WO2010004517A1 - Triazole derivatives useful for the treatment of diseases - Google Patents

Triazole derivatives useful for the treatment of diseases Download PDF

Info

Publication number
WO2010004517A1
WO2010004517A1 PCT/IB2009/052986 IB2009052986W WO2010004517A1 WO 2010004517 A1 WO2010004517 A1 WO 2010004517A1 IB 2009052986 W IB2009052986 W IB 2009052986W WO 2010004517 A1 WO2010004517 A1 WO 2010004517A1
Authority
WO
WIPO (PCT)
Prior art keywords
formula
pharmaceutically acceptable
compound
ethyl
asthma
Prior art date
Application number
PCT/IB2009/052986
Other languages
French (fr)
Inventor
Lyn Howard Jones
Dannielle Frances Roberts
Ross Sinclair Strang
Original Assignee
Pfizer 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
Priority to AP2010005508A priority Critical patent/AP2010005508A0/en
Priority to BRPI0915897A priority patent/BRPI0915897A2/en
Priority to MX2011000352A priority patent/MX2011000352A/en
Priority to ES09786555T priority patent/ES2398428T3/en
Priority to NZ589613A priority patent/NZ589613A/en
Priority to EP09786555A priority patent/EP2300443B1/en
Priority to CN2009801271934A priority patent/CN102089290B/en
Application filed by Pfizer Limited filed Critical Pfizer Limited
Priority to KR1020117000612A priority patent/KR101235962B1/en
Priority to CA2726686A priority patent/CA2726686C/en
Priority to EA201001821A priority patent/EA201001821A1/en
Priority to JP2011517301A priority patent/JP5608162B2/en
Priority to AU2009269610A priority patent/AU2009269610B2/en
Priority to DK09786555.4T priority patent/DK2300443T3/en
Publication of WO2010004517A1 publication Critical patent/WO2010004517A1/en
Priority to IL209730A priority patent/IL209730A/en
Priority to TNP2010000612A priority patent/TN2010000612A1/en
Priority to MA33507A priority patent/MA32466B1/en
Priority to ZA2011/00534A priority patent/ZA201100534B/en
Priority to HK11108477.2A priority patent/HK1154382A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D249/00Heterocyclic compounds containing five-membered rings having three nitrogen atoms as the only ring hetero atoms
    • C07D249/02Heterocyclic compounds containing five-membered rings having three nitrogen atoms as the only ring hetero atoms not condensed with other rings
    • C07D249/081,2,4-Triazoles; Hydrogenated 1,2,4-triazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41961,2,4-Triazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/04Drugs for disorders of the respiratory system for throat disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/08Bronchodilators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/16Central respiratory analeptics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/06Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings

Definitions

  • This invention relates to compounds of general formula (1 ):
  • A, X, Y, n, R 1 and R 2 have the meanings indicated below, and to processes and intermediates for the preparation of, compositions containing and the uses of such derivatives.
  • ⁇ 2 adrenergic agonists and cholinergic muscarinic antagonists are well-established therapeutic agents for the treatment of obstructive respiratory diseases such as COPD and Asthma.
  • obstructive respiratory diseases such as COPD and Asthma.
  • Currently used inhaled ⁇ 2 agonists include both short acting agents such as salbutamol (q.i.d.), and terbutaline (t.i.d) and longer acting agents such as salmeterol and formoterol (b.i.d.) and produce bronchodilation via stimulation of adrenergic receptors on airway smooth muscle.
  • Inhaled muscarinic antagonists in clinical use include the short acting ipratropium bromide (q.i.d.), oxitropium bromide (q.i.d) and the long acting tiotropium (q.d.).
  • Muscarinic antagonists produce bronchodilation by inhibiting the cholinergic tone of airways primarily by antagonising the action of acetylcholine on muscarinic receptors present on airway smooth muscle.
  • the invention relates to the compounds of general formula (1 ):
  • A is selected from:
  • X is -(CH 2 ) m - where m is an integer comprised between 7 to 12 inclusive, or is of formula:
  • Y is selected from wherein ** and *** represent the attachment points, ** being linked to X; n is 0 or 1 ;
  • R 1 is selected from cyclopentyl, cyclohexyl, phenyl, furanyl and thiophenyl; and,
  • R 2 is selected from phenyl, furanyl and thiophenyl; or the p harmaceutically acceptable salts thereof, or the pharmaceutically acceptable solvates of said compounds or salts.
  • the compounds of formula (1) are ⁇ 2 adrenergic receptor agonists and muscarinic receptor antagonists that are particularly useful for the treatment of diseases and/or conditions involving said receptors, by showing excellent potency, in particular when administered via the inhalation route.
  • the amine of formula (2) is reacted with a bromide of formula (3) optionally in the presence of a solvent or mixture of solvents (e.g. dimethyl sulphoxide, toluene, ⁇ /, ⁇ /-dimethylformamide, propionitrile, acetonitrile, butyronitrile, dichloromethane), optionally in the presence of a suitable base (e.g. triethylamine, diisopropylethylamine, potassium carbonate, sodium hydrogen carbonate), at a temperature comprised between 8O 0 C and 120 0 C, for 12 to 72 hours.
  • a solvent or mixture of solvents e.g. dimethyl sulphoxide, toluene, ⁇ /, ⁇ /-dimethylformamide, propionitrile, acetonitrile, butyronitrile, dichloromethane
  • a suitable base e.g. triethylamine, diisopropylethylamine, potassium carbonate, sodium
  • the protecting groups such as tert- butyldimethylsilyl and benzyl can then be removed using standard methodology for cleaving oxygen protecting groups such as those found in the text book T. W. Greene, Protective Groups in Organic Synthesis (Wiley-lnterscience Publication, 1981 ).
  • the amine of formula (2) may be prepared from the corresponding protected amine of formula (4):
  • R a and R represent suitable protecting groups such as tert-butoxycarbonyl or together represent phthalimide.
  • the amine of formula (4) is deprotected using standard methodology for cleaving nitrogen protecting groups such as those found in the text book T. W. Greene, Protective Groups in Organic Synthesis (Wiley-lnterscience Publication, 1981 ).
  • the amine of formula (4) may be prepared by reacting the corresponding amine of formula (5):
  • R a and R b are as previously defined and LG is a suitable leaving group such as bromide or mesylate
  • the amine of formula (5) is reacted with a compound of formula (6) in a suitable solvent (e.g. dimethyl sulphoxide, toluene, ⁇ /, ⁇ /-dimethylformamide, propionitrile, acetonitrile, methyl ethyl ketone), in the presence of a suitable base (e.g. triethylamine, diisopropylethylamine) and optionally in the presence of a nucleophilic additive (e.g. sodium iodide) at a temperature comprised between 5O 0 C and 90 0 C, for 18 to 48 hours.
  • a suitable solvent e.g. dimethyl sulphoxide, toluene, ⁇ /, ⁇ /-dimethylformamide, propionitrile, acetonitrile, methyl ethyl ketone
  • a suitable base e.g. triethylamine, diisopropylethylamine
  • the compounds of formula (5) may also be used as muscarinic antagonists. In particular they may be useful for the treatment of diseases which can be alleviated by the modulation of the activity of the muscarinic receptor.
  • the compounds of formula (6) where LG is bromide may be prepared by reacting the corresponding amine nucleophiles R a R b NH with the corresponding dibromide of formula (7):
  • phthalimide or di-tert-butyl iminodicarboxylate (R a R b NH) is reacted with a suitable base such as sodium hydride (60% dispersion in oil), in a suitable solvent such as N, N- dimethylformamide or tetrahydrofuran, at a temperature comprised between 0 0 C and room temperature for up to 1 hour, followed by addition of the dibromide of formula (7), at a temperature comprised between 0 0 C and 150 0 C, for 6-48 hours.
  • a suitable base such as sodium hydride (60% dispersion in oil)
  • a suitable solvent such as N, N- dimethylformamide or tetrahydrofuran
  • the amines of formula R a R b NH are commercially available.
  • Compound of formula (10) may be prepared from compounds of formula (8) and (9) by Heck reaction (process step (i)).
  • Typical conditions comprise reaction of compound (8) with compound (9), tri-o- tolylphosphine, palladium(ll)acetate and a suitable base such as diisopropylethylamine, in a suitable solvent such as acetonitrile, at 90 0 C for 21 hours.
  • Compound of formula (11 ) may be prepared from compound of formula (10) by hydrogenation (process step (ii)).
  • Typical conditions comprise reaction of compound (10) with ammonium formate and 30% palladium hydroxide on carbon, in suitable solvents such as ethyl acetate and ethanol, at 8O 0 C for 18 hours.
  • Alternative conditions comprise reaction of compound (10) with hydrogen gas and rhodium tris(triphenylphosphine)chloride, in suitable solvents such as ethyl acetate and ethanol, at 20psi and room temperature for 24 hours.
  • Compound of formula (6a) where LG is bromide may be prepared from compound of formula (11 ) by bromination (process step (iii)).
  • Typical conditions comprise reaction of compound (11 ) with phosphorous tribromide, in a suitable solvent such as toluene, at reflux for 4 hours.
  • Compound of formula (6a) where LG is mesylate may be prepared from compound of formula (1 1 ) by mesylation (process step (iii)).
  • Typical conditions comprise reaction of compound (1 1 ) with methanesulfonyl chloride, in a suitable solvent such as methylethyl ketone, with a suitable base such as triethylamine, at O 0 C to room temperature for 1 -4 hours.
  • the amines of formula (5) may be prepared from the corresponding protected amines of formula (12): where R c represents a suitable protecting group such as tert-butoxycarbonyl or benzyloxycarbonyl.
  • the amine of formula (12) is deprotected using standard methodology for cleaving nitrogen protecting groups such as those found in the text book T. W. Greene, Protective Groups in Organic Synthesis (Wiley-lnterscience Publication, 1981 ).
  • the amines of formula (12) may be prepared by reacting the corresponding compounds of formula (13):
  • LG is a suitable leaving group such as bromide or mesylate, with compounds of formula (14):
  • compounds of formula (13) are reacted with compounds of formula (14) in a suitable solvent (e.g. ⁇ /, ⁇ /-dimethylformamide, propionitrile, acetonitrile, acetone), in the presence of a suitable base (e.g. potassium carbonate, caesium carbonate, triethylamine, diisopropylethylamine) at a temperature between 7O 0 C and 15O 0 C, for 5 to 48 hours.
  • a suitable solvent e.g. ⁇ /, ⁇ /-dimethylformamide, propionitrile, acetonitrile, acetone
  • a suitable base e.g. potassium carbonate, caesium carbonate, triethylamine, diisopropylethylamine
  • the compounds of formula (13) where R c is tert-butoxycarbonyl, p and q represent 1 or 2, n is 0 and LG is mesylate are commercially available.
  • the compounds of formula (13) where R c is tert- butoxycarbonyl, p and q are 1 , n is 1 and LG is mesylate may be prepared as described in U.S. Pat. Appl. Publ., 2005101586.
  • the compounds of formula (13) where R c is tert-butoxycarbonyl, p is 1 , q is 2, n is 1 and LG is bromide or mesylate are commercially available.
  • the compounds of formula (13) where R c is tert-butoxycarbonyl or benzyloxycarbonyl, p and q are 2, n is 1 and LG is bromide are commercially available.
  • compounds of formula (14) may be prepared from the corresponding compounds of formula (17):
  • the compounds of formula (17) may be prepared by reacting the compound of formula (18):
  • the compound of formula (18) is reacted with a suitable base such as n-butyl lithium, in a suitable solvent such as tetrahydrofuran, at a temperature between -78 0 C and room temperature for 1 hour, followed by addition of a compound of formula (19) in tetrahydrofuran at a temperature between -78 0 C and room temperature for 18 hours.
  • a suitable base such as n-butyl lithium
  • a suitable solvent such as tetrahydrofuran
  • the compounds of formula (18) may be prepared as described in Tetrahedron: Asymmetry, 8(9), 1491- 1500; 1997.
  • A is of formula:
  • X is (CH 2 ) 9 or is of formula:
  • X is (CH 2 ⁇ .
  • X is of formula:
  • Y is of formula: wherein ** and *** represent the attachment points, ** being linked to X.
  • R 1 is phenyl or cyclohexyl.
  • R 2 is phenyl
  • n 1
  • the carbon atom bearing these two substituents can be in the (R) or (S) configuration.
  • the carbon atom bearing R 1 and R 2 is in the (R) configuration when R 1 and R 2 are different.
  • A is of formula: wherein * represent the attachment point of A to the carbon bearing the hydroxy;
  • X is-(CH 2 ) 9 - or is of formula:
  • Y is of formula: wherein ** and *** represent the attachment points, ** being linked to X; n is 1;
  • R 1 is cyclohexyl or phenyl and R 2 is phenyl; or the pharmaceutically acceptable salts thereof, or the pharmaceutically acceptable solvates of said compounds or salts, are further preferred.
  • Pharmaceutically acceptable salts of the compounds of formula (1 ) include the acid addition and base salts thereof. Suitable acid addition salts are formed from acids which form non-toxic salts. Examples include the acetate, aspartate, benzoate, besylate, bicarbonate/carbonate, bisulphate/sulphate, borate, camsylate, citrate, edisylate, esylate, formate, fumarate, gluceptate, gluconate, glucuronate, hexafluorophosphate, hibenzate, hydrochloride/chloride, hydrobromide/bromide, hydroiodide/iodide, isethionate, lactate, malate, maleate, malonate, mesylate, methylsulphate, naphthylate, 1 ,5- naphthalenedisulfonate, 2-napsylate, nicotinate, nitrate, orotate, oxalate, palmitate, pamoate, phosphat
  • Suitable base salts are formed from bases which form non-toxic salts. Examples include the aluminium, arginine, benzathine, calcium, choline, diethylamine, diolamine, glycine, lysine, magnesium, meglumine, olamine, potassium, sodium, tromethamine and zinc salts. Hemisalts of acids and bases may also be formed, for example, hemisulphate and hemicalcium salts.
  • suitable salts see "Handbook of Pharmaceutical Salts: Properties, Selection, and Use” by Stahl and Wermuth (Wiley-VCH, Weinheim, Germany, 2002).
  • the resulting salt may precipitate out and be collected by filtration or may be recovered by evaporation of the solvent.
  • the degree of ionisation in the resulting salt may vary from completely ionised to almost non-ionised.
  • the compounds of the invention may exist in both unsolvated and solvated forms.
  • 'solvate' is used herein to describe a molecular complex comprising the compound of the invention or a salt thereof and a stoichiometric amount of one or more pharmaceutically acceptable solvent molecules, for example, ethanol.
  • solvent molecules for example, ethanol.
  • 'hydrate' is employed when said solvent is water.
  • complexes such as clathrates, drug-host inclusion complexes wherein, in contrast to the aforementioned solvates, the drug and host are present in stoichiometric or non-stoichiometric amounts.
  • complexes of the drug containing two or more organic and/or inorganic components which may be in stoichiometric or non-stoichiometric amounts.
  • the resulting complexes may be ionised, partially ionised, or non-ionised.
  • references to compounds of formula (1 ) include references to salts, solvates and complexes thereof and to solvates and complexes of salts thereof.
  • the compounds of the invention include compounds of formula (1 ) as hereinbefore defined, including all polymorphs and crystal habits thereof, prodrugs and isomers thereof (including optical, geometric and tautomeric isomers) as hereinafter defined and isotopically-labeled compounds of formula (1 ).
  • 'pro-drugs' of the compounds of formula (1 ) are also within the scope of the invention.
  • certain derivatives of compounds of formula (1 ) which may have little or no pharmacological activity themselves can, when administered into or onto the body, be converted into compounds of formula (1 ) having the desired activity, for example, by hydrolytic cleavage.
  • Such derivatives are referred to as 'prodrugs'.
  • Further information on the use of prodrugs may be found in 'Pro-drugs as Novel Delivery Systems, Vol. 14, ACS Symposium Series (T. Higuchi and W. Stella) and 'Bioreversible Carriers in Drug Design', Pergamon Press, 1987 (ed. E. B Roche, American Pharmaceutical Association).
  • Prodrugs in accordance with the invention can, for example, be produced by replacing appropriate functionalities present in the compounds of formula (1 ) with certain moieties known to those skilled in the art as 'pro-moieties' as described, for example, in "Design of Prodrugs" by H. Bundgaard (Elsevier, 1985).
  • prodrugs in accordance with the invention include:
  • the compound of formula (1 ) contains a primary or secondary amino functionality (-NH 2 or -NHR where R ⁇ H), an amide thereof, for example, a compound wherein, as the case may be, one or both hydrogens of the amino functionality of the compound of formula (1 ) is/are replaced by (C- ⁇ -Cio)alkanoyl.
  • metabolites of compounds of formula (1 ) that is, compounds formed in vivo upon administration of the drug.
  • Some examples of metabolites in accordance with the invention include (i) where the compound of formula (1 ) contains a methyl group, an hydroxymethyl derivative thereof (-CH 3 ⁇ -CH 2 OH): (ii) where the compound of formula (1 ) contains an alkoxy group, an hydroxy derivative thereof (-
  • the compounds of formula (1 ) wherein R 1 and R 2 are different can exist as stereoisomers. Where structural isomers are interconvertible via a low energy barrier, tautomeric isomerism ('tautomerism') can occur. This can take the form of proton tautomerism in compounds of formula (1 ) containing, for example, an imino, keto, or oxime group, or so-called valence tautomerism in compounds which contain an aromatic moiety. It follows that a single compound may exhibit more than one type of isomerism.
  • the present invention includes the compounds of formula (1 ) wherein the triazole moiety may be present as different regio-isomers.
  • the racemate (or a racemic precursor) may be reacted with a suitable optically active compound, for example, an alcohol, or, in the case where the compound of formula (1 ) contains an acidic or basic moiety, an acid or base such as tartaric acid or 1-phenylethylamine.
  • a suitable optically active compound for example, an alcohol, or, in the case where the compound of formula (1 ) contains an acidic or basic moiety, an acid or base such as tartaric acid or 1-phenylethylamine.
  • the resulting diastereomeric mixture may be separated by chromatography and/or fractional crystallization and one or both of the diastereoisomers converted to the corresponding pure enantiomer(s) by means well known to a skilled person.
  • Chiral compounds of the invention may be obtained in enantiomerically- enriched form using chromatography, typically HPLC, on an asymmetric resin with a mobile phase consisting of a hydrocarbon, typically heptane or hexane, containing from 0 to 50% by volume of isopropanol, typically from 2% to 20%, and from 0 to 5% by volume of an alkylamine, typically 0.1 % diethylamine. Concentration of the eluate affords the enriched mixture.
  • chromatography typically HPLC
  • a mobile phase consisting of a hydrocarbon, typically heptane or hexane, containing from 0 to 50% by volume of isopropanol, typically from 2% to 20%, and from 0 to 5% by volume of an alkylamine, typically 0.1 % diethylamine.
  • Stereoisomeric conglomerates may be separated by conventional techniques known to those skilled in the art - see, for example, "Stereochemistry of Organic Compounds" by E. L. Eliel (Wiley, New York, 1994).
  • the present invention includes all pharmaceutically acceptable isotopically-labelled compounds of formula (1 ) wherein one or more atoms are replaced by atoms having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number which predominates in nature.
  • isotopes suitable for inclusion in the compounds of the invention include isotopes of hydrogen, such as 2 H and 3 H, carbon, such as 11 C, 13 C and 14 C, nitrogen, such as 13 N and 15 N, oxygen, such as 15 O, 17 O and 18 O, and sulphur, such as 35 S.
  • isotopically-labelled compounds of formula (1 ), for example, those incorporating a radioactive isotope, are useful in drug and/or substrate tissue distribution studies.
  • the radioactive isotopes tritium, i.e. 3 H, and carbon-14, i.e. 14 C, are particularly useful for this purpose in view of their ease of incorporation and ready means of detection.
  • substitution with heavier isotopes such as deuterium, i.e. 2 H, may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements, and hence may be preferred in some circumstances.
  • Isotopically-labeled compounds of formula (1 ) can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described in the accompanying Examples and Preparations using an appropriate isotopically-labeled reagents in place of the non- labeled reagent previously employed.
  • solvates in accordance with the invention include those wherein the solvent of crystallization may be isotopically substituted, e.g. D 2 O, d 6 -acetone, d 6 -DMSO.
  • the compounds of formula (1 ), the pharmaceutically acceptable salts thereof and/or the pharmaceutically acceptable solvates of said compounds or salts, are valuable pharmaceutically active compounds, which are suitable for the therapy and prophylaxis of numerous disorders in which the ⁇ 2 receptor and/or the muscarinic receptor are involved .
  • these compounds are useful for the therapy and prophylaxis of numerous disorders in which agonism of the ⁇ 2 receptor and antagonism of the muscarinic receptor may induce benefit, in particular the allergic and non-allergic airways diseases.
  • Compounds of the invention intended for pharmaceutical use may be administered as crystalline or amorphous products. They may be obtained, for example, as solid plugs, powders, or films by methods such as precipitation, crystallization, freeze drying, spray drying, or evaporative drying. Microwave or radio frequency drying may be used for this purpose.
  • excipient is used herein to describe any ingredient other than the compound(s) of the invention. The choice of excipient will to a large extent depend on factors such as the particular mode of administration, the effect of the excipient on solubility and stability, and the nature of the dosage form.
  • compositions suitable for the delivery of compounds of the present invention and methods for their preparation will be readily apparent to those skilled in the art. Such compositions and methods for their preparation may be found, for example, in 'Remington's Pharmaceutical Sciences', 19th Edition (Mack Publishing Company, 1995).
  • the compounds of the invention may be administered orally.
  • Oral administration may involve swallowing, so that the compound enters the gastrointestinal tract, or buccal or sublingual administration may be employed by which the compound enters the blood stream directly from the mouth.
  • Formulations suitable for oral administration include solid formulations such as tablets, capsules containing particulates, liquids, or powders, lozenges (including liquid-filled), chews, multi- and nano- particulates, gels, solid solution, liposome, films, ovules, sprays and liquid formulations.
  • Liquid formulations include suspensions, solutions, syrups and elixirs. Such formulations may be employed as fillers in soft or hard capsules and typically comprise a carrier, for example, water, ethanol, polyethylene glycol, propylene glycol, methylcellulose, or a suitable oil, and one or more emulsifying agents and/or suspending agents. Liquid formulations may also be prepared by the reconstitution of a solid, for example, from a sachet.
  • the compounds of the invention may also be used in fast-dissolving, fast-disintegrating dosage forms such as those described in Expert Opinion in Therapeutic Patents, 1 1 (6), 981-986, by Liang and Chen (2001 ).
  • the drug may make up from 1 weight % to 80 weight % of the dosage form, more typically from 5 weight % to 60 weight % of the dosage form.
  • tablets generally contain a disintegrant.
  • disinteg rants include sodium starch glycolate, sodium carboxymethyl cellulose, calcium carboxymethyl cellulose, croscarmellose sodium, crospovidone, polyvinylpyrrolidone, methyl cellulose, microcrystalline cellulose, lower alkyl -substituted hydroxypropyl cellulose, starch, pregelatinised starch and sodium alginate.
  • the disintegrant will comprise from 1 weight % to 25 weight %, preferably from 5 weight % to 20 weight % of the dosage form.
  • Binders are generally used to impart cohesive qualities to a tablet formulation. Suitable binders include microcrystalline cellulose, gelatin, sugars, polyethylene glycol, natural and synthetic gums, polyvinylpyrrolidone, pregelatinised starch, hydroxypropyl cellulose and hydroxypropyl methylcellulose. Tablets may also contain diluents, such as lactose (monohydrate, spray-dried monohydrate, anhydrous and the like), mannitol, xylitol, dextrose, sucrose, sorbitol, microcrystalline cellulose, starch and dibasic calcium phosphate dihydrate.
  • lactose monohydrate, spray-dried monohydrate, anhydrous and the like
  • mannitol xylitol
  • dextrose sucrose
  • sorbitol microcrystalline cellulose
  • starch dibasic calcium phosphate dihydrate
  • Tablets may also optionally comprise surface active agents, such as sodium lauryl sulfate and polysorbate 80, and glidants such as silicon dioxide and talc.
  • surface active agents such as sodium lauryl sulfate and polysorbate 80
  • glidants such as silicon dioxide and talc.
  • surface active agents may comprise from 0.2 weight % to 5 weight % of the tablet, and glidants may comprise from 0.2 weight % to 1 weight % of the tablet.
  • Tablets also generally contain lubricants such as magnesium stearate, calcium stearate, zinc stearate, sodium stearyl fumarate, and mixtures of magnesium stearate with sodium lauryl sulphate.
  • Lubricants generally comprise from 0.25 weight % to 10 weight %, preferably from 0.5 weight % to 3 weight % of the tablet.
  • ingredients include anti-oxidants, colourants, flavouring agents, preservatives and taste- masking agents.
  • Exemplary tablets contain up to about 80% drug, from about 10 weight % to about 90 weight % binder, from about 0 weight % to about 85 weight % diluent, from about 2 weight % to about 10 weight % disintegrant, and from about 0.25 weight % to about 10 weight % lubricant.
  • Tablet blends may be compressed directly or by roller to form tablets. Tablet blends or portions of blends may alternatively be wet-, dry-, or melt-granulated, melt congealed, or extruded before tabletting.
  • the final formulation may comprise one or more layers and may be coated or uncoated; it may even be encapsulated.
  • Consumable oral films for human or veterinary use are typically pliable water-soluble or water-swellable thin film dosage forms which may be rapidly dissolving or mucoadhesive and typically comprise a compound of formula (1 ), a film-forming polymer, a binder, a solvent, a humectant, a plasticiser, a stabiliser or emulsifier, a viscosity-modifying agent and a solvent. Some components of the formulation may perform more than one function.
  • the compound of formula (1 ) may be water-soluble or insoluble.
  • a water-soluble compound typically comprises from 1 weight % to 80 weight %, more typically from 20 weight % to 50 weight %, of the solutes. Less soluble compounds may comprise a greater proportion of the composition, typically up to 88 weight % of the solutes.
