WO2002067909A1 - New method of bronchodilatory therapy - Google Patents

New method of bronchodilatory therapy Download PDF

Info

Publication number
WO2002067909A1
WO2002067909A1 PCT/GB2002/000809 GB0200809W WO02067909A1 WO 2002067909 A1 WO2002067909 A1 WO 2002067909A1 GB 0200809 W GB0200809 W GB 0200809W WO 02067909 A1 WO02067909 A1 WO 02067909A1
Authority
WO
WIPO (PCT)
Prior art keywords
selective
alkyl
agonist
human patient
receptor agonist
Prior art date
Application number
PCT/GB2002/000809
Other languages
French (fr)
Inventor
Giampietro Ventresca
Original Assignee
Glaxo Group Limited
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Glaxo Group Limited filed Critical Glaxo Group Limited
Publication of WO2002067909A1 publication Critical patent/WO2002067909A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • A61K31/52Purines, e.g. adenine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7076Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines containing purines, e.g. adenosine, adenylic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/08Bronchodilators

Definitions

  • This invention relates to a novel method of bronchodilatory therapy and to the novel use of a class of molecules in the manufacture of a medicament for the treatment of patients having need of bronchodilation.
  • Inflammation is a primary response to tissue injury or microbial invasion and is characterised by leukocyte adhesion to the endothelium, diapedesis and activation within the tissue.
  • Leukocyte activation can result in the generation of toxic oxygen species (such as superoxide anion), and the release of granule products (such as peroxidases and proteases).
  • Circulating leukocytes include neutrophils, eosinophils, basophils, monocytes and lymphocytes.
  • Different forms of inflammation involve different types of infiltrating leukocytes, the particular profile being regulated by the profile of adhesion molecule, cytokine and chemotactic factor expression within the tissue.
  • Adenosine agonists may also down- regulate other classes of leucocytes (Elliot and Leonard, 1989; Peachell et al, 1989). Studies on whole animals have shown the anti-inflammatory effects of methotrexate to be mediated through adenosine and A2 receptor activation (Asako et al, 1993; Cronstein et al, 1993 and 1994). Adenosine itself, and compounds that raise circulating levels of adenosine also show anti- inflammatory effects in vivo (Green et al, 1991; Rosengren et al, 1995). A2 receptor agonists have also been shown to have effects on capsaicin sensitive afferent sensory nerves in airways tissue (Marimoto et al). A number of A2a receptor agonists have been described in the literature - for example:
  • A2a agonists having claimed selective activity are described in WO00/23457, WO00/77018, WO01/94368 and WO02/00676 (Pfizer).
  • Glaxo and Pfizer A2a receptor agonists are all derivatives of adenosine which are derivatised in respect of modified substitution the purine ring and/or the ribose moiety.
  • A2a receptor agonists have also been described in WO00/78776, WO00/78777, WO00/78778 and WO00/78779 (CV Therapeutics) and in WO00/72799 and US patent 5,877,180 (University of Virginia Foundation).
  • adenosine derivatives in the treatment of inflammatory conditions of the respiratory tract is the fact that adenosine has been shown to elicit dose-dependent bronchoconstriction in allergic and non allergic asthmatics (Cushley et al). Adenosine monophosphate has been reported to exhibit similar effects in atopic non-asthmatics (Phillips et al).
  • Step 1 For the mildest asthmatics (“Step 1”), the prescribed therapy is occasional use of relief bronchodilators.
  • the only licensed bronchodilators are the short acting beta-2 agonists (eg albuterol, terbutaline) which are specifically recommended for these mild patients or long acting beta-2 agonists (eg salmeterol, formoterol) which are recommended for more severe patients.
  • beta-2 agonists eg albuterol, terbutaline
  • beta-2 agonists eg salmeterol, formoterol
  • Step 2-3 it is first recommended to add an inhaled anti-inflammatory agent such as a corticosteroid (such as fluticasone propionate, beclomethasone dipropionate or budesonide) or a mast cell stabiliser such as sodium cromoglycate or nedocromil sodium.
  • a corticosteroid such as fluticasone propionate, beclomethasone dipropionate or budesonide
  • Steps 4-5 Use of long acting beta-2 agonists or oral theophylline may also be appropriate in more severe patients within this category.
  • asthmatics For yet more severe asthmatics (“Steps 4-5") it is recommended to use high doses of inhaled steroids together with one or more of: long acting beta-2 agonists, oral theophylline, inhaled ipratropium, mast cell stabilisers finally adding oral prednisolone for the most severe patients.
  • bronchodilatory therapy which may be used regularly (as in the case of the long- acting beta-2 agonists) or on-demand in the case of crisis (eg as in the case of short acting beta-2 agonists).
  • beta-2 agonists show evidence of anti-inflammatory effects in vitro and in animal models, there remains controversy as to whether or not anti-inflammatory effects of such agents are exhibited in man. Furthermore, the commonly used anti-inflammatory medicaments are believed to have no bronchodilatory activity.
  • selective A2a receptor agonists cause bronchodilation in asthmatic patients.
  • selective A2a agonists show anti-inflammatory effects in animal models and are expected to show anti-inflammatory effects in human subjects.
  • selective A2a agonists appear to fall into a novel and useful class of molecules which combine a bronchodilator effect with an anti-inflammatory effect and will be especially useful in the treatment of inflammatory airways disease such as asthma and COPD.
  • a method of inducing bronchodilation in a human patient which comprises administering to that patient an effective amount of a selective A2a receptor agonist.
  • a method of inducing bronchodilation in a human patient suffering from an inflammatory disorder eg asthma or COPD
  • an inflammatory disorder eg asthma or COPD
  • a method of relieving bronchoconstriction in a human patient which comprises administering to that patient an effective amount of a selective A2a receptor agonist.
  • a method of relieving bronchoconstriction in a human patient suffering from an inflammatory disorder eg asthma or COPD
  • an inflammatory disorder eg asthma or COPD
  • a pharmaceutical composition for the inducing of bronchodilation in a human patient which comprises an effective amount of a selective A2a receptor agonist.
  • a pharmaceutical composition for inducing bronchodilation in a human patient suffering from an inflammatory disorder eg asthma or COPD
  • an inflammatory disorder eg asthma or COPD
  • a pharmaceutical composition for the relief of bronchoconstriction in a human patient which comprises an effective amount of a selective A2a receptor agonist.
  • a pharmaceutical composition for the relief of bronchoconstriction in a human patient suffering from an inflammatory disorder eg asthma or COPD
  • an inflammatory disorder eg asthma or COPD
  • a selective A2a receptor agonist in the manufacture of a medicament for the induction of bronchodilation in a human patient.
  • a selective A2a receptor agonist in the manufacture of a medicament for the induction of bronchodilation in a human patient suffering from an inflammatory disorder (eg asthma or COPD).
  • a selective A2a receptor agonist in the manufacture of a medicament for the relief of bronchoconstriction in a human patient.
  • a selective A2a receptor agonist in the manufacture of a medicament for the relief of bronchoconstriction in a human patient suffering from an inflammatory disorder (eg asthma or COPD).
  • the bronchodilatory relief will advantageously be acute (i.e. rapid onset) relief as well as chronic relief.
  • Selective A2a receptor agonists will generally act at the A2a receptor with an agonist potency which is at least 2 times (preferably 5, especially at least 10 times) that at which they act at the A1 , A2b and A3 receptors when measured in assay systems expressing or containing comparable levels of each receptor.
  • Selective A2a receptor agonists will generally have an affinity for the A2a receptor which is at least 2 times (preferably 5, especially at least 10 times) greater than that for the A1 , A2b and A3 receptors. However A2a agonists with high affinity for A1, A2b or A3 receptors may still behave as selective A2a agonists if they have affinity but little or no agonist activity at these other adenosine receptors (eg if they are antagonists of some or all of them).
  • the selective A2a receptor agonist will also demonstrate A3 antagonist activity.
  • Agonist activity of selective A2a agonists eg at A2a, A1 and A3 receptors may be determined by reference to the following method:
  • Agonist selectivity of compounds against other human adenosine receptors may be determined using Chinese hamster ovary (CHO) cells transfected with the gene for the relevant human adenosine receptor following a method based on that of Castanon and Spevak, 1994 .
  • the CHO cells are also transfected with cyclic AMP response elements promoting the gene for secreted placental alkaline phosphatase (SPAP) (Wood, 1995).
  • test compounds may be determined by their effects on basal levels of cAMP (A2a and A2b) or on forskolin-enhanced cAMP (A1 and A3) as reflected by changes in levels of SPAP.
  • EC 50 values for compounds may then be determined as a ratio to that of the non-selective agonist N-ethyl carboxamide adenosine (NECA).
  • NECA non-selective agonist N-ethyl carboxamide adenosine
  • Affinity for receptor sub-types may be determined following the methods of Klotz et al (A1 and A2a ), Robeva et al (A2b) and Varani et al (A3).
  • antagonist affinity may be determined from a competition experiment involving use of a known sub-type selective agonist ligand.
  • a known A3 selective agonist is IB-MECA (iodobenzylmethylcarboximidoadenosine).
  • the selective A2a receptor agonist is a derivative of adenosine.
  • Derivatives include compounds which have modified functionality on the purine and/or the ribose moieties of adenosine.
  • a first set of preferred selective A2a receptor agonists is defined by compounds of formula (I) as follows: wherein R 1 and R 2 independently represent a group selected from:
  • Z 2 represents C or N
  • R 3 represents C ⁇ alkyl or cyclopropyl, save that where Z 2 represents C, R 3 may also represent CH 2 OH.
