WO2012066335A1 - Phenol compounds als toll -like receptor 7 agonists - Google Patents

Phenol compounds als toll -like receptor 7 agonists Download PDF

Info

Publication number
WO2012066335A1
WO2012066335A1 PCT/GB2011/052245 GB2011052245W WO2012066335A1 WO 2012066335 A1 WO2012066335 A1 WO 2012066335A1 GB 2011052245 W GB2011052245 W GB 2011052245W WO 2012066335 A1 WO2012066335 A1 WO 2012066335A1
Authority
WO
WIPO (PCT)
Prior art keywords
amino
methyl
formula
compound
methylpyrimidin
Prior art date
Application number
PCT/GB2011/052245
Other languages
French (fr)
Inventor
Andrew Bailey
Adrian John Highton
Thomas Mcinally
Tobias Mochel
Daisuke Urabe
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 WO2012066335A1 publication Critical patent/WO2012066335A1/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

Definitions

  • the present invention relates to novel phenol compounds and, more particularly, to novel phenol compounds that act as TLR7 agonists.
  • This invention also relates to methods for the preparation of such compounds and novel intermediates 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 (LC)), 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
  • 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 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.
  • the phenol compounds of the present invention are capable of activating TLR7 in vitro.
  • the phenol 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.
  • some compounds of the invention also have a surprisingly advantageous hERG profile.
  • Compounds that have significant activities against the hERG ion channel are relevant to inducing QT prolongation and are disfavored because such activity is implicated in the development of Torsades de Pointes and cardiac death.
  • Q is a direct bond, -0-(C 2 - 4 alkylene)- or -(Ci_ 4 alkylene)-;
  • R 1 is selected from hydrogen, Ci_ 4 alkyl, Ci_ 3 alkoxyC 2 - 4 alkyl, hydroxyC 2 _ 4 alkyl, C 3 _ 5 cycloalkyl, tetrahydropyranyl, benzyl, C-linked pyrrolidinyl, C-linked piperidinyl and (R x )(R Y )N-C 2 - 4 alkyl, wherein R x and R Y each independently represent hydrogen or Ci_ 3 alkyl; and
  • R 2 is selected from hydrogen, Ci_ 4 alkyl and Ci_ 3 alkoxyC 2 - 4 alkyl;
  • R 1 and R 2 together with the nitrogen to which they are attached form a saturated heterocyclyl selected from azetidin-l-yl, pyrrolidin-l-yl, piperidin-l-yl, morpholin-4-yl, piperazin-l-yl, azepan-l-yl, 1,4-diazepan-l-yl; wherein one or two carbon atoms within said heterocyclyl may be substituted by oxo and wherein the available nitrogen of said piperazin-l-yl and 1,4-diazepan-l-yl is optionally substituted by Ci_ 3 alkyl,
  • 5-membered aromatic heterocyclyl containing a total of 1-3 nitrogen atoms; wherein said 5-membered aromatic heterocyclyl is optionally substituted on a ring carbon atom by Ci_ 3 alkyl;
  • R 3 is selected from hydrogen, hydroxymethyl and 2-hydroxyethyl
  • the "available nitrogen" of a piperazin-l-yl and 1,4- diazepan-l-yl is at the 4-positions of the piperazin-l-yl and the 1,4-diazepan-l-yl.
  • 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 alkylene is intended to mean divalent group of a saturated carbon chain of 1 to 4 carbon atoms in length which may be straight-chained or branched, preferably straight chained.
  • Ci_ 4 alkylene includes, but is not limited to, methylene, ethylene, trimethylene, tetramethylene.
  • C 2 - 4 alkylene is to be construed accordingly.
  • 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.
  • “Ci_ 4 alkyl” includes, but is not limited to, methyl, ethyl, propyl, isopropyl, butyl, and tert-butyl.
  • the term “C 2 _ 4 alkyl” and “Ci_ 3 alkyl” are to be construed accordingly.
  • Ci_ 3 alkoxyC 2 - 4 alkyl is intended to mean a saturated carbon chain of 1 to 3 carbon atoms in length, which may be straight-chained or branched, linked via oxygen to another saturated carbon chain of 2 to 4 carbon atoms in length, which may be straight- chained or branched.
  • “Ci_ 3 alkoxyC 2 - 4 alkyl” includes, but is not limited to, methoxyethyl, methoxypropyl, ethoxypropyl, propoxyethyl and methoxypropyl.
  • hydroxyC 2 - 4 alkyl is intended to mean a saturated carbon chain of 2 to 4 carbon atoms in length, which may be straight-chained or branched, wherein one of the hydrogen atoms has been replaced by a hydroxy group.
  • hydroxyC 2 - 4 alkyl includes, but is not limited to, hydroxyethyl, 2-hydroxypropyl, 3-hydroxypropyl, 1- hydroxyisopropyl and 4-hydroxybutyl.
  • (R x )(R Y )N-C 2 - 4 alkyl is intended to mean a saturated carbon chain of 2 to 4 carbon atoms in length, which may be straight-chained or branched, wherein one of the hydrogen atoms has been replaced by a group "(R X )(R Y )N-", wherein R x and R Y each independently represent hydrogen or Ci_ 3 alkyl.
  • the "Ci_ 3 alkyl” is intended to mean a saturated carbon chain of 1 to 3 carbon atoms in length which may be straight-chained or branched, including methyl, ethyl, propyl and isopropyl.
  • R 1 , R 2 , R 3 and Q may take any of the values defined herein R 1 , R 2 , R 3 and Q respectively.
  • variable groups R 1 , R 2 , R 3 and Q 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.
  • Q is -0-(C 2 - 4 alkylene)-.
  • Q is -O-CH2CH2- or -O-CH2CH2CH2-.
  • Q is -(Ci_ 4 alkylene)-.
  • Q is -CH 2 - or -CH 2 CH 2 CH 2 -.
  • Q is -CH 2 CH 2 CH 2 -.
  • Q is -0-(C 2 _ 4 alkylene)- or -(Ci_ 4 alkylene)-.
  • Q is -0-(C 2 _ 4 alkylene)- or -(Ci_ 4 alkylene)- wherein said alkylene is a straight chain alkylene.
  • R 1 is hydrogen
  • R 1 is Ci_ 4 alkyl, Ci_ 3 alkoxyC 2 _ 4 alkyl, hydroxyC 2 _ 4 alkyl, C 3 _ 5 Cycloalkyl,
  • R 1 is Ci_ 4 alkyl, Ci_ 3 alkoxyC 2 _ 4 alkyl or hydroxyC 2 _ 4 alkyl.
  • R 1 is selected from methyl, ethyl, 2-methoxyethyl and 2-hydroxyethyl.
  • R 2 is hydrogen
  • R 2 is Ci_ 4 alkyl or Ci_ 3 alkoxyC 2 _ 4 alkyl.
  • R 2 is Ci_ 4 alkyl.
  • R 2 is methyl or ethyl.
  • R 2 is Ci_ 3 alkoxyC 2 _ 4 alkyl.
  • 1,4-diazepan-l-yl is optionally substituted by Ci_ 3 alkyl, hydroxyCi_ 3 alkyl or
  • 5-membered aromatic heterocyclyl containing a total of 1-3 nitrogen atoms; wherein said 5-membered aromatic heterocyclyl is optionally substituted on a ring carbon atom by Ci_ 3 alkyl.
  • R 1 is Ci_ 4 alkyl, Ci_ 3 alkoxyC 2 - 4 alkyl or hydroxyC 2 - 4 alkyl; and R 2 is Ci_ 4 alkyl; or R 1 and R 2 together with the nitrogen to which they are attached form a pyrrolidine- 1-yl or piperazin-l-yl wherein said piperazin-l-yl is substituted by Ci_ 3 alkyl.
  • R 1 is methyl, ethyl, 2-methoxyethyl or 2-hydroxyethyl; and R 2 is methyl or ethyl; or R 1 and R 2 together with the nitrogen to which they are attached form a pyrrolidine- 1-yl or 4-methylpiperazin-l-yl.
  • R 3 is hydrogen
  • R 3 is hydroxymethyl
  • R 3 is 2-hydroxyethyl.
  • R 3 is hydroxymethyl or 2-hydroxyethyl.
  • Q is -0-(C 2 - 4 alkylene)- or -(Ci_ 4 alkylene)-;
  • R 3 is selected from hydrogen, hydroxymethyl and 2-hydroxyethyl.
  • 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, that is 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.
  • 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).
  • the compound of Formula (I) may be prepared by reacting a compound of Formula (II), or a salt thereof,
  • R 1 , R 2 and Q may take any of the values for R 1 , R 2 and Q as defined herein, and where R 4 may take any of the values for R 3 as defined herein or R 4 may be a protected hydroxyalkyl group: PG 1 -0-(CH 2 ) n -, wherein n is 1 or 2 and PG 1 is a protecting group.
  • n is 1.
  • n is 2.
  • PG 1 is selected from a benzyl-based protecting group and a silyl-based protecting group.
  • a benzyl-based protecting group may be benzyl.
  • a silyl-based protecting group may be a tri(Ci_ 4 alkyl)silyl group, for example a triemthylsilyl (TMS) or fert-butyldimethylsilyl (TBDMS) protecting group.
  • TMS triemthylsilyl
  • TDMS fert-butyldimethylsilyl
  • a compound of Formula (II), or a salt thereof as defined herein.
  • the preparation of a compound of Formula (I) from a compound of Formula (II) may be carried out with or without any hydroxy group within R 4 being protected. Any protecting group used on a hydroxyl group within R 4 may be removed before or after the reaction with BBr 3 (the chemistry that reveals the phenol functionality).
  • the R 5A group of the compound of Formula (III-A) may be a substituted or unsubstituted hydrocarbyl group.
  • R 5A may be a hydrocarbyl group selected from benzyl and Ci_ 4 alkyl, for example methyl or ethyl.
  • the LG 2A group of the compound of Formula (III-B) 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 (III- A) and Formula (III-B): For example, reaction of a compound of Formula (III-B) 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 (III-A). 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 butylamine or 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 (TFA) at elevated temperature (for example 50-200°C) using conventional or microwave heating.
  • TFA trifluoroacetic acid
  • the R 4 group in the compound of Formula (VII) and the compound of Formula (VIII) is independently selected from same R 4 values described hereinbefore in connection with the compound of Formula (II).
  • the compound of Formula (VII) and/or the compound of Formula (VIII) and/or the compound of Formula (II- A), when R 4 is other than hydrogen, has an (5)-stereocentre at the asymmetric carbon atom where the R 4 group is attached.
  • 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 (II- A) may be prepared by reaction of a compound of (VIII) with the appropriate amine (FINR ⁇ 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 CH 2 CI 2 and acetic acid in the presence of activated molecular sieves, or by using NaBH 4 in a suitable solvent such as methanol.
  • the R group of the compound of Formula (IX) may be a substituted or unsubstituted hydrocarbyl group.
  • R 5B may be a hydrocarbyl group selected from benzyl and Ci_4alkyl, for example methyl or ethyl.
  • the LG 2B 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_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.
  • 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 (XI) 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 butylamine or 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 TFA at elevated temperature (for example 50-200°C) using conventional or microwave heating.
  • a suitable solvent such as propionitrile, butanol or 1 ,4-dioxane
  • the compound of Formula (XIV) has an (iS)-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
  • the compounds of Formula (XIV- A) and (XV- A) are compounds of Formula (XIV) and (XV) respectively, as hereinbefore defined, wherein R 4 is either hydrogen or PG 1 -0-(CH 2 ) n -, as defined hereinbefore.
  • R 4 is either hydrogen or PG 1 -0-(CH 2 ) n - wherein n is 1 or 2, and wherein PG 1 is a tert-butyldimethylsilyl (TBDMS) protecting group; or a salt thereof.
  • Step (i) Compounds of Formula (XV- A) may be prepared from compounds of Formula (XIV- A) 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 CH 3 SO 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 (XVI) may be converted into a compound of Formula (II- A) 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), when R 4 is other than hydrogen, has an (iS)-stereocentre at the carbon atom to which the R 4 group is attached.
  • the R 7 group in the compound of Formula (XXII) may take any of the values of R 7 described herein.
  • R 7 may be a 2,4,6-trimethylphenyl group.
  • the PG 3 group in the compounds of Formula (XXII) and Formula (XXVII) may be any suitable protecting group for an alcohol, for example a tri(Ci_ 4 alkyl)silyl protecting group, such as TBDMS.
  • TBDMS tri(Ci_ 4 alkyl)silyl protecting group
  • the R 4 group in the compound of Formula (XXVI) is other than hydrogen then the alcohol group within the R 4 group is protected by a benzyl-based protecting group PG 4 , for example with a benzyl group.
  • Step (x): Compounds of Formula (XXVII) may be prepared by reacting a compound of Formula (XXII) with an excess of a compound of Formula (XXVI) in a suitable solvent, such as propionitrile, butanol or 1,4-dioxane, in the presence of TFA at a suitable temperature, for example 50-150°C. Alternatively, the reaction may be performed in a microwave at a suitable temperature, for example 50-200°C.
  • a suitable solvent such as propionitrile, butanol or 1,4-dioxane
  • the reaction may be carried out using an acid such as hydrochloric acid or TFA, in a suitable solvent, such as 1,4-dioxane or MeOH at a suitable temperature, for example 0-50°C.
  • Step (xii): Compounds of Formula (XXIX) may be prepared by reacting a compound of Formula (XXVIII) with a hydrocarbyl sulfonyl chloride: R 7 S0 2 C1, for example mesyl chloride, trimethylbenzene sulfonyl chloride or triisopropylbenzenesulfonyl chloride, in a suitable solvent, such as CH 2 C1 2 or THF in the presence of a suitable base, such as metal carbonate base, or an organic base, such as tertiary amine base, for example, triethylamine, at a suitable temperature, for example 0-50°C.
  • a suitable solvent such as CH 2 C1 2 or THF
  • a suitable base such as metal carbonate base
  • organic base such as tertiary amine base, for example, triethylamine
  • reaction may be performed in a microwave at a suitable temperature, for example 50-200°C.
  • the reaction may be carried out with a catalyst such as Pd/C under H 2 atmosphere (1-20 bar) in a suitable solvent, such as MeOH at a suitable temperature, for example 20-100°C, optionally in the presence of an additive such as hydrochloric acid or acetic acid.
  • the compound of Formula (XXII) may be prepared as shown in Scheme 6.
  • Step (i): Compounds of Formula (XVII) may be prepared using an alkylation reaction, for example by reacting a compound of Formula (XVI) with (3-bromopropoxy)- tert-butyldimethylsilane in the case where PG 3 is TBDMS, in a suitable solvent, such as DMF in the presence of a suitable base, such as K 2 CO 3 at a suitable temperature, for example 0-50°C.
