WO2007036734A1 - Pyrazolo[3,4-b]pyridine compound, and its use as a pde4 inhibitor - Google Patents

Pyrazolo[3,4-b]pyridine compound, and its use as a pde4 inhibitor Download PDF

Info

Publication number
WO2007036734A1
WO2007036734A1 PCT/GB2006/003627 GB2006003627W WO2007036734A1 WO 2007036734 A1 WO2007036734 A1 WO 2007036734A1 GB 2006003627 W GB2006003627 W GB 2006003627W WO 2007036734 A1 WO2007036734 A1 WO 2007036734A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
salt
pharmaceutically acceptable
formula
mammal
Prior art date
Application number
PCT/GB2006/003627
Other languages
French (fr)
Inventor
Siegfried Benjamin Christensen, Iv
Stuart Holman
Suzanne Elaine Keeling
Amyn Pyarali Sayani
Original Assignee
Glaxo Group Limited
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Glaxo Group Limited filed Critical Glaxo Group Limited
Priority to US12/067,994 priority Critical patent/US20080255186A1/en
Priority to EP06779579A priority patent/EP1940836A1/en
Priority to JP2008532874A priority patent/JP2009510044A/en
Publication of WO2007036734A1 publication Critical patent/WO2007036734A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • 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
    • A61P11/02Nasal agents, e.g. decongestants
    • 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
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/04Antipruritics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/02Drugs for disorders of the nervous system for peripheral neuropathies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/22Anxiolytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/24Antidepressants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/14Decongestants or antiallergics
    • 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
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P39/00General protective or antinoxious agents
    • A61P39/02Antidotes
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/08Vasodilators for multiple indications