  • R 4 and R 5 independently represent hydrogen, C ⁇ alkyl, aryl, arylC,. 6 alkyl- or
  • NR 4 R 5 together may represent pyridinyl, pyrrolidinyl, piperidinyl, morpholinyl, azetidinyl, azepinyl, piperazinyl or N-C ⁇ alkylpiperazinyl;
  • R 6 represents OH, NH 2 , NHCOCH 3 or halogen
  • R 7 represents hydrogen, C ⁇ alkyl, -C.,. 6 alkylaryl or -COC L ealkyl;
  • X represents NR 7 , O, S, SO or SO 2 ;
  • p represents 0 or 1 ;
  • a and b independently represent an integer 0 to 4 provided that a + b is in the range 3 to 5;
  • c, d and e independently represent an integer 0 to 3 provided that c + d + e is in the range 2 to 3;
  • Z Z 3 and Z 4 will independently represent C, N, O or S and, in the case of C and N, together with a sufficient number of hydrogen atoms to provide the ring with aromatic character. At least one of Z 1 , Z 2 , Z 3 , Z 4 and Z 5 will represent a heteroatom. Preferably at least one of Z ⁇ Z 3 and Z 4 will represent a nitrogen atom. More preferably at least one of Z ⁇ Z 3 and Z 4 will represent a nitrogen atom and at least one of the remainder will represent C or N. We prefer that two or three of Z ⁇ Z 2 , Z 3 and Z 4 are heteroatoms.
  • references to C x . y alkyl include references to an aliphatic hydrocarbon grouping containing x to y carbon atoms which may be straight chain or branched and may be saturated or unsaturated. References to alkoxy may also be interpreted similarly.
  • references to aryl include references to mono- and bicyclic carbocyclic aromatic rings (e.g. phenyl, naphthyl) and heterocyclic aromatic rings, for example containing 1-3 hetero atoms selected from N, O and S (e.g. pyridinyl, pyrimidinyl, thiophenyl, imidazolyl, quinolinyl, furanyl, pyrrolyl, oxazolyl) all of which may be optionally substituted, e.g. by C ⁇ alkyl, halogen, hydroxy, nitro, C,. 6 alkoxy, cyano, amino, SO 2 NH 2 or -CH 2 OH.
  • mono- and bicyclic carbocyclic aromatic rings e.g. phenyl, naphthyl
  • heterocyclic aromatic rings for example containing 1-3 hetero atoms selected from N, O and S (e.g. pyridinyl, pyrimidinyl, thiophenyl, imi
  • C 3 . 8 cycloalkyl for R 1 and R 2 include monocyclic alkyl groups (e.g. cyclopentyl, cyclohexyl) and bicyclic alkyl groups (e.g. norbornyl such as exo- norborn-2-yl).
  • Examples of (aryl) 2 CHCH 2 - for R 1 and R 2 include Ph 2 CHCH 2 - or such a group in which one or both phenyl moieties is substituted, e.g. by halogen or C ⁇ alkyl.
  • Examples of C 3 . 8 cycloalkylC 1 . 6 alkyl- for R 1 and R 2 include ethylcyclohexyl.
  • Examples of C,. 8 alkyl for R 1 and R 2 include -(CH 2 ) 2 C(Me) 3 , -CH(Et) 2 and
  • CH 2 C(Me)CH 2 CH 2 -.
  • Examples of arylC,. 6 alkyl- for R 1 and R 2 include -(CH 2 ) 2 Ph, -CH 2 Ph or either in which Ph is substituted (one or more times) by halogen (e.g. iodine), amino, methoxy, hydroxy, -CH 2 OH or SO 2 NH 2 ; -(CH 2 ) 2 pyridinyl (e.g. -(CH 2 ) 2 pyridin-2-yl) optionally substituted by amino; (CH 2 ) 2 imidazolyl (e.g. 1 H-imidazol-4-yl) or this group in which imidazoyl is N-substituted by C,. 6 alkyl (especially methyl).
  • halogen e.g. iodine
  • amino methoxy, hydroxy, -CH 2 OH or SO 2 NH 2
  • -(CH 2 ) 2 pyridinyl e.g. -(CH 2 )
  • R 4 R 5 N-C 1 ⁇ alkyl- for R 1 and R 2 examples include ethyl-piperidin-1-yl, ethyl- pyrrolidin-1-yl, ethyl-morpholin-1-yl, -(CH 2 ) 2 NH(pyridin-2-yl) and -(CH ⁇ NK,.
  • Examples of C ⁇ alkyl-CH C ⁇ OH)- for R 1 and R 2 include Me 2 CHCH(CH 2 OH)-.
  • Examples of arylC ⁇ alkyl-CHfCH H)- for R 1 and R 2 include PhCH 2 CH(CH 2 OH)- particularly
  • R 1 and R 2 examples include PhCH 2 C(CH 2 OH) 2 -.
  • Examples of C 3 . 8 cycloalkyl independently substituted by one or more -(CH 2 ) P R 6 groups (eg 1 , 2 or 3 such groups) for R 1 and R 2 include 2-hydroxy-cyclopentyl and 4-aminocyclohexyl (especially trans-4-amino-cyclohexyl).
  • R 1 and R 2 include pyrrolidin-3-yl, piperidin-3-yl, piperidin-4-yl, tetrahydro-1,1- dioxide thiophen-3-yl, tetrahydropyran-4-yl, tetrahydrothiopyran-4-yl and 1 ,1- dioxo-hexahydro-1.lamda.6-thiopyran-4-yl, or a derivative in which the ring nitrogen is substituted by C h alky! (e.g. methyl), C ⁇ alkylacyl (e.g. acetyl), arylC,. 6 alkyl- (e.g. benzyl).
  • C h alky! e.g. methyl
  • C ⁇ alkylacyl e.g. acetyl
  • arylC arylC,. 6 alkyl- (e.g. benzyl).
  • Examples of -C ⁇ alkyl-OH groups for R 1 and R 2 include -CH 2 CH 2 OH and -CH(CH 2 OH)CH(CH 3 ) 2 .
  • Examples of C,. 8 haloalkyl for R 1 and R 2 include -CH 2 CH 2 CI and (CH 3 ) 2 CIC(CH 2 ) 3 -. Examples of groups of formula
  • R 1 and R 2 include 2-oxopyrrolidin-4-yl, 2-oxopyrrolidin-3-yl, 2-oxopyrrolidin-5- yl or a derivative in which the ring nitrogen is substituted by C ⁇ alkyl (e.g. methyl) or benzyl.
  • aryl for R 1 and R 2 include phenyl optionally substituted by halogen
  • R 1 and R 2 is -(CH 2 ) 2 SO 2 NHCH 2 Ph.
  • C ⁇ alkyl for R 7 is methyl
  • an example of C ⁇ alkylaryl for R 7 is benzyl
  • an example of -COC,. 6 alkyl for R 7 is acetyl.
  • R 1 and R 2 do not both represent hydrogen.
  • R 1 to represent aryl 2 CHCH 2 -, C h alky!-, hydrogen or arylC ⁇ alky
  • R 2 to represent arylC L galkyl-CHfCH H)-, R 4 R 5 N-C 1 . 6 alkyl-,
  • R 3 to represent methyl, ethyl, n-propyl, isopropyl, cyclopropyl or CH 2 OH (when Z 2 represents C), particularly methyl, ethyl or cyclopropyl, especially ethyl.
  • R 4 and R 5 independently to represent hydrogen or aryl or NR 4 R 5 together to represent pyrrolidinyl, piperidinyl, morpholinyl, azetidinyl, azepinyl, piperazinyl or N-methylpiperazinyl.
  • R 6 represents OH or NH 2 .
  • a represents 2 and that b represents 1 or 2.
  • X to represent NR 7 (e.g. NH), O, S or SO 2 , particularly O, S or NH.
  • c represents 0, and either that d represents 1 and e represents 1 or d represents 0 and e represents 2.
  • R 7 represents hydrogen.
  • R 1 to represent Ph 2 CHCH 2 -, hydrogen or CH(Et) 2 , especially hydrogen or CH(Et) 2 .
  • Group (a) is especially preferred; group (c) is also particularly preferred.
  • Z 2 to represent C.
  • Z 4 to represent N.
  • the groups above illustrated as (i) triazolyl, (ii) 4'- 1 ,2,4 oxadiazolyl, (iii) 4'-1,3,4 oxadiazolyl and (vi) N-alkyl triazolyl are preferred.
  • the group above illustrated as (i) triazolyl is most preferred.
  • Z ⁇ Z 2 , Z 3 and Z 4 together with the carbon atom form a 5-membered heterocyclic aromatic ring and variables R 1 and R 2 are as previously defined (Z ⁇ Z 2 , Z 3 and Z 4 will independently represent C, N, O or S and, in the case of C and N, together with a sufficient number of hydrogen atoms to provide the ring with aromatic character. At least one of Z Z 2 , Z 3 and Z 4 will represent a heteroatom. Preferably at least one of Z ⁇ Z 2 , Z 3 and Z 4 will represent a nitrogen atom. More preferably at least one of Z ⁇ Z 2 , Z 3 and Z 4 will represent a nitrogen atom, two of the remainder independently will represent C or N and the fourth will represent C, N or O. In each case sufficient hydrogen atoms will be provided to give the ring aromatic character. However, we prefer that Z ⁇ Z 2 , Z 3 and Z 4 do not all represent nitrogen); and
  • Z ⁇ Z 2 , Z 3 and Z 4 together with Z 5 form a 5-membered heterocyclic aromatic ring and variables R 1 and R 2 are as previously defined (Z ⁇ Z 2 , Z 3 and Z 4 will independently represent C, N, O or S and, in the case of C and N, together with a sufficient number of hydrogen atoms to provide the ring with aromatic character. At least one of Z ⁇ Z 2 , Z 3 , and Z 4 will represent carbon. We prefer that one or two of Z ⁇ Z 2 , Z 3 , and Z 4 represent N and the remainder represent C. Carbon atoms in the ring may be substituted by hydroxy).
  • R 8 is alkyl or cyclopropylmethyl
  • R 9 is phenyl-alkylene or napthyl-alkylene, said alkylene chain being optionally further substituted by phenyl or napthyl, each phenyl or napthyl being optionally substituted by one or more substituents each independently selected from alkyl, alkoxy, halo and cyano; n is 1 or 2;
  • A is NR a , NR a C(O), NR a C(O)NR a , NR a C(O)O, OC(O)NR a , C(O)NR a , NR a SO 2 , SO 2 NR a , O, S or SO 2 ,
  • R a is H, alkyl or benzyl optionally ring-substituted by one or more substituents each independently selected from alkyl, alkoxy, halo and cyano;
  • R 10 is a group of the formula -(CH 2 ) q -R p -B; q is O, 1 or 2; R p is a bond, alkylene, cycloalkylene, phenylene or naphthylene, said cycloalkylene, phenylene and naphthylene each being optionally substituted by one or more substituents each independently selected from alkyl, alkoxy, halo and alkoxyalkylene; B is (i) H, -NR b R b , R b R b N-alkylene, -OR b , -COOR b , -OCOR b , -SO 2 R b , -CN, - SO 2 NR R b , -NR b COR b , NR b SO 2 R b or -CONR b R b , in which each R b is the same or different and is selected form H, alkyl, phenyl and
  • R p is a bond, p is 0 and B is H only when A is NR a , NR a C(O)NR a , OC(O)NR a , C(O)NR a -, SO 2 NR a , O or S, (ii) an optionally-substituted, fully- or partially-saturated or-unsaturated, mono-or bicyclic, heterocyclic group, which is linked to R p by a ring carbon atom, or (iii) N-linked azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl or morpholinyl, each optionally substituted by one or more alkyl substituents, with the proviso that -(CH 2 ) p -R D -is not-CH 2 -; and where A is NR a , C(O)NR a , OC(O)NR a or SO 2 NR a
  • R 11 is hydrogen or C,-C 6 alkyl optionally substituted by 1 or 2 substituents each independently selected from phenyl and naphthyl, said phenyl and naphthyl being optionally substituted by C,-C 6 alkyl, C r C 6 alkoxy, halo or cyano;
  • R 12 is H or C r C 6 alkyl;
  • K is C C 6 alkylene;
  • R 13 is (i) hydrogen, C C ⁇ alkyl, -COOR 14 , -CN, -CONR 14 R 14 , C 3 -C 8 cycloalkyl, phenyl or naphthyl, said C 3 -C 8 cycloalkyl, phenyl and naphthyl being optionally substituted by C r C 6 alkyl, phenyl, C r C 6 alkoxy (C r C 6 ) alkyl, R 1 R N(C r C 6 ) alkyl, halo (C r C 6 ) alkyl, fluoro (C r C 6 ) alkoxy, (C 2 -C 5 ) alkanoyl, halo, -OR 14 , cyano, -COOR 14 , C 3 -C 8 cycloalkyl, -S(O) m R 15 , -NR 14 R 14 , -SO 2 NR 14 R 14 , - CONR
  • R 14 is H, C r C 6 alkyl, C 3 -C 8 cycloalkyl or phenyl;
  • R 15 is C r C 6 alkyl, C 3 -C 8 cycloalkyl or phenyl
  • R 16 is H, C r C 6 alkyl, C 3 -C 8 cycloalkyl, phenyl, naphthyl or het;
  • R 17 is C r C 6 alkyl, C 3 -C 8 cycloalkyl, phenyl, naphthyl or het; m is 0, 1 or 2; and "het", used in the definitions of R 16 and R 17 , means C-linked pyrrolyl, imidazoyl, triazolyl, thienyl, furyl, thiazolyl, oxazolyl, thiadiazolyl, oxadiazolyl, pyridinyl, pyrimidinyl, pyridazinyl, pyrazinyl, indolyl, isoindolyl, quinolinyl, isoquinolinyl, benzimidazolyl, quinazolinyl, phthalazinyl, benzoxazoyl or quinoxalinyl, each being optionally substituted by C r C 6 alkyl, C C 6 alkoxy, cyano or halo.