  • a suitable solvent such as DMF
  • a suitable base such as K 2 CO 3
  • Step (ii): Compounds of Formula (XVIII) may be prepared using any reducing agent that is known to be capable of reducing an aldehyde to the corresponding alcohol, for example by reacting a compound of Formula (XVII) with NaBH 4 in a suitable solvent, such as MeOH at a suitable temperature, for example 0°C to 50°C.
  • a suitable solvent such as MeOH
  • Step (iii): Compounds of Formula (XIX) may be prepared by reacting a compound of Formula (XVII) with a hydrocarbylsulfonyl chloride and a salt such as LiCl, in a suitable solvent, such as THF in the presence of a suitable base, such as DIPEA, at a suitable temperature, for example 0-50°C.
  • Step (iv): Compounds of Formula (XX) can be prepared by reaction of a compound of Formula (III-A), for example, methylacetoacetate, 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 (XIX). The reaction is then preferably heated, for example at 50-100°C, optionally in the presence of an additive such as KI.
  • a compound of Formula (III-A) for example, methylacetoacetate
  • a suitable solvent such as THF or DMF
  • Step (v): Compounds of Formula (XXI) may be prepared by reacting a compound of Formula (XX) with guanidine or guanidine carbonate in a suitable solvent, such as MeOH or EtOH, and at a suitable temperature, for example 50-150°C.
  • a suitable solvent such as MeOH or EtOH
  • Step (vi): Compounds of Formula (XXII) may be prepared by reacting a compound of Formula (XXI) with a suitable a hydrocarbyl sulfonyl chloride: R 7 S0 2 C1, 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 tetramethylpropylenediamine (TMPDA) at a suitable temperature, for example 0-50°C.
  • Step (vii): Compounds of Formula (XXIV) may be prepared by reacting a compound of Formula (XXIII) with di-fert-butyl dicarbonate. The reaction may be carried out in a suitable solvent, such methanol and water in the presence of a suitable base, such as, NaOH at a suitable temperature, for example 0-50°C.
  • a suitable solvent such methanol and water
  • a suitable base such as, NaOH
  • Step (viii): Compounds of Formula (XXV) may be prepared by reacting a compound of Formula (XXIV) with the appropriate benzyl bromide or chloride in a suitable solvent, such as DMF in the presence of a suitable base, such as DIPEA, at a suitable temperature, for example 0-50°C.
  • Step (ix) Compounds of Formula (XXVI) may be prepared by reacting a compound of Formula (XXV) with an acid such as hydrochloric acid or TFA in a suitable solvent, such as 1,4-dioxane or MeOH at a suitable temperature, for example 0-50°C.
  • an acid such as hydrochloric acid or TFA
  • a suitable solvent such as 1,4-dioxane or MeOH
  • Scheme 8 Step (i): Compounds of Formula (XXXIII) can be prepared by reaction of a compound of Formula (XXXII) 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 (III- A) wherein LG 4 represents a suitable leaving group, for example bromo or chloro. The reaction is then preferably heated, for example at 50-100°C, optionally in the presence of an additive such as KI.
  • a base such as NaH
  • a suitable solvent such as THF or DMF
  • Step (ii): Compounds of Formula (XXXIV) may be prepared by reacting a compound of Formula (XXXIII) with guanidine or guanidine carbonate in a suitable solvent, such as MeOH or EtOH, and at a suitable temperature, for example 50-150°C.
  • a suitable solvent such as MeOH or EtOH
  • Step (iii): Compounds of Formula (XXXV) may be prepared by reacting a compound of Formula (XXXIV) with a a hydrocarbyl sulfonyl chloride: R 7 S0 2 C1, 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 TMPDA or DABCO at a suitable temperature, for example 0-50°C.
  • Step (iv): Compounds of Formula (XXVI) may be prepared by reacting a compound of Formula (XXXV) with a suitable acrylamide such as N,N-dimethyl- acrylamide. The reaction may be carried out in a suitable base such as triethylamine in the presence of palladium (II) acetate and tri-ort/zo-tolylphosphine at a suitable temperature for example 70-80°C.
  • a suitable base such as triethylamine
  • palladium (II) acetate and tri-ort/zo-tolylphosphine at a suitable temperature for example 70-80°C.
  • Step (v): Compounds of Formula (XXXVII) may be prepared by treating a compound of Formula (XXXVI) with H 2 in the presence of palladium on carbon, in a suitable solvent such as ethanol or EtOAc.
  • Step (vi): Compounds of Formula (XXXVIII) may be prepared by reacting a compound of Formula (XXXVII) with an excess of the appropriate amine, in a suitable solvent, such as propionitrile, butanol, butyronitrile or 1,4-dioxane, in the presence of TFA at a suitable temperature, for example 50-150°C. Alternatively, the reaction may be performed in a microwave at a suitable temperature, for example 50-100°C.
  • a suitable solvent such as propionitrile, butanol, butyronitrile or 1,4-dioxane
  • Step (vii): Compound of Formula (VI V) may be prepared by reacting a compound of Formula (XXXVIII) with BH 3 -dimethyl sulphide complex in a suitable solvent such as THF at a suitable temperature, for example 60°C.
  • a suitable solvent such as THF
  • Compounds of Formula (II-B) may also be prepared as shown in Scheme 9.
  • Step (i): Compounds of a Formula (XLIII) may be prepared by reacting compounds of Formula (XLI) and (XLII) in a suitable solvent such as DMF in the presence of Pd-118 (1,1 -bis(di-tert-butylphosphino)-ferrocenepalladium(II) dichloride), tetrabutylammonium chloride, hydrate and N-cyclohexyl-N-methylcyclo-hexanamine at a sutiable temperature, for example 100°C.
  • LG 5 is a leaving group, for example a chloro or bromo.
  • PG 5 is a suitable protecting group, for example a benzyl-based protecting group, for example benzyl.
  • Step (ii): Compounds of Formula (XLIV) may be prepared by reacting a compound of Formula (XLIII) with guanidine or guanidine carbonate, in a suitable solvent, such as MeOH or EtOH, and at a suitable temperature, for example 50-150°C.
  • a suitable solvent such as MeOH or EtOH
  • Step (iii): Compounds of Formula (XLV) may be prepared by reacting a compound of Formula (XLIV) with POCI 3 , at a suitable temperature, for example 50-110°C.
  • Step (iv): Compounds of Formula (XL VI) may be prepared by reacting a compound of Formula (XLV) with an excess of the appropriate amine, in a suitable solvent, such as propionitrile ,butanol or 1,4-dioxane, in the presence of TFA at a suitable temperature, for example 50-150°C.
  • the reaction may be performed in a microwave at a suitable temperature, for example 50-200°C.
  • Step (v): -Compounds of Formula (XL VII) may be prepared by reacting a compound of Formula (XL VI) with a suitable protecting group.
  • PG 6 may be a tri(Ci_ 4 alkyl)silyl group, for example TBDMS or TMS.
  • PG 6 is a silicon based protecting group, it may be added using, for example TBDMSCl, or TMSCl, in the presence of a suitable base, for example imidazole, in a suitable solvent such as DMF.
  • Step (vii): Compounds of Formula (XLIX) may be prepared by reacting a compound of a Formula (XL VIII) with the appropriate amino alcohol, in a suitable solvent such as THF using Mitsunobu conditions, for example using diisopropyl azodicarboxylate and tripheny lphosphine .
  • Step (viii): Compounds of Formula (XXXI) may be prepared by using appropriate deprotection conditions known to be suitable for removing the PG 6 group.
  • deprotection conditions known to be suitable for removing the PG 6 group.
  • PG 6 were a silicon-based protecting group
  • the deprotection could involve reaction of a compound of Formula (XLIX) with a fluoride salt, for example tetra-n-butylammonium fluoride (TBAF), in a suitable solvent such as CH 2 C1 2 .
  • a deproection may be performed using SCX resin with a suitable solvent such as acetonitrile.
  • 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.
  • hydrofluoroalkane 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.
  • surfactant e.g., oleic acid or lecithin
  • cosolvent e.g., ethanol
  • 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 z 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 a 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.
  • 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. In any aspect or embodiment described herein 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, miriplatin, carboplatin, 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
  • epipodophyllotoxins like etoposide and teniposide, amsacrine, topotecan and
  • 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-l-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 inter leukin 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 inter leukin 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) 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;
  • kits 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.
  • DABCO l,4-diazabicyclo[2.2.2]octane
  • FCC flash column chromatography using silica
  • TFA TFA
  • DIPEA diisopropylethylamine
  • DMSO dimethylsulfoxide
  • aq aqueous
  • eq. molar equivalents
  • 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 molar.
  • Proton nuclear magnetic resonance data (“ 1 H NMR”) was generally obtained at 300-500 MHz and using deuterated DMSO unless otherwise stated.
  • Example 1A 2-[(2-Amino-4- ⁇ [(3S -l-hvdroxyhexan-3-yllamino ⁇ -6-methylpyrimidin- 5-yl)methyll-5- ⁇ [(2-hvdroxyethyl)(methyl)aminolmethyl ⁇ phenol bis saccharin salt
  • the title compound may be prepared by the steps described below:
  • Butylamine (1.26 g; 5eq) was reacted using the general method described for step (iv) of Example 1, and with heating for 3h. Purification by FCC, eluting with 0-20% CH 3 OH (and 0.7M NH 3 ) in CH 2 C1 2 gave the sub-title compound (0.7 g) as an oil; LC-MS: m/z 326 APCI+.
  • Example 3 2- ⁇ [2- Amino-4-(butylamino)-6-methylpyrimidin-5-yll methyl ⁇ -5- ⁇ (4- methylpiperazin-l-vDmethyllphenol
  • 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:
  • the title compound may be prepared by the steps described below:
  • the crude material was dissolved in CH 3 OH (25 mL), acidified with acetic acid (1 mL) and loaded on to a 50g SCX cartridge. The impurities were washed through with CH 3 OH (125 mL) and discarded. The product was eluted with 1M NH 3 /CH 3 OH (250 mL) and concentrated in vacuo. The product was purified by FCC, eluting with 10% CH 3 OH in EtOAc to give the subtitle compound (1 g) as a yellow gum; LC-MS: m/z 370 APCI+.
  • the title compound may be prepared by the steps described below:
  • Example 8 2-[(2-Amino-4- ⁇ [(2S -l-hvdroxypentan-2-yllamino ⁇ -6-methylpyrimidin-5- vDmethyll -5- [(dimethylamino)methyll phenol
  • 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:
  • PBr 3 (0.95 mL) was added drop-wise to a vigorously stirred solution of (4-bromo-2- methoxyphenyl)methanol (4.4 g) in CH 2 CI 2 (100 mL) at 0°C. After 40 mins the mixture was slowly quenched with ice/water and the organic solution was separated and the aqueous phase was extracted with CH 2 CI 2 . The organic solutions were combined, washed with water, dried (MgS0 4 ), and concentrated in vacuo to give the sub-title compound (5.2 g); 1H NMR: 7.19 (d, 1H), 7.07 (dd, 1H), 7.01 (d, 1H), 4.49 (s, 2H), 3.89 (s, 3H).
  • the title compound may be prepared by the steps described below:
  • Example 12 2-[(2-Amino-4- ⁇ [(3S -l-hvdroxyhexan-3-yllamino ⁇ -6-methylpyrimidin- 5-vDmethyll -5- [3-(dimethylamino)propoxyl phenol
  • the title compound may be prepared by the steps described below:
  • Methanesulfonyl chloride (4.02 mL) was added to a mixture of ⁇ 4-[3-(tert- butyldimethylsilyloxy)-propoxy]-2-methoxyphenyl ⁇ methanol (8.47 g), DIPEA (13.4 mL) and LiCl (3.29 g) in THF (105 mL) at RT. The mixture was then stirred for 40 mins and then diluted with water and brine.
  • Example 13 2-[(2-Amino-4- ⁇ [(3S -l-hvdroxyhexan-3-yllamino ⁇ -6-methylpyrimidin- 5-vDmethyll -5- ⁇ r(2-methoxyethyl)(methyl)aminol methyljphenol
  • the title compound may be prepared by the steps described below:
  • the title compound may be prepared by the steps described below:
  • Methyl 2-[4-(benzyloxy)-2-methoxybenzyl]-3-oxobutanoate (870 mg) was dissolved in CH 3 OH (14 mL) and guanidine hemicarbonate (687 mg) was added. The mixture was then heated to 60°C for 5h and allowed to cool to RT. The mixture was then filtered and washed with diethyl ether and water. The filtrate was concentrated in vacuo and more diethyl ether and water added.
  • Imidazole (94 mg) was added to a solution of (35)-3-( ⁇ 2-amino-5-[4-(benzyloxy)-2- methoxybenzyl]-6-methylpyrimidin-4-yl ⁇ amino)-hexan-l-ol (310 mg) and tert-butyl- dimethylsilyl chloride (207 mg) in DMF (2.5 mL) under an atmosphere of N 2 , and the mixture was stirred 24h. Further tert-butyldimethylsilyl chloride (104 mg) and imidazole (47 mg) were then added and the mixture was stirred for a further 3h.
  • Diisopropyl azodicarboxylate (117 ⁇ ) was added to 4-[(2-amino-4- ⁇ [(35)-l- ⁇ [tert- butyl(dimethyl)silyl]oxy ⁇ hexan-3-yl]amino ⁇ -6-methyl-pyrimidin-5-yl)methyl]-3- methoxyphenol (94 mg), 2-(dimethylamino)ethanol (99 ⁇ ) and triphenylphosphine (156 mg) in THF (2 mL) under N 2 at 0°C.
  • 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 5 o value.
  • 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; and
  • 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.
  • 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).
  • 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.
  • PatchPlate TM 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 TM 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 ⁇ -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 electrophysiology (-82m V) and then making the necessary offset adjustment in the Ion WorksTM 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 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).