Definitions

  • the present invention relates to a pyrazolo[3,4-b]pyridine compound or a salt thereof, processes for their preparation, intermediates usable in these processes, and pharmaceutical compositions containing the compound or a salt thereof.
  • the invention also relates to the use of the pyrazolo[3,4-b]pyridine compound or a salt thereof in therapy, for example as inhibitors of phosphodiesterase type IV (PDE4) and/or for the treatment and/or prophylaxis of inflammatory and/or allergic diseases such as atopic dermatitis.
  • PDE4 phosphodiesterase type IV
  • WO 2004/024728 A2 (PCT/EP2003/011814, filed on 12 September 2003, published on 25 March 2004, Glaxo Group Limited), and incorporated herein by reference in its entirity as though fully set forth, discloses pyrazolo[3,4-b]pyridine compounds or salts thereof with a 4-NHR3 group and a 5-C(O)-X group (wherein X is NR4R5 or OR ⁇ a ), according to the following formula (I):
  • pyrazolo[3,4-b]pyridine compounds of formula (I) and salts thereof disclosed therein are disclosed as being inhibitors of phosphodiesterase type IV (PDE4).
  • WO 2004/056823 A1 (PCT/EP2003/014867, filed on 19 December 2003, published on 8 July 2004, Glaxo Group Limited), and incorporated herein by reference in its entirity as though fully set forth, discloses and claims further pyrazolo[3,4-b]pyridine compounds or salts thereof. WO 2004/056823 A1 also discloses the use of these compounds as PDE4 inhibitors.
  • WO 2004/024728 has been reviewed, and WO 2004/056823 mentioned, in Expert Opin. Ther. Patents, 2005 (January edition), 15(1), 111-114.
  • WO 2005/058892 A1 (PCT/EP2004/014490, filed on 17 December 2004, published on 30 June 2005, Glaxo Group Limited), and incorporated herein by reference in its entirity as though fully set forth, discloses further pyrazolo[3,4-b]pyridine compounds or salts thereof and their use as PDE4 inhibitors.
  • PCT/GB2005/000983 PCT/GB2005/000983
  • WO 2005/090354 A1 PCT/GB2005/000987
  • WO 2005/090352 A1 PCT/EP2005/003038
  • WO 2005/090353 A1 PCT/GB2005/000976
  • the present invention therefore provides ⁇ /- ⁇ [1 ,6-diethyl-4-(tetrahydro-2H-pyran-4- ylamino)-1H-pyrazolo[3,4-b]pyridin-5-yl]methyl ⁇ -3-methyl-5-isoxazolecarboxamide
  • the present invention provides ⁇ /- ⁇ [1 ,6-diethyl-4-(tetrahydro-2H-pyran-4- ylamino)-1 H-pyrazolo[3,4-jb]pyridin-5-yl]methyl ⁇ -3-methyl-5-isoxazolecarboxamide or a pharmaceutically acceptable salt thereof.
  • the present invention provides ⁇ /- ⁇ [1 ,6-diethyl-4-(tetrahydro-2/-/-pyran-4- ylamino)-1H-pyrazolo[3,4-b]pyridin-5-yl]methyl ⁇ -3-methyl-5-isoxazolecarboxamide.
  • This is the so-called “free base” form, also called “the compound of the invention” or similar.
  • the above-defined compound or salt of the present invention is a compound of formula (I) or a salt thereof (in particular, the compound or a pharmaceutically acceptable salt thereof): wherein: R 1 is ethyl; R 2 is ethyl;
  • R 3 is a heterocyclic group of sub-formula (bb) which is not substituted on a ring carbon:
  • R a is a hydrogen atom (H);
  • R b is a hydrogen atom (H);
  • R 4 is a hydrogen atom (H);
  • R 5 is -C(O)-(CH2) n -Ar; wherein n is 0;
  • an "alkyl” group or moiety may be straight-chain or branched.
  • Alkyl groups for example C-i ⁇ 'M or C ⁇ galkyl or C-
  • alkoxy such as C-i ⁇ alkoxy or C- ⁇ alkoxy or Ci_ 2 alkoxy includes methoxy, ethoxy, propyloxy, and oxy derivatives of the alkyls listed above.
  • Alkylsulfonyl such as C- ⁇ alkylsulfonyl includes methylsulfonyl (methanesulfonyl), ethylsulfonyl, and others derived from the alkyls listed above.
  • Alkylsulfonyloxy such as C-
  • “Fluoroalkyl” includes alkyl groups with one, two, three, four, five or more fluorine substituents, for example C-j_4fluoroalkyl or Ci_3fluoroalkyl or C-
  • “Fluoroalkoxy” includes C- ⁇ fluoroalkoxy or C-).
  • fluoroalkoxy such as trifluoromethoxy, pentafluoroethoxy, monofluoromethoxy, difluoromethoxy, etc.
  • fluoroalkylsulfonyl such as Ci_4fluoroalkylsulfonyl includes trifluoromethanesulfonyl, pentafluoroethylsulfonyl, etc.
  • halogen atom present in compounds, for example in the compounds of formula (I), means a fluorine, chlorine, bromine or iodine atom ("fluoro", “chloro”, “bromo” or “iodo”), for example fluoro, chloro or bromo.
  • R 3 is tetrahydro-2H-pyran-4-yl; that is NHR 3 is of sub-formula (h) as shown below:
  • the compound of the invention or the salt thereof can suitably be for external topical administration (e.g. to a mammal such as a human or pig), in particular for topical administration to the skin.
  • external topical administration e.g. to a mammal such as a human or pig
  • the compound of the invention or a pharmaceutically acceptable salt thereof can suitably be contained / comprised in a pharmaceutical composition suitable and/or adapted for external topical administration (e.g. to a mammal such as a human or pig), in particular in a pharmaceutical composition suitable and/or adapted for topical administration to the skin (e.g. to a mammal such as a human or pig).
  • a pharmaceutical composition suitable and/or adapted for external topical administration e.g. to a mammal such as a human or pig
  • a pharmaceutical composition suitable and/or adapted for topical administration to the skin e.g. to a mammal such as a human or pig.
  • the compound of formula (I) may be used in the form of a pharmaceutically acceptable salt thereof.
  • Pharmaceutically acceptable salts can include acid addition salts.
  • a pharmaceutically acceptable acid addition salt of the compound of the invention can be formed (or can have been formed) by mixing (e.g. intimately mixing) ⁇ /- ⁇ [1 ,6-diethyl-4-(tetrahydro-2H ⁇ pyran-4-ylamino)-1 H ⁇ pyrazolo[3,4- ⁇ b]pyridin-5- yl]methyl ⁇ -3-methyl-5-isoxazolecarboxamide with a pharmaceutically acceptable acid having a pKa of 1.5 or less.
  • a pharmaceutically acceptable acid addition salt is optionally formed by mixing (e.g. intimately mixing) a compound of formula (I) with a suitable pharmaceutically acceptable inorganic or organic acid (such as hydrobromic, hydrochloric, sulfuric, nitric, phosphoric, p-toluenesulfonic, benzenesulfonic, methanesulfonic, ethanesulfonic, or naphthalenesulfonic such as 2- naphthalenesulfonic acid), optionally in (e.g. dissolved in) a suitable solvent, and/or optionally in (e.g.
  • a suitable pharmaceutically acceptable inorganic or organic acid such as hydrobromic, hydrochloric, sulfuric, nitric, phosphoric, p-toluenesulfonic, benzenesulfonic, methanesulfonic, ethanesulfonic, or naphthalenesulfonic such as 2- na
  • the compound of formula (I) is for example dissolved in a suitable solvent, and/or in an organic solvent such as methanol, ethanol, isopropanol, ethyl acetate, acetonitrile and/or dichloromethane, and the pharmaceutically acceptable inorganic or organic acid (e.g. in solution or in isolated form) is then added, to prepare the pharmaceutically acceptable salt, which is usually isolated for example by crystallisation and filtration.
  • an organic solvent such as methanol, ethanol, isopropanol, ethyl acetate, acetonitrile and/or dichloromethane
  • an anhydrous solution e.g. 1-5M, e.g. 2M
  • aqueous hydrochloric acid or HCI gas
  • an aqueous, methanolic or ethanolic solution of sulfuric acid or nitric acid respectively is optionally added to a solution of the compound of formula (I) in a water-miscible organic solvent.
  • a pharmaceutically acceptable acid addition salt of a compound of formula (I) can comprise or be for example a hydrobromide, hydrochloride, sulfate, nitrate, phosphate, p-toluenesulfonate, benzenesulfonate, methanesulfonate, ethanesulfonate, or naphthalenesulfonate (e.g. 2- naphthalenesulfonate) salt of the compound of formula (I).
  • Other non-pharmaceutically acceptable salts e.g. oxalates or trifluoroacetates, may be used, for example in the isolation of compounds of the invention, and are included within the scope of this invention.
  • the invention includes within its scope all possible stoichiometric and non- stoichiometric forms of the salts of the compounds of formula (I). Also included within the scope of the invention are all solvates, hydrates and polymorphs of compounds and salts of the invention.
  • Certain salts of the compound of the invention may be present as isomers, e.g. positional, geometric or optical isomers.
  • the present invention includes within its scope all such isomers, including racemates, enantiomers and mixtures thereof.
  • Certain of the groups, e.g. heteroaromatic ring systems or amide moiety(ies), included in the compound of formula (I) or a salt thereof may exist in one or more tautomeric forms.
  • the present invention includes within its scope all such tautomeric forms, including mixtures.
  • R a and R b are both a hydrogen atom (H), R 4 is H, R 5 is -C(O)-(CH2)n ⁇ Ar; wherein n is 0 and wherein Ar is as defined herein, and R ⁇ , R ⁇ and R ⁇ are as defined herein.
  • an amine of formula (II) or a salt thereof can be reacted with a compound Ar-C(O)-X " * , wherein X ⁇ is a leaving group substitutable by the NH2 amine moiety of the compound of formula (II):
  • Ar-C(O)-X ⁇ is typically an activated derivative of carboxylic acid Ar-C(O)-OH.
  • a compound Ar-C(O)-X 1 can for example be the acid chloride Ar-C(O)CI (wherein X 1 is a chlorine atom (Cl)).
  • a suitable non-aqueous non-alcohol organic solvent e.g. anhydrous solvent
  • acetonitrile e.g. anhydrous
  • DMF room temperature
  • the acid chloride Ar-C(O)CI can typically be formed from the carboxylic acid (a) by reaction with thionyl chloride, either in a suitable organic solvent such as chloroform or without solvent, or (b) by reaction with oxalyl chloride (optionally also with e.g. diethyl formamide or DMF), for example in a suitable organic solvent such as dichloromethane.
  • the compound Ar-C(O)-X 1 can be an activated derivative of the carboxylic acid Ar-C(O)-OH wherein the leaving group X 1 is
  • the activated compound Ar-C(O)-X-I can optionally be formed from the carboxylic acid Ar-C(O)-OH by the following reaction (a).
  • reaction (a) the carboxylic acid Ar-C(O)-OH is reacted with (i), (ii), (iii) or (iv):
  • HBTU O-(benzotriazol-1-yl)- ⁇ /, ⁇ /, ⁇ /',/V'-tetramethyluronium hexafluorophosphate
  • TBTU 2-(1 H-benzotriazol-1-yl)-1 ,1 ,3,3-tetramethyluronium tetrafluoroborate
  • reaction (a) is e.g. carried out at room temperature (e.g. about 18 to about 25 0 C), and/or for a reaction time of from 2 to 48 hours or 6 to 48 hours such as 12 to 24 hours, for example about 15-18 hours.
  • reaction (a) can optionally be carried out under anhydrous conditions.
  • the activated compound Ar-C(O)-X 1 is for example formed from the carboxylic acid Ar-C(O)-OH by the following reaction (b).
  • EDC 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide or a salt thereof
  • this reaction (b) is for example carried out in the presence of a nonaqueous non-alcohol organic solvent (e.g. anhydrous solvent) such as dimethyl formamide (DMF) or acetonitrile and/or e.g. at room temperature and/or e.g. under anhydrous conditions.
  • a nonaqueous non-alcohol organic solvent e.g. anhydrous solvent
  • DMF dimethyl formamide
  • acetonitrile e.g. at room temperature and/or e.g. under anhydrous conditions.
  • reaction (b) the reaction Of Ar-C(O)-OH with the carbodiimide is optionally carried out in the presence of 1-hydroxybenzotriazole (HOBT), in which case,
  • HOBT 1-hydroxybenzotriazole
  • an activated compound is often the compound which reacts with the amine of formula (II) or a salt thereof (cf. reaction (a) above).
  • the amine compound of formula (II) can be prepared by hydrogenation of an azide compound of formula (Ilia):
  • Hydrogenation conditions can for example include H2 / palladium on carbon (e.g. 5 to
  • the azide compound of formula (Ilia) can generally be prepared by reaction of a compound of formula (lllb), wherein X ⁇ b js a leaving group displaceable by azide, with a metal azide such as sodium azide, lithium azide or potassium azide:
  • Typical conditions for the (lllb) to (Ilia) reaction can e.g. include DMSO solvent (e.g. dry) or DMF solvent (e.g. dry) and/or reaction at room temperature. See for example Reference Intermediate 7 and Intermediate 8.
  • Compounds of formula (INb), in particular wherein X 3b is Cl or an organic sulfonate, can generally or sometimes be prepared by conversion (e.g. chlorination) of an alcohol compound of formula (IMc).
  • This can for example be by reaction with SOCI2 (thionyl chloride) for chlorination (when X 3 I 3 is Cl), or by reaction with methanesulfonyl chloride (when X 3b is methanesulfonate) or p-toluenesulfonyl chloride (when X 3b is p-toluenesulfonate).
  • the chlorination reaction with thionyl chloride may require heating, e.g. to about 60 to about 90 0 C, for example at about 85 0 C:
  • azide compounds of formula (Ilia) may be preparable directly from the alcohol compound of formula (lllc).
  • reacting compounds of formula (lllc) with an azide, e.g. sodium azide, in the presence of carbon tetrabromide and triphenylphosphine may give a compound of formula (Ilia) (e.g see Toyota et. al.
  • the amine compound of formula (II) or a salt thereof can be prepared directly from the compound of formula (MIb) or a salt thereof (for example wherein X 3b is a chlorine atom, for example from the benzenesulfonate salt of the compound of formula (lllb) e.g. wherein X 3b is a chlorine atom), without converting (lllb) to the azide compound of formula (Ilia):
  • this reaction of (lllb) or a salt thereof to the amine (II) or a salt thereof can generally be carried out by reaction of the compound of formula (lllb) (for example wherein X 3 * 3 is or comprises a chlorine atom) with an aminating agent such as lithium hexamethyldisilazide, sodium hexamethyldisilazide or potassium hexamethyldisilazide (in particular lithium hexamethyldisilazide, e.g. with slow mixing/addition), in a suitable non-aqueous non-alcohol organic solvent (e.g.
  • an aminating agent such as lithium hexamethyldisilazide, sodium hexamethyldisilazide or potassium hexamethyldisilazide (in particular lithium hexamethyldisilazide, e.g. with slow mixing/addition)
  • a suitable non-aqueous non-alcohol organic solvent e.
  • anhydrous solvent such as tetrahydrofuran
  • a suitable temperature of, for example, about 25 to about 50 0 C, e.g. ca. 30-45 0 C or ca. 35-40 0 C.
  • the reaction is suitably followed by treatment with an aqueous acid such as aqueous hydrochloric acid (e.g. 2-10M, e.g. about 5M), e.g. at a suitable temperature such as from O 0 C to room temperature, for example at about 1O 0 C.
  • aqueous acid such as aqueous hydrochloric acid (e.g. 2-10M, e.g. about 5M)
  • a suitable temperature such as from O 0 C to room temperature, for example at about 1O 0 C.
  • the alcohol compound of formula (I I Ic) or a salt thereof is converted into the amine of formula (II) or a salt thereof, without substantially purifying and/or without substantially isolating the compound of formula (lllb) or the salt thereof wherein X 3b is a chlorine atom.
  • the compound of formula (lllb) or a salt thereof wherein X 3b is a chlorine atom can for example be in the form of the benzenesulfonate salt:
  • the alcohol compound of formula (IUc) can be prepared by reduction of an ester compound of formula (IV), wherein R e is Q> ⁇ .4 straight-chain alkyl such as ethyl, in the presence of a reducing agent.
  • the reducing agent can for example be diisobutylaluminium hydride or lithium borohydride (LJBH4).
  • LJBH4 lithium borohydride
  • the reduction can e.g. be in dichloromethane and/or toluene solvent (e.g. see Intermediate 4 or Reference Intermediate 3 herein) and/or can e.g.
  • Compounds of formula (IV) can generally be prepared according to a method, for example as described by Yu et. al. in J. Med Chem., 2001, 44, 1025-1027, by reaction of a compound of formula (V) with an amine of formula R 3 NH2 or a salt thereof such as a hydrochloride salt thereof.
  • the reaction is for example carried out in the presence of a tertiary amine base such as triethylamine or N.N-diisopropylethylamine, and/or in an organic solvent such as 1-methyl-2-pyrrolidinone (NMP), ethanol, dioxane or acetonitrile.
  • the reaction may comprise heating e.g. heating to ca.
  • 60-180 0 C or 80-180 0 C for example ca. 60-100 0 C (e.g. ca. 80-90 0 C) or ca. 110-160 0 C, for example depending on the reflux temperature or boiling point of the solvent(s) used.
  • Intermediate 2 see e.g. Intermediate 2 herein, or see e.g. the process shown in Yu et. al. in J. Med. Chem., 2001 , 44, 1025-1027 (see steps c+d of Scheme 1 therein):
  • the amine of formula R 3 NH2 is commercially available, e.g. from Combi-Blocks Inc. and/or from Peakdale Molecular Ltd. See Intermediate 12 one possible synthesis of the hydrochloride salt of R 3 NH2-
  • the compound of formula (Vl) is reacted with a dialkyl (i-chloroalkylidene)propanedioate with heating, for example in a suitable organic solvent such as toluene, and for example in the presence of a suitable base such as triethylamine, e.g. at a suitable temperature such as the reflux temperature of the solvent.
  • a suitable organic solvent such as toluene
  • a suitable base such as triethylamine
  • POCI3 phosphorous oxychloride
  • the conditions for the reaction of the intermediate [formed from (Vl) and the dialkyl (i-chloroalkylidene)propanedioate] with phosphorous oxychloride (POCI3) can include heating, e.g. heating at the reflux temperature of phosphorous oxychloride.
  • a compound of formula (XV) is prepared by reaction of a dialkyl malonate of formula (XVI) with magnesium chloride and a suitable non-aqueous base such as triethylamine, in a suitable solvent (e.g. anhydrous solvent) such as acetonitrile, at a suitable temperature such as ca. 5-10 0 C, followed by addition of an acid chloride of formula (XVII) (propanoyl chloride when R ⁇ is ethyl), at a suitable temperature such as between 10 0 C and room temperature.
  • a suitable solvent e.g. anhydrous solvent
  • an acid chloride of formula (XVII) propanoyl chloride when R ⁇ is ethyl
  • the reaction is for example carried out under anhydrous conditions:
  • the desired amino pyrazole of formula (Vl), which (for R 1 is ethyl) is 1 -ethyl- 1 H-pyrazol- 5-amine, is commercially available, e.g. is commercially available from one or more of the following suppliers: Aldrich Chemical Company Inc., Albemarle Corporation, Art- Chem GmbH, Enamine and/or TimTec Corporation.
  • the amino pyrazole (Vl) is optionally prepared, for example, by reaction of cyanoethyl hydrazine with a suitable aldehyde of formula R 40 CHO in a solvent such as ethanol, with heating, followed by reduction, for example reduction with sodium in a suitable solvent such as t-butanol.
  • the leaving group displaceable by the amine is for example R ⁇ A 1 j n the compound of formula (Va) shown below, wherein R*-A j s a bromine atom or an alkoxy group OR 35 such as OCi ⁇ alkyl (in particular OEt) or a group -O-S(O)2-R 37 .
  • R 37 can e.g. be C- ⁇ alkyl such as methyl, C ⁇ fluoroalkyl such as CF3, phenyl, or
  • reaction of the compound of formula (Va) with the amine of formula R 3 NH2 or a salt thereof is optionally carried out with or without solvent, and e.g. may comprise heating:
  • the present invention therefore also provides a method of (process for) preparing a compound of formula (I) le salt) thereof:
  • another aspect of the present invention provides a process for preparing a compound of formula (I) or a salt (e.g. pharmaceutically acceptable salt) thereof: wherein R 1 , R 2 , R 3 , R 4 , R 5 , R a and R* 3 are as defined herein, which process comprises:
  • process (A) preferred, suitable or optional features of process (A), can be as described above for Process A (e.g. those described above as preferred, suitable, optional or example features of this process), with all necessary changes being made.
  • a compound Ar-C(O)-X 1 can for example be the acid chloride Ar-C(O)CI (i.e. X 1 is a chlorine atom (Cl)).
  • the compound Ar-C(O)-X 1 can be an activated derivative of the carboxylic acid Ar-C(O)-OH wherein the leaving group X 1 is
  • R c1 and R C2 can e.g. both be cyclohexyl; or one of R C 1 and R C 2 j s ethyl and the other of R c1 and R c2 is 3-dimethylaminopropyl.
  • the present invention also provides: (B) a process for preparing a pharmaceutically acceptable salt of a compound of formula (I) comprising conversion of the compound of formula (I) or a salt thereof into a or the desired pharmaceutically acceptable salt of the compound of formula (I).
  • a pharmaceutically acceptable salt can be an acid addition salt.
  • a pharmaceutically acceptable acid addition salt is optionally prepared by reaction of a compound of formula (I) with a suitable inorganic or organic acid (e.g. as described hereinabove).
  • the present invention also provides a compound of formula (I) or a salt thereof, prepared by (or obtainable by) a process as defined herein.
  • the present invention also provides a compound of formula (I) or a pharmaceutically acceptable salt thereof for use as an active therapeutic substance in a mammal such as a human.
  • the compound or salt can be for use in the treatment and/or prophylaxis of any of the diseases / conditions described herein (e.g. for use in the treatment and/or prophylaxis of an inflammatory and/or allergic disease in a mammal such as a human; or e.g. for use in the treatment and/or prophylaxis of cognitive impairment (e.g. in a neurological disorder such as Alzheimer's disease) or depression in a mammal such as a human) and/or can be for use as a phosphodiesterase 4 (PDE4) inhibitor.
  • “Therapy” may include treatment and/or prophylaxis.
  • the compound or salt is in particular for use in the treatment and/or prophylaxis of an inflammatory and/or allergic skin disease, such as atopic dermatitis or psoriasis (in particular atopic dermatitis), in a mammal such as a human.
  • an inflammatory and/or allergic skin disease such as atopic dermatitis or psoriasis (in particular atopic dermatitis)
  • a mammal such as a human.
  • a compound of formula (I) or a pharmaceutically acceptable salt thereof in the manufacture of a medicament (e.g. pharmaceutical composition) for the treatment and/or prophylaxis of any of the diseases / conditions described herein in a mammal such as a human, e.g. for the treatment and/or prophylaxis of an inflammatory and/or allergic disease in a mammal such as a human, or e.g. for the treatment and/or prophylaxis of cognitive impairment (e.g. in a neurological disorder such as Alzheimer's disease) or depression in a mammal.
  • a medicament e.g. pharmaceutical composition
  • a method of treatment and/or prophylaxis of any of the diseases / conditions described herein in a mammal (e.g. human) in need thereof e.g. a method of treatment and/or prophylaxis of an inflammatory and/or allergic disease, cognitive impairment (e.g. in a neurological disorder such as Alzheimer's disease or schizophrenia) or depression in a mammal (e.g. human) in need thereof, which method comprises administering to the mammal (e.g. human) a therapeutically effective amount of a compound of formula (I) as herein defined or a pharmaceutically acceptable salt thereof.
  • Phosphodiesterase 4 inhibitors are thought to be, or may be, potentially useful in the treatment and/or prophylaxis of a variety of diseases / conditions, especially inflammatory and/or allergic diseases, in a mammal such as a human, for example: asthma, chronic obstructive pulmonary disease (COPD) (e.g. chronic bronchitis and/or emphysema), atopic dermatitis, urticaria, rhinitis (e.g.
  • COPD chronic obstructive pulmonary disease
  • COPD chronic obstructive pulmonary disease
  • atopic dermatitis e.g. chronic bronchitis and/or emphysema
  • urticaria urticaria
  • rhinitis e.g.
  • rhinitis allergic rhinitis
  • vernal conjunctivitis vernal conjunctivitis
  • eosinophilic granuloma psoriasis
  • rheumatoid arthritis rheumatoid arthritis
  • septic shock ulcerative colitis
  • Crohn's disease reperfusion injury of the myocardium and brain
  • chronic glomerulonephritis e.g. in a neurological disorder such as Alzheimer's disease or schizophrenia
  • depression e.g. inflammatory pain
  • Ulcerative colitis and/or Crohn's disease are collectively often referred to as inflammatory bowel disease.
  • Phosphodiesterase 4 inhibitors may also be potentially useful in the treatment and/or prophylaxis of anxiety in a mammal such as a human.
  • the inflammatory and/or allergic disease can for example be chronic obstructive pulmonary disease (COPD), asthma, rheumatoid arthritis, rhinitis (e.g. allergic rhinitis), psoriasis or atopic dermatitis in a mammal (e.g. human).
  • COPD chronic obstructive pulmonary disease
  • asthma rheumatoid arthritis
  • rhinitis e.g. allergic rhinitis
  • psoriasis atopic dermatitis in a mammal (e.g. human).
  • the treatment and/or prophylaxis is of an inflammatory and/or allergic skin disease such as psoriasis or atopic dermatitis (in particular atopic dermatitis) in a mammal (e.g. human).
  • the treatment and/or prophylaxis is of atopic dermatitis in a mammal such as a human or pig, in particular in a human, in particular in a human aged 21 years or less, e.g. 18 years or less.
  • a mammal such as a human or pig
  • external topical administration to the mammal of the compound of formula (I) or a pharmaceutically acceptable salt thereof e.g. topical administration to the skin e,g. to skin affected by the atopic dermatitis
  • inhaled administration is usually not suitable.
  • Atopic dermatitis has been proposed to include two general sub-classes: (1) an "allergic (extrinsic)” type of atopic dermatitis which generally occurs in the context of sensitization to environmental allergens and/or which is generally accompanied by elevated serum IgE levels; and (2) an "non-allergic (intrinsic)” type of atopic dermatitis generally with little or no detectable sensitization and/or generally with normal or low serum IgE levels (N. Novak et al., J. Allergy Clin. Immunol., 2003, 112, 252-262; and T.C. Roos et al., Drugs, 2004, 64(23), 2639-2666, see e.g.
  • the compound of formula (I) or the pharmaceutically acceptable salt thereof can therefore be for the treatment and/or prophylaxis of allergic (extrinsic) atopic dermatitis and/or non- allergic (intrinsic) atopic dermatitis in a mammal (e.g. human or pig, in particular a human).
  • a mammal e.g. human or pig, in particular a human.
  • External topical administration means topical administration to an external body part (i.e. excluding, for example, the lung or mouth, but including the lips), in particular excluding the eye.
  • External topical administration is for example topical administration to the skin, for example to the skin of an arm, hand, leg, foot, head (e.g. face), neck and/or torso of a mammal such as a human.
  • External topical administration can for example be to those parts of a mammal's skin affected by or susceptible to atopic dermatitis.
  • PDE4 inhibitors for example cilomilast and roflumilast, are thought to be effective in the treatment of COPD.
  • PDE4 inhibitors for example cilomilast and roflumilast.
  • S.L. Wolda Emerging Drugs, 2000, 5(3), 309- 319
  • Z. Huang et al. Current Opinion in Chemical Biology, 2001 , 5: 432-438
  • H.J. Dyke et al. Expert Opinion on Investigational Drugs, January 2002, 11 (1 ), 1 -13
  • C.Burnouf et al. Current Pharmaceutical Design, 2002, 8(14), 1255-1296
  • A.M.Doherty Current Opinion Chem. Biol., 1999, 3(4), 466-473
  • the PDE4 inhibitor cilomilast (Ariflo TM) at 15 mg orally twice daily appears to improve forced expiratory volume in 1s (FEV 1 ) in COPD patients (C.H.Compton et al., The Lancet, 2001 , vol.358, 265-270), and appears to have antiinflammatory effects in COPD patients (E.Gamble et al., Am. J. Respir. Crit. Care Med., 2003, 168, 976-982).
  • On cilomilast see also R.D. Border et al., Chest, 2003, vol. 124 Suppl. 4, p.170S (abstract) and J. D. Eddleston et al., Am. J. Respir. Crit.
  • COPD is often characterised by the presence of airflow obstruction due to chronic bronchitis and/or emphysema (e.g., see S.L. Wolda, Emerging Drugs, 2000, 5(3), 309- 319).
  • oral, inhaled or parenteral administration to the mammal of the compound of formula (I) or a pharmaceutically acceptable salt thereof is optionally used, e.g. oral or inhaled administration.
  • PDE4 inhibitors are thought to be effective in the treatment and/or prophylaxis of asthma (e.g. see M.A.Giembycz, Drugs, Feb. 2000, 59(2), 193-212; Z. Huang et al., Current Opinion in Chemical Biology, 2001, 5: 432-438; HJ. Dyke et al., Expert Opinion on Investigational Drugs, January 2002, 11(1), 1-13; C.Burnouf et al., Current Pharmaceutical Design, 2002, 8(14), 1255-1296; A.M.Doherty, Current Opinion Chem. Biol., 1999, 3(4), 466-473; PJ. Barnes, Nature Reviews - Drug Discovery, October 2004, 831-844; BJ.
  • the PDE4 inhibitor roflumilast given orally at 500 ug once daily for 9 days, is reported to be effective in improving rhinal airflow during the treatment period (compared to placebo), in humans with histories of allergic rhinitis but asymptomatic at screening, and who were challenged with intranasal allergen provocation (pollen extracts) daily beginning the third day of treatment and each time approx. 2 hours after study drug administration (see B.M. Schmidt et al., J. Allergy & Clinical Immunology, 108(4), 2001, 530-536).
  • rhinitis e.g. allergic rhinitis
  • intranasal, oral or parenteral administration of the compound of formula (I) or a pharmaceutically acceptable salt thereof is optionally used.
  • the compound of formula (I) or the pharmaceutically acceptable salt thereof is for the treatment and/or prophylaxis of rhinitis, such as allergic rhinitis (e.g. seasonal allergic rhinitis or perennial allergic rhinitis) or non-allergic rhinitis (e.g. vasomotor rhinitis), in a mammal such as a human.
  • rhinitis such as allergic rhinitis (e.g. seasonal allergic rhinitis or perennial allergic rhinitis) or non-allergic rhinitis (e.g. vasomotor rhinitis)
  • rhinitis such as allergic rhinitis (e.g. seasonal allergic rhinitis or perennial allergic rhinitis) or non-allergic rhinitis (e.g. vasomotor rhinitis)
  • rhinitis such as allergic rhinitis (e.g. seasonal allergic rhinitis or perennial allergic rhin
  • PDE4 inhibitors are thought to be, or may be, effective in the treatment of rheumatoid arthritis and multiple sclerosis (e.g. see H.J. Dyke et al., Expert Opinion on Investigational Drugs, January 2002, 11(1), 1-13; C.Bumouf et al., Current Pharmaceutical Design, 2002, 8(14), 1255-1296; and A.M.Doherty, Current Opinion Chem. Biol., 1999, 3(4), 466-473; and references cited in these publications).
  • oral or parenteral administration is optionally used for rheumatoid arthritis.
  • PDE4 inhibitors have been suggested as having analgesic properties and thus being effective in the treatment of pain (A.Kumar et al., Indian J. Exp. Biol., 2000, 38(1), 26- 30).
  • the treatment and/or prophylaxis can be of cognitive impairment e.g. cognitive impairment in a neurological disorder (such as Alzheimer's disease or schizophrenia); and/or administration of the compound or salt can optionally be oral.
  • the treatment and/or prophylaxis can comprise cognitive enhancement e.g. in a neurological disorder.
  • cognition background see for example: H.T.Zhang et al. in: Psychopharmacology, June 2000, 150(3), 311-316 and Neuropsychopharmacology, 2000, 23(2), 198-204; and T. Egawa et al., Japanese J. Pharmacol., 1997, 75(3), 275- 81.
  • PDE4 inhibitors such as rolipram have been suggested as having antidepressant properties (e.g. J. Zhu et al., CNS Drug Reviews, 2001 , 7(4), 387-398; O'Donnell, Expert Opinion on Investigational Drugs, 2000, 9(3), 621-625; HT. Zhang et al.,
  • PDE4 inhibition has been suggested for the treatment of inflammatory bowel disease (e.g. ulcerative colitis and/or Crohn's disease), see K.H. Banner and M.A.Trevethick, Trends Pharmacol. Sci., August 2004, 25(8), 430-436.
  • inflammatory bowel disease e.g. ulcerative colitis and/or Crohn's disease
  • the compounds or salts of the present invention are usually administered as a pharmaceutical composition.
  • the present invention therefore provides in a further aspect a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof and one or more pharmaceutically acceptable carriers and/or excipients.
  • the pharmaceutical composition can be for use in the treatment and/or prophylaxis of any of the conditions described herein, for example chronic obstructive pulmonary disease (COPD) 1 asthma, rheumatoid arthritis, allergic rhinitis, psoriasis or atopic dermatitis in a mammal (e.g. human).
  • COPD chronic obstructive pulmonary disease
  • the invention also provides a method of preparing a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of formula (I), as herein defined, or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable carriers and/or excipients, the method comprising mixing the compound or salt with the one or more pharmaceutically acceptable carriers and/or excipients.
  • the invention also provides a pharmaceutical composition prepared by said method.
  • the compounds of formula (I) and/or the pharmaceutical composition may be administered, for example, by external topical (e.g. skin topical), oral, parenteral (e.g. intravenous, subcutaneous, or intramuscular), inhaled, or nasal administration.
  • external topical e.g. skin topical
  • oral e.g. parenteral
  • parenteral e.g. intravenous, subcutaneous, or intramuscular
  • inhaled administration involves topical administration to the lung e.g. by aerosol or dry powder composition.
  • the pharmaceutical composition can be suitable for (e.g. adapted for) external topical (e.g. skin topical), oral, parenteral (e.g. intravenous, subcutaneous, or intramuscular), inhaled, or nasal administration, e.g. to a mammal such as a human.
  • the pharmaceutical composition can for example be suitable for external topical (e.g. skin topical) or oral administration, e.g. to a mammal such as a human.
  • the pharmaceutical composition can optionally be in unit dose form.
  • the unit dose form can for example be:
  • a tablet or capsule for oral administration e.g. for oral administration to a human
  • a rupturable or peel-openable sealed dose container containing a dry powder inhalable pharmaceutical composition e.g. a plurality of which are usually disposed inside a suitable inhalation device
  • a vial, ampoule or filled syringe for parenteral administration e.g. comprising a solution or suspension of the compound or pharmaceutically acceptable salt in a suitable carrier such as an aqueous carrier or e.g. containing a lyophilised parenteral pharmaceutical composition (the vial or ampoule can optionally be manufactured using a blow-fill-seal process).
  • the composition can be in a form adapted for the administration of varying amounts of composition as desired by the user, such as a spreadable or sprayable external topical composition such as a cream, an ointment, a gel, or a liquid.
  • a spreadable or sprayable external topical composition such as a cream, an ointment, a gel, or a liquid.
  • compositions suitable for external topical administration are provided.
  • the pharmaceutical composition of the invention can for example be suitable for (e.g. adapted for) external topical administration (e.g. skin topical administration, i.e. topical administration to the skin), for example to a mammal such as a human.
  • external topical administration e.g. skin topical administration, i.e. topical administration to the skin
  • the pharmaceutical composition suitable for external topical administration can suitably be for the treatment and/or prophylaxis of atopic dermatitis in a mammal such as a human, e.g. by external topical administration.
  • External topical administration is defined above under the “medical uses” section.
  • External topical administration can for example be to those parts of the skin affected by or susceptible to the disease or condition e.g. atopic dermatitis, in particular in a mammal (e.g. human) suffering from or susceptible to atopic dermatitis.
  • An external-topical pharmaceutical composition e.g. skin topical pharmaceutical composition
  • An external-topical pharmaceutical composition e.g. skin topical pharmaceutical composition, of particular interest is an ointment.
  • the compound of formula (I) [ ⁇ /- ⁇ [1 ,6-diethyl-4-(tetrahydro-2H- pyran-4-ylamino)-1/-/-pyrazolo[3,4-6]pyridin-5-yl]methyl ⁇ -3-methyl-5- isoxazolecarboxamide] or the pharmaceutically acceptable salt thereof can be present in 0.1% to 10%, such as 0.2% to 10%, or 0.2% to 5%, or 0.5% to 5%, in particular 1% to 10% (e.g. about 2%, about 4% or about 6%), or 1% to 5% (e.g.
  • 1.5% to 5% or 1.5% to 5% such as about 2% or about 4%), or 0.5% to 3% (e.g. 0.5% or about 2%), or 1% to 3% (e.g. about 2%), by weight of the composition (w/w).
  • the compound of formula (I) or the pharmaceutically acceptable salt thereof can in particular comprise or be the compound (i.e. as the "free base” form).
  • the compound of formula (I) or the pharmaceutically acceptable salt thereof can optionally be in a particle-size-reduced form, for example obtained or obtainable by micronisation.
  • This can be, for example, for use in a pharmaceutical composition suitable for (e.g. adapted for) external topical (e.g. skin topical) administration. See the Particle size reduction sub-section herein, within the lnhalable pharmaceutical compositions section, for more details.
  • a preliminary screen which can aim to estimate roughly the aqueous solubility of a compound or salt of the invention, can include (as an approximate summary): (i) creating a ca. 1OmM solution of the compound in DMSO, (ii) diluting a portion of this DMSO solution by mixing about 19 parts by volume of pH 7.4 aqueous phosphate buffered saline (PBS) buffer with 1 part by volume of the ca. 1OmM DMSO solution, (iii) "filtering" the mixture with the aid of centrifugation, and then (iv) measuring the concentration of the dissolved compound in the "filtrate".
  • PBS aqueous phosphate buffered saline
  • ⁇ pophilicity The clogP (calculated Jog of the octanol/water partition coeficient (P)) of a particular compound or salt of the invention can estimate the lipophilicity of the compound or salt.
  • Solubilising and/or skin-penetration-enhancing agents An external-topical pharmaceutical composition, e.g. an ointment or an oil-in-water cream or water-in-oil cream, can for example include an agent which acts as a skin-penetration enhancer for and/or a solubiliser of the compound of formula (I) or the salt thereof.
  • the skin- penetration-enhancing- and/or solubilising- agent can for example be propylene glycol, diethylene glycol monoethyl ether (e.g. TRANSCUTOL TM) and/or caprylocaproyl macrogolglycerides (e.g. LABRASOL TM), in particular propylene glycol.
  • the solubiliser and/or skin-penetration enhancer suitably does not comprise DMSO.
  • the solubiliser and/or skin-penetration enhancer is in particular both a solubiliser and skin-penetration enhancer, and/or can be present in 0.5% to 50%, in particular 5% to 50%, for example 7% to 30%, such as 7% to 25%, e.g. about 10% to about 20% (e.g. about 10% or about 20%), by weight of the composition (w/w).
  • the skin-penetration enhancer is for delivery of the compound of formula (I) or salt thereof ("active agent" or "drug") through the skin. Solubilization of the drug also helps.
  • the solubilising and/or skin-penetration-enhancing agents should ideally (a) be safe and/or tolerable, (b) have as low a potential for skin irritancy as possible consistent with being an effective skin penetration enhancer, and (c) be compatibile with the active pharmaceutical ingredient.
  • the agent optionally functions both as a solubilising agent and a skin-penetration-enhancing agent.
  • An external-topical pharmaceutical composition e.g. an ointment or an oil- in-water cream or water-in-oil cream
  • a surfactant e.g. as an emulsifier
  • the total surfactant content can for example be 0.3% to 20%, e.g. 0.5% to 15% or 0.5% to 12% or 0.5% to 10% or 1 % to 12% or 3% to 10%, by weight of the composition (w/w).
  • the surfactant can for example comprise one or more of the following: a polyoxyl C-
  • 4_20 a 'kyl ether such as polyoxyl cetyl ether or polyoxyl stearyl ether
  • 4_20 a 'kyl ether such as polyoxyl cetyl ether or polyoxyl stearyl ether
  • glycerol monostearate e.g. Arlacel 165 TM
  • sorbitan monostearate e.g. Span 60 TM
  • sorbitan monostearate e.g. Span 60 TM
  • cetyl alcohol and/or stearyl alcohol e.g. wherein the total of any cetyl alcohol and any stearyl alcohol present is 0.1% to 15% w/w, e.g. 1% to 10% w/w such as about 2% to about 5% w/w
  • SDS sodium dodecyl sulphate
  • Polyoxyl stearyl ether (steareth) can e.g. be polyoxyl 2 stearyl ether (steareth 2) or polyoxyl 21 stearyl ether (steareth 21).
  • DMSO-containing solutions One possible external-topical pharmaceutical composition is a solution of the compound of formula (I) or the pharmaceutically acceptable salt thereof present at ca. 0.5% to ca. 2.5% w/w in a DMSO-containing solvent such as in DMSO/acetone or in DMSO/water; for example a solution of the compound or salt present at ca. 0.5% to ca. 2.5% w/w in DMSO/acetone (1:1).
  • DMSO-containing solutions often being capable of high skin penetration, are often good experimental preclinical formulations for use in animals e.g. pigs, but their likely skin irritancy generally make them less suitable for use in humans such as patients, e.g. atopic dermatitis patients.
  • An external-topical pharmaceutical composition can for example be an ointment or an oil-in-water cream or water-in-oil cream.
  • An ointment is of particular interest.
  • the ointment or cream typically contains an oil phase (oily ointment base).
  • the oil phase (oily ointment base) typically comprises an oil and/or a fat, for example of a consistency suitable for skin-spreadability.
  • the oil phase (oily ointment base) can for example comprise (e.g. be) an oil, wherein the oil comprises (e.g. is) white soft paraffin (white petrolatum) and/or a silicone oil and/or a mineral oil (such as liquid paraffin). (Mineral oil can also be used as a solubiliser and/or emollient).
  • the oil phase comprises (e.g. is) an oil, wherein the oil comprises (e.g. is) white soft paraffin (white petrolatum) and/or a silicone oil.
  • the external-topical pharmaceutical composition is an ointment
  • the oil phase (oily ointment base) comprises (e.g. is) an oil
  • the oil comprises (e.g. consists essentially of) white soft paraffin (white petrolatum) and a silicone oil.
  • the white soft paraffin (white petrolatum), e.g. in an ointment or cream, can be of various grades, for example (for Penreco supplier) Penreco Regent White TM grade, Penreco Snow White TM grade, or Penreco Ultima White TM grade; in particular high melting point white petrolatum (high melting point white soft paraffin) (e.g. of Penreco Ultima White TM grade).
  • the white petrolatum can be present at 25% to 99.9% w/w or 45% to 99.5% w/w or 50% to 99.5% w/w or 45% to 99% w/w or 50% to 99% w/w or 45% to 85% w/w or 45% to 75% w/w (i.e. by weight of the composition).
  • the silicone oil e.g. in an ointment or cream, in particular in an ointment, can for example be present at: 5% to 60 % w/w such as 5% to 50% w/w, in particular 10% to 50% w/w such as 15% to 40% w/w, suitably 20% to 35% w/w such as about 25% w/w (measured as the total silicone oil content, by weight of the composition).
  • the silicone oil can be solid or liquid.
  • the silicone oil e.g. in an ointment or cream, can for example comprise (e.g. be): decamethyl-cyclopentasiloxane (e.g. ST-Cyclomethicone 5-NF TM, available from Dow Corning), stearoxytrimethylsilane [Me(CH2)i7 ⁇ -SiMe3], polydimethylsiloxane (dimethicone), hexamethyldisiloxane (e.g. ca. 0.65 cSt viscosity at 25°C), octamethyltrisiloxane (e.g. ca.
  • the silicone oil e.g. in an ointment or cream, can in particular comprise (e.g. be): decamethyl-cyclopentasiloxane, stearoxytrimethylsilane [IvIe(CI- ⁇ )-17 ⁇ -SiMe3], or polydimethylsiloxane (dimethicone), or mixtures of any of the foregoing.
  • the silicone oil e.g. in an ointment or cream, can comprise (e.g. be) decamethyl- cyclopentasiloxane.
  • the decamethyl-cyclopentasiloxane can be ST-Cyclomethicone 5-NF TM, available from Dow Corning, and which is described by Dow Corning as being a polydimethyl- cyclosiloxane having a decamethyl-cyclopentasiloxane content of >95% and having a octamethyl-cyclotetrasiloxane content of ⁇ 1.0%.
  • the decamethyl-cyclopentasiloxane can for example be present at 5% to 60 % w/w such as 5% to 50% w/w, in particular 10% to 50% w/w such as 15% to 40% w/w, suitably 20% to 35% w/w such as about 25% w/w (i.e. by weight of the composition).
  • Stearoxytrimethylsilane [Me(CH2)i7 ⁇ -SiMe3] can for example be present as a mixture of stearoxytrimethylsilane and stearyl alcohol for example Silky Wax 10 TM which is available from Dow Corning.
  • Stearoxytrimethylsilane (and/or stearoxytrimethylsilane and stearyl alcohol mixture), e.g. in an ointment or cream e.g. ointment can for example be present at 1% to 30% w/w or 2% to 20% w/w or 5% to 20% w/w such as about 10% w/w.
  • Polydimethylsiloxane (dimethicone), whose structure is given in the Merck Index 12th edition 1996 as Me3Si-O-[-Si(CH3)2-O-] n -SiMe3, can for example have a viscosity at
  • polydimethylsiloxane can have a viscosity at 25 0 C of: 20 cSt ( ⁇ 10%) ("dimethicone 20"), 100 cSt ( ⁇ 5%), 350 cSt ( ⁇ 5%) ("dimethicone 350”), 1000 cSt ( ⁇ 5%), or 12500 cSt ( ⁇ 5%); grades of polydimethylsiloxane having these five different viscosities are available from Dow Corning as Q7-9120 TM Silicone Fluid.
  • Polydimethylsiloxane e.g. in an ointment, can e.g. be present at 0.1% to 15% w/w such as 0.5% to 10% w/w e.g. 0.5% to 5% w/w.
  • Microcrystalline wax or beeswax or beeswax substitute can alternatively or additionally be used as an oil / fat in the oil phase.
  • one or more fats like straight or branched chain mono- or di- alkyl esters such as isopropyl myristate, isopropyl palmitate, diisopropyl adipate, isocetyl stearate, isostearyl isostearate, decyl oleate, butyl stearate, 2-ethylhexyl palmitate, propylene glycol diester of coconut fatty acids, or a mixed ester of 2-ethyl hexanoic acid with a blend of cetyl or stearyl alcohols (e.g. known as Crodamol CAP) may be used in the oil phase (some of these are also solubilisers and/or surfactants). These may be used singly or in combination depending on the properties required.
  • solubilisers and/or surfactants e.g. known as Crodamol CAP
  • the oil phase (oily ointment base) can for example be present at 25% to 99.9% w/w or 25% to 99.5% w/w or 25% to 85% w/w (in particular 45% to 99.5% w/w or 45% to 99% w/w, or 50% to 99.5% w/w or 50% to 99% w/w or 50% to 80% w/w, or 70% to 99.5% w/w or 80% to 99.5% w/w) in an ointment (e.g. as an emulsion, or e.g. as a homogeneous single phase (which does not exclude the compound or salt being at least partly in suspension)).
  • an ointment e.g. as an emulsion, or e.g. as a homogeneous single phase (which does not exclude the compound or salt being at least partly in suspension).
  • the oil phase can for example be present at 25% to 85% w/w (e.g. 35% to 70% w/w) in an water-in-oil cream (e.g. emulsion), or at 8% to 55% w/w (e.g. 10% to 45% w/w) in an oil-in-water cream (e.g. emulsion).
  • oil phase oil phase
  • emulsion base water-in-oil cream
  • 8% to 55% w/w e.g. 10% to 45% w/w
  • oil-in-water cream e.g. emulsion
  • an external-topical pharmaceutical composition is an ointment comprising (e.g. consisting essentially of): - the compound of formula (I) [ ⁇ /- ⁇ [1 ,6-diethyl-4-(tetrahydro-2H-pyran ⁇ 4-ylamino)-1 H- pyrazolo[3,4-b]pyridin-5-yl]methyl ⁇ -3-methyl-5-isoxazolecarboxamide] or the pharmaceutically acceptable salt thereof present at 0.5% to 10% w/w (in particular 1 % to 10% w/w or 1% to 5% w/w, or 1% to 3% w/w, e.g. about 2% w/w);
  • - white petrolatum present at 25% to 99.9% w/w, in particular 45% to 99.5% w/w or 50% to 99.5% w/w or 45% to 99% w/w or 50% to 99% w/w or 45% to 85% w/w or 45% to 75% w/w (e.g. about 60-85% w/w, e.g. about 73-75% w/w) (i.e. by weight of the composition); and
  • a silicone oil present at: 5% to 60 % w/w such as 5% to 50% w/w, in particular 10% to 50% w/w such as 15% to 40% w/w, suitably 20% to 35% w/w such as about 25% w/w (measured as the total silicone oil content, by weight of the composition).
  • an external-topical pharmaceutical composition is an ointment comprising (e.g. consisting essentially of):
  • - white petrolatum present at 25% to 99.9% w/w, in particular 45% to 99.5% w/w or 50% to 99.5% w/w or 45% to 99% w/w or 50% to 99% w/w or 45% to 85% w/w or 45% to 75% w/w (e.g. about 60-85% w/w, e.g. about 73-75% w/w) (i.e. by weight of the composition); and
  • - decamethyl-cyclopentasiloxane e.g. (e.g. ST-Cyclomethicone 5-NF TM) present at 5% to 60 % w/w such as 5% to 50% w/w, in particular 10% to 50% w/w such as 15% to 40% w/w, suitably 20% to 35% w/w such as about 25% w/w (i.e. by weight of the composition).
  • an external-topical pharmaceutical composition is an ointment comprising: - the compound of formula (I) or the pharmaceutically acceptable salt thereof present at 0.2% to 5% w/w (e.g. 0.5% to 5% w/w, or 1% to 3% w/w, e.g. about 2% w/w);
  • an oil phase (oily ointment base) present at 25% to 99% w/w or 50% to 99% w/w or 25% to 85% w/w or 50% to 80% w/w
  • the oil phase can comprise white petrolatum present at 25 to 75% w/w or 45 to 75% w/w, and optionally also comprising mineral oil (e.g. as solubiliser and emollient) present at 2.5% to 15% w/w such as 4% to 12% w/w);
  • one or more surfactants e.g. polyoxyl stearyl ether
  • - one or more agents acting as a skin-penetration enhancer in particular acting as both a solubiliser and skin-penetration enhancer and/or in particular a hydrophilic skin- penetration enhancer such as propylene glycol
  • a skin-penetration enhancer in particular acting as both a solubiliser and skin-penetration enhancer and/or in particular a hydrophilic skin- penetration enhancer such as propylene glycol
  • a skin-penetration enhancer in particular acting as both a solubiliser and skin-penetration enhancer and/or in particular a hydrophilic skin- penetration enhancer such as propylene glycol
  • antioxidants e.g. butylated hydroxyanisole
  • preservatives e.g. present in total at 0.01 to 4% w/w such as 0.05 to 1% w/w (e.g. methylparaben present at 0.05 to 2% w/w and/or propylparaben present at 0.01 to 2% w/w).
  • the above example composition can optionally be a homogeneous single phase.
  • the oil phase e.g. when using propylene glycol or another hydrophilic solubiliser and penetration enhancer
  • the oil phase (oily ointment base) and a hydrophilic phase containing the hydrophilic solubiliser and penetration enhancer e.g. propylene-glycol-containing phase
  • a hydrophilic phase containing the hydrophilic solubiliser and penetration enhancer e.g. propylene-glycol-containing phase
  • Ointment compositions having two phases can optionally be prepared using an emulsification process whereby the hydrophilic phase (e.g. propylene-glycol-containing phase) and oil phase are first prepared in separate vessels.
  • the hydrophilic phase can optionally contain a penetration enhancer such as propylene glycol, and optionally some or all of the compound of formula (I) or salt thereof.
  • the oil phase can optionally contain a surfactant.
  • Temperatures of both phases are maintained at elevated temperatures, such as about 45-9O 0 C or about 45-8O 0 C or about 55-9O 0 C or about 55-8O 0 C (e.g. about 60-65 0 C), or from above 70 to 90 0 C, the oil phase temperature being sufficiently high (e.g.
  • ointment emulsion is allowed to cool, e.g. to about 15-35 0 C such as to about 17-3O 0 C, in particular while the agitation continues e.g. at lower speeds.
  • the ointment emulsion can then optionally be dispensed from the manufacturing vessel and filled into primary packaging, for example tubes or sachets.
  • an ointment can comprise a polyethylene glycol base, e.g. present at 25 to 98% w/w such as 50 to 95% w/w, instead of or as well as an oily ointment base.
  • a polyethylene glycol base e.g. present at 25 to 98% w/w such as 50 to 95% w/w, instead of or as well as an oily ointment base.
  • An external-topical pharmaceutical composition can be a cream, e.g. a water- in-oil cream or an oil-in-water cream.
  • Water-in-oil creams usually have an increased aqueous content compared to ointments.
  • the water-in-oil cream can be a water-in-oil cream emulsion. That is, in particular, in the water-in-oil cream, an oil phase and an aqueous phase can have been emulsified to form a water-in-oil cream emulsion.
  • an external-topical pharmaceutical composition is a water-in-oil cream (e.g. cream emulsion) comprising:
  • the compound of formula (I) or pharmaceutically acceptable salt thereof present at 0.2% to 10% w/w or 0.2% to 5% w/w (in particular 0.5% to 10% w/w or 1% to 10% w/w, such as 0.5% to 5% w/w or 1% to 5% w/w, or 1% to 3% w/w, e.g. about 2% w/w);
  • oil phase oil phase (oily ointment base) present at 25% to 85% w/w or 35% to 70% w/w (for example comprising white petrolatum present at 25% to 75% w/w or 30% to 65% w/w
  • mineral oil e.g. as solubiliser and emollient
  • mineral oil e.g. as solubiliser and emollient
  • - water present at 2% to 30% w/w, e.g. 5% to 25% or 10% to 22% w/w (e.g. about 20% w/w); - one or more surfactants (e.g. polyoxyl stearyl ether such as polyoxyl 2 stearyl ether) present in total at 0.5% to 12% w/w, such as 3% to 10% w/w (e.g. about 8% w/w); and
  • surfactants e.g. polyoxyl stearyl ether such as polyoxyl 2 stearyl ether
  • one or more agents acting as a skin-penetration enhancer in particular acting as both a solubiliser and skin-penetration enhancer and/or in particular a hydrophilic skin- penetration enhancer such as propylene glycol) present in total at 0.5% to 50% w/w, such as 5% to 50% w/w or 7% to 30% w/w (e.g. about 20% w/w, e.g. about 20% w/w of propylene glycol).
  • the above water-in-oil cream can also optionally comprise:
  • antioxidants e.g. butylated hydroxyanisole
  • preservatives e.g. present in total at 0.01 to 4% w/w such as 0.05 to 1 % w/w (e.g. methylparaben present at 0.05 to 2% w/w and/or propylparaben present at 0.01 to 2% w/w).
  • Oil-in-water creams usually have an increased aqueous content compared to ointments and water-in-oil creams.
  • the oil-in-water cream can be an oil-in- water cream emulsion. That is, in particular, in the oil-in-water cream, an oil phase and an aqueous phase can have been emulsified to form an oil-in-water cream emulsion.
  • Oil-in-water creams can for example be high-occlusion creams, wherein, after topical administration to the skin, moisture loss from the skin and/or from the cream is reduced or limited by means of sufficiently high coverage of the skin and/or by providing a sufficient barrier at the site of application.
  • An oil-in-water cream can in particular contain one or more emollients (hydrating agents), such as silicones (e.g. dimethicone, e.g. dimethicone 360 or dimethicone 20), a high-viscosity wax such as microcrystalline wax, and/or mineral oil.
  • emollients such as silicones (e.g. dimethicone, e.g. dimethicone 360 or dimethicone 20), a high-viscosity wax such as microcrystalline wax, and/or mineral oil.
  • silicones e.g. dimethicone, e.g. dimethicone 360 or dimethicone 20
  • a high-viscosity wax such as microcrystalline wax
  • mineral oil suitably there is a sufficiently high water content, for example wherein the water is present in 15% to 60% w/w, 20% to 50% w/w, or 25% to 40% w/w.
  • an external-topical pharmaceutical composition is a oil-in-water cream (e.g. cream emulsion) comprising:
  • the compound of formula (I) or pharmaceutically acceptable salt thereof present at 0.2% to 10% w/w or 0.2% to 5% w/w (in particular 0.5% to 10% w/w or 1% to 10% w/w, such as 0.5% to 5% w/w or 1% to 5% w/w, or 1% to 3% w/w, e.g. about 2% w/w);
  • oil phase containing one or more ingredients capable of acting as emollients, the oil phase being present at 5% to 60% w/w or in particular 20% to 60% w/w or 30% to 60% w/w such as 30% to 55% w/w;
  • one or more surfactants e.g. polyoxyl stearyl ether such as polyoxyl 2 stearyl ether
  • surfactants e.g. polyoxyl stearyl ether such as polyoxyl 2 stearyl ether
  • one or more agents acting as a skin-penetration enhancer in particular acting as both a solubiliser and skin-penetration enhancer and/or in particular a hydrophilic skin- penetration enhancer such as propylene glycol) present in total at 0.5% to 50% w/w, in particular 5% to 50% w/w or 7% to 25% w/w (e.g. about 20% w/w, e.g. about 20% w/w of propylene glycol).
  • the above oil-in-water cream can also optionally comprise:
  • solubilisers e.g. isopropyl myristate
  • buffers e.g. citric acid and/or dibasic sodium phosphate
  • the oil phase can in particular comprise mineral oil (e.g. as emollient and solubiliser) present at 15% to 50% w/w or 20% to 45% w/w, and/or can in particular comprise a high-viscosity wax such as microcrystalline wax (e.g. as emollient) present at 5% to 25% w/w such as 8% to 15% w/w, and/or can in particular comprise a silicone (such as dimethicone e.g. dimethicone 360 or dimethicone 20, e.g. as emollient) present at 0.5% to 20% such as 0.5% to 10% or 1% to 5% w/w.
  • mineral oil e.g. as emollient and solubiliser
  • a high-viscosity wax such as microcrystalline wax (e.g. as emollient) present at 5% to 25% w/w such as 8% to 15% w/w
  • a silicone such as dimethicone e.g
  • the one or more surfactants can for example comprise: glycerol monostearate present at 0.5% to 10% w/w, and/or sorbitan monostearate present at 0.05% to 10% w/w, and/or [cetyl alcohol and/or stearyl alcohol] present in total at 0.1% to 15% or 1 to 10% w/w.
  • Cream emulsions e.g. water-in-oil or oil-in-water cream emulsions
  • an aqueous phase is prepared, e.g. prepared before emulsification.
  • the aqueous phase usually contains water and a solubiliser and/or skin- penetration enhancer such as propylene glycol, and optionally contains some or all of the compound of formula (I) or salt thereof, and/or optionally contains surfactant.
  • the oil phase e.g. containing white petrolatum and/or mineral oil, and/or optionally containing surfactant, can be prepared in a separate vessel.
  • Temperatures of both phases are suitably maintained at (or heated to) elevated temperatures, such as about 45-9O 0 C or about 45-8O 0 C or about 45-75 0 C, for example about 55-90 0 C or about 55-8O 0 C or about 55-75 0 C (in particular at about 60-65 0 C), or e.g. from above 70 to 90 0 C, the oil phase temperature being sufficiently high (e.g. about 45-9O 0 C or about 55-9O 0 C or from above 70 to 90 0 C) to melt the oil phase. While hot, one phase is suitably added to another while mixing, e.g.
  • the resulting emulsion is typically allowed to cool, e.g. to about 15-35 0 C such as to about 17-3O 0 C (e.g. to about 17-22 0 C) or to about 18-3O 0 C, for example while the agitation continues e.g. at lower speeds.
  • the cream emulsion can then optionally be dispensed from the manufacturing vessel and filled into primary packaging, for example tubes or sachets.
  • a pharmaceutical composition of the invention suitable for external topical administration can be administered once daily, twice daily or more than twice daily, to external body part(s), e.g. on the skin such as at a site of diseased skin, e.g. skin suffering from atopic dermatitis.
  • compositions suitable for oral or parenteral administration are provided.
  • a pharmaceutical composition suitable for oral administration can be liquid or solid; for example it can be a syrup, suspension or emulsion, a tablet, a capsule or a lozenge.
  • a liquid formulation can generally consist of a suspension or solution of the compound or pharmaceutically acceptable salt in a suitable pharmaceutically acceptable liquid carriers), for example an aqueous solvent such as water, ethanol or glycerine, or a non-aqueous solvent, such as polyethylene glycol or an oil.
  • a suitable pharmaceutically acceptable liquid carriers for example an aqueous solvent such as water, ethanol or glycerine, or a non-aqueous solvent, such as polyethylene glycol or an oil.
  • the formulation may also contain a suspending agent, preservative, flavouring and/or colouring agent.
  • the pharmaceutical composition is in unit dose form, such as a tablet or capsule for oral administration, e.g. for oral administration to a human.
  • a pharmaceutical composition suitable for oral administration being a tablet can comprise one or more pharmaceutically acceptable carriers and/or excipients suitable for preparing tablet formulations.
  • the carrier can for example be or include lactose, cellulose (for example microcrystalline cellulose), or mannitol.
  • the tablet can also or instead contain one or more pharmaceutically acceptable excipients, for example a binding agent such as hydroxypropylmethylcellulose or povidone (polyvinylpyrrolidone), a lubricant e.g. an alkaline earth metal stearate such as magnesium stearate, and/or a tablet disintegrant such as sodium starch glycollate, croscarmellose sodium, or crospovidone (cross-linked polyvinylpyrrolidone).
  • a binding agent such as hydroxypropylmethylcellulose or povidone (polyvinylpyrrolidone)
  • a lubricant e.g. an alkaline earth metal stearate such as magnesium stearate
  • the pharmaceutical composition being a tablet can be prepared by a method comprising the steps of: (i) mixing the compound of formula (I), as herein defined, or a pharmaceutically acceptable salt thereof, with the one or more pharmaceutically acceptable carriers and/or excipients, (ii) compressing the resulting mixture (which is usually in powder form) into tablets, and (iii) optionally coating the tablet with a tablet film-coating material.
  • a pharmaceutical composition suitable for oral administration being a capsule can be prepared using encapsulation procedures.
  • pellets or powder containing the active ingredient can be prepared using a suitable pharmaceutically acceptable carrier and then filled into a hard gelatin capsule.
  • a dispersion or suspension can be prepared using any suitable pharmaceutically acceptable carrier, for example an aqueous gum or an oil and the dispersion or suspension then filled into a soft gelatin capsule.
  • the compound of formula (I) or the pharmaceutically acceptable salt thereof can in one embodiment comprise (e.g. be) the compound of the invention (i.e. as the "free base” form).
  • a pharmaceutical composition suitable for (e.g. adapted for) parenteral (e.g. intravenous, subcutaneous, or intramuscular) administration can comprise a solution or suspension of the compound or pharmaceutically acceptable salt in a sterile pharmaceutically acceptable aqueous carrier (e.g. sterile water) or parenterally acceptable oil.
  • a sterile pharmaceutically acceptable aqueous carrier e.g. sterile water
  • parenterally acceptable oil e.g. sterile water
  • the solution can be lyophilised.
  • a lyophilised pharmaceutical composition suitable for (e.g. adapted for) parenteral administration may, in use, optionally be reconstituted with a suitable solvent, e.g. sterile water or a sterile parenterally acceptable aqueous solution, just prior to administration.
  • compositions suitable for inhalable or intranasal administration, and particle-size reduction are provided.
  • compositions suitable for (e.g. adapted for) nasal or inhaled administration may conveniently be formulated as aerosols, drops, gels or dry powders.
  • Aerosol formulations can comprise a solution or fine suspension of the active substance in a pharmaceutically acceptable aqueous or nonaqueous solvent. Aerosol formulations can be presented in single or multidose quantities in sterile form in a sealed container, which can take the form of a cartridge or refill for use with an atomising device or inhaler. Alternatively the sealed container may be a unitary dispensing device such as a single dose nasal inhaler or an aerosol dispenser fitted with a metering valve (metered dose inhaler) which is intended for disposal once the contents of the container have been exhausted.
  • a metering valve metered dose inhaler
  • the dosage form comprises an aerosol dispenser
  • it can contain a suitable propellant under pressure such as compressed air, carbon dioxide, or an organic propellant such as a chlorofluorocarbon (CFC) or hydrofluorocarbon (HFC).
  • CFC chlorofluorocarbon
  • HFC hydrofluorocarbon
  • Suitable CFC propellants include dichlorodifluoromethane, trichlorofluoromethane and dichlorotetrafluoroethane.
  • Suitable HFC propellants include 1 ,1 ,1 ,2,3,3,3- heptafluoropropane and 1 ,1 ,1 ,2-tetrafluoroethane.
  • the aerosol dosage forms can also take the form of a pump-atomiser.
  • compositions suitable e.g. adapted for) inhaled administration
  • the compound or salt of formula (I) can be in a particle-size-reduced form.
  • the size-reduced form can for example be obtained or obtainable by micronisation. Micronisation usually involves subjecting the compound/salt to collisional and/or abrasional forces in a fast-flowing circular or spiral/vortex-shaped airstream often including a cyclone component.
  • the particle size of the size-reduced (e.g. micronised) compound or salt can be defined by a D50 value of about 0.5 to about 10 microns, e.g. about 1 to about 7 microns or about 1 to about 5 microns (e.g.
  • the compound or salt of formula (I) can have a particle size defined by: a D10 of about 0.3 to about 3 microns (e.g. about 0.5 to about 2 microns, or about 1 micron), and/or a D50 of about 0.5 to about 10 microns or about 1 to about 7 microns or (e.g. about 1 to about 5 microns or about 2 to about 5 microns or about 2 to about 4 microns), and/or a D90 of about 1 to about 30 microns or about 2 to about 20 microns or about 2 to about 15 microns or about 3 to about 15 microns (e.g. about 5 to about 15 microns or about 5 to about 10 microns or about 2 to about 10 microns); for example as measured using laser diffraction.
  • a D10 of about 0.3 to about 3 microns e.g. about 0.5 to about 2 microns, or about 1 micron
  • D90, D50 and D10 respectively mean that 90%, 50% and 10% of the material is less than the micron size specified.
  • D50 is the median particle size.
  • DV90, DV50 and DV10 respectively mean that 90%, 50% and 10% by volume of the material is less than the micron size specified.
  • DM90, DM50 and DM10 respectively mean that 90%, 50% and 10% by weight of the material is less than the micron size specified.
  • Laser diffraction measurement of particle size can use a dry method (wherein a suspension of the compound/salt in an airflow crosses the laser beam) or a wet method [wherein a suspension of the compound/salt in a liquid dispersing medium, such as isooctane or (e.g. if compound is soluble in isooctane) 0.1 % Tween 80 in water, crosses the laser beam].
  • particle size can for example be calculated using the Fraunhofer calculation; and/or optionally a Malvern Mastersizer or Sympatec apparatus is used for measurement.
  • particle size measurement and/or analysis by laser diffraction can use any or all of (e.g.
  • a Malvern Mastersizer longbed version a dispersing medium of 0.1 % Tween 80 in water, a stir rate of ca. 1500 rpm, ca. 3 mins sonification prior to final dispersion and analysis, a 300 RF (Reverse Fourier) lens, and/or the Fraunhofer calculation with Malvern software.
  • Micronisation Example Micronisation of a compound or salt of one of the Examples
  • the parent (unmicronised) and micronised materials are analyzed for particle size by laser diffraction and crystallinity by PXRD.
  • Jetpharma MC1 Micronizer Nitrogen supply Air tank with 275psi rate tubing
  • the Jetpharma MC1 Micronizer comprises a horizontal disc-shaped milling housing having: a tubular compound inlet (e.g. angled at ca. 30 degrees to the horizontal) for entry of a suspension of unmicronised compound of formula (I) or salt in a gasflow, a separate gas inlet for entry of gases, a gas outlet for exit of gases, and a collection vessel (micronizer container) for collecting micronised material.
