Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
SUBSTITUTED QUINOLINES AS ANTITUMOR AGENTS
Document Type and Number:
WIPO Patent Application WO/2002/044166
Kind Code:
A1
Abstract:
The invention provides a compound of Formula (Ia), or a pharmaceutically acceptable salt, pro-drug or solvate thereof. The invention also provides a process for the preparation of a compound of Formula (Ia), pharmaceutical compositions of a compound of Formula (Ia) and methods for the treatment or prevention of cancer comprising administering an effective amount of a compound of Formula (1a).

Inventors:
BOYLE FRANCIS THOMAS (GB)
GIBSON KEITH HOPKINSON (GB)
FOOTE KEVIN MICHAEL (GB)
Application Number:
PCT/GB2001/004737
Publication Date:
June 06, 2002
Filing Date:
October 26, 2001
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
ASTRAZENECA AB (SE)
ASTRAZENECA UK LTD (GB)
BOYLE FRANCIS THOMAS (GB)
GIBSON KEITH HOPKINSON (GB)
FOOTE KEVIN MICHAEL (GB)
International Classes:
C07D217/26; A61K31/4709; A61K31/496; A61K31/5377; A61K31/541; A61K31/55; A61P35/00; A61P43/00; C07D215/54; C07D215/56; C07D401/12; C07D405/12; C07D409/12; C07D413/12; C07D417/12; C07D521/00; (IPC1-7): C07D401/12; A61K31/4709; A61P35/00; C07D215/54; C07D417/12
Domestic Patent References:
WO1998043960A11998-10-08
WO2000018740A12000-04-06
WO2000018761A12000-04-06
WO1998013350A11998-04-02
WO2000068201A12000-11-16
Attorney, Agent or Firm:
Bryant, Tracey (Global Intellectual Property Mereside, Alderley Par, Macclesfield Cheshire SK10 4TG, GB)
Download PDF:
Claims:
Claims
1. A compound of Formula (Ia), or a pharmaceutically acceptable salt, prodrug or solvate thereof ; wherein: nisOorl ; Y is selected fromNH,O,S, orNR7where R7 is alkyl of 16 carbon atoms; W is cyano, fluoro, chloro or bromo; R is cycloalkyl of 3 to 7 carbon atoms, which may be optionally substituted with one or more alkyl of 1 to 6 carbon atom groups ; or is a pyridinyl, pyrimidinyl, or phenyl ring; wherein the pyridinyl, pyrimidinyl, or phenyl ring may be optionally monodi, or trisubstituted with a substituent selected from the group consisting of halogen, alkyl of 16 carbon atoms, alkenyl of 26 carbon atoms, alkynyl of 26 carbon atoms, azido, hydroxyalkyl of 16 carbon atoms, halomethyl, alkoxymethyl of 27 carbon atoms, alkanoyloxymethyl of 27 carbon atoms, alkoxy of 16 carbon atoms, alkylthio of 16 carbon atoms, hydroxy, trifluoromethyl, cyano, nitro, carboxy, carboalkoxy of 27 carbon atoms, carboalkyl of 27 carbon atoms, phenoxy, phenyl, thiophenoxy, benzoyl, benzyl, amino, alkylamino of 16 carbon atoms, dialkylamino of 2 to 12 carbon atoms, phenylamino, benzylamino, alkanoylamino of 16 carbon atoms, alkenoylamino of 38 carbon atoms, alkynoylamino of 38 carbon atoms, and benzoylamino ; or R is a groupR8XR9 where Rg is a divalent cycloalkyl of 3 to 7 carbon atoms, which may be optionally further substituted with one or more alkyl of 1 to 6 carbon atom groups; or is a divalent pyridinyl, pyimidinyl, or phenyl ring; wherein the pyridinyl, pyrimidinyl, or phenyl ring may be optionally further substituted with one or more groups selected from halogen, alkyl of 16 carbon atoms, alkenyl of 26 carbon atoms, alkynyl of 26 carbon atoms, azido, hydroxyalkyl of 16 carbon atoms, halomethyl, alkoxymethyl of 27 carbon atoms, alkanoyloxymethyl of 27 carbon atoms, alkoxy of 16 carbon atoms, alkylthio of 16 carbon atoms, hydroxy, trifluoromethyl, cyano, nitro, carboxy, carboalkoxy of 27 carbon atoms, carboalkyl of 27 carbon atoms, phenoxy, phenyl, thiophenoxy, benzoyl, benzyl, amino, alkylamino of 16 carbon atoms, dialkylamino of 2 to 12 carbon atoms, phenylamino, benzylamino, alkanoylamino of 16 carbon atoms, alkenoylamino of 38 carbon atoms, alkynoylamino of 38 carbon atoms, and benzoylamino ; where X is selected fromNH,O,S, CH2 orNR7where R7 is alkyl of 16 carbon atoms, and R9 is a group (CH2) Rio where m is 0 or an integer of from 13 and Rl° is an optionally substituted aryl or optionally substituted cycloalkyl ring of up to 10 carbon atoms, or Rl° is a optionally substituted heterocyclic ring or an Noxide of any nitrogen containing ring ; Rl, R2, R4 are independently selected from hydrogen, hydroxy, halogeno, cyano, nitro, trifluoromethyl, C13alkyl,NR11R12 (wherein Rll and Rl2, which may be the same or different each represents hydrogen, or C13akyl), or a group R13Xl (CH2) x wherein x is 0 or an integer of from 1 to 3, Xl represents a direct bond,O,CH2, OC(O), C(O), S, SO, SO2, NR14C(O), NR14C(O)O, C(O)NR15, C(O)ONR15, SO2NR16, NR17SO2 NR18or NR18NR18 (wherein R14, R15, R16, R"and R"each independently represents hydrogen, C13alkyl or C13alkoxyC23alkyl), and R13 is hydrogen, optionally substituted hydrocarbyl, or optionally substituted heterocyclyl; and R3 is selected from (i) a group of formulaXlRx(OH) p where Xl is as defined above, Rx is an alkylene, alkenylene or alkynylene chain, optionally interposed with a heteroatom or a heteocyclic ring and p is 1 or 2; provided that when Rx is alkylene Ru must be interposed with a heteroatom or a heterocyclic ring and at least one (OH) p is situated on the alkylene chain between Xl and the interposed heteroatom or heterocyclic ring; (ii) a group of formula R13a(CH2)yX1(CH2)x where Rl3 ais as defined for R13 above, and Xl and x are as defined above, y is 0 or an integer between 1 and 5, wherein (CH2) y is optionally interposed by an Xl group; provided that Rl3a is selected from phenyl or optionally substituted heterocyclyl and the optional substituents for phenyl and aromatic heterocyclyl rings are selected from : C25alkenyl, C25alkynyl, C15alkanoyl, C15alkoxycarbonyl, C13alkanoylC13alkyl, C13alkoxyC13alkyl, C15alkylthio, C15alkylsulphanyl, C15alkylsulphonyl, N,Ndic15alkylamino, cyanoC15alkyl and the optional substituents for nonaromatic heterocyclyl rings further include hydroxyClsalkoxy, amino, aminoC15alkyl, NC15alkylamino, carboxy, cyano, CONRzzRzz'andNRzz" CORzz'" (wherein Rzz, Rzz'm, Rzz' and Rzz'" each independently represent hydrogen, C15alkyl or C13alkoxyC13alkyl) ; (iii) a group of formulaX1RyNRzRy'SRy" where X1 is as defined above, Ry, RY and RY are independently selected from alkyl, alkenyl or alkynyl chains, and R2 is hydrogen or alkyl, or RZ and Ry"are joined together to form an optionally substituted nitrogen and sulphur containing ring; provided that at least one of RY, RY and RY is an alkenyl or alkynyl chain; (iv) a group of formulaXlRxXla (C3 6cycloalkyl) where Xi is as defined above, Xla is as defined for Xl above and Rx is an alkylene, alkenylene or alkynylene chain, optionally interposed with a heteroatom provided that when Ru ils alkylene and a heteroatom is interposed adjacent to C36cycloalkyl, C36cycloalkyl does not include cyclopentyl or cyclohexyl; (v) a group of the formula X1C15alkyl where Xl is as defined above and C15alkyl is substituted by one more substituents independently selected from chloro and cyano; (vi) a group of the formulaX1c13alkylCONR18NR18R20 where R18 is as defined above and R20 is selected from hydrogen or Ci5alkoxycarbonyl ; or (vii) a heterocyclic ring.
2. A compound of Formula (I) or a pharmaceutically acceptable salt, prodrug or solvate thereof; wherein: n is 0 or 1 ; Y is selected fromNH,O,S, orNR7where R7 is alkyl of 16 carbon atoms; W is cyano, fluoro, chloro or bromo; R6 is cycloalkyl of 3 to 7 carbon atoms, which may be optionally substituted with one or more alkyl of 1 to 6 carbon atom groups; or is a pyridinyl, pyrimidinyl, or phenyl ring; wherein the pyridinyl, pyrimidinyl, or phenyl ring may be optionally monodi, or trisubstituted with a substituent selected from the group consisting of halogen, alkyl of 16 carbon atoms, alkenyl of 26 carbon atoms, alkynyl of 26 carbon atoms, azido, hydroxyalkyl of 16 carbon atoms, halomethyl, alkoxymethyl of 27 carbon atoms, alkanoyloxymethyl of 27 carbon atoms, alkoxy of 16 carbon atoms, alkylthio of 16 carbon atoms, hydroxy, trifluoromethyl, cyano, nitro, carboxy, carboalkoxy of 27 carbon atoms, carboalkyl of 27 carbon atoms, phenoxy, phenyl, thiophenoxy, benzoyl, benzyl, amino, alkylamino of 16 carbon atoms, dialkylamino of 2 to 12 carbon atoms, phenylamino, benzylamino, alkanoylamino of 16 carbon atoms, alkenoylamino of 38 carbon atoms, alkynoylamino of 38 carbon atoms, and benzoylamino; or R is a groupR8XR9 where R8 is a divalent cycloalkyl of 3 to 7 carbon atoms, which may be optionally further substituted with one or more alkyl of 1 to 6 carbon atom groups; or is a divalent pyridinyl, pyimidinyl, or phenyl ring; wherein the pyridinyl, pyrimidinyl, or phenyl ring may be optionally further substituted with one or more groups selected from halogen, alkyl of 16 carbon atoms, alkenyl of 26 carbon atoms, alkynyl of 26 carbon atoms, azido, hydroxyalkyl of 16 carbon atoms, halomethyl, alkoxymethyl of 27 carbon atoms, alkanoyloxymethyl of 27 carbon atoms, alkoxy of 16 carbon atoms, alkylthio of 16 carbon atoms, hydroxy, trifluoromethyl, cyano, nitro, carboxy, carboalkoxy of 27 carbon atoms, carboalkyl of 27 carbon atoms, phenoxy, phenyl, thiophenoxy, benzoyl, benzyl, amino, alkylamino of 16 carbon atoms, dialkylamino of 2 to 12 carbon atoms, phenylamino, benzylamino, alkanoylamino of 16 carbon atoms, alkenoylamino of 38 carbon atoms, alkynoylamino of 38 carbon atoms, and benzoylamino; where X is selected fromNH,O,S, CH2 orNR7where R7 is alkyl of 16 carbon atoms, and R9 is a group (CH2)mR10 where m is 0 or an integer of from 13 and Rl° is an optionally substituted aryl or optionally substituted cycloalkyl ring of up to 10 carbon atoms, or Rl° is a optionally substituted heterocyclic ring or an Noxide of any nitrogen containing ring ; Rl, R2, R4 are independently selected from hydrogen, hydroxy, halogeno, cyano, nitro, trifluoromethyl, CI3alkyl,NR"R (wherein Rl l and Rl2, which may be the same or different each represents hydrogen, or C13alkyl), or a group R13X1(CH2)x wherein x is 0 or an integer of from 1 to 3, Xl represents a direct bond,O,CH2, OC(O), C(O), S, SO, SO2M, NR14C(O), NR14C(O)O, C(ONR15, C(O)ONR15M, SO2NR16, NR17SO2 or NR18 (wherein R14, R15, R16, R17 an dR18 each independently represents hydrogen, C13alkyl or Ci3alkoxyC23alkyl)), and R13 is hydrogen, optionally substituted hydrocarbyl, or optionally substituted heterocyclyl; and R3 is selected from (i) a group of formulaX1Rx(OH) p where Xl is as defined above, Rx is an alkylene, alkenylene or alkynylene chain, optionally interposed with a heteroatom or a heteocyclic ring and p is 1 or 2; provided that when Rx is alkylene Ru must be interposed with a heteroatom or a heterocyclic ring and at least one (OH) p is situated on the alkylene chain between Xl and the interposed heteroatom or heterocyclic ring; (ii) a group of formula R13aX20(CH2)x where R13a is as defined for R13 above and x is as defined above and X20 is a group NR14C(O), C(O)NR15 or NR18 where R14, R15 and Ria axe as defined above; provided that R 13a is selected from phenyl or optionally substituted heterocyclyl and the optional substituents for phenyl and aromatic heterocyclyl rings are selected from: C25alkenyl, hydroxyC25alkenyl, C25alkynyl, C15alkanoyl, C1 5alkoxycarbonyl, C13alkanoylC13alkyl, C13alkoxyC13alkyl, C15alkylthio, C1 5alkylsulphanyl, C15alkylsulphonyl, NC15alkylamino, N,NdiC15alkylamino, guanidino, nitro, cyanoCl5alkyl and aryl and the optional substituents for non aromatic heterocyclyl rings further include hydroxyC15alkoxy, amino, amino 5alkyl, NC1salkylamino, carboxy, cyano, CONRzzRzz' and NRzz" CORzz''' (wherein Rzz, Rzz' aNd Rzz'' and Rzz''' each independently represent hydrogen, Ci 5alkyl or Ci3alkoxyCi3alkyl) ; (iii) a group of formula X1RyNRzRy'SRy'' where X1 is as defined above, RY, Ry' and RY are independently selected from alkyl, alkenyl or alkynyl chains, and Rz is hydrogen or alkyl, or RZ and RY are joined together to form an optionally substituted nitrogen and sulphur containing ring; provided that when Rz and RY are joined RY is an alkenyl or alkynyl chain and when Rz and RY are not joined at least one of Ry, RY and RY is an alkenyl or alkynyl chain; or (iv) a group of formulaX1Rz' (C46cycloalkyl) where Xl is as defined above and Ru ils an alkylene, alkenylene or alkynylene chain, optionally interposed with a heteroatom provided that when Rx' is alkylene and a heteroatom is interposed adjacent to C46cycloalkyl, C46cycloalkyl does not include cyclopentyl or cyclohexyl.
3. A compound according to claim 1 or claim 2 wherein Rs is cyano.
4. A compound according to claim 1 or 3 wherein R3 is selected from group (i).
5. A compound according to claim 4 wherein Xl is selected fromO,NR14C (O), or NRI8 (wherein R14 and Rl8 are as defined above), X isOand Rx is C16alkylene group interposed withNR75where R7s is hydrogen or C14alkyl (wherein C14alkyl is optionally substituted by hydroxy).
6. A compound according to any one of claims 1 to 3 wherein R3 is selected from group (ii).
7. A compound according to claim 6 when dependent on claim 1 wherein Xl is selected from O, NR14C(O), or NR18 (wherein R14 and Rl8 are as defined above),, X isO, x is 0, y is an integer between 1 and 5, Rs is cyano and Rl3a is selected from azetidinyl, pyrrolidinyl, tetrahydrofuranyl, 1,3dioxolanyl, 1,3oxazolidinyl, 1,2,4oxadiazolidinyl, 1, 1dioxoctetrahydrothiophenyl, morpholinyl, piperidinyl, piperazinyl, 1,3dioxanyl, tetrahydropyranyl, 1, 1dioxotetrahydrothiopyranyl, thiomorpholinyl, 1oxo thiomorpholinyl, 1, ldioxo thiomorpholinyl, 2,3dihydropyrrolyl, imidazolyl, pyrazolyl, 1,3thiazolyl, 1,3,4oxadiazolyl, isoxazolyl, 1,2,4thiadiazolyl, furanyl, 2,5dihydro 1,2,4oxadiazolyl and pyrimidinyl.
8. A compound according to claim 7 wherein Xl isOand Rl3a is selected from: 4methoxyethylpiperazin1yl, 4acetylpiperazin1yl, 3methylsulphonylpyrrolidin1yl and 4ethylsulphonylpiperazin1yl.
9. A compound according to claim 1 or claim 3 where R3 is selected from group (iv).
10. A compound according to claim 9 wherein Xl is selected fromO,NR14C (O), or NRI8 (wherein R14 and R18 are as defined above), X is O, X1a is NR18(wherein R18 is as defined above) and R8 is C15alkylen.
11. A compound according to claim 10 wherein Xl isOand C36cycloalkyl is selected from: cyclopropyl and cyclobutyl.
12. A compound according to any one of the preceding claims where the substituent on Rlo is a group of the subformula (ii): Z(CH2)pR100R101 (ii) wherein Zis a direct bond or a group of subformula (iii) X121R751X131(R761X141)s(R771X151)s (iii) wherein Xl2l, Xl3l each X141 and each X'51 are independently selected fromO,C (O), C(O)O, S, SO, SO2, NR781C(O), NR781C(O)O, CONR791, C(O)ONR791, SO2NR301, NR811 SO2 or NR821(wherein R781, R791, R801, R8ll and R821 each independently represents hydrogen, C13alkyl optionally substituted by hydroxy, or Ci3alkoxyC23alkyl) and each X131m, X141 and Xls may additionally be a direct bond; s and s'are independently selected from 0,1,2 or 3; R751 R761 and R771 are independently selected from C16alkylene, C26alkenylene or C26alkynylene optionally substituted by halo and hydroxy or R751, R761 and R771 can each independently be direct bonds; Rloo is an optionally substituted divalent heterocyclic group, C15alkylene, or divalent C37cycloalkyl, Rlol is hydrogen, amino or a group of subformula (iv) X161R831(X171R341)tX181R851 (iv) wherein X161, XIII and each Xl7l are each independently selected from a direct bond,0, C(O), SM, SO, SO2, NR361C(O), NR361C(O)O, CONR871, C(O)ONR871M, SO2NR881, NR891SO2 or NR901(wherein R361 R371, R881, R391 and Wol each independently represents hydrogen, C13alkyl or C13alkoxyC23alkyl), R831 and each R341 are independently selected from C16alkylene, C26alkenylene or C26alkynylene, R851 is selected from hydrogen, C16alkyl, C26alkenyl or C26alkynyl, and tis0, 1,2 or 3; and pis 0, 1, 2or3.
13. A compound selected from: 7 (3azetidin1yl2oxopropoxy)6methoxy4 { [4 (2 methoxyphenoxy) phenyl] amino}quinoline3carbomitrile ; 7 {3 [4 (ethylsulfonyl) piperazin1yl] propoxy}6methoxy4 { [4 (2 methoxyphenoxy) phenyl] amino} quinoline3carbonitrile ; 6methoxy7{3[4(2methoxyethyl0piperain1yl]propoxy}4{ [4(2 methoxyphenoxy) phenyl] amino} quinoline3carbonitrile; 6methoxy4{[4(2methoxyphenoxy) phenyl] amino}7 {3 [3 (methylsulfonyl) pyrrolidin1yl] propoxy} quinoline3carbonitrile ; and 7 [3 (4acetylpiperazin1yl) propoxy]6methoxy4 { [4 (2 methoxyphenoxy) phenyl] amino} quinoline3carbonitrile; 73[bis(2hydroxyethyl)amino]propoxy}6methoxy4{[4(2 methoxyphenoxy) phenyl] amino} quinoline3carbonitrile; 7[3(cyclopropylamio)propoxy]4{[2fluoro4(1, 3thiazol2 yloxy) phenyl] amino}6methoxyquinoline3carbonitrile ; 7 [3(cyclopropylamino) propoxy]4 { [2fluoro4(isothiazol3yloxy) phenyl] amino} 6methoxyquinoline3carbonitrile ; 2{2[4({3cyano7[3(cyclopropylamino)propoxy]6methoxyquionlin4 yl} amino) phenoxy] phenoxy}Nmethylacetamide ; and 7[3(cyclopropylamino_propoxy]6methoxy4{[4(2 methoxyphenoxy) phenyl] amino} quinoline3carbonitrile ; and pharmaceuticalacceptable salts, prodrugs or solvate thereof.
14. A process for the preparation of a compound of Formula (I) or a compound of Formula (Ia) by reacting a compound of Formula (III) where Rl, R2', R3, R4' represent R1m, R2, R3 and R4 respectively as defined in relation to Formula (1) or Formula (Ia) or a precursor thereof, Rs is as defined in relation to Formula (1) or Formula (Ia) and Z'is a leaving group, with a compound of Formula (IV) H Y(CH2)nR6' (IV) where Y, and n are as defined in realtion of Formula (UI) or Formula (Ia), and R6' is a group R as defined in relation to Formula (I) or Formula (Ia) or a precursor thereof; and thereafter if necessary or desired converting precursor groups Rl, R2', R3', R4 and R6 to groups of formula Rl, R2, R3, R4 and R6 respectively, or converting a group R1, R2, R3, R4 and R6 to a different such group.
15. A compound according to any one of claims 1 to 13 or a pharmaceutically acceptable salt, prodrug or solvate thereof for use in the method of treatment of the human of animal body by therapy.
16. The use of a compound according to any one of claims 1 to 13 or a pharmaceutically acceptable salt, prodrug or solvate thereof in the manufacture of a medicament for use as an antiproliferative agent in the containment and/or treatment of solid tumour disease.
17. SO2, NR14C(O), NR14C(O)O, C(O)NR15, C(O)ONR15, SO2NR16M, NR17SO2 NR18 or NR18NR18 (wherein R14, R15, R16, R.
18. and Rl8 each independently represents hydrogen, C13alky or c13alkoxyC23alkyl)), and R13 is hydrogen, optionally substituted hydrocarbyl, or optionally substituted heterocyclyl; and R3 is selected from (i) a group of formulaX'W (OH) p where Xl is as defined above, Rx is an alkylene, alkenylene or alkynylene chain, optionally interposed with a heteroatom or a heteocyclic ring and p is 1 or 2 ; (ii) a group of formula R13a(CH2)yX1(CH2)x where R13 a is as defined for R13 above, and X1 and x are as defined above, y is 0 or an integer between 1 and 5, wherein (CH2) y is optionally interposed by an Xi group; (iii) a group of formulaXlRYNRZRYSRY where Xl is as defined above, RY, RY and RY are independently selected from alkyl, alkenyl or alkynyl chains, and Rz is hydrogen or alkyl, or Rz and RY are joined together to from an optionally substituted nitrogen and sulphur containing ring; (iv) a group of formulaX1Rx' (C36cycloalkyl) where Xl is as defined above and Ru ils an alkylene, alkenylene or alkynylene chain, optionally interposed with a heteroatom a group of the formulaX'C15alkyl where Xl is as defined above and c15alkyl is substituted by one more substituents independently selected from chloro and cyano; (v) a group of the formulaX1C13alkylCONR18NR18R20 where Rl8 is as defined above and R20 is selected from hydrogen or C15alkoxycarbonyl ; or (vi) a heterocyclic ring. The invention also provides a process for the preparation of a compound of Formula (Ia), pharmaceutical compositions of a compound of Formula (Ia) and methods for the treatment or prevention of cancer comprising administering an effective amount of a compound of Formula (la).
Description:
SUBSTITUTED QUINOLINES AS ANTITUMOR AGENTS The present invention relates to certain novel quinoline derivatives as well as to their use as pharmaceuticals, in particular as inhibitors of specific kinase enzymes, such as MEK enzymes. Further aspects of the invention include pharmaceutical compositions and methods of treatment of proliferative disease such as cancer using said compounds.

Cancer is a disease in which cells grow and divide in an uncontrolled fashion. This uncontrolled growth arises from abnormalities in signal transduction pathways that are used by normal cells to regulate cell growth and division in response to various signalling molecules. Normal cells do not proliferate unless stimulated to do so by specific signal molecules located outside the cell derived from nearby cells or tissues. Growth factors bind to the cell membrane via specific receptors which have intrinsic enzyme activity. These receptors relay the growth signal to the cell nucleus via a series of signalling proteins. In cancer, a number of defects in signal pathways are apparent. For example, cancer cells may produce their own growth factors which bind to their cognate receptors, resulting in an autocrine loop, or receptors may be mutated or overexpressed leading to an increased, continuous signal to proliferate. In addition, negative regulators of cell growth may be lost.

Oncogenes are cancer related genes which often encode abnormal versions of signal pathway components, such as receptor tyrosine kinases, serine-threonine kinases, or downstream signaling molecules such as the ras genes, which code for closely related small guanine nucleotide binding proteins which hydrolyse bound guanosine triphosphate (GTP) to guanosine diphosphate (GDP). Ras proteins are active in promoting cell growth and transformation when they are bound to GTP and inactive when they are bound to GDP.

Transforming mutants of p2lras are defective in their GTPase activity and hence remain in the active GTP bound state. The ras oncogene is known to play an integral role in certain cancers, and has been found to contribute to the formation of over 20% of all cases of human cancer.

When activated by ligand, cell surface receptors which are coupled to the mitogenic response, such as growth factor receptors, initiate a chain of reactions which leads to the activation of guanine nucleotide exchange activity on ras. When in its active GTP-bound state, a number of proteins interact directly with ras at the plasma membrane resulting in signal transmission through several distinct pathways. The best characterised effector protein is the product of the raf proto-oncogene. The interaction of raf and ras is a key regulatory step in the control of cell proliferation. Ras-mediated activation of the raf serine-threonine kinase in turn

activates the dual-specificity MEK (MEK1 and MEK2), which is the immediate upstream activator of mitogen activated protein kinase (MAPKs known as extracellular signal regulated protein kinases or ERK1 and ERK2). To date, no substrates of MEK other than MAPK have been identified, though recent reports indicate that MEK may also be activated by other upstream signal proteins such as MEK kinase or MEKK1 and PKC. Activated MAPK translocates and accumulates in the nucleus, where it can phosphorylate and activate transcription factors such as Elk-1 and Sapla, leading to the enhanced expression of genes such as that for c-fos.

The ras-dependent raf-MEK-MAPK cascade is one of the key signalling pathways responsible for transmitting and amplifying mitogenic signals from cell surface to the nucleus resulting in changes in gene expression and cell fate. This ubiquitous pathway appears essential for normal cell proliferation and constitutive activation of this pathway is sufficient to induce cellular transformation. Transforming mutants of p2lras are constitutively active, resulting in raf, MEK and MAPK activity and cell transformation. Inhibition of MEK activity using either antisense raf, a dominant negative MEK mutant or the selective inhibitor PD098059 have been shown to block the growth and morphological transformation of ras-transformed fibroblasts.

The mechanism of activation of raf, MEK and MAPK is through phosphorylation on specific serine, threonine or tyrosine residues. Activated raf and other kinases phosphorylate MEK1 on S218 and S222 and MEK2 on S222 and S226. This results in MEK activation and subsequent phosphorylation and activation of ERK1 on T190 and Y192 and ERK2 on T 183 and Y185 by the dual specificity MEKs. Whilst MEK can be activated by a number of protein kinases, and active MAPKs phosphorylate and activate a number of substrate proteins including transcription factors and other protein kinases, MEKs appear specific and sole activators of MAPKs and could act as a focal point for cross-cascade regulation. MEK1 and MEK2 isoforms show unusual specificity and also contain a proline-rich insert between catalytic subdomains IX and X which is not present in any of the other known MEK family members. These differences between MEK and other protein kinases, together with the known role of MEK in proliferative signalling suggest that it may be possible to discover and employ selective MEK inhibitors as therapeutic agents for use in proliferative disease.

WO 98/43960 discloses a range of 3-cyano quinoline compounds and their use in the treatment of cancer. Certain of the compounds are demonstrated as being inhibitors of Epidermal Growth Factor Receptor Kinase, and to inhibit cancer cell growth. Other

quinoline derivatives which inhibit the effect of growth factors such as VEGF are described in <BR> <BR> W098/13350.<BR> <BR> <P> Copending International patent application nos PCT/GBOO/01697, PCT/GBOO/01707 and PCT/GB00/01698 describe certain quinoline derivatives which are inhibitors of the kinase activity of MEK and as a result, can produce therapeutically useful effects in the treatment of proliferative disease and in particular cancer. The applicants have found that certain particular compounds of this type, where the substituent at the 7-position on the quinoline ring has certain characteristics, produce particularly good results.