Abstract

The invention concerns compounds of Formula (I): wherein R1, R2, R3 and Q are as defined in the description. The present invention also relates to processes for the preparation of such compounds, novel intermediates useful in the preparation of such compounds, pharmaceutical compositions containing them and their use as toll-like receptor 7 agonists.

Description

PHENOL COMPOUNDS ALS TOLL -LIKE RECEPTOR 7 AGONISTS
Field of the Invention
The present invention relates to novel phenol compounds and, more particularly, to novel phenol compounds that act as TLR7 agonists. This invention also relates to methods for the preparation of such compounds and novel intermediates 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.
Background of the Invention
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 (LC)), 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 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.
Summary of the Invention
As now demonstrated herein, the phenol compounds of the present invention are capable of activating TLR7 in vitro. As a consequence of this activity, the phenol 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, some 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, some compounds of the invention also have a surprisingly advantageous hERG profile. Compounds that have significant activities against the hERG ion channel are relevant to inducing QT prolongation and are disfavored 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 provided a compound of Formula (I):
Figure imgf000004_0001
(I)
wherein:
Q is a direct bond, -0-(C2-4alkylene)- or -(Ci_4alkylene)-;
R1 is selected from hydrogen, Ci_4alkyl, Ci_3alkoxyC2-4alkyl, hydroxyC2_4alkyl, C3_5cycloalkyl, tetrahydropyranyl, benzyl, C-linked pyrrolidinyl, C-linked piperidinyl and (Rx)(RY)N-C2-4alkyl, wherein Rx and RY each independently represent hydrogen or Ci_3alkyl; and
R2 is selected from hydrogen, Ci_4alkyl and Ci_3alkoxyC2-4alkyl;
or R1 and R2 together with the nitrogen to which they are attached form a saturated heterocyclyl selected from azetidin-l-yl, pyrrolidin-l-yl, piperidin-l-yl, morpholin-4-yl, piperazin-l-yl, azepan-l-yl, 1,4-diazepan-l-yl; wherein one or two carbon atoms within said heterocyclyl may be substituted by oxo and wherein the available nitrogen of said piperazin-l-yl and 1,4-diazepan-l-yl is optionally substituted by Ci_3alkyl,
hydroxyCi_3alkyl or Ci_2alkoxyC2-3alkyl;
or R1 and R2 together with the nitrogen to which they are attached form a
5-membered aromatic heterocyclyl containing a total of 1-3 nitrogen atoms; wherein said 5-membered aromatic heterocyclyl is optionally substituted on a ring carbon atom by Ci_3alkyl;
R3 is selected from hydrogen, hydroxymethyl and 2-hydroxyethyl;
or a pharmaceutically acceptable salt thereof. For the avoidance of doubt, the "available nitrogen" of a piperazin-l-yl and 1,4- diazepan-l-yl is at the 4-positions of the piperazin-l-yl and the 1,4-diazepan-l-yl.
It is to be understood that, insofar as 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. Similarly, 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_4alkylene" is intended to mean divalent group of a saturated carbon chain of 1 to 4 carbon atoms in length which may be straight-chained or branched, preferably straight chained. For example, "Ci_4alkylene" includes, but is not limited to, methylene, ethylene, trimethylene, tetramethylene. The term "C2-4alkylene"is to be construed accordingly.
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, but is not limited to, methyl, ethyl, propyl, isopropyl, butyl, and tert-butyl. The term "C2_4alkyl" and "Ci_3alkyl" are to be construed accordingly.
The term "Ci_3alkoxyC2-4alkyl" is intended to mean a saturated carbon chain of 1 to 3 carbon atoms in length, which may be straight-chained or branched, linked via oxygen to another saturated carbon chain of 2 to 4 carbon atoms in length, which may be straight- chained or branched. For example, "Ci_3alkoxyC2-4alkyl" includes, but is not limited to, methoxyethyl, methoxypropyl, ethoxypropyl, propoxyethyl and methoxypropyl.
The term "hydroxyC2-4alkyl" is intended to mean a saturated carbon chain of 2 to 4 carbon atoms in length, which may be straight-chained or branched, wherein one of the hydrogen atoms has been replaced by a hydroxy group. For example "hydroxyC2-4alkyl" includes, but is not limited to, hydroxyethyl, 2-hydroxypropyl, 3-hydroxypropyl, 1- hydroxyisopropyl and 4-hydroxybutyl.
The term "(Rx)(RY)N-C2-4alkyl" is intended to mean a saturated carbon chain of 2 to 4 carbon atoms in length, which may be straight-chained or branched, wherein one of the hydrogen atoms has been replaced by a group "(RX)(RY)N-", wherein Rx and RY each independently represent hydrogen or Ci_3alkyl. The "Ci_3alkyl" is intended to mean a saturated carbon chain of 1 to 3 carbon atoms in length which may be straight-chained or branched, including methyl, ethyl, propyl and isopropyl.
In one embodiment there is provided a compound of Formula (I) of Formula (IA):
Figure imgf000006_0001
(IA)
or a pharmaceutically acceptable salt thereof, wherein the values of R1, R2, R3 and Q may take any of the values defined herein R1, R2, R3 and Q respectively.
The variable groups R1, R2, R3 and Q 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.
(1) Q is a direct bond.
(2) Q is -0-(C2-4alkylene)-.
(3) Q is -O-CH2CH2- or -O-CH2CH2CH2-. (4) Q is -(Ci_4alkylene)-.
(5) Q is -CH2- or -CH2CH2CH2-.
(6) Q is -CH2-.
(7) Q is -CH2CH2CH2-.
(8) Q is -0-(C2_4alkylene)- or -(Ci_4alkylene)-.
(9) Q is -0-(C2_4alkylene)- or -(Ci_4alkylene)- wherein said alkylene is a straight chain alkylene.
(10) R1 is hydrogen.
(11) R1 is Ci_4alkyl, Ci_3alkoxyC2_4alkyl, hydroxyC2_4alkyl, C3_5Cycloalkyl,
tetrahydropyranyl, benzyl, C-linked pyrrolidinyl, C-linked piperidinyl and (RX)(RY)N-C2_4alkyl, wherein Rx and RY each independently represent hydrogen or Ci_3alkyl.
(12) R1 is Ci_4alkyl, Ci_3alkoxyC2_4alkyl or hydroxyC2_4alkyl.
(13) R1 is selected from methyl, ethyl, 2-methoxyethyl and 2-hydroxyethyl.
(14) R2 is hydrogen.
(15) R2 is Ci_4alkyl or Ci_3alkoxyC2_4alkyl.
(16) R2 is Ci_4alkyl.
(17) R2 is methyl or ethyl.
(18) R2 is Ci_3alkoxyC2_4alkyl.
(19) R1 and R2 together with the nitrogen to which they are attached form a
saturated heterocyclyl selected from azetidin-l-yl, pyrrolidin-l-yl,
piperidin-l-yl, morpholin-4-yl, piperazin-l-yl, azepan-l-yl, 1,4-diazepan-l-yl; wherein one or two carbon atoms within said heterocyclyl may be substituted by oxo and wherein the available nitrogen of said piperazin-l-yl and
1,4-diazepan-l-yl is optionally substituted by Ci_3alkyl, hydroxyCi_3alkyl or
C i _2alkoxy C2_3 alkyl .
(20) R1 and R2 together with the nitrogen to which they are attached form a
pyrrolidine- 1-yl or piperazin-l-yl wherein said piperazin-l-yl is substituted by Ci_3alkyl.
(21) R1 and R2 together with the nitrogen to which they are attached form a
5-membered aromatic heterocyclyl containing a total of 1-3 nitrogen atoms; wherein said 5-membered aromatic heterocyclyl is optionally substituted on a ring carbon atom by Ci_3alkyl.
(22) R1 is Ci_4alkyl, Ci_3alkoxyC2-4alkyl or hydroxyC2-4alkyl; and R2 is Ci_4alkyl; or R1 and R2 together with the nitrogen to which they are attached form a pyrrolidine- 1-yl or piperazin-l-yl wherein said piperazin-l-yl is substituted by Ci_3alkyl.
(23) R1 is methyl, ethyl, 2-methoxyethyl or 2-hydroxyethyl; and R2 is methyl or ethyl; or R1 and R2 together with the nitrogen to which they are attached form a pyrrolidine- 1-yl or 4-methylpiperazin-l-yl.
(24) R3 is hydrogen.
(25) R3 is hydroxymethyl.
(26) R3 is 2-hydroxyethyl.
(27) R3 is hydroxymethyl or 2-hydroxyethyl.
(28) Q is -0-(C2-4alkylene)- or -(Ci_4alkylene)-;
R1 Ci_4alkyl, Ci_3alkoxyC2-4alkyl or hydroxyC2-4alkyl; and R2 is Ci_4alkyl; or R1 and R2 together with the nitrogen to which they are attached form a pyrrolidine- 1-yl or piperazin-l-yl wherein said piperazin-l-yl is substituted by Ci_3alkyl; and
R3 is selected from hydrogen, hydroxymethyl and 2-hydroxyethyl.
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:
2-[(2-amino-4- { [(35)- 1 -hydroxyhexan-3 -yl] amino } -6-methylpyrimidin-5 - yl)methyl]-5- {[(2-hydroxyethyl)(methyl)amino]methyl}phenol;
2- {[2-amino-4-(butylamino)-6-methylpyrimidin-5-yl]methyl} -5-(pyrrolidin- 1 - ylmethyl)phenol;
2- { [2-amino-4-(butylamino)-6-methylpyrimidin-5 -yljmethyl} -5 - [(4- methylpiperazin- 1 -yl)methyl]phenol;
2-[(2-amino-4- { [(35)- 1 -hydroxyhexan-3 -yl] amino } -6-methylpyrimidin-5 - yl)methyl]-5-[(dimethylamino)methyl]phenol;
2-[(2-amino-4- { [(35)- 1 -hydroxyhexan-3 -yl] amino } -6-methylpyrimidin-5 - yl)methyl] -5 - [(4-methylpiperazin- 1 -yl)methyl]phenol; 2-[(2-amino-4- { [(35)- 1 -hydroxyhexan-3 -yl] amino } -6-methylpyrimidin-5 - yl)methyl] -5 - [(diethylamino)methyl]phenol;
2-[(2-amino-4- { [(35)- 1 -hydroxyhexan-3 -yl] amino } -6-methylpyrimidin-5 - yl)methyl]-5-{[ethyl(methyl)amino]methyl}phenol;
2-[(2-amino-4- { [(25)- 1 -hydroxypentan-2-yl] amino } -6-methylpyrimidin-5 - yl)methyl]-5-[(dimethylamino)methyl]phenol;
2-[(2-amino-4- { [(25)- 1 -hydroxypentan-2-yl] amino } -6-methylpyrimidin-5 - yl)methyl]-5-(pyrrolidin-l-ylmethyl)phenol;
2-[(2-amino-4- { [(25)- 1 -hydroxypentan-2-yl] amino } -6-methylpyrimidin-5 - yl)methyl]-5-[3-(dimethylamino)propyl]phenol;
2-[(2-amino-4- { [(35)- 1 -hydroxyhexan-3 -yl] amino } -6-methylpyrimidin-5 - yl)methyl]-5-[3-(dimethylamino)propyl]phenol;
2-[(2-amino-4- { [(35)- 1 -hydroxyhexan-3 -yl] amino } -6-methylpyrimidin-5 - yl)methyl] -5 - [3 -(dimethylamino)propoxy]phenol;
2-[(2-amino-4- { [(35)- 1 -hydroxyhexan-3 -yl] amino } -6-methylpyrimidin-5 - yl)methyl]-5-{[(2-methoxyethyl)(methyl)amino]methyl}phenol; and
2-[(2-amino-4- { [(35)- 1 -hydroxyhexan-3 -yl] amino } -6-methylpyrimidin-5 - yl)methyl] -5 - [2-(dimethylamino)ethoxy ]phenol;
or a pharmaceutically acceptable salt thereof.
In one embodiment of the invention there is provided any Example described herein, or a pharmaceutically acceptable salt thereof.
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, that is 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 Enzvmology, 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).
Preparation of Compounds of Formula (I)
The compound of Formula (I) may be prepared by reacting a compound of Formula (II), or a salt thereof,
Figure imgf000011_0001
(Π) wherein R1, R2 and Q may take any of the values for R1, R2 and Q as defined herein, and where R4 may take any of the values for R3 as defined herein or R4 may be a protected hydroxyalkyl group: PG1-0-(CH2)n-, wherein n is 1 or 2 and PG1 is a protecting group. In one embodiment n is 1. In another embodiment n is 2. In one embodiment PG1 is selected from a benzyl-based protecting group and a silyl-based protecting group. A benzyl-based protecting group may be benzyl. A silyl-based protecting group may be a tri(Ci_4alkyl)silyl group, for example a triemthylsilyl (TMS) or fert-butyldimethylsilyl (TBDMS) protecting group.
Therefore in one aspect of the invention there is provided a compound of Formula (II), or a salt thereof, as defined herein. The preparation of a compound of Formula (I) from a compound of Formula (II) may be carried out with or without any hydroxy group within R4 being protected. Any protecting group used on a hydroxyl group within R4 may be removed before or after the reaction with BBr3 (the chemistry that reveals the phenol functionality).
Compounds of Formula (II) where Q is -CH2- [a compound of Formula (II- A)] may be prepared as shown in Scheme 1 :
Figure imgf000012_0001
Scheme 1
The R5A group of the compound of Formula (III-A) may be a substituted or unsubstituted hydrocarbyl group. Conveniently R5A may be a hydrocarbyl group selected from benzyl and Ci_4alkyl, for example methyl or ethyl. The LG2A group of the compound of Formula (III-B) 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 (III- A) and Formula (III-B): For example, reaction of a compound of Formula (III-B) 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 (III-A). 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 herein, or a salt thereof.
Step (iv) - Compounds of Formula (VII) may be prepared by reacting a compound of Formula (VI) with an excess of butylamine or 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 (TFA) 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 same R4 values described hereinbefore in connection with the compound of Formula (II). In further embodiments, the compound of Formula (VII) and/or the compound of Formula (VIII) and/or the compound of Formula (II- A), when R4 is other than hydrogen, has an (5)-stereocentre at the asymmetric carbon atom where the R4 group is attached. 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 (II- A) may be prepared by reaction of a compound of (VIII) with the appropriate amine (FINR^2) 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 CH2CI2 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 (VIII) may also be prepared as shown in Scheme 3.
Figure imgf000015_0001
Figure imgf000015_0002
(VIII)
Scheme 3
The R group of the compound of Formula (IX) may be a substituted or unsubstituted hydrocarbyl group. Conveniently R5B may be a hydrocarbyl group selected from benzyl and Ci_4alkyl, for example methyl or ethyl. The LG2B 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 (XI) 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 butylamine or 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 TFA 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 same R4 values described hereinbefore in connection with the compound of Formula (II). In a further embodiment the compound of Formula (XIV) has an (iS)-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.
Alternatively a compound of Formula (II-A) may be prepared as shown in Scheme
4.
The compounds of Formula (XIV- A) and (XV- A) are compounds of Formula (XIV) and (XV) respectively, as hereinbefore defined, wherein R4 is either hydrogen or PG1-0-(CH2)n-, as defined hereinbefore.
In one embodiment there is provided a compound of Formula (XIV- A) as defined hereinbefore wherein R4 is either hydrogen or PG1-0-(CH2)n- wherein n is 1 or 2, and wherein PG1 is a tert-butyldimethylsilyl (TBDMS) protecting group; or a salt thereof.
Step (i) Compounds of Formula (XV- A) may be prepared from compounds of Formula (XIV- A) 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
Figure imgf000018_0001
(XVI)
Scheme 4
Step (ii) 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 (XVI) may be converted into a compound of Formula (II- A) 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), when R4 is other than hydrogen, has an (iS)-stereocentre at the carbon atom to which the R4 group is attached.
Compounds of Formula (II) where Q is -0-(C2_4alkylene)- [compounds of Formula (II-B) may be prepared ac rding to the route shown in Scheme 5.
Figure imgf000019_0001
(XXVII)
Figure imgf000019_0002
(II-B)
Scheme 5
The R7 group in the compound of Formula (XXII) may take any of the values of R7 described herein. For example, R7 may be a 2,4,6-trimethylphenyl group. The PG3 group in the compounds of Formula (XXII) and Formula (XXVII) may be any suitable protecting group for an alcohol, for example a tri(Ci_4alkyl)silyl protecting group, such as TBDMS. Where the R4 group in the compound of Formula (XXVI) is other than hydrogen then the alcohol group within the R4 group is protected by a benzyl-based protecting group PG4, for example with a benzyl group. Step (x): Compounds of Formula (XXVII) may be prepared by reacting a compound of Formula (XXII) with an excess of a compound of Formula (XXVI) in a suitable solvent, such as propionitrile, butanol or 1,4-dioxane, in the presence of TFA at a suitable temperature, for example 50-150°C. Alternatively, the reaction may be performed in a microwave at a suitable temperature, for example 50-200°C.
Step (xi): Compounds of Formula (XXVIII) may be prepared by using the appropriate deprotection conditions for removing the PG3 group. For example, where PG3 is a TBDMS group, the reaction may be carried out using an acid such as hydrochloric acid or TFA, in a suitable solvent, such as 1,4-dioxane or MeOH at a suitable temperature, for example 0-50°C.