  • the compound of formula (1 ) may be in the form of multiparticulate beads.
  • the film-forming polymer may be selected from natural polysaccharides, proteins, or synthetic hydrocolloids and is typically present in the range 0.01 to 99 weight %, more typically in the range 30 to 80 weight %.
  • ingredients include anti-oxidants, colorants, flavourings and flavour enhancers, preservatives, salivary stimulating agents, cooling agents, co-solvents (including oils), emollients, bulking agents, anti-foaming agents, surfactants and taste-masking agents.
  • Films in accordance with the invention are typically prepared by evaporative drying of thin aqueous films coated onto a peelable backing support or paper. This may be done in a drying oven or tunnel, typically a combined coater dryer, or by freeze-drying or vacuuming.
  • Solid formulations for oral administration may be formulated to be immediate and/or modified release.
  • Modified release formulations include delayed-, sustained-, pulsed-, controlled-, targeted and programmed release.
  • Suitable modified release formulations for the purposes of the invention are described in US Patent No. 6,106,864. Details of other suitable release technologies such as high energy dispersions and osmotic and coated particles are to be found in Pharmaceutical Technology On-line, 25(2), 1-14, by Verma et al (2001 ). The use of chewing gum to achieve controlled release is described in WO 00/35298.
  • the compounds of the invention may also be administered directly into the blood stream, into muscle, or into an internal organ.
  • Suitable means for parenteral administration include intravenous, intraarterial, intraperitoneal, intrathecal, intraventricular, intraurethral, intrasternal, intracranial, intramuscular and subcutaneous.
  • Suitable devices for parenteral administration include needle (including microneedle) injectors, needle-free injectors and infusion techniques.
  • Parenteral formulations are typically aqueous solutions which may contain excipients such as salts, carbohydrates and buffering agents (preferably to a pH of from 3 to 9), but, for some applications, they may be more suitably formulated as a sterile non-aqueous solution or as a dried form to be used in conjunction with a suitable vehicle such as sterile, pyrogen-free water.
  • excipients such as salts, carbohydrates and buffering agents (preferably to a pH of from 3 to 9)
  • a suitable vehicle such as sterile, pyrogen-free water.
  • parenteral formulations under sterile conditions may readily be accomplished using standard pharmaceutical techniques well known to those skilled in the art.
  • solubility of compounds of formula (1 ) used in the preparation of parenteral solutions may be increased by the use of appropriate formulation techniques, such as the incorporation of solubility- enhancing agents.
  • Formulations for parenteral administration may be formulated to be immediate and/or modified release.
  • Modified release formulations include delayed-, sustained-, pulsed-, controlled-, targeted and programmed release.
  • compounds of the invention may be formulated as a solid, semi-solid, or thixotropic liquid for administration as an implanted depot providing modified release of the active compound.
  • examples of such formulations include drug-coated stents and poly(c//-lactic-coglycolic)acid (PGLA) microspheres.
  • the compounds of the invention may also be administered topically to the skin or mucosa, that is, dermally or transdermally.
  • Typical formulations for this purpose include gels, hydrogels, lotions, solutions, creams, ointments, dusting powders, dressings, foams, films, skin patches, wafers, implants, sponges, fibres, bandages and microemulsions. Liposomes may also be used.
  • Typical carriers include alcohol, water, mineral oil, liquid petrolatum, white petrolatum, glycerin, polyethylene glycol and propylene glycol. Penetration enhancers may be incorporated - see, for example, J Pharm Sci, 88 (10), 955-958 by Finnin and Morgan (October 1999).
  • topical administration include delivery by electroporation, iontophoresis, phonophoresis, sonophoresis and microneedle or needle-free (e.g. PowderjectTM, BiojectTM, etc.) injection.
  • Formulations for topical administration may be formulated to be immediate and/or modified release.
  • Modified release formulations include delayed-, sustained-, pulsed-, controlled-, targeted and programmed release.
  • the compounds of the invention can also be administered intranasally or by inhalation, typically in the form of a dry powder (either alone, as a mixture, for example, in a dry blend with lactose, or as a mixed component particle, for example, mixed with phospholipids, such as phosphatidylcholine) from a dry powder inhaler or as an aerosol spray from a pressurised container, pump, spray, atomiser (preferably an atomiser using electrohydrodynamics to produce a fine mist), or nebuliser, with or without the use of a suitable propellant, such as 1 ,1 ,1 ,2-tetrafluoroethane or 1 ,1 ,1 ,2,3,3,3-heptafluoropropane.
  • the powder may comprise a bioadhesive agent, for example, chitosan or cyclodextrin.
  • the pressurised container, pump, spray, atomizer, or nebuliser contains a solution or suspension of the compound(s) of the invention comprising, for example, ethanol, aqueous ethanol, or a suitable alternative agent for dispersing, solubilising, or extending release of the active, a propellant(s) as solvent and an optional surfactant, such as sorbitan trioleate, oleic acid, or an oligolactic acid.
  • a solution or suspension of the compound(s) of the invention comprising, for example, ethanol, aqueous ethanol, or a suitable alternative agent for dispersing, solubilising, or extending release of the active, a propellant(s) as solvent and an optional surfactant, such as sorbitan trioleate, oleic acid, or an oligolactic acid.
  • the drug product Prior to use in a dry powder or suspension formulation, the drug product is micronised to a size suitable for delivery by inhalation (typically less than 5 microns). This may be achieved by any appropriate comminuting method, such as spiral jet milling, fluid bed jet milling, supercritical fluid processing to form nanoparticles, high pressure homogenisation, or spray drying.
  • comminuting method such as spiral jet milling, fluid bed jet milling, supercritical fluid processing to form nanoparticles, high pressure homogenisation, or spray drying.
  • Capsules made, for example, from gelatin or hydroxypropylmethylcellulose
  • blisters and cartridges for use in an inhaler or insufflator may be formulated to contain a powder mix of the compound of the invention, a suitable powder base such as lactose or starch and a performance modifier such as I- leucine, mannitol, or magnesium stearate.
  • the lactose may be anhydrous or in the form of the monohydrate, preferably the latter.
  • Other suitable excipients include dextran, glucose, maltose, sorbitol, xylitol, fructose, sucrose and trehalose.
  • a suitable solution formulation for use in an atomiser using electrohydrodynamics to produce a fine mist may contain from 1 ⁇ g to 20mg of the compound of the invention per actuation and the actuation volume may vary from 1 ⁇ l to 10O ⁇ l.
  • a typical formulation may comprise a compound of formula (1 ), propylene glycol, sterile water, ethanol and sodium chloride.
  • Alternative solvents which may be used instead of propylene glycol include glycerol and polyethylene glycol.
  • Suitable flavours such as menthol and levomenthol, or sweeteners, such as saccharin or saccharin sodium, may be added to those formulations of the invention intended for inhaled/intranasal administration.
  • Formulations for inhaled/intranasal administration may be formulated to be immediate and/or modified release using, for example, PGLA.
  • Modified release formulations include delayed-, sustained-, pulsed-, controlled-, targeted and programmed release.
  • the dosage unit is determined by a prefilled capsule, blister or pocket or by a system that utilises a gravimetrically fed dosing chamber .
  • Units in accordance with the invention are typically arranged to administer a metered dose or "puff" containing from 1 to 5000 ⁇ g of (compound name here), or a salt thereof.
  • the overall daily dose will typically be in the range 1 ⁇ g to 20 mg which may be administered in a single dose or, more usually, as divided doses throughout the day.
  • the compounds of formula (1 ) are particularly suitable for an administration by inhalation, in particular using a dry powder inhaler.
  • the compounds of the invention may be administered rectally or vaginally, for example, in the form of a suppository, pessary, or enema.
  • Cocoa butter is a traditional suppository base, but various alternatives may be used as appropriate.
  • Formulations for rectal/vaginal administration may be formulated to be immediate and/or modified release.
  • Modified release formulations include delayed-, sustained-, pulsed-, controlled-, targeted and programmed release.
  • the compounds of the invention may also be administered directly to the eye or ear, typically in the form of drops of a micronised suspension or solution in isotonic, pH-adjusted, sterile saline.
  • Other formulations suitable for ocular and aural administration include ointments, biodegradable (e.g. absorbable gel sponges, collagen) and non-biodegradable (e.g. silicone) implants, wafers, lenses and particulate or vesicular systems, such as niosomes or liposomes.
  • a polymer such as crossed-linked polyacrylic acid, polyvinylalcohol, hyaluronic acid, a cellulosic polymer, for example, hydroxypropylmethylcellulose, hydroxyethylcellulose, or methyl cellulose, or a heteropolysaccharide polymer, for example, gelan gum, may be incorporated together with a preservative, such as benzalkonium chloride.
  • a preservative such as benzalkonium chloride.
  • Such formulations may also be delivered by iontophoresis.
  • Formulations for ocular/aural administration may be formulated to be immediate and/or modified release.
  • Modified release formulations include delayed-, sustained-, pulsed-, controlled-, targeted, or programmed release.
  • the compounds of the invention may be combined with soluble macromolecular entities, such as cyclodextrin and suitable derivatives thereof or polyethylene glycol-containing polymers, in order to improve their solubility, dissolution rate, taste-masking, bioavailability and/or stability for use in any of the aforementioned modes of administration.
  • soluble macromolecular entities such as cyclodextrin and suitable derivatives thereof or polyethylene glycol-containing polymers
  • Drug-cyclodextrin complexes are found to be generally useful for most dosage forms and administration routes. Both inclusion and non-inclusion complexes may be used.
  • the cyclodextrin may be used as an auxiliary additive, i.e. as a carrier, diluent, or solubiliser. Most commonly used for these purposes are alpha-, beta- and gamma- cyclodextrins, examples of which may be found in International Patent Applications Nos. WO 91/1 1172, WO 94/02518 and WO 98/55148.
  • compositions may conveniently be combined in the form of a kit suitable for co-administration of the compositions.
  • the kit of the invention comprises two or more separate pharmaceutical compositions, at least one of which contains a compound of formula (1 ) in accordance with the invention, and means for separately retaining said compositions, such as a container, divided bottle, or divided foil packet.
  • a container, divided bottle, or divided foil packet An example of such a kit is the familiar blister pack used for the packaging of tablets, capsules and the like.
  • the kit of the invention is particularly suitable for administering different dosage forms, for example parenteral, for administering the separate compositions at different dosage intervals, or for titrating the separate compositions against one another.
  • the kit typically comprises directions for administration and may be provided with a so-called memory aid.
  • the total daily dose of the compounds of the invention is typically in the range 0.001 mg to 5000mg depending, of course, on the mode of administration.
  • an intravenous daily dose may only require from 0.001 mg to 40mg.
  • the total daily dose may be administered in single or divided doses and may, at the physician's discretion, fall outside of the typical range given herein.
  • These dosages are based on an average human subject having a weight of about 65kg to 70kg. The physician will readily be able to determine doses for subjects whose weight falls outside this range, such as infants and the elderly.
  • references herein to "treatment” include references to curative, palliative and prophylactic treatment.
  • the compounds of the formula (1 ), or pharmaceutically acceptable salts thereof and/or pharmaceutically acceptable solvates of said compounds or salts can also be used as a combination with one or more additional therapeutic agents to be co-administered to a patient to obtain some particularly desired therapeutic end result such as the treatment of pathophysiologically-relevant disease processes including, but not limited to (i) bronchoconstriction, (ii) inflammation, (iii) allergy, (iv) tissue destruction, (v) signs and symptoms such as breathlessness, cough.
  • additional therapeutic agents including, but not limited to (i) bronchoconstriction, (ii) inflammation, (iii) allergy, (iv) tissue destruction, (v) signs and symptoms such as breathlessness, cough.
  • the second and more additional therapeutic agents may also be a compound of the formula (1 ), or pharmaceutically acceptable salts thereof and/or pharmaceutically acceptable solvates of said compounds or salts, or one or more ⁇ 2 agonists known in the art. More typically, the second and more therapeutic agents will be selected from a different class of therapeutic agents.
  • the terms "co-administration”, “co-administered” and “in combination with”, referring to the compounds of formula (1 ) and one or more other therapeutic agents is intended to mean, and does refer to and include the following:
  • Suitable examples of other therapeutic agents which may be used in combination with the compound(s) of formula (1 ), or pharmaceutically acceptable salts thereof and/or pharmaceutically acceptable solvates of said compounds or salts, include, but are by no means limited to:
  • LTRAs Leukotriene antagonists
  • Histamine receptor antagonists including H1 and H3 antagonists
  • PDE inhibitors e.g. PDE3, PDE4 and PDE5 inhibitors
  • COX inhibitors both non-selective and selective COX-1 or COX-2 inhibitors (NSAIDs);
  • Anti-tumor necrosis factor (anti-TNF- ⁇ ) agents (o) Anti-tumor necrosis factor (anti-TNF- ⁇ ) agents;
  • Adhesion molecule inhibitors including VLA-4 antagonists
  • MMPs matrix metalloproteases
  • cytokine signalling pathyways such as p38 MAP kinase, PI3 kinase, JAK kinase, syk kinase, EGFR or MK-2;
  • Agents that can be classed as mucolytics or anti-tussive (bb) Agents which enhance responses to inhaled corticosteroids; (cc) Antibiotics and antivral agents effective against micro-organisms which can colonise the respiratory tract;
  • HDAC inhibitors HDAC inhibitors; (ee) CXCR2 antagonists; (ff) lntegrin antagonists; (gg) Chemokines; (hh)Epithelial sodium channel (ENaC) blockers or Epithelial sodium channel (ENaC) inhibitors;
  • Adhesion factors including VLAM, ICAM, and ELAM.
  • cytokine signalling pathyways such as p38 MAP kinase or syk kinase;
  • LTRAs Leukotriene antagonists
  • glucocorticosteroids in particular inhaled glucocorticosteroids with reduced systemic side effects, including prednisone, prednisolone, flunisolide, triamcinolone acetonide, beclomethasone dipropionate, budesonide, fluticasone propionate, ciclesonide, and mometasone furoate, or muscarinic M3 receptor antagonists or anticholinergic agents including in particular ipratropium salts, namely bromide, tiotropium salts, namely bromide, oxitropium salts, namely bromide, perenzepine, and telenzepine are further preferred.
  • ipratropium salts namely bromide, tiotropium salts, namely bromide, oxitropium salts, namely bromide, perenzepine, and telenzepine
  • the compounds of formula (1 ) have the ability to interact with the ⁇ 2 receptor and cholinergic muscarinic receptors, and thereby have a wide range of therapeutic applications, as described further below, because of the essential role which the ⁇ 2 receptor and muscarinic receptors play in the physiology of all mammals.
  • a further aspect of the present invention relates to the compounds of formula (1 ), or pharmaceutically acceptable salts thereof and/or pharmaceutically acceptable solvates of said compounds or salts, for use in the treatment of diseases, disorders, and conditions in which the ⁇ 2 receptor and /or muscarinic receptors are involved.
  • the present invention also concerns the compounds of formula (1 ), or pharmaceutically acceptable salts thereof and/or pharmaceutically acceptable solvates of said compounds or salts, for use in the treatment of diseases, disorders, and conditions selected from the group consisting of: • asthma of whatever type, etiology, or pathogenesis, in particular asthma that is a member selected from the group consisting of atopic asthma, non-atopic asthma, allergic asthma, atopic bronchial IgE-mediated asthma, bronchial asthma, essential asthma, true asthma, intrinsic asthma caused by pathophysiologic disturbances, extrinsic asthma caused by environmental factors, essential asthma of unknown or inapparent cause, non-atopic asthma, bronchitic asthma, emphysematous asthma, exercise-induced asthma, allergen induced asthma, cold air induced asthma, occupational asthma, infective asthma caused by bacterial, fungal, protozoal, or viral infection, non-allergic asthma, incipient asthma, whez infant syndrome and bronchiolytis;
  • obstructive or inflammatory airways diseases of whatever type, etiology, or pathogenesis in particular an obstructive or inflammatory airways disease that is a member selected from the group consisting of chronic eosinophilic pneumonia, chronic obstructive pulmonary disease (COPD), COPD that includes chronic bronchitis, pulmonary emphysema or dyspnea associated or not associated with COPD, COPD that is characterized by irreversible, progressive airways obstruction, adult respiratory distress syndrome (ARDS), exacerbation of airways hyper-reactivity consequent to other drug therapy and airways disease that is associated with pulmonary hypertension;
  • COPD chronic osinophilic pneumonia
  • COPD chronic obstructive pulmonary disease
  • COPD that includes chronic bronchitis, pulmonary emphysema or dyspnea associated or not associated with COPD
  • COPD that is characterized by irreversible, progressive airways obstruction, adult respiratory distress syndrome (ARDS), exacerb
  • bronchitis of whatever type, etiology, or pathogenesis in particular bronchitis that is a member selected from the group consisting of acute bronchitis, acute laryngotracheal bronchitis, arachidic bronchitis, catarrhal bronchitis, croupus bronchitis, dry bronchitis, infectious asthmatic bronchitis, productive bronchitis, staphylococcus or streptococcal bronchitis and vesicular bronchitis;
  • bronchiectasis of whatever type, etiology, or pathogenesis, in particular bronchiectasis that is a member selected from the group consisting of cylindric bronchiectasis, sacculated bronchiectasis, fusiform bronchiectasis, capillary bronchiectasis, cystic bronchiectasis, dry bronchiectasis and follicular bronchiectasis.
  • a still further aspect of the present invention also relates to the use of the compounds of formula (1 ), or pharmaceutically acceptable salts thereof and/or pharmaceutically acceptable solvates of said compounds or salts, for the manufacture of a drug having a ⁇ 2 agonist activity and an M3 antagonist activity.
  • the present inventions concerns the use of the compounds of formula (1 ), or pharmaceutically acceptable salts thereof and/or pharmaceutically acceptable solvates of said compounds or salts, for the manufacture of a drug for the treatment of diseases and/or conditions involving the beta 2 and muscarinic receptors, in particular the diseases and/or conditions listed above.
  • the present invention provides a particularly interesting method to treat a mammal, including a human being, with an effective amount of a compound of formula (1 ), or pharmaceutically acceptable salts thereof and/or pharmaceutically acceptable solvates of said compounds or salts. More precisely, the present invention provides a particularly interesting method for the treatment of a ⁇ 2- mediated diseases and/or conditions involving the beta 2 and muscarinic receptors, in a mammal, including a human being, in particular the diseases and/or conditions listed above, comprising administering said mammal with an effective amount of a compound of formula (1 ), its pharmaceutically acceptable salts and/or solvates of said compounds or salts.
  • the powder X-ray diffraction pattern was determined using a Bruker-AXS Ltd. D4 powder X-ray diffractometer fitted with an automatic sample changer, a theta-theta goniometer, automatic beam divergence slit, and a PSD Vantec-1 detector.
  • the sample was prepared for analysis by mounting on a low background cavity silicon wafer specimen mount. The peaks obtained were aligned against a silicon reference standard.
  • the analyses were performed with the goniometer running in continuous mode set for a 0.2 second count per 0.018° step over a two theta range of 2° to 55°
  • the title compound was prepared according to the following procedure: Tert-butyl 4-( ⁇ 3-[(R)-cyclohexyl(hydroxy)phenylmethyl]-1H-1 ,2,4-triazol-1-yl ⁇ methyl)piperidine-1- carboxylate (Preparation 3a, 2.0Og, 4.40mmol) was dissolved in dioxane (11 ml) and stirred vigorously to achieve solubilisation. 4M HCI in dioxane (5.54mL, 22.1 mmol) was then added. After stirring at room temperature for 24 hours the solvent was removed in vacuo, and the residue partitioned between dichloromethane (5OmL) and saturated aqueous sodium bicarbonate solution (5OmL).
  • the title compound may be prepared according to the following procedure:
  • the title compound may be prepared according to the following procedure: Cyclohexyl-phenyl-(1-piperidin-4-ylmethyl-1H-[1 ,2,4]triazol-3-yl)-methanol (Preparation 4, 0.4Og, 1.13mmol) and 2- ⁇ 2-[4-(2-Bromo-ethyl)-phenyl]-ethyl ⁇ -isoindole-1 ,3-dione (Preparation 11 , 0.404g, 1.13mmol) and diisopropylethylamine (0.59ml, 3.38mmol) were dissolved in methyl ethyl ketone (8mL).
  • the title compound was also prepared according to the following procedure: Methanesulfonic acid 2- ⁇ 4-[2-(1 ,3-dioxo-1 ,3-dihydro-isoindol-2-yl)-ethyl]-phenyl ⁇ -ethyl ester (Preparation 35, 20.0Og, 53.56mmol) was dissolved in acetonitrile (8OmL) and stirred at room temperature. Sodium iodide (16.06g, 107.12mmol) was added portionwise and the resulting slurry heated to 8O 0 C for 18h. The reaction mixture was cooled to 5O 0 C and further acetonitrile (8OmL) was added. To this mixture was added cyclohexyl-phenyl-(1-piperidin-4-ylmethyl-1H-[1 ,2,4]triazol-3-yl)-methanol (Preparation 4, 15.87g,
  • Triazole (1 Og, 144.8mnnol) and pyrrolidine (11.3g, 159.0mnnol) were dissolved ethanol (60ml) and formaldehyde (37% aqueous solution, 12.9ml, 159.0mmol) was added. After stirring at reflux for 4 hours reaction was allowed to stand at room temperature for 18 hours. The solvent was removed in vacuo, and the residue partitioned between dichloromethane (150ml) and water (8OmL) . The aqueous phase was separated and extracted with additional dichloromethane (80ml). The combined organic layers were washed with brine (100ml), dried over magnesium sulphate, filtered and the solvent removed in vacuo to furnish the title compound as a yellow oil, in 55% yield, 12.2g.
  • the title compound was prepared from tert-butyl 4-( ⁇ 3-[cyclohexyl(hydroxy)phenylmethyl]-1H-1 ,2,4- triazol-1-yl ⁇ methyl)piperidine-1-carboxylate (Preparation 19, 450mg, 0.990mmol) using the same method as described in preparation 4 to give a clear oil, in 91 % yield, 320mg.
  • Benzyl 4-(bromomethyl)piperidine-1-carboxylate (5g, 16mnnol) was dissolved in dimethylformannide (40ml) and diphenyl-(1 H-[1 ,2,4]triazol-3-yl)-methanol (Tetrahedron: Asymmetry, 8(9), 1491-1500; 1997; 3.35g, 13.65mmol) added, followed by potassium carbonate (3.69g, 26.7mmol). After stirring at 7O 0 C for 18 hours the solvent was removed in vacuo, and the residue partitioned between ethyl acetate (150ml) and water (15OmL).
  • the crystalline form produced by the process described above also has the characteristics shown in the corresponding Powder X-ray diffraction pattern of Figure 1/1.
  • the main characteristic peaks are at 12.7, 18.1 , 21.0, 22.2 and 23.6 degrees 2-theta ⁇ 0.1 degrees 2-theta and are further given in table 1 below.
  • Cell Pellets from CHO (Chinese Hamster Ovary) cells recombinantly expressing the human muscarinic M 3 receptor were homogenised in 2OmM HEPES (pH7.4) and centrifuged at 48000 x g for 20min at 4°C. The pellet was re-suspended in buffer and the homogenisation and centrifugation steps repeated. The resulting pellet was re-suspended in 1 ml buffer per 1 ml original packed cell volume and the homogenisation step repeated. Protein estimation was carried out on the suspension and 1 ml aliquots of ⁇ 1 mg/ml frozen at -8O 0 C.
  • Membranes (5 ⁇ g/well) were incubated with 3 H-NMS (at a concentration 5 x K D ) plus/minus test compound for 24hr at RT (room temperature) in a 1 ml polystyrene 96-well deep well block.
  • the final assay volume was 200 ⁇ l, comprising of: 20 ⁇ l plus/minus test compound; 20 ⁇ l 3 H-NMS (Perkin Elmer
  • Binding was defined with 1 ⁇ M Atropine. Assay buffer was 2OmM Hepes (pH 7.4).
  • IC 50 is the concentration of unlabelled drug which inhibits by 50% the specific radioligand binding.
  • [L] is the free radioligand concentrations and K D and K, are the equilibrium dissociation constants of the radioligand and unlabelled drug respectively.
  • CHO Choinese Hamster Ovary cells recombinantly expressing the human adrenergic ⁇ 2 receptor were maintained in growth media composed of F12:DMEM (Gibco 21331-020) containing 10% Foetal Bovine Serum (FBS: Sigma F4135), 10 ⁇ g/ml puromycin (Sigma P8833), 0.1 mg/ml Geneticin G418 (Sigma G7034) and 2mM L-glutamine (Sigma G7513). The cells were kept in sterile conditions at 37°C, in an atmosphere containing 5% CO 2 .
  • Cells were harvested for assay when they reached 80-90% confluency using trypsin 0.25% (Sigma T4049) incubated with the cells for 5 min at 37°C in an atmosphere containing 5% CO 2 . Detached cells were collected in warmed growth media (composition described above), and re-suspended in assay media (F12:DMEM (Gibco 21331-020) containing 1 % Foetal Bovine Serum (FBS: Sigma F4135) and 2mM L-glutamine (Sigma G7513)) to give a viable cell concentration of 0.25x10 6 cells/ml.
  • F12:DMEM Gibco 21331-020
  • FBS Foetal Bovine Serum
  • 2mM L-glutamine Sigma G7513
  • the PBS plate wash procedure was repeated, followed by the addition of 50 ⁇ l PBS plus 0.5mM IBMX and 50 ⁇ l PBS plus 2% DMSO and the plates returned to the incubator for 30mins.
  • Concentration ranges of test compounds were prepared in PBS containing 2% DMSO.
  • the PBS plate wash procedure was repeated, and then 50 ⁇ l PBS plus 0.5mM IBMX was added to the assay plates, followed by the addition of 50 ⁇ l of each compound test concentration to the appropriate well.
  • 50 ⁇ l per well of 2% DMSO or 20OnM Formoterol was added to the control wells, giving a final assay concentration of 1 % DMSO or 10OnM Formoterol. Plates were returned to the incubator for a further 30mins.

