WO2012066336A1 - Benzylamine compounds as toll -like receptor 7 agonists - Google Patents

Benzylamine compounds as toll -like receptor 7 agonists Download PDF

Info

Publication number
WO2012066336A1
WO2012066336A1 PCT/GB2011/052246 GB2011052246W WO2012066336A1 WO 2012066336 A1 WO2012066336 A1 WO 2012066336A1 GB 2011052246 W GB2011052246 W GB 2011052246W WO 2012066336 A1 WO2012066336 A1 WO 2012066336A1
Authority
WO
WIPO (PCT)
Prior art keywords
amino
methyl
methylpyrimidin
compound
formula
Prior art date
Application number
PCT/GB2011/052246
Other languages
French (fr)
Inventor
Thomas Mcinally
Tobias Mochel
Futoshi Hasegawa
Seiji Hori
Original Assignee
Astrazeneca Ab
Dainippon Sumitomo Pharma Co., Ltd.
Astrazeneca Uk 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 Astrazeneca Ab, Dainippon Sumitomo Pharma Co., Ltd., Astrazeneca Uk Limited filed Critical Astrazeneca Ab
Publication of WO2012066336A1 publication Critical patent/WO2012066336A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/46Two or more oxygen, sulphur or nitrogen atoms
    • C07D239/48Two nitrogen atoms
    • C07D239/49Two nitrogen atoms with an aralkyl radical, or substituted aralkyl radical, attached in position 5, e.g. trimethoprim
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • 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
    • C07D403/10Heterocyclic 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 linked by a carbon chain containing aromatic rings
    • 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
    • C07D403/12Heterocyclic 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 linked by a chain containing hetero atoms as chain links