  • the previously mentioned compounds may all be used in the form of physiologically acceptable salts if desired eg in order to optimise the biological or physical properties of the molecule.
  • the representation of formulae (I), (II), (III), (IV), (V) and (VI) indicate the absolute stereochemistry. When sidechains contain chiral centres the invention extends to mixtures of enantiomers (including racemic mixtures) and diastereoisomers as well as individual enantiomers. Generally it is preferred to use a compound of formula (I) in the form of a purified single enantiomer.
  • the selective A2a receptor agonist may beneficially be combined in therapy with one or more further therapeutic agents, for example:
  • Short acting beta-2 agonists eg albuterol (eg as free base or sulphate) or terbutaline (eg as sulphate);
  • beta-2 agonists eg salmeterol (eg as xinafoate) or formoterol (eg as fumarate);
  • Glucocorticosteroids eg fluticasone propionate, beclomethasone dipropionate, mometasone furoate, ciclesonide, budesonide, rofleponide, triamcinolone acetonide;
  • Anti-cholinergic medicaments eg ipratropium (eg as bromide), oxitropium, tiotropium;
  • Non-steroidal anti-inflammatory and/or anti-allergic compounds eg ketotifen, cromoglycate (eg as sodium), nedocromil (eg as sodium);
  • DA2-beta-2 agonist compounds eg 4-hydroxy-7-[2-[[2-[[3-(2- phenylethoxy)propyl]sulfonyl]ethyl]amino]ethyl-2(3H)-benzothiazolone; PDE4 inhibitors, such as cilomilast.
  • any of the above mentioned medicaments may be used in alternative salt forms and, where appropriate, in the form of enantiomeric mixtures (eg racemates) or single enantiomers.
  • Combinations of medicaments in therapy may either be administered sequentially or (preferably) simultaneously.
  • the A2a agonist according to the invention may be formulated for administration in any convenient way, and the invention therefore also includes within its scope pharmaceutical compositions for use in anti-inflammatory therapy, comprising a compound of formula (I) or a physiologically acceptable salt or solvate thereof together, if desirable, with one or more physiologically acceptable diluents or carriers.
  • the compounds according to the invention may, for example, be formulated for oral, buccal, parenteral, topical, ocular or rectal administration, preferably for oral, parenteral or topical (e.g. by aerosol) administration.
  • Tablets and capsules for oral administration may contain conventional excipients such as binding agents, for example syrup, acacia, gelatin, sorbitol, tragacanth, mucilage of starch, cellulose or polyvinyl pyrrolidone; fillers, for example, lactose, microcrystalline cellulose, sugar, maize- starch, calcium phosphate or sorbitol; lubricants, for example, magnesium stearate, stearic acid, talc, polyethylene glycol or silica; disintegrants, for example, potato starch, croscarmellose sodium or sodium starch glycollate; or wetting agents such as sodium lauryl sulphate.
  • the tablets may be coated according to methods well known in the art.
  • Oral liquid preparations may be in the form of, for example, aqueous or oily suspensions, solutions, emulsions, syrups or elixirs, or may be presented as a dry product for constitution with water or other suitable vehicle before use.
  • Such liquid preparations may contain conventional additives such as suspending agents, for example, sorbitol syrup, methyl cellulose, glucose/sugar syrup, gelatin, hydroxymethyl cellulose, carboxymethyl cellulose, aluminium stearate gel or hydrogenated edible fats; emulsifying agents, for example, lecithin, sorbitan mono-oleate or acacia; non-aqueous vehicles (which may include edible oils), for example almond oil, fractionated coconut oil, oily esters, propylene glycol or ethyl alcohol; or preservatives, for example, methyl or propyl 2- hydroxy benzoates or sorbic acid.
  • the preparations may also contain buffer salts, flavouring, colouring and/or sweetening agents (
  • the compounds may also be formulated as suppositories, e.g. containing conventional suppository bases such as cocoa butter or other glycerides.
  • Compounds according to the invention may also be formulated for parenteral administration by bolus injection or continuous infusion and may be presented in unit dose form, for instance as ampoules, vials, small volume infusions or pre- filled syringes, or in multi-dose containers with an added preservative.
  • the compositions may take such forms as solutions, suspensions, or emulsions in aqueous or non-aqueous vehicles, and may contain formulatory agents such as anti-oxidants, buffers, antimicrobial agents and/or tonicity adjusting agents.
  • the active ingredient may be in powder form for constitution with a suitable vehicle, e.g. sterile, pyrogen-free water, before use.
  • the dry solid presentation may be prepared by filling a sterile powder aseptically into individual sterile containers or by filling a sterile solution aseptically into each container and freeze-drying.
  • topical administration as used herein, we include administration by insufflation and inhalation.
  • preparation for topical administration include ointments, creams, lotions, powders, pessaries, sprays, aerosols, capsules or cartridges for use in an inhaler or insufflator, solutions for nebulisation or drops (e.g. eye or nose drops).
  • Ointments and creams may, for example, be formulated with an aqueous or oily base with the addition of suitable thickening and/or gelling agents and/or solvents.
  • bases may thus, for example, include water and/or an oil such as liquid paraffin or a vegetable oil such as arachis oil or castor oil or a solvent such as a polyethylene glycol.
  • Thickening agents which may be used include soft paraffin, aluminium stearate, cetostearyl alcohol, polyethylene glycols, microcrystalline wax and beeswax.
  • Lotions may be formulated with an aqueous or oily base and will in general also contain one or more emulsifying agents, stabilising agents, dispersing agents, suspending agents or thickening agents.
  • Powders for external application may be formed with the aid of any suitable powder base, for example, talc, lactose or starch. Drops may be formulated with an aqueous or non-aqueous base also comprising one or more dispersing agents, solubilising agents or suspending agents.
  • Spray compositions may be formulated, for example, as aqueous solutions or suspensions or as aerosols delivered from pressurised packs, with the use of a suitable propellant, e.g. dichlorodifluoromethane, trichlorofluoromethane, dichlorotetra-fluoroethane, 1 ,1 ,1 ,2,3,3,3-heptafluoropropane, 1 ,1 ,1,2- tetrafluoroethane, carbon dioxide or other suitable gas.
  • a suitable propellant e.g. dichlorodifluoromethane, trichlorofluoromethane, dichlorotetra-fluoroethane, 1 ,1 ,1 ,2,3,3,3-heptafluoropropane, 1 ,1 ,1,2- tetrafluoroethane, carbon dioxide or other suitable gas.
  • Intranasal sprays may be formulated with aqueous or non-aqueous vehicles with the addition of agents such as thickening agents, buffer salts or acid or alkali to adjust the pH, isotonicity adjusting agents or anti-oxidants.
  • agents such as thickening agents, buffer salts or acid or alkali to adjust the pH, isotonicity adjusting agents or anti-oxidants.
  • Capsules and cartridges of for example gelatin, or blisters of for example laminated aluminium foil, for use in an inhaler or insufflator may be formulated containing a powder mix of a compound of the invention and a suitable powder base such as lactose or starch.
  • Solutions for inhalation by nebulisation may be formulated with an aqueous vehicle with the addition of agents such as acid or alkali, buffer salts, isotonicity adjusting agents or antimicrobials. They may be sterilised by filtration or heating in an autoclave, or presented as a non-sterile product.
  • Selective A2a agonist compounds may conveniently be administered in amounts of, for example, 0.001 ⁇ g/kg to 50mg/kg body weight, preferably 0.01 ⁇ g/kg to 10mg/kg body weight, 1 to 4 times daily or on demand.
  • the precise dose will of course depend on the age and condition of the patient, the activity of the molecule selected and the particular route of administration chosen.
  • the preferred route of administration is topical administration to the lung (eg administration by inhalation).
  • a preferred A2a agonist according to the invention is (2R,3R,4S,5R)-2-[6-Amino- 2-(1S-hydroxymethyl-2-phenyl-ethylamino)-purin-9-yl]-5-(2-ethyl-2H-tetrazol-5- yl)-tetrahydro-furan-3,4-diol and salts thereof (eg the maleate) which may be prepared according to the method of Example 11 in W098/28319.
  • a preferable dose of this compound is 0.06-125 ⁇ g administered by inhalation each day.
  • Another compound of interest is:
  • WRC0474 SHA211
  • WRC0470 which may be prepared as described in WO00/72799 and references contained therein.
  • the bronchodilatory medicaments according to the invention may be useful in the treatment of diseases of the respiratory tract which include actual or a susceptibility to bronchoconstriction or wherein bronchodilation would be beneficial.
  • the principal such diseases are asthma and chronic obstructive pulmonary disease (COPD).
  • COPD chronic obstructive pulmonary disease
  • Other diseases for which this therapy may be suitable include adult respiratory distress syndrome (ARDS), emphysema, cystic fibrosis and atopic/allergic diseases having a bronchial component eg allergic rhinitis.
  • ARDS adult respiratory distress syndrome
  • emphysema cystic fibrosis
  • atopic/allergic diseases having a bronchial component eg allergic rhinitis.
  • the bronchodilatory medicaments according to the invention may also be useful in the treatment of cough.
  • Figure 1 Mean FEV1 change (in litres) from baseline with time in Cohort 1 following the experiment of the Example.
  • Figure 2 Mean FEV1 change (in litres) from baseline with time in Cohort 2 following the experiment of the Example.
  • Dry powder placebo consisted of a Diskhaler blister containing lactose.
  • Dry powder drug formulation consisted of a Diskhaler blister containing 25 ⁇ g drug per blister and lactose.
  • C 5 doses of matched placebo via nebuliser
  • B A single high dose of 75 ⁇ g of drug , then 2 doses each of 25 ⁇ g of drug and then 2 doses each of placebo via Diskhaler inhaler (total daily dose received