  • the milling housing has two chambers: (a) an outer annular chamber in gaseous connection with the gas inlet, the chamber being for receiving pressurised gas (e.g.
  • the annular wall (ring R) has a plurality of narrow-bored holes connecting the inner and outer chambers and circumferentially-spaced-apart around the annular wall.
  • the holes opening into the inner chamber are directed at an angle (directed part-way between radially and tangentially), and in use act as nozzles directing pressurised gas at high velocity from the outer chamber into the inner chamber and in an inwardly-spiral path (vortex) around the inner chamber (cyclone).
  • the compound inlet is in gaseous communication with the inner chamber via a nozzle directed tangentially to the inner chamber, within and near to the annular wall / ring R.
  • Upper and lower broad-diameter exit vents in the central axis of the inner milling chamber connect to (a) (lower exit) the collection vessel which has no air outlet, and (b) (upper exit) the gas outlet.
  • a venturi inlet (V) Inside and coaxial with the tubular compound inlet and longitudinally-movable within it is positioned a venturi inlet (V) for entry of gases.
  • the compound inlet also has a bifurcation connecting to an upwardly-directed material inlet port for inputting material.
  • the narrow head of the venturi inlet (V) is suitably positioned below and slightly forward of the material inlet port, so that when the venturi delivers pressurised gas (e.g. air or nitrogen) the feed material is sucked from the material inlet port into the gas stream through the compound inlet and is accelerated into the inner milling chamber tangentially at a subsonic speed. Inside the milling chamber the material is further accelerated to a supersonic speed by the hole/nozzle system around the ring (R ) (annular wall) of the milling chamber. The nozzles are slightly angled so that the acceleration pattern of the material is in the form of an inwardly-directed vortex or cyclone.
  • pressurised gas e.g. air or nitrogen
  • the material inside the milling chamber circulates rapidly and particle collisions occur during the process, causing larger particles to fracture into smaller ones.
  • "Centrifugal" acceleration in the vortex causes the larger particles to remain at the periphery of the inner chamber while progressively smaller particles move closer to the centre until they exit the milling chamber, generally through the lower exit, at low pressure and low velocity.
  • the particles that exit the milling chamber are heavier than air and settle downward thorugh the lower exit into the collection vessel (micronizer container), while the exhaust gas rises (together with a minority of small particles of micronised material) and escapes into the atmosphere at low pressure and low velocity.
  • the micronizer is assembled.
  • the narrow head of the venturi inlet is positioned below and slightly forward of the material inlet port and is measured with a micro-caliper to make sure that it is inserted correctly.
  • the ring (R ) and venturi (V) pressures are adjusted according to the values specified in the experimental design (refer to experimental section below) by adjusting the valves on the pressure gauges on the micronizer.
  • the setup is checked for leakage by observing if there is any fluctuation in the reading of the pressure gauges.
  • venturi (V) pressure is kept at least 2 bars greater than the ring (R ) pressure to prevent regurgitation of material, e.g. outwardly from the material inlet port.
  • Balance performance is checked with calibration weights. Specified amount of the parent material is fed into the input container of the micronizer using a spatula. The input container plus material is weighed. The equipment pressure is monitored during the micronization process.
  • the nitrogen supply is shut off and the micronised material is allowed to settle into the micronizer container.
  • the micronised powder in the micronizer container (collection vessel) and the cyclone (above the recovery vessel) are collected together into a pre-weighed and labelled collection vial. The weight of the micronised material is recorded.
  • the input container is re-weighed in order to calculate the amount of input material by difference.
  • the micronizer is disassembled and residual PDE4 compound on the micronizer inner surface is rinsed with 70/30 isopropyl alcohol / water and collected into a flask. The micronizer is then thoroughly cleaned in a Lancer washing machine and dried before subsequent runs are performed.
  • Procedure 1 can optionally be carried out generally using a procedure and an apparatus generally as described above or similar to those described, using generally the following experimental parameters:
  • % yield [(Material from collection vessel + Material from cyclone) / Material input amount] x100.
  • Procedure 2 Optional Experimental Parameters
  • the optional experimental parameters can for example be as follows: Material Venturi Intended
  • the pharmaceutical composition may for example be a dry powder inhalable composition.
  • a dry powder inhalable composition can comprise a powder base such as lactose or starch, the compound of formula (I) or salt thereof (suitably in particle-size-reduced form, e.g. in micronised form), and optionally a ternary agent such as L-leucine, mannitol, trehalose, magnesium stearate and/or cellobiose octaacetate (e.g. alpha-D-isomer of cellobiose octaacetate, e.g. available from Aldrich).
  • cellobiose octaacetate and storage stability see WO 03/088943.
  • the dry powder inhalable composition can comprise a dry powder blend of lactose and the compound of formula (I) or salt thereof.
  • the lactose can be lactose hydrate e.g. lactose monohydrate and/or can be inhalation-grade and/or fine-grade lactose.
  • the particle size of the lactose can for example be defined by 90% or more (by weight or by volume) of the lactose particles being less than 1000 microns (micrometres) (e.g. 10-1000 microns e.g. 30-1000 microns) in diameter, and/or 50% or more of the lactose particles being less than 500 microns (e.g. 10-500 microns) in diameter.
  • the particle size of the lactose can for example be defined by 90% or more of the lactose particles being less than 300 microns (e.g. 10-300 microns e.g. 50-300 microns) in diameter, and/or
  • the particle size of the lactose can be defined by 90% or more of the lactose particles being less than 100-200 microns in diameter, and/or 50% or more of the lactose particles being less than 40-70 microns in diameter. It can be about 3 to about 30% (e.g. about 10%) (by weight or by volume) of the particles are less than 50 microns or less than 20 microns in diameter.
  • a suitable inhalation-grade lactose is E9334 lactose (10% fines) (Borculo Domo Ingredients, Hanzeplein 25, 8017 JD Zwolle, Netherlands).
  • the compound of formula (I) or salt thereof can for example be present in about 0.1% to about 70% (e.g. about 1% to about 50%, e.g. about 5% to about 40%, e.g. about 20 to about 30%) by weight of the composition.
  • the dry powder blend is, for example, prepared by mixing the required amount of the compound/salt (e.g. 10 mg, 1% w/w) with inhalation-grade lactose containing 10% fines (e.g. 990 mg, 99% w/w) in a TeflonTM (polytetrafluoroethene) pot in a Mikro-dismembrator ball-mill (but without a ball bearing) at 3 A speed (ca. 2000-2500 rpm) for about 4 hours at each blend concentration.
  • the Mikro-dismembrator available from B.
  • blends can include: 10% w/w compound/salt (50 mg) + 90% w/w lactose (450 mg, inhalation-grade lactose containing 10% fines).
  • Serial dilution of the 1% w/w blend can achieve e.g. 0.1% and 0.3% w/w blends.
  • a pharmaceutical composition for inhaled administration can be incorporated into a plurality of sealed dose containers (e.g. containing the dry powder composition) mounted longitudinally in a strip or ribbon inside a suitable inhalation device.
  • the container can be rupturable or peel- openable on demand and the dose, e.g. of the dry powder composition, can be administered by inhalation via a device such as the DISKUS TM device, marketed by GlaxoSmithKline.
  • the DISKUS TM inhalation device can e.g. be substantially as described in GB 2,242,134 A.
  • At least one container for the pharmaceutical composition in powder form (the at least one container in particular being a plurality of sealed dose containers mounted longitudinally in a strip or ribbon) is defined between two members peelably secured to one another; the device comprises: means defining an opening station for the said at least one container; means for peeling the members apart at the opening station to open the container; and an outlet, communicating with the opened container, through which a user can inhale the pharmaceutical composition in powder form from the opened container.
  • a pharmaceutical composition suitable for (e.g. adapted for) external topical administration e.g. an ointment or an oil-in-water or water-in-oil composition
  • the compound of formula (I) or the pharmaceutically acceptable salt thereof can be present in 0.1% to 10%, such as 0.2% to 10% or 0.2% to 5%, or 0.5% to 10% or 0.5% to 5%, or 1 % to 10% or 1 % to 5%, or 0.5% to 3%, or 1 % to 3% (e.g. about 0.5% or in particular about 2%), by weight of the composition (w/w).
  • an external-topical pharmaceutical composition can be administered once daily, twice daily or more than twice daily, to external body part(s), e.g. to the skin such as at a site of diseased skin. The amount administered is usually such as substantially to cover the site(s) of diseased skin.
  • a pharmaceutical composition of the invention can e.g. be in unit dose form such as a tablet or capsule for oral administration, e.g. for oral administration to a human.
  • a or each dosage unit for oral or parenteral administration can for example contain from 0.01 to 3000 mg, such as 0.5 to 1000 mg, of a compound of the formula (I) or a pharmaceutically acceptable salt thereof, calculated as the free base.
  • a or each dosage unit for nasal or inhaled administration can for example contain from 0.001 to 50 mg, such as 0.01 to 5 mg, of a compound of the formula (I) or a pharmaceutically acceptable salt thereof, calculated as the free base.
  • a pharmaceutically acceptable compound or salt of the invention can for example be administered to a mammal (e.g. human) in a daily oral or parenteral dose of 0.001 mg to 50 mg per kg body weight per day (mg/kg/day), for example 0.01 to 20 mg/kg/day or 0.03 to 10 mg/kg/day or 0.1 to 2 mg/kg/day, of the compound of the formula (I) or a pharmaceutically acceptable salt thereof, calculated as the free base.
  • a pharmaceutically acceptable compound or salt of the invention can for example be administered to a mammal (e.g. human) in a daily nasal or inhaled dose of: 0.0001 to 5 mg/kg/day or 0.0001 to 1 mg/kg/day, e.g. 0.001 to 1 mg/kg/day or 0.001 to 0.3 mg/kg/day or 0.001 to 0.1 mg/kg/day or 0.005 to 0.3 mg/kg/day, of the compound of the formula (I) or a pharmaceutically acceptable salt thereof, calculated as the free base.
  • the pharmaceutically acceptable compounds or salts of the invention can for example be administered to a human in a daily dose (for an adult patient) of, for example, an oral or parenteral dose of 0.01 mg to 3000 mg per day or 0.5 to 1000 mg per day e.g. 2 to 500 mg per day, or a nasal or inhaled dose of 0.001 to 50 mg per day or 0.01 to 30 mg per day or 0.01 to 5 mg per day or 0.02 to 2 mg per day, of the compound of the formula (I) or a pharmaceutically acceptable salt thereof, calculated as the free base.
  • a daily dose for an adult patient
  • an oral or parenteral dose 0.01 mg to 3000 mg per day or 0.5 to 1000 mg per day e.g. 2 to 500 mg per day
  • a nasal or inhaled dose of 0.001 to 50 mg per day or 0.01 to 30 mg per day or 0.01 to 5 mg per day or 0.02 to 2 mg per day, of the compound of the formula (I) or a pharmaceutically acceptable salt thereof, calculated as the free base
  • the compounds, salts and/or pharmaceutical compositions according to the invention may also be used in combination with another therapeutically active agent, for example, a ⁇ 2 adrenoreceptor agonist, an anticholinergic compound (e.g. muscarinic (M) receptor antagonist), an anti-histamine, an anti-allergic, an anti-inflammatory agent, an antiinfective agent or an immunosuppressant.
  • a ⁇ 2 adrenoreceptor agonist e.g. muscarinic (M) receptor antagonist
  • an anti-histamine e.g. muscarinic (M) receptor antagonist
  • an anti-histamine e.g. muscarinic (M) receptor antagonist
  • an anti-histamine e.g. muscarinic (M) receptor antagonist
  • an anti-histamine e.g. muscarinic (M) receptor antagonist
  • an anti-histamine e.g. muscarinic (M) receptor antagonist
  • the invention thus provides, in a further aspect, a combination comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof together with another therapeutically active agent, for example, a ⁇ 2 -adrenoreceptor agonist, an anti-histamine, an anti-allergic, an anti-inflammatory agent, an antiinfective agent or an immunosuppressant.
  • a ⁇ 2 -adrenoreceptor agonist for example, a ⁇ 2 -adrenoreceptor agonist, an anti-histamine, an anti-allergic, an anti-inflammatory agent, an antiinfective agent or an immunosuppressant.
  • the ⁇ 2 -adrenoreceptor agonist is salmeterol (e.g. as racemate or a single enantiomer such as the R-enantiomer), salbutamol, formoterol, salmefamol, fenoterol or terbutaline, or a salt thereof (e.g.
  • ⁇ 2 -adrenoreceptor agonists are optionally used, especially those having a therapeutic effect over a 12-24 hour period such as salmeterol or formoterol.
  • the ⁇ 2 -adrenoreceptor agonist is for inhaled administration, e.g. once per day and/or for simultaneous inhaled administration; and in particular the ⁇ 2 -adrenoreceptor agonist can be in particle-size-reduced form e.g. as defined herein.
  • the ⁇ 2 -adrenoreceptor agonist combination can for example be for treatment and/or prophylaxis of COPD or asthma.
  • Salmeterol or a pharmaceutically acceptable salt thereof, e.g. salmeterol xinofoate, is suitably administered to humans at an inhaled dose of 25 to 50 micrograms twice per day (measured as the free base).
  • Examples of long acting ⁇ 2 -adrenoreceptor agonists which could be used include those described in WO 02/066422A, WO 03/024439, WO 02/070490 and WO 02/076933.
  • ⁇ 2 -adrenoreceptor agonists disclosed in WO 02/066422 include: 3-(4- ⁇ [6-( ⁇ (2R)-2-hydroxy-2-[4-hydroxy-3-(hydroxymethyl)- phenyl]ethyl ⁇ amino)hexyl]oxy ⁇ butyl)benzenesulfonamide and 3-(3- ⁇ [7-( ⁇ (2R)-2-hydroxy-2-[4-hydroxy-3-hydroxymethyl)phenyl]ethyl ⁇ - amino)heptyl]oxy ⁇ propyl)benzenesulfonamide.
  • ⁇ 2 -adrenoreceptor agonist disclosed in WO 03/024439 is:
  • An anti-histamine usable in a combination of a compound of formula (I) or salt can for example be for oral administration (e.g. as a combined composition such as a combined tablet), and can be for treatment and/or prophylaxis of allergic rhinitis.
  • antihistamines include methapyrilene, or H1 antagonists such as cetirizine, loratadine (e.g. Clarityn TM) 1 desloratadine (e.g. Clarinex TM) or fexofenadine (e.g. Allegra TM)
  • the invention also provides, in a further aspect, a combination comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof together with an anticholinergic compound, e.g. a muscarinic (M) receptor antagonist such as an M-] , M2,
  • an anticholinergic compound e.g. a muscarinic (M) receptor antagonist such as an M-] , M2,
  • /M2, or M3 receptor antagonist in particular a M3 receptor antagonist, such as a M3 receptor antagonist which selectively antagonises (e.g. antagonises 10 times or more strongly) the M3 receptor over the Mi and/or M2 receptor.
  • a M3 receptor antagonist such as a M3 receptor antagonist which selectively antagonises (e.g. antagonises 10 times or more strongly) the M3 receptor over the Mi and/or M2 receptor.
  • PDE4 inhibitors see for example WO 03/011274 A2 and WO 02/069945 A2 / US 2002/0193393 A1 and US 2002/052312 A1 , and some or all of these publications give examples of anticholinergic compounds / muscarinic (M) receptor antagonists which may be used with the compounds of formula (I) or salts, and/or suitable pharmaceutical compositions.
  • the muscarinic receptor antagonist can comprise or be an ipratropium salt (e.g. ipratropium bromide), an oxitropium salt (e.g. oxitropium bromide), or a tiotropium salt (e.g. tiotropium bromide); see e.g. EP 418 716 A1 for tiotropium.
  • an ipratropium salt e.g. ipratropium bromide
  • an oxitropium salt e.g. oxitropium bromide
  • a tiotropium salt e.g. tiotropium bromide
  • the anticholinergic compound or muscarinic (M) receptor antagonist can for example be for inhaled administration, in particular in particle-size- reduced form e.g. as defined herein.
  • both the muscarinic (M) receptor antagonist and the compound of formula (I) or the pharmaceutically acceptable salt thereof are for inhaled administration.
  • the anticholinergic compound or muscarinic receptor antagonist and the compound of formula (I) or salt are optionally for simultaneous administration.
  • the muscarinic receptor antagonist combination is optionally for treatment and/or prophylaxis of COPD.
  • a combination comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof together with another anti-inflammatory agent such as an anti-inflammatory corticosteroid; or a non- steroidal anti-inflammatory drug (NSAID) such as a leukotriene antagonist (e.g. montelukast), an iNOS inhibitor, a tryptase inhibitor, a elastase inhibitor, a beta-2 integrin antagonist, a adenosine 2a agonist, a CCR3 antagonist, or a 5-lipoxogenase inhibitor; or an antiinfective agent (e.g. an antibiotic or an antiviral).
  • An iNOS inhibitor can e.g. be for oral administration.
  • iNOS inhibitors inducible nitric oxide synthase inhibitors
  • examples of iNOS inhibitors include those disclosed in WO 93/13055, WO 98/30537, WO 02/50021 , WO 95/34534 and WO 99/62875.
  • Example combinations, in particular for external topical administration include, for example, a combination comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof together with an immunosuppressant, e.g. a calcineurin inhibitor such as pimecrolimus or tacrolimus.
  • an immunosuppressant e.g. a calcineurin inhibitor such as pimecrolimus or tacrolimus.
  • the immunosuppressant can in particular be an externally-topically administrable immunosuppressant such as pimecrolimus (e.g. pimecrolimus at ca. 1% w/w concentration in a topical composition such as a cream, and/or e.g. Elidel TM) or tacrolimus (e.g.
  • the externally-topically administrable immunosuppressant can be administered or administrable in a external-topical composition separately from the compound or salt of the invention, or it can be contained with the compound of formula (I) or pharmaceutically acceptable salt in a combined externally-topically-administrable composition.
  • a topical composition such as an ointment, and/or e.g. Protopic TM.
  • the externally-topically administrable immunosuppressant can be administered or administrable in a external-topical composition separately from the compound or salt of the invention, or it can be contained with the compound of formula (I) or pharmaceutically acceptable salt in a combined externally-topically-administrable composition.
  • For external topical administration e.g.
  • the anti-infective agent can include (e.g. be) an externally-topically-administrable antibacterial, such as mupiricin or a salt thereof (e.g. mupiricin calcium salt) (e.g. Bactroban TM) or such as an externally-topically- administrable pleuromutilin antibacterial (e.g. rumblemulin or a salt thereof, which can be present in about 1% w/w by weight of an externally-topically-administrable pharmaceutical composition, such as an ointment).
  • the anti-infective agent can include an externally- topically-administrable antifungal such as clortrimazole, clotrimazole or ketoconazole.
  • a combination with an anti-itch compound may optionally be used.
  • the anti-inflammatory corticosteroid is fluticasone propionate (e.g. see US patent 4,335,121), beclomethasone 17-propionate ester, beclomethasone 17,21 -dipropionate ester, dexamethasone or an ester thereof, mometasone or an ester thereof (e.g.
  • mometasone furoate ), ciclesonide, budesonide, flunisolide, or a compound as described in WO 02/12266 A1 (e.g. as claimed in any of claims 1 to 22 therein), or a pharmaceutically acceptable salt of any of the above.
  • the anti-inflammatory corticosteroid is a compound as described in WO 02/12266 A1, then it can for example be Example 1 therein ⁇ which is 6 ⁇ ,9 ⁇ -difluoro-17 ⁇ -[(2-furanylcarbonyl)oxy]-11 ⁇ -hydroxy-16 ⁇ -methyl-3- oxo-androsta-1 ,4-diene-17 ⁇ -carbothioic acid S-fluoromethyl ester ⁇ or Example 41 therein ⁇ which is 6 ⁇ ,9 ⁇ -difluoro-11 ⁇ -hydroxy-16 ⁇ -methyl-17 ⁇ -[(4-methyl-1 ,3-thiazole-5- carbonyl)oxy]-3-oxo-androsta-1 ,4-diene-17 ⁇ -carbothioic acid S-fluoromethyl ester ⁇ , or a pharmaceutically acceptable salt thereof.
  • the anti-inflammatory corticosteroid can for example be for external topical, intranasal or inhaled administration.
  • Fluticasone propionate can be used for inhaled administration to a human, for example either (a) at a dose of 250 micrograms once per day or (b) at a dose of 50 to 250 micrograms twice per day.
  • a combination comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof together with ⁇ 2 -adrenoreceptor agonist and an anti-inflammatory corticosteroid, for example as described in WO 03/030939 A1.
  • This combination can for example be for treatment and/or prophylaxis of asthma, COPD or allergic rhinitis.
  • the ⁇ 2 -adrenoreceptor agonist and/or the anti-inflammatory corticosteroid can be as described above and/or as described in WO 03/030939 A1.
  • the ⁇ 2 -adrenoreceptor agonist can e.g. be salmeterol or a pharmaceutically acceptable salt thereof (e.g. salmeterol xinafoate) and the antiinflammatory corticosteroid can e.g. be fluticasone propionate.
  • compositions comprising a combination as defined above together with one or more pharmaceutically acceptable carriers and/or excipients represent a further aspect of the invention.
  • the individual compounds of such combinations may be administered either sequentially or simultaneously in separate or combined pharmaceutical composition.
  • the combination as defined herein can be for simultaneous inhaled administration and is disposed in a combination inhalation device.
  • a combination inhalation device is another aspect of the invention.
  • Such a combination inhalation device can comprise a combined pharmaceutical composition for simultaneous inhaled administration (e.g. dry powder composition), the composition comprising all the individual compounds of the combination, and the composition being incorporated into a plurality of sealed dose containers mounted longitudinally in a strip or ribbon inside the inhalation device, the containers being rupturable or peel-openable on demand; for example such inhalation device can be substantially as described in GB 2,242,134 A (DISKUS TM) and/or as described above.
  • DISKUS TM substantially as described in GB 2,242,134 A
  • the combination inhalation device can be such that the individual compounds of the combination are administrable simultaneously but are stored separately (or wholly or partly stored separately for triple combinations), e.g. in separate pharmaceutical compositions, for example as described in PCT/EP03/00598 filed on 22 January 2003, published as WO 03/061743 (e.g. as described in the claims thereof e.g. claim 1).
  • the invention also provides a method of preparing a combination as defined herein, the method comprising either
  • the invention also provides a combination as defined herein, prepared by a method as defined herein.
  • the biological activity of the compounds or salts of the invention can be measured in the assay methods shown below, or in generally similar or generally analogous assay methods.
  • Human recombinant PDE4B in particular the 2B splice variant thereof (HSPDE4B2B), is disclosed in WO 94/20079 and also M. M. McLaughlin et al., "A low Km, rolipram- sensitive, cAMP-specific phosphodiesterase from human brain: cloning and expression of cDNA, biochemical characterisation of recombinant protein, and tissue distribution of mRNA", J. Biol. Chem., 1993, 268, 6470-6476.
  • human recombinant PDE4B is described as being expressed in the PDE- deficient yeast Saccharomyces cerevisiae strain GL62, e.g. after induction by addition of 150 uM CUSO4, and 100,000 x g supernatant fractions of yeast cell lysates are described for use in the harvesting of PDE4B enzyme.
  • HSPDE4D3A Human recombinant PDE4D (HSPDE4D3A) is disclosed in P. A. Baecker et al., "Isolation of a cDNA encoding a human rolipram-sensitive cyclic AMP phoshodiesterase (PDE IV D )", Gene, 1994, 138, 253-256.
  • Human recombinant PDE5 is disclosed in K. Loughney et al., "Isolation and characterisation of cDNAs encoding PDE5A, a human cGMP-binding, cGMP-specific 3',5'-cyclic nucleotide phosphodiesterase", Gene, 1998, 216, 139-147.
  • PDE3 can be purified from bovine aorta as described by H. Coste and P. Grondin, "Characterisation of a novel potent and specific inhibitor of type V phosphodiesterase", Biochem. Pharmacol., 1995, 50, 1577-1585.
  • PDE6 can be purified from bovine retina as described by: P. Catty and P. Deterre, "Activation and solubilization of the retinal cGMP-specific phosphodiesterase by limited proteolysis", Eur. J. Biochem., 1991, 199, 263-269; A. Tar et al. "Purification of bovine retinal cGMP phosphodiesterase", Methods in Enzymology, 1994, 238, 3-12; and/or D. Srivastava et al. "Effects of magnesium on cyclic GMP hydrolysis by the bovine retinal rod cyclic GMP phosphodiesterase", Biochem. J., 1995, 308, 653-658. Inhibition of PDE 3, PDE 4B, PDE 4D, PDE 5 or PDE 6 activity: radioactive Scintiliation Proximity Assay (SPA)
  • SPA radioactive Scintiliation Proximity Assay
  • the ability of compounds to inhibit catalytic activity at PDE4B or 4D can optionally be determined by Scintillation Proximity Assay (SPA) in a 96-well format.
  • SPA Scintillation Proximity Assay
  • Test compounds (as a solution in DMSO, suitably about 2 microlitre (ul) volume of DMSO solution) are preincubated at ambient temperature (room temperature, e.g. 19- 23 0 C) in Wallac lsoplates (code 1450-514) with PDE enzyme in 5OmM Tris-HCI buffer pH 7.5 , 8.3mM MgCI 2 , 1.7mM EGTA, 0.05% (w/v) bovine serum albumin for 10-30 minutes (usually 30 minutes). The enzyme concentration is adjusted so that no more than 20% hydrolysis of the substrate defined below occurs in control wells without compound, during the incubation.
  • Plates containing assay mixture are mixed on an orbital shaker for 5 minutes and incubated at ambient temperature for 1 hour.
  • Phosphodiesterase SPA beads (Amersham Pharmacia Biotech, code RPNQ 0150) are added (about 1mg per well) to terminate the assay. Plates are sealed and shaken and allowed to stand at ambient temperature for 35 minutes to 1 hour (suitably 35 minutes) to allow the beads to settle.
  • Bound radioactive product is measured using a WALLAC TRILUX 1450 Microbeta scintillation counter.
  • 10 concentrations e.g. 1.5nM - 3OuM
  • Curves are analysed using ActivityBase and XLfit (ID Business Solutions Limited, 2 Ocean Court, Surrey Research Park, Guildford, Surrey GU2 7QB, United Kingdom). Results are expressed as plC 5 o values.
  • PDE4B or PDE4D inhibition can be measured in the following Fluorescence Polarisation (FP) assay:
  • the ability of compounds to inhibit catalytic activity at PDE4B (human recombinant) or PDE4D (human recombinant) can optionally be determined by IMAP Fluorescence Polarisation (FP) assay (IMAP Explorer kit, available from Molecular Devices Corporation, Sunnydale, CA, USA; Molecular Devices code: R8062) in 384-well format.
  • FP IMAP Fluorescence Polarisation
  • the IMAP FP assay is able to measure PDE activity in an homogenous, nonradioactive assay format.
  • the FP assay uses the ability of immobilised trivalent metal cations, coated onto nanoparticles (tiny beads), to bind the phosphate group of FI-AMP that is produced on the hydrolysis of fluorescein-labelled (Fl) cyclic adenosine mono- phosphate (FI-cAMP) to the non-cyclic FI-AMP form.
  • FI-cAMP substantially does not bind.
  • Binding of FI-AMP product to the beads (coated with the immobilised trivalent cations) slows the rotation of the bound FI-AMP and leads to an increase in the fluorescence polarisation ratio of parallel to perpendicular light. Inhibition of the PDE reduces/inhibits this signal increase.
  • Test compounds small volume, e.g. ca. 0.5 to 1 microlitres (ul), suitably ca. 0.5 ul, of solution in DMSO
  • ambient temperature room temperature, e.g. 19-23 0 C
  • PDE enzyme in 1OmM Tris-HCI buffer pH 7.2, 1OmM MgCI 2 , 0.1% (w/v) bovine serum albumin, and 0.05% NaN 3 for 10-30 minutes.
  • the enzyme level is set by experimentation so that reaction is linear throughout the incubation.
  • Fluorescein adenosine 3',5'-cyclic phosphate (from Molecular Devices Corporation, Molecular Devices code: R7091) is added to give about 4OnM final concentration (final assay volume usually ca. 20-40 ul, suitably ca. 20 ul). Plates are mixed on an orbital shaker for 10 seconds and incubated at ambient temperature for 40 minutes. IMAP binding reagent (as described above, from Molecular Devices Corporation, Molecular Devices code: R7207) is added (6OuI of a 1 in 400 dilution in binding buffer of the kit stock solution) to terminate the assay. Plates are allowed to stand at ambient temperature for 1 hour.
  • FP Fluorescence Polarisation
  • 10 concentrations e.g. 1.5nM - 3OuM
  • Curves are analysed using ActivityBase and XLfit (ID Business Solutions Limited, 2 Ocean Court, Surrey Research Park, Guildford, Surrey GU2 7QB, United Kingdom). Results are expressed as plC 50 values.
  • reagents can be dispensed using MultidropTM (available from
  • the PDE4B (or PDE4D) inhibition values measured using the SPA and FP assays can differ slightly.
  • the compound of the invention (e.g. as prepared in Example 1) has also been tested for PDE3, PDE5 and PDE6 inhibition, e.g. using (independently for each PDE type) either a Fluorescence Polarisation (FP) general type of assay or using an SPA general type of assay.
  • the compound of the invention (e.g. as prepared in Example 1) exhibits a larger PDE4B plC 50 value than its PDE3, PDE5 and PDE6 plC 50 values, i.e. it inhibits PDE4B more strongly than it inhibits PDE3, PDE5 and PDE6 (in the particular assays used).
  • Emesis Some known PDE4 inhibitors can cause emesis and/or nausea to greater or lesser extents, especially after systemic exposure e.g. after oral administration (e.g. see Z. Huang et al., Current Opinion in Chemical Biology, 2001 , 5: 432-438, see especially pages 433-434 and refs cited therein). Therefore, it would be preferable, but not essential, if a PDE4 inhibitory compound or salt of the invention were to cause only limited or manageable emetic side-effects, e.g. after oral or parenteral or external-topical administration.
  • Emetic side-effects can for example be measured by the emetogenic potential of the compound or salt when administered to ferrets; for example one can measure the time to onset, extent, frequency and/or duration of vomiting, retching and/or writhing in ferrets after oral or parenteral administration of the compound or salt. See for example In vivo Assay 3 hereinafter for one optional measurement method for anti- inflammatory effect, emetic side-effects and therapeutic index (Tl) in the ferret. See also for example A.
  • emetic side-effects and therapeutic index (Tl) after oral administration in rats can be conveniently measured by monitoring the pica feeding behaviour of rats after administration of the compound or salt of the invention (see In Vivo Assay 2 below).
  • TNF- ⁇ TNF-alpha
  • a 96-well plate (96 MicroWellTM Plates NunclonTM ⁇ - High Flange Design, Fisher Scientific UK, Bishop Meadow Road, Loughborough LE 11 5 RG, Sheffieldshire, UK) is prepared by initially adding to column 1 ca. 1OmM of test compound dissolved in DMSO. For a more potent compound, a more diluted solution in DMSO may be used. The compound is further diluted with DMSO into columns 2 to 9 by 8 successive 3-fold dilutions using the Biomek® FX Laboratory Automation Workstation (Beckman Coulter, Inc., 4300 N. Harbor Boulevard, P.O. Box 3100, Fullerton, CA 92834-3100 USA).
  • PBMC cells peripheral blood mononuclear cells
  • PBMC cells peripheral blood mononuclear cells
  • heparinised human blood using 1% v/v Heparin Sodium 1000IU/ml Endotoxin Free, Leo Laboratories Ltd., Cashel Road, Dublin 12 . Ireland, Cat No: PL0043/0149
  • AccuspinTM System-Histopaque ® -1077 essentially (Sigma-Aldrich Company Ltd., The Old Brickyard New Rd, Gillingham Dorset SP8 4XT).
  • About 20 ml of blood is overlaid onto 15ml Histopaque ® in AccuspinTM tubes. The tube is then centrifuged at about 80Og for ca. 20 minutes.
  • the cells are collected from the interface, washed by centrifugation (ca. 130Og, ca. 10 minutes) and resuspended in RPM11640 medium (Low endotoxin RPMH 640 medium, Cat No: 31870-025, Invitrogen Corporation Invitrogen Ltd, 3 Fountain Drive, Inchinnan Business Park, Paisley PA4 9RF, UK) containing 10% foetal calf serum, 1% L-glutamine (Invitrogen Corporation, Cat No: 25030024) and 1% penicillin/streptomycin (Invitrogen Corporation, Cat No: 15140-122). Viable cells are counted by trypan blue staining and diluted to 1x1 O ⁇ viable cells/ml. About 50 ⁇ l (about
  • TNF- ⁇ concentrations of TNF- ⁇ are determined by electrochemiluminescence assay using the Meso Scale Discovery (MSD) technology (Meso Scale Discovery, 9238 Gaither Road, Gaithersburg, Maryland 20877, USA). See the "TNF- ⁇ (TNF-alpha) MSD Assay” described below for typical details.
  • MSD Meso Scale Discovery
  • Results can be expressed as plC50 values for inhibition of TNF- ⁇ (TNF-alpha) production in PBMCs, and it should be appreciated that these results can be subject to variability or error.
  • TNFa TNF-alpha
  • Human PBMC peripheral blood mononuclear cell assay
  • Test compounds are prepared as a ca. 1OmM stock solution in DMSO and a dilution series prepared in DMSO with 8 successive 3-fold dilutions, either directly from the 1OmM stock solution or from a more dilute solution in DMSO.
  • the compound is added to assay plates using a Biomek Fx liquid handling robot.
  • PBMC cells peripheral blood mononuclear cells
  • peripheral blood mononuclear cells peripheral blood mononuclear cells
  • the cells are collected from the interface, washed by centrifugation (ca. 130Og, ca. 10 minutes) and resuspended in assay buffer (RPMI 1640 containing 10% foetal calf serum, 1% L-glutamine and 1% penicillin / streptomycin) at 1x10 6 cells/ml.
  • Ca. 50 ⁇ l (ca. 5OuI) of cells are added to microtitre wells containing ca. 0.5 or ca. 1.O ⁇ l (ul) of an appropriately diluted compound solution.
  • Ca. 75 ⁇ l (ul) of LPS (lipopolysaccharide) (ca. 1 ng/ml final) is added and the samples are incubated at 37 0 C, 5% CO2, for 20 hours.
  • TNF- ⁇ concentrations of TNF- ⁇ are determined by electrochemiluminescence assay using the IGEN technology or by ELISA (see below).
  • Results can be expressed as plC50 values for inhibition of TNF- ⁇ (TNF-alpha) production in PBMCs, and it should be appreciated that these results can be subject to variability or error.
  • the assay may measure the effect of PDE4 inhibitors after loss by protein binding, it might possibly also be relevant to extemally-topically-administrable PDE4 inhibitors as protein-binding-loss of compound is possible during transport through the skin.
  • Test compounds are prepared as a ca. 1OmM stock solution in DMSO and a dilution series prepared in DMSO with 8 successive 3-fold dilutions, either directly from the 1OmM stock solution or from a more dilute solution in DMSO.
  • the compound is added to assay plates using a Biomek Fx liquid handling robot.
  • Plasma TNF ⁇ content is determined by electrochemiluminescence assay using the MSD technology (see below), the IGEN technology (see below) or by enzyme linked immunosorbant assay (ELISA) (see below).
  • Results can be expressed as plC50 values for inhibition of TNF- ⁇ (TNF-alpha) production in Human Whole Blood, and it should be appreciated that these results can be subject to variability or error.
  • Human Whole Blood assay results :
  • the mean and/or measured plC50 values for inhibition of TNF- ⁇ (TNF-alpha) production in Human Whole Blood are generally as follows (subject to variability or error):
  • TNF- ⁇ (TNF-alpha) MSD Assay Using the Biomek FX, 25 ⁇ l (25ul) of MSD Human Serum Cytokine Assay Diluent (Meso Scale Discovery, 9238 Gaither Road, Gaithersburg, Maryland 20877) is added to a 96-well High-Bind MSD plate pre-coated with anti-hTNF alpha capture antibody (MA6000) and then incubated for 24 hours at 4°C to prevent non-specific binding. About 20 ⁇ l (ul) of supernatant from the PBMC plate or about 40 ⁇ l (ul) of supernatant from the whole blood (WB) plate are then transferred from columns 1-11 to columns 1-11 of the MSD plate using the Biomek FX.
  • TNF- ⁇ standard About 20 ⁇ l (ul) of TNF- ⁇ standard (Cat No. 210-TA; R&D Systems Inc., 614 McKinley Place NE, Minneapolis, MN 55413, USA) are added to column 12 of the MSD plate to generate a standard calibration curve (about 0 to 30000 pg/ml final).
  • PBMC assay For the PBMC assay, about 20 ⁇ l (ul) of diluted sulfo-TAG antibody (ca. 1 ⁇ g/ml final) is added to each well, and the plates / wells are shaken at room temperature for 2 hours. Finally, about 90 ⁇ l (ul) of MSD Read Buffer P (diluted to 2.5 times with distilled water) is added and the plates are read on a MSD Sector 6000.
  • TNF-a (TNF-alpha) IGEN Assay Ca. 50 ⁇ l supernatant from either whole blood or PBMC assay plates is transferred to a 96 well polypropylene plate. Each plate also contains a TNF- ⁇ standard curve (ca. 0 to 30000 pg/ml: R+D Systems, 210-TA). Ca. 50 ⁇ l (ul) of streptavidin/biotinylated anti- TNF- ⁇ antibody mix, ca. 25 ⁇ l ruthenium tagged anti-TNF- ⁇ monoclonal and ca. 100 ⁇ l PBS containing 0.1% bovine serum albumin are added to each well and the plates are sealed and shaken for ca. 2 hours before being read on an IGEN instrument.
  • TNF- ⁇ (TNF-alpha) ELISA Assay enzyme linked immunosorhant assay
  • Human TNF- ⁇ can be assayed using a commercial ELISA assay kit (AMS Biotechnology, 211-90-164-40) according to the manufacturers' instructions but with TNF- ⁇ calibration curves prepared using Pharmingen TNF- ⁇ (cat No. 555212).
  • in vitro enzymatic PDE4B inhibition assay(s) described above or generally similar or generally analogous assays should be regarded as being the primary test(s) of biological activity.
  • additional in vivo biological tests which are optional only, and which are not an essential measure of activity, efficacy or side-effects, and which have not necessarily been carried out, are described below.
  • the pig DTH (delayed type hypersensitivity) model of contact hypersensitivity utilizes the Th2-mediated inflammatory response in pig skin to mimic the pathology of atopic dermatitis in humans.
  • the model measures the potential anti-inflammatory effect of compounds, topically-applied to the skin, on the acute DTH (delayed type hypersensitivity) response in castrated male Yorkshire pigs.
  • pigs domestic Yorkshire pigs, 15-18 kg at time of sensitization, castrated males
  • DNFB dinitrofluorobenzene
  • DMSO:acetone:olive oil ca. 1:5:3
  • DNFB 400 microlitre solution total
  • the pigs are then challenged with ca. 0.6% (w/v) DNFB applied to randomized sites on the shaved back of the pigs (ca. 90 micrograms/site; sites are identified and numbered by grid made with marking pen).
  • the treatments are performed at the challenge sites at about 2 hours prior to and about 6 hours after challenge (for DMSO / acetone solutions/suspensions containing the PDE4 inhibitor, to maximize exposure to drug), or at about 30 minutes after and about 6 hours after challenge (for topical ointments or creams containing the PDE4 inhibitor, representing a more clinically relevant treatment protocol).
  • treatment sites can optionally also be visually evaluated for lesion area.
  • a single specific cream composition being either Composition Example 5 or
  • Compounds or pharmaceutical compositions showing apparent activity in the above pig DTH model may have potential utility via skin topical administration in the treatment and/or prophylaxis of atopic dermatitis, e.g. in humans.
  • the preliminary nature of In Vivo Assay A, and the potential for error or for variation in results when the assay is repeated is noted.
  • LPS-induced pulmonary neutrophilia in rats effect of orally administered PDE4 inhibitors Pulmonary neutrophil influx is thought to be a significant component to the family of pulmonary diseases like chronic obstructive pulmonary disease (COPD) which can involve chronic bronchitis and/or emphysema (G. F. Filley, Chest. 2000; 117(5); 251s- 260s).
  • COPD chronic obstructive pulmonary disease
  • the purpose of this neutrophilia model is to study the potentially antiinflammatory effects in vivo of orally administered PDE4 inhibitors on neutrophilia induced by inhalation of aerosolized lipopolysaccharide (LPS), modelling the neutrophil inflammatory component(s) of COPD. See the literature section below for scientific background.
  • test compound for example suspended in about 0.5% methylcellulose (obtainable from Sigma-Aldrich, St Louis, MO, USA) in water or (b) vehicle only, delivered orally in a dose volume of ca. 10 ml/kg.
  • dose response curves can for example be generated using the following approx. doses of PDE4 inhibitors: 2.0, 0.4, 0.08, 0.016 and 0.0032 mg/kg (or alternatively approx. 10, 2.0, 0.4, 0.08 and 0.016 mg/kg).
  • dose response curves can for example be generated using the following approx. doses of PDE4 inhibitors: 2.0, 0.4, 0.08, 0.016 and 0.0032 mg/kg (or alternatively approx. 10, 2.0, 0.4, 0.08 and 0.016 mg/kg).
  • aerosolized LPS Serotype E.
  • CoIi 026:B6 prepared by trichloroacetic acid extraction, obtainable from Sigma-Aldrich, St Louis, MO, USA), generated from a nebulizer containing a ca. 100 ⁇ g/ml LPS solution (ca. 100 ug/ml). Rats are exposed to the LPS aerosol at a rate of ca. 4 L/min for ca. 20 minutes. LPS exposure is carried out in a closed chamber with internal dimensions of roughly 45 cm length x 24 cm width x 20 cm height. The nebulizer and exposure chamber are contained in a certified fume hood. At about 4 hours-post LPS exposure the rats are euthanized by overdose with pentobarbital at ca. 90 mg/kg, administered intraperitoneal ⁇ .
  • Bronchoalveolar lavage (BAL) is performed through a 14 gauge blunt needle into the exposed trachea. Five, 5 ml washes are performed to collect a total of 25 ml of BAL fluid. Total cell counts and leukocyte differentials are performed on BAL fluid in order to calculate neutrophil influx into the lung. Percent neutrophil inhibition at each dose (cf. vehicle) is calculated and a variable slope, sigmoidal dose-response curve is generated, usually using Prism Graph- Pad. The dose-response curve is used to calculate an ED50 value (in mg per kg of body weight) for inhibition by the PDE4 inhibitor of the LPS-induced neutrophilia.
  • ED50 value in mg per kg of body weight
  • PDE4 inhibitors are thought to inhibit inflammation in various in vitro and in vivo models by increasing intracellular levels of cAMP of many immune cells (e.g. lymphocytes, monocytes).
  • cAMP a side effect of some PDE4 inhibitors in some species is emesis.
  • many rat models of inflammation are well characterized, they can be used in procedures (see e.g. In Vivo Assay 1 above) to show beneficial anti-inflammatory effects of PDE 4 inhibitors.
  • rats have no emetic response (they have no vomit reflex), so that the relationship between beneficial antiinflammatory effects of PDE 4 inhibitors and emesis is difficult to study directly in rats.
  • Pica feeding is a behavioural response to illness in rats wherein rats eat non-nutritive substances such as earth or in particular clay (e.g. kaolin) which may help to absorb toxins.
  • Pica feeding can be induced by motion and chemicals (especially chemicals which are emetic in humans), and can be inhibited pharmacologically with drugs that inhibit emesis in humans.
  • the Rat Pica Model, In Vivo Assay 2 can determine the level of pica response of rats to PDE 4 inhibition at pharmacologically relevant doses in parallel to in vivo anti-inflammatory Assays in (a separate set of) rats (e.g. In Vivo Assay 1 above).
  • Anti-inflammatory and pica assays in the same species together can provide data on the "therapeutic index" (Tl) in the rat of the compounds/salts of the invention.
  • the Rat Tl can for example be calculated as the ratio of a) the potentially-emetic Pica Response ED50 dose from Assay 2 to b) the rat anti-inflammatory ED50 dose (e.g.
  • the rats are housed individually in cages without bedding or "enrichment".
  • the rats are kept off of the cage floor by a wire screen.
  • Pre-weighed food cups containing standard rat chow and clay pellets are placed in the cage.
  • the clay pellets obtainable from Languna Clay Co, City of Industry, CA, USA, are the same size and shape as the food pellets.
  • the rats are acclimated to the clay for 72 hours, during which time the cups and food and clay debris from the cage are weighed daily on an electronic balance capable of measuring to the nearest 0.1 grams. By the end of the 72 hour acclimation period the rats generally show no interest in the clay pellets.
  • the rats are placed in clean cages and the food cups weighed. Rats that are still consuming clay regularly are removed from the study.
  • the animals are split into treatment groups and dosed orally with a dose of the compound or salt of the invention (different doses for different treatment groups) or with vehicle alone, at a dose volume of ca. 2 ml/kg.
  • the compound or salt can for example be in the form of a suspension in about 0.5% methylcellulose (obtainable Sigma-Aldrich, St. Louis, MO, USA) in water.
  • the food and clay cups and cage debris are weighed the following day and the total clay and food consumed that night by each individual animal is calculated.
  • a dose response is calculated by first converting the data into quanta! response, where animals are either positive or negative for the pica response.
  • a rat is "pica positive” if it consumes greater than or equal to 0.3 grams of clay over the mean of its control group.
  • the D50 value is usually calculated using logistic regression performed by the Statistica software statistical package.
  • a Pica Response ED50 value in mg per kg of body weight can then be calculated.
  • the Pica Response ED50 value can be compared to the neutrophilia-inhibition ED50 values for the same compound administered orally to the rat (measurable by In
  • Rat Therapeutic index (Tl) (50/50) Pica Response ED50 value rat neutrophilia-inhibition ED50 value
  • the Therapeutic Index (Tl) calculated this way can be substantially different to, and for example (without being bound) can be substantially higher than, the Tl (D20/D50) calculated in the ferret (see In vivo Assay 3 below).
  • the In Vivo Assay 2 (pica) can use only a single oral dose of the test compound (e.g. 10 mg/kg orally).
  • Literature Beavo JA, Contini, M., Heaslip, R.J. Multiple cyclic nucleotide phosphodiesterases. MoI Pharmacol. 1994; 46:399-405.
  • PDE4 inhibitors are prepared for oral (p.o.) administration by dissolving in a fixed volume
  • Acetone is evaporated by directing a flow of nitrogen gas onto the solution. Once the acetone is removed, the solution is made up to final volume with distilled water.
  • LPS is dissolved in phosphate buffered saline.
  • the diet comprises SDS diet C pelleted food given ad lib with Whiskers ⁇ M ca t food given 3 times per week.
  • the animals are supplied with pasteurised animal grade drinking water changed daily.
  • PDE4 inhibitors are administered orally (p.o.), using a dose volume of ca. 1 ml/kg. Ferrets are fasted overnight but allowed free access to water.
  • the animals are orally dosed with vehicle or PDE 4 inhibitor using a ca. 15cm dosing needle that is passed down the back of the throat into the oesophagus. After dosing, the animals are returned to holding cages fitted with perspex doors to allow observation, and given free access to water. The animals are constantly observed and any emetic episodes (retching and vomiting) or behavioural changes are recorded. The animals are allowed access to food ca. 60 - 90 minutes after p.o. dosing.
  • the trachea is then cannulated with polypropylene tubing and the lungs lavaged twice with ca. 20 ml heparinised (10 units/ml) phosphate buffered saline (PBS).
  • PBS phosphate buffered saline
  • the bronchoalveolar lavage (BAL) samples are centrifuged at ca. 1300 rpm for ca. 7 minutes. The supernatant is removed and the resulting cell pellet re-suspended in ca. 1 ml PBS.
  • a cell smear of re-suspended fluid is prepared and stained with Leishmans stain to allow differential cell counting. A total cell count is made using the remaining re-suspended sample. From this, the total number of neutrophils in the BAL sample is determined.
  • the Ferret Therapeutic index (Tl) (D20/D50) calculated using this in vivo Assay 3 can be substantially different to, and for example (without being bound) can be substantially lower than, the Rat Tl (50/50) calculated using the rat oral inflammation and pica feeding Assays 1+2.
  • Intermediates can represent syntheses of intermediate compounds intended for use in the synthesis of one or more of the “Examples”, and/or “Intermediates” can represent syntheses of intermediate compounds which can be used in the synthesis of compounds of formula (I) or salts thereof.
  • “Examples” are examples of a compound or salt of the invention, i.e. examples of a compound of formula (I) or a salt thereof.
  • TRET retention time (e.g. from LCMS) Room temperature (ambient temperature): this is usually in the range of about 18 to about 25 0 C.
  • Waters ZQ mass spectrometer operating in positive ion electrospray mode, mass range 100-1000 amu.
  • UV wavelength 215-330 nm
  • Injection Volume 5 ⁇ l (5 microlitres)
  • Solvent A 0.05% v/v solution of formic acid in a mixture of [95% acetonitrile and 5% water]
  • Solvent B aqueous solution of [0.1% v/v formic acid + IOmMolar ammonium acetate]
  • T RE ⁇ retention times quoted herein are inherently variable (e.g. the variability can be about +/- 0.2 min or more.). Variability can arise e.g. when samples are run on different Waters machines, or on the same Waters machine at different times of day or under slightly different conditions, even when the same type of column and identical flow rates, injection volumes, solvents and gradients are used.
  • a mass spectrometer attached to the end of the column can detect peaks arising from eluted compounds.
  • UV wavelength usually 200-320 nm
  • Gradient systems mixtures of Solvent A and Solvent B are used according to a choice of 5 generic gradient profiles (expressed as % Solvent B in the mixture), ranging from a start of 0 to 50% Solvent B, with all finishing at 100% Solvent B to ensure total elution.
  • Method 1 Generally, two alternative solvent systems have been used, Method 1 and Method 2:
  • Solvent A 0.1% v/v aqueous formic acid solution
  • Solvent B 0.05% v/v solution of formic acid in a mixture of [95% acetonitrile and 5% water]
  • Solvent B solution of 0.1% v/v trifluoroacetic acid in acetonitrile
  • Albemarle Corporation 451 Florida Street, Baton Rouge, LA 70801 , USA; or Albemarle Europe SPRL, Pare Scientific de LLN, Rue du Bosquet 9, B-1348 Louvain-la-Neuve, Belgium
  • 1-ethyl-1H-pyrazol- 5-amine e.g. available from Aldrich Chemical Company Inc., Albemarle Corporation, Art- Chem GmbH, Enamine and/or TimTec Corporation
  • Triethylamine (230 ml) is added dropwise to a mixture of diethyl (i-chloropropylidene)propanedioate (208g) and 1-ethyl-1H-pyrazol-5-amine (101g) in toluene (2.65L). The mixture is heated under reflux for 16 hours. The reaction mixture is cooled to room temperature, and the solid removed by filtration. The filtrate is evaporated under reduced pressure. The residue is heated under reflux in phosphorus oxychloride (POCI3, 2.65L) for 16hrs.
  • POCI3, 2.65L phosphorus oxychloride
  • Ethyl 4-chloro-1 ,6-diethyl-1/-/-pyrazolo[3,4-jb]pyridine-5-carboxylate (0.5Og) (e.g. this can be as prepared in Intermediate 1) was dissolved in 1-methyl-2-pyrrolidinone (5ml) and treated with tetrahydro-2H-pyran-4-amine hydrochloride (0.49g) [e.g. this can be as prepared in Intermediate 12, see below] and DIPEA (0.60ml) at 120 0 C overnight. The mixture was allowed to cool and was partitioned between ethyl acetate (3 x 50ml) and water (50ml). The organic layer was separated, dried and evaporated in vacuo.
  • Intermediate 4 can be prepared, in an analogous manner to Reference Intermediate 3, from ethyl 1 ,6-diethyl-4-(tetrahydro-2H-pyran-4-ylamino)-1H-pyrazolo[3,4- ⁇ b]pyridine-5- carboxylate (e.g. which can be as prepared in Intermediate 2).
  • the first reaction was as follows: To ethyl 1 ,6-diethyl-4- (tetrahydro-2H-pyran-4-ylamino)-1 H-pyrazolo[3,4- ⁇ b]pyridine-5-carboxylate (7.5g) (e.g. which can be as prepared in Intermediate 2) in dichloromethane (75ml) under nitrogen at 0 0 C was added a solution of diisobutyaluminium hydride in toluene (1.5M, 43ml), dropwise, keeping the temperature at 0 0 C. The addition took 9.5mins.
  • the reaction mixture is then cooled down to 20 ⁇ 3°C and water (3 vol) is added dropwise over 20mins followed by 10.8M NaOH (6.0 vol) (dropwise over 10 mins), ensuring the temperature remains below 40 0 C.
  • the temperature of the reaction is then adjusted to 40 ⁇ 3°C and stirred vigorously for at least 90 minutes.
  • the organic layer is separated and the aqueous layer is transferred to a Schott bottle. Water is added to the organic layer (2.5 vols) followed by NaOH (10.8M, 2 vols), the biphase is stirred vigorously at 40 0 C for at least 30 minutes.
  • the organic layer is again separated and the organic solution is concentrated in vacuo to 3vol, and the product precipitates as yellow solid.
  • Toluene (3vol) is then added and the slurry is reconcentrated in vacuo to 3vol.
  • the suspension is then cooled to 10 ⁇ 3°C and aged for at least 30min.
  • the solid is then filtered and washed with toluene (3vol).
  • the product is then dried in vacuo at 45 0 C to constant temperature.
  • aqueous layer ex-step 12 and 14 can be charged to a vessel and the contents adjusted to 10 ⁇ 3°C.
  • the stirred solution is acidified using 5M H 2 SO 4 (11 vols) which is added over approx 1 hour (caution, exothermic).
  • Reference Intermediate 5 5-(chloromethyl)-1-ethyl-W-(tetrahydro-2H-pyran-4-yl)- 1H-pyrazoIo[3,4-Jb]pyridin-4-amine
  • Intermediate 6 can be prepared, in an analogous manner to Reference Intermediate 5, from [1 ,6-diethyl-4-(tetrahydro-2H-pyran-4-ylamino)-1 H-pyrazolo[3,4-ib]pyridin-5- yljmethanol (e.g. which can be as prepared in Intermediate 4).
  • Intermediate 8 can be prepared, in an analogous manner to Reference Intermediate 7, from 5-(chloromethyl)-1,6-diethyl-N-(tetrahydro-2W-pyran-4-yl)-1W- pyrazolo[3,4-/)]pyridin-4-amine (e.g. which can be as prepared in Intermediate 6).
  • [1 ,6-diethyl-4-(tetrahydro-2H-pyran-4-ylamino)-1 H-pyrazolo[3,4- ⁇ ]pyridin-5-yl]methanol (e.g. which can be as prepared in Intermediate 4, e.g. Alternative Synthesis B thereof) is suspended in anisole and treated with solid benzensulfonic acid. This suspension is aged at room temperature for 30 minutes. Thionyl chloride is added at about 20 degrees C over 20 minutes, and stirred for 20 minutes. The reaction is then sampled for HPLC.
  • the mixture is cooled to 10 degrees C and hydrochloric acid (5M) is added.
  • the phases are separated and the lower aqueous layer is transferred back into the vessel, and the organic phase is discarded.
  • Methyl-THF methyl-tetrahydrofuran
  • the layers are separated and the lower aqueous layer is back extracted with further methyl-THF.
  • the combined organic phase is washed with brine.
  • the amount of the desired 5-(aminomethyl)-1 ,6-diethyl- ⁇ /-(tetrahydro-2H-pyran-4- yl)-1H-pyrazolo[3,4-jb]pyridin-4-amine is quantified via yieldaliser, and an appropriate amount of concentrated hydrochloric acid is added at 55-60 degrees C.
  • the suspension is held for 2 hours at about 50 degrees C, and then cooled to 10 degrees C over 3 hours and held at this temperature for at least 3 hours.
  • Anisole (0.25vol) is used to wash the line.
  • Reaction mixture is cooled to 20-25 0 C and can be held at this point.
  • This mixture (ex-step 10) is added to lithium hexamethyldisilazide in THF (1.26M in THF, 6.51vol, 5.73wt) over 60-70 minutes at 35-40°C.
  • the lower aqueous layer is retained, and the upper anisole layer discarded.
  • the batch can be held at 20-25 0 C at this point.
  • the yieldaliser system is used to determine the absolute amount of 5-(aminomethyl)-1 ,6-diethyl- ⁇ /- (tetrahydro-2H-pyran-4-yl)-1/-/-pyrazolo[3,4-jb]pyridin-4-amine present.
  • reaction mixture is heated to 55-60 0 C and treated with concentrated hydrochloric acid [1.03 equivalents relative to the 5-(aminomethyl)-1 ,6-diethyl- ⁇ /-
  • the mixture is aged at 50-55 0 C for 2 hours and then cooled to 5-10 0 C over 3 hours.
  • the off white solid cake is dried in a vacuum oven at 60 0 C to constant temperature.
  • Example 1 An example of a specific synthesis of Example 1 is as follows:
  • the mixture was treated with DIPEA (0.128ml) and stirred at room temperature under nitrogen for 15h.
  • Dichloromethane (50ml) was added to the reaction mixture and the solution was washed with dilute aqueous sodium chloride solution (2 x 50ml).
  • the organic phase was separated using a hydrophobic frit (70ml) and loaded directly onto an SPE cartridge (10g, aminopropyl) which had been pre-washed with methanol.
  • the cartridge was eluted with methanol (x2) and the fractions collected and blown down / evaporated to dryness.
  • S-Methyl- ⁇ -isoxazolecarboxylic acid is thought to be commercially available from one or more of the following suppliers: Chemical Block Stock Library, Chemical Block Building Blocks, Fluorochem, Scientific Exchange, Aurora Screening Library, Oakwood Products Catalog, Ambinter Stock Screening Collection, TimTec Building Blocks and Reagents, TimTec Overseas Stock, Enamine Building Blocks, Enamine Screening Library, lnterchim Intermediates, AsInEx Express Gold Collection, and/or MicroChemistry Building Blocks. See near the start of the "Intermediates and Examples" section hereinabove, for the addresses of some of these suppliers.
  • the carboxylic acid R'COOH ( ) (0.12mmol) is treated with a solution of HATU (0.12mmol) in DMF (0.25ml), and DIPEA (0.052ml, ca. 0.3mmol) is added.
  • the solution is shaken for 10mins, and is treated with a solution of 5-(aminomethyl)-1 ,6-diethyl- ⁇ /- (tetrahydro-2H-pyran-4-yl)-1H-pyrazolo[3,4-6]pyridin-4-amine (O.immol, ca. 30.3mg) (e.g. which can be as prepared in Intermediate 10) in DMF (0.2ml).
  • the resulting solution is shaken for IOmins and left to stand for 18h (e.g. at room temperature), and then the DMF is removed in a Genevac vacuum centrifuge.
  • the residue is dissolved in chloroform (0.3ml), is applied to an SPE cartridge (1g, aminopropyl) which has been pre- washed with chloroform (6ml), and is eluted sequentially with chloroform (3ml) and 10% methanol in ethyl acetate (3ml).
  • Fractions containing the desired product are concentrated in vacuo in a Genevac vacuum centrifuge, and where necessary the residue is purified by mass directed autoprep HPLC (e.g. acetonitrile/water).
  • the compound is dissolved in chloroform, and is further purified by loading onto a SPE cartridge (0.5g, aminopropyl) which has been prewashed with chloroform, eluting with 10% methanol in ethyl acetate.
  • the reduced liquors (the reduced slurry liquids) were purified by column chromatography (silica, 400 ml) using 1 : 0.96 : 0.4 DCM : EtOAc : MeOH (2L) as eluent, giving, after reduction in vacuo, a further batch of product.
  • compositions suitable for external topical administration (e.g. topical administration to skin).
  • composition suitable for external topical administration (e.g. topical administration to skin), and comprises 2% w/w of the compound of the invention ⁇ /- ⁇ [1 ,6-diethyl-4-(tetrahydro-2H-pyran-4-ylamino)- 1W-pyrazolo[3,4-d]pyridin-5-yl]methyl ⁇ -3-methyl-5-isoxazolecarboxamide (i.e. the "free base" form):
  • the white petrolatum (a solid at room temperature, and e.g. which can optionally be high melting point white petrolatum such as Penreco Ultima White TM grade white petrolatum) and the decamethyl-cyclopentasiloxane (a liquid at room temperature) are heated together, in a beaker in a hot water bath, to a temperature of approximately 60-65 0 C, to melt the white petrolatum.
  • white petrolatum a solid at room temperature, and e.g. which can optionally be high melting point white petrolatum such as Penreco Ultima White TM grade white petrolatum
  • decamethyl-cyclopentasiloxane (a liquid at room temperature)
  • the drug substance (the compound of the invention, which, for example, can be as prepared in Example 1 , Synthesis C) is added slowly into the melted oil phase and is stirred with a spatula and dispersed completely.
  • the mixture is homogenised under high shear conditions (using setting #5, a high setting, on an Ultra-turrax T25 homogenizer), using the small homogenizer shaft, for 10 minutes.
  • the formulation is allowed to cool to room temperature (e.g. about 17 to about 22 0 C) while stirring.
  • the ointment formulation is filled into a 3OmL transparent container.
  • composition is an ointment suitable for external topical administration (e.g. topical administration to skin), but it does not include any compound of the invention. Thus it can be used as a comparator placebo ointment, when testing (e.g. in "In Vivo Assay A” herein) a corresponding ointment containing the compound of the invention such as Composition Example 1 (Ointment D).
  • Placebo Ointment AP Approximately the following procedure is used to prepare Placebo Ointment AP:
  • the white petrolatum (which can optionally be high melting point white petrolatum such as Penreco Ultima White TM grade white petrolatum) and the decamethyl- cyclopentasiloxane are heated together, in a small beaker in a hot water bath, to a temperature of approximately 60-65 0 C.
  • the mixture is homogenised under high shear conditions (using setting #5, a high setting, on an Ultra-turrax T25 homogenizer), for approximately 10 minutes.
  • the formulation is allowed to cool and is stirred until room temperature (e.g. about 17 to about 22 0 C) is reached.
  • room temperature e.g. about 17 to about 22 0 C
  • the ointment formulation is packed into a 125mL transparent plastic container.
  • composition suitable for external topical administration (e.g. topical administration to skin), and comprises 2% w/w of the compound of the invention /V- ⁇ [1 ,6-diethyl-4-(tetrahydro-2W-pyran-4-ylamino)- 1H-pyrazolo[3,4- ⁇ b]pyridin-5-yl]methyl ⁇ -3-methyl-5-isoxazolecarboxamide (i.e. the "free base" form):
  • composition is an ointment suitable for external topical administration (e.g. topical administration to skin), but it does not include any compound of the invention.
  • it can be used as a comparator placebo ointment, when testing (e.g. in "In Vivo Assay A” herein) a corresponding ointment containing the compound of the invention such as Composition Example 3 (Ointment D2).
  • Placebo Ointment AP is used to prepare Placebo Ointment AP2.
  • composition is believed to be a water-in-oil cream emulsion, for external topical administration (e.g. topical administration to skin), and comprises propylene glycol and 2% w/w of the compound of the invention ⁇ /- ⁇ [1 ,6- diethyl-4-(tetrahydro-2H-pyran-4-ylamino)-1H-pyrazolo[3,4- ⁇ ]pyridin-5-yl]methyl ⁇ -3- methyl-5-isoxazolecarboxamide (i.e. the "free base" form):
  • the white petrolatum (which can optionally be high melting point white petrolatum such as Penreco Ultima White TM grade white petrolatum), the mineral oil and the steareth-2 (Volpo s-2 TM) are heated together, via a hot water bath, to a temperature of approximately 60-65 0 C to form an oily phase.
  • white petrolatum which can optionally be high melting point white petrolatum such as Penreco Ultima White TM grade white petrolatum
  • the mineral oil and the steareth-2 (Volpo s-2 TM) are heated together, via a hot water bath, to a temperature of approximately 60-65 0 C to form an oily phase.
  • the drug substance (the compound of the invention, which, for example, can be as prepared in Example 1, Synthesis C) is dispersed in water and sonicated for approximately 10 minutes. Propylene glycol is added to this aqueous phase and the mixture is sonicated for another 10 minutes. The drug substance is at least partly in suspension.
  • the aqueous phase is heated to approximately the same temperature as the oily phase (e.g. is heated to approximately 60-65 0 C), and then the aqueous phase is added slowly to the oily phase while homogenizing the mixture under high shear conditions (using setting #5, a high setting, e.g. on an Ultra-turrax T25 homogenizer), for approximately 10 minutes.
  • the formulation After homogenization, the formulation is allowed to cool to room temperature (e.g. about 17 to about 22 0 C) with constant mixing with a spatula.
  • room temperature e.g. about 17 to about 22 0 C
  • the cream formulation is packed into a 2OmL scintillation vial.
  • composition is believed to be a water-in-oil cream emulsion, for external topical administration, but it does not include any compound of the invention. Thus it can be used as a comparator placebo cream, when testing (e.g. in "In Vivo Assay A” herein) a corresponding cream containing the compound of the invention such as Composition Example 5 (Water-in-oil cream Cr-D).
  • the white petrolatum (which can optionally be high melting point white petrolatum such as Penreco Ultima White TM grade white petrolatum), the mineral oil and the steareth-2 (Volpo s-2 TM) are heated together, via a hot water bath, to a temperature of approximately 60-65 0 C to form an oily phase.
  • the aqueous phase (propylene glycol and water) is heated to a temperature of approximately 60-65 °C.
  • the aqueous phase is added slowly to the oily phase while homogenizing the mixture at low speed, and then the speed is increased to high shear conditions (using setting #5, a high setting, e.g. on an Ultra-turrax T25 homogenizer).
  • the mixture is homogenized, while being kept hot via a hot water bath, for approximately 10 minutes.
  • the formulation is then allowed to cool and stirred until room temperature (e.g. about 17 to about 22 0 C) is reached.
  • room temperature e.g. about 17 to about 22 0 C
  • the cream formulation is packed into a 125ml_ transparent plastic container.
  • composition is believed to be a water-in-oil cream emulsion, for external topical administration (e.g. topical administration to skin), and comprises propylene glycol and 2% w/w of the compound of the invention /V- ⁇ [1 ,6- diethyl-4-(tetrahydro-2/-/-pyran-4-ylamino)-1H-pyrazolo[3,4-fe]pyridin-5-yl]methyl ⁇ -3- methyl-5-isoxazolecarboxamide (i.e. the "free base" form):
  • composition is believed to be a water-in-oil cream emulsion, for external topical administration, but it does not include any compound of the invention. Thus it can be used as a comparator placebo cream, when testing (e.g. in "In Vivo Assay A” herein) a corresponding cream containing the compound of the invention such as Composition Example 7 (Water-in-oil cream Cr-D2).

