Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
OXYTOCIN INHIBITORS
Document Type and Number:
WIPO Patent Application WO/2004/020414
Kind Code:
A1
Abstract:
This invention relates to compounds of formula (I).

Inventors:
ARMOUR DUNCAN ROBERT (GB)
BELL ANDREW SIMON (GB)
EDWARDS PAUL JOHN (GB)
ELLIS DAVID (GB)
HEPWORTH DAVID (GB)
LEWIS MARK LLEWELLYN (GB)
SMITH CHRISTOPHER RONALD (GB)
Application Number:
PCT/IB2003/003705
Publication Date:
March 11, 2004
Filing Date:
August 13, 2003
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
PFIZER LTD (GB)
ARMOUR DUNCAN ROBERT (GB)
BELL ANDREW SIMON (GB)
EDWARDS PAUL JOHN (GB)
ELLIS DAVID (GB)
HEPWORTH DAVID (GB)
LEWIS MARK LLEWELLYN (GB)
SMITH CHRISTOPHER RONALD (GB)
PFIZER (US)
International Classes:
A61K31/16; A61P15/04; A61P15/10; C07C233/65; C07C237/22; C07C255/57; C07D213/81; C07D213/82; C07D319/18; C07D401/12; C07D405/12; C07D521/00; (IPC1-7): C07D213/82; C07D319/18; C07D213/81; C07D405/12; C07D521/00; C07D401/12; C07C255/57; A61K31/44; A61K31/4427; A61P15/04; A61P15/10
Domestic Patent References:
WO2003037274A22003-05-08
WO2003007888A22003-01-30
WO2002044142A22002-06-06
WO1999006340A21999-02-11
Foreign References:
US2496882A1950-02-07
EP0189774A11986-08-06
US4060402A1977-11-29
Other References:
DATABASE CHEMCATS CHEMICAL ABSTRACT SERVICE, COLUMBUS, OHIO, US; XP002264114
DATABASE CHEMCATS CHEMICAL ABSTRACT SERVICE, COMLUBUS, OHIO, US; XP002264115
DATABASE CAPLUS [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; SASAKI, YOSHIO ET AL: "Rational estimation of the QSAR (quantitative structure-activity relationships) descriptors.sigma.S.degree., and their applications for medicinals now available", XP002264116, retrieved from STN Database accession no. 119:195101
BRAGG R A ET AL: "'meso-Selective' functionalisation of N-benzyl-alpha-methylbenzylamine derivatives by alpha-lithiation and alkylation", TETRAHEDRON LETTERS, ELSEVIER SCIENCE PUBLISHERS, AMSTERDAM, NL, vol. 43, no. 11, 11 March 2002 (2002-03-11), pages 1955 - 1959, XP004339077, ISSN: 0040-4039
DATABASE CAPLUS [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; DUNINA, V. V. ET AL: "Optically active organic thiones and their complexes. IV. Synthesis of N-sec-butyl- and N-.alpha.-phenylethylthioamides", XP002264117, retrieved from STN Database accession no. 87:167684
DATABASE CAPLUS [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; ARIES, ROBERT: "Nicotinoylaralkylamines", XP002264118, retrieved from STN Database accession no. 79:146413
DATABASE CAPLUS [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; DAVID, SERGE ET AL: "Hydrogenation and ring opening of certain hydroxypyrimidines", XP002264119, retrieved from STN Database accession no. 63:71981
DATABASE CAPLUS [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; POTAPOV, V. M. ET AL: "Stereochemical studies. XIII. Rotational dispersion of derivatives of.alpha.-p-tolylethylamine and 2-aminobutane", XP002264120, retrieved from STN Database accession no. 58:14418
Attorney, Agent or Firm:
Lumb, Trevor J. c/o Simpson (Alison Urquhart-Dykes & Lord, 30 Welbeck Stree, London W1G 8ER, GB)
Download PDF:
Claims:
Claims
1. A compound of formula (I) wherein: R'is selected from: a) phenyl, which is optionally substituted by 13 groups each independently selected from C1C6 alkyl, CF3, halo, CN, NR7R8, OCF3, SOR6, SO2R6 and OC1C6 alkyl, wherein said alkyl group may be optionally substituted by : C3C8 cycloalkyl group, and b) Aromatic Heterocycle, which is optionally substituted by 13 groups each independently selected from C1C6 alkyl, NH2, CF3, halo, OH, OC1C6 alkyl, SR6, SOR6, SO2R6, NR7R8 wherein R8 may be optionally substituted by NH2, phenyl or Heterocycle, and OPh wherein Ph may be optionally substituted by 13 groups each independently selected from halo and C1 C6 alkyl ; R2 is selected from : a) phenyl, which is optionally substituted by C1C6 alkyl, halo, CN, NR7R8, OC1C6 alkyl, OCF3, CF3 and SO2R6, b) OPh, which is optionally substituted by C1C6 alkyl, halo, OC1C6 alkyl, OCF3, CF3 and SO2R6 c) C3C8 cycloalkyl which is optionally fused to phenyl, d) Aromatic Heterocycle, e) R', f) C (O) NR6R6, and g) Heterocycle, which is optionally substituted by C (O) R6 ; R3 is selected from : a) phenyl, said phenyl being optionally fused to Heterocycle and said phenyl or said fused phenyl being optionally substituted by 13 groups each independently selected from: C1C6 alkyl, CF3, halo, CN, OCF3, S02R 6 andOClC6alkyl, b) Heterocycle, c) R, d) 38 membered cycloalkyl group, which is optionally substituted by CiCe alkyl, and e) Aromatic Heterocycle, which is optionally substituted by C1C6 alkyl ; 4iS hydrogen or CH3 ; R5 is selected from: a) CONH2, CONHR6, CONR6R6, R6, NH2, NHR6, OH, OR6, OC(O)NHR6, NHC(O)H, NHC(O)R6, and b) Aromatic Heterocycle, which is optionally substituted by 13 groups each independently selected from C1C6 alkyl, NH2, CF3, halo, SR6, OH, OC1C6 alkyl, NHR6 wherein the R6 moiety may be optionally substituted by phenyl or Heterocycle, and OPh wherein Ph may be optionally substituted by 13 groups each independently selected from halo and C16 alkyl ; R6 is C16 alkyl ; R7 is hydrogen or C1C6 alkyl ; R8 is C1. C6 alkyl ; or NR7R8 forms a monocyclic saturated ring system containing between 3 and 7 ring atoms ; x is 0, 1 or 2, y is 0, 1 or 2, and z is 0,1 or 2, and wherein : Aromatic Heterocycle may be defined as a 56 membered aromatic heterocycle containing 14 heteroatoms each independently selected from N, O and S, said ring optionally, fused with a phenyl or a 38 membered cycloalkyl group ; Heterocycle is a 58 membered saturated or partially saturated ring containing 13 heteroatoms each independently selected from N, O and S, said ring optionally fused with phenyl ; a tautomer thereof or a pharmaceutical acceptable salt, solvate or polymorph of said compound or tautomer.
2. A compound according to claim 1 wherein R1 is selected from: a) phenyl, which is optionally substituted by 13 groups each independently selected from C1C6 alkyl, CF3, halo, CN, NR7R8, SO2R6 and OC1C6 alkyl, and b) Aromatic Heterocycle, wherein said Aromatic Heterocycle is selected from pyridyl, pyrazinyi, pyrimidinyl, quinolinyl, quinoxalinyl, isoxazolyl and pyrazolyl, each aromatic heterocycle optionally substituted by 13 groups each independentiy selected from C1C6 alkyl, SR6, S02R6, NH2, CF3, halo, OH, OC1C6 alkyl, NR7R8 wherein R8 may be optionally substituted by NIH2, phenyl or Heterocycle, and OPh wherein Ph may be optionally substituted by 13 groups each independently selected from halo and C1C6 alkyl ; R2 is selected from: a) phenyl, which is optionally substituted by C1C6 alkyl, halo, OC1C6 alkyl, OCF3, NR7R8, CF3 or SO2R6, b) OPh, which is optionally substituted by C1 C6 alkyl or halo, c) cyclopropyl or 1or 2indanyl, d) pyrazolyl, which is optionally substituted by R6, e) R6, f) C (O) N(CH3)2, and g) a 56 membered saturated ring containing 1 nitrogen atom, said ring being substituted by C (O) R6 ; R3 is selected from : a) phenyl, said phenyl being optionally fused to Heterocycle and said phenyl or said fused phenyl being optionally substituted by 13 groups each independently selected from C1C6 alkyl, halo, CN and OC1C6 alkyl, b) R6, c) cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl, which is optionally substituted by C1C6 alkyl ; and d) Aromatic Heterocycle, wherein said Aromatic Heterocycle may be defined as a 56 membered aromatic heterocycle containing 1 or 2 nitrogen atoms, said ring optionally fused with a phenyl or a 38 membered cycloalkyl group. R4 is H; R5 is selected from : CONH2, CONHR6, CONR6R6 and R6 ; Fi6 is methyl ; xis 1; y is 0 ; and z is 0 or 1.
3. A compound according to claim 2 wherein R1 is selected from : a) phenyl, which is optionally substituted by 13 groups each independently selected from C1C6 alkyl, CF3, halo, CN, NR7R8, S02R6 and OC1C6 alkyl, and b) Aromatic Heterocycle, wherein said Aromatic Heterocycle is selected from : i) pyridyl, which is optionally substituted by 13 groups each independently selected from C1C6 alkyl, SO2R6, NH2, CF3, CN, halo, OH, OC1C6 alkyl, NR7R8 wherein R8 may be optionally substituted by NH2, phenyl or Heterocycle, and OPh wherein Ph may be optionally substituted by 13 groups each independently selected from, halo and C1C6 alkyl; <BR> <BR> ii) pyrimidinyl, which is optionally substituted by 13 groups each<BR> <BR> independently selected from C1C6 alkyl, SO2R6, NH2, CF3, CN, halo, OH, OC1C6 alkyl, NR7R8 wherein R8 may be optionally substituted by l\lH2, phenyl or Heterocycle, and OPh wherein Ph may be optionally substituted by 13 groups each independently selected from halo and C1C6 alkyl; <BR> <BR> <BR> iii) pyrazinyl, which is optionally substituted by 13 groups each<BR> <BR> <BR> <BR> <BR> independently selected from C1C6 alkyl, NH2, SR6 and halo; iv) quinolinyl; v) quinoxalinyl, which is optionally substituted by OH ; vi) isoxazolyi, which is optionally substituted by 13 groups each independently selected from : C1C6 alkyl; and vii) pyrazol ; R2 is selected from : a) phenyl, which is optionally substituted by methyl, halo, methoiy, CF3 <BR> <BR> <BR> <BR> or S02CH3,<BR> <BR> <BR> <BR> <BR> <BR> <BR> b) cyclopropyl or 1or 2indanyl, c) pyrazolyl, which is optionally substituted by methyl, d) C(O)N(CH3) 2, and e) piperidinyl substituted by C (O) R6. R3 is selected from : a) phenyl, said phenyl being optionally fused to 1, 4dioxan and said phenyl or said fused phenyl being optionally substituted by 13 groups each independently selected from C1C6 alkyl, halo, CN and OC1C6 alkyl; b) R6, c) cyclopropyl, which is optionally substituted by C1C6 alkyl; and d) Aromatic Heterocycle, wherein said Aromatic Heterocycle is selected from pyrazolyl, or pyridyl, both optionally substituted by C1C6 alkyl ; R5 is CONH2 or CH3 ; and z is 0.
4. A compound according to any one of claims 1 to 3 wherein R'is. phenyl, 2 or 3pyridyl or 2, 4pyrimidinyl, said moieties being optionally substituted by 1 3 groups each independently selected from C1C6 alkyl, halo, OC1C6 alkyl, CN, SO2R6, NHR7, NHCH2CH2NH2 and CF3;.
5. A compound according to claim 4 wherein R'. is phenyl, 2or 3pyridyl or 2, 4pyrimidinyl, said moieties being optionally substituted by 13 groups each independently selected from methyl, fluoro, chloro, methoxy, ethoxy, npropoxy, CN, S02CH3, NH2, NHCH3, NHCH2CH2NH2, and CF3.
6. A compound according to any one of claims 1 to 5 wherein R 2 is selected from: a) phenyl, which is optionally substituted by methyl, fluoro, chloro, metho ? ny, CF3 or SO2CH3, b) pyrazolyl, which is optionally substituted by methyl, and c) C (O) N (CH3) 2.
7. A compound according to claim 6 wherein R2 is phenyl, para fluorophenyl, parachlorophenyl, paramethylphenyl, 2, 5dimethylphenyl, o methylphenyl and paramethoxyphenyl.
8. A compound according to any one of claims 1 to 7 wherein R3 is selected from : a) phenyl, said phenyl being optionally fused to 1, 4dioxan and said phenyl or said fused phenyl being optionally substituted by 12 groups each independently selected from methyl, methoxy, ethoxy, fluoro, chloro and CN ; b) isopropyl; c) cyclopropyl ; and d) pyrazolyl and pyridyl, both optionally substituted by methyl.
9. A compound according to claim 8 wherein R3 is 3methoxyphenyl or 1, 4 benzodioxanyl.
10. A compound according to any one of claims 1 to wherein ; R5 is CONH2.
11. A compound according to claim 1 selected from: 2AminoN[2amino1(2methylphenyl)2oxoethyl]N(4 chlorobenzyl)nicotinamide, N[2Amino1(3methoxyphenyl)2oxoethyl]4cyanoN(4 methylbenzyl) benzamid, N[3Amino1(3methoxyphenyl)3oxopropyl]4methylN(4 methylbenzyl) nicotinamide, 2AminoN[(1S)3amino3oxo1phenylpropyl]N(4 methylbenzyl) nicotinamide, 5Chloro2methylthioN[2amino1{1,4benzodioxan6yl}2oxoethyl]N(4 methylbenzyl)pyrimidine4carboxamide, 5Chloro2aminoN[2amino1{1,4benzodioxan6yl}2oxoethyl]N(4 methylbenzyl) pyrimidine4carboxamide, and 2AminoN[carbamoyl(2,3dihydrobenzo[1,4]dioxin6yl)methyl]4,6dimethyl N (4methylbenzyl)nicotinamide ; and tautomers thereof and pharmaceutically acceptable salts, solvates and polymorphs of said compound or tautomer.
12. A pharmaceutical composition comprising a compound of formula (I) as claimed in any one of claims 1 to 11, or pharmaceutical acceptable salts, solvates or polymorphs thereof, and a pharmaceutical acceptable diluent or carrier.
13. A compound of formula (I) as claimed in any one of claims 1 to 11, or a pharmaceutical acceptable salt, solvate or polymorph thereof, for use as a medicament.
14. A method of treatment of a disorder or condition where inhibition of Oxytocin is known, or can be shown, to produce a beneficial effect, in a mammal, comprising administering to said mammal a therapeutical effective amount of a compound of formula (I) as claimed in any one of claims 1 to 11, or a pharmaceutically acceptable salt, solvate or polymorph thereof.
15. Use of a compound of formula (I) as claimed in any one of claims 1 to 11, or a pharmaceutical acceptable salt, solvate or polymorph thereof, in the preparation of a medicament for the treatment of a disorder or condition where inhibition of Oxytocin is known, or can be shown, to produce a beneficial effect.
16. Use according to either claim 14 or claim 15, wherein the disorder or condition is selected from sexual dysfunction (including premature ejaculation), preterm labour, complications in labour, appetite and feeding disorders, obesity, benign prostatic hyperplasia, premature birth, dysmenorrhoea, congestive heart failure, arterial hypertension, liver cirrhosis, nephrotic hypertension, occular hypertension, obsessive compulsive disorder and neuropsychiatric disorders.
17. Use according to claim 16, wherein the disorder or condition is premature ejaculation.
Description:
Oxytocin Inhibitors The present invention relates to a class of substituted amides with activity as Oxytocin inhibitors, uses thereof, processes for the preparation thereof and compositions containing said inhibitors. These inhibitors have utility in a variety of therapeutic areas including sexual dysfunction, particularly premature ejaculation (P. E.).

The present invention provides for compounds of formula (I) wherein: R1 is selected from: a) phenyl, which is optionally substituted by 1-3 groups each independently selected from C1 C6 alkyl, CF3, halo, CN, NR7R8, OCF3, SOR6, SOIR6 and OC1 C6 alkyl, wherein said alkyl group may be optionally substituted by a C3 C8 cycloalkyl group, and b) Aromatic Heterocycle, which is optionally substituted by 1-3 groups each independently selected from C1 C6 alkyl, NH2 CF3, halo, OH, OC1-C6 alkyl, SR6, SOR6, SO2R6, NR7R8 wherein R may be optionally substituted by NH2, phenyl or Heterocycle, and OPh wherein Ph may be optionally substituted by 1-3 groups each independently selected from halo and C1 C6 alkyl ;

R2 is selected from : a) phenyl, which is optionally substituted by C1-C6 alkyl, halo, CN, NR OC1-C6 alkyl, OCF3, CF3 and SO2R6, b) OPh, which is optionally substituted by C1-C6 alkyl, halo, OC1-C6 alkyl, OCF3, CF3 and SO2R6, c) C3-C8 cycloalkyl which is optionally fused to phenyl, d) Aromatic Heterocycle, e) R6, f) C (O) NR6R6, and g) Heterocycle, which is optionally substituted by C (O) R6 ; R3 is selected from : a) phenyl, said phenyl being optionally fused to Heterocycle and said phenyl or said fused phenyl being optiona ! ly substituted by 1-3 groups each independently selected from: C1-C6 alkyl, CF3, halo, CN, OCF3, S02RE3 and OC1-C6 alkyl, b) Heterocycle, c) R6, d) 3-8 membered cycloalkyl group, which is optionally substituted by C1 C6 alkyl, ana' e) Aromatic Heterocycle, which is optionally substituted by Cl-C6 alkyl ; R4 is hydrogen or CH3 ; R5 is selected from : a) CONH2, CONHR6, CONR6R6, R6, NH2, NHR6, OH, OR6, OC(O)NHR6, NHC NHC(O)R6, and b) Aromatic Heterocycle, which is optionally substituted by 1-3 groups each independently selected from C1-C6 alkyl, NH2, CF3, halo, SR6, OH, OC1-C6 alkyl, NHR6 wherein the R6 moiety may be optionally substituted by phenyl or Heterocycle, and OPh wherein Ph may be optionally

substituted by 1-3 groups each independently selected from halo and Cl-6 alkyl ; R6 is C1-6 alkyl; R7 is hydrogen or C1 C6 alkyl ; R8 is Cl-C6alkyl ; or NR7R8 forms a monocyclic saturated ring system containing between 3 and 7 ring atoms; x is 0, 1 or 2, y is 0, 1 or 2, and z is 0,1 or 2, and wherein : Aromatic Heterocycle may be defined as a 5-6 membered aromatic heterocycle containing 1-4 heteroatoms each independently selected from N, O and S, said ring optionally fused with a phenyl or a 3-8 membered cycloalkyl group; Heterocycle is a 5-8 membered saturated or partially saturated ring containing 1-3 heteroatoms each independently selected from N, 0 and S, said ring optionally fused with phenyl.