Landscapes

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

Abstract

The invention relates to compounds of formula (1) and to processes for the preparation of, intermediates used in the preparation of, compositions containing and the uses of, such derivatives. The compounds according to the present invention are useful in numerous diseases, disorders and conditions, in particular inflammatory, allergic and respiratory diseases, disorders and conditions.

Description

TRIAZOLE DERIVATIVES USEFUL FOR THE TREATMENT OF DISEASES
This invention relates to compounds of general formula (1 ):
Figure imgf000002_0001
in which A, X, Y, n, R1 and R2 have the meanings indicated below, and to processes and intermediates for the preparation of, compositions containing and the uses of such derivatives.
β2 adrenergic agonists and cholinergic muscarinic antagonists are well-established therapeutic agents for the treatment of obstructive respiratory diseases such as COPD and Asthma. Currently used inhaled β2 agonists include both short acting agents such as salbutamol (q.i.d.), and terbutaline (t.i.d) and longer acting agents such as salmeterol and formoterol (b.i.d.) and produce bronchodilation via stimulation of adrenergic receptors on airway smooth muscle. Inhaled muscarinic antagonists in clinical use include the short acting ipratropium bromide (q.i.d.), oxitropium bromide (q.i.d) and the long acting tiotropium (q.d.). Muscarinic antagonists produce bronchodilation by inhibiting the cholinergic tone of airways primarily by antagonising the action of acetylcholine on muscarinic receptors present on airway smooth muscle. A number of published studies have demonstrated that the combined administration of inhaled β2 agonists with inhaled muscarinic antagonists (whether short or long acting) to patients with obstructive lung disease results in superior improvements in lung function, symptoms and quality of life measures compared to patients receiving either single class of agent alone. Studies to date have been restricted to combination studies with single pharmacology agents, however combination of both pharmacologies within a single molecule would be desirable as this could yield increased bronchodilator efficacy with similar therapeutic index to the single agents or similar efficacy with superior therapeutic index. In addition, combining both pharmacologies in a single molecule would allow the potential for combination with anti-inflammatory agents thus giving a triple therapy from a single inhaler.
Accordingly, there is a need for alternative compounds active as beta 2 receptor agonists and muscarinic antagonists, being suitable for the treatment of respiratory diseases, preferably by the inhalation route. Such compounds would have an appropriate pharmacological profile, for example in term of potency, pharmacokinetics or duration of action. In addition, as these compounds may be used for the treatment of chronic diseases, such as asthma or COPD, they would preferably have a low potential for interaction with co-administered compounds.
The invention relates to the compounds of general formula (1 ):
Figure imgf000002_0002
wherein: A is selected from:
Figure imgf000003_0001
wherein * represent the attachment point of A to the carbon bearing the hydroxy;
X is -(CH2)m- where m is an integer comprised between 7 to 12 inclusive, or is of formula:
Figure imgf000003_0002
Y is selected from
Figure imgf000003_0003
wherein ** and *** represent the attachment points, ** being linked to X; n is 0 or 1 ;
R1 is selected from cyclopentyl, cyclohexyl, phenyl, furanyl and thiophenyl; and,
R2 is selected from phenyl, furanyl and thiophenyl; or the p harmaceutically acceptable salts thereof, or the pharmaceutically acceptable solvates of said compounds or salts.
The compounds of formula (1) are β2 adrenergic receptor agonists and muscarinic receptor antagonists that are particularly useful for the treatment of diseases and/or conditions involving said receptors, by showing excellent potency, in particular when administered via the inhalation route.
Compounds of formula (1 ) may be prepared in a variety of ways. The routes below illustrate one such way of preparing these compounds, in which A, X, Y, n, R1 and R2 are as previously defined for the compounds of the formula (1 ) unless otherwise stated. The skilled person will appreciate that other routes may be equally as practicable. (1 )
The amine derivative of the formula (1 ):
Figure imgf000003_0004
may be prepared by reaction of an amine of formula (2):
Figure imgf000003_0005
with a bromide of formula (3):
Figure imgf000004_0001
where A1 represents A protected with a suitable phenol protecting group such as benzyl.
In a typical procedure, the amine of formula (2) is reacted with a bromide of formula (3) optionally in the presence of a solvent or mixture of solvents (e.g. dimethyl sulphoxide, toluene, Λ/,Λ/-dimethylformamide, propionitrile, acetonitrile, butyronitrile, dichloromethane), optionally in the presence of a suitable base (e.g. triethylamine, diisopropylethylamine, potassium carbonate, sodium hydrogen carbonate), at a temperature comprised between 8O0C and 1200C, for 12 to 72 hours. The protecting groups such as tert- butyldimethylsilyl and benzyl can then be removed using standard methodology for cleaving oxygen protecting groups such as those found in the text book T. W. Greene, Protective Groups in Organic Synthesis (Wiley-lnterscience Publication, 1981 ).
Compounds of formula (3) where A1 is represented below may be prepared according to the methods disclosed in the following references:
Figure imgf000004_0004
The amine of formula (2) may be prepared from the corresponding protected amine of formula (4):
Figure imgf000004_0002
where Ra and R represent suitable protecting groups such as tert-butoxycarbonyl or together represent phthalimide.
In a typical procedure, the amine of formula (4) is deprotected using standard methodology for cleaving nitrogen protecting groups such as those found in the text book T. W. Greene, Protective Groups in Organic Synthesis (Wiley-lnterscience Publication, 1981 ).
The amine of formula (4) may be prepared by reacting the corresponding amine of formula (5):
Figure imgf000004_0003
where p and q are independently selected from 1 or 2, with a compound of formula (6):
Figure imgf000005_0001
wherein Ra and Rb are as previously defined and LG is a suitable leaving group such as bromide or mesylate
In a typical procedure, the amine of formula (5) is reacted with a compound of formula (6) in a suitable solvent (e.g. dimethyl sulphoxide, toluene, Λ/,Λ/-dimethylformamide, propionitrile, acetonitrile, methyl ethyl ketone), in the presence of a suitable base (e.g. triethylamine, diisopropylethylamine) and optionally in the presence of a nucleophilic additive (e.g. sodium iodide) at a temperature comprised between 5O0C and 900C, for 18 to 48 hours.
In addition to their utility for the preparation of the compounds of formula (1 ), the compounds of formula (5) may also be used as muscarinic antagonists. In particular they may be useful for the treatment of diseases which can be alleviated by the modulation of the activity of the muscarinic receptor.
The compounds of formula (6) where LG is bromide may be prepared by reacting the corresponding amine nucleophiles RaRbNH with the corresponding dibromide of formula (7):
Br-X-Br (7)
In a typical procedure, phthalimide or di-tert-butyl iminodicarboxylate (RaRbNH) is reacted with a suitable base such as sodium hydride (60% dispersion in oil), in a suitable solvent such as N, N- dimethylformamide or tetrahydrofuran, at a temperature comprised between 00C and room temperature for up to 1 hour, followed by addition of the dibromide of formula (7), at a temperature comprised between 00C and 1500C, for 6-48 hours.
The amines of formula RaRbNH are commercially available.
Compounds of formula (7) where X is -(CH2)m- are commercially available.
Compounds of formula (7) where X is of formula:
Figure imgf000005_0002
may be prepared as described in Journal of Organic Chemistry, 46(22), 4608-10; 1981.
Alternatively, compounds of formula (6) where Ra and Rb toge ther represent phthalimide and X is of formula:
- (CH2J2
Figure imgf000005_0003
may be prepared as described in scheme 1 below:
Figure imgf000006_0001
Scheme 1
wherein Ra and Rb together represent phthalimide.
Compounds of formula (8) and (9) are commercially available.
Compound of formula (10) may be prepared from compounds of formula (8) and (9) by Heck reaction (process step (i)). Typical conditions comprise reaction of compound (8) with compound (9), tri-o- tolylphosphine, palladium(ll)acetate and a suitable base such as diisopropylethylamine, in a suitable solvent such as acetonitrile, at 900C for 21 hours.
Compound of formula (11 ) may be prepared from compound of formula (10) by hydrogenation (process step (ii)). Typical conditions comprise reaction of compound (10) with ammonium formate and 30% palladium hydroxide on carbon, in suitable solvents such as ethyl acetate and ethanol, at 8O0C for 18 hours. Alternative conditions comprise reaction of compound (10) with hydrogen gas and rhodium tris(triphenylphosphine)chloride, in suitable solvents such as ethyl acetate and ethanol, at 20psi and room temperature for 24 hours.
Compound of formula (6a) where LG is bromide may be prepared from compound of formula (11 ) by bromination (process step (iii)). Typical conditions comprise reaction of compound (11 ) with phosphorous tribromide, in a suitable solvent such as toluene, at reflux for 4 hours.
Compound of formula (6a) where LG is mesylate may be prepared from compound of formula (1 1 ) by mesylation (process step (iii)). Typical conditions comprise reaction of compound (1 1 ) with methanesulfonyl chloride, in a suitable solvent such as methylethyl ketone, with a suitable base such as triethylamine, at O0C to room temperature for 1 -4 hours.
The amines of formula (5) may be prepared from the corresponding protected amines of formula (12):
Figure imgf000007_0001
where Rc represents a suitable protecting group such as tert-butoxycarbonyl or benzyloxycarbonyl.
In a typical procedure, the amine of formula (12) is deprotected using standard methodology for cleaving nitrogen protecting groups such as those found in the text book T. W. Greene, Protective Groups in Organic Synthesis (Wiley-lnterscience Publication, 1981 ).
The amines of formula (12) may be prepared by reacting the corresponding compounds of formula (13):
Figure imgf000007_0002
wherein LG is a suitable leaving group such as bromide or mesylate, with compounds of formula (14):
Figure imgf000007_0003
In a typical procedure, compounds of formula (13) are reacted with compounds of formula (14) in a suitable solvent (e.g. Λ/,Λ/-dimethylformamide, propionitrile, acetonitrile, acetone), in the presence of a suitable base (e.g. potassium carbonate, caesium carbonate, triethylamine, diisopropylethylamine) at a temperature between 7O0C and 15O0C, for 5 to 48 hours.
Compounds of formula (13) are commercially available or can be prepared according to methods known to the skilled person.
In particular, the compounds of formula (13) where Rc is tert-butoxycarbonyl, p and q represent 1 or 2, n is 0 and LG is mesylate are commercially available. The compounds of formula (13) where Rc is tert- butoxycarbonyl, p and q are 1 , n is 1 and LG is mesylate may be prepared as described in U.S. Pat. Appl. Publ., 2005101586. The compounds of formula (13) where Rc is tert-butoxycarbonyl, p is 1 , q is 2, n is 1 and LG is bromide or mesylate are commercially available. Finally, the compounds of formula (13) where Rc is tert-butoxycarbonyl or benzyloxycarbonyl, p and q are 2, n is 1 and LG is bromide are commercially available
Compounds of formula (14) may be prepared from the corresponding amides of formula (15):
Figure imgf000008_0001
In a typical procedure, compounds of formula (15) are reacted with N,N-dimethylformamide dimethylacetal at 9O0C for 2 hours. Excess N,N-dimethylformamide dimethylacetal is removed in vacuo and the residue azeotroped with a suitable solvent such as toluene. Hydrazine hydrate is added, followed by glacial acetic acid, and heated at 9O0C for 2 hours.
Compounds of formula (15) may be prepared from the corresponding acids of formula (16):
Figure imgf000008_0002
In a typical procedure, compounds of formula (16) are reacted with carbonyldiimidazole in a suitable solvent such as dichloromethane or tetrahydrofuran at room temperature for 1 hour, followed by addition of aqueous 0.880 ammonia solution at room temperature for 18 hours.
Compounds of formula (16) and their corresponding single enantiomers, as appropriate, are either commercially available, known in the literature, may be prepared as described in the literature, or by analogous methods to those in the literature. Examples of relevant literature with such information or reference to such information include, but are not limited to: WO2002/053564, WO2006/048225, WO2001/04118, WO2003/057694, WO2004/052857, WO96/33973, Journal of Organic Chemistry, 46(14), 2885-9; 1981 , Journal of Organic Chemistry (1961 ), 26, 1573-7, Chimica Therapeutica (1966), (4), 238-45 and Journal of Medicinal Chemistry, 44(20), 3244-3253; 2001 .
For example, the compound of formula (16a):
Figure imgf000008_0003
is commercially available as (2R)-2-cyclohexyl-2-hydroxy-2-phenylacetic acid.
Alternatively, compounds of formula (14) may be prepared from the corresponding compounds of formula (17):
Figure imgf000008_0004
In a typical procedure, compounds of formula (17) are reacted with sodium borohydride in a suitable solvent such as ethanol, at reflux, for 3 hours.
The compounds of formula (17) may be prepared by reacting the compound of formula (18):
with the compounds of formula (19):
Figure imgf000009_0001
In a typical procedure, the compound of formula (18) is reacted with a suitable base such as n-butyl lithium, in a suitable solvent such as tetrahydrofuran, at a temperature between -780C and room temperature for 1 hour, followed by addition of a compound of formula (19) in tetrahydrofuran at a temperature between -780C and room temperature for 18 hours.
The compounds of formula (18) may be prepared as described in Tetrahedron: Asymmetry, 8(9), 1491- 1500; 1997.
Compounds of formula (19) are either commercially available, may be prepared as described in the literature, or by analogous methods to those described in the literature. Examples of relevant literature with such information include, but are not limited to:
Tetrahedron Letters, 49(11 ), 1884-1888; 2008; Tetrahedron Letters, 46(44), 7627-7630; 2005 Synthesis, (13), 1970-1978; 2007; Journal of Organic Chemistry, 55(4), 1286-91 ; 1990;U.S. 5969159; Tetrahedron Letters, 47(10), 1649-1651 ; 2006; Journal of Chemical Research, Synopses, (9), 280-1 ; 1984; Synthesis, (3), 242-3; 1991 ; Journal of the Chemical Society, Perkin Transactions 2: Physical Organic Chemistry (1972-1999), (11 ), 1741-51 ; 1989; and Synthetic Communications, 34(23), 4249- 4256; 2004.
The preparation of compounds of formula (1 ) may require the protection of potential reactive functionality in addition to those methods already described. In such a case, examples of compatible protecting groups and their particular methods of protection and deprotection are described in "Protecting Groups in Organic Synthesis" by T.W. Greene and P. Wutz (Wiley-lnterscience Publication, 1981 ) or "Protecting groups" by P. J. Kocienski (Georg Thieme Verlag, 1994).
Compounds of formula (1 ) as well as intermediates for their preparation can be purified and isolated according to various well-known methods, for example crystallisation or chromatography.
Subgroups of compounds of formula (1 ) containing the following substituents, or combinations of the following substituents, are preferred: Preferably, A is of formula:
Figure imgf000010_0001
wherein * represent the attachment point of A to the carbon bearing the hydroxyl.
Preferably, X is (CH2)9 or is of formula:
Figure imgf000010_0002
In a preferred embodiment, X is (CH2^.
In another preferred embodiment, X is of formula:
Figure imgf000010_0003
Preferably, Y is of formula:
Figure imgf000010_0004
wherein ** and *** represent the attachment points, ** being linked to X.
Preferably, R1 is phenyl or cyclohexyl.
Preferably, R2 is phenyl.
Preferably, n is 1.
When R1 and R2 are different, the carbon atom bearing these two substituents can be in the (R) or (S) configuration. Preferably, the carbon atom bearing R1 and R2 is in the (R) configuration when R1 and R2 are different.
The compounds of formula (1 ) wherein: A is of formula:
Figure imgf000011_0001
wherein * represent the attachment point of A to the carbon bearing the hydroxy; X is-(CH2)9- or is of formula:
Figure imgf000011_0002
Y is of formula:
Figure imgf000011_0003
wherein ** and *** represent the attachment points, ** being linked to X; n is 1;
R1 is cyclohexyl or phenyl and R2 is phenyl; or the pharmaceutically acceptable salts thereof, or the pharmaceutically acceptable solvates of said compounds or salts, are further preferred.
The following compounds are more preferred:
5-[(1R)-2-({9-[4-({3-[(R)-cyclohexyl(hydroxy)phenylmethyl]-1H-1 ,2,4-triazol-1-yl}methyl)piperidin-1- yl]nonyl}amino)-1-hydroxyethyl]-8-hydroxyquinolin-2(1H)-one,
5-[(1R)-2-({9-[4-({3-[(R)-cyclohexyl(hydroxy)phenylmethyl]-1H-1 ,2,4-triazol-1-yl}methyl)piperidin-1- yl]nonyl}amino)-1-hydroxyethyl]-8-hydroxyquinolin-2(1 H)-one naphthalene-1 ,5-disulfonate salt,
5-[(1R)-2-{[2-(4-{2-[4-({3-[(R)-cyclohexyl(hydroxy)phenylmethyl]-1H-1 ,2,4-triazol-1-yl}methyl)piperidin-1- yl]ethyl}phenyl)ethyl]amino}-1-hydroxyethyl]-8-hydroxyquinolin-2(1H)-one,
5-[(1R)-2-{[2-(4-{2-[4-({3-[(R)-cyclohexyl(hydroxy)phenylmethyl]-1H-1 ,2,4-triazol-1-yl}methyl)piperidin-1- yl]ethyl}phenyl)ethyl]amino}-1-hydroxyethyl]-8-hydroxyquinolin-2(1H)-one naphthalene-1 ,5-disulfonate salt,
5-[(1R)-2-{[2-(4-{2-[4-({3-[(R)-cyclohexyl(hydroxy)phenylmethyl]-1H-1 ,2,4-triazol-1-yl}methyl)piperidin-1- yl]ethyl}phenyl)ethyl]amino}-1-hydroxyethyl]-8-hydroxyquinolin-2(1H)-one succinate salt,
5-[(1R)-2-{[2-(4-{2-[4-({3-[(R)-cyclohexyl(hydroxy)phenylmethyl]-1H-1 ,2,4-triazol-1-yl}methyl)piperidin-1- yl]ethyl}phenyl)ethyl]amino}-1-hydroxyethyl]-8-hydroxyquinolin-2(1H)-one fumarate salt,
5-[(1R)-2-({9-[4-({3-[cyclohexyl(hydroxy)phenylmethyl]-1H-1 ,2,4-triazol-1-yl}methyl)piperidin-1- yl]nonyl}amino)-1-hydroxyethyl]-8-hydroxyquinolin-2(1H)-one,
8-hydroxy-5-[(1R)-1-hydroxy-2-({9-[4-({3-[hydroxy(diphenyl)methyl]-1H-1 ,2,4-triazol-1-yl}methyl)piperidin-
1-yl]nonyl}amino)ethyl]quinolin-2(1H)-one, and
8-hydroxy-5-[(1R)-1-hydroxy-2-{[2-(4-{2-[4-({3-[hydroxy(diphenyl)methyl]-1H-1 ,2,4-triazol-1- yl}methyl)piperidin-1-yl]ethyl}phenyl)ethyl]amino}ethyl]quinolin-2(1H)-one. 5-[(1 R)-2-{[2-(4-{2-[4-({3-[(R)-cyclohexyl(hydroxy)phenylmethyl]-1H-1 ,2,4-triazol-1-yl}methyl)piperidin-1- yl]ethyl}phenyl)ethyl]amino}-1-hydroxyethyl]-8-hydroxyquinolin-2(1 H)-one is even more preferred.
5-[(1 R)-2-{[2-(4-{2-[4-({3-[(R)-cyclohexyl(hydroxy)phenylmethyl]-1H-1 ,2,4-triazol-1-yl}methyl)piperidin-1- yl]ethyl}phenyl)ethyl]amino}-1-hydroxyethyl]-8-hydroxyquinolin-2(1 H)-one naphthalene-1 ,5-disulfonate salt is most preferred.
Pharmaceutically acceptable salts of the compounds of formula (1 ) include the acid addition and base salts thereof. Suitable acid addition salts are formed from acids which form non-toxic salts. Examples include the acetate, aspartate, benzoate, besylate, bicarbonate/carbonate, bisulphate/sulphate, borate, camsylate, citrate, edisylate, esylate, formate, fumarate, gluceptate, gluconate, glucuronate, hexafluorophosphate, hibenzate, hydrochloride/chloride, hydrobromide/bromide, hydroiodide/iodide, isethionate, lactate, malate, maleate, malonate, mesylate, methylsulphate, naphthylate, 1 ,5- naphthalenedisulfonate, 2-napsylate, nicotinate, nitrate, orotate, oxalate, palmitate, pamoate, phosphate/hydrogen phosphate/dihydrogen phosphate, saccharate, stearate, succinate, tartrate, tosylate and trifluoroacetate salts. Suitable base salts are formed from bases which form non-toxic salts. Examples include the aluminium, arginine, benzathine, calcium, choline, diethylamine, diolamine, glycine, lysine, magnesium, meglumine, olamine, potassium, sodium, tromethamine and zinc salts. Hemisalts of acids and bases may also be formed, for example, hemisulphate and hemicalcium salts. For a review on suitable salts, see "Handbook of Pharmaceutical Salts: Properties, Selection, and Use" by Stahl and Wermuth (Wiley-VCH, Weinheim, Germany, 2002).
Pharmaceutically acceptable salts of compounds of formula (1 ) may be prepared by one or more of three methods:
(i) by reacting the compound of formula (1 ) with the desired acid or base;
(ii) by removing an acid- or base-labile protecting group from a suitable precursor of the compound of formula (1 ) or by ring-opening a suitable cyclic precursor, for example, a lactone or lactam, using the desired acid or base; or (iii) by converting one salt of the compound of formula (1 ) to another by reaction with an appropriate acid or base or by means of a suitable ion exchange column.
All three reactions are typically carried out in solution. The resulting salt may precipitate out and be collected by filtration or may be recovered by evaporation of the solvent. The degree of ionisation in the resulting salt may vary from completely ionised to almost non-ionised.
The compounds of the invention may exist in both unsolvated and solvated forms. The term 'solvate' is used herein to describe a molecular complex comprising the compound of the invention or a salt thereof and a stoichiometric amount of one or more pharmaceutically acceptable solvent molecules, for example, ethanol. The term 'hydrate' is employed when said solvent is water.
Included within the scope of the invention are complexes such as clathrates, drug-host inclusion complexes wherein, in contrast to the aforementioned solvates, the drug and host are present in stoichiometric or non-stoichiometric amounts. Also included are complexes of the drug containing two or more organic and/or inorganic components which may be in stoichiometric or non-stoichiometric amounts. The resulting complexes may be ionised, partially ionised, or non-ionised. For a review of such complexes, see J Pharm Sci, 64 (8), 1269-1288 by Haleblian (August 1975).
Hereinafter all references to compounds of formula (1 ) include references to salts, solvates and complexes thereof and to solvates and complexes of salts thereof.
The compounds of the invention include compounds of formula (1 ) as hereinbefore defined, including all polymorphs and crystal habits thereof, prodrugs and isomers thereof (including optical, geometric and tautomeric isomers) as hereinafter defined and isotopically-labeled compounds of formula (1 ).
As indicated, so-called 'pro-drugs' of the compounds of formula (1 ) are also within the scope of the invention. Thus certain derivatives of compounds of formula (1 ) which may have little or no pharmacological activity themselves can, when administered into or onto the body, be converted into compounds of formula (1 ) having the desired activity, for example, by hydrolytic cleavage. Such derivatives are referred to as 'prodrugs'. Further information on the use of prodrugs may be found in 'Pro-drugs as Novel Delivery Systems, Vol. 14, ACS Symposium Series (T. Higuchi and W. Stella) and 'Bioreversible Carriers in Drug Design', Pergamon Press, 1987 (ed. E. B Roche, American Pharmaceutical Association).