Abstract

The invention provides N-{[1,6-diethyl-4-(tetrahydro-2H-pyran-4-ylamino)-1H-pyrazolo[3,4-b]pyridin-5-yl]methyl}-3-methyl-5-isoxazolecarboxamide Formula (I): or a salt thereof, such as the compound or a pharmaceutically acceptable salt thereof. The invention also provides the use of the compound or salt as an inhibitor of phosphodiesterase type IV (PDE4) and/or for the treatment and/or prophylaxis of an inflammatory and/or allergic disease, especially an inflammatory and/or allergic skin disease such as atopic dermatitis, in a mammal such as a human. External topical administration, e.g. topical administration to the skin, of the compound or salt is preferred, and external-topical pharmaceutical compositions are provided

Description

Pyrazolo[3,4-b]pyridine compound, and its use as a PDE4 inhibitor
The present invention relates to a pyrazolo[3,4-b]pyridine compound or a salt thereof, processes for their preparation, intermediates usable in these processes, and pharmaceutical compositions containing the compound or a salt thereof. The invention also relates to the use of the pyrazolo[3,4-b]pyridine compound or a salt thereof in therapy, for example as inhibitors of phosphodiesterase type IV (PDE4) and/or for the treatment and/or prophylaxis of inflammatory and/or allergic diseases such as atopic dermatitis.
BACKGROUND TO THE INVENTION
WO 2004/024728 A2 (PCT/EP2003/011814, filed on 12 September 2003, published on 25 March 2004, Glaxo Group Limited), and incorporated herein by reference in its entirity as though fully set forth, discloses pyrazolo[3,4-b]pyridine compounds or salts thereof with a 4-NHR3 group and a 5-C(O)-X group (wherein X is NR4R5 or OR^a), according to the following formula (I):
Figure imgf000002_0001
In WO 2004/024728 A2, the pyrazolo[3,4-b]pyridine compounds of formula (I) and salts thereof disclosed therein are disclosed as being inhibitors of phosphodiesterase type IV (PDE4).
WO 2004/056823 A1 (PCT/EP2003/014867, filed on 19 December 2003, published on 8 July 2004, Glaxo Group Limited), and incorporated herein by reference in its entirity as though fully set forth, discloses and claims further pyrazolo[3,4-b]pyridine compounds or salts thereof. WO 2004/056823 A1 also discloses the use of these compounds as PDE4 inhibitors.
WO 2004/024728 has been reviewed, and WO 2004/056823 mentioned, in Expert Opin. Ther. Patents, 2005 (January edition), 15(1), 111-114.
WO 2005/058892 A1 (PCT/EP2004/014490, filed on 17 December 2004, published on 30 June 2005, Glaxo Group Limited), and incorporated herein by reference in its entirity as though fully set forth, discloses further pyrazolo[3,4-b]pyridine compounds or salts thereof and their use as PDE4 inhibitors.
Further pyrazolo[3,4-b]pyridine compounds or salts thereof, and their use as PDE4 inhibitors, are disclosed in copending patent publications WO 2005/090348 A1
(PCT/GB2005/000983), WO 2005/090354 A1 (PCT/GB2005/000987), WO 2005/090352 A1 (PCT/EP2005/003038), and WO 2005/090353 A1 (PCT/GB2005/000976) (all Glaxo Group Limited, all PCT-filed on 15 March 2005 and all published on 29 September 2005).
THE INVENTION
We have now found a new pyrazolo[3,4-b]pyridine compound which inhibits phosphodiesterase type IV (PDE4), and which for example appears to be suitable for inclusion in an external-topical pharmaceutical composition and/or which appears to be usable by external topical administration (e.g. to a mammal such as a human or pig), in particular by topical administration to the skin.
The present invention therefore provides Λ/-{[1 ,6-diethyl-4-(tetrahydro-2H-pyran-4- ylamino)-1H-pyrazolo[3,4-b]pyridin-5-yl]methyl}-3-methyl-5-isoxazolecarboxamide
Figure imgf000003_0001
or a salt thereof.
In particular, the present invention provides Λ/-{[1 ,6-diethyl-4-(tetrahydro-2H-pyran-4- ylamino)-1 H-pyrazolo[3,4-jb]pyridin-5-yl]methyl}-3-methyl-5-isoxazolecarboxamide or a pharmaceutically acceptable salt thereof.
In particular, the present invention provides Λ/-{[1 ,6-diethyl-4-(tetrahydro-2/-/-pyran-4- ylamino)-1H-pyrazolo[3,4-b]pyridin-5-yl]methyl}-3-methyl-5-isoxazolecarboxamide. This is the so-called "free base" form, also called "the compound of the invention" or similar.
The above-defined compound or salt of the present invention (hereinafter "the / a compound or salt of the invention" or "the compound of the invention or the salt thereof or similar) is a compound of formula (I) or a salt thereof (in particular, the compound or a pharmaceutically acceptable salt thereof):
Figure imgf000004_0001
wherein: R1 is ethyl; R2 is ethyl;
R3 is a heterocyclic group of sub-formula (bb) which is not substituted on a ring carbon:
Figure imgf000004_0002
(bb) in which n1 is 1 ; and in which Y is O;
and wherein:
Ra is a hydrogen atom (H); Rb is a hydrogen atom (H);
R4 is a hydrogen atom (H); R5 is -C(O)-(CH2)n-Ar; wherein n is 0;
(that is: NR4R5 is -NH-C(O)-Ar)
and Ar has the sub-formula (z) which is sub-formula (z9):
Figure imgf000004_0003
In compounds which may be usable in the preparation of the compounds of formula (I), an "alkyl" group or moiety may be straight-chain or branched. Alkyl groups, for example C-i^'M or C^galkyl or C-|_4alkyl or C<|_3alkyl or C-^alkyl, which may be employed include C^.galkyl or C^alkyl or C^alkyl or C-^alkyl such as methyl, ethyl, n-propyl, n-butyl, n-pentyl, or n-hexyl or any branched isomers thereof such as isopropyl, t-butyl, sec-butyl, isobutyl, 3-methylbutan-2-yl, 2-ethylbutan-1-yl, or the like. A corresponding meaning is intended for "alkoxy", and like terms derived from alkyl. For example, "alkoxy" such as C-i^alkoxy or C-^alkoxy or Ci_2alkoxy includes methoxy, ethoxy, propyloxy, and oxy derivatives of the alkyls listed above. "Alkylsulfonyl" such as C-^alkylsulfonyl includes methylsulfonyl (methanesulfonyl), ethylsulfonyl, and others derived from the alkyls listed above. "Alkylsulfonyloxy" such as C-|_4alkylsulfonyloxy includes methanesulfonyloxy (methylsulfonyloxy), ethanesulfonyloxy, et a/.
"Fluoroalkyl" includes alkyl groups with one, two, three, four, five or more fluorine substituents, for example C-j_4fluoroalkyl or Ci_3fluoroalkyl or C-|_2fluoroalkyl such as monofluoromethyl, difluoromethyl, trifluoromethyl, pentafluoroethyl, 2,2,2-trifluoroethyl (CF3CH2-), 2,2-difluoroethyl (CHF2CH2-), 2-fluoroethyl (CH2FCH2-), etc. "Fluoroalkoxy" includes C-^fluoroalkoxy or C-).2fluoroalkoxy such as trifluoromethoxy, pentafluoroethoxy, monofluoromethoxy, difluoromethoxy, etc. "Fluσroalkylsulfonyl" such as Ci_4fluoroalkylsulfonyl includes trifluoromethanesulfonyl, pentafluoroethylsulfonyl, etc.
A halogen atom ("halo") present in compounds, for example in the compounds of formula (I), means a fluorine, chlorine, bromine or iodine atom ("fluoro", "chloro", "bromo" or "iodo"), for example fluoro, chloro or bromo.
In the compound of formula (I) or the salt thereof of the present invention, R3 is tetrahydro-2H-pyran-4-yl; that is NHR3 is of sub-formula (h) as shown below:
Figure imgf000005_0001
(h)
In the sub-formulae (h) above, the -NH- connection point of the NHR3 group to the 4- position of the pyrazolopyridine of formula (I) is underlined.
The compound of the invention or the salt thereof can suitably be for external topical administration (e.g. to a mammal such as a human or pig), in particular for topical administration to the skin.
The compound of the invention or a pharmaceutically acceptable salt thereof can suitably be contained / comprised in a pharmaceutical composition suitable and/or adapted for external topical administration (e.g. to a mammal such as a human or pig), in particular in a pharmaceutical composition suitable and/or adapted for topical administration to the skin (e.g. to a mammal such as a human or pig). SALTS, SOLVATES, ISOMERS, TAUTOMERIC FORMS, MOLECULAR WEIGHTS, ETC.
Because of their potential use in medicine, the compound of formula (I) may be used in the form of a pharmaceutically acceptable salt thereof. Pharmaceutically acceptable salts can include acid addition salts.
For example, a pharmaceutically acceptable acid addition salt of the compound of the invention can be formed (or can have been formed) by mixing (e.g. intimately mixing) Λ/-{[1 ,6-diethyl-4-(tetrahydro-2H~pyran-4-ylamino)-1 H~pyrazolo[3,4-<b]pyridin-5- yl]methyl}-3-methyl-5-isoxazolecarboxamide with a pharmaceutically acceptable acid having a pKa of 1.5 or less.
In one embodiment, a pharmaceutically acceptable acid addition salt is optionally formed by mixing (e.g. intimately mixing) a compound of formula (I) with a suitable pharmaceutically acceptable inorganic or organic acid (such as hydrobromic, hydrochloric, sulfuric, nitric, phosphoric, p-toluenesulfonic, benzenesulfonic, methanesulfonic, ethanesulfonic, or naphthalenesulfonic such as 2- naphthalenesulfonic acid), optionally in (e.g. dissolved in) a suitable solvent, and/or optionally in (e.g. dissolved in) an organic solvent such as methanol, ethanol, isopropanol, ethyl acetate, acetonitrile and/or dichloromethane, to give the salt, which is usually isolated for example by crystallisation and filtration. In one embodiment, the compound of formula (I) is for example dissolved in a suitable solvent, and/or in an organic solvent such as methanol, ethanol, isopropanol, ethyl acetate, acetonitrile and/or dichloromethane, and the pharmaceutically acceptable inorganic or organic acid (e.g. in solution or in isolated form) is then added, to prepare the pharmaceutically acceptable salt, which is usually isolated for example by crystallisation and filtration. For preparation of a hydrochloride salt, an anhydrous solution (e.g. 1-5M, e.g. 2M) of HCI in dioxane or diethyl ether, or aqueous hydrochloric acid, or HCI gas, is optionally used as the acid. For preparation of a sulfate or nitrate salt, in one embodiment an aqueous, methanolic or ethanolic solution of sulfuric acid or nitric acid respectively is optionally added to a solution of the compound of formula (I) in a water-miscible organic solvent.
A pharmaceutically acceptable acid addition salt of a compound of formula (I) can comprise or be for example a hydrobromide, hydrochloride, sulfate, nitrate, phosphate, p-toluenesulfonate, benzenesulfonate, methanesulfonate, ethanesulfonate, or naphthalenesulfonate (e.g. 2- naphthalenesulfonate) salt of the compound of formula (I). Other non-pharmaceutically acceptable salts, e.g. oxalates or trifluoroacetates, may be used, for example in the isolation of compounds of the invention, and are included within the scope of this invention.
The invention includes within its scope all possible stoichiometric and non- stoichiometric forms of the salts of the compounds of formula (I). Also included within the scope of the invention are all solvates, hydrates and polymorphs of compounds and salts of the invention.
Certain salts of the compound of the invention (or the tautomer of the amide moiety in the compound of the invention), included in the present invention, may be present as isomers, e.g. positional, geometric or optical isomers. The present invention includes within its scope all such isomers, including racemates, enantiomers and mixtures thereof.
Certain of the groups, e.g. heteroaromatic ring systems or amide moiety(ies), included in the compound of formula (I) or a salt thereof may exist in one or more tautomeric forms. The present invention includes within its scope all such tautomeric forms, including mixtures.
SYNTHETIC PROCESS ROUTES
The following non-limiting processes can generally be used to prepare the compound of the invention:
Figure imgf000008_0001
In the compound of Formula (I), Ra and Rb are both a hydrogen atom (H), R4 is H, R5 is -C(O)-(CH2)n~Ar; wherein n is 0 and wherein Ar is as defined herein, and R^ , R^ and R^ are as defined herein.
Process A
To prepare a compound of formula (I), an amine of formula (II) or a salt thereof can be reacted with a compound Ar-C(O)-X"* , wherein X^ is a leaving group substitutable by the NH2 amine moiety of the compound of formula (II):
Figure imgf000008_0002
Ar-C(O)-X^ is typically an activated derivative of carboxylic acid Ar-C(O)-OH.
In the reaction of (II) to (I), a compound Ar-C(O)-X1 can for example be the acid chloride Ar-C(O)CI (wherein X1 is a chlorine atom (Cl)). For use of Ar-C(O)CI, the reaction is typically carried out in the presence of a base such as diisopropylethylamine ('P^NEt = DlPEA) and/or in a suitable non-aqueous non-alcohol organic solvent (e.g. anhydrous solvent) such as acetonitrile (e.g. anhydrous) or DMF, for example at room temperature (e.g. about 18 to about 25 0C).
The acid chloride Ar-C(O)CI can typically be formed from the carboxylic acid (a) by reaction with thionyl chloride, either in a suitable organic solvent such as chloroform or without solvent, or (b) by reaction with oxalyl chloride (optionally also with e.g. diethyl formamide or DMF), for example in a suitable organic solvent such as dichloromethane.
Alternatively, the compound Ar-C(O)-X1 can be an activated derivative of the carboxylic acid Ar-C(O)-OH wherein the leaving group X1 is
Figure imgf000009_0001
The activated compound Ar-C(O)-X-I, where X1 is as shown above, can optionally be formed from the carboxylic acid Ar-C(O)-OH by the following reaction (a). In reaction (a), the carboxylic acid Ar-C(O)-OH is reacted with (i), (ii), (iii) or (iv):
(i) O-(7-azabenzotriazol-1 -yl)-N,Λ/,Λ/',Λ/-tetramethyluronium hexafluorophosphate (HATU) (when X2 is N), or
(ii) O-(benzotriazol-1-yl)-Λ/,Λ/,Λ/',/V'-tetramethyluronium hexafluorophosphate (HBTU) (when X2 is CH), or (iii) 2-(1 H-benzotriazol-1-yl)-1 ,1 ,3,3-tetramethyluronium tetrafluoroborate (TBTU) (when X2 is CH), or
(iv) benzotriazol-1-yl-oxy-trispyrrolidinophosphonium hexafluorophosphate (PyBOP).
In one embodiment of reaction (a), reaction (a) is e.g. carried out in the presence of a tertiary amine base such as diisopropylethylamine ('Pr2NEt = DIPEA), and/or in the presence of a non-aqueous non-alcohol organic solvent (e.g. anhydrous solvent) such as dimethyl formamide (DMF, e.g. anhydrous DMF) or acetonitrile e.g. anhydrous acetonitrile. In one embodiment, reaction (a) is e.g. carried out at room temperature (e.g. about 18 to about 25 0C), and/or for a reaction time of from 2 to 48 hours or 6 to 48 hours such as 12 to 24 hours, for example about 15-18 hours. For example, reaction (a) can optionally be carried out under anhydrous conditions.
In an alternative embodiment, the activated compound Ar-C(O)-X1 is for example formed from the carboxylic acid Ar-C(O)-OH by the following reaction (b). In optional reaction (b), the carboxylic acid Ar-C(O)-OH is reacted with with a suitable organic di-substituted carbodiimide (RC1N=C=NRC2 wherein RC1 and RC2 are independent organic groups such as, independently, C-|_4alkyl (e.g. ethyl), cyclohexyl or 3-dimethylaminopropyl), such as 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide or a salt thereof (EDC) e.g. the hydrochloride salt, or such as dicyclohexylcarbodiimide (DCC), and the resulting carbodiimide-acid adduct Ar-C(O)-X1 wherein X1 is O-C(NHRC1)=NRC2 for example then reacts with the amine of formula (II) or a salt thereof.
In one embodiment, this reaction (b) is for example carried out in the presence of a nonaqueous non-alcohol organic solvent (e.g. anhydrous solvent) such as dimethyl formamide (DMF) or acetonitrile and/or e.g. at room temperature and/or e.g. under anhydrous conditions. In one embodiment, the reaction is carried out in the presence of a tertiary amine base such as diisopropylethylamine ('P^NEt = DIPEA).
In one alternative to reaction (b), the reaction Of Ar-C(O)-OH with the carbodiimide is optionally carried out in the presence of 1-hydroxybenzotriazole (HOBT), in which case,
an activated compound
Figure imgf000010_0001
is often the compound which reacts with the amine of formula (II) or a salt thereof (cf. reaction (a) above).
The amine compound of formula (II) can be prepared by hydrogenation of an azide compound of formula (Ilia):
Figure imgf000010_0002
Hydrogenation conditions can for example include H2 / palladium on carbon (e.g. 5 to
10% palladium on carbon), for example with ethanol, methanol, isopropyl alcohol or ethyl acetate (in particular ethanol) solvent.
The azide compound of formula (Ilia) can generally be prepared by reaction of a compound of formula (lllb), wherein X^b js a leaving group displaceable by azide, with a metal azide such as sodium azide, lithium azide or potassium azide:
Figure imgf000010_0003
Typical conditions for the (lllb) to (Ilia) reaction, e.g. with sodium azide or lithium azide, can e.g. include DMSO solvent (e.g. dry) or DMF solvent (e.g. dry) and/or reaction at room temperature. See for example Reference Intermediate 7 and Intermediate 8. In one embodiment, X^b js a chlorine atom (Cl) or an organic sulfonate such as methanesulfonate, trifluoromethanesulfonate CF3SO2- or p-toluenesulfonate, in particular a chlorine atom. Compounds of formula (INb), in particular wherein X3b is Cl or an organic sulfonate, can generally or sometimes be prepared by conversion (e.g. chlorination) of an alcohol compound of formula (IMc). This can for example be by reaction with SOCI2 (thionyl chloride) for chlorination (when X3I3 is Cl), or by reaction with methanesulfonyl chloride (when X3b is methanesulfonate) or p-toluenesulfonyl chloride (when X3b is p-toluenesulfonate). The chlorination reaction with thionyl chloride may require heating, e.g. to about 60 to about 90 0C, for example at about 85 0C:
Figure imgf000011_0001
In one alternative method, azide compounds of formula (Ilia) may be preparable directly from the alcohol compound of formula (lllc). For example, reacting compounds of formula (lllc) with an azide, e.g. sodium azide, in the presence of carbon tetrabromide and triphenylphosphine may give a compound of formula (Ilia) (e.g see Toyota et. al.
Journal of 65(21), 7110-7113):
Figure imgf000011_0002
Figure imgf000011_0003
In another alternative process which is of particular interest, the amine compound of formula (II) or a salt thereof can be prepared directly from the compound of formula (MIb) or a salt thereof (for example wherein X3b is a chlorine atom, for example from the benzenesulfonate salt of the compound of formula (lllb) e.g. wherein X3b is a chlorine atom), without converting (lllb) to the azide compound of formula (Ilia):
Figure imgf000011_0004
For example, this reaction of (lllb) or a salt thereof to the amine (II) or a salt thereof can generally be carried out by reaction of the compound of formula (lllb) (for example wherein X3*3 is or comprises a chlorine atom) with an aminating agent such as lithium hexamethyldisilazide, sodium hexamethyldisilazide or potassium hexamethyldisilazide (in particular lithium hexamethyldisilazide, e.g. with slow mixing/addition), in a suitable non-aqueous non-alcohol organic solvent (e.g. anhydrous solvent) such as tetrahydrofuran, for example at a suitable temperature of, for example, about 25 to about 50 0C, e.g. ca. 30-450C or ca. 35-400C. The reaction is suitably followed by treatment with an aqueous acid such as aqueous hydrochloric acid (e.g. 2-10M, e.g. about 5M), e.g. at a suitable temperature such as from O0C to room temperature, for example at about 1O0C.
In a simplified process, wherein the compound of formula (lllb) has X3b as a chlorine atom, in one suitable embodiment the alcohol compound of formula (I I Ic) or a salt thereof is converted into the amine of formula (II) or a salt thereof, without substantially purifying and/or without substantially isolating the compound of formula (lllb) or the salt thereof wherein X3b is a chlorine atom. In this embodiment, the compound of formula (lllb) or a salt thereof wherein X3b is a chlorine atom can for example be in the form of the benzenesulfonate salt:
Figure imgf000012_0001
(II), orsalt therof
Figure imgf000012_0002
(not purified and/or not isolated)
The alcohol compound of formula (IUc) can be prepared by reduction of an ester compound of formula (IV), wherein Re is Q>\ .4 straight-chain alkyl such as ethyl, in the presence of a reducing agent. The reducing agent can for example be diisobutylaluminium hydride or lithium borohydride (LJBH4). For diisobutylaluminium hydride reduction, the reduction can e.g. be in dichloromethane and/or toluene solvent (e.g. see Intermediate 4 or Reference Intermediate 3 herein) and/or can e.g. be carried out at about 0°C and/or under nitrogen and/or with careful temperature control and/or with gradual addition or mixing of diisobutylaluminium hydride. For lithium borohydride (IJBH4), see for example Intermediate 4, Alternative Synthesis B herein:
Figure imgf000013_0001
Compounds of formula (IV) can generally be prepared according to a method, for example as described by Yu et. al. in J. Med Chem., 2001, 44, 1025-1027, by reaction of a compound of formula (V) with an amine of formula R3NH2 or a salt thereof such as a hydrochloride salt thereof. The reaction is for example carried out in the presence of a tertiary amine base such as triethylamine or N.N-diisopropylethylamine, and/or in an organic solvent such as 1-methyl-2-pyrrolidinone (NMP), ethanol, dioxane or acetonitrile. The reaction may comprise heating e.g. heating to ca. 60-1800C or 80-1800C, for example ca. 60-100 0C (e.g. ca. 80-900C) or ca. 110-160 0C, for example depending on the reflux temperature or boiling point of the solvent(s) used. For an example process, see e.g. Intermediate 2 herein, or see e.g. the process shown in Yu et. al. in J. Med. Chem., 2001 , 44, 1025-1027 (see steps c+d of Scheme 1 therein):
Figure imgf000013_0002
(V) (IV)
The amine of formula R3NH2 is commercially available, e.g. from Combi-Blocks Inc. and/or from Peakdale Molecular Ltd. See Intermediate 12 one possible synthesis of the hydrochloride salt of R3NH2-
In one embodiment, a compound of formula (V) is for example prepared by reaction of a compound of formula (Vl) with (R2)(CI)C=C(CC>2Re)2 (a compound of formula (XIV)), followed by reaction with phosphorous oxychloride (POCI3). The compound
(R2)(CI)C=C(CC>2Re)2 can for example be diethyl (i-chloropropylidene)propanedioate (wherein R2 is Et and Re is Et):
Figure imgf000014_0001
In one embodiment of the (Vl) to (V) process, the compound of formula (Vl) is reacted with a dialkyl (i-chloroalkylidene)propanedioate with heating, for example in a suitable organic solvent such as toluene, and for example in the presence of a suitable base such as triethylamine, e.g. at a suitable temperature such as the reflux temperature of the solvent. The conditions for the reaction of the intermediate [formed from (Vl) and the dialkyl (i-chloroalkylidene)propanedioate] with phosphorous oxychloride (POCI3) can include heating, e.g. heating at the reflux temperature of phosphorous oxychloride.
For an example of the compound (Vl) to compound (V) process, see for example the Intermediate 1 synthesis hereinafter wherein R^ = ethyl and R^ = ethyl and Re is ethyl.
In one embodiment, a compound (R2)(CI)C=C(Cθ2Re)2 (a compound of formula (XIV)) is prepared by reaction of a compound of formula (XV), with phosphorus oxychloride (POCI3) in the presence of a suitable base such as tributylamine, at a suitable temperature su
Figure imgf000014_0002
(XV) (XIV)
In one embodiment, a compound of formula (XV) is prepared by reaction of a dialkyl malonate of formula (XVI) with magnesium chloride and a suitable non-aqueous base such as triethylamine, in a suitable solvent (e.g. anhydrous solvent) such as acetonitrile, at a suitable temperature such as ca. 5-10 0C, followed by addition of an acid chloride of formula (XVII) (propanoyl chloride when R^ is ethyl), at a suitable temperature such as between 10 0C and room temperature. In one embodiment, the reaction is for example carried out under anhydrous conditions:
Figure imgf000014_0003
The compounds of formulae (XVI) and (XVII) where Re and R2 respectively represent ethyl are commercially available, e.g. from Aldrich or elsewhere.
The desired amino pyrazole of formula (Vl), which (for R1 is ethyl) is 1 -ethyl- 1 H-pyrazol- 5-amine, is commercially available, e.g. is commercially available from one or more of the following suppliers: Aldrich Chemical Company Inc., Albemarle Corporation, Art- Chem GmbH, Enamine and/or TimTec Corporation. However, alternatively, the amino pyrazole (Vl) is optionally prepared, for example, by reaction of cyanoethyl hydrazine with a suitable aldehyde of formula R40CHO in a solvent such as ethanol, with heating, followed by reduction, for example reduction with sodium in a suitable solvent such as t-butanol. (See for example the method(s) described by Dorgan et. al. in J. Chem. Soc, Perkin Trans. 1, (4), 938-42; 1980.) R40 should be chosen so as to contain one less carbon atom than R^ , so that for example R40 = methyl will afford R1 = ethyl.
Figure imgf000015_0001
( Vl )
In an alternative to the reaction of compound (V) to prepare a 4-amino 5-ester pyrazolopyridine compound of Formula (IV), instead of the 4-chloro substituent in the compound of formula (V), it is likely that a different non-fluorine halogen atom such as a bromine atom, or another suitable leaving group which is displaceable by an amine of formula R3NH2, can sometimes be used at the 4-position of the pyrazolopyridine. In one embodiment, the leaving group displaceable by the amine is for example R^A1 jn the compound of formula (Va) shown below, wherein R*-A js a bromine atom or an alkoxy group OR35 such as OCi^alkyl (in particular OEt) or a group -O-S(O)2-R37. Here, R37 can e.g. be C-^alkyl such as methyl, C^fluoroalkyl such as CF3, phenyl, or
4-methyl-phenyl. The reaction of the compound of formula (Va) with the amine of formula R3NH2 or a salt thereof is optionally carried out with or without solvent, and e.g. may comprise heating:
Figure imgf000016_0001
(Va) (IV)
Compounds (Va) wherein RLA is the group -O-S(O)2-R37 are "kely to be preparable by reaction of the corresponding 4-hydroxy-pyrazolopyridine with R37_S(O)2-CI such as CF3-S(O)2-CI. the compound of formula (Va) wherein RLA is OEt and Re is Et is likely to be preparable, e.g. via the following reaction from the 4-chloro-pyrazolopyridine (V):
Figure imgf000016_0002
Process Summary
The present invention therefore also provides a method of (process for) preparing a compound of formula (I) le salt) thereof:
Figure imgf000016_0003
, using the final step of Process A1 generally as described hereinabove,
and optionally converting the compound of formula (I) into a salt thereof e.g. a pharmaceutically acceptable salt thereof.
Therefore, another aspect of the present invention provides a process for preparing a compound of formula (I) or a salt (e.g. pharmaceutically acceptable salt) thereof:
Figure imgf000017_0001
wherein R1, R2, R3, R4, R5, Ra and R*3 are as defined herein, which process comprises:
(A) reacting an amine of formula (II) or a salt thereof
Figure imgf000017_0002
with a compound Ar-C(O)-X1 , wherein X1 is a leaving group substitutable by the NH2 amine moiety of the compound of formula (II);
and optionally converting the compound of formula (I) into a salt thereof e.g. a pharmaceutically acceptable salt thereof.
In this process aspect of the invention, preferred, suitable or optional features of process (A), can be as described above for Process A (e.g. those described above as preferred, suitable, optional or example features of this process), with all necessary changes being made.
In process (A), In the reaction of (II) to (I), a compound Ar-C(O)-X1 can for example be the acid chloride Ar-C(O)CI (i.e. X1 is a chlorine atom (Cl)). Alternatively, the compound Ar-C(O)-X1 can be an activated derivative of the carboxylic acid Ar-C(O)-OH wherein the leaving group X1 is
Figure imgf000017_0003
Alternatively, in the compound Ar-C(O)-X1 , X1 can be O-C(NHRC1 )=NRC2 wherein RC1 and RC2 are independent organic groups such as, independently, Ci_4alkyl (e.g. ethyl), cyclohexyl or 3-dimethylaminopropyl. In one embodiment, Rc1 and RC2 can e.g. both be cyclohexyl; or one of RC1 and RC2 js ethyl and the other of Rc1 and Rc2 is 3-dimethylaminopropyl.
The present invention also provides: (B) a process for preparing a pharmaceutically acceptable salt of a compound of formula (I) comprising conversion of the compound of formula (I) or a salt thereof into a or the desired pharmaceutically acceptable salt of the compound of formula (I). Such processes can for example be as described herein, e.g. as described in the Salts section hereinabove. For example, a pharmaceutically acceptable salt can be an acid addition salt. In one embodiment, a pharmaceutically acceptable acid addition salt is optionally prepared by reaction of a compound of formula (I) with a suitable inorganic or organic acid (e.g. as described hereinabove).
The present invention also provides a compound of formula (I) or a salt thereof, prepared by (or obtainable by) a process as defined herein.
MEDICAL USES
The present invention also provides a compound of formula (I) or a pharmaceutically acceptable salt thereof for use as an active therapeutic substance in a mammal such as a human. The compound or salt can be for use in the treatment and/or prophylaxis of any of the diseases / conditions described herein (e.g. for use in the treatment and/or prophylaxis of an inflammatory and/or allergic disease in a mammal such as a human; or e.g. for use in the treatment and/or prophylaxis of cognitive impairment (e.g. in a neurological disorder such as Alzheimer's disease) or depression in a mammal such as a human) and/or can be for use as a phosphodiesterase 4 (PDE4) inhibitor. "Therapy" may include treatment and/or prophylaxis.
The compound or salt is in particular for use in the treatment and/or prophylaxis of an inflammatory and/or allergic skin disease, such as atopic dermatitis or psoriasis (in particular atopic dermatitis), in a mammal such as a human.
Also provided is the use of a compound of formula (I) or a pharmaceutically acceptable salt thereof in the manufacture of a medicament (e.g. pharmaceutical composition) for the treatment and/or prophylaxis of any of the diseases / conditions described herein in a mammal such as a human, e.g. for the treatment and/or prophylaxis of an inflammatory and/or allergic disease in a mammal such as a human, or e.g. for the treatment and/or prophylaxis of cognitive impairment (e.g. in a neurological disorder such as Alzheimer's disease) or depression in a mammal.
Also provided is a method of treatment and/or prophylaxis of any of the diseases / conditions described herein in a mammal (e.g. human) in need thereof, e.g. a method of treatment and/or prophylaxis of an inflammatory and/or allergic disease, cognitive impairment (e.g. in a neurological disorder such as Alzheimer's disease or schizophrenia) or depression in a mammal (e.g. human) in need thereof, which method comprises administering to the mammal (e.g. human) a therapeutically effective amount of a compound of formula (I) as herein defined or a pharmaceutically acceptable salt thereof.
Phosphodiesterase 4 inhibitors are thought to be, or may be, potentially useful in the treatment and/or prophylaxis of a variety of diseases / conditions, especially inflammatory and/or allergic diseases, in a mammal such as a human, for example: asthma, chronic obstructive pulmonary disease (COPD) (e.g. chronic bronchitis and/or emphysema), atopic dermatitis, urticaria, rhinitis (e.g. allergic rhinitis), allergic conjunctivitis, vernal conjunctivitis, eosinophilic granuloma, psoriasis, rheumatoid arthritis, septic shock, ulcerative colitis, Crohn's disease, reperfusion injury of the myocardium and brain, chronic glomerulonephritis, endotoxic shock, adult respiratory distress syndrome, multiple sclerosis, cognitive impairment (e.g. in a neurological disorder such as Alzheimer's disease or schizophrenia), depression, or pain (e.g. inflammatory pain). Ulcerative colitis and/or Crohn's disease are collectively often referred to as inflammatory bowel disease.
Phosphodiesterase 4 inhibitors, e.g. perhaps the compound or salt of the invention, may also be potentially useful in the treatment and/or prophylaxis of anxiety in a mammal such as a human.
In the treatment and/or prophylaxis, with the compound or salt of the invention, the inflammatory and/or allergic disease can for example be chronic obstructive pulmonary disease (COPD), asthma, rheumatoid arthritis, rhinitis (e.g. allergic rhinitis), psoriasis or atopic dermatitis in a mammal (e.g. human).
In particular, with the compound or salt of the invention, the treatment and/or prophylaxis is of an inflammatory and/or allergic skin disease such as psoriasis or atopic dermatitis (in particular atopic dermatitis) in a mammal (e.g. human).
Suitably, the treatment and/or prophylaxis is of atopic dermatitis in a mammal such as a human or pig, in particular in a human, in particular in a human aged 21 years or less, e.g. 18 years or less. For treatment and/or prophylaxis of atopic dermatitis in a mammal, external topical administration to the mammal of the compound of formula (I) or a pharmaceutically acceptable salt thereof (e.g. topical administration to the skin e,g. to skin affected by the atopic dermatitis) is suitably used, though alternatively oral or parenteral administration can be used. For treatment and/or prophylaxis of atopic dermatitis, inhaled administration is usually not suitable.
"Atopic dermatitis" has been proposed to include two general sub-classes: (1) an "allergic (extrinsic)" type of atopic dermatitis which generally occurs in the context of sensitization to environmental allergens and/or which is generally accompanied by elevated serum IgE levels; and (2) an "non-allergic (intrinsic)" type of atopic dermatitis generally with little or no detectable sensitization and/or generally with normal or low serum IgE levels (N. Novak et al., J. Allergy Clin. Immunol., 2003, 112, 252-262; and T.C. Roos et al., Drugs, 2004, 64(23), 2639-2666, see e.g. pages 2640-2641). The compound of formula (I) or the pharmaceutically acceptable salt thereof can therefore be for the treatment and/or prophylaxis of allergic (extrinsic) atopic dermatitis and/or non- allergic (intrinsic) atopic dermatitis in a mammal (e.g. human or pig, in particular a human).
"External topical" administration means topical administration to an external body part (i.e. excluding, for example, the lung or mouth, but including the lips), in particular excluding the eye.
"External topical" administration is for example topical administration to the skin, for example to the skin of an arm, hand, leg, foot, head (e.g. face), neck and/or torso of a mammal such as a human. External topical administration can for example be to those parts of a mammal's skin affected by or susceptible to atopic dermatitis.
For the use of PDE4 inhibitors in atopic dermatitis, see for example:
- J. M. Hanifin et al., "Type 4 phosphodiesterase inhibitors have clinical and in vitro antiinflammatory effects in atopic dermatitis", J. Invest. Dermatol., 1996, 107(1), 51-56; which reports reductions of inflammatory parameters in atopic dermatitis patients treated with PDE4 inhibitor CP80,633 (0.5% ointment, twice daily topical application); - C.E.M. Griffiths et al., "Randomized comparison of the type 4 phosphodiesterase inhibitor cipamfylline cream, cream vehicle and hydrocortisone 17-butyrate cream for the treatment of atopic dermatitis", Br. J. Dermatol., 2002, 147(2), 299-307, which reports that cipamfylline (0.15%) cream is significantly more effective than vehicle, but significantly less effective than hydrocortisone 17-butyrate (0.1%) cream, in the treatment of atopic dermatitis patients;
- T.C. Roos et al., "Recent advances in treatment strategies for atopic dermatitis", Drugs, 2004, 64(23), 2639-2666 (see e.g. page 2657 and refs. 201-209 therein);
- A.M.Doherty, Current Opinion Chem. Biol., 1999, 3(4), 466-473 (e.g. see p. 470); and
- HJ. Dyke et al., Expert Opinion Invest. Drugs, 2002, 11(1), 1-13 (e.g. see p.7 and refs. 74, 75 and 76 cited therein); and references cited in the above references. For the use of the PDE4 inhibitors SB 207499 (cilomilast) and AWD 12-281 in mouse models of the allergic type of dermatitis, see W. Baumer et al., Eur. J. Pharmacol., 2002, 446, 195-200 and W. Baumer et al., J. Pharmacy Pharmacol., 2003, 55, 1107-1114.
PDE4 inhibitors, for example cilomilast and roflumilast, are thought to be effective in the treatment of COPD. For example, see S.L. Wolda, Emerging Drugs, 2000, 5(3), 309- 319; Z. Huang et al., Current Opinion in Chemical Biology, 2001 , 5: 432-438; H.J. Dyke et al., Expert Opinion on Investigational Drugs, January 2002, 11 (1 ), 1 -13; C.Burnouf et al., Current Pharmaceutical Design, 2002, 8(14), 1255-1296; A.M.Doherty, Current Opinion Chem. Biol., 1999, 3(4), 466-473; A.M. Vignola, Respiratory Medicine, 2004, 98, 495-503; D. Spina, Drugs, 2003, 63(23), 2575-2594; and references cited in the aforementioned publications; and G. Krishna et al., Expert Opinion on Investigational Drugs, 2004, 13(3), 255-267 (see especially pp. 259-261 and refs. 102-111 and 201 therein).
The PDE4 inhibitor cilomilast (Ariflo ™) at 15 mg orally twice daily appears to improve forced expiratory volume in 1s (FEV1) in COPD patients (C.H.Compton et al., The Lancet, 2001 , vol.358, 265-270), and appears to have antiinflammatory effects in COPD patients (E.Gamble et al., Am. J. Respir. Crit. Care Med., 2003, 168, 976-982). On cilomilast, see also R.D. Border et al., Chest, 2003, vol. 124 Suppl. 4, p.170S (abstract) and J. D. Eddleston et al., Am. J. Respir. Crit. Care Med., 2001 , 163, A277 (abstract). The PDE4 inhibitor roflumilast appears to show small improvements in FEVi in COPD patients (see BJ. Lipworth, The Lancet, 2005, 365, 167-175, and refs 49-50 therein).
COPD is often characterised by the presence of airflow obstruction due to chronic bronchitis and/or emphysema (e.g., see S.L. Wolda, Emerging Drugs, 2000, 5(3), 309- 319).
For treatment and/or prophylaxis of COPD or asthma in a mammal (e.g. human), oral, inhaled or parenteral administration to the mammal of the compound of formula (I) or a pharmaceutically acceptable salt thereof is optionally used, e.g. oral or inhaled administration.
PDE4 inhibitors are thought to be effective in the treatment and/or prophylaxis of asthma (e.g. see M.A.Giembycz, Drugs, Feb. 2000, 59(2), 193-212; Z. Huang et al., Current Opinion in Chemical Biology, 2001, 5: 432-438; HJ. Dyke et al., Expert Opinion on Investigational Drugs, January 2002, 11(1), 1-13; C.Burnouf et al., Current Pharmaceutical Design, 2002, 8(14), 1255-1296; A.M.Doherty, Current Opinion Chem. Biol., 1999, 3(4), 466-473; PJ. Barnes, Nature Reviews - Drug Discovery, October 2004, 831-844; BJ. Lipworth, The Lancet, 2005, 365, 167-175; and references cited in the aforementioned publications). The PDE4 inhibitor roflumilast, given orally at 500 ug once daily for 9 days, is reported to be effective in improving rhinal airflow during the treatment period (compared to placebo), in humans with histories of allergic rhinitis but asymptomatic at screening, and who were challenged with intranasal allergen provocation (pollen extracts) daily beginning the third day of treatment and each time approx. 2 hours after study drug administration (see B.M. Schmidt et al., J. Allergy & Clinical Immunology, 108(4), 2001, 530-536).
For treatment and/or prophylaxis of rhinitis (e.g. allergic rhinitis), intranasal, oral or parenteral administration of the compound of formula (I) or a pharmaceutically acceptable salt thereof is optionally used.
In one optional embodiment, the compound of formula (I) or the pharmaceutically acceptable salt thereof is for the treatment and/or prophylaxis of rhinitis, such as allergic rhinitis (e.g. seasonal allergic rhinitis or perennial allergic rhinitis) or non-allergic rhinitis (e.g. vasomotor rhinitis), in a mammal such as a human.
PDE4 inhibitors are thought to be, or may be, effective in the treatment of rheumatoid arthritis and multiple sclerosis (e.g. see H.J. Dyke et al., Expert Opinion on Investigational Drugs, January 2002, 11(1), 1-13; C.Bumouf et al., Current Pharmaceutical Design, 2002, 8(14), 1255-1296; and A.M.Doherty, Current Opinion Chem. Biol., 1999, 3(4), 466-473; and references cited in these publications). For rheumatoid arthritis, oral or parenteral administration is optionally used.
PDE4 inhibitors have been suggested as having analgesic properties and thus being effective in the treatment of pain (A.Kumar et al., Indian J. Exp. Biol., 2000, 38(1), 26- 30).
In the invention, the treatment and/or prophylaxis can be of cognitive impairment e.g. cognitive impairment in a neurological disorder (such as Alzheimer's disease or schizophrenia); and/or administration of the compound or salt can optionally be oral. For example, the treatment and/or prophylaxis can comprise cognitive enhancement e.g. in a neurological disorder. For cognition background, see for example: H.T.Zhang et al. in: Psychopharmacology, June 2000, 150(3), 311-316 and Neuropsychopharmacology, 2000, 23(2), 198-204; and T. Egawa et al., Japanese J. Pharmacol., 1997, 75(3), 275- 81.
PDE4 inhibitors such as rolipram have been suggested as having antidepressant properties (e.g. J. Zhu et al., CNS Drug Reviews, 2001 , 7(4), 387-398; O'Donnell, Expert Opinion on Investigational Drugs, 2000, 9(3), 621-625; HT. Zhang et al.,
Neuropsychopharmacology, October 2002, 27(4), 587-595; J. M. O'Donnell and H.-T. Zhang, Trends Pharmacol. Sci., March 2004, 25(3), 158-163; and T.E.Renau, Curr. Opinion Invest Drugs, 2004, 5(1), 34-39).
PDE4 inhibition has been suggested for the treatment of inflammatory bowel disease (e.g. ulcerative colitis and/or Crohn's disease), see K.H. Banner and M.A.Trevethick, Trends Pharmacol. Sci., August 2004, 25(8), 430-436.
PHARMACEUTICAL COMPOSITIONS, ROUTES OF ADMINISTRATION, AND DOSAGE REGIMENS
For use in medicine, the compounds or salts of the present invention are usually administered as a pharmaceutical composition.
The present invention therefore provides in a further aspect a pharmaceutical composition comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof and one or more pharmaceutically acceptable carriers and/or excipients.
The pharmaceutical composition can be for use in the treatment and/or prophylaxis of any of the conditions described herein, for example chronic obstructive pulmonary disease (COPD)1 asthma, rheumatoid arthritis, allergic rhinitis, psoriasis or atopic dermatitis in a mammal (e.g. human).
The invention also provides a method of preparing a pharmaceutical composition comprising a compound of formula (I), as herein defined, or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable carriers and/or excipients, the method comprising mixing the compound or salt with the one or more pharmaceutically acceptable carriers and/or excipients.
The invention also provides a pharmaceutical composition prepared by said method.
The compounds of formula (I) and/or the pharmaceutical composition may be administered, for example, by external topical (e.g. skin topical), oral, parenteral (e.g. intravenous, subcutaneous, or intramuscular), inhaled, or nasal administration. Inhaled administration involves topical administration to the lung e.g. by aerosol or dry powder composition.
Accordingly, the pharmaceutical composition can be suitable for (e.g. adapted for) external topical (e.g. skin topical), oral, parenteral (e.g. intravenous, subcutaneous, or intramuscular), inhaled, or nasal administration, e.g. to a mammal such as a human. The pharmaceutical composition can for example be suitable for external topical (e.g. skin topical) or oral administration, e.g. to a mammal such as a human.
The pharmaceutical composition can optionally be in unit dose form. The unit dose form can for example be:
(a) a tablet or capsule for oral administration e.g. for oral administration to a human; (b) a rupturable or peel-openable sealed dose container containing a dry powder inhalable pharmaceutical composition (e.g. a plurality of which are usually disposed inside a suitable inhalation device); (c) a vial, ampoule or filled syringe for parenteral administration e.g. comprising a solution or suspension of the compound or pharmaceutically acceptable salt in a suitable carrier such as an aqueous carrier or e.g. containing a lyophilised parenteral pharmaceutical composition (the vial or ampoule can optionally be manufactured using a blow-fill-seal process).
Alternatively, the composition can be in a form adapted for the administration of varying amounts of composition as desired by the user, such as a spreadable or sprayable external topical composition such as a cream, an ointment, a gel, or a liquid.
Pharmaceutical compositions suitable for external topical administration
The pharmaceutical composition of the invention can for example be suitable for (e.g. adapted for) external topical administration (e.g. skin topical administration, i.e. topical administration to the skin), for example to a mammal such as a human. The pharmaceutical composition suitable for external topical administration can suitably be for the treatment and/or prophylaxis of atopic dermatitis in a mammal such as a human, e.g. by external topical administration.
"External topical administration" is defined above under the "medical uses" section. External topical administration can for example be to those parts of the skin affected by or susceptible to the disease or condition e.g. atopic dermatitis, in particular in a mammal (e.g. human) suffering from or susceptible to atopic dermatitis.
An external-topical pharmaceutical composition, e.g. skin topical pharmaceutical composition, can for example be an ointment, a cream (usually an oil-in-water or water- in-oil pharmaceutical composition, usually an emulsion), an aqueous gel, or a microemulsion. The pharmaceutical composition can alternatively be a DMSO- containing solution such as a DMSO/acetone solution or DMSO/water solution (DMSO = dimethyl sulfoxide); a DMSO-containing solution can be used for experimental animal tests, but is not usually desirable for use in humans.
An external-topical pharmaceutical composition, e.g. skin topical pharmaceutical composition, of particular interest is an ointment.
In the external-topical pharmaceutical composition, e.g. an ointment or an oil-in-water or water-in-oil composition, the compound of formula (I) [Λ/-{[1 ,6-diethyl-4-(tetrahydro-2H- pyran-4-ylamino)-1/-/-pyrazolo[3,4-6]pyridin-5-yl]methyl}-3-methyl-5- isoxazolecarboxamide] or the pharmaceutically acceptable salt thereof can be present in 0.1% to 10%, such as 0.2% to 10%, or 0.2% to 5%, or 0.5% to 5%, in particular 1% to 10% (e.g. about 2%, about 4% or about 6%), or 1% to 5% (e.g. 1.5% to 5% or 1.5% to 5%, such as about 2% or about 4%), or 0.5% to 3% (e.g. 0.5% or about 2%), or 1% to 3% (e.g. about 2%), by weight of the composition (w/w).
In the external-topical pharmaceutical composition of the invention, the compound of formula (I) or the pharmaceutically acceptable salt thereof can in particular comprise or be the compound (i.e. as the "free base" form).
In one optional embodiment, the compound of formula (I) or the pharmaceutically acceptable salt thereof can optionally be in a particle-size-reduced form, for example obtained or obtainable by micronisation. This can be, for example, for use in a pharmaceutical composition suitable for (e.g. adapted for) external topical (e.g. skin topical) administration. See the Particle size reduction sub-section herein, within the lnhalable pharmaceutical compositions section, for more details.
Aqueous solubility: A preliminary screen, which can aim to estimate roughly the aqueous solubility of a compound or salt of the invention, can include (as an approximate summary): (i) creating a ca. 1OmM solution of the compound in DMSO, (ii) diluting a portion of this DMSO solution by mixing about 19 parts by volume of pH 7.4 aqueous phosphate buffered saline (PBS) buffer with 1 part by volume of the ca. 1OmM DMSO solution, (iii) "filtering" the mixture with the aid of centrifugation, and then (iv) measuring the concentration of the dissolved compound in the "filtrate". Although some DMSO (about 5% by volume) is usually present in this solubility screen "filtrate", the results can be a very approximate estimate of aqueous solubility, e.g. at room temperature.
ϋpophilicity: The clogP (calculated Jog of the octanol/water partition coeficient (P)) of a particular compound or salt of the invention can estimate the lipophilicity of the compound or salt.
Solubilising and/or skin-penetration-enhancing agents: An external-topical pharmaceutical composition, e.g. an ointment or an oil-in-water cream or water-in-oil cream, can for example include an agent which acts as a skin-penetration enhancer for and/or a solubiliser of the compound of formula (I) or the salt thereof. The skin- penetration-enhancing- and/or solubilising- agent can for example be propylene glycol, diethylene glycol monoethyl ether (e.g. TRANSCUTOL ™) and/or caprylocaproyl macrogolglycerides (e.g. LABRASOL ™), in particular propylene glycol. The solubiliser and/or skin-penetration enhancer suitably does not comprise DMSO. The solubiliser and/or skin-penetration enhancer is in particular both a solubiliser and skin-penetration enhancer, and/or can be present in 0.5% to 50%, in particular 5% to 50%, for example 7% to 30%, such as 7% to 25%, e.g. about 10% to about 20% (e.g. about 10% or about 20%), by weight of the composition (w/w). The skin-penetration enhancer is for delivery of the compound of formula (I) or salt thereof ("active agent" or "drug") through the skin. Solubilization of the drug also helps. The solubilising and/or skin-penetration-enhancing agents should ideally (a) be safe and/or tolerable, (b) have as low a potential for skin irritancy as possible consistent with being an effective skin penetration enhancer, and (c) be compatibile with the active pharmaceutical ingredient. Note that the agent optionally functions both as a solubilising agent and a skin-penetration-enhancing agent.