Definitions

  • the present invention relates to novel benzylamine compounds and, more particularly, to certain benzylamine compounds that act as TLR7 agonists and at the same time show advantageous selectivity over TLR8 and hERG.
  • This invention also relates to methods for the preparation of such compounds and intermediates useful in the preparation thereof, to pharmaceutical compositions containing such compounds, to the use of such compounds in the preparation of medicaments, and to the use of such compounds in the treatment of conditions mediated by TLR7, such as allergic diseases, autoimmune diseases, viral diseases and, in particular, cancer.
  • TLRs Toll-like receptors
  • DCs dendritic cells
  • TLRs recognise molecular motifs on pathogens called pathogen-associated molecular patterns (PAMPs).
  • PAMPs pathogen-associated molecular patterns
  • TLRs recognise different pathogen-derived ligands, for example TLRs 2 (bacterial lipoproteins), 3 (double-stranded RNA/poly (I:C)), 4 (lipopolysaccharides), 5 (flagellin), 7 (single- stranded RNA) and 9 (CpG-containing DNA).
  • Ligation of TLRs on antigen-presenting cells, such as DCs leads to production of proinflammatory cytokines, DC maturation and priming of the adaptive immune system.
  • TLR7 and TLR9 are expressed by plasmacytoid dendritic cells (pDCs) and ligand recognition leads to the secretion of interferon-a (INF-a).
  • pDCs plasmacytoid dendritic cells
  • IFN-a interferon-a
  • TLR7 agonists include the imidazoquinoline, imiquimod, which has been used to treat a number of dermatological conditions e.g. genital warts, molluscum contagiosum and melanoma.
  • imiquimod which has been used to treat a number of dermatological conditions e.g. genital warts, molluscum contagiosum and melanoma.
  • topically applied imiquimod (AldaraTM, Graceway Pharmaceuticals, Bristol, TN) demonstrated therapeutic responses in cutaneous metastatic melanoma and lentigo maligna and has been approved for the treatment of superficial basal cell carcinoma (BCC).
  • 852A is another imidazoquinoline, which unlike imiquimod, is suitable for systemic administration.
  • 852A is in phase II clinical trials in a number of cancer indications, including melanoma.
  • TLR7 agonists which are expected to be more effective in the treatment of disease, for example cancer, by reason of their superior potency and/or advantageous physical properties (for example, higher
  • TLR7 agonists for example 852A.
  • the benzylamine compounds of the present invention are capable of activating TLR7 in vitro.
  • the compounds of the present invention are expected to have value in the prevention or treatment of human disease, for example cancer, either as a monotherapy or in combination with other chemotherapeutic agents or radiotherapy regimens.
  • TLR7 and TLR8 differ in their cellular expression and as a result stimulation with selective agonists induces different cytokine profiles.
  • TLR8 stimulation results in enhanced levels of pro-inflammatory cytokines including TNFa, IL- ⁇ and IL-6 (Gorden et al (2005) J. Immunol. 174, 1259-1268). Conversely, TLR8 stimulation results in lower levels of IFNa.
  • a TLR7 selective agonist would favour induction of IFNa, which is important in suppression of Th2 cytokines (Huber et al (2010) J. Immunol. 185; 813-817) that are elevated in allergic disease.
  • Th2 cytokines Hauber et al (2010) J. Immunol. 185; 813-817) that are elevated in allergic disease.
  • compounds selective for TLR7 compared to TLR8 the induction of proinflammatory cytokines would be reduced thus avoiding inflammatory responses in man.
  • compounds of the invention also have a surprisingly advantageous hERG profile.
  • Compounds that have significant activity against the hERG ion channel are disfavoured because such activity is implicated in the
  • n 1 or 2;
  • R 1 is Ci_ 4 alkyl, methoxyC 2 - 3 alkyl, hydroxyC 2 _ 4 alkyl, -(CH 2 ) y CONH 2 wherein y is 1 or 2, or azetidin-3-yl, wherein said azetidin-3-yl is optionally substituted on nitrogen by Ci_ 3 alkyl; and
  • R 2 is hydrogen or Ci_ 4 alkyl
  • R 1 and R 2 together with the nitrogen to which they are attached form a pyrrolidin-l-yl, lH-imidazol-l-yl or a piperazin-l-yl; wherein said piperazin-l-yl is optionally substituted on the available nitrogen by Ci_ 3 alkyl;
  • the "available nitrogen" of a piperazin-l-yl is at the 4-position of the piperazin-l-yl.
  • optically active or racemic forms may exist in optically active or racemic forms by virtue of one or more asymmetric carbon atoms
  • the invention includes in its definition any such optically active or racemic form which possesses the above-mentioned activity.
  • the synthesis of optically active forms may be carried out by standard techniques of organic chemistry well known in the art, for example by synthesis from optically active starting materials or by resolution of a racemic form.
  • the above-mentioned activity may be evaluated using the standard laboratory techniques referred to hereinafter.
  • Ci_ 4 alkyl is intended to mean a saturated carbon chain of 1 to 4 carbon atoms in length which may be straight-chained or branched. However references to individual alkyl groups such as “propyl” are specific for the straight chain version only and references to individual branched-chain alkyl groups such as tert-butyl are specific for the branched chain version only. For example, “Ci_ 4 alkyl” includes methyl, ethyl, propyl, isopropyl, butyl, n-butyl, isobutyl and tert-butyl. The term “Ci_3alkyl” or “C 2 _3alkyl” is to be construed accordingly.
  • R 1 , R 2 and n may take any of the values defined herein for R 1 , R 2 and n respectively.
  • variable groups R 1 , R 2 and n may also take the values as indicated below. Such values may be used together with any of the definitions, claims, aspects or embodiments defined herein to provide further embodiments or claims of the invention, and unless the context does not permit, any number of said variable group definitions may be used in any combination with each other to form further embodiments, aspects and claims.
  • R 1 is Ci_ 4 alkyl.
  • R 1 is methoxyC 2 - 3 alkyl.
  • R 1 is hydroxyC 2 - 4 alkyl.
  • R 1 is -(CH 2 ) y CONH 2 wherein y is 1 or 2.
  • R 1 is azetidin-3-yl, wherein said azetidin-3-yl is optionally substituted on nitrogen by Ci_ 3 alkyl.
  • R 1 is methyl, 2-hydroxy ethyl, azetidin-3-yl, l-methylazetidin-3-yl, 2- methoxyethyl or -(CH 2 ) y CONH 2 wherein y is 1 or 2; and R 2 is hydrogen or methyl; or R 1 and R 2 together with the nitrogen to which they are attached form a piperazin-l-yl, lH-imidazol-l-yl, 4-methylpiperazin-l-yl or pyrrolidin-l-yl.
  • R 1 is methyl or ethyl.
  • R 1 is 2-hydroxyethyl.
  • R 1 is azetidin-3-yl.
  • R 1 is l-methylazetidin-3-yl.
  • R 1 is -CH 2 CONH 2 .
  • R 1 is -CH 2 CH 2 CONH 2 .
  • R 2 is hydrogen
  • R 2 is Ci_ 4 alkyl.
  • R 2 is methyl or ethyl.
  • R 1 is methyl, ethyl, 2-hydroxyethyl, azetidin-3-yl, l-methylazetidin-3-yl, 2- methoxyethyl or -(CH 2 ) y CONH 2 wherein y is 1 or 2; and R 2 is hydrogen or methyl.
  • R 1 and R 2 together with the nitrogen to which they are attached form a piperazin-l-yl, lH-imidazol-l-yl, 4-methylpiperazin-l-yl or pyrrolidin-l-yl.
  • the R 3 group of the compound of Formula (II) may be a substituted or
  • R 3 may be a hydrocarbyl group selected from benzyl and Ci_ 4 alkyl, for example methyl or ethyl.
  • the LG 1 group of the compound of Formula (III) may be any convenient leaving group which are well-known to the skilled person, for example a substituted or unsubstituted hydrocarbylsulfonyloxy leaving group, for example a tosylate or mesylate group, or a halo leaving group such as iodo or more conveniently, a bromo or chloro leaving group.
  • Step (i) - Compounds of Formula (IV) may be prepared by a standard alkylation reaction using compounds of Formula (II) and Formula (III): For example, reaction of a compound of Formula (III) with a base, such as NaH, in a suitable solvent, such as THF or DMF, at a suitable temperature, for example 0-20°C, followed by addition of a compound of Formula (II). The reaction may be heated, for example at 50-100°C, optionally in the presence of a catalytic amount of an iodide salt, such as KI.
  • a base such as NaH
  • a suitable solvent such as THF or DMF
  • Step (ii) - Compounds of Formula (V) may be prepared by reacting a compound of Formula (IV) with guanidine or guanidine carbonate in a suitable solvent, such as methanol or ethanol, at elevated temperature, for example 50-150°C.
  • a suitable solvent such as methanol or ethanol
  • the compound of Formula (V) may be isolated as a salt.
  • Step (iii) - Compounds of Formula (VI) may be prepared by reacting a compound of Formula (V) with POCI3, at elevated temperature, for example 50-110°C.
  • the compound of Formula (V) may be isolated as a salt.
  • Step (iv) - Compounds of Formula (VII) may be prepared by reacting a compound of Formula (VI) with an excess of the appropriate amino alcohol (where the amino alcohol may optionally have its alcohol group protected) in a suitable solvent, such as propionitrile, butanol or 1,4-dioxane, in the presence of trifluoroacetic acid at elevated temperature (for example 50-200°C) using conventional or microwave heating.
  • a suitable solvent such as propionitrile, butanol or 1,4-dioxane
  • trifluoroacetic acid at elevated temperature (for example 50-200°C) using conventional or microwave heating.
  • the R 4 group in the compound of Formula (VII) and the compound of Formula (VIII) is independently selected from hydrogen, a silicon-based protecting group or a benzyl-based protecting group.
  • R 4 is selected from hydrogen, trimethylsilyl, tert-butyldimethylsilyl and benzyl. In one embodiment R 4 is hydrogen. In one embodiment R 4 is a silicon-based protecting group or a benzyl-based protecting group. In one embodiment R 4 is a silicon- based protecting group. The silicon-based protecting group may be a tri(Ci_ 4 alkyl)silyl group, for example a trimethylsilyl group or a tert-butyldimethylsilyl group. In one embodiment R 4 is tert-butyldimethylsilyl.
  • the compound of Formula (VII) and/or the compound of Formula (VIII) has an (5)-stereocentre at the asymmetric carbon atom in the amino alcohol part of the molecule.
  • the optionally used protecting group incorporated into the compound of Formula (VII) and/or Formula (VIII) may be removed at any convenient point in the synthesis using standard deprotection conditions that are well known to the skilled person.
  • the compounds of Formula (VII) and the compound of Formula (VIII) may be isolated as a salt.
  • Step (v) - Compounds of Formula (VIII) may be prepared by reacting a compound of Formula (VII) with Raney nickel in a suitable solvent mixture such as pyridine, acetic acid and water, at a suitable temperature, for example 20-50°C.
  • a suitable solvent mixture such as pyridine, acetic acid and water
  • Step (vi) - Compounds of Formula (I) may be prepared by reaction of a compound of (VIII) with the appropriate amine (FTNR R 2 ) under reductive amination conditions, which are well known to the skilled person.
  • the reductive amination may be carried out using a suitable reducing agent such as sodium triacetoxyborohydride in a suitable solvent such as dichloromethane and acetic acid in the presence of activated molecular sieves, or by using NaBH 4 in a suitable solvent such as methanol.
  • the R 5 group of the compound of Formula (IX) may be a substituted or unsubstituted hydrocarbyl group.
  • R 5 may be a hydrocarbyl group selected from benzyl and Ci_ 4 alkyl, for example methyl or ethyl.
  • the LG 2 group of the compound of Formula (X) may be any convenient leaving group which are well-known to the skilled person, for example a substituted or unsubstituted hydrocarbylsulfonyloxy leaving group, for example a tosylate or mesylate group, or a halo leaving group such as iodo or more conveniently, a bromo or chloro leaving group.
  • R 6 group of the compound of Formula (X) may be a substituted or unsubstituted hydrocarbyl group.
  • R 6 may be a hydrocarbyl group selected from benzyl and Ci_ 4 alkyl, for example methyl or ethyl.
  • Step (i) - Compounds of Formula (XI) may be prepared by a standard alkylation reaction using compounds of Formula (IX) and Formula (X): For example, reaction of a compound of Formula (IX) with a base, such as NaH, in a suitable solvent, such as THF or DMF, at a suitable temperature, for example 0-20°C, followed by addition of a compound of Formula (X). The reaction may be heated, for example at 50-100°C, optionally in the presence of a catalytic amount of an iodide salt, such as KI.
  • a base such as NaH
  • a suitable solvent such as THF or DMF
  • Step (ii) - Compounds of Formula (XII) may be prepared by reacting a compound of Formula (IV) with guanidine or guanidine carbonate in a suitable solvent, such as methanol or ethanol, at elevated temperature, for example 50-150°C.
  • a suitable solvent such as methanol or ethanol
  • the compound of Formula (XII) may be isolated as a salt.
  • R 7 group in the compound of Formula (XIII) may be a substituted or unsubstituted hydrocarbyl group, for example a Ci_ 4 alkyl group or a phenyl group optionally substituted by 1 , 2 or 3 Ci_ 4 alkyl groups, for example R 7 may conveniently be methyl, ethyl, 2,4,6-trimethylphenyl or 2,4,6-triisopropylphenyl.
  • (XIII) may be prepared by reacting a compound of Formula (XII) with a hydrocarbyl sulfonyl chloride: R 7 S0 2 C1, for example trimethylbenzene sulfonyl chloride or
  • reaction may be carried out in a suitable solvent, such THF in the presence of a suitable base, such as, triethylamine or tetramethyl- propylenediamine, at a suitable temperature, for example 0-50°C.
  • a suitable solvent such as THF
  • a suitable base such as, triethylamine or tetramethyl- propylenediamine
  • Step (iv) - Compounds of Formula (XIV) may be prepared by reacting a compound of Formula (XIII) with an excess of the appropriate amino alcohol (where the amino alcohol may optionally have its alcohol group protected) in a suitable solvent, such as propionitrile, butanol or 1 ,4-dioxane, in the presence of trifluoroacetic acid at elevated temperature (for example 50-200°C) using conventional or microwave heating.
  • a suitable solvent such as propionitrile, butanol or 1 ,4-dioxane
  • trifluoroacetic acid at elevated temperature (for example 50-200°C) using conventional or microwave heating.
  • the R 4 group in the compound of Formula (XIV) and the compound of Formula (XV) is independently selected from hydrogen, a silicon-based protecting group or a benzyl-based protecting group.
  • R 4 is selected from hydrogen, trimethylsilyl, tert- butyldimethylsilyl and benzyl. In one embodiment R 4 is hydrogen. In one embodiment R 4 is a silicon-based protecting group or a benzyl-based protecting group. In one embodiment R 4 is a silicon-based protecting group. The silicon-based protecting group may be a tri(Ci_ 4 alkyl)silyl group, for example a trimethylsilyl group or a tert-butyldimethylsilyl group. In one embodiment R 4 is tert-butyldimethylsilyl.
  • the compound of Formula (XIV) has an (5)-stereocentre at the asymmetric carbon atom in the amino alcohol part of the molecule.
  • the optionally used protecting group incorporated into the compound of Formula (XIV) and/or Formula (XV) may be removed at any convenient point in the synthesis using standard deprotection conditions that are well known to the skilled person.
  • the compound of Formula (XIV) may be isolated as a salt.
  • Step (v) - Compounds of Formula (XV) may be prepared by reacting a compound of Formula (XIV) with any standard reducing agent known to be capable of reducing an ester to the corresponding alcohol.
  • any standard reducing agent known to be capable of reducing an ester to the corresponding alcohol.
  • LiBH 4 may be used, in a suitable solvent mixture such as THF and methanol at a suitable temperature, for example 20-60°C.
  • the compound of Formula (XV) may be isolated as a salt.
  • Step (vi) - Compounds of Formula (VIII) may be prepared by reacting a compound of Formula (XV) in the presence of a suitable oxidising agent such as Mn0 2 in a suitable solvent mixture such as THF and CH 2 C1 2 at a suitable temperature, for example 20-60°C.
  • a suitable oxidising agent such as Mn0 2
  • a suitable solvent mixture such as THF and CH 2 C1 2
  • Step (vii) - Compounds of Formula (I) may be prepared by reaction of a compound of (VIII) with the appropriate amine (FTNR R 2 ) under reductive amination conditions, which are well known to the skilled person.
  • the reductive amination may be carried out using a suitable reducing agent such as sodium triacetoxyborohydride in a suitable solvent such as dichloromethane and acetic acid in the presence of activated molecular sieves, or by using NaBH 4 in a suitable solvent such as methanol.
  • the compound of Formula (XIV- A) is a compound of Formula (XIV) as hereinbefore defined, wherein R 4 is hydrogen.
  • the compounds of Formula (XIV-B) and (XV-A) are compounds of Formula (XIV) and (XV) respectively, as hereinbefore defined, wherein R 4 is a silicon protecting group, for example a tri(Ci_ 4 alkyl)silyl protecting group (where each of the three Ci_ 4 alkyl groups may be the same or different).
  • R 4 is a tert-butyldimethylsilyl (TBDMS) protecting group, or a salt thereof.
  • R 4 is a tert-butyldimethylsilyl (TBDMS) protecting group, or a salt thereof.
  • Step (i) Compounds of Formula (XIV-B) may be prepared reaction of compounds of Formula (XIV- A) by reaction with tri(Ci_ 4 alkyl)silyl chloride, for example tert- butyldimethylsilyl chloride, in the presence of a base, for example an organic nitrogen base, for example an amine base such as triethylamine in a suitable solvent such as DMF at a suitable temperature, for example 20°C.
  • a base for example an organic nitrogen base, for example an amine base such as triethylamine in a suitable solvent such as DMF at a suitable temperature, for example 20°C.
  • Step (ii) Compounds of Formula (XV- A) may be prepared from compounds of Formula (XIV-B) by reacting with a reducing agent that is known to the skilled person for its capability for reducing esters to alcohols, for example LiBH 4 , in a suitable solvent mixture such as THF and methanol, at a suitable temperature, for example 20-60°C
  • a reducing agent that is known to the skilled person for its capability for reducing esters to alcohols, for example LiBH 4
  • a suitable solvent mixture such as THF and methanol
  • LG 3 group in the compound of Formula (XVI) is a leaving group where suitable leaving groups are well known to the skilled person.
  • LG 3 may be chloro or bromo.
  • Compounds of Formula (XVI) may be prepared by reacting a compound of Formula (XV- A) with a hydrocarbyl sulfonyl bromide or chloride in the presence of LiBr or LiCl, for example CH3SO 2 CI with LiCl, in a suitable solvent such as THF at a suitable temperature, for example 20°C, followed by treatment with an acid such as HC1 in dioxane, a suitable solvent such as methanol at a suitable temperature, for example 20°C.
  • a suitable solvent such as THF at a suitable temperature, for example 20°C
  • an acid such as HC1 in dioxane
  • a suitable solvent such as methanol
  • Compounds of Formula (XIV) may be converted into a compound of Formula (I) by an alkylation reaction using the appropriate amine (FfNR ⁇ R 2 ) using alkylation conditions that are well known to the skilled person.
  • the alkylation may be achieved using a suitable base such as Na 2 C0 3 , in a suitable solvent, such as acetonitrile.
  • the compound of Formula (XVI) may be isolated as a salt.
  • a compound of Formula (XVI), as defined herein, or a salt thereof in one embodiment, LG 3 is chloro.
  • the compound of Formula (XVI) has an (5)-stereocentre at the asymmetric carbon atom in the amino alcohol part of the molecule.
  • LG 4 and LG 5 are each independently a suitable leaving group wherein LG 4 in a compound of Formula (XIX) is not removed in the reaction conditions used in Step (iii) and Step (iv).
  • LG 4 group may be a hydrocarbyl sulphonyl group: -OS0 2 R 8 wherein R 8 may be a optionally substituted phenyl group such as /?-methylphenyl, 2,4,6- trimethylphenyl or 2,4,6-triisopropylphenyl.
  • LG 5 group may be a halogen such as chloro, bromo, iodo or alternatively may be a hydrocarbyl sulphonyl group: -OS0 2 R 9 wherein R 9 may be a Ci_ 4 alkyl such as methyl.
  • Step (i) - A compound of Formula (XVII) may be prepared by reacting a compound of Formula (XII) with any standard reducing agent known to be capable of reducing an ester to the corresponding alcohol.
  • any standard reducing agent known to be capable of reducing an ester to the corresponding alcohol.
  • L1AIH 4 or LiBHEt 3 may be used, in a suitable solvent mixture such as THF and methanol at a suitable temperature, for example 20-60°C.
  • a compound of Formula (XVIII) may be prepared by reacting a compound of Formula (XVII) with a hydrocarbyl sulphonyl chloride: R 8 S0 2 C1, for example 2, 4, 6-trimethylbenzene sulphonyl chloride or 2, 4, 6-triisopropylbenzenesulfonyl chloride.
  • the reaction may be carried out in a suitable solvent, such as THF in the presence of a suitable base, such as, triethylamine or diisopropylethylamine, at a suitable temperature, for example 0-100°C.
  • a compound of Formula (XIX) may be prepared by reacting a compound of Formula (XVIII) with a hydrocarbyl sulphonyl chloride: R 9 S0 2 C1, for example methane sulphonyl chloride when R 9 is methyl.
  • the reaction may be carried out in a suitable solvent, such THF in the presence of a suitable base, such as, triethylamine or diisopropylethylamine, and optionally lithium chloride or lithium bromide when LG 5 is halogen, at a suitable temperature, for example 0-100°C, preferably 0-30°C.
  • Step (iv) - A compound of Formula (XIX) may be successively converted to a compound of Formula (XX) by an alkylation reaction using the appropriate amine
  • alkylation may be achieved using a suitable base such as Na 2 C0 3 , K 2 C0 3 , or diisopropyethylamine in a suitable solvent, such as acetonitrile or THF.
  • a suitable base such as Na 2 C0 3 , K 2 C0 3 , or diisopropyethylamine in a suitable solvent, such as acetonitrile or THF.
  • a suitable solvent such as acetonitrile or THF.
  • Step (v) - A compound of Formula (I) may be prepared by reacting a compound of Formula (XX) with an excess of the appropriate amino alcohol (where the amino alcohol may optionally have its alcohol group protected) in a suitable solvent, such as propionitrile, butanol or 1,4-dioxane, in the presence of trifluoroacetic acid at elevated temperature (for example 50-200°C) using conventional or microwave heating.
  • a suitable solvent such as propionitrile, butanol or 1,4-dioxane
  • trifluoroacetic acid at elevated temperature (for example 50-200°C) using conventional or microwave heating.
  • a suitable pharmaceutically-acceptable salt of a compound of the Formula (I) is, for example, an acid-addition salt of a compound of the Formula (I), for example an acid- addition salt with an inorganic or organic acid such as hydrochloric, hydrobromic, sulphuric, trifluoroacetic, citric or maleic acid.
  • an inorganic or organic acid such as hydrochloric, hydrobromic, sulphuric, trifluoroacetic, citric or maleic acid.
  • the compounds of the invention may be administered in the form of a pro-drug, i.e. a compound that is broken down in the human or animal body to release a compound of the invention.
  • a pro-drug may be used to alter the physical properties and/or the pharmacokinetic properties of a compound of the invention.
  • a pro-drug can be formed when the compound of the invention contains a suitable group or substituent to which a property-modifying group can be attached.
  • Examples of pro-drugs include in-vivo cleavable amide derivatives that may be formed at an amino group in a compound of the Formula (I).
  • the present invention includes those compounds of the Formula (I) as defined hereinbefore when made available by organic synthesis and when made available within the human or animal body by way of cleavage of a pro-drug thereof. Accordingly, the present invention includes those compounds of the Formula (I) that are produced by organic synthetic means and also such compounds that are produced in the human or animal body by way of metabolism of a precursor compound, that is a compound of the Formula (I) may be a synthetically-produced compound or a metabolically-produced compound.
  • a suitable pharmaceutically-acceptable pro-drug of a compound of the Formula (I) is one that is based on reasonable medical judgement as being suitable for administration to the human or animal body without undesirable pharmacological activities and without undue toxicity.
  • pro-drug Various forms of pro-drug have been described, for example in the following documents:- a) Methods in Enzymology, Vol. 42, p. 309-396, edited by K. Widder, et al.
  • a suitable pharmaceutically-acceptable pro-drug of a compound of the Formula (I) that possesses an amino group is, for example, an in-vivo cleavable amide derivative thereof.
  • Suitable pharmaceutically-acceptable amides from an amino group include, for example an amide formed with Ci_ioalkanoyl groups such as an acetyl, benzoyl, phenylacetyl and substituted benzoyl and phenylacetyl groups. Examples of ring substituents on the phenylacetyl and benzoyl groups include aminomethyl,
  • the in-vivo effects of a compound of the Formula (I) may be exerted in part by one or more metabolites that are formed within the human or animal body after administration of a compound of the Formula (I). As stated hereinbefore, the in-vivo effects of a compound of the Formula (I) may also be exerted by way of metabolism of a precursor compound (a pro-drug).
  • a pharmaceutical composition which comprises a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore in association with a pharmaceutically- acceptable diluent or carrier.
  • the pharmaceutical composition may be used in the treatment of cancer.
  • the composition may be in a form suitable for oral administration, for example as a tablet or capsule; for parenteral injection (including intravenous, subcutaneous, intramuscular, intravascular or infusion) as a sterile solution, suspension or emulsion; for topical administration as an ointment or cream; or for rectal administration as a
  • the compound of Formula (I), or a pharmaceutically acceptable salt thereof could also be administered as an air spray for inhalation.
  • the air spray e.g., spray, aerosol, dry powder preparation, etc.
  • a dry powder preparation may also be used.
  • An aerosol appropriate for inhalation may be either a suspension or solution, and would typically contain the compound of Formula (I), or a pharmaceutically acceptable salt thereof, and any appropriate propellants such as a fluorocarbon or hydrogen-containing chlorofluorocarbon or a mixture thereof.
  • hydro fluoroalkane particularly 1,1,1,2-tetrafluoroethane, heptafluoroalkane (HFA) such as 1,1,1,2,3,3,3-heptafluoro-n-propane, or a mixture thereof.
  • HFA heptafluoroalkane
  • An aerosol may optionally contain an additional preparation excipient well-known to those skilled in the art such as surfactant (e.g., oleic acid or lecithin) and cosolvent (e.g., ethanol), etc.
  • an aerosol preparation could be delivered using the inhaler known as
  • the compound of the invention may be admixed with an adjuvant or a carrier, for example, lactose, saccharose, sorbitol, mannitol; a starch, for example, potato starch, corn starch or amylopectin; a cellulose derivative; a binder, for example, gelatine or polyvinylpyrrolidone; and/or a lubricant, for example, magnesium stearate, calcium stearate, polyethylene glycol, a wax, paraffin, and the like, and then compressed into tablets.
  • an adjuvant or a carrier for example, lactose, saccharose, sorbitol, mannitol
  • a starch for example, potato starch, corn starch or amylopectin
  • a cellulose derivative for example, gelatine or polyvinylpyrrolidone
  • a lubricant for example, magnesium stearate, calcium stearate, polyethylene glycol, a wax
  • the cores may be coated with a concentrated sugar solution which may contain, for example, gum arabic, gelatine, talcum and titanium dioxide.
  • a concentrated sugar solution which may contain, for example, gum arabic, gelatine, talcum and titanium dioxide.
  • the tablet may be coated with a suitable polymer dissolved in a readily volatile organic solvent.
  • the compound of the invention may be admixed with, for example, a vegetable oil or polyethylene glycol.
  • Hard gelatine capsules may contain granules of the compound using either the above-mentioned excipients for tablets.
  • liquid or semisolid formulations of the compound of the invention may be filled into hard gelatine capsules.
  • Liquid preparations for oral application may be in the form of syrups or
  • suspensions for example, solutions containing the compound of the invention, the balance being sugar and a mixture of ethanol, water, glycerol and propylene glycol.
  • liquid preparations may contain colouring agents, flavouring agents, saccharine and/or carboxymethylcellulose as a thickening agent or other excipients known to those skilled in art.
  • the compound of Formula (I) will normally be administered to a warm-blooded animal at a unit dose within the range 5-5000 mg/m 2 body area of the animal, i.e.
  • a unit dose form such as a tablet or capsule will usually contain, for example 1-250 mg of active ingredient.
  • a daily dose in the range of 1-50 mg/kg is employed.
  • the daily dose will necessarily be varied depending upon the host treated, the particular route of administration, and the severity of the illness being treated. Accordingly the optimum dosage may be determined by the practitioner who is treating any particular patient.
  • treatment is intended to have its normal everyday meaning of dealing with a disease in order to entirely or partially relieve one, some or all of its symptoms, or to correct or compensate for the underlying pathology.
  • prophylaxis is intended to have its normal everyday meaning and includes primary prophylaxis to prevent the development of the disease and secondary prophylaxis whereby the disease has already developed and the patient is temporarily or permanently protected against exacerbation or worsening of the disease or the development of new symptoms associated with the disease.
  • the compounds defined in the present invention are effective activators of TLR7 in vitro. Accordingly, the compounds of the present invention are expected to be potentially useful agents in the treatment of diseases or medical conditions mediated alone or in part by TLR7. For example, the following diseases and conditions listed in paragraphs 1 to 8 below may be treatable with compounds of the present invention.
  • obstructive diseases of the airways including: asthma, including bronchial, allergic, intrinsic, extrinsic, exercise-induced, drug-induced (including aspirin and NSAID-induced) and dust-induced asthma, both intermittent and persistent and of all severities, and other causes of airway hyper-responsiveness; chronic obstructive pulmonary disease (COPD); bronchitis, including infectious and eosinophilic bronchitis; emphysema; bronchiectasis; cystic fibrosis; sarcoidosis; farmer's lung and related diseases;
  • COPD chronic obstructive pulmonary disease
  • lung fibrosis including cryptogenic fibrosing alveolitis, idiopathic interstitial pneumonias, fibrosis complicating anti-neoplastic therapy and chronic infection, including tuberculosis and aspergillosis and other fungal infections; complications of lung transplantation; vasculitic and thrombotic disorders of the lung vasculature, and pulmonary hypertension; antitussive activity including treatment of chronic cough associated with inflammatory and secretory conditions of the airways, and iatrogenic cough; acute and chronic rhinitis including rhinitis medicamentosa, and vasomotor rhinitis; perennial and seasonal allergic rhinitis including rhinitis nervosa (hay fever); nasal polyposis; acute viral infection including the common cold, and infection due to respiratory syncytial virus, influenza, coronavirus (including SARS) and adenovirus;