Abstract

As a first aspect of the invention we provide a method of inducing bronchodilation in a human patient which comprises administering to that patient an effective amount of a selective A2a receptor agonist.

Description

New method of bronchodilatory therapy
This invention relates to a novel method of bronchodilatory therapy and to the novel use of a class of molecules in the manufacture of a medicament for the treatment of patients having need of bronchodilation.
Inflammation is a primary response to tissue injury or microbial invasion and is characterised by leukocyte adhesion to the endothelium, diapedesis and activation within the tissue. Leukocyte activation can result in the generation of toxic oxygen species (such as superoxide anion), and the release of granule products (such as peroxidases and proteases). Circulating leukocytes include neutrophils, eosinophils, basophils, monocytes and lymphocytes. Different forms of inflammation involve different types of infiltrating leukocytes, the particular profile being regulated by the profile of adhesion molecule, cytokine and chemotactic factor expression within the tissue.
There is evidence from both in vitro and in vivo studies to suggest that compounds active at the adenosine A2a receptor will have anti-inflammatory actions. The area has been reviewed by Cronstein (1994). Studies on isolated neutrophils show an A2 receptor-mediated inhibition of superoxide generation, degranulation, aggregation and adherence (Cronstein et al, 1983 and 1985; Burkey and Webster, 1993; Richter, 1992; Skubitz et al, 1988). When agents selective for the A2a receptor over the A2b receptor (eg CGS21680) have been used, the profile of inhibition appears consistent with an action on the A2a receptor subtype (Dianzani et al, 1994). Adenosine agonists may also down- regulate other classes of leucocytes (Elliot and Leonard, 1989; Peachell et al, 1989). Studies on whole animals have shown the anti-inflammatory effects of methotrexate to be mediated through adenosine and A2 receptor activation (Asako et al, 1993; Cronstein et al, 1993 and 1994). Adenosine itself, and compounds that raise circulating levels of adenosine also show anti- inflammatory effects in vivo (Green et al, 1991; Rosengren et al, 1995). A2 receptor agonists have also been shown to have effects on capsaicin sensitive afferent sensory nerves in airways tissue (Marimoto et al). A number of A2a receptor agonists have been described in the literature - for example:
Certain substituted 4'-carboxamido and 4'-thioamido adenosine derivatives which are useful for the treatment of inflammatory diseases are described in International Patent Application Nos. WO94/17090, WO96/02553, WO96/02543 (Glaxo Group).
Selective A2a agonists are described in WO98/28319, WO99/38877, WO99/41267, WO99/67263, WO99/67264, WO99/67265 and WO99/67266 (all of Glaxo Group).
Other A2a agonists having claimed selective activity are described in WO00/23457, WO00/77018, WO01/94368 and WO02/00676 (Pfizer).
These Glaxo and Pfizer A2a receptor agonists are all derivatives of adenosine which are derivatised in respect of modified substitution the purine ring and/or the ribose moiety.
A2a receptor agonists have also been described in WO00/78776, WO00/78777, WO00/78778 and WO00/78779 (CV Therapeutics) and in WO00/72799 and US patent 5,877,180 (University of Virginia Foundation).
A concern with the use of adenosine derivatives in the treatment of inflammatory conditions of the respiratory tract is the fact that adenosine has been shown to elicit dose-dependent bronchoconstriction in allergic and non allergic asthmatics (Cushley et al). Adenosine monophosphate has been reported to exhibit similar effects in atopic non-asthmatics (Phillips et al). The explanation for the bronchoconstriction effect of adenosine is not known, although it has been variously hypothesised that it is mediated via the adenosine A3 receptor (Kohno et al, 1996), the A1 receptor (Ali et al and Ghai et al) and A2 receptors (Gustafsson et al). Asthmatic-type responses have also been reported as being mediated via the A2b receptor (Feoktistov et al). Standard therapies for chronic asthmatics are set out in international guidelines (see eg British Thoracic Society et al Thorax (1993) 48 (suppl 1) S1-24). For the mildest asthmatics ("Step 1"), the prescribed therapy is occasional use of relief bronchodilators. The only licensed bronchodilators are the short acting beta-2 agonists (eg albuterol, terbutaline) which are specifically recommended for these mild patients or long acting beta-2 agonists (eg salmeterol, formoterol) which are recommended for more severe patients. With increased severity ("Steps 2-3") it is first recommended to add an inhaled anti-inflammatory agent such as a corticosteroid (such as fluticasone propionate, beclomethasone dipropionate or budesonide) or a mast cell stabiliser such as sodium cromoglycate or nedocromil sodium. Use of long acting beta-2 agonists or oral theophylline may also be appropriate in more severe patients within this category. For yet more severe asthmatics ("Steps 4-5") it is recommended to use high doses of inhaled steroids together with one or more of: long acting beta-2 agonists, oral theophylline, inhaled ipratropium, mast cell stabilisers finally adding oral prednisolone for the most severe patients.
In summary, standard therapy for all but the mildest patients includes an anti- inflammatory component which will generally have no effect for immediate relief of the symptoms of asthma. Asthmatic patients therefore additionally receive bronchodilatory therapy which may be used regularly (as in the case of the long- acting beta-2 agonists) or on-demand in the case of crisis (eg as in the case of short acting beta-2 agonists).
Whilst beta-2 agonists show evidence of anti-inflammatory effects in vitro and in animal models, there remains controversy as to whether or not anti-inflammatory effects of such agents are exhibited in man. Furthermore, the commonly used anti-inflammatory medicaments are believed to have no bronchodilatory activity.
Surprisingly we have discovered a class of compounds that has bronchodilatory activity and is also expected to possess anti-inflammatory activity. More specifically, we have discovered that contrary to expectations, selective A2a receptor agonists cause bronchodilation in asthmatic patients. As just noted, selective A2a agonists show anti-inflammatory effects in animal models and are expected to show anti-inflammatory effects in human subjects. Thus selective A2a agonists appear to fall into a novel and useful class of molecules which combine a bronchodilator effect with an anti-inflammatory effect and will be especially useful in the treatment of inflammatory airways disease such as asthma and COPD.
Thus as a first aspect of the invention we provide a method of inducing bronchodilation in a human patient which comprises administering to that patient an effective amount of a selective A2a receptor agonist. As a subsidiary aspect we provide a method of inducing bronchodilation in a human patient suffering from an inflammatory disorder (eg asthma or COPD) which comprises administering to that patient an effective amount of a selective A2a receptor agonist.
We also provide a method of relieving bronchoconstriction in a human patient which comprises administering to that patient an effective amount of a selective A2a receptor agonist. As a subsidiary aspect we provide a method of relieving bronchoconstriction in a human patient suffering from an inflammatory disorder (eg asthma or COPD) which comprises administering to that patient an effective amount of a selective A2a receptor agonist.
There is also provided a pharmaceutical composition for the inducing of bronchodilation in a human patient which comprises an effective amount of a selective A2a receptor agonist. As a subsidiary aspect we provide a pharmaceutical composition for inducing bronchodilation in a human patient suffering from an inflammatory disorder (eg asthma or COPD) which comprises an effective amount of a selective A2a receptor agonist.
We also provide a pharmaceutical composition for the relief of bronchoconstriction in a human patient which comprises an effective amount of a selective A2a receptor agonist. As a subsidiary aspect we provide a pharmaceutical composition for the relief of bronchoconstriction in a human patient suffering from an inflammatory disorder (eg asthma or COPD) which comprises an effective amount of a selective A2a receptor agonist. There is also provided the use of a selective A2a receptor agonist in the manufacture of a medicament for the induction of bronchodilation in a human patient. As a subsidiary aspect we provide the use of a selective A2a receptor agonist in the manufacture of a medicament for the induction of bronchodilation in a human patient suffering from an inflammatory disorder (eg asthma or COPD).
We also provide the use of a selective A2a receptor agonist in the manufacture of a medicament for the relief of bronchoconstriction in a human patient. As a subsidiary aspect we provide the use of a selective A2a receptor agonist in the manufacture of a medicament for the relief of bronchoconstriction in a human patient suffering from an inflammatory disorder (eg asthma or COPD).
The bronchodilatory relief will advantageously be acute (i.e. rapid onset) relief as well as chronic relief.
Selective A2a receptor agonists will generally act at the A2a receptor with an agonist potency which is at least 2 times (preferably 5, especially at least 10 times) that at which they act at the A1 , A2b and A3 receptors when measured in assay systems expressing or containing comparable levels of each receptor.
Selective A2a receptor agonists will generally have an affinity for the A2a receptor which is at least 2 times (preferably 5, especially at least 10 times) greater than that for the A1 , A2b and A3 receptors. However A2a agonists with high affinity for A1, A2b or A3 receptors may still behave as selective A2a agonists if they have affinity but little or no agonist activity at these other adenosine receptors (eg if they are antagonists of some or all of them).
Preferably the selective A2a receptor agonist will also demonstrate A3 antagonist activity.
Agonist activity of selective A2a agonists eg at A2a, A1 and A3 receptors may be determined by reference to the following method: Agonist selectivity of compounds against other human adenosine receptors may be determined using Chinese hamster ovary (CHO) cells transfected with the gene for the relevant human adenosine receptor following a method based on that of Castanon and Spevak, 1994 . The CHO cells are also transfected with cyclic AMP response elements promoting the gene for secreted placental alkaline phosphatase (SPAP) (Wood, 1995). The effect of test compounds may be determined by their effects on basal levels of cAMP (A2a and A2b) or on forskolin-enhanced cAMP (A1 and A3) as reflected by changes in levels of SPAP. EC50 values for compounds may then be determined as a ratio to that of the non-selective agonist N-ethyl carboxamide adenosine (NECA).
A "no effect" result in the above test is evidence either of no affinity for the receptor or of antagonistic activity.
Affinity for receptor sub-types may be determined following the methods of Klotz et al (A1 and A2a ), Robeva et al (A2b) and Varani et al (A3).