Prodrugs in accordance with the invention can, for example, be produced by replacing appropriate functionalities present in the compounds of formula (1 ) with certain moieties known to those skilled in the art as 'pro-moieties' as described, for example, in "Design of Prodrugs" by H. Bundgaard (Elsevier, 1985).
Some examples of prodrugs in accordance with the invention include:
(i) where the compound of formula (1 ) contains an alcohol functionality (-OH), an ether thereof, for example, a compound wherein the hydrogen of the alcohol functionality of the compound of formula (1 ) is replaced by (C-ι-C6)alkanoyloxymethyl; and
(ii) where the compound of formula (1 ) contains a primary or secondary amino functionality (-NH2 or -NHR where R ≠ H), an amide thereof, for example, a compound wherein, as the case may be, one or both hydrogens of the amino functionality of the compound of formula (1 ) is/are replaced by (C-ι-Cio)alkanoyl.
Further examples of replacement groups in accordance with the foregoing examples and examples of other prodrug types may be found in the aforementioned references.
Moreover, certain compounds of formula (1 ) may themselves act as prodrugs of other compounds of formula
(1 )-
Also included within the scope of the invention are metabolites of compounds of formula (1 ), that is, compounds formed in vivo upon administration of the drug. Some examples of metabolites in accordance with the invention include (i) where the compound of formula (1 ) contains a methyl group, an hydroxymethyl derivative thereof (-CH3 → -CH2OH): (ii) where the compound of formula (1 ) contains an alkoxy group, an hydroxy derivative thereof (-
OR → -OH); (iii) where the compound of formula (1 ) contains a tertiary amino group, a secondary amino derivative thereof (-NR1R2 → -NHR1 or -NHR2); (iv) where the compound of formula (1 ) contains a secondary amino group, a primary derivative thereof (-NHR1 → -NH2); (v) where the compound of formula (1 ) contains a phenyl moiety, a phenol derivative thereof (-Ph — >
-PhOH); and (vi) where the compound of formula (1 ) contains an amide group, a carboxylic acid derivative thereof
(-CONH2 → COOH).
The compounds of formula (1 ) wherein R1 and R2 are different can exist as stereoisomers. Where structural isomers are interconvertible via a low energy barrier, tautomeric isomerism ('tautomerism') can occur. This can take the form of proton tautomerism in compounds of formula (1 ) containing, for example, an imino, keto, or oxime group, or so-called valence tautomerism in compounds which contain an aromatic moiety. It follows that a single compound may exhibit more than one type of isomerism. In particular, the present invention includes the compounds of formula (1 ) wherein the triazole moiety may be present as different regio-isomers.
Included within the scope of the present invention are all stereoisomers and tautomeric forms of the compounds of formula (1 ), including compounds exhibiting more than one type of isomerism, and mixtures of one or more thereof. Also included are acid addition or base salts wherein the counterion is optically active, for example, d-lactate or /-lysine, or racemic, for example, c//-tartrate or c//-arginine.
Conventional techniques for the preparation/isolation of individual enantiomers include chiral synthesis from a suitable optically pure precursor or resolution of the racemate (or the racemate of a salt or derivative) using, for example, chiral high pressure liquid chromatography (HPLC).
Alternatively, the racemate (or a racemic precursor) may be reacted with a suitable optically active compound, for example, an alcohol, or, in the case where the compound of formula (1 ) contains an acidic or basic moiety, an acid or base such as tartaric acid or 1-phenylethylamine. The resulting diastereomeric mixture may be separated by chromatography and/or fractional crystallization and one or both of the diastereoisomers converted to the corresponding pure enantiomer(s) by means well known to a skilled person.
Chiral compounds of the invention (and chiral precursors thereof) may be obtained in enantiomerically- enriched form using chromatography, typically HPLC, on an asymmetric resin with a mobile phase consisting of a hydrocarbon, typically heptane or hexane, containing from 0 to 50% by volume of isopropanol, typically from 2% to 20%, and from 0 to 5% by volume of an alkylamine, typically 0.1 % diethylamine. Concentration of the eluate affords the enriched mixture.
Stereoisomeric conglomerates may be separated by conventional techniques known to those skilled in the art - see, for example, "Stereochemistry of Organic Compounds" by E. L. Eliel (Wiley, New York, 1994).
The present invention includes all pharmaceutically acceptable isotopically-labelled compounds of formula (1 ) wherein one or more atoms are replaced by atoms having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number which predominates in nature.
Examples of isotopes suitable for inclusion in the compounds of the invention include isotopes of hydrogen, such as 2H and 3H, carbon, such as 11C, 13C and 14C, nitrogen, such as 13N and 15N, oxygen, such as 15O, 17O and 18O, and sulphur, such as 35S.
Certain isotopically-labelled compounds of formula (1 ), for example, those incorporating a radioactive isotope, are useful in drug and/or substrate tissue distribution studies. The radioactive isotopes tritium, i.e. 3H, and carbon-14, i.e. 14C, are particularly useful for this purpose in view of their ease of incorporation and ready means of detection.
Substitution with heavier isotopes such as deuterium, i.e. 2H, may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements, and hence may be preferred in some circumstances.
Substitution with positron emitting isotopes, such as 11C, 15O and 13N, can be useful in Positron Emission Topography (PET) studies for examining substrate receptor occupancy.
Isotopically-labeled compounds of formula (1 ) can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described in the accompanying Examples and Preparations using an appropriate isotopically-labeled reagents in place of the non- labeled reagent previously employed.
Pharmaceutically acceptable solvates in accordance with the invention include those wherein the solvent of crystallization may be isotopically substituted, e.g. D2O, d6-acetone, d6-DMSO.
The compounds of formula (1 ), the pharmaceutically acceptable salts thereof and/or the pharmaceutically acceptable solvates of said compounds or salts, are valuable pharmaceutically active compounds, which are suitable for the therapy and prophylaxis of numerous disorders in which the β2 receptor and/or the muscarinic receptor are involved . In particular, these compounds are useful for the therapy and prophylaxis of numerous disorders in which agonism of the β2 receptor and antagonism of the muscarinic receptor may induce benefit, in particular the allergic and non-allergic airways diseases.
Compounds of the invention intended for pharmaceutical use may be administered as crystalline or amorphous products. They may be obtained, for example, as solid plugs, powders, or films by methods such as precipitation, crystallization, freeze drying, spray drying, or evaporative drying. Microwave or radio frequency drying may be used for this purpose.
They may be administered alone or in combination with one or more other compounds of the invention or in combination with one or more other drugs (or as any combination thereof). Generally, they will be administered as a formulation in association with one or more pharmaceutically acceptable excipients. The term "excipient" is used herein to describe any ingredient other than the compound(s) of the invention. The choice of excipient will to a large extent depend on factors such as the particular mode of administration, the effect of the excipient on solubility and stability, and the nature of the dosage form.
Pharmaceutical compositions suitable for the delivery of compounds of the present invention and methods for their preparation will be readily apparent to those skilled in the art. Such compositions and methods for their preparation may be found, for example, in 'Remington's Pharmaceutical Sciences', 19th Edition (Mack Publishing Company, 1995).
The compounds of the invention may be administered orally. Oral administration may involve swallowing, so that the compound enters the gastrointestinal tract, or buccal or sublingual administration may be employed by which the compound enters the blood stream directly from the mouth.
Formulations suitable for oral administration include solid formulations such as tablets, capsules containing particulates, liquids, or powders, lozenges (including liquid-filled), chews, multi- and nano- particulates, gels, solid solution, liposome, films, ovules, sprays and liquid formulations.
Liquid formulations include suspensions, solutions, syrups and elixirs. Such formulations may be employed as fillers in soft or hard capsules and typically comprise a carrier, for example, water, ethanol, polyethylene glycol, propylene glycol, methylcellulose, or a suitable oil, and one or more emulsifying agents and/or suspending agents. Liquid formulations may also be prepared by the reconstitution of a solid, for example, from a sachet.
The compounds of the invention may also be used in fast-dissolving, fast-disintegrating dosage forms such as those described in Expert Opinion in Therapeutic Patents, 1 1 (6), 981-986, by Liang and Chen (2001 ).
For tablet dosage forms, depending on dose, the drug may make up from 1 weight % to 80 weight % of the dosage form, more typically from 5 weight % to 60 weight % of the dosage form. In addition to the drug, tablets generally contain a disintegrant. Examples of disinteg rants include sodium starch glycolate, sodium carboxymethyl cellulose, calcium carboxymethyl cellulose, croscarmellose sodium, crospovidone, polyvinylpyrrolidone, methyl cellulose, microcrystalline cellulose, lower alkyl -substituted hydroxypropyl cellulose, starch, pregelatinised starch and sodium alginate. Generally, the disintegrant will comprise from 1 weight % to 25 weight %, preferably from 5 weight % to 20 weight % of the dosage form.
Binders are generally used to impart cohesive qualities to a tablet formulation. Suitable binders include microcrystalline cellulose, gelatin, sugars, polyethylene glycol, natural and synthetic gums, polyvinylpyrrolidone, pregelatinised starch, hydroxypropyl cellulose and hydroxypropyl methylcellulose. Tablets may also contain diluents, such as lactose (monohydrate, spray-dried monohydrate, anhydrous and the like), mannitol, xylitol, dextrose, sucrose, sorbitol, microcrystalline cellulose, starch and dibasic calcium phosphate dihydrate.
Tablets may also optionally comprise surface active agents, such as sodium lauryl sulfate and polysorbate 80, and glidants such as silicon dioxide and talc. When present, surface active agents may comprise from 0.2 weight % to 5 weight % of the tablet, and glidants may comprise from 0.2 weight % to 1 weight % of the tablet.
Tablets also generally contain lubricants such as magnesium stearate, calcium stearate, zinc stearate, sodium stearyl fumarate, and mixtures of magnesium stearate with sodium lauryl sulphate. Lubricants generally comprise from 0.25 weight % to 10 weight %, preferably from 0.5 weight % to 3 weight % of the tablet.
Other possible ingredients include anti-oxidants, colourants, flavouring agents, preservatives and taste- masking agents.
Exemplary tablets contain up to about 80% drug, from about 10 weight % to about 90 weight % binder, from about 0 weight % to about 85 weight % diluent, from about 2 weight % to about 10 weight % disintegrant, and from about 0.25 weight % to about 10 weight % lubricant.
Tablet blends may be compressed directly or by roller to form tablets. Tablet blends or portions of blends may alternatively be wet-, dry-, or melt-granulated, melt congealed, or extruded before tabletting. The final formulation may comprise one or more layers and may be coated or uncoated; it may even be encapsulated.
The formulation of tablets is discussed in Pharmaceutical Dosage Forms: Tablets, Vol. 1 , by H. Lieberman and L. Lachman (Marcel Dekker, New York, 1980).
Consumable oral films for human or veterinary use are typically pliable water-soluble or water-swellable thin film dosage forms which may be rapidly dissolving or mucoadhesive and typically comprise a compound of formula (1 ), a film-forming polymer, a binder, a solvent, a humectant, a plasticiser, a stabiliser or emulsifier, a viscosity-modifying agent and a solvent. Some components of the formulation may perform more than one function. The compound of formula (1 ) may be water-soluble or insoluble. A water-soluble compound typically comprises from 1 weight % to 80 weight %, more typically from 20 weight % to 50 weight %, of the solutes. Less soluble compounds may comprise a greater proportion of the composition, typically up to 88 weight % of the solutes. Alternatively, the compound of formula (1 ) may be in the form of multiparticulate beads.
The film-forming polymer may be selected from natural polysaccharides, proteins, or synthetic hydrocolloids and is typically present in the range 0.01 to 99 weight %, more typically in the range 30 to 80 weight %.
Other possible ingredients include anti-oxidants, colorants, flavourings and flavour enhancers, preservatives, salivary stimulating agents, cooling agents, co-solvents (including oils), emollients, bulking agents, anti-foaming agents, surfactants and taste-masking agents.
Films in accordance with the invention are typically prepared by evaporative drying of thin aqueous films coated onto a peelable backing support or paper. This may be done in a drying oven or tunnel, typically a combined coater dryer, or by freeze-drying or vacuuming.
Solid formulations for oral administration may be formulated to be immediate and/or modified release. Modified release formulations include delayed-, sustained-, pulsed-, controlled-, targeted and programmed release.
Suitable modified release formulations for the purposes of the invention are described in US Patent No. 6,106,864. Details of other suitable release technologies such as high energy dispersions and osmotic and coated particles are to be found in Pharmaceutical Technology On-line, 25(2), 1-14, by Verma et al (2001 ). The use of chewing gum to achieve controlled release is described in WO 00/35298.
The compounds of the invention may also be administered directly into the blood stream, into muscle, or into an internal organ. Suitable means for parenteral administration include intravenous, intraarterial, intraperitoneal, intrathecal, intraventricular, intraurethral, intrasternal, intracranial, intramuscular and subcutaneous. Suitable devices for parenteral administration include needle (including microneedle) injectors, needle-free injectors and infusion techniques.
Parenteral formulations are typically aqueous solutions which may contain excipients such as salts, carbohydrates and buffering agents (preferably to a pH of from 3 to 9), but, for some applications, they may be more suitably formulated as a sterile non-aqueous solution or as a dried form to be used in conjunction with a suitable vehicle such as sterile, pyrogen-free water.
The preparation of parenteral formulations under sterile conditions, for example, by lyophilisation, may readily be accomplished using standard pharmaceutical techniques well known to those skilled in the art. The solubility of compounds of formula (1 ) used in the preparation of parenteral solutions may be increased by the use of appropriate formulation techniques, such as the incorporation of solubility- enhancing agents.
Formulations for parenteral administration may be formulated to be immediate and/or modified release. Modified release formulations include delayed-, sustained-, pulsed-, controlled-, targeted and programmed release. Thus compounds of the invention may be formulated as a solid, semi-solid, or thixotropic liquid for administration as an implanted depot providing modified release of the active compound. Examples of such formulations include drug-coated stents and poly(c//-lactic-coglycolic)acid (PGLA) microspheres.
The compounds of the invention may also be administered topically to the skin or mucosa, that is, dermally or transdermally. Typical formulations for this purpose include gels, hydrogels, lotions, solutions, creams, ointments, dusting powders, dressings, foams, films, skin patches, wafers, implants, sponges, fibres, bandages and microemulsions. Liposomes may also be used. Typical carriers include alcohol, water, mineral oil, liquid petrolatum, white petrolatum, glycerin, polyethylene glycol and propylene glycol. Penetration enhancers may be incorporated - see, for example, J Pharm Sci, 88 (10), 955-958 by Finnin and Morgan (October 1999).
Other means of topical administration include delivery by electroporation, iontophoresis, phonophoresis, sonophoresis and microneedle or needle-free (e.g. Powderject™, Bioject™, etc.) injection.
Formulations for topical administration may be formulated to be immediate and/or modified release. Modified release formulations include delayed-, sustained-, pulsed-, controlled-, targeted and programmed release.
The compounds of the invention can also be administered intranasally or by inhalation, typically in the form of a dry powder (either alone, as a mixture, for example, in a dry blend with lactose, or as a mixed component particle, for example, mixed with phospholipids, such as phosphatidylcholine) from a dry powder inhaler or as an aerosol spray from a pressurised container, pump, spray, atomiser (preferably an atomiser using electrohydrodynamics to produce a fine mist), or nebuliser, with or without the use of a suitable propellant, such as 1 ,1 ,1 ,2-tetrafluoroethane or 1 ,1 ,1 ,2,3,3,3-heptafluoropropane. For intranasal use, the powder may comprise a bioadhesive agent, for example, chitosan or cyclodextrin.
The pressurised container, pump, spray, atomizer, or nebuliser contains a solution or suspension of the compound(s) of the invention comprising, for example, ethanol, aqueous ethanol, or a suitable alternative agent for dispersing, solubilising, or extending release of the active, a propellant(s) as solvent and an optional surfactant, such as sorbitan trioleate, oleic acid, or an oligolactic acid.
Prior to use in a dry powder or suspension formulation, the drug product is micronised to a size suitable for delivery by inhalation (typically less than 5 microns). This may be achieved by any appropriate comminuting method, such as spiral jet milling, fluid bed jet milling, supercritical fluid processing to form nanoparticles, high pressure homogenisation, or spray drying.
Capsules (made, for example, from gelatin or hydroxypropylmethylcellulose), blisters and cartridges for use in an inhaler or insufflator may be formulated to contain a powder mix of the compound of the invention, a suitable powder base such as lactose or starch and a performance modifier such as I- leucine, mannitol, or magnesium stearate. The lactose may be anhydrous or in the form of the monohydrate, preferably the latter. Other suitable excipients include dextran, glucose, maltose, sorbitol, xylitol, fructose, sucrose and trehalose.
A suitable solution formulation for use in an atomiser using electrohydrodynamics to produce a fine mist may contain from 1 μg to 20mg of the compound of the invention per actuation and the actuation volume may vary from 1 μl to 10Oμl. A typical formulation may comprise a compound of formula (1 ), propylene glycol, sterile water, ethanol and sodium chloride. Alternative solvents which may be used instead of propylene glycol include glycerol and polyethylene glycol.
Suitable flavours, such as menthol and levomenthol, or sweeteners, such as saccharin or saccharin sodium, may be added to those formulations of the invention intended for inhaled/intranasal administration.
Formulations for inhaled/intranasal administration may be formulated to be immediate and/or modified release using, for example, PGLA. Modified release formulations include delayed-, sustained-, pulsed-, controlled-, targeted and programmed release.
In the case of dry powder inhalers and aerosols, the dosage unit is determined by a prefilled capsule, blister or pocket or by a system that utilises a gravimetrically fed dosing chamber . Units in accordance with the invention are typically arranged to administer a metered dose or "puff" containing from 1 to 5000 μg of (compound name here), or a salt thereof. The overall daily dose will typically be in the range 1 μg to 20 mg which may be administered in a single dose or, more usually, as divided doses throughout the day.
The compounds of formula (1 ) are particularly suitable for an administration by inhalation, in particular using a dry powder inhaler.
The compounds of the invention may be administered rectally or vaginally, for example, in the form of a suppository, pessary, or enema. Cocoa butter is a traditional suppository base, but various alternatives may be used as appropriate.
Formulations for rectal/vaginal administration may be formulated to be immediate and/or modified release. Modified release formulations include delayed-, sustained-, pulsed-, controlled-, targeted and programmed release. The compounds of the invention may also be administered directly to the eye or ear, typically in the form of drops of a micronised suspension or solution in isotonic, pH-adjusted, sterile saline. Other formulations suitable for ocular and aural administration include ointments, biodegradable (e.g. absorbable gel sponges, collagen) and non-biodegradable (e.g. silicone) implants, wafers, lenses and particulate or vesicular systems, such as niosomes or liposomes. A polymer such as crossed-linked polyacrylic acid, polyvinylalcohol, hyaluronic acid, a cellulosic polymer, for example, hydroxypropylmethylcellulose, hydroxyethylcellulose, or methyl cellulose, or a heteropolysaccharide polymer, for example, gelan gum, may be incorporated together with a preservative, such as benzalkonium chloride. Such formulations may also be delivered by iontophoresis.
Formulations for ocular/aural administration may be formulated to be immediate and/or modified release. Modified release formulations include delayed-, sustained-, pulsed-, controlled-, targeted, or programmed release.
The compounds of the invention may be combined with soluble macromolecular entities, such as cyclodextrin and suitable derivatives thereof or polyethylene glycol-containing polymers, in order to improve their solubility, dissolution rate, taste-masking, bioavailability and/or stability for use in any of the aforementioned modes of administration.
Drug-cyclodextrin complexes, for example, are found to be generally useful for most dosage forms and administration routes. Both inclusion and non-inclusion complexes may be used. As an alternative to direct complexation with the drug, the cyclodextrin may be used as an auxiliary additive, i.e. as a carrier, diluent, or solubiliser. Most commonly used for these purposes are alpha-, beta- and gamma- cyclodextrins, examples of which may be found in International Patent Applications Nos. WO 91/1 1172, WO 94/02518 and WO 98/55148.
Inasmuch as it may desirable to administer a combination of active compounds, for example, for the purpose of treating a particular disease or condition, it is within the scope of the present invention that two or more pharmaceutical compositions, at least one of which contains a compound in accordance with the invention, may conveniently be combined in the form of a kit suitable for co-administration of the compositions.