According to the first feature of the present invention there is provided a compound of Formula (I) or a pharmaceutically acceptable salt, pro-drug or solvate thereof ; wherein: n is 0 or 1 ; Y is selected from-NH-,-O-,-S-, or-NR7-where R7 is alkyl of 1-6 carbon atoms; R5 is cyano, fluoro, chloro or bromo; R6 is cycloalkyl of 3 to 7 carbon atoms, which may be optionally substituted with one or more alkyl of 1 to 6 carbon atom groups; or is a pyridinyl, pyrimidinyl, or phenyl ring ; wherein the pyridinyl, pyrimidinyl, or phenyl ring may be optionally mono-di-, or tri-substituted with a substituent selected from the group consisting of halogen, alkyl of 1-6 carbon atoms, alkenyl of 2-6 carbon atoms, alkynyl of 2-6 carbon atoms, azido, hydroxyalkyl of 1-6 carbon atoms, halomethyl, alkoxymethyl of 2-7 carbon atoms, alkanoyloxymethyl of 2-7 carbon atoms, alkoxy of 1-6 carbon atoms, alkylthio of 1-6 carbon atoms, hydroxy, trifluoromethyl, cyano, nitro, carboxy, carboalkoxy of 2-7 carbon atoms, carboalkyl of 2-7 carbon atoms, phenoxy, phenyl, thiophenoxy, benzoyl, benzyl,

amino, alkylamino of 1-6 carbon atoms, dialkylamino of 2 to 12 carbon atoms, phenylamino, benzylamino, alkanoylamino of 1-6 carbon atoms, alkenoylamino of 3-8 carbon atoms, alkynoylamino of 3-8 carbon atoms, and benzoylamino ; or R6 is a group-R8-X-R9 where R8 is a divalent cycloalkyl of 3 to 7 carbon atoms, which may be optionally further substituted with one or more alkyl of 1 to 6 carbon atom groups; or is a divalent pyridinyl, pyimidinyl, or phenyl ring; wherein the pyridinyl, pyrimidinyl, or phenyl ring may be optionally further substituted with one or more groups selected from halogen, alkyl of 1-6 carbon atoms, alkenyl of 2-6 carbon atoms, alkynyl of 2-6 carbon atoms, azido, hydroxyalkyl of 1-6 carbon atoms, halomethyl, alkoxymethyl of 2-7 carbon atoms, alkanoyloxymethyl of 2-7 carbon atoms, alkoxy of 1-6 carbon atoms, alkylthio of 1-6 carbon atoms, hydroxy, trifluoromethyl, cyano, nitro, carboxy, carboalkoxy of 2-7 carbon atoms, carboalkyl of 2-7 carbon atoms, phenoxy, phenyl, thiophenoxy, benzoyl, benzyl, amino, alkylamino of 1-6 carbon atoms, dialkylamino of 2 to 12 carbon atoms, phenylamino, benzylamino, alkanoylamino of 1-6 carbon atoms, alkenoylamino of 3-8 carbon atoms, alkynoylamino of 3-8 carbon atoms, and benzoylamino; where X is selected from-NH-, -O-, -S-, CH2 or-NR7-where R7 is alkyl of 1-6 carbon atoms, and is a group (CH2) mRI° where m is 0, or an integer of from 1-3 and Rlo is an optionally substituted aryl or optionally substituted cycloalkyl ring of up to 10 carbon atoms, or Rlo is a optionally substituted heterocyclic ring or an N-oxide of any nitrogen containing ring; Ru, R2, R4 are independently selected from hydrogen, hydroxy, halogeno, cyano, nitro, trifluoromethyl, C1-3alkyl, -NR11R12- (wherein R11 and R12, which may be the same or different each represents hydrogen, or C1-3alkyl), or a group R13-Xl- (CH2) x wherein x is 0 or an integer of from 1 to 3, Xl represents a direct bond, -O-, -CH2-, -OC (O)-,-C (O)-,-S-, -SO-, -SO2-, -NR14C(O)-, -NR14C(O)O-, -C(O)NR15-, -C(O)ONR5-, -SO2NR16-, -NRI7So2-or-NR18- (wherein Rl4, Rl5, Rl6, Rl7 and Rl8 each independently represents hydrogen, C1-3alkyl or Ci-3aIkoxyC2-3alkyl)), and R13 is hydrogen, optionally substituted hydrocarbyl, or optionally substituted heterocyclyl; and R3 is selected from (i) a group of formula-Xl-Rx-(OH) p where Xl is as defined above, Rx is an alkylene, alkenylene or alkynylene chain, optionally interposed with a heteroatom or a heteocyclic ring and p is 1 or 2; provided that when Rx is alkylene Ru must be interposed with a heteroatom or a heterocyclic ring and at least one (OH) p is situated on the alkylene chain between XI and the interposed heteroatom or heterocyclic ring; (ii) a group of formula R13a-X20-(CH2)x where Rl3a is as defined for R13 above and x is as defined above and X20 is a group -NR14C(O)-, -C(O)NR15- or -NR18-where R14, R15 and R'8 are as defined above; provided Rl3a is selected from phenyl or optionally substituted heterocyclyl and the optional substituents for phenyl and aromatic heterocyclyl rings are selected from: Cz- salkenyl, hydroxyC2-5alkenyl, C2-5alkynyl, Cl-salkanoyl, Cl-5allcoxycarbonyl, C1-3alkanoylC1-3alkyl, C1-3alkoxyC1-3alkyl, C1-5alkylthio, C1-5alkylsulphanyl, C1-5alkylsulphonyl, N-C1-5alkylamino, N,N-di-C1-5alkylamino, guanidino, nitro, cyanoCl-5alkyl and aryl and the optional substituents for non-aromatic heterocyclyl rings further include hydroxyCi-salkoxy, amino, aminoC1-5alkyl, N-C1-5alkylamino, carboxy, cyano, -CONRzzRzz' and-NRzz''CORzz"' (wherein Rzz, Rzz', Rzz" and Rzz"' each independently represent hydrogen, C1-5alkyl or Ci-BalkoxyCi-salkyI) ; (iii) a group of formula -X1-Ry-NRz-Ry'-S-Ry" where X1 is as defined above, Ry, RY and Ry"are independently selected from alkyl, alkenyl or alkynyl chains, and Rz is hydrogen or alkyl, or Rz and Ru are joined together to form an optionally substituted nitrogen and sulphur containing ring; provided that when Rz and RY are joined RY is an alkenyl or alkynyl chain and when RZ and RY are not joined at least one of Ry, RY and RY is an alkenyl or alkynyl chain; or (iv) a group of formula-Xl-Rx-(C46cycloalkyl) where Xl is as defined above and Ru ils an alkylene, alkenylene or alkynylene chain, optionally interposed with a heteroatom provided that when Ru ils alkylene and a heteroatom is interposed adjacent to C4-6cycloalkyl,C4-6cycloalkyl does not include cyclopentyl or cyclohexyl.

According to a further aspect of the first feature of the present invention there is provided a compound of Formula (1), or a pharmaceutically acceptable salt, pro-drug or solvate thereof, wherein R3 is selected from group (i), (ii) or (iv).

According to a second feature of the present invention there is provided a compound of Formula (Ia), or a pharmaceutically acceptable salt, pro-drug or solvate thereof; wherein: n is 0 or 1 ; Y is selected from-NH-,-O-,-S-, or-NR7-where R7 is alkyl of 1-6 carbon atoms; W is cyano, fluoro, chloro or bromo; R is cycloalkyl of 3 to 7 carbon atoms, which may be optionally substituted with one or more alkyl of 1 to 6 carbon atom groups; or is a pyridinyl, pyrimidinyl, or phenyl ring; wherein the pyridinyl, pyrimidinyl, or phenyl ring may be optionally mono-di-, or tri-substituted with a substituent selected from the group consisting of halogen, alkyl of 1-6 carbon atoms, alkenyl of 2-6 carbon atoms, alkynyl of 2-6 carbon atoms, azido, hydroxyalkyl of 1-6 carbon atoms, halomethyl, alkoxymethyl of 2-7 carbon atoms, alkanoyloxymethyl of 2-7 carbon atoms, alkoxy of 1-6 carbon atoms, alkylthio of 1-6 carbon atoms, hydroxy, trifluoromethyl, cyano, nitro, carboxy, carboalkoxy of 2-7 carbon atoms, carboalkyl of 2-7 carbon atoms, phenoxy, phenyl, thiophenoxy, benzoyl, benzyl, amino, alkylamino of 1-6 carbon atoms, dialkylamino of 2 to 12 carbon atoms, phenylamino, benzylamino, alkanoylamino of 1-6 carbon atoms, alkenoylamino of 3-8 carbon atoms, alkynoylamino of 3-8 carbon atoms, and benzoylamino ; or R is a group-R8-X-R9 where R8 is a divalent cycloalkyl of 3 to 7 carbon atoms, which may be optionally further substituted with one or more alkyl of 1 to 6 carbon atom groups; or is a divalent pyridinyl, pyimidinyl, or phenyl ring; wherein the pyridinyl, pyrimidinyl, or phenyl ring may be optionally further substituted with one or more groups selected from halogen, alkyl of 1-6 carbon atoms, alkenyl of 2-6 carbon atoms, alkynyl of 2-6 carbon atoms, azido, hydroxyalkyl of 1-6 carbon atoms, halomethyl, alkoxymethyl of 2-7 carbon atoms, alkanoyloxymethyl of 2-7 carbon atoms, alkoxy of 1-6 carbon atoms, alkylthio of 1-6 carbon atoms, hydroxy, trifluoromethyl, cyano, nitro, carboxy, carboalkoxy of 2-7 carbon

atoms, carboalkyl of 2-7 carbon atoms, phenoxy, phenyl, thiophenoxy, benzoyl, benzyl, amino, alkylamino of 1-6 carbon atoms, dialkylamino of 2 to 12 carbon atoms, phenylamino, benzylamino, alkanoylamino of 1-6 carbon atoms, alkenoylamino of 3-8 carbon atoms, alkynoylamino of 3-8 carbon atoms, and benzoylamino; where X is selected from -NH-, -O-, -S-, CH2 or -NR7'- where R7' is alkyl of 1-6 carbon atoms, and R9 is a group (CH2) mR10 where m is 0, or an integer of from 1-3 and Rio is an optionally substituted aryl or optionally substituted cycloalkyl ring of up to 10 carbon atoms, or Rio is a optionally substituted heterocyclic ring or an N-oxide of any nitrogen containing ring; Rl, R2, R4 are independently selected from hydrogen, hydroxy, halogeno, cyano, nitro, trifluoromethyl, CI-3alkyl,-NR R- (whereinR"andR, whichmaybe the same or different each represents hydrogen, or C1-3alkyl), or a group Rl3-Xl-(CH2) x wherein x is 0 or an integer of from 1 to 3, Xl represents a direct bond, -O-, -CH2-, -OC (O)-,-C (O)-,-S-, -SO-,-SO2-,-NRIaC (O)-,-NR14C (0) 0-,-C (O) NRis-,-C () ONRIS-,-SO2NRt6-, -NR17SO2- -NR18- or -NR18NR18- (wherein R, Ris, Rl6, R and Rl8 each independently represents hydrogen, C1-3alkyl or C1-3alkoxyC2-3alkyl)), and R13 is hydrogen, optionally substituted hydrocarbyl, or optionally substituted heterocyclyl; and R3 is selected from (i) a group of formula-X'-W- (OH) p where Xl is as defined above, Rx is an alkylene, alkenylene or alkynylene chain, optionally interposed with a heteroatom or a heteocyclic ring and p is 1 or 2; provided that when Rx is alkylene Ru must be interposed with a heteroatom or a heterocyclic ring and at least one (OH) p is situated on the alkylene chain between X and the interposed heteroatom or heterocyclic ring; (ii) a group of formula R13a-(CH2)y-X1-(CH2)x where Rl3 a is as defined for Rl3 above, and Xl and x are as defined above, y is 0 or an integer between 1 and 5, wherein (CH2) y is optionally interposed by an Xl group; provided that Rl3a is selected from phenyl or optionally substituted heterocyclyl and the optional substituents for phenyl and aromatic heterocyclyl rings are selected from: C2-5alkenyl,C2-5alkynyl, C1-5alkanoyl, C1-5alkoxycarbonyl, C1-3alkanonylC1-3alkyl, C1-3alkoxyC1-3alkyl, C1-5alkylthio, C1-5alkylsulphanyl, C1-5alkylsuphonyl, N, N-di-C1-5alkylamino, cyanoC1-5alkyl and the optional substituents for non-aromatic rings further include hydroxyCl-5alkoxy, amino, aminoCI-salkyl, N-Cl-salkylamino, carboxy, cyano, -CONRzzRzz' and -NRzz" CO Rzz"' (wherein RZZ, RZZ, RZZ and RZZ- each independently represent hydrogen, C1-5alkyl or C1-3alkoxyC1-3alkyl) ; (iii) a group of formula-Xl-RY-NRZ-RY-S-RY where Xl is as defined above, Ry, Ry'and Ru are independently selected from alkyl, alkenyl or alkynyl chains, and Rz is hydrogen or alkyl, or Rz and RY are joined together to form an optionally substituted nitrogen and sulphur containing ring; provided that at least one of RY, RY and RY is an alkenyl or alkynyl chain; (iv) a group of formula-Xl-Rx-(C36cycloalkyl) where Xl is as defined above and Ru ils an alkylene, alkenylene or alkynylene chain, optionally interposed with a heteroatom provided that when Ru ils alkylene and a heteroatom is interposed adjacent to C3-6cycloalkyl, C3-6cycloalkyl does not include cyclopentyl or cyclohexyl; (v) a group of the formula-Xl-Cl-5alkyl where Xl is as defined above and C1-5alkyl is substituted by one more substituents independently selected from chloro and cyano; (vi) a group of the formula -X1-C1-3alkyl-CO-NR18NR18-R20 where Rl3 is as defined above and Wo is selected from hydrogen or C1-5alkoxycarbonyl ; or (vii) a heterocyclic ring.

According to a further aspect of the second feature of the present invention there is provided a compound of Formula (Ia), or a pharmaceutically acceptable salt, pro-drug or solvate thereof, wherein R3 is selected from group (i), (ii) or (iv).

According to a further aspect of the second feature of the present invention there is provided a compound of Formula (Ia), or a pharmaceutically acceptable salt, pro-drug or solvate thereof, wherein R3 is selected from group (ii) and Rl3a is optionally substituted heterocyclyl.

According to a further aspect of the second feature of the present invention there is provided a compound of Formula (Ia), or a pharmaceutically acceptable salt, pro-drug or solvate thereof, wherein R3 is selected from group (ii), x is 0 and Rl3a is optionally substituted heterocyclyl.

According to a further aspect of the second feature of the present invention there is provided a compound of Formula (Ia), or a pharmaceutically acceptable salt, pro-drug or solvate thereof, wherein R3 is selected from group (ii), x is 0 and Rl3a is optionally substituted heterocyclyl attached to -(CH2)y- via a ring heteroatom and the optional substituents on heterocyclyl further include C1-4alkyl or di-C1-4alkyl attached to the ring carbon atoms adjacent to the heteroatom linked to -(CH2)y-. Preferably R13a is 2,4-di-Cz 4alkylazetin-l-yl,

2,5-di-CI 4alkylpyrrolin-l-yl or 2, 6-di-Co 4alkylpiperidin-l-yl. More preferably Rl3 is 2,5-dimethylpyrrolin-1-yl or 2,6-dimethylpiperidin-1-yl.

In this specification the term'alkyl'when used either alone or as a suffix includes straight chained, branched structures. Unless otherwise stated, these groups may contain up to 10, preferably up to 6 and more preferably up to 4 carbon atoms. Similarly the terms"alkenyl" and"alkynyl"refer to unsaturated straight or branched structures containing for example from 2 to 10, preferably from 2 to 6 carbon atoms. Cyclic moieties such as cycloalkyl, cycloalkenyl and cycloalkynyl are similar in nature but have at least 3 carbon atoms. Terms such as "alkoxy"comprise alkyl groups as is understood in the art.

The tenn"halo"or"halogeno"includes fluoro, chloro, bromo and iodo. References to aryl groups include aromatic carbocylic groups such as phenyl and naphthyl. The term "heterocyclyl"or"heterocyclic"includes aromatic or non-aromatic rings, for example containing from 4 to 20, suitably from 5 to 8 ring atoms, at least one of which, and preferably from 1-4 of which is a heteroatom such as oxygen, sulphur or nitrogen. Examples of such groups include furyl, thienyl, pyrrolyl, pyrrolidinyl, imidazolyl, triazolyl, thiazolyl, tetrazolyl, oxazolyl, isoxazolyl, pyrazolyl, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazinyl, quinolinyl, isoquinolinyl, quinoxalinyl, benzothiazolyl, benzoxazolyl, benzothienyl or benzofuryl. Examples of non-aromatic heterocyclyl groups include morpholino, piperidino, azetidine, tetrahydrofuryl, tetrahydropyridyl. In the case of bicyclic rings, these may comprise an aromatic and non-aromatic portion.

"Heteroaryl"refers to those groups described above which have an aromatic character.

The term"aralkyl"refers to aryl substituted alkyl groups such as benzyl.

Other expressions used in the specification include"hydrocarbyl"which refers to any structure comprising carbon and hydrogen atoms. The moiety may be saturated or unsaturated. For example, these may be alkyl, alkenyl, alkynyl, aryl, aralkyl, cycloalkyl, cycloalkenyl or cycloalkynyl, or combinations thereof.

Examples of such combinations are alkyl, alkenyl or alkynyl substituted with aryl, aralkyl, cycloalkyl, cycloalkenyl or cycloalkynyl, or an aryl, heterocyclyl, alkoxy, aralkyl, cycloalkyl, cycloalkenyl or cycloalkynyl substituted with alkyl, alkenyl, alkynyl or alkoxy, but others may be envisaged.

In particular hydrocarbyl groups include alkyl, alkenyl, alkynyl, aryl, aralkyl, cycloalkyl, cycloalkenyl or cycloalkynyl.

The term"interposed"used in relation to heteroatoms in hydrocarbyl chains means that the chains include a heteroatom such as sulphur, oxygen or nitrogen either at an intermediate position along their length or at an end of the chain.

The term"interposed"used in relation to heterocyclic rings in hydrocarbyl chains means that the chains include a heterocyclic ring either at an intermediate position along their length or at an end of the chain.

Suitable pharmaceutically acceptable salts of compounds of Formula (I) include acid addition salts such as methanesulfonate, fumarate, hydrochloride, hydrobromide, citrate, maleate and salts formed with phosphoric and sulphuric acid. A preferred pharmaceutically acceptable salt is a hydrochloride salt.

Thus, the alkyl portion of the alkyl, alkoxy, alkanoyloxy, alkoxymethyl, alkanoyloxymethyl, alkylsuphinyl, alkylsulphonyl, allcylsulfonamido, carboalkoxy, carboalkyl, alkanoylamino aminoalkyl, alkylaminoalkyl, N, N-dicycloalkylaminoalkyl, hydroxyalkyl, and alkoxyalkyl substituents include both straight chain as well as branched carbon chains. The cycloalkyl portions of N-cycloalkyl-N-alkylaminoalkyl and N, N-dicycloalkylaminoalkyl substituents include both simple carbocycles as well as carbocycles containing alkyl substituents. The alkenyl portion of the alkenyl, alkenoyloxymethyl, alkenyloxy, alkenylsulfonamido, substituents include both straight chain as well as branched carbon chains and one or more sites of unsaturation. The alkynyl portion of the alkynyl, alkynoyloxymethyl, allcynylsulfonamido, alkynyloxy, substituents include both straight chain as well as branched carbon chains and one or more sites of unsaturation. Carboxy is defined as a-CO2H radical.

Carboalkoxy of 2-7 carbon atoms is defined as a-C02R"radical, where R"is an alkyl radical of 1-6 carbon atoms. Carboalkyl is defined as a-COR"radical, where R"is an alkyl radical of 1-6 carbon atoms. Alkanoyloxy is defined as a-OCOR"radical, where R"is an alkyl radical of 1-6 carbon atoms. Alkanoyloxymethyl is defined as R"CO2CH2-radical, where R" is an alkyl radical of 1-6 carbon atoms. Alkoxymethyl is defined at R"OCH2-radical, where R"is an alkyl radical of 1-6 carbon atoms. Alkylsulphinyl is defined as R"SO-radical, where R"is an alkyl radical of 1-6 carbon atoms. Alkylsulphonyl is defined as R"S02-radical, where R"is alkyl radical of 1-6 carbon atoms. Alkylsulfonamido, alkenylsulfonamido, alkynylsulfonamido are defined as R"SO2NH-radical, where R"is an alkyl radical of 1-6 carbon atoms, an alkenyl radical of 2-6 carbon atoms, or an alkynyl radical of 2-6 carbon atoms, respectively. N-alkylcarbamoyl is defined as R"NHCO-radical, where R"is an alkyl radical of 1-6 carbon atoms. N, N-dialkylcarbamoyl is defined as R"R'NCO-radical, where

R"is an alkyl radical of 1-6 carbon atoms, R'is an alkyl radical of 1-6 carbon atoms and R', and R"may be the same or different. When X is substituted, it is preferred that it is mono-, di-, or tri-substituted, with monosubstituted being most preferred. It is preferred that of the substituents, Rl, R2 and R4 at least one is hydrogen and it is most preferred that two or three be hydrogen. An azacycloalkyl-N-alkyl substituent refers to a monocyclic heterocycle that contains a nitrogen atom on which is substituted a straight or branched chain alkyl radical. A morpholino-N-alkyl substituent is a morpholine ring substituted on the nitrogen atom with a straight or branch chain alkyl radical. A pipeazino-N-alkyl substituent is a piperazine ring substituted on one of the nitrogen atoms with a straight or branch chain alkyl radical. A N-alkyl-piperidino-N-allçyl substituent is a piperidine ring substituted on one of the nitrogen atoms with a straight or branched chain alkyl group and on the other nitrogen atom with a straight or branch chain alkyl radical.

When any group contains an alkyl portion, the alkyl portion contains preferably 1-6 carbon atoms, more preferably 1-4 carbon atoms, particularly methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl or tert-butyl. When any group contains an alkenyl or alkynyl portion, the alkenyl or alkynyl portion contains preferably 2-6 carbon atoms, more preferably 2-4 carbon atoms.

The compounds of this invention may contain an asymmetric carbon; in such cases, the compounds of this invention cover the racemate and the individual R and S entantiomers, and in the case were more than one asymmetric carbon exists, the individual diasteromers, their racemates and individual entantiomers.

When R3 is a group of formula-Xl-Rx-(OH) p, suitable examples of Xl are-O-, -NR14C (O)-,-C (o) NR15- or-NRlB-where R14, Rls and Rl8 are as defined above. Preferably Xl is selected from-O-,-NR14C (O)-, or-NRlB-. In particular Xl is-O-. Particular examples of Rx are C2-6alkylene chains which is interposed by at least one heteroatom such as nitrogen, or a heterocyclic ring, in particular, a saturated 5-6 membered heterocyclic ring. The ring suitably contains at least from one to three and preferably one heteroatom which is suitably nitrogen. In particular, the heterocyclic ring is a saturated heterocyclic ring. The hydroxy groups may be attached to the alkylene portion of the group Razor where present, the heterocyclic ring. In particular, these groups are groups of sub-formula (i) - X'- (CH2) j- (T)- ( (CH2) k-OH) p (i)

where Xl and p is as defined above, T is a 5 or 6 membered nitrogen containing ring, j is 2,3, 4, or 5, and k is 0,1,2 or 3. Preferably Rx is interposed by a nitrogen atom or is not interposed by a heteroatom or a heterocyclic ring.

Where the alkylene chain Rx is interposed with a nitrogen atom, it is suitably in the form or a group-NR75-where R75 is hydrogen or C1-4alkyl (optionally substituted by hydroxy), in particular C1-3alkyl or hydroxyC1-3alkyl.

Where R3 is a group of formula R13a-X20-(CH2)x, x is suitably 0. Preferred groups for Rl3a are heterocyclic rings, preferably saturated heterocyclic rings.

Where R3 is a group of formula Rl3a-(CH2) y-Xl-(CH2) x, x is suitably 0 and y is an integer between 1 and 4.

Preferred groups for Rl3a are heterocyclic rings, preferably saturated heterocyclic rings.

Preferred saturated heterocyclic rings are selected from azetidinyl, pyrrolidinyl, tetrahydrofuranyl, 1,3-dioxolanyl, 1,3-oxazolidineyl 1,2,4-oxadiazolidinyl, tetrahydrothiophenyl, 1-oxo-tetrahydrothiophenyl, 1, 1-dioxo-tetrahydrothiophenyl, morpholinyl, piperidinyl, piperazinyl, 1,3-dioxanyl, tetrahydropyranyl, tetrahydrothiopyranyl, 1-oxo-tetrahydrothiopyranyl, 1, 1-dioxo-tetrahydrothiopyranyl, thiomorpholinyl, 1- oxothiomorpholinyl and 1,1-dioxo-thiomorpholinyl. More preferred saturated heterocyclic rings are selected from azetidinyl, pyrrolidinyl, tetrahydrofuranyl, 1,3-dioxolanyl, 1,3- oxazolidinyl, 1,2,4-oxadiazolidinyl, 1, 1-dioxoctetrahydrothiophenyl, morpholinyl, piperidinyl, piperazinyl, 1,3-dioxanyl, tetrahydropyranyl, 1, 1-dioxo-tetrahydrothiopyranyl, thiomorpholinyl, 1-oxo-thiomorpholinyl and 1, 1-dioxo thiomorpholinyl. Preferred aromatic heterocyclyl rings are selected from: furanyl, thiophenyl, pyrrolyl, pyrrolinyl, imidazolyl, imidazolinyl, pyrazolyl, pyrazolinyl, triazolyl, thiazolyl, isothiazolyl, oxadiazolyl, oxazolyl, isoxazolyl, thiadiazolyl, 2,5-dihydro-1,2,4-oxadiazolyl, pyrazinyl, pyridazinyl, pyrimidinyl and triazinyl. More preferred aromatic heterocyclyl rings are selected from: 2,3- dihydropyrrolyl, imidazolyl, pyrazolyl, 1,3-thiazolyl, 1,3,4-oxadiazolyl, isoxazolyl, 1,2,4- thiadiazolyl, furanyl, 2,5-dihydro-1,2,4-oxadiazolyl and pyrimidinyl. Suitably R14, R15 and R18 are hydrogen or 1-3 alkyl and preferably hydrogen.

Preferred substituents on Rl3a are selected from nitro, C2-5alkenyl, Ci-3alkoxyCi-3alkyl, Cl-salkanoyl and Ci-salkylsulphonyl. More preferred substituents on R are selected from Ci-3alkoxyCi-3alkyl, Ci-salkanoyl and C1-5alkylsulphonyl. Most preferred substituents on Rl3a are selected from acetyl, methoxyethyl, methylsulphonyl and ethylsulphonyl. Preferred substituted heterocyclic rings at Rl3a are selected from: 2,6-dimethylmorpholino, 4-

metlioxyethylpiperazin-1-yl, 4-acetylpiperazin-1-yl, 3-methylsulphonylpyrrolidin-1-yl and 4- ethylsulphonylpiperazin-1-yl.

Particular examples of groups of formula -X1-Ry-NRz-Ry'-S-Ry" for R3 are groups where X1 is -O-, -NR14C(O)-, -C(O)NR15- or -NR18- where R14, R15 and Rl8 are as defined above. Preferably Xl is selected from -O-, -NR14C(O)-, or -NR18-. In particular is-0-.

Suitably RI is a C2-6alkenylene group. Suitably RY is a C2-3alkylene group. Suitably Rz is hydrogen or C1-3alkyl and in particular is hydrogen.

In one embodiment however, the group -NRz-Ry'-S-Ry'' is a thiomorpholine ring.

Where R3 is a group of formula -X1-Rx'-(C4-6cycloalkyl) or a group of the formula-Xl- Rx-(C3 (C3-6cycloalkyl), X1 is suitably -O-, -NR14C(O)-, -C(O)NR15- or -NR18- where R14, R15 and R'8 are as defined above. Preferably Xl is selected from-O-,-NR14C (O)-, or-NR18-. In particular Xl is-O-. Particular examples of Rx' are C2-5alkylene groups which suitably include at least one heteroatom in particular a group -NR75- where R75 is as defined above.