Surfactants: An external-topical pharmaceutical composition, e.g. an ointment or an oil- in-water cream or water-in-oil cream, can, in one embodiment, include a surfactant (e.g. as an emulsifier), for example for achieving emulsification of compositions having two or more phases. The total surfactant content can for example be 0.3% to 20%, e.g. 0.5% to 15% or 0.5% to 12% or 0.5% to 10% or 1 % to 12% or 3% to 10%, by weight of the composition (w/w). The surfactant can for example comprise one or more of the following: a polyoxyl C-|2-22a'M ether (e.g. a polyoxyl C-|4_20a'kyl ether such as polyoxyl cetyl ether or polyoxyl stearyl ether) (e.g. present at 0.5% to 10% w/w, e.g. 2.5% to 10% w/w such as about 5% to about 8% w/w), glycerol monostearate (e.g. Arlacel 165 ™) (e.g. present at 0.5% to 10% w/w, e.g. about 2% w/w), sorbitan monostearate (e.g. Span 60 ™) (e.g. present at 0.05% to 10% w/w, e.g. about 1% w/w), cetyl alcohol and/or stearyl alcohol (e.g. wherein the total of any cetyl alcohol and any stearyl alcohol present is 0.1% to 15% w/w, e.g. 1% to 10% w/w such as about 2% to about 5% w/w), and sodium dodecyl sulphate (SDS) (e.g. present at 0.3% to 2% w/w such as about 1% w/w). Polyoxyl stearyl ether (steareth) can e.g. be polyoxyl 2 stearyl ether (steareth 2) or polyoxyl 21 stearyl ether (steareth 21).
DMSO-containing solutions: One possible external-topical pharmaceutical composition is a solution of the compound of formula (I) or the pharmaceutically acceptable salt thereof present at ca. 0.5% to ca. 2.5% w/w in a DMSO-containing solvent such as in DMSO/acetone or in DMSO/water; for example a solution of the compound or salt present at ca. 0.5% to ca. 2.5% w/w in DMSO/acetone (1:1). DMSO-containing solutions, often being capable of high skin penetration, are often good experimental preclinical formulations for use in animals e.g. pigs, but their likely skin irritancy generally make them less suitable for use in humans such as patients, e.g. atopic dermatitis patients.
Ointments (and oil phase in ointments and creams):
An external-topical pharmaceutical composition can for example be an ointment or an oil-in-water cream or water-in-oil cream. An ointment is of particular interest. The ointment or cream typically contains an oil phase (oily ointment base). The oil phase (oily ointment base) typically comprises an oil and/or a fat, for example of a consistency suitable for skin-spreadability. The oil phase (oily ointment base) can for example comprise (e.g. be) an oil, wherein the oil comprises (e.g. is) white soft paraffin (white petrolatum) and/or a silicone oil and/or a mineral oil (such as liquid paraffin). (Mineral oil can also be used as a solubiliser and/or emollient).
In particular, the oil phase (oily ointment base) comprises (e.g. is) an oil, wherein the oil comprises (e.g. is) white soft paraffin (white petrolatum) and/or a silicone oil.
Preferably, the external-topical pharmaceutical composition is an ointment, and the oil phase (oily ointment base) comprises (e.g. is) an oil, wherein the oil comprises (e.g. consists essentially of) white soft paraffin (white petrolatum) and a silicone oil.
The white soft paraffin (white petrolatum), e.g. in an ointment or cream, can be of various grades, for example (for Penreco supplier) Penreco Regent White ™ grade, Penreco Snow White ™ grade, or Penreco Ultima White ™ grade; in particular high melting point white petrolatum (high melting point white soft paraffin) (e.g. of Penreco Ultima White ™ grade). The white petrolatum can be present at 25% to 99.9% w/w or 45% to 99.5% w/w or 50% to 99.5% w/w or 45% to 99% w/w or 50% to 99% w/w or 45% to 85% w/w or 45% to 75% w/w (i.e. by weight of the composition).
The silicone oil, e.g. in an ointment or cream, in particular in an ointment, can for example be present at: 5% to 60 % w/w such as 5% to 50% w/w, in particular 10% to 50% w/w such as 15% to 40% w/w, suitably 20% to 35% w/w such as about 25% w/w (measured as the total silicone oil content, by weight of the composition).
The silicone oil can be solid or liquid. The silicone oil, e.g. in an ointment or cream, can for example comprise (e.g. be): decamethyl-cyclopentasiloxane (e.g. ST-Cyclomethicone 5-NF ™, available from Dow Corning), stearoxytrimethylsilane [Me(CH2)i7θ-SiMe3], polydimethylsiloxane (dimethicone), hexamethyldisiloxane (e.g. ca. 0.65 cSt viscosity at 25°C), octamethyltrisiloxane (e.g. ca. 1.0 cSt viscosity at 25°C), decamethyltetrasiloxane, dodecamethylpentasiloxane, or hydroxy-terminated polydimethylsiloxane (e.g. ST-Dimethiconol 40 ™, Dow Corning), or mixtures of any of the foregoing. The silicone oil, e.g. in an ointment or cream, can in particular comprise (e.g. be): decamethyl-cyclopentasiloxane, stearoxytrimethylsilane [IvIe(CI-^)-17θ-SiMe3], or polydimethylsiloxane (dimethicone), or mixtures of any of the foregoing. Preferably, the silicone oil, e.g. in an ointment or cream, can comprise (e.g. be) decamethyl- cyclopentasiloxane.
The decamethyl-cyclopentasiloxane can be ST-Cyclomethicone 5-NF ™, available from Dow Corning, and which is described by Dow Corning as being a polydimethyl- cyclosiloxane having a decamethyl-cyclopentasiloxane content of >95% and having a octamethyl-cyclotetrasiloxane content of <1.0%. The decamethyl-cyclopentasiloxane can for example be present at 5% to 60 % w/w such as 5% to 50% w/w, in particular 10% to 50% w/w such as 15% to 40% w/w, suitably 20% to 35% w/w such as about 25% w/w (i.e. by weight of the composition).
Stearoxytrimethylsilane [Me(CH2)i7θ-SiMe3] can for example be present as a mixture of stearoxytrimethylsilane and stearyl alcohol for example Silky Wax 10 ™ which is available from Dow Corning. Stearoxytrimethylsilane (and/or stearoxytrimethylsilane and stearyl alcohol mixture), e.g. in an ointment or cream e.g. ointment, can for example be present at 1% to 30% w/w or 2% to 20% w/w or 5% to 20% w/w such as about 10% w/w.
Polydimethylsiloxane (dimethicone), whose structure is given in the Merck Index 12th edition 1996 as Me3Si-O-[-Si(CH3)2-O-]n-SiMe3, can for example have a viscosity at
25°C of from about 20 to about 12500 cSt (centistokes), such as a viscosity at 250C of from about 20 to about 350 cSt or from about 20 to about 100 cSt. For example, polydimethylsiloxane (dimethicone) can have a viscosity at 250C of: 20 cSt (±10%) ("dimethicone 20"), 100 cSt (±5%), 350 cSt (±5%) ("dimethicone 350"), 1000 cSt (±5%), or 12500 cSt (±5%); grades of polydimethylsiloxane having these five different viscosities are available from Dow Corning as Q7-9120 ™ Silicone Fluid. Polydimethylsiloxane (dimethicone), e.g. in an ointment, can e.g. be present at 0.1% to 15% w/w such as 0.5% to 10% w/w e.g. 0.5% to 5% w/w.
Microcrystalline wax or beeswax or beeswax substitute can alternatively or additionally be used as an oil / fat in the oil phase.
Alternatively or additionally, one or more fats like straight or branched chain mono- or di- alkyl esters such as isopropyl myristate, isopropyl palmitate, diisopropyl adipate, isocetyl stearate, isostearyl isostearate, decyl oleate, butyl stearate, 2-ethylhexyl palmitate, propylene glycol diester of coconut fatty acids, or a mixed ester of 2-ethyl hexanoic acid with a blend of cetyl or stearyl alcohols (e.g. known as Crodamol CAP) may be used in the oil phase (some of these are also solubilisers and/or surfactants). These may be used singly or in combination depending on the properties required.
The oil phase (oily ointment base) can for example be present at 25% to 99.9% w/w or 25% to 99.5% w/w or 25% to 85% w/w (in particular 45% to 99.5% w/w or 45% to 99% w/w, or 50% to 99.5% w/w or 50% to 99% w/w or 50% to 80% w/w, or 70% to 99.5% w/w or 80% to 99.5% w/w) in an ointment (e.g. as an emulsion, or e.g. as a homogeneous single phase (which does not exclude the compound or salt being at least partly in suspension)).
The oil phase (oily ointment base) can for example be present at 25% to 85% w/w (e.g. 35% to 70% w/w) in an water-in-oil cream (e.g. emulsion), or at 8% to 55% w/w (e.g. 10% to 45% w/w) in an oil-in-water cream (e.g. emulsion). Example ointments:
One particular example of an external-topical pharmaceutical composition is an ointment comprising (e.g. consisting essentially of): - the compound of formula (I) [Λ/-{[1 ,6-diethyl-4-(tetrahydro-2H-pyran~4-ylamino)-1 H- pyrazolo[3,4-b]pyridin-5-yl]methyl}-3-methyl-5-isoxazolecarboxamide] or the pharmaceutically acceptable salt thereof present at 0.5% to 10% w/w (in particular 1 % to 10% w/w or 1% to 5% w/w, or 1% to 3% w/w, e.g. about 2% w/w);
- white petrolatum present at 25% to 99.9% w/w, in particular 45% to 99.5% w/w or 50% to 99.5% w/w or 45% to 99% w/w or 50% to 99% w/w or 45% to 85% w/w or 45% to 75% w/w (e.g. about 60-85% w/w, e.g. about 73-75% w/w) (i.e. by weight of the composition); and
- a silicone oil present at: 5% to 60 % w/w such as 5% to 50% w/w, in particular 10% to 50% w/w such as 15% to 40% w/w, suitably 20% to 35% w/w such as about 25% w/w (measured as the total silicone oil content, by weight of the composition).
One preferable example an external-topical pharmaceutical composition is an ointment comprising (e.g. consisting essentially of):
- the compound of formula (I) [Λ/-{[1 ,6-diethyl-4-(tetrahydro-2H-pyran-4-ylamino)-1H- pyrazolo[3,4-jb]pyridin-5-yl]methyl}-3-methyl-5-isoxazolecarboxamide] or the pharmaceutically acceptable salt thereof present at 0.5% to 10% w/w (in particular 1% to 10% w/w or 1 % to 5% w/w, or 1 % to 3% w/w, e.g. about 2% w/w);
- white petrolatum present at 25% to 99.9% w/w, in particular 45% to 99.5% w/w or 50% to 99.5% w/w or 45% to 99% w/w or 50% to 99% w/w or 45% to 85% w/w or 45% to 75% w/w (e.g. about 60-85% w/w, e.g. about 73-75% w/w) (i.e. by weight of the composition); and
- decamethyl-cyclopentasiloxane (e.g. (e.g. ST-Cyclomethicone 5-NF ™) present at 5% to 60 % w/w such as 5% to 50% w/w, in particular 10% to 50% w/w such as 15% to 40% w/w, suitably 20% to 35% w/w such as about 25% w/w (i.e. by weight of the composition).
One example of an external-topical pharmaceutical composition is an ointment comprising: - the compound of formula (I) or the pharmaceutically acceptable salt thereof present at 0.2% to 5% w/w (e.g. 0.5% to 5% w/w, or 1% to 3% w/w, e.g. about 2% w/w);
- an oil phase (oily ointment base) present at 25% to 99% w/w or 50% to 99% w/w or 25% to 85% w/w or 50% to 80% w/w (for example, the oil phase can comprise white petrolatum present at 25 to 75% w/w or 45 to 75% w/w, and optionally also comprising mineral oil (e.g. as solubiliser and emollient) present at 2.5% to 15% w/w such as 4% to 12% w/w);
- one or more surfactants (e.g. polyoxyl stearyl ether) present in total at 0.5% to 10% w/w or 3% to 10% w/w; and - one or more agents acting as a skin-penetration enhancer (in particular acting as both a solubiliser and skin-penetration enhancer and/or in particular a hydrophilic skin- penetration enhancer such as propylene glycol) present in total at 0.5% to 50% w/w, such as 5% to 50% w/w or 7% to 30% w/w (e.g. about 20% w/w, e.g. of propylene glycol); and
- optionally one or more antioxidants (e.g. butylated hydroxyanisole), e.g. present in total at 0.001 to 2% w/w such as 0.02 to 2% w/w; and
- optionally one or more preservatives, e.g. present in total at 0.01 to 4% w/w such as 0.05 to 1% w/w (e.g. methylparaben present at 0.05 to 2% w/w and/or propylparaben present at 0.01 to 2% w/w).
The above example composition, including the oil "phase" and the penetration enhancer, can optionally be a homogeneous single phase. However, in one embodiment of the above example ointment composition, e.g. when using propylene glycol or another hydrophilic solubiliser and penetration enhancer, the oil phase (oily ointment base) and a hydrophilic phase containing the hydrophilic solubiliser and penetration enhancer (e.g. propylene-glycol-containing phase) have been emulsified to form an ointment emulsion.
Ointment compositions having two phases can optionally be prepared using an emulsification process whereby the hydrophilic phase (e.g. propylene-glycol-containing phase) and oil phase are first prepared in separate vessels. The hydrophilic phase can optionally contain a penetration enhancer such as propylene glycol, and optionally some or all of the compound of formula (I) or salt thereof. The oil phase can optionally contain a surfactant. Temperatures of both phases are maintained at elevated temperatures, such as about 45-9O0C or about 45-8O0C or about 55-9O0C or about 55-8O0C (e.g. about 60-650C), or from above 70 to 90 0C, the oil phase temperature being sufficiently high (e.g. from above 70 to 90 0C) to melt the oil phase. While hot, one phase is added to another while mixing, e.g. using a high shear mixer, to effect emulsification, optionally keeping the temperature above 700C such as from above 70 to 90 0C. The resulting ointment emulsion is allowed to cool, e.g. to about 15-350C such as to about 17-3O0C, in particular while the agitation continues e.g. at lower speeds. The ointment emulsion can then optionally be dispensed from the manufacturing vessel and filled into primary packaging, for example tubes or sachets.
Optionally, an ointment can comprise a polyethylene glycol base, e.g. present at 25 to 98% w/w such as 50 to 95% w/w, instead of or as well as an oily ointment base.
Creams: An external-topical pharmaceutical composition can be a cream, e.g. a water- in-oil cream or an oil-in-water cream.
Water-in-oil creams: These usually have an increased aqueous content compared to ointments. In particular, the water-in-oil cream can be a water-in-oil cream emulsion. That is, in particular, in the water-in-oil cream, an oil phase and an aqueous phase can have been emulsified to form a water-in-oil cream emulsion.
One example of an external-topical pharmaceutical composition is a water-in-oil cream (e.g. cream emulsion) comprising:
- the compound of formula (I) or pharmaceutically acceptable salt thereof present at 0.2% to 10% w/w or 0.2% to 5% w/w (in particular 0.5% to 10% w/w or 1% to 10% w/w, such as 0.5% to 5% w/w or 1% to 5% w/w, or 1% to 3% w/w, e.g. about 2% w/w);
- an oil phase (oily ointment base) present at 25% to 85% w/w or 35% to 70% w/w (for example comprising white petrolatum present at 25% to 75% w/w or 30% to 65% w/w
(e.g. about 40% w/w), and optionally also comprising mineral oil (e.g. as solubiliser and emollient) present at 2.5% to 15% w/w or 4% to 12% w/w, e.g. about 10% w/w);
- water present at 2% to 30% w/w, e.g. 5% to 25% or 10% to 22% w/w (e.g. about 20% w/w); - one or more surfactants (e.g. polyoxyl stearyl ether such as polyoxyl 2 stearyl ether) present in total at 0.5% to 12% w/w, such as 3% to 10% w/w (e.g. about 8% w/w); and
- one or more agents acting as a skin-penetration enhancer (in particular acting as both a solubiliser and skin-penetration enhancer and/or in particular a hydrophilic skin- penetration enhancer such as propylene glycol) present in total at 0.5% to 50% w/w, such as 5% to 50% w/w or 7% to 30% w/w (e.g. about 20% w/w, e.g. about 20% w/w of propylene glycol).
The above water-in-oil cream can also optionally comprise:
- one or more antioxidants (e.g. butylated hydroxyanisole), e.g. present in total at 0.001 to 2% w/w such as 0.02 to 2% w/w; and/or
- one or more preservatives, e.g. present in total at 0.01 to 4% w/w such as 0.05 to 1 % w/w (e.g. methylparaben present at 0.05 to 2% w/w and/or propylparaben present at 0.01 to 2% w/w).
Oil-in-water creams: These usually have an increased aqueous content compared to ointments and water-in-oil creams. In particular, the oil-in-water cream can be an oil-in- water cream emulsion. That is, in particular, in the oil-in-water cream, an oil phase and an aqueous phase can have been emulsified to form an oil-in-water cream emulsion.
Oil-in-water creams can for example be high-occlusion creams, wherein, after topical administration to the skin, moisture loss from the skin and/or from the cream is reduced or limited by means of sufficiently high coverage of the skin and/or by providing a sufficient barrier at the site of application.
An oil-in-water cream can in particular contain one or more emollients (hydrating agents), such as silicones (e.g. dimethicone, e.g. dimethicone 360 or dimethicone 20), a high-viscosity wax such as microcrystalline wax, and/or mineral oil. In an oil-in-water cream, suitably there is a sufficiently high water content, for example wherein the water is present in 15% to 60% w/w, 20% to 50% w/w, or 25% to 40% w/w.
One example of an external-topical pharmaceutical composition is a oil-in-water cream (e.g. cream emulsion) comprising:
- the compound of formula (I) or pharmaceutically acceptable salt thereof present at 0.2% to 10% w/w or 0.2% to 5% w/w (in particular 0.5% to 10% w/w or 1% to 10% w/w, such as 0.5% to 5% w/w or 1% to 5% w/w, or 1% to 3% w/w, e.g. about 2% w/w);
- an oil phase (oily ointment base) containing one or more ingredients capable of acting as emollients, the oil phase being present at 5% to 60% w/w or in particular 20% to 60% w/w or 30% to 60% w/w such as 30% to 55% w/w;
- water present at 12% to 75% w/w or 15% to 75% w/w or 15% to 60% w/w, in particular 15% to 50% w/w or 20% to 40% w/w;
- one or more surfactants (e.g. polyoxyl stearyl ether such as polyoxyl 2 stearyl ether) present in total at 0.5% to 12% w/w, e.g. 3% to 10% w/w; and
- one or more agents acting as a skin-penetration enhancer (in particular acting as both a solubiliser and skin-penetration enhancer and/or in particular a hydrophilic skin- penetration enhancer such as propylene glycol) present in total at 0.5% to 50% w/w, in particular 5% to 50% w/w or 7% to 25% w/w (e.g. about 20% w/w, e.g. about 20% w/w of propylene glycol).
The above oil-in-water cream can also optionally comprise:
- one or more solubilisers (e.g. isopropyl myristate), e.g. present at 0.5% to 20% w/w, e.g. 3 to 12% w/w; and/or - one or more buffers (e.g. citric acid and/or dibasic sodium phosphate), e.g. present in total at 0.05 to 5% w/w.
In the above example oil-in-water cream composition, the oil phase can in particular comprise mineral oil (e.g. as emollient and solubiliser) present at 15% to 50% w/w or 20% to 45% w/w, and/or can in particular comprise a high-viscosity wax such as microcrystalline wax (e.g. as emollient) present at 5% to 25% w/w such as 8% to 15% w/w, and/or can in particular comprise a silicone (such as dimethicone e.g. dimethicone 360 or dimethicone 20, e.g. as emollient) present at 0.5% to 20% such as 0.5% to 10% or 1% to 5% w/w.
In the above example oil-in-water cream composition, the one or more surfactants can for example comprise: glycerol monostearate present at 0.5% to 10% w/w, and/or sorbitan monostearate present at 0.05% to 10% w/w, and/or [cetyl alcohol and/or stearyl alcohol] present in total at 0.1% to 15% or 1 to 10% w/w.
Cream emulsions, e.g. water-in-oil or oil-in-water cream emulsions, can generally be prepared by a process in which an aqueous phase is prepared, e.g. prepared before emulsification. The aqueous phase usually contains water and a solubiliser and/or skin- penetration enhancer such as propylene glycol, and optionally contains some or all of the compound of formula (I) or salt thereof, and/or optionally contains surfactant. The oil phase, e.g. containing white petrolatum and/or mineral oil, and/or optionally containing surfactant, can be prepared in a separate vessel. Temperatures of both phases are suitably maintained at (or heated to) elevated temperatures, such as about 45-9O0C or about 45-8O0C or about 45-750C, for example about 55-900C or about 55-8O0C or about 55-750C (in particular at about 60-650C), or e.g. from above 70 to 90 0C, the oil phase temperature being sufficiently high (e.g. about 45-9O0C or about 55-9O0C or from above 70 to 90 0C) to melt the oil phase. While hot, one phase is suitably added to another while mixing, e.g. using a high shear mixer, to effect emulsification, for example keeping the temperature 450C or above, or 550C or above such as above 7O0C e.g. from above 70 to 90 0C. The resulting emulsion is typically allowed to cool, e.g. to about 15-350C such as to about 17-3O0C (e.g. to about 17-220C) or to about 18-3O0C, for example while the agitation continues e.g. at lower speeds. The cream emulsion can then optionally be dispensed from the manufacturing vessel and filled into primary packaging, for example tubes or sachets.
Typically, a pharmaceutical composition of the invention suitable for external topical administration can be administered once daily, twice daily or more than twice daily, to external body part(s), e.g. on the skin such as at a site of diseased skin, e.g. skin suffering from atopic dermatitis.
Pharmaceutical compositions suitable for oral or parenteral administration
A pharmaceutical composition suitable for oral administration can be liquid or solid; for example it can be a syrup, suspension or emulsion, a tablet, a capsule or a lozenge.
A liquid formulation (e.g. oral) can generally consist of a suspension or solution of the compound or pharmaceutically acceptable salt in a suitable pharmaceutically acceptable liquid carriers), for example an aqueous solvent such as water, ethanol or glycerine, or a non-aqueous solvent, such as polyethylene glycol or an oil. The formulation may also contain a suspending agent, preservative, flavouring and/or colouring agent.
In one embodiment, the pharmaceutical composition is in unit dose form, such as a tablet or capsule for oral administration, e.g. for oral administration to a human.
A pharmaceutical composition suitable for oral administration being a tablet can comprise one or more pharmaceutically acceptable carriers and/or excipients suitable for preparing tablet formulations. The carrier can for example be or include lactose, cellulose (for example microcrystalline cellulose), or mannitol. The tablet can also or instead contain one or more pharmaceutically acceptable excipients, for example a binding agent such as hydroxypropylmethylcellulose or povidone (polyvinylpyrrolidone), a lubricant e.g. an alkaline earth metal stearate such as magnesium stearate, and/or a tablet disintegrant such as sodium starch glycollate, croscarmellose sodium, or crospovidone (cross-linked polyvinylpyrrolidone). The pharmaceutical composition being a tablet can be prepared by a method comprising the steps of: (i) mixing the compound of formula (I), as herein defined, or a pharmaceutically acceptable salt thereof, with the one or more pharmaceutically acceptable carriers and/or excipients, (ii) compressing the resulting mixture (which is usually in powder form) into tablets, and (iii) optionally coating the tablet with a tablet film-coating material.
A pharmaceutical composition suitable for oral administration being a capsule can be prepared using encapsulation procedures. For example, pellets or powder containing the active ingredient can be prepared using a suitable pharmaceutically acceptable carrier and then filled into a hard gelatin capsule. Alternatively, a dispersion or suspension can be prepared using any suitable pharmaceutically acceptable carrier, for example an aqueous gum or an oil and the dispersion or suspension then filled into a soft gelatin capsule.
In a pharmaceutical composition for oral administration of the invention, the compound of formula (I) or the pharmaceutically acceptable salt thereof can in one embodiment comprise (e.g. be) the compound of the invention (i.e. as the "free base" form).
A pharmaceutical composition suitable for (e.g. adapted for) parenteral (e.g. intravenous, subcutaneous, or intramuscular) administration can comprise a solution or suspension of the compound or pharmaceutically acceptable salt in a sterile pharmaceutically acceptable aqueous carrier (e.g. sterile water) or parenterally acceptable oil. Alternatively, the solution can be lyophilised. A lyophilised pharmaceutical composition suitable for (e.g. adapted for) parenteral administration may, in use, optionally be reconstituted with a suitable solvent, e.g. sterile water or a sterile parenterally acceptable aqueous solution, just prior to administration.
Pharmaceutical compositions suitable for inhalable or intranasal administration, and particle-size reduction
Compositions suitable for (e.g. adapted for) nasal or inhaled administration may conveniently be formulated as aerosols, drops, gels or dry powders.
Aerosol formulations, e.g. for inhaled administration, can comprise a solution or fine suspension of the active substance in a pharmaceutically acceptable aqueous or nonaqueous solvent. Aerosol formulations can be presented in single or multidose quantities in sterile form in a sealed container, which can take the form of a cartridge or refill for use with an atomising device or inhaler. Alternatively the sealed container may be a unitary dispensing device such as a single dose nasal inhaler or an aerosol dispenser fitted with a metering valve (metered dose inhaler) which is intended for disposal once the contents of the container have been exhausted.
Where the dosage form comprises an aerosol dispenser, it can contain a suitable propellant under pressure such as compressed air, carbon dioxide, or an organic propellant such as a chlorofluorocarbon (CFC) or hydrofluorocarbon (HFC). Suitable CFC propellants include dichlorodifluoromethane, trichlorofluoromethane and dichlorotetrafluoroethane. Suitable HFC propellants include 1 ,1 ,1 ,2,3,3,3- heptafluoropropane and 1 ,1 ,1 ,2-tetrafluoroethane. The aerosol dosage forms can also take the form of a pump-atomiser.
Particle size reduction of compound of formula (I) or salt thereof
In, for example, pharmaceutical compositions suitable (e.g. adapted for) inhaled administration, the compound or salt of formula (I) can be in a particle-size-reduced form. The size-reduced form can for example be obtained or obtainable by micronisation. Micronisation usually involves subjecting the compound/salt to collisional and/or abrasional forces in a fast-flowing circular or spiral/vortex-shaped airstream often including a cyclone component. The particle size of the size-reduced (e.g. micronised) compound or salt can be defined by a D50 value of about 0.5 to about 10 microns, e.g. about 1 to about 7 microns or about 1 to about 5 microns (e.g. as measured using laser diffraction). For example, the compound or salt of formula (I) can have a particle size defined by: a D10 of about 0.3 to about 3 microns (e.g. about 0.5 to about 2 microns, or about 1 micron), and/or a D50 of about 0.5 to about 10 microns or about 1 to about 7 microns or (e.g. about 1 to about 5 microns or about 2 to about 5 microns or about 2 to about 4 microns), and/or a D90 of about 1 to about 30 microns or about 2 to about 20 microns or about 2 to about 15 microns or about 3 to about 15 microns (e.g. about 5 to about 15 microns or about 5 to about 10 microns or about 2 to about 10 microns); for example as measured using laser diffraction.
In particle size measurements, D90, D50 and D10 respectively mean that 90%, 50% and 10% of the material is less than the micron size specified. D50 is the median particle size. DV90, DV50 and DV10 respectively mean that 90%, 50% and 10% by volume of the material is less than the micron size specified. DM90, DM50 and DM10 respectively mean that 90%, 50% and 10% by weight of the material is less than the micron size specified.
Laser diffraction measurement of particle size can use a dry method (wherein a suspension of the compound/salt in an airflow crosses the laser beam) or a wet method [wherein a suspension of the compound/salt in a liquid dispersing medium, such as isooctane or (e.g. if compound is soluble in isooctane) 0.1 % Tween 80 in water, crosses the laser beam]. With laser diffraction, particle size can for example be calculated using the Fraunhofer calculation; and/or optionally a Malvern Mastersizer or Sympatec apparatus is used for measurement. For example, particle size measurement and/or analysis by laser diffraction can use any or all of (e.g. all of) the following: a Malvern Mastersizer longbed version, a dispersing medium of 0.1 % Tween 80 in water, a stir rate of ca. 1500 rpm, ca. 3 mins sonification prior to final dispersion and analysis, a 300 RF (Reverse Fourier) lens, and/or the Fraunhofer calculation with Malvern software.
An illustrative non-limiting example of a small-scale micronisation process is now given:
Micronisation Example: Micronisation of a compound or salt of one of the Examples
• Purpose: To micronise a compound or salt of one of the Examples (described hereinafter), e.g. in an amount of approximately 600-1000 mg thereof, using a Jetpharma MC1 micronizer.
• The parent (unmicronised) and micronised materials are analyzed for particle size by laser diffraction and crystallinity by PXRD.
Micronisation Example: Equipment and material
Equipment/material Description and specification
Jetpharma MC1 Micronizer Nitrogen supply: Air tank with 275psi rate tubing
Analytical balance Sartorius Analytical
Top loader balance Mettler PM400
Digital Caliper VWR Electronic caliper
Materials to be micronised a compound or salt of one of the Examples
(Procedure 1)
Materials to be micronised a compound or salt of one of the Examples
(Procedure2)
The Jetpharma MC1 Micronizer comprises a horizontal disc-shaped milling housing having: a tubular compound inlet (e.g. angled at ca. 30 degrees to the horizontal) for entry of a suspension of unmicronised compound of formula (I) or salt in a gasflow, a separate gas inlet for entry of gases, a gas outlet for exit of gases, and a collection vessel (micronizer container) for collecting micronised material. The milling housing has two chambers: (a) an outer annular chamber in gaseous connection with the gas inlet, the chamber being for receiving pressurised gas (e.g. air or nitrogen), and (b) a discshaped inner milling chamber within and coaxial with the outer chamber for micronising the input compound / salt, the two chambers being separated by an annular wall. The annular wall (ring R) has a plurality of narrow-bored holes connecting the inner and outer chambers and circumferentially-spaced-apart around the annular wall. The holes opening into the inner chamber are directed at an angle (directed part-way between radially and tangentially), and in use act as nozzles directing pressurised gas at high velocity from the outer chamber into the inner chamber and in an inwardly-spiral path (vortex) around the inner chamber (cyclone). The compound inlet is in gaseous communication with the inner chamber via a nozzle directed tangentially to the inner chamber, within and near to the annular wall / ring R. Upper and lower broad-diameter exit vents in the central axis of the inner milling chamber connect to (a) (lower exit) the collection vessel which has no air outlet, and (b) (upper exit) the gas outlet. Inside and coaxial with the tubular compound inlet and longitudinally-movable within it is positioned a venturi inlet (V) for entry of gases. The compound inlet also has a bifurcation connecting to an upwardly-directed material inlet port for inputting material. In use, the narrow head of the venturi inlet (V) is suitably positioned below and slightly forward of the material inlet port, so that when the venturi delivers pressurised gas (e.g. air or nitrogen) the feed material is sucked from the material inlet port into the gas stream through the compound inlet and is accelerated into the inner milling chamber tangentially at a subsonic speed. Inside the milling chamber the material is further accelerated to a supersonic speed by the hole/nozzle system around the ring (R ) (annular wall) of the milling chamber. The nozzles are slightly angled so that the acceleration pattern of the material is in the form of an inwardly-directed vortex or cyclone. The material inside the milling chamber circulates rapidly and particle collisions occur during the process, causing larger particles to fracture into smaller ones. "Centrifugal" acceleration in the vortex causes the larger particles to remain at the periphery of the inner chamber while progressively smaller particles move closer to the centre until they exit the milling chamber, generally through the lower exit, at low pressure and low velocity. The particles that exit the milling chamber are heavier than air and settle downward thorugh the lower exit into the collection vessel (micronizer container), while the exhaust gas rises (together with a minority of small particles of micronised material) and escapes into the atmosphere at low pressure and low velocity.
Micronisation Example: Procedure:
The micronizer is assembled. The narrow head of the venturi inlet is positioned below and slightly forward of the material inlet port and is measured with a micro-caliper to make sure that it is inserted correctly. The ring (R ) and venturi (V) pressures are adjusted according to the values specified in the experimental design (refer to experimental section below) by adjusting the valves on the pressure gauges on the micronizer. The setup is checked for leakage by observing if there is any fluctuation in the reading of the pressure gauges.
Note that the venturi (V) pressure is kept at least 2 bars greater than the ring (R ) pressure to prevent regurgitation of material, e.g. outwardly from the material inlet port.
Balance performance is checked with calibration weights. Specified amount of the parent material is fed into the input container of the micronizer using a spatula. The input container plus material is weighed. The equipment pressure is monitored during the micronization process.
Upon completion of the micronising run, the nitrogen supply is shut off and the micronised material is allowed to settle into the micronizer container. The micronised powder in the micronizer container (collection vessel) and the cyclone (above the recovery vessel) are collected together into a pre-weighed and labelled collection vial. The weight of the micronised material is recorded. The input container is re-weighed in order to calculate the amount of input material by difference. The micronizer is disassembled and residual PDE4 compound on the micronizer inner surface is rinsed with 70/30 isopropyl alcohol / water and collected into a flask. The micronizer is then thoroughly cleaned in a Lancer washing machine and dried before subsequent runs are performed.
Micronisation Example: Optional Experimental Parameters
Procedure 1: Optional Experimental Parameters
This experiment, Procedure 1 , can optionally be carried out generally using a procedure and an apparatus generally as described above or similar to those described, using generally the following experimental parameters:
Material Venturi Proc- input Pressure (V) / edure amount ring (R ) no. (g) Pressure
(bar) 1 ca. 0.9 g V = 5 to 7 bar
R = 3 to 4 bar
% yield = [(Material from collection vessel + Material from cyclone) / Material input amount] x100.
The above optional parameters can be varied using the skilled person's knowledge.
Procedure 2: Optional Experimental Parameters The optional experimental parameters can for example be as follows: Material Venturi Intended
Procinput Pressure (V) / feed-rate edure amount (g) ring (R ) no. Pressure (bar)
2 ca. 0.9 g V = 8 to 10 bar 180 to 200
R = 5.5 to 6 mg/min bar
The above optional parameters can be varied using the skilled person's knowledge Dry powder inhalable compositions
For pharmaceutical compositions suitable (e.g. adapted for) inhaled administration, the pharmaceutical composition may for example be a dry powder inhalable composition. Such a composition can comprise a powder base such as lactose or starch, the compound of formula (I) or salt thereof (suitably in particle-size-reduced form, e.g. in micronised form), and optionally a ternary agent such as L-leucine, mannitol, trehalose, magnesium stearate and/or cellobiose octaacetate (e.g. alpha-D-isomer of cellobiose octaacetate, e.g. available from Aldrich). For cellobiose octaacetate and storage stability, see WO 03/088943.
The dry powder inhalable composition can comprise a dry powder blend of lactose and the compound of formula (I) or salt thereof. The lactose can be lactose hydrate e.g. lactose monohydrate and/or can be inhalation-grade and/or fine-grade lactose. The particle size of the lactose can for example be defined by 90% or more (by weight or by volume) of the lactose particles being less than 1000 microns (micrometres) (e.g. 10-1000 microns e.g. 30-1000 microns) in diameter, and/or 50% or more of the lactose particles being less than 500 microns (e.g. 10-500 microns) in diameter. The particle size of the lactose can for example be defined by 90% or more of the lactose particles being less than 300 microns (e.g. 10-300 microns e.g. 50-300 microns) in diameter, and/or
50% or more of the lactose particles being less than 100 microns in diameter. Optionally, the particle size of the lactose can be defined by 90% or more of the lactose particles being less than 100-200 microns in diameter, and/or 50% or more of the lactose particles being less than 40-70 microns in diameter. It can be about 3 to about 30% (e.g. about 10%) (by weight or by volume) of the particles are less than 50 microns or less than 20 microns in diameter. For example, without limitation, a suitable inhalation-grade lactose is E9334 lactose (10% fines) (Borculo Domo Ingredients, Hanzeplein 25, 8017 JD Zwolle, Netherlands).
In the dry powder inhalable composition the compound of formula (I) or salt thereof can for example be present in about 0.1% to about 70% (e.g. about 1% to about 50%, e.g. about 5% to about 40%, e.g. about 20 to about 30%) by weight of the composition.
An illustrative non-limiting example of a dry powder inhalable composition follows:
Dry Powder Formulation Example - Dry powder Lactose Blend Preparation
Using a size-reduced e.g. micronised form of the compound of formula (I) or salt thereof (e.g. as prepared in the Micronisation Example herein), the dry powder blend is, for example, prepared by mixing the required amount of the compound/salt (e.g. 10 mg, 1% w/w) with inhalation-grade lactose containing 10% fines (e.g. 990 mg, 99% w/w) in a Teflon™ (polytetrafluoroethene) pot in a Mikro-dismembrator ball-mill (but without a ball bearing) at 3A speed (ca. 2000-2500 rpm) for about 4 hours at each blend concentration. The Mikro-dismembrator (available from B. Braun Biotech International, Schwarzenberger Weg 73-79, D-34212 Melsungen, Germany; www.bbraunbiotech.com) comprises a base with an upwardly-projecting and sidewardly-vibratable arm to which is attached the Teflon ™ pot. The vibration of the arm achieves blending.
Other blends can include: 10% w/w compound/salt (50 mg) + 90% w/w lactose (450 mg, inhalation-grade lactose containing 10% fines).
Serial dilution of the 1% w/w blend can achieve e.g. 0.1% and 0.3% w/w blends.
Dry powder inhalation devices
Optionally, in particular for dry powder inhalable compositions, a pharmaceutical composition for inhaled administration can be incorporated into a plurality of sealed dose containers (e.g. containing the dry powder composition) mounted longitudinally in a strip or ribbon inside a suitable inhalation device. The container can be rupturable or peel- openable on demand and the dose, e.g. of the dry powder composition, can be administered by inhalation via a device such as the DISKUS ™ device, marketed by GlaxoSmithKline. The DISKUS TM inhalation device can e.g. be substantially as described in GB 2,242,134 A. In such device at least one container for the pharmaceutical composition in powder form (the at least one container in particular being a plurality of sealed dose containers mounted longitudinally in a strip or ribbon) is defined between two members peelably secured to one another; the device comprises: means defining an opening station for the said at least one container; means for peeling the members apart at the opening station to open the container; and an outlet, communicating with the opened container, through which a user can inhale the pharmaceutical composition in powder form from the opened container.
Dosage regimens
In a pharmaceutical composition suitable for (e.g. adapted for) external topical administration, e.g. an ointment or an oil-in-water or water-in-oil composition, the compound of formula (I) or the pharmaceutically acceptable salt thereof can be present in 0.1% to 10%, such as 0.2% to 10% or 0.2% to 5%, or 0.5% to 10% or 0.5% to 5%, or 1 % to 10% or 1 % to 5%, or 0.5% to 3%, or 1 % to 3% (e.g. about 0.5% or in particular about 2%), by weight of the composition (w/w). Typically, an external-topical pharmaceutical composition can be administered once daily, twice daily or more than twice daily, to external body part(s), e.g. to the skin such as at a site of diseased skin. The amount administered is usually such as substantially to cover the site(s) of diseased skin.
A pharmaceutical composition of the invention can e.g. be in unit dose form such as a tablet or capsule for oral administration, e.g. for oral administration to a human. In the pharmaceutical composition of the invention, a or each dosage unit for oral or parenteral administration can for example contain from 0.01 to 3000 mg, such as 0.5 to 1000 mg, of a compound of the formula (I) or a pharmaceutically acceptable salt thereof, calculated as the free base. A or each dosage unit for nasal or inhaled administration can for example contain from 0.001 to 50 mg, such as 0.01 to 5 mg, of a compound of the formula (I) or a pharmaceutically acceptable salt thereof, calculated as the free base.
When a parenteral or oral composition is used, a pharmaceutically acceptable compound or salt of the invention can for example be administered to a mammal (e.g. human) in a daily oral or parenteral dose of 0.001 mg to 50 mg per kg body weight per day (mg/kg/day), for example 0.01 to 20 mg/kg/day or 0.03 to 10 mg/kg/day or 0.1 to 2 mg/kg/day, of the compound of the formula (I) or a pharmaceutically acceptable salt thereof, calculated as the free base.
When a parenteral or oral composition is used, a pharmaceutically acceptable compound or salt of the invention can for example be administered to a mammal (e.g. human) in a daily nasal or inhaled dose of: 0.0001 to 5 mg/kg/day or 0.0001 to 1 mg/kg/day, e.g. 0.001 to 1 mg/kg/day or 0.001 to 0.3 mg/kg/day or 0.001 to 0.1 mg/kg/day or 0.005 to 0.3 mg/kg/day, of the compound of the formula (I) or a pharmaceutically acceptable salt thereof, calculated as the free base.
The pharmaceutically acceptable compounds or salts of the invention can for example be administered to a human in a daily dose (for an adult patient) of, for example, an oral or parenteral dose of 0.01 mg to 3000 mg per day or 0.5 to 1000 mg per day e.g. 2 to 500 mg per day, or a nasal or inhaled dose of 0.001 to 50 mg per day or 0.01 to 30 mg per day or 0.01 to 5 mg per day or 0.02 to 2 mg per day, of the compound of the formula (I) or a pharmaceutically acceptable salt thereof, calculated as the free base.
COMBINATIONS
The compounds, salts and/or pharmaceutical compositions according to the invention may also be used in combination with another therapeutically active agent, for example, a β2 adrenoreceptor agonist, an anticholinergic compound (e.g. muscarinic (M) receptor antagonist), an anti-histamine, an anti-allergic, an anti-inflammatory agent, an antiinfective agent or an immunosuppressant.
The invention thus provides, in a further aspect, a combination comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof together with another therapeutically active agent, for example, a β2-adrenoreceptor agonist, an anti-histamine, an anti-allergic, an anti-inflammatory agent, an antiinfective agent or an immunosuppressant. In one particular embodiment, the β2-adrenoreceptor agonist is salmeterol (e.g. as racemate or a single enantiomer such as the R-enantiomer), salbutamol, formoterol, salmefamol, fenoterol or terbutaline, or a salt thereof (e.g. pharmaceutically acceptable salt thereof), for example the xinafoate salt of salmeterol, the sulphate salt or free base of salbutamol or the fumarate salt of formoterol. Long-acting β2-adrenoreceptor agonists are optionally used, especially those having a therapeutic effect over a 12-24 hour period such as salmeterol or formoterol. In particular, the β2-adrenoreceptor agonist is for inhaled administration, e.g. once per day and/or for simultaneous inhaled administration; and in particular the β2-adrenoreceptor agonist can be in particle-size-reduced form e.g. as defined herein. The β2-adrenoreceptor agonist combination can for example be for treatment and/or prophylaxis of COPD or asthma. Salmeterol or a pharmaceutically acceptable salt thereof, e.g. salmeterol xinofoate, is suitably administered to humans at an inhaled dose of 25 to 50 micrograms twice per day (measured as the free base).
Examples of long acting β2-adrenoreceptor agonists which could be used include those described in WO 02/066422A, WO 03/024439, WO 02/070490 and WO 02/076933.
Particular examples of β2-adrenoreceptor agonists disclosed in WO 02/066422 include: 3-(4-{[6-({(2R)-2-hydroxy-2-[4-hydroxy-3-(hydroxymethyl)- phenyl]ethyl}amino)hexyl]oxy}butyl)benzenesulfonamide and 3-(3-{[7-({(2R)-2-hydroxy-2-[4-hydroxy-3-hydroxymethyl)phenyl]ethyl}- amino)heptyl]oxy}propyl)benzenesulfonamide.
A particular example of a β2-adrenoreceptor agonist disclosed in WO 03/024439 is:
4-{(1R)-2-[(6-{2-[(2,6-dichlorobenzyl)oxy]ethoxy}hexyl)amino]-1-hydroxyethyl}-2- (hydroxymethyl)phenol.
An anti-histamine usable in a combination of a compound of formula (I) or salt can for example be for oral administration (e.g. as a combined composition such as a combined tablet), and can be for treatment and/or prophylaxis of allergic rhinitis. Examples of antihistamines include methapyrilene, or H1 antagonists such as cetirizine, loratadine (e.g. Clarityn TM)1 desloratadine (e.g. Clarinex TM) or fexofenadine (e.g. Allegra TM)
The invention also provides, in a further aspect, a combination comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof together with an anticholinergic compound, e.g. a muscarinic (M) receptor antagonist such as an M-] , M2,
M-|/M2, or M3 receptor antagonist, in particular a M3 receptor antagonist, such as a M3 receptor antagonist which selectively antagonises (e.g. antagonises 10 times or more strongly) the M3 receptor over the Mi and/or M2 receptor. For combinations of anticholinergic compounds / muscarinic (M) receptor antagonist with PDE4 inhibitors, see for example WO 03/011274 A2 and WO 02/069945 A2 / US 2002/0193393 A1 and US 2002/052312 A1 , and some or all of these publications give examples of anticholinergic compounds / muscarinic (M) receptor antagonists which may be used with the compounds of formula (I) or salts, and/or suitable pharmaceutical compositions. For example, the muscarinic receptor antagonist can comprise or be an ipratropium salt (e.g. ipratropium bromide), an oxitropium salt (e.g. oxitropium bromide), or a tiotropium salt (e.g. tiotropium bromide); see e.g. EP 418 716 A1 for tiotropium.
The anticholinergic compound or muscarinic (M) receptor antagonist, e.g. M3 receptor antagonist, can for example be for inhaled administration, in particular in particle-size- reduced form e.g. as defined herein. In one optional embodiment, both the muscarinic (M) receptor antagonist and the compound of formula (I) or the pharmaceutically acceptable salt thereof are for inhaled administration. For example, the anticholinergic compound or muscarinic receptor antagonist and the compound of formula (I) or salt are optionally for simultaneous administration. The muscarinic receptor antagonist combination is optionally for treatment and/or prophylaxis of COPD.
Other possible combinations include, for example, a combination comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof together with another anti-inflammatory agent such as an anti-inflammatory corticosteroid; or a non- steroidal anti-inflammatory drug (NSAID) such as a leukotriene antagonist (e.g. montelukast), an iNOS inhibitor, a tryptase inhibitor, a elastase inhibitor, a beta-2 integrin antagonist, a adenosine 2a agonist, a CCR3 antagonist, or a 5-lipoxogenase inhibitor; or an antiinfective agent (e.g. an antibiotic or an antiviral). An iNOS inhibitor can e.g. be for oral administration. Examples of iNOS inhibitors (inducible nitric oxide synthase inhibitors) include those disclosed in WO 93/13055, WO 98/30537, WO 02/50021 , WO 95/34534 and WO 99/62875.
Example combinations, in particular for external topical administration (e.g. versus atopic dermatitis), include, for example, a combination comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof together with an immunosuppressant, e.g. a calcineurin inhibitor such as pimecrolimus or tacrolimus. The immunosuppressant can in particular be an externally-topically administrable immunosuppressant such as pimecrolimus (e.g. pimecrolimus at ca. 1% w/w concentration in a topical composition such as a cream, and/or e.g. Elidel ™) or tacrolimus (e.g. tacrolimus at from about 0.03% to about 0.1% w/w concentration in a topical composition such as an ointment, and/or e.g. Protopic ™). The externally-topically administrable immunosuppressant can be administered or administrable in a external-topical composition separately from the compound or salt of the invention, or it can be contained with the compound of formula (I) or pharmaceutically acceptable salt in a combined externally-topically-administrable composition. For external topical administration, e.g. versus an inflammatory and/or allergic skin disease such as atopic dermatitis or psoriasis, in a combination of the compound or salt of the invention together with an anti-infective agent, the anti-infective agent can include (e.g. be) an externally-topically-administrable antibacterial, such as mupiricin or a salt thereof (e.g. mupiricin calcium salt) (e.g. Bactroban ™) or such as an externally-topically- administrable pleuromutilin antibacterial (e.g. retapamulin or a salt thereof, which can be present in about 1% w/w by weight of an externally-topically-administrable pharmaceutical composition, such as an ointment). Alternatively or additionally, for external topical administration, the anti-infective agent can include an externally- topically-administrable antifungal such as clortrimazole, clotrimazole or ketoconazole.
For external topical administration, e.g. versus atopic dermatitis, a combination with an anti-itch compound may optionally be used.