  • eyes blepharitis; conjunctivitis, including perennial and vernal allergic
  • nephritis including interstitial and glomerulonephritis; nephrotic syndrome; cystitis including acute and chronic (interstitial) cystitis and Hunner's ulcer; acute and chronic urethritis, prostatitis, epididymitis, oophoritis and salpingitis; vulvovaginitis; Peyronie's disease; erectile dysfunction (both male and female);
  • allograft rejection acute and chronic following, for example, transplantation of kidney, heart, liver, lung, bone marrow, skin or cornea or following blood transfusion; or chronic graft versus host disease;
  • thrombocytopaenic purpura eosinophilic fasciitis, hyper-IgE syndrome, antiphospholipid syndrome and Sazary syndrome;
  • oncology treatment of common cancers including bladder, head and neck, prostate, breast, lung, ovarian, pancreatic, bowel and colon, stomach, skin and brain tumors and malignancies affecting the bone marrow (including the leukaemias) and
  • lymphoproliferative systems such as Hodgkin's and non-Hodgkin's lymphoma; including the prevention and treatment of metastatic disease and tumour recurrences, and
  • infectious diseases virus diseases such as genital warts, common warts, plantar warts, hepatitis B, hepatitis C, herpes simplex virus, molluscum contagiosum, variola, human immunodeficiency virus (HIV), human papilloma virus (HPV), cytomegalovirus (CMV), varicella zoster virus (VZV), rhinovirus, adenovirus, coronavirus, influenza, parainfluenza; bacterial diseases such as tuberculosis and mycobacterium avium, leprosy; other infectious diseases, such as fungal diseases, chlamydia, Candida, aspergillus, cryptococcal meningitis, Pneumocystis carnii, cryptosporidiosis, histoplasmosis, toxoplasmosis, trypanosome infection and leishmaniasis.
  • virus diseases such as genital warts, common warts, plantar wart
  • the compound of Formula (I) will be administered to a mammal, more particularly a human being.
  • the compound of Formula (I) will be administered to a mammal, more particularly a human being.
  • said disease mediated through TLR7 is cancer.
  • said cancer is selected from bladder cancer, head and neck cancer, prostate cancer, breast cancer, lung cancer, uterus cancer, pancreatic cancer, liver cancer, renal cancer, ovarian cancer, colon cancer, stomach cancer, skin cancer, cerebral tumor, malignant myeloma and lymphoproliferative tumors.
  • said disease mediated through TLR7 is asthma, COPD, allergic rhinitis, allergic conjunctivitis, atopic dermatitis, hepatitis B, hepatitis C, HIV, HPV, bacterial infections or dermatosis.
  • a compound of Formula (I) as defined hereinbefore, or a pharmaceutically acceptable salt thereof for the preparation of a medicament for the treatment of a disease mediated through TLR7.
  • said disease mediated through TLR7 is cancer.
  • said cancer is selected from bladder cancer, head and neck cancer, prostate cancer, breast cancer, lung cancer, uterus cancer, pancreatic cancer, liver cancer, renal cancer, ovarian cancer, colon cancer, stomach cancer, skin cancer, cerebral tumor, malignant myeloma and lymphoproliferative tumors.
  • said disease mediated through TLR7 is asthma, COPD, allergic rhinitis, allergic conjunctivitis, atopic dermatitis, hepatitis B, hepatitis C, HIV, HPV, bacterial infections or dermatosis.
  • said cancer is selected from bladder cancer, head and neck cancer, prostate cancer, breast cancer, lung cancer, uterus cancer, pancreatic cancer, liver cancer, renal cancer, ovarian cancer, colon cancer, stomach cancer, skin cancer, cerebral tumor, malignant myeloma and lymphoproliferative tumors.
  • a compound of Formula (I) as defined hereinbefore, or a pharmaceutically acceptable salt thereof for the preparation of a medicament for the treatment of asthma, COPD, allergic rhinitis, allergic conjunctivitis, atopic dermatitis, hepatitis B, hepatitis C, HIV, HPV, bacterial infections or dermatosis.
  • a method of using compound of Formula (I) as defined hereinbefore, or a pharmaceutically acceptable salt thereof, for the treatment of cancer comprises administering to said animal an effective amount of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, as defined herein.
  • said cancer is selected from bladder cancer, head and neck cancer, prostate cancer, breast cancer, lung cancer, uterus cancer, pancreatic cancer, liver cancer, renal cancer, ovarian cancer, colon cancer, stomach cancer, skin cancer, cerebral tumor, malignant myeloma and lymphoproliferative tumors.
  • a method of treating a human suffering from a disease in which activation of TLR7 is beneficial comprising the steps of administering to a person in need thereof of a therapeutically effective amount of a compound of Formula (I) as defined hereinbefore, or a
  • the disease in which activation of TLR7 is beneficial is cancer.
  • said cancer is selected from bladder cancer, head and neck cancer, prostate cancer, breast cancer, lung cancer, uterus cancer, pancreatic cancer, liver cancer, renal cancer, ovarian cancer, colon cancer, stomach cancer, skin cancer, cerebral tumor, malignant myeloma and lymphoproliferative tumors.
  • the disease in which activation of TLR7 is beneficial is asthma, COPD, allergic rhinitis, allergic conjunctivitis, atopic dermatitis, hepatitis B, hepatitis C, HIV, HPV, bacterial infections or dermatosis.
  • the cancer may be bladder cancer. In any aspect or embodiment described herein the cancer may be head and neck cancer.
  • the cancer may be prostate cancer.
  • the cancer may be breast cancer.
  • the cancer may be lung cancer.
  • the cancer may be uterus cancer.
  • the cancer may be pancreatic cancer.
  • the cancer may be liver cancer.
  • the cancer may be renal cancer.
  • the cancer may be ovarian cancer.
  • the cancer may be colon cancer.
  • the cancer may be stomach cancer.
  • the cancer may be skin cancer.
  • the cancer may be cerebral tumor.
  • the cancer may be malignant myeloma cancer.
  • anti-cancer treatment may be applied as a sole therapy or may involve, in addition to the compound of the invention, conventional surgery or radiotherapy or chemotherapy.
  • chemotherapy may include one or more of the following categories of anti-tumour agents :-
  • antiproliferative/antineoplastic drugs and combinations thereof as used in medical oncology, such as alkylating agents (for example cis-platin, oxaliplatin, carboplatin, miriplatin, cyclophosphamide, nitrogen mustard, melphalan, chlorambucil, busulphan, temozolamide and nitrosoureas); antimetabolites (for example gemcitabine and antifolates such as fluoropyrimidines like 5-fluorouracil and tegafur, raltitrexed, methotrexate, cytosine arabinoside, and hydroxyurea); antitumour antibiotics (for example anthracyclines like adriamycin, bleomycin, doxorubicin, daunomycin, epirubicin, amrubicin, idarubicin, mitomycin-C, dactinomycin and mithramycin); antimitotic agents (for example vinca alkal
  • cytostatic agents such as antioestrogens (for example tamoxifen, fulvestrant, toremifene, raloxifene, droloxifene and iodoxyfene), antiandrogens (for example bicalutamide, flutamide, nilutamide and cyproterone acetate), LHRH antagonists or LHRH agonists (for example goserelin, leuprorelin and buserelin), progestogens (for example megestrol acetate), aromatase inhibitors (for example as anastrozole, letrozole, vorazole and exemestane) and inhibitors of 5a-reductase such as finasteride;
  • antioestrogens for example tamoxifen, fulvestrant, toremifene, raloxifene, droloxifene and iodoxyfene
  • antiandrogens for example
  • anti-invasion agents for example c-Src kinase family inhibitors like 4-(6-chloro-2,3- methylenedioxyanilino)-7- [2-(4-methylpiperazin- 1 -yl)ethoxy] -5 -tetrahydropyran-4- yloxyquinazoline (AZD0530; International Patent Application WO 01/94341), N-(2- chloro-6-methylphenyl)-2- ⁇ 6-[4-(2-hydroxyethyl)piperazin- 1 -yl]-2-methylpyrimidin-4- ylamino ⁇ thiazole-5-carboxamide (dasatinib, BMS-354825; J. Med.
  • anti-invasion agents for example c-Src kinase family inhibitors like 4-(6-chloro-2,3- methylenedioxyanilino)-7- [2-(4-methylpiperazin- 1 -yl)ethoxy] -5
  • inhibitors of growth factor function include growth factor antibodies and growth factor receptor antibodies (for example the anti-erbB2 antibody trastuzumab [HerceptinTM], the anti-EGFR antibody panitumumab, the anti-erbBl antibody cetuximab [Erbitux, C225] and any growth factor or growth factor receptor antibodies disclosed by Stern et al. Critical reviews in oncology/haematology, 2005, Vol. 54, ppl 1-29); such inhibitors also include tyrosine kinase inhibitors, for example inhibitors of the epidermal growth factor family (for example EGFR family tyrosine kinase inhibitors such as
  • N-(3-chloro-4-fluorophenyl)-7-methoxy-6-(3-morpholinopropoxy)quinazolin-4-amine (gefitinib, ZD 1839), N-(3-ethynylphenyl)-6,7-bis(2-methoxyethoxy)quinazolin-4-amine (erlotinib, OSI-774) and 6-acrylamido-N-(3-chloro-4-fluorophenyl)-7-(3- morpholinopropoxy)-quinazolin-4-amine (CI 1033), erbB2 tyrosine kinase inhibitors such as lapatinib); inhibitors of the hepatocyte growth factor family; inhibitors of the insulin growth factor family; inhibitors of the platelet-derived growth factor family such as imatinib and/or nilotinib (AMN107); inhibitors of serine/threonine kinases (for example Ras/Raf signal
  • antiangiogenic agents such as those which inhibit the effects of vascular endothelial growth factor, [for example the anti-vascular endothelial cell growth factor antibody bevacizumab (AvastinTM) and for example, a VEGF receptor tyrosine kinase inhibitor such as vandetanib (ZD6474), vatalanib (PTK787), sunitinib (SU11248), axitinib (AG-013736), pazopanib (GW 786034) and 4-(4-fluoro-2-methylindol-5-yloxy)-6-methoxy-7-(3- pyrrolidin-l-ylpropoxy)quinazoline (AZD2171; Example 240 within WO 00/47212), compounds such as those disclosed in International Patent Applications W097/22596, WO 97/30035, WO 97/32856 and WO 98/13354 and compounds that work by other
  • vascular damaging agents such as Combretastatin A4 and compounds disclosed in International Patent Applications WO 99/02166, WO 00/40529, WO 00/41669,
  • an endothelin receptor antagonist for example zibotentan (ZD4054) or atrasentan;
  • antisense therapies for example those which are directed to the targets listed above, such as ISIS 2503, an anti-ras antisense;
  • (ix) gene therapy approaches including for example approaches to replace aberrant genes such as aberrant p53 or aberrant BRCA1 or BRCA2, GDEPT (gene-directed enzyme pro-drug therapy) approaches such as those using cytosine deaminase, thymidine kinase or a bacterial nitroreductase enzyme and approaches to increase patient tolerance to chemotherapy or radiotherapy such as multi-drug resistance gene therapy; and
  • GDEPT gene-directed enzyme pro-drug therapy
  • (x) immunotherapy approaches including for example ex-vivo and in-vivo approaches to increase the immunogenicity 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, approaches to decrease the function of immune suppressive cells such as regulatory T cells, myeloid-derived suppressor cells or IDO (indoleamine 2,3,-deoxygenase)-expressing dendritic cells, and approaches using cancer vaccines consisting of proteins or peptides derived from tumour-associated antigens such as NY-ESO-1, MAGE-3, WT1 or Her2/neu.
  • cytokines such as interleukin 2, interleukin 4 or granulocyte-macrophage colony stimulating factor
  • a pharmaceutical product comprising a compound of the formula (I) as defined hereinbefore and an additional anti-tumour substance as defined hereinbefore for the conjoint treatment of cancer.
  • a pharmaceutical product comprising a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore, and an additional anti-tumour substance as defined hereinbefore for the conjoint treatment of cancer.
  • a pharmaceutical product comprising a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore, and an additional anti-tumour substance for the conjoint treatment of cancer.
  • a combination suitable for use in the treatment of cancer comprising a compound of Formula (I) as defined hereinbefore, or a pharmaceutically acceptable salt thereof, and any one of the anti tumour agents listed under (i) - (ix) above.
  • a pharmaceutical composition which comprises a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in combination with an anti-tumour agent selected from one listed under (i) - (ix) herein above, in association with a pharmaceutically acceptable diluent or carrier.
  • a pharmaceutical composition which comprises a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in combination with an anti-tumour agent selected from one listed under (i) - (ix) herein above, in association with a pharmaceutically acceptable diluent or carrier for use in the treatment of cancer.
  • a compound of the Formula (I), or a pharmaceutically acceptable salt thereof in combination with an anti- tumour agent selected from one listed under (i) - (ix) herein above for use in the treatment of cancer in a warm-blooded animal, such as man.
  • a method of treating cancer in a warm-blooded animal, such as man, in need of such treatment which comprises administering to said animal an effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in combination with an anti-tumour agent selected from one listed under (i) - (ix) herein above.
  • kits comprising a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in combination with an anti-tumour agent selected from one listed under (i) - (ix) herein above.
  • a kit comprising:
  • an anti-tumour agent selected from one listed under (i) - (ix) herein above; in a second unit dosage form;
  • a kit comprising a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in combination with an additional anti-tumour agent.
  • a kit comprising:
  • the compounds of Formula (I) may be useful as vaccine adjuvants.
  • the invention still further provides a method of treating, or reducing the risk of, a disease or condition, which method comprises administering to a patient in need thereof a therapeutically effective amount of a vaccine and a compound of formula (I), or a pharmaceutically acceptable salt thereof, as defined herein.
  • the invention still further provides a method of increasing the response to a vaccine in a patient, which method comprises administering to a patient in need thereof a therapeutically effective amount of a vaccine and a compound of formula (I), or a pharmaceutically acceptable salt thereof, as defined herein.
  • W091, W092 or W093 packed with silica gel that is suface- modified by aminopropyl purchased from Yamazen Science, Inc);
  • DMSO dimethylsulfoxide;
  • sat. saturated aqueous solution;
  • RT room temperature;
  • LC- MS liquid chromatography with mass spectrometry;
  • m/z measured mass to charge ratio;
  • SCX refers to solid phase extraction with a sulfonic acid sorbent;
  • M molarity;
  • EtOH ethanol.
  • Proton nuclear magnetic resonance data (' a H NMR”) was generally obtained at 300-500 MHz and using deuterated DMSO unless otherwise stated.
  • the title compound may be prepared by the sequence of steps described below:
  • Methyl 4-[(2-amino-4-hydroxy-6-methylpyrimidin-5-yl)methyl]-3-methoxybenzoate (8.6 g) was added to POCl 3 and the mixture was heated at 100°C for 15h. The solvent was then removed in vacuo. The resulting residue was diluted with water and the pH adjusted to 7 by the addition of sat. NaHCOs. This mixture was heated at 50°C for 2h, and was then extracted with EtOAc.
  • Methyl 4-[(2-amino-4- ⁇ [(25)- 1 -hydroxypentan-2-yl]amino ⁇ -6-methylpyrimidin-5- yl)methyl]-3-methoxybenzoate (220 mg) was dissolved into a mixture of THF (3 mL) and water (3 mL), and LiOH solution (3M, 0.944 mL) was added. The mixture was heated in a CEM DiscoverTM microwave for 6h at 120°C. The mixture was concentrated in vacuo and the resulting residue was dissolved in water and the mixture acidified to pH7 using 2M HCl.
  • BH 3 -THF complex (1M, 3 mL) was added to 4-[(2-amino-4- ⁇ [(25)-l-hydroxypentan-2- yl]amino ⁇ -6-methylpyrimidin-5-yl)methyl]-3-methoxybenzoic acid (85 mg) and the mixture was heated to 80°C for 2h. CH 3 OH was then carefully added and the mixture was heated to 80°C for lh. The mixture was concentrated in vacuo and the resulting residue was dissolved in CH 3 OH and loaded onto an SCX cartridge. The cartridge was washed with CH 3 OH and then eluted with a solution of 0.7M NH 3 in CH 3 OH.
  • the title compound may be prepared according to the sequence of steps described below:
  • Example 1 A ( 2S)-2- ⁇ ( 2- Amino-5- ⁇ 4- ⁇ ( dimethylamino)methyll -2-methoxybenzyl ⁇ -6- methylpyrimidin-4-yl)aminolpentan-l-ol bis saccharin salt
  • the title compound may be prepared by the sequence of steps described below:
  • LiAlH 4 (8.4 mg) was added to a solution of tert-butyl 3-( ⁇ 4-[(2-amino-4- ⁇ [(2S)-l- hydroxypentan-2-yl] amino ⁇ -6-methylpyrimidin-5 -yl)methyl] -3 -methoxybenzyl ⁇ - amino)azetidine-l-carboxylate (preparation: see step (i) of Example 5, above; 57 mg) in THF (1.5 mL) and the mixture was stirred under reflux for 5h. Further LiAlH 4 (17 mg) was then added, and the mixture was stirred under reflux for 5h.
  • the title compound may be prepared by the sequence of steps described below:
  • Example 2 above, to give the title compound (40 mg) as a colourless gum; 1H NMR (CDCI3): 6.90-6.86 (m, 2H), 6.78 (dd, IH), 4.70-4.66 (m, 3H), 4.13-4.03 (m, IH), 3.89 (s, 3H), 3.75 (s, 2H), 3.65 (s, 2H), 3.64 (t, 2H), 3.38 (ddd, IH), 3.38 (ddd, IH), 2.77 (t, 2H), 2.33 (s, 3H), 1.84-1,72 (m, IH), 1.45-1.34 (m, IH), 1.28-1.16 (m, 2H), 1.12-0.98 (m, 2H), 0.74 (t, 3H).
  • Lithium triethylborohydride (93 mL) was added to a solution of methyl 4-((2-amino-4- hydroxy-6-methylpyrimidin-5-yl)methyl)-3-methoxybenzoate (5.0 g, 17 mmol) in THF (25mL) over 5 min and the reaction mixture was stirred at r.t. for 3.5 h.
  • Water 60 mL and 2N-HC1 (40 mL) was added to the mixture. The organic solvent was removed by evaporation. 2N-HC1 (16 mL) was added and the mixture was stirred at r.t. for 4 h. The mixture was neutralized by saturated aqueous NaHC0 3 .
  • 2-Mesitylenesulfonyl chloride (7.2 g, 33 mmol) was added to a suspension of diisopropylethylamine (5.5 mL, 33 mmol) and the product from step (i) (6.1 g, 22 mmol) in THF (200 mL) and the mixture was stirred under reflux for 12 h. The resulting mixture was diluted with water and extracted with EtOAc. The combined organic layer was washed with brine, dried over Na 2 S0 4 and concentrated under reduced pressure.
  • Methansulfonyl chloride (1.4 mL, 17 mmol) was added to a mixture of lithium chloride (0.74 g, 17 mmol) and the product from step (ii) (4.0 g, 8.7 mmol) in THF (45 mL). The mixture was stirred at r.t. for 1.5 h, diluted with water and extracted with EtOAc.
  • Trifluoroacetic acid (0.46 mL, 6.0 mmol) was added to the mixture of (S)-3-aminohexan- l-ol (2.1 g, 18 mmol) and the product from step (iv) (3.0 g, 6.0 mmol) in propionitrile (15 mL). The mixture was heated at 120°C for 10 h and cooled to r.t. The solvent was removed by evaporation.
  • Example 10 TV 2 - ⁇ 4- ⁇ (2- Amino-4- ⁇ ⁇ (3S)- l-hvdroxyhexan-3-yll aminol-6-methyl- pyrimidin-5-yl)methyll-3-methoxybenzyl ⁇ glycinamide
  • Example 11 TV 3 - ⁇ 4- if 2- Amino-4- ⁇ ⁇ ( 35)- l-hydroxyhexan-3-yll aminol-6-methyl- pyrimidin-5-yl)methyll-3-methox benzyl ⁇ -
  • the title compound may be prepared by the sequence of steps described below:
  • fert-Butyldimethylsilylchloride (1.95 g) was added to a solution of methyl 4-[(2-amino-4- ⁇ [(35)- 1 -hydroxyhexan-3 -yljamino ⁇ -6-methylpyrimidin-5 -yl)methyl] -3 -methoxybenzoate (preparation: see step (i) of Example 7, above; 4 g) in N,N-dimethylformamide (20 mL) and triethylamine (2.1 mL), and the mixture was stirred at RT for 1.5h.
  • LiBH 4 (2M in THF, 0.77 mL) was added to a solution of methyl 4-[(2- ⁇ [ieri-butyl- (dimethyl)silyl] amino ⁇ -4- ⁇ [(35)- 1 - ⁇ [fert-butyl(dimethyl)silyl]oxy ⁇ hexan-3 -yl] amino ⁇ -6- methylpyrimidin-5-yl)methyl]-3-methoxybenzoate (0.65 g) in THF (5 mL) and CH3OH (0.5 mL), and the mixture was stirred at 60°C for 1.5h. Further LiBH 4 (2M in THF, 0.39 mL) was then added, and the mixture was stirred under reflux for 3h.
  • the title compound may be prepared by the sequence of steps described below:
  • Guanidinium carbonate (8.51 g) was added in one portion to a solution of ethyl 2-(4- cyano-2-methoxybenzyl)-3-oxobutanoate (13 g) in CH 3 OH (200 mL). The resulting mixture was stirred at 65°C for 16h, and then allowed to cool. Water (200 mL) was added and the mixture neutralized by the addition of acetic acid.
  • Example 16 A ( 3S)-3- ⁇ [2- Amino-5-( 2-methoxy-4- ⁇ ⁇ ( 2-methoxyethylH methvDaminol - methyl ⁇ benzyl)-6-methylpyrimidin-4-yllamino ⁇ hexan-l-ol bis-saccharin salt,
  • Example 17A ( 3S)-3- ⁇ ( 2- Amino-5- ⁇ 2-methoxy-4- ⁇ ( 4-methylpiperazin- 1-vDmethyll - benzyl ⁇ -6-methylpyrimidin-4-yl)aminolhexan-l-ol bis saccharin salt
  • Example 18A f3S -3-f ⁇ 2-Amino-5-[2-methoxy-4-fpyrrolidin-l-ylmethyl)benzyll-6- methylpyrimidin-4-yl ⁇ amino)hexan-l-ol bis saccharin salt
  • the title compound may be prepared by the sequence of steps described below:
  • Example 19 A (35V3- ⁇ ⁇ 2- Amino-5-(4- ⁇ r -hvdroxyethyl)(methyl)aminol methyl ⁇ -2- methoxybenzyl)-6-methylpyrimidin-4-yllamino ⁇ hexan-l-ol bis saccharin salt
  • the title compound may be prepared by the steps described below:
  • the title compound may be prepared by the steps described below:
  • the title compound may be prepared by the steps described below:
  • Recombinant human TLR7 was stably expressed in a HEK293 cell line already stably expressing the pNiFty2-SEAP reporter plasmid; integration of the reporter gene was maintained by selection with the antibiotic zeocin.
  • the most common variant sequence of human TLR7 (represented by the EMBL sequence AF240467) was cloned into the mammalian cell expression vector pUNO and transfected into this reporter cell-line.
  • Transfectants with stable expression were selected using the antibiotic blasticidin.
  • expression of secreted alkaline phosphatase (SEAP) is controlled by an NFkB/ELAM-1 composite promoter comprising five NFkB sites combined with the proximal ELAM-1 promoter.
  • TLR signaling leads to the translocation of NFkB and activation of the promoter results in expression of the SEAP gene.
  • TLR7-specific activation was assessed by determining the level of SEAP produced following overnight incubation of the cells at 37°C with the standard compound in the presence of 0.1% (v/v) dimethylsulfoxide (DMSO).
  • DMSO dimethylsulfoxide
  • TLR7 activity for compounds of the present invention was assessed using the human TLR7 assay and the results are shown in Table 1 below wherein the degree of TLR7 activation for each compound is expressed as a pEC 5 o value.
  • HEK293-hTLR8 cells stably transfected with human TLR8 (pUNO expression vector) and pNiFty2-SEAP (Secretory Alkaline Phosphatase) reporter plasmid were maintained in DMEM, FCS 10% (v/v), 2 mM /-glutamine, non-essential amino acids, 10 ⁇ g/mL blasticidin S and 10 ⁇ g/mL zeocin.
  • the human TLR8 sequence used was identical to GenBank sequence AAZ95441.1. Cells were seeded in tissue culture treated clear flat bottom polystyrene 96 well plates at 2> ⁇ 10 4 cells/well.
  • Dose response curves were generated by addition of test compounds and incubated for 20h at 37°C in an atmosphere of 5% C0 2 .
  • the SEAP released was quantified using /?-nitrophenyl phosphate as a substrate and the absorbance at 405 nm was determined by a microplate reader.
  • TLR8/NF-kB/SEAPorterTM HEK 293 Cell Line (Imgenex Corporation) is a stably co- transfected cell line which expresses full-length human TLR8 and the secreted alkaline phosphatase (SEAP) reporter gene under the transcriptional control of an NF- ⁇ response element. TLR8 expression in this cell line has been tested by flow cytometry. Transfectants with stable expression were selected using the antibiotic blasticidin and geneticin. TLR signaling leads to the translocation of NF- ⁇ and activation of the promoter results in expression of the SEAP gene. TLR8-specific activation was assessed by determining the level of SEAP produced following overnight incubation of the cells at 37°C with the standard compound in the presence of 0.1% (v/v) dimethylsulfoxide (DMSO).
  • DMSO dimethylsulfoxide
  • Concentration dependent induction of SEAP production by compounds was expressed as the concentration of compound which produced half of the maximal level of SEAP induction for that compound (EC 50 ).
  • TLR8 activity for compounds of the present invention was assessed using the human TLR8 assay and the results are shown in Table 2 below wherein the degree of TLR8 activation for each compound is expressed as a pEC 50 value.
  • the hERG-expressing Chinese hamster ovary Kl (CHO) cells described by (Persson, Carlsson, Duker, & Jacobson, 2005) are grown to semi-confluence at 37°C in a humidified environment (5% C0 2 ) in F-12 Ham medium containing L-glutamine, 10% foetal calf serum (FCS) and 0.6mg/mL hygromycin (all available from Sigma- Aldrich). Prior to use, the monolayer is washed using a pre-warmed (37°C) 3mL aliquot of Versene 1 :5,000 (Invitrogen).
  • Phosphate-Buffered Saline containing calcium (0.9mM) and magnesium (0.5mM) PBS; Invitrogen
  • PBS Phosphate-Buffered Saline containing calcium (0.9mM) and magnesium (0.5mM)
  • PBS Invitrogen
  • the resulting supernatant is discarded and the pellet gently re-suspended in 3mL of PBS.
  • a 0.5mL aliquot of cell suspension is removed and the number of viable cells (based on trypan blue exclusion) is determined in an automated reader (Cedex; Innovatis) so that the cell re-suspension volume can be adjusted with PBS to give the desired final cell concentration. It is the cell concentration at this point in the assay that is quoted when referring to this parameter.
  • CHO-Kvl .5 cells which are used to adjust the voltage offset on Ion WorksTM HT, are maintained and prepared for use in the same way.
  • a ⁇ -test Ion WorksTM HT from Essen Instrument was used. There is no capability to warm solutions in this device hence it is operated at ⁇ r.t. ( ⁇ 21°C), as follows.
  • the reservoir in the "Buffer” position is loaded with 4 mL of PBS and that in the "Cells” position with the CHO-hERG cell suspension described above.
  • a 96-well plate (V-bottom, Greiner Bio- one) containing the compounds to be tested (at 3 -fold above their final test concentration) is placed in the "Plate 1" position and a PatchPlateTM is clamped into the PatchPlateTM station. Each compound plate is laid-out in 12 columns to enable ten, 8-point
  • concentration-effect curves to be constructed the remaining two columns on the plate are taken up with vehicle (final concentration 0.33% DMSO), to define the assay baseline, and a supra-maximal blocking concentration of cisapride (final concentration 10 ⁇ ) to define the 100% inhibition level.
  • the fluidics-head (F-Head) of IonWorksTM HT then adds 3.5 ⁇ . of PBS to each well of the PatchPlateTM and its underside is perfused with "internal" solution that had the following composition (in mM): K-Gluconate (100 parts), KC1 (40 parts), MgCl 2 (3.2 parts), EGTA (3 parts) and HEPES (5 parts, pH 7.25-7.30 using 10M KOH).
  • the electronics-head After priming and de-bubbling, the electronics-head (E-head) then moves round the PatchPlateTM performing a hole test (i.e. applying a voltage pulse to determine whether the hole in each well is open).
  • the F-head then dispenses 3.5 ⁇ of the cell suspension described above into each well of the PatchPlateTM and the cells are given 200 seconds to reach and seal to the hole in each well.
  • the E-head moves round the PatchPlateTM to determine the seal resistance obtained in each well.
  • the solution on the underside of the PatchPlateTM is changed to "access" solution that has the following composition (in mM): KC1 (140 parts), EGTA (1 part), MgCl 2 (1 part) and HEPES (20 parts, pH 7.25-7.30 using 10M KOH) plus 100 ⁇ g/mL of amphotericin B (Sigma- Aldrich).
  • KC1 140 parts
  • EGTA 1 part
  • MgCl 2 MgCl 2
  • HEPES 20 parts, pH 7.25-7.30 using 10M KOH
  • 100 ⁇ g/mL of amphotericin B Sigma- Aldrich
  • the E-head moves round the PatchPlateTM 48 wells at a time to obtain pre-compound hERG current measurements.
  • the F-head then adds 3.5 ⁇ of solution from each well of the compound plate to 4 wells on the PatchPlateTM (the final DMSO concentration is 0.33% in every well).
  • the pre- and post-compound hERG current is evoked by a single voltage pulse consisting of a 20 second period holding at -70mV, a 160millisecond step to -60mV (to obtain an estimate of leak), a lOOmillisecond step back to -70mV, a 1 second step to + 40mV, a 2 second step to -30mV and finally a 500 millisecond step to -70mV.
  • a single voltage pulse consisting of a 20 second period holding at -70mV, a 160millisecond step to -60mV (to obtain an estimate of leak), a lOOmillisecond step back to -70mV, a 1 second step to + 40mV, a 2 second step to -30mV and finally a 500 millisecond step to -70mV.
  • Currents are leak- subtracted based on the estimate of current evoked during the +10mV step at the start of the voltage pulse protocol. Any voltage offsets in Ion WorksTM
  • a depolarising voltage ramp is applied to CHO-Kvl .5 cells and the voltage noted at which there was an inflection point in the current trace (i.e. the point at which channel activation is seen with a ramp protocol).
  • the voltage at which this occurred has previously been determined using the same voltage command in conventional electrophysiology and found to be -15mV (data not shown); thus an offset potential could be entered into the Ion WorksTM HT software using this value as a reference point.
  • any offset is adjusted by determining the hERG tail current reversal potential in Ion WorksTM HT, comparing it with that found in conventional
  • Pre- and post-scan hERG current magnitude was measured automatically from the leak subtracted traces by the Ion WorksTM HT software by taking a 40ms average of the current during the initial holding period at -70 mV (baseline current) and subtracting this from the peak of the tail current response.
  • the acceptance criteria for the currents evoked in each well are: pre-scan seal resistance >60 ⁇ , pre-scan hERG tail current amplitude >150pA; post-scan seal resistance >60 ⁇ .
  • the degree of inhibition of the hERG current can be assessed by dividing the post-scan hERG current by the respective pre-scan hERG current for each well. References: Persson, F. et al, J Cardiovasc.ElectrophysioL, 16, 329-341 (2005), and Schroeder, K., et al, J Biomol Screen., 8, 50-64, (2003). hERG analysis - Method 2
  • the hERG potassium current is measured in a hERG-stably-expressing Chinese hamster ovary Kl (CHO) cells.
  • the experiments were performed using an automated planar patch-clamp system QPatch HT (Sophion Bioscience A/S).
  • QPatch HT Sophion Bioscience A/S
  • the application of pressure for forming gigaseals and whole-cell patch clamp configuration were established using the QPatch assay software. Patch-clamp experiments were performed in voltage-clamp mode and whole-cell currents were recorded from individual cells. The following stimulation protocol was applied to investigate the effects of compounds on hERG potassium channel.
  • the membrane potential was held at -80 mV and repetitively (every 15 s) depolarized to +20 mV for 5 s after the pulse to -50 mV for 20 ms served to define the baseline, followed by repolarizing step to -50 mV for 5 s to evaluate of the tail current amplitude.
  • Experiments were conducted at room temperature (22 ⁇ 2°C).
  • the test solution includes:
  • Extracellular solution 2mM of CaCl 2 , lmM of MgCl 2 , lOmM of HEPES, 4mM of KC1, 145 mM of NaCl, and lOmM of Glucose;
  • Intracellular solution 5.4mM of CaCl 2 , 1.8mM of MgCl 2 , lOmM of HEPES, 31mM of KOH, lOmM of EGTA, 120mM of KC1, and 4mM of ATP.