Suitably the C4-6cycloalkyl group is cyclopropyl. hi particular R, Ra and R4 are selected from hydrogen, hydroxy, halogeno, cyano, nitro, trifluoromethyl, C1-3alkyl, -NR11R12- (wherein Rll and R12 are as defined above), or R13X1-(CH2)x- (wherein x is 0 or an integer of from 1-3, Xl represents a direct bond,-0-, -CH2-, -OC(O)-, -C(O)-, -S-, -SO-, -SO2-, -NR14C(O)-, -NR14C(O)O-, -C(O)NR15-, -C(O)ONR15-, -SO2NR16-, -NR17SO2- or -NR18- (wherein R14, R15, R16, R and R18 each independently represents hydrogen, C1-3alkyl or C1-3alkyoxyC2-3alkyl)), and R13 is any one of the following twenty-two groups 1') C1-5alkyl which may be unsubstituted or which may be substituted with one or more groups selected from hydroxy, oxiranyl, fluoro, chloro, bromo and amino (including C1-3alkyl and trifluoromethyl); 2')-RaX2C (O) Rl9 (wherein X2 represents -O- or -NR20- (in which Wo represents hydrogen, C1-3alkyl or Cl-3alkoxyC2-3alkyl) and Rl9 represents C1-3alkyl, -NR21R22 or -OR23 (wherein Ray Raz and R23 which may be the same or different each represents hydrogen, C1-5alkyl. hydroxyC1-5alkyl or C1-3alkoxyC2-3alkyl)) ; 3') -RbX3R24 (wherein X3 represents-0-, C (O)-S-,-SO-,-SO2-,-OC (O)-, -NR25C (O)-, -NR25C(O)O-, -C(O)NR26-, -C(O)ONR26-, -SO2NR27-, -NR28SO2- or -NR29- (wherein R25, R26, R27, Wl and R29 each independently represents hydrogen, C1-3alkyl, hydroxy C1-4alkyl or Ci-3alkoxyC2-3alkyl and R24 represents hydrogen, C1-6alkyl, C2-6alkenyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, phenyl or a 5-6-membered saturated heterocyclic group

with 1-2 heteroatoms, selected independently from O, S and N, which C1-6alkyl group may bear 1, 2 or 3 substituents selected from oxo, hydroxy, halogeno, cyclopropyl, amino, C1-4alkylamino,C1-4alkanoyldi-C1-4alkylamino, C1-4alkylthio, C1-4alkoxy and which cyclic group may bear 1 or 2 substituents selected from oxo, hydroxy, halogeno, cyano, C1-4cyanoalkyl, C1-4alkyl, C1-4hydroxyalkyl, C1-4alkoxy, C1-4alkoxyC1-4alkyl, C1-4alkylsulphonylC1-4alkyl, C1-4alkoxycarbonyl, C1-4aminoalkyl, C1-4alkylamino, di (C1-4alkyl) amino, C1-4alkylaminoC1-4alkyl, di (C1-4alkyl)aminoC1-4alkyl, Ci-4alkylaminoCi-4alkoxy, di (Cl-4alkyl) aminoCl-4alkoxy and a group -(-O-)f(Rb') gD (wherein f is 0 or 1, g is 0 or 1 and D is a C3-6cycloalkyl group or a 5-6-membered saturated heterocyclic group with 1-2 heteroatoms, selected independently from O, S and N, which cyclic group may bear one or more substituents selected from Cl-4alkyl)) ; 4') -RcX4Rc' X5R30 (wherein X4 and X5 which may be the same or different are each-O-, C (O), -S-, -SO-, -SO2-, -NR31C(O)-, -NR31C(O)O-, -C(O)NR32-, -C(O)ONR32-, -SO2NR33-, -NR34SO2- or -NR35- (wherein R31,R32,R33,R34 and R35 each independently represents hydrogen, C1-3alkyl or C1-3alkoxyC2-3alkyl and R30 represents hydrogen, Cl-3alkyl, hydroxyCl-3alkylorCl-3alkoxyC2-3alkyl) ; 5') RdR36 (wherein R36 is a 4-6-membered cycloalkyl or saturated heterocyclic ring (linked via carbon or nitrogen) with 1-2 heteroatoms, selected independently from O, S and N, which cycloalkyl or heterocyclic group may bear 1 or 2 substituents selected from oxo, hydroxy, halogeno, cyano, C1-4alkyl, hydroxyC1-4alkyl, cyanoC1-4alkyl, cyclopropyl, Cl-4alkylsulphonylCl-4alkyl, Cl-4alkoxycarbonyl, carboxamido, C1-4aminoalkyl, C1-4alkylamino, di (C1-4alkyl) amino, Ci-4alkylaminoCi-4alkyl, Ci-4alkanoyl, di (C1-4alkyl)aminoC1-4alkyl, C1-4alkylaminoC1-4alkoxy, di (C1-4alkyl)aminoC1-4alkoxy nitro, amino, C1-4alkoxy, C1-4hydroxyalkoxy, carboxy, trifluoromethyl,-C (O) NR65R66, -NR67C(O)R68 (wherein R65,R66,R67 and R68, which may be the same or different, each represents hydrogen, C1-4alkyl, hydroxyC1-4alkyl or C1-3alkoxyC2-3alkyl) and a group -(-O-) f (Rb) gringD (wherein f is 0 or 1, g is 0 or 1 and ring D is a cyclic group selected from C3-6cycloalkyl, aryl or 5-6-membered saturated or unsaturated heterocyclic group with 1-2 heteroatoms, selected independently from O, S and N, which cyclic group may bear one or more substituents selected from halo and C1-4alkyl) ; 6') ReX6R37 (wherein X6 represents a direct bond,-O-,-C (O)-,-S-,-SO-,-SO2-,-NR38CO-, -NR38C(O)O-, -CONR39-, -C(O)ONR39-, -SO2NR40-, -NR41SO2- or -NR42- (wherein R38,R39, wo, R41 and R42 each independently represents hydrogen, C1-3alkyl or Ci-3alkoxyC2-3alkyl)

and R37 is a pyridone group, a phenyl group or a 5-6-membered aromatic heterocyclic group (linked via carbon or nitrogen) with 1-3 heteroatoms selected from 0, N and S, which pyridone, phenyl or aromatic heterocyclic group may carry up to 5 substituents selected from hydroxy, nitro, halogeno, amino, C1-4alkyl, C1-4alkoxy, C1-4hydroxyalkyl, C1-4aminoalkyl, C1-4alkylamino, C1-4hydroxyalkoxy, oxo, cyanoC1-4alkyl, cyclopropyl, C1-4alkylsulphonylC1-4alkyl, C1-4alkoxycarbonyl, di (C1-4alkyl) amino, C1-4alkylaminoC1-4alkyl, C1-4alkanoyl, di (C1-4alkyl)aminoC1-4alkyl, C1-4alkylaminoC1-4alkoxy, di(C1-4alkyl)aminoC1-4alkoxy, carboxy, carboxamido, trifluoromethyl, cyano, -C(O)NR43R44, -NR45C(O)R46 (wherein R43,R44,R45 and R46, which may be the same or different, each represents hydrogen, C1-4alkyl, hydroxyC1-4alkyl or C1-3alkoxyC2-3alkyl) and a group -(-O-) Rb) gringD (wherein f is 0 or 1, g is 0 or 1 and ring D is a cyclic group selected from C3-6cycloalkyl, aryl or 5-6-membered saturated or unsaturated heterocyclic group with 1-2 heteroatoms, selected independently from 0, S and N, which cyclic group may bear one or more substituents selected from halo and C1-4alkyl) ; 7') - RfR36 (wherein R36 is as defined in (5') hereinbefore); 8') -RgR36 (wherein R36 is as defined in (5') hereinbefore); 9') X7R47 (wherein X7 is-S02-,-O-or-CONR48R49- (wherein R48 and R49, which may be the same or different, each represents hydrogen, C1-3alkyl or Cl-3alkoxyC2-3alkyl) and R47 represents C1-5alkyl which may be unsubstituted or which may be substituted with one or more groups selected from hydroxy, fluoro and amino) with the provisos that when X7 is -S02-, Xl is-O-, when X7 is-O-, Xl is carbonyl, when X7 is -CONR48R49-, Xl is-O-or NR13 (wherein R48, R49 and R18 are as defined in (6') hereinbefore); 10')-RhR37 (wherein R37 is as defined in (6') hereinbefore); 11') -RiR37 (wherein R37 is as defined in (6') hereinbefore); 12')-Ri X8R37 (wherein X8 represents -O-, -C (O)-,-S-,-SO-,-S02-,-OC (O)-, -NR50C(O)-, -NR50C(O)O-, -C(O)NR51-, -C(O)ONR51-, -SO2NR52-, -NR53SO2- or -NR54- (wherein R50, R51, R52, R53 and R54 each independently represents hydrogen, C1-3alkyl, hydroxyC1-3alkyl or Ci-3alkoxyC2-3alkyl) and R37 is as defined in (6') hereinbefore); 13')-RkX9R37 (wherein X9 represents -O-, C (O), -S-, -SO-, -SO2-, -NR55C(O)-, -NR55C(O)O-, -C(O)NR56-, -C(O)ONR56-, -SO2NR57-, -NR58SO2- or -NR59- (wherein R55, R56, W R58 and R49 each independently represents hydrogen, C1-3alkyl, hydroxyC1-3alkyl or Cl-3alkoxyC2-3alkyl) and R37 is as defined in (6') hereinbefore); 14') -RmX10Rm'R37 (wherein Xlo represents -O-, -C (O)-, -S-, -SO-, -SO2-, -NR60C(O)-,

-NR60C(O)O-, -C(O)NR61-, -C(O)ONR61-, -SO2NR62-, -NR63SO2- or -NR64- (wherein R60, R61, R62, R63 and R64 each independently represents hydrogen, C1-3alkyl, hydroxyC1-3alkyl or Ci-salkoxyCz-salkyI) and R37 is as defined in (6') hereinbefore); 15') R36 (where R36 is as defined in (5') hereinbefore) ; 16') -Rn X10Rn'R36 (wherein Xlo is as defined in (14') above and R36 is as defined in (5') hereinbefore) ; 17') -Rp X10-Rp'R37 (wherein X10 is as defined in (14') above and R37 are as defined in (6') hereinbefore); 18') C2-5alkenyl which may be unsubstituted or which may be substituted with one or more groups selected from hydroxy, fluoro, amino, C1-4alkylamino, N,N-di(C1-4alkyl) amino, aminosulphonyl, N-Cl-4allcylaminosulphonyl and N, N-di (C1-4alkyl)aminosulphonyl ; 19') C2-5alkynyl which may be unsubstituted or which may be substituted with one or more groups selected from hydroxy, fluoro, amino, C1-4alkylamino, N,N-di(C1-4alkyl) amino, aminosulphonyl, N-C1-4alkylaminosulphonyl and N, N-di (C1-4alkyl) aminosulphonyl; 20') -RtX10Rt'R36 (wherein Xl° is as defined in (14') above and R36 is as defined in (5') hereinbefore); 21') -Ru X10 Ru'R36 (wherein X10 is as defined in (14') above and R36 is as defined in (5') hereinbefore); and 22') -RvR69(Rv')q(X10),R70(wherein X10 is as defined in (14') above, q is 0 or 1, r is 0 or 1, and R 69 is a Ci-3alkylene group or a cyclic group selected from cyclopropyl, cyclobutyl, cyclopentylene, cyclohexylene or a 5-6-membered saturated heterocyclic group with 1-2 heteroatoms, selected independently from O, S and N, which Ci-salkylene group may bear 1 or 2 substituents selected from oxo, hydroxy, halogeno and C1-4alkoxy and which cyclic group may bear 1 or 2 substituents selected from oxo, hydroxy, halogeno, cyano, C1-4cyanoalkyl, C1-4alkyl, C1-4hydroxyalkyl, C1-4alkoxy, C1-4alkoxyC1-4alkyl, C1-4alkylsulphonylC1-4alkyl, Ci-4alkoxycarbonyl, Ci-4aminoalkyl, Ci-4alkylamino, di (C1-4alkyl) amino, C1-4alkylaminoC1-4alkyl, i (C1-4alkyl)aminoC1-4alkyl, C1-4alkylaminoCl-4alkoxy, di (Cl-4alkyl) aminoCl-4alkoxy and a group -(-O-) f (Rb) gringD (wherein f is 0 or 1, g is 0 or 1 and ring D is a cyclic group selected from C3-6cycloalkyl, aryl or 5-6-membered saturated or unsaturated heterocyclic group with 1-2 heteroatoms, selected independently from O, S and N, which cyclic group may bear one or

more substituents selected from halo and C1-4alkyl) ; and R70 is hydrogen, C1-3alkyl, or a cyclic group selected from cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and a 5-6-membered saturated heterocyclic group with 1-2 heteroatoms, selected independently from O, S and N, which C1-3alkyl group may bear 1 or 2 substituents selected from oxo, hydroxy, halogeno, C1-4alkoxy and which cyclic group may bear 1 or 2 substituents selected from oxo, hydroxy, halogeno, cyano, C1-4cyanoalkyl, C1-4alkyl, C1-4hydroxyalkyl, C1-4alkoxy, C1-4alkoxyC1-4alkyl, C1-4alkylsulphonylC1-4alkyl, C1-4alkoxycarbonyl, C1-4aminoalkyl, C1-4alkylamino, di (C1-4alkyl) amino, C1-4alkylaminoC1-4alkyl, di (C1-4alkyl)aminoC1-4alkyl, C1-4alkylaminoC1-4alkoxy, di (C1-4alkyl)aminoC1-4alkoxy and a group -(-O-)f(Rb') gringD (wherein f is 0 or 1, g is 0 or 1 and D is a cyclic group selected from C3-6cycloalkyl, aryl or 5-6-membered saturated or unsaturated heterocyclic group with 1-2 heteroatoms, selected independently from O, S and N, which cyclic group may bear one or more substituents selected from halo and C1-4alkyl) ; and wherein R, Rb, Rb, Rc, Rc', Rd, Re, Ri len, Rm' Rn, Rn', Rp', Rt', Ru', Rv and Rv' are independently selected from C1-8alkylene groups optionally substituted by one or more substituents selected from hydroxy, halogeno, amino; and Re may additionally be a bond ; and Rf, Rh, Rj, Rp and Rt are independently selected from C2-8alkenylene groups optionally substituted by one or more substituents selected from hydroxy, halogeno, amino, and Rt may additionally be a bond; and Rg, Rk and Ru are independently selected from by C2-8alkynylene groups optionally susbstituted by one or more substituents selected from hydroxy, halogeno, amino, subject to the proviso set out above.

In one embodiment, at least one group Rl, R2 or R4 is a group R13-X1-(CH2)x- (wherein x is 0 or an integer of from 1-3, Xl represents-NR14C (0) 0-, or C (O) ONRIS-, (wherein R13R14 and R15 are as defined above). A particular example of such a group for R, R2, R3 or R4 is a group-NHC (O) OR" where R13 is as defined above, and in particular is a group of formula (6') such as benzyl.

In particular, at least one group Ru, R2 or R4 is selected from R13X1-(CH2)x- (wherein x is 0 or an integer of from 1-3, Xl represents a direct bond,-O-,-CH2-,-OC (O)-,-C (O)-,-S-, -SO-, -SO2-, -NR14C(O)-, -NR14C(O)O-, -C(O)NR15-, -C(O)ONR15-, -SO2NR16-, -NR17SO2- or-NRl3- (wherein R, R15, R, R and R each independently represents hydrogen, C1-3alkyl or Ci-3aIkoxyC2-3alkyl)), and R13 is any one of the following twenty-two groups; 1") C1-5alkyl substituted with one or more groups selected from oxiranyl, chloro or bromo;

2")-Ra'X2C (O) Rl9 (wherein X2 and Rl9 are as defined in (2') above, and Ra'is a C1-8alkylene groups substituted by one or more substituents selected from hydroxy, halogeno, amino, 3") -RbX3R24 (wherein either R24 is any of the groups defined in (3') above and X3 is - C (O),-NR25C (0) 0-,-C (O) ONR26- (wherein R25 and R26 are as defined in (3') above), or X3 is any other groups defined in (3') above and R24 represents C1-3alkyl, C3-6alkyl, C2-6alkenyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl or phenyl, wherein (a) the C3-6alkyl group may bear 1, 2 or 3 substituents selected from oxo, hydroxy, halogeno, cyclopropyl, amino, C1-4alkylamino,C1-4alkanoyldi-C1-4alkylamino, C1-4alkylthio, C1-4alkoxy ; (b) the C1-3 alkyl group may be similarly substituted to the C3-6alkyl provided it includes at least one substitutent selected from cyclopropyl, amino, C1-4alkylamino, C1-4alkanoyldi-C1-4alkylamino , C1-4alkylthio, C1-4alkoxy ; (c) the cyclopropyl or cyclobutyl may bear 1 or 2 substituents selected from oxo, hydroxy, halogeno, cyano, C1-4cyanoalkyl, C1-4alkyl, C1-4hydroxyalkyl, C1-4alkoxy, C1-4alkoxyC1-4alkyl, C1-4alkylsulphonylC1-4alkyl, C1-4alkoxycarbonyl, Ci-4aminoalkyl, Ci-4alkylamino, di (Cl-4alkyl) amino, C1-4alkylaminoC1-4alkyl, di (C1-4alkyl) aminoCl-4alkyl, Cl-4alkylaminoCl-4alkoxy, di (C1-4alkyl) aminoCl-4alkoxy and a group-(-O-) f (Rb) gD (wherein f, g Rb, and D are as defined above); (d) the cyclopentyl or cyclohexyl may be similarly substituted provided it includes at least one substituent selected from cyano, C1-4cyanoalkyl, C1-4alkoxyC1-4alkyl, C1-4alkylsulphonylC1-4alkyl, C1-4alkoxycarbonyl, C1-4aminoalkyl, C1-4alkylamino, di (C1-4alkyl) amino, C1-4alkylaminoC1-4alkyl, di (C1-4alkyl)aminoC1-4alkyl, C1-4alkylaminoC1-4alkoxy, di (C1-4alkyl)aminoC1-4alkoxy and a group- (-O-) f (Rb') gD (wherein f, g Rb' and D are as defined above), or X3 and R24 are any of the groups defined in 3'above and Rb is other than C1-5alkylene; 4") -RcX4Rc'X5R30 (wherein RC, Rc' and R30 are as defined in (4') above and X4 and X5 are as defined above provided at least one of these is selected from C (O),-NR3lC (0) 0-, or -C (O) ONR32- (wherein R31 and R32 are as defined in (4') above), or X4 and X5 are any of the groups defined in (4') above, and either Wo is hydroxyCl-3allcyl or C1-3alkoxyC2-3alkyl), or at least one ofR° or R° is other than an unsubstituted C1-5alkylene group; 5") RdR36. (wherein Ra is as defined above, and R36 is a 4-6-membered cycloalkyl or a saturated heterocyclic ring (linked via carbon or nitrogen including for example from 4 to 7 atoms) with 1-2 heteroatoms, selected independently from O, S and N, which cycloalkyl or heterocyclic group may bear 1 or 2 substituents listed in (5') above, provided that where R36 is

a 5 or 6 membered heterocyclic ring, either it carries at least one substitutent selected from cyano, cyanoC1-4alkyl, cyclopropyl, C1-4alkylsulphonylC1-4alkyl, C1-4alkoxycarbonyl, carboxamido, Cl-4aminoalkyl, CI-4alkylamino, di (C1-4alkyl) amino, C1-4alkylaminoCl-4alkyl, CI-4alkanoyl, di (Cl-4alkyl) aminoCl-4alkyl, Cl-4alkylaminoCl-4allcoxy, di (CI-4alkyl) aminoCl-4alkoxy nitro, amino, Ci-4hydroxyalkoxy, carboxy, trifluoromethyl, -C(O)NR65R66, -NR67C(O)R68 (wherein R65, R66, R67 and R68, are as defined in (5') above and a group-(-O-) f (Rb) gringD (wherein f, g, Rb and D are as defined above); R36 is any of the groups defined in 5'above and Rd is other than C1-5alkyl ; 6") ReX6R37 (wherein Re and R37 are any of the groups defined above, provided that X6 represents-C (O)-,-NR38C (0) O-, or-C (O) ONR39-, (wherein R38 and R39 are as defined above) or, x6 is any other group listed in (6') above, provided that either R37 is a pyridone group, a phenyl group or a 5-6-membered aromatic heterocyclic group as (linked via carbon or nitrogen) with 1-3 heteroatoms selected from O, N and S, which pyridone, phenyl or aromatic heterocyclic group which is substituted as described in (6') provided it carries at least one substituent selected from nitro, amino, C1-4hydroxyalkoxy, oxo, cyanoCl-4allcyl, cyclopropyl, Cl-4alkylsulphonylCl-4alkyl, Cl-4alkoxycarbonyl, di (C1-4alkyl0amino, C1-4alkylaminoC1-4alkyl, C1-4alkanoyl, di (C1-4alkyl)aminoC1-4alkyl, C1-4alkylaminoC1-4alkoxy, di (C1-4alkyl)aminoC1-4alkoxy, carboxamido, trifluoromethyl, or a group -(-O-)f(Rb') gringD (wherein f, g, Rb' and D are as defined above) or Re is other than a direct bond or a C1- 5alkylene group; 7") - RfR36 (wherein Rf and R36 are as defined in (7') above provided that where Rf is unsubstituted C2-6 alkenylene, R36 is as defined in (5") hereinbefore) ; 8") -Rg R36 (wherein R g and R36 are as defined in (8') above provided that where Rg is unsubstituted C2-6 alkynylene, R36 is as defined in (5") hereinbefore) ; 10")-RIlR37 (wherein Rh and R37 are as defined in (10') above provided that where Rh is unsubstituted C2-6 alkenylene, R37 is W7 is a pyridone group, a phenyl group or a 5-6-membered aromatic heterocyclic group as (linked via carbon or nitrogen) with 1-3 heteroatoms selected from O, N and S, which pyridone, phenyl or aromatic heterocyclic group which is substituted as described in (6') provided it carries at least one substituent selected from nitro, amino, C1-4hydroxyalkoxy, oxo, cyanoC1-4alkyl, cyclopropyl, C1-4alkylsulphonylC1-4alkyl, C1-4alkoxycarbonyl, di (C1-4alkyl) amino, C1-4alkylaminoC1-4alkyl, C1-4alkanoyl, di (Cl-4alkyl) aminoCl_4alkyl, Cl-4alkylaminoCl-4alkoxy,

di (Cl-4alkyl) aminoCl-4alkoxy, carboxamido, trifluoromethyl, or a group-(-O-) f (Rb) gringD (wherein f, g, Rb and D are as defined above)); 11")-R'R37 (wherein Ri and R37 are as defined in (11') above provided that where Ri is unsubstituted 2-6 alkynylene, R37 R37 is a pyridone group, a phenyl group or a 5-6-membered aromatic heterocyclic group as (linked via carbon or nitrogen) with 1-3 heteroatoms selected from O, N and S, which pyridone, phenyl or aromatic heterocyclic group which is substituted as described in (6') provided it carries at least one substituent selected from nitro, amino, Cl-4hydroxyalkoxy, oxo, cyanoCl-4allcyl, cyclopropyl, C1-4alkylsulphonylC1-4alkyl, C1-4alkoxycarbonyl, di (C1-4alkyl) amino, Cl-4allcylaminoCl-4alkyl, Ci-4alkanoyl, di (C1-4alkyl)aminoC1-4alkyl, C1-4alkylaminoC1-4alkoxy, di (CI-4allcyl) aminoCl-4alkoxy, carboxamido, trifluoromethyl, or a group-(-O-) f (Rb) gringD (wherein f, g, Rb and D are as defined above); 12") -Rj X8R37 (wherein Rj and R37 are as defined in (12') above, and X8 is-C (O)-, -NR50C(O)O- or -C(O)ONR51-. (wherein R50 and R51 are as defined in (12') above, or X8 is any other group listed in (12'above) and either R37 is a pyridone group, a phenyl group or a 5-6-membered aromatic heterocyclic group as (linked via carbon or nitrogen) with 1-3 heteroatoms selected from O, N and S, which pyridone, phenyl or aromatic heterocyclic group which is substituted as described in (6') provided it carries at least one substituent selected from nitro, amino, C1-4hydroxyalkoxy, oxo, cyanoCi-4alkyl, cyclopropyl, Cl-4alkylsulphonylCl-4alkyl, Cl-4alkoxycarbonyl, di (Cl-4alkyl) amino, C1-4alkylaminoC1-4alkyl, Ci-4alkanoyl, di (C1-4alkyl) aminoCl-4alkyl, Cl-4alkylaminoCl-4alkoxy, di (CI-4alkyl) aminoCl-4alkoxy, carboxamido, trifluoromethyl, or a group -(-O-)f(Rb')gringD (wherein f, g, Rb and D are as defined above); or Ri is other than unsubstituted Cz- 6alkenylene ; 13")-RkX9R37 (wherein Rk and R37 are as defined in (13') above, and X9 is-C (O)-, -NR55C(O)O- or -C(O)ONR56-. (wherein R55 and R56 are as defined in (13') above, or X9 is any other group listed in (13'above) and either R37 is a pyridone group, a phenyl group or a 5-6-membered aromatic heterocyclic group as (linked via carbon or nitrogen) with 1-3 heteroatoms selected from O, N and S, which pyridone, phenyl or aromatic heterocyclic group which is substituted as described in (6') provided it carries at least one substituent selected from nitro, amino, C1-4hydroxyalkoxy, oxo, cyanoC1-4alkyl, cyclopropyl, C1-4alkylsulphonylC1-4alkyl, C1-4alkoxycarbonyl, di (C1-4alkyl)amino, C1-4alkylaminoC1-4alkyl, CI-4alkanoyl, di (C1-4alkyl)aminoC1-4alkyl, C1-4alkylaminoC1-4alkoxy,

di (C1-4alkyl)aminoC1-4alkoxy, carboxamido, trifluoromethyl, or a group -(-O-)f(Rb') gringD (wherein f, g, Rb and D are as defined above); or Rk is other than unsubstituted C2-6alkynylene 14") -RmX10Rm'R37 (wherein Rm, Rm' and R37 are as defined in (14') above, and Xl° represents -C(O)-, --NR60C(O)O- or -C(O0ONR61-, (wherein R60 and R6l are as defined in (14') above, or where Xlo is any other group listed in (14') above, and either R37 is a pyridone group, a phenyl group or a 5-6-membered aromatic heterocyclic group as (linked via carbon or nitrogen) with 1-3 heteroatoms selected from O, N and S, which pyridone, phenyl or aromatic heterocyclic group which is substituted as described in (6') provided it carries at least one substituent selected from nitro, amino, C1-4hydroxyalkoxy, oxo, cyanoC1-4alkyl, cyclopropyl, C1-4alkylsulphonylC1-4alkyl, C1-4alkoxycarbonyl, di (C1-4alkyl) amino, C1-4alkylaminoC1-4alkyl, C1-4alkanoyl, di (C1-4alkyl)aminoC1-4alkyl, C1-4alkylaminoC1-4alkoxy, di (CI-4alkyl) aminoCl-4alkoxy, carboxamido, trifluoromethyl, or a group-(-O-) f (Rb) gringD (wherein f, g, Rb and D are as defined above), or at least one of Rm or Rm' is other than unsubstituted C1-3alkylene) ; 15") R36 (where R36 is as defined in (5") hereinbefore); 16") -Rn X10Rn'R36 (wherein R", Rn' and R36 are as defined in (16') above and Xl° represents -C (O)-,--NR60C (O) O-or-C (O) ONR6l-, (wherein R60 and R6l are as defined in (14') above or Xl° is any of the other groups set out in (14') above and either R36 is as defined in (5") hereinbefore) or at least one of Rn or Rn is other than unsubstituted C1-3alkyl ; 17")-RP X10-Rp'R37 (wherein Xl° is as defined in (14') above and RP, Rp' and R37 are as defined in (6') hereinbefore); 18") C2-5alkcnyl which may be unsubstituted or which may be substituted with one or more groups selected from hydroxy, fluoro, amino, C1-4alkylamino, N,N-di(C1-4alkyl) amino, aminosulphonyl, N-C1-4alkylaminosulphonyl and N, N-di (C1-4alkyl) aminosulphonyl; 19") C2-5alkynyl which may be unsubstituted or which may be substituted with one or more groups selected from hydroxy, fluoro, amino, C1-4alkylamino, N,N-di(C1-4alkyl) amino, aminosulphonyl, N-C1-4alkylaminosulphonyl and N, N-di (C1-4alkyl)aminosulphonyl ; 20") -RtX10Rt'R36 (wherein Xl° is as defined in (14') above and R36 is as defined in (5') hereinbefore); 21") -Ru X10 Ru'R36 (wherein Xl° is as defined in (14') above and W is as defined in (5') hereinbefore); and

22") - Rv R69(Rv') q (X10) rR70 (wherein Xl° is as defined in (14') above, q is 0 or 1, r is 0 or 1, and R69 is a Cl-3alkylene group or a cyclic group selected from cyclopropyl, cyclobutyl, cyclopentylene, cyclohexylene or a 5-6-membered saturated heterocyclic group with 1-2 heteroatoms, selected independently from O, S and N, which C1-3alkylene group may bear 1 or 2 substituents selected from oxo, hydroxy, halogeno and C1-4alkoxy and which cyclic group may bear 1 or 2 substituents selected from oxo, hydroxy, halogeno, cyano, Ci-4cyanoalkyl, C1-4alkyl, C1-4hydroxyalkyl, C1-4alkoxy, C1-4alkoxyC1-4alkyl, C1-4alkylsulphonylC1-4alkyl, C1-4alkoxycarbonyl, C1-4aminoalkyl, C1-4alkylamino, di (Cl-4alkyl) amino, Cl-4alkylaminoCl-4alkyl, i (Cl-4alkyl) aminoCl-4alkyl, Cl-4alkylaminoCl-4alkoxy, di (C1-4alkyl)aminoC1-4alkoxy and a group -(-O-)f(Rb')gringD (wherein f is 0 or 1, g is 0 or 1 and ring D is a cyclic group selected from C3-6cycloalkyl, aryl or 5-6-membered saturated or unsaturated heterocyclic group with 1-2 heteroatoms, selected independently from O, S and N, which cyclic group may bear one or more substituents selected from halo and C1-4alkyl) ; and R70 is hydrogen, C1-3alkyl, or a cyclic group selected from cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and a 5-6-membered saturated heterocyclic group with 1-2 heteroatoms, selected independently from O, S and N, which C1-3alkyl group may bear 1 or 2 substituents selected from oxo, hydroxy, halogeno, C1-4alkoxy and which cyclic group may bear 1 or 2 substituents selected from oxo, hydroxy, halogeno, cyano, C1-4cyanoalkyl, C1-4alkyl, C1-4hydroxyalkyl, C1-4alkoxy, C1-4alkoxyC1-4alkyl, C1-4alkylsulphonylC1-4alkyl, C1-4alkoxycarbonyl, C1-4aminoalkyl, C1-4alkylamino, di (C1-4alkyl) amino, C14alkylaminoC,-4alkyl, di (CI-4alkyl) aminoCI-4alkyl, Cl-4alkylaminoCl-4alkoxy, di (Cl-4alkyl) aminoCl-4alkoxy and a group -(-O-)f(C1-4alkyl) gringD (wherein f is 0 or 1, g is 0 or 1 and D is a cyclic group selected from C3-6cycloalkyl, aryl or 5-6-membered saturated or unsaturated heterocyclic group with 1-2 heteroatoms, selected independently from O, S and N, which cyclic group may bear one or more substituents selected from halo and C1-4alkyl) ; and wherein Ra, Rb, Rb', R°, R°', Rd, Re, R', Rrn, Rm' Rn, Rn', Rp', Rt', Ru', Rv and Rv' are independently selected from Cl-8alkylene groups optionally substituted by one or more substituents selected from hydroxy, halogeno, amino, Rf, Rh, Rj, Rp and Rt are independently selected from C2-8alkenylene groups optionally substituted by one or more substituents selected from hydroxy, halogeno, amino, and Rt may additionally be a bond; and

Rg, Rk, and Ru are independently selected from by C2-8alkynylene groups optionally susbstituted by one or more substituents selected from hydroxy, halogeno or amino.