In a combination comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof together with an anti-inflammatory corticosteroid (which can for example be for treatment and/or prophylaxis of asthma, COPD or allergic rhinitis), then optionally the anti-inflammatory corticosteroid is fluticasone propionate (e.g. see US patent 4,335,121), beclomethasone 17-propionate ester, beclomethasone 17,21 -dipropionate ester, dexamethasone or an ester thereof, mometasone or an ester thereof (e.g. mometasone furoate), ciclesonide, budesonide, flunisolide, or a compound as described in WO 02/12266 A1 (e.g. as claimed in any of claims 1 to 22 therein), or a pharmaceutically acceptable salt of any of the above. If the anti-inflammatory corticosteroid is a compound as described in WO 02/12266 A1, then it can for example be Example 1 therein {which is 6α,9α-difluoro-17α-[(2-furanylcarbonyl)oxy]-11 β-hydroxy-16α-methyl-3- oxo-androsta-1 ,4-diene-17β-carbothioic acid S-fluoromethyl ester} or Example 41 therein {which is 6α,9α-difluoro-11β-hydroxy-16α-methyl-17α-[(4-methyl-1 ,3-thiazole-5- carbonyl)oxy]-3-oxo-androsta-1 ,4-diene-17β-carbothioic acid S-fluoromethyl ester}, or a pharmaceutically acceptable salt thereof. The anti-inflammatory corticosteroid can for example be for external topical, intranasal or inhaled administration. Fluticasone propionate can be used for inhaled administration to a human, for example either (a) at a dose of 250 micrograms once per day or (b) at a dose of 50 to 250 micrograms twice per day.
Also provided is a combination comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof together with β2-adrenoreceptor agonist and an anti-inflammatory corticosteroid, for example as described in WO 03/030939 A1. This combination can for example be for treatment and/or prophylaxis of asthma, COPD or allergic rhinitis. The β2-adrenoreceptor agonist and/or the anti-inflammatory corticosteroid can be as described above and/or as described in WO 03/030939 A1. In this "triple" combination, the β2-adrenoreceptor agonist can e.g. be salmeterol or a pharmaceutically acceptable salt thereof (e.g. salmeterol xinafoate) and the antiinflammatory corticosteroid can e.g. be fluticasone propionate.
The combinations referred to above may be presented for use in the form of a pharmaceutical composition and thus a pharmaceutical composition comprising a combination as defined above together with one or more pharmaceutically acceptable carriers and/or excipients represent a further aspect of the invention.
The individual compounds of such combinations may be administered either sequentially or simultaneously in separate or combined pharmaceutical composition.
In one embodiment, the combination as defined herein can be for simultaneous inhaled administration and is disposed in a combination inhalation device. Such a combination inhalation device is another aspect of the invention. Such a combination inhalation device can comprise a combined pharmaceutical composition for simultaneous inhaled administration (e.g. dry powder composition), the composition comprising all the individual compounds of the combination, and the composition being incorporated into a plurality of sealed dose containers mounted longitudinally in a strip or ribbon inside the inhalation device, the containers being rupturable or peel-openable on demand; for example such inhalation device can be substantially as described in GB 2,242,134 A (DISKUS TM) and/or as described above. Alternatively, the combination inhalation device can be such that the individual compounds of the combination are administrable simultaneously but are stored separately (or wholly or partly stored separately for triple combinations), e.g. in separate pharmaceutical compositions, for example as described in PCT/EP03/00598 filed on 22 January 2003, published as WO 03/061743 (e.g. as described in the claims thereof e.g. claim 1).
The invention also provides a method of preparing a combination as defined herein, the method comprising either
(a) preparing a separate pharmaceutical composition for administration of the individual compounds of the combination either sequentially or simultaneously, or
(b) preparing a combined pharmaceutical composition for administration of the individual compounds of the combination simultaneously, wherein the pharmaceutical composition comprises the combination together with one or more pharmaceutically acceptable carriers and/or excipients.
The invention also provides a combination as defined herein, prepared by a method as defined herein. BIOLOGICAL TEST METHODS
PDE 3, PDE 4B, PDE 4D, PDE 5, PDE 6 in vitro assay methods
The biological activity of the compounds or salts of the invention can be measured in the assay methods shown below, or in generally similar or generally analogous assay methods.
Possible PDE enzyme sources and literature references
Human recombinant PDE4B, in particular the 2B splice variant thereof (HSPDE4B2B), is disclosed in WO 94/20079 and also M. M. McLaughlin et al., "A low Km, rolipram- sensitive, cAMP-specific phosphodiesterase from human brain: cloning and expression of cDNA, biochemical characterisation of recombinant protein, and tissue distribution of mRNA", J. Biol. Chem., 1993, 268, 6470-6476. For example, in Example 1 of WO 94/20079, human recombinant PDE4B is described as being expressed in the PDE- deficient yeast Saccharomyces cerevisiae strain GL62, e.g. after induction by addition of 150 uM CUSO4, and 100,000 x g supernatant fractions of yeast cell lysates are described for use in the harvesting of PDE4B enzyme.
Human recombinant PDE4D (HSPDE4D3A) is disclosed in P. A. Baecker et al., "Isolation of a cDNA encoding a human rolipram-sensitive cyclic AMP phoshodiesterase (PDE IVD)", Gene, 1994, 138, 253-256.
Human recombinant PDE5 is disclosed in K. Loughney et al., "Isolation and characterisation of cDNAs encoding PDE5A, a human cGMP-binding, cGMP-specific 3',5'-cyclic nucleotide phosphodiesterase", Gene, 1998, 216, 139-147.
PDE3 can be purified from bovine aorta as described by H. Coste and P. Grondin, "Characterisation of a novel potent and specific inhibitor of type V phosphodiesterase", Biochem. Pharmacol., 1995, 50, 1577-1585.
PDE6 can be purified from bovine retina as described by: P. Catty and P. Deterre, "Activation and solubilization of the retinal cGMP-specific phosphodiesterase by limited proteolysis", Eur. J. Biochem., 1991, 199, 263-269; A. Tar et al. "Purification of bovine retinal cGMP phosphodiesterase", Methods in Enzymology, 1994, 238, 3-12; and/or D. Srivastava et al. "Effects of magnesium on cyclic GMP hydrolysis by the bovine retinal rod cyclic GMP phosphodiesterase", Biochem. J., 1995, 308, 653-658. Inhibition of PDE 3, PDE 4B, PDE 4D, PDE 5 or PDE 6 activity: radioactive Scintiliation Proximity Assay (SPA)
The ability of compounds to inhibit catalytic activity at PDE4B or 4D (human recombinant), PDE3 (from bovine aorta), PDE5 (human recombinant) or PDE6 (from bovine retina) can optionally be determined by Scintillation Proximity Assay (SPA) in a 96-well format.
Test compounds (as a solution in DMSO, suitably about 2 microlitre (ul) volume of DMSO solution) are preincubated at ambient temperature (room temperature, e.g. 19- 230C) in Wallac lsoplates (code 1450-514) with PDE enzyme in 5OmM Tris-HCI buffer pH 7.5 , 8.3mM MgCI2, 1.7mM EGTA, 0.05% (w/v) bovine serum albumin for 10-30 minutes (usually 30 minutes). The enzyme concentration is adjusted so that no more than 20% hydrolysis of the substrate defined below occurs in control wells without compound, during the incubation. For the PDE3, PDE4B and PDE4D assays, [5',8- 3H]Adenosine 3',5'-cyclic phosphate (Amersham Pharmacia Biotech, code TRK.559; or Amersham Biosciences UK Ltd, Pollards Wood, Chalfont St Giles, Buckinghamshire HP8 4SP, UK) is added to give 0.05uCi per well and about 1OnM final concentration. For the PDE5 and PDE6 assays, [8-3H]Guanosine 3',5'-cyclic phosphate (Amersham Pharmacia Biotech, code TRK.392) is added to give O.OδuCi per well and about 36nM final concentration. Plates containing assay mixture, suitably approx. 100 ul volume of assay mixture, are mixed on an orbital shaker for 5 minutes and incubated at ambient temperature for 1 hour. Phosphodiesterase SPA beads (Amersham Pharmacia Biotech, code RPNQ 0150) are added (about 1mg per well) to terminate the assay. Plates are sealed and shaken and allowed to stand at ambient temperature for 35 minutes to 1 hour (suitably 35 minutes) to allow the beads to settle. Bound radioactive product is measured using a WALLAC TRILUX 1450 Microbeta scintillation counter. For inhibition curves, 10 concentrations (e.g. 1.5nM - 3OuM) of each compound are assayed. Curves are analysed using ActivityBase and XLfit (ID Business Solutions Limited, 2 Ocean Court, Surrey Research Park, Guildford, Surrey GU2 7QB, United Kingdom). Results are expressed as plC5o values.
In an alternative to the above radioactive SPA assay, PDE4B or PDE4D inhibition can be measured in the following Fluorescence Polarisation (FP) assay:
Inhibition ofPDE4B orPDE4D activity: Fluorescence Polarisation (FP) assay
The ability of compounds to inhibit catalytic activity at PDE4B (human recombinant) or PDE4D (human recombinant) can optionally be determined by IMAP Fluorescence Polarisation (FP) assay (IMAP Explorer kit, available from Molecular Devices Corporation, Sunnydale, CA, USA; Molecular Devices code: R8062) in 384-well format.
The IMAP FP assay is able to measure PDE activity in an homogenous, nonradioactive assay format. The FP assay uses the ability of immobilised trivalent metal cations, coated onto nanoparticles (tiny beads), to bind the phosphate group of FI-AMP that is produced on the hydrolysis of fluorescein-labelled (Fl) cyclic adenosine mono- phosphate (FI-cAMP) to the non-cyclic FI-AMP form. FI-cAMP substantially does not bind. Binding of FI-AMP product to the beads (coated with the immobilised trivalent cations) slows the rotation of the bound FI-AMP and leads to an increase in the fluorescence polarisation ratio of parallel to perpendicular light. Inhibition of the PDE reduces/inhibits this signal increase.
Test compounds (small volume, e.g. ca. 0.5 to 1 microlitres (ul), suitably ca. 0.5 ul, of solution in DMSO) are preincubated at ambient temperature (room temperature, e.g. 19-230C) in black 384-well microtitre plates (supplier: NUNC, code 262260) with PDE enzyme in 1OmM Tris-HCI buffer pH 7.2, 1OmM MgCI2, 0.1% (w/v) bovine serum albumin, and 0.05% NaN3 for 10-30 minutes. The enzyme level is set by experimentation so that reaction is linear throughout the incubation. Fluorescein adenosine 3',5'-cyclic phosphate (from Molecular Devices Corporation, Molecular Devices code: R7091) is added to give about 4OnM final concentration (final assay volume usually ca. 20-40 ul, suitably ca. 20 ul). Plates are mixed on an orbital shaker for 10 seconds and incubated at ambient temperature for 40 minutes. IMAP binding reagent (as described above, from Molecular Devices Corporation, Molecular Devices code: R7207) is added (6OuI of a 1 in 400 dilution in binding buffer of the kit stock solution) to terminate the assay. Plates are allowed to stand at ambient temperature for 1 hour. The Fluorescence Polarisation (FP) ratio of parallel to perpendicular light is measured using an Analyst™ plate reader (from Molecular Devices Corporation). For inhibition curves, 10 concentrations (e.g. 1.5nM - 3OuM) of each compound are assayed. Curves are analysed using ActivityBase and XLfit (ID Business Solutions Limited, 2 Ocean Court, Surrey Research Park, Guildford, Surrey GU2 7QB, United Kingdom). Results are expressed as plC50 values. In the FP assay, reagents can be dispensed using Multidrop™ (available from
Thermo Labsystems Oy, Ratastie 2, PO Box 100, Vantaa 01620, Finland).
For a given PDE4 inhibitor, the PDE4B (or PDE4D) inhibition values measured using the SPA and FP assays can differ slightly.
Biological Data obtained for the compound of the invention (e.g. as prepared in Example 1 ) (PDE4B and PDE4D inhibitory activity, as an average of 2 or more readings (n = 2 or more, e.g. 3) are generally as follows, based on measurements only, generally using FP assay(s) generally as described above or generally similar or generally analogous to those described above. In each of the SPA and FP assays, absolute accuracy of measurement is not possible, and the readings given are generally thought to be accurate only up to very approximately ± 0.5 of a log unit, depending on the number of readings made and averaged:
Figure imgf000049_0001
The compound of the invention (e.g. as prepared in Example 1) has also been tested for PDE3, PDE5 and PDE6 inhibition, e.g. using (independently for each PDE type) either a Fluorescence Polarisation (FP) general type of assay or using an SPA general type of assay. The compound of the invention (e.g. as prepared in Example 1) exhibits a larger PDE4B plC50 value than its PDE3, PDE5 and PDE6 plC50 values, i.e. it inhibits PDE4B more strongly than it inhibits PDE3, PDE5 and PDE6 (in the particular assays used).
Emesis: Some known PDE4 inhibitors can cause emesis and/or nausea to greater or lesser extents, especially after systemic exposure e.g. after oral administration (e.g. see Z. Huang et al., Current Opinion in Chemical Biology, 2001 , 5: 432-438, see especially pages 433-434 and refs cited therein). Therefore, it would be preferable, but not essential, if a PDE4 inhibitory compound or salt of the invention were to cause only limited or manageable emetic side-effects, e.g. after oral or parenteral or external-topical administration. Emetic side-effects can for example be measured by the emetogenic potential of the compound or salt when administered to ferrets; for example one can measure the time to onset, extent, frequency and/or duration of vomiting, retching and/or writhing in ferrets after oral or parenteral administration of the compound or salt. See for example In vivo Assay 3 hereinafter for one optional measurement method for anti- inflammatory effect, emetic side-effects and therapeutic index (Tl) in the ferret. See also for example A. Robichaud et al., "Emesis induced by inhibitors of [PDE IV] in the ferret", Neuropharmacology, 1999, 38, 289-297, erratum Neuropharmacology, 2001 , 40, 465- 465. However, optionally, emetic side-effects and therapeutic index (Tl) after oral administration in rats can be conveniently measured by monitoring the pica feeding behaviour of rats after administration of the compound or salt of the invention (see In Vivo Assay 2 below).
Other optional in vitro assays:
Inhibition of TNF-α (TNF-alpha) Production in Human PBMC (peripheral blood mononuclear cell) assay (MSD technology)
This is an optional supplementary test.
A 96-well plate (96 MicroWell™ Plates Nunclon™Δ - High Flange Design, Fisher Scientific UK, Bishop Meadow Road, Loughborough LE 11 5 RG, Leicestershire, UK) is prepared by initially adding to column 1 ca. 1OmM of test compound dissolved in DMSO. For a more potent compound, a more diluted solution in DMSO may be used. The compound is further diluted with DMSO into columns 2 to 9 by 8 successive 3-fold dilutions using the Biomek® FX Laboratory Automation Workstation (Beckman Coulter, Inc., 4300 N. Harbor Boulevard, P.O. Box 3100, Fullerton, CA 92834-3100 USA). Column 10 is used as a DMSO negative control (High Signal, 0% response), whilst column 11, which contains 10 mM of the PDE4 inhibitor roflumilast, is used as a positive control (Low Signal, 100% response). About 1 μl (about 1 ul) of compound is transferred to the compound plate using the Biomek® FX.
PBMC cells (peripheral blood mononuclear cells) are prepared from heparinised human blood (using 1% v/v Heparin Sodium 1000IU/ml Endotoxin Free, Leo Laboratories Ltd., Cashel Road, Dublin 12 . Ireland, Cat No: PL0043/0149) from normal volunteers using the Accuspin™ System-Histopaque®-1077 essentially (Sigma-Aldrich Company Ltd., The Old Brickyard New Rd, Gillingham Dorset SP8 4XT). About 20 ml of blood is overlaid onto 15ml Histopaque® in Accuspin™ tubes. The tube is then centrifuged at about 80Og for ca. 20 minutes. The cells are collected from the interface, washed by centrifugation (ca. 130Og, ca. 10 minutes) and resuspended in RPM11640 medium (Low endotoxin RPMH 640 medium, Cat No: 31870-025, Invitrogen Corporation Invitrogen Ltd, 3 Fountain Drive, Inchinnan Business Park, Paisley PA4 9RF, UK) containing 10% foetal calf serum, 1% L-glutamine (Invitrogen Corporation, Cat No: 25030024) and 1% penicillin/streptomycin (Invitrogen Corporation, Cat No: 15140-122). Viable cells are counted by trypan blue staining and diluted to 1x1 O^ viable cells/ml. About 50μl (about
5OuI) of diluted cells and about 75μl (about 75ul) of LPS (ca. 1 ng/ml final; Sigma Cat No: L-6386) are added to the compound plate, which is then incubated at 37 0C, 5% CO2, for 20 hours.
The supernatant is removed and the concentrations of TNF-α are determined by electrochemiluminescence assay using the Meso Scale Discovery (MSD) technology (Meso Scale Discovery, 9238 Gaither Road, Gaithersburg, Maryland 20877, USA). See the "TNF-α (TNF-alpha) MSD Assay" described below for typical details.
Results can be expressed as plC50 values for inhibition of TNF-α (TNF-alpha) production in PBMCs, and it should be appreciated that these results can be subject to variability or error.
Inhibition of TNFa (TNF-alpha) Production in Human PBMC (peripheral blood mononuclear cell) assay (IGEN technology)
This is an optional supplementary test.
Test compounds are prepared as a ca. 1OmM stock solution in DMSO and a dilution series prepared in DMSO with 8 successive 3-fold dilutions, either directly from the 1OmM stock solution or from a more dilute solution in DMSO. The compound is added to assay plates using a Biomek Fx liquid handling robot.
PBMC cells (peripheral blood mononuclear cells) are prepared from heparinised human blood from normal volunteers by centrifugation on histopaque at ca. 1000g for ca. 30 minutes. The cells are collected from the interface, washed by centrifugation (ca. 130Og, ca. 10 minutes) and resuspended in assay buffer (RPMI 1640 containing 10% foetal calf serum, 1% L-glutamine and 1% penicillin / streptomycin) at 1x106 cells/ml. Ca. 50μl (ca. 5OuI) of cells are added to microtitre wells containing ca. 0.5 or ca. 1.Oμl (ul) of an appropriately diluted compound solution. Ca. 75μl (ul) of LPS (lipopolysaccharide) (ca. 1 ng/ml final) is added and the samples are incubated at 37 0C, 5% CO2, for 20 hours.
The supernatant is removed and the concentrations of TNF-α are determined by electrochemiluminescence assay using the IGEN technology or by ELISA (see below).
Results can be expressed as plC50 values for inhibition of TNF-α (TNF-alpha) production in PBMCs, and it should be appreciated that these results can be subject to variability or error.
Inhibition of TNFa (TNF-alpha) Production in Human Whole Blood
This is an optional supplementary test, e.g. for potentially orally-administrable PDE4 inhibitors. Also, as the assay may measure the effect of PDE4 inhibitors after loss by protein binding, it might possibly also be relevant to extemally-topically-administrable PDE4 inhibitors as protein-binding-loss of compound is possible during transport through the skin.
Test compounds are prepared as a ca. 1OmM stock solution in DMSO and a dilution series prepared in DMSO with 8 successive 3-fold dilutions, either directly from the 1OmM stock solution or from a more dilute solution in DMSO. The compound is added to assay plates using a Biomek Fx liquid handling robot.
Heparinised blood drawn from normal volunteers is dispensed (ca. 100μl = ca. 10OuI) into microtitre plate wells containing ca. 0.5 or ca. 1.Oμl (ul, microlitres) of an appropriately diluted test compound solution. After ca. 1 hr incubation at ca. 37 0C, 5% CO2, ca. 25μl (ca. 25ul) of LPS (lipopolysaccharide) solution (S. typhosa) in RPM1 1640
(containing 1% L-glutamine and 1% Penicillin / streptomycin) is added (ca. 50ng/ml final). The samples are incubated at ca. 37°C, 5% CO2, for ca. 20 hours, and ca. 10Oμl
(ca. 10OuI) physiological saline (0.138% NaCI) is added, and diluted plasma is collected using a Platemate or Biomek FX liquid handling robot after centrifugation at ca. 1300 g for ca. 10 min. Plasma TNFα content is determined by electrochemiluminescence assay using the MSD technology (see below), the IGEN technology (see below) or by enzyme linked immunosorbant assay (ELISA) (see below).
Results can be expressed as plC50 values for inhibition of TNF-α (TNF-alpha) production in Human Whole Blood, and it should be appreciated that these results can be subject to variability or error. Human Whole Blood assay results:
For the compound of the invention (e.g. as prepared in Example 1 ), and generally when using the one of the above assays, or a generally similar or generally analogous assay, the mean and/or measured plC50 values for inhibition of TNF-α (TNF-alpha) production in Human Whole Blood are generally as follows (subject to variability or error):
Figure imgf000053_0001
TNF-α (TNF-alpha) MSD Assay Using the Biomek FX, 25μl (25ul) of MSD Human Serum Cytokine Assay Diluent (Meso Scale Discovery, 9238 Gaither Road, Gaithersburg, Maryland 20877) is added to a 96-well High-Bind MSD plate pre-coated with anti-hTNF alpha capture antibody (MA6000) and then incubated for 24 hours at 4°C to prevent non-specific binding. About 20μl (ul) of supernatant from the PBMC plate or about 40μl (ul) of supernatant from the whole blood (WB) plate are then transferred from columns 1-11 to columns 1-11 of the MSD plate using the Biomek FX. About 20 μl (ul) of TNF-α standard (Cat No. 210-TA; R&D Systems Inc., 614 McKinley Place NE, Minneapolis, MN 55413, USA) are added to column 12 of the MSD plate to generate a standard calibration curve (about 0 to 30000 pg/ml final).
For the Whole Blood assay, plates are washed after 2 hours shaking with a Skanwasher 300 version B (Skatron Instruments AS. PO Box 8, N-3401 Lier, Norway). About 40μl (ul) of diluted sulfo-TAG antibody (ca. 1 μg/ml final) is added, the plates are shaken at room temperature for 1 hours, and the plates washed again as above. About 150μl (ul) of Read Buffer T (2X) is added to the plates, which are then read on a MSD Sector 6000.
For the PBMC assay, about 20 μl (ul) of diluted sulfo-TAG antibody (ca. 1 μg/ml final) is added to each well, and the plates / wells are shaken at room temperature for 2 hours. Finally, about 90μl (ul) of MSD Read Buffer P (diluted to 2.5 times with distilled water) is added and the plates are read on a MSD Sector 6000.
Data Analysis
Data analysis is performed with ActivityBase/XC50 module (ID Business Solutions Ltd., 2 Occam Court, Surrey Research Park, Guildford, Surrey, GU2 7QB UK). Data are normalized and expressed as %inhibition using the formula 100*((U-C1)/(C2-C1)) where U is the unknown value, C1 is the average of the high signal (0%) control wells (column 10), and C2 is the average of the low signal (100%) control wells (column 11). Curve fitting is performed with the following equation: y = A+((B-A)/(1+(10Λx/10ΛC)ΛD)), where A is the minimum response, B is the maximum response, C is the log10(IC50), and D is the Hill slope. The results for each compound are recorded as plC50 values (-C in the above equation).
TNF-a (TNF-alpha) IGEN Assay Ca. 50μl supernatant from either whole blood or PBMC assay plates is transferred to a 96 well polypropylene plate. Each plate also contains a TNF-α standard curve (ca. 0 to 30000 pg/ml: R+D Systems, 210-TA). Ca. 50μl (ul) of streptavidin/biotinylated anti- TNF-α antibody mix, ca. 25μl ruthenium tagged anti-TNF-α monoclonal and ca. 100μl PBS containing 0.1% bovine serum albumin are added to each well and the plates are sealed and shaken for ca. 2 hours before being read on an IGEN instrument.
TNF-α (TNF-alpha) ELISA Assay (enzyme linked immunosorhant assay)
Human TNF-α can be assayed using a commercial ELISA assay kit (AMS Biotechnology, 211-90-164-40) according to the manufacturers' instructions but with TNF-α calibration curves prepared using Pharmingen TNF-α (cat No. 555212).
In Vivo Biological Assays
The in vitro enzymatic PDE4B inhibition assay(s) described above or generally similar or generally analogous assays should be regarded as being the primary test(s) of biological activity. However, some additional in vivo biological tests, which are optional only, and which are not an essential measure of activity, efficacy or side-effects, and which have not necessarily been carried out, are described below.
In Vivo Assay A:
Activity of topically-applied compounds in a pig model of atopic dermatitis: Effect of compounds, applied by skin topical administration, on the dinitrofluorobenzene (DNFB)-induced delayed type hypersensitivity (DTH) response in pigs
General Study Design:
The pig DTH (delayed type hypersensitivity) model of contact hypersensitivity utilizes the Th2-mediated inflammatory response in pig skin to mimic the pathology of atopic dermatitis in humans. The model measures the potential anti-inflammatory effect of compounds, topically-applied to the skin, on the acute DTH (delayed type hypersensitivity) response in castrated male Yorkshire pigs.
, In general in the assay, pigs (domestic Yorkshire pigs, 15-18 kg at time of sensitization, castrated males) are sensitized by topical application of ca. 10% (w/v) dinitrofluorobenzene (DNFB) dissolved in DMSO:acetone:olive oil (ca. 1:5:3) (ca. 40 mg DNFB, 400 microlitre solution total) to the ears (outer) and groin (inner) on day -12 and - 5. The pigs are then challenged with ca. 0.6% (w/v) DNFB applied to randomized sites on the shaved back of the pigs (ca. 90 micrograms/site; sites are identified and numbered by grid made with marking pen).
On the day of challenge, the treatments are performed at the challenge sites at about 2 hours prior to and about 6 hours after challenge (for DMSO / acetone solutions/suspensions containing the PDE4 inhibitor, to maximize exposure to drug), or at about 30 minutes after and about 6 hours after challenge (for topical ointments or creams containing the PDE4 inhibitor, representing a more clinically relevant treatment protocol).
After 24 hrs and 48 hrs post challenge, test sites are visually evaluated for intensity and extent of erythema by measuring the diameter of the reaction at its widest point and assigning scores of 0 to 4 for each of erythema intensity and erythema extent. Induration (a measure of swelling) is also scored 0 to 4. Scores for erythema intensity, erythema extent and induration are assigned according to the following criteria: Intensity of Erythema: 0=normal, 1=minimal, barely visible, 2=mild, 3=moderate, 4=severe. Extent of Erythema (not raised): 0=no edema, 1=macules of pin head size, 2=lentil sized macules, 3=confluent macules, 4=diffuse over entire site. Induration (palpable): 0=normal, 1=nodules of pin head size, 2=doughy lentil sized nodules, 3=confluent firm nodules, 4=diffuse hard lesion. The summed visual score at ca. 24 and 48 hours includes the individual scores for erythema intensity, erythema extent, and induration; so the maximal summed score for each site would be 12. High summed scores can generally indicate a high inflammatory response. Visual scores are subject to some inaccuracy / error.
Differences in the summed score between adjacent control (placebo) and treatment sites on the grids are calculated (see experimental design for details). This difference value is then used to determine the percent inhibition compared to the summed score for the control (placebo) sites. The more negative the difference value, the greater the calculated inhibition. Percent inhibition of (percent inhibition compared to) the mean summed score can be calculated.
About 24 and 48 hours after challenge, treatment sites can optionally also be visually evaluated for lesion area.
Experimental Design An 8 row by 3 column grid is outlined on the back of each pig. Due to potential heterogeneity in skin thickness, etc, across various regions of the back, the assignment of treatments and their controls were assigned to each row/column in a systematic manner. Specifically, each row contained a treatment and its control (n=1 or 2 of each in the 3 columns). Row assignments were randomized across pigs to ensure that treatments were not always in the same region of the pig's backs. Column assignments were also randomized: the number of treatments per row (either 1 or 2) was randomly chosen, and treatment and control assignments were randomized across the 3 columns.
Data Analysis Proper statistical analysis will incorporate features of the experimental design, which is generically known as a randomized block design (a block being a row on the grid). For each row, the average treatment and control response is calculated. The difference, d, between treatment and control is determined, and this is the response variable. The set of differences for each treatment and pig is analyzed using a 2 factor analysis of variance (ANOVA), with pig and treatment as factors. The average difference of each treatment group is tested against 0 using results from the two factor ANOVA; p-values < 0.05 are considered significant. (However, given the number of tests, a significant p- value cutoff of <0.01 might optionally (but not necessarily) be used to protect against possible false positive results.)
Statistical analysis which does not incorporate features of the randomized block design generally will not be as powerful (i.e. will tend to generate higher p-values). For example, pooling treatment and control data across all rows and columns from all pigs and conducting t-tests on treatment vs. control will be expected to have larger variance estimates because heterogeneity within and between pigs are part of the variance. In the 2 factor ANOVA on the differences, within and between effects are factored out of the variance resulting in more powerful tests. See Statistics for Experimenters, Box, Hunter and Hunter, 2nd edition, chapter 4 (Wiley and Sons, 2005).
Data are presented as Change (average difference in Matched pair scores) and p value (2 factor ANOVA via SAS software).
Results: Pig DTH assay - Study #8
A single specific ointment composition, being either Composition Example 1 or Composition Example 3 (Ointment D or D2) as disclosed hereinafter, and containing 2% w/w of the compound of the invention, and white petrolatum and ST-Cyclomethicone 5- NF ™, significantly (p < 0.05) inhibited inflammation scores at the 48 hour time point, but not at the 24 hour time point (placebo/control = Reference Composition Example 2 or 4 = Placebo Ointment AP or AP2, disclosed hereinafter). See the results tabulated below under the label "Modified White Petrolatum Ointment".
A single specific cream composition, being either Composition Example 5 or
Composition Example 7 (Water-in-oil cream Cr-D or Cr-D2) as disclosed hereinafter, and containing inter alia propylene glycol and 2% w/w of the compound of the invention, significantly (p < 0.05) inhibited inflammation scores at the 48 hour time point, but not at the 24 hour time point (placebo/control = Reference Composition Example 6 or 8 = Placebo Water-in-oil cream Cr-AP or Cr-AP2, disclosed hereinafter). See the results tabulated below under the label "Cream".
24 hr 48 hr
Modified White Modified White
Petrolatum Ointment Petrolatum Ointment
P P
Drug % Change study value Drug % Change study value
2 0.15 8 0.766 2 -1.10 8 0.031
24 hr 48 hr
Cream Cream
P P
Drug % Change study value Drug % Change study value
2 0.10 8 0.843 2 -1.10 8 0.031
Results: Pig DTH assay - Study #9
A single specific ointment composition, being either Composition Example 3 or Composition Example 1 (Ointment D2 or D) as disclosed hereinafter, and containing 2% w/w of the compound of the invention, and white petrolatum and ST-Cyclomethicone 5- NF ™, significantly (p < 0.05) inhibited inflammation scores at the 24 and 48 hour time point (placebo/control = Reference Composition Example 4 or 2 = Placebo Ointment AP2 or AP, disclosed hereinafter). See the results tabulated below under the label "Modified White Petrolatum Ointment".
A single specific cream composition, being either Composition Example 7 or Composition Example 5 (Water-in-oil cream Cr-D2 or Cr-D) as disclosed hereinafter, and containing inter alia propylene glycol and 2% w/w of the compound of the invention, appeared to be somewhat inhibitory (negative Change observed), but the differences (with p ≥ 0.05) did not reach statistical significance (placebo/control = Reference Composition Example 8 or 6 = Placebo Water-in-oil cream Cr-AP2 or Cr-AP, disclosed hereinafter). See the results tabulated below under the label "Cream".
24 hr 48 hr
Modified White Modified White
Petrolatum Ointment Petrolatum Ointment
P P
Drug % Change study value Drug % Change study value about
2 -1.29 9 0.028 2 -1.92 9 0.000
24 hr 48 hr
Cream Cream
P P
Drug % Change study value Drug % Change study value
2 -1.00 9 0.086 -0.65 9 0.156
Compounds or pharmaceutical compositions showing apparent activity in the above pig DTH model (In Vivo Assay A) may have potential utility via skin topical administration in the treatment and/or prophylaxis of atopic dermatitis, e.g. in humans. The preliminary nature of In Vivo Assay A, and the potential for error or for variation in results when the assay is repeated (see e.g. results of Study #8 cf. those of Study #9) is noted.
in Vivo Assay 1. LPS-induced pulmonary neutrophilia in rats: effect of orally administered PDE4 inhibitors Pulmonary neutrophil influx is thought to be a significant component to the family of pulmonary diseases like chronic obstructive pulmonary disease (COPD) which can involve chronic bronchitis and/or emphysema (G. F. Filley, Chest. 2000; 117(5); 251s- 260s). The purpose of this neutrophilia model is to study the potentially antiinflammatory effects in vivo of orally administered PDE4 inhibitors on neutrophilia induced by inhalation of aerosolized lipopolysaccharide (LPS), modelling the neutrophil inflammatory component(s) of COPD. See the literature section below for scientific background.
Male Lewis rats (Charles River, Raleigh, NC, USA) weighing approximately 300- 400 grams are pretreated with either (a) test compound, for example suspended in about 0.5% methylcellulose (obtainable from Sigma-Aldrich, St Louis, MO, USA) in water or (b) vehicle only, delivered orally in a dose volume of ca. 10 ml/kg. Generally, dose response curves can for example be generated using the following approx. doses of PDE4 inhibitors: 2.0, 0.4, 0.08, 0.016 and 0.0032 mg/kg (or alternatively approx. 10, 2.0, 0.4, 0.08 and 0.016 mg/kg). About thirty minutes following pretreatment, the rats are exposed to aerosolized LPS (Serotype E. CoIi 026:B6 prepared by trichloroacetic acid extraction, obtainable from Sigma-Aldrich, St Louis, MO, USA), generated from a nebulizer containing a ca. 100 μg/ml LPS solution (ca. 100 ug/ml). Rats are exposed to the LPS aerosol at a rate of ca. 4 L/min for ca. 20 minutes. LPS exposure is carried out in a closed chamber with internal dimensions of roughly 45 cm length x 24 cm width x 20 cm height. The nebulizer and exposure chamber are contained in a certified fume hood. At about 4 hours-post LPS exposure the rats are euthanized by overdose with pentobarbital at ca. 90 mg/kg, administered intraperitoneal^. Bronchoalveolar lavage (BAL) is performed through a 14 gauge blunt needle into the exposed trachea. Five, 5 ml washes are performed to collect a total of 25 ml of BAL fluid. Total cell counts and leukocyte differentials are performed on BAL fluid in order to calculate neutrophil influx into the lung. Percent neutrophil inhibition at each dose (cf. vehicle) is calculated and a variable slope, sigmoidal dose-response curve is generated, usually using Prism Graph- Pad. The dose-response curve is used to calculate an ED50 value (in mg per kg of body weight) for inhibition by the PDE4 inhibitor of the LPS-induced neutrophilia.
Alternative method: In an alternative simpler embodiment of the procedure, a single oral dose of 10 mg/kg, or more usually 1.0 mg/kg or 0.3 mg/kg, of the PDE4 inhibitor (or vehicle) is administered to the rats, and percent neutrophil inhibition is calculated and reported for that specific dose. Literature: Filley G. F. Comparison of the structural and inflammatory features of COPD and asthma. Chest. 2000; 117(5) 251 s-260s.
Howell RE, Jenkins LP, Fielding LE, and Grimes D. Inhibition of antigen-induced pulmonary eosinophilia and neutrophilia by selective inhibitors of phosphodiesterase types 3 and 4 in brown Norway rats. Pulmonary Pharmacology. 1995; 8: 83-89. Spond J, Chapman R, Fine J, Jones H, Kreutner W, Kung TT, Minnicozzi M. Comparison of PDE 4 inhibitors, Rolipram and SB 207499 (Ariflo™), in a rat model of pulmonary neutrophilia. Pulmonary Pharmacology and Therapeutics. 2001; 14: 157- 164.
Underwood DC, Osborn RR, Bochnowicz S, Webb EF, Rieman DJ, Lee JC, Romanic AM, Adams JL, Hay DWP, and Griswold DE. SB 239063, a p38 MAPK inhibitor, reduces neutrophilia, inflammatory cytokines, MMP-9, and fibrosis in lung. Am J Physiol Lung Cell MoI Physiol. 2000; 279: L895-L902.
In Vivo Assay 2. Rat Pica Model of emesis
Background: Selective PDE4 inhibitors are thought to inhibit inflammation in various in vitro and in vivo models by increasing intracellular levels of cAMP of many immune cells (e.g. lymphocytes, monocytes). However, a side effect of some PDE4 inhibitors in some species is emesis. Because many rat models of inflammation are well characterized, they can be used in procedures (see e.g. In Vivo Assay 1 above) to show beneficial anti-inflammatory effects of PDE 4 inhibitors. However rats have no emetic response (they have no vomit reflex), so that the relationship between beneficial antiinflammatory effects of PDE 4 inhibitors and emesis is difficult to study directly in rats.
However, in 1991 , Takeda et al. (see Literature section below) demonstrated that the pica feeding response is analogous to emesis in rats. Pica feeding is a behavioural response to illness in rats wherein rats eat non-nutritive substances such as earth or in particular clay (e.g. kaolin) which may help to absorb toxins. Pica feeding can be induced by motion and chemicals (especially chemicals which are emetic in humans), and can be inhibited pharmacologically with drugs that inhibit emesis in humans. The Rat Pica Model, In Vivo Assay 2, can determine the level of pica response of rats to PDE 4 inhibition at pharmacologically relevant doses in parallel to in vivo anti-inflammatory Assays in (a separate set of) rats (e.g. In Vivo Assay 1 above). Anti-inflammatory and pica assays in the same species together can provide data on the "therapeutic index" (Tl) in the rat of the compounds/salts of the invention. The Rat Tl can for example be calculated as the ratio of a) the potentially-emetic Pica Response ED50 dose from Assay 2 to b) the rat anti-inflammatory ED50 dose (e.g. measured by rat neutrophilia-inhibition in eg In Vivo Assay 1), with larger Tl ratios possibly indicating lower emesis at many anti-inflammatory doses. This might allow a choice of a non-emetic or low-emetic pharmaceutical dose of the compounds or salts of the invention which has an anti-inflammatory effect. It is recognised however that achieving a low-emetic PDE4 inhibitory compound is not essential to the invention.
Procedure: On the first day of the experiment, the rats are housed individually in cages without bedding or "enrichment". The rats are kept off of the cage floor by a wire screen. Pre-weighed food cups containing standard rat chow and clay pellets are placed in the cage. The clay pellets, obtainable from Languna Clay Co, City of Industry, CA, USA, are the same size and shape as the food pellets. The rats are acclimated to the clay for 72 hours, during which time the cups and food and clay debris from the cage are weighed daily on an electronic balance capable of measuring to the nearest 0.1 grams. By the end of the 72 hour acclimation period the rats generally show no interest in the clay pellets.
At the end of 72 hours the rats are placed in clean cages and the food cups weighed. Rats that are still consuming clay regularly are removed from the study. Immediately prior to the dark cycle (the time when the animals are active and should be eating) the animals are split into treatment groups and dosed orally with a dose of the compound or salt of the invention (different doses for different treatment groups) or with vehicle alone, at a dose volume of ca. 2 ml/kg. In this oral dosing, the compound or salt can for example be in the form of a suspension in about 0.5% methylcellulose (obtainable Sigma-Aldrich, St. Louis, MO, USA) in water. The food and clay cups and cage debris are weighed the following day and the total clay and food consumed that night by each individual animal is calculated.
A dose response is calculated by first converting the data into quanta! response, where animals are either positive or negative for the pica response. A rat is "pica positive" if it consumes greater than or equal to 0.3 grams of clay over the mean of its control group. The D50 value is usually calculated using logistic regression performed by the Statistica software statistical package. A Pica Response ED50 value in mg per kg of body weight can then be calculated.
The Pica Response ED50 value can be compared to the neutrophilia-inhibition ED50 values for the same compound administered orally to the rat (measurable by In
Vivo Assay 1 above), so that a Therapeutic Index (Tl) in rats can be calculated thus:
Rat Therapeutic index (Tl) (50/50) = Pica Response ED50 value rat neutrophilia-inhibition ED50 value
In general, the Therapeutic Index (Tl) calculated this way can be substantially different to, and for example (without being bound) can be substantially higher than, the Tl (D20/D50) calculated in the ferret (see In vivo Assay 3 below).
Alternatively, e.g. for a simpler test, the In Vivo Assay 2 (pica) can use only a single oral dose of the test compound (e.g. 10 mg/kg orally). Literature: Beavo JA, Contini, M., Heaslip, R.J. Multiple cyclic nucleotide phosphodiesterases. MoI Pharmacol. 1994; 46:399-405.
Spond J, Chapman R, Fine J, Jones H, Kreutner W, Kung TT, Minnicozzi M. Comparison of PDE 4 inhibitors, Rolipram and SB 207499 (Ariflo™), in a rat model of pulmonary neutrophilia. Pulmonary Pharmacology and Therapeudtics. 2001 ; 14:157- 164.
Takeda N, Hasegawa S, Morita M, and Matsunaga T. Pica in rats is analogous to emesis: an animal model in emesis research. Pharmacology, Biochemistry and Behavior. 1991 ; 45:817-821. Takeda N, Hasegawa S, Morita M, Horii A, Uno A, Yamatodani A and Matsunaga
T. Neuropharmacological mechanisms of emesis. I . Effects of antiemetic drugs on motion- and apomorphine-induced pica in rats. Meth Find Exp Clin Pharmacol. 1995; 17(9) 589-596.
Takeda N, Hasegawa S, Morita M, Horii A, Uno A, Yamatodani A and Matsunaga T. Neuropharmacological mechanisms of emesis. Il . Effects of antiemetic drugs on cisplatin-induced pica in rats. Meth Find Exp CHn Pharmacol. 1995; 17(9) 647-652.
In Vivo Assay 3. Evaluation of Therapeutic Index of Orally-administered PDE 4 inhibitors in the conscious ferret 1.1 Materials
The following materials can be used for these studies:
PDE4 inhibitors are prepared for oral (p.o.) administration by dissolving in a fixed volume
(ca. 1 ml) of acetone and then adding cremophor to ca. 20% of the final volume.
Acetone is evaporated by directing a flow of nitrogen gas onto the solution. Once the acetone is removed, the solution is made up to final volume with distilled water.
LPS is dissolved in phosphate buffered saline.
1.2 Animals
Male ferrets (Mustela Pulorius Furo, weighing 1 - 2 kg) are transported and allowed to acclimatise for not less than 7 days. The diet comprises SDS diet C pelleted food given ad lib with Whiskers^M cat food given 3 times per week. The animals are supplied with pasteurised animal grade drinking water changed daily.
1.3 Experimental Protocol(s)
1.3.1 Dosing with PDE4 inhibitors
PDE4 inhibitors are administered orally (p.o.), using a dose volume of ca. 1 ml/kg. Ferrets are fasted overnight but allowed free access to water. The animals are orally dosed with vehicle or PDE 4 inhibitor using a ca. 15cm dosing needle that is passed down the back of the throat into the oesophagus. After dosing, the animals are returned to holding cages fitted with perspex doors to allow observation, and given free access to water. The animals are constantly observed and any emetic episodes (retching and vomiting) or behavioural changes are recorded. The animals are allowed access to food ca. 60 - 90 minutes after p.o. dosing.
1.3.2 Exposure to LPS
About thirty minutes after oral dosing with compound or vehicle control, the ferrets are placed into sealed perspex containers and exposed to an aerosol of LPS (ca. 30 μg/ml = ca. 30 ug/ml) for ca. 10 minutes. Aerosols of LPS are generated by a nebuliser (DeVilbiss, USA) and this is directed into the perspex exposure chamber. Following a 10-minute exposure period, the animals are returned to the holding cages and allowed free access to water, and at a later stage, food. General observation of the animals continues for a period of at least 2.5 hours post oral dosing. All emetic episodes and behavioural changes are recorded.
1.3.3 Bronchoalveolar lavage and cell counts
About six hours after LPS exposure the animals are killed by overdose of sodium pentobarbitone administered intraperitoneally. The trachea is then cannulated with polypropylene tubing and the lungs lavaged twice with ca. 20 ml heparinised (10 units/ml) phosphate buffered saline (PBS). The bronchoalveolar lavage (BAL) samples are centrifuged at ca. 1300 rpm for ca. 7 minutes. The supernatant is removed and the resulting cell pellet re-suspended in ca. 1 ml PBS. A cell smear of re-suspended fluid is prepared and stained with Leishmans stain to allow differential cell counting. A total cell count is made using the remaining re-suspended sample. From this, the total number of neutrophils in the BAL sample is determined.
1.3.4 Pharmacodynamic readouts The following parameters are recorded: a) % inhibition of LPS-induced pulmonary neutrophilia to determine the dose of PDE4 inhibitor which gives 50% inhibition (D50). b) Emetic episodes - the number of vomits and retches are counted to determine the dose of PDE4 inhibitor that gives a 20% incidence of emesis (D20). c) A therapeutic index (Tl), using this assay, is then calculated for each PDE4 inhibitor using the following equation:
Ferret Therapeutic index (Tl) (D20/D50) = D20 incidence of emesis in ferret
D50 inhibition of neutrophilia in ferret
It is noted that the Ferret Therapeutic index (Tl) (D20/D50) calculated using this in vivo Assay 3 can be substantially different to, and for example (without being bound) can be substantially lower than, the Rat Tl (50/50) calculated using the rat oral inflammation and pica feeding Assays 1+2.
All publications, including but not limited to patents and patent applications, cited in this specification are herein incorporated by reference as if each individual publication were specifically and individually indicated to be incorporated by reference herein as though fully set forth. EXAMPLES
The various aspects of the invention will now be described by reference to the following examples. These examples are merely illustrative and are not to be construed as a limitation of the scope of the present invention.
In this section, "Intermediates" can represent syntheses of intermediate compounds intended for use in the synthesis of one or more of the "Examples", and/or "Intermediates" can represent syntheses of intermediate compounds which can be used in the synthesis of compounds of formula (I) or salts thereof. "Examples" are examples of a compound or salt of the invention, i.e. examples of a compound of formula (I) or a salt thereof.
Abbreviations used herein:
AcOH acetic acid
Ac2O acetic anhydride
BEMP 2-t-butylimino-2-diethylamino-1 ,3-dimethylperhydro-1 ,3,2- diazaphosphazine
BOC2O di tert-butyl carbonate
DMSO dimethyl sulfoxide
DCM dichloromethane
DMF dimethyl formamide
DIPEA diisopropylethyl amine ('P^NEt)
EDC 1 -(3-Dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride
EtOAc ethyl acetate
Et2O diethyl ether
Et3N triethylamine
EtOH ethanol
HATU 0-(7-Azabenzotriazol-1-yl)-/V,/V,N',N'-tetramethyluronium hexafluorophosphate
HBTU 0-(Benzotriazol-1-yl)-Λ/,/V,Λ/',Λ/'-tetramethyluronium hexafluorophosphate
HOBT hydroxybenzotriazole = 1-hydroxybenzotriazole
Lawesson's reagent 2,4-bis(4-methoxyphenyl)-1 ,3-dithia-2,4-diphosphetane-2,4- disulphide
MeCN acetonitrile
MeOH methanol
PyBOP Benzotriazole-1-yl-oxy-trispyrrolidinophosphonium hexafluorophosphate
THF Tetrahydrofuran
TFA Trifluoroacetic acid τint internal temperature of the reaction mixture HPLC high performance liquid chromatography h hours min minutes
LCMS liquid chromatography / mass spectroscopy NMR nuclear magnetic resonance (in which: s = singlet, d = doublet, t = triplet, q = quartet, dd = doublet of doublets, m = multiplet, H = no. of protons)
SPE solid phase extraction
TLC thin layer chromatography
TRET retention time (e.g. from LCMS) Room temperature (ambient temperature): this is usually in the range of about 18 to about 25 0C.
General Experimental Details
Machine Methods used herein:
LCMS (liquid chromatography / mass spectroscopy)
Waters ZQ mass spectrometer operating in positive ion electrospray mode, mass range 100-1000 amu.
UV wavelength : 215-330 nm
Column : 3.3cm x 4.6mm ID (internal diameter), 3μm (3 micrometres) ABZ+PLUS
Flow Rate : 3ml/min
Injection Volume : 5μl (5 microlitres) Solvent A : 0.05% v/v solution of formic acid in a mixture of [95% acetonitrile and 5% water]
Solvent B : aqueous solution of [0.1% v/v formic acid + IOmMolar ammonium acetate]
Gradient : Mixtures of Solvent A and Solvent B are used according to the following gradient profiles (expressed as % Solvent A in the mixture): 0% A/0.7min, 0-100% A/3.5min, 100% A/1.1 min, 100-0% A/0.2min
It should be noted that retention times (TREτ) quoted herein are inherently variable (e.g. the variability can be about +/- 0.2 min or more.). Variability can arise e.g. when samples are run on different Waters machines, or on the same Waters machine at different times of day or under slightly different conditions, even when the same type of column and identical flow rates, injection volumes, solvents and gradients are used.