The pharmaceutical acceptable salts of the compounds of the formula (I) include the acid addition and the base salts thereof.

A pharmaceutical acceptable salt of a compound of the formula (I) may be readily prepared by mixing together solutions of a compound of the formula (I) and the desired acid or base, as appropriate. The salt may precipitate from

solution and be collected by filtration or may be recovered by evaporation of the solvent.

Suitable acid addition salts are formed from acids which form non-toxic salts and examples are the hydrochloride, hydrobromide, hydroiodide, sulphate, bisulphate, nitrate, phosphate, hydrogen phosphate, acetate, maleat, fumarate, lactate, tartrate, citrate, gluconate, succinate, saccharate, benzoate, methanesulphonate, ethanesulphonate, benzenesulphonate, p-toluenesulphonate and pamoate salts.

Suitable base salts are formed from bases which form non-toxic salts and examples are the sodium, potassium, aluminium, calcium, magnesium, zinc and diethanolamine salts.

For a review on suitable salts see Berge et al, J. Pharm. Sci., 66, 1-19, 1977.

The pharmaceutically acceptable solvates of the compounds of the formula (I) include the hydrates thereof.

Also included within the present scope of the compounds of the formula (i) are polymorphs thereof.

Also included within the present scope of the compounds of the formula (I) are atropisomers thereof.

A compound of the formula (I) contains one or more asymmetric carbon atoms and therefore exists in two or more stereoisomeric forms. Where a compound of the formula (I) contains an alkenyl or alkenylene group, cis (Z) and trans (E) isomerism may also occur. The present invention includes the individual stereoisomers of the compounds of the formula (I) and, where appropriate, the individual tautomeric forms thereof, together with mixtures thereof.

Separation of diastereoisomers or cis and trans isomers may be achieved by conventional techniques, e. g. by fractional crystallisation, chromatography or H. P. L. C. of a stereoisomeric mixture of a compound of the formula (I) or a suitable salt or derivative thereof. An individual enantiomer of a compound of the formula (I) may also be prepared from a corresponding optically pure intermediate or by resolution, such as by H. P. L. C. of the corresponding racemate using a suitable chiral support or by fractional crystallisation of the diastereoisomeric salts formed by reaction of the corresponding racemate with a suitable optically active acid or base, as appropriate.

Unless otherwise indicated, an alkyl. or alkoxy group may be straight or branched and contain 1 to 8 carbon atoms, preferably 1 to 6 and particularly 1 to 4 carbon atoms. Examples of alkyl include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, pentyl and hexyl. Examples of alkoxy include methoxy, ethoxy, isopropoxy and n-butoxy.

Unless otherwise indicated, a cycloalkyl or cycloalkoxy group may contain 3 to 10 ring-atoms, may be either monocyclic or, when there are an appropriate number of ring atoms, polycyclic. Examples of cycloalkyl groups are cyclopropyl, cyclopentyl, cyclohexyl and adamantyl.

Examples of Aromatic Heterocycle are furyl, thienyl, pyrrolyl, oxazolyl, thiazolyl, <BR> <BR> <BR> imidazoiyl, pyrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, tetrazolyl, triazinyl. In addition, the term heteroaryl includes fused heteroaryl groups, for example benzimidazolyl, <BR> <BR> <BR> benzoxazolyl, imidazopyridinyl, benzoxazinyl, benzothiazinyl, oxazolopyridinyl, benzofuranyl, quinolinyl, quinazolinyl, quinoxalinyl, benzothiazolyl, phthalimido, benzofuranyl, benzodiazepinyl, indolyl and isoindolyl.

Examples of Heterocycle are oxiranyl, azetidinyl, tetrahydrofuranyl, thiolanyl, pyrrolidinyl, pyrrolinyl, imidazolidinyl, imidazolinyl, sulfolanyl, dioacolanyl, dihydropyrariyl, tetrahydropyranyl, piperidinyl, pyrazolinyl, pyrazolidinyl,

dioxanyl, morpholinyl, dithianyl, thiomorpholinyl, piperazinyl, azepinyl, oxazepinyl, thiazepinyl, thiazolinyl and diazapanyl.

Halo means fluoro, chloro, bromo or iodo.

Unless otherwise indicated, the term substituted means substituted by one or more defined groups. In the case where groups may be selected from a number of alternative groups, the selected groups may be the same or different.

Preferably R'is selected from : a) phenyl, which is optionally substituted by 1-3 groups each independently selected from C1 C6 alkyl, CF3, halo, CN, NR7R8, SO2F6 and OC1-C6 alkyl, and b) Aromatic Heterocycle, wherein said Aromatic Heterocycle is selected from pyridy !, pyrazinyl, pyrimidinyl,, quinolinyl, quinoxalinyl ;. isoxazolyl and pyrazolyl, each aromatic heterocycle optionally substituted by 1-3 groups each independently selected from C1-C6 alkyl, SR6, SO2R6, NH2, CF3, halo, OH, OC1-C6 alkyl, NR7R8 wherein R8 may be optionally substituted by NH2, phenyl or Heterocycle, and OPh wherein Ph may be optionally substituted by 1-3 groups each independently selected from halo and C1-C6 alkyl.

More preferably R'is selected from : a) phenyl, which is optionally substituted by 1-3 groups each independently selected from C1-C6 alkyl, CF3, halo, CN, NR'R8, S02Rs and OC1 C6 alkyl, and b) Aromatic Heterocycle, wherein said Aromatic Heterocycle is selected from : i) pyridyl, which is optionally substituted by 1-3 groups each independently selected from C1-C6 alkyl, SO2R6, NH2, CF3, CN, halo, OH, OC1-C6 alkyl, NR7R8 wherein R8 may be

optionally substituted by NH2, phenyl or Heterocycle, and OPh wherein Ph may be optionally substituted by 1-3 groups each independently selected from halo and C1 C6 alkyl ; ii) pyrimidinyl, which is optionally substituted by 1-3 groups each independently selected from C1-C6 alkyl, SO2R6, NH2, CF3, CN, halo, OH, OC1-C6 alkyl, NR7R8 wherein R3 may be optionally substituted by NH2, phenyl or Heterocycle, and OPh wherein Ph may be optionally substituted by 1-3 groups each independently selected from halo and Ci-Ce alkyl ; iii) pyrazinyl, which is optionally substituted by 1-3 groups each independently selected from C1-C6 alkyl, NH2, SR6 and halo ; iv) quinoliny ! ; v) quinoxalinyl, which is optionally substituted by OH; vi) isoxazolyl, which is optionally substituted by 1-3 groups each independently selected from : C1-C6 alkyl ; and vii) pyrazol.

Yet more preferably R.'is phenyl, 2-or 3-pyridyl or 2, 4-pyrimidinyi, said moieties being optionally substituted by 1-3 groups each independently selected from C1-C6 alkyl, halo, OC1-C6 alkyl, CN, SO2R6, NHR7, NHCH2CH2NH2 and CF3.

Most preferably R1 is phenyl, 2-or 3-pyridyl or 2, 4-pyrimidinyl, said moieties being optionally substituted by 1-3 groups each independently selected from methyl, fluoro, chloro, methoxy, ethoxy, n-propoxy, CN, SO2CH3, NH2, NHCH3, NHCH2CH2NH2, and CFg.

Preferably R2 is selected from : a) phenyl, which is optionally substituted by C1 C6 alkyl, halo, OC1-C6 alkyl, OCF3, NR7R8, CF3 or SO2R6,

b) OPh, which is optionally substituted by C1 C6 alkyl or halo, c) cyclopropyl or 1-or 2-indanyl, d) pyrazolyl, which is optionally substituted by R6, e) R6, f) C (O) IN (CH3) 2, and g) 5-6 membered saturated ring containing 1 nitrogen atom, said ring being substituted by C (O) R6 ; More preferably R2 is selected from: a) phenyl, which is optionally substituted by methyl, halo, methoy, CF3 or S02CH3, b) cyclopropyl or 1-or 2-indanyl, c) pyrazolyl, which is optionally substituted by methyl, d) C (O) N (CH3) 2, and e) piperidinyl substituted by C (O) R6 ; Yet more preferably R2 is selected from : a) phenyl, which is optionally substituted by methyl, fluoro, chloro, methoxy, CF3 or SO2CH3, b) pyrazolyl, which is optionally substituted by methyl, and c) C(O)N(CH3)2.

Most preferably R2 is phenyl, para-fluorophenyl, para-chlorophenyl, para-methylphenyl, 2, 5-dimethylphenyl, o-methylphenyl and para-methoxyphenyl.

Preferably R3 is selected from : a) phenyl, said phenyl being optionally fused to Heterocycle and said phenyl or said fused phenyl being optionally substituted by 1-3 groups each independently selected from C1-C6 alkyl, halo, CN and OC1-C6 alkyl, b) R6,

c) cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl, which is optionally substituted by C1 C6 alkyl ; and d) Aromatic Heterocycle, wherein said Aromatic Heterocycle may be defined as a 5-6 membered aromatic heterocycle containing 1 or 2 nitrogen atoms, said ring optionally fused with a phenyl or a 3-8 membered cycloalkyl group.

More preferably R is selected from: a) phenyl, said phenyl being optionally fused to 1,4-dioxan and said phenyl or said fused phenyl being optionally substituted by 1-3 groups each independently selected from C1-C6 alkyl, halo, CN and OC1-C6 alkyl; b) Rs, c) cyclopropyl, which is optionally substituted by C1-C6 alkyl ; and d) Aromatic Heterocycle, wherein said Aromatic Heterocycle is selected from pyrazolyl or pyridyl, both optionally substituted by C1 C6 alkyl.

Yet more preferably R3 is selected from: a) phenyl, said phenyl being optionally fused to 1,4-dioxan and said phenyl or said fused phenyl being optionally substituted by 1-2 groups each independently selected from methyl, methoxy, ethoxy, fluoro, chloro and CN ; b) isopropyl ; c) cyclopropyl ; and d) pyrazolyl and pyridyl, both optionally substituted by methyl.

Most preferably R3 is selected from 3-methoxyphenyl and 1, 4-benzodioxanyl.

Preferably R4 iS H.

Preferably R5 is selected from: CONH2, CONHR, CONR6R6 and R

More preferably R5 is CONH2 or CH3.

Most preferably R5 is CONH2.

Preferably R6 is methyl.

Preferably x is 1.

Preferably y is 0.

Preferably z is 0 or 1.

Most preferably z is 0.

Preferably, Aromatic Heterocycle may be defined as a 5-6 membered aromatic heterocycle containing 1-3 heteroatoms each independently selected from N, O and S, said ring optionally fused with a phenyl or a 3-8 membered cycloalkyl group.

More preferably, Aromatic Heterocycle may be defined as a 5-6 membered aromatic heterocycle containing 1-2 heteroatoms each independently selected from N, O and S, said ring optionally fused with a phenyl.

Preferably, Aromatic Heterocycle is selected from pyridyl, pyrazinyl, pyrimidinyl, quinolinyl, quinoxalinyl, isoxazolyl and pyrazolyl.

Heterocycle is a 5-8 membered saturated or partially saturated ring containing 1-3 heteroatoms each independently selected from N, 0 and S, said ring optionally fused with phenyl.

Preferably, Heterocycle is a 5-6 membered saturated ring containing 1-3 heteroatoms each independently selected from N, O and S.

Preferably, Heterocycle is a 5-6 membered saturated ring containing 1 nitrogen atom.

All of the above reactions and the preparations of novel starting materials using in the preceding methods are conventional and appropriate reagents and reaction conditions for their performance or preparation as well as procedures for isolating the desired products will be well-know to those skilled in the art with reference to literature precedents and the Examples and Preparations hereto.

Compounds of formula (I) where R1, R2, R3 and y are as described above, x is 1, z is 0, R4 is H and R5 is CONH2 may be prepared by the following process as described in Scheme 1 :

Scheme 1 Compounds of formula (I) may be prepared by reacting compounds of formula (II), (III), (IV) and (V), where R7 may be H or Ph, under the conditions of process step (a) Ugi 4 component condensation-the acid, amine, aldehyde

and isonitrile are reacted together to give the desired compounds using the method of Keating, T. A. and Armstrong, R. W., J. Am. Chem. Soc. (1995), 117 (29), 7842-3.

Typicallv-the acid, amine, aldehyde and isonitrile are stirred together in a suitable solvent such as methanol, ethanol, THF, Et20, DME, DMF, DMSO at a temperature of 0 °C to the reflux temperature of the solvent for 1-48 hours.

The mixture is then treated with an acid such as HCI, H2SO4, AcOH in a suitable solvent such as methanol, ethanol, THF, Et2O, DME, DMF, DMSO at a temperature of 0 °C to the reflux temperature of the solvent for an additional 1- 48 hours.

Preferably - a mixture of the acid, 1.1 equivalents of the amine and 1.0 equivalents of the aldehyde in methanol was treated with 1.0 equivalents of (4- isocyano-3-cyclohexen-1-yl.)-benzene (Baldwin, J., E.; O'Neil, It A. Tetrahedron Lett., (1990), 31 (14), 2047-50) and the mixture was stirred at room temperature for 18 hours. The mixture was then heated to 50°C for 4 hours. The cooled mixture was treated with cHCI in THF (7% by volume) and stirred at room temperature for another 18 hours.

Compounds of formula (VI) and (Vil) are also produced as by products of process step (a)

Compounds of formula (VI) may be converted into compounds of formula (Vil) and subsequently transformed into compounds of formula (I).

It will be understood by one skilled in the art, that the Ugi 4 component condensation sometimes forms the acid (VH) directty without the intermediacy of the ester (VI). This is believed to be due to the formation of a munchnone intermediate which is trapped by water, Keating, T. A. ; Armstrong, R. W. J. Am. Chem. Soc. (1996) 118, 2574. The relative proportions depend primarily upon the starting materials used. However, certain conditions may be used in process step (a) to increase the relative proportions of (Vil) to (VI) : Typically - the acid, amine, aldehyde and isonitrile are stirred together in a suitable solvent such as methanol, ethanol, THF, Et2O, DME, DMF, DMSO at a temperature of 0 °C to the reflux temperature of the solvent for 1-48 hours.

The mixture is treated with an acid such as HCI, H2SO4, AcOH or acid chloride such as acetyl chloride in a suitable alcoholic solvent such as MeOH or EtOH, the choice of alcohol dictating the ester that is ultimately formed Preferably-a mixture of the acid, 1, 1 equivalents of the amine and 1.0 , equivalents of the aldehyde in methanol was treated with 1. 0 equivalents of the isonitrile and the mixture was stirred at room temperature for 18 hours. The mixture was s then heated to 50°C for 4 hours. The mixture is treated with 5. 0 equivalents of acetyl chloride and heating continued for 4 hours.

As discussed above, compounds of formula (I) where R1, R2 and R3 are as described above,) is 1, y is 0, z is 0, R4 is H and R5 is CONHR6 or CONH2 may be prepared from compounds of formula (VI) and (Vil) by the following process as described in Scheme 2:

Scheme 2 Compounds of formula (Vil) may be prepared by reacting compounds of formula (VI), under the conditions of process step (b) Ester hydrolysis-the ester can be treated with either an acid or a base, with heating in a suitable

solvent to effect the hydrolysis. Alternatively, if R7 = benzyl, the ester can be removed by hydrogenolysis using an appropriate catalyst.

Typically-the ester is treated with a metal hydroxide (Li, Na, K) in an aqueous solvent, MeOH, EtOH, THF, dioxan at a temperature of 0 °C to the reflux temperature of the solvent for 1-48 hours.

Preferably-a methanolic solution of the ester was stirred at room temperature for 18 hours in the presence of approximately 3 equivalents of aqueous sodium hydroxide.

Compounds of formula (I) may be prepared by reacting compounds of formula (Vil), under the conditions of process step (c) Amide bond formation-such reactions may be carried out under a wide variety of conditions well known to the skilled man.

Tvpicallv-the carboxylic acid may be activated by treatment with an agent such as CDI, TFFH, or a combination of reagents such as PyAOP and HOAt, or by the intermediacy of the acid chloride, for example prepared by the use of oxalyl chloride and catalytic DMF. Alternatively, the reaction may be carried out by the addition of a peptide coupling agent such as HATU, or HBTU, or DCC or DIC to a mixture of the acid and amine. The reaction is carried out in a suitable solvent such as DCM, pyridine, DMF, DMA or NMP at a temperature of 0 °C to the reflux temperature of the solvent.