In many cases, it is preferred that where such groups include a bridging group Ra, Rb, Razz Rc Rc', Rd, Re, Ri Rm Rm' Rn, Rn', Rp', Rt', Ru', Rv, Rv' Rf, Rh, Rj, Rp, Rt Rg, Rk, Rm or Ru, said bridging group carries a substitutent as defined above, and in particular a hydroxy substitutent, in order to block metabolism.

In particular, at least one of Rl, R2, R3 or R4 is a group of formula Xl-R13 where R13 is a group as defined in (3"), (5"), (19") or (22").

When said group is a group of formula 3", particularly suitable groups R24 are cyclopropyl or any C1-6 alkyl group substituted by cyclopropyl. Suitably in said groups, X3 is a group NR29 where R29 is as defined in 3'above and in particular is hydrogen.

When said group is a group of formula 5", particularly suitable examples are compounds where R36 is a saturated 7-membered heterocyclic ring or R36 is a 5 or 6 membered heterocyclic ring such a a morpholine, piperidine or tetrahydropyridyl ring, which carries at least one substitutent selected from CI-4alkanoyl such as acetyl, or-C (o) NR65R66,- (wherein R65 and R66 are as defined in (5') above and in particular are hydrogen.

When said group is a group of formula 19", it is preferably an unsubstituted alkynyl group such as prop-2-ynyl.

When said group is a group of formula 22"above, it is suitably a group in which R69 is a 5-6-membered saturated heterocyclic group with 1-2 heteroatoms, such as piperidinyl. Suitably R70 is hydrogen or C1-3alkyl such as methyl. Suitably also, Xl° is oxygen. Rv and Rv' are suitably the same or different and are Cl-5 alkylene groups in particular C2-3alkylenegroups.

In one embodiment, at least one of R1, R2, or R4 is a member selected from the group consisting of hydrogen, hydroxy, halogeno, cyano, nitro, trifluoromethyl, C1-3alkyl, -NR11R12 (wherein R11 and R12, which may be the same or different, each represents hydrogen or C1-3alkyl), or a group R13-X1-(CH2)x wherein x is 0 to 3, Xl represents -O-, -CH2-, -OCO-, carbonyl, -S-, -SO-, -SO2-, -NR14CO-, -CONR15-, -SO2NR16-, -NR17SO2- or -NR18- (wherein R, R, R, R and Rl8 each independently represents hydrogen, C1-3alkyl or Ci-3alkoxyC2-3alkyl) and R13 is selected from one of the following sixteen groups: 1) C1-5alkyl which may be unsubstituted or which may be substituted with one or more groups selected from hydroxy, fluoro and amino;

2) C1-5alkylX2COR19 (wherein X2 represents -O- or -NR20- (wherein R2° represents hydrogen, C1-3alkyl or Ci-3alkoxyC2-3alkyl) and Rl9 represents-NR2lR22-or-oR23- (wherein R21, R22 and R23 which may be the same or different each represents hydrogen, C1-3alkyl or Cl-3alkoxyC2-3alkyl)) ; 3) C1-5alkylX3R24 (wherein X3 represents -O-, -S-, -SO-, -SO2-, -OCO-, -NR25CO-, -CONR26-, -SO2NR27-, -NR28SO2- or -NR29- (wherein R25, R26, R27, R28 and R29 each independently represents hydrogen, C1-3alkyl or C1-3alkoxyC2-3alkyl) and R24 represents hydrogen, C1-3alkyl, cyclopentyl, cyclohexyl or a 5 or 6 membered saturated heterocyclic group with one or two heteroatoms, selected independently from O, S and N, which Cl-3alkyl group may bear one or two substituents selected from oxo, hydroxy, halogeno and C1-4alkoxy and which cyclic group may bear one or two substituents selected from oxo, hydroxy, halogeno, C1-4alkyl, C1-4hydroxyalkyl and C1-4alkoxy) ; 4) C1-5alkylX4C1-5alkylX5R30 (wherein X4 and X5 which may be the same or different are each -O-, -S-, -SO-, -SO2-, -NR31CO-, -CONR32-, -SO2NR33-, -NR34SO2- or -NR35- (wherein R3 R32, R33, R34 and R35 each independently represents hydrogen, C1-3alkyl or Cl-3alkoxyC2-3alkyl) and R30 represents hydrogen or C1-3alkyl) ; 5) C1-5alkylR36 (wherein R36 is a 5 or 6 membered saturated heterocyclic group with one or two heteroatoms, selected independently from O, S and N, which heterocyclic group may bear one or two substituents selected from oxo, hydroxy, halogeno, C1-4alkyl, C1-4hydroxyalkyl and C1-4alkoxy); 6) (CH2) qX6R37 (wherein q is an integer from 0 to 5, x6 represents a direct bond,-O-,-S-, -SO-, -SO2-, -NR38CO-, -CONR39-, -SO2NR40-, -NR41SO2- or -NR42- (wherein R38, R39, R40, R41 and R42 each independently represents hydrogen, C1-3alkyl or Ci-3alkoxyC2-3alkyl) and R37 is a phenyl group, a pyridone group or a 5 or 6 membered aromatic heterocyclic group with 1 to 3 heteroatoms selected from O, N and S, which phenyl, pyridone or aromatic heterocyclic group may carry up to 5 substituents selected from hydroxy, halogeno, amino, C1-4alkyl, C1-4alkoxy, C1-4hydroxyalkyl, C1-4hydroxyalkoxy, C1-4aminoalkyl, C1-4alkylamino, carboxy, cyano, -CONR43R44 and -NR45COR46 (wherein R43, R44, R45 and R46, which may be the same or different, each represents hydrogen, C1-4alkyl or Cl-3alkoxyC2-3alkyl)) ; 7) C2-6alkenylR36 (wherein R36 is as defined hereinbefore); 8) C2-6alkynylR36 (wherein R36 is as defined hereinbefore); 9) X7R47 (wherein X7 is -SO2-, -O- or -CONR48R49- (wherein R48 and R49, which may be the same or different, each represents hydrogen, C1-3alkyl or Ci-3alkoxyC2-3alkyl) and R47

represents C1-5alkyl which may be unsubstituted or which may be substituted with one or more groups selected from hydroxy, fluoro and amino) with the provisos that when X7 is -S02-, Xl is-O-, when X7 is-O-, Xl is carbonyl, when X7 is -CONR48R49-, X1 is -O- or NR18 (wherein R48, R49 and Rl8 are as defined hereinbefore); 10) C2-6alkenylR37 (wherein R37 is as defined hereinbefore); 11) C2-6alkynylR37 (wherein R37 is as defined hereinbefore); 12) C2-6alkenylX8R37 (wherein x8 represents -O-, -S-, -SO-, -SO2-, -NR50CO-, -CONR51-, -SO2NR52-, -NR53SO2- or -NR54- (wherein R50, R51, R52, R53 and R54 each independently represents hydrogen, C1-3alkyl or C1-3alkoxyC2-3alkyl) and R37 is as defined hereinbefore); 13) C2-6alkynylX9R37 (wherein X9 represents -O-, -S-, -SO-, -SO2-, -NR55CO-, -CONR56-, -SO2NR57-, -NR58SO2- or -NR59- (wherein R55, R56, R57, R58 and R59 each independently represents hydrogen, C1-3alkyl or Ci-3alkoxyC2-3alkyl) and R37 is as defined hereinbefore); 14) C1-3alkylX10C1-3alkylR37 (wherein Xlo represents -O-, -S-, -SO-, -SO2-, -NR60CO-, -CONR61-, -SO2NR62-, -NR63SO2- or -NR64- (wherein R60, R61, R62, R63 and R64 each independently represents hydrogen, C1-3alkyl or Cl-3alkoxyC2-3alkyl) and R37 is as defined hereinbefore); 15) R36 (wherein R36 is as defined hereinbefore); and 16) C1-3alkylX10C1-3alkylR36 (wherein Xlo and R36 are as defined hereinbefore).

Suitably, R1 and R4 are hydrogen.

Suitably R2 is other than hydrogen and in particular are a group of formula-Xl-R13 as defined above.

The group R2 may be any of the groups defined above for Ru, R2 or R4. Preferably however said other group is a small group such as hydrogen or C1-6 alkoxy such as methoxy.

A further feature of the invention provides compounds of Formula (I) or compounds of Formula (II) wherein R1, R2 and R4 are selected from hydrogen or groups within the definition of R3 and R3 is selected from a group within the definition of Rl, R2 and R4, provided that at least one of R1, R2 and R4 is not hydrogen.

Preferably, RS is cyano.

Suitable groups Y include oxygen or-NH-and most preferably are-NH-.

In particular, in the compound of Formula (1), n is 0.

Preferably R6 is a group -R8-X-R9-.

Suitably R8 is phenyl.

Suitably X is oxygen or-NH-and most preferably oxygen.

Suitably R9 is a group Rl° as defined above (where m is 0).

Examples of optional substituents for aryl, carbocyclic or heterocyclic groups Rio include one or more groups selected from hydroxy; halo; nitro; cyano; carboxy; C1-6alkoxy ; C1-6alkyl ; C2-6alkenyl ; C2-6alkynyl ; C2-6alkenyloxy ; C2-6alkynyloxy ; C3-6cycloalkyl ; amino; mono-or di-CI-6alkyl amino; heterocyclyl optionally substituted with C1-6alkyl, oxo or C1- 6alkylthioCl-6alkyl ; C (O) Ra ; C (O) ORa ; S (O) dRa ; NRaC (O) Rb ; C (O) NRaS (O) dRb ; C (O) NRaRb ; NRaC (O) NRbR ; NRaS (O) dub or N (S (O) dRb) S (O) dR° where d is 0,1 or 2 and Ra, Rb and Rc are independently selected from hydrogen, C1-6alkyl, aryl, C3-6cycloalkyl or heterocyclyl, and wherein any alkyl, alkenyl or alkynyl group or moiety contained within the substituent on Rlo may themselves be optionally substituted with one or more groups selected from hydroxy; cyano; nitro; halo; carboxy; carboalkoxy of 2-7 carbon atoms, C3-6cycloalkyl, heterocyclyl optionally substituted with C1-6alkyl or oxo; C (O) Rd ; C (O) ORd NRdRe ; S (O) eRd ; NRdC (O) Re ; C (O) NRdRe ; NRdC (O) NReRf or NRdS (O) eRe where e is 0,1 or 2 and Rd, Re and Rf are independently selected from hydrogen or C1-6alkyl optionally substituted with one or more groups selected from hydroxy; cyano; nitro; halo; carboxy; carboalkoxy of 2-7 carbon atoms ; C3-6cycloalkyl ; heterocyclyl optionally substituted with C1-6alkyl or oxo; C (O) Rg ; C (O) ORg NRgRh; S (O) e Rg ; NRhC (O) RI ; C (O) NRgRh ; NRgC (O) NRl'Ri or NRgS (O) eRh where e is as defined above and Rg, Rh and Ri are independently selected from hydrogen, Cl-6alkyl or heterocyclyl optionally substituted with C1-6alkyl. Alternatively, two substituents on adjacent atoms may be joined to form the second ring of a bicyclic ring system wherein the said second ring is optionally substituted with one or more of the groups listed above for Rl° and optionally contains one or more heteroatoms.

In some embodiments, the substituent on the group Rl° is a complex chain. Thus, for example, a substituent may comprise a substituted alkyl chain which is optionally interposed with heteroatoms such as groups of sub-formula (i) -Xa-R70-(Xb-R71)q-(Xc)s-R72 wherein Xa, Xb and X° are independently selected from any of the groups listed above for Xl, R70 and R7l are independently selected from C1-6alkylene, C2-6alkenylene or C2-6alkynylene groups any of which may be optionally substituted with hydroxy; cyano; nitro; halo; carboxy, carboalkoxy of 2-7 carbon atoms or C3-6cycloalkyl ; R72 is hydrogen or an C1-6alkyl, C2-6 alkenyl or C2-6alkynyl group any of which may be optionally substituted with hydroxy; cyano; nitro; halo; carboxy; C3-6cycloalkyl and in

particular cyclopropyl ; or an optionally substituted heterocyclic group, in particular a group as defined above for R36, and q and s are independently 0 or 1.

A further example of a substituent on Rlo is a group of the sub-formula (ii) -Z-(CH2)pR100-R101 (ii) wherein -Z- is a direct bond or a group of sub-formula (iii) -X121-R751-X131-(R761-X141)s-(R771-X151)s'- (iii) wherein X121,X131 each Xl4l and each Xl5l are independently selected from-O-,-C (O)-, - C SO O-, -S-, -SO-, -SO2-, -NR781C(O)-, -NR781C(O)O-, -CONR791-, -C(O)ONR791-, -SO2NR801-, -NR811SO2- or -NR821- (wherein R781, R791, R801, R811 and R821 each independently represents hydrogen, C1-3alkyl optionally substituted by hydroxy, or Ci-3alkoxyC2-3alkyl) and each X 131, X141 and X151 may additionally be a direct bond; sands'isO, 1,2 or, 3; R751 R761 and R771 are independently selected from C1-6alkylene, C2-6alkenylene or C2-6alkynylene optionally substituted by halo and hydroxy or R751, R761 and R771 can each independently be direct bonds; Rloo is an optionally substituted divalent heterocyclic group, C1-5alkylene, or divalent C3-7cycloalkyl, Rlol is hydrogen, amino or a group of sub-formula (iv) -X161R8131-(X171R841)t-X181R851 (iv) wherein X161, Xi and each Xl7l are each independently selected from a direct bond,-0-, -C(O)-, -S-, -SO-, -SO2-, -NR861C(O)-, -NR861C(O)O-, -CONR871-, -C ONR871-, -SO2NR881-, -NR891SO2- or -NR901- (wherein R861, R871, R881, R891 and R901 each independently represents hydrogen, C1-3alkyl or C 1-3alkoxyC2-3alkyl), R831 and each R841 are independently selected from C1-6alkylene, C2-6alkenylene or C2-6alkynylene, R851 is selected from hydrogen, C1-6alkyl, C2-6alkenyl or C2-6alkylyl, and <BR> <BR> <BR> tis0, 1, 2or3.<BR> <BR> <BR> <BR> <BR> <BR> <BR> pis0, 1, 2Or3

In some embodiments, Rlo is a heterocyclic ring containing one or 2 oxygen atoms.

Particular examples of groups Rlo include phenyl or cycloalkyl of from 3-8 and preferably of 6 carbon atoms which are substituted at the alpha position. Preferably however, R10 is substituted phenyl.

Preferably Rlo is a substituted with an optionally substituted alkoxy group wherein substituents are as described above. For example Rl° is phenyl substituted at the alpha (ortho position) by methoxy,-OCH2 (C (O) NHCH2) fC (O) NH (R73), where f is 0 or 1, and R73 is CI-4 alkyl such as methyl or C3-6cycloalkyl such as cyclopropyl.

Examples of heterocyclic rings Rl° include 3-7 membered rings, up to two of which may be oxygen atoms. Such groups include: where each R65 is independently selected from hydrogen or C1-6alkyl and especially methyl.

In such compounds, m in R9 is suitably 1,2 or 3.

Other examples of heterocyclic groups Rlo include pyridyl, thiazolyl, pyrazinyl, pyrimidinyl, oxadiazole.

Particular examples of groups Rl° include divalent phenyl, pyridyl or C3-8cycloalkyl.

Most preferably however, Rlo is optionally substituted phenylene. Rloo is preferably a group selected from a divalent C1-2 alkylene, or divalent C3-4 cycloalkyl, pyridyl, or pyrrolidinyl or phenylene.

In a preferred embodiment R8 and Rlo are both phenylene; Y is-NH and X is oxygen; and n, m and p are all 0.

Preferably Rloi is hydrogen, Suitable further substituents for R100 and R10 include those listed above for pyridyl, pyrimidinyl and phenyl groups R8. A particularly preferred substituent for Rlo is fluoro.

Suitable examples of variables within sub-formula (i) defined above are as follows: X12 is suitably-0- ;

R7s is suitably Cl-6alkylene and preferably the group-C (RA RB)-where RA and RB are each independently selected from C 1-3 alkyl, C 2-3 alkenyl, C 2-3 alkynyl and halo, most preferably both either C1 alkyl or fluoro.

R76 and R77 are suitably the same or different and are preferably independently selected from Cl-6alkylene and in particular methylene groups and a direct bond. Most preferably R77 is a direct bond.

X13 is preferably the group-CoNR79-or-NR78C (O)-, most preferably the group- CoNR79-, where R78 and R79 are selected from hydrogen or C1-3alkyl and are more preferably hydrogen; X14 is suitably-C (O)-, -CONR79-m, where R78 and R79 are selected from hydrogen or C1-3alkyl and are preferably hydrogen, or a direct bond; s, q and p are all preferably 0.

Where Z is a group of sub-formula (i), a particularly preferred group is selected from -O-C(CH3)2-CONR791- -O-C(F)2- -O-C(CH3)2-CONR791-CH2-CONR791- -O-C(CH3)2-CONR791-CH2-CONR791-(CH2)2-NR821- -O-C(F)2-CONR791- -0-C (F) 2-CONR791-CH2-CONR791- -(CH2)2-CONR791-CH2-NR821- -O-C(CH3)2-CON791-(CH2)2-SO2- -O-C(CH3)2-CONR791-(CH2)2-S- -0-C (CH3) 2-CONR791-(CH2)2-O- where R791, R82l are preferably as hereinbefore defined and preferably are methyl or hydrogen, most preferably hydrogen.

Alternatively Z is preferably a direct bond.

Examples of suitable heterocyclic groups Rloo are 5-or 6-membered aromatic or non- aromatic rings which contain up to 4 and preferably up to 3 heteroatoms. A particular example of a non-aromatic group Rloo is piperazine or morpholine or piperidine linked via carbon or nitrogen but preferably by nitrogen atoms whilst an example of an aromatic group is oxadiazole.

Where Z is a group of sub-formula (iii), R"D may be substituted by a simple C-16alkyl group such as methyl. However, where Z is a direct bond, more complex substituents are required to be present on the ring Rloo. In such instances, at least one group Xl6l, X171 or Xl8l, and preferably at least X181, in the group of sub-formula (iv) is other than a direct bond.

Preferably, at least one such group, and most preferably Xl is-S-,-S (O)- or-S (O) 2-.

Suitable further substituents for Rl° include those listed above for pyridyl, pyrimidinyl and phenyl groups R6.

A preferred sub-group of compounds of the invention comprise composed of Formula (I) or Formula (Ia) wherein: R1 and R4 are each hydrogen; R2 is C1-5alkoxy; is selected from group (i) or group (ii); R5 is cyano; Y is-NH- ; n is 0; R6 is a group of the formula-R8-X-R9 ; R8 is a divalent phenyl ring, optionally substituted by halogeno, preferably fluoro; X is-0-; R9 is a group (CH2)mR10; m is 0; and Rl° is optionally substituted phenyl; or pharmaceutical-acceptable salts, pro-drugs or solvate thereof. a further preferred sub-group of compounds of Formula (I) are compounds of Formula (II) where R1, R2 R3, R4 and R5 are as defined above and R66 is optionally substituted C1-6alkyl in particular methyl and R67 is selected from hydrogen, halogen, alkyl of 1-6 carbon atoms, alkenyl of 2-6 carbon atoms, alkynyl of 2-6 carbon atoms, azido, hydroxyalkyl of 1-6 carbon atoms, halomethyl, alkoxymethyl of 2-7 carbon atoms, alkanoyloxymethyl of 2-7 carbon atoms, alkoxy of 1-6 carbon atoms, alkylthio of 1-6 carbon atoms, hydroxy, trifluoromethyl, cyano, nitro, carboxy, carboalkoxy of 2-7 carbon atoms, carboalkyl of 2-7 carbon atoms, phenoxy, phenyl, thiophenoxy, benzoyl, benzyl, amino, alkylamino of 1-6 carbon atoms, dialkylamino of 2 to 12 carbon atoms, phenylamino, benzylamino, alkanoylamino of 1-6 carbon atoms, alkenoylamino of 3-8 carbon atoms, alkynoylamino of 3-8 carbon atoms, and benzoylamino.

Suitably R66 is C1-6 alkyl such as methyl. Preferably however it is a substituted Cl-4 alkyl group, wherein the substitutents are selected from hydroxy, NRdRe, S (O) eRd, VRAC (O) Re ; C (O) NRdRe ; NRdC (O) NReRf ; NRdS (O) eRe where e, Rd, Re and Rf are as defined above.

In particular, R66 is a group-CH2 (C (O) NHCH2) pC (O) NH (R73), where p and R73 are as defined above.

Preferably W7 is hydrogen.

A further group of compounds according to the invention are compounds of Formula

wherein R130 is a group of formula (i) as hereinbefore defined and preferably is selected from: -NR78 C (O)-R9-R6,-NR82-Rloo-Rioyand Cl-4 allcoxyl ; hydrogen or halo, particularly fluoro, where k", R82, Rloo and Rlol are as hereinbefore defined; *131 is a hydrogen or halo, particularly fluoro ; and R132 is a C1-4 alkoxy and pharmaceutical-acceptable salts, pro-drugs or solvate thereof.

A preferred group of compounds of the invention comprises 7-[(3-(4,4-difluoropiperidin-1-yl)propoxy]-6-methoxy-4-{[3-( 2-methoxyphenoxy) phenyl] amino} quinoline-3-carbonitrile ; <BR> <BR> <BR> <BR> 7- [3- (4-isopropylpiperazin-1-yl) propoxy]-6-methoxy-4- { [3- (2-methoxyphenoxy) phenyl]<BR> <BR> <BR> <BR> <BR> <BR> <BR> amino} quinoline-3-carbonitrile ; 7- [3- (3, 3-difluoropyrrolidin-1-yl) propoxy]-6-methoxy-4- {[3-(2-methoxyphenoxy) phenyl] amino} quinoline-3-carbonitrile; 7-[3-(1,1-dioxidothiomorpholin-4-yl)propoxy)]-6-methoxy-4-{[ 4-(2- methoxyphenoxy) phenyl] amino} quinoline-3-carbonitrile ; 6-methoxy-4- { [4- (2-methoxyphenoxy) phenyl] amino}-7- (3-morpholin-4-yl-2- oxopropoxy) quinoline-3-carbonitrile; 6-methoxy-4-f [4- (2-methoxyphenoxy) phenyl] amino}-7- (2-oxo-3-pyrrolidin-1- ylpropoxy) quinoline-3-carbonitrile; 7-[3-(2,6-dimethylpiperidin-1-yl)-2-oxopropoxy]-6-methoxy-4- {[4-(2- methoxyphenoxy) phenyl] amino} quinoline-3-carbonitrile; and

7-f{3- [ (1, 1-dioxidotetrahydrothien-3-yl) (methyl) amino] propoxy}-6-methoxy-4-{[4-(2- methoxyphenoxy) phenyl] amino} quinoline-3-carbonitrile and pharmaceutical-acceptable salts, pro-drugs or solvate thereof.

A further preferred group of compounds of the invention comprises 7- (3-azetidin-1-yl-2-oxopropoxy)-6-methoxy-4- f [4- (2- methoxyphenoxy) phenyl] amino} quinoline-3-carbonitrile ; 7- {3-[4-(ethylsulfonyl)piperazin-1-yl]propoxy}-6-ethoxy-4-{[4- (2- methoxyphenoxy) phenyl] amino} quinoline-3-carbonitrile ; 6-methoxy-7- {3- [4- (2-methoxyethyl) piperazin-1-yl] propoxy}-4- {[4-(2- methoxyphenoxy) phenyl] amino} quinoline-3-carbonitrile ; 6-methoxy-4- [4-(2-methoxyphenoxy)phenyl]amino}-7-{3-[3-(methylsulfonyl)p yrrolidin- 1-yl] quinoline-3-carbonitrile ; and 7- [3- (4-acetylpiperazin-1-yl) propoxy]-6-methoxy-4- {[4-(2- methoxyphenoxy) phenyl] amino} quinoline-3-carbonitrile ; 7- {3- [bis (2-hydroxyethyl) amino] propoxy}-6-methoxy-4- { [4- (2- methoxyphenoxy) phenyl] amino} quinoline-3-carbonitrile ; 7- [3- (cyclopropylamino) propoxy]-4-1 [2-fluoro-4- (1, 3-thiazol-2-yloxy) phenyl] amino}-6- methoxyquinoline-3-carbonitrile; 7- [3-(cyclopropylamino)propoxy]-4-{[2-fluoro-4-(isothiazol-3-y loxy) phenyl] amino}-6- methoxyquinoline-3-carbonitrile ; 2-[2-[4-({3-cyano-7-[3-(cyclopropylamino) propoxy]-6-methoxyquinolin-4- yl} amino) phenoxy] phenoxy}-N-methylacetamide ; and 7-[3-(cyclopropylamino)propoxy]-6-methoxy-4-{[4-(2- methoxyphenoxy) phenyl] amino} quinoline-3-carbonitrile ; and pharmaceutical-acceptable salts, pro-drugs or solvate thereof.

Particular examples of compounds of Formula (I) or Formula (Ia) are listed in Tables 1,2,3and4.