Step (xii): Compounds of Formula (XXIX) may be prepared by reacting a compound of Formula (XXVIII) with a hydrocarbyl sulfonyl chloride: R7S02C1, for example mesyl chloride, trimethylbenzene sulfonyl chloride or triisopropylbenzenesulfonyl chloride, in a suitable solvent, such as CH2C12 or THF in the presence of a suitable base, such as metal carbonate base, or an organic base, such as tertiary amine base, for example, triethylamine, at a suitable temperature, for example 0-50°C.
Step (xiii): Compounds of Formula (II-B) may be prepared by reacting a
1 2
compound of Formula (XXIX) with an excess of the appropriate (R )(R )NH compound, in a suitable solvent, such as DMF, acetonitrile or water, and at a suitable temperature, for example 50-150°C, optionally in the presence of an additive such as triethylamine or DIPEA. Alternatively, the reaction may be performed in a microwave at a suitable temperature, for example 50-200°C.
Step (xiv): In cases where the R4 group in the compound of Formula (XXIX) is other than hydrogen, initially formed compounds of Formula (II-B) that have a benzyl- based protecting group within R4 may deprotected using suitable deprotection conditions to provide a compound of Formula (II-B) where R4 is hydroxymethyl or 2-hydroxyethyl. For example, the reaction may be carried out with a catalyst such as Pd/C under H2 atmosphere (1-20 bar) in a suitable solvent, such as MeOH at a suitable temperature, for example 20-100°C, optionally in the presence of an additive such as hydrochloric acid or acetic acid.
The compound of Formula (XXII) may be prepared as shown in Scheme 6.
Figure imgf000021_0001
Figure imgf000021_0002
Figure imgf000021_0003
Figure imgf000021_0004
(XXI) (XXII)
Scheme 6
Step (i): Compounds of Formula (XVII) may be prepared using an alkylation reaction, for example by reacting a compound of Formula (XVI) with (3-bromopropoxy)- tert-butyldimethylsilane in the case where PG3 is TBDMS, in a suitable solvent, such as DMF in the presence of a suitable base, such as K2CO3 at a suitable temperature, for example 0-50°C.
Step (ii): Compounds of Formula (XVIII) may be prepared using any reducing agent that is known to be capable of reducing an aldehyde to the corresponding alcohol, for example by reacting a compound of Formula (XVII) with NaBH4 in a suitable solvent, such as MeOH at a suitable temperature, for example 0°C to 50°C.
Step (iii): Compounds of Formula (XIX) may be prepared by reacting a compound of Formula (XVII) with a hydrocarbylsulfonyl chloride and a salt such as LiCl, in a suitable solvent, such as THF in the presence of a suitable base, such as DIPEA, at a suitable temperature, for example 0-50°C.
Step (iv): Compounds of Formula (XX) can be prepared by reaction of a compound of Formula (III-A), for example, methylacetoacetate, 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 (XIX). The reaction is then preferably heated, for example at 50-100°C, optionally in the presence of an additive such as KI.
Step (v): Compounds of Formula (XXI) may be prepared by reacting a compound of Formula (XX) with guanidine or guanidine carbonate in a suitable solvent, such as MeOH or EtOH, and at a suitable temperature, for example 50-150°C.
Step (vi): Compounds of Formula (XXII) may be prepared by reacting a compound of Formula (XXI) with a suitable 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 tetramethylpropylenediamine (TMPDA) at a suitable temperature, for example 0-50°C.
Compounds of Formula (XXVI) where R4 is other than hydrogen and where the hydroxy group within R4 is protected by a benzyl-based protecting group, may be prepared as shown in Scheme 7.
Figure imgf000022_0001
(XXV) (XXVI)
Scheme 7
Step (vii): Compounds of Formula (XXIV) may be prepared by reacting a compound of Formula (XXIII) with di-fert-butyl dicarbonate. The reaction may be carried out in a suitable solvent, such methanol and water in the presence of a suitable base, such as, NaOH at a suitable temperature, for example 0-50°C.
Step (viii): Compounds of Formula (XXV) may be prepared by reacting a compound of Formula (XXIV) with the appropriate benzyl bromide or chloride in a suitable solvent, such as DMF in the presence of a suitable base, such as DIPEA, at a suitable temperature, for example 0-50°C.
Step (ix) Compounds of Formula (XXVI) may be prepared by reacting a compound of Formula (XXV) with an acid such as hydrochloric acid or TFA in a suitable solvent, such as 1,4-dioxane or MeOH at a suitable temperature, for example 0-50°C.
Compounds of Formula (II) where Q is -CH2CH2CH2- [a compound of Formula -C)] may be prepared as shown in Scheme 8.
Figure imgf000023_0001
Scheme 8 Step (i): Compounds of Formula (XXXIII) can be prepared by reaction of a compound of Formula (XXXII) 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 (III- A) wherein LG4 represents a suitable leaving group, for example bromo or chloro. The reaction is then preferably heated, for example at 50-100°C, optionally in the presence of an additive such as KI.
Step (ii): Compounds of Formula (XXXIV) may be prepared by reacting a compound of Formula (XXXIII) with guanidine or guanidine carbonate in a suitable solvent, such as MeOH or EtOH, and at a suitable temperature, for example 50-150°C.
Step (iii): Compounds of Formula (XXXV) may be prepared by reacting a compound of Formula (XXXIV) with a 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 TMPDA or DABCO at a suitable temperature, for example 0-50°C.
Step (iv): Compounds of Formula (XXVI) may be prepared by reacting a compound of Formula (XXXV) with a suitable acrylamide such as N,N-dimethyl- acrylamide. The reaction may be carried out in a suitable base such as triethylamine in the presence of palladium (II) acetate and tri-ort/zo-tolylphosphine at a suitable temperature for example 70-80°C.
Step (v): Compounds of Formula (XXXVII) may be prepared by treating a compound of Formula (XXXVI) with H2 in the presence of palladium on carbon, in a suitable solvent such as ethanol or EtOAc.
Step (vi): Compounds of Formula (XXXVIII) may be prepared by reacting a compound of Formula (XXXVII) with an excess of the appropriate amine, in a suitable solvent, such as propionitrile, butanol, butyronitrile or 1,4-dioxane, in the presence of TFA at a suitable temperature, for example 50-150°C. Alternatively, the reaction may be performed in a microwave at a suitable temperature, for example 50-100°C.
Step (vii): Compound of Formula (VI V) may be prepared by reacting a compound of Formula (XXXVIII) with BH3-dimethyl sulphide complex in a suitable solvent such as THF at a suitable temperature, for example 60°C. Compounds of Formula (II-B) may also be prepared as shown in Scheme 9.
Figure imgf000025_0001
Scheme 9
Step (i): Compounds of a Formula (XLIII) may be prepared by reacting compounds of Formula (XLI) and (XLII) in a suitable solvent such as DMF in the presence of Pd-118 (1,1 -bis(di-tert-butylphosphino)-ferrocenepalladium(II) dichloride), tetrabutylammonium chloride, hydrate and N-cyclohexyl-N-methylcyclo-hexanamine at a sutiable temperature, for example 100°C. LG5 is a leaving group, for example a chloro or bromo. PG5 is a suitable protecting group, for example a benzyl-based protecting group, for example benzyl.
Step (ii): Compounds of Formula (XLIV) may be prepared by reacting a compound of Formula (XLIII) with guanidine or guanidine carbonate, in a suitable solvent, such as MeOH or EtOH, and at a suitable temperature, for example 50-150°C.
Step (iii): Compounds of Formula (XLV) may be prepared by reacting a compound of Formula (XLIV) with POCI3, at a suitable temperature, for example 50-110°C. Step (iv): Compounds of Formula (XL VI) may be prepared by reacting a compound of Formula (XLV) with an excess of the appropriate amine, in a suitable solvent, such as propionitrile ,butanol or 1,4-dioxane, in the presence of TFA at a suitable temperature, for example 50-150°C. Alternatively, the reaction may be performed in a microwave at a suitable temperature, for example 50-200°C.
Step (v): -Compounds of Formula (XL VII) may be prepared by reacting a compound of Formula (XL VI) with a suitable protecting group. For example PG6 may be a tri(Ci_4alkyl)silyl group, for example TBDMS or TMS. In the case where PG6 is a silicon based protecting group, it may be added using, for example TBDMSCl, or TMSCl, in the presence of a suitable base, for example imidazole, in a suitable solvent such as DMF.
Step (vi): Compounds of Formula (XL VIII) may be prepared by using standard deprotection conditions that are known to be suitable for removing a PG5 group. For example, when PG5 is benzyl, this could involve stirring compounds of a Formula
(XL VII) in a suitable solvent such as ethanol or EtOAc in the presence of H2 and palladium on carbon.
Step (vii): Compounds of Formula (XLIX) may be prepared by reacting a compound of a Formula (XL VIII) with the appropriate amino alcohol, in a suitable solvent such as THF using Mitsunobu conditions, for example using diisopropyl azodicarboxylate and tripheny lphosphine .
Step (viii): Compounds of Formula (XXXI) may be prepared by using appropriate deprotection conditions known to be suitable for removing the PG6 group. For example, if PG6 were a silicon-based protecting group, then the deprotection could involve reaction of a compound of Formula (XLIX) with a fluoride salt, for example tetra-n-butylammonium fluoride (TBAF), in a suitable solvent such as CH2C12. Alternatively, such a deproection may be performed using SCX resin with a suitable solvent such as acetonitrile.
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. The intermediates having an R4 group where R4 is other than hydrogen have a chiral centre at the carbon atom to which the R4 group is attached. In further embodiments there is provided each of such intermediates, or a salt thereof, wherein the intermediate has the (S)- stereochemical configuration.
It will be appreciated by those skilled in the art that in the processes of the present invention certain functional groups such as hydroxyl or amino groups in the reagents may need to be protected by protecting groups. Thus, the preparation of the compounds of Formula (I) may involve, at an appropriate stage, the removal of one or more protecting groups.
The protection and deprotection of functional groups is described in 'Protective Groups in Organic Chemistry', edited by J.W.F. McOmie, Plenum Press (1973) and 'Protective Groups in Organic Synthesis', 3rd edition, T.W. Greene and P.G.M. Wuts, Wiley-Interscience (1999).
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 hydrofluoroalkane, 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/mz 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 a 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 a 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
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, miriplatin, carboplatin, 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-l-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) 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 (insulinlike growth factor) kinase inhibitors; aurora kinase inhibitors (for example AZD1152, PH739358, VX-680, MLN8054, R763, MP235, MP529, VX-528 AND AX39459), 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 inter leukin 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) 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.
Chemical Synthesis and Biological Assays
In this specification, the following abbreviations may be used: "EtOAc" = ethyl acetate; "EtOH" = ethanol; "min(s)" = minute(s); "THF" = tetrahydrofuran; "h" = hour(s);
"DABCO" = l,4-diazabicyclo[2.2.2]octane; "FCC" = flash column chromatography using silica; "TFA" = TFA; "DIPEA" = diisopropylethylamine; "DMSO" = dimethylsulfoxide; "sat." = saturated aqueous solution; "aq" = aqueous; "eq." = molar equivalents; "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" = molar. Proton nuclear magnetic resonance data ("1H 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; "ddd" = doublet of doublet of doublets; "dt" = doublet of triplets. Preparative reverse phase high performance liquid chromatography ("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 : 2- Γ(2-Αηιΐηο-4-{ [(3S)-l-hvdroxyhexan-3-vH amino}-6-methylpyrimidin-5- vDmethyll -5-{ [(2-hydr oxyethyl)(methyl)aminol methyljphenol
Figure imgf000039_0001
The title compound may be prepared by the steps described below: (i) Ethyl 2-(4-cvano-2-methoxybenzyl)-3-oxobutanoate
Figure imgf000040_0001
NaH (60% in mineral oil, 4.42 g) was added portion-wise over lOmins to a solution of ethyl acetoacetate (13.48 mL) in THF (60 mL) at 0°C and the resulting suspension was stirred at 0°C for lOmins. Then, a solution of 4-(bromomethyl)-3-methoxybenzonitrile (20 g) in THF (40 mL) was added portion- wise over lOmins, and 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 30mins. The resulting aqueous mixture was extracted with EtOAc (3 x 200 mL). The combined organic solutions 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 21 '4 APCI-.
(ii) 4-[(2-Amino-4-hydroxy-6-methylpyrimidin-5-yl)methyll-3-methoxybenzonitrile
Figure imgf000040_0002
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 was then neutralized by the addition of acetic acid. The resulting precipitate was collected by filtration and the solid was 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 271 APCI+.
(iii) 4-[(2-Amino-4-chloro-6-meth lpyrimidin-5-yl)methyll-3-methoxybenzonitrile
Figure imgf000040_0003
4-[(2-Amino-4-hydroxy-6-methylpyrimidin-5 -yl)methyl] -3 -methoxybenzonitrile (2.3 g) was added to POCI3 (20 mL) and the suspension was heated at 100°C for 16h. The remaining POCI3 was removed in vacuo and the resulting residue was diluted with ice/water (20 mL). The pH of the suspension was adjusted to ~7 by the addition of NaHCC>3 and this mixture was heated at 50°C for lh. The resulting solid was collected by filtration and dried under vacuum to give the sub-title compound (2.2 g) as a white solid; LC-MS: m/z 289 APCI+
(iv) 4- IY2- Amino-4- { \(3S)- 1 -hydroxyhexan-3 -yll amino) -6-methylpyrimidin-5 -vDmethyll - 3 -methoxybenzonitrile
Figure imgf000041_0001
(5)-3-Aminohexan-l-ol (804 mg) was combined with 4-[(2-amino-4-chloro-6- methylpyrimidin-5-yl)methyl]-3-methoxybenzonitrile (660 mg) in 1,4-dioxane (5 mL) and the mixture was stirred and heated in a CEM Discover™ microwave at 180°C for 9h. The solvents were then 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 -yl] amino) -6-methylpyrimidin-5 -yOmethyl] - 3 -methoxybenzaldehyde
Figure imgf000041_0002
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 mixture was stirred for 16h at 45°C before cooling to RT. The solids were removed by filtration and washed with acetic acid (1 mL). The resulting solution was diluted with CH2CI2 (150 mL) and was washed with water (x2), dried (MgS04), and concentrated in vacuo. The residue was azeotroped (x3) with toluene to give the sub-title compound (400 mg) as a yellow foam; LC-MS: m/z 373 APCI+.
(vi) (35V3- (r2-Amino-5-(2-methoxy-4- (Γ(2- methoxyethyl)(methyl)aminolmethyl|benzyl)-6-methylpyrimidin-4-yllamino|hexan-l-ol
Figure imgf000042_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 -hydroxyhexan-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 (3 A) were added. The mixture was cooled to 0°C and sodium triacetoxyborohydride (156 mg) was added and then the mixture was stirred for 3h. The mixture was diluted with CH2CI2 and CH3OH was then added. Purification by FCC eluting with 10% CH3OH in CH2C12 with 1% 7M NH3/CH3OH gave the sub-title compound (105 mg) as a gum; LC-MS: m/z 446 Multimode+.
(vii) 2- IY2- Amino-4- { IY351)- 1 -hydroxyhexan-3 -yll amino) -6-methylpyrimidin-5 -vDmethyll- 5-(r(2-hvdroxyethyl)(methyl)amino1methyl|phenol
Figure imgf000042_0002
A solution of BBr3 (1M in CH2CI2, 1010 μί) was added drop-wise to a solution of (35)-3- {[2-Amino-5-(2-methoxy-4-{[(2-methoxyethyl)(methyl)amino]methyl}benzyl)-6- methylpyrimidin-4-yl]amino}hexan-l-ol (90 mg) in CH2CI2 (10 mL), and the mixture was stirred at RT for 16h. CH3OH (4 mL) was added and the mixture stirred for 30mins. HC1 (4M in 1,4-dioxane, 3 mL) was added. The mixture was stirred for 30mins and the solvents were then removed in vacuo. The resulting residue was dissolved in CH3OH, neutralised with NH3 and the crude product was purified by RPHPLC to give the title compound (26 mg) as a gum; LC-MS: m/z 418 APCI+. Example 1A: 2-[(2-Amino-4-{[(3S -l-hvdroxyhexan-3-yllamino}-6-methylpyrimidin- 5-yl)methyll-5-{[(2-hvdroxyethyl)(methyl)aminolmethyl}phenol bis saccharin salt