Preferably-equimolar amounts of the acid and amine, 1.1 equivalents of HBTU and 2-4 equiva) ents of D ! PEA in DMF were stirred at room temperature for 18 hours. Compounds of formula (I) where R1, R2, R3 and y are as described above, x is 1, z is 0, R4 is H and R5 is CONHR6 may be prepared by the following process as described in Scheme 3:

Scheme 3 Compounds of formula (I) may be prepared by reacting compounds of formula (II), (III), (IV) and (VIII), where R8 may be H or Ph, under the conditions of process step (a) Ugi 4 component condensation as described herein.

Typically - the acid, amine, aldehyde and isonitrile are stirred together in a suitable solvent such as methanol, ethanol, THF, Et20, DME, DMF, DMSO at a temperature of 0 °C to the reflux temperature of the solvent for 1-48 hours.

Preferably - a mixture of the acid, 1. 1 equivalents of the amine and 1.0 equivalents of the aldehyde in methanol was treated with 1.0 equivalents of the isonitrile and the mixture was stirred at room temperature for 18 hours. The mixture was then heated to 500C for 4 hours.

A variety of methods are obvious to one skilled in the art for the preparation of isonitriles of formula (VIII). For example the isothiocyanate can be converted into the corresponding isonitrile using 3-methyl-2-phenyl- [1,3, 2] oxazaphospholidine according to the method of Mukaiyama, T; Yokota, Y. Bull. Chem. Soc. Jpn. , (1965) 38, 858 or the polymer-supported equivalent of Ley, S. V.; Taylor, S. J., Bioorg. Med. Chem. Lett. (2002) 12 (14), 1813.

Alternative preparations of isonitriles include the methods of Weber, W. P.; Gokel, G. W., Tet. Lett. (1972) 1637, of Ugi, I Angew. Chem. Int. Ed. Engl.

(1965) 4, 472 and of Creedon, S. M. et al, J. Chem. Soc. Perkin Trans. 1 (1998), 1015.

Compounds of formula (I) where R1, R2, R3, R4, z and y are as described above, x is 1 and R5 is CONR6R6 may be prepared by the following process as described in Scheme 4:

Scheme 4

Compounds of formula (XI) may be prepared by reacting compounds of formula (IX) and (X), under the conditions of process step (d) Reductive amination- dehydration of an amine and aidehyde followed by reduction of the imine so formed, by a suitable metal hydride reducing agent, usually requiring Lewis acid catalysis in a suitable solvent at room temperature Tvpically-the amine and aldehyde are mixed together in a suitable solvent such as methanol, ethanol, THF, Et20, DCM or DCE and are treated with a suitable reducing agent, such as sodium triacetoxyborohydride or sodium cyanoborohydride and a catalytic quantity of acetic acid, and then stirred at room temperature for 1-48 hours. Alternatively, the amine and aldehyde are premixed for a time of 1-24 hours in a, suitable solvent such as methanol, elthanol, THF, Et2O, DCI\/I or DCE, followed by the reducing agent such as sodium borohydride, sodium triacetoxyborohydride, sodium cyanoborohydride or lithium aluminium hydride and stirring continued for 1-48 hours at a temperature of 0 °C to the reflux temperature of the solvent.

Preferably-a mixture of the amine, 1. 05 equivalents of the aldehyde and 1.3 equivalents of sodium triacetoxyborohydride in DCM and catalytic acetic acid was stirred at room temperature for 18 hours.

Compounds of formula (XII) may be prepared by reacting compounds of formula () C9), under the conditions of process step (c) Amide bond formation as described herein.

Preferably-a mixture of the acid and 2.0 equivalents of oxalyl chloride with catalytic DMF in DCM was stirred at 0 °C for 2 hours. The solvent was removed in vacuo and the resultant crude acid chloride treated with 1. 0 equivalents of the amine and 4 equivalents of DIPEA in DCM stirring at room temperature for 18 hours.

Compounds of formula (XIII), may be prepared by reacting compounds of formula (XII), under the conditions of process step (b) Ester hydrolysis as described herein.

Compounds of formula (I) may be prepared by reacting compounds of formula (XIV), where R9 and R'° are C1-6 alkyl, or may link together to form a 4-7 containing nitrogen heterocycle, and (XIII), under the conditions of process step (c) Amide bond formation as described herein.

Compounds of formula (I) where R1, R2, R3, R4, z and y are as described above, x is 1 and R5 is CONR6R6 may be prepared by the following process as described in Scheme 5:

Scheme 5

Compounds of formula (XVII) may be prepared by reacting compounds of formula (XVI), where R6 is as defined herein, and (XV), where P is a suitable nitrogen protecting group, under the conditions of process step (c) Amide bond formation as described herein.

Suitable nitrogen protecting groups are well described in the art and for example can be found in references such as Greene T. W., Wuts, P. G. M.

Protective Groups in Organic Synthesis, Wiley-Interscience and Kocienski, P. J.

Protecting Groups, Thieme.

Compounds of formula (XVIII) may be prepared by reacting compounds of formula (XVII), under the conditions of process step (e) Removal of a nitrogen protecting group.

The conditions required for removal of the protecting group are often specific to that protecting group; conditions for their removal may be found in references such as Greene T. W., Wuts, P. G. M. Protective Groups in Organic Synthesis, Wiley-Interscie. nce and Kocienski, P. J. Protecting Groups, Thieme. If P is BOC, deprotection is acid catalysed removal of protecting group using a suitable solvent at room temperature Typically-the protected amine is treated with an excess of an acid such as HCI or TFA at room temperature in a solvent such as 1, 4-dioxane, ethyl acetate, DCM or THF.

Preferably-the amine in dichloromethane was treated with 4N HCI in 1, 4- dioxane and stirred at room temp for 18 hours.

Compounds of formula (XIX) may be prepared by reacting compounds of formula (iX) and (XVIII), under the conditions of process step (d) Reductive amination as described herein.

Compounds of formula (I) may be prepared by reacting compounds of formula () and (XIX), under the conditions of process step (c) Amide bond formation as described herein Compounds of formula (I) where R1, R2, R3, R4, z and y are as described above, x is 1 and R5 is C1-6 alkyl may be prepared by the following process as described in Scheme 6:

Scheme 6 Compounds of formula (XIX) may be prepared by reacting compounds of formula (XVIII) and (IX), under the conditions of process step (d) Reductive amination as described herein..

Compounds of formula (1) may be prepared by reacting compounds of formula (XIX) and (II), under the conditions of process step (c) Amide bond formation as described herein.

Compounds of formula (I), wherein R1 is phenyl or Aromatic Heterocycle, comprising an SO2Me substituent may be prepared by oxidation of the corresponding compound of formula ( !) comprising an SMe substituent.

Typically, the oxidation is carried out by addition of an oxidant to the sulfide in a solvent at ambient temperature. Preferably, the solvent is dichlorornethane and the oxidant is 3-chloroperoxybenzoic acid.

Compounds of formula (I), wherein Rr is phenyl or Aromatic Heterocycle, comprising an NR7R8 substituent may be prepared by reaction of the corresponding compound of formula (I) comprising an SO2Me substituent with an amine, HNR7R8. Typically, the reaction is carried out by addition of the

amine to the sulfone in an organic solvent at a temperature of 0°C, followed by warming to room temperature for 2 hours. Preferably, the solvent is THF.

The compounds of the invention are useful because they have pharmacological activity in mammals, including humans. More particularly, they are useful in the treatment or prevention of a disorder in which elevated levels of oxytocin or an excessive response to a normal level of oxytocin are implicated. Disease states that may be mentioned include sexual dysfunction, particularly premature ejaculation, preterm labour, complications in labour, appetite and feeding disorders, obesity, benign prostatic hyperplasia, premature birth, dysmenorrhoea, congestive heart failure, arterial hypertension, liver cirrhosis, nephrotic hypertens, ion, occular hypertension, obsessive compulsive disorder and neuropsychiatric disorders.

Sexual dysfunction (SD) is a significant clinical problem which can affect both mates and females. The causes of SD may be both organic as well as psychological. Organic aspects of SD are typically caused by underlying vascular diseases, such as those associated with hypertension or diabetes rnellitus, by prescription medication and/or by psychiatric disease such as depression. Physiological factors include fear, performance anxiety and interpersonal conflict. SD impairs sexual performance, diminishes self-esteem and disrupts personal relationships thereby inducing personal distress. In the clinic, SD disorders have been divided into female sexual dysfunction (FSD) disorders and male sexual dysfunction (fdSD) disorders (Melman et al 1999 J.

Urology 161 5-11). FSD is best defined as the difficulty or inability of a woman to find satisfaction in sexual expression. Male sexual dysfunction (MSD) is generally associated with either erectile dysfunction, also known as male erectile dysfunction (MED) and/or ejaculatory disorders such as premature ejaculation, anorgasmia (unable to achieve orgasm) or desire disorders such as hypoactive sexual desire disorder (lack of interest in sex).

PE is a relatively common sexual dysfunction in men. It has been defined in several different ways but the most widely accepted is the Diagnostic and Statistical Manual of Mental Disorders IV one which states: "PE is a lifelong persistent or recurrent ejaculation with minimal sexual stimulation before, upon or shortly after penetration and before the patient wishes it. The clinician must take into account factors that affect duration of the excitement phase, such as age, novelty of the sexual partner or stimulation, and frequency of sexual activity. The disturbance causes marked distress of interpersonal difficulty." The International Classification of Diseases 10 definition states: "There is an inability to delay ejaculation sufficiently to enjoy lovemaking, manifest as either of the following : (1) occurrence of ejaculation before or very soon after the beginning of intercourse (if a time limit is required : before or within 15 seconds of the beginning of intercourse); (2) ejaculation occurs in the absence of sufficient erection to make intercourse possible. The problem is not the result of prolonged abstinence from sexual activity" Other definitions, which have been used include classification on the following criteria: o Related to partner's orgasm Duration between penetration and ejaculation Number of thrust and capacity for voluntary control Psychological factors may be involved in PE, with relationship problems, anxiety, depression, prior sexual failure all playing a role.

Ejaculation is dependent on the sympathetic and parasympathetic nervous systems. Efferent impulses via the sympathetic nervous system to the vas deferens and the epididymis produce smooth muscle contraction, moving sperm into the posterior urethra. Similar contractions of the seminal vesicles, prostatic glands and the bulbouretheral glands increase the volume and fluid content of semen. Expulsion of semen is mediated by efferent impulses originating from the nucleus of Onuf in the spinal cord, which pass via the parasympathetic nervous system and cause rhythmic contractions of, the bulbocavernous, ischiocavernous and pelvic floor muscles. Cortical control of ejaculation is still under debate in humans. In the rat the medial pre-optic area and the paraventricular nucleus of the hypothalamus seem to be involved in ejaculation.

Ejaculation comprises two separate components-emission and ejaculation.

Emission is the deposition of seminal fluid and sperm from the. distal epididymis, vas deferens, seminal vesicles and prostrate into the prostatic urethra. Subsequent to this deposition is the forcible expulsion of the seminal contents from the urethra meatus. Ejaculation is distinct from orgasm, which is purely a cerebral event. Often the two processes are coincidental.

A pulse of oxytocin, in, peripheral. serum accompanies ejaculation in mammals.

In man oxytocin but not vasopressin plasma concentrations are significantly raised at or around ejaculation. Oxytocin does not induce ejaculation itself ; this process is 100% under nervous control via al-adrenoceptor/sympathetic nerves originating from the lumbar region of the spinal cord. The systemic pulse of oxytocin may have a direct role in the peripheral ejaculatory response.

It could serve to modulate the contraction of ducts and glandular lobules throughout the male genital tract, thus influencing the fluid volume of different ejaculate components for example. Oxytocin released centraliy into the brain could influence sexual behaviour, subjective appreciation of arousal (orgasm) and latency to subsequent ejaculation.

Accordingly, one aspect of the invention provides for the use of a compound of formula (I) in the preparation of a medicament for the prevention or treatment of sexual dysfunction, preferably male sexual dysfunction, most preferably premature ejaculation.

It has been demonstrated in the scientific literature that the number of oxytocin receptors in a womans body increases during pregnancy, most markedly before the onset of labour. Without being bound by any theory it is known that the inhibition of oxytocin can assist in preventing preterm labour and in resolving complications in labour.

Accordingly, another aspect of the invention provides for the use of a compound of formula (I) in the preparation of a medicament for the prevention or treatment of preterm labour and complications in labour.

Oxytocin has a role in feeding ;, it stimulates a desire to eat. By inhibiting oxytocin it is possible to reduce the desire to eat. Accordingly oxytocin inhibitors are useful in treating appetite and feeding disorders. Further by reducing appetite, oxytocin inhibitors can help to treat obesity.

Accordingly, a further aspect of the invention provides for the use of a compound of formula (I) in the preparation of a medicament for the prevention or treatment of obesity, appetite and feeding disorders.

Oxytocin is implicated as one of the causes of benign prostatic hyperplasia (BPH). Analysis of prostate tissue have shown that patients with BPH have increased levels of oxytocin. Oxytocin antagonists can help treat this condition.

Accordingly, another aspect of the invention provides for the use of a compound of formula (I) in the preparation of a medicament for the prevention or treatment of benign prostatic hyperplasia.

It is to be appreciated that all references herein to treatment include curative, palliative and prophylactic treatment.

The compounds of the present invention may be coadministered with one or more agents selected from: 1) One or more SSRIS such as paroxetine, 3- [ (dimethylamino) methyl]-4- [4- (methylsulfanyl) phenoxy] benzenesulfonamide (Example 28, WO 0172687), 3- [(dimethylamino)methyl]-4-[3-methyl-4- (methylsulfanyl) phenoxy] benzenesulfonamide (Example 12, WO 0218333), N- methyl-N-({3-[3-methyl-4-(methylsulfanyl)phenoxy]-4-pyridiny l}methyl)amine (Example 38, PCT Application no PCT/IB02/01032).

2) One or more local anaesthetics; 3) One or more beta adrenoceptor agonists; 4) one or more a-adrenergic receptor antagonists (also known as a-adrenoceptor blockers, a-receptor blockers or a-blockers) ; suitable a1- adrenergic receptor antagonists mciude ; phentotamine, prazosin, phentolamine mesylate, trazodone alfuzosin, indoramin, naftopidil, tamsulosin, phenoxybenzamine, rauwolfa alkaloids, Recordati 15/2739, SNAP 1069, SNAP 5089, RS17053, SL 89. 0591, doxazosin, Example 19 of W09830560, terazosin and abanoquil ; suitable a2-adrenergic receptor antagonists include dibenarnine, tolazoline, trimazosin, efaroxan, yohimbine, idazoxan clonidine and dibenarnine; suitable non-selective a-adrenergic receptor antagonists include dapiprazole ; further a-adrenergic receptor antagonists are described in PCT application W099/30697 published on, 4th June 1998 and US patents : 4, 188, 390; 4,026, 894 ; 3,511, 836; 4, 315, 007; 3,527, 761; 3,997, 666; 2,503, 059; 4,703, 063; 3,381, 009; 4,252, 721 and 2, 599, 000 each of which is incorporated herein by reference; 5) one or more cholesterol lowering agents such as statins (e. g. atorvastatin/Lipitor - trade mark) and fibrates ; 6) one or more of vasoactive intestinal protein (VIP), VIP mimetic, VIP analogue, more particularly. mediated by one or more of the VIP receptor

subtypes VPAC1, VPAC or PACAP (pituitory adenylate cyclase activating peptide), one or more of a VIP receptor agonist or a VIP analogue (eg Ro-125-1553) or a VIP fragment, one or more of a a-adrenoceptor antagonist with VIP combination eg Invicorp, Aviptadil) ; 7) one or more of a serotonin receptor agonist, antagonist or modulator, more particularly agonists, antagonists or modulators for example 5HT1A (including VML 670), 5HT2A, 5HT2C, 5HT3,5HT6 and/or 5HT7 receptors, including those described in WO-09902159, WO-00002550 and/or WO- 00028993; 8) one or more NEP inhibitors, preferably wherein said NEP is EC 3. 4. 24. 11 and more preferably wherein said NEP inhibitor is a selective inhibitor for EC 3. 4.24. 11, more preferably a selective NEP inhibitor is a selective inhibitor for EC 3. 4. 24. 11, which has an IC50 of less than 100nM (e. g. ompatrilat, sampatrilat) suitable NEP inhibitor compounds are described in EP- A-1097719 ; IC50 values against NEP and ACE may be determined using methods described in published patent application EP1097719-A1, paragraphs [0368] to [0376]; 9) one or more of an agonist or modulator for vasopressin receptors, such as relcoraptan (SR 49059) 10) Apomorphine-teachings on the use of apomorphine as a pharmaceutical may be found in US-A-5945117 ; 11) Dopamine D2 agonists (e. g. Premiprixal, Pharmacia Upjohn compound number PNU95666); 12) Melanocortin receptor agonists (e. g. Metanotan ll) ; 13) PGE1 receptor agonists (e. g. alprostadil) ; 14) Mono amine transport inhibitors, particularly Noradrenaline Re-uptake Inhibitors (NRIs) (e. g. Reboxetine), other Serotonin Re-uptake Inhibitors (SR ! s) (e. g. paroxetine) or Dopamine Re-uptake Inhibitors (DRIs) ; 15) 5-HT1A antagonists (e. g. robalzotan) ; and 16) PDE inhibitors such as PDE2 (e. g. erythro-9- (2-hydroxyl-3-nonyl)- adenine) and Example 100 of EP 0771799-incorporated herein by reference) and in particular a PDE5 inhibitor such as the pyrazol [4,3-

d] pyrimidin-7-ones disclosed in EP-A-0463756 ; the pyrazol [4, 3- d] pyrimidin-7-ones disclosed in EP-A-0526004 ; the pyrazol [4,3- d] pyrimidin-7-ones disclosed in published international patent application WO 93/06104; the isomeric pyrazol [3, 4-d] pyrimidin-4-ones disclosed in published international patent application WO 93/07149 ; the quinazolin- 4-ones disclosed in published international patent application WO 93/12095 ; the pyrido [3,2-d] pyrimidin-4-ones disclosed in published international patent application WO 94/05661; the purin-6-ones disclosed in published international patent application WO 94/00453 ; the pyrazol [4,3-d] pyrimidin-7-ones disclosed in published international patent application WO 98/49166 ; the pyrazol [4, 3-d] pyrimidin-7-ones disclosed in published international patent application WO 99/54333 ; the pyrazol [4,3-d] pyrimidin-4-ones disclosed in EP-A-0995751; the pyrazol [4, 3-d] pyrimidin-7-ones disclosed in published international patent application WO 00/24745 ; the pyrazol [4,3-d] pyrimidin-4-ones disclosed in EP-A-0995750 ; the compounds disclosed in published international application W095/19978 ; the compounds disclosed in published international application WO 99l24433 and the compounds disclosed in published international application WO 93/07124.