Table 1 Compd. R3 R76 No. 1 OH OCH3 * \ozon 2 OCH3 OH 3 O (CH2) 3NH (CH2) 20H OCH3 4 gOH OCH3 * ON 5 OH OCH3 OH 6 O (CH2) 3N (CH3) (CH2) 20H OCH3 7 \Oz NJ OH OCH3 8 OCH3 OH OH 9 OCH3 zou Compd. R3 R76 No. 10 o H 12 rN" NI I O zu 11 * N --) r 12 % NNJ II N\CHa O 13 H 0 o 0 H % \N/N' N 15 O (CH2) 3NH (CH2) 2SCH3 OCH3 16 OCH3 N s 17 N 17 H HN H 0 Table 2 Compd. R2 R3 R" No. 18 OCH3 OCH3 N "O-CH, 19 OCH3 OCH2C (O) NHCH3 \OZON H 20 OCH2C=CH OCH3 O II N H. zozo 21 OCH3 OCH3 ON H 22 OCH3 OCH2C (O) NHCH3 H <1 H 23 OCH3 soz N OCH3 N 24 OCH3 NOnNX OCH3 \ O o 25 OCH3 OCH3 OUZO 0 26 OCH3 sow NX OCH3 NH 0 27OCH2C=CHOCH3H ?"dz 0 28 OCH2C=-CH H ro 0 N Compd. R2 ~ R3 RX No. 29 OCH3 oN (cH3) 2 OCH3 ""0 \ON 30 OCH3 OCH2CH (OH) CH2N (CH3) 2 OCH3 31 OCH3 OCH2C (O) NHCH3 H H H H V 32 OCH3 OCH3 0 N H 33 OCH3 0 asz zu "o<J 34 OCH3 oN OCH3 OH 35 OCH3 OCH3 OU OH 36 OCH3 OCH3 0 "N N-S 0 37 OCH3 OH OCH3 UN OH 38 OCH3 0 OCH3 OH 39 OCH3 OCH3 N + 0'0 Table 3 Compd. R3 R Rlo No. 40 \ON F H S 41 H N-cz 1- L H Table 4 Compd. R3 Rl° No. * i 42 0 0 zu

Compd. 3 Rl° No. 43 y Y3 F 44 yN. i N * 0 45 0--'-'-N 40 L * zu 47 0 4snot 45 o ~'NF o p O * 40 sO/ 0 0 Zu 49 kS oi OH S * V 50 o 0 oN IN 'OH 51 s.. o S Compd. R3 Rl° No. 52 O O 53 O O oi zu / u 54 0y 0 0 * zu 55 0,,, zu 56 N 0 " / 57 *. , o ) 59 ou zon * 59 y o s Compd. R3 Rl° No. 60-O-CH2-CN zu 61 y, zu * zu 1 0 O * OH Table 5 Compd R3 Rb No. 63 H oi Table 6 Compd R3 Rl o No. 4 z ~/OCH2C (O) NHCH3 NO 0y CH3 Compounds of Formula (I) are suitably prepared by reacting a compound of formual (III) where Rl, R2', R3', R4' represent R1, R2, R3 and R4 respectively as defined in relation to Formula (I) or a precursor thereof, Rs is as defined in relation to Formula (1) and Z'is a leaving group, with a compound of Formula (IV) H-Y(CH2)nR6' (IV)

where Y, and n are as defined in relation to Formula (I), and R6 is a group R6 as defined in relation to Formula (1) or a precursor thereof; and thereafter if necessary or desired converting precursor groups Rl, Ruz', R3', R4', and R6' to groups of formula Rl, R2, R3, R4 and R respectively, or converting a group Rl, R2, R3, R4 and R6 to a different such group.

Suitable leaving groups for Z'include halogen such as bromo or chloro, or a mesylate or tosylate group or a substituted phenoxy group.

The reaction is suitably carried out in an organic solvent such as an alcohol for example propanol or cyclohexanol at elevated temperatures, for example of from 50 to 150°C, for example at about 105°C.

Conversion reactions in which precursor groups Rl, R2', le" R4 are converted to groups of formula R1, R2, R3 and R4 respectively, or groups Rl, R2, R3 and R4 are converted to different such group can be carried out using conventional chemistry as outlined hereinafter.

Particular precursor groups Rl, R2'm, R3', R4 are groups of formula R13'-X1-(CH2)x wherein x and Xl are as defined herein, and R13' is C1-5alkyl which is substituted with halo other than fluoro, and in particular chloro or bromo. The chloro or bromo group may readily be converted into many other groups R13 as defined in relation to claim 1." Similarly conversion reactions involving groups R may be effected using conventional chemistry. For example substituent groups on a group Rl° within the group R6 may be changed, for example by changing acids to esters or amides etc.

Alternatively, compounds of Formula (1) where R is a group-R8-X-R9 are prepared by reacting a compound of Formula (V) where R1', R2', R3', R4' are as defined in relation to Formula (III) R8, X, Y and n are as defined in relatjion to Formula (I), with a compound of Formula (VI) R9'-Z" (VI)

where R9' is a group R9 as defined in relation to Formula (1) or a precursor thereof and Z"is a leaving group ; and thereafter if necessary or desired converting precursor groups Rl, R2, R3, R4 and R9 to groups of formula Rl, R2, R3, R4 and R9 respectively, or converting a group Rl, R2, R3, R4 and R9 to a different such group. Suitable leaving groups for Z"include halogen such a bromo or chloro, or a mesylate or tosylate group.

The reaction is suitably carried out in an organic solvent such as DMF at elevated temperatures, for example of from 40 to 120°C, for example at about 80°C. Conversion reactions are conventional and can be derived from literature information..

Compounds of Formula (III) and (V) are either known compounds or they can be prepared from known compounds by conventional methods, for example as described in WO 98/43960 and WO 98/13350.

Compounds of Formula (IV) are also known compounds (see for example Rev. Chim.

(Bucharest) (1988), 39 (6), 477-82 and DD 110651: 74.01.05) or they can be prepared from known compounds using conventional methods. For example, where Y is NH, compounds of Formula (IV) are suitably prepared by reduction of a compound of formula (VET) O2N(CH2)nR8XR9' (VII) where X, R8, R9' and n are as defined above. It may be convenient to convert precursor groups R9 to groups R9 or groups R9 to other such groups at the level of compound of Formula (VH) or (IV) using conventional chemistry.

Compounds of Formula (VI) are also known compounds or they can be prepared from known compounds by conventional methods.

Compounds of the invention are useful in the inhibition of MEK enzyme activity and can be used in the treatment of proliferative disease. They will suitably be in the form of a pharmaceutical composition, in combination with a pharmaceutically acceptable carrier. Such compositions form a further aspect of the invention.

The compositions of the invention may be in a form suitable for oral use (for example as tablets, lozenges, hard or soft capsules, aqueous or oily suspensions, emulsions, dispersible powders or granules, syrups or elixirs), for topical use (for example as creams, ointments, gels, or aqueous or oily solutions or suspensions), for administration by inhalation (for

example as a finely divided powder or a liquid aerosol), for administration by insufflation (for example as a finely divided powder) or for parenteral administration (for example as a sterile aqueous or oily solution for intravenous, subcutaneous, intramuscular or intramuscular dosing or as a suppository for rectal dosing).

The compositions of the invention may be obtained by conventional procedures using conventional pharmaceutical excipients, well known in the art. Thus, compositions intended for oral use may contain, for example, one or more colouring, sweetening, flavouring and/or preservative agents.

Suitable pharmaceutically acceptable excipients for a tablet formulation include, for example, inert diluents such as lactose, sodium carbonate, calcium phosphate or calcium carbonate, granulating and disintegrating agents such as corn starch or algenic acid; binding agents such as starch; lubricating agents such as magnesium stearate, stearic acid or talc; preservative agents such as ethyl or propyl p-hydroxybenzoate, and anti-oxidants, such as ascorbic acid. Tablet formulations may be uncoated or coated either to modify their disintegration and the subsequent absorption of the active ingredient within the gastrointestinal tract, or to improve their stability and/or appearance, in either case, using conventional coating agents and procedures well known in the art.

Compositions for oral use may be in the form of hard gelatin capsules in which the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules in which the active ingredient is mixed with water or an oil such as peanut oil, liquid paraffin, or olive oil.

Aqueous suspensions generally contain the active ingredient in finely powdered form together with one or more suspending agents, such as sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethylcellulose, sodium alginate, polyvinyl-pyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents such as lecithin or condensation products of an alkylene oxide with fatty acids (for example polyoxyethylene stearate), or condensation products of ethylene oxide with long chain aliphatic alcohols, for example heptadecaethyleneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example heptadecaethyleneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and

hexitol anhydrides, for example polyethylene sorbitan monooleate. The aqueous suspensions may also contain one or more preservatives (such as ethyl or propyl p-hydroxybenzoate, anti-oxidants (such as ascorbic acid), colouring agents, flavouring agents, and/or sweetening agents (such as sucrose, saccharine or aspartame).

Oily suspensions may be formulated by suspending the active ingredient in a vegetable oil (such as arachis oil, olive oil, sesame oil or coconut oil) or in a mineral oil (such as liquid paraffin). The oily suspensions may also contain a thickening agent such as beeswax, hard paraffin or cetyl alcohol. Sweetening agents such as those set out above, and flavouring agents may be added to provide a palatable oral preparation. These compositions may be preserved by the addition of an anti-oxidant such as ascorbic acid.

Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water generally contain the active ingredient together with a dispersing or wetting agent, suspending agent and one or more preservatives. Suitable dispersing or wetting agents and suspending agents are exemplified by those already mentioned above. Additional excipients such as sweetening, flavouring and colouring agents, may also be present.

The pharmaceutical compositions of the invention may also be in the form of oil-in-water emulsions. The oily phase may be a vegetable oil, such as olive oil or arachis oil, or a mineral oil, such as for example liquid paraffin or a mixture of any of these. Suitable emulsifying agents may be, for example, naturally-occurring gums such as gum acacia or gum tragacanth, naturally-occurring phosphatides such as soya bean, lecithin, and esters or partial esters derived from fatty acids and hexitol anhydrides (for example sorbitan monooleate) and condensation products of the said partial esters with ethylene oxide such as polyoxyethylene sorbitan monooleate. The emulsions may also contain sweetening, flavouring and preservative agents.

Syrups and elixirs may be formulated with sweetening agents such as glycerol, propylene glycol, sorbitol, aspartame or sucrose, and may also contain a demulcent, preservative, flavouring and/or colouring agent.

The pharmaceutical compositions may also be in the form of a sterile injectable aqueous or oily suspension, which may be formulated according to known procedures using one or more of the appropriate dispersing or wetting agents and suspending agents, which have been mentioned above. A sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example a solution in 1,3-butanediol.

Suppository formulations may be prepared by mixing the active ingredient with a suitable non-irritating excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug. Suitable excipients include, for example, cocoa butter and polyethylene glycols.

Topical formulations, such as creams, ointments, gels and aqueous or oily solutions or suspensions, may generally be obtained by formulating an active ingredient with a conventional, topically acceptable, vehicle or diluent using conventional procedure well known in the art.

Compositions for administration by insufflation may be in the form of a finely divided powder containing particles of average diameter of, for example, 30je, or much less, the powder itself comprising either active ingredient alone or diluted with one or more physiologically acceptable carriers such as lactose. The powder for insufflation is then conveniently retained in a capsule containing, for example, 1 to 50mg of active ingredient for use with a turbo-inhaler device, such as is used for insufflation of the known agent sodium cromoglycate.

Compositions for administration by inhalation may be in the form of a conventional pressurised aerosol arranged to dispense the active ingredient either as an aerosol containing finely divided solid or liquid droplets. Conventional aerosol propellants such as volatile fluorinated hydrocarbons or hydrocarbons may be used and the aerosol device is conveniently arranged to dispense a metered quantity of active ingredient.

For further information on Formulation the reader is referred to Chapter 25.2 in Volume 5 of Comprehensive Medicinal Chemistry (Corwin Hansch; Chairman of Editorial Board), Pergamon Press 1990.

The amount of active ingredient that is combined with one or more excipients to produce a single dosage form will necessarily vary depending upon the host treated and the particular route of administration. For example, a formulation intended for oral administration to humans will generally contain, for example, from 0.5 mg to 2 g of active agent compounded with an appropriate and convenient amount of excipients which may vary from about 5 to about 98 percent by weight of the total composition. Dosage unit forms will generally contain about 1 mg to about 500 mg of an active ingredient. For further information on Routes of Administration and Dosage Regimes the reader is referred to Chapter 25.3 in

Volume 5 of Comprehensive Medicinal Chemistry (Corwin Hansch ; Chairman of Editorial Board), Pergamon Press 1990.

The size of the dose for therapeutic or prophylactic purposes of a compound of the Formula I will naturally vary according to the nature and severity of the conditions, the age and sex of the animal or patient and the route of administration, according to well known principles of medicine. As mentioned above, compounds of the Formula I are useful in treating diseases or medical conditions which are due alone or in part to the effects MEK enzymes.

In using a compound of the Formula I for therapeutic or prophylactic purposes it will generally be administered so that a daily dose in the range, for example, 0.5 mg to 75 mg per kg body weight is received, given if required in divided doses. In general lower doses will be administered when a parenteral route is employed. Thus, for example, for intravenous administration, a dose in the range, for example, 0.5 mg to 30 mg per kg body weight will generally be used. Similarly, for administration by inhalation, a dose in the range, for example, 0.5 mg to 25 mg per kg body weight will be used. Oral administration is however preferred.

In a further aspect, the invention provides a method of treating proliferative disease by administering a compound of Formula (I) as described above, or a pharmaceutical composition as described above.

Yet a further aspect of the invention provides the use of a compound of Formula (1) as defined above, in the preparation of a medicament for use in the inhibition of MEK enzyme activitiy and in particular for the treatment of proliferative disease such as cancer.

According to a further aspect of the invention there is provided a compound of the Formula (I), Formula (Ia), Formula (II) or Formula (hui) or a phannaceutically-acceptable salt or prodrug or solvate thereof, as defined hereinbefore, for use in a method of treatment of the human or animal body by therapy.

We have found that the compounds of the present invention possess potent anti- tumour activity which it is believed is obtained by way of inhibition the MAPK pathway and, in particular, inhibition of MEK enzyme Accordingly the compounds of the present invention are of value as anti-proliferative agents.

Thus according to this aspect of the invention there is provided the use of a compound of Formula (I), Formula (Ia), Formula (It) or Formula (ici) or a pharmaceutically-acceptable

salt, prodrug or solvate thereof, as defined hereinbefore in the manufacture of a medicament for use as an anti-proliferative agent in the containment and/or treatment of solid tumour disease.

According to a further feature of this aspect of the invention there is provided a method for producing an anti-proliferative effect by the containment and/or treatment of solid tumour disease in a warm-blooded animal, such as man, in need of such treatment which comprises administering to said animal an effective amount of a compound of Formula (I), Formula (Ia), Formula (II) or Formula (E) or a pharmaceutically-acceptable salt thereof, prodrug or solvate thereof as defined hereinbefore.

According to a further aspect of the invention there is provided the use of a compound of Formula (1), Formula (Ia), Formula (D) or Formula (E) or a pharmaceutically-acceptable salt thereof, prodrug or solvate thereof as defined hereinbefore in the manufacture of a medicament for use in the prevention or treatment of solid tumour disease in a warm-blooded animal such as man.

According to a further feature of this aspect of the invention there is provided a method for the prevention or treatment of solid tumour disease in a warm-blooded animal, such as man, in need of such treatment which comprises administering to said animal an effective amount of a compound of Formula (1), Formula (Ia), Formula (H) » or Fonnula (m) or a pharmaceutically-acceptable salt thereof, prodrug or solvate thereof as defined hereinbefore.

According to a further aspect of the invention there is provided the use of a compound of Formula (1), Formula (Ia), Formula (D) or Formula (E) or a pharmaceutically-acceptable salt thereof, prodrug or solvate thereof as defined hereinbefore in the manufacture of a medicament for use in the prevention or treatment of those tumours which are sensitive to inhibition of MEK enzymes.

According to a further feature of this aspect of the invention there is provided a method for the prevention or treatment of those tumours which are sensitive to inhibition of MEK enzymes which comprises administering to said animal an effective amount of a compound of Formula (1), Formula (Ia) Formula (II) or Formula (m) or a pharmaceutically-acceptable salt thereof, prodrug or solvate thereof as defined hereinbefore.

The anti-proliferative treatment defined hereinbefore may be applied as a sole therapy or may involve, in addition to the compounds of the invention, conventional surgery or

radiotherapy or chemotherapy. Such chemotherapy may include one or more of the following categories of anti-tumour agents:- (i) anti-invasion agents (for example metalloproteinase inhibitors like marimastat and inhibitors of urokinase plasminogen activator receptor function); (ii) other antiproliferative or antineoplastic drugs and combinations thereof, as used in medical oncology, such as alkylating agents (for example cis-platin, carboplatin, cyclophosphamide, nitrogen mustard, melphalan, chlorambucil, busulphan and nitrosoureas) ; antimetabolites (for example antifolates such as fluoropyrimidines like 5-fluorouracil and tegafur, raltitrexed, methotrexate, cytosine arabinoside and hydroxyurea, or, for example, one of the preferred antimetabolites disclosed in European Patent Application No. 562734 such as (2S)-2- {_-fluoro-p-[N- {2,7-dimethyl-4-oxo-3,4-dihydroquinazolin-6-ylmethyl)- N- (prop-2-ynyl) amino] benzamido}-4- (tetrazol-5-yl) butyric acid); antitumour antibiotics (for example anthracyclines like adriamycin, bleomycin, doxorubicin, daunomycin, epirubicin, idarubicin, mitomycin-C, dactinomycin and mithramycin) ; antimitotic agents (for example vinca alkaloids like vincristine, vinblastine, vindesine and vinorelbine and taxoids like taxol and taxotere); and topoisomerase inhibitors (for example epipodophyllotoxins like etoposide and teniposide, amsacrine, topotecan and camptothecin); (iii) cytostatic agents such as antioestrogens (for example tamoxifen, toremifene, raloxifene, droloxifene and iodoxyfene), antiandrogens (for example bicalutamide, flutamide, nilutamide and cyproterone acetate), LHRH antagonists or LHRH agonists (for example goserelin, leuprorelin and buserelin), progestogens (for example megestrol acetate), aromatase inhibitors (for example as anastrozole, letrazole, vorazole and exemestane) and inhibitors of 5a-reductase such as finasteride ; (iv) inhibitors of growth factor function, for example such inhibitors include growth factor antibodies, growth factor receptor antibodies, tyrosine kinase inhibitors and serine/threonine kinase inhibitors, for example inhibitors of the epidermal growth factor family (for example the EGFR tyrosine kinase inhibitors N- (3-chloro-4-fluorophenyl)-7-methoxy- 6- (3-morpholinopropoxy) quinazolin-4-amine (ZD1839), N- (3-ethynylphenyl)- 6,7-bis (2-methoxyethoxy) quinazolin-4-amine (CP 358774) and 6-acrylamido-N- (3-chloro- 4-fluorophenyl)-7- (3-morpholinopropoxy) quinazolin-4-amine (CI 1033)), for example inhibitors of the platelet-derived growth factor family and for example inhibitors of the hepatocyte growth factor family; and

(v) antiangiogenic agents such as those which inhibit vascular endothelial growth factor such as the compounds disclosed in International Patent Applications WO 97/22596, WO 97/30035, WO 97/32856 and WO 98/13354 and those that work by other mechanisms (for example linomide, inhibitors of integrin avp3 function and angiostatin).

Such conjoint treatment may be achieved by way of the simultaneous, sequential or separate dosing of the individual components of the treatment. Such combination products employ the compounds of this invention within the dosage range described hereinbefore and the other pharmaceutically-active agent within its approved dosage range.

According to this aspect of the invention there is provided a pharmaceutical product comprising a compound of Formula (I) or Formula (Ia) or a pharmaceutically-acceptable salt thereof, prodrug or solvate thereof as defined hereinbefore and an additional anti-tumour agent as defined hereinbefore for the conjoint treatment of cancer.

Although the compounds of the present invention are primarily of value as therapeutic agents for use in warm-blooded animals (including man), they are also useful whenever it is required to inhibit the effects of MEK enzyme. Thus, they are useful as pharmacological standards for use in the development of new biological tests and in the search for new pharmacological agents.

The invention will now be particularly described by way of example.

Examples Ill the examples the following abbreviations have been used.

DMF N, N-dimethylformamide THF tetrahydrofuran DMSO dimethyl acetamide DEAD diethyl azodicarboxylate Ph3 P triphenylphosphine EDC ethylene dichloride (1,2-dichloroethane) DCM dichloromethane (methylenechloride) DMAP dimethylaminepyridine HOBT N-hydroxybenzotriazole EDAC 1- (3-dimethylaminopropyl)-3-ethyl-carbodiimide KHMDS potassium hexamethyldisilazane (potassium bis (trimethylsilyl) amide) Preparation of Key Intermediates Preparation A Chloroquinoline intermediates These can be prepared for example using the following scheme where"Bz"represents benzyl. ETOOC E ci T cri MeO EtO MeO ETOOC OOET POC6 MeO OOET --- I \ I/i Bz0 HZ Bz0 Bz0 H (A1) (A2) (A3) ci ci ci NaOHMeO ef OOH t sOcX MeO tONH2 POCb MeO tN EtOH BZO 2 NH3 BZO BZO (A4) (A5) (A6) ci TFA meo N Cl--"Br MeO N thioanisole. Ho KOtBu (A7) (A8) A mixture of (Al) (10.36g., 45.3 mmole) and diethylethoxymethylene malonate (9mL, 45.3 mmole) was heated at 110 ° C for 1 hour and then allowed to cool overnight. The mixture was evaporated and the product (A2) used in the next step without further purification.

Mass Spectrum m/e 400 (M++H).

Preparation of (A3) A mixture of (A2) (assumed 45,. 3 mmole) and phosphoryl chloride (83.3mL, 906 mmole) was heated at 115 ° C for 18 hours. After cooling, the solution was evaporated to remove excess phosphoryl chloride. The residue was treated with ice and aqueous ammonia to hydrolyse the remaining phosphoryl chloride. The solid product was filtered off and dried in a vacuum oven to give a cream coloured solid, 9. Og (53% yield).

Mass Spectrum m/e 372 (M++H).

Preparation of (A4) A mixture of (A3) (9.0g, 24.2 mmole) was stirred in ethanol (48.3mL) for 15 minutes at ambient temperature to give a smooth suspension. Aqueous sodium hydroxide solution (2.0M, 48.3mL, 96.7 mmole) was added and the mixture stirred for 18 hours at ambient temperature. The ethanol was removed by rotary evaporation and the resulting solution was

acidified to pH 2 with hydrochloric acid while stirring. The precipitate was filtered off and dried in a vacuum oven to give an orange solid, 7.19g (86% yield).

Mass Spectrum m/e 344 (M++H).

Preparation of (A5) A mixture of (A4) (7.18g, 20.9 mmole) and thionyl chloride (90 mL) was refluxed for 2 hours.

After cooling the excess thionyl chloride was removed by rotary evaporation and the residue was suspended in acetone (175mL) and the resulting suspension cooled in an ice-bath.

Aqueous ammonia (S. G. 0.880,20mL) was added gradually, keeping the temperature below 10 ° C. The resulting suspension was filtered off, washed with water and air-dried to give a solid, 5.15g (75% yield).

Mass Spectrum m/e 343 (M++H).

Preparation of (A6) A mixture of (A5) (20.55g, 60 mmole) and phosphoryl chloride (250mL) was heated and stirred at 120 ° C for 4 hours when the starting material had dissolved. Heating and stirring was continued at 110 ° C for 18 hours. After cooling, the solution was evaporated to remove excess phosphoryl chloride. Last traces of phosphoryl chloride were removed by azeotroping with toluene. The residue was treated with ice and aqueous ammonia to remove acidity. The solid product was filtered off and dried in a vacuum oven to give a grey solid, 19.23g (99% yield).

(This may also be prepared as described in WO 9843960) Mass Spectrum m/e 325 (M++H).

Preparation of (A7) A mixture of (A6) (19.23g, 60.0 mmole) and trifluoroacetic acid (300 ml) and thioanisole (35ml) was refluxed in a nitrogen atmosphere for 3 hours. After cooling the trifluoroacetic acid was removed by rotary evaporation and the oily residue was stirred with ice and water and basified with aqueous ammonia (S. G. 0.880). The resulting suspension was filtered and the solid was washed successively with water, ethyl acetate and diethyl ether and then dried to give a khaki solid, 13.74g (97% yield).

Mass Spectrum m/e 235 (M++H).

Preparation of (A8) Potassium tert-butoxide (5. 0g) was added portionwise to a stirred solution of (A7) (10 g) in DMA (200ml) cooled to 5°C under an atmosphere of nitrogen. The mixture was stirred at

ambient temperature for 30min. and then cooled to 5°C. l-chloro-3-bromopropane (7.4g) was added followed by tetrabutylammonium iodide (1.57g) and 18-crown-6 (0. 5g) and the mixture stirred at ambient temperature for 16hr. The DMA was removed in vacuo and the residue partitioned between ethyl acetate and water. The organic extracts were dried and evaporated to dryness. The product was purified by flash column chromatography on silica eluting with 1-2 % methanol in dichloromethane to give (A8) as a white solid (6.5g).

Mass Spectrum m/e 311 (M++H).

Preparation B Preparation of (B2) Intermediate (B2) was prepared as described in Rev. Chim. (Bucharest), 1988,39 (6), 477- 482.

Preparation of (B3) A mixture of the 4-chloro-3-cyano-7-hydroxy-6-methoxyquinoline (17.6g) and 4- (2- methoxyphenoxy)-aniline (17. 2g) in 1-propanol (600ml) was stirred and refluxed for 4 hours.

The mixture was allowed to cool to room temperature overnight and the product was filtered off and washed with 1-propanol and then dried under high vacuum. The product was obtained as a yellow hydrochloride salt, 32.2g (96% yield).

Mass Spectrum m/e 414 (M++H)

Preparation C Preparation of (C1) 4-fluoro-nitrobenzene and ethyl 3-hydroxybenzoate were reacted together in DMA in the presence of potassium butoxide for 2hours at 150°C to yield Intermediate Cl.

Mass Spectrum m/e 283.27 (M-H+)- Preparation of (C2) Intermediate (Cl) was reduced to the corresponding aniline by reduction, at room temperature in ethyl acetate solution, with hydrogen and catalytic 5% Pd/C to give Intermediate (C2).

Mass Spectrum m/e 258.22 (M+H) + Preparation of (C3) Intermediate (C2) was converted to the carboxylic acid by hydrolysis with 2M aqueous sodium hydroxide solution in ethanol for 16hr at room temperature to give Intermediate (C3).

Mass Spectrum m/e 230.12 (M+H) + Example 1 Preparation of Compound 1 in Table 1 Step 1 Intermediate (A6) from Preparation 1 (Preparation of 6) above (10.28g, 0.030moles) was mixed with 4- (2-methoxyphenoxy) aniline (7.74g, 0.036moles), prepared as described in Rev.

Chim. (Bucharest) (1988), 39 (6), 477-482, in 1-propanol (170ml) and the mixture was stirred

and heated at 115°C for 5 hours. The mixture was cooled to ambient temperature and then filtered. The crystals were washed with a small volume of 1-propanol and then dried to give 4- (4- (2-methoxyphenoxy)-anilino)-3-cyano-6-methoxy-7- (benzyloxy) quinoline, Mass Spectrum m/e 504 (M++H).

NMR Spectrum (d-6-DMSO, 5 values) 3.73 (s, 3H), 3.97 (s, 3H), 5.32 (s, 2H), 6.95 (m, 3H), 7.05 (d, 1H), 7.18 (m, 2H), 7.38 (m, 5H), 7.51 (d, 2H), 7.58 (s, 1H), 8.17 (s, 1H), 8.87 (s, 1H), 11.13 (broad, 1H).

Step 2 The product from step (1) (7.2g, 14.3mmole) trifluoroacetic acid (40ml) and thioanisole (8.38ml, 71. 5mmole) was refluxed in a nitrogen atmosphere for 3 hours. After cooling the trifluoroacetic acid was removed by rotary evaporation and the oily residue was stirred with ice and water and basified with aqueous ammonia (S. G. 0.880). The resulting suspension was filtered and the solid was washed successively with water, ethyl acetate and diethyl ether and then dried to give 4-(4-(2-methoxyphenoxy)-anilino)-3-cyano-6-methoxy-7-hydroxy quinoline.