2-[(2-Amino-4- {[(35)- 1 -hydroxyhexan-3-yl]amino} -6-methylpyrimidin-5-yl)methyl]-5- {[(2-hydroxyethyl)(methyl)amino]methyl}phenol (preparation: see step (vii) of Example 1, above; 26 mg) was dissolved in CH3CN. Then saccharin (2 eq.) was added to give a solution. The solvent was removed by blowing N2 over the solution. The solid was then dried under vacuum to give the title compound (50 mg) as a white solid; 1H NMR: 11.89 (s, 1H), 10.32 (s, 1H), 9.38 (s, 1H), 7.62 (d, 8H), 7.37 (s, 2H), 7.00-6.83 (m, 3H), 5.34 (s, 1H), 4.36 (s, 2H), 4.28-4.10 (m, 2H), 3.71 (s, 4H), 3.16 (s, 2H), 3.05 (d, 2H), 2.67 (d, 4H), 2.23 (s, 3H), 1.69-1.53 (m, 2H), 1.24 (s, 1H), 1.16-1.05 (m, 2H), 0.79 (t, 3H); LC-MS: m/z 418 multimode+.
Example 2 : 2- { [2- Amino-4-(butylamino)-6-methylpyrimidin-5-yll methyl}-5-
(pyrrolidin-l-ylmethyl)phenol
Figure imgf000043_0001
The title compound may be prepared by the steps described below:
(i) 4-{[2-Amino-4-(butylamino)-6-meth lpyrimidin-5-yllmethyl|-3-methoxybenzonitrile
Figure imgf000043_0002
Butylamine (1.26 g; 5eq) was reacted using the general method described for step (iv) of Example 1, and with heating for 3h. Purification by FCC, eluting with 0-20% CH3OH (and 0.7M NH3) in CH2C12 gave the sub-title compound (0.7 g) as an oil; LC-MS: m/z 326 APCI+.
(ii) 4- {r2-Amino-4-(butylamino)-6-methylpyrimidin-5-yl1methyl|-3- methoxybenzaldehyde
Figure imgf000043_0003
4-{[2-Amino-4-(butylamino)-6-methylpyrimidin-5-yl]methyl}-3-methoxybenzonitri (0.7 g) was reacted using the general method described for step (v) of Example 1 to give the sub-title compound (0.7 g) as a yellow foam; LC-MS: m/z 329 APCI+.
(iii) A4-Butyl-5-[2-methoxy-4-(pyrrolidin-l-ylmethyl)benzyll-6-methylpyrimidine-2,4- diamine
Figure imgf000044_0001
4-{[2-Amino-4-(butylamino)-6-methylpyrimidin-5-yl]methyl}-3-methoxybenzaldehyde (400 mg) and pyrrolidine (260 mg) were reacted together using the general method described for step (vi) of Example 1. After stirring for 3h the solvent was removed in vacuo and CH3OH was added. Purification by RPHPLC gave the sub-title compound (150 mg) as an oil; LC-MS: m/z 384 APCI+.
(iv) 2- { r2-Amino-4-(butylamino)-6-methylpyrimidin-5 -yllmethyll -5 -(pyrrolidin- 1 - ylmethvDphenol
Figure imgf000044_0002
N4-Butyl-5-[2-methoxy-4-(pyrrolidin-l-ylmethyl)benzyl]-6-methylpyrimidine-2,4-diamine (150 mg) was reacted using the general method described for step (vii) of Example 1 to give the title compound (27 mg) as a white solid; 1H NMR: 9.62 (s, 1H), 6.80 (d, 1H), 6.68 (d, 1H), 6.59 (dd, 1H), 6.04 (s, 1H), 5.61 (d, 2H), 3.55 (s, 2H), 3.40 (d, 2H), 3.23 (dd, 2H), 2.36 (d, 4H), 2.07 (s, 3H), 1.72-1.61 (m, 4H), 1.46-1.37 (m, 2H), 1.24-1.13 (m, 2H), 0.82 (t, 3H); LC-MS: m/z 370 APCI+.
Example 3 : 2- { [2- Amino-4-(butylamino)-6-methylpyrimidin-5-yll methyl}-5- \(4- methylpiperazin-l-vDmethyllphenol
Figure imgf000044_0003
The title compound may be prepared by the steps described below:
(i) A^-Butyl-S- {2-methoxy-4-[(4-methylpiperazin- 1 -yl)methyl]benzyl| -6- methylpyrimidine-2 ,4-diamine
Figure imgf000045_0001
4-{[2-Amino-4-(butylamino)-6-methylpyrimidin-5-yl]methyl}-3-methoxybenzaldehyde (preparation: see step (ii) of Example 2, above; 200 mg) and 1-methylpiperazine (183 mg) were reacted together using the general method described for step (vi) of Example 1. After stirring for 16h the solvent was removed in vacuo and CH3OH added. Purification by RPHPLC gave the sub-title compound (31 mg) as a white solid; 1H NMR: 6.89 (s, 1H), 6.72 (d, 1H), 6.66 (d, 1H), 5.95 (s, 1H), 5.65 (s, 1H), 3.83 (s, 2H), 3.58 (s, 2H), 3.41-3.36 (m, 2H), 3.31 (s, 4H), 3.22 (dd, 2H), 2.36 (s, 2H), 2.13 (s, 3H), 1.98 (s, 3H), 1.44-1.35 (m, 2H), 1.24 (s, 2H), 1.21-1.10 (m, 2H), 0.89-0.78 (m, 3H); LC-MS: m/z 413 APCI+.
(ii) 2-{[2-Amino-4-(butylamino)-6-methylpyrimidin-5-yllmethyl|-5-[(4-methylpiperazin- 1 -yl)methyl]phenol
Figure imgf000045_0002
N4-Butyl-5-{2-methoxy-4-[(4-methylpiperazin-l-yl)methyl]benzyl}-6-methylpyrimidine- 2,4-diamine (100 mg) was reacted using the general method described in step (vii) of Example 1 to give the title compound (15 mg) as a white solid; 1H NMR: 9.63 (s, 1H), 6.79 (s, 1H), 6.68 (d, 1H), 6.57 (d, 1H), 6.02 (s, 1H), 5.61 (d, 2H), 3.54 (s, 2H), 3.31 (s, 2H), 3.29 (s, 2H), 3.23 (dd, 2H), 2.41-2.20 (m, 6H), 2.14 (d, 3H), 2.07 (s, 3H), 1.44-1.35 (m, 2H), 1.16 (dt, 2H), 0.88-0.77 (m, 3H); LC-MS: m/z 399 APCI+.
Example 4: 2-[(2-Amino-4-{[(3S -l-hvdroxyhexan-3-yllamino}-6-methylpyrimidin-5- vDmethyll -5- [(dimethylamino)methyll phenol
Figure imgf000046_0001
The title compound may be prepared by the steps described below:
(i) 4- \(2- Amino-4- { \(3S)- 1 -hydroxyhexan-3 -yll amino) -6-methylpyrimidin-5 -vDmethyll -3 - methoxybenzonitrile
Figure imgf000046_0002
(5)-3-Aminohexan-l-ol (264 mg, 1.3 eq.) was reacted using the general method described in step (iv) of Example 1 , 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- [(2- Amino-4- { \(3S)- 1 -hydroxyhexan-3 -yl] amino) -6-methylpyrimidin-5 -yQmethyl]- 3 -methoxybenzaldehyde
Figure imgf000046_0003
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 1 , with stirring for 3h, to give the sub-title compound (350 mg) as a yellow foam; LC-MS: m/z 373 APCI+.
(iii) 2- IY2- Amino-4- { \(3S)- 1 -hydroxyhexan-3 -yll amino) -6-methylpyrimidin-5 -vDmethyll-
5- [(dimethylamino)methyl]phenol
Figure imgf000047_0001
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 1. After stirring for 16h the solvent was removed in vacuo and CH3OH was added. Purification by RPHPLC gave a gum (35 mg). This material was reacted with BBr3 (1M in CH2CI2, 2 mL) using the general method described for step (vii) of Example 1, with stirring for lh, to give the title compound (16 mg) as a solid; 1H NMR: 9.69 (s, 1H), 6.78 (s, 1H), 6.74 (d, 1H), 6.59 (d, 1H), 5.65 (t, 2H), 4.41-4.33 (m, 1H), 4.20-4.11 (m, 1H), 3.56 (s, 2H), 3.24 (d, 3H), 2.11 (d, 3H), 2.08 (d, 6H), 1.59 (dt, 1H), 1.49-1.19 (m, 5H), 1.14-0.97 (m, 2H), 0.75 (t, 3H); LC-MS: m/z 388 APCI+.
Example 5: 2-[(2-Amino-4-{[(3S -l-hvdroxyhexan-3-yllamino}-6-methylpyrimidin-5- yl)methyll-5-[(4-methylpiperazin-l- l)methyllphenol
Figure imgf000047_0002
The title compound may be prepared by the steps described below:
(i) (361-3 - r(2-amino-5 - (2-methoxy-4-r(4-methylpiperazin- 1 -vDmethvHbenzyl} -6- methylpyrimidin-4-yl)amino1hexan- 1 -ol
Figure imgf000047_0003
4-[(2-Amino-4- {[(3S)- 1 -hydroxyhexan-3-yl]amino} -6-methylpyrimidin-5-yl)methyl]-3- methoxybenzaldehyde (preparation: see step (ii) of Example 4, above; 120 mg) and 1- methylpiperazine (97 mg) were reacted together using the general method described in step (vi) of Example 1. After stirring for 16h the solvent was removed in vacuo and CH3OH was added. Purification by RPHPLC gave the sub-title compound (50 mg) as a white solid; LC-MS: m/z 457 APCI+.
(ii) 2- [(2- Amino-4- { \(3S)- 1 -hydroxyhexan-3 -yl] amino) -6-methylpyrimidin-5 -yQmethyl]- 5-r(4-methylpiperazin- 1 -vDmethyllphenol
Figure imgf000048_0001
(35)-3-[(2-amino-5- {2-methoxy-4-[(4-methylpiperazin- 1 -yl)methyl]benzyl} -6- methylpyrimidin-4-yl)amino]hexan-l-ol (50 mg) was reacted using the general method described in step (vii) of Example 1 to give the title compound (15 mg) as a white solid; 1H NMR: 9.70 (s, IH), 6.79 (s, IH), 6.75 (d, IH), 6.59 (d, IH), 5.64 (d, 2H), 4.38 (s, IH), 4.14 (dt, IH), 3.55 (s, 2H), 3.31 (s, 2H), 3.29 (s, 2H), 3.23 (d, IH), 2.34 (dd, 6H), 2.12 (s, 6H), 1.58 (dt, IH), 1.37 (m, 3H), 1.05 (m, 2H), 0.74 (t, 3H). LC-MS: m/z 443 APCI+.
Example 6: 2-[(2-Amino-4-{[(3S -l-hvdroxyhexan-3-yllamino}-6-methylpyrimidin-5- vDmethyll -5- [(diethylamino)meth ll phenol
Figure imgf000048_0002
The title compound may be prepared by the steps described below:
(i) 2-Amino-5-(4-cyano-2-methoxybenzyl)-6-methylpyrimidin-4-yl 2,4,6-tri(propan-2-yl)- benzenesulfonate
Figure imgf000048_0003
4-[(2-Amino-4-hydroxy-6-methylpyrimidin-5-yl)methyl]-3-methoxybenzonitrile
(preparation: see step (ii) of Example 1, 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 (MgS04), 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) -6-methylpyrimidin-5 -yQmethyl]- 3 -methoxybenzonitrile
Figure imgf000049_0001
2-Amino-5-(4-cyano-2-methoxybenzyl)-6-methylpyrimidin-4-yl 2,4,6-tri(propan-2-yl)- benzenesulfonate (g), (5)-3-aminohexan-l-ol (1.965 g) and TFA (419 μί) 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) -6-methylpyrimidin-5 -yQmethyl]- 3 -methoxybenzaldehyde
Figure imgf000049_0002
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 1 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 imgf000050_0001
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 1. 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+.
(v) 2- |Y2-Amino-4- { \(3S)- 1 -hydroxyhexan-3 -yl] amino) -6-methylpyrimidin-5 -yDmethyl] -
5- r(diethylamino)methyl1phenol
Figure imgf000050_0002
(35)-3-[(2-Amino-5-{4-[(diethylamino)methyl]-2-methoxybenzyl}-6-methylpyrimidin-4- yl)amino]hexan-l-ol (200 mg) was reacted using the general method described in step (vii) of Example 1 to give the title compound (18 mg) a white solid; 1H NMR: 9.66 (s, 1H), 6.83 (s, 1H), 6.74 (d, 1H), 6.60 (d, 1H), 5.67 (d, 2H), 4.38 (d, 1H), 4.14 (dt, 1H), 3.55 (s, 2H), 3.40-3.36 (m, 2H), 3.31 (s, 2H), 2.43-2.35 (m, 4H), 2.13 (s, 3H), 1.59 (td, 1H), 1.37 (m, 3H), 1.24 (s, 1H), 1.13-1.01 (m, 2H), 0.94 (t, 6H), 0.75 (t, 3H); LC-MS: m/z 416 multimode+. Example 7: 2-[(2-Amino-4-{[(3S -l-hvdroxyhexan-3-yllamino}-6-methylpyrimidin-5- yl)methyll-5-{[ethvKmethyl)amin methyl}phenol
Figure imgf000051_0001
The title compound may be prepared by the steps described below:
(i) (3S)-3- i Γ2-Αηιίηο-5 -(4- { rethylfmethvOaminolmethyl) -2-methoxybenzyl)-6- methylpyrimidin-4-yl] amino) hexan- 1 -ol
Figure imgf000051_0002
4-[(2 -Amino-4- {[(35)- 1 -hydroxyhexan-3-yl]amino} -6-methylpyrimidin-5-yl)methyl]-3- methoxybenzaldehyde (preparation: see step (iii) of Example 6; 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+.
(ii) 2- [(2- Amino-4- { \(3S)- 1 -hydroxyhexan-3 -yl] amino) -6-methylpyrimidin-5 -yQmethyl]- 5 - ( rethyl(methyl)aminolmethyl) phenol
Figure imgf000051_0003
(3iS)-3 - { [2- Amino-5 -(4- { [ethyl(methyl)amino]methyl} -2-methoxybenzyl)-6-methyl- pyrimidin-4-yl]amino}hexan-l-ol (160 mg) was reacted using the general method described in step (vii) of Example 1 to give the title compound (41 mg) as a white solid; 1H NMR: 9.68 (s, 1H), 6.80 (s, 1H), 6.74 (d, 1H), 6.58 (t, 1H), 5.65 (t, 2H), 4.37 (d, 1H), 4.20- 4.10 (m, 1H), 3.60-3.53 (m, 2H), 3.31 (d, 4H), 2.38-2.27 (m, 3H), 2.12 (s, 3H), 2.05 (s, 3H), 1.59 (td, 1H), 1.48-1.31 (m, 3H), 1.12-1.03 (m, 2H), 1.02-0.93 (m, 3H), 0.75 (t, 3H); LC-MS: m/z 402 multimode+.
Example 8: 2-[(2-Amino-4-{[(2S -l-hvdroxypentan-2-yllamino}-6-methylpyrimidin-5- vDmethyll -5- [(dimethylamino)methyll phenol
Figure imgf000052_0001
The title compound may be prepared by the steps described below:
(i) 4- [(2- Amino-4- i\(2S)- 1 -hydroxypentan-2-yllaminol -6-methylpyrimidin-5-yl)methyl1- 3 -methoxybenzonitrile
Figure imgf000052_0002
(5)-2-Aminopentan-l-ol (2.88 g) was reacted using the general method described in step (ii) of Example 6. 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 CH2CI2 gave the sub-title compound (2.7 g) as a yellow gum; LC-MS: m/z 356 APCI+. (ii) 4-[(2 -Amino-4- {[(261-1 -hydroxypentan-2-yllamino|-6-methylpyrimidin-5-yl)methyll- 3 -methoxybenzaldehyde
Figure imgf000052_0003
4-[(2 -Amino-4- {[(25)- 1 -hydroxypentan-2-yl]amino} -6-methylpyrimidin-5-yl)methyl]-3- methoxybenzonitrile (2.7 g) was reacted using the general method described in step (v) of Example 1 to give the sub-title compound (2.4 g) as a yellow foam; LC-MS: m/z 359
APCI+. (iii) (26 -2-r(2-Amino-5- (4-r(dimethylamino)methyl1-2-methoxybenzyl| -6- methylpyrimidin-4-yl)aminolpentan- 1 -ol
Figure imgf000053_0001
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 using the general method described in step (vi) of Example 1. After stirring for 16h the solvent was removed in vacuo and CH3OH and DMSO were added. Purification by RPHPLC gave the title compound (87.6 mg) as a colourless gum; 1H NMR: 11.92 (s, IH), 9.52 (s, IH), 7.67-7.54 (m, 7H), 7.13 (s, IH), 6.93 (s, 2H), 4.75 (t, IH), 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); LC-MS: m/z 388 multimode+.
(iv) 2-[(2-Amino-4- {[(261-1 -hydroxypentan-2-yllamino|-6-methylpyrimidin-5-yl)methyll- 5-r(dimethylamino)methyl1phenol
Figure imgf000053_0002
(25)-2-[(2-Amino-5-{4-[(dimethylamino)methyl]-2-methoxybenzyl}-6-methylpyrimidin- 4-yl)amino]pentan-l-ol (80 mg) was reacted using the general method described in step
(vii) of Example 1 to give the title compound (16 mg) as a white solid; H NMR: 6.78 (s,
IH), 6.74 (d, IH), 6.58 (d, IH), 4.04 (d, IH), 3.54 (d, 2H), 3.26-3.19 (m, 3H), 2.13 (s, 3H), 2.08 (d, 6H), 1.50 (ddd, 2H), 1.27 (ddd, 3H), 1.13-0.97 (m, 2H), 0.74 (t, 3H); LC-MS: m/z 374 Multimode+.
Example 9: 2-[(2-Amino-4-{[(2S -l-hvdroxypentan-2-yllamino}-6-methylpyrimidin-5- yl)methyll-5-(pyrrolidin-l-ylmethyl)phenol
Figure imgf000054_0001
The title compound may be prepared by the steps described below:
(i) (2S)-2-( (2- Amino-5 - r2-methoxy-4-(pyrrolidin- 1 -ylmethvDbenzvH -6-methylp yrimidin- 4-ylj amino)pentan- 1 -ol
Figure imgf000054_0002
Pyrrolidine (280 μί) was reacted using the general method described in step (iii) of Example 8 to give the sub-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), 4.1 1-4.04 (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), 1.1 1-0.96 (m, 2H), 0.75 (t, 3H); LC-MS: m/z 414 multimode+ (ii) 2-[(2 -Amino-4- {[(261-1 -hydroxypentan-2-yllamino|-6-methylpyrimidin-5-yl)methyll- 5 -(pyrrolidin- 1 -y lmethy Dphenol
Figure imgf000054_0003
(2iS)-2-( {2- Amino-5 - [2-methoxy-4-(pyrrolidin- 1 -ylmethyl)benzyl] -6-methylpyrimidin-4- yl}amino)pentan-l-ol (100 mg) was reacted using the general method described for step (vii) of Example 1 to give the title compound (40 mg) as a white solid; 1H NMR: 9.65 (s, 1H), 6.80 (s, 1H), 6.73 (d, 1H), 6.58 (t, 1H), 5.65-5.54 (m, 3H), 4.06 (t, 1H), 3.55 (s, 2H), 3.41 (s, 2H), 3.24 (dd, 1H), 2.37 (s, 4H), 2.13 (s, 3H), 1.66 (s, 4H), 1.27 (ddd, 2H), 1.13- 0.96 (m, 2H), 0.75 (t, 3H); LC-MS: m/z 400 multimode+.
Example 10: 2-[(2-Amino-4-{[(2S -l-hvdroxypentan-2-yllamino}-6-methylpyrimidin-
5-vDmethyll -5- [3-(dimethylamino)propyll phenol
Figure imgf000055_0001
The title compound may be prepared by the steps described below:
(i) (4-Bromo-2-methoxyphenyl)methanol
Figure imgf000055_0002
L1BH4 (0.66 g) was added portion- wise to a solution of methyl 4-bromo-2- methoxybenzoate (5 g) in dry THF (50 mL) and EtOH (10 mL) was added. The reaction was stirred for 18h. The mixture was then poured into aq. NaOH (1M, 300 mL) and extracted with EtOAc. The organics were combined, washed with water then sat. brine before being dried (MgS04), and concentrated in vacuo to give the sub-title compound (4.4 g) as a gum; 1H NMR: 7.30 (d, 1H), 7.17-7.11 (m, 2H), 5.10 (t, 1H), 4.43 (d, 2H), 3.79 (s, 3H).
(ii) 4-Bromo- 1 -(bromomethvD-2-methoxybenzene
Figure imgf000055_0003
PBr3 (0.95 mL) was added drop-wise to a vigorously stirred solution of (4-bromo-2- methoxyphenyl)methanol (4.4 g) in CH2CI2 (100 mL) at 0°C. After 40 mins the mixture was slowly quenched with ice/water and the organic solution was separated and the aqueous phase was extracted with CH2CI2. The organic solutions were combined, washed with water, dried (MgS04), and concentrated in vacuo to give the sub-title compound (5.2 g); 1H NMR: 7.19 (d, 1H), 7.07 (dd, 1H), 7.01 (d, 1H), 4.49 (s, 2H), 3.89 (s, 3H).
(iii) Ethyl 2-(4-bromo-2-methoxybenz l)-3-oxobutanoate
Figure imgf000055_0004
NaH (0.92 g, 60% in oil) was added portion- wise over 10 mins to a solution of the ethyl 3- oxobutanoate (2.9 g) in THF (60 mL) at 0°C and the mixture was stirred for 10 mins. A solution of 4-bromo-l-(bromomethyl)-2-methoxybenzene (5.2 g) in THF (40 mL) was added portion- wise over 10 mins and the mixture was warmed to 70°C and the stirred for 16h. The mixture was allowed to cool and then poured into ice/water (300 mL) and stirred for 30 mins. The aqueous mixture was extracted with EtOAc and the combined organic extracts were dried and concentrated in vacuo. Purification by FCC, eluting with 10% EtOAc in isohexane gave the sub-title compound as a solid (4 g); LC/MS: m/z 328 APCI+. (iv) 2-Amino-5-(4-bromo-2-methoxybenzyl)-6-methylpyrimidin-4-ol