In the case of compounds exhibiting sub-type receptor binding, antagonist affinity may be determined from a competition experiment involving use of a known sub-type selective agonist ligand. For example a known A3 selective agonist is IB-MECA (iodobenzylmethylcarboximidoadenosine).
Preferably the selective A2a receptor agonist is a derivative of adenosine. Derivatives include compounds which have modified functionality on the purine and/or the ribose moieties of adenosine.
A first set of preferred selective A2a receptor agonists is defined by compounds of formula (I) as follows:
Figure imgf000009_0001
wherein R1 and R2 independently represent a group selected from:
(i) C3.8cycloalkyl-;
(ϋ) hydrogen;
(iii) aryl2CHCH2-;
(iv) C^cycloalkylC^alkyl-;
(v) C^alkyl-;
(vi) arylC,_6alkyl-;
(vϋ) R^N-C^alkyl-;
Figure imgf000009_0002
(ix) arylC1.5alkyl-CH(CH2OH)-;
(x) arylC1.5alkyl-C(CH2OH)2-;
(xi) C3.8cycloalkyl independenl
-(CH2)PR6 groups;
(xii) H2NC(=NH)NHC1.6 ,alkyl-;
( (xxiiiiii)) aa ggrroouupp ooff ffoorrmmuullaa
Figure imgf000009_0003
or such a group in which one methylene carbon atom adjacent to X, or both if such exist, is substituted by methyl; (xiv) -C^alkyl-OH;
(xv) -C^haloalkyl; (xvi) a group of formula
Figure imgf000009_0004
(xvii) aryl; and (xviii) -(CH2)fSO2NHg(C^alkyl-)2.g or -(CH2)fS02NHg(arylCMalkyl-) where f is
2 or 3 and g is an integer 0 to 2;
Z2 represents C or N;
Z1, Z3 and Z4 together with Z2 and the carbon atom form a 5-membered heterocyclic aromatic ring;
R3 represents C^alkyl or cyclopropyl, save that where Z2 represents C, R3 may also represent CH2OH.
R4 and R5 independently represent hydrogen, C^alkyl, aryl, arylC,.6alkyl- or
NR4R5 together may represent pyridinyl, pyrrolidinyl, piperidinyl, morpholinyl, azetidinyl, azepinyl, piperazinyl or N-C^alkylpiperazinyl;
R6 represents OH, NH2, NHCOCH3 or halogen;
R7 represents hydrogen, C^alkyl, -C.,.6alkylaryl or -COCLealkyl;
X represents NR7, O, S, SO or SO2; p represents 0 or 1 ; a and b independently represent an integer 0 to 4 provided that a + b is in the range 3 to 5; c, d and e independently represent an integer 0 to 3 provided that c + d + e is in the range 2 to 3;
Z Z3 and Z4 will independently represent C, N, O or S and, in the case of C and N, together with a sufficient number of hydrogen atoms to provide the ring with aromatic character. At least one of Z1, Z2, Z3, Z4 and Z5 will represent a heteroatom. Preferably at least one of Z\ Z3 and Z4 will represent a nitrogen atom. More preferably at least one of Z\ Z3and Z4will represent a nitrogen atom and at least one of the remainder will represent C or N. We prefer that two or three of Z\ Z2, Z3 and Z4 are heteroatoms.
References to Cx.yalkyl include references to an aliphatic hydrocarbon grouping containing x to y carbon atoms which may be straight chain or branched and may be saturated or unsaturated. References to alkoxy may also be interpreted similarly.
References to aryl include references to mono- and bicyclic carbocyclic aromatic rings (e.g. phenyl, naphthyl) and heterocyclic aromatic rings, for example containing 1-3 hetero atoms selected from N, O and S (e.g. pyridinyl, pyrimidinyl, thiophenyl, imidazolyl, quinolinyl, furanyl, pyrrolyl, oxazolyl) all of which may be optionally substituted, e.g. by C^alkyl, halogen, hydroxy, nitro, C,. 6alkoxy, cyano, amino, SO2NH2 or -CH2OH.
Examples of C3.8cycloalkyl for R1 and R2 include monocyclic alkyl groups (e.g. cyclopentyl, cyclohexyl) and bicyclic alkyl groups (e.g. norbornyl such as exo- norborn-2-yl).
Examples of (aryl)2CHCH2- for R1 and R2 include Ph2CHCH2- or such a group in which one or both phenyl moieties is substituted, e.g. by halogen or C^alkyl.
Examples of C3.8cycloalkylC1.6alkyl- for R1 and R2 include ethylcyclohexyl. Examples of C,.8alkyl for R1 and R2 include -(CH2)2C(Me)3, -CH(Et)2 and
CH2=C(Me)CH2CH2-.
Examples of arylC,.6alkyl- for R1 and R2 include -(CH2)2Ph, -CH2Ph or either in which Ph is substituted (one or more times) by halogen (e.g. iodine), amino, methoxy, hydroxy, -CH2OH or SO2NH2; -(CH2)2 pyridinyl (e.g. -(CH2)2pyridin-2-yl) optionally substituted by amino; (CH2)2imidazolyl (e.g. 1 H-imidazol-4-yl) or this group in which imidazoyl is N-substituted by C,.6alkyl (especially methyl).
Examples of R4R5N-C1^alkyl- for R1 and R2 include ethyl-piperidin-1-yl, ethyl- pyrrolidin-1-yl, ethyl-morpholin-1-yl, -(CH2)2NH(pyridin-2-yl) and -(CH^NK,.
Examples of C^alkyl-CH C^OH)- for R1 and R2 include Me2CHCH(CH2OH)-. Examples of arylC^alkyl-CHfCH H)- for R1 and R2 include PhCH2CH(CH2OH)- particularly
Figure imgf000011_0001
Examples of
Figure imgf000011_0002
for R1 and R2 include PhCH2C(CH2OH)2-.
Examples of C3.8 cycloalkyl independently substituted by one or more -(CH2)PR6 groups (eg 1 , 2 or 3 such groups) for R1 and R2 include 2-hydroxy-cyclopentyl and 4-aminocyclohexyl (especially trans-4-amino-cyclohexyl).
Examples of H2NC(=NH)NHC1.6alkyl for R1 and R2 include H2NC(=NH)NH(CH2)2-
Examples of groups of formula
,(CH2
-< X
/
(CH2 2)'b for R1 and R2 include pyrrolidin-3-yl, piperidin-3-yl, piperidin-4-yl, tetrahydro-1,1- dioxide thiophen-3-yl, tetrahydropyran-4-yl, tetrahydrothiopyran-4-yl and 1 ,1- dioxo-hexahydro-1.lamda.6-thiopyran-4-yl, or a derivative in which the ring nitrogen is substituted by Chalky! (e.g. methyl), C^alkylacyl (e.g. acetyl), arylC,. 6alkyl- (e.g. benzyl).
Examples of -C^alkyl-OH groups for R1 and R2 include -CH2CH2OH and -CH(CH2OH)CH(CH3)2.
Examples of C,.8haloalkyl for R1 and R2 include -CH2CH2CI and (CH3)2CIC(CH2)3-. Examples of groups of formula
Figure imgf000012_0001
for R1 and R2 include 2-oxopyrrolidin-4-yl, 2-oxopyrrolidin-3-yl, 2-oxopyrrolidin-5- yl or a derivative in which the ring nitrogen is substituted by C^alkyl (e.g. methyl) or benzyl. Examples of aryl for R1 and R2 include phenyl optionally substituted by halogen
(e.g. fluorine, especially 4-fluorine).
An example of a -(CH^SO^Hg^^alky z^ group f°r R1 and R2is
-(CH2)2SO2NHMe, and an example of a -(CH2)fSO2NHg(arylC1^alkyl)2.g group for
R1 and R2is -(CH2)2SO2NHCH2Ph.
An example of C^alkyl for R7 is methyl, an example of C^alkylaryl for R7 is benzyl, and an example of -COC,.6alkyl for R7 is acetyl.
We prefer that R1 and R2 do not both represent hydrogen. We prefer R1 to represent aryl2CHCH2-, Chalky!-, hydrogen or arylC^alky
We prefer R2 to represent arylCLgalkyl-CHfCH H)-, R4R5N-C1.6alkyl-,
-CH(CH2OH)C1.3alkyl, 4-aminocyclohexyl, pyrrolidinyl or arylCH2CH2-, especially where aryl represents (l-C^alkyl-IH-imidazoyl-4-yl).
We prefer R3 to represent methyl, ethyl, n-propyl, isopropyl, cyclopropyl or CH2OH (when Z2 represents C), particularly methyl, ethyl or cyclopropyl, especially ethyl. We prefer R4 and R5 independently to represent hydrogen or aryl or NR4R5 together to represent pyrrolidinyl, piperidinyl, morpholinyl, azetidinyl, azepinyl, piperazinyl or N-methylpiperazinyl.
We prefer that p represents 0. We prefer that R6 represents OH or NH2. We prefer that a represents 2 and that b represents 1 or 2. We prefer X to represent NR7 (e.g. NH), O, S or SO2, particularly O, S or NH. We prefer that c represents 0, and either that d represents 1 and e represents 1 or d represents 0 and e represents 2. We prefer that R7 represents hydrogen.
We particularly prefer R1 to represent Ph2CHCH2-, hydrogen or CH(Et)2, especially hydrogen or CH(Et)2.
We particularly prefer R2 to represent ethyl-piperidin-1-yl, PhCH2CH(CH2OH)-, - CH(CH2OH)(CH(CH3)2, trans-4-amino-cyclohexyl, 2-(1-methyl-1 H-imidazoyl-4- yl)CH2CH2-, ethyl-morpholin-1-yl, pyrrolidin-3-yl, ethyl-pyridin-2-yl, H2NC(=NH)NH(CH2)2-, cyclopentyl or ethylcyclohexyl, especially PhCH2CH(CH2OH)-, 2-(1-methyl-1H-imidazoyl-4-yl)CH2CH2- or ethyl-piperidin-1- yi-
In a first subset of compounds we prefer that the moiety
Figure imgf000013_0001
Group (a) is especially preferred; group (c) is also particularly preferred. In a second subset of compounds we prefer Z2 to represent C. We prefer Z4 to represent N.
We prefer that the moiety
Figure imgf000014_0001
represents one of the following groups:
Figure imgf000014_0002
The above groups may be referred to hereinafter as (i) = triazolyl; (ii) = 4'-1 ,2,4 oxadiazolyl; (iii) = 4'-1,3,4 oxadiazolyl; (iv) = 1 ,3 oxazolyl; (v) = 1 ,3,4 thiadiazolyl; and (vi) = N-alkyl triazolyl. The groups above illustrated as (i) triazolyl, (ii) 4'- 1 ,2,4 oxadiazolyl, (iii) 4'-1,3,4 oxadiazolyl and (vi) N-alkyl triazolyl are preferred. The group above illustrated as (i) triazolyl is most preferred.
Other selective A2a agonist compounds may be defined by the following formulae (II) and (III):
Figure imgf000015_0001
wherein Z\ Z2, Z3 and Z4 together with the carbon atom form a 5-membered heterocyclic aromatic ring and variables R1 and R2 are as previously defined (Z\ Z2, Z3 and Z4will independently represent C, N, O or S and, in the case of C and N, together with a sufficient number of hydrogen atoms to provide the ring with aromatic character. At least one of Z Z2, Z3 and Z4 will represent a heteroatom. Preferably at least one of Z\ Z2, Z3 and Z4 will represent a nitrogen atom. More preferably at least one of Z\ Z2, Z3 and Z4 will represent a nitrogen atom, two of the remainder independently will represent C or N and the fourth will represent C, N or O. In each case sufficient hydrogen atoms will be provided to give the ring aromatic character. However, we prefer that Z\ Z2, Z3 and Z4 do not all represent nitrogen); and
Figure imgf000015_0002
wherein Z\ Z2, Z3 and Z4 together with Z5 form a 5-membered heterocyclic aromatic ring and variables R1 and R2 are as previously defined (Z\ Z2, Z3 and Z4 will independently represent C, N, O or S and, in the case of C and N, together with a sufficient number of hydrogen atoms to provide the ring with aromatic character. At least one of Z\ Z2, Z3, and Z4 will represent carbon. We prefer that one or two of Z\ Z2, Z3, and Z4 represent N and the remainder represent C. Carbon atoms in the ring may be substituted by hydroxy).
Compounds of formula (I), (II) and (III) may be prepared following the methods of WO98/28319, WO99/38877, WO99/41267, WO99/67263, WO99/67264, WO99/67265 and WO99/67266 (all of Glaxo Group).
Other selective A2a agonist compounds may be defined by the following formula (IV):
Figure imgf000016_0001
wherein:
R8 is alkyl or cyclopropylmethyl;
R9 is phenyl-alkylene or napthyl-alkylene, said alkylene chain being optionally further substituted by phenyl or napthyl, each phenyl or napthyl being optionally substituted by one or more substituents each independently selected from alkyl, alkoxy, halo and cyano; n is 1 or 2;
A is NRa, NRaC(O), NRaC(O)NRa, NRaC(O)O, OC(O)NRa, C(O)NRa, NRaSO2, SO2NRa, O, S or SO2,
Ra is H, alkyl or benzyl optionally ring-substituted by one or more substituents each independently selected from alkyl, alkoxy, halo and cyano;
R10 is a group of the formula -(CH2)q-Rp-B; q is O, 1 or 2; Rp is a bond, alkylene, cycloalkylene, phenylene or naphthylene, said cycloalkylene, phenylene and naphthylene each being optionally substituted by one or more substituents each independently selected from alkyl, alkoxy, halo and alkoxyalkylene; B is (i) H, -NRbRb, RbRbN-alkylene, -ORb, -COORb, -OCORb, -SO2Rb, -CN, - SO2NR Rb, -NRbCORb, NRbSO2Rb or -CONRbRb, in which each Rb is the same or different and is selected form H, alkyl, phenyl and benzyl, provided that,