Mass Spectrum m/e 414 (M++H).

NMR Spectrum (d-6-DMSO, 8 values) 3.75 (s, 3H), 3.91 (s, 3H), 6.89 (d, 2H), 6.94 (m, 1H), 7.02 (d, 1H), 7.16 (m, 3H), 7.23 (m, 1H), 7.73 (s, 1H), 8.31 (s, 1H), 9.33 (s, 1H), 10.31 (broad, 1H).

Step 3 The product of step 2 (206.5mg, 0. 5mmole) and 1-chloro-3-bromopropane (95mg, 0.6mmole) was dissolved in dimethylsulphoxide (5ml) in the presence of potassium butoxide (0. 55ml, 1. OM in THF, 0. 55mmole) in and the mixture held at room temperature for 18 hours. Water was added and the mixture extracted with ethyl acetate. The combined extracts were washed with water and brine then dried (MgS04) and evaporated to give 4- (4- (2-methoxyphenoxy)- anilino)-3-cyano-6-methoxy-7- (3-chloropropoxy) quinoline, intermediate 1, (189mg, 77%) as a yellow gum.

Mass Spectrum m/e 490 (M>+H) Step 4 A mixture of4- (4- (2-methoxyphenoxy)-anilino)-3-cyano-6-methoxy-7- (3- chloropropoxy) quinoline, intermediate 1, an excess (5 molar equivalents) of 2- (hydroxymethyl) piperidine and sodium iodide (1 molar equivalent) were heated together in DMA at 60°C for 18hours. The reaction mixture was evaporated and the residue partitioned

between water and ethyl acetate. The combined organic extracts were washed with water and brine then dried (MgS04) and evaporated. The residue was chromatographed eluting with a gradient of dichloromethane and dichloromethane containing 10% of methylamine (33% in ethanol) solution from 0% to 100%. Fractions containing product were evaporated.

Compound No. 1 was obtained as the hydrochloride salt by treatment of the residue in ethanol with 1. OM hydrogen chloride (2 molar equivalents) in ether.

Mass Spectrum m/e 569 (M++H).

Example 2 Preparation of Compound 2 in Table 1 ole f [Y°TS N S t/eN '0 N I \ \ N "0 N-HCI salt Cl~O N Ho Ho (1) (2) A mixture of intermediate 1, Example 1, Step 3, excess of 4-hydroxypiperidine (44 molar equivalents) and sodium iodide (1 molar equivalent in DMA) were heated, without solvent, together at 60°C for 20hours. The reaction mixture was evaporated and the residue partitioned between water and ethyl acetate. The combined organic extracts were washed with water and brine then dried (MgS04) and evaporated. The residue was chromatographed.

Fractions containing product were evaporated. Compound No. 2 was obtained as the hydrochloride salt by treatment of the residue in ethanol with 1. OM hydrogen chloride (2 molar equivalents) in ether.

Mass Spectrum m/e 555 (MF+H).

NMR Spectrum (d-6-DMSO, 8 values) 1.73 (m, 2H), 1.97 (m, 2H), 2.33 (m, 2H), 2.96 (m, 1H), 3.19 (m, 2H), 3.31 (m, 2H), 3.47 (m, 2H), 3.73 (s, 3H), 3.99 (s, 3H), 4.26 (t, 2H), 6.96 (m, 3H), 7.05 (d, 1H), 7.19 (m, 2H), 7.39 (d, 2H), 7.56 (s, 1H), 8.23 (s, 1H), 8.88 (s, 1H), 11.22 (broad, 1H) Example 3 Preparation of Compound 3 in Table 1 0-" No t \ I O N I N "C)-o-6 N' (a b- ----N (i) (2) A mixture of intermediate 1, Example 1, Step 3, and excess of 4-hydroxyethanol (25 molar equivalents) were heated in n-propanol solution at 60°C for 72hours. The reaction mixture was worked up as described in Example l, Step 4 and Compound No. 3 was obtained as the hydrochloride salt.

Mass Spectrum m/e 515 (M++H).

NMR Spectrum (d-6-DMSO, 8 values) 2.25 (m, 2H), 3.03 (m, 2H), 3.12 (m, 2H), 3.68 (m, 2H), 3.73 (s, 3H), 3.97 (s, 3H), 4.28 (t, 2H), 6.96 (m, 3H), 7.05 (d, 1H), 7.18 (m, 2H), 7.39 (d, 2H), 7.55 (s, 1H), 8.22 (s, 1H), 8.88 (s, 1H), 8.96 (broad, 1H), 11.20 (broad, 1H) Example 4 Preparation of Compound 4 in Table 1 ou 0, 1 I o f i N w I I i oxo ou N C, Cl~o tN +-tN HCl salt oWo ---i (1) (2) By using the procedure described in Example 1, Step 4, but using 2-hydroxymethylpyrrolidine in place of 2-hydroxymetliylpiperidine, the title compound was obtained after chromatographic purification.

Mass Spectrum m/e 555 (M++H).

Example 5 Preparation of Compound 5 in Table 1 oll o . 0 ? N " i0 w N I \N T HON N0 I ri HCI Salt (1) (2) (D

By using the procedure described in Example 1, Step 4, but using 4-hydroxymethylpiperidine in place of 2-hydroxymethylpiperidine, the title compound was obtained after chromatographic purification.

Mass Spectrum m/e 569 (M++H).

Example 6 Preparation of Compound 6 in Table 1 ole zozo N t, 0 N Ho i NONO N HCI Salt ci---'o N I (1) (2)

By using the procedure described in Example 1, Step 4, but using N-methyl-2- hydroxyethylamine in place of 2-hydroxymethylpiperidine, the title compound was obtained after chromatographic purification.

Mass Spectrum m/e 529 (M++H).

Example 7 Preparation of Compound 7 in Table 1 OLE 0 0 w 11"N., r 0) 1 (-,)-- 'T I I HO'-'o N (1) (2)

By using the procedure described in Example 1, Step 4, but using 3-hydroxymethylpiperidine in place of 2-hydroxymethylpiperidine, the title compound was obtained after chromatographic purification.

Mass Spectrum m/e 569 (M++H).

Example 8 Preparation of Compound 8 in Table 1 0 zozo NEZ ^ \ \N i \N + H07VN , r I' I N HCl Salt Cl~ot HO HoC N HCI salt HO

By using the procedure described in Example 1, Step 4, but using 4,4-dihydroxypiperidine in place of 2-hydroxymethylpiperidine, and including 5 molar equivalents of potassium carbonate in the reaction mixture, the title compound was obtained after chromatographic purification.

Mass Spectrum m/e 571 (M++H).

NMR Spectrum (d-6-DMSO, 8 values) 2.39 (m, 2H), 2.98 (m, 2H), 3.39 (broad, 8H), 3.76 (s, 3H), 4.09 (s, 3H), 4.35 (t, 2H), 6.99 (m, 3H), 7.07 (m, 1H), 7.22 (m, 2H), 7.41 (d, 2H), 7.53 (s, 1H), 8.17 (s, 1H), 8.89 (s, 1H), 11.00 (broad, 1H), 11.49 (broad, 1H) Example 9 Preparation of Compound 9 in Table 1 o o I o I i N I I I Zozo N NON HO Np I N HCI Salt T ci"-, 0 N (1) (2)

By using the procedure described in Example 1, Step 4, but using 3-hydroxypyrrolidine in place of 2-hydroxymethylpiperidine, the title compound was obtained after chromatographic purification.

Mass Spectrum m/e 541 (M+H).

Example 10 Preparation of Compound 10 in Table 1 This compound was prepared using the following scheme.

Step 1 Intermediate (1) was obtained by reaction of benzylcliloroformate (25.7ml, 0. 18mol, 1.2 equiv.) and 2-methoxy-5-nitroaniline (25g, 1. Oequiv.) in pyridine (190m1) at room temperature for 18 hours.

NMR Spectrum (d-6-DMSO, 8 values) 3.93 (s, 3H), 5.18 (s, 2H), 7.21 (d, 1H), 7.37 (m, 5H), 7.98 (m, 1H), 8. 69 (m, 1H), 9.10 (s, 1H).

Step 2 Intermediate (2) was prepared by treating intermediate (1) (28.7g, 94. 97mmol, 1.0 equiv.) in ethyl acetate (550ml) with SnCl2. 2H20 (85. 7g, 0.38mol, 4 equiv.) at 105°C for 5hrs. After this, ethoxymethylenecyanoacetate (16. 0g, 95mmol, 1.0 equiv.) in ethanol (200m1) was added and the mixture held at 90°C for 90mins. The mixture was cooled, and Intermediate (2) was isolated by filtration, washed with ethanol and dried under vacuum.

NMR Spectrum (d-6-DMSO, 8 values) (-3 : 1 mixture of isomers) 1.22 (m, 3H), 3.76 (s, 3H), 4.16 (m, 2H), 5.13 (s, 2H), 7.02 (m, 2H), 7.35 (m 5H), 7.72 (m, 0.25H), 7.80 (m, 0.75H), 8.14 (bs, 0.75H), 8.26 (d, 0.25H), 8.66 (s, 0.25H), 8.71 (s, 0.75H), 10.65 (d, 0.25H), 10.78 (bs, 0.75H).

Step 3 Intermediate (2) was converted to Intermediate (3) in the above scheme by contact with Dowthenn A for 4 hours at 250-260°C.

Mass Spectrum m/e 350 (M+H) +.

Step 4 The product of Step 3 was chlorinated using POC13 in MeCN at 110°C for 2hrs. Intermediate (4) in the above scheme was obtained.

Mass Spectrum m/e 368,370 (M+H) +.

Step 5 4- (2-N-methylcarboxamidomethoxy-phenoxy) nitrobenzene, obtained by reacting 4- fluoronitrobenzene with (2-N-methylcarboxamidomethoxy) phenol for 2.5 hours at 150°C in DMA in the presence of potassium butoxide, was dissolved in ethyl acetate and hydrogenated for 18 hours in the presence of a 10% palladium/carbon catalyst. The catalyst was filtered, the solution concentrated in vacuo, and upon standing 4- (2-N-methylcarboxamidomethoxy- phenoxy) aniline, intermediate 5, was obtained as crystals.

Mass Spectrum m/e 273 (M+H) + Step 6 Intermediate 5 was reacted with intermediate 4, in n-propanol solution at 100°C for 90mins.

Intermediate 6 was obtained.

Mass Spectrum m/e 604 (M+H) +.

NMR Spectrum (d-6-DMSO, 8 values) 2.60 (d, 3H), 4.00 (s, 3H), 4.47 (2H, s), 5.24 (s, 2H), 7.03 (m, 5H), 7.12 (m, 2H), 7.37 (m, 5H), 7.46 (m, 2H), 7.53 (m, 1H), 8.13 (s, 1H), 8.58 (s, 1H), 8.79 (s, 1H), 9.42 (s, 1H).

Step 7 Intermediate 6 was converted to intermediate 7 by treatment with 33% HBr in AcOH in the presence ofthioanisole at 0°C to RT for 3hr. After chromatographic purification, intermediate 7 was obtained.

Mass Spectrum m/e 470 (M+H) +.

Step 8 A solution of intermediate 7 in DCM was coupled with N-piperidine-3-propionic acid using 1- (3-dimethylaminopropyl)-3-ethylcarbodiimide (EDAC) and dimethylaminopyridine (DMAP) at room temperature for 6 days to yield, after chromatographic purification, the title compound.

Mass Spectrum m/e 609 (M+H) +.

NMR Spectrum (d-6-DMSO, 8 values) 1.74 (m, 6H), 2.62 (d, 3H), 2.89 (m, 2H), 3.14 (m, 4H), 4.08 (s, 3H), 4.53 (2H, s), 7.05 (m, 5H), 7.13 (m, 2H), 7.42 (d, 2H), 7.55 (m, 1H), 8.24 (s, 1H), 8.85 (s, 1H), 8.89 (s, 1H).

Example 11 Preparation of Compound 11 in Table 1 N O'Y Ns HN, la e 1,, N i o o i o o i HAN hiN HN \ \, N N N HzNvN a~N4N OJ (2) Erample i 1

Step 1 Intermediate 1, Example 10, Step 7, was converted to intermediate 2 by reaction with 1- bromo-3-chloropropane by a procedure analogous to that described in Preparation of Key Intermediates, Preparation of (8). Intermediate 2 was obtained.

Mass Spectrum m/e 546,548 (M+H) +.

Step 2 Intermediate 2 was reacted with morpholine in the presence of sodium iodide at room temperature for 9 days to give, after chromatographic purification, the title compound Mass Spectrum m/e 595 (M+H) +.

NMR Spectrum (d-6-DMSO, 8 values) 2.08 (m, 2H), 2.60 (d, 3H), 3.04 (m, 2H), 3.16 (m, 2H), 3.40 (m, 2H), 3.80 (m, 2H), 3.92 (m, 2H), 4.00 (s, 3H), 4.46 (2H, s), 6.95 (1H, s), 7.04 (m, 5H), 7.16 (m, 2H), 7.39 (d, 2H), 7.54 (m, 1H), 7.92 (s, 1H), 8.80 (s, 1H).

Example 12 Preparation of Compound 12 in Table 1 Q nu 0 I I I HN \ I'/- 0 sN ClA G N N N 0 ZIZI , a-,- ,,) (1) (Example 12)

Intermediate 1, Example 11, Step 1, was converted to the title compound by a process analogous to that described in Example 11, Step 2, using piperidine in place of morpholine.

The title compound was obtained after chromatographic purification.

Mass Spectrum m/e 593 (M+H) +.

NMR Spectrum (d-6-DMSO, 8 values) 1.37 (m, 1H), 1.76 (m, 5H), 2.06 (m, 2H), 2.62 (d, 3H), 2.84 (m, 2H), 3.07 (m, 2H), 3.39 (m, 2H), 4. 01 (s, 3H), 4.33 (2H, s), 7.00 (2H, m), 7.08 (m, 4H), 7.16 (m, 2H), 7.39 (d, 2H), 7.55 (m, 1H), 7.96 (s, 1H), 8.79 (s, 1H).

Example 13 Preparation of Compound 13 in Table 1 ou N 1 : 1 /O O v 11 HN I I/ Zu i i N N N HzN/N 0 (1) (Example 12) Intermediate 1 (Example 10, Step 7) was converted to the title product by a process analogous to that of Example 10, Step 8, but using N-morpholine-3-propionic acid in place of N- piperidine-3-propionic acid. After chromatographic purification, the title compound was obtained.

Mass Spectrum m/e 611 (M+H) +.

NMR Spectrum (d-6-DMSO + d-4-AcOH, 8 values) 2.62 (s, 3H), 3.14 (m, 4H), 3.42 (m, 4H), 4.08 (s, 3H), 4.45 (2H, s), 7.04 (m, 5H), 7.13 (m, 2H), 7.42 (d, 2H), 8.21 (m, 1H), 8.89 (s, 1H).

Example 14 Preparation of Compound 14 in Table 1 Oi Oi Oi oto voto voto N, co I N 'N O O igO I 0 N F F 0 !" O' F 0 N I I N N I I , N E ° C N- p NW OvN 0 0 o (Example 14) (4) Step 1 Intermediate 1 (Example 1, Step2) was reacted in DCM solution with triflic anhydride in the presence of lutidine and DMAP to give intermediate 2.

Mass Spectrum m/e 546 (M++H).

Step 2 Intermediate 2 was reacted in DMF/methanol solution with carbon monoxide in the presence of palladium acetate, 1, 3-bis (diphenylphosphino) propane and triethylamine at 70°C to give intermediate 3.

Mass Spectrum m/e 456 (M++H).

Step 3 Intermediate 3 was hydrolysed with lithium hydroxide in a mixture of methanol, THF and water to give intermediate 4.

Mass Spectrum m/e 442 (M++H).

Step 4 Intermediate 4 was coupled with N-2-aminoethylmorpholine in DMF solution using HATU to give, after chromatographic purification on silica using as eluant 0-4% methanol in DCM, the title compound.

Mass Spectrum m/e 554 (M++H).

NMR Spectrum (d-6-DMSO, 6 values) 2.30-2.60 (m, 4H), 2.87-2.95 (m, 1H), 3.15-3.30 (m, 1H), 3.37-3.70 (m, 6H), 3.73 (s, 3H), 4.00 (s, 3H), 6.94 (m, 3H), 7.02 (m, 1H), 7.17 (m, 2H), 7.29 (m, 2H), 7.91 (s, 1H), 8.17 (s, 1H). 1H), 8.43 (s, 1H), 8.48 (br. s, 1H), 9.70 (s, 1H).

Example 15 Preparation of Compound 15 in Table 1 ole N JS I1 1-1 N tN, N'or 6 O Cl~OJ% NJ oS N o5 NO4NJ HCI salt oWo''%'NJ (1) (2) (Example 15)

By using the procedure described in Example 1, Step 4, but using 2-methylthioethylamine in place of 2-hydroxymethylpiperidine, the title compound was obtained after chromatographic purification.

Mass Spectrum m/e 545 (M++H).

Example 16 Preparation of Compound 16 in Table 1 oll o'i ilolw Nwl li --6 Na b Cl~OtN + CN > CN~O N HCI salt R (1) (2) (Example 16)

By using the procedure described in Example 2, but using thiomopholine in place of 4- hydroxypiperidine, the title compound was obtained after chromatographic purification.

Mass Spectrum m/e 557 (M++H).

NMR Spectrum (d-6-DMSO, 8 values) 2.32 (m, 2H), 2.83 (d, 2H), 3.21 (m, 6H), 3.74 (s, 3H), 3.78 (m, 2H), 3.99 (s, 3H), 4.29 (t, 2H), 6.97 (m, 3H), 7.05 (d, 1H), 7.19 (m, 2H), 7.39 (d, 2H), 7.51 (s, 1H), 8. 16 (s, 1H), 8.88 (s, 1H), 11.00 (broad, 1H) Example 17 Preparation of Compound 17 in Table 1 > Ns ow \ fT°Y° fY° 0-6 1, 0 1 1 1 I \ I \ \ u IIN I-N I (Example 17) Step 1 4- (2-N-methylcarboxamidomethoxy-phenoxy) aniline (Example 10, Step 5) was reacted with 4-chloro-3-cyano-6-inethoxy-7- (3-chloropropoxy) quinoline (Preparation of Key Intermediates, Preparation of (8)) in 1-propanol and the mixture was stirred and heated at 100°C for 18 hours. The mixture was cooled to ambient temperature and then filtered. The crystals were washed with a small volume of 1-propanol and then dried to give 4- (4- (2-N- methylcarboxamidomethoxy-phenoxy)-anilino)-3-cyano-6-methoxy -7- (3- chloropropoxy) quinoline, intermediate 1.

Mass Spectrum m/e 547.5 (M+H) +.

Step 2 Intermediate 1 was reacted with cyclopentylamine at 100°C for 3hr to give, after chromatographic purification, the title compound.

Mass Spectrum m/e 596 (M+H) +.

NMR Spectrum (d-6-DMSO + d-4-AcOH, 8 values) 1.62 (m, 6H), 1.95 (m, 2H), 2.21 (m, 2H), 2.60 (s, 3H), 3.08 (m, 2H), 3.50 (m, 2H), 3.99 (s, 3H), 4.28 (m, 2H), 4.45 (s, 2H), 7.04 (d, 2H), 7.10 (m, 4H), 7.40 (d, 2H), 7.47 (s, 1H), 8. 12 (s, 1H), 8.87 (s, 1H).

Example 18 Preparation of Compound No. 18 in Table 2 Step 1 Intermediate (A6) from Preparation A above (10.28g, 0.030moles) was mixed with 4- (2- methoxyphenoxy) aniline (7.74g, 0.036moles), prepared as described in Rev. Chim.

(Bucharest) (1988), 39 (6), 477-482, in 1-propanol (170ml) and the mixture was stirred and heated at 115°C for 5 hours. The mixture was cooled to ambient temperature and then filtered. The crystals were washed with a small volume of 1-propanol and then dried to give 4- (4- (2-methoxyphenoxy)-anilino)-3-cyano-6-methoxy-7- (benzyloxy) quinoline,

Mass Spectrum m/e 504 (M++H).

NMR Spectrum (d-6-DMSO, 8 values) 3.73 (s, 3H), 3.97 (s, 3H), 5.32 (s, 2H), 6.95 (m, 3H), 7.05 (d, 1H), 7.18 (m, 2H), 7.38 (m, 5H), 7.51 (d, 2H), 7.58 (s, 1H), 8.17 (s, 1H), 8.87 (s, 1H), 11.13 (broad, 1H).

Step 2 The product from step (1) (7.2g, 14.3mmole) trifluoroacetic acid (40ml) and thioanisole (8.38ml, 71. 5mmole) was refluxed in a nitrogen atmosphere for 3 hours. After cooling the trifluoroacetic acid was removed by rotary evaporation and the oily residue was stirred with ice and water and basified with aqueous ammonia (S. G. 0.880). The resulting suspension was filtered and the solid was washed successively with water, ethyl acetate and diethyl ether and then dried to give 4- (4- (2-methoxyphenoxy)-anilino)-3-cyano-6-methoxy-7-hydroxyquino line.

Mass Spectrum m/e 414 (M++H).

NMR Spectrum (d-6-DMSO, 8 values) 3.75 (s, 3H), 3.91 (s, 3H), 6.89 (d, 2H), 6.94 (m, 1H), 7.02 (d, 1H), 7.16 (m, 3H), 7.23 (m, 1H), 7.73 (s, 1H), 8.31 (s, 1H), 9.33 (s, 1H), 10. 31 (broad, 1H).

Step 3 The product of step 2 (206.5mg, 0. 5mmole) and 1-chloro-3-bromopropane (95mg, 0.6mmole) was dissolved in dimethylsulphoxide (5ml) in the presence of potassium butoxide (0. 55ml, 1. OM in THF, 0. 55mmole) in and the mixture held at room temperature for 18 hours. Water was added and the mixture extracted with ethyl acetate. The combined extracts were washed with water and brine then dried (MgS04) and evaporated to give 4- (4- (2-methoxyphenoxy)- anilino)-3-cyano-6-methoxy-7- (3-chloropropoxy) quinoline (189mg, 77%) as a yellow gum.

Mass Spectrum m/e 490 (M++H) Step 4 A mixture of 4- (4- (2-methoxyphenoxy)-anilino)-3-cyano-6-methoxy-7- (3- chloropropoxy) quinoline, an excess (44 molar equivalents) of N- (2-methoxyethyl) piperidine (available commercially) and sodium iodide (1 molar equivalent) were heated together at 50°C for 20hours. The reaction mixture was evaporated and the residue partitioned between water and ethyl acetate. The combined organic extracts were washed with water and brine then dried (MgS04) and evaporated. The residue was chromatographed on a Bond Elut (lOg) cartridge eluting with a gradient of dichloromethane and dichloromethane containing 10% of methylamine (33% in ethanol) solution from 0% to 100%. Fractions containing product were

evaporated. Compound No. 1 was obtained as the hydrochloride salt by treatment of the residue in ethanol with 1. OM hydrogen chloride (2 molar equivalents) in ether.

Mass Spectrum m/e 598 (M++H).

NMR Spectrum (d-6-DMSO, 8 values) 2.33 (m, 2H), 3.28 (s, 3H), 3.36 (m, 4H), 3.51 (m, 6H), 3.71 (m, 4H), 3.73 (s, 3H), 3.99 (s, 3H), 4.31 (t, 2H), 6.96 (m, 3H), 7.04 (d, 1H), 7.18 (m, 2H), 7.39 (d, 2H), 7.56 (s, 1H), 8.26 (s, 1H), 8.89 (s, 1H), 11.28 (broad, 1H) Example 19 Preparation of Compound No. 19 in Table 2 Step 1 4-(2-N-methylcarboxamidomethoxy-phenoxy) nitrobenzene, obtained by reacting 4- fluoronitrobenzene with (2-N-methylcarboxamidomethoxy) phenol for 2.5 hours at 150°C in DMA in the presence of potassium butoxide, was dissolved in ethyl acetate and hydrogenated for 18 hours in the presence of a 10% palladium/carbon catalyst. The catalyst was filtered, the solution concentrated in vacuo, and upon standing 4- (2-N-methylcarboxamidomethoxy- phenoxy) aminobenzene was obtained as crystals.

Mass Spectrum m/e 273 (M+H) + Step 2 Intermediate (A8) from Preparation A above (0.60g, 2.3mmol) was mixed with the product of step 1 (0.68g, 2.5mmol) in 1-propanol and the mixture was stirred and heated at 100°C for 18 hours. The mixture was cooled to ambient temperature and then filtered. The crystals were washed with a small volume of 1-propanol and then dried to give 4- (4- (2-N- methylcarboxamidomethoxy-phenoxy)-anilino)-3-cyano-6-methoxy -7- (3- chloropropoxy) quinoline.

Mass Spectrum m/e 547.5 (M+H) + Step 3 The product from step 2 (0.2g, 0.37mmol) was mixed with cyclopropylamine (5ml) and stirred for 4 days at room temperature. The mixture was concentrated in vacuo and the residue washed with ether then extracted with DCM/water (3x). The combined DCM extracts were dried, evaporated and the residue chromatographed on silica gel using 10% methanol in DCM containing 0.5% 0.880 aqueous ammonia solution. Fractions containing the product

were evaporated and the residue treated with ethereal hydrogen chloride and Compound 19 was isolated as the hydrochloride salt (0.16g, 79%).

Mass Spectrum m/e 568 (M+H) +.

NMR Spectrum (d-6-DMSO + d-4-AcOH, 8 values) 0.76 (m, 2H), 0.91 (m, 2H), 2.21 (m, 2H), 2.60 (s, 3H), 2.74 (m, 1H), 3.17 (m, 2H), 3.95 (s, 3H), 4.28 (m, 2H), 4.45 (s, 2H), 7.02 (m, 6H), 7.37 (d, 2H), 7.43 (s, 1H), 8.04 (s, 1H), 8.74 (s, 1H).

Example 20 Preparation of Compound No 20 in Table 2 Step 1 Using processes analogous to that described for the preparation of Intermediate (A7) above but using 3-methoxy-4-benzyloxy-aniline instead of starting material (Al), 3-cyano-4-chloro- 6-hydroxy-7-methoxyquinoline was obtained.

Mass spectrum m/e 234 (M++H).

Step 2 The product from step 1 (lg) was reacted with propargyl bromide (0.95g) in the presence of potassium butoxide (0.53g), tetrabutylammonium iodide (0. 16g) and 18C-6 crown ether (0. 05g), in solution in DMA (100ml, 10°C to ambient temperature). 3-cyano-4-chloro-6- propargyloxy-7-methoxyquinoline (0.73g) was obtained.

Mass Spectrum m/e 273 (M++H).

Step 3 4-nitro-fluorobenzene and 2- (carboxymethoxy) phenol were reacted together in DMA in the presence of potassium butoxide for 2hours at 150°C to yield 4- (2-carboxymethoxy- phenoxy) nitrobenzene.

Mass Spectrum m/e 288 (M-H+)- Step 4 A solution of the product of step 3 in DMA was then coupled with methyl glycine using 1- (3- dimethylaminopropyl)-3-ethylcarbodiimide (EDAC), dimethylaminopyridine (DMAP) and N- hydroxybenzotriazole (HOBT) to yield the intermediate of formula

Mass Spectrum m/e 361.17 (M++H).

Step 5 Reduction of the product of step 4 to the corresponding amine, was effected by hydrogenation in the presence of a 5% Pd/C catalyst.

Mass ectrum m/e 331.14 (M++H).

Step 6 Reacting the product of step 5 in methanol with cyclopropylamine at room temperature for 3 hours resulted in the production of the intermediate of formula Mass Spectrum m/e 356. 23 (M++H).

Step 7 The products of steps 2 and 6 were reacted together using reaction conditions analogous to those described in Example 18 step 1, but at a temperature of 100°C for 2hours, Compound No. 20 was obtained.

NMR Spectrum (d-6-DMSO, 8 values) 0.00-0.08 (m, 2H), 0.20-0.30 (m, 2H), 2.19-2.32 (m, 1H), 3.30 (s, 2H), 3.32 (t, 1H), 3.64 (s, 3H), 4.19 (s, 2H), 4.67 (s, 2H), 6.60-6.75 (m, 4H), 6.75-6.88 (m, 2H), 7.07 (d, 2H), 7.10 (s, 1H), 7.50 (t, 1H), 7.60 (d, 1H), 7.86 (s, 1H), 8.55 (s, 1H), 10.60-10.70 (br. s, 1H)..

