) * 2013-10-16 2014-01-22 苏州云轩医药科技有限公司 Amido thiazole-pyridine heterocycle compound with activity of hedgehog path antagonist
CN111909083A (en) * 2014-06-25 2020-11-10 赛尔基因昆蒂赛尔研究公司 Histone demethylase inhibitors
WO2015200709A1 (en) * 2014-06-25 2015-12-30 Quanticel Pharmaceuticals, Inc. Histone demethylase inhibitors
US9447046B2 (en) 2014-06-25 2016-09-20 Celegene Quanticel Research, Inc. Histone demethylase inhibitors
US10106534B2 (en) 2014-06-25 2018-10-23 Celgene Quanticel Research, Inc. Histone demethylase inhibitors
EA031200B1 (en) * 2014-06-25 2018-11-30 Селджен Квонтисел Рисёрч, Инк. Histone methylase inhibitors
CN111909083B (en) * 2014-06-25 2023-08-15 赛尔基因昆蒂赛尔研究公司 Inhibitors of histone demethylase
US9994561B2 (en) 2014-06-25 2018-06-12 Celgene Quanticel Research, Inc. Histone demethylase inhibitors
CN106660982B (en) * 2014-06-25 2020-09-08 赛尔基因昆蒂赛尔研究公司 Histone demethylase inhibitors
US9242968B2 (en) 2014-06-25 2016-01-26 Quanticel Pharmaceuticals, Inc. Histone demethylase inhibitors
CN106660982A (en) * 2014-06-25 2017-05-10 赛尔基因昆蒂赛尔研究公司 Histone demethylase inhibitors
US10385047B2 (en) 2014-06-25 2019-08-20 Celgene Quanticel Research, Inc. Histone demethylase inhibitors
US9586902B2 (en) 2014-06-25 2017-03-07 Celgene Quanticel Research, Inc. Histone demethylase inhibitors
US9815828B2 (en) 2014-06-25 2017-11-14 Celgene Quanticel Research, Inc. Histone demethylase inhibitors
US10206930B2 (en) 2015-08-14 2019-02-19 New York University Methods for treating T-cell acute lymphoblastic leukemia
CN106608869B (en) * 2015-10-27 2020-08-04 中国科学院上海药物研究所 Histone demethylase JMJD3 inhibitor and preparation method and application thereof
CN106608869A (en) * 2015-10-27 2017-05-03 中国科学院上海药物研究所 Histone demethylase JMJD3 inhibitor, preparation method and application thereof
CN106854181A (en) * 2015-12-08 2017-06-16 湖南华腾制药有限公司 A kind of preparation method of pyrimidine compound
US10265279B2 (en) 2016-03-15 2019-04-23 Oryzon Genomics, S.A. Combinations of LSD1 inhibitors for use in the treatment of solid tumors
WO2017157825A1 (en) 2016-03-15 2017-09-21 F. Hoffmann-La Roche Ag Combinations of lsd1 inhibitors for use in the treatment of solid tumors
US11400099B2 (en) 2016-09-23 2022-08-02 Thomas Jefferson University Drug-induced epigenetic remodeling to prevent fibrosis
WO2018106984A1 (en) 2016-12-09 2018-06-14 Constellation Pharmaceuticals, Inc. Markers for personalized cancer treatment with lsd1 inhibitors
CN106810538A (en) * 2017-01-09 2017-06-09 湖南华腾制药有限公司 A kind of preparation method of 2 pyrrole radicals pyrimidine derivatives
EP3579834A4 (en) * 2017-02-10 2021-04-28 Temple Otorongo LLC Treatment of diabetes and associated metabolic conditions with epigenetic modulators
US11524011B2 (en) * 2017-09-01 2022-12-13 The Johns Hopkins University H3K27 demethylase inhibitors in pediatric and juvenile osteoporosis
WO2019046378A1 (en) * 2017-09-01 2019-03-07 The Johns Hopkins University H3k27 demethylase inhibitors in pediatric and juvenile osteoporosis
EP3793550A4 (en) * 2018-05-15 2022-04-13 Arbutus Biopharma Corporation Substituted 2,2'-bipyrimidinyl compounds, analogues thereof, and methods using same
CN113387938A (en) * 2020-03-11 2021-09-14 上海科技大学 Substituted pyrimidine compound, preparation method, intermediate and application thereof
CN113387938B (en) * 2020-03-11 2023-08-18 上海科技大学 Substituted pyrimidine compound, preparation method, intermediate and application thereof
CN115466245A (en) * 2022-06-29 2022-12-13 深圳大学 Hydroxamic acid derivative of pyrimidopyridine and preparation method and application thereof
CN115466245B (en) * 2022-06-29 2024-04-16 深圳大学 Hydroxamic acid derivative of pyrimidyl bipyridine, and preparation method and application thereof
US12083125B2 (en) 2022-07-07 2024-09-10 Thomas Jefferson University Drug-induced epigenetic remodeling to prevent fibrosis

Also Published As

Publication number Publication date
GB201112607D0 (en) 2011-09-07

Similar Documents

Publication Publication Date Title
WO2012052390A1 (en) N-2-(2-pyridinyl)-4-pyrimidinyl-beta-alanine derivatives as inhibitors of histone demethylase jmjd3
EP2330909B1 (en) Chemical compounds
US10336744B2 (en) 1AP E3 ligase directed proteolysis targeting chimeric molecules
JP5908493B2 (en) Indole
JP5889875B2 (en) Azaindazole
US9073924B2 (en) Azaindazoles
US9114141B2 (en) Indoles
JP5864546B2 (en) Indazole
EP2362775B1 (en) Chemical compounds
WO2013143597A1 (en) Demethylase enzymes inhibitors
US20080318947A1 (en) Inhibitors of Akt Activity
AU2014286342A1 (en) Benzothiophene derivatives as estrogen receptor inhibitors
WO2015000868A1 (en) Novel compounds
US20130225524A1 (en) Chemical Compounds
US20130303551A1 (en) Pyrimidinone derivatives as fatty acid synthase inhibitors
US20130172384A1 (en) Fatty acid synthase inhibitors
WO2008110508A1 (en) Pyrrolo-pyridine derivatives for the treatment of disorders associated with inappropriate ikk1 activity

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: 11772943

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 11772943

Country of ref document: EP

Kind code of ref document: A1