Mass directed autoprep HPLC
The preparative HPLC column generally used is a Supelcosil ABZplus (10cm x 2.12cm internal diameter; particle size 5μm = 5 micrometres) e.g. for eluent containing formic acid, or is a C18 column (10cm x 2.12cm internal diameter; particle size 5μm = 5 micrometres) e.g. for eluent containing trifluoroacetic acid . A mass spectrometer attached to the end of the column can detect peaks arising from eluted compounds. UV wavelength : usually 200-320 nm
Flow : 20ml/min
Injection Volume: 0.5 to 1ml
Gradient systems: mixtures of Solvent A and Solvent B are used according to a choice of 5 generic gradient profiles (expressed as % Solvent B in the mixture), ranging from a start of 0 to 50% Solvent B, with all finishing at 100% Solvent B to ensure total elution.
Generally, two alternative solvent systems have been used, Method 1 and Method 2:
Method 1
Solvent A : 0.1% v/v aqueous formic acid solution Solvent B : 0.05% v/v solution of formic acid in a mixture of [95% acetonitrile and 5% water]
It is thought that compounds isolated by this method can sometimes be isolated as formate salts.
Method 2 Solvent A: 0.1 % v/v aqueous trifluoroacetic acid solution
Solvent B: solution of 0.1% v/v trifluoroacetic acid in acetonitrile
It is thought that compounds isolated by this method can sometimes be isolated as trifluoroacetate salts.
Intermediates and Examples
Reagents not detailed in the text below are usually commercially available from chemicals suppliers, e.g. established suppliers such as Sigma-Aldrich. The addresses and/or contact details of the suppliers for some of the starting materials mentioned in the Intermediates and Examples below or the Assays above, or suppliers of miscellaneous chemicals in general, are as follows:
- Albemarle Corporation, 451 Florida Street, Baton Rouge, LA 70801 , USA; or Albemarle Europe SPRL, Pare Scientific de LLN, Rue du Bosquet 9, B-1348 Louvain-la-Neuve, Belgium
- Acros Organics, A Division of Fisher Scientific Company, 500 American Road, Morris Plains, NJ 07950, USA
- Apin Chemicals Ltd., 82 C Milton Park, Abingdon, Oxon OX14 4RY, United Kingdom
- Aldrich (catalogue name), Sigma-Aldrich Company Ltd., Dorset, United Kingdom, telephone: +44 1202 733114; Fax: +44 1202 715460; ukcustsv@eurnotes.sial.com; or
- Aldrich (catalogue name), Sigma-Aldrich Corp., P.O. Box 14508, St. Louis, MO 63178- 9916, USA; telephone: +1-314-771-5765; fax: +1-314-771-5757; custserv@sial.com; or
- Aldrich (catalogue name), Sigma-Aldrich Chemie GmbH, Munich, Germany; telephone: +49 89 6513 0; Fax: +49 89 6513 1169; deorders@eurnotes.sial.com. - Amersham Biosciences UK Ltd, Pollards Wood, Chalfont St Giles, Buckinghamshire HP8 4SP, United Kingdom
- Art-Chem GmbH: (a) Lininsky prospect 47, Moscow 119991 , Russia, or (b) Campus Berlin-Buch, Haus B55, Robert-Roessle Strasse 10, 13125 Berlin, Germany - AstaTech, Inc., 8301 Torresdale Ave., 19C, Philadelphia, PA 19136, USA
- Bayer AG, Business Group Basic and Fine Chemicals, D-51368 Leverkusen, Germany
- Chemical Building Blocks (catalogue name), Ambinter, 46 quai Louis Bleriot, Paris, F-75016, France - Combi-Blocks Inc., 7949 Silverton Avenue, Suite 915, San Diego, CA 92126, USA
- Enamine, 23 A. Motrosova Street, Kiev 01103, Ukraine
- Fluka Chemie AG, lndustriestrasse 25, P.O. Box 260, CH-9471 Buchs, Switzerland
- Fluorochem Ltd., Wesley Street, Old Glossop, Derbyshire SK13 7RY, United Kingdom
- lnterchim Intermediates (catalogue name), Interchim, 213 Avenue Kennedy, BP 1140, Montlucon, Cedex, 03103, France
- Lancaster Synthesis Ltd., Newgate, White Lund, Morecambe, Lancashire LA3 3DY, United Kingdom
- Matrix Scientific, P.O. Box 25067, Columbia, SC 29224-5067, USA
- MicroChemistry Building Blocks (catalogue name), MicroChemistry-RadaPharma, Shosse Entusiastov 56, Moscow, 111123, Russia
- Molecular Devices Corporation, Sunnydale, CA, USA
- Oakwood Products Inc., 1741 , Old Dunbar Road, West Columbia, SC, 29172, USA
- Peakdale Molecular Ltd., Peakdale Science Park, Sheffield Road, Chapel-en-le-Frith, High Peak SK23 OPG, United Kingdom - SALOR (catalogue name) (Sigma Aldrich Library of Rare Chemicals), Aldrich Chemical Company Inc, 1001 West Saint Paul Avenue, Milwaukee, Wl 53233, USA
- Sigma (catalogue name), Sigma-Aldrich Corp., P.O. Box 14508, St. Louis, MO 63178- 9916, USA; see "Aldrich" above for other non-US addresses and other contact details
- SIGMA-RBI, One Strathmore Road, Natick, MA 01760-1312, USA - TimTec Corporation (e.g. "TimTec Overseas Stock"), 100 Interchange Boulevard, Newark, DE 19711 , USA
- "TimTec Building Blocks A or B", or "TimTec Stock Library", TimTec, Inc., P O Box 8941 , Newark, DE 19714-8941 , USA
INTERMEDIATES
Intermediate 1 Ethyl 4-chloro-1,6-diethyl-1H-pyrazolo[3,4-Jb]pyridine-5- carboxylate
Figure imgf000067_0001
Diethyl propanoylpropanedioate which is Et-C(O)-C(CO2Et)2 [e.g. see J. Org. Chem.,
(1976), 41 (24), 3857] (2.74g) was dissolved in phosphoryl chloride (30ml), tributylamine (3ml) was added cautiously, and the mixture was heated to 11O0C for 5h. The mixture was concentrated under reduced pressure to remove the phosphoryl chloride, and the residue was dissolved in diethyl ether (about 20ml) and n-hexane added until two layers formed. The top layer (diethyl ether) was collected and the diethyl ether/hexane extraction procedure repeated twice more. The combined ether extracts were washed with 1 N HCI solution (2 x 50ml), 1 N sodium hydroxide solution and water, and were dried and evaporated to give diethyl (i-chloropropylidene)propanedioate (1.61g). LCMS showed MH+ = 235; TRET = 3.21 min.
A mixture of diethyl (i-chloropropylidene)propanedioate (1.61g) and 1-ethyl-1H-pyrazol- 5-amine [e.g. available from Aldrich Chemical Company Inc., Albemarle Corporation, Art- Chem GmbH, Enamine and/or TimTec Corporation] (0.755g) in toluene (40ml) was treated with triethylamine (1.8ml) and the mixture heated at 12O0C for 16h. The solvent was evaporated under reduced pressure and the residue was dissolved in phosphoryl chloride (50ml) and heated at 1100C overnight. The mixture was evaporated under reduced pressure to remove the phosphoryl chloride, and the residue was partitioned between ethyl acetate and saturated aqueous sodium hydrogen carbonate solution. The organic layer was separated, dried (sodium sulphate) and evaporated under reduced pressure. The residue was purified on an SPE cartridge (silica, 5Og) eluting with 5% ethyl acetate in cyclohexane to give the title compound as a yellow oil (1.22g). LCMS showed MH+ = 282; TRET = 3.40min.
Intermediate 1 (Alternative Synthesis B) Ethyl 4-chloro-1,6-diethyl-1H- pyrazolo[3,4-Jb]pyridine-5-carboxylate
Figure imgf000067_0002
Triethylamine (230 ml) is added dropwise to a mixture of diethyl (i-chloropropylidene)propanedioate (208g) and 1-ethyl-1H-pyrazol-5-amine (101g) in toluene (2.65L). The mixture is heated under reflux for 16 hours. The reaction mixture is cooled to room temperature, and the solid removed by filtration. The filtrate is evaporated under reduced pressure. The residue is heated under reflux in phosphorus oxychloride (POCI3, 2.65L) for 16hrs. Excess phosphorus oxychloride is removed under reduced pressure and the cooled mixture is poured onto a mixture of saturated aqueous NaHCOβ solution (4L) and EtOAc (1.5L). The organic layer is separated and the aqueous layer further extracted with ethyl acetate (2x1 L). The combined EtOAc extracts are washed with saturated aqueous NaHCθ3 solution (2x2L) and dried (Na2SO4).
Evaporation of solvent under reduced pressure affords the crude product. The crude product is purified by chromatography (silica gel, 60-120 mesh, 3.5 kg), eluting with 3% EtOAc in hexane. Fractions containing the product are pooled and evaporated to give the title compound.
Intermediate 2 Ethyl 1,6-diethyl-4-(tetrahydro-2H-pyran-4-ylamino)-1H- pyrazolo[3,4-Jb]pyridine-5-carboxylate
Figure imgf000068_0001
Ethyl 4-chloro-1 ,6-diethyl-1/-/-pyrazolo[3,4-jb]pyridine-5-carboxylate (0.5Og) (e.g. this can be as prepared in Intermediate 1) was dissolved in 1-methyl-2-pyrrolidinone (5ml) and treated with tetrahydro-2H-pyran-4-amine hydrochloride (0.49g) [e.g. this can be as prepared in Intermediate 12, see below] and DIPEA (0.60ml) at 1200C overnight. The mixture was allowed to cool and was partitioned between ethyl acetate (3 x 50ml) and water (50ml). The organic layer was separated, dried and evaporated in vacuo. The residue was purified on an SPE cartridge (e.g. solid phase can be ca. 2Og or ca. 5Og; e.g. can be silica) eluting with from 5% to 20% ethyl acetate in cyclohexane to give the title compound (0.413g). LCMS showed MH+ = 347; TRET = 3.05min.
Intermediate 2 (Alternative Synthesis B) Ethyl 1,6-diethyl-4-(tetrahydro-2H- pyran-4-ylamino)-1H-pyrazolo[3,4-Jb]pyridine-5-carboxylate
Figure imgf000068_0002
To a solution of ethyl 4-chloro-1 ,6-diethyl-1H-pyrazolo[3,4-ιb]pyridine-5-carboxylate (36g, 127.8mmol) (which can e.g. be as prepared in Intermediate 1 ) in 1-methyl-2-pyrrolidinone (300ml) is added DIPEA (44.5ml, 255.6mmol). Tetrahydro-2H- pyran-4-amine (e.g. available from Peakdale and/or Combi-Blocks Inc., 15.5g, 153.3mmol) is added and the reaction mixture is heated at 1150C with stirring overnight. The cooled mixture is poured into water (1200ml), which may form an oily mixture. This is extracted with EtOAc (4x250ml), and the organic extracts are combined, washed with water (50ml), 5% aqueous LiCI solution (50ml), dried (MgSC>4), filtered and evaporated.
The residue is purified by silica gel (1kg) chromatography eluting with 2:1 cyclohexane:EtOAc (6000ml) followed by 1 :1 cyclohexane:EtOAc (3000ml). The fractions containing product are pooled and evaporated to give the title compound.
Reference Intermediate 3 [1-ethyl-4-(tetrahydro-2H-pyran-4-ylamino)-1H- pyrazolo[3,4-Jb]pyridin-5-yl]methanol
Figure imgf000069_0001
A solution of 1M diisobutylaluminium hydride in dichloromethane (80ml) was added dropwise to a stirred solution of ethyl 1-ethyl-4-(tetrahydro-2H-pyran-4~ylarnino)-1H- pyrazolo[3,4-jb]pyridine-5-carboxylate (13.8Og) [e.g. see Intermediate 32 and/or Example 3 of WO 2004/024728 A2, and/or Intermediate 4 of WO 2005/058892 A1] in dichloromethane (75ml) at O0C under a nitrogen atmosphere. The reaction mixture was maintained below 50C during the addition, and was then stirred at about O0C. The reaction mixture was then quenched by addition of aqueous potassium sodium tartrate (10% solution), diluted with water (150ml) and the organic phase separated. The aqueous phase was extracted with ethyl acetate (2 x 250ml) and the combined organics were dried (magnesium sulphate) and evaporated. The residue was divided into three portions and each portion was purified by column chromatography on silica gel (100g) eluting with a gradient of from 0 to 100% ethyl acetate in cyclohexane followed by from 0 to 20% methanol in ethyl acetate, and appropriate fractions were combined and evaporated, to give the title compound as a white solid (10.29g). LCMS showed MH+ = 277; TRET = 1.81 min.
Intermediate 4 [1 ,6-diethyl-4-(tetrahydro-2H-pyran-4-ylamino)-1 H- pyrazolo[3,4-Jb]pyridin-5-yl]methanol
Figure imgf000070_0001
Intermediate 4 can be prepared, in an analogous manner to Reference Intermediate 3, from ethyl 1 ,6-diethyl-4-(tetrahydro-2H-pyran-4-ylamino)-1H-pyrazolo[3,4-ιb]pyridine-5- carboxylate (e.g. which can be as prepared in Intermediate 2).
One specific synthesis of Intermediate 4:
Two reactions were done. The first reaction was as follows: To ethyl 1 ,6-diethyl-4- (tetrahydro-2H-pyran-4-ylamino)-1 H-pyrazolo[3,4-<b]pyridine-5-carboxylate (7.5g) (e.g. which can be as prepared in Intermediate 2) in dichloromethane (75ml) under nitrogen at 0 0C was added a solution of diisobutyaluminium hydride in toluene (1.5M, 43ml), dropwise, keeping the temperature at 0 0C. The addition took 9.5mins. Stirring at 0 0C was continued for 30mins; then the reaction was quenched with saturated aqueous sodium potassium tartrate solution (30ml). There was much effervescence. The mixture gelled which made work-up more difficult. Ethyl acetate and water were added and the layers separated. The aqueous phase was extracted with more ethyl acetate and the combined organic extracts were washed with brine, dried and evaporated to give a white solid (6.13g). The second duplicate reaction was generally the same (also using a 9.5mins addition), and gave 6.59g of product. The two batches of product from each reaction were each dissolved in chloroform and were filtered. The solutions from each duplicate reaction were combined and then evaporated to give the title compound as a white solid (12.57g). LCMS showed MH+ 305; TRET = 1.84min.
Intermediate 4 (Alternative Synthesis B) [1 ,6-diethyl-4-(tetrahydro-2H-pyran-4- ylamino)-1 H-pyrazolo[3,4-b]pyridin-5-yl]methanol
Figure imgf000070_0002
Intermediate 4 (Alternative Synthesis B) Process Scheme
Figure imgf000071_0001
inorganics
Intermediate 4 (Alternative Synthesis B) Process Summary
Note: All or most of this process is carried out under a nitrogen atmosphere. All weights, volumes and equivalents are relative to ethyl 1 ,6-diethyl-4-(tetrahydro-2H-pyran-4- ylamino)-1H-pyrazolo[3,4-o]pyridine-5-carboxylate.
To a stirred solution of lithium borohydride (3eq, 4.34vol, 2M in THF) and toluene (4 vols) at 64-68 0C, is added a solution of ethyl 1 ,6-diethyl-4-(tetrahydro-2H-pyran-4-ylamino)- 1H-pyrazolo[3,4-ιb]pyridine-5-carboxylate (1wt) in THF (1vol) and methanol (9eq, 1.05vol) over 2.5 hours. The reaction is monitored by HPLC and is typically complete 60 minutes after the addition. The reaction mixture is then cooled down to 20±3°C and water (3 vol) is added dropwise over 20mins followed by 10.8M NaOH (6.0 vol) (dropwise over 10 mins), ensuring the temperature remains below 40 0C. The temperature of the reaction is then adjusted to 40±3°C and stirred vigorously for at least 90 minutes. The organic layer is separated and the aqueous layer is transferred to a Schott bottle. Water is added to the organic layer (2.5 vols) followed by NaOH (10.8M, 2 vols), the biphase is stirred vigorously at 40 0C for at least 30 minutes. The organic layer is again separated and the organic solution is concentrated in vacuo to 3vol, and the product precipitates as yellow solid. Toluene (3vol) is then added and the slurry is reconcentrated in vacuo to 3vol. The suspension is then cooled to 10±3°C and aged for at least 30min. The solid is then filtered and washed with toluene (3vol). The product is then dried in vacuo at 450C to constant temperature.
Intermediate 4 (Alternative Synthesis B) Procedure (can be run in 1.0L or 10.0-L equipment)
Note: All or most of this process is carried out under a nitrogen atmosphere. All weights, volumes and equivalents are relative to ethyl 1 ,6-diethyl-4-(tetrahydro-2H-pyran-4- ylamino)-1H-pyrazolo[3,4-6]pyridine-5-carboxylate.
1. Purge vessel with nitrogen.
2. Charge toluene (4 vols) to the vessel, mark 3 volumes during addition. 3. Charge LiBH4 (2M in THF, 4.34 vols) to vessel. 4. Heat contents to 64-68 0C.
5. Charge a Schott bottle with ethyl 1 ,6-diethyl-4-(tetrahydro~2H-pyran-4-ylamino)-1H- pyrazolo[3,4-ϋ]pyridine-5-carboxylate (1wt), add THF (1 vol) and MeOH (1.05vols) and stir to dissolve contents. 6. Add the solution from step 5 to the LiBH4 solution via peristaltic pump fitted with silicon tubing over 2.5 hours. Maintain Tjn^ at 64-68 0C.
7. Stir the mixture for at 50-70 minutes at 64-68 0C.
8. Sample the reaction and check for completeness by generic HPLC (typically complete at 60 minutes). The reaction can be held at 2O0C overnight at this point. Do not heat reaction beyond 70 minutes; start to cool at 70 minutes.
9. Cool contents of vessel to 20±3 0C, add water (3vols) to the vessel via peristaltic pump fitted with silicon tubing over at least 20 minutes. Ensure Tjn^ <40 0C.
10. Adjust contents to 20±3 0C and add 10.8M sodium hydroxide (6.0 vols) to the vessel via peristaltic pump fitted with silicon tubing over 10 minutes, stir vigorously. 11. Adjust contents to 40±3 0C and stir for at least 90 minutes then allow layers to separate (separation <3minutes).
12. Remove bottom aqueous layer from vessel. The organic layer can be held at this point at 2O0C overnight.
13. Add water (2.5 vols) and NaOH (10.8M, 2 vols) to the organic layer, stir biphase for at least 30 minutes at 40 0C and then allow layers to separate.
14. Remove bottom aqueous layer from vessel.
15. Concentrate the vessel contents to 3vols using vacuum distillation.
16. Add toluene to the vessel (3vols) and reconcentrate the contents to 3vol via vacuum distillation. 17. Cool the contents to 10±3 0C and stir for at least 30 minutes. The batch can be held overnight at this point.
18. Filter off the product and wash the cake with toluene (1x3vol) before sucking the cake dry (filtration times are both less than 2 minutes).
19. Transfer product to a vacuum oven and dry to constant temperature at 45-50 0C under vacuum (drying complete in less than 24 hours).
20. A yellow solid is obtained of [1 ,6-diethyl-4-(tetrahydro-2/-/-pyran-4-ylamino)~1H- pyrazolo[3,4-ό] pyrid in-5-yl] methanol .
21. For dispposal of aqueous layer, aqueous layer ex-step 12 and 14 can be charged to a vessel and the contents adjusted to 10±3°C. The stirred solution is acidified using 5M H2SO4 (11 vols) which is added over approx 1 hour (caution, exothermic). Check pH <3. Reference Intermediate 5 5-(chloromethyl)-1-ethyl-W-(tetrahydro-2H-pyran-4-yl)- 1H-pyrazoIo[3,4-Jb]pyridin-4-amine
Figure imgf000073_0001
[1 -ethyl-4-(tetrahydro-2H-pyran-4-yla Jmino)- H-pyrazolo[3,4-ιb]pyridin-5-yl]methanol (80mg) (e.g. which can be as prepared in Reference Intermediate 3) was treated with thionyl chloride (1ml), heated at 8O0C for 1h and then allowed to cool. The orange solution was evaporated to dryness and the residue azeotroped with toluene (2 x 5ml) to give the title compound as a brown foam. LCMS of the compound in methanol showed MH+ 291 , TRET = 1 -90min, which is consistent with the 5-(chloromethyl)-pyrazolo[3,4- jb]pyridine derivative reacting with the methanol during LCMS to give the corresponding 5-(methoxymethyl)-pyrazolo[3,4-jb]pyridine derivative.
Intermediate 6 5-(chloromethyl)-1 ,6-diethyl-tø-(tetrahydro-2H-pyran-4-yI)-1 H- pyrazolo[3,4-Jb]pyridin-4-amine
Figure imgf000073_0002
Intermediate 6 can be prepared, in an analogous manner to Reference Intermediate 5, from [1 ,6-diethyl-4-(tetrahydro-2H-pyran-4-ylamino)-1 H-pyrazolo[3,4-ib]pyridin-5- yljmethanol (e.g. which can be as prepared in Intermediate 4).
One specific synthesis of Intermediate 6:
[1 ,6-diethyl-4-(tetrahydro-2H-pyran-4-ylamino)-1H-pyrazolo[3,4-ib]pyridin-5-yl]methanol (0.12Og) (e.g. which can be Intermediate 4) is treated with thionyl chloride (1.5ml), and the solution is heated at 80 0C for 1.5h and then allowed to cool. The reaction mixture is concentrated to dryness and then azeotroped with toluene (5ml) to leave the title compound.
Reference Intermediate 7 5-(azidomethyl)-1-ethyl-Λf-(tetrahydro-2H-pyran-4-yl)- 1H-pyrazolo[3,4-Jb]pyridin-4-amine
Figure imgf000074_0001
[1-ethyl-4-(tetrahydro-2/-/-pyran-4-ylamino)-1W-pyrazolo[3,4-jb]pyridin-5-yl]methanol (0.552 g) (e.g. which can be as prepared in Reference Intermediate 3) was treated with thionyl chloride (2ml, excess) and the solution was heated at 800C for 2h and then allowed to cool. The thionyl chloride was evaporated off and the resulting solid was azeotroped with toluene (2 x 5ml) to leave a yellow solid.
A solution of this solid, presumed to be 5-(chloromethyl)-1-ethyl-Λ/-(tetrahydro- 2W-pyran-4-yl)-1W-pyrazolo[3,4-fe]pyridin-4-amine, (50mg) in anhydrous dimethylsulphoxide (0.20ml) was treated with lithium azide (9mg) and the solution stirred at room temperature for 2Oh. A further portion of lithium azide (15mg) was then added, and, after a further day of stirring at room temperature, water (0.25ml) was added. The solution was extracted with dichloromethane (2 x 5ml) and the combined organic extracts were passed through a hydrophobic frit (6ml) then blown to dryness to leave a clear colourless gum. This was dissolved in dichloromethane (0.5ml) and applied to an SPE cartridge (silica; 1g). The cartridge was eluted with 50% ethyl acetate in cyclohexane and fractions containing the desired material were combined and blown to dryness to give the title compound as a clear colourless gum (10mg). LCMS showed MH+ = 302; TRET = 2.06min.
Intermediate 8 5-(azidomethyl)-1,6-diethyl-yV-(tetrahydro-2H-pyran-4-yl)-1H- pyrazolo[3,4-Jb]pyridin-4-amine
Figure imgf000074_0002
It is thought that Intermediate 8 can be prepared, in an analogous manner to Reference Intermediate 7, from 5-(chloromethyl)-1,6-diethyl-N-(tetrahydro-2W-pyran-4-yl)-1W- pyrazolo[3,4-/)]pyridin-4-amine (e.g. which can be as prepared in Intermediate 6).
One specific synthesis of Intermediate 8:
To [1 ,6-diethyl-4-(tetrahydro-2W-pyran-4-ylamino)-1 H-pyrazolo[3,4-/?]pyridin-5- yl]methanol (12.57g) (e.g. which can be as prepared in Intermediate 4) was added thionyl chloride (51 ml, 83.6g) and the mixture was heated to 80 °C. There was effervescence during the addition, and the reaction mixture turned yellow, and then red on heating. After 5h excess thionyl chloride was removed on an evaporator and the wine-red residue was azeotroped with toluene (75ml) to leave a residue containing 5-
(chloromethyl)-1 ,6-diethyl-Λ/-(tetrahydro-2H-pyran-4-yl)-1/-/-pyrazolo[3,4-b]pyridin-4- amine.
This residue was dissolved in DMF (100ml) and treated with sodium azide (4.Og) and the mixture was stirred at room temperature overnight. Water was added, followed by 2M sodium hydroxide solution to adjust from a pH of about 3.5 to a pH of about 8. Ethyl acetate was added and the layers separated. The aqueous phase was extracted with more ethyl acetate. Then the combined organics were washed with water, aqueous lithium chloride solution and brine, and were dried and evaporated to give an orange-brown oil which was subject to high vacuum. This was purified by column chromatography using 400ml of 7734 silica gel eluting with a 3 : 2 mixture of cyclohexane : ethyl acetate, and fractions containing a mixture of two products (as nmeasured by TLC) were collected and reduced to give the title compound as a golden oil which started to crystallise under high vacuum (9.74g). LCMS showed MH+ = 330; TRET = 2.30min.
Reference Intermediate 9 5-(aminomethyl)-1-ethyl-</V-(tetrahydro-2H-pyran-4-yl)- 1H-pyrazolo[3,4-/j]pyridin-4-amine
Figure imgf000075_0001
A solution of 5-(azidomethyl)-1-ethyl-/V-(tetrahydro-2H-pyran-4-yl)-1H-pyrazolo[3,4-
6]pyridin-4-amine (0.351g) (e.g. which can be as prepared in Reference Intermediate 7) in ethanol (30ml) was added to 5% palladium on carbon (wet) (0.05Og) and the mixture was stirred at room temperature for 20 hours under an atmosphere of hydrogen. The mixture was filtered first through a glass fibre filter and then through a plug of celite, which was then washed with ethanol (about 50ml). The combined filtrate and washings were evaporated to dryness to give the title compound as a dirty green gum (0.318g). LCMS showed MH+ = 276; TRET = 1.63min.
Intermediate 10 5-(aminomethyO-1,6-diethyl-/V-(tetrahydro-2W-pyran-4-yl)-1W- pyrazolo[3,4-Jb]pyridin-4-amine
Figure imgf000075_0002
It is thought that Intermediate 10 can be prepared, in an analogous manner to Reference Intermediate 9, from 5-(azidomethyl)-1 ,6-diethyl-Λ/-(tetrahydro-2H-pyran-4-yl)-1tf- pyrazolo[3,4-6]pyridin-4-amine (e.g. which can be as prepared in Intermediate 8).
One specific synthesis of Intermediate 10:
To 10% by weight palladium on carbon (0.4g) was added ethanol (10ml) followed by 5-(azidomethyl)-1 ,6-diethyl-Λ/-(tetrahydro-2H-pyran-4-yl)-1H-pyrazolo[3,4-ib]pyridin-4- amine (4g) (e.g. which can be as prepared in Intermediate 8) in ethanol (90ml) in a hydrogenation flask. The mixture was then hydrogenated at room temperature and atmospheric pressure with stirring overnight. Then, the palladium on carbon catalyst was filtered off twice to give a solution. The solvent was removed to give the title compound as a grey solid (2.59g). LCMS showed MH+ = 304; TRET = 1.63min. LCMS showed MH+ = 304; TRET = 1.63min.
Intermediate 11 5-(aminomethyl)-1 ,6-diethyl-W-(tetrahydro-2H-pyran-4-yI)-1 H- pyrazolo[3,4-Jb]pyridin-4-amine hydrochloride
Figure imgf000076_0001
Intermediate 11 Process Scheme:
Figure imgf000076_0002
SOCI2 (1.15eq), anisole
Benzenesulphonic acid (1eq)
Toluene LiHMDS (2.5eq), THF
5M HCI, NaOH, Me-THF, cHCI
Figure imgf000076_0003
Intermediate 11 lntermediate 11 Process Summary
Note: All or most of this process is carried out under a nitrogen atmosphere.
[1 ,6-diethyl-4-(tetrahydro-2H-pyran-4-ylamino)-1 H-pyrazolo[3,4-ύ]pyridin-5-yl]methanol (e.g. which can be as prepared in Intermediate 4, e.g. Alternative Synthesis B thereof) is suspended in anisole and treated with solid benzensulfonic acid. This suspension is aged at room temperature for 30 minutes. Thionyl chloride is added at about 20 degrees C over 20 minutes, and stirred for 20 minutes. The reaction is then sampled for HPLC. The resulting solution of 5-(chloromethyl)-1 ,6-diethyl-Λ/-(tetrahydro-2/-/-pyran-4-yl)-1 H- pyrazolo[3,4-jb]pyridin-4-amine benzenesulfonate is treated with toluene and then placed under medium vacuum (100 mbar) to remove hydrogen chloride and sulfur dioxide, then the vacuum is increased (50 mbar) to azeotrope off excess thionyl chloride and toluene.
This solution of 5-(chloromethyl)-1 ,6-diethyl-Λ/-(tetrahydro-2H-pyran-4-yl)-1 H- pyrazolo[3,4-(b]pyridin-4-amine benzenesulfonate is added to a solution of lithium hexamethyldisilazide (LiHMDS) in tetrahydrofuran at 35-40 degrees C over about an hour and the reaction is usually complete after a further 10 minutes.
The mixture is cooled to 10 degrees C and hydrochloric acid (5M) is added. The phases are separated and the lower aqueous layer is transferred back into the vessel, and the organic phase is discarded. Methyl-THF (methyl-tetrahydrofuran) is added and then the biphasic mixture is adjusted to pH >13 with sodium hydroxide solution (10M). The layers are separated and the lower aqueous layer is back extracted with further methyl-THF. The combined organic phase is washed with brine. The amount of the desired 5-(aminomethyl)-1 ,6-diethyl-Λ/-(tetrahydro-2H-pyran-4- yl)-1H-pyrazolo[3,4-jb]pyridin-4-amine is quantified via yieldaliser, and an appropriate amount of concentrated hydrochloric acid is added at 55-60 degrees C. The suspension is held for 2 hours at about 50 degrees C, and then cooled to 10 degrees C over 3 hours and held at this temperature for at least 3 hours. The slurry is filtered and washed with methyl-THF and dried at 60 0C to constant weight or batch temperature, to give 5-(aminomethyl)-1 ,6-diethyl-Λ/-(tetrahydro-2H- pyran-4-yl)-1H-pyrazolo[3,4-/b]pyridin-4-amine hydrochloride (Intermediate 11).
Intermediate 11 Detailed Procedure: (can be run in 1.0/10 L equipment)
Note: All or most of this process is carried out under a nitrogen atmosphere.
1. [1 ,6-diethyl-4-(tetrahydro-2/-/-pyran-4-ylamino)-1 /-/-pyrazolo[3,4-ό]pyridin-5- yl]methanol (1wt) is suspended in anisole (6vol) at 20-250C. 2. Benzenesulfonic acid (1 equivalent, 0.52wt) is added.
3. The temperature is adjusted to 20-250C and the reaction is aged for 30 minutes. 4. Thionyl chloride (1.15 equivalents, 0.45wt, 0.275vol) is added over about 20 minutes at 20-250C.
5. Anisole (0.25vol) is used to wash the line.
6. Sample after 20 minutes at 20-250C. Reaction is judged complete when <4% of [1 ,6-diethyl-4-(tetrahydro-2H-pyran-4-ylamino)-1 /-/-pyrazolo[3,4-jb]pyridin-5-yl]methanol remains when compared to the 5-(methoxymethyl)-pyrazolo[3,4-/b]pyridine derivative derived from a methanol quench of 5-(chloromethyl)-1 ,6-diethyl-Λ/-(tetrahydro-2/-/-pyran- 4-yl)-1 H-pyrazolo[3,4-ό]pyridin-4-amine benzenesulfonate.
7. Vessel fill volume is noted, and toluene (2vol) added. 8. The mixture is heated to a jacket temperature of 40 degrees C and held under moderate vacuum (100 mbar) for 1 hour with agitation.
9. Vacuum is increased to about 50 mbar and contents are distilled to marked volume above (about 7.5vol). The Contents should not be heated above 50 degrees C. (Distillation time around 80 minutes, p = 45-50 mbar, Tjn^ = 20-48 0C.)
10. Reaction mixture is cooled to 20-250C and can be held at this point.
11. This mixture (ex-step 10) is added to lithium hexamethyldisilazide in THF (1.26M in THF, 6.51vol, 5.73wt) over 60-70 minutes at 35-40°C.
12. Sample after a further 10 minutes at 35-40°C. Reaction is judged complete if <4% of the 5-(methoxymethyl)-pyrazolo[3,4-jb]pyridine derivative is seen relative to 5-(aminomethyl)-1 ,6-diethyl-Λ/-(tetrahydro-2H-pyran-4-yl)-1 H-pyrazolo[3,4-b]pyridin-4- amine (after quench into methanol) by HPLC.
13. Mixture is cooled to 1O0C and hydrochloric acid (5M, 3.6 vol) is added over 20 minutes. The phases are stirred vigorously for 10 minutes.
14. The lower aqueous layer is retained, and the upper anisole layer discarded. The batch can be held at 20-25 0C at this point.
15. The brown aqueous layer is treated with methyl-tetrahydrofuran (8vol).
16. Sodium hydroxide solution (ca 1.8 vol, 10M) is added to the biphasic mixture to give a resulting aqueous layer with pH >13. The temperature is raised to 3O0C and the mixture is stirred vigorously for 20 minutes (separation time 5 minutes in 10L CLR equipment).
17. The layers are separated and the lower aqueous layer (yellow) is backwashed with methyl-tetrahydrofuran (2vol). The aqueous layer is sampled and analysed for residual 5-(aminomethyl)-1,6-diethyl-Λ/-(tetrahydro-2/-/-pyran-4-yl)-1/-/-pyrazolo[3,4-ό]pyridin-4- amine prior to disposal. 18. Brine (15%w/w NaCI, 2vol) is added to the combined brown organic layers the biphase is vigorously stirred at 30 0C and separated. The lower aqueous layer is removed. 19. The volume of the organic layer is measured and a sample taken. The yieldaliser system is used to determine the absolute amount of 5-(aminomethyl)-1 ,6-diethyl-Λ/- (tetrahydro-2H-pyran-4-yl)-1/-/-pyrazolo[3,4-jb]pyridin-4-amine present.
20. The reaction mixture is heated to 55-600C and treated with concentrated hydrochloric acid [1.03 equivalents relative to the 5-(aminomethyl)-1 ,6-diethyl-Λ/-
(tetrahydro-2H-pyran-4-yl)-1H-pyrazolo[3,4-b]pyridin-4-amine present, about 0.24-0.30 vol].
21. The mixture is aged at 50-550C for 2 hours and then cooled to 5-100C over 3 hours.
22. The mixture is aged at 5-1O0C for at least 3 hours. 23. Mixture is filtered and washed with methyl-THF (2 x 2vol). The mother liquors are kept until a dried weight and yield of product is obtained. (Filtration times all less then 90seconds on a scale of about 70Og.)
24. The off white solid cake is dried in a vacuum oven at 60 0C to constant temperature.
Intermediate 12: Tetrahydro-2H-pyran-4-amine hydrochloride, which is 4-aminotetrahydro-2H-pyran hydrochloride
Figure imgf000079_0001
Stepi: N,N-dibenzyltetrahydro-2H-pyran-4-amine
Dibenzylamine (34.5g) and acetic acid (6.7ml) are added to a stirred solution of tetrahydro-4H-pyran-4-one (16.4g, e.g. commercially available from Aldrich) in dichloromethane (260ml) at 0 °C to 5 0C. After 2.5h at 0 0C to 5 0C, sodium triacetoxyborohydride (38.9g) is added portionwise, and the mixture is allowed to warm to room temperature. After stirring at room temperature overnight, the reaction mixture is washed successively with 2M-sodium hydroxide (200ml and 50ml), water (2 x 50ml) and brine (50ml), then dried and evaporated. The residue (which may be an oil) is stirred with methanol (50ml) at 4 0C for 30min to give the product (e.g. as a solid).
Step 2: Tetrahydro-2H-pyran-4-amine hydrochloride
Λ/,Λ/-dibenzyltetrahydro-2H-pyran-4-amine (20.5g) is dissolved in ethanol (210ml) and hydrogenated over 10% palladium on carbon catalyst (4g) at 100 psi for 72h at room temperature. The reaction mixture is filtered and the filtrate is adjusted to pH 1 with 2M- hydrogen chloride in diethyl ether. Evaporation of solvents gives a residue (which may be a solid) which is triturated with diethyl ether to give the product (which may be a solid). EXAMPLES
Example 1 (Synthesis A) /V-{[1,6-diethyl-4-(tetrahydro-2H-pyran-4-ylamino)-1W- pyrazolo[3,4-Jb]pyridin-5-y|]methyl}-3-methyl-5-isoxazolecarboxamide
Figure imgf000080_0001
An example of a specific synthesis of Example 1 is as follows:
S-Methyl-δ-isoxazolecarboxylic acid (92mg) was suspended in dry dichloromethane (2ml) and treated at 200C with oxalyl chloride (0.064ml) and diethyl formamide (1 drop). Effervescence occurred over ca. 10mins, and after 40mins stirring at room temperature the solution was added dropwise to a solution of 5-(aminomethyl)-1 ,6-diethyl-Λ/- (tetrahydro-2W-pyran-4-yl)-1H-pyrazolo[3,4-jb]pyridin-4-amine (198mg) (e.g. which can be as prepared in Intermediate 10) in anhydrous acetonitrile (4ml). The mixture was treated with DIPEA (0.128ml) and stirred at room temperature under nitrogen for 15h. Dichloromethane (50ml) was added to the reaction mixture and the solution was washed with dilute aqueous sodium chloride solution (2 x 50ml). The organic phase was separated using a hydrophobic frit (70ml) and loaded directly onto an SPE cartridge (10g, aminopropyl) which had been pre-washed with methanol. The cartridge was eluted with methanol (x2) and the fractions collected and blown down / evaporated to dryness. The residual gum was dissolved in dichloromethane and purified by SPE cartridge (10g, silica) on a Flashmaster 2 eluting with a gradient of 0 - 100% ethyl acetate in cyclohexane over 40 mins. Two fractions (from two chromatographic peaks) were collected and evaporated separately, and each was purified by mass directed autoprep HPLC. Relevant fractions from each were evaporated to dryness. The two residual gums (125 mg and 48mg respectively) were combined and purified by SPE cartridge (2g, aminopropyl) which had been pre-washed with methanol. Elution with methanol (x2), collection of methanol, and evaporation to dryness gave the title compound as a white foam (173mg). LCMS showed MH+ = 413; TRET = 2.20min.
Note: S-Methyl-δ-isoxazolecarboxylic acid is thought to be commercially available from one or more of the following suppliers: Chemical Block Stock Library, Chemical Block Building Blocks, Fluorochem, Scientific Exchange, Aurora Screening Library, Oakwood Products Catalog, Ambinter Stock Screening Collection, TimTec Building Blocks and Reagents, TimTec Overseas Stock, Enamine Building Blocks, Enamine Screening Library, lnterchim Intermediates, AsInEx Express Gold Collection, and/or MicroChemistry Building Blocks. See near the start of the "Intermediates and Examples" section hereinabove, for the addresses of some of these suppliers.
Example 1 (Synthesis B)
Figure imgf000081_0001
The carboxylic acid R'COOH (
Figure imgf000081_0002
) (0.12mmol) is treated with a solution of HATU (0.12mmol) in DMF (0.25ml), and DIPEA (0.052ml, ca. 0.3mmol) is added. The solution is shaken for 10mins, and is treated with a solution of 5-(aminomethyl)-1 ,6-diethyl-Λ/- (tetrahydro-2H-pyran-4-yl)-1H-pyrazolo[3,4-6]pyridin-4-amine (O.immol, ca. 30.3mg) (e.g. which can be as prepared in Intermediate 10) in DMF (0.2ml). The resulting solution is shaken for IOmins and left to stand for 18h (e.g. at room temperature), and then the DMF is removed in a Genevac vacuum centrifuge. The residue is dissolved in chloroform (0.3ml), is applied to an SPE cartridge (1g, aminopropyl) which has been pre- washed with chloroform (6ml), and is eluted sequentially with chloroform (3ml) and 10% methanol in ethyl acetate (3ml). Fractions containing the desired product are concentrated in vacuo in a Genevac vacuum centrifuge, and where necessary the residue is purified by mass directed autoprep HPLC (e.g. acetonitrile/water). Where necessary, the compound is dissolved in chloroform, and is further purified by loading onto a SPE cartridge (0.5g, aminopropyl) which has been prewashed with chloroform, eluting with 10% methanol in ethyl acetate.
Example 1 (Synthesis C) /V-{[1,6-diethyl-4-(tetrahydro-2W-pyran-4-ylamino)-1H-pyrazolo[3,4-fo]pyridin-5- yl]methyl}-3-methyl-5-isoxazolecarboxamide
Figure imgf000081_0003
S-Methyl-δ-isoxazolecarboxylic acid (6.59 g, 0.052 mol, 1.0 equivalent), O-(benzotriazol- 1-yl)-Λ/,N,Λr,N'-tetramethyluronium hexafluorophosphate (HBTU) (20.88g, 1.08 equivalents) and diisopropylethylamine (18 ml, 2 equivalents) in dimethyl formamide (DMF, 314ml, 20 volumes) were stirred under nitrogen for 10 minutes. 5-(aminomethyl)- 1 ,6-diethyl-Λ/-(tetrahydro-2H-pyran-4-yl)-1 H-pyrazolo[3,4-ιb]pyridin-4-annine (15.7 g, 0.052 mol, 1.0 equivalent) (e.g. which can be as prepared in Intermediate 10) was added to the mixture with extra DMF (20ml) to wash it into the flask. The reaction mixture was left to stir for three (3) hours, and then left under nitrogen overnight. The reaction mixture was partitioned between saturated sodium hydrogen carbonate solution (3 L) and dicloromethane (800 mL). The aqueous layer was extracted again with dicloromethane (800 mL). The combined organic layers were backwashed with water (2 x 500 mL), dried over MgSC>4 (50 g), filtered, reduced in vacuo, and cooled to give a crude brown oil (about 3Og) which appeared to include DMF.
This crude brown oil was slurried in 1 : 0.98 : 0.2 DCM : EtOAc : MeOH, whereupon the crude product precipitated, was filtered and the filtered solid was washed with diethyl ether (100 ml) to give a damp mass (12 g). The reduced liquors (the reduced slurry liquids remaining after filtration and the diethyl ether washings) were purified by column chromatography (silica, 800 ml), using 1 : 0.98 : 0.2 DCM : EtOAc : MeOH (4L) as eluent, to give a further batch of product (4 g).
Both batches of product were combined (appeared impure by 1H NMR), and were purified by column chromatography (silica, 800 ml), using 3 to 4 % MeOH in DCM (3L) as eluent, to give 13 g of material which still appeared impure (by TLC).
This resulting impure solid (13 g) was slurried in 1 : 0.96 : 0.4 of DCM : EtOAc : MeOH and the solid remaining was separated by filtration and dried, to give the title compound Λ/-{[1 ,6-diethyl-4-(tetrahydro-2H-pyran-4-yIamino)-1 H-pyrazolo[3,4-jb]pyridin- 5-yl]methyl}-3-methyl-5-isoxazolecarboxamide as an off-white solid (9 g). The reduced liquors (the reduced slurry liquids) were purified by column chromatography (silica, 400 ml) using 1 : 0.96 : 0.4 DCM : EtOAc : MeOH (2L) as eluent, giving, after reduction in vacuo, a further batch of product.
The two batches of product were combined to give 10.714 g in total of the title compound A/-{[1 ,6-diethyl-4-(tetrahydro-2H-pyran-4-ylamino)-1 H-pyrazolo[3,4-ιb]pyridin- 5-yl]methyl}-3-methyl-5-isoxazolecarboxamide as an off-white solid (more than about 95% purity as measured by NMR in CDCI3 solvent (not the NMR given below)).
1H NMR (500MHz in d6-DMSO, δ (delta) ppm): 9.38 (br t, 1 H), 8.04 (s, 1H), 7.00
(s, 1 H), 6.64 (d, 1 H), 4.52 (d, 2H), 4.33 (q, 2H), 4.12 (br s, 1H), 3.88 (d, 2H), 3.57 (t, 2H), 2.95 (q, 2H)1 2.29 (s, 3H), 1.93 (d, 2H), 1.54 (q, 2H), 1.35 (t, 3H), 1.24 (t, 3H). Plus trace other peaks: possibly solvent. PHARMACEUTICAL COMPOSITION EXAMPLES
The following are examples of pharmaceutical compositions (formulations) suitable for external topical administration (e.g. topical administration to skin).
Composition Example 1 (Ointment D)
The following pharmaceutical composition (formulation) is an ointment suitable for external topical administration (e.g. topical administration to skin), and comprises 2% w/w of the compound of the invention Λ/-{[1 ,6-diethyl-4-(tetrahydro-2H-pyran-4-ylamino)- 1W-pyrazolo[3,4-d]pyridin-5-yl]methyl}-3-methyl-5-isoxazolecarboxamide (i.e. the "free base" form):
Product: Ointment D
Batch Size: 20 g
Concentration in composition Theoretical Actual
Ingredient (% w/w) Amount (g) Amount (g) white petrolatum 73 14.6 14.69 decamethyl-cyclopentasiloxane (ST-Cyclomethicone 5-NF ™) 25 5.01
compound of the invention (i.e. "free base" form) 0.4 0.4006
TOTAL 20 20.1006
Preparation Procedure:
Approximately the following procedure is used to prepare Ointment D:
The white petrolatum (a solid at room temperature, and e.g. which can optionally be high melting point white petrolatum such as Penreco Ultima White ™ grade white petrolatum) and the decamethyl-cyclopentasiloxane (a liquid at room temperature) are heated together, in a beaker in a hot water bath, to a temperature of approximately 60-65 0C, to melt the white petrolatum.
The drug substance (the compound of the invention, which, for example, can be as prepared in Example 1 , Synthesis C) is added slowly into the melted oil phase and is stirred with a spatula and dispersed completely. The mixture is homogenised under high shear conditions (using setting #5, a high setting, on an Ultra-turrax T25 homogenizer), using the small homogenizer shaft, for 10 minutes. The formulation is allowed to cool to room temperature (e.g. about 17 to about 22 0C) while stirring. The ointment formulation is filled into a 3OmL transparent container.
Reference Composition Example 2 (Placebo Ointment AP)
The following pharmaceutical composition (formulation) is an ointment suitable for external topical administration (e.g. topical administration to skin), but it does not include any compound of the invention. Thus it can be used as a comparator placebo ointment, when testing (e.g. in "In Vivo Assay A" herein) a corresponding ointment containing the compound of the invention such as Composition Example 1 (Ointment D).
Product: Placebo Ointment AP
Batch Size: 100 g
Concentration in composition Theoretical Actual Amount
Ingredient (% w/w) Amount (g) (9) white petrolatum 75 75 75.61 decamethyl-cyclopentasiloxane
(ST-Cyclomethicone 5-NF ™) 25 25 25.00
TOTAL 100 100.61
Preparation Procedure:
Approximately the following procedure is used to prepare Placebo Ointment AP:
The white petrolatum (which can optionally be high melting point white petrolatum such as Penreco Ultima White ™ grade white petrolatum) and the decamethyl- cyclopentasiloxane are heated together, in a small beaker in a hot water bath, to a temperature of approximately 60-65 0C.
The mixture is homogenised under high shear conditions (using setting #5, a high setting, on an Ultra-turrax T25 homogenizer), for approximately 10 minutes.
The formulation is allowed to cool and is stirred until room temperature (e.g. about 17 to about 22 0C) is reached. The ointment formulation is packed into a 125mL transparent plastic container.
Composition Example 3 (Ointment D2)
The following pharmaceutical composition (formulation) is an ointment suitable for external topical administration (e.g. topical administration to skin), and comprises 2% w/w of the compound of the invention /V-{[1 ,6-diethyl-4-(tetrahydro-2W-pyran-4-ylamino)- 1H-pyrazolo[3,4-ιb]pyridin-5-yl]methyl}-3-methyl-5-isoxazolecarboxamide (i.e. the "free base" form):
Product: Ointment D2
Batch Size: 20 g
Concentration in composition Theoretical Actual
Ingredient (% w/w) Amount (g) Amount (g) white petrolatum 73 14.6 14.66 decamethyl-cyclopentasiloxane (ST-Cyclomethicone 5-NF ™) 25 5.09
compound of the invention (i.e. "free base" form) 0.4 0.4012
TOTAL 20 20.1512
Preparation Procedure:
Substantially the same preparation procedure as in Composition Example 1 (Ointment
D) is used to prepare Ointment D2.
Reference Composition Example 4 (Placebo Ointment AP2)
The following pharmaceutical composition (formulation) is an ointment suitable for external topical administration (e.g. topical administration to skin), but it does not include any compound of the invention. Thus it can be used as a comparator placebo ointment, when testing (e.g. in "In Vivo Assay A" herein) a corresponding ointment containing the compound of the invention such as Composition Example 3 (Ointment D2).
Product: Placebo Ointment AP2
Batch Size: 100 g
Concentration in composition Theoretical Actual Amount
Ingredient (% w/w) Amount (g) (9) white petrolatum 75 75 75.52 decamethyl-cyclopentasiloxane (ST-Cyclomethicone 5-NF ™) 25 25 25.03
TOTAL 100 100.55 Preparation Procedure:
Substantially the same preparation procedure as in Reference Composition Example 2
(Placebo Ointment AP) is used to prepare Placebo Ointment AP2.
Composition Example 5 (Water-in-oil cream Cr-D)
The following pharmaceutical composition (formulation) is believed to be a water-in-oil cream emulsion, for external topical administration (e.g. topical administration to skin), and comprises propylene glycol and 2% w/w of the compound of the invention Λ/-{[1 ,6- diethyl-4-(tetrahydro-2H-pyran-4-ylamino)-1H-pyrazolo[3,4-ό]pyridin-5-yl]methyl}-3- methyl-5-isoxazolecarboxamide (i.e. the "free base" form):
Product: Water-in-oil cream Cr-D
Batch Size: 20 g
Concentration in composition Theoretical Actual
Ingredient (% w/w) Amount (g) Amount (g) white petrolatum 40 8 8.05 mineral oil 10 2 2.00 steareth-2 = polyoxyl 2 stearyl ether
(Volpo s-2 ™) (surfactant) 8 1.6 1.62
Aαueous phase propylene glycol 20 4 4.01 purified water 20 4 4.01 compound of the invention (i.e. "free base" form) 2 0.4 0.4001
TOTAL 20 20.0901
Preparation Procedure:
Approximately the following procedure is used to prepare Water-in-oil cream Cr-D:
The white petrolatum (which can optionally be high melting point white petrolatum such as Penreco Ultima White ™ grade white petrolatum), the mineral oil and the steareth-2 (Volpo s-2 ™) are heated together, via a hot water bath, to a temperature of approximately 60-65 0C to form an oily phase.
In a separate container, the drug substance (the compound of the invention, which, for example, can be as prepared in Example 1, Synthesis C) is dispersed in water and sonicated for approximately 10 minutes. Propylene glycol is added to this aqueous phase and the mixture is sonicated for another 10 minutes. The drug substance is at least partly in suspension. The aqueous phase is heated to approximately the same temperature as the oily phase (e.g. is heated to approximately 60-65 0C), and then the aqueous phase is added slowly to the oily phase while homogenizing the mixture under high shear conditions (using setting #5, a high setting, e.g. on an Ultra-turrax T25 homogenizer), for approximately 10 minutes.
After homogenization, the formulation is allowed to cool to room temperature (e.g. about 17 to about 22 0C) with constant mixing with a spatula. The cream formulation is packed into a 2OmL scintillation vial.
Reference Composition Example 6 (Placebo Water-in-oil cream Cr-AP)
The following pharmaceutical composition (formulation) is believed to be a water-in-oil cream emulsion, for external topical administration, but it does not include any compound of the invention. Thus it can be used as a comparator placebo cream, when testing (e.g. in "In Vivo Assay A" herein) a corresponding cream containing the compound of the invention such as Composition Example 5 (Water-in-oil cream Cr-D).
Product: Placebo Water-in-oil cream Cr-AP Batch Size: 100g
Concentration in composition Theoretical Actual Amount
Ingredient (% w/w) Amount (g) (9) white petrolatum 42 42 42.98 mineral oil 10 10 10.06 steareth-2 = polyoxyl 2 stearyl ether (Volpo s-2 ™) (surfactant) 8 8 8.04
Aqueous phase propylene glycol 20 20 20.06 purified water 20 20 20.03
TOTAL 100 101.17
Preparation Procedure:
Approximately the following procedure is used to prepare Placebo Water-in-oil cream
Cr-AP:
The white petrolatum (which can optionally be high melting point white petrolatum such as Penreco Ultima White ™ grade white petrolatum), the mineral oil and the steareth-2 (Volpo s-2 ™) are heated together, via a hot water bath, to a temperature of approximately 60-65 0C to form an oily phase. In another beaker, the aqueous phase (propylene glycol and water) is heated to a temperature of approximately 60-65 °C. The aqueous phase is added slowly to the oily phase while homogenizing the mixture at low speed, and then the speed is increased to high shear conditions (using setting #5, a high setting, e.g. on an Ultra-turrax T25 homogenizer). The mixture is homogenized, while being kept hot via a hot water bath, for approximately 10 minutes.
The formulation is then allowed to cool and stirred until room temperature (e.g. about 17 to about 22 0C) is reached. The cream formulation is packed into a 125ml_ transparent plastic container.
Composition Example 7 (Water-in-oil cream Cr-D2)
The following pharmaceutical composition (formulation) is believed to be a water-in-oil cream emulsion, for external topical administration (e.g. topical administration to skin), and comprises propylene glycol and 2% w/w of the compound of the invention /V-{[1 ,6- diethyl-4-(tetrahydro-2/-/-pyran-4-ylamino)-1H-pyrazolo[3,4-fe]pyridin-5-yl]methyl}-3- methyl-5-isoxazolecarboxamide (i.e. the "free base" form):
Product: Water-in-oil cream Cr-D2
Batch Size: 20 g
Concentration in composition Theoretical Actual
Ingredient (% w/w) Amount (g) Amount (g) white petrolatum 40 8 8.01 mineral oil 10 2 • 2.02 steareth-2 = polyoxyl 2 stearyl ether
(Volpo s-2 ™) (surfactant) 8 1.6 1.64
Aqueous phase propylene glycol 20 4 4.03 purified water 20 4 4.07 compound of the invention (i.e. "free base" form) 2 0.4 0.4004
TOTAL 20 20.1704
Preparation Procedure:
Substantially the same preparation procedure as in Composition Example 5 (Water-in-oil cream Cr-D) is used to prepare Water-in-oil cream Cr-D2, except that the cream formulation is packed into a 3OmL transparent container. Reference Composition Example 8 (Placebo Water-in-oil cream Cr-AP2)
The following pharmaceutical composition (formulation) is believed to be a water-in-oil cream emulsion, for external topical administration, but it does not include any compound of the invention. Thus it can be used as a comparator placebo cream, when testing (e.g. in "In Vivo Assay A" herein) a corresponding cream containing the compound of the invention such as Composition Example 7 (Water-in-oil cream Cr-D2).
Product: Placebo Water-in-oil cream Cr-AP2 Batch Size: 10Og
Concentration in composition Theoretical Actual Amount
Ingredient (% w/w) Amount (g) (9) white petrolatum 42 42 42.71 mineral oil 10 10 10.15 steareth-2 = polyoxyl 2 stearyl ether (Volpo s-2 ™) (surfactant) 8 8 8.01
Aqueous phase propylene glycol 20 20 20.00 purified water 20 20 20.02
TOTAL 100 101.89
Preparation Procedure:
Substantially the same preparation procedure as in Reference Composition Example 6 (Placebo Water-in-oil cream Cr-AP) is used to prepare Placebo Water-in-oil cream Cr-AP2.