(a) when B is -OCOR , -SO2Rb,-NR CORb or -NRbSO2Rb, then the terminal Rb is not H, and
(b) Rp is a bond, p is 0 and B is H only when A is NRa, NRaC(O)NRa, OC(O)NRa, C(O)NRa-, SO2NRa, O or S, (ii) an optionally-substituted, fully- or partially-saturated or-unsaturated, mono-or bicyclic, heterocyclic group, which is linked to Rp by a ring carbon atom, or (iii) N-linked azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl or morpholinyl, each optionally substituted by one or more alkyl substituents, with the proviso that -(CH2)p-RD-is not-CH2-; and where A is NRa, C(O)NRa, OC(O)NRa or SO2NRa, Ra and R10 taken together with the nitrogen atom to which they are attached can form an azetidine, pyrrolidine, piperidine or piperazine ring, optionally by one or more alkyl substituents; and wherein within this definition the expressions "alkyl", "alkylene", "alkoxy" and the like will be understood to relate to such groups preferably containing 1 to 6, especially 1-4 carbon atoms.
These compounds may be prepared following the method of WO00/23457.
Other selective A2a agonist compounds may be defined by the following formula (V):
Figure imgf000017_0001
wherein: R11 is hydrogen or C,-C6 alkyl optionally substituted by 1 or 2 substituents each independently selected from phenyl and naphthyl, said phenyl and naphthyl being optionally substituted by C,-C6 alkyl, CrC6 alkoxy, halo or cyano; R12 is H or CrC6 alkyl; K is C C6 alkylene;
R13 is (i) hydrogen, C Cβ alkyl, -COOR14, -CN, -CONR14R14, C3-C8 cycloalkyl, phenyl or naphthyl, said C3-C8 cycloalkyl, phenyl and naphthyl being optionally substituted by CrC6 alkyl, phenyl, CrC6 alkoxy (CrC6) alkyl, R1 R N(CrC6) alkyl, halo (CrC6) alkyl, fluoro (CrC6) alkoxy, (C2-C5) alkanoyl, halo, -OR14, cyano, -COOR14, C3-C8 cycloalkyl, -S(O)mR15, -NR14R14, -SO2NR14R14, - CONR14R14, -NR1 COR15 or-NR14SO2R15 or -NR14SO2R15, or (ii) when A is C2-C6 alkylene, -NR14R14, -OR14, -OCOR15, -SO2R15, -S02NR14R14 or -NR14COR15, or (iii) a C-linked, 4- to 11 -membered ring, mono- or bicyclic, heterocycle having either from 1 to 4 ring nitrogen atom(s), or 1 or 2 nitrogen and 1 oxygen or 1 sulphur ring atoms, being optionally C-substituted by oxo, CrC6 alkoxy(C1- C6) alkyl, R16R16N(CrC6) alkyl, halo (CrC6) alkyl, fluoro (CrC6) alkoxy, fluoro (C2- C5) alkanoyl, halo, cyano, -OR16, R17, -COR16, -NR16R16, -COOR16, -S(0)mR17, - SO2NR16R16, -CONR16, -NR16SO2R17 or -NR16COR17 and optionally N-substituted by C C6 alkoxy (CrC6) alkyl, R16R16N(C2-C6) alkyl, halo (CrC6) alkyl, fluoro (C2- C5) alkanoyl, R17, -COR16, -COOR17, -SO2R17, -SO2NR16R16 or -CONR16R16, or (iv) when A is C2-C6 alkylene, N-linked azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, homopiperazinyl or morpholinyl, each being optionally C-substituted by C C6 alkyl, phenyl, CrC6 alkoxy (CrC6) alkyl, R1 R14N(CrC6) alkyl, halo (C C6) alkyl, fluoro (C C6) alkoxy, C2-C5 alkanoyl, halo, -OR14, cyano, -COOR14, C3- C8 cycloalkyl, -S(O)mR15, -NR1 R14, -S02NR14R14, -CONR14R14, -NR14COR15 or - NR1 SO2R15, and said piperazinyl and homopiperazinyl being optionally N- substituted by
Figure imgf000018_0001
alkyl, phenyl, CrC6 alkoxy (C2-C6) alkyl, R14R14(C2-C6) alkyl, fluoro (CrC6) alkyl, C2-C5 alkanoyl, -COOR11, C3-C8 cycloalkyl, -S02R15, - SO2NR14R14 or -CONR14R14;
R14 is H, CrC6 alkyl, C3-C8 cycloalkyl or phenyl;
R15 is CrC6 alkyl, C3-C8 cycloalkyl or phenyl;
R16 is H, CrC6 alkyl, C3-C8 cycloalkyl, phenyl, naphthyl or het;
R17 is CrC6 alkyl, C3-C8 cycloalkyl, phenyl, naphthyl or het; m is 0, 1 or 2; and "het", used in the definitions of R16 and R17, means C-linked pyrrolyl, imidazoyl, triazolyl, thienyl, furyl, thiazolyl, oxazolyl, thiadiazolyl, oxadiazolyl, pyridinyl, pyrimidinyl, pyridazinyl, pyrazinyl, indolyl, isoindolyl, quinolinyl, isoquinolinyl, benzimidazolyl, quinazolinyl, phthalazinyl, benzoxazoyl or quinoxalinyl, each being optionally substituted by CrC6 alkyl, C C6 alkoxy, cyano or halo.
These compounds may be prepared following the method of WO00/77018.
Other selective A2a agonist compounds may be defined by the following formula (VI):
Figure imgf000019_0001
wherein:
R18 represents -NH-N=CH-cyclohexyl, -NH-N=CH-CH2-CHMe2, -O-CH2-CH2-(4- methyl-phenyl), -CC-(CH2)2.15-Me (wherein -CC- represents two carbons connected to each other by a triple bond) or -O-(CH2)2-naphthyl, preferably -NH- N=CH-cyclohexyl or -NH-N=CH-CH2-CHMe2; and R19 represents HOCH2- or EtHNCO-, preferably HOCH2-.
Compounds of formula (VI) wherein R19 represents HOCH2- and R18 represents - NH-N=CH-cyclohexyl or -NH-N=CH-CH2-CHMe are known by the code numbers WRC0474 (SHA211) and WRC0470 respectively.
Compounds of formula (VI) may be prepared as described in WO00/72799 and references contained therein.
The previously mentioned compounds may all be used in the form of physiologically acceptable salts if desired eg in order to optimise the biological or physical properties of the molecule. The representation of formulae (I), (II), (III), (IV), (V) and (VI) indicate the absolute stereochemistry. When sidechains contain chiral centres the invention extends to mixtures of enantiomers (including racemic mixtures) and diastereoisomers as well as individual enantiomers. Generally it is preferred to use a compound of formula (I) in the form of a purified single enantiomer.
In use according to the invention the selective A2a receptor agonist may beneficially be combined in therapy with one or more further therapeutic agents, for example:
Short acting beta-2 agonists, eg albuterol (eg as free base or sulphate) or terbutaline (eg as sulphate);
Long acting beta-2 agonists, eg salmeterol (eg as xinafoate) or formoterol (eg as fumarate);
Glucocorticosteroids, eg fluticasone propionate, beclomethasone dipropionate, mometasone furoate, ciclesonide, budesonide, rofleponide, triamcinolone acetonide;
Anti-cholinergic medicaments, eg ipratropium (eg as bromide), oxitropium, tiotropium;
Non-steroidal anti-inflammatory and/or anti-allergic compounds, eg ketotifen, cromoglycate (eg as sodium), nedocromil (eg as sodium);
DA2-beta-2 agonist compounds, eg 4-hydroxy-7-[2-[[2-[[3-(2- phenylethoxy)propyl]sulfonyl]ethyl]amino]ethyl-2(3H)-benzothiazolone; PDE4 inhibitors, such as cilomilast.
It will be understood that any of the above mentioned medicaments may be used in alternative salt forms and, where appropriate, in the form of enantiomeric mixtures (eg racemates) or single enantiomers.
Combinations of medicaments in therapy may either be administered sequentially or (preferably) simultaneously.
The A2a agonist according to the invention (together if appropriate with any other pharmaceutically active substances) may be formulated for administration in any convenient way, and the invention therefore also includes within its scope pharmaceutical compositions for use in anti-inflammatory therapy, comprising a compound of formula (I) or a physiologically acceptable salt or solvate thereof together, if desirable, with one or more physiologically acceptable diluents or carriers.
There is also provided a process for preparing such a pharmaceutical formulation which comprises mixing the ingredients.
The compounds according to the invention may, for example, be formulated for oral, buccal, parenteral, topical, ocular or rectal administration, preferably for oral, parenteral or topical (e.g. by aerosol) administration.
Tablets and capsules for oral administration may contain conventional excipients such as binding agents, for example syrup, acacia, gelatin, sorbitol, tragacanth, mucilage of starch, cellulose or polyvinyl pyrrolidone; fillers, for example, lactose, microcrystalline cellulose, sugar, maize- starch, calcium phosphate or sorbitol; lubricants, for example, magnesium stearate, stearic acid, talc, polyethylene glycol or silica; disintegrants, for example, potato starch, croscarmellose sodium or sodium starch glycollate; or wetting agents such as sodium lauryl sulphate. The tablets may be coated according to methods well known in the art. Oral liquid preparations may be in the form of, for example, aqueous or oily suspensions, solutions, emulsions, syrups or elixirs, or may be presented as a dry product for constitution with water or other suitable vehicle before use. Such liquid preparations may contain conventional additives such as suspending agents, for example, sorbitol syrup, methyl cellulose, glucose/sugar syrup, gelatin, hydroxymethyl cellulose, carboxymethyl cellulose, aluminium stearate gel or hydrogenated edible fats; emulsifying agents, for example, lecithin, sorbitan mono-oleate or acacia; non-aqueous vehicles (which may include edible oils), for example almond oil, fractionated coconut oil, oily esters, propylene glycol or ethyl alcohol; or preservatives, for example, methyl or propyl 2- hydroxy benzoates or sorbic acid. The preparations may also contain buffer salts, flavouring, colouring and/or sweetening agents (e.g. mannitol) as appropriate. For buccal administration the compositions may take the form of tablets or lozenges formulated in conventional manner.
The compounds may also be formulated as suppositories, e.g. containing conventional suppository bases such as cocoa butter or other glycerides.
Compounds according to the invention may also be formulated for parenteral administration by bolus injection or continuous infusion and may be presented in unit dose form, for instance as ampoules, vials, small volume infusions or pre- filled syringes, or in multi-dose containers with an added preservative. The compositions may take such forms as solutions, suspensions, or emulsions in aqueous or non-aqueous vehicles, and may contain formulatory agents such as anti-oxidants, buffers, antimicrobial agents and/or tonicity adjusting agents. Alternatively, the active ingredient may be in powder form for constitution with a suitable vehicle, e.g. sterile, pyrogen-free water, before use. The dry solid presentation may be prepared by filling a sterile powder aseptically into individual sterile containers or by filling a sterile solution aseptically into each container and freeze-drying.
By topical administration as used herein, we include administration by insufflation and inhalation. Examples of various types of preparation for topical administration include ointments, creams, lotions, powders, pessaries, sprays, aerosols, capsules or cartridges for use in an inhaler or insufflator, solutions for nebulisation or drops (e.g. eye or nose drops).
Ointments and creams may, for example, be formulated with an aqueous or oily base with the addition of suitable thickening and/or gelling agents and/or solvents. Such bases may thus, for example, include water and/or an oil such as liquid paraffin or a vegetable oil such as arachis oil or castor oil or a solvent such as a polyethylene glycol. Thickening agents which may be used include soft paraffin, aluminium stearate, cetostearyl alcohol, polyethylene glycols, microcrystalline wax and beeswax. Lotions may be formulated with an aqueous or oily base and will in general also contain one or more emulsifying agents, stabilising agents, dispersing agents, suspending agents or thickening agents.
Powders for external application may be formed with the aid of any suitable powder base, for example, talc, lactose or starch. Drops may be formulated with an aqueous or non-aqueous base also comprising one or more dispersing agents, solubilising agents or suspending agents.