The pyrazol [4, 3-d] pyrimidin-7-ones disclosed in published international application WO 01/271112 ; the pyrazol [4,3-d] pyrimidin-7-ones disclosed in published international application WO 01/27113 ; the compounds disclosed in EP-A-1092718 and the compounds disclosed in EP-A-1092719.

Preferred PDE5 inhibitors for use with the invention: <BR> <BR> <BR> <BR> <BR> <BR> <BR> 5- [2-ethoxy-5- (4-methyl-1-piperazinylsulphonyl) phenyl]-1-m, ethyl-3-n-propyl-1, 6- dihydro-7H-pyrazolo [4, 3-d] pyrimidin-7-one (sildenafil) also known as 1-[[3-(6, 7- dihydro-1-methyl-7-oxo-3-propyl-1 H-pyrazolo [4, 3-d] pyrimidin-5-yl)-4- ethoxyphenyl] sulphonyB]-4-methylpiperazine (see EP-A-0463756) ;

5- (2-ethoxy-5-morpholinoacetylphenyl)-1-methyl-3-n-propyl-1, 6-dihydro-7H- pyrazol [4, 3-d] pyrimidin-7-one (see EP-A-0526004) ; <BR> <BR> <BR> <BR> <BR> 3-ethyl-5- [5- (4-ethylpiperazin-1-ylsulphonyl)-2-n-propoxyphenyl]-2- (pyridin-2- yl) methyl-2, 6-dihydro-7H-pyrazolo [4, 3-d] pyrimidin-7-one (see W098/49166) ; <BR> <BR> <BR> <BR> <BR> 3-ethyl-5- [5- (4-ethylpiperazin-1-ylsulphonyl)-2- (2-methoxyethoxy) pyridin-3-yl]-2- (pyridi 2-yl)methyl-2,6-dihydro-7H-pyrazolo[4,3-d]pyrimidin-7-one (see W099/54333) ; (+)-3-ethyl-5- [5- (4-ethylpiperazin-1-y ! sulphonyl)-2- (2-methoxy-1 (R)- methylethoxy) pyridin-3-yl]-2-methyl-2, 6-dihydro-7H-pyrazolo [4, 3-d] pyrimidin-7- one, also known as 3-ethyl-5-{5-[4-ethylpiperazin-1-ylsulphonyl]-2-([(1R)-2- <BR> <BR> <BR> methoxy-1-methylethyl] oxy) pyridin-3-yl}-2-methyl-2, 6-dihydro-7H-pyrazolo [4, 3-<BR> <BR> <BR> <BR> djpyrimidin-7-one (see W099/54333) ;.

5-[2-ethoxy-5-(4-ethylpiperazin-1-ylsulphonyl)pyridin-3-e thyl-2-[2- methoxyethyl]-2, 6-dihydro-7H-pyrazolo[4,3-d]pyrimidin-7-one, also known as 1- {6-ethoxy-5- [3-ethyl-6, 7-dihydro-2- (2-methoxyethyl)-7-oxo-2H-pyrazolo [4,3- d]pyrimidin-5-yl]-3-pyridylsulphonyl}-4-ethylpiperazine (see WO 01/27113, Example 8); 5-[2-iso-Butoxy-5-(4-ethylpiperazin-1-ylsulphonyl)pyridin-3- yl]-3-ethyl-2-(1- <BR> <BR> methylpiperidin-4-yl)-2, 6-dihydro-7H-pyrazolo [4, 3-d] pyrimidin-7-one (see WO 01/27113, Example 15) ; 5-[2-Ethoxy-5-(4-ethylpiperazin-1-ylsulphonyl)pyridin-3-yl]- 3-ethyl-2, 6- dihydro-7H-pyrazolo [4, 3-d] pyrimidin-7-one (see WO 01/27113, Example 66); 5-(5-Acetyl-2-propoxy-3-pyridinyl)-3-ethyl-2-(1-isopropyl-3- azetidinyl)-2, 6- dihydro-7H-pyrazolo[4,3-d]pyrimidin-7-one (see WO 01/27112, Example 124) ;

5- (5-Acetyl-2-butoxy-3-pyridinyl)-3-ethyl-2- (1-ethyl-3-azetidinyl)-2, 6-dihydro-7H- pyrazol [4, 3-d] pyrimidin-7-one (see WO 01/27112, Example 132) ; (6R, 12aR)-2, 3,6, 7, 12, 12a-hexahydro-2-methyl-6- (3, 4-methylenedioxyphenyl)- pyrazin [2', 1' : 6,1] pyrido [3,4-b] indole-1, 4-dione (IC-351), i. e. the compound of examples 78 and 95 of published international application W095/19978, as well as the compound of examples 1,3, 7 and 8 ; <BR> <BR> <BR> <BR> <BR> <BR> 2- [2-ethoxy-5- (4-ethyl-piperazin-1-yl-1-sulphohyl)-phenyl]-5-methyl-7-prop yl-3H- imidazo [5,1-f] [1,2, 4] triazin-4-one (vardenafil) also known as 1-[[3-(3,4-dihydro- 5-methyl-4-oxo-7-propylimidazo [5, 1-f]-as-triazin-2-yl)-4- ethoxyphenyl] s. ulphonyl]-4-ethylpiperazine, i. e. the compound of examples 20, 19,337 and 336 of published international application WO99/24433 ; and the compound of example 11 of published international application WO93/07124 (EISAI) ; and compounds 3 and 14 from Rotella D P, J. Med Chem., 2000,43, 1257.

Still further PDE5 inhibitors for use with the invention include : 4-bromo-5- (pyridylmethylamino)-6[3-(4-chlorophenyl)-propoxy]-3(2H)pyri dazinone ; 1-[4- [(1,3-benzodioxol-5- ylmethyl)amiono]-6-chloro-2-quinozolinyl]-4-piperidine- carboxylic acid, monosodium salt; (+)-cis-5, 6a, 7,9, 9, 9a-hexahydro-2-[4 (trifluoromethyl)-phenylmethyl-5-methyl-cyclopent-4, 5] imidazo [2,1-b] purin- 4 (3H) one; furazlocillin ; cis-2-hexyl-5-methyl-3, 4, 5, 6a, 7,8, 9, 9a- octahydrocyclopent [4, 5]-imidazo [2,1-b] purin-4-one; 3-acetyl-1-(2-chlorobenzyl)- 2-propylindole-6-carbonylate ; 3-acetyl-1-(2-chlorobenzyl)-2-propylindole-6- carboxylate ; 4-bromo-5-(3-pyridylmethylamino)-6-(3-(4-chlorophenyl)propox y)- 3- (2H) pyridazinone ; G-methyl-5 (5-morpholinoacetyl-2-n-propoxyphenyl)-3-n- propyl-1, 6-dihydro-7H-pyrazolo (4,3-d) pyrimidin-7-one ; 1- [4- [ (1, 3-benzodioxol-5- ylmethyl)amino]-6-chloro-2- quinazolinyl]-4-piperidinecarboxylic acid, monosodium salt ; Pharmaprojects No. 4516 (Glaxo Wellcome) ; Pharmaprojects

No. 5051 (Bayer) ; Pharmaprojects No. 5064 (Kyowa Hakko; see WO 96/26940); Pharmaprojects No. 5069 (Schering Plough) ; GF-196960 (Glaxo Welcome) ; E-8010 and E-4010 (Eisai); Bay-38-3045 & 38-9456 (Bayer) and Sch-51866.

The contents of the published patent applications and journal articles and in particular the general formulae of the therapeutically active compounds of the claims and exemplified compounds therein are incorporated herein in their entirety by reference thereto.

More preferred PDE5 inhibitors for use with the invention are selected from the group : <BR> <BR> <BR> 5- [2-ethoxy-5- (4-methyl-1-piperazinylsulphonyl) phenyl]-1-methyl-3-n-propyl-1, 6- dihydro-7H-pyrazolo [4, 3-d] pyrimidin-7-one (sildenafil) ; (6R, 12aR)-2, 3,6, 7,12, 12a-hexahydro-2-methyl-6- (3, 4-methylenedioxyphenyl)- pyrazino [2', 1' : 6,1] pyrido [3,4-b] indole-1, 4-dione (IC-351) ; 2-[2-ethoxy-5-(4-ethyl-piperazin-1-yl-1-sulphonyl)-phenyl]-5 -methyl-7-propyl-3H- imidazo [5,1-f] [1, 2, 4] triazin-4-one (. vardenafil) ; and 5- [2-ethoxy-5- (4-ethylpiperazin-1-ylsulphonyl) pyridin-3-yl]-3-ethyl-2- [2- methoxyethyl]-2, 6-dihydro-7H-pyrazolo [4,3-d] pyrimidin-7-one or 5- (5-Acetyl-2- butoxy-3-pyridinyl)-3-ethyl-2- (1-ethyl-3-azetidinyl)-2, 6-dihydro-7H-pyrazolo [4, 3- d] pyrimidin-7-one and pharmaceutical acceptable salts thereof.

A particularly preferred PDE5 inhibitor is 5- [2-ethoxy-5- (4-methyl-1- piperazinylsulphonyl) phenyl]-1-methyl-3-n-propyl-1, 6-dihydro-7H-pyrazolo [4,3- d] pyrimidin-7-one (sildenafil) (also known as 1-[[3-(6,7-dihydro-1-methyl-7-oxo- 3-propyl-1 H-pyrazolo [4, 3-d] pyrimidin-5-yl)-4-ethoxyphenyl] sulphonyl]-4- methylpiperazine) and pharmaceutical acceptable salts thereof. Sildenafil citrate is a preferred salt.

The compounds of the formula (I) can also be administered together with A. Oxytocin Receptor Ligand Binding IC50 Assay i) Buffers Cell Growth Medium Hams F12 Nutrient Mix 10 % FCS 2 mM L-Glutamine 400 Rg/ml G418 15 mM HEPES Membrane Prep. Buffer 50 mM Tris-HCI, pH7.8 10 mM Mg Cl2 Protease Inhibitors Freezing Buffer 50 mM Tris-HCl, pH7.8 10 MM Mg C12 20% Glycerol Assay Medium 50 mM Tris-HCl, pH7.8 10 mM Mg Cl2 0.25% BSA Max. 0.5 µM (arg8)-vasotocin made in 2.5 % DMSO/50 mM Tris- HCL, pH 7.8, 10 mM MgCl2 Min. 2.5 % DMSO/50 mM Tris-HCL, pH 7.8, 10 mM MgCl2 ii) Compound Dilution (Final concentration of 10 µM in the assay)

a) HTA stock compounds at 4 mM in 100 % DMSO b) Dilute compounds to 200 RM in dH2O. c) Further dilute compounds to 100 µM in 100 mM Tris-HCl, pH 7.8, 20 mM MgCl2. This gives final concentrations of 2.5 % DMSO, 50 mM Tris-HCl, ph 7.8, 10 mM MgCl2. d) Using the diluted stock, prepare 1 : 2 dilutions over 10 points in 50 mM Tris-HCI, pH 7.8, 10 mM MgCI2, 2.5% DMSO with the TECAN Genesis. e) Dispense 10 ul of the compound into a 384 well Optiplate according to the plate layout required for analysis by ECADA leaving space for the standard (arg8)-vasotocin IC50. These plates can be stored at 4°C. f) On the day of the assay, add 10 gui of Max. to the + wells and 10 ßl of Min. to the-wells, and a 1 : 2 serial dilution over 10 points in duplicate of the (arg8)-vasotocin with a top concentration of 100 nM (20 nM final). iii) Maintenance of the Oxytocin Receptor - CHO Cells @ The cell line is routinely maintained as a continuous culture in 50 mi growth medium in 225 cm2 flasks.

Cells are passaged by removing. the medium from the monolayer, washing with PBS and incubating. with Trypsin until cells show. signs of dissociation. After knocking the cells from the bottom of the flask, cells are resuspended in growth medium and seeded into 225 cm2 flasks at a concentration of 8x105 cells/flask.

IV) GROWTH OF CELLS IN ROLLER BOTTLES Cells are seeded into 10 x 850 cm2 roller bottles at a density of 6 i (~ o6 cells/bottle and are allowed to reach near confluence.

Cells are removed from the bottles using trypsin, as described above, and the cells are seeded into 100 x roller bottles (i. e. 1 : 10 split ratio).

Cells are again allowed to reach near confluence before removing the growth medium, adding 40 ml PBS/bottle and harvesting by scraping using the CellMate. The cell suspension is then centrifuged at 2000 rpm, washed in PBS, centrifuged again and pellets are frozen in aliquots at-80°C. v) Membrane Preparations Cell pellets are retrieved from the freezer, thawed on ice and resuspended in 3 ml of membrane prep buffer per ml packed cell volume.

The suspension is then homogenised using a mechanical homogeniser for several bursts of 5 secs on ice before centrifuging at 25, 000 x g for 30 mins.

After resuspending the pellet in 1 ml of freezing buffer per 1 ml. of the original packed cell volume the suspension is briefly homogenised to remove small lumps. Protein concentrations are then measured and the membrane suspension is finally frozen in aliquots at a minimum of 5 mg/mi at-80°C. vi) Assay Membranes are thawed on ice before diluting to 1 mg/ml in assay buffer.

SPA beads are resuspended at, 50 mg/ml in assay buffer. From these concentrations, beads are pre-coupled with membranes by incubating 30 µg of protein per mg of bead on a top-to-tail shaker for 2 hours at 4 °C. The bead/membranes are then centrifuged at 2000 rpm for 10 mins and the pellet is resuspended at 3 mg/mi.

All manipulations of the 1251-OVTA are carried out using tips that have been silanised using SigmaCote. All bottles and tubes are also silanised. The 125-I-OVTA is diluted in 1 ml assay buffer per 50 RCi of lyophilised ligand. A 5 µl sample is then counted in duplicate using liquid scintillation counting

(protocol 61 on Wallac Counter) and the concentration of the ligand is calculated (see example below). This is to overcome any loss of ligand due to stickiness. Using the measured concentration, the 125I-OVTA is diluted to 0.3 nM in assay buffer.

Example : If 5 pLi gives 500000 dpm and the specific activity of the ligand is 2200Ci/mmol then: Concentration = 500000/(2.2x2200x5) nM 20 ul of the bead/membrane preparation is added to the prepared Optiplates using the Multi-drop. The bead/membrane preparation is kept in suspension using a stirring flask. 20 µl of the 125I-OVTA is then added to each well of the Optiplate using the Multi-drop. Following a 4 hour incubation at room temperature, the plates are counted using the TopCount NXT for 30s/well.

The compounds of the present invention all exhibit better than 70% inhibition of oxytocin at 10µM.

2-Amino-N-[carbamoyl-(2,3-dihydro-benzo[1,4]dioxin-6-yl)- methyl]-4,6-dimethyl- N- (4-methyl-benzyl)-nicotinamide (Example 257) has a Ki of 9. 4nM <BR> <BR> (R or S)-2-Amino-N- [carbamoyl- { (3-methoxyphenyl)-methyl}]-4, 6-dimethyl-N- (4- methyl-benzyl)-nicotinamide (Example 258-single enantiomer) has a Ki of 9. 4nM The compounds of the formula (I) can be administered alone but will generally be administered in admixture with a suitable pharmaceutical excipient, diluent

or carrier selected with regard to the intended route of administration and standard pharmaceutical practice.

The present invention provides for a composition comprising a compound of formula (I) and a pharmaceutically acceptable diluent or carrier.

For example, the compounds of the formula (I) can be administered orally, buccally or sublingually in the form of tablets, capsules, ovules, elixirs, solutions or suspensions, which may contain flavouring or colouring agents, for immediate-, delayed-, modified-, sustained-, pulsed-or controlled-release applications.

Such tablets may contain excipients such as microcrystalline cellulose, lactose, sodium citrate, calcium carbonate, dibasic calcium phosphate and glycine, disintegrants such as starch (preferably corn, potato or tapioca starch), sodium starch glycollate, croscarmellose sodium and certain complex silicates, and <BR> <BR> <BR> <BR> granulation binders such as polyvinylpyrrolidone"hydroxypropylmethylcellulose (HPMC), hydroxypropylcellulose (HPC), sucrose, gelatin and acacia.

Additionally, lubricating agents such as magnesium stearate, stearic acid, glyceryl behenate and talc may be included.

Solid compositions of a similar type may also be employed as fillers in gelatin capsules. Preferred excipients in this regard include lactose, starch, a cellulose, milk sugar or high molecular weight polyethylene glycols. For aqueous suspensions and/or elixirs, the compounds of the formula (I) may be combined with various sweetening or flavouring agents, colouring matter or dyes, with emulsifying and/or suspending agents and with diluents such as water, ethanol, propylene glycol and glycerin, and combinations thereof.