Claims

1. Λ/-{[1 ,6-diethyl-4-(tetrahydro-2H-pyran-4-ylamino)-1 H-pyrazolo[3,4-ϋ]pyridin-5- yl]methyl}-3-methyl-5-isoxazolecarboxamide
Figure imgf000090_0001
or a salt thereof.
2. A compound as claimed in claim 1 or a pharmaceutically acceptable salt thereof.
3. Λ/-{[1 ,6-diethyl-4-(tetrahydro-2H-pyran-4-ylamino)-1 H-pyrazolo[3,4-ιb]pyridin-5- yl]methyl}-3-methyl-5-isoxazolecarboxamide
Figure imgf000090_0002
4. A compound or salt as claimed in claim 2, wherein the pharmaceutically acceptable salt comprises a pharmaceutically acceptable acid addition salt.
5. A compound or salt as claimed in claim 2, wherein the pharmaceutically acceptable salt is a pharmaceutically acceptable acid addition salt.
6. A compound or salt as claimed in claim 4 or 5, wherein the pharmaceutically acceptable acid addition salt has been formed by mixing Λ/-{[1 ,6-diethyl-4-(tetrahydro- 2H-pyran-4-ylamino)-1H-pyrazolo[3,4-ib]pyridin-5-yl]methyl}-3-methyl-5- isoxazolecarboxamide with a pharmaceutically acceptable acid having a pKa of 1.5 or less.
7. A compound or salt as claimed in claim 4 or 5, wherein the pharmaceutically acceptable acid addition salt of the Λ/-{[1 ,6-diethyl-4-(tetrahydro-2H-pyran-4-ylamino)- 1 H-pyrazolo[3,4-β]pyridin-5-yl]methyl}-3-methyl-5-isoxazolecarboxamide comprises a hydrobromide, hydrochloride, sulfate, nitrate, phosphate, p-toluenesulfonate, benzenesulfonate, methanesulfonate, ethanesulfonate, or naphthalenesulfonate salt thereof.
8. A compound or salt as claimed in claim 4 or 5, wherein the pharmaceutically acceptable acid addition salt of the Λ/-{[1 ,6-diethyl-4-(tetrahydro-2H-pyran-4-ylamino)- 1 /-/-pyrazolo[3,4-ib]pyridin-5-yl]methyl}-3-methyl-5-isoxazolecarboxamide is a hydrobromide, hydrochloride, sulfate, nitrate, phosphate, p-toluenesulfonate, benzenesulfonate, methanesulfonate, ethanesulfonate, or naphthalenesulfonate salt thereof.
9. A process for preparing a compound of formula (I) or a salt thereof:
Figure imgf000091_0001
wherein R^ is ethyl; R2 is ethyl;
R3 is a heterocyclic group of sub-formula (bb) which is not substituted on a ring carbon:
Figure imgf000091_0002
(bb) in which n^ is 1 ; and in which Y is O; and wherein:
Ra is a hydrogen atom (H); R^ is a hydrogen atom (H); R4 is a hydrogen atom (H); R5 is -C(O)-(C^)n-Ar; wherein n is 0; and Ar has the sub-formula (z) which is sub-formula (z9):
Figure imgf000091_0003
, which process comprises:
(A) reacting an amine of formula (II) or a salt thereof
Figure imgf000092_0001
with a compound Ar-C(O)-X1 , wherein X1 is a leaving group substitutable by the NH2 amine moiety of the compound of formula (II);
and optionally converting the compound of formula (I) into a salt thereof;
or (B), in a process for preparing a pharmaceutically acceptable salt of a compound of formula (I), converting the compound of formula (I) or a salt thereof into a desired pharmaceutically acceptable salt of the compound of formula (I).
10. A process as claimed in claim 9, which is a process for preparing a compound of formula (I) or a pharmaceutically acceptable salt thereof, and wherein:
- in process (A), in the compound Ar-C(O)-X1 , X1 is a chlorine atom (Cl),
Figure imgf000092_0002
or X Λη is \ wherein X2 is CH or N; or X1 is O-C(NHRC1)=NRC2 wherein RC1 and RC2 are independently C-j^alkyl, cyclohexyl or 3-dimethylaminopropyl;
and/or
- in process (B), the process comprises conversion of the compound of formula (I) or a salt thereof into a desired pharmaceutically acceptable acid addition salt of the compound of formula (I).
11. A compound or salt as claimed in any one of claims 1 to 8, for use as an active therapeutic substance in a mammal.
12. A pharmaceutical composition comprising a compound of formula (I), as defined in any of claims 1 to 8, or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable carriers and/or excipients.
13. A pharmaceutical composition as claimed in claim 12, for use in the treatment and/or prophylaxis of an inflammatory and/or allergic disease, cognitive impairment or depression in a mammal.
14. A pharmaceutical composition as claimed in claim 12 or 13, which is suitable for external topical administration to a mammal such as a human.
15. A pharmaceutical composition as claimed in claim 14, which is suitable for topical administration to the skin of a mammal such as a human.
16. A pharmaceutical composition as claimed in claim 14 or 15, which is for the treatment and/or prophylaxis of atopic dermatitis in a mammal such as a human, by external topical administration to the mammal.
17. A pharmaceutical composition as claimed in claim 14, 15 or 16, wherein the compound as defined in claim 1 or the pharmaceutically acceptable salt thereof is present in 0.5% to 10%, by weight of the composition (w/w).
18. A pharmaceutical composition as claimed in claim 17, wherein the compound as defined in claim 1 or the pharmaceutically acceptable salt thereof is present in 1 % to 5%, by weight of the composition (w/w).
19. A pharmaceutical composition as claimed in claim 15, 16, 17 or 18, wherein the compound as defined in claim 1 or the pharmaceutically acceptable salt thereof is N- {[1 ,6-diethyl-4-(tetrahydro-2H-pyran-4-ylamino)-1 /-/-pyrazolo[3,4-ύ]pyridin-5-yl]methyl}-3- methyl-5-isoxazolecarboxamide.
20. A pharmaceutical composition as claimed in any one of claims 14 to 19, which is an ointment.
21. An pharmaceutical composition as claimed in claim 20, being an ointment and comprising:
- the compound as defined in claim 1 or the pharmaceutically acceptable salt thereof present at 0.5% to 10% w/w; - white petrolatum present at 45% to 99.5% w/w (i.e. by weight of the composition); and
- a silicone oil present at 5% to 50% w/w (measured as the total silicone oil content, by weight of the composition)).
22. An pharmaceutical composition as claimed in claim 20 or 21, being an ointment and comprising:
- the compound as defined in claim 1 or the pharmaceutically acceptable salt thereof present at 1 % to 10% w/w;
- white petrolatum present at 45% to 99.5% w/w; and - decamethyl-cyclopentasiloxane present at 5% to 50% w/w.
23. The use of a compound of formula (I), as defined in any of claims 1 to 8, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment and/or prophylaxis of an inflammatory and/or allergic disease in a mammal.
24. The use as claimed in claim 23, wherein the inflammatory and/or allergic disease is an inflammatory and/or allergic skin disease in a mammal such as a human.
25. The use as claimed in claim 23, wherein the inflammatory and/or allergic disease is atopic dermatitis in a mammal such as a human.
26. The use as claimed in claim 23, wherein the inflammatory and/or allergic disease is chronic obstructive pulmonary disease (COPD), asthma, rheumatoid arthritis, allergic rhinitis, psoriasis or atopic dermatitis in a mammal such as a human.
27. The use of a compound of formula (I), as defined in any of claims 1 to 8, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment and/or prophylaxis of asthma, chronic obstructive pulmonary disease (COPD), atopic dermatitis, urticaria, allergic rhinitis, allergic conjunctivitis, vernal conjunctivitis, eosinophilic granuloma, psoriasis, rheumatoid arthritis, septic shock, ulcerative colitis, Crohn's disease, reperfusion injury of the myocardium and brain, chronic glomerulonephritis, endotoxic shock, adult respiratory distress syndrome, multiple sclerosis, cognitive impairment, depression, or inflammatory pain, in a mammal such as a human.
28. A method of treatment and/or prophylaxis of an inflammatory and/or allergic disease, cognitive impairment or depression in a mammal, such as a human, in need thereof, which method comprises administering to the mammal a therapeutically effective amount of a compound of formula (I) as defined in any of claims 1 to 8 or a pharmaceutically acceptable salt thereof.
29. A method as claimed in claim 28, which is a method of treatment and/or prophylaxis of an inflammatory and/or allergic disease in a mammal, such as a human, in need thereof, and wherein the inflammatory and/or allergic disease is chronic obstructive pulmonary disease (COPD), asthma, rheumatoid arthritis, allergic rhinitis, psoriasis or atopic dermatitis in the mammal.
PCT/GB2006/003627 2005-09-29 2006-09-29 Pyrazolo[3,4-b]pyridine compound, and its use as a pde4 inhibitor WO2007036734A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US12/067,994 US20080255186A1 (en) 2005-09-29 2006-09-29 Pyrazolo[3,4-B]Pyridine Compound, and Its Use as a Pde4 Inhibitor
EP06779579A EP1940836A1 (en) 2005-09-29 2006-09-29 Pyrazolo[3,4-b]pyridine compound, and its use as a pde4 inhibitor
JP2008532874A JP2009510044A (en) 2005-09-29 2006-09-29 Pyrazolo [3,4-b] pyridine compounds and their use as PDE4 inhibitors

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US72159705P 2005-09-29 2005-09-29
US60/721,597 2005-09-29

Publications (1)

Publication Number Publication Date
WO2007036734A1 true WO2007036734A1 (en) 2007-04-05

Family

ID=37685723

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/GB2006/003626 WO2007036733A1 (en) 2005-09-29 2006-09-29 Pyrazolo[3,4-b]pyridine compounds, and their use as pde4 inhibitors
PCT/GB2006/003627 WO2007036734A1 (en) 2005-09-29 2006-09-29 Pyrazolo[3,4-b]pyridine compound, and its use as a pde4 inhibitor

Family Applications Before (1)

Application Number Title Priority Date Filing Date
PCT/GB2006/003626 WO2007036733A1 (en) 2005-09-29 2006-09-29 Pyrazolo[3,4-b]pyridine compounds, and their use as pde4 inhibitors

Country Status (7)

Country Link
US (2) US20080255186A1 (en)
EP (2) EP1940836A1 (en)
JP (2) JP5323484B2 (en)
AT (1) ATE503756T1 (en)
DE (1) DE602006021044D1 (en)
ES (1) ES2363795T3 (en)
WO (2) WO2007036733A1 (en)

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008015416A1 (en) 2006-08-01 2008-02-07 Glaxo Group Limited Pyrazolo[3,4-b]pyridine compounds, and their use as pde4 inhibitors
WO2010097248A1 (en) 2009-01-13 2010-09-02 Glaxo Group Limited Pyrimidinecarboxamide derivatives as inhibitors of syk kinase
US9266886B2 (en) 2014-02-03 2016-02-23 Vitae Pharmaceuticals, Inc. Dihydropyrrolopyridine inhibitors of ROR-gamma
US9481674B1 (en) 2016-06-10 2016-11-01 Vitae Pharmaceuticals, Inc. Dihydropyrrolopyridine inhibitors of ROR-gamma
US9663515B2 (en) 2014-11-05 2017-05-30 Vitae Pharmaceuticals, Inc. Dihydropyrrolopyridine inhibitors of ROR-gamma
US9796710B2 (en) 2014-10-14 2017-10-24 Vitae Pharmaceuticals, Inc. Dihydropyrrolopyridine inhibitors of ROR-gamma
US9845308B2 (en) 2014-11-05 2017-12-19 Vitae Pharmaceuticals, Inc. Isoindoline inhibitors of ROR-gamma
US10301261B2 (en) 2015-08-05 2019-05-28 Vitae Pharmaceuticals, Llc Substituted indoles as modulators of ROR-gamma
US10829481B2 (en) 2016-01-29 2020-11-10 Vitae Pharmaceuticals, Llc Benzimidazole derivatives as modulators of ROR-gamma
US10913739B2 (en) 2017-07-24 2021-02-09 Vitae Pharmaceuticals, LLC (121374) Inhibitors of RORγ
US11008340B2 (en) 2015-11-20 2021-05-18 Vitae Pharmaceuticals, Llc Modulators of ROR-gamma
US11186573B2 (en) 2017-07-24 2021-11-30 Vitae Pharmaceuticals, Llc Inhibitors of ROR gamma

Families Citing this family (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ATE503756T1 (en) * 2005-09-29 2011-04-15 Glaxo Group Ltd PYRAZOLOA3,4-BUPYRIDINE COMPOUNDS AND THEIR USE AS PDE4 INHIBITORS
GB0614570D0 (en) * 2006-07-21 2006-08-30 Glaxo Group Ltd Compounds
EP2124944B1 (en) * 2007-03-14 2012-02-15 Ranbaxy Laboratories Limited Pyrazolo[3,4-b]pyridine derivatives as phosphodiesterase inhibitors
CN101687810A (en) 2007-04-26 2010-03-31 武田药品工业株式会社 Bicyclic compound and pharmaceutical use thereof
WO2009100169A1 (en) * 2008-02-06 2009-08-13 Glaxo Group Limited Dual pharmacophores - pde4-muscarinic antagonistics
WO2009100170A1 (en) * 2008-02-06 2009-08-13 Glaxo Group Limited Dual pharmacophores - pde4-muscarinic antagonistics
WO2010046791A1 (en) * 2008-09-19 2010-04-29 Ranbaxy Laboratories Limited Phosphodiestarase inhibitors
BR112012022868A2 (en) * 2010-03-16 2018-06-05 Merck Patent Gmbh morpholinylquinazolines
US8487102B2 (en) * 2010-04-20 2013-07-16 Hoffmann-La Roche Inc. Pyrrazolopyridine compounds as dual NK1/NK3 receptor antagonists
US20140113898A1 (en) * 2010-11-08 2014-04-24 Zalicus Pharmaceuticals Ltd. Bisarylsulfone and dialkylarylsulfone compounds as calcium channel blockers
EP2574607A1 (en) * 2011-09-06 2013-04-03 F. Hoffmann-La Roche AG PDE10 modulators
CA2903572A1 (en) * 2013-03-15 2014-10-23 Epizyme, Inc. Substituted benzene compounds
US9243001B2 (en) 2013-03-15 2016-01-26 Epizyme, Inc. Substituted benzene compounds
US9011934B2 (en) 2013-06-14 2015-04-21 SatisPharma, LLC Multi-purpose anti-itch treatment
DE102014009164A1 (en) * 2014-06-03 2015-12-03 Herbalist & Doc Gesundheitsgesellschaft Mbh Silicone oil containing nasal formulations
US9452178B1 (en) 2014-10-22 2016-09-27 SatisPharma, LLC Acne formulations, treatments, and pressure sensitive patches for delivery of acne formulations
WO2018114657A1 (en) 2016-12-20 2018-06-28 Syngenta Participations Ag N-cyclobutyl-thiazol-5-carboxamides with nematicidal activity
US11098010B2 (en) 2017-03-21 2021-08-24 Arbutus Biopharma Corporation Substituted dihydroindene-4-carboxamides and analogs thereof, and methods using same
AR112346A1 (en) * 2017-08-10 2019-10-16 Lilly Co Eli DERIVATIVES OF [1,2,4] TRIAZOLO
US20210031012A1 (en) 2018-01-26 2021-02-04 Progenity, Inc. Treatment of a disease of the gastrointestinal tract with a pde4 inhibitor
WO2020106757A1 (en) 2018-11-19 2020-05-28 Progenity, Inc. Ingestible device for delivery of therapeutic agent to the gastrointestinal tract
US11707610B2 (en) 2019-12-13 2023-07-25 Biora Therapeutics, Inc. Ingestible device for delivery of therapeutic agent to the gastrointestinal tract

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004024728A2 (en) * 2002-09-16 2004-03-25 Glaxo Group Limited Pyrazolo[3,4-b]pyridine compounds, and their use as phosphodiesterase inhibitors
WO2004056823A1 (en) * 2002-12-23 2004-07-08 Glaxo Group Limited PYRAZOLO[3,4-b]PYRIDINE COMPOUNDS, AND THEIR USE AS PHOSPHODIESTERASE INHIBITORS

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ATE503756T1 (en) * 2005-09-29 2011-04-15 Glaxo Group Ltd PYRAZOLOA3,4-BUPYRIDINE COMPOUNDS AND THEIR USE AS PDE4 INHIBITORS

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004024728A2 (en) * 2002-09-16 2004-03-25 Glaxo Group Limited Pyrazolo[3,4-b]pyridine compounds, and their use as phosphodiesterase inhibitors
WO2004056823A1 (en) * 2002-12-23 2004-07-08 Glaxo Group Limited PYRAZOLO[3,4-b]PYRIDINE COMPOUNDS, AND THEIR USE AS PHOSPHODIESTERASE INHIBITORS

Cited By (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008015416A1 (en) 2006-08-01 2008-02-07 Glaxo Group Limited Pyrazolo[3,4-b]pyridine compounds, and their use as pde4 inhibitors
WO2010097248A1 (en) 2009-01-13 2010-09-02 Glaxo Group Limited Pyrimidinecarboxamide derivatives as inhibitors of syk kinase
US10047085B2 (en) 2014-02-03 2018-08-14 Vitae Pharmaceuticals, Inc. Dihydropyrrolopyridine inhibitors of ROR-gamma
US9266886B2 (en) 2014-02-03 2016-02-23 Vitae Pharmaceuticals, Inc. Dihydropyrrolopyridine inhibitors of ROR-gamma
US11535614B2 (en) 2014-02-03 2022-12-27 Vitae Pharmaceuticals, Llc Dihydropyrrolopyridine inhibitors of ROR-gamma
US9624217B2 (en) 2014-02-03 2017-04-18 Vitae Pharmaceuticals, Inc. Dihydropyrrolopyridine inhibitors of ROR-gamma
US10807980B2 (en) 2014-02-03 2020-10-20 Vitae Pharmaceuticals, Llc Dihydropyrrolopyridine inhibitors of ROR-gamma
US10399976B2 (en) 2014-02-03 2019-09-03 Vitae Pharmaceuticals, Llc Dihydropyrrolopyridine inhibitors of ROR-gamma
US9796710B2 (en) 2014-10-14 2017-10-24 Vitae Pharmaceuticals, Inc. Dihydropyrrolopyridine inhibitors of ROR-gamma
US10087184B2 (en) 2014-10-14 2018-10-02 Vitae Pharmaceuticals, Inc. Dihydropyrrolopyridine inhibitors of RORγ
US9845308B2 (en) 2014-11-05 2017-12-19 Vitae Pharmaceuticals, Inc. Isoindoline inhibitors of ROR-gamma
US11001583B2 (en) 2014-11-05 2021-05-11 Vitae Pharmaceuticals, Llc Dihydropyrrolopyridine inhibitors of ROR-gamma
US9663515B2 (en) 2014-11-05 2017-05-30 Vitae Pharmaceuticals, Inc. Dihydropyrrolopyridine inhibitors of ROR-gamma
US10301261B2 (en) 2015-08-05 2019-05-28 Vitae Pharmaceuticals, Llc Substituted indoles as modulators of ROR-gamma
US10829448B2 (en) 2015-08-05 2020-11-10 Vitae Pharmaceuticals, Llc Substituted benzoimidazoles as modulators of ROR-γ
US11008340B2 (en) 2015-11-20 2021-05-18 Vitae Pharmaceuticals, Llc Modulators of ROR-gamma
US10829481B2 (en) 2016-01-29 2020-11-10 Vitae Pharmaceuticals, Llc Benzimidazole derivatives as modulators of ROR-gamma
US9481674B1 (en) 2016-06-10 2016-11-01 Vitae Pharmaceuticals, Inc. Dihydropyrrolopyridine inhibitors of ROR-gamma
US10913739B2 (en) 2017-07-24 2021-02-09 Vitae Pharmaceuticals, LLC (121374) Inhibitors of RORγ
US11186573B2 (en) 2017-07-24 2021-11-30 Vitae Pharmaceuticals, Llc Inhibitors of ROR gamma

Also Published As

Publication number Publication date
DE602006021044D1 (en) 2011-05-12
EP1940835B1 (en) 2011-03-30
JP2009510043A (en) 2009-03-12
EP1940835A1 (en) 2008-07-09
WO2007036733A1 (en) 2007-04-05
JP2009510044A (en) 2009-03-12
ATE503756T1 (en) 2011-04-15
US20090131431A1 (en) 2009-05-21
EP1940836A1 (en) 2008-07-09
ES2363795T3 (en) 2011-08-16
JP5323484B2 (en) 2013-10-23
US20080255186A1 (en) 2008-10-16

Similar Documents

Publication Publication Date Title
WO2007036734A1 (en) Pyrazolo[3,4-b]pyridine compound, and its use as a pde4 inhibitor
US7709497B2 (en) Pyrazolo[3,4-b]pyridine compound, and its use as a PDE4 inhibitor
JP4872068B2 (en) Pyrazolo [3,4-b] pyridine compounds and their use as phosphodiesterase inhibitors
US20070167485A1 (en) Pyrazolo[3,4-b]pyridine compounds, and their use as pde4 inhibitors
JP2007514704A (en) Pyrazolo [3,4-b] pyridine compounds and their use as phosphodiesterase inhibitors
EP1751151A1 (en) Pyrazolo &#39;3,4-b! pyridine compounds, and their use as phosphodiesterase type 4 (pde4) inhibitors
US20090318494A1 (en) Pyrazolo[3,4-b]pyridine compounds, and their use as pde4 inhibitors
US20090326003A1 (en) Pyrazolo (3, 4-b) pyridine derivatives as pde4 inhibitors
ES2362445T3 (en) PYRAZOLO [3,4-B] PYRIDINE COMPOUNDS AND THEIR USE AS PDE4 INHIBITORS.
MXPA06010486A (en) Pyrazolo[3,4-b]pyridine compound, and its use as a pde4 inhibitor
EP1737857A1 (en) Pyrazolo[3,4-b] pyridine compounds, and their use as phosphodiesterase inhibitors

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 12067994

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 2008532874

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2006779579

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2006779579

Country of ref document: EP