Thus the kit of the invention comprises two or more separate pharmaceutical compositions, at least one of which contains a compound of formula (1 ) in accordance with the invention, and means for separately retaining said compositions, such as a container, divided bottle, or divided foil packet. An example of such a kit is the familiar blister pack used for the packaging of tablets, capsules and the like.
The kit of the invention is particularly suitable for administering different dosage forms, for example parenteral, for administering the separate compositions at different dosage intervals, or for titrating the separate compositions against one another. To assist compliance, the kit typically comprises directions for administration and may be provided with a so-called memory aid. For administration to human patients, the total daily dose of the compounds of the invention is typically in the range 0.001 mg to 5000mg depending, of course, on the mode of administration. For example, an intravenous daily dose may only require from 0.001 mg to 40mg. The total daily dose may be administered in single or divided doses and may, at the physician's discretion, fall outside of the typical range given herein.
These dosages are based on an average human subject having a weight of about 65kg to 70kg. The physician will readily be able to determine doses for subjects whose weight falls outside this range, such as infants and the elderly.
For the avoidance of doubt, references herein to "treatment" include references to curative, palliative and prophylactic treatment.
According to another embodiment of the present invention, the compounds of the formula (1 ), or pharmaceutically acceptable salts thereof and/or pharmaceutically acceptable solvates of said compounds or salts, can also be used as a combination with one or more additional therapeutic agents to be co-administered to a patient to obtain some particularly desired therapeutic end result such as the treatment of pathophysiologically-relevant disease processes including, but not limited to (i) bronchoconstriction, (ii) inflammation, (iii) allergy, (iv) tissue destruction, (v) signs and symptoms such as breathlessness, cough. The second and more additional therapeutic agents may also be a compound of the formula (1 ), or pharmaceutically acceptable salts thereof and/or pharmaceutically acceptable solvates of said compounds or salts, or one or more β2 agonists known in the art. More typically, the second and more therapeutic agents will be selected from a different class of therapeutic agents. As used herein, the terms "co-administration", "co-administered" and "in combination with", referring to the compounds of formula (1 ) and one or more other therapeutic agents, is intended to mean, and does refer to and include the following:
• simultaneous administration of such combination of compound(s) of formula (1 ) and therapeutic agent(s) to a patient in need of treatment, when such components are formulated together into a single dosage form which releases said components at substantially the same time to said patient,
• substantially simultaneous administration of such combination of compound(s) of formula (1 ) and therapeutic agent(s) to a patient in need of treatment, when such components are formulated apart from each other into separate dosage forms which are taken at substantially the same time by said patient, whereupon said components are released at substantially the same time to said patient,
• sequential administration of such combination compound(s) of formula (1 ) and therapeutic agent(s) to a patient in need of treatment, when such components are formulated apart from each other into separate dosage forms which are taken at consecutive times by said patient with a significant time interval between each administration, whereupon said components are released at substantially different times to said patient; and
• sequential administration of such combination of compound(s) of formula (1 ) and therapeutic agent(s) to a patient in need of treatment, when such components are formulated together into a single dosage form which releases said components in a controlled manner whereupon they are concurrently, consecutively, and/or overlapingly administered at the same and/or different times by said patient, where each part may be administered by either the same or different route.
Suitable examples of other therapeutic agents which may be used in combination with the compound(s) of formula (1 ), or pharmaceutically acceptable salts thereof and/or pharmaceutically acceptable solvates of said compounds or salts, include, but are by no means limited to:
(a) 5-Lipoxygenase (5-LO) inhibitors or 5-lipoxygenase activating protein (FLAP) antagonists;
(b) Leukotriene antagonists (LTRAs) including antagonists of LTB4, LTC4, LTD4, and LTE4;
(c) Histamine receptor antagonists including H1 and H3 antagonists;
(d) OC1- and oc2-adrenoceptor agonist vasoconstrictor sympathomimetic agents for decongestant use;
(e) PDE inhibitors, e.g. PDE3, PDE4 and PDE5 inhibitors;
(f) Beta 2 receptor agonists;
(g) muscarinic M3 receptor antagonist or anticholinergic agents; (h) Theophylline;
(i) Sodium cromoglycate;
(j) COX inhibitors both non-selective and selective COX-1 or COX-2 inhibitors (NSAIDs);
(k) Prostaglandin receptor antagonists and inhibitors of prostaglandin synthase;
(I) Oral and inhaled glucocorticosteroids;
(m) Dissociated agonists of the corticoid receptor (DAGR);
(n) Monoclonal antibodies active against endogenous inflammatory entities;
(o) Anti-tumor necrosis factor (anti-TNF-α) agents;
(p) Adhesion molecule inhibitors including VLA-4 antagonists;
(q) KJnJn-B1 - and B2 -receptor antagonists;
(r) Immunosuppressive agents, including inhibitors of the IgE pathway and cyclosporine;
(s) Inhibitors of matrix metalloproteases (MMPs);
(t) Tachykinin NK1, NK2 and NK3 receptor antagonists;
(u) Protease inhibitors such as elastase inhibitors;
(v) Adenosine A2a receptor agonists and A2b antagonists;
(w) Inhibitors of urokinase;
(x) Compounds that act on dopamine receptors, such as D2 agonists;
(y) Modulators of the NFiφ pathway, such as IKK inhibitors;
(z) modulators of cytokine signalling pathyways such as p38 MAP kinase, PI3 kinase, JAK kinase, syk kinase, EGFR or MK-2;
(aa) Agents that can be classed as mucolytics or anti-tussive; (bb) Agents which enhance responses to inhaled corticosteroids; (cc) Antibiotics and antivral agents effective against micro-organisms which can colonise the respiratory tract;
(dd) HDAC inhibitors; (ee) CXCR2 antagonists; (ff) lntegrin antagonists; (gg) Chemokines; (hh)Epithelial sodium channel (ENaC) blockers or Epithelial sodium channel (ENaC) inhibitors;
(ii) P2Y2 Agonists and other Nucleotide receptor agonists;
(jj) Inhibitors of thromboxane;
(kk) Inhibitors of PGD2 synthesis and PGD2 receptors (DP1 and DP2/CRTH2);
(II) Niacin; and
(mm) Adhesion factors including VLAM, ICAM, and ELAM.
According to the present invention, combination of the compounds of formula (1 ) with:
- H3 antagonists;
- Muscarinic M3 receptor antagonists;
- PDE4 inhibitors;
- glucocorticosteroids;
- Adenosine A2a receptor agonists;
- Modulators of cytokine signalling pathyways such as p38 MAP kinase or syk kinase; or
- Leukotriene antagonists (LTRAs) including antagonists of LTB4, LTC4, LTD4, and LTE4 are further preferred.
According to the present invention, combination of the compounds of formula (1 ) with: glucocorticosteroids, in particular inhaled glucocorticosteroids with reduced systemic side effects, including prednisone, prednisolone, flunisolide, triamcinolone acetonide, beclomethasone dipropionate, budesonide, fluticasone propionate, ciclesonide, and mometasone furoate, or muscarinic M3 receptor antagonists or anticholinergic agents including in particular ipratropium salts, namely bromide, tiotropium salts, namely bromide, oxitropium salts, namely bromide, perenzepine, and telenzepine are further preferred.
It is to be appreciated that all references herein to treatment include curative, palliative and prophylactic treatment.
The compounds of formula (1 ) have the ability to interact with the β2 receptor and cholinergic muscarinic receptors, and thereby have a wide range of therapeutic applications, as described further below, because of the essential role which the β2 receptor and muscarinic receptors play in the physiology of all mammals.
Therefore, a further aspect of the present invention relates to the compounds of formula (1 ), or pharmaceutically acceptable salts thereof and/or pharmaceutically acceptable solvates of said compounds or salts, for use in the treatment of diseases, disorders, and conditions in which the β2 receptor and /or muscarinic receptors are involved. More specifically, the present invention also concerns the compounds of formula (1 ), or pharmaceutically acceptable salts thereof and/or pharmaceutically acceptable solvates of said compounds or salts, for use in the treatment of diseases, disorders, and conditions selected from the group consisting of: • asthma of whatever type, etiology, or pathogenesis, in particular asthma that is a member selected from the group consisting of atopic asthma, non-atopic asthma, allergic asthma, atopic bronchial IgE-mediated asthma, bronchial asthma, essential asthma, true asthma, intrinsic asthma caused by pathophysiologic disturbances, extrinsic asthma caused by environmental factors, essential asthma of unknown or inapparent cause, non-atopic asthma, bronchitic asthma, emphysematous asthma, exercise-induced asthma, allergen induced asthma, cold air induced asthma, occupational asthma, infective asthma caused by bacterial, fungal, protozoal, or viral infection, non-allergic asthma, incipient asthma, wheezy infant syndrome and bronchiolytis;
• chronic or acute bronchoconstriction, chronic bronchitis, small airways obstruction, and emphysema;
• obstructive or inflammatory airways diseases of whatever type, etiology, or pathogenesis, in particular an obstructive or inflammatory airways disease that is a member selected from the group consisting of chronic eosinophilic pneumonia, chronic obstructive pulmonary disease (COPD), COPD that includes chronic bronchitis, pulmonary emphysema or dyspnea associated or not associated with COPD, COPD that is characterized by irreversible, progressive airways obstruction, adult respiratory distress syndrome (ARDS), exacerbation of airways hyper-reactivity consequent to other drug therapy and airways disease that is associated with pulmonary hypertension;
• bronchitis of whatever type, etiology, or pathogenesis, in particular bronchitis that is a member selected from the group consisting of acute bronchitis, acute laryngotracheal bronchitis, arachidic bronchitis, catarrhal bronchitis, croupus bronchitis, dry bronchitis, infectious asthmatic bronchitis, productive bronchitis, staphylococcus or streptococcal bronchitis and vesicular bronchitis;
• acute lung injury; and
• bronchiectasis of whatever type, etiology, or pathogenesis, in particular bronchiectasis that is a member selected from the group consisting of cylindric bronchiectasis, sacculated bronchiectasis, fusiform bronchiectasis, capillary bronchiectasis, cystic bronchiectasis, dry bronchiectasis and follicular bronchiectasis.
A still further aspect of the present invention also relates to the use of the compounds of formula (1 ), or pharmaceutically acceptable salts thereof and/or pharmaceutically acceptable solvates of said compounds or salts, for the manufacture of a drug having a β2 agonist activity and an M3 antagonist activity. In particular, the present inventions concerns the use of the compounds of formula (1 ), or pharmaceutically acceptable salts thereof and/or pharmaceutically acceptable solvates of said compounds or salts, for the manufacture of a drug for the treatment of diseases and/or conditions involving the beta 2 and muscarinic receptors, in particular the diseases and/or conditions listed above.
As a consequence, the present invention provides a particularly interesting method to treat a mammal, including a human being, with an effective amount of a compound of formula (1 ), or pharmaceutically acceptable salts thereof and/or pharmaceutically acceptable solvates of said compounds or salts. More precisely, the present invention provides a particularly interesting method for the treatment of a β2- mediated diseases and/or conditions involving the beta 2 and muscarinic receptors, in a mammal, including a human being, in particular the diseases and/or conditions listed above, comprising administering said mammal with an effective amount of a compound of formula (1 ), its pharmaceutically acceptable salts and/or solvates of said compounds or salts.
The following examples illustrate the preparation of the compounds of formula (1 ):
FIGURE
Figure 1/1 : PXRD Pattern of Example 2a
PROTOCOLS
For all examples below, the following experimental conditions were used:
Powder X-ray diffraction method (PXRD)
The powder X-ray diffraction pattern was determined using a Bruker-AXS Ltd. D4 powder X-ray diffractometer fitted with an automatic sample changer, a theta-theta goniometer, automatic beam divergence slit, and a PSD Vantec-1 detector. The sample was prepared for analysis by mounting on a low background cavity silicon wafer specimen mount. The peaks obtained were aligned against a silicon reference standard. The specimen was rotated whilst being irradiated with copper K-alpha-i X -rays (wavelength = 1.5406 Angstroms) with the X-ray tube operated at 40kV/35mA. The analyses were performed with the goniometer running in continuous mode set for a 0.2 second count per 0.018° step over a two theta range of 2° to 55°
PREPARATIONS
Preparation 1 (2R)-2-cyclohexyl-2-hvdroxy-2-phenylacetamide
Figure imgf000026_0001
(2R)-2-Cyclohexyl-2-hydroxy-2-phenylacetic acid (preparation 36, 4.87g, 20.8mmol) was dissolved in dichloromethane (15OmL) and carbonyldiimidazole (3.37g, 20.8mmol) added in one portion. After stirring for 1 hour at room temperature, 0.880 ammonia (21 mL) was added and stirring continued at room temperature for 18 hours. The organic layer was separated, washed with brine (5OmL), dried over magnesium sulphate, filtered and the solvent removed in vacuo to furnish the title compound as a white foam in 92% yield, 4.56g. LRMS: APCI ESI m/z 232 [M-H]"
1H NMR (400 MHz, CHLOROFORM-c/) δ = 0.92 (m, 1 H), 1.15 (m, 5H), 1.76 (m, 4H), 2.40 (m, 1 H), 5.38 (br.s, 1 H), 6.52 (br.s, 1 H), 7.28 (m, 1 H), 7.37 (m, 2H), 7.62 (m, 2H) ppm.
Preparation 2
(R)-cyclohexyl(phenyl)1 H-1 ,2,4-triazol-3-ylmethanol
Figure imgf000027_0001
(2R)-2-cyclohexyl-2-hydroxy-2-phenylacetamide (Preparation 1 , 4.497g, 19.27mnnol) was dissolved in
N,N-dimethylfornnannide dimethylacetal (60ml). After stirring at 9O0C for 2 hours the solvent was removed in vacuo and residue azeotroped with toluene (100ml) to give a yellow oil. Hydrazine hydrate
(1.11 ml, 22.9mmol) was added followed by glacial acetic acid (90ml). After stirring at 9O0C for 2 hours the solvent was removed in vacuo and the residue partitioned between ethyl acetate (250ml) and saturated aqueous sodium bicarbonate solution (25OmL). The organic layer was separated, dried over magnesium sulphate, filtered and the solvent removed in vacuo. The residue was purified by column chromatography on silica gel eluting with ethyl acetate to furnish the title compound as a white solid, in
100% yield, 4.9g.
LRMS: ESI m/z 256 [M-H]"
1H NMR (400 MHz, CHLOROFORM-c/) δ = 1.08 (m, 3H), 1.36 (m, 2H), 1.70 (m, 5H), 2.46 (m, 1 H), 3.08
(br.s, 1 H), 7.23 (m, 1 H), 7.34 (m, 2H), 7.65 (m, 2H), 8.00(s, 1 H) ppm.
Enantiomeric excess: 99.3%, calculated using a Chiralpak AS-H Column (250 x 4.6 mm) eluting with
79% heptane, 31 % isopropyl alcohol at a flow rate of 1 ml/min. Retention time of desired (R)-enantiomer
13.61 mins, retention time of undesired (S)-enantiomer 1 1.31 mins (identified from racemate).
Preparation 3 tert-butyl 4-({3-[(R)-cvclohexyl(hvdroxy)phenylmethyl1-1H-1 ,2,4-triazol-1-yl)methyl)piperidine-1- carboxylate
Figure imgf000027_0002
Tert-butyl 4-(bromomethyl)piperidine-1-carboxylate (6.75g, 24.2mmol) was dissolved in DMF (62ml) and (R)-cyclohexyl(phenyl)1 H-1 ,2,4-triazol-3-ylmethanol (Preparation 2, 5.2g, 20mmol) added, followed by potassium carbonate (5.58g, 40.4mmol). After stirring at 7O0C for 18 hours the solvent was removed in vacuo, and the residue partitioned between ethyl acetate (150ml) and water (15OmL). The organic layer was separated, dried over magnesium sulphate, filtered and the solvent removed in vacuo. The residue was purified by column chromatography on silica gel eluting with ethyl acetate:heptane, (1 :4 to
1 :1 , by volume) to furnish the title compound as a white foam, in 51 % yield, 4.7g (contains 10% of the other triazole regio-isomer).
LRMS: APCI ESI m/z 487 [M+H+MeOH]+
1H NMR (400 MHz, METHANOL-c/4) δ = 1.05-1.36 (m, 9H), 1.43 (s, 9H), 1.47-1.75 (m, 5H), 2.09 (m, 1 H),
2.38 (m, 1 H), 2.72 (m, 2H), 4.08 (m, 4H), 7.17 (m, 1 H), 7.26 (m, 2H), 7.58 (d, 2H), 8.30 (s, 1 H) ppm. Alternatively, the title compound was prepared according to the following procedure: (R)-cyclohexyl(phenyl)1 H-1 ,2,4-triazol-3-ylmethanol (Preparation 2, 3.Og, 11.7mnnol) was dissolved in acetone (6OmL) and tert-butyl 4-(bromomethyl)piperidine-1-carboxylate (3.24g, 1 1.7mmol) added, followed by cesium carbonate (7.6Og, 23.3mmol). After stirring at 7O0C for 5 hours, the reaction was allowed to cool to room temperature and stirred for a further 16 hours. The solvent was removed in vacuo, and the residue partitioned between ethyl acetate (5OmL) and water (5OmL). The organic layer was separated, dried over magnesium sulphate, filtered and the solvent removed in vacuo. 100mg of this residue was dissolved in acetonitrile (0.5ml) and left to evaporate, almost to dryness, at which point the resulting oil began to crystallise. The remainder of the residue was dissolved in acetonitrile (55ml) and seeded with the crystalline material obtained above. Crystallisation was allowed to occur over 18 hours and the resulting solid collected by filtration to furnish the title compound as a white solid, in 64% yield, 3.4g.
LRMS: APCI ESI m/z 487 [M+H+MeOH]+
1H NMR (400 MHz, METHANOL-c/4) δ = 1.05-1.36 (m, 9H), 1.43 (s, 9H), 1.47-1.75 (m, 5H), 2.09 (m, 1 H), 2.38 (m, 1 H), 2.72 (m, 2H), 4.08 (m, 4H), 7.17 (m, 1 H), 7.26 (m, 2H), 7.58 (d, 2H), 8.30 (s, 1 H) ppm.
Preparation 4
(R)-cvclohexyl(phenyl)[1 -(piperidin-4-ylmethyl)-1 H-1 ,2,4-triazol-3-yl1methanol
Figure imgf000028_0001
Tert-butyl 4-({3-[(R)-cyclohexyl(hydroxy)phenylmethyl]-1H-1 ,2,4-triazol-1-yl}methyl)piperidine-1- carboxylate (Preparation 3, 5.6Og, 12.3mmol) was dissolved in DCM (31 ml) and 2M HCI in ether (31 ml, 60mmol) added. After stirring at room temperature for 5 hours the solvent was removed in vacuo, and the residue partitioned between dichloromethane (200ml) and saturated aqueous sodium bicarbonate solution (200ml). The organic layer was separated, dried over magnesium sulphate, filtered and the solvent removed in vacuo to furnish the title compound as a white foam, in 92% yield, 4.03g (contains 10% of other triazole regio-isomer). LRMS: APCI ESI m/z 377 [M+Na]+
1H NMR (400 MHz, METHANOL-c/4) δ = 1.04-1.36 (m, 9H), 1.38-1.57 (m, 2H), 1.58-1.81 (m, 3H), 2.00 (m, 1 H), 2.38 (m, 1 H), 2.58 (m, 2H), 3.06 (m, 2H), 4.06 (d, 2H), 7.17 (m, 1 H), 7.25 (m, 2H), 7.58 (d, 2H), 8.31 (s, 1 H) ppm.
Alternatively, the title compound was prepared according to the following procedure: Tert-butyl 4-({3-[(R)-cyclohexyl(hydroxy)phenylmethyl]-1H-1 ,2,4-triazol-1-yl}methyl)piperidine-1- carboxylate (Preparation 3a, 2.0Og, 4.40mmol) was dissolved in dioxane (11 ml) and stirred vigorously to achieve solubilisation. 4M HCI in dioxane (5.54mL, 22.1 mmol) was then added. After stirring at room temperature for 24 hours the solvent was removed in vacuo, and the residue partitioned between dichloromethane (5OmL) and saturated aqueous sodium bicarbonate solution (5OmL). The organic layer was separated, dried over magnesium sulphate, filtered and the solvent removed in vacuo to furnish the title compound as a colourless oil, in 96% yield, 1.5g. LRMS: APCI ESI m/z 377 [IVRNa]+
1H NMR (400 MHz, METHANOL-c/4) δ = 1.04-1.36 (m, 9H), 1.38-1.57 (m, 2H), 1.58-1.81 (m, 3H), 2.00 (m, 1 H), 2.38 (m, 1 H), 2.58 (m, 2H), 3.06 (m, 2H), 4.06 (d, 2H), 7.17 (m, 1 H), 7.25 (m, 2H), 7.58 (d, 2H), 8.31 (s, 1 H) ppm.
Preparation 5
Di-tert-butyl {9-[4-({3-[(R)-cvclohexyl(hvdroxy)phenylmethyl1-1H-1 ,2,4-triazol-1-yl)methyl)piperidin-1- ylinonvDimidodicarbonate
Figure imgf000029_0001
(R)-cyclohexyl(phenyl)[1-(piperidin-4-ylmethyl)-1H-1 ,2,4-triazol-3-yl]methanol (Preparation 4, 2.0Og, 5.64mmol) and di-tert-butyl (9-bromononyl)imidodicarbonate (US04167167, 2.38g, 5.64mmol) were dissolved in acetonitrile (60ml) and triethylamine (2.35ml, 16.9mmol) added. After stirring at 5O0C for 18 hours, the solvent was removed in vacuo, and the residue purified by column chromatography on silica gel eluting with dichloromethane:methanol:880 ammonia (98:2:0.2, by volume) to furnish the title compound as a clear oil, in 60% yield, 2.6g (contains 10% of other triazole regio-isomer) LRMS: APCI ESI m/z 696 [M+H]+
1H NMR (400 MHz, METHANOL-c/4) δ = 1.14 (m, 4H), 1.25-1.38 (m, 18H) 1.41-1.51 (m, 18H), 1.51-1.75 (m, 6H), 1.99 (m, 2H), 2.14 (m, 2H), 2.42 (m, 2H), 3.06 (m, 2H), 3.55 (m, 2H), 4.09 (m, 2H), 7.16 (m, 1 H), 7.26 (m, 2H), 7.58 (m, 2H), 8.32 (m, 1 H) ppm.
Preparation 6
(R)-(I -Ifi-O-aminononvDpiperidin^-ylimethvD-IH-I ^Λ-triazol-S-ylϊcvclohexyl) phenylmethanol
Figure imgf000029_0002
Di-tert-butyl {9-[4-({3-[(R)-cyclohexyl(hydroxy)phenylnnethyl]-1H-1 ,2,4-triazol-1-yl}nnethyl)piperidin -1- yl]nonyl}imidodicarbonate (Preparation 5, 2.6g, 3.7mnnol) was dissolved in dichloromethane (15ml) and hydrogen chloride in diethylether (2M, 15ml, 30mmol) added. After stirring at room temperature for 5 hours the solvent was removed in vacuo, the residue was partitioned between dichloromethane (200ml) and saturated aqueous sodium bicarbonate solution (200ml). The organic layer was separated, dried over magnesium sulphate, filtered and the solvent removed in vacuo. The residue was purified by column chromatography on silica gel eluting with dichloromethane:methanol:880 ammonia (97.5:2.5:0.25, by volume) to furnish the title compound as a clear oil, in 70% yield, 1.29g (contains 10% of other triazole regio-isomer). LRMS: APCI m/z 496 [M+H]+
1H NMR (400 MHz, METHANOL-d4) δ = 1.12 (m, 4H), 1.23-1.34 (m, 18H), 1.49 (m, 6H), 1.64 (m, 2H), 1.94 (m, 2H), 2.31 (m, 2H), 2.63 (m, 2H), 2.93 (m, 2H), 4.07 (m, 2H), 7.16 (m, 1 H), 7.25 (m, 2H), 7.58 (m, 2H), 8.31 (s, 1 H) ppm.
Preparation 7
8-(benzyloxyV5-r(1 R)-1-(rtert-butyl(dimethvnsilvnoxy)-2-a9-r4-a3-r(RVcvclohexyl(hvdroxy) phenylmethyl1-1H-1 ,2,4-triazol-1-yl)methyl)piperidin-1-yl1nonyl)amino)ethyl1quinolin-2(1 H)-one
Figure imgf000030_0001
(R)-(I -{[1-(9-aminononyl)piperidin-4-yl]methyl}-1H-1 ,2,4-triazol-3-yl)(cyclohexyl) phenylmethanol
(Preparation 6, 350mg, 0.706mg), 8-(benzyloxy)-5-[(1 R)-2-bromo-1-{[tert- butyl(dimethyl)silyl]oxy}ethyl]quinolin-2(1 H)-one (WO200509286, 345mg, 0.706mmol) and sodium hydrogen carbonate (88.9mg, 1.06mmol) were combined in acetonitrile (7ml_). After stirring at 9O0C for 72 hours the solvent was removed in vacuo and residue partitioned between dichloromethane (50ml) and saturated aqueous sodium bicarbonate solution (5OmL). The organic layer was separated, dried over magnesium sulphate, filtered and the solvent removed in vacuo. The residue was purified by column chromatography on silica gel eluting with dichloromethane:methanol:880 ammonia (99:1 :0.1 to 95:5:0.5 by volume) to furnish the title compound as a clear glass, in 37% yield, 240mg (contains 10% of other triazole regio-isomer). LRMS: APCI ESI m/z 904 [M+H]+