Example 21 Preparation of Compound No. 21 in Table 2 4- (4- (2-methoxyphenoxy)-anilino)-3-cyano-6-methoxy-7- (3-chloropropoxy) quinoline (245mg, 0. 5mmole) prepared as described in Example 18 step 3, was dissolved in 1-propanol with an excess (25 molar equivalents) of cyclopropylamine and heated at 60°C for 72 hours.

The reaction mixture was evaporated and the residue was chromatographed on a Bond Elut (10g) cartridge eluting with a gradient of dichloromethane and dichloromethane containing 10% of methylamine (33% in ethanol) solution from 0% to 100%. Fractions containing product were evaporated and Compound No. 21 was obtained as the hydrochloride salt by treatment of the residue in ethanol with 1. OM hydrogen chloride (2 molar equivalents) in ether.

Mass Spectrum m/e 511 (M++H).

NMR Spectrum (d-6-DMSO, d values) 0.76 (m, 2H), 0.91 (m, 2H), 2.25 (m, 2H), 2.75 (m, 1H), 3.18 (m, 2H), 3.74 (s, 3H), 3.99 (s, 3H), 4.31 (t, 2H), 6.97 (m, 3H), 7.05 (m, 1H), 7.19 (m, 2H), 7.39 (d, 2H), 7.49 (s, 1H), 8.15 (s, 1H), 8.87 (s, 1H), 9.26 (broad, 2H), 11.00 (broad, 1H) Example 22 Preparation of Compound No. 22 in Table 2 This compound was prepared using the following scheme. o 0 po l : J / JN N i O N N-I O N N mO N (2) (3) cri O p O % N _/I I '. " O'N N vif/ HN'ao Example22 (5) Step 1 Intermediate (1) was obtained by reaction of benzylchloroformate (25.7ml, 0. 18mol, 1.2 equiv.) and 2-methoxy-5-nitroaniline (25g, l. Oequiv.) in pyridine (190m1) at room temperature for 18 hours.

NMR Spectrum (d-6-DMSO, 8 values) 3.93 (s, 3H), 5.18 (s, 2H), 7.21 (d, 1H), 7.37 (m, 5H), 7.98 (m, 1H), 8.69 (m, 1H), 9.10 (s, 1H).

Step 2 Intermediate (2) was prepared by treating starting material (1) (28.7g, 94.97mmol, 1.0 equiv.) in ethyl acetate (550ml) with SnC12. 2H20 (85. 7g, 0.38mol, 4 equiv.) at 105°C for 5hrs. After this, ethoxymethylenecyanoacetate (16. 0g, 95mmol, 1.0 equiv.) in ethanol (200ml) was added and the mixture held at 90°C for 90mins. The mixture was cooled, and Intermediate (2) was isolated by filtration, washed with ethanol and dried under vacuum.

NMR Spectrum (d-6-DMSO, 5 values) (-3 : 1 mixture of isomers) 1.22 (m, 3H), 3.76 (s, 3H), 4.16 (m, 2H), 5.13 (s, 2H), 7.02 (m, 2H), 7.35 (m 5H), 7.72 (m, 0.25H), 7.80 (m, 0.75H), 8. 14 (bs, 0.75H), 8.26 (d, 0.25H), 8.66 (s, 0.25H), 8.71 (s, 0.75H), 10.65 (d, 0. 25H), 10. 78 (bs, 0.75H).

Step 3 Intermediate (2) was converted to Intermediate (3) in the above scheme by contact with Dowtherm A for 4 hours at 250-260°C.

Mass Spectrum m/e 350 (M+H) +.

Step 4 The product of Step 3 was halogenated using POC13 in MeCN at 110°C for 2hrs. Intermediate (4) in the above scheme was obtained.

Mass Spectrum m/e 368,370 (M+H) +.

Step 5 Intermediate (5) prepared as described in Example 19 step 1 was reacted with the product of step 4, using conditions analogous to those described in Example 18 step 1 but using temperatures of 100°C for 90mins. The title compound was obtained.

Mass Spectrum m/e 604 (M+H) +.

NMR Spectrum (d-6-DMSO, 8 values) 2.60 (d, 3H), 4.00 (s, 3H), 4.47 (2H, s), 5.24 (s, 2H), 7.03 (m, 5H), 7.12 (m, 2H), 7.37 (m, 5H), 7.46 (m, 2H), 7.53 (m, 1H), 8.13 (s, 1H), 8.58 (s, 1H), 8.79 (s, 1H), 9.42 (s, 1H).

Example 23 Preparation of Compound No. 23 in Table 2 4- (4- (2-methoxyphenoxy)-anilino)-3-cyano-6-metlioxy-7- (3-chloropropoxy) quinoline (245mg, 0. 5mmole) prepared as described in Example 18 step 3, was dissolved in dimethylacetamide (5ml) with an excess (5 molar equivalents) of hexamethyleneimine and sodium iodide (1 molar equivalent) and heated at 60°C for 18 hours. At the end of this period, the reaction mixture was evaporated and the residue partitioned between water (5ml) and ethyl acetate (1 Oml). The aqueous phase was extracted with a further portion of ethyl acetate (3ml) and the combined organic extracts were washed with water and brine then dried (MgS04) and evaporated. Compound 6 was purified by Bond Elut chromatography and converted to the hydrochloride salt by the procedure described in Example 21.

Mass Spectrum m/e 553 (M++H).

Example 24 Preparation of Compound No. 24 in Table 2 Using a method analogous to that described in Example 23 but with N-acetylpiperazine as the amine, the title compound was obtained.

Mass Spectrum m/e 583 (M++H).

Example 25 Preparation of Compound No. 25 in Table 2 Step 1 A solution of 4- (4- (2-methoxyphenoxy)-anilino)-3-cyano-6-methoxy-7-hydroxyquino line (3.6g, 8. Ommole), prepared as described in Example 18 step 2, in DMSO (40ml) was reacted with (2S)- (+)-glycidyl tosylate (2.3g, 1 Ommole) in the presence of potassium carbonate (2.8g, 20mmole) at room temperature for 18 hours. 4- (4- (2-methoxyphenoxy)-anilino)-3-cyano-6- methoxy-7-glycidyloxy quinoline was obtained.

Mass Spectrum m/e 470 (M++H) Step 2 The product from step 1 together with 20 molar equivalents of morpholine and 0.5 molar equivalents NaI in dimethyl acetamide were heated to 60°C for 18hours to yield the title compound.

Mass Spectrum m/e 557 (M++H).

NMR Spectrum (d-6-DMSO, 8 values at 373K) 3.37 (m, 6H), 3.80 (s, 3H), 3.96 (m, 4H), 4.02 (s, 3H), 4.28 (m, 2H), 4.57 (m, 1H), 6.99 (m, 3H), 7.06 (d, 1H), 7.19 (m, 2H), 7.34 (d, 2H), 7.59 (s, 1H), 8. 06 (s, 1H), 8. 63 (s, 1H), 10.28 (broad, 1H) Example 26 Preparation of Compound No. 26 in Table 2 4- (4- (2-methoxyphenoxy)-anilino)-3-cyano-6-methoxy-7- (3-chloropropoxy) quinoline (107.5mg, 0.22mmole), prepared as described in Example 18 step 3 was reacted with 4- amidopiperidine (44 molar equivalents) and 1 molar equivalent sodium iodide in N, N- dimethylacetamide (3ml). The mixture was heated to 50°C for 20 hours after which the title compound was isolated as the hydrochloride salt using the work up and salt formation procedure described in Example 18, step 4.

Mass Spectrum m/e 582 (M++H).

NMR Spectrum (d-6-DMSO, 8 values) 2.00 (m, 4H), 2.33 (m, 2H), 3.10 (m, 2H), 3.30 (t, 2H), 3.55 (m, 2H), 3.77 (s, 3H), 3.98 (s, 3H), 4.33 (t, 2H), 6.97 (m, 3H), 7.03 (d, 1H), 7.15 (m, 2H), 7.32 (m, 2H), 7.54 (s, 1H), 8.19 (s, 1H), 8. 88 (s, 1H), 10.14 (broad, 1H), 11.07 (broad, 1H) Example 27 Preparation of Compound 27 in Table 2 Step 1 4-nitro-fluorobenzene and 2-(carboxymethoxy) phenol were reacted together in DMA in the presence of potassium butoxide for 2hours at 150°C to yield 4- (2- (carboxymethoxy) phenoxy) nitrobenzene.

Mass Spectrum m/e 288 (M-H+)- Step 2 Reduction of the product of step 1 and concomitant esterification was effected by hydrogenation in EtOAc/MeOH/lM HC1 at room temperature for 18 hours which gave the corresponding methyl ester, 4-(2-methoxycarbonylmethoxy) phenoxy) aniline.

Mass Spectrum m/e 274 (M+H Step 3 Reacting the product of step 2 in methanol with cyclopropylamine at room temperature for 3 hours resulted in the production of 4-(2-(cyclopropyl-amido-methoxy)-phenoxy) aniline of formula Mass Spectrum m/e 299.5 (M+H) + Step 4 Using a method analogous to that described in Example 20 but using the product of step (3) in place of the product of Step 6 of Example 20, the title compound was obtained.

NMR Spectrum (d-6-DMSO, 8 values) 0.00-0.08 (m, 2H), 0.17-0.28 (m, 2H), 2.20-2.30 (m, 1H), 3.26 (t, 1H), 3.60 (s, 3H), 4.05 (s, 2H), 4.64 (s, 2H), 6.55-6.71 (m, 5H), 6.71-6.81 (m, 1H), 7.03 (d, 2H), 7.09 (s, 1H), 7.40 (d, 1H), 7.88 (s, 1H), 8.53 (s, 1H), 10.71-10. 78 (br. s, 1H).

Example 28 Preparation of Compound 28 in Table 2 The compound in this instance was prepared using the following scheme: 0 0 0 0 0 0 \ OEt 0 I I OEt I I OEt (1) 12) (3) < \ () DS (3) 1-o N () 0 kOAA"OYY\-NH, ci 0 0 0 0 w I \ W W I I NHz HO I I NHZ 0", 0 NH2 HO :, :) ! NH, I-Y C (8) N , 0 N^"-'o N N CI, N' ci cl N'V 0 HO : 0 - (su J J (8) (9) (10) ?"dz HN o¢jw sV HN I I' Campound 28 N 0,-, J Step 1 Intermediate (1) was prepared by reacting together methylenedioxyaniline (51g, 0.37mol) and diethylethoxymethylenemalonate (75.1ml, 0.37mol) in ethanol at 80°C for lhour.

NMR Spectrum (d-6-DMSO, 8 values) 1.30 (t, 3H), 1.37 (t, 3H), 4.22 (q, 2H), 4.29 (q, 2H), 6.99 (s, 2H), 6.56 (m, 1H), 6.66 (d, 1H), 6.79 (d, 1H), 8.37 (d, 1H), 10.94 (bd, 1H).

Step 2 The product of step 1 was converted to Intermediate (2) above by contact with Dowtherm A for 1 hour at 250-260°C.

NMR Spectrum (d-6-DMSO, $# values) 1.24 (t, 3H), 4.17 (q, 2H), 6.13 (s, 2H), 7.03 (s, 1H), 7.43 (s, 1H), 8.49 (s, 1H).

Step 3 Reaction of the product of step (2) (20.3g,) with 4-methoxybenzylchloride (11. 9ml, 81.6mmol) in DMA (200ml) in the presence of potassium carbonate for 3 hours at 100°C yielded Intermediate 3 Mass Spectrum m/e 382 (M+H) +.

Step 4 The product of step 3 mixed with formamide (91.8mmol, 3.5equiv.) in DMA was heated at 60°C. Sodium methoxide (25% in methanol, 3.9ml, 0.65 equiv.) added and heating of the mixture continued for 90mins to yield Intermediate 4 in the above scheme.

Mass Spectrum m/e 353 (M+H) +.

Step 5 The product of step 4 was mixed with 2-hydroxyethylmorpholine, potassium (tert)-butoxide and DMA and the mixture heated at 120°C for 25mins. Intermediate (5) was obtained.

Mass Spectrum m/e 454 (M+H) +.

Step 6 Intermediate (5) was mixed with benzylalcohol, PPh3 in DCM and DEAD added, and the mixture allowed to react at room temperature for 18hrs. Intermediate 6 was isolated.

Mass Spectrum m/e 544 (M+H) +.

Step 7 The product of step 6 was then reacted with POC13 in MeCN for 15 hours at 110°C to yield Intermediate 7 in the above scheme.

Mass Spectrum m/e 424,426 (M+H) +.

Step 8 Futher processing of Intermediate (7) using reaction conditions analogous to those described in Example 18 step 2 but with a temperature of 80°C held for 18hrs produced Intermediate 8.

Mass Spectrum m/e 334, 336 (M+H) +.

Step 9 Intermediate (9) was produced by reacting the product of step 8 with propargylbromide using conditions analogous to those described in Example 20 step 2. The reaction was carried out over 18 hours at room temperature.

Mass Spectrum m/e 372,374 (M+H) +.

Step 10 Intermediate (10) prepared as described in Example 27 step 3 was reacted with Intermediate (9) above using conditions analogous to those described in Example 18 step 1 but maintaining the reaction mixture at 100°C for 3hrs. The title compound was obtained.

Mass Spectrum m/e 634 (M+H) +.

NMR Spectrum (d-6-DMSO + d-4-AcOH, 8 values) 0.40 (m, 2H), 0.60 (m, 2H), 1.84 (m, 1H), 2.61 (m, 2H), 3.68 (m, 3H), 3.87 (m, 2H), 4.42 (s, 2H), 4.64 (m, 2H), 4.99 (m, 2H), 7.02 (m, 5H), 7.14 (m, 1H), 7.38 (d, 2H), 7.35 (s, 1H), 8.16 (s, 1H), 8.81 (s, 1H).

Example 29 Preparation of Compound No. 29 in Table 2 Using a method analogous to that described in Example 23 but with 2- (N, N-dimethylamido)- pyrrolidine as the amine, the title compound was obtained.

Mass Spectrum m/e 596 (M++H).

Example 30 Preparation of Compound 30 in Table 2 The title compound was obtained using a method analogous to that described in Example 25 but with dimethylamine used instead of morpholine in step 2.

Mass Spectrum m/e 515 (M++H).

NMR Spectrum (d-6-DMSO, 6 values) 2.87 (m, 6H), 3.32 (m, 2H), 3.76 (s, 3H), 4.01 (s, 3H), 4.21 (d, 2H), 4.45 (m, 1H), 6.17 (broad, 1H), 6.99 (m, 3H), 7.08 (m, 1H), 7.21 (m, 2H), 7.41 (m, 2H), 7.60 (s, 1H), 8.23 (s, 1H), 8. 89 (s, 1H), 9.90 (broad, 1H), 11. 10 (broad, 1H) Example 31 Preparation of Compound No. 31 in Table 2 rr c- 0 0 I I ol,, 0 HN I I I I N HN'v CI N N v/uN PN<N N N N (1) Example 31

The title compound was prepared using a method analogous to that described in Example 19 above except that cyclopropylmethylamine was used as the amine in step 3 and the reaction was allowed to proceed over 9 days.

Mass Spectrum m/e 581 (M+H) +.

NMR Spectrum (d-6-DMSO, 8 values) 0.37 (m, 2H), 0.58 (m, 2H), 1.07 (m, 1H), 2.04 (m, 2H), 2.65 (d, 3H), 2.82 (m, 2H), 3.00 (m, 2H), 4.03 (s, 3H), 4.47 (s, 2H), 6.95 (s, 1H), 7.05 (m, 4H), 7.17 (m, 3H), 7.41 (d, 2H), 7.58 (m, 1H), 7.95 (s, 1H), 8.82 (s, 1H), 8.91 (bs, 1H).

Example 32 Preparation of Compound No. 32 in Table 2 f ° fr°Ti TYS' >-'---"A. TYj'HC! sa! t N t NJS 3 N 0 N HCI Salt O N N0 N O H (1) (2) Example32 The title compound was prepared from intermediate (1) by reacting with 20 molar equivalents of cyclopropylamine at a temperature of 60°C for 18hr in dimethyl acetamide.

Mass Spectrum m/e 527 (M++H) NMR Spectrum (d-6-DMSO, 8 values) 0.83 (m, 2H), 1.01 (m, 2H), 2.84 (m, 1H), 3.22 (m, 1H), 3.82 (s, 3H), 4.06 (s, 3H), 4.29 (d, 2H), 4.44 (m, 1H), 7.04 (m, 3H), 7.13 (m, 1H), 7.27 (m, 2H), 7.46 (d, 2H), 7.60 (s, 1H), 8.24 (s, 1H), 8. 94 (s, 1H), 9.16 (broad, 1H), 9.25 (broad, 1H), 11.08 (broad, 1H) Intermediate (1) Intermediate (1) was prepared by reacting Intermediate (B3) with (2S)- (+)-Glycidyl tosylate in the presence of anhydrous potassium carbonate at room temperature for 72 hours in DMSO.

Mass Spectrum m/e 470 (M++H)

Example 33 Preparation of Compound No. 33 in Table 2 ou -1 ilolw Nwl li nez -6 N''a I/,/lJN/S VNO/N CIO N (1) (2) Example 33 The title compound was prepared by reacting Intermediate (1) with 1 molar equivalent NaI, 5 molar equivalents anhydrous potassium carbonate and 5 molar equivalents 3- (methylsulfonyl) pyrrolidine (2) at a temperature of 60°C for 18 hours in dimethyl acetamide.

Mass Spectrum m/e 603 (M++H) NMR Spectrum (d-6-DMSO, 8 values, 373K) 2.32 (m, 2H), 3.10 (s, 3H), 3.34 (m, 8H), 3.77 (s, 3H), 3.98 (s, 3H), 4.18 (m, 1H), 4.34 (t, 2H), 6.97 (m, 3H), 7.05 (m, 1H), 7.17 (m, 2H), 7.31 (d, 2H), 7.51 (s, 1H), 8. 01 (s, 1H), 8.61 (s, 1H), 10.17 (broad, 1H) Intermediate (1) Intermediate (1) was prepared by reacting Intermediate (B3) with 1-chloro-3-bromopropane at room temperature for 18hours in the presence of KOtBu (1. OM in THF) in DMSO Mass Spectrum m/e 490 (M++H) Example 34 Preparation of Compound No. 34 in Table 2 qui ou /O \/O \ N N X t N JS t vwl,N N0 N G Example 34 The title compound was prepared by reacting Intermediate (1) with 5 molar equivalents of azetidine at a temperature of 60°C or 20 hours in dimethyl acetamide.

Mass Spectrum m/e 527 (M++H) NMR Spectrum (d-6-DMSO, 8 values) 2.19 (m, 1H), 3.09 (m, 1H), 3.33 (m, 3H), 3.74 (s, 3H), 3.76 (m, 1H), 3.97 (s, 3H), 4.13 (m, 4H), 4.20 (m, 1H), 5.91 (broad, 1H), 6.97 (m, 3H), 7.05 (m, 1H), 7.19 (m, 2H), 7.34 (d, 2H), 7.47 (s, 1H), 8.05 (s, 1H), 8.73 (s, 1H), 8.88 (broad doublet, 1H), 10.26 (broad, 1H), 10.57 (broad, 1H) Intermediate (1) Intermediate (1) was prepared by reacting Intermediate (B3) with (2S)- (+)-Glycidyl tosylate in the presence of anhydrous potassium carbonate in DMSO at room temperature for 72 hours.

Mass Spectrum m/e 470 (M++H) Example 35 Preparation of Compound No. 35 in Table 2 ?" Y°vS fT°n i o i o N 'o I N + N-- ; N HCI Salt O N NO N (1) (2) Example 35

The title compound was prepared by reacting the Intermediate (1) with 5 molar equivalent of 2,6-dimethylpiperidine at a temperature of 60°C for 20 hours in dimethyl acetamide.

Mass Spectrum m/e 583 (M++H) NMR Spectrum (d-6-DMSO, 8 values, 373K) 1.35 (s, 2H), 1.50 (m, 4H), 1.71 (m, 6H), 3.45 (m, 2H), 3.77 (s, 3H), 3.98 (s, 3H), 4.27 (m, 2H), 4.47 (m, 1H), 6.96 (m, 3H), 7.03 (m, 1H), 7.17 (m, 2H), 7.29 (d, 2H), 7.48 (s, 1H), 7.96 (s, 1H), 8. 57 (s, 1H), 9.78 (broad, 2H) Intermediate (1) Intermediate (1) was prepared by reacting Intermediate (B3) with (2S)- (+)-Glycidyl tosylate in the presence of anhydrous potassium carbonate at room temperature for 72 hours in DMSO.

Mass Spectrum m/e 470 (M++H) Example 36 Preparation of Compound No. 36 in Table 2 ou ou ow ow NJSt NJS t .. I 0 D 1 (-)" -"r-N,, S, N,) HCI salt < S, NC < HCI It Cl~O4N O ° CNOv sa (1) (2) Exampte36 The title compound was prepared by reacting Intermediate (1) with 6 molar equivalents l- (ethylsulfonyl) piperazine (2) in the presence of 1 molar equivalent NaI and 5 molar equivalents anhydrous potassium carbonate at a temperature of 60°C for 18 hours in dimethyl acetamide.

Mass Spectrum m/e 632 (M++H) NMR Spectrum (d-6-DMSO, 8 values) 1.19 (t, 3H), 1.96 (m, 2H), 2.44 (m, 6H), 3.03 (q, 2H), 3.17 (m, 4H), 3.74 (s, 3H), 3.90 (s, 3H), 4.18 (t, 2H), 6.91 (m, 3H), 7.02 (m, 1H), 7.17 (m, 2H), 7.24 (d, 2H), 7.30 (s, 1H), 7.73 (s, 1H), 8.37 (s, 1H), 9.38 (s, 1H) Intermediate (1) oll oll 0, 1 Z, 11 N I I HO-N I I' (B1) (1) Inter mediate (1) was prepared by reacting Intermediate (B3) with 1-chloro-3-bromopropane in the presence of KOtBu (1. OM in THF) at room temperature for 18 hours in DMSO.

Mass Spectrum m/e 490 (M++H) Example 37 Preparation of Compound No. 37 in Table 2 f N N , p I \ N + 001 i I \ \ N HCI Scalt NJS HCI salt --'0 N 0 (2) Example 37 0 The title compound was prepared by reacting Intermediate (1) with 6 molar equivalents N- (2- hydroxyethyl)-2-hydroxyethylamine (2) in the presence of lmolar equivalent NaI and 5 molar equivalents anhydrous potassium carbonate at a temperature of 60°C for 18 hours in dimethyl acetamide.

Mass Spectrum m/e 559 (M++H) NMR Spectrum (d-6-DMSO, 8 values) 2.31 (m, 2H), 3.29 (m, 4H), 3.37 (m, 2H), 3.72 (s, 3H), 3.78 (m, 4H), 3.99 (s, 3H), 4.27 (t, 2H), 6.93 (m, 3H), 7.04 (m, 1H), 7.17 (m, 2H), 7.38 (d, 2H), 7.57 (s, 1H), 8.24 (s, 1H), 8.87 (s, 1H), 9.93 (broad, 1H)

Intermediate (1) Intermediate (1) was prepared by reacting Intermediate (B3) with 1-chloro-3-bromopropane in the presence of KOtBu (1. OM in THF) at room temperature for 18 hours in DMSO.

Mass Spectrum m/e 490 (M++H) Example 38 Preparation of Compound No. 38 in Table 2 0"f O O , Ll 1! 0 NJa t NJS t NEZ , p I WN + yC I \N 0 0"T 0 N (B1) (2) Example 38 The title compound was prepared by reacting Intermediate (B3) with 1.2 molar equivalents l-chloro-3-methoxypropan-2-ol (2) in the presence of 2.2 molar equivalents anhydrous potassium carbonate at a temperature of 65°C for 66hr followed by a temperature of 85°C for 24 hours in dimethyl formamide.

Mass Spectrum m/e 502 (M++H) Example 39 Preparation of Compound No. 39 in Table 2 f oo ow , Ni ilol I/N N \/ i \ \ N + NC N I \N ip Ns ouN (1) (2) 0-l. Example 39 The title compound was prepared by reacting Intermediate (B3) with 1.2 molar equivalents 1- (2-chloroethyl)-2-methyl-5-nitro-lH-imidazole (2) in the presence of 2.2 molar equivalents

anhydrous potassium carbonate at a temperature of 65°C for 66 hours followed by 85°C for 24 hours in dimethyl formamide.

Mass Spectrum m/e 567 (M++H) Example 40 Preparation of Compound No. 40 in Table 3 The title compound was prepared according to the following scheme: F 0 N N () ClAO T ,, CIO N (A8) F) aO 11 F 0 N N : Cr N'N ION C. N \ \ \ \ N N ci"o N Example 40 (2) In the final step the title compound was prepared by reaction of Intermediate (2) with cyclopropylamine in the presence of NaI at temperature of 50°C for 24hr.

Mass Spectrum m/e 506 (M++H) NMR Spectrum (d-6-DMSO, 8 values) 0.75 (d, 2H), 0.9 (br s, 2H), 2.2 (m, 2H), 2.75 (br s, 1H), 3.2 (br s, 2H), 4.0 (s, 3H), 4.3 (m, 2H), 7.35 (m, 3H), 7.55 (s, 1H), 7.6 (d, 1H), 7.65 (t, 1H), 8.3 (s, 1H), 8.9 (s, 1H), 9.4 (br s, 2H) Intermediate (1) Intermediate (1) was prepared by reaction of 2-bromothiazole and 4-amino-3-fluorophenol in the presence of KOtBu at a temperature of 150°C for lhr in dimethylacetamide.

Mass Spectrum m/e 211 (M++H) Intermediate (2) Intermediate (2) was prepared by reaction of Intermediate (1) with Intermediate (A8) at a temperature of 110°C for 5hr in n-propanol.

Mass Spectrum m/e 485 (M++H)

Example 41 Preparation of Compound No. 41 in Table 3 0 N 0 N 0 N I, N \ \ \ \ i N"'+ >NH, 1. >-N---O-'-N' Example 41 The title compound was prepared by reaction of Intermediate (1) with cyclopropylamine in the presence of sodium iodide at 50°C for 24 hours.

Mass Spectrum m/e 488 (M++H) NMR Spectrum (d-6-DMSO, 8 values) 0.7 (m, 2H), 0.9 (m, 2H), 2.25 (m, 2H), 2.7 (m, 1H), 3.15 (m, 2H), 4.0 (s, 3H), 4.3 (t, 2H), 6.9 (d, 1H), 7.35 (d, 2H), 7.5 (d, 2H), 7.6 (s, 1H), 8.3 (s, 1H), 8. 9 (s, 1H), 9.05 (d, 1H), 9.4 (br s, 2H), 11.4 (br s, 1H) Intermediate (1) Intermediate 1 Intermediate (1) was prepared by reaction of Intermediate (A8) with Intermediate (2) in n- propanol at a temperature of 110°C for 5 hours.

Mass Spectrum m/e 467 (M++H) Intermediate (2) was prepared according to the following scheme: Intermediate (3) l-Fluoro-4-nitrobenzene was reacted with 3-hydroxy-isothiazole in the presence of KOtBu in dimethylacetamide at a temperature of 150°C for lhour.

Intermediate 2 Intermediate (2) was prepared by the reduction of Intermediate (3) using palladium over carbon in ethyl acetate for 36 hours Mass Spectrum m/e 193 (M++H) Example 42 Preparation of Compound No. 42 in Table 4 / v 0 o_ CL N i I w w \ % CI O/N/H CI O/N/ w w (A8) (1) Example 42 4-chloro-7- (3-chloropropoxy)-6-methoxyquinoline-3-carbonitrile (900 mg, 2.9 mmol) and 3- (2-methoxyphenoxy) aniline (Reference: WO 2001056990) were heated together in n-propanol for 4 hours. The resulting solid was filtered and washed first with cold n-propanol and then with diethyl ether and then dried in vacuo to leave a solid (1.22g, 80 %).

Mass Spectrum ES 488 (MH-), 490 (MH+) H NMR spectrum (300 MHz, DMSO) : 2.22-2.32 (m, 2H), 3.74 (s, 3H), 3.83 (t, 2H), 3.97 (s , 3H), 4.30 (t, 2H), 6.90 (m, 2H), 6.98 (dd, 1H), 7.10 (m, 2H), 7.14-7.22 (m, 2H), 7.41 (t, 1H), 7.48 (s, 1H), 8.04 (s, 1H), 8.90 (s, 1H)m, 10.6 (br s, 1H).

Intermediate (1) This prepared by a method analogous in Intermediate (B2) in Preparation B above.