The compounds of the formula (I) can also be administered parenterally, for example, intravenously, intra-arterially, intraperitoneally, intrathecally, intraventricularly, intraurethrally, intrasternally, intracranially, intramuscularly or

subcutaneously, or they may be administered by infusion techniques. For such parenteral administration they are best used in the form of a sterile aqueous solution which may contain other substances, for example, enough salts or glucose to make the solution isotonic with blood. The aqueous solutions should be suitably buffered (preferably to a pH of from 3 to 9), if necessary. The preparation of suitable parenteral formulations under sterile conditions is readily accomplished by standard pharmaceutical techniques well-known to those skilled in the art.

The compounds of formula (I) can also be administered intranasally or by inhalation and are conveniently delivered in the form of a dry powder inhaler or an aerosol spray presentation from a pressurised container, pump, spray, atomiser or nebuliser, with or without the use of a suitable propellant, e. g. dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, a hydrofluoroalkane such as 1,1, 1, 2-tetrafluoroethane (HFA 134A [trade mark] ) or 1, 1, 1, 2,3, 3, 3-heptafluoropropane (HFA 227EA [trade mark] ), carbon dioxide or other suitable gas. In the case of a pressurised aerosol, the dosage unit may be determined by providing a valve to deliver a metered amount. The pressurised container, pump, spray, atomiser or nebuliser may contain a solution or suspension of the active compound, e. g. using a mixture of ethanol and the propellant as the solvent, which may additionally contain a lubricant, e. g. sorbitan trioleate. Capsules and cartridges (made, for example, from gelatin) for use in an inhaler or insufflator may be formulated to contain a powder mix of a compound of the formula (I) and a suitable powder base such as lactose or starch.

Alternatively, the compounds of the formula (I) can be administered in the form of a suppository or pessary, or they may be applied topically in the form of a gel, hydrogel, lotion, solution, cream, ointment or dusting powder. The compounds of the formula (I) may also be dermally or transdermally administered, for example, by the use of a skin patch. They may also be administered by the pulmonary or rectal routes.

They may also be administered by the ocular route. For ophthalmic use, the compounds can be formulated as micronised suspensions in isotonic, pH adjusted, sterile saline, or, preferably, as solutions in isotonic, pH adjusted, sterile saline, optionally in combination with a preservative such as a benzylalkonium chloride. Alternatively, they may be formulated in an ointment such as petrolaturn.

For application topically to the skin, the compounds of the formula (I) can be formulated as a suitable ointment containing the active compound suspended or dissolved in, for example, a mixture with one or more of the following : mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene polyoxypropylene compound, emulsifying wax and water.

Alternatively, they can be formulated as a suitable lotion or cream, suspended or dissolved in, for example, a mixture of one or more of the following : mineral oil, sorbitan monostearate, a polyethylene glycol, iiquid paraffin, polysorlDate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.

The compounds of the formula (i) may also be used in combination with a cyclodextrin. Cyclodextrins are known to form inclusion and non-inclusion, complexes with drug molecules. Formation of a drug-cyclodextrin complex may modify the solubility, dissolution rate, bioavailability and/or stability property of a drug molecule. Drug-cyclodextrin complexes are generally useful for most dosage forms and administration routes. As an alternative to direct complexation with the drug the cyclodextrin may be used as an auxiliary additive, e. g. as a carrier, diluent or solubiliser. Alpha-, beta-and gamma- cyclodextrins are most commonly used and suitable examples are described in WO-A-91/11172, WO-A-94/02518 and WO-A-98/55148.

The invention is further illustrated by the following, non-limiting examples.

Abbreviations and Definitions: ArbocelTM Filtration agent, from J. Rettenmaier & Sohne, Germany Amberlyst# 15 ton exchange resin, available from Aldrich Chemical Company APCI Atmospheric Pressure Chemical lonisation atm Pressure in atmospheres (1 atm = 760 Torr = 101.3 kPa) BiotageT Chromatography performed using Flash 75 silica gel cartridge, from Biotage, UK BOC tert-Butyloxycarbonyl gròup br Broad c Concentration used for optical rotation measurements in g per 100ml (1 mg/mlisc0. 10) cat Catalytic d Doublet dd Doublet of doublets Degussa# 101 10 wt% palladium on activated carbon, Degussa type E101 available from. Aldrich Chemical Company Develosil Supplied by Phenomenex-manufactured by Nomura Combi-RP C3o Chemical Co. Composed of spherical silica particles (size 3 hplc column µm or 5 pm) which have a chemically bonded surface of C30 chains. These particles are packed into stainless steel columns. of dimensions 2 cm internal diameter and 25 cm long.

Dowex ton exchange resin, from Aldrich Chemical Company ee Enantiomeric excess HRMS High Resolution Mass Spectrocopy (electrospray ionisation positive scan) HyfloTM Hyflo supercel0, from Aldrich Chemical Company liq Liquid LRMS Low Resolution Mass Spectroscopy (electrospray or thermospray ionisation positive scan)

LRMS (ES-) Low Resolution Mass Spectroscopy (electrospray ionisation negative scan) m Multiplet m/z Mass spectrum peak MCITM gel High porous polymer, CHP20P 75-150µm, from Mitsubishi Chemical Corporation Phenomenex Supplied by Phenomenex. Composed of spherical silica Luna C18 hp ! c partictes (size 5 um or 10 pm) which have a chemically column bonded surface of C18 chains. These particles are packed into a stainless steel column of dimensions 2. 1 cm internal diameter and 25 cm long. psi Pounds per square inch (1 psi = 6.9 kPa) q Quartet Rf Retention factor on TLC s Singlet, Sep-Pake Reverse phase C18 silica gel cartridge, Waters Corporation Triplet TLC Thin Layer Chromatography Chemical shift Unless otherwise provided herein: PYBOPO means Benzotriazol-1-yloxytris(pyrrolidino) phosphonium hexafluorophosphate ; PyBrOP# means bromo-tris-pyrrolidino-phosphonium hexafluorophosphate ; CDl means N,N'-carbonyldiimidazole ; WSCDI means 1- (3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride ; Mukaiyama's reagent means 2-chloro-1-methylpyridinium iodide; DCC means N, N'-dicyclohexylcarbodiimide ; D ! PEA means diisopropylethylamine ; HATU means O-(7-azabenzotriazol-1-yl)-1, 1,3, 3 tetramethyluronium hexafluorophosphate ;

HBTU means O-benzotriazol-1-yl-N,N,N',N'-tetramethyluronium hexafluorophosphate ; HOAT means 1-hydroxy-7-azabenzotriazole ; HOBT means 1-hydroxybenzotriazole hydrate; Honig's base means N-ethyldiisopropylamine ; Et3N means triethylamine ; NMM means N-methylmorpholine ; NMP means 1-methyl-2-pyrrolidinone ; DMA means dimethylacetamide DME means ethylene glycol dimethyl ether; DMAP means 4-dimethylaminopyridine ; NMO means 4-methylmorpholine N-oxide ; KHMDS means potassium bis (trimethylsilyl) amide ; NaHMDS means sodium bis (trimethylsilyl) amide ; DIAD means diisopropyl azodicarboxylate ; DIC means 2-dimethylaminoisopropyl chloride hydrochloride ; DEAD means diethyl azodicarboxylate ; DIBAL means diisobutylaluminium hydride; Dess-Martin periodinane means 1,1, 1-triacetoxy-1, 1-dihydro-1, 2-benziodoxol- 3 (1H)-one ; TBDMS-Cl means tert-butyldimethylchlorosilane; TFFH means tetramethylfluoroformamidinium hexafluorophosphate ; TUS-CI means chlorotrimethylsilane ; BOC means tert-butoxycarbonyl ; CBz means benzyloxycarbonyl ; MeOH means methanol, EtOH means ethanol, and EtOAc means ethyl acetate ; THF means tetrahydrofuran, DMSO means dimethyl sulphoxide, and DCM means dichloromethane ; DMF means N, N-dimethylformamide ; AcOH means acetic acid, TFA means trifluoroacetic acid.

EXAMPLE 1 2-Amino-N-[2-amino-1-(2-methylphenyl)-2-oxoethyl]-N-(4- chlorobenzyl) nicotinamide

Solutions of 4-chlorobenzylamine (708mg, 5mmol) in methanol (1 Oml), followed by o-tolualdehyde (601 mg, 5mmol) in methane (1 Oml) then the compound from preparation 6 (916mg, 5mmol) in methanol/cyclohexane (95: 5, by volume) were added consecutively to a suspension of 2-aminonicotinic acid (691 mg, 5mmol) in methanol (20mi), and the mixture stirred at 50°C for 7 hours, then at room temperature for a further 18 hours. The reaction mixture was concentrated under reduced pressure, the, residue dissolved in a solution of hydrochloric acid in tetrahydrofuran (25moi, 0. 6M) and the reaction stirred at room temperature for 24 hours. The mixture was evaporated under reduced pressure, the residue suspended in dichloromethane (200m1), triethylamine added, until dissolution occurred, then the solution washed with saturated aqueous ammonium chloride solution (2x50ml). The organic solution was then dried (MgSO4) and evaporated under reduced pressure. The crude product was purified by column chromatography on silica gel using a gradient elution of dichloromethane : methanol : triethylamine (100: 0: 0 to 90: 10 : 1) to afford the title compound, 968mg.

1Hnmr (CDCl3, 400MHz) # : 2.15 (s, 3H), 4. 28 (d, 1H), 4. 73 (d, 1H), 5.60 (m, 3H), 6.48-6. 56 (m, 3H), 6.92-7. 19 (m, 5H), 7. 25 (s, 2H), 7. 41 (m, 2H), 8.06 (d, 1H).

LRMS : m/z (ES+) 409, 411 [MH+] Examples 2 to 218

Exemples 2 to 8

Solutions of 4-methylbenzylamine 950µl, 0. 6M in methanol), benzadiooane aldehyde (100µl, 0. 3M in methanol) and the isocyanate from preparation 6 (50µl, 0. 61i\/1 in methanol) were added to the appropriate acids (30µmol). The reactions were sealed and agitated at rt for 18 hours then heated at 50°C for 3 hours. The solvents were removed under reduced pressure, hydrochloric acid

in tetrahydrofuran (500µl, 0. 6M) was added, and the vessels were resealed and agitated again at room temperature for a further 24 hours. The solvents were removed under reduced pressure, the residues dissolved in dimethyl sulphoxide (500, u1) and purified by HPLC, using a Phenomonex Luna 150x10mm, 10µm column, in acetonitile : 0. 1% aqueous diethylamine, at 8mimis-', at 225nM, using the following gradient. Time (min) % acetonitrile 0.00-0. 50 5 0.50-0. 60 5-20 0. 60-6. 50 20-95 6.5-8. 5 95 8. 5-8.6 95-5

Ex. No. R1 Retention time/min 2 < 6. 163 CH - c3 F 3 6. 372 / 8. Cf-13 4 H3Cw 6. 355 1 caf3 5 F 5. 883 I./ if 6 6. 267 1 F F F 75. 988 F F 6. 4. 37 CHUG EXAMPLE 9

Example 9 was prepared from methyl (3-carboxaldehyde-2-pyridine) carboxylate, 4-methylbenzylamine, 6-methyl-2-pyridine carboxylic acid and the isocyanate from preparation 6, following a similar procedure to that described in example 2.

Retention time/min : 6. 647 Examples 10 to 24 The required acids (R1CO2H) (200µl, 0. 25m solution in methanol), (1-methyl-2- pyrrolidinone added to aid dissolution where necessary), followed by the amines (R2NH2) (100µl, 0.5 M solution in methanol) were transferred to 96 well

plates. Solutions of the appropriate aldehydes R3COH (100µl, 0.5 M solution in methanol) were prepared and then added, followed by a solution of the compound from preparation 6 (100µl, 0. 5M in cyclohexane : methanol, 1: 19, by volume), and the plates sealed. The plates were heated at 50°C for 24 hours under a nitrogen atmosphere, then allowed to cool and the solvents removed in vacuo.

A solution of hydrochloric acid in tetrahydrofuran (0. 5ml, 0.6N) was added and the reactions sealed and shaken at room temperature for 72 hours. The solvent was removed in vacuo, and the residues neutralised by the addition of triethylamine (50A1), then dissolved in methyl sulphoxide/water (approx. 0. 5ml.

90: 10, by volume). The solutions were purified b. y HPLC,. on a Phenomenex Magellen 5µ C18 column (150x10mm), using an elution gradient of acetonitrile : 0.05% aqueous trifluoroacetic acid. (15: 85 to 90: 10 to 98 : 2) at 6mlmin-1 and-detected at 210nm, and the-solvents evaporated in vacuo, to afford the title compounds.

The final compounds were analysed on a Phenomenex Magellen 5µ C18 column (30x4. 6mm) ; using acetonitile : 0.05% aqueous trifluoroacetic acid, at. a rate of 3. mimin-1, at 225nM and 255 nM, using the following gradient : Time (min) % acetonitrile 0.00-2. 55-95. 2. 5-3. 00. 95 3.00-3. 50 5 Ex. R1 R2 R3 RT/min No . 1 H3. . 03 I I I. fV NH2 H3C 1 \ _ _ H3 \ __ . 21 ci 12 1. 13 /HaC / N NHz H3C O 13 1. 18 N-a NH2 ci N NN2 CI 14 CH 1. 24 I I N NH2 H3C 15 H3 1. 22 N-a NH ? H3c 16 1. 23. H 3c N-a N H H3c 0 ure3 CH, 17 CH3 0 CH 3 1. 22 N < NI H H3C 0 > CH3 9 Cl3/ ,, CH3 y i Hs' H3c / N3C fV iVH2 19 CH3 1. 28 /1-IsGw I/ H3C O / H3C N (VH2 20 CH3 1. 28 3 N CH3 H3 1. 18 hic HIC H3C N NHz 22 CH3 1. 39 H 3c 3 N3C CH3 H3C N NH2 23 CH3 H3c 1. 38 H3 c Hug / H3C N NH2 24. cH3 \ 1. 4. 3 H C NH 3 2 Examples 25 to 62 The required acids (R1CO2H) (200µl, 0.25M solution in 1-methyl-2- pyrrolidinone), followed by the amines (R2NH2) (100µl, 0.5 M solution in methanol) were transferred to 96 well plates. Solutions of the appropriate aidehydes R3COH (100µl, 0.5 M solution in methanol) were prepared and then added, followed by a solution of the compound from preparation 6 (100R1, 0. 5M

in cyclohexane : methanol, 1 : 13, by volume), and the plates sealed. The plates were heated at 50°C for 24 hours under a nitrogen atmosphere, then allowed to cool and the solvents removed in vacuo.

A solution of hydrochloric acid in tetrahydrofuran (0. 5moi, 0. 6N) was added and the reactions sealed and shaken at room temperature for 24 hours. The solvent was removed in vacuo, and the residues dissolved in methyl sulphoxide/water (0. 5ml, 90: 10, by volume). The solutions were purified by HPLC, on a Phenomenex Magellen 5µ C18 column (150x10mm), using an elution gradient of acetonitrile : 0. 05% aqueous trifluoroacetic acid (15: 85 to 90: 10 to 98 : 2) at 6mlmin-1 over 10 minutes and detected at 210nm, and the solvents evaporated in vacuo, to afford the title compounds.

The final compounds were analysed on a Phenomenex Magellen 5g C18 column (30x4. 6mm), using acetonitile;0. 05% aqueous trifluoroacetic acid, at a rate of 3mlmin-', at 225nM and 255 nM, using the following gradient : Time (min) % acetonitrile 0.00-2. 5 5-95 2.5-3. 00 3.00-3. 50 Ex. R1 R2 R3 RT/min No 25 1. 21 1"' co : 0", 26 ' i H3-1. 32 i I L X NHn mCH3 27 H3c 1. 29 N-aNH2 0 28 NH2 O NNR, HgC" 29 I IV NH2 H3C 29 0. 6'9 I I I N NH2 H3C O 30 \ H3 1. 3 % \ HsCW I// NV NH2 O 31 s1. 23 31 CH 3 1. 23 Hic I/H3Cw I/ N NH2 O 32 0 : : c 1. 2'1 , nana2 33'H3c l. 36 N'a N H 2 F N NH2 F hic N-a NH 2 F N NHz F cl N'a N H 2 F N NHz F O 36 H3c 1. 55' I I I NH2 CF3 37 1-34 \ N NH2 H3c 37 sCH3 1. 34 3 o H3 1. 33 I I I N NH2 F 39 H3c A. I i/I/ CL, CH3 40 cl3 1. 35 ull-I N NH I L ; H3 : cl, 1' co N NH O. . CHUS 43 '1. 41 N-a N H CH3 N i H GH3 CH3 s-F _ \ I H3 I. + I o i NH CH3 Chez 44 H3c 1. 61 N-a N H CH 3 CH3 I u [-13 45 H3c 1. 31 1 1 hic CL, I 46 H3c 1. 5k I I I N NH H3C I 47 CH3 1. 45 I I N NH H3C. . CH C3 4 1. 57 I . I I N i H H3C. H3C CH3 L ; H3 49 1. 44 /, CH3 I/ fV i H O CH3 1 3 50 CH3'1. 36 o CH3 N NH H CHUG UH3 51 cl3 1. 31 1 H c u N'a N H 0 I UN3 52 1. 47 zizi Cl, 1 53 H3c 1. 47 i I I N NH F I 54 F CH3 1. 38 N-ANH. (V NH. ut3 55 cl3 1. 37 NNH F"" N NH F I 56 NC 1. 31 N-ANH F N NH F I I 57 CH 3 0. 98 H C- 3 CH3 N H s I H 3c CH 3 0 CH, 0 58 CH, 9, 1. 51 NANA N fVH CH3 59b . , i N3 1. 04 N CNg H ¢ ; 0 N N nu (I ; H3 60 CH 3 H3 c r, / N nu I H 3c CHg | 60 t9 H oNe / fV NN CH3 N-a N H a XCH3 < X N IH I CH3/

a=2- (2-fluorophenoxy) ethylamine was obtained from ChemDiv Inc. Building Blocks b= 2- (1 H-pyrazol-1-yl) ethylamine was obtained from Peakdale EXAMPLES 63 TO 218 The following example of general formula :