Spray compositions may be formulated, for example, as aqueous solutions or suspensions or as aerosols delivered from pressurised packs, with the use of a suitable propellant, e.g. dichlorodifluoromethane, trichlorofluoromethane, dichlorotetra-fluoroethane, 1 ,1 ,1 ,2,3,3,3-heptafluoropropane, 1 ,1 ,1,2- tetrafluoroethane, carbon dioxide or other suitable gas.
Intranasal sprays may be formulated with aqueous or non-aqueous vehicles with the addition of agents such as thickening agents, buffer salts or acid or alkali to adjust the pH, isotonicity adjusting agents or anti-oxidants.
Capsules and cartridges of for example gelatin, or blisters of for example laminated aluminium foil, for use in an inhaler or insufflator may be formulated containing a powder mix of a compound of the invention and a suitable powder base such as lactose or starch.
Solutions for inhalation by nebulisation may be formulated with an aqueous vehicle with the addition of agents such as acid or alkali, buffer salts, isotonicity adjusting agents or antimicrobials. They may be sterilised by filtration or heating in an autoclave, or presented as a non-sterile product.
Selective A2a agonist compounds may conveniently be administered in amounts of, for example, 0.001 μg/kg to 50mg/kg body weight, preferably 0.01μg/kg to 10mg/kg body weight, 1 to 4 times daily or on demand. The precise dose will of course depend on the age and condition of the patient, the activity of the molecule selected and the particular route of administration chosen. The preferred route of administration is topical administration to the lung (eg administration by inhalation).
A preferred A2a agonist according to the invention is (2R,3R,4S,5R)-2-[6-Amino- 2-(1S-hydroxymethyl-2-phenyl-ethylamino)-purin-9-yl]-5-(2-ethyl-2H-tetrazol-5- yl)-tetrahydro-furan-3,4-diol and salts thereof (eg the maleate) which may be prepared according to the method of Example 11 in W098/28319. A preferable dose of this compound is 0.06-125μg administered by inhalation each day.
Other compounds of particular interest are:
(2S,3S,4R,5R)-2-(3-Ethyl-isoxazol-5-yl)-5-{6-(1-ethyl-propylamino)-2-[2-(1- methyl-1H-imidazol-4-yl)-ethylamino]-purin-9-yl}-tetrahydro-furan-3,4-diol and salts thereof; and (2S,3S,4R,5R)-2-(3-Ethyl-isoxazol-5-yl)-5-[6-(1-ethyl-propylamino)-2-(2- piperidin-1-yl-ethylamino)-purin-9-yl]-tetrahydro-furan-3,4-diol and salts thereof which may be prepared according to the methods of Examples 5 and 6 respectively in WO99/38877. A preferable dose of these compounds is 0.06-
250μg administered by inhalation each day.
Other compounds of interest are:
(2R,3R,4S,5S)-2-[6-(1-Ethyl-propylamino)-2-(2-piρeridin-1-yl-ethylamino)-purin-
9-yl]-5-(3-methyl-isoxazol-5-yl)-tetrahydro-furan-3,4-diol;
N-(4-{6-(2,2-Diphenyl-ethylamino)-9-[5S-(3-ethyl-isoxazol-5-yl)-3R,4S-dihydroxy- tetrahydro-furan-2R-yl]-9H-purin-2-ylamino}-cyclohexyl)-acetamide;
(2R,3R,4S,5S)-2-{6-(2,2-Diphenyl-ethylamino)-2-[2-(pyridin-2-ylamino)- ethylamino]-purin-9-yl}-5-(3-ethyl-isoxazol-5-yl)-tetrahydro-furan-3,4-diol;
(2R,3R,4S,5S)-2-{6-(3,3-Dimethyl-butylamino)-2-[2-(1-methyl-1H-imidazol-4-yl)- ethylamino]-purin-9-yl}-5-(3-ethyl-isoxazol-5-yl)-tetrahydro-furan-3,4-diol; and (2R,3R,4S,5S)-2-[6-(1-Ethyl-propylamino)-2-(2-piperidin-1-yl-ethyIamino)-purin-
9-yl]-5-(3-hydroxymethyl-isoxazol-5-yl)-tetrahydro-furan-3,4-diol and salts thereof which may be prepared according to the methods of Examples 55, 24, 16, 7 and
57 respectively in WO99/38877. A preferable dose of these compounds is 0.06- 250μg administered by inhalation each day. Other compounds of interest are:
(2R,3R,4S,5R)-2-[6-(2,2-Diphenyl-ethylamino)-2-(1S-hydroxymethyl-2-phenyl- ethylamino)-purin-9-yl]-5-(5-ethyl-4H-[1,2,4]triazol-3-yl)-tetrahydro-furan-3,4-diol; (2R,3R,4S,5R)-2-{6-(2,2-Diphenyl-ethylamino)-2-[2-(1-methyl-1H-imidazol-4-yl)- ethylamino]-purin-9-yl}-5-(5-ethyl-[1 ,2,4]oxadiazol-3-yl)-tetrahydro-furan-3,4-diol; which may be prepared according to the methods of Examples 5 and 13 respectively in WO99/67264. A preferable dose of these compounds is 0.06- 250μg administered by inhalation each day.
Other compounds of interest are:
N-({9-[(2R,3R,4S,5R)-3,4-dihydroxy-5-(methoxymethyl)tetrahydro-2-furanyl]-6- [(2,2-diphenylethyl)amino]-9H-purin-2-yl}methyl)-2-methyl-1- propanesulphonamide and salts thereof (eg the potassium salt); and (2R,3R,4S,5R)-2-(6-[(2,2-diphenylethyl)amino]-2-{[1 -isopropyl-4- piperidinyl)amino]methyl}-9H-purin-9-yl)-5-(methoxymethyl)tetrahydro-3,4- furandiol and salts thereof (eg the dihydroxybutanedioate salt) which may be prepared according to the methods of Examples 15(74) and 16(75) respectively in WO00/23457.
Another compound of interest is:
9-[(2R,3R,4S,5R)-3,4-Dihydroxy-5-(hydroxymethyl)tetrahydro-2-furanyl]-6-[(2,2- diphenylethyl)amino]-N-[2-(1-piperidinyl)ethyl]-9H-purine-2-carboxamide and salts thereof ) which may be prepared according to the method of Example 9 in WOOO/77018.
Other compounds of interest are WRC0474 (SHA211) and WRC0470 which may be prepared as described in WO00/72799 and references contained therein.
The bronchodilatory medicaments according to the invention may be useful in the treatment of diseases of the respiratory tract which include actual or a susceptibility to bronchoconstriction or wherein bronchodilation would be beneficial. The principal such diseases are asthma and chronic obstructive pulmonary disease (COPD). Other diseases for which this therapy may be suitable include adult respiratory distress syndrome (ARDS), emphysema, cystic fibrosis and atopic/allergic diseases having a bronchial component eg allergic rhinitis. The bronchodilatory medicaments according to the invention may also be useful in the treatment of cough.
Brief Description of the Figures:
Figure 1: Mean FEV1 change (in litres) from baseline with time in Cohort 1 following the experiment of the Example.
Figure 2: Mean FEV1 change (in litres) from baseline with time in Cohort 2 following the experiment of the Example.
References:
Ali S, Mustafa SJ and Metzger WJ (1994) J Pharmacol Exper Therapeut 268(3),
1328-34; Asako H, Wolf, RE, Granger, DN (1993), Gastroenterology 104, pp 31-37;
Burkey TH, Webster, RO, (1993), Biochem. Biophys Acta 1175, pp 312-318;
Castanon MJ, Spevak W, (1994), Biochem. Biophys Res. Commun. 198, pp
626-631;
Cronstein BN, Kramer SB, Weissmann G, Hirschhorn R, (1983), Trans. Assoc. Am. Physicians 96, pp 384-91 ;
Cronstein BN, Kramer SB, Rosenstein ED, Weissmann G, Hirschhorn R, (1985),
Ann N.Y. Acad. Sci. 451, pp 291-301;
Cronstein BN, Naime D, Ostad E, (1993), J. Clin. Invest. 92, pp 2675-82;
Cronstein BN, Naime D, Ostad E, (1994), Adv. Exp. Med. Biol., 370, pp 411-6; Cronstein BN, (1994), J. Appl. Physiol. 76, pp 5-13;
Cushley MJ, Tattersfield E and Holgate ST (1983) Br J Clin Pharmacol 15, 161-
165;
Dianzani C, Brunelleschi S, Viano I, Fantozzi R, (1994), Eur. J. Pharmacol 263, pp 223-226; Elliot KRF, Leonard EJ, (1989), FEBS Letters 254, pp 94-98;
Feoktistov I and Biaggioni I (1997) Pharmacol Revs 49, 381-402;
Ghai G, Zimmerman MB and Hopkins MF (1987) Life Sciences 41(10) 1215-24;
Green PG, Basbaum Al, Helms C, Levine JD, (1991), Proc. Natl. Acad Sci. 88, pp 4162-4165; Gustafsson LE, Wiklund NP and Cederqvist B (1986) Eur J Pharmacol 120(2), 179-85;
Klotz KN. Hessling J. Hegler J. Owman C. Kull B. Fredholm BB. Lohse MJ (1998) Naunyn-Schmiedebergs Archives of Pharmacology. 357(1):1-9; Kohno Y, Xiao-duo J, Mawhorter SD, Koshiba M, Jacobson KA. (1996).BIood 88 p3569-3574.
Marimoto et al (1993) Eur J Pharmacol 240, 121-126; Peachell PT, Lichtenstein LM, Schleimer RP, (1989), Biochem Pharmacol 38, 1717-1725; Phillips GD, Ng WH, Church MK and Holgate ST (1990) Am Rev Respir Dis 141 , 9-13; Richter J, (1992), J. Leukocyte Biol. 51, pp 270-275;
Robeva AS. Woodard RL. Jin XW. Gao ZH. Bhattacharya S. Taylor HE. Rosin DL. Linden J. (1996) Drug Development Research. 39(3-4):243-252; Rosengren S, Bong GW, Firestein GS, (1995), J. Immunol. 154, pp 5444-5451;
Skubitz KM, Wickman NW, Hammerschmidt DE, (1988), Blood 72, pp 29-33;
Varani K. Cacciari B. Baraldi PG. Dionisotti S. Ongini E. Borea PA. (1998) Life
Sciences. 63(5):PL 81-PL 87;
Wood KV. (1995) Curr Opinion Biotechnology 6 p50-58.
Example
A randomised, double blind, double dummy, placebo controlled, cross-over study to investigate the effects of cumulative inhaled doses of nebulised and dry-powder (2R,3R,4S,5R)-2-[6-Amino-2-(1S-hydroxymethyl-2-phenyl- ethylamino)-purin-9-yl]-5-(2-ethyl-2H-tetrazol-5-yl)-tetrahydro-furan-3,4-diol
("drug") on airway hyper-responsiveness in mild asthmatic patients.
Protocol
The study was conducted in 24 mild asthmatics in 2 centres. There were 2 Cohorts in each centre - Cohort 1 received nebulised doses of drug or placebo via HaloLite nebuliser and Cohort 2 received dry powder doses of drug or placebo via Diskhaler inhaler. Subjects attended the unit for a screening visit before treatment commenced. They were required to demonstrate stable PC20 (PC20 is the provocative concentration causing a 20% fall in FEV1 from baseline) in response to adenosine monophosphate (AMP) challenge within 7 days prior to randomisation, at the beginning of the trial and at the end. Nebulised placebo consisted of 0.9% sodium chloride solution in water. Nebulised drug formulation consisted of 500μg/ml drug and 0.9% sodium chloride in water.
Dry powder placebo consisted of a Diskhaler blister containing lactose. Dry powder drug formulation consisted of a Diskhaler blister containing 25μg drug per blister and lactose.
Cohort 1 (n=6) received on 3 separate days:
A: 5 cumulative low doses of 0.06, 0.12, 0.24, 0.48 and 0.96 μg of drug via nebuliser (Haloϋ'te) (total daily dose received = 1.86 μg). B: 5 cumulative high doses of 0.96, 1.92 3.84, 7.68 and 15.36 μg of drug via nebuliser (HaloLite) (total daily dose received = 29.76 μg). C: 5 doses of matched placebo via nebuliser
Cohort 2 (n = 6) received on 3 separate days:
A: 5 cumulative low doses each of 25 μg of drug via Diskhaler inhaler (total daily dose received = 125 μg). B: A single high dose of 75 μg of drug , then 2 doses each of 25 μg of drug and then 2 doses each of placebo via Diskhaler inhaler (total daily dose received
= 125 μg). C: 5 doses of matched placebo via Diskhaler inhaler.
Patients were monitored for changes in FEV1 before and after AMP challenge and dosing.
Results
The results are shown in Figures 1 and 2. The arrows indicate the 5 dosing time points.
Conclusion
From Figures 1 and 2 it can be seen that administration of (2R,3R,4S,5R)-2-[6- Amino-2-(1S-hydroxymethyl-2-phenyl-ethylamino)-purin-9-yl]-5-(2-ethyl-2H- tetrazol-5-yl)-tetrahydro-furan-3,4-diol according to the study design resulted in a rapid dose-dependent increase in FEV1 of up to 150-200ml after administration of drug compared with placebo. This provides consistent evidence for a rapid-onset bronchodilatory effect in asthmatic patients of a magnitude which would be of therapeutic benefit.
' Patents and patent applications referred to herein are incorporated by reference.
Throughout the specification and the claims which follow, unless the context requires otherwise, the word 'comprise', and variations such as 'comprises' and 'comprising', will be understood to imply the inclusion of a stated integer or step or group of integers but not to the exclusion of any other integer or step or group of integers or steps.