Example 43 Preparation of Compound No. 43 in Table 4 n\l Q-. P o- N N-- C 0 a I Nwa Example 42 Example 43 7- (3-chloropropoxy)-6-methoxy-4- f [3- (2-methoxyphenoxy) phenyl] amino} quinoline-3- carbonitrile (200 mg, 0.41mmol) [Example 42] and 4,4-difluoropiperidine (200 mg, 1.8 mmol) and sodium iodide (184 mg, 1.23 mmol) were heated in dimethylacetamide (lOml) at

40 C for 12 hours. The reaction was evaporated and the residue was partitioned between dichloromethane and water. The organic layer was separated and the aqueous layer re- extracted with dichloromethane. The organics were combined, dried over magnesium sulfate, filtered and then evaporated in vacuo to leave a gum. The crude product was triturated with diethyl ether to leave the title compound as a solid (28 mg, 12 %).

H NMR spectrum (400MHz, DMSO + acetic acid at 373K ! 2.19 (m, 6 H), 3.12 (m, 6H), 3.79 (s, 3H), 3.90 (s, 3H), 4.28 (t, 2 H), 6.73 (m, 2H), 6.88 (d, 1H), 6.95 (m, 1H), 7.08 (d, 1H), 7.15 (m, 2H), 7.32 (t, 1H), 7.43 (s, 1H), 7.65 (s, 1H), 8. 50 (s, 1H).

Mass Spectrum ES 575 (MH+) Example 44 Preparation of Compound No. 44 in Table 4 Example 42 Example 44 7-(3-chloropropoxy)-6-methoxy-4- {[3-(2-methoxyphenoxy) phenyl] amino} quinoline-3- carbonitrile (200mg, 0.41 mmol) was dissolved in 1-isopropylpiperazine (2 ml) and heated at 45 C for 12 hours. The reaction mixture was partitioned between dichloromethane and water.

The organic layer was separated and the aqueous layer re-extracted with dichloromethane.

The organics were combined, dried over magnesium sulfate, filtered and then evaporated in vacuo. The residue was purified by flash chromatography, eluting over a gradient from dichloromethane to 10 % methanol in dichloromethane to give an orange oil. The oil was triturated in hexane leave the title compound as a solid (9lmg, 38%).

H NMR spectrum (300 MHz, CD13) 1.08 (d, 6H), 2.14 (m, 2H), 2.55 (m, 11H), 3.66 (s, 3H), 3.82 (s, 3H), 4.24 (t, 2H), 6.62 (m, 1H), 6.78 (m, 3H), 6.89-7.04 (m, 4H), 7.15 (m, 1H), 7.34 (s, 1H), 8. 62 (s, 1H).

Mass Spectrum ES 580 (MH-), 582 (MH+) Example 45 Preparation of Compound No. 45 in Table 4 Example 42 Example 45 7-(3-chloropropoxy)-6-methoxy-4-{[3-(2-methoxyphenoxy) phenyl] amino} quinoline-3- carbonitrile (300 mg, 0.6 mmol) [Example 42], 3,3-difluoropyrolidine hydrochloride (171 mg, 1.2 mmol), sodium iodide (180 mg, 1.2 mmol) and potassium carbonate (414 mg, 3.0 mmol) were heated at 85°C in dimethylformamide (20ml) for 12 hours. The reaction mixture was evaporated and the residue was partitioned between dichloromethane and water.

The organic layer was separated and the aqueous layer re-extracted with dichloromethane.

The organics were combined, dried over magnesium sulfate, filtered and then evaporated in vacuo. The residue was purified by flash chromatography, eluting with a gradient of dichloromethane to 4 % methanol in dichloromethane to give an oil. The oil was triturated with diethyl ether to leave the title compound as a solid (108 mg, 32 %).

H NMR spectrum (300 MHz, DMSO): 1.95 (m, 2H), 2.23 (m, 2H), 2.58 (t, 2H), 2.71 (t, 2H), 2.90 (t, 2H), 3.74 (s, 3H), 3.89 (s, 3H), 4.19 (t, 2H), 6.64 (m, 2H), 6.84 (d, 1H), 6.95 (m, 1H), 7.07 (d, 1H), 7.15 (m, 2H), 7.27 (t, 1H), 7.30 (s, 1H), 7.63 (s, 1H), 8.44 (s, 1H), 9.39 (s, 1H).

Mass Spectrum ES 559 (MH-), 561 (MH+) Example 46 Preparation of Compound No. 46 in Table 4 ou 0, 1 ° ou ou NJS t NJS t 11 1 :), Si HCI salt ° J! J Example 42 (2) o, si Example 46

The title compound was prepared by reaction of Example 42 with 5 molar equivalents of Intermediate (2) (1-oxothiomorpholine) in the presence of 1 molar equivalent of NaI and 5 molar equivalents of anhydrous potassium carbonate at a temperature of 60°C for 18 hours in dimethyl acetamide.

Mass Spectrum m/e 573 (M++H) NMR Spectrum (d-6-DMSO, 8 values) 2.44 (m, 2H), 3.24 (d, 2H), 3.45 (m, 6H), 3.71 (m, 2H), 3.81 (s, 3H), 4.06 (s, 3H), 4.38 (t, 2H), 7.04 (m, 3H), 7.13 (m, 1H), 7.26 (m, 2H), 7.46 (d, 2H), 7.56 (s, 1H), 8.21 (s, 1H), 8.95 (s, 1H), 11.06 (broad, 1H), 11.44 (broad, 1H) Intermediate (2) (1-oxothiomorpholine) Intermediate (2) was prepared as described in Chadha et al (1983) J. Med. Chem. (1983), 26 (6), 916-22 Example 47 Preparation of Compound No. 47 in Table 4 ou ouzo N'Cro-6 0 Na N-v O I N N N'O I N N HCI Scalt ci 0 N 0 z: SJ Example 42 (2) ó Example 46 The title compound was prepared by reaction of Example 42 with 5 molar equivalents of Intermediate (2) (1, 1-dioxothiomorpholine) in the presence of lmolar equivalent ofNaI and 5 molar equivalents of anhydrous potassium carbonate at a temperature of 60°C for 120 hours in dimethyl acetamide.

Mass Spectrum m/e 589 (M++H) NMR Spectrum (d-6-DMSO, 8 values) 2.32 (m, 2H), 3.42 (m, 8H), 3.63 (m, 2H), 3.74 (s, 3H), 3.99 (s, 3H), 4.30 (t, 2H), 6.98 (m, 3H), 7.05 (m, 1H), 7.19 (m, 2H), 7.40 (d, 2H), 7.48 (s, 1H), 8.15 (s, 1H), 8.91 (s, 1H), 11.05 (broad, 1H) Intermediate (2) (1, 1,-dioxomorpholine) Intermediate (2) was prepared as described in Lazer et al (1994) J. Med. Chem. 37 (7), 913- 23.

Example 48 Preparation of Compound No. 48 in Table 4 0.11 0. oxo ow o i NJS t NJS t N N N w w N + °, s ,, 5 ° w w N I-ICI Salt ci 0 N Exaple 42 Example 47 The title compound was prepared by reactin of Example 42 with 5 molar equivalents of 1, 1-dioxo-3-amino-tetrahydrothiophene in the presence of lmolar equivalent ofNaI and 5 molar equivalents of anhydrous potassium carbonate at a temperature of 60°C for 144 hours in dimethyl acetamide.

Mass Spectrum m/e 589 (M++H) NMR Spectrum (d-6-DMSO, 8 values, 373K) 2.28 (m, 2H), 2.40 (m, 1H), 2.65 (m, 1H), 3.18 (m, 2H), 3.48 (m, 4H), 3.78 (s, 3H), 3.98 (s, 3H), 4.09 (m, 1H), 4.35 (t, 2H), 6.97 (m, 3H), 7.03 (m, 1H), 7.17 (m, 2H), 7.31 (d, 2H), 7.50 (s, 1H), 7.99 (s, 1H), 8.61 (s, 1H), 10.14 (broad, 1H) Example 49 Preparation of Compound No. 49 in Table 4 ?" NEZ , N- 0 NJS t NX t wOtN CN zOe HCI salt mO N CN<O N OHH S J O (1) Example 49 The title compound was prepared by reaction of Intermediate (1) wih 20 molar equivalents of thiomorpholine at a temperature of 60°C for 18 hours in dimethyl acetamide.

Mass Spectrum m/e 573 (M++H) NMR Spectrum (d-6-DMSO, 8 values) 2.85 (m, 2H), 3.26 (m, 6H), 3.74 (s, 3H), 3.76 (m, 2H), 3.99 (s, 3H), 4.19 (m, 2H), 4.54 (m, 1H), 6.11 (broad, 1H), 6.97 (m, 3H), 7.05 (m, 1H), 7.19 (m, 2H), 7.38 (d, 2H), 7.54 (s, 1H), 8. 15 (s, 1H), 8.84 (s, 1H), 10.36 (broad, 1H), 10.91 (broad, 1H)

Intermediate (1) Intermediate (1) was prepared by reaction of Intermediate (B3) with (2S)- (+)-Glycidyl tosylate in the presence of anhydrous potassium carbonate at room temperature for 72 hours in DMSO.

Mass Spectrum m/e 470 (M++H) Example 50 Preparation of Compound No. 50 in Table 4 oll 0. 0, 1-11 i ° I I/+ Q N, N 0, 6 , I I I Nla 6 zon O ) I- _", 0 (1) Example 50 Intermediate (1) was reacted with 1, 5-anhydro-2,4-dideoxy-3-C-methyl-4-(methylamino)-D- erythro-pentitol in the presence of NaI and potassium carbonated at a temperature of 1000C for 24 hours in DMA.

Mass Spectrum m/e 599.27 (M++H).

NMR Spectrum (d-6-DMSO, b values) 1.42 (s, 3H), 1.74 (m, 2H), 2.36 (m, 2H), 2.74&2.97 (2d, 3H), 3.2-3.8 (m, 6H), 3.74 (s, 3H), 3.9 (s, 3H), 4.13-4.35 (m, 3H), 6.95 (m, 3H), 7.04 (d, 1H), 7.19 (m, 2H), 7.4 (d, 2H), 7.55 (s, 1H), 8.22 (s, 1H), 8.9 (s, 1H), 9.7 (br. s, 1H), 11.2 (v. br. s, 1H).

Example 51 Preparation of Compound No. 51 in Table 4 I oN N \I o \N HCI salt 1, j (- CI O N N o, r l HCI salt (1) (2) "Exampte51

The title compound was prepared by reaction of Intermediate (1) with 5 molar equivalents of Intermediate (2) in the presence of 1 molar equivalent of Nal and 5 molar equivalents of anhydrous potassium carbonate at a temperature of 60°C for 44 hours in dimethyl acetamide.

Mass Spectrum m/e 550 (M++H) NMR Spectrum (d-6-DMSO, 8 values) 2.43 (m, 2H), 3.26 (d, 2H), 3.45 (m, 6H), 3.71 (m, 2H), 4.06 (s, 3H), 4.38 (t, 2H), 7.35 (d, 1H), 7.40 (d, 1H), 7.60 (m, 5H), 8. 17 (s, 1H), 8.93 (s, 1H), 10.92 (broad, 1H), 11.29 (broad, 1H) Intermediate (1) Intermediate (1) was prepared as described in Example 41.

Intermediate (2) Intermediate (2) was prepared as desribed in Chadha et al (1983) J. Med. Chem. 26 (6), 916- 22.

Example 52 Preparation of Compound No. 52 in Table 4 0. 11 roll-6 ° N I I i \ \ N + Br I \N N JS t N JS t JU. y y-0 HO N Example 52 The title compound was prepared by reacton of Intermediate (B3) with 1.2 molar equivalents of 2-bromomethytetrahydrofuran in the presence of 2.2 molar equivalents of anhydrous potassium carbonate at a temperature of 65°C for 44 hours in dimethyl formamide.

Mass Spectrum m/e 498 (M++H) Example 53 Preparation of Compound No. 53 in Table 4 oo ow O N JUS I ; zz : p wN + i/B I \N i I \ \ Ho cr 0 0 N N Example 53 The title compound was prepared by reaction of Intermediate (B3) with 1.2 molar equivalents of 2-bromomethyltetrahydropyran in the presence of 2.2 molar equivalents anhydrous potassium carbonate at a temperature of 65°C for 44 hours in dimethyl formamide.

Mass Spectrum m/e 512 (M++H) Example 54 Preparation of Compound No. 54 in Table 4 ou 0, ° I I I i o, o N'Cro-6 0--, ( 1\ 6 i0 I \ \ wN NJ I HO N N N (B3) (2) Example 53 The title compound was prepared by reaction of Intermediate (B3) with 1.2 molar equivalents of 5- (S)-para-toluenesulphonyloxymethyl-3-isopropyl-1, 3-oxazolan-2-one in the presence of 2.2 molar equivalents of anhydrous potassium carbonate at a temperatue of 65°C for 44 hours in dimethyl formamide.

Mass Spectrum m/e 555 (M++H) Example 55 Preparation of Compound No. 55 in Table 4 / /ru o N'au N I I p I N + O-N"CI HO N, o/ O-N (B3) Example 55

The title compound ws prepared by reaction of Intermediate (B3) with 1.2 molar equivalents of 3- (chloromethyl)-5-methylisoxazole in the presence of 2.2 molar equivalents anhydrous potassium carbonate at a temperature of 65°C for 44 hours in dimethyl formamide.

Mass Spectrum m/e 509 (M++H) Example 56 Preparation of Compound No. 56 in Table 4 ou 0 o 0, N. v i0 (\ HO-N =N I/i N CNO N HO N N Example 56 The title compound was prepared by reaction of Intermediate (B3) with 1.2 molar equivalents of 1- (2-chloroethyl)-pyrazole in the presence of 2.2 molar equivalents of anhydrous potassium carbonate at a temperature of 100°C for 42 hours in dimethyl formamide.

Mass Spectrum m/e 508 (M++H) Example 57 Preparation of Compound No. 57 in Table 4 0 0 °TS fVn N'C 0 0. N O ; N su N N' CI O N/N O N (1) Example 57 The title compound was prepared by reaction of Intermediate (1) with 6 molar equivalents of 1, 1-dioxo-3-N-methylamino-tetrahydrothiophene in the presence of 1 molar equivalent of NaI and 5 molar equivalents of anhydrous potassium carbonate at a temperature of 60°C for 18 hours in dimethyl acetamide.

Mass Spectrum m/e 603 (M++H) NMR Spectrum (d-6-DMSO, 8 values) 2.34 (m, 4H), 2.68 (m, 1H), 3.23 (m, 2H), 3.46 (m, 4H), 3.72 (s, 3H), 3.98 (s, 3H), 4.26 (m, 3H), 6.96 (m, 3H), 7.05 (m, 1H), 7.18 (m, 2H), 7.40 (d, 2H), 7.56 (s, 1H), 8.22 (s, 1H), 8.90 (s, 1H) Intermediate (1) Intermediate (1) was prepared by reaction of Intermediate (B3) with 1-chloro-3-bromopropane in the presence of KOtBu (1. OM in THF) at room temperature for 18 hours in DMSO.

Mass Spectrum m/e 490 (MF+H) Example 58 Preparation of Compound No. 58 in Table 4 Step 1 Intermediate (1) was prepared by alkylation of Intermediate (A7) with N- (3-chloropropyl)- morpholine in toluene solution at 110°C for 18 hours in the presence of cesium carbonate, tetra-n-butyl ammonium iodide and 4A sieves Mass Spectrum m/e 511 (M+H) +.

Step 2 Intermediate (2) was prepared from salicyl hydrazide by reaction of the hydrazide with triethylorthoformate to give, after chromatographic purification, the 2-hydroxyphenyloxadiazole, Mass Spectrum m/e 163 (M++H), then the 2-hydroxyphenyloxadiazole was reacted with 4-fluoronitrobenzene, in a procedure analogous to that in Preparation B to give, after chromatographic purification, 2- (4- nitrophenoxy) phenyloxadiazole.

Mass Spectrum m/e 284 (M++H), 2- (4-Nitrophenoxy) phenyloxadiazole was reduced in ethyl acetate solution with hydrogen and 5% Pd/C to produce Intermediate (2).

Mass Spectrum m/e 254 (M++H) Step 3 Intermediate (1) was reacted with Intermediate (2) in n-propanol solution at 110°C for 16 hours in the presence of 1 molar equivalent of 1. OM ethereal HC1 to give, after chromatographic purification, the title compound Mass Spectrum m/e 579 (M++H).

Example 59 Preparation of Compound No. 59 in Table 4 oll 0.1 ow on CN NEZ "HCI salt N Ja t N X t Go Example 59 The title compound was prepared by reaction of Intermediate (1) with 20 molar equivalents of pyrrolidine at a temperature of 60°C for 18 hours in dimethyl acetamide.

Mass Spectrum m/e 541 (M++H) NMR Spectrum (d-6-DMSO, 8 values) 1.95 (m, 4H), 3.11 (m, 2H), 3.35 (m, 2H), 3.61 (m, 2H), 3.74 (s, 3H), 3.99 (s, 3H), 4.19 (m, 2H), 4.38 (m, 1H), 6.09 (broad, 1H), 6.97 (m, 3H), 7.05 (m, 1H), 7.19 (m, 2H), 7.38 (d, 2H), 7.55 (s, 1H), 8.17 (s, 1H), 8.85 (s, 1H), 10.10 (broad, 1H), 10.98 (broad, 1H) Intermediate (1) Intermediate (1) was prepared by reaction of Intermediate (B3) with (2S)- (+)-Glycidyl tosylate in the presence of anhydrous potassium carbonate at room temperature for 72 hours in DMSO.

Mass Spectrum m/e 470 (M++H) Example 60 Preparation of Compound No. 60 in Table 4 ou ou 0 ° I I N I I N i C_ci o i HO O-N N » <O N (B3) Example 60 The title compound was prepared by reaction of Intermediate (B3) with 1.2 molar equivalents 3- (chloromethyl)-1, 2,4-oxadiazole in the presence of 2.2 molar equivalents anhydrous potassium carbonate at a temperature of 100°C for 72 hours in dimethyl formamide.

Mass Spectrum m/e 453 (M++H) NMR Spectrum (d-6-DMSO, 8 values) 3.74 (s, 3H), 3.94 (s, 3H), 5.38 (s, 2H), 6.91 (m, 3H), 7.02 (m, 1H), 7.17 (m, 2H), 7.28 (d, 2H), 7.50 (s, 1H), 7.86 (s, 1H), 8.42 (s, 1H), 9.50 (s, 1H) Example 61 Preparation of Compound No. 61 in Table 4 0 0. -6 I' NJS t 0), :-,)---, . (1) Example 61 (1) 2 Example 61 The title compound was prepared by reaction of Intermediate (1) with 1.2 molar equivalents sodium methanesulfinate at a temperature of 100°C for 18 hours in dimethyl acetamide.

Mass Spectrum m/e 534 (M++H) NMR Spectrum (d-6-DMSO, 8 values) 2.23 (m, 2H), 3.03 (s, 3H), 3.28 (t, 2H), 3.73 (s, 3H), 3.91 (s, 3H), 4.25 (t, 2H), 6.91 (m, 3H), 7.02 (m, 1H), 7.15 (m, 2H), 7.23 (d, 2H), 7.31 (s, 1H), 7.75 (s, 1H), 8.37 (s, 1H), 9.40 (s, 1H)

Intermediate (1) Intermediate (1) was prepared by reaction of Intermediate (B3) with 1-chloro-3-bromopropane in the presence of KOtBu (1. OM in THF) at room temperature for 18 hours in DMSO.

Mass Spectrum m/e 490 (M++H) Example 62 Preparation of Compound No. 62 in Table 4 oll oll 0, 1 N I I Nazi N N O i i \ \ N CI I/ i NO N O. l. 'N- 0 Example 62 The title compound was prepared by reaction of Intermediate (1) with 1, 5-anhydro-2,4- dideoxy-3-C-methyl-4- (methylamino)-D-erythro-pentitol in the presence of NaI and potassium carbonate at a temperature of 1000C for 24 hours.

Mass Spectrum m/e 613 (M++H).

NMR Spectrum (d-6-DMSO, 8 values) 1.40 (s, 3H), 1.64-1.78 (m, 2H), 1.78-2.09 (m, 4H), 2.69 (d, 3H), 3.14-3.52 (m, 3H), 3.6 (t, 2H), 3.71 (s, 3H), 3.65-3.8 (m, 1H), 3.98 (s, 3H), 4.1- 4.29 (m, 3H), 6.9-7.0 (m, 3H), 7.03 (d, 1H), 7.2 (m, 2H), 7.38 (d, 2H), 7.94 (s, 1H), 8.13 (s, 1H), 8.85 (s, 1H), 9.43 (br. s, 1H).

Intermediate (1)

Intermediate (1) was prepared by reaction of Intermediate (B3) with 1-chloro-3-bromopropane in the presence of KOtBu (1. OM in THF) at room temperature for 18 hours in DMSO.

Mass Spectrum m/e 490 (M++H) Example 63 Preparation of Compound 63 in Table 5 3 o a iN I w ° w o I w ° I w N + I w w' i i i i i. han 0 ci N (C1) H, N O 0 O HzN o ci 0 N (CI) (2) (A8) O i \ \ NO N CIO I N ZU (Example63) Step 1 Intermediate (C1) was converted to the primary amide by treatment with formamide in DMA at room temperature for 5 minutes followed by treatment with sodium methoxide (25% in methanol) for Ihr at 100°C to yield, after chromatographic purification, Intermediate (2).

Mass Spectrum m/e 229.35 (M+H) + Step 2 Intermediate (2) was reacted with Intermediate (A8) in n-propanol solution at 105°C for 3 hours to give Intermediate (4).

Mass Spectrum m/e 503.27 (M+H) + Step 3 Intermediate (4) was reacted with morpholine in the manner described for Example 11, Step 2 to yield, after chromatographic purification, the title compound.

Mass Spectrum m/e 554.3 (M+H)- NMR Spectrum (d-6-DMSO, 6 values) 1.96 (m, 2H), 2.32-2.52 (m, 6H), 3.52-3.60 (m, 4H), 3.92 (s, 3H), 4.19 (t, 2H), 7.07-7.16 (m, 3H), 7.29-7.50 (m, 6H), 7.60 (d, 1H), 7.74 (s, 1H), 7.89 (s, 1H), 8. 39 (s, 1H), 9.47 (s, 1H).

Example 64 Preparation of Compound 64 in Table 6 0 0 0 o, N'OH H2Nfa (1)' (2) ! P'0 Cl~Ot a N (4) 0 (2) 0 0 ci 0 ZON (4) \/ i0 I/\ N O. E i0 I \ \ iN O NMO N CIMO N (5) Compound No 64 Step 1 4-Fluoro-nitrobenzene and 4-hydroxyphenol were reacted together in DMA in the presence of potassium butoxide in a manner analogous to the preparation of 4- (4-nitrophenoxy)-phenol (Rev. Chim. (Bucharest), 1988,39 (6), 477-482) to give Intermediate (1).

Step 2 Intermediate (1) was reacted with ethyl bromoacetate in acetone solution for 3hr at 65°C in the presence of potassium carbonate and then the nitro group reduced to the corresponding aniline by reduction, at room temperature in ethyl acetate solution, with hydrogen and catalytic 5% Pd/C to give, after chromatographic purification, intermediate (2).

Mass Spectrum m/e 288.63 (M+H) + NMR Spectrum (d-6-DMSO, 8 values) 1.19 (t, 3H), 4.14 (q, 2H), 4.68 (s, 2H), 4.88 (s, 2H), 6.55 (d, 2H), 6.68 (d, 2H), 6.75-6.87 (m, 4H).

Step 3 Intermediate (2) was converted to the corresponding N-methylamide by reaction with methylamine in methanol for 96hr at 65°C. The product was purified by chromatography to give intermediate (3).

Mass Spectrum m/e 273.59 (M+H) + NMR Spectrum (d-6-DMSO, S values) 2.63 (d, 3H), 4.37 (s, 2H), 4.88 (s, 2H), 6.54 (d, 2H), 6.68 (d, 2H), 6.80 (d, 2H), 6.89 (d, 2H), 7.86 (bs, 1H).

Step 4 Intermediate (3) was reacted with intermediate (A8) in n-propanol solution at 105°C for 3hr to give intermediate (5).

Mass Spectrum m/e 547.92 (M+H) + NMR Spectrum (d-6-DMSO, 8 values) 2.19 (m, 2H), 2.66 (d, 3H), 3.83 (t, 2H), 3.99 (s, 3H), 4.30 (t, 2H), 4.44 (s, 2H), 7.00 (s, 4H), 7.07 (d, 2H), 7.44 (d, 2H), 7.50 (s, 1H), 8.00 (bs, 1H), 8.15 (s, 1H), 8. 90 (s, 1H).

Step 5 Intermediate (5) was reacted with 2,6-dimethylmorpholine, for 96 hours at room temperature in the presence of sodium iodide to yield, after chromatographic purification, the title compound.

Mass Spectrum m/e 627.16 (M+H) + NMR Spectrum (d-6-DMSO D4 Acetic, 8 values) 1.12 (s, 3H), 1.14 (s, 3H), 2.34 (2H, m), 2.60-2.71 (m, 5H), 3.26 (m, 2H), 3.50 (d, 2H) 3.90-4.01 (m, 5H), 4.30 (t, 2H), 4.42 (s, 2H), 6.99 (s, 4H), 7.07 (d, 2H), 7.45 (d, 2H), 7.50 (s, 1H), 8.16 (s, 1H), 8.91 (s, 1H).

Assay for inhibitors of the MAP kinase pathway To evaluate inhibitors of the MAPK pathway a coupled assay was carried out which measures phosphorylation of serine/threonine residues present in the substrate in the presence or absence of inhibitor. Recombinant glutathione S-transferase fusion protein containing human p45MEKl (GST-MEK) was activated by c-raf (Sf9 insect cell lysate from triple baculoviral infection with c-raf/ras/lck) and used for the assay. Active GST-MEK was first used to activate a recombinant glutathione S-transferase fusion protein containing p44MAP kinase (GST-MAPK) in the presence of ATP and Mg2+ for 60min at room temperature in the presence or absence of potential inhibitors. The activated GST-MAPK was then incubated with myelin basic protein (MBP) as substrate for 10min at room temperature in the presence of ATP, Mg2+ and 33P-ATP. The reaction was stopped by addition of 20% v/v phosphoric acid. Incorporation of 33P into the myelin basic protein was determined by capture of the substrate on a filter mat, washing and counting using scintillation methods. The extent of inhibition was determined by comparison with untreated controls.

The final assay solution contained 10mM Tris, pH 7.5,0.05mM EGTA, 8.33uM [y33P] ATP, 8.33mM Mg (OAc) 2, 0. 5mM sodium orthovanadate, 0.05% w/v BSA, 6.5ng GST-MEK, lpg GST-MAPK and 16.5 gag MBP in a reaction volume of 60, u1.

Compounds tested of the present invention had ICso results typically less than 0. 5uM.

For example, Compound No 11 gave an IC50 of 0.0013uM.

In vitro MAP kinase assay To determine whether compounds were inhibiting GST-MEK or GST-MAPK, a direct assay of MAPK activity was employed. GST-MAPK was activated by a constitutively active GST-MEK fusion protein containing two point mutations (S217E, S221E) and used for the assay in the presence and absence of potential inhibitors. The activated GST-MAPK was incubated with substrate (MBP) for 60min at room temperature in the presence of ATP, Mg2+ and 33P-ATP. The reaction was stopped by addition of 20% v/v phosphoric acid.

Incorporation of 33P into the myelin basic protein was determined by capture of the substrate on a filter mat, washing and counting using scintillation methods.

The final assay solution contained 12mM Tris, pH 7.5,0.06mM EGTA, 30uM [y33P] ATP, lOmM Mg (OAc) 2, 0.6mM sodium orthovanadate, 0.06% w/v BSA, 28ng GST-MAPK and 16.51lg MBP in a reaction volume of 60p1.

Compounds of the invention showed activity in this screen.

Cell proliferation assays Cells were seeded into multi-well plates at 20 000-40 000 cells/ml in growth medium containing 5% FCS and incubated overnight at 37°C. The compounds were prepared in fresh medium at an appropriate concentration and added to the wells containing the cells. These were then incubated for a further 72 hours. Cells were then either removed from the wells by incubating with trypsin/EDTA and counted using a Coulter counter, or treated with XTT/PMS in PBSA and optical densities read at 450nm. Compounds tested of the present invention had IC50 results typically less than 30µ. For example, Compound No 11 gave an IC50 of 1. 3µM in HT29 human colon tumour cells.