Abstract

The invention concerns compounds of Formula (I), and pharmaceutically acceptable salts thereof: where such compounds act as TLR7 agonists and at the same time show advantageous selectivity over TLR8 and hERG, wherein the variable groups R1, R2 and n are as defined herein. The invention also relates to methods for the preparation of such compounds and intermediates useful in the preparation thereof, to pharmaceutical compositions containing such compounds, to the use of such compounds in the preparation of medicaments, and to the use of such compounds in the treatment of conditions mediated by TLR7, such as allergic diseases, autoimmune diseases, viral diseases and, in particular, cancer.

Description

BENZYLAMINE COMPOUNDS AS TOLL -LIKE RECEPTOR 7 AGONISTS
The present invention relates to novel benzylamine compounds and, more particularly, to certain benzylamine compounds that act as TLR7 agonists and at the same time show advantageous selectivity over TLR8 and hERG. This invention also relates to methods for the preparation of such compounds and intermediates useful in the preparation thereof, to pharmaceutical compositions containing such compounds, to the use of such compounds in the preparation of medicaments, and to the use of such compounds in the treatment of conditions mediated by TLR7, such as allergic diseases, autoimmune diseases, viral diseases and, in particular, cancer.
Toll-like receptors (TLRs) are expressed on a variety of immune cells, including macrophages and dendritic cells (DCs). TLRs recognise molecular motifs on pathogens called pathogen-associated molecular patterns (PAMPs). To date, 13 TLRs have been identified in man, these include TLRs 1, 2, 4, 5 and 6, which are confined to the cell surface and TLRs 3, 7, 8 and 9 which are expressed in endosomes. Different TLRs recognise different pathogen-derived ligands, for example TLRs 2 (bacterial lipoproteins), 3 (double-stranded RNA/poly (I:C)), 4 (lipopolysaccharides), 5 (flagellin), 7 (single- stranded RNA) and 9 (CpG-containing DNA). Ligation of TLRs on antigen-presenting cells, such as DCs, leads to production of proinflammatory cytokines, DC maturation and priming of the adaptive immune system. TLR7 and TLR9 are expressed by plasmacytoid dendritic cells (pDCs) and ligand recognition leads to the secretion of interferon-a (INF-a). Preclinical studies investigating the effects of activation of TLRs, using bacterial or viral components, dosed as monotherapy and/or combined with anti-tumor agents, have shown tumour growth inhibition in a variety of murine tumour models.
Several small molecule TLR7 agonists have been described, including the imidazoquinoline, imiquimod, which has been used to treat a number of dermatological conditions e.g. genital warts, molluscum contagiosum and melanoma. In the case of melanoma, topically applied imiquimod (Aldara™, Graceway Pharmaceuticals, Bristol, TN) demonstrated therapeutic responses in cutaneous metastatic melanoma and lentigo maligna and has been approved for the treatment of superficial basal cell carcinoma (BCC). Preclinical and clinical studies indicate that imiquimod is likely to function through the induction of type 1 IFN and IFN-inducible genes, which in turn can have direct effects on tumour cell growth and/or harness components of the adaptive immune system. 852A is another imidazoquinoline, which unlike imiquimod, is suitable for systemic administration. Currently 852A is in phase II clinical trials in a number of cancer indications, including melanoma.
Nevertheless, there remains a need for further TLR7 agonists which are expected to be more effective in the treatment of disease, for example cancer, by reason of their superior potency and/or advantageous physical properties (for example, higher
permeability, and/or lower plasma protein binding) and/or favourable toxicity profiles and/or favourable metabolic profiles in comparison with other known TLR7 agonists, for example 852A.
As demonstrated herein, the benzylamine compounds of the present invention are capable of activating TLR7 in vitro. As a consequence of this activity, the compounds of the present invention are expected to have value in the prevention or treatment of human disease, for example cancer, either as a monotherapy or in combination with other chemotherapeutic agents or radiotherapy regimens.
As a further feature of the invention, compounds of the invention have surprisingly advantageous selectivity for TLR7 over TLR8. TLR7 and TLR8 differ in their cellular expression and as a result stimulation with selective agonists induces different cytokine profiles. TLR8 stimulation (either as a TLR8 selective agonist or a TLR7/8 dual agonist) results in enhanced levels of pro-inflammatory cytokines including TNFa, IL-Ιβ and IL-6 (Gorden et al (2005) J. Immunol. 174, 1259-1268). Conversely, TLR8 stimulation results in lower levels of IFNa. Therefore, a TLR7 selective agonist would favour induction of IFNa, which is important in suppression of Th2 cytokines (Huber et al (2010) J. Immunol. 185; 813-817) that are elevated in allergic disease. In addition, by making compounds selective for TLR7 compared to TLR8 the induction of proinflammatory cytokines would be reduced thus avoiding inflammatory responses in man.
As a further feature of the invention, compounds of the invention also have a surprisingly advantageous hERG profile. Compounds that have significant activity against the hERG ion channel are disfavoured because such activity is implicated in the
development of Torsades de Pointes and cardiac death. According to a first aspect of the present invention, there is therefore provided a compound of Formula (I):
Figure imgf000004_0001
wherein:
n is 1 or 2;
R1 is Ci_4alkyl, methoxyC2-3alkyl, hydroxyC2_4alkyl, -(CH2)yCONH2 wherein y is 1 or 2, or azetidin-3-yl, wherein said azetidin-3-yl is optionally substituted on nitrogen by Ci_3alkyl; and
R2 is hydrogen or Ci_4alkyl;
or R1 and R2 together with the nitrogen to which they are attached form a pyrrolidin-l-yl, lH-imidazol-l-yl or a piperazin-l-yl; wherein said piperazin-l-yl is optionally substituted on the available nitrogen by Ci_3alkyl;
or a pharmaceutically acceptable salt thereof.
For the avoidance of doubt, the "available nitrogen" of a piperazin-l-yl is at the 4-position of the piperazin-l-yl.
It is to be understood that, certain of the compounds of Formula (I) defined above may exist in optically active or racemic forms by virtue of one or more asymmetric carbon atoms, the invention includes in its definition any such optically active or racemic form which possesses the above-mentioned activity. The synthesis of optically active forms may be carried out by standard techniques of organic chemistry well known in the art, for example by synthesis from optically active starting materials or by resolution of a racemic form. The above-mentioned activity may be evaluated using the standard laboratory techniques referred to hereinafter.
It is to be understood that certain compounds of Formula (I) above may exist in unsolvated forms as well as solvated forms, such as, for example, hydrated forms. It is to be understood that the present invention encompasses all such solvated forms that activate TLR7.
It is also to be understood that certain compounds of the Formula (I) may exist in crystalline form and exhibit polymorphism. The present invention encompasses all such forms that activate TLR7.
The term "Ci_4alkyl" is intended to mean a saturated carbon chain of 1 to 4 carbon atoms in length which may be straight-chained or branched. However references to individual alkyl groups such as "propyl" are specific for the straight chain version only and references to individual branched-chain alkyl groups such as tert-butyl are specific for the branched chain version only. For example, "Ci_4alkyl" includes methyl, ethyl, propyl, isopropyl, butyl, n-butyl, isobutyl and tert-butyl. The term "Ci_3alkyl" or "C2_3alkyl" is to be construed accordingly.
In one embodiment there is provided a compound of Formula (I) of Formula (IA):
Figure imgf000005_0001
or a pharmaceutically acceptable salt thereof, wherein the values of R1, R2 and n may take any of the values defined herein for R1, R2 and n respectively.
The variable groups R1, R2 and n may also take the values as indicated below. Such values may be used together with any of the definitions, claims, aspects or embodiments defined herein to provide further embodiments or claims of the invention, and unless the context does not permit, any number of said variable group definitions may be used in any combination with each other to form further embodiments, aspects and claims.
(i) n is 1.
(ii) n is 2.
(iii) R1 is Ci_4alkyl.
(iv) R1 is methoxyC2-3alkyl.
(v) R1 is hydroxyC2-4alkyl.
(vi) R1 is -(CH2)yCONH2 wherein y is 1 or 2. (vii) R1 is azetidin-3-yl, wherein said azetidin-3-yl is optionally substituted on nitrogen by Ci_3alkyl.
(viii) R1 is methyl, 2-hydroxy ethyl, azetidin-3-yl, l-methylazetidin-3-yl, 2- methoxyethyl or -(CH2)yCONH2 wherein y is 1 or 2; and R2 is hydrogen or methyl; or R1 and R2 together with the nitrogen to which they are attached form a piperazin-l-yl, lH-imidazol-l-yl, 4-methylpiperazin-l-yl or pyrrolidin-l-yl.
(ix) R1 is methyl or ethyl.
(x) R1 is methyl.
(xi) R1 is 2-hydroxyethyl.
(xii) R1 is azetidin-3-yl.
(xiii) R1 is l-methylazetidin-3-yl.
(xiv) R1 is -CH2CONH2.
(xv) R1 is -CH2CH2CONH2.
(xvi) R2 is hydrogen.
(xvii) R2 is Ci_4alkyl.
(xviii) R2 is methyl or ethyl.
(xix) R2 is methyl.
(xx) R1 is methyl, ethyl, 2-hydroxyethyl, azetidin-3-yl, l-methylazetidin-3-yl, 2- methoxyethyl or -(CH2)yCONH2 wherein y is 1 or 2; and R2 is hydrogen or methyl.
(xxi) R1 and R2 together with the nitrogen to which they are attached form a piperazin-l-yl, lH-imidazol-l-yl, 4-methylpiperazin-l-yl or pyrrolidin-l-yl.
In one embodiment of the invention there is provided any Example described herein, or a pharmaceutically acceptable salt thereof.
In one embodiment there is provided a compound of Formula (I) selected from:
(25)-2-[(2-amino-5-{4-[(dimethylamino)methyl]-2-methoxybenzyl}-6- methylpyrimidin-4-yl)amino]pentan- 1 -ol;
(2iS)-2-( {2-amino-5-[2-methoxy-4-(piperazin- 1 -ylmethyl)benzyl]-6- methylpyrimidin-4-yl} amino)pentan- 1 -ol;
(25)-2-[(2-amino-5-{2-methoxy-4-[(methylamino)methyl]benzyl}-6- methylpyrimidin-4-yl)amino]pentan- 1 -ol; (25)-2-{[2-amino-5-(4-{[(2-hydroxyethyl)amino]methyl}-2-methoxy-benzyl)-6- methylpyrimidin-4-yl]amino}pentan- 1 -ol;
(25)-2-[(2-amino-5 - {4- [(azetidin-3 -ylamino)methyl] -2-methoxybenzyl} -6- methylpyrimidin-4-yl)amino]pentan- 1 -ol;
(2S)-2- { [2-amino-5-(2-methoxy-4- { [( 1 -methylazetidin-3-yl)amino]methyl} - benzyl)-6-methylpyrimidin-4-yl] amino } pentan- 1 -ol;
(35)-3-[(2-amino-5-{2-methoxy-4-[(methylamino)methyl]benzyl}-6- methylpyrimidin-4-yl)amino]hexan- 1 -ol;
(35)-3-( {2-amino-5-[2-methoxy-4-(piperazin- 1 -ylmethyl)benzyl]-6- methylpyrimidin-4-yl} amino)hexan- 1 -ol;
(35)-3-{[2-amino-5-(4-{[(2-hydroxyethyl)amino]methyl}-2-methoxy-benzyl)-6- methylpyrimidin-4-yl]amino } hexan- 1 -ol;
N2- {4-[(2-amino-4- {[(35)- 1 -hydroxyhexan-3-yl]amino} -6-methyl-pyrimidin-5- yl)methyl]-3-methoxybenzyl}glycinamide;
N3- {4-[(2-amino-4- {[(35)- 1 -hydroxyhexan-3-yl]amino} -6-methyl-pyrimidin-5- yl)methyl]-3-methoxybenzyl}-P-alaninamide;
(35)-3-[(2-amino-5-{4-[(azetidin-3-ylamino)methyl]-2-methoxybenzyl}-6- methylpyrimidin-4-yl)amino]hexan- 1 -ol;
(35)-3-{[2-amino-5-(2-methoxy-4-{[(l-methylazetidin-3-yl)amino]- methyl} benzyl)-6-methylpyrimidin-4-yl] amino } hexan- 1 -ol;
N2- {4-[(2-amino-4- {[(35)- 1 -hydroxyhexan-3-yl]amino} -6-methyl-pyrimidin-5- yl)methyl]-3-methoxybenzyl}-N2-methylglycinamide;
(35)-3-( {2-amino-5-[4-(lH-imidazol- 1 -ylmethyl)-2-methoxybenzyl]-6- methylpyrimidin-4-yl} amino)hexan- 1 -ol;
(35)-3-{[2-amino-5-(2-methoxy-4-{[(2-methoxyethyl)(methyl)amino]- methyl} benzyl)-6-methylpyrimidin-4-yl] amino } hexan- 1 -ol;
(35)-3-[(2-amino-5-{2-methoxy-4-[(4-methylpiperazin-l-yl)methyl]-benzyl}-6- methylpyrimidin-4-yl)amino]hexan- 1 -ol;
(35)-3-( {2-amino-5-[2-methoxy-4-(pyrrolidin- 1 -ylmethyl)benzyl]-6- methylpyrimidin-4-yl} amino)hexan- 1 -ol;
(35)-3 - { [2-amino-5 -(4- { [(2-hydroxyethyl)(methyl)amino]methyl} -2- methoxybenzyl)-6-methy lpyrimidin-4-yl] amino } hexan- 1 -ol; (2S)-2-( {2-amino-5-[2-methoxy-4-(pyrrolidin- 1 -ylmethyl)benzyl]-6- methylpyrimidin-4-yl} amino)pentan- 1 -ol;
(25)-2-[(2-amino-5-{2-methoxy-4-[(4-methylpiperazin-l-yl)methyl]-benzyl}-6- methylpyrimidin-4-yl)amino]pentan- 1 -ol;
(35)-3-[(2-amino-5-{4-[(diethylamino)methyl]-2-methoxybenzyl}-6- methylpyrimidin-4-yl)amino]hexan- 1 -ol;
(3iS)-3 - { [2-amino-5 -(4- { [ethyl(methyl)amino]methyl} -2-methoxybenzyl)-6- methylpyrimidin-4-yl]amino } hexan- 1 -ol; and
(35)-3-[(2-amino-5-{4-[(dimethylamino)methyl]-2-methoxybenzyl}-6- methylpyrimidin-4-yl)amino]hexan- 1 -ol;
or a pharmaceutically acceptable salt thereof.
Compounds of Formula (I) may be prepared by the sequence shown in Scheme 1 :
Figure imgf000008_0001
(VI) (VII) (VIII)
Scheme 1
The R3 group of the compound of Formula (II) may be a substituted or
unsubstituted hydrocarbyl group. Conveniently R3 may be a hydrocarbyl group selected from benzyl and Ci_4alkyl, for example methyl or ethyl. The LG1 group of the compound of Formula (III) may be any convenient leaving group which are well-known to the skilled person, for example a substituted or unsubstituted hydrocarbylsulfonyloxy leaving group, for example a tosylate or mesylate group, or a halo leaving group such as iodo or more conveniently, a bromo or chloro leaving group.
Step (i) - Compounds of Formula (IV) may be prepared by a standard alkylation reaction using compounds of Formula (II) and Formula (III): For example, reaction of a compound of Formula (III) with a base, such as NaH, in a suitable solvent, such as THF or DMF, at a suitable temperature, for example 0-20°C, followed by addition of a compound of Formula (II). The reaction may be heated, for example at 50-100°C, optionally in the presence of a catalytic amount of an iodide salt, such as KI.
Step (ii) - Compounds of Formula (V) may be prepared by reacting a compound of Formula (IV) with guanidine or guanidine carbonate in a suitable solvent, such as methanol or ethanol, at elevated temperature, for example 50-150°C. The compound of Formula (V) may be isolated as a salt.
In one aspect of the invention there is provided a compound of Formula (V) as defined in Scheme 1 , or a salt thereof.
Step (iii) - Compounds of Formula (VI) may be prepared by reacting a compound of Formula (V) with POCI3, at elevated temperature, for example 50-110°C. The compound of Formula (V) may be isolated as a salt.
In one aspect of the invention there is provided a compound of Formula (VI) as defined in Scheme 1, or a salt thereof.
Step (iv) - Compounds of Formula (VII) may be prepared by reacting a compound of Formula (VI) with an excess of the appropriate amino alcohol (where the amino alcohol may optionally have its alcohol group protected) in a suitable solvent, such as propionitrile, butanol or 1,4-dioxane, in the presence of trifluoroacetic acid at elevated temperature (for example 50-200°C) using conventional or microwave heating. The R4 group in the compound of Formula (VII) and the compound of Formula (VIII) is independently selected from hydrogen, a silicon-based protecting group or a benzyl-based protecting group. Conveniently, R4 is selected from hydrogen, trimethylsilyl, tert-butyldimethylsilyl and benzyl. In one embodiment R4 is hydrogen. In one embodiment R4 is a silicon-based protecting group or a benzyl-based protecting group. In one embodiment R4 is a silicon- based protecting group. The silicon-based protecting group may be a tri(Ci_4alkyl)silyl group, for example a trimethylsilyl group or a tert-butyldimethylsilyl group. In one embodiment R4 is tert-butyldimethylsilyl. In further embodiments the compound of Formula (VII) and/or the compound of Formula (VIII) has an (5)-stereocentre at the asymmetric carbon atom in the amino alcohol part of the molecule. The optionally used protecting group incorporated into the compound of Formula (VII) and/or Formula (VIII) may be removed at any convenient point in the synthesis using standard deprotection conditions that are well known to the skilled person. The compounds of Formula (VII) and the compound of Formula (VIII) may be isolated as a salt.
In one aspect of the invention there is provided a compound of Formula (VII), as defined herein, or a salt thereof.
In one aspect there is provided a compound of Formula (VIII), as defined herein, or a salt thereof.
Step (v) - Compounds of Formula (VIII) may be prepared by reacting a compound of Formula (VII) with Raney nickel in a suitable solvent mixture such as pyridine, acetic acid and water, at a suitable temperature, for example 20-50°C.
Step (vi) - Compounds of Formula (I) may be prepared by reaction of a compound of (VIII) with the appropriate amine (FTNR R2) under reductive amination conditions, which are well known to the skilled person. For example, the reductive amination may be carried out using a suitable reducing agent such as sodium triacetoxyborohydride in a suitable solvent such as dichloromethane and acetic acid in the presence of activated molecular sieves, or by using NaBH4 in a suitable solvent such as methanol.
Compounds of Formula (VIII) may also be prepared as shown in Scheme 2.
Figure imgf000011_0001
Figure imgf000011_0002
(XV) (VIII)
Scheme 2
The R5 group of the compound of Formula (IX) may be a substituted or unsubstituted hydrocarbyl group. Conveniently R5 may be a hydrocarbyl group selected from benzyl and Ci_4alkyl, for example methyl or ethyl. The LG2 group of the compound of Formula (X) may be any convenient leaving group which are well-known to the skilled person, for example a substituted or unsubstituted hydrocarbylsulfonyloxy leaving group, for example a tosylate or mesylate group, or a halo leaving group such as iodo or more conveniently, a bromo or chloro leaving group. The R6 group of the compound of Formula (X) may be a substituted or unsubstituted hydrocarbyl group. Conveniently R6 may be a hydrocarbyl group selected from benzyl and Ci_4alkyl, for example methyl or ethyl. Step (i) - Compounds of Formula (XI) may be prepared by a standard alkylation reaction using compounds of Formula (IX) and Formula (X): For example, reaction of a compound of Formula (IX) with a base, such as NaH, in a suitable solvent, such as THF or DMF, at a suitable temperature, for example 0-20°C, followed by addition of a compound of Formula (X). The reaction may be heated, for example at 50-100°C, optionally in the presence of a catalytic amount of an iodide salt, such as KI.
Step (ii) - Compounds of Formula (XII) may be prepared by reacting a compound of Formula (IV) with guanidine or guanidine carbonate in a suitable solvent, such as methanol or ethanol, at elevated temperature, for example 50-150°C. The compound of Formula (XII) may be isolated as a salt.
Therefore, in one aspect of the invention there is provided a compound of Formula
(XII) , as defined herein, or a salt thereof.
Step (iii) - The R7 group in the compound of Formula (XIII) may be a substituted or unsubstituted hydrocarbyl group, for example a Ci_4alkyl group or a phenyl group optionally substituted by 1 , 2 or 3 Ci_4alkyl groups, for example R7 may conveniently be methyl, ethyl, 2,4,6-trimethylphenyl or 2,4,6-triisopropylphenyl. Compounds of Formula
(XIII) may be prepared by reacting a compound of Formula (XII) with a hydrocarbyl sulfonyl chloride: R7S02C1, for example trimethylbenzene sulfonyl chloride or
triisopropylbenzenesulfonyl chloride. The reaction may be carried out in a suitable solvent, such THF in the presence of a suitable base, such as, triethylamine or tetramethyl- propylenediamine, at a suitable temperature, for example 0-50°C. The compound of Formula (XIII) may be isolated as a salt.
Therefore, in one aspect of the invention there is provided a compound of Formula (XIII), as defined herein, or a salt thereof.
Step (iv) - Compounds of Formula (XIV) may be prepared by reacting a compound of Formula (XIII) with an excess of the appropriate amino alcohol (where the amino alcohol may optionally have its alcohol group protected) in a suitable solvent, such as propionitrile, butanol or 1 ,4-dioxane, in the presence of trifluoroacetic acid at elevated temperature (for example 50-200°C) using conventional or microwave heating. The R4 group in the compound of Formula (XIV) and the compound of Formula (XV) is independently selected from hydrogen, a silicon-based protecting group or a benzyl-based protecting group. Conveniently, R4 is selected from hydrogen, trimethylsilyl, tert- butyldimethylsilyl and benzyl. In one embodiment R4 is hydrogen. In one embodiment R4 is a silicon-based protecting group or a benzyl-based protecting group. In one embodiment R4 is a silicon-based protecting group. The silicon-based protecting group may be a tri(Ci_ 4alkyl)silyl group, for example a trimethylsilyl group or a tert-butyldimethylsilyl group. In one embodiment R4 is tert-butyldimethylsilyl. In a further embodiment the compound of Formula (XIV) has an (5)-stereocentre at the asymmetric carbon atom in the amino alcohol part of the molecule. The optionally used protecting group incorporated into the compound of Formula (XIV) and/or Formula (XV) may be removed at any convenient point in the synthesis using standard deprotection conditions that are well known to the skilled person. The compound of Formula (XIV) may be isolated as a salt.
In one aspect of the invention there is provided a compound of Formula (XIV) as defiend herein, or a salt thereof.
Step (v) - Compounds of Formula (XV) may be prepared by reacting a compound of Formula (XIV) with any standard reducing agent known to be capable of reducing an ester to the corresponding alcohol. For example, LiBH4 may be used, in a suitable solvent mixture such as THF and methanol at a suitable temperature, for example 20-60°C. The compound of Formula (XV) may be isolated as a salt.
In one aspect of the invention there is provided a compound of Formula (XV) as defiend herein, or a salt thereof.
Step (vi) - Compounds of Formula (VIII) may be prepared by reacting a compound of Formula (XV) in the presence of a suitable oxidising agent such as Mn02 in a suitable solvent mixture such as THF and CH2C12 at a suitable temperature, for example 20-60°C.
Step (vii) - Compounds of Formula (I) may be prepared by reaction of a compound of (VIII) with the appropriate amine (FTNR R2) under reductive amination conditions, which are well known to the skilled person. For example, the reductive amination may be carried out using a suitable reducing agent such as sodium triacetoxyborohydride in a suitable solvent such as dichloromethane and acetic acid in the presence of activated molecular sieves, or by using NaBH4 in a suitable solvent such as methanol.
Alternatively compounds of Formula (XIV) may be converted into a compound of Formula (I) as shown in Scheme 3 :
Figure imgf000014_0001
Figure imgf000014_0002
(XV-A) (XVI)
Scheme 3
The compound of Formula (XIV- A) is a compound of Formula (XIV) as hereinbefore defined, wherein R4 is hydrogen. The compounds of Formula (XIV-B) and (XV-A) are compounds of Formula (XIV) and (XV) respectively, as hereinbefore defined, wherein R4 is a silicon protecting group, for example a tri(Ci_4alkyl)silyl protecting group (where each of the three Ci_4alkyl groups may be the same or different).
In one embodiment there is provided a compound of Formula (XIV-B) as defined hereinbefore wherein R4 is a tert-butyldimethylsilyl (TBDMS) protecting group, or a salt thereof.
In one embodiment there is provided a compound of Formula (XV-A) as defined hereinbefore wherein R4 is a tert-butyldimethylsilyl (TBDMS) protecting group, or a salt thereof.
Step (i) Compounds of Formula (XIV-B) may be prepared reaction of compounds of Formula (XIV- A) by reaction with tri(Ci_4alkyl)silyl chloride, for example tert- butyldimethylsilyl chloride, in the presence of a base, for example an organic nitrogen base, for example an amine base such as triethylamine in a suitable solvent such as DMF at a suitable temperature, for example 20°C.
Step (ii) Compounds of Formula (XV- A) may be prepared from compounds of Formula (XIV-B) by reacting with a reducing agent that is known to the skilled person for its capability for reducing esters to alcohols, for example LiBH4, in a suitable solvent mixture such as THF and methanol, at a suitable temperature, for example 20-60°C
Step (iii) The LG3 group in the compound of Formula (XVI) is a leaving group where suitable leaving groups are well known to the skilled person. For example LG3 may be chloro or bromo.
In one embodiment there is provided a compound of Formula (XVI) wherein LG3 is chloro, or a salt thereof.
Compounds of Formula (XVI) may be prepared by reacting a compound of Formula (XV- A) with a hydrocarbyl sulfonyl bromide or chloride in the presence of LiBr or LiCl, for example CH3SO2CI with LiCl, in a suitable solvent such as THF at a suitable temperature, for example 20°C, followed by treatment with an acid such as HC1 in dioxane, a suitable solvent such as methanol at a suitable temperature, for example 20°C.
Compounds of Formula (XIV) may be converted into a compound of Formula (I) by an alkylation reaction using the appropriate amine (FfNR^R2) using alkylation conditions that are well known to the skilled person. For example, the alkylation may be achieved using a suitable base such as Na2C03, in a suitable solvent, such as acetonitrile. The compound of Formula (XVI) may be isolated as a salt.
Therefore in a further aspect of the invention there is provided a compound of Formula (XVI), as defined herein, or a salt thereof. In one embodiment LG3 is chloro. In one embodiment the compound of Formula (XVI) has an (5)-stereocentre at the asymmetric carbon atom in the amino alcohol part of the molecule.
Alternatively, a compound of the Formula (I) may be prepared by the sequence shown in Scheme 4:
Figure imgf000016_0001
Figure imgf000016_0002
(XIX) (XX)
Figure imgf000016_0003
(I)
Scheme 4
1 2 6
wherein R , R and n are the same as defined in Formula (I), R is the same as defined in Formula (X), and LG4 and LG5 are each independently a suitable leaving group wherein LG4 in a compound of Formula (XIX) is not removed in the reaction conditions used in Step (iii) and Step (iv). LG4 group may be a hydrocarbyl sulphonyl group: -OS02R8 wherein R8 may be a optionally substituted phenyl group such as /?-methylphenyl, 2,4,6- trimethylphenyl or 2,4,6-triisopropylphenyl. LG5 group may be a halogen such as chloro, bromo, iodo or alternatively may be a hydrocarbyl sulphonyl group: -OS02R9 wherein R9 may be a Ci_4alkyl such as methyl. Step (i) - A compound of Formula (XVII) may be prepared by reacting a compound of Formula (XII) with any standard reducing agent known to be capable of reducing an ester to the corresponding alcohol. For example, L1AIH4 or LiBHEt3 may be used, in a suitable solvent mixture such as THF and methanol at a suitable temperature, for example 20-60°C.
Step (ii) - A compound of Formula (XVIII) may be prepared by reacting a compound of Formula (XVII) with a hydrocarbyl sulphonyl chloride: R8S02C1, for example 2, 4, 6-trimethylbenzene sulphonyl chloride or 2, 4, 6-triisopropylbenzenesulfonyl chloride. The reaction may be carried out in a suitable solvent, such as THF in the presence of a suitable base, such as, triethylamine or diisopropylethylamine, at a suitable temperature, for example 0-100°C.
Step (iii) - A compound of Formula (XIX) may be prepared by reacting a compound of Formula (XVIII) with a hydrocarbyl sulphonyl chloride: R9S02C1, for example methane sulphonyl chloride when R9 is methyl. The reaction may be carried out in a suitable solvent, such THF in the presence of a suitable base, such as, triethylamine or diisopropylethylamine, and optionally lithium chloride or lithium bromide when LG5 is halogen, at a suitable temperature, for example 0-100°C, preferably 0-30°C.
Step (iv) - A compound of Formula (XIX) may be successively converted to a compound of Formula (XX) by an alkylation reaction using the appropriate amine
(HNRXR2) using alkylation conditions that are well known to the skilled person. For example, the alkylation may be achieved using a suitable base such as Na2C03, K2C03, or diisopropyethylamine in a suitable solvent, such as acetonitrile or THF. The compound of Formula (XX) may be isolated as a salt.
Step (v) - A compound of Formula (I) may be prepared by reacting a compound of Formula (XX) with an excess of the appropriate amino alcohol (where the amino alcohol may optionally have its alcohol group protected) in a suitable solvent, such as propionitrile, butanol or 1,4-dioxane, in the presence of trifluoroacetic acid at elevated temperature (for example 50-200°C) using conventional or microwave heating. The compounds described herein in the Schemes and associated text, and later in the detailed experimental section, are useful intermediates for the preparation of the compounds of Formula (I) and may be isolated as a free base or as a salt. Therefore, in further aspects and embodiments of the invention there is provided an intermediate described herein, or a salt thereof, wherein any of the variable groups described for said intermediate may take any of the values described herein in connection with that group.
A suitable pharmaceutically-acceptable salt of a compound of the Formula (I) is, for example, an acid-addition salt of a compound of the Formula (I), for example an acid- addition salt with an inorganic or organic acid such as hydrochloric, hydrobromic, sulphuric, trifluoroacetic, citric or maleic acid.
The compounds of the invention may be administered in the form of a pro-drug, i.e. a compound that is broken down in the human or animal body to release a compound of the invention. A pro-drug may be used to alter the physical properties and/or the pharmacokinetic properties of a compound of the invention. A pro-drug can be formed when the compound of the invention contains a suitable group or substituent to which a property-modifying group can be attached. Examples of pro-drugs include in-vivo cleavable amide derivatives that may be formed at an amino group in a compound of the Formula (I).
Accordingly, the present invention includes those compounds of the Formula (I) as defined hereinbefore when made available by organic synthesis and when made available within the human or animal body by way of cleavage of a pro-drug thereof. Accordingly, the present invention includes those compounds of the Formula (I) that are produced by organic synthetic means and also such compounds that are produced in the human or animal body by way of metabolism of a precursor compound, that is a compound of the Formula (I) may be a synthetically-produced compound or a metabolically-produced compound.
A suitable pharmaceutically-acceptable pro-drug of a compound of the Formula (I) is one that is based on reasonable medical judgement as being suitable for administration to the human or animal body without undesirable pharmacological activities and without undue toxicity.
Various forms of pro-drug have been described, for example in the following documents:- a) Methods in Enzymology, Vol. 42, p. 309-396, edited by K. Widder, et al.
(Academic Press, 1985);
b) Design of Pro-drugs, edited by H. Bundgaard, (Elsevier, 1985);
c) A Textbook of Drug Design and Development, edited by Krogsgaard-Larsen and H. Bundgaard, Chapter 5 "Design and Application of Pro-drugs", by H. Bundgaard p. 113- 191 (1991);
d) H. Bundgaard, Advanced Drug Delivery Reviews, 8, 1-38 (1992);
e) H. Bundgaard, et al., Journal of Pharmaceutical Sciences, 77, 285 (1988);
f) N. akeva, et al. Chem. Pharm. Bull, 32, 692 (1984);
g) T. Higuchi and V. Stella, "Pro-Drugs as Novel Delivery Systems", A.C.S.
Symposium Series, Volume 14; and
h) E. Roche (editor), "Bioreversible Carriers in Drug Design", Pergamon Press, 1987.