Figure imgf000056_0001
Guanidine hemicarbonate (2.17 g) was added to a solution of ethyl 2-(4-bromo-2- methoxybenzyl)-3 -oxobutanoate (3.97 g) in CH3OH (90 mL). The resulting mixture was stirred at 65°C for 16h and then allowed to cool. Water (150 mL) was added and the mixture neutralised by addition of acetic acid. The mixture was stirred in an ice bath for 15 mins and the resulting precipitate was collected by filtration and the solid washed with CH3OH (20 mL) to provide the sub-title compound as a solid (2.38 g); LC/MS: m/z 325 APCI+.
(v) 2-Amino-5-(4-bromo-2-methoxybenzyl)-6-methylpyrimidin-4-yl 2,4,6- triisopropylbenzenesulfonate
A mixture of 2-amino-5-(4-bromo-2-methoxybenzyl)-6-methylpyrimidin-4-ol (2.38 g), 2,4,6-triisopropylbenzene-l-sulfonyl chloride (2.89 g) and DABCO (1.07g) in THF (100 mL) was heated at 30°C for 7h. The mixture was then partitioned between EtOAc and brine, the phases were separated and the organic solution was dried (Na2S04) and concentrated in vacuo to give a solid. This solid was tritrurated with isohexane (60 mL) and diethyl ether (2 mL) to give the sub-title compound (3.87 g) as a solid; LC-MS: m/z 591 APCI+.
(vi) (E)-2- Amino-5 - (4-[3 -(dimethylamino)-3 -oxoprop- 1 -enyl] -2-methoxybenzyl| -6- methylpyrimidin-4-yl 2,4,6-triisopropylbenzenesulfonate
Figure imgf000057_0001
A solution of 2-amino-5-(4-bromo-2-methoxybenzyl)-6-methylpyrimidin-4-yl 2,4,6- triisopropylbenzenesulfonate (3.87 g), and N,N-dimethylacrylamide (0.65 g) in
triethylamine (25 mL) was treated with palladium (II) acetate (74 mg) and txi-ortho- tolylphosphine (190 mg) and the mixture was then heated at relux for 24h. The solvent was then removed in vacuo. Purification by FCC, eluting with EtOAc, gave the sub-title compound (2.7g) as a solid; LC-MS: m/z 609 APCI+.
(vii) 2- Amino-5 - {4- Γ3 -(dimethylamino)- 3 -oxopropyll -2-methoxybenzyl| -6- methylpyrimidin-4-yl 2,4,6-triisopropylbenzenesulfonate
Figure imgf000057_0002
A solution of (E)-2-amino-5-{4-[3-(dimethylamino)-3-oxoprop-l-enyl]-2- methoxybenzyl}-6-methylpyrimidin-4-yl 2,4,6-triisopropylbenzenesulfonate (2.7 g) in EtOH (120 mL) and EtOAc (80 mL) in the presence of 10% palladium on carbon (200 mg) was stirred vigorously under a 3 bar pressure of ¾ for 3h. Solids were then removed by filtration and the filtrate was concentrated in vacuo to give the sub-title (2.7g) as a gum;
LC-MS: m/z 611 APCI+. (viii) 3- (4-Γ(2-Αηιίηο-4- U(2S)- 1 -hydroxypentan-2-yllaminol -6-methylpyrimidin-5-yl)- meth ll-3-methoxyphenyl|-N,N-dimethylpropanamide 2,4,6-triisopropylsulfonic acid salt
As 2,4,6-triisopropylsulfonic acid salt
Figure imgf000058_0001
A mixture of 2-amino-5-{4-[3-(dimethylamino)-3-oxopropyl]-2-methoxybenzyl}-6- methylpyrimidin-4-yl 2,4,6-triisopropylbenzenesulfonate (0.95 g), (5)-2-aminopentan-l-ol (0.48 g) and trifluoroacetic acid (0.12 mL) in butyronitrile (10 mL) were heated at 120°C for 30h. The mixture was then concentrated in vacuo. Purification by FCC, eluting with 8% CH3OH/CH2CI2 gave the sub-title compound (0.9 g) as a gum; LC-MS: m/z 430 APCI+.
(ix) (26 -2-[(2-Amino-5- (4-[3-(dimethylamino)propyll-2-methoxybenzyl| -6-methyl- pyrimidin-4-yl)amino"|pentan- 1 - l
Figure imgf000058_0002
A solution of 3-{4-[(2-amino-4-{[(25)-l-hydroxypentan-2-yl]amino}-6-methylpyrimidin- 5-yl)-methyl]-3-methoxyphenyl}-N,N-dimethylpropanamide 2,4,6-triisopropylsulfonic acid salt (0.9 g) in THF (30 mL) was treated with borane -methyl sulphide complex (6.29 mL, 2M in THF) and heated at 60°C for 30 mins. The mixture was then cooled to RT and treated drop-wise with a solution of N,N'-dimethylethane-l,2-diamine (1.84 g) in CH3OH (30 mL) and then heated at 60°C under N2 for 8h. The mixture was then cooled to RT and the solvent removed in vacuo. The residue was partitioned between water and CH2CI2 and the phases were separated. The organic solution was washed with water before being dried and concentrated in vacuo. Purification by FCC, eluting with 6-7% CH3OH in CH2CI2 with 1%) triethylamine, gave the sub-title compound (0.25 g) as a solid; 1H NMR: 6.81 (s, 1H),
6.71 (d, 1H), 6.64 (d, 1H), 5.67 (s, 2H), 5.42 (d, 1H), 4.60 (s, 1H), 4.12-4.04 (m, 1H), 3.84 (s, 3H), 3.58 (s, 2H), 3.40-3.21 (m, 2H), 2.56-2.51 (m, 2H), 2.19 (t, 2H), 2.12 (s, 6H), 2.06 (s, 3H), 1.70-1.62 (m, 2H), 1.52-1.43 (m, 1H), 1.28-1.19 (m, 1H), 1.10-1.00 (m, 2H), 0.76 (t, 3H); LC-MS: m/z 416 APCI+. (x) 2-r(2-Amino-4-{r(25 l-hydroxypentan-2- 5 -[3 -(dimethylamino)propyl]ph nol
Figure imgf000059_0001
BBr3 (2.31 mL, 1M in CH2CI2) was added drop-wise over 5 mins to an ice cold solution of (2S)-2-[(2-amino-5-{4-[3-(dimethylamino)propy
4-yl)amino]pentan-l-ol (240 mg) in CH2CI2 (10 mL) and the mixture was then stirred at RT for 3h. The mixture was then cooled in an ice bath and CH3OH (3 mL) was added drop-wise and the mixture then stirred for 15 mins at RT. The mixture was again cooled in an ice bath and treated dropwise with HC1 (4M in 1,4-dioxane 0.77 mL). The mixture was stirred for 15 mins at RT and the solvent was then removed in vacuo. Purification by RPHPLC gave the title compound (120 mg) as a white solid; 1H NMR: 9.59 (s, 1H), 6.69 (t, 1H), 6.64 (d, 1H), 6.50 (d, 1H), 5.72-5.49 (m, 3H), 4.61 (s, 1H), 4.13-3.92 (m, 1H), 3.53 (s, 2H), 3.37-3.31 (m, 1H), 3.23 (dd, 1H), 2.43 (t, 2H), 2.14 (t, 2H), 2.13 (s, 3H), 2.08 (s, 6H), 1.64-1.55 (m, 2H), 1.55-1.45 (m, 1H), 1.32-1.22 (m, 1H), 1.11-0.96 (m, 2H), 0.75 (t, 3H); LC-MS: m/z 402 APCI+.
Example 11: 2- \(2- Amino-4- { [(35)- l-hydroxyhexan-3-yll aminol-6-methylpyrimidin-
5-vDmethyll -5- [3-(dimethylamino)propyll phenol
Figure imgf000059_0002
The title compound may be prepared by the steps described below:
(i) 3 - (4- [(2- Amino-4- { \(3S)- 1 -hydroxyhexan-3 -yl] amino) -6-methylpyrimidin-5 - yl)methyl]-3-methoxyphenyl| -N,N-dimethylpropanamide 2,4,6-triisopropylsulfonic acid salt
Figure imgf000060_0001
as 2,4,6-triisopropylsulfonic acid salt
2 -Amino-5 - {4- [3 -(dimethylamino)-3 -oxopropyl] -2-methoxybenzyl} -6-methylpyrimidin-4- yl 2,4,6-triisopropylbenzenesulfonate (preparation: see step (vii) of Example 10, above; 0.95 g), (5)-3-aminohexan-l-ol (0.54 g) and trifluoroacetic acid (0.12 mL) were reacted together using the general method described for step (viii) of Example 10 to give the subtitle compound (0.8 g) as a gum; LC-MS: m/z 444 APCI+.
(ii) (361-3 - [(2- Amino-5 - (4- [3 -(dimethylamino)propyl] -2-methoxybenzyl| -6-methyl- pyrimidin-4-yl)amino1hexan- 1 -ol
Figure imgf000060_0002
3 - {4- [(2- Amino-4- { [(35)- 1 -hydroxy-hexan-3 -yl] amino} -6-methy lpyrimidin-5 -yl)methyl] - 3-methoxyphenyl}-N,N-dimethyl-propanamide 2,4,6-triisopropylsulfonic acid salt (0.8 g) was reacted using the general method described in step (ix) of Example 10 to give the subtitle compound (0.24 g) as a gum: 1H NMR: 6.81 (s, 1H), 6.68 (d 1H), 6.64 (d, 1H), 5.68 (s, 2H), 5.51 (d, 1H), 4.38 (s, 1H), 4.17 (dt, 1H), 3.84 (s, 3H), 3.58 (s, 2H), 3.34-3.24 (m, 2H), 2.55-2.51 (m, 2H), 2.17 (t, 2H), 2.10 (s, 6H), 2.02 (d, 3H), 1.66 (dt, 2H), 1.61-1.53 (m, 1H), 1.46-1.30 (m, 3H), 1.13-1.02 (m, 2H), 0.77 (t, 3H); LC-MS: m/z 430 APCI+. (iii) 2- [(2- Amino-4- { \(3S)- 1 -hydroxyhexan-3 -yl] amino) -6-methylpyrimidin-5 -yOmethyl]- 5 -[3 -(dimethylamino)propyllphenol
Figure imgf000060_0003
(3iS)-3-[(2 -Amino-5- {4-[3-(dimethylamino)propyl]-2-methoxybenzyl} -6-methylpyrimidin- 4-yl)amino]hexan-l-ol (230 mg) was reacted using the general method described in step (x) of Example 10 to give the title compound (60 mg) as a white solid: 1H NMR: 9.63 (s, 1H), 6.71 (d, 1H), 6.64 (s, 1H), 6.51 (d, 1H), 5.68-5.61 (m, 3H), 4.40 (s, 1H), 4.18-4.10 (m, 1H), 3.54 (s, 2H), 3.32-3.20 (m, 2H), 2.43 (t, 2H), 2.14 (t, 2H), 2.12 (s, 3H), 2.09 (s, 6H), 1.64-1.53 (m, 3H), 1.46-1.30 (m, 3H), 1.12-0.98 (m, 2H), 0.75 (t, 3H); LC-MS: m/z 416 APCI+.
Example 12: 2-[(2-Amino-4-{[(3S -l-hvdroxyhexan-3-yllamino}-6-methylpyrimidin- 5-vDmethyll -5- [3-(dimethylamino)propoxyl phenol
Figure imgf000061_0001
The title compound may be prepared by the steps described below:
(i) 4-[3-(tert-Butyldimethylsilyloxy)propoxyl-2-methoxybenzaldehyde
Figure imgf000061_0002
A mixture of 4-hydroxy-2-methoxybenzaldehyde (10 g), (3-bromopropoxy)-tert-butyl- dimethylsilane (25 g) and K2C03 (13.6 g) in DMF (100 niL) was stirred at RT for 5h. The mixture was then diluted with water and extracted with EtOAc, the combined organic solutions were washed with water and brine, dried (Na2S04) and concentrated. Purification by FCC, eluting with 0-15% EtOAc in hexane, gave the sub-title compound (21.3g) as a yellow oil (as a mixture with (3-bromopropoxy)-tert-butyldimethylsilane); LC-MS: m/z 325 [MH+].
(ii) {4-r3-(tert-Butyldimethylsilyloxy)propoxy1-2-methoxyphenyl|methanol
Figure imgf000061_0003
NaBH4 (1.24 g) was added to the solution of 4-[3-(tert-butyldimethylsilyloxy)propoxy]-2- methoxybenzaldehyde (21.3 g) in THF (100 mL) and CH3OH (15 mL) and the mixture was stirred at RT for 2.5h. The mixture was then diluted with water and brine, and extracted with EtOAc. The combined organic solutions were dried (Na2S04) and concentrated in vacuo. Purification by FCC, eluting with 8-29% EtOAc in hexane, gave the subtitle compound (18.5 g) as a colourless oil; 1H NMR (CDC13): 7.14 (d, 1H), 6.42-6.48 (m, 2H), 4.61 (d, 2H), 4.06 (t, 2H), 3.84 (s, 3H), 3.80 (t, 2H), 2.15 (t, 1H), 1.94-2.02 (m,
2H), 0.89 (s, 9H), 0.04 (s, 6H); LC-MS: m/z 309 [M-(OH)].
(iii) tert-Butyl {3-[4-(chloromethyl)-3-methoxyphenoxylpropoxy| dimethylsilane
Figure imgf000062_0001
Methanesulfonyl chloride (4.02 mL) was added to a mixture of {4-[3-(tert- butyldimethylsilyloxy)-propoxy]-2-methoxyphenyl}methanol (8.47 g), DIPEA (13.4 mL) and LiCl (3.29 g) in THF (105 mL) at RT. The mixture was then stirred for 40 mins and then diluted with water and brine. The mixture was extracted with EtOAc, the combined organic solutions were washed with brine, dried (Na2S04) and concentrated in vacuo to give the sub-title compound (10 g) as a yellow oil; 1H NMR (CDC13): 7.23 (dd, 1H), 6.44- 6.49 (m, 2H), 4.63 (s, 2H), 4.06 (t, 2H), 3.85 (s, 3H), 3.79 (t, 2H), 1.94-2.02 (m, 2H), 0.89 (s, 9H), 0.04 (s, 6H).
(iv) Methyl 2- {4-r3-(tert-butyldimethylsilyloxy)propoxy1-2-methoxybenzyl| -3- oxobutanoate
Figure imgf000062_0002
Methyl acetoacetate (4.18 mL) was added to the suspension of NaH (55% oil dispersion, 1.7 g) in DMF (60 mL) at 0°C and the mixture was stirred at RT for 0.5h. tert-Butyl{3-[4- (chloromethyl)-3-methoxyphenoxy]propoxy} dimethylsilane (10 g) in DMF (60 mL) and KI (4.73 g) were added and the mixture was stirred at 80°C for 6h. The mixture was then cooled to RT and diluted with water and extracted with EtOAc. The combined organic solutions were washed with water and brine, dried (Na2S04) and concentrated in vacuo. Purification by FCC, eluting with 0-17% EtOAc in hexane, gave of the sub-title compound
(9.9 g) as a yellow oil; 1H NMR (CDCI3): 6.99 (d, 1H), 6.42 (d, 1H), 6.37 (dd, 1H), 4.02 (t, 2H), 3.89 (t, 1H), 3.80 (s, 3H), 3.79 (t, 2H), 3.67 (s, 3H), 2.99-3.14 (m, 2H), 2.17 (s, 3H), 1.92-2.00 (m, 2H), 0.88 (s, 9H), 0.04 (s, 6H). (v) 2-Amino-5- (4-r3-(tert-butyldimethylsilyloxy)propoxy1-2-methoxybenzyl| -6- methylpyrimidin-4-ol
Figure imgf000063_0001
Guanidium carbonate (6.4g) was added to a solution of methyl 2-{4-[3-(tert-butyldimethyl- silyloxy)propoxy]-2-methoxybenzyl}-3-oxobutanoate (11.6 g) in CH3OH (116 mL) and the mixture was heated at 75°C for 8h. The mixture was then cooled to RT and
concentrated in vacuo. The residue was diluted with EtOAc (50 mL) and water (50 mL), and the mixture was stirred for 5.5h. The resulting precipitate was collected and washed with water and EtOAc to give the subtitle compound as a white solid; 1H NMR: 10.75 (br s, 1H), 6.71 (d, 1H), 6.48 (d, 1H), 6.36 (dd, 1H), 6.30 (br s, 2H), 3.97 (t, 2H), 3.77 (s, 3H), 3.73 (t, 2H), 3.45 (s, 2H), 1.92 (s, 3H), 1.82-1.90 (m, 2H), 0.85 (s, 9H), 0.02 (s, 6H); LC- MS: m/z = 434 [MH+].
(vi) 2-Amino-5- (4-r3-(tert-butyldimethylsilyloxy)propoxy1-2-methoxybenzyl| -6- methylpyrimidin-4-yl 2,4,6-trimethylbenzenesulfonate
Figure imgf000063_0002
2-Mesitylenesulfonyl chloride (4.96 g) was added to a solution of N,N,N',N'-tetramethyl- 1,3-propanediamine (3.79 mL) and 2-amino-5-{4-[3-(tert-butyldimethylsilyloxy)propoxy]- 2-methoxybenzyl}-6-methylpyrimidin-4-ol (6.55 g) in THF (66 mL) and the mixture was stirred at RT for 3h. The mixture was then diluted with water and extracted with EtOAc. The combined organic solutions were washed with brine, dried (Na2S04) and concentrated in vacuo. Purification by FCC, eluting with 23-44% EtOAc in hexane, gave the sub-title compound (8.8 g) as a white solid: 1H NMR (CDCI3): 6.93 (s, 2H), 6.71 (d, 1H), 6.41 (d, 1H), 6.33 (dd, 1H), 4.66 (br s, 2H), 4.02 (t, 2H), 3.78 (s, 3H), 3.77-3.82 (m, 2H), 3.75 (s, 2H), 2.59 (s, 6H), 2.31 (s, 3H), 2.25 (s, 3H), 1.93-2.01 (m, 2H), 0.89 (s, 9H), 0.05 (s, 6H); LC-MS: m/z = 616 [MH ].
(vii) (S)-tert- utyl l-hydroxyhexan-3-ylcarbamate
"OH
BocHN^^^^
Di-tert-butyl dicarbonate (2.62 g) was added to a solution of (5)-3-aminohexan-l-ol (1.17 g) in aq. NaOH (5M, 2.4 mL), water (2.6 mL) and 1,4-dioxane (10 mL) and the mixture was stirred at RT for 2h. The mixture was then diluted with water and extracted with EtOAc. The combined organic solutions were dried (Na2S04) and concentrated in vacuo to give the sub-title compound (2.39 g) as a yellow oil; LC-MS: m/z = 218 [MH+].
(viii) (S)-tert-Butyl l-(benzyloxy)hexan-3-ylcarbamate
BocHN'^^^
NaH (55% oil dispersion, 240 mg) was added to a solution of {S)-tert-bvXy\ 1- hydroxyhexan-3-ylcarbamate (1.2 g) and benzyl bromide (0.654 mL) in DMF (20 mL) at 0°C and the mixture was stirred for 2h. The mixture was then diluted with water and extracted with EtOAc. The combined organic solutions were washed with water and brine, dried (Na2S04) and concentrated in vacuo. Purification by FCC eluting with 0-10% EtOAc in hexane, gave the sub-title compound (1.16g) as a colourless oil; LC-MS: m/z = 308 [MH+].
(ix) (S)- l-(Benzyloxy)hexan-3 -amine
-^^OBn
HsN^^^
HC1 (4M in 1,4-dioxane, 20 mL) was added to (S)-tert-butyl l-(benzyloxy)hexan-3- ylcarbamate (1.16 g) and the mixture was stirred at RT for 5h. The mixture was then concentrated in vacuo, diluted with 40%> aq. K2CO3 and extracted with EtOAc. The combined organic solutions were dried (Na2S04) and concentrated in vacuo to give the sub-title compound (789 mg) as a yellow oil; 1H NMR (CDCI3): 7.25-7.37 (m, 5H), 4.51 (s, 2H), 3.53-3.64 (m, 2H), 2.86-2.94 (m, 1H), 1.22-1.80 (m, 6H), 0.91 (t, 3H); LC-MS: m/z = 208 [MH+]. (x) N4-r(36 -l-(Benzyloxy)hexan-3-yl1-5-r4-(3-(rter^butyl(dimethyl)silyl1oxylpropoxy)- 2-methoxybenzyll-6-methylpyrimidine-2,4-diamine
Figure imgf000065_0001
TFA (98 μί) was added to a solution of 2-amino-5-{4-[3-(tert-butyl- dimethylsilyloxy)propoxy]-2-methoxybenzyl}-6-methylpyrimidin-4-yl 2,4,6-trimethyl- benzenesulfonate (preparation: see step (vi) of Example 12 above; 781 mg) and (S)-l- (benzyloxy)hexan-3 -amine (789 mg) at RT and the mixture was then stirred at 120°C for 15h. The mixture was then cooled to RT and concentrated in vacuo. The residue was diluted with 40% aq. K2CO3 and extracted with CHCl3/EtOH (3/1). The combined organic solutions were dried (Na2S04) and concentrated in vacuo. The residue was diluted with EtOAc, stirred, filtered, and concentrated in vacuo. Purification by FCC, eluting with 0- 50%) EtOAc in hexane, gave the sub-title compound (805mg) as yellow oil; LC-MS: m/z = 624 [MH+].
(xi) 3-(4-[(2-Amino-4-{[(36 -l-(benzyloxy)hexan-3-yllamino|-6-methylpyrimidin-5- yl)methyl]-3-methoxyphenoxy|propan- 1 -ol
Figure imgf000065_0002