were prepared from 2-amino-4, 6-dimethylnicotinic acid (obtained from Bionet Research Ltd.), the compound from preparation 6 and the appropriate amine and aldehyde, following the procedure described for the preparation of examples 25 to 62. Ex No R2 R3 Rt/min 63 1. 36 I/I/ c3 6g. H3 1. 37 65 1. 36 HgC 66 H3c l. 27 3 67 1. 47 8 n NCv H3C CH3 69 coo : 1. 21 69° 0 1. 17 71 ~ 1. 43 CH3 CH3 72 H3c 1. 43 CHU CH3 73 H3C/t0 1. 43 73 1. 43 CH3 _ _ \ \ 1. 33 i r CH, c3 chug 3 c3 . 43 CH3 O CH3 77 Ne 1. 27 . 78 1. 29 I/ / 7H3 iVC CH 34F /if CHUG CH3 79 < ~. 34 CHg C ! 82 cH3 1. 5 HsC \ . 3 83 H3C>< 1. 51 zu CH3 CH3 84 H3c 1. 5 CH3 H3C 85 H3c 1. 52 CH3 CH3 86 CH3 1. 57 CH3 HIC 87 cH3 1. 46 F CI-ILS 3 t Cl3 F CHg F 89 F 1. 4 F 3 ! 90 < 1. 35 CH3 \F F 91 F 1. 39 CH3 F f, 92 1. 14 chug CH3 93 N 1. 03 94 1. 03 CHU zozos CHg 96aCK, o N 95 H3c 1. 23 CH3 96a i H3 1. 0 o Hs N/ 97 H3 H3 1. 54, gg H3c 1. 46 / 99 H3c,,-, 1. 38 F 100 H3C m F 1. 42 W I 101 H3C wS F w 1. 4 F 102 H3c H3c 1. 26 103 1. 39 /, CH3/ O CN3 104 H3C w 1. 49 /, CH3 4 105 1. 49 H3 O hic 106 /CrH3//rH3 107 T h3 i. 4 107 CH3 1. 4 v ./ 108 1. 4 /, CH3/ 110 F 109 H3 F. 1.'43 /iCH3/ 1. 0 Nc . 35 , CI-13 O 1. 36 H3 O 1C 112 3c 1. 31 "0 113 1. 59 114 GH3/ 114 H3C v 1. 57 cl3 /, c3/ p CH3 H3 C : c CH H3C 116 1. 59 (DICH3 CL, CH3 k 7 tH3 1. 64 R < z CH3 N A 118 +. 26 119 1. 23 11 \ ___ 1. 2 iCHs O 1 9 H co \ . 23 3 CH3 120 H3c 1. 15 H3c 121 H3c,-o 1. 15 Hic HgC 1. 15 22 //Hs O 122 H3 cl,"o 1. 15 3 3 24/O_-__ 1. 11 H3C NC 126 H 3cl-lo F 1. 1 3 F 126 H o F 1. 15 3 127 H3c,,, o H3c CH3 1. 3 3/H3C 128 H3c 1. 32 Chug N C'3 129 H 3cllo H3c 1. 07 130 CH3 1. 23 hic o HsC I/ O 131 1. 2 NsW I / 3 0 NC"C 132 H3c 1. 41 Ici H3C0//CH 3 133 H3c 1. 2 If CH3 CH3 134 H3C < H3C w 1. 22 CHg CH3 135 H3c CH 1. 12 //ice3 CH3 O 136 H3c CH3 1. 25 o /I \ CH3 137 H3c NC 1. 39 . I- 138 H3c 1. 45 CH3 F l v l, CH3 F 139 H3c F 1. 47 CHUG 1. 23 140 H3C < C 1. 23 Chug ; 141 H3c H3c 1. 09 u W CH3 142 H3C <. 1. 66 If H3C CH3 143 H 3c H3c 1. 67 Hic H3C 144 H3 . 55 H3c (xo CH3 H3C O 145 H3c CH3 1. 56 3 ! ! I 146 H3c'l. 57 H3C. F 147 H3c F 1. 59 Hic H3C., 148. H3c NC 1. 5 Hic HIC 149 H3c H3c 1. 45 H3C 150 H3c 1. 52 H3C 151 CH3 1. 41 F 1. 64 / H3C 153 H3 0. 99 / N 3""3 CH3 154 H3 1. 0 Y N 0 1 55/> H3C v 1. 69 \I I/ 156 1. 36 wF CH3 157 H 3c 1. 36 158 1. 37 'F 1. 57 I-ICH3 F H3C 159 V . 2 //iCH3 F O 160 C H3 1. 27 a \ I 161 (C Nc 1. 23 I/I/ 1. 29 162 1. 29 163 F 1. 3 F F I/I/ F 64.- c 1. 41 F 165 H 3c 1. 46 CH3 166 H3C w 1. 45 hic F H3C CH, F GH3. 168 CH3 1. 47 HIC Hic H3C 169 CH3 1. 4 F F F \ F F 1. 32 fez F F 172 F 1. 52 CHg c3 _-1 3 F H3 1. 5 i 74 F w 1. 51 Hc"IC H3C 175 F 1. 42 W 0\C O 176 F \ j H3 1. 42 o. 177 F NC 1. 36 4 AO 1 79 F <ts < 1. eo NHz i/I/ F 180 F F 1. 43 181 F 1. 48 C) 82 F v H3C w 1. 62 Cl3 CH3 183 F H3c 1. 59 Hic H3C 184 F H3c 1. 61 Cl, CH3 185 CH3 1. 64 H3c I H3C 6 F \ cH3 1. 56 F 7 F F'. 47 F I 1sa F v F v i. 45 F 189 F F 1. 31 F I F 190 F NC 1. 42 / N 192 F \. N \ 1. 11 192 F H3c 1. 3 191 1. 4 I/ / CH3 194 H3 1. 38 Fic 195 F H3 C", 1. 41 196"lo 1. 3 Hic Foc 96 'H c \ . 3 3 F 197 1. 26 CON 198 1. 24 I/I/ F 199 1. 24 NO 200 1. 31 F 201 1. 32 202 1. 37 F 203 H3c 1. 48 Fj CH3 F CI 203, H3 1. 48 F CH3 204 » H3c 1. 4s F hic 3 205 H30 1. 5 He 206 : ; t49 \ Hic 207 CH3 1. 45 \ F 208 1. 35 F 209 F 1. 2 F F F 210 F 1. 3 F F 211 NC 1. 3 F 212 HOC 1. 19 F 213 H3C N NA 0. 96 F" 214 Cl1. 49 214 I W W 215 m H3 1. 4$ 216 F 1. 42 217 Nc 1. 32 218 j HaC < 1 12B /lez a-1-methyl-1 H-pyrazole-4-carbaldehyde was obtained from ChemCollect EXAMPLES 219 TO 233

Solutions of 4-methylbenzylamine (100µl, 0.5M in methanol) and m- anisaldehyde (100µl, 0. 5M in methanol) were added to a solution of the appropriate acids (100µl, 0.5M in methanol/1-methyl-2-pyrrolidinone 1: 1). A solution of the isocyanate from preparation 6 (100µl, 0.5M in methanoUcyclohexane, 1 : 1) was then added, the reactions sealed and heated at 50°C for 24 hours. The solvents were removed under reduced pressure, hydrochloric acid in tetrahydrofuran (500µl, 0. 6M) was added, and the vessels were resealed and agitated again at room temperature for a further 24 hours.

The solvents were removed under reduced pressure, the residues dissolved in methyl sulphoxide, and purified by HPLC, using a Phenomonex Luna 150x10mm, 10µm column, in acetonitile : 0.1% aqueous diethylamine, at 8mimis'\ at 225nM, using the following gradient. Time (min) % acetonitrile 0.00-0. 50 5 0.50-0. 60 5-20 0. 60-6. 50 20-95 Ex. No. R1 Retention time/min 219 5. 366 1- (NHZ 220 5. 391 if CH3 221 a 5. 279 COO C'a zæ ~ 5. 838 -J c9 223 H C o 5. 265 9 N 224b HS 5. 782 N 2Z5 N, 5. 559 Nay cl 226 4. 693 N 227 4. 749 Br 228 H2Nw 6. 321 N 229 4. 777 Hic hic . 230 5. 642 231. _ _ s. 321 232 5. 670 11 232 N^ 5. 670 110'Y 233c Cl 4. 832 N\ ; +N HAN H, cl HaC

a- (4-trifluoromethyl) nicotinic acid obtained from Maybridge b=starting nicotinic acid prepared as in WO 9954333 c=5-chloro-2-methylsulphanyl-pyrimidine-4-carboxylic acid obtained from Maybridge EXAMPLE 234 N-[2-Amino-1-(3-methoxyphenyl0-2-oxoethyl]-4-cyano-N-(4- methvibentyl) benzamide

A mixture of 4-cyanobenzoic acid (584mg, 4mmol), m-anisaldehyde (486µl, 4mmol), 4-methylbenzylamine (509Si, 4mmol) and the compound from preparation 6 (752mg, 4mmol) in methanol (15mi) was stirred at room temperature for 18 hours. The reaction was then stirred at 50°C for a further 4 hours, and the mixture concentrated under reduced pressure. The residue was dissolved in a solution of hydrochloric acid in tetrahydrofuran (0.6N, 15ml), and the solution stirred at room temperature for 2 hours, then evaporated under reduced pressure. The product was washed with 1 N sodium hydroxide solution, and extracted with ethyl acetate. The combined organic extracts were washed with brine, dried (MgSO4) and evaporated under reduced pressure. The crude product was purified by column chromatography on silica using methanol : dichloromethane (1: 99) as eluant to afford the title compound as a yellow foam, 1. 38g.

'Hnmr (CDCI3, 400MHz) : fi 2.27 (s, 3H), 3.75 (s, 3H), 4.35 (bd, 1H), 4.58 (bd, 1H), 5. 43-5.70 (m, 3H), 6. 88 (m, 4H), 6.99 (m, 3H), 7.22 (m, 2H), 7. 50~7. 63 (m, 3H).

LRMS : m/z (ES+) 436 [MNa+] Microanalysis found: C, 71. 63 ; H, 5.66 ; N, 10.05. C25H23N3O3;0.3H2O requires C, 71. 69 ; H, 5.68 ; N, 10. 03%. EXAMPLES 235 TO 239 The following compounds of general formula : were prepared from m-anisaldehyde, 4-methylbenzyiamine, the compound from 5 preparation 6 and the appropriate acid, following a similar procedure to that described in example 234. Ex. R 1 Yieldl% No. 235 F 30'Hnmr (CDCI3, 400MHz) S : 2. 25 (s, 3H), 3. 75 (s, 3H), 4. 38 (d, 1 H), 4. 58 (d, 1 H), 5. 40-5. 60 (m, 3H), 6. 84 (m, 4H), F 7. 00 (m, 4H), 7. 24 (m, 3H). LRMS ; rn/z (ES4) 447 iMNasl Microanalysis found : C, 67. 62 ; H, 5, 21 ; N, 6. 51. C24H22F2N203 requires C, 67. 92 ; H, 5. 22 ; N, 6. 60%. 236 68'Hnmr (CDC13, 400MHz) 8 : 2. 24 (s, 3H), 3. 78 (s, 3H), 4. 36 (d, 1 H), 4. 58 (d, F F 1H), 5. 50 (bs, 2H), 6. 78-7. 02 (m, 10H), 7. 25 (m, 2H). HRMS : m/z (ES' 849. 3238 [2M+Hl+ C24H22F2N203 requires 849. 3269 237 F 20'Hnmr (CDCI3, 400MHz) 8 : 2. 23 (s, 3H), 3. 78 (2xs, 6H), 4. 38 (d, 1 H), 4. 59 H3Cs 4s (d, 1 H), 5. 57 (s, 1 H), 6. 42 (m, 2H), 6. 66 (m, 3H), 7. 00 (m, 4H), 7. 24 (m, 1 H). LRMS : m/z (ES+) 477 [MNapl 85'Hnmr (DMSOde, 400MHz) (rotamers) 8 : 2. 12, 2. 18 (2xs, 3H), 2. 40, 2. 58 (2xs, 3H), 3. 64 (s, 3H), 4. 25, 4, 43 (2xd, 1 H), 4. 79, 4. 90 (2xd, 1 H), 5. 82, 5. 99 (2xs, 1 H), 6. 64-6. 96 (m, 7H), 7. 08-7. 23 (m, 3H), 7. 37, 7. 59, 7. 82 (3un, 3H). Microanalysis found : C, 71. 21 ; H, 6. 32 ; N, 10. 22. C24H2eN303 requires C, 71. 44 ; H, 6. 25 ; N, 10. 41% 239a NNN3 25'Hnmr (DMSOds, 4o0MHz) 8 : 2. 16 (s, 3H), 2. 22 (s, 3H), 2. 84 (s, 3H), 3. 58 (s, 3H), 4. 16 (d, 1 H), 4. 40 (d, 1 H), 5. 80 (s, Cow 1 H), 6. 30 (d, 1 H), 6. 60 (m, 3H), 6. 78 3 (m, 1 H), 6. 81 (m, 3H), 7. 16 (m, 1 H), 7. 19 (d, 1 H), 7. 41 (s, 1 H), 7. 60 (s, 1 H). LRMS : m/z (ES+) 455 [MNal

a=6-methyl-2-methylamino-nicotininc acid was obtained from Peakdale EXAMPLE 240 N-[3-Amino-1-(3-methoxyphenyl)-3-oxopropyl]-4-methyl-N-(4- methylbenzyl)nicotinamide

A solution of the acid from preparation 13 (176mg, 0. 42mol), O-(1H- benzotriazol-1-yl)-N,N,N',N'-tetramethyluronium hexafluorophosphate (157mg, 0. 42mol) and N-ethyldiisopropylamine (147µl, 0. 84mmol) in dichloromethane (20moi) was stirred at room temperature for an hour. 0.88 Ammonia (0. 5mil) was added and the reaction stirred at room temperature for 18 hours. The mixture was washed with sodium bicarbonate solution (20ml), water (20ml) and brine (20mi) then dried (Na2SO4) and evaporated under reduced pressure. The residue was purified by column chromatography on silica gel using an elution gradient of dichloromethane : methanol : O. 88 ammonia (100 : 0: 0 to 97.5 : 2.5 : 0.25) to afford the title compound, 79mg.

HRMS: m/z (ES+) 418.2117 [MH+] EXAMPLE 241 2-Aminr (1S)-3-amino-3-oxo-1-phenylpropyl]-N-(4- methylbenzyl)nicotinamide The amine from preparation 11 (100mg, 0. 37mmol), 2-aminonicotininc acid (47. 1mg, 0. 34mmol) and 0- (1H-benzotriazol-1-yl)-N, N, N', N'-tetramethyluronium

hexafluorophosphate (141 mg, 0. 37mol) were added to a solution of N- ethyldiisopropylamine (195µl, 1.125mmol) in N, N-dimethylformamide (2ml), and the solution stirred at room temperature for 18 hours, then at 85°C for a further 48 hours. The cooled mixture was concentrated under reduced pressure and the residue partitioned between sodium carbonate solution (10moi) and ethyl acetate (10ml). The layers were separated, the organic phase washed with water (l0ml) and brine (5ml), dried (Na2S04) and evaporated under reduced pressure. The residual brown oil was purified by column chromatography on silica gel using an elution gradeint of dichloromethane : methanol (100: 0 to 90: 10) to afford the title compound as a brown oil, 25. 1mg.

'H-nmr (CDCl3, 400MHz) # : 2.21 (s, 3H), 2.64 (m, 1H), 3.19 (m, 1H), 4.21 (m, 1 H), 4.58 (m, 1 H), 5.80-6. 05 (m, 2H), 6.38-6. 72 (m, 3H), 7.25 (m, 9H), 7.95 (s, 1H).

HRMS: m/z (ES+) 389.1972 lMH+l Cz3H24N402 requires 389.1972.

Example 242 5-Chioro-2-methylthio-N-[2-amino-1-{1,4-benzodioxan-6-yl}-2- oxoethyl]-N-(4- methylbenzyl)pyrimidine-4-carboamide To a solution of methyl-5-chloro-2-methylthiopyrimidine-4-carboxylate (1.0g, 4. 88mmol) in methanol (40ml) was added 4-methylbenzylamine (0. 71 g, 5. 86mmol), 1,4 benzodioxan-6-carboxaldehyde (0. 96g, 5. 86mmol) and 4- phenylcyclohex-1-enylisonitrile (1. 075g, 5. 86mmol). After 3 hours the solvent

was evaporated and the residue was dissolved in 10ml of 1. 2N HCl in THF.

After 1 hour the solvent was evaporated and the residue dissolved in ethyl acetate (100ml). This solution was washed with water (50mut), saturated NaHCO3 solution (50mi) and brine (50ml), dried over MgSO4, filtered and evaporated. The residue was purified by chromatography on silica gel (50gm), eluting with a gradient of pentane-ethyl acetate (9: 1 to 4 : 6 in 10% increments of EtOAc) to afford the title compound as a solid, 2.28g, (93%).

'Hnmr (CDC13, 400MHz) 8 : 2.2 (s, 3H), 2.8 (s, 3H), 4.1-4. 5 (m, 6H), 5. 0-5.2 (m, 1 H), 5. 5-5.8 (m, 2H), 6.5-7. 3 (m, 3H), 8.3 (s, 1 H) LRMS: m/z (APCI 521 [MNal Microanalysis found: C, 57. 20 ; H, 4.70 ; N, 10.81. C24H23CIN4OS requires C, 57. 77 ; H, 4.65 ; N, 11.23% Example 243 5-Chloro-2-methanesulfonyl-N-[2-amino-1-[1,4-benzodioxan-6-y l]-2-oxoethyl]- N- (4-methylbenzyl)pyrimidine-4-carboxamide To a solution of the sulfide from example 242 (2.04g, 4. 08mmol) in CH2Cl2 (30mi) was added 3-chloroperoxybenzoic acid (3. 10g, 8. 99mmol) portionwise over 10 minutes at ambient temperature. After 18 hours the solution was washed with 10% NaHSO3 solution (10ml), saturated NaHCO3 solution (2 x

10ml), brine (10ml), dried over MgSO4, filtered and evaporated to give the title compound as a yellow solid, 1.5g, (69%).