A suitable pharmaceutically-acceptable pro-drug of a compound of the Formula (I) that possesses an amino group is, for example, an in-vivo cleavable amide derivative thereof. Suitable pharmaceutically-acceptable amides from an amino group include, for example an amide formed with Ci_ioalkanoyl groups such as an acetyl, benzoyl, phenylacetyl and substituted benzoyl and phenylacetyl groups. Examples of ring substituents on the phenylacetyl and benzoyl groups include aminomethyl,
N-alkylaminomethyl, N,N-dialkylaminomethyl, morpholinomethyl, piperazin-l-ylmethyl and 4-(Ci_4alkyl)piperazin- 1 -ylmethyl.
The in-vivo effects of a compound of the Formula (I) may be exerted in part by one or more metabolites that are formed within the human or animal body after administration of a compound of the Formula (I). As stated hereinbefore, the in-vivo effects of a compound of the Formula (I) may also be exerted by way of metabolism of a precursor compound (a pro-drug).
According to a further aspect of the invention there is provided a pharmaceutical composition which comprises a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore in association with a pharmaceutically- acceptable diluent or carrier. The pharmaceutical composition may be used in the treatment of cancer. The composition may be in a form suitable for oral administration, for example as a tablet or capsule; for parenteral injection (including intravenous, subcutaneous, intramuscular, intravascular or infusion) as a sterile solution, suspension or emulsion; for topical administration as an ointment or cream; or for rectal administration as a
suppository.
The compound of Formula (I), or a pharmaceutically acceptable salt thereof, could also be administered as an air spray for inhalation. The air spray (e.g., spray, aerosol, dry powder preparation, etc.) could be optionally formulated as an aqueous solution or suspension, or as an aerosol delivered from a pressurized pack such as a pressurised metered dose inhaler by using, for example, a liquefied propellant. A dry powder preparation may also be used. An aerosol appropriate for inhalation may be either a suspension or solution, and would typically contain the compound of Formula (I), or a pharmaceutically acceptable salt thereof, and any appropriate propellants such as a fluorocarbon or hydrogen-containing chlorofluorocarbon or a mixture thereof. Specifically, it may contain hydro fluoroalkane, particularly 1,1,1,2-tetrafluoroethane, heptafluoroalkane (HFA) such as 1,1,1,2,3,3,3-heptafluoro-n-propane, or a mixture thereof. An aerosol may optionally contain an additional preparation excipient well-known to those skilled in the art such as surfactant (e.g., oleic acid or lecithin) and cosolvent (e.g., ethanol), etc.
Specifically, an aerosol preparation could be delivered using the inhaler known as
"Turbuhaler™".
For oral administration the compound of the invention may be admixed with an adjuvant or a carrier, for example, lactose, saccharose, sorbitol, mannitol; a starch, for example, potato starch, corn starch or amylopectin; a cellulose derivative; a binder, for example, gelatine or polyvinylpyrrolidone; and/or a lubricant, for example, magnesium stearate, calcium stearate, polyethylene glycol, a wax, paraffin, and the like, and then compressed into tablets. If coated tablets are required, the cores, prepared as described above, may be coated with a concentrated sugar solution which may contain, for example, gum arabic, gelatine, talcum and titanium dioxide. Alternatively, the tablet may be coated with a suitable polymer dissolved in a readily volatile organic solvent.
For the preparation of soft gelatine capsules, the compound of the invention may be admixed with, for example, a vegetable oil or polyethylene glycol. Hard gelatine capsules may contain granules of the compound using either the above-mentioned excipients for tablets. Also liquid or semisolid formulations of the compound of the invention may be filled into hard gelatine capsules. Liquid preparations for oral application may be in the form of syrups or
suspensions, for example, solutions containing the compound of the invention, the balance being sugar and a mixture of ethanol, water, glycerol and propylene glycol. Optionally such liquid preparations may contain colouring agents, flavouring agents, saccharine and/or carboxymethylcellulose as a thickening agent or other excipients known to those skilled in art.
The compound of Formula (I) will normally be administered to a warm-blooded animal at a unit dose within the range 5-5000 mg/m2 body area of the animal, i.e.
approximately 0.1-100 mg/kg, and this normally provides a therapeutically-effective dose. A unit dose form such as a tablet or capsule will usually contain, for example 1-250 mg of active ingredient. Preferably a daily dose in the range of 1-50 mg/kg is employed. However the daily dose will necessarily be varied depending upon the host treated, the particular route of administration, and the severity of the illness being treated. Accordingly the optimum dosage may be determined by the practitioner who is treating any particular patient.
For further information on Routes of Administration and Dosage Regimes the reader is referred to Chapter 25.3 in Volume 5 of Comprehensive Medicinal Chemistry (Corwin Hansch; Chairman of Editorial Board), Pergamon Press 1990.
In the context of the present specification, the term "therapy" also includes
"prophylaxis" unless there are specific indications to the contrary. The terms "therapeutic" and "therapeutically" should be construed accordingly.
As used herein, the term "treatment" is intended to have its normal everyday meaning of dealing with a disease in order to entirely or partially relieve one, some or all of its symptoms, or to correct or compensate for the underlying pathology.
As used herein, the term "prophylaxis" is intended to have its normal everyday meaning and includes primary prophylaxis to prevent the development of the disease and secondary prophylaxis whereby the disease has already developed and the patient is temporarily or permanently protected against exacerbation or worsening of the disease or the development of new symptoms associated with the disease.
The compounds defined in the present invention are effective activators of TLR7 in vitro. Accordingly, the compounds of the present invention are expected to be potentially useful agents in the treatment of diseases or medical conditions mediated alone or in part by TLR7. For example, the following diseases and conditions listed in paragraphs 1 to 8 below may be treatable with compounds of the present invention.
1. respiratory tract: obstructive diseases of the airways including: asthma, including bronchial, allergic, intrinsic, extrinsic, exercise-induced, drug-induced (including aspirin and NSAID-induced) and dust-induced asthma, both intermittent and persistent and of all severities, and other causes of airway hyper-responsiveness; chronic obstructive pulmonary disease (COPD); bronchitis, including infectious and eosinophilic bronchitis; emphysema; bronchiectasis; cystic fibrosis; sarcoidosis; farmer's lung and related diseases;
hypersensitivity pneumonitis; lung fibrosis, including cryptogenic fibrosing alveolitis, idiopathic interstitial pneumonias, fibrosis complicating anti-neoplastic therapy and chronic infection, including tuberculosis and aspergillosis and other fungal infections; complications of lung transplantation; vasculitic and thrombotic disorders of the lung vasculature, and pulmonary hypertension; antitussive activity including treatment of chronic cough associated with inflammatory and secretory conditions of the airways, and iatrogenic cough; acute and chronic rhinitis including rhinitis medicamentosa, and vasomotor rhinitis; perennial and seasonal allergic rhinitis including rhinitis nervosa (hay fever); nasal polyposis; acute viral infection including the common cold, and infection due to respiratory syncytial virus, influenza, coronavirus (including SARS) and adenovirus;
2. skin: psoriasis, atopic dermatitis, contact dermatitis or other eczematous
dermatoses, and delayed-type hypersensitivity reactions; phyto- and photodermatitis;
seborrhoeic dermatitis, dermatitis herpetiformis, lichen planus, lichen sclerosus et atrophica, pyoderma gangrenosum, skin sarcoid, discoid lupus erythematosus, pemphigus, pemphigoid, epidermolysis bullosa, urticaria, angioedema, vasculitides, toxic erythemas, cutaneous eosinophilias, alopecia areata, male-pattern baldness, Sweet's syndrome, Weber- Christian syndrome, erythema multiforme; cellulitis, both infective and non-infective; panniculitis; cutaneous lymphomas, non-melanoma skin cancer and other dysplastic lesions; drug-induced disorders including fixed drug eruptions;
3. eyes: blepharitis; conjunctivitis, including perennial and vernal allergic
conjunctivitis; iritis; anterior and posterior uveitis; choroiditis; autoimmune, degenerative or inflammatory disorders affecting the retina; ophthalmitis including sympathetic ophthalmitis; sarcoidosis; infections including viral , fungal, and bacterial;
4. genitourinary: nephritis including interstitial and glomerulonephritis; nephrotic syndrome; cystitis including acute and chronic (interstitial) cystitis and Hunner's ulcer; acute and chronic urethritis, prostatitis, epididymitis, oophoritis and salpingitis; vulvovaginitis; Peyronie's disease; erectile dysfunction (both male and female);
5. allograft rejection: acute and chronic following, for example, transplantation of kidney, heart, liver, lung, bone marrow, skin or cornea or following blood transfusion; or chronic graft versus host disease;
6. other auto-immune and allergic disorders including rheumatoid arthritis, irritable bowel syndrome, systemic lupus erythematosus, multiple sclerosis, Hashimoto's thyroiditis, Graves' disease, Addison's disease, diabetes mellitus, idiopathic
thrombocytopaenic purpura, eosinophilic fasciitis, hyper-IgE syndrome, antiphospholipid syndrome and Sazary syndrome;
7. oncology: treatment of common cancers including bladder, head and neck, prostate, breast, lung, ovarian, pancreatic, bowel and colon, stomach, skin and brain tumors and malignancies affecting the bone marrow (including the leukaemias) and
lymphoproliferative systems, such as Hodgkin's and non-Hodgkin's lymphoma; including the prevention and treatment of metastatic disease and tumour recurrences, and
paraneoplastic syndromes; and,
8. infectious diseases: virus diseases such as genital warts, common warts, plantar warts, hepatitis B, hepatitis C, herpes simplex virus, molluscum contagiosum, variola, human immunodeficiency virus (HIV), human papilloma virus (HPV), cytomegalovirus (CMV), varicella zoster virus (VZV), rhinovirus, adenovirus, coronavirus, influenza, parainfluenza; bacterial diseases such as tuberculosis and mycobacterium avium, leprosy; other infectious diseases, such as fungal diseases, chlamydia, Candida, aspergillus, cryptococcal meningitis, Pneumocystis carnii, cryptosporidiosis, histoplasmosis, toxoplasmosis, trypanosome infection and leishmaniasis.
It is envisaged that for the methods of treatment mentioned herein, the compound of Formula (I) will be administered to a mammal, more particularly a human being.
Similarly, for the uses of a compound of Formula (I) for the treatment of diseases or medical conditions mentioned herein, it is envisaged that the compound of Formula (I) will be administered to a mammal, more particularly a human being.
According to a another aspect of the invention, there is therefore provided a compound of Formula (I) as defined hereinbefore, or a pharmaceutically acceptable salt thereof, for use as a medicament.
According to a further aspect of the invention, there is provided a compound of Formula (I) as defined hereinbefore, or a pharmaceutically acceptable salt thereof for use in the treatment of a disease mediated through TLR7. In one embodiment of the invention, said disease mediated through TLR7 is cancer. In a further embodiment of the invention, said cancer is selected from bladder cancer, head and neck cancer, prostate cancer, breast cancer, lung cancer, uterus cancer, pancreatic cancer, liver cancer, renal cancer, ovarian cancer, colon cancer, stomach cancer, skin cancer, cerebral tumor, malignant myeloma and lymphoproliferative tumors. In one embodiment of the invention, said disease mediated through TLR7 is asthma, COPD, allergic rhinitis, allergic conjunctivitis, atopic dermatitis, hepatitis B, hepatitis C, HIV, HPV, bacterial infections or dermatosis.
According to a further aspect of the invention, there is provided the use of a compound of Formula (I) as defined hereinbefore, or a pharmaceutically acceptable salt thereof for the preparation of a medicament for the treatment of a disease mediated through TLR7. In one embodiment of the invention, said disease mediated through TLR7 is cancer. In a further embodiment of the invention, said cancer is selected from bladder cancer, head and neck cancer, prostate cancer, breast cancer, lung cancer, uterus cancer, pancreatic cancer, liver cancer, renal cancer, ovarian cancer, colon cancer, stomach cancer, skin cancer, cerebral tumor, malignant myeloma and lymphoproliferative tumors. In one embodiment of the invention, said disease mediated through TLR7 is asthma, COPD, allergic rhinitis, allergic conjunctivitis, atopic dermatitis, hepatitis B, hepatitis C, HIV, HPV, bacterial infections or dermatosis.
According to a further aspect of the invention, there is provided the use of a compound of Formula (I) as defined hereinbefore, or a pharmaceutically acceptable salt thereof, for the preparation of a medicament for the treatment of cancer. In one
embodiment of the invention, said cancer is selected from bladder cancer, head and neck cancer, prostate cancer, breast cancer, lung cancer, uterus cancer, pancreatic cancer, liver cancer, renal cancer, ovarian cancer, colon cancer, stomach cancer, skin cancer, cerebral tumor, malignant myeloma and lymphoproliferative tumors.
According to a further aspect of the invention, there is provided the use of a compound of Formula (I) as defined hereinbefore, or a pharmaceutically acceptable salt thereof, for the preparation of a medicament for the treatment of asthma, COPD, allergic rhinitis, allergic conjunctivitis, atopic dermatitis, hepatitis B, hepatitis C, HIV, HPV, bacterial infections or dermatosis.
In one aspect of the invention there is provided a compound of Formula (I) or a pharmaceutically acceptable salt thereof for use in the treatment of cancer.
According to a further aspect of the invention, there is provided a method of using compound of Formula (I) as defined hereinbefore, or a pharmaceutically acceptable salt thereof, for the treatment of cancer. Accordingly there is therefore provided a method of treating cancer in a warm-blooded animal, such as man, in need of such treatment, which comprises administering to said animal an effective amount of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, as defined herein.In one embodiment of the invention, said cancer is selected from bladder cancer, head and neck cancer, prostate cancer, breast cancer, lung cancer, uterus cancer, pancreatic cancer, liver cancer, renal cancer, ovarian cancer, colon cancer, stomach cancer, skin cancer, cerebral tumor, malignant myeloma and lymphoproliferative tumors.
According to a further aspect of the invention, there is provided a method of using compound of Formula (I) as defined hereinbefore, or a pharmaceutically acceptable salt thereof, for the treatment of asthma, COPD, allergic rhinitis, allergic conjunctivitis, atopic dermatitis, hepatitis B, hepatitis C, HIV, HPV, bacterial infections or dermatosis.
According to a further aspect of the invention, there is provided a method of treating a human suffering from a disease in which activation of TLR7 is beneficial, comprising the steps of administering to a person in need thereof of a therapeutically effective amount of a compound of Formula (I) as defined hereinbefore, or a
pharmaceutically acceptable salt thereof. In one embodiment of the invention, the disease in which activation of TLR7 is beneficial is cancer. In a further embodiment of the invention, said cancer is selected from bladder cancer, head and neck cancer, prostate cancer, breast cancer, lung cancer, uterus cancer, pancreatic cancer, liver cancer, renal cancer, ovarian cancer, colon cancer, stomach cancer, skin cancer, cerebral tumor, malignant myeloma and lymphoproliferative tumors. In one embodiment of the invention, the disease in which activation of TLR7 is beneficial is asthma, COPD, allergic rhinitis, allergic conjunctivitis, atopic dermatitis, hepatitis B, hepatitis C, HIV, HPV, bacterial infections or dermatosis.
In any aspect or embodiment described herein the cancer may be bladder cancer. In any aspect or embodiment described herein the cancer may be head and neck cancer.
In any aspect or embodiment described herein the cancer may be prostate cancer.
In any aspect or embodiment described herein the cancer may be breast cancer.
In any aspect or embodiment described herein the cancer may be lung cancer.
In any aspect or embodiment described herein the cancer may be uterus cancer.
In any aspect or embodiment described herein the cancer may be pancreatic cancer.
In any aspect or embodiment described herein the cancer may be liver cancer.
In any aspect or embodiment described herein the cancer may be renal cancer.
In any aspect or embodiment described herein the cancer may be ovarian cancer.
In any aspect or embodiment described herein the cancer may be colon cancer.
In any aspect or embodiment described herein the cancer may be stomach cancer.
In any aspect or embodiment described herein the cancer may be skin cancer.
In any aspect or embodiment described herein the cancer may be cerebral tumor.
In any aspect or embodiment described herein the cancer may be malignant myeloma cancer.
In any aspect or embodiment described herein the cancer may be
lymphoproliferative tumors.
The anti-cancer treatment defined hereinbefore may be applied as a sole therapy or may involve, in addition to the compound of the invention, conventional surgery or radiotherapy or chemotherapy. Such chemotherapy may include one or more of the following categories of anti-tumour agents :-
(i) other antiproliferative/antineoplastic drugs and combinations thereof, as used in medical oncology, such as alkylating agents (for example cis-platin, oxaliplatin, carboplatin, miriplatin, cyclophosphamide, nitrogen mustard, melphalan, chlorambucil, busulphan, temozolamide and nitrosoureas); antimetabolites (for example gemcitabine and antifolates such as fluoropyrimidines like 5-fluorouracil and tegafur, raltitrexed, methotrexate, cytosine arabinoside, and hydroxyurea); antitumour antibiotics (for example anthracyclines like adriamycin, bleomycin, doxorubicin, daunomycin, epirubicin, amrubicin, idarubicin, mitomycin-C, dactinomycin and mithramycin); antimitotic agents (for example vinca alkaloids like vincristine, vinblastine, vindesine and vinorelbine and taxoids like taxol and taxotere and polokinase inhibitors); and topoisomerase inhibitors (for example epipodophyllotoxins like etoposide and teniposide, amsacrine, topotecan and camptothecin);
(ii) cytostatic agents such as antioestrogens (for example tamoxifen, fulvestrant, toremifene, raloxifene, droloxifene and iodoxyfene), antiandrogens (for example bicalutamide, flutamide, nilutamide and cyproterone acetate), LHRH antagonists or LHRH agonists (for example goserelin, leuprorelin and buserelin), progestogens (for example megestrol acetate), aromatase inhibitors (for example as anastrozole, letrozole, vorazole and exemestane) and inhibitors of 5a-reductase such as finasteride;
(iii) anti-invasion agents [for example c-Src kinase family inhibitors like 4-(6-chloro-2,3- methylenedioxyanilino)-7- [2-(4-methylpiperazin- 1 -yl)ethoxy] -5 -tetrahydropyran-4- yloxyquinazoline (AZD0530; International Patent Application WO 01/94341), N-(2- chloro-6-methylphenyl)-2- {6-[4-(2-hydroxyethyl)piperazin- 1 -yl]-2-methylpyrimidin-4- ylamino}thiazole-5-carboxamide (dasatinib, BMS-354825; J. Med. Chem., 2004, 47, 6658- 6661) and bosutinib (SKI-606), and metalloproteinase inhibitors like marimastat, inhibitors of urokinase plasminogen activator receptor function or antibodies to Heparanase];
(iv) inhibitors of growth factor function: for example such inhibitors include growth factor antibodies and growth factor receptor antibodies (for example the anti-erbB2 antibody trastuzumab [Herceptin™], the anti-EGFR antibody panitumumab, the anti-erbBl antibody cetuximab [Erbitux, C225] and any growth factor or growth factor receptor antibodies disclosed by Stern et al. Critical reviews in oncology/haematology, 2005, Vol. 54, ppl 1-29); such inhibitors also include tyrosine kinase inhibitors, for example inhibitors of the epidermal growth factor family (for example EGFR family tyrosine kinase inhibitors such as
N-(3-chloro-4-fluorophenyl)-7-methoxy-6-(3-morpholinopropoxy)quinazolin-4-amine (gefitinib, ZD 1839), N-(3-ethynylphenyl)-6,7-bis(2-methoxyethoxy)quinazolin-4-amine (erlotinib, OSI-774) and 6-acrylamido-N-(3-chloro-4-fluorophenyl)-7-(3- morpholinopropoxy)-quinazolin-4-amine (CI 1033), erbB2 tyrosine kinase inhibitors such as lapatinib); inhibitors of the hepatocyte growth factor family; inhibitors of the insulin growth factor family; inhibitors of the platelet-derived growth factor family such as imatinib and/or nilotinib (AMN107); inhibitors of serine/threonine kinases (for example Ras/Raf signalling inhibitors such as farnesyl transferase inhibitors, for example sorafenib (BAY 43-9006), tipifarnib (Rl 15777), BRAF inhibitor (PLX-4032) and lonafarnib (SCH66336)), inhibitors of cell signalling through MEK and/or AKT kinases, c-kit inhibitors, abl kinase inhibitors, PI3 kinase inhibitors, Plt3 kinase inhibitors, CSF-1R kinase inhibitors, IGF receptor (insulin-like growth factor) kinase inhibitors; aurora kinase inhibitors (for example AZD1152, PH739358, VX-680, MLN8054, R763, MP235, MP529, VX-528 AND AX39459) and cyclin dependent kinase inhibitors such as CDK2 and/or CDK4 inhibitors and BRAF inhibitors such as Vemurafenib;
(v) antiangiogenic agents such as those which inhibit the effects of vascular endothelial growth factor, [for example the anti-vascular endothelial cell growth factor antibody bevacizumab (Avastin™) and for example, a VEGF receptor tyrosine kinase inhibitor such as vandetanib (ZD6474), vatalanib (PTK787), sunitinib (SU11248), axitinib (AG-013736), pazopanib (GW 786034) and 4-(4-fluoro-2-methylindol-5-yloxy)-6-methoxy-7-(3- pyrrolidin-l-ylpropoxy)quinazoline (AZD2171; Example 240 within WO 00/47212), compounds such as those disclosed in International Patent Applications W097/22596, WO 97/30035, WO 97/32856 and WO 98/13354 and compounds that work by other
mechanisms (for example linomide, inhibitors of integrin ανβ3 function and angiostatin)];
(vi) vascular damaging agents such as Combretastatin A4 and compounds disclosed in International Patent Applications WO 99/02166, WO 00/40529, WO 00/41669,
WO 01/92224, WO 02/04434 and WO 02/08213;
(vii) an endothelin receptor antagonist, for example zibotentan (ZD4054) or atrasentan;
(viii) antisense therapies, for example those which are directed to the targets listed above, such as ISIS 2503, an anti-ras antisense;
(ix) gene therapy approaches, including for example approaches to replace aberrant genes such as aberrant p53 or aberrant BRCA1 or BRCA2, GDEPT (gene-directed enzyme pro-drug therapy) approaches such as those using cytosine deaminase, thymidine kinase or a bacterial nitroreductase enzyme and approaches to increase patient tolerance to chemotherapy or radiotherapy such as multi-drug resistance gene therapy; and
(x) immunotherapy approaches, including for example ex-vivo and in-vivo approaches to increase the immunogenicity 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, approaches to decrease the function of immune suppressive cells such as regulatory T cells, myeloid-derived suppressor cells or IDO (indoleamine 2,3,-deoxygenase)-expressing dendritic cells, and approaches using cancer vaccines consisting of proteins or peptides derived from tumour-associated antigens such as NY-ESO-1, MAGE-3, WT1 or Her2/neu.
According to this aspect of the invention there is provided a pharmaceutical product comprising a compound of the formula (I) as defined hereinbefore and an additional anti-tumour substance as defined hereinbefore for the conjoint treatment of cancer.
According to this aspect of the invention there is provided a pharmaceutical product comprising a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore, and an additional anti-tumour substance as defined hereinbefore for the conjoint treatment of cancer.
According to this aspect of the invention there is provided a pharmaceutical product comprising a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore, and an additional anti-tumour substance for the conjoint treatment of cancer.
According to this aspect of the invention there is provided a combination suitable for use in the treatment of cancer comprising a compound of Formula (I) as defined hereinbefore, or a pharmaceutically acceptable salt thereof, and any one of the anti tumour agents listed under (i) - (ix) above.
Therefore in a further aspect of the invention there is provided a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in combination with an anti- tumour agent selected from one listed under (i) - (ix) herein above.
Herein, where the term "combination" is used it is to be understood that this refers to simultaneous, separate or sequential administration. In one aspect of the invention "combination" refers to simultaneous administration. In another aspect of the invention "combination" refers to separate administration. In a further aspect of the invention "combination" refers to sequential administration. Where the administration is sequential or separate, the delay in administering the second component should not be such as to lose the beneficial effect of the combination.
According to a further aspect of the invention there is provided a pharmaceutical composition which comprises a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in combination with an anti-tumour agent selected from one listed under (i) - (ix) herein above, in association with a pharmaceutically acceptable diluent or carrier.
According to a further aspect of the invention there is provided a pharmaceutical composition which comprises a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in combination with an anti-tumour agent selected from one listed under (i) - (ix) herein above, in association with a pharmaceutically acceptable diluent or carrier for use in the treatment of cancer.
According to another feature of the invention there is provided the use of a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, in combination with an anti-tumour agent selected from one listed under (i) - (ix) herein above, in the manufacture of a medicament for use in cancer in a warm-blooded animal, such as man.
According to another feature of the invention there is provided a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, in combination with an anti- tumour agent selected from one listed under (i) - (ix) herein above for use in the treatment of cancer in a warm-blooded animal, such as man.
Therefore in an additional feature of the invention, there is provided a method of treating cancer in a warm-blooded animal, such as man, in need of such treatment which comprises administering to said animal an effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in combination with an anti-tumour agent selected from one listed under (i) - (ix) herein above.
According to a further aspect of the present invention there is provided a kit comprising a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in combination with an anti-tumour agent selected from one listed under (i) - (ix) herein above.
According to a further aspect of the present invention there is provided a kit comprising:
a) a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in a first unit dosage form;
b) an anti-tumour agent selected from one listed under (i) - (ix) herein above; in a second unit dosage form; and
c) container means for containing said first and second dosage forms. According to a further aspect of the present invention there is provided a kit comprising a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in combination with an additional anti-tumour agent.
According to a further aspect of the present invention there is provided a kit comprising:
a) a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in a first unit dosage form;
b) a second anti-tumour agent in a second unit dosage form; and
c) container means for containing said first and second dosage forms.
In one aspect of the invention the compounds of Formula (I) may be useful as vaccine adjuvants.
As a further aspect of the invention there is provided a compound of formula (I), or a pharmaceutically acceptable salt thereof, as defined herein, for use as a vaccine adjuvant.
As a further aspect of the invention there is provided the use of a compound of formula (I), or a pharmaceutically acceptable salt thereof, as defined herein, as a vaccine adjuvant, in the manufacture of a vaccine for the treatment of a disease or condition.
The invention still further provides a method of treating, or reducing the risk of, a disease or condition, which method comprises administering to a patient in need thereof a therapeutically effective amount of a vaccine and a compound of formula (I), or a pharmaceutically acceptable salt thereof, as defined herein.
The invention still further provides a method of increasing the response to a vaccine in a patient, which method comprises administering to a patient in need thereof a therapeutically effective amount of a vaccine and a compound of formula (I), or a pharmaceutically acceptable salt thereof, as defined herein.
Experimental Procedures (Chemical Syntheses and Biological Assays)
In the experimental procedures described below, the following abbreviations may be used: "EtOAc" = ethyl acetate; "min(s)" = minute(s); "THF" = tetrahydrofuran; "h" = hour(s); "FCC" = flash column chromatography using silica; "Amino FCC" = flash column chromatography using silica that is suface-modified by aminopropyl (HiFlash Column Amino 40 μΜ 60 A; Cat. No. W091, W092 or W093 packed with silica gel that is suface- modified by aminopropyl, purchased from Yamazen Science, Inc); "DMSO" = dimethylsulfoxide; "sat." = saturated aqueous solution; "RT" = room temperature; "LC- MS" = liquid chromatography with mass spectrometry; "m/z" = measured mass to charge ratio; "SCX" refers to solid phase extraction with a sulfonic acid sorbent; "M" = molarity; "EtOH" = ethanol. Proton nuclear magnetic resonance data ('aH NMR") was generally obtained at 300-500 MHz and using deuterated DMSO unless otherwise stated.
Abbreviations used for 1H NMR are: "s" = singlet, "d" = doublet; "t" = triplet; "q" = quartet; "m" = multiplet; "dd" = doublet of doublets; "br s" = broad singlet; "dt" = doublet of triplets, "td" = triplet of doublets, etc. Preparative reverse phase HPLC ("RPHPLC") was carried out using a Phenomenex Gemini™ CI 8 5 μιη column, using CH3CN in an aqueous 0.1% NH3 solution as eluent. Fractions were collected following detection by UV spectroscopy at a wavelength such as 220 or 254 nm.
Example 1: f2S -2-[f2-Amino-5-{4-[fdimethylamino)methyll-2-methoxybenzyl}-6- methylpyrimidin-4-yl)aminolpentan-l-ol
Figure imgf000032_0001
The title compound may be prepared by the sequence of steps described below:
(i) Methyl 4 2-(ethoxycarbonv0-3-oxobutyl1-3-methoxybenzoate
Figure imgf000032_0002