HC1 (12M, 529 μΕ) was added to a solution of N4-[(35)-l-(benzyloxy)hexan-3-yl]-5-[4-(3- {[tert-butyl(dimethyl)silyl]oxy}propoxy)-2-methoxybenzyl]-6-methylpyrimidine-2,4- diamine (805 mg) in CH3OH (10 mL) and the mixture was stirred at RT for lh. The mixture was then concentrated in vacuo, diluted with aq. NaHC03 and extracted with EtOAc. The combined organic solutions were dried (Na2S04) and concentrated in vacuo. Purification by FCC, eluting with 55-100% EtOAc in hexane and 7% MeOH in CHC13 gave the sub-title compound (476 mg) as a yellow oil; 1H NMR (CDC13): 7.21-7.34 (m, 5H), 6.79 (d, 1H), 6.44 (d, 1H), 6.35 (dd, 1H), 4.75 (d, 1H), 4.53 (br s, 2H), 4.33 (s, 2H), 4.20-4.31 (m, 1H), 4.05 (t, 2H), 3.83 (s, 3H), 3.83 (t, 2H), 3.57 (s, 2H), 3.23-3.34 (m, 2H), 2.29 (s, 3H), 1.96-2.04 (m, 2H), 1.70-1.85 (m, 1H), 1.45-1.58 (m, 1H), 1.19-1.45 (m, 2H), 1.05-1.19 (m, 2H), 0.80 (t, 3H); LC-MS: m/z = 510 [MH+].
(xii) 3- (4-[(2-Amino-4- {[(351)- 1 -(benzyloxy)hexan-3-yll amino) -6-methylpyrimidin-5- yl)methvH-3-methoxyphenoxylpropyl methanesulfonate
Figure imgf000066_0001
Mesyl chloride (86 μί) was added to a solution of 3-{4-[(2-amino-4-{[(35)-l- (benzyloxy)hexan-3-yl]amino}-6-methylpyrimidin-5-yl)methyl]-3- methoxyphenoxy}propan-l-ol (470 mg) and triethylamine (0.258 mL) in THF (10 mL) at 0 °C and the mixture was then stirred for 40 mins. Further mesyl chloride (215 μί) and triethylamine (0.774 mL) were added to the mixture at 0°C until the reaction was complete. The resulting mixture was diluted with NaHC03 solution and extracted with EtOAc. The combined organic solutions were dried (Na2S04) and concentrated in vacuo to give the sub-title compound (559 mg) as a yellow oil: 1H NMR (CDC13): 7.21-7.34 (m, 5H), 6.79 (d, 1H), 6.43 (d, 1H), 6.33 (dd, 1H), 4.75 (d, 1H), 4.52 (br s, 2H), 4.41 (t, 2H), 4.34 (s, 2H), 4.20-4.32 (m, 1H), 4.01 (t, 2H), 3.84 (s, 3H), 3.57 (s, 2H), 3.25-3.36 (m, 2H), 2.97 (s, 3H), 2.29 (s, 3H), 2.14-2.22 (m, 2H), 1.70-1.85 (m, 1H), 1.46-1.58 (m, 1H), 1.32-1.44 (m, 1H), 1.20-1.32 (m, 1H), 1.05-1.18 (m, 2H), 0.79 (t, 3H).
(xiii) N4-[(36 -l-(Benzyloxy)hexan-3-yll-5-(4-[3-(dimethylamino)propoxyl-2- methoxybenzyl|-6-methylpyrimidine-2 4-diamine
Figure imgf000066_0002
3- {4-[(2-Amino-4- {[(35)- 1 -(benzyloxy)hexan-3-yl] amino} -6-methylpyrimidin-5- yl)methyl]-3-methoxyphenoxy}propyl methanesulfonate (140 mg) and dimethylamine (50% in water, 1 mL) in CH3CN (2 mL) was stirred at 80°C for 1.5h. The resulting mixture was diluted with 40% aq. K2CO3 and extracted with EtOAc. The combined organic solutions were dried (Na2S04) and concentrated in vacuo. Purification by FCC, eluting with 0-7%) MeOH in CHCI3, gave the sub-title compound (116 mg) as a colourless oil; 1H NMR (CDCI3): 7.21-7.34 (m, 5H), 6.78 (d, IH), 6.44 (d, IH), 6.35 (dd, IH), 4.75 (d, IH), 4.49 (br s, 2H), 4.33 (s, 2H), 4.20-4.32 (m, IH), 3.94 (t, 2H), 3.83 (s, 3H), 3.57 (s, 2H), 3.25-3.32 (m, 2H), 2.37-2.46 (m, 2H), 2.30 (s, 3H), 2.23 (s, 6H), 1.87-1.96 (m, 2H), 1.74- 1.85 (m, IH), 1.45-1.58 (m, IH), 1.20-1.44 (m, 2H), 1.05-1.18 (m, 2H), 0.79 (t, 3H); LC- MS: m/z = 537 [MH+].
(xiv) (36 -3-[(2-Amino-5-(4-[3-(dimethylamino)propoxyl-2-methoxybenzyl|-6- methylpyrimidin-4-vDamino hexan- 1 -ol
Figure imgf000067_0001
HC1 (5% in CH3OH, 2 mL) was added to a solution of N4-[(35)-l-(benzyloxy)hexan-3-yl]- 5-{4-[3-(dimethylamino)propoxy]-2-methoxybenzyl}-6-methylpyrimidine-2,4-diamine (116 mg) and 10% palladium on carbon (500 mg) in CH3OH (6 mL) and the mixture was stirred at RT for 3h under H2. The mixture was then filtered and concentrated in vacuo and the residue diluted with 40% aq. K2CO3 and extracted with EtOAc. The combined organic solutions were dried (Na2S04) and concentrated in vacuo. Purification by FCC, eluting with 0-10%) MeOH in CHCI3, gave the sub-title compound (84mg) as a colourless oil; 1H
NMR (CDCI3): 6.81 (d, IH), 6.43-6.47 (m, IH), 6.35-6.40 (m, IH), 4.70 (br s, IH), 4.56 (d, IH), 4.00-4.15 (m, IH), 3.95 (t, 2H), 3.85 (s, 3H), 3.60 (s, 2H), 3.40-3.50 (m, IH), 3.24-3.35 (m, IH), 2.42 (t, 2H), 2.31 (s, 3H), 2.22 (s, 6H), 1.87-1.96 (m, 2H), 1.73-1.85 (m, IH), 1.31-1.46 (m, IH), 0.98-1.26 (m, 4H), 0.75 (t, 3H). (xy) 2- \(2 -Amino-4- { \(3S)- 1 -hydroxyhexan-3 -yll amino) -6-methylpyrimidin-5 -vQmethyll - 5 -[3 -(dimethylamino)propoxylphenol
Figure imgf000068_0001
BBr3 (1M in CH2C12, 0.46 mL) was added to a solution of (3S)-3-[(2-amino-5-{4-[3- (dimethylamino)propoxy]-2-methoxybenzyl}-6-methylpyrimidin-4-yl)amino]hexan-l-ol (41 mg) in CH2C12 (1 mL) and the mixture was stirred at 45°C for 1 lh. The mixture was then diluted with 40% aq. K2C03 and extracted with EtOAc. The combined organic solutions were dried (Na2S04) and concentrated in vacuo. Purification by FCC, eluting with 0-10% MeOH in CHC13 gave the title compound (32 mg) as a white solid; 1H NMR: 9.76 (br s, 1H), 6.66 (d, 1H), 6.37 (d, 1H), 6.25 (dd, 1H), 5.58-5.66 (m 3H), 4.30-4.50 (m, 1H), 4.07-4.20 (m, 1H), 3.85 (t, 2H), 3.48 (s, 3H), 2.29 (t, 2H), 2.10 (s, 6H), 2.07-2.13 (m, 3H), 1.72-1.82 (m, 2H), 1.52-1.64 (m, 1H), 1.30-1.50 (m, 3H), 1.00-1.12 (m, 2H), 0.75 (t, 3H); LC-MS: m/z = 432 [MH+].
Example 13: 2-[(2-Amino-4-{[(3S -l-hvdroxyhexan-3-yllamino}-6-methylpyrimidin- 5-vDmethyll -5- { r(2-methoxyethyl)(methyl)aminol methyljphenol
Figure imgf000068_0002
The title compound may be prepared by the steps described below:
(i) 4- [(2- Amino-4- { \(3S)- 1 -hydroxyhexan-3 -yl] amino) -6-methylpyrimidin-5 -yQmethyl] -3 - hydroxybenzonitrile
Figure imgf000069_0001
BBr3 (3.79 mL) was added to a solution of 4-[(2-amino-4-chloro-6-methylpyrimidin-5- yl)methyl]-3-methoxybenzonitrile (preparation: step (iii) of Example 1, above; 280 mg) in CH2CI2 (10 mL), and the mixture was stirred for 16h. CH3OH (4 mL) was then added and after 20 mins HC1 (4M in dioxane, 0.947 mL) was added and the mixture stirred for a further 20 mins. The mixture was then concentrated in vacuo. The crude product was purified by RPHPLC to give the sub-title compound (150mg) as a colourless gum; LC-MS: m/z 356 APCI+.
(ii) 4- [(2- Amino-4- { \(3S)- 1 -hydroxyhexan-3 -yl] amino) -6-methylpyrimidin-5 -yQmethyl]- 3 -hydroxybenzaldehyde
Figure imgf000069_0002
4- [(2 -Amino-4- {[(35)- 1 -hydroxyhexan-3-yl]amino} -6-methylpyrimidin-5-yl)methyl]-3- hydroxybenzonitrile (150 mg) was reacted using the general method described for step (v) of Example 1, to give the sub-title compound (100 mg) as a yellow foam; LC-MS: m/z 432 APCI+.
(iii) 2- IY2- Amino-4- { \(3S)- 1 -hydroxyhexan-3 -yll amino) -6-methylpyrimidin-5 -vDmethyll-
5- ([(2-methoxyethyl)(methyl)aminolmethyl)phenol
Figure imgf000069_0003
4- [(2-Amino-4- {[(35)- 1 -hydroxyhexan-3-yl]amino} -6-methylpyrimidin-5-yl)methyl]-3- hydroxybenzaldehyde (100 mg) and 2-methoxy-N-methylethanamine (74.6 mg) were reacted using the general method described for step (vi) of Example 1. The mixture was stirred for 16 h. The solvent was evaporated and CH3OH added. The crude product was purified by RPHPLC to afford the title (35mg) compound as a white solid; 1H NMR: 9.69 (s, 1H), 6.80 (s, 1H), 6.74 (d, 1H), 6.59 (d, 1H), 5.73-5.62 (m, 3H), 4.38 (s, 1H), 4.22-4.10 (m, 1H), 3.56 (s, 2H), 3.42 (t, 2H), 3.37 (dd, 1H), 3.24 (dd, 1H), 3.21 (s, 3H), 2.46 (t, 2H), 2.11 (d, 6H), 1.59 (td, 1H), 1.48-1.30 (m, 3H), 1.15-0.97 (m, 2H), 0.75 (t, 3H); LC-MS: m/z = 432 [MH+].
Example 14: 2-[(2-Amino-4-{[(3S -l-hvdroxyhexan-3-yllamino}-6-methylpyrimidin-
5- vDmethyll -5- [2-(dimethylamino)ethoxyl phenol
Figure imgf000070_0001
The title compound may be prepared by the steps described below:
(i) Methyl 2 4-(benzyloxy)-2-methoxybenzyl"|-3-oxobutanoate
Figure imgf000070_0002
DMF (7 mL) was added to a mixture of "Pd-118" (dichloro [Ι,Γ bis(di-tert- butylphosphino)]ferrocene palladium (II)) (73 mg) and tetrabutyl-ammonium chloride hydrate (264 mg) and 4-(benzyloxy)-l-bromo-2-methoxybenzene (1.31 g), under N2. Methyl 3-hydroxy-2-methylenebutyrate (543 μί) and N-cyclohexyl-N- methylcyclohexanamine (2.84 mL) were added and the solution was heated at 100°C for 18h and then allowed to cool to RT. The mixture was diluted with CH3OH and loaded on to a SCX cartridge. The compound was eluted with CH3OH and the filtrate concentrated in vacuo. Purification by FCC eluting with 0-10% EtOAc in isohexane gave the sub-title compound (888 mg) as a yellow oil; 1H NMR: 7.43 (d, 2H), 7.39 (t, 2H), 7.35-7.31 (m, IH), 7.01 (d, IH), 6.52 (d, IH), 6.45 (dd, IH), 5.03 (s, 2H), 3.89 (t, IH), 3.79 (s, 3H), 3.69
(s, 3H), 3.15-3.00 (m, 2H), 2.18 (s, 3H); LC-MS m/z 341 APCI-.
(ii) 2- Amino-5 - |"4-(benzyloxy)-2-methoxybenzyl] -6-methylpyrimidin-4-ol
Figure imgf000071_0001
Methyl 2-[4-(benzyloxy)-2-methoxybenzyl]-3-oxobutanoate (870 mg) was dissolved in CH3OH (14 mL) and guanidine hemicarbonate (687 mg) was added. The mixture was then heated to 60°C for 5h and allowed to cool to RT. The mixture was then filtered and washed with diethyl ether and water. The filtrate was concentrated in vacuo and more diethyl ether and water added. After scratching the crude material the resulting solid was collected by filtration, washed with water and dried under vacuum to give the sub-title compound (467 mg) as a pale yellow solid; 1H NMR: 7.44 (d, 2H), 7.39 (t, 2H), 7.32 (t, IH), 6.73 (d, IH), 6.60 (d, IH), 6.46 (dd, IH), 6.35 (s, IH), 5.05 (s, 2H), 3.77 (s, 3H), 3.45 (s, 2H), 1.93 (s, 3H); LC-MS m/z 352 APCI+.
(iii) 2-Amino-5-[4-(benzyloxy)-2-methoxybenzyll-6-methylpyrimidin-4-yl 2,4,6- triisopropylbenzenesulfonate
Figure imgf000071_0002
A mixture of 2-amino-5-[4-(benzyloxy)-2-methoxybenzyl]-6-methylpyrimidin-4-ol (467 mg), 2,4,6-triisopropylbenzene-l-sulfonyl chloride (523 mg) and DABCO (194 mg) in THF (13 mL) was heated at 50°C for 3h and then allowed to cool to RT. The mixture was partitioned between EtOAc (100 mL) and water (50 mL) and the phases were separated. The organic solution was washed with further water (50 mL), sat. NaHC03 (2 x 50 mL) and brine (50 mL). The solution was then dried (MgS04) and concentrated in vacuo to give the sub-title compound (807 mg) as a yellow solid; 1H NMR (CDCI3): 7.43 (d, 2H), 7.39 (t, 2H), 7.32 (dd, IH), 7.16 (s, 2H), 6.78 (d, IH), 6.50 (d, IH), 6.41 (dd, IH), 5.02 (s, 2H), 4.67 (s, 2H), 4.19-4.09 (m, 2H), 3.76 (s, 5H), 2.97-2.86 (m, IH), 2.27 (s, 3H), 1.25 (d, 6H), 1.20 (d, 12H); LC-MS m/z 618 APCI+.
(iv) (36 -3-((2-Amino-5-[4-(benzyloxy)-2-methoxybenzyll-6-methylpyrimidin-4- yl| amino)-hexan- 1 -ol
Figure imgf000072_0001
2-Amino-5-[4-(benzyloxy)-2-methoxybenzyl]-6-methylpyrimidin-4-yl 2,4,6- triisopropylbenzenesulfonate (807 mg) and (5)-3-aminohexan-l-ol (459 mg) were reacted using the general method described for step (viii) of Example 10, heating for 20 h.
Purification by RPHPLC gave the sub-title compound (311 mg) as a yellow foam; 1H NMR (CDCls): 7.43-7.36 (m, 4H), 7.35-7.30 (m, IH), 6.85 (d, IH), 6.55 (d, IH), 6.47 (dd, IH), 5.13 (s, IH), 5.03 (s, 2H), 4.55 (d, 3H), 4.15-4.04 (m, IH), 3.86 (s, 3H), 3.63 (s, 2H), 3.48 (s, IH), 3.31 (t, IH), 2.34 (s, 3H), 1.86-1.75 (m, IH), 1.46-1.36 (m, IH), 1.27-1.17 (m, IH), 1.16-0.99 (m, 3H), 0.77 (t, 3H); LC-MS m/z 451 APCI+.
(v) 5-[4-(Benzyloxy)-2-methoxybenzyll-A4-[(36 -l-([ter^butyl(dimethyl)silylloxy|hexan- 3 -yl] -6-methylpyrimidine-2,4-diamine
Figure imgf000072_0002
Imidazole (94 mg) was added to a solution of (35)-3-({2-amino-5-[4-(benzyloxy)-2- methoxybenzyl]-6-methylpyrimidin-4-yl}amino)-hexan-l-ol (310 mg) and tert-butyl- dimethylsilyl chloride (207 mg) in DMF (2.5 mL) under an atmosphere of N2, and the mixture was stirred 24h. Further tert-butyldimethylsilyl chloride (104 mg) and imidazole (47 mg) were then added and the mixture was stirred for a further 3h. The solvent was removed in vacuo and the residue was dissolved in CH2CI2 (50 mL) and the resulting solution was washed with water (4 x 50 mL), dried (Na2S04) and concentrated in vacuo to give the sub-title compound (506 mg) as a yellow oil; LC-MS m/z 565 APCI+.
(vi) 4-[(2-Amino-4- {[(361-1 -([tert-butyl(dimethyl)silylloxy|hexan-3-yllamino|-6-methyl- pyrimidin-5 -yQmethy 1] -3 -methoxyphenol
Figure imgf000073_0001
A mixture of 5-[4-(benzyloxy)-2-methoxybenzyl]-N4-[(35)-l-{[fert-butyl(dimethyl)silyl]- oxy}hexan-3-yl]-6-methylpyrimidine-2,4-diamine (390 mg) and palladium on carbon (147 mg) in EtOH (7 mL) was reacted with H2 (3 bar) for 4h. The mixture was filtered and washed with EtOH and a solution of 10% EtOH in CH2C12 and the filtrate was
concentrated in vacuo. The resulting foam was scratched and treated with isohexane to give an off-white solid which was filtered, washed with further isohexane and dried under vacuum to give the sub-title compound (295 mg) as an off-white solid; 1H NMR (CDC13): 6.70 (d, 1H), 6.43 (d, 1H), 6.16 (dd, 1H), 4.87 (s, 1H), 4.57 (s, 2H), 4.25-4.16 (m, 1H), 3.86 (s, 3H), 3.57 (s, 2H), 3.53-3.43 (m, 2H), 2.31 (s, 3H), 1.74-1.64 (m, 1H), 1.54-1.38 (m, 2H), 1.30-1.20 (m, 1H), 1.08 (ddd, 2H), 0.85 (s, 9H), 0.79 (t, 3H), -0.04 (s, 6H); LC- MS m/z 475 APCI+.
(vii) (36 -3-[(2-Amino-5-(4-[2-(dimethylamino)ethoxyl-2-methoxybenzyl|-6- methylpyrimidin-4-yl)amino"|hexan- 1 -ol
Figure imgf000073_0002
Diisopropyl azodicarboxylate (117 μί) was added to 4-[(2-amino-4-{[(35)-l-{[tert- butyl(dimethyl)silyl]oxy}hexan-3-yl]amino}-6-methyl-pyrimidin-5-yl)methyl]-3- methoxyphenol (94 mg), 2-(dimethylamino)ethanol (99 μί) and triphenylphosphine (156 mg) in THF (2 mL) under N2 at 0°C. The mixture was then warmed to 60°C for lOh and then further triphenylphosphine (156 mg), 2-(dimethylamino)ethanol (99 μί) and diisopropyl azodicarboxylate (1 17 μί) were added and the mixture heated at 60°C for another 2h. The mixture was then allowed to cool to RT and then poured onto an SCX cartridge. The mixture was eluted first with CH3OH and then 1M NH3 in CH3OH and the basic fraction collected and concentrated in vacuo. The resulting residue was purified by RPHPLC to give the sub-title compound (53 mg) as a colourless gum; 1H NMR: 6.65 (d, IH), 6.54 (d, IH), 6.40 (dd, IH), 5.67 (s, 2H), 5.50 (d, IH), 4.40 (t, IH), 4.18 (dt, IH), 3.99 (t, 2H), 3.83 (s, 3H), 3.53 (s, 2H), 3.39-3.25 (m, 2H), 2.61-2.55 (m, 2H), 2.19 (s, 6H), 2.02 (s, 3H), 1.58 (td, IH), 1.47-1.30 (m, 3H), 1.16-1.02 (m, 2H), 0.77 (t, 3H); LC-MS m/z 432 multimode+.
(Viii) 2- IY2- Amino-4- { IY351)- 1 -hvdroxyhexan-3 -yll amino) -6-methylpyrimidin-5 - vDmethyll -5 - r2-(dimethylamino)ethoxy1phenol
Figure imgf000074_0001
A solution of BBr3 (1M in CH2CI2, 185 μί) was added drop-wise over 5 mins to a solution of (35)-3-[(2-amino-5- {4-[2-(dimethylamino)ethoxy]-2-methoxybenzyl} -6- methylpyrimidin-4-yl)amino]hexan-l-ol (20 mg) in CH2CI2 (1 mL) at 0°C. The mixture was stirred vigorously at RT under N2 for 3h. The mixture was then cooled to 0°C and further BBr3 (1M in CH2CI2, 185 μί) was added. The mixture was allowed to warm to RT for 30 mins and was then cooled to 0°C and CH3OH (1 mL) was added dropwise. The mixture was stirred at RT for 15 mins and then cooled to 0°C and treated, dropwise, with HC1 (4M in 1,4-dioxane, 58 μί). The mixture was stirred for 15 mins at RT and the solvent removed in vacuo. The residue was azeotroped with acetonitrile. Purification by RPHPLC gave the title compound (8.2 mg) as a colourless gum; 1H NMR: 10.07 (s, IH), 6.91 (d, 2H), 6.75 (d, IH), 6.44 (d, IH), 6.33 (dd, IH), 4.38 (s, IH), 4.32-4.24 (m, IH), 4.00 (t, 2H), 3.55 (s, 2H), 3.31 (d, 2H), 2.82 (s, 2H), 2.37 (s, 6H), 2.22 (s, 3H), 1.68-1.50 (m, 2H), 1.49-1.34 (m, 2H), 1.14-1.01 (m, 2H), 0.78 (t, 3H); LC-MS m/z 418 multimode+. The ability of compounds to activate TLR7 in vitro was assessed using the human TLR7 assay described below. 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.
Table 1
Figure imgf000075_0001
Human 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 pEC5o value.
Table 2
Figure imgf000076_0001
All of the compounds in Table 2 were assayed by using Method 1, except that Method 2 was used for the compound of Example 12. hERG ( human ether-a-go-go ) Analysis - Method 1
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 are shown in Table 3, below. hERG Analysis - Method 2
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 mins. 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 3.5μΕ 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 3.5μί 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 mins 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 3.5 Ε 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 (-82m V) 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).
Table 3
Figure imgf000080_0001
All of the compounds in Table 3 were assayed by using Method 2, except that Method 1 was used for the compound of Example 12. As shown in Table 3, the compounds of the invention exhibit favourable hERG inhibition activity.