'Hnmr (CDCl3, 400MHz) 8 : 2.2 &2.38 (2 x s, 3H), 3.2 & 3.38 (2 x s, 3H) 4.1- 4.6 (m, 6.5H), 5.15-5. 25 (m, 0. 5H), 5.4-5. 8 (br m, 1H), 6.05 (s, 1H) 6. 7-7. 3 (m, 7H), 8. 6 & 8. 7 (2 x s, 1 H) LRMS: m/z (APCIs) 531 [MEn Example 244 5-Chloro-2-ethylamino-N-[2-amino-1-{1,4-benzodioxan-6-yl}-2- oxoethyl]-N-(4- methylbenzyl)pyrimidine-4-carboxamide Ethylamin was passed through an ice-cold solution of the sulfone from example 243 (0.15g, 0. 28mmol) in THF (3ml) for 20 minutes. After 2 hours at room temperature, the solvent was evaporated. The residue was dissolved in ethyl acetate (10mi) and the solution was washed with water (10ml), saturated NaHCO3 solution (2 x 10ml0, brine (10ml), dried over MgSO4, filtered and evaporated. The residue was chromatographed on silica gel (10g) eluting with a gradient of pentane-ethyl acetate from 90/10 to 30/70 in 10% increments of ethyl acetate to give the title compound as a foam 107mg, (76%). nHnmr (CDCl3, 400MHz) 8 : 1.15-1. 3 (complex t, 3H), 2. 25 & 2.30 (2 x s, 3H), 3. 3 - 3. 45 (complex q, 2H) 4. 1-4. 6 (m, 5. 5H), 4.9-5. 00 (m, 0. 5H), 5.2-6. 0 (m's, 3H), 6.6-7. 2 (m, 7H), 8. 15 & 8.2 (2 x s, 1 H)

LRMS : m/z (APCl+) 518 [MNal Microanalysis found: C, 59.76; H, 5.36 ; N, 13. 63. C2sHCIN504, 0. 05 CH2CIz requires C, 60.15 ; H, 5.26 ; N, 14. 00% Example 245 5-Chloro-2-ethylamino-N-[2-amino-1-{1,4-benzodioxan-6-yl}-2- oxoethyl]-N-(4- methylbenzyl)pyrimidine-4-carboxamide The title compound was obtained from the sulfone of example 243, using the method from example 244 and using ammonia as the amine, as a foam, 91. 5mg, (89%) 1Hnmr (CDCI3, 400MHz) 8 : 2. 25 & 2.30 (2 x s, 3H), 4.1-4. 6 (m, 5.5H), 4.9- 4.95 (d, 0.5H), 5.2-6. 0 (m's, 5H), 6.6-7. 2 (m, 7H), 8. 15 & 8.2 (2 x s, 1H) LRMS: m/z (APCIf) 468 [MH+] Microanalysis found : C, 57.25 ; H, 4.80 ; N, 14.14. C23H22CIN5O4. 0. 2 CH2CI2 requires C, 57.47 ; H, 4. 68 ; N, 14. 44% Example 246 5-Chloro-2-ethylamino-N-[2-amino-1-{1,4-benzodioxan-6-yl}-2- oxoethyl]-N-(4- methylbenzyl)pyrimidine-4-carboxamide

To an ice-cold solution of the product from preparation 14 (276mg, 0. 45mmol) and anisole (245mg, 2. 26mmol) in CH2C12 (6mt) was added trifluoroacetic acid (6ml), After 2hrs the reaction mixture was evaporated to dryness. The residue was dissolved in fresh CH2C12 (20ml) and the solution was washed with 10% Na2CO3 solution (20ml), saturated NaHCO3 solution (20ml) brine (20ml) dried over MgSO4, filtered and evaporated. The residue was purified by chromatography on silica gel (10gm), eluting with a gradient of CH2CI2-CH30H- NH4OH (99: 1: 0.1 to 90: 10: 1 in 1% increments of CH30H and 0. 1% increments of NH40H) to afford the title compound as a foam, 69mg, (30%).

'Hnmr (CDCl3, 400MHz) 8 : 1. 95-2. 6 (s @ 2.3, 3H & br s, 2H), 2. 9-3. 1 (m, 2H), 3. 4-3.65 (m, 2H), 4. 1-4. 8 (m, 6H), 5. 25 & 5.5 (2 x s, 1H), 5. 7- 6. 4 (m, 2H), 6.6-7. 1 (m, 7H), 8.1 &8.2 (2 x s, 1H) LRMS : m/z (APCl+) 511 [MH+] Microanalysis found: C, 56.68 ; H, 5. 40 ; N, 15. 34. C25H27CIN6O4 : 0.25 CH2CI2 requires C, 56. 98 ; H, 5. 21; N, 15.79% Example 247 5-Chloro-2-methylthio-N-[2-amino-1-{3-methoxyphenyll}2-oxoet hyl]-N-(4- methylbenzyl)pyrimidine-4-carboxamide

This was prepared in the same manner as the product from example 242, using 3-methoxybenzaldehyde as the aldehyde component, to return the title compound as a solid, 1.74g, (76%).

'Hnmr (CDCl3, 400MHz) # : 2. 25 & 2.35 (2 x s, 3H), 2.38 & 2.50 (2 x s, 3H), 3.65 & 3.78 (2 x s, 3H) 4. 35-4.6 (q, 2H), 5. 8 (s, 1 H), 6. 8-7. 3 (m, 8H), 8.4 (s, 1 H) LRMS: m/z (APCI*) 471/473 [MH+] Microanalysis found : C, 57. 34 ; H, 5. 11 ; N, 11.36. C23H23CIN4O3S : 0, 2CH2CI2 requires C, 57.48 ; H, 4.86 ; N, 11. 58% Example 248 5-Chloro-2-methylthio-N-[2-amino-1-{3-methoxyphenyll}2-oxoet hyl]-N-(4- methylbenzyl)pyrimidine-4-carboxamide

This was prepared in the same manner as the product of example 243, using the sufide from example 247, to return the title compound as a foam, 0.97g, (53%). lHnmr (CDCI3, 400MHz) 8 : 2. 2 &2.38 (2 x s, 3H), 3.25 & 3.35 (2 x s, 3H) 3. 63 & 3.8 (2 x s, 3H), 4. 35-4. 62 (q, 2H), 5. 3 & 6. 1 (2 x s, 1H), 5.5-5. 9 (br d, 2H) 6.7 - 7. 3 (m, 8H), 8.65 (s, 1 H) LRMS: m/z (APCl+) 503/505 [MH+] Microanalysis found: C, 53.25 ; H, 4.47 ; N, 10.50. C23H23CIN4O5S, 0.25CH2Cl2 requires C, 52.67 ; H, 4.42 ; N, 10. 68% Example 249 5-Chloro-2-amino-N-[2-amino-1-{3-methoxyphenyl}-2-oxoethyl]- N-(4- methvlbenzvl) pyrimidine-4-carboxamide The title compound was prepared from the sulfone of example 248, using the method from example 244 and ammonia as the amine, to return the title compound as a foam, 22mg, (12%) 'Hnmr (CDCI3, 400MHz) S : 2.25 & 2.30 (2 x s, 3H), 3.65 & 3.78 (2 x s, 3H), 4.3 - 4. 6 (m, 1. 75H), 4. 95-5. 05 (d, 0. 25H), 5.4-6. 0 (m's, 5H), 6.6-7. 3 (m, 8H), 8.15 & 8. 18 (2 x s, 1 H) LRMS : m/z (APCl+) 462/464 [MNa+] Example 250 5-Chloro-2-amino-N-[2-amino-1-{3-methoxyphenyl}-2-oxoethyl]- N-(4- methylbenzyl)pyrimidine-4-carboxamide

This was prepared in the same manner as the product of example 249, using dimethylamine as the amine, to give the title compound as a foam, 0. 06g, (31 %). tHnmr (CDCl3, 400MHz) 3 : 2. 25 & 2.30 (2 x s, 3H), 3.1 & 3, 18 (2 x s, 6H), 3.65 & 3.75 (2 x s, 3H), 4.30-4. 6 (m, 1.75H), 4.90-5. 00 (d, 0.25H), 5.39 & 5.59 (2 x s, 1 H), 5.3-5. 65 (br m, 1 H), 5.8-6. 1 (br m, 1 H), 6. 7 - 7. 3 (m, 8H), 8.2 & 8.25 (2xs, 1H) LRMS : m/z (APCl+) 468/470 [MH+] Example 251 5-Chloro-2-amino-N-[2-amino-1-{3-methoxyphenyl}-2-oxoethyl]- N-(4- sus

This was prepared in the same manner as the product of example 249, using methylamine as the amine, to give the title compound as a foam, 0.24g, (88%).

'Hnmr (CDCI3, 400MHz) #: 2.25 & 2.30 (2 x s, 3H), 2.9-3. 0 (m, 3H), 3. 65 & 3.78 (2 x s, 3H), 4.35-4. 6 (m, 1.75H), 4. 98-5. 05 (d, 0. 25H), 5.2-6. 00 (m, 3H), 6.6-7. 3 (m, 8H), 8.1-8. 2 (br s, 1 H) LRMS : m/z (APIS) 454/456 [MH+1 Microanalysis found : C, 59.51 ; H, 5.44 ; N, 14.34. C23H24CIN5O3; 0.2 CH2Cl2 requires C, 59.17 ; H, 5.22 ; N, 14.87% Example 252 5-Chloro-2-amino-N-[2-amino-1-{3-methoxyphenyl}-2-oxoethyl]- N-(4- methylbenzyl)pyrimidine-4-carboxamide This was prepared in the same manner as the product from example 242 using 5-methyl-2-methylthiopyrimidine-4-carboxylic acid (preparation 17) and 3- methoxybenzaldehyde as the acid and aldehyde components to give the title compound as a solid, 0. 97g, (72%).

'Hnmr (CDCI3, 400MHz) # : 2. 0-2. 2 (m, 6H), 2.4 & 2.55 (2 x s, 3H), 3.6 & 3.65 (2 x s, 3H), 4.3-4. 5 (m, 1.5H), 4. 95-5. 0 (d, 0. 5H), 5. 3 (s, 0.5H), 6.05 (s, 0. 5H), 6.6-7. 4 (m, 9.5H), 7.6-7. 7 (br s, 0.5H), 8.35 (s, 0. 5H), 8.62 (s, 0. 5H) LRMS : m/z (APCl+) 451 [MH+]

Microanalysis found: C, 63.31 ; H, 5.98 ; N, 11. 98. C24H26N403S : 02. H2O, 0.1 EtOAc requires C, 63. 30 ; H, 5. 92 ; N, 12.10% Example 253 5-Chloro-2-amino-N-[2-amino-1-{3-methoxyphenyl}-2-oxoethyl]- N-(4- methylbenzyl)pyrimidine-4-carboxamide This was prepared in the same manner as the product of example 243, using the sulfide from example 252, to give the title compound as a solid, 0.7g, (74%).

'Hnmr (CDC13, 400MHz) 8 : 2, 1 (s, 3H), 2.18 & 2.35 (2 x s, 3H), 3.25 & 3.40 (2 x s, 3H), 3.6 & 3.65 (2 x s, 3H), 4. 25-4. 5 (q, 1. 5H), 4. 95-5. 0 (d, 0.5H), 5.28 (s, 0. 5H), 6.15 (s, 0. 5H), 6.58-7. 4 (m, 9. 5H), 7. 6-7. 75 (br s, 0.5H), 8.75 (s, 0. 5H), 9.1 (s, 0.5H) LRMS: m/z (APCl+) 483 [MH+] Microanalysis found : C, 59.27 ; H, 5. 36 ; N, 11.48. C24H26N4OsS requires C, 59.79 ; H, 5.43 ; N, 11. 61% Example 254 5-Chloro-2-amino-N-[2-amino-1-{3-methoxyphenyl}-2-oxoethyl]- N-(4- methvlbenzv !) pvrimidine-4-carboxamide

This was prepared in the same manner as the product of example 244, using the sulfone from example 253 and dimethylamine as the amine, to give the title compound as a foam, 0.14g, (74%).

1Hnmr (CDCl3, 400MHz) # : 2.05 (s, 3H), 2.25 & 2.30 (2 x s, 3H), 3.1 & 3.18 (2 x s, 6H), 3.65 & 3.75 (2 x s, 3H), 4.30-4. 6 (m, 1. 75H), 4. 90-4. 95 (d, 0.25H), 5.5-5. 65 (m, 2H), 5. 9-6. 05 (br m, 1H), 6. 65-7. 2 (m, 8H), 8.1 & 8.15 (2 x s, 1H) LRMS : m/z (APCIs) 448 [MH+1 Microanalysis found : C, 65.62 ; H, 6. 62 ; N, 14. 75. C25H29N5O3; 0. 10 CH2CI2, 0.2 EtOAc requires C, 65.68 ; H, 6.58 ; N, 14.79% Example 255 5-Methyl-2-methylamino-N-[2-amino-1-{3-methoxyphenyl}-2-oxoe thyl]-N-(4- methylbenzyl)pyrimidine-4-carboxamide

This was prepared in the same manner as the product from example 254, using methylamine as the amine, to give the title compound as a foam, 0. 15g, (85%).

1Hnmr (CDCl3, 400MHz) # : 2.0-2. 15 (m, 3H), 2.25 & 2. 30 (2 x s, 3H), 2.9-3. 0 (m, 3H), 3.65 & 3. 75 (2 x s, 3H), 4. 35-4. 6 (m, 1.75H), 4.98-5. 05 (d, 0.25H), 5. 2-5. 35 (m, 1 H), 5.5 (s, 0. 25H), 5.62 (s, 0. 75H), 5.7-6. 1 (br m, 2H), 6.6-7. 3 (m, 8H), 8.02 & 8.05 (2 x s, 1H) LRMS: m/z (APCR) 434 [MHI Microanalysis found: C, 64.50 ; H, 6.32 ; N, 15. 22. C24H27N5O3 : 0.15 CH2CI2 requires C, 65.00 ; H, 6. 17 ; N, 15. 69% Example 256 3-Methyl-2-methylamino-N-[2-amino-1-{3-methoxyphenyl}-2-oxoe thyl]-N-(4- methylbenzyl) pvrazine-2-carboxamide This was prepared in the same manner as the product from example 242 using 3-methylpyrazine-2-carboxylic acid (JOC, 2002, p556) and 3- methoxybenzatdehyde as the acid and aldehyde components to give the title compound as a foam, 0.5g, (63%).

1Hnmr (CDCl3, 400MHz) # : 2.20 & 2.32 (2 x s, 3H), 2.4 & 2.55 (2 x s, 3H), 3. 65 & 3.78 (2 x s, 3H), 4. 30-4. 6 (m, 1. 80H), 5. 10-5. 15 (d, 0. 20H), 5.45 & 5.85 (2

x s, 1H), 5. 5-5. 70 (br m, 1H), 5.9 -6. 05 (br m, 1H), 6. 5-7. 3 (m, 8H), 8. 3- 8, 45 (m, 1H) LRMS : m/z (APCl+) 405 [MId11 Microanalysis found: C, 67. 23 ; H, 6.09 ; N, 13.40. C23H24N4O3 : 0.25 H20, 0. 1 EtOAc requires C, 67.27 ; H, 6. 10 ; N, 13. 41% Examc ! e257 2-Amino-N-[carbamoyl-(2,3-dihydro-benzo[1,4]dioxin-6-yl)-met hyl]-4,6-dimethyl- N-t4-methvl-benzyl)-nicotinamide A mixture of 2-amino4, 6-dimethyinicotinic acid (obtained from Bionet Research <BR> <BR> <BR> Ltd. ), (665mg, Ommol), 1, 4-benzodioxan-6-carboxaldehyde (656mg, 4mmol), 4- methylbenzylamine (509µl, 4mmol) and the compound from preparation 6 (752mg, 4mmol) in methane) (15m !) was stirred at room temperature for 18 hours. The reaction was then stirred at 50°C for a further 4 hours, and the mixture concentrated under reduced pressure. The residue was dissolved in a solution of hydrochloric acid in tetrahydrofuran (0. 6N, 15ml), and the solution stirred at room temperature for 4 hours, then evaporated under reduced pressure. The product was washed with 2N sodium hydroxide solution, and extracted with ethyl acetate. The combined organic extracts were washed with brine, dried (MgSO4) and evaporated under reduced pressure. The crude product was purified by column chromatography on silica gel using

methanol : dichloromethane (4: 96) as eluant to afford a yellow foam which was triturated with diethyl ether to give the title compound as a yellow powder, 360mg.

LRMS : m/z (ES+) 461 fMH+J, 483 [MNafll Microanalysis found : C, 66. 94; H, 6.20 ; N, 11.59.

C26H28N404 ; 0. 2H20 ; 0. 3 (CH3CH2) z0 requires C, 67. 17; H, 6.51 ; N, 11. 52%.