NaH (60%) in mineral oil, 1.45 g) was added portion-wise over 10 mins to a solution of ethyl acetoacetate (4.4 mL) in THF (60 mL) at 0°C. The mixture was then stirred for 10 mins. Then, a solution of methyl 4-(bromomethyl)-3-methoxybenzoate (7.5 g) in THF (40 mL) was added and the mixture was warmed to 70°C and stirred for 15h. The mixture was then poured into ice/water (300 mL) and stirred for 30 mins. The resulting aqueous mixture was extracted with EtOAc. The organic extracts were combined, dried and concentrated in vacuo to provide crude product. The reaction was repeated on the same scale. The two batches of crude product were combined and purified by FCC, eluting with 20-30% EtOAc in isohexane, to give the sub-title compound (14.7 g) as a colourless oil; 1H NMR: 7.48 (dd, 1H), 7.45 (d, 1H), 7.24 (d, 1H), 4.05 (q, 2H), 3.95 (dd, 1H), 3.86 (s, 3H), 3.84 (s, 3H), 3.10 (dd, 1H), 3.00 (dd, 1H), 2.17 (s, 3H), 1.09 (t, 3H).
(ii) Methyl 4- [(2-amino-4-hydroxy-6-methylpyrimidin-5 -yQmethyl] -3 -methoxybenzoate
Figure imgf000033_0001
Guanidinium carbonate (7.15 g) was added to a solution of methyl 4-[2-(ethoxycarbonyl)- 3 -oxobutyl] -3 -methoxybenzoate (8.95 g) in EtOH (60 mL) and the mixture was then heated at 80°C for 15h. After cooling the resulting solid was collected by filtration. This solid was then suspended in water and collected by filtration. The solid was then washed with EtOAc and dried to give the sub-title compound (5.8 g) as a colourless solid, which was used without further purification; 1H NMR: 10.78 (s, 1H), 7.46 (d, 1H), 7.45 (s, 1H), 6.98 (d, 1H), 6.34 (s, 2H), 3.89 (s, 3H), 3.83 (s, 3H), 3.61 (s, 2H), 1.93 (s, 3H); LC-MS: m/z 304.
(iii) Methyl 4- [(2-amino-4-chloro-6-methylpyrimidin-5 -yDmethyl] -3 -methoxybenzoate
Figure imgf000033_0002
Methyl 4-[(2-amino-4-hydroxy-6-methylpyrimidin-5-yl)methyl]-3-methoxybenzoate (8.6 g) was added to POCl3 and the mixture was heated at 100°C for 15h. The solvent was then removed in vacuo. The resulting residue was diluted with water and the pH adjusted to 7 by the addition of sat. NaHCOs. This mixture was heated at 50°C for 2h, and was then extracted with EtOAc. The combined extracts were dried and concentrated in vacuo to give the sub-title compound (9.05 g) as a colourless solid which was used without further purification; 1H NMR: 7.50 (s, 2H), 7.49 (d, 1H), 6.90 (s, 1H), 6.81 (d, 1H), 3.92 (s, 3H), 3.90 (s, 3H), 3.84 (s, 2H), 2.16 (s, 3H).
(iv) Methyl 4- [(2-amino-4- { \(2S)- 1 -hydroxypentan-2-yl] amino I -6-methylpyrimidin-5 - vDmethyll -3 -methoxybenzo ate
Figure imgf000034_0001
(5)-2-Aminopentan-l-ol (0.481 g) was added to a suspension of methyl 4-[(2-amino-4- chloro-6-methylpyrimidin-5-yl)methyl]-3-methoxybenzoate (0.5 g) in dioxane (5 mL). The mixture was heated in a CEM Discover™ microwave at 170°C for 9h. The mixture was then cooled and concentrated in vacuo. Purification by FCC, eluting with 5% CH3OH in CH2CI2 gave the sub-title compound (0.25 g) as a orange gum; 1H NMR: 7.53-7.42 (m, 2H), 6.89 (d, 1H), 5.75 (d, 2H), 5.58 (d, 1H), 4.61-4.54 (m, 1H), 4.17-4.08 (m, 1H), 3.92 (s, 3H), 3.83 (s, 3H), 3.69 (d, 2H), 3.28-3.23 (m, 1H), 2.00 (d, 3H), 1.53-1.40 (m, 1H), 1.35-1.19 (m, 2H), 1.16-1.02 (m, 2H), 0.77 (t, 3H); LC-MS: m/z 389.
(v) 4- r(2-Amino-4- { \(2S)- 1 -hydroxypentan-2-νΠ amino I -6-methylpyrimidin-5 -vDmethyll- 3-methoxybenzoic acid
Figure imgf000034_0002
Methyl 4-[(2-amino-4- {[(25)- 1 -hydroxypentan-2-yl]amino} -6-methylpyrimidin-5- yl)methyl]-3-methoxybenzoate (220 mg) was dissolved into a mixture of THF (3 mL) and water (3 mL), and LiOH solution (3M, 0.944 mL) was added. The mixture was heated in a CEM Discover™ microwave for 6h at 120°C. The mixture was concentrated in vacuo and the resulting residue was dissolved in water and the mixture acidified to pH7 using 2M HCl. The resulting precipitate was filtered off and dried under high vacuum overnight to give the sub-title compound (90 mg); 1H NMR: 7.53-7.42 (m, 2H), 6.92 (d, 1H), 6.61 (d, 2H), 4.21 (d, 1H), 3.91 (d, 3H), 3.75 (s, 2H), 2.09 (s, 3H), 1.53-1.42 (m, 1H), 1.40-1.20 (m, 2H), 1.09 (q, 2H), 0.78 (t, 3H); LC-MS: m/z 375. (vi) (25V2-({2-Amino-5-[4-(hydroxymethy^
yl| amino)pentan- 1 -ol
Figure imgf000035_0001
BH3-THF complex (1M, 3 mL) was added to 4-[(2-amino-4-{[(25)-l-hydroxypentan-2- yl]amino}-6-methylpyrimidin-5-yl)methyl]-3-methoxybenzoic acid (85 mg) and the mixture was heated to 80°C for 2h. CH3OH was then carefully added and the mixture was heated to 80°C for lh. The mixture was concentrated in vacuo and the resulting residue was dissolved in CH3OH and loaded onto an SCX cartridge. The cartridge was washed with CH3OH and then eluted with a solution of 0.7M NH3 in CH3OH. Concentration in vacuo gave the sub-title compound (40 mg) as a colourless gum; 1H NMR: 6.94 (s, 1H), 6.74 (q, 2H), 5.66 (s, 2H), 5.48 (t, 1H), 5.14 (s, 1H), 4.59 (s, 1H), 4.45 (s, 2H), 4.15-4.04 (m, 1H), 3.84 (d, 3H), 3.59 (d, 2H), 3.25 (dd, 1H), 2.03 (s, 3H), 1.54-1.44 (m, 1H), 1.32- 1.23 (m, 2H), 1.11 (m, 2H), 0.81-0.76 (t, 3H); LC-MS: m/z 361.
(vii) 4- IY2- Amino-4- { \(2S)- 1 -hydroxypentan-2-νΠ amino I -6-methylpyrimidin-5 -vD- methyll -3 -methoxybenzaldehyde
Figure imgf000035_0002
Mn02 (9 g) was added to a solution of (25)-2-({2-amino-5-[4-(hydroxymethyl)-2- methoxybenzyl]-6-methylpyrimidin-4-yl}amino)pentan-l-ol (930 mg) in CH2C12 (15 mL) and the mixture was stirred at RT for 3h. The resulting mixture was filtered through a pad of diatomaceous earth (Celite™), and the filtrate was concentrated in vacuo to give the sub-title compound (706 mg) as a yellow solid; 1H NMR: 9.92 (s, 1H), 7.44 (d, 1H), 7.41 (dd, 1H), 6.96 (d, 1H), 5.70 (br s, 2H), 5.55 (d, 1H), 4.55 (dd, 1H), 4.15-4.07 (m, 1H), 3.94 (s, 3H), 3.71 (s, 2H), 3.38-3.20 (m, 2H), 1.98 (s, 3H), 1.52-1.41 (m, 1H), 1.32-1.20 ( m, 1H), 1.12-1.05 (m, 2H), 0.75 (t, 3H). (Viii) (26 -2-[(2-Amino-5- (4-[(dimethylamino)methyll-2-methoxybenzyl| -6-methyl-
Figure imgf000036_0001
Dimethylamine (2M in THF, 125 μί) was added to a solution of 4-[(2-amino-4-{[(25)-l- hydroxypentan-2-yl] amino } -6-methylpyrimidin-5 -yl)-methyl] -3 -methoxybenzaldehyde (30 mg) in THF (1 mL), and the mixture was stirred at RT for 40 mins. Sodium triacetoxy- borohydride (71 mg) was added, and the resulting mixture was stirred at RT for 6h. Sat. Na2C03 and water were added. The mixture was extracted with EtOAc (3 x 10 mL). The combined organic extracts were washed with brine, dried (Na2S04) and concentrated in vacuo. Purification by Amino FCC, eluting with CHCI3/CH3OH, gave the title compound (24 mg) as a colourless gum; 1H NMR: 6.88 (d, 1H), 6.74 (d, 1H), 6.71 (dd, 1H), 5.63 (br s, 2H), 5.40 (d, 1H), 4.58 (dd, 1H), 4.12-4.01 (m, 1H), 3.82 (s, 3H), 3.59 (s, 2H), 3.37-3.21 (m, 4H), 2.10 (s, 6H), 2.05 (s, 3H), 1.52-1.40 (m, 1H), 1.29-1.17 (m, 1H), 1.08-0.99 (m, 2H), 0.73 (t, 3H).
Alternatively, the title compound may be prepared according to the sequence of steps described below:
(ix) 2-Amino-5-(4-cvano-2-methoxybenzyl)-6-methylpyrimidin-4-yl 2,4,6- triisopropylbenzenesulfonate
Figure imgf000036_0002
4-[(2-Amino-4-hydroxy-6-methylpyrimidin-5-yl)methyl]-3-methoxybenzonitrile
(preparation: see step (ii) of Example 16, below; 9.6 g) was mixed with 2,4,6- triisopropylbenzene-l-sulfonyl chloride (13.98 g) and l,4-diazabicyclo[2.2.2]octane (5.18 g) in THF (250 mL) and this mixture was heated at 30°C for 7h. The mixture was partitioned between EtOAc and sat. NaCl solution. The combined organic extracts were dried (MgS04), and concentrated in vacuo. The residue was triturated with a mixture of isohexane (30 mL) and diethyl ether (1 mL) to give the sub-title compound (17 g) as a white solid; LC-MS: m/z 537 APCI+
(x) 4- \(2 -Amino-4- { \(2S)- 1 -hydroxypentan-2-νΠ amino I -6-methylpyrimidin-5 -vDmethvH- 3 -methoxybenzonitrile
Figure imgf000037_0001
2-Amino-5-(4-cyano-2-methoxybenzyl)-6-methylpyrimidin-4-yl 2,4,6-triisopropyl- benzenesulfonate (5 g), (5)-2-aminopentan-l-ol (2.88 g) and trifluoroacetic acid (0.699 mL) were mixed together in butyronitrile (50 mL) and heated at 120°C for 30h. The solvent was removed in vacuo, and the residue was dissolved in CH3OH. Purification by FCC, eluting with 10% CH3OH (containing 5% 0.7M NH3) in CH2C12 gave the sub-title compound (2.7 g) as a yellow gum; LC-MS: m/z 356 APCI+
(xi) 4- [(2- Amino-4- { \(2S)- 1 -hydroxypentan-2-yl] amino I -6-methylpyrimidin-5 -yDmethyl]- 3 -methoxybenzaldehyde
Figure imgf000037_0002
4-[(2 -Amino-4- { [(2S)- 1 -hydroxypentan-2-yl] amino } -6-methylpyrimidin-5 -yl)methyl] -3 - methoxybenzonitrile (2.7 g) was dissolved in pyridine (28 mL), acetic acid (14 mL) and water (14 mL), and sodium hypophosphite monohydrate (5.64 g) was added, followed by Raney nickel (1.34 g) according to the method described for step (v) of Example 16, below, to give the sub-title compound (2.4 g) as a yellow foam; LC-MS: m/z 359 APCI+ (xii) (26 -2-r(2-Amino-5- (4-r(dimethylamino)methyl1-2-methoxybenzyl| -6- methylpyrimidin-4-yl)amino"|pentan- 1 - l
Figure imgf000037_0003
Dimethylamine (2M in THF, 1.67 mL) and 4-[(2-amino-4-{[(25)-l-hydroxypentan-2- yl]amino}-6-methylpyrimidin-5-yl)methyl]-3-methoxybenzaldehyde (400 mg) were reacted together according to the general method described in step (vi) of Example 16, below. After stirring for 16h the solvent was removed in vacuo and CH3OH and DMSO was added. Purification by RPHPLC gave the title compound (87.6 mg) as a colourless gum; LC-MS: m/z 388 multimode+
Example 1 A: ( 2S)-2- \( 2- Amino-5- {4- \( dimethylamino)methyll -2-methoxybenzyl}-6- methylpyrimidin-4-yl)aminolpentan-l-ol bis saccharin salt
Figure imgf000038_0001
salt
(2iS)-2-[(2 -Amino-5- {4-[(dimethylamino)methyl]-2-methoxybenzyl}-6-methylpyrimidin- 4-yl)amino]pentan-l-ol (the title compound of Example 1, above; 7.6 mg) was dissolved in CH3CN and saccharin (2 eq.) was added to give a solution. The solvent was removed by blowing N2 over the solution. The solid was then dried in vacuo to give the title compound (15 mg) as a solid; 1H NMR: 11.92 (s, 1H), 9.52 (s, 1H), 7.67-7.54 (m, 7H), 7.13 (s, 1H), 6.93 (s, 2H), 4.75 (t, 1H), 4.25 (d, 2H), 3.88 (s, 3H), 3.74 (s, 2H), 3.39 (ddd, 3H), 2.69 (s, 3H), 2.17 (s, 3H), 2.08 (s, 2H), 1.55-1.41 (m, 2H), 1.35 (ddd, 2H), 1.07 (td, 2H), 0.78 (t, 3H).
Example 2: f2S -2-f{2-Amino-5-[2-methoxy-4-fpiperazin-l-ylmethyl)benzyll-6- methylpyrimidin-4-yl}amino)pentan-l-ol
Figure imgf000038_0002
Piperazine (36 mg) was added to a solution of 4-[(2-amino-4-{[(25)-l-hydroxypentan-2- yljamino} -6-methylpyrimidin-5-yl)-methyl]-3-methoxybenzaldehyde (preparation: see step (vii) of Example 1, above; 30 mg) in CH3OH (1 mL), and the mixture was stirred under reflux for 2h. After cooling, NaBH4 (16 mg) was added, and the resulting mixture was stirred at RT for lh. Water was then added and the mixture was extracted with EtOAc (3 x 10 mL). The combined organic extracts were washed with brine, dried (Na2S04) and concentrated in vacuo. Purification by Amino FCC, eluting with CHCI3/CH3OH gave the title compound (27 mg) as a colourless gum; 1H NMR (CDC13): 6.88-6.82 (m, 2H), 6.77 (dd, 1H), 4.91 (d, 1H), 4.65 (br s, 2H), 4.01-3.91 (m, 1H), 3.87 (s, 3H), 3.66 (s, 2H), 3.59 (dd, 1H), 3.44-3.36 (m, 3H), 2.86-2.83 (m, 4H), 2.36-2.31 (m, 7H), 1.44-1.33 (m, 1H), 1.29-1.16 (m, 1H), 1.10-1.00 (m, 2H), 0.76 (t, 3H).
Example 3 : ( 2S)-2- \( 2-Amino-5-{2-methoxy-4-[( methylamino)methyll benzylj-6- methylpyrimidin-4-yl)aminolpentan-l-ol
Figure imgf000039_0001
Methylamine (2M in THF, 0.12 mL) was reacted 4-[(2-amino-4-{[(25)-l-hydroxypentan- 2-yl]amino}-6-methylpyrimidin-5-yl)-methyl]-3-methoxybenzaldehyde using the general method described for step (viii) of Example 1 , to give the title compound (27 mg) as a colourless gum; 1H NMR: 6.94 (s, 1H), 6.75-6.69 (m, 2H), 5.62 (br s, 2H), 5.42 (d, 1H), 4.58 (dd, 1H), 4.11-4.03 (m, 1H), 3.83 (s, 3H), 3.58 (s, 2H), 3.56 (s, 2H), 3.37-3.22 (m, 2H), 2.22 (s, 3H), 2.03(s, 3H), 1.52-1.41 (m, 1H), 1.30-1.18 (m, 1H), 1.10-1.02 (m, 2H), 0.75 (t, 3H).
Example 4 : ( 2S)-2- { [2- Amino-5-( 4- { \( 2-hydroxyethyl)aminol methyll-2-methoxy- benzyl)-6-methylpyrimidin-4-yllamino}pentan-l-ol
Figure imgf000039_0002
2-Aminoethanol (50 μί) was reacted with 4-[(2-amino-4-{[(25)-l-hydroxypentan-2- yl]amino}-6-methylpyrimidin-5-yl)-methyl]-3-methoxybenzaldehyde using the general method described for Example 2, to give the title compound (61 mg) as a colourless gum; 1H NMR: 6.95 (s, 1H), 6.75 (d, 1H), 6.71 (d, 1H), 5.62 (br s, 2H), 5.42 (d, 1H), 4.58 (t, 1H), 4.44 (dd, 1H), 4.12-4.03 (m, 1H), 3.83 (s, 3H), 3.64 (s, 2H), 3.58 (s, 2H), 3.43 (q, 2H), 3.33-3.23 (m, 2H), 2.54-2.48 (m, 2H), 2.03 (s, 3H), 1.52-1.41 (m, 1H), 1.30-1.20 (m, 1H), 1.1 1-1.02 (m, 2H), 0.75 (t, 3H).
Example 5 : ( 2S)-2- \( 2- Amino-5- {4- \( azetidin-3-ylamino)methyll -2-methoxybenzyl}-6- methylpyrimidin-4-yl)aminolpentan-l-ol
Figure imgf000040_0001
The title compound may be prepared by the sequence of steps described below:
(Ί) tert-Butyl 3 -( (4- [(2-amino-4- { \(2S)- 1 -hydroxypentan-2-yl] amino I -6-methylpyrimidin-
5-yl)methyl]-3-methoxybenzyl| amino)azetidine- 1 -carboxylate
Figure imgf000040_0002
tert-Butyl 3-aminoazetidine-l -carboxylate (144 mg) was reacted 4-[(2-amino-4- {[(25)-l- hydroxypentan-2-yl] amino } -6-methylpyrimidin-5 -yl)-methyl] -3 -methoxybenzaldehyde using the general method described for Example 2, above, to give the sub-title compound (105 mg) as a colourless oil; 1H NMR: 6.93 (s, 1H), 6.69 (d, 1H), 6.73 (d, 1H), 5.62 (br s, 2H), 5.44 (d, 1H), 4.57 (t, 1H), 4.1 1-4.04 (m, 1H), 3.83 (s, 3H), 3.59-3.32 (m, 1 1H), 2.01 (s, 3H), 1.50-1.42 (m, 1H), 1.35 (s, 9H), 1.34-1.04 (m, 3H), 0.76 (t, 3H).
(ii) (2S)-2-\(2 -Amino-5- {4-[(azetidin-3-ylamino)methyll-2-methoxybenzyl| -6-
Figure imgf000041_0001
HC1 (4M in dioxane, 1.5 mL) was added to a solution of tert-butyl 3-({4-[(2-amino-4- { [(2iS)- 1 -hydroxypentan-2-yl] amino } -6-methylpyrimidin-5 -yl)methyl] -3 -methoxy- benzyl}amino)azetidine-l-carboxylate (50 mg) in CH3OH (1 mL) and the mixture was stirred at RT for 3h. The mixture was then concentrated in vacuo. Sat. Na2C03 was added, and the mixture was extracted with CHCl3-EtOH (3: 1). The combined organic extracts were washed with brine, dried (Na2S04) and concentrated in vacuo. Purification by Amino FCC, eluting with CHCI3/CH3OH gave the sub-title compound (21 mg) as a colourless gum; 1H NMR (CDC13): 6.86 (d, 1H), 6.83 (d, 1H), 6.73 (dd, 1H), 4.88 (d, 1H), 4.71 (br s, 2H), 4.01-3.92 (m, 1H), 3.86 (s, 3H), 3.69-3.64 (m, 7H), 3.55 (dd, 1H), 3.41-3.35 (m, 3H), 2.29 (s, 3H), 1.46-1.33 (m, 1H), 1.29-1.03 (m, 3H), 0.78 (t, 3H).
Example 6 : ( 2S)-2- { 12- Amino-5-( 2-methoxy-4-{ Γ ( l-methylazetidin-3- yl)aminolmethyl}-benzyl)-6-meth lpyrimidin-4-yllamino}pentan-l-ol
Figure imgf000041_0002
LiAlH4 (8.4 mg) was added to a solution of tert-butyl 3-({4-[(2-amino-4-{[(2S)-l- hydroxypentan-2-yl] amino } -6-methylpyrimidin-5 -yl)methyl] -3 -methoxybenzyl} - amino)azetidine-l-carboxylate (preparation: see step (i) of Example 5, above; 57 mg) in THF (1.5 mL) and the mixture was stirred under reflux for 5h. Further LiAlH4 (17 mg) was then added, and the mixture was stirred under reflux for 5h. After cooling to RT, water (25 μί), 15% aqueous NaOH (25 μί), and water (75 μί) were added sequentially. The mixture was stirred at RT for lh, then filtered through a pad of diatomaceous earth (Celite™), and the filtrate was concentrated in vacuo. Purification by Amino FCC, eluting with CHCI3/CH3OH gave the title compound (15 mg) as a colourless gum; 1H NMR (CDCI3): 6.86 (d, 1H), 6.83 (d, 1H), 6.73 (dd, 1H), 4.88 (d, 1H), 4.67 (br s, 2H), 4.01-3.94 (m, 1H), 3.87 (s, 3H), 3.65 (s, 2H), 3.64 (s, 2H), 3.60-3.50 (m, 3H), 3.44-3.35 (m, 2H), 2.73 (dd, 2H), 2.30 (s, 3H), 2.27 (s, 3H), 1.45-1.34 (m, 1H), 1.29-1.04 (m, 3H), 0.79 (t, 3H).
Example 7: f3S -3-[f2-Amino-5-{2-methoxy-4-[fmethylamino)methyllbenzyl}-6- methylpyrimidin-4-yl)aminolhexan-l- l
Figure imgf000042_0001
The title compound may be prepared by the sequence of steps described below:
(Ί) Methyl 4- [(2-amino-4- { \(3S)- 1 -hydroxyhexan-3 -yl] amino I -6-methylpyrimidin-5 - y Dmethy 1] -3 -methoxybenzo ate
Figure imgf000042_0002
(5)-3-Aminohexan-l-ol (0.72 g) was reacted methyl 4-[(2-amino-4-chloro-6- methylpyrimidin-5-yl)methyl]-3-methoxybenzoate using the general method described in step (iv) of Example 1 , above, to give the sub-title compound (0.53 g) as a white solid; 1H NMR: 7.47 (s, 1H), 7.45 (d, 1H), 6.83 (d, 1H), 5.72 (br s, 2H), 5.68 (d, 1H), 4.37 (t, 1H), 4.23-4.15 (m, 1H), 3.91 (s, 3H), 3.82 (s, 3H), 3.68 (s, 2H), 3.32-3.26 (m, 2H), 1.94 (s, 3H), 1.62-1.51 (m, 1H), 1.46-1.31 (m, 3H), 1.16-1.04 (m, 2H), 0.76 (t, 3H).
(ii) (36 -3-((2-Amino-5-r4-(hvdroxymethyl)-2-methoxybenzyl1-6-methylpyrimidin-4- yl| amino)hexan- 1 -ol
Figure imgf000042_0003
LiBH4 (2M in THF, 2 mL) was added to a solution of methyl 4-[(2-amino-4-{[(3S)-l- hydroxyhexan-3 -yl] amino } -6-methylpyrimidin-5 -yl)methyl] -3 -methoxybenzoate (530 mg) in THF (5 mL) and CH3OH (0.5 mL), and the mixture was stirred at 60°C for 1.5h. After cooling to RT, HC1 (4M, 2.5 mL) was added and the mixture was stirred at RT for 20 mins. The mixture was basified with sat. Na2C03, and then extracted with EtOAc (3 x 15 mL). The combined organic extracts were washed with brine, dried (Na2S04) and concentrated in vacuo. The residue was treated with hexane/EtOAc (1 : 1, 5mL) and the resulting solid was collected by filtration to give the title compound (295 mg) as a colourless gum; 1H NMR: 6.93 (s, 1H), 6.74 (d, 1H), 6.68 (d, 1H), 5.68 (br s, 2H), 5.60 (d, 1H), 5.12 (t, 1H), 4.44 (d, 2H), 4.39 (dd, 1H), 4.22-4.14 (m, 1H), 3.83 (s, 3H), 3.59 (s, 2H), 3.32-3.26 (m, 2H), 1.98 (s, 3H), 1.63-1.52 (m, 1H), 1.47-1.31 (m, 3H), 1.19-1.04 (m, 2H), 0.77 (t, 3H). (iii) 4- IY2- Amino-4- { \(3S)- 1 -hydroxyhexan-3 -yll amino I -6-methylpyrimidin-5 -vDmethyll - 3 -methoxybenzaldehyde
Figure imgf000043_0001
Mn02 (2.8 g) was added to a suspension of (35)-3-({2-amino-5-[4-(hydroxymethyl)-2- methoxybenzyl]-6-methylpyrimidin-4-yl}amino)hexan-l-ol (280 mg) in CH2C12 (10 mL) and the mixture was stirred at RT for 20 mins. THF (10 mL) was added and the resulting mixture was stirred at RT for 3.5h. The mixture was then filtered through a pad of diatomaceous earth (Celite™), and the filtrate was concentrated in vacuo to give the subtitle compound (239 mg) as a yellow solid; 1H NMR: 9.92 (s, 1H), 7.44 (s, 1H), 7.42 (d, 1H), 6.92 (d, 1H), 5.73 (br s, 2H), 5.71 (d, 1H), 4.38 (br s, 1H), 4.24-4.16 (m, 1H), 3.94 (s, 3H), 3.71 (s, 2H), 3.32-3.27 (m, 2H), 1.94 (s, 3H), 1.63-1.52 (m, 1H), 1.47-1.32 (m, 3H), 1.19-1.05 (m, 2H), 0.76 (t, 3H).
(iv) (36 -3-[(2-Amino-5- (2-methoxy-4-[(methylamino)methyllbenzyl| -6-methyl- pyrimidin-4-yl)amino"|hexan- 1 -ol
Figure imgf000043_0002
4-[(2 -Amino-4- { [(35)- 1 -hydroxyhexan-3 -yl] amino } -6-methylpyrimidin-5 -yl)methyl] -3 - methoxybenzaldehyde (150 mg) and methylamine (2M in THF, 0.27 mL) were reacted using the general method described for Example 2, above, to give the title compound (30 mg) as a colourless gum; 1H NMR: 6.94 (s, IH), 6.74 (d, IH), 6.68 (d, IH), 5.66 (br s, 2H), 5.54 (d, IH), 4.38 (br s, IH), 4.21-4.12 (m, IH), 3.83 (s, 3H), 3.58 (s, 2H), 3.57 (s, 2H), 3.32-3.25 (m, 2H), 2.22 (s, 3H), 2.00 (s, 3H), 1.62-1.51 (m, IH), 1.46-1.31 (m, 3H), 1.14- 1.04 (m, 2H), 0.76 (t, 3H).
Example 8: f3S -3-f{2-Amino-5-[2-methoxy-4-fpiperazin-l-ylmethyl)benzyll-6- methylpyrimidin-4-yl}amino)hex -l- l
Figure imgf000044_0001
4-[(2-Amino-4- { [(35)- 1 -hydroxyhexan-3 -yl] amino } -6-methylpyrimidin-5 -yl)methyl] -3 - methoxybenzaldehyde (preparation: see step (iii) of Example 7, above; 45 mg) and piperizine (42 mg) were reacted according to the general method described for Example 2, above, to give the title compound (41 mg) as a colourless gum; 1H NMR (CDCI3): 6.86- 6.84 (m, 2H), 6.75 (dd, IH), 4.71 (br s, 2H), 4.60 (d, IH), 4.10-4.00 (m, IH), 3.86 (s, 3H), 3.63 (s, 2H), 3.39 (s, 2H), 3.36 (ddd, IH), 3.36 (ddd, IH), 2.83-2.80 (m, 4H), 2.33-2.30 (m, 7H), 1.81-1.70 (m, IH), 1.41-1.30 (m, IH), 1.25-1.16 (m, 2H), 1.05-0.93 (m, 2H), 0.70 (t, 3H).
Example 9 : ( 3S)-3- { [2- Amino-5-( 4- { \( 2-hydroxyethyl)aminol methyll-2-methoxy- benzyl)-6-methylpyrimidin-4-yllamin hexan-l-ol
Figure imgf000044_0002
4-[(2-Amino-4- { [(35)- 1 -hydroxyhexan-3 -yl] amino } -6-methylpyrimidin-5 -yl)methyl] -3 - methoxybenzaldehyde (preparation: see step (iii) of Example 7, above; 45 mg) and 2- aminoethanol (30 μί) were reacted togther using the general method described for
Example 2, above, to give the title compound (40 mg) as a colourless gum; 1H NMR (CDCI3): 6.90-6.86 (m, 2H), 6.78 (dd, IH), 4.70-4.66 (m, 3H), 4.13-4.03 (m, IH), 3.89 (s, 3H), 3.75 (s, 2H), 3.65 (s, 2H), 3.64 (t, 2H), 3.38 (ddd, IH), 3.38 (ddd, IH), 2.77 (t, 2H), 2.33 (s, 3H), 1.84-1,72 (m, IH), 1.45-1.34 (m, IH), 1.28-1.16 (m, 2H), 1.12-0.98 (m, 2H), 0.74 (t, 3H).
Example 9: Alternative process:
(i) 2-amino-5-(4-(hydroxymethyl)-2-methoxybenzyl)-6-methylpyrimidin-4-ol
Figure imgf000045_0001
Lithium triethylborohydride (93 mL) was added to a solution of methyl 4-((2-amino-4- hydroxy-6-methylpyrimidin-5-yl)methyl)-3-methoxybenzoate (5.0 g, 17 mmol) in THF (25mL) over 5 min and the reaction mixture was stirred at r.t. for 3.5 h. Water (60 mL) and 2N-HC1 (40 mL) was added to the mixture. The organic solvent was removed by evaporation. 2N-HC1 (16 mL) was added and the mixture was stirred at r.t. for 4 h. The mixture was neutralized by saturated aqueous NaHC03. The precipitate was collected by filtration, washed with water and dried in vacuo at 50 °C to afford 4.7 g (quant.) of the subtitle compound as a white solid; 1H NMR: 10.76 (IH, br s), 6.89 (IH, s), 6.77-6.70 (2H, m), 6.30 (2H, br s), 5.10 (IH, t), 4.42 (2H, d), 3.79 (3H, s), 3.51 (2H, s), 1.91 (3H, s).
(ii) 2-amino-5-(4-(hydroxymethyl)-2-methoxybenzyl)-6-methylpyrimidin-4-yl 2,4,6- trimethylbenzenesulfonate
Figure imgf000045_0002
2-Mesitylenesulfonyl chloride (7.2 g, 33 mmol) was added to a suspension of diisopropylethylamine (5.5 mL, 33 mmol) and the product from step (i) (6.1 g, 22 mmol) in THF (200 mL) and the mixture was stirred under reflux for 12 h. The resulting mixture was diluted with water and extracted with EtOAc. The combined organic layer was washed with brine, dried over Na2S04 and concentrated under reduced pressure. The residue was purified by flash column chromatography on amino silica gel to afford 8.6 g (85%) of the subtitle compound as a white solid; 1H NMR (CDC13): 6.92 (2H, s), 6.86 (1H, s), 6.77 (2H, s), 4.67 (2H, br s), 4.64 (2H, d), 3.82 (3H, s), 3.81 (2H, s), 2.57 (6H, s), 2.29 (3H, s), 2.21 (3H, s), 1.81 (lH, t).
(iii) 2-amino-5-(4-(chloromethyl)-2-methoxybenzyl)-6-methylpyrimidin-4-yl 2,4,6- trimethylbenzenesulfonate
Figure imgf000046_0001
Methansulfonyl chloride (1.4 mL, 17 mmol) was added to a mixture of lithium chloride (0.74 g, 17 mmol) and the product from step (ii) (4.0 g, 8.7 mmol) in THF (45 mL). The mixture was stirred at r.t. for 1.5 h, diluted with water and extracted with EtOAc. The combined organic layer was washed with brine, dried over Na2S04 and concentrated under reduced pressure to afford the crude product as a pale yellow oil, which was used for next reaction without further purification; 1H NMR (CDC13): 6.91 (2H, s), 6.83 (1H, s), 6.81-6.74 (2H, m), 4.69 (2H, br s), 4.54 (2H, s), 3.82 (3H, s), 3.80 (2H, s), 2.54 (6H, s), 2.29 (3H, s), 2.24 (3H, s).
(iv) 2-amino-5-(4-((2-hydroxyethylamino)methyl)-2-methoxybenzyl)-6-methylpyrimidin- 4-yl 2,4,6-trimethylbenzenesulfonate
Figure imgf000046_0002
2-Aminoethanol (2.7 g, 44 mmol) was added to the mixture of K2CO3 (6.0 g, 44 mmol), KI (0.15 g, 0.87 mmol) and the crude product from step (iii) in CH3CN (30 mL). After the mixture was stirred at 60°C for 4 h, the reaction mixture was cooled to r.t., filtered, and concentrated under reduced pressure. The residue was purified by flash column
chromatography on amino silica gel to afford 3.0 g (69% for 2 steps) of the subtitle compound as a colourless amorphous solid; 1H NMR (CDCI3): 6.92 (2H, s), 6.78 (1H, s), 6.77-6.70 (2H, m), 4.66 (2H, br s), 3.81 (3H, s), 3.79 (2H, s), 3.75 (2H, s), 3.65 (2H, t), 2.79 (2H, t), 2.57 (6H, s), 2.29 (3H, s), 2.23 (3H, s).
(v) (S)-3-(2-amino-5-(4-((2-hvdroxyethylamino)methyl)-2-methoxybenzyl)-6- methylpyrimidin-4-ylamino)hexan- 1 -ol
Figure imgf000047_0001
Trifluoroacetic acid (0.46 mL, 6.0 mmol) was added to the mixture of (S)-3-aminohexan- l-ol (2.1 g, 18 mmol) and the product from step (iv) (3.0 g, 6.0 mmol) in propionitrile (15 mL). The mixture was heated at 120°C for 10 h and cooled to r.t. The solvent was removed by evaporation. The residue was purified by flash column chromatography on amino silica gel to afford 1.3 g (52%) of the subtitle compound as a colourless gum; 1H NMR (CDCI3): 6.90-6.86 (m, 2H), 6.78 (dd, 1H), 4.70-4.66 (m, 3H), 4.13-4.03 (m, 1H), 3.89 (s, 3H), 3.75 (s, 2H), 3.65 (s, 2H), 3.64 (t, 2H), 3.38 (ddd, 1H), 3.38 (ddd, 1H), 2.77 (t, 2H), 2.33 (s, 3H), 1.84-1,72 (m, 1H), 1.45-1.34 (m, 1H), 1.28-1.16 (m, 2H), 1.12-0.98 (m, 2H), 0.74 (t, 3H).
Example 10: TV2- {4- \(2- Amino-4- { \(3S)- l-hvdroxyhexan-3-yll aminol-6-methyl- pyrimidin-5-yl)methyll-3-methoxybenzyl}glycinamide
Figure imgf000047_0002
4-[(2 -Amino-4- { [(35)- 1 -hydroxyhexan-3 -yl] amino } -6-methylpyrimidin-5 -yl)methyl] -3 - methoxybenzaldehyde (preparation: see step (iii) of Example 7, above; 45 mg) and glycinamide hydrochloride (53 mg) were reacted together using the general method of Example 2, above, to give the title compound (35 mg) as a pale yellow gum; 1H NMR (CDC13): 6.95-6.89 (m, 2H), 6.80-6.78 (m, 2H), 5.43 (br s, IH), 4.75 (br s, 2H), 4.68 (d, IH), 4.15-4.06 (m, IH), 3.89 (s, 3H), 3.74 (s, 2H), 3.65 (s, 2H), 3.42 (ddd, IH), 3.28 (ddd, IH), 3.26 (s, 2H), 2.33 (s, 3H), 1.46-1.35 (m, IH), 1.28-1.16 (m, 3H), 1.14-1.01 (m, 2H), 0.75 (t, 3H).
Example 11: TV3- {4- if 2- Amino-4- { ί ( 35)- l-hydroxyhexan-3-yll aminol-6-methyl- pyrimidin-5-yl)methyll-3-methox benzyl}-|¾-alaninamide
Figure imgf000048_0001
4-[(2 -Amino-4- { [(35)- 1 -hydroxyhexan-3 -yl] amino } -6-methylpyrimidin-5 -yl)methyl] -3 - methoxybenzaldehyde (preparation: see step (iii) of Example 7, above; 45 mg) and 3- aminopropanamide hydrochloride (60 mg) were reacted together using the general method described for Example 2, above, to give the title compound (27 mg) as a colourless gum; 1H NMR (CDCI3): 7.15 (br s, IH), 6.87 (d, IH), 6.80-6.74 (m, 2H), 5.73 (br s, IH), 4.72 (br s, 2H), 4.57 (d, IH), 4.11-4.01 (m, IH), 3.87 (s, 3H), 3.72 (s, 2H), 3.64 (s, 2H), 3.39 (ddd, IH), 3.25 (ddd, IH), 2.84 (t, 2H), 2.36 (t, 2H), 2.29 (s, 3H), 1.81-1.70 (m, IH), 1.43- 1.32 (m, IH), 1.26-1.14 (m, 2H), 1.10-0.98 (m, 2H), 0.72 (t, 3H).
Example 12 : ( 3S)-3- [(2- Amino-5- {4- \( azetidin-3-ylamino)methyll -2-methoxybenzyl}- 6-methylpyrimidin-4-yl)aminolh xan-l-ol
Figure imgf000048_0002
4-[(2 -Amino-4- { [(35)- 1 -hydroxyhexan-3 -yl] amino } -6-methylpyrimidin-5 -yl)methyl] -3 - methoxybenzaldehyde (preparation: see step (iii) of Example 7, above; 40 mg) was reacted with tert-butyl 3-aminoazetidine-l-carboxylate using the general method described for steps (i) and (ii) of Example 5, above, to give the title compound (18 mg) as a colourless gum; 1H NMR (CDC13): 6.86 (d, 1H), 6.83 (s, 1H), 6.74 (d, 1H), 4.68 (br s, 2H), 4.56 (d, 1H), 4.11-4.01 (m, 1H), 3.87 (s, 3H), 3.73-3.57 (m, 7H), 3.42-3.36 (m, 4H), 3.26 (ddd, 1H), 2.30 (s, 3H), 1.82-1.71 (m, 1H), 1.43-0.97 (m, 5H), 0.73 (t, 3H).
Example 13: f3S -3-{[2-Amino-5-f2-methoxy-4-{[fl-methylazetidin-3-yl)aminol- methyl}benzyl)-6-methylpyrimidin-4-yllamino}hexan-l-ol
Figure imgf000049_0001
4-[(2-Amino-4- { [(35)- 1 -hydroxyhexan-3 -yl] amino } -6-methylpyrimidin-5 -yl)methyl] -3 - methoxybenzaldehyde (preparation: see step (iii) of Example 7, above) was reacted with tert-butyl 3-aminoazetidine-l-carboxylate using the general method described for step (i) of Example 5, subsequently LiAlH4 using the general method described for Example 6 above to give the title compound (28 mg) as a colourless gum; 1H NMR (CDC13): 6.86 (d, 1H), 6.83 (d, 1H), 6.73 (dd, 1H), 4.65 (br s, 2H), 4.57 (d, 1H), 4.11-3.99 (m, 1H), 3.87 (s, 3H), 3.64 (s, 4H), 3.53-3.49 (m, 2H), 3.42-3.34 (m, 2H), 3.25 (ddd, 1H), 2.71 (dd, 2H), 2.30 (s, 3H), 2.26 (s, 3H), 1.82-1.71 (m, 1H), 1.44-0.97 (m, 5H), 0.73 (t, 3H).
Example 14 : TV2- {4- \( 2- Amino-4- { \( 35)- l-hydroxyhexan-3-yll aminol-6-methyl-
Figure imgf000049_0002
4-[(2 -Amino-4- { [(35)- 1 -hydroxyhexan-3 -yl] amino } -6-methylpyrimidin-5 -yl)methyl] -3 - methoxybenzaldehyde (preparation: see step (iii) of Example 7, above; 45 mg) and N1- methylglycinamide hydrochloride (60 mg) were reacted together using the general method described for Example 2, above, to give the title compound (32 mg) as a colourless gum; 1H NMR (CDC13): 7.58 (d, 1H), 7.04 (s, 1H), 6.85-6.78 (m, 2H), 4.94 (d, 2H), 4.68 (s, 1H), 4.42 (dd, 1H), 4.13-4.03 (m, 1H), 3.89 (s, 3H), 3.65-3.55 (m, 3H), 3.39-3.19 (m, 2H), 3.09 (dd, 1H), 2.25 (s, 3H), 1.94-1.90 (m, 3H), 1.82-1.70 (m, 1H), 1.29-1.18 (m, 2H), 1.14-1.00 (m, 2H), 0.78-0.70 (m, 3H).
Example 15 : ( 3S)-3-( {2-Amino-5- [4-( IH-imidazol- l-ylmethyl)-2-methoxybenzyll -6- methylpyrimidin-4-yl}amino)hexan-l-ol
Figure imgf000050_0001
The title compound may be prepared by the sequence of steps described below:
Figure imgf000050_0002
silylloxyl hexan-3 -yll amino I -6-methylpyrimidin-5 -yl)methyl1-3 -methoxybenzoate
S
Figure imgf000050_0003
fert-Butyldimethylsilylchloride (1.95 g) was added to a solution of methyl 4-[(2-amino-4- { [(35)- 1 -hydroxyhexan-3 -yljamino } -6-methylpyrimidin-5 -yl)methyl] -3 -methoxybenzoate (preparation: see step (i) of Example 7, above; 4 g) in N,N-dimethylformamide (20 mL) and triethylamine (2.1 mL), and the mixture was stirred at RT for 1.5h. tert- Butyldimethylsilylchloride (1.5 g) and triethylamine (0.75 mL) were added again, and the mixture was stirred at RT for 4h. Sat. NaHC03 and water were then added. The mixture was extracted with EtOAc (2 x 50 mL). The combined organic extracts were washed with sat. Na2C03, brine and then dried (Na2S04) and concentrated in vacuo. Purification by FCC, eluting with hexane/EtOAc, gave the sub-title compound (0.65 g) as a yellow oil; 1H NMR: 7.48 (d, 1H), 7.44 (dd, 1H), 6.83 (d, 1H), 5.68 (d, 1H), 5.26 (br s, 1H), 4.26-4.18 (m, 1H), 3.92 (s, 3H), 3.82 (s, 3H), 3.70 (s, 2H), 3.44 (t, 2H), 1.98 (s, 3H), 1.62-1.49 (m, 1H), 1.41-1.22 (m, 2H), 1.19-1.07 (m, 2H), 0.91 (s, 9H), 0.79-0.74 (s, 12H), 0.20 (s, 3H), 0.19 (s, 3H), -0.09 (s, 3H), -0.10 (s, 3H).
(u) 14- IY2- ( I fert-ButvK dimethvOsilyll amino |-4-([(35νΐ-( I fert-butvK dimethvOsilyll - oxyl hexan-3 -yllamino I -6-methylpyrimidin-5 -vQmethyll -3 -methoxyphenyll methanol S
Figure imgf000051_0001
LiBH4 (2M in THF, 0.77 mL) was added to a solution of methyl 4-[(2- {[ieri-butyl- (dimethyl)silyl] amino } -4- { [(35)- 1 - { [fert-butyl(dimethyl)silyl]oxy } hexan-3 -yl] amino } -6- methylpyrimidin-5-yl)methyl]-3-methoxybenzoate (0.65 g) in THF (5 mL) and CH3OH (0.5 mL), and the mixture was stirred at 60°C for 1.5h. Further LiBH4 (2M in THF, 0.39 mL) was then added, and the mixture was stirred under reflux for 3h. After cooling to RT, water and sat. NH4C1 were added. The mixture was extracted with EtOAc (3 x 15 mL). The combined organic extracts were washed with brine, dried (Na2S04) and concentrated in vacuo. Purification by Amino FCC, eluting with hexane/EtOAc, gave the sub-title compound (369 mg) as a colourless gum; 1H NMR (CDC13): 6.92-6.90 (m, 2H), 6.80 (d, 1H), 4.62 (m, 2H), 4.23-4.13 (m, 1H), 3.90 (s, 3H), 4.38 (m, 1H), 3.62 (s, 2H), 3.49-3.34 (m, 2H), 2.27 (s, 3H), 1.73-1.64 (m, 1H), 1.50-1.20(m, 3H), 1.13-1.01 (m, 2H), 0.93 (s, 9H), 0.82 (s, 9H), 0.24 (s, 3H), 0.23 (s, 3H), -0.06 (s, 3H), -0.07 (s, 3H).
(iii) (3S)-3-( (2-Amino-5 - r4-(chloromethyl)-2-methoxybenzyl1 -6-methylpyrimidin-4- yl| amino)hexan- 1 -ol
Figure imgf000051_0002
LiCl (76 mg) and methanesulfonyl chloride (92 μί) were added to a solution of {4-[(2- { [tert-butyl(dimethyl)silyl]amino } -4- { [(35)- 1 - { [fert-butyl(dimethyl)silyl]oxy } hexan-3 - yl]amino}-6-methylpyrimidin-5-yl)methyl]-3-methoxyphenyl}methanol (360 mg) in THF (4 mL), and the mixture was stirred at RT for 4h. Water was added and the mixture was extracted with EtOAc (3 x 15 mL). The combined organic extracts were washed with brine, dried (Na2S04) and concentrated in vacuo to give crude product (433 mg) as colourless gum. This material (371 mg) was dissolved in HC1 (4M in dioxane, 3 mL) and CH3OH (1 mL) and the mixture was stirred at RT for 2h. Sat. NaHC03 was added and the resulting mixture was extracted with EtOAc (3 x 15 mL). The combined organic extracts were washed with brine, dried (Na2S04) and concentrated in vacuo. Purification by Amino FCC, eluting with CHCI3/CH3OH, gave the sub-title compound (153 mg) as a colourless gum; 1H NMR (CDC13): 6.92-6.84 (m, 3H), 4.67 (br s, 2H), 4.53-4.51 (m, 3H), 4.14-4.03 (m, IH), 3.91 (s, 3H), 3.67 (s, 2H), 3.46 (ddd, IH), 3.29 (ddd, IH), 2.31 (s, 3H), 1.84-1.76 (m, IH), 1.45-1.34(m, IH), 1.26-0.95 (m, 4H), 0.73 (t, 3H).