Claims

Claims
1. A compound of Formula (I) :
Figure imgf000081_0001
(I)
wherein:
Q is a direct bond, -0-(C2-4alkylene)- or -(Ci_4alkylene)-;
R1 is selected from hydrogen, Ci_4alkyl, Ci_3alkoxyC2-4alkyl, hydroxyC2_4alkyl, C3-5cycloalkyl, tetrahydropyranyl, benzyl, C-linked pyrrolidinyl, C-linked piperidinyl and (RX)(RY)N-C2_4alkyl, wherein Rx and RY each independently represent hydrogen or Ci_3alkyl; and
R2 is selected from hydrogen, Ci_4alkyl and Ci_3alkoxyC2-4alkyl;
or R1 and R2 together with the nitrogen to which they are attached form a saturated heterocyclyl selected from azetidin-l-yl, pyrrolidin-l-yl, piperidin-l-yl, morpholin-4-yl, piperazin-l-yl, azepan-l-yl, 1,4-diazepan-l-yl; wherein one or two carbon atoms within said heterocyclyl may be substituted by oxo and wherein the available nitrogen of said piperazin-l-yl and 1,4-diazepan-l-yl is optionally substituted by Ci_3alkyl,
hydroxyCi_3alkyl or Ci_2alkoxyC2-3alkyl;
or R1 and R2 together with the nitrogen to which they are attached form a
5-membered aromatic heterocyclyl containing a total of 1-3 nitrogen atoms; wherein said 5-membered aromatic heterocyclyl is optionally substituted on a ring carbon atom by Ci_3alkyl;
R3 is selected from hydrogen, hydroxymethyl and 2-hydroxyethyl;
or a pharmaceutically acceptable salt thereof.
2. A compound of Formula (I),as claimed in claim 1, selected from: 2-[(2-amino-4- {[(35)- 1 -hydroxyhexan-3-yl]amino} -6-methylpyrimidin-5- yl)methyl]-5-{[(2-hydroxyethyl)(methyl)amino]methyl}phenol;
2- {[2-amino-4-(butylamino)-6-methylpyrimidin-5-yl]methyl} -5-(pyrrolidin- 1 - ylmethyl)phenol;
2-{[2-amino-4-(butylamino)-6-methylpyrimidin-5-yl]methyl}-5-[(4- methylpiperazin- 1 -yl)methyl]phenol;
2-[(2-amino-4- {[(35)- 1 -hydroxyhexan-3-yl]amino} -6-methylpyrimidin-5- yl)methyl]-5-[(dimethylamino)methyl]phenol;
2-[(2-amino-4- {[(35)- 1 -hydroxyhexan-3-yl]amino} -6-methylpyrimidin-5- yl)methyl] -5 - [(4-methylpiperazin- 1 -yl)methyl]phenol;
2-[(2-amino-4- {[(35)- 1 -hydroxyhexan-3-yl]amino} -6-methylpyrimidin-5- yl)methyl] -5 - [(diethylamino)methyl]phenol;
2-[(2-amino-4- {[(35)- 1 -hydroxyhexan-3-yl]amino} -6-methylpyrimidin-5- yl)methyl] -5 - { [ethyl(methyl)amino]methyl} phenol;
2-[(2-amino-4- {[(25)- 1 -hydroxypentan-2-yl]amino} -6-methylpyrimidin-5- yl)methyl]-5-[(dimethylamino)methyl]phenol;
2-[(2-amino-4- {[(25)- 1 -hydroxypentan-2-yl]amino} -6-methylpyrimidin-5- yl)methyl]-5-(pyrrolidin-l-ylmethyl)phenol;
2-[(2-amino-4- {[(25)- 1 -hydroxypentan-2-yl]amino} -6-methylpyrimidin-5- yl)methyl]-5 - [3 -(dimethylamino)propyl]phenol;
2-[(2-amino-4- {[(35)- 1 -hydroxyhexan-3-yl]amino} -6-methylpyrimidin-5- yl)methyl]-5 - [3 -(dimethylamino)propyl]phenol;
2-[(2-amino-4- {[(35)- 1 -hydroxyhexan-3-yl]amino} -6-methylpyrimidin-5- yl)methyl] -5 - [3 -(dimethylamino)propoxy]phenol;
2-[(2-amino-4- {[(35)- 1 -hydroxyhexan-3-yl]amino} -6-methylpyrimidin-5- yl)methyl]-5-{[(2-methoxyethyl)(methyl)amino]methyl}phenol; and
2-[(2-amino-4- {[(35)- 1 -hydroxyhexan-3-yl]amino} -6-methylpyrimidin-5- yl)methyl] -5 - [2-(dimethylamino)ethoxy]phenol;
or a pharmaceutically acceptable salt thereof. A pharmaceutical composition which comprises a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as claimed in claim 1 or claim 2, in association with a pharmaceutically-acceptable diluent or carrier.
A compound of Formula (I), or a pharmaceutically acceptable salt thereof, as claimed in claim 1 or claim 2, for use as a medicament.
A compound of Formula (I), or a pharmaceutically acceptable salt thereof, as claimed in claim 1 or claim 2, for use in the treatment of cancer.
Use of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, as claimed in claim 1 or claim 2, for the preparation of a medicament for 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 the compound of Formula (I), or a pharmaceutically acceptable salt thereof, as claimed in claim 1 or claim 2.
PCT/GB2011/052245 2010-11-19 2011-11-17 Phenol compounds als toll -like receptor 7 agonists WO2012066335A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US41570310P 2010-11-19 2010-11-19
US61/415,703 2010-11-19

Publications (1)

Publication Number Publication Date
WO2012066335A1 true WO2012066335A1 (en) 2012-05-24

Family

ID=45099136

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2011/052245 WO2012066335A1 (en) 2010-11-19 2011-11-17 Phenol compounds als toll -like receptor 7 agonists

Country Status (1)

Country Link
WO (1) WO2012066335A1 (en)

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014023813A1 (en) * 2012-08-10 2014-02-13 Janssen R&D Ireland Alkylpyrimidine derivatives for the treatment of viral infections and further diseases
US9045472B2 (en) 2010-12-16 2015-06-02 Astrazeneca Ab Imidazoquinoline compounds
US9376398B2 (en) 2012-05-18 2016-06-28 Sumitomo Dainippon Pharma Co., Ltd Carboxylic acid compounds
WO2017061532A1 (en) * 2015-10-07 2017-04-13 大日本住友製薬株式会社 Pyrimidine compound
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
US10253003B2 (en) 2012-11-16 2019-04-09 Janssen Sciences Ireland Uc Heterocyclic substituted 2-amino quinazoline derivatives for the treatment of viral infections
US10259814B2 (en) 2012-10-10 2019-04-16 Janssen Sciences Ireland Uc Pyrrolo[3,2-d]pyrimidine derivatives for the treatment of viral infections and other diseases
US10259793B2 (en) 2013-02-21 2019-04-16 Janssen Sciences Ireland Uc 2-aminopyrimidine derivatives for the treatment of viral infections
US10266543B2 (en) 2013-03-29 2019-04-23 Janssen Sciences Ireland Uc Macrocyclic deaza-purinones for the treatment of viral infections
US10272085B2 (en) 2011-04-08 2019-04-30 Janssen Sciences Ireland Uc Pyrimidine derivatives for the treatment of viral infections
US10280167B2 (en) 2011-11-09 2019-05-07 Janssen Sciences Ireland Uc Purine derivatives for the treatment of viral infections
US10280180B2 (en) 2012-07-13 2019-05-07 Janssen Sciences Ireland Uc Macrocyclic purines for the treatment of viral infections
US10316043B2 (en) 2013-07-30 2019-06-11 Janssen Sciences Ireland Unlimited Company Thieno[3,2-d]pyrimidines derivatives for the treatment of viral infections
US10377738B2 (en) 2013-05-24 2019-08-13 Janssen Sciences Ireland Unlimited Company Pyridone derivatives for the treatment of viral infections and further diseases
US10385054B2 (en) 2013-06-27 2019-08-20 Janssen Sciences Ireland Unlimited Company Pyrrolo[3,2-d]pyrimidine derivatives for the treatment of viral infections and other diseases
WO2020162705A1 (en) 2019-02-08 2020-08-13 성균관대학교산학협력단 Toll-like receptor 7 or 8 agonist-cholesterol complex, and use of same
US10968184B2 (en) 2016-09-29 2021-04-06 Janssen Sciences Ireland Unlimited Company Pyrimidine prodrugs for the treatment of viral infections and further diseases
US11053256B2 (en) 2016-07-01 2021-07-06 Janssen Sciences Ireland Unlimited Company Dihydropyranopyrimidines for the treatment of viral infections
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
WO2022031011A1 (en) 2020-08-04 2022-02-10 성균관대학교산학협력단 Kinetically acting adjuvant ensemble
WO2022031021A1 (en) 2020-08-04 2022-02-10 성균관대학교산학협력단 Mrna vaccine comprising adjuvant capable of kinetic control
US11597704B2 (en) 2018-03-01 2023-03-07 Janssen Sciences Ireland Unlimited Company 2,4-diaminoquinazoline derivatives and medical uses thereof

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 (17)

* Cited by examiner, † Cited by third party
Title
"Bioreversible Carriers in Drug Design", 1987, PERGAMON PRESS
"Comprehensive Medicinal Chemistry", vol. 5, 1990, PERGAMON PRESS
"Design of Pro-drugs", 1985, ELSEVIER
"Methods in Enzymology", vol. 42, 1985, ACADEMIC PRESS, pages: 309 - 396
"Protective Groups in Organic Chemistry", 1973, PLENUM 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", 1991, article "Design and Application of Pro-drugs", 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 LL-19
T. HIGUCHI; V. STELLA: "A.C.S. Symposium Series", vol. 14, article "Pro-Drugs as Novel Delivery Systems"
T.W. GREENE; P.G.M. WUTS: "Protective Groups in Organic Synthesis", 1999, WILEY-INTERSCIENCE

Cited By (66)

* 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
US11541050B2 (en) 2011-04-08 2023-01-03 Janssen Sciences Ireland Uc Pyrimidine derivatives for the treatment of viral infections
US10272085B2 (en) 2011-04-08 2019-04-30 Janssen Sciences Ireland Uc Pyrimidine derivatives for the treatment of viral infections
US10780089B2 (en) 2011-04-08 2020-09-22 Janssen Sciences Ireland Uc Pyrimidine derivatives for the treatment of viral infections
US10420767B2 (en) 2011-04-08 2019-09-24 Janssen Sciences Ireland Uc Pyrimidine derivatives for the treatment of viral infections
US11104678B2 (en) 2011-11-09 2021-08-31 Janssen Sciences Ireland Unlimited Company Purine derivatives for the treatment of viral infections
US10280167B2 (en) 2011-11-09 2019-05-07 Janssen Sciences Ireland Uc Purine derivatives for the treatment of viral infections
US9376398B2 (en) 2012-05-18 2016-06-28 Sumitomo Dainippon Pharma Co., Ltd Carboxylic acid compounds
RU2636146C2 (en) * 2012-05-18 2017-11-21 Сумитомо Дайниппон Фарма Ко., Лтд. Carboxylic acids compounds
US10562861B2 (en) 2012-05-18 2020-02-18 Sumitomo Dainippon Pharma Co., Ltd. Carboxylic acid compounds
US11299465B2 (en) 2012-05-18 2022-04-12 Sumitomo Dainippon Pharma Co., Ltd. Carboxylic acid compounds
US20190119220A1 (en) * 2012-05-18 2019-04-25 Sumitomo Dainippon Pharma Co., Ltd. Carboxylic acid compounds
US10150743B2 (en) 2012-05-18 2018-12-11 Sumitomo Dainippon Pharma Co., Ltd. Carboxylic acid compounds
US10280180B2 (en) 2012-07-13 2019-05-07 Janssen Sciences Ireland Uc Macrocyclic purines for the treatment of viral infections
US10822349B2 (en) 2012-07-13 2020-11-03 Janssen Sciences Ireland Unlimited Company Macrocyclic purines for the treatment of viral infections
JP2015524462A (en) * 2012-08-10 2015-08-24 ヤンセン・サイエンシズ・アイルランド・ユーシー Alkylpyrimidine derivatives for treating viral infections and other diseases
CN109232439A (en) * 2012-08-10 2019-01-18 爱尔兰詹森科学公司 For treating the alkyl derivative of virus infection and other disease
WO2014023813A1 (en) * 2012-08-10 2014-02-13 Janssen R&D Ireland Alkylpyrimidine derivatives for the treatment of viral infections and further diseases
US9284304B2 (en) 2012-08-10 2016-03-15 Janssen Sciences Ireland Uc Substituted pyrimidines as toll-like receptor modulators
CN104781239A (en) * 2012-08-10 2015-07-15 爱尔兰詹森科学公司 Alkylpyrimidine derivatives for the treatment of viral infections and further diseases
EA029266B1 (en) * 2012-08-10 2018-02-28 Янссен Сайенсиз Айрлэнд Юси Alkylpyrimidine derivatives for the treatment of viral infections
AU2013301450B2 (en) * 2012-08-10 2018-01-18 Janssen Sciences Ireland Uc Alkylpyrimidine derivatives for the treatment of viral infections and further diseases
US10259814B2 (en) 2012-10-10 2019-04-16 Janssen Sciences Ireland Uc Pyrrolo[3,2-d]pyrimidine derivatives for the treatment of viral infections and other diseases
US11220504B2 (en) 2012-10-10 2022-01-11 Janssen Sciences Ireland Unlimited Company Pyrrolo[3,2-d] pyrimidine derivatives for the treatment of viral infections and other diseases
US10253003B2 (en) 2012-11-16 2019-04-09 Janssen Sciences Ireland Uc Heterocyclic substituted 2-amino quinazoline derivatives for the treatment of viral infections
US10723707B2 (en) 2012-11-16 2020-07-28 Janssen Sciences Ireland Unlimited Company Heterocyclic substituted 2-amino quinazoline derivatives for the treatment of viral infections
US10647684B2 (en) 2013-02-21 2020-05-12 Janssen Sciences Ireland Unlimited Company 2-aminopyrimidine derivatives for the treatment of viral infections
US10259793B2 (en) 2013-02-21 2019-04-16 Janssen Sciences Ireland Uc 2-aminopyrimidine derivatives for the treatment of viral infections
US11702426B2 (en) 2013-03-29 2023-07-18 Janssen Sciences Ireland Unlimited Company Macrocyclic deaza-purinones for the treatment of viral infections
US10266543B2 (en) 2013-03-29 2019-04-23 Janssen Sciences Ireland Uc Macrocyclic deaza-purinones for the treatment of viral infections
US10829494B2 (en) 2013-03-29 2020-11-10 Janssen Sciences Ireland Unlimited Company Macrocyclic deaza-purinones for the treatment of viral infections
US10377738B2 (en) 2013-05-24 2019-08-13 Janssen Sciences Ireland Unlimited Company Pyridone derivatives for the treatment of viral infections and further diseases
US10865193B2 (en) 2013-05-24 2020-12-15 Janssen Sciences Ireland Unlimited Company Pyridone derivatives for the treatment of viral infections and further diseases
US10385054B2 (en) 2013-06-27 2019-08-20 Janssen Sciences Ireland Unlimited Company Pyrrolo[3,2-d]pyrimidine derivatives for the treatment of viral infections and other diseases
US10781216B2 (en) 2013-06-27 2020-09-22 Janssen Sciences Ireland Unlimited Company Pyrrolo [3,2-d]pyrimidine derivatives for the treatment of viral infections and other diseases
US10822347B2 (en) 2013-07-30 2020-11-03 Janssen Sciences Ireland Unlimited Company Thieno[3,2-d]pyrimidines derivatives for the treatment of viral infections
US10316043B2 (en) 2013-07-30 2019-06-11 Janssen Sciences Ireland Unlimited Company Thieno[3,2-d]pyrimidines derivatives for the treatment of viral infections
CN108290845B (en) * 2015-10-07 2021-08-03 大日本住友制药株式会社 Pyrimidine compounds
EP3868741A1 (en) 2015-10-07 2021-08-25 Sumitomo Dainippon Pharma Co., Ltd. Pyrimidine compound
WO2017061532A1 (en) * 2015-10-07 2017-04-13 大日本住友製薬株式会社 Pyrimidine compound
AU2016334826B2 (en) * 2015-10-07 2020-08-27 Sumitomo Pharma Co., Ltd. Pyrimidine compound
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
KR20180056774A (en) * 2015-10-07 2018-05-29 다이닛본 스미토모 세이야꾸 가부시끼가이샤 Pyrimidine compound
JP7195398B2 (en) 2015-10-07 2022-12-23 住友ファーマ株式会社 pyrimidine compound
US11266738B2 (en) 2015-10-07 2022-03-08 Sumitomo Dainippon Pharma Co., Ltd. Substituted pyrimidines as vaccine adjuvants
JPWO2017061532A1 (en) * 2015-10-07 2018-08-02 大日本住友製薬株式会社 Pyrimidine compounds
KR102651207B1 (en) * 2015-10-07 2024-03-25 스미토모 파마 가부시키가이샤 pyrimidine compounds
RU2759917C2 (en) * 2015-10-07 2021-11-18 Сумитомо Дайниппон Фарма Ко., Лтд. Pyrimidine compound
CN108290845A (en) * 2015-10-07 2018-07-17 大日本住友制药株式会社 Pyrimidine compound
JP2022010266A (en) * 2015-10-07 2022-01-14 大日本住友製薬株式会社 Pyrimidine compound
US11911466B2 (en) 2015-10-07 2024-02-27 Sumitomo Pharma Co., Ltd. Substituted pyrimidines as vaccine adjuvants
US11053256B2 (en) 2016-07-01 2021-07-06 Janssen Sciences Ireland Unlimited Company Dihydropyranopyrimidines for the treatment of viral infections
US10968184B2 (en) 2016-09-29 2021-04-06 Janssen Sciences Ireland Unlimited Company Pyrimidine prodrugs for the treatment of viral infections and further diseases
US11253476B2 (en) 2017-03-29 2022-02-22 Sumitomo Dainippon Pharma Co., Ltd. Vaccine adjuvant formulation
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
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
US11597704B2 (en) 2018-03-01 2023-03-07 Janssen Sciences Ireland Unlimited Company 2,4-diaminoquinazoline derivatives and medical uses thereof
WO2020162705A1 (en) 2019-02-08 2020-08-13 성균관대학교산학협력단 Toll-like receptor 7 or 8 agonist-cholesterol complex, and use of same
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
WO2022031011A1 (en) 2020-08-04 2022-02-10 성균관대학교산학협력단 Kinetically acting adjuvant ensemble
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

Similar Documents

Publication Publication Date Title
WO2012066335A1 (en) Phenol compounds als toll -like receptor 7 agonists
US11299465B2 (en) Carboxylic acid compounds
WO2012066336A1 (en) Benzylamine compounds as toll -like receptor 7 agonists
US9533978B2 (en) 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
WO2012067268A1 (en) Cyclic amide compounds and their use in the treatment of disease
US8198285B2 (en) Pyrazine derivatives
US11866436B2 (en) Benzo[c][2,6]naphthyridine derivatives, compositions and therapeutic uses thereof
US20210122746A1 (en) Febrifugine derivatives

Legal Events

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

Ref document number: 11791619

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: 11791619

Country of ref document: EP

Kind code of ref document: A1