1H NMR (400 MHz, METHANOL-c/4) δ = 0.07 (s, 3H), 0.37 (s, 3H), 1.17 (s, 9H), 1.37-1.49 (m, 4H), 1.52- 1.63 (m, 18H), 1.76 (m, 6H), 1.92 (m, 2H), 2.20 (m, 2H), 2.60 (m, 2H), 2.88 (m, 2H), 3.01 (m, 1 H), 3.18 (m, 3H), 4.35 (m, 2H), 5.50 (m, 1 H), 5.61 (s, 2H), 6.95 (d, 1 H), 7.41-7.70 (m, 8H), 7.79 (m, 2H), 7.86 (m, 2H), 8.59 (s, 1 H), 8.74 (d, 1 H) ppm.
Preparation 8 δ-fd RVI-lftert-butvKdimethvnsilylloxyl^-αQ-μ-αS-^RVcvclohexyKhvdroxy'lphenylmethyll-IH-I ^Λ- triazol-i-vDπnethvDpiperidin-i-ylinonvDaπnino^ethyli-δ-hvdroxyquinolin^d HVone
Figure imgf000031_0001
8-(benzyloxy)-5-[(1 R)-1-{[tert-butyl(dimethyl)silyl]oxy}-2-({9-[4-({3-[(R)-cyclohexyl(hydroxy) phenylmethyl]-1H-1 ,2,4-triazol-1-yl}methyl)piperidin-1-yl]nonyl}amino)ethyl]quinolin-2(1 H)-one (Preparation 7, 230mg, 0.255mnnol) was dissolved in ethanol (10ml) and palladium hydroxide [20wt.%(dry basis) on carbon(wet)] (5mg) added followed by ammonium formate (161 mg, 2.55mmol). After stirring at reflux for 1 hour the reaction mixture was filtered through arbocel® and the solvent removed in vacuo. The residue was purified by column chromatography on silica gel eluting with dichloromethane:methanol:880 ammonia (97.5:2.5:0.25 to 90:10:1 , by volume) to furnish the title compound as a glass, in 87% yield, 180mg. LRMS: APCI ESI m/z 813 [M+H]+
1H NMR (400 MHz, METHANOL-c/4) δ = 0.00 (s, 3H), 0.30 (s, 3H), 1.10 (s, 9H), 1.45-1.94 (m, 28H), 2.21 (m, 3H), 2.58 (m, 3H ), 2.85 (m, 2H ), 2.95 (m, 1 H), 3.17 (m, 3H), 4.29 (m, 2H), 5.42 (m, 1 H), 6.85 (d, 1 H), 7.14 (d, 1 H), 7.32 -7.48 (m, 4H), 7.78 (m, 2H), 8.52 (s, 1 H), 8.65 (d, 1 H) ppm.
Preparation 9
2-{2-[4-(2-Hvdroxy-ethyl)-phenyl1-vinyl)-isoindole-1 ,3-dione
Figure imgf000031_0002
To a solution of 2-(4-bromophenyl)ethanol (48.3g) in acetonitrile (480 ml_) was added diisopropylethylamine (46.6g), N-vinylphthalimide (43.7g) and tri-o-tolylphosphine (7.31g) and the mixture was purged with nitrogen gas three times. Palladium acetate (2.7g) was added and the mixture was stirred at 9O0C for 21 hr under nitrogen. The reaction was cooled and the precipitated product collected by filtration. The resulting solid was re-dissolved in dichloromethane and ethyl acetate and filtered through silica gel. The filtrate was concentrated in vacuo to give the title compound, 24g. 1H NMR (400MHz, CDCI3) δ = 2.81 -2.84 (t, 2H), 3.82-3.90 (t, 2H), 7.23-7.26 (d, 2H) 7.32-7.36 (d, 1 H), 7.40-7.43 (d, 2H), 7.61-7.64 (d, 1 H), 7.66-7.78 (d, 2H), 7.86-7.88 (d, 2H) ppm.
Preparation 10 2-{2-[4-(2-Hvdroxy-ethyl)-phenyl1-ethyl)-isoindole-1 ,3-dione
Figure imgf000032_0001
To a stirred solution of 2-{2-[4-(2-Hydroxy-ethyl)-phenyl]-vinyl}-isoindole-1 ,3-dione (Preparation 9, 1Og) in ethanol and ethyl acetate (350 ml_ each) was added 30% palladium hydroxide on carbon (1.44g) followed by ammonium formate (21.5g). The reaction was heated to 8O0C for 4 hours. Additional palladium hydroxide on carbon (1.44g) and ammonium formate (21.5g) were added and the reaction was stirred at 8O0C for 18 hours. The reaction was cooled, filtered through a pad of Arbocel™ and rinsed with methanol. The solvent was removed under reduced pressure and the resulting white solid was partitioned between dichloromethane (200ml) and water (100ml). The aqueous layer was separated and extracted with further dichloromethane (2 x 50 ml_). The combined organic layers were dried (sodium sulphate) and the solvent removed in vacuo to give the title compound as an off-white solid, 9.11g. 1H NMR (400MHz, CDCI3) δ = 2.80-2.83 (t, 2H), 2.92-3.00 (t, 2H), 3.82-3.86 (t, 2H), 3.87-3.96 (t, 2H), 7.14-7.22 (2x d, 4H), 7.70-7.72 (dd, 2H), 7.82-7.84 (dd, 2H) ppm.
Alternatively, the title compound may be prepared according to the following procedure:
2-{2-[4-(2-hydroxy-ethyl)phenyl]-vinyl}-isoindole-1 ,3-dione (Preparation 9, 62.Og, 21 1.37mmol) was dissolved in ethyl acetate (120OmL). To this was added rhodium tris(triphenylphosphine) chloride, (12.7g, 13.7mmol) and the mixture hydrogenated at 20psi, room temperature for 24 hours. The reaction was filtered and concentrated in vacuo. The residue was dissolved in ethyl acetate (100OmL) and passed through a pad of silica gel, washing with ethyl acetate. The solvent was removed in vacuo to yield a light brown solid which was recrystallised from ethyl acetate:heptane (4:1 , by volume) to yield the title compound as an off-white crystalline solid, in 85% yield, 53g,
1H NMR (400MHz, CDCI3) δ = 2.80-2.83 (t, 2H), 2.92-3.00 (t, 2H), 3.82-3.86 (t, 2H), 3.87-3.96 (t, 2H), 7.14-7.22 (2x d, 4H), 7.70-7.72 (dd, 2H), 7.82-7.84 (dd, 2H) ppm.
Preparation 11
2-{2-[4-(2-Bromo-ethyl)-phenyl1-ethyl)-isoindole-1 ,3-dione
Figure imgf000032_0002
A solution of 2-{2-[4-(2-Hydroxy-ethyl)-phenyl]-ethyl}-isoindole-1 ,3-dione (Preparation 10, 22.37g) and phosphorus tribromide (8.2Og) in toluene (500 mL) was refluxed for 4 hours. The mixture was allowed to cool to room temperature, diluted with ethyl acetate (300 mL) and carefully quenched with sodium bisulphite/sodium bicarbonate (1 :1 ) in water (100 mL). The organic layer was separated and washed with further sodium bisulphite/sodium bicarbonate (1 :1 ) in water (100 mL), dried over sodium sulphate and concentrated in vacuo. The resulting solid (24.26g) was triturated with heptane:tert-butyl methyl ether (100 mL; 9:1 , by volume) to give the title compound as a pale green solid, 17.94g. 1H NMR (400MHz, CDCI3) δ = 2.93-3.00 (t, 2H), 3.07-3.18 (t, 2H), 3.53-3.59 (t, 2H), 3.88-3.94 (t, 2H), 7.13-7.23 (2xd, 4H), 7.70-7.73 (dd, 2H), 7.83-7.85 (dd, 2H) ppm.
Preparation 12
2-[2-(4-{2-[4-({3-[(R)-cvclohexyl(hvdroxy)phenylmethyl1-1H-1 ,2,4-triazol-1-yl)methyl)piperidin-1- Vl1ethyl)phenyl)ethyl1-1 H-isoindole-1 ,3(2H)-dione
Figure imgf000033_0001
Cyclohexyl-phenyl-^-piperidin^-ylmethyl-IH-π ^^triazol-S-yO-methanol (Preparation 4, 8.2g, 2.98mmol), 2-{2-[4-(2-Bromo-ethyl)-phenyl]-ethyl}-isoindole-1 ,3-dione (Preparation 11 , 7.56g, 21.1 mmol) and triethylamine (13.3ml, 95.9mnnol) were dissolved in acetonitrile (100ml). After stirring at 9O0C for 48 hours the solvent was removed in vacuo, and the residue partitioned between dichloromethane (200ml) and water (20OmL). The organic layer was separated, dried over magnesium sulphate, filtered and the solvent removed in vacuo. The residue was purified by column chromatography on silica gel eluting with dichloromethane:methanol:880 ammonia (98:2:0.2, by volume) and the resulting material then triturated in tert-butyl methyl ether to furnish title compound as a clear solid in 28% yield, 3.4g. LRMS: APCI ESI m/z 632 [M+H]+
1H NMR (400 MHz, METHANOL-c/4) δ = 1.13 (m, 3H), 1.22-1.37 (m, 5H), 1.43 (m, 1 H), 1.50-1.74 (m, 5H), 1.94 (m, 1 H), 2.03 (m, 2H), 2.38 (m, 1 H), 2.52 (m, 2H), 2.73 (m, 2H), 2.90-3.02 (m, 4H), 3.86 (t, 2H), 4.08 (d, 2H), 7.06-7.17 (m, 5H), 7.25 (m, 2H), 7.58 (m, 2H), 7.78 (m, 4H), 8.32 (s, 1 H) ppm.
Alternatively, the title compound may be prepared according to the following procedure: Cyclohexyl-phenyl-(1-piperidin-4-ylmethyl-1H-[1 ,2,4]triazol-3-yl)-methanol (Preparation 4, 0.4Og, 1.13mmol) and 2-{2-[4-(2-Bromo-ethyl)-phenyl]-ethyl}-isoindole-1 ,3-dione (Preparation 11 , 0.404g, 1.13mmol) and diisopropylethylamine (0.59ml, 3.38mmol) were dissolved in methyl ethyl ketone (8mL). After stirring at 9O0C for 24 hours the reaction was allowed to cool slowly to room temperature. Crystallisation occured and the resulting solid was collected by filtration and dried in vacuo to yield the title compound as a cream coloured solid, in 74% yield, 0.532g. LRMS: APCI ESI m/z 632 [M+H]+
1H NMR (400 MHz, METHANOL-c/4) δ = 1.13 (m, 3H), 1.22-1.37 (m, 5H), 1.43 (m, 1 H), 1.50-1.74 (m, 5H), 1.94 (m, 1 H), 2.03 (m, 2H), 2.38 (m, 1 H), 2.52 (m, 2H), 2.73 (m, 2H), 2.90-3.02 (m, 4H), 3.86 (t, 2H), 4.08 (d, 2H), 7.06-7.17 (m, 5H), 7.25 (m, 2H), 7.58 (m, 2H), 7.78 (m, 4H), 8.32 (s, 1 H) ppm.
Alternatively, the title compound was also prepared according to the following procedure: Methanesulfonic acid 2-{4-[2-(1 ,3-dioxo-1 ,3-dihydro-isoindol-2-yl)-ethyl]-phenyl}-ethyl ester (Preparation 35, 20.0Og, 53.56mmol) was dissolved in acetonitrile (8OmL) and stirred at room temperature. Sodium iodide (16.06g, 107.12mmol) was added portionwise and the resulting slurry heated to 8O0C for 18h. The reaction mixture was cooled to 5O0C and further acetonitrile (8OmL) was added. To this mixture was added cyclohexyl-phenyl-(1-piperidin-4-ylmethyl-1H-[1 ,2,4]triazol-3-yl)-methanol (Preparation 4, 15.87g,
53.56mmol) and diisopropylethylamine (9.79ml_, 56.24mmol) and heating was continued for 6 hours, then allowed to cool to room temperature. Water (16OmL) was added and the resulting slurry stirred overnight at room temperature. The solid was collected by filtration and washed with acetonitrile
(5x20mL), then dried in vacuo at 450C for 8 hours to yield the title compound as a brown solid in 68% yield, 20.9g,
LRMS: APCI ESI m/z 632 [M+H]+
1H NMR (400 MHz, METHANOL-c/4) δ = 1.13 (m, 3H), 1.22-1.37 (m, 5H), 1.43 (m, 1 H), 1.50-1.74 (m,
5H), 1.94 (m, 1 H), 2.03 (m, 2H), 2.38 (m, 1 H), 2.52 (m, 2H), 2.73 (m, 2H), 2.90-3.02 (m, 4H), 3.86 (t,
2H), 4.08 (d, 2H), 7.06-7.17 (m, 5H), 7.25 (m, 2H), 7.58 (m, 2H), 7.78 (m, 4H), 8.32 (s, 1 H) ppm.
Preparation 13
(RH1-[(1-(2-[4-(2-aminoethvnphenyllethyl)piperidin-4-vnmethyll-1H-1 ,2,4-triazol-3- ylKcvclohexyDphenylmethanol
Figure imgf000034_0001
2-[2-(4-{2-[4-({3-[(R)-cyclohexyl(hydroxy)phenylmethyl]-1H-1 ,2,4-triazol-1-yl}methyl)piperidin-1- yl]ethyl}phenyl)ethyl]-1H-isoindole-1 ,3(2H)-dione (Preparation 12, 3.4g, 5.38mmol) was suspended in ethanol (20ml) and hydrazine monohydrate (2.61 ml, 53.8ml) added. After stirring at reflux for 2 hours, the reaction was cooled to room temperature and the precipitate collected by filtration and washed with ethanol (200ml). The filtrate was concentrated in vacuo to furnish the title compound as a white solid, in
81 % yield, 2.57g.
LRMS: ESI m/z 502 [M+H]+
1H NMR (400 MHz, METHANOL-c/4) δ = 1.04-1.75 (m, 14H), 1.90-2.08 (m, 3H), 2.38 (m, 1 H), 2.54 (m,
2H), 2.68 - 2.78 (m, 4H), 2.84 (m, 2H), 2.99 (m, 2H), 3.96 (d, 2H), 7.09-7.17 (m, 4H), 7.12 (m, 1 H), 7.25
(m, 2H), 7.59 (m, 2H), 8.31 (s, 1 H) ppm.
Preparation 14
8-(benzyloxy')-5-[(1 R)-1-([tert-butyl(dimethvnsilylloxy)-2-([2-(4-(2-[4-((3-[(R)-cvclohexyl (hvdroxy)phenylmethyl1-1H-1 ,2,4-triazol-1-yl)methyl)piperidin-1-yl1ethyl)phenyl)ethyl1 amino)ethyl1quinolin-2(1 H)-one
Figure imgf000035_0001
The title compound was prepared from (R)-{1-[(1-{2-[4-(2-aminoethyl)phenyl]ethyl}piperidin-4-yl)methyl]- 1H-1 ,2,4-triazol-3-yl}(cyclohexyl)phenylmethanol (Preparation 13, 2.57g, 5.12 mmol) and δ-(benzyloxy)- 5-[(1 R)-2-bromo-1-{[tert-butyl(dimethyl)silyl]oxy}ethyl]quinolin-2(1 H)-one (WO2005/09286, 2.5Og, 5.12mmol) using the same method as described in preparation 7 to give a white solid, in 40% yield, 2.3g. LRMS: ESI m/z 909 [M+H]+
1H NMR (400 MHz, METHANOL-c/4) δ = -0.28 (s, 3H), -0.04 (s, 3H), 0.76 (s, 9H), 1.04-1.76 (m, 14H), 1.93 (m, 3H), 2.39 (m, 1 H), 2.53 (m, 2H), 2.64-2.79 (m, 5H), 2.79-2.94 (m, 3H), 2.99 (m, 2H), 4.07 (d, 2H), 5.15 (m, 1 H), 5.30 (s, 2H), 6.65 (m, 1 H), 7.06 (m, 4H), 7.15 (m, 3H), 7.20-7.43 (m, 5H), 7.49 (d, 2H), 7.59 (d, 2H), 8.32 (s, 1 H), 8.39 (d,1 H) ppm.
Preparation 15
5-r(1 R)-1-(rtert-butyl(dimethvnsilvnoxy)-2-(r2-(4-(2-r4-a3-r(RVcvclohexyl(hvdroxy) phenylmethyll-1H-
1 ,2,4-triazol-1-yl)methyl)piperidin-1-yl1ethyl)phenyl)ethyl1amino)ethyl1-8-hvdroxyquinolin-2(1 H)-one
Figure imgf000035_0002
The title compound was prepared from 8-(benzyloxy)-5-[(1 R)-1-{[tert-butyl(dimethyl)silyl]oxy}-2-{[2-(4-{2- [4-({3-[(R)-cyclohexyl(hydroxy)phenylmethyl]-1H-1 ,2,4-triazol-1-yl}methyl)piperidin-1- yl]ethyl}phenyl)ethyl]amino}ethyl]quinolin-2(1 H)-one (Preparation 14, 2.3Og, 2.81 mmol)_using the same method as described in preparation 8, to give a yellow solid, in 91 % yield, 2.1Og. LRMS: ESI m/z 819 [M+H]+
1H NMR (400 MHz, METHANOL-c/4) δ = -0.28 (s, 3H), -0.04 (s, 3H), 0.77 (s, 9H), 1.02-1.22 (m, 3H), 1.22-1.51 (m, 6H), 1.51-1.76 (m, 5H), 1.95 (m, 1 H), 2.08 (m, 2H), 2.39 (m, 1 H), 2.57 (m, 2H), 2.69 - 2.81 (m, 5H), 2.84 - 2.94 (m, 3H), 3.07 (m, 2H), 4.08 (d, 2H), 5.13 (m, 1 H), 6.62 (d, 1 H), 6.91 (d, 1 H), 6.99- 7.11 (m, 5H), 7.16 (m, 1 H), 7.25 (m, 2H), 7.59 (d, 2H), 8.32 (s, 1 H), 8.38 (d, 1 H) ppm.
Preparation 16 1-(pyrrolidin-1-ylmethyl)-1H-1 ,2,4-triazole
Figure imgf000036_0001
Triazole (1 Og, 144.8mnnol) and pyrrolidine (11.3g, 159.0mnnol) were dissolved ethanol (60ml) and formaldehyde (37% aqueous solution, 12.9ml, 159.0mmol) was added. After stirring at reflux for 4 hours reaction was allowed to stand at room temperature for 18 hours. The solvent was removed in vacuo, and the residue partitioned between dichloromethane (150ml) and water (8OmL) . The aqueous phase was separated and extracted with additional dichloromethane (80ml). The combined organic layers were washed with brine (100ml), dried over magnesium sulphate, filtered and the solvent removed in vacuo to furnish the title compound as a yellow oil, in 55% yield, 12.2g.
1H NMR (400 MHz, CHLOROFORM-c/) δ = 1.75 (m, 4H), 2.70 (m, 4H), 5.13 (s, 2H), 7.94 (s, 1 H), 8.13 (s, 1 H) ppm.
Preparation 17
Figure imgf000036_0002
1-(pyrrolidin-1-ylmethyl)-1H-1 ,2,4-triazole (Preparation 16, 12.Og, 78.84mmol) was dissolved in tetrahydrofuran (12OmL) and the solution cooled to -780C. n-Butyl lithium (2.5M in hexanes, 34.7ml, 86.7mmol) was then added dropwise over 30 mins. After warming to room temperature and stirring for 1 hour the reaction mixture was cooled to -780C and a solution of cyclohexyl(phenyl)methanone (16.3g, 86.7mmol) in tetrahydrofuran (3OmL) added dropwise. After warming to room temperature over 18 hr, water (100ml) was added and the solvent removed in vacuo. The residue was partitioned with ethyl acetate (200ml), the aqueous phase separated and re-extracted with ethyl acetate (2x 200ml). The combined organic layers were dried over magnesium sulphate, filtered and the solvent removed in vacuo to furnish the title compound as a yellow oil, in 100% yield, 27.28g.
1H NMR (400 MHz, CHLOROFORM-c/) δ = 1.00-1.94 (m, 14H), 2.49 (m 1 H), 2.71 (m, 4H), 5.08 (s, 2H), 7.19 (m, 1 H), 7.30 (m, 2H), 7.72 (m, 2H), 8.00 (s, 1 H) ppm.
Preparation 18
Cvclohexyl(phenyl)1 H-1 ,2,4-triazol-3-ylmethanol
Figure imgf000036_0003
CyclohexyKphenyOti^pyrrolidin-i-ylmethyO-IH-i ^^-triazol-S-yljmethanol (Preparation 17, 27.28g, 80.12mmol) was dissolved in ethanol (400ml) and sodium borohydride (3.03g, 80.1 mmol) added portionwise. After stirring at reflux for 3 hours the reaction was left to cool to room temperature over
18hrs. The solvent was removed in vacuo and the residue purified by column chromatography on silica gel eluting with pentane:ethyl acetate (3:1 to 0:1 , by volume) to furnish the title compound as a white foam, in 79% yield, 16.4g.
LRMS: APCI ESI m/z 256 [M-H]"
1H NMR (400 MHz, CHLOROFORM-c/) δ = 0.90-1.80 (m, 10H), 2.46 (m 1 H), 7.25 (m, 1 H), 7.33 (m, 2H),
7.67 (m, 2H), 8.00 (s, 1 H) ppm.
Preparation 19 Tert-butyl-4-({3-[cvclohexyl(hvdroxy)phenylmethyl1-1H-1 ,2,4-triazol-1-yl)methyl)piperidine-1-carboxylate
Figure imgf000037_0001
The title compound was prepared from cyclohexyl(phenyl)1 H-1 ,2,4-triazol-3-ylmethanol (Preparation 18,
500mg, 1.94mmol) and tert-butyl 4-(bromomethyl)piperidine-1-carboxylate (649mg, 2.33mmol) using the same method as described in preparation 3, to give a clear oil, in 47% yield, 415mg.
LRMS: APCI ESI m/z 381 [M+H]+
1H NMR (400 MHz, METHANOL-c/4) δ = 1.05-1.45 (m, 9H), 1.43 (s, 9H), 1.52 (m, 2H), 1.67 (m, 3H), 2.09
(m, 1 H), 2.38 (m, 1 H), 2.72 (m, 2H), 4.08 (m, 4H), 7.17 (m, 1 H), 7.26 (m, 2H), 7.58 (d, 2H), 8.30 (s, 1 H) ppm.
Preparation 20
CvclohexyKphenvDH -(piperidin-4-ylmethyl)-1 H-1 ,2,4-triazol-3-yl1methanol
Figure imgf000037_0002
The title compound was prepared from tert-butyl 4-({3-[cyclohexyl(hydroxy)phenylmethyl]-1H-1 ,2,4- triazol-1-yl}methyl)piperidine-1-carboxylate (Preparation 19, 450mg, 0.990mmol) using the same method as described in preparation 4 to give a clear oil, in 91 % yield, 320mg.
LRMS: APCI ESI m/z 378 [M+Na]+
1H NMR (400 MHz, METHANOL-c/4) δ = 1.04 -1.35 (m, 9H), 1.39-1.55 (m, 2H), 1.59-1.75 (m, 3H),
2.03(m, 1 H), 2.38 (m, 1 H), 2.5 (m, 2H), 3.01 (m, 2H), 4.05 (d, 2H), 7.15 (m, 1 H), 7.27 (m, 2H), 7.58 (d,
2H), 8.31 (s, 1 H) ppm.
Preparation 21 Di-tert-butyl {Q-K-dS-fcvclohexyKhvdroxy^phenylπnethyli-IH-I ^Λ-triazol-i-vDπnethvD-piperidin-i- ylinonvDimidodicarbonate
Figure imgf000038_0001
The title compound was prepared from cyclohexyl(phenyl)[1-(piperidin-4-ylmethyl)-1H-1 ,2,4-triazol-3- yl]methanol (Preparation 20, 300mg, 0.846mmol) and di-tert-butyl (9-bromononyl)imidodicarbonate (US04167167, 357mg, 0.846mmol) using the same method as described in preparation 5, to give a white foam, in 61 % yield, 360mg. LRMS: APCI ESI m/z 696 [M+H]+
1H NMR (400 MHz, METHANOL-c/4) δ = 1.14 (m, 4H), 1.23 - 1.37 (m, 18H) 1.46-1.60 (m, 24H) 1.68 (m, 2H) 1.96 (m, 2H) 2.33 (m, 2H) 2.95(m, 2H) 3.55 (m, 2H) 4.07 (m, 2H) 7.16 (m, 1 H) 7.26 (m, 2H) 7.58 (m, 2H) 8.31 (s, 1 H) ppm.
Preparation 22 (i-lfi-O-aminononvDpiperidin^-ylimethvD-IH-I ^Λ-triazol-S-ylϊcvclohexyDphenylmethanol
Figure imgf000038_0002
The title compound was prepared from di-tert-butyl {9-[4-({3-[cyclohexyl(hydroxy)phenyl methyl]-1H-
1 ,2,4-triazol-1-yl}methyl)piperidin-1-yl]nonyl}imidodicarbonate (Preparation 21 , 360mg, 0.57mmol) using the same method as described in preparation 6, to give a clear oil, in 90% yield, 230mg.
LRMS: APCI m/z 496 [M+H]+
1H NMR (400 MHz, METHANOL-c/4) δ = 1.12 (m, 4H), 1.22-1.39 (m, 18H), 1.39-1.58 (m, 6H), 1.64 (m,
2H), 1.94 (m, 2H), 2.31 (m, 2H), 2.65 (m, 2H), 2.93 (m, 2H), 4.06 (d, 2H), 7.16 (m, 1 H), 7.25 (m, 2H),
7.58 (d, 2H), 8.31 (s, 1 H) ppm.
Preparation 23
8-(benzyloxy)-5-[(1 R)-1-{[tert-butyl(dimethyl)silyl1oxy)-2-({9-[4-({3-[cvclohexyl(hvdroxy)phenyl methyli- 1H-1 ,2,4-triazol-1-yl)methyl)piperidin-1-yl1nonyl)-amino)ethyl1quinolin-2(1 H)-one
Figure imgf000039_0001
The title compound was prepared from (1-{[1-(9-aminononyl)piperidin-4-yl]methyl}-1H-1 ,2,4-triazol-3- yl)(cyclohexyl)phenylmethanol (Preparation 22, 150mg, 0.303mg) and 8-(benzyloxy)-5-[(1 R)-2-bromo-1- {[tert-butyl(dimethyl)silyl]oxy}ethyl]quinolin-2(1 H)-one (WO05/09286, 148mg, 0.303mmol) using the same method as described in preparation 7, to give a clear oil, in 24% yield, 66mg. LRMS: APCI m/z 904 [M+H]+
1H NMR (400 MHz, METHANOL-c/4) δ = 0.07 (s, 3H), 0.37 (s, 3H), 1.15 (s, 9H), 1.32-1.65(m, 22H), 1.76 (m, 6H), 1.92 (m, 2H), 2.20 (m, 2H), 2.58 (m, 2H), 2.86 (m, 2H), 3.01 (m, 1 H), 3.19 (m, 3H), 4.35 (m, 2H), 5.52 (m, 1 H), 5.61 (s, 2H), 6.95 (d, 1 H), 7.4-7.69 (m, 8H), 7.79 (m, 2H), 7.86 (m, 2H), 8.59 (s, 1 H), 8.74 (d, 1 H) ppm.
Preparation 24 δ-fd RVI-lftert-butvKdimethvnsilylloxyl^-gg-μ-αS-fcvclohexyKhvdroxy'lphenylmethyll-IH-I ^Λ-triazol-i- yl)methyl)piperidin-1-yl1nonyl)amino)ethyl1-8-hvdroxyquinolin-2(1 H)-one
The title compound was prepared from 8-(benzyloxy)-5-[(1 R)-1-{[tert-butyl(dimethyl)silyl]oxy}-2-({9-[4-({3- [cyclohexyl(hydroxy)phenylmethyl]-1H-1 ,2,4-triazol-1-yl}methyl)piperidin-1-yl]nonyl}amino)ethyl]quinolin- 2(1 H)-one (Preparation 23, 60mg, 0.066mmol) using the same method as described in preparation 8, to give a clear glass, in 70% yield, 38mg. LRMS: ESI m/z 813 [M+H]+
1H NMR (400 MHz, METHANOL-c/4) δ = 0.05 (s, 3H), 0.35 (s, 3H), 1.16 (s, 9H), 1.35- 1.62 (m, 22H), 1.68-1.85 (m, 6H), 1.85-2.02 (m, 2H), 2.22 (m, 2H), 2.63 (m, 2H), 2.90 (m, 2H), 3.04 (m, 1 H), 3.23 (m, 3H), 4.34 (m, 2H), 5.47 (m, 1 H), 6.87 (d, 1 H), 7.19 (d, 1 H), 7.42 (m, 2H), 7.53 (m, 2H), 7.85 (m, 2H), 8.58 (s, 1 H), 8.70 (d, 1 H) ppm.
Preparation 25
Benzyl 4-({3-[hvdroxy(diphenyl)methyl1-1H-1 ,2,4-triazol-1-yl)methyl)piperidine-1-carboxylate
Figure imgf000040_0001
Benzyl 4-(bromomethyl)piperidine-1-carboxylate (5g, 16mnnol) was dissolved in dimethylformannide (40ml) and diphenyl-(1 H-[1 ,2,4]triazol-3-yl)-methanol (Tetrahedron: Asymmetry, 8(9), 1491-1500; 1997; 3.35g, 13.65mmol) added, followed by potassium carbonate (3.69g, 26.7mmol). After stirring at 7O0C for 18 hours the solvent was removed in vacuo, and the residue partitioned between ethyl acetate (150ml) and water (15OmL). The organic layer was separated, dried over magnesium sulphate, filtered and the solvent removed in vacuo to furnish the title compound as a white solid in 81 % yield, 5.2g. 1H NMR (400 MHz, CHLOROFORM-c/) δ = 1.11 -1.40 (m, 5H), 1.50-1.77 (m, 2H), 2.75 (m, 2H), 4.20 (m, 2H), 5.09(m, 2H), 7.23-7.46 (m, 15H), 8.01 (s, 1 H) ppm.
Preparation 26
DiphenylH -(piperidin-4-ylmethyl)-1 H-1 ,2,4-triazol-3-yl1methanol
Figure imgf000040_0002
Benzyl 4-({3-[hydroxy(diphenyl)methyl]-1H-1 ,2,4-triazol-1-yl}methyl)piperidine-1-carboxylate (Preparation 25, 5.2g, 16.78mmol) was dissolved in ethanol (300ml) and ammonium formate (6g) and palladium hydroxide [20wt.%(dry basis) on carbon(wet)] (1g) added. After stirring at reflux for 1 hour the reaction mixture was cooled to room temperature and filtered through arbocel®. The solvent was removed in vacuo and the residue partitioned between dichloromethane (500ml) and water (50OmL). The aqueous layer was separated and then basified with aqueous sodium hydroxide solution (2N, 200ml) and extracted with dichloromethane (500ml). This organic layer was then dried over magnesium sulphate, filtered and the solvent removed in vacuo. The crude material was recrystallised from acetonitrile to furnish the title compound as a white solid, in 51 % yield, 1.9g.
1H NMR (400 MHz, METHANOL-c/4) δ = 1.56 (m, 2H), 1.86 (m, 2H), 2.35 (m, 1 H), 2.99 (m, 2H), 3.40 (m, 2H), 4.38 (m, 2H), 7.28-7.47 (m, 11 H) ppm.
Preparation 27
Di-tert-butyl {9-[4-({3-[hydroxy(diphenyl)methyl1-1H-1 ,2,4-triazol-1-yl)methyl)piperidin-1- ylinonvDimidodicarbonate
Figure imgf000041_0001
The title compound was prepared from diphenyl[1 -(piperidin-4-ylmethyl)-1H-1 ,2,4-triazol-3-yl]methanol
(Preparation 26, 2.0Og, 5.740mmol) and di-tert-butyl(9-bromononyl) imidodicarbonate (US04167167,
2.42g, 5.740mmol) using the same method as described in preparation 5, to give a clear oil, in 43% yield, 1.7g.
LRMS: APCI ESI m/z 691 [M+H]+
1H NMR (400 MHz, CHLOROFORM-c/) δ = 1.18-1.35 (m, 11 H), 1.41 (m, 3H), 1.49 (s, 18H), 1.57 (m,
4H), 1.94 (m, 3H), 2.38 (m, 2H) 2.96 (m, 2H) 3.54 (m, 2H), 4.00 (m, 2H) 7.22-7.48 (m, 10H), 7.99 (s, 1 H) ppm.
Preparation 28 (1-{[1-(9-aminononyl)piperidin-4-yl1methyl)-1H-1 ,2,4-triazol-3-yl)(diphenyl)methanol
Figure imgf000041_0002
Di-tert-butyl {9-[4-({3-[hydroxy(diphenyl)methyl]-1H-1 ,2,4-triazol-1-yl}methyl)piperidin-1-yl]nonyl} imidodicarbonate (Preparation 27, 1.7g, 2.46mmol) was dissolved ethanol (30ml) and hydrogen chloride in ether (2M, 30ml, 60mmol) added. After stirring at room temperature for 18 hours the solvent was removed in vacuo, and the residue partitioned between dichloromethane (200ml) and saturated aqueous sodium bicarbonate solution (200ml). The organic layer was separated, dried over magnesium sulphate, filtered and the solvent removed in vacuo. The residue was purified by column chromatography on silica gel eluting with dichloromethane:methanol:880 ammonia (90:10:1 to 80:20:2, by volume) to furnish the title compound as a white solid, in 60% yield, 725mg.
1H NMR (400 MHz, METHANOL-c/4) δ = 1.30 (m, 11 H), 1.40-1.62 (m, 7H), 1.93 (m, 3H), 2.28 (m, 2H), 2.61 (m, 2H), 2.94 (m, 2H), 4.09 (m, 2H), 7.20-7.36 (m, 10H), 8.36 (s, 1 H) ppm.
Preparation 29
8-(benzyloxy')-5-[(1 R)-1-([tert-butyl(dimethvnsilylloxy)-2-((9-[4-((3-[hvdroxy(diphenvnmethyll-1H-1 ,2,4- triazol-1-yl)methyl)piperidin-1-yl1nonyl)amino)ethyl1quinolin-2(1 H)-one
Figure imgf000042_0001