(iv) (361-3 -( {2-Amino-5-r4-(lH-imidazol- 1 -ylmethyl)-2-methoxybenzyl"|-6- methylpyrimidin-4-yl| amino)hexa - 1 -ol
Figure imgf000052_0001
Imidazole (35 mg) and K2C03 (70 mg) were added to a solution of (35)-3-({2-amino-5-[4- (chloromethyl)-2-methoxybenzyl]-6-methylpyrimidin-4-yl}amino)hexan-l-ol (40 mg) in CH3CN (1 mL), and the mixture was stirred at 60°C for 1.5h. The mixture was filtered, and the filtrate was concentrated in vacuo. Purification by Amino FCC, eluting with
CHC13/CH30H, gave the sub-title compound (29 mg) as a colourless gum; 1H NMR (CDCI3): 7.48 (s, IH), 7.03 (s, IH), 6.99 (d, IH), 6.82 (s, IH), 6.67 (dd, IH), 6.55 (d, IH), 5.03 (s, 2H), 4.73 (br s, 2H), 4.46 (d, IH), 4.12-4.02 (m, IH), 3.79 (s, 3H), 3.63 (s, 2H), 3.43 (ddd, IH), 3.27 (ddd, IH), 2.27 (s, 3H), 1.82-1.71 (m, IH), 1.42-1.29 (m, IH), 1.23- 1.08 (m, 2H), 1.04-0.92 (m, 2H), 0.70 (t, 3H). Example 16 : ( 3S -3- { [2-Amino-5-( 2-methoxy-4- { \( 2-methoxyethylH methvDaminol methyl}benzyl)-6-methylpyrimidin-4- llamino}hexan-l-ol
Figure imgf000053_0001
The title compound may be prepared by the sequence of steps described below:
(Ί) Ethyl 2-(4-cvano-2-methoxybenzyl)-3-oxobutanoate
Figure imgf000053_0002
NaH (60% in mineral oil, 4.42 g) was added portion-wise over 10 min to a solution of ethyl acetoacetate (13.48 mL) in THF (60 mL) at 0°C. The resulting suspension was stirred at 0°C for 10 mins. Then, a solution of 4-(bromomethyl)-3-methoxybenzonitrile (20 g) in THF (40 mL) was added portion- wise over 10 min. The mixture was warmed to 70°C and stirred for 16h. The mixture was allowed to cool and then poured cautiously into ice/water (300 mL) and stirred for 30 mins. The aqueous was extracted with EtOAc (3 x 200 mL). The combined organic extracts were dried (MgS04) and concentrated in vacuo.
Purification by FCC, eluting with 20% EtOAc in isohexane gave the sub-title compound (15 g) as a colourless oil; 1H NMR (CDC13): 7.17 (d, 1H), 7.11 (dd, 1H), 6.99 (s, 1H), 4.16-3.99 (m, 2H), 3.81 (d, 3H), 3.09 (ddd, 2H), 2.15 (s, 3H), 1.52 (s, 1H), 1.12 (dt, 3H); LC-MS: m/z 274 APCI-
(ii) 4-r(2-Amino-4-hvdroxy-6-methylpyrimidin-5-yl)methyl1-3-methoxybenzonitrile
Figure imgf000053_0003
Guanidinium carbonate (8.51 g) was added in one portion to a solution of ethyl 2-(4- cyano-2-methoxybenzyl)-3-oxobutanoate (13 g) in CH3OH (200 mL). The resulting mixture was stirred at 65°C for 16h, and then allowed to cool. Water (200 mL) was added and the mixture neutralized by the addition of acetic acid. The precipitate was collected by filtration and the solid washed with EtOH/ water mixture (1 : 1 , 20 mL) and EtOAc (20 mL) to give the sub-title compound (9 g) as a yellow solid; 1H NMR: 10.80 (s, 1H), 7.48-7.24 (m, 2H), 7.01 (d, 1H), 6.37 (s, 2H), 3.89 (s, 3H), 3.60 (s, 2H), 1.94 (s, 3H). LC-MS: m/z 111 APCI+
(iii) 4-r(2-Amino-4-chloro-6-methylpyrimidin-5-yl)methyl1-3-methoxybenzonitrile
Figure imgf000054_0001
4-[(2-Amino-4-hydroxy-6-methylpyrimidin-5 -yl)methyl] -3 -methoxybenzonitrile (2.3 g) was added to POCl3 (20 mL) and the suspension was heated at 100°C for 16h. The POCI3 was removed in vacuo and the residue was diluted with ice/water (20 mL). The pH of the suspension was adjusted to ~7 by the addition of NaHC03 and this mixture was heated at 50°C for lh. The solid was collected by filtration and dried in vacuo to give the sub-title compound (2.2 g) as a white solid; LC-MS: m/z 289 APCI+
(iv) 4- [(2- Amino-4- { \(3S)- 1 -hydroxyhexan-3 -yl] amino I -6-methylpyrimidin-5 -yOmethyl]- 3 -methoxybenzonitrile
Figure imgf000054_0002
(5)-3-Aminohexan-l-ol (804 mg) was combined with 4-[(2-amino-4-chloro-6-methyl- pyrimidin-5-yl)methyl] -3 -methoxybenzonitrile (660 mg) in 1,4-dioxane (5 mL) and the stirred mixture was heated in a CEM Discover™ microwave at 180°C for 9h. The solvents were removed in vacuo. Purification by FCC, eluting with 10% CH3OH (and 0.7M NH3) in CH2CI2 gave the sub-title compound (500 mg) as a gum; LC-MS: m/z 370 APCI+
(v) 4- \(2 -Amino-4- { \(3S)- 1 -hydroxyhexan-3 -yll amino I -6-methylpyrimidin-5 -vQmethyll- 3 -methoxybenzaldehyde
Figure imgf000054_0003
4-[(2-Amino-4- { [(35)- 1 -hydroxyhexan-3 -yl] amino } -6-methylpyrimidin-5 -yl)methyl] -3 - methoxybenzonitrile (500 mg) was dissolved in pyridine (6 mL), acetic acid (3 mL) and water (3 mL), and sodium hypophosphite monohydrate (1004 mg) was added, followed by Raney nickel (238 mg). The reaction was stirred for 16h at 45°C before cooling to RT. The catalyst was removed by filtration and the solids washed with acetic acid (1 mL). The resulting solution was diluted with CH2CI2 (150 mL) and was washed twice with water, dried (MgSC^) and concentrated in vacuo. The residue was azeotroped three times with toluene to give the sub-title compound (400 mg) as a yellow foam; LC-MS: m/z 373 APCI+
(vi) (361-3-1 [2-Amino-5 -(2-methoxy-4- { [(2-methoxyethyl)(methyl)aminolmethyl| - benzyl)-6-methylpyrimidin-4-vH amino I hexan- 1 -ol
Figure imgf000055_0001
A solution of 2-methoxy-N-methylethanamine (131 mg) in CH2CI2 (3 mL) was added to a solution of 4-[(2-amino-4- {[(35)- 1 -hy droxyhexan-3-yl] amino } -6-methylpyrimidin-5- yl)methyl]-3-methoxybenzaldehyde (183 mg) in CH2CI2 (3 mL) and acetic acid (33.8 μί) at RT. Activated molecular sieves (3A) were added. The mixture was cooled to 0°C, sodium triacetoxyborohydride (156 mg) was then added and then the mixture was stirred for 3h. The mixture was then diluted with CH2CI2 and CH3OH added. Purification by FCC eluting with 10% CH3OH in CH2C12 with 1% 7M NH3/CH3OH gave the title compound (105 mg) as a gum; LC-MS: m/z 446 Multimode+.
Example 16 A: ( 3S)-3- { [2- Amino-5-( 2-methoxy-4-{ \( 2-methoxyethylH methvDaminol - methyl}benzyl)-6-methylpyrimidin-4-yllamino}hexan-l-ol bis-saccharin salt,
Figure imgf000055_0002
As bis-saccharin salt (3iS)-3 - { [2- Amino-5 -(2-methoxy-4- { [(2-methoxyethy l)(methy l)amino] -methyl} benzyl)-6- methylpyrimidin-4-yl] amino }hexan-l-ol (the title compound of Example 16, above; 12 mg) was dissolved in CH3CN and saccharin (2 eq.) was added to give a solution. The solvent was removed by blowing N2 over the solution. The solid was then dried in vacuo to give the title compound (21 mg) as a solid; 1H NMR: 7.71 (s, 1H), 7.63 (dd, 7H), 7.35 (d, 1H), 7.21 (s, 1H), 6.98 (d, 1H), 6.90 (s, 1H), 4.35 (s, 2H), 4.25 (s, 1H), 3.88 (s, 3H), 3.75 (s, 2H), 3.64 (s, 2H), 3.22-3.11 (m, 2H), 2.71 (s, 3H), 2.14 (s, 3H), 2.07 (s, 3H), 1.91 (s, 1H), 1.61 (s, 2H), 1.48-1.41 (m, 2H), 1.17-1.07 (m, 2H), 0.80 (t, 3H).
Example 17: ( 3S)-3- \( 2- Amino-5- {2-methoxy-4- \( 4-methylpiperazin- 1-vDmethyll - benzyl}-6-methylpyrimidin-4-yl)aminolhexan-l-ol
Figure imgf000056_0001
4-[(2-Amino-4- { [(35)- 1 -hydroxyhexan-3 -yl] amino } -6-methylpyrimidin-5 -yl)methyl] -3 - methoxybenzaldehyde (preparation: see step (v) of Example 16, above; 100 mg) and 1- methylpiperazine (81 mg) were reacted according to the general method described for step (vi) of Example 16. After stirring for 16h the solvent was removed in vacuo and CH3OH was added. The mixture was purified by RPHPLC to give the title compound (27 mg) as a gum; LC-MS: m/z 457 multimode+
Example 17A: ( 3S)-3- \( 2- Amino-5- {2-methoxy-4- \( 4-methylpiperazin- 1-vDmethyll - benzyl}-6-methylpyrimidin-4-yl)aminolhexan-l-ol bis saccharin salt
Figure imgf000056_0002
as bis saccharin salt (35)-3-[(2-Amino-5-{2-methoxy-4-[(4-methylpiperazin-l-yl)methyl]benzyl}-6-methyl- pyrimidin-4-yl)amino]hexan-l-ol (the title compound of Example 17, above; 27 mg) was dissolved CH3CN and saccharin (2 eq.) was added to give a solution. The solvent was removed by blowing N2 over the solution. The solid was then dried in vacuo to give the title compound (49 mg) as a solid; 1H NMR: 7.68-7.54 (m, 6H), 6.95 (s, 1H), 6.81 (s, 2H), 4.37 (s, 2H), 4.10 (d, 1H), 3.85 (s, 3H), 3.70 (s, 2H), 3.49 (s, 2H), 3.17 (d, 3H), 2.15 (s, 3H), 2.04-1.96 (m, 1H), 1.65-1.55 (m, 2H), 1.43 (d, 2H), 1.24 (s, 2H), 1.11 (dd, 2H), 0.79 (t, 3H).
Example 18 : ( 3S)-3-( {2-Amino-5- [2-methoxy-4-( pyrrolidin- l-ylmethyDbenzyll -6- methylpyrimidin-4-yl}amino)hexan-l-ol
Figure imgf000057_0001
4-[(2-Amino-4- { [(35)- 1 -hydroxyhexan-3 -yl] amino } -6-methylpyrimidin-5 -yl)methyl] -3 - methoxybenzaldehyde (preparation: see step (v) of Example 16, above; 100 mg) and pyrrolidine (57.3 mg) were reacted according to the general method described for step (vi) of Example 16 to give the title compound (23.7 mg) as a gum; LC-MS: m/z 428 multimode+
Example 18A: f3S -3-f{2-Amino-5-[2-methoxy-4-fpyrrolidin-l-ylmethyl)benzyll-6- methylpyrimidin-4-yl}amino)hexan-l-ol bis saccharin salt
Figure imgf000057_0002
as bis saccharin salt
(3iS)-3 -( {2- Amino-5 -[2-methoxy-4-(pyrrolidin- 1 -ylmethyl)benzyl] -6-methylpyrimidin-4- yl}amino)hexan-l-ol (the title compound of Example 18, above; 23.7 mg) was dissolved in CH3CN and saccharin (2 eq.) was added to give a solution. The solvent was removed by blowing N2 over the solution. The solid was then dried in vacuo to give the title compound (44 mg) as a solid; 1H NMR: 7.69-7.63 (m, 2H), 7.62-7.56 (m, 6H), 7.34 (s, 1H), 7.19 (d, 1H), 6.98 (d, 1H), 6.93-6.87 (m, 1H), 4.41-4.27 (m, 3H), 4.05-4.00 (m, 1H), 3.91-3.85 (m, 3H), 3.75 (d, 2H), 3.08 (s, 2H), 2.13 (dd, 3H), 2.05-1.96 (m, 3H), 1.89 (d, 2H), 1.67-1.59 (m, 2H), 1.48-1.41 (m, 2H), 1.26-1.15 (m, 2H), 1.13 (ddd, 2H), 0.83-0.77 (m, 3H).
Example 19 : (35V3- { r2-Amino-5-(4- { r -hvdroxyethyl)(methyl)aminol methyl}-2- methoxybenzyl)-6-methylpyrimidin-4-yllamino}hexan-l-ol
Figure imgf000058_0001
The title compound may be prepared by the sequence of steps described below:
(Ί) 4- [(2- Amino-4- { \(3S)- 1 -hydroxyhexan-3 -yl] amino I -6-methylpyrimidin-5 -yPmethyl] -3 - methoxybenzaldehyde
Figure imgf000058_0002
4-[(2 -Amino-4- { [(35)- 1 -hydroxyhexan-3 -yl] amino } -6-methylpyrimidin-5 -yl)methyl] -3 - methoxybenzonitrile (preparation: see step (iv) of Example 16, above; 375 mg) was reacted according to the general method described for step (v) of Example 16, stirring for 3h, to give the sub-title compound (350 mg) as a yellow foam; LC-MS: m/z 373 APCI+ (ii) (361-3- (r2-Amino-5-(2-methoxy-4- {r(2-methoxyethyl)(methyl)amino1methyl|benzyl)- 6-methylpyrimidin-4-yllamino I hexan- 1 -ol
Figure imgf000058_0003
4-[(2 -Amino-4- { [(35)- 1 -hydroxyhexan-3 -yl] amino } -6-methylpyrimidin-5 -yl)methyl] -3 - methoxybenzaldehyde (120 mg) was reacted 2-methoxy-N-methylethanamine using the general method described in step (vi) of Example 16. After stirring for 16h the solvent was removed in vacuo and CH3OH was added. Purification by RPHPLC gave the sub-title compound (100 mg) as a gum; LC-MS: m/z 446 APCI+
(iii) (3S)-3- i \2- Amino-5 -(4- { r(2-hvdroxyethyl)(methyl)amino1methyl| -2-methoxybenzvD- 6-methylpyrimidin-4-vHamino I hexan- 1 -ol
Figure imgf000059_0001
(35)-3-{[2-Amino-5-(2-methoxy-4- {[(2-methoxyethyl)(methyl)amino]methyl}benzyl)-6- methylpyrimidin-4-yl]amino}hexan-l-ol (50 mg) in CH2CI2 (5 mL) was cooled to -70°C and BBr3 (1 12 μί) was carefully added. The mixture was allowed to warm to RT, then the mixture was cooled to -70°C and more BBr3 (1 12 μΐ,) was added. The mixture was stirred for 30 mins at RT. CH3OH (2 mL) was added the mixture was stirred for 10 mins then HCl (4M in dioxane, 28 μί) was added and the solvent was removed in vacuo. The residue was dissolved in CH3OH and the pH adjusted to 7 by the addition of NH3. Purification was by RPHPLC to afford a gum. This was re-purified by RPHPLC eluting with 0.1%
trifluoroacetic acid in CH3CN. This material was passed through a macroporous carbonate resin cartridge to give the title compound (1 1.8 mg) as a gum; LC-MS: m/z 432
multimode+
Example 19 A: (35V3- { Γ2- Amino-5-(4- { r -hvdroxyethyl)(methyl)aminol methyl}-2- methoxybenzyl)-6-methylpyrimidin-4-yllamino}hexan-l-ol bis saccharin salt
Figure imgf000059_0002
as bis saccharin salt
(3iS)-3 - { [2- Amino-5 -(4- { [(2-hydroxyethyl)(methyl)amino]methyl} -2-methoxybenzyl)-6- methylpyrimidin-4-yl] amino} hexan- l-ol (the title compound of Example 19, above; 1 1.8 mg) was dissolved in CH3CN and saccharin (2 eq.) was added to give a solution. The solvent was removed by blowing N2 over the solution. The solid was then dried in vacuo to give the title compound (18 mg) as a solid; 1H NMR: 1 1.94 (s, 1H), 9.43 (s, 1H), 7.68-7.56 (m, 8H), 7.35 (d, 2H), 7.20 (s, 1H), 6.98 (d, 1H), 6.91 (d, 1H), 5.35 (d, 1H), 4.41-4.17 (m, 4H), 3.88 (s, 3H), 3.74 (s, 4H), 3.08 (d, 2H), 2.71 (s, 3H), 2.14 (s, 3H), 2.08 (s, 1H), 1.68- 1.54 (m, 2H), 1.44 (dd, 2H), 1.24 (s, 1H), 1.19-1.05 (m, 2H), 0.80 (t, 3H).
Example 20: f2S -2-f{2-Amino-5-[2-methoxy-4-fpyrrolidin-l-ylmethyl)benzyll-6- methylpyrimidin-4-yl}amino)pentan-l-ol
Figure imgf000060_0001
Pyrrolidine (280 μί) was reacted with 4-[(2-amino-4-{[(25)-l-hydroxypentan-2-yl]- amino}-6-methylpyrimidin-5-yl)methyl]-3-methoxybenzaldehyde according to the general method described in step (xii) of Example 1 to give the title compound (17 mg) as a colourless gum; 1H NMR: 6.90 (s, 1H), 6.75 (d, 2H), 5.64 (s, 2H), 5.40 (d, 1H), 4.59 (t, 1H), 4.11-4.04 (m, 1H), 3.84 (s, 3H), 3.59 (s, 2H), 3.52 (d, 2H), 3.28-3.22 (m, 1H), 2.41- 2.36 (m, 4H), 2.06 (s, 3H), 1.51-1.42 (m, 1H), 1.31-1.19 (m, 2H), 1.11-0.96 (m, 2H), 0.75 (t, 3H); LC-MS: m/z 414 multimode+
Example 21 : ( 2S)-2- \( 2- Amino-5- {2-methoxy-4- \( 4-methylpiperazin- 1-vDmethyll - benzyl}-6-methylpyrimidin-4-yl)aminolpentan-l-ol
Figure imgf000060_0002
1-Methylpiperazine (112 mg) was reacted with 4-[(2-amino-4-{[(25)-l-hydroxypentan-2- yl]amino}-6-methylpyrimidin-5-yl)methyl]-3-methoxybenzaldehyde according to the general method described in step (xii) of Example 1 to give the title compound (23 mg) as a colourless gum; 1H NMR: 6.89 (s, 1H), 6.78-6.72 (m, 2H), 5.64 (s, 2H), 5.39 (d, 1H), 4.59 (t, 1H), 4.13-4.03 (m, 1H), 3.83 (s, 3H), 3.58 (d, 2H), 3.38 (d, 2H), 3.24 (dt, 1H), 2.34 (d, 5H), 2.13 (s, 3H), 2.06 (s, 3H), 1.46 (dt, 1H), 1.29-1.20 (m, 2H), 1.12-0.97 (m, 3H), 0.74 (t, 3H); LC-MS: m/z 443 multimode+ Example 22 : ( 3S)-3- \( 2- Amino-5- {4- \( diethylamino)methyll -2-methoxybenzyl}-6- methylpyrimidin-4-yl)aminolhexan-l-ol
Figure imgf000061_0001
The title compound may be prepared by the steps described below:
(Ί) 2-Amino-5-(4-cyano-2-methoxybenzyl)-6-methylpyrimidin-4-yl 2,4,6-tri(propan-2-yl)- benzenesulfonate
Figure imgf000061_0002
4-[(2-Amino-4-hydroxy-6-methylpyrimidin-5-yl)methyl]-3-methoxybenzonitrile
(preparation: see step (ii) of Example 16, above; 9.6 g) was mixed with 2,4,6-triisopropyl- benzene-l-sulfonyl chloride (13.98 g) and l,4-diazabicyclo[2.2.2]octane (5.18 g) in THF (250 mL) and this mixture was heated at 30°C for 7h. The mixture was then partitioned between EtOAc and sat. brine. The combined organic solutions were dried (MgSC^), and concentrated in vacuo. The residue was triturated with isohexane (30 mL) and diethyl ether (1 mL) to give the sub-title compound (17 g) as a white solid; LC-MS: m/z 537 APCI+.
(ii) 4- \(2- Amino-4- { \(3S)- 1 -hydroxyhexan-3 -yl] amino I -6-methylpyrimidin-5 -yDmethyl] - 3 -methoxybenzonitrile
Figure imgf000061_0003
2-Amino-5-(4-cyano-2-methoxybenzyl)-6-methylpyrimidin-4-yl 2,4,6-tri(propan-2-yl)- benzenesulfonate (3.00 g, 5.59 mmol), (5)-3-aminohexan-l-ol (1.965 g, 16.77 mmol) and TFA (419 μί, 5.59 mmol) were mixed together in butyronitrile (30 mL) and the mixture was heated at 120°C for 30h. The solvent was then removed in vacuo. The residue was diluted with EtOAc and washed with 2M NaOH. The organic solution was dried (Na2S04), and concentrated in vacuo. The crude material was dissolved in CH3OH (25 mL), acidified with acetic acid (1 mL) and loaded on to a 50g SCX cartridge. The impurities were washed through with CH3OH (125 mL) and discarded. The product was eluted with 1M
NH3/CH3OH (250 mL) and concentrated in vacuo. The product was purified by FCC, eluting with 10% CH3OH in EtOAc to give the subtitle compound (1 g) as a yellow gum; LC-MS: m/z 370 APCI+.
(iii) 4- [(2- Amino-4- { \(3S)- 1 -hydroxyhexan-3 -yl] amino I -6-methylpyrimidin-5 -yPmethyl] - 3 -methoxybenzaldehyde
Figure imgf000062_0001
4-[(2 -Amino-4- { [(35)- 1 -hydroxyhexan-3 -yl] amino } -6-methylpyrimidin-5 -yl)methyl] -3 - methoxybenzonitrile (850 mg) was reacted using the general method described in step (v) of Example 16 to give the sub-title compound (860 mg) as a yellow foam; LC-MS: m/z 373 APCI+.
(iv) (36)-3-[(2-Amino-5- (4-[(diethylamino)methyll-2-methoxybenzyl| -6- methylpyrimidin-4-yl)amino"|hexan- 1 - l
Figure imgf000062_0002
4-[(2 -Amino-4- { [(35)- 1 -hydroxyhexan-3 -yl] amino } -6-methylpyrimidin-5 -yl)methyl] -3 - methoxybenzaldehyde (310 mg) and diethylamine (183 mg) were reacted together using the general method described in step (vi) of Example 16. After stirring for 16h the solvent was removed in vacuo and CH3OH was added. The solvent was removed in vacuo and the crude material was dissolved in CH3OH (10 mL), acidified with acetic acid (0.2 mL) and loaded on to a lOg SCX cartridge. The impurities were washed through with CH3OH (25 mL) and discarded. The product was eluted with 1M NH3/CH3OH (50 mL) and
concentrated in vacuo (50 mL) to give the sub-title compound (200 mg) as a gum; LC-MS: m/z 430 APCI+.
Example 23 : ( 3S)-3- { [2- Amino-5-( 4- { lethvKmethvDaminol methyll-2-methoxybenzyl)-
6-methylpyrimidin-4-yllamino}hexan-l-ol
Figure imgf000063_0001
The title compound may be prepared by the steps described below:
4-[(2 -Amino-4- { [(35)- 1 -hydroxyhexan-3 -yl] amino } -6-methylpyrimidin-5 -yl)methyl] -3 - methoxybenzaldehyde (preparation: see step (iii) of Example 22; 310 mg) and N- methylethan-amine (148 mg) were reacted together using the general method described in step (iv) of Example 6 to give the sub-title compound (160 mg) as a gum; LC-MS: m/z 416
APCI+.
Example 24: f3S -3-[f2-Amino-5-{4-[fdimethylamino)methyll-2-methoxybenzyl}-6- methylpyrimidin-4-yl)aminolhexan-l-ol
Figure imgf000063_0002
The title compound may be prepared by the steps described below:
(i) 4- [(2- Amino-4- { \(3S)- 1 -hydroxyhexan-3 -yl] amino I -6-methylpyrimidin-5 -yOmethyl] -3 - methoxybenzonitrile
Figure imgf000064_0001
(S)-3-Aminohexan-l-ol (264 mg, 1.3 eq.) was reacted using the general method described in step (iv) of Example 16, with the addition of triethylamine (0.24 mL, 1 eq.) and heating for 3h. Purification by FCC eluting with 0-20% CH3OH (and 0.7M NH3) in CH2C12 gave the sub-title compound (260 mg) as an oil; LC-MS: m/z 370 APCI+.
(ii) 4- IY2- Amino-4- { \(3S)- 1 -hydroxyhexan-3 -yll amino I -6-methylpyrimidin-5 -vDmethyll - 3 -methoxybenzaldehyde
Figure imgf000064_0002
4-[(2 -Amino-4- { [(35)- 1 -hydroxyhexan-3 -yl] amino } -6-methylpyrimidin-5 -yl)methyl] -3 - methoxybenzonitrile (375 mg) was reacted using the general method described in step (v) of Example 16, with stirring for 3h, to give the sub-title compound (350 mg) as a yellow foam; LC-MS: m/z 373 APCI+.
(iii) (36)-3-[(2-Amino-5- (4-[(dimethylamino)methyll-2-methoxybenzyl| -6- methylpyrimidin-4-vDaminolhexan- 1 -ol
Figure imgf000064_0003
4-[(2 -Amino-4- { [(35)- 1 -hydroxyhexan-3 -yl] amino } -6-methylpyrimidin-5 -yl)methyl] -3 - methoxybenzaldehyde (120 mg) and dimethylamine (483 μί) were reacted together using the general method described in step (vi) of Example 16. After stirring for 16h the solvent was removed in vacuo and CH3OH was added. Purification by RPHPLC gave a gum (35 mg). Human TLR7 assay
Recombinant human TLR7 was stably expressed in a HEK293 cell line already stably expressing the pNiFty2-SEAP reporter plasmid; integration of the reporter gene was maintained by selection with the antibiotic zeocin. The most common variant sequence of human TLR7 (represented by the EMBL sequence AF240467) was cloned into the mammalian cell expression vector pUNO and transfected into this reporter cell-line.
Transfectants with stable expression were selected using the antibiotic blasticidin. In this reporter cell-line, expression of secreted alkaline phosphatase (SEAP) is controlled by an NFkB/ELAM-1 composite promoter comprising five NFkB sites combined with the proximal ELAM-1 promoter. TLR signaling leads to the translocation of NFkB and activation of the promoter results in expression of the SEAP gene. TLR7-specific activation was assessed by determining the level of SEAP produced following overnight incubation of the cells at 37°C with the standard compound in the presence of 0.1% (v/v) dimethylsulfoxide (DMSO). Concentration dependent induction of SEAP production by compounds was expressed as the concentration of compound which produced half of the maximal level of SEAP induction for that compound (EC50). TLR7 activity for compounds of the present invention was assessed using the human TLR7 assay and the results are shown in Table 1 below wherein the degree of TLR7 activation for each compound is expressed as a pEC5o value.
Ex. No. TLR7 (pEC50) Ex. No. TLR7 (pEC50)
1 8.64 13 7.72
1A 8.06 14 7.26
2 7.20 15 8.56
3 7.92 16A >8.71
4 7.02 17A >8.71
5 6.42 18A >8.71
6 6.81 19A >8.3
7 8.52 20 >8.23
8 8.23 21 >8.2
9 8.01
10 7.35
11 7.09
12 7.71
Table 1
TLR8 assay - Method 1
HEK293-hTLR8 cells, stably transfected with human TLR8 (pUNO expression vector) and pNiFty2-SEAP (Secretory Alkaline Phosphatase) reporter plasmid were maintained in DMEM, FCS 10% (v/v), 2 mM /-glutamine, non-essential amino acids, 10 μg/mL blasticidin S and 10 μg/mL zeocin. The human TLR8 sequence used was identical to GenBank sequence AAZ95441.1. Cells were seeded in tissue culture treated clear flat bottom polystyrene 96 well plates at 2>< 104 cells/well. Dose response curves were generated by addition of test compounds and incubated for 20h at 37°C in an atmosphere of 5% C02. The SEAP released was quantified using /?-nitrophenyl phosphate as a substrate and the absorbance at 405 nm was determined by a microplate reader.
Human TLR8 assay - Method 2
TLR8/NF-kB/SEAPorter™ HEK 293 Cell Line (Imgenex Corporation) is a stably co- transfected cell line which expresses full-length human TLR8 and the secreted alkaline phosphatase (SEAP) reporter gene under the transcriptional control of an NF-κΒ response element. TLR8 expression in this cell line has been tested by flow cytometry. Transfectants with stable expression were selected using the antibiotic blasticidin and geneticin. TLR signaling leads to the translocation of NF-κΒ and activation of the promoter results in expression of the SEAP gene. TLR8-specific activation was assessed by determining the level of SEAP produced following overnight incubation of the cells at 37°C with the standard compound in the presence of 0.1% (v/v) dimethylsulfoxide (DMSO).
Concentration dependent induction of SEAP production by compounds was expressed as the concentration of compound which produced half of the maximal level of SEAP induction for that compound (EC50). TLR8 activity for compounds of the present invention was assessed using the human TLR8 assay and the results are shown in Table 2 below wherein the degree of TLR8 activation for each compound is expressed as a pEC50 value.
Table 2
Figure imgf000067_0001
hERG Analysis - Method 1
Cell culture
The hERG-expressing Chinese hamster ovary Kl (CHO) cells described by (Persson, Carlsson, Duker, & Jacobson, 2005) are grown to semi-confluence at 37°C in a humidified environment (5% C02) in F-12 Ham medium containing L-glutamine, 10% foetal calf serum (FCS) and 0.6mg/mL hygromycin (all available from Sigma- Aldrich). Prior to use, the monolayer is washed using a pre-warmed (37°C) 3mL aliquot of Versene 1 :5,000 (Invitrogen). After aspiration of this solution the flask is incubated at 37°C in an incubator with a further 2mL of Versene 1 :5,000 for a period of 6 minutes. Cells are then detached from the bottom of the flask by gentle tapping and lOmL of Dulbecco's
Phosphate-Buffered Saline containing calcium (0.9mM) and magnesium (0.5mM) (PBS; Invitrogen) is then added to the flask and aspirated into a 15mL centrifuge tube prior to centrifugation (50g, for 4 mins). The resulting supernatant is discarded and the pellet gently re-suspended in 3mL of PBS. A 0.5mL aliquot of cell suspension is removed and the number of viable cells (based on trypan blue exclusion) is determined in an automated reader (Cedex; Innovatis) so that the cell re-suspension volume can be adjusted with PBS to give the desired final cell concentration. It is the cell concentration at this point in the assay that is quoted when referring to this parameter. CHO-Kvl .5 cells, which are used to adjust the voltage offset on Ion Works™ HT, are maintained and prepared for use in the same way.
Electrophysiology
The principles and operation of this device have been described by (Schroeder, Neagle, Trezise, & Worley, 2003). Briefly, the technology is based on a 384-well plate (PatchPlate) in which a recording is attempted in each well by using suction to position and hold a cell on a small hole separating two isolated fluid chambers. Once sealing has taken place, the solution on the underside of the PatchPlate is changed to one containing amphotericin B. This permeablises the patch of cell membrane covering the hole in each well and, in effect, allows a perforated, whole-cell patch clamp recording to be made.
A β-test Ion Works™ HT from Essen Instrument was used. There is no capability to warm solutions in this device hence it is operated at ~r.t. (~21°C), as follows. The reservoir in the "Buffer" position is loaded with 4 mL of PBS and that in the "Cells" position with the CHO-hERG cell suspension described above. A 96-well plate (V-bottom, Greiner Bio- one) containing the compounds to be tested (at 3 -fold above their final test concentration) is placed in the "Plate 1" position and a PatchPlate™ is clamped into the PatchPlate™ station. Each compound plate is laid-out in 12 columns to enable ten, 8-point
concentration-effect curves to be constructed; the remaining two columns on the plate are taken up with vehicle (final concentration 0.33% DMSO), to define the assay baseline, and a supra-maximal blocking concentration of cisapride (final concentration 10 μΜ) to define the 100% inhibition level. The fluidics-head (F-Head) of IonWorks™ HT then adds 3.5μΙ. of PBS to each well of the PatchPlate™ and its underside is perfused with "internal" solution that had the following composition (in mM): K-Gluconate (100 parts), KC1 (40 parts), MgCl2 (3.2 parts), EGTA (3 parts) and HEPES (5 parts, pH 7.25-7.30 using 10M KOH). After priming and de-bubbling, the electronics-head (E-head) then moves round the PatchPlate™ performing a hole test (i.e. applying a voltage pulse to determine whether the hole in each well is open). The F-head then dispenses 3.5μΕ of the cell suspension described above into each well of the PatchPlate™ and the cells are given 200 seconds to reach and seal to the hole in each well. Following this, the E-head moves round the PatchPlate™ to determine the seal resistance obtained in each well. Next, the solution on the underside of the PatchPlate™ is changed to "access" solution that has the following composition (in mM): KC1 (140 parts), EGTA (1 part), MgCl2 (1 part) and HEPES (20 parts, pH 7.25-7.30 using 10M KOH) plus 100μg/mL of amphotericin B (Sigma- Aldrich). After allowing 9 minutes for patch perforation to take place, the E-head moves round the PatchPlate™ 48 wells at a time to obtain pre-compound hERG current measurements. The F-head then adds 3.5μΕ of solution from each well of the compound plate to 4 wells on the PatchPlate™ (the final DMSO concentration is 0.33% in every well). This is achieved by moving from the most dilute to the most concentrated well of the compound plate to minimise the impact of any compound carry-over. After approximately 3.5 mins incubation, the E-head then moves around all 384-wells of the PatchPlate™ to obtain post- compound hERG current measurements. In this way, non-cumulative concentration-effect curves can be produced where, providing the acceptance criteria are achieved in a sufficient percentage of wells (see below), the effect of each concentration of test compound is based on recording from between 1 and 4 cells.
The pre- and post-compound hERG current is evoked by a single voltage pulse consisting of a 20 second period holding at -70mV, a 160millisecond step to -60mV (to obtain an estimate of leak), a lOOmillisecond step back to -70mV, a 1 second step to + 40mV, a 2 second step to -30mV and finally a 500 millisecond step to -70mV. In between the pre- and post-compound voltage pulses there is no clamping of the membrane potential. Currents are leak- subtracted based on the estimate of current evoked during the +10mV step at the start of the voltage pulse protocol. Any voltage offsets in Ion Works™ HT were adjusted in one of two ways. When determining compound potency, a depolarising voltage ramp is applied to CHO-Kvl .5 cells and the voltage noted at which there was an inflection point in the current trace (i.e. the point at which channel activation is seen with a ramp protocol). The voltage at which this occurred has previously been determined using the same voltage command in conventional electrophysiology and found to be -15mV (data not shown); thus an offset potential could be entered into the Ion Works™ HT software using this value as a reference point. When determining the basic electrophysiological properties of hERG, any offset is adjusted by determining the hERG tail current reversal potential in Ion Works™ HT, comparing it with that found in conventional
electrophysiology (-82mV) and then making the necessary offset adjustment in the Ion Works™ HT software. The current signal is sampled at 2.5kHz.
Pre- and post-scan hERG current magnitude was measured automatically from the leak subtracted traces by the Ion Works™ HT software by taking a 40ms average of the current during the initial holding period at -70 mV (baseline current) and subtracting this from the peak of the tail current response. The acceptance criteria for the currents evoked in each well are: pre-scan seal resistance >60ΜΩ, pre-scan hERG tail current amplitude >150pA; post-scan seal resistance >60ΜΩ. The degree of inhibition of the hERG current can be assessed by dividing the post-scan hERG current by the respective pre-scan hERG current for each well. References: Persson, F. et al, J Cardiovasc.ElectrophysioL, 16, 329-341 (2005), and Schroeder, K., et al, J Biomol Screen., 8, 50-64, (2003). hERG analysis - Method 2
The hERG potassium current is measured in a hERG-stably-expressing Chinese hamster ovary Kl (CHO) cells. The experiments were performed using an automated planar patch-clamp system QPatch HT (Sophion Bioscience A/S). The application of pressure for forming gigaseals and whole-cell patch clamp configuration were established using the QPatch assay software. Patch-clamp experiments were performed in voltage-clamp mode and whole-cell currents were recorded from individual cells. The following stimulation protocol was applied to investigate the effects of compounds on hERG potassium channel. : The membrane potential was held at -80 mV and repetitively (every 15 s) depolarized to +20 mV for 5 s after the pulse to -50 mV for 20 ms served to define the baseline, followed by repolarizing step to -50 mV for 5 s to evaluate of the tail current amplitude. Experiments were conducted at room temperature (22±2°C).
Effects of compounds were determined from cumulative applications of increasing 4 concentrations and calculated as percent of blocked current. The data points were fitted with Hill equation to calculate half-maximal inhibition concentrations.
The test solution includes:
Extracellular solution (mM): 2mM of CaCl2, lmM of MgCl2, lOmM of HEPES, 4mM of KC1, 145 mM of NaCl, and lOmM of Glucose; and
Intracellular solution (mM): 5.4mM of CaCl2, 1.8mM of MgCl2, lOmM of HEPES, 31mM of KOH, lOmM of EGTA, 120mM of KC1, and 4mM of ATP.
The results of the hERG analysis are shown in Table 3 :
Figure imgf000071_0001
Table 3