Example 258 & Example 259 Compound from example 18 was separated via chiral HPLC into its two enantiomers, using a Chiralpak AD250 20mm column, in 50% hexane 50% isopropylalcohol, at 220nm over 40 min at a flow rate of 10ml/min, Enantiomeric excesses were determined by HPLC analysis using a Chiralpak AD250 4.6mm column, in 50% hexane 50% isopropylalcohol, at 220nm over 30 min at a flow rate of 1 mUmin Example 258 (R or S)-2-Amino-N-[carbamoyl-{(3-methoxyphenyl)-methyl}]-4,6-dime thyl-N- (4- methvl-benzvl)-nicotinamide * single enantiomer Enantiomer 1 : retention time 14. 87 min, >99% ee LRMS : m/z (ES+) 433 [MH 85% ; 456 [MNa+], 100% Example 259 (R or S)-2-Amino-N-[carbamoyl-{(3-methoxyphenyl)-methyl}]-4,6-dime thyl-4- methvl-benzvi)-nicotinamide

* single enantiomer Enantiomer 2: retention time 20. 20 min, 98. 89 ee LRMS : m/z (ES+) 433 [MH41, 95%; 455 [MNal, 100%

Preparation 1 2-Methylamino-nicotinic acid

Methylamine hydrochloride (8. 6g, 128mol) was added portionwise to a mixture of 2-chloronicotinic acid (10. 0g, 64mmol), potassium carbonate (35. 4g, 256mol) and copper (1) bromide (920mg, 6. 4mmol) in N, N-dimethylformamide (100mi), and the reaction heated at 100°C for 18 hours, then cooled. The resulting precipitate was removed by filtration, washing through with additional methanol and the filtrate evaporated under reduced pressure. The residue was redissolved in 2N sodium hydroxide solution (200ml), washed with diethyl ether

(4x80mi), then the pH of the aqueous solution adjusted to 6, using concentrated hydrochloric acid. This aqueous solution was evaporated under reduced pressure, the residue dissolved in methanol, poly (4-vinylpyridine), 2% cross- linked (5g) was added, and the mixture stirred at room temperature for 18 hours. The resin was removed by filtration, and the procedure then repeated.

The solution was evaporated under reduced pressure, and the solid recrystallised from ethanol, to afford the title compound, 5.2g.

'H-nmr (DMSO-d6, 270MHz) 8 2.91 (s, 3H), 6.50 (dd, 1H), 8.01 (dd, 1H), 8.13 (m, 1H), 8.37 (m, 1H).

LRMS: m/z (ES+) 153 [MHl Preparation 2 2-Ethvlamino-nicotinic acid Potassium carbonate (9. 21g, 66. 6mrnol) was added to a mixture of 2- chloronicotinic acid (5. 0g, 31. 7mol), ethylamin hydrochloride (5.18g, 63. 4mmol) and copper (1) bromide (450mg, 3. 17mmol) in N, N- dimethytformamide (50ml), and the reaction heated at 100°C for 1.5 hours, then cooled. The resulting solid was removed by filtration, and the filtrate evaporated under reduced pressure. The residual blueJgreen soíid was triturated with acetone, and the resulting solid filtered off and dried in vacuo to afford the title compound.

The filtrate was evaporated under reduced pressure, the residue triturated with diethyl ether and the solid filtered and dried in vacuo, to afford additional product, 2.2g in total.

Preparation 3 2-Benzylamino-nicotinic acid

Potassium carbonate (4. 82g, 34. 9mmol) was added to a mixture of 2- chloronicotinic acid (5. 0g, 31. 7mol), benzylamine (3. 47ml, 31.7mmol) and copper (t) bromide (450mg, 3. 17mmol) in N, N-dimethytformamide (50ml), and the reaction heated at 100°C for 2 hours, then cooled. The resulting solid was filtered off and the filtrate evaporated under reduced pressure. The residue was partitioned between 4N sodium hydroxide solution (25ml) and dichloromethane (25ml) and the layers separated. The organic solution was extracted with water (2x), and the combined aqueous solutions, neutralised using concentrated hydrochloric acid. The resulting solid was filtered off, washed with cold water and dried in vacuo at 50°C, to afford the title compound, 1. 6g.

H-nmr (DMSOd6, 300MHz) 8 : 4. 69 (d, 2H), 6.62 (dd, 1H), 7.31 (m, 5H), 8.10 (dd, 1H), 8.24 (dd, 1H), 8. 48 (bs, 1H).

Preparation 4 2-{[3-(4-Morpholinyl)propyl]aminolnicotinic acid 3- (4-Morpholino)-1-propylamine (9. 239, 40mmol) was added to a solution of 2- chloronicotininc acid (5g, 32mol), potassium carbonate (4.42g, 3mmol) and copper (I) bromide (460mg, 3. 2mmol) in N, N-dimethylformamide, and the

mixture stirred at 110°C for 21 hours. The cooled mixture was filtered and the filtrate concentrated under reduced pressure, and the residue azeotroped with toluene.

The residue was dissolved in methanol (40mi), and poly (4-vinylpyridine) 2% cross-linked was added, the mixture stirred for an hour, then filtered. The filtrate was concentrated under reduced pressure, the residue dissolved in a minimum volume of dichloromethane, and added dropwise into diethyl ether (250mi), yielding an oil. The solvent was decanted off, the oi) re-dissoived in a minimum volume of dichioromethane and again added dropwise to diethyl ether. The resulting precipitate was filtered and dried to afford the title compound, 4. 3g.

1H-nmr (D2O, 270MHz) # : 1. 86-1. 97 (m, 2H), 2. 74-2.92 (m, 6H), 3.41 (m, 2H), 3. 83 (m, 4H), 6. 67 (m, 1 H), 8.01 (d, 1 H), 8. 06 (d, 1 H).

LRMS: m/z (TSP+) 266 [MHI Preparation 5 2-Fiuoro-5-formylbenzonitrile Iso-propyl magnesium bromide (1 8mi, 1 M in tetrahydrofuran, 18mmol) was added dropwise to an ice-cooled solution of 5-bromo-2-fluorobenzonitrile (3g, 15. 1mmol) in tetrahydrofuran (25ml), and once addition was complete, the mixture was allowed to warm to room temperature and stirred for a further 2 hours. N, N-Dimethylformamide (3. 5ml, 45. 2mmol) was added and the reaction stirred for 3 hours. Water was added and the mixture extracted with ethyl acetate (3x). The combined organic extracts were washed with 6% aqueous magnesium sulphate solution, and brine, then dried (Na2SO4) and concentrated under reduced pressure. The product was purified by column chromatography

on silica gel using an elution gradient of ethyl acetate: hexane (10 : 90 to 20 : 80) to afford the title compound as a light yellow solid, 1.01g.

1H-nmr (CDCl3) 8 : 3.27 (s, 3H), 6.83 (d, 1H), 7.83 (dd, 1H), 7.94 (d, 1H), 9.72 (s, 1H).

Preparation 6 (4-Isocyano-cyclohex-3-enyl)-benzene Potassium tert-butoxide (34. 3g, 306mol) was added portionwise to a solution of 4-phenyl-1-formamidocyctohexene (13. 6g, 68mmol) (Biiorg. Med. Chem; 8 ; 6; 2000; 1343) in tert-butanol (150mi), and the mixture stirred for 2 hours, with sufficient heating to ensure solution. Phosphorous oxychloride (7.82g, 51 mmol) was added dropwise, with cooling of the reaction vessel, and once addilon was complete, the reaction was stirred at room temperature for 24 hours. TLC analysis showed starting material remaining, so additional potassium tert- butoxide (3.8g, 34mmol) and phosphorous oxychloride (1. 57ml, 17mmol) were added, and the reaction stirred for a further 45 minutes. The mixture was concentrated under reduced pressure, the residue poured into brine (500moi) and extracted with dichloromethane (100ml, 3x50ml). The combined organic solutions were washed with water (100mI), brine (200ml), dried (Na2SO4) and evaporated under reduced pressure. The crude product was purified by column chromatography on silica gel using hexane: ethyl acetate (90: 10) as eluant to afford the title compound, 5. 8g.

'H nmr (CDCl3, 270MHz) # : 1.80-1. 90 (m, 1H), 1.97-2. 08 (m, 1H), 2. 23-2. 47 (m, 4H), 2. 72-2. 82 (m, 1H), 6.11 (s, 1H), 7.17-7. 34 (m, 5H).

Preparation 7 Methyl 3-amino-3-(3-methoxyphenyl)propanoate

A solution of P-amino-3-methoxy-benzenepropanoic acid (WO 0041469) (9. 38g, 52mmol) in concentrated hydrochloric acid (10ml) and methanol (115ml), was heated under reflux for 7 hours, then cooled. The reaction was evaporated under reduced pressure, and the residue partitioned between ethyl acetate (300ml) and 1 N sodium hydroxide solution (300ml). The layers were separated, and the organic phase evaporated under reduced pressure to afford the title compound as a colourless oil, 8.8g.

1H nmr (CDCl3, 400MHz 2.77 (m, 4H), 3. 68 (s, 3H), 3.80 (s, 3H), 4.42 (t, 1H), 6, 80 (m, 1 H), 6. 96 (m, 2H), 7.24 (m, 1 H).

LRMS: m/z (ES+) 232 [MNal Preparation 8 tert-Butvl (1 S)-3-amino-3-oxo-1-phenvloropvtcarbamate A mixture of 1-hydroxybenzotriazole hydrate (3.46g, 25. 6mmol), 1- (3- dimethylaminopropyl)-3-ethylcarbodiimide hydrochforide (4. 91g, 25. 6mmol), and (S)-N-tert-butoxycarbonyl-3-amino-3-phenylpropanoic acid (6. 80g,

25. 6mmol) in dichloromethane (250mi) was stirred at room temperature for 1 hour. 0.88 Ammonia (20ml) was added and the mixture stirred at room temperature for a further 18 hours. The resulting solid was filtered off and washed with sodium bicarbonate solution, then water and dried under vacuum at 50°C to afford the title compound, 6.78g.

'H nmr (CDCI3, 400MHz) 8 : 1.42 (s, 9H), 2.78 (s, 2H), 5.04 (m, 1 H), 5.35 (s, 1 H), 5.74 (s, 1 H), 5. 84 (s, 1 H), 7.24 (m, 5H).

LRMS : m/z (ES+) 287 [MNa+] [α]D = -38.73° (c = 0.25, methanol) Preparation 9 (3S)-3-Amino-3-phenvlpropanamide 4M Hydrochloric acid in dioxan (50ml) was added to a solution of the protected amine from preparation 8 (6.50g, 24. 6mmol) in methane ! (20m !), and the solution stirred at room temperature for 18 hours. The solution was evaporated under reduced pressure to give a white solid. This was dissolved in water (50ml), 1M sodium hydroxide (27ml, 27mmol) added and the solution allowed to stir for 18 hours at room temperature. The aqueous solution was extracted with dichloromethane and then ethyl acetate and the combined organic extracts were dried (Na2SO4) and evaporated under reduced pressure to afford the title compound as a white solid, 1. 32g.

1H nmr (DMSOd6, 400MHz) # : 2.32 (d, 3H), 4. 18 (t, 2H), 6.78 (s, 1H), 7.18 (m, 1H), 7.22-7. 40 (m, 5H).

LRMS : m/z (APCI) 165 [MH+] Preparation 10 Methyl 3-(3-methoxyphenyl)-3-[(4-methylbenzyl)amino]propanoate

A mixture of the amine from preparation 7 (8-. 8g, 42mol), p-tolualdehyde (5. 10g, 42mmol), acetic acid (1ml), and sodium triacetoxyborohydride (11. 57g, 55mmoi) in dichloromethane (500ml) was stirred at room temperature for 18 hours. 2N Hydrochloric acid (50m !) was added, and the solution stirred for a further 30 minutes. The reaction was basified using sodium bicarbonate solution, the phases separated, and the aqueous layer extracted with dichloromethane (500ml). The combined organic solutions were dried (Na2SO4) and evaporated under reduced pressure to give an oil. This was purified by. column chromatography on silica gel usingvan elution gradient of dichloromethane : methanol : 0. 88 ammonia (100: 0: 0 to 97.5 : 2.5 : 0.25) to afford the title compound, 7. 68g.

'H nmr (CDCl3, 400MHz) #: 2. 36. (s, 3H), 2. 70 (m, 1H), 2. 83 (m, 1 H), 3. 54 (d, 1H), 3. 63 (m, 4H), 3. 82 (s, 3H), 4.12 (m, 1H), 6. 82 (m, 1H), 6.99 (m, 2H), 7. 12 (d, 2H), 7.18 (d, 2H), 7.24 (m, 1H).....

LEMS : m/z (ES+) 336 [MNa+] Preparation 11 (3S)-3-[(4-Methylbenzyl)amino]-3-phenylpropanamide

The title compound was obtained in 62% yield from p-tolualdehyde and the amine from preparation 9, following the procedure decribed in preparation 10.

H nmr (CDCl3, 400MHz) # : 2.38 (s, 3H), 2.51 (dd, 1 H), 2.62 (dd, 1 H), 3. 58 (d, 1 H), 3.63 (m, 4H), 4. 03 (dd, 1 H), 5.40 (s, 1 H), 7.20-7. 40 (m, 6H).

LRMS : m/z (ES+) 269 [MH+] Preparation 1. 2 Methyl 3-(3-methoxyphenyl)-3-{(4-methylbenzyl)[(4-methyl-3- pyridinyl)carbonyl]amino}propanoate

Oxalyl chloride (731 mg, 5. 76mmol) and N, N-dimethylformamide (1 drop) were added to a solution of 4-methyinicotininc acid (500mg, 2. 88mmo !) in dichloromethane (25mi) and the solution stirred at room temperature for 4 hours. The reaction was evaporated under reduced pressure, to give 4-methyl- nicotinoyl chloride.

A solution of the freshly prepared acid chloride, the amine from preparation 10 (400mg, 1. 28mol), and N-ethyidiisopropylamine (0. 67ml, 3. 84mmol) in dichloromethane (50mut) was stirred at room temperature for 18 hours. The reaction was washed with sodium bicarbonate solution (50mi), water (50ml) then brine (50ml), dried (Na2S04) and evaporated under reduced pressure. The residue was purified by column chromatography on silica gel using an eluton gradient of dichloromethane : methanol : 0. 88 ammonia (100 : 0: 0 to 97.5 : 2.5 : 0.25) to afford the title compound, 289mg.

LRMS : m/z (ES+) 433 [MH+] Preparation 13 3-(3-Methoxyphenyl)-N-(4-methylbenzyl)-N-[(4-methyl-3-pyridi nyl)carbonyl]-ß- alahine Lithium hydroxide (30mg, 0. 74mmol) was added to a solution of the ester from preparation 12 (289mg, 0. 67mmol) in water (10ml) and tetrahydrofuran (10ml), and the mixture stirred at room temperature for 18 hours. The solution was concentrated under reduced pressure and the aqueous residue washed with ethyl acetate (20ml). The aqueous solution was acidified to pH 5 using 2N

hydrochloric acid, then extracted with ethyl acetate (20ml). This solution was dried (Na2SO4) and evaporated under reduced pressure. The residue was purified by column chromatography on silica gel using dichloromethane : methanol : 0.88 ammonia (100: 0: 0 to 97.5 : 2.5 : 0.25) to afford the title compound, 176mg.

HRMS : m/z (ES+) 418.2117 [MH+], C25H27N3O3 requires 418. 2125.

Preparation 14 5-Chloro-2- (2-t-butoxvcarbonylaminoethyl) amino-N-f2-amino-1-1, 4- benzodioxan-6-yl}-2-oxoethyl-N-(4-methylbenzyl)pyrimidine-4- carboxamide

The title compound was made from the sulfone from example 243 and N-Boc- ethylenediamine (commercially available from Aldrich) using the method of example 244 and was used immediately in the next step Preparation 15 Methvl-2-keto-3-methyl-4-dimethvlaminobut-3-enoate

Methyl-2-ketobutyrate (5g, 43mmol) was added at ambient temperature to dimethylformamide dimethyl acetal (5.13g, 43mmol, 5. 72ml). The solution was stirred overnight under nitrogen and then the volatile components were removed in vacuo at 40°C. The yellow residue was used without further purification.

1Hnmr (CDCl3, 400MHz) # : 2.0 (s, 3H), 3. 18 (s, 6H), 3.8 (s, 3H), 7.05-7. 15 (br s, 1H) LRMS : mlz (APCI+) 172 [MH+] Preparation 16 Methyl-5-methyl-2-methylthiopyrimidine-4-carboxylate Sodium metal (1.05g, 45. 5mmol) was added to methanol (40ml) at ambient temperature. After dissolution of the metal, S-methylisothiouronium sulfate (4.34g, 22. 8mmol) was added in one portion, followed by methyl-2-keto-3- methyl-4-dimethylaminobut-3-enoate (3.9g, 22. 8mmol, preparation 15) dropwise as a solution in 5ml of methanol. The reaction was stirred at ambient temperature for 1 hr and then heated to 50°C for 3hrs. After cooling, the solvent was evaporated. The residue was partitioned between ethyl acetate (50ml) and water (30m !). The organic phase was separated and washed with brine (20moi), dried over MgSO4, filtered and evaporated. The residue was purified by chromatography on silica using a gradient of ethyl acetate/pentane as eluent, starting with 95/5 and increasing to give the title compound as a solid, 0.9g, (20%).

1Hnmr (CDCl3, 400MHz) # : 2. 4 (s, 3H), 2. 58 (s,. 3H), 3. 98 (s, 3H), 8. 50 (s, AIH)

LRMS : m/z (APCl+) 199 [MH+] Preparation 17 5-methyl-2-methylthiopyrimidine-4-carboxylic acid Sodium hydroxide (0.37g, 4. 6mmol) was added dropwise as a solution in water (2ml) to a solution of the ester, from preparation 16, in methanol (5ml) at ambient temperature. After 20 minutes the : reaction mixture was acidified with 2M HCI and the methanol evaporated. The resulting aqueous suspension was extracted with CH2CI2 (3 x 10m The combined extracts were dried over MgS04, filtered and evaporated. The solid residue was broken up in ether and filtered to give the title compound as a solid, 0.57g, (67%).

1Hnmr (CDCis, 400MHz) # : 2. 25 (s, 3H), 2.45 (s, 3H), 8. 35 (s, 1 H) LRMS : m/z (APCl+) 135 [MH+]