C-{[1-(9-aminononyl)piPeric'in-4-yl]ιπnethyl}-1H-1 ,2,4-triazol-3-yl)(diphenyl)methanol (Preparation 28, 710mg, 1.45mnnol) and 8-(benzyloxy)-5-[(1 R)-2-bromo-1-{[tert-butyl(dinnethyl)silyl]oxy}ethyl] quinolin- 2(1 H)-one (WO200509286, 708mg, 1.45mmol) were dissolved in dichloromethane (5ml) and dimethylsulphoxide (100μl) and diisopropylethylamine (253μl) added. After stirring in a sealed vessel at 950C for 48 hours the solvent was removed in vacuo. The residue was purified by column chromatography on silica gel eluting with dichloromethane:methanol:880 ammonia (90:10:1 , by volume) to furnish the title compound as a clear oil, in 38% yield, 500mg. LRMS: APCI m/z 898 [M+H]+
Preraration 30
5-[(1 R)-1-([tert-butyl(dimethvnsilylloxy)-2-((9-[4-((3-[hvdroxy(diphenyl)methyll-1H-1 ,2,4-triazol-1- yl)methyl)piperidin-1-yl1nonyl)amino)ethyl1-8-hvdroxyquinolin-2(1 H)-one
Figure imgf000042_0002
The title compound was prepared from 8-(benzyloxy)-5-[(1 R)-1-{[tert-butyl(dimethyl)silyl]oxy}-2-({9-[4-({3- [hydroxy(diphenyl)methyl]-1H-1 ,2,4-triazol-1-yl}methyl)piperidin-1-yl]nonyl}amino)ethyl] quinolin-2(1 H)- one (Preparation 29, 500mg, 0.056mmol) using the same method as described in preparation 8, to give a yellow glass, in 59% yield, 267mg. LRMS: ESI m/z 807 [M+H]+
1H NMR (400 MHz, METHANOL-c/4) δ = 0.00 (s, 3H), 0.20(s, 3H), 1.00 (s, 9H), 1.06-1.82 (m, 18H), 2.19 (m, 3H), 2.55 (m, 2H), 2.63 (m, 1 H), 2.73 (m, 1 H ), 3.05 (m, 1 H ), 3.13 (m, 1 H), 3.25 (m, 2H), 4.17 (m, 2H), 5.22 (m, 1 H), 6.78 (m, 2H), 7.13-7.48 (m, 7H), 7.58 (m, 4H), 8.17 (m, 1 H), 8.50 (m, 1 H) ppm.
Preparation 31
2-[2-(4-{2-[4-({3-[hvdroxy(diphenyl)methyl1-1H-1 ,2,4-triazol-1-yl)methyl)piperidin-1-yl1ethyl)phenyl)ethyl1- 1 H-isoindole-1 ,3(2H)-dione
Figure imgf000043_0001
The title compound was prepared from diphenyl[1-(piperidin-4-ylmethyl)-1H-1 ,2,4-triazol-3-yl]methanol (Preparation 26, 778mg, 2.23mmol) and 2-{2-[4-(2-bromoethyl)phenyl]ethyl}-1H-isoindole-1 ,3(2H)-dione (Preparation 1 1 , 800mg, 2.23mmol) using the same method as described in preparation 12, to give a white solid, in 71 % yield, 1.0Og. LRMS: ESI m/z 627 [M+H]+
1H NMR (400 MHz, METHANOL-c/4) δ = 1.24-2.03 (m, 7H), 2.54 (m, 2H), 2.75 (m, 2H), 2.97 (m, 4H), 3.88 (m, 2H), 3.99 (m, 2H), 7.10 (m, 2H), 7.20 (m, 2H), 7.29 (m, 7H), 7.44 (m, 3H), 7.70 (m, 2H), 7.82 (m, 2H), 7.99 (s, 1 H) ppm.
Preparation 32 {1-[(1-{2-[4-(2-aminoethyl)phenyl1ethyl)piperidin-4-yl)methyl1-1H-1 ,2,4-triazol-3-yl)(diphenyl)methanol
Figure imgf000043_0002
The title compound was prepared from 2-[2-(4-{2-[4-({3-[hydroxy(diphenyl)methyl]-1H-1 ,2,4-triazol-1- yl}methyl)piperidin-1-yl]ethyl}phenyl)ethyl]-1H-isoindole-1 ,3(2H)-dione (Preparation 31 , 1.0g, 1.59mmol) using the same method as described in preparation 13, to give a colourless oil, in 75% yield, 790mg. LRMS: ESI m/z 497 [M+H]+
1H NMR (400 MHz, METHANOL-c/4) δ = 1.37 (m, 2H) 1.58 (m, 2H) 1.93 (m, 1 H) 2.05 (m, 2H) 2.54 (m, 2H) 2.73 (m, 4H) 2.83 (m, 2H) 3.02 (m, 2H) 4.09 (d, 2H) 7.13 (m, 4H) 7.26 (m, 6H) 7.33 (m, 4H), 8.38 (s, 1 H) ppm.
Preparation 33
8-(benzyloxy')-5-[(1 R)-1-([tert-butyl(dimethvnsilylloxy)-2-([2-(4-(2-[4-((3-[hvdroxy(diphenvn methyll-1H-
1 ,2,4-triazol-1-yl)methyl)piperidin-1-yl1ethyl)phenyl)ethyl1-amino)ethyl1quinolin-2(1 H)-one Si
Figure imgf000044_0001
The title compound was prepared from {1-[(1-{2-[4-(2-aminoethyl)phenyl]ethyl}piperidin-4-yl)methyl]-1H- 1 ,2,4-triazol-3-yl}(diphenyl)methanol (Preparation 32, 590mg, 1.2mmol) and 8-(benzyloxy)-5-[(1 R)-2- bromo-1-{[tert-butyl(dimethyl)silyl]oxy}ethyl]quinolin-2(1 H)-one (WO2005/09286, 581 mg, 1.19mmol) using the same method as described in preparation 7, to give a white solid, in 67% yield, 740mg. LRMS: ESI m/z 903 [M+H]+
1H NMR (400 MHz, METHANOL-c/4) δ = -0.28 (s, 3H), -0.04 (s, 3H), 0.76 (s, 9H), 1.38 (m, 2H), 1.59 (m, 2H), 1.92 (m, 1 H), 2.05 (m, 2H), 2.54 (m, 2H), 2.69-2.94 (m, 8H), 3.03 (m, 2H), 4.11 (m, 2H), 5.15 (m, 1 H), 5.32 (s, 2H), 6.65 (d, 1 H), 7.08 (m, 4H), 7.16 (m, 2H), 7.27 (m, 6H), 7.35 (m, 7H), 7.50 (m, 2H), 8.40 (s,2H) ppm.
Preparation 34
5-[(1 R)-1-([tert-butyl(dimethvnsilylloxy)-2-([2-(4-(2-[4-((3-[hvdroxy(diphenyl)methyll-1H-1 ,2,4-triazol-1- yl)methyl)piperidin-1-yl1ethyl)phenyl)ethyl1amino)ethyl1-8-hvdroxyquinolin-2(1 H)-one
^Si
Figure imgf000044_0002
8-(benzyloxy)-5-[(1 R)-1-{[tert-butyl(dimethyl)silyl]oxy}-2-{[2-(4-{2-[4-({3-[hydroxy(diphenyl) methyl]-1H- 1 ,2,4-triazol-1-yl}methyl)piperidin-1-yl]ethyl}phenyl)ethyl]amino}ethyl]quinolin-2(1 H)-one (Preparation 33, 740mg, 0.82mmol), palladium hydroxide [20wt.%(dry basis) on carbon(wet)] (23mg, 0.164mmol) and methylcyclohexyldiene (231 mg, 2.46mmol) were dissolved in MeOH (20ml). After stirring at 5O0C for 18 hours, additional methylcyclohexyldiene (115.5mg, 1.23mmol) was added and stirring continued at 5O0C for a further 18 hours. The reaction mixture was allowed to cool, filtered through arbocel® and the solvent removed in vacuo to furnish the titile compound as yellow solid, in 87% yield, 580mg. LRMS: ESI m/z 814 [M+H]+
1H NMR (400 MHz, METHANOL-c/4) δ = -0.28 (s, 3H), -0.04 (s, 3H), 0.77 (s, 9H), 1.38(m, 2H), 1.60 (m, 2H), 1.92 (m, 1 H), 2.07 (m, 2H), 2.57 (m, 2H), 2.70-2.96 (m, 8H), 3.05 (m, 2H), 4.10 (m, 2H), 5.14 (m, 1 H), 6.59 (d, 1 H), 6.92 (d, 1 H), 7.08 (m, 4H), 7.15 (m, 1 H), 7.25 (m, 6H), 7.35 (m, 4H), 8.33 (d, 1 H), 8.40 (s, 1 H) ppm.
Preparation 35
Methanesulfonic acid 2-{4-[2-(1 ,3-dioxo-1 ,3-dihvdro-isoindol-2-yl)-ethyl1-phenyl)-ethyl ester
Figure imgf000045_0001
A solution of 2-{2-[4-(2-hydroxy-ethyl)phenyl]-ethyl}-isoindole-1 ,3-dione (Preparation 10, 21.Og, 71.25mnnol) in methyl ethyl ketone (315ml_) was cooled to O0C. Triethylamine (14.9ml_, 106.87mnnol) was added followed by the dropwise addition of methane sulfonyl chloride (6.07ml_, 78.37mmol). The reaction was stirred at 0-40C for 1 hour and then allowed to warm to room temperature. An aqueous solution of potassium carbonate (1 M, 21OmL) was added and the mixture stirred. The organic and aqueous layers were separated and the aqueous re-extracted with methyl ethyl ketone (5OmL). The combined organic layers were concentrated to 4OmL by distillation. Tert-butyl methyl ether (30OmL) was slowly added to the hot solution causing crystallisation to occur. The mixture was allowed to cool to room temperature and then allowed to stir for 72 hours. The solid was removed by filtration and washed with tert-butyl methyl ether (3x 25mL). The solid was dried in vacuo at 450C for 4 hours to yield the title compound as a light brown solid, in 92% yield, 24.6g.
1H NMR (400MHz, CDCI3) δ = 2.82 (s, 3H), 2.96-3.02 (m, 4H), 3.89-3.93 (t, 2H), 4.37-4.40 (t, 2H), 7.13- 7.23 (2xd, 4H), 7.70-7.72 (dd, 2H), 7.81-7.83 (dd, 2H) ppm.
Preparation 36 (2R)-Cvclohexyl-hvdroxy-phenyl-acetic acid
Figure imgf000045_0002
To a slurry of D-tyrosine methyl ester (90.Og, 460mmol) in acetonitrile (810ml) and water (65ml) was added racemic 2-cyclohexyl-2-hydroxy-2-phenylacetic acid (216.Og, 921 mmol). The mixture was heated to reflux for 1 hour and then allowed to cool to room temperature overnight. The mixture was further cooled to 40C for 4 hours and then filtered, washing with cold acetonitrile (400ml) to yield a white solid (169.9g). This solid was partitioned between methyl tert-butyl ether (1.35L) and 0.5M aqueous hydrochloric acid (765ml, 1 eq). The organic layer was separated and washed with 0.5M aqueous hydrochloric acid (380ml, 0.5eq), dried over sodium sulphate, filtered and the solvent removed in vacuo to furnish the title compound as a white solid, 84.5g. LRMS: APCI ESI m/z 233 [M-H]"
1H NMR (400 MHz, DMSO-c/6) δ = 0.95-1.14 (m, 4H), 1.19-1.29 (m, 1 H), 1.32-1.42 (m, 1 H), 1.49-1.63 (m, 3H), 1.69-1.78 (m, 1 H), 2.11-2.19 (m, 1 H), 7.21-7.25 (m, 1 H), 7.30-7.34 (m, 2H), 7.57-7.60 (m, 2H) ppm. EXAMPLES Example 1 δ-fd R^-αQ-K-αS-^RVcvclohexyKhvdroxy^phenylmethyli-IH-I ^Λ-triazol-i-yllmethvDpiperidin-i- yl1nonyl)amino)-1-hvdroxyethyl1-8-hvdroxyquinolin-2(1 H)-one
Figure imgf000046_0001
5-[(1 R)-1-{[tert-butyl(dimethyl)silyl]oxy}-2-({9-[4-({3-[(R)-cyclohexyl(hydroxy)phenylmethyl]-1H-1 ,2,4- triazol-1-yl}nnethyl)piperidin-1-yl]nonyl}annino)ethyl]-8-hydroxyquinolin-2(1 H)-one (Preparation 8, 13.06g, 16.06mnnol) was dissolved in methanol (430ml) and ammonium fluoride (2.97g, 80.3mmol) was added. After stirring at 4O0C for 18 hours the solvent was removed in vacuo and the residue partitioned between dichloromethane/methanol (950ml/50ml) and saturated aqueous sodium bicarbonate solution (50OmL). The organic layer was separated and the aqueous layer further extracted with dichloromethane/methanol (450ml/50ml). The combined organic layers were dried over magnesium sulphate, filtered and the solvent removed in vacuo. The residue was purified by column chromatography on silica gel eluting with dichloromethane:methanol:880 ammonia (90:10:1 , by volume) to furnish the title compound as a yellow foam, in 72.3% yield, 8.12g. LRMS: ESI m/z 699 [M+H]+
1H NMR (400 MHz, METHANOL-c/4) δ =: 1.05-1.77 (m, 28H), 1.98 (m, 3H), 2.35 (m, 3H) 2.75 (m, 2H), 2.95 (m, 4H), 4.09 (m, 2H), 5.24 (m, 1 H), 6.65 (d, 1 H), 6.95 (d, 1 H), 7.15-7.27 (m, 4H), 7.60 (m, 2H), 8.33 (s, 1 H), 8.38 (d, 1 H) ppm.
Example 1a
5-[(1 R)-2-({9-[4-({3-[(R)-cvclohexyl(hvdroxy)phenylmethyl1-1H-1 ,2,4-triazol-1-yl)methyl)piperidin-1- yl1nonyl)amino)-1-hvdroxyethyl1-8-hvdroxyquinolin-2(1 H)-one; naphthalene-1 ,5-disulfonate salt
Figure imgf000046_0002
5-[(1 R)-2-({9-[4-({3-[(R)-cyclohexyl(hydroxy)phenylmethyl]-1H-1 ,2,4-triazol-1yl}methyl) piperidin-1- yl]nonyl}amino)-1-hydroxyethyl]-8-hydroxyquinolin-2(1 H)-one (Example 1 , 2.Og, 2.86mmol) was dissolved in methanol (15ml) and a solution of naphthalene-1 ,5-disulfonic acid (825. Omg, 2.86mmol) in water (3ml) added dropwise. The mixture was stirred at room temperature overnight. The resulting solid was removed by filtration and dried in vacuo to yield 2.03g of white solid. 50mg of this material was treated with methanokwater (70:30 by volume, 1ml) and the resulting suspension heated to 7O0C and then slow cooled to 2O0C over 24 hours. The solid was recovered by filtration, washing with water and dried in vacuo to yield the title compound as a crystalline white solid, 19mg.
1H NMR (400 MHz, DMSO-d) δ =: 0.95-1.70 (m, 28H), 1.96-2.29 (m, 3H), 2.71-3.13 (m, 7H), 3.42 (m, 2H), 4.07 (m, 2H), 5.30 (m, 1H), 6.58 (d, 1H), 6.95 (d, 1H), 7.13 (m, 2H), 7.23 (m, 2H), 7.41 (m, 2H), 7.56 (m, 2H), 7.92 (m, 2H), 8.17 (d, 1 H), 8.42 (s, 1H ), 8.89 (d, 2H) ppm.
Example 2
5-r(1RV2-(r2-(4-(2-r4-((3-r(RVcvclohexyl(hvdroxybhenylmethyll-1H-1.2.4-triazol-1-yl)methylbiperidin-1- yllethyl)phenyl)ethyllamino)-1-hvdroxyethyll-8-hvdroxyαuinolin-2(1H)-one
Figure imgf000047_0001
The title compound was prepared from 5-[(1R)-1-{[tert-butyl(dimethyl)silyl]oxy}-2-{[2-(4-{2-[4-({3-[(R)- cyclohexyKhydroxyJphenylmethyll-IH-I ^Λ-triazol-i-ylJmethylJpiperidin-i-yllethyl} phenyl)ethyl]amino}ethyl]-8-hydroxyquinolin-2(1H)-one (Preparation 15, 2.3Og, 2.81 mmol) using the same method as described in example 1 , to give a yellow solid, in 77% yield, 1.4Og. LRMS: ESI m/z 705 [M+H]+
1H NMR (400 MHz, METHANOL-^) δ= 1.08-1.48 (m, 9H), 1.48-1.81 (m, 5H), 1.94 (m, 1H), 2.08 (m, 2H), 2.38 (m, 1 H), 2.54 (m, 2H), 2.66-2.97 (m, 8H), 3.04 (m, 2H), 4.07 (d, 2H), 5.18 (m, 1 H), 6.61 (d, 1H), 6.92 (d, 1H), 7.07 (m, 4H), 7.15 (m, 2H), 7.25 (m, 2H), 7.56 (d, 2H), 8.31 (m, 2H) ppm.
Example 2a
5-r(1RV2-(r2-(4-(2-r4-((3-r(RVcvclohexyl(hvdroxybhenylmethyll-1H-1.2.4-triazol-1-yl)methylbiperidin-1- yllethyl}phenyl)ethyllamino}-1-hvdroxyethyll-8-hvdroxyαuinolin-2(1H)-one: naphthalene- 1 ,5-disulfonate salt
Figure imgf000047_0002
5-[(1R)-2-{[2-(4-{2-[4-({3-[(R)-cyclohexyl(hydroxy)phenylmethyl]-1H-1 ,2,4-triazol-1-yl}methyl)piperidin-1- yl]ethyl}phenyl)ethyl]amino}-1-hydroxyethyl]-8-hydroxyquinolin-2(1H)-one (Example 2, 935 mg, 1.33
46 mmol) was dissolved in methanol (10 ml_). A solution of 1 ,5-naphthalenedisulphonic acid tetrahydrate (478 mg, 1.33 mmol) in methanol (5 ml_) was added and the precipitate that formed was redissolved by adding water (5 ml_) and heating. The resulting solution was evaporated in vacuo, re-suspended in methanol (50 ml_) and heated to reflux for 30 seconds. The supernatant was decanted from the residue and the residue re-suspended in methanol (50 ml_) and heated at reflux with stirring for 4 days. The resulting precipitate was collected by filtration and dried under vacuum to provide the title compound as a colourless crystalline solid, 990 mg.
1H NMR (400 MHz, DMSO-c/6) δ = 0.94-1.49 (m, 8H), 1.50-1.74 (m, 6H), 1.99-2.12 (m, 1 H), 2.19-2.27 (m, 1 H), 2.83-3.01 (m, 6H), 3.04-3.24 (m, 6H), 3.49-3.59 (m, 2H), 4.05-4.11 (d , 2H), 5.07 (s, 1 H), 5.30- 5.36 (m, 1 H), 6.14-6.19 (d, 1 H), 6.57-6.59 (d, 1 H), 6.97-6.99 (d, 1 H), 7.12-7.19 (m, 6H), 7.24-7.28 (t, 2H), 7.39-7.43 (t, 2H), 7.56-7.58 (d, 2H), 7.94-7.96 (d, 2H), 8.15-8.18 (d, 1 H), 8.45 (s, 1 H), 8.59-8.74 ( bm, 2H), 8.88-8.90 (d, 2H), 9.04-9.19 (bm, 1 H), 10.41 (s, 1 H), 10.45 (s, 1 H) ppm.
The crystalline form produced by the process described above (Form 1 ) also has the characteristics shown in the corresponding Powder X-ray diffraction pattern of Figure 1/1. The main characteristic peaks are at 12.7, 18.1 , 21.0, 22.2 and 23.6 degrees 2-theta ± 0.1 degrees 2-theta and are further given in table 1 below.
Table 1 - Charateristic PXRD peaks for Example 2a
Figure imgf000048_0002
Example 2b δ-fd R^-l^^^-μ-αS-^RVcvclohexyKhvdroxy'lphenylmethyll-IH-I ^Λ-triazol-i-vDmethvnpiperidin-i- yl1ethyl)phenyl)ethyl1amino)-1-hvdroxyethyl1-8-hvdroxyquinolin-2(1 H)-one; succinate salt
Figure imgf000048_0001
5-[(1 R)-2-{[2-(4-{2-[4-({3-[(R)-cyclohexyl(hydroxy)phenylmethyl]-1H-1 ,2,4-triazol-1-yl}methyl)piperidin-1- yl]ethyl}phenyl)ethyl]amino}-1-hydroxyethyl]-8-hydroxyquinolin-2(1 H)-one (Example 2, 50mg, 0.07 mmol) was dissolved in methanol (4 ml_). A solution of succinic acid (8.4 mg, 0.07 mmol) in methanol (1 ml_) was added and the resulting cloudy solution allowed to evaporate on standing overnight. The resulting gum was redissolved in ethanol (2.5 ml_) and water (2.5 ml_) with heating and then allowed to cool to room temperature with stirring overnight. The resulting precipitate was collected by filtration and dried under vacuum to provide the title compound as a colourless crystalline solid, 28 mg. 1H NMR (400 MHz, DMSO-c/6) δ = 0.97-1.85 (m, 15H), 1.88-1.93 (m, 2H), 2.19-2.25 (m, 1 H), 2.34 (s, 4H), 2.44-2.47 (m, 2H), 2.64-2.68 (m, 2H), 2.71-2.75 (m, 2H), 2.84-2.93 (m, 6H), 4.02 (d, 2H), 5.10-5.13 (m, 1 H), 6.52 (d, 1 H), 6.93 (d, 1 H), 7.07-7.16 (m, 6H), 7.25 (t, 2H), 7.56 (d, 2H), 8.15 (d, 1 H), 8.38 (s, 1 H) ppm.
Example 2c δ-fd R^-l^^^-μ-αS-^RVcvclohexyKhvdroxy'lphenylmethyll-IH-I ^Λ-triazol-i-vDmethvnpiperidin-i- yl1ethyl)phenyl)ethyl1amino)-1-hvdroxyethyl1-8-hvdroxyquinolin-2(1 H)-one; fumarate salt
Figure imgf000049_0001
5-[(1 R)-2-{[2-(4-{2-[4-({3-[(R)-cyclohexyl(hydroxy)phenylmethyl]-1H-1 ,2,4-triazol-1-yl}methyl)piperidin-1- yl]ethyl}phenyl)ethyl]amino}-1-hydroxyethyl]-8-hydroxyquinolin-2(1 H)-one (Example 2, 50mg, 0.07 mmol) was dissolved in methanol (4 ml_). A solution of fumaric acid (8.2 mg, 0.07 mmol) in methanol ( 1 ml_) was added and the resulting cloudy solution allowed to evaporate on standing overnight. The resulting gum was redissolved in ethanol (2.5 ml_) and water (2.5 ml_) with heating and then allowed to cool to room temperature with stirring overnight. The resulting precipitate was collected by filtration and dried under vacuum to provide the title compound as a colourless crystalline solid, 38mg. 1H NMR (400 MHz, DMSO-c/6) δ = 0.99-1.84 (m, 15H), 1.91-1.98 (m, 2H), 2.18-2.24 (m, 1 H), 2.46-2.49 (m, 2H), 2.65-2.69 (m, 2H), 2.78-2.82 (m, 2H), 2.90-3.02 (m, 6H), 4.03 (d, 2H), 5.19-5.22 (m, 1 H), 6.51 (m, 3H), 6.95 (d, 1 H), 7.09-7.16 (m, 6H), 7.23-7.27 (m, 2H), 7.56 (d, 2H), 8.18 (d, 1 H), 8.39 (s, 1 H) ppm.
Example 3
5-[(1 R)-2-({9-[4-({3-[cvclohexyl(hvdroxy)phenylmethyl1-1H-1 ,2,4-triazol-1-yl)methyl)piperidin-1- yl1nonyl)amino)-1-hvdroxyethyl1-8-hvdroxyquinolin-2(1 H)-one
Figure imgf000050_0001
The title compound was prepared from 5-[(1 R)-1-{[tert-butyl(dimethyl)silyl]oxy}-2-({9-[4-({3- [cyclohexyl(hydroxy)phenylmethyl]-1H-1 ,2,4-triazol-1-yl}methyl)piperidin-1-yl]nonyl}amino) ethyl]-8- hydroxyquinolin-2(1 H)-one (Preparation 24, 18mg, 0.022mmol) using the same method as described in example 1 , to give a clear glass, in 65% yield, 10mg. LRMS:APCI ESI m/z 699 [M+H]+
1H NMR (400 MHz, METHANOL-c/4) δ =: 1.07-1.76 (m, 30H), 1.98 (m, 2H), 2.36 (m, 2H), 2.75 (m, 2H), 2.95 (m, 4H), 4.07 (m, 2H), 5.24 (m, 1 H), 6.65 (d, 1 H), 6.95 (d, 1 H), 7.12-7.28 (m, 4H), 7.58 (m, 2H), 8.31 (s, 1 H), 8.37 (d, 1 H) ppm.
Example 4
8-hvdroxy-5-[(1 R)-1-hvdroxy-2-({9-[4-({3-[hvdroxy(diphenyl)methyl1-1H-1 ,2,4-triazol-1-yl)methyl)piperidin- 1-yl1nonyl)amino)ethyl1quinolin-2(1 H)-one
Figure imgf000050_0002
The title compound was prepared from 5-[(1 R)-1-{[tert-butyl(dimethyl)silyl]oxy}-2-({9-[4-({3- [hydroxy(diphenyl)methyl]-1H-1 ,2,4-triazol-1-yl}methyl)piperidin-1-yl]nonyl}amino)ethyl]-8- hydroxyquinolin-2(1 H)-one (Preparation 30, 260mg, 0.322mmol) using the same method as described in Example 1 , to give a yellow glass, in 49% yield, 110mg. LRMS:APCI ESI m/z 693 [M+H]+
1H NMR (400 MHz, METHANOL-c/4) δ =: 1.24-1.39 (m, 12H), 1.42-1.62 (m, 6H), 1.87-2.03 (m, 3H), 2.34 (m, 2H), 2.74 (m, 2H), 2.93 (m, 4H), 4.07 (m, 2H), 5.24 (m, 1 H), 6.63 (d, 1 H), 6.95 (d, 1 H), 7.17-7.30 (m, 7H), 7.30-7.37 (m, 4H), 8.35 (m, 2H) ppm.
Example 5
8-hvdroxy-5-[(1 R)-1-hvdroxy-2-([2-(4-(2-[4-((3-[hvdroxy(diphenyl)methyll-1H-1 ,2,4-triazol-1- yl)methyl)piperidin-1-yl1ethyl)phenyl)ethyl1amino)ethyl1quinolin-2(1 H)-one
Figure imgf000051_0001
The title compound was prepared from 5-[(1 R)-1-{[tert-butyl(dimethyl)silyl]oxy}-2-{[2-(4-{2-[4-({3- [hydroxy(diphenyl)methyl]-1H-1 ,2,4-triazol-1-yl}methyl)piperidin-1-yl]ethyl}phenyl)ethyl] amino}ethyl]-8-hydroxyquinolin-2(1 H)-one (Preparation 34, 580mg, 0.71 mmol) using the same method as described in example 1 , to give a yellow solid, in 60% yield, 300mg. LRMS:APCI ESI m/z 699 [M+H]+
1H NMR (400 MHz, METHANOL-c/4) δ = 1.37(m, 2H), 1.58 (m, 2H), 1.91 (m, 1 H), 2.07 (m, 2H), 2.54 (m, 2H), 2.82 (m, 8H), 3.04 (m, 2H), 4.11 (m, 2H), 5.18 (m, 1 H), 6.62 (d, 1 H), 6.92 (d, 1 H), 7.05 (m, 4H), 7.15 (m, 1 H), 7.25 (m, 6H), 7.34 (m, 4H), 8.33 (d, 1 H), 8.38 (s, 1 H) ppm.
Binding affinity assessment at the human recombinant M^ muscarinic receptor
Membrane preparation
Cell Pellets from CHO (Chinese Hamster Ovary) cells recombinantly expressing the human muscarinic M3 receptor were homogenised in 2OmM HEPES (pH7.4) and centrifuged at 48000 x g for 20min at 4°C. The pellet was re-suspended in buffer and the homogenisation and centrifugation steps repeated. The resulting pellet was re-suspended in 1 ml buffer per 1 ml original packed cell volume and the homogenisation step repeated. Protein estimation was carried out on the suspension and 1 ml aliquots of ~1 mg/ml frozen at -8O0C.
hM3 competition binding Assay Protocol
Membranes (5μg/well) were incubated with 3H-NMS (at a concentration 5 x KD) plus/minus test compound for 24hr at RT (room temperature) in a 1 ml polystyrene 96-well deep well block. The final assay volume was 200μl, comprising of: 20μl plus/minus test compound; 20μl 3H-NMS (Perkin Elmer
NEN 636) and 160μl membrane solution. Total Binding was defined with 0.1 % DMSO; Non-Specific
Binding was defined with 1μM Atropine. Assay buffer was 2OmM Hepes (pH 7.4).
Once all assay components were added, plates were covered and incubated at room temperature for 24 hrs with shaking. The assay was terminated by rapidly filtering through GF/B Unifilter plates pre-soaked with 0.5% polyethylenimine, using a Packard filtermate harvester, the filter plate was then washed with
3x1 ml 40C assay buffer. The filter plates were dried at 45°C for 1 hour. The bottoms of the filter plates were sealed and 50μl/well of Microscint '0' added, the top of the plates were sealed with a Topseal.
Following 90mins, the plates were read on an NXT Topcount (1 minute read time per well).
The resulting data was expressed as a percentage of the specific binding (Specific binding = Total binding - Non-Specific Binding). % specific binding versus test compound concentration was plotted to determine an IC50 from a sigmoid curve using an in-house data analysis programme. IC50 values corrected to Ki values by applying the Cheng-Prussoff equation:
Cheng-Prussoff equation: IC 50
K1 =
1 + [L]/KD where IC50 is the concentration of unlabelled drug which inhibits by 50% the specific radioligand binding. [L] is the free radioligand concentrations and KD and K, are the equilibrium dissociation constants of the radioligand and unlabelled drug respectively.
Functional assessment of agonist potency and efficacy using a whole cell cAMP assay in CHO cells expressing the hβ? receptor
Cell Culture
CHO (Chinese Hamster Ovary) cells recombinantly expressing the human adrenergic β2 receptor were maintained in growth media composed of F12:DMEM (Gibco 21331-020) containing 10% Foetal Bovine Serum (FBS: Sigma F4135), 10μg/ml puromycin (Sigma P8833), 0.1 mg/ml Geneticin G418 (Sigma G7034) and 2mM L-glutamine (Sigma G7513). The cells were kept in sterile conditions at 37°C, in an atmosphere containing 5% CO2.
hβ? cAMP Assay Protocol
Cells were harvested for assay when they reached 80-90% confluency using trypsin 0.25% (Sigma T4049) incubated with the cells for 5 min at 37°C in an atmosphere containing 5% CO2. Detached cells were collected in warmed growth media (composition described above), and re-suspended in assay media (F12:DMEM (Gibco 21331-020) containing 1 % Foetal Bovine Serum (FBS: Sigma F4135) and 2mM L-glutamine (Sigma G7513)) to give a viable cell concentration of 0.25x106 cells/ml. 100μl of this suspension was added to each well of a tissue culture treated 96 well view-plate (PerkinElmer 6005181 ) and the plate incubated in an atmosphere containing 5% CO2 at 37°C overnight. The following day, cells were washed (4 x 200μl, Skatron Skanstacker 300) with Phosphate Buffered Saline (PBS). The plates were then pat dried and the media replaced with 50μl PBS plus 0.5mM isobutylmethylxanthine (IBMX) (Sigma I5879) before returning to the incubator for 30mins. The PBS plate wash procedure was repeated, followed by the addition of 50μl PBS plus 0.5mM IBMX and 50μl PBS plus 2% DMSO and the plates returned to the incubator for 30mins. Concentration ranges of test compounds were prepared in PBS containing 2% DMSO. The PBS plate wash procedure was repeated, and then 50μl PBS plus 0.5mM IBMX was added to the assay plates, followed by the addition of 50μl of each compound test concentration to the appropriate well. 50μl per well of 2% DMSO or 20OnM Formoterol was added to the control wells, giving a final assay concentration of 1 % DMSO or 10OnM Formoterol. Plates were returned to the incubator for a further 30mins. At the end of the incubation period the PBS plate wash procedure was repeated a final time as described above. The plates were pat dried and 30μl per well PBS added followed by 40μl per well cAMP Il ED/Substrate mix and 40μl per well cAMP Il EA-Ab/lysis mix (DiscoverX 90-0034-03). The plates were then incubated in the dark for a minimum of 4 hours at room temperature. The plates were read using a Fusion plate reader (luminescence protocol, each well read for 1 second). Concentration effect curves were plotted and EC50 & Emax values determined using a A- parameter sigmoid fit generated using an in-house data analysis programme. Formoterol and salmeterol were run in every assay as reference standards. It has thus been found that compounds of formula (1 ) according to the present invention that have been tested in the above assays show hβ2 receptor agonist activity and hM3 receptor antagonist activity as listed in the table below:
Figure imgf000053_0001