Claims

1. A compound of Formula (I)
Figure imgf000072_0001
wherein:
n is 1 or 2;
R1 is Ci_4alkyl, methoxyC2_3alkyl, hydroxyC2_4alkyl, -(CH2)yCONH2 wherein y is 1 or 2, or azetidin-3-yl, wherein said azetidin-3-yl is optionally substituted on nitrogen by Ci_3alkyl; and
R2 is hydrogen or Ci_4alkyl;
or R1 and R2 together with the nitrogen to which they are attached form a pyrrolidin-l-yl, lH-imidazol-l-yl or a piperazin-l-yl; wherein said piperazin-l-yl is optionally substituted on the available nitrogen by Ci_3alkyl;
or a pharmaceutically acceptable salt thereof.
2. A compound of Formula (I) of Formula (IA):
Figure imgf000072_0002
or a pharmaceutically acceptable salt thereof, wherein the values of R1, R2 and n are as claimed in claim 1.
3. A compound according to claim 1 or claim 2, or a pharmaceutically acceptable salt thereof, wherein n is 1.
4. A compound according to claim 1 or claim 2, or a pharmaceutically acceptable salt thereof, wherein n is 2.
5. A compound according to claim 1, selected from:
(25)-2-[(2-amino-5-{4-[(dimethylamino)methyl]-2-methoxybenzyl}-6- methylpyrimidin-4-yl)amino]pentan- 1 -ol;
(2iS)-2-( {2-amino-5-[2-methoxy-4-(piperazin- 1 -ylmethyl)benzyl]-6- methylpyrimidin-4-yl} amino)pentan- 1 -ol;
(25)-2-[(2-amino-5-{2-methoxy-4-[(methylamino)methyl]benzyl}-6- methylpyrimidin-4-yl)amino]pentan- 1 -ol;
(25)-2-{[2-amino-5-(4-{[(2-hydroxyethyl)amino]methyl}-2-methoxy-benzyl)-6- methylpyrimidin-4-yl]amino}pentan- 1 -ol;
(25)-2-[(2-amino-5-{4-[(azetidin-3-ylamino)methyl]-2-methoxybenzyl}-6- methylpyrimidin-4-yl)amino]pentan- 1 -ol;
(2S)-2- { [2-amino-5-(2-methoxy-4- { [( 1 -methylazetidin-3-yl)amino]methyl} - benzyl)-6-methylpyrimidin-4-yl] amino } pentan- 1 -ol;
(35)-3-[(2-amino-5-{2-methoxy-4-[(methylamino)methyl]benzyl}-6- methylpyrimidin-4-yl)amino]hexan- 1 -ol;
(3iS)-3-( {2-amino-5-[2-methoxy-4-(piperazin- 1 -ylmethyl)benzyl]-6- methylpyrimidin-4-yl} amino)hexan- 1 -ol;
(35)-3-{[2-amino-5-(4-{[(2-hydroxyethyl)amino]methyl}-2-methoxy-benzyl)-6- methylpyrimidin-4-yl]amino } hexan- 1 -ol;
N2- {4-[(2-amino-4- {[(35)- 1 -hydroxyhexan-3-yl]amino} -6-methyl-pyrimidin-5- yl)methyl]-3-methoxybenzyl}glycinamide;
N3- {4-[(2-amino-4- {[(35)- 1 -hydroxyhexan-3-yl]amino} -6-methyl-pyrimidin-5- yl)methyl]-3-methoxybenzyl}-P-alaninamide;
(35)-3-[(2-amino-5-{4-[(azetidin-3-ylamino)methyl]-2-methoxybenzyl}-6- methylpyrimidin-4-yl)amino]hexan- 1 -ol; (3iS)-3- { [2-amino-5 -(2-methoxy-4- { [( 1 -methylazetidin-3 -yl)amino] - methyl} benzyl)-6-methylpyrimidin-4-yl] amino } hexan- 1 -ol;
N2- {4-[(2-amino-4- {[(35)- 1 -hydroxyhexan-3-yl]amino} -6-methyl-pyrimidin-5- yl)methyl]-3-methoxybenzyl}-N2-methylglycinamide;
(3iS)-3-( {2-amino-5-[4-(lH-imidazol- 1 -ylmethyl)-2-methoxybenzyl]-6- methylpyrimidin-4-yl} amino)hexan- 1 -ol;
(35)-3-{[2-amino-5-(2-methoxy-4-{[(2-methoxyethyl)(methyl)amino]- methyl} benzyl)-6-methylpyrimidin-4-yl] amino } hexan- 1 -ol;
(35)-3-[(2-amino-5-{2-methoxy-4-[(4-methylpiperazin-l-yl)methyl]-benzyl}-6- methylpyrimidin-4-yl)amino]hexan- 1 -ol;
(35)-3-( {2-amino-5-[2-methoxy-4-(pyrrolidin- 1 -ylmethyl)benzyl]-6- methylpyrimidin-4-yl} amino)hexan- 1 -ol;
(3iS)-3 - { [2-amino-5 -(4- { [(2-hydroxyethyl)(methyl)amino]methyl} -2- methoxybenzyl)-6-methy lpyrimidin-4-yl] amino } hexan- 1 -ol;
(2iS)-2-( {2-amino-5-[2-methoxy-4-(pyrrolidin- 1 -ylmethyl)benzyl]-6- methylpyrimidin-4-yl} amino)pentan- 1 -ol;
(25)-2-[(2-amino-5-{2-methoxy-4-[(4-methylpiperazin-l-yl)methyl]-benzyl}-6- methylpyrimidin-4-yl)amino]pentan- 1 -ol;
(35)-3-[(2-amino-5-{4-[(diethylamino)methyl]-2-methoxybenzyl}-6- methylpyrimidin-4-yl)amino]hexan- 1 -ol;
(35)-3-{[2-amino-5-(4-{[ethyl(methyl)amino]methyl}-2-methoxybenzyl)-6- methylpyrimidin-4-yl]amino } hexan- 1 -ol; and
(35)-3-[(2-amino-5-{4-[(dimethylamino)methyl]-2-methoxybenzyl}-6- methylpyrimidin-4-yl)amino]hexan- 1 -ol;
or a pharmaceutically acceptable salt thereof.
6. A pharmaceutical composition which comprises a compound according to any one of claims 1 to 5, or a pharmaceutically acceptable salt thereof, in association with a pharmaceutically acceptable diluent or carrier.
7. A compound according to any one of claims 1 to 5, or a pharmaceutically
acceptable salt thereof, for use as a medicament. A compound according to any one of claims 1 to 5, or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer.
Use of a compound according to any one of claims 1 to 5, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for use in the treatment of cancer.
A method of treating cancer in a warm-blooded animal, such as man, in need of such treatment which comprises administering to said animal an effective amount of a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined in any one of claims 1 to 5.
PCT/GB2011/052246 2010-11-19 2011-11-17 Benzylamine compounds as toll -like receptor 7 agonists WO2012066336A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US41569510P 2010-11-19 2010-11-19
US61/415,695 2010-11-19

Publications (1)

Publication Number Publication Date
WO2012066336A1 true WO2012066336A1 (en) 2012-05-24

Family

ID=45099137

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2011/052246 WO2012066336A1 (en) 2010-11-19 2011-11-17 Benzylamine compounds as toll -like receptor 7 agonists

Country Status (1)

Country Link
WO (1) WO2012066336A1 (en)

Cited By (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013172479A1 (en) * 2012-05-18 2013-11-21 Dainippon Sumitomo Pharma Co., Ltd. Carboxylic acid compounds
US9045472B2 (en) 2010-12-16 2015-06-02 Astrazeneca Ab Imidazoquinoline compounds
US9334268B2 (en) 2014-04-22 2016-05-10 Hoffman-La Roche Inc. 4-amino-imidazoquinoline compounds
US9475775B2 (en) 2015-03-06 2016-10-25 Hoffmann-La Roche Inc. Benzazepine dicarboxamide compounds
WO2017046112A1 (en) 2015-09-17 2017-03-23 F. Hoffmann-La Roche Ag Sulfinylphenyl or sulfonimidoylphenyl benzazepines
WO2017061532A1 (en) * 2015-10-07 2017-04-13 大日本住友製薬株式会社 Pyrimidine compound
WO2017202703A1 (en) 2016-05-23 2017-11-30 F. Hoffmann-La Roche Ag Benzazepine dicarboxamide compounds with secondary amide function
WO2017216054A1 (en) 2016-06-12 2017-12-21 F. Hoffmann-La Roche Ag Dihydropyrimidinyl benzazepine carboxamide compounds
WO2018106606A1 (en) * 2016-12-05 2018-06-14 Apros Therapeutics, Inc. Pyrimidine compounds containing acidic groups
WO2018181420A1 (en) 2017-03-29 2018-10-04 大日本住友製薬株式会社 Vaccine adjuvant formulation
WO2019066571A2 (en) 2017-09-28 2019-04-04 연세대학교 산학협력단 Method for producing myeloid-derived suppressor cells, myeloid-derived suppressor cells produced thereby, and uses thereof
TWI666212B (en) * 2014-05-01 2019-07-21 瑞士商諾華公司 Compounds and compositions as toll-like receptor 7 agonists
US10370338B2 (en) 2016-05-23 2019-08-06 Hoffmann-La Roche Inc. Benzazepine dicarboxamide compounds with tertiary amide function
US10407431B2 (en) 2014-05-01 2019-09-10 Novartis Ag Compounds and compositions as toll-like receptor 7 agonists
WO2020007275A1 (en) 2018-07-03 2020-01-09 江苏恒瑞医药股份有限公司 Pyridopyrimidine derivative, preparation method therefor and medical use thereof
WO2020162705A1 (en) 2019-02-08 2020-08-13 성균관대학교산학협력단 Toll-like receptor 7 or 8 agonist-cholesterol complex, and use of same
US10786502B2 (en) 2016-12-05 2020-09-29 Apros Therapeutics, Inc. Substituted pyrimidines containing acidic groups as TLR7 modulators
WO2020204172A1 (en) 2019-04-05 2020-10-08 大日本住友製薬株式会社 Water soluble adjuvant
US10857153B2 (en) 2018-06-04 2020-12-08 Apros Therapeutics, Inc. Pyrimidine compounds containing acidic groups
WO2020255038A1 (en) 2019-06-18 2020-12-24 Janssen Sciences Ireland Unlimited Company Combination of hepatitis b virus (hbv) vaccines and pyridopyrimidine derivatives
WO2021136488A1 (en) 2020-01-02 2021-07-08 江苏恒瑞医药股份有限公司 Crystal form of pyridopyrimidine derivative and preparation method thereof
WO2021177679A1 (en) 2020-03-02 2021-09-10 성균관대학교산학협력단 Live-pathogen-mimetic nanoparticles based on pathogen cell wall skeleton, and production method thereof
WO2022031021A1 (en) 2020-08-04 2022-02-10 성균관대학교산학협력단 Mrna vaccine comprising adjuvant capable of kinetic control
WO2022031057A1 (en) 2020-08-04 2022-02-10 성균관대학교산학협력단 Conjugate of functional drug and toll-like receptor 7 or 8 agonist of which active site is temporarily inactivated and use thereof
WO2022031011A1 (en) 2020-08-04 2022-02-10 성균관대학교산학협력단 Kinetically acting adjuvant ensemble
RU2782066C2 (en) * 2016-12-05 2022-10-21 Эйпрос Терапьютикс, Инк. Pyrimidine compounds containing acid groups

Citations (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997022596A1 (en) 1995-12-18 1997-06-26 Zeneca Limited Quinazoline derivatives
WO1997030035A1 (en) 1996-02-13 1997-08-21 Zeneca Limited Quinazoline derivatives as vegf inhibitors
WO1997032856A1 (en) 1996-03-05 1997-09-12 Zeneca Limited 4-anilinoquinazoline derivatives
WO1998013354A1 (en) 1996-09-25 1998-04-02 Zeneca Limited Quinazoline derivatives and pharmaceutical compositions containing them
WO1999002166A1 (en) 1997-07-08 1999-01-21 Angiogene Pharmaceuticals Ltd. Use of colchinol derivatives as vascular damaging agents
WO2000040529A1 (en) 1999-01-07 2000-07-13 Angiogene Pharmaceuticals Ltd. Colchinol derivatives as vascular damaging agents
WO2000041669A2 (en) 1999-01-15 2000-07-20 Angiogene Pharmaceuticals Ltd. Benzimidazole vascular damaging agents
WO2000047212A1 (en) 1999-02-10 2000-08-17 Astrazeneca Ab Quinazoline derivatives as angiogenesis inhibitors
WO2001092224A1 (en) 2000-05-31 2001-12-06 Astrazeneca Ab Indole derivatives with vascular damaging activity
WO2001094341A1 (en) 2000-06-06 2001-12-13 Astrazeneca Ab Quinazoline derivatives for the treatment of tumours
WO2002004434A1 (en) 2000-07-07 2002-01-17 Angiogene Pharmaceuticals Limited Colchinol derivatives as vascular damaging agents
WO2002008213A1 (en) 2000-07-07 2002-01-31 Angiogene Pharmaceuticals Limited Colchinol derivatives as angiogenesis inhibitors
WO2009067081A1 (en) * 2007-11-22 2009-05-28 Astrazeneca Ab Pyrimidine derivatives for the treatment of asthma, copd, allergic rhinitis, allergic conjunctivitis, atopic dermatitis, cancer, hepatitis b, hepatitis c, hiv, hpv, bacterial infections and dermatosis
WO2010133885A1 (en) * 2009-05-21 2010-11-25 Astrazeneca Ab Novel pyrimidine derivatives and their use in the treatment of cancer and further diseases

Patent Citations (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997022596A1 (en) 1995-12-18 1997-06-26 Zeneca Limited Quinazoline derivatives
WO1997030035A1 (en) 1996-02-13 1997-08-21 Zeneca Limited Quinazoline derivatives as vegf inhibitors
WO1997032856A1 (en) 1996-03-05 1997-09-12 Zeneca Limited 4-anilinoquinazoline derivatives
WO1998013354A1 (en) 1996-09-25 1998-04-02 Zeneca Limited Quinazoline derivatives and pharmaceutical compositions containing them
WO1999002166A1 (en) 1997-07-08 1999-01-21 Angiogene Pharmaceuticals Ltd. Use of colchinol derivatives as vascular damaging agents
WO2000040529A1 (en) 1999-01-07 2000-07-13 Angiogene Pharmaceuticals Ltd. Colchinol derivatives as vascular damaging agents
WO2000041669A2 (en) 1999-01-15 2000-07-20 Angiogene Pharmaceuticals Ltd. Benzimidazole vascular damaging agents
WO2000047212A1 (en) 1999-02-10 2000-08-17 Astrazeneca Ab Quinazoline derivatives as angiogenesis inhibitors
WO2001092224A1 (en) 2000-05-31 2001-12-06 Astrazeneca Ab Indole derivatives with vascular damaging activity
WO2001094341A1 (en) 2000-06-06 2001-12-13 Astrazeneca Ab Quinazoline derivatives for the treatment of tumours
WO2002004434A1 (en) 2000-07-07 2002-01-17 Angiogene Pharmaceuticals Limited Colchinol derivatives as vascular damaging agents
WO2002008213A1 (en) 2000-07-07 2002-01-31 Angiogene Pharmaceuticals Limited Colchinol derivatives as angiogenesis inhibitors
WO2009067081A1 (en) * 2007-11-22 2009-05-28 Astrazeneca Ab Pyrimidine derivatives for the treatment of asthma, copd, allergic rhinitis, allergic conjunctivitis, atopic dermatitis, cancer, hepatitis b, hepatitis c, hiv, hpv, bacterial infections and dermatosis
WO2010133885A1 (en) * 2009-05-21 2010-11-25 Astrazeneca Ab Novel pyrimidine derivatives and their use in the treatment of cancer and further diseases

Non-Patent Citations (15)

* Cited by examiner, † Cited by third party
Title
"Bioreversible Carriers in Drug Design", 1987, PERGAMON PRESS
"Design of Pro-drugs", 1985, ELSEVIER
"Methods in Enzymology", vol. 42, 1985, ACADEMIC PRESS, pages: 309 - 396
CORWIN HANSCH: "Comprehensive Medicinal Chemistry", vol. 5, 1990, PERGAMON PRESS
GORDEN ET AL., J. IMMUNOL., vol. 174, 2005, pages 1259 - 1268
H. BUNDGAARD ET AL., JOURNAL OF PHARMACEUTICAL SCIENCES, vol. 77, 1988, pages 285
H. BUNDGAARD, ADVANCED DRUG DELIVERY REVIEWS, vol. 8, 1992, pages 1 - 38
H. BUNDGAARD: "A Textbook of Drug Design and Development", DESIGN AND APPLICATION OF PRO-DRUGS, 1991, pages 113 - 191
HUBER ET AL., J. IMMUNOL., vol. 185, 2010, pages 813 - 817
J. MED. CHEM., vol. 47, 2004, pages 6658 - 6661
N. KAKEYA ET AL., CHEM. PHARM. BULL., vol. 32, 1984, pages 692
PERSSON, F. ET AL., J CARDIOVASC.ELECTROPHYSIOL., vol. 16, 2005, pages 329 - 341
SCHROEDER, K. ET AL., JBIOMOL SCREEN., vol. 8, 2003, pages 50 - 64
STERN ET AL., CRITICAL REVIEWS IN ONCOLOGY/HAEMATOLOGY, vol. 54, 2005, pages 1 - 29
T. HIGUCHI; V. STELLA: "Pro-Drugs as Novel Delivery Systems", A.C.S. SYMPOSIUM SERIES, vol. 14

Cited By (63)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9045472B2 (en) 2010-12-16 2015-06-02 Astrazeneca Ab Imidazoquinoline compounds
KR102194585B1 (en) 2012-05-18 2020-12-23 다이닛본 스미토모 세이야꾸 가부시끼가이샤 Carboxylic acid compounds
CN104302628A (en) * 2012-05-18 2015-01-21 大日本住友制药株式会社 Carboxylic acid compounds
KR20150023410A (en) * 2012-05-18 2015-03-05 다이닛본 스미토모 세이야꾸 가부시끼가이샤 Carboxylic acid compounds
US10562861B2 (en) 2012-05-18 2020-02-18 Sumitomo Dainippon Pharma Co., Ltd. Carboxylic acid compounds
US9376398B2 (en) 2012-05-18 2016-06-28 Sumitomo Dainippon Pharma Co., Ltd Carboxylic acid compounds
WO2013172479A1 (en) * 2012-05-18 2013-11-21 Dainippon Sumitomo Pharma Co., Ltd. Carboxylic acid compounds
US11299465B2 (en) 2012-05-18 2022-04-12 Sumitomo Dainippon Pharma Co., Ltd. Carboxylic acid compounds
RU2636146C2 (en) * 2012-05-18 2017-11-21 Сумитомо Дайниппон Фарма Ко., Лтд. Carboxylic acids compounds
US9447097B2 (en) 2014-04-22 2016-09-20 Hoffmann-La Roche Inc. 4-amino-imidazoquinoline compounds
US9334268B2 (en) 2014-04-22 2016-05-10 Hoffman-La Roche Inc. 4-amino-imidazoquinoline compounds
TWI666212B (en) * 2014-05-01 2019-07-21 瑞士商諾華公司 Compounds and compositions as toll-like receptor 7 agonists
US10844068B2 (en) 2014-05-01 2020-11-24 Novartis Ag Compounds and compositions as toll-like receptor 7 agonists
US10407431B2 (en) 2014-05-01 2019-09-10 Novartis Ag Compounds and compositions as toll-like receptor 7 agonists
US9597333B2 (en) 2015-03-06 2017-03-21 Hoffmann-La Roche Inc. Benzazepine dicarboxamide compounds
US9822065B1 (en) 2015-03-06 2017-11-21 Hoffmann-La Roche Inc. Benzazepine dicarboxamide compounds
US9475775B2 (en) 2015-03-06 2016-10-25 Hoffmann-La Roche Inc. Benzazepine dicarboxamide compounds
WO2017046112A1 (en) 2015-09-17 2017-03-23 F. Hoffmann-La Roche Ag Sulfinylphenyl or sulfonimidoylphenyl benzazepines
JPWO2017061532A1 (en) * 2015-10-07 2018-08-02 大日本住友製薬株式会社 Pyrimidine compounds
EP3868741A1 (en) 2015-10-07 2021-08-25 Sumitomo Dainippon Pharma Co., Ltd. Pyrimidine compound
JP7195398B2 (en) 2015-10-07 2022-12-23 住友ファーマ株式会社 pyrimidine compound
CN108290845A (en) * 2015-10-07 2018-07-17 大日本住友制药株式会社 Pyrimidine compound
KR102651207B1 (en) * 2015-10-07 2024-03-25 스미토모 파마 가부시키가이샤 pyrimidine compounds
US11266738B2 (en) 2015-10-07 2022-03-08 Sumitomo Dainippon Pharma Co., Ltd. Substituted pyrimidines as vaccine adjuvants
WO2017061532A1 (en) * 2015-10-07 2017-04-13 大日本住友製薬株式会社 Pyrimidine compound
JP2022010266A (en) * 2015-10-07 2022-01-14 大日本住友製薬株式会社 Pyrimidine compound
KR20180056774A (en) * 2015-10-07 2018-05-29 다이닛본 스미토모 세이야꾸 가부시끼가이샤 Pyrimidine compound
US11911466B2 (en) 2015-10-07 2024-02-27 Sumitomo Pharma Co., Ltd. Substituted pyrimidines as vaccine adjuvants
AU2016334826B2 (en) * 2015-10-07 2020-08-27 Sumitomo Pharma Co., Ltd. Pyrimidine compound
CN108290845B (en) * 2015-10-07 2021-08-03 大日本住友制药株式会社 Pyrimidine compounds
JP2021001210A (en) * 2015-10-07 2021-01-07 大日本住友製薬株式会社 Pyrimidine compound
TWI718188B (en) * 2015-10-07 2021-02-11 日商大日本住友製藥股份有限公司 Pyrimidine compounds
US10632192B2 (en) 2015-10-07 2020-04-28 Sumitomo Dainippon Pharma Co., Ltd. Pyrimidine compounds and their use as vaccine adjuvants
WO2017202703A1 (en) 2016-05-23 2017-11-30 F. Hoffmann-La Roche Ag Benzazepine dicarboxamide compounds with secondary amide function
US10544102B2 (en) 2016-05-23 2020-01-28 Hoffmann-La Roche Inc. Benzazepine dicarboxamide compounds with secondary amide function
US10370338B2 (en) 2016-05-23 2019-08-06 Hoffmann-La Roche Inc. Benzazepine dicarboxamide compounds with tertiary amide function
WO2017216054A1 (en) 2016-06-12 2017-12-21 F. Hoffmann-La Roche Ag Dihydropyrimidinyl benzazepine carboxamide compounds
US10590112B2 (en) 2016-06-12 2020-03-17 Hoffmann-La Roche Inc. Dihydropyrimidinyl benzazepine carboxamide compounds
US11046671B2 (en) 2016-06-12 2021-06-29 Hoffmann-La Roche Inc. Dihydropyrimidinyl benzazepine carboxamide compounds
CN110177780A (en) * 2016-12-05 2019-08-27 阿普罗斯治疗公司 Pyrimidine compound containing acidic groups
US10287253B2 (en) 2016-12-05 2019-05-14 Apros Therapeutics, Inc. Substituted pyrimidines containing acidic groups as TLR7 modulators
RU2782066C2 (en) * 2016-12-05 2022-10-21 Эйпрос Терапьютикс, Инк. Pyrimidine compounds containing acid groups
US10786502B2 (en) 2016-12-05 2020-09-29 Apros Therapeutics, Inc. Substituted pyrimidines containing acidic groups as TLR7 modulators
JP7071392B2 (en) 2016-12-05 2022-05-18 アプロス セラピューティクス, インコーポレイテッド Pyrimidine compound containing an acidic group
IL266875A (en) * 2016-12-05 2019-07-31 Apros Therapeutics Inc Pyrimidine compounds containing acidic groups
JP2020504170A (en) * 2016-12-05 2020-02-06 アプロス セラピューティクス, インコーポレイテッド Pyrimidine compounds containing acidic groups
WO2018106606A1 (en) * 2016-12-05 2018-06-14 Apros Therapeutics, Inc. Pyrimidine compounds containing acidic groups
US11173157B2 (en) 2016-12-05 2021-11-16 Apros Therapeutics, Inc. Substituted pyrimidines containing acidic groups as TLR7 modulators
US11833247B2 (en) 2017-03-29 2023-12-05 Sumitomo Pharma Co., Ltd. Vaccine adjuvant formulation
WO2018181420A1 (en) 2017-03-29 2018-10-04 大日本住友製薬株式会社 Vaccine adjuvant formulation
US11253476B2 (en) 2017-03-29 2022-02-22 Sumitomo Dainippon Pharma Co., Ltd. Vaccine adjuvant formulation
US11806364B2 (en) 2017-09-28 2023-11-07 Industry-Academic Cooperation Foundation, Yonsei University Method for producing myeloid-derived suppressor cells, myeloid-derived suppressor cells produced thereby, and methods thereof
WO2019066571A2 (en) 2017-09-28 2019-04-04 연세대학교 산학협력단 Method for producing myeloid-derived suppressor cells, myeloid-derived suppressor cells produced thereby, and uses thereof
US10857153B2 (en) 2018-06-04 2020-12-08 Apros Therapeutics, Inc. Pyrimidine compounds containing acidic groups
WO2020007275A1 (en) 2018-07-03 2020-01-09 江苏恒瑞医药股份有限公司 Pyridopyrimidine derivative, preparation method therefor and medical use thereof
WO2020162705A1 (en) 2019-02-08 2020-08-13 성균관대학교산학협력단 Toll-like receptor 7 or 8 agonist-cholesterol complex, and use of same
WO2020204172A1 (en) 2019-04-05 2020-10-08 大日本住友製薬株式会社 Water soluble adjuvant
WO2020255038A1 (en) 2019-06-18 2020-12-24 Janssen Sciences Ireland Unlimited Company Combination of hepatitis b virus (hbv) vaccines and pyridopyrimidine derivatives
WO2021136488A1 (en) 2020-01-02 2021-07-08 江苏恒瑞医药股份有限公司 Crystal form of pyridopyrimidine derivative and preparation method thereof
WO2021177679A1 (en) 2020-03-02 2021-09-10 성균관대학교산학협력단 Live-pathogen-mimetic nanoparticles based on pathogen cell wall skeleton, and production method thereof
WO2022031057A1 (en) 2020-08-04 2022-02-10 성균관대학교산학협력단 Conjugate of functional drug and toll-like receptor 7 or 8 agonist of which active site is temporarily inactivated and use thereof
WO2022031021A1 (en) 2020-08-04 2022-02-10 성균관대학교산학협력단 Mrna vaccine comprising adjuvant capable of kinetic control
WO2022031011A1 (en) 2020-08-04 2022-02-10 성균관대학교산학협력단 Kinetically acting adjuvant ensemble

Similar Documents

Publication Publication Date Title
WO2012066336A1 (en) Benzylamine compounds as toll -like receptor 7 agonists
US20220267282A1 (en) Carboxylic acid compounds
WO2012066335A1 (en) Phenol compounds als toll -like receptor 7 agonists
EP2432768B1 (en) Novel pyrimidine derivatives and their use in the treatment of cancer and further diseases
WO2012067269A1 (en) Aminoalkoxyphenyl compounds and their use in the treatment of disease
EP3596061A1 (en) Compounds useful in the treatment or prevention of a prmt5-mediated disorder
WO2012067268A1 (en) Cyclic amide compounds and their use in the treatment of disease
RU2633694C2 (en) Dyetherned phenylaminopyrimidine and pharmaceutical composition containing such connection
WO2020188467A1 (en) Condensed tricyclic compound used as kinase inhibitor
KR20230154209A (en) Benzo[C][2,6]naphthyridine derivatives, compositions and therapeutic uses thereof
US20100292222A1 (en) Chemical compounds 751

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: 11791620

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: 11791620

Country of ref document: EP

Kind code of ref document: A1