Claims

Claims
1. A method of inducing bronchodilation in a human patient which comprises administering to that patient an effective amount of a selective A2a receptor agonist.
2. A method of inducing bronchodilation in a human patient suffering from an inflammatory disorder (eg asthma or COPD) which comprises administering to that patient an effective amount of a selective A2a receptor agonist.
3. A method of relieving bronchoconstriction in a human patient which comprises administering to that patient an effective amount of a selective
A2a receptor agonist.
4. A method of relieving bronchoconstriction in a human patient suffering from an inflammatory disorder (eg asthma or COPD) which comprises administering to that patient an effective amount of a selective A2a receptor agonist.
5. A pharmaceutical composition for the inducing of bronchodilation in a human patient which comprises an effective amount of a selective A2a receptor agonist.
6. A pharmaceutical composition for inducing bronchodilation in a human patient suffering from an inflammatory disorder (eg asthma or COPD) which comprises an effective amount of a selective A2a receptor agonist.
7. A pharmaceutical composition for the relief of bronchoconstriction in a human patient which comprises an effective amount of a selective A2a receptor agonist.
8. A pharmaceutical composition for the relief of bronchoconstriction in a human patient suffering from an inflammatory disorder (eg asthma or COPD) which comprises an effective amount of a selective A2a receptor agonist.
9. The use of a selective A2a receptor agonist in the manufacture of a medicament for the induction of bronchodilation in a human patient.
10. The use of a selective A2a receptor agonist in the manufacture of a medicament for the induction of bronchodilation in a human patient suffering from an inflammatory disorder (eg asthma or COPD).
11. The use of a selective A2a receptor agonist in the manufacture of a medicament for the relief of bronchoconstriction in a human patient.
12. The use of a selective A2a receptor agonist in the manufacture of a medicament for the relief of bronchoconstruction in a human patient suffering from an inflammatory disorder (eg asthma or COPD).
13. A composition, method or use according to any one of claims 1-12 wherein the selective A2a agonist is also an A3 antagonist.
14. A composition, method or use according to any one of claims 3, 4, 7, 8, 11 or 12 wherein the relief is acute relief.
15. A composition, method or use according to any one of claims 1-12 wherein the selective A2a agonist is a derivative of adenosine.
16. A composition, method or use according to claim 15 wherein the A2a agonist is (2R,3R,4S,5R)-2-[6-Amino-2-(1S-hydroxymethyl-2-phenyl-ethylamino)- purin-9-yl]-5-(2-ethyl-2H-tetrazol-5-yl)-tetrahydro-furan-3,4-diol or a salt thereof.
17. A composition, method or use according to claim 15 wherein the A2a agonist is (2S,3S,4R,5R)-2-(3-Ethyl-isoxazol-5-yl)-5-{6-(1 -ethyl-propylamino)-2-[2-(1 - methyl-1 H-imidazol-4-yI)-ethylamino]-purin-9-yl}-tetrahydro-furan-3,4-diol or a salt thereof.
18. A composition, method or use according to any one of claims 1 to 12 wherein the selective A2a agonist is administered in combination with a beta- 2 agonist.
19. A composition, method or use according to any one of claims 1 to 12 wherein the selective A2a agonist is administered by inhalation.
PCT/GB2002/000809 2001-02-23 2002-02-22 New method of bronchodilatory therapy WO2002067909A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GB0104555.8 2001-02-23
GBGB0104555.8A GB0104555D0 (en) 2001-02-23 2001-02-23 New Therapeutic method

Publications (1)

Publication Number Publication Date
WO2002067909A1 true WO2002067909A1 (en) 2002-09-06

Family

ID=9909388

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2002/000809 WO2002067909A1 (en) 2001-02-23 2002-02-22 New method of bronchodilatory therapy

Country Status (2)

Country Link
GB (1) GB0104555D0 (en)
WO (1) WO2002067909A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006128674A2 (en) * 2005-05-31 2006-12-07 Novartis Ag Combinations of indacaterol derivatives and other agents for the treatment of airway diseases
US7737126B2 (en) 2004-05-24 2010-06-15 Glaxo Group Limited Purine derivative
US7985740B2 (en) 2005-07-19 2011-07-26 Glaxo Group Limited Purine derivatives as agonists of the adenosine A2A receptor

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998028319A1 (en) * 1996-12-24 1998-07-02 Glaxo Group Limited 2-(purin-9-yl)-tetrahydrofuran-3,4-diol derivatives
WO1999038877A2 (en) * 1998-01-31 1999-08-05 Glaxo Group Limited 2-(PURIN-9-yl)-TETRAHYDROFURAN-3,4-DIOL DERIVATIVES
WO1999067265A1 (en) * 1998-06-23 1999-12-29 Glaxo Group Limited 2-(purin-9-yl)-tetrahydrofuran-3,4-diol derivatives
WO1999067264A1 (en) * 1998-06-23 1999-12-29 Glaxo Group Limited 2-(purin-9-yl)-tetrahydrofuran-3,4-diol derivatives
WO2000077018A2 (en) * 1999-06-15 2000-12-21 Pfizer Limited Purine derivatives

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998028319A1 (en) * 1996-12-24 1998-07-02 Glaxo Group Limited 2-(purin-9-yl)-tetrahydrofuran-3,4-diol derivatives
WO1999038877A2 (en) * 1998-01-31 1999-08-05 Glaxo Group Limited 2-(PURIN-9-yl)-TETRAHYDROFURAN-3,4-DIOL DERIVATIVES
WO1999067265A1 (en) * 1998-06-23 1999-12-29 Glaxo Group Limited 2-(purin-9-yl)-tetrahydrofuran-3,4-diol derivatives
WO1999067264A1 (en) * 1998-06-23 1999-12-29 Glaxo Group Limited 2-(purin-9-yl)-tetrahydrofuran-3,4-diol derivatives
WO2000077018A2 (en) * 1999-06-15 2000-12-21 Pfizer Limited Purine derivatives

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7737126B2 (en) 2004-05-24 2010-06-15 Glaxo Group Limited Purine derivative
WO2006128674A2 (en) * 2005-05-31 2006-12-07 Novartis Ag Combinations of indacaterol derivatives and other agents for the treatment of airway diseases
WO2006128674A3 (en) * 2005-05-31 2007-04-05 Novartis Ag Combinations of indacaterol derivatives and other agents for the treatment of airway diseases
US7985740B2 (en) 2005-07-19 2011-07-26 Glaxo Group Limited Purine derivatives as agonists of the adenosine A2A receptor

Also Published As

Publication number Publication date
GB0104555D0 (en) 2001-04-11

Similar Documents

Publication Publication Date Title
EP0948509B1 (en) 2-(purin-9-yl)-tetrahydrofuran-3,4-diol derivatives
ES2228173T3 (en) PROCEDURE AND COMPOSITIONS FOR THE PREVENTION OF TOLERANCE TO BRONCODILATORS.
EP1090022B1 (en) 2-(purin-9-yl)-tetrahydrofuran-3,4-diol derivatives
EP1397140A2 (en) Combination of an adenosine a2a-receptor agonist and tiotropium or a derivative thereof for treating obstructive airways
ES2745064T3 (en) Super fine formoterol formulation
US6534486B1 (en) 2-(purin-9-yl)-Tetrahydrofuran-3,4-diol derivatives
JP2002518511A (en) 2- (purine-9-yl) -tetrahydrofuran-3,4-diol derivative
KR20010034473A (en) 2-(Purin-9-yl)-tetrahydrofuran-3,4-diol Derivatives
PL206965B1 (en) Combinations of formoterol and a tiotropium salt
EP1274434A1 (en) Medical combinations comprising tiotropium and rofleponide
JPH07509476A (en) Medications for the treatment or prevention of high intraocular pressure
WO2002067909A1 (en) New method of bronchodilatory therapy
US20040009963A1 (en) Use of salmeterol and fluticasone propionate combination
EP1434587A1 (en) Pharmaceutical combinations comprising salmeterol and fluticasone proprionate for the treatment of asthma
EP1274436A1 (en) Medical compositions comprising (r,r)-formoterol und rofleponide
US20070134165A1 (en) Use of Ciclesonide for the Treatment of Respiratory Disease in a Smoking Patient
US20030109485A1 (en) Pharmaceutical combination
US20040019025A1 (en) Medical compositions comprising (r,r)-formoterol and rofleponide
US3198703A (en) Method of producing sympathomimetic activity by isopropyl-2&#39;-phenoxymethyl-2-imidazoline
EP1462148A2 (en) Methods and compositions for the prevention of tolerance to bronchodilators
BRPI0708197A2 (en) combination of steroids and methylxanthine compounds

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ OM PH PL PT RO RU SD SE SG SI SK SL TJ TM TN TR TT TZ UA UG US UZ VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase

Ref country code: JP

WWW Wipo information: withdrawn in national office

Country of ref document: JP