Claims

1. A compound of general formula (1 ),
Figure imgf000054_0001
wherein:
A is selected from:
H
Figure imgf000054_0002
and
Figure imgf000054_0003
wherein * represent the attachment point of A to the carbon bearing the hydroxy;
X is -(CH2)m- where m is an integer comprised between 7 to 12 inclusive, or is of formula:
Figure imgf000054_0004
Y is selected from
Figure imgf000054_0005
wherein ** and *** represent the attachment points, ** being linked to X; n is 0 or 1 ;
R1 is selected from cyclopentyl, cyclohexyl, phenyl, furanyl and thiophenyl; and,
R2 is selected from phenyl, furanyl and thiophenyl; or the pharmaceutically acceptable salts thereof, or the pharmaceutically acceptable solvates of said compounds or salts.
2. A compound according to claim 1 , or a pharmaceutically acceptable salt thereof or a pharmaceutically acceptable solvate of said compound or salt, wherein A is of formula:
Figure imgf000054_0006
wherein * represent the attachment point of A to the carbon bearing the hydroxyl.
3. A compound according to claim 1 or 2, or a pharmaceutically acceptable salt thereof or a pharmaceutically acceptable solvate of said compound or salt, wherein X is -(CH2^- or is of formula:
4. A compound according to any one of claims 1 to 3, or a pharmaceutically acceptable salt thereof or a pharmaceutically acceptable solvate of said compound or salt, wherein X is of formula:
Figure imgf000055_0001
wherein ** and *** represent the attachment points, ** being linked to X.
5. A compound according to any one of claims 1 to 4, or a pharmaceutically acceptable salt thereof or a pharmaceutically acceptable solvate of said compound or salt, wherein R1 is phenyl or cyclohexyl.
6. A compound according to any one of claims 1 to 5, or a pharmaceutically acceptable salt thereof or a pharmaceutically acceptable solvate of said compound or salt, wherein R2 is phenyl.
7. A compound according to any one of claims 1 to 6, or a pharmaceutically acceptable salt thereof or a pharmaceutically acceptable solvate of said compound or salt, wherein n is 1.
8. A compound according to claim 1 , or a pharmaceutically acceptable salt thereof or a pharmaceutically acceptable solvate of said compound or salt, wherein:
A is of formula:
Figure imgf000055_0002
wherein * represent the attachment point of A to the carbon bearing the hydroxy; X is -(CH2)9- or is of formula:
Figure imgf000055_0003
Y is of formula:
Figure imgf000055_0004
wherein ** and *** represent the attachment points, ** being linked to X; n is 1 ;
R1 is cyclohexyl or phenyl and R2 is phenyl.
9. A compound according to claim 1 which is selected from the group consisting of: 5-[(1 R)-2-({9-[4-({3-[(R)-cyclohexyl(hydroxy)phenylmethyl]-1H-1 ,2,4-triazol-1-yl}methyl)piperidin-1- yl]nonyl}amino)-1-hydroxyethyl]-8-hydroxyquinolin-2(1 H)-one,
5-[(1 R)-2-({9-[4-({3-[(R)-cyclohexyl(hydroxy)phenylmethyl]-1H-1 ,2,4-triazol-1-yl}methyl)piperidin-1- yl]nonyl}amino)-1-hydroxyethyl]-8-hydroxyquinolin-2(1 H)-one; naphthalene-1 ,5-disulfonate salt,
5-[(1 R)-2-{[2-(4-{2-[4-({3-[(R)-cyclohexyl(hydroxy)phenylmethyl]-1H-1 ,2,4-triazol-1-yl}methyl)piperidin-1- yl]ethyl}phenyl)ethyl]amino}-1-hydroxyethyl]-8-hydroxyquinolin-2(1 H)-one,
5-[(1 R)-2-{[2-(4-{2-[4-({3-[(R)-cyclohexyl(hydroxy)phenylmethyl]-1H-1 ,2,4-triazol-1-yl}methyl)piperidin-1- yl]ethyl}phenyl)ethyl]amino}-1-hydroxyethyl]-8-hydroxyquinolin-2(1 H)-one naphthalene-1 ,5-disulfonate salt,
5-[(1 R)-2-{[2-(4-{2-[4-({3-[(R)-cyclohexyl(hydroxy)phenylmethyl]-1H-1 ,2,4-triazol-1-yl}methyl)piperidin-1- yl]ethyl}phenyl)ethyl]amino}-1-hydroxyethyl]-8-hydroxyquinolin-2(1 H)-one succinate salt,
5-[(1 R)-2-{[2-(4-{2-[4-({3-[(R)-cyclohexyl(hydroxy)phenylmethyl]-1H-1 ,2,4-triazol-1-yl}methyl)piperidin-1- yl]ethyl}phenyl)ethyl]amino}-1-hydroxyethyl]-8-hydroxyquinolin-2(1 H)-one funnarate salt,
5-[(1 R)-2-({9-[4-({3-[cyclohexyl(hydroxy)phenylmethyl]-1H-1 ,2,4-triazol-1-yl}methyl)piperidin-1- yl]nonyl}amino)-1-hydroxyethyl]-8-hydroxyquinolin-2(1 H)-one,
8-hydroxy-5-[(1 R)-1-hydroxy-2-({9-[4-({3-[hydroxy(diphenyl)methyl]-1H-1 ,2,4-triazol-1-yl}methyl)piperidin-
1-yl]nonyl}amino)ethyl]quinolin-2(1 H)-one, and
8-hydroxy-5-[(1 R)-1-hydroxy-2-{[2-(4-{2-[4-({3-[hydroxy(diphenyl)methyl]-1H-1 ,2,4-triazol-1- yl}nnethyl)piperidin-1-yl]ethyl}phenyl)ethyl]annino}ethyl]quinolin-2(1 H)-one.
10. A compound according to claim 1 which is 5-[(1 R)-2-{[2-(4-{2-[4-({3-[(R)- cyclohexyl(hydroxy)phenylmethyl]-1H-1 ,2,4-triazol-1-yl}methyl)piperidin-1-yl]ethyl}phenyl)ethyl] amino}- 1-hydroxyethyl]-8-hydroxyquinolin-2(1 H)-one naphthalene-1 ,5-disulfonate salt.
11. A pharmaceutical composition comprising at least an effective amount of a compound of the formula (1 ) as described in any one of claims 1 to 10 or a pharmaceutically acceptable salt thereof or a pharmaceutically acceptable solvate of said compound or salt.
12. A compound of the formula (1 ) as described in any one of claims 1 to 10 or a pharmaceutically acceptable salt thereof or a pharmaceutically acceptable solvate of said compound or salt, for use as a medicament.
13. A compound of the formula (1 ) as described in any one of claims 1 to 10 or a pharmaceutically acceptable salt thereof or a pharmaceutically acceptable solvate of said compound or salt, for use in the treatment of diseases, disorders, and conditions selected from the group consisting of:
• asthma of whatever type, etiology, or pathogenesis, in particular asthma that is a member selected from the group consisting of atopic asthma, non-atopic asthma, allergic asthma, atopic bronchial IgE-mediated asthma, bronchial asthma, essential asthma, true asthma, intrinsic asthma caused by pathophysiologic disturbances, extrinsic asthma caused by environmental factors, essential asthma of unknown or inapparent cause, non-atopic asthma, bronchitic asthma, emphysematous asthma, exercise-induced asthma, allergen induced asthma, cold air induced asthma, occupational asthma, infective asthma caused by bacterial, fungal, protozoal, or viral infection, non-allergic asthma, incipient asthma, wheezy infant syndrome and bronchiolytis;
• chronic or acute bronchoconstriction, chronic bronchitis, small airways obstruction, and emphysema;
• obstructive or inflammatory airways diseases of whatever type, etiology, or pathogenesis, in particular an obstructive or inflammatory airways disease that is a member selected from the group consisting of chronic eosinophilic pneumonia, chronic obstructive pulmonary disease (COPD), COPD that includes chronic bronchitis, pulmonary emphysema or dyspnea associated or not associated with COPD, COPD that is characterized by irreversible, progressive airways obstruction, adult respiratory distress syndrome (ARDS), exacerbation of airways hyper-reactivity consequent to other drug therapy and airways disease that is associated with pulmonary hypertension;
• bronchitis of whatever type, etiology, or pathogenesis, in particular bronchitis that is a member selected from the group consisting of acute bronchitis, acute laryngotracheal bronchitis, arachidic bronchitis, catarrhal bronchitis, croupus bronchitis, dry bronchitis, infectious asthmatic bronchitis, productive bronchitis, staphylococcus or streptococcal bronchitis and vesicular bronchitis;
• acute lung injury; and
• bronchiectasis of whatever type, etiology, or pathogenesis, in particular bronchiectasis that is a member selected from the group consisting of cylindric bronchiectasis, sacculated bronchiectasis, fusiform bronchiectasis, capillary bronchiectasis, cystic bronchiectasis, dry bronchiectasis and follicular bronchiectasis.
14. A combination of a compound according to any one of claims 1 to 10, or a pharmaceutically acceptable salt thereof or a pharmaceutically acceptable solvate of said compound or salt, with other therapeutic agent(s) selected from:
(a) 5-Lipoxygenase (5-LO) inhibitors or 5-lipoxygenase activating protein (FLAP) antagonists;
(b) Leukotriene antagonists (LTRAs) including antagonists of LTB4, LTC4, LTD4, and LTE4;
(c) Histamine receptor antagonists including H1 and H3 antagonists;
(d) oti- and α2-adrenoceptor agonist vasoconstrictor sympathomimetic agents for decongestant use;
(e) PDE inhibitors, e.g. PDE3, PDE4 and PDE5 inhibitors;
(f) Beta 2 receptor agonists;
(g) muscarinic M3 receptor antagonist or anticholinergic agents;
(h) Dual compounds active as β2 agonists and muscarinic M3 receptor antagonists;
(i) Theophylline;
(j) Sodium cromoglycate;
(k) COX inhibitors both non-selective and selective COX-1 or COX-2 inhibitors (NSAIDs);
(I) Prostaglandin receptor antagonists and inhibitors of prostaglandin synthase;
(m) Oral and inhaled glucocorticosteroids;
(n) Dissociated agonists of the corticoid receptor (DAGR);
(o) Monoclonal antibodies active against endogenous inflammatory entities;
(p) Anti-tumor necrosis factor (anti-TNF-α) agents;
(q) Adhesion molecule inhibitors including VLA-4 antagonists; (r) KJnJn-B1 - and B2 -receptor antagonists;
(s) Immunosuppressive agents, including inhibitors of the IgE pathway and cyclosporine;
(t) Inhibitors of matrix metalloproteases (MMPs);
(u) Tachykinin NK1, NK2 and NK3 receptor antagonists;
(v) Protease inhibitors such as elastase inhibitors;
(w) Adenosine A2a receptor agonists and A2b antagonists;
(x) Inhibitors of urokinase;
(y) Compounds that act on dopamine receptors, such as D2 agonists;
(z) Modulators of the NFiφ pathway, such as IKK inhibitors;
(aa)modulators of cytokine signalling pathyways such as p38 MAP kinase, PI3 kinase, JAK kinase, syk kinase, EGFR or MK-2;
(bb)Agents that can be classed as mucolytics or anti-tussive; (cc)Agents, which enhance responses to inhaled corticosteroids; (dd)Antibiotics and antivral agents effective against micro-organisms which can colonise the respiratory tract; (ee) HDAC inhibitors; (ff) CXCR2 antagonists; (gg) lntegrin antagonists; (hh) Chemokines;
(ii) Epithelial sodium channel (ENaC) blockers or Epithelial sodium channel (ENaC) inhibitors; (jj) P2Y2 Agonists and other Nucleotide receptor agonists; (kk) Inhibitors of thromboxane;
(II) Inhibitors of PGD2 synthesis and PGD2 receptors (DP1 and DP2/CRTH2); (mm) Niacin; and
(nn)Adhesion factors including VLAM, ICAM, and ELAM.
15. A compound of formula:
H
Figure imgf000058_0001
where p and q are independently selected from 1 or 2 and n, R1 and R2 are as defined in claim 1.
PCT/IB2009/052986 2008-07-11 2009-07-09 Triazole derivatives useful for the treatment of diseases WO2010004517A1 (en)

Priority Applications (18)

Application Number Priority Date Filing Date Title
DK09786555.4T DK2300443T3 (en) 2008-07-11 2009-07-09 Triazole derivatives useful for the treatment of diseases
MX2011000352A MX2011000352A (en) 2008-07-11 2009-07-09 Triazole derivatives useful for the treatment of diseases.
ES09786555T ES2398428T3 (en) 2008-07-11 2009-07-09 Triazole derivatives useful for the treatment of diseases
NZ589613A NZ589613A (en) 2008-07-11 2009-07-09 Triazole derivatives useful for the treatment of diseases
EP09786555A EP2300443B1 (en) 2008-07-11 2009-07-09 Triazole derivatives useful for the treatment of diseases
CN2009801271934A CN102089290B (en) 2008-07-11 2009-07-09 Triazole derivatives useful for the treatment of diseases
EA201001821A EA201001821A1 (en) 2008-07-11 2009-07-09 TRIAZOLE DERIVATIVES USEFUL FOR THE TREATMENT OF DISEASES
KR1020117000612A KR101235962B1 (en) 2008-07-11 2009-07-09 Triazole derivatives useful for the treatment of diseases
CA2726686A CA2726686C (en) 2008-07-11 2009-07-09 Triazole derivatives useful for the treatment of diseases
AP2010005508A AP2010005508A0 (en) 2008-07-11 2009-07-09 Triazole derivatives useful for the treatment of diseases.
JP2011517301A JP5608162B2 (en) 2008-07-11 2009-07-09 Triazole derivatives useful for treating diseases
AU2009269610A AU2009269610B2 (en) 2008-07-11 2009-07-09 Triazole derivatives useful for the treatment of diseases
BRPI0915897A BRPI0915897A2 (en) 2008-07-11 2009-07-09 triazole derivatives, pharmaceutical compositions and combinations thereof and their uses
IL209730A IL209730A (en) 2008-07-11 2010-12-02 Triazole derivatives useful for the treatment of diseases
TNP2010000612A TN2010000612A1 (en) 2009-07-09 2010-12-27 TRIAZOLE DERIVATIVES USEFUL FOR THE TREATMENT OF DISEASE
MA33507A MA32466B1 (en) 2008-07-11 2011-01-10 TRIAZOLE DERIVATIVES USEFUL FOR THE TREATMENT OF DISEASES
ZA2011/00534A ZA201100534B (en) 2008-07-11 2011-01-20 Triazole derivatives useful for the treatment of diseases
HK11108477.2A HK1154382A1 (en) 2008-07-11 2011-08-12 Triazole derivatives useful for the treatment of diseases

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US7983708P 2008-07-11 2008-07-11
US61/079,837 2008-07-11

Publications (1)

Publication Number Publication Date
WO2010004517A1 true WO2010004517A1 (en) 2010-01-14

Family

ID=41078162

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2009/052986 WO2010004517A1 (en) 2008-07-11 2009-07-09 Triazole derivatives useful for the treatment of diseases

Country Status (30)

Country Link
US (1) US8263623B2 (en)
EP (1) EP2300443B1 (en)
JP (1) JP5608162B2 (en)
KR (1) KR101235962B1 (en)
CN (1) CN102089290B (en)
AP (1) AP2010005508A0 (en)
AR (1) AR072488A1 (en)
AU (1) AU2009269610B2 (en)
BR (1) BRPI0915897A2 (en)
CA (1) CA2726686C (en)
CO (1) CO6321248A2 (en)
CR (1) CR11818A (en)
DK (1) DK2300443T3 (en)
DO (1) DOP2011000010A (en)
EA (1) EA201001821A1 (en)
EC (1) ECSP11010754A (en)
ES (1) ES2398428T3 (en)
HK (1) HK1154382A1 (en)
IL (1) IL209730A (en)
MA (1) MA32466B1 (en)
MX (1) MX2011000352A (en)
NI (1) NI201100006A (en)
NZ (1) NZ589613A (en)
PE (1) PE20110151A1 (en)
SV (1) SV2011003802A (en)
TW (1) TW201006822A (en)
UA (1) UA99524C2 (en)
UY (1) UY31976A (en)
WO (1) WO2010004517A1 (en)
ZA (1) ZA201100534B (en)

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011143105A1 (en) * 2010-05-10 2011-11-17 Gilead Sciences, Inc. Bifunctional quinoline derivatives
US8367829B2 (en) 2010-05-10 2013-02-05 Gilead Sciences, Inc. Bi-functional pyrazolopyridine compounds
US9233108B2 (en) 2011-11-11 2016-01-12 Almirall, S.A. Cyclohexylamine derivatives having β2 adrenergic agonist and M3 muscarinic antagonist activities
US9315463B2 (en) 2010-05-13 2016-04-19 Almirall, S.A. Cyclohexylamine derivatives having β2 adrenergic agonist and M3 muscarinic antagonist activities
US9518050B2 (en) 2012-12-18 2016-12-13 Almirall, S.A. Cyclohexyl and quinuclidinyl carbamate derivatives having β2 adrenergic agonist and M3 muscarinic antagonist activity
US9562039B2 (en) 2013-02-27 2017-02-07 Almirall, S.A. Salts of 2-amino-1-hydroxyethyl-8-hydroxyquinolin-2(1H)-one derivatives having both β2 adrenergic receptor agonist and M3 muscarinic receptor antagonist activities
US9579316B2 (en) 2013-07-25 2017-02-28 Almirall, S.A. Salts of 2-amino-1-hydroxyethyl-8-hydroxyquinolin-2(1H)-one derivatives having both muscarinic receptor antagonist and β2 adrenergic receptor agonist activities
US9643961B2 (en) 2010-05-13 2017-05-09 Almirall, S.A. Cyclohexylamine derivatives having β2 adrenergic antagonist and M3 muscarinic antagonist activities
WO2018108089A1 (en) 2016-12-14 2018-06-21 北京硕佰医药科技有限责任公司 Class of bifunctional compounds with quaternary ammonium salt structure
US10005771B2 (en) 2014-09-26 2018-06-26 Almirall, S.A. Bicyclic derivatives having β2 adrenergic agonist and M3 muscarinic antagonist activities
US10342786B2 (en) 2017-10-05 2019-07-09 Fulcrum Therapeutics, Inc. P38 kinase inhibitors reduce DUX4 and downstream gene expression for the treatment of FSHD
US10456390B2 (en) 2013-07-25 2019-10-29 Almirall, S.A. Combinations comprising MABA compounds and corticosteroids
US11291659B2 (en) 2017-10-05 2022-04-05 Fulcrum Therapeutics, Inc. P38 kinase inhibitors reduce DUX4 and downstream gene expression for the treatment of FSHD

Families Citing this family (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR101840852B1 (en) 2011-10-10 2018-03-22 삼성전자주식회사 Surface temperature management method of mobile device and memory thermal management method of multichip package
KR101857529B1 (en) 2011-11-08 2018-05-15 삼성전자주식회사 Nonvolatile memory device and driving method thereof
DE102012111829A1 (en) 2011-12-06 2013-06-06 Samsung Electronics Co., Ltd. Method for operating non-volatile memory device such as flash memory device, involves performing error checking and correction (ECC) workflow to specific pages of read data from first portion of memory cells of memory device
KR101859646B1 (en) 2011-12-16 2018-05-18 삼성전자주식회사 Secure data protecting memory device, data protecting method using the secure data
DE102013100596B4 (en) 2012-01-27 2023-09-07 Samsung Electronics Co. Ltd. Non-volatile memory system with programming and erasing methods and block management methods
KR101903440B1 (en) 2012-02-21 2018-10-02 삼성전자주식회사 Nonvolatile memory device and threshold adjusting method of ground selection transistor thereof
KR101996004B1 (en) 2012-05-29 2019-07-03 삼성전자주식회사 Programming method of nonvolatile memory device and memory system having the same
DE102013105356A1 (en) 2012-05-29 2013-12-05 Samsung Electronics Co., Ltd. Method for operating non-volatile memory device for use in smart-TV system, involves detecting errors generated during programming operation for programming portion of non-volatile multi-bit memory cells in non-volatile memory device
KR102000634B1 (en) 2012-06-07 2019-07-16 삼성전자주식회사 Nonvolatile memory device and erase method thereof
KR102025263B1 (en) 2012-10-05 2019-09-25 삼성전자주식회사 Memory system and read reclaim method thereof
KR102025251B1 (en) 2012-10-31 2019-09-25 삼성전자주식회사 Nonvolatile memory device and program method
KR102068342B1 (en) 2013-03-07 2020-01-20 삼성전자주식회사 Memory controller and memory system including the same
KR102187643B1 (en) 2013-12-04 2020-12-08 삼성전자주식회사 Memory system and user device including the same
CN107194205B (en) * 2017-05-31 2020-11-24 浙江大学 JAK2 kinase inhibitor with bactericidal activity and virtual screening method thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004074246A2 (en) * 2003-02-14 2004-09-02 Theravance Inc. Biphenyl derivatives having beta2 adrenergic receptor agonist and muscarinic receptor antagonist activity
WO2004089892A2 (en) * 2003-04-01 2004-10-21 Theravance, Inc. Diarylmethyl and related compounds having beta2 andrenergic receptor agonist and muscarinic receptor antagonist activity
WO2008023157A1 (en) * 2006-08-21 2008-02-28 Argenta Discovery Limited Nitrogen containing heterocyclic compounds useful as m3-receptor modulators

Family Cites Families (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996033973A1 (en) 1995-04-28 1996-10-31 Banyu Pharmaceutical Co., Ltd. 1,4-disubstituted piperidine derivatives
GB9603755D0 (en) 1996-02-22 1996-04-24 Pfizer Ltd Therapeutic agents
ES2165768B1 (en) 1999-07-14 2003-04-01 Almirall Prodesfarma Sa NEW DERIVATIVES OF QUINUCLIDINE AND PHARMACEUTICAL COMPOSITIONS THAT CONTAIN THEM.
CN1250545C (en) 2000-12-28 2006-04-12 阿尔米雷尔普罗迪斯制药有限公司 Quinuclidine derivatives and their use as M3 antagonists
DE10200943A1 (en) 2002-01-12 2003-07-24 Boehringer Ingelheim Pharma Process for the preparation of scopine esters
EP2177511A2 (en) 2002-12-10 2010-04-21 Ranbaxy Laboratories Limited Process for preparing 3,6-disubstituted azabicyclo derivatives
CN101239971B (en) * 2003-02-14 2011-07-20 施万制药 Biphenyl derivatives
EP1460064A1 (en) 2003-03-14 2004-09-22 Pfizer Limited Indole-2-carboxamide derivatives useful as beta-2 agonists
ATE435862T1 (en) 2003-05-28 2009-07-15 Theravance Inc AZABICYCLOALKAN COMPOUNDS AS MUSCARINE RECEPTOR ANTAGONISTS
PL1708992T3 (en) * 2004-01-22 2007-12-31 Pfizer Sulfonamide derivatives for the treatment of diseases
EP1730103B1 (en) 2004-03-23 2010-05-26 Pfizer Limited Formamide derivatives useful as adrenoceptor
US7538141B2 (en) * 2004-03-23 2009-05-26 Alan Daniel Brown Compounds for the treatment of diseases
EP1833822A2 (en) * 2004-08-16 2007-09-19 Theravance, Inc. Compounds having beta2 adrenergic receptor agonist and muscarinic receptor antagonist activity
GB0424284D0 (en) 2004-11-02 2004-12-01 Novartis Ag Organic compounds
GB0516313D0 (en) 2005-08-08 2005-09-14 Argenta Discovery Ltd Azole derivatives and their uses
TW200745084A (en) * 2006-03-08 2007-12-16 Astrazeneca Ab Novel compounds

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004074246A2 (en) * 2003-02-14 2004-09-02 Theravance Inc. Biphenyl derivatives having beta2 adrenergic receptor agonist and muscarinic receptor antagonist activity
WO2004089892A2 (en) * 2003-04-01 2004-10-21 Theravance, Inc. Diarylmethyl and related compounds having beta2 andrenergic receptor agonist and muscarinic receptor antagonist activity
WO2008023157A1 (en) * 2006-08-21 2008-02-28 Argenta Discovery Limited Nitrogen containing heterocyclic compounds useful as m3-receptor modulators

Cited By (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8367829B2 (en) 2010-05-10 2013-02-05 Gilead Sciences, Inc. Bi-functional pyrazolopyridine compounds
US8394829B2 (en) 2010-05-10 2013-03-12 Gilead Sciences, Inc. Bi-functional quinoline analogs
US8450490B2 (en) 2010-05-10 2013-05-28 Gilead Sciences, Inc. Bi-functional pyrazolopyridine compounds
JP2013526520A (en) * 2010-05-10 2013-06-24 ギリアード サイエンシーズ, インコーポレイテッド Bifunctional quinoline derivative
WO2011143105A1 (en) * 2010-05-10 2011-11-17 Gilead Sciences, Inc. Bifunctional quinoline derivatives
US9643961B2 (en) 2010-05-13 2017-05-09 Almirall, S.A. Cyclohexylamine derivatives having β2 adrenergic antagonist and M3 muscarinic antagonist activities
US9315463B2 (en) 2010-05-13 2016-04-19 Almirall, S.A. Cyclohexylamine derivatives having β2 adrenergic agonist and M3 muscarinic antagonist activities
US10300072B2 (en) 2011-11-11 2019-05-28 Almirall, S.A. Cyclohexylamine derivatives having β2 adrenergic agonist and M3 muscarinic antagonist activities
US9233108B2 (en) 2011-11-11 2016-01-12 Almirall, S.A. Cyclohexylamine derivatives having β2 adrenergic agonist and M3 muscarinic antagonist activities
US9549934B2 (en) 2011-11-11 2017-01-24 Almirall, S.A. Cyclohexylamine derivatives having β2 adrenergic agonist and M3 muscarinic antagonist activities
US9757383B2 (en) 2011-11-11 2017-09-12 Almirall, S.A. Cyclohexylamine derivatives having β2 adrenergic agonist and M3 muscarinic antagonist activities
US9518050B2 (en) 2012-12-18 2016-12-13 Almirall, S.A. Cyclohexyl and quinuclidinyl carbamate derivatives having β2 adrenergic agonist and M3 muscarinic antagonist activity
US9562039B2 (en) 2013-02-27 2017-02-07 Almirall, S.A. Salts of 2-amino-1-hydroxyethyl-8-hydroxyquinolin-2(1H)-one derivatives having both β2 adrenergic receptor agonist and M3 muscarinic receptor antagonist activities
US10456390B2 (en) 2013-07-25 2019-10-29 Almirall, S.A. Combinations comprising MABA compounds and corticosteroids
US9579316B2 (en) 2013-07-25 2017-02-28 Almirall, S.A. Salts of 2-amino-1-hydroxyethyl-8-hydroxyquinolin-2(1H)-one derivatives having both muscarinic receptor antagonist and β2 adrenergic receptor agonist activities
US10005771B2 (en) 2014-09-26 2018-06-26 Almirall, S.A. Bicyclic derivatives having β2 adrenergic agonist and M3 muscarinic antagonist activities
WO2018108089A1 (en) 2016-12-14 2018-06-21 北京硕佰医药科技有限责任公司 Class of bifunctional compounds with quaternary ammonium salt structure
US10342786B2 (en) 2017-10-05 2019-07-09 Fulcrum Therapeutics, Inc. P38 kinase inhibitors reduce DUX4 and downstream gene expression for the treatment of FSHD
US10537560B2 (en) 2017-10-05 2020-01-21 Fulcrum Therapeutics. Inc. P38 kinase inhibitors reduce DUX4 and downstream gene expression for the treatment of FSHD
US11291659B2 (en) 2017-10-05 2022-04-05 Fulcrum Therapeutics, Inc. P38 kinase inhibitors reduce DUX4 and downstream gene expression for the treatment of FSHD
US11479770B2 (en) 2017-10-05 2022-10-25 Fulcrum Therapeutics, Inc. Use of p38 inhibitors to reduce expression of DUX4

Also Published As

Publication number Publication date
CN102089290A (en) 2011-06-08
IL209730A0 (en) 2011-02-28
NI201100006A (en) 2011-07-20
AU2009269610A1 (en) 2010-01-14
PE20110151A1 (en) 2011-03-05
EA201001821A1 (en) 2011-08-30
CA2726686C (en) 2014-03-11
ECSP11010754A (en) 2011-02-28
IL209730A (en) 2013-11-28
UA99524C2 (en) 2012-08-27
US8263623B2 (en) 2012-09-11
US20100010040A1 (en) 2010-01-14
MX2011000352A (en) 2011-02-22
NZ589613A (en) 2012-06-29
AP2010005508A0 (en) 2010-12-31
ES2398428T3 (en) 2013-03-19
CA2726686A1 (en) 2010-01-14
DK2300443T3 (en) 2013-02-18
TW201006822A (en) 2010-02-16
KR20110018423A (en) 2011-02-23
CN102089290B (en) 2013-09-04
KR101235962B1 (en) 2013-02-21
MA32466B1 (en) 2011-07-03
SV2011003802A (en) 2011-03-23
DOP2011000010A (en) 2011-01-31
ZA201100534B (en) 2011-10-26
UY31976A (en) 2010-02-26
HK1154382A1 (en) 2012-04-20
AR072488A1 (en) 2010-09-01
CO6321248A2 (en) 2011-09-20
AU2009269610B2 (en) 2012-02-02
CR11818A (en) 2011-01-14
EP2300443B1 (en) 2012-12-12
JP5608162B2 (en) 2014-10-15
BRPI0915897A2 (en) 2015-11-03
JP2011527672A (en) 2011-11-04
EP2300443A1 (en) 2011-03-30

Similar Documents

Publication Publication Date Title
EP2300443B1 (en) Triazole derivatives useful for the treatment of diseases
US10150752B2 (en) Substituted triazole derivatives as oxytocin antagonists
KR101064787B1 (en) Amine derivatives
US20060111416A1 (en) Octahydropyrrolo[3,4-C]pyrrole derivatives
EP2074094A1 (en) Sulfonamide derivatives as adrenergic agonists and muscarinic antagonists
JP2009526037A (en) Triazolopyridine compound
WO2006056848A1 (en) Octahydropyrrolo[3,4-c]pyrrole derivatives
JPH10287634A (en) Benzene derivatives
WO2008035157A1 (en) Azetidine derivatives as muscarinic receptor antagonists
WO2007017752A1 (en) Substituted triazole derivatives as oxytocin antagonists
EP1671972A1 (en) Octahydropyrrolo[3,4-c]pyrrole derivatives
EP1680423A1 (en) Azabenzodiazepines as phosphodiesterase-4 inhibitors

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200980127193.4

Country of ref document: CN

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

Ref document number: 09786555

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 201011818

Country of ref document: CR

Ref document number: CR2010-011818

Country of ref document: CR

WWE Wipo information: entry into national phase

Ref document number: 589613

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 2726686

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 209730

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 2009269610

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2010122098

Country of ref document: EG

WWE Wipo information: entry into national phase

Ref document number: 2009786555

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 10160881

Country of ref document: CO

Ref document number: 9143/DELNP/2010

Country of ref document: IN

Ref document number: 201001821

Country of ref document: EA

WWE Wipo information: entry into national phase

Ref document number: 000007-2011

Country of ref document: PE

ENP Entry into the national phase

Ref document number: 2009269610

Country of ref document: AU

Date of ref document: 20090709

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: D2011002

Country of ref document: CU

WWE Wipo information: entry into national phase

Ref document number: 12011500055

Country of ref document: PH

WWE Wipo information: entry into national phase

Ref document number: DZP2011000020

Country of ref document: DZ

ENP Entry into the national phase

Ref document number: 20117000612

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: MX/A/2011/000352

Country of ref document: MX

ENP Entry into the national phase

Ref document number: 2011517301

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 12054

Country of ref document: GE

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: A201014259

Country of ref document: UA

ENP Entry into the national phase

Ref document number: PI0